patent_id
stringlengths
7
8
description
stringlengths
125
2.47M
length
int64
125
2.47M
11858893
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS The following definitions are provided for the purpose of understanding the present subject matter and for construing the appended patent claims. Definitions Throughout the application, where compositions are described as having, including, or comprising specific components, or where processes are described as having, including, or comprising specific process steps, it is contemplated that compositions of the present teachings can also consist essentially of, or consist of, the recited components, and that the processes of the present teachings can also consist essentially of, or consist of, the recited process steps. It is noted that, as used in this specification and the appended claims, the singular forms “a”, “an”, and “the” include plural references unless the context clearly dictates otherwise. In the application, where an element or component is said to be included in and/or selected from a list of recited elements or components, it should be understood that the element or component can be any one of the recited elements or components, or the element or component can be selected from a group consisting of two or more of the recited elements or components. Further, it should be understood that elements and/or features of a composition or a method described herein can be combined in a variety of ways without departing from the spirit and scope of the present teachings, whether explicit or implicit herein. The use of the terms “include,” “includes”, “including,” “have,” “has,” or “having” should be generally understood as open-ended and non-limiting unless specifically stated otherwise. The use of the singular herein includes the plural (and vice versa) unless specifically stated otherwise. In addition, where the use of the term “about” is before a quantitative value, the present teachings also include the specific quantitative value itself, unless specifically stated otherwise. As used herein, the term “about” refers to a ±10% variation from the nominal value unless otherwise indicated or inferred. As used herein, “halo” or “halogen” refers to fluoro, chloro, bromo, and iodo. As used herein, “alkyl” refers to a straight-chain or branched saturated hydrocarbon group. Examples of alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and z′-propyl), butyl (e.g., n-butyl, z′-butyl, sec-butyl, tert-butyl), pentyl groups (e.g., n-pentyl, z′-pentyl, -pentyl), hexyl groups, and the like. In various embodiments, an alkyl group can have 1 to 40 carbon atoms (i.e., C1-C40 alkyl group), for example, 1-30 carbon atoms (i.e., C1-C30 alkyl group). In some embodiments, an alkyl group can have 1 to 6 carbon atoms, and can be referred to as a “lower alkyl group” or a “C1-C6 alkyl group”. Examples of lower alkyl groups include methyl, ethyl, propyl (e.g., n-propyl and z′-propyl), and butyl groups (e.g., n-butyl, z′-butyl, sec-butyl, tert-butyl). In some embodiments, alkyl groups can be substituted as described herein. An alkyl group is generally not substituted with another alkyl group, an alkenyl group, or an alkynyl group. As used herein, “alkenyl” refers to a straight-chain or branched alkyl group having one or more carbon-carbon double bonds. Examples of alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl groups, and the like. The one or more carbon-carbon double bonds can be internal (such as in 2-butene) or terminal (such as in 1-butene). In various embodiments, an alkenyl group can have 2 to 40 carbon atoms (i.e., C2-C40 alkenyl group), for example, 2 to 20 carbon atoms (i.e., C2-C20 alkenyl group) or 2 to 6 carbon atoms (i.e., C2-C6 alkenyl group). In some embodiments, alkenyl groups can be substituted as described herein. An alkenyl group is generally not substituted with another alkenyl group, an alkyl group, or an alkynyl group. The term “substituted alkyl” as used herein refers to an alkyl group in which 1 or more (up to about 5, for example about 3) hydrogen atoms is replaced by a substituent independently selected from the group: —O, —S, acyl, acyloxy, optionally substituted alkoxy, optionally substituted amino (wherein the amino group may be a cyclic amine), azido, carboxyl, (optionally substituted alkoxy)carbonyl, amido, cyano, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, halogen, hydroxyl, nitro, sulfamoyl, sulfanyl, sulfinyl, sulfonyl, and sulfonic acid. Some of the optional substituents for alkyl are hydroxy, halogen exemplified by chloro and bromo, acyl exemplified by methylcarbonyl; alkoxy, and heterocyclyl exemplified by morpholino and piperidino. Other alkyl substituents as described herein may further be contemplated. The term “substituted alkenyl” refers to an alkenyl group in which 1 or more (up to about 5, for example about 3) hydrogen atoms is replaced by a substituent independently selected from those listed above with respect to a substituted alkyl. Other alkenyl substituents as described herein may further be contemplated. As used herein, “heteroatom” refers to an atom of any element other than carbon or hydrogen and includes, for example, nitrogen, oxygen, silicon, sulfur, phosphorus, and selenium. As used herein, “aryl” refers to an aromatic monocyclic hydrocarbon ring system or a polycyclic ring system in which two or more aromatic hydrocarbon rings are fused (i.e., having a bond in common with) together or at least one aromatic monocyclic hydrocarbon ring is fused to one or more cycloalkyl and/or cycloheteroalkyl rings. An aryl group can have 6 to 24 carbon atoms in its ring system (e.g., C6-C24 aryl group), which can include multiple fused rings. In some embodiments, a polycyclic aryl group can have 8 to 24 carbon atoms. Any suitable ring position of the aryl group can be covalently linked to the defined chemical structure. Examples of aryl groups having only aromatic carbocyclic ring(s) include phenyl, 1-naphthyl (bicyclic), 2-naphthyl (bicyclic), anthracenyl (tricyclic), phenanthrenyl (tricyclic), pentacenyl (pentacyclic), and like groups. Examples of polycyclic ring systems in which at least one aromatic carbocyclic ring is fused to one or more cycloalkyl and/or cycloheteroalkyl rings include, among others, benzo derivatives of cyclopentane (i.e., an indanyl group, which is a 5,6-bicyclic cycloalkyl/aromatic ring system), cyclohexane (i.e., a tetrahydronaphthyl group, which is a 6,6-bicyclic cycloalkyl/aromatic ring system), imidazoline (i.e., a benzimidazolinyl group, which is a 5,6-bicyclic cycloheteroalkyl/aromatic ring system), and pyran (i.e., a chromenyl group, which is a 6,6-bicyclic cycloheteroalkyl/aromatic ring system). Other examples of aryl groups include benzodioxanyl, benzodioxolyl, chromanyl, indolinyl groups, and the like. In some embodiments, aryl groups can be substituted as described herein. In some embodiments, an aryl group can have one or more halogen substituents, and can be referred to as a “haloaryl” group. Perhaloaryl groups, i.e., aryl groups where all of the hydrogen atoms are replaced with halogen atoms (e.g., —C6F5), are included within the definition of “haloaryl”. In certain embodiments, an aryl group is substituted with another aryl group and can be referred to as a biaryl group. Each of the aryl groups in the biaryl group can be substituted as disclosed herein. As used herein, “heteroaryl” refers to an aromatic monocyclic ring system containing at least one ring heteroatom selected from oxygen (O), nitrogen (N), sulfur (S), silicon (Si), and selenium (Se) or a polycyclic ring system where at least one of the rings present in the ring system is aromatic and contains at least one ring heteroatom. Polycyclic heteroaryl groups include those having two or more heteroaryl rings fused together, as well as those having at least one monocyclic heteroaryl ring fused to one or more aromatic carbocyclic rings, non-aromatic carbocyclic rings, and/or non-aromatic cycloheteroalkyl rings. A heteroaryl group, as a whole, can have, for example, 5 to 24 ring atoms and contain 1-5 ring heteroatoms (i.e., 5-20 membered heteroaryl group). The heteroaryl group can be attached to the defined chemical structure at any heteroatom or carbon atom that results in a stable structure. Generally, heteroaryl rings do not contain O—O, S—S, or S—O bonds. However, one or more N or S atoms in a heteroaryl group can be oxidized (e.g., pyridine N-oxide thiophene S-oxide, thiophene S,S-dioxide). Examples of heteroaryl groups include, for example, the 5- or 6-membered monocyclic and 5-6 bicyclic ring systems shown below: where T is O, S, NH, N-alkyl, N-aryl, N-(arylalkyl) (e.g., N-benzyl), SiH2, SiH(alkyl), Si(alkyl)2, SiH(arylalkyl), Si(arylalkyl)2, or Si(alkyl)(arylalkyl). Examples of such heteroaryl rings include pyrrolyl, furyl, thienyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazolyl, tetrazolyl, pyrazolyl, imidazolyl, isothiazolyl, thiazolyl, thiadiazolyl, isoxazolyl, oxazolyl, oxadiazolyl, indolyl, isoindolyl, benzofuryl, benzothienyl, quinolyl, 2-methylquinolyl, isoquinolyl, quinoxalyl, quinazolyl, benzotriazolyl, benzimidazolyl, benzothiazolyl, benzisothiazolyl, benzisoxazolyl, benzoxadiazolyl, benzoxazolyl, cinnolinyl, 1H-indazolyl, 2H-indazolyl, indolizinyl, isobenzofuryl, naphthyridinyl, phthalazinyl, pteridinyl, purinyl, oxazolopyridinyl, thiazolopyridinyl, imidazopyridinyl, furopyridinyl, thienopyridinyl, pyridopyrimidinyl, pyridopyrazinyl, pyridopyrdazinyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl groups, and the like. Further examples of heteroaryl groups include 4,5,6,7-tetrahydroindolyl, tetrahydroquinolinyl, benzothienopyridinyl, benzofuropyridinyl groups, and the like. In some embodiments, heteroaryl groups can be substituted as described herein. The term “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, “optionally substituted alkyl” means either “alkyl” or “substituted alkyl,” as defined herein. It will be understood by those skilled in the art with respect to any chemical group containing one or more substituents that such groups are not intended to introduce any substitution or substitution patterns that are sterically impractical and/or physically non-feasible. The term “isomers” or “stereoisomers” as used herein relates to compounds that have identical molecular formulae but that differ in the arrangement of their atoms in space. Stereoisomers that are not mirror images of one another are termed “diastereoisomers” and stereoisomers that are non-superimposable mirror images are termed “enantiomers,” or sometimes optical isomers. A carbon atom bonded to four non-identical substituents is termed a “chiral center.” Certain compounds herein have one or more chiral centers and therefore may exist as either individual stereoisomers or as a mixture of stereoisomers. Configurations of stereoisomers that owe their existence to hindered rotation about double bonds are differentiated by their prefixes cis and trans (or Z and E), which indicate that the groups are on the same side (cis or Z) or on opposite sides (trans or E) of the double bond in the molecule according to the Cahn-Ingold-Prelog rules. All possible stereoisomers are contemplated herein as individual stereoisomers or as a mixture of stereoisomers. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which the presently described subject matter pertains. Where a range of values is provided, for example, concentration ranges, percentage ranges, or ratio ranges, it is understood that each intervening value, to the tenth of the unit of the lower limit, unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the described subject matter. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and such embodiments are also encompassed within the described subject matter, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the described subject matter. Throughout the application, descriptions of various embodiments use “comprising” language. However, it will be understood by one of skill in the art, that in some specific instances, an embodiment can alternatively be described using the language “consisting essentially of” or “consisting of”. “Subject” as used herein refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, and pet companion animals such as household pets and other domesticated animals such as, but not limited to, cattle, sheep, ferrets, swine, horses, poultry, rabbits, goats, dogs, cats and the like. “Patient” as used herein refers to a subject in need of treatment of a condition, disorder, or disease, such as an acute or chronic airway disorder or disease. For purposes of better understanding the present teachings and in no way limiting the scope of the teachings, unless otherwise indicated, all numbers expressing quantities, percentages or proportions, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term “about”. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. In the instant application the terms “benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one derivatives”, “tetracyclic aza-Podophyllotoxin analogues of benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one”, “aza-Podophyllotoxin analogue compounds”, “benzimidazofuropyrimidin-1-one derivatives”, and the like may be used interchangeably. Aza-Podophyllotoxin Analogue Compounds In one embodiment, the present subject matter may include aza-Podophyllotoxin analogue compounds having the Structure I: or a pharmaceutically acceptable salt, ester stereoisomer, or solvate thereof, wherein:represents a single or double bond;n is 1 or 2;W represents an oxygen or sulfur;X represents an oxygen, sulfur, CH2, or —NR7wherein R7represents a hydrogen atom or a linear or branched (C1-C6)alkyl group, a (C3-C9)cycloalkyl group, a (C1-C6)alkyl-aryl group, an aryl group, or a heteroaryl group;R1represents:a hydrogen atom,aryl,heteroaryl,(C3-C9) cycloalkyl group,linear or branched (C1-C6) alkyl group, optionally substituted by an aryl group, a heteroaryl group, a hydroxy group, a linear or branched (C1-C6) alkoxy group, a carboxylic acid group, a group of formula —CONR8R9or —NR8R9, wherein R8and R9, which may be the same or different, each independently represent a linear or branched (C1-C6)alkyl group optionally substituted by a hydroxy groupor an amino group (itself optionally substituted by one or two linear or branched (C1-C6) alkyl groups), or R8and R9, together with the nitrogen atom to which they are attached, form a nitrogen-containing heterocycle,an amino group optionally substituted by one or more aryl groups, heteroaryl groups, or linear or branched (C1-C6) alkyl groups or (C3-C6) cycloalkyl groups optionally substituted by a carboxylic acid group, or by a group of formula —CONR8R9or —NR8R9, or—OR10wherein R10represents a hydrogen atom, an aryl group, a heteroaryl group, or a linear or branched (C1-C6) alkyl group or a (C3-C6) cycloalkyl group optionally substituted by a carboxylic acid group, or by a group of formula —CONR8R9or —NR8R9;R2, R3, R4, and R5, which may be the same or different, each represent:a hydrogen atom,a halogen atom,a hydroxy group,a nitro group,a linear or branched (C1-C6) polyhaloalkyl group,a linear or branched (C1-C6) alkyl group,—OR10,an amino group,a substituted amino group optionally substituted by one or more aryl groups, heteroaryl groups, or linear or branched (C1-C6) alkyl groups or (C3-C6) cycloalkyl groups optionally substituted by a carboxylic acid group, by a carbonyl group, or by a group of formula —CONR8R9or —NR8R9,—OPO(OH)2,an acyl group,a carboxylic acid group,a sulfonyl group,a sulfonamide group, ora cyano group; andR6is a hydrogen atom, a linear or branched (C1-C6) alkyl group, a (C3-C6) cycloalkyl group, or an aryl or heteroaryl group optionally substituted in one or more positions with one or more of R2, R3, R4, and R5, each of which may be the same or different. In a further embodiment, the present subject matter relates to an aza-Podophyllotoxin analogue compound of formula I, wherein n is 1. In another embodiment, the present subject matter relates to compounds of formula I, wherein W is O. In yet another embodiment, the present subject matter relates to compounds of formula I, wherein X is S, NH, or O. In still yet another embodiment, the present subject matter relates to compounds of formula I, wherein R1is H. In a further embodiment, the present subject matter relates to compounds of formula I, wherein R2, R3, R4, and R5are each H. In another embodiment, the present subject matter relates to a compound of formula I, wherein R6is phenyl or benzo[d][1,3]dioxolyl, each of which may be optionally substituted with one or more substituents independently selected from the group consisting of hydroxy, methoxy, methyl, N(CH3)2, NH2, —NHCOCH3, fluorine, bromine, and chlorine. In one embodiment in this regard, R6is phenyl substituted by 1, 2, or 3 methoxy groups. In another embodiment in this regard, R6is phenyl substituted by 1 or 2 hydroxy groups, or by one hydroxy group and one methyl group. In an embodiment, the present subject matter relates to a compound selected from the group consisting of:11-(4-methoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (1),11-(3,4-dimethoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (2),11-(3,4,5-trimethoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (3),11-(benzo[d][1,3]dioxol-5-yl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (4),11-(3,4-dihydroxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (5),11-(4-hydroxy-3-methoxyphenyl)-4,11-dihydro-1H,3H benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (6),11-(p-tolyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (7),11-(4-(dimethylamino)phenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (8),11-(4-aminophenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (9),N-(4-(1-oxo-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-11-yl)phenyl)acetamide (10),11-(3,4,5-trimethoxyphenyl)-2,3,4,11-tetrahydro-1H-benzo[4,5]imidazo[1,2-a]pyrrolo[3,4-d]pyrimidin-1-one (11),11-(3,4,5-trimethoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]thieno[3,4-d]pyrimidin-1-one (12),11-(4-fluorophenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (13),11-(4-chlorophenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (14),11-(4-bromophenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (15), and a pharmaceutically acceptable salt, ester stereoisomer, or solvate thereof. In the present subject matter, the structures of the above compounds are illustrated as follows: and a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof. It is to be understood that the present subject matter covers all combinations of substituent groups referred to herein. The present compounds may contain, e.g., when isolated in crystalline form, varying amounts of solvents. Accordingly, the present subject matter includes all solvates of the present compounds of formula I and pharmaceutically acceptable stereoisomers, esters, and/or salts thereof. Hydrates are one example of such solvates. Further, the present subject matter includes all mixtures of possible stereoisomers of the embodied compounds, independent of the ratio, including the racemates. Salts of the present compounds, or the salts of the stereoisomers thereof, include all inorganic and organic acid addition salts and salts with bases, especially all pharmaceutically acceptable inorganic and organic acid addition salts and salts with bases, particularly all pharmaceutically acceptable inorganic and organic acid addition salts and salts with bases customarily used in pharmacy. Examples of acid addition salts include, but are not limited to, hydrochlorides, hydrobromides, phosphates, nitrates, sulfates, acetates, trifluoroacetates, citrates, D-gluconates, benzoates, 2-(4-hydroxy-benzoyl)benzoates, butyrates, sub salicylates, maleates, laurates, malates, lactates, fumarates, succinates, oxalates, tartrates, stearates, benzenesulfonates (besilates), toluenesulfonates (tosilates), methanesulfonates (mesilates) and 3-hydroxy-2-naphthoates. Examples of salts with bases include, but are not limited to, lithium, sodium, potassium, calcium, aluminum, magnesium, titanium, ammonium, meglumine and guanidinium salts. The salts include water-insoluble and, particularly, water-soluble salts. The present compounds, the salts, the stereoisomers and the salts of the stereoisomers thereof may contain, e.g., when isolated in crystalline form, varying amounts of solvents. Included within the present scope are, therefore, all solvates of the compounds of formula I, as well as the solvates of the salts, the stereoisomers and the salts of the stereoisomers of the compounds of formula I. The present compounds may be isolated and purified in a manner known per se, e.g., by distilling off the solvent in vacuo and recrystallizing the residue obtained from a suitable solvent or subjecting it to one of the customary purification methods, such as column chromatography on a suitable support material. Salts of the compounds of formula I and the stereoisomers thereof can be obtained by dissolving the free compound in a suitable solvent (by way of non-limiting example, a ketone such as acetone, methylethylketone or methylisobutylketone; an ether such as diethyl ether, tetrahydrofurane or dioxane; a chlorinated hydrocarbon such as methylene chloride or chloroform; a low molecular weight aliphatic alcohol such as methanol, ethanol or isopropanol; a low molecular weight aliphatic ester such as ethyl acetate or isopropyl acetate; or water) which contains the desired acid or base, or to which the desired acid or base is then added. The acid or base can be employed in salt preparation, depending on whether a mono- or polybasic acid or base is concerned and depending on which salt is desired, in an equimolar quantitative ratio or one differing therefrom. The salts are obtained by filtering, reprecipitating, precipitating with a non-solvent for the salt or by evaporating the solvent. Salts obtained can be converted into the free compounds which, in turn, can be converted into salts. In this manner, pharmaceutically unacceptable salts, which can be obtained, for example, as process products in the manufacturing on an industrial scale, can be converted into pharmaceutically acceptable salts by processes known to the person skilled in the art. Pure diastereomers and pure enantiomers of the present compounds can be obtained, e.g., by asymmetric synthesis, by using chiral starting compounds in synthesis and by splitting up enantiomeric and diastereomeric mixtures obtained in synthesis. Preferably, the pure diastereomeric and pure enantiomeric compounds are obtained by using chiral starting compounds in synthesis. Enantiomeric and diastereomeric mixtures can be split up into the pure enantiomers and pure diastereomers by methods known to a person skilled in the art. Preferably, diastereomeric mixtures are separated by crystallization, in particular fractional crystallization, or chromatography. Enantiomeric mixtures can be separated, e.g., by forming diastereomers with a chiral auxiliary agent, resolving the diastereomers obtained and removing the chiral auxiliary agent. As chiral auxiliary agents, for example, chiral acids can be used to separate enantiomeric bases and chiral bases can be used to separate enantiomeric acids via formation of diastereomeric salts. Furthermore, diastereomeric derivatives such as diastereomeric esters can be formed from enantiomeric mixtures of alcohols or enantiomeric mixtures of acids, respectively, using chiral acids or chiral alcohols, respectively, as chiral auxiliary agents. Additionally, diastereomeric complexes or diastereomeric clathrates may be used for separating enantiomeric mixtures. Alternatively, enantiomeric mixtures can be split up using chiral separating columns in chromatography. Another suitable method for the isolation of enantiomers is enzymatic separation. Synthesis: In one embodiment, the present subject matter further relates to a process for synthesizing the aza-podophyllotoxin analogue compounds of formula/structure I, the process comprising the following general reaction scheme: The synthesis of the derivatives of the current subject matter of structure I relies on a one pot multiple component reaction by using diverse aldehydes, 2-aminobenzimidazole and activated methylene derivative such as tetronic acid. Pharmaceutical Compositions: In another embodiment, the present subject matter is directed to pharmaceutical compositions comprising a therapeutically effective amount of the compounds as described herein together with one or more pharmaceutically acceptable carriers, excipients, or vehicles. In some embodiments, the present compositions can be used for combination therapy, where other therapeutic and/or prophylactic ingredients can be included therein. The present subject matter further relates to a pharmaceutical composition, which comprises at least one of the present compounds together with at least one pharmaceutically acceptable auxiliary. In an embodiment, the pharmaceutical composition comprises one or two of the present compounds, or one of the present compounds. Non-limiting examples of suitable excipients, carriers, or vehicles useful herein include liquids such as water, saline, glycerol, polyethylene glycol, hyaluronic acid, ethanol, and the like. Suitable excipients for nonliquid formulations are also known to those of skill in the art. A thorough discussion of pharmaceutically acceptable excipients and salts useful herein is available in Remington's Pharmaceutical Sciences, 18th Edition. Easton, Pa., Mack Publishing Company, 1990, the entire contents of which are incorporated by reference herein. The present compounds are typically administered at a therapeutically or pharmaceutically effective dosage, e.g., a dosage sufficient to provide treatment for an acute or chronic airway disease or disorder. Administration of the compounds or pharmaceutical compositions thereof can be by any method that delivers the compounds systemically and/or locally. These methods include oral routes, parenteral routes, intraduodenal routes, and the like. While human dosage levels have yet to be optimized for the present compounds, generally, a daily dose is from about 0.01 to 10.0 mg/kg of body weight, for example about 0.1 to 5.0 mg/kg of body weight. The precise effective amount will vary from subject to subject and will depend upon the species, age, the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. The subject may be administered as many doses as is required to reduce and/or alleviate the signs, symptoms, or causes of the disease or disorder in question, or bring about any other desired alteration of a biological system. In employing the present compounds for treatment of a bacterial infection, any pharmaceutically acceptable mode of administration can be used with other pharmaceutically acceptable excipients, including solid, semi-solid, liquid or aerosol dosage forms, such as, for example, tablets, capsules, powders, liquids, suspensions, suppositories, aerosols or the like. The present compounds can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, and the like, for the prolonged administration of the compound at a predetermined rate, preferably in unit dosage forms suitable for single administration of precise dosages. The present compounds may also be administered as compositions prepared as foods for foods or animals, including medical foods, functional food, special nutrition foods and dietary supplements. A “medical food” is a product prescribed by a physician that is intended for the specific dietary management of a disorder or health condition for which distinctive nutritional requirements exist and may include formulations fed through a feeding tube (referred to as enteral administration or gavage administration). A “dietary supplement” shall mean a product that is intended to supplement the human diet and may be provided in the form of a pill, capsule, tablet, or like formulation. By way of non-limiting example, a dietary supplement may include one or more of the following dietary ingredients: vitamins, minerals, herbs, botanicals, amino acids, and dietary substances intended to supplement the diet by increasing total dietary intake, or a concentrate, metabolite, constituent, extract, or combinations of these ingredients, not intended as a conventional food or as the sole item of a meal or diet. Dietary supplements may also be incorporated into foodstuffs, such as functional foods designed to promote control of glucose levels. A “functional food” is an ordinary food that has one or more components or ingredients incorporated into it to give a specific medical or physiological benefit, other than a purely nutritional effect. “Special nutrition food” means ingredients designed for a particular diet related to conditions or to support treatment of nutritional deficiencies. Generally, depending on the intended mode of administration, the pharmaceutically acceptable composition will contain about 0.1% to 90%, for example about 0.5% to 50%, by weight of a compound or salt of the present compounds, the remainder being suitable pharmaceutical excipients, carriers, etc. One manner of administration for the conditions detailed above is oral, using a convenient daily dosage regimen which can be adjusted according to the degree of affliction. For such oral administration, a pharmaceutically acceptable, non-toxic composition is formed by the incorporation of any of the normally employed excipients, such as, for example, mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, sodium crosscarmellose, glucose, gelatin, sucrose, magnesium carbonate, and the like. Such compositions take the form of solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations and the like. The present compositions may take the form of a pill or tablet and thus the composition may contain, along with the active ingredient, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose and derivatives thereof, and the like. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc. an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, for example, sodium acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine acetate, triethanolamine oleate, etc. For oral administration, a pharmaceutically acceptable non-toxic composition may be formed by the incorporation of any normally employed excipients, such as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, cellulose derivatives, sodium crosscarmellose, glucose, sucrose, magnesium carbonate, sodium saccharin, talcum and the like. Such compositions take the form of solutions, suspensions, tablets, capsules, powders, sustained release formulations and the like. For a solid dosage form, a solution or suspension in, for example, propylene carbonate, vegetable oils or triglycerides, may be encapsulated in a gelatin capsule. Such diester solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545, the contents of each of which are incorporated herein by reference. For a liquid dosage form, the solution, e.g., in a polyethylene glycol, may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be easily measured for administration. Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and the like, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in U.S. Pat. Nos. Re. 28,819 and 4,358,603, the contents of each of which are hereby incorporated by reference. Another manner of administration is parenteral administration, generally characterized by injection, either subcutaneously, intramuscularly or intravenously. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like. In addition, if desired, the pharmaceutical compositions to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, solubility enhancers, and the like, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate, cyclodextrins, etc. Another approach for parenteral administration employs the implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject. However, percentages of active ingredient of 0.01% to 10% in solution are employable and will be higher if the composition is a solid which will be subsequently diluted to the above percentages. The composition may comprise 0.2% to 2% of the active agent in solution. Nasal solutions of the active compound alone or in combination with other pharmaceutically acceptable excipients can also be administered. Formulations of the active compound or a salt may also be administered to the respiratory tract as an aerosol or solution for a nebulizer, or as a microtine powder for insufflation, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation have diameters of less than 50 microns, for example less than 10 microns. Methods of Use: The present compounds have valuable pharmaceutical properties, which make them commercially utilizable. Accordingly, the present subject matter further relates to use of the present compounds for the treatment of diseases such as cancers. In another embodiment of the present subject matter, the aforementioned compound derivatives demonstrated in vitro anticancer action against human cancer cell lines comprising MCF-7 (breast cancer), A-549 (lung cancer), and HL-60 (leukemia). Accordingly, the present subject matter relates to methods of treating a cancer in a patient by administering one or more of the compounds presented herein to a patient in need thereof. In certain embodiments, the cancer is breast cancer, lung cancer, or leukemia. In another embodiment of the present subject matter, the concentration of the present compounds engaged for in vitro study against lung cancer cell lines for IC50 was in the range of about 0.058 to 0.211 μM at an exposure period of at least 48 hrs. In another embodiment of the present subject matter, the concentration of the present compounds engaged for in vitro study against breast cancer cell lines for IC50 was in the range of about 0.045 to 0.374 μM at an exposure period of at least 48 hrs. In another embodiment of the present subject matter, the concentration of the present compounds engaged for in vitro study against leukemia cell lines for IC50 was in the range of 0.061 to 0.281 μM at an exposure period of at least 48 hrs. Pharmacological Activity The cytotoxicity activity of the compounds of the current subject matter was assessed against three cancer cell lines such as A-549 (lung cancer), MCF-7 (breast cancer) and HL-60 (leukemia) cancer cells. The biological results, reported in Table 1, demonstrated that all the derivatives of Formula I displayed potent in vitro anti-proliferative activity with nanomolar concentration on all cancer cell lines ranging from 45 nM to 374 nM. By way of example, the compound of Example 13 showed one of the most potent cytotoxic derivatives in the series with IC50(concentration of cytotoxic agent which inhibits proliferation of the treated cells by 50%) of 71 nM, 45 nM and 61 nM against A-549, MCF-7 and HL-60 cancer cells respectively. In conclusion, the benzimidazofuropyrimidin-1-one derivatives of the current subject matter showing highly potent cytotoxic agents may find valuable application in treating a variety of cancers either as a sole active agent or in combination with other active ingredients. In Vitro Cytotoxic Activity Assay Compounds 1-15 were screened for their in vitro cytotoxic behavior utilizing a 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay against selected cancer human cell lines consisting of MCF-7 (human breast cancer), A-549 (lung cancer) and HL-60 (leukemia) (T. Mosmann, J. Immunol. Meth., 1983, 65, 55-63). The cells were cultured at 37° C. in RMPI1640 medium supplemented with 10% fetal bovine serum, 50 IU/mL penicillin, and 50 μg/mL streptomycin in a 5% CO2 incubator. All cells were sub-cultured 3 times/week by trypsinization. Viable cells were seeded and allowed to adhere for 12 h before a test drug was added in 96-well plates at an initial density of 1.0×105 cells/mL. Tumour cell lines were separately exposed to various concentrations of the tested compounds followed by incubation at a temperature of 37° C. during 24 h inside a medium of fresh RMPI 1640. Cells were subsequently incubated at 37° C. using MTT at 0.5 mg/mL during 4 h. After removal of supernatant, formazan crystals were dissolved in isopropanol and the optical density was measured at 570 nm. Etopside and Cisplatin were used as a positive control. The results are summarized with Etopside and Cisplatin as standard drugs in Table 1. TABLE 1In vitro cytotoxic activity (IC50, μM) ofbenzimidazofuropyrimidinones 1-15Anti-proliferative activity: IC50(μM)EntryA-549MCF-7HL-6010.0920.1260.08220.1200.1040.09630.0810.1150.12840.1040.0970.08450.1510.1540.17860.0950.1050.13370.0670.0690.07280.0920.0870.12990.1230.1620.143100.2110.3740.281110.0860.1250.104120.0750.1110.085130.0710.0450.061140.0580.0740.069150.1050.0810.092Etoposide11.9232.810.31Cisplatin9.2515.861.16aCancer cell lines: A-549 (lung cancer), MCF-7 (breast cancer) and HL-60 (leukemia). The biological results demonstrated that the compounds of Formula I displayed potent in vitro anti-proliferative activity against cancer cell lines as compared to the control drugs. The following examples relate to various methods of manufacturing certain specific compounds as described herein. EXAMPLES Example 1 11-(4-methoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (1) To a mixture of 2-aminobenzimidazole (1 mmol), 4-methoxybenzaldehyde (1.2 mmol), tetronic acid (1.2 mmol) and 10% L-proline (10 mg) in 5 mL ethanol were heated at reflux for 1 hour. After cooling at room temperature, the precipitate obtained was filtered off, washed with ethanol and then recrystallized to yield the expected product. Elemental Analysis: Calculated: C, 68.46; H, 4.54; N, 12.61. Found: C, 68.35; H, 4.66; N, 12.72. Example 2 11-(3,4-dimethoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (2) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 3,4-dimethoxybenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 66.11; H, 4.72; N, 11.56. Found C, 66.04; H, 4.77; N, 11.48. Example 3 11-(3,4,5-trimethoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (3) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 3,4,5-trimethoxybenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 64.12; H, 4.87; N, 10.68. Found C, 64.31; H, 4.78; N, 10.55. Example 4 11-(benzo[d][1,3]dioxol-5-yl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (4) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, piperonal and tetronic acid. Elemental Analysis: Calculated C, 65.70; H, 3.77; N, 12.10. Found C, 65.89; H, 3.75; N, 12.02. Example 5 11-(3,4-dihydroxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (5) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 3,4-dihydroxybenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 64.48; H, 3.91; N, 12.53. Found C, 64.29; H, 3.82; N, 12.61. Example 6 11-(4-hydroxy-3-methoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (6) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 4-hydroxy-3-methoxybenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 65.32; H, 4.33; N, 12.03. Found C, 65.57; H, 4.29; N, 11.88. Example 7 11-(p-tolyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (7) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, p-tolualdehyde and tetronic acid. Elemental Analysis: Calculated C, 71.91; H, 4.76; N, 13.24. Found C, 70.03; H, 4.61; N, 13.35. Example 8 11-(4-(dimethylamino)phenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (8) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 4-dimethylaminobenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 69.35; H, 5.24; N, 16.17. Found C, 69.06; H, 5.18; N, 16.09. Example 9 11-(4-aminophenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (9) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 4-aminobenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 67.92; H, 4.43; N, 17.60. Found C, 68.15; H, 4.37; N, 17.66. Example 10 N-(4-(1-oxo-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-11-yl)phenyl)acetamide (10) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, N-(4-formylphenyl)acetamide and tetronic acid. Elemental Analysis: Calculated C, 66.66; H, 4.48; N, 15.55. Found C, 66.48; H, 4.39; N, 15.61. Example 11 11-(3,4,5-trimethoxyphenyl)-2,3,4,11-tetrahydro-1H-benzo[4,5]imidazo[1,2-a]pyrrolo[3,4-d]pyrimidin-1-one (11) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 3,4,5-trimethoxybenzaldehyde and pyrrolidine-2,4-dione. Elemental Analysis: Calculated C, 64.28; H, 5.14; N, 14.28. Found C, 64.19; H, 5.06; N, 14.32. Example 12 11-(3,4,5-trimethoxyphenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]thieno[3,4-d]pyrimidin-1-one (12) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 3,4,5-trimethoxybenzaldehyde and thiotetronic acid. Elemental Analysis: Calculated C, 61.60; H, 4.68; N, 10.26. Found C, 61.48; H, 4.73; N, 10.31. Example 13 11-(4-fluorophenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (13) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 4-fluorobenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 67.29; H, 3.76; N, 13.08. Found C, 67.44; H, 3.64; N, 13.12. Example 14 11-(4-chlorophenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (14) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 4-chlorobenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 64.01; H, 3.58; N, 12.44. Found C, 63.87; H, 3.52; N, 12.53. Example 15 11-(4-bromophenyl)-4,11-dihydro-1H,3H-benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one (15) The expected product is obtained in accordance with the compound described in Example 1, starting from 2-aminobenzimidazole, 4-bromobenzaldehyde and tetronic acid. Elemental Analysis: Calculated C, 56.56; H, 3.16; N, 10.99. Found C, 56.39; H, 3.28; N, 11.14. It is to be understood that the tetracyclic aza-Podophyllotoxin analogues, for example, benzo[4,5]imidazo[1,2-a]furo[3,4-d]pyrimidin-1-one compounds, use/application, method of making, and properties of said compounds are not limited to the specific embodiments or examples described above, but encompasses any and all embodiments within the scope of the generic language of the following claims enabled by the embodiments described herein, or otherwise shown in the drawings or described above in terms sufficient to enable one of ordinary skill in the art to make and use the instant subject matter compounds.
47,436
11858894
DETAILED DESCRIPTION OF THE EMBODIMENTS Below, preferred embodiments will be provided in order to assist in the understanding of the present disclosure. However, these examples are provided only for illustration of the present invention, and should not be construed as limiting the present invention to these examples. Preparation Example 1: Preparation of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine (free base) Step 1-1) Preparation of 2-(2,4-difluorophenyl)-2-((3-methoxy-2-(methoxycarbonyl)-3-oxoprop-1-en-1-yl)amino)acetic acid 2,4-Difluorophenyl glycine (150.0 g, 801.5 mmol), dimethyl 2-(methoxymethylene)malonate (126.9 g, 728.6 mmol) and sodium acetate (65.8 g, 801.5 mmol) were added to methanol (800.0 ml), and the mixture was then refluxed at 60° C. for 4 hours. The reaction mixture was cooled to room temperature and then concentrated under reduced pressure to remove about 70% of methanol, and then filtered. The obtained solid was dried under reduced pressure to give 190.0 g of the title compound. (Yield: 79.2%). 1H-NMR (500 MHz, CDCl3): 8.02-7.99 (m, 1H), 7.45-7.40 (m, 1H), 7.00-6.95 (m, 2H), 5.16 (s, 1H), 3.74 (s, 3H), 3.76 (s, 3H) Step 1-2) Preparation of methyl 5-(2,4-difluorophenyl)-4-hydroxy-1H-pyrrol-3-carboxylate Acetic anhydride (1731.2 ml) and triethylamine (577.1 ml) were added to 2-(2,4-difluorophenyl)-2-((3-methoxy-2-(methoxycarbonyl)-3-oxoprop-1-en-1-yl)amino)acetic acid (190.0 g, 577.1 mmol) prepared in the step 1-1. The reaction mixture was refluxed at 140° C. for 30 minutes and then cooled to 0° C. To the reaction mixture, ice water (577.1 ml) was added at 0° C., stirred at room temperature for 1 hours and then extracted with ethyl acetate. The obtained extract was dried over anhydrous magnesium sulfate, and concentrated under reduced pressure. The resulting compound was filtered using silica gel to remove solids, and then concentrated under reduced pressure. Tetrahydrofuran (140.0 ml) and water (120.0 ml) were added to the resulting residue, and the mixture was cooled at 0° C. and sodium hydroxide (46.17 g, 1154.2 mmol) was then added thereto. The reaction mixture was stirred at 0° C. for 30 minutes, neutralized with 1N aqueous hydrochloric acid solution and then extracted with ethyl acetate. The obtained extract was dried over anhydrous magnesium sulfate, and then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (ethyl acetate:n-hexane=1:4 (v/v)) to give 22.0 g of the title compound. (Yield: 15.1%). 1H-NMR (500 MHz, CDCl3): 8.80 (s, 1H), 8.17-8.12 (m, 2H), 7.13 (d, 1H), 6.95 (t, 1H), 6.86-6.83 (m, 1H), 3.88 (s, 3H) Step 1-3) Preparation of methyl 5-(2,4-difluorophenyl)-4-methoxy-1H-pyrrol-3-carboxylate Methyl 5-(2,4-difluorophenyl)-4-hydroxy-1H-pyrrol-3-carboxylate (22.0 g, 86.9 mmol) prepared in the step 1-2 was dissolved in tetrahydrofuran (434.5 ml) and methanol (173.9 ml). To the reaction mixture, (trimethylsilyl)diazomethane (2.0 M diethyl ether solution, 173.8 ml) was added, and stirred at room temperature for 48 hours. Water was added to the reaction mixture and extracted with ethyl acetate. The obtained extract was dried over anhydrous magnesium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (ethyl acetate:n-hexane=1:4 (v/v)) to give 18.1 g of the title compound. (Yield: 75.3%) 1H-NMR (500 MHz, CDCl3): 8.78 (s, 1H), 8.12 (m, 1H), 7.30 (d, 1H), 6.95 (t, 1H), 6.88 (t, 1H), 3.87 (s, 3H), 3.85 (s, 3H) Step 1-4) Preparation of methyl 5-(2,4-difluorophenyl)-4-methoxy-1-((3-fluorophenyl)sulfonyl)-IH-pyrrol-3-carboxylate Methyl 5-(2,4-difluorophenyl)-4-methoxy-1H-pyrrol-3-carboxylate (18.0 g, 67.4 mmol) prepared in the step 1-3 was dissolved in dimethylformamide (335.0 ml). To the obtained solution, sodium hydride (60%, dispersion in liquid paraffin) (4.0 g, 101.0 mmol) was added at room temperature and the mixture was stirred at room temperature for 10 minutes. To the reaction mixture, 3-fluorobenzenesulfonyl chloride (13.37 ml, 101.0 mmol) was added, and the mixture was stirred at room temperature for 1 hour. Water was added to the reaction mixture and extracted with ethyl acetate. The obtained extract was dried over anhydrous magnesium sulfate, and then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (ethyl acetate:n-hexane=1:4 (v/v)) to give the title compound (26.1 g). (Yield: 91.1%). 1H-NMR (500 MHz, CDCl3): 7.98 (s, 1H), 7.43-7.39 (m, 1H), 7.30 (t, 1H), 7.23 (d, 1H), 7.15 (q, 1H), 7.67 (q, 1H), 6.91 (t, 1H), 6.77 (t, 1H), 3.87 (s, 3H), 3.61 (s, 3H) Step 1-5) Preparation of 5-(2,4-difluorophenyl)-4-methoxy-1-((3-fluorophenyl)sulfonyl)-IH-pyrrol-3-carbaldehyde Methyl 5-(2,4-difluorophenyl)-4-methoxy-1-((3-fluorophenyl)sulfonyl)-IH-pyrrol-3-carboxylate (26.0 g, 61.1 mmol) prepared in the step 1-4 was dissolved in tetrahydrofuran (300.0 ml). Diisobutyl aluminum hydride (1.0 M tetrahydrofuran solution) (183.4 ml, 183.4 mmol) was added to the obtained solution at 0° C., and the mixture was stirred at room temperature for 1 hour, neutralized with 1N hydrochloric acid solution and then extracted with ethylacetate. The obtained extract was dried over anhydrous magnesium sulfate, and then concentrated under reduced pressure. The resulting residue was dissolved in dichloromethane (300.0 ml), and then celite (26.0 g) and pyridinium chlorochromate (39.5 g, 183.4 mmol) were added thereto. The reaction mixture was stirred at room temperature for 1 hour and then filtered to remove a solid, and the obtained filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (ethyl acetate:n-hexane=1:2 (v/v)) to give the title compound (17.2 g). (Yield: 70.9%). 1H-NMR (500 MHz, CDCl3): 9.89 (s, 1H), 7.99 (s, 1H), 7.45-7.41 (m, 1H), 7.33 (s, 1H), 7.25 (d, 1H), 7.18 (q, 1H), 7.05 (s, 1H), 6.92 (t, 1H), 6.77 (t, 1H), 3.63 (s, 3H) Step 1-6) Preparation of 1-(5-(2,4-difluorophenyl)-4-methoxy-1-((3-fluorophenyl)sulfonyl)-IH-pyrrol-3-yl)-N-methylmethanamine 5-(2,4-difluorophenyl)-4-methoxy-1-((3-fluorophenyl)sulfonyl)-IH-pyrrol-3-carbaldehyde (17.0 g, 43.0 mmol) prepared in the step 1-5 was dissolved in methanol (430.0 ml). Methylamine (9.8 M methanol solution) (87.8 ml, 860.0 mmol) was added to the obtained solution, and the mixture was stirred at room temperature for 30 minutes. Sodium borohydride (16.3 g, 430.0 mmol) was added to the reaction mixture, and the mixture was stirred at room temperature for 30 minutes. Water was added to the reaction mixture and extracted with ethyl acetate. The obtained extract was dried over anhydrous magnesium sulfate, and then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (ethyl acetate:n-hexane=1:2 (v/v)) to give the title compound (15.2 g). (Yield: 86.1%). 1H-NMR (500 MHz, CDCl3): 7.39-7.35 (m, 1H), 7.26-7.20 (m, 2H), 7.15 (q, 1H), 7.06 (d, 1H), 6.87 (t, 1H), 6.78 (t, 1H), 3.60 (d, 2H), 3.44 (s, 3H), 2.45 (s, 3H) Preparation Example 2: Preparation of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine (15.0 g, 36.6 mmol) prepared in Preparation Example 1 was dissolved in ethyl acetate (36.6 ml) to which hydrochloric acid solution (2.0 M diethyl ether solution) (36.6 ml, 73.1 mmol) was added. The reaction mixture was stirred at room temperature for 1 hour and then filtered, and the obtained solid was dried under reduced pressure to give the title compound (15.1 g). (Yield: 92.5%). Molecular weight 446.87 1H-NMR (500 MHz, MeOD): 7.69 (s, 1H), 7.58-7.53 (m, 1H), 7.45 (t, 1H), 7.30 (d, 1H), 7.20-7.15 (m, 2H), 7.02-6.94 (m, 2H), 4.07 (d, 2H), 3.46 (s, 3H), 2.71 (s, 3H) Hereinafter, in the following examples, 1-(5-(2,4-difluorophenyl)-4-methoxy-1-((3-fluorophenyl)sulfonyl)-IH-pyrrol-3-yl)-N-methylmethanamine (free base) prepared in Preparation Example 1 and 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine hydrochloride prepared in Preparation Example 2 were used. Example 1-1: Preparation of Crystalline Form I of Hydrochloride by an Evaporative Crystallization Method 300 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride was dissolved in 5 ml of ethanol to prepare a solution. Then, ethanol was evaporated from the prepared solution at room temperature for 1 day. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 250 mg of crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride. Example 1-2: Preparation of Crystalline Form I of Hydrochloride by a Drowning-Out Crystallization Method 300 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride was dissolved in 5 ml of ethanol to prepare a solution. Then, 5 ml of n-hexane was added to the prepared solution and stirred at 50 rpm at room temperature for 1 day. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 235 mg of crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride. Example 2: Preparation of Crystalline Form II of Hydrochloride by an Evaporative Crystallization Method 20 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride was dissolved in 1 ml of methanol to prepare a solution. Then, methanol was evaporated from the prepared solution at room temperature for 1 day. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 15 mg of crystalline form II of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride. Example 3-1: Preparation of Crystalline Form of Succinate by an Evaporative Crystallization Method 300 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine free base and 86.3 mg of succinic acid were dissolved in 5 ml of methanol to prepare a solution. Then, methanol was evaporated from the prepared solution at room temperature for 2 days. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 340 mg of crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine succinate. Example 3-2: Preparation of Crystalline Form of Succinate by a Drowning-Out Crystallization Method 300 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine free base and 86.3 mg of succinic acid were dissolved in 5 ml of methanol to prepare a solution. Then, 5 ml of n-hexane was added to the prepared solution and stirred at 50 rpm at room temperature for 4 hours. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 300 mg of crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine succinate. Example 4-1: Preparation of Crystalline Form of Tartrate by an Evaporative Crystallization Method 300 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine free base and 109.7 mg of tartaric acid were dissolved in 5 ml of methanol to prepare a solution. Then, methanol was evaporated from the prepared solution at room temperature for 2 days. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 385 mg of crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine tartrate. Example 4-2: Preparation of Crystalline Form of Tartrate by a Drowning-Out Crystallization Method 300 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine free base and 109.7 mg of tartaric acid were dissolved in 5 ml of ethanol to prepare a solution. Then, 5 ml of n-hexane was added to the prepared solution and stirred at 50 rpm at room temperature for 4 hours. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 340 mg of crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine tartrate. Example 5-1: Preparation of Crystalline Form I of Fumarate by an Evaporative Crystallization Method 300 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine free base and 84.8 mg of fumaric acid were dissolved in 5 ml of ethanol to prepare a solution. Then, ethanol was evaporated from the prepared solution at room temperature for 2 days. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 340 mg of crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine fumarate. Example 5-2: Preparation of Crystalline Form I of Fumarate by a Reactive Crystallization Method 300 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine free base was dissolved in 5 ml of ethanol, and 109.7 mg of fumaric acid was dissolved in 3 ml of ethanol to prepare respective solutions. Then, the prepared two solutions were mixed and stirred at 50 rpm for 2 hours at room temperature. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 314 mg of crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine fumarate. Example 6-1: Preparation of Crystalline Form II of Fumarate by a Solvent-Mediated Polymorphic Transition Method 300 mg of crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine fumarate was dissolved in 5 ml of ethanol to prepare a solution. Then, the prepared solution was stirred at 50 rpm at room temperature for 16 hours. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 250 mg of crystalline form II of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine fumarate Example 6-2: Preparation of Crystalline Form II of Fumarate by a Solid-State Polymorphic Transition Method 300 mg of crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine fumarate was dried under vacuum at a temperature of 50° C. for 24 hours. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 300 mg of crystalline form II of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine fumarate Comparative Example 1: Preparation of Crystalline Form of Free Base by a Cooling Crystallization Method 100 mg of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine free base was cooled at a low temperature of 4° C. for 2 weeks. After a crystal was produced, the crystal was separated by filtration under reduced pressure to obtain 100 mg of crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methyl methanamine free base. Test Example 1: Inhibitory Effects on Proton Pump (H+/K+-ATPase) Activity The inhibitory effects on proton pump (H+/K+-ATPase) activity of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride prepared in Preparation Example 2 were measured as follows. Gastric vesicles were prepared from a hog stomach according to a known method (Edd C. Rabon et al., Preparation of Gastric H+,K+-ATPase, Methods in enzymology, vol. 157 Academic Press Inc., (1988), pp. 649-654). The protein contents of gastric vesicles thus prepared were quantitatively measured with Bicinchoninic Acid (BCA) kit (Thermo). 80 μl of (a predetermined concentration of a test compound, 0.5% DMSO, 2.5 mM MgCl2, 12.5 mM KCl, 1.25 mM EDTA, 60 mM Tris-HCl, pH7.4) was added to each well of 96-well plates. 10 μl of a reaction solution containing gastric vesicles (60 mmol/l, Tris-HCl buffer, pH 7.4) and 10 μl of a Tris buffer solution containing adenosine triphosphate (10 mM ATP, Tris-HCl buffer solution, pH 7.4) were added to each well and subjected to enzymatic reaction at 37° C. for 40 minutes. 50 μl of malachite green solution (0.12% malachite green solution in 6.2 N sulfuric acid, 5.8% ammonium molybdate and 11% Tween 20 were mixed at a ratio of 100:67:2) was added thereto to stop the enzyme reaction, and 50 μl of 15.1% sodium citrate was added thereto. The amount of monophosphate (Pi) in the reaction solution was measured at 570 nm by using a microplate reader (FLUOstar Omega, BMG). The inhibition rate (%) was measured from the activity value of the control group and the activity value of the test compounds at various concentrations. The concentration (IC50) that inhibits H+/K+-ATPase activity by 50% was calculated from each % inhibition value of the compounds using Logistic 4-parameter function of Sigmaplot 8.0 program. As a result, 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride prepared in Preparation Example 2 exhibited an IC50value of 0.024 μM. Thus, a salt of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine according to one embodiment of the present invention had excellent proton pump inhibitory activity and thus can be used for a pharmaceutical composition for the prevention and treatment of gastrointestinal injury due to gastrointestinal tract ulcer, gastritis, reflux esophagitis, orH. pylori. Test Example 2: X-Ray Powder Diffraction Analysis X-ray powder diffraction analysis was performed for the crystalline forms prepared in the Examples and Comparative Examples, and the results were shown inFIGS.1to7. In this case, the X-ray powder diffraction analysis was carried out using a CuKα target in the range of diffraction angles (2θ) of 5° to 35° with an X-ray powder diffraction spectrometer (D8 Advance, Bruker) under conditions of a voltage of 45 kV, a current amount of 40 mA, a divergence and scattering slit of 1°, a light receiving slit of 0.2 mm, and a scanning speed of 3°/min (0.4 seconds/0.02° interval). Referring toFIG.1, it could be confirmed that the crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride prepared in Example 1-1 had peaks at diffraction angles (2θ) of 5.8°, 9.7°, 10.0°, 12.8°, 13.2°, 17.4°, 18.5°, 19.5°, 19.8°, 20.10, 21.8°, 25.9°, 26.5° and 28.2° in an X-ray powder diffraction pattern. Referring toFIG.2, it could be confirmed that the crystalline form II of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride prepared in Example 2 had peaks at diffraction angles (2θ) of 9.2°, 9.8°, 10.0°, 12.9°, 13.2°, 13.4°, 13.8°, 15.0°, 18.4°, 19.6° and 20.2° in an X-ray powder diffraction pattern. Referring toFIG.3, it could be confirmed that the crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine succinate prepared in Example 3-1 had peaks at diffraction angles (2θ) of 8.0°, 11.2°, 12.0°, 14.9°, 20.0°, 22.1° and 24.1° in an X-ray powder diffraction pattern. Referring toFIG.4, it could be confirmed that the crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine tartrate prepared in Example 4-1 had peaks at diffraction angles (2θ) of 11.7°, 13.0°, 13.5°, 14.5°, 18.3°, 19.5°, 20.3°, 21.5° and 23.5° in an X-ray powder diffraction pattern. Referring toFIG.5, it could be confirmed that the crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine fumarate prepared in Example 5-1 had peaks at diffraction angles (2θ) of 7.9°, 11.9°, 20.0° and 24.0° in an X-ray powder diffraction pattern. Referring toFIG.6, it could be confirmed that the crystalline form II of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine fumarate prepared in Example 6-1 had peaks at diffraction angles (2θ) of 8.4°, 10.5°, 18.3° and 19.02° in an X-ray powder diffraction pattern. Referring toFIG.7, it could be confirmed that the crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine free base prepared in Comparative Example 1 had peaks at diffraction angles (2θ) of 8.7°, 10.4°, 12.4°, 17.08°, 17.48°, 21.6°, 25.06°, 26.03°, 28.70° and 29.6° in an X-ray powder diffraction pattern. Test Example 3: Differential Scanning Calorimetry Analysis The differential scanning calorimetry analysis was carried out for the crystalline forms prepared in the Examples and Comparative Example and the results were shown inFIG.8toFIG.14. In this case, the differential scanning calorimetry analysis was carried out with raising the temperature from 200° C. to 300° C. at a scanning rate of 10° C./min under a nitrogen purification in a sealed pan using a differential scanning calorimeter (DSC Q20, TA Instruments Co., Ltd.). Referring toFIG.8, it could be confirmed that the crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride prepared in Example 1-1 had an endothermic initiation temperature of 215.02° C. and exhibited a maximum endothermic peak at an endothermic temperature of 217.11° C. in a differential scanning calorimetry analysis. Referring toFIG.9, it could be confirmed that the crystalline form II of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine hydrochloride prepared in Example 2 had an endothermic initiation temperature of 213.14° C. and exhibited a maximum endothermic peak at an endothermic temperature of 215.7° C. in a differential scanning calorimetry analysis. Referring toFIG.10, it could be confirmed that the crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine succinate prepared in Example 3-1 had an endothermic initiation temperature of 132.3° C. and exhibited a maximum endothermic peak at an endothermic temperature of 133.9° C. in a differential scanning calorimetry analysis. Referring toFIG.11, it could be confirmed that the crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine tartrate prepared in Example 4-1 had an endothermic initiation temperature of 146.34° C. and exhibited a maximum endothermic peak at an endothermic temperature of 148.27° C. in a differential scanning calorimetry analysis. Referring toFIG.12, it could be confirmed that the crystalline form I of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine fumarate prepared in Example 5-1 had an endothermic initiation temperature of 164.97° C. and exhibited a maximum endothermic peak at an endothermic temperature of 167.46° C. in a differential scanning calorimetry analysis. Referring toFIG.13, it could be confirmed that the crystalline form II of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine fumarate prepared in Example 6-1 had an endothermic initiation temperature of 179.47° C. and exhibited a maximum endothermic peak at an endothermic temperature of 189.05° C. in a differential scanning calorimetry analysis. Referring toFIG.14, it could be confirmed that the crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine free base prepared in Comparative Example 1 had an endothermic initiation temperature of 79.76° C. and exhibited a maximum endothermic peak at an endothermic temperature of 83.45° C. in a differential scanning calorimetry analysis. As can be seen fromFIGS.8to14, it could be confirmed that the crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine free base prepared in Comparative Example 1 had a lower endothermic initiation temperature and a lower endothermic temperature with the maximum endothermic peak, compared to the crystalline forms of the salts prepared in Examples. Thus, it was confirmed that the crystalline form of 1-(5-(2,4-difluorophenyl)-1-((3-fluorophenyl)sulfonyl)-4-methoxy-1H-pyrrol-3-yl)-N-methylmethanamine free base was not suitable for the production of pharmaceuticals due to its low melting point, while the crystalline forms of the salts according to the Examples were pharmaceutically applicable. Test Example 4: Hygroscopicity Test The hygroscopicity test was carried out for the crystalline forms prepared in the above Examples. First, 40 mg of the crystalline forms of the Examples were tightly sealed and stored in each glass desiccator containing a saturated aqueous solution of several salts for at least two days under the condition of constant relative humidity as shown in Table 1 below. Subsequently, the result of measurement of weight change for each of these crystalline forms showed that weight change due to moisture was not observed. Accordingly, it could be seen that the crystalline forms prepared in the Examples did not have hygroscopicity. TABLE 1RelativeDesiccatorhumidityTypes of salt-saturated aqueous solution133%MgCl2-saturated aqueous solution253%Mg(NO3)2•6H2O-saturated aqueous solution364%NaNO2-saturated aqueous solution475%NaCl-saturated aqueous solution593%KNO3-aqueous solution Test Example 5: Stability Confirmation Test The stability test was carried out for the crystalline forms prepared in the Examples to evaluate the degree to which impurities were formed during storage under severe conditions (moisture-proof condition and high-humidity exposure condition). The results of the stability test under the moisture-proof condition were shown in Table 2 below, and the results of the stability test under the high-humidity exposure condition were shown in Table 3 below. For the stability test, vials containing 10 mg of each sample which was precisely weighed and taken were prepared in the planned quantity, and they were stored by dividing into the moisture-proof condition (60° C. and less than 10% relative humidity) and under the high-humidity exposure condition (60° C. and 95% relative humidity). However, under the high-humidity exposure condition, a stopper of the vial was not used to keep so that the sample is in sufficient contact with a moisture in the air. At a fixed point of time after the initiation of the test, two vials per point of time were taken (number of samples per test n=2). 10 ml of methanol was added to each vial to dissolve the sample, which was then centrifuged. The resulting supernatant was analyzed using a liquid chromatography. The peak area was determined by integration for all detected peaks, and the relative peak area for the main component and the total impurity was calculated and expressed as an average value. TABLE 2InitialPeak areaPeak areaAfter 2 weeksAfter 4 weeksTypes ofof mainof totalPeak area ofPeak area ofPeak area ofPeak area ofcrystallinecomponentimpuritiesmain componenttotal impuritiesmain componenttotal impuritiesform(%)(%)(%)(%)(%)(%)ExampleCrystalline99.820.1899.800.1999.800.201-1form I ofhydrochlorideExampleCrystalline99.550.4599.610.3999.550.453-1form ofsuccinateExampleCrystalline99.520.4899.540.4699.480.524-1form oftartrateExampleCrystalline99.380.6299.360.6499.370.635-1form II offumarate TABLE 3After 1 weekInitialPeakAfter 2 weeksAfter 4 weeksPeak areaPeak area ofPeak areaarea ofPeak areaPeak areaPeak areaPeak areaof maintotalof maintotalof mainof totalof mainof totalType of crystallinecomponentimpuritiescomponentimpuritiescomponentimpuritiescomponentimpuritiesform(%)(%)(%)(%)(%)(%)(%)(%)ExampleCrystalline form I99.820.1899.810.1999.800.2099.800.201-1of hydrochlorideExampleCrystalline form of99.550.4599.560.4499.530.4799.470.543-1succinateExampleCrystalline form of99.520.4899.480.5299.430.5799.230.774-1tartrateExampleCrystalline form I99.380.6299.400.6099.320.6899.300.705-1of fumarate As shown in Tables 2 and 3, it could be confirmed that the crystalline forms prepared in the Examples did not show a decrease in the peak area of the main component and an increase in the peak area of the total impurities which were significant under the moisture-proof condition and the high-humidity exposure condition. Therefore, it was confirmed that the crystalline forms produced in the Examples suppressed an increase of impurities regardless of the influence of humidity under severe conditions and exhibited excellent chemical stability. Test Example 6: Solubility Test in Water The solubility test in water was carried out for the crystal form prepared in the Examples, and the results were shown in Table 4 below. For the solubility test in water, a sample of less than 10 mg was first precisely weighed and taken and placed into a vial, to which 50 μl of deionized water was added, shaking for 30 seconds and ultrasonic shaking for 1 minute were carried out, and these processes were repeated several times. The water solubility was calculated by measuring the amount of water used to dissolve all the samples. TABLE 4Solubility in waterType of crystalline form(mg/ml)Example 1-1Crystalline form I of11.11hydrochlorideExample 3-1Crystalline form of succinate7.20Example 4-1Crystalline form of tartrate6.90Example 5-1Crystalline form I of fumarate1.73-2.60ComparativeCrystalline form of free baseLess than 0.16Example 1 As shown in Table 4, it could be seen that the crystalline forms prepared in the Examples had a water solubility of 10 times or more as compared with that of the crystalline form of the free base prepared in Comparative Example 1. In addition, the crystalline forms prepared in the Examples showed high solubility in the order of crystalline form I of hydrochloride, crystalline form of succinate, crystalline form of tartrate and crystalline form I of fumarate.
31,017
11858895
The figures together with the following detailed description make apparent to those skilled in the art how the disclosure may be implemented in practice. DETAILED DESCRIPTION Various compositions, systems or processes will be described below to provide an example of an embodiment of each claimed subject matter. No embodiment described below limits any claimed subject matter and any claimed subject matter may cover processes, compositions or systems that differ from those described below. The claimed subject matter is not limited to compositions, processes or systems having all of the features of any one composition, system or process described below or to features common to multiple or all of the compositions, systems or processes described below. It is possible that a composition, system, or process described below is not an embodiment of any claimed subject matter. Any subject matter disclosed in a composition, system or process described below that is not claimed in this document may be the subject matter of another protective instrument, for example, a continuing patent application, and the applicant(s), inventor(s) or owner(s) do not intend to abandon, disclaim or dedicate to the public any such subject matter by its disclosure in this document. As used herein and in the claims, the singular forms, such “a”, “an” and “the” include the plural reference and vice versa unless the context clearly indicates otherwise. Throughout this specification, unless otherwise indicated, “comprise,” “comprises” and “comprising” are used inclusively rather than exclusively, so that a stated integer or group of integers may include one or more other non-stated integers or groups of integers. Various compositions, systems or processes will be described below to provide an example of an embodiment of each claimed subject matter. No embodiment described below limits any claimed subject matter and any claimed subject matter may cover processes, compositions or systems that differ from those described below. The claimed subject matter is not limited to compositions, processes or systems having all of the features of any one composition, system or process described below or to features common to multiple or all of the compositions, systems or processes described below. It is possible that a composition, system, or process described below is not an embodiment of any claimed subject matter. Any subject matter disclosed in a composition, system or process described below that is not claimed in this document may be the subject matter of another protective instrument, for example, a continuing patent application, and the applicant(s), inventor(s) or owner(s) do not intend to abandon, disclaim or dedicate to the public any such subject matter by its disclosure in this document. When ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and sub-combinations of ranges and specific embodiments therein are intended to be included. Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term “about.” The term “about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary between 1% and 15% of the stated number or numerical range, as will be readily recognized by context. Furthermore any range of values described herein is intended to specifically include the limiting values of the range, and any intermediate value or sub-range within the given range, and all such intermediate values and sub-ranges are individually and specifically disclosed (e.g., a range of 1 to 5 includes 1, 1.5, 2, 2.75, 3, 3.90, 4, and 5). Similarly, other terms of degree such as “substantially” and “approximately” as used herein mean a reasonable amount of deviation of the modified term such that the end result is not significantly changed. These terms of degree should be construed as including a deviation of the modified term if this deviation would not negate the meaning of the term it modifies. Unless otherwise defined, scientific and technical terms used in connection with the formulations described herein shall have the meanings that are commonly understood by those of ordinary skill in the art. The terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention, which is defined solely by the claims. All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference in its entirety. Terms and Definitions The term “psilocybin”, refers to a chemical compound having the structure set forth inFIG.1. The term “indole prototype structure” refers to the chemical structure shown inFIG.2. It is noted that specific carbon atoms and a nitrogen atom in the indole prototype structure are numbered. Reference may be made to these carbon and nitrogen numbers herein, for example C2, C4, N1, and so forth. Furthermore, reference may be made to chemical groups attached to the indole prototype structure in accordance with the same numbering, for example R4and R6reference chemical groups attached to the C4and C6atom, respectively. In addition, R3Aand R3B, in this respect, reference chemical groups extending from the ethyl-amino group extending in turn from the C3atom of the prototype indole structure. The term “aminated psilocybin derivative” refers to a psilocybin derivative compound to which an amino group has been bonded to psilocybin or a psilocybin derivative. The nitrogen of the amino group may bear 1-3 substituents (i.e., be a N-substituted amino group). N-substituents can be an alkyl, aryl, acyl, sulfonyl groups or combinations thereof. Reference may be made to specific carbon atoms which may be aminated. For example, a 5-amino-psilocybin derivative refers to a psilocybin derivative in which carbon atom number 5 (as identified in the indole prototype structure) possesses an amino or N-substituted amino group, or, similarly, 7-amino-psilocybin derivative refers to a psilocybin derivative in which carbon atom number 7 (as identified in the indole prototype structure) possess an amino or N-substituted amino group. Thus, for example, aminated psilocybin derivatives include, single amino derivatives, 2-amino, 4-amino, 5-amino, 6-amino, and 7-amino psilocybin derivatives, for example, and multiple amino derivatives, such as, for example, 5,7-di-amino psilocybin derivatives, and 2,5,7-tri-amino psilocybin derivatives. The term aminated psilocybin derivatives further includes chemical compounds having the chemical formula (I): wherein at least one of R2, R4, R5, R6, or R7is an amino group or an N-substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom or an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group, and wherein R3Aand R3Bare a hydrogen atom, an alkyl group, an aryl group, or an acyl group. Furthermore, it is noted that when R4is a phosphate group, the term aminated psilocybin derivatives includes compounds having the formula (XIX): wherein at least one of R2, R5, R6, or R7is a hydrogen atom, and wherein R2, R5, R6, or R7which are not a hydrogen atom are an amino or N-substituted amino group, and wherein R3Aand R3Bare a hydrogen atom, an alkyl group or aryl group. The term further includes salts of aminated psilocybins, such as a sodium salt, a potassium salt etc. The term “amino group” and “amino”, as used herein refers to a molecule containing one atom of nitrogen bonded to hydrogen atoms and having the formula —NH2. An amino group also may be protonated and having the formula —NH3+. An amino group through its nitrogen atom may be chemically bonded to another entity. Furthermore, it is noted that an entity attached to an amino group may be referred to herein as an “aminated” entity, e.g., an aminated psilocybin derivative is a psilocybin derivative possessing either an amino group or a N-substituted amino group. The term “N-substituted amino group”, as used herein, refers to an amino group wherein at least one of the hydrogen atoms has been substituted by another atom or group, such as, for example, an alkyl group, an acyl group, an aryl group a sulfonyl group etc., excluding, however, an amino group wherein both of the hydrogen atoms are substituted by oxygen atoms to thereby form a nitro group. An N-substituted amino group also may be protonated, and the amino group through its nitrogen atom may be chemically bonded to another entity. Thus, N-substituted amino group may be represented herein as: Furthermore N-substituted amino groups include:(i) chemical group (XX) (an alkyl group, an aryl group): wherein R′ and R″ are each independently selected from a hydrogen atom, an alkyl group, and an aryl group, provided however that at least one of R′, and R″ is not a hydrogen atom;(ii) chemical group (XXI): wherein R′, R″ and R′″ are each independently selected from a hydrogen atom, an alkyl group, and an aryl group, provided however that at least one of R′, R″, and R′″ is not a hydrogen atom;(iii) chemical group (XXII) (an acyl group): wherein R′ and R″ are each independently selected from a hydrogen atom, an alkyl group and an aryl group;(iv) chemical group (XXIII) (a sulfonyl group): wherein R′, and R″ are each independently selected from a hydrogen atom, an alkyl group and an aryl group; or(v) chemical group (XXIV) (a sulfonate group): wherein R″ is selected from a hydrogen atom, an alkyl group, and an aryl group. The nitrogen atom of chemical groups (XXII), (XXIII) and (XXIV) can also be positively charged and be further substituted with H, or R′″. It is noted that R′, R″ and R′″ can herein additionally include numerical subscripts, such as 5a, 6b, 7b etc., and be represented, for example, as R′5a, R″6bor R′″7a, respectively. Where such numerical values are included, they reference chemical entity extending from the amino group extending in turn from the thus numbered C atom of the prototype indole structure. Thus, for example, R′5ais a chemical entity extending from an aminated group attached to the C5atom of the indole ring structure, R′2ais a chemical entity extending from an aminated group attached to the C2atom of the indole ring structure, and so forth. Furthermore, it is noted that an entity attached to an N-substituted amino group may be referred to herein as an “aminated” entity, e.g., an aminated psilocybin derivative is a psilocybin derivative possessing either an amino group or a N-substituted amino group. The term “sulfonyl”, as used herein, refers to a molecule containing one sulfur atom bonded to two oxygen atoms, and one other entity and having the formula: wherein R may be a variety of entities including a hydroxy group, an alkyl group, or an aryl group. A sulfonyl group through its sulfur atom may be chemically bonded to another entity. Furthermore, it is noted that an entity attached to a sulfonyl group may be referred to herein as a “sulfonylated”. The term “phosphate group”, as used herein, is a molecule containing one atom of phosphorus, covalently bound to four oxygen atoms (three single bonds and one double bond). Of the four oxygen atoms, one oxygen atom may be a hydroxy, and one of the non-hydroxylated oxygen atom may be chemically bonded to another entity. The terms “hydroxy group”, and “hydroxy”, as used herein, refer to a molecule containing one atom of oxygen bonded to one atom of hydrogen, and having the formula —OH. A hydroxy through its oxygen atom may be chemically bonded to another entity. The terms “nitro” and “nitro group”, as used herein, refer to a molecule containing one atom of nitrogen bonded to two atoms of oxygen and having the formula —NO2. A nitro group through its nitrogen atom may be chemically bonded to another entity. Furthermore, it is noted that an entity attached to a nitro group may be referred to herein as a “nitrated” entity, e.g., a nitrated psilocybin derivative is a psilocybin derivative possessing a nitro group. The term “alkyl”, as used herein, refers to a straight and/or branched chain, saturated alkyl radical containing from one to “p” carbon atoms (“C1-Cp-alkyl”) and includes, depending on the identity of “p”, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, isobutyl, t-butyl, 2,2-dimethylbutyl, n-pentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, n-hexyl and the like, where the variable p is an integer representing the largest number of carbon atoms in the alkyl radical Alkyl groups further include hydrocarbon groups arranged in a chain having the chemical formula —CnH2n+1, including, without limitation, methyl groups (—CH3), ethyl groups (—C2H5), propyl groups (—C3H7), and butyl groups (—C4H9). The term “O-alkyl”, as used herein, refers to a hydrocarbon group arranged in a chain having the chemical formula —O—CnH2n+1. O-alkyl groups include, without limitation, O-methyl groups (—O—CH3), O-ethyl groups (—O—C2H5), O-propyl groups (—O—C3H7) and O-butyl groups (—O—C4H9). The term “aryl”, as used herein, refers to a monocyclic, bicyclic, or tricyclic aromatic ring system containing, depending on the number of atoms in the rings, for example, from 6 to 14 carbon atoms (C6-C14-aryl) or from 6 to 10 carbons (C6-C10-aryl), and at least 1 aromatic ring and includes phenyl, naphthyl, anthracenyl, 1,2-dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, fluorenyl, phenanthrenyl, biphenylenyl, indanyl, indenyl and the like. The term “acyl”, as used herein, refers to a carbon atom double bonded to an oxygen and single bonded to an alkyl group. The carbon atom further can be bonded to another entity. An acyl group can be described by the chemical formula: —C(═O)—CnH2n+1. The term “O-acyl group” refers to an acyl group in which the carbon atom is single bonded to an additional oxygen atom. The additional oxygen atom can be bonded to another entity. An O-acyl group can be described by the chemical formula: —O—C(═O)—CnH2n+1. Furthermore, depending on the carbon chain, length specific O-acyl groups may be termed an acetyl group (n=1), a propanoyl group (n=2), propoxycarbonyl group (n=3), a butoxycarbonyl group (n=4) etc. The term “azido”, as used herein refers to a chemical group having the formula: —N═N+═N−. The term “5-HT2Areceptor”, as used herein, refers to a subclass of a family of receptors for the neurotransmitter and peripheral signal mediator serotonin. 5-HT2Areceptors can mediate a plurality of central and peripheral physiologic functions of serotonin. Central nervous system effects can include mediation of hallucinogenic effects of hallucinogenic compounds. The term “modulating 5-HT2Areceptors”, as used herein, refers to the ability of a compound disclosed herein to alter the function of 5-HT2Areceptors. A 5-HT2Areceptor modulator may activate the activity of a 5-HT2Areceptor, may activate or inhibit the activity of a 5-HT2Areceptor depending on the concentration of the compound exposed to the 5-HT2Areceptor, or may inhibit the activity of a 5-HT2Areceptor. Such activation or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or maybe manifest only in particular cell types. The term “modulating 5-HT2Areceptors,” also refers to altering the function of a 5-HT2Areceptor by increasing or decreasing the probability that a complex forms between a 5-HT2Areceptor and a natural binding partner to form a multimer. A 5-HT2Areceptor modulator may increase the probability that such a complex forms between the 5-HT2Areceptor and the natural binding partner, may increase or decrease the probability that a complex forms between the 5-HT2Areceptor and the natural binding partner depending on the concentration of the compound exposed to the 5-HT2Areceptor, and or may decrease the probability that a complex forms between the 5-HT2Areceptor and the natural binding partner. Furthermore, the term includes allosteric modulation of the receptor 5-HT2A, i.e., modulation of the 5-HT2Areceptor through interaction with the 5-HT2Areceptor that is topographically different than the orthosteric site recognized by the cell's endogenous agonist, such modulation further including positive allosteric modulation (PAM), negative allosteric modulation (NAM) and silent allosteric modulation (SAM). The term “5-HT2Areceptor-mediated disorder”, as used herein, refers to a disorder that is characterized by abnormal 5-HT2Areceptor activity. A 5-HT2Areceptor-mediated disorder may be completely or partially mediated by modulating 5-HT2Areceptors. In particular, a 5-HT2Areceptor-mediated disorder is one in which modulation of 5-HT2Areceptors results in some effect on the underlying disorder e.g., administration of a 5-HT2Areceptor modulator results in some improvement in at least some of the subjects being treated. The term “5-HT1Areceptor”, as used herein, refers to a subclass of a family of receptors for the neurotransmitter and peripheral signal mediator serotonin. 5-HT1Areceptors can mediate a plurality of central and peripheral physiologic functions of serotonin. Ligand activity at 5-HT1Ais generally not associated with hallucination, although many hallucinogenic compounds are known to modulate 5-HT1Areceptors to impart complex physiological responses (Inserra et al., 2020, Pharmacol Rev 73: 202). The term “modulating 5-HT1Areceptors”, as used herein, refers to the ability of a compound disclosed herein to alter the function of 5-HT1Areceptors. A 5-HT2Areceptor modulator may activate the activity of a 5-HT1Areceptor, may activate or inhibit the activity of a 5-HT1Areceptor depending on the concentration of the compound exposed to the 5-HT1Areceptor, or may inhibit the activity of a 5-HT1Areceptor. Such activation or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or maybe manifest only in particular cell types. The term “modulating 5-HT1Areceptors,” also refers to altering the function of a 5-HT1Areceptor by increasing or decreasing the probability that a complex forms between a 5-HT1Areceptor and a natural binding partner to form a multimer. A 5-HT1Areceptor modulator may increase the probability that such a complex forms between the 5-HT1Areceptor and the natural binding partner, may increase or decrease the probability that a complex forms between the 5-HT2Areceptor and the natural binding partner depending on the concentration of the compound exposed to the 5-HT1Areceptor, and or may decrease the probability that a complex forms between the 5-HT1Areceptor and the natural binding partner. Furthermore, the term includes allosteric modulation of the receptor 5-HT1A, i.e., modulation of the 5-HT1Areceptor through interaction with the 5-HT1Areceptor that is topographically different than the orthosteric site recognized by the cell's endogenous agonist, such modulation further including positive allosteric modulation (PAM), negative allosteric modulation (NAM) and silent allosteric modulation (SAM). The term “5-HT1Areceptor-mediated disorder”, as used herein, refers to a disorder that is characterized by abnormal 5-HT1Areceptor activity. A 5-HT1Areceptor-mediated disorder may be completely or partially mediated by modulating 5-HT2Areceptors. In particular, a 5-HT1Areceptor-mediated disorder is one in which modulation of 5-HT1Areceptors results in some effect on the underlying disorder e.g., administration of a 5-HT1Areceptor modulator results in some improvement in at least some of the subjects being treated. The term “pharmaceutical formulation”, as used herein, refers to a preparation in a form which allows an active ingredient, including a psychoactive ingredient, contained therein to provide effective treatment, and which does not contain any other ingredients which cause excessive toxicity, an allergic response, irritation, or other adverse response commensurate with a reasonable risk/benefit ratio. The pharmaceutical formulation may contain other pharmaceutical ingredients such as excipients, carriers, diluents, or auxiliary agents. The term “recreational drug formulation”, as used herein, refers to a preparation in a form which allows a psychoactive ingredient contained therein to be effective for administration as a recreational drug, and which does not contain any other ingredients which cause excessive toxicity, an allergic response, irritation, or other adverse response commensurate with a reasonable risk/benefit ratio. The recreational drug formulation may contain other ingredients such as excipients, carriers, diluents, or auxiliary agents. The term “effective for administration as a recreational drug”, as used herein, refers to a preparation in a form which allows a subject to voluntarily induce a psychoactive effect for non-medical purposes upon administration, generally in the form of self-administration. The effect may include an altered state of consciousness, satisfaction, pleasure, euphoria, perceptual distortion, or hallucination. The term “effective amount”, as used herein, refers to an amount of an active agent, pharmaceutical formulation, or recreational drug formulation, sufficient to induce a desired biological or therapeutic effect, including a prophylactic effect, and further including a psychoactive effect. Such effect can include an effect with respect to the signs, symptoms or causes of a disorder, or disease or any other desired alteration of a biological system. The effective amount can vary depending, for example, on the health condition, injury stage, disorder stage, or disease stage, weight, or sex of a subject being treated, timing of the administration, manner of the administration, age of the subject, and the like, all of which can be determined by those of skill in the art. The terms “treating” and “treatment”, and the like, as used herein, are intended to mean obtaining a desirable physiological, pharmacological, or biological effect, and includes prophylactic and therapeutic treatment. The effect may result in the inhibition, attenuation, amelioration, or reversal of a sign, symptom or cause of a disorder, or disease, attributable to the disorder, or disease, which includes mental and psychiatric diseases and disorders. Clinical evidence of the prevention or treatment may vary with the disorder, or disease, the subject, and the selected treatment. The term “pharmaceutically acceptable”, as used herein, refers to materials, including excipients, carriers, diluents, or auxiliary agents, that are compatible with other materials in a pharmaceutical or recreational drug formulation and within the scope of reasonable medical judgement suitable for use in contact with a subject without excessive toxicity, allergic response, irritation, or other adverse response commensurate with a reasonable risk/benefit ratio. The term “psilocybin biosynthetic enzyme complement”, as used herein, refers to one or more polypeptides which alone or together are capable of facilitating the chemical conversion of a psilocybin precursor compound and form another psilocybin precursor compound, or an aminated psilocybin derivative compound. A psilocybin biosynthetic enzyme complement can include, for example, a tryptophan synthase subunit B polypeptide, a tryptophan decarboxylase and/or a N-acetyl transferase. The term “tryptophan synthase subunit B polypeptide” as used herein, refers to any and all enzymes comprising a sequence of amino acid residues which is (i) substantially identical to the amino acid sequences constituting any tryptophan synthase subunit B polypeptide set forth herein, including, for example, SEQ. ID NO: 9, or (ii) encoded by a nucleic acid sequence capable of hybridizing under at least moderately stringent conditions to any nucleic acid sequence encoding any tryptophan synthase subunit B polypeptide set forth herein, but for the use of synonymous codons. The term “tryptophan decarboxylase” as used herein, refers to any and all enzymes comprising a sequence of amino acid residues which is (i) substantially identical to the amino acid sequences constituting any tryptophan decarboxylase polypeptide set forth herein, including, for example, SEQ. ID NO: 12, or (ii) encoded by a nucleic acid sequence capable of hybridizing under at least moderately stringent conditions to any nucleic acid sequence encoding any tryptophan decarboxylase set forth herein, but for the use of synonymous codons. The term “N-acetyl transferase” as used herein, refers to any and all enzymes comprising a sequence of amino acid residues which is (i) substantially identical to the amino acid sequences constituting any N-acetyl transferase polypeptide set forth herein, including, for example, SEQ. ID NO: 5, or (ii) encoded by a nucleic acid sequence capable of hybridizing under at least moderately stringent conditions to any nucleic acid sequence encoding any N-acetyl transferase set forth herein, but for the use of synonymous codons. The term “N-methyl transferase” as used herein, refers to any and all enzymes comprising a sequence of amino acid residues which is (i) substantially identical to the amino acid sequences constituting any N-methyl transferase polypeptide set forth herein, including, for example, SEQ. ID NO: 14, or (ii) encoded by a nucleic acid sequence capable of hybridizing under at least moderately stringent conditions to any nucleic acid sequence encoding any N-methyl transferase set forth herein, but for the use of synonymous codons. The terms “nucleic acid sequence encoding tryptophan synthase subunit B polypeptide”, as may be used interchangeably herein, refer to any and all nucleic acid sequences encoding a tryptophan synthase subunit B polypeptide, including, for example, SEQ. ID NO: 8. Nucleic acid sequences encoding a tryptophan synthase subunit B polypeptide further include any and all nucleic acid sequences which (i) encode polypeptides that are substantially identical to the tryptophan synthase subunit B polypeptide sequences set forth herein; or (ii) hybridize to any tryptophan synthase subunit B polypeptide nucleic acid sequences set forth herein under at least moderately stringent hybridization conditions or which would hybridize thereto under at least moderately stringent conditions but for the use of synonymous codons. The terms “nucleic acid sequence encoding tryptophan decarboxylase”, and “nucleic acid sequence encoding a tryptophan decarboxylase polypeptide”, as may be used interchangeably herein, refer to any and all nucleic acid sequences encoding a tryptophan decarboxylase, including, for example, SEQ. ID NO: 11. Nucleic acid sequences encoding a tryptophan decarboxylase polypeptide further include any and all nucleic acid sequences which (i) encode polypeptides that are substantially identical to the tryptophan decarboxylase polypeptide sequences set forth herein; or (ii) hybridize to any tryptophan decarboxylase nucleic acid sequences set forth herein under at least moderately stringent hybridization conditions or which would hybridize thereto under at least moderately stringent conditions but for the use of synonymous codons. The terms “nucleic acid sequence encoding N-acetyl transferase”, and “nucleic acid sequence encoding an N-acetyl transferase polypeptide”, as may be used interchangeably herein, refer to any and all nucleic acid sequences encoding an N-acetyl transferase, including, for example, SEQ. ID NO: 4. Nucleic acid sequences encoding an N-acetyl transferase polypeptide further include any and all nucleic acid sequences which (i) encode polypeptides that are substantially identical to the N-acetyl transferase polypeptide sequences set forth herein; or (ii) hybridize to any N-acetyl transferase nucleic acid sequences set forth herein under at least moderately stringent hybridization conditions or which would hybridize thereto under at least moderately stringent conditions but for the use of synonymous codons. The terms “nucleic acid sequence encoding an N-methyl transferase”, and “nucleic acid sequence encoding an N-methyl transferase polypeptide”, as may be used interchangeably herein, refer to any and all nucleic acid sequences encoding an N-methyl transferase, including, for example, SEQ. ID NO: 13. Nucleic acid sequences encoding an N-methyl transferase polypeptide further include any and all nucleic acid sequences which (i) encode polypeptides that are substantially identical to the N-methyl transferase polypeptide sequences set forth herein; or (ii) hybridize to any N-methyl transferase nucleic acid sequences set forth herein under at least moderately stringent hybridization conditions or which would hybridize thereto under at least moderately stringent conditions but for the use of synonymous codons. The terms “nucleic acid”, or “nucleic acid sequence”, as used herein, refer to a sequence of nucleoside or nucleotide monomers, consisting of naturally occurring bases, sugars and intersugar (backbone) linkages. The term also includes modified or substituted sequences comprising non-naturally occurring monomers or portions thereof. The nucleic acids of the present disclosure may be deoxyribonucleic nucleic acids (DNA) or ribonucleic acids (RNA) and may include naturally occurring bases including adenine, guanine, cytosine, thymidine, and uracil. The nucleic acids may also contain modified bases. Examples of such modified bases include aza and deaza adenine, guanine, cytosine, thymidine and uracil, and xanthine and hypoxanthine. A sequence of nucleotide or nucleoside monomers may be referred to as a polynucleotide sequence, nucleic acid sequence, a nucleotide sequence, or a nucleoside sequence. The term “polypeptide”, as used herein in conjunction with a reference SEQ. ID NO, refers to any and all polypeptides comprising a sequence of amino acid residues which is (i) substantially identical to the amino acid sequence constituting the polypeptide having such reference SEQ. ID NO, or (ii) encoded by a nucleic acid sequence capable of hybridizing under at least moderately stringent conditions to any nucleic acid sequence encoding the polypeptide having such reference SEQ. ID NO, but for the use of synonymous codons. A sequence of amino acid residues may be referred to as an amino acid sequence, or polypeptide sequence. The term “nucleic acid sequence encoding a polypeptide”, as used herein in conjunction with a reference SEQ. ID NO, refers to any and all nucleic acid sequences encoding a polypeptide having such reference SEQ. ID NO. Nucleic acid sequences encoding a polypeptide, in conjunction with a reference SEQ. ID NO, further include any and all nucleic acid sequences which (i) encode polypeptides that are substantially identical to the polypeptide having such reference SEQ. ID NO; or (ii) hybridize to any nucleic acid sequences encoding polypeptides having such reference SEQ. ID NO under at least moderately stringent hybridization conditions or which would hybridize thereto under at least moderately stringent conditions but for the use of synonymous codons. By the term “substantially identical” it is meant that two amino acid sequences preferably are at least 70% identical, and more preferably are at least 85% identical and most preferably at least 95% identical, for example 96%, 97%, 98% or 99% identical. In order to determine the percentage of identity between two amino acid sequences the amino acid sequences of such two sequences are aligned, using for example the alignment method of Needleman and Wunsch (J. Mol. Biol., 1970, 48: 443), as revised by Smith and Waterman (Adv. Appl. Math., 1981, 2: 482) so that the highest order match is obtained between the two sequences and the number of identical amino acids is determined between the two sequences. Methods to calculate the percentage identity between two amino acid sequences are generally art recognized and include, for example, those described by Carillo and Lipton (SIAM J. Applied Math., 1988, 48:1073) and those described in Computational Molecular Biology, Lesk, e.d. Oxford University Press, New York, 1988, Biocomputing: Informatics and Genomics Projects. Generally, computer programs will be employed for such calculations. Computer programs that may be used in this regard include, but are not limited to, GCG (Devereux et al., Nucleic Acids Res., 1984, 12: 387) BLASTP, BLASTN and FASTA (Altschul et al., J. Mol. Biol., 1990:215:403). A particularly preferred method for determining the percentage identity between two polypeptides involves the Clustal W algorithm (Thompson, J D, Higgines, D G and Gibson T J, 1994, Nucleic Acid Res 22(22): 4673-4680 together with the BLOSUM 62 scoring matrix (Henikoff S & Henikoff, J G, 1992, Proc. Natl. Acad. Sci. USA 89: 10915-10919 using a gap opening penalty of 10 and a gap extension penalty of 0.1, so that the highest order match obtained between two sequences wherein at least 50% of the total length of one of the two sequences is involved in the alignment. By “at least moderately stringent hybridization conditions” it is meant that conditions are selected which promote selective hybridization between two complementary nucleic acid molecules in solution. Hybridization may occur to all or a portion of a nucleic acid sequence molecule. The hybridizing portion is typically at least 15 (e.g., 20, 25, 30, 40 or 50) nucleotides in length. Those skilled in the art will recognize that the stability of a nucleic acid duplex, or hybrids, is determined by the Tm, which in sodium containing buffers is a function of the sodium ion concentration and temperature (Tm=81.5° C.-16.6 (Log 10 [Na+])+0.41(% (G+C)−600/I), or similar equation). Accordingly, the parameters in the wash conditions that determine hybrid stability are sodium ion concentration and temperature. In order to identify molecules that are similar, but not identical, to a known nucleic acid molecule a 1% mismatch may be assumed to result in about a 1° C. decrease in Tm, for example if nucleic acid molecules are sought that have a >95% identity, the final wash temperature will be reduced by about 5° C. Based on these considerations those skilled in the art will be able to readily select appropriate hybridization conditions. In preferred embodiments, stringent hybridization conditions are selected. By way of example the following conditions may be employed to achieve stringent hybridization: hybridization at 5× sodium chloride/sodium citrate (SSC)/5×Denhardt's solution/1.0% SDS at Tm (based on the above equation) −5° C., followed by a wash of 0.2×SSC/0.1% SDS at 60° C. Moderately stringent hybridization conditions include a washing step in 3×SSC at 42° C. It is understood however that equivalent stringencies may be achieved using alternative buffers, salts, and temperatures. Additional guidance regarding hybridization conditions may be found in: Current Protocols in Molecular Biology, John Wiley & Sons, N.Y., 1989, 6.3.1.-6.3.6 and in: Sambrook et al., Molecular Cloning, a Laboratory Manual, Cold Spring Harbor Laboratory Press, 1989, Vol. 3. The term “functional variant”, as used herein in reference to polynucleotides or polypeptides, refers to polynucleotides or polypeptides capable of performing the same function as a noted reference polynucleotide or polypeptide. Thus, for example, a functional variant of the polypeptide set forth in SEQ. ID NO: 2, refers to a polypeptide capable of performing the same function as the polypeptide set forth in SEQ. ID NO: 2. Functional variants include modified a polypeptide wherein, relative to a noted reference polypeptide, the modification includes a substitution, deletion, or addition of one or more amino acids. In some embodiments, substitutions are those that result in a replacement of one amino acid with an amino acid having similar characteristics. Such substitutions include, without limitation (i) glutamic acid and aspartic acid; (i) alanine, serine, and threonine; (iii) isoleucine, leucine, and valine, (iv) asparagine and glutamine, and (v) tryptophan, tyrosine, and phenylalanine. Functional variants further include polypeptides having retained or exhibiting an enhanced psilocybin biosynthetic bioactivity. The term “chimeric”, as used herein in the context of nucleic acids, refers to at least two linked nucleic acids which are not naturally linked. Chimeric nucleic acids include linked nucleic acids of different natural origins. For example, a nucleic acid constituting a microbial promoter linked to a nucleic acid encoding a plant polypeptide is considered chimeric. Chimeric nucleic acids also may comprise nucleic acids of the same natural origin, provided they are not naturally linked. For example a nucleic acid constituting a promoter obtained from a particular cell-type may be linked to a nucleic acid encoding a polypeptide obtained from that same cell-type, but not normally linked to the nucleic acid constituting the promoter. Chimeric nucleic acids also include nucleic acids comprising any naturally occurring nucleic acids linked to any non-naturally occurring nucleic acids. The terms “substantially pure” and “isolated”, as may be used interchangeably herein describe a compound, e.g., a secondary metabolite, psilocybin or a psilocybin derivative, polynucleotide, or a polypeptide, which has been separated from components that naturally accompany it. Typically, a compound is substantially pure when at least 60%, more preferably at least 75%, more preferably at least 90%, 95%, 96%, 97%, or 98%, and most preferably at least 99% of the total material (by volume, by wet or dry weight, or by mole percent or mole fraction) in a sample is the compound of interest. Purity can be measured by any appropriate method, e.g., in the case of polypeptides, by chromatography, gel electrophoresis or HPLC analysis. The term “recovered” as used herein in association with a chemical compound, refers to a more or less pure form of the chemical compound. General Implementation As hereinbefore mentioned, the present disclosure relates to psilocybin derivatives. In particular, the present disclosure provides novel aminated psilocybin derivatives. In general, the herein provided compositions exhibit functional properties which deviate from the functional properties of psilocybin. Thus, for example, the aminated psilocybin derivatives, can exhibit pharmacological properties which deviate from psilocybin. Furthermore, the aminated derivatives may psilocybin derivatives may exhibit physico-chemical properties which differ from psilocybin. Thus, for example, aminated psilocybin derivatives may exhibit superior solubility in a solvent, for example, an aqueous solvent. The aminated psilocybin derivatives in this respect are useful in the formulation of pharmaceutical and recreational drug formulations. Furthermore, the aminated psilocybin compounds of the present disclosure may be used as a feedstock material for deriving further psilocybin derivatives. In one embodiment, the aminated psilocybin derivatives of the present disclosure can conveniently be synthetically produced. The practice of this method avoids the extraction of psilocybin from mushrooms and the performance of subsequent chemical reactions to achieve aminated derivatives. Furthermore, the growth of mushrooms can be avoided thus limiting the dependence on climate and weather, and potential legal and social challenges associated with the cultivation of mushrooms containing psychoactive compounds. The method can efficiently yield substantial quantities of aminated psilocybin derivatives. In what follows selected embodiments are described with reference to the drawings. Initially example aminated psilocybin derivatives will be described. Thereafter example methods of using and making the aminated psilocybin derivatives will be described. Accordingly, in one aspect, the present disclosure provides derivatives of a compound known as psilocybin of which the chemical structure is shown inFIG.1. The derivatives herein provided are, in particular, derivatives of psilocybin including an amino group or N-substituted amino group. Thus, in one aspect, the present disclosure provides, in accordance with the teachings herein, in at least one embodiment, a chemical compound or salt thereof having formula (I): wherein at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group, O-alkyl group, a hydroxy group, or a phosphate group, and wherein R3Aand R3Bare each independently a hydrogen, an alkyl group, an aryl group or an acyl group. Thus, referring to the chemical compound having formula (I), initially it is noted that, in an aspect thereof, at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group. Thus, referring to the chemical compound having the formula (I), initially it is noted that, in an aspect hereof, at least one of R2, R5, R6, or R7is an amino group or an N-substituted amino group. In a further aspect, at least one of R2, R5, R6, or R7can be a N-substituted amino group, wherein one, or at least one, hydrogen atom is substituted by a group selected from an alkyl group, an aryl group, an acyl group, or a sulfonyl group. Thus, for example, in one embodiment, the N-substituted amino group can be a chemical group having the formula (XX): wherein R′, and R″ are each independently selected from a hydrogen atom, an alkyl group, and an aryl group, provided however, that at least one of R′ and R″ is not a hydrogen atom. In a further example embodiment, the N-substituted amino group can be a chemical group having the formula chemical group (XXI): wherein R′, R″ and R′″ are each independently selected from a hydrogen atom, an alkyl group and an aryl group, provided however that at least one of R′, R″, and R′″ is not a hydrogen atom. In a further example embodiment, the N-substituted amino group can be a chemical group having the formula (XXII) (an acyl group): wherein R′, and R′″ are each independently selected from a hydrogen atom, an alkyl group, and an aryl group. In a further example embodiment, the N-substituted amino group can be a chemical group having the formula (XXIII) (a sulfonyl group): wherein R′ and R″ are each independently selected from a hydrogen atom, an alkyl group and an aryl group. In yet a further example embodiment, the N-substituted amino group can be a chemical group having the formula (XXIV) (a sulfo group): wherein R″ is selected from a hydrogen atom, an alkyl group, and an aryl group. The nitrogen atom of chemical groups (XXII), (XXIII) and (XXIV) can also be positively charged and be further substituted with H, or R′″. Continuing to refer to the chemical compound having formula (I), in a further aspect, R2, R5, R6, or R7can be a N-substituted amino-group wherein two, or at least two, hydrogen atoms are substituted by a group independently selected from an alkyl group, an aryl group, an acyl group or a sulfonyl group. In at least one embodiment, at least one of R2, R5, R6, or R7can be a N-substituted amino-group (i.e., an ammonium group), wherein three hydrogen atoms are substituted by a group independently selected from an alkyl group, or an aryl group, wherein the nitrogen atom of the N-substituted group carries a positive charge. Continuing to refer to the chemical compound having formula (I), in a further aspect hereof, R3Aand R3Bcan each independently be a hydrogen atom, an alkyl group, an acyl group or an aryl group. Thus, R3Aand R3Bcan each be a hydrogen atom, or R3Aand R3Bcan each be an alkyl group, such as a methyl group, ethyl group, propyl group, or longer chain alkyl group, or R3Aand R3Bcan be each be an acyl group, or R3Aand R3Bcan each be an aryl group. Furthermore, one of R3Aand R3Bcan be a hydrogen atom, and one of R3Aand R3Bcan be an alkyl group. One of R3Aand R3Bcan be a hydrogen atom, and one of R3Aand R3Bcan be an acyl group. One of R3Aand R3Bcan be a hydrogen atom, and one of R3Aand R3Bcan be an aryl group. One of R3Aand R3Bcan be an alkyl group, and one of R3Aand R3Bcan be an aryl group. One of R3Aand R3Bcan be an alkyl group, and one of R3Aand R3Bcan be an acyl group. One of R3Aand R3Bcan be an acyl group, and one of R3Aand R3Bcan be an aryl group. Continuing to refer to the chemical compound having formula (I), in a further aspect hereof, R4, when it is not an amino group or N-substituted amino group can be is a hydrogen atom, an alkyl group, O-alkyl group, a hydroxy group, or a phosphate group. Continuing to refer to the chemical compound having formula (I), in a further aspect hereof, the non-aminated groups R2, R5, R6, or R7can be a hydrogen atom or an alkyl or O-alkyl group. Referring now toFIGS.3A-3D, examples of aminated psilocybin derivatives in accordance herewith, wherein one of R2, R5, R6, or R7are aminated, and the non-aminated groups R2, R5, R6, or R7are hydrogen atoms are: the 2-amino-psilocybin derivative compound depicted inFIG.3A, the 5-amino-psilocybin derivative depicted inFIG.3B, the 6-amino-psilocybin derivative depicted inFIG.3C, the 7-amino-psilocybin derivative depicted inFIG.3D, Referring now toFIGS.3E-3H, examples of aminated psilocybin derivatives in accordance herewith, wherein one of R2, R5, R6, or R7are aminated, and the non-aminated groups R2, R5, R6, or R7are hydrogen atoms are: the 2-N,N-substituted amino-psilocybin derivative compound depicted inFIG.3E, the 5-N,N-substituted amino-psilocybin derivative depicted inFIG.3F, the 6-N,N-substituted amino-psilocybin derivative depicted inFIG.3G, and the 7-N,N-substituted amino-psilocybin derivative depicted inFIG.3H. It is noted that inFIGS.3E-3H, R′2a, R″2b, R′5a, R″5b, R′6a, R″6b, R′7a, and R″7bcan each be independently selected from an alkyl group, an aryl group, an acyl group, a sulfonyl group, a sulfo group, or a hydrogen atom, provided however, that N-substituted amino psilocybin derivatives do not include psilocybin derivatives wherein in both R′2a, and R″2b, or both R′5a, and R″5b, or both R′6aand R″6b, or both R′7a, and R″7bare hydrogen atoms. In a further aspect hereof, the non-aminated groups R2, R5, R6, or R7can be a hydrogen atom or an alkyl or O-alkyl group. Referring now toFIGS.3I-3L, examples of aminated psilocybin derivatives in accordance herewith, wherein one of R2, R5, R6, or R7are aminated, and the non-aminated groups R2, R5, R6, or R7are alkyl or O-alkyl groups are: the 2-amino-6-ethyl-psilocybin derivative compound depicted inFIG.3I, the 5-amino-7-methyl psilocybin derivative depicted inFIG.3J, the 5-ethoxy-6-amino-psilocybin derivative depicted inFIG.3K, the 6-O-methyl-7-amino-psilocybin derivative depicted inFIG.3L, Referring now toFIGS.3M-3P, examples of aminated psilocybin derivatives in accordance herewith, wherein one of R2, R5, R6, or R7are aminated, and the non-aminated groups R2, R5, R6, or R7are hydrogen atoms are: the 2-N,N-substituted amino-7methyl-psilocybin derivative compound depicted inFIG.3M, the 5-N,N-substituted amino-6-ethyl-psilocybin derivative depicted inFIG.3N, the 5-O-methyl-6-N,N-substituted amino-psilocybin derivative depicted inFIG.3O, and the 5-O-ethyl-7-N,N-substituted amino-psilocybin derivative depicted inFIG.3P. It is noted that inFIGS.3M-3P, R′2a, R″2b, R′5a, R″5b, R′6a, R″6b, R′7a, and R″7bcan each be independently selected from an alkyl group, an aryl group, an acyl group, a sulfonyl group, a sulfo group, or a hydrogen atom, provided however, that N-substituted amino psilocybin derivatives do not include psilocybin derivatives wherein in both R′2a, and R″2b, or both R′5a, and R″5b, or both R′6aand R″6b, or both R′7a, and R″7bare hydrogen atoms. Referring now toFIGS.4A-4F, examples of aminated psilocybin derivatives in accordance herewith, wherein two of R2, R5, R6, or R7are aminated, and the non-aminated groups R2, R5, R6, or R7are hydrogen atoms are: the 2,5-di-amino-psilocybin derivative compound depicted inFIG.4A, the 2,6-di-amino-psilocybin derivative depicted inFIG.4B, the 2,7-di-amino-psilocybin derivative depicted inFIG.4C, the 5,6-di-amino-psilocybin derivative depicted inFIG.4D, the 5,7-di-amino-psilocybin derivative depicted inFIG.4E, and the 6,7-di-amino-psilocybin derivative depicted inFIG.4F. Referring now toFIGS.4G-4J, examples of aminated psilocybin derivatives in accordance herewith, wherein three of R2, R5, R6, or R7are aminated, and the non-aminated groups R2, R5, R6, or R7are hydrogen atoms are: the 2,5,6-tri-amino-psilocybin derivative compound depicted inFIG.4G, the 2,5,7-tri-amino-psilocybin derivative depicted inFIG.4H, the 2,6,7-tri-amino-psilocybin derivative depicted inFIG.4I, and the 5,6,7-tri-amino-psilocybin derivative depicted inFIG.4J. Referring now toFIG.4Kan example of a aminated psilocybin derivatives in accordance herewith, wherein all four of R2, R5, R6, or R7are aminated is the 2,5,6,7-tetra-amino-psilocybin derivative depicted inFIG.4K. Referring now toFIGS.4L-4Q, examples of aminated psilocybin derivatives in accordance herewith, wherein two of R2, R5, R6, or R7are aminated, and the non-aminated groups R2, R5, R6, or R7are hydrogen atoms are: the 2-N,5-N,N-di-substituted-amino-psilocybin derivative compound depicted inFIG.4L, the 2-N,6-N,N-di-substituted-amino-psilocybin derivative depicted inFIG.4M, the 2-N,7-N,N-di-substituted-amino-psilocybin derivative depicted inFIG.4N, the 5-N,6-N,N-di-substituted-amino-psilocybin derivative depicted inFIG.4O, the 5-N,7-N,N-di-substituted-amino-psilocybin derivative depicted inFIG.4P, and the 6-N,N-7-N,N-di-substituted-amino-psilocybin derivative depicted inFIG.4Q. As hereinbefore noted, the substituents may be a group selected from an alkyl group, an aryl group, an acyl group, or a sulfonyl group. Thus, by way of example only, in the 5-N,6-N,N-di-substituted-amino-psilocybin derivative depicted inFIG.4Oat least one of R′5a, of R″5b, and at least one of R′6a, and R″6b, is an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. Continuing to refer toFIGS.4L-4Q, it is noted that, in other embodiments, instead of being di-substituted, only one of the aminated groups may be an N-substituted amino group while the other aminated group is an amino group. Thus, for example, referring toFIG.4P, in such embodiments, only R″7aor R′7bmay be N-substituted, while R′5aand R″5bmay each be a hydrogen atom, thus forming an amino group, or conversely, only R″5aor R′5bmay be N-substituted, while R′7aand R″7bmay be hydrogen atoms thus forming an amino group. It is to be understood that any and all embodiments including the aminated psilocybin derivatives shown inFIGS.4L-4Q, provided that at least one of the R′2a, R″2b, R′5a, R″5b, R′6a, R″6b, R′7a, and R″7bis an N-substituted amino group are also included herein. As hereinbefore noted, the substituents may be a group selected from an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. Referring now toFIGS.4R-4U, examples of aminated psilocybin derivatives in accordance herewith, wherein three of R2, R5, R6, or R7are aminated, and the non-aminated groups R2, R5, R6, or R7are hydrogen atoms are: the 2-N,5-N,6-N,N-tri-substituted-amino-psilocybin derivative compound depicted inFIG.4R, the 2-N,5-N,7-N,N-tri-substituted-amino-psilocybin derivative depicted inFIG.4S, the 2-N,6-N,7-N,N-tri-substituted amino-psilocybin derivative depicted inFIG.4T, and the 5-N,6-N,7-N,N-tri-substituted-amino-psilocybin derivative depicted inFIG.4U. As hereinbefore noted, the substituents may be a group selected from an alkyl group, an aryl group, an acyl group, or a sulfonyl group. Thus, by way of example only, in the 5-N,6-N,7-N,N-tri-substituted-amino-psilocybin derivative depicted inFIG.4Uat least one of R′5a, of R″5b, and at least one of R′6a, and R″6b, and at least one of R′7a, and R″7bis an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. Continuing to refer toFIGS.4R-4U, it is noted that, in other embodiments, instead of being tri-substituted, only one or two of the aminated groups may be an N-substituted amino group while the other aminated groups is/are an amino group. Thus, for example, referring toFIG.4S, in such embodiments, only R″7aor R′7bmay be N-substituted, while R′5aand R″5band R′2aand R″2bmay all be a hydrogen atoms, thus forming two an amino groups, or R″5aor R′5bmay be N-substituted, and R′7aand R″7bmay N-substituted, but R′2aand R″2bmay be hydrogen atoms thus forming an amino group. It is to be understood that any and all embodiments including aminated psilocybin derivatives shown inFIGS.4R-4U, provided that at least one of the R′2a, R″2b, R′5a, R″5b, R′6a, R″6b, R′7a, and R″7bis an N-substituted amino group are also included herein. As hereinbefore noted, the substituents may be a group selected from an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. Referring now toFIG.4V, an example of an aminated psilocybin derivatives in accordance herewith, wherein all four of R2, R5, R6, or R7are aminated is the 2-N,5-N,6-N,7-N,N-tetra-substituted-amino-psilocybin derivative depicted inFIG.4K. As hereinbefore noted, the substituents may be a group selected from an alkyl group, an aryl group, an acyl group, or a sulfonyl group. Thus, by way of example only, in the 2-N,N-5-N,6-N,7-N,N-tri-substituted-amino-psilocybin derivative depicted inFIG.4Vat least one of R′2a, of R″2band R′5a, of R″5b, at least one of R′6a, and R″6b, at least one of R′7a, and R″7bis an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. Continuing to refer toFIG.4V, it is noted that, in other embodiments, instead of being tetra-substituted, only one, two or three of the aminated groups may be an N-substituted amino group while the other aminated groups is/are an amino group. Thus, for example, referring toFIG.4V, in such embodiments, only R″7aor R′7bmay be N-substituted, while R′5aand R″5b, R′6aand R″6band R′2aand R″2bmay all be a hydrogen atoms, thus forming three amino groups, or, for example R″5aor R′5bmay be N-substituted, and R′7aand R″7bmay N-substituted, but R′2aand R″2band R′6aand R″6bmay be hydrogen atoms thus forming two amino groups. It is to be understood that any and all embodiments including the aminated psilocybin derivatives shown inFIGS.4V, provided that at least one of the R′2a, R″2b, R′5a, R″5b, R′6a, R″6b, R′7a, and R″7bis an N-substituted amino group are also included herein. As hereinbefore noted, the substituents may be a group selected from an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. In a further aspect, R4, can be an O-alkyl group. Referring now toFIGS.5A,5B,6A,6B,7A, and7Bexamples of aminated psilocybin derivatives in accordance herewith, wherein R5, and/or R7are amino groups and R4is an O-alkyl group are: the 4-O-methyl-5-amino-psilocybin derivative depicted inFIG.5A, the 4-O-ethyl-5-amino-psilocybin derivative depicted inFIG.5B, the 4-O-methyl-7-amino-psilocybin derivative depicted inFIG.6A, the 4-O-ethyl-7-amino-psilocybin derivative depicted in FIG. GB, the 4-O-methyl-5,7-di-amino-psilocybin derivative depicted inFIG.7A, the 4-O-ethyl-5,7-di-amino-psilocybin derivative depicted inFIG.7B. It is noted that in these specific examples only 5-amino, 7-amino, and 5,7-di-O-alkyl psilocybin derivatives are shown. Further examples of O-alkyl psilocybin derivatives included herein are any and all O-alkyl psilocybin derivatives which may be selected by referring to the chemical formulas shown inFIGS.4A-4J, wherein R4is an O-alkyl group. It will thus be clearly understood thatFIGS.5A,5B,6A,6B,7A, and7Brepresent examples only of aminated psilocybin derivatives having chemical formula (I) wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom. Other aminated psilocybin derivatives wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom can readily be selected, and thus are included in the O-alkylated aminated psilocybin derivatives compounds of the present disclosure. It is noted that the example aminated psilocybin derivatives shown inFIGS.5A,5B,6A,6B,7A, and7Bare aminated psilocybin derivatives compounds by virtue of their amino groups. ConsideringFIGS.5A,5B,6A,6B,7A, and7B, in conjunction withFIGS.4L,4N, and4P, it is noted, and it will be clear that, in other embodiments, included herein are, further, aminated psilocybin derivativesFIGS.5A,5B,6A,6B,7A, and7B, wherein instead of an amino group the psilocybin derivative possesses at least on N-substituted amino group, i.e., R′7aor R″7bis substituted by a group selected from an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. In a further aspect, R4, can be an O-acyl group. Referring now toFIGS.5C,5D,6C,6D,7C, and7Dexamples of aminated psilocybin derivatives in accordance herewith, wherein R5, and/or R7are amino groups and R4is an O-acyl group are: the 4-acetyl-5-amino-psilocybin derivative depicted inFIG.5C, the 4-propanoyl-5-amino-psilocybin derivative depicted inFIG.5D, the 4-acetyl-7-amino-psilocybin derivative depicted in FIG. GC, the 4-propanoyl-7-amino-psilocybin derivative depicted inFIG.6D, the 4-acetyl-5,7-di-amino-psilocybin derivative depicted inFIG.7C, the 4-propanoyl-5,7-di-amino-psilocybin derivative depicted inFIG.7D. It is noted that in these specific examples only 5-amino, 7-amino, and 5,7-di-O-acyl psilocybin derivatives are shown. Further examples of O-acyl psilocybin derivatives included herein are any and all O-acyl psilocybin derivatives which may be selected by referring to the chemical formulas shown inFIGS.4A-4J, wherein R4is an O-acyl group. It will thus be clearly understood thatFIGS.5C,5D,6C,6D,7C, and7Drepresent examples only of O-acylated psilocybin derivatives having chemical formula (I) wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom. Other aminated psilocybin derivatives wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom can readily be selected, and thus are included in the aminated O-acylated psilocybin derivatives compounds of the present disclosure. It is noted that the example aminated psilocybin derivatives shown inFIGS.5C,5D,6C,6D,7C, and7Dare aminated psilocybin derivatives compounds by virtue of their amino groups. Considering5C,5D,6C,6D,7C, and7D, in conjunction withFIGS.4L,4N, and4P, it is noted, and it will be clear that, in other embodiments, included herein are, further, aminated psilocybin derivatives5C,5D,6C,6D,7C, and7D, wherein instead of an amino group the psilocybin derivative possesses at least on N-substituted amino group, i.e., R′7a or R″7bis substituted by a group selected from an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. In a further aspect, R4can be a hydroxy group. Referring now toFIGS.5E,6E, and7Eexamples of aminated psilocybin derivatives in accordance herewith, wherein R5, and/or R7are amino groups and are R4is a hydroxy group are: the 4-hydroxy-5-amino-psilocybin derivative depicted inFIG.5E, the 4-hydroxy-7-amino-psilocybin derivative depicted inFIG.6E, and the 4-hydroxy-5,7-di-amino-psilocybin derivative depicted inFIG.7E, It is noted that in these specific examples only 5-amino, 7-amino, and 5,7-di-hydroxy-psilocybin derivatives are shown. Further examples of hydroxy-psilocybin derivatives included herein are any and all hydroxy-psilocybin derivatives which may be selected by referring to the chemical formulas shown inFIGS.4A-4J, wherein R4is a hydroxy group. It will thus be clearly understood thatFIGS.5E,6E, and7Erepresent examples only of hydroxy psilocybin derivatives having chemical formula (I) wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom. Other aminated psilocybin derivatives wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom can readily be selected, and thus are included in the aminated hydroxy psilocybin derivatives compounds of the present disclosure. It is noted that the example aminated psilocybin derivatives shown inFIGS.5E,6E, and7Eare aminated psilocybin derivatives compounds by virtue of their amino groups. ConsideringFIGS.5E,6E, and7E, in conjunction withFIGS.4L,4N, and4P, it is noted, and it will be clear that, in other embodiments, included herein are, further, aminated psilocybin derivativesFIGS.5E,6E, and7E, wherein instead of an amino group the psilocybin derivative possesses at least on N-substituted amino group, i.e., R′7aor R″7bis substituted by a group selected from an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. In a further aspect, R4can be a phosphate group. Referring now toFIGS.5F,6F, and7Fexamples of aminated psilocybin derivatives in accordance herewith, wherein R5, and/or R7are amino groups and are R4is a phosphate group are: the 4-phospho-5-amino-psilocybin derivative depicted inFIG.5F, the 4-phospho-7-amino-psilocybin derivative depicted inFIG.6F, and the 4-phosphate-5,7-amino-psilocybin derivative depicted inFIG.7F, It is noted that in these specific examples only 5-amino, 7-amino, and 5,7-di-phospho-psilocybin derivatives are shown. Further examples of phosphate-psilocybin derivatives included herein are any and all phosphate-psilocybin derivatives which may be selected by referring to the chemical formulas shown inFIGS.4A-4J, wherein R4is a phosphate group. It will thus be clearly understood thatFIGS.5F,6F, and7Frepresent examples only of phosphate psilocybin derivatives having chemical formula (I) wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom. Other aminated psilocybin derivatives wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom can readily be selected, and thus are included in the aminated phosphate psilocybin derivatives compounds of the present disclosure. It is noted that the example aminated psilocybin derivatives shown inFIGS.5F,6F, and7Fare aminated psilocybin derivatives compounds by virtue of their amino groups. ConsideringFIGS.5F,6F, and7F, in conjunction withFIGS.4L,4N, and4P, it is noted, and it will be clear that, in other embodiments, included herein are, further, aminated psilocybin derivativesFIGS.5F,6F, and7F, wherein instead of an amino group the psilocybin derivative possesses at least on N-substituted amino group, i.e., R′7aor R″7bis substituted by a group selected from an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. In a further aspect, R4can be a hydrogen atom. Referring now toFIGS.5G,6G, and7Gexamples of aminated psilocybin derivatives in accordance herewith, wherein R5, and/or R7are amino groups and are R4is a hydrogen atom are: the 5-amino-psilocybin derivative depicted inFIG.5G, the 7-amino-psilocybin derivative depicted inFIG.6G, and the 5,7-di-amino-psilocybin derivative depicted inFIG.7G, It is noted that in these specific examples only 5-amino, 7-amino, and 5,7-di-amino-hydro-psilocybin derivatives are shown. Further examples of hydro-psilocybin derivatives included herein are any and all hydro-psilocybin derivatives which may be selected by referring to the chemical formulas shown inFIGS.4A-4J, wherein R4is a hydrogen atom. It will thus be clearly understood thatFIGS.5G,6G, and7Grepresent examples only of hydro psilocybin derivatives having chemical formula (I) wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom. Other aminated psilocybin derivatives wherein non-aminated groups R2, R5, R6, or R7are a hydrogen atom can readily be selected, and thus are included in the aminated hydro psilocybin derivatives compounds of the present disclosure. It is noted that the example aminated psilocybin derivatives shown inFIGS.5G,6G, and7Gare aminated psilocybin derivatives compounds by virtue of their amino groups. ConsideringFIGS.5G,6G, and7G, in conjunction withFIGS.4L,4N, and4P, it is noted, and it will be clear that, in other embodiments, included herein are, further, aminated psilocybin derivativesFIGS.5G,6G, and7G, wherein instead of an amino group the psilocybin derivative possesses at least one N-substituted amino group, i.e., R′7aor R″7bis substituted by a group selected from an alkyl group, an aryl group, an acyl group, a sulfo group, or a sulfonyl group. Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (III): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (IV): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (V): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (VI): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (VII): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (VIII): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (IX): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (X): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (XI): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (XII): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (XIII): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (XIV): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (XV): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (XVI): Furthermore, in one embodiment, an aminated psilocybin derivative according to the present disclosure can be a chemical compound having the formula (XVII): Furthermore, it is noted that the aminated psilocybin derivatives of the present disclosure include salts thereof, including pharmaceutically acceptable salts. Thus, the nitrogen atom of the ethyl-amino group extending in turn from the C3atom may be protonated, and the positive charge may be balanced by, for example, chloride or sulfate ions, to thereby form a chloride salt or a sulfate salt. Furthermore, in compounds wherein R4is a phosphate group, the phosphate group may be de-protonated, and the negative charge may be balanced by, for example, sodium ions or potassium ions, to thereby form a sodium salt or a potassium salt. Furthermore, it is noted that when R4is a phosphate group, the term aminated psilocybin derivative also includes compounds having the formula (XVIII): wherein at least one of R2, R5, R6, or R7is an amino group or N-substituted amino group, and wherein any R2, R5, R6, or R7which are not an amino group or N-substituted amino group are a hydrogen atom, an alkyl group or O-alkyl group, and wherein R3Aand R3Bare each independently a hydrogen atom, an alkyl group, and aryl group or an acyl group. Further included are salts of aminated psilocybins having the formula (XVIII), such as a sodium salt, a potassium salt etc. Thus, to briefly recap, the present disclosure provides aminated psilocybin derivatives. The disclosure provides, in particular, a chemical compound or salt thereof having formula (I): wherein in an aspect, at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group. In an aspect, in formula (I), each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group. In a further aspect, in formula (I), R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group. Yet in a further aspect, R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an aryl group, or an acyl group. In one embodiment of the disclosure, a chemical compound or salt thereof having formula (I) is included: wherein at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, alkyl group or O-alkyl group, a hydroxy group, or a phosphate group, and wherein R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an aryl group, or an acyl group. In one embodiment, at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom or a (C1-C20)-alkyl group or (C1-C20)—O-alkyl group. In another embodiment, each non-aminated R2, R5, R6, or R7is a hydrogen atom, a methyl group, ethyl group, a propyl group, an O-methyl group, an O-ethyl group, or an O-propyl group. In another embodiment, each non-aminated R2, R5, R6, or R7is a hydrogen atom or a (C1-C10)-alkyl group or (C1-C10)—O-alkyl group. In another embodiment, each non-aminated R2, R5, R6, or R7is a hydrogen atom, a methyl group, ethyl group, a propyl group, an O-methyl group, an O-ethyl group, or an O-propyl group. In another embodiment, each non-aminated R2, R5, R6, or R7is a hydrogen atom or a (C1-C6)-alkyl group or (C1-C6)—O-alkyl group. In another embodiment, each non-aminated R2, R5, R6, or R7is a hydrogen atom, a methyl group, ethyl group, a propyl group, an O-methyl group, an O-ethyl group, or an O-propyl group. In another embodiment, when R4is not aminated, R4is a hydrogen atom, a (C1-C20)-alkyl group or (C1-C20)—O-alkyl group, a hydroxy group, or a phosphate group. In another embodiment, when R4is not aminated, R4is a hydrogen atom, a (C1-C10)-alkyl group or (C1-C10)—O-alkyl group, a hydroxy group, or a phosphate group. In another embodiment, when R4is not aminated, R4is a hydrogen atom, a (C1-C6)-alkyl group or (C1-C6)—O-alkyl group, a hydroxy group, or a phosphate group. In another embodiment, when R4is not aminated, R4is a hydrogen atom, a methyl group, an ethyl group, a propyl group, a phosphate group, an O-methyl group, an O-ethyl group, or an O-propyl group. In another embodiment, R3Aand R3Bare each independently a hydrogen atom, a (C1-C20)-alkyl group, a (C6-C14)-aryl group, or a —C(═O)(C1-C20)-alkyl group. In another embodiment, R3Aand R3Bare each independently a hydrogen atom, a (C1-C10)-alkyl group, a (C6-C10)-aryl group, or a —C(═O)(C1-C10)-alkyl group. In another embodiment, R3Aand R3Bare each independently a hydrogen atom, a (C1-C6)-alkyl group, a phenyl group, or a —C(═O)(C1-C6)-alkyl group. In another embodiment, R3Aand R3Bare each independently a hydrogen atom, a methyl group, an ethyl group, a propyl group, a phenyl group, —C(═O)—CH3, —C(═O)—CH2CH3, or —C(═O)—CH2CH2CH3. In one embodiment of the disclosure, a chemical compound or salt thereof having formula (I) is included: wherein R2, R5, R6, and R7are independently or simultaneously a hydrogen atom, an alkyl group or O-alkyl group or an amino group or N-substituted amino group, R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an aryl group, or an acyl group; and R4is hydrogen atom, alkyl group or O-alkyl group, an amino group or N-substituted amino group, a hydroxy group, or a phosphate group; wherein at least one of R2, R4R5, R6, and R7is an amino group or N-substituted amino group. In one embodiment, R2, R5, R6, and R7are independently or simultaneously a hydrogen atom, (C1-C20)-alkyl group or (C1-C20)—O-alkyl group or an amino group or N-substituted amino group. In one embodiment, R2, R5, R6, and R7are independently or simultaneously a hydrogen atom, (C1-C10)-alkyl group or (C1-C10)—O-alkyl group or an amino group or N-substituted amino group. In one embodiment, R2, R5, R6, and R7are independently or simultaneously a hydrogen atom, (C1-C6)-alkyl group or (C1-C6)—O-alkyl group or an amino group or N-substituted amino group. In one embodiment, R2, R5, R6, and R7are independently or simultaneously a hydrogen atom, methyl, ethyl, propyl, O-methyl, O-ethyl, O-propyl, or an amino group or N-substituted amino group. In one embodiment, R4is a hydrogen atom, (C1-C20)-alkyl group or (C1-C20)—O-alkyl group, an amino group or N-substituted amino group or a phosphate group. In one embodiment, R4is a hydrogen atom, (C1-C10)-alkyl group or (C1-C10)—O-alkyl group, an amino group or N-substituted amino group or a phosphate group. In one embodiment, R4is a hydrogen atom, (C1-C6)-alkyl group or (C1-C6)—O-alkyl group, an amino group or N-substituted amino group, a hydroxy group, or a phosphate group. In one embodiment, R4is a hydrogen atom, methyl, ethyl, propyl, O-methyl, O-ethyl, O-propyl, an amino group or N-substituted amino group, a hydroxy group, or a phosphate group. In another embodiment, R3Aand R3Bare each independently a hydrogen atom, a (C1-C20)-alkyl group, a (C6-C14)-aryl group, or a —C(═O)(C1-C20)-alkyl group. In another embodiment, R3Aand R3Bare each independently a hydrogen atom, a (C1-C10)-alkyl group, a (C6-C10)-aryl group, or a —C(═O)(C1-C10)-alkyl group or O-alkyl group. In another embodiment, R3Aand R3Bare each independently a hydrogen atom, a (C1-C6)-alkyl group, a phenyl group, or a —C(═O)(C1-C6)-alkyl group. In another embodiment, R3Aand R3Bare a hydrogen atom, a methyl group, an ethyl group, a propyl group, a phenyl group, —C(═O)—CH3, —C(═O)—CH2CH3, or —C(═O)—CH2CH2CH3. The aminated psilocybin derivatives of the present disclosure may be used to prepare a pharmaceutical or recreational drug formulation. Thus in one embodiment, the present disclosure further provides in another aspect, pharmaceutical and recreational drug formulations comprising aminated psilocybin derivatives. Accordingly, in one aspect, the present disclosure provides in a further embodiment a pharmaceutical or recreational drug formulation comprising a chemical compound having formula (I): wherein at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group, and wherein R3Aand R3Bare each independently a hydrogen atom an alkyl group, an aryl group, or an acyl group, or a slat of the chemical compound, together with a diluent, carrier or excipient. The pharmaceutical or recreational drug formulations may be prepared as liquids, tablets, capsules, microcapsules, nanocapsules, trans-dermal patches, gels, foams, oils, aerosols, nanoparticulates, powders, creams, emulsions, micellar systems, films, sprays, ovules, infusions, teas, decoctions, suppositories, etc. and include a pharmaceutically acceptable salt or solvate of the aminated psilocybin compound together with an excipient. The term “excipient” as used herein means any ingredient other than the chemical compound of the disclosure. As will readily be appreciated by those of skill in art, the selection of excipient may depend on factors such as the particular mode of administration, the effect of the excipient on solubility of the chemical compounds of the present disclosure and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in “Remington's Pharmaceutical Sciences”, 22ndEdition (Pharmaceutical Press and Philadelphia College of Pharmacy at the University of the Sciences, 2012). The dose when using the compounds of the present disclosure can vary within wide limits, and as is customary and is known to those of skill in the art, the dose can be tailored to the individual conditions in each individual case. The dose depends, for example, on the nature and severity of the illness to be treated, on the condition of the patient, on the compound employed or on whether an acute or chronic disease state is treated, or prophylaxis is conducted, on the mode of delivery of the compound, or on whether further active compounds are administered in addition to the compounds of the present disclosure. Representative doses of the present disclosure include, but are not limited to, about 0.001 mg to about 5000 mg, about 0.001 mg to about 2500 mg, about 0.001 mg to about 1000 mg, about 0.001 mg to about 500 mg, about 0.001 mg to about 250 mg, about 0.001 mg to about 100 mg, about 0.001 mg to about 50 mg, and about 0.001 mg to about 25 mg. Representative doses of the present disclosure include, but are not limited to, about 0.0001 to about 1,000 mg, about 10 to about 160 mg, about 10 mg, about 20 mg, about 40 mg, about 80 mg, or about 160 mg. Multiple doses may be administered during the day, especially when relatively large amounts are deemed to be needed, for example 2, 3 or 4, doses. Depending on the subject and as deemed appropriate from the patient's physician or care giver it may be necessary to deviate upward or downward from the doses described herein. The pharmaceutical and drug formulations comprising the aminated psilocybin derivatives of the present disclosure may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth. Formulations suitable for oral administration include both solid and liquid formulations. Solid formulations include tablets, capsules (containing particulates, liquids, microcapsules, or powders), lozenges (including liquid-filled lozenges), chews, multi- and nano-particulates, gels, solid solutions, liposomal preparations, microencapsulated preparations, creams, films, ovules, suppositories, and sprays. Liquid formulations include suspensions, solutions, syrups, and elixirs. Such formulations may be employed as fillers in soft or hard capsules and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet. Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinized starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch, and dibasic calcium phosphate dihydrate. Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80. When present, surface active agents may comprise from 0.2% (w/w) to 5% (w/w) of the tablet. Tablets may further contain lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate. Lubricants generally comprise from 0.25% (w/w) to 10% (w/w), from 0.5% (w/w) to 3% (w/w) of the tablet. In addition to the aminated psilocybin derivative, tablets may contain a disintegrant. Examples of disintegrants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropyl cellulose, starch, pregelatinized starch and sodium alginate. Generally, the disintegrant will comprise from 1% (w/w) to 25% (w/w) or from 5% (w/w) to 20% (w/w) of the dosage form. Other possible auxiliary ingredients include anti-oxidants, colourants, flavouring agents, preservatives, and taste-masking agents. For tablet dosage forms, depending on the desired effective amount of the chemical compound, the chemical compound of the present disclosure may make up from 1% (w/w) to 80% (w/w) of the dosage form, more typically from 5% (w/w) to 60% (w/w) of the dosage form. Exemplary tablets contain up to about 80% (w/w) of the chemical compound, from about 10% (w/w) to about 90% (w/w) binder, from about 0% (w/w) to about 85% (w/w) diluent, from about 2% (w/w) to about 10% (w/w) disintegrant, and from about 0.25% (w/w) to about 10% (w/w) lubricant. The formulation of tablets is discussed in “Pharmaceutical Dosage Forms: Tablets”, Vol. 1-Vol. 3, by CRC Press (2008). The pharmaceutical and recreational drug formulations comprising the aminated psilocybin derivatives of the present disclosure may also be administered directly into the blood stream, into muscle, or into an internal organ. Thus, the pharmaceutical and recreational drug formulations can be administered parenterally (for example, by subcutaneous, intravenous, intraarterial, intrathecal, intraventricular, intracranial, intramuscular, or intraperitoneal injection). Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (in one embodiment, to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile water. Formulations comprising the aminated psilocybin derivatives of the present disclosure for parenteral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. Thus the chemical compounds of the disclosure may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound. Examples of such formulations include drug-coated stents and poly(dl-lactic-coglycolic) acid (PGLA) microspheres. The pharmaceutical or recreational drug formulations of the present disclosure also may be administered topically to the skin or mucosa, i.e., dermally or transdermally. Example pharmaceutical and recreational drug formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, cosmetics, oils, eye drops, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Example carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporate (see: for example, Finnin, B. and Morgan, T. M., 1999 J. Pharm. Sci, 88 (10), 955-958). Other means of topical administration include delivery by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free (e.g., Powderject™, Bioject™, etc.) injection. Pharmaceutical and recreational drug formulations for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous, or organic solvents, or mixtures thereof, and powders. The liquid or solid pharmaceutical compositions can contain suitable pharmaceutically acceptable excipients. In some embodiments, the pharmaceutical compositions are administered by the oral or nasal respiratory route for local or systemic effect. Pharmaceutical compositions in pharmaceutically acceptable solvents can be nebulized by use of inert gases. Nebulized solutions can be inhaled directly from the nebulizing device or the nebulizing device can be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder pharmaceutical compositions can be administered, e.g., orally, or nasally, from devices that deliver the formulation in an appropriate manner. In further embodiments, in which the aminated psilocybin compounds of present disclosure are used as a recreational drug, the compounds may be included in compositions such as a food or food product, a beverage, a food seasoning, a personal care product, such as a cosmetic, perfume or bath oil, or oils (both for topical administration as massage oil, or to be burned or aerosolized). The chemical compounds of the present disclosure may also be included in a “vape” product, which may also include other drugs, such as nicotine, and flavorings. The pharmaceutical formulations comprising the chemical compounds of the present disclosure may be used to treat a subject, and in particular to treat a psychiatric disorder in a subject. Accordingly, the present disclosure includes in a further embodiment, a method for treating a psychiatric disorder, the method comprising administering to a subject in need thereof a pharmaceutical formulation comprising a chemical compound or salt thereof having formula (I): wherein at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group, and wherein R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an aryl group, or an acyl group, together with a diluent, carrier, or excipient. Psychiatric disorders that may be treated include, for example, neurodevelopmental disorders such as intellectual disability, global development delay, communication disorders, autism spectrum disorder, and attention-deficit hyperactivity disorder (ADHD); bipolar and related disorders, such as mania, and depressive episodes; anxiety disorder, such as generalized anxiety disorder (GAD), agoraphobia, social anxiety disorder, specific phobias (natural events, medical, animal, situational, for example), panic disorder, and separation anxiety disorder; stress disorders, such as acute stress disorder, adjustment disorders, post-traumatic stress disorder (PTSD), and reactive attachment disorder; dissociative disorders, such as dissociative amnesia, dissociative identity disorder, and depersonalization/derealization disorder; somatoform disorders, such as somatic symptom disorders, illness anxiety disorder, conversion disorder, and factitious disorder; eating disorders, such as anorexia nervosa, bulimia nervosa, rumination disorder, pica, and binge-eating disorder; sleep disorders, such as narcolepsy, insomnia disorder, hypersomnolence, breathing-related sleep disorders, parasomnias, and restless legs syndrome; disruptive disorders, such as kleptomania, pyromania, intermittent explosive disorder, conduct disorder, and oppositional defiant disorder; depressive disorders, such as disruptive mood dysregulation disorder, major depressive disorder, persistent depressive disorder (dysthymia), premenstrual dysphoric disorder, substance/medication-induced depressive disorder, postpartum depression, and depressive disorder caused by another medical condition for example, psychiatric and existential distress within life-threatening cancer situations (ACS Pharmacol Transl Sci 4: 553-562; J Psychiatr Res 137: 273); substance-related disorders, such as alcohol-related disorders, cannabis related disorders, inhalant-use related disorders, stimulant use disorders, and tobacco use disorders; neurocognitive disorders, such as delirium; schizophrenia; compulsive disorders, such as obsessive compulsive disorders (OCD), body dysmorphic disorder, hoarding disorder, trichotillomania disorder, excoriation disorder, substance/medication induced obsessive-compulsive disorder, and obsessive-compulsive disorder related to another medical condition; and personality disorders, such as antisocial personality disorder, avoidant personality disorder, borderline personality disorder, dependent personality disorder, histrionic personality disorder, narcissistic personality disorder, obsessive-compulsive personality disorder, paranoid personality disorder, schizoid personality disorder, and schizotypal personality disorder. In an aspect, the compounds of the present disclosure may be used to be contacted with a 5-HT2Areceptor to thereby modulate the 5-HT2Areceptor. Such contacting includes bringing a compound of the present disclosure and 5-HT2Areceptor together under in vitro conditions, for example, by introducing the compounds in a sample containing a 5-HT2Areceptor, for example, a sample containing purified 5-HT2Areceptors, or a sample containing cells comprising 5-HT2Areceptors. In vitro conditions further include the conditions described in Example 4 hereof. Contacting further includes bringing a compound of the present disclosure and 5-HT2Areceptor together under in vivo conditions. Such in vivo conditions include the administration to an animal or human subject, for example, of a pharmaceutically effective amount of the compound of the present disclosure, when the compound is formulated together with a pharmaceutically active carrier, diluent, or excipient, as hereinbefore described, to thereby treat the subject. Upon having contacted the 5-HT2Areceptor, the compound may activate the 5-HT2Areceptor or inhibit the 5-HT2Areceptor. Thus, in a further aspect, the condition that may be treated in accordance herewith can be any 5-HT2Areceptor mediated disorder. Such disorders include, but are not limited to schizophrenia, psychotic disorder, attention deficit hyperactivity disorder, autism, and bipolar disorder. In an aspect, the compounds of the present disclosure may be used to be contacted with a 5-HT1Areceptor to thereby modulate the 5-HT1Areceptor. Such contacting includes bringing a compound of the present disclosure and 5-HT1Areceptor together under in vitro conditions, for example, by introducing the compounds in a sample containing a 5-HT1Areceptor, for example, a sample containing purified 5-HT1Areceptors, or a sample containing cells comprising 5-HT1Areceptors. In vitro conditions further include the conditions described in Example 1 hereof. Contacting further includes bringing a compound of the present disclosure and 5-HT1Areceptor together under in vivo conditions. Such in vivo conditions include the administration to an animal or human subject, for example, of a pharmaceutically effective amount of the compound of the present disclosure, when the compound is formulated together with a pharmaceutically active carrier, diluent, or excipient, as hereinbefore described, to thereby treat the subject. Upon having contacted the 5-HT2Areceptor, the compound may activate the 5-HT1Areceptor or inhibit the 5-HT1Areceptor. Thus, in a further aspect, the condition that may be treated in accordance herewith can be any 5-HT1Areceptor mediated disorder. Such disorders include, but are not limited to schizophrenia, psychotic disorder, attention deficit hyperactivity disorder, autism, and bipolar disorder. The chemical compounds of the present disclosure may also be used as a feedstock material for other psilocybin derivatives. Thus in one embodiment, the chemical compounds of the present disclosure may be in used manufacture of a pharmaceutical or recreational drug formulation, wherein the manufacture may comprise derivatizing a chemical compound having the formula wherein at least one of R2, R4, R5, R6or R7is an amino group or N-substituted amino group, wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, alkyl group or O-alkyl group, wherein R4when it is not aminated is a phosphate group, a hydrogen atom, a hydroxy group, an alkyl group, or O-alkyl group, and wherein R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an aryl group, or an acyl group, or a salt of the chemical compound. In order to use the compound having formula (I) as a feedstock, one or more amino group or N-substituted amino groups may be substituted by any atoms or groups, for example hydrocarbon groups. Those of skill in the art will be generally familiar with methods that may be used to substitute amino group or N-substituted amino groups. In this respect, guidance may be found in Schnepel C. et al. (2017) Chem. Eur. J. 23:12064-12086; Durak L. J. et al. (2016) ACS Catal. 6: 1451; Runguphan W. et al. (2013) Org Lett 15: 2850; Corr M. J. et al. (2017) Chem. Sci. 8: 2039; and Roy A. D. et al. Chem. Comm. 4831. Turning now to methods of making the aminated psilocybin derivatives of the present disclosure, it is initially noted that the aminated psilocybin derivatives of the present disclosure may be prepared in any suitable manner, including by any organic chemical synthesis methods, biosynthetic methods, or a combination thereof. One suitable method of making the aminated psilocybin derivatives of the present disclosure initially involves selecting and obtaining or preparing a reactant psilocybin derivative compound, and reacting the compound under suitable conditions to form an aminated psilocybin derivative. Suitable reactant psilocybin derivative compounds include compounds comprising an indole prototype structure (see:FIG.2), including, for example, a chemical compound having formula (II): wherein at least one of R2, R4, R5, R6, or R7is a reactive group selected from a nitro group, an azido group, or a hydrogen atom, and wherein R2, R4, R5, R6, or R7which are not a reactive group, are a hydrogen atom, an alkyl of O-alkyl group, and wherein R3Aand R3Bare each independently a hydrogen atom, an alkyl group, and acyl group, or an aryl group. Reactant psilocybin derivative compound (II) comprises a plurality of compounds, some examples of which will next be described. In one example embodiment, the reactant psilocybin derivative can be selected to be a chemical compound wherein R4is an O-alkyl group, R2, R5, R6, and R7are a hydrogen atom, and R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an acyl group, or an aryl group, such as, for example, the reactant psilocybin derivative shown inFIGS.8A and8B. In one example embodiment, the reactant psilocybin derivative can be selected to be a chemical compound wherein R4is an O-acyl group, R2, R5, R6, and R7are a hydrogen atom, and R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an acyl group, or an aryl group, such as, for example, the reactant psilocybin derivative shown inFIGS.8C and8D. In one example embodiment, the reactant psilocybin derivative can be selected to be a chemical compound wherein R4is a hydroxyl group, R2, R5, R6, and R7are a hydrogen atom, and R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an acyl group, or an aryl group, such as, for example, the reactant psilocybin derivative shown inFIG.8E. In one example embodiment, the reactant psilocybin derivative can be selected to be a chemical compound wherein R4is a phosphate group, R2, R5, R6, and R7are a hydrogen atom, and R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an acyl group, or an aryl group, such as, for example, the reactant psilocybin derivative shown inFIG.8F. In one example embodiment, the reactant psilocybin derivative can be selected to be a chemical compound wherein R4is a hydrogen atom, R2, R5, R6, and R7are a hydrogen atom, and R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an acyl group, or an aryl group, such as, for example, the reactant psilocybin derivative shown inFIG.8G. The reactant psilocybin derivative compounds may be provided in a more or less chemically pure form, for example, in the form of a psilocybin derivative preparation having a purity of at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or at least 99.9%. The psilocybin derivative may be chemically synthesized, or obtained from a fine chemical manufacturer. In one example embodiment, the reactant psilocybin derivative compound, may be reacted with a nitrogenous compound with a nitrogenous compound, including, for example, a nitrogenous compound selected from nitric acid (HNO3), a nitrate salt, an acyl nitrate, trifluoromethansulfonyl nitrate, nitrosonium tetrafluoroborate (NO2BF4), and trifluoracetyl nitrate to initially form a nitrated psilocybin compound which can then be reacted under reducing conditions to form an aminated psilocybin compound. Alternatively, in order to form the aminated psilocybin derivatives of the present disclosure a nitrated or azido group containing compound reactant psilocybin compounds may be obtained and reacted under reducing conditions to form the aminated psilocybin derivatives of the present disclosure. Referring now toFIGS.9A-9B, shown therein is an example reaction wherein 5-nitro-4-O-methyl-psilocybin derivative is converted to the 5-amino-4-O-methyl-psilocybin and some example N-substituted derivatives. Referring now toFIG.9A, shown therein is an example chemical reaction wherein nitrosonium tetrafluoroborate (NO2BF4) is reacted with a 4-O-methyl-psilocybin derivative (FIG.8A) in a chemical reaction which results initially in the formation of an intermediate psilocybin derivative, notably a 4-O-methyl-5-nitro-psilocybin derivative. Subsequently, in the presence of hydrogen, the nitro group of the intermediate psilocybin derivative is reduced to an amino group under the catalytic hydrogenolysis condition with the help of palladium on charcoal. This affords the 5-amino-4-O-methyl-psilocybin derivative. Subsequent N-substitutions on the formed amino group (FIG.9B) can be carried out using (1) N-acylations, such as N-acetylation with acetic anhydride or N-sulfonylation with sulfur trioxide-pyridine complex, (2) selective N-alkylation via a reaction with an aldehyde followed by a reduction of the intermediate imine, such as the reaction with acetaldehyde to form an intermediate imine, followed by a reduction with sodium borohydride. Referring now toFIG.9D, shown therein is an example of multistep synthesis of two 4-O-methyl-psilocybin derivative respectively aminated at C5(compound 9D-8, corresponding with the compound having chemical formula (IX), set forth herein) and C7(compound 9D-10, corresponding with the compound having chemical formula (XII), set forth herein) using 4-methoxyindole (compound 9D-1) as a starting compound. Thus, starting from 9D-1, a regioselective 2-nitrovinylation can be carried out using 1-(dimethylamino)-2-nitroethylene as an electrophile in the presence of trifluoroacetic acid as a catalyst. The reaction can provide the desired (E)-3-(2-nitroethenyl)-4-methoxyindole (9D-2) which can be directly reduced using sodium borohydride in a mixture of ethanol and THF, to provide the desired 3-(2-nitroethyl)-4-methoxyindole (9D-3), for example, yielding 33% (2 steps). The nitro group of the side chain can be further reduced using lithium aluminum hydride in THE to afford the intermediate 4-O-methy-tryptamine (9D-4), for example, yielding 57%. To facilitate subsequent nitration, compound 9D-4 can be protected with an excess of di-tert-butyl decarbonate in the presence of 4-N,N-dimethylaminopyridine to provide the tri-Boc-protected 4-O-methy-tryptamine (9D-5), for example, yielding 51%. Compound 9D-5 can then be subjected to a reaction with benzoyl nitrate, generated by mixing benzoyl chloride and silver nitrate in anhydrous acetonitrile. This can provide three nitrated products respectively at C2(compound 9D-6a, e.g., 7% yield), C5(compound 9D-6b, 14% yield), and C7(compound 9D-6c, 12%). Compound 9D-6b can subsequently be subjected to a reduction using ammonium formate in the presence of 10% palladium on charcoal in methanol at room temperature to afford the 5-aminated intermediate 9D-7 (e.g., 88% yield) which can be fully deprotected subsequently by a treatment with trifluoroacetic acid to furnish the desired 5-amino-4-O-methyl-psilocybin derivative (9D-8, e.g., 56% yield, corresponding with the compound having chemical formula (IX), set forth herein). Analogously, compound 9D-6c can also subjected to a reduction using ammonium formate in the presence of 10% palladium on charcoal in methanol at room temperature to afford the 7-aminated intermediate 9D-9 (e.g., 67% yield) which can further be fully deprotected by a treatment with trifluoroacetic acid to furnish the desired 7-amino-4-O-methyl-psilocybin derivative 9D-10, (e.g., 70% yield, corresponding with the compound having chemical formula (XII), set forth herein). Thus, referring to the reactant psilocybin derivative compound having formula (II), the conditions can comprise: (i) appropriately protecting the side-chain amino group with one or two protecting groups (R3a, R3b) along with or without the protection of N1using R1. It is noted that the protecting groups R1, R3a, R3bcan be, for example, an alkyl or an acyl group, such as an acetyl group or substituted acetyl group, such as trifluoroacetyl, or other groups, such as a carbamate group, e.g., fluorenylmethyloxycarbonyl (Fmoc), benzyloxycarbonyl, or tert-butyloxycarbonyl (Boc) which can, for example, be prepared by reacting with di-tert-butyl dicarbonate in the presence of 4-N,N-dimethylaminopyridine (DMAP). It is noted that the protection of N1with R1is optional depending on the nature of electrophile which is used in the nitration reaction (referring toFIG.9D, see: e.g., reaction 9D-5 to 9D-6a, 9D-6b, 9D-6c), as well as the specific reaction conditions, e.g., pH, temperatures, solvents, catalysts; (ii) reacting the protected reactant psilocybin compound with a nitrogenous compound selected from nitric acid (HNO3); a nitrate salt, such as AgNO3; an acyl nitrate such as trifluoromethansulfonyl nitrate; benzoyl nitrate, nitrosonium tetrafluoroborate (NO2BF4), and trifluoracetyl nitrate to form a nitrated compound having chemical formula (XXXI): wherein at least one of R2, R4, R5, R6, or R7is a nitro group, and wherein each non-nitrated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not nitrated is a hydrogen atom, an alkyl group, O-alkyl group, a hydroxy group, or a phosphate group, and wherein R3Aand R3Bare a protective group, and wherein R1is a protective group or a hydrogen atom, and then (iii) reacting the nitrated compound under a reducing condition to form an aminated compound having chemical formula (XXXII): wherein at least one of R2, R4, R5, R6, or R7is an amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group, O-alkyl group, a hydroxy group, or a phosphate group, and wherein R3Aand R3Bare a protective group, and wherein R1is a protective group or a hydrogen atom, then (iv) optionally substituting the at least one amino group at R2, R4, R5, R6, or R7group to form a N-substituted derivative, and then (v) removing the protecting groups (R1, R3a, R3b) at N1and the side chain amino functionality, to thereby form a compound having chemical formula (I), and then (vi) optionally substituting the amino group of the side chain to form at least one N-substituted group. Furthermore, referring now toFIG.9C, shown therein is an alternative method for making the psilocybin derivatives of the present disclosure, notably a direct amination method using hydrogen peroxide and ammonia with the help of Cu/SiO2as a catalyst (T. Yu, R. Yang, S. Xia, G. Li, and C. HuCatal. Sci. Technol.,2014, 4, 3159-3167). The presence of a catalyst is required in view of the fact that the reaction at practical temperatures and pressures is thermodynamically unfavorable. Such catalysts may include a platinum-containing catalyst, which may be held at high temperature, or a reducible metal oxide, for example, oxides of Fe, Ni, Co, Sn, Sb, Bi or Cu, as further described in, for example, Canadian Patent 553,988, and U.S. Pat. Nos. 2,948,755; and 4,031,106. Similarly, referring further toFIG.9A, having obtained the nitrated compound, the reduction reaction is preferably conducted in the presence of a catalyst capable of providing electrons, for example, a Cu, Sn, Ni or Pt containing catalyst. The reduction reaction under acidic conditions may initially lead to the formation of an intermediate psilocybin derivative possessing a cationic NH3+group, which may be reacted with for example sodium hydroxide to form an amino group or a N-substituted amino group. Thus, it will be clear that in one embodiment, in an aspect, in the chemical compound having formula (II), at least one of R2, R4, R5, R6, or R7in the reactant psilocybin derivative compound can be a hydrogen atom, and the reaction conditions can comprise reacting the reactant psilocybin compound with ammonia and hydrogen peroxide in the presence of a catalyst, such as Cu/SiO2, to form the chemical compound having formula (I) and to then optionally substitute the at least one amino group in the chemical compound having formula (I) to form at least one N-substituted group. Furthermore, referring toFIG.9D, 1-3 protecting groups can be introduced to protect the selected substrate compound shown inFIG.8A, and the protecting groups can be a group different from Boc, such as an alkyl and/or acyl group like trifluoroacetyl, trichloroacetyl, dichloroacetyl, chloroacetyl, benzyloxycarbonyl, fluorenylmethyloxycarbonyl (Fmoc) etc. It will now be clear that, in an aspect hereof, the protected reactant psilocybin derivatives disclosed herein may be reacted with a nitrogenous compound, such as, for example, nitric acid (HNO3), a nitrate salt, an acyl nitrate such as trifluoromethansulfonyl nitrate, benzoyl nitrate and trifluoracetyl nitrate, and ammonia to form the aminated psilocybin derivatives of the present disclosure. Thus, in addition to reactant psilocybin derivative shown inFIG.8A, the example reactant psilocybin derivatives shown inFIGS.8B-8Gmay also be reacted with a suitable reagent to form example aminated psilocybin derivatives of the present disclosure. The 4-O-methyl-psilocybin derivative depicted anFIG.8Amay be reacted to form, for example, the 4-O-methyl-5-amino-psilocybin derivative depicted inFIG.5A(as already noted), the 4-O-methyl-7-amino-psilocybin derivative depicted inFIG.6A, and the 4-O-methyl-5,7-di-amino-psilocybin depicted inFIG.7A. Similarly, the 4-O-ethyl-psilocybin derivative depicted anFIG.8Bmay be reacted in similar reaction sequence to form, for example, the 4-O-ethyl-5-amino-psilocybin derivative depicted inFIG.5B, the 4-O-ethyl-7-amino-psilocybin derivative depicted inFIG.6B, and the 4-O-ethyl-5,7-di-amino-psilocybin depicted inFIG.7B. Similarly, the 4-acetyl-psilocybin derivative depicted anFIG.8Cmay be reacted in similar reaction sequence to form, for example, the 4-O-acetyl-5-amino-psilocybin derivative depicted inFIG.5C, the 4-O-acetyl-7-amino-psilocybin derivative depicted inFIG.6C, and the 4-O-acetyl-5,7-di-amino-psilocybin depicted inFIG.7C. Similarly, the 4-propanoyl-psilocybin derivative depicted anFIG.8Dmay be reacted in similar reaction sequence to form, for example, the 4-O-propanoyl-5-amino-psilocybin derivative depicted inFIG.5D, the 4-O-propanoyl-7-amino-psilocybin derivative depicted inFIG.6D, and the 4-O-propanoyl-5,7-di-amino-psilocybin depicted inFIG.7D. Similarly, the 4-hydroxy-psilocybin derivative depicted anFIG.8Emay be reacted in similar reaction sequence to form, for example, the 4-hydroxy-5-amino-psilocybin derivative depicted inFIG.5E, the 4-hydroxy-7-amino-psilocybin derivative depicted inFIG.6E, and the 4-hydroxy-5,7-di-amino-psilocybin depicted inFIG.7E. Similarly, the 4-phospho-psilocybin derivative depicted anFIG.8Fmay be reacted in similar reaction sequence to form, for example, the 4-phosphate-5-amino-psilocybin derivative depicted inFIG.5F, the 4-phosphate-7-amino-psilocybin derivative depicted inFIG.6F, and the 4-phosphate-5,7-di-amino-psilocybin depicted inFIG.7F. Similarly, the psilocybin derivative depicted anFIG.8Gmay be reacted to form, for example, the 5-amino-psilocybin derivative depicted inFIG.5G, the 47-amino-psilocybin derivative depicted inFIG.6G, and the 5,7-di-amino-psilocybin depicted inFIG.7Gas well as other analogs. It is noted that the performance of the reactions, in example different embodiments, may involve amination of different carbon atoms, i.e., the C2, C5, C6and/or C7atom. In general, reaction conditions may be selected so that different carbon atoms or combinations thereof are aminated. Thus, for example, using either a C5-nitrated or C5-azido-substituted psilocybin or derivative as a starting material, the nitro or the azido group can be reduced to afford the 5-amino-psilocybin or derivative. The methods can be used to prepare any other mono-, di- or multi-aminated psilocybin derivatives from their corresponding nitrated or azido-substituted substrates (see: Kadam, H. K; Tilve, S. G. RSC Adv. 2015, 5, 83391-83407). Typical reduction conditions can be selected from a range of conventional conditions, such as catalytic hydrogenolysis with the help of heavy metal such as palladium on charcoal, palladium hydroxide on charcoal, Raney Nickel, platinum oxide; palladium on charcoal with ammonium formate; reactive metal such as zinc, iron or copper in an acidic media or with an salt, such as zinc/ammonium chloride; organic phosphine such as triphenylphosphine or trimethylphosphine (H.-C, Wu, J.-Q. Yu, J. B. Spencer, Org. Lett., 2004, 6, 4675-4678); sulfur containing reducing agent such as sodium hydrosulfite, sodium sulfide, hydrogen sulfide; tin(I) chloride; organic silanes (R. J. Rahaim, R. E. Maleczka, Jr., Org. Lett., 2005, 7, 5087-5090). The amination on the psilocybin and derivatives can also be achieved from a precursor substrate containing either an acyl azide (—CON3) or amide (—CONH2) functionality at any of the C2, C5, C6, C7positions via respectively the Curtis rearrangement (Scriven, E. F. V.; Turnbull, K., Chemical Reviews. 1988, 88, 297-368) or Hoffmann rearrangement (Baumgarten, H.; Smith, H; Stakiis, A, J. Org. Chem 1975, 40 (24): 3554-3561). The obtained amines can be further substituted with N-alkylation or N-acylation or a combination of the two, and it can also be modified with a sulfur containing acylating agent such as sulfur trioxide-pyridine, sulfonyl chloride. Furthermore, the obtained amines can also be reacted with an aldehyde or ketone for form the corresponding imines that can be reduced subsequently. The reactions may be conducted in any suitable reaction vessel (e.g., a tube, bottle). Suitable solvents that may be used are for example, water, alcohol (such as methanol, ethanol, tetrahydrofuran (THF), N,N-dimethylformamide (DMF), dimethylsulfoxide (DMSO), or a combination of solvents. Suitable temperatures may range from, for example, e.g., from about 20° C. to about 100° C. Furthermore, reaction times may be varied. As will readily be appreciated by those of skill in the art, the reaction conditions may be optimized, for example by preparing several psilocybin derivative reactants preparations and azido and reacting these in different reaction vessels under different reaction conditions, for example, at different temperatures, using different solvents, using different catalysts etc., evaluating the obtained aminated psilocybin derivative reaction product, adjusting reaction conditions, and selecting a desired reaction condition. Further general guidance regarding appropriate reaction conditions for performing amination reactions may be found in, for example Kadarn, H. K.: Tilve, S. G. RSC Adv. 2015, 5, 83391-83407. In another aspect of the present disclosure, the aminated psilocybin compounds may be made biosynthetically. Accordingly, the present disclosure further includes, in one embodiment, a method of making an aminated psilocybin derivative the method comprising:(a) contacting a aminated psilocybin precursor compound with a host cell comprising a psilocybin biosynthetic enzyme complement, and(b) growing the host cell to produce an aminated psilocybin derivative or salts thereof having the formula (I): wherein at least one of R2, R4, R5, R6, or R7is an amino group or an N-amino substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group, and wherein R3Aand R3Bare each independently a hydrogen atom, an alkyl group, an aryl group, or an acyl group. Implementation of the foregoing example embodiment initially involves providing aminated psilocybin precursor compounds and host cells having a psilocybin biosynthetic enzyme complement. Accordingly, next, example aminated psilocybin precursor compounds and example host cells that may be selected and used in accordance with the present disclosure will be described. Thereafter, example methodologies and techniques will be described to contact and use the aminated psilocybin precursor compounds and cells to produce example aminated psilocybin compounds. A variety of aminated psilocybin precursor compounds may be selected, prepared, and used. In some embodiments, for example, the aminated psilocybin precursor compound is a compound comprising an aminated indole prototype structure. Examples of such compounds are an aminated indole, e.g., 2-amino-indole, 4-amino-indole, 5-amino-indole, 6-amino-indole, and 7-amino-indole; and aminated tryptophan derivatives, e.g., 2-amino-tryptophan, 4-amino-tryptophan, 5-amino-tryptophan, 6-amino-tryptophan, and 7-amino-tryptophan. Further aminated psilocybin precursor compounds that may be used include aminated indoles, having the formula (XXIX): wherein at least one of R2, R4, R5, R6and R7is an amino group or N-substituted amino group, wherein R2, R4, R5, R6and R7when they are not aminated are hydrogen atoms, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group. Further aminated psilocybin precursor compounds that may be used include compounds having the formula (XXVII): wherein at least one of R2, R4, R5, R6and R7is an amino group or an N-substituted group, wherein R2, R4, R5, R6and R7when they are not aminated are hydrogen atoms, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group Turning now to the host cells that can be used in accordance with the present disclosure, it is initially noted that a variety of host cells may be selected in accordance with the present disclosure, including microorganism host cells, plant host cells, and animal host cells. In accordance herewith the host cell includes a psilocybin biosynthetic enzyme complement. Such cells can be obtained in at least two ways. First, in some embodiments, host cells may be selected in which a psilocybin biosynthetic enzyme complement is naturally present. Generally cells naturally producing psilocybin for example, cells of fungal species belonging to the genusPsilocybe, are suitable in this respect. Second, in some embodiments, a host cell that not naturally produces psilocybin may be modulated to produce a psilocybin biosynthetic enzyme complement. Thus, for example, a nucleic acid sequence encoding a psilocybin biosynthetic enzyme complement may be introduced into a host cell, and upon cell growth the host cells can make the psilocybin biosynthetic enzyme complement. Typically a nucleic acid sequence encoding one or more enzymes constituting a psilocybin biosynthetic enzyme complement further includes one or more additional nucleic acid sequences, for example, a nucleic acid sequences controlling expression of the one or more enzymes, and these one or more additional nucleic acid sequences together with the nucleic acid sequence encoding the one or more enzymes can be said to form a chimeric nucleic acid sequence. A host cell which upon cultivation expresses the chimeric nucleic acid can be selected and used in accordance with the present disclosure. Suitable host cells in this respect include, for example, microbial cells, such as bacterial cells, yeast cells, for example, and algal cells or plant cells. A variety of techniques and methodologies to manipulate host cells to introduce nucleic acid sequences in cells and attain expression exists and are well known to the skilled artisan. These methods include, for example, cation based methods, for example, lithium ion or calcium ion based methods, electroporation, biolistics, and glass beads based methods. As will be known to those of skill in the art, depending on the host cell selected, the methodology to introduce nucleic acid material in the host cell may vary, and, furthermore, methodologies may be optimized for uptake of nucleic acid material by the host cell, for example, by comparing uptake of nucleic acid material using different conditions. Detailed guidance can be found, for example, in Sambrook et al., Molecular Cloning, a Laboratory Manual, Cold Spring Harbor Laboratory Press, 2012, Fourth Ed. It is noted that the chimeric nucleic acid is a non-naturally occurring chimeric nucleic acid sequence and can be said to be heterologous to the host cell. In some embodiments, the one or more enzymes constituting a psilocybin enzyme complement can be selected from by a nucleic acid sequence selected from the nucleic acid sequences consisting of:(a) SEQ. ID NO: 4, SEQ. ID NO: 8, SEQ. ID NO: 11 and SEQ. ID NO: 13;(b) a nucleic acid sequence that is substantially identical to any one of the nucleic acid sequences of (a);I a nucleic acid sequence that is substantially identical to any one of the nucleic acid sequences of (a) but for the degeneration of the genetic code;(d) a nucleic acid sequence that is complementary to any one of the nucleic acid sequences of (a);(e) a nucleic acid sequence encoding a polypeptide having any one of the amino acid sequences set forth in SEQ. ID NO: 5, SEQ. ID NO: 9, SEQ. ID NO: 12 and SEQ. ID NO: 14;(f) a nucleic acid sequence that encodes a functional variant of any one of the amino acid sequences set forth in SEQ. ID NO: 5, SEQ. ID NO: 9, SEQ. ID NO: 12 and SEQ. ID NO: 14; and(g) a nucleic acid sequence that hybridizes under stringent conditions to any one of the nucleic acid sequences set forth in (a), (b), (c), (d), (e) or (f). Thus any of the nucleic acid sequence set forth in (a), (b), (c), (d), (e), (f) or (g) may be selected and introduced into a host cell. In particular, however the nucleic acid sequence is selected in conjunction with the selected psilocybin precursor compound, as hereinafter further discussed in reference withFIG.10. One example host cell that conveniently may be used isEscherichia coli. The preparation of theE. colivectors may be accomplished using commonly known techniques such as restriction digestion, ligation, gel electrophoresis, DNA sequencing, the polymerase chain reaction (PCR) and other methodologies. A wide variety of cloning vectors is available to perform the necessary steps required to prepare a recombinant expression vector. Among the vectors with a replication system functional inE. coli, are vectors such as pBR322, the pUC series of vectors, the M13 mp series of vectors, pBluescript etc. Suitable promoter sequences for use inE. coliinclude, for example, the T7 promoter, the T5 promoter, tryptophan (trp) promoter, lactose (lac) promoter, tryptophan/lactose (tac) promoter, lipoprotein (lpp) promoter, and A phage PL promoter. Typically, cloning vectors contain a marker, for example, an antibiotic resistance marker, such as ampicillin or kanamycin resistance marker, allowing selection of transformed cells. Nucleic acid sequences may be introduced in these vectors, and the vectors may be introduced inE. coliby preparing competent cells, electroporation or using other well known methodologies to a person of skill in the art.E. colimay be grown in an appropriate medium, such as Luria-Broth medium and harvested. Recombinant expression vectors may readily be recovered from cells upon harvesting and lysing of the cells. Another example host cell that may be conveniently used is a yeast cell. Example yeast host cells that can be used are yeast cells belonging to the genusCandida, Kluyveromyces, Saccharomyces, Schizosaccharomyces, Pichia, Hansenula, andYarrowia. In specific example embodiments, the yeast cell can be aSaccharomyces cerevisiaecell, aYarrowia lipolyticacell, orPichia pastoriscell. A number of vectors exist for the expression of recombinant proteins in yeast host cells. Examples of vectors that may be used in yeast host cells include, for example, Yip type vectors, YEp type vectors, YRp type vectors, YCp type vectors, pGPD-2, pAO815, pGAPZ, pGAPZα, pHIL-D2, pHIL-S1, pPIC3.5K, pPIC9K, pPICZ, pPICZα, pPIC3K, pHWO10, pPUZZLE and 2 μm plasmids. Such vectors are known to the art and are, for example, described in Cregg et al, Mol Biotechnol. (2000) 16(1): 23-52. Suitable promoter sequences for use in yeast host cells are also known and described, for example, in Mattanovich et al., Methods Mol Biol, 2012, 824:329-58, and in Romanos et al, 1992, Yeast 8: 423-488. Examples of suitable promoters for use in yeast host cells include promoters of glycolytic enzymes, like triosephosphate isomerase (TPI), phosphoglycerate kinase (PGK), glyceraidehyde-3-phosphate dehydrogenase (GAPDH or GAP) and variants thereof, lactase (LAC) and galactosidase (GAL),P. pastorisglucose-6-phosphate isomerase promoter (PPGI), the 3-phosphoglycerate kinase promoter (PPGK), the glycerol aldehyde phosphate dehydrogenase promoter (PGAP), translation elongation factor promoter (PTEF),S. cerevisiaeenolase (ENO-1),S. cerevisiaegalactokinase (GAL1),S. cerevisiaealcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase (ADH1, ADH2/GAP),S. cerevisiaetriose phosphate isomerase (TPI),S. cerevisiaemetallothionein (CUP1), andS. cerevisiae3-phosphoglycerate kinase (PGK), and the maltase gene promoter (MAL). Marker genes suitable for use in yeast host cells are also known to the art. Thus, antibiotic resistance markers, such as ampicillin resistance markers, can be used in yeast, as well as marker genes providing genetic functions for essential nutrients, for example, leucine (LEU2), tryptophan (TRP1 and TRP2), uracil (URA3, URA5, URA6), histidine (HIS3), and the like. Methods for introducing vectors into yeast host cells can, for example, be found in S. Kawai et al., 2010, Bioeng. Bugs 1(6): 395-403. Further, guidance with respect to the preparation of expression vectors and introduction thereof into host cells, including inE. colicells, yeast cells, and other host cells, may be found in, for example: Sambrook et al., Molecular Cloning, a Laboratory Manual, Cold Spring Harbor Laboratory Press, 2012, Fourth Ed. Thus, to briefly recap, a host cell comprising a chimeric nucleic acid comprising (i) a nucleic acid sequence controlling expression in a host cell and (ii) a nucleic acid sequence encoding a psilocybin biosynthetic enzyme complement, can be prepared in accordance with the present disclosure. In accordance herewith, host cells are grown to multiply and to express a chimeric nucleic acid. Expression of the chimeric nucleic acid results in the biosynthetic production in the host cell of a psilocybin biosynthetic enzyme complement. Growth media and growth conditions can vary depending on the host cell that is selected, as will be readily appreciated to those of ordinary skill in the art. Growth media typically contain a carbon source, one or several nitrogen sources, essential salts including salts of potassium, sodium, magnesium, phosphate and sulphate, trace metals, water soluble vitamins, and process aids including but not limited to antifoam agents, protease inhibitors, stabilizers, ligands, and inducers. Example carbon sources are e.g., mono- or disaccharides. Example nitrogen sources are, e.g., ammonia, urea, amino acids, yeast extract, corn steep liquor and fully or partially hydrolyzed proteins. Example trace metals are e.g., Fe, Zn, Mn, Cu, Mo and H3BO3. Example water soluble vitamins are e.g. biotin, pantothenate, niacin, thiamine, p-aminobenzoic acid, choline, pyridoxine, folic acid, riboflavin, and ascorbic acid. Further, specific example media include liquid culture media for the growth of yeast cells and bacterial cells including, Luria-Bertani (LB) broth for bacterial cell cultivation, and yeast extract peptone dextrose (YEPD or YPD), for yeast cell cultivation. Further media and growth conditions can be found in Sambrook et al., Molecular Cloning, a Laboratory Manual, Cold Spring Harbor Laboratory Press, 2012, Fourth Ed. In order for the host cells to produce the aminated psilocybin compound, the cells are provided with a precursor compound. Thus in accordance herewith, host cells may be contacted with a psilocybin precursor compound. In some embodiments, a psilocybin precursor compound can be exogenously supplied, for example, by including a psilocybin precursor compound in the growth medium of the host cells, and growing the host cells in a medium including the psilocybin precursor compound. Referring next toFIG.10, shown therein is an example biosynthetic pathway showing the conversion of example psilocybin precursor compounds to form an aminated psilocybin. Thus, as can be appreciated fromFIG.10, various psilocybin precursor compounds may be selected and prepared in aminated form, in conjunction with a psilocybin biosynthetic enzyme complement. Thus, by way of example, aminated tryptophan (e.g., 2-,5-, 6-, or 7-aminated tryptophan) may be selected and contacted with a host cell comprising a psilocybin biosynthetic enzyme complement comprising tryptophan decarboxylase and optionally N-acetyl transferase, and upon growth of the cells aminated psilocybin derivatives can be formed. By way of further example, aminated indole (e.g., 2-,5-, 6-, or 7-aminated indole) may be selected and contacted with a host cell comprising a psilocybin biosynthetic enzyme complement comprising tryptophan synthase subunit B polypeptide and tryptophan decarboxylase and optionally N-acetyl transferase, and upon growth of the cells aminated psilocybin derivatives can be formed In some embodiments, the psilocybin precursor compound can be an aminated psilocybin precursor compound which is exogenously supplied to a host cell, for example by inclusion in the host cell's growth medium. Thus, for example, referring toFIG.10, it will be understood that in accordance herewith, for example, 7-amino-indole or 7-amino-tryptophan, may be included in the growth medium of a host cell comprising a psilocybin biosynthetic enzyme complement. Referring toFIG.10, in a further example embodiment, the aminated psilocybin precursor compound can be an aminated indole, having the formula (XXIX): wherein at least one of R2, R4, R5, R6and R7is an amino group or N-substituted amino group, wherein R2, R4, R5, R6and R7when they are not aminated are hydrogen atoms, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl, O-alkyl or O-aryl group, a hydroxy group, or a phosphate group, the psilocybin biosynthetic enzyme complement can comprise:(i) a tryptophan synthase subunit B polypeptide encoded by a nucleic acid selected from:(a) SEQ. ID NO: 8;(b) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a);(c) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a) but for the degeneration of the genetic code;(d) a nucleic acid sequence that is complementary to the nucleic acid sequence of (a);(e) a nucleic acid sequence encoding a polypeptide having any one of the amino acid sequences set forth in SEQ. ID NO: 9;(f) a nucleic acid sequence that encodes a functional variant of any one of the amino acid sequences set forth in SEQ. ID NO: 9; and(g) a nucleic acid sequence that hybridizes under stringent conditions to any one of the nucleic acid sequences set forth in (a), (b), (c), (d), (e) or (f); and(ii) a tryptophan decarboxylase encoded by a nucleic acid sequence selected from:(a) SEQ. ID NO: 11;(b) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a);(c) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a) but for the degeneration of the genetic code;(d) a nucleic acid sequence that is complementary to the nucleic acid sequence of (a);(e) a nucleic acid sequence encoding a polypeptide having any one of the amino acid sequences set forth in SEQ. ID NO: 12;(f) a nucleic acid sequence that encodes a functional variant of any one of the amino acid sequences set forth in SEQ. ID NO: 12; and(g) a nucleic acid sequence that hybridizes under stringent conditions to any one of the nucleic acid sequences set forth in (a), (b), (c), (d), (e) or (f); and the formed aminated psilocybin derivative can be a compound having formula (XXVIII): wherein at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group, and wherein at least one of R3Aand R3Bare hydrogen atom. Referring further toFIG.10, in another example embodiment, the aminated psilocybin precursor compound can be a compound, having the formula (XXVII): wherein at least one of R2, R4, R5, R6and R7is an amino group or an N-substituted amino group, wherein R2, R4, R5, R6and R7when they are not aminated are hydrogen atoms, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group; the psilocybin biosynthetic enzyme complement can comprise: a tryptophan decarboxylase encoded by a nucleic acid sequence selected from:(a) SEQ. ID NO: 11;(b) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a);(c) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a) but for the degeneration of the genetic code;(d) a nucleic acid sequence that is complementary to the nucleic acid sequence of (a);(e) a nucleic acid sequence encoding a polypeptide having any one of the amino acid sequences set forth in SEQ. ID NO: 12;(f) a nucleic acid sequence that encodes a functional variant of any one of the amino acid sequences set forth in SEQ. ID NO: 12; and(g) a nucleic acid sequence that hybridizes under stringent conditions to any one of the nucleic acid sequences set forth in (a), (b), (c), (d), (e) or (f); and the formed aminated psilocybin derivative can be a compound having formula (XXVIII): wherein at least one of R2, R4, R5, R6, or R7is an amino group or N-substituted amino group, and wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a hydrogen atom, an alkyl group or O-alkyl group, a hydroxy group, or a phosphate group, and wherein at least one of R3Aand R3Bare hydrogen atom. In some embodiments, in formula (XXVIII) R3Aand R3Bare each a hydrogen atom. Referring again toFIG.10, the psilocybin biosynthetic enzyme complement can, in addition to the aforementioned tryptophan decarboxylase and tryptophan synthase subunit B polypeptide further comprise an N-acetyl transferase. In at least one embodiment, in an aspect, the N-acetyl transferase can be an enzyme encoded by a nucleic acid sequence selected from:(a) SEQ. ID NO: 4;(b) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a);(c) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a) but for the degeneration of the genetic code;(d) a nucleic acid sequence that is complementary to the nucleic acid sequence of (a);(e) a nucleic acid sequence encoding a polypeptide having any one of the amino acid sequences set forth in SEQ. ID NO: 5;(f) a nucleic acid sequence that encodes a functional variant of any one of the amino acid sequences set forth in SEQ. ID NO: 5; and(g) a nucleic acid sequence that hybridizes under stringent conditions to any one of the nucleic acid sequences set forth in (a), (b), (c), (d), (e) or (f). In at least one embodiment, in an aspect, the formed aminated psilocybin compound can have the formula (XXX): wherein at least one of R2, R4, R5, R6or R7is an amino group or N-substituted amino group, wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, an alkyl group or O-alkyl group, wherein R4when it is not aminated is a phosphate group, a hydrogen atom or an alkyl group or O-alkyl group. Referring again toFIG.10, the psilocybin biosynthetic enzyme complement can, in addition to the aforementioned tryptophan decarboxylase and tryptophan synthase subunit B polypeptide further comprise an N-methyl transferase. In at least one embodiment, in an aspect, the N-methyl transferase can be an enzyme encoded by a nucleic acid sequence selected from:(a) SEQ. ID NO: 13;(b) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a);(c) a nucleic acid sequence that is substantially identical to the nucleic acid sequence of (a) but for the degeneration of the genetic code;(d) a nucleic acid sequence that is complementary to the nucleic acid sequence of (a);(e) a nucleic acid sequence encoding a polypeptide having any one of the amino acid sequences set forth in SEQ. ID NO: 14;(f) a nucleic acid sequence that encodes a functional variant of the amino acid sequence set forth in SEQ. ID NO: 14; and(g) a nucleic acid sequence that hybridizes under stringent conditions to any one of the nucleic acid sequences set forth in (a), (b), (c), (d), (e) or (f). In at least one embodiment, in an aspect, the formed aminated psilocybin compound can have the chemical formula (XXXIII): wherein at least one of R2, R4, R5, R6or R7is an amino group or substituted amino group, wherein each non-aminated R2, R5, R6, or R7is a hydrogen atom, or an alkyl group or O-alkyl group, wherein R4when it is not aminated is a phosphate group, a hydrogen atom or an alkyl group or O-alkyl group, and wherein at least one of R3aand R3bis an amino group, and wherein a non-aminated R3aand R3bis a hydrogen atom. It will be clear to those of skill in the art that a significant variety of different aminated psilocybin precursor compounds may be selected.FIG.10in this respect provides guidance and allows a person of skill in the art to select appropriate psilocybin precursor compounds and a matching a psilocybin biosynthetic enzyme complement. Upon production by the host cells of the aminated psilocybin compounds in accordance with the methods of the present disclosure, the aminated psilocybin compounds may be extracted from the host cell suspension, and separated from other constituents within the host cell suspension, such as media constituents and cellular debris. Separation techniques will be known to those of skill in the art and include, for example, solvent extraction (e.g., butane, chloroform, ethanol), column chromatography based techniques, high-performance liquid chromatography (HPLC), for example, and/or countercurrent separation (CCS) based systems. The recovered aminated psilocybin compounds may be obtained in a more or less pure form, for example, a preparation of aminated psilocybin compounds of at least about 60% (w/w), about 70% (w/w), about 80% (w/w), about 90% (w/w), about 95% (w/w), about 96% (w/w), about 97% (w/w), about 98% (w/w) or about 99% (w/w) purity may be obtained. Thus, in this manner, aminated psilocybin derivatives in more or less pure form may be prepared. Similarly, other methods of making the aminated psilocybin compounds that may be used in accordance herewith may yield preparations of aminated compounds of at least about 60% (w/w), about 70% (w/w), about 80% (w/w), about 90% (w/w), about 95% (w/w), about 96% (w/w), about 97% (w/w), about 98% (w/w), or about 99% (w/w) purity. It will now be clear form the foregoing that novel aminated psilocybin derivatives are disclosed herein. The aminated psilocybin compounds may be formulated for use as a pharmaceutical drug or recreational drug. The aminated psilocybin compounds may also be used as a feedstock to produce other psilocybin derivatives. Hereinafter are provided examples of specific implementations for performing the methods of the present disclosure, as well as implementations representing the compositions of the present disclosure. The examples are provided for illustrative purposes only, and are not intended to limit the scope of the present disclosure in any way. Summary of Sequences SEQ. ID NO: 1 sets forth a nucleic acid sequence of pCDM4 vector. SEQ. ID NO: 2 sets forth a nucleic acid sequence encoding a synthetic FLAG epitope tag polypeptide. SEQ. ID NO: 3 sets forth deduced amino acid sequence of a synthetic FLAG epitope tag polypeptide. SEQ. ID NO: 4 sets forth a nucleic acid sequence encoding aStreptomyces griseofuscusPsmF N-acetyltransferase polypeptide. SEQ. ID NO: 5 sets forth a deduced amino acid sequence of aStreptomyces griseofuscusPsmF N-acetyltransferase polypeptide. SEQ. ID NO: 6 sets forth a nucleic acid sequence encoding a synthetic V5 epitope tag polypeptide. SEQ. ID NO: 7 sets forth deduced amino acid sequence of a synthetic V5 epitope tag polypeptide. SEQ. ID NO: 8 sets forth a nucleic acid sequence encoding a mutatedThermotoga maritimaTmTrpB-2F3 tryptophan synthase subunit B polypeptide. SEQ. ID NO: 9 sets forth a deduced amino acid sequence of a mutatedThermotoga maritimaTmTrpB-2F3 tryptophan synthase subunit B polypeptide. SEQ. ID NO: 10 sets forth a nucleic acid sequence of pETM6-H10 vector SEQ. ID NO: 11 sets forth a nucleic acid sequence encoding aBacillus atrophaeusBaTDC tryptophan decarboxylase polypeptide. SEQ. ID NO: 12 sets forth a deduced amino acid sequence of aBacillus atrophaeusBaTDC tryptophan decarboxylase polypeptide. SEQ. ID NO: 13 sets forth anEscherichia colicodon optimized nucleic acid sequence encoding aRhinella marinaN-methyltransferase polypeptide. SEQ. ID NO: 14 sets forth a deduced amino acid sequence of aRhinella marinaN-methyltransferase polypeptide. SEQUENCESSEQ. ID NO: 1GCTAACAGCGCGATTTGCTGGTGACCCAATGCGACCAGATGCTCCACGCCCAGTCGCGTACCGTCTTCATGGGAGAAAATAATACTGTTGATGGGTGTCTGGTCAGAGACATCAAGAAATAACGCCGGAACATTAGTGCAGGCAGCTTCCACAGCAATGGCATCCTGGTCATCCAGCGGATAGTTAATGATCAGCCCACTGACGCGTTGCGCGAGAAGATTGTGCACCGCCGCTTTACAGGCTTCGACGCCGCTTCGTTCTACCATCGACACCACCACGCTGGCACCCAGTTGATCGGCGCGAGATTTAATCGCCGCGACAATTTGCGACGGCGCGTGCAGGGCCAGACTGGAGGTGGCAACGCCAATCAGCAACGACTGTTTGCCCGCCAGTTGTTGTGCCACGCGGTTGGGAATGTAATTCAGCTCCGCCATCGCCGCTTCCACTTTTTCCCGCGTTTTCGCAGAAACGTGGCTGGCCTGGTTCACCACGCGGGAAACGGTCTGATAAGAGACACCGGCATACTCTGCGACATCGTATAACGTTACTGGTTTCACATTCACCACCCTGAATTGACTCTCTTCCGGGCGCTATCATGCCATACCGCGAAAGGTTTTGCGCCATTCGATGGTGTCCGGGATCTCGACGCTCTCCCTTATGCGACTCCTGCATTAGGAAGCAGCCCAGTAGTAGGTTGAGGCCGTTGAGCACCGCCGCCGCAAGGAATGGTGCATGCAAGGAGATGGCGCCCAACAGTCCCCCGGCCACGGGGCCTGCCACCATACCCACGCCGAAACAAGCGCTCATGAGCCCGAAGTGGCGAGCCCGATCTTCCCCATCGGTGATGTCGGCGATATAGGCGCCAGCAACCGCACCTGTGGCGCCGGTGATGCCGGCCACGATGCGTCCGGCGTAGCCTAGGATCGAGATCGATCTCGATCCCGCGAAATTAATACGACTCACTATAGGGGAATTGTGAGCGGATAACAATTCCCCTCTAGAAATAATTTTGTTTAACTTTAAGAAGGAGATATACATATGGCAGATCTCAATTGGATATCGGCCGGCCACGCGATCGCTGACGTCGGTACCCTCGAGTCTGGTAAAGAAACCGCTGCTGCGAAATTTGAACGCCAGCACATGGACTCGTCTACTAGTCGCAGCTTAATTAACCTAAACTGCTGCCACCGCTGAGCAATAACTAGCATAACCCCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTAGCGAAAGGAGGAGTCGACACTGCTTCCGGTAGTCAATAAACCGGTAAACCAGCAATAGACATAAGCGGCTATTTAACGACCCTGCCCTGAACCGACGACCGGGTCATCGTGGCCGGATCTTGCGGCCCCTCGGCTTGAACGAATTGTTAGACATTATTTGCCGACTACCTTGGTGATCTCGCCTTTCACGTAGTGGACAAATTCTTCCAACTGATCTGCGCGCGAGGCCAAGCGATCTTCTTCTTGTCCAAGATAAGCCTGTCTAGCTTCAAGTATGACGGGCTGATACTGGGCCGGCAGGCGCTCCATTGCCCAGTCGGCAGCGACATCCTTCGGCGCGATTTTGCCGGTTACTGCGCTGTACCAAATGCGGGACAACGTAAGCACTACATTTCGCTCATCGCCAGCCCAGTCGGGCGGCGAGTTCCATAGCGTTAAGGTTTCATTTAGCGCCTCAAATAGATCCTGTTCAGGAACCGGATCAAAGAGTTCCTCCGCCGCTGGACCTACCAAGGCAACGCTATGTTCTCTTGCTTTTGTCAGCAAGATAGCCAGATCAATGTCGATCGTGGCTGGCTCGAAGATACCTGCAAGAATGTCATTGCGCTGCCATTCTCCAAATTGCAGTTCGCGCTTAGCTGGATAACGCCACGGAATGATGTCGTCGTGCACAACAATGGTGACTTCTACAGCGCGGAGAATCTCGCTCTCTCCAGGGGAAGCCGAAGTTTCCAAAAGGTCGTTGATCAAAGCTCGCCGCGTTGTTTCATCAAGCCTTACGGTCACCGTAACCAGCAAATCAATATCACTGTGTGGCTTCAGGCCGCCATCCACTGCGGAGCCGTACAAATGTACGGCCAGCAACGTCGGTTCGAGATGGCGCTCGATGACGCCAACTACCTCTGATAGTTGAGTCGATACTTCGGCGATCACCGCTTCCCTCATACTCTTCCTTTTTCAATATTATTGAAGCATTTATCAGGGTTATTGTCTCATGAGCGGATACATATTTGAATGTATTTAGAAAAATAAACAAATAGCCAGCTCACTCGGTCGCTACGCTCCGGGCGTGAGACTGCGGCGGGCGCTGCGGACACATACAAAGTTACCCACAGATTCCGTGGATAAGCAGGGGACTAACATGTGAGGCAAAACAGCAGGGCCGCGCCGGTGGCGTTTTTCCATAGGCTCCGCCCTCCTGCCAGAGTTCACATAAACAGACGCTTTTCCGGTGCATCTGTGGGAGCCGTGAGGCTCAACCATGAATCTGACAGTACGGGCGAAACCCGACAGGACTTAAAGATCCCCACCGTTTCCGGCGGGTCGCTCCCTCTTGCGCTCTCCTGTTCCGACCCTGCCGTTTACCGGATACCTGTTCCGCCTTTCTCCCTTACGGGAAGTGTGGCGCTTTCTCATAGCTCACACACTGGTATCTCGGCTCGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTAAGCAAGAACTCCCCGTTCAGCCCGACTGCTGCGCCTTATCCGGTAACTGTTCACTTGAGTCCAACCCGGAAAAGCACGGTAAAACGCCACTGGCAGCAGCCATTGGTAACTGGGAGTTCGCAGAGGATTTGTTTAGCTAAACACGCGGTTGCTCTTGAAGTGTGCGCCAAAGTCCGGCTACACTGGAAGGACAGATTTGGTTGCTGTGCTCTGCGAAAGCCAGTTACCACGGTTAAGCAGTTCCCCAACTGACTTAACCTTCGATCAAACCACCTCCCCAGGTGGTTTTTTCGTTTACAGGGCAAAAGATTACGCGCAGAAAAAAAGGATCTCAAGAAGATCCTTTGATCTTTTCTACTGAACCGCTCTAGATTTCAGTGCAATTTATCTCTTCAAATGTAGCACCTGAAGTCAGCCCCATACGATATAAGTTGTAATTCTCATGTTAGTCATGCCCCGCGCCCACCGGAAGGAGCTGACTGGGTTGAAGGCTCTCAAGGGCATCGGTCGAGATCCCGGTGCCTAATGAGTGAGCTAACTTACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCGTGCCAGCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCCAGGGTGGTTTTTCTTTTCACCAGTGAGACGGGCAACAGCTGATTGCCCTTCACCGCCTGGCCCTGAGAGAGTTGCAGCAAGCGGTCCACGCTGGTTTGCCCCAGCAGGCGAAAATCCTGTTTGATGGTGGTTAACGGCGGGATATAACATGAGCTGTCTTCGGTATCGTCGTATCCCACTACCGAGATGTCCGCACCAACGCGCAGCCCGGACTCGGTAATGGCGCGCATTGCGCCCAGCGCCATCTGATCGTTGGCAACCAGCATCGCAGTGGGAACGATGCCCTCATTCAGCATTTGCATGGTTTGTTGAAAACCGGACATGGCACTCCAGTCGCCTTCCCGTTCCGCTATCGGCTGAATTTGATTGCGAGTGAGATATTTATGCCAGCCAGCCAGACGCAGACGCGCCGAGACAGAACTTAATGGGCCCSEQ. ID NO: 2GACTACAAGGATGACGATGACAAASEQ. ID NO: 3DYKDDDDKSEQ. ID NO: 4ATGAACACCTTCAGAACAGCCACTGCCAGAGACATACCTGATGTAGCAGCAACTCTTACGGAAGCCTTCGCAACTGATCCACCCACGCAGTGGGTGTTCCCCGACGGTACTGCCGCCGTCAGCAGGTTCTTTACACATGTTGCAGATAGGGTTCACACGGCCGGTGGTATTGTTGAGCTACTACCAGACAGAGCCGCCATGATTGCATTGCCACCACACGTGAGGCTGCCAGGAGAAGCTGCCGACGGAAGGCAGGCGGAAATTCAGAGAAGGCTGGCAGACAGGCACCCGCTGACACCTCACTACTACCTGCTGTTTTACGGAGTTAGAACGGCACACCAGGGTTCGGGATTGGGCGGAAGAATGCTGGCCAGATTAACTAGCAGAGCTGATAGGGACAGGGTGGGTACATATACTGAGGCATCCACCTGGCGTGGCGCTAGACTGATGCTGAGACATGGATTCCATGCTACAAGGCCACTAAGATTGCCAGATGGACCCAGCATGTTTCCACTTTGGAGAGATCCAATCCATGATCATTCTGATTAGSEQ. ID NO: 5MNTFRTATARDIPDVAATLTEAFATDPPTQWVFPDGTAAVSRFFTHVADRVHTAGGIVELLPDRAAMIALPPHVRLPGEAADGRQAEIQRRLADRHPLTPHYYLLFYGVRTAHQGSGLGGRMLARLTSRADRDRVGTYTEASTWRGARLMLRHGFHATRPLRLPDGPSMFPLWRDPIHDHSDSEQ. ID NO: 6GGTAAGCCAATTCCAAATCCTTTGTTGGGTTTGGACTCCACCSEQ. ID NO: 7GKPIPNPLLGLDSTSEQ. ID NO: 8ATGAAAGGATATTTCGGACCATACGGTGGCCAGTACGTACCAGAAATATTAATGGGTGCCTTAGAGGAGTTAGAGGCAGCATACGAGGAGATTATGAAGGATGAGAGCTTCTGGAAGGAGTTCAACGATCTACTGAGGGATTACGCAGGCAGACCAACGCCATTGTACTTTGCCAGGAGATTGTCTGAGAAGTACGGCGCCCGTGTTTACTTGAAGCGTGAGGATCTGCTGCACACTGGAGCACACAAGATAAATAACGCTATCGGACAGGTTTTATTGGCCAAATTAATGGGGAAGACACGTATCATAGCCGAGACGGGAGCTGGGCAGCATGGAGTCGCTACTGCTACCGCTGCTGCCCTGTTCGGAATGGAATGTGTGATCTACATGGGTGAAGAGGACACAATCAGACAGAAGTTGAACGTGGAGCGTATGAAATTATTAGGGGCTAAAGTTGTCCCTGTTAAGTCTGGCAGTAGGACCTTGAAGGATGCGATAGACGAGGCTTTGAGAGACTGGATTACTAATTTACAGACAACATATTATGTTATCGGATCTGTTGTTGGTCCCCACCCTTACCCAATTATCGTAAGGAATTTCCAGAAGGTTATCGGTGAGGAGACCAAGAAGCAAATACCAGAAAAGGAAGGTCGTTTGCCAGACTATATAGTTGCCTGCGTAGGCGGCGGTAGCAATGCCGCAGGTATATTTTACCCATTCATAGACTCTGGAGTAAAGCTGATAGGTGTTGAGGCAGGTGGCGAGGGATTGGAGACAGGTAAACACGCAGCCTCGTTATTAAAGGGTAAAATTGGCTATTTACATGGATCGAAGACCTTTGTTCTACAAGATGACTGGGGTCAAGTCCAAGTGAGCCATTCGGTGTCAGCTGGTCTTGACTATTCAGGAGTAGGACCTGAGCATGCTTATTGGAGAGAGACAGGGAAGGTTCTGTACGACGCAGTGACTGACGAAGAGGCTTTGGACGCATTTATAGAGTTATCAAGACTAGAGGGCATTATACCCGCTTTAGAGTCATCGCATGCTCTAGCATATTTGAAGAAGATAAATATAAAAGGTAAGGTTGTGGTGGTCAACCTATCAGGGAGAGGGGATAAAGACCTGGAGTCAGTCTTAAACCATCCATACGTGAGAGAAAGAATTAGATGASEQ. ID NO: 9MKGYFGPYGGQYVPEILMGALEELEAAYEEIMKDESFWKEFNDLLRDYAGRPTPLYFARRLSEKYGARVYLKREDLLHTGAHKINNAIGQVLLAKLMGKTRIIAETGAGQHGVATATAAALFGMECVIYMGEEDTIRQKLNVERMKLLGAKVVPVKSGSRTLKDAIDEALRDWITNLQTTYYVIGSVVGPHPYPIIVRNFQKVIGEETKKQIPEKEGRLPDYIVACVGGGSNAAGIFYPFIDSGVKLIGVEAGGEGLETGKHAASLLKGKIGYLHGSKTFVLQDDWGQVQVSHSVSAGLDYSGVGPEHAYWRETGKVLYDAVTDEEALDAFIELSRLEGIIPALESSHALAYLKKINIKGKVVVVNLSGRGDKDLESVLNHPYVRERIRSEQ. ID NO: 10GAAGAATTGTGAGCGGATAACAATTCCCCTCTAGAAATAATTTTGTTTAACTTTAAGAAGGAGATATACATATGGCAGATCTCAATTGGATATCGGCCGGCCACGCGATCGCTGACGTCGGTACCCTCGAGTCTGGTAAAGAAACCGCTGCTGCGAAATTTGAACGCCAGCACATGGACTCGTCTACTAGTCGCAGCTTAATTAACCTAAACTGCTGCCACCGCTGAGCAATAACTAGCATAACCCCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTAGCGAAAGGAGGAGTCGACTATATCCGGATTGGCGAATGGGACGCGCCCTGTAGCGGCGCATTAAGCGCGGCGGGTGTGGTGGTTACGCGCAGCGTGACCGCTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCGCTTTCTTCCCTTCCTTTCTCGCCACGTTCGCCGGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCTTTAGGGTTCCGATTTAGTGCTTTACGGCACCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCACGTAGTGGGCCATCGCCCTGATAGACGGTTTTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAGTGGACTCTTGTTCCAAACTGGAACAACACTCAACCCTATCTCGGTCTATTCTTTTGATTTATAAGGGATTTTGCCGATTTCGGCCTATTGGTTAAAAAATGAGCTGATTTAACAAAAATTTAACGCGAATTTTAACAAAATATTAACGTTTACAATTTCTGGCGGCACGATGGCATGAGATTATCAAAAAGGATCTTCACCTAGATCCTTTTAAATTAAAAATGAAGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCTGACAGTTACCAATGCTTAATCAGTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAGTTGCCTGACTCCCCGTCGTGTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTGCAATGATACCGCGAGACCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCGGAAGGGCCGAGCGCAGAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTTGCCGGGAAGCTAGAGTAAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTACAGGCATCGTGGTGTCACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATCAAGGCGAGTTACATGATCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGATCGTTGTCAGAAGTAAGTTGGCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCTCTTACTGTCATGCCATCCGTAAGATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCTGAGAATAGTGTATGCGGCGACCGAGTTGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGCCACATAGCAGAACTTTAAAAGTGCTCATCATTGGAAAACGTTCTTCGGGGCGAAAACTCTCAAGGATCTTACCGCTGTTGAGATCCAGTTCGATGTAACCCACTCGTGCACCCAACTGATCTTCAGCATCTTTTACTTTCACCAGCGTTTCTGGGTGAGCAAAAACAGGAAGGCAAAATGCCGCAAAAAAGGGAATAAGGGCGACACGGAAATGTTGAATACTCATACTCTTCCTTTTTCAATCATGATTGAAGCATTTATCAGGGTTATTGTCTCATGAGCGGATACATATTTGAATGTATTTAGAAAAATAAACAAATAGGTCATGACCAAAATCCCTTAACGTGAGTTTTCGTTCCACTGAGCGTCAGACCCCGTAGAAAAGATCAAAGGATCTTCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCTGCTTGCAAACAAAAAAACCACCGCTACCAGCGGTGGTTTGTTTGCCGGATCAAGAGCTACCAACTCTTTTTCCGAAGGTAACTGGCTTCAGCAGAGCGCAGATACCAAATACTGTCCTTCTAGTGTAGCCGTAGTTAGGCCACCACTTCAAGAACTCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTGTTACCAGTGGCTGCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAGTTACCGGATAAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACAGCCCAGCTTGGAGCGAACGACCTACACCGAACTGAGATACCTACAGCGTGAGCTATGAGAAAGCGCCACGCTTCCCGAAGGGAGAAAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGGGAGCTTCCAGGGGGAAACGCCTGGTATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTTGAGCGTCGATTTTTGTGATGCTCGTCAGGGGGGCGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTTTTTACGGTTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCCCCTGATTCTGTGGATAACCGTATTACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGAACGACCGAGCGCAGCGAGTCAGTGAGCGAGGAAGCGGAAGAGCGCCTGATGCGGTATTTTCTCCTTACGCATCTGTGCGGTATTTCACACCGCATATATGGTGCACTCTCAGTACAATCTGCTCTGATGCCGCATAGTTAAGCCAGTATACACTCCGCTATCGCTACGTGACTGGGTCATGGCTGCGCCCCGACACCCGCCAACACCCGCTGACGCGCCCTGACGGGCTTGTCTGCTCCCGGCATCCGCTTACAGACAAGCTGTGACCGTCTCCGGGAGCTGCATGTGTCAGAGGTTTTCACCGTCATCACCGAAACGCGCGAGGCAGCTGCGGTAAAGCTCATCAGCGTGGTCGTGAAGCGATTCACAGATGTCTGCCTGTTCATCCGCGTCCAGCTCGTTGAGTTTCTCCAGAAGCGTTAATGTCTGGCTTCTGATAAAGCGGGCCATGTTAAGGGCGGTTTTTTCCTGTTTGGTCACTGATGCCTCCGTGTAAGGGGGATTTCTGTTCATGGGGGTAATGATACCGATGAAACGAGAGAGGATGCTCACGATACGGGTTACTGATGATGAACATGCCCGGTTACTGGAACGTTGTGAGGGTAAACAACTGGCGGTATGGATGCGGCGGGACCAGAGAAAAATCACTCAGGGTCAATGCCAGCGCTTCGTTAATACAGATGTAGGTGTTCCACAGGGTAGCCAGCAGCATCCTGCGATGCAGATCCGGAACATAATGGTGCAGGGCGCTGACTTCCGCGTTTCCAGACTTTACGAAACACGGAAACCGAAGACCATTCATGTTGTTGCTCAGGTCGCAGACGTTTTGCAGCAGCAGTCGCTTCACGTTCGCTCGCGTATCGGTGATTCATTCTGCTAACCAGTAAGGCAACCCCGCCAGCCTAGCCGGGTCCTCAACGACAGGAGCACGATCATGCTAGTCATGCCCCGCGCCCACCGGAAGGAGCTGACTGGGTTGAAGGCTCTCAAGGGCATCGGTCGAGATCCCGGTGCCTAATGAGTGAGCTAACTTACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCGTGCCAGCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCCAGGGTGGTTTTTCTTTTCACCAGTGAGACGGGCAACAGCTGATTGCCCTTCACCGCCTGGCCCTGAGAGAGTTGCAGCAAGCGGTCCACGCTGGTTTGCCCCAGCAGGCGAAAATCCTGTTTGATGGTGGTTAACGGCGGGATATAACATGAGCTGTCTTCGGTATCGTCGTATCCCACTACCGAGATGTCCGCACCAACGCGCAGCCCGGACTCGGTAATGGCGCGCATTGCGCCCAGCGCCATCTGATCGTTGGCAACCAGCATCGCAGTGGGAACGATGCCCTCATTCAGCATTTGCATGGTTTGTTGAAAACCGGACATGGCACTCCAGTCGCCTTCCCGTTCCGCTATCGGCTGAATTTGATTGCGAGTGAGATATTTATGCCAGCCAGCCAGACGCAGACGCGCCGAGACAGAACTTAATGGGCCCGCTAACAGCGCGATTTGCTGGTGACCCAATGCGACCAGATGCTCCACGCCCAGTCGCGTACCGTCTTCATGGGAGAAAATAATACTGTTGATGGGTGTCTGGTCAGAGACATCAAGAAATAACGCCGGAACATTAGTGCAGGCAGCTTCCACAGCAATGGCATCCTGGTCATCCAGCGGATAGTTAATGATCAGCCCACTGACGCGTTGCGCGAGAAGATTGTGCACCGCCGCTTTACAGGCTTCGACGCCGCTTCGTTCTACCATCGACACCACCACGCTGGCACCCAGTTGATCGGCGCGAGATTTAATCGCCGCGACAATTTGCGACGGCGCGTGCAGGGCCAGACTGGAGGTGGCAACGCCAATCAGCAACGACTGTTTGCCCGCCAGTTGTTGTGCCACGCGGTTGGGAATGTAATTCAGCTCCGCCATCGCCGCTTCCACTTTTTCCCGCGTTTTCGCAGAAACGTGGCTGGCCTGGTTCACCACGCGGGAAACGGTCTGATAAGAGACACCGGCATACTCTGCGACATCGTATAACGTTACTGGTTTCACATTCACCACCCTGAATTGACTCTCTTCCGGGCGCTATCATGCCATACCGCGAAAGGTTTTGCGCCATTCGATGGTGTCCGGGATCTCGACGCTCTCCCTTATGCGACTCCTGCATTAGGAAGCAGCCCAGTAGTAGGTTGAGGCCGTTGAGCACCGCCGCCGCAAGGAATGGTGCATGCAAGGAGATGGCGCCCAACAGTCCCCCGGCCACGGGGCCTGCCACCATACCCACGCCGAAACAAGCGCTCATGAGCCCGAAGTGGCGAGCCCGATCTTCCCCATCGGTGATGTCGGCGATATAGGCGCCAGCAACCGCACCTGTGGCGCCGGTGATGCCGGCCACGATGCGTCCGGCGTAGCCTAGGATCGAGATCGATCTCGATCCCGCGAAATTAATACGACTCACTACGSEQ. ID NO: 11ATGATGTCTGAAAATTTGCAATTGTCAGCTGAAGAAATGAGACAATTGGGTTACCAAGCAGTTGATTTGATCATCGATCACATGAACCATTTGAAGTCTAAGCCAGTTTCAGAAACAATCGATTCTGATATCTTGAGAAATAAGTTGACTGAATCTATCCCAGAAAATGGTTCAGATCCAAAGGAATTGTTGCATTTCTTGAACAGAAACGTTTTTAATCAAATTACACATGTTGATCATCCACATTTCTTGGCTTTTGTTCCAGGTCCAAATAATTACGTTGGTGTTGTTGCAGATTTCTTGGCTTCTGGTTTTAATGTTTTTCCAACTGCATGGATTGCTGGTGCAGGTGCTGAACAAATCGAATTGACTACAATTAATTGGTTGAAATCTATGTTGGGTTTTCCAGATTCAGCTGAAGGTTTATTTGTTTCTGGTGGTTCAATGGCAAATTTGACAGCTTTGACTGTTGCAAGACAGGCTAAGTTGAACAACGATATCGAAAATGCTGTTGTTTACTTCTCTGATCAAACACATTTCTCAGTTGATAGAGCATTGAAGGTTTTAGGTTTTAAACATCATCAAATCTGTAGAATCGAAACAGATGAACATTTGAGAATCTCTGTTTCAGCTTTGAAGAAACAAATTAAAGAAGATAGAACTAAGGGTAAAAAGCCATTCTGTGTTATTGCAAATGCTGGTACTACAAATTGTGGTGCTGTTGATTCTTTGAACGAATTAGCAGATTTGTGTAACGATGAAGATGTTTGGTTGCATGCTGATGGTTCTTATGGTGCTCCAGCTATCTTGTCTGAAAAGGGTTCAGCTATGTTGCAAGGTATTCATAGAGCAGATTCTTTGACTTTAGATCCACATAAGTGGTTGTTCCAACCATACGATGTTGGTTGTGTTTTGATCAGAAACTCTCAATATTTGTCAAAGACTTTTAGAATGATGCCAGAATACATCAAGGATTCAGAAACTAACGTTGAAGGTGAAATTAATTTCGGTGAATGTGGTATCGAATTGTCAAGAAGATTCAGAGCTTTGAAGGTTTGGTTGTCTTTTAAAGTTTTCGGTGTTGCTGCTTTTAGACAAGCAATCGATCATGGTATCATGTTAGCAGAACAAGTTGAAGCATTTTTGGGTAAAGCAAAAGATTGGGAAGTTGTTACACCAGCTCAATTGGGTATCGTTACTTTTAGATACATTCCATCTGAATTGGCATCAACAGATACTATTAATGAAATTAATAAGAAATTGGTTAAGGAAATCACACATAGAGGTTTCGCTATGTTATCTACTACAGAATTGAAGGAAAAGGTTGTTATTAGATTGTGTTCAATTAATCCAAGAACTACAACTGAAGAAATGTTGCAAATCATGATGAAGATTAAAGCATTGGCTGAAGAAGTTTCTATTTCATACCCATGTGTTGCTGAATAASEQ. ID NO: 12MMSENLQLSAEEMRQLGYQAVDLIIDHMNHLKSKPVSETIDSDILRNKLTESIPENGSDPKELLHFLNRNVFNQITHVDHPHFLAFVPGPNNYVGVVADFLASGFNVFPTAWIAGAGAEQIELTTINWLKSMLGFPDSAEGLFVSGGSMANLTALTVARQAKLNNDIENAVVYFSDQTHFSVDRALKVLGFKHHQICRIETDEHLRISVSALKKQIKEDRTKGKKPFCVIANAGTTNCGAVDSLNELADLCNDEDVWLHADGSYGAPAILSEKGSAMLQGIHRADSLTLDPHKWLFQPYDVGCVLIRNSQYLSKTFRMMPEYIKDSETNVEGEINFGECGIELSRRFRALKVWLSFKVFGVAAFRQAIDHGIMLAEQVEAFLGKAKDWEVVTPAQLGIVTFRYIPSELASTDTINEINKKLVKEITHRGFAMLSTTELKEKVVIRLCSINPRTTTEEMLQIMMKIKALAEEVSISYPCVAESEQ. ID NO: 13ATGTTTGGTGTACAAGACACCCCGCAACATATATGCTACGAGCCTCAGCAGCGTAAGGTCAGTGAGAGAACATCACGTAACAGATCTCGTTCTAAATCACTGGACCCGGACAGCTTGCGCGAGAAAGGAAAGAAGACGCAACACCGTGAGGCGGATTGTTTCTTCGGTGAAGACAACCGGATGGAAAACTCCTACTCTGCGCAAATGTACATTGACGAGTTCGACCCTGTACACTATTACCAAACCTATTATTCCTCAGGGAAGGGCGGCATTGCTCGTGAGTGGACAGATTTTGCTTTGCAAAACTTGCATGAAACGTTCGGGCCTGGCGGGGTTAAAGGTGACATTCTTATTGACTTCGGTGCTGGGCCGACAATATATCAGCTTCTGAGCGCATGTGAGGTTTTCAATAGCATTATTACATCCGACTTTCTTGAGCAAAACCGCGAGCAACTTGAGAAATGGCTTCGAAAGGACCCGGACGCCCTTGACTGGTCCCATTTCACGAAGTACGTTTGCGAGCTCGAAGGCAACCGGGACAACTGGGAAAAGAAAGAGGAAACCCTGCGCCGAAAGGTTACCAAGGTGCTTAAATGTGACGCACTGGCCGAGAAGCCTTTCGACGACGTGCCAATGCCAGAGGCTGACTGTCTGATCTCATGCCTGTGTTTAGAGAACCCTTGTCAAGACCAGGAAGCTTACATTAACATATTGAAGAAGTTAAAAGAGCTCTTGAAACCGGGCGGCCACATCATTATACAGTCCATATTGAACTGCTCGTATTACCATATTGGCAATAGCTGCTTCTCACATTTGTCGTTAAGCAAGGACGACGTGGAGAAATCGTTTAAGGAAGCTGGCTACGAAATCGTCAAATTGAAGGTTCTTCCACGCTCAGTTATGTCGGAAATGGAAATCAGCGACTCAAATGGCTACTACTTCATCCACGCTCGGAAACCGCAAAAGGAGTAASEQ. ID NO: 14MFGVQDTPQHICYEPQQRKVSERTSRNRSRSKSLDPDSLREKGKKTQHREADCFFGEDNRMENSYSAQMYIDEFDPVHYYQTYYSSGKGGIAREWTDFALQNLHETFGPGGVKGDILIDFGAGPTIYQLLSACEVFNSIITSDFLEQNREQLEKWLRKDPDALDWSHFTKYVCELEGNRDNWEKKEETLRRKVTKVLKCDALAEKPFDDVPMPEADCLISCLCLENPCQDQEAYINILKKLKELLKPGGHIIIQSILNCSYYHIGNSCFSHLSLSKDDVEKSFKEAGYEIVKLKVLPRSVMSEMEISDSNGYYFIHARKPQKE EXAMPLES Example 1—Chemical Synthesis of a First, a Second and a Third Aminated Psilocybin Derivative Referring toFIG.9D, shown therein is a 7-step synthesis for two 4-O-methyl-psilocybin derivatives respectively aminated at C5and C7from 4-methoxyindole. The first step involved the regioselective 3-nitrovinylation of 4-methoxyindole (9D-1). Under argon, 4-methoxy indole (9D-1) (4000 mg, 27.18 mmol, 1.00 eq) and 1-(dimethylamino)-2-nitroethylene (3472 mg, 29.90 mmol, 1.10 eq) were dissolved in trifluoroacetic acid (20.8 mL, 272 mmol, 10.0 eq) and allowed to stir at room temperature for 3 hours until complete as determined by TLC (1:1 ethyl acetate-hexanes). The dark red solution was diluted with ethyl acetate (100 mL) and carefully poured over saturated sodium bicarbonate solution (200 mL). This biphasic mixture was then separated, and the aqueous phase extracted with ethyl acetate (4×100 mL). The combined organic extracts were washed with brine, dried with MgSO4, and concentrated under reduced pressure to yield the crude product 9D-2 (5600 mg) that was used directly without any further purification. The second step involved the conjugated reduction of the alkene functionality of compound 9D-2. To a solution of crude compound 9D-2 (5600 mg) in ethanol (50 mL) and THE (50 mL) under ambient air was added sodium borohydride (5141 mg, 136 mmol) in small portions, waiting for effervescence to decrease between additions. This mixture was allowed to stir at room temperature for 18 hours at which point reaction was complete as determined by TLC (1:1 ethyl acetate-hexanes). The reaction was quenched by pouring over ice-water (200 ml) and extracted with DCM (4×100 mL). The combined organic extracts were dried with MgSO4and concentrated under reduced pressure to yield the crude product as a brown solid. Purification by column chromatography on silica gel using a 10% to 50% ethyl acetate-hexanes gradient to yield compound 9D-3 as a white solid (1950 mg, 8.94 mg, 33% over 2 steps).1H NMR (400 MHz, CDCl3): δ (ppm)=8.02 (s, 1H), 7.15 (t, J=8.0 Hz, 1H), 7.00 (dd, J=8.3, 0.7 Hz, 1H), 6.94 (dd, J=2.5, 1.0 Hz, 1H), 6.55 (dd, J=7.8, 0.7 Hz, 1H), 4.76 (t, J=7.2 Hz, 2H), 3.97 (s, 3H), 3.57 (td, J=7.2, 0.8 Hz, 2H). The third step involved the reduction of the nitro functionality of compound 3. Under argon in a flame-dried flask compound 9D-3 (800 mg, 3.63 mmol, 1.00 eq) was dissolved in anhydrous THE (20 mL) and cooled to 0° C. To this solution was added 1 M lithium aluminum hydride in THE (18.2 mL, 18.2 mmol, 5.00 eq), causing a colour change to yellow. The reaction mixture was heated to reflux for 2.5 hours, yielding a milky-white solution. After cooling to 0° C., the reaction was quenched with 10% water-THE (15 mL) and allowed to stir for 10 minutes. The precipitated white solids were filtered off and the filter-cake rinsed with THE (10 mL), dichloromethane (10 mL) and methanol (10 mL). The organic filtrate was dried with MgSO4and concentrated under reduced pressure to yield compound 9D-4 as an off-white solid (395 mg, 2.08 mmol, 57%).1H NMR (400 MHz, CDCl3): δ (ppm)=8.12 (s, 1H), 7.09 (t, J=7.9 Hz, 1H), 6.97 (d, J=8.1 Hz, 1H), 6.89 (d, J=2.2 Hz, 1H), 6.50 (d, J=7.8 Hz, 1H), 3.92 (s, 3H), 3.02 (s, 4H). The fourth step involved the full protection of the side-chain amino group and the N1 of compound 9D-4. Under argon in a flame-dried flask, compound 9D-4 (245 mg, 1.29 mmol, 1.00 eq) was dissolved in anhydrous acetonitrile (10 mL). To this solution, di-tert-butyl dicarbonate (2811 mg, 12.9 mmol, 10.0 eq) and 4-dimethylaminopyridine (157 mg, 1.29 mmol, 1.00 eq) was added, and the reaction mixture allowed to stir at room temperature for 20 hours. Water (20 mL) was added, and the mixture was extracted with dichloromethane (4×30 mL). The combined organic extracts were washed with brine (25 mL), dried with MgSO4, and concentrated under vacuum to yield the crude product as a dark red oil. Purification by column chromatography on silica gel using an 8% to 15% ethyl acetate-hexanes gradient yielded compound 9D-5 as an oily white solid (325 mg, 0.663 mmol, 51%).1H NMR (400 MHz, CDCl3): δ (ppm)=7.74 (d, J=8.4 Hz, 1H), 7.23-7.13 (m, 2H), 6.63 (d, J=7.9 Hz, 1H), 3.91 (m, 5H), 3.08 (ddd, J=7.5, 6.3, 1.0 Hz, 2H), 1.63 (s, 9H), 1.38 (s, 18H). The fifth step involved the regioselective nitration of compound 9D-5. A flame-dried round-bottom flask was charged with compound 9D-5 (325 mg, 0.662 mmol, 1.00 eq), silver nitrate (124 mg, 0.729 mmol, 1.10 eq), and dry acetonitrile (2.5 mL), then cooled to 0° C. under argon atmosphere. Benzoyl chloride (102 mg, 0.729 mmol, 1.10 eq) was diluted with dry acetonitrile (0.5 mL) and added dropwise to the reaction mixture, which was then allowed to stir at 0° C. for 3 hours. The reaction mixture was diluted with ethyl acetate (10 mL) and the precipitated salts were removed via vacuum filtration and washed with ethyl acetate (5 mL). The organic filtrate was washed with water (3×20 mL) and saturated Na2CO3(20 mL), then dried with MgSO4and solvent removed in vacuo. The crude mixture was purified by column chromatography on silica gel using a gradient of 5 to 15% ethyl acetate-hexanes to afford compounds 9D-6a (25 mg, 0.047 mmol, 7%), 9D-6b (48 mg, 0.090 mmol, 14%), and 9D-6c (45 mg, 0.078 mmol, 12%) in order of elution as yellow solids. Compound 9D-6a:1H NMR (400 MHz, CDCl3): δ (ppm)=7.62 (dd, J=8.5, 0.7 Hz, 1H), 7.41 (t, J=8.3 Hz, 1H), 7.41 (s, 1H), 6.70 (dd, J=8.1, 0.7 Hz, 1H), 4.07-4.02 (m, 2H), 3.94 (s, 3H), 3.37-3.31 (m, 2H), 1.55 (s, 9H), 1.33 (s, 18H). Compound 9D-9D-6b:1H NMR (400 MHz, CDCl3): δ (ppm)=8.00-7.94 (m, 1H), 7.87 (d, J=9.0 Hz, 1H), 7.38 (s, 1H), 4.01 (s, 3H), 3.94 (t, J=7.6 Hz, 2H), 3.10 (t, J=7.6 Hz, 2H), 1.65 (s, 9H), 1.41 (s, 18H). Compound 9D-6c:1H NMR (400 MHz, CDCl3): δ (ppm)=7.82 (d, J=8.8 Hz, 1H), 6.64 (d, J=8.8 Hz, 1H), 4.02 (s, 3H), 3.91 (dd, J=7.4, 6.4 Hz, 2H), 3.08 (td, J=6.8, 6.4, 1.0 Hz, 2H), 1.57 (s, 9H), 1.40 (s, 18H). The sixth step involved the reduction of the nitro group. To a vigorously stirring solution of compound 9D-6b (27 mg, 0.050 mmol, 1.0 eq) in methanol (2 mL) was added 10% palladium on activated charcoal (23 mg, 0.021 mmol, 0.50 eq) followed by ammonium formate (46 mg, 0.70 mmol, 17 eq). The reaction mixture was allowed to stir at room temperature for 2 hours until complete as determined by TLC (1:4 ethyl acetate-hexanes). The catalyst was removed and methanol was removed under vacuum. The residue was taken up in dichloromethane (10 mL), washed with brine (10 mL), the organic phase dried with MgSO4and concentrated under reduced pressure to yield compound 9D-7 as a colourless film (22 mg, 0.044 mmol, 88%).1H NMR (400 MHz, CDCl3): δ (ppm)=7.72 (s, 1H), 7.26-7.18 (m, 1H), 6.76 (d, J=8.7 Hz, 1H), 3.96-3.91 (m, 2H), 3.84 (s, 3H), 3.09-3.00 (m, 2H), 1.62 (s, 9H), 1.43 (s, 18H). Similarly, the nitro group of compound 9D-6c can also be reduced in an analogous manner. To a vigorously stirring solution of compound 9D-6c (22 mg, 0.041 mmol, 1.0 eq) in methanol (2 mL) was added 10% palladium on activated charcoal (22 mg, 0.021 mmol, 0.50 eq) followed by ammonium formate (44 mg, 0.70 mmol, 17 eq). The reaction mixture was allowed to stir at room temperature for 2 hours and monitored by TLC (1:4 ethyl acetate-hexanes). The catalyst was removed and methanol was removed under vacuum. The residue was taken up in dichloromethane (10 mL), washed with brine (10 mL), the organic phase dried with MgSO4and concentrated under reduced pressure to yield compound 9D-9 as a brown film (14 mg, 0.028 mmol, 67%).1H NMR (600 MHz, CDCl3): δ (ppm)=7.13 (s, 1H), 6.69 (d, J=8.3 Hz, 1H), 6.57 (dt, J=8.5, 1.0 Hz, 1H), 3.93-3.87 (m, 2H), 3.85 (s, 3H), 3.04 (t, J=6.9 Hz, 2H), 1.61 (s, 9H), 1.40 (s, 18H). The seventh step involved the removal of all protecting group. To a solution of 9D-7 (22 mg, 0.044 mmol, 1.0 eq) in dichloromethane (1.0 mL) and methanol (0.1 mL) was added trifluoroacetic acid (0.15 mL, 1.5 mmol, 35 eq) dropwise. The reaction mixture was heated to 40° C. for 2 hours, then allowed to stir at room temperature for 18 hours. As the reaction was incomplete, the reaction was heated for a further 20 hours at 40° C. The reaction mixture was concentrated under reduced pressure to yield compound 9D-8 (5 mg, 0.2 mmol, 56%).1H NMR (600 MHz, D2O): δ (ppm)=7.24 (d, J=8.5 Hz, 1H), 7.22 (s, 1H), 7.03 (d, J=8.5 Hz, 1H), 3.81 (s, 3H), 3.23 (t, J=7.0 Hz, 2H), 3.10 (t, J=7.0 Hz, 2H). It is noted that compound 9D-8 corresponds with an example compound having chemical formula (IX): set forth herein. Similarly, compound 9D-9 can also be deprotected in an analogous manner. To a solution of 9D-9 (14 mg, 0.028 mmol, 1.0 eq) in dichloromethane (1.0 mL) and methanol (0.1 mL) was added trifluoroacetic acid (0.064 mL, 0.83 mmol, 30 eq). The reaction mixture was heated to 40° C. for 20 hours. The reaction mixture was concentrated under reduced pressure to yield compound 9D-10 (4 mg, 0.2 mmol, 70%).1H NMR (600 MHz, D2O): δ (ppm)=7.25 (s, 1H), 7.18 (d, J=8.3 Hz, 1H), 6.66 (d, J=8.3 Hz, 1H), 3.96 (s, 3H), 3.30 (t, J=6.8 Hz, 2H), 3.21 (t, J=6.9 Hz, 2H). It is noted that compound 9D-10 corresponds with an example compound having chemical formula (XII): set forth herein. Assessment of Cell Viability Upon Treatment of Aminated Psilocybin Derivatives To establish suitable ligand concentrations for competitive binding assays, PrestoBlue assays were first performed. The PrestoBlue assay measures cell metabolic activity based on tetrazolium salt formation, and is a preferred method for routine cell viability assays (Terrasso et al., 2017, J Pharmacol Toxicol Methods 83: 72). Results of these assays were conducted using both control ligands (e.g., psilocybin, psilocin, DMT) and novel derivatives, in part as a pre-screen for any remarkable toxic effects on cell cultures up to concentrations of 1 mM. A known cellular toxin (Triton X-100, Pyrgiotakis G. et al., 2009, Ann. Biomed. Eng. 37: 1464-1473) was included as a general marker of toxicity. Drug-induced changes in cell health within simple in vitro systems such as the HepG2 cell line are commonly adopted as first-line screening approaches in the pharmaceutical industry (Weaver et al, 2017, Expert Opin Drug Metab Toxicol 13: 767). HepG2 is a human hepatoma that is most commonly used in drug metabolism and hepatotoxicity studies (Donato et al., 2015, Methods Mol Biol 1250: 77). Herein, HepG2 cells were cultured using standard procedures using the manufacture's protocols (ATCC, HB-8065). Briefly, cells were cultured in Eagle's minimum essential medium supplemented with 10% fetal bovine serum and grown at 37° C. in the presence of 5% CO2. To test the various compounds with the cell line, cells were seeded in a clear 96-well culture plate at 20,000 cells per well. After allowing cells to attach and grow for 24 hours, compounds were added at 1 μM, 10 μM, 100 μM, and 1 mM. Methanol was used as vehicle, at concentrations 0.001, 0.01, 0.1, and 1%. As a positive control for toxicity, TritonX concentrations used were 0.0001, 0.001, 0.01 and 0.1%. Cells were incubated with compounds for 48 hours before accessing cell viability with the PrestoBlue assay following the manufacture's protocol (ThermoFisher Scientific, P50200). PrestoBlue reagent was added to cells and allowed to incubate for 1 hour before reading. Absorbance readings were performed at 570 nm with the reference at 600 nm on a SpectraMax iD3 plate reader. Non-treated cells were assigned 100% viability. Bar graphs show the mean +/−SD, n=3. Significance was determined by 2-way ANOVA followed by Dunnett's multiple comparison test and is indicated by *** (P<0.0001), **(P<0.001), *(P<0.005). Data acquired for the derivative having chemical formula (IX) is displayed as “IX” on the x-axis ofFIG.11A.). Data acquired for the derivative having chemical formula (XII) is displayed as “XII” on the x-axis ofFIG.11B. Radioligand Receptor Binding Assays. Evaluation of drug binding is an essential step to characterization of all drug-target interactions (Fang 2012, Exp Opin Drug Discov 7:969). The binding affinity of a drug to a target is traditionally viewed as an acceptable surrogate of its in vivo efficacy (NGAez et al., 2012, Drug Disc Today 17: 10). Competition assays, also called displacement or modulation binding assays, are a common approach to measure activity of a ligand at a target receptor (Flanagan 2016, Methods Cell Biol 132: 191). In these assays, standard radioligands acting either as agonists or antagonists are ascribed to specific receptors. In the case of G protein-coupled receptor 5-HT2A, [3H]ketanserin is a well-established antagonist used routinely in competition assays to evaluate competitive activity of novel drug candidates at the 5-HT2Areceptor (Maguire et al., 2012, Methods Mol Biol 897: 31). Thus, to evaluate activity of novel psilocybin derivatives at the 5-HT2Areceptor, competition assays using [3H]ketanserin were employed as follows. SPA beads (RPNQ0010), [3H] ketanserin (NET1233025UC), membranes containing 5-HT2A(ES-313-M400UA), and isoplate-96 microplate (6005040) were all purchased from PerkinElmer. Radioactive binding assays were carried out using Scintillation Proximity Assay (SPA). For saturation binding assays, mixtures of 10 ug of membrane containing 5-HT2Areceptor was pre-coupled to 1 mg of SPA beads at room temperature in a tube rotator for 1 hour in binding buffer (50 mM Tris-HCl pH7.4, 4 mM CaCl2, 1 mM ascorbic acid, 10 μM pargyline HCl). After pre-coupling, the beads and membrane were aliquoted in an isoplate-96 microplate with increasing amounts of [3H]ketanserin (0.1525 nM to 5 nM) and incubated for two hours at room temperature in the dark with shaking. After incubation, the samples were read on a MicroBeta 2 Microplate Counter (Perkin Elmer). Determination of non-specific binding was carried out in the presence of 20 μM of spiperone (S7395-250MG, Sigma). Equilibrium binding constants for ketanserin (Kd) were determined from saturation binding curves using the ‘one-site saturation binding analysis’ method of GraphPad PRISM software (Version 9.2.0). Competition binding assays were performed using fixed (1 nM) [3H]ketanserin and different concentrations of tryptophan (3 nM to 1 mM), psilocin (30 pM to 10 μM) or unlabeled test compound (3 nM to 1 mM) similar to the saturation binding assay. Kivalues were calculated from the competition displacement data using the competitive binding analysis from GraphPad PRISM software. Tryptophan was included as a negative control as it has no activity at the 5-HT2Areceptor. In contrast, psilocin was used as a positive control since it has established binding activity at the 5-HT2Areceptor (Kim et al., 2020, Cell 182: 1574).FIG.11Ddepicts the saturation binding curves for [3H]ketanserin at the 5-HT2Areceptor. Panel 1 shows the specific saturation ligand binding of [3H]ketanserin (from 0.1525 nM to 5 nM) to membranes containing 5-HT2Areceptor, which was obtained after subtracting non-specific binding values (shown in Panel 2). Specific binding in counts per minute (cpm) was calculated by subtracting non-specific binding from total binding. Specific binding (pmol/mg) was calculated from pmol of [3H]ketanserin bound per mg of protein in the assay. The Kdwas calculated by fitting the data with the one-site binding model of PRISM software (version 9.2.0).FIG.11E(top panel) shows the competition binding curve for psilocin as a positive control (binding).FIG.11E(bottom panel) shows the competition binding curve for tryptophan as a negative control (no binding).FIG.11Fshows competition binding curve for compound with formula (IX), designated “IX” in the figure.FIG.11Gshows competition binding curve for compound with formula (XII), designated “XII” in the figure. Cell Lines and Control Ligands Used to Assess Activity at 5-HT1A. CHO-K1/Gα15(GenScript, M00257) (−5-HT1A) and CHO-K1/5-HT1A/Gα15(GenScript, M00330) (+5-HT1A) cells lines were used. Briefly, CHO-K1/Gα15is a control cell line that constitutively expresses Gα15which is a promiscuous Gqprotein. This control cell line lacks any transgene encoding 5-HT1Areceptors, but still responds to forskolin; thus, cAMP response to forskolin should be the same regardless of whether or not 5-HT1Aagonists are present. Conversely, CHO-K1/5-HT1A/Gα15cells stably express 5-HT1Areceptor in the CHO-K1 host background. Notably, Gα15is a promiscuous G protein known to induce calcium flux response, present in both control and 5-HT1Acell lines. In +5-HT1Acells, Gα15may be recruited in place of Gαi/o, which could theoretically dampen cAMP response (Rojas and Fiedler 2016, Front Cell Neurosci 10: 272). Thus, we included two known 5-HT1Aagonists, DMT (Cameron and Olson 2018, ACS Chem Neurosci 9: 2344) and serotonin (Rojas and Fiedler 2016, Front Cell Neurosci 10: 272) as positive controls to ensure sufficient cAMP response was observed, thereby indicating measurable recruitment of Gαi/oprotein to activated 5-HT1Areceptors. In contrast, tryptophan is not known to activate, or modulate in any way, 5-HT1Areceptors, and was thus used as a negative control. Cells were maintained in complete growth media as recommended by supplier (GenScript) which is constituted as follows: Ham's F12 Nutrient mix (HAM's F12, GIBCO #11765-047) with 10% fetal bovine serum (FBS) (Thermo Scientific #12483020), 200 μg/ml zeocin (Thermo Scientific #R25005) and/or 100 μg/ml hygromycin (Thermo Scientific #10687010). The cells were cultured in a humidified incubator with 37° C. and 5% CO2. Cells maintenance was carried out as recommended by the cell supplier. Briefly, vials with cells were removed from the liquid nitrogen and thawed quickly in 37° C. water bath. Just before the cells were completely thawed the vial's outside was decontaminated by 70% ethanol spray. The cell suspension was then retrieved from the vial and added to warm (37° C.) complete growth media, and centrifuged at 1,000 rpm for 5 minutes. The supernatant was discarded, and the cell pellet was then resuspended in another 10 ml of complete growth media, and added to the 10 cm cell culture dish (Greiner Bio-One #664160). The media was changed every third day until the cells were about 90% confluent. The ˜90% confluent cells were then split 10:1 for maintenance or used for experiment. Evaluation of 5-HT1AReceptor Modulation As 5-HT1Aactivation inhibits cAMP formation, the ability of test molecules to modulate 5-HT1Aresponse was measured via changes in the levels of cAMP produced due to application of 4 μM forskolin. The change in intracellular cAMP levels due to the treatment of novel molecules was measured using cAMP-Glo Assay kit (Promega #V1501). Briefly, +5-HT1Acells were seeded on 1-6 columns and base −5-HT1Acells were seeded on columns 7-12 of the white walled clear bottom 96-well plate (Corning, #3903). Both cells were seeded at the density of 30,000 cells/well in 100 μl complete growth media and cultured 24 hrs in humidified incubator at 37° C. and 5% CO2. On the experiment day, the media of cells was replaced with serum/antibiotic free culture media. Then the cells were treated for 20 minutes with test molecules dissolved in induction medium (serum/antibiotic free culture media containing 4 μM forskolin, 500 μM IBMX (isobutyl-1-methylxanthine, Sigma-Aldrich, Cat. #17018) and 100 μM (RO 20-1724, Sigma-Aldrich, Cat. #B8279)). Forskolin induced cAMP formation whereas IBMX and RO 20-1724 inhibited the degradation of cAMP. PKA was added to the lysate, mixed, and subsequently the substrate of the PKA was added. PKA was activated by cAMP, and the amount of ATP consumed due to PKA phosphorylation directly corresponded to cAMP levels in the lysate. Reduced ATP caused reduced conversion of luciferin to oxyluciferin, conferring diminished luminescence as the result of 5-HT1Aactivation. Conversely, enhanced luminescence was expected in cases where 5-HT1Areceptor modulation—imparted by a test molecule—caused downstream increases in ATP, thus imparting enhanced conversion of luciferin to oxyluciferin.FIG.11Ishows increased luminescence resulting from decreased dosages of forskolin (and decreased cAMP) in +5HT1Acell culture.FIG.11Jillustrates reduced luminescence (i.e., increased cAMP) in the presence of fixed (4 μM) forskolin as dosages of DMT decrease, revealing 5-HT1Aactivity of DMT.FIG.11Killustrates no trend in luminescence (i.e., no trend in cAMP levels) in the presence of fixed (4 μM) forskolin, as dosages of tryptophan decrease, revealing a lack of 5-HT1Amodulation for tryptophan.FIG.11Lillustrates increased % cAMP levels in the presence of fixed (4 μM) forskolin as dosages of serotonin decrease, revealing 5-HT1Abinding activity of serotonin in +5HT1Acell cultures. Conversely, this trend of increasing % cAMP levels with decreasing serotonin is not observed in −5HT1Acell cultures. FIG.11Millustrates increased % cAMP levels in the presence of fixed (4 μM) forskolin as dosages of compound (IX) decrease, revealing 5-HT1Abinding activity of compound (IX) in +5HT1Acell cultures. Conversely, this trend of increasing % cAMP levels with decreasing compound (IX) is not observed in −5HT1Acell cultures. Note that compound (IX) is shown simply as (IX) along the x-axis. ForFIGS.11I-11M, error bars represent results of three experiments (n=3). Example 2—Biochemical Synthesis of a Fourth Aminated Psilocybin Derivative E. colistrain Ec-1 was constructed as follows. For plasmid cloning, Top10 or XL1-blue strains were used depending on antibiotic markers. Standard LB media was used for culturing. For gene expression and feeding experiments, the parent host strain employed was BL21 (DE3). The plasmid pETM6-H10-TmTrpB-2F3-V5-BaTDC-FLAG was created by first cloning the in-frame, C-terminally V5-tagged (SEQ. ID NO: 6, SEQ. ID NO: 7) TmTrpB-2F3 (SEQ. ID NO: 8, SEQ. ID NO: 9) into the NdeI/XhoI site of pETM6-H10 (SEQ. ID NO: 10) to create pETM6-H10-TmTrpB-2F3-V5. This intermediate plasmid was digested with SpeI and SalI, and in-frame, C-terminally FLAG tagged (SEQ. ID NO: 2, SEQ. ID NO: 3) BaTDC (SEQ. ID NO: 11, SEQ. ID NO: 12) was cloned into the site with XbaI and SalI, nullifying the SpeI restriction site. In this setup, the T7 polymerase was able to drive the expression of the polycistronic DNA containing both TmTrpB-2F3 and BaTDC. The target plasmid pETM6-H10-TmTrpB-2F3-V5-BaTDC-FLAG was transformed into BL21 (DE3) cells, and ampicillin was used to select for the correct clones containing the plasmid. Scaled-up culturing of engineeredE. coliwas conducted as follows: seed cultures were inoculated in AMM (Jones et al. 2015, Sci Rep. 5: 11301) medium overnight. The overnight culture was then divided into two flasks containing 500 mL each of AMM medium additionally containing 0.5% (w/v) serine, 1M IPTG, 50 ug/L ampicillin, and 100 mg/L aminated indole feedstock (5,7-dimethyl-1H-indol-4-ylamine; 1clickchemistry, www.1 clickchemistry.com) for conversion by Ec-1. Cultures were grown for 24 h. Cultures were then centrifuged (10,000 g×5 minutes) to remove cellular content, and culture broth containing secreted derivative was stored at −80° C. until further processing. Analysis and Purification, Analysis was carried out using high-resolution LC-HESI-LTQ-Orbitrap-XL MS (Thermo Fisher Scientific), employing a modified version of a method described previously (Chang et al., 2015, Plant Physiol. 169: 1127-1140), with the exception that liquid chromatography was carried out using an UltiMate 3000 HPLC (Thermo Fisher Scientific) equipped with a Poroshell 120 SB-C18 column (Agilent Technologies) instead of an Accela HPLC system (Thermo Fisher Scientific) equipped with a Zorbax C18 column (Agilent Technologies). Briefly, 100 microliters of culture media were dried and resuspended in 100 microliters of DMSO. One tenth (10 microliters) of this suspension was injected at a flow rate of 0.5 mL/min and a gradient of solvent A (water with 0.1% of formic acid) and solvent B (ACN with 0.1% formic acid) as follows: 100% to 0% (v/v) solvent A over 5 min; isocratic at 0% (v/v) for 1 min; 0% to 100% (v/v) over 0.1 min; and isocratic at 100% (v/v) for 1.9 min. Total run time was 8 minutes. Heated ESI source and interface conditions were operated in positive ion mode as follows: vaporizer temperature, 400° C.; source voltage, 3 kV; sheath gas, 60 au, auxiliary gas, 20 au; capillary temperature, 380° C.; capillary voltage, 6 V; tube lens, 45 V. Instrumentation was performed as a single, HR scan event using Orbitrap detection of m/z in the range of 100-500 m/z. Ion injection time was 300 ms with scan time of 1 s. External and internal calibration procedures ensured <2 ppm error to facilitate elemental formulae predictions. Singly protonated product with exact m/z and expected elemental formula matching the singly protonated form of 3-(2-aminoethyl)-5,7-dimethyl-1H-indol-4-amine, having chemical formula (VI): eluted at 2.9 minutes (EIC, see:FIG.12A). Although peak splitting can be minimized through the use of DMSO as injection solvent (Kaufman and Jegle 2005, Agilent Technologies Technical Bulletin 5989-2485EN), this phenomenon persisted owing to ion pairing effects between matrix components (Tarafder et al. 2010, J Chromatogr A 1217:7065-7073). As per standard procedures (Menéndez-Perdomo et al. 2021, Mass Spectrom 56: 34683) further analysis using high energy collisions (HCD) was achieved in a dedicated, post-LTQ, nitrogen collision cell. Orbitrap-based, HR fragment detection was employed (normalized collision energy, NCE 35), enabling opportunity to assign elemental formulae to subsequent diagnostic ion species characteristic of the targeted aminated psilocybin derivative with formula (VI) as follows (FIG.12B, Table 1) (Servillo L et al., 2013, J. Agric. Chem. 61: 5156-5162). TABLE I% RelativeIonicEmpiricalm/zabundancespeciesformula204.1131100[M + H]+C12H15N3187.122955[M + H − NH2]+C12H16N2202.492021186.110619189.09766.0200.09735.0162.10274.1150.68543.892.87853.861.09623.8 Example 3—Biochemical Synthesis of a Fifth Aminated Psilocybin Derivative Escherichia colistrain Ec-1 was used to biosynthesize aminated psilocybin derivative with formula (XIV) from aminated indole feedstock. The construction of Ec-1 is described in Example 2. Scaled-up culturing and material storage of engineeredE. coliwas conducted as described in Example 2, except that 6-methyl-1H-indol-4-ylamine (Combi-Blocks, www.combi-blocks.com) was used in place of 5,7-dimethyl-1H-indol-4-ylamine. To assess product, high-resolution LC-HESI-LTQ-Orbitrap-XL MS analysis was conducted as described in Example 2. Singly protonated product with exact m/z and expected elemental formula matching the singly protonated form of 3-(2-aminoethyl)-6-methyl-1H-indol-4-amine having chemical formula (XIV): eluted at 2.4 minutes (EIC, see:FIG.13A). As per standard procedures (Menéndez-Perdomo et al. 2021, Mass Spectrom 56: 34683) high energy collisions (HCD) were achieved in a dedicated, post-LTQ, nitrogen collision cell. Orbitrap-based, HR fragment detection was employed (normalized collision energy, NCE 35), enabling opportunity to assign elemental formulae to subsequent diagnostic ion species characteristic of a compound of formula (XIV), as follows (FIG.13B, Table II) (Servillo L. et al., 2013, J. Agric. Chem. 61: 5156-5162), TABLE II% RelativeIonicm/zabundancespeciesΔ ppm173.10707100[M + H − NH2]+1.44130.0648920144.0805716190.133732.7[M + H]+1.00161.107152.6118.064851.9202.350101.5160.075511.0 Example 4—Biochemical Synthesis of a Sixth Aminated Psilocybin Derivative Escherichia colistrain Ec-2 was used to biosynthesize aminated psilocybin derivative, where the amino group terminating the 2-carbon aliphatic side chain was conjugated to an acetyl group. Ec-2 was constructed using the same method as for Ec-1 (see: Example 2), except that an additional plasmid was assembled and transformed into cells along with pETM6-H10-TmTrpB-2F3-V5-BaTDC-FLAG. This additional plasmid encoded a promiscuous and efficientStreptomyces griseofuscusN-acetyltransferase enzyme named PsmF (SEQ. ID NO: 5). This additional plasmid was assembled as follows: from plasmid pCDM4 (SEQ. ID NO: 1), the plasmid pCDM4-PsmF-FLAG was created by inserting an in-frame, C-terminally FLAG-tagged (SEQ. ID NO: 2, SEQ. ID NO: 3) PsmF gene (SEQ. ID NO: 4, SEQ. ID NO: 5) into the NdeI/XhoI site of pCDM4. The two target plasmids pCDM4-PsmF-FLAG and pETM6-H10-TmTrpB-2F3-V5-BaTDC-FLAG were transformed into BL21 (DE3) cells, and antibiotics ampicillin plus streptomycin were used to select for the correct clones containing both plasmids. Scaled up culturing, analysis, purification, toxicology, and pharmacological testing were performed as described in Example 2, except that 1H-indol-7-ylamine (Combi-Blocks, www.combi-blocks.com) was used in place of 5,7-dimethyl-1H-indol-4-ylamine. Purification of the target product was achieved as follows: to 0.75 L ofE. coliculture, 10M NaOH solution was added until the pH reached ˜7. The culture was then extracted by ethyl acetate (4×500 ml). The organic layer was dried over Na2SO4, followed by concentration under reduced pressure. The residue was purified by flash chromatography on silica gel methanol-dichloromethane (2->4%) as eluent, to give the compound as light yellow solid (15 mg). NMR and HRMS data were as follows:1H NMR (400 MHz, CD3OD): δ=1.91 (s, 3H), 2.22 (s, 3H), 2.93 (t, J=6.9 Hz, 2H), 3.46 (t, J=7.2 Hz, 2H), 6.99 (t, J=7.8 Hz, 1H), 7.10 (m, 2H), 7.44 (dd, J=7.9, 1.0 Hz, 1H). Selective13C NMR (100 MHz, CD3OD): δ=21.2, 21.8, 24.7, 40.0, 112.5, 115.2, 115.8, 118.3, 122.1, 122.3, 129.4, 170.5, 171.8. HRMS (ESI) m/z: calcd. for C14H17N3O2[M+H]+260.1394, found 260.1392. Purity was assessed at 95%. This characterization confirmed a structure corresponding to compound (XIII): Assessment of Cell Viability Upon Treatment of Aldehyde Psilocybin Derivative Cell viability was assessed as described for Example 1, except the compound with formula (XIII) was evaluated in place of the compounds with formulae (IX) and (XII).FIG.11Cshows PrestoBlue assay results for compound with formula (XIII), depicted on the x-axis as “XIII”. Radioligand Receptor Binding Assays. Activity at 5-HT2Areceptor was assessed as described for Example 1, except the compound with formula (XIII) was evaluated in place of the compounds with formulae (IX) and (XII).FIG.11Hshows radioligand competition assay results for compound with formula (XIII), depicted on the x-axis simply as “XIII”. Example 5—Biochemical Synthesis of a Seventh Aminated Psilocybin Derivative Escherichia colistrain Ec-2 was used to biosynthesize aminated psilocybin derivative with formula (VII) from aminated indole feedstock. The construction of Ec-2 is described in Example 4. Scaled-up culturing and material storage of engineeredE. coliwas conducted as described in Example 2. To assess product, high-resolution LC-HESI-LTQ-Orbitrap-XL MS analysis was conducted as described in Example 2. Singly protonated product with exact m/z and expected elemental formula matching the singly protonated form of N-[2-(4-amino-5,7-dimethyl-1H-indol-3-yl)ethyl]acetamide having chemical formula (VII): eluted at 2.9 minutes (EIC, see:FIG.14A). Although peak splitting can be minimized through the use of DMSO as injection solvent (Kaufman and Jegle 2005, Agilent Technologies Technical Bulletin 5989-2485EN), this phenomenon persisted owing to ion pairing effects between matrix components (Tarafder et al 2010, J Chromatogr A 1217:7065-7073). As per standard procedures (Menéndez-Perdomo et al. 2021, Mass Spectrom 56: 34683) high energy collisions (HCD) were achieved in a dedicated, post-LTQ, nitrogen collision cell. Orbitrap-based, HR fragment detection was employed (normalized collision energy, NCE 35), enabling opportunity to assign elemental formulae to subsequent diagnostic ion species characteristic of a compound of formula (VII), as follows (FIG.14B, Table 111) (Servillo L. et al, 2013, J. Agric. Chen. 61: 5156-5162). TABLE III% RelativeIonicEmpiricalm/zAbundancespeciesformula187.1228100[M + H − N-acetyl]+C12H15N2246.160123[M + H]+C14H20N3O229.133516[M + H − NH2]+C14H17N2O187.12571.2203.22311.0217.13340.9187.11910.693.05290.666.12400.6 Example 6—Biochemical Synthesis of an Eighth Aminated Psilocybin Derivative Escherichia colistrain Ec-2 was used to biosynthesize aminated psilocybin derivative with formula (IV) from aminated indole feedstock. The construction of Ec-2 is described in Example 4. Scaled-up culturing and material storage of engineeredE. coliwas conducted as described in Example 2, except that 1H-indol-6-ylamine was used in place of 5,7-dimethyl-1H-indol-4-ylamine. To assess product, high-resolution LC-HESI-LTQ-Orbitrap-XL MS analysis was conducted as described in Example 2. Singly protonated product with exact m/z and expected elemental formula matching the singly protonated form of N-[2-(6-amino-1H-indol-3-yl)ethyl]acetamide having chemical formula (IV): eluted at 2.3 minutes (EIC, see:FIG.15A). As per standard procedures (Menéndez-Perdomo et al. 2021, Mass Spectrom 56: 34683) high energy collisions (HCD) were achieved in a dedicated, post-LTQ, nitrogen collision cell. Orbitrap-based, HR fragment detection was employed (normalized collision energy, NCE 35), enabling opportunity to assign elemental formulae to subsequent diagnostic ion species characteristic of a compound of formula (IV), as follows (FIG.15B, Table IV) (Servillo L. et al., 2013, J. Agric. Chem. 61: 5156-5162). TABLE IV% RelativeIonicEmpiricalm/zAbundanceSpeciesFormula159.0916100[M + H − NH2—C2H3O]+C10H11N2201.102212[M + H − NH2]+C12H13N2O218.12885.1[M + H]+C12H16N3O202.49311.2159.09461.1160.43830.5167.46640.383.35050.356.98680.3 Example 7—Biochemical Synthesis of a Ninth Aminated Psilocybin Derivative Escherichia colistrain Ec-2 was used to biosynthesize aminated psilocybin derivative with formula (III) from aminated indole feedstock. The construction of Ec-2 is described in Example 4. Scaled-up culturing and material storage of engineeredE. coliwas conducted as described in Example 2, except that 1H-indol-4-ylamine (Combi-Blocks; www.combi-blocks.com) was used in place of 5,7-dimethyl-1H-indol-4-ylamine. To assess product, high-resolution LC-HESI-LTQ-Orbitrap-XL MS analysis was conducted as described in Example 2. Singly protonated product with exact m/z and expected elemental formula matching the singly protonated form of N-[2-(4-amino-1H-indol-3-yl)ethyl]acetamide having chemical formula (III): eluted at 2.5 minutes (EIC, see:FIG.16A). Although peak splitting can be minimized through the use of DMSO as injection solvent (Kaufman and Jegle 2005, Agilent Technologies Technical Bulletin 5989-2485EN), this phenomenon persisted owing to ion pairing effects between matrix components (Tarafder et al. 2010, J Chromatogr A 1217:7065-7073). As per standard procedures (Menéndez-Perdomo et al. 2021, Mass Spectrom 56: 34683) high energy collisions (HCD) were achieved in a dedicated, post-LTQ, nitrogen collision cell. Orbitrap-based, HR fragment detection was employed (normalized collision energy, NCE 35), enabling opportunity to assign elemental formulae to subsequent diagnostic ion species characteristic of a compound of formula (III), as follows (FIG.16B, Table V) (Servillo L. et al., 2013, J. Agric. Chem. 61: 5156-5162). TABLE V% RelativeIonicEmpiricalm/zAbundanceSpeciesFormula159.0915100218.128856[M + H]+C12H16N3O201.102223C12H13N2O202.49012.1189.10221.0C11H13N2O176.11820.6C10H14N3200.10490.6122.03540.5183.09160.461.09650.4 Example 8—Biochemical Synthesis of a Tenth Aminated Psilocybin Derivative Escherichia colistrain Ec-2 was used to biosynthesize aminated psilocybin derivative with formula (XV) from aminated indole feedstock. The construction of Ec-2 is described in Example 4. Scaled-up culturing and material storage of engineeredE. coliwas conducted as described in Example 2, except that 6-methyl-1H-indol-4-ylamine (BLDPharm; www.bldpharm.com) was used in place of 5,7-dimethyl-1H-indol-4-ylamine. To assess product, high-resolution LC-HESI-LTQ-Orbitrap-XL MS analysis was conducted as described in Example 2. Singly protonated product with exact m/z and expected elemental formula matching the singly protonated form of N-[2-(4-amino-6-methyl-1H-indol-3-yl)ethyl]acetamide having chemical formula (XV): eluted at 2.9 minutes (EIC, see:17A). As per standard procedures (Menéndez-Perdomo et al. 2021, Mass Spectrom 56: 34683) high energy collisions (HCD) were achieved in a dedicated, post-LTQ, nitrogen collision cell. Orbitrap-based, HR fragment detection was employed (normalized collision energy, NCE 35), enabling opportunity to assign elemental formulae to subsequent diagnostic ion species characteristic of a compound of formula (XV), as follows (17B, Table VI) (Servillo L. et al., 2013, J. Agric. Chem. 61: 5156-5162). TABLE VI% RelativeIonicm/zabundancespeciesΔ ppm173.10712100[M + H − C2H5NO]+1.55232.1443664[M + H]+0.30215.1177214201.987031.3203.117721.166.124330.895.311060.878.589060.7 Example 9—Biochemical Synthesis of an Eleventh Aminated Psilocybin Derivative Escherichia colistrain Ec-2 was used to biosynthesize aminated psilocybin derivative with formula (XVI) from aminated indole feedstock. The construction of Ec-2 is described in Example 4. Scaled-up culturing and material storage of engineeredE. coliwas conducted as described in Example 2, except that 7-methyl-1H-indol-5-ylamine (BLDPharm; www.bldpharm.com) was used in place of 5,7-dimethyl-1H-indol-4-ylamine. To assess product, high-resolution LC-HESI-LTQ-Orbitrap-XL MS analysis was conducted as described in Example 2. Singly protonated product with exact m/z and expected elemental formula matching the singly protonated form of N-[2-(5-amino-7-methyl-1H-indol-3-yl)ethyl]acetamide having chemical formula (XVI): eluted at 2.3 minutes (EIC, see:FIG.18A). Although peak splitting can be minimized through the use of DMSO as injection solvent (Kaufman and Jegle 2005, Agilent Technologies Technical Bulletin 5989-2485EN), this phenomenon persisted owing to ion pairing effects between matrix components (Tarafder et al. 2010, J Chromatogr A 1217:7065-7073). As per standard procedures (Menéndez-Perdomo et al. 2021, Mass Spectrom 56: 34683) high energy collisions (HCD) were achieved in a dedicated, post-LTQ, nitrogen collision cell. Orbitrap-based, HR fragment detection was employed (normalized collision energy, NCE 35), enabling opportunity to assign elemental formulae to subsequent diagnostic ion species characteristic of a compound of formula (XVI), as follows (FIG.18B, Table VII) (Servillo L. et al., 2013, J. Agric. Chem. 61: 5156-5162). TABLE VII% RelativeIonicm/zAbundanceFragmentΔ ppm173.10714100.00[M + H − C2H5NO]+1.04232.1444911.17[M + H]+0.26215.117889.63117.998820.95201.972390.95214.133760.63 Example 10—Biochemical Synthesis of a Twelfth Aminated Psilocybin Derivative Escherichia colistrain Ec-3 was used to biosynthesize aminated psilocybin derivative, where the amino group terminating the 2-carbon aliphatic side chain was singly methylated. Ec-3 was constructed using the same method as for Ec-1 (see: Example 2), except that an additional plasmid was assembled and transformed into cells along with pETM6-H10-TmTrpB-2F3-V5-BaTDC-FLAG. This additional plasmid encoded a promiscuous and efficientRhinella marinaN-methyltransferase enzyme named RmNMT (SEQ. ID NO: 14). This additional plasmid was assembled as follows: from plasmid pCDM4 (SEQ. ID NO: 1), the plasmid pCDM4-RmNMT-FLAG was created by inserting an in-frame, C-terminally FLAG-tagged (SEQ. ID NO: 2, SEQ. ID NO: 3) RmNMT gene (SEQ. ID NO: 13) into the NdeI/XhoI site of pCDM4. The two target plasmids pCDM4-RmNMT-FLAG and pETM6-H10-TmTrpB-2F3-V5-BaTDC-FLAG were transformed into BL21 (DE3) cells, and antibiotics ampicillin plus streptomycin were used to select for the correct clones containing both plasmids. Scaled up culturing and analysis were performed as described in Example 2, except that 6-methyl-1H-indol-4-ylamine (BLDPharm, www.bldpharm.com) was used in place of 5,7-dimethyl-1H-indol-4-ylamine. To assess product, high-resolution LC-HESI-LTQ-Orbitrap-XL MS analysis was conducted as described in Example 2. Singly protonated product with exact m/z and expected elemental formula matching the singly protonated form of 6-methyl-3-[2-(methylamino)ethyl]-1H-indol-4-amine having chemical formula (XVII): eluted at 2.4 minutes (EIC, see:FIG.19). Although peak splitting and poor peak shape can be minimized through the use of DMSO as injection solvent (Kaufman and Jegle 2005, Agilent Technologies Technical Bulletin 5989-2485EN), this phenomenon persisted owing to ion pairing effects between matrix components (Tarafder et al. 2010, J Chromatogr A 1217:7065-7073).
195,780
11858896
DETAILED DESCRIPTION This disclosure relates DMT analogues or crystalline DMT analogues. According to the disclosure a DMT analogue or a crystalline DMT analogue includes crystalline N-ethyl-N-propyl-tryptamine (EPT) hydrofumarate, N-methyl-N-allyl-tryptamine (MALT) hydrofumarate, crystalline N-methyl-N-allyl-tryptamine (MALT) hydrofumarate, N—N-dibutyl-tryptamine (DBT) iodide, crystalline N—N-dibutyl-tryptamine (DBT) iodide, and crystalline N—N-diisopropyl-tryptamine (DiPT) hydrofumarate. This disclosure also relates to pharmaceutical compositions containing the DMT analogues or crystalline DMT analogues crystalline EPT hydrofumarate, MALT hydrofumarate, crystalline MALT hydrofumarate, DBT iodide, crystalline DBT iodide, crystalline MPT iodide, crystalline MiPT fumarate, or crystalline DiPT hydrofumarate according to the disclosure. The therapeutic uses of crystalline EPT hydrofumarate, MALT hydrofumarate, crystalline MALT hydrofumarate, DBT iodide, crystalline DBT iodide, or crystalline DiPT hydrofumarate according to the disclosure, are described below as well as compositions containing them. Crystalline EPT hydrofumarate, crystalline MALT hydrofumarate, crystalline DBT iodide, or crystalline DiPT hydrofumarate according to the disclosure, and the methods used to characterize them are described below. The novel and crystalline EPT hydrofumarate, MALT hydrofumarate, crystalline MALT hydrofumarate, DBT iodide, crystalline DBT iodide, or crystalline DiPT hydrofumarate compounds of the disclosure may be used to prepare other salts, including pharmaceutically acceptable salts, by anion exchange techniques known in the art to exchange the fumarate anion for another desired anion. N-methyl-N-allyl-tryptamine (MALT) hydrofumarate and N—N-dibutyl-tryptamine (DBT) iodide are themselves novel compounds. N-ethyl-N-propyl-tryptamine (EPT) hydrofumarate has the following structural formula: N-methyl-N-allyl-tryptamine (MALT) hydrofumarate has the following structural formula: N—N-dibutyl-tryptamine (DBT) iodide has the following structural formula: N—N-diisopropyl-tryptamine (DiPT) hydrofumarate has the following structural formula: Methods of Treatment and Therapeutic Uses In some embodiments a DMT analogue or a crystalline DMT analogue according to the disclosure, and the methods and the compositions—particularly the pharmaceutical compositions—of the disclosure are used to regulate the activity of a neurotransmitter receptor by administering a therapeutically effective dose of a DMT analogue or a crystalline DMT analogue of the disclosure. In another embodiment, a DMT analogue or a crystalline DMT analogue according to the disclosure, and the methods and the compositions—particularly the pharmaceutical compositions—of the disclosure are used to treat inflammation and/or pain by administering a therapeutically effective dose of a DMT analogue or a crystalline DMT analogue of the disclosure. Methods of the disclosure administer a therapeutically effective amount of a DMT analogue or a crystalline DMT analogue of the disclosure to prevent or treat a disease or condition, such as those discussed below for a subject in need of treatment. A DMT analogue or a crystalline DMT analogue of the disclosure may be administered neat or as a composition comprising a DMT analogue or a crystalline DMT analogue of the disclosure as discussed below. A DMT analogue or a crystalline DMT analogue of the disclosure may be used to prevent and/or treat a psychological disorder. The disclosure provides a method for preventing and/or treating a psychological disorder by administering to a subject in need thereof a therapeutically effective amount of a DMT analogue or a crystalline DMT analogue of the disclosure, including the exemplary embodiments discussed herein. The psychological disorder may be chosen from depression, psychotic disorder, schizophrenia, schizophreniform disorder (acute schizophrenic episode); schizoaffective disorder; bipolar I disorder (mania, manic disorder, manic-depressive psychosis); bipolar II disorder; major depressive disorder; major depressive disorder with psychotic feature (psychotic depression); delusional disorders (paranoia); Shared Psychotic Disorder (Shared paranoia disorder); Brief Psychotic disorder (Other and Unspecified Reactive Psychosis); Psychotic disorder not otherwise specified (Unspecified Psychosis); paranoid personality disorder; schizoid personality disorder; schizotypal personality disorder; anxiety disorder; social anxiety disorder; substance-induced anxiety disorder; selective mutism; panic disorder; panic attacks; agoraphobia; attention deficit syndrome, post-traumatic stress disorder (PTSD), premenstrual dysphoric disorder (PMDD), and premenstrual syndrome (PMS). A DMT analogue or a crystalline DMT analogue of the disclosure may be used to prevent and/or treat a brain disorder. The disclosure provides a method for preventing and/or treating a brain disorder by administering to a subject in need thereof a therapeutically effective amount of a DMT analogue or a crystalline DMT analogue of the disclosure, including the exemplary embodiments discussed above. The brain disorder is chosen from Huntington's disease, Alzheimer's disease, dementia, and Parkinson's disease. A DMT analogue or a crystalline DMT analogue of the disclosure may be used to prevent and/or treat developmental disorders, delirium, dementia, amnestic disorders and other cognitive disorders, psychiatric disorders due to a somatic condition, drug-related disorders, schizophrenia and other psychotic disorders, mood disorders, anxiety disorders, somatoform disorders, factitious disorders, dissociative disorders, eating disorders, sleep disorders, impulse control disorders, adjustment disorders, or personality disorders. The disclosure provides a method for preventing and/or treating these disorders by administering to a subject in need thereof a therapeutically effective amount of a DMT analogue or a crystalline DMT analogue of the disclosure, including the exemplary embodiments discussed above. A DMT analogue or a crystalline DMT analogue of the disclosure may be used to prevent and/or treat inflammation and/or pain, such as for example inflammation and/or pain associated with inflammatory skeletal or muscular diseases or conditions. The disclosure provides a method for preventing and/or treating an inflammation and/or pain by administering to a subject in need thereof a therapeutically effective amount of a DMT analogue or a crystalline DMT analogue of the disclosure, including the exemplary embodiments discussed herein. Generally speaking, treatable “pain” includes nociceptive, neuropathic, and mix-type. A method of the disclosure may reduce or alleviate the symptoms associated with inflammation, including but not limited to treating localized manifestation of inflammation characterized by acute or chronic swelling, pain, redness, increased temperature, or loss of function in some cases. A method of the disclosure may reduce or alleviate the symptoms of pain regardless of the cause of the pain, including but not limited to reducing pain of varying severity, i.e. mild, moderate and severe pain, acute pain and chronic pain. A method of the disclosure is effective in treating joint pain, muscle pain, tendon pain, burn pain, and pain caused by inflammation such as rheumatoid arthritis. Skeletal or muscular diseases or conditions which may be treated include but are not limited to musculoskeletal sprains, musculoskeletal strains, tendinopathy, peripheral radiculopathy, osteoarthritis, joint degenerative disease, polymyalgia rheumatica, juvenile arthritis, gout, ankylosing spondylitis, psoriatic arthritis, systemic lupus erythematosus, costochondritis, tendonitis, bursitis, such as the common lateral epicondylitis (tennis elbow), medial epicondylitis (pitchers elbow) and trochanteric bursitis, temporomandibular joint syndrome, and fibromyalgia. A DMT analogue or a crystalline DMT analogue of the disclosure may be used to modulate activity of a mitogen activating protein (MAP), comprising administering a composition of the invention. In one embodiment, the mitogen activating protein (MAP) comprises a MAP kinase (MAPk). MAPKs provide a wide-ranging signaling cascade that allow cells to quickly respond to biotic and abiotic stimuli. Exemplary MAPKs include, but are not limited to, Tropomyosin Receptor Kinase A (TrkA), P38-alpha, Janus Kinase 1 (JAK1), and c-Jun N-Terminal Kinase 3 (JNK3). TrkA is a high affinity catalytic receptor of nerve growth factor (NGF) protein. TrkA regulates NGF response, influencing neuronal differentiation and outgrowth as well as programmed cell death. p38-alpha is involved with the regulation of pro-inflammatory cytokines, including TNF-α. In the central nervous system, p38-alpha regulates neuronal death and neurite degeneration, and it is a common target of Alzheimer's disease therapies. JAK1 influences cytokine signaling, including IL-2, IL-4, IFN-alpha/beta, IFN-γ, and IL-10, and it is implicated in brain aging. JNK3 is neuronal specific protein isoform of the JNKs. It is involved with the regulation of apoptosis. JNK3 also plays a role in modulating the response of cytokines, growth factors, and oxidative stress. As used herein, the term “modulating activity of a mitogen activating protein” refers to changing, manipulating, and/or adjusting the activity of a mitogen activating protein. In one embodiment, modulating the activity of a MAP, such as a MAPK, can influence neural health, neurogenesis, neural growth and differentiation, and neurodegenerative diseases. A DMT analogue or a crystalline DMT analogue of the disclosure may be used to modulate neurogenesis, comprising administering a composition of the invention. As used herein, the term “modulating neurite outgrowth” refers to changing, manipulating, and/or adjusting the growth and development of neural projections, or “neurites.” In one embodiment, neurogenesis comprises modulating the growth of new neurites, the number of neurites per neuron, and/or neurite length. In one embodiment, modulating neurite outgrowth comprises increasing and/or enhancing the rate and/or length at which neurites develop. A DMT analogue or a crystalline DMT analogue of the disclosure may be used to modulate neurite outgrowth, comprising administering a composition of the invention. As used herein, the term “modulating neurogenesis” refers to changing, manipulating, and/or adjusting the growth and development of neural tissue. In one embodiment, neurogenesis comprises adult neurogenesis, in which new neural stem cells are generated from neural stem cells in an adult animal. In one embodiment, modulating neurogenesis comprises increasing and/or enhancing the rate at which new neural tissue is developed. Compositions The disclosure also relates to compositions comprising an effective amount of DMT analogues or crystalline DMT analogues of the disclosure, especially pharmaceutical compositions comprising a therapeutically effective amount of DMT analogues or crystalline DMT analogues of the disclosure and a pharmaceutically acceptable carrier (also known as a pharmaceutically acceptable excipient). As discussed above, DMT analogues or crystalline DMT analogues of the disclosure may be, for example, therapeutically useful to prevent and/or treat the psychological and other disorders discussed above. A composition or a pharmaceutical composition of the disclosure may be in any form which contains DMT analogues or crystalline DMT analogues of the disclosure. The composition may be, for example, a tablet, capsule, liquid suspension, injectable, topical, or transdermal. The compositions or pharmaceutical compositions generally contain, for example, about 1% to about 99% by weight of DMT analogues or crystalline DMT analogues of the disclosure and, for example, 99% to 1% by weight of at least one suitable pharmaceutical excipient. In one embodiment, the composition may be between about 5% and about 75% by weight of DMT analogues or crystalline DMT analogues of the disclosure with the rest being at least one suitable pharmaceutical excipient or at least one other adjuvant, as discussed below. Published US applications US 2018/0221396 A1 and US 2019/0142851 A1 disclose compositions comprising a combination of a first purified psilocybin derivative with a second purified psilocybin derivative, with one or two purified cannabinoids or with a purified terpene. Various ratios of these components in the composition are also disclosed. The disclosures of US 2018/0221396 A1 and US 2019/0142851 A1 are incorporated herein by reference. According to this disclosure, DMT analogues or crystalline DMT analogues of the disclosure may be used as the “first purified psilocybin derivative” in the compositions described in US 2018/0221396 A1 and US 2019/0142851 A1. Accordingly, this disclosure provides a composition comprising as a first component: DMT analogues or crystalline DMT analogues of the disclosure; and a second component selected from (a) a serotonergic drug, (b) a purified psilocybin derivative, (c) one or two purified cannabinoids, and (d) a purified terpene; with the rest being at least one suitable pharmaceutical excipient or at least one other adjuvant, as discussed below. When used in such compositions as a first component comprising one or more of the DMT analogues or crystalline DMT analogues of the disclosure (crystalline EPT hydrofumarate, MALT hydrofumarate, crystalline MALT hydrofumarate, DBT iodide, crystalline DBT iodide, and crystalline DiPT hydrofumarate) with a second component selected from (a) a serotonergic drug, (b) a purified psilocybin derivative, (c) one or two purified cannabinoids, and (d) a purified terpene, the compositions represent particular embodiments of the invention. Compositions having a combination of EPT hydrofumarate, MALT hydrofumarate, DBT iodide, or DiPT hydrofumarate as a first component with a second component selected from(e) an adrenergic drug, (f) a dopaminergic drug, (g) a purified erinacine, and (h) a purified hericenone represent additional particular embodiments of the invention represented by the compositions having the DMT analogues or crystalline DMT analogues of the disclosure. In some embodiments, the first and second components can be administered at the same time (e.g., together in the same composition), or at separate times over the course of treating a patient in need thereof. Such a composition may be a pharmaceutical composition wherein the components are present individually in therapeutically effective amounts or by combination in a therapeutically effective amount to treat a disease, disorder, or condition as described herein. A serotonergic drug refers to a compound that binds to, blocks, or otherwise influences (e.g., via an allosteric reaction) activity at a serotonin receptor as described in paragraphs [0245]-[0253] of US 2018/0221396 A1 and [0305]-[0311] US 2019/0142851 A1 as well as the disclosed exemplary embodiments, incorporated here by reference. Exemplary psilocybin derivatives include but are not limited to psilocybin itself and the psilocybin derivates described in paragraphs [0081]-[0109] of US 2018/0221396 A1 and [082]-[0110] US 2019/0142851 A1 as well as the disclosed exemplary embodiments. Exemplary cannabinoids include but are not limited to the cannabinoids described in paragraphs [0111]-[0159] of US 2018/0221396 A1 and [0112]-[0160] US 2019/0142851 A1 as well as the disclosed exemplary embodiments. Exemplary terpenes include but are not limited to the terpenes described in paragraphs [0160]-[0238] of US 2018/0221396 A1 and [0161]-[0300] US 2019/0142851 A1 as well as the disclosed exemplary embodiments. This invention also relates to a composition or a pharmaceutical formulation that may comprise, consist essentially of, or consist of a DMT analogue or crystalline DMT analogue of the disclosure as a first component; and a second component selected from (e) an adrenergic drug, (f) a dopaminergic drug, (g) a purified erinacine, and (h) a purified hericenone; with the rest being at least one suitable pharmaceutical excipient or at least one other adjuvant, as discussed below. When used in such compositions as a first component the DMT analogues or crystalline DMT analogues of the disclosure (crystalline EPT hydrofumarate, MALT hydrofumarate, crystalline MALT hydrofumarate, DBT iodide, crystalline DBT iodide, or crystalline DiPT hydrofumarate) with a second component selected from (e) an adrenergic drug, (f) a dopaminergic drug, (g) a purified erinacine, and (h) a purified hericenone, the compositions represent particular embodiments of the invention. Compositions having a combination of EPT hydrofumarate, MALT hydrofumarate, DBT iodide, or DiPT hydrofumarate as a first component with a second component selected from (e) an adrenergic drug, (f) a dopaminergic drug, (g) a purified erinacine, and (h) a purified hericenone represent additional particular embodiments of the invention represented by the compositions having the DMT analogues or crystalline DMT analogues of the disclosure. In some embodiments, the first and second components can be administered at the same time (e.g., together in the same composition), or at separate times over the course of treating a patient in need thereof. Such a composition may be a pharmaceutical composition wherein the components are present individually in therapeutically effective amounts or by combination in a therapeutically effective amount to treat a disease, disorder, or condition as described herein. A pharmaceutical formulation of the disclosure may comprise, consist essentially of, or consist of (a) DMT analogues or crystalline DMT analogues of the disclosure and (b) a second active compound selected from a serotonergic drug, a purified psilocybin derivative, a purified cannabinoid, or a purified terpene, an adrenergic drug, a dopaminergic drug, a purified erinacine, and a purified hericenone and (c) a pharmaceutically acceptable excipient. DMT analogues or crystalline DMT analogues of the disclosure and the second active compound are each present in a therapeutically effective amount using a purposefully engineered and unnaturally occurring molar ratios. Exemplary molar ratios of DMT analogues or crystalline DMT analogues of the disclosure to the second active compound in a composition of the disclosure include but are not limited to from about 0.1:100 to about 100:0.1, from about 1:100 to about 100:1, from about 1:50 to about 50:1, from about 1:25 to about 25:1, from about 1:20 to about 20:1, from about 1:10 to about 10:1, from about 1:5 to about 5:1, from about 1:2 to about 2:1 or may be about 1:1. A pharmaceutical formulation of the disclosure may comprise a composition of the disclosure and a serotonergic drug, a purified psilocybin derivative, a purified cannabinoid, or a purified terpene, each present in a therapeutically effective amount using a purposefully engineered and unnaturally occurring molar ratios. Published US applications US 2018/0221396 A1 and US 2019/0142851 A1 disclose compositions comprising a combination of a purified psilocybin derivative with a second purified psilocybin derivative, with one or two purified cannabinoids or with a purified terpene. The disclosures of US 2018/0221396 A1 and US 2019/0142851 A1 are incorporated herein by reference. According to this disclosure a composition containing DMT analogues or crystalline DMT analogues as discussed above may be used in place of a “purified psilocybin derivative” in the compositions described in US 2018/0221396 A1 and US 2019/0142851 A1. Accordingly, the disclosure provides a pharmaceutical formulation comprising as a first component a DMT analogue or a crystalline DMT analogue of the disclosure and a second component selected from (a) a serotonergic drug, (b) a purified psilocybin derivative, (c) one or two purified cannabinoids, (d) a purified terpene, (e) an adrenergic drug, (f) a dopaminergic drug, (g) a purified erinacine, and (h) a purified hericenone; with the rest being at least one suitable pharmaceutical excipient or at least one other adjuvant, as discussed below. Such a composition may be a pharmaceutical composition wherein the components are present individually in therapeutic effective amounts or by combination in a therapeutically effective amount to treat a disease, disorder, or condition as described herein. A serotonergic drug refers to a compound that binds to, blocks, or otherwise influences (e.g., via an allosteric reaction) activity at a serotonin receptor as described in paragraphs [0245]-[0253] of US 2018/0221396 A1 and [0305]-[0311] US 2019/0142851 A1 as well as the disclosed exemplary embodiments, incorporated here by reference. Some exemplary serotonergic drugs include the following molecules: 6-Allyl-N,N-diethyl-NL, N,N-Dibutyl-T, N,N-Diethyl-T, N,N-Diisopropyl-T, 5-Methyoxy-alpha-methyl-T, N,N-Dimethyl-T, 2,alpha-Dimethyl-T, alpha,N-Dimethyl-T, N,N-Dipropyl-T, N-Ethyl-N-isopropyl-T, alpha-Ethyl-T, 6,N,N-Triethyl-NL, 3,4-Dihydro-7-methoxy-1-methyl-C, 7-Methyoxy-1-methyl-C, N,N-Dibutyl-4-hydroxy-T, N,N-Diethyl-4-hydroxy-T, N,N-Diisopropyl-4-hydroxy-T, N,N-Dimethyl-4-hydroxy-T, N,N-Dimethyl-5-hydroxy-T, N, N-Dipropyl-4-hydroxy-T, N-Ethyl-4-hydroxy-N-methyl-T, 4-Hydroxy-N-isopropyl-N-methyl-T, 4-Hydroxy-N-methyl-N-propyl-T, 4-Hydroxy-N,N-tetramethylene-T Ibogaine, N,N-Diethyl-L, N-Butyl-N-methyl-T, N,N-Diisopropyl-4,5-methylenedioxy-T, N,N-Diisopropyl-5,6-methylenedioxy-T, N,N-Dimethyl-4,5-methylenedioxy-T, N,N-Dimethyl-5,6-methylenedioxy-T, N-Isopropyl-N-methyl-5,6-methylenedioxy-T, N,N-Diethyl-2-methyl-T, 2,N,N-Trimethyl-T, N-Acetyl-5-methoxy-T, N,N-Diethyl-5-methoxy-T, N,N-Diisopropyl-5-methoxy-T, 5-Methoxy-N,N-dimethyl-T, N-Isopropyl-4-methoxy-N-methyl-T, N-Isopropyl-5-methoxy-N-methyl-T, 5,6-Dimethoxy-N-isopropyl-N-methyl-T, 5-Methoxy-N-methyl-T, 5-Methoxy-N,N-tetramethylene-T, 6-Methoxy-1-methyl-1,2,3,4-tetrahydro-C, 5-Methoxy-2,N,N-trimethyl-T, N,N-Dimethyl-5-methylthio-T, N-Isopropyl-N-methyl-T, alpha-Methyl-T, N-Ethyl-T, N-Methyl-T, 6-Propyl-N L, N,N-Tetramethylene-T, Tryptamine, and 7-Methoxy-1-methyl-1,2,3,4-tetrahydro-C, alpha,N-Dimethyl-5-methoxy-T. For additional information regarding these compounds See Shulgin, A. T., & Shulgin, A. (2016). Tihkal: The Continuation. Berkeley, Calif.: Transform Press. In one embodiment, a serotonergic drug is chosen from alprazolam, amphetamine, aripiprazole, azapirone, a barbiturate, bromazepam, bupropion, buspirone, a cannabinoid, chlordiazepoxide, citalopram, clonazepam, clorazepate, dextromethorphan, diazepam, duloxetine, escitalopram, fluoxetine, flurazepam, fluvoxamine, lorazepam, lysergic acid diethylamide, lysergamide, 3,4-methylenedioxymethamphetamine, milnacipran, mirtazapine, naratriptan, paroxetine, pethidine, phenethylamine, psicaine, oxazepam, reboxetine, serenic, serotonin, sertraline, temazepam, tramadol, triazolam, a tryptamine, venlafaxine, vortioxetine, and/or derivatives thereof. In a exemplary embodiment, the serotonergic drug is 3,4-methylenedioxymethamphetamine. Exemplary psilocybin derivatives include but are not limited to psilocybin itself and the psilocybin derivates described in paragraphs [0081]-[0109] of US 2018/0221396 A1 and [082]-[0110] US 2019/0142851 A1 as well as the disclosed exemplary embodiments, incorporated here by reference. In one embodiment, the compositions disclosed herein comprise one or more purified psilocybin derivatives chosen from: [3-(2-Dimethylaminoethyl)-1H-indol-4-yl] dihydrogen phosphate, 4-hydroxytryptamine, 4-hydroxy-N,N-dimethyltryptamine, [3-(2-methylaminoethyl)-1H-indol-4-yl]dihydrogen phosphate, 4-hydroxy-N-methyltryptamine, [3-(aminoethyl)-1H-indol-4-yl] dihydrogen phosphate, [3-(2-trimethylaminoethyl)-1H-indol-4-yl] dihydrogen phosphate, and 4-hydroxy-N,N,N-trimethyltryptamine. Exemplary cannabinoids include but are not limited to the cannabinoids described in paragraphs [0111]-[0159] of US 2018/0221396 A1 and [0112]-[0160] US 2019/0142851 A1 as well as the disclosed exemplary embodiments, incorporated here by reference. Examples of cannabinoids within the context of this disclosure include the following molecules: Cannabichromene (CBC), Cannabichromenic acid (CBCA), Cannabichromevarin (CBCV), Cannabichromevarinic acid (CBCVA), Cannabicyclol (CBL), Cannabicyclolic acid (CBLA), Cannabicyclovarin (CBLV), Cannabidiol (CBD), Cannabidiol monomethylether (CBDM), Cannabidiolic acid (CBDA), Cannabidiorcol (CBD-C1), Cannabidivarin (CBDV), Cannabidivarinic acid (CBDVA), Cannabielsoic acid B (CBEA-B), Cannabielsoin (CBE), Cannabielsoin acid A (CBEA-A), Cannabigerol (CBG), Cannabigerol monomethylether (CBGM), Cannabigerolic acid (CBGA), Cannabigerolic acid monomethylether (CBGAM), Cannabigerovarin (CBGV), Cannabigerovarinic acid (CBGVA), Cannabinodiol (CBND), Cannabinodivarin (CBDV), Cannabinol (CBN), Cannabinol methylether (CBNM), Cannabinol-C2 (CBN-C2), Cannabinol-C4 (CBN-C4), Cannabinolic acid (CBNA), Cannabiorcool (CBN-C1), Cannabivarin (CBV), Cannabitriol (CBT), Cannabitriolvarin (CBTV), 10-Ethoxy-9-hydroxy-delta-6a-tetrahydrocannabinol, Cannbicitran (CBT), Cannabiripsol (CBR), 8,9-Dihydroxy-delta-6a-tetrahydrocannabinol, Delta-8-tetrahydrocannabinol (A8-THC), Delta-8-tetrahydrocannabinolic acid (A8-THCA), Delta-9-tetrahydrocannabinol (THC), Delta-9-tetrahydrocannabinol-C4 (THC-C4), Delta-9-tetrahydrocannabinolic acid A (THCA-A), Delta-9-tetrahydrocannabinolic acid B (THCA-B), Delta-9-tetrahydrocannabinolic acid-C4 (THCA-C4), Delta-9-tetrahydrocannabiorcol (THC-C1), Delta-9-tetrahydrocannabiorcolic acid (THCA-C1), Delta-9-tetrahydrocannabivarin (THCV), Delta-9-tetrahydrocannabivarinic acid (THCVA), 10-Oxo-delta-6a-tetrahydrocannabinol (OTHC), Cannabichromanon (CBCF), Cannabifuran (CBF), Cannabiglendol, Delta-9-cis-tetrahydrocannabinol (cis-THC), Tryhydroxy-delta-9-tetrahydrocannabinol (triOH-THC), Dehydrocannabifuran (DCBF), and 3,4,5,6-Tetrahydro-7-hydroxy-alpha-alpha-2-trimethyl-9-n-propyl-2,6-metha-no-2H-1-benzoxocin-5-methanol. In one embodiment, the purified cannabinoid is chosen from THC, THCA, THCV, THCVA, CBC, CBCA, CBCV, CBCVA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBGV, or CBGVA. Exemplary terpenes include but are not limited to the terpenes described in paragraphs [0160]-[0238] of US 2018/0221396 A1 and [0161]-[0300] US 2019/0142851 A1 as well as the disclosed exemplary embodiments, incorporated here by reference. In one embodiment, a purified terpene is chosen from acetanisole, acetyl cedrene, anethole, anisole, benzaldehyde, bornyl acetate, borneol, cadinene, cafestol, caffeic acid, camphene, camphor, capsaicin, carene, carotene, carvacrol, carvone, caryophyllene, caryophyllene, caryophyllene oxide, cedrene, cedrene epoxide, cecanal, cedrol, cembrene, cinnamaldehyde, cinnamic acid, citronellal, citronellol, cymene, eicosane, elemene, estragole, ethyl acetate, ethyl cinnamate, ethyl maltol, eucalyptol/1,8-cineole, eudesmol, eugenol, euphol, farnesene, farnesol, fenchone, geraniol, geranyl acetate, guaia-1(10),11-diene, guaiacol, guaiol, guaiene, gurjunene, herniarin, hexanaldehyde, hexanoic acid, humulene, ionone, ipsdienol, isoamyl acetate, isoamyl alcohol, isoamyl formate, isoborneol, isomyrcenol, isoprene, isopulegol, isovaleric acid, lavandulol, limonene, gamma-linolenic acid, linalool, longifolene, lycopene, menthol, methyl butyrate, 3-mercapto-2-methylpentanal, beta-mercaptoethanol, mercaptoacetic acid, methyl salicylate, methylbutenol, methyl-2-methylvalerate, methyl thiobutyrate, myrcene, gamma-muurolene, nepetalactone, nerol, nerolidol, neryl acetate, nonanaldehyde, nonanoic acid, ocimene, octanal, octanoic acid, pentyl butyrate, phellandrene, phenylacetaldehyde, phenylacetic acid, phenylethanethiol, phytol, pinene, propanethiol, pristimerin, pulegone, retinol, rutin, sabinene, squalene, taxadiene, terpineol, terpine-4-ol, terpinolene, thujone, thymol, umbelliferone, undecanal, verdoxan, or vanillin. In one embodiment, a purified terpene is chosen from bornyl acetate, alpha-bisabolol, borneol, camphene, camphor, carene, caryophyllene, cedrene, cymene, elemene, eucalyptol, eudesmol, farnesene, fenchol, geraniol, guaiacol, humulene, isoborneol, limonene, linalool, menthol, myrcene, nerolidol, ocimene, phellandrene, phytol, pinene, pulegone, sabinene, terpineol, terpinolene, or valencene. As used herein, the term “adrenergic drug” refers to a compound that binds, blocks, or otherwise influences (e.g., via an allosteric reaction) activity at an adrenergic receptor. In one embodiment, an adrenergic drug binds to an adrenergic receptor. In one embodiment, an adrenergic drug indirectly affects an adrenergic receptor, e.g., via interactions affecting the reactivity of other molecules at the adrenergic receptor. In one embodiment, an adrenergic drug is an agonist, e.g., a compound activating an adrenergic receptor. In one embodiment, an adrenergic drug is an antagonist, e.g., a compound binding but not activating an adrenergic receptor, e.g., blocking a receptor. In one embodiment, an adrenergic drug is an effector molecule, e.g., a compound binding to an enzyme for allosteric regulation. In one embodiment, an adrenergic drug acts (either directly or indirectly) at more than one type of receptor (e.g., 5HT, dopamine, adrenergic, acetylcholine, etc.). In one embodiment, an adrenergic drug is an antidepressant. In one embodiment, an adrenergic drug is a norepinephrine transporter inhibitor. In one embodiment, an adrenergic drug is a vesicular monoamine transporter inhibitor. In one embodiment, an adrenergic drug is chosen from adrenaline, agmatine, amoxapine, aptazapine, atomoxetine, bupropion, clonidine, doxepin, duloxetine, esmirtazpine, mianserin, ketanserin, mirabegron, mirtazapine, norepinephrine, phentolamine, phenylephrine, piperoxan, reserpine, ritodrine, setiptiline, tesofensine, timolol, trazodone, trimipramine, or xylazine. As used herein, the term “dopaminergic drug” refers to a compound that binds, blocks, or otherwise influences (e.g., via an allosteric reaction) activity at a dopamine receptor. In one embodiment, a dopaminergic drug binds to a dopamine receptor. In one embodiment, a dopaminergic drug indirectly affects a dopamine receptor, e.g., via interactions affecting the reactivity of other molecules at the dopamine receptor. In one embodiment, a dopaminergic drug is an agonist, e.g., a compound activating a dopamine receptor. In one embodiment, a dopaminergic drug is an antagonist, e.g., a compound binding but not activating a dopamine receptor, e.g., blocking a receptor. In one embodiment, a dopaminergic drug is an effector molecule, e.g., a compound binding to an enzyme for allosteric regulation. In one embodiment, a dopaminergic drug acts (either directly or indirectly) at more than one type of receptor (e.g., 5HT, dopamine, adrenergic, acetylcholine, etc.). In one embodiment, a dopaminergic drug is a dopamine transporter inhibitor. In one embodiment, a dopaminergic drug is a vesicular monoamine transporter inhibitor. In one embodiment, a dopaminergic drug is chosen from amineptine, apomorphine, benzylpiperazine, bromocriptine, cabergoline, chlorpromazine, clozapine, dihydrexidine, domperidone, dopamine, fluphenazine, haloperidol, ketamine, loxapine, methamphetamine, olanzapine, pemoline, perphenazine, pergolide, phencyclidine, phenethylamine, phenmetrazine, pimozide, piribedil, a psychostimulant, reserpine, risperidone, ropinirole, tetrabenazine, or thioridazine. In one embodiment, the compositions and methods disclosed herein include one or more purified erinacine molecules. In one embodiment, the compositions and methods disclosed herein comprise purified erinacine A. In one embodiment, the compositions and methods disclosed herein comprise erinacine B. In one embodiment, the compositions and methods disclosed herein comprise erinacine C. In one embodiment, the compositions and methods disclosed herein comprise erinacine D. In one embodiment, the compositions and methods disclosed herein comprise erinacine E. In one embodiment, the compositions and methods disclosed herein comprise erinacine F. In one embodiment, the compositions and methods disclosed herein comprise erinacine G. In one embodiment, the compositions and methods disclosed herein comprise erinacine H. In one embodiment, the compositions and methods disclosed herein comprise erinacine I. In one embodiment, the compositions and methods disclosed herein comprise erinacine.J. In one embodiment, the compositions and methods disclosed herein comprise erinacine K In one embodiment, the compositions and methods disclosed herein comprise erinacine P. In one embodiment, the compositions and methods disclosed herein comprise erinacine Q. In one embodiment, the compositions and methods disclosed herein comprise erinacine R. In one embodiment, the compositions and methods disclosed herein comprise erinacine S. In one embodiment, the compositions and methods disclosed herein include one or more purified hericenone molecules. In one embodiment, the compositions and methods disclosed herein comprise purified hericenone A. In one embodiment, the compositions and methods disclosed herein comprise purified hericenone B. In one embodiment, the compositions and methods disclosed herein comprise purified hericenone C. In one embodiment, the compositions and methods disclosed herein comprise purified hericenone D. In one embodiment, the compositions and methods disclosed herein comprise purified hericenone E. In one embodiment, the compositions and methods disclosed herein comprise purified hericenone F. In one embodiment, the compositions and methods disclosed herein comprise purified hericenone G. In one embodiment, the compositions and methods disclosed herein comprise purified hericenone H. Exemplary compositions of DMT analogues of the disclosure and a second compound selected from a serotonergic drug, a purified psilocybin derivative, a purified cannabinoid, a purified terpene, an adrenergic drug, a dopaminergic drug, a purified erinacine, or a purified hericenone in exemplary molar ratios are shown in Table 1. TABLE 1Molar ratio of DMTMolar ratio of DMTMolar ratio of DMTanalogues: secondanalogues: secondanalogues: secondSecond Compoundcompoundcompoundcompound3,4-About 1:100 toAbout 1:25 toAbout 1:5 tomethylenedioxymethamphetamineabout 100:1about 25:1about 5:1CitalopramAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1EscitalopramAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1FluoxetineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1ParoxetineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1SertralineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1[3-(2-Dimethylaminoethyl)-1H-About 1:100 toAbout 1:25 toAbout 1:5 toindol-4-yl] dihydrogen phosphateabout 100:1about 25:1about 5:14-hydroxytryptamineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:14-hydroxy-N,N-dimethyltryptamineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1[3-(2-methylaminoethyl)-1H-indol-About 1:100 toAbout 1:25 toAbout 1:5 to4-yl] dihydrogen phosphateabout 100:1about 25:1about 5:14-hydroxy-N-methyltryptamineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1[3-(aminoethyl)-1H-indol-4-yl]About 1:100 toAbout 1:25 toAbout 1:5 todihydrogen phosphateabout 100:1about 25:1about 5:1[3-(2-trimethylaminoethyl)-1H-About 1:100 toAbout 1:25 toAbout 1:5 toindol-4-yl] dihydrogen phosphateabout 100:1about 25:1about 5:14-hydroxy-N,N,N-About 1:100 toAbout 1:25 toAbout 1:5 totrimethyltryptamineabout 100:1about 25:1about 5:1THCAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1CBCAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1CBDAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1CBGAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1MyrceneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1PineneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1CaryophylleneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1LimoneneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1HumuleneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1LinaloolAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1AdrenalineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1AmineptineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1Erinacine AAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1Hericenone AAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1 Exemplary pharmaceutical compositions of DMT analogues of the disclosure and a second compound selected from a serotonergic drug, a purified psilocybin derivative, a purified cannabinoid, or a purified terpene and an excipient with exemplary molar ratios of DMT analogues of the disclosure to the second compound are shown in Table 2. TABLE 2Molar ratio DMTMolar ratio of DMTMolar ratio of DMTanalogues: secondanalogues: secondanalogues: secondSecond Compoundcompoundcompoundcompound3,4-About 1:100 toAbout 1:25 toAbout 1:5 tomethylenedioxymethamphetamineabout 100:1about 25:1about 5:1CitalopramAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1EscitalopramAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1FluoxetineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1ParoxetineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1SertralineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1[3-(2-Dimethylaminoethyl)-1H-About 1:100 toAbout 1:25 toAbout 1:5 toindol-4-yl] dihydrogen phosphateabout 100:1about 25:1about 5:14-hydroxytryptamineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:14-hydroxy-N,N-dimethyltryptamineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1[3-(2-methylaminoethyl)-1H-indol-About 1:100 toAbout 1:25 toAbout 1:5 to4-yl] dihydrogen phosphateabout 100:1about 25:1about 5:14-hydroxy-N-methyltryptamineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1[3-(aminoethyl)-1H-indol-4-yl]About 1:100 toAbout 1:25 toAbout 1:5 todihydrogen phosphateabout 100:1about 25:1about 5:1[3-(2-trimethylaminoethyl)-1H-About 1:100 toAbout 1:25 toAbout 1:5 toindol-4-yl] dihydrogen phosphateabout 100:1about 25:1about 5:14-hydroxy-N,N,N-About 1:100 toAbout 1:25 toAbout 1:5 totrimethyltryptamineabout 100:1about 25:1about 5:1THCAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1CBCAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1CBDAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1CBGAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1MyrceneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1PineneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1CaryophylleneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1LimoneneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1HumuleneAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1LinaloolAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1AdrenalineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1AmineptineAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1Erinacine AAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1Hericenone AAbout 1:100 toAbout 1:25 toAbout 1:5 toabout 100:1about 25:1about 5:1 An “effective amount” or a “therapeutically effective amount” of DMT analogues or crystalline DMT analogues according to the disclosure is generally in the range of about 0.1 to about 100 mg daily (oral dose), of about 0.1 to about 50 mg daily (oral dose) of about 0.25 to about 25 mg daily (oral dose), of about 0.1 to about 5 mg daily (oral dose) or of about 0.5 to about 2.5 mg daily (oral dose). The actual amount required for treatment of any particular patient may depend upon a variety of factors including, for example, the disease being treated and its severity; the specific pharmaceutical composition employed; the age, body weight, general health, sex, and diet of the patient; the mode of administration; the time of administration; the route of administration; and the rate of excretion; the duration of the treatment; any drugs used in combination or coincidental with the specific compound employed; and other such factors well known in the medical arts. These factors are discussed in Goodman and Gilman's “The Pharmacological Basis of Therapeutics,” Tenth Edition, A. Gilman, J. Hardman and L Limbird, eds., McGraw-Hill Press, 155-173 (2001), which is incorporated herein by reference. DMT analogues or crystalline DMT analogues according to the disclosure, compositions and pharmaceutical compositions containing them may be used in combination with other agents that are generally administered to a patient being treated for psychological and other disorders discussed above. They may also be co-formulated with one or more of such agents in a single pharmaceutical composition. Depending on the type of composition or pharmaceutical composition, the excipient or pharmaceutically acceptable carrier may be chosen from any one or a combination of carriers known in the art. The choice of the pharmaceutically acceptable carrier depends upon the pharmaceutical form and the desired method of administration to be used. Exemplary carriers include those that do not substantially alter DMT analogues or crystalline DMT analogues of the disclosure or produce undesirable biological effects or otherwise interact in a deleterious manner with any other component(s) of the pharmaceutical composition. The compositions or pharmaceutical compositions of the disclosure may be prepared by methods known in the pharmaceutical formulation art, for example, see Remington's Pharmaceutical Sciences, 18th Ed., (Mack Publishing Company, Easton, Pa., 1990), which is incorporated herein by reference. In a solid dosage form, DMT analogues or crystalline DMT analogues of the disclosure may be admixed with at least one pharmaceutically acceptable excipient such as, for example, sodium citrate or dicalcium phosphate or (a) fillers or extenders, such as, for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders, such as, for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia, (c) humectants, such as, for example, glycerol, (d) disintegrating agents, such as, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate, (e) solution retarders, such as, for example, paraffin, (f) absorption accelerators, such as, for example, quaternary ammonium compounds, (g) wetting agents, such as, for example, cetyl alcohol, and glycerol monostearate, magnesium stearate and the like (h) adsorbents, such as, for example, kaolin and bentonite, and (i) lubricants, such as, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Excipients or pharmaceutically acceptable adjuvants known in the formulation art may also be used in the pharmaceutical compositions of the disclosure. These include, but are not limited to, preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prevention of the action of microorganisms may be ensured by inclusion of various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. If desired, a composition or a pharmaceutical composition of the disclosure may also contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, antioxidants, and the like, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylated hydroxytoluene, etc. Solid dosage forms as described above may be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain pacifying agents and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Non-limiting examples of embedded compositions that may be used are polymeric substances and waxes. The active compounds may also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients. Suspensions, in addition to the active compounds, may contain suspending agents, such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like. Solid dosage forms for oral administration, which includes capsules, tablets, pills, powders, and granules, may be used. In such solid dosage forms, the active compound may be mixed with at least one inert, pharmaceutically acceptable excipient (also known as a pharmaceutically acceptable carrier). Administration of DMT analogues or crystalline DMT analogues of the disclosure in pure form, with a permeation enhancer, with stabilizers (e.g., antioxidants), or in an appropriate pharmaceutical composition may be carried out via any of the accepted modes of administration or agents for serving similar utilities. Thus, administration may be, for example, orally, buccally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, or intrasystemically, in the form of solid, semi-solid, lyophilized powder, liquid dosage forms, such as, for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, suspensions, or aerosols, or the like, such as, for example, in unit dosage forms suitable for simple administration of precise dosages. One route of administration may be oral administration, using a convenient daily dosage regimen that can be adjusted according to the degree of severity of the disease-state to be treated. EXAMPLES Example 1: Crystalline N-ethyl-N-propyl-tryptammonium hydrofumarate Preparation: Single crystals of N-ethyl-N-propyl-tryptammonium (EPT) hydrofumarate suitable for X-ray analysis were obtained from the slow evaporation of an aqueous solution of a commercial sample of EPT fumarate (The Indole Shop, Canada). Single Crystal Characterization: Crystal data, data collection and structure refinement details for crystalline N-ethyl-N-propyl-tryptammonium (EPT) hydrofumarate are summarized in Table 3. TABLE 3EPT hydrofumarateChemical formulaC4H3O4•C15H23N2Mr346.42Crystal system, space groupMonoclinic, P21Temperature (K)297a, b, c (Å)7.4839 (8), 14.1752 (14), 9.6461 (10)α, β, γ (°)90, 110.537 (3), 90V (Å3)958.28 (17)Z2Radiation typeMo Kαμ (mm−1)0.08Crystal size (mm)0.42 × 0.2 × 0.1DiffractometerBruker D8 Venture CMOSAbsorption correctionMulti-scan SADABS2016/2 (Bruker, 2016 February) was used forabsorption correction. wR2(int) was 0.0636 before and0.0481 after correction. The Ratio of minimum tomaximum transmission is 0.9438. The λ/2 correctionfactor is not present.Tmin, Tmax0.703, 0.745No. of measured, independent and21982, 3570, 3368observed [I > 2σ(I)] reflectionsRint0.028(sin q/l)max(Å−1)0.611R[F2> 2σ(F2)], wR(F2), S0.034, 0.093, 1.03No. of reflections3570No. of parameters261No. of restraints7H-atom treatmentH atoms treated by a mixture of independent andconstrained refinementΔmax, Δmin(e Å−3)0.13, −0.13Absolute structureFlack x determined using 1509 quotients [(I+) − (I−)]/[(I+) + (I−)] (Parsons, Flack and Wagner, Acta Cryst.B69 (2013) 249-259).Absolute structure parameter0.1 (2)Computer programs: OLEX3 (Bruker, 2018), APEX3 (Bruker, 2018), SAINT (Bruker, 2018), SHELXT2014 (Sheldrick, 2015a), SHELXL2018 (Sheldrick, 2015b), OLEX2 (Dolomanov et al., 2009), pubICIF (Westrip, 2010). The molecular structure of crystalline N-ethyl-N-propyltryptammonium (EPT) hydrofumarate showing the atom labelling is shown inFIG.1. Displacement ellipsoids are drawn at the 50% probability level. Dashed bonds indicate the disordered component of the structure. Hydrogen bonds are shown as dashed lines. InFIG.1only one component of the disorder is shown.FIG.2shows the hydrogen bonding of a hydrofumarate ion in the structure of crystalline N-ethyl-N-propyltryptammonium hydrofumarate, with hydrogen bonds shown as dashed lines. Displacement ellipsoids are drawn at the 50% probability level. Only one component of the disorder is shown, and hydrogen atoms not involved in hydrogen bonding are omitted for clarity. Symmetry codes: (i) −1+x, y, z; (ii) 1+x, y, z; (iii) 2−x, −1/2+y, 1−z.FIG.3shows the unit cell of crystalline N-ethyl-N-propyl-tryptammonium (EPT) hydrofumarate. The crystal packing of crystalline EPT hydrofumarate is shown along the a axis. The hydrogen bonds are shown as dashed lines. Displacement ellipsoids are drawn at the 50% probability level. Hydrogen atoms not involved in hydrogen bonding are omitted for clarity. InFIG.1only one component of the disorder is shown. The tryptammonium cations and the hydrofumarate anions of the EPT salt are held together in infinite two-dimensional networks parallel to (001) through N—H. . . O and O—H. . . O hydrogen bonds. The indole N—H hydrogen bonds to the carbonyl oxygen of the carboxylic acid of a hydrofumarate molecule. The ammonium N—H and the carboxylic acid O—H each hydrogen bond to one of the carboxylate oxygens (FIG.2). The packing of N-ethyl-N-propyl-tryptammonium hydrofumarate is shown inFIG.3. Simulated Powder X-ray Diffraction (PXRD) Pattern FIG.4is a simulated X-ray powder diffraction (XRPD) pattern for crystalline N-ethyl-N-propyl-tryptammonium (EPT) hydrofumarate generated from its single crystal data. Table 4 lists the angles, °2θ±0.2° 2θ, and d-spacing of the peaks identified in the experimental XRPD pattern ofFIG.4. The entire list of peaks, or a subset thereof, may be sufficient to characterize the cocrystal. For example, the cocrystal may be characterized by at least two peaks selected from the peaks at 11.6, 15.9 and 21.2°2θ±0.2°2θ as well as by a XRPD pattern substantially similar toFIG.4. TABLE 4d-spacing2 Theta(deg)Intensity9.039.894.7.6211.62669.8447.0912.5326.8267.0112.616.202466.8113.01006.2186.2814.11839.7566.1414.44075.25.5815.918046.364.9817.815375.64.9118.08620.684.7818.556514.6019.31696.4684.5319.68778.844.5219.6254.7644.3820.31580.7684.3020.66799.284.1921.210155.43.9722.423493.643.9122.74862.123.8822.91817.3123.8623.018618.523.8123.31087.7363.7423.81622.893.6224.61833.883.5425.116106.723.5025.44534.83.4126.1556.983.4026.26586.283.3626.5349053.3126.9721.243.3126.9573.223.3127.045.37343.3027.017353.643.3027.0540.8723.2627.32692.9243.2227.72637.9883.2027.82.074523.1628.23142.3243.1428.4835.2043.1428.43552.6963.1328.5543.0243.0729.1846.5963.0129.62255.53.0029.8287.1924 Example 2: N-methyl-N-allyl-tryptammonium (MALT) hydrofumarate and crystalline MALT hydrofumarate Preparation: 134 mg of a commercial sample of N-methyl-N-allyl-tryptamine (The Indole Shop, Canada), which is a waxy solid that does not crystallize well, was dissolved in 10 mL of methanol, and 68 mg of fumaric acid was added. The mixture was refluxed for 12 hours and solvent was removed in vacuo to obtain a waxy, yellow product. The material was recrystallized from ethanol to yield colorless single crystals suitable for X-ray diffraction. The product was also characterized by nuclear magnetic resonance.1H NMR (400 MHz, D2O): d 7.69 (d, J=7.9 Hz, 1H, ArH), 7.34 (s, 1H, ArH), 7.29 (t, J=7.1 Hz, 1 H, ArH), 7.21 (t, J=7.1 Hz, 1H, ArH), 6.66 (s, 2H, CH), 6.92-5.82 (m, 1H, CH), 5.60-5.56 (m, 2H, CH2), 3.88-3.83 (m, 1H, CH2), 3.77-3.72 (m, 1H, CH2), 3.68-3.57 (m, 1H, CH2), 3.44-3.37 (m, 1H, CH2), 3.34-3.21 (m, 2H, CH2), 2.90 (s, 3H, CH3).13C NMR (100 MHz, D20): d 172.2 (COOH), 137.0 (CH), 135.5 (ArC), 127.3 (ArC), 126.9 (ArC), 126.2 (ArC), 124.8 (ArC), 122.9 (ArC), 120.1 (ArC), 118.9 (ArC), 112.7 (sp2q, 109.0 (sp2C), 58.7 (AkC), 55.6 (AkC), 40.1 (AkC), 20.6 (AkC). Single Crystal Characterization: Crystal data, data collection and structure refinement details are summarized in Table 5. TABLE 5MALT HydrofumarateChemical formulaC4H3O4•C14H19N2Mr330.37Crystal system, space groupOrthorhombic, P212121Temperature (K)297a, b, c (Å)7.9845 (7), 8.5641 (6), 25.649 (2)α, β, γ (°)90, 90, 90V (Å3)1753.9 (3)Z4Radiation typeMo Kαμ (mm−1)0.09Crystal size (mm)0.42 × 0.24 × 0.15DiffractometerBruker D8 Venture CMOSAbsorption correctionMulti-scanSADABS2016/2 (Bruker, 2016 February) was used forabsorption correction. wR2(int) was 0.0704 before and0.0622 after correction. The Ratio of minimum tomaximum transmission is 0.9131. The λ/2 correctionfactor is not present.Tmin, Tmax0.681, 0.745No. of measured,49712, 3318, 3036independent andobserved [I > 2σ(I)]reflectionsRint0.046(sin q/l)max(Å−1)0.610R[F2> 2σ(F2)], wR(F2), S0.053, 0.147, 1.10No. of reflections3318No. of parameters228No. of restraints4H-atom treatmentH atoms treated by a mixture of independent andconstrained refinementΔmax, Δmin(e Å−3)0.25, −0.17Absolute structureFlack x determined using 1177 quotients [(I+) − (I−)]/[(I+) + (I−)] (Parsons, Flack and Wagner, Acta Cryst.B69 (2013) 249-259).Absolute structure0.0 (3)parameterComputer programs: OLEX3 (Bruker, 2018), APEX3 (Bruker, 2018), SAINT (Bruker, 2018), SHELXT2014 (Sheldrick, 2015a), SHELXL2018 (Sheldrick, 2015b), OLEX2 (Dolomanov et al., 2009), pubICIF (Westrip, 2010). The molecular structure of N-methyl-N-allyltryptammonium hydrofumarate showing the atom labelling is shown inFIG.5. Displacement ellipsoids are drawn at the 50% probability level. Hydrogen bonds are shown as dashed lines.FIG.4shows the hydrogen bonding of a hydrofumarate ion in the structure of N-methyl-N-allyltryptammonium hydrofumarate, with hydrogen bonds shown as dashed lines. Displacement ellipsoids are drawn at the 50% probability level. Hydrogen atoms not involved in hydrogen bonding are omitted for clarity. Symmetry codes: (i) 1+x, y, z; (ii) 1−x, 1/2+y, 1/2−z; (iii) −1+x, y, z; (iv) −x, 1/2+y, 1/2−z.FIG.7shows the crystal packing of MALT hydrofumarate shown along the b axis. The hydrogen bonds are shown as dashed lines. Displacement ellipsoids are drawn at the 50% probability level. Hydrogen atoms not involved in hydrogen bonding are omitted for clarity. The unit cell of N-methyl-N-allyl-tryptammonium hydrofumarate contains one tryptammonium cation and one hydrofumarate anion (FIG.7). The tryptammonium has a near planar indole, with a mean deviation from planarity of 0.007 Å. The ethyl-amino group is turned away from the plane of the indole, with a C1-C8-C9-C10 torsion angle of −105.5 (5). The hydrofumarate is also near planar, with a r.m.s. deviation of 0.055 Å. The carboxylate is partially delocalized, with C—O distances of 1.239 (5) A and 1.259 (4) A. The tryptammonium cations and the hydro-fumarate anions of the MALT salt are held together in infinite two-dimensional networks parallel to (001) through N—H. . . O and O—H. . . O hydrogen bonds. The ammonium N—H hydrogen bonds to one of the carboxyl-ate oxygens. The indole N—H has a three-centre (bifurcated) hydrogen bond with the carbonyl oxygen of a carboxylic acid of a hydrofumarate anion, and a carboxyl-ate oxygen of a different hydrofumarate anion. The carb-oxy-lic acid O—H hydrogen bonds to a carboxyl-ate oxygen of another hydrofumarate anion. The packing of the N-methyl-N-allyl-tryptammonium hydrofumarate is shown inFIG.7. Simulated Powder X-ray Diffraction (PXRD) Pattern:FIG.8is a simulated X-ray powder diffraction (XRPD) pattern for crystalline N-ethyl-N-propyl-tryptammonium (EPT) hydrofumarate generated from its single crystal data. Table 6 lists the angles, °2θ±0.2°2θ, and d-spacing of the peaks identified in the experimental XRPD pattern ofFIG.8. The entire list of peaks, or a subset thereof, may be sufficient to characterize the cocrystal. For example, the cocrystal may be characterized by at least two or at least three peaks selected from the peaks at 11.6, 13.0, 13.8, 16.6 and 18.3°2θ±0.2°2θ as well as by a XRPD pattern substantially similar toFIG.8. TABLE 6d-spacing2 Theta(deg)Intensity12.826.90.2428688.1210.98574.847.6211.68656.487.1212.470.46766.7813.013427.566.4113.82507.646.0514.6387.2045.8415.124206.645.8415.19471.485.6915.531756.325.3116.630182.85.1317.21.1813085.0017.71084.8524.8218.35484.6644.4020.111304.484.3220.417683.924.3220.51595.14.2820.6801.5464.2720.649909.44.2220.9585.544.0621.782909.23.9922.117132.883.9422.4321.75123.8522.92544.7283.8323.185018.43.8223.16811.843.8123.2473.643.7723.428270.923.7723.4896.223.7323.634492.323.6224.452610.83.6224.413947.523.6224.42417.7283.5824.613977.923.5624.881.62763.4825.324432.43.4525.646305.63.4525.662.193363.3926.03119.163.3726.281174.43.3326.55491.923.3326.5132.21763.2926.91413.8883.2527.16170.4563.2127.547700.43.1528.03461.23.1528.024178.963.1028.48100.963.0429.05070.0243.0329.26579.723.0029.4907.988 Example 3: N—N-dibutyl-tryptamine (DBT) iodide and crystalline DBT iodide Preparation: 208 mg of tryptamine was dissolved in 10 mL of THF and 1.50 mL of 1-lodobutane. The mixture was refluxed under nitrogen for 5 days. The solvent was removed in vacuo to yield a brown oil. The oil was recrystallized from acetone to yield colorless crystalline solid. Crystals suitable for X-ray diffraction were obtained from slow evaporation of an ethanol solution. The product was also characterized by nuclear magnetic resonance.1H NMR (400 MHz, D20) δ 7.70 (d, J=8.0 Hz, 1H, ArH), 7.55 (d, J=7.6 Hz, 1H, ArH), 7.34-7.28 (m, 2H, ArH), 7.22 (t, J=8.0 Hz, 1H, ArH), 3.53 (t, J=7.3 Hz, 2H, CH2), 3.29-3.18 (m, 6H, CH2), 1.69-1.61 (m, 4H, CH2), 1.36-1.31 (m, 4H, CH2), 0.90 (t, J=7.4 Hz, 6H, CH3). Single Crystal Characterization: Crystal data, data collection and structure refinement details are summarized in Table 7. TABLE 7DBT iodideChemical formulaI•C18H29N2Mr400.33Crystal system, space groupOrthorhombic, PbcaTemperature (K)273a, b, c (Å)10.506 (2), 14.860 (3), 24.540 (5)V (Å3)3831.0 (13)Z8Radiation typeMo Kαμ (mm−1)1.67Crystal size (mm)0.25 × 0.1 × 0.02DiffractometerBruker APEX-II CCDAbsorption correctionMulti-scanSADABS2016/2 (Bruker, 2016 February) was used forabsorption correction. wR2(int) was 0.0617 before and0.0540 after correction. The Ratio of minimum tomaximum transmission is 0.9173. The λ/2 correctionfactor is Not present.Tmin, Tmax0.684, 0.745No. of measured,49317, 3510, 2546independent andobserved [I > 2σ(I)]reflectionsRint0.090R[F2> 2σ(F2)], WR(F2), S0.046, 0.070, 1.13No. of reflections3510No. of parameters218No. of restraints26H-atom treatmentH atoms treated by a mixture of independent andconstrained refinementΔmax, Δmin(e Å−3)0.39, −0.59Data Collection: SAINT V8.40A (Bruker, 2019); data reduction: SAINT V8.40A (Bruker, 2019); program(s) used to solve structure: SHELXT 2014/5 (Sheldrick, 2014); program(s) used to refine structure: SHELXL 2018 March (Sheldrick, 2015); molecular graphics: Olex2 1.3 (Dolomanov et al., 2009); software used to prepare material for publication: Olex2 1.3 (Dolomanov et al., 2009). Simulated Powder X-ray Diffraction (PXRD) Pattern:FIG.11is a simulated X-ray powder diffraction (XRPD) pattern for crystalline N—N-dibutyl-tryptamine (DBT) iodide generated from its single crystal data. Table 8 lists the angles, *28±0.2′20, and d-spacing of the peaks identified in the experimental XRPD pattern ofFIG.11. The entire list of peaks, or a subset thereof, may be sufficient to characterize the cocrystal. For example, the cocrystal may be characterized by the peaks at 7.2, 14.4, and 16.1°′2θ±0.2°2θ as well as by a XRPD pattern substantially similar toFIG.11. TABLE 8d-spacing2 Theta (deg)intensity12.277.2113830.48.1010.9737207.9811.11261567.4311.911537.267.1112.41193727.0312.6848.5926.3613.910904.926.1414.42989645.9215.01067.7925.8915.012243.445.5016.1312882.85.4416.315.294565.3016.741496.45.2516.940316.44.9917.8277.16564.9517.9312203.24.8718.246.220724.8518.3370604.84.8318.423.83964.7318.76726.884.5919.3394.77044.4120.1144141.64.3120.641681.844.2920.76212.164.2620.84361564.2421.09222644.2321.027615.844.2121.13280.1364.1021.75521.044.0921.73456304.0521.9378.93443.9922.32496.7043.9322.6214.12723.8523.173189.283.8223.312258643.8123.353887.63.8023.49882.83.7223.916135843.6924.1105475.23.6724.2399020.83.6224.631706.723.6024.75386643.5824.8322.59043.5725.094490.43.5625.0133301.23.5225.3451.14323.4925.52796.7843.4725.7646.19763.4625.728358.43.3926.321790.883.3826.32128183.3826.43802683.3726.4179711.23.3726.4161576.83.3126.9191729.63.3027.0435268.83.2827.1260997.63.2527.517226.323.2327.62.3899043.2327.69942.83.2227.712246.563.1828.1349707.23.1728.1129469.23.1528.3339687.23.1528.314438.723.1428.43742843.1128.7339841.63.0729.12601.763.0729.117122.83.0429.3114762.43.0429.3102896.83.0429.4103504.83.0329.419609.243.0229.5178769.63.0129.73428.3682.9830.061798.32 Example 4: Crystalline N—N-diisopropyl-tryptamine (DiPT) hydrofumarate Preparation: Single crystals of DiPT hydrofumarate suitable for X-ray diffraction analysis were obtained from the slow evaporation of a methanol/isopropanol solution of a commercial sample of DiPT hydrofumarate (ChemLogix). Single Crystal Characterization: Crystal data, data collection and structure refinement details are summarized in Table 9. TABLE 9DiPT hydrofumarateChemical formulaC4H3O4•C16H25N2Mr360.44Crystal system, space groupMonoclinic, P21/cTemperature (K)297a, b, c (Å)9.7954 (5), 13.6386 (6), 14.8273 (7)β (°)101 (2)V (Å3)1944.81 (16)Z4Radiation typeMo Kαμ (mm−1)0.09Crystal size (mm)0.27 × 0.22 × 0.21DiffractometerBruker APEX-II CCDAbsorption correctionMulti-scanSADABS2016/2 (Bruker, 2016 February) was used forabsorption correction. wR2(int) was 0.0555 before and0.0499 after correction. The Ratio of minimum tomaximum transmission is 0.9552. The λ/2 correctionfactor is Not present.Tmin, Tmax0.712, 0.745No. of measured,53570, 3531, 2885independent andobserved [I > 2σ(I)]reflectionsRint0.044R[F2> 2σ(F2)], WR(F2), S0.040, 0.104, 1.05No. of reflections3531No. of parameters272No. of restraints42H-atom treatmentH atoms treated by a mixture of independent andconstrained refinementΔmax, Δmin(e Å−3)0.17, −0.14Data collection: Bruker APEX3; cell refinement: Bruker SAINT; data reduction: Bruker SAINT; program(s) used to solve structure: SHELXS97 (Sheldrick 2008); program(s) used to refine structure: SHELXL 2018 March (Sheldrick, 2015); molecular graphics: Olex2 1.3 (Dolomanov et al., 2009); software used to prepare material for publication: Olex2 1.3 (Dolomanov et al., 2009). TABLE 10d-spacing2Theta (deg)Intensity9.958.93382.9049.629.23565.287.8611.240.64887.4111.91651.8287.2812.11941.4686.8213.0172.44366.5113.69262.926.4213.811601.726.4213.8122.79886.1814.32056.2845.8115.24652.65.5615.910369.245.4016.4492285.3416.63577.845.0217.722182.724.9817.827775.724.9717.89396.764.8118.454127.84.6719.0510084.5719.422565.124.5719.44120.764.5319.6468024.4519.910349.844.4220.1770.3384.3420.411266.844.2021.1288.36044.2021.14781.244.1321.55352.884.1121.65895.284.0422.018942.643.9522.5156.52243.9522.59694.43.9322.611565.363.8722.9725.2483.8722.9504203.8723.04874.283.8623.018405.083.7124.068049.63.7124.028430.443.7024.03944.863.6624.310979.163.6524.320738.83.6424.44366.063.6424.41285.4583.5724.99.031163.5225.310309.563.5225.31113.6843.4725.6570.5963.4625.76687.523.4126.12203.923.3226.837.225363.3226.952.6763.3226.923237.083.3226.922812.483.3027.0115.94683.2727.340811.63.2527.420670.323.2127.712939.843.2127.87986.963.2127.8138013.2127.83110.543.2127.816023.83.2127.858745.23.1828.07332.283.1728.111668.923.1728.13724.9843.1628.25747.783.1328.55139.083.1228.521870.563.1228.65422.363.1228.625361.763.1028.83668.0643.0928.92589.983.0828.91089.963.0529.2890.7323.0229.6190.663.0129.64503.96 REFERENCES Ascic, E., Hansen, C. L., Le Quement, S. T. & Nielsen, T. E. (2012). Chem. Commun., 48, 3345-3347.Blei, F., Dörner, S., Fricke, J., Baldeweg, F., Trottmann, F., Komor, A., Meyer, F., Hertweck, C. & Hoffmeister, D. (2020). Chem. Eur. J., 26, 729-734.Brandt, S. D., Freeman, S., Fleet, I. A. & Alder, J. F. (2005b). Analyst, 130, 1258-1262.Brandt, S. D., Freeman, S., Fleet, I. A., McGagh, P. & Alder, J. F. (2005a). Analyst, 130, 330-344.Bruker (2018). APEX3, SAINT, and SADABS. Bruker AXS Inc., Madison, Wisconsin, USA.Cameron, L P. & Olson, D. E. (2018). ACS Chem. Neurosci., 9, 2344-2357.Dolomanov, O. V., Bourhis, L J., Gildea, R. J., Howard, J. A. K. & Puschmann, H. (2009). J. Appl. Cryst. 42, 339-341.Jimenez-Garrido, D. F., G6mez-Sousa, M., Ona, G., Dos Santos, R. G., Hallak, J. E. C., Alcdzar-C6rcoles, M. A. & Bouso, J. C. (2020). Sci. Rep. 10, 4075.Johnson, M. W., Hendricks, P. S., Barrett, F. S. & Griffiths, R. R. (2019). Pharmacol. Therapeut. 197, 83-102.Sheldrick, G. M. (2015a). Acta Cryst. A71, 3-8.Sheldrick, G. M. (2015b). Acta Cryst. C71, 3-8.Sherwood, A. M., Halberstadt, A. L., Klein, A. K., McCorvy, J. D., Kaylo, K. W., Kargbo, R. B. & Meisenheimer, P. (2020). J. Nat. Prod. 83, 461-467.Shulgin, A. T. & Shulgin, A. (2016). TiKHAL: The Continuation. Isomerdesign. Available at:http://isomerdesign.com/PiHKAL/read.php?domain=tk&id=56. Accessed 19 Mar. 2020.
65,962
11858897
DETAILED DESCRIPTION Compounds of the Application A first aspect of the application relates to a compound of Formula (I): or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, wherein:each R1is independently (C1-C6) alkyl, (C1-C6) haloalkyl, (C1-C6) alkoxy, (C1-C6) haloalkoxy, halogen, NO2, NH2, OH, or CN;R2is H or (C1-C3) alkyl;R3is H, (C1-C6) alkyl, or (C1-C6) haloalkyl;R4is H, (C1-C6) alkyl, or (C1-C6) haloalkyl;each R5is independently (C1-C6) alkyl, (C1-C6) haloalkyl, (C1-C6) alkoxy, (C1-C6) haloalkoxy, halogen, NO2, NH2, OH, or CN;R6is CN, COOH, N((C1-C6) alkyl)-(CH2)1-4—N((C1-C6) alkyl)2, (C1-C6) alkyl substituted with at least one OH, (C2-C6) alkenyl, C6-C10aryl, heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, or heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the (C2-C6) alkenyl, aryl, heteroaryl, and heterocyclyl are each optionally substituted with one or more Q-T;Q is a bond or (C1-C6) alkyl linker;T is (C1-C6) alkyl, (C1-C6) alkylamino, di(C1-C6) alkylamino, amino, aminocarbonyl, (C1-C6) alkylaminocarbonyl, di(C1-C6) alkylaminocarbonyl, OH, S(O)qF, or heterocyclyl comprising a 5 or 6-membered ring and 1-3 heteroatoms selected from N, O and S, wherein when R6is (C2-C6) alkenyl, T is not (C1-C6) alkyl;q is 1 or 2;m is 0, 1, 2, or 3; andn is 1, 2, or 3. (1a) In some embodiments of Formula (I), m is 0. (1b) In some embodiments of Formula (I), m is 1. (1c) In some embodiments of Formula (I), m is 2. (1d) In some embodiments of Formula (I), m is 3. (2a) In some embodiments of Formula (I), at least one R1is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl). (2b) In some embodiments of Formula (I), at least one R1is (C1-C6) haloalkyl (e.g., CH2C1, CHCl2, CCl3, CH2F, CHF2, or CF3). (2c) In some embodiments of Formula (I), at least one R1is (C1-C6) alkoxy (e.g., methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, t-butoxy, pentoxy, or hexyloxy). (2d) In some embodiments of Formula (I), at least one R1is (C1-C6) haloalkoxy (e.g., C1CH2O, FCH2O, Cl2CHO, F2CHO, Cl3CO, or F3CO). (2e) In some embodiments of Formula (I), at least one R1is halogen (e.g., F, C1, Br, or I), NO2, NH2, OH, or CN. (3a) In some embodiments of Formula (I), R2is H. (3b) In some embodiments of Formula (I), R2is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). (4a) In some embodiments of Formula (I), R3is H. (4b) In some embodiments of Formula (I), R3is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl) or (C1-C6) haloalkyl (e.g., CH2C1, CHCl2, CCl3, CH2F, CHF2, or CF3). (4c) In some embodiments of Formula (I), R3is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl). (4d) In some embodiments of Formula (I), R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R3is methyl. (5a) In some embodiments of Formula (I), R4is H. (5b) In some embodiments of Formula (I), R4is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl) or (C1-C6) haloalkyl (e.g., CH2C1, CHCl2, CCl3, CH2F, CHF2, or CF3). (5c) In some embodiments of Formula (I), R4is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl). (5d) In some embodiments of Formula (I), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R4is methyl. (6a) In some embodiments of Formula (I), R3and R4are each (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl). (6b) In some embodiments of Formula (I), R3and R4are each (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R3and R4are each methyl. (7a) In some embodiments of Formula (I), n is 1. (7b) In some embodiments of Formula (I), n is 2. (7c) In some embodiments of Formula (I), n is 3. (8a) In some embodiments of Formula (I), at least one R5is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl). (8b) In some embodiments of Formula (I), at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, at least one R5is methyl. In other further embodiments, at least one R5is ethyl. In other further embodiments at least one R5is propyl. (8c) In some embodiments of Formula (I), at least one R5is (C1-C6) haloalkyl (e.g., CH2C1, CHCl2, CCl3, CH2F, CHF2, or CF3). (8d) In some embodiments of Formula (I), at least one R5is (C1-C6) alkoxy (e.g., methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, t-butoxy, pentoxy, or hexyloxy). (8e) In some embodiments of Formula (I), at least one R5is (C1-C6) haloalkoxy (e.g., C1CH2O, FCH2O, C12CHO, F2CHO, C13CO, or F3CO). (8f) In some embodiments of Formula (I), at least one R5is halogen (e.g., F, C1, Br, or I), NO2, NH2, OH, or CN. (8g) In some embodiments of Formula (I), n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), and the remaining R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), (C1-C6) haloalkyl (e.g., CH2C1, CHCl2, CCl3, CH2F, CHF2, or CF3), (C1-C6) alkoxy (e.g., methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, t-butoxy, pentoxy, or hexyloxy), (C1-C6) haloalkoxy (e.g., C1CH2O, FCH2O, C12CHO, F2CHO, C13CO, or F3CO), halogen (e.g., F, C1, Br, or I), NO2, NH2, OH, or CN. In further embodiments, n is 2 or 3, and at least one R5is methyl or ethyl, and the remaining R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), (C1-C6) haloalkyl (e.g., CH2C1, CHCl2, CCl3, CH2F, CHF2, or CF3), (C1-C6) alkoxy (e.g., methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, t-butoxy, pentoxy, or hexyloxy), (C1-C6) haloalkoxy (e.g., C1CH2O, FCH2O, C12CHO, F2CHO, Cl3CO, or F3CO), halogen (e.g., F, C1, Br, or I), NO2, NH2, OH, or CN. In other further embodiments, n is 2 or 3, and at least one R5ethyl, and the remaining R5is (C1-C6) haloalkyl (e.g., CH2C1, CHCl2, CCl3, CH2F, CHF2, or CF3), (C1-C6) alkoxy (e.g., methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, t-butoxy, pentoxy, or hexyloxy), (C1-C6) haloalkoxy (e.g., C1CH2O, FCH2O, C12CHO, F2CHO, C13CO, or F3CO), halogen (e.g., F, C1, Br, or I), NO2, NH2, OH, or CN. (9) In some embodiments of Formula (I), R6is CN, COOH, N((C1-C6) alkyl)-(CH2)1-4—N4C1-C6) alkyl)2, (C1-C6) alkyl substituted with at least one OH, (C2-C6) alkenyl, heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, or heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the (C2-C6) alkenyl, aryl, heteroaryl, and heterocyclyl are each optionally substituted with one or more Q-T. (9a) In some embodiments of Formula (I), R6is CN. (9b) In some embodiments of Formula (I), R6is COOH. (9c) In some embodiments of Formula (I), R6is N((C1-C6) alkyl)-(CH2)1-4N((C1-C6) alkyl)2, wherein the (C1-C6) alkyl is selected from methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, and hexyl. In further embodiments, R6is N((C1-C3) alkyl)-(CH2)1-4—N((C1-C3) alkyl)2, wherein the (C1-C3) alkyl is selected from methyl, ethyl, n-propyl, and i-propyl. In further embodiments, R6is N((C1-C3) alkyl)-(CH2)1-2—N((C1-C3) alkyl)2, wherein the (C1-C3) alkyl is selected from methyl, ethyl, n-propyl, and i-propyl. In further embodiments, R6is N(CH3)CH2CH2N(CH3)2. (9d) In some embodiments of Formula (I), R6is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl) substituted with at least one OH (e.g., one OH, two OH, or three OH). In further embodiments, R6is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl) substituted with at least one OH (e.g., one OH, two OH, or three OH). In further embodiments, R6is ethyl substituted with at least one OH. In further embodiments, R6is 1,2-dihydroxyethyl. (9e) In some embodiments of Formula (I), R6is (C2-C6) alkenyl (e.g., ethenyl, propenyl, butenyl, pentenyl, or hexenyl), optionally substituted with one or more Q-T. (9f) In some embodiments of Formula (I), R6is heteroaryl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, etc.), optionally substituted with one or more Q-T. In further embodiments, R6is heteroaryl comprising a 5-membered ring and at least one nitrogen atom (e.g., pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, etc.), optionally substituted with one or more Q-T. In further embodiments, R6is furan-3-yl, thiophen-2-yl, pyrazol-3-yl, pyrazol-4-yl, isoxazol-4-yl, or 1,2,3-triazol-4-yl, optionally substituted with one or more Q-T. In further embodiments, R6is pyrazol-4-yl, optionally substituted with one or more Q-T. (9g) In some embodiments of Formula (I), R6is a heteroaryl comprising a 6-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyranyl, thiopyranyl, diazinyl, thiazinyl, dioxinyl, triazinyl, etc.), optionally substituted with one or more Q-T. In further embodiments, R6is heteroaryl comprising a 6-membered ring and at least one nitrogen atom (e.g., pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, diazinyl, thiazinyl, triazinyl, etc.), optionally substituted with one or more Q-T. In further embodiments, R6is pyridin-4-yl, pyridin-3-yl, or pyrimidin-5-yl, optionally substituted with one or more Q-T. (9h) In some embodiments of Formula (I), R6is heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, triazolidinyl, oxadiazolidinyl, isoxadiazolidinyl, thiadiazolidinyl, isothiadiazolidinyl, etc.), optionally substituted with one or more Q-T. In some embodiments of Formula (I), R6is heterocyclyl comprising a 5-membered ring and at least one nitrogen atom (e.g., pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, triazolidinyl, oxadiazolidinyl, isoxadiazolidinyl, thiadiazolidinyl, isothiadiazolidinyl, etc.), optionally substituted with one or more Q-T. (9i) In some embodiments of Formula (I), R6is CN, COOH, N((C1-C6) alkyl)-(CH2)1-4—N((C1-C6) alkyl)2, (C1-C6) alkyl substituted with at least one OH, heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, or heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the heteroaryl and heterocyclyl are each optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. (9j) In some embodiments of Formula (I), R6is heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, or heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the heteroaryl and heterocyclyl are each optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. (9k) In some embodiments of Formula (I), R6is heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the heteroaryl is optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. (9l) In some embodiments of Formula (I), R6is C6-C10aryl optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. In some embodiments of Formula (I), R6is C6-C10aryl optionally substituted with one or more Q-T, wherein T is S(O)qF. In some embodiments of Formula (I), R6is C6-C10aryl optionally substituted with one or more Q-T, wherein T is S(O)2F. (9m) In some embodiments of Formula (I), R6is phenyl optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. In some embodiments of Formula (I), R6is phenyl optionally substituted with one or more Q-T, wherein T is S(O)qF. In some embodiments of Formula (I), R6is phenyl optionally substituted with one or more Q-T, wherein T is S(O)2F. (10a) In some embodiments of Formula (I), Q is a bond. (10b) In some embodiments of Formula (I), Q is a (C1-C6) alkyl linker (e.g., methyl linker (—CH2—), ethyl linker (—CH2CH2— or —CH(CH3)—), propyl linker (—CH2CH2CH2—, —CH(CH3)CH2—, or —C(CH3)2—), butyl linker (—CH2CH2CH2CH2—, —CH(CH3)CH2CH2—, —CH2CH(CH3)CH2—, —C(CH3) 2CH2—, or —CH(CH3)CH(CH3)—), pentyl linker (—CH2CH2CH2CH2CH2—, —CH(CH3)CH2CH2CH2—, —CH2CH(CH3)CH2CH2—, —C(CH3)2CH2CH2—, or —CH2C(CH3)2CH2—), or hexyl linker (—CH2CH2CH2CH2CH2CH2—). In further embodiments, Q is a (C1-C3) alkyl linker (e.g., methyl linker (—CH2—), ethyl linker (—CH2CH2— or —CH(CH3)—), or propyl linker (—CH2CH2CH2—, —CH(CH3)CH2—, or —C(CH3)2—)). In further embodiments, Q is —CH2—, —CH2CH2—, —CH2CH2CH2—, or —C(CH3)2—. (11) In some embodiments of Formula (I), T is (C1-C6) alkyl, (C1-C6) alkylamino, di(C1-C6) alkylamino, amino, aminocarbonyl, (C1-C6) alkylaminocarbonyl, di(C1-C6) alkylaminocarbonyl, OH, or heterocyclyl comprising a 5 or 6-membered ring and 1-3 heteroatoms selected from N, O and S, wherein when R6is (C2-C6) alkenyl, T is not (C1-C6) alkyl. (11a) In some embodiments of Formula (I), T is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl). In further embodiments, T is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, T is methyl. (11b) In some embodiments of Formula (I), T is (C1-C6) alkylamino (e.g., methylamino, ethylamino, propylamino, butylamino, pentylamino, or hexylamino). (11c) In some embodiments of Formula (I), T is di(C1-C6) alkylamino (e.g., dimethylamino, diethylamino, dipropylamino, dibutylamino, dipentylamino, or dihexylamino). In further embodiments, T is di(C1-C3) alkylamino (e.g., dimethylamino, diethylamino, or dipropylamino). In further embodiments, T is dimethylamino. (11d) In some embodiments of Formula (I), T is amino. (11e) In some embodiments of Formula (I), T is aminocarbonyl (i.e., NH2C(O)). (11f) In some embodiments of Formula (I), T is (C1-C6) alkylaminocarbonyl (e.g., methylaminocarbonyl, ethylaminocarbonyl, propylaminocarbonyl, butylaminocarbonyl, pentylaminocarbonyl, or hexylaminocarbonyl). In further embodiments, T is methylaminocarbonyl (i.e., CH3NHC(O)). (11g) In some embodiments of Formula (I), T is di (C1-C6) alkylaminocarbonyl (e.g., dimethylaminocarbonyl, diethylaminocarbonyl, dipropylaminocarbonyl, dibutylaminocarbonyl, dipentylaminocarbonyl, or dihexylaminocarbonyl). In further embodiments, T is dimethylaminocarbonyl (i.e., (CH3)2NC(O)). (11h) In some embodiments of Formula (I), T is OH. (11i) In some embodiments of Formula (I), T is heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, triazolidinyl, oxadiazolidinyl, isoxadiazolidinyl, thiadiazolidinyl, isothiadiazolidinyl, etc.). (11j) In some embodiments of Formula (I), T is heterocyclyl comprising a 6-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., piperidinyl, tetrahydropyranyl, piperazinyl, morpholinyl, thiomorpholinyl, dioxanyl, etc.). In further embodiments, T is morpholinyl. In other further embodiments, T is tetrahydropyranyl. In other further embodiments, T is piperidinyl. (11k) In some embodiments of Formula (I), T is S(O)F. In some embodiments of Formula (I), T is S(O)2F. In some embodiments of Formula (I), each of the substituents defined for any one of R1, R2, R3, R4, R5, R6, Q, T, q, m, and n can be combined with any of the substituents defined for the remainder of R1, R2, R3, R4, R5, R6, Q, T, q, m, and n. (12) In some embodiments, m is 0, and R2is H. (13) In some embodiments, m is 0, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), and R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, and R3and R4are each methyl. (14) In some embodiments, R2is H, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), and R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R2is H, and R3and R4are each methyl. (15) In some embodiments, m is 0, R2is H, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), and R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, R2is H, and R3and R4are each methyl. (16) In some embodiments, m is 0, and n is 1. (17) In some embodiments, R2is H, and n is 1. (18) In some embodiments, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), and n is 1. In further embodiments, R3and R4are each methyl, and n is 1. (19) In some embodiments, m is 0, R2is H, and n is 1. (20) In some embodiments, m is 0, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), and n is 1. In further embodiments, m is 0, R3and R4are each methyl, and n is 1. (21) In some embodiments, R2is H, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), and n is 1. In further embodiments, R2is H, R3and R4are each methyl, and n is 1. (22) In some embodiments, m is 0, R2is H, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), and n is 1. In further embodiments, m is 0, R2is H, R3and R4are each methyl, and n is 1. (23) In some embodiments, n is 1, and R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, n is 1, and R5is ethyl. (24) In some embodiments, m is 0, n is 1, and R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, n is 1, and R5is ethyl. (25) In some embodiments, R2is H, n is 1, and R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R2is H, n is 1, and R5is ethyl. (26) In some embodiments, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), n is 1, and R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R3and R4are each methyl, n is 1, and R5is ethyl. (27) In some embodiments, m is 0, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), n is 1, and R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, R3and R4are each methyl, n is 1, and R5is ethyl. (28) In some embodiments, R2is H, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), n is 1, and R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R2is H, R3and R4are each methyl, n is 1, and R5is ethyl. (29) In some embodiments, m is 0, R2is H, n is 1, and R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, R2is H, n is 1, and R5is ethyl. (30) In some embodiments, m is 0, R2is H, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), n is 1, and R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, R2is H, R3and R4are each methyl, n is 1, and R5is ethyl. (31) In some embodiments, n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, n is 2 or 3, and at least one R5is ethyl. (32) In some embodiments, m is 0, n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, n is 2 or 3, and at least one R5is ethyl. (33) In some embodiments, R2is H, n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R2is H, n is 2 or 3, and at least one R5is ethyl. (34) In some embodiments, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R3and R4are each methyl, n is 2 or 3, and at least one R5is ethyl. (35) In some embodiments, m is 0, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, R3and R4are each methyl, n is 2 or 3, and at least one R5is ethyl. (36) In some embodiments, R2is H, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R2is H, R3and R4are each methyl, n is 2 or 3, and at least one R5is ethyl. (37) In some embodiments, m is 0, R2is H, n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, R2is H, n is 2 or 3, and at least one R5is ethyl. (38) In some embodiments, m is 0, R2is H, R3is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), R4is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl), n is 2 or 3, and at least one R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, m is 0, R2is H, R3and R4are each methyl, n is 2 or 3, and at least one R5is ethyl. (39) In some embodiments, R1, R2, R3, R4, R5, q, m, and n are each as described in any one of (12)-(38), and R6is as described in any one of (9)-(9m). (40) In some embodiments, R1, R2, R3, R4, R5, q, m, and n are each as described in any one of (12)-(38), and R6is as described in any one of (9a)-(9k). (41) In some embodiments, R1, R2, R3, R4, R5, q, m, and n are each as described in any one of (12)-(38), and R6is as described in any one of (9i)-(9k). (42) In some embodiments, R1, R2, R3, R4, R5, q, m, and n are each as described in any one of (12)-(38), and R6is as described in any one of (9f)-(9h). (43) In some embodiments, R1, R2, R3, R4, R5, q, m, and n are each as described in any one of (12)-(38), and R6is as described in (90. (44) In some embodiments, R1, R2, R3, R4, R5, q, m, and n are each as described in any one of (12)-(38), and R6is as described in (9l) or (9m). (45) In some embodiments, R1, R2, R3, R4, R5, q, m, and n are each as described in any one of (12)-(38), and R6is as described in (9m). In some embodiments, the compound of Formula (I) has the structure of Formula (II): or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, wherein n1 is 0, 1, or 2, and R1, R2, R3, R4, R5, R6, Q, T, and m are each as defined herein above in Formula (I). In some embodiments of Formula (II), n1 is 0. In some embodiments of Formula (II), n1 is 1. In some embodiments of Formula (II), n1 is 2. In some embodiments of Formula (II), each of R1, R2, R3, R4, R5, R6, Q, T, q, and m can be selected from the substituent groups described above in Formula (I). In some embodiments of Formula (II), each of the substituents defined for any one of R1, R2, R3, R4, R5, R6, Q, T, q, m, and n1 can be combined with any of the substituents defined for the remainder of R1, R2, R3, R4, R5, R6, Q, T, q, m, and n1. In some embodiments, the compound of Formula (I) has the structure of Formula (IIa) or (IIb): or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, wherein R5and R6are each as defined herein above in Formula (I). In some embodiments of Formula (IIa), R5is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl). In some embodiments of Formula (IIa), R5is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, R5is methyl. In other further embodiments, R5is ethyl. In other further embodiments R5is propyl. In some embodiments of Formula (IIa), R5is ethyl. In some embodiments of Formula (IIa), R5is (C1-C6) haloalkyl (e.g., CH2C1, CHCl2, CCl3, CH2F, CHF2, or CF3). In some embodiments of Formula (IIa), R5is (C1-C6) alkoxy (e.g., methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, t-butoxy, pentoxy, or hexyloxy). In some embodiments of Formula (IIa), R5is (C1-C6) haloalkoxy (e.g., C1CH2O, FCH2O, C12CHO, F2CHO, C13CO, or F3CO). In some embodiments of Formula (IIa), R5is halogen (e.g., F, C1, Br, or I), NO2, NH2, OH, or CN. In some embodiments of Formula (IIa) or (IIb), R6is CN. In some embodiments of Formula (IIa) or (IIb), R6is COOH. In some embodiments of Formula (IIa) or (IIb), R6is N((C1-C6) alkyl)-(CH2)1-4—N((C1-C6) alkyl)2, wherein the (C1-C6) alkyl is selected from methyl, ethyl, n-propyl, propyl, n-butyl, i-butyl, t-butyl, pentyl, and hexyl. In further embodiments, R6is N((C1-C3) alkyl)-(CH2)1-4—N((C1-C3) alkyl)2, wherein the (C1-C3) alkyl is selected from methyl, ethyl, n-propyl, and i-propyl. In further embodiments, R6is N((C1-C3) alkyl)-(CH2)1-2—N((C1-C3) alkyl)2, wherein the (C1-C3) alkyl is selected from methyl, ethyl, n-propyl, and i-propyl. In further embodiments, R6is N(CH3)CH2CH2N(CH3)2. In some embodiments of Formula (IIa) or (IIb), R6is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl) substituted with at least one OH (e.g., one OH, two OH, or three OH). In further embodiments, R6is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl) substituted with at least one OH (e.g., one OH, two OH, or three OH). In further embodiments, R6is ethyl substituted with at least one OH. In further embodiments, R6is 1,2-dihydroxyethyl. In some embodiments of Formula (IIa) or (IIb), R6is (C2-C6) alkenyl (e.g., ethenyl, propenyl, butenyl, pentenyl, or hexenyl), optionally substituted with one or more Q-T. In some embodiments of Formula (IIa) or (IIb), R6is heteroaryl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, etc.), optionally substituted with one or more Q-T. In further embodiments, R6is heteroaryl comprising a 5-membered ring and at least one nitrogen atom (e.g., pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, etc.), optionally substituted with one or more Q-T. In further embodiments, R6is furan-3-yl, thiophen-2-yl, pyrazol-3-yl, pyrazol-4-yl, isoxazol-4-yl, or 1,2,3-triazol-4-yl, optionally substituted with one or more Q-T. In further embodiments, R6is pyrazol-4-yl, optionally substituted with one or more Q-T. In some embodiments of Formula (IIa) or (IIb), R6is a heteroaryl comprising a 6-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyranyl, thiopyranyl, diazinyl, thiazinyl, dioxinyl, triazinyl, etc.), optionally substituted with one or more Q-T. In further embodiments, R6is heteroaryl comprising a 6-membered ring and at least one nitrogen atom (e.g., pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, diazinyl, thiazinyl, triazinyl, etc.), optionally substituted with one or more Q-T. In further embodiments, R6is pyridin-4-yl, pyridin-3-yl, or pyrimidin-5-yl, optionally substituted with one or more Q-T. In some embodiments of Formula (IIa) or (IIb), R6is heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, triazolidinyl, oxadiazolidinyl, isoxadiazolidinyl, thiadiazolidinyl, isothiadiazolidinyl, etc.), optionally substituted with one or more Q-T. In some embodiments of Formula (IIa) or (IIb), R6is heterocyclyl comprising a 5-membered ring and at least one nitrogen atom (e.g., pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, triazolidinyl, oxadiazolidinyl, isoxadiazolidinyl, thiadiazolidinyl, isothiadiazolidinyl, etc.), optionally substituted with one or more Q-T. In some embodiments of Formula (IIa) or (IIb), R6is CN, COOH, N((C1-C6) alkyl)-(CH2)1-4—N((C1-C6) alkyl)2, (C1-C6) alkyl substituted with at least one OH, heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, or heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the heteroaryl and heterocyclyl are each optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. In some embodiments of Formula (IIa) or (IIb), R6is heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, or heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the heteroaryl and heterocyclyl are each optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. In some embodiments of Formula (IIa) or (IIb), R6is heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the heteroaryl is optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. In some embodiments of Formula (IIa) or (IIb), R6is C6-C10aryl optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. In some embodiments of Formula (I), R6is C6-C10aryl optionally substituted with one or more Q-T, wherein T is S(O)qF. In some embodiments of Formula (I), R6is C6-C10aryl optionally substituted with one or more Q-T, wherein T is S(O)2F. In some embodiments of Formula (IIa) or (IIb), R6is phenyl optionally substituted with one or more Q-T, each of which may be selected from the substituents as described herein. In some embodiments of Formula (I), R6is phenyl optionally substituted with one or more Q-T, wherein T is S(O)qF. In some embodiments of Formula (I), R6is phenyl optionally substituted with one or more Q-T, wherein T is S(O)2F. In some embodiments of Formula (IIa) or (IIb), Q is a bond. In some embodiments of Formula (IIa) or (IIb), Q is a (C1-C6) alkyl linker (e.g., methyl linker (—CH2—), ethyl linker (—CH2CH2— or —CH(CH3)—), propyl linker (—CH2CH2CH2—, —CH(CH3)CH2—, or —C(CH3)2—), butyl linker (—CH2CH2CH2CH2—, —CH(CH3)CH2CH2—, —CH2CH(CH3)CH2—, —C(CH3)2CH2—, or —CH(CH3)CH(CH3)—), pentyl linker (—CH2CH2CH2CH2CH2—, —CH(CH3)CH2CH2CH2—, —CH2CH(CH3)CH2CH2—, —C(CH3)2CH2CH2—, or —CH2C(CH3)2CH2—), or hexyl linker (—CH2CH2CH2CH2CH2CH2—). In further embodiments, Q is a (C1-C3) alkyl linker (e.g., methyl linker (—CH2—), ethyl linker (—CH2CH2— or —CH(CH3)—), or propyl linker (—CH2CH2CH2—, —CH(CH3)CH2—, or —C(CH3)2—)). In further embodiments, Q is —CH2—, —CH2CH2—, —CH2CH2CH2—, or —C(CH3)2—. In some embodiments of Formula (IIa) or (IIb), T is (C1-C6) alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, or hexyl). In further embodiments, T is (C1-C3) alkyl (e.g., methyl, ethyl, n-propyl, or i-propyl). In further embodiments, T is methyl. In some embodiments of Formula (IIa) or (IIb), T is (C1-C6) alkylamino (e.g., methylamino, ethylamino, propylamino, butylamino, pentylamino, or hexylamino). In some embodiments of Formula (IIa) or (IIb), T is di(C1-C6) alkylamino (e.g., dimethylamino, diethylamino, dipropylamino, dibutylamino, dipentylamino, or dihexylamino). In further embodiments, T is di(C1-C3) alkylamino (e.g., dimethylamino, diethylamino, or dipropylamino). In further embodiments, T is dimethylamino. In some embodiments of Formula (IIa) or (IIb), T is amino. In some embodiments of Formula (IIa) or (IIb), T is aminocarbonyl (i.e., NH2C(O)). In some embodiments of Formula (IIa) or (IIb), T is (C1-C6) alkylaminocarbonyl (e.g., methylaminocarbonyl, ethylaminocarbonyl, propylaminocarbonyl, butylaminocarbonyl, pentylaminocarbonyl, or hexylaminocarbonyl). In further embodiments, T is methylaminocarbonyl (i.e., CH3NHC(O)). In some embodiments of Formula (IIa) or (IIb), T is di (C1-C6) alkylaminocarbonyl (e.g., dimethylaminocarbonyl, diethylaminocarbonyl, dipropylaminocarbonyl, dibutylaminocarbonyl, dipentylaminocarbonyl, or dihexylaminocarbonyl). In further embodiments, T is dimethylaminocarbonyl (i.e., (CH3)2NC(O)). In some embodiments of Formula (IIa) or (IIb), T is OH. In some embodiments of Formula (IIa) or (IIb), T is heterocyclyl comprising a 5-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, triazolidinyl, oxadiazolidinyl, isoxadiazolidinyl, thiadiazolidinyl, isothiadiazolidinyl, etc.). In some embodiments of Formula (IIa) or (IIb), T is heterocyclyl comprising a 6-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., piperidinyl, tetrahydropyranyl, piperazinyl, morpholinyl, thiomorpholinyl, dioxanyl, etc.). In further embodiments, T is morpholinyl. In other further embodiments, T is tetrahydropyranyl. In other further embodiments, T is piperidinyl. In some embodiments of Formula (IIa) or (IIb), T is S(O)F. In some embodiments of Formula (IIa) or (IIb), T is S(O)2F. In some embodiments of Formula (IIa) or (IIb), each of the substituents defined for any one of R5, R6, Q, T, and q can be combined with any of the substituents defined for the remainder of R5, R6, Q, T, and q. Another aspect of the application relates to a compound of Formula (Ia): or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for inhibiting a mutant ALK (e.g., ALK G1202R); treating or preventing a disease or disorder (e.g., cancer) in which a mutant ALK (e.g., ALK G1202R) plays a role; treating or preventing cancer in a subject identified as being in need of inhibition of a mutant ALK (e.g., ALK G1202R) for the treatment or prevention of the cancer; treating or preventing a disease or disorder (e.g., cancer) resistant to an ALK targeted therapy, such as a therapy with Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922; treating or preventing cancer, wherein the cancer cell comprises the mutant ALK (e.g., ALK G1202R); inhibiting SRPK (e.g., SRPK1 and/or SRPK2); regulating (e.g., inhibiting) VEGF mediated angiogenesis; treating or preventing a disease or disorder in which a VEGF mediated angiogenesis plays a role (e.g., AMD or angiogenesis-dependent cancers); treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD); or treating or preventing an angiogenesis-dependent cancer (e.g., tumorous cancer) (e.g., in a subject identified in need of regulation of VEGF mediated angiogenesis for the treatment or prevention of an angiogenesis-dependent cancer), wherein: each R1is independently (C1-C6) alkyl, (C1-C6) haloalkyl, (C1-C6) alkoxy, (C1-C6) haloalkoxy, halogen, NO2, NH2, OH, or CN; R2is H or (C1-C3) alkyl; R3is H, (C1-C6) alkyl, or (C1-C6) haloalkyl; R4is H, (C1-C6) alkyl, or (C1-C6) haloalkyl; each R5is independently (C1-C6) alkyl, (C1-C6) haloalkyl, (C1-C6) alkoxy, (C1-C6) haloalkoxy, halogen, NO2, NH2, OH, or CN; R6′ is CN, COOH, N((C1-C6) alkyl)-(CH2)1-4—N((C1-C6) alkyl)2, (C1-C6) alkyl substituted with at least one OH, (C2-C6) alkenyl, C6-C10aryl, heteroaryl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, or heterocyclyl comprising a 5- or 6-membered ring and 1-3 heteroatoms selected from N, O and S, wherein the (C2-C6) alkenyl, aryl, heteroaryl, and heterocyclyl are each optionally substituted with one or more Q-T; Q is a bond or (C1-C6) alkyl linker; T is (C1-C6) alkyl, (C1-C6) alkylamino, di(C1-C6) alkylamino, amino, aminocarbonyl, (C1-C6) alkylaminocarbonyl, di(C1-C6) alkylaminocarbonyl, OH, S(O)qF, or heterocyclyl comprising a 5 or 6-membered ring and 1-3 heteroatoms selected from N, O and S, wherein when R6′ is (C2-C6) alkenyl, T is not (C1-C6) alkyl; q is 1 or 2; m is 0, 1, 2, or 3; and n is 1, 2, or 3. In some embodiments of Formula (Ia), each of R1, R2, R3, R4, R5, Q, T, m, and n can be selected from the substituents as defined for R1, R2, R3, R4, R5, Q, T, m, and n in Formula (I). In some embodiments of Formula (Ia), m is as defined in (1a)-(1d) of Formula (I). In some embodiments of Formula (Ia), R1is as defined in (2a)-(2e) of Formula (I). In some embodiments of Formula (Ia), R2is as defined in (3a)-(3b) of Formula (I). In some embodiments of Formula (Ia), R3is as defined in (4a)-(4d) and (6a)-(6b) of Formula (I). In some embodiments of Formula (Ia), R4is as defined in (5a)-(5d) and (6a)-(6b) of Formula (I). In some embodiments of Formula (Ia), n is as defined in (7a)-(7c) and (8g) of Formula (I). In some embodiments of Formula (Ia), R5is as defined in (8a)-(8g) of Formula (I). In some embodiments of Formula (Ia), Q is as defined in (10a)-(10b) of Formula (I). In some embodiments of Formula (Ia), T is as defined in (11)-(11m) of Formula (I). In some embodiments of Formula (Ia), R6′ can be selected from the substituents as defined in (9)-(9m) for R6in Formula (I). In some embodiments of Formula (Ia), R6′ is heterocyclyl comprising a 6-membered ring and 1-3 heteroatoms selected from N, O and S (e.g., piperidinyl, tetrahydropyranyl, piperazinyl, morpholinyl, thiomorpholinyl, dioxanyl, etc.), optionally substituted with one or more Q-T. In some embodiments of Formula (Ia), R6′ is heterocyclyl comprising a 5-membered ring and at least one nitrogen atom (e.g., piperidinyl, piperazinyl, morpholinyl, etc.), optionally substituted with one or more Q-T. In some embodiments of Formula (Ia), each of the substituents defined for any one of R1, R2, R3, R4, R5, R6′, Q, T, q, m, and n can be combined with any of the substituents defined for the remainder of R1, R2, R3, R4, R5, R6′, Q, T, q, m, and n. In some embodiments of Formula (Ia), R1, R2, R3, R4, R5, q, m, and n can be combined as described in (12)-(38) in Formula (I), and R6′ is as described above. In some embodiments of Formula (Ia), R1, R2, R3, R4, R5, R6′, Q, T, q, m, and n can be combined as described above. In some embodiments, a compound of Formula (Ia) is for inhibting SRPK (e.g., SRPK1 and/or SRPK2), regulating (e.g., inhibiting) VEGF mediated angiogenesis, or treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD). Another aspect of the application relates to a method of inhibiting a mutant ALK (e.g., ALK G1202R); treating or preventing a disease or disorder (e.g., cancer) in which a mutant ALK (e.g., ALK G1202R) plays a role; treating or preventing cancer in a subject identified as being in need of inhibition of a mutant ALK (e.g., ALK G1202R) for the treatment or prevention of the cancer; treating or preventing a disease or disorder (e.g., cancer) resistant to an ALK targeted therapy, such as a therapy with Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922; treating or preventing cancer, wherein the cancer cell comprises the mutant ALK (e.g., ALK G1202R); inhibiting SRPK (e.g., SRPK1 and/or SRPK2); regulating (e.g., inhibiting) VEGF mediated angiogenesis; treating or preventing a disease or disorder in which a VEGF mediated angiogenesis plays a role (e.g., AMD or angiogenesis-dependent cancers); treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD); or treating or preventing an angiogenesis-dependent cancer (e.g., tumorous cancer) (e.g., in a subject identified in need of regulation of VEGF mediated angiogenesis for the treatment or prevention of an angiogenesis-dependent cancer), comprising administering to a subject in need thereof an effective amount of a compound of Formula (Ia), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In some embodiments, the application relates to a method of inhibiting SRPK (e.g., SRPK1 and/or SRPK2), regulating (e.g., inhibiting) VEGF mediated angiogenesis, or treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD). In some embodiments, the application relates to a compound of Formula (Ia), of the following structure: or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for inhibiting a mutant ALK (e.g., ALK G1202R); treating or preventing a disease or disorder (e.g., cancer) in which a mutant ALK (e.g., ALK G1202R) plays a role; treating or preventing cancer in a subject identified as being in need of inhibition of a mutant ALK (e.g., ALK G1202R) for the treatment or prevention of the cancer; treating or preventing a disease or disorder (e.g., cancer) resistant to an ALK targeted therapy, such as a therapy with Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922; treating or preventing cancer, wherein the cancer cell comprises the mutant ALK (e.g., ALK G1202R); inhibiting SRPK (e.g., SRPK1 and/or SRPK2); regulating (e.g., inhibiting) VEGF mediated angiogenesis; treating or preventing a disease or disorder in which a VEGF mediated angiogenesis plays a role (e.g., AMD or angiogenesis-dependent cancers); treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD); or treating or preventing an angiogenesis-dependent cancer (e.g., tumorous cancer) (e.g., in a subject identified in need of regulation of VEGF mediated angiogenesis for the treatment or prevention of an angiogenesis-dependent cancer). In some embodiments, the application relates to a compound of Formula (Ia), of the following structure: or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for inhibiting SRPK (e.g., SRPK1 and/or SRPK2), regulating (e.g., inhibiting) VEGF mediated angiogenesis, or treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD). Another aspect of the application relates to a method of inhibiting a mutant ALK (e.g., ALK G1202R); treating or preventing a disease or disorder (e.g., cancer) in which a mutant ALK (e.g., ALK G1202R) plays a role; treating or preventing cancer in a subject identified as being in need of inhibition of a mutant ALK (e.g., ALK G1202R) for the treatment or prevention of the cancer; treating or preventing a disease or disorder (e.g., cancer) resistant to an ALK targeted therapy, such as a therapy with Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922; treating or preventing cancer, wherein the cancer cell comprises the mutant ALK (e.g., ALK G1202R); inhibiting SRPK (e.g., SRPK1 and/or SRPK2); regulating (e.g., inhibiting) VEGF mediated angiogenesis; treating or preventing a disease or disorder in which a VEGF mediated angiogenesis plays a role (e.g., AMD or angiogenesis-dependent cancers); treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD); or treating or preventing an angiogenesis-dependent cancer (e.g., tumorous cancer) (e.g., in a subject identified in need of regulation of VEGF mediated angiogenesis for the treatment or prevention of an angiogenesis-dependent cancer), comprising administering to a subject in need thereof an effective amount of a compound of the following structure: or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In some embodiments, the application relates to a method of inhibting SRPK (e.g., SRPK1 and/or SRPK2), regulating (e.g., inhibiting) VEGF mediated angiogenesis, or treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD), comprising administering to a subject in need thereof an effective amount of a compound of the following structure: or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. Non-limiting illustrative compounds of the application include: CmpdNo.StructureCompound Name62-(4-(3-cyano-9-ethyl-6,6-dimethyl- 11-oxo-6,11-dihydro-5H- benzo[b]carbazol-8-yl)-1H-pyrazol- 1-yl)-N,N-dimethylacetamide79-ethyl-6,6-dimethyl-8-(1-methyl- 1H-pyrazol-4-yl)-11-oxo-6,11- dihydro-5H-benzo[b]carbazole-3- carbonitrile89-ethyl-6,6-dimethyl-11-oxo-8-(1H- pyrazol-4-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile99-ethyl-6,6-dimethyl-11-oxo-8-(1H- pyrazol-3-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile109-ethyl-8-(isoxazol-4-yl)-6,6- dimethyl-11-oxo-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile119-ethyl-8-(furan-3-yl)-6,6- dimethyl-11-oxo-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile129-ethyl-6,6-dimethyl-11-oxo-8- (thiophen-2-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile139-ethyl-6,6-dimethyl-11-oxo-8-(1H- 1,2,3-triazol-5-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile14(9-ethyl-6,6-dimethyl-11-oxo-8- (pyridin-4-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile159-ethyl-6,6-dimethyl-11-oxo-8- (pyridin-3-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile169-ethyl-6,6-dimethyl-11-oxo-8- (pyrimidin-5-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile178-(4-(dimethylamino)piperidin-1- yl)-9-ethyl-6,6-dimethyl-11-oxo- 6,11-dihydro-5H-benzo[b]carbazole- 3-carbonitrile189-ethyl-6,6-dimethyl-11-oxo-8- (piperazin-1-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile199-ethyl-6,6-dimethyl-8-(4- methylpiperazin-1-yl)-11-oxo-6,11- dihydro-5H-benzo[b]carbazole-3- carbonitrile209-ethyl-6,6-dimethyl-8-morpholino- 11-oxo-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile219-ethyl-6,6-dimethyl-11-oxo-6,11- dihydro-5H-benzo[b]carbazole-3,8- dicarbonitrile22(R)-8-(1,2-dihydroxyethyl)-9-ethyl- 6,6-dimethyl-11-oxo-6,11-dihydro- 5H-benzo[b]carbazole-3-carbonitrile23(S)-8-(1,2-dihydroxyethyl)-9-ethyl- 6,6-dimethyl-11-oxo-6,11-dihydro- 5H-benzo[b]carbazole-3-carbonitrile243-cyano-9-ethyl-6,6-dimethyl-11- oxo-6,11-dihydro-5H- benzo[b]carbazole-8-carboxylic acid25(E)-9-ethyl-6,6-dimethyl-8-(3- morpholinoprop-1-en-1-yl)-11-oxo- 6,11-dihydro-5H-benzo[b]carbazole- 3-carbonitrile269-ethyl-8-(6-hydroxypyridin-3-yl)- 6,6-dimethyl-11-oxo-6,11-dihydro- 5H-benzo[b]carbazole-3-carbonitrile278-(6-aminopyridin-3-yl)-9-ethyl-6,6- dimethyl-11-oxo-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile288-((2 (dimethylamino)ethyl)(methyl)amino)- 9-ethyl-6,6-dimethyl-11-oxo- 6,11-dihydro-5H-benzo[b]carbazole- 3-carbonitrile299-ethyl-6,6-dimethyl-11-oxo-8-(1- (tetrahydro-2H-pyran-4-yl)-1H- pyrazol-4-yl)-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile309-ethyl-6,6-dimethyl-11-oxo-8-(1- (piperidin-4-yl)-1H-pyrazol-4-yl)- 6,11-dihydro-5H-benzo[b]carbazole- 3-carbonitrile318-(1-(2-(dimethylamino)ethyl)-1H- pyrazol-4-yl)-9-ethyl-6,6-dimethyl- 11-oxo-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile328-(1-(3-(dimethylamino)propyl)-1H- pyrazol-4-yl)-9-ethyl-6,6-dimethyl- 11-oxo-6,11-dihydro-5H- benzo[b]carbazole-3-carbonitrile332-(4-(3-cyano-9-ethyl-6,6-dimethyl- 11-oxo-6,11-dihydro-5H- benzo[b]carbazol-8-yl)-1H-pyrazol- 1-yl)-N-methylacetamide342-(4-(3-cyano-9-ethyl-6,6-dimethyl- 11-oxo-6,11-dihydro-5H- benzo[b]carbazol-8-yl)-1H-pyrazol- 1-yl)acetamide352-(4-(3-cyano-9-ethyl-6,6-dimethyl- 11-oxo-6,11-dihydro-5H- benzo[b]carbazol-8-yl)-1H-pyrazol- 1-yl)-N,N,2-trimethylpropanamide364-(3-cyano-9-ethyl-6,6-dimethyl-11- oxo-6,11-dihydro-5H- benzo[b]carbazol-8-yl)-N,N- dimethyl-1H-pyrazole-1- carboxamide373-(3-cyano-9-ethyl-6,6-dimethyl-11- oxo-6,11-dihydro-5H- benzo[b]carbazol-8- yl)benzenesulfonyl fluoride384-(3-cyano-9-ethyl-6,6-dimethyl-11- oxo-6,11-dihydro-5H- benzo[b]carbazol-8- yl)benzenesulfonyl fluoride A compound of the present application is capable of binding to the ATP binding site in ALK. In some embodiments, a compound of the present application is capable of binding to the ATP binding site in a wild-type ALK. In some embodiments, a compound of the present application is capable of binding to the ATP binding site in ALK comprising one or more mutations. In some embodiments, a compound of the present application is capable of modulating (e.g., inhibiting or decreasing) the activity of a wild-type ALK and/or ALK comprising one or more mutations. In some embodiments, the mutant ALK comprises one or more mutations selected from C1156Y, F1174L, L1196M, L1152R, 1151 Tins, G1202R, G1269A, and S1206Y. In some embodiments, the mutant ALK comprises at least the mutation G1202R, optionally in combination with one or more other ALK mutations (e.g., C1156Y, F1174L, L1196M, L1152R, 1151 Tins, G1269A, and S1206Y). In further embodiments, a compound of the present application is capable of modulating (e.g., inhibiting or decreasing) the activity of a wild-type ALK and/or an ALK mutant comprising one or more mutations selected from C1156Y, F1174L, L1196M, L1152R, 1151 Tins, G1202R, G1269A, and S1206Y. In further embodiments, a compound of the present application is capable of modulating (e.g., inhibiting or decreasing) the activity of a wild-type ALK and/or a mutant ALK comprising at least the mutation G1202R, optionally in combination with one or more other ALK mutations (e.g., C1156Y, F1174L, L1196M, L1152R, 1151 Tins, G1269A, and S1206Y). In some embodiments, a compound of the present application is capable of modulating (e.g., inhibiting or decreasing) the activity of SRPK (e.g., SRPK1 and/or SRPK2). In still further embodiments, modulation of SRPK (e.g., SRPK1 and/or SRPK2) activity may regulate VEGF mediated angiogenesis. In some embodiments, the inhibition of SRPK (e.g., SRPK1 and/or SRPK2), ALK and ALK mutants by a compound of the present application is measured by IC50. In some embodiments, the inhibition of SRPK (e.g., SRPK1 and/or SRPK2), ALK and ALK mutants by a compound of the present application is measured by EC50. Potency of the inhibitor can be determined by EC50value. A compound with a lower EC50value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher EC50value. In some embodiments, the substantially similar conditions comprise determining an ALK-dependent phosphorylation level, in vitro or in vivo (e.g., in Ba/F3cells or a tumor cell transduced with a wild-type ALK, a mutant ALK, or a fragment of thereof). In some embodiments, the substantially similar conditions comprise determining an SRPK (e.g., SRPK1 and/or SRPK2)-dependent phosphorylation level, in vitro or in vivo. Potency of the inhibitor can also be determined by IC50value. A compound with a lower IC50value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher IC50value. In some embodiments, the substantially similar conditions comprise determining an ALK-dependent phosphorylation level, in vitro or in vivo (e.g., in Ba/F3cells or a tumor cell transduced with a wild-type ALK, a mutant ALK, or a fragment of thereof). In some embodiments, the substantially similar conditions comprise determining an SRPK (e.g., SRPK1 and/or SRPK2)-dependent phosphorylation level, in vitro or in vivo. The inhibition of wild-type ALK and ALK comprising one or more mutations by a compound of the present application can also be measured using cellular proliferation assays where cell proliferation is dependent on kinase activity. For example, Ba/F3cells or cancer cell lines (e.g., NSCLC) transfected with a wild-type ALK, or an ALK mutant comprising one or more mutations selected from C1156Y, F1174L, L1196M, L1152R, 1151 Tins, G1202R, G1269A, and S1206Y can be used. Proliferation assays are performed at a range of inhibitor concentrations and EC50's or IC50's are calculated. An alternative method to measure effects on ALK activity is to assay ALK phosphorylation. For example, a wild-type ALK or mutant ALK (e.g., C1156Y, F1174L, L1196M, L1152R, 1151 Tins, G1202R, G1269A, and/or S1206Y) can be transfected into Ba/F3cells or cancer cell lines (e.g., NSCLC) (which may or may not normally express endogenous ALK), and the ability of the inhibitor to inhibit ALK phosphorylation can be assayed. The effects on ALK phosphorylation can be measured by Western blotting using phospho-specific ALK antibodies. The inhibition of SRPK (e.g., SRPK1 and/or SRPK2) by a compound of the present application can also be measured using cell vascularization assays (e.g., choroid neovasculariation assay) where vascularization is dependent on SRPK (e.g., SRPK1 and/or SRPK2) activity. Alternatively, The inhibition of SRPK (e.g., SRPK1 and/or SRPK2) by a compound of the present application can also be evaluated by measuring VEGF splicing. In some embodiments, a compound of the application exhibits greater inhibition of ALK comprising one or more mutations as described herein relative to a wild-type ALK. In certain embodiments, a compound of the application exhibits at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of ALK comprising one or more mutations as described herein relative to a wild-type ALK. In some embodiments, a compound of present application is more potent than one or more known ALK inhibitors, including but not limited to, Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922, at inhibiting the activity of a wild-type ALK. For example, the compounds can be at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold more potent (e.g., as measured by IC50) than Alectinib, Ceritinib, Brigatinib, Crizotinib, and/or PF-06463922, at inhibiting the activity of a wild-type ALK. In some embodiments, a compound of present application is more potent than one or more known ALK inhibitors, including but not limited to, Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922, at inhibiting the activity of ALK comprising one or more mutations as described herein. For example, the compounds can be at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold more potent (e.g., as measured by IC50) than Alectinib, Ceritinib, Brigatinib, Crizotinib, and/or PF-06463922, at inhibiting the activity of ALK comprising one or more mutations as described herein. In some embodiments, a compound of present application is more potent than one or more known ALK inhibitors, including but not limited to, Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922, at inhibiting the activity of ALK containing at least the G1202R mutation. For example, the compounds can be at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold more potent (e.g., as measured by IC50) than Alectinib, Ceritinib, Brigatinib, Crizotinib, and/or PF-06463922, at inhibiting the activity of ALK containing at least the G1202R mutation. In some embodiments, a compound of present application is more potent at inhibiting the activity of ALK comprising one or more mutations as described herein, but less potent at inhibiting the activity of a wild-type ALK, than one or more known ALK inhibitors, including but not limited to, Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922. For example, the compounds can be at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold more potent (e.g., as measured by IC50) at inhibiting the activity of ALK comprising one or more mutations as described herein, but at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold less potent (e.g., as measured by IC50) at inhibiting the activity of a wild-type ALK, than Alectinib, Ceritinib, Brigatinib, Crizotinib, and/or PF-06463922. In some embodiments, a compound of present application is more potent at inhibiting the activity of ALK containing at least the G1202R mutation, but less potent at inhibiting the activity of a wild-type ALK, than one or more known ALK inhibitors, including but not limited to, Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922. For example, the compounds can be at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold more potent (e.g., as measured by IC50) at inhibiting the activity of ALK containing at least the G1202R mutation, but at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold less potent (e.g., as measured by IC50) at inhibiting the activity of a wild-type ALK, than Alectinib, Ceritinib, Brigatinib, Crizotinib, and/or PF-06463922. In some embodiments, a compound of present application is more potent at inhibiting the activity of ALK comprising one or more mutations as described herein, and more potent at inhibiting the activity of a wild-type ALK, than one or more known ALK inhibitors, including but not limited to, Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922. For example, the compounds can be at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold more potent (e.g., as measured by IC50) at inhibiting the activity of ALK comprising one or more mutations as described herein, and at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold more potent (e.g., as measured by IC50) at inhibiting the activity of a wild-type ALK, than Alectinib, Ceritinib, Brigatinib, Crizotinib, and/or PF-06463922. In some embodiments, a compound of present application is more potent at inhibiting the activity of ALK containing at least the G1202R mutation, and more potent at inhibiting the activity of a wild-type ALK, than one or more known ALK inhibitors, including but not limited to, Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922. For example, the compounds can be at least about 2-fold, about 3-fold, about 5-fold, about 10-fold, about 25-fold, about 50-fold or about 100-fold more potent (e.g., as measured by IC50) at inhibiting the activity of ALK containing at least the G1202R mutation, and at least about 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or about 100-fold more potent (e.g., as measured by IC50) at inhibiting the activity of a wild-type ALK, than Alectinib, Ceritinib, Brigatinib, Crizotinib, and/or PF-06463922. In some embodiments, a compound of the application displays high brain exposure (or brain permeability). Brain exposure can be measured by various methods known in the art. For example, brain exposure can be measured by calculating the ratio between the concentration of a compound of the application in the brain and the concentration of the compound in the plasma (i.e., B:P ratio). In some embodiments, a compound of the application has a B:P ratio of at least 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1.0 at 2 hours after administration of the compound to a subject. In some embodiments, a compound of the application displays higher brain exposure than one or more known ALK inhibitors, including but not limited to, Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922. In some embodiments, a compound of the application has a B:P ratio that is at least about 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or about 100-fold of the B:P ratio of one or more known ALK inhibitors. Definitions Listed below are definitions of various terms used to describe this application. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group. The term “alkyl,” as used herein, refers to saturated, straight- or branched-chain hydrocarbon radicals containing, in certain embodiments, between one and six, or one and eight carbon atoms, respectively. Examples of C1-C6alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, n-hexyl radicals; and examples of C1-C8alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, n-hexyl, heptyl, octyl radicals. The term “alkenyl,” as used herein, denotes a monovalent group derived from a hydrocarbon moiety containing, in certain embodiments, from two to six, or two to eight carbon atoms having at least one carbon-carbon double bond. The double bond may or may not be the point of attachment to another group. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, heptenyl, octenyl and the like. The term “alkoxy” refers to an —O-alkyl radical. The term “heteroaryl,” as used herein, refers to a mono- or poly-cyclic (e.g., bi-, or tri-cyclic or more) fused or non-fused, radical or ring system having at least one aromatic ring, having from five to ten ring atoms of which at least one ring atom is selected from S, O, and N; zero, one, or two ring atoms are additional heteroatoms independently selected from S, O, and N; and the remaining ring atoms are carbon. Heteroaryl includes, but is not limited to, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl, and the like. In accordance with the application, any of the heteroaryls and substituted heteroaryls described herein, can be any aromatic group. Aromatic groups can be substituted or unsubstituted. The term “heterocyclyl,” as used herein, refers to a non-aromatic 3-, 4-, 5-, 6- or 7-membered ring or a bi- or tri-cyclic group fused of non-fused system, where (i) each ring contains between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, (ii) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, and (iv) the nitrogen heteroatom may optionally be quaternized. Representative heterocyclyl groups include, but are not limited to, [1,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, and tetrahydrofuryl. The term “alkylamino” refers to a group having the structure —NH(C1-C12alkyl), e.g., —NH(C1-C6alkyl), where C1-C6alkyl is as previously defined. The term “dialkylamino” refers to a group having the structure —N(C1-C12alkyl)2, e.g., —N(C1-C6alkyl)2, where C1-C6alkyl is as previously defined. The terms “hal,” “halo,” and “halogen,” as used herein, refer to an atom selected from fluorine, chlorine, bromine, and iodine. The term “alkyl linker” is intended to include C1, C2, C3, C4, C5or C6straight chain (linear) saturated aliphatic hydrocarbon groups and C3, C4, C5or C6branched saturated aliphatic hydrocarbon groups. For example, C1-C6alkyl linker is intended to include C1, C2, C3, C4, C5and C6alkyl linker groups. Examples of alkyl linker include, moieties having from one to six carbon atoms, such as, but not limited to, methyl linker (—CH2—), ethyl linker (—CH2CH2— or —CH(CH3)—), propyl linker (—CH2CH2CH2—, —CH(CH3)CH2—, or —C(CH3)2—), butyl linker (—CH2CH2CH2CH2—, —CH(CH3)CH2CH2—, —CH2CH(CH3)CH2—, —C(CH3) 2CH2—, or —CH(CH3)CH(CH3)—), pentyl linker (—CH2CH2CH2CH2CH2—, —CH(CH3)CH2CH2CH2—, —CH2CH(CH3)CH2CH2—, —C(CH3)2CH2CH2—, or —CH2C(CH3)2CH2—), and hexyl linker (—CH2CH2CH2CH2CH2CH2—). As described herein, a compound of the application may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the application. It will be appreciated that the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted.” In general, the term “substituted”, whether preceded by the term “optionally” or not, refers to the replacement of hydrogen in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, and the substituent may be either the same or different at every position. It is understood that the aryls, heteroaryls, alkyls, and the like can be substituted. The term “cancer” includes, but is not limited to, the following cancers: epidermoid Oral: buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma, and teratoma; Lung: bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, larynx, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel or large intestines (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), colon, colon-rectum, colorectal, rectum; Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, biliary passages; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma (serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma (malignant lymphoma) hairy cell; lymphoid disorders; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma; medullary thyroid carcinoma, undifferentiated thyroid cancer, multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary thyroid cancer, pheochromocytoma, paraganglioma; and Adrenal glands: neuroblastoma. Thus, the term “cancerous cell” as provided herein, includes a cell afflicted by any one of the above-identified conditions. The term “AMD” herein refers to age related macular degeneration. The term “ALK” herein refers to anaplastic lymphoma kinase. The term “SRPK1” herein refers to serine-rich protein kinase-1. The term “SRPK2” herein refers to serine-rich protein kinase-1. The term “VEGF” herein refers to vascular endothelial growth factor. The term “subject” as used herein refers to a mammal. A subject therefore refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, and the like. Preferably the subject is a human. When the subject is a human, the subject may be referred to herein as a patient. “Treat”, “treating”, and “treatment” refer to a method of alleviating or abating a disease and/or its attendant symptoms. As used herein, “preventing” or “prevent” describes reducing or eliminating the onset of the symptoms or complications of the disease, condition, or disorder. As used herein the term “AF802” or “Alectinib” refers to a compound having the chemical structure: As used herein the term “LDK378” or “Ceritinib” refers to a compound having the chemical structure: As used herein the term “AP26113” or “Brigatinib” refers to a compound having the chemical structure: As used herein the term “Crizotinib” or “Xalkori” refers to a compound having the chemical structure: As used herein the term “PF-06463922” refers to a compound having the chemical structure: As used herein, the term “pharmaceutically acceptable salt” refers to those salts of the compounds formed by the process of the present application which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977). The salts can be prepared in situ during the final isolation and purification of a compound of the application, or separately by reacting the free base function with a suitable organic acid. Examples of pharmaceutically acceptable salts include, but are not limited to, salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid, or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid, or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aryl sulfonate. As used herein, the term “pharmaceutically acceptable ester” refers to esters of the compounds formed by the process of the present application which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic, and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates. The term “pharmaceutically acceptable prodrugs” as used herein refers to those prodrugs of the compounds formed by the process of the present application which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of a compound of the present application. “Prodrug”, as used herein means a compound which is convertible in vivo by metabolic means (e.g., by hydrolysis) to afford any compound delineated by the formulae of the instant application. Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, vol. 4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed). “Design and Application of Prodrugs, Textbook of Drug Design and Development, Chapter 5, 113-191 (1991); Bundgaard, et al., Journal of Drug Deliver Reviews, 8:1-38(1992); Bundgaard, J. of Pharmaceutical Sciences, 77:285 et seq. (1988); Higuchi and Stella (eds.) Prodrugs as Novel Drug Delivery Systems, American Chemical Society (1975); and Bernard Testa & Joachim Mayer, “Hydrolysis In Drug And Prodrug Metabolism: Chemistry, Biochemistry And Enzymology,” John Wiley and Sons, Ltd. (2002). This application also encompasses pharmaceutical compositions containing, and methods of treating disorders through administering, pharmaceutically acceptable prodrugs of a compound of the application. For example, a compound of the application having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs. Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues is covalently joined through an amide or ester bond to a free amino, hydroxy or carboxylic acid group of a compound of the application. The amino acid residues include but are not limited to the 20 naturally occurring amino acids commonly designated by three letter symbols and also includes 4-hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone. Additional types of prodrugs are also encompassed. For instance, free carboxyl groups can be derivatized as amides or alkyl esters. Free hydroxy groups may be derivatized using groups including but not limited to hemisuccinates, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxy carbonyls, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 1 15. Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups. Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers wherein the acyl group may be an alkyl ester, optionally substituted with groups including but not limited to ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed. Prodrugs of this type are described in J. Med. Chem. 1996, 39, 10. Free amines can also be derivatized as amides, sulfonamides or phosphonamides. All of these prodrug moieties may incorporate groups including but not limited to ether, amine and carboxylic acid functionalities. Combinations of substituents and variables envisioned by this application are only those that result in the formation of stable compounds. The term “stable”, as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject). The application also provides for a pharmaceutical composition comprising a compound disclosed herein, or a pharmaceutically acceptable ester, salt, or prodrug thereof, together with a pharmaceutically acceptable carrier. In another aspect, the application provides a method of synthesizing a compound disclosed herein. The synthesis of a compound of the application can be found herein and in the Examples below. Another aspect is an isotopically labeled compound of any of the formulae delineated herein. Such compounds have one or more isotope atoms which may or may not be radioactive (e.g.,3H,2H,14C,13C,18F,35S,32P,125I, and131I) introduced into the compound. Such compounds are useful for drug metabolism studies and diagnostics, as well as therapeutic applications. A compound of the application can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid. Alternatively, a pharmaceutically acceptable base addition salt of a compound of the application can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base. Alternatively, the salt forms of a compound of the application can be prepared using salts of the starting materials or intermediates. The free acid or free base forms of a compound of the application can be prepared from the corresponding base addition salt or acid addition salt from, respectively. For example a compound of the application in an acid addition salt form can be converted to the corresponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like). A compound of the application in a base addition salt form can be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc.). Prodrugs of a compound of the application can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et al., (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). For example, appropriate prodrugs can be prepared by reacting a non-derivatized compound of the application with a suitable carbamylating agent (e.g., 1,1-acyloxyalkylcarbanochloridate, para-nitrophenyl carbonate, or the like). Protected derivatives of a compound of the application can be made by means known to those of ordinary skill in the art. A detailed description of techniques applicable to the creation of protecting groups and their removal can be found in T. W. Greene, “Protecting Groups in Organic Chemistry”, 3rd edition, John Wiley and Sons, Inc., 1999. Acids and bases useful in the methods herein are known in the art. Acid catalysts are any acidic chemical, which can be inorganic (e.g., hydrochloric, sulfuric, nitric acids, aluminum trichloride) or organic (e.g., camphorsulfonic acid, p-toluenesulfonic acid, acetic acid, ytterbium triflate) in nature. Acids are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions. Bases are any basic chemical, which can be inorganic (e.g., sodium bicarbonate, potassium hydroxide) or organic (e.g., triethylamine, pyridine) in nature. Bases are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions. In addition, some of a compound of this application have one or more double bonds, or one or more asymmetric centers. Such compounds can occur as racemates, racemic mixtures, single enantiomers, individual diastereomers, diastereomeric mixtures, and cis- or trans- or E- or Z-double isomeric forms, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-, or as (D)- or (L)- for amino acids. All such isomeric forms of these compounds are expressly included in the present application. Optical isomers may be prepared from their respective optically active precursors by the procedures described herein, or by resolving the racemic mixtures. The resolution can be carried out in the presence of a resolving agent, by chromatography or by repeated crystallization or by some combination of these techniques which are known to those skilled in the art. Further details regarding resolutions can be found in Jacques, et al., Enantiomers, Racemates, and Resolutions (John Wiley & Sons, 1981). “Isomerism” means compounds that have identical molecular formulae but differ in the sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers”. Stereoisomers that are not mirror images of one another are termed “diastereoisomers”, and stereoisomers that are non-superimposable mirror images of each other are termed “enantiomers” or sometimes optical isomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a “racemic mixture”. A carbon atom bonded to four non-identical substituents is termed a “chiral center”. “Chiral isomer” means a compound with at least one chiral center. Compounds with more than one chiral center may exist either as an individual diastereomer or as a mixture of diastereomers, termed “diastereomeric mixture”. When one chiral center is present, a stereoisomer may be characterized by the absolute configuration (R or S) of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al., Angew. Chem. Inter. Edit. 1966, 5, 385; errata 511; Cahn et al., Angew. Chem. 1966, 78, 413; Cahn and Ingold, J. Chem. Soc. 1951 (London), 612; Cahn et al., Experientia 1956, 12, 81; Cahn, J. Chem. Educ. 1964, 41, 116). “Geometric isomer” means the diastereomers that owe their existence to hindered rotation about double bonds. These configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold-Prelog rules. Furthermore, the structures and other compounds discussed in this application include all atropic isomers thereof “Atropic isomers” are a type of stereoisomer in which the atoms of two isomers are arranged differently in space. Atropic isomers owe their existence to a restricted rotation caused by hindrance of rotation of large groups about a central bond. Such atropic isomers typically exist as a mixture, however as a result of recent advances in chromatography techniques; it has been possible to separate mixtures of two atropic isomers in select cases. “Tautomer” is one of two or more structural isomers that exist in equilibrium and is readily converted from one isomeric form to another. This conversion results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. Tautomers exist as a mixture of a tautomeric set in solution. In solid form, usually one tautomer predominates. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent and pH. The concept of tautomers that are interconvertable by tautomerizations is called tautomerism. Of the various types of tautomerism that are possible, two are commonly observed. In keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs. Ring-chain tautomerism arises as a result of the aldehyde group (—CHO) in a sugar chain molecule reacting with one of the hydroxy groups (—OH) in the same molecule to give it a cyclic (ring-shaped) form as exhibited by glucose. Common tautomeric pairs are: ketone-enol, amide-nitrile, lactam-lactim, amide-imidic acid tautomerism in heterocyclic rings (e.g., in nucleobases such as guanine, thymine and cytosine), amine-enamine and enamine-enamine. A compound of this application may also be represented in multiple tautomeric forms, in such instances, the application expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the application expressly includes all such reaction products). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. The configuration of any carbon-carbon double bond appearing herein is selected for convenience only and is not intended to designate a particular configuration unless the text so states; thus a carbon-carbon double bond depicted arbitrarily herein as trans may be cis, trans, or a mixture of the two in any proportion. All such isomeric forms of such compounds are expressly included in the present application. In the present specification, the structural formula of the compound represents a certain isomer for convenience in some cases, but the present application includes all isomers, such as geometrical isomers, optical isomers based on an asymmetrical carbon, stereoisomers, tautomers, and the like. Additionally, a compound of the present application, for example, the salts of the compounds, can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules. Non-limiting examples of hydrates include monohydrates, dihydrates, etc. Non-limiting examples of solvates include ethanol solvates, acetone solvates, etc. A compound of the present application can be conveniently prepared, or formed during the process of the application, as solvates (e.g., hydrates). Hydrates of a compound of the present application can be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol. “Solvate” means solvent addition forms that contain either stoichiometric or non stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate; and if the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one molecule of the substance in which the water retains its molecular state as H2O. The synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization. As can be appreciated by the skilled artisan, further methods of synthesizing a compound of the formulae herein will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. In addition, the solvents, temperatures, reaction durations, etc. delineated herein are for purposes of illustration only and one of ordinary skill in the art will recognize that variation of the reaction conditions can produce the desired bridged macrocyclic products of the present application. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof. A compound of this application may be modified by appending various functionalities via any synthetic means delineated herein to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion. A compound of the application is defined herein by their chemical structures and/or chemical names. Where a compound is referred to by both a chemical structure and a chemical name, and the chemical structure and chemical name conflict, the chemical structure is determinative of the compound's identity. The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof. Methods of Synthesizing the Compounds A compound of the present application may be made by a variety of methods, including standard chemistry. The synthetic processes of the application can tolerate a wide variety of functional groups, therefore various substituted starting materials can be used. The processes generally provide the desired final compound at or near the end of the overall process, although it may be desirable in certain instances to further convert the compound to a pharmaceutically acceptable salt, ester, or prodrug thereof. Suitable synthetic routes are depicted in the schemes below. A compound of the present application can be prepared in a variety of ways using commercially available starting materials, compounds known in the literature, or from readily prepared intermediates, by employing standard synthetic methods and procedures either known to those skilled in the art, or which will be apparent to the skilled artisan in light of the teachings herein. Standard synthetic methods and procedures for the preparation of organic molecules and functional group transformations and manipulations can be obtained from the relevant scientific literature or from standard textbooks in the field. Although not limited to any one or several sources, classic texts such as Smith, M. B., March, J., March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5thedition, John Wiley & Sons: New York, 2001; and Greene, T. W., Wuts, P. G. M., Protective Groups in Organic Synthesis, 3rdedition, John Wiley & Sons: New York, 1999, incorporated by reference herein, are useful and recognized reference textbooks of organic synthesis known to those in the art. The following descriptions of synthetic methods are designed to illustrate, but not to limit, general procedures for the preparation of a compound of the present application. A compound disclosed herein may be prepared by methods known in the art of organic synthesis as set forth in part by the following synthetic schemes. In the schemes described below, it is well understood that protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles or chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Greene and P. G. M. Wuts, “Protective Groups in Organic Synthesis”, Third edition, Wiley, New York 1999). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection processes, as well as the reaction conditions and order of their execution, shall be consistent with the preparation of a compound disclosed herein. Those skilled in the art will recognize if a stereocenter exists in a compound disclosed herein. Accordingly, the present application includes both possible stereoisomers (unless specified in the synthesis) and includes not only racemic compounds but the individual enantiomers and/or diastereomers as well. When a compound is desired as a single enantiomer or diastereomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate. Resolution of the final product, an intermediate, or a starting material may be affected by any suitable method known in the art. See, for example, “Stereochemistry of Organic Compounds” by E. L. Eliel, S. H. Wilen, and L. N. Mander (Wiley-lnterscience, 1994). The compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, and/or enzymatic processes. All the abbreviations used in this application are found in “Protective Groups in Organic Synthesis” by John Wiley & Sons, Inc, or the MERCK INDEX by MERCK & Co., Inc, or other chemistry books or chemicals catalogs by chemicals vendor such as Aldrich, or according to usage know in the art. A compound of the present application can be prepared in a number of ways well known to those skilled in the art of organic synthesis. By way of example, a compound of the present application can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. Preferred methods include but are not limited to those methods described below. A compound of the present application can be synthesized by following the steps outlined in General Scheme A. Starting materials are either commercially available or made by known procedures in the reported literature or as illustrated. The general way of preparing a compound of Formula (I) is exemplified in General Scheme A. Compound 37-b is reacted with Compound A in a suitable solvent, e.g., 1,4-dioxane, through Suzuki coupling to yield a compound of Formula (I). Compound 37-b is reacted with Compound B in a suitable solvent, e.g., 1,4-dioxane, through Suzuki coupling to yield Compound 40, which can be used as an intermediate to prepare other compounds of Formula (I). Scheme 1 shows the synthetic route to Compound 6. Starting material Compound 37 (commercially available) is subjected to Suzuki coupling conditions followed by ester hydrolysis to afford the carboxylic acid Compound 38. Compound 38 is then reacted with dimethyalmine HCl and HATU to provide Compound 6. Compounds with an alkyl heterocyclic ring substituent are prepared by subjecting Compound 37 to Buchwald-Hartwig coupling conditions using the desired amine (Scheme 2). Scheme 3 provides the synthetic route to prepare Compound 13 and a compound of the present application with similar structures. Scheme 4 provides the synthetic route to prepare Compounds 22 and 24 and a compound of the present application with similar structures. A mixture of enantiomers, diastereomers, and/or cis/trans isomers resulting from the processes described above can be separated into their single components by chiral salt technique, chromatography using normal phase, or reverse phase or chiral column, depending on the nature of the separation. It should be understood that in the description and formulae shown above, the various groups R1-R6, m, and n are as defined herein, except where otherwise indicated. Furthermore, for synthetic purposes, the compounds in the Schemes are mere representatives with elected sustituents to illustrate the general synthetic methodology of a compound disclosed herein. Biological Assays Growth Inhibition Assay A compound of the present application is tested in various cells (e.g., Ba/F3cells, or tumor cell lines such as NSCLC) untransduced or transduced with wild-type ALK or ALK comprising one or more mutations (e.g., mutations described herein) in a growth inhibition assay at a fixed concentration or a series of concentrations. The cells are treated with the compounds for different durations, after which the percentage of viable cells was determined via a MTS assay. If necessary, the IC50's or EC50's are then calculated. Western Blotting Cell lysates from cells treated with a compound of the present application is equalized to protein content and loaded onto a gel with running buffer. Proteins are transferred from the gel to membranes, which are probed with primary antibodies against the protein (e.g., ALK). Secondary antibodies are added and after washing, the amount of the protein is determined, e.g., by detecting the signal intensity of a HRP substrate reagent with an imager. Methods of the Application In another aspect, the application provides a method of inhibiting a kinase (e.g., SRPK (e.g., SRPK1 and/or SRPK2), ALK or a mutant ALK (e.g., ALK G1202R)) with a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In some embodiments, the ALK comprises one or more mutations selected from C1156Y, F1174L, L1196M, L1152R, 1151 Tins, G1202R, G1269A, and S1206Y. In further embodiments, the mutant ALK comprises at least the mutation G1202R. Another aspect of the application provides a method of treating or preventing a disease, the method comprising administering to a subject in need thereof an effective amount of a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In some embodiments, the disease is mediated by a kinase. In further embodiments, the kinase is ALK (e.g., a wild-type ALK or mutant ALK described herein). In another embodiment, the kinase is SRPK (e.g., SRPK1 and/or SRPK2). In some embodiments, the disease is mediated by ALK (e.g., ALK plays a role in the initiation or development of the disease). In further embodiments, the disease is mediated by a mutant ALK described herein. In further embodiments, the ALK mutant comprises at least the mutation G1202R. In some embodiments, the disease is mediated by SRPK (e.g., SRPK1 and/or SRPK2) (e.g., SRPK1 and/or SRPK2 plays a role in the initiation or development of the disease). In further embodiments any disease ordisorder associated with abnormal angiogenesis or abnormal over-production of proangiogenic VEGF isoforms. In certain embodiments, the disease is cancer or a proliferative disease. In further embodiments, the cancer is non-small-cell lung cancer (NSCLC), inflammatory myofibroblastic tumors (IMT), diffuse large B cell lymphoma (DLBCL), squamous cell carcinoma, neuroblastoma, adult and pediatric renal cell carcinomas, glioblastoma multiforme, anaplastic thyroid cancer, colon cancer, breast cancer, prostate cancer, liver cancer, pancreas cancer, brain cancer, kidney cancer, ovarian cancer, stomach cancer, skin cancer, bone cancer, gastric cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, hepatocellular carcinoma, papillary renal carcinoma, head and neck squamous cell carcinoma, leukemias, lymphomas, myelomas, or solid tumors. In further embodiments, the cancer is NSCLC or neuroblastoma. In some embodiments, the cancer is a cancer of the central nervous system (CNS). In some embodiments, the cancer is a cancer from the replase of a cancer previously treated with an ALK targeted therapy, such as a therapy with Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922. In another aspect, the application provides a method of treating or preventing cancer, wherein the cancer cell comprise activated ALK, comprising administering to a subject in need thereof an effective amount of a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In some embodiments, the activated ALK is a wild-type ALK. In other embodiments, the activated ALK is a mutant ALK described herein. In further embodiments, the activated ALK is a mutant ALK comprising at least the mutation G1202R. Another aspect of the application provides a method of treating or preventing cancer in a subject, wherein the subject is identified as being in need of ALK inhibition for the treatment of cancer, comprising administering to the subject an effective amount of a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In some embodiments, the subject is in need of inhibition of a wild-type ALK. In other embodiments, the subject is in need of inhibition of a mutant ALK described herein. In further embodiments, the subject is in need of inhibition of a mutant ALK comprising at least the mutation G1202R. Another aspect of the application provides a method of treating or preventing a disease or disorder (e.g., cancer), wherein the disease or disorder is resistant to an ALK targeted therapy, such as a therapy with Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922. In another aspect, the application provides a method of treating or preventing cancer, wherein the cancer cell comprises a mutant ALK, comprising administering to a subject in need thereof an effective amount of a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In some embodiments, the ALK mutant comprises one or more mutations described herein. In further embodiments, the ALK mutant comprises at least the mutation G1202R. Another aspect of the application provides a method of treating or preventing resistance to a known ALK inhibitor, including but not limited to, Alectinib, Ceritinib, Brigatinib, Crizotinib or PF-06463922, comprising administering to a subject in need thereof an effective amount of a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In certain embodiments, the application provides a method of treating any of the disorders described herein, wherein the subject is a human. In certain embodiments, the application provides a method of preventing any of the disorders described herein, wherein the subject is a human. In another aspect, the application provides a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating or preventing a disease in which ALK plays a role. In still another aspect, the application provides a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for inhibiting a kinase (e.g., ALK or a mutant ALK (e.g., ALK G1202R), or SRPK (e.g., SRPK1 and/or SRPK2)); treating or preventing a disease or disorder (e.g., cancer) in which a kinase (e.g., ALK or a mutant ALK (e.g., ALK G1202R), or SRPK (e.g., SRPK1 and/or SRPK2)) plays a role; treating or preventing cancer, wherein the cancer cell comprises activated ALK or a mutant ALK; treating or preventing cancer in a subject identified as being in need of inhibition of ALK or the mutant ALK for the treatment or prevention of cancer; treating or preventing a disease or disorder (e.g., cancer) resistant to an ALK targeted therapy, such as a therapy with Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922; regulating (e.g., inhibiting) VEGF mediated angiogenesis; treating or preventing a disease or disorder in which a VEGF mediated angiogenesis plays a role (e.g., AMD or angiogenesis-dependent cancers); treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD; and/or treating or preventing an angiogenesis-dependent cancer (e.g., tumorous cancer) (e.g., in a subject identified in need of regulation of VEGF mediated angiogenesis for the treatment or prevention of an angiogenesis-dependent cancer). In still another aspect, the application provides use of a compound disclosed herein (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for inhibiting a kinase (e.g., ALK or a mutant ALK (e.g., ALK G1202R), or SRPK (e.g., SRPK1 and/or SRPK2)); treating or preventing a disease or disorder (e.g., cancer) in which a kinase (e.g., ALK or a mutant ALK (e.g., ALK G1202R), or SRPK (e.g., SRPK1 and/or SRPK2)) plays a role; treating or preventing cancer, wherein the cancer cell comprises activated ALK or a mutant ALK; treating or preventing cancer in a subject identified as being in need of inhibition of ALK or the mutant ALK for the treatment or prevention of cancer; treating or preventing a disease or disorder (e.g., cancer) resistant to an ALK targeted therapy, such as a therapy with Alectinib (AF802), Ceritinib (LDK378), Brigatinib (AP26113), Crizotinib (Xalkori), and/or PF-06463922; regulating (e.g., inhibiting) VEGF mediated angiogenesis; treating or preventing a disease or disorder in which a VEGF mediated angiogenesis plays a role (e.g., AMD or angiogenesis-dependent cancers); treating or preventing AMD (e.g., in a subject identified in need of regulation (e.g., inhibition) of VEGF mediated angiogenesis for the treatment or prevention of AMD; and/or treating or preventing an angiogenesis-dependent cancer (e.g., tumorous cancer) (e.g., in a subject identified in need of regulation of VEGF mediated angiogenesis for the treatment or prevention of an angiogenesis-dependent cancer). One aspect of this application provides compounds that are useful for the treatment of diseases, disorders, and conditions characterized by excessive or abnormal cell proliferation. Such diseases include, but are not limited to, a proliferative or hyperproliferative disease. Examples of proliferative and hyperproliferative diseases include, without limitation, cancer. The term “cancer” includes, but is not limited to, the following cancers: breast; ovary; cervix; prostate; testis, genitourinary tract; esophagus; larynx, glioblastoma; neuroblastoma; stomach; skin, keratoacanthoma; lung, epidermoid carcinoma, large cell carcinoma, small cell carcinoma, lung adenocarcinoma; bone; colon; colorectal; adenoma; pancreas, adenocarcinoma; thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma; seminoma; melanoma; sarcoma; bladder carcinoma; liver carcinoma and biliary passages; kidney carcinoma; myeloid disorders; lymphoid disorders, Hodgkin's, hairy cells; buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx; small intestine; colonrectum, large intestine, rectum, brain and central nervous system; chronic myeloid leukemia (CML), and leukemia. The term “cancer” includes, but is not limited to, the following cancers: myeloma, lymphoma, or a cancer selected from gastric, renal, or and the following cancers: head and neck, oropharangeal, non-small cell lung cancer (NSCLC), endometrial, hepatocarcinoma, Non-Hodgkins lymphoma, and pulmonary. The term “cancer” refers to any cancer caused by the proliferation of malignant neoplastic cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, lymphomas and the like. For example, cancers include, but are not limited to, mesothelioma, leukemias and lymphomas such as cutaneous T-cell lymphomas (CTCL), noncutaneous peripheral T-cell lymphomas, lymphomas associated with human T-cell lymphotrophic virus (HTLV) such as adult T-cell leukemia/lymphoma (ATLL), B-cell lymphoma, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic myelogenous leukemia, acute myelogenous leukemia, lymphomas, and multiple myeloma, non-Hodgkin lymphoma, acute lymphatic leukemia (ALL), chronic lymphatic leukemia (CLL), Hodgkin's lymphoma, Burkitt lymphoma, adult T-cell leukemia lymphoma, acute-myeloid leukemia (AML), chronic myeloid leukemia (CML), or hepatocellular carcinoma. Further examples include myelodisplastic syndrome, childhood solid tumors such as brain tumors, neuroblastoma, retinoblastoma, Wilms' tumor, bone tumors, and soft-tissue sarcomas, common solid tumors of adults such as head and neck cancers (e.g., oral, laryngeal, nasopharyngeal and esophageal), genitourinary cancers (e.g., prostate, bladder, renal, uterine, ovarian, testicular), lung cancer (e.g., small-cell and non-small cell), breast cancer, pancreatic cancer, melanoma and other skin cancers, stomach cancer, brain tumors, tumors related to Gorlin's syndrome (e.g., medulloblastoma, meningioma, etc.), and liver cancer. Additional exemplary forms of cancer which may be treated by the subject compounds include, but are not limited to, cancer of skeletal or smooth muscle, stomach cancer, cancer of the small intestine, rectum carcinoma, cancer of the salivary gland, endometrial cancer, adrenal cancer, anal cancer, rectal cancer, parathyroid cancer, and pituitary cancer. Additional cancers that the compounds described herein may be useful in preventing, treating and studying are, for example, colon carcinoma, familiary adenomatous polyposis carcinoma and hereditary non-polyposis colorectal cancer, or melanoma. Further, cancers include, but are not limited to, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, gastric carcinoma, adenocarcinoma, thyroid cancer (medullary and papillary thyroid carcinoma), renal carcinoma, kidney parenchyma carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, testis carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, gall bladder carcinoma, bronchial carcinoma, multiple myeloma, basalioma, teratoma, retinoblastoma, choroidea melanoma, seminoma, rhabdomyosarcoma, craniopharyngeoma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, Ewing sarcoma, and plasmocytoma. In one aspect of the application, the present application provides for the use of one or more a compound of the application in the manufacture of a medicament for the treatment of cancer, including without limitation the various types of cancer disclosed herein. This application further embraces the treatment or prevention of cell proliferative disorders such as hyperplasias, dysplasias and pre-cancerous lesions. Dysplasia is the earliest form of pre-cancerous lesion recognizable in a biopsy by a pathologist. The subject compounds may be administered for the purpose of preventing said hyperplasias, dysplasias or pre-cancerous lesions from continuing to expand or from becoming cancerous. Examples of pre-cancerous lesions may occur in skin, esophageal tissue, breast and cervical intraepithelial tissue. Another aspect of the application provides compounds that are useful for the treatment or prevention of any disease or disorder associated with abnormal angiogenesis or abnormal over-production of pro-angiogenicVEGF isoforms. Such diseases and disorders include, for example, vascular disease (e.g., vasoconstriction and disorders characterized by vasoconstriction, and cardiovascular disease), malignant and benign neoplasia (e.g., angiogenesis-dependent cancers, for example tumorous cancers), tumor metastasis, inflammatory disorders, diabetes, diabetic retinopathy and other complications of diabetes (e.g., diabetic neovascularisation), trachoma, retrolental hyperplasia, neovascular glaucoma, age-related macular degeneration, haemangioma, immune rejection of implanted corneal tissue, corneal angiogenesis associated with ocular injury or infection, Osier-Webber Syndrome, myocardial angiogenesis, wound granulation, telangiectasia, hemophiliac joints, angiofibroma, telangiectasia psoriasis scleroderma, pyogenic granuloma, coronary collaterals, ischemic limb abgiogenesis, rubeosis, obesity, arthritis (e.g., rheumatoid arthritis), hematopoieses, vasculogenesis, gingivitis, atherosclerosis, endometriosis, neointimal hyperplasia, psoriasis, hirsutism and proliferative retinopathy. In some embodiments, the application provides compounds that are useful for the treatment or prevention of AMD. The anti-angiogenic treatment according to the present application may also include non-therapeutic treatments performed on healthy subjects, for example to inhibit vascular development for cosmetic purposes. Other disorders in which the alternatively spliced VEGF isoform has been implicated, include but are not limited to microvascular hyperpermeability disorders, disorders of epithelial cell survival and disorders of fenestrations of epithelial filtration membranes. Examples of such conditions include, for example, proteinuria, uraemia, microalbuminuria, hypoalbuminemia, renal hyperfiltration, nephrotic syndrome, renal failure, pulmonary hypertension, capillary hyperpermeability, microaneurysms, oedema and vascular complications of diabetes (e.g., diabetic retinopathy, both proliferative and non-proliferative, and diabetic nephropathy). Exemplary microvascular hyperpermeability disorders include, but are not limited to renal disorders, for example a permeability disorder of the glomerular filtration barrier (e.g., a permeability disorder of the podocytes). Examples of disorders where treatment to support epithelial cell survival would be effective incude acute pulmonary fibrotic disease, adult respiratory distress syndrome, adult respiratory distress syndrome, advanced cancer, allergic respiratory disease, alveolar injury, angiogenesis, arthritis, ascites, asthma, asthma or edema following burns, atherosclerosis, autoimmune diseases, bone resorption, bullous disorder associated with subepidermal blister formation including bullous pemphigoid, cardiovascular condition, certain kidney diseases associated with proliferation of glomerular or mesangial cells, chronic and allergic inflammation, chronic lung disease, chronic occlusive pulmonary disease, cirrhosis, corneal angiogenisis, corneal disease, coronary and cerebral collateral vascularization, coronary restenosis, damage following heart disease, dermatitis herpetiformis, diabetes, diabetic nephropathy, diabetic retinopathy, endotoxic shock, erythema multiforme, fibrosis, glomerular nephritis, glomerulonophritis, graft rejection, gram negative sepsis, hemangioma, hepatic cirrhosis, hepatic failure, Herpes Zoster, host-versus-graft reaction (ischemia reperfusion injury and allograft rejections of kidney, liver, heart, and skin), impaired wound healing in infection, infection by Herpes simplex, infection from human immunodeficiency virus (HIV), inflammation, cancer, inflammatory bowel disease (Crohn's disease and ulcerative colitis), inflammatory conditions, in-stent restenosis, in-stent stenosis, ischemia, ischemic retinal-vein occlusion, ischemic retinopathy, Kaposi's sarcoma, keloid, liver disease during acute inflammation, lung allograft rejection (obliterative bronchitis), lymphoid malignancy, macular degeneration retinopathy of prematurity, myelodysplastic syndromes, myocardial angiogenesis, neovascular glaucoma, non-insulin-dependent diabetes mellitus (NIDDM), obliterative bronchiolitis, ocular conditions or diseases, ocular diseases associated with retinal vessel proliferation, Osier-Weber-Rendu disease, osteoarthritis, ovarian hyperstimulation syndrome, Paget's disease, pancreatitis, pemphigoid, polycystic kidney disease, polyps, postmenopausal osteoperosis, preeclampsia, psoriasis, pulmonary edema, pulmonary fibrosis, pulmonary sarcoidosis, restenosis, restenosis, retinopathy including diabetic retinopathy, retinopathy of prematurity, age related macular degeneration, rheumatoid arthritis, rubeosis, sarcoidosis, sepsis, stroke, synovitis, systemic lupus erythematosus, throiditis, thrombic micoangiopathy syndromes, transplant rejection, trauma, tumor-associated angiogenesis, vascular graft restenosis, vascular graft restenosis, von Hippel Lindau disease, and wound healing. Yet another aspect of the application provides compounds that are useful for the treatment or prevention of macular dystrophy. Non-limiting examples of muscular dystrophy include Stargardt disease/fundus flavimaculatus, Stargardt-like macular dystrophy, Autosomal dominant bull's eye macular dystrophy, Best macular dystrophy, Adult vitelliform dystrophy, Pattern dystrophy, Doyne honeycomb retinal dystrophy, North Carolina macular dystrophy, Autosomal dominant macular dystrophy resembling MCDR1, North Carolina-like macular dystrophy associated with deafness, Progressive bifocal chorioretinal atrophy, Sorsby's fundus dystrophy, Central areolar choroidal dystrophy, Dominant cystoid macular dystrophy, Juvenile retinoschisis; Occult Macular Dystrophy, Non-familial Occult Macular Dystrophy. The disorder may particularly be a disorder of the retinal epithelium, such as geographic atrophy, or age related macular degeneration. In still another aspect the application provides compounds that are useful for the treatment or prevention of neuropathic and neurodegenerative disorders. Neuropathic disorders to be treated or prevented according to the present application include neuropathic pain and diabetic and other neuropathies. Neurodegenerative disorders to be treated or prevented according to the present application include neurodegeneration of the cognitive and non-cognitive types, neuromuscular degeneration, motor-sensory neurodegeneration, and ocular neurodegeneration. In a further aspect of the application, treatment of neuropathic and neurodegenerative disorders provides for the treatment or prevention of pain (e.g., neuropathic pain), dementia, age-related cognitive impairment, Alzheimer's disease, senile dementia of the Alzheimer's type (SDAT), Lewy body dementia, vascular dementia, Parkinson's disease, postencephalitic Parkinsonism, depression, schizophrenia, muscular dystrophy including facioscapulohumeral muscular dystrophy (FSH), Duchenne muscular dystrophy, Becker muscular dystrophy and Bruce's muscular dystrophy, Fuchs' dystrophy, myotonic dystrophy, corneal dystrophy, reflex sympathetic dystrophy syndrome (RSDSA), neurovascular dystrophy, myasthenia gravis, Lambert Eaton disease, Huntington's disease, motor neurone diseases including amyotrophic lateral sclerosis (ALS), multiple sclerosis, postural hypotension, traumatic neuropathy or neurodegeneration, for example following stroke or following an accident, (e.g., traumatic head injury or spinal cord injury), Batten's disease, Cockayne syndrome, Down syndrome, corticobasal ganglionic degeneration, multiple system atrophy, cerebral atrophy, olivopontocerebellar atrophy, dentatorubral atrophy, pallidoluysian atrophy, spinobulbar atrophy, optic neuritis, sclerosing pan-encephalitis (SSPE), attention deficit disorder, post-viral encephalitis, post-poliomyelitis syndrome, Fahr's syndrome, Joubert syndrome, Guillain-Barre syndrome, lissencephaly, Moyamoya disease, neuronal migration disorders, autistic syndrome, polyglutamine disease, Niemann-Pick disease, progressive multifocal leukoencephalopathy, pseudotumor cerebri, Refsum disease, Zellweger syndrome, supranuclear palsy, Friedreich's ataxia, spinocerebellar ataxia type 2, Rhett syndrome, Shy-Drager syndrome, tuberous sclerosis, Pick's disease, chronic fatigue syndrome, neuropathies including hereditary neuropathy, diabetic neuropathy and mitotic neuropathy, prion-based neurodegeneration, including Creutzfeldt-Jakob disease (CJD), variant CJD, new variant CJD, bovine spongiform encephalopathy (BSE), GSS, FFI, Kuru and Alper's syndrome, Joseph's disease, acute disseminated encephalomyelitis, arachnoiditis, vascular lesions of the central nervous system loss of extremity neuronal function, Charcot-Marie-Tooth disease, Krabbe's disease, leukodystrophies, susceptibility to heart failure, asthma, epilepsy, auditory neurodegeneration, macular degeneration, pigmentary retinitis and glaucoma-induced optic nerve degeneration. In a still another aspect of the application, treatment of neuropathic and neurodegenerative disorders provides for the treatment of psychiatric disorders, which includes without limitation anxiety disorders (e.g., acute stress disorder, panic disorder, agoraphobia, social phobia, specific phobia, obsessive-compulsive disorder, sexual anxiety disorders, post-traumatic stress disorder, body dysmorphic disorder and generalized anxiety disorder), childhood disorders (e.g., attention-deficit hyperactivity disorder (ADHD), Asperger's disorder, autistic disorder, conduct disorder, oppositional defiant disorder, separation anxiety disorder and Tourette's disorder), eating disorders (for example, anorexia nervosa and bulimia nervosa), mood disorders (e.g., depression, major depressive disorder, bipolar disorder (manic depression), seasonal affective disorder (SAD), cyclothymic disorder and dysthymic disorder), sleeping disorders, cognitive psychiatric disorders (e.g., delirium, amnestic disorders), personality disorders (e.g., paranoid personality disorder, schizoid personality disorder, schizotypal personality disorder, antisocial personality disorder, borderline personality disorder, histrionic personality disorder, narcissistic personality disorder, avoidant personality disorder, dependent personality disorder andobsessive-compulsive personality disorder), psychotic disorders (e.g., schizophrenia, delusional disorder, brief psychotic disorder, schizophreniform disorder, schizoaffective disorder and shared psychotic disorder), and substance-related disorders (e.g., alcohol dependence, amphetamine dependence, cannabis dependence, cocaine dependence, hallucinogen dependence, inhalant dependence, nicotine dependence, opioid dependence, phencyclidine dependence and sedative dependence). In another aspect of the application, the componds disclosed herein may be used to treat VEGFR2-mediated non-inflammatory pain. VEGFR2-mediated non-inflammatory pain to be treated or prevented according to the present application includes non-inflammatory neuropathic and nociceptive pain where the VEGFR2 receptor is involved in the cause or transmission of the pain. For example, the compounds according to the present application have activity against non-inflammatory allodynia and pain (antiallodynic and analgesic activity). Pain states of this type include chronic pain, whether of the intermittent or constant form. Such pain states may include, for example, low back pain, neuralgia, atypical pains such as atypical facial pain, pain exhibited post-surgery, post-injury (e.g., after surgery or injury causing nerve damage) or in association with cancer or with cancer therapy such as cytotoxic or radiation therapy, or neuropathy associated with diabetes (diabetic neuropathy, insulin neuritis) or other systemic or autoimmune disease or pathology, or the treatment thereof, alcoholism or HIV infection, ageing associated neuropathy, or neuropathy of unknown origin. In accordance with the foregoing, the present application further provides a method for preventing or treating any of the diseases or disorders described above in a subject in need of such treatment, which method comprises administering to said subject a therapeutically effective amount of a compound of the application, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. For any of the above uses, the required dosage will vary depending on the mode of administration, the particular condition to be treated and the effect desired. In another aspect, the application provides a method of labeling a SRPK protein with a compound of the present application, comprising interacting the SRPK protein with a compound of the present application (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein). In one embodiment, the SRPK protein is SRPK1. In another embodiment, the SRPK protein is SRPK2. In one aspect, the SRPK protein (e.g., SRPK1) is labeled at one or more amino acid residues. In one embodiment, the SRPK protein (e.g., SRPK1) is labeled at one amino acid residue. In one embodiment, the SRPK1 protein is labeled at amino acid residue Y227. In one embodiment, the SRPK1 protein is labeled at amino acid residue Y227 with a compound of the present application (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein). In one embodiment, the SRPK1 protein is labeled at amino acid residue Y227 with Compound 37. In one embodiment, interacting the SRPK protein with a compound of the present application (e.g., a compound of any of Formula I, Ia, II, IIa, or IIb, or any specific compound, such as Compounds 6-38, disclosed herein) involves binding of the compound with the SRPK protein. In one embodiment, the compound is bound to the SRPK protein at one or more amino acid residues. In one embodiment, the compound is bound to the SRPK protein at one amino acid residue. In one embodiment, the compound is bound to the SRPK protein at amino acid residue Y227. The compound used to label the SRPK protein can itself be labeled. For example, the compound can be labeled radioactively or fluorescently, according to methods known in the art. Pharmaceutical Compositions In another aspect, the application provides a pharmaceutical composition comprising a compound disclosed herein, or a pharmaceutically acceptable ester, salt, or prodrug thereof, together with a pharmaceutically acceptable carrier. A compound of the application can be administered as pharmaceutical compositions by any conventional route, in particular enterally, e.g., orally, e.g., in the form of tablets or capsules, or parenterally, e.g., in the form of injectable solutions or suspensions, topically, e.g., in the form of lotions, gels, ointments or creams, or in a nasal or suppository form. Pharmaceutical compositions comprising a compound of the present application in free form or in a pharmaceutically acceptable salt form in association. For example, oral compositions can be tablets or gelatin capsules comprising the active ingredient together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners. Injectable compositions can be aqueous isotonic solutions or suspensions, and suppositories can be prepared from fatty emulsions or suspensions. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances. Suitable formulations for transdermal applications include an effective amount of a compound of the present application with a carrier. A carrier can include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. Matrix transdermal formulations may also be used. Suitable formulations for topical application, e.g., to the skin and eyes, are preferably aqueous solutions, ointments, creams or gels well-known in the art. Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives. The pharmaceutical compositions of the present application comprise a therapeutically effective amount of a compound of the present application formulated together with one or more pharmaceutically acceptable carriers. As used herein, the term “pharmaceutically acceptable carrier” means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The pharmaceutical compositions of this application can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, or as an oral or nasal spray. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, com, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing a compound of this application with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. Solid compositions of a similar type may also be employed as fillers in soft and hard filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. Dosage forms for topical or transdermal administration of a compound of this application include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this application. The ointments, pastes, creams and gels may contain, in addition to an active compound of this application, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Powders and sprays can contain, in addition to a compound of this application, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons. Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. According to the methods of treatment of the present application, disorders are treated or prevented in a subject, such as a human or other animal, by administering to the subject a therapeutically effective amount of a compound of the application, in such amounts and for such time as is necessary to achieve the desired result. The term “therapeutically effective amount” of a compound of the application, as used herein, means a sufficient amount of the compound so as to decrease the symptoms of a disorder in a subject. As is well understood in the medical arts a therapeutically effective amount of a compound of this application will be at a reasonable benefit/risk ratio applicable to any medical treatment. In general, a compound of the application will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents. A therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. Therapeutic amounts or doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents. Upon improvement of a subject's condition, a maintenance dose of a compound, composition or combination of this application may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. The subject may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms. It will be understood, however, that the total daily usage of the compounds and compositions of the present application will be decided by the attending physician within the scope of sound medical judgment. The specific inhibitory dose for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts. The term “pharmaceutical combination” as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that the active ingredients, e.g., a compound of the application and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients, e.g., a compound of the application and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g., the administration of three or more active ingredients. Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylenepolyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes, oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate, agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water, isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator. The protein kinase inhibitors or pharmaceutical salts thereof may be formulated into pharmaceutical compositions for administration to animals or humans. These pharmaceutical compositions, which comprise an amount of the protein inhibitor effective to treat or prevent a protein kinase-mediated condition and a pharmaceutically acceptable carrier, are other embodiments of the present application. In another aspect, the application provides a kit comprising a compound capable of inhibiting kinase activity selected from one or more compounds of disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and instructions for use in treating cancer. In another aspect, the application provides a kit comprising a compound capable of inhibiting ALK activity selected from a compound disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. In a further aspect, the application provides a kit comprising a compound capable of inhibiting SRPK (e.g., SRPK1 and/or SRPK2) activity selected from a compound disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof. The application is further illustrated by the following examples and synthesis schemes, which are not to be construed as limiting this application in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to illustrate certain embodiments and that no limitation to the scope of the application is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present application and/or scope of the appended claims. EXAMPLES Analytical Methods, Materials, and Instrumentation All reactions were monitored by thin layer chromatography (TLC) with 0.25 mm E. Merck pre-coated silica gel plates (60 F254) and Waters LCMS system (Waters 2489 UV/Visible Detector, Waters 3100 Mass, Waters 515 HPLC pump, Waters 2545 Binary Gradient Module, Waters Reagent Manager, Waters 2767 Sample Manager) using SunFire™ C18column (4.6×50 mm, 5 μm particle size): solvent gradient=100% A at 0 min, 1% A at 5 min; solvent A=0.035% TFA in Water; solvent B=0.035% TFA in CH3CN; flow rate: 2.5 mL/min. Purification of reaction products was carried out by flash chromatography using CombiFlash®Rf with Teledyne Isco RediSep®Rf High Performance Gold or Silicycle SiliaSep™ High Performance columns (4 g, 12 g, 24 g, 40 g, 80 g, or 120 g). The purity of all compounds was over 95% and was analyzed with Waters LCMS system.1HNMR and13C NMR spectra were obtained using a Varian Inova-400 (400 MHz for 1H, and 75 MHz for 13C) spectrometer. Chemical shifts are reported relative to chloroform (6=7.24) for1H NMR or dimethyl sulfoxide (6=2.50) for1H NMR and dimethyl sulfoxide (6=39.51) for13C NMR. Data are reported as (br=broad, s=singlet, d=doublet, t=triplet, q=quartet, m=multiplet). Abbreviations used in the following examples and elsewhere herein are: atm atmospherebr broadDIPEA N,N-diisopropylethylamineDMF N,N-dimethylformamideDMSO dimethyl sulfoxideDIEA diisopropylethylaminEtOAc ethyl acetateHCl hydrochloric acidh hour(s)HATU bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluoro-phosphateHPLC high-performance liquid chromatographyLCMS liquid chromatography—mass spectrometrym multipletMeOH methanolMHz megahertzmin minutesMS mass spectrometryNMR nuclear magnetic resonancePd2(dba)3tris(dibenzylideneaceione)dipaliadium(0)Pd(dppf)Cl2bis(diphenylphosphino)ferrocene]palladium(II) dichlorideppm parts per millionTHF tetrahydrofuranTLC thin layer chromatographyTBAF tetra-n-butylammonium fluorideXphos 2-dicyclohexylphosphino-2″,4″,6″-triisopropylbiphenyl Example 1: Synthesis of 9-ethyl-6,6-dimethyl-8-(1-methyl-1H-pyrazol-4-yl)-11-oxo-6,11-dihydro-5H-benzo[b]carbazole-3-carbonitrile (Compound 7) 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (34 mg, 0.16 mmol) and 37 (60 mg, 0.13 mmol) were dissolved in 1,4-dioxane (5 mL) and 2M Na2CO3sat. aq. solution (0.17 mL, 0.34 mmol) and thoroughly degassed. Pd(dppf)C12(6 mg, 0.008 mmol) and t-Butyl XPhos (4 mg, 0.005 mmol) were added and mixture was heated to 100° C. in a sealed vial. After stirring for 1 hour, LC-MS analysis indicated the reaction was finished. The reaction mixture was filtered through celite and purified by reversed-phase HPLC using a gradient of 30-100% CH3CN/H2O with 0.035% TFA to give the desired compound as a beige solid.1H NMR (DMSO-d6, 400 MHz) δ 12.75 (s, 1H), 8.35 (d, J=8 Hz, 1H), 8.11 (s, 1H) 8.07 (s, 1H), 8.01 (S, 1H), 7.79 (d, J=8 Hz, 2H), 7.63 (d, J=8 Hz, 2H), 3.93 (s, 3H), 2.82 (q, J=7.2 Hz, 2H) 1.79 (s, 6H), 1.17 (t, J=7.2 Hz, 3H), MS m/z 395.73 [M+1]. Example 2: Synthesis of 8-(4-(dimethylamino)piperidin-1-yl)-9-ethyl-6,6-dimethyl-11-oxo-6,11-dihydro-5H-benzo[b]carbazole-3-carbonitrile (Compound 17) N,N-dimethylpiperidin-4-amine (30 mg, 0.15 mmol), NaOt-Bu (70 mg, 0.73 mmol) and 37 (40 mg, 0.09 mmol) were dissolved in 1,4-Dioxane (3 mL), and the mixture thoroughly degassed. Pd2(dba)3(5 mg, 0.05 mmol) and tri(o-tolyl) (3 mg, 0.09 mmol) were added. The mixture was heated to 110° C. for 4 hours. LC-MS analysis showed conversion to the desired product. The mixture was filtered and purified by reversed-phase HPLC using a gradient of 10-60% CH3CN/H2O with 0.035% TFA to give the desired compound as a brown solid (18 mg, 45% yield).1H NMR (DMSO-d6, 400 MHz) δ 12.76 (s, 1H), 8.31 (d, J=8 Hz, 2H), 8.04 (s, 1H) 7.98 (s, 1H), 7.58 (d, J=8 Hz, 1H), 7.34 (s, 1H), 6.51 (s, 1H), 4.07 (m, 4H), 2.80 (s, 6H), 3.14 (m, 4H), 2.71 (q, J=7.2 Hz, 2H) 1.74 (s, 6H), 1.23 (t, J=8 Hz, 3H), MS m/z 382.43 [M+1]. Example 3: Synthesis of 9-ethyl-6,6-dimethyl-11-oxo-8-(1H-1,2,3-triazol-5-yl)-6,11-dihydro-5H-benzo[b]carbazole-3-carbonitrile (Compound 13) Ethylnyltrimethylsilane (70 μL, 0.5 mmol) and 37 (200 mg, 0.45 mmol) were dissolved in Diethylamine (2 mL). Triphenylphosphine (15 mg, 0.054 mmol) and CuI (10 mg, 0.05 mmol) were added and the solution degassed. Pd(OAc)2(5 mg, 0.022 mmol) was added and the mixture heated to 90° C. for 4 h. The mixture was filtered through celite and purified by reversed-phase HPLC using a gradient of 40-100% CH3CN/H2O with 0.035% TFA to give the desired compound as a white solid (131 mg, 70% yield). This material was dissolved in THF (5 mL) and TBAF 1M in THF (0.95 mL, 0.95 mmol) was added. The mixture was stirred for 5 h at rt. The reaction was quenched with water and extracted with EtOAc (3×50 mL) washed with brine, dried over MgSO4, and condensed to give the alkyne as a white solid in quantitative yield. TMS-Azide (30 μL, 0.22 mmol) was added to a solution of the alkyne 39 (50 mg, 0.15 mmol) and CuI (3 mg, 0.007 mmol) in a 9:1 mixture of DMF/MeOH (1 mL) and stirred at 100° C. for 4 h. The mixture was filtered through celite and purified by reversed-phase HPLC using a gradient of 40-100% CH3CN/H2O with 0.035% TFA to give the desired compound as a white solid (20 mg, 36% yield).1H NMR (DMSO-d6, 400 MHz) δ 13.15 (s, 1H), 12.06 (br, 1H), 8.31 (d, J=8 Hz, 1H), 8.11 (s, 1H) 8.04 (s, 1H), 7.95 (s, 1H), 7.71 (s, 1H), 7.60 (d, J=7.2 Hz, 1H), 2.91 (q, J=7.2 Hz, 2H) 1.81 (s, 6H), 1.17 (t, J=7.2 Hz, 3H), MS m/z 382.19 [M+1]. Example 4: Synthesis of (R)-8-(1,2-dihydroxyethyl)-9-ethyl-6,6-dimethyl-11-oxo-6,11-dihydro-5H-benzo[b]carbazole-3-carbonitrile (Compound 22) Step 1. 9-ethyl-6,6-dimethyl-11-oxo-8-vinyl-6,11-dihydro-5H-benzo[b]carbazole-3-carbonitrile (40) The procedure used to prepare Compound 7 was used to prepare Compound 40 (32 mg, 68% yield). Step 2. (R)-8-(1,2-dihydroxyethyl)-9-ethyl-6,6-dimethyl-11-oxo-6,11-dihydro-5H-benzo[b]carbazole-3-carbonitrile (Compound 22) AD-mix 13 (206 mg) and Compound 40 (50 mg, 0.15 mmol) were dissolved in a 0° C. solution of H2O (3 mL) and t-BuOH. The mixture was slowly warmed to rt and stirred for 12 h at rt. LC-MS analysis showed complete conversion of the starting material to the desired product. The reaction mixture was filtered and purified by reversed-phase HPLC using a gradient of 20-70% CH3CN/H2O with 0.035% TFA to give the desired compound as a white solid (28 mg, 51% yield).1H NMR (DMSO-d6, 400 MHz) δ 12.75 (s, 1H), 8.32 (d, J=8 Hz, 1H), 7.99 (m, 2H), 7.85 (s, 1H), 7.63 (d, J=8 Hz, 1H), 5.38 (d, J=4 Hz, 1H), 4.87 (m, 2H), 3.48 (m, 2H), 2.75 (m, 2H), 1.75 (s, 6H), 1.25 (t, J=7 Hz, 3H), MS m/z 375.74 [M+1]. Example 5: Synthesis of the Compounds of the Present Application The following compounds in Table 1 were synthesized according to the procedures outlined in Examples 1-4. TABLE 1Cmpd No.MS (m/z) and/or1H NMR data6MS m/z 466.19 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.77 (s, 1H), 8.57(S, 1H), 8.34 (d, J = 8 Hz, 1H), 8.13 (d, J = 8 Hz, 2H), 8.02 (s, 1H), 7.86 (S, 1H),7.64 (d, J = 8 Hz, 1H), 5.21 (s, 2H), 3.01 (s, 3H), 2.89 (s, 3H), 2.83 (q, J = 8 Hz,2H), 1.80 (s, 6H), 1.22 (t, J = 8 Hz, 3H);13C NMR 100 MHz (DMSO-d6) δ179.61, 167.01, 160.63, 147.78, 146.10, 139.58, 138.93, 136.61, 136.16, 131.37,129.83, 128.13, 127.3, 126.22, 125.33, 122.09, 120.42, 116.85, 109.97, 105.15,53.34, 36.74, 36.36, 35.68, 30.39, 26.32, 15.27.8MS m/z 381.65 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 13.6 (s, 1H), 8.34 (d,J = 8 Hz, 1H), 8.11 (s, 1H) 8.01 (s, 1H), 7.97 (s, 1H), 7.78 (s, 1H), 7.60 (d, J =7.2 Hz, 1H), 3.46 (br, 1H), 2.85 (q, J = 7.2 Hz, 2H) 1.79 (s, 6H), 1.22 (t, J = 7.2Hz, 3H).9MS m/z: 381.73 (M + 1);1H NMR (DMSO-d6, 400 MHz) δ 12.78 (s, 1H), 8.33(d, J = 8 Hz, 1H), 8.14 (s, 1H), 8.01 (d, J = 8 Hz, 2H), 7.83 (s, 1H), 7.76 (s, 1H),7.64 (d, J = 8 Hz, 1H), 6.89 (m, 1H), 2.81 (q, J = 8 Hz, 2H), 1.85 (s, 6H), 1.21 (t,J = 8 Hz, 3H).10MS m/z: 382.43 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.87 (s, 1H), 9.25(s, 1H), 9.02 (s, 1H), 8.31 (d, J = 8 Hz, 1H), 8.13 (s, 1H) 7.98 (s, 1H), 7.84 (S,1H), 7.6 (d, J = 8 Hz, 1H), 2.75 (q, J = 7.2 Hz, 2H) 1.76 (s, 6H), 1.17 (t, J = 8Hz, 3H).11MS m/z: 381.48 (M + 1);1H NMR (DMSO-d6, 400 MHz) δ 12.81 (s, 1H), 8.34(d, J = 8 Hz, 1H), 8.15 (s, 1H), 7.96 (s, 1H), 7.90, (s, 1H), 7.70 (s, 1H), 7.61 (d, J =8 Hz, 1H), 7.43 (d, J = 8 Hz, 1H), 7.32 (d, J = 6 Hz, 1H), 2.81 (q, J = 8 Hz, 2),1.81 (s, 6H), 1.26 (t, J = 8 Hz, 3H).12MS m/z: 397.26 (M + 1);1H NMR (DMSO-d6, 400 MHz) δ 12.78 (s, 1H), 8.36(d, J = 8 Hz, 1H), 8.15 (s, 1H), 8.02 (s, 1H), 7.71 (m, 2H), 7.65 (m, 2H), 7.32 (d,J = 6 Hz, 1H), 2.81 (q, J = 8 Hz, 2H), 1.78 (s, 6H), 1.21 (t, J = 8 Hz, 3H).14MS m/z 392.31 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.88 (s, 1H), 8.85(m, 2H), 8.36 (d, J = 8 Hz, 1H), 8.23 (s, 1H) 8.04 (s, 1H), 7.78 (s, 1H), 7.65 (d, J =7.2 Hz, 1H), 2.70 (q, J = 8 Hz, 2H) 1.79 (s, 6H), 1.13 (t, J = 8 Hz, 3H).15MS m/z 392.48 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.85 (s, 1H), 8.78(m, 2H), 8.36 (d, J = 8 Hz, 1H), 8.22 (s, 1H), 8.13 (d, J = 8 Hz, 1H), 8.03 (s,1H), 7.77 (s, 1H), 7.73 (m, 1H), 7.65 (d, J = 7.2 Hz, 1H), 2.66 (q, J = 8 Hz, 2H)1.79 (s, 6H), 1.13 (t, J = 8 Hz, 3H).16MS m/z 393.71 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.83 (s, 1H), 9.28 (s,1H), 8.95 (s, 2H), 8.36 (d, J = 8 Hz, 1H), 8.22 (s, 1H), 8.03 (s, 1H), 7.84 (s, 1H),7.64 (d, J = 7.2 Hz, 1H), 2.67 (q, J = 8 Hz, 2H) 1.8 (s, 6H), 1.13 (t, J = 7.2 Hz,3H).18MS m/z 399.69 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.81 (s, 1H), 8.29 (d,J = 8 Hz, 1H), 8.06 (s, 1H) 7.98 (s, 1H), 7.59 (d, J = 7 Hz, 1H), 7.35 (s, 1H),3.14 (m, 4H), 2.71 (q, J = 7.2 Hz, 2H), 2.49 (m, 4H), 1.74 (s, 6H), 1.25 (t, J = 8Hz, 3H).19MS m/z 413.27 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.75 (s, 1H), 8.27 (d,J = 8 Hz, 1H), 8.06 (s, 1H) 7.96 (s, 1H), 7.58 (d, J = 7 Hz, 1H), 7.35 (s, 1H),3.21 (m, 4H), 2.71 (q, J = 7.2 Hz, 2H), 2.10 (m, 4H), 1.76 (s, 6H), 1.17 (t, J = 8Hz, 3H).20MS m/z 400.49 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.76 (s, 1H), 8.31 (d,J = 8 Hz, 1H), 8.02 (s, 1H) 7.97 (s, 1H), 7.76 (d, J = 8 Hz, 1H), 7.58 (s, 1H),7.34 (S, 1H), 3.78 (m, 2H), 2.98 (m, 2H) 2.71 (q, J = 7.2 Hz, 2H) 1.76 (s, 6H),1.23 (t, J = 8 Hz, 3H).21MS m/z 340.53 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.92 (s, 1H), 8.44 (s,1H), 8.33 (d, J = 8 Hz, 1H), 8.24 (s, 1H) 8.05 (s, 1H), 7.66 (d, J = 8 Hz, 1H),2.92 (q, J = 7.2 Hz, 2H), 1.79 (s, 6H), 1.31 (t, J = 8 Hz, 3H).22MS m/z 375.74 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.75 (s, 1H), 8.32 (d,J = 8 Hz, 1H), 7.99 (m, 2H), 7.85 (s, 1H), 7.63 (d, J = 8 Hz, 1H), 5.38 (d, J = 4Hz, 1H), 4.87 (m. 2H), 3.48 (m, 2H), 2.75 (m, 2H), 1.75 (s, 6H), 1.25 (t, J = 7Hz, 3H).23MS m/z 375.68 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.82 (s, 1H), 8.34 (d.J = 8 Hz, 1H), 8.03 (m, 2H), 7.85 (s, 1H), 7.63 (d, J = 8 Hz, 1H), 5.40 (d, J = 4Hz, 1H), 4.78 (m, 2H), 3.69 (m, 2H), 2.68 (m, 2H), 1.81 (s, 6H), 1.13 (t, J = 7Hz, 3H).24MS m/z 359.43 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.89 (s, 1H), 8.34 (d,J = 8 Hz, 1H), 8.13 (d, J = 8 Hz, 2H), 8.03 (s, 1H), 7.64 (d, J = 8 Hz, 1H), 2.98(q, J = 7.2 Hz, 2H) 1.76 (s, 6H), 1.23 (t, J = 8 Hz, 3H).25MS m/z 440.84 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.82 (s, 1H), 8.30 (d,J = 8 Hz, 1H), 8.03 (m, 3H), 8.03 (s, 1H), 7.64 (d, J = 8 Hz, 1H), 7.19 (d, J = 16Hz, 1H), 6.48 (m, 1H), 3.99 (m 4H), 2.80 (q, J = 8 Hz, 2H) 1.79 (s, 6H), 1.20 (t,J = 7.2 Hz, 3H).26MS m/z 408.29 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.88 (s, 1H), 8.34 (d,J = 8 Hz, 1H), 8.18 (s, 1H), 8.06 (d, J = 7 Hz, 2H), 7.98 (d, J = 8 Hz, 1H), 7.89(br, 1H), 7.73 (s, 1H), 7.64 (d, J = 8 Hz, 1H), 7.03 (d, J = 8 Hz, 1H), 2.68 (q, J =7.2 Hz, 2H), 1.78 (s, 6H), 1.18 (t, J = 7 Hz, 3H).27MS m/z 407.72 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.78 (s, 1H), 11.84(br, 1H), 8.36 (d, J = 8 Hz, 1H), 8.14 (s, 1H), 7.67 (s, 1H), 7.63 (d, J = 8 Hz,1H), 7.55 (d, J = 7.2 Hz, 1H), 7.46 (s, 1H), 6.46 (d, J = 8.2 Hz, 1H), 2.69 (q, J =8 Hz, 2H), 1.78 (s, 6H), 1.17 (t, J = 7 Hz, 3H).28MS m/z: 415.33 [M + 1];1H NMR (400 MHz, DMSO-d6) δ 12.78 (br, 1H), 8.29(d, J = 4 Hz, 1H), 8.05 (s, 1H), 7.99 (s, 1H), 7.58 (d, J = 7.2 Hz, 1H), 7.47 (s,1H), 2.8 (s, 3H), 2.73 (m, 5H), 2.48 (s, 6H), 2.73 (m, 5H), 1.79 (s, 6H), 0.88 (t, J =8 Hz, 3H).29MS m/z 465.84 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.76 (s, 1H), 8.34 (d,J = 8 Hz, 1H), 8.18 (s, 1H), 8.11 (S, 1H), 8.01 (S, 1H), 7.78 (d, J = 8 Hz, 2H),7.62 (d, J = 8 Hz, 1H), 4.49 (q, J = 7.2 Hz, 1H), 3.98 (m, 4H), 3.50 (m 4H), 2.85(q, J = 7.2 Hz, 2H), 1.76 (s, 6H), 1.23 (t, J = 8 Hz, 3H).30MS m/z 464.47 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.82 (s, 1H), 8.33 (d,J = 8 Hz, 1H), 8.13 (d, J = 8 Hz, 2H), 8.03 (S, 1H), 7.87 (s, 1H), 7.79 (s, 1H),7.63 (d, J = 8 Hz, 2H), 4.59 (m, 1H), 3.47 (m, 2H), 3.13 (m, 2H), 2.85 (q, J = 7.2Hz, 2H), 2.26 (m, 4H), 1.79 (s, 6H), 1.23 (t, J = 8 Hz, 3H).31MS m/z: 452.57 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.83 (s, 1H), 8.34(d, J = 8 Hz, 1H), 8.22 (s, 1H), 8.14 (s, 1H), 8.03 (s, 1H), 7.95 (s, 1H), 7.78 (s,1H), 7.63 (d, J = 8 Hz, 1H), 4.21 (m, 2H), 3.65 (m, 2H), 2.86 (s, 6H), 1.79 (s,6H), 1.24 (t, J = 8 Hz, 3H).32MS m/z 466.37 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 13.05 (s, 1H), 8.36 (d,J = 8 Hz, 1H), 8.18 (s, 1H), 8.11 (S, 1H), 8.15 (S, 1H), 8.06 (S, 1H), 7.90 (S,1H), 7.82 (S, 1H), 7.62 (d, J = 8 Hz, 1H), 4.32 (m, 2H), 3.15 (m, 2H), 2.86 (m,5H), 2.27 (m, 2H), 1.83 (s, 6H), 1.26 (t, J = 8 Hz, 3H);13C NMR 100 MHz(DMSO-d6) δ 179.61, 160.69, 150.93, 146.15, 139.55, 139.25, 136.12, 136.26,130.09, 128.13, 127.41, 126.17, 125.25, 122.09, 120.52, 116.95, 109.94, 105.11,54.78, 48.95, 42.70, 36.75, 30.35, 26.31, 25.38, 15.20.33MS m/z 452.59 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.87 (s, 1H), 8.51(S, 1H), 8.31 (d, J = 8 Hz, 1H), 8.13 (d, J = 8 Hz, 2H), 8.02 (s, 1H), 7.86 (S, 1H),7.64 (d, J = 8 Hz, 1H), 6.43 (s, 1H), 5.32 (s, 2H), 3.27 (s, 3H), 2.83 (q, J = 8 Hz,2H), 1.80 (s, 6H), 1.22 (t, J = 8 Hz, 3H).34MS m/z: 438.59 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.86 (s, 1H), 8.63(S, 1H), 8.25 (d, J = 8 Hz, 1H), 8.08 (d, J = 8 Hz, 2H), 7.96 (s, 1H), 7.87 (S, 1H),7.64 (d, J = 8 Hz, 1H), 5.31 (s, 2H), 2.87 (q, J = 8 Hz, 2H), 1.77 (s, 6H), 1.18 (t,J = 8 Hz, 3H)35MS m/z: 494.61 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.76 (s, 1H), 8.33(d, J = 8 Hz, 1H), 8.22 (s, 1H), 8.11 (s, 1H), 8.02 (s, 1H), 7.87 (s, 1H), 7.82 (s,1H), 7.64 (d, J = 8 Hz, 1H), 2.85 (q, J = 8 Hz, 2H), 1.81 (s, 6H), 1.77 (s, 6H),1.19 (t, J = 8 Hz, 3H).36MS m/z 452.68 [M + 1];1H NMR (DMSO-d6, 400 MHz) δ 12.77 (s, 1H), 8.52(S, 1H), 8.34 (d, J = 8 Hz, 1H), 8.13 (d, J = 8 Hz, 2H), 8.02 (s, 1H), 7.86 (S, 1H),7.63 (d, J = 8 Hz, 1H), 2.83 (q, J = 8 Hz, 2H), 2.51 (S, 6H), 1.85 (s, 6H), 1.22 (t,J = 8 Hz, 3H). Example 6: Biochemical Studies Growth and inhibition of growth was assessed by MTS assay and was performed according to previously established methods (Zhou et al., Nature 2009). The MTS assay is a colorimetric method for determining the number of viable cells that is based on the bioreduction of MTS by cells to a formazan product that is soluble in cell culture medium and can be detected spectrophotometrically. Ba/F3cells of activated ALK and different ALK secondary mutations were exposed to treatment for 72 hours and the number of cells used per experiment determined empirically and has been previously established (Zhou et al., Nature 2009). All experimental points were set up in six wells and all experiments were repeated at least three times. The data was graphically displayed using GraphPad Prism version 5.0 for Windows, (GraphPad Software; www.graphpad.com). The curves were fitted using a nonlinear regression model with a sigmoidal dose response. Example 7: Activities Against ALK Mutants in Comparison with Clinical ALK Inhibitors A compound of the present application, together with clinical ALK inhibitors, was tested against a panel of the most common secondary ALK mutants. EML4-ALKWTor secondary mutants transformed Ba/F3cells, or untransduced Ba/F3as cytotoxicity control, were treated with ALK inhibitors in a dose escalation MTS assay and assessed for viability after 72 hours. Average IC50values (nM) (n=3) are shown in Table 2. As shown in Table 2, the G1202R mutant confers resistance to all clinical stage ALK inhibitors. In contrast, Compound 6 displays potent activity against the G1202R mutant as well as the most common reported mutants. TABLE 2wtC1156YF1174LL1196ML1152R1151 TinsG1202RG1269AS1206YUntrans.Alectinib223901697220792>103Ceritinib4116410164274766844457335512Brigatinib531232300142562121921Crizotinib561448154964585732851265927PF-064639221.31.60.2219.03877154.2N/ACompound 622258196107232591 Example 8: Activity Against G1202R Mutant ALK of the Compounds of the Present Application The compounds of the present application were tested against Ba/F3cells in a single point inhibition assay. EML4-ALK or EML4-ALKG1202Rtransformed Ba/F3cells or untransduced Ba/F3control were treated with a single dose (1 μM) of the compounds of the present application. Percent viability of untreated control for each compound was determined by MTS assay after 72 hours. The results are shown in Table 3. TABLE 3Cmpd No.wtG1202RParental Ba/F360071711658011791117102312111681121212104113678814251321513811634150172918181774190.10.1372024682119287022439542345359245739682593103120264125271242622827832900.254301153111343200303300134381291350071360044 Example 9: Activities Against ALK Mutants of the Compounds of the Present Application Compounds that showed potent inhibition of the G1202R mutant without showing potent inhibition of untransduced Ba/F3cells in the single point inhibition assay were tested against a panel of the most common secondary ALK mutants. The compounds of the present application were tested against Ba/F3cells in a dose escalation MTS assay in EML4-ALKWTor secondary mutant transformed Ba/F3cells. Untransduced Ba/F3control was used as a cytotoxicity control. Viability of the cells was determined after 72 hours, and the IC50's (nM) were shown in Table 4. TABLE 4Cmpd.wtC1156YF1174LL1196ML1152R1151TinsG1202RG1269AS1206YUntrans. Ba/F362225819610723259176115084370575044092120010416518645994422284376229460564950918127316792582123555451059116131634345821801867287324131151771886293455321425616093014172221069520347832625519292719961720347850256462978581055357816202017753924304024872622971613714629768312245480548778116133832148199428667142311831356121491219115239857736565811248371162825153591647 Example 10: Inhibition of Proliferation of Cells Bearing ALK Mutants by the Compounds of the Present Application The inhibitory activity of the compounds of the present application, as well as the clinical stage ALK inhibitors Alectinib, Ceritinib (LDK378), AP26113 (Brigatinib) and Crizotinib (Xalkori), were test against different ALK mutants in a panel of cell lines derived from NSCLC (H3122, DFCI76, and DFCI114), and neuroblastoma (Kelly, LAN-1, SH-SYSY (F1174L), SK-N-SH (F1174L), LAN-5 (R1275Q), SMS-KCNR (R1275Q), CHLA-20 (R1275Q), SK-N-BE2 (wt), SK-N-FI (wt), and SK-N-AS (wt) (Table 5 andFIGS.4A,4B, and5). Cells were seeded at 4000 per well in 96 well plates and exposed to each compound in triplicate at 1 nM to 10 μM for 72 hours. Cell viability was evaluated using CellTiter-Glo Luminescent Cell Viability Assay (Promega) following manufacturer's instruction. IC50values were calculated by nonlinear regression (variable slope) using GraphPad Prism 5 software. Each experiment was repeated for at least twice. These selected cell lines showed varied patterns of sensitivity to the growth inhibitory activity of the compounds, which likely reflects a combination of the degree to which the antiproliferative activity is ‘on-target’ to ALK versus other targets of these compounds and the degree to which each of these cell lines are dependent upon ALK kinase activity. For example, Compounds 6 and 32 possessed submicromolar EC50s across the entire panel of cell lines, and Compound 6 showed a marked increase in potency against all of the neuroblastoma cell lines and the ALK™ sensitive H3122 cells. The L1152R EML4-ALK mutant Ba/F3cells were more potently inhibited by Compound 6 than Alectinib (Table 5 andFIGS.6A and6B) possibly due to the fact that in DFCI76 the EML4-ALK activity of Compound 6 was masked by the activation of EGFR signaling, an additional known resistance mechanism in DFCI76. The mutant EGFR PC9 cell line was not inhibited by Compound 6, further demonstrating the on-target effect of this compound. Compound 32 was more potent in the neuroblastoma cell lines than Compound 6. TABLE 5EC50’s of the compounds of the present application against a panel ofNSCLC and neuroblastoma cell lines transduced with ALK mutantsEC50(nM)Cell LineMYCNHistology31326AlectinibLDK378AP26113CrizotinibH3122NSCL1395915532DFCI76NSCL4530195117230233(L1152R)DFCI114NSCL5358634192071891615(G1269A)KellyAmplifiedNeuroblastoma16491147434142127211(F1174L)LAN-1AmplifiedNeuroblastoma494265571200454928531346(F1174L)SH-SY5YNon-AmplifiedNeuroblastoma4512644131150186986523(F1174L)SK-N-SHNon-AmplifiedNeuroblastoma2521612458723031988370(F1174L)LAN-5AmplifiedNeuroblastoma15283192617122790232(R1275Q)SMS-KCNRAmplifiedNeuroblastoma1297413376592535179(R1275Q)CHLA-20Non-AmplifiedNeuroblastoma119922184303638667439(R1275Q)SK-N-BE2AmplifiedNeuroblastoma114975262315545932928710(wt)SK-N-FI (wt)Non-AmplifiedNeuroblastoma914567973240134926451469SK-N-AS (wt)Non-AmplifiedNeuroblastoma871465775213910457761473 TABLE 6IC50's of Compound 6 against Ba/F3 or tumor cells transduced with various ALKmutants in comparison with clinical ALK inhibitorsCrizotinibCeritinibAfectinibCompound 6IC50 (μM)IC50 (μM)IC50 (μM)IC50 (μM)Ba/F3EML4-ALK WT0.050 ± 0.0160.026 ± 0.0140.004 ± 0.0030.004 ± 0.003EML4-ALK G1202R0.315 ± 0.0960.624 ± 0.1590.235 ± 0.0250.003 ± 0.002EML4-ALK C1156Y0.184 ± 0.0390.153 ± 0.0220.008 ± 0.0100.010 ± 0.012EML4-ALK F1174L0.130 ± 0.0450.095 ± 0.0050.009 ± 0.0120.011 ± 0.011EML4-ALK L1196M0.482 ± 0.1020.048 ± 0.0150.132 ± 0.0560.074 ± 0.049EML4-ALK L1152R0.697 ± 0.0622.364 ± 0.5410.350 ± 0.0690.195 ± 0.005EML4-ALK 1151Tins0.724 ± 0.1150.205 ± 0.0520.062 ± 0.0130.079 ± 0.030EML4-ALK G1269A0.553 ± 0.0350.035 ± 0.0190.024 ± 0.0140.015 ± 0.011EML4-ALK S1206Y0.133 ± 0.0590.029 ± 0.0060.008 ± 0.0100.010 ± 0.009CD74-ROS0.017 ± 0.0050.678 ± 0.109del19EGFR0.281 ± 0.001Untransduced1.859 ± 0.0596.064 ± 0.480>100.605 ± 0.013Tumor Cell LineH31220.056 ± 0.0210.013 ± 0.0050.007 ± 0.0040.004 ± 0.002DFC176 (L1152R)0.248 ± 0.0640.092 ± 0.0310.026 ± 0.0270.030 ± 0.034DFCl114 (G1269A)1.697 ± 0.2620.041 ± 0.0290.194 ± 0.0500.876 ± 0.131PC90.595 ± 0.011 Example 11: KINOMEscAN™ Analysis of the Compounds of the Present Application The kinase selectivity of the compounds of the present application was assessed using the KINOMEscAN™ methodology across a panel of 456 kinases (Ambit Biosciences, San Diego, Calif.). Compounds 6 and 32 were screened at a concentration of 1 μM. Both compounds were slightly less selective than Alectinib. Compound 6 was more selective than compound 32 with 34 interactions mapped compared to 39 with an S-score (1)=0.06, which may explain the increase in cytotoxicity against the neuroblastoma cell lines (FIG.3). Dose—response analysis using Compound 6 revealed inhibition of CSNK2A1<10 μM, IRAK1 with an IC50=14 nM, IRAK 4 with an IC50=465 nM, CLK4 with an IC50=14 nM, RET with an IC50=3 nM, RET V804L with an IC50=13 nM, and RET V804M with an IC50=12 nM. Dose—response analysis using compound 32 revealed inhibition of CSNK2A1<10 μM, IRAK1 with an IC50=15 nM, IRAK 4 with an IC50=234 nM, CLK4 with an IC50=4 nM, RET with an IC50=2 nM, RET V804L with an IC50=9 nM, and RET V804M with an IC50=23 nM. Example 12: Pharmacokinetics and CNS Bioavailability of the Compounds of the Present Application The mouse pharmacokinetic profile of Compound 6 demonstrated good oral bioavailability (87% F), a half-life of 1.69 hours, and a plasma exposure of 64,635 (min*ng/mL, AUClast), following an oral dose of 10 mg/kg (Table 7). Additionally, 2 hours after an oral dose of 10 mg/kg, Compound 6 showed a plasma exposure of 0.34 μM, and a brain exposure of 0.03 μM which equates to a brain/plasma concentration ratio of 0.1. Compared to Compound 6, Compound 32 showed lower oral bioavailability (26% F), a half-life of 4.7 hours, and a plasma exposure of 109,909 (min*ng/mL, AUClast), following an oral dose of 10 mg/kg (Table 8). Additionally, 2 hours after an oral dose of 10 mg/kg, Compound 32 showed a plasma exposure of 0.21 μM, and a brain exposure of 0.03 μM which equates to a brain/plasma concentration ratio of 0.14. TABLE 7Pharmacokinetic properties of Compounds 6 and 32DoseAUClastAUCINF_obsAUCCl_obs(mg/T1/2TmaxCmax(min*ng/(μM •(min*ng/(%(mL/min/Vss_obsmatrixroutekg)(hr)(hr)(ng/mL)(μM)mL)hr)mL)extrap)kg)(L/kg)% F6plasmai.v.21.640.083525075.7176985327.567806211.743.550.38—p.o.101.690.833730.8646352.31670363.66154.83—8732plasmai.v.23.060.0810052.16858073.079661912.6823.274.98—p.o.104.790.926401.381099093.9415485929.7167.98—26 TABLE 8In Vivo CNS Availability of Compounds 6 and 32DoseTimeConc.Conc.Cmpdmatrixroute(mg/kg)(hr)(ng/mL)(μM)i.v.229602.066plasmap.o.101590.34i.v.28760.16p.o.10160.04i.v.22190.046brainp.o.10120.03i.v.2820.004p.o.1020.004i.v.221000.2132plasmap.o.10960.21i.v.2220.05p.o.108770.17i.v.2253.80.1232brainp.o.1012.10.03i.v.235.40.08p.o.10828.10.06 Example 13: Molecular Modeling Molecular modeling study based upon the co-crystal structure of ALK with Alectinib (PDB: 3AOX) (Sakamoto, H. et al., Cancer Cell 2011, 19, 679) was performed to assess the structure-activity relationship of inhibition of ALK and/or ALK mutants by the compounds of the present application. The modeling showed that Compound 6 makes the same backbone hinge contact as Alectinib, however, Compound 6 forms two additional hydrogen bond interactions between the guanidine moiety of R1120 and the carbonyl group of the dimethyl acetamide group (FIG.1A). Furthermore, in the G1202R mutant, Compound 6 forms an additional hydrogen bond interaction between the guanidine moiety of R1202 and the nitrogen of the pyrazole ring (FIG.1B). The modeling study predicted that the methylene spacer between the pyrazole ring and the dimethylacetamide moiety is preferable for the carbonyl amide of Compound 6 to interact with the guanidine moiety of R1120. Example 14: SRPK1 Inhibitory Activities of the Compounds of the Present Application, Measured as IC50(nM), are Shown in Table 9 TABLE 9Cmpd.SRPK1 (IC50(nM))SRPK2 (IC50(nM))Alectinib1128.273.0112.491.15N/A105.0914.5130.8847.46142.6113.2162.5312.517>103N/A192.668.57311.916.6364.7523.63735.6N/A3841N/A EQUIVALENTS Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific embodiments described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.
172,517
11858898
DETAILED DESCRIPTION OF THE INVENTION The present invention provides crystalline freebase forms of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (formula I). Surprisingly, the crystalline freebase forms of the invention have been found not to be deliquescent, even when exposed to atmospheric moisture. Additionally, the crystalline freebase forms of the invention have acceptable levels of hygroscopicity and acceptable melting points. For example, the crystalline freebase Form III has a melting point of about 125° C. and the crystalline freebase Form IV has a melting point of about 119° C. Among other uses, the crystalline freebase forms of the invention are useful for preparing pharmaceutical compositions expected to have utility in treating pulmonary disorders. Accordingly, one aspect of the invention relates to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a crystalline freebase of the invention. Definitions When describing the compounds, compositions, methods and processes of the invention, the following terms have the following meanings unless otherwise indicated. Additionally, as used herein, the singular forms “a,” “an” and “the” include the corresponding plural forms unless the context of use clearly dictates otherwise. The terms “comprising”, “including,” and “having” are intended to be inclusive and mean that there may be additional elements other than the listed elements. All numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth used herein are to be understood as being modified in all instances by the term “about,” unless otherwise indicated. Accordingly, the numbers set forth herein are approximations that may vary depending upon the desired properties sought to be obtained by the present invention. At least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each number should at least be construed in light of the reported significant digits and by applying ordinary rounding techniques. Both Form III and Form IV are anhydrous freebase crystal polymorphs. When reference is made to “a crystalline freebase of the invention”, it is understood that the term includes Form III and Form IV. As used herein, the phrase “having the formula” or “having the structure” is not intended to be limiting and is used in the same way that the term “comprising” is commonly used. The term “pharmaceutically acceptable” refers to a material that is not biologically or otherwise unacceptable when used in the invention. For example, the term “pharmaceutically acceptable carrier” refers to a material that can be incorporated into a composition and administered to a patient without causing unacceptable biological effects or interacting in an unacceptable with other components of the composition. Such pharmaceutically acceptable materials typically have met the required standards of toxicological and manufacturing testing, and include those materials identified as suitable inactive ingredients by the U.S. Food and Drug Administration. The term “therapeutically effective amount” means an amount sufficient to effect treatment when administered to a patient in need thereof, i.e., the amount of drug needed to obtain the desired therapeutic effect. For example, a therapeutically effective amount for treating a pulmonary disorder is an amount of compound needed to, for example, reduce, suppress, eliminate or prevent the symptoms of asthma or chronic obstructive pulmonary disease (“COPD”), or to treat the underlying cause of asthma or COPD. In one embodiment, a therapeutically effective amount is that amount needed to produce bronchodilation. On the other hand, the term “effective amount” means an amount sufficient to obtain a desired result, which may not necessarily be a therapeutic result. For example, when studying a system comprising a muscarinic receptor, an “effective amount” may be the amount needed to antagonize the receptor. The term “treating” or “treatment” as used herein means the treating or treatment of a disease or medical condition (such as COPD) in a patient such as a mammal (particularly a human) that includes: (a) preventing the disease or medical condition from occurring, that is, prophylactic treatment of a patient; (b) ameliorating the disease or medical condition such as by eliminating or causing regression of the disease or medical condition in a patient; (c) suppressing the disease or medical condition such as by slowing or arresting the development of the disease or medical condition in a patient; or (d) alleviating the symptoms of the disease or medical condition in a patient. For example, the term “treating COPD” would include preventing COPD from occurring, ameliorating COPD, suppressing COPD, and alleviating the symptoms of COPD. The term “patient” is intended to include those mammals, such as humans, that are in need of treatment or disease prevention or that are presently being treated for disease prevention or treatment of a specific disease or medical condition. The term “patient” also includes test subjects in which compounds of the invention are being evaluated or test subjects being used in a assay, for example an animal model. Synthesis The crystalline freebase forms of the invention can be synthesized from readily available starting materials as described below and in the Examples. While there may be several methods that can be used to produce each crystalline freebase form, it is noted, however, that the crystalline content as well as the habit of the crystals (size and shape) may vary, based partly upon the method of preparation, as well as on the solvent composition. It will be appreciated that while specific process conditions (i.e. crystallization temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. In some instances, reactions or crystallizations were conducted at room temperature and no actual temperature measurement was taken. It is understood that room temperature can be taken to mean a temperature within the range commonly associated with the ambient temperature in a laboratory environment, and will typically be in the range of about 25° C. to about 50° C. In other instances, reactions or crystallizations were conducted at room temperature and the temperature was actually measured and recorded. All weights, volumes and equivalents are relative to the biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl) benzoyl]methylamino}ethyl)piperidin-4-yl ester (or salt form) starting material. Generally, the crystallizations are conducted in a suitable inert diluent or solvent system, examples of which include, but are not limited to, methanol, ethanol, isopropanol, isobutanol, ethyl acetate, acetonitrile, dichloromethane, methyl t-butyl ether, and the like, and mixtures thereof. Upon completion of any of the foregoing crystallizations, the crystalline compounds can be isolated from the reaction mixture by any conventional means such as precipitation, concentration, centrifugation and the like. The biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl) benzoyl]methylamino}ethyl)piperidin-4-yl ester, as well as its salts such as the diphosphate salt, employed in the invention can be readily prepared from commercially available starting materials and reagents using the procedures described in the Examples, or using the procedures described in U.S. Patent Publication No. 2005/0203133 to Mammen et al. and U.S. Patent Publication No. 2007/0112027 to Axt et al. The molar ratios described in the methods of the invention can be readily determined by various methods available to those skilled in the art. For example, such molar ratios can be readily determined by1H NMR. Alternatively, elemental analysis and HPLC methods can be used to determine the molar ratio. Form III Form III crystalline freebase of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoyl-piperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester can be prepared from the ester or the diphosphate salt of the ester. In one embodiment, the Form III crystalline freebase is prepared by contacting biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]-methylamino}ethyl)piperidin-4-yl ester with acetonitrile. Typically, the ratio of milligrams of the ester to total milliliters of acetonitrile is about 100:1, with the acetonitrile being added in two steps. Generally, this reaction is conducted while repeatedly cycling through a temperature range of 0-40° C. The solids are then isolated by vacuum filtration and dried. In another embodiment, the Form III crystalline freebase is prepared using a seed crystal of the Form III crystalline freebase and the diphosphate salt of the ester. This method involves: a) forming a seed crystal of the crystalline freebase Form III; b) dissolving the diphosphate salt of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester in isopropyl acetate and water to form a solution; c) and adding the seed crystal to the solution. More specifically, the diphosphate salt of the ester (1 wt) is slurried in isopropyl acetate (17.5 vol) and water (10 vol) at 20±3° C. under nitrogen. The suspension is warmed to 53±3° C. and 10M NaOH (0.5 vol) is added. The mixture is stirred at that temperature for a short time, then the layers are separated and the basic aqueous layer is removed. Water (5 vol) is added to the organic layer, and stirred. The layers are separated and the water layer is removed. Isopropyl acetate (17.5 vol) is added and about 10 volumes of distillate are collected by atmospheric distillation. This step is repeated with additional isopropyl acetate (10 vol). After the second distillation, the temperature of the clear solution is reduced to 53±3° C., then seeded with a suspension of crystalline freebase Form III (0.005 wt; 0.5 wt %) in isopropyl acetate (0.08 vol). The resulting suspension is stirred at 53±3° C. for at least 2 hours, then cooled to 10±3° C. at an approximate cooling rate of 0.19° C./min. The suspension is stirred at 10±3° C. for at least 2 hours and then is collected by filtration. The resulting filter cake is washed with isopropyl acetate (2×3 volumes) and the product is then dried to yield the Form III crystalline freebase. Form IV In one embodiment, the Form IV crystalline freebase is prepared using a seed crystal of the Form III crystalline freebase. This method involves: a) forming a seed crystal of the crystalline freebase Form III; b) dissolving the crystalline freebase Form III in acetonitrile to form a solution; c) and adding the seed crystal to the solution. Typically, the weight ratio of seed to ester is in the range of about 2:250. Typically, the ratio of grams of crystalline freebase Form III to total milliliters of acetonitrile is within the range of about 2:10 to 3:30, with 2.5:16 being one range. The acetonitrile is usually added in several aliquots. Generally, this reaction is conducted while repeatedly cycling through a temperature range of 0-40° C. The solids are then isolated by vacuum filtration and dried. Crystalline Properties As is well known in the field of powder x-ray diffraction, relative peak heights of powder x-ray diffraction (PXRD) spectra are dependent on a number of factors relating to sample preparation and instrument geometry, while peak positions are relatively insensitive to experimental details. PXRD patterns for the crystalline freebase Form III and Form IV were obtained as set forth in Example 5. Thus, in one embodiment, a crystalline compound of the invention is characterized by a PXRD pattern having certain peak positions. Each crystalline freebase form of the invention exhibits a different PXRD pattern, but with certain common peaks. Thus, in one embodiment, the invention relates to crystalline freebase forms of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl) piperidin-4-yl ester characterized by a powder x-ray diffraction pattern comprising diffraction peaks at 2θ values selected from 6.6±0.1, 13.1±0.1, 18.6±0.1, 19.7±0.1, and 20.2±0.1. In one embodiment, the crystalline freebase Form III is characterized by a powder x-ray diffraction pattern comprising diffraction peaks at 2θ values of 6.6±0.1, 13.1±0.1, 18.6±0.1, 19.7±0.1, and 20.2±0.1; and further characterized by having five or more additional diffraction peaks at 2θ values selected from 8.8±0.1, 10.1±0.1, 11.4±0.1, 11.6±0.1, 14.8±0.1, 15.2±0.1, 16.1±0.1, 16.4±0.1, 16.9±0.1, 17.5±0.1, 18.2±0.1, 19.3±0.1, 19.9±0.1, 20.8±0.1, 21.1±0.1, 21.7±0.1, and 22.3±0.1. In another embodiment, the crystalline freebase Form III is characterized by a powder x-ray diffraction comprising diffraction peaks at 2θ values selected from 6.6±0.1, 11.4±0.1, 13.1±0.1, 16.1±0.1, 17.5±0.1, 18.2±0.1, 18.6±0.1, 19.3±0.1, 19.7±0.1, 19.9±0.1, 20.2±0.1, 20.8±0.1, 21.1±0.1, 21.7±0.1, and 22.3±0.1. In yet another embodiment, the crystalline freebase Form III is characterized by a PXRD pattern in which the peak positions are substantially in accordance with those shown inFIG.1. In one embodiment, the crystalline freebase Form IV is characterized by a powder x-ray diffraction pattern comprising diffraction peaks at 2θ values of 6.6±0.1, 13.1±0.1, 18.6±0.1, 19.7±0.1, and 20.2±0.1; and further characterized by having five or more additional diffraction peaks at 2θ values selected from 10.6±0.1, 15.0±0.1, 16.0±0.1, 17.3±0.1, 17.7±0.1, 20.9±0.1, 21.4±0.1, 22.6±0.1, 24.6±0.1, and 27.8±0.1. In another embodiment, the crystalline freebase Form IV is characterized by a powder x-ray diffraction pattern comprising diffraction peaks at 2θ values selected from 6.6±0.1, 13.1±0.1, 15.0±0.1, 17.3±0.1, 17.7±0.1, 18.6±0.1, 19.7±0.1, 20.2±0.1, 20.9±0.1, 21.4±0.1, and 22.6±0.1. In yet another embodiment, the crystalline freebase Form IV is characterized by a PXRD pattern in which the peak positions are substantially in accordance with those shown inFIG.2. In yet another embodiment, a crystalline freebase of the invention is characterized by a differential scanning calorimetry (DSC) thermogram. DSC thermograms were obtained as set forth in Example 6. Melting points reported herein are estimated on the basis of the melt onset registered during DSC analysis. Thus, in one embodiment, a crystalline compound of the invention is characterized by its DSC thermograph. In one embodiment, the crystalline freebase Form III is characterized by a DSC thermograph which shows an onset of endothermic heat flow at about 123° C. and a melting point of about 125° C., as seen inFIG.4. In another embodiment, the crystalline freebase Form IV is characterized by a DSC thermograph which shows one onset of endothermic heat flow at about 66° C., a second onset of endothermic heat flow at about 119° C., and a melting point of about 119° C., as seen inFIG.5. Thermogravimetric analysis (TGA) was performed on the crystalline freebase forms of the invention as described in Example 6. Thus, in one embodiment, a crystalline freebase is characterized by its TGA trace. In one embodiment, the crystalline freebase Form III is characterized by the TGA trace depicted inFIG.6. In another embodiment, the crystalline freebase Form IV is characterized by the TGA trace depicted inFIG.7. A gravimetric vapor sorption (GVS) assessment was performed on the crystalline freebase forms of the invention as described in Example 7. The crystalline freebase forms of the invention have been demonstrated to have a reversible sorption/desorption profiles with acceptable levels of hygroscopicity. For example, the crystalline freebase Form III showed a reversible water uptake of <2% wt/wt between 0 and 90% relative humidity at 25° C. Additionally, the crystalline freebase Form III has been found to be stable upon exposure to elevated temperature and humidity. These properties of the crystalline freebase forms of the invention are further illustrated in the Examples below. Utility The compound of formula I possesses muscarinic receptor antagonist activity and therefore, a crystalline freebase form of the compound of formula I is expected to be useful for treating medical conditions mediated by muscarinic receptors, i.e., medical conditions that are ameliorated by treatment with a muscarinic receptor antagonist. Such medical conditions include, by way of example, pulmonary disorders or diseases including those associated with reversible airway obstruction such as chronic obstructive pulmonary disease (e.g., chronic and wheezy bronchitis and emphysema), asthma, pulmonary fibrosis, allergic rhinitis, rhinorrhea, and the like. Other medical conditions that can be treated with muscarinic receptor antagonists are genitourinary tract disorders such as overactive bladder or detrusor hyperactivity and their symptoms; gastrointestinal tract disorders such as irritable bowel syndrome, diverticular disease, achalasia, gastrointestinal hypermotility disorders and diarrhea; cardiac arrhythmias such as sinus bradycardia; Parkinson's disease; cognitive disorders such as Alzheimer's disease; dismenorrhea; and the like. Accordingly, in one embodiment, the invention relates to a method for treating a pulmonary disorder, the method comprising administering to a patient a therapeutically effective amount of a crystalline freebase of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoyl piperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester. When used to treat a pulmonary disorder, a crystalline freebase of the invention will typically be administered by inhalation in multiple doses per day, in a single daily dose or a single weekly dose. Generally, the dose for treating a pulmonary disorder will range from about 10 μg/day to 200 μg/day. When administered by inhalation, a crystalline freebase of the invention typically will have the effect of producing bronchodilation. Accordingly, in another embodiment, the invention relates to a method of producing bronchodilation in a patient, the method comprising administering to a patient a bronchodilation-producing amount of a crystalline freebase of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl] methylamino}ethyl)piperidin-4-yl ester. Generally, the therapeutically effective dose for producing bronchodilation will range from about 10 μg/day to 200 μg/day. In one embodiment, the invention relates to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient a therapeutically effective amount of a crystalline freebase of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester. When used to treat a COPD or asthma, a crystalline freebase of the invention will typically be administered by inhalation in multiple doses per day or in a single daily dose. Generally, the dose for treating COPD or asthma will range from about 10 μg/day to 200 μg/day. As used herein, COPD includes chronic obstructive bronchitis and emphysema (see, for example, Barnes, Chronic Obstructive Pulmonary Disease,New England Journal of Medicine343:269-78 (2000)). When used to treat a pulmonary disorder, a crystalline freebase of the invention is optionally administered in combination with other therapeutic agents. Accordingly, in a particular embodiment, the pharmaceutical compositions and methods of the invention further comprise a therapeutically effective amount of a β2-adrenoreceptor agonist, a corticosteroid, a non-steroidal anti-inflammatory agent, or combination thereof. In another embodiment, a crystalline freebase of the invention is used to antagonize a muscarinic receptor in biological system, and a mammal in particular such as mice, rats, guinea pigs, rabbits, dogs, pigs, humans and so forth. In this embodiment, a therapeutically effective amount of a crystalline freebase of the invention is administered to the mammal. If desired, the effects of antagonizing the muscarinic receptor can then determined using conventional procedures and equipment. The properties and utility of a crystalline freebase of the invention, such as the muscarinic receptor antagonizing activity, can be demonstrated using various in vitro and in vivo assays that are well-known to those skilled in the art. For example, representative assays are described in further detail in the following Examples and include by way of illustration and not limitation, assays that measure hM1, hM2, hM3, hM4, and hM5muscarinic receptor binding (for example, as described in Assay 1). Useful functional assays to determine the muscarinic receptor antagonizing activity of a crystalline freebase of the invention include by way of illustration and not limitation, assays that measure ligand-mediated changes in intracellular cyclic adenosine monophosphate (cAMP), ligand-mediated changes in activity of the enzyme adenylyl cyclase (which synthesizes cAMP), ligand-mediated changes in incorporation of guanosine 5′-O-(γ-thio)triphosphate ([35S]GTPγS) into isolated membranes via receptor catalyzed exchange of [35S]GTPγS for GDP, ligand-mediated changes in free intracellular calcium ions (measured, for example, with a fluorescence-linked imaging plate reader or FLIPR® from Molecular Devices, Inc.), and the like. Exemplary assays are described in Assay 2. The crystalline freebase is expected to antagonize or decrease the activation of muscarinic receptors in any of the assays listed above, or assays of a similar nature, and will typically be used in these studies at a concentration ranging from about 0.1-100 nanomolar. Thus, the aforementioned assays are useful in determining the therapeutic utility, for example, the bronchodilating activity, of a crystalline freebase of the invention. Other properties and utilities of a crystalline freebase of the invention can be demonstrated using various in vitro and in vivo assays well-known to those skilled in the art. For example, the in vivo potency of a crystalline freebase can be measured in an animal model such as the Einthoven model. Briefly, the bronchodilator activity of a crystalline freebase is evaluated in an anesthetized animal (the Einthoven model), which uses ventilation pressure as a surrogate measure of airway resistance. See, for example, Einthoven (1892)Pfugers Arch.51:367-445; and Mohammed et al. (2000)Pulm Pharmacol Ther.13(6):287-92, as well as Assay 3 which describes a rat Einthoven model. Another useful in vivo assay is the rat antisialagogue assay (for example, as described in Assay 4). Pharmaceutical Compositions and Formulations A crystalline freebase of the invention is typically administered to a patient in the form of a pharmaceutical composition or formulation. Such pharmaceutical compositions may be administered to the patient by any acceptable route of administration including, but not limited to, inhaled, oral, nasal, topical (including transdermal) and parenteral modes of administration. However, it will be understood by those skilled in the art that, once a crystalline freebase of the invention has been formulated, it may no longer be in crystalline form, i.e., the crystalline freebase may be dissolved in a suitable carrier. Accordingly, in one embodiment, the invention relates to a pharmaceutical composition comprising a pharmaceutically acceptable carrier or excipient and a crystalline freebase of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl] methylamino}ethyl)piperidin-4-yl ester. The pharmaceutical composition may contain other therapeutic and/or formulating agents if desired. The pharmaceutical compositions of the invention typically contain a therapeutically effective amount of a crystalline freebase of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester, as the active agent. Typically, such pharmaceutical compositions will contain from about 0.01 to about 95% by weight of the active agent; including, from about 0.01 to about 30% by weight; such as from about 0.01 to about 10% by weight of the active agent. Any conventional carrier or excipient may be used in the pharmaceutical compositions of the invention. The choice of a particular carrier or excipient, or combination of carriers or excipients, will depend on the mode of administration being used to treat a particular patient or type of medical condition or disease state. In this regard, the preparation of a suitable pharmaceutical composition for a particular mode of administration is well within the scope of those skilled in the pharmaceutical arts. Additionally, the ingredients for such compositions are commercially available from, for example, Sigma, P.O. Box 14508, St. Louis, MO 63178. By way of further illustration, conventional formulation techniques are described inRemington: The Science and Practice of Pharmacy,20thEdition, Lippincott Williams & White, Baltimore, Maryland (2000); and H. C. Ansel et al.,Pharmaceutical Dosage Forms and Drug Delivery Systems,7thEdition, Lippincott Williams & White, Baltimore, Maryland (1999). Representative examples of materials that can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols such as propylene glycol; polyols such as glycerin, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; compressed propellant gases such as chlorofluorocarbons and hydrofluorocarbons; and other non-toxic compatible substances employed in pharmaceutical compositions. The pharmaceutical compositions of the invention are typically prepared by thoroughly and intimately mixing or blending the crystalline freebase with a pharmaceutically acceptable carrier and one or more optional ingredients. If necessary or desired, the resulting uniformly blended mixture can then be shaped or loaded into tablets, capsules, pills, canisters, cartridges, dispensers and the like using conventional procedures and equipment. In one embodiment, the pharmaceutical compositions of the invention are suitable for inhaled administration. Suitable pharmaceutical compositions for inhaled administration will typically be in the form of an aerosol or a powder. Such compositions are generally administered using well-known delivery devices such as a nebulizer inhaler, a metered-dose inhaler (MDI), a dry powder inhaler (DPI) or a similar delivery device. In a specific embodiment of the invention, a pharmaceutical composition comprising the active agent is administered by inhalation using a nebulizer inhaler. Such nebulizer devices typically produce a stream of high velocity air that causes the pharmaceutical composition comprising the active agent to spray as a mist that is carried into the patient's respiratory tract. Accordingly, when formulated for use in a nebulizer inhaler, the crystalline freebase active agent is typically dissolved in a suitable carrier to form a solution. Suitable nebulizer devices include the Respimat® Soft Mist™ Inhaler (Boehringer Ingelheim), the AERx® Pulmonary Delivery System (Aradigm Corp.), and the PARI LC Plus Reusable Nebulizer (Pari GmbH). A representative pharmaceutical composition for use in a nebulizer inhaler comprises an isotonic aqueous solution comprising from about 0.05 μg/mL to about 10 mg/mL of a crystalline freebase of the invention. In one embodiment, the aqueous nebulizer formulation is isotonic. In one embodiment, such a solution has a pH of about 4-6. In a particular embodiment, the aqueous nebulizer formulation is buffered with citrate buffer to a pH of about 5. In another particular embodiment, the aqueous formulation contains from about 0.1 mg/mL to about 1.0 mg/mL free base equivalents of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl) piperidin-4-yl ester. In another specific embodiment of the invention, the pharmaceutical composition comprising the active agent is administered by inhalation using a DPI. Such DPIs typically administer the active agent as a free-flowing powder that is dispersed in a patient's air-stream during inspiration. In order to achieve a free flowing powder, the crystalline freebase active agent is typically formulated with a suitable excipient such as lactose, starch, mannitol, dextrose, polylactic acid, polylactide-co-glycolide, and combinations thereof. Micronization is a common method of reducing crystal size to that suitable for pulmonary delivery. Typically, a crystalline freebase active agent is micronized and combined with a suitable carrier to form a suspension of micronized particles of respirable size, where “micronized particles” or “micronized form” means at least about 90% of the particles have a diameter of less than about 10 μm. Other methods of reducing particle size may also be used such as fine milling, chopping, crushing, grinding, milling, screening, trituration, pulverization, and so forth, as long as the desired particle size can be obtained. A representative pharmaceutical composition for use in a DPI comprises dry lactose having a particle size between about 1 μm and about 100 μm and micronized particles of a crystalline freebase of the invention. Such a dry powder formulation can be made, for example, by combining the lactose with the crystalline freebase active agent and then dry blending the components. Alternatively, if desired, the crystalline freebase active agent can be formulated without an excipient. The pharmaceutical composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device. Examples of DPI delivery devices include Diskhaler (GlaxoSmithKline, Research Triangle Park, NC; see, e.g., U.S. Pat. No. 5,035,237 to Newell et al.); Diskus (GlaxoSmithKline; see, e.g., U.S. Pat. No. 6,378,519 to Davies et al.); Turbuhaler (AstraZeneca, Wilmington, DE; see, e.g., U.S. Pat. No. 4,524,769 to Wetterlin); Rotahaler (GlaxoSmithKline; see, e.g., U.S. Pat. No. 4,353,365 to Hallworth et al.) and Handihaler (Boehringer Ingelheim). Further examples of suitable DPI devices are described in U.S. Pat. No. 5,415,162 to Casper et al., U.S. Pat. No. 5,239,993 to Evans, and U.S. Pat. No. 5,715,810 to Armstrong et al., and references cited therein. The disclosures of the aforementioned patents are incorporated herein by reference in their entirety. In yet another specific embodiment of the invention, a pharmaceutical composition comprising a crystalline freebase active agent is administered by inhalation using an MDI, which typically discharges a measured amount of the active agent using compressed propellant gas. Accordingly, pharmaceutical compositions administered using an MDI typically comprise a solution or suspension of the crystalline freebase active agent in a liquefied propellant. Any suitable liquefied propellant may be employed including chlorofluorocarbons such as CCl3F, and hydrofluoroalkanes (HFAs) such as 1,1,1,2-tetrafluoroethane (HFA 134a) and 1,1,1,2,3,3,3-heptafluoro-n-propane, (HFA 227). Due to concerns about chlorofluorocarbons affecting the ozone layer, formulations containing HFAs are generally preferred. Additional optional components of HFA formulations include co-solvents such as ethanol or pentane, and surfactants such as sorbitan trioleate, oleic acid, lecithin, and glycerin. See, for example, U.S. Pat. No. 5,225,183 to Purewal et al., EP 0717987 A2 (Minnesota Mining and Manufacturing Company), and WO 92/22286 (Minnesota Mining and Manufacturing Company, the disclosures of which are incorporated herein by reference in their entirety. A representative pharmaceutical composition for use in a metered-dose inhaler comprises from about 0.01 to 5% by weight of a freebase crystalline compound of the invention; from about 0 to 20% by weight ethanol; and from about 0 to 5% by weight surfactant; with the remainder being an HFA propellant. Such compositions are typically prepared by adding chilled or pressurized hydrofluoroalkane to a suitable container containing the crystalline freebase active agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the crystalline freebase active agent is micronized and then combined with the propellant. The formulation is then loaded into an aerosol canister, which forms a portion of a metered-dose inhaler device. Examples of metered-dose inhaler devices developed specifically for use with HFA propellants are described in U.S. Pat. No. 6,006,745 to Marecki and U.S. Pat. No. 6,143,277 to Ashurst et al. Alternatively, a suspension formulation can be prepared by spray drying a coating of surfactant on micronized particles of the active agent. See, for example, WO 99/53901 (Glaxo Group Ltd.) and WO 00/61108 (Glaxo Group Ltd.). The disclosures of the aforementioned patents and publications are incorporated herein by reference in their entirety. For additional examples of processes of preparing respirable particles, and formulations and devices suitable for inhalation dosing see U.S. Pat. No. 6,268,533 to Gao et al., U.S. Pat. No. 5,983,956 to Trofast; U.S. Pat. No. 5,874,063 to Briggner et al.; and U.S. Pat. No. 6,221,398 to Jakupovic et al.; and WO 99/55319 (Glaxo Group Ltd.) and WO 00/30614 (AstraZeneca AB); the disclosures of which are incorporated herein by reference in their entirety. In another embodiment, the pharmaceutical compositions of the invention are suitable for oral administration. Suitable pharmaceutical compositions for oral administration may be in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil liquid emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a crystalline freebase of the invention as an active ingredient. The pharmaceutical composition may be packaged in a unit dosage form. When intended for oral administration in a solid dosage form (i.e., as capsules, tablets, pills and the like), a pharmaceutical composition of the invention will typically comprise a crystalline freebase of the invention as the active ingredient and one or more pharmaceutically acceptable carriers such as sodium citrate or dicalcium phosphate. Optionally or alternatively, such solid dosage forms may also comprise: fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders such as carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants such as glycerol; disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and/or sodium carbonate; solution retarding agents such as paraffin; absorption accelerators such as quaternary ammonium compounds; wetting agents such as cetyl alcohol and/or glycerol monostearate; absorbents such as kaolin and/or bentonite clay; lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and/or mixtures thereof; coloring agents; and buffering agents. Release agents, wetting agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the pharmaceutical compositions of the invention. Examples of pharmaceutically acceptable antioxidants include: water-soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfate sodium sulfite and the like; oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and metal-chelating agents such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Coating agents for tablets, capsules, pills and like, include those used for enteric coatings such as cellulose acetate phthalate (CAP), polyvinyl acetate phthalate (PVAP), hydroxypropyl methylcellulose phthalate, methacrylic acid-methacrylic acid ester copolymers, cellulose acetate trimellitate (CAT), carboxymethyl ethyl cellulose (CMEC), hydroxypropyl methyl cellulose acetate succinate (HPMCAS), and the like. If desired, the pharmaceutical compositions of the invention may also be formulated to provide slow or controlled release of the active ingredient using, by way of example, hydroxypropyl methyl cellulose in varying proportions; or other polymer matrices, liposomes and/or microspheres. In addition, the pharmaceutical compositions of the invention may optionally contain opacifying agents and may be formulated so that they release the active ingredient only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The crystalline freebase active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients. Suitable liquid dosage forms for oral administration include, by way of illustration, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. Such liquid dosage forms typically comprise the active ingredient and an inert diluent such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (especially cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Suspensions, in addition to the active ingredient, may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof. A crystalline freebase of the invention can also be administered transdermally using known transdermal delivery systems and excipients. For example, the crystalline freebase can be admixed with permeation enhancers such as propylene glycol, polyethylene glycol monolaurate, azacycloalkan-2-ones and the like, and incorporated into a patch or similar delivery system. Additional excipients including gelling agents, emulsifiers and buffers, may be used in such transdermal compositions if desired. A crystalline freebase of the invention can also be co-administered with other therapeutic agents. This combination therapy involves using the crystalline freebase combined with one or more of these secondary agents, either formulated together (e.g., packaged together in a single formulation) or formulated separately (e.g., packaged as separate unit dosage forms). Methods of formulating multiple agents together in the same formulation or in separate unit dosage forms, are well known in the art. The term “unit dosage form” refers to a physically discrete unit suitable for dosing a patient, i.e., each unit containing a predetermined quantity of a compound of the invention calculated to produce the desired therapeutic effect either alone or in combination with one or more additional units. For example, such unit dosage forms may be capsules, tablets, pills, and the like. The additional therapeutic agent(s) can be selected from other bronchodilators (e.g., PDE3inhibitors, adenosine 2b modulators and β2adrenergic receptor agonists); anti-inflammatory agents (e.g., steroidal anti-inflammatory agents such as corticosteroids; non-steroidal anti-inflammatory agents (NSAIDs), and PDE4inhibitors); other muscarinic receptor antagonists (i.e., antichlolinergic agents); antiinfective agents (e.g., Gram positive and Gram negative antibiotics or antivirals); antihistamines; protease inhibitors; and afferent blockers (e.g., D2agonists and neurokinin modulators). One particular embodiment of the invention relates to a composition comprising (a) a pharmaceutically acceptable carrier and a therapeutically effective amount of a crystalline freebase of the invention; and (b) a pharmaceutically acceptable carrier and a therapeutically effective amount of an agent selected from a steroidal anti-inflammatory agent such as a corticosteroid; a β2adrenergic receptor agonist; a phosphodiesterase-4 inhibitor; or a combination thereof; wherein the crystalline freebase and the agent are formulated together or separately. In another embodiment, (b) is a pharmaceutically acceptable carrier and a therapeutically effective amount of a β2adrenergic receptor agonist and a steroidal anti-inflammatory agent. The secondary agents can be used in the form of pharmaceutically acceptable salts or solvates, and if appropriate, as optically pure stereoisomers. Representative β2adrenergic receptor agonists that can be used in combination with a crystalline freebase of the invention include, but are not limited to, salmeterol, salbutamol, formoterol, salmefamol, fenoterol, terbutaline, albuterol, isoetharine, metaproterenol, bitolterol, pirbuterol, levalbuterol and the like, or pharmaceutically acceptable salts thereof. Other β2adrenergic receptor agonists that can be used include, but are not limited to, 3-(4-{[6-({(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)-phenyl]ethyl}amino)-hexyl]oxy}butyl)benzenesulfonamide and 3-(-3-{[7-({(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl]ethyl}-amino)heptyl]oxy}-propyl)benzenesulfonamide and related compounds described in WO 02/066422 (Glaxo Group Ltd.); 3-[3-(4-{[6-([(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl]ethyl}amino)hexyl]oxy}butyl)-phenyl]imidazolidine-2,4-dione and related compounds described in WO 02/070490 (Glaxo Group Ltd.); 3-(4-{[6-({(2R)-2-[3-(formylamino)-4-hydroxyphenyl]-2-hydroxyethyl}amino)hexyl]oxy}butyl)-benzenesulfonamide, 3-(4-{[6-({(2S)-2-[3-(formylamino)-4-hydroxyphenyl]-2-hydroxyethyl}amino)hexyl]oxy}butyl)-benzenesulfonamide, 3-(4-{[6-({(2R/S)-2-[3-(formylamino)-4-hydroxyphenyl]-2-hydroxyethyl}amino)hexyl]oxy}butyl)-benzenesulfonamide, N-(t-butyl)-3-(4-{[6-({(2R)-2-[3-(formylamino)-4-hydroxyphenyl]-2-hydroxyethyl}amino)hexyl]-oxy}butyl) benzenesulfonamide, N-(tert-butyl)-3-(4-{[6-({(2S)-2-[3-(formylamino)-4-hydroxyphenyl]-2-hydroxyethyl}amino)-hexyl]oxy}butyl)-benzenesulfonamide, N-(tert-butyl)-3-(4-{[6-({(2R/S)-2-[3-(formylamino)-4-hydroxyphenyl]-2-hydroxyethyl}amino)hexyl]-oxy}butyl)benzenesulfonamide and related compounds described in WO 02/076933 (Glaxo Group Ltd.); 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol and related compounds described in WO 03/024439 (Glaxo Group Ltd.); N-{2-[4-((R)-2-hydroxy-2-phenylethylamino)phenyl]ethyl}-(R)-2-hydroxy-2-(3-formamido-4-hydroxyphenyl)ethylamine and related compounds described in U.S. Pat. No. 6,576,793 to Moran et al.; N-{2-[4-(3-phenyl-4-methoxy phenyl)aminophenyl]ethyl}-(R)-2-hydroxy-2-(8-hydroxy-2(1H)-quinolinon-5-yl) ethylamine and related compounds described in U.S. Pat. No. 6,653,323 to Moran et al.; and pharmaceutically acceptable salts thereof. In a particular embodiment, the β2-adrenoreceptor agonist is a crystalline monohydrochloride salt of N-{2-[4-((R)-2-hydroxy-2-phenylethylamino)phenyl]ethyl}-(R)-2-hydroxy-2-(3-formamido-4-hydroxyphenyl) ethylamine. When employed, the 02-adrenoreceptor agonist will be present in the pharmaceutical composition in a therapeutically effective amount. Typically, the β2-adrenoreceptor agonist will be present in an amount sufficient to provide from about 0.05 μg to 500 μg per dose. The disclosures of the aforementioned patents and publications are incorporated herein by reference in their entirety. Representative steroidal anti-inflammatory agents that can be used in combination with a crystalline freebase of the invention include, but are not limited to, methyl prednisolone, prednisolone, dexamethasone, fluticasone propionate, 6α,9α-difluoro-17α-[(2-furanyl carbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxoandrosta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester, 6α,9α-difluoro-11β-hydroxy-16α-methyl-3-oxo-17α-propionyloxy-androsta-1,4-diene-17β-carbothioic acid S-(2-oxo-tetrahydrofuran-3S-yl) ester, beclomethasone esters (e.g., the 17-propionate ester or the 17,21-dipropionate ester), budesonide, flunisolide, mometasone esters (e.g., the furoate ester), triamcinolone acetonide, rofleponide, ciclesonide, butixocort propionate, RPR-106541, ST-126 and the like, or pharmaceutically-acceptable salts thereof. When employed, the steroidal anti-inflammatory agent will be present in the composition in a therapeutically effective amount. Typically, the steroidal anti-inflammatory agent will be present in an amount sufficient to provide from about 0.05 μg to 500 μg per dose. An exemplary combination is a crystalline freebase of the invention, co-administered with salmeterol as the β2adrenergic receptor agonist, and fluticasone propionate as the steroidal anti-inflammatory agent. Another exemplary combination is a e crystalline freebase of the invention, co-administered with a crystalline monohydrochloride salt of N-{2-[4-((R)-2-hydroxy-2-phenylethylamino)phenyl]ethyl}-(R)-2-hydroxy-2-(3-formamido-4-hydroxyphenyl) ethylamine as the 02-adrenoreceptor agonist, and 6α,9α-difluoro-17α-[(2-furanylcarbonyl) oxy]-11β-hydroxy-16α-methyl-3-oxoandrosta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester as the steroidal anti-inflammatory agent. As noted above, these agents can be formulated together or separately. Other suitable combinations include, for example, other anti-inflammatory agents, e.g., NSAIDs (e.g., sodium cromoglycate, nedocromil sodium, and phosphodiesterase (PDE) inhibitors such as theophylline, PDE4 inhibitors and mixed PDE3/PDE4 inhibitors); leukotriene antagonists (e.g., monteleukast); inhibitors of leukotriene synthesis; iNOS inhibitors; protease inhibitors such as tryptase and elastase inhibitors; beta-2 integrin antagonists and adenosine receptor agonists or antagonists (e.g., adenosine 2a agonists); cytokine antagonists (e.g., chemokine antagonists such as, an interleukin antibody (αIL antibody), specifically, an αIL-4 therapy, an αIL-13 therapy, or a combination thereof); or inhibitors of cytokine synthesis. Representative phosphodiesterase-4 (PDE4) inhibitors or mixed PDE3/PDE4 inhibitors that can be used in combination with a crystalline freebase of the invention include, but are not limited to cis 4-cyano-4-(3-cyclopentyloxy-4-methoxyphenyl) cyclohexan-1-carboxylic acid, 2-carbomethoxy-4-cyano-4-(3-cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-1-one; cis-[4-cyano-4-(3-cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-1-ol]; cis-4-cyano-4-[3-(cyclopentyloxy)-4-methoxyphenyl]cyclohexane-1-carboxylic acid and the like, or pharmaceutically acceptable salts thereof. Other representative PDE4 or mixed PDE4/PDE3 inhibitors include AWD-12-281 (elbion); NCS-613 (INSERM); D-4418 (Chiroscience and Schering-Plough); CI-1018 or PD-168787 (Pfizer); benzodioxole compounds described in WO99/16766 (Kyowa Hakko); K-34 (Kyowa Hakko); V-1 1294A (Napp); roflumilast (Byk-Gulden); pthalazinone compounds described in WO99/47505 (Byk-Gulden); Pumafentrine (Byk-Gulden, now Altana); arofylline (Almirall-Prodesfarma); VM554/UM565 (Vernalis); T-440 (Tanabe Seiyaku); and T2585 (Tanabe Seiyaku). Representative muscarinic antagonists (i.e., anticholinergic agents) that can be used in combination with a crystalline freebase of the invention include, but are not limited to, atropine, atropine sulfate, atropine oxide, methylatropine nitrate, homatropine hydrobromide, hyoscyamine (d, l) hydrobromide, scopolamine hydrobromide, ipratropium bromide, oxitropium bromide, tiotropium bromide, methantheline, propantheline bromide, anisotropine methyl bromide, clidinium bromide, copyrrolate (Robinul), isopropamide iodide, mepenzolate bromide, tridihexethyl chloride (Pathilone), hexocyclium methylsulfate, cyclopentolate hydrochloride, tropicamide, trihexyphenidyl hydrochloride, pirenzepine, telenzepine, AF-DX 116 and methoctramine and the like, or a pharmaceutically acceptable salt thereof; or, for those compounds listed as a salt, alternate pharmaceutically acceptable salt thereof. Representative antihistamines (i.e., H1-receptor antagonists) that can be used in combination with a crystalline freebase of the invention include, but are not limited to, ethanolamines such as carbinoxamine maleate, clemastine fumarate, diphenylhydramine hydrochloride and dimenhydrinate; ethylenediamines such as pyrilamine amleate, tripelennamine hydrochloride and tripelennamine citrate; alkylamines such as chlorpheniramine and acrivastine; piperazines such as hydroxyzine hydrochloride, hydroxyzine pamoate, cyclizine hydrochloride, cyclizine lactate, meclizine hydrochloride and cetirizine hydrochloride; piperidines such as astemizole, levocabastine hydrochloride, loratadine or its descarboethoxy analogue, terfenadine and fexofenadine hydrochloride; azelastine hydrochloride; and the like, or a pharmaceutically acceptable salt thereof; or, for those compounds listed as a salt, alternate pharmaceutically acceptable salt thereof. Unless otherwise indicated, exemplary suitable doses for the other therapeutic agents administered in combination with a crystalline freebase of the invention are in the range of about 0.05 μg/day to 100 mg/day. The following formulations illustrate representative pharmaceutical compositions of the invention, as well as exemplary methods of preparation. One or more secondary agents can optionally be formulated with a crystalline freebase of the invention (primary active agent). Alternately, the secondary agents(s) can be formulated separately and co-administered with the primary active agent, either simultaneously or sequentially. For example, in one embodiment, a single dry powder formulation can be manufactured to include both the crystalline freebase of the invention and one or more secondary agents. In another embodiment, one formulation is manufactured to contain the crystalline freebase of the invention and separate formulation(s) are manufactured to contain the secondary agent(s). Such dry powder formulations can then be packaged in separate blister packs and administered with a single DPI device. Exemplary Dry Powder Formulation for Administration by Inhalation 0.2 mg of a crystalline freebase of the invention is micronized and then blended with 25 mg of lactose. The blended mixture is then loaded into a gelatin inhalation cartridge. The contents of the cartridge are administered using a powder inhaler. Exemplary Dry Powder Formulation for Administration by a Dry Powder Inhaler A dry powder is prepared having a bulk formulation ratio of micronized crystalline freebase of the invention (active agent) to lactose of 1:200. The powder is packed into a dry powder inhalation device capable of delivering between about 10 μg and 100 μg of active agent per dose. Exemplary Formulations for Administration by a Metered Dose Inhaler A suspension containing 5 wt % of a crystalline freebase of the invention (active agent) and 0.1 wt % lecithin is prepared by dispersing 10 g of the crystalline freebase as micronized particles with a mean size less than 10 μm in a solution formed from 0.2 g of lecithin dissolved in 200 mL of demineralized water. The suspension is spray dried and the resulting material is micronized to particles having a mean diameter less than 1.5 μm. The particles are loaded into cartridges with pressurized 1,1,1,2-tetrafluoroethane. Alternately, a suspension containing 5 wt % of a crystalline freebase of the invention, 0.5 wt % lecithin, and 0.5 wt % trehalose is prepared by dispersing 5 g of the crystalline freebase as micronized particles with a mean size less than 10 μm in a colloidal solution formed from 0.5 g of trehalose and 0.5 g of lecithin dissolved in 100 mL of demineralized water. The suspension is spray dried and the resulting material is micronized to particles having a mean diameter less than 1.5 μm. The particles are loaded into canisters with pressurized 1,1,1,2-tetrafluoroethane. Exemplary Aqueous Aerosol Formulation for Administration by Nebulizer A pharmaceutical composition is prepared by dissolving 0.5 mg of a crystalline freebase of the invention (active agent) in 1 mL of a 0.9% sodium chloride solution acidified with citric acid. The mixture is stirred and sonicated until the active agent is dissolved. The pH of the solution is adjusted to a value in the range of from 3 to 8 (typically about 5) by the slow addition of NaOH. Exemplary Hard Gelatin Capsule Formulation for Oral Administration The following ingredients are thoroughly blended and then loaded into a hard gelatin capsule: 250 mg of a crystalline freebase of the invention, 200 mg of lactose (spray-dried), and 10 mg of magnesium stearate, for a total of 460 mg of composition per capsule. Exemplary Suspension Formulation for Oral Administration The following ingredients are mixed to form a suspension containing 100 mg of active ingredient per 10 mL of suspension. IngredientsAmounta crystalline freebase of the invention1.0gfumaric acid0.5gsodium chloride2.0gmethyl paraben0.15gpropyl paraben0.05ggranulated sugar25.5gsorbitol (70% solution)12.85gVeegum k (Vanderbilt Co.)1.0gflavoring0.035mLcolorings0.5mgdistilled waterq.s. to 100 mL Exemplary Injectable Formulation The following ingredients are blended and the pH is adjusted to 4±0.5 using 0.5 N HCl or 0.5 N NaOH. IngredientsAmounta crystalline freebase of the invention0.2gsodium acetate buffer solution (0.4M)2.0mLHCl (0.5N) or NaOH (0.5N)q.s. to pH 4water (distilled, sterile)q.s. to 20 mL EXAMPLES The following Preparations and Examples are provided to illustrate specific embodiments of the invention. These specific embodiments, however, are not intended to limit the scope of the invention in any way unless specifically indicated. The following abbreviations have the following meanings unless otherwise indicated and any other abbreviations used herein and not defined have their standard meaning.AC adenylyl cyclaseBSA bovine serum albumincAMP 3′-5′ cyclic adenosine monophosphateCHO Chinese hamster ovarycM5cloned chimpanzee M5 receptorDCM dichloromethanedPBS Dulbecco's phosphate buffered salineEDTA ethylenediaminetetraacetic acidEtOAc ethyl acetateFBS fetal bovine serumFLIPR fluorometric imaging plate readerHBSS Hank's buffered salt solutionHEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acidhM1cloned human M1 receptorhM2cloned human M2 receptorhM3cloned human M3 receptorhM4cloned human M4 receptorhM5cloned human M5 receptorHOBT N-hydroxybenzotriazoleHPLC high-performance liquid chromatographyMCh methylcholineMeCN acetonitrile Any other abbreviations used herein but not defined have their standard, generally accepted meaning. Unless noted otherwise, reagents, starting materials and solvents were purchased from commercial suppliers (such as Sigma-Aldrich, Fluka, and the like) and were used without further purification. Preparation 1 Biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl Ester The diphosphate salt of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (16 g) was dissolved in a biphasic mixture of water (100 mL) and EtOAc (200 mL). NaOH (2 N, 75 mL) was added over a period of 5 minutes. The mixture was then stirred for 30 minutes. The phases were separated and the aqueous phase was extracted with EtOAc (200 mL). The combined organic phases were concentrated. DCM (100 mL) was added, and the mixture evaporated to dryness. The solids were dried in an oven for about 48 hours to yield the title compound (9.6 g). Example 1 Crystalline Freebase of Biphenyl-2-ylcarbamic Acid 1-(2-{[4-(4-Carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl Ester (Form III) Biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (102.4 mg) was dissolved in MeCN (500 μL). The solution was stirred at room temperature for 80 minutes and a white solid precipitate formed. The mixture was placed in the shaker block to thermocycle (0-40° C. in one hour blocks) for 48 hours. A white, dense, immobile solid was observed. MeCN (500 μL) was added to mobilize the slurry. The mixture was then placed back in the shaker block for 2 hours. The solids were isolated by vacuum filtration using a sinter funnel, then placed in the piston dryer at 40° C. under full vacuum for 15.5 hours, to yield 76.85 mg of the title crystalline compound. Example 2 Crystalline Freebase of Biphenyl-2-ylcarbamic Acid 1-(2-{[4-(4-Carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl Ester (Form III) Diphosphate salt of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoyl-piperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (C35H43N5O4·2H3PO4; MW 793.75; 632.9 g) was slurried in isopropyl acetate (11.08 L) and water (6.33 L) at room temperature under nitrogen. The suspension was warmed to 53±3° C. and 10M NaOH (317 mL) was added to the stirred mixture, while maintaining the temperature of the mixture above 50° C. The mixture was stirred for approximately 5 minutes at 53±3° C. before allowing the layers to settle. The layers were then separated and the aqueous layer was removed. Water (3.16 L) was added to the organic layer while maintaining the temperature of the mixture above 50° C. The mixture was stirred for 5 minutes at 53±3° C. before allowing the layers to settle. The layers were separated and the water layer was removed. Isopropyl acetate (6.33 L) was added and then about 10 volumes of distillate were collected by atmospheric distillation. This step was repeated with additional isopropyl acetate (3.2 L). After the second distillation, the temperature of the clear solution was reduced to 53±3° C., then seeded with a suspension of the biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester crystalline freebase (Form III; 3.2 g) in isopropyl acetate (51 mL). The resulting suspension was stirred at 53±3° C. for 2 hours, then cooled to 10±3° C. over 4 hours. The suspension was stirred at 10±3° C. for at least 2 hours and then the solids were collected by filtration. The resulting filter cake was washed with isopropyl acetate (2×1.9 L) and the product was dried in vacuo at 50° C. to yield the title crystalline compound (C35H43N5O4; MW 597.76; 382.5 g, 80.3% yield). Example 3 Recrystallization of Crystalline Freebase of Biphenyl-2-ylcarbamic Acid 1-(2-{[4-(4-Carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl Ester (Form III) Crystalline freebase of biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (Form III; C35H43N5O4; MW 597.76; 372.5 g) was slurried in toluene (5.6 L) at 20±3° C. under nitrogen. The suspension was warmed to 82±3° C., and held at this temperature until complete dissolution was observed. The solution was then clarified into the crystallizer vessel, followed by rinsing with toluene (373 μL). Solids were observed in the crystallizer vessel, and the vessel was re-heated to 82±3° C. to effect dissolution, then cooled to 58±3° C. and seeded with a pre-sonicated (approximately 1 minute) of crystalline freebase (Form III; 1.9 g) in toluene (8 μL). The resulting suspension was allowed to stand at 58±3° C. for at least 4 hours, then cooled to 20±3° C. over 2 hours (approximate cooling rate of 0.33° C./min). The suspension was stirred at 20±3° C. for at least 1 hour, then the solids were collected by filtration. The resulting filter cake was washed with toluene (2×1.2 L) and the product was dried in vacuo at 52±3° C. to yield the title crystalline compound (345.3 g, 92.7% yield). Example 4 Crystalline Freebase of Biphenyl-2-ylcarbamic Acid 1-(2-{[4-(4-Carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl Ester (Form IV) Biphenyl-2-ylcarbamic acid 1-(2-{[4-(4-carbamoylpiperidin-1-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (prepared as described in Preparation 1; 2.5 g) was dissolved in MeCN (10 mL) to yield a viscous oily pale yellow material. Additional MeCN (5 mL) was added to dilute the material. The solution was seeded with crystalline freebase (20 mg; Form III prepared as described in Example 1) and stirred at room temperature for 90 minutes. A large amount of white precipitate (small crystals) was observed. The slurry was analyzed under a polarized light microscope and found to be birefringent. Additional MeCN (3 mL) was added and the slurry was placed in a Metz Syn10 block to thermocycle (0-40° C. in one hour blocks) at 800 rpm overnight. The Metz Syn10 is a 10 position parallel reaction station that is static. Agitation of the solution/slurry was by a cross magnetic stirrer bar. The shaker block was a separate piece of equipment that was heated and cooled by an external Julabo bath. The material was removed at 0° C. It was observed that the slurry had settled out, leaving a pale yellow solution above the white precipitate. The slurry was stirred and placed back in the shaker block to thermocycle. The material was removed at 40° C., and stirred at a high agitation rate at room temperature for 80 minutes. The slurry was again analyzed and found to be birefringent. The filter cake was isolated by vacuum filtration using a sinter funnel. MeCN (3 mL) was used to wet the filter paper and the filter cake was washed with MeCN prior to filtration. The cake was deliquored under vacuum for 40 minutes to yield 2.3 g of a flowing white powder. The material was placed in a piston dryer at 40° C. for 65 hours, to yield 2.2 g of the title crystalline compound as a white powder (99.6% purity). The majority of the Raman spectra of the product was consistent with that of the Form III starting material. However, three shifts were noted: Form IIIProduct878 cm−1881 cm−1775 cm−1772 cm−1485 cm−1488 cm−1 The product was then analyzed by powder X-ray diffraction, differential scanning calorimetry, and thermal gravimetric analysis. It was determined that the product was a different freebase crystalline from than the Form III starting material, and was designated Form IV. Example 5 Powder X-Ray Diffraction Powder X-ray diffraction (PXRD) patterns of the crystalline freebase Forms III (from Example 1) and IV (from Example 4) were acquired on a PANalytical X'Pert Pro powder diffractomer, equipped with an XCelerator detector. The acquisition conditions were radiation: Cu Ku; generator tension: 40 kV; generator current: 45 mA; start angle 2.0° 2θ; end angle 40.0° 2θ, step size: 0.0167° 2θ. The time per step was 31.750 seconds. The sample was prepared by mounting a few milligrams of sample on a Silicon wafer (zero background) plate, resulting in a thin layer of powder. Characteristic peak positions and calculated d-spacings are summarized below, only reporting those peaks with greater than 14% relative intensity. These were calculated from the raw data using Highscore software. The experimental error in the peak positions is approximately ±0.1° 2θ. Relative peak intensities will vary due to preferred orientation. Pos.d-spacingRel. Int.[°2Th.][Å][%]Form III6.613.553.88.810.114.810.18.814.111.47.821.711.67.614.713.16.829.314.86.015.215.25.815.816.15.530.116.45.413.916.95.213.817.55.125.518.24.938.418.64.823.619.34.623.119.74.5100.019.94.573.520.24.422.820.84.372.721.14.251.521.74.121.722.34.031.0Form IV6.613.427.110.68.413.713.16.842.015.05.958.416.05.515.017.35.141.217.75.045.618.64.8100.019.74.581.220.24.429.720.94.234.821.44.174.822.63.934.324.63.618.127.83.216.1 A representative PXRD pattern for the crystalline freebase Form III is shown inFIG.1. A representative PXRD pattern for the crystalline freebase Form IV is shown inFIG.2. Example 6 Thermal Analysis Differential scanning calorimetry (DSC) thermograms of the crystalline freebase Forms III (from Example 1) and IV (from Example 4) were obtained using a TA Instruments calorimeter. The samples were weighed into an aluminum pan, a pan lid placed on top and lightly crimped without sealing the pan. The experiments were conducted using a heating rate of 10° C./min. A representative DSC thermograph for the crystalline freebase Form III is shown inFIG.4. The DSC thermograph demonstrates that Form III is characterized by a DSC thermograph which shows an onset of endothermic heat flow at 123.1° C. (enthalpy 67.7 J/g). A representative DSC thermograph for the crystalline freebase Form IV is shown inFIG.5. The DSC thermograph demonstrates that Form IV is characterized by a DSC thermograph which shows a small endotherm and a main endotherm, i.e., a small first onset of endothermic heat flow occurring at 65.6° C. (enthalpy 0.8 J/g) and a main second onset of endothermic heat flow occurring at 118.8° C. (enthalpy 66.8 J/g). Thermal gravimetric analysis (TGA) data were obtained using a TA Instruments Q500 instrument. The samples were heated in an open aluminium pan at a heating rate of 10° C./min to 200° C. A representative TGA trace for the crystalline freebase Form III is shown inFIG.6, and indicates that negligible weight loss was observed prior to sample degradation. A representative TGA trace for the crystalline freebase Form IV is shown inFIG.7, and indicates that approximately 0.3% weight loss was observed prior to sample melt, which is consistent with the loss of residual solvent. Example 7 Gravimetric Vapor Sorption Assessment Gravimetric vapor sorption (GVS) studies were performed using a Surface Measurements System DVS-1 instrument for generation of full sorption isotherm using water vapor perfusion at 25° C. A sample size of approximately 7 mg was placed into a clean and dry tared sample mesh pan and weighed using the internal balance. The target relative humidity (RH) ranges were from 30% to 90%, then 90% to 0% and 0% to 30% with 10% steps. The point of equilibrium was automatically determined using a 0.02 dm/dt asymptote setting. GVS studies on a sample of the crystalline freebase Form III conducted at 25° C. demonstrated that the material had a low propensity to take up moisture over the range 0% RH to 90% RH. The sample showed a reversible water uptake of <2% w/w between 0 and 90% RH at 25° C. This GVS trace demonstrates that Form III has an acceptable weight gain when exposed to a broad humidity range. Example 8 Micronization Samples of the crystalline freebase Form III were micronized using either an APTM 4” micronizer and the particle size determined buy laser light diffraction. Amt of crystalline materialParticle size of micronized material (μm)input (g)yield (g)X10X50X9060.1150.731.272.695.25 For reference, the particle size of the input crystalline freebase Form III was X10=5.58 μm X50=18.2 μm, and X90=49.7 μm. Micronization yielded particles in the respirable size range. Micronization resulted in a reduction in crystallinity but retained the essential characteristics of the pre-micronized material. No changes were observed in the PXRD after storage for 3 months at 40° C./20% relative humidity, at 40° C./75% relative humidity (uncapped), and at 50° C./ambient humidity. The DSC thermograph for the crystalline freebase Form III showed a sharp melt at 125° C., before and after micronization. There was an additional small thermal event in the micronized material at 87° C., likely due to crystallization. After storage for 3 months at 40° C./20% RH, 40° C./75% RH naked, and 50° C./ambient humidity, the micronized material showed a sharp melt at 125° C. with no evidence of amorphous content. Example 9 Lactose Compatibility Two formulations of the crystalline freebase Form III were evaluated as to stability for 3 months at 40° C./20% relative humidity (RH), at 40° C./75% RH (uncapped), and at 50° C./ambient humidity. 0.08 wt/wt % (10 μg DPI dose) and 2 wt/wt % (250 μg DPI dose) formulations were prepared as a blend with lactose alone, or with lactose and 1 wt/wt % magnesium stearate. The stability of all formulations was found to be acceptable. Example 10 pH Solubility and Stability The crystalline freebase Form III shows good solubility (greater than approximately 2 mg/mL) in media up to pH 7. Solubility in water is 0.66 mg/mL with a natural pH of 8.9. Solubility in simulated lung fluid is 0.46 mg/mL with no change observed between 4 hour and 24 hour solubility measurements. The crystalline freebase Form III solutions are stable in pH 4 and pH 6 buffers for up to 7 days at 50° C. or exposed to light. The solutions are stable in water and saline for 7 days at room temperature, protected from light. Assay 1 Radioligand Binding Assay Membrane Preparation from Cells Expressing hM1, hM2, hM3and hM4Muscarinic Receptor Subtypes CHO cell lines stably expressing cloned human hM1, hM2, hM3and hM4muscarinic receptor subtypes, respectively, were grown to near confluency in medium consisting of HAM's F-12 supplemented with 10% FBS and 250 μg/mL Geneticin. The cells were grown in a 5% CO2, 37° C. incubator and lifted with 2 mM EDTA in dPBS. Cells were collected by 5 minute centrifugation at 650×g, and cell pellets were either stored frozen at −80° C. or membranes were prepared immediately. For membrane preparation, cell pellets were resuspended in lysis buffer and homogenized with a Polytron PT-2100 tissue disrupter (Kinematica AG; 20 seconds×2 bursts). Crude membranes were centrifuged at 40,000×g for 15 minutes at 4° C. The membrane pellet was then resuspended with resuspension buffer and homogenized again with the Polytron tissue disrupter. The protein concentration of the membrane suspension was determined by the method described in Lowry, O. et al.,Journal of Biochemistry193:265 (1951). All membranes were stored frozen in aliquots at −80° C. or used immediately. Aliquots of prepared hM5receptor membranes were purchased directly from Perkin Elmer and stored at −80° C. until use. Radioligand Binding Assay on Muscarinic Receptor Subtypes hM1, hM2, hM3, hM4and hM5 Radioligand binding assays were performed in 96-well microtiter plates in a total assay volume of 1000 μL. CHO cell membranes stably expressing either the hM1, hM2, hM3, hM4or hM5muscarinic subtype were diluted in assay buffer to the following specific target protein concentrations (μg/well): 10 μg for hM1, 10-15 μg for hM2, 10-20 μg for hM3, 10-20 μg for hM4, and 10-12 μg for hM5. The membranes were briefly homogenized using a Polytron tissue disruptor (10 seconds) prior to assay plate addition. Saturation binding studies for determining KDvalues of the radioligand were performed using L-[N-methyl-3H]scopolamine methyl chloride ([3H]-NMS) (TRK666, 84.0 Ci/mmol, Amersham Pharmacia Biotech, Buckinghamshire, England) at concentrations ranging from 0.001 nM to 20 nM. Displacement assays for determination of Kivalues of test compounds were performed with [3H]-NMS at 1 nM and eleven different test compound concentrations. The test compounds were initially dissolved to a concentration of 40 μM in dilution buffer and then serially diluted 5× with dilution buffer to final concentrations ranging from 400 fM to 4 μM. The addition order and volumes to the assay plates were as follows: 825 μL assay buffer with 0.1% BSA, 25 μL radioligand, 100 μL diluted test compound, and 50 μL membranes. Assay plates were incubated for 6 hours at 37° C. Binding reactions were terminated by rapid filtration over GF/B glass fiber filter plates (Perkin Elmer Inc., Wellesley, MA) pre-treated in 0.3% polyethyleneimine (PEI). Filter plates were rinsed three times with wash buffer (10 mM HEPES) to remove unbound radioactivity. Plates were then air dried, and 50 μL Microscint-20 liquid scintillation fluid (PerkinElmer Inc., Wellesley, MA) was added to each well. The plates were then counted in a PerkinElmer Topcount liquid scintillation counter (PerkinElmer Inc., Wellesley, MA). Binding data were analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, Inc., San Diego, CA) using the one-site competition model. Kivalues for test compounds were calculated from observed IC50values and the KDvalue of the radioligand using the Cheng-Prusoff equation (Cheng Y; Prusoff W. H.Biochemical Pharmacology22(23):3099-108 (1973)). K values were converted to pKivalues to determine the geometric mean and 95% confidence intervals. These summary statistics were then converted back to Kivalues for data reporting. In this assay, a lower Kivalue indicates that the test compound has a higher binding affinity for the receptor tested. The compound of formula I was found to have a Kivalue of less than about 5 nM for the M3muscarinic receptor subtype when tested in this or a similar assay. Assay 2 Muscarinic Receptor Functional Potency Assays Blockade of Agonist-Mediated Inhibition of cAMP Accumulation In this assay, the functional potency of a test compound is determined by measuring the ability of the test compound to block oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-K1 cells expressing the hM2receptor. cAMP assays are performed in a radioimmunoassay format using the Flashplate Adenylyl Cyclase Activation Assay System with125I-cAMP (NEN SMP004B, PerkinElmer Life Sciences Inc., Boston, MA), according to the manufacturer's instructions. Cells are rinsed once with dPBS and lifted with Trypsin-EDTA solution (0.05% trypsin/0.53 mM EDTA) as described in Assay 1. The detached cells are washed twice by centrifugation at 650×g for five minutes in 50 mLs dPBS. The cell pellet is then re-suspended in 10 mL dPBS, and the cells are counted with a Coulter Z1 Dual Particle Counter (Beckman Coulter, Fullerton, CA). The cells are centrifuged again at 650×g for five minutes and re-suspended in stimulation buffer to an assay concentration of 1.6×106-2.8×106cells/mL. The test compound is initially dissolved to a concentration of 400 μM in dilution buffer (dPBS supplemented with 1 mg/mL BSA (0.1%)), and then serially diluted with dilution buffer to final molar concentrations ranging from 100:M to 0.1 nM. Oxotremorine is diluted in a similar manner. To measure oxotremorine inhibition of AC activity, 25 μL forskolin (25 μM final concentration diluted in dPBS), 25 μL diluted oxotremorine, and 50 μL cells are added to agonist assay wells. To measure the ability of a test compound to block oxotremorine-inhibited AC activity, 25 μL forskolin and oxotremorine (25 μM and 5 μM final concentrations, respectively, diluted in dPBS) 25 μL diluted test compound, and 50 μL cells are added to remaining assay wells. Reactions are incubated for 10 minutes at 37° C. and stopped by addition of 100 μL ice-cold detection buffer. Plates are sealed, incubated overnight at room temperature and counted the next morning on a PerkinElmer TopCount liquid scintillation counter (PerkinElmer Inc., Wellesley, MA). The amount of cAMP produced (pmol/well) is calculated based on the counts observed for the samples and cAMP standards, as described in the manufacturer's user manual. Data are analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, Inc., San Diego, CA) using the non-linear regression, one-site competition equation. The Cheng-Prusoff equation is used to calculate the Ki, using the EC50of the oxotremorine concentration-response curve and the oxotremorine assay concentration as the KDand [L], respectively. The Kivalues are converted to pKivalues to determine the geometric mean and 95% confidence intervals. These summary statistics are then converted back to Kivalues for data reporting. In this assay, a lower Kivalue indicates that the test compound has a higher functional activity at the receptor tested. The compound of formula I was found to have a Kivalue of less than about 5 nM for blockade of oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-K1 cells expressing the hM2receptor, when tested in this or a similar assay. Blockade of Agonist-Mediated [S]GTPγS-Binding In a second functional assay, the functional potency of test compounds can be determined by measuring the ability of the compounds to block oxotremorine-stimulated [35S]GTPγS binding in CHO-K1 cells expressing the hM2receptor. At the time of use, frozen membranes are thawed and then diluted in assay buffer with a final target tissue concentration of 5-10 μg protein per well. The membranes are briefly homogenized using a Polytron PT-2100 tissue disrupter and then added to the assay plates. The EC90value (effective concentration for 90% maximal response) for stimulation of [35S]GTPγS binding by the agonist oxotremorine is determined in each experiment. To determine the ability of a test compound to inhibit oxotremorine-stimulated [35S]GTPγS binding, the following is added to each well of 96 well plates: 25 μL of assay buffer with [35S]GTPγS (0.4 nM), 25 μL of oxotremorine (EC90) and GDP (3 μM), 25 μL of diluted test compound and 25 μL CHO cell membranes expressing the hM2receptor. The assay plates are then incubated at 37° C. for 60 minutes. The assay plates are filtered over 1% BSA-pretreated GF/B filters using a PerkinElmer 96-well harvester. The plates are rinsed with ice-cold wash buffer for 3×3 seconds and then air or vacuum dried. Microscint-20 scintillation liquid (50 μL) is added to each well, and each plate is sealed and radioactivity counted on a topcounter (PerkinElmer). Data are analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, Inc., San Diego, CA) using the non-linear regression, one-site competition equation. The Cheng-Prusoff equation is used to calculate the Ki, using the IC50values of the concentration-response curve for the test compound and the oxotremorine concentration in the assay as the KDand [L], ligand concentration, respectively. In this assay, a lower Kivalue indicates that the test compound has a higher functional activity at the receptor tested. The compound of formula I was found to have a Kivalue of less than about 5 nM for blockade of oxotremorine-stimulated [35S]GTPγS-binding in CHO-K1 cells expressing the hM2receptor, when tested in this or a similar assay. Blockade of Agonist-Mediated Calcium Release Via FLIPR Assays Muscarinic receptor subtypes (M1, M3and M5receptors), which couple to Gqproteins, activate the phospholipase C (PLC) pathway upon agonist binding to the receptor. As a result, activated PLC hydrolyzes phosphatyl inositol diphosphate (PIP2) to diacylglycerol (DAG) and phosphatidyl-1,4,5-triphosphate (IP3), which in turn generates calcium release from intracellular stores, i.e., endoplasmic and sarcoplasmic reticulum. The FLIPR (Molecular Devices, Sunnyvale, CA) assay capitalizes on this increase in intracellular calcium by using a calcium sensitive dye (Fluo-4AM, Molecular Probes, Eugene, OR) that fluoresces when free calcium binds. This fluorescence event is measured in real time by the FLIPR, which detects the change in fluorescence from a monolayer of cells cloned with human M1and M3, and chimpanzee M5receptors. Antagonist potency can be determined by the ability of antagonists to inhibit agonist-mediated increases in intracellular calcium. For FLIPR calcium stimulation assays, CHO cells stably expressing the hM1, hM3and cM5receptors are seeded into 96-well FLIPR plates the night before the assay is done. Seeded cells are washed twice by Cellwash (MTX Labsystems, Inc.) with FLIPR buffer (10 mM HEPES, pH 7.4, 2 mM calcium chloride, 2.5 mM probenecid in HBSS without calcium and magnesium) to remove growth media and leaving 50 μL/well of FLIPR buffer. The cells are then incubated with 50 μL/well of 4 μM FLUO-4AM (a 2× solution was made) for 40 minutes at 37° C., 5% carbon dioxide. Following the dye incubation period, cells are washed two times with FLIPR buffer, leaving a final volume of 50 μL/well. To determine antagonist potency, the dose-dependent stimulation of intracellular Ca2+release for oxotremorine is first determined so that antagonist potency can later be measured against oxotremorine stimulation at an EC90concentration. Cells are first incubated with compound dilution buffer for 20 minutes, followed by agonist addition, which is performed by the FLIPR. An EC90value for oxotremorine is generated according to the method detailed in the FLIPR measurement and data reduction section below, in conjunction with the formula ECF=((F/100−F){circumflex over ( )}*EC50. An oxotremorine concentration of 3×ECFis prepared in stimulation plates such that an EC90concentration of oxotremorine is added to each well in the antagonist inhibition assay plates. The parameters used for the FLIPR are: exposure length of 0.4 seconds, laser strength of 0.5 watts, excitation wavelength of 488 nm, and emission wavelength of 550 nm. Baseline is determined by measuring the change in fluorescence for 10 seconds prior to addition of agonist. Following agonist stimulation, the FLIPR continuously measures the change of fluorescence every 0.5 to 1 second for 1.5 minutes to capture the maximum fluorescence change. The change of fluorescence is expressed as maximum fluorescence minus baseline fluorescence for each well. The raw data is analyzed against the logarithm of drug concentration by nonlinear regression with GraphPad Prism (GraphPad Software, Inc., San Diego, CA) using the built-in model for sigmoidal dose-response. Antagonist Kivalues are determined by Prism using the oxotremorine EC50value as the KDand the oxotremorine EC90for the ligand concentration according to the Cheng-Prusoff equation (Cheng & Prusoff, 1973). In this assay, a lower Kivalue indicates that the test compound has a higher functional activity at the receptor tested. The compound of formula I was found to have a Kivalue of less than about 5 nM for blockade of agonist-mediated calcium release in CHO cells stably expressing the hM3receptor, when tested in this or a similar assay. Assay 3 Rat Einthoven Assay This in vivo assay is used to assess the bronchoprotective effects of test compounds exhibiting muscarinic receptor antagonist activity. All test compounds are diluted in sterile water and are dosed via the inhalation route (IH). The rats (Sprague-Dawley, male, 250-350 g) are exposed to the aerosol generated from an LC Star Nebulizer Set and driven by a mixture of gases (5% CO2/95% atmospheric air). Each test compound solution is nebulized over a 10 minute time period in a pie shaped dosing chamber capable of holding six rats. At predetermined time points after inhalation of compound, the Einthoven assay is performed. Thirty minutes prior to the start of pulmonary evaluation, the animals are anesthetized with inactin (thiobutabarbital, 120 mg/kg IP). The jugular vein is catheterized with saline filled polyethylene catheters (PE-50) and used to infuse MCh. The trachea is then dissected and cannulated with a 14G needle and used for rat ventilation during pulmonary evaluation. Once surgery is complete, rats are ventilated using a piston respirator set at a stroke volume of 1 ml/100 g body weight but not exceeding 2.5 ml volume, and at a rate of 90 strokes per minute. The changes in pressure that occur with each breath are measured. Baseline values are collected for at least 2.5 minutes then rats are challenged non-cumulatively with 2-fold incremental increases of the bronchoconstrictor MCh (5, 10, 20, 40 and 80 μg/ml). The MCh is infused for 2.5 minutes from a syringe pump at a rate of 2 mL/kg/min. The animals are euthanized upon completion of the studies. Changes in ventilation pressure (cm H20) in treated animals are expressed as % inhibition of MCh response relative to control animals. In this assay, a higher % inhibition value indicates that the test compound has a bronchoprotective effect. The compound of formula I, when tested in this assay at a dose of 100 μg/ml, is expected to exhibit greater than 35% inhibition, possibly greater than 70% inhibition, and even more possibly greater than 90% inhibition. 1.5 hr ID50Determination Standard muscarinic antagonists were evaluated in the rat Einthoven assay 1.5 hrs post-dose. The order of potency (ID50s) for the five standards tested was determined to be: ipratropium (4.4 μg/ml)>tiotropium (6 μg/ml)>des-methyl-tiotropium (12 μg/ml)>glycopyrrolate (15 μg/ml)>LAS-34237 (24 μg/ml). The potency of the test compound is similarly determined at 1.5 hrs post-dose. 6 and 24 hr ID50Determination Standards tiotropium and ipratropium were also evaluated 24 hr and/or 6 hr post-dose in the rat Einthoven assay. Ipratropium (10 and 30 μg/ml) was about 3-fold less potent 6-hr post-dose compared to its 1.5 hr potency. The observed loss of activity at this time point (6 hr) is consistent with its relatively short duration of action in the clinic. Tiotropium showed a slow onset of effect with peak bronchoprotection being achieved 6-hr post-dose. Its 6 hr and 24 hr potency values were not significantly different from each other and were about 2-fold more potent compared to its 1.5 hr potency. The onset of action of the test compound, as well as the 6 and 24 hr potency values, is similarly determined. Assay 4 Rat Antisialagogue Assay Rats (Sprague-Dawley, male, 250-350 g) are dosed, anesthetized and cannulated as described for Assay 3. At predetermined time points and after surgery, animals are placed on their dorsal side at a 20° incline with their head in a downward slope. A pre-weighed gauze pad is inserted in the animal's mouth and the muscarinic agonist pilocarpine (PILO) (3 mg/kg, iv.) is administered. Saliva produced during 10 minutes post-PILO is measured gravimetrically by determining the weight of the gauze pad before and after PILO. Antisialagogue effects are expressed as % inhibition of salivation relative to control animals. 1, 6 and 24 hr ID50Determination The rat antisialagogue assay was developed to assess systemic exposure and calculate the lung selectivity index (LSI) of test compounds. The standard, tiotropium, was evaluated in this model at 1, 6, and 24 hr post-dose. Tiotropium was found to be most potent at inhibiting pilocarpine-induced salivation 6 hrs post dose. This finding is consistent with the peak effects observed in the Einthoven assay. This model is a modified version of the procedure described in Rechter, “Estimation of anticholinergic drug effects in mice by antagonism against pilocarpine-induced salivation”Ata Pharmacol Toxicol24:243-254 (1996). The mean weight of saliva in vehicle-treated animals, at each pre-treatment time, is calculated and used to compute % inhibition of salivation, at the corresponding pre-treatment time, at each dose. Exemplary compounds of the invention that are tested in this assay are expected to exhibit ID50values less than 100 μg/ml (measured at 24 hours), with some compounds expected to exhibit an ID50value less than 30 μg/ml, some less than 20 μg/ml, and some less than 15 μg/ml. The ratio of the anti-sialagogue ID50to bronchoprotective ID50is used to compute the apparent lung selectivity index of the test compound. Generally, compounds having an apparent lung selectivity index greater than about 5 are preferred. While the present invention has been described with reference to specific aspects or embodiments thereof, it will be understood by those of ordinary skilled in the art that various changes can be made or equivalents can be substituted without departing from the true spirit and scope of the invention. Additionally, to the extent permitted by applicable patent statues and regulations, all publications, patents and patent applications cited herein are hereby incorporated by reference in their entirety to the same extent as if each document had been individually incorporated by reference herein.
89,258
11858899
DETAILED DESCRIPTION One skilled in the art will recognize that the various embodiments may be practiced without one or more of the specific details described herein, or with other replacement and/or additional methods, materials, or components. In other instances, well-known structures, materials, or operations are not shown or described in detail herein to avoid obscuring aspects of various embodiments of the invention. Similarly, for purposes of explanation, specific numbers, materials, and configurations are set forth herein in order to provide a thorough understanding of the invention. Furthermore, it is understood that the various embodiments shown in the figures are illustrative representations and are not necessarily drawn to scale. Reference throughout this specification to “one embodiment” or “an embodiment” means that a particular feature, structure, material, or characteristic described in connection with the embodiment is included in at least one embodiment of the invention, but does not denote that they are present in every embodiment. Thus, the appearances of the phrases “in an embodiment” or “in another embodiment” in various places throughout this specification are not necessarily referring to the same embodiment of the invention. Further, “a component” may be representative of one or more components and, thus, may be used herein to mean “at least one.” The term “therapeutically effective amount” means an amount of a compound according to the disclosure which, when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utility. Such an amount would be sufficient to elicit the biological or medical response of a tissue, system, or patient that is sought by a researcher or clinician. The amount of a compound of according to the disclosure which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological activity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of treatment, the type of disease-state or disorder being treated and its severity, drugs used in combination with or coincidentally with the compounds of the disclosure, and the age, body weight, general health, sex, and diet of the patient. Such a therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the prior art, and this disclosure. The present invention relates generally to the complexation of a supramolecular host and guest cocrystal to form a supramolecular assembly. Supramolecular assemblies are generally described as inclusion/host-guest complexes comprised of two or more molecules which bind through non-covalent interactions. In one embodiment, the present invention involves the application of these complexes as supramolecular drugs in the pharmaceutical/cosmetic sciences. To develop a microbiologically potent supramolecular drug, the present invention complexes a macrocyclic host with a guest molecule, such as an NSAID. Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used to treat inflammation, pain, and fever that are associated with bacterial infections. Pirfenidone (5-methyl-1-phenylpyridin-2-one) (“PFD”), an FDA-approved antifibrotic drug used for the treatment of idiopathic pulmonary fibrosis, is also an NSAID. The presence of an anti-bacterial pyridone nucleus, which is found in broad-spectrum fluoroquinolone antibiotics, along with recent studies showing NSAIDs might have antibacterial properties, indicates the potential utility of PFD. An example of the supramolecular assemblies of the present invention are assemblies which contains polyphenolic resorcin[4]arenes as host and pirfenidone for skin conditions such as acne. In another embodiment, calixarene is used as the host with pirfenidone. These assemblies have advantages over other conventional therapies recommended for the treatment of acne vulgaris. These novel 1:1 cocrystal complexes have the potential to overcome the drawbacks and complexity of multiple therapies in acne by providing clinical benefits needed, all in one. These new cocrystals can be used as anti-fibrotic, anti-inflammatory, antioxidant and antimicrobial agents in the prevention, treatment and reversal of acne and post-acne lesions. Through the medical application of the pirfenidone, the formation of skin scars can be inhibited. It can also be used to reduce the redness/inflammation of the skin, whereas resorcin[4]arenes can be used as free radical scavenger to halt the formation of new acne breakouts, and to help repair acne skin. Pirfenidone Pirfenidone (5-methyl-1-phenylpyridin-2-one), a pyridone derivative, is an FDA-approved pharmaceutical active (seeFIG.2A). It is a new drug molecule with antifibrotic and anti-inflammatory effects utilized to treat idiopathic lung fibrosis. It is commercially accessible in the market under the brand name of ESBRIET. Pirfenidone (PFD) shows antifibrotic effect through multiple mechanisms, including attenuation of fibroblast proliferation, differentiation, and related collagen synthesis and regulation of fibrotic growth factors and cytokines. More specifically, it modulates diverse cytokines action, involving TGF-, TNF-, epidermal growth factor, platelet-derived growth factor, VEGF, IGF-1, fibroblast growth factor, interferon-, interleukin (IL)-1, IL-6, and IL-8 and it has shown promising effects in vitro and in vivo settings. Results showed it acts on both the inflammatory and the fibrotic phases. Clinical features of acne include seborrhoea, prevalence of acne bacteria, non-inflammatory lesions, inflammatory lesions and various degrees of scarring. The deep nodular acne lesions are difficult to treat and remain even after acne is treated. So, the present invention should treat post acne lesions based on in vitro and in vivo effects reported in literature for PFD. In addition, the polyphenolic host provide antioxidant effect by reducing oxidative stress which is leading cause of acne breakouts. These beneficial effects of PFD could lead to treatment for other skin diseases/conditions, such as scleroderma, acne, hypertrophic scars. However, PFD is accompanied by elevated liver enzyme levels and severe dermatological and gastrointestinal adverse effects, particularly phototoxicity and skin rash. These side effects have hindered using the medication as a potential topical agent for different skin diseases/conditions. In order to lessen the effects of PFD-induced phototoxicity, it is generally recommended to avoid exposure to sunlight by using photoprotective clothing and sunscreens. The present invention takes an alternative way to protect the photosensitive PFD by complexing with a polyphenolic host. We have found that a chemical or conformational perturbation from a host molecule can influence the mechanism of PFD action, which in turn alters its clinical behavior and associated side effects. The present invention discloses the first cocrystal structure of the PFD with RsCx macrocycle with two different tail lengths. In one embodiment, the present invention assembles a resorcin[4]arene-based drug cocrystal with PFD, in which part of the macrocycle is not just acting as a drug carrier. However, the macrocycle chemistry potentially highlights a noticeable effect on the clinical behavior of the drug molecule. Also, polyphenolic resorcin[4]arenes have anti-bacterial properties, which can be beneficial in treating acne bacteria. Resorcinol Resorcinol[4]arenes represent a class of cyclic polyphenolic compounds obtained from the condensation reaction of resorcinol with several aldehydes in acidic solutions. Interestingly, the flexibility in changes of electron-rich upper-rim bunches and lower-rim alkyl chains with distinctive substituents driven to a wide assortment of tunable host molecules. Amidst cyclic polyphenolic cavitands, resorcinarenes have broadly been inspected in host-guest chemistry due to their conical shape to develop valuable (bio)materials and sensors. A variety of guests, from cationic to neutral molecules, have been found to embed into this cavity, through C—H . . . π, cation . . . π and π . . . π interactions. This ability to act as a host, together with its adaptability and affinity towards hydrogen bonding, makes resorcinol[4]arenes a perfect candidate for cocrystallizations. Despite enjoying omnipresent investigation in chemical studies, cyclic polyphenol-based host-guest chemistry including pharmaceutical actives is undoubtedly in its earliest stages. The present invention uses cyclic poplyphenols like resorcinol[4] for cocrystallization with phototoxic drugs due to their potential to protect chemical induced ROS generation based on the polphenolic antioxidant activity. One objective of the present invention is to generate a cocrystal complex of PFD with a polyphenolic macrocycle host through non-covalent supramolecular interactions for treating acne type skin conditions. One embodiment of the present invention uses RsC1, which is antibacterial in nature. The complex has both anti-acne and anti-bacterial applications. The chemical structure of C-methylresorcin[4]arene (RsC1) is shown inFIG.2B. The chemical structure for RsC1-PFD (1:1) Cocrystal (C46H48 N2 O10) is shown inFIG.2C. In another embodiment, C-butylresorcin[4]arene (RsC4) is used. Calixarene The field of supramolecular chemistry has engaged in relentless development for several years, in which macrocyclic cavitands are crucial units. Familiar illustrations of such synthetic supramolecular cavitands incorporate cyclodextrins, calixarenes and analogs, pillarenes, and cucurbiturils. These cavitands basically contrast in symmetry, shape, and hydrophilicity. Among other macrocycles, the family of macrocycles known as calixarenes has held particular importance due to their bowl-shaped conformation. They are considered to be the model for host-guest binding and self-assembly. Calixarenes are phenol-based macrocycles, whereas their sister derivatives (resorcinarenes and pyrogalloarene) are synthetic polyphenols. The calixarene molecules may be characterized as calix(N)arenes in which N is an integer within the range of 4-8. Thus the calixarenes ranging from calix(4)arene to calix(8)arene and their derivatives can be employed in carrying out the invention. The calixarene molecules may be distally substituted with a substituent selected from the group consisting of methyl, ethyl, propyl, butyl amyl or phenyl groups. Cocrystal The present inventive cocrystal of host and guest is moderately lipophilic in nature based on molecular structure. The lipophilic nature helps in skin permeation for localized effect as required in treating acne topically. Chemically, they are linked by non-covalent bond which helps in dissociation and providing individual biological effects such as antioxidant, anti-inflammatory, anti-bacterial and anti-scarring. The cocrystals of the present invention can be included in a number of formulations, including formulations for the treatment of acne and antibacterial formulations. In an embodiment, a formulation for use according to the invention is suitable for topical or local application to the skin, in particular human skin. The ingredients are combined with a “cosmetically-acceptable topical carrier,” i.e., a carrier for topical use that is capable of having the other ingredients dispersed or dissolved therein, and possessing acceptable properties rendering it safe to use topically. A formulation which is “suitable for” topical or local application may also be adapted for topical or local application. A formulation for use according to the invention may be in the form of a fluid, for example a lotion, cream, ointment, varnish, foam, paste, gel or other viscous or semi-viscous fluid, or a less viscous fluid such as might be used in sprays or aerosols. It may take the form of a solution, suspension or emulsion. It may take the form of a powder or of granules, which may be designed to be added to liquid (e.g. water) prior to use. In an embodiment the formulation is, or may be, applied to a substrate such as a sponge, swab, brush, pad, tissue, cloth, wipe, skin patch or dressing (which includes a bandage, plaster, skin adhesive or other material designed for application to a tissue surface), to facilitate its administration. For use in the treatment of acne, the formulation may for example take the form of a lotion, cream, ointment, varnish, foam, paste or gel or it may be, or be capable of being, applied to a substrate of the type described above. The compositions of the present invention may be prepared using methodology that is well known by an artisan of ordinary skill in the field of cosmetics formulation. The cocrystals of the present invention may be included in formulations at various concentrations, depending on the specific end use and carrier. In one embodiment, the cocrystals comprise from about 2 to about 10 wt % of a formulation. In another embodiment, the cocrystals comprise from about 4 to about 8 wt % of a formulation. In the examples below, host-guest complexation was studied: spectroscopy (solution mix) and crystallography (solvent evaporation in solid-state) techniques using NMR, UV-vis and X-ray to identify the stoichiometry and in-vitro anti-bacterial studies. It was found that the complex between PFD-RsC1 were held together by hydrogen-bonding and hydrophobic interactions and exhibits a superior behavior over the drug alone by improving the MIC value against Gram-positive and negative bacteria. The present invention is useful in a variety of applications, including skin care applications for acne vulgaris via a single treatment. The present invention may also decrease resistance associated with antibiotics use for treatment. The RsC1-PFD complex of the present invention may also be useful on acne bacteria (Propionibacterium acnes). EXAMPLES Example 1 The following 1H NMR and diffusion ordered NMR (DOSY) results were found for PFD, RSC4 and RSC4:PFD (1:1 and 2:1). All samples were dissolved in d3-ACN. All the reagents and solvents involved in these examples were employed as purchased and used without further purification unless otherwise noted. All chemical shifts are reported in ppm with residual solvents or TMS (tetramethylsilane) as the internal standards. A saturated solution of PFD, RSC4 and individual guest with varying ratios of (RSC4:PFD, 1:1 and 2:1) was prepared in d3-ACN with an internal standard of 1 v/v % TMS for NMR measurements. The 1H-NMR spectra were obtained using a 400 MHz NMR spectrometer (Bruker AV-400). All experiments were done at 25° C. To minimize convection in DOSY measurement all samples were placed in 3 mm tubes and the Bruker pulse sequence named “ledbpgp2s” was used with Z gradient strength stepping from 5 to 95% (total 16 data points). The probe's maximum Z gradient strength was 53.5 G/cm. The diffusion delay (d20) was set to either 50 ms or 90 ms and the gradient pulse (p30) was set to 2.2 ms. Diffusion coefficient was obtained by fitting peak intensity as a function of gradient strength using Bruker supplied DOSY2D program. The reported diffusion coefficient was average of multiple experiments. Example 2 Tests were conducted to check the antioxidant/antibacterial activity of macrocycle and host-guest compounds. 5 uL of 0.01M compound, 5 uL (premixed, 0.01M compound+1M Pyrogallol, 1:1) and 5 uL (premixed, 0.01M compound+0.88M H2O2, 1:1) were added on paper discs placed on plates. 5 uL of 1M pyrogallol, 5 uL of 0.88M H2O2 were used as control (oxidants). TABLE 1Stock SolutionSampleWeightVolumeStockCodeCompound(mg)(mL)solutionSolventR1RsC15.4410.01MDMSODPFD9.2650.01MDMSORDRsC1-PFD7.8810.01MDMSO(1:1) CocrystalR4RsC47.1210.01MDMSOP6PgC69.0310.01MDMSOPRP2R2C47.4410.01MDMSOVKVK-28110.01MDMSOPyPyrogallol6.3051MWater Example 3 In the following tables, Decrease/Increase means wrt pure H2O2 or pyrogallol. If ZI decreased (shrink) as compared to pyrogallol and H2O2, then they are having antioxidant activity, and if the zone increases then it is showing oxidant activity. If the pure compound shows ZI, then it means the compound has anti-bacterial activity. SeeFIGS.3A and3B. Zone=mm of zone width around disk. TABLE 2Observations w.r.t. Zone of inhibition (ZI) forStaphylococcus aureus(G + ve) bacteria5 uL of5 uL of5 uL of5 uL of5 uLSample0.01M(Compound +(Compound +0.88Mof 1MCode(a,b,c)CompoundaH2O2)bPyrogallol)cH2O2pyrogallolInference*R4, 1, 2No ZIZI decreasedZI decreasedZIZILow radicalslightlyslightly(4.6 mm)(3.3 mm)scavenger(3.6 mm)(2.3 mm)P6, 3, 4No ZIZI decreasedNoticeableZIZILow radicalslightlydecrease in ZI(4.6 mm)(3.3 mm)scavenger(3.8 mm)(2 mm)VK, 5, 6No ZIZI decreasedZI decreasedZIZILow radicalslightlyslightly(4.6 mm)(3.3 mm)scavenger(3.7 mm)(2.3 mm)*R1, 7, 8Slight ZIZI decreasedZI decreasedZIZIAnti-bacterial(1.2 mm)slightlyslightly(4.6 mm)(3.3 mm)potential/Low(4.2 mm)(2.5 mm)radicalscavengerD, 9, 10No ZIZI decreasedNo ZIZIZIModerateslightly (3.8(4.6 mm)(3.3 mm)radicalmm)scavengerRD, 11, 12Slight ZISignificantNoticeableZIZIAnti-bacterial(1.1 mm)decrease in ZIdecrease in ZI(4.6 mm)(3.3 mm)potential/High(1.7 mm)(1.8 mm)radicalscavengerPR, 13, 14No ZINoticeableZIdecreaseZIZIModerate OHdecrease ZIslightly(4.6 mm)(3.3 mm)radical(2.8 mm)(2.4 mm)scavengerarepresents compoundbrepresents 5 uL (premixed, 0.01M compound + 0.88M H2O2, 1:1)crepresents 5 uL (premixed, 0.01M compound + 1M Pyrogallol, 1:1)*represents that both R1 and R4 compound has main structure but only differs in carbon tail length R1 is a macrocycle which is showing slight anti-bacterial activity againstStaphylococcus aureus(G+ve) bacteria, but when complexed with D (i.e., RD) shows antibacterial as well as antioxidant activity. The drug itself shows low to high free radical scavenging effect in the presence of H2O2 and pyrogallol. R1 and R4 being the same molecule, only differing in tail length, has shown low anti-bacterial activity (R1) to none (R4) respectively. TABLE 3Observations w.r.t. Zone of inhibition (ZI) forPseudomonas aeruginosa(G-ve) bacteria5 uL of5 uL of5 uL of5 uL of5 uLSample0.01M(Compound +(Compound +0.88Mof 1MCode(a,b,c)CompoundaH2O2)bPyrogallol)cH2O2pyrogallolInference*R4, 1, 2No ZIZI decreasedSlight increaseZI (2.7ZI (1.1Low radicalslightlyZImm)mm)scavenger(2.1 mm)(1.4 mm)P6, 3, 4No ZIZI decreasedZI is same asZI (2.7ZI (1.1Low radicalslightlypyrogallol (1.2mm)mm)scavenger(2.2 mm)mm)VK, 5, 6No ZIZI decreasedSignificantZI (2.7ZI (1.1Moderateslightlydecrease in ZImm)mm)radical(2.2 mm)(0.7 mm)scavenger*R1, 7, 8No ZIZI decreasedZI is same asZI (2.7ZI (1.1Low radicalslightlypyrogallol (1.2mm)mm)scavenger(2.1 mm)mm)D, 9, 10No ZIZI is same asZI is same asZI (2.7ZI (1.1No radicalH2O2pyrogallol (1.2mm)mm)scavenger(2.7 mm)mm)effectRD, 11, 12No ZIZI decreasedNo ZIZI (2.7ZI (1.1Moderateslightlymm)mm)radical(2.2 mm)scavengerPR, 13, 14No ZINo ZINo ZIZI (2.7ZI (1.1Highmm)mm)radicalscavenger Decrease/Increase wrt pure H2O2 or pyrogallol. If ZI decreased (shrink) as compared to pyrogallol and H2O2, then they are providing antioxidant activity. SeeFIG.4. R1 is amacrocycle which is showing low antioxidant activity but when complexed with D (i.e., RD) shows increased antioxidant activity. The drug itself shows no free radical scavenging effect in the presence of H2O2 and pyrogallol. TABLE 4Observations w.r.t. Zone of inhibition (ZI) forklebsiella pneumoniae(G -ve) bacteria5 uL of5 uL of5 uL of5 uL of5 uLSample0.01M(Compound +(Compound +0.88Mof 1MCode(a,b,c)CompoundaH2O2)bPyrogallol)cH2O2pyrogallolInference*R4, 1, 2No ZINo ZINo ZIZI (3ZI (0.9High OHmm)mm)radicalscavengerP6, 3, 4No ZIZI decreasedNo ZIZI (3ZI (0.9Moderateslightlymm)mm)radical(2 mm)scavengerVK, 5, 6No ZIZI decreasedNo ZIZI (3ZI (0.9Moderateslightlymm)mm)radical(2.3 mm)scavenger*R1, 7, 8No ZIZI decreasedNo ZIZI (3ZI (0.9Moderateslightlymm)mm)radical(2.4 mm)scavengerD, 9, 10No ZIZI decreasedNo ZIZI (3ZI (0.9Moderateslightlymm)mm)radical(2.2 mm)scavengerRD, 11, 12No ZIZI decreasedNo ZIZI (3ZI (0.9Moderateslightlymm)mm)radical(2.2 mm)scavengerPR, 13, 14No ZIZI decreasedNo ZIZI (3ZI (0.9Moderateslightlymm)mm)radical(2.6 mm)scavenger Decrease/Increase wrt pure H2O2 or pyrogallol. If ZI decreased (shrink) as compared to pyrogallol and H2O2, then they are providing antioxidant activity. SeeFIG.5. TABLE 5Observations w.r.t. Zone of inhibition (ZI) forE. Coli(G-ve) bacteria5 uL of5 uL of5 uL of5 uL of5 uLSample0.01M(Compound +(Compound +0.88Mof 1MCode(a,b,c)CompoundaH2O2)bPyrogallol)cH2O2pyrogallolInference*R4, 1, 2No ZINoticeableNo ZIZI (2.9ZI (0.9Moderatedecrease ZImm)mm)radical(1.7 mm)scavengerP6, 3, 4No ZINoticeableNo ZIZI (2.9ZI (0.9Moderatedecrease ZImm)mm)radical(1.7 mm)scavengerVK, 5, 6No ZINo ZINo ZIZI (2.9ZI (0.9High radicalmm)mm)scavenger*R1, 7, 8No ZINoticeableNo ZIZI (2.9ZI (0.9Moderatedecrease ZImm)mm)radical(1.9 mm)scavengerD, 9, 10No ZINoticeableNo ZIZI (2.9ZI (0.9Moderate OHdecrease ZImm)mm)radical(1.7 mm)scavengerRD, 11, 12No ZISignificantNo ZIZI (2.9ZI (0.9High radicaldecrease inmm)mm)scavengerZI(1.1mm)PR, 13, 14No ZINoticeableNo ZIZI (2.9ZI (0.9High OHdecrease ZImm)mm)radical(2 mm)scavenger Decrease/Increase wrt pure H2O2 or pyrogallol. If ZI decreased (shrink) as compared to pyrogallol and H2O2, then they are providing antioxidant activity. SeeFIG.6. Example 4 3 mg/ml stock solutions of RsC1, PFD and RsC1-PFD were prepared in DMSO (bcoz host soluble in DMSO solution). From the stock, a working solution 4× (highest conc. for each compound in plate) was prepared in LB media. 50 uL of LB media were first added in each well (96 well plate). Then a 4× solution of each respective compound was added to the first row (A4-A12) except A1-A3 (Positive control/bacterial innoculum). Then the 4× was serially diluted up to well G for all 3 compounds. (A-G). Then, a 50 uL aliquot of the 100-fold diluted bacterial inoculum was added in each test well. Individual test concentrations (in triplicate wells in a 96-well plate) for the given compounds were achieved by serial dilution by using LB medium. Total volume in each well was 100 uL. The final-test concentration range for the individual test compounds forC. acnewere as follows: Macrocycle, PFD and RsC1-PFD complex (128-2 ug/mL). The results are shown inFIG.10, which is a bar plot of absorbance (at 600 nm) at various test compound concentrations, showing the percent inhibition of pathogen growth in the presence of decreasing concentrations of the test compounds using a twofold dilution series. RsC1-PFD showed inhibition at 16 μg/mL, whereas RsC1 alone only showed 35% inhibition at the same concentration. Example 5 3 mg/ml stock solutions of PFD, RsC4-PFD (1:1 complex), and RsC4 were prepared in DMSO (bcoz host soluble in DMSO solution). From the stock, a working solution 4× (highest conc. for each compound in plate) was prepared in LB media. 50 uL of LB media was first added in each well (96 well plate). Then a 4× solution of the respective compound was added to the first row (A4-A12) except A1-A3 (Positive control/bacterial innoculum). Then, the 4× was serially diluted up to well G for all 3 compounds. (A-G). Then, a 50 uL aliquot of the 100-fold diluted bacterial inoculum was added in each test well. Individual test concentrations (in triplicate wells in a 96-well plate) for the given compounds were achieved by serial dilution by using LB medium. Total volume in each well is 100 uL. The final-test concentration range for the individual test compounds forC. acneas follows: Macrocycle, PFD and RsC4-PFD complex (128-2 ug/mL). The results are shown inFIG.11. Example 6 3 mg/ml stock solutions of PFD, RsC1, RsC1-PFD (1:1 complex), RsC4, and RsC4-PFD (1:1 complex) were prepared in DMSO (bcoz host soluble in DMSO solution). From the stock, a working solution 4× (highest conc. for each compound in plate) was prepared in LB media. 50 uL of LB media was first added in each well (96 well plate). Then a 4× solution of the respective compound was added to the first row (A4-A12) except A1-A3 (Positive control/bacterial innoculum). Then, the 4× was serially diluted up to well G for all 3 compounds. (A-G). Then, a 50 uL aliquot of the 100-fold diluted bacterial inoculum was added in each test well. Individual test concentrations (in triplicate wells in a 96-well plate) for the given compounds were achieved by serial dilution by using LB medium. Total volume in each well is 100 uL. The final-test concentration range for the individual test compounds forC. acnes, S. aureusandP. aeruginosaare as follows: PFD, RsC1, RsC4, RsC1-PFD complex and RsC4-PFD complex (128-2 ug/mL). The results are shown in Table 6. TABLE 6Antibacterial activities of cocrystals and their individual componentsTestMIC (μg/mL); GI (%)compoundS. aureus(MU50)P. aeruginosa(BAMF)C. acnesPFD128; 45128; 35128; 98RsC116; 9864; 4764; 96RsC1-PFD (1)8; 99128; 4316; 93RsC4128; 5264; 56128; 98RsC4-PFD (2)128; 26128; 39128; 99C. acnes:Cutibacterium acnes;S. aureus:Staphylococcus aureus;P. aeruginosa:Pseudomonas aeruginosa;MIC: Minimum inhibitory concentration;GI: Growth inhibition.PFD: pirfenidone;RsC1: C-methylresorcin[4]arene;1:1 cocrystal of RsC1-PFD (1);RsC4: C-butylresorcin[4]arene;1:1 cocrystal of RsC4-PFD (2). FIG.1Ashows a Job's plot constructed from the chemical shift change (Δδ) of the phenyl ring protons (#2) of PFD in 1H NMR spectra by varying the ratio between PFD and RsC4.FIG.1Bshows a Job's plot constructed from the chemical shift change (Δδ) of the aromatic protons (#C) of RsC4 in 1H NMR spectra by varying the ratio between PFD and RsC4. Diffusion coefficient of PFD in equimolar mixture has changed drastically compared to PFD alone (above spectra,FIGS.1A and1B) which indicates the PFD bind to RsC4 in the mixture and diffuses at slower rate compared to PFD alone. The interaction between host and guest were loose/weak, therefore RsC4 and PFD peaks do not correspond to the same diffusion constant and have different position on the y axis. The diffusion coefficient of solvent ACN-d3 is 3.17×10-9. The broad, mobile OH proton at 7.7 ppm of host RsC4 has disappeared in the spectra. The y-axis is shown as log D. FIG.7is a graph showing the antibacterial activity of PFD alone and RsC1-PFD complex againstS. aureus. The percent on each bar indicates growth inhibition at a specific concentration (minimum inhibitory concentration).FIG.8is a graph showing the antibacterial activity of PFD alone and RsC1-PFD complex againstP. aeruginosa. The percent on each bar indicates growth inhibition at a specific concentration (minimum inhibitory concentration).FIG.9is a Job's plot (NMR titration), Stoichiometry 1:1. All documents cited are incorporated herein by reference; the citation of any document is not to be construed as an admission that it is prior art with respect to the present invention. It is to be further understood that where descriptions of various embodiments use the term “comprising,” and/or “including” those skilled in the art would understand that in some specific instances, an embodiment can be alternatively described using language “consisting essentially of” or “consisting of.” While particular embodiments of the present invention have been illustrated and described, it would be obvious to one skilled in the art that various other changes and modifications can be made without departing from the spirit and scope of the invention. It is therefore intended to cover in the appended claims all such changes and modifications that are within the scope of this invention.
27,791
11858900
DETAILED DESCRIPTION OF THE APPLICATION For purposes of the present application, the following definitions will be used (unless expressly stated otherwise): The term “a compound of the application” or “compounds of the application” refers to any compound disclosed herein, e.g., a compound of any of the formulae described herein, including formulae A, I, Ia, II, IIIa-IIIc, IVa-IVc, V, VI, and VII, and/or an individual compound explicitly disclosed herein. Whenever the term is used in the context of the present application it is to be understood that the reference is being made to the free base, a deuterium labeled compound, and the corresponding pharmaceutically acceptable salts or solvates thereof, provided that such is possible and/or appropriate under the circumstances. The term “pharmaceutical” or “pharmaceutically acceptable” when used herein as an adjective, means substantially non-toxic and substantially non-deleterious to the recipient. By “pharmaceutical formulation” it is further meant that the carrier, solvent, excipient, and salt must be compatible with the active ingredient of the formulation (e.g., a compound of the application). It is understood by those of ordinary skill in this art that the terms “pharmaceutical formulation” and “pharmaceutical composition” are generally interchangeable, and they are so used for the purposes of this application. Some of the compounds of the present application may exist in unsolvated as well as solvated forms such as, for example, hydrates. “Solvate” means a solvent addition form that contains either a stoichiometric or non stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate, when the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one of the substances in which the water retains its molecular state as H2O, such combination being able to form one or more hydrate. In the hydrates, the water molecules are attached through secondary valencies by intermolecular forces, in particular hydrogen bridges. Solid hydrates contain water as so-called crystal water in stoichiometric ratios, where the water molecules do not have to be equivalent with respect to their binding state. Examples of hydrates are sesquihydrates, monohydrates, dihydrates or trihydrates. Equally suitable are the hydrates of salts of the compounds of the application. Physiologically acceptable, i.e., pharmaceutically compatible or pharmaceutically acceptable, salts can be salts of the compounds of the application with inorganic or organic acids. Preference is given to salts with inorganic acids, such as, for example, hydrochloric acid, hydrobromic acid, phosphoric acid or sulphuric acid, or to salts with organic carboxylic or sulphonic acids, such as, for example, acetic acid, trifluoroacetic acid, propionic acid, maleic acid, fumaric acid, malic acid, citric acid, tartaric acid, lactic acid, benzoic acid, or methanesulphonic acid, ethanesulphonic acid, benzenesulphonic acid, toluenesulphonic acid or naphthalenedisulphonic acid. Other pharmaceutically compatible salts which may be mentioned are salts with customary bases, such as, for example, alkali metal salts (for example sodium or potassium salts), alkaline earth metal salts (for example calcium or magnesium salts) or ammonium salts, derived from ammonia or organic amines, such as, for example, diethylamine, triethylamine, ethyldiisopropylamine, procaine, dibenzylamine, N-methylmorpholine, dihydroabietylamine or methylpiperidine. Representative salts include the following: acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt, oleate, oxalate, pamottle (embonate), palmitate, pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, sulfate, subacetate, succinate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate. The compounds of the application may contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers. It is intended that all of the possible optical isomers and diastereomers in mixtures and as pure or partially purified compounds are included within the ambit of the application. The application is meant to comprehend all such isomeric forms of these compounds. The independent syntheses of these diastereomers or their chromatographic separations may be achieved as known in the art by appropriate modification of the methodology disclosed herein. Their absolute stereochemistry may be determined by the X-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. In the present specification, the structural formula of the compound represents a certain isomer for convenience in some cases, but the present application includes all isomers, such as geometrical isomers, optical isomers based on an asymmetrical carbon, stereoisomers, tautomers, and the like. “Isomerism” means compounds that have identical molecular formulae but differ in the sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers”. Stereoisomers that are not mirror images of one another are termed “diastereoisomers”, and stereoisomers that are non-superimposable mirror images of each other are termed “enantiomers” or sometimes optical isomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a “racemic mixture”. “Chiral isomer” means a compound with at least one chiral center. Compounds with more than one chiral center may exist either as an individual diastereomer or as a mixture of diastereomers, termed “diastereomeric mixture”. When one chiral center is present, a stereoisomer may be characterized by the absolute configuration (R or S) of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al.,Angew. Chem. Inter. Edit.1966, 5, 385; errata 511; Cahn et al.,Angew. Chem.1966, 78, 413; Cahn and Ingold,J. Chem. Soc.1951 (London), 612; Cahn et al.,Experientia1956, 12, 81; Cahn,J. Chem. Educ.1964, 41, 116). “Geometric isomer” means the diastereomers that owe their existence to hindered rotation about double bonds. These configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold-Prelog rules. Furthermore, the structures and other compounds discussed in this application include all atropic isomers thereof. “Atropic isomers” are a type of stereoisomer in which the atoms of two isomers are arranged differently in space. Atropic isomers owe their existence to a restricted rotation caused by hindrance of rotation of large groups about a central bond. Such atropic isomers typically exist as a mixture, however as a result of recent advances in chromatography techniques; it has been possible to separate mixtures of two atropic isomers in select cases. “Tautomer” is one of two or more structural isomers that exist in equilibrium and is readily converted from one isomeric form to another. This conversion results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. Tautomers exist as a mixture of a tautomeric set in solution. In solid form, usually one tautomer predominates. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent and pH. The concept of tautomers that are interconvertable by tautomerizations is called tautomerism. Of the various types of tautomerism that are possible, two are commonly observed. In keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs. Ring-chain tautomerism arises as a result of the aldehyde group (—CHO) in a sugar chain molecule reacting with one of the hydroxy groups (—OH) in the same molecule to give it a cyclic (ring-shaped) form as exhibited by glucose. Common tautomeric pairs are: ketone-enol, amide-nitrile, lactam-lactim, amide-imidic acid tautomerism in heterocyclic rings (e.g., in nucleobases such as guanine, thymine and cytosine), amine-enamine and enamine-enamine. In one example, are tautomers to each other. It is to be understood that the compounds of the present application may be depicted as different tautomers. It should also be understood that when compounds have tautomeric forms, all tautomeric forms are intended to be included in the scope of the present application, and the naming of the compounds does not exclude any tautomer form. If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated. The separation can be carried out by methods well known in the art, such as contacting a racemic mixture of compounds with an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography. The diastereomeric mixture is often a mixture of diasteriomeric salts formed by contacting a racemic mixture of compounds with an enantiomerically pure acid or base. The diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue. The racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which are well known in the art. The application also includes one or more metabolites of a compound of the application. The present application also comprehends deuterium labeled compounds of each of the formulae described herein or the individual compounds specifically disclosed, wherein a hydrogen atom is replaced by a deuterium atom. The deuterium labeled compounds comprise a deuterium atom having an abundance of deuterium that is substantially greater than the natural abundance of deuterium, e.g., 0.015%. The term “deuterium enrichment factor” as used herein means the ratio between the deuterium abundance and the natural abundance of a deuterium. In one aspect, a compound of the application has a deuterium enrichment factor for each deuterium atom of at least 3500 (52.5% deuterium incorporation at each deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Deuterium labeled compounds can be prepared using any of a variety of art-recognized techniques. For example, deuterium labeled compounds of each of the formulae described herein or the compounds listed in Table 1 can generally be prepared by carrying out the procedures described herein, by substituting a readily available deuterium labeled reagent for a non-deuterium labeled reagent. A compound of the application or a pharmaceutically acceptable salt or solvate thereof that contains the aforementioned deuterium atom(s) is within the scope of the application. Further, substitution with deuterium, i.e.,2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life and/or reduced dosage requirements. As used herein, the term “treat”, “treating”, or “treatment” herein, is meant decreasing the symptoms, markers, and/or any negative effects of a disease, disorder or condition in any appreciable degree in a patient who currently has the condition. The term “treat”, “treating”, or “treatment” includes alleviating symptoms of a disease, disorder, or condition, e.g., alleviating the symptoms of epilepsy. In some embodiments, treatment may be administered to a subject who exhibits only early signs of the condition for the purpose of decreasing the risk of developing the disease, disorder, and/or condition. As used herein, the term “prevent”, “prevention”, or “preventing” refers to any method to partially or completely prevent or delay the onset of one or more symptoms or features of a disease, disorder, and/or condition. Prevention may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition. As used herein, “subject” means a human or animal (in the case of an animal, more typically a mammal). In one embodiment, the subject is a human. In one embodiment, the subject is a male. In one embodiment, the subject is a female. As used herein, the term a “fluorinated derivative” is a derivative compound that has the same chemical structure as the original compound, except that at least one atom is replaced with a fluorine atom or with a group of atoms containing at least one fluorine atom. The problem to be solved by the present application is the identification of novel compounds for the treatment and/or prevention of epilepsy and/or other diseases or disorders ameliorated by KCNQ2/3 potassium channel opening. Although drugs for epilepsy and related disorders are available, these drugs are often not suitable for many patients for a variety of reasons. Many epilepsy drugs are associated with adverse effects. For example, many of the available epilepsy drugs are believed to significantly increase the risk of birth defects if taken during the first trimester of pregnancy. Other adverse side effects include urinary retention, neuro-psychiatric symptoms including hallucinations and psychosis, dizziness and somnolence, QT-prolonging effect, and increased risk of suicidal behavior and ideation. Some epilepsy drugs require administration of high doses due to extensive metabolism into inactive or less potent metabolites. The present application provides the solution of new fluorinated 2-amino-4-(benzylamino)phenylcarbamate compounds for treating epilepsy and other diseases or disorders ameliorated by KCNQ2/3 potassium channel opening. The compounds described herein have the advantage of providing improved potency, selectivity, tissue penetration, half-life, and/or metabolic stability. Compounds of the Application The present application relates to a compound of formula A: or a pharmaceutically acceptable salt or solvate thereof, wherein: X1, X2, X3, and X9are each independently H, deuterium, F, NH2, or a C1-C4alkyl optionally substituted with one or more F; X10is C(O)(C7X7)nX6or CO2(C7X7)nX6; X4is H, C1-C4alkyl, C2-C6alkenyl, or C2-C6alkynyl; X5is phenyl-(CX8X8)m, wherein the phenyl is substituted with one or more substituents independently selected from deuterium, C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5, and wherein at least one substituent is selected from C1-C4alkyl substituted with one or more F, F, and SF5, or X4and X5, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclic ring comprising 1 or 2 heteroatoms selected from N, O, and S, wherein the heterocyclic ring is optionally substituted with one or more substituents independently selected from deuterium, C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5, wherein at least one substituent is selected from C1-C4alkyl substituted with one or more F, F, and SF5, or two substituents attached to adjacent carbon atoms on the heterocyclic ring, together with the carbon atoms to which they are attached, form a phenyl substituted with one or more substituents independently selected from deuterium, C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5, wherein the phenyl is substituted with at least one substituent selected from C1-C4alkyl substituted with one or more F, F, and SF5; X6is H or deuterium; each X7is independently H, C1-C4alkyl, or deuterium, or two X7, together with the carbon atom to which they are attached, form a 3- to 6-membered carbocyclic ring or a 3- to 6-membered heterocyclic ring comprising 1 or 2 heteroatoms selected from N, O, and S; each X5is independently H, deuterium, C1-C4alkyl, C1-C4alkyl substituted with one or more F, or F; m is 1, 2, or 3; and n is 1, 2, or 3, wherein when X1, X2, X3, and X6are each H, n is 2, each X7is H, X5is 4-fluorobenzyl, X9is NH2, and X10is CO2(C7X7)nX6, then X4is not propenyl or propynyl. In one embodiment, the compound of the present application is a compound of formula A, wherein when X1, X2, X3, and X6are each H, n is 2, each X7is H, and X5is 4-fluorobenzyl, then X4is not propenyl or propynyl. In one embodiment, the compound of the present application is a compound of formula A, wherein when X10is CO2(C7X7)nX6, then X4is not H. In one embodiment, the compound of formula A is of formula I: or a pharmaceutically acceptable salt or solvate thereof, wherein X1, X2, and X3are each independently H, deuterium, or F, and X4, X5, X6, X7, X8, m, and n are each as defined above in formula A, wherein when X1, X2, X3, and X6are each H, n is 2, each X7is H, and X5is 4-fluorobenzyl, then X4is not propenyl or propynyl. In one embodiment, the compound of formula A is of formula Ia: or a pharmaceutically acceptable salt or solvate thereof, wherein X2, X3, X4, X5, X6, X7, X8, m, and n are each as defined above in formula A. For a compound of formula A, I, or Ia, X1, X2, X3, X4, X5, X6, X7, X8, X9, X10, m, and n can each be, where applicable, selected from the groups described herein below, and any group described herein for any of X1, X2, X3, X4, X5, X6, X7, X8, X9, X10, m, and n can be combined, where applicable, with any group described herein for one or more of the remainder of X1, X2, X3, X4, X5, X6, X7, X8, X9, X10, m, and n. In one embodiment, at least one of X1, X2, X3, and X9is NH2. In one embodiment, one of X1, X2, X3, and X9is NH2. In one embodiment, X9is NH2. In one embodiment, one of X1, X2, X3, and X9is NH2, and the remainder of X1, X2, X3, and X9are each independently H, deuterium, F, C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, X9is NH2, and X1, X2, and X3are each independently H, deuterium, F, C1-C4alkyl (e.g., methyl, ethyl, propyl, propyl, butyl, i-butyl, or t-butyl), or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, X9is NH2, and X1, X2, and X3are each independently H, deuterium, or F. In one embodiment, X9is NH2, and X3is F. In one embodiment, X9is NH2, X3is F, and X1and X2are each independently H or deuterium. In one embodiment, at least one of X1, X2, X3, and X9is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, at least two of X1, X2, X3, and X9is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, X1and X9are each independently C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), and X2and X3are each independently H, deuterium, F, or NH2. In one embodiment, X1and X9are each independently C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), and X2and X3are each independently H, deuterium, or F. In one embodiment, X1and X9are each independently methyl, CF3, CHF2, or CH2F, and X2and X3are each independently H, deuterium, or F. In one embodiment, at least one of X1and X9is methyl. In one embodiment, X1and X9are each methyl. In one embodiment, X1and X9are each methyl, and X2and X3are each independently H, deuterium, or F. In one embodiment, X10is CO2(C7X7)nX6. In one embodiment, X10is C(O)(C7X7)nX6. In one embodiment, X1, X2, and X3are each H. In one embodiment, at least one of X1, X2, and X3is deuterium or F. In one embodiment, X1is deuterium or F, and X2and X3are each H. In one embodiment, X1is F, and X2and X3are each H. In one embodiment, X2is deuterium or F, and X1and X3are each H. In one embodiment, X2is F, and X1and X3are each H. In one embodiment, X3is deuterium or F, and X1and X2are each H. In one embodiment, X3is F, and X1and X2are each H. In one embodiment, at least two of X1, X2, and X3are deuterium or F. In one embodiment, X1and X2are each independently deuterium or F, and X3is H. In one embodiment, X1and X2are each F, and X3is H. In one embodiment, X1and X3are each independently deuterium or F, and X2is H. In one embodiment, X1and X3are each F, and X2is H. In one embodiment, X2and X3are each independently deuterium or F, and X1is H. In one embodiment, X2and X3are each F, and X1is H. In one embodiment, X4is H. In one embodiment, X4is H, only when X10is C(O)(C7X7)nX6. In one embodiment, X4is C1-C4alkyl, C2-C6alkenyl, or C2-C6alkynyl. In one embodiment, X4is C1-C4alkyl, C2-C6alkenyl, or C2-C6alkynyl, when X10is CO2(C7X7)nX6. In one embodiment, X4is C1-C4alkyl selected from methyl, ethyl, propyl, i-propyl, butyl, i-butyl, and t-butyl. In one embodiment, X4is C2-C6alkenyl or C2-C6alkynyl. In one embodiment, X4is C2-C6alkenyl selected from ethenyl, propenyl (e.g., 1-propenyl or 2-propenyl), butenyl (e.g., 1-butenyl, 2-butenyl, or 3-butenyl), pentenyl (e.g., 1-pentenyl, 2-pentenyl, 3-pentenyl, or 4-pentenyl), and hexenyl (e.g., 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, or 5-hexenyl). In one embodiment, X4is 1-propenyl or 2-propenyl. In one embodiment, X4is C2-C6alkynyl selected from ethynyl, propynyl (e.g., 1-propynyl or 2-propynyl), butynyl (e.g., 1-butynyl, 2-butynyl, or 3-butynyl), pentynyl (e.g., 1-pentynyl, 2-pentynyl, 3-pentynyl, or 4-pentynyl), and hexynyl (e.g., 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, or 5-hexynyl). In one embodiment, X4is 1-propynyl or 2-propynyl. In one embodiment, X5is phenyl-(CX8X8), phenyl-(CX8X8)2, or phenyl-(CX8X8)3, wherein the phenyl is substituted with one or more substituents independently selected from deuterium, C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) and C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F) and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In one embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3and F. In one embodiment, the substituent is attached at the para-position on the phenyl ring. In one embodiment, the substituent(s) are attached at the meta-position(s) on the phenyl ring. In one embodiment, the substituent(s) are attached at the ortho-position(s) on the phenyl ring. In one embodiment, X5is 4-fluoro-benzyl, 4-trifluoromethyl-benzyl, or 3-trifluoromethyl-benzyl. In one embodiment, each X5is H. In one embodiment, at least one X5is deuterium, C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), or F. In one embodiment, at least one X5is deuterium. In one embodiment, at least one X5is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), or F. In one embodiment, at least one X5is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, at least one X5is C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F) or F. In one embodiment, at least one X5is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl). In one embodiment, at least one X5is C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, at least one X5is F. In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclic ring comprising 1 or 2 heteroatoms selected from N, O, and S (e.g., pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, piperidinyl, piperazinyl, tetrahydropyranyl, tetrahydrothiapyranyl, dioxanyl, morpholinyl, oxazinanyl, thiazinanyl, or oxathianyl). In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclic ring comprising 1 heteroatom selected from N, O, and S. In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a 5- or 6-membered heterocyclic ring comprising 1 heteroatom selected from N, O, and S. In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a 5- or 6-membered heterocyclic ring comprising 1 heteroatom selected from N and O. In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a pyrrolidinyl or piperidinyl ring. In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclic ring substituted with one or more substituents independently selected from deuterium, C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), F, and SF5. In one embodiment, the heterocyclic ring is substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), and F. In one embodiment, the heterocyclic ring is substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) and C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, the heterocyclic ring is substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F) and F. In one embodiment, the heterocyclic ring is substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In one embodiment, the heterocyclic ring is substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In one embodiment, the heterocyclic ring is substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In one embodiment, the heterocyclic ring is substituted with one or more substituents independently selected from CF3and F. In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclic ring substituted with two or more substituents, wherein two substituents attached to adjacent carbon atoms on the heterocyclic ring, together with the carbon atoms to which they are attached, form a phenyl substituted with one or more substituents independently selected from deuterium, C1-C4alkyl (e.g., methyl, ethyl, propyl, propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) and C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F) and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In one embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3and F. In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a heterocyclic ring selected from wherein the nitrogen atom is the nitrogen atom bonded to X4and X5. In one embodiment, X6is H. In one embodiment, X6is deuterium. In one embodiment, each X7is H. In one embodiment, at least one X7is deuterium. In one embodiment, at least one X7is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl). In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- to 6-membered carbocyclic ring (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl). In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- to 6-membered heterocyclic ring comprising 1 or 2 heteroatoms selected from N, O, and S (e.g., aziridinyl, oxiranyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, piperidinyl, piperazinyl, tetrahydropyranyl, tetrahydrothiapyranyl, dioxanyl, morpholinyl, oxazinanyl, thiazinanyl, or oxathianyl). In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- to 6-membered heterocyclic ring comprising 1 heteroatom selected from N, O, and S. In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- or 4-membered heterocyclic ring comprising 1 heteroatom selected from N, O, and S. In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- or 4-membered heterocyclic ring comprising 1 heteroatom selected from N and O. In one embodiment, m is 1. In one embodiment, m is 2. In one embodiment, m is 3. In one embodiment, n is 1. In one embodiment, n is 2. In one embodiment, n is 3. Any of the substituent groups described above for any of X1, X2, X3, X4, X5, X6, X7, X8, X9, X10, m, and n can be combined with any of the substituent groups described above for one or more of the remainder of X1, X2, X3, X4, X5, X6, X7, X8, X9, X10, m, and n.(1a) In one embodiment, X1, X2, and X3are each H.(1b) In one embodiment, X1and X2are each H, and X3is deuterium or F.(1c) In one embodiment, X1and X2are each H, and X3is F.(1d) In one embodiment, X1and X9are each methyl, and X3is F.(2a) In one embodiment, X6is H.(2b) In one embodiment, X6is deuterium.(3a) In one embodiment, each X7is H.(3b) In one embodiment, at least one X7is C1-C4alkyl.(3c) In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- to 6-membered carbocyclic ring or a 3- to 6-membered heterocyclic ring comprising 1 to 2 heteroatoms selected from N, O, and S. In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- to 6-membered heterocyclic ring comprising 1 heteroatom selected from N, O, and S. In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- or 4-membered heterocyclic ring comprising 1 heteroatom selected from N, O, and S. In one embodiment, at least two X7, together with the carbon atom to which they are attached, form a 3- or 4-membered heterocyclic ring comprising 1 heteroatom selected from N and O.(4a) In one embodiment, n is 1.(4b) In one embodiment, n is 2.(4c) In one embodiment, n is 3.(A1) In one embodiment, X1, X2, and X3are each as defined in (1a), and X7is as defined in (3a).(A2) In one embodiment, X1, X2, and X3are each as defined in (1b), and X7is as defined in (3a).(A3) In one embodiment, X1, X2, and X3are each as defined in (1c), and X7is as defined in (3a).(A4) In one embodiment, X1, X3, and X9are each as defined in (1d), and X7is as defined in (3a).(B1) In one embodiment, X1, X2, and X3are each as defined in (1a), and X7is as defined in (3b).(B2) In one embodiment, X1, X2, and X3are each as defined in (1b), and X7is as defined in (3b).(B3) In one embodiment, X1, X2, and X3are each as defined in (1c), and X7is as defined in (3b).(B4) In one embodiment, X1, X3, and X9are each as defined in (1d), and X7is as defined in (3b).(C1) In one embodiment, X1, X2, and X3are each as defined in (1a), and X7is as defined in (3c).(C2) In one embodiment, X1, X2, and X3are each as defined in (1b), and X7is as defined in (3c).(C3) In one embodiment, X1, X2, and X3are each as defined in (1c), and X7is as defined in (3c).(C4) In one embodiment, X1, X3, and X9are each as defined in (1d), and X7is as defined in (3c).(D1) In one embodiment, X1, X2, X3, X7, and X9are each as defined in any one of (A1)-(A4), and X6is as defined in (2a) or (2b). In one embodiment, X6is as defined in (2a).(D2) In one embodiment, X1, X2, X3, X7, and X9are each as defined in any one of (B1)-(B4), and X6is as defined in (2a) or (2b). In one embodiment, X6is as defined in (2a).(D3) In one embodiment, X1, X2, X3, X7, and X9are each as defined in any one of (C1)-(C4), and X6is as defined in (2a) or (2b). In one embodiment, X6is as defined in (2a).(E1) In one embodiment, X1, X2, X3, X7, and X9are each as defined in any one of (A1)-(A4), (B1)-(B4), or (C1)-(C4), and n is as defined in (4a).(E2) In one embodiment, X1, X2, X3, X7, and X9are each as defined in any one of (A1)-(A4), (B1)-(B4), or (C1)-(C4), and n is as defined in (4b).(E3) In one embodiment, X1, X2, X3, X7, and X9are each as defined in any one of (A1)-(A4), (B1)-(B4), or (C1)-(C4), and n is as defined in (4c).(5a) In one embodiment, X4is C1-C4alkyl, and X5is phenyl-(CX8X8)m. In a further embodiment, the phenyl is substituted with one or more groups independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl and C1-C4alkyl substituted with one or more F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3and F.(5b) In one embodiment, X4is C2-C6alkenyl, and X5is phenyl-(CX8X8)m. In a further embodiment, the phenyl is substituted with one or more groups independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl and C1-C4alkyl substituted with one or more F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3and F.(5c) In one embodiment, X4is C2-C6alkynyl, and X5is phenyl-(CX8X8)m. In a further embodiment, the phenyl is substituted with one or more groups independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl and C1-C4alkyl substituted with one or more F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3and F.(5d) In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclic ring comprising 1 to 2 heteroatoms selected from N, O, and S, optionally substituted with one or more substituents independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5. In one embodiment, the heterocyclic ring is optionally substituted with one or more substituents independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, and F. In one embodiment, the heterocyclic ring is optionally substituted with one or more substituents independently selected from C1-C4alkyl and C1-C4alkyl substituted with one or more F. In one embodiment, the heterocyclic ring is optionally substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F and F. In one embodiment, the heterocyclic ring is optionally substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In one embodiment, the heterocyclic ring is optionally substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In one embodiment, the heterocyclic ring is optionally substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In one embodiment, the heterocyclic ring is optionally substituted with one or more substituents independently selected from CF3and F.(5e) In one embodiment, X4and X5, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclic ring comprising 1 to 2 heteroatoms selected from N, O, and S, substituted with two or more substituents, wherein two substituents attached to adjacent carbon atoms on the heterocyclic ring, together with the carbon atoms to which they are attached, form a phenyl substituted with one or more substituents independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl and C1-C4alkyl substituted with one or more F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In a further embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3and F.(6a) In one embodiment, m is 1.(6b) In one embodiment, m is 2.(6c) In one embodiment, m is 3.(7a) In one embodiment, each X5is H.(7b) In one embodiment, at least one X5is deuterium.(7c) In one embodiment, at least one X5is C1-C4alkyl, C1-C4alkyl substituted with one or more F, or F.(F1a) In one embodiment, X4and X5are each as defined in (5a), and m is as defined in any one of (6a)-(6c). In a further embodiment, m is as defined in (6a).(F1b) In one embodiment, X4and X5are each as defined in (5b), and m is as defined in any one of (6a)-(6c). In a further embodiment, m is as defined in (6a).(F1c) In one embodiment, X4and X5are each as defined in (5c), and m is as defined in any one of (6a)-(6c). In a further embodiment, m is as defined in (6a).(G1a) In one embodiment, X4, X5, and m are each as defined in any one of (F1a)-(F1c), and X5is as defined in (7a).(G1b) In one embodiment, X4, X5, and m are each as defined in any one of (F1a)-(F1c), and X5is as defined in (7b).(G1c) In one embodiment, X4, X5, and m are each as defined in any one of (F1a)-(F1c), and X5is as defined in (7c).(H1a) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, and X3are each as defined in (1a).(H1b) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, and X3are each as defined in (1b).(H1c) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, and X3are each as defined in (1c).(H1d) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X3, and X9are each as defined in (1d).(H1e) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X7is as defined in (3a).(H1f) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X7is as defined in (3b).(H1g) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X7is as defined in (3c).(H1h) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (A1).(H1i) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (A2).(H1j) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (A3).(H1k) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X3, X7, and X9are each as defined (A4).(H1l) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (B1).(H1m) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (B2).(H1n) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (B3).(H1o) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X3, X7, and X9are each as defined (B4).(H1p) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (C1).(H1q) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (C2).(H1r) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, and X7are each as defined (C3).(H1s) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X3, X7, and X9are each as defined (C4).(H1t) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, X6, X7, and X9are each as defined (D1).(H1u) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, X6, X7, and X9are each as defined (D2).(H1v) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, X6, X7, and X9are each as defined (D3).(H1w) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, X7, X9, and n are each as defined (E1).(H1x) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, X7, X9, and n are each as defined (E2).(H1y) In one embodiment, X4and X5are each as defined in any one of (5a)-(5e), and X1, X2, X3, X7, X9, and n are each as defined (E3).(I1a) In one embodiment, X1, X2, and X3are each as defined in (1a), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1b) In one embodiment, X1, X2, and X3are each as defined in (1b), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1c) In one embodiment, X1, X2, and X3are each as defined in (1c), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1d) In one embodiment, X1, X3, and X9are each as defined in (1d), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1e) In one embodiment, X1, X2, and X3are each as defined in (1a), and X4, X5, X8, and m are each as defined in any one of (G1a)-(G1c).(I1f) In one embodiment, X1, X2, and X3are each as defined in (1b), and X4, X5, X8, and m are each as defined in any one of (G1a)-(G1c).(I1g) In one embodiment, X1, X2, and X3are each as defined in (1c), and X4, X5, X8, and m are each as defined in any one of (G1a)-(G1c).(I1h) In one embodiment, X1, X3, and X9are each as defined in (1d), and X4, X5, X8, and m are each as defined in any one of (G1a)-(G1c).(I1i) In one embodiment, X1, X2, X3, and X7are each as defined (A1), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1j) In one embodiment, X1, X2, X3, and X7are each as defined (A2), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1k) In one embodiment, X1, X2, X3, and X7are each as defined (A3), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1l) In one embodiment, X1, X3, X7, and X9are each as defined (A4), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1m) In one embodiment, X1, X2, X3, and X7are each as defined (B1), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1n) In one embodiment, X1, X2, X3, and X7are each as defined (B2), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1o) In one embodiment, X1, X2, X3, and X7are each as defined (B3), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1p) In one embodiment, X1, X3, X7, and X9are each as defined (B4), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1q) In one embodiment, X1, X2, X3, and X7are each as defined (C1), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1r) In one embodiment, X1, X2, X3, and X7are each as defined (C2), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1 s) In one embodiment, X1, X2, X3, and X7are each as defined (C3), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1t) In one embodiment, X1, X3, X7, and X9are each as defined (C4), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1u) In one embodiment, X1, X2, X3, X6, X7, and X9are each as defined (D1), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1v) In one embodiment, X1, X2, X3, X6, X7, and X9are each as defined (D2), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1w) In one embodiment, X1, X2, X3, X6, X7, and X9are each as defined (D3), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1x) In one embodiment, X1, X2, X3, X7, X9, and n are each as defined (E1), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1y) In one embodiment, X1, X2, X3, X7, X9, and n are each as defined (E2), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(I1z) In one embodiment, X1, X2, X3, X7, X9, and n are each as defined (E3), and X4, X5, and m are each as defined in any one of (F1a)-(F1c).(J1) In one embodiment, X1, X2, X3, X4, X5, X6, X7, X8, X9, m, and n are each, where applicable, as defined in any one of (1a)-(I1z), X10is C(O)(C7X7)nX6.(J2) In one embodiment, X1, X2, X3, X4, X5, X6, X7, X8, X9, m, and n are each, where applicable, as defined in any one of (1a)-(I1z), X10is CO2(C7X7)nX6. In one embodiment, the compound of formula A is of formula II or VI: or a pharmaceutically acceptable salt or solvate thereof, wherein X3, X4, X5, X8, and m are each as defined above in formula A. X3, X4, X5, X8, and m can each be selected from any of the substituents described above in formula A, and any of the substituents described above for any of X3, X4, X5, X8, and m can be combined with any of the substituents described above for one or more of the remainder of X3, X4, X5, X8, and m. In one embodiment, the compound of formula A is of formula V or VII: or a pharmaceutically acceptable salt or solvate thereof, wherein X3, X4, X5, X8, and m are each as defined above in formula A. X3, X4, X5, X8, and m can each be selected from any of the substituents described above in formula A, and any of the substituents described above for any of X3, X4, X5, X8, and m can be combined with any of the substituents described above for one or more of the remainder of X3, X4, X5, X8, and m. In one embodiment, the compound of formula A is of formula IIIa: or a pharmaceutically acceptable salt or solvate thereof, wherein: X3, X4, and m are each as defined above in formula A; t1 is 1, 2, 3, 4, or 5; and each Z1is independently C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, or SF5, wherein at least one Z1is C1-C4alkyl substituted with one or more F, F, or SF5, wherein when X3is H, t1 is 1, and Z1is 4-fluoro, then X4is not propenyl or propynyl. In one embodiment, the compound of formula A is of formula IIIb or IIIc: or a pharmaceutically acceptable salt or solvate thereof, wherein: X3, X4, and m are each as defined above in formula A; t1 is 1, 2, 3, 4, or 5; and each Z1is independently C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, or SF5, wherein at least one Z1is C1-C4alkyl substituted with one or more F, F, or SF5. For a compound of formula IIIa, IIIb, or IIIc, t1 and Z1can each be, where applicable, selected from the groups described herein below, and any group described herein for any of t1 and Z1can be combined, where applicable, with any group described herein for the remainder of t1 and Z1. In one embodiment, t1 is 1, 2, or 3. In one embodiment, t1 is 1 or 2. In one embodiment, t1 is 1. In one embodiment, t1 is 2. In one embodiment, t1 is 3. In one embodiment, t1 is 4. In one embodiment, t1 is 5. In one embodiment, at least one Z1is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), F, or SF5. In one embodiment, at least one Z1is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), or F. In one embodiment, at least one Z1is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, at least one Z1is C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F) or F. In one embodiment, at least one Z1is CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, or SF5. In a further embodiment, at least one Z1is CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, or F. In a further embodiment, at least one Z1is CF3, CHF2, CH2F, or F. In a further embodiment, at least one Z1is CF3or F. X3, X4, and m can each be selected from any of the substituents described above in formula A, and any of the substituents described above for any of X3, X4, and m can be combined with any of the substituents described above for one or more of the remainder of X3, X4, and m, and can further be combined with any of the substituents described for any of t1 and Z1. In one embodiment, t1 is 1, and Z1is CF3or F. In one embodiment, t1 is 1, Z1is CF3or F, and m is 1. In one embodiment, t1 is 1, Z1is CF3or F, and X4is propenyl or propynyl. In one embodiment, t1 is 1, Z1is CF3or F, m is 1, and X4is propenyl or propynyl. In one embodiment, t1 is 1, Z1is CF3or F, X4is propenyl or propynyl, and X3is H or F. In one embodiment, t1 is 1, Z1is CF3, m is 1, X4is propenyl or propynyl, and X3is H or F. In one embodiment, t1 is 1, Z1is CF3or F, m is 1, X4is propenyl or propynyl, and X3is F. In one embodiment, the compound of formula A is of formula IVa, IVb, or IVc: or a pharmaceutically acceptable salt or solvate thereof, wherein: X3is as defined above in formula A; q is 1, 2, or 3; t2 is 1, 2, 3, or 4; and each Z2is independently C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, or SF5, wherein at least one Z2is C1-C4alkyl substituted with one or more F, F, or SF5, or two Z2, together with adjacent carbon atoms to which they are attached, form a phenyl substituted with one or more substituents independently selected from C1-C4alkyl, C1-C4alkyl substituted with one or more F, F, and SF5, wherein the phenyl is substituted with at least one substituent selected from C1-C4alkyl substituted with one or more F, F, and SF5. For a compound of formula IVa, IVb, or IVc, q, t2, and Z2can each be, where applicable, selected from the groups described herein below, and any group described herein for any of q, t2, and Z2can be combined, where applicable, with any group described herein for one or more of the remainder of q, t2, and Z2. In one embodiment, q is 1. In one embodiment, q is 2. In one embodiment, q is 3. In one embodiment, t2 is 1, 2, or 3. In one embodiment, t2 is 1 or 2. In one embodiment, t2 is 1. In one embodiment, t2 is 2. In one embodiment, t2 is 3. In one embodiment, t2 is 4. In one embodiment, at least one Z2is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), F, or SF5. In one embodiment, at least one Z2is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), or F. In one embodiment, at least one Z2is C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) or C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, at least one Z2is C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F) or F. In one embodiment, at least one Z2is CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, or SF5. In a further embodiment, at least one Z2is CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, or F. In a further embodiment, at least one Z2is CF3, CHF2, CH2F, or F. In a further embodiment, at least one Z2is CF3or F. In one embodiment, two Z2, together with adjacent carbon atoms to which they are attached, form a phenyl substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl), C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F), and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl) and C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F). In one embodiment, the phenyl is substituted with one or more substituents independently selected from C1-C4alkyl substituted with one or more F (e.g., methyl, ethyl, propyl, i-propyl, butyl, i-butyl, or t-butyl, each of which is substituted with one or more F) and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, F, and SF5. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3, CHF2, CH2F, CH2CF3, CH2CHF2, CH2CH2F, and F. In one embodiment, the phenyl is substituted with one or more groups independently selected from CF3, CHF2, CH2F, and F. In one embodiment, the phenyl is substituted with one or more substituents independently selected from CF3and F. Any of the substituents described above for any of q, t2, and Z2can be combined with any of the substituents described above for any of the remainder of q, t2, and Z2, and can further be combined with any of the substituents described above for X3. In one embodiment, a compound of the present application is selected from the compounds in Tables 1a and 1b. TABLE 1aCpmdNo.Structure12345678911121314151617181920212223242526 TABLE 1bCpmdNo.Structure27282930313233343536 In one embodiment, a compound of the application is a pharmaceutically acceptable salt. In one embodiment, a compound of the application is a solvate. In one embodiment, a compound of the application is a hydrate. The present application relates to pharmaceutical compositions comprising one of the compounds of the application as an active ingredient. In one embodiment, the application provides a pharmaceutical composition comprising at least one compound of formula A, I, Ia, II, IIIa, IIIb, IIIc, IVa, IVb, IVc, V, VI, or VII, or a pharmaceutically acceptable salt or solvate thereof, and one or more pharmaceutically acceptable carrier or excipient. In one embodiment, the application provides a pharmaceutical composition comprising at least one compound of Table 1, or a pharmaceutically acceptable salt or solvate thereof, and one or more pharmaceutically acceptable carrier or excipient. The present application relates to a method of synthesizing a compound of the application or a pharmaceutically acceptable salt or solvate thereof. A compound of the application can be synthesized using a variety of methods known in the art, such as those described in U.S. Pat. No. 8,916,133, the contents of which are incorporated by reference in their entirety. The schemes and description below depict general routes for the preparation of a compound of the application. For example, compounds of the present application can be synthesized by following the steps outlined in Schemes 1-6 which comprise different sequences of assembling intermediates 3a, 3b, 3c, 3d, 3e, 4a, 4b, 4c, 5a, 5b, 5c, 5d, 5e, 7a, 7b, 7c, 7d, 7e, 7f, 8a, 8b, 8c, 8d, 8e, 8f, 9a, 9b, 9c, 9d, 9e, and 9f. Starting materials are either commercially available or made by known procedures in the reported literature or as illustrated. wherein X3, X4, Z1, t1, and m are as defined herein above. The general way of preparing representative compounds of the present application using intermediates 3a, 3b, 3c, 3d, and 3e is outlined in Scheme 1. Alkylation of amine 3a with bromide 3b (i.e., an alkyl bromide, allyl bromide, etc.) in the presence of a base, e.g., diisopropylethylamine (DIPEA), in a solvent, e.g., dimethylformamide (DMF), and optionally at an elevated temperature provides intermediate 3c. Nucleophilic addition of 3c to fluoride 3d in the presence of a base, e.g., triethylamine (Et3N), in a solvent, e.g., dimethylsulfoxide (DMSO), and optionally at an elevated temperature provides intermediate 3e. Reduction of 3e using a metal catalyst, e.g., Zinc (Zn), and ammonium chloride (NH4Cl) in the presence of a base, e.g., DIPEA and in a solvent, e.g., methanol (MeOH), and consequent esterification with an agent, e.g., ethyl chloroformate, in the presence of a base, e.g., diisopropylethylamine (DIPEA), and optionally at an elevated temperature provides compounds of formula IIIa. wherein X3, Z2, and t2 are as defined herein above. The general way of preparing representative compounds of the present application using intermediates 4a, 4b, and 4c is outlined in Scheme 2. Nucleophilic addition of 4a to fluoride 4b in the presence of a base, e.g., triethylamine (Et3N), in a solvent, e.g., dimethylsulfoxide (DMSO), and optionally at an elevated temperature provides intermediate 4c. Reduction of 4c using a metal catalyst, e.g., Zinc (Zn), and ammonium chloride (NH4Cl) in the presence of a base, i.e., DIPEA and in a solvent, i.e., methanol (MeOH), and consequent esterification with an agent, e.g., ethyl chloroformate, in the presence of a base, e.g., diisopropylethylamine (DIPEA), and optionally at an elevated temperature provides compounds of formula IVa. wherein X3, X4, Z1, t1, and m are as defined herein above. The general way of preparing representative compounds of the present application using intermediates 5a, 5b, 5c, 5d, and 5e is outlined in Scheme 3. Nucleophilic addition of 5a to fluoride 5b in the presence of a base, e.g., triethylamine (Et3N), in a solvent, e.g., dimethylsulfoxide (DMSO), and optionally at an elevated temperature provides intermediate 5c. Alkylation of amine 5c with bromide 5d (i.e., an alkyl bromide, allyl bromide, etc.) in the presence of a base, e.g., diisopropylethylamine (DIPEA), in a solvent, e.g., dimethylformamide (DMF), and optionally at an elevated temperature provides intermediate 5e. Reduction of 5e using a metal catalyst, e.g., Zinc (Zn), and ammonium chloride (NH4Cl) in the presence of a base, e.g., DIPEA and in a solvent, e.g., water, and consequent esterification with an agent, e.g., ethyl chloroformate, in the presence of a base, e.g., diisopropylethylamine (DIPEA), and optionally at an elevated temperature provides compounds of formula IIIa. wherein X3, X4, Z1, t1, and m are as defined herein above. The general way of preparing representative compounds of the present application using intermediates 7a, 7b, 7c, 7d, 7e, and 7f is outlined in Scheme 5. Nucleophilic addition of 7a to fluoride 7b in the presence of a base, e.g., triethylamine (Et3N), in a solvent, e.g., dimethylsulfoxide (DMSO), and optionally at an elevated temperature provides intermediate 7c. Reduction of 7c using a metal catalyst, e.g., Zinc (Zn), and ammonium chloride (NH4Cl) in the presence of a base, e.g., DIPEA and in a solvent, e.g., methanol (MeOH), and consequent protection using BOC2O in the presence of a base, e.g., NaHCO3, provides intermediate 7d. Alkylation of amine 7d with bromide 7e (i.e., an alkyl bromide, allyl bromide, etc.) in the presence of a base, e.g., diisopropylethylamine (DIPEA), in a solvent, e.g., dimethylformamide (DMF), and optionally at an elevated temperature provides intermediate 7f. Deprotection of 7f in the presence of an acid, e.g., trifluoroacetic acid (TFA), and optionally at an elevated temperature, and consequent esterification with an agent, e.g., ethyl chloroformate, in the presence of a base, e.g., diisopropylethylamine (DIPEA), and optionally at an elevated temperature provides compounds of formula IIIa. wherein X3and X5are as defined herein above. The general way of preparing representative compounds of the present using intermediates 8a, 8b, 8c, 8d, 8e, 8f, and 8g is outlined in Scheme 6. Nucleophilic addition of 8a to fluoride 8b in the presence of a base, e.g., triethylamine (Et3N), in a solvent, e.g., dimethylsulfoxide (DMSO), and optionally at an elevated temperature provides intermediate 8c. Protection of 8c with Boc2O in the presence of a base, e.g., sodium hydride (NaH) and/or 4-dimethylaminopyridine (DMAP), in a solvent, e.g., tetrahydrofuran (THF), and optionally at an elevated temperature provides intermediate 8d. Reduction of 8d using a metal catalyst, e.g., Zinc (Zn), and ammonium chloride (NH4Cl), in a solvent, e.g., methanol (MeOH), and optionally at an elevated temperature provides intermediate 8e. Acetylation of 8e with tert-butylacetyl chloride in the presence of a base, e.g., diisopropylethylamine (DIPEA), in a solvent, e.g., dichlorimethane (DCM), and optionally at an elevated temperature provides intermediate 8f. Deprotection of 7f in the presence of an acid, e.g., hydrochloric acid (HCL), in a solvent, e.g., DCM and/or diethyl ether (Et2O), and optionally at an elevated temperature provides compounds of formula VIa. wherein X3, X4, and X5are as defined herein above. The general way of preparing representative compounds of the present application using intermediates 9a, 9b, 9c, 9d, 9e, 9f is outlined in Scheme 7. Methylation of 9a with trimethylboroxine in the presence of a metal catalyst, e.g., tetrakis(triphenylphosphine)palladium (Pd(PPh3)4)), and a base, e.g., potassium carbonate (K2CO3), in a solvent, e.g., dimethylsulfoxide (DMSO), and optionally at an elevated temperature provides intermediate 9b. Reduction of 9b using a metal catalyst, e.g., Zinc (Zn), and ammonium chloride (NH4Cl), in a solvent, e.g., water and ethyl acetate (EtOAc), and optionally at an elevated temperature provides intermediate 9c. Bromination of 9c with N-bromosuccinimide (NB S), in the presence of an acid, e.g., acetic acid, and optionally at an elevated temperature provides intermediate 9d. Acetylation of 9d with tert-butylacetyl chloride in the presence of a base, e.g., diisopropylethylamine (DIPEA), in a solvent, e.g., acetonitrile (MeCN), and optionally at an elevated temperature provides intermediate 9e. Coupling 9e with 9f in the presence of a metal catalyst, e.g., tris(dibenzylideneacetone)dipalladium (Pd2(dba)3), and in the presence of a base, e.g., potassium tert-butoxide (t-BuOK), in a solvent, e.g., toluene, and optionally at an elevated temperature provides compounds of formula VII. The present application also comprehends deuterium labeled compounds, wherein one or more hydrogen atoms is replaced by a deuterium atom having an abundance of deuterium at that position that is substantially greater than the natural abundance of deuterium, which is 0.015%. Deuterium labeled compounds can be prepared by using any of a variety of art-recognized techniques. For example, deuterium labeled compounds of any of the formulae described herein and compounds listed in Table 1 of this application can be prepared. In one aspect, a deuterium labeled compound of the application is a pharmaceutically acceptable salt. In one aspect, a deuterium labeled compound of the application is a solvate. In one aspect, a deuterium labeled compound of the application is a hydrate. The present application relates to pharmaceutical compositions comprising one of the deuterium labeled compounds of the application as an active ingredient. In one aspect, the application provides a pharmaceutical composition comprising at least one deuterium labeled compound of any of the formulae described herein or a pharmaceutically acceptable salt or solvate thereof and one or more pharmaceutically acceptable carrier or excipient. The present application relates to a method of synthesizing a deuterium labeled compound of the application or a pharmaceutically acceptable salt or solvate thereof. The deuterium labeled compounds of the application can be prepared using any of a variety of art-recognized techniques, such as those described in U.S. Pat. No. 8,916,133, the contents of which are incorporated by reference in their entirety. For example, a deuterium labeled compound can be prepared by starting with deuterium labeled Compound 1 and/or substituting a readily available deuterium labeled reagent for a non-deuterium labeled reagent. The scheme and description below depicts a general route for the incorporation of deuterium label to produce a deuterium labeled compound of the application. Scheme 1A outlines a preparation for a deuterium labeled compound of the application. The preparation begins with Compound A (from Scheme 1A described herein). In Step 1, the nitro group of Compound A is reduced and then the deuterium label is introduced via formation of a carbamate containing one or more deuterium. For example, the nitro group of Compound A can be reduced using zinc powder and ammonium chloride in methanol and the carbamate can be formed using ethyl-d5chloroformate to provide a deuterium labeled compound. In some embodiments, temporary protecting groups may be used to prevent other reactive functionality, such as amines, thiols, alcohols, phenols, and carboxylic acids, from participating or interfering in the fluorination reaction. Representative amine protecting groups include, for example, tert-butoxycarbonyl and trityl (removed under acid conditions), Fmoc (removed by the use of secondary amines such as piperidine), and benzyloxycarbonyl (removed by strong acid or by catalytic hydrogenolysis). The trityl group may also be used for the protection of thiols, phenols, and alcohols. In certain embodiments the carboxylic acid protecting groups include, for example, tert-butyl ester (removed by mild acid), benzyl ester (usually removed by catalytic hydrogenolysis), and alkyl esters such as methyl or ethyl (usually removed by mild base). All protecting groups may be removed at the conclusion of the synthesis using the conditions described above for the individual protecting groups, and the final product may be purified by techniques which would be readily apparent to one of ordinary skill in the art, in combination with the teachings described herein. Biological Assays Assessment of KCNQ2/3 Channel Activation Activity Biological activities of the compounds of the application can be assessed by using various methods known in the art. For example, the KCNQ2/3 channel activation activity of the compounds of the application can be evaluated through an in vitro assay described below. The in vitro effects of a compound of the application on cloned KCNQ2/3 potassium channels (e.g., encoded by the human KCNQ2/3 gene) are evaluated using a patch clamp system. Compounds of the application are tested at various concentrations (e.g., 0.01, 0.1, 1, 10 and 100 μM) for a certain duration of exposure (e.g., 5 min). The baseline for each recording is established. A single test compound concentration is applied for a certain duration of exposure after the vehicle. Each recording ends with treatment with a supramaximal dose of linopirdine. The % activation is calculated using the following equation by using leak subtracted responses: vehicle_response-compound_responsevehicle_response-flupirtine_response Maximal Electroshock Seizure Test (MES) In MES test, the ability of different doses of the test compound in preventing seizure induced by an electrical stimulus, delivered through the corneal electrodes primed with a drop of anesthetic/electrolyte solution is tested. Mice are restrained and released immediately following corneal stimulation that allows for the observation of the entire seizure episode. A maximal seizure in a test animal includes four distinct phases that includes, hind leg flexor component tonic phase (Phase I), hind leg extensor component of the tonic phase (Phase II), intermittent, whole-body clonus (Phase III), and muscular relaxation (Phase IV) followed by seizure termination (Woodbury & Davenport, 1952; Racine et al., 1972). Test compounds are tested for their ability to abolish hind limb tonic extensor component that indicates the compound's ability to inhibit MES-induced seizure spread. Compounds are pre-administered (i.p) and tested at various time points for the abolishment of hind limb tonic extensor component after electrical stimulus. Corneal-Kindled Mouse Model of Partial Seizures In corneal kindled seizure model, mice are kindled electrically with stimulation delivered through corneal electrodes primed with tetracaine hydrochloride in saline, twice daily, until 5 consecutive stage V seizures are induced. Mice are considered kindled when they display at least 5 consecutive stage V seizures according to the Racine scale (Racine et al., 1972) including, mouth and facial clonus (stage I), Stage I plus head nodding (Stage II), Stage II plus forelimb clonus (Stage III), Stage III plus rearing (Stage IV), and stage IV plus repeated rearing and falling (Stage V) (Racine et al., 1972). At the completion of the kindling acquisition, mice are permitted a 3-day stimulation-free period prior to any drug testing. On the day of the experiment, fully kindled mice are pre-administered (i.p) with increasing doses of the test compound and challenged with the corneal kindling stimulus. Mice are scored as protected (seizure score of <3) or not protected, (seizure score ≥4) based on the Racine scoring (Racine et al., 1972). Pharmaceutical Compositions The present application relates to pharmaceutical compositions comprising a compound of the application as an active ingredient. In one embodiment, the application provides a pharmaceutical composition comprising at least one compound of each of the formulae described herein, or a pharmaceutically acceptable salt or solvate thereof, and one or more pharmaceutically acceptable carriers or excipients. In one embodiment, the application provides a pharmaceutical composition comprising at least one compound selected from Table 1. As used herein, the term “composition” is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The compounds of the application can be formulated for oral administration in forms such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups and emulsions. The compounds of the application can also be formulated for intravenous (bolus or in-fusion), intraperitoneal, topical, subcutaneous, intramuscular or transdermal (e.g., patch) administration, all using forms well known to those of ordinary skill in the pharmaceutical arts. The formulation of the present application may be in the form of an aqueous solution comprising an aqueous vehicle. The aqueous vehicle component may comprise water and at least one pharmaceutically acceptable excipient. Suitable acceptable excipients include those selected from the group consisting of a solubility enhancing agent, chelating agent, preservative, tonicity agent, viscosity/suspending agent, buffer, and pH modifying agent, and a mixture thereof. Any suitable solubility enhancing agent can be used. Examples of a solubility enhancing agent include cyclodextrin, such as those selected from the group consisting of hydroxypropyl-β-cyclodextrin, methyl-β-cyclodextrin, randomly methylated-β-cyclodextrin, ethylated-β-cyclodextrin, triacetyl-β-cyclodextrin, peracetylated-β-cyclodextrin, carboxymethyl-β-cyclodextrin, hydroxyethyl-β-cyclodextrin, 2-hydroxy-3-(trimethylammonio)propyl-β-cyclodextrin, glucosyl-β-cyclodextrin, sulphated β-cyclodextrin (S13-CD), maltosyl-β-cyclodextrin, β-cyclodextrin sulfobutyl ether, branched-β-cyclodextrin, hydroxypropyl-γ-cyclodextrin, randomly methylated-γ-cyclodextrin, and trimethyl-γ-cyclodextrin, and mixtures thereof. Any suitable chelating agent can be used. Examples of a suitable chelating agent include those selected from the group consisting of ethylenediaminetetraacetic acid and metal salts thereof, disodium edetate, trisodium edetate, and tetrasodium edetate, and mixtures thereof. Any suitable preservative can be used. Examples of a preservative include those selected from the group consisting of quaternary ammonium salts such as benzalkonium halides (preferably benzalkonium chloride), chlorhexidine gluconate, benzethonium chloride, cetyl pyridinium chloride, benzyl bromide, phenylmercury nitrate, phenylmercury acetate, phenylmercury neodecanoate, merthiolate, methylparaben, propylparaben, sorbic acid, potassium sorbate, sodium benzoate, sodium propionate, ethyl p-hydroxybenzoate, propylaminopropyl biguanide, and butyl-p-hydroxybenzoate, and sorbic acid, and mixtures thereof. The aqueous vehicle may also include a tonicity agent to adjust the tonicity (osmotic pressure). The tonicity agent can be selected from the group consisting of a glycol (such as propylene glycol, diethylene glycol, triethylene glycol), glycerol, dextrose, glycerin, mannitol, potassium chloride, and sodium chloride, and a mixture thereof. The aqueous vehicle may also contain a viscosity/suspending agent. Suitable viscosity/suspending agents include those selected from the group consisting of cellulose derivatives, such as methyl cellulose, ethyl cellulose, hydroxyethylcellulose, polyethylene glycols (such as polyethylene glycol 300, polyethylene glycol 400), carboxymethyl cellulose, hydroxypropylmethyl cellulose, and cross-linked acrylic acid polymers (carbomers), such as polymers of acrylic acid cross-linked with polyalkenyl ethers or divinyl glycol (Carbopols—such as Carbopol 934, Carbopol 934P, Carbopol 971, Carbopol 974 and Carbopol 974P), and a mixture thereof. In order to adjust the formulation to an acceptable pH (typically a pH range of about 5.0 to about 9.0, more preferably about 5.5 to about 8.5, particularly about 6.0 to about 8.5, about 7.0 to about 8.5, about 7.2 to about 7.7, about 7.1 to about 7.9, or about 7.5 to about 8.0), the formulation may contain a pH modifying agent. The pH modifying agent is typically a mineral acid or metal hydroxide base, selected from the group of potassium hydroxide, sodium hydroxide, and hydrochloric acid, and mixtures thereof, and preferably sodium hydroxide and/or hydrochloric acid. These acidic and/or basic pH modifying agents are added to adjust the formulation to the target acceptable pH range. Hence it may not be necessary to use both acid and base—depending on the formulation, the addition of one of the acid or base may be sufficient to bring the mixture to the desired pH range. The aqueous vehicle may also contain a buffering agent to stabilize the pH. When used, the buffer is selected from the group consisting of a phosphate buffer (such as sodium dihydrogen phosphate and disodium hydrogen phosphate), a borate buffer (such as boric acid, or salts thereof including disodium tetraborate), a citrate buffer (such as citric acid, or salts thereof including sodium citrate), and ε-aminocaproic acid, and mixtures thereof. The formulation may further comprise a wetting agent. Suitable classes of wetting agents include those selected from the group consisting of polyoxypropylene-polyoxyethylene block copolymers (poloxamers), polyethoxylated ethers of castor oils, polyoxyethylenated sorbitan esters (polysorbates), polymers of oxyethylated octyl phenol (Tyloxapol), polyoxyl 40 stearate, fatty acid glycol esters, fatty acid glyceryl esters, sucrose fatty esters, and polyoxyethylene fatty esters, and mixtures thereof. Oral compositions generally include an inert diluent or an edible pharmaceutically acceptable carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. Methods of Use The present application relates to methods for the use of compounds of the application. The compounds of the application have a useful pharmacological activity spectrum and are therefore particularly suitable for the prophylaxis and/or treatment of diseases or disorders. The present application provides a method of treating or preventing diseases or disorders, comprising administering a therapeutically effective amount of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, to a subject in need thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for the treatment or prevention of diseases or disorders. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for treating or preventing diseases or disorders. In one embodiment, the disease or disorder is a condition which can be ameliorated by KCNQ2/3 potassium channel opening. In one embodiment, the disease or disorder is selected from epilepsy, neurotransmission disorder, CNS disorder, neurodegenerative disease (e.g., Alzheimer's disease, ALS, motor neuron disease, Parkinson's disease, macular degeneration, or glaucoma), cognitive disorder (e.g., degenerative dementia (including senile dementia, Alzheimer's disease, Pick's disease, Huntington's chorea, Parkinson's disease, and Creutzfeldt-Jakob disease); vascular dementia (including multi-infarct dementia); dementia associated with intracranial space occupying lesions, trauma, infections or related conditions (including HIV infection), metabolism, toxins, anoxia, or vitamin deficiency; mild cognitive impairment associated with ageing, particularly Age Associated Memory Loss, or learning deficiencies), bipolar disorder (e.g., Type I or II bipolar disorder), unipolar depression, anxiety, migraine, ataxia, myokimia, tinnitus, functional bowel disorders (e.g., non-ulcer dyspepsia, non-cardiac chest pain, or irritable bowel syndrome), cancer, inflammatory disease, ophthalmic disease (e.g., retinitis, retinopathies, uveitis, or acute injury to the eye tissue), asthma, allergic rhinitis, respiratory distress syndrome, gastrointestinal conditions (e.g., inflammatory bowel disease, Chron's disease, gastritis, irritable bowel syndrome, or ulcerative colitis), and inflammation in such diseases as vascular disease, migraine, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodoma, type I diabetes, myasthenia gravis, multiple sclerosis, sorcoidosis, nephrotic syndrome, Bechet's syndrome, polymyositis, gingivitis, conjunctivitis, and myocardial ischemia. In one embodiment, the application provides a method of producing an anti-epileptic, muscle relaxing, fever reducing, peripherally analagesic, and/or anti-convulsive effect in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for producing an anti-epileptic, muscle relaxing, fever reducing, peripherally analagesic, and/or anti-convulsive effect. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for producing an anti-epileptic, muscle relaxing, fever reducing, peripherally analagesic, and/or anti-convulsive effect. In one embodiment, the application provides compounds that are useful as an anticonvulsant. They are therefore useful in treating or preventing epilepsy. Compounds of the application may be used to improve the condition of a host, typically a human being, suffering from epilepsy. They may be employed to alleviate the symptoms of epilepsy in a host. “Epilepsy” is intended to include the following seizures: simple partial seizures, complex partial seizures, secondary generalized seizures, generalized seizures including absence seizures, myoclonic seizures, clonic seizures, tonic seizures, tonic clonic seizures and atonic seizures. Partial-onset seizures are the most common type of seizure in adult patients. For partial seizures, there is a focal epileptic zone (site of seizure onset), and seizure activity is initially limited to one hemisphere. Partial seizures can be further sub-divided into simple partial (without impairment of consciousness), complex partial (with impairment of consciousness with or following a simple partial onset) and secondarily generalized (i.e., partial seizures, either simple or complex, which evolve to generalized tonic-clonic seizures). Simple partial seizures, depending on the anatomical site of origin of the seizure, may have motor, somatosensory or special sensory, autonomic or psychic signs or symptoms. In one embodiment, the application provides a method of treating a subject suffering from or susceptible to epilepsy, comprising administering to the subject an effective amount of a compound of the application or a pharmaceutically acceptable salt or solvate thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject suffering from or susceptible to epilepsy for the treatment of epilepsy. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for treating a subject suffering from or susceptible to epilepsy. In one embodiment, the application provides a method for the adjunctive treatment of adults with partial-onset seizures, comprising administering to the subject an effective amount of a compound of the application or a pharmaceutically acceptable salt thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for adjunctive treatment of adults with partial-onset seizures. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for adjunctive treatment of adults with partial-onset seizures. In one embodiment, the present application provides a method of treating or preventing epilepsy, comprising administering a therapeutically effective amount of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, to a subject in need thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for the treatment or prevention of epilepsy. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for treating or preventing epilepsy. In one embodiment, a compound of the application is administered in combination with one or more anti-epileptic drugs (AEDs). There are different types of AEDs. For example, narrow-spectrum AEDs include phenytoin (Dilantin), phenobarbital, carbamazepine (Tegretol), oxcarbazepine (Trileptal), gabapentin (Neurontin), pregabalin (Lyrica), lacosamide (Vimpat), and vigabatrin (Sabril). Broad spectrum AEDs include valproic acid (Depakote), lamotrigine (Lamictal), topiramate (Topamax), zonisamide (Zonegran), levetiracetam (Keppra), clonazepam (Klonopin), and rufinamide (Banzel). In one embodiment, the AED is any AED. In one embodiment, the AED is a narrow spectrum AED. In one embodiment, the AED is a broad spectrum AED In one embodiment, the application provides compounds that are useful as analgesics. The compounds are therefore useful in treating or preventing pain. They may be used to improve the condition of a host, typically a human being, suffering from pain. They may be employed to alleviate pain in a host. Thus, the compounds may be used as a pre-emptive analgesic to treat acute pain such as musculoskeletal pain, post-operative pain and surgical pain, chronic pain such as chronic inflammatory pain (e.g., rheumatoid arthritis and osteoarthritis), neuropathic pain (e.g., post herpetic neuralgia, trigeminal neuralgia and sympathetically maintained pain) and pain associated with cancer and fibromyalgia. The compounds may also be used in the treatment or prevention of pain associated with migraine. The compounds may also be used in the treatment of the pain (both chronic and acute), fever and inflammation of conditions such as rheumatic fever; symptoms associated with influenza or other viral infections, such as the common cold; lower back and neck pain; headache; toothache; sprains and strains; myositis; neuralgia; synovitis; arthritis, including rheumatoid arthritis; degenerative joint diseases, including osteoarthritis; gout and ankylosing spondylitis; tendinitis; bursitis; skin related conditions, such as psoriasis, eczema, burns and dermatitis; injuries, such as sports injuries and those arising from surgical and dental procedures. In one embodiment, the application provides a method of producing an analgesic effect in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the application or a pharmaceutically acceptable salt or solvate thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for producing an analgesic effect. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for producing an analgesic effect. In one embodiment, the analgesic effect is a neuroprotective effect. In one embodiment, the analgesic effect is a centrally acting analgesic effect. In one embodiment, the application provides a method of treating or preventing a neurotransmission disorder, CNS disorder, neurodegenerative disease (e.g., Alzheimer's disease, ALS, motor neuron disease, Parkinson's disease, macular degeneration and glaucoma), cognitive disorder, bipolar disorder (e.g., Type I or II bipolar disorder), unipolar depression, or anxiety in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the application or a pharmaceutically acceptable salt or solvate thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for treating or preventing a neurotransmission disorder, CNS disorder, neurodegenerative disease (e.g., Alzheimer's disease, ALS, motor neuron disease, Parkinson's disease, macular degeneration and glaucoma), cognitive disorder, bipolar disorder (e.g., Type I or II bipolar disorder), unipolar depression, or anxiety. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for treating or preventing a neurotransmission disorder, CNS disorder, neurodegenerative disease (e.g., Alzheimer's disease, ALS, motor neuron disease, Parkinson's disease, macular degeneration and glaucoma), cognitive disorder, bipolar disorder (e.g., Type I or II bipolar disorder), unipolar depression, or anxiety. In one embodiment, the application provides a method of treating or preventing migraine, ataxia, myokimia, tinnitus, and functional bowel disorders (e.g., non-ulcer dyspepsia, non-cardiac chest pain, or irritable bowel syndrome) in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the application or a pharmaceutically acceptable salt or solvate thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for treating or preventing migraine, ataxia, myokimia, tinnitus, and functional bowel disorders (e.g., non-ulcer dyspepsia, non-cardiac chest pain, or irritable bowel syndrome). The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for treating or preventing migraine, ataxia, myokimia, tinnitus, and functional bowel disorders (e.g., non-ulcer dyspepsia, non-cardiac chest pain, or irritable bowel syndrome). In one embodiment, the application provides compounds that are useful in the treatment of CNS disorders such as bipolar disorder, alternatively known as manic depression. The compounds may thus be used to improve the condition of a human patient suffering from bipolar disorder. They may be used to alleviate the symptoms of bipolar disorder in a host. The compounds may also be used in the treatment of unipolar depression, ataxia, myokimia and anxiety. In one embodiment, the application provides compounds that are useful in the treatment of neurodegenerative diseases, such as Alzheimer's disease, ALS, motor neuron disease, Parkinson's disease, macular degeneration and glaucoma. The compounds of the application may also be useful in neuroprotection and in the treatment of neurodegeneration following stroke, cardiac arrest, pulmonary bypass, traumatic brain injury, spinal cord injury or the like. In one embodiment, compounds of the application are further useful in the treatment of tinnitus. In one embodiment, the application provides compounds that are useful in the treatment of functional bowel disorders which include non-ulcer dyspepsia, non-cardiac chest pain and in particular irritable bowel syndrome. Irritable bowel syndrome is a gastrointestinal disorder characterized by the presence of abdominal pain and altered bowel habits without any evidence of organic disease. The compounds may thus be used to alleviate pain associated with irritable bowel syndrome. The condition of a human patient suffering from irritable bowel syndrome may thus be improved. In one embodiment, the application provides a method of preventing or reducing dependence on, or preventing or reducing tolerance, or reverse tolerance, to a dependence-inducing agent in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the application or a pharmaceutically acceptable salt or solvate thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for preventing or reducing dependence on, or preventing or reducing tolerance, or reverse tolerance, to a dependence-inducing agent. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for preventing or reducing dependence on, or preventing or reducing tolerance, or reverse tolerance, to a dependence-inducing agent. Examples of dependence inducing agents include opioids (e.g., morphine), CNS depressants (e.g., ethanol), psychostimulants (e.g., cocaine) and nicotine. In one embodiment, the application provides a method of treating or preventing cancer, inflammatory disease, or ophthalmic disease in a subject in need thereof comprising administering to the subject an effective amount of a compound of the application or a pharmaceutically acceptable salt or solvate thereof. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for treating or preventing cancer, inflammatory disease, or ophthalmic disease. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for treating or preventing cancer, inflammatory disease, or ophthalmic disease. In one embodiment, the application provides compounds that inhibit cellular and neoplastic transformation and metastatic tumor growth and hence are useful in the treatment of certain cancerous diseases, such as colonic cancer. In one embodiment, the application provides compounds that inhibit inflammatory processes and therefore are of use in the treatment of asthma, allergic rhinitis and respiratory distress syndrome; gastrointestinal conditions such as inflammatory bowel disease, Chron's disease, gastritis, irritable bowel syndrome and ulcerative colitis; and the inflammation in such diseases as vascular disease, migraine, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodoma, type I diabetes, myasthenia gravis, multiple sclerosis, sorcoidosis, nephrotic syndrome, Bechet's syndrome, polymyositis, gingivitis, conjunctivitis and myocardial ischemia. In one embodiment, the application provides compounds that are useful in the treatment of ophthalmic diseases such as retinitis, retinopathies, uveitis, and acute injury to the eye tissue. In one embodiment, the application provides compounds that are useful for the treatment of cognitive disorders such as dementia, particularly degenerative dementia (including senile dementia, Alzheimer's disease, Pick's disease, Huntington's chorea, Parkinson's disease and Creutzfeldt-Jakob disease), and vascular dementia (including multi-infarct dementia), as well as dementia associated with intracranial space occupying lesions, trauma, infections and related conditions (including HIV infection), metabolism, toxins, anoxia and vitamin deficiency; and mild cognitive impairment associated with ageing, particularly Age Associated Memory Loss; and learning deficiencies. In one embodiment, the application provides a method of producing an anxiolytic effect in a subject in need thereof comprising administering to the subject an effective amount of a compound of the application or a pharmaceutically acceptable salt or solvate thereof. In one embodiment, the application provides a method for the treatment of anxiety and its related psychological and physical symptoms. Anxiolytics have been shown to be useful in the treatment of anxiety disorders. The present application also provides the use of a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for the preparation of a medicament for administration to a subject for producing an anxiolytic effect. The present application also provides a compound of the application, or a pharmaceutically acceptable salt or solvate thereof, for producing an anxiolytic effect. In one embodiment, the application provides compounds for treatment. In one embodiment, the application provides compounds for prophylaxis. In one embodiment, the application provides compound for alleviation of established symptoms. Administration may for example be in the form of tablets, capsules, pills, coated tablets, suppositories, ointments, gels, creams, powders, dusting powders, aerosols or in liquid form. Liquid application forms that may for example be considered are: oils or alcoholic or aqueous solutions as well as suspensions and emulsions. In one embodiment, the application provides forms of application that are tablets that contain between 30 and 60 mg or solutions that contain between 0.1 to 5 percent by weight of active substance. In one embodiment, a compound of the application is used in human medicine. In one embodiment, the compound of the application is used in veterinary medicine. In one embodiment, a compound of the application is used in agriculture. In one embodiment, a compound of the application is used alone or mixed with other pharmacologically active substances. The following Examples are illustrative and should not be interpreted in any way so as to limit the scope of the application. EXAMPLES Example 1a: Compound 1 Step 1: Synthesis of Compound a To a stirred solution of 4-fluorobenzylamine (1 equivalent (equiv)) dissolved in dimethylformamide (DMF) is added allyl bromide (1.5 equiv) and diisopropyl ethylamine (2 equiv) dropwise, and the resulting mixture is heated to 80° C. After 2 hours, the reaction mixture is cooled, diluted with water, and extracted with ethyl acetate (EtOAc). The organic layer is then washed with saturated brine, dried with anhydrous sodium sulfate, filtered, and concentrated under vacuum. The resulting residue is purified by silica gel liquid chromatography to provide Compound a. Step 2: Synthesis of Compound b To a stirred suspension of 2,3-difluoro-6-nitroaniline (1 equiv) in dry dimethyl sulfoxide (DMSO) is added Compound a (3 equiv) followed by Et3N (1.2 equiv) and 12 (catalytic amount). The resulting mixture is heated to 120° C. and stirred at 120° C. for 24 hours. Upon complete consumption of the starting material (as determined by thin layer chromatography (TLC)), the reaction mixture is cooled to RT, diluted with water (25 mL), and extracted with EtOAc (2×25 mL). The combined organic layers are dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a crude product, which is purified by silica gel column chromatography to afford Compound b. Step 3: Synthesis of Compound 1 To a stirred solution of Compound b (1 equiv) in methanol is added zinc powder (5 equiv) followed by the dropwise addition of ammonium chloride solution (5 equiv). After stirring at room temperature (RT) for 5 hours, N,N-diisopropylethylamine (DIPEA) (1.25 equiv) and ethyl chloroformate (1 equiv) are then added at 10° C., and the stirring is continued for another 3 hours at RT. Upon complete consumption of the starting material (as determined by TLC), the reaction mixture is diluted with water and stirred for 1 hour to give a solid product. The obtained solid is filtered, dissolved in EtOAc, and any un-dissolved solid is removed by filtration. The filtrate is concentrated to provide Compound 1 which is crystallized using n-hexane. Example 1b: Compound 1 Step 1: Synthesis of 2-Fluoro-N1-(4-fluorobenzyl)-4-nitrobenzene-1,3-diamine 2,3-Difluoro-6-nitroaniline (10.0 g, 79.9 mmole) was dissolved in anhydrous dimethylsulfoxide (90 mL). 4-fluorobenzylamine (9.3 g, 53.3 mmole) was added triethylamine (17.7 mL) and solid iodine (80 mg) were added and the mixture was heated at reflux for 4 h. under argon. The reaction was dissolved in ethyl acetate (200 mL) and extracted with water (3×100 mL). A yellow solid precipitated out of the organic layer to give 2-fluoro-M-(4-fluorobenzyl)-4-nitrobenzene-1,3-diamine (13.6 g, 91% yield). Step 2: Synthesis of di-tert-butyl (3-fluoro-4-((4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate 2-Fluoro-N1-(4-fluorobenzyl)-4-nitrobenzene-1,3-diamine (13.55 g, 48.53 mmole) was dissolved in methanol (60 mL) and tetrahydrofuran (60 mL). The mixture was cooled in an ice bath and zinc powder (31.70 g, 485.3 mmole) was added followed by ammonium chloride (26.0 g, 485.3 mmole) in DI water (64 mL) over 30 min. Ethyl acetate (200 mL) was added and the mixture was extracted with water (200 mL) and the organic layer was evaporated to dryness. The residue was dissolved in tetrahydrofuran (200 mL) and di-tert-butyldicarbonate (15.9 g, 72.8 mmole) was added followed by solid sodium bicarbonate (8.15 g, 97.06 mmole) and then DI water (150 mL). The reaction was stirred for an 18 h. at ambient temperature. The reaction was filtered and evaporated to dryness. Ethyl acetate (200 mL) was added and then 3M NH4OH (2×200 mL). The organic layer was evaporated to dryness. It was chromatographed on a silica gel column (200 g) packed in hexane. The column polarity was increased to 16% ethyl acetate over 5 CV, held at 16% ethyl acetate for 2 CV, increased to 32% ethyl acetate over 4 CV, and then to 53% ethyl acetate over 2 CV. Flow rate at 100 mL/min. t 100 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give di-tert-butyl (3-fluoro-4-(4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate (9.84 g, 45% yield). Step 3: Synthesis of di-tert-butyl (4-(allyl(4-fluorobenzyl)amino)-3-fluoro-1,2-phenylene)dicarbamate Di-tert-butyl (3-fluoro-4-((4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate (2.03 g, 4.52 mmole) was dissolved in anhydrous dimethylformamide (10 mL). Diisopropylethylamine (1.6 mL, 9.0 mmole) was added followed by allyl bromide (0.710 mg, 5.87 mmole). The mixture was heated in an 110° C. oil bath under argon for 6h. The reaction was diluted in ethyl acetate (100 mL) and extracted with water (100 mL). The aqueous layer was washed with ethyl acetate (100 mL). The organic layers were washed with water, 2×50 mL and then brine (50 mL) and filtered through a 1 PS filter to dry and evaporated to dryness. The crude material was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 9% ethyl acetate over 4 CV, held at 9% ethyl acetate over 7 CV and then increased to 33% ethyl acetate over 12 CV. The flow rate was 25 mL/min. Fractions (22 mL each) containing the first product were pooled and stripped to give of di-tert-butyl (4-(allyl(4-fluorobenzyl)amino)-3-fluoro-1,2-phenylene)dicarbamate (1.35 g, 61% yield). Step 4: Synthesis of ethyl (4-(allyl(4-fluorobenzyl)amino)-2-amino-3-fluorophenyl)carbamate (Compound 1) The organic layer was evaporated to dryness, dissolved in methanol (5 mL) and tetrahydrofuran (5 mL) and cooled in an ice bath when N, N-diisopropylethylamine (1.2 mL, 6.72 mmole) was added followed by ethyl chloroformate (0.128 mL, 1.19 mmole) dropwise. The reaction was stirred at ambient temperature for 0.5 h and was evaporated to dryness. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 30% ethyl acetate in chloroform over 7 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (4-(allyl(4-fluorobenzyl)amino)-2-amino-3-fluorophenyl)carbamate (0.115 g, 29% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.28 (m, 2H), 6.99 (t, 2H), 6.83 (d, 1H), 6.38 (t, 1H), 6.25 (br s, 1H), 5.80-5.89 (m, 1H), 5.18 (t, 2H), 4.30 (s, 2H), 4.25 (q, 2H), 3.84 (br s, 1H), 3.70 (d, 2H), 1.60 (br s, 1H), 1.32 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd1=8.2 min. Example 2a: Compound 2 Step 1: Synthesis of Compound c To a stirred solution of 4-trifluoromethylbenzylamine (1 equiv) dissolved in DMF is added allyl bromide (1.5 equiv) and diisopropyl ethylamine (2 equiv) dropwise, and the resulting mixture is heated to 80° C. After 2 hours, the reaction mixture is cooled, diluted with water, and extracted with ethyl acetate. The organic layer is then washed with saturated brine, dried with anhydrous sodium sulfate, filtered, and concentrated under vacuum. The resulting residue is purified by silica gel liquid chromatography to give Compound c. Step 2: Synthesis of Compound d To a stirred suspension of 2, 3-difluoro-6-nitroaniline (1 equiv) in dry DMSO is added Compound c (3 equiv) followed by Et3N (1.2 equiv) and 12 (catalytic amount). The reaction mixture is heated to 120° C. and stirred at 120° C. for 24 h. Upon complete consumption of the starting material (as determined by TLC), the reaction mixture is cooled to RT, diluted with water (25 mL), and extracted with EtOAc (2×25 mL). The combined organic layers are dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a crude product, which is purified by silica gel column chromatography to afford Compound d. Step 3: Synthesis of Compound 2 To a stirred solution of Compound d (1 equiv) in methanol is added zinc powder (5 equiv) followed by the dropwise addition of ammonium chloride solution (5 equiv). After stirring at RT for 5 hours, DIPEA (1.25 equiv) and ethyl chloroformate (1 equiv) are then added to reaction mixture at 10° C., and the stirring is continued for another 3 hours at RT. Upon complete consumption of the starting material (as determined by TLC), the reaction mixture is diluted with water and stirred for 1h to give a solid product. The obtained solid is filtered, dissolved in EtOAc, and any un-dissolved solid is removed by filtration. The filtrate is concentrated to provide Compound 2 which is crystallized using n-hexane. Example 2b: Compound 2 Step 1: Synthesis of N-(4-(trifluoromethyl)benzyl)prop-2-en-1-amine 4-(Trifluoromethyl)benzylamine (8.76 g, 50 mmole) was cooled in an ice bath when allyl bromide (2.60 g, 20.0 mmole) in DCM (30 mL) was added dropwise over 1 h. The reaction was filtered and evaporated to dryness. It was chromatographed on a silica gel column (100 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 10 CV, at 50 mL/min. Fractions (22 mL each) containing the second band were pooled and stripped to give N-(4-(trifluoromethyl)benzyl)prop-2-en-1-amine (1.42 g, 10.5% yield). Step 2: Synthesis of N′-allyl-2-fluoro-4-nitro-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine 2,3-Difluoro-6-nitroaniline (0.377 g, 2.17 mmole) was dissolved in anhydrous dimethylsulfoxide (4 mL). N-(4-(trifluoromethyl)benzyl)prop-2-en-1-amine (0.700 g, 3.25 mmole) was added followed by triethylamine (0.722 mL) and solid iodine (1 mg). The mixture was heated at reflux for 18 h. under argon. The reaction was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was washed with 3×30 mL water and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 16 CV, at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give N1-allyl-2-fluoro-4-nitro-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.58 g, 71% yield). Step 3: Synthesis of ethyl (4-(allyl(4-(trifluoromethyl)benzyl)amino)-2-amino-3-fluorophenyl)carbamate (Compound 2) N1-allyl-2-fluoro-4-nitro-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.392 g, 1.06 mmole) was dissolved in methanol (10 mL). Zinc powder (347 mg, 5.30 mmole) was added followed by ammonium chloride (284 mg, 5.30 mmole) in DI water (1.0 mL). The mixture was stirred under argon at ambient temperature for 2 h. and then cooled to 10° C. in an ice bath. N, N-diisopropylethylamine (0.221 mL, 1.27 mmole) was added, followed by ethyl chloroformate, dropwise (285 mg, 2.66 mmole) and the reaction was stirred at ambient temperature for 18 h. The reaction was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was washed with 2×10 mL water and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (25 g) packed in chloroform. The column polarity was increased to 20% ethyl acetate/chloroform over 10 CV, at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (4-(allyl(4-(trifluoromethyl)benzyl)amino)-2-amino-3-fluorophenyl)carbamate (0.331 g, 51% yield). NMR Spectroscopy:1H NMR (CDCl3, 300 MHz): δ 7.56 (d, 2H), 7.43 (d, 2H), 7.24 (s, 1H), 6.85 (d, 1H), 6.34 (t, 1H), 6.20 (br s, 1H), 5.80-5.94 (m, 1H), 5.16-5.22 (m, 2H), 4.39 (s, 2H), 4.22 (q, 2H), 3.80-4.17 (br s, 1H), 3.75 (d, 2H), 1.33 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd2=12.2 min. Example 3: Compound 3 Step 1: Synthesis of N-(3-(trifluoromethyl)benzyl)prop-2-en-1-amine 3-(Trifluoromethyl)benzylamine (10.0 g, 57.1 mmole) was dissolved in anhydrous acetonitrile (36 mL). Potassium carbonate (7.90 g, 57.1 mmole) was added and the mixture was cooled in an ice bath when allyl bromide (4.91 g, 40.6 mmole) in anhydrous acetonitrile (3.5 mL) was added over 30 min. The reaction was stirred for 2 h at 0° C. and then warmed to ambient temperature. After 1 h, the mixture was cooled in an ice bath and allyl bromide (0.987 mL, 11.4 mmole) was added dropwise and then warmed to ambient temperature. It was filtered on a glass fiber filter and then evaporated. It was chromatographed on a silica gel column (100 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 6 CV, at 50 mL/min. Fractions (22 mL each) containing the second band were pooled and stripped to give N-(3-(trifluoromethyl)benzyl)prop-2-en-1-amine (3.0 g, 24% yield). Step 2: Synthesis of N1-allyl-2-fluoro-4-nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine 2,3-Difluoro-6-nitroaniline (270 mg, 2.32 mmole) was dissolved in anhydrous dimethylsulfoxide (5 mL). N-(3-(trifluoromethyl)benzyl)prop-2-en-1-amine (0.500 g, 2.32 mmole) was added triethylamine (0.708 mL) and solid iodine (1 mg). The mixture was heated at reflux for 18 h. under argon. The reaction was dissolved in dichloromethane (20 mL) and extracted with water (20 mL). The aqueous layer was washed with dichloromethane (20 mL). The organic layers were washed with water, 2×20 mL brine and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 40% ethyl acetate over 10 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give N1-allyl-2-fluoro-4-nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.320 g, 56% yield). Step 3: Synthesis of ethyl (4-(allyl(3-(trifluoromethyl)benzyl)amino)-2-amino-3-fluorophenyl)carbamate (Compound 3) N1-allyl-2-fluoro-4-nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.305 g, 0.826 mmole) was dissolved in methanol (4 mL) and tetrahydrofuran (4 mL). Zinc powder (0.540 g, 8.26 mmole) was added followed by ammonium chloride (442 mg, 8.26 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 15 min. The reaction was cooled in an ice bath and N, N-diisopropylethylamine (0.330 mL, 1.89 mmole) was added followed by ethyl chloroformate (159 mg, 1.49 mmole) dropwise. The reaction was stirred at ambient temperature for 12 h. The reaction was cooled in an ice bath and N, N-diisopropylethylamine (0.460 mL, 2.63 mmole) was added followed by ethyl chloroformate (244 mg, 2.30 mmole) dropwise. The reaction was stirred at ambient temperature for 18 h and was filtered and evaporated. The crude oil was dissolved in ethyl acetate (20 mL) and extracted with water (20 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 45% ethyl acetate in chloroform over 15 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (4-(allyl(3-(trifluoromethyl)benzyl)amino)-2-amino-3-fluorophenyl)carbamate (0.119 g, 30% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.57 (s, 1H), 7.48 (m, 2H), 7.41 (t, 1H), 6.84 (d, 1H), 6.33 (t, 1H), 6.19 (br s, 1H), 5.80-5.89 (m, 1H), 5.16 (s, 1H), 5.13 (d, 1H), 4.35 (s, 2H), 4.21 (q, 2H), 3.85 (br s, 2H), 3.70 (d, 2H), 1.30 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd3=11.8 min. Example 4: Compound 4 Step 1: Synthesis of di-tert-butyl (4-(allyl(4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate Di-tert-butyl (4-((4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate (0.937 g, 2.17 mmole) was dissolved in anhydrous dimethylformamide (10 mL). Diisopropylethylamine (0.755 mL, 4.34 mmole) was added followed by allyl bromide (0.237 mL, 2.82 mmole). The mixture was heated in an 110° C. oil bath under argon for 2h. The reaction was diluted in ethyl acetate (100 mL) and extracted with water (100 mL). The aqueous layer was washed with ethyl acetate (100 mL). The organic layers were washed with water, 2×50 mL and then brine (50 mL) and filtered through a 1 PS filter to dry and evaporated to dryness. The crude material was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 12% ethyl acetate over 6 CV, held at 12% ethyl acetate over 2 CV and then increased to 40% ethyl acetate over 14 CV. The flow rate was 25 mL/min. Fractions (22 mL each) containing the first product were pooled and stripped to give di-tert-butyl (4-(allyl(4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate (0.780 g, 76% yield). Step 2: Synthesis of ethyl (4-(allyl(4-fluorobenzyl)amino)-2-aminophenyl)carbamate (Compound 4) Di-tert-butyl (4-(allyl(4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate (0.700 g, 1.48 mmole) was dissolved in dichloromethane (7 mL) and trifluoroacetic acid (7 mL) was added and the reaction was stirred at ambient temperature under argon for 65 min. The organic layer was evaporated to dryness, dissolved in methanol (7 mL) and tetrahydrofuran (7 mL) and cooled in an ice bath when N, N-diisopropylethylamine (1.6 mL, 9.20 mmole) was added followed by ethyl chloroformate (0.175 g, 1.63 mmole) dropwise. The reaction was stirred at ambient temperature for 1 h and was evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 39% ethyl acetate over 6CV, held at 39% ethyl acetate over 2 CV and then increased to 100% ethyl acetate over 9 CV. Flow rate at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (4-(allyl(4-fluorobenzyl)amino)-2-aminophenyl)carbamate (0.209 g, 41% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.21 (t, 2H), 7.00 (m, 3H), 6.18 (m, 3H), 5.90 (m, 1H), 5.21 (m, 2H), 4.47 (s, 2H), 4.20 (q, 2H), 3.95 (s, 3H), 1.30 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd4=6.4 min. Example 5—Compound 5 Step 1: Synthesis of N1-allyl-4-nitro-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine 5-Fluoro-2-nitroaniline (0.566 g, 3.62 mmole) was dissolved in anhydrous dimethylsulfoxide (5 mL). N-(4-(trifluoromethyl)benzyl)prop-2-en-1-amine (1.17 g, 5.44 mmole) was added triethylamine (1.80 mL) and solid iodine (1 mg) were added and the mixture was heated at reflux for an additional 18 h. under argon. The reaction was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was washed with 3×30 mL water and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (50 g) packed in hexanes. The column polarity was increased to 40% ethyl acetate/chloroform over 8 CV, at 50 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give N1-allyl-4-nitro-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.182 g, 14% yield). Step 2: Synthesis of ethyl (4-(allyl(4-(trifluoromethyl)benzyl)amino)-2-aminophenyl)carbamate (Compound 5) N1-allyl-4-nitro-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.182 g, 0.518 mmole) was dissolved in methanol (3 mL). Zinc powder (169 mg, 2.59 mmole) was added followed by ammonium chloride (139 mg, 2.59 mmole) in DI water (1.0 mL). The mixture was stirred under argon at ambient temperature for 30 min., and then zinc powder (169 mg, 2.59 mmole) was added followed by ammonium chloride (139 mg, 2.59 mmole) in DI water (1.0 mL) and tetrahydrofuran (3 mL). After 30 min., the mixture was cooled in an ice bath. N, N-diisopropylethylamine (0.378 mL, 2.18 mmole) was added, followed by ethyl chloroformate, dropwise (166 mg, 1.55 mmole) and the reaction was stirred at ambient temperature for 18 h. The reaction was filtered on a Buchner funnel with #4 Whatman filter paper. The filtrate was diluted with ethyl acetate (15 mL) and extracted with water (15 mL). The organic layer was washed with brine (15 mL), dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10g) packed in chloroform. The column polarity was increased to 3% ethyl acetate/chloroform over 2 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (4-(allyl(4-(trifluoromethyl)benzyl)amino)-2-aminophenyl)carbamate (0.017 g, 8.3% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.59 (d, 2H), 7.38 (d, 2H), 6.96 (m, 1H), 6.18 (m, 2H), 6.08 (br s, 1H), 5.87 (m, 1H), 5.21 (m, 2H), 4.55 (s, 2H), 4.22 (q, 2H), 3.99 (s, 2H), 1.32 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd5=9.3 min. Example 6: Compound 6 Step 1: Synthesis of 4-Nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine 5-Fluoro-2-nitroaniline (10.24 g, 58.46 mmole) was dissolved in anhydrous dimethylsulfoxide (90 mL). 3-fluorobenzylamine (6.1 g, 39.0 mmole) was added triethylamine (13.0 mL) and solid iodine (90 mg) were added and the mixture was heated at reflux for 4 h. under argon. The reaction was dissolved in ethyl acetate (200 mL) and extracted with water (3×200 mL). The combined aqueous layers were washed with (300 mL) ethyl acetate, combined and then evaporated to dryness. The crude material was triturated with hexane/ethyl acetate (7:3, 100 mL) and dried under high vacuum to give 4-nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine (8.29 g, 77% yield). Step 2: Synthesis of di-tert-butyl (4-((tert-butoxycarbonyl)(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate 4-Nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine (10.8 g, 34.7 mmole) was dissolved in methanol (50 mL) and tetrahydrofuran (50 mL). Zinc powder (22.7 g, 347 mmole) was added followed by ammonium chloride (18.6 g, 347 mmole) in DI water (46 mL) over 30 min. The mixture was stirred under argon at ambient temperature for 30 min. The reaction was filtered on a celite pad which was washed with methanol (200 mL) and the mixture was evaporated to dryness. Ethyl acetate (200 mL) was added and the mixture was extracted with water (200 mL) and brine (50 mL) and evaporated to dryness. The residue was dissolved in tetrahydrofuran (150 mL) and di-tert-butyldicarbonate (22.1 g, 101.3 mmole) was added followed by solid sodium bicarbonate (11.63 g, 138.4 mmole) and then DI water (100 mL). The reaction was stirred for an 18 h. at ambient temperature. The reaction was evaporated to dryness. and ethyl acetate (200 mL) was added. The organic layer was extracted with water (3×200 mL) and brine (50 mL) and evaporated to dryness. It was chromatographed on a silica gel column (200 g) packed in hexane. The column polarity was increased to 35% ethyl acetate in hexanes over 9 CV, at 100 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give di-tert-butyl (4-((tert-butoxycarbonyl)(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (13.1 g, 65% yield). Step 3: Synthesis of di-tert-butyl (4-((3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate Di-tert-butyl (4-((tert-butoxycarbonyl)(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (13.0 g, 22.4 mmole) was dissolved in dichloromethane (75 mL) and trifluoroacetic acid (50 mL) was added and the reaction was stirred at ambient temperature under argon for 60 min. The reaction was evaporated to give an off-white solid. The solid was dissolved in dioxane (125 mL) and di-tert-butyldicarbonate (10.24 g, 46.94 mmole) was added followed by solid sodium bicarbonate (7.51 g, 89.4 mmole) and then DI water (50 mL). The reaction was heated to 40° C. with stirring for 18 h., under argon. The reaction was evaporated and ethyl acetate (200 mL) was added. The organic layer was extracted with 3M NH4OH (2×100 mL) and brine (50 mL), dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (200 g) packed in hexane. The column polarity was increased to 45% ethyl acetate in hexanes over 12 CV, at 100 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give di-tert-butyl (4-((3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (1.3 g, 65% yield). Step 4: Synthesis of di-tert-butyl (4-(allyl(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate Di-tert-butyl (4-((3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (2.0 g, 4.2 mmole) was dissolved in anhydrous dimethylformamide (20 mL). Diisopropylethylamine (2.2 mL, 12.5 mmole) was added followed by allyl bromide (0.803 mL, 9.55 mmole). The mixture was heated in an 110° C. oil bath under argon for 2h. The reaction was diluted in ethyl acetate (200 mL) and extracted with water (200 mL), and then brine (50 mL) and filtered through a 1 PS filter to dry and evaporated to dryness. The crude material was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 37% ethyl acetate in hexanes over 14 CV. The flow rate was 25 mL/min. Fractions (22 mL each) containing the first product were pooled and stripped to give of di-tert-butyl (4-(allyl(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (1.63 g, 75% yield). Step 5: Synthesis of ethyl (4-(allyl(3-(trifluoromethyl)benzyl)amino)-2-aminophenyl)carbamate (Compound 6) Di-tert-butyl (4-(allyl(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (1.0 g, 1.92 mmole) was dissolved in dichloromethane (7 mL) and trifluoroacetic acid (7 mL) was added and the reaction was stirred at ambient temperature under argon for 90 min. The organic layer was evaporated to dryness, dissolved in methanol (5 mL) and tetrahydrofuran (5 mL) and cooled in an ice bath when N, N-diisopropylethylamine (2.1 mL, 11.9 mmole) was added followed by ethyl chloroformate (0.225 mL, 2.11 mmole) dropwise. The reaction was stirred at ambient temperature for 18 h and was filtered and evaporated. The crude oil was dissolved in ethyl acetate (20 mL) and extracted with water (20 mL). The organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (25 g) packed in chloroform. The column polarity was increased to 100% ethyl acetate in hexanes over 26 CV, at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (4-(allyl(3-(trifluoromethyl)benzyl)amino)-2-aminophenyl)carbamate (0.150 g, 20% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.48 (s, 2H), 7.41 (s, 2H), 6.92 (d, 1H), 6.15 (d, 1H), 6.07 (s, 1H), 6.00 (br s, 1H), 5.85 (m, 1H), 5.15-5.20 (m, 2H), 4.50 (s, 2H), 4.18 (q, 2H), 3.95 (s, 2H), 3.75 (br s, 2H), 1.26 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd6=8.8 min. Example 7: Compound 7 Step 1: Synthesis of 2-Fluoro-N1-(4-fluorobenzyl)-4-nitrobenzene-1,3-diamine 2,3-Difluoro-6-nitroaniline (10.0 g, 79.9 mmole) was dissolved in anhydrous dimethylsulfoxide (90 mL). 4-fluorobenzylamine (9.3 g, 53.3 mmole) was added triethylamine (17.7 mL) and solid iodine (80 mg) were added and the mixture was heated at reflux for 4 h. under argon. The reaction was dissolved in ethyl acetate (200 mL) and extracted with water (3×100 mL). A yellow solid precipitated out of the organic layer to give 2-fluoro-N1-(4-fluorobenzyl)-4-nitrobenzene-1,3-diamine (13.6 g, 91% yield). Step 2: Synthesis of di-tert-butyl (3-fluoro-4-((4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate 2-Fluoro-N1-(4-fluorobenzyl)-4-nitrobenzene-1,3-diamine (13.55 g, 48.53 mmole) was dissolved in methanol (60 mL) and tetrahydrofuran (60 mL). The mixture was cooled in an ice bath and zinc powder (31.70 g, 485.3 mmole) was added followed by ammonium chloride (26.0 g, 485.3 mmole) in DI water (64 mL) over 30 min. Ethyl acetate (200 mL) was added and the mixture was extracted with water (200 mL) and the organic layer was evaporated to dryness. The residue was dissolved in tetrahydrofuran (200 mL) and di-tert-butyldicarbonate (15.9 g, 72.8 mmole) was added followed by solid sodium bicarbonate (8.15 g, 97.06 mmole) and then DI water (150 mL). The reaction was stirred for an 18 h. at ambient temperature. The reaction was filtered and evaporated to dryness. Ethyl acetate (200 mL) was added and then 3M NH4OH (2×200 mL). The organic layer was evaporated to dryness. It was chromatographed on a silica gel column (200 g) packed in hexane. The column polarity was increased to 16% ethyl acetate over 5 CV, held at 16% ethyl acetate for 2 CV, increased to 32% ethyl acetate over 4 CV, and then to 53% ethyl acetate over 2 CV. Flow rate at 100 mL/min. t 100 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give di-tert-butyl (3-fluoro-4-((4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate (9.84 g, 45% yield). Step 3: Synthesis of di-tert-butyl (3-fluoro-4-((4-fluorobenzyl)(prop-2-yn-1-yl)amino)-1,2-phenylene)dicarbamate Di-tert-butyl (3-fluoro-4-((4-fluorobenzyl)amino)-1,2-phenylene)dicarbamate (2.00 g, 4.45 mmole) was dissolved in anhydrous dimethylformamide (20 mL), 80% propargyl bromide in toluene (0.618 mL, 5.78 mmole) and diisopropylethylamine (1.50 mL, 8.90 mmole). The mixture was heated in a 90° C. oil bath, under argon, for 0.5 h. 80% propargyl bromide in toluene (0.65 mL, 5.78 mmole) and diisopropylethylamine (1.50 mL, 8.90 mmole) were added and heated at 100° C. for 4.5h. 80% propargyl bromide in toluene (0.618 mL, 5.78 mmole) and diisopropylethylamine (1.50 mL, 8.90 mmole) were added and the reaction was heated at 100° for 18 h. 80% propargyl bromide in toluene (0.618 mL, 5.78 mmole) was added and the mixture was heated for 2 h. at 100° C. The reaction was diluted in ethyl acetate (100 mL) and extracted with water (100 mL) and brine (50 mL), filtered through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 40% ethyl acetate over 15 CV, at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give (1.01 g, 47% yield) di-tert-butyl (3-fluoro-4-(4-fluorobenzyl)(prop-2-yn-1-yl)amino)-1,2-phenylene)dicarbamate (1.35 g, 61% yield). Step 4: Synthesis of ethyl (2-amino-3-fluoro-4-((4-fluorobenzyl)(prop-2-yn-1-yl)amino)phenyl)carbamate (Compound 7) Di-tert-butyl (3-fluoro-4-((4-fluorobenzyl)(prop-2-yn-1-yl)amino)-1,2-phenylene)dicarbamate (0.480 g, 0.985 mmole) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) was added and the reaction was stirred at ambient temperature under argon for 90 min. The reaction was evaporated to give red oil which was dissolved in methanol (5 mL) and tetrahydrofuran (5 mL) and cooled in an ice bath when N, N-diisopropylethylamine (1.2 mL, 6.1 mmole) was added followed by ethyl chloroformate (129 mg, 1.2 mmole) dropwise. The reaction was stirred at ambient temperature for 18 h and was filtered and evaporated. The crude oil was dissolved in ethyl acetate (20 mL) and extracted with water (20 mL). The organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 30% ethyl acetate in chloroform over 7 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-3-fluoro-4-((4-fluorobenzyl)(prop-2-yn-1-yl)amino)phenyl)carbamate (0.073 g, 20% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.41 (m, 2H), 7.12 (t, 2H), 6.88 (m, 1H), 6.61 (m, 1H), 6.22 (br s, 1H), 4.30 (s, 2H), 4.25 (m, 2H), 3.88 (s, 2H), 3.78 (s, 2H), 2.28 (s, 1H), 1.35 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd7=10.6 min. Example 8—Compound 8 Step 1: Synthesis of N-(4-(trifluoromethyl)benzyl)prop-2-yn-1-amine 4-(Trifluoromethyl)benzylamine (6.93 g, 40 mmole) was dissolved in anhydrous acetonitrile (18 mL). Potassium carbonate (5.5 g, 40 mmole) was added and the mixture was cooled in an ice bath when 80% propargyl bromide in toluene (2.6 g, 20 mmole) was added over 10 min. The reaction was warmed to ambient temperature and stirred for 18 h. It was filtered and evaporated. The crude oil was dissolved in ethyl acetate (50 mL) and extracted with water (50 mL) and then brine (50 mL) and dried over sodium sulfate. It was chromatographed on a silica gel column (100g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 10 CV, at 50 mL/min. Fractions (22 mL each) containing the second band were pooled and stripped to give N-(4-(trifluoromethyl)benzyl)prop-2-yn-1-amine (3.0 g, 43% yield). Step 2: Synthesis of 2-Fluoro-4-nitro-N1-(prop-2-yn-1-yl)-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine 2,3-Difluoro-6-nitroaniline (0.681 g, 3.91 mmole) was dissolved in anhydrous dimethylsulfoxide (5 mL). N-(4-(trifluoromethyl)benzyl)prop-2-yn-1-amine (1.75 g, 8.21 mmole)) was added followed by triethylamine (2.3 mL) and solid iodine (2 mg). The mixture was heated at reflux for 18 h. under argon. The reaction was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The aqueous layer was washed with 2×10 mL dichloromethane. The organic layers were combined and washed with brine (30 mL) and evaporated to dryness. The crude material was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 100% chloroform over 20 CV, at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give of 2-fluoro-4-nitro-N1-(prop-2-yn-1-yl)-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.36 g, 25% yield). Step 3: Synthesis of ethyl (2-amino-3-fluoro-4-(prop-2-yn-1-yl(4-(trifluoromethyl)benzyl)amino)phenyl)carbamate (Compound 8) 2-Fluoro-4-nitro-N1-(prop-2-yn-1-yl)-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.350 g, 0.953 mmole) was dissolved in methanol (3 mL). Zinc powder (312 mg, 4.76 mmole) was added followed by ammonium chloride (255 mg, 4.76 mmole) in DI water (1.0 mL). The mixture was stirred under argon at ambient temperature for 18 h., and then zinc powder (312 mg, 4.76 mmole) were added followed by ammonium chloride (255 mg, 4.76 mmole) in DI water (1.0 mL). After 25 min., the mixture was cooled in an ice bath. N, N-diisopropylethylamine (0.364 mL, 2.01 mmole) was added, followed by ethyl chloroformate, dropwise (204 mg, 1.90 mmole) and the reaction was stirred at ambient temperature for 0.5 h. The reaction was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (50g) packed in chloroform. The column polarity was increased to 65% ethyl acetate/chloroform over 11 CV, at 50 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-3-fluoro-4-(prop-2-yn-1-yl(4-(trifluoromethyl)benzyl)amino)phenyl)-carbamate (0.087 g, 19% yield). NMR Spectroscopy:1H NMR (CDCl3, 300 MHz): 7.59 (q, 4H), 6.95 (d, 1H), 6.62 (t, 1H), 6.24 (br s, 1H), 4.40 (s, 2H), 4.24 (q, 2H), 3.80 (d, 2H), 2.05-2.50 (m, 2H), 1.38 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd8=12.3 min. Example 9: Compound 9 Step 1: Synthesis of N-(3-(trifluoromethyl)benzyl)prop-2-yn-1-amine 3-(Trifluoromethyl)benzylamine (5.0 g, 28.6 mmole) was dissolved in anhydrous acetonitrile (18 mL). Potassium carbonate (4.00 g, 28.6 mmole) was added and the mixture was cooled in an ice bath when 80% propargyl bromide in toluene (3.0 mL, 19 mmole) was added over 30 min. The reaction was warmed to ambient temperature and stirred for 2 h. It was filtered and evaporated. It was chromatographed on a silica gel column (50 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 9 CV, at 50 mL/min. Fractions (22 mL each) containing the second band were pooled and stripped to give N-(3-(trifluoromethyl)benzyl)prop-2-yn-1-amine (2.72 g, 45% yield). Step 2: Synthesis of 2-Fluoro-4-nitro-N1-(prop-2-yn-1-yl)-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine 2,3-Difluoro-6-nitroaniline (1.09 g, 6.26 mmole) was dissolved in anhydrous dimethylsulfoxide (8 mL). N-(3-(trifluoromethyl)benzyl)prop-2-yn-1-amine (2.00 g, 9.39 mmole) was added triethylamine (2.86 mL) and solid iodine (1 mg). The mixture was heated at reflux for 23 h. under argon. The reaction was dissolved in ethyl acetate (40 mL) and extracted with water (40 mL). The aqueous layer was washed with ethyl acetate (20 mL). The organic layers were washed with water, 2×20 mL brine and filtered through a 1 PS filter to dry. The crude material was chromatographed on a silica gel column (50 g) packed in hexanes. The column polarity was increased to 40% ethyl acetate over 15 CV, at 50 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 2-fluoro-4-nitro-N1-(prop-2-yn-1-yl)-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.393 g, 17% yield). Step 3: Synthesis of ethyl (2-amino-3-fluoro-4-(prop-2-yn-1-yl(3-(trifluoromethyl)benzyl)amino)phenyl)carbamate (Compound 9) 2-Fluoro-4-nitro-N1-(prop-2-yn-1-yl)-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine (0.380 g, 1.04 mmole) was dissolved in methanol (6 mL) and tetrahydrofuran (6 mL). Zinc powder (0.677 g, 10.4 mmole) was added followed by ammonium chloride (553 mg, 10.35 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 30 min. The reaction was cooled in an ice bath and N, N-diisopropylethylamine (0.515 mL, 2.96 mmole) was added followed by ethyl chloroformate (249 mg, 2.33 mmole) dropwise. The reaction was stirred at ambient temperature for 18 h and was filtered and evaporated. The crude oil was dissolved in ethyl acetate (20 mL) and extracted with water (20 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 20% ethyl acetate in chloroform over 7 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give of ethyl (2-amino-3-fluoro-4-(prop-2-yn-1-yl(3-(trifluoromethyl)benzyl)amino)phenyl)carbamate (0.249 g, 58% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.39-7.70 (m, 5H), 6.90 (d, 1H), 6.59 (t, 1H), 6.23 (br s, 1H), 4.35 (s, 2H), 4.19 (m, 2H), 3.56-3.92 (m, 3H), 2.25 (s, 1H), 1.25 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd9=12.0 min. Example 10: Compound 11 Step 1: Synthesis of 4-Nitro-N1-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine 5-Fluoro-2-nitroaniline (10.24 g, 58.46 mmole) was dissolved in anhydrous dimethylsulfoxide (90 mL). (4-(trifluoromethyl)phenyl)methanamine (6.1 g, 39.0 mmole) was added followed by triethylamine (13.0 mL) and solid iodine (70 mg). The mixture was heated at reflux for 1 h. under argon. Ethyl acetate (150 mL) was added and the organics were extracted with water (3×150 mL). The combined aqueous layers were washed with dichloromethane (300 mL), all organics were combined, filtered through a 1 PS filter and then evaporated to dryness. The crude material was dissolved in boiling ethyl acetate (20 mL) and hexanes were added to cloud point. Upon cooling, crystals formed. The solid was filtered off on a #54 Whatman filter paper on a Buchner filter and dried under high vacuum to give 4-nitro-M-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine (8.61 g, 71% yield). Step 2: Synthesis of di-tert-butyl (4-((4-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate 4-Nitro-M-(4-(trifluoromethyl)benzyl)benzene-1,3-diamine (8.17 g, 26.2 mmole) was dissolved in methanol (30 mL) and tetrahydrofuran (30 mL). The mixture was cooled in an ice bath and zinc powder (17.16 g, 262.5 mmole) was added followed by ammonium chloride (14.0 g, 262.5 mmole) in DI water (40 mL) over 60 min. The reaction was filtered on a celite pad and solids were washed with methanol (200 mL) and the filtrate was evaporated to dryness. Ethyl acetate (100 mL) was added and the mixture was extracted with water (100 mL) and then brine (50 mL) The organic layer was evaporated to dryness. The residue was dissolved in tetrahydrofuran (171 mL) and di-tert-butyldicarbonate (8.60 g, 39.36 mmole) was added followed by solid sodium bicarbonate (6.60 g, 78.75 mmole) and then DI water (86 mL). The reaction was stirred for an 18 h. at ambient temperature. The reaction was filtered and evaporated to dryness. Ethyl acetate (200 mL) was added and then washed with 3M NH4OH (2×200 mL). The organic layer was evaporated to dryness. It was chromatographed on a silica gel column (100 g) packed in hexane. The column polarity was increased to 60% ethyl acetate in hexanes over 14 CV. Flow rate at 50 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give di-tert-butyl (4-((4-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (5.9 g, 47% yield). Step 3: Synthesis of di-tert-butyl (4-(prop-2-yn-1-yl(4-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate Di-tert-butyl (4-((4-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (4.88 g, 10.13 mmole) was dissolved in anhydrous dimethylformamide (50 mL). 80% propargyl bromide in toluene (2.50 mL, 23.3 mmole) was added followed by diisopropylethylamine (5.30 mL, 30.4 mmole). The mixture was heated at reflux, under argon, for 2h. The reaction was evaporated to dryness and the crude material was chromatographed on a suction silica gel column (50 g) packed in hexanes. The column was washed with hexanes (250 mL), 10% ethyl acetate in hexanes (250 mL), 10% ethyl acetate in hexanes (250 mL), 20% ethyl acetate in hexanes (250 mL), 30% ethyl acetate in hexanes (500 mL), Fractions (125 mL each) containing the product were pooled and stripped to give di-tert-butyl (4-(prop-2-yn-1-yl(4-(trifluoromethyl)benzyl)-amino)-1,2-phenylene)dicarbamate (1.42 g, 27% yield). Step 4: Synthesis of ethyl (2-amino-4-(prop-2-yn-1-yl(4-(trifluoromethyl)benzyl)amino)phenyl)carbamate (Compound 11) Di-tert-butyl (4-(prop-2-yn-1-yl(4-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (1.40 g, 2.69 mmole) was dissolved in dichloromethane (15 mL) and trifluoroacetic acid (15 mL) was added and the reaction was stirred at ambient temperature under argon for 45 min. The organic layer was evaporated to dryness, dissolved in methanol (10 mL) and tetrahydrofuran (10 mL) and cooled in an ice bath when N, N-diisopropylethylamine (3.00 mL, 16.7 mmole) was added followed by ethyl chloroformate (0.317 g, 2.63 mmole) dropwise. The reaction was stirred at ambient temperature for 1 h and was evaporated. The crude oil was dissolved in ethyl acetate (40 mL) and extracted with water (40 mL). The organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 33% ethyl acetate over 7CV, held at 33% ethyl acetate over 4 CV and then increased to 84% ethyl acetate over 10 CV. Flow rate at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-4-(prop-2-yn-1-yl(4-(trifluoromethyl)benzyl)amino)phenyl)carbamate (0.462 g, 44% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.55 (d, 2H), 7.40 (d, 2H), 6.98 (d, 1H), 6.20-6.25 (m, 2H), 6.07 (br s, 1H), 4.53 (s, 2H), 4.18 (q, 2H), 3.95 (s, 2H), 3.78 (br s, 2H), 2.21 (s, 1H), 1.25 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd11=8.8 min. Example 11: Compound 12 Step 1: Synthesis of 4-Nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine 5-Fluoro-2-nitroaniline (10.24 g, 58.46 mmole) was dissolved in anhydrous dimethylsulfoxide (90 mL). 3-fluorobenzylamine (6.1 g, 39.0 mmole) was added triethylamine (13.0 mL) and solid iodine (90 mg) were added and the mixture was heated at reflux for 4 h. under argon. The reaction was dissolved in ethyl acetate (200 mL) and extracted with water (3×200 mL). The combined aqueous layers were washed with (300 mL) ethyl acetate, combined and then evaporated to dryness. The crude material was triturated with hexane/ethyl acetate (7:3, 100 mL) and dried under high vacuum to give 4-nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine (8.29 g, 77% yield). Step 2: Synthesis of di-tert-butyl (4-((tert-butoxycarbonyl)(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate 4-Nitro-N1-(3-(trifluoromethyl)benzyl)benzene-1,3-diamine (10.8 g, 34.7 mmole) was dissolved in methanol (50 mL) and tetrahydrofuran (50 mL). Zinc powder (22.7 g, 347 mmole) was added followed by ammonium chloride (18.6 g, 347 mmole) in DI water (46 mL) over 30 min. The mixture was stirred under argon at ambient temperature for 30 min. The reaction was filtered on a celite pad which was washed with methanol (200 mL) and the mixture was evaporated to dryness. Ethyl acetate (200 mL) was added and the mixture was extracted with water (200 mL) and brine (50 mL) and evaporated to dryness. The residue was dissolved in tetrahydrofuran (150 mL) and di-tert-butyldicarbonate (22.1 g, 101.3 mmole) was added followed by solid sodium bicarbonate (11.63 g, 138.4 mmole) and then DI water (100 mL). The reaction was stirred for an 18 h. at ambient temperature. The reaction was evaporated to dryness. and ethyl acetate (200 mL) was added. The organic layer was extracted with water (3×200 mL) and brine (50 mL) and evaporated to dryness. It was chromatographed on a silica gel column (200 g) packed in hexane. The column polarity was increased to 35% ethyl acetate in hexanes over 9 CV, at 100 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give di-tert-butyl (4-((tert-butoxycarbonyl)(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (13.1 g, 65% yield). Step 3: Synthesis of di-tert-butyl (4-((3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate Di-tert-butyl (4-((tert-butoxycarbonyl)(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (13.0 g, 22.4 mmole) was dissolved in dichloromethane (75 mL) and trifluoroacetic acid (50 mL) was added and the reaction was stirred at ambient temperature under argon for 60 min. The reaction was evaporated to give an off-white solid. The solid was dissolved in dioxane (125 mL) and di-tert-butyldicarbonate (10.24 g, 46.94 mmole) was added followed by solid sodium bicarbonate (7.51 g, 89.4 mmole) and then DI water (50 mL). The reaction was heated to 40° C. with stirring for 18 h., under argon. The reaction was evaporated and ethyl acetate (200 mL) was added. The organic layer was extracted with 3M NH4OH (2×100 mL) and brine (50 mL), dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (200 g) packed in hexane. The column polarity was increased to 45% ethyl acetate in hexanes over 12 CV, at 100 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give di-tert-butyl (4-((3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (1.3 g, 65% yield). Step 4: Synthesis of Di-tert-butyl (4-(prop-2-yn-1-yl(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate Di-tert-butyl (4-((3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (2.00 g, 4.15 mmole) was dissolved in anhydrous dimethylformamide (20 mL). Diisopropylethylamine (2.20 mL, 12.5 mmole) was added followed by 80% propargyl bromide in toluene (1.50 mL, 14.3 mmole). The mixture was heated in a 110° C. oil bath under argon for 2h. The reaction was diluted in ethyl acetate (200 mL) and extracted with water (200 mL). The organic layer was evaporated to dryness. The crude material was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 40% ethyl acetate over 15 CV, at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give di-tert-butyl (4-(prop-2-yn-1-yl(3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (1.76 g, 82% yield). Step 5: Synthesis of ethyl (2-amino-4-(prop-2-yn-1-yl(3-(trifluoromethyl)benzyl)amino) phenyl)carbamate (Compound 12) Di-tert-butyl (4-(prop-2-yn-1-yl (3-(trifluoromethyl)benzyl)amino)-1,2-phenylene)dicarbamate (1.0 g, 1.9 mmole) was dissolved in dichloromethane (7 mL) and trifluoroacetic acid (7 mL) was added and the reaction was stirred at ambient temperature under argon for 90 min. The organic layer was evaporated to dryness, dissolved in methanol (5 mL) and tetrahydrofuran (5 mL) and cooled in an ice bath when N, N-diisopropylethylamine (2.1 mL, 11.9 mmole) was added followed by ethyl chloroformate (0.225 mL, 2.11 mmole) dropwise. The reaction was stirred at ambient temperature for 18 h and was filtered and evaporated. The crude oil was dissolved in ethyl acetate (20 mL) and extracted with water (20 mL). The organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (25 g) packed in hexane. The column polarity was increased to 33% ethyl acetate over 4 CV, held at 33% ethyl acetate over 4 CV and then increased to 100% ethyl acetate over 13 CV. Flow rate at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-4-(prop-2-yn-1-yl(3-(trifluoromethyl)benzyl)amino)phenyl)-carbamate (0.360 g, 48% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.60 (s, 1H), 7.50 (m, 2H), 7.40-7.45 (m, 1H), 7.24 (s, 1H), 7.00 (t, 1H), 6.30 (d, 1H), 6.23 (s, 1H), 6.02 (br s, 1H), 4.60 (s, 2H), 4.2 (q, 2H), 3.87 (s, 2H), 2.30 (br s, H), 2.21 (s, 1H), 1.24 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd12=8.3 min. Example 12: Compound 13 Step 1: Synthesis of Compound e To a stirred suspension of 2, 3-difluoro-6-nitroaniline (1 equiv) in dry DMSO is added 5-fluoroisoindoline (3 equiv) followed by Et3N (1.2 equiv) and 12 (catalytic amount). The reaction mixture is heated to 120° C. and stirred at 120° C. for 24 h. Upon complete consumption of the starting material (as determined by TLC), the reaction mixture is cooled to RT, diluted with water (25 mL), and extracted with EtOAc (2×25 mL). The combined organic layers are dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a crude product, which is purified by silica gel column chromatography to afford Compound e. Step 2: Synthesis of Compound 13 To a stirred solution of Compound e (1 equiv) in methanol is added zinc powder (5 equiv) followed by the dropwise addition of ammonium chloride solution (5 equiv). After stirring at RT for 5 hours, DIPEA (1.25 equiv) and ethyl chloroformate (1 equiv) are then added to reaction mixture at 10° C., and the stirring is continued for another 3 hours at RT. Upon complete consumption of the starting material (as determined by TLC), the reaction mixture is diluted with water and stirred for 1 hour to give a solid product. The obtained solid is filtered, dissolved in EtOAc, and any un-dissolved solid is removed by filtration. The filtrate concentrated to provide Compound 13 which is crystallized using n-hexane. Example 13: Compound 13 Step 1: Synthesis of 2-Fluoro-3-(5-fluoroisoindolin-2-yl)-6-nitroaniline 2,3-Difluoro-6-nitroaniline (0.311 g, 1.78 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 5-fluoroisoindoline hydrochloride (0.465 g, 2.68 mmole) was added triethylamine (0.814 mL) and solid iodine (1 mg). The mixture was heated at reflux for 4 h. under argon. The reaction was slurried in 7:3 chloroform/isopropanol (5 mL) and filtered on a #54 Whatman filter on a buchner funnel. The solid was washed with 7:3 chloroform/isopropanol (2×5 mL) and dried at ambient temp. under high vacuum to give a white solid 2-fluoro-3-(5-fluoroisoindolin-2-yl)-6-nitroaniline (0.455 g, 88% yield). Step 2: Synthesis of Ethyl (2-amino-3-fluoro-4-(5-fluoroisoindolin-2-yl)phenyl)carbamate (Compound 13) 2-Fluoro-3-(5-fluoroisoindolin-2-yl)-6-nitroaniline (0.440 g, 1.51 mmole) was dissolved in methanol (5 mL) and tetrahydrofuran (5 mL). Zinc powder (0.988 g, 15.1 mmole) was added followed by ammonium chloride (808 mg, 15.1 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 45 min. The reaction was cooled in an ice bath and N, N-diisopropylethylamine (0.604 mL, 3.45 mmole) was added followed by ethyl chloroformate (291 mg, 2.72 mmole) dropwise. The reaction was stirred at ambient temperature for 12 h. The reaction was cooled in an ice bath and N, N-diisopropylethylamine (0.300 mL, 1.74 mmole) was added followed by ethyl chloroformate (150 mg, 1.30 mmole) dropwise. The reaction was stirred at ambient temperature for 2 h and was filtered and evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 45% ethyl acetate in chloroform over 14 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-3-fluoro-4-(5-fluoroisoindolin-2-yl)phenyl)carbamate (0.033 g, 6.3% yield). NMR Spectroscopy:1H NMR (CD3OD, 500 MHz): δ 7.30 (m, 1H), 7.05 (d, 1H), 6.98 (m, 1H), 6.80 (s, 1H), 6.20 (m, 1H), 4.68 (m, 4H), 4.18 (m, 2H), 1.25 (s, 4H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd13=10.1 min. Example 14a: Compound 14 Step 1: Synthesis of Compound f To a stirred suspension of 2, 3-difluoro-6-nitroaniline (1 equiv) in dry DMSO is added 5-trifluoromethylisoindoline (3 equiv) followed by Et3N (1.2 equiv) and I2(catalytic amount). The reaction mixture is heated to 120° C. and stirred at 120° C. for 24 hours. Upon complete consumption of the starting material (as determined by TLC), the reaction mixture is diluted with water (25 mL), and extracted with EtOAc (2×25 mL). The combined organic layers are dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a crude product, which is purified by silica gel column chromatography to afford Compound f. Step 2: Synthesis of Compound 14 To a stirred solution of Compound f (1 equiv) in methanol is added zinc powder (5 equiv) followed by the dropwise addition of ammonium chloride solution (5 equiv). After stirring at RT for 5 hours, DIPEA (1.25 equiv) and ethyl chloroformate (1 equiv) are added to reaction mixture at 10° C., and the stirring is continued for another 3 hours at RT. Upon complete consumption of the starting material (as determined by TLC), the reaction mixture is diluted with water and stirred for 1 hour to give a solid product. The obtained solid is filtered, dissolved in EtOAc, and any un-dissolved solid is removed by filtration. The filtrate is concentrated to provide Compound 14 which is crystallized using n-hexane. Example 14b: Compound 14 Step 1: Synthesis of 2-Fluoro-6-nitro-3-(5-(trifluoromethyl)isoindolin-2-yl)aniline 2,3-Difluoro-6-nitroaniline (0.233 g, 1.34 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 5-(trifluoromethyl)isoindoline hydrochloride (0.448 g, 2.00 mmole) was added triethylamine (0.61 mL) and solid iodine (1 mg). The mixture was heated at reflux for 2 h. under argon. The reaction was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The aqueous layer was washed with dichloromethane (10 mL), organics pooled and washed with brine (5 mL) and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in hexane. The column polarity was increased to 100% ethyl acetate over 12 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 2-fluoro-6-nitro-3-(5-(trifluoromethyl)-isoindolin-2-yl)aniline (0.480 g, 100% yield). Step 2: Synthesis of ethyl (2-amino-3-fluoro-4-(5-(trifluoromethyl)isoindolin-2-yl)phenyl)carbamate (Compound 14) 2-Fluoro-6-nitro-3-(5-(trifluoromethyl)isoindolin-2-yl)aniline (0.470 g, 1.378 mmole) was dissolved in methanol (5 mL) and tetrahydrofuran (5 mL). Zinc powder (0.900 g, 13.78 mmole) was added followed by ammonium chloride (737 mg, 13.78 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 45 min. and then cooled in an ice bath. Triethylamine (0.55 mL, 3.96 mmole) was added, followed by ethyl chloroformate dropwise (265 mg, 2.48 mmole) and the reaction was stirred at ambient temperature for 105 min. The reaction was filtered and evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column was washed with 13 CV of chloroform and then polarity was increased to 45% ethyl acetate/chloroform over 14 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-3-fluoro-4-(5-(trifluoromethyl)isoindolin-2-yl)phenyl)carbamate (0.146 g, 28% yield). NMR Spectroscopy:1H NMR (DMSO, 300 MHz): δ 8.50 (br s, 1H), 7.78 (s, 1H), 7.63 (q, 3H), 6.82 (d, 1H), 6.10 (t, 1H), 4.75 (s, 5H), 4.08 (q, 2H), 1.20 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd14=11.8 min. Example 15: Compound 15 5-(5-Fluoroisoindolin-2-yl)-2-nitroaniline (0.334 g, 1.22 mmole) was dissolved in methanol (5 mL) and tetrahydrofuran (5 mL). Zinc powder (0.800 g, 12.23 mmole) was added followed by ammonium chloride (654 mg, 12.23 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 2 h. The reaction was cooled in an ice bath and N, N-diisopropylethylamine (0.489 mL, 2.81 mmole) was added followed by ethyl chloroformate (235 mg, 2.20 mmole) dropwise. The reaction was stirred at ambient temperature for 60 min. and was filtered and evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 20 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-4-(5-fluoroisoindolin-2-yl)phenyl)carbamate (0.130 g, 34% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.28 (m, 1H), 7.03 (m, 3H), 6.10 (d, 3H), 4.60 (m, 4H), 4.21 (m, 2H), 3.98 (br s, 2H), 1.32 (s, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd15=6.5 min. Example 16: Compound 16 Step 1: Synthesis of 2-Nitro-5-(5-(trifluoromethyl)isoindolin-2-yl)aniline 5-Fluoro-2-nitroaniline (0.237 g, 1.52 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 5-(trifluoromethyl)isoindoline hydrochloride (0.510 g, 2.28 mmole) was added triethylamine (0.72 mL) and solid iodine (1 mg). The mixture was heated at reflux for 12 h. under argon. The reaction was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The reaction was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The organic layer was washed with 3×30 mL water and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 10 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give of 2-nitro-5-(5-(trifluoromethyl)isoindolin-2-yl)aniline (0.225 g, 46% yield). Step 2: Synthesis of ethyl (2-amino-4-(5-(trifluoromethyl)isoindolin-2-yl)phenyl)carbamate (Compound 16) 2-Nitro-5-(5-(trifluoromethyl)isoindolin-2-yl)aniline (0.252 g, 0.779 mmole) was dissolved in methanol (5 mL) and tetrahydrofuran (5 mL). Zinc powder (0.509 g, 7.79 mmole) was added followed by ammonium chloride (417 mg, 7.79 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 45 min triethylamine (0.55 mL, 3.96 mmole) was added, and the reaction was cooled in an ice bath. Ethyl chloroformate was added dropwise (150 mg, 1.40 mmole) and the reaction was stirred at ambient temperature for 2 h. The reaction was filtered and evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column was washed with 12 CV of chloroform and then polarity was increased to 50% ethyl acetate/chloroform over 20 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-4-(5-(trifluoromethyl)isoindolin-2-yl)phenyl)carbamate (0.113 g, 40% yield). NMR Spectroscopy:1H NMR (DMSO, 500 MHz): δ 8.28 (br s, 1H), 7.80 (m, 1H), 7.60-7.75 (m, 3H), 6.97 (s, 1H), 6.03 (s, 1H), 5.95 (m, 1H), 4.98 (d, 1H), 4.75 (s, 2H), 4.70 (s, 1H), 4.60 (s, 1H), 4.08 (q, 2H), 1.22 (s, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd16=8.3 min. Example 17: Compound 17 Step 1: Synthesis of 2-Fluoro-3-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-6-nitroaniline 2,3-Difluoro-6-nitroaniline (0.31 g, 1.8 mmole) was dissolved in anhydrous dimethylsulfoxide (3 mL). 6-fluoro-1,2,3,4-tetrahydroisoquinoline hydrochloride (0.50 g, 2.7 mmole) was added followed by triethylamine (0.84 mL) and solid iodine (1 mg). The mixture was heated at reflux for 3 h. under argon. The reaction was dissolved in ethyl acetate (50 mL) and extracted with water (50 mL). The aqueous layer was extracted with 2×25 mL chloroform/isopropanol (7:3). The organic layers were washed with 3×25 mL water and then brine (50 mL) and dried over sodium sulfate. The oily solid was chromatographed on a silica gel column (25 g) packed in hexanes. The column polarity was increased to 50% ethyl acetate/hexanes over 8 CV, at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 2-fluoro-3-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-6-nitroaniline (0.4 g, 74% yield). Step 2: Synthesis of ethyl (2-amino-3-fluoro-4-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (Compound 17) 2-Fluoro-3-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-6-nitroaniline (0.388 g, 1.27 mmole) was dissolved in methanol (10 mL) and tetrahydrofuran (10 mL). Zinc powder (415 mg, 6.35 mmole) was added followed by ammonium chloride (340 mg, 6.35 mmole) in DI water (1 mL). The mixture was stirred under argon at ambient temperature for 2 h. and then cooled to 10° C. in an ice bath. N, N-diisopropylethylamine (0.565 mL, 3.24 mmole) was added, followed by ethyl chloroformate dropwise (406 mg, 3.80 mmole) and the reaction was stirred at ambient temperature for 18 h. It was filtered and evaporated. The crude oil was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The aqueous layer was extracted with additional dichloromethane (3×10 mL), the organic layers were pooled and evaporated to dryness. It was chromatographed on a silica gel column (25g) packed in chloroform. The column polarity was increased to 20% ethyl acetate/chloroform over 11 CV, at 25 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-3-fluoro-4-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (0.225 g, 51% yield). NMR Spectroscopy:1H NMR (CDCl3, 300 MHz): δ 7.05 (m, 1H), 6.80-7.00 (m, 3H), 6.47 (t, 1H), 6.25 (br s, 1H), 4.25 (m, 4H), 3.52-4.15 (br s, 2H), 3.40 (m, 2H), 2.98 (m, 2H), 1.30 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd17=8.4 min. Example 18: Compound 18 Step 1: Synthesis of 2-Fluoro-6-nitro-3-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline 2,3-Difluoro-6-nitroaniline (0.238 g, 1.37 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 7-(trifluoromethyl)-1,2,3,4-tetrahydroisoquinoline hydrochloride (0.487 g, 2.05 mmole) was added triethylamine (0.643 mL) and solid iodine (1 mg). The mixture was heated at reflux for 12 h. under argon. The reaction was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The aqueous layer was washed with dichloromethane (10 mL), organics pooled and washed with brine (5 mL) and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 12 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 2-fluoro-6-nitro-3-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline (0.388 g, 80% yield). Step 2: Synthesis of Ethyl (2-amino-3-fluoro-4-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (Compound 18) 2-Fluoro-6-nitro-3-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline (0.430 g, 1.21 mmole) was dissolved in methanol (4 mL) and tetrahydrofuran (6 mL). Zinc powder (791 mg, 12.1 mmole) was added followed by ammonium chloride (647 mg, 12.1 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 45 min. and then cooled in an ice bath. N, N-diisopropylethylamine (0.421 mL, 2.42 mmole) was added, followed by ethyl chloroformate dropwise (194 mg, 1.82 mmole) and the reaction was stirred at ambient temperature for 45 min. It was filtered and evaporated. The crude oil was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The aqueous layer was extracted with additional dichloromethane (3×10 mL), the organic layers were pooled and evaporated to dryness. It was chromatographed on a silica gel column (10g) packed in chloroform. The column polarity was increased to 50% ethyl acetate/chloroform over 9 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give of ethyl (2-amino-3-fluoro-4-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (0.150 g, 31% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.42 (d, 1H), 7.39 (s, 1H), 7.28 (d, 1H), 6.98 (d, 1H), 6.45 (t, 1H), 6.25 (br s, 1H), 4.32 (s, 2H), 4.25 (q, 2H), 3.92 (br s, 2H), 3.45 (t, 2H), 3.06 (t, 2H), 1.38 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd18=7.5 min. Example 19: Compound 19 Step 1: Synthesis of 2-Fluoro-6-nitro-3-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline 2,3-Difluoro-6-nitroaniline (0.238 g, 1.37 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 6-(trifluoromethyl)-1,2,3,4-tetrahydroisoquinoline hydrochloride (0.49 g, 2.05 mmole) was added triethylamine (0.64 mL) and solid iodine (1 mg). The mixture was heated at reflux for 2 h. under argon. The reaction was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The aqueous layer was washed with dichloromethane (10 mL), organics pooled and washed with brine (5 mL) and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 12 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 2-fluoro-6-nitro-3-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline (0.420 g, 87% yield) Step 2: Synthesis of ethyl (2-amino-3-fluoro-4-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (Compound 19) 2-Fluoro-6-nitro-3-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline (0.420 g, 1.18 mmole) was dissolved in methanol (5 mL) and tetrahydrofuran (5 mL). Zinc powder (0.693 g, 10.6 mmole) was added followed by ammonium chloride (567 mg, 10.6 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 0.5 h. and then cooled in an ice bath. N, N-diisopropylethylamine (0.423 mL, 2.43 mmole) was added, followed by ethyl chloroformate dropwise (227 mg, 2.12 mmole) and the reaction was stirred at ambient temperature for 1 h. The reaction was filtered and evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 37% ethyl acetate/chloroform over 15 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-3-fluoro-4-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (0.153 g, 33% yield). NMR Spectroscopy:1H NMR (DMSO, 300 MHz): δ 8.60 (br s, 1H), 7.55 (d, 2H), 7.40 (d, 1H), 6.85 (d, 1H), 6.30 (t, 1H), 4.80 (s, 2H), 4.21 (s, 2H), 4.10 (q, 2H), 3.30 (m, 2H), 2.99 (m, 2H), 1.32 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd19=11.4 min. Example 20: Compound 20 Step 1: Synthesis of 2-Fluoro-3-(7-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-6-nitroaniline 2,3-Difluoro-6-nitroaniline (0.310 g, 1.78 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 7-fluoro-1,2,3,4-tetrahydroisoquinoline hydrochloride (0.50 g, 2.66 mmole) was added triethylamine (0.840 mL) and solid iodine (1 mg). The mixture was heated at reflux for 12 h. under argon. The reaction was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The aqueous layer was washed with dichloromethane (10 mL), organics pooled and washed with brine (5 mL) and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 12 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 2-fluoro-3-(7-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-6-nitroaniline (0.500 g, 92% yield). Step 2: Synthesis of ethyl (2-amino-3-fluoro-4-(7-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (Compound 20) 2-Fluoro-3-(7-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-6-nitroaniline (0.500 g, 1.64 mmole) was dissolved in methanol (5 mL) and tetrahydrofuran (5 mL). Zinc powder (1.07 g, 16.4 mmole) was added followed by ammonium chloride (877 mg, 16.4 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 3.5 h. and then cooled in an ice bath. N, N-diisopropylethylamine (0.666 mL, 3.77 mmole) was added, followed by ethyl chloroformate dropwise (298 mg, 2.80 mmole) and the reaction was stirred at ambient temperature for 12 h. The reaction was cooled in an ice bath. N, N-diisopropylethylamine (0.350 mL) was added, followed by ethyl chloroformate dropwise (162 mg,). The reaction was stirred at ambient temperature for 1 h. It was filtered and evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10g) packed in chloroform. The column polarity was increased to 50% ethyl acetate/chloroform over 9 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give of ethyl (2-amino-3-fluoro-4-(7-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (0.150 g, 26% yield). NMR Spectroscopy:1H NMR (CDCl3, 300 MHz): δ 7.10 (m, 1H), 6.80-7.00 (m, 3H), 6.48 (t, 1H), 6.25 (br s, 1H), 4.25 (m, 4H), 3.43 (m, 2H), 2.92 (t, 3H), 1.32 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd20=8.9 min. Example 21: Compound 21 Step 1: Synthesis of 5-(6-Fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-2-nitroaniline 5-Fluoro-2-nitroaniline (0.278 g, 1.78 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 6-fluoro-1,2,3,4-tetrahydroisoquinoline hydrochloride (0.50 g, 2.7 mmole) was added triethylamine (0.837 mL) and solid iodine (1 mg). The mixture was heated at reflux for 1.5 h. under argon. The reaction was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was washed with 3×30 mL water and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 15% ethyl acetate/chloroform over 10 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 5-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-2-nitroaniline (0.43 g, 72% yield). Step 2: Synthesis of ethyl (2-amino-4-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (Compound 21) 5-(6-Fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-2-nitroaniline (0.432 g, 1.28 mmole) was dissolved in methanol (4 mL) and dioxane (4 mL). Zinc powder (837 mg, 12.8 mmole) was added followed by ammonium chloride (685 mg, 12.8 mmole) in DI water (2.0 mL). The mixture was stirred under argon at ambient temperature for 30 min. The mixture was cooled in an ice bath. N, N-diisopropylethylamine (0.556 mL, 3.20 mmole) was added, followed by ethyl chloroformate, dropwise (274 mg, 2.56 mmole) and the reaction was stirred at ambient temperature for 18 h. To the reaction was added ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 100% ethyl acetate/chloroform over 12 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-4-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (0.065 g, 15.4% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.08 (m, 2H), 6.90 (m, 2H), 6.42 (m, 2H), 6.15 (br s, 1H), 4.36 (s, 2H), 4.22 (q, 2H), 2.50 (m, 2H), 2.98 (m, 2H), 1.32 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd21=5.4 min. Example 22: Compound 22 Step 1: Synthesis of 2-Nitro-5-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline 5-Fluoro-2-nitroaniline (0.219 g, 1.40 mmole) was dissolved in anhydrous dimethylsulfoxide (5 mL). 7-(trifluoromethyl)-1,2,3,4-tetrahydroisoquinoline hydrochloride (0.500 g, 2.10 mmole) was added triethylamine (0.660 mL) and solid iodine (1 mg) were added and the mixture was heated at reflux for an additional 3 h. under argon. The reaction was diluted with dichloromethane (10 mL) and extracted with water (10 mL). The organic layer was washed with 3×30 mL water and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate/chloroform over 12 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 2-nitro-5-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline (0.410 g, 76% yield). Step 2: Synthesis of ethyl (2-amino-4-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (Compound 22) 2-Nitro-5-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline (0.380 g, 1.13 mmole) was dissolved in methanol (4 mL) and tetrahydrofuran (6 mL). Zinc powder (737 mg, 11.3 mmole) was added followed by ammonium chloride (604 mg, 11.3 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 20 min. and then cooled to 10° C. in an ice bath. N, N-diisopropylethylamine (0.393 mL, 2.26 mmole) was added, followed by ethyl chloroformate dropwise (181 mg, 1.70 mmole) and the reaction was stirred at ambient temperature for 45 min. It was filtered and evaporated. The crude oil was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The aqueous layer was extracted with additional dichloromethane (3×10 mL), the organic layers were pooled and evaporated to dryness. It was chromatographed on a silica gel column (10g) packed in chloroform. The column polarity was increased to 50% ethyl acetate/chloroform over 9 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-4-(7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (0.112 g, 26% yield). NMR Spectroscopy:1H NMR (CDCl3, 500 MHz): δ 7.43 (m, 2H), 7.28 (d, 1H), 7.08 (d, 1H), 6.39-6.44 (m, 2H), 6.17 (br s, 1H), 4.42 (s, 2H), 4.22 (q, 2H), 3.90 (br s, 2H), 3.55 (t, 2H), 3.02 (t, 2H), 1.32 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd22=11.3 min. Example 23: Compound 23 Step 1: Synthesis of 2-Nitro-5-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline 5-Fluoro-2-nitroaniline (0.219 g, 1.40 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 6-(trifluoromethyl)-1,2,3,4-tetrahydroisoquinoline hydrochloride (0.50 g, 2.1 mmole) was added triethylamine (0.66 mL) and solid iodine (1 mg). The mixture was heated at reflux for 5 h. under argon. The reaction was dissolved in dichloromethane (10 mL) and extracted with water (10 mL). The organic layer was washed with 3×30 mL water and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in hexanes. The column polarity was increased to 100% ethyl acetate over 10 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 2-nitro-5-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline (0.23 g, 42% yield). Step 2: Synthesis of ethyl (2-amino-4-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (Compound 23) 2-Nitro-5-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)aniline (0.230 g, 0.682 mmole) was dissolved in methanol (5 mL) and tetrahydrofuran (5 mL). Zinc powder (0.396 g, 6.06 mmole) was added followed by ammonium chloride (324 mg, 6.06 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 0.5 h. and then cooled in an ice bath. N, N-diisopropylethylamine (0.242 mL, 1.39 mmole) was added, followed by ethyl chloroformate dropwise (313 mg, 1.23 mmole) and the reaction was stirred at ambient temperature for 1 h. The reaction was filtered and evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 50% ethyl acetate/chloroform over 20 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-4-(6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (0.10 g, 39% yield). NMR Spectroscopy:1H NMR (CDCl3, 300 MHz): δ 7.45 (m, 2H), 7.25 (d, 1H), 7.10 (d, 1H), 6.43 (m, 2H), 6.18 (br s, 1H), 4.42 (s, 2H), 4.22 (q, 2H), 3.58 (m, 2H), 3.05 (m, 2H), 1.30 (t, 3H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd23=7.6 min. Example 24: Compound 24 Step 1: Synthesis of 5-(7-Fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-2-nitroaniline 5-Fluoro-2-nitroaniline (0.278 g, 1.78 mmole) was dissolved in anhydrous dimethylsulfoxide (6 mL). 6-fluoro-1,2,3,4-tetrahydroisoquinoline hydrochloride (0.50 g, 2.7 mmole) was added triethylamine (0.837 mL) and solid iodine (1 mg). The mixture was heated at reflux for 1.5 h. under argon. The reaction was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). The organic layer was washed with 3×30 mL water and then dried through a 1 PS filter and evaporated to dryness. The crude material was chromatographed on a silica gel column (10 g) packed in chloroform. The column polarity was increased to 15% ethyl acetate/chloroform over 10 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give 5-(7-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-2-nitroaniline (0.43 g, 84% yield). Step 2: Synthesis of ethyl (2-amino-4-(7-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (Compound 24) 5-(7-Fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-2-nitroaniline (0.430 g, 1.49 mmole) was dissolved in methanol (15 mL) and tetrahydrofuran (15 mL). Zinc powder (0.979 g, 15.0 mmole) was added followed by ammonium chloride (800 mg, 15.0 mmole) in DI water (2 mL). The mixture was stirred under argon at ambient temperature for 4 h. and then cooled in an ice bath. N, N-diisopropylethylamine (0.593 mL, 3.43 mmole) was added, followed by ethyl chloroformate dropwise (271 mg, 2.53 mmole) and the reaction was stirred at ambient temperature for 12 h. The reaction was filtered and evaporated. The crude oil was dissolved in ethyl acetate (10 mL) and extracted with water (10 mL). the organic layer was then dried through a 1 PS filter and evaporated to dryness. It was chromatographed on a silica gel column (10g) packed in chloroform. The column polarity was increased to 20% ethyl acetate/chloroform over 8 CV, at 12 mL/min. Fractions (22 mL each) containing the product were pooled and stripped to give ethyl (2-amino-4-(7-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)phenyl)carbamate (0.028 g, 6% yield). NMR Spectroscopy:1H NMR (CDCl3, 300 MHz): δ 7.10 (m, 2H), 6.80-6.98 (m, 2H), 6.48 (m, 2H), 6.15 (br s, 1H), 4.38 (s, 2H), 4.22 (q, 2H), 2.55 (m, 2H), 2.98 (t, 2H), 1.30 (m, 4H). HPLC: Grace Alltima C18 reverse phase HPLC column (250×4.6 mm); Mobile Phase A: 0.1% TFA in water; Mobile Phase B: 0.1% TFA in 100% acetonitrile; Flow rate: 1 mL/min; Temperature: 30° C.; Injection Volume: 0.1-5 μL; Detection Wavelengths: 215-260 nm; Gradient: 0 min (60% A, 40% B), 15 min (10% A, 90% B), 18 min (10% A, 90% B), 18.1 min (60% A, 40% B), 20 min (60% A, 40% B). tCmpd24=5.7 min. Example 25: Compound 27 Under nitrogen, to N-(4-bromo-3-fluoro-2,6-dimethyl-phenyl)-3,3-dimethyl-butanamide (316 mg, 1.00 mmol, 1.00 equiv) in toluene (5 mL) at 23° C. were added 6-fluoro-1,2,3,4-tetrahydroisoquinoline (166 mg, 1.10 mmol, 1.10 equiv), DavePhos (47 mg, 0.12 mmol, 12 mol %), Pd2(dba)3(37 mg, 0.040 mmol, 4.0 mol %), and t-BuOK (168 mg, 1.50 mmol, 1.50 equiv). After stirring for 24 hr at 90° C., the reaction mixture was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 70 mg the title compound (18% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.02 (dd, J=9.0, 8.7 Hz, 1H), 6.89-6.77 (m, 3H), 6.61 (br s, 1H), 4.27 (br s, 2H), 3.43 (br s, 2H), 3.03 (br s, 2H), 2.24 (s, 2H), 2.17-2.10 (m, 6H), 1.08 (s, 9H). Example 26: Compound 28 Under nitrogen, to N-(4-bromo-3-fluoro-2,6-dimethyl-phenyl)-3,3-dimethyl-butanamide (316 mg, 1.00 mmol, 1.00 equiv) in toluene (5 mL) at 23° C. were added N-(4-fluorobenzyl)prop-2-yn-1-amine (180 mg, 1.10 mmol, 1.10 equiv), DavePhos (47 mg, 0.12 mmol, 12 mol %), Pd2(dba)3(37 mg, 0.040 mmol, 4.0 mol %), and t-BuOK (168 mg, 1.50 mmol, 1.50 equiv). After stirring for 2 d at 110° C., the reaction mixture was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 25 mg the title compound (6% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.35 (dd, J=8.7, 5.7 Hz, 2H), 6.96 (dd, J=9.0, 8.7 Hz, 2H), 7.08 (d, J=7.5 Hz, 1H), 6.96 (br s, 1H), 3.88 (s, 2H), 3.58 (s, 2H), 2.23 (s, 1H), 2.12-2.08 (m, 8H), 1.10 (s, 9H). Example 27: Compound 29 Step 1: Synthesis of 2-Fluoro-N1-[(4-fluorophenyl)methyl]-4-nitro-benzene-1,3-diamine Under air, to 2,3-difluoro-6-nitroaniline (335 g, 1.92 mol, 1.00 equiv) in DMSO (400 mL) at 23° C. was added 4-fluorobenzylamine (395 mL, 3.46 mol, 1.80 equiv), Et3N (642 mL, 4.61 mol, 2.40 equiv), and 12 (243 mg, 0.960 mmol, 0.0500 mol %). After stirring at 100° C. with a reflux condenser for 3 hr, the reaction mixture was cooled to 23° C. and was poured into water (2 L). The resulting suspension was filtered and the solids were washed with water (3×800 mL). The solids were collected and dried inside an oven set to 110° C. for 8 hr to afford 530 g of the title compound as yellow solids (99% yield). NMR Spectroscopy:1H NMR (400 MHz, CDCl3, 23° C., δ): 7.87 (dd, J=9.6, 1.6 Hz, 1H), 7.31-7.28 (m, 2H), 7.08-7.03 (m, 2H), 6.11-6.03 (m, 3H), 4.82 (br s, 1H), 4.44 (d, J=5.2 Hz, 2H). Step 2: Synthesis of tert-Butyl N-[3-[bis(tert-butoxycarbonyl)amino]-2-fluoro-4-nitro-phenyl]-N-[(4-fluorophenyl)-methyl]carbamate Under nitrogen, to 2-fluoro-N1-[(4-fluorophenyl)methyl]-4-nitro-benzene-1,3-diamine (5.58 g, 20.0 mmol, 1.00 equiv) in THF (200 mL) at 23° C. were added DMAP (244 mg, 2.00 mmol, 10.0 mol %), NaH (1.44 g, 60.0 mmol, 3.00 equiv), and Boc2O (13.8 mL, 60.0 mmol, 3.00 equiv). After stirring for 1.5 hr at 60° C., the reaction mixture was cooled to 23° C. and water (200 mL) was added dropwise. The phases were separated and the aqueous phase was extracted with EtOAc (2×200 mL). The combined organic phases were washed with brine (200 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 8.35 g of the title compound (72% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.81 (dd, J=9.0, 1.8 Hz, 1H), 7.24-7.15 (m, 3H), 6.95 (dd, J=8.4, 8.4 Hz, 2H), 4.80 (s, 2H), 1.41 (s, 9H), 1.36 (s, 18H). Step 3: Synthesis of tert-Butyl N-[4-amino-3-[bis(tert-butoxycarbonyl)amino]-2-fluoro-phenyl]-N-[(4-fluorophenyl)methyl]carbamate Under air, to tert-butyl N-[3-[bis(tert-butoxycarbonyl)amino]-2-fluoro-4-nitro-phenyl]-N-[(4-fluorophenyl)methyl]carbamate (8.35 g, 14.4 mmol, 1.00 equiv) in MeOH (144 mL) at 23° C. were added Zn powder (4.71 g, 72.1 mmol, 5.00 equiv) and NH4Cl (3.85 g, 72.1 mmol, 5.00 equiv) in H2O (20 mL). After stirring for 3 hr at 23° C., the reaction mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo, and H2O (200 mL) and EtOAc (200 mL) were added to the residue. The phases were separated and the aqueous phase was extracted with EtOAc (2×200 mL). The combined organic phases were washed with brine (200 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 6.00 g of the title compound (76% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.17 (dd, J=8.4, 5.4 Hz, 2H), 6.92 (dd, J=8.4, 8.4 Hz, 2H), 6.69 (dd, J=5.1, 5.1 Hz, 1H), 6.38 (d, J=5.1 Hz, 1H), 4.66 (br s, 2H), 1.38 (s, 27H). Step 4: Synthesis of tert-Butyl N-[3-[bis(tert-butoxycarbonyl)amino]-4-(3,3-dimethylbutanoylamino)-2-fluoro-phenyl]-N-[(4-fluorophenyl)methyl]carbamate Under nitrogen, to tert-butyl N-[4-amino-3-[bis(tert-butoxycarbonyl)amino]-2-fluoro-phenyl]-N-[(4-fluorophenyl)methyl]carbamate (5.90 g, 10.7 mmol, 1.00 equiv) in DCM (26 mL) at 0° C. were added DIPEA (2.06 mL, 11.8 mmol, 1.10 equiv) and tert-butylacetyl chloride (1.65 mL, 11.8 mmol, 1.10 equiv). After stirring for 3 hr at 23° C., the reaction mixture was concentrated in vacuo, and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 4.30 g of the title compound (62% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.98 (d, J=9.0 Hz, 1H), 7.21-6.90 (m, 6H), 4.71 (s, 2H), 2.18 (s, 2H), 1.39 (s, 9H), 1.34 (s, 18H), 1.06 (s, 9H). Step 5: Synthesis of N-[2-amino-3-fluoro-4-[(4-fluorophenyl)methylamino]phenyl]-3,3-dimethyl-butanamide (Compound 29) Under nitrogen, to tert-butyl N-[3-[bis(tert-butoxycarbonyl)amino]-4-(3,3-dimethylbutanoylamino)-2-fluoro-phenyl]-N-[(4-fluorophenyl)methyl]carbamate (1.30 g, 2.01 mmol, 1.00 equiv) in DCM (10 mL) at 23° C. was added HCl (2.0 M in Et2O, 10.1 mL, 20.1 mmol, 10.0 equiv). After stirring for 15 hr at 23° C., NaHCO3(aq) (100 mL) was added to the reaction mixture. The phases were separated and the aqueous phase was extracted with EtOAc (2×100 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 690 mg of the title compound (99% yield). NMR Spectroscopy:1H NMR (300 MHz, DMSO-d6, 23° C., δ): 9.00 (s, 1H), 7.36 (dd, J=8.4, 5.4 Hz, 2H), 7.11 (dd, J=8.4, 8.4 Hz, 2H), 6.56 (d, J=8.7 Hz, 1H), 5.92 (dd, J=6.0, 6.0 Hz, 1H), 5.83 (dd, J=8.7, 8.7 Hz, 1H), 4.54 (s, 2H), 4.27 (d, J=6.0 Hz, 2H), 2.09 (s, 2H), 1.00 (s, 9H). Example 28: Compound 30 Step 1: Synthesis of 1-Bis(tert-butoxylcarbonyl)amino-2,3-difluoro-6-nitrobenzene Under nitrogen, to 2,3-difluoro-6-nitroaniline (3.48 g, 20.0 mmol, 1.00 equiv) in THF (100 mL) at 23° C. were added DMAP (122 mg, 1.00 mmol, 5.00 mol %), NaH (1.44 g, 60.0 mmol, 3.00 equiv), and Boc2O (13.8 mL, 60 mmol, 3.00 equiv). After stirring for 1 h at 60° C., the reaction mixture was cooled to 23° C. and water (100 mL) was added. The solution was then neutralized with 1N HCl (aq) and was extracted with EtOAc (2×100 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by chromatography on silica gel eluting with hexanes/EtOAc to afford 6.20 g of the title compound (83% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.98-7.90 (m, 1H), 7.37-7.27 (m, 1H), 1.40 (s, 18H). Step 2: Synthesis of tert-Butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[(4-fluorophenyl)methylamino]-6-nitro-phenyl]carbamate Under nitrogen, to 1-bis(tert-butoxylcarbonyl)amino-2,3-difluoro-6-nitrobenzene (1.87 g, 5.00 mmol, 1.00 equiv) in DMSO (5 mL) at 23° C. were added 4-fluorobenzylamine (1.03 mL, 9.00 mmol, 1.80 equiv) and Et3N (0.837 mL, 6.00 mmol, 1.20 equiv). After stirring for 1.5 hr at 23° C., the reaction mixture was cooled to 23° C. and water (10 mL) was added. The solution was then neutralized with 1N HCl (aq) and was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by chromatography on silica gel eluting with hexanes/EtOAc to afford 2.00 g of the title compound (83% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.97 (d, J=9.0 Hz, 1H), 7.31 (dd, J=8.7, 5.7 Hz, 2H), 7.07 (dd, J=8.7, 8.7 Hz, 2H), 6.59 (dd, J=8.4, 8.4 Hz, 1H), 4.98 (br s, 1H), 4.44 (br s, 2H), 1.42 (s, 18H). Step 3: Synthesis of tert-Butyl N-tert-butoxycarbonyl-N-[6-(3,3-dimethylbutanoylamino)-2-fluoro-3-[(4-fluorophenyl)methyl-prop-2-ynyl-amino]phenyl]carbamate Under nitrogen, to tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[(4-fluorophenyl)-methylamino]-6-nitro-phenyl]carbamate (2.00 g, 4.17 mmol, 1.00 equiv) in THF (4.2 mL) at 23° C. were added propargyl bromide (0.948 mL, 12.5 mmol, 3.00 equiv) and NaH (300 mg, 12.5 mmol, 3.00 equiv). After stirring for 1 hr at 23° C., the reaction mixture was cooled to 0° C. and water (10 mL) was added. The reaction mixture was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford a crude alkylation product, which was used in the next step without further purification. Under air, to the crude product obtained above in MeOH (42 mL) at 23° C. were added Zn powder (1.36 g, 20.9 mmol, 5.00 equiv) and NH4Cl (1.12 g, 20.9 mmol, 5.00 equiv) in H2O (5 mL). After stirring for 3 hr at 23° C., the reaction mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo, and H2O (50 mL) and EtOAc (50 mL) were added to the residue. The phases were separated and the aqueous phase was extracted with EtOAc (2×50 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford a crude reduction product, which was used in the next step without further purification. Under nitrogen, to the crude reduction product obtained above in MeCN (4.2 mL) at 23° C. were added DIPEA (1.31 mL, 7.51 mmol, 1.80 equiv) and tert-butylacetyl chloride (1.05 mL, 7.51 mmol, 1.80 equiv). After stirring for 1 hr at 23° C., the reaction mixture was concentrated in vacuo, and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 1.90 g of the title compound (78% yield over 3 steps). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.90 (d, J=8.7 Hz, 1H), 7.37 (dd, J=8.7, 5.7 Hz, 2H), 7.16 (dd, J=9.0, 9.0 Hz, 1H), 7.06 (s, 1H), 6.99 (dd, J=8.7, 8.4 Hz, 2H), 4.27 (s, 2H), 3.76 (d, J=2.4 Hz, 2H), 2.25 (t, J=2.4 Hz, 1H), 2.19 (s, 2H), 1.40 (s, 18H), 1.07 (s, 9H). Step 4: Synthesis of N-(2-amino-3-fluoro-4-((4-fluorobenzyl)(prop-2-yn-1-yl)amino)phenyl)-3,3-dimethylbutanamide (Compound 30) Under nitrogen, to tert-butyl N-tert-butoxycarbonyl-N-[6-(3,3-dimethylbutanoylamino)-2-fluoro-3-[(4-fluorophenyl)methyl-prop-2-ynyl-amino]phenyl]carbamate (1.90 g, 3.24 mmol, 1.00 equiv) in DCM (8 mL) at 23° C. was added HCl (2.0 M in Et2O, 16.2 mL, 20.1 mmol, 10.0 equiv). After stirring for 15 hr at 23° C., NaHCO3(aq) (10 mL) was added to the reaction mixture. The phases were separated and the aqueous phase was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford 1.20 g of the title compound (96% yield). NMR Spectroscopy:1H NMR (300 MHz, methanol-d4, 23° C., δ): 7.34 (dd, J=8.7, 5.7 Hz, 2H), 6.96 (dd, J=9.0, 9.0 Hz, 2H), 6.71 (d, J=8.7 Hz, 1H), 6.50 (dd, J=9.0, 8.7 Hz, 1H), 4.22 (s, 2H), 3.69 (d, J=2.4 Hz, 2H), 2.56 (t, J=2.4 Hz, 1H), 2.19 (s, 2H), 1.07 (s, 9H). Example 29: Compound 31 Step 1: Synthesis of tert-Butyl N-tert-butoxycarbonyl-N-[2-fluoro-6-nitro-3-[[4-(trifluoromethyl)phenyl]-methylamino]phenyl]carbamate Under nitrogen, to 1-bis(tert-butoxylcarbonyl)amino-2,3-difluoro-6-nitrobenzene (1.87 g, 5.00 mmol, 1.00 equiv) in DMSO (5 mL) at 23° C. were added 4-(trifluoromethyl)benzylamine (1.28 mL, 9.00 mmol, 1.80 equiv) and Et3N (0.837 mL, 6.00 mmol, 1.20 equiv). After stirring for 1.5 hr at 23° C., the reaction mixture was cooled to 23° C. and water (10 mL) was added. The solution was then neutralized with 1N HCl (aq) and was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by chromatography on silica gel eluting with hexanes/EtOAc to afford 1.90 g of the title compound (72% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.95 (d, J=9.0 Hz, 1H), 7.63 (d, J=7.8 Hz, 2H), 7.48 (d, J=7.8 Hz, 2H), 6.54 (dd, J=8.4, 8.4 Hz, 1H), 5.08 (br s, 1H), 4.56 (br s, 2H), 1.41 (s, 18H). Step 2: Synthesis of tert-Butyl N-tert-butoxycarbonyl-N-[6-(3,3-dimethylbutanoylamino)-2-fluoro-3-[prop-2-ynyl-[[4-(trifluoromethyl)phenyl]methyl]amino]phenyl]carbamate Under nitrogen, to tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-6-nitro-3-[[4-(trifluoromethyl)phenyl]methylamino]phenyl]carbamate (1.90 g, 3.59 mmol, 1.00 equiv) in THF (3.6 mL) at 23° C. were added propargyl bromide (0.816 mL, 10.8 mmol, 3.00 equiv) and NaH (258 mg, 10.8 mmol, 3.00 equiv). After stirring for 1 hr at 23° C., the reaction mixture was cooled to 0° C. and water (10 mL) was added. The reaction mixture was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford a crude alkylation product, which was used in the next step without further purification. Under air, to the crude product obtained above in MeOH (36 mL) at 23° C. were added Zn powder (1.17 g, 18.0 mmol, 5.00 equiv) and NH4Cl (960 mg, 18.0 mmol, 5.00 equiv) in H2O (5 mL). After stirring for 3 hr at 23° C., the reaction mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo, and H2O (50 mL) and EtOAc (50 mL) were added to the residue. The phases were separated and the aqueous phase was extracted with EtOAc (2×50 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford a crude reduction product, which was used in the next step without further purification. Under nitrogen, to the crude reduction product obtained above in MeCN (3.6 mL) at 23° C. were added DIPEA (1.13 mL, 6.46 mmol, 1.80 equiv) and tert-butylacetyl chloride (0.902 mL, 6.46 mmol, 1.80 equiv). After stirring for 1 hr at 23° C., the reaction mixture was concentrated in vacuo, and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 1.90 g of the title compound (83% yield over 3 steps). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.91 (d, J=8.7 Hz, 1H), 7.62-7.50 (m, 4H), 7.15 (dd, J=9.0, 9.0 Hz, 1H), 7.07 (s, 1H), 4.36 (s, 2H), 3.73 (d, J=2.4 Hz, 2H), 2.25 (t, J=2.4 Hz, 1H), 2.19 (s, 2H), 1.40 (s, 18H), 1.07 (s, 9H). Step 3: Synthesis of N-[2-amino-3-fluoro-4-[prop-2-ynyl-[[4-(trifluoromethyl)phenyl]methyl]amino]phenyl]-3,3-dimethyl-butanamide (Compound 31) Under nitrogen, to tert-butyl N-tert-butoxycarbonyl-N-[6-(3,3-dimethylbutanoylamino)-2-fluoro-3-[prop-2-ynyl-[[4-(trifluoromethyl)phenyl]methyl]amino]phenyl]carbamate (1.90 g, 2.99 mmol, 1.00 equiv) in DCM (7.5 mL) at 23° C. was added HCl (2.0 M in Et2O, 15.0 mL, 29.9 mmol, 10.0 equiv). After stirring for 15 hr at 23° C., NaHCO3(aq) (10 mL) was added to the reaction mixture. The phases were separated and the aqueous phase was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by chromatography on silica gel eluting with hexanes/EtOAc to afford 600 mg of the title compound (46% yield). NMR Spectroscopy:1H NMR (300 MHz, methanol-d4, 23° C., δ): 7.58-7.50 (m, 4H), 6.70 (d, J=9.0 Hz, 1H), 6.50 (dd, J=9.0, 8.7 Hz, 1H), 4.35 (s, 2H), 3.76 (d, J=2.4 Hz, 2H), 2.59 (t, J=2.4 Hz, 1H), 2.20 (s, 2H), 1.03 (s, 9H). Example 30: Compound 32 Step 1: Synthesis of 2-Fluoro-3-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-6-nitroaniline Under nitrogen, to 2,3-difluoro-6-nitro-aniline (1.74 g, 10.0 mmol, 1.00 equiv) in DMSO (10 mL) at 23° C. were added N-[(4-fluoro-2-methyl-phenyl)methyl]ethanamine (1.67 g, 10.0 mmol, 1.00 equiv) and Et3N (1.67 mL, 12.0 mmol, 1.20 equiv). After stirring for 1 hr at 100° C., the reaction mixture was cooled to 23° C. and water (100 mL) was added. The solution was then neutralized with 1N HCl (aq) and was extracted with EtOAc (2×100 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by chromatography on silica gel eluting with hexanes/EtOAc to afford 1.38 g of the title compound (45% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.87 (d, J=9.0 Hz, 1H), 7.08 (dd, J=7.8 Hz, 7.8 Hz, 1H), 6.95-6.85 (m, 2H), 6.33 (dd, J=8.4, 8.4 Hz, 1H), 4.49 (s, 2H), 3.66 (t, J=6.0 Hz, 2H), 2.99 (t, J=6.0 Hz, 2H). Step 2: Synthesis of tert-Butyl N-tert-butoxycarbonyl-N-[3-[ethyl-[(4-fluoro-2-methyl-phenyl)methyl]amino]-2-fluoro-6-nitro-phenyl]carbamate Under nitrogen, to 2-fluoro-3-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-6-nitroaniline (1.38 g, 4.29 mmol, 1.00 equiv) in THF (21 mL) at 23° C. were added DMAP (26.2 mg, 0.214 mmol, 5.00 mol %), NaH (309 mg, 12.9 mmol, 3.00 equiv), and Boc2O (2.96 mL, 12.9 mmol, 3.00 equiv). After stirring for 1 h at 60° C., the reaction mixture was cooled to 23° C. and water (50 mL) was added. The solution was then neutralized with 1N HCl (aq) and was extracted with EtOAc (2×50 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by chromatography on silica gel eluting with hexanes/EtOAc to afford 2.17 g of the title compound (97% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.96 (d, J=9.0 Hz, 1H), 7.10 (dd, J=7.8 Hz, 7.8 Hz, 1H), 6.95-6.83 (m, 3H), 4.46 (s, 2H), 3.64 (t, J=6.0 Hz, 2H), 2.99 (t, J=6.0 Hz, 2H), 1.41 (s, 18H). Step 3: Synthesis of tert-Butyl N-tert-butoxycarbonyl-N-[6-(3,3-dimethylbutanoylamino)-2-fluoro-3-(6-fluoro-3,4-dihydro-1H-isoquinolin-2-yl)phenyl]carbamate Under air, to tert-butyl N-tert-butoxycarbonyl-N-[3-[ethyl-[(4-fluoro-2-methyl-phenyl)methyl]amino]-2-fluoro-6-nitro-phenyl]carbamate (2.17 g, 4.29 mmol, 1.00 equiv) in MeOH (43 mL) at 23° C. were added Zn powder (1.40 g, 21.5 mmol, 5.00 equiv) and NH4Cl (1.15 g, 21.5 mmol, 5.00 equiv) in H2O (5 mL). After stirring for 3 hr at 23° C., the reaction mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo, and H2O (50 mL) and EtOAc (50 mL) were added to the residue. The phases were separated and the aqueous phase was extracted with EtOAc (2×50 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford a crude reduction product, which was used in the next step without further purification. Under nitrogen, to the crude reduction product obtained above in MeCN (4.3 mL) at 23° C. were added DIPEA (1.35 mL, 7.72 mmol, 1.80 equiv) and tert-butylacetyl chloride (1.08 mL, 7.72 mmol, 1.80 equiv). After stirring for 1 hr at 23° C., the reaction mixture was concentrated in vacuo, and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 2.20 g of the title compound (89% yield over 2 steps). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.87 (d, J=8.7 Hz, 1H), 7.10-6.80 (m, 4H), 4.23 (s, 2H), 3.38 (t, J=6.0 Hz, 2H), 2.96 (t, J=6.0 Hz, 2H), 2.16 (s, 2H), 1.41 (s, 18H), 1.07 (s, 9H). Step 4: Synthesis of ethyl (3-fluoro-4-(6-fluoro-3,4-dihydroisoquinolin-2(1H)-yl)-2,6-dimethylphenyl)-carbamate (Compound 32) Under nitrogen, to tert-butyl N-tert-butoxycarbonyl-N-[6-(3,3-dimethylbutanoylamino)-2-fluoro-3-(6-fluoro-3,4-dihydro-1H-isoquinolin-2-yl)phenyl]carbamate (2.20 g, 3.83 mmol, 1.00 equiv) in DCM (10 mL) at 23° C. was added HCl (2.0 M in Et2O, 19.2 mL, 38.3 mmol, 10.0 equiv). After stirring for 15 hr at 23° C., NaHCO3(aq) (100 mL) was added to the reaction mixture. The phases were separated and the aqueous phase was extracted with EtOAc (2×100 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford 1.20 g of the title compound (84% yield). NMR Spectroscopy:1H NMR (300 MHz, methanol-d4, 23° C., δ): 7.16-7.10 (m, 1H), 6.94-6.79 (m, 3H), 6.46 (dd, J=8.7, 8.7 Hz, 1H), 4.19 (s, 2H), 3.36 (t, J=6.0 Hz, 2H), 2.96 (t, J=6.0 Hz, 2H), 2.27 (s, 2H), 1.11 (s, 9H). Example 31: Compound 33 Step 1: Synthesis of tert-Butyl N-tert-butoxycarbonyl-N-[2-fluoro-6-nitro-3-[6-(trifluoromethyl)-3,4-dihydro-1H-isoquinolin-2-yl]phenyl]carbamate Under nitrogen, to 1-bis(tert-butoxylcarbonyl)amino-2,3-difluoro-6-nitrobenzene (1.87 g, 5.00 mmol, 1.00 equiv) in DMSO (5 mL) at 23° C. were added 6-(trifluoromethyl)-1,2,3,4-tetrahydroisoquinoline (1.01 g, 5.00 mmol, 1.00 equiv) and Et3N (1.74 mL, 12.5 mmol, 2.50 equiv). After stirring for 1.5 hr at 23° C., the reaction mixture was cooled to 23° C. and water (10 mL) was added. The solution was then neutralized with 1N HCl (aq) and was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by chromatography on silica gel eluting with hexanes/EtOAc to afford 1.39 g of the title compound (50% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.97 (d, J=9.0 Hz, 1H), 7.47 (d, J=8.1 Hz, 1H), 7.41 (s, 1H), 7.30 (d, J=8.1 Hz, 1H), 6.94 (dd, J=8.7, 8.4 Hz, 1H), 4.54 (s, 2H), 3.67 (t, J=6.0 Hz, 2H), 3.07 (t, J=6.0 Hz, 2H), 1.42 (s, 18H). Step 2: Synthesis of tert-Butyl N-tert-butoxycarbonyl-N-[6-(3,3-dimethylbutanoylamino)-2-fluoro-3-[6-(trifluoromethyl)-3,4-dihydro-1H-isoquinolin-2-yl]phenyl]carbamate Under air, to tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-6-nitro-3-[6-(trifluoromethyl)-3,4-dihydro-1H-isoquinolin-2-yl]phenyl]carbamate (1.39 g, 2.50 mmol, 1.00 equiv) in MeOH (25 mL) at 23° C. were added Zn powder (817 mg, 12.5 mmol, 5.00 equiv) and NH4Cl (669 mg, 12.5 mmol, 5.00 equiv) in H2O (5 mL). After stirring for 3 hr at 23° C., the reaction mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo, and H2O (50 mL) and EtOAc (50 mL) were added to the residue. The phases were separated and the aqueous phase was extracted with EtOAc (2×50 mL). The combined organic phases were washed with brine (100 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford a crude reduction product, which was used in the next step without further purification. Under nitrogen, to the crude reduction product obtained above in MeCN (2.5 mL) at 23° C. were added DIPEA (0.784 mL, 4.50 mmol, 1.80 equiv) and tert-butylacetyl chloride (0.628 mL, 4.50 mmol, 1.80 equiv). After stirring for 1 hr at 23° C., the reaction mixture was concentrated in vacuo, and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 1.25 g of the title compound (80% yield over 2 steps). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.95 (d, J=8.7 Hz, 1H), 7.44 (d, J=8.1 Hz, 1H), 7.37 (s, 1H), 7.30-7.18 (m, 2H), 7.09 (s, 1H), 4.37 (s, 2H), 3.48 (t, J=6.0 Hz, 2H), 3.10 (t, J=6.0 Hz, 2H), 2.20 (s, 2H), 1.42 (s, 18H), 1.08 (s, 9H). Step 3: Synthesis of N-[2-amino-3-fluoro-4-[6-(trifluoromethyl)-3,4-dihydro-1H-isoquinolin-2-yl]phenyl]-3,3-dimethyl-butanamide (Compound 33) Under nitrogen, to tert-butyl N-tert-butoxycarbonyl-N-[6-(3,3-dimethylbutanoylamino)-2-fluoro-3-[6-(trifluoromethyl)-3,4-dihydro-1H-isoquinolin-2-yl]phenyl]carbamate (1.25 g, 2.00 mmol, 1.00 equiv) in DCM (5 mL) at 23° C. was added HCl (2.0 M in Et2O, 10.0 mL, 20.0 mmol, 10.0 equiv). After stirring for 15 hr at 23° C., NaHCO3(aq) (10 mL) was added to the reaction mixture. The phases were separated and the aqueous phase was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo to afford 570 mg of the title compound (67% yield). NMR Spectroscopy:1H NMR (300 MHz, methanol-d4, 23° C., δ): 7.42-7.25 (m, 3H), 6.75 (d, J=8.7 Hz, 1H), 6.41 (dd, J=8.7, 8.7 Hz, 1H), 4.23 (s, 2H), 3.36 (t, J=6.0 Hz, 2H), 2.96 (t, J=6.0 Hz, 2H), 2.21 (s, 2H), 1.04 (s, 9H). Example 32: Compound 34 Step 1: Synthesis of 1-Fluoro-2,4-dimethyl-3-nitrobenzene Under air, to 1-bromo-4-fluoro-3-methyl-2-nitro-benzene (4.68 g, 20.0 mmol, 1.00 equiv) in DME-H2O (10 mL-10 mL) at 23° C. were added trimethylboroxine (1.76 g, 14.0 mmol, 0.700 equiv), K2CO3(4.15 g, 30.0 mmol, 1.50 equiv), and Pd(PPh3)4(2.31 g, 2.00 mmol, 10.0 mol %). After stirring for 3 d at 100° C., the reaction mixture was cooled to 23° C. The phases were separated and the aqueous phase was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 3.00 g of the title compound (89% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.05-6.99 (m, 2H), 2.21 (s, 3H), 2.15 (d, J=1.8 Hz, 3H). Step 2: Synthesis of 3-Fluoro-2,6-dimethylaniline Under air, to 1-fluoro-2,4-dimethyl-3-nitrobenzene (3.00 g, 17.7 mmol, 1.00 equiv) in MeOH (177 mL) at 23° C. were added Zn powder (5.80 g, 88.7 mmol, 5.00 equiv) and NH4Cl (4.74 g, 5.00 mmol, 5.00 equiv) in H2O (10 mL). After stirring for 3 hr at 23° C., the reaction mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo, and H2O (100 mL) and EtOAc (100 mL) were added to the residue. The phases were separated and the aqueous phase was extracted with EtOAc (2×100 mL). The combined organic phases were washed with brine (200 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 2.00 g of the title compound (81% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 6.87 (dd, J=7.5, 7.5 Hz, 1H), 6.48 (dd, J=9.0, 7.5 Hz, 1H), 2.19 (s, 3H), 2.14 (d, J=1.8 Hz, 3H). Step 3: Synthesis of 4-Bromo-3-fluoro-2,6-dimethylaniline Under air, to 3-fluoro-2,6-dimethylaniline (2.00g, 14.4 mmol, 1.00 equiv) in AcOH (14 mL) at 23° C. was added NBS (2.56 g, 14.4 mmol, 1.00 equiv). After stirring for 10 min at 23° C., the reaction mixture was poured into water (100 mL). Potassium carbonate was added to neutralize the solution, after which was extracted with EtOAc (2×100 mL). The combined organic phases were washed with brine (200 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 1.66 g of the title compound (53% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.07 (d, J=4.5 Hz, 1H), 2.14 (s, 6H). Step 4: Synthesis of ethyl (4-bromo-3-fluoro-2,6-dimethylphenyl)carbamate Under nitrogen, to 4-bromo-3-fluoro-2,6-dimethylaniline (830 mg, 3.81 mmol, 1.00 equiv) in MeCN (3.8 mL) at 0° C. were added DIPEA (797 μl, 5.72 mmol, 1.50 equiv) and ethyl chloroformate (798 μl, 5.72 mmol, 1.50 equiv). After stirring for 4 hr at 23° C., NaHCO3(aq) (10 mL) was added to the reaction mixture. The phases were separated and the aqueous phase was extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine (10 mL) and dried (MgSO4). The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 935 mg of the title compound (78% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.26 (d, J=4.5 Hz, 1H), 6.65 (br s, 1H), 2.18 (s, 2H), 2.14 (s, 6H), 1.14 (s, 9H). Step 5: Synthesis of N-(3-fluoro-4-((4-fluorobenzyl)amino)-2,6-dimethylphenyl)-3,3-dimethylbutanamide (Compound 34) Under nitrogen, to N-(4-bromo-3-fluoro-2,6-dimethyl-phenyl)-3,3-dimethyl-butanamide (316 mg, 1.00 mmol, 1.00 equiv) in toluene (5 mL) at 23° C. are added 4-fluorobenzylamine (125 mg, 1.00 mmol, 1.00 equiv), DavePhos (47 mg, 0.12 mmol, 12 mol %), Pd2(dba)3(37 mg, 0.040 mmol, 4.0 mol %), and t-BuOK (168 mg, 1.50 mmol, 1.50 equiv). After stirring for 2 hr at 90° C., the reaction mixture is concentrated in vacuo and the residue is purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford the title compound. Example 33: Compound 35 Under nitrogen, to N-(4-bromo-3-fluoro-2,6-dimethyl-phenyl)-3,3-dimethyl-butanamide (316 mg, 1.00 mmol, 1.00 equiv) in toluene (5 mL) at 23° C. were added N-[[4-(trifluoromethyl)phenyl]methyl]prop-2-yn-1-amine (235 mg, 1.10 mmol, 1.10 equiv), DavePhos (47 mg, 0.12 mmol, 12 mol %), Pd2(dba)3(37 mg, 0.040 mmol, 4.0 mol %), and t-BuOK (168 mg, 1.50 mmol, 1.50 equiv). After stirring for 2 d at 110° C., the reaction mixture was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 120 mg the title compound (27% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.57 (d, J=8.7 Hz, 2H), 7.50 (d, J=8.7 Hz, 2H), 7.08 (d, J=7.5 Hz, 1H), 6.96 (br s, 1H), 4.00 (s, 2H), 3.65 (s, 2H), 2.27 (s, 1H), 2.13-2.10 (m, 8H), 1.12 (s, 9H). Example 34: Compound 36 Under nitrogen, to N-(4-bromo-3-fluoro-2,6-dimethyl-phenyl)-3,3-dimethyl-butanamide (158 mg, 0.500 mmol, 1.00 equiv) in toluene (2.5 mL) at 23° C. were added 6-(trifluoromethyl)-1,2,3,4-tetrahydroisoquinoline hydrochloric acid salt (178 mg, 0.750 mmol, 1.50 equiv), DavePhos (47 mg, 0.12 mmol, 24 mol %), Pd2(dba)3(37 mg, 0.040 mmol, 8.0 mol %), and t-BuOK (168 mg, 1.50 mmol, 3.00 equiv). After stirring for 1 hr at 100° C., the reaction mixture was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc to afford 72 mg the title compound (33% yield). NMR Spectroscopy:1H NMR (300 MHz, CDCl3, 23° C., δ): 7.40-7.32 (m, 2H), 7.14 (d, J=8.7 Hz, 1H), 6.79-6.70 (m, 1H), 6.54 (br s, 1H), 4.25 (br s, 2H), 3.37 (t, J=6.0 Hz, 2H), 3.03 (t, J=6.0 Hz, 2H), 2.23 (s, 2H), 2.13-2.08 (m, 6H), 1.08 (s, 9H). Example 35: Assessment of KCNQ2/3 Channel Activation Activity The in vitro effects of a compound of the application on cloned KCNQ2/3 potassium channels (encoded by the human KCNQ2/3 gene and expressed in HEK293 cells) are evaluated at room temperature using the QPatch HT® (Sophion Bioscience A/S, Denmark), an automatic parallel patch clamp system. Each test compound is evaluated at 0.01, 0.1, 1, 10 and 100 μM with each concentration tested in at least two cells (n≥2). The duration of exposure to each test compound concentration is 5 minutes. The baseline for each recording is established using a 5-10 minute vehicle application (HBPS+0.3% DMSO). A single test compound concentration is applied for a period of 5 minutes after the vehicle, followed by a 3 minute application of 30 μM flupirtine. Each recording ends with a supramaximal dose of 30 μM linopirdine. The % activation is calculated using the following equation by using leak subtracted responses: vehicle_response-compound_responsevehicle_response-flupirtine_response Example 36: Electrophysiology (Kalappa et al.,J. Neurosci.,35, 8829 (2015)) HEK293T cells are transfected with recombinant DNA (3-5 μg) using Lipofectamine 2000 (Invitrogen, Carlsbad, Calif.) and recorded 48 hours after transfection. All experiments are performed at room temperature using conventional whole-cell patch clamp technique. Recording electrodes are filled with internal solution containing (in mM): 132 K-Gluconate, 10 KCl, 4 Mg·ATP, 20 HEPES, and 1 EGTA·KOH, pH 7.2-7.3, and have resistances of 3-5 MΩ. The standard bath solution contains (in mM): 144 NaCl, 2.5 KCl, 2.25 CaCl2, 1.2 MgCl2, 10 HEPES, and 22 D-Glucose, pH 7.2-7.3. Series resistance is compensated by 75%. Osmolarity is adjusted to 300-305 mOsm and pH to 7.2-7.3 with NaOH. Voltage pulses are applied at 30s intervals from a holding potential of −85 mV to various test pulses before jumping down to −70 mV. These values are adjusted for the calculated junction potential of −15 mV. Data are acquired through a Multiclamp 700B amplifier (Molecular Devices, Sunnyvale, Calif.), low-pass filtered at 2 kHz and sampled at 10 kHz. The construct for testing KCNQ2/3 electrophysiology is created as described previously (Soh and Tzingounis,Mol. Pharmaco.,78, 1088 (2010)). Example 37: Maximal Electroshock Seizure Test (MES) In MES test, the ability of different doses of the test compound in preventing seizure induced by an electrical stimulus of 0.2 s in duration (50 mA at 60 Hz), delivered through the corneal electrodes primed with a drop of anesthetic/electrolyte solution (0.5% tetracaine hydrochloride in 0.9% saline) is tested. Mice are restrained by hand and released immediately following corneal stimulation that allows for the observation of the entire seizure episode. A maximal seizure in a test animal includes four distinct phases that includes, hind leg flexor component tonic phase (Phase I), hind leg extensor component of the tonic phase (Phase II), intermittent, whole-body clonus (Phase III), and muscular relaxation (Phase IV) followed by seizure termination (Woodbury & Davenport, 1952; Racine et al., 1972). Test compounds are tested for their ability to abolish hind limb tonic extensor component that indicates the compound's ability to inhibit MES-induced seizure spread. Compounds are pre-administered (i.p) and tested at 0.25, 0.5, 1 and 4 h time points for the abolishment of hind limb tonic extensor component after electrical stimulus. Example 38: Corneal-Kindled Mouse Model of Partial Seizures In corneal kindled seizure model, mice are kindled electrically with 3 s stimulation, 8 mA, 60 Hz delivered through corneal electrodes primed with 0.5% tetracaine hydrochloride in 0.9% saline, twice daily, until 5 consecutive stage V seizures are induced. Mice are considered kindled when they display at least 5 consecutive stage V seizures according to the Racine scale (Racine et al., 1972) including, mouth and facial clonus (stage I), Stage I plus head nodding (Stage II), Stage II plus forelimb clonus (Stage III), Stage III plus rearing (Stage IV), and stage IV plus repeated rearing and falling (Stage V) (Racine et al., 1972). At the completion of the kindling acquisition, mice are permitted a 3-day stimulation-free period prior to any drug testing. On the day of the experiment, fully kindled mice are pre-administered (i.p) with increasing doses of the test compound and challenged with the corneal kindling stimulus of 3 mA for 3 seconds 15 min. Mice are scored as protected (seizure score of <3) or not protected, (seizure score ≥4) based on the Racine scoring (Racine et al., 1972). Example 39: Assessment of Recombinantly Expressed Human Kv7.2/7.3 Xhannels Activation Ability The in vitro effects of a compound of the present application recombinantly expressed human Kv7.2/7.3 channels are assessed on Syncropatch high throughput electrophysiology platform. Cell Preparations: CHO cells stably expressing human Kv7.2/7.3 channels were cultured in Ham's F-12 media (Hyclone, Cat #SH30022.02) supplemented with 10% Fetal Bovine Serum, 1×MEM non-essential amino acids, and 400m/ml G418 at 37° C. in 5% CO2. On the day of Syncropatch, the cells were washed once in DPBS (Hyclone, Cat #SH30028.03) for approximately 30 seconds. 1 ml of 1×0.015% Trypsin-EDTA GIBCO Cat #25300-054) was added and swirled around to cover the bottom of the flask, and allowed to sit on the cells for about 4 minutes (approximately 90% of the cells were lifted by light tapping of the flask). 10 ml of cold media (Ham's F-12 media (Hyclone, Cat #SH30022.02) supplemented with 10% Fetal Bovine Serum, 1×MEM non-essential amino acids, and 400m/ml G418) was added to inactivate Trypsin. The cells were then triturated until a single cell suspension was achieved, and the cell count was performed. The cells were then diluted to a concentration of 5×105/ml and placed into the “cell hotel” on the deck of the Syncropatch at 10° C. for about 1 hour to recover. 40 μL of the cell suspension was dispensed into each well of a 384-well Syncropatch chip by the onboard pipettor at the beginning of each Syncropatch assay. Test Solution Preparations: The compounds to be tested were dissolved in DMSO to give 10 mM stock solutions. Eight-point dose response curves were created by performing semi-log serial dilutions from 10 mM compound stock solutions in 100% DMSO. Concentration-response curves were transferred to assay plates to give two-fold final compound concentration to account for the two-fold dilution with drug addition on the SyncroPatch. Final DMSO concentration in the assay was 0.3%. Final assay test concentrations were 30 μM to 0.01 μM or 1 μM to 0.0003 μM. Negative (0.3% DMSO) and positive (30 μM ML213) controls were included in each test run to assess pharmacological responsiveness. Assessment Protocol: Electrophysiological studies of the compounds were performed using the Nanion SyncroPatch automated patch clamp platform. Compound effects on Kv7 channels were assayed using a voltage protocol as shown inFIG.1. Kv7 channels were evaluated using a voltage protocol in which cells were voltage-clamped at a holding potential of −110 mV. Potassium currents were activated with a series of voltage steps from −110 mV to +50 mV in 10 mV intervals with 5.5 seconds between successive voltage steps. Each voltage step was 3 seconds in duration and immediately followed by a 1 second voltage step to −120 mV to generate an inward “tail” current to allow construction of activation (G-V) curves by plotting normalized peak tail current versus the potential of the activating voltage step. To obtain normalized values, peak current amplitudes for successive depolarizing pulses were normalized against the maximum tail current amplitude generated at +50 mV (Tatulian et al., Journal of Neuroscience 2001, 21 (15)). Data Analysis: Data was collected on the Syncropatch platform using PatchControl software (Nanion) and processed and analyzed using DataControl Software (Nanion). Normalized percent activation was calculated and activation curves were fit with a Boltzmann function to determine the midpoint voltage of activation (G-V midpoint) for both pre-compound and post-compound conditions for each of the 384-wells of a sealchip with Pipeline Pilot (Accelrys). The difference in G-V midpoint between pre-compound and post-compound conditions (A V0.5) was plotted as a function of concentration and concentration-response curves were fit with a three-parameter logistic equation {Y=Bottom+(Top−Bottom)/(1+10{circumflex over ( )}(Log EC50−X))} for determination of the EC50(Graphpad Prism). Assessment Results: Exemplary compounds of the present application were tested for their ability to produce a concentration-dependent hyperpolarizing shift in the midpoint of activation for heteromeric Kv7.2/7.3 channels. Eight of the compounds produced a quantifiable hyperpolarizing shift in activation as determined by a concentration-dependent shift in the midpoint that could be fit with a 3-parameter logistic equation. These data were combined with the initial 8-point concentration-response data in a single fit. Potency and efficacy data for each compound are summarized in Table 2 andFIGS.2A-2F. TABLE 2Cmpd #EC50(95% CI)Cmpd #EC50(95% CI)1B2A3C4A6A7A8A9A10 (control)B11A12A17A18A19B20A21B22A23B24BX (control)AA: 0.1 to 1.0 μM,B: 1.0 to 5 μM,C: 5 to 25 μM,D: 25 to 50 μM EQUIVALENTS Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific embodiments described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.
235,995
11858901
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT Compound of Formula (I) or a Salt Thereof. In a first aspect, the present invention relates to a compound of formula (I) (3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione) or a salt thereof. In the context of the invention, the term “salt” must be understood as an ionic compound formed by a cation of the amino group of the compound of formula (I), and a counterion (an anion), such as, for example, an anion of an acid, whether inorganic (such as, for example, hydrochloride, hydrobromide, hydroiodide, sulfate, nitrate, and phosphate, among others) or organic (such as, for example, acetate, trifluoroacetate, maleate, fumarate, citrate, oxalate, succinate, tartrate, malate, mandelate, methanesulfonate, and p-toluenesulfonate, among others). Preferably, the salt of compound (I) is a hydrochloride. In a preferred embodiment, the compound of the first aspect is 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia). In a preferred embodiment, said 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethyl benzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) is in solid form. In a preferred embodiment, the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethyl benzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) has an X-ray powder diffractogram measured with CuKα radiation with peaks at 8.5, 9.9, 14.7, 16.9, 19.5, 21.0, and 22.9° 2θ±0.2°θ. In a preferred embodiment, the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethyl benzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) has an X-ray powder diffractogram measured with CuKα radiation essentially as that ofFIG.2. The X-ray diffractograms can be recorded using a powder diffraction system with a copper anode emitting CuKα radiation with a wavelength of 1.54 Å, in particular, following the method described in the examples. In a preferred embodiment, the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethyl benzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) has a differential scanning calorimetry (DSC) diagram comprising an endothermic peak having an onset temperature of about 240.7° C.±2° C. and an exothermic peak having an onset temperature of about 248.6° C.±2° C. In a preferred embodiment, the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethyl benzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) has a differential scanning calorimetry (DSC) diagram essentially as that ofFIG.1. The differential scanning calorimetry diagram can be obtained as described in the examples. The onset temperature or “T onset” refers to the temperature resulting from extrapolating the baseline before the start of the transition and the baseline during energy absorption (tangent to the curve). It can be calculated as defined in standard DIN ISO 11357-1:2016(E). Process for Preparing the Compound of Formula (I) or a Salt Thereof. In a second aspect, the present invention relates to a process for preparing a compound of formula (I) or a salt thereof as defined in the first aspect, wherein the process comprises:a) reacting the 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethyl benzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) with iodine monochloride in the presence of an organic solvent to give 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) b) optionally treating the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) with a base to give the compound of formula (I) andc) optionally treating the compound of formula (I) with an acid to give a salt of the compound of formula (I). Step a) of the process yields 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia). The organic solvent used in step a) is any organic solvent suitable for carrying out the reaction. Examples of suitable organic solvents are dichloromethane, C1-C4alkanols, such as methanol, ethanol, n-propanol, isopropanol, n-butanol, sec-butanol, iso-butanol, tert-butanol, and a mixtures thereof; preferably dichloromethane, methanol and mixtures thereof; more preferably methanol; even more preferably a mixture of dichloromethane and methanol. In a preferred embodiment, in step a) 4 to 8 mL of organic solvent are used with respect to each gram of 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II), preferably 5 to 8 mL. In a preferred embodiment, step a) is performed at a temperature of 45° C. to 55° C., more preferably 48° C. to 52° C. Preferably, it is maintained under stirring in the temperature range indicated of 1.5 to 3 hours. In a particular embodiment, in step a) at least 2 mol of iodine monochloride are used with respect to each mol of 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II), preferably 2 to 4 mol, more preferably 2.5 to 3.5 mol. In a preferred embodiment, step a) is performed by adding a solution of 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) in the organic solvent, preferably methanol or dichloromethane, to a solution of iodine monochloride in the organic solvent, preferably methanol. The organic solvent of the solution of 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) can be the same as or different from the organic solvent of the solution of iodine monochloride (provided that the solvents are miscible with one another). In a particular embodiment it is the same, for example methanol. In another particular embodiment it is different, for example, dichloromethane is the organic solvent used in the solution of compound (II) and methanol is the organic solvent used in the solution of iodine monochloride. In a particular embodiment, the concentration of iodine monochloride in the solution of iodine monochloride in the organic solvent is 0.2 to 0.6 g/ml. In particular, the concentration of 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) in the solution of 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) in the organic solvent is 0.2 to 0.8 g/mL, in particular 0.2 to 0.6 g/mL. Preferably, the addition is performed at a temperature of 20° C. to 35° C., more preferably 20° C. to 25° C. After the addition, it is heated at a temperature of 45° C. to 55° C., more preferably 48° C. to 52° C. Preferably, it is maintained under stirring for 1.5 to 3 hours after the addition. In a particular embodiment, after the formation reaction of 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) in step a), this salt is isolated from the reaction medium. In particular, it is isolated by removing the solvent. In particular, the solvent can be removed by distillation at a pressure of less than 101325 Pa (less than 1 atm). Preferably, acetone is added after removing the solvent, in particular 2 to 5 ml of acetone are added with respect to each gram of 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia), and the resulting 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) in solid form is isolated by filtration. Step b) of the process is optional. Said step is performed to obtain 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione (1) in the form of a free base. Step b) comprises treating the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) with a base. Any base capable of yielding the compound of formula (I) in the form of a free base can be used. Bases suitable for said step b) are, for example, sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate, among others. Preferably said bases are used in solution in water. In particular, at least one mol of base is used with respect to each mol of 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia). In particular, the free base of the compound of formula (I) obtained is isolated by extraction with a water-immiscible organic solvent followed by removing the solvent, preferably by distillation at a pressure of less than 101325 Pa (less than 1 atm). The expression “water-immiscible organic solvent” refers to an organic solvent that is not water-miscible in at least one proportion of said solvent and water, i.e., in at least one proportion of said solvent and water two phases are produced. The phases can be rapidly identified by means of visual inspection. Miscibility/immiscibility is determined at a temperature in the range of 20° C. to 25° C. Examples of water-immiscible organic solvents are ethyl acetate, butyl acetate, isopropyl acetate, methyl ethyl ketone, methyl tert-butyl ether, diisopropyl ether, dichloromethane, chloroform, cyclohexane, hexane, heptane, pentane, toluene, xylene, and mixtures thereof. In a particular embodiment, the treatment of step b) is performed for 0.5 h to 2 h. In particular, said treatment is performed at a temperature of 20° C. to 35° C., more preferably 20° C. to 25° C. Step c) of the process is optional. Said step is performed to obtain a salt of the compound of formula (I), in particular a salt of the compound of formula (I) that is not the hydrochloride, since the hydrochloride is obtained directly in step a) of the process. Step c) comprises treating the compound of formula (I) in the form of a free base obtained in step b) with an acid to give a salt of the compound of formula (I). In a particular embodiment, the treatment of step c) is performed for 0.5 h to 2 h. In particular, said treatment is performed at a temperature of 20° C. to 35° C., more preferably 20 to 25° C. Acids suitable for said step are inorganic acids (such as, for example, HBr, HI, sulfuric acid, nitric acid, and phosphoric acid, among others) or organic acids (such as, for example, acetic acid, trifluoroacetic acid, maleic acid, fumaric acid, citric acid, oxalic acid, succinic acid, tartaric acid, malic acid, mandelic acid, methanesulfonic acid, and p-toluenesulfonic acid, among others). Preferably, the acid is not HCl, since the resulting salt would be the hydrochloride (Ia) which has been obtained in step a). Preferably said acids are used in solution in water, organic solvent (such as, for example, C1-C4alkanol), and mixtures thereof. In particular, at least one mol of acid is used with respect to each mol of 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia). In particular, the salt of the compound of formula (I) obtained is isolated by removing the solvent, preferably by distillation at a pressure of less than 101325 Pa (less than 1 atm). In a preferred embodiment, steps b) and c) are not performed, i.e., the product obtained by the process of the second aspect is 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia). In another embodiment, step b) is performed and, therefore, the product obtained by the process of the second aspect is 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione (I), i.e., in the form of a free base. In another embodiment, steps b) and c) are performed. Therefore, the product obtained by the process of the second aspect is a 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione salt. Preferably, the salt obtained in step c) is not the hydrochloride (Ia). In a preferred embodiment, the 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) used in step a) is obtained by reacting 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III) with a compound of formula (IV), wherein LG is a leaving group, in an organic solvent, and in the presence of a base. The radical “t-BOC”, “t-Boc”, “BOC”, or “Boc” refers to a tert-butoxycarbonyl group. The leaving group LG of the compound of formula (IV) can be selected from the group consisting of mesylate, tosylate, triflate, iodide, bromide, and chloride. Preferably the leaving group LG of the compound of formula (IV) is mesylate. Therefore, preferably the compound of formula (IV) is (R)-2-((tert-butoxycarbonyl)amino)-2-phenylethyl methanesulfonate (IVa). Bases suitable for reacting 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III) with the compound of formula (IV) are potassium carbonate, sodium carbonate, potassium hydrogen carbonate, sodium hydrogen carbonate, potassium hydroxide, and sodium hydroxide, among others; preferably the base is potassium carbonate (K2CO3). In a particular embodiment, at least 1 mol of base is used with respect to each mol of compound of formula (III), preferably 1 to 4 mol, more preferably 1 to 2 mol. Organic solvents suitable for reacting 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III) with the compound of formula (IV) are ketones (such as, for example, acetone and methyl isobutyl ketone), alcohols (such as, for example, methanol, ethanol, isopropanol, n-butanol, sec-butanol, tert-butanol, 1,2-propanediol, amyl alcohol, and isoamyl alcohol), and esters (such as, for example, ethyl acetate, isopropyl acetate, and tert-butyl acetate), N,N-dimethylformamide (DMF), and mixtures thereof; preferably the organic solvent is selected from the group consisting of acetone, DMF and mixtures thereof; more preferably, the organic solvent is acetone or DMF. In a particular embodiment, 3 to 7 mL of organic solvent are used with respect to each gram of 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III). In a particular embodiment, the reaction of 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III) with the compound of formula (IV) is performed in the presence of a phase-transfer catalyst. Preferably, the phase-transfer catalyst is present when the organic solvent used in the reaction is acetone. Preferably, the phase-transfer catalyst is not present when the organic solvent used in the reaction is DMF. Phase-transfer catalysts suitable are quaternary ammonium salts, such as, for example, tetrabutylammonium iodide and tetramethylammonium iodide; preferably the phase-transfer catalyst is tetrabutylammonium iodide (TBAI). In particular, 0.1 to 0.5 mol of phase-transfer catalyst are used with respect to each mol of 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III), preferably 0.15 to 0.25 mol. In a particular embodiment, the reaction of 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III) with the compound of formula (IV) is performed at a temperature of 50° C. to 70° C. In one embodiment, the reaction is performed at a temperature of 50° C. to 60° C., in particular when the organic solvent used in the reaction is acetone. In another embodiment, the reaction is performed at a temperature of 60° C. to 70° C., in particular when the organic solvent used in the reaction is DMF. Preferably, said treatment is performed for 12 h to 20 h. The reaction of 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III) with the compound of formula (IV) yields 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II). In a particular embodiment, said compound (II) is isolated by removing the solvent, preferably by distillation at a pressure of less than 101325 Pa (less than 1 atm). After removing the solvent or directly on the reaction mixture (i.e. without performing the step removing the solvent), water and a water-immiscible organic solvent, preferably ethyl acetate and/or tert-butyl methyl ether, are added. Optionally, the pH is adjusted to 7 to 8, preferably by adding aqueous hydrochloric acid. The water-immiscible organic solvent phase is separated, and said solvent is removed, preferably by distillation at a pressure of less than 101325 Pa (less than 1 atm). Process for Preparing Elagolix Sodium (V) The third aspect of the invention relates to a process for preparing elagolix sodium (V) comprising:a) reacting the compound of formula (I) or a salt thereof as defined in the first aspect with 2-fluoro-3-methoxyphenylboronic acid (VI) in the presence of a palladium catalyst, a phosphine-type ligand, and a base to give 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) optionally performing steps b1) to b3):b1) treating the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) with HCl to give 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (VIIa) b2) isolating the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (VIIa); andb3) treating the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (VIIa) with a base to give 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII);c) reacting the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) obtained in step a) or in step b3) with a C1-4alkyl 4-halobutyrate to obtain a compound of formula (VIII) wherein R represents a C1-4alkyl group; andd) hydrolyzing the ester group of the compound of formula (VIII) by treatment with NaOH to obtain elagolix sodium (V) Step a) of the process is reacting the compound of formula (I) or a salt thereof as defined in the first aspect with 2-fluoro-3-methoxyphenylboronic acid (VI) in the presence of a palladium catalyst, a phosphine-type ligand, and a base to give 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII). In a preferred embodiment, in step a) the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (Ia) is reacted. Palladium catalysts suitable for the reaction are palladium(II) acetate (Pd(OAc)2), palladium(II) bromide and palladium(II) chloride; preferably palladium(II) acetate. In a particular embodiment, in step a) 0.001 to 0.05 mol of palladium catalyst are used with respect to each mol of the compound of formula (I) or a salt thereof, preferably 0.005 to 0.05 mol, more preferably 0.0085 to 0.025 mol. Phosphine-type ligands suitable for the reaction are tri-tert-butyltetraphosphonium tetrafluoroborate, triphenylphosphine, tricyclohexylphosphine, tri-tert-butylphosphine, tri-n-butylphosphine, tri-n-butylphosphonium tetrafluoroborate, and triethylphosphonium tetrafluoroborate, among others; preferably the phosphine-type ligand is tri-tert-butyltetraphosphonium tetrafluoroborate. In a particular embodiment, in step a) 0.005 to 0.05 mol of phosphine-type ligand are used for each mol of compound of formula (I) or a salt thereof, preferably 0.0085 to 0.025 mol. Bases suitable for the reaction are potassium phosphate, potassium carbonate, potassium hydroxide, or hydrates thereof, preferably potassium phosphate or a hydrate thereof. In a particular embodiment, in step a) 2 to 6 mol of base are used with respect to each mol of compound of formula (I) or a salt thereof, preferably 3 to 5 mol. In another embodiment, in step a) 2 to 4 mol of base are used with respect to each mol of compound of formula (I) or a salt thereof. In a preferred embodiment, in step a) the palladium catalyst is palladium(II) acetate, the phosphine-type ligand is tri-tert-butyltetraphosphonium tetrafluoroborate, and/or the base is potassium phosphate or a hydrate thereof. In a more preferred embodiment, in step a) the palladium catalyst is palladium(II) acetate, the phosphine-type ligand is tri-tert-butyltetraphosphonium tetrafluoroborate, and the base is potassium phosphate or a hydrate thereof. In a particular embodiment, step a) is performed in a suitable solvent. Examples of suitable solvents are mixtures of water and organic solvent (such as acetone, tetrahydrofuran, 2-methyltetrahydrofuran and dioxane); preferably the solvent is a mixture of water and acetone. In a particular embodiment, the proportion by volume of the water and the organic solvent in the mixture is 1:5 to 5:1, preferably 1:2 to 2:1, more preferably about 1:1. In a particular embodiment, the reaction of step a) is performed at a temperature of 50° C. to 70° C. Preferably, said treatment is performed for 3 h to 24 h. Preferably step a) is performed under inert atmosphere, such as, for example, a nitrogen or argon atmosphere. The reaction of step a) of the third aspect yields 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII). In a particular embodiment, said compound (VII) is isolated by separating the aqueous phase; treating the remaining organic phase with an aqueous acid solution, preferably with a pH of 1 to 2 (preferably phosphoric acid); separating the organic phase; treating the remaining aqueous phase with a base (preferably potassium carbonate) to a pH of 9 to 10 and with water-immiscible organic solvent (preferably isopropyl acetate); separating the aqueous phase; and removing the organic solvent from the resulting organic phase, preferably by distillation at a pressure of less than 101325 Pa (less than 1 atm). In a preferred embodiment, step a) further comprises crystallizing the compound (VII) obtained from a suitable solvent, such as, for example, a mixture of acetone and water. In a particular embodiment, the proportion by volume of acetone and water in the mixture is 99:1 to 50:50, preferably 80:20 to 55:45, more preferably 70:30 to 60:40. In another particular embodiment, the proportion by volume of acetone and water in the mixture is from 80:20 to 45:55, preferably 70:30 to 45:55. In another particular embodiment, the proportion by volume of acetone and water is about 2:1. In another particular embodiment, the proportion by volume of acetone and water is about 1:1. In a particular embodiment, 5 to 10 mL of solvent are used with respect to each gram of the compound (VII), preferably 6 to 8 mL of solvent with respect to each gram of the compound (VII). In a preferred embodiment, the compound of formula (VII) is obtained as a solid, in particular a crystalline solid. Thus, in another aspect, the invention relates to a compound of formula (VII) in the form of a solid, preferably a crystalline solid. In a preferred embodiment, the compound of formula (VII) in solid form has an X-ray powder diffractogram measured with CuKα radiation with peaks at 4.6, 9.1, 11.7, 12.2, 13.2, 16.0, 18.2, 18.6, 22.2, 24.7 2θ±0.2°θ. In another preferred embodiment, the compound of formula (VII) in solid form has an X-ray powder diffractogram measured with CuKα radiation essentially as that ofFIG.4. In another preferred embodiment, the compound of formula (VII) in solid form has an X-ray powder diffractogram measured with CuKα radiation essentially as that ofFIG.6. The X-ray diffractograms can be recorded using a powder diffraction system with a copper anode emitting CuKα radiation with a wavelength of 1.54 Å, in particular, following the method described in the examples. In a preferred embodiment, the compound of formula (VII) in solid form has a differential scanning calorimetry (DSC) diagram comprising an endothermic peak having an onset temperature of in the range of 170 to 175° C. In another preferred embodiment, the compound of formula (VII) in solid form has a differential scanning calorimetry (DSC) diagram essentially as that ofFIG.3. In another preferred embodiment, the compound of formula (VII) in solid form has a differential scanning calorimetry (DSC) diagram essentially as that ofFIG.5. The differential scanning calorimetry diagram can be obtained as described in the examples. The onset temperature or “T onset” refers to the temperature resulting from extrapolating the baseline before the start of the transition and the baseline during energy absorption (tangent to the curve). It can be calculated as defined in standard DIN ISO 11357-1:2016(E). In a preferred embodiment, the compound of formula (I) or a salt thereof used in step a) is prepared by a process as defined in the second aspect. Next, optional steps b1) to b3) can be performed. Said steps, in particular steps b1) and b2), may be performed to increase the purity of 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) obtained in step a). In step b1), the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) obtained in step a) is treated with HCl to give 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (VIIa). Said treatment is preferably performed with an aqueous solution of HCl and in the presence of an organic solvent, such as, for example, isopropyl acetate. In particular, at least 1 mol of HCl is added with respect to each mol of 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII), preferably 1 to 3 mol, more preferably 1 to 1.5 mol. In a particular embodiment, the treatment of step b1) is performed at a temperature of 20° C. to 35° C., more preferably 20° C. to 25° C. Preferably, said treatment is performed for 10 min to 60 min. The treatment of step b1) yields 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (VIIa), which is isolated in step b2). Said isolation of step b2) can be performed by removing the solvent, preferably by distillation at a pressure of less than Pa (less than 1 atm). In a preferred embodiment, after removing the solvent the hydrochloride (VIa) is crystallized in a suitable organic solvent, such as, for example, 2-methyltetrahydrofuran or tetrahydrofuran (preferably 2-methyltetrahydrofuran). The solid hydrochloride (VIa) obtained is separated from the crystallization solvent, for example by filtration. Next, step b3) of treating 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione hydrochloride (VIIa) with a base to give 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII), is performed. In a particular embodiment, the base used in step b3) is selected from the group consisting of diisopropylethylamine, triethylamine, tert-butylamine, and diethylamine, preferably diisopropylethylamine. In a particular embodiment, step b3) is performed in the presence of an organic solvent, preferably selected from the group consisting of dimethylsulfoxide, toluene, and dimethylformamide, more preferably dimethylformamide. Step b3) is preferably performed at a temperature in the range of 20° C. to 35° C., more preferably 20° C. to 25° C. In one embodiment, steps b1)-b3) are performed. In a preferred embodiment, steps b1)-b3) are not performed. Next, step c) is of reacting the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) obtained in step a) or in step b3) with a C1-4alkyl 4-halobutyrate is performed to obtain a compound of formula (VIII) wherein R represents a C1-4alkyl group. If optional steps b1) to b3) have not been performed, the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) obtained in step a) is used in step c). If optional steps b1) to b3) have been performed, the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) obtained in step b3) is used in step c). Preferably, steps b1) to b3) are not performed. Therefore, in a preferred embodiment, the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) obtained in step a) is used in step c). The term “halo” in the expression “C1-4alkyl 4-halobutyrate” refers to a halogen atom, i.e., an atom selected from the group consisting of fluorine, chlorine, bromine, and iodine, preferably bromine. The term “C1-4alkyl” in the expression “C1-4alkyl 4-halobutyrate” refers to a radical having a linear or branched chain consisting of hydrogen atoms and 1 to 4 carbon atoms, with no unsaturations and being bound to the rest of the molecule by means of a single bond. Examples of C1-4alkyl are methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, iso-butyl, and tert-butyl, preferably ethyl. In a preferred embodiment, the C1-4alkyl 4-halobutyrate used in step c) is ethyl 4-bromobutyrate, such that the product obtained in said step c) is 4-((R)-2-[5-(2-fluoro-3-methoxyphenyl)-3-(2-fluoro-6-trifluoromethylbenzyl)-4-methyl-2,6-dioxo-3,6-dihydro-2H-pyrimidin-1-yl]-1-phenylethylamino)-butyric acid ethyl ester (VIIIa) In a particular embodiment, at least 1 mol of C1-4alkyl 4-halobutyrate is added with respect to each mol of 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII), preferably 1 to mol, more preferably 1.3 to 1.7 mol. In a particular embodiment, in step c) a base selected from the group consisting of diisopropylethylamine, triethylamine, tert-butylamine, and diethylamine, preferably diisopropylethylamine, is used. In a particular embodiment, the reaction in step c) is performed in the presence of a phase-transfer catalyst. Suitable phase-transfer catalysts are quaternary ammonium salts, such as, for example, tetrabutylammonium iodide and tetramethylammonium iodide; preferably the phase-transfer catalyst is tetrabutylammonium iodide (TBAI). In particular, 0.1 to 0.5 mol of phase-transfer catalyst are used with respect to each mol of 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII), preferably 0.15 to 0.25 mol. In a particular embodiment, step c) is performed at a temperature in the range of 70° C. to 100° C., preferably 80° C. to 90° C., more preferably 80° C. to 85° C. maintaining stirring for a time comprised between 8 and 24 hours, preferably between 8 and 20 hours, more preferably between 12 and 16 hours. In a particular embodiment, the compound of formula (VIII) obtained in step c) (preferably compound (VIIIa)), is purified before performing step d). In a particular embodiment, purification is performed by i) adding to the reaction medium of step c) water and a water-immiscible organic solvent, preferably isopropyl acetate, ii) treating the organic phase of step i) with an aqueous acid solution, iii) separating said aqueous solution from the organic phase, iv) neutralizing the aqueous solution with a base, v) adding the same solvent mentioned in step i) again, vi) separating the organic phase from said solvent containing the product (VIII), and v) removing the solvent to obtain the product (VIII). Next, step d) of hydrolysis of the ester group of the compound of formula (VIII) is performed by treatment with NaOH to obtain elagolix sodium (V). In a particular embodiment, step d) of hydrolysis is performed in a solvent which is selected from the group consisting of water, C1-C4alkanol, mixtures of C1-C4alkanol and water, preferably isopropanol or a mixture of isopropanol and water. In a particular embodiment, step d) of hydrolysis is performed using as a base an aqueous solution of NaOH at a temperature in the range of 20° C. to 50° C., more preferably 30° C. to 40° C. In a fourth aspect, the invention relates to the use of a compound of formula (I) or a salt thereof as defined in the first aspect, preferably the hydrochloride (Ia), in a process for preparing elagolix sodium (V). In particular embodiments of the fourth aspect of the invention, the use of the compound (I) or a salt thereof as defined in the first aspect, preferably the hydrochloride (Ia), is performed following the process described in the third aspect of the invention. In the context of the present invention, the terms “approximate” and “about” refer to the value these expression characterize ±5% of said value. In the context of the present invention, the term “acid” refers to a substance capable of donating a proton (to a base). In the context of the present invention, the term “base” refers to a substance capable of accepting a proton (from an acid). To help better understand the preceding ideas, several examples of the experimental methods and embodiments of the present invention are described below. Said examples are merely illustrative. EXAMPLES Example 1: Obtaining 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) 10 g (33.09 mmol) of 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III), 6.8 g (49.62 mmol) of K2CO3and 2.4 g (6.6 mmol) of tetrabutylammonium iodide were mixed with 50 mL of acetone at the temperature of about 20° C. Subsequently, 13.6 g (43.12 mmol) of (R)-2-((tert-butoxycarbonyl)amino)-2-phenylethyl methanesulfonate (IVa) were added and the obtained mixture was heated at the temperature of about 55° C. and maintained under stirring for about 16 hours at said temperature. Once this maintenance was finished, the solvent was vacuum distilled and 50 mL of ethyl acetate and 50 mL of water were added to the residue thus obtained. A 1 M aqueous solution of HCl was slowly added, maintaining the temperature between 20 and 25° C. until achieving a pH of between 7 and 8. The aqueous phase was separated and treated with 3 fractions of 30 mL each of ethyl acetate. All the organic extracts were pooled and the solvent was removed by means of vacuum to obtain a slightly yellowish oily residue to which 45 mL of methanol were added, obtaining complete dissolution of the residue. Example 2: Obtaining 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethyl benzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione Hydrochloride Salt (Ia) 16.1 g (99.24 mmol) of iodine monochloride (ICI) were dissolved in 40 mL of methanol at the temperature of about 10° C. The methanol solution previously obtained according to the methodology described in Example 1 comprising 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) was added to the iodine monochloride solution, maintaining the temperature between 20 and 25° C. Once the addition was finished, the obtained solution was heated to about 50° C. and was maintained under stirring for 2 hours at the mentioned temperature. Once the maintenance was finished, the solvent was vacuum distilled and 50 mL of acetone were slowly added to the obtained oily residue at the temperature of between and 25° C. The addition of acetone caused a solid precipitate to appear almost immediately. The obtained mixture was maintained for 1 hour under stirring at the mentioned temperature. The resulting solid was isolated by filtration, washed with two fractions of 25 mL of acetone, and finally dried at the temperature of 50° C. to obtain 15.6 g (80.8% yield) of a white solid corresponding to the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride salt (Ia) (UHPLC purity: 98.9%). 1H-NMR (d6-DMSO, 400 MHz) δ (ppm): 8.70 (2H, s broad), 7.65-7.48 (3H, m), 7.40-7.32 (5H, m), 5.40-5.29 (2H, dd), 4.47 (1H, t), 4.25 (2H, dd), 2.65 (3H, s). 13C-NMR (d6-DMSO, 100 MHz) δ (ppm): 161.87, 159.47, 159.41, 154.19, 150.98, 134.70, 129.93, 129.84, 129.01, 128.58, 127.38, 122.61, 122.34, 122.22, 121.34, 121.10, 74.80, 52.26, 45.45, 44.60, 25.66. The DSC of this compound is shown inFIG.1and the XRPD is shown inFIG.2. Example 3: Obtaining 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione Hydrochloride Salt (VIIa) 12.3 g (21.09 mmol) of 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride salt (Ia), 4.6 g (27.42 mmol) of 2-fluoro-3-methoxyphenylboronic acid (VI), and 17.9 g (84.36 mmol) of K3PO4·3H2O were dissolved in 74 mL of a 50:50 mixture of acetone/water at the temperature of about 20° C. under nitrogen atmosphere. Subsequently 24 mg (0.1 mmol) of palladium(II) acetate and 62 mg (0.21 mmol) of tri-tert-butyl-tetraphosphonium tetrafluoroborate were added. The mixture thus obtained was heated at the temperature of about 60° C. and maintained at said temperature for 16 hours under nitrogen atmosphere. Once the maintenance was finished, the reaction mixture was cooled to the temperature of about 20° C., and 50 mL of isopropyl acetate were added. The aqueous phase was separated and treated with 2 fractions of 50 mL each of isopropyl acetate. All the organic extracts were pooled, and 100 mL of water and H3PO4were added to a pH of the mixture of between 1 and 2. The aqueous phase was separated, and 50 mL of isopropyl acetate were added and, slowly, K2CO3was also added to a pH of between 9 and 10. The aqueous phase was separated and treated successively with two fractions of 50 mL each of isopropyl acetate. The solvent was removed from the thus pooled organic phases by vacuum distillation to obtain 11.36 g of an orange colored oil comprising 3-((R)-2-amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) (UHPLC purity: 89.9%). The obtained oil was dissolved in 57 mL of isopropyl acetate, and 1.9 mL of a 12 M aqueous solution of HCl were slowly added, maintaining the temperature between 20 and 25° C. The obtained mixture was maintained under stirring at the indicated temperature for 30 minutes. Once the maintenance was finished, the solvent was vacuum distilled, and 100 mL of 2-methyltetrahydrofuran were added to the obtained oily residue at the temperature of between 20 and 25° C. The resulting mixture was heated at the reflux temperature and maintained under stirring for 10 minutes. Subsequently, the obtained solution was slowly cooled to an approximate temperature of 20° C., observing the occurrence of a whitish solid. After a maintenance for 30 minutes at the indicated temperature, the resulting solid was isolated by filtration, washed with two fractions of 15 mL of 2-methyltetrahydrofuran, and finally dried at the temperature of 50° C. to obtain 10.8 g (87.8% yield) of a white solid corresponding to the 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione hydrochloride salt (VIIa) (UHPLC purity: 99.03%). Example 4: Obtaining 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) 150.0 g (257.0 mmol) of 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride salt (Ia), 48.0 g (282.4 mmol) of 2-fluoro-3-methoxyphenylboronic acid (VI), and 205.0 g (769.8 mmol) of K3PO4·3H2O were dissolved in 1200 mL of a 50:50 mixture of acetone/water at the temperature of about 20° C. and nitrogen atmosphere. Subsequently 290 mg (1.3 mmol) of palladium(II) acetate and 750 mg (2.6 mmol) of tri-tert-butyl-tetraphosphonium tetrafluoroborate were added. The mixture thus obtained was heated at the temperature of about 60° C. and was maintained at said temperature for 20 hours under nitrogen atmosphere. Once the maintenance was finished, the reaction mixture was cooled to the temperature of about 20° C. and the aqueous phase was separated. The solvent was removed from the organic phase by vacuum distillation, and 750 mL of isopropyl acetate and 750 mL of water were added to the obtained residue. The aqueous phase thus obtained was separated and treated with 450 mL of isopropyl acetate. The two organic extracts were pooled, and a solution previously prepared from 50 mL of 85% H3PO4and from 600 mL of water was added. The aqueous phase was separated, and a solution previously prepared from 20 mL of 85% H3PO4and from 600 mL of water was added to the organic phase. The aqueous phase thus obtained was separated. The two combined aqueous phases were treated with two fractions of 450 mL of isopropyl acetate. 600 mL of isopropyl acetate and, slowly, K2CO3were added to the aqueous phase to a pH of about 9. The aqueous phase was separated and treated with 600 mL of isopropyl acetate. The solvent was removed from the thus pooled organic phases by vacuum distillation to obtain 141.2 g of a slightly brown colored solid comprising 3-((R)-2-amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) (UHPLC purity: 94.3%). The obtained solid was dissolved in 1120 mL of a 50:50 mixture of acetone/water at the temperature of about 20° C. The obtained mixture was heated at the reflux temperature and maintained 15 minutes under stirring. The solution thus obtained was slowly cooled until the occurrence of a solid precipitate (at the temperature of about 50° C.). The mixture was maintained under stirring at the temperature of about 40° C. for 2 hours and was then slowly cooled to the temperature of about 20° C. After a maintenance for 2 hours at the indicated temperature, the resulting solid was isolated by filtration, washed with two fractions of 150 mL each of a 2:1 acetone/water mixture, and finally dried at the temperature of 50° C. to obtain 125.0 g (89.2% yield) of a white solid corresponding to 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) (UHPLC purity: 99.4%). The DSC of this compound is shown inFIG.3and the XRPD is shown inFIG.4. Example 5: Obtaining 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) Following the same conditions of the method described in Example 4, starting from 25 g of (42.8 mmol) of 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride salt (Ia), 24.7 g of a solid corresponding to 3-((R)-2-amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) were obtained (UHPLC purity: 94.09%). The obtained solid was dissolved in 100 mL of acetone and 50 mL of water at the temperature of about 20° C. The obtained mixture was heated at the reflux temperature and maintained 10 minutes under stirring. The solution thus obtained was slowly cooled until the occurrence of a solid precipitate (at the temperature of about 40° C.). The mixture was maintained under stirring at the temperature of about 40° C. for 2 hours and was then slowly cooled to the temperature of about 20° C. After a maintenance for 2 hours at the indicated temperature, the resulting solid was isolated by means of filtration, washed with two fractions of 30 mL each of a 4:2 acetone/water mixture, and finally dried at the temperature of 50° C. to obtain 20.8 g (88.9% yield) of a white solid corresponding to 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) (UHPLC purity: 99.62%). Example 6: Obtaining 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) 550 g (1.82 mol) of 1-(2-fluoro-6-trifluoromethyl-benzyl)-6-methyl-1H-pyrimidine-2,4-dione (III) and 914.3 g (6.62 mol) of K2CO3were mixed with 2.75 L of N,N-dimethylformamide (DMF) at the temperature of about 20° C. under a nitrogen atmosphere. Subsequently, 918.4 g (2.91 mol) of (R)-2-((tert-butoxycarbonyl)amino)-2-phenylethyl methanesulfonate (IVa) were added and the obtained mixture was heated at the temperature of about 65° C. and maintained under stirring for about 16 hours at said temperature. Once this maintenance was finished, 5.5 L of tert-butyl methyl ether and 2.75 L of water were added to the reaction mixture maintaining the temperature between 35 and 40° C. The aqueous-DMF phase was separated and treated with one fraction of 2.75 L tert-butyl methyl ether. The aqueous-DMF phase was separated, the two organic extracts were pooled and 2.75 L of water were added. The aqueous phase was separated and 2.75 L of a 2N NaOH aqueous solution was added on the organic phase. The aqueous phase was separated and the solvent of the organic phase was removed by means of vacuum to obtain a yellowish oily residue to which 1425 mL of dichloromethane were added, obtaining complete dissolution of the residue. Example 7: Obtaining 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethyl benzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione Hydrochloride Salt (Ia) 886.48 g (5.46 mol) of iodine monochloride (ICI) were dissolved in 3320 mL of methanol at a temperature between 10 and 20° C. The dichloromethane solution previously obtained according to the methodology described in Example 6 comprising 3-((R)-2-(tert-butoxycarbonyl)amino-2-phenylethyl)-1-(2-fluoro-6-trifluoro methylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (II) (949.1 g, theoretical) was added to the iodine monochloride solution, maintaining the temperature between 20 and 25° C. Once the addition was finished, the obtained solution was heated to about 50° C. and was maintained under stirring for 3 hours at the mentioned temperature. Once the maintenance was finished, the solvent was vacuum distilled and 1000 mL of acetone were slowly added to the obtained residue at the temperature of between 20 and 25° C. and the solvent was removed by means of vacuum. 3 L of acetone were added to the residue thus obtained at the temperature of between 20 and 25° C. and the mixture was maintained under stirring for 3 hours at the mentioned temperature. The resulting solid was isolated by filtration, washed with three fractions of 500 mL each of acetone, and finally dried at the temperature of 50° C. to obtain 870.3 g (81.9% yield) of a white solid corresponding to the 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride salt (Ia) (UHPLC purity: 98.73%). Example 8: Obtaining 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) 750.0 g (1.285 mol) of 3-((R)-2-(amino-2-phenylethyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-5-iodo-6-methyl-1H-pyrimidine-2,4-dione hydrochloride salt (Ia), 240.21 g (1.413 mol) of 2-fluoro-3-methoxyphenylboronic acid (VI), and 1026.59 g (3.855 mol) of K3PO4·3H2O were dissolved in 6 L of a 50:50 mixture of acetone/water at the temperature of about 20° C. and under nitrogen atmosphere. Subsequently 1.442 g (6.4 mmol) of palladium(II) acetate and 3.728 g (12.8 mmol) of tri-tert-butyl-tetraphosphonium tetrafluoroborate were added. The mixture thus obtained was heated at the temperature of about 60° C. and was maintained at said temperature for 4 hours under nitrogen atmosphere. Once the maintenance was finished, the reaction mixture was cooled to the temperature of about 30° C. and the aqueous phase was separated. The solvent was removed from the organic phase by vacuum distillation, and 3750 mL of isopropyl acetate and 3750 mL of water were added to the obtained residue. The aqueous phase thus obtained was separated and treated with 1500 mL of isopropyl acetate. The two organic extracts were pooled, and a solution previously prepared from 250 mL of 85% H3PO4and from 2 L of water was added. The aqueous phase was separated, and a solution previously prepared from 50 mL of 85% H3PO4and from 2 L of water was added to the organic phase. The aqueous phase thus obtained was separated. The two pooled aqueous phases were treated with two fractions of 2 L each of isopropyl acetate. 3 L of isopropyl acetate were added to the aqueous phase and, slowly, K2CO3was also added to a pH of about 8.5. The aqueous phase was separated and treated with 1500 mL of isopropyl acetate. The solvent was removed from the pooled organic phases by vacuum distillation. 750 mL of acetone were added to the residue thus obtained and the solvent was removed by vacuum distillation. The obtained residue was dissolved in 4.2 L of a 2:1 mixture of acetone/water at the temperature of about 20° C. The obtained mixture was heated at the reflux temperature and maintained 15 minutes under stirring at the indicated temperature. The solution thus obtained was slowly cooled until the occurrence of a solid precipitate (at the temperature of about 50° C.). The mixture was maintained under stirring at the temperature of between 40 and 45° C. for about 2 hours and was then slowly cooled to the temperature of about 20° C. After a maintenance for 2 hours at the indicated temperature, the resulting solid was isolated by filtration, washed with two fractions of 750 mL each of a 2:1 acetone/water mixture, and finally dried at the temperature of 50° C. to obtain 615.4 g (87.8% yield) of a white solid corresponding to 3-((R)-2-(amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) (UHPLC purity: 99.82%). 1H-NMR (d6-DMSO, 400 MHz) δ (ppm): 7.64 (1H, m), 7.52-7.48 (2H, m), 7.28-7.22 (4H, m), 7.20-7.13 (3H, m), 6.73-6.55 (1H, m), 5.29 (2H, s), 4.91-4.83 (1H, m), 4.18-3.93 (3H, m), 3.85 (3H, s), 2.09 (3H, s). 13C-NMR (d6-DMSO, 100 MHz) δ (ppm): 166.85, 161.95, 160.41, 159.51, 150.85, 150.83, 150.77, 150.69, 148.34, 147.42, 141.58, 129.73, 128.07, 127.25, 126.93, 124.01, 123.57, 122.57, 121.30, 121.06, 113.38, 106.78, 60.64, 55.95, 47.10, 42.86, 28.84, 18.04. The DSC of this compound is shown inFIG.5and the XRPD is shown inFIG.6. Example 9: Obtaining 4-((R)-2-[5-(2-fluoro-3-methoxyphenyl)-3-(2-fluoro-6-trifluoromethylbenzyl)-4-methyl-2,6-dioxo-3,6-dihydro-2H-pyrimidin-1-yl]-1-phenylethyl amino)-butyric Acid Ethyl Ester (VIIIa) 100 g (183.3 mmol) of 3-((R)-2-amino-2-phenylethyl)-5-(2-fluoro-3-methoxyphenyl)-1-(2-fluoro-6-trifluoromethylbenzyl)-6-methyl-1H-pyrimidine-2,4-dione (VII) and 13.54 g of tetrabutylammonium iodide (36.7 mmol) were mixed with 500 mL of isopropyl acetate at the temperature of about 20° C. 120 mL of diisopropylethylamine, subsequently 500 mL of water, and finally 39.5 mL (276.0 mmol) of ethyl 4-bromobutyrate were slowly added. The resulting mixture was heated at the temperature of 80-85° C. and maintained under stirring at said temperature for 16 hours. Once the maintenance was finished, the reaction mass was cooled at the temperature of about 20° C., and the aqueous phase was separated and treated with 1000 mL of isopropyl acetate. The aqueous phase was separated, and a solution previously prepared from 100 mL of 85% H3PO4and from 2000 mL of water was added to the pooled organic phases. The aqueous phase was separated, and a solution previously prepared from 40 mL of 85% H3PO4and from 1000 mL of water was added to the organic phase. The aqueous phase thus obtained was separated. The two combined aqueous phases were treated with 600 mL of isopropyl acetate. The organic phase was separated, and 1500 mL of isopropyl acetate and, slowly, K2CO3were added to the aqueous phase until a pH of about 8. The aqueous phase was separated and treated with 1000 mL of isopropyl acetate. The thus pooled organic phases were treated with 1000 mL of an aqueous solution of 8% NaHCO3. The solvent was removed from the organic phase thus obtained by means of vacuum distillation to obtain 111.3 g of a residue comprising 4-((R)-2-[5-(2-fluoro-3-methoxyphenyl)-3-(2-fluoro-6-trifluoromethylbenzyl)-4-methyl-2,6-dioxo-3,6-dihydro-2H-pyrimidin-1-yl]-1-phenylethylamino)-butyric acid ethyl ester (VIIIa) (UHPLC: 90.21%). Example 10: Obtaining 4-((R)-2-[5-(2-fluoro-3-methoxyphenyl)-3-(2-fluoro-6-trifluoro methylbenzyl)-4-methyl-2,6-dioxo-3,6-dihydro-2H-pyrimidin-1-yl]-1-phenylethylamino)-butyric Acid Sodium Salt (V) The residue obtained in the preceding example comprising 4-((R)-2-[5-(2-fluoro-3-methoxyphenyl)-3-(2-fluoro-6-trifluoromethylbenzyl)-4-methyl-2,6-dioxo-3,6-dihydro-2H-pyrimidin-1-yl]-1-phenylethylamino)-butyric acid ethyl ester (VIIIa) was dissolved in 240 mL of isopropanol. An aqueous solution previously prepared with 18.4 g of NaOH (460.0 mmol) and 240 mL of water was added to said solution at the temperature of about 20° C. The resulting mixture was heated at the temperature of about 35° C. and maintained under stirring at said temperature for 2 hours. Once the maintenance was finished, the solvent was removed with a vacuum at the maximum temperature of 45° C., and 700 mL of water and 200 mL of isopropyl acetate were added. The aqueous phase was separated and treated with two fractions of 200 mL each of isopropyl acetate. 200 g of NaCl were added, and the pH of the solution was adjusted to about 12 by means of an aqueous solution of 20% NaOH. 500 mL of methyl isobutyl ketone were subsequently added to the treated aqueous phase. The aqueous phase was separated, and two fractions of 500 mL each of methyl isobutyl ketone were added to same. The pooled organic phases were treated with a fraction of 300 mL of an aqueous solution of 20% NaCl. The solvent was removed from the organic phase by vacuum distillation at the maximum temperature of about 45° C., and the obtained residue was dissolved in 500 mL of methyl isobutyl ketone. The obtained mixture was filtered through a filter made up of a layer of diatomaceous earth and a 0.20 micra filter, washing the filter with two fractions of 70 mL each of methyl isobutyl ketone. The filtered solution was slowly added to 2000 mL of heptane, maintaining the temperature at about 20° C. The resulting mixture was maintained under stirring at the indicated temperature for 60 minutes. The resulting solid was isolated by filtration, washed with two fractions of 200 mL each of heptane, and finally dried at the temperature of 40° C. to obtain 89.8 g (75.0% yield) of a virtually white solid corresponding to 4-((R)-2-[5-(2-fluoro-3-methoxyphenyl)-3-(2-fluoro-6-trifluoromethylbenzyl)-4-methyl-2,6-dioxo-3,6-dehydro-2H-pyrimidin-1-yl]-1-phenylethylamino)-butyric acid sodium salt (V) (elagolix sodium salt), with a purity of 99.49% by UHPLC. Assay Conditions Analysis by proton nuclear magnetic resonance (1H-NMR) and13C-NMR was performed in a 400 MHz Brucker Avance III spectrometer. The chemical shifts are referenced to the DMSO-d6signal (2.49 ppm for proton and 39.5 ppm for carbon). DSC analysis was performed in a Mettler Toledo 822e apparatus with STARe SW15 software. Parameters: heating range of 30 to 300° C. with a ramp of 10° C./min and N2flow of 50 ml/min. The measurement is taken with a perforated closed capsule. XRPD analysis of compound (Ia) was performed using a Siemens D-500 model X-ray powder diffractometer equipped with a copper anode using Cu Karadiation. Scanning parameters: 4-50 degrees 2θ, continuous scan, ratio: 1.2 degrees/minute. XRPD analysis of compound (VII) was performed using a BRUKER D2 PHASER model X-ray powder diffractometer equipped with a copper anode using Cu Karadiation (1.54060 Å). Scanning parameters: 3-50 degrees 2θ, continuous scan, ratio: 5.6 degrees/minute. The purity of the obtained products was analyzed by ultra-high-performance liquid chromatography (UHPLC) technique in a Waters Acquity model apparatus, provided with a photodiode detector and thermostated oven for the column. The column used is an HSST3 column (2.1×100 mm and 1.8 μm) and mobile phases A (50 mM ammonium acetate pH 5.2), B (acetonitrile) and C (water) were used with the following analysis conditions:Flow rate: (mL/min): 0.3Column T (° C.): 40Wavelength (nm): 270 (for elagolix), 210 (for the remaining compounds)Inj. vol. (μL): 1Acquisition time (min): 10Diluent: acetonitrile/water (1:1)Gradient: t (min)% A% B% C0525700.552570659057.559058525701052570
57,976
11858902
DETAILED DESCRIPTION For the purposes of promoting an understanding of the principles of the present disclosure, reference will now be made to embodiments illustrated in drawings, and specific language will be used to describe the same. It will nevertheless be understood that no limitation of the scope of this disclosure is thereby intended. In one embodiment, the present disclosure provides a compound of Formula I: or a stereoisomer thereof. In one embodiment, the present disclosure provides a composition, wherein the composition comprises the compound of Formula I. In one embodiment regarding the compound of Formula I, wherein the compound may comprise E and/or Z stereoisomer. In one embodiment, the present disclosure provides a method of using the compound of Formula I, wherein the compound is used as a metal-free primary explosive. In one embodiment regarding the method of using the compound of Formula I, wherein the compound of claim1has a thermal stability up to 227° C. and can undergo a deflagration to detonation transition (DDT) to initiate detonation of a larger body of secondary explosive. The present disclosure was influenced by the sensitivity issues represented by azo-coupled compounds such as compounds illustrated inFIG.1, with 1,1′-azobis(tetrazole) being able to undergo a DDT but too sensitive, and 1,1-azobis-1,2,3-triazole being too stable and not undergoing a DDT, precluding use. The present disclosure provides the study that examined the azo-coupling of 2-amino-4-nitro-1,2,3-triazole 1 and 1-amino-4-nitro-1,2,3-triazole 2 (Scheme 1). These compounds are intermediates between the insensitive 1,1′azobis-1,2,3-triazole and 1,1′-azobis(tetrazole) and had a potential to possess properties that would be ideal in a metal-free primary explosive: being sensitive enough to undergo a DDT, however also being thermally and mechanically stable. Precursor compounds 1 and 2 were synthesized according to literature methods. (See Wozniak, D. R. et al., Sensitive Energetics from the N-Amination of 4-Nitro-1,2,3-Triazole,ChemistryOpen2020, (9), 1-7, doi[dot]org/10[dot]1002/open[dot]202000053). In previous studies, there was success in procuring catenated nitrogen chains through coupling of N—NH2bonds with sodium dichloroisocyanurate; this same methodology was applied in this disclosure. The precursor compounds 1 and 2 were reacted separately in acetonitrile at 0° C. with an aqueous acidic solution of sodium dischloroisocyanurate (Scheme 1). The reactions yielded pure Example 1 (1,2-bis(4-nitro-2H-1,2,3-triazol-2-yl)diazene; or 2,2′-azobis(4-nitro-1,2,3-triazole)) and Example 2 (1,2-bis(4-nitro-1H-1,2,3-triazol-1-yl)diazene; or 1,1′-azobis(4-nitro-1,2,3-triazole)) in 71% and 61% yields respectively. No further purification was necessary.1H and13C NMR spectra were collected for both products. Example 1 and Example 2 had proton peaks present at 9.36 ppm and 10.10 ppm respectively. Example 1 carbon peaks were present at 155.12 ppm, 135.53 ppm. Example 2 carbon peaks were present at 153.02 ppm 129.60 ppm. When comparing the NMR spectra of precursor 1 to Example 1, it was observed that the C—H carbon shifts from 8.53 ppm in precursor compound 1. Both carbon peaks shifted downfield from 149.9 ppm and 129.83 ppm seen in the parent compounds. Colorless crystals of Example 1 and Example 2 suitable for x-ray crystallographic analysis were obtained from slow evaporation from acetone over several days at room temperature (FIG.2andFIG.3). Both products crystalize in orthorhombic crystal systems with high densities of 1.840 g cm−3and 1.818 g cm−3respectively. The length azo double bond Example 1 is 1.251 Å, which closely matches that of the N8compound mentioned previously. The azo double bonds in Example 2 are slightly shorter with lengths of 1.244 Å and 1.246 Å. Example 2 has a calculated heat of formation of 866.4 kJ mol−1, and an impressive calculated detonation velocity of 9014 m s−1. It was the more sensitive of the two isomers, with impact sensitivity of <1 J and friction sensitivity of <5 N. Example 1 has a heat of formation of 841.4 kJ mol−1but with its higher density was calculated to have a detonation velocity of 9068 m s−1. It is slightly more stable against mechanical stimuli compared to Example 2 with an impact sensitivity between 4.0-4.5 J and friction sensitivity between 36-40 N. This makes it a material with a detonation velocity approaching that of HMX (9193 m s−1) while possessing sensitivity to classify it as a primary explosive. Besides the slight differences in detonation performances and stability against mechanical stimuli, the differences in thermal stability of Example 1 and Example 2 is rather impressive. Example 2 is stable up to 160° C. where it detonates with enough force to decimate the aluminum pan, sample arm, and portions of the reference arm of the DSC/TGA apparatus (FIG.4). Due to its low thermal stability, Example 2, while powerful, is not a good candidate as a primary explosive. However, Example 1 is thermally stable up to an impressive 227° C. This high thermal stability makes this material an attractive candidate as a new metal-free primary explosive. It is quite surprise to find out that, although both Example 1 and Example 2 have the same number of nitrogen atoms, the more symmetric orientation of triazole nitrogen atoms to the azo group appears to make Example 1 unexpectedly suitable for a primary explosive. The combination of eight nitrogen atoms and the more symmetric orientation of Example 1 clearly has played some unexpected role and has made Example 1 unexpectedly suitable for a primary explosive. In order to be used as a primary explosive, energetic materials must be able to undergo a DDT. To test this, very small amounts of Example 1 and Example 2 were placed on the tip of a metal spatula and held to a blow-torch. In open air, Example 2 decomposes producing a yellow flame and sharp snap. Example 1 only produces a yellow flame and hissing noise in open air, however with slight confinement such as a single layer of aluminum foil, it produces a loud snapping sound when ignited. This indicates that in sufficiently confined spaces, Example 1 can undergo a DTT and is a viable candidate as a primary explosive. In conclusion, this disclosure provides the synthesis and characterization of two new energetic azo compounds, Example 1 and Example 2. Due to the sensitivity and high thermal stability of Example 1, combined with its excellent detonation velocity and ability to undergo a DDT, this compound is a promising candidate for use as a metal-free primary explosive. To the best of our knowledge, it possesses the most appropriate properties to function in this role where its high thermal stability, reasonable mechanical stability, and ability to undergo a DDT in small amounts make it more suitable than currently known metal-free primary explosives. Those skilled in the art will recognize that numerous modifications can be made to the specific implementations described above. The implementations should not be limited to the particular limitations described. Other implementations may be possible.
7,169
11858903
DETAILED DESCRIPTION FIG.1illustrates a melamine offgas quencher1which receives: melamine-containing offgas via line2; liquid ammonia via line3; an aqueous solvent via line4; cold gaseous nitrogen via line5. The melamine-containing offgas in line2come from the synthesis section of a melamine plant, e.g. a non-catalytic high-pressure melamine synthesis section. The liquid ammonia may be introduced via one or more sprays. For example the lines31and32inFIG.1denote different ammonia sprayers fed by the main header3. The aqueous solvent of line4may be demineralized water or recycled water or recycled mother liquor from the melamine plant. It may contain traces of ammonia and/or CO2. The cold gaseous nitrogen5, which is an optional, is colder and therefore denser than the washed offgas. As illustrated, liquid ammonia is introduced above the inlet of the offgas. The cold nitrogen of line5is introduced below the offgas inlet and the aqueous solvent is introduced below the nitrogen inlet at the bottom of the quencher1. Due to the location of the introduction points of the above described streams, the quencher1operates basically as a two-zone equipment. The upper zone10operates in a gaseous phase. The offgas entering at line2travel upward and contact the liquid ammonia sprayed at lines31,32in a counter-current regime. As a consequence of this, the melamine contained in the offgas is solidified and precipitates; melamine-free anhydrous offgas are extracted from top of the quencher1at line6. Optionally, the washed offgas are mixed with a CO2stream7. The resulting stream8is sent to a tied-in urea plant for recycle, e.g. via offgas condensation and recycle of the so obtained carbamate-containing solution. The lower zone11operates in a liquid phase. The solid melamine removed from the offgas is dissolved in the aqueous medium and partially fills the bottom of the quencher1forming a liquid level12. As illustrated, in operation the input line4of the aqueous solvent remains preferably below the liquid level12, whilst the nitrogen line5is above the liquid level12. The melamine-containing solution is removed via line9for further processing. The cold nitrogen, due to its density, tends to form a layer just above the liquid level12, which separates the zones10and11, particularly to avoid that gaseous CO2passes into the liquid phase. In some embodiments, the lower portion of the quencher1(substantially corresponding to the zone11) may be of a reduced diameter. This melamine solution at line9can be sent to a downstream equipment for further purification. Before purification, the melamine solution withdrawn for the quencher1may be mixed with a melamine solution obtained from a step of melamine melt quenching, or the solution at line9may be sent directly to a step of filtration/crystallization but without the need of a dedicated offgas stripping stage. Example 1 Reference is made to a high-pressure melamine plant with a capacity of 40000 tons/year wherein the offgas are released from the melamine synthesis section at 380° C. and 80 barg (bar gauge). A total of 13.0 tons/hour (t/h) of offgas are released, including 6.6 t/h of NH3, 5.9 t/h of CO2and 0.5 t/h of melamine. Said offgas is washed with ammonia introduced in liquid state at 20° C. and 80 barg. The offgas is cooled down to 169° C. The operating pressure of the quencher is 25 barg. The ammonia required for cooling down the offgas is 3.3 t/h. The offgas obtained from the quencher at 169° C. and 25 barg is free of melamine and contains 9.9 t/h of ammonia and 5.9 t/h of CO2. At the bottom of the quencher, 4.5 t/h of water or water recycling solution at 140° C. are fed in order to obtain 5 t/h of a melamine solution containing 10% by weight of melamine. The solution is free or substantially free of dissolved offgas. Example 2 This invention can be carried out even in a more advantageous embodiment by increasing the temperature of the washed offgas considering that the need of keeping a low-enough temperature in the offgas quencher (in order to minimize the water content in the washed offgas) is set aside. Increasing the washed offgas temperature decreases the amount of required quenching ammonia. The offgas stream of example 1 is washed with ammonia in liquid state at 20° C. and 80 barg and cooled to 250° C. The operating pressure of the quencher is 40 barg. The ammonia required for cooling down the offgas is 2.0 t/h. The offgas obtained from the quencher at 250° C. and 40 barg is free of melamine and contains 8.6 t/h of ammonia and 5.9 t/h of CO2. At the bottom of the quencher, 2.2 t/h of water or water recycling solution at 170° C. are fed in order to obtain 2.7 t/h of a solution containing 18.5% by weight of melamine, which is free or substantially free of dissolved offgas.
4,818
11858904
DETAILED DESCRIPTION An improved process to produce large isoxazoline compound particles which comprises initiating crystallization and then maintaining the temperature of the crystallization in the metastable region by removing, reheating and recycling a portion of the solvent thereby allowing the existing crystals to grow larger while minimizing the formation of newer smaller crystals. Crystallization is initiated by nucleation, which happens either spontaneously or is induced by vibration or seed particles. Nucleated crystals are small crystals formed when there is a drop in the temperature of a saturated solution. If nucleation sets in too quickly, too many too small crystals will grow. In the case of isoxazoline compounds and fluralaner in particular, the seed crystals are typically less than 10 μm length. The process of crystallization starts with the addition of nucleated material (seed crystals) to a solution of isoxazoline compound in solution to achieve surface properties of the starting crystals that are amenable to growth. As crystallization is initiated, a slurry of isoxazoline compound particles in the solvent is formed. This initial slurry is kept at a relatively high temperature (52-54° C.) to facilitate reasonable growth rates and avoid further nucleation. At lower temperatures, growth rates are significantly slower, and the risk of nucleation is greater. A portion of the batch of isoxazoline compound particle slurry is removed, heated to dissolve any crystals that have formed and returned to the crystallizer to provide continuous supersaturation to drive crystal growth. This recycle rate cannot be too low slow since under these growth conditions thin plates are preferentially formed, which are susceptible to breakage. The recycle rate cannot be too high, since under these conditions either nucleation, or aggregation can occur. Once the starting slurry has grown to a sufficient point, the slurry is cooled at a rate that avoids nucleation to a temperature where the desired crystal dimensions are achieved. The return of the dissolved isoxazoline compound solution to the crystallizer vessel is conducted at rate of approximately 0.25 to 0.75 batch volumes per hour to achieve continuous crystal growth of the isoxazoline compound particles. After sufficient particle size growth is achieved from the repeated removal of slurry material from and return of dissolved isoxazoline to the crystallizer, the crystallizer is cooled to about 0° C., preferable about −10° C. over 10-48 hours, preferably 12-20 hours to further relieve supersaturation and achieve growth to the desired dimensions. It has been found that injectable compositions comprising particles of isoxazoline compounds with a defined particle size produced by the inventive process show desirable bioavailability and duration of efficacy, while causing minimal irritation at the injection site. Such compositions also provide desirable safety profiles toward the warm-blooded and bird animal recipients. In addition, it has been discovered that a single administration of such compositions generally provides potent activity against one or more parasites (e.g., ectoparasites, e.g. fleas, ticks or mites), while also tending to provide fast onset of activity, long duration of activity, and/or desirable safety profiles. Definitions Scanning electron microscopy (SEM) is an analytical instrument that uses a focused beam of high-energy electrons to generate a variety of signals at the surface of solid specimens. The signals reveal information about the sample including external morphology (texture), chemical composition, and crystalline structure and orientation of materials making up the sample. Solvent with temperature dependent solubility for the solute means that the solubility of the solute in the solvent varies with temperature. Generally, this means the solubility increases with increased temperature. Temperature sensitivity of fluralaner solubility in isopropanol (IPA) is shown inFIG.1with the x-axis showing temperature and the y-axis showing the solubility of fluralaner in expressed in mg/mL. The meta-stable region of the solubility temperature curve is the region where existing crystals will grow, but no new crystals are formed. Crystallizer vessel is a vessel in which crystallization occurs. Saturation is the state of a solution when it holds the maximum equilibrium quantity of dissolved matter at a given temperature. Supersaturation is when a solution contains more solute than the saturated solution at equilibrium. Slurry is a thin suspension. Batch is the solvent plus solute. Batch volume is the volume of the batch. As used herein, particle size data reported are volume weighted as measured by conventional particle techniques well known to those skilled in the art, such as static light scattering (also known as laser diffraction), image analysis or sieving. More discussion of particle size measurement is provided below. Mechanical resiliency is the resistance of crystals or particles to break into smaller crystals or particles when exposed to pressure or stress from other sources. Mechanical resiliency can be measured by a pressure titration using the Sympatec HELOS. This instrument can simultaneously measure the particle size distribution. In this experiment, pressure is applied to the crystals to disperse or separate them one from another. The change in the particle size distribution measurement of d50 is monitored as the pressure on the crystals is increased from 1 bar to 3 bars. Preferably, the isoxazoline compound particles of the subject invention will not decrease their particle size distribution measurement of d50 by more than 30-40% when the dispersion pressure is increased from 1 to 3 bar. In an embodiment of an isoxazoline for use in the invention, T is selected from wherein in T-1, T-3 and T-4, the radical Y=hydrogen, halogen, methyl, halomethyl, ethyl, or haloethyl. In an embodiment of an isoxazoline for use in the invention, Q is selected from wherein R3, R4, X and ZAare as defined above, and ZB= ZD= In an embodiment an isoxazoline for use in the invention is as presented in Table 1. TABLE 1(R1)nR2R3R4TYQZX3-Cl, 5-ClCF3CH2CF3HT-2—Q-1—CO3-Cl, 5-ClCF3CH2CH3HT-2—Q-1—CO3-Cl, 5-ClCF3CH2CH2OCH3HT-2—Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-2—Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CH3HT-2—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3HT-2—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CH3HT-2—Q-1—CO3-CF3, 5-ClCF3CH2C(O)NHCH2CF3HT-2—Q-1—CO3-CF3, 5-ClCF3CH2C(O)NHCH2CH3HT-2—Q-1—CO3-Cl, 5-ClCF3—T-2—Q-6ZB-7CO3-Cl, 5-ClCF3——T-2—Q-7ZB-7CO3-Cl, 5-ClCF3——T-2—Q-5ZB-7CO3-Cl, 5-ClCF3——T-2—Q-2ZD-1CO3-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-3CH3Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CCHT-3CH3Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CNHT-3CH3Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CH3HT-3CH3Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3HT-3CH3Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CH3HT-3CH3Q-1—CO3-Cl, 4-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-3CH3Q-1—CO3-Cl, 4-Cl, 5-ClCF3CH2C(O)NHCH2CH3HT-3CH3Q-1—CO3-Cl, 4-F, 5-ClCF3CH2C(O)NHCH2CF3HT-3CH3Q-1—CO3-Cl, 4-F, 5-ClCF3CH2C(O)NHCH2CH3HT-3CH3Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-20—Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CH3HT-20—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3CH3T-20—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CH3CH3T-20—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3HT-20—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CH3HT-20—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3HT-21—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CH3HT-21—Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-21—Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CH3HT-21—Q-1—CO3-Cl, 5-ClCF3CH2CH2SCH3HT-21—Q-1—CO3-Cl, 4-Cl, 5-ClCF3C(O)CH3HT-22FQ-1—CH23-Cl, 4-Cl, 5-ClCF3C(O)CH(CH3)2HT-22FQ-1—CH23-Cl, 4-Cl, 5-ClCF3C(O)-cyclo-propylHT-22FQ-1—CH23-Cl, 4-F, 5-ClCF3C(O)CH3HT-22FQ-1—CH23-Cl, 4-Cl, 5-ClCF3C(O)CH2CH3HT-22FQ-1—CH23-Cl, 4-F, 5-ClCF3C(O)CH3HT-22ClQ-1—CH23-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-1CH3Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CH3HT-1CH3Q-1—CO3-Cl, 5-ClCF3R3-1 (Z)HT-1CH3Q-1—CO3-Cl, 5-ClCF3R3-1 (E)HT-1CH3Q-1—CO In an embodiment an isoxazoline for use in the invention is as presented in Table 2. TABLE 2(R1)nR2R3R4TYQZX3-Cl, 5-ClCF3CH2CF3HT-2—Q-1—CO3-Cl, 5-ClCF3CH2CH3HT-2—Q-1—CO3-Cl, 5-ClCF3CH2CH2OCH3HT-2—Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-2—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3HT-2—Q-1—CO3-CF3, 5-ClCF3CH2C(O)NHCH2CF3HT-2—Q-1—CO3-Cl, 5-ClCF3—T-2—Q-6ZB-73-Cl, 5-ClCF3——T-2—Q-7ZB-73-Cl, 5-ClCF3——T-2—Q-5ZB-73-Cl, 5-ClCF3——T-2—Q-2ZD-13-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-3CH3Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CCHT-3CH3Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CNHT-3CH3Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3HT-3CH3Q-1—CO3-Cl, 4-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-3CH3Q-1—CO3-Cl, 4-F, 5-ClCF3CH2C(O)NHCH2CF3HT-3CH3Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-20—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3CH3T-20—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3HT-20—Q-1—CO3-CF3, 5-CF3CF3CH2C(O)NHCH2CF3HT-21—Q-1—CO3-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-21—Q-1—CO3-Cl, 5-ClCF3CH2CH2SCH3HT-21—Q-1—CO3-Cl, 4-Cl, 5-ClCF3C(O)CH3HT-22FQ-1—CH23-Cl, 4-Cl, 5-ClCF3C(O)CH(CH3)2HT-22FQ-1—CH23-Cl, 4-Cl, 5-ClCF3C(O)-cyclo-propylHT-22FQ-1—CH23-Cl, 4-F, 5-ClCF3C(O)CH3HT-22FQ-1—CH23-Cl, 4-Cl, 5-ClCF3C(O)CH2CH3HT-22FQ-1—CH23-Cl, 4-F, 5-ClCF3C(O)CH3HT-22ClQ-1—CH23-Cl, 5-ClCF3CH2C(O)NHCH2CF3HT-1CH3Q-1—CO3-Cl, 5-ClCF3R3-1 (Z)HT-1CH3Q-1—CO3-Cl, 5-ClCF3R3-1 (E)HT-1CH3Q-1—CO In an embodiment an isoxazoline for use in the invention is the compound: wherein R1a, R1b, R1care independently from each other: hydrogen, Cl or CF3. Preferably R1aand R1care Cl or CF3, and R1bis hydrogen, T is wherein Y is methyl, bromine, Cl, F, CN or C(S)NH2; n=1 or 2; and Q is as described above. In an embodiment of an isoxazoline as defined herein, R3is H, and R4is: —CH2—C(O)—NH—CH2—CF3, —CH2—C(O)—NH—CH2—CH3, —CH2—CH2—CF3or —CH2—CF3. The isoxazoline for use in the invention also includes pharmaceutically acceptable salts, esters, and/or N-oxides thereof. In addition, the reference to an isoxazoline compound refers equally to any of its polymorphic forms or stereoisomers. With respect to stereospecific forms, the pharmaceutical composition according to the invention may employ a racemic mixture of an isoxazoline for use in the invention, containing equal amounts of the enantiomers of such isoxazoline compound as described above. Alternatively, the pharmaceutical composition may use isoxazoline compounds that contain enriched stereoisomers compared to the racemic mixture in one of the enantiomers of the isoxazoline as defined herein. Also, the pharmaceutical composition may use an essentially pure stereoisomer of such isoxazoline compounds. Such enriched- or purified stereoisomer preparations of an isoxazoline for use in the invention, may be prepared by methods known in the art. Examples are chemical processes utilizing catalytic asymmetric synthesis, or the separation of diastereomeric salts (see e.g.: WO 2009/063910, and JP 2011/051977, respectively). In an embodiment of the pharmaceutical composition according to the invention, the isoxazoline is one or more selected from the group consisting of fluralaner, afoxolaner, lotilaner or sarolaner. In one embodiment the compound of Formula (I) is 4-[5-(3,5-Dichlorophenyl)-5-trifluoromethyl-4,5-dihydro isoxazol-3-yl]-2-methyl-N-[(2,2,2-trifluoro-ethylcarbamoyl)-methyl]-benzamide (CAS RN 864731-61-3-USAN fluralaner). In an embodiment, the fluralaner is S-fluralaner. In another embodiment the compound of Formula (I) is 4-[5-[3-Chloro-5-(trifluoromethyl)phenyl]-4,5-dihydro-5-(trifluoromethyl)-3-isoxazolyl]-N-[2-oxo-2-[(2,2,2-trifluoroethyl)amino]ethyl]-1-naphthalenecarboxamide (CAS RN 1093861-60-9, USAN-afoxolaner) that was disclosed in WO2007/079162. In an embodiment of the pharmaceutical composition according to the invention the isoxazoline is lotilaner (CAS RN: 1369852-71-0; 3-methyl-N-[2-oxo-2-(2,2,2-trifluoroethylamino)ethyl]-5-[(5S)-5-(3,4,5-trichlorophenyl)-5-(trifluoromethyl)-4H-1,2-oxazol-3-yl]thiophene-2-carboxamide). In an embodiment of the pharmaceutical composition according to the invention the isoxazoline is sarolaner (CAS RN: 1398609-39-6; 1-(5′-((5S)-5-(3,5-dichloro-4-fluorophenyl)-5-(trifluoromethyl)-4,5-dihydroisoxazol-3-yl)-3′-H-spiro(azetidine-3,1′-(2) benzofuran)-1-yl)-2-(methylsulfonyl) ethanone). In another embodiment, the compound of Formula (I) is (Z)-4-[5-(3,5-Dichlorophenyl)-5-trifluoromethyl-4,5-dihydroisoxazol-3-yl]-N-[(methoxyimino)methyl]-2-methylbenzamide (CAS RN 928789-76-8). In another embodiment the compound of Formula (I) is 4-[5-(3,5-dichlorophenyl)-5-(trifluoromethyl)-4H-isoxazol-3-yl]-2-methyl-N-(thietan-3-yl)benzamide (CAS RN 1164267-94-0) that was disclosed in WO2009/0080250. In an embodiment, the compound according to the invention is 5-[5-(3,5-Dichlorophenyl)-4,5-dihydro-5-(trifluoromethyl)-3-isoxazolyl]-3-methyl-N-[2-oxo-2-[(2,2,2-trifluoroethyl)amino]ethyl]-2-thiophenecarboxamide (CAS RN: 1231754-09-8), which was disclosed in WO 2010/070068. An embodiment of the invention is a process for preparing isoxazoline compound particles wherein the isoxazoline compound is a compound of Formula (I) comprisinga) Combining an isoxazoline compound in a crystallizer vessel with a solvent which has a temperature dependent solubility of the isoxazoline compound;b) Heating the crystallizer vessel until the isoxazoline compound is dissolved in the solvent;c) Cooling the crystallizer vessel to 48-55° C. to form a batch of supersaturated isoxazoline compound in the solvent;i) adding crystalline seed of the isoxazoline compound to the crystallizer vessel to initiate crystallization and particle growth;ii) Forming a slurry of isoxazoline compound particles and solvent in the crystallizer vessel;d) Maintaining the temperature of the crystallizer vessel to 48-55° C.;e) Removing a portion of the batch and heating the removed portion to fully dissolve the isoxazoline compound particles in the solvent; wherein the rate of removal is at a rate of approximately 0.25 to 0.75 batch volumes per hour; and wherein the batch volume is the volume of the supersaturated isoxazoline compound solution created in step c);f) Returning the dissolved isoxazoline compound solution to the crystallizer vessel; wherein the rate of return is equal to the rate of removal of step e); andg) Cooling the crystallizer vessel to achieve isoxazoline compound particles of the desired dimensions; wherein the desired particle dimensions are particles having a volume weighted particle size distribution (d50) as measured by a light scattering instrument of between 75 and 120 μm and an average particle thickness greater than 10 μm, preferably greater than 20 μm. In an embodiment, the isoxazoline compound is fluralaner. In an embodiment, the solvent is methanol or acetone. In yet another embodiment, the solvent is an acetate or acetonitrile. In an embodiment, the solvent is selected from the group of dimethyl acetamide (DMA), N-methylpyrrolidone (NMP), dimethyl sulfoxide (DMSO), N,N-diethy-m-toluamide (DEET), 2-pyrrolidone, acetone, g-hexalactone, glycofurol (tetraglycol), methyl ethyl ketone, diethylene glycol monoethyl ether (Transcutol®), ethyl lactate, dimethylisosorbide, ethyl acetate, macrogol glycerol caprylcaprate (Labrasol®), dipropylene glycol monomethyl ether (Dowanol™ DPM), glycerol formal, benzyl alcohol, methanol, polyethylene glycol 200, propylene carbonate, 1-methoxy-2-propyl acetate (Dowanol™ PMA), isopropylidene glycerol (solketal), ethyl alcohol, glycerol triacetate (triacetin), isopropyl alcohol, propylene glycol, triglycerides medium chain (Miglyol® 812), ethyl oleate, toluene, ethyl acetate or mixtures thereof. In an embodiment, the solvent is isopropanol. In an embodiment, the solvent is a mixture of toluene and ethyl acetate. In an embodiment, the crystallizer of step b is heated to a temperature greater than 60° C., preferably about 65° C. In an embodiment, in step c) the crystallizer vessel is cooled to achieve supersaturation, preferably to a temperature of about 48-55° C., more preferably to a temperature of about 52-54° C. In an embodiment, the removed portion is heated to a temperature greater than 60° C., preferably about 65° C. In an embodiment, the removed portion is heated via a heat exchanger or in a second vessel. In an embodiment, the rate of removal in step e) is 0.40 to 0.46 batch volumes per hour. In an embodiment, the rate of removal is maintained for about 4 to 24 hours, preferably about 6 hours. In an embodiment, the crystallizer vessel of step g) is cooled to a temperature of around 0° C. or less, preferably about −10° C. An additional embodiment of any of the above processes, further comprising a step of filtering the isoxazoline compound particles of step g). In an embodiment, the temperature of the filtering is maintained at a temperature of 0° C. or less, preferably at −10° C. In an embodiment, the filtered isoxazoline particles are dried. Embodiments of the invention are the isoxazoline compound particles produced by any of processes disclosed herein. An embodiment of the invention is a isoxazoline compound particle composition comprising particles with a thickness of greater than 10 μm, preferably greater than 20 μm as measured by scanning electron microscopy (SEM), and a mechanical resiliency as measured by a pressure titration using the Sympatec HELOS, wherein the particle size distribution (d50) of the particles does not decrease by more than 40% from 1 to 3 bar dispersion pressure. In an embodiment, the particle size distribution (d50) of the particles does not decrease by more than 35% from 1 to 3 bar dispersion pressure. In an embodiment, the particle size distribution (d50) of the particles does not decrease by more than 30% from 1 to 3 bar dispersion pressure. In an embodiment, the isoxazoline compound particle composition comprising particles with a thickness of greater than 10 μm but less than 100 μm, preferably greater than 20 μm but less than 90 μm, preferably greater than 30 μm but less than 80 μm as measured by scanning electron microscopy (SEM). In an embodiment, the isoxazoline compound particle composition comprising particles with a thickness of greater than 10 μm, preferably greater than 20 μm. In an embodiment, the isoxazoline compound has a particle size distribution of D50 as measured by a static light scattering instrument of from about 25 microns to about 250 microns, particle size of from about 11 microns to about 250 microns, particle size of from about 50 microns to about 150 microns, particle size of from about 75 microns to about 125 microns, particle size of from about 75 microns to about 150 microns, particle size of from about 90 microns to about 110 microns or a particle size of from about 30 microns to about 100 microns. Particle size distribution describes the relative amount of particles present according to size. D10 is a particle size distribution that expresses the size that 10% of the particles are smaller than. D50 is a particle size measurement distribution that expresses the size that 50% of the particles are smaller than. D90 is a particle size measurement distribution that expresses the size that 90% of the particles are smaller than. In a particular embodiment, the D10 of particle size is about 10 μm, about 20 μm, about 30 μm, about 40 μm, about 50 μm, about 60 μm, or about 80 μm. In a particular embodiment, the D50 of particle size is about 50 μm, about 75 μm, about 80 μm, about 90 μm, about 100 μm, about 110 μm, about 120 μm, about 130 μm about 140 μm or about 150 μm. In a particular embodiment, the D90 of particle size is about 100 μm, about 130 μm, about 150 μm, about 175 μm, about 200 μm, or about 250 μm. In a particular embodiment, the D10 of the particle size is about 20 to 35 μm, the D50 of the particle size is about 90 to 105 μm and the D90 of the particle size is about 155 to 175 μm. In a particular embodiment, the D10 of the particle size is about 25 to 30 μm, the D50 of the particle size is about 95 to 100 μm and the D90 of the particle size is about 160 to 170 μm. In a particular embodiment, the D10 of the particle size is about 10 to 20 μm, the D50 of the particle size is about 85 to 110 μm and the D90 of the particle size is about 170 to 185 μm. In a particular embodiment, the D10 of the particle size is about 10 to 15 μm, the D50 of the particle size is about 95 to 105 μm and the D90 of the particle size is about 175 to 180 μm. In a particular embodiment, the D10 of the particle size is about 10 to 25 μm, the D50 of the particle size is about 40 to 60 μm and the D90 of the particle size is about 95 to 100 μm. In a particular embodiment, the D10 of the particle size is about 15 to 20 μm, the D50 of the particle size is about 45 to 55 μm and the D90 of the particle size is about 90 to 95 μm. In a particular embodiment, the D10 of the particle size is about 30 to 50 μm and the D50 of the particle size is about 70 to 130 μm. In a particular embodiment, the D10 of the particle size is about 35 to 45 μm and the D50 of the particle size is about 90 to 110 μm. In a particular embodiment, the D10 of the particle size is about 40 μm and the D50 of the particle size is about 100 μm. The volume weighted particle size can be measured by sieving, microscopy or laser diffraction (Malvern or Sympatec) The volume weighted particle size measurement can be performed with a Malvern Mastersizer 2000 with the Hydro 2000G measuring cell, or with a Horiba LA-910 laser scattering particle size distribution analyzer. The volume weighted particle size can be measured by a Sympatec Helos instrument. In an embodiment, the isoxazonline compound is fluralaner. EXAMPLES Example 1—Process to Form Large Particle Size Fluralaner Fluralaner was added at a concentration of 100 mg/mL in IPA, with 60 g added to 600 mL of isopropanol. This composition was heated to 65° C. over 1 hour, and aged for one hour to ensure full dissolution. The solution was cooled over 20 minutes to 50° C. and seeded with 0.6 g of crystalline fluralaner seed. The batch was further cooled to 20° C. over two hours to establish the starting particles. The batch was heated to 54° C., at which point a stream of the batch was removed and heated to an elevated temperature until fully dissolved (>65° C.). The removal rate and return rate to the crystallizer were set to approximately 4.4-4.8 mL/min. The recycle loop continued for 6 hours, at which point the ×50 particle size dimension is approximately 40 μm. The batch was aged at 54° C. for 6 hours to further relieve supersaturation, then cooled to 45° C. over 6 hours, and further cooled to 0° C. over 16 hours. SeeFIG.3for a schematic of the process equipment. The resultant slurry was filtered and dried to produce fluralaner particles. The dried fluralaner particles were measured to determine the particle dimensions and mechanical resiliency. Example 2—Determination of the Particle Size and Mechanical Resiliency of the Fluralaner Particles The volume weighted particle size of the fluralaner crystals was measured by laser diffraction (Sympatec Helos) to determine the particle size distribution. The mechanical resiliency was also determined during a pressure titration experiment.FIG.4shows the particle size distribution fluralaner crystals not produced by the inventive process. In this case, this material is the product of the previous commercial process using an unseeded, distallative crystallization process from an ethyl acetate, toluene solvent system. Of note is the lower particle size and general wider distribution of sizes of the particles. The fragile nature of particles not representative of the inventive process is demonstrated inFIG.5, which shows the results of the pressure titration experiment. In this experiment, the particle size distribution was monitored as the particles are exposed to increasing pressure from 1 bar to 3 bar.FIG.5shows that as the pressure increased, the median particle size (d50) decreases from 110 μm to 60 μm, a loss of around 45%. Moreover, the particle size distribution curve broadens and shifts towards smaller particle sizes. This is evidence that these particles are being broken under the increased pressure and is an indication that the particles were very thin. In contrast,FIG.10shows the particle size distribution of the fluralaner crystals produced by the inventive process. These particles have a larger d50 than the particles ofFIG.5. Furthermore, in the pressure titration test, for the particles produced by the inventive process, the d50 was reduced by only around 25% of the original value. This is an indication of the increased mechanical resiliency of these particles. Also of note is the fact that under the elevated pressures, the distribution does not broaden in the same fashion as the particles from the unoptimized process shown inFIG.5. FIG.7shows the particle size distribution and pressure titration of an additional batch of fluralaner particles that were produced by the inventive process. In this case, the original d50 of around 103 μm was reduced to around 67 μm, a loss of around 35%. FIG.8is a scanning electron microscopy image of fluralaner particles that were not produced the inventive process. Of note is that these crystals are rather thin. FIG.9is a scanning electron microscopy image of fluralaner particles that were produce by the inventive process. In contrast to the particles shown inFIG.8, these particles are large (around 100 μm) and thick (around 10-20 μm). Example 3:—Process to Form Large Particle Size Fluralaner at the 6 L Scale Fluralaner was added at a concentration of 100 mg/mL in isopropanol (IPA), with 600 g added to 6 L of isopropanol. This composition was heated to 65° C. over 1 hour, and aged for one hour to ensure full dissolution. The solution was cooled over 20 minutes to 50° C. and seeded with 6 g of crystalline fluralaner seed, in this instance unmilled seed with an d50 of approximately 10 μm. The batch was further cooled to 20° C. over two hours to establish the starting particles. The batch was heated to 54° C., at which point 1.2 L of the batch was removed and heated to an elevated temperature until all solids were fully dissolved (>65° C.). A recirculation loop was then started, with the removal rate and return rate to the crystallizer set to approximately 44-48 mL/min. The recycle loop continued for 3 hours, at which point the d50 particle size dimension is approximately 45 μm. The batch was aged at 54° C. for 6 hours to further relieve supersaturation, then cooled to 45° C. over 6 hours, and further cooled to 0° C. over 16 hours. The resultant slurry was filtered and dried to produce fluralaner particles. The dried fluralaner particles were measured to determine the particle dimensions and inform mechanical resiliency of the particles, and shown inFIG.10. SeeFIG.11for an SEM image of the resulting particles. Example 4:—Process to Form Large Particle Size Fluralaner at the Pilot Scale Fluralaner was added at a concentration of 100 mg/mL in IPA, with 60 kg added to 600 L of isopropanol. This composition was heated to 65° C. over 1 hour, and aged for one hour to ensure full dissolution. The solution was cooled over 20 minutes to 50° C. and seeded with 600 g of crystalline fluralaner seed, again with unmilled seed crystals having an d50 of approximately 10 μm. The batch was further cooled to 20° C. over two hours to establish the starting particles. The batch was heated to 54° C., at which point 120 L of the batch was removed and heated to an elevated temperature until fully dissolved (>65° C.). The removal rate and return rate to the crystallizer were set to approximately 4.4-4.8 L/min. The recycle loop continued for 2.75 hours, at which point the d50 particle size dimension is approximately 40 μm. The batch was aged at 54° C. for 6 hours to further relieve supersaturation, then cooled to 45° C. over 6 hours, and further cooled to 0° C. over 16 hours. SeeFIG.12for a schematic of the process equipment. The resultant slurry was filtered and dried to produce fluralaner particles. Agitation was limited during the filtration and drying. The material was delumped at low speed in a conical mill. The dried fluralaner particles were measured to determine the particle dimensions and mechanical resiliency. SeeFIG.13for the particle size distribution and mechanical resiliency. SeeFIG.14for an SEM image of the resulting particles. Example 5:—Process to Form Large Particle Size Fluralaner from an Alternative Solvent System Fluralaner was added at a concentration of 100 mg/mL in 5:3 (volume basis) of Toluene:Ethyl Acetate, with 60 g added to 600 mL of solvent. This composition was heated to 65° C. over 1 hour, and aged for one hour to ensure full dissolution. The solution was cooled over 20 minutes to 50° C. and seeded with 0.6 g of crystalline fluralaner seed, again with unmilled seed crystals having an d50 of approximately 10 μm. The batch was further cooled to 20° C. over two hours to establish the starting particles. The batch was heated to 54° C., at which point 120 mL of the batch was removed and heated to an elevated temperature until fully dissolved (>65° C.). The removal rate and return rate to the crystallizer were set to approximately 4.3 mL/min. The recycle loop continued for 2.2 hours, at which point the ×50 particle size dimension is approximately 50 μm. The batch was aged at 54° C. for 5 hours to further relieve supersaturation, then cooled to 45° C. over 6 hours, and further cooled to 0° C. over 16 hours. The dried fluralaner particles were measured to determine the particle dimensions and mechanical resiliency. SeeFIG.15for the particle size distribution and mechanical resiliency as measured with the pressure titration on the Sympatec static light scattering system. SeeFIG.16for an SEM image of the resulting particles. These results show that using the recirculation process, the target particle size and mechanical resiliency can be achieved. It should be noted that the solvent may impact the morphology, as is observed inFIG.16, where the surfaces of the crystals are slightly modified from the surfaces of crystals grown from isopropanol.
30,172
11858905
DETAILED DESCRIPTION The following description is presented to enable any person skilled in the art to make and use the present disclosure and is provided in the context of a particular application and its requirements. Various modifications to the disclosed embodiments will be readily apparent to those skilled in the art, and the general principles defined herein may be applied to other embodiments and applications without departing from the spirit and scope of the present disclosure. Thus, the present disclosure is not limited to the embodiments shown but is to be accorded the widest scope consistent with the claims. The terminology used herein is to describe particular example embodiments only and is not intended to be limiting. As used herein, the singular forms “a,” “an,” and “the” may be intended to include the plural forms as well, unless the context clearly indicates otherwise. It will be further understood that the terms “comprises,” “comprising,” “includes,” and/or “including” when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. These and other features, and characteristics of the present disclosure, as well as the methods of operation and functions of the related elements of structure and the combination of parts and economies of manufacture, may become more apparent upon consideration of the following description with reference to the accompanying drawing(s), all of which form a part of this specification. It is to be expressly understood, however, that the drawing(s) is for the purpose of illustration and description only and are not intended to limit the scope of the present disclosure. It is understood that the drawings are not to scale. According to an aspect of the present disclosure, a plurality of compounds are provided. The compounds are capable of inhibiting CatL and may be used for treating or preventing a CatL-related disease in a subject. As used herein, the term “inhibiting CatL” refers to decreasing the activity of CatL and/or the content of CatL in a local part (e.g., in vitro and/or in vivo). These compounds provided by the present disclosure are referred to as “Compounds” herein for convenience. In some embodiments, the Compounds may be represented by formula (I-a): In some embodiments, in formula (I-a), W may be CO or SO2. When W is CO, the Compounds may be represented by formula (I-b): When W is SO2, the Compounds may be represented by formula (I-c): In some embodiments, R1and R2may be independently selected from H, a —CH2-group, and an alkyl group. In some embodiments, R1and R2may be unconnected or connected via a single bond. In some embodiments, R3may be an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, or a heterocyclic group, and R3may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. In some embodiments, X1 may be a CH group or N. In some embodiments, X2may be O, S, or N—R4, and R4may be selected from H, an alkyl group, an aryl group, and a heterocyclic group. In some embodiments, X3may be H, an alkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, or a heterocyclic group, and X3may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, and an alkoxy group. In some embodiments, Z1may be a CH group, C—R5, or N. In some embodiments, Z2may be a CH group, C—R6, or N. In some embodiments, Z3may be a CH group, C—R7, or N. In some embodiments, Z4may be a CH group, C—R8, or N. In some embodiments, R5-R8may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, —CN, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and each of R5-R8may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, a heterocyclic group, a cycloalkyl group, an aryl group, and an alkoxy group. In some embodiments, the Compounds may be represented by formula (II): In some embodiments, the Compounds may be represented by formula (III): In some embodiments, the Compounds may be represented by formula (IV): In some embodiments, R0may be H, an alkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, or a heterocyclic group, and R0may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, and an alkoxy group. In some embodiments, the Compounds may be represented by formula (V): In some embodiments, at least one of R5-R8is H. For example, R5-R8are H. In some embodiments, at least one of R5-R8is halogen or —CN, and the other of R5-R8are H. In some embodiments, at least one of R5-R8is a pyrazole group, which is optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and the other of R5-R8are H. For example, one of R5-R8is a group represented by formula (VI-a): In some embodiments, R9may be H, an alkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, or a heterocyclic group. In some embodiments, R10and R11may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group. Formula (VI-a) may be connected with the benzoxazole group shown in formula (II), formula (III), formula (IV), or formula (V) at methyl. In such cases, the Compounds may be represented by, for example, formula (II-a), formula (II-b), formula (II-c), or formula (II-d): In some embodiments, R5-R10are H, and the Compounds may be represented by, for example, formula (II-a1): In some embodiments, at least one of R5-R8is a cycloalkyl group or a heterocyclic group, which is optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and the other of R5-R8are H. For example, one of R5-R8may be a group represented by formula (VI-b): In some embodiments, X4may be S, O, SO2, N, C, or C-L1. L1may be selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and L1may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. In some embodiments, X5may be N or C. In some embodiments, R12does not exist, or may be selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. In some embodiments, R13-R16may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and each of R13-R16may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. Formula (VI-b) may be connected with the benzoxazole group shown in formula (II), formula (III), formula (IV), or formula (V) at X5. In some embodiments, X4 and X5 in Formula (VI-b) may be both N, and the Compounds may be represented by, for example, formula (II-e), formula (II-f, formula (II-g), or formula (II-h): In some embodiments, in Formula (VI-b), X4 and X5are both N, R12is a methyl group, and R13-R16are H, in such cases, the Compounds may be represented by, for example, formula (II-g1): As another example, one of R5-R8may be a group represented by formula (VI-c): In some embodiments, X6may be S, O, SO2, N, C, or C-L2. L2may be selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group. L2may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. In some embodiments, R17does not exist, or may be selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. In some embodiments, R18—R21may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and each of R18-R21may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. Formula (VI-c) may be connected with the benzoxazole group shown in formula (II), formula (III), formula (IV), or formula (V) at methyl. In some embodiments, X6in Formula (VI-c) may be N, and the Compounds may be represented by, for example, formula (II-i), (II-j), (II-k), or (II-l): In some embodiments, in Formula (VI-c), X6is N, R18-R21are H, and R17is a methyl group, —CH2—CHF2, or —C2H4—OCH3, in such cases, the Compounds may be represented by, for example, formula (II-k1), formula (II-k2), and formula (II-k2),: As yet another example, one of R5-R8may be a group represented by formula (VI-d): In some embodiments, R22may be H, an alkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, or a heterocyclic group, and R22may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, and an alkoxy group. In some embodiments, R23-R25may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group. In such cases, the Compounds may be represented by, for example, formula (II-m), (II-n), (II-o), or (II-p): In some embodiments, in Formula (VI-d), R22is a methyl group, and R23—R25are H, in such cases, the Compounds may be represented by, for example, (11-01): In some embodiments, at least one of R5—R8is a heteroaryl group, which is optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and the other of R5-R8are H. For example, one of R5—R8may be a group represented by formula (VI-e): In some embodiments, R26-R29may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, a heterocyclic group, and N—R38, wherein R38may be selected from H, an alkyl group, an aryl group, a heterocyclic group, and a ketone group. Formula (VI-e) may be connected with the benzoxazole group shown in formula (II), formula (III), formula (IV), or formula (V) at methyl. In such cases, the Compounds may be represented by, for example, formula (II-q), formula (II-r), formula (II-s), or formula (II-t): In some embodiments, in Formula (VI-e), R27—R29are H, and R26is a methyl group, —CHF2, or a cyclopropyl group, accordingly, the Compounds may be represented by, for example, formula (II-r1), formula (II-r2), and formula (II-r3): As another example, one of R5-R8may be a group represented by formula (VI-f: In some embodiments, R30-R33may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, a heterocyclic group, and N—R38, wherein R38may be selected from H, an alkyl group, an aryl group, a heterocyclic group, and a ketone group. Formula (VI-f may be connected with the benzoxazole group shown in formula (II), formula (III), formula (IV), or formula (V) at methyl. In such cases, the Compounds may be represented by, for example, formula (II-u), formula (II-v), formula (II-w), or formula (II-x): In some embodiments, in Formula (VI-f, R31-R33are H, and R30is a methyl group, —NH2, —NC2H6, —NHCOCH3, or —NHCH3, accordingly, the Compounds may be represented by, for example, formula (II-v1), formula (II-v2), formula (II-v3), formula (II-v4), and formula (II-v5): As yet another example, one of R5—R8may be a group represented by formula (VI-g): In some embodiments, R34-R37may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, a heterocyclic group, and N—R38, wherein R38may be selected from H, an alkyl group, an aryl group, a heterocyclic group, and a ketone group. Formula (VI-g) may be connected with the benzoxazole group shown in formula (II), formula (III), formula (IV), or formula (V) at methyl. In such cases, the Compounds may be represented by, for example, formula (II-y): In some embodiments, in Formula (VI-g), R35—R37are H, and R34is a methyl group, accordingly, the Compounds may be represented by, for example, formula (II-y1): In some embodiments, R3may be an aryl group, and R3may be optionally substituted by one or more groups selected from halogen, a cycloalkyl group, a fluoroalkyl group, a methyl group, an ethyl group, a propyl group, and a butyl group. For example, R3may be benzene halide. In some embodiments, R3may be a heterocyclic group, and R3may be optionally substituted by one or more groups selected from halogen, a cycloalkyl group, a fluoroalkyl group, a methyl group, an ethyl group, a propyl group, and a butyl group. For example, R3is a group represented by formula (VII-a): In some embodiments, R39-R42may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, a heterocyclic group, and N—R49, wherein R49is selected from H, an alkyl group, an aryl group, a heterocyclic group, and a ketone group. Formula (VII-a) may be connected with —CO— or —SO2— shown in formula (II), formula (III), formula (IV), or formula (V) at methyl. In such cases, the Compounds may be represented by, for example, formula (II-aa): In some embodiments, R40-R42are H, and R39is a methyl group, accordingly, the Compounds may be represented by, for example, formula (II-aa1): In some embodiments, R3may be selected from an imidazole group, a pyrrole group, a pyrazole group, a triazole, a piperidine group, a pyridine group, a pyrimidine group, and a pyridazine group, and R3may be optionally substituted by one or more groups selected from a cycloalkyl group, a fluoroalkyl group, a methyl group, and a tertiary butyl group. For example, R3is a group represented by formula (VII-b): In some embodiments, R43may be H, an alkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, or a heterocyclic group. In some embodiments, R44and R45may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and each of R44and R45may be optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. Formula (VII-b) may be connected with —CO— or —SO2— shown in formula (II), formula (III), formula (IV), or formula (V) at methyl. In such cases, the Compounds may be represented by, for example, formula (II-bb): In some embodiments, R45is H, R43is a methyl group, a cyclopropyl group, or and R44is a tert-butyl group, a cyclopropyl group, or As another example, R3is a group represented by formula (VII-c): In some embodiments, R46may be H, an alkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, or a heterocyclic group. In some embodiments, R47and R48may be independently selected from H, halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an alkoxy group, an aryl group, a heteroaryl group, and a heterocyclic group, and each of R47and R48is optionally substituted by one or more groups selected from halogen, a hydroxyl group, an alkyl group, a fluoroalkyl group, a cycloalkyl group, an aryl group, a heterocyclic group, and an alkoxy group. Formula (VII-c) may be connected with —CO— or —SO2— shown in formula (II), formula (III), formula (IV), or formula (V) at methyl. In such cases, the Compounds may be represented by, for example, formula (II-cc): In some embodiments, R48is H, R46is a methyl group, a —CHF2, or —CF3, and R47is a cyclopropyl group. In some embodiments, R1and R2may be H. In some embodiments, R1and R2may be —CH2— groups connected via a single bond. Some exemplary compounds as CatL inhibitors provided by the present disclosure are shown in Table 1: TABLE 1exemplary compounds as CatL inhibitorsCompound 1Compound 2Compound 3Compound 4Compound 5Compound 6Compound 7Compound 8Compound 9Compound 10Compound 11Compound 12Compound 13Compound 14Compound 15Compound 16Compound 17Compound 18Compound 19Compound 20Compound 21Compound 22Compound 23Compound 24Compound 25Compound 26Compound 27Compound 28Compound 29Compound 30Compound 31Compound 32Compound 33Compound 34Compound 35Compound 36Compound 37Compound 38Compound 39Compound 40Compound 41Compound 42Compound 43Compound 44Compound 45Compound 46Compound 47Compound 48 It should be noted that the Compounds listed above in Table 1 are provided merely for illustration purposes. Other Compounds represented by the formulas presented in the present disclosure (e.g., formula (I-a), formula (I-b), formula (I-c), formula (II), formula (III), formula (IV), and formula (V)) are also within the scope of the present disclosure. According to another aspect of the present disclosure, a composition is provided. The composition may include at least one of the Compounds described previously, an isomer thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a solvate thereof, or a pharmaceutically acceptable salt thereof, in a pharmaceutically effective amount. In some embodiments, the composition may further include a pharmaceutically acceptable carrier. For instance, the carrier may include a coating layer, a capsule, a microcapsule, a nano-capsule, or the like, or any combination thereof. It should be noted that the carrier may need to be non-toxic and may not have significant impacts on the activity of the key ingredients in the pharmaceutical composition (e.g., the Compounds described above). In some embodiments, the carrier may protect the key ingredients against some undesired conditions, such as oxidation, decomposition, or inactivation of the key ingredients. For instance, enzymes or relatively low-pH in the stomach may cause the decomposition or inactivation of the key ingredients. The carrier may help maintain or increase the efficacy of the pharmaceutical composition by protecting the key ingredients in the pharmaceutical composition. In some embodiments, the carrier may be used for the controlled release of the key ingredients. The controlled release may include but is not limited to slow release, sustained release, targeted release, or the like. For instance, the carrier may include hydrogel capsules, microcapsules, or nano-capsules made of collagen, gelatin, chitosan, alginate, polyvinyl alcohol, polyethylene oxide, starch, cross-linked starch, or the like, or any combination thereof. In some embodiments, the carrier may facilitate a controlled release of the key ingredients (e.g., at least one of the Compounds described previously) in the pharmaceutical composition. In some embodiments, the composition may be administered to the subject via an oral administration, an injection administration, an inhalation administration, or a topical administration. In some embodiments, the injection administration may include subcutaneous injection, intramuscular injection, intravenous injection, or the like. In some embodiments, the injection administration may include the injection of the composition into a tumor or a region close to the tumor. In some embodiments, the injection administration may include injection of the composition into the kidney, liver, heart, thyroid, or joints. In some embodiments, the inhalation administration may include applying the composition dispersed via an aerosol spray, mist, or powder. In some embodiments, the topical administration may include applying the composition on the skin to attenuate cancer such as skin cancer, or lymphoma. In some embodiments, the topical administration may include vaginal administration, rectal administration, nasal administration, auricular administration, intramedullary administration, intra-articular administration, intra-pleural administration, or the like, or any combination thereof. In some embodiments, the composition may be administered to the subject via a combination of different means of administration. In some embodiments, the method may include administering the composition to the subject three times a day, two times a day, one time a day, once every two days, etc. In some embodiments, a method of treating a disease in a subject is provided. The method may include administering the composition described earlier to the subject. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal. In some embodiments, the subject is male. In some embodiments, the subject is female. In some embodiments, the subject is suffering from a disease or pathological condition. In some embodiments, the disease may be caused by a viral infection. The Compounds provided by the present disclosure exhibit remarkable anti-viral effects. Thus, the Compounds may be used for treating diseases related to virus infections. For example, the disease may include a severe acute respiratory syndrome (SARS), severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) infection, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, or a coronavirus disease 19 (COVID-19). Merely by way of example, the Compounds may be used for treating the long-term effects of coronavirus (long COVID) or post-acute sequelae of COVID-19 (PASC). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a coronavirus that has caused the coronavirus disease 2019 (COVID-19) pandemics in the past three years. It imposes a great threat to public health and social-economical systems worldwide. To date, most COVID-19 treatments have been focused on targeting the viral spike protein (S protein) and viral proteases (mainly 3C-like protease and papain-like protease). These treatments have been proven to be effective in preventing SARS-CoV-2 infection and severe COVID-19 symptoms. However, more immune-evasive and contagious SARS-CoV-2 variants continue emerging and spreading around the world. This demands innovative strategies to develop new antiviral medicines to combat COVID-19. As another example, the disease may include herpes simplex virus (HSV) infection. Infection with HSV, known as herpes, is common globally. Some medications are available to reduce the severity and frequency of symptoms, but they may not cure the infection. Cystatin C is a human cysteine proteinase inhibitor present in extracellular fluids. Cystatin C and a tripeptide derivative (Z-LVG-CHN2) that mimics its proteinase-binding center, were tested for possible antiviral activity against HSV type 1 and poliovirus type 1. (J Virol. 1990 February; 64(2): 941-943.) Thus, the compounds provided by the present disclosure may be used for treating HSV infection. As yet another example, the disease may include respiratory syncytial virus (RSV) infection. Human RSV is a globally prevalent cause of lower respiratory tract infection in all age groups. RSV infection is frequently reported in infants, the elderly, and immunocompromised patients. RSV is highly contagious and can be fatal. There is no vaccine currently available to prevent RSV infection. Current anti-viral drugs for the treatment of RSV infection have notable limitations. There is an urgent need to search for novel medicines that can meet clinical demands. RSV infection increases the expression and activity of several host proteases, including MMP and cathepsin families of proteases. The induced host protease response can facilitate RSV infection and may have a major role in disease progression. A selective cathepsin L inhibitor alone or in combination with the inhibition of additional host proteases has the potential to enhance RSV clearance and prevent RSV-induced airway hyperresponsiveness and allergic response. It was reported that, cathepsin inhibitor E64 or ribavirin prevented airway hyperresponsiveness and enhanced viral clearance in RSV infected mice. (Mucosal Immunol. 2015 January; 8(1): 161-175.) Thus, the Compounds provided by the present disclosure may be used for treating RSV infection. As yet another example, the disease may include an ebola virus infection or a middle east respiratory syndrome (MERS). In some embodiments, the disease may be Acute respiratory distress syndrome (ARDS), or ARDS-induced multiple organ failures (e.g., of lung, kidney, liver). In some embodiments, the disease may be acute kidney injury (AKI). For example, the AKI may be caused by anti-cancer drugs, microbial infections, parasites, etc. In some embodiments, the disease may be liver injury or liver fibrosis. In some embodiments, the disease may be cancer. In some embodiments, the disease may be osteoporosis. In some embodiments, the disease may be inflammation. In some embodiments, the disease may be atherosclerosis. In some embodiments, the disease may be a renal disease or a bone disease. In some embodiments, the disease may be diabetes. In some embodiments, the method may include orally administering the composition to the subject, injecting the composition to the subject, or administering the composition to the subject via a topical administration. According to another aspect of the present disclosure, a method of inhibiting cathepsin L in a subject is provided. The method may include administering the composition described earlier to the subject. In some embodiments, administering the composition to the subject may include: orally administering the composition to the subject, inhaling the composition to the subject, injecting the composition to the subject, or administering the composition to the subject via a topical administration. According to another aspect of the present disclosure, a use of the Compounds described earlier for inhibiting cathepsin L in a subject is provided. The use of the Compounds may include steps mentioned in the method for inhibiting CatL. According to yet another aspect of the present disclosure, a use of at least one of the Compounds described earlier for preparing a composition for treating a disease in a subject is provided. The present disclosure is further described according to the following examples, which should not be construed as limiting the scope of the present disclosure. EXAMPLES Abbreviations Å=angstrom Ac=acetyl Ac2O=acetic anhydride Boc2O=di-tert-butyl dicarbonate DCM=dichloromethane DIPEA=N,N-Diisopropylethylamine or N-ethyl-N-isopropyl-propan-2-amine DMAP=dimethylamino pyridine DMA=dimethyl acetamide DME=dimethoxyethane DMF=dimethylformamide DMSO=dimethyl sulfoxide EtOAc/EA=Ethyl Acetate EtOH=ethanol FA=formic acid HATU=1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate HOAc=acetic acid KOAc=potassium acetate LiHMDS=lithium bis(trimethylsilyl)amide MeMgBr=methylmagnesium bromide MeOH=methanol NaOAc=sodium acetate NBS=N-bromosuccinimide Pd(dppf)2Cl2=[1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) PE=petroleum ether PTSA=p-Toluenesulfonic acid monohydrate rt=room (ambient) temperature T3P=2,4,6-Tripropyl-1,3,5,2,4,6-trioxatriphosphorinane-2,4,6-trioxide TEA=triethylamine TFA=trifluoroacetic acid THF=tetrahydrofuran TsCI=p-toluene sulfonyl chloride UV=ultra-violet X-Phos=2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl Example 1-Inhibition of CatL by the Compounds In Vitro Biological Data Human Cathepsin L (CatL) Enzymatic Assay Human cathepsin L enzymatic assay was performed in assay buffer (50 mM MES pH 5.5, 2.5 mM DTT, 0.5 mM EDTA) to assess the inhibition of human CatL by tested compounds. 60 μL of the compounds were added to 384-well dilution plate. The compound solution was diluted for 1:3 in succession in DMSO for each column for 10 pts. 0.05 μL diluted compound solution was added in each row to 384 assay plates (Corning 4514) using Echo (LABCYTE 655), each column containing 2 replicates. 5 μL working solution of human CatL enzyme (Abcam #ab81780) was added to 384-well assay plate, centrifuge 1000 rpm for 1 min. The mixture was incubated at 2500 for 15 min, then add 5 μL working solution of CatL substrate (Genscript #C7360HB140_5) to initiate the reaction (CatL: 0.05 nM, Substrate: 500 nM). Incubate at 2500 for 30 min. Reading Ex370 nm and Em460 nm fluorescence signals with BMG CLARIO Star Plusaucu. Percent inhibition for each compound was calculated and IC50(half maximal inhibitory concentration) was fitted from non-linear regression by XLfit 5.5.0. The results of CatL enzymatic assays are shown in the following Table 2. TABLE 2IC50of Compounds 1-48 for inhibiting CatLEnzymatic inhibitionCompoundIC50(nM)114.127.535.9434.9512267.0721.1817.4930.51051.51194.612235133.9145341519.416139172011850.71944.6204.2218.2225.323361249.72536.7261.4278.52812.3291.3301243111.732>1000332.9340.6351.13679.337125381.7391.6401.5415.3424.5430.84416.5452.5461.0476.6483.5 This example shows that compounds provided by the present disclosure may be used effectively as CatL inhibitors. According to the results, many of the compounds 1-48 exhibit remarkable inhibition capacities with respect to CatL. Specifically, the IC50of Compounds 1-4, 6-9, 13, 15, 19-22, 24-29, 31, and 44 for inhibiting CatL was less than 50 nM. Moreover, the IC50of Compounds 2, 3, 6, 13, 20-22, 24, 26, 27, 29, 33-35, 38-43, and 45-48 for inhibiting CatL was less than 10 nM. Example 2-Pseudovirus Infection Assay In order to infect the host, SARS-CoV-2 needs to enter host cells for viral replication. This depends on the proper cleavage and activation of the viral S protein by host cell proteases, primarily furin, TMPRSS2, and cathepsin L (also known as CatL or CTSL). TMPRSS2 and furin cleave viral S protein at different sites, priming the virus for entry into host cells. CatL then cleaves S protein into smaller fragments, promoting the fusion between virus and endosome membrane, allowing the release of the viral genome into host cells for viral replication. The CatL cleavage sites are highly conserved among all known SARS-CoV-2 variants. Therefore, the inhibition of CatL alone or in combination with the inhibition of additional host proteases will likely prevent the proper processing of S protein, and the infection of SARS-CoV-2 and its variants. Pseudoviruses (PsVs) incorporated with S protein from SARS-CoV-2, or mutants were constructed using a reported procedure. For this VSV-based PsV system, the backbone was provided by VSV-G pseudotyped virus (G*AG-VSV) that packages expression cassettes for firefly luciferase instead of VSV-G in the VSV genome. For quantification of PsV, viral RNA was extracted by using the QlAamp Viral RNA Mini Kit (Cat. No. 52906, QIAGEN), and the reverse transcription was performed with RevertAid™ First Strand cDNA Synthesis Kit (Fermentas K1622) according to the manufacturer. The real-time qPCR was then performed on the LightCycler® 96 Real-Time PCR System (Roche) using SYBR Green I Master Mix reagent (Roche). The P protein gene of VSV virus was quantified and the viral copy number is calculated accordingly. The forward primers were: TCTCGTCTGGATCAGGCGG (SEQ ID NO: 1); the reverse primer is: TGCTCTTCCACTCCATCCTCTTGG (SEQ ID NO: 2). All PsVs were normalized to the same amount as previously described (See Zhao, M. M. et al. Novel cleavage sites identified in SARS-CoV-2 spike protein reveal the mechanism for cathepsin L-facilitated viral infection and treatment strategies. Cell Discovery. 8: 53-70 (2022)). Vero E6 cells were maintained in high glucose Dulbecco's modified Eagle's medium (DMEM) (Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS, Gibco, Carlsbad, CA), 100 units/mL Penicillin-Streptomycin (Gibco). All the cells were maintained at 37 00 in a humidified atmosphere containing 95% air and 5% CO2. To assess the anti-viral effect of tested compounds, Vero E6 cells were seeded in 96-well cell culture plates, then co-treated with different concentrations of tested compounds and SARS-CoV-2 PsVs (100 μL of the normalized PsV was added to each well). After 24 h incubation at 3700, the activities of firefly luciferase were measured in cell lysates using luciferase substrate (PerkinElmer, BRITELITE PLUS 100 mL KIT, Cat. No. 6066761) following the manufacturer's instructions. Luciferase activity was quantified using a luminometer (Promega). The infection rate was calculated from a control reaction containing only vehicle. The results of pseudovirus infection assays are shown in the following Table 3. TABLE 3Results of compounds 1-48 in Pseudovirus infection assay% Infection rate% InfectionCompoundat 500 nMrate at 5 uM1693925512365104815159181675207964889269997731097961186811355101410397157920189272.61910177.320406.32160122233132393982434225541526252.2274013287038297.38.83099101316710321019533395.934410.935403.136110125379610438341.039324.0405712419057.742345.643180.8448255.5457.00.02467.00.08475615.9486322.4 This example indicates that the Compounds provided by the present disclosure may effectively protect cells from viral infections and may be used for treating or preventing a disease related to viral infections (e.g., SARS-CoV-2). According to the results in Table 3, many of Compounds 1-48 are capable of protecting cells from pseudovirus infections in dose-dependent manners from 500 nM to 5 uM, and some Compounds (e.g., Compound 29, 45, 46) demonstrated nearly full protection at 500 nM. Example 3-Preparation of Compounds 1-48 General Procedure A The general procedure A for preparing a compound A-7 is illustrated inFIG.1. As illustrated inFIG.1, the general procedure A may include steps A-D. Step A. Benzoxazole Ring Formation Ethyl (tert-butoxycarbonyl)-L-asparaginate (A-1, 1 equiv.) was dissolved in 1,2-dichloroethane (0.2 M), Et3O+BF4−(1.2 equiv.) was added in portions under nitrogen. The resulting mixture was stirred for 24 h at RT (room temperature). A solution of (substituted) aminophenol (A-2, 1 equiv.) in 2 mL ethanol (2 M) was transferred into the solution through a syringe. The mixture was heated to 90° C. and stirred for 24 h. The reaction mixture was cooled to room temperature, saturated NaHCO3(aq.) was added, and then the aqueous layer was extracted with dichloromethane. The combined organic layer was washed with brine and dried over anhydrous Na2SO4. After filtration and concentration, the crude product was purified by chromatography (EtOAc in PE, 25% to 50%) to give the desired product A-3. Step B. Ester Hydrolysis A solution of ester A-3 (1 equiv.) in THF (0.1 M) and 1N LiOH (aq., 3 equiv.) was stirred for 3 h at rt. The reaction was adjusted to pH=7 by HCl (1 M) then extracted with DCM/MeOH (v/v, 10:1). The combined organic layer was separated, dried (MgSO4) and concentration to obtain the desired product A-4. Step C. Amide Coupling (C-Terminal) To a stirred solution of acid A-4 (1 equiv.), 1-aminocyclopropane-1-carbonitrile hydrochloride A-5 (1.2 equiv.) and T3P (50 wt % EA solution, 1.1 equiv.) in DCM (0.2 M) was added DIPEA (4 equiv.). The reaction mixture was stirred under N2atmosphere at rt for 3 hrs. The reaction mixture was concentrated and purified by chromatography (EtOAc in PE, 40% to 100%) to give the desired product A-6. Step D. N-Boc Deprotection A solution of protected amine A-6 in HCOOH (0.4 M) was stirred at 25° C. for 5 h. The mixture was blown by nitrogen to dryness at 20° C., basified with saturated NaHCO3(aq.) and extracted with EA. The combined organic layer was concentrated to give the desired product A-7. General Procedure B-1 The general procedure B-1 for preparing a compound A-8 based on Compound A-7 is illustrated inFIG.2. A solution of amine A-7 (1 equiv.), acyl chloride (1.1 equiv.) and DIPEA (3 equiv.) in DCM (0.5 M) was stirred under at 25° C. for 2 hrs. The reaction mixture was concentrated, the residue was purified by prep-High Performance Liquid Chromatography (HPLC) [(Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA); 15%-80%)] to give the desired product A-8. General Procedure B-2 The general procedure B-2 for preparing Compound A-8 based on Compound A-7 is illustrated inFIG.3. To a stirred solution of amine A-7, acid (1.2 equiv.) and DIPEA (4 equiv.) in DCM (0.1 M) was added T3P (50 wt % DMF solution, 1.2 equiv.) dropwise. The reaction was stirred at 25° C. for 3 h. The reaction mixture was quenched with water and extracted with DCM. The combined organic layer was dried over Na2SO4and concentrated under vacuum to give a crude product, which was purified by prep-HPLC [(Gemini-C18, 150×21.2 mm, 5 um; ACN-H2O (0.1% FA); 15%-80%)] to give the desired product A-8. General Procedure B-3 The general procedure B-3 for preparing Compound A-8 based on Compound A-7 is illustrated inFIG.4. To a stirred solution of amine A-7 (70 mg, 0.2 mmol) in DMF (0.2 M) was added acid (1 equiv.), DIPEA (5 equiv.) and HATU (3 equiv.). The reaction mixture was stirred at rt under N2for 2 hrs. After the reaction completed, H2O was added to the reaction mixture, and then extracted with EA. The combined organic layer was washed with brine, dried over anhydrous Na2SO4. After filtration, the solution was concentrated under vacuum, and the residue was purified by prep-HPLC to give the desired product A-8. Synthesis of Intermediates Preparation of Intermediate I-1: 3-(tert-butyl)-1-cyclopropyl-1H-pyrazole-5-carboxylic acid FIG.5is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-1 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl 3-(tert-butyl)-1-cyclopropyl-1H-pyrazole-5-carboxylate To a solution of cyclopropylhydrazine hydrochloride (1.5 g, 0.0138 mol) in EtOH (40 mL) was added 5 N aq. NaOH solution (3 mL) and stirred for 10 min at 0° C. Then the mixture was added to an ethanol solution of ethyl 5,5-dimethyl-2,4-dioxohexanoate (4.14 g, 0.02 mol). The resulting mixture was stirred at 60° C. for 16 hrs. The mixture was concentrated and the residue was purified by flash column (PE/EA=50:1) to give the product as colorless oil (1.79 g, 50.3%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 237.1 [M+H]+. Step 2. Preparation of 3-(tert-butyl)-1-cyclopropyl-1H-pyrazole-5-carboxylic acid To a solution of ethyl 3-(tert-butyl)-1-cyclopropyl-1H-pyrazole-5-carboxylate (1.79 g, 7.6 mmol) in THF (18 mL), H2O (6 mL), and MeOH (6 mL) was added LiOH (3.19 g, 76 mmol) and stirred at RT for 5 hrs. The solvent was removed under reduced pressure. The residue was dissolved in H2O (20 mL) and adjusted to pH 7 by using 1 N aq. HCl. Then the mixture was extracted with DCM (100 mL×2). The combined organic layer was washed with brine (50 mL), then dried over anhydrous Na2SO4. After filtration, the solution was concentrated under vacuum to give the product as a light pink solid (1.45 g, 86%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 209.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 13.16 (s, 1H), 6.67 (s, 1H), 4.27 (d, J=3.8 Hz, 1H), 1.21 (s, 9H), 1.11-1.04 (m, 2H), 0.95 (dd, J=7.2, 2.4 Hz, 2H). Preparation of Intermediate I-2: 1-cyclopropyl-3-(trifluoromethyl)-1H-pyrazole-4-carboxylic acid FIG.6is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-2 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl 1-cyclopropyl-3-(trifluoromethyl)-1H-pyrazole-4-carboxylate To a solution of ethyl (Z)-2-(ethoxymethylene)-4,4,4-trifluoro-3-oxobutanoate (2 g, 8.3 mmol) in toluene (20 mL) at 0° C. was added cyclopropylhydrazine hydrochloride (0.54 g, 4.9 mmol). The mixture was stirred at 50° C. under N2for 16 hrs. After completion, the mixture was concentrated under vacuum. The residue was purified by prep-TLC (PE:EA=4:1) to give the product as a white solid (500 mg, 22%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 249.2 [M+H]+. Step 2. Preparation of 1-cyclopropyl-3-(trifluoromethyl)-1H-pyrazole-4-carboxylic acid A solution of ethyl 1-cyclopropyl-3-(trifluoromethyl)-1H-pyrazole-4-carboxylate (620 mg, 2.48 mmol) in 1 N aq. LiOH (5 mL) and THF(5 mL) was stirred at RT for 16 hours. The mixture was acidified to pH 3-4 with 5 N aqueous HCl and extracted with EA (20 mL×3). The combined organic layers were washed with brine (20 mL×2), and dried over Na2SO4. Then by filtration, the filtrate was concentrated to give the product as a white solid (447 mg, 70%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass (m/z): 221.0 [M+H]+. Preparation of Intermediate I-3: 1-cyclopropyl-3-(difluoromethyl)-1H-pyrazole-4-carboxylic acid FIG.7is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-3 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl (Z)-2-(ethoxymethylene)-4,4-difluoro-3-oxobutanoate A solution of ethyl 4,4-difluoro-3-oxobutanoate (5 g, 30 mmol) and (diethoxy methoxy) ethane (10 mL, 58 mmol) in acetic acid anhydride (30 mL) was stirred at 140° C. under N2for 6 hrs. After completion, the mixture was concentrated under vacuum to give the product as light-yellow oil (5 g, 74%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass (m/z): Mass(m/z): 222.0 [M+H]+. Step 2. Preparation of ethyl 1-cyclopropyl-3-(difluoromethyl)pyrazole-4-carboxylate To a solution of ethyl (Z)-2-(ethoxymethylene)-4,4-difluoro-3-oxobutanoate (1.55 g, 0.45 mmol) in toluene (10 mL) at 0° C. was added cyclopropylhydrazine hydrochloride (0.46 g, 4.2 mmol). The mixture was stirred at 50° C. under N2for 16 hrs. After completion, the mixture was concentrated under vacuum. The residue was purified by prep-TLC (PE:EA=5:1) to give the product as a light-yellow solid (210 mg, 11%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 231.0 [M+H]+. Step 3. Preparation of 1-cyclopropyl-3-(difluoromethyl)-1H-pyrazole-4-carboxylic acid A solution of ethyl 1-cyclopropyl-3-(trifluoromethyl)-1H-pyrazole-4-carboxylate (210 mg, 0.9 mmol) in 1 N aq. LiOH (2 mL) and THF (2 mL) was stirred at RT for 16 hours. The mixture was acidified to pH 3-4 with 5 N aqueous HCl and extracted with EA (20 mL×3). The combined organic layers were washed with brine (20 mL×2), and dried over Na2SO4. Then by filtration, the filtrate was concentrated to give the product as a white solid (150 mg, 77%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass (m/z): 203.1 [M+H]+. Preparation of Intermediate I-4: 3-(tert-butyl)-1-(oxetan-3-yl)-1H-pyrazole-5-carboxylic acid FIG.8is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-4 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl 3-(tert-butyl)-1-(oxetan-3-yl)-pyrazole-5-carboxylate To a solution of ethyl 3-(tert-butyl)-1H-pyrazole-5-carboxylate (1.6 g, 8 mmol) and K2CO3(2.24 g, 16 mmol) in DMF (8 mL) was added 3-iodooxetane (1.3 mL, 12 mmol). The reaction was stirred at 75° C. for 24 hrs. The reaction mixture was quenched with ice water, and extracted with EtOAc (30 mL×3). The combined organic layer was washed with water (30 mL×3) and brine (30 mL), dried with anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by flash chromatography (PE/EtOAc=4:1) to give the product as a white solid (1.9 g, 94%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 253.2 [M+H]+. Step 2. Preparation of 3-(tert-butyl)-1-(oxetan-3-yl)-pyrazole-5-carboxylic acid To a solution of ethyl 3-(tert-butyl)-1-(oxetan-3-yl)-pyrazole-5-carboxylate (500 mg, 2 mmol) in THF/H2O (5:1, 10 mL) was added LiOH.H2O (420 mg, 10 mmol). The reaction mixture was stirred at RT for 3 hrs. The reaction solution was adjusted pH to 5˜6 by using 1N aq.HCl. Then the mixture was extracted with EtOAc (30 mL×2). The combined organic layer was washed with brine (30 mL), then dried over with anhydrous Na2SO4. The mixture was filtered, and the filtrate was concentrated under vacuum to afford desired product (400 mg, 89%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 225.1 [M+H]+. Preparation of Intermediate I-5: 1,3-dicyclopropyl-1H-pyrazole-5-carbonyl chloride FIG.9is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-5 according to some embodiments of the present disclosure. Step 1. Preparation of methyl 1,3-dicyclopropyl-1H-pyrazole-5-carboxylate To a solution of the cyclopropylhydrazine dihydrochloride (2813 mg, 19.4 mmol) in EtOH (50 mL) was added 5N NaOH (3 mL). After stirred for 10 min at 0° C. A solution of Methyl 4-cyclopropyl-2,4-dioxobutanoate (2200 mg, 12.9 mmol) in EtOH (50 mL) was added and the resulting mixture was stirred at 60° C. for 14 hrs. The solvent was removed under reduced pressure and the residue was purified by Combiflash column (PE/EA=0˜ 50%) to give the product (800 mg, 27%) as colorless oil. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 206.9 [M+H]+. Step 2. Preparation of 1,3-dicyclopropyl-1H-pyrazole-5-carboxylic acid To a solution of methyl 1,3-dicyclopropyl-1H-pyrazole-5-carboxylate (170 mg, 0.82 mmol) in THF/H2O (5:1, 6 mL) was added LiOH·H2O (346 mg, 8.2 mmol). The reaction mixture was stirred at RT for 4 hrs. The reaction solution was adjusted pH to 5˜ 6 by using 1 N aq.HCl. Then the mixture was extracted with EA (20 mL×2). The combined organic layer was washed with brine (30 mL), then dried over with anhydrous Na2SO4. The mixture was filtered, and the filtrate was concentrated under vacuum to afford compound product (140 mg, 79.5%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 193.1 [M+H]+. Step 3. Preparation of 1,3-dicyclopropyl-1H-pyrazole-5-carbonyl chloride To a solution of the 1,3-dicyclopropyl-1H-pyrazole-5-carboxylic acid (140 mg, 0.73 mmol) in DCM (10 mL) was added oxalyl chloride (140 mg, 1.1 mmol) and DMF (0.05 ml). The reaction mixture was stirred at 0° C. for 1 hour. The solvent was removed under reduced pressure to afford crude product. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 207.1 [M−Cl+MeOH]+. Preparation of Intermediate I-6: 1-cyclopropyl-3-methyl-1H-pyrazole-4-carbonyl chloride FIG.10is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-6 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl 1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxylate A solution of ethyl (Z)-2-(ethoxymethylene)-3-oxobutanoate (5 g, 0.02 mol) and cyclopropylhydrazine (1.94 g, 0.02 mol) in EA (50 mL) was stirred under reflux for 3 hours. The mixture was concentrated under reduced pressure and the residue was purified by flash column (PE/EA=5:1) to give the product as orange oil (0.53 g, 9.6%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 195.1 [M+H]+. Step 2. Preparation of 1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxylic acid To a solution of ethyl 1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxylate (0.53 g, 0.7 mmol) in 1N aq. LiOH (10 mL) and THF (10 mL). The reaction mixture was stirred for 24 hours at 25° C. After completion, the mixture was concentrated under vacuum. The residue was dissolved in water (10 mL), adjusted pH to 7 with 1N aq. HCl, and extracted with ethyl acetate (10 mL×2). The combined organic layer was washed with brine (10 mL), dried over Na2SO4, filtered, and concentrated under vacuum to give the product as a yellow solid (380 mg, 85.7%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 167.2 [M+H]+. Step 3. Preparation of 1-cyclopropyl-3-methyl-1H-pyrazole-4-carbonyl chloride To a solution of 1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxylic acid (150 mg, 0.9 mmol) in DCM (2 mL) was added oxalic dichloride (103 mg, 0.8 mmol) and a drop of DMF. The reaction mixture was stirred at 25° C. for 16 hrs. The mixture are concentrated under vacuum to give the product as a yellow oil (150 mg, 81%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 181.0 [M-Cl+MeOH]+. Preparation of Intermediate I-7: 2-cyclopropyl-5-(1-methylcyclopropyl)pyrazole-3-carboxylic acid FIG.11is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-7 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl 4-(1-methylcyclopropyl)-2,4-dioxobutanoate To a mixture of 1-(1-methylcyclopropyl)ethenone (3.00 g, 30.6 mmol) and diethyl oxalate (4.47 g, 30.6 mmol) in THF (30.0 mL) was added LiHMDS (30.6 mL, 30.6 mmol). The reaction was stirred at −70° C. for 16 hrs. The reaction mixture was quenched with NH4Cl solution (100 mL) at 0° C., then extracted with EA (100 mL×3). The combined organic layers were washed with brine (100 mL×3), dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by flash column (PE: EA=0˜50%) to give the desired product ethyl 4-(1-methylcyclopropyl)-2,4-dioxobutanoate as brown oil (2.80 g, 42%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 199.1 [M+H]+. Step 2. Preparation of ethyl 2-cyclopropyl-5-(1-methylcyclopropyl)pyrazole-3-carboxylate A mixture of ethyl 4-(1-methylcyclopropyl)-2,4-dioxobutanoate cyclopropyl hydrazine (3.07 g, 21.1 mmol) in EtOH (10.0 mL) was adjusted pH to 10 with aq.NaOH (5N) at 0° C. Then the mixture was added to a solution of ethyl 4-(1-methylcyclopropyl)-2,4-dioxobutanoate (2.80 g, 14.1 mmol) in EtOH (20.0 mL) at 0° C. The reaction was stirred at 60° C. for 16 hrs. The reaction mixture was concentrated under reduced pressure. The residue was purified by flash column (PE:EA=0-5%) to give the product ethyl 2-cyclopropyl-5-(1-methylcyclopropyl)pyrazole-3-carboxylate as colorless oil (2.20 g, 60%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 235.1 [M+H]+. Step 3. Preparation of 2-cyclopropyl-5-(1-methylcyclopropyl)pyrazole-3-carboxylic acid To a mixture of ethyl 2-cyclopropyl-5-(1-methyl cyclopropyl) pyrazole-3-carboxylate (2.20 g, 9.40 mmol) in THF/H2O (3:1, 24.0 mL) was added LiOH (680 mg, 28.2 mmol). The reaction was stirred at RT for 16 hrs. The reaction mixture was concentrated under reduced pressure. The residue was diluted with water (50 mL), then adjusted pH to 4 with aq. HCl (2 M). The mixture was extracted with EA (100 mL×3), washed with brine (100 mL), dried over Na2SO4, and concentrated under reduced pressure to give the product 2-cyclopropyl-5-(1-methylcyclopropyl) pyrazole-3-carboxylic acid as a white solid (1.20 g, 55%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 207.1 [M+H]+. Preparation of Intermediate I-8: (S)-3-(6-bromobenzo[d]oxazol-2-yl)-2-((tert-butoxycarbonyl) amino)propanoic acid FIG.12is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-8 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl (S)-3-(6-bromobenzo[d]oxazol-2-yl)-2-((tert-butoxycarbonyl)amino)propanoate To a solution of ethyl (tert-butoxycarbonyl)-L-asparaginate (20.7 g, 79.78 mmol) in DCE (300 mL) was added Triethyloxonium tetrafluoroborate (15.1 g, 79.78 mmol). The reaction mixture was stirred at 25° C. under N2for 16 hrs. To the resulting mixture was added 2-amino-5-bromophenol (15 g, 79.78 mmol). The reaction mixture was stirred at 85° C. under N2for 16 hrs. The mixture was diluted with water (500 mL), and extracted with DCM (300 mL×2). The organic phase was evaporated, and the residue was purified by silica gel column chromatography (PE:EA=5:1) to give the product as a black oil (19 g, 46%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 413.0 415.0[M+H]+. Step 2. Preparation of (S)-3-(6-bromobenzo[d]oxazol-2-yl)-2-((tert-butoxycarbonyl) amino)propanoic acid To a solution of ethyl (S)-3-(6-bromobenzo[d]oxazol-2-yl)-2-((tert-butoxycarbonyl)amino)propanoate (6 g, 14.52 mmol) in THF (45 mL) and H2O (15 mL) was added LiOH·H2O (914 mg, 21.78 mmol). The reaction mixture was stirred at 25° C. under N2for 2 hrs. The mixture was adjusted pH to 6˜ 7 with 2 N aq.HCl. The mixture was washed with water (50 mL), and the mixture was extracted with EA (50 mL×3). The organic phase was washed with brine (50 mL×2), dried over Na2SO4, and evaporated to give the product as black oil (5.2 g, 74%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 385.0 387.0[M+H]+. Preparation of Intermediate I-9: (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-2-[(3-chlorophenyl)formamido]-N-(1-cyanocyclopropyl)propanamide FIG.13is a schematic diagram illustrating an exemplary procedure for preparing Intermediate I-9 according to some embodiments of the present disclosure. Step 1. Preparation of tert-butyl N-[(1S)-2-(6-bromo-1,3-benzoxazol-2-yl)-1-[(1-cyanocyclopropyl)carbamoyl]ethyl]carbamate To a solution of (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-2-{[(tert-butoxy) carbonyl]amino}propanoic acid (3.3 g, 0.0086 mol) in DMF (30 mL) was added DIEA (3.33 g, 0.0258 mol), HATU (4.10 g, 0.0129 mol), and 1-aminocyclopropane-1-carbonitrile (1.06 g, 0.0129 mol). The solution was stirred at 25° C. under N2for 2 hrs. Water (50 mL) was added, and the mixture was extracted with EA (40 mL×3). The combined organic layer was washed with brine (30 mL×3), then dried over with anhydrous Na2SO4. After filtration, the solution was concentrated under vacuum, and the crude product was purified by Combiflash (EA/PE=20%-25%) to afford tert-butyl N-[(1S)-2-(6-bromo-1,3-benzoxazol-2-yl)-1-[(1-cyanocyclopropyl)carbamoyl] ethyl]carbamate as a yellow solid (1.6 g, 39.53%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 449.3 [M+H]+. Step 2. Preparation of (2S)-2-amino-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propanamide A solution of tert-butyl N-[(1S)-2-(6-bromo-1,3-benzoxazol-2-yl)-1-[(1-cyanocyclopropyl)carbamoyl]ethyl]carbamate (1.6 g, 0.0036 mmol) in FA (10 mL) was stirred at 25° C. for 1 hr. The result was concentrated under vacuum to afford (2S)-2-amino-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propanamide as a yellow solid (900 mg, 69%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass (m/z): 349.1 [M+H]+. Step 3. Preparation of (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-2-[(3-chlorophenyl)formamido]-N-(1-cyanocyclopropyl)propanamide To a solution of (2S)-2-amino-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (600 mg, 1.71 mmol) in DMF (6 mL) was added 3-chlorobenzoic acid (349 mg, 2.23 mmol), HATU (820 mg, 2.23 mmol), and DIEA (666 mg, 5.15 mmol). The solution was stirred at 25° C. under N2for 2 hrs. Water (50 mL) was added, and the mixture was extracted with EA (40 mL×3). The combined organic layer was washed with brine (30 mL×3), then dried over with anhydrous Na2SO4. After filtration, the solution was concentrated under vacuum, and the crude product was purified by Combiflash (EA/PE=25%-30%) to afford (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-2-[(3-chlorophenyl)formamido]-N-(1-cyanocyclopropyl)propanamide as a yellow solid (450 mg, 51%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 487.2 [M+H]+. Example 3.1: Preparation of Compound 1 ((S)—N-(3-(benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide) FIG.14is a schematic diagram illustrating an exemplary procedure for preparing Compound 1 according to some embodiments of the present disclosure. Following general procedure A, from 2-aminophenol, the (S)-2-amino-3-(benzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide was obtained as a yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 271.0 [M+H]+. Following general procedure B-1, from (S)-2-amino-3-(benzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (50 mg), the desired product Compound 1 was obtained as a white solid (25 mg, 33%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 409.0 [M+H]+.1H NMR (400 MHz, CDCl3) δ 8.34 (d, J=7.1 Hz, 1H), 8.17 (s, 1H), 7.87 (t, J=1.7 Hz, 1H), 7.74 (d, J=7.8 Hz, 1H), 7.68 (m, 1H), 7.54 (m, 2H), 7.42 (t, J=7.9 Hz, 1H), 7.36 (m, 2H), 5.16 (td, J=7.0, 4.4 Hz, 1H), 3.69 (dd, J=16.6, 4.3 Hz, 1H), 3.37 (dd, J=16.6, 6.9 Hz, 1H), 1.51 (m, 2H), 1.24 (m, 2H). Example 3.2: Preparation of Compound 2 ((S)—N-(3-(4-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide) FIG.15is a schematic diagram illustrating an exemplary procedure for preparing Compound 2 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-3-bromo-phenol, the (S)-2-amino-3-(4-bromobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide was obtained as a yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 349.02, 351.02 [M+H]+ Following general procedure B-1, from (S)-2-amino-3-(4-bromobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (40 mg), the desired product Compound 2 was obtained as a white solid (4 mg, 7%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 487.01, 489.01 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 9.09 (s, 1H), 9.03 (d, J=7.9 Hz, 1H), 7.85 (t, J=1.8 Hz, 1H), 7.76-7.71 (m, 1H), 7.64 (dd, J=7.9, 0.9 Hz, 1H), 7.60-7.52 (m, 2H), 7.47 (t, J=7.9 Hz, 1H), 7.25 (t, J=8.0 Hz, 1H), 4.96-4.89 (m, 1H), 3.49 (dd, J=15.6, 5.6 Hz, 1H), 3.36 (d, J=8.9 Hz, 1H), 1.44 (m, 2H), 1.08 (m, 2H). Example 3.3: Preparation of Compound 3 ((S)—N-(3-(7-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide) FIG.16is a schematic diagram illustrating an exemplary procedure for preparing Compound 3 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-6-bromo-phenol, the (S)-2-amino-3-(7-bromobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide was obtained as a yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 349.1 [M+H]+ Following general procedure B-1, from 2-amino-6-bromo-phenol, the (S)-2-amino-3-(7-bromobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (35 mg), the desired product Compound 3 was obtained as a white solid (17.2 mg, 35%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 486.8 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.11 (s, 1H), 9.05 (d, J=7.9 Hz, 1H), 7.90 (t, J=1.8 Hz, 1H), 7.79 (d, J=7.8 Hz, 1H), 7.67 (m, 1H), 7.61 (m, 2H), 7.51 (t, J=7.9 Hz, 1H), 7.29 (t, J=8.0 Hz, 1H), 4.97 (td, J=8.5, 5.6 Hz, 1H), 3.54 (dd, J=15.6, 5.5 Hz, 1H), 3.39 (dd, J=12.5, 5.7 Hz, 1H), 1.48 (m, 2H), 1.13 (m, 2H). Example 3.4: Preparation of Compound 4 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-3-(6-fluorobenzo[d]oxazol-2-yl)-1-oxopropan-2-yl)benzamide) FIG.17is a schematic diagram illustrating an exemplary procedure for preparing Compound 4 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-5-fluoro-phenol, the (S)-2-amino-N-(1-cyanocyclopropyl)-3-(6-fluorobenzo[d]oxazol-2-yl)propenamide was obtained as a yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 289.0 [M+H]+ Following general procedure B-1, from (S)-2-amino-N-(1-cyanocyclopropyl)-3-(6-fluorobenzo[d]oxazol-2-yl)propenamide (60 mg), the desired product Compound 4 was obtained as a white solid (20.2 mg, 23%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 427.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.10 (s, 1H), 9.04 (d, J=7.9 Hz, 1H), 7.89 (s, 1H), 7.78 (d, J=7.8 Hz, 1H), 7.66 (m, 3H), 7.50 (d, J=7.9 Hz, 1H), 7.24-7.18 (m, 1H), 4.95 (dd, J=14.0, 8.3 Hz, 1H), 3.48 (dd, J=15.6, 5.9 Hz, 2H), 1.46 (t, J=6.0 Hz, 2H), 1.13-1.03 (m, 2H). Example 3.5: Preparation of Compound 5 ((S)-3-chloro-N-(3-(7-cyanobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)benzamide) FIG.18is a schematic diagram illustrating an exemplary procedure for preparing Compound 5 according to some embodiments of the present disclosure. Following general procedure A, from 3-amino-2-hydroxybenzonitrile, the (S)-2-amino-3-(7-cyanobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide was obtained as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 296.0 [M+H]+ Following general procedure B-1, from (S)-2-amino-3-(7-cyanobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (52 mg), the desired product Compound 5 was obtained as a white solid (12.5 mg, 16%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 434.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.09 (s, 1H), 9.04 (d, J=8.0 Hz, 1H), 8.02 (dd, J=8.0, 1.0 Hz, 1H), 7.87-7.80 (m, 2H), 7.74 (m, 1H), 7.62-7.56 (m, 1H), 7.48 (m, 2H), 4.95 (td, J=8.4, 5.8 Hz, 1H), 3.54 (dd, J=15.5, 5.8 Hz, 1H), 3.39 (dd, J=15.5, 8.7 Hz, 1H), 1.48-1.36 (m, 2H), 1.15-0.98 (m, 2H). Example 3.6: Preparation of Compound 6 ((S)-3-chloro-N-(3-(7-chlorobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)benzamide) FIG.19is a schematic diagram illustrating an exemplary procedure for preparing Compound 6 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-6-chloro-phenol, the (S)-2-amino-3-(7-chlorobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide was obtained as a yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 305.0 [M+H]+ Following general procedure B-1, from (S)-2-amino-3-(7-chlorobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (58 mg), the desired product Compound 6 was obtained as a white solid (68.9 mg, 82%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 443.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.11 (s, 1H), 9.06 (d, J=7.9 Hz, 1H), 7.89 (t, J=1.9 Hz, 1H), 7.79-7.77 (m, 1H), 7.67-7.64 (m, 1H), 7.64-7.61 (m, 1H), 7.51 (t, J=7.9 Hz, 1H), 7.48-7.46 (m, 1H), 7.35 (t, J=8.0 Hz, 1H), 5.01-4.90 (m, 1H), 3.58-3.49 (m, 1H), 3.45-3.36 (m, 1H), 1.52-1.41 (m, 2H), 1.20-1.05 (m, 2H). Example 3.7: Preparation of Compound 7 ((S)-3-chloro-N-(3-(4-chlorobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)benzamide) FIG.20is a schematic diagram illustrating an exemplary procedure for preparing Compound 7 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-3-chloro-phenol, the (S)-2-amino-3-(4-chlorobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide was obtained as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 305.0 [M+H]+ Following general procedure B-1, from (S)-2-amino-3-(4-chlorobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propanamide (50 mg), the desired product Compound 7 was obtained as a white solid (29 mg, 40%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 443.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.11 (s, 1H), 9.06 (d, J=8.0 Hz, 1H), 7.89 (t, J=1.7 Hz, 1H), 7.78 (d, J=7.8 Hz, 1H), 7.64 (m, 2H), 7.51 (t, J=7.9 Hz, 1H), 7.44 (dd, J=8.0, 0.9 Hz, 1H), 7.38 (t, J=8.0 Hz, 1H), 4.97 (dd, J=14.2, 8.1 Hz, 1H), 3.57-3.50 (m, 1H), 3.42-3.36 (m, 1H), 1.46 (m, 2H), 1.17-1.04 (m, 2H). Example 3.8: Preparation of Compound 8 ((S)-3-chloro-N-(3-(5-chlorobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)benzamide) FIG.21is a schematic diagram illustrating an exemplary procedure for preparing Compound 8 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-4-chloro-phenol, the (S)-2-amino-3-(5-chlorobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide was obtained as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 305.0 [M+H]+ Following general procedure B-1, from (S)-2-amino-3-(5-chlorobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propanamide (60 mg), the desired product Compound 8 was obtained as a white solid (28.9 mg, 33%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 442.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.14 (s, 1H), 9.08 (d, J=7.9 Hz, 1H), 7.92-7.86 (m, 1H), 7.79 (dd, J=4.9, 2.7 Hz, 2H), 7.71 (d, J=8.7 Hz, 1H), 7.62 (dd, J=8.0, 1.1 Hz, 1H), 7.52 (q, J=7.5 Hz, 1H), 7.41 (dd, J=8.7, 2.1 Hz, 1H), 5.04-4.89 (m, 1H), 3.50 (m, 1H), 3.39 (d, J=8.7 Hz, 1H), 1.53-1.41 (m, 2H), 1.17-1.02 (m, 2H). Example 3.9: Preparation of Compound 9 ((S)-3-chloro-N-(3-(6-chlorobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)benzamide) FIG.22is a schematic diagram illustrating an exemplary procedure for preparing Compound 9 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-5-chloro-phenol, the (S)-2-amino-3-(6-chlorobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide was obtained as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 305.0 [M+H]+ Following general procedure B-1, from (S)-2-amino-3-(6-chlorobenzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (55 mg), the desired product Compound 9 was obtained as a white solid (30.1 mg, 42%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 442.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.10 (s, 1H), 9.04 (d, J=7.9 Hz, 1H), 7.88 (m, 2H), 7.78 (d, J=7.8 Hz, 1H), 7.68 (d, J=8.5 Hz, 1H), 7.62 (m, 1H), 7.51 (t, J=7.9 Hz, 1H), 7.39 (dd, J=8.5, 1.9 Hz, 1H), 4.96 (dd, J=14.0, 8.2 Hz, 1H), 3.50 (dd, J=15.6, 5.9 Hz, 1H), 3.37 (d, J=8.7 Hz, 1H), 1.46 (m, 2H), 1.09 (m, 2H). Example 3.10: Preparation of Compound 10 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-1-oxo-3-(5-(trifluoromethyl)benzo[d]oxazol-2-yl)propan-2-yl)benzamide) FIG.23is a schematic diagram illustrating an exemplary procedure for preparing Compound 10 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-4-trifluoromethyl-phenol, the (S)-2-amino-N-(1-cyanocyclopropyl)-3-(5-(trifluoromethyl)benzo[d]oxazol-2-yl)propenamide was obtained as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 339.0 [M+H]+ Following general procedure B-1, from (S)-2-amino-N-(1-cyanocyclopropyl)-3-(5-(trifluoromethyl)benzo[d]oxazol-2-yl)propanamide (60 mg), the desired product Compound 10 was obtained as a white solid (30.8 mg, 36%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 477.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.07 (s, 1H), 9.02 (d, J=7.8 Hz, 1H), 8.08-8.04 (m, 1H), 7.89-7.83 (m, 2H), 7.73 (dd, J=6.8, 5.4 Hz, 2H), 7.62-7.56 (m, 1H), 7.47 (t, J=7.9 Hz, 1H), 4.96 (td, J=8.3, 5.9 Hz, 1H), 3.55-3.32 (m, 2H), 1.43 (m, 2H), 1.06 (m, 2H). Example 3.11: Preparation of Compound 11 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-1-oxo-3-(6-(trifluoromethyl)benzo[d]oxazol-2-yl)propan-2-yl)benzamide) FIG.24is a schematic diagram illustrating an exemplary procedure for preparing Compound 11 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-5-trifluoromethyl-phenol, the (S)-2-amino-N-(1-cyanocyclopropyl)-3-(6-(trifluoromethyl)benzo[d]oxazol-2-yl)propanamide was obtained as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 339.0 [M+H]+ Following general procedure B-1, from (S)-2-amino-N-(1-cyanocyclopropyl)-3-(6-(trifluoromethyl)benzo[d]oxazol-2-yl)propanamide (35 mg), the desired product Compound 11 was obtained as a white solid (25 mg, 51%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 477.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.08 (s, 1H), 9.03 (d, J=7.9 Hz, 1H), 8.16-8.10 (m, 1H), 7.84 (m, 2H), 7.76-7.71 (m, 1H), 7.67 (m, 1H), 7.58 (m, 1H), 7.47 (t, J=7.9 Hz, 1H), 4.96 (td, J=8.3, 6.0 Hz, 1H), 3.52 (m, 1H), 3.41-3.32 (m, 1H), 1.47-1.34 (m, 2H), 1.13-1.05 (m, 2H). Example 3.12: Preparation of Compound 12 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-1-oxo-3-(7-(trifluoromethyl)benzo[d]oxazol-2-yl)propan-2-yl)benzamide) FIG.25is a schematic diagram illustrating an exemplary procedure for preparing Compound 12 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-6-trifluoromethyl-phenol, the (S)-2-amino-N-(1-cyanocyclopropyl)-3-(7-(trifluoromethyl)benzo[d]oxazol-2-yl)propenamide was obtained as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 339.0 [M+H]+. Following general procedure B-1, from (S)-2-amino-N-(1-cyanocyclopropyl)-3-(7-(trifluoromethyl)benzo[d]oxazol-2-yl)propenamide (60 mg), the desired product Compound 12 was obtained as a white solid (35.4 mg, 42%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 476.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.11 (s, 1H), 9.05 (d, J=8.0 Hz, 1H), 8.01 (d, J=7.8 Hz, 1H), 7.87 (d, J=1.8 Hz, 1H), 7.74 (m, 2H), 7.65-7.59 (m, 1H), 7.52 (m, 2H), 4.99 (dd, J=13.5, 8.7 Hz, 1H), 3.58 (dd, J=15.7, 5.4 Hz, 1H), 3.41 (dd, J=15.7, 9.0 Hz, 1H), 1.48 (m, 2H), 1.14 (m, 2H). Example 3.13: Preparation of Compound 13 ((S)—N-(3-(benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-(tert-butyl)-1-methyl-1H-pyrazole-5-carboxamide) FIG.26is a schematic diagram illustrating an exemplary procedure for preparing Compound 13 according to some embodiments of the present disclosure. Following general procedure B-2, from (S)-2-amino-3-(benzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (50 mg), the desired product Compound 13 was purified by prep-HPLC [(Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA); 15%-80%)] and obtained as a white solid (15 mg, 19%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 435.1 [M+H]+.1H NMR (400 MHz, CDCl3) δ 8.19 (s, 1H), 7.99 (d, J=6.9 Hz, 1H), 7.67 (m, 1H), 7.54 (s, 1H), 7.38 (m, 2H), 6.55 (s, 1H), 5.06 (td, J=7.2, 4.0 Hz, 1H), 4.13 (s, 3H), 3.68 (dd, J=16.9, 3.9 Hz, 1H), 3.32 (dd, J=16.9, 7.4 Hz, 1H), 1.55 (s, 2H), 1.34 (d, J=7.1 Hz, 9H), 1.22 (m, 2H). Example 3.14: Preparation of Compound 14 ((S)—N-(3-(benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-6-methylpicolinamide) FIG.27is a schematic diagram illustrating an exemplary procedure for preparing Compound 14 according to some embodiments of the present disclosure. Following general procedure B-2, from (S)-2-amino-3-(benzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (50 mg), the desired product Compound 14 was purified by prep-HPLC [(Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA); 15%-80%)] and obtained as a white solid (6.1 mg, 9%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 390.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.18 (s, 1H), 8.97 (d, J=8.3 Hz, 1H), 7.84 (t, J=7.6 Hz, 1H), 7.78 (d, J=7.3 Hz, 1H), 7.62 (m, 2H), 7.45 (d, J=7.5 Hz, 1H), 7.35-7.26 (m, 2H), 4.95 (dd, J=14.5, 6.4 Hz, 1H), 3.52-3.39 (m, 2H), 2.52 (s, 3H), 1.47-1.38 (m, 2H), 1.11-0.99 (m, 2H). Example 3.15: Preparation of Compound 15 ((S)—N-(3-(benzo[d]oxazol-2-yl)-1-((cyanomethyl)amino)-1-oxopropan-2-yl)-1,3-dicyclopropyl-1H-pyrazole-5-carboxamide) FIG.28is a schematic diagram illustrating an exemplary procedure for preparing Compound 15 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl (S)-3-(benzo[d]oxazol-2-yl)-2-(1,3-dicyclopropyl-1H-pyrazole-5-carboxamido) propanoate To a solution of 1,3-dicyclopropyl-1 H-pyrazole-5-carbonyl chloride (150 mg, 0.73 mmol) and ethyl (2S)-2-amino-3-(1,3-benzoxazol-2-yl) propanoate (170 mg, 0.73 mmol) in DCM (30 mL) was added DIPEA (282 mg, 2.2 mmol). The reaction mixture was stirred at 0° C. for 1 hour. The solvent was removed under reduced pressure and the residue was purified by Prep-Thin Layer Chromatography (TLC) (PE/EA=3/1) to give the product (170 mg, 56%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 408.9 [M+H]+. Step 2. Preparation of (S)-3-(benzo[d]oxazol-2-yl)-2-(1,3-dicyclopropyl-1H-pyrazole-5 carboxamido) propanoic acid To a solution of ethyl (S)-3-(benzo[d]oxazol-2-yl)-2-(1,3-dicyclopropyl-1H-pyrazole-5-carboxamido) propanoate (170 mg, 0.42 mmol) in THF/H2O (5:1, 12 mL) was added LiOH.H2O (175 mg, 4.2 mmol). The reaction mixture was stirred at rt for 1 hour. The reaction solution was adjusted pH to 5-6 by using 1 N aq. HCl. Then the mixture was extracted with EA (20 mL×2). The combined organic layer was washed with brine (10 mL), then dried over with anhydrous Na2SO4. The mixture was filtered, the filtrate was concentrated under vacuum to give the desired product (150 mg, 90%) as yellow oil. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 381.2 [M+H]+. Step 3. Preparation of (S)—N-(3-(benzo[d]oxazol-2-yl)-1-((cyanomethyl)amino)-1-oxopropan-2-yl)-1,3-dicyclopropyl-1H-pyrazole-5-carboxamide To a solution of (S)-3-(benzo[d]oxazol-2-yl)-2-(1,3-dicyclopropyl-1H-pyrazole-5-carboxamido) propanoic acid (150 mg, 0.39 mmol) and 2-aminoacetonitrile hydrochloride (40 mg, 0.44 mmol) in DCM (15 mL) was added DIPEA (255 mg, 2.0 mmol). After stirring for 10 min at 0° C., T3P (50% in EA, 753 mg, 1.2 mmol) was added and the mixture was stirred at rt for 4 hrs. The solvent was removed under reduced pressure and the residue was purified by Combi-flash (PE/EA=0˜ 50%) to give the desired product Compound 15 (35.7 mg, 22%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 419.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.89-8.75 (m, 2H), 7.73-7.62 (m, 2H), 7.43-7.28 (m, 2H), 6.47 (s, 1H), 5.10-5.01 (m, 1H), 4.28-4.13 (m, 3H), 3.54 (dd, J=15.6, 5.2 Hz, 1H), 3.37 (s, 1H), 3.30 (s, 1H), 1.86-1.77 (m, 1H), 1.03-0.76 (m, 6H), 0.59-0.50 (m, 2H). Example 3.16: Preparation of Compound 16 ((S)—N-(3-(benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxamide) FIG.29is a schematic diagram illustrating an exemplary procedure for preparing Compound 16 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl (S)-3-(benzo[d]oxazol-2-yl)-2-(1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxamido)propanoate To a solution of ethyl (S)-2-amino-3-(benzo[d]oxazol-2-yl)propanoate (190 mg, 0.81 mmol) and DIPEA (410 mg, 4.06 mmol) in DCM (5 mL) was added 1-cyclopropyl-3-methyl-1H-pyrazole-4-carbonyl chloride (150 mg, 0.81 mmol). The reaction mixture was stirred at 25° C. for 16 hrs. The mixture was concentrated under reduced pressure to give the crude product as yellow oil (210 mg, 54%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 383.1 [M+H]+. Step 2. Preparation of (S)-3-(benzo[d]oxazol-2-yl)-2-(1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxamido)propanoic acid A solution of ethyl (S)-3-(benzo[d]oxazol-2-yl)-2-(1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxamido)propanoate (230 mg, 0.6 mmol) in THF (2 mL) and 1 N aq.LiOH (2 mL) was stirred at 25° C. for 2 hours. After completion, the mixture was concentrated under vacuum. The residue was dissolved in water (10 mL), adjusted pH to 7 with 1N aq. HCl, and extracted with ethyl acetate (10 mL×2). The combined organic layer was washed with brine (10 mL), dried over Na2SO4, filtered, and concentrated under vacuum to give the desired product as yellow oil (140 mg, 52.56%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 355.1 [M+H]+. Step 3. Preparation of (S)—N-(3-(benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxamide A solution of (S)-3-(benzo[d]oxazol-2-yl)-2-(1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxamido)propanoic acid (140 mg, 0.39 mmol), 1-aminocyclopropane-1-carbonitrile (70 mg, 0.59 mmol), T3P (50% in EA, 754 mg, 2.37 mmol), and DIPEA (255 mg, 1.97 mmol) in DCM (5 mL) was stirred at 25° C. for 16 hours. The residue was diluted with NaHCO3(10 mL) and extracted with DCM (10 mL×2). The combined organic layer was washed with brine (10 mL), dried over Na2SO4, filtered, and concentrated under vacuum, and the residue was purified by Prep-HPLC [Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% TFA), 30-50] to give the desired product Compound 16 as a white solid (38.4 mg, 22%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 419.1 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.05 (s, 1H), 7.67-7.46 (m, 2H), 7.45-7.30 (m, 2H), 4.98 (dd, J=7.8, 6.2 Hz, 1H), 3.62-3.46 (m, 2H), 3.37 (dd, J=15.4, 7.8 Hz, 1H), 3.28 (dt, J=3.2, 1.6 Hz, 3H), 1.44 (dd, J=6.2, 3.4 Hz, 2H), 1.15 (dd, J=18.2, 1.6 Hz, 2H), 1.02-0.99 (m, 4H). Example 3.17: Preparation of Compound 17 ((S)—N-(1-((1-cyanocyclopropyl)amino)-3-(5,6-difluorobenzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-methyl-1H-pyrazole-4-carboxamide) FIG.30is a schematic diagram illustrating an exemplary procedure for preparing Compound 17 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-4,5-difluorophenol, the (S)-2-amino-N-(1-cyanocyclopropyl)-3-(5,6-difluorobenzo[d]oxazol-2-yl)propanamide was obtained as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 306.8 [M+H]+. Following general procedure B-1, from(S)-2-amino-N-(1-cyanocyclopropyl)-3-(5,6-difluorobenzo [d]oxazol-2-yl)propanamide (83 mg), the desired product Compound 17 was obtained as a white solid (10 mg, 8%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 454.8 [M+H]+.1H NMR (400 MHz, MeOD) δ 8.10 (s, 1H), 7.61 (ddd, J=17.4, 9.6, 7.2 Hz, 2H), 5.02 (d, J=6.2 Hz, 1H), 3.61 (d, J=5.4 Hz, 1H), 3.55 (dd, J=15.4, 6.2 Hz, 1H), 3.40 (dd, J=15.6, 7.8 Hz, 1H), 2.33 (s, 3H), 1.50 (d, J=2.2 Hz, 2H), 1.23 (dd, J=8.2, 2.2 Hz, 2H), 1.06 (d, J=5.6 Hz, 4H). Example 3.18: Preparation of Compound 18 ((S)—N-(3-(benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(trifluoromethyl)-1H-pyrazole-4-carboxamide) FIG.31is a schematic diagram illustrating an exemplary procedure for preparing Compound 18 according to some embodiments of the present disclosure. Following general procedure B-2, from (S)-2-amino-3-(benzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (81 mg), the desired product Compound 18 was obtained as a white solid (38 mg, 22%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 473.1 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.24 (s, 1H), 7.60 (s, 1H), 7.54 (d, J=0.8 Hz, 1H), 7.39-7.27 (m, 2H), 4.97 (d, J=1.0 Hz, 1H), 3.80-3.67 (m, 1H), 3.49 (d, J=6.4 Hz, 1H), 3.40 (d, J=7.8 Hz, 1H), 1.44 (d, J=3.0 Hz, 2H), 1.18-1.04 (m, 6H). Example 3.19: Preparation of Compound 19 ((S)—N-(3-(benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(difluoromethyl)-1 H-pyrazole-4-carboxamide) FIG.32is a schematic diagram illustrating an exemplary procedure for preparing Compound 19 according to some embodiments of the present disclosure. Following general procedure B-2, from (S)-2-amino-3-(benzo[d]oxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (50 mg), the desired product Compound 19 was obtained as a white solid (14.5 mg, 17%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 455.1 [M+H]+. NMR (400 MHz, DMSO-d6) δ 8.22 (s, 1H), 7.62-7.51 (m, 2H), 7.38-7.29 (m, 2H), 7.02 (t, J=54.0 Hz, 1H), 4.99 (s, 1H), 3.76-3.68 (m, 1H), 3.50 (d, J=6.2 Hz, 1H), 3.40 (d, J=7.8 Hz, 1H), 1.44 (s, 2H), 1.19-1.02 (m, 6H). Example 3.20: Preparation of Compound 20 ((S)-3-(tert-butyl)-N-(3-(7-chlorobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-1H-pyrazole-5-carboxamide) FIG.33is a schematic diagram illustrating an exemplary procedure for preparing Compound 20 according to some embodiments of the present disclosure. Following general procedure B-3, from (2S)-2-amino-3-(7-chloro-5-fluoro-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (200 mg), the desired product Compound 20 was obtained as a white solid (37 mg, 19%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 495.2 [M+H]+.1H NMR (400 MHz, MeOD) δ 7.58 (dd, J=7.8, 1.2 Hz, 1H), 7.39 (dd, J=8.0, 1.2 Hz, 1H), 7.33 (t, J=7.8 Hz, 1H), 6.62 (s, 1H), 5.05 (dd, J=8.6, 5.6 Hz, 1H), 3.98-3.91 (m, 1H), 3.62 (dd, J=15.6, 5.8 Hz, 1H), 3.45 (dd, J=15.4, 8.6 Hz, 1H), 1.49 (t, J=5.6 Hz, 2H), 1.28 (s, 1H), 1.25 (s, 9H), 1.23 (d, J=2.6 Hz, 1H), 1.06 (dd, J=6.4, 3.8 Hz, 1H), 1.00-0.95 (m, 1H), 0.92-0.82 (m, 2H). Example 3.21: Preparation of Compound 21 ((S)-3-(tert-butyl)-N-(3-(7-chloro-5-fluorobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-1 H-pyrazole-5-carboxamide) FIG.34is a schematic diagram illustrating an exemplary procedure for preparing Compound 21 according to some embodiments of the present disclosure. Following general procedure A, from 2-amino-6-chloro-4-fluorophenol, the (2S)-2-amino-3-(7-chloro-5-fluoro-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propanamide was obtained as a yellow oil. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 323.2 [M+H]+. Following general procedure B-3, from (2S)-2-amino-3-(7-chloro-5-fluoro-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (49 mg), the desired product Compound 21 was obtained as a white solid (4.7 mg, 3%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass (m/z):513.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.12 (s, 1H), 8.80 (d, J=8.0 Hz, 1H), 7.64 (dd, J=8.4, 2.2 Hz, 1H), 7.55 (dd, J=9.4, 2.0 Hz, 1H), 6.69 (s, 1H), 4.92 (d, J=5.8 Hz, 1H), 4.19 (dd, J=7.4, 3.8 Hz, 1H), 3.53 (dd, J=15.4, 5.4 Hz, 1H), 1.48 (t, J=4.1 Hz, 2H), 1.37-1.08 (m, 12H), 1.04 (dd, J=10.0, 6.2 Hz, 1H), 0.94 (dd, J=9.8, 5.8 Hz, 1H), 0.90-0.75 (m, 2H). Example 3.22: Preparation of Compound 22 ((S)-3-(tert-butyl)-N-(3-(7-chloro-5-fluorobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-(oxetan-3-yl)-1H-pyrazole-5-carboxamide) FIG.35is a schematic diagram illustrating an exemplary procedure for preparing Compound 22 according to some embodiments of the present disclosure. Following general procedure B-3, from (2S)-2-amino-3-(7-chloro-5-fluoro-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide 100 mg), the desired product Compound 22 was obtained as a white solid (6 mg, 4%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass (m/z): 529.2[M+H]+.1HNMR (400 MHz, DMSO-d6) δ 9.15 (s, 1H), 8.96 (d, J=4.8 Hz, 1H), 7.65-7.55 (m, 2H), 6.85 (s, 1H), 5.88-5.79 (m, 1H), 4.75-4.95 (m, 5H), 3.55-3.48 (m, 1H), 1.55-1.42 (m, 2H), 1.38-1.55 (m, 10H), 1.19-1.05 (m, 2H). Example 3.23: Preparation of Compound 23 ((S)—N-(3-(4-(1H-pyrazol-4-yl)benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide) FIG.36is a schematic diagram illustrating an exemplary procedure for preparing Compound 23 according to some embodiments of the present disclosure. Step 1. Preparation of tert-butyl (S)-4-(2-(2-((tert-butoxycarbonyl)amino)-3-ethoxy-3-oxopropyl)benzo[d] oxazol-4-yl)-1H-pyrazole-1-carboxylate A solution of ethyl (S)-3-(4-bromobenzo[d]oxazol-2-yl)-2-((tert-butoxycarbonyl)amino)propanoate (450 mg, 1.09 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole-1-carboxylate (384 mg, 1.31 mmol), PdCl2(dppf (70.65 mg, 0.10 mmol), Na2CO3(231 mg, 2.18 mmol) in 1,4-dioxane/H2O (12 mL/2 mL) was stirred at 90° C. for 1 h. The reaction mixture was quenched with water and extracted with EA (60 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum to give a crude product, which was purified by flash chromatography (MeOH in DCM, 2% to 10%) to obtain the desired product (200 mg, 74%) as a pale solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 501.1 [M+H]+. Step 2. Preparation of (S)-3-(4-(1-(tert-butoxycarbonyl)-1 H-pyrazol-4-yl)benzo[d]oxazol-2-yl)-2-((tert-butoxy carbonyl)amino)propanoic acid A solution of tert-butyl (S)-4-(2-(2-((tert-butoxycarbonyl)amino)-3-ethoxy-3-oxopropyl)benzo[d]oxazol-4-yl)-1H-pyrazole-1-carboxylate (350 mg, 0.7 mmol), LiOH (19.06 mg, 0.80 mmol) in THF/H2O (15 mL/4 mL) was stirred at 25° C. for 1 h. The reaction mixture was quenched with water and extracted with EA (60 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum to give the desired product (280 mg, 85%) as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 473.1 [M+H]+. Step 3. Preparation of tert-butyl (S)-4-(2-(2-((tert-butoxycarbonyl)amino)-3-((1-cyanocyclopropyl)amino)-3-oxopropyl)benzo[d]oxazol-4-yl)-1H-pyrazole-1-carboxylate A solution of (S)-3-(4-(1-(tert-butoxycarbonyl)-1H-pyrazol-4-yl)benzo[d]oxazol-2-yl)-2-((tert-butoxy carbonyl)amino)propanoic acid (280 mg, 0.59 mmol), A-5 (84 mg, 0.71 mmol), DIPEA (228 mg, 1.77 mmol) and T3P (50% in EA, 402 mg, 0.63 mmol) in DCM (5 mL) was stirred at 25° C. under N2for 3 hrs. The reaction mixture was quenched with water and extracted with DCM (30 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum to give a crude product, which was purified by flash chromatography (MeOH in DCM, 2% to 10%) to obtain the desired product (130 mg, 41%) as a pale solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 537.1 [M+H]+. Step 4. Preparation of (S)-3-(4-(1H-pyrazol-4-yl)benzo[d]oxazol-2-yl)-2-amino-N-(1-cyanocyclopropyl) propanamide A solution of tert-butyl (S)-4-(2-(2-((tert-butoxycarbonyl)amino)-3-((1-cyanocyclopropyl)amino)-3-oxopropyl)benzo[d]oxazol-4-yl)-1H-pyrazole-1-carboxylate (130 mg, 0.24 mmol) in FA (2 mL) was stirred at 25° C. for 3 hrs. The mixture was concentrated under reduced pressure to give a residue. The residue was quenched with Sat. NaHCO3and extracted with EtOAc (60 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum to give the crude desired product (30 mg, 37%) as a light-yellow solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 437.1 [M+H]+. Step 5. Preparation of (S)—N-(3-(4-(1 H-pyrazol-4-yl)benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide To a solution of (S)-3-(4-(1 H-pyrazol-4-yl)benzo[d]oxazol-2-yl)-2-amino-N-(1-cyanocyclopropyl) propenamide (30 mg, 0.09 mmol) and K2CO3(24.66 mg, 0.18 mmol) in EA (2 mL) and H2O (3 mL) was added a solution of 3-chlorobenzoyl chloride (18.73 mg, 0.11 mmol) in EA (0.5 mL) dropwise. After the addition, the reaction mixture was stirred at 25° C. for 0.5 h. Then the reaction mixture was quenched with water and extracted with EA (20 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum to give a crude product, which was purified by prep-HPLC [(Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA); 15%-95%)] to give the desired product Compound 23 (4.1 mg, 10%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 475.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 13.18 (s, 1H), 9.15 (s, 1H), 9.10 (d, J=7.8 Hz, 1H), 8.43 (s, 2H), 8.19-8.12 (m, 1H), 7.86 (s, 1H), 7.76 (d, J=8.0 Hz, 1H), 7.64 (d, J=6.7 Hz, 1H), 7.58 (d, J=9.1 Hz, 1H), 7.45 (s, 1H), 7.31 (t, J=7.8 Hz, 1H), 5.06 (d, J=4.1 Hz, 1H), 3.55 (d, J=4.2 Hz, 1H), 3.45 (s, 1H), 1.47 (m, 2H), 1.17-1.06 (m, 2H). Example 3.24: Preparation of Compound 24 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)benzamide) FIG.37is a schematic diagram illustrating an exemplary procedure for preparing Compound 24 according to some embodiments of the present disclosure. Step 1. Preparation of ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanoate Six mixtures of ethyl (S)-3-(6-bromobenzo[d]oxazol-2-yl)-2-((tert-butoxycarbonyl)amino) propanoate (50 mg, 0.73 mmol), 1-methylpiperazine (73 mg, 0.73 mmol), Cs2CO3(474.5 mg, 1.46 mmol) and RuPhos-Pd-G2 (57 mg, 0.073 mmol) in toluene (5 mL) under nitrogen were stirred at 110° C. for 18 hrs. The reaction mixtures were cooled to room temperature, combined, concentrated under vacuum to give a crude product. The crude product was then purified on silica gel, eluting with MeOH in DCM (5% to 10%) to give the desired product (180 mg, 57%) as a light brown oil. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 433.1 [M+H]+. Step 2. Preparation of (S)-2-((tert-butoxycarbonyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanoic acid A solution of ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanoate (160 mg, 0.37 mmol) and Me3SnOH (134 mg, 0.74 mmol) in DCE (6 mL) under nitrogen was heated to 80° C. and stirred for 3 hrs. After concentration, the crude product was then purified on silica gel, eluted with MeOH in DCM (5% to 10%) to give the desired product (80 mg, 53.3%) as a light brown solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 405.1 [M+H]+. Step 3. Preparation of tert-butyl (S)-(1-((1-cyanocyclopropyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)carbamate A solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d] oxazol-2-yl)propanoic acid (70 mg, 0.17 mmol), A-5 (25 mg, 0.21 mmol), HATU (97 mg, 0.26 mmol), and DIPEA (66 mg, 0.51 mmol) in DMF (3 mL) was stirred under nitrogen at 25° C. for 3 hrs. The reaction mixture was quenched with water (10 mL) and extracted with EA (35 mL×3). The combined organic layer was dried over Na2SO4and concentrated to dryness and then purified on silica gel, eluted with MeOH in DCM (5% to 10%) to obtain the desired product (60 mg, 75%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 469.1 [M+H]+. Step 4. Preparation of (S)-2-amino-N-(1-cyanocyclopropyl)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanamide To a solution of tert-butyl (S)-(1-((1-cyanocyclopropyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)carbamate (60 mg, 0.13 mmol) in FA (2 mL) was stirred under nitrogen at 25° C. for 5 hrs. The mixture was blown by nitrogen to dryness at 20° C. Then the residue was basified with saturated sodium bicarbonate and extracted with EA (20 mL×3). The combined organic layer was concentrated to dryness to give the desired product (30 mg, 63.8%) as a light white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 369.1 [M+H]+. Step 5. Preparation of (S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)benzamide To a solution of (S)-2-amino-N-(1-cyanocyclopropyl)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanamide (30 mg, 0.08 mmol) in EA (3 mL) was added K2CO3(22 mg, 0.16 mmol) in water (0.5 mL) and then followed by 3-chlorobenzoyl chloride (14 mg, 0.08 mmol) in EA (1 mL) dropwise, after the addition, the reaction mixture was stirred under at 25° C. for 1 h. The reaction mixture was quenched with water (5 mL) and extracted with EA (15 mL×3). The combined organic layer was concentrated to dryness, and the residue was purified by prep-HPLC [(Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA); 15%-95%)] to give the desired product Compound 24 (4.9 mg, 12%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 507.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.08 (s, 1H), 9.00 (d, J=7.9 Hz, 1H), 7.89 (t, J=1.8 Hz, 1H), 7.78 (d, J=7.8 Hz, 1H), 7.64-7.59 (m, 1H), 7.51 (t, J=7.9 Hz, 1H), 7.44 (d, J=8.8 Hz, 1H), 7.14 (d, J=2.2 Hz, 1H), 6.97 (dd, J=8.8, 2.3 Hz, 1H), 4.94-4.88 (m, 1H), 3.43 (d, J=5.7 Hz, 1H), 3.26 (d, J=8.8 Hz, 1H), 3.17-3.11 (m, 4H), 2.48-2.43 (m, 4H), 2.22 (s, 3H), 1.46-1.47 (m, 2H), 1.13-1.02 (m, 2H). Example 3.25: Preparation of Compound 25 ((S)—N-(1-((cyanomethyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1,3-dicyclopropyl-1 H-pyrazole-5-carboxamide) FIG.38is a schematic diagram illustrating an exemplary procedure for preparing Compound 25 according to some embodiments of the present disclosure. Step 1. Preparation of tert-butyl (S)-(1-((cyanomethyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)carbamate To a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanoic acid (500 mg, 1.24 mmol) in DMF (20 mL) was added DIPEA (798 mg, 6.2 mmol), HATU (1410 mg, 3.72 mmol), and 2-aminoacetonitrile (114 mg, 1.86 mmol). The reaction mixture was stirred at RT for 2 hrs. After the reaction completed, H2O (30 mL) was added to the reaction mixture, and then extracted with EA (30 mL×3). The combined organic layer was washed with brine (50 mL×3), then dried over anhydrous Na2SO4. After filtration, the solution was concentrated under vacuum, and the residue was purified by Combi-flash (DCM/MeOH=0˜10%) to give the product tert-butyl (S)-(1-((cyanomethyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)carbamate as yellow oil (310 mg, 56%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 442.9 [M+H]+. Step 2. Preparation of (S)-2-amino-N-(cyanomethyl)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanamide A solution of tert-butyl (S)-(1-((cyanomethyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)carbamate (310 mg, 0.7 mmol) in FA (5 mL) was stirred at RT for 3 hrs. The resulting mixture was blown dry with compressed air to give the product (S)-2-amino-N-(cyanomethyl)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanamide as brown oil (160 mg, 66%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 343.1 [M+H]+. Step 3. Preparation of (S)—N-(1-((cyanomethyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1,3-dicyclopropyl-1H-pyrazole-5-carboxamide Following general procedure B-3, from(S)-2-amino-N-(cyanomethyl)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanamide (70 mg), the desired product Compound 25 was obtained as a white solid (5 mg, 4%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 517.3 [M+H]+.1H NMR (400 MHz, MeOD) δ 7.55 (d, J=8.8 Hz, 1H), 7.26 (d, J=2.2 Hz, 1H), 7.12 (dd, J=8.8, 2.4 Hz, 1H), 6.39 (s, 1H), 5.16 (dd, J=8.8, 5.6 Hz, 1H), 4.21 (s, 2H), 3.93 (ddd, J=11.2, 7.4, 4.0 Hz, 2H), 3.62 (dd, J=15.6, 5.6 Hz, 2H), 3.39 (dd, J=15.4, 8.8 Hz, 2H), 3.00 (s, 3H), 1.87 (ddd, J=13.4, 8.6, 5.0 Hz, 1H), 1.05 (dd, J=9.0, 4.8 Hz, 1H), 0.98-0.78 (m, 6H), 0.65 (dt, J=6.2, 4.2 Hz, 2H). Example 3.26: Preparation of Compound 26 ((S)-3-(tert-butyl)-N-(1-((1-cyanocyclopropyl)amino)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-1 H-pyrazole-5-carboxamide) FIG.39is a schematic diagram illustrating an exemplary procedure for preparing Compound 26 according to some embodiments of the present disclosure. Following general procedure B-3, from (S)-2-amino-N-(1-cyanocyclopropyl)-3-(6-(4-methylpiperazin-1-yl)benzo[d]oxazol-2-yl)propanamide (80 mg), the desired product Compound 26 was obtained as a white solid (14 mg, 11%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 559.3 [M+H]+.1H NMR (400 MHz, MeOD) δ 7.43 (d, J=8.8 Hz, 1H), 7.14 (d, J=2.2 Hz, 1H), 7.01 (dd, J=8.8, 2.4 Hz, 1H), 6.50 (s, 1H), 4.91 (dd, J=8.2, 6.0 Hz, 1H), 3.88-3.79 (m, 1H), 3.45 (dd, J=15.4, 6.2 Hz, 2H), 3.29 (d, J=8.2 Hz, 4H), 2.83 (s, 3H), 1.39 (d, J=2.6 Hz, 2H), 1.19 (d, J=1.6 Hz, 2H), 1.15 (s, 9H), 1.14-1.09 (m, 2H), 1.02-0.69 (m, 6H). Example 3.27: Preparation of Compound 27 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-3-(6-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)benzamide) FIG.40is a schematic diagram illustrating an exemplary procedure for preparing Compound 27 according to some embodiments of the present disclosure. To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide (100 mg, 0.20 mmol) in dioxane/H2O (10:1, 5 mL) was added 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine (91 mg, 0.41 mmol), K3PO4(87 mg, 0.411 mmol), and Pd(dppf)Cl2(15 mg, 0.02 mmol). The solution was stirred at 80° C. under N2for 2 hrs. After filtration, the solution was concentrated under vacuum, and the crude product was purified by flash chromatography to give the desired product Compound 27 (18.6 mg, 17.27%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass (m/z): 504.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.09 (s, 1H), 9.03 (d, J=7.8 Hz, 1H), 7.89 (t, J=1.8 Hz, 1H), 7.78 (d, J=7.9 Hz, 1H), 7.67 (d, J=1.3 Hz, 1H), 7.64-7.60 (m, 1H), 7.58 (d, J=8.3 Hz, 1H), 7.52 (d, J=7.9 Hz, 1H), 7.43 (dd, J=8.4, 1.6 Hz, 1H), 6.21 (s, 1H), 4.96 (dd, J=14.0, 8.3 Hz, 1H), 3.51-3.35 (m, 4H), 3.02 (d, J=3.0 Hz, 2H), 2.57 (d, J=4.8 Hz, 2H), 2.28 (s, 3H), 1.47 (d, J=2.8 Hz, 2H), 1.12-1.06 (m, 2H). Example 3.28: Preparation of Compound 28 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-3-(6-(1-(2,2-difluoroethyl)-1,2,3,6-tetrahydropyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)benzamide) FIG.41is a schematic diagram illustrating an exemplary procedure for preparing Compound 28 according to some embodiments of the present disclosure. To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide (130 mg, 0.47 mmol) in dioxane/H2O (10/1, 10 mL) was added 1-(2,2-difluoroethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,6-tetrahydropyridine (84 mg, 0.57 mmol), K3PO4(113 mg, 0.94 mmol), and Pd(dppf)Cl2(20 mg, 0.05 mmol). The reaction mixture was stirred at 100° C. under N2for 16 hrs. The solvent was removed under reduced pressure and the residue was purified by Combi-flash (DCM/MeOH=0˜ 10%) to give the desired product Compound 28 as a yellow solid (35 mg, 13%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 554.2 [M+H]+.1H NMR (400 MHz, MeOD) δ 7.83 (t, J=1.8 Hz, 1H), 7.75-7.71 (m, 1H), 7.62 (d, J=1.4 Hz, 1H), 7.58-7.53 (m, 2H), 7.49-7.43 (m, 2H), 6.18 (d, J=5.6 Hz, 1H), 6.04 (s, 1H), 5.05 (dd, J=8.2, 6.4 Hz, 1H), 4.91 (d, J=1.8 Hz, 1H), 4.85 (s, 1H), 3.59 (dd, J=15.4, 6.4 Hz, 1H), 3.44 (dd, J=15.4, 8.2 Hz, 1H), 3.33 (d, J=3.4 Hz, 2H), 2.95-2.86 (m, 4H), 1.48 (dd, J=5.2, 2.2 Hz, 2H), 1.19 (dd, J=17.0, 2.4 Hz, 2H). Example 3.29: Preparation of Compound 29 ((S)-3-(tert-butyl)-N-(1-((1-cyanocyclopropyl)amino)-3-(6-(1-(2,2-difluoroethyl)-1,2,3,6-tetrahydropyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-1 H-pyrazole-5-carboxamide) FIG.42is a schematic diagram illustrating an exemplary procedure for preparing Compound 29 according to some embodiments of the present disclosure. Step 1. Preparation of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl) amino)-1-oxopropan-2-yl)-3-(tert-butyl)-1-cyclopropyl-1H-pyrazole-5-carboxamide To a solution of (2S)-2-amino-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (1.6 g, 4.58 mmol) and 5-tert-butyl-2-cyclopropyl pyrazole-3-carboxylic acid (954 mg, 4.58 mmol) in DCM (20 mL) was added T3P (50% in EA, 5.83 g, 9.16 mmol) and DIPEA (1.18 g, 9.16 mmol). The reaction mixture was stirred at 25° C. under N2for 3 hrs. The reaction mixture was concentrated and the residue was purified by silica gel column chromatography (PE:EA=1:1) to give the product as a yellow solid (1.3 g, 47%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 539.1 541.1[M+H]+. Step 2. Preparation of (S)-3-(tert-butyl)-N-(1-((1-cyanocyclopropyl)amino)-3-(6-(1-(2,2-difluoroethyl)-1,2,3,6-tetrahydropyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-1H-pyrazole-5-carboxamide To a solution of (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-2-[(5-tert-butyl-2-cyclopropylpyrazol-3-yl)formamido]-N-(1-cyanocyclopropyl)propenamide (150 mg, 0.28 mmol) and 1-(2,2-difluoroethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine (114 mg, 0.42 mmol) in dioxane (3 mL) and H2O (0.3 mL) was added K3PO4(118 mg, 0.56 mmol) and Pd(dppf)Cl2(20 mg, 0.028 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The reaction mixture was concentrated and the residue was purified by Pre-HPLC [Gemini-C18 150×21.2 mm, Sum, mobile phase: ACN-H2O (0.1% FA), gradient: 20-40] to give the desired product Compound 29 as a white solid (30 mg, 19%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 606.3[M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.13 (s, 1H), 8.79 (d, J=8.0 Hz, 1H), 7.73 (s, 1H), 7.62 (d, J=8.2 Hz, 1H), 7.47 (d, J=8.4 Hz, 1H), 6.69 (s, 1H), 6.23 (s, 1H), 4.99-4.88 (m, 1H), 4.23-4.15 (m, 1H), 3.49 (dd, J=15.4, 5.8 Hz, 1H), 3.39-3.29 (m, 6H), 3.09-2.56 (m, 4H), 1.53-1.40 (m, 2H), 1.24-1.17 (m, 9H), 1.16-1.07 (m, 2H), 1.06-1.01 (m, 1H), 0.97-0.91 (m, 1H), 0.87-0.78 (m, 2H). Example 3.30: Preparation of Compound 30 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-3-(6-(4-methyl-2-oxopiperazin-1-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)benzamide) FIG.43is a schematic diagram illustrating an exemplary procedure for preparing Compound 30 according to some embodiments of the present disclosure. To a mixture of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide (200 mg, 0.4 mmol) in dioxane (10.0 mL) was added K2CO3(113 mg, 0.8 mmol), 4-methylpiperazin-2-one (93 mg, 0.8 mmol), Cul (15 mg, 0.08 mmol), and DMEDA (7 mg, 0.08 mmol). The reaction mixture was degassed with N2and stirred at 110° C. for 16 hrs. The reaction mixture was concentrated under reduced pressure and the residue was purified by Prep-HPLC [Gemini-C18 150×21.2 mm, Sum; Mobile phase: MeCN/H2O (0.1% FA); Ratio: 10-25] to give the desired product Compound 30 (12 mg, 6%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 521.1 [M+H]+.1H NMR (400 MHz, MeOD) δ 7.84 (t, J=1.8 Hz, 1H), 7.75-7.72 (m, 1H), 7.68 (d, J=8.5 Hz, 1H), 7.59 (d, J=1.6 Hz, 1H), 7.57-7.54 (m, 1H), 7.45 (t, J=7.8 Hz, 1H), 7.29 (dd, J=8.4, 1.8 Hz, 1H), 5.06 (dd, J=8.2, 6.2 Hz, 1H), 3.79-3.76 (m, 2H), 3.61 (dd, J=15.4, 6.2 Hz, 1H), 3.47 (dd, J=15.4, 8.2 Hz, 1H), 3.29 (s, 2H), 2.89 (t, J=5.4 Hz, 2H), 2.44 (s, 3H), 1.52-1.44 (m, 2H), 1.26-1.16 (m, 2H). Example 3.31: Preparation of Compound 31 ((S)—N-(3-(benzo[d]thiazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide) FIG.44is a schematic diagram illustrating an exemplary procedure for preparing Compound 31 according to some embodiments of the present disclosure. Step 1. Preparation of allyl (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(benzo[d]thiazol-2-yl)propanoate To a solution of (S)-3-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-4-(allyloxy)-4-oxobutanoic acid (1 g, 2.53 mmol), (COCl)2(0.38 g, 3.0 mmol) in DCM (25 mL) was added 1 drop DMF and then the reaction mixture was stirred at 25° C. under N2for 2 h. Then the solvent was removed under vacuum to give a residue, the residue was dissolved in anhydrous toluene (15 mL). After that, 2-aminobenzenethiol (0.33 g, 2.6 mmol) and DIPEA (0.97 g, 7.5 mmol) was added and the reaction mixture was heated to 40° C. and stirred for 2 hours. After cooled to room temperature, the reaction mixture was removed and then quenched with water (30 mL) and extracted with DCM (120 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum to give the crude desired product, which was purified by flash chromatography (EA in PE, 30% to 50%) to give the desired product (0.47 g, 32%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 485.1 [M+H]+. Step 2. Preparation of (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(benzo[d]thiazol-2-yl)propanoic acid To a solution of allyl (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(benzo[d]thiazol-2-yl) propanoate (470 mg, 0.96 mmol) in DCM (12 mL) was added phenylsilane (215.43 mg, 2.9 mmol) and followed by tetrakis(triphenylphosphine)palladium (559.23 mg, 0.48 mmol) under nitrogen protection. After the addition, the reaction mixture was stirred at 25° C. for 3 h. The solvent was removed under vacuum to give a crude product, which was purified by flash chromatography (MeOH in DCM, 2%-10%) to obtain the desired product (350 mg, 81%) as a pale solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 445.1 [M+H]+. Step 3. Preparation of (9H-fluoren-9-yl)methyl (S)-(3-(benzo[d]thiazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)carbamate A solution of (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(benzo[d]thiazol-2-yl)propanoic acid (350 mg, 0.78 mmol), 1-aminocyclopropane-1-carbonitrile hydrogen chloride (67.73 mg, 0.82 mmol), DIPEA (203.06 mg, 0.57 mmol) and HATU (358.45 mg, 0.45 mmol) in DMF (5 mL) was stirred at 25° C. under N2for 3 hrs. The reaction mixture was quenched with water and extracted with EA (30 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum to give a crude product, which was purified by flash chromatography (MeOH in DCM, 2% to 10%) to obtain the desired product (90 mg, 20%) as a pale solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 509.1 [M+H]+. Step 4. Preparation of (S)-2-amino-3-(benzo[d]thiazol-2-yl)-N-(1-cyanocyclopropyl)propanamide To a solution of (9H-fluoren-9-yl)methyl (S)-(3-(benzo[d]thiazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)carbamate (90 mg, 0.17 mmol) in MeCN (5 mL) was added Et2NH (1 mL). The reaction mixture was stirred at 25° C. under N2for 3 hrs. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC [(Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA); 15-90)] to give the desired product (26 mg, 54%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 287 [M+H]+. Step 5. Preparation of (S)—N-(3-(benzo[d]thiazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-chlorobenzamide To a solution of (S)-2-amino-3-(benzo[d]thiazol-2-yl)-N-(1-cyanocyclopropyl)propanamide and DIPEA (16.25 mg, 0.12 mmol) in DCM (1 mL) was added a solution of 3-chlorobenzoyl chloride (7.7 mg, 0.04 mmol) in DCM (0.5 mL) dropwise. After the addition, the reaction mixture was stirred at 25° C. for 0.5 h. Then the reaction mixture was quenched with water and extracted with DCM (20 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum to give a crude product, which was purified by prep-HPLC [(Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA); 15%-90%)] to give the desired product Compound 31 (11.2 mg, 31%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 425.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.63 (d, J=6.4 Hz, 1H), 8.24 (s, 1H), 8.01-7.97 (m, 1H), 7.92 (t, J=1.8 Hz, 1H), 7.88 (d, J=7.8 Hz, 1H), 7.80-7.77 (m, 1H), 7.55-7.49 (m, 2H), 7.41 (m, 2H), 5.03 (m, 1H), 3.83 (m, 1H), 3.49-3.43 (m, 1H), 1.53-1.47 (m, 2H), 1.23-1.13 (m, 2H). Example 3.32: Preparation of Compound 32 ((S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-3-(1-methyl-1 H-benzo[d]imidazol-2-yl)-1-oxopropan-2-yl)benzamide) FIG.45is a schematic diagram illustrating an exemplary procedure for preparing Compound 32 according to some embodiments of the present disclosure. Step 1. Preparation of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(1-methyl-1 H-benzo[d]imidazol-2-yl)propanoate A mixture of (S)-3-((tert-butoxycarbonyl)amino)-4-methoxy-4-oxobutanoic acid (1.3 g, 5.26 mmol), N1-methylbenzene-1,2-diamine (284 mg, 5.26 mmol), HOBt (858 mg, 6.31 mmol), EDCl (1.21 g, 6.31 mmol), Et3N (1.59 g, 15.78 mmol) in DCM (20 mL) under nitrogen was stirred for 18 hrs at 25° C. The reaction mixture was quenched with 15 mL of water and extracted with DCM (30 mL×3). The combined organic layer was dried over Na2SO4and concentrated under vacuum, purified on silica gel (EtOAc in PE, 50% to 100%) to give a mixture of NH and NMe-amides, as a light brown oil. This mixture (1.18 mg, 3.4 mmol) in AcOH (10 mL) under nitrogen was heated to 60° C. and stirred for 18 h. The reaction was cooled to room temperature and the solvent was removed to give a residue, the residue was dissolved with EtOAc (30 mL) and basified with Sat. NaHCO3(20 mL), the organic layer was separated, and the aqueous layer was extracted with EtOAc (30 mL×3). The combined organic layer was washed with brine and dried over anhydrous Na2SO4. After filtration and concentration, the crude product was then purified on silica gel, eluted with EtOAc in petroleum ether (35% to 100%) to give the desired product (700 mg, 62.5%) as a light-yellow oil. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 334.1 [M+H]+. Step 2. Preparation of (S)-2-((tert-butoxycarbonyl)amino)-3-(1-methyl-1H-benzo[d]imidazol-2-yl)propanoic acid To a solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(1-methyl-1 H-benzo[d]imidazol-2-yl)propanoate (500 mg, 1.50 mmol) in THF (8 mL) was added a solution of LiOH (72 mg, 3.0 mmol) in water (1.5 mL) slowly. After the addition, the reaction mixture was stirred for 5 hrs. Then the reaction mixture was acidified with HCl (1M) to pH=3 and extracted with EtOAc (30 mL×3). The combined organic layer was washed with brine and dried over anhydrous Na2SO4. The solvent was removed under vacuum to give the crude product (330 mg, 69%) as a light brown solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 320.1 [M+H]+. Step 3. Preparation of tert-butyl (S)-(1-((1-cyanocyclopropyl)amino)-3-(1-methyl-1H-benzo[d]imidazol-2-yl)-1-oxopropan-2-yl)carbamate A solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(1-methyl-1H-benzo[d]imidazol-2-yl)propanoic acid (330 mg, 1.03 mmol), A-5 (147 mg, 1.24 mmol), HATU (471 mg, 1.24 mmol), and DIPEA (401.7 mg, 3.09 mmol) in DMF (5 mL) was stirred under nitrogen at 25° C. for 3 hrs. The reaction mixture was quenched with water (10 mL) and extracted with EA (35 mL×3). The combined organic layer was dried over Na2SO4and concentrated to dryness and then purified on silica gel, eluted with (EA in PE, 30% to 100%) to obtain the desired product (250 mg, 63.4%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 384.1 [M+H]+. Step 4. Preparation of (S)-2-amino-N-(1-cyanocyclopropyl)-3-(1-methyl-1 H-benzo[d]imidazol-2-yl)propanamide A solution of tert-butyl (S)-(1-((1-cyanocyclopropyl)amino)-3-(1-methyl-1H-benzo[d]imidazol-2-yl)-1-oxopropan-2-yl)carbamate (70 mg, 0.18 mmol) in FA (2 mL) was stirred under nitrogen at 25° C. for 5 hrs. The mixture was blown by nitrogen to dryness at 20° C. Then the residue was basified with saturated sodium bicarbonate and extracted with EA (20 mL×3). And then the combined organic layer was concentrated to dryness to give the desired product (22 mg, 43%) as a pale solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 284.1 [M+H]+. Step 5. Preparation of (S)-3-chloro-N-(1-((1-cyanocyclopropyl)amino)-3-(1-methyl-1H-benzo[d]imidazol-2-yl)-1-oxopropan-2-yl)benzamide To a solution of (S)-2-amino-N-(1-cyanocyclopropyl)-3-(1-methyl-1H-benzo[d]imidazol-2-yl)propanamide (22 mg, 0.08 mmol) and DIPEA (20.64 mg, 0.16 mmol) in DCM (2 mL) was added a solution of 3-chlorobenzoyl chloride (14 mg, 0.08 mmol) in DCM (0.5 mL) dropwise, after the addition, the reaction mixture was stirred at 25° C. for 1 h. The reaction mixture was quenched with water (5 mL) and extracted with DCM (15 mL×3). The combined organic layer was concentrated to dryness, and the residue was purified by prep-HPLC [(Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA); 15%-95%)] to give the desired product Compound 32 (4.2 mg, 12.4%) as a white solid. Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 422.0 [M+H]+.1H NMR (400 MHz, DMSO-d) 6 9.01 (s, 1H), 8.98 (d, J=7.6 Hz, 1H), 7.91 (s, 1H), 7.79 (d, J=7.9 Hz, 1H), 7.61 (d, J=9.0 Hz, 1H), 7.51 (t, J=7.8 Hz, 2H), 7.17 (m, 2H), 4.96 (dd, J=14.3, 7.6 Hz, 1H), 3.78 (s, 3H), 3.37 (dd, J=16.5, 6.6 Hz, 2H), 1.47-1.36 (m, 2H), 1.02 (m, 2H). Example 3.33: Preparation of Compound 33 (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-(tert-butyl)-1-cyclopropyl-1H-pyrazole-5-carboxamide FIG.46is a schematic diagram illustrating an exemplary procedure for preparing Compound 33 according to some embodiments of the present disclosure. To a solution of (2S)-2-amino-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)propenamide (1.6 g, 4.58 mmol) and 5-tert-butyl-2-cyclopropyl pyrazole-3-carboxylic acid (954 mg, 4.58 mmol) in DCM (20 mL) was added T3P (50% in EA, 5.83 g, 9.16 mmol) and DIEA (1.18 g, 9.16 mmol). The reaction mixture was stirred at 25° C. under N2for 3 hrs. The reaction mixture was concentrated and the residue was purified by silica gel column chromatography (PE:EA=1:1) to give the desired product Compound 33 as a yellow solid (1.3 g, 47%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 539.1 541.1[M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.17 (s, 1H), 8.84 (d, J=8.0 Hz, 1H), 8.00 (d, J=1.7 Hz, 1H), 7.64 (d, J=8.4 Hz, 1H), 7.52 (dd, J=8.4, 1.8 Hz, 1H), 6.68 (s, 1H), 4.99-4.88 (m, 1H), 4.26-4.13 (m, 1H), 3.49 (dd, J=15.4, 5.8 Hz, 1H), 3.34-3.29 (m, 1H), 1.55-1.39 (m, 2H), 1.20 (s, 9H), 1.13-0.80 (m, 6H). Example 3.34: Preparation of Compound 34 (S)—N-(3-(6-(6-aminopyridin-3-yl)benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl) amino)-1-oxopropan-2-yl)-3-(tert-butyl)-1-cyclopropyl-1H-pyrazole-5-carboxamide FIG.47is a schematic diagram illustrating an exemplary procedure for preparing Compound 34 according to some embodiments of the present disclosure. To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-3-(tert-butyl)-1-cyclopropyl-1H-pyrazole-5-carboxamide (150 mg, 0.28 mmol) and (6-aminopyridin-3-yl)boronic acid (58 mg, 0.42 mmol) in dioxane (3 mL) and H2O (0.3 mL) was added K3PO4(118 mg, 0.56 mmol) and Pd(dppf)Cl2(20 mg, 0.028 mmol). The reaction mixture was stirred at 90° C. under N2for 10 hrs. The reaction mixture was concentrated and the residue was purified by Pre-TLC (DCM: MeOH=20:1) to give the desired product Compound 34 as a brown solid (120 mg, 72%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 553.3[M+H]+.1H NMR (400 MHz, DMSO-d) δ 9.08 (s, 1H), 8.75 (d, J=8.0 Hz, 1H), 8.25 (d, J=2.2 Hz, 1H), 7.80 (d, J=1.2 Hz, 1H), 7.71 (dd, J=8.6, 2.4 Hz, 1H), 7.62 (d, J=8.2 Hz, 1H), 7.50 (dd, J=8.2, 1.6 Hz, 1H), 6.65 (s, 1H), 6.49 (d, J=8.6 Hz, 1H), 6.07 (s, 2H), 4.96-4.85 (m, 1H), 4.21-4.13 (m, 1H), 3.45 (dd, J=15.6, 5.8 Hz, 1H), 3.30-3.24 (m, 1H), 1.49-1.38 (m, 2H), 1.16 (s, 9H), 1.12-1.05 (m, 2H), 1.01-0.96 (m, 1H), 0.93-0.88 (m, 1H), 0.83-0.76 (m, 2H). Example 3.35: Preparation of Compound 35 (S)—N-(1-((cyanomethyl)amino)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide FIG.48is a schematic diagram illustrating an exemplary procedure for preparing Compound 35 according to some embodiments of the present disclosure. Step 1. Preparation of tert-butyl N-[(1S)-2-(6-bromo-1,3-benzoxazol-2-yl)-1-[(cyanomethyl)carbamoyl]ethyl]carbamate To a mixture of (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-2-{[(tert-butoxy)carbonyl] amino}propanoic acid (2.90 g, 7.5 mmol) in DMF (30.0 mL) was added 2-aminoacetonitrile (0.69 g, 7.5 mmol), DIEA (2.91 g, 22.5 mmol), and HATU (5.70 g, 15 mmol). The reaction was stirred at RT for 2 hrs. Water (100 mL) was added and the reaction mixture was extracted with EA (100 mL×3). The combined organic layers were washed with brine (100 mL×3), dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by flash column (PE:EA=0-50%) to give the desired product tert-butyl N-[(1 S)-2-(6-bromo-1,3-benzoxazol-2-yl)-1-[(cyano methyl)carbamoyl]ethyl]carbamate as a white solid (2.7 g, 76%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 423.0 [M+H]+. Step 2. Preparation of tert-butyl (S)-(1-((cyanomethyl)amino)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)carbamate To a solution of tert-butyl (S)-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((cyanomethyl)amino)-1-oxopropan-2-yl)carbamate (1400 mg, 3.31 mmol) in dioxane/H2O (10/1, 30 mL) was added (2-methylpyridin-4-yl)boronic acid (679 mg, 4.96 mmol), K3PO4(2103 mg, 9.92 mmol), and 1,1-Bis(diphenylphosphino) ferrocenepalladiumdichloride (242 mg, 0.33 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The solvent was removed under reduced pressure and the residue was purified by Combiflash (PE/EA=0˜ 80%) to give the product as a brown solid (840 mg, 58%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 436.1 [M+H]+. Step 3. Preparation of (S)-2-amino-N-(cyanomethyl)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)propanamide To a solution of (S)-(1-((cyanomethyl)amino)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)carbamate (790 mg, 1.81 mmol) in MeCN (15 mL) was added TMSI (907 mg, 4.54 mmol). The reaction mixture was stirred at RT under N2for 0.5 hr. H2O (50 mL) was added to the reaction mixture, and then adjusted to pH 8 by using sat. NaHCO3and extracted with EA (50 mL×3). The combined organic layer was washed with brine (30 mL×2), then dried over with anhydrous Na2SO4to give the product as a yellow solid (550 mg, 58%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 336.2 [M+H]+. Step 4. Preparation of (S)—N-(1-((cyanomethyl)amino)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1 H-pyrazole-5-carboxamide To a solution of (S)-2-amino-N-(cyanomethyl)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)propanamide (130 mg, 0.39 mmol) in DMF (10 mL) was added 2-cyclopropyl-5-(1-methylcyclopropyl)pyrazole-3-carboxylic acid (80 mg, 0.39 mmol), DIEA (250 mg, 1.94 mmol), and HATU (370 mg, 1.16 mmol). The reaction mixture was stirred at RT under N2for 2 hrs. After the reaction completed, H2O (30 mL) was added to the reaction mixture, and then extracted with EA (30 mL×3). The combined organic layer was washed with brine (50 mL×2), then dried over with anhydrous Na2SO4. After filtration, the solution was concentrated under vacuum, and the residue was purified by Combiflash (DCM/MeOH=0˜ 10%) to give the desired product Compound 35 as a white solid (140 mg, 68%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 523.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.86 (t, J=6.8 Hz, 2H), 8.51 (d, J=5.4 Hz, 1H), 8.14 (s, 1H), 7.78 (s, 2H), 7.67 (s, 1H), 7.58 (d, J=5.2 Hz, 1H), 6.61 (s, 1H), 5.09 (d, J=5.4 Hz, 1H), 4.21 (dd, J=7.6, 3.8 Hz, 1H), 4.17 (dd, J=5.6, 2.0 Hz, 2H), 3.59 (dd, J=15.6, 5.4 Hz, 1H), 3.42-3.36 (m, 1H), 2.54 (s, 3H), 1.33 (s, 3H), 1.01 (dd, J=6.3, 3.8 Hz, 1H), 0.91 (dd, J=10.0, 6.2 Hz, 1H), 0.85-0.74 (m, 4H), 0.68 (dd, J=6.2, 3.8 Hz, 2H). Example 3.36: Preparation of Compound 36 (S)-2-((3-bromophenyl)sulfonamido)-N-(cyanomethyl)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)propanamide FIG.49is a schematic diagram illustrating an exemplary procedure for preparing Compound 36 according to some embodiments of the present disclosure. To a solution of (S)-2-amino-N-(cyanomethyl)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)propanamide (80 mg, 0.24 mmol) in DCM (5 mL) was added 3-bromobenzenesulfonyl chloride (60 mg, 0.24 mmol) and TEA (73 mg, 0.72 mmol) at 0° C. The reaction mixture was stirred at RT under N2for 3 hrs. The resulting mixture was concentrated. The residue was purified via Prep-TLC (DCM/MeOH=20/1) to give the desired product Compound 36 (S)-2-((3-bromophenyl)sulfonamido)-N-(cyanomethyl)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)propanamide as a white solid (20 mg, 15%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 554.0 [M+H]+.1H NMR (400 MHz, MeOD) δ 8.48 (d, J=5.4 Hz, 1H), 7.88-7.83 (m, 2H), 7.74 (dd, J=8.2, 1.6 Hz, 1H), 7.69-7.62 (m, 3H), 7.59 (d, J=5.4 Hz, 1H), 7.42-7.38 (m, 1H), 7.16 (t, J=7.8 Hz, 1H), 4.49 (dd, J=9.6, 5.0 Hz, 1H), 4.14 (s, 2H), 3.41 (dd, J=15.4, 5.0 Hz, 1H), 3.21 (dd, J=15.4, 9.6 Hz, 1H), 2.63 (s, 3H).). Example 3.37: Preparation of Compound 37 (S)-2-((3-chlorophenyl)sulfonamido)-N-(cyanomethyl)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)propanamide FIG.50is a schematic diagram illustrating an exemplary procedure for preparing Compound 37 according to some embodiments of the present disclosure. To a solution of (S)-2-amino-N-(cyanomethyl)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)propanamide (80 mg, 0.24 mmol) in DCM (5 mL) was added 3-chlorobenzenesulfonyl chloride (50 mg, 0.24 mmol) and TEA (73 mg, 0.72 mmol) at 0° C. The reaction mixture was stirred at RT under N2for 3 hrs. The resulting mixture was concentrated. The residue was purified via Prep-TLC (DCM/MeOH=20/1) to give the desired product Compound 37 (S)-2-((3-chlorophenyl)sulfonamido)-N-(cyanomethyl)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)propanamide as a white solid (30 mg, 20%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 510.0 [M+H]+.1H NMR (400 MHz, MeOD) δ 8.48 (d, J=5.4 Hz, 1H), 7.86 (d, J=1.2 Hz, 1H), 7.75-7.69 (m, 2H), 7.66 (d, J=8.4 Hz, 2H), 7.60 (dt, J=10.6, 2.8 Hz, 2H), 7.25 (t, J=4.6 Hz, 2H), 4.49 (dd, J=9.4, 5.0 Hz, 1H), 4.13 (s, 2H), 3.41 (dd, J=15.4, 5.0 Hz, 1H), 3.21 (dd, J=15.4, 9.4 Hz, 2H), 2.63 (s, 3H). Example 3.38: Preparation of Compound 38 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide FIG.51is a schematic diagram illustrating an exemplary procedure for preparing Compound 38 according to some embodiments of the present disclosure. Step 1. Preparation of (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]formamido}propenamide To a mixture of (2S)-2-amino-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl) propenamide (454 mg, 1.30 mmol) and 2-cyclopropyl-5-(1-methylcyclopropyl)pyrazole-3-carboxylic acid (295 mg, 1.43 mmol) in DCM (10.0 mL) was added DIEA (504 mg, 3.90 mmol) and T3P (50% in EA, 1.65 g, 2.60 mmol). The reaction was stirred at RT for 16 hrs. The reaction mixture was diluted with water (100 mL) and extracted with EA (100 mL×3). The combined organic layers were washed with NaHCO3(100 mL×3), brine (100 mL), dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by flash column (PE: EA=0-60%) to give the product (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]formamido}propanamide as a white solid (360 mg, 49%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 537.1 [M+H]+.1H NMR (400 MHz, MeOD) δ 7.83 (d, J=1.6 Hz, 1H), 7.54 (dt, J=8.4, 5.2 Hz, 2H), 6.51 (s, 1H), 5.04 (dd, J=8.2, 6.2 Hz, 1H), 4.01-3.89 (m, 1H), 3.59 (dd, J=15.4, 6.2 Hz, 1H), 3.43 (dd, J=15.4, 8.2 Hz, 1H), 1.57-1.46 (m, 2H), 1.40 (s, 3H), 1.23 (td, J=12.6, 4.6 Hz, 2H), 1.11-1.05 (m, 1H), 1.02-0.80 (m, 5H), 0.70 (q, J=4.0 Hz, 2H). Step 2. Preparation of (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl) amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (300 mg, 0.56 mmol) in dioxane/H2O (10/1, 15 mL) was added (2-methylpyridin-4-yl)boronic acid (77 mg, 0.56 mmol), K3PO4(237 mg, 1.12 mmol), and 1,1′-Bis(diphenylphosphino)ferrocenepalladiumdichloride (45 mg, 0.06 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The resulting mixture was concentrated. The residue was purified via Prep-TLC (DCM/MeOH=20/1) to give the desired product Compound 38 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(2-methylpyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide as a white solid (208 mg, 68%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 550.2 [M+H]+.1H NMR (400 MHz, MeOD) δ 8.46 (d, J=5.2 Hz, 1H), 8.00 (s, 1H), 7.76 (d, J=0.9 Hz, 2H), 7.65 (s, 1H), 7.56 (d, J=5.2 Hz, 1H), 6.51 (s, 1H), 5.15-5.01 (m, 1H), 3.97-3.94 (m, 1H), 3.65-3.60 (m, 1H), 3.49-3.43 (m, 1H), 2.61 (s, 3H), 1.50 (d, J=3.0 Hz, 2H), 1.38 (s, 3H), 1.26-1.19 (m, 2H), 1.09-1.02 (m, 1H), 0.96-0.93 (m, 1H), 0.90-0.79 (m, 4H), 0.68 (q, J=4.0 Hz, 2H). Example 3.39: Preparation of Compound 39 (2S)—N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]formamido}-3-{6-[6-(dimethylamino)pyridin-3-yl]-1,3-benzoxazol-2-yl}propanamide FIG.52is a schematic diagram illustrating an exemplary procedure for preparing Compound 39 according to some embodiments of the present disclosure. To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl) amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (80 mg, 0.1489 mmol) in dioxane/H2O (10/1, 2 mL) was added (6-(dimethylamino)pyridin-3-yl)boronic acid (37 mg, 0.223 mmol), K3PO4(63 mg, 0.297 mmol), and 1,1′-Bis(diphenylphosphino)ferrocenepalladiumdichloride (7 mg, 0.089 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The solvent was removed under reduced pressure and the residue was purified by Prep-HPLC [Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA), 25-50] to give the desired product Compound 39 as a white solid (25 mg, 27%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 579.0 [M+H]+.1H NMR (400 MHz, DMSO-d) δ 9.12 (s, 1H), 8.77 (d, J=7.8 Hz, 1H), 8.48 (d, J=2.2 Hz, 1H), 7.90 (s, 1H), 7.68 (d, J=8.2 Hz, 1H), 7.59 (d, J=8.2 Hz, 1H), 6.75 (d, J=8.6 Hz, 1H), 6.61 (s, 1H), 4.94 (dd, J=14.0, 8.2 Hz, 1H), 4.24-4.12 (m, 1H), 3.48 (dd, J=15.4, 5.6 Hz, 1H), 3.07 (d, J=9.8 Hz, 6H), 1.48 (d, J=2.4 Hz, 2H), 1.34 (s, 3H), 1.23 (s, 1H), 1.11 (q, J=10.6 Hz, 2H), 1.04-0.97 (m, 1H), 0.97-0.89 (m, 1H), 0.85-0.77 (m, 4H), 0.68 (dd, J=6.0, 3.8 Hz, 2H). Example 3.40: Preparation of Compound 40 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(1-(2,2-difluoroethyl)-1,2,3,6-tetrahydropyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide FIG.53is a schematic diagram illustrating an exemplary procedure for preparing Compound 40 according to some embodiments of the present disclosure. To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl) amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (100 mg, 0.18 mmol) in dioxane/H2O (10/1, 10 mL) was added 1-(2,2-difluoroethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,6-tetrahydro pyridine (51 mg, 0.18 mmol), K3PO4(76 mg, 0.36 mmol), and 1,1′-Bis(diphenylphosphino)ferrocenepalladiumdichloride (16 mg, 0.02 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The resulting mixture was concentrated. The residue was purified by Prep-TLC (DCM/MeOH=10/1) to give the desired product Compound 40 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(1-(2,2-difluoroethyl)-1,2,3,6-tetrahydropyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide as a white solid (8 mg, 7%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 604.1 [M+H]+.1H NMR (400 MHz, MeOD) δ 7.64 (s, 1H), 7.59 (d, J=8.4 Hz, 1H), 7.49 (d, J=8.4 Hz, 1H), 6.51 (s, 1H), 6.20 (d, J=4.2 Hz, 1H), 5.99 (dt, J=55.8, 4.2 Hz, 1H), 5.04 (dd, J=8.2, 6.2 Hz, 1H), 3.94 (td, J=7.4, 3.8 Hz, 1H), 3.59 (dd, J=15.2, 6.2 Hz, 1H), 3.43 (dd, J=15.2, 8.4 Hz, 1H), 3.37-3.34 (m, 2H), 2.99-2.87 (m, 3H), 2.65 (s, 2H), 1.49 (t, J=8.2 Hz, 2H), 1.40 (s, 3H), 1.31 (s, 1H), 1.30-1.15 (m, 2H), 1.06 (dt, J=8.2, 5.2 Hz, 1H), 1.00-0.77 (m, 5H), 0.70 (q, J=3.8 Hz, 2H). Example 3.41: Preparation of Compound 41 (2S)—N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]formamido}-3-[6-(6-acetamidopyridin-3-yl)-1,3-benzoxazol-2-yl]propanamide FIG.54is a schematic diagram illustrating an exemplary procedure for preparing Compound 41 according to some embodiments of the present disclosure. Step 1. Preparation of (S)—N-(3-(6-(6-aminopyridin-3-yl) benzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl) amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (300 mg, 0.56 mmol) in dioxane/H2O (10/1, 15 mL) was added (6-aminopyridin-3-yl)boronic acid (77 mg, 0.56 mmol), K3P04 (237 mg, 1.12 mmol), and 1,1′-Bis(diphenylphosphino)ferrocenepalladiumdichloride (45 mg, 0.06 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The resulting mixture was concentrated. The residue was purified by Prep-TLC (DCM: MeOH=10:1) to give the product as a white solid (140 mg, 45%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 551.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.12 (s, 1H), 8.76 (d, J=8.0 Hz, 1H), 8.29 (d, J=2.4 Hz, 1H), 7.85 (s, 1H), 7.77 (dd, J=8.6, 2.4 Hz, 1H), 7.66 (d, J=8.2 Hz, 1H), 7.54 (dd, J=8.4, 1.4 Hz, 1H), 6.61 (s, 1H), 6.55 (d, J=8.6 Hz, 1H), 6.17 (s, 2H), 4.93 (dd, J=14.2, 8.4 Hz, 1H), 4.26-4.12 (m, 1H), 3.48 (dd, J=15.6, 5.8 Hz, 1H), 3.29 (s, 1H), 1.51-1.44 (m, 2H), 1.34 (s, 3H), 1.12-1.07 (m, 2H), 1.04-0.98 (m, 1H), 0.95-0.89 (m, 1H), 0.88-0.75 (m, 4H). Step 2. Preparation of (2S)—N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]formamido}-3-[6-(6-acetamidopyridin-3-yl)-1,3-benzoxazol-2-yl]propanamide To a mixture of (2S)-3-[6-(6-aminopyridin-3-yl)-1,3-benzoxazol-2-yl]-N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]form amido}propenamide (45 mg, 0.081 mmol) in Pyridine (1 mL) was added acetyl acetate (16 mg, 0.163 mmol). The reaction was stirred at RT under N2for 18 hrs. The solvent was removed under reduced pressure and the residue was purified by Prep-HPLC [Gemini-C18, 150×21.2 mm, Sum; ACN-H2O (0.1% FA), 15-40] to give the desired product Compound 41 as a white solid (15 mg, 29%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 593.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 10.58 (s, 1H), 9.08 (s, 1H), 8.73 (d, J=8.0 Hz, 1H), 8.65 (d, J=1.8 Hz, 1H), 8.11 (d, J=2.4 Hz, 2H), 8.00 (d, J=1.4 Hz, 1H), 7.67 (dt, J=8.4, 5.0 Hz, 2H), 6.57 (s, 1H), 4.91 (td, J=8.2, 5.8 Hz, 1H), 4.26-4.04 (m, 1H), 3.51-3.42 (m, 1H), 3.30 (dd, J=15.6, 8.8 Hz, 1H), 2.08 (s, 3H), 1.47-1.41 (m, 2H), 1.30 (s, 3H), 1.23-1.19 (m, 3H), 1.14-1.02 (m, 2H), 0.99-0.94 (m, 1H), 0.91-0.86 (m, 1H), 0.84-0.72 (m, 5H), 0.64 (dd, J=6.2, 3.8 Hz, 2H). Example 3.42: Preparation of Compound 42 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(6-methylpyridin-3-yl)benzo[d] oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide FIG.55is a schematic diagram illustrating an exemplary procedure for preparing Compound 42 according to some embodiments of the present disclosure. To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl) amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (150 mg, 0.28 mmol) in dioxane/H2O (10:1, 10 mL) was added (6-methylpyridin-3-yl)boronic acid (57 mg, 0.42 mmol), K3PO4(118 mg, 0.56 mmol), and 1,1′-Bis(diphenylphosphino)ferrocenepalladiumdichloride (20 mg, 0.03 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The resulting mixture was concentrated to give the crude product. The residue was purified via Prep-TLC (DCM/MeOH=20/1) to give the desired product Compound 42 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(6-methylpyridin-3-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide as a white solid (70 mg, 45%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 550.2[M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.12 (s, 1H), 8.80 (dd, J=14.0, 5.2 Hz, 2H), 8.06-8.00 (m, 2H), 7.76 (d, J=8.4 Hz, 1H), 7.68 (dd, J=8.4, 1.6 Hz, 1H), 7.36 (d, J=8.2 Hz, 1H), 6.61 (s, 1H), 4.95 (d, J=5.8 Hz, 1H), 4.19 (d, J=3.8 Hz, 1H), 3.49 (d, J=5.8 Hz, 1H), 3.37 (d, J=8.8 Hz, 1H), 1.48 (d, J=2.4 Hz, 2H), 1.34 (s, 3H), 1.12 (d, J=10.8 Hz, 2H), 1.00 (s, 1H), 0.93 (s, 1H), 0.80 (dd, J=5.6, 3.8 Hz, 4H), 0.68 (dd, J=6.2, 3.8 Hz, 2H). Example 3.43: Preparation of Compound 43 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(6-(ethylamino)pyridin-3-yl)benzo[d] oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide FIG.56is a schematic diagram illustrating an exemplary procedure for preparing Compound 43 according to some embodiments of the present disclosure. Step 1. Preparation of (6-(ethylamino)pyridin-3-yl)boronic acid To a solution of 5-bromo-N-ethylpyridin-2-amine (100 mg, 0.5 mmol) in dioxane (10 mL) was added 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (190 mg, 0.75 mmol), KOAc (98 mg, 1 mmol), and Pd(dppf)Cl2(40 mg, 0.05 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The solvent was removed under reduced pressure to give the crude product (6-(ethylamino)pyridin-3-yl)boronic acid as yellow oil (90 mg, purity: 60%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 167.3 [M+H]+. Step 2. Preparation of (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(6-(ethylamino) pyridine-3-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methyl cyclopropyl)-1H-pyrazole-5-carboxamide To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyano cyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (100 mg, 0.19 mmol) in dioxane/H2O (10:1, 10 mL) was added (6-(ethylamino)pyridin-3-yl)boronic acid (46 mg, 0.28 mmol), K3PO4(79 mg, 0.38 mmol), and Pd(dppf)Cl2(14 mg, 0.02 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The solvent was removed under reduced pressure and the residue was purified by Combiflash column (DCM/MeOH=0˜10%) to give the desired product Compound 43 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(6-(ethylamino)pyridin-3-yl) benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide as a white solid (43 mg, 39%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 579.2 [M+H]+.1H (400 MHz, MeOD) δ 8.23 (d, J=2.2 Hz, 1H), 7.78-7.72 (m, 2H), 7.67 (d, J=8.4 Hz, 1H), 7.54 (dd, J=8.2, 1.6 Hz, 1H), 6.63 (d, J=8.8 Hz, 1H), 6.52 (s, 1H), 5.06 (dd, J=8.4, 6.0 Hz, 1H), 4.00-3.92 (m, 1H), 3.61 (dd, J=15.2, 6.2 Hz, 1H), 3.45 (dd, J=15.4, 8.4 Hz, 1H), 3.40-3.35 (m, 2H), 1.51 (d, J=2.8 Hz, 2H), 1.39 (s, 3H), 1.28 (d, J=7.2 Hz, 3H), 1.25-1.16 (m, 2H), 1.10-1.03 (m, 1H), 0.97 (dd, J=10.0, 6.0 Hz, 1H), 0.88 (ddd, J=13.6, 8.6, 4.0 Hz, 4H), 0.69 (q, J=4.0 Hz, 2H). Example 3.44: Preparation of Compound 44 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(6-methylpyridin-2-yl)benzo[d] oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide FIG.57is a schematic diagram illustrating an exemplary procedure for preparing Compound 44 according to some embodiments of the present disclosure. Step 1. Preparation of (S)—N-(1-((1-cyanocyclopropyl)amino)-1-oxo-3-(6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazol-2-yl)propan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide To a solution of (S)—N-(3-(6-bromobenzo[d]oxazol-2-yl)-1-((1-cyanocyclopropyl)amino)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (500 mg, 0.93 mmol) in dioxane (15 mL) was added 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (354 mg, 1.39 mmol), KOAc (183 mg, 1.86 mmol), and Pd(dppf)Cl2(68 mg, 0.09 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The solvent was removed under reduced pressure and the residue was purified by Combiflash column (DCM/MeOH=0˜5%) to give the product (S)—N-(1-((1-cyanocyclopropyl)amino)-1-oxo-3-(6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazol-2-yl)propan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide as brown oil (400 mg, 73%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 585.3 [M+H]+. Step 2. Preparation of (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(6-methylpyridin-2-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide To a solution of (S)—N-(1-((1-cyanocyclopropyl)amino)-1-oxo-3-(6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazol-2-yl)propan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (40 mg, 0.07 mmol) in dioxane/H2O (10:1, 5 mL) was added 2-bromo-6-methylpyridine (13 mg, 0.07 mmol), K3PO4(33 mg, 0.14 mmol), and Pd(dppf)Cl2(6 mg, 0.01 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The solvent was removed under reduced pressure and the residue was purified by Combiflash column (DCM/MeOH=0˜10%) to give the desired product Compound 44 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(6-methylpyridin-2-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide as a yellow solid (4 mg, 10%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 550.2 [M+H]+.1H NMR (400 MHz, MeOD) δ 8.18 (d, J=1.2 Hz, 1H), 7.97 (dd, J=8.4, 1.6 Hz, 1H), 7.77 (d, J=7.6 Hz, 1H), 7.70 (dd, J=17.6, 8.2 Hz, 2H), 7.24 (d, J=7.6 Hz, 1H), 6.51 (s, 1H), 5.06 (dd, J=8.4, 6.2 Hz, 1H), 3.98-3.92 (m, 1H), 3.60 (s, 1H), 3.48 (d, J=8.4 Hz, 1H), 2.61 (s, 3H), 1.49 (d, J=2.8 Hz, 2H), 1.38 (s, 3H), 1.24-1.18 (m, 2H), 1.07-1.01 (m, 1H), 0.96 (dd, J=10.0, 6.0 Hz, 1H), 0.88 (dt, J=9.8, 5.2 Hz, 4H), 0.68 (q, J=4.0 Hz, 2H). Example 3.45: Preparation of Compound 45 (2S)-2-[(5-tert-butyl-2-cyclopropylpyrazol-3-yl)formamido]-N-(cyanomethyl)-3-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}propanamide FIG.58is a schematic diagram illustrating an exemplary procedure for preparing Compound 45 according to some embodiments of the present disclosure. Step 1. Preparation of 1-(2-methoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine To a mixture of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,6-tetrahydropyridine (1.0 g, 4.07 mmol) in DMF (15.0 mL) was added DIEA (1.58 g, 12.2 mmol) and 1-bromo-2-methoxyethane (623 mg, 4.48 mmol). The reaction was stirred at 60° C. for 16 hrs. The reaction mixture was diluted with water (50 mL) and then extracted with EA (50 mL×3). The combined organic layer was washed with brine (100 mL×3), dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by flash column (DCM:MeOH=15:1) to give the desired product 1-(2-methoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine as colorless oil (1.5 g, 96.5%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 268.2 [M+H]+. Step 2. Preparation of tert-butyl N-[(1S)-1-[(cyanomethyl)carbamoyl]-2-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}ethyl]carbamate To a mixture of 1-(2-methoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine (196 mg, 0.732 mmol) in 1,4-dioxane/H2O (10:1, 11.0 mL) was added tert-butyl N-[(1S)-2-(6-bromo-1,3-benzoxazol-2-yl)-1-[(cyanomethyl) carbamoyl]ethyl]carbamate (310 mg, 0.732 mmol), K2CO3(304 mg, 2.20 mmol), and Pd(dppf)Cl2(59.8 mg, 0.0732 mmol). The reaction mixture was stirred under N2at 90° C. for 6 hrs. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was diluted with water (20 mL) and then extracted with EA (20 mL×3). The combined organic layer was washed with brine (50 mL×2), dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by flash column (DCM:MeOH=20:1) to give the product tert-butyl N-[(1 S)-1-[(cyanomethyl)carbamoyl]-2-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}ethyl]carbamate as a white solid (150 mg, 42%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 484.2 [M+H]+. Step 3. Preparation of (2S)-2-amino-N-(cyanomethyl)-3-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}propanamide To a mixture of tert-butyl N-[(1S)-1-[(cyanomethyl)carbamoyl]-2-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}ethyl]carbamate (150 mg, 0.310 mmol) in MeCN (5.0 mL) was added TMSI (155 mg, 0.776 mmol). The reaction was stirred at RT for 0.5 hr. The reaction mixture was concentrated under reduced pressure to give the product (2S)-2-amino-N-(cyanomethyl)-3-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}propanamide as a brown solid (100 mg, 75.7%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 384.2 [M+H]+. Step 4. Preparation of (2S)-2-[(5-tert-butyl-2-cyclopropylpyrazol-3-yl)formamido]-N-(cyanomethyl)-3-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}propanamide To a mixture of (120 mg, 0.248 mmol) in DMF (5.0 mL) was added 5-tert-butyl-2-cyclopropylpyrazole-3-carboxylic acid (62 mg, 0.298 mmol), DIEA (96 mg, 0.745 mmol), and HATU (189 mg, 0.496 mmol). The reaction was stirred at RT for 2 hrs. The reaction mixture was diluted with water (20 mL) and then extracted with EA (20 mL×3). The combined organic layer was washed with brine (50 mL×3), dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM:MeOH=15:1) to give the desired product Compound 45 (2S)-2-[(5-tert-butyl-2-cyclopropylpyrazol-3-yl)formamido]-N-(cyanomethyl)-3-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}propanamide as a white solid (15 mg, 10%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 547.8 [M+H]+.1H NMR (400 MHz, CDCl3) δ 8.12 (d, J=6.8 Hz, 1H), 8.01 (t, J=5.6 Hz, 1H), 7.57 (d, J=8.2 Hz, 1H), 7.51 (s, 1H), 7.39 (d, J=8.4 Hz, 1H), 6.55 (s, 1H), 6.08 (s, 1H), 5.19 (dd, J=11.0, 6.8 Hz, 1H), 4.18 (t, J=4.6 Hz, 2H), 3.75-3.67 (m, 2H), 3.68 (d, J=4.0 Hz, 1H), 3.53 (s, 2H), 3.39 (s, 3H), 3.33 (dd, J=16.8, 6.8 Hz, 1H), 3.07 (s, 2H), 2.97 (s, 2H), 2.77 (s, 2H), 1.43-1.39 (m, 1H), 1.27 (d, J=12.2 Hz, 9H), 1.01 (d, J=8.0 Hz, 2H), 0.88 (t, J=6.8 Hz, 2H). Example 3.46: Preparation of Compound 46 (2S)—N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]formamido}-3-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}propanamide FIG.59is a schematic diagram illustrating an exemplary procedure for preparing Compound 46 according to some embodiments of the present disclosure. To a mixture of (2S)-3-(6-bromo-1,3-benzoxazol-2-yl)-N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]formamido}propanamide (200 mg, 0.372 mmol) and 1-(2-methoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine (249 mg, 0.931 mmol) in dioxane/H2O (10:1, 11 mL) was added K3PO4(158 mg, 0.744 mmol) and Pd(dppf)Cl2(30 mg, 0.037 mmol). The reaction was stirred at 90° C. under N2for 16 hrs. The reaction mixture was filtered and the filtrate was extracted with H2O (100 mL) and EtOAc (50×3 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, and concentrated under reduced pressure. The crude product was purified by SFC [Column: chiralpak-AD; mobile phase: CO2-IPA (DEA)] to give the desired product Compound 46 (2S)—N-(1-cyanocyclopropyl)-2-{[2-cyclopropyl-5-(1-methylcyclopropyl)pyrazol-3-yl]form amido}-3-{6-[1-(2-methoxyethyl)-3,6-dihydro-2H-pyridin-4-yl]-1,3-benzoxazol-2-yl}propenamide as a white solid (25 mg, 11%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 598.2 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 9.10 (s, 1H), 8.75 (d, J=8.0 Hz, 1H), 7.70 (s, 1H), 7.61 (d, J=8.4 Hz, 1H), 7.45 (d, J=8.4 Hz, 1H), 6.59 (s, 1H), 6.22 (s, 1H), 4.92 (dd, J=14.0, 8.2 Hz, 1H), 4.20-4.16 (m, 1H), 3.54 (s, 2H), 3.47 (dd, J=15.6, 5.8 Hz, 1H), 3.28 (s, 3H), 3.33 (s, 3H), 2.79 (s, 3H), 2.59 (s, 2H), 1.49-1.45 (m, 2H), 1.34 (s, 3H), 1.14-0.98 (m, 3H), 0.94-0.89 (m, 1H), 0.86-0.77 (m, 4H), 0.68 (dd, J=6.2, 3.8 Hz, 2H). Example 3.47: Preparation of Compound 47 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(2-(difluoromethyl)pyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methyl cyclopropyl)-1H-pyrazole-5-carboxamide FIG.60is a schematic diagram illustrating an exemplary procedure for preparing Compound 47 according to some embodiments of the present disclosure. To a solution of (S)—N-(1-((1-cyanocyclopropyl)amino)-1-oxo-3-(6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazol-2-yl)propan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (100 mg, 0.17 mmol) in dioxane/H2O (10:1, 10 mL) was added 4-bromo-2-(difluoromethyl)pyridine (43 mg, 0.17 mmol), K3PO4(73 mg, 0.34 mmol), and Pd(dppf)Cl2(14 mg, 0.02 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The solvent was removed under reduced pressure and the residue was purified by Combiflash column (DCM/MeOH=0˜ 10%) to give the desired product Compound 47 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(2-(difluoromethyl)pyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-y)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide as a white solid (7 mg, 7%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 586.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.15 (s, 1H), 8.79 (dd, J=19.8, 6.6 Hz, 2H), 8.26 (s, 1H), 8.06 (s, 1H), 7.98 (d, J=4.8 Hz, 1H), 7.90-7.80 (m, 2H), 7.02 (t, J=54.8 Hz, 1H), 6.61 (s, 1H), 4.97 (d, J=6.0 Hz, 1H), 4.23-4.15 (m, 1H), 3.52 (d, J=5.6 Hz, 1H), 3.41-3.35 (m, 1H), 1.48 (s, 2H), 1.33 (s, 3H), 1.12 (d, J=10.4 Hz, 2H), 1.04-0.89 (m, 2H), 0.86-0.74 (m, 4H), 0.68 (dd, J=6.2, 3.8 Hz, 2H). Example 3.48: Preparation of Compound 48 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(2-cyclopropylpyridin-4-yl)benzo[d] oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide FIG.61is a schematic diagram illustrating an exemplary procedure for preparing Compound 48 according to some embodiments of the present disclosure. To a solution of (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(2-cyclopropyl pyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide (150 mg, 0.26 mmol) in dioxane/H2O (10:1, 10 mL) was added 4-bromo-2-cyclopropylpyridine (51 mg, 0.26 mmol), K3PO4(109 mg, 0.51 mmol), and Pd(dppf)Cl2(19 mg, 0.03 mmol). The reaction mixture was stirred at 90° C. under N2for 16 hrs. The solvent was removed under reduced pressure and the residue was purified by Combiflash column (DCM/MeOH=0˜10%) to give the desired product Compound 48 (S)—N-(1-((1-cyanocyclopropyl)amino)-3-(6-(2-cyclopropylpyridin-4-yl)benzo[d]oxazol-2-yl)-1-oxopropan-2-yl)-1-cyclopropyl-3-(1-methylcyclopropyl)-1H-pyrazole-5-carboxamide as a white solid (32 mg, 21%). Mass spectrometry was conducted on the resultant compound, and the test results are as follows: Mass(m/z): 576.3 [M+H]+.1H (400 MHz, MeOD) δ 8.40 (d, J=5.2 Hz, 1H), 7.99 (s, 1H), 7.76 (d, J=1.0 Hz, 2H), 7.54 (d, J=1.2 Hz, 1H), 7.47 (dd, J=5.2, 1.8 Hz, 1H), 6.51 (s, 1H), 5.06 (dd, J=8.4, 6.2 Hz, 1H), 3.98-3.92 (m, 1H), 3.63 (dd, J=15.4, 6.2 Hz, 1H), 3.46 (dd, J=15.4, 8.4 Hz, 1H), 2.21-2.16 (m, 1H), 1.49 (t, J=6.2 Hz, 2H), 1.38 (s, 3H), 1.24-1.18 (m, 2H), 1.09-1.05 (m, 2H), 1.03 (dd, J=4.6, 2.8 Hz, 2H), 0.98-0.78 (m, 6H), 0.68 (q, J=4.0 Hz, 2H). Having thus described the basic concepts, it may be rather apparent to those skilled in the art after reading this detailed disclosure that the foregoing detailed disclosure is intended to be presented by way of example only and is not limiting. Various alterations, improvements, and modifications may occur and are intended to those skilled in the art, though not expressly stated herein. These alterations, improvements, and modifications are intended to be suggested by this disclosure, and are within the spirit and scope of the exemplary embodiments of this disclosure. Moreover, certain terminology has been used to describe embodiments of the present disclosure. For example, the terms “one embodiment,” “an embodiment,” and “some embodiments” mean that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present disclosure. Therefore, it is emphasized and should be appreciated that two or more references to “an embodiment” or “one embodiment” or “an alternative embodiment” in various portions of this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures or characteristics may be combined as suitable in one or more embodiments of the present disclosure. Furthermore, the recited order of processing elements or sequences, or the use of numbers, letters, or other designations, therefore, is not intended to limit the claimed processes and methods to any order except as may be specified in the claims. Although the above disclosure discusses through various examples what is currently considered to be a variety of useful embodiments of the disclosure, it is to be understood that such detail is solely for that purpose and that the appended claims are not limited to the disclosed embodiments, but, on the contrary, are intended to cover modifications and equivalent arrangements that are within the spirit and scope of the disclosed embodiments. For example, although the implementation of various components described above may be embodied in a hardware device, it may also be implemented as a software-only solution, e.g., an installation on an existing server or mobile device. Similarly, it should be appreciated that in the foregoing description of embodiments of the present disclosure, various features are sometimes grouped together in a single embodiment, figure, or description thereof to streamline the disclosure aiding in the understanding of one or more of the various embodiments. This method of disclosure, however, is not to be interpreted as reflecting an intention that the claimed subject matter requires more features than are expressly recited in each claim. Rather, claim subject matter lie in less than all features of a single foregoing disclosed embodiment.
157,543
11858906
DETAILED DESCRIPTION OF THE EXEMPLARY EMBODIMENTS Below, a detailed description will be given of the present disclosure. In each drawing of the present disclosure, sizes or scales of components may be enlarged or reduced from their actual sizes or scales for better illustration, and known components may not be depicted therein to clearly show features of the present disclosure. Therefore, the present disclosure is not limited to the drawings. When describing the principle of the embodiments of the present disclosure in detail, details of well-known functions and features may be omitted to avoid unnecessarily obscuring the presented embodiments. In drawings, for convenience of description, sizes of components may be exaggerated for clarity. For example, since sizes and thicknesses of components in drawings are arbitrarily shown for convenience of description, the sizes and thicknesses are not limited thereto. Furthermore, throughout the description, the terms “on” and “over” are used to refer to the relative positioning, and mean not only that one component or layer is directly disposed on another component or layer but also that one component or layer is indirectly disposed on another component or layer with a further component or layer being interposed therebetween. Also, spatially relative terms, such as “below”, “beneath”, “lower”, and “between”, may be used herein for ease of description to refer to the relative positioning. Throughout the specification, when a portion may “include” a certain constituent element, unless explicitly described to the contrary, it may not be construed to exclude another constituent element but may be construed to further include other constituent elements. Further, throughout the specification, the word “on” means positioning on or below the object portion, but does not essentially mean positioning on the lower side of the object portion based on a gravity direction. The amine compound according to the present invention may be represented by the following [Chemical Formula A] or [Chemical Formula B]: wherein, the substituent Ar1is one selected from a substituted or unsubstituted alkyl of 1 to 30 carbon atoms, a substituted or unsubstituted haloalkyl of 1 to 30 carbon atoms, a substituted or unsubstituted aryl of 6 to 50 carbon atoms, a substituted or unsubstituted cycloalkyl of 3 to 30 carbon atoms, a substituted or unsubstituted heteroaryl of 2 to 50 carbon atoms, a substituted or unsubstituted alkylsilyl of 1 to 30 carbon atoms, and a substituted or unsubstituted arylsilyl of 5 to 30 carbon atoms, the linker L1is one selected from a single bond, a substituted or unsubstituted arylene of 6 to 30 carbon atoms, and a substituted or unsubstituted heteroarylene of 1 to 30 carbon atoms, the substituents R1and R2, which may be same or different, are each independently one selected from a substituted or unsubstituted alkyl of 1 to 30 carbon atoms, a substituted or unsubstituted haloalkyl of 1 to 30 carbon atoms, a substituted or unsubstituted aryl of 6 to 50 carbon atoms, a substituted or unsubstituted arylalkyl of 7 to 50 carbon atoms, a substituted or unsubstituted cycloalkyl of 3 to 30 carbon atoms, a substituted or unsubstituted heteroaryl of 2 to 50 carbon atoms, a substituted or unsubstituted alkoxy of 1 to 30 carbon atoms, a substituted or unsubstituted aryloxy of 6 to 30 carbon atoms, a substituted or unsubstituted alkylthioxy of 1 to 30 carbon atoms, a substituted or unsubstituted arylthioxy of 5 to 30 carbon atoms, a substituted or unsubstituted alkylamine of 1 to 30 carbon atoms, a substituted or unsubstituted arylamine of 5 to 30 carbon atoms, a substituted or unsubstituted alkylsilyl of 1 to 30 carbon atoms, a substituted or unsubstituted arylsilyl of 5 to 30 carbon atoms, a cyano, a nitro, and a halogen, R1and R2may be connected to each other to form a ring, and each of the carbon atoms on the aromatic rings of the indenodibenzofuran fused ring moiety in Chemical Formulas A and B may be bound with a hydrogen or deuterium, except for the carbon atoms bound with Ar1or L1, wherein the term “substituted” in the expression “substituted or unsubstituted” means having at least one substituent selected from the group consisting of a deuterium, a cyano, a halogen, a hydroxy, a nitro, an alkyl of 1 to 24 carbon atoms, a cycloalkyl of 3 to 24 carbon atoms, a halogenated alkyl of 1 to 24 carbon atoms, an alkenyl of 2 to 24 carbon atoms, an alkynyl of 2 to 24 carbon atoms, a heteroalkyl of 1 to 24 carbon atoms, an aryl of 6 to 24 carbon atoms, an arylalkyl of 7 to 24 carbon atoms, an alkylaryl of 7 to 24 carbon atoms, a heteroaryl of 2 to 24 carbon atoms, a heteroarylalkyl of 2 to 24 carbon atoms, an alkoxy of 1 to 24 carbon atoms, an alkylamino of 1 to 24 carbon atoms, a diarylamino of 12 to 24 carbon atoms, a diheteroarylamino of 2 to 24 carbon atoms, an aryl(heteroaryl)amino of 7 to 24 carbon atoms, an alkylsilyl of 1 to 24 carbon atoms, an arylsilyl of 6 to 24 carbon atoms, an aryloxy of 6 to 24 carbon atoms, and an arylthionyl of 6 to 24 carbon atoms. The expression indicating the number of carbon atoms, such as “a substituted or unsubstituted alkyl of 1 to 30 carbon atoms”, “a substituted or unsubstituted aryl of 6 to 50 carbon atoms”, etc. means the total number of carbon atoms of, for example, the alkyl or aryl radical or moiety alone, exclusive of the number of carbon atoms of substituents attached thereto. For instance, a phenyl group with a butyl at the para position falls within the scope of an aryl of 6 carbon atoms, even though it is substituted with a butyl radical of 4 carbon atoms. As used herein, the term “aryl” means an organic radical derived from an aromatic hydrocarbon by removing one hydrogen that is bonded to the aromatic hydrocarbon. The aromatic system may include a fused ring that is formed by adjacent substituents on the aryl radical. Concrete examples of the aryl include phenyl, o-biphenyl, m-biphenyl, p-biphenyl, o-terphenyl, m-terphenyl, p-terphenyl, naphthyl, anthryl, phenanthryl, pyrenyl, indenyl, fluorenyl, tetrahydronaphthyl, perylenyl, chrysenyl, naphthacenyl, and fluoranthenyl. At least one hydrogen atom of the aryl may be substituted by a deuterium atom, a halogen atom, a hydroxy, a nitro, a cyano, a silyl, an amino (—NH2, —NH(R), —N(R′) (R″) wherein R′ and R″ are each independently an alkyl of 1 to 10 carbon atoms, in this case, called “alkylamino”), an amidino, a hydrazine, a hydrazone, a carboxyl, a sulfonic acid, a phosphoric acid, an alkyl of 1 to 24 carbon atoms, a halogenated alkyl of 1 to 24 carbon atoms, an alkenyl of 1 to 24 carbon atoms, an alkynyl of 1 to 24 carbon atoms, a heteroalkyl of 1 to 24 carbon atoms, an aryl of 6 to 24 carbon atoms, an arylalkyl of 6 to 24 carbon atoms, a heteroaryl of 2 to 24 carbon atoms, or a heteroarylalkyl of 2 to 24 carbon atoms. The term “heteroaryl substituent” used in the compound of the present disclosure refers to a hetero aromatic radical of 2 to 50 carbon atoms, preferably 2 to 24 carbon atoms, bearing 1 to 3 heteroatoms selected from among N, O, P, Si, S, Ge, Se, and Te. In the aromatic radical, two or more rings may be fused. One or more hydrogen atoms on the heteroaryl may be substituted by the same substituents as on the aryl. In addition, the term “heteroaromatic ring”, as used herein, refers to an aromatic hydrocarbon ring bearing at least one heteroatom as aromatic ring member. In the heteroaromatic ring, one to three carbon atoms of the aromatic hydrocarbon may be substituted by at least one selected particularly from N, O, P, Si, S, Ge, Se, and Te. As used herein, the term “alkyl” refers to an alkane missing one hydrogen atom and includes linear or branched structures. Examples of the alkyl substituent useful in the present disclosure include methyl, ethyl, propyl, isopropyl, isobutyl, sec-butyl, tert-butyl, pentyl, isoamyl, and hexyl. At least one hydrogen atom of the alkyl may be substituted by the same substituent as in the aryl. The term “cyclo” as used in substituents of the present disclosure refers to a structure responsible for a mono- or polycyclic ring of saturated hydrocarbons in an alkyl. Concrete examples of cycloalkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methylcyclopentyl, methylcyclohexyl, ethylcyclopentyl, ethylcyclohexyl, adamantyl, dicyclopentadienyl, decahydronaphthyl, norbornyl, bornyl, and isobornyl. One or more hydrogen atoms on the cycloalkyl may be substituted by the same substituents as on the aryl. The term “alkoxy” as used in the compounds of the present disclosure refers to an alkyl or cycloalkyl singularly bonded to oxygen. Concrete examples of the alkoxy include methoxy, ethoxy, propoxy, isobutoxy, sec-butoxy, pentyloxy, iso-amyloxy, hexyloxy, cyclobutyloxy, cyclopentyloxy, adamantyloxy, dicyclopentyloxy, bornyloxy, and isobornyloxy. One or more hydrogen atoms on the alkoxy may be substituted by the same substituents as on the aryl. Concrete examples of the arylalkyl used in the compounds of the present disclosure include phenylmethyl (benzyl), phenylethyl, phenylpropyl, naphthylmethyl, and naphthylethyl. One or more hydrogen atoms on the arylalkyl may be substituted by the same substituents as on the aryl. Concrete examples of the silyl radicals used in the compounds of the present disclosure include trimethylsilyl, triethylsilyl, triphenylsilyl, trimethoxysilyl, dimethoxyphenylsilyl, diphenylmethylsilyl, diphenylvinlysilyl, methylcyclobutylsilyl, and dimethyl furylsilyl. One or more hydrogen atoms on the silyl may be substituted by the same substituents as on the aryl. As used herein, the term “alkenyl” refers to an unsaturated hydrocarbon group that contains a carbon-carbon double bond between two carbon atoms and the term “alkynyl” refers to an unsaturated hydrocarbon group that contains a carbon-carbon triple bond between two carbon atoms. As used herein, the term “alkylene” refers to an organic aliphatic radical regarded as derived from a linear or branched saturated hydrocarbon alkane by removal of two hydrogen atoms from different carbon atoms. Concrete examples of the alkylene include methylene, ethylene, propylene, isopropylene, isobutylene, sec-butylene, tert-butylene, pentylene, iso-amylene, hexylene, and so on. One or more hydrogen atoms on the alkylene may be substituted by the same substituents as on the aryl. Furthermore, as used herein, the term “diarylamino” refers to an amine radical having two identical or different aryl groups bonded to the nitrogen atom thereof, the term “diheteroarylamino” refers to an amine radical having two identical or different heteroaryl groups bonded to the nitrogen atom thereof, and the term “aryl(heteroaryl)amino” refers to an amine radical having an aryl group and a heteroaryl group both bonded to the nitrogen atom thereof. As more particular examples accounting for the term “substituted” in the expression “substituted or unsubstituted” used for compounds of Chemical Formulas A and B, the compounds may be substituted by at least one substituents selected from the group consisting of a deuterium atom, a cyano, a halogen, a hydroxy, a nitro, an alkyl of 1 to 12 carbon atoms, a halogenated alkyl of 1 to carbon atoms, an alkenyl of 2 to 12 carbon atoms, an alkynyl of 2 to 12 carbon atoms, a cycloalkyl of 3 to 12 carbon atoms, a heteroalkyl of 1 to 12 carbon atoms, an aryl of 6 to 18 carbon atoms, an arylalkyl of 7 to 20 carbon atoms, an alkylaryl of 7 to 20 carbon atoms, a heteroaryl of 2 to 18 carbon atoms, a heteroarylalkyl of 2 to 18 carbon atoms, an alkoxy of 1 to 12 carbon atoms, an alkylamino of 1 to 12 carbon atoms, a diarylamino of 12 to 18 carbon atoms, a diheteroarylamino of 2 to 18 carbon atoms, an aryl(heteroaryl)amino of 7 to 18 carbon atoms, an alkylsilyl of 1 to 12 carbon atoms, an arylsilyl of 6 to 18 carbon atoms, an aryloxy of 6 to 18 carbon atoms, an arylthionyl of 6 to 18 carbon atoms. The amine compound of the present invention, represented by Chemical Formula A or B, is characterized by the structure in which the 9,9-dimethyl fluorene moiety represented by diagram A, is bonded at position 7 (or 2) with the oxygen atom and in the benzofuran ring moiety and at position 6 (or 3) with an aromatic carbon atom near the carbon atom bonded to the oxygen atom in the benzofuran ring moiety to form an indenodibenzofuran fused ring structure which is the polycondensed ring system of “6-membered aromatic ring/5-membered ring having two methyl groups bound thereto/6-membered aromatic ring/5-membered ring bearing an oxygen atom/6-membered aromatic ring”, wherein the amine radical is connected to the terminal aromatic ring having the substituent Ar1thereon or to the middle 6-membered ring in the indenodibenzofuran ring structure of Chemical Formula A, or the amine radical is connected to the middle 6-membered ring having the substituent Ar1thereon or to the terminal 6-membered aromatic ring in the indenodibenzofuran ring structure of Chemical Formula B, wherein the substituent Ar1is selected from a substituted or unsubstituted alkyl of 1 to 30 carbon atoms, a substituted or unsubstituted haloalkyl of 1 to 30 carbon atoms, a substituted or unsubstituted aryl of 6 to 50 carbon atoms, a substituted or unsubstituted cycloalkyl of 3 to 30 carbon atoms, a substituted or unsubstituted heteroaryl of 2 to 50 carbon atoms, a substituted or unsubstituted alkylsilyl of 1 to 30 carbon atoms, and a substituted or unsubstituted arylsilyl of 5 to 30 carbon atoms. Compared to a structure in which the indenodibenzofuran framework has only the amine radical bonded thereto, the structural feature in which the substituent Ar1such as an aryl, a heteroaryl, an alkyl, etc. is further added to the middle aromatic ring or to the terminal aromatic ring of the indenodibenzofuran framework is made more planar and thus exhibits a higher deposition yield, whereby the organic light-emitting diode of the present disclosure can achieve high emission efficiency, compared to conventional organic light-emitting diodes. According to an embodiment, the amine radical in Chemical Formulas A and B may be connected to the aromatic ring which has the substituent Ar1bonded thereto. According to an embodiment, the amine radical in Chemical Formulas A and B may be connected to the aromatic ring which does not have the substituent Ar1bonded thereto. In an embodiment, the substituent Ar1in Chemical Formulas A and B may be one selected from a substituted or unsubstituted an aryl of 6 to 18 carbon atoms and a substituted or unsubstituted a heteroaryl of 2 to 18 carbon atoms, and the aryl of 6 to 18 carbon atoms for Ar1may be exemplified by phenyl, biphenyl, terphenyl, naphthalenyl, phenanthrenyl, and anthracenyl. In addition, the substituent Ar1may be represented by the following Structural Formula A: wherein “-*” denotes a bonding site at which the substituent Ar1is bonded to a carbon atom within the terminal aromatic ring or middle aromatic ring of the dibenzofuran moiety in Chemical Formulas A and B and, R41to R45, which may be the same or different, are each independently selected from a hydrogen, a deuterium, a cyano, a halogen, a hydroxy, a nitro, an alkyl of 1 to 24 carbon atoms, a cycloalkyl of 3 to 24 carbon atoms, a halogenated alkyl of 1 to 24 carbon atoms, an alkenyl of 2 to 24 carbon atoms, an alkynyl of 2 to carbon atoms, a heteroalkyl of 1 to 24 carbon atoms, an aryl of 6 to 24 carbon atoms, an arylalkyl of 7 to 24 carbon atoms, an alkylaryl of 7 to 24 carbon atoms, a heteroaryl of 2 to 50 carbon atoms, a heteroarylalkyl of 2 to 24 carbon atoms, an alkoxy of 1 to carbon atoms, an alkylamino of 1 to 24 carbon atoms, an arylamino of 6 to 24 carbon atoms, a heteroarylamino of 1 to 24 carbon atoms, an alkylsilyl of 1 to 24 carbon atoms, an arylsilyl of 6 to 24 carbon atoms, and an aryloxy of 6 to 24 carbon atoms. According to an embodiment, at least one of the substituents R1and R2in Chemical Formulas A and B may be any one selected from a substituted or unsubstituted an aryl of 6 to 18 carbon atoms and a substituted or unsubstituted a heteroaryl of 2 to 18 carbon atoms. In this regard, the substituents R1and R2, which may be same or different, may be each independently a substituted or unsubstituted an aryl of 6 to 18 carbon atoms or a substituted or unsubstituted a heteroaryl of 2 to 18 carbon atoms. When the substituents R1and R2, which may be same or different, are each independently an aryl of 6 to 18 carbon atoms, each of them may be selected from phenyl, biphenyl, and terphenyl. According to an embodiment, at least one of the substituents Ar1, R1, and R2in Chemical Formulas A and B may be a deuterium-substituted of 6 to 18 carbon atoms. In this regard, at least one of the substituents Ar1, R1, and R2may be a deuterium-substituted phenyl. In an embodiment, the substituent Ar1may be bonded to the terminal benzene ring of the dibenzofuran moiety at position 1, 2, 3, or 4 in Chemical Formula A, and the substituent Ar1may be bonded to middle aromatic ring of the dibenzofuran moiety at the carbon adjacent to the aromatic carbon atom bonded to the oxygen atom or at the carbon atom opposite thereto in Chemical Formula B. In an embodiment, the linker L1in Chemical Formula A or B may be selected from a single bond and a substituted or unsubstituted arylene of 6 to 18 carbon atoms or may be selected from a single bond, phenylene, naphthalenylene, and biphenylene. In addition, concrete examples of the amine compound represented by Chemical Formula A or B include the following [Compound 1] to [Compound 72]: In a particular embodiment thereof, the present invention provides an organic light-emitting diode comprising: a first electrode; a second electrode facing the first electrode; and an organic layer interposed between the first electrode and the second electrode, wherein the organic layer comprises at least one of the amine compounds represented by Chemical Formulas A and B. Having such structural characteristics, the organic light-emitting diode according to the present disclosure can drive at high luminous efficiency. In this regard, the organic light-emitting diode according to the present disclosure may include at least one of a hole injection layer, a hole transport layer, a functional layer capable of both hole injection and hole transport, an electron blocking layer, a light-emitting layer, an electron transport layer, an electron injection layer, and a capping layer. Throughout the description of the present disclosure, the phrase “(an organic layer) includes at least one organic compound” may be construed to mean that “(an organic layer) may include a single organic compound species or two or more difference species of organic compounds falling within the scope of the present disclosure”. In a further particular embodiment of the present disclosure, the organic light-emitting diode may comprise: the first electrode as an anode; the second electrode as a cathode; and the organic layer interposed between the anode and the cathode, the organic layer including a hole transport layer or a hole injection layer, wherein the amine compound according to the present disclosure may be used in the hole transport layer. That is, at least one of the amine compounds represented by Chemical Formulas A and B may be used as a material for a hole transport layer in the organic light-emitting diode. According to a particular embodiment, the hole transport layer may be divided into two or more sub-hole transport layers employing respective different materials therein, wherein the amine compound may use at least one of the sub-layers. In an exemplary embodiment thereof, the present disclosure provides an organic light-emitting comprising: a first electrode; a second electrode facing the first electrode; and an organic layer interposed between the first electrode and the second electrode and including at least one of the amine compounds represented by Chemical Formulas A and B, wherein the hole transport layer includes a first hole transport layer and a second hole transport layer different in material from the first hole transport layer and employing the amine compound. In addition, the present disclosure provides an organic light-emitting diode comprising: an anode as a first electrode; a cathode as a second electrode facing the first electrode; and a hole injection layer, a first hole transport layer, a second hole transport layer, a light-emitting layer, an electron transport layer, and an electron injection layer sequentially disposed in that order between the anode and the cathode, wherein the first hole transport layer and the second hole transport layer are different in terms of material and the second hole transport layer employs an amine compound represented by Chemical Formula A or B. Here, the light-emitting layer in the organic light-emitting diode of the present disclosure includes a host and a dopant. As the host, an anthracene derivative represented by the following Chemical Formula C may be used, but without limitations thereto: wherein. R11to R18, which may be the same or different, are each independently selected from a hydrogen, a deuterium, a substituted or unsubstituted alkyl of 1 to 30 carbon atoms, an alkenyl of 2 to 24 carbon atoms, an alkynyl of 2 to 24 carbon atoms, a substituted or unsubstituted aryl of 6 to 50 carbon atoms, a substituted or unsubstituted cycloalkyl of 3 to 30 carbon atoms, a substituted or unsubstituted heterocycloalkyl of 1 to 30 carbon atoms, a substituted or unsubstituted heteroaryl of 2 to 50 carbon atoms, a substituted or unsubstituted alkoxy of 1 to 30 carbon atoms, a substituted or unsubstituted aryloxy of 6 to 30 carbon atoms, a substituted or unsubstituted alkylthioxy of 1 to 30 carbon atoms, a substituted or unsubstituted arylthioxy of 6 to 30, a substituted or unsubstituted alkylamine of 1 to 30 carbon atoms, a substituted or unsubstituted arylamine of 6 to 30 carbon atoms, a substituted or unsubstituted alkylsilyl of 1 to 30 carbon atoms, a substituted or unsubstituted arylsilyl of 6 to 30 carbon atoms, a nitro, a cyano, and a halogen, Ar9and Ar10, which may be the same or different, are each independently selected from a substituted or unsubstituted alkyl of 1 to 30 carbon atoms, a substituted or unsubstituted aryl of 6 to 50 carbon atoms, a substituted or unsubstituted cycloalkyl of 3 to 30 carbon atoms, and a substituted or unsubstituted heteroaryl of 2 to carbon atoms; L13, which functions as a linker, is a single bond or is selected from a substituted or unsubstituted arylene of 6 to 20 carbon atoms, and a substituted or unsubstituted heteroarylene of 2 to 20 carbon atoms; and k is an integer of 1 to 3, wherein when k is 2 or greater, the corresponding L13's are each the same or different. For a more exemplary host, Ar9in Chemical Formula C may be a substituent represented by the following Chemical Formula C-1: wherein, R21to R25, which may be the same or different, are as defined for R1to R10, above; and may each be linked to an adjacent one to form a saturated or unsaturated cyclic ring. In this case, L13may be a single bond or a substituted or unsubstituted arylene of 6 to 20 carbon atoms, and k may be 1 or 2, with the proviso that when k is 2, corresponding L13's may be the same or different. According to one embodiment, the anthracene derivative may be one selected from the compounds represented by the following <Chemical Formula 22> to <Chemical Formula 60>: In addition, the light-emitting layer may employ as a dopant compound at least one selected from the compounds represented by the following Chemical Formulas D1 to D3: wherein,A31, A32, E1, and F1, which may be the same or different, are each independently a substituted or unsubstituted aromatic hydrocarbon ring of 6 to 50 carbon atoms, or a substituted or unsubstituted heteroaromatic ring of 2 to 40 carbon atoms;wherein two adjacent carbon atoms within the aromatic ring of A31and two adjacent carbon atoms within the aromatic ring of A32form a 5-membered ring with a carbon atom connected to both substituents R51and R52, thus establishing a fused ring structure;linkers L21to L32, which may be same or different, are each independently selected from a single bond, a substituted or unsubstituted alkylene of 1 to 60 carbon atoms, a substituted or unsubstituted alkenylene of 2 to 60 carbon atoms, a substituted or unsubstituted alkynylene of 2 to 60 carbon atoms, a substituted or unsubstituted cycloalkylene of 3 to 60 carbon atoms, a substituted or unsubstituted heterocycloalkylene of 2 to 60 carbon atoms, a substituted or unsubstituted arylene of 6 to 60 carbon atoms, and a substituted or unsubstituted heteroarylene of 2 to 60 carbon atoms; W is selected from N—R53, CR54R55, SiR56R57, GeR58R59, O, S, and Se; R51to R59and Ar21to Ar28, which may be same or different, are each independently selected from a hydrogen atom, a deuterium, a substituted or unsubstituted alkyl of 1 to 30 carbon atoms, a substituted or unsubstituted aryl of 6 to 50 carbon atoms, a substituted or unsubstituted alkenyl of 2 to 30 carbon atoms, a substituted or unsubstituted alkynyl of 2 to 20 carbon atoms, a substituted or unsubstituted cycloalkyl of 3 to 30 carbon atoms, a substituted or unsubstituted cycloalkenyl of 5 to 30 carbon atoms, a substituted or unsubstituted heteroaryl of 2 to 50 carbon atoms, a substituted or unsubstituted heterocycloalkyl of 2 to 30 carbon atoms, a substituted or unsubstituted alkoxy of 1 to 30 carbon atoms, a substituted or unsubstituted aryloxy of 6 to 30 carbon atoms, a substituted or unsubstituted alkylthioxy of 1 to 30 carbon atoms, a substituted or unsubstituted arylthioxy of 5 to 30 carbon atoms, a substituted or unsubstituted alkylamine of 1 to 30 carbon atoms, a substituted or unsubstituted arylamine of 5 to 30 carbon atoms, a substituted or unsubstituted alkylsilyl of 1 to 30 carbon atoms, a substituted or unsubstituted arylsilyl of 5 to 30 carbon atoms, a substituted or unsubstituted alkyl germanium of 1 to 30 carbon atoms, a substituted or unsubstituted aryl germanium of 1 to 30 carbon atoms a cyano, a nitro, and a halogen, wherein R51and R52may be connected to each other to form a mono- or polycyclic aliphatic or aromatic ring which may bear at least one heteroatom selected from N, O, P, Si, S, Ge, Se, and Te as a ring member; p11 to p14, r11 to r14, and s11 to s14 are each independently an integer of 1 to 3, under which when any of them is 2 or greater, the corresponding linkers L21to L32may be the same or different, x1 is an integer of 1 or 2, and y1 and z1, which may be the same or different, are each independently an integer of 0 to 3, a bond may be made between at least one pair selected from Ar21and Ar22, Ar23and Ar24, Ar25and Ar26, and Ar27and Ar28to form a ring; two adjacent carbon atoms of the A32ring moiety of Chemical Formula D1 may occupy respective positions * of Structural Formula Q11to form a fused ring, two adjacent carbon atoms of the A31ring moiety of Chemical Formula D2 may occupy respective positions * of Structural Formula Q12to form a fused ring and two adjacent carbon atoms of the A32ring moiety of Chemical Formula D2 may occupy respective positions * of structural Formula Q11to form a fused ring wherein, X1is selected from B, P, and P═O, T1 to T3, which may be the same or different, are each independently a substituted or unsubstituted aromatic hydrocarbon ring of 6 to 50 carbon atoms, or a substituted or unsubstituted heteroaromatic ring of 2 to 40 carbon atoms; Y1is selected from N—R61, CR62R63, O, S, and SiR64R65; Y2is selected from N—R66, CR66R68, O, S, and SiR69R70; wherein R61to R70, which may be the same or different, are each independently selected from a hydrogen atom, a deuterium, a substituted or unsubstituted alkyl of 1 to 30 carbon atoms, a substituted or unsubstituted aryl of 6 to 50 carbon atoms, a substituted or unsubstituted cycloalkyl of 3 to 30 carbon atoms, a substituted or unsubstituted heteroaryl of 2 to 50 carbon atoms, a substituted or unsubstituted alkoxy of 1 to 30 carbon atoms, a substituted or unsubstituted aryloxy of 6 to 30 carbon atoms, a substituted or unsubstituted alkylthioxy of 1 to 30 carbon atoms, a substituted or unsubstituted arylthioxy of 5 to 30 carbon atoms, a substituted or unsubstituted alkylamine of 1 to 30 carbon atoms, a substituted or unsubstituted arylamine of 5 to 30 carbon atoms, a substituted or unsubstituted alkylsilyl of 1 to 30 carbon atoms, a substituted or unsubstituted arylsilyl of 5 to 30 carbon atoms, a cyano, and a halogen, wherein R61to R70may each be linked to at least one of T1 to T3 to further form a mono- or polycyclic aliphatic or aromatic ring. Here, the term “substituted” in the expression “substituted or unsubstituted” used for Chemical Formulas D1 to D3 means having at least one substituent selected from the group consisting of a deuterium, a cyano, a halogen, a hydroxy, a nitro, an alkyl of 1 to carbon atoms, a halogenated alkyl of 1 to 24 carbon atoms, an alkenyl of 2 to 24 carbon atoms, an alkynyl of 2 to 24 carbon atoms, a heteroalkyl of 1 to 24 carbon atoms, an aryl of 6 to 24 carbon atoms, an arylalkyl of 7 to 24 carbon atoms, a heteroaryl of 2 to 24 carbon atoms or a heteroarylalkyl of 2 to 24 carbon atoms, an alkoxy of 1 to 24 carbon atoms, an alkylamino of 1 to 24 carbon atoms, an arylamino of 6 to 24 carbon atoms, a heteroarylamino of 1 to 24 carbon atoms, an alkylsilyl of 1 to 24 carbon atoms, an arylsilyl of 6 to 24 carbon atoms, and an aryloxy of 6 to 24 carbon atoms. Among the dopant compounds according to the present disclosure, the boron compound represented by Chemical Formula D3 may have on the aromatic hydrocarbon rings or heteroaromatic rings of T1 to T3 a substituent selected from a deuterium, an alkyl of 1 to 24 carbon atoms, an aryl of 6 to 24 carbon atoms, an alkylamino of 1 to 24 carbon atoms, and an arylamino of 6 to 24 carbon atoms, wherein the alkyl radicals or the aryl radicals in the alkylamino of 1 to 24 carbon atoms and the arylamino of 6 to 24 carbon atoms on the rings may be linked to each other, and particularly selected from an alkyl of 1 to 12 carbon atoms, an aryl of 6 to 18 carbon atoms, an alkylamino of 1 to 12 carbon atoms, and an arylamino of 6 to 18 carbon atoms, wherein the alkyl radicals or aryl radicals in the alkylamino of 1 to 12 carbon atoms and the arylamino of 6 to 18 carbon atoms on the rings may be linked to each other. In addition, concrete examples of the dopant compound used for the light-emitting layer, represented by one of Chemical Formulas D1 and D2, include compounds represented by the following Chemical Formulas d1 to D239: In addition, concrete examples of the compound represented by Chemical Formula D3 include the compounds represented by the following Chemical Formulas D101 to D130: The content of the dopant in the light-emitting layer may range from about 0.01 to 20 parts by weight, based on 100 parts by weight of the host, but is not limited thereto. In addition to the above-mentioned dopants and hosts, the light-emitting layer may further include various hosts and dopant materials. Below, the organic light-emitting diode of the present disclosure is explained with reference toFIG.1. FIG.1is a schematic cross-sectional view of the structure of an organic light-emitting diode according to an embodiment of the present disclosure. As shown inFIG.1, the organic light-emitting diode according to an embodiment of the present disclosure comprises an anode20, a hole transport layers40and45, an organic light-emitting layer50containing a host and a dopant, an electron transport layer60, and a cathode80, wherein the anode and the cathode serve as a first electrode and a second electrode, respectively, with the interposition of the two hole transport layers40and45between the anode and the light-emitting layer, and the electron transport layer between the light-emitting layer and the cathode. The hole transport layers40and45are composed of a first hole transport layer40and a second hole transport layer45, wherein the second hole transport layer45is interposed between the first hole transport layer40and the light-emitting layer50. In this regard, the amine compound represented by Chemical Formula A may be used as a material for the second hole transport layer45in the organic light-emitting diode of the present disclosure. Having such a structural characteristic, the organic light-emitting diode according to the present disclosure can be driven with high luminous efficiency. Furthermore, the organic light-emitting diode according to an embodiment of the present disclosure may comprise a hole injection layer30between the anode20and the hole transport layers40and45, and an electron injection layer70between the electron transport layer60and the cathode80. Reference is made toFIG.1with regard to the organic light emitting diode of the present disclosure and the fabrication thereof. First, a substrate10is coated with an anode electrode material to form an anode20. So long as it is used in a typical organic light emitting diode, any substrate may be used as the substrate10. Preferable is an organic substrate or transparent plastic substrate that exhibits excellent transparency, surface smoothness, ease of handling, and waterproofness. As the anode electrode material, indium tin oxide (ITO), indium zinc oxide (IZO), tin oxide (SnO2), or zinc oxide (ZnO), which are transparent and superior in terms of conductivity, may be used. A hole injection layer material is applied on the anode20by thermal deposition in a vacuum or by spin coating to form a hole injection layer30. As a material for the hole injection layer30, the amine compound represented by Chemical Formula A may be used. No particular limitations are imparted to the hole injection layer material, as long as it is one that is typically used in the art. For example, mention may be made of 2-TNATA [4,4′,4″-tris(2-naphthylphenyl-phenylamino)-triphenylamine], NPD[N, N′-di(1-naphthyl)-N,N′-diphenylbenzidine)], TPD[N,N′-diphenyl-N,N′-bis(3-methylphenyl)-1,1′-biphenyl-4,4′-diamine], DNTPD[N,N′-diphenyl-N,N′-bis-[4-(phenyl-m-tolyl-amino)-phenyl]-biphenyl-4,4′-diamine], but the present disclosure is not limited thereby. Subsequently, thermal deposition in a vacuum or by spin coating may also be conducted to form a hole transport layer40with a hole transport layer material on the hole injection layer30. When the hole transport layer is composed of two or more sub-layers, each of them may be separately formed by deposition or spin coating. In addition, the amine compound represented by Chemical Formula A may be contained as a material for the hole transport layers40and45. In a particular embodiment, the hole transport layer is composed of a first hole transport layer40and a second hole transport layer45, wherein the amine compound represented by Chemical Formula A is used in the second hole transport layer45. So long as it is typically used in the art, any material may be selected for the first hole transport layer without particular limitation. Examples include, but are not limited to, N,N′-bis(3-methylphenyl)-N,N′-diphenyl-[1,1-biphenyl]-4,4′-diamine (TPD) and N,N′-di(naphthalen-1-yl)-N,N′-diphenylbenzidine (a-NPD). Then, an organic light-emitting layer50is deposited on the hole transport layers40and45by deposition in a vacuum or by spin coating. Herein, the light-emitting layer50may contain a host and a dopant and the materials are as described above. In some embodiments of the present disclosure, the light-emitting layer50particularly ranges in thickness from 50 to 2,000 Å. Here, an electron transport layer60is deposited on the organic light emitting layer by deposition in a vacuum or by spin coating. A material for use in the electron transport layer60functions to stably carry the electrons injected from the electron injection electrode (cathode), and may be an electron transport material known in the art. Examples of the electron transport material known in the art include quinoline derivatives, particularly, tris(8-quinolinorate)aluminum (Alq3), Liq, TAZ, BAlq, beryllium bis(benzoquinolin-10-olate) (Bebq2), Compound 201, Compound 202, BCP, and oxadiazole derivatives such as PBD, BMD, and BND, but are not limited thereto: In the organic light emitting diode of the present disclosure, an electron injection layer (EIL) that functions to facilitate electron injection from the cathode may be deposited on the electron transport layer. The material for the EIL is not particularly limited. Any material that is conventionally used in the art can be available for the electron injection layer without particular limitations. Examples include CsF, NaF, LiF, Li2O, and BaO. Deposition conditions for the electron injection layer may vary, depending on compounds used, but may be generally selected from condition scopes that are almost the same as for the formation of hole injection layers. The electron injection layer may range in thickness from about 1 Å to about 100 Å, and particularly from about 3 Å to about 90 Å. Given the thickness range for the electron injection layer, the diode can exhibit satisfactory electron injection properties without actually elevating a driving voltage. In order to facilitate electron injection, the cathode80may be made of a material having a small work function, such as metal or metal alloy such as lithium (Li), magnesium (Mg), calcium (Ca), aluminum (Al), aluminum-lithium (Al—Li), magnesium-indium (Mg—In), and magnesium-silver (Mg—Ag). Alternatively, ITO or IZO may be employed to form a transparent cathode for a top-emitting organic light-emitting diode. Moreover, the organic light-emitting diode of the present disclosure may further comprise a light-emitting layer containing a blue, green, or red luminescent material that emits radiations in the wavelength range of 380 nm to 800 nm. That is, the light-emitting layer in the present disclosure has a multi-layer structure wherein the blue, green, or red luminescent material may be a fluorescent material or a phosphorescent material. Furthermore, at least one selected from among the layers may be deposited using a single-molecule deposition process or a solution process. Here, the deposition process is a process by which a material is vaporized in a vacuum or at a low pressure and deposited to form a layer, and the solution process is a method in which a material is dissolved in a solvent and applied for the formation of a thin film by means of inkjet printing, roll-to-roll coating, screen printing, spray coating, dip coating, spin coating, etc. Also, the organic light-emitting diode of the present disclosure may be applied to a device selected from among flat display devices, flexible display devices, monochrome or grayscale flat illumination devices, and monochrome or grayscale flexible illumination devices. A better understanding of the present disclosure may be obtained through the following examples which are set forth to illustrate, but are not to be construed as limiting the present invention. Preparation Example 1: Synthesis of Compound 1 Synthesis of Intermediate 1-1 In a reactor, 2-bromo-dimethylfluorene (100 g, 0.366 mol) and methanol (1,000 mL) were stirred together and NaOMe (114 g, 2.196 mol) and CuI (20 g, 0.110 mol) were added dropwise before stirring overnight under reflux. After completion of the reaction, drops of distilled water (100 mL) were added. Subsequently, the reaction mixture was filtered. The filtrate was dissolved in methylene chloride (300 mL) and purified through a column. After enrichment, recrystallization in an excess of methanol afforded Intermediate 1-1 (69 g, yield 84%). Synthesis of Intermediate 1-2 In a reactor, Intermediate 1-1 (69 g, 0.308 mol) was stirred together with IMF (897 mL). NBS (58.15 g, 0.327 mol) was dropwise added at room temperature and stirred overnight. After completion of the reaction, drops of distilled water (69 mL) were added. The reaction mixture was filtered, dissolved in methylene chloride (207 mL), and purified through a column. After enrichment, recrystallization in an excess of methanol afforded Intermediate 1-2 (60 g, yield 64%). Synthesis of Intermediate 1-3 In a reactor, Intermediate 1-2 (46 g, 0.157 mol) was stirred together with THF (460 mL) and chilled to −78° C. at which drops of n-BuLi (113 mL, 0.182 mol) were slowly added. After 2 hours, trimethyl borate (15.85 g, 0.187 mol) was dropwise added. The reaction was terminated with an excess of 2M HCl. Extraction was made with ethyl acetate/distilled water, and the organic layer thus formed was pooled, concentrated, and added to methanol. Filtration and drying afforded Intermediate 1-3 (19 g, yield 50%). Synthesis of Intermediate 1-4 In a reactor, Intermediate 1-3 (20 g, 0.074 mol), 1-bromo-2-fluoro-3-iodobenzene (26.95 g, 0.089 mol), Pd(pph3)4 (1.638 g, 0.0014 mol), hydrazine (0.11 g, 0.0022 mol), and sodium tetraborate (19.5 g, 0.097 mol) were placed, added with 1,4-dioxane (140 mL) and distilled water (60 ml), and stirred together overnight at 90° C. Extraction was made with ethyl acetate/distilled water and the organic layer thus formed was concentrated and purified through a column to afford Intermediate 1-4 (20 g, yield 68%). Synthesis of Intermediate 1-5 Intermediate 1-4 (20 g, 0.055 mol) and methylene chloride (200 mL) was placed in a reactor, cooled to 0° C., and added with drops of BBr3 (18.8 g, 0.076 mol). The mixture was warmed to room temperature, stirred for 5 hours, and added with drops of distilled water (600 mL). Extraction was made with ethyl acetate/distilled water. The organic layer thus formed was concentrated to afford Intermediate 1-5 (19 g, yield 94%). Synthesis of Intermediate 1-6 In a reactor, Intermediate 1-5 (19 g, 0.049 mol), potassium carbonate (13.7 g, 0.098 mol), and NMP (190 mL) were stirred together for 3 hours under reflux. The reaction mixture was cooled to room temperature and added with drops of 2M HCl (570 mL, 30 vol.) to form precipitates. The precipitates were filtered and then slurried in methanol to afford Intermediate 1-6 (17 g, yield 94%). Synthesis of Intermediate 1-7 Intermediate 1-6 (17 g, 0.046 mol), phenyl boronic acid (6.2 g, 0.051 mol), potassium carbonate (12.9 g, 0.093 mol), pd(pph3)4 (1.6 g, 0.001 mol), toluene (119 mL), ethanol (51 mL), and distilled water (34 mL) were stirred together overnight under reflux. The reaction mixture was cooled to room temperature and subjected to extraction with ethyl acetate/distilled water. The organic layer thus formed was concentrated and purified through a column to afford Intermediate 1-7 (16 g, 95%). Synthesis of Intermediate 1-8 In a reactor, Intermediate 1-7 (16 g, 0.044 mol) and THF (160 mL) were chilled to −78° C. and slowly added with drops of n-BuLi (33.2 mL, 0.053 mol). Thereafter, the mixture was warmed to room temperature and stirred for 12 hours. Trimethyl borate (0.053 mol) was dropwise added, followed by terminating the reaction with an excess of 2M HCl. The reaction mixture was subjected to extraction with ethyl acetate/distilled water and the organic layer thus formed was pooled and concentrated. The concentrate was slurried in methanol, filtered, and dried to afford Intermediate 1-8 (8 g, yield 37%). Synthesis of Compound 1 In a reactor, Intermediate 1-8 (8 g, 0.019 mol) and N,N-bis(4-biphenylyl)-N-(4-bromophenyl)amine (10.3 g, 0.021 mol), Pd (pph3) 4 (0.45 g, 0.0004 mol), and potassium carbonate (5.25 g, 0.038 mol) were stirred together with toluene (56 mL), ethanol (24 ml), and distilled water (16 ml), overnight under reflux. Extraction was made with ethyl acetate/distilled water and the organic layer thus formed was concentrated and hot filtered with toluene. The filtrate was concentrated and recrystallized in acetone to afford Compound 1 (3 g, yield 20%). Preparation Example 2. Synthesis of Compound 20 Synthesis of Intermediate 2-1 In a reactor, SM (15 g, 0.041 mol) synthesized with reference to Korean Patent Number KR 2018-0037889 Å, bis(4-biphenylyl)amine (14.6 g, 0.045 mol), Pd2dba3 (0.76 g, 0.003 mol), tributyl phosphine (0.25 g, 0.001 mol), sodium tert-butoxide (4.37 g, 0.045 mol), and toluene (150 mL) were stirred together for 3 hour under reflux. The reaction mixture was hot filtered with toluene and subjected to extraction with ethyl acetate/distilled water. The organic layer thus formed was concentrated and purified through a column to afford Intermediate 2-1 (16 g, yield 64%). Synthesis of Intermediate 2-2 In a reactor, Intermediate 2-1 (16 g, 0.026 mol) and IMF (160 mL) were chilled to 0° C., added with drops of NBS (4.71 g, 0.026 mol), and stirred for 6 hours. After completion of the reaction, the reaction mixture was added with drops of distilled water (480 mL), filtered, and slurried in an excess of methanol to afford Intermediate 2-2 (11 g, yield 61%). Synthesis of Compound 20 In a reactor, Intermediate 2-2 (11 g, 0.016 mol), phenyl boronic acid (2.1 g, 0.017 mol), potassium carbonate (4.4 g, 0.032 mol), pd(pph3)4 (0.37 g, 0.0003 mol), toluene (77 mL), ethanol (33 mL), and distilled water (22 mL) were stirred overnight under reflux. The reaction mixture was added with drops of methanol (330 mL), filtered, hot filtered with toluene, and recrystallized in acetone to afford Compound 20 (3 g, yield 30%). Preparation Example 3: Synthesis of Compound 55 Synthesis of Intermediate 3-1 In a reactor, the starting material (100 g, 0.366 mol) and methanol (1000 mL) were stirred together. NaOMe (114 g, 2.196 mol) and CuI (20 g, 0.110 mol) were added dropwise, followed by stirring overnight under reflux. After completion of the reaction, distilled water (100 mL) was added. The reaction mixture was filtered and the filtrate was dissolved in dichloromethane (300 mL) and allowed to pass through a column. After concentration, recrystallization in an excess of methanol afforded Intermediate 3-1 (69 g, yield 84%). Synthesis of Intermediate 3-2 In a reactor, Intermediate 3-1 (69 g, 0.308 mol) and DMF (897 mL) were stirred together. Then, NBS (58.15 g, 0.327 mol) was dropwise added for 1 hour and stirred overnight at room temperature. After completion of the reaction, distilled water (69 mL) was dropwise added. The reaction mixture was filtered and the filtrate was dissolved and allowed to pass through a column. After concentration, recrystallization in an excess of methanol afforded Intermediate 3-2 (60 g, yield 64%). Synthesis of Intermediate 3-3 In a reactor, Intermediate 3-2 (46 g, 0.157 mol) and THF (460 mL) were stirred together and chilled to −78° C. before slowly adding drops of n-BuLi (113 mL, 0.182 mol). After 2 hours, trimethyl borate (15.85 g, 0.187 mol) was dropwise added. At room temperature, the reaction was terminated with an excess of 2 M HCl. The reaction mixture was subjected to extraction with ethyl acetate/distilled water and the organic layer thus formed was pooled and concentrated. The concentrate was added with methanol, filtered, and dried to afford Intermediate 3-3 (20 g, yield 50%). Synthesis of Intermediate 3-4 In a reactor, Intermediate 3-3 (20 g, 0.074 mol), 1-bromo-4-chloro-2-fluorobenzene (18.74 g, 0.089 mol), Pd(pph3)4 (1.638 g, 0.0014 mol), hydrazine (0.11 g, 0.022 mol), and sodium tetraborate (19.5 g, 0.097 mol) were placed and stirred, together with 1,4-dioxane (140 mL) and distilled water (60 ml), overnight at 90° C. Extraction was made with ethyl acetate/distilled water and the organic layer thus formed was purified through a column to afford Intermediate 3-4 (20 g, yield 68%). Synthesis of Intermediate 3-5 In a reactor, Intermediate 3-4 (20 g, 0.056 mol) and dichloromethane (200 mL) was stirred together. Br2 (10.87 g, 0.061 mol) was dropwise added and then stirred for 1 hour. After completion of the reaction, recrystallization in an excess of methanol afforded Intermediate 3-5 (21 g, yield 86%). Synthesis of Intermediate 3-6 In a reactor, Intermediate 3-5 (21 g, 0.048 mol), phenyl boronic acid (7.1 g 0.058 mol), Pd(pph3)4 (1.12 g, 0.00096 mol), and potassium carbonate (13.3 g, 0.097 mol) were stirred, together with toluene (147 mL), ethanol (63 ml), and distilled water (42 ml), overnight at 90° C. Extraction was made with ethyl acetate/distilled water and the organic layer thus formed was concentrated and purified through a column to afford Intermediate 3-6 (18 g, yield 86%). Synthesis of Intermediate 3-7 In a reactor, a mixture of Intermediate 3-6 (18 g, 0.041 mol) and dichloromethane (180 mL) was chilled to 0° C., followed by adding drops of BBr3 (12.6 g, 0.051 mol). At room temperature, the mixture was stirred for 5 hours and added with drops of distilled water (540 mL). Extraction was made with ethyl acetate/distilled water and the organic layer thus formed was concentrated to afford Intermediate 3-7 (15 g, yield 86%). Synthesis of Intermediate 3-8 In a reactor, Intermediate 3-7 (15 g, 0.036 mol), potassium carbonate (9.99 g, 0.0723 mol), and NMP (150 mL) were stirred together at 150° C. for 3 hours. The reaction mixture was cooled to room temperature and added with drops of 2M HCl (450 mL). The solid thus formed was filtered and slurried in methanol to afford Intermediate 3-8 (12 g, yield 84%). Synthesis of Compound 55 In a reactor, Intermediate 3-8 (12 g, 0.031 mol), N-phenyl[1,1′-biphenyl]-4-amine (8.2 g, 0.0033 mol), bis(tri-tert-butlyphosphine)palldium (0.31 g, 0.0004 mol), sodium tert-butoxide (5.84 g, 0.061 mol), and toluene (144 ml) were stirred overnight at 100° C. Extraction was made with ethyl acetate/distilled water and the organic layer thus formed was concentrated and hot filtered in toluene. The filtrate was concentrated and recrystallized in dichloromethane and acetone to afford Compound 55 (5 g, yield 37%). Preparation Example 4: Synthesis of Compound 67 The same procedure as in Preparation Example 1 was carried out, with the exception of using N-[1,1′-biphenyl]-4-yl-N-(4-bromophenyl)-1-dibenzofuranamine instead of N, N-bis(4-biphenylyl)-N-(4-bromophenyl)amine, to afford Compound 67 (4.5 g, yield 27%). Preparation Example 5: Synthesis of Compound 68 The same procedure as in Preparation Example 1 was carried out, with the exception of using 2-naphthyl boronic acid instead of phenyl boronic acid for the synthesis of Intermediate 1-7 to afford Compound (5 g, yield 25%). Preparation Example 6: Synthesis of Compound 69 The same procedure as in Preparation Example 1 was carried out, with the exception of using 9-phenanthrene boronic acid instead of phenyl boronic acid for the synthesis of Intermediate 1-7 to afford Compound 69 (4.2 g, yield 21%). Preparation Example 7: Synthesis of Compound 70 The same procedure as in Preparation Example 2 was carried out, with the exception of using 9-phenanthrene boronic acid instead of phenyl boronic acid for the synthesis of Compound 20 to afford Compound 70 (4.5 g, yield 28%). Preparation Example 8: Synthesis of Compound 71 The same procedure as in Preparation Example 2 was carried out, with the exception of using N-[4-(1-naphthalenyl)phenyl][1,1′-biphenyl]-4-amine instead of bis(4-biphenylyl)amine for the synthesis of Intermediate 2-1 to afford Compound 71 (4 g, yield 29%). Preparation Example 9: Synthesis of Compound 72 The same procedure as in Preparation Example 2 was carried out, with the exception of using 2-naphthyl boronic acid instead of phenyl boronic acid for the synthesis of Compound 20 to afford Compound 72 (3.7 g, yield 32%). Examples 1 to 9: Fabrication of Organic Light-Emitting Diodes An ITO glass substrate was patterned to have a translucent area of 2 mm×2 mm and cleansed. The ITO glass was mounted in a vacuum chamber that was then set to have a base pressure of 1×10−7torr. On the ITO glass substrate, films were sequentially formed of DNTPD (450 Å), [Chemical Formula G] (200 Å), and the compound listed in Table 1, below, as a material for the second hole transport layer (50 Å). Subsequently, a light-emitting layer (250 Å) was formed of a combination of [Chemical Formula BH] and [Chemical Formula BD] (97:3). Then, [Chemical Formula E-2] was deposited to form an electron transport layer (300 Å) on which an electron injecting layer of [Chemical Formula E-1] (10 Å) was formed and then covered with an Al layer (1000 Å) to fabricate an organic light-emitting diode. The organic light-emitting diodes thus obtained were measured at 0.4 mA for luminescence properties: Comparative Examples 1 to 3 Organic light emitting diodes were fabricated in the same manner as in the Examples, with the exception of using the following Chemical Formula B, C, or D instead of the compounds according to the present disclosure. The luminescence of the organic light-emitting diodes thus obtained was measured at 0.4 mA. The organic light emitting diodes fabricated in Examples 1 to 9 and Comparative Examples 1 to 3 were measured for driving voltage and efficiency, and the results are summarized in Table, below. TABLE 1DrivingEfficiencyNo.Volt. (V)(Cd/A)Ex. 1Compound 13.968.51Ex. 2Compound 204.018.42Ex. 3Compound 553.938.15Ex. 4Compound 673.828.48Ex. 5Compound 683.958.54Ex. 6Compound 693.988.67Ex. 7Compound 703.968.62Ex. 8Compound 713.948.50Ex. 9Compound 723.958.57C. Ex. 1Chemical4.047.60Formula BC. Ex. 2Chemical3.957.85Formula CC. Ex. 3Chemical3.967.36Formula D As is understood from data of Table 1, the organic light-emitting diodes according to the present disclosure exhibited excellent luminous efficiency, compared to those of the Comparative Examples 1 to 3.
53,927
11858907
DETAILED DESCRIPTION As used herein the following definitions shall apply unless otherwise indicated. Further many of the groups defined herein can be optionally substituted. The listing of substituents in the definition is exemplary and is not to be construed to limit the substituents defined elsewhere in the specification. The term “alkyl” refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, and 1,1-dimethylethyl (t-butyl). The term “(C1-6)alkyl” refers to an alkyl group as defined above having up to 6 carbon atoms. The term “alkenyl” refers to an aliphatic hydrocarbon group containing a carbon-carbon double bond and which may be a straight or branched or branched chain having about 2 to about 10 carbon atoms, e.g., ethenyl, 1-propenyl, 2-propenyl (allyl), iso-propenyl, 2-methyl-1-propenyl, 1-butenyl, and 2-butenyl. The term “(C2-6)alkenyl” refers to an alkenyl group as defined above having up to 6 carbon atoms. The term “alkynyl” refers to a straight or branched chain hydrocarbyl radical having at least one carbon-carbon triple bond, and having in the range of 2 to up to 12 carbon atoms (with radicals having in the range of 2 to up to 10 carbon atoms presently being preferred) e.g., ethynyl, propynyl, and butnyl. The term “(C2-6) alkynyl” refers to an alkynyl group as defined above having up to 6 carbon atoms. The term “alkoxy” denotes an alkyl, cycloalkyl, or cycloalkylalkyl group as defined above attached via an oxygen linkage to the rest of the molecule. The term “substituted alkoxy” refers to an alkoxy group where the alkyl constituent is substituted (i.e., —O-(substituted alkyl) wherein the term “substituted alkyl” is the same as defined above for “alkyl”. For example “alkoxy” refers to the group —O-alkyl, including from 1 to 8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, and cyclohexyloxy. The term “cycloalkyl” denotes a non-aromatic mono or multicyclic ring system of about 3 to 12 carbon atoms such as cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Examples of multicyclic cycloalkyl groups include perhydronaphthyl, adamantyl and norbornyl groups, bridged cyclic groups, and sprirobicyclic groups, e.g., sprio (4,4) non-2-yl. The term “(C3-8) cycloalkyl” refers to a cycloalkyl group as defined above having up to 8 carbon atoms. The term “cycloalkylalkyl” refers to a cyclic ring-containing radical containing in the range of about 3 up to 8 carbon atoms directly attached to an alkyl group which are then attached to the main structure at any carbon from the alkyl group that results in the creation of a stable structure such as cyclopropylmethyl, cyclobutylethyl, and cyclopentylethyl. The term “cycloalkenyl” refers to cyclic ring-containing radicals containing in the range of about 3 up to 8 carbon atoms with at least one carbon-carbon double bond such as cyclopropenyl, cyclobutenyl, and cyclopentenyl. The term “cycloalkenylalkyl” refers to a cycloalkenyl group directly attached to an alkyl group which are then attached to the main structure at any carbon from the alkyl group that results in the creation of a stable structure. The term “aryl” refers to aromatic radicals having in the range of 6 up to 20 carbon atoms such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, and biphenyl. The term “arylalkyl” refers to an aryl group as defined above directly bonded to an alkyl group as defined above, e.g., —CH2C6H5and —C2H5C6H5. The term “heterocyclic ring” refers to a non-aromatic 3 to 15 member ring radical which consists of carbon atoms and at least one heteroatom selected from nitrogen, phosphorus, oxygen and sulfur. For purposes of this invention, the heterocyclic ring radical may be a mono-, bi-, tri- or tetracyclic ring system, which may include fused, bridged or spiro ring systems, and the nitrogen, phosphorus, carbon, oxygen or sulfur atoms in the heterocyclic ring radical may be optionally oxidized to various oxidation states. In addition, the nitrogen atom may be optionally quaternized. The heterocyclic ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure. The term “heterocyclyl” refers to a heterocylic ring radical as defined above. The heterocylcyl ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure. The term “heterocyclylalkyl” refers to a heterocylic ring radical as defined above directly bonded to an alkyl group. The heterocyclylalkyl radical may be attached to the main structure at carbon atom in the alkyl group that results in the creation of a stable structure. Examples of such heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. The term “heteroaryl” refers to an optionally substituted 5 to 14 member aromatic ring having one or more heteroatoms selected from N, O, and S as ring atoms. The heteroaryl may be a mono-, bi- or tricyclic ring system. Examples of such “heterocyclic ring” or “heteroaryl” radicals include, but are not limited to, oxazolyl, thiazolyl, imidazolyl, pyrrolyl, furanyl, pyridinyl, pyrimidinyl, pyrazinyl, benzofuranyl, indolyl, benzothiazolyl, benzoxazolyl, carbazolyl, quinolyl, isoquinolyl, azetidinyl, acridinyl, benzodioxolyl, benzodioxanyl, benzofuranyl, carbazolyl, cinnolinyl, dioxolanyl, indolizinyl, naphthyridinyl, perhydroazepinyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, quinazolinyl, quinoxalinyl, tetrazoyl, tetrahydroisoquinolyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxoazepinyl, azepinyl, 4-piperidonyl, pyrrolidinyl, pyridazinyl, oxazolinyl, oxazolidinyl, triazolyl, indanyl, isoxazolyl, isoxazolidinyl, morpholinyl, thiazolinyl, thiazolidinyl, isothiazolyl, quinuclidinyl, isothiazolidinyl, isoindolyl, indolinyl, isoindolinyl, octahydroindolyl, octahydroisoindolyl, decahydroisoquinolyl, benzimidazolyl, thiadiazolyl, benzopyranyl, tetrahydrofuryl, tetrahydropyranyl, thienyl, benzothienyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, dioxaphospholanyl, oxadiazolyl, chromanyl, and isochromanyl. The heteroaryl ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure. The term “substituted heteroaryl” also includes ring systems substituted with one or more oxide (—O—) substituents, such as pyridinyl N-oxides. The term “heteroarylalkyl” refers to a heteroaryl ring radical as defined above directly bonded to an alkyl group. The heteroarylalkyl radical may be attached to the main structure at any carbon atom from alkyl group that results in the creation of a stable structure. The term “cyclic ring” refers to a cyclic ring containing 3 to 10 carbon atoms. The term “substituted” unless otherwise specified, refers to substitution with any one or any combination of the following substituents which may be the same or different and are independently selected from hydrogen, hydroxy, halogen, carboxyl, cyano, nitro, oxo (═O), thio (═S), substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted cycloalkenyl, substituted or unsubstituted cycloalkenylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroarylalkyl, substituted or unsubstituted heterocyclic ring, substituted heterocyclylalkyl ring, substituted or unsubstituted guanidine, —COORx, —C(O)Rx, —C(S)Rx, —C(O)NRxRy, —C(O)ONRxRy, —NRyRz, —NRxCONRyRz, —N(Rx)SORy, —N(Rx)SO2Ry, —(═N—N(Rx)Ry), —NRxC(O)ORy, —NRxRy, —NRxC(O)Ry—, —NRxC(S)Ry—NRxC(S)NRyRz, —SONRxRy—, —SO2NRxRy—, —ORx, —ORxC(O)NRyRz, —ORxC(O)ORy—, —OC(O)Rx, —OC(O)NRxRy, —RxNRyC(O)Rz, —RxORy, —RxC(O)ORy, —RxC(O)NRyRz, —RxC(O)Rx, —RxOC(O)Ry, —SRx, —SORx, —SO2Rx, and —ONO2, wherein Rx, Ryand Rzin each of the above groups can be hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted cycloalkenyl, substituted or unsubstituted amino, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroarylalkyl, substituted or unsubstituted heterocyclic ring, or substituted heterocyclylalkyl ring, or any two of Rx, Ryand Rzmay be joined to form a substituted or unsubstituted saturated or unsaturated 3-10 membered ring, which may optionally include heteroatoms which may be the same or different and are selected from O, NRx(e.g., Rxcan be hydrogen or C1-6alkyl) or S. Substitution or the combinations of substituents envisioned by this invention are preferably those that result in the formation of a stable or chemically feasible compound. The term stable as used herein refers to the compounds or the structure that are not substantially altered when subjected to conditions to allow for their production, detection and preferably their recovery, purification and incorporation into a pharmaceutical composition. The substituents in the aforementioned “substituted” groups cannot be further substituted. For example, when the substituent on “substituted alkyl” is “substituted aryl”, the substituent on “substituted aryl” cannot be “substituted alkenyl”. The term “halo”, “halide”, or, alternatively, “halogen” means fluoro, chloro, bromo or iodo. The terms “haloalkyl,” “haloalkenyl,” “haloalkynyl” and “haloalkoxy” include alkyl, alkenyl, alkynyl and alkoxy structures that are substituted with one or more halo groups or with combinations thereof. For example, the terms “fluoroalkyl” and “fluoroalkoxy” include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine. The term “protecting group” or “PG” refers to a substituent that is employed to block or protect a particular functionality. Other functional groups on the compound may remain reactive. For example, an “amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include, but are not limited to, acetyl, trifluoroacetyl, tert-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethylenoxycarbonyl (Fmoc). Similarly, a “hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable hydroxy-protecting groups include, but are not limited to, acetyl and silyl. A “carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Suitable carboxy-protecting groups include, but are not limited to, —CH2CH2SO2Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, -2-(p-toluenesulfonyl)ethyl, 2-(p-nitrophenylsulfenyl)ethyl, 2-(diphenylphosphino)-ethyl, and nitroethyl. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991. Certain of the compounds described herein contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that can be defined, in terms of absolute stereochemistry, as (R)- or (S)-. The present chemical entities, pharmaceutical compositions and methods are meant to include all such possible isomers, including racemic mixtures, optically pure forms and intermediate mixtures. For instance, non-limiting examples of intermediate mixtures include a mixture of isomers in a ratio of 10:90, 13:87, 17:83, 20:80, or 22:78. Optically active (R)- and (S)-isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. The term “tautomers” refers to compounds, which are characterized by relatively easy interconversion of isomeric forms in equilibrium. These isomers are intended to be covered by this invention. “Tautomers” are structurally distinct isomers that interconvert by tautomerization. “Tautomerization” is a form of isomerization and includes prototropic or proton-shift tautomerization, which is considered a subset of acid-base chemistry. “Prototropic tautomerization” or “proton-shift tautomerization” involves the migration of a proton accompanied by changes in bond order, often the interchange of a single bond with an adjacent double bond. Where tautomerization is possible (e.g. in solution), a chemical equilibrium of tautomers can be reached. An example of tautomerization is keto-enol tautomerization. A specific example of keto-enol tautomerization is the interconversion of pentane-2,4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerization is phenol-keto tautomerization. A specific example of phenol-keto tautomerization is the interconversion of pyridin-4-ol and pyridin-4(1H)-one tautomers. A “leaving group or atom” is any group or atom that will, under the reaction conditions, cleave from the starting material, thus promoting reaction at a specified site. Suitable examples of such groups unless otherwise specified are halogen atoms and mesyloxy, p-nitrobenzensulphonyloxy and tosyloxy groups. The term “prodrug” refers to a compound, which is an inactive precursor of a compound, converted into its active form in the body by normal metabolic processes. Prodrug design is discussed generally in Hardma, et al. (Eds.), Goodman and Gilman's The Pharmacological Basis of Therapeutics, 9th ed., pp. 11-16 (1996). A thorough discussion is provided in Higuchi, et al., Prodrugs as Novel Delivery Systems, Vol. 14, ASCD Symposium Series, and in Roche (ed.), Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press (1987). To illustrate, prodrugs can be converted into a pharmacologically active form through hydrolysis of, for example, an ester or amide linkage, thereby introducing or exposing a functional group on the resultant product. The prodrugs can be designed to react with an endogenous compound to form a water-soluble conjugate that further enhances the pharmacological properties of the compound, for example, increased circulatory half-life. Alternatively, prodrugs can be designed to undergo covalent modification on a functional group with, for example, glucuronic acid, sulfate, glutathione, amino acids, or acetate. The resulting conjugate can be inactivated and excreted in the urine, or rendered more potent than the parent compound. High molecular weight conjugates also can be excreted into the bile, subjected to enzymatic cleavage, and released back into the circulation, thereby effectively increasing the biological half-life of the originally administered compound. The term “ester” refers to a compound, which is formed by reaction between an acid and an alcohol with elimination of water. An ester can be represented by the general formula RCOOR′. These prodrugs and esters are intended to be covered within the scope of this invention. Additionally the instant invention also includes the compounds which differ only in the presence of one or more isotopically enriched atoms for example replacement of hydrogen with deuterium or tritium, or the replacement of a carbon by13C- or14C-enriched carbon. The compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (125I) or carbon-14 (14C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention. Pharmaceutically acceptable salts forming part of this invention include salts derived from inorganic bases such as Li, Na, K, Ca, Mg, Fe, Cu, Zn, and Mn; salts of organic bases such as N,N′-diacetylethylenediamine, glucamine, triethylamine, choline, hydroxide, dicyclohexylamine, metformin, benzylamine, trialkylamine, and thiamine; chiral bases such as alkylphenylamine, glycinol, and phenyl glycinol; salts of natural amino acids such as glycine, alanine, valine, leucine, isoleucine, norleucine, tyrosine, cystine, cysteine, methionine, proline, hydroxy proline, histidine, ornithine, lysine, arginine, and serine; quaternary ammonium salts of the compounds of invention with alkyl halides, alkyl sulphates such as MeI and (Me)2SO4; non-natural amino acids such as D-isomers or substituted amino acids; guanidine; and substituted guanidine wherein the substituents are selected from nitro, amino, alkyl, alkenyl, alkynyl, ammonium or substituted ammonium salts and aluminum salts. Salts may include acid addition salts where appropriate which are sulphates, nitrates, phosphates, perchlorates, borates, hydrohalides, acetates, tartrates, maleates, citrates, fumarates, succinates, palmoates, methanesulphonates, benzoates, salicylates, benzenesulfonates, ascorbates, glycerophosphates, and ketoglutarates. When ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included. The term “about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary from, for example, between 1% and 15% of the stated number or numerical range. The term “comprising” (and related terms such as “comprise” or “comprises” or “having” or “including”) includes those embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, that “consist of” or “consist essentially of” the described features. The following abbreviations and terms have the indicated meanings throughout: PI3-K=Phosphoinositide 3-kinase; PI=phosphatidylinositol; PDK=Phosphoinositide Dependent Kinase; DNA-PK=Deoxyribose Nucleic Acid Dependent Protein Kinase; PTEN=Phosphatase and Tensin homolog deleted on chromosome Ten; PIKK=Phosphoinositide Kinase Like Kinase; AIDS=Acquired Immuno Deficiency Syndrome; HIV=Human Immunodeficiency Virus; MeI=Methyl Iodide; POCl3=Phosphorous Oxychloride; KCNS=Potassium IsoThiocyanate; TLC=Thin Layer Chromatography; MeOH=Methanol; and CHCl3=Chloroform. Abbreviations used herein have their conventional meaning within the chemical and biological arts. The term “cell proliferation” refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal. The term “co-administration,” “administered in combination with,” and their grammatical equivalents, as used herein, encompasses administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time. Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present. The term “effective amount” or “therapeutically effective amount” refers to that amount of a compound described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below. The therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells, e.g. reduction of platelet adhesion and/or cell migration. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried. As used herein, “treatment,” “treating,” or “ameliorating” are used interchangeably. These terms refers to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. For prophylactic benefit, the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made. A “therapeutic effect,” as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof. The term “subject” or “patient” refers to an animal, such as a mammal, for example a human. The methods described herein can be useful in both human therapeutics and veterinary applications. In some embodiments, the patient is a mammal, and in some embodiments, the patient is human. “Radiation therapy” means exposing a patient, using routine methods and compositions known to the practitioner, to radiation emitters such as alpha-particle emitting radionuclides (e.g., actinium and thorium radionuclides), low linear energy transfer (LET) radiation emitters (i.e. beta emitters), conversion electron emitters (e.g. strontium-89 and samarium-153-EDTMP, or high-energy radiation, including without limitation x-rays, gamma rays, and neutrons. “Signal transduction” is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response. A modulator of a signal transduction pathway refers to a compound which modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway. A modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule. The term “selective inhibition” or “selectively inhibit” as applied to a biologically active agent refers to the agent's ability to selectively reduce the target signaling activity as compared to off-target signaling activity, via direct or indirect interaction with the target. The term “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes, but is not limited to, any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, one or more suitable diluents, fillers, salts, disintegrants, binders, lubricants, glidants, wetting agents, controlled release matrices, colorants/flavoring, carriers, excipients, buffers, stabilizers, solubilizers, and combinations thereof. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions of the invention is contemplated. Supplementary active ingredients can also be incorporated into the compositions. In some embodiments, one or more subject compounds bind specifically to a PI3 kinase or a protein kinase selected from the group consisting of mTor, DNA-dependent protein kinase (Pubmed protein accession number (PPAN) AAA79184), AbI tyrosine kinase (CAA52387), Bcr-Abl, hemopoietic cell kinase (PPAN CAI19695), Src (PPAN CAA24495), vascular endothelial growth factor receptor 2 (PPAN ABB82619), epidermal growth factor receptor (PPAN AG43241), EPH receptor B4 (PPAN EAL23820), stem cell factor receptor (PPAN AAF22141), Tyrosine-protein kinase receptor TIE-2 (PPAN Q02858), fms-related tyrosine kinase 3 (PPAN NP_004110), platelet-derived growth factor receptor alpha (PPAN NP_990080), RET (PPAN CAA73131), and any other related protein kinases, as well as any functional mutants thereof. In some embodiments, the IC50 of a subject compound for pi 10α, pi 10β, pi 10γ, or pi 10δ is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5 nM. In some embodiments, the IC50 of a subject compound for mTor is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5 nM. In some other embodiments, one or more subject compounds exhibit dual binding specificity and are capable of inhibiting a PI3 kinase (e.g., a class I PI3 kinase) as well as a protein kinase (e.g., mTor) with an IC50 value less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5 nM. In some embodiments, the compounds of the present invention exhibit one or more functional characteristics disclosed herein. For example, one or more subject compounds bind specifically to a PI3 kinase. In some embodiments, the IC50 of a subject compound for pi 10α, pi 10β, pi 10γ, or pi 10δ is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than about 1 nM, less than about 0.5 nM, less than about 100 pM, or less than about 50 pM. In some embodiments, one or more of the subject compounds may selectively inhibit one or more members of type I or class I phosphatidylinositol 3-kinases (PI3-kinase) with an IC50 value of about 100 nM, 50 nM, 10 nM, 5 nM, 100 pM, 10 pM or 1 pM, or less as measured in an in vitro kinase assay. In some embodiments, one or more of the subject compound may selectively inhibit one or two members of type I or class I phosphatidylinositol 3-kinases (PI3-kinase) consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In some aspects, some of the subject compounds selectively inhibit PI3-kinase δ as compared to all other type I PI3-kinases. In other aspects, some of the subject compounds selectively inhibit PI3-kinase δ and PI3-kinase γ as compared to the rest of the type I PI3-kinases. In yet other aspects, some of the subject compounds selectively inhibit PI3-kinase α and PI3-kinase β as compared to the rest of the type I PI3-kinases. In still yet some other aspects, some of the subject compounds selectively inhibit PI3-kinase δ and PI3-kinase α as compared to the rest of the type I PI3-kinases. In still yet some other aspects, some of the subject compounds selectively inhibit PI3-kinase δ and PI3-kinase β as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase δ and PI3-kinase α as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase α and PI3-kinase γ as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase γ and PI3-kinase β as compared to the rest of the type I PI3-kinases. In yet another aspect, an inhibitor that selectively inhibits one or more members of type I PI3-kinases, or an inhibitor that selectively inhibits one or more type I PI3-kinase mediated signaling pathways, alternatively can be understood to refer to a compound that exhibits a 50% inhibitory concentration (IC50) with respect to a given type I PI3-kinase, that is at least at least 10-fold, at least 20-fold, at least 50-fold, at least 100-fold, at least 1000-fold, or lower, than the inhibitor's IC50 with respect to the rest of the other type I PI3-kinases. As used herein, the term “PI3-kinase δ selective inhibitor” generally refers to a compound that inhibits the activity of the PI3-kinase δ isozyme more effectively than other isozymes of the PI3K family. A PI3-kinase δ selective inhibitor compound is therefore more selective for PI3-kinase δ than conventional PI3K inhibitors such as wortmannin and LY294002, which are “nonselective PI3K inhibitors.” Inhibition of PI3-kinase δ may be of therapeutic benefit in treatment of various conditions, e.g., conditions characterized by an inflammatory response including but not limited to autoimmune diseases, allergic diseases, and arthritic diseases. Importantly, inhibition of PI3-kinase δ function does not appear to affect biological functions such as viability and fertility. “Inflammatory response” as used herein is characterized by redness, heat, swelling and pain (i.e., inflammation) and typically involves tissue injury or destruction. An inflammatory response is usually a localized, protective response elicited by injury or destruction of tissues, which serves to destroy, dilute or wall off (sequester) both the injurious agent and the injured tissue. Inflammatory responses are notably associated with the influx of leukocytes and/or leukocyte (e.g., neutrophil) chemotaxis. Inflammatory responses may result from infection with pathogenic organisms and viruses, noninfectious means such as trauma or reperfusion following myocardial infarction or stroke, immune responses to foreign antigens, and autoimmune diseases. Inflammatory responses amenable to treatment with the methods and compounds according to the invention encompass conditions associated with reactions of the specific defense system as well as conditions associated with reactions of the non-specific defense system. The therapeutic methods of the invention include methods for the amelioration of conditions associated with inflammatory cell activation. “Inflammatory cell activation” refers to the induction by a stimulus (including but not limited to, cytokines, antigens or auto-antibodies) of a proliferative cellular response, the production of soluble mediators (including but not limited to cytokines, oxygen radicals, enzymes, prostanoids, or vasoactive amines), or cell surface expression of new or increased numbers of mediators (including but not limited to, major histocompatibility antigens or cell adhesion molecules) in inflammatory cells (including but not limited to monocytes, macrophages, T lymphocytes, B lymphocytes, granulocytes (polymorphonuclear leukocytes including neutrophils, basophils, and eosinophils) mast cells, dendritic cells, Langerhans cells, and endothelial cells). It will be appreciated by persons skilled in the art that the activation of one or a combination of these phenotypes in these cells can contribute to the initiation, perpetuation, or exacerbation of an inflammatory condition. “Autoimmune disease” as used herein refers to any group of disorders in which tissue injury is associated with humoral or cell-mediated responses to the body's own constituents. “Transplant rejection” as used herein refers-to any immune response directed against grafted tissue (including organs or cells (e.g., bone marrow), characterized by a loss of function of the grafted and surrounding tissues, pain, swelling, leukocytosis, and thrombocytopenia). “Allergic disease” as used herein refers to any symptoms, tissue damage, or loss of tissue function resulting from allergy. “Arthritic disease” as used herein refers to any disease that is characterized by inflammatory lesions of the joints attributable to a variety of etiologies. “Dermatitis” as used herein refers to any of a large family of diseases of the skin that are characterized by inflammation of the skin attributable to a variety of etiologies. As previously described, the term “PI3-kinase δ selective inhibitor” generally refers to a compound that inhibits the activity of the PI3-kinase δ isozyme more effectively than other isozymes of the PI3K family. The relative efficacies of compounds as inhibitors of an enzyme activity (or other biological activity) can be established by determining the concentrations at which each compound inhibits the activity to a predefined extent and then comparing the results. Typically, the preferred determination is the concentration that inhibits 50% of the activity in a biochemical assay, i.e., the 50% inhibitory concentration or “IC50”. IC50 determinations can be accomplished using conventional techniques known in the art. In general, an IC50 can be determined by measuring the activity of a given enzyme in the presence of a range of concentrations of the inhibitor under study. The experimentally obtained values of enzyme activity then are plotted against the inhibitor concentrations used. The concentration of the inhibitor that shows 50% enzyme activity (as compared to the activity in the absence of any inhibitor) is taken as the IC50 value. Analogously, other inhibitory concentrations can be defined through appropriate determinations of activity. For example, in some settings it can be desirable to establish a 90% inhibitory concentration, i.e., IC90, etc. Accordingly, a PI3-kinase δ selective inhibitor alternatively can be understood to refer to a compound that exhibits a 50% inhibitory concentration (IC50) with respect to PI3-kinase δ, that is at least 10-fold, in another aspect at least 20-fold, and in another aspect at least 30-fold, lower than the IC50 value with respect to any or all of the other class I PI3K family members. In an alternative embodiment of the invention, the term PI3-kinase δ selective inhibitor can be understood to refer to a compound that exhibits an IC50 with respect to PI3-kinase δ that is at least 50-fold, in another aspect at least 100-fold, in an additional aspect at least 200-fold, and in yet another aspect at least 500-fold, lower than the IC50 with respect to any or all of the other PI3K class I family members. A PI3-kinase δ selective inhibitor is typically administered in an amount such that it selectively inhibits PI3-kinase δ activity, as described above. The methods of the invention may be applied to cell populations in vivo or ex vivo. “In vivo” means within a living individual, as within an animal or human or in a subject's body. In this context, the methods of the invention may be used therapeutically or prophylactically in an individual. “Ex vivo” or “In vitro” means outside of a living individual. Examples of ex vivo cell populations include in vitro cell cultures and biological samples including but not limited to fluid or tissue samples obtained from individuals. Such samples may be obtained by methods known in the art. Exemplary biological fluid samples include blood, cerebrospinal fluid, urine, and saliva. Exemplary tissue samples include tumors and biopsies thereof. In this context, the invention may be used for a variety of purposes, including therapeutic and experimental purposes. For example, the invention may be used ex vivo or in vitro to determine the optimal schedule and/or dosing of administration of a PI3-kinase δ selective inhibitor for a given indication, cell type, individual, and other parameters. Information gleaned from such use may be used for experimental or diagnostic purposes or in the clinic to set protocols for in vivo treatment. Other ex vivo uses for which the invention may be suited are described below or will become apparent to those skilled in the art. Pharmaceutical Compositions The invention provides a pharmaceutical composition comprising one or more compounds of the present invention. The pharmaceutical composition may include one or more additional active ingredients as described herein. The pharmaceutical composition may be administered for any of the disorders described herein In some embodiments, the invention provides pharmaceutical compositions for treating diseases or conditions related to an undesirable, over-active, harmful or deleterious immune response in a mammal. Such undesirable immune response can be associated with or result in, e.g., asthma, emphysema, bronchitis, psoriasis, allergy, anaphylaxsis, auto-immune diseases, rhuematoid arthritis, graft versus host disease, and lupus erythematosus. The pharmaceutical compositions of the present invention can be used to treat other respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing. In some embodiments, the invention provides pharmaceutical compositions for the treatment of disorders such as hyperproliferative disorder including but not limited to cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS related (e.g. Lymphoma and Kaposi's Sarcoma) or Viral-Induced cancer. In some embodiments, the pharmaceutical composition is for the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)). The invention also relates to a composition for treating a disease related to vasculogenesis or angiogenesis in a mammal which can manifest as tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer. The invention also provides compositions for the treatment of liver diseases (including diabetes), pancreatitis or kidney disease (including proliferative glomerulonephritis and diabetes-induced renal disease) or pain in a mammal. The invention further provides a composition for the prevention of blastocyte implantation in a mammal. The subject pharmaceutical compositions are typically formulated to provide a therapeutically effective amount of a compound of the present invention as the active ingredient, or a pharmaceutically acceptable salt, ester, or prodrug thereof. Where desired, the pharmaceutical compositions contain a compound of the present invention as the active ingredient or a pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, such as inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants. The subject pharmaceutical compositions can be administered alone or in combination with one or more other agents, which are also typically administered in the form of pharmaceutical compositions. Where desired, the subject compounds and other agent(s) may be mixed into a preparation or both components may be formulated into separate preparations to use them in combination separately or at the same time. Methods include administration of an inhibitor by itself, or in combination as described herein, and in each case optionally including one or more suitable diluents, fillers, salts, disintegrants, binders, lubricants, glidants, wetting agents, controlled release matrices, colorants/flavoring, carriers, excipients, buffers, stabilizers, solubilizers, and combinations thereof. Preparations of various pharmaceutical compositions are known in the art. See, e.g., Anderson, Philip O.; Knoben, James E.; Troutman, William G, eds., Handbook of Clinical Drug Data, Tenth Edition, McGraw-Hill, 2002; Pratt and Taylor, eds., Principles of Drug Action, Third Edition, Churchill Livingston, New York, 1990; Katzung, ed., Basic and Clinical Pharmacology, Ninth Edition, McGraw Hill, 2003; Goodman and Gilman, eds., The Pharmacological Basis of Therapeutics, Tenth Edition, McGraw Hill, 2001; Remingtons Pharmaceutical Sciences, 20th Ed., Lippincott Williams & Wilkins., 2000; Martindale, The Extra Pharmacopoeia, Thirty-Second Edition (The Pharmaceutical Press, London, 1999), all of which are incorporated by reference herein in their entirety. The compounds or pharmaceutical composition of the present invention can be administered by any route that enables delivery of the compounds to the site of action, such asoral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical administration (e.g. transdermal application), rectal administration, via local delivery by catheter or stent or through inhalation. The compounds can also be administered intraadiposally or intrathecally. The compositions can be administered in solid, semi-solid, liquid or gaseous form, or may be in dried powder, such as lyophilized form. The pharmaceutical compositions can be packaged in forms convenient for delivery, including, for example, solid dosage forms such as capsules, sachets, cachets, gelatins, papers, tablets, capsules, suppositories, pellets, pills, troches, and lozenges. The type of packaging will generally depend on the desired route of administration. Implantable sustained release formulations are also contemplated, as are transdermal formulations. Routes of Administration In the methods according to the invention, the inhibitor compounds may be administered by various routes. For example, pharmaceutical compositions may be for injection, or for oral, nasal, transdermal or other forms of administration, including, e.g., by intravenous, intradermal, intramuscular, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., aerosolized drugs) or subcutaneous injection (including depot administration for long term release e.g., embedded-under the-splenic capsule, brain, or in the cornea); by sublingual, anal, or vaginal administration, or by surgical implantation, e.g., embedded under the splenic capsule, brain, or in the cornea. The treatment may consist of a single dose or a plurality of doses over a period of time. In general, the methods of the invention involve administering effective amounts of a modulator of the invention together with one or more pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers, as described above. The subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages. The pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, and adjuvants. In one aspect, the invention provides methods for oral administration of a pharmaceutical composition of the invention. Oral solid dosage forms are described generally in Remington's Pharmaceutical Sciences, supra at Chapter 89. Solid dosage forms include tablets, capsules, pills, troches or lozenges, and cachets or pellets. Also, liposomal or proteinoid encapsulation may be used to formulate the compositions (as, for example, proteinoid microspheres reported in U.S. Pat. No. 4,925,673). Liposomal encapsulation may include liposomes that are derivatized with various polymers (e.g., U.S. Pat. No. 5,013,556). The formulation may include a compound of the invention and inert ingredients which protect against degradation in the stomach and which permit release of the biologically active material in the intestine. Toxicity and therapeutic efficacy of the PI3-kinase δ selective compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). Additionally, this information can be determined in cell cultures or experimental animals additionally treated with other therapies including but not limited to radiation, chemotherapeutic agents, photodynamic therapies, radiofrequency ablation, anti-angiogenic agents, and combinations thereof. The amount of the compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. However, an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. by dividing such larger doses into several small doses for administration throughout the day. In some embodiments, a compound of the invention is administered in a single dose. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes may be used as appropriate. A single dose of a compound of the invention may also be used for treatment of an acute condition. In practice of the methods of the invention, the pharmaceutical compositions are generally provided in doses ranging from 1 pg compound/kg body weight to 1000 mg/kg, 0.1 mg/kg to 100 mg/kg, 0.1 mg/kg to 50 mg/kg, and 1 to 20 mg/kg, given in daily doses or in equivalent doses at longer or shorter intervals, e.g., every other day, twice weekly, weekly, or twice or three times daily. The inhibitor compositions may be administered by an initial bolus followed by a continuous infusion to maintain therapeutic circulating levels of drug product. Those of ordinary skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual to be treated. The frequency of dosing will depend on the pharmacokinetic parameters of the agents and the route of administration. The optimal pharmaceutical formulation will be determined by one skilled in the art depending upon the route of administration and desired dosage [see, for example, Remington's Pharmaceutical Sciences, pp. 1435-1712, the disclosure of which is hereby incorporated by reference]. Such formulations may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered agents. Depending on the route of administration, a suitable dose may be calculated according to body weight, body surface area or organ size. Further refinement of the calculations necessary to determine the appropriate dosage for treatment involving each of the above mentioned formulations is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein, as well as the pharmacokinetic data observed in human clinical trials. Appropriate dosages may be ascertained by using established assays for determining blood level dosages in conjunction with an appropriate physician considering various factors which modify the action of drugs, e.g., the drug's specific activity, the severity of the indication, and the responsiveness of the individual, the age, condition, body weight, sex and diet of the individual, the time of administration and other clinical factors. As studies are conducted, further information will emerge regarding the appropriate dosage levels and duration of treatment for various diseases and conditions capable of being treated with the methods of the invention. In some embodiments, a compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary. Administration of the agents of the invention may continue as long as necessary. In some embodiments, an agent of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, an agent of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, an agent of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects. An effective amount of a compound of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intraarterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant. The compounds of the invention may be administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation in light of the instant disclosure. When a compound of the invention, is administered in a composition that comprises one or more agents, and the agent has a shorter half-life than the compound of the invention unit dose forms of the agent and the compound of the invention may be adjusted accordingly. The inhibitors of the invention may be covalently or noncovalently associated with a carrier molecule including but not limited to a linear polymer (e.g., polyethylene glycol, polylysine, dextran, etc.), a branched-chain polymer (see U.S. Pat. Nos. 4,289,872 and 5,229,490; PCT Publication No. WO 93/21259), a lipid, a cholesterol group (such as a steroid), or a carbohydrate or oligosaccharide. Specific examples of carriers for use in the pharmaceutical compositions of the invention include carbohydrate-based polymers such as trehalose, mannitol, xylitol, sucrose, lactose, sorbitol, dextrans such as cyclodextran, cellulose, and cellulose derivatives. Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated. Other carriers include one or more water soluble polymer attachments such as polyoxyethylene glycol, or polypropylene glycol as described U.S. Pat. Nos. 4,640,835, 4,496,689, 4,301,144, 4,670,417, 4,791,192 and 4,179,337. Still other useful carrier polymers known in the art include monomethoxy-polyethylene glycol, poly-(N-vinyl pyrrolidone)-polyethylene glycol, propylene glycol homopolymers, a polypropylene oxidelethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol, as well as mixtures of these polymers. Derivitization with bifunctional agents is useful for cross-linking a compound of the invention to a support matrix or to a carrier. One such carrier is polyethylene glycol (PEG). The PEG group may be of any convenient molecular weight and may be straight chain or branched. The average molecular weight of the PEG can range from about 2 kDa to about 100 kDa, in another aspect from about 5 kDa to about 50 kDa, and in a further aspect from about 5 kDa to about 10 kDa. The PEG groups will generally be attached to the compounds of the invention via acylation, reductive alkylation, Michael addition, thiol alkylation or other chemoselective conjugation/ligation methods through a reactive group on the PEG moiety (e.g., an aldehyde, amino, ester, thiol, ci-haloacetyl, maleimido or hydrazino group) to a reactive group on the target inhibitor compound (e.g., an aldehyde, amino, ester, thiol, a-haloacetyl, maleimido or hydrazino group). Cross-linking agents can include, e.g., esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3′-dithiobis (succinimidylpropionate), and bifunctional maleimides such as bis-N-maleimido-1,8-octane. Derivatizing agents such as methyl-3-[(p-azidophenyl)dithiolpropioimidate yield photoactivatable intermediates that are capable of forming crosslinks in the presence of light. Alternatively, reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287; 3,691,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440 may be employed for inhibitor immobilization. Method of Treatment The invention also provides methods of using the compounds or pharmaceutical compositions of the present invention to treat disease conditions, including but not limited to diseases associated with malfunctioning of one or more types of PI3 kinase. A detailed description of conditions and disorders mediated by pi 10δ kinase activity is set forth in WO 2001/81346 and US 2005/043239, both of which are incorporated herein by reference in their entireties for all purposes. The treatment methods provided herein comprise administering to the subject a therapeutically effective amount of a compound of the invention. In one embodiment, the present invention provides a method of treating an inflammation disorder, including autoimmune diseases in a mammal. The method comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. The disorders, diseases, or conditions treatable with a compound provided herein, include, but are not limited to,inflammatory or allergic diseases, including systemic anaphylaxis and hypersensitivity disorders, atopic dermatitis, urticaria, drug allergies, insect sting allergies, food allergies (including celiac disease and the like), anaphylaxis, serum sickness, drug reactions, insect venom allergies, hypersensitivity pneumonitis, angioedema, erythema multiforme, Stevens-Johnson syndrome, atopic keratoconjunctivitis, venereal keratoconjunctivitis, giant papillary conjunctivitis, and mastocytosis;inflammatory bowel diseases, including Crohn's disease, ulcerative colitis, ileitis, enteritis, and necrotizing enterocolitis;vasculitis, and Behcet's syndrome;psoriasis and inflammatory dermatoses, including dermatitis, eczema allergic contact dermatitis, viral cutaneous pathologies including those derived from human papillomavirus, HIV or RLV infection, bacterial, flugal, and other parasital cutaneous pathologies, and cutaneous lupus erythematosus;asthma and respiratory allergic diseases, including allergic asthma, exercise induced asthma, allergic rhinitis, otitis media, hypersensitivity lung diseases, chronic obstructive pulmonary disease and other respiratory problems;autoimmune diseases and inflammatory conditions, including but are not limited to acute disseminated encephalomyelitis (ADEM), Addison's disease, antiphospholipid antibody syndrome (APS), aplastic anemia, autoimmune hepatitis, coeliac disease, Crohn's disease, Diabetes mellitus (type 1), Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome (GBS), Reynaud's syndrome, Hashimoto's disease, lupus erythematosus, systemic lupus erythematosus (SLE), multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome (OMS), optic neuritis, Ord's thyroiditis, oemphigus, polyarthritis, primary biliary cirrhosis, psoriasis, rheumatoid arthritis, psoriatic arthritis, gouty arthritis, spondylitis, reactive arthritis, chronic or acute glomerulonephritis, lupus nephritis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis (also known as “giant cell arteritis”), warm autoimmune hemolytic anemia, Wegener's granulomatosis, alopecia universalis, Chagas' disease, chronic fatigue syndrome, dysautonomia, endometriosis, hidradenitis suppurativa, interstitial cystitis, neuromyotonia, sarcoidosis, scleroderma, ulcerative colitis, connective tissue disease, autoimmune pulmonary inflammation, autoimmune thyroiditis, autoimmune inflammatory eye disease, vitiligo, and vulvodynia. Other disorders include bone-resorption disorders and thromobsis;tissue or organ transplant rejection disorders including but not limited to graft rejection (including allograft rejection and graft-v-host disease (GVHD)), e.g., skin graft rejection, solid organ transplant rejection, bone marrow transplant rejection;fever;cardiovascular disorders, including acute heart failure, hypotension, hypertension, angina pectoris, myocardial infarction, cardiomyopathy, congestive heart failure, atherosclerosis, coronary artery disease, restenosis, and vascular stenosis;cerebrovascular disorders, including traumatic brain injury, stroke, ischemic reperfusion injury and aneurysm;cancers of the breast, skin, prostate, cervix, uterus, ovary, testes, bladder, lung, liver, larynx, oral cavity, colon and gastrointestinal tract (e.g., esophagus, stomach, pancreas), brain, thyroid, blood, and lymphatic system;fibrosis, connective tissue disease, and sarcoidosis;genital and reproductive conditions, including erectile dysfunction;gastrointestinal disorders, including gastritis, ulcers, nausea, pancreatitis, and vomiting;neurologic disorders, including Alzheimer's disease;sleep disorders, including insomnia, narcolepsy, sleep apnea syndrome, and Pickwick Syndrome;pain, myalgias due to infection;renal disorders;ocular disorders, including glaucoma;infectious diseases, including HIV;sepsis; septic shock; endotoxic shock; gram negative sepsis; gram positive sepsis; toxic shock syndrome; multiple organ injury syndrome secondary to septicemia, trauma, or hemorrhage;pulmonary or respiratory conditions including but not limited to asthma, chronic bronchitis, allergic rhinitis, adult respiratory distress syndrome (ARDS), severe acute respiratory syndrome (SARS), chronic pulmonary inflammatory diseases (e.g., chronic obstructive pulmonary disease), silicosis, pulmonary sarcoidosis, pleurisy, alveolitis, vasculitis, pneumonia, bronchiectasis, hereditary emphysema, and pulmonary oxygen toxicity;ischemic-reperfusion injury, e.g., of the myocardium, brain, or extremities;fibrosis including but not limited to cystic fibrosis; keloid formation or scar tissue formation;central or peripheral nervous system inflammatory conditions including but not limited to meningitis (e.g., acute purulent meningitis), encephalitis, and brain or spinal cord injury due to minor trauma;Sjorgren's syndrome; diseases involving leukocyte diapedesis; alcoholic hepatitis; bacterial pneumonia; community acquired pneumonia (CAP); Pneumocystis carinii pneumonia (PCP); antigen-antibody complex mediated diseases; hypovolemic shock; acute and delayed hypersensitivity; disease states due to leukocyte dyscrasia and metastasis; thermal injury; granulocyte transfusion associated syndromes; cytokine-induced toxicity; stroke; pancreatitis; myocardial infarction, respiratory syncytial virus (RSV) infection; and spinal cord injury. In certain embodiments, the cancer or cancers treatable with the methods provided herein includes, but is or are not limited to,leukemias, including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblasts, promyelocyte, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome or a symptom thereof (such as anemia, thrombocytopenia, neutropenia, bicytopenia or pancytopenia), refractory anemia (RA), RA with ringed sideroblasts (RARS), RA with excess blasts (RAEB), RAEB in transformation (RAEB-T), preleukemia, and chronic myelomonocytic leukemia (CMML);chronic leukemias, including, but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, and hairy cell leukemia;polycythemia vera;lymphomas, including, but not limited to, Hodgkin's disease and non-Hodgkin's disease;multiple myelomas, including, but not limited to, smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma, and extramedullary plasmacytoma;Waldenstrom's macroglobulinemia;monoclonal gammopathy of undetermined significance;benign monoclonal gammopathy;heavy chain disease;bone and connective tissue sarcomas, including, but not limited to, bone sarcoma, osteosarcoma, chondrosarcoma, Ewing's sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcomas, angiosarcoma (hemangiosarcoma), fibrosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, metastatic cancers, neurilemmoma, rhabdomyosarcoma, and synovial sarcoma;brain tumors, including, but not limited to, glioma, astrocytoma, brain stem glioma, ependymoma, oligodendroglioma, nonglial tumor, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, and primary brain lymphoma;breast cancer, including, but not limited to, adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, primary cancers, Paget's disease, and inflammatory breast cancer;adrenal cancer, including, but not limited to, pheochromocytom and adrenocortical carcinoma;thyroid cancer, including, but not limited to, papillary or follicular thyroid cancer, medullary thyroid cancer, and anaplastic thyroid cancer;pancreatic cancer, including, but not limited to, insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor;pituitary cancer, including, but limited to, Cushing's disease, prolactin-secreting tumor, acromegaly, and diabetes insipidus;eye cancer, including, but not limited, to ocular melanoma such as iris melanoma, choroidal melanoma, and cilliary body melanoma, and retinoblastoma;vaginal cancer, including, but not limited to, squamous cell carcinoma, adenocarcinoma, and melanoma;vulvar cancer, including, but not limited to, squamous cell carcinoma, melanoma, adenocarcinoma, basal cell carcinoma, sarcoma, and Paget's disease;cervical cancers, including, but not limited to, squamous cell carcinoma, and adenocarcinoma;uterine cancer, including, but not limited to, endometrial carcinoma and uterine sarcoma;ovarian cancer, including, but not limited to, ovarian epithelial carcinoma, borderline tumor, germ cell tumor, and stromal tumor;esophageal cancer, including, but not limited to, squamous cancer, adenocarcinoma, adenoid cystic carcinoma, mucoepidermoid carcinoma, adenosquamous carcinoma, sarcoma, melanoma, plasmacytoma, verrucous carcinoma, and oat cell (small cell) carcinoma;stomach cancer, including, but not limited to, adenocarcinoma, fungating (polypoid), ulcerating, superficial spreading, diffusely spreading, malignant lymphoma, liposarcoma, fibrosarcoma, and carcinosarcoma;colon cancer;rectal cancer;liver cancer, including, but not limited to, hepatocellular carcinoma and hepatoblastoma;gallbladder cancer, including, but not limited to, adenocarcinoma;cholangiocarcinomas, including, but not limited to, pappillary, nodular, and diffuse;lung cancer, including, but not limited to, non-small cell lung cancer, squamous cell carcinoma (epidermoid carcinoma), adenocarcinoma, large-cell carcinoma, and small-cell lung cancer;testicular cancer, including, but not limited to, germinal tumor, seminoma, anaplastic, classic (typical), spermatocytic, nonseminoma, embryonal carcinoma, teratoma carcinoma, and choriocarcinoma (yolk-sac tumor);prostate cancer, including, but not limited to, adenocarcinoma, leiomyosarcoma, and rhabdomyosarcoma;penal cancer;oral cancer, including, but not limited to, squamous cell carcinoma;basal cancer;salivary gland cancer, including, but not limited to, adenocarcinoma, mucoepidermoid carcinoma, and adenoidcystic carcinoma;pharynx cancer, including, but not limited to, squamous cell cancer and verrucous;skin cancer, including, but not limited to, basal cell carcinoma, squamous cell carcinoma and melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, and acral lentiginous melanoma;kidney cancer, including, but not limited to, renal cell cancer, adenocarcinoma,hypernephroma, fibrosarcoma, and transitional cell cancer (renal pelvis and/or uterer);Wilms' tumor;bladder cancer, including, but not limited to, transitional cell carcinoma, squamous cell cancer, adenocarcinoma, and carcinosarcoma; and other cancer, including, not limited to, myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangio-endotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, and papillary adenocarcinomas See Fishman et al., 1985, Medicine, 2d Ed., J.B. Lippincott Co., Philadelphia and Murphy et al., 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America. It will be appreciated that the treatment methods of the invention are useful in the fields of human medicine and veterinary medicine. Thus, the individual to be treated may be a mammal, preferably human, or other animals. For veterinary purposes, individuals include but are not limited to farm animals including cows, sheep, pigs, horses, and goats; companion animals such as dogs and cats; exotic and/or zoo animals; laboratory animals including mice, rats, rabbits, guinea pigs, and hamsters; and poultry such as chickens, turkeys, ducks, and geese. In some embodiments, the method of treating inflammatory or autoimmune diseases comprises administering to a subject (e.g. a mammal) a therapeutically effective amount of one or more compounds of the present invention that selectively inhibit PI3K-δ and/or PI3K-γ as compared to all other type I PI3 kinases. Such selective inhibition of PI3K-δ and/or PI3K-γ may be advantageous for treating any of the diseases or conditions described herein. For example, selective inhibition of PI3K-δ may inhibit inflammatory responses associated with inflammatory diseases, autoimmune disease, or diseases related to an undesirable immune response including but not limited to asthma, emphysema, allergy, dermatitis, rhuematoid arthritis, psoriasis, lupus erythematosus, or graft versus host disease. Selective inhibition of POK-δ may further provide for a reduction in the inflammatory or undesirable immune response without a concomittant reduction in the ability to reduce a bacterial, viral, and/or fungal infection. Selective inhibition of both PI3 K-δ and PI3K-γ may be advantageous for inhibiting the inflammatory response in the subject to a greater degree than that would be provided for by inhibitors that selectively inhibit PI3K-δ or PI3K-γ alone. In one aspect, one or more of the subject methods are effective in reducing antigen specific antibody production in vivo by about 2-fold, 3-fold, 4-fold, 5-fold, 7.5-fold, 10-fold, 25-fold, 50-fold, 100-fold, 250-fold, 500-fold, 750-fold, or about 1000-fold or more. In another aspect, one or more of the subject methods are effective in reducing antigen specific IgG3 and/or IgGM production in vivo by about 2-fold, 3-fold, 4-fold, 5-fold, 7.5-fold, 10-fold, 25-fold, 50-fold, 100-fold, 250-fold, 500-fold, 750-fold, or about 1000-fold or more. In one aspect, one of more of the subject methods are effective in ameliorating symptoms associated with rhuematoid arthritis including but not limited to a reduction in the swelling of joints, a reduction in serum anti-collagen levels, and/or a reduction in joint pathology such as bone resorption, cartilage damage, pannus, and/or inflammation. In another aspect, the subject methods are effective in reducing ankle inflammation by at least about 2%, 5%, 10%, 15%, 20%, 25%, 30%, 50%, 60%, or about 75% to 90%. In another aspect, the subject methods are effective in reducing knee inflammation by at least about 2%, 5%, 10%, 15%, 20%, 25%, 30%, 50%, 60%, or about 75% to 90% or more. In still another aspect, the subject methods are effective in reducing serum anti-type II collagen levels by at least about 10%, 12%, 15%, 20%, 24%, 25%, 30%, 35%, 50%, 60%, 75%, 80%, 86%, 87%, or about 90% or more. In another aspect, the subject methods are effective in reducing ankle histopathology scores by about 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 90% or more. In still another aspect, the subject methods are effective in reducing knee histopathology scores by about 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 90% or more. In other embodiments, the present invention provides methods of using the compounds or pharmaceutical compositions to treat respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing. For example, methods are provided to treat obstructive pulmonary disease. Chronic obstructive pulmonary disease (COPD) is an umbrella term for a group of respiratory tract diseases that are characterized by airflow obstruction or limitation. Conditions included in this umbrella term are: chronic bronchitis, emphysema, and bronchiectasis. In another embodiment, the compounds described herein are used for the treatment of asthma. Also, the compounds or pharmaceutical compositions described herein may be used for the treatment of endotoxemia and sepsis. In one embodiment, the compounds or pharmaceutical compositions described herein are used to for the treatment of rheumatoid arthritis (RA). In yet another embodiment, the compounds or pharmaceutical compositions described herein is used for the treatment of contact or atopic dermatitis. Contact dermatitis includes irritant dermatitis, phototoxic dermatitis, allergic dermatitis, photoallergic dermatitis, contact urticaria, systemic contact-type dermatitis and the like. Irritant dermatitis can occur when too much of a substance is used on the skin of when the skin is sensitive to certain substance. Atopic dermatitis, sometimes called eczema, is a kind of dermatitis, an atopic skin disease. The invention also relates to a method of treating a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In some embodiments, said method relates to the treatment of cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS-related (e.g. Lymphoma and Kaposi's Sarcoma) or viral-induced cancer. In some embodiments, said method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)). The invention also relates to a method of treating diseases related to vasculogenesis or angiogenesis in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In some embodiments, said method is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer. Patients that can be treated with compounds of the present invention, or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative of said compounds, according to the methods of this invention include, for example, patients that have been diagnosed as having psoriasis; restenosis; atherosclerosis; BPH; breast cancer such as a ductal carcinoma in duct tissue in a mammary gland, medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian cancer, including epithelial ovarian tumors such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated from the ovary into the abdominal cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix epithelial including squamous cell carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer selected from the following: an adenocarcinoma or an adenocarinoma that has migrated to the bone; pancreatic cancer such as epitheliod carcinoma in the pancreatic duct tissue and an adenocarcinoma in a pancreatic duct; bladder cancer such as a transitional cell carcinoma in urinary bladder, urothelial carcinomas (transitional cell carcinomas), tumors in the urothelial cells that line the bladder, squamous cell carcinomas, adenocarcinomas, and small cell cancers; leukemia such as acute myeloid leukemia (AML), acute lymphocytic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, hairy cell leukemia, myelodysplasia, myeloproliferative disorders, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), mastocytosis, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), and myelodysplastic syndrome (MDS); bone cancer; lung cancer such as non-small cell lung cancer (NSCLC), which is divided into squamous cell carcinomas, adenocarcinomas, and large cell undifferentiated carcinomas, and small cell lung cancer; skin cancer such as basal cell carcinoma, melanoma, squamous cell carcinoma and actinic keratosis, which is a skin condition that sometimes develops into squamous cell carcinoma; eye retinoblastoma; cutaneous or intraocular (eye) melanoma; primary liver cancer (cancer that begins in the liver); kidney cancer; thyroid cancer such as papillary, follicular, medullary and anaplastic; AIDS-related lymphoma such as diffuse large B-cell lymphoma, B-cell immunoblastic lymphoma and small non-cleaved cell lymphoma; Kaposi's Sarcoma; viral-induced cancers including hepatitis B virus (HBV), hepatitis C virus (HCV), and hepatocellular carcinoma; human lymphotropic virus-type 1 (HTLV-I) and adult T-cell leukemia/lymphoma; and human papilloma virus (HPV) and cervical cancer; central nervous system cancers (CNS) such as primary brain tumor, which includes gliomas (astrocytoma, anaplastic astrocytoma, or glioblastoma multiforme), Oligodendroglioma, Ependymoma, Meningioma, Lymphoma, Schwannoma, and Medulloblastoma; peripheral nervous system (PNS) cancers such as acoustic neuromas and malignant peripheral nerve sheath tumor (MPNST) including neurofibromas and schwannomas, malignant fibrous cytoma, malignant fibrous histiocytoma, malignant meningioma, malignant mesothelioma, and malignant mixed Müllerian tumor; oral cavity and oropharyngeal cancer such as, hypopharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, and oropharyngeal cancer; stomach cancer such as lymphomas, gastric stromal tumors, and carcinoid tumors; testicular cancer such as germ cell tumors (GCTs), which include seminomas and nonseminomas, and gonadal stromal tumors, which include Leydig cell tumors and Sertoli cell tumors; thymus cancer such as to thymomas, thymic carcinomas, Hodgkin disease, non-Hodgkin lymphomas carcinoids or carcinoid tumors; rectal cancer; and colon cancer. The invention also relates to a method of treating diabetes in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In addition, the compounds described herein may be used to treat acne. In addition, the compounds described herein may be used for the treatment of arteriosclerosis, including atherosclerosis. Arteriosclerosis is a general term describing any hardening of medium or large arteries. Atherosclerosis is a hardening of an artery specifically due to an atheromatous plaque. Further the compounds described herein may be used for the treatment of glomerulonephritis. Glomerulonephritis is a primary or secondary autoimmune renal disease characterized by inflammation of the glomeruli. It may be asymptomatic, or present with hematuria and/or proteinuria. There are many recognized types, divided in acute, subacute or chronic glomerulonephritis. Causes are infectious (bacterial, viral or parasitic pathogens), autoimmune or paraneoplastic. Additionally, the compounds described herein may be used for the treatment of bursitis, lupus, acute disseminated encephalomyelitis (ADEM), addison's disease, antiphospholipid antibody syndrome (APS), aplastic anemia, autoimmune hepatitis, coeliac disease, Crohn's disease, diabetes mellitus (type 1), goodpasture's syndrome, graves' disease, guillain-barre syndrome (GBS), hashimoto's disease, inflammatory bowel disease, lupus erythematosus, myasthenia gravis, opsoclonus myoclonus syndrome (OMS), optic neuritis, ord's thyroiditiSjOstheoarthritis, uveoretinitis, pemphigus, polyarthritis, primary biliary cirrhosis, reiter's syndrome, takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, alopecia universalis, chagasi disease, chronic fatigue syndrome, dysautonomia, endometriosis, hidradenitis suppurativa, interstitial cystitis, neuromyotonia, sarcoidosis, scleroderma, ulcerative colitis, vitiligo, vulvodynia, appendicitis, arteritis, arthritis, blepharitis, bronchiolitis, bronchitis, cervicitis, cholangitis, cholecystitis, chorioamnionitis, colitis, conjunctivitis, cystitis, dacryoadenitis, dermatomyositis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, gingivitis, hepatitis, hidradenitis, ileitis, iritis, laryngitis, mastitis, meningitis, myelitis, myocarditis, myositis, nephritis, omphalitis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vasculitis, or vulvitis. The invention also relates to a method of treating a cardiovascular disease in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. Examples of cardiovascular conditions include, but are not limited to, atherosclerosis, restenosis, vascular occlusion and carotid obstructive disease. In another aspect, the present invention provides methods of disrupting the function of a leukocyte or disrupting a function of an osteoclast. The method includes contacting the leukocyte or the osteoclast with a function disrupting amount of a compound of the invention. In another aspect of the present invention, methods are provided for treating ophthalmic disease by administering one or more of the subject compounds or pharmaceutical compositions to the eye of a subject. The invention further provides methods of modulating kinase activity by contacting a kinase with an amount of a compound of the invention sufficient to modulate the activity of the kinase. Modulate can be inhibiting or activating kinase activity. In some embodiments, the invention provides methods of inhibiting kinase activity by contacting a kinase with an amount of a compound of the invention sufficient to inhibit the activity of the kinase. In some embodiments, the invention provides methods of inhibiting kinase activity in a solution by contacting said solution with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said solution. In some embodiments, the invention provides methods of inhibiting kinase activity in a cell by contacting said cell with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said cell. In some embodiments, the invention provides methods of inhibiting kinase activity in a tissue by contacting said tissue with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said tissue. In some embodiments, the invention provides methods of inhibiting kinase activity in an organism by contacting said organism with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said organism. In some embodiments, the invention provides methods of inhibiting kinase activity in an animal by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said animal. In some embodiments, the invention provides methods of inhibiting kinase activity in a mammal by contacting said mammal with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said mammal. In some embodiments, the invention provides methods of inhibiting kinase activity in a human by contacting said human with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said human. In some embodiments, the % of kinase activity after contacting a kinase with a compound of the invention is less than 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 95, or 99% of the kinase activity in the absence of said contacting step. In some embodiments, the kinase is a lipid kinase or a protein kinase. In some embodiments, the kinase is selected from the group consisting of PI3 kinase including different isorforms such as PI3 kinase α, PI3 kinase β, PI3 kinase γ, PI3 kinase δ; DNA-PK; mTor; AbI, VEGFR, Ephrin receptor B4 (EphB4); TEK receptor tyrosine kinase (HE2); FMS-related tyrosine kinase 3 (FLT-3); Platelet derived growth factor receptor (PDGFR); RET; ATM; ATR; hSmg-1; Hck; Src; Epidermal growth factor receptor (EGFR); KIT; Insulin Receptor (IR) and IGFR. The invention further provides methods of modulating PI3 kinase activity by contacting a PI3 kinase with an amount of a compound of the invention sufficient to modulate the activity of the PI3 kinase. Modulate can be inhibiting or activating PI3 kinase activity. In some embodiments, the invention provides methods of inhibiting PI3 kinase activity by contacting a PI3 kinase with an amount of a compound of the invention sufficient to inhibit the activity of the PI3 kinase. In some embodiments, the invention provides methods of inhibiting PI3 kinase activity. Such inhibition can take place in solution, in a cell expressing one or more PI3 kinases, in a tissue comprising a cell expressing one or more PI3 kinases, or in an organism expressing one or more PI3 kinases. In some embodiments, the invention provides methods of inhibiting PI3 kinase activity in an animal (including mammal such as humans) by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of the PI3 kinase in said animal. The ability of the compounds of the invention to treat arthritis can be demonstrated in a murine collagen-induced arthritis model [Kakimoto, et al., Cell. Immunol., 142:326-337 (1992)], in a rat collagen-induced arthritis model [Knoerzer, et al., Toxicol. Pathol., 25:13-19-(1997)], in a rat adjuvant arthritis model [Halloran, et al., Arthritis Rheum., 39:810-819 (1996)], in a rat streptococcal cell wall-induced arthritis model [Schimmer, et al., J. Immunol., 160:1466-1477 (1998)], or in a SCID-mouse human rheumatoid arthritis model [Oppenheimer-Marks, et al., J. Clin. Invest., 101: 1261-1272(1998)]. The ability of the compounds of the invention to treat Lyme arthritis can be demonstrated according to the method of Gross, et al., Science, 218:703-706, (1998). The ability of the compounds of the invention to treat asthma can be demonstrated in a murine allergic asthma model according to the method of Wegner, et al., Science, 247:456-459 (1990), or in a murine non-allergic asthma model according to the method of Bloemen, et al, Am. J. Respir. Crit. Care Med., 153:521-529 (1996). The ability of the compounds of the invention to treat inflammatory lung injury can be demonstrated in a murine oxygen-induced lung injury model according to the method of Wegner, et al., Lung, 170:267-279 (1992), in a murine immune complex-induced lung injury model according to the method of Mulligan, et al., J. Immunol., 154:1350-1363 (1995), or in a murine acid-induced lung injury model according to the method of Nagase, et al., Am. J. Respir. Crit. Care Med., 154:504-510(1996). The ability of the compounds of the invention to treat inflammatory bowel disease can be demonstrated in a murine chemical-induced colitis model according to the method of Bennett, et al., J. Pharmacol. Exp. Ther., 280:988-1000 (1997). The ability of the compounds of the invention to treat autoimmune diabetes can be demonstrated in an NOD mouse model according to the method of Hasagawa, et al., Int. Immunol., 6:831-838 (1994), or in a murine streptozotocin-induced diabetes model according to the method of Herrold, et al., Cell Immunol., 157:489-500 (1994). The ability of the compounds of the invention to treat inflammatory liver injury can be demonstrated in a murine liver injury model according to the method of Tanaka, et al., J. Immunol., 151:5088-5095 (1993). The ability of the compounds of the invention to treat inflammatory glomerular injury can be demonstrated in a rat nephrotoxic serum nephritis model according to the method of Kawasaki, et al., J. Immunol., 150: 1074-1083 (1993). The ability of the compounds of the invention to treat radiation-induced enteritis can be demonstrated in a rat abdominal irradiation model according to the method of Panes, et al., Gastroenterology, 108:1761-1769 (1995). The ability of the PI3K delta selective inhibitors to treat radiation pneumonitis can be demonstrated in a murine pulmonary irradiation model according to the method of Hallahan, et al., Proc. Natl. Acad. Sci (USA), 94:6432-6437 (1997). The ability of the compounds of the invention to treat reperfusion injury can be demonstrated in the isolated heart according to the method of Tamiya, et al., Immunopharmacology, 29:53-63 (1995), or in the anesthetized dog according to the model of Hartman, et al., Cardiovasc. Res., 30:47-54 (1995). The ability of the compounds of the invention to treat pulmonary reperfusion injury can be demonstrated in a rat lung allograft reperfusion injury model according to the method of DeMeester, et al., Transplantation, 62:1477-1485 (1996), or in a rabbit pulmonary edema model according to the method of Horgan, et al., Am. J. Physiol., 261:H1578-H1584 (1991). The ability of the compounds of the invention to treat stroke can be demonstrated in a rabbit cerebral embolism stroke model according to the method of Bowes, et al., Exp. Neurol., 119:215-219 (1993), in a rat middle cerebral artery ischemia-reperfusion model according to the method of Chopp, et al., Stroke, 25:869-875 (1994), or in a rabbit reversible spinal cord ischemia model according to the method of Clark, et al., Neurosurg., 75:623-627 (1991). The ability of the compounds of the invention to treat cerebral vasospasm can be demonstrated in a rat experimental vasospasm model according to the method of Oshiro, et al., Stroke, 28:2031-2038 (1997). The ability of the compounds of the invention to treat peripheral artery occlusion can be demonstrated in a rat skeletal muscle ischemia/reperfusion model according to the method of Gute, et al., Mol. Cell Biochem., 179:169-187 (1998). The ability of the compounds of the invention to treat graft rejection can be demonstrated in a murine cardiac allograft rejection model according to the method of Isobe, et al., Science, 255:1125-1127 (1992), in a murine thyroid gland kidney capsule model according to the method of Talento, et al., Transplantation, 55:418-422 (1993), in a cynomolgus monkey renal allograft model according to the method of Cosimi, et al., J. Immunol., 144:4604-4612 (1990), in a rat nerve allograft model according to the method of Nakao, et al., Muscle Nerve, 18:93-102 (1995), in a murine skin allograft model according to the method of Gorczynski and Wojcik, J. Immunol., 152:2011-2019 (1994), in a murine corneal allograft model according to the method of He, et al., Opthalmol. Vis. Sci., 35:3218-3225 (1994), or in a xenogeneic pancreatic islet cell transplantation model according to the method of Zeng, et al., Transplantation, 58:681-689 (1994). The ability of the compounds of the invention to treat graft-versus-host disease (GVHD) can be demonstrated in a murine lethal GVHD model according to the method of Harning, et al., Transplantation, 52:842-845 (1991). The ability of the compounds of the invention to treat cancers can be demonstrated in a human lymphoma metastasis model (in mice) according to the method of Aoudjit, et al., J. Immunol., 161:2333-2338 (1998). Combination Treatment The present invention also provides methods for combination therapies in which an agent known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes are used in combination with a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In one aspect, such therapy includes but is not limited to the combination of the subject compound with chemotherapeutic agents, therapeutic antibodies, and radiation treatment, to provide a synergistic or additive therapeutic effect. In one aspect, the compounds or pharmaceutical compositions of the present invention may present synergistic or additive efficacy when administered in combination with agents that inhibit IgE production or activity. Such combination can reduce the undesired effect of high level of IgE associated with the use of one or more PI3Kδ inhibitors, if such effect occurs. This may be particularly useful in treatment of autoimmune and inflammatory disorders (AIID) such as rheumatoid arthritis. Additionally, the administration of PI3Kδ or PI3Kδ/γ inhibitors of the present invention in combination with inhibitors of mTOR may also exhibit synergy through enhanced inhibition of the PI3K pathway. In a separate but related aspect, the present invention provides a combination treatment of a disease associated with PI3Kδ comprising administering a PI3K δ inhibitor and an agent that inhibits IgE production or activity. Other exemplary PI3Kδ inhibitors are applicable for this combination and they are described, e.g., U.S. Pat. No. 6,800,620. Such combination treatment is particularly useful for treating autoimmune and inflammatory diseases (AIID) including but not limited to rheumatoid arthritis. Agents that inhibit IgE production are known in the art and they include but are not limited to one or more of TEI-9874, 2-(4-(6-cyclohexyloxy-2-naphtyloxy)phenylacetamide)benzoic acid, rapamycin, rapamycin analogs (i.e. rapalogs), TORC1/mTORC1 inhibitors, mTORC2/TORC2 inhibitors, and any other compounds that inhibit TORC1/mTORC1 and mTORC2/TORC2. Agents that inhibit IgE activity include, for example, anti-IgE antibodies such as for example Omalizumab and TNX-901. For treatment of autoimmune diseases, the subject compounds or pharmaceutical compositions can be used in combination with commonly prescribed drugs including but not limited to Enbrel®, Remicade®, Humira®, Avonex®, and Rebif®. For treatment of respiratory diseases, the subject compounds or pharmaceutical compositions can be administered in combination with commonly prescribed drugs including but not limited to Xolair®, Advair®, Singulair®, and Spiriva®. The compounds of the invention may be formulated or administered in conjunction with other agents that act to relieve the symptoms of inflammatory conditions such as encephalomyelitis, asthma, and the other diseases described herein. These agents include non-steroidal anti-inflammatory drugs (NSAIDs), e.g. acetylsalicylic acid; ibuprofen; naproxen; indomethacin; nabumetone; tolmetin; etc. Corticosteroids are used to reduce inflammation and suppress activity of the immune system. The most commonly prescribed drug of this type is Prednisone. Chloroquine (Aralen) or hydroxychloroquine (Plaquenil) may also be very useful in some individuals with lupus. They are most often prescribed for skin and joint symptoms of lupus. Azathioprine (Imuran) and cyclophosphamide (Cytoxan) suppress inflammation and tend to suppress the immune system. Other agents, e.g. methotrexate and cyclosporin are used to control the symptoms of lupus. Anticoagulants are employed to prevent blood from clotting rapidly. They range from aspirin at very low dose which prevents platelets from sticking, to heparin/coumadin. In another one aspect, this invention also relates to a pharmaceutical composition for inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, in combination with an amount of an anti-cancer agent (e.g. a chemotherapeutic agent). Many chemotherapeutics are presently known in the art and can be used in combination with the compounds of the invention. In some embodiments, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens. Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-peptide small molecules such as Gleevec (Imatinib Mesylate), Velcade (bortezomib), Iressa (gefitinib), Sprycel (Dasatinib), and Adriamycin as well as a host of chemotherapeutic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN™); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, Casodex™, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, pκ)tfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK.R™; razoxane; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethyl amine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxanes, e.g. paclitaxel (TAXOL™, Bristol-Myers Squibb Oncology, Princeton, NJ) and docetaxel (TAXOTERE™, Rhone-Poulenc Rorer, Antony, France); retinoic acid; esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included as suitable chemotherapeutic cell conditioners are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen (Nolvadex™), raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide (Casodex), leuprolide, and goserelin (Zoladex); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; camptothecin-11 (CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO), 17α-Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Megestrolacetate, Methylprednisolone, Methyl-testosterone, Prednisolone, Triamcinolone, chlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Medroxyprogesteroneacetate, matrix metalloproteinase inhibitors, EGFR inhibitors, Pan Her inhibitors, VEGF inhibitors, including as anti-VEGF antibodies such as Avastin, and small molecules such as ZD6474 and SU6668, vatalanib, BAY-43-9006, SU11248, CP-547632, and CEP-7055. Anti-Her2 antibodies (such as Herceptin from Genentech) may also be utilized. Suitable EGFR inhibitors include gefitinib, erlotinib, and cetuximab. Pan Her inhibitors include canertinib, EKB-569, and GW-572016. Further suitable anticancer agents include, but are not limited to, Src inhibitors, MEK-1 kinase inhibitors, MAPK kinase inhibitors, PI3 kinase inhibitors, and PDGF inhibitors, such as imatinib. Also included are anti-angiogenic and antivascular agents which, by interrupting blood flow to solid tumors, render cancer cells quiescent by depriving them of nutrition. Castration which also renders androgen dependent carcinomas non-proliferative, may also be utilized. Also included are IGF1R inhibitors, inhibitors of non-receptor and receptor tyrosine kinases, and inhibitors of integrin signalling. Additional anticancer agents include microtubule-stabilizing agents 7-O-methylthiomethylpaclitaxel (disclosed in U.S. Pat. No. 5,646,176), 4-desacetyl-4-methylcarbonatepaclitaxel, 3′-tert-butyl-3′-N-tert-butyloxycarbonyl-4-desacetyl-3′-dephenyl-3′-N-debenzoyl-4-O-methoxycarbonyl-paclitaxel (disclosed in U.S. Ser. No. 09/712,352 filed on Nov. 14, 2000), C-4 methyl carbonate paclitaxel, epothilone A, epothilone B, epothilone C, epothilone D, desoxyepothilone A, desoxyepothilone B, [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7-11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17 oxabicyclo [14.1.0]heptadecane-5,9-dione (disclosed in WO 99/02514), [1S-[1R*,3R* (E),7R*10S*,11R*,12R*,16S*]]-3-[2-[2-(aminomethyl)-4-thiazolyl]-1-methyl ethenyl]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-4-17-dioxabicyclo[14.1.0]-heptadecane-5,-9-dione (as disclosed in U.S. Pat. No. 6,262,094) and derivatives thereof; and microtubule-disruptor agents. Also suitable are CDK inhibitors, an antiproliferative cell cycle inhibitor, epidophyllotoxin; an antineoplastic enzyme; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors. Additional cytotoxic agents include, hexamethyl melamine, idatrexate, L-asparaginase, camptothecin, topotecan, pyridobenzoindole derivatives, interferons, and interleukins. Where desired, the compounds or pharmaceutical composition of the present invention can be used in combination with commonly prescribed anti-cancer drugs such as Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, and Velcade® This invention further relates to a method for using the compounds or pharmaceutical composition in combination with radiation therapy in inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the invention in this combination therapy can be determined as described herein. Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy. The term “brachytherapy,” as used herein, refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended without limitation to include exposure to radioactive isotopes (e.g. At-211, 1-131, 1-125, Y-90, Re-186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids. By way of non-limiting example, the radiation source can be a radionuclide, such as 1-125, 1-131, Yb-169, Ir-192 as a solid source, 1-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays. The radioactive material can also be a fluid made from any 5 solution of radionuclides), e.g., a solution of 1-125 or 1-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive micro spheres. Without being limited by any theory, the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, which amount is effective is sensitizing abnormal cells to treatment with radiation. The amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein. The compounds or pharmaceutical compositions of the present invention can be used in combination with an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents. Anti-angiogenesis agents, such as MMP-2 (matrix-metalloprotienase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-II (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the present invention and pharmaceutical compositions described herein. Examples of useful COX-II inhibitors include CELEBREX™ (alecoxib), valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published Oct. 24, 1996), WO 96/27583 (published Mar. 7, 1996), European Patent Application No. 97304971.1 (filed Jul. 8, 1997), European Patent Application No. 99308617.2 (filed Oct. 29, 1999), WO 98/07697 (published Feb. 26, 1998), WO 98/03516 (published Jan. 29, 1998), WO 98/34918 (published Aug. 13, 1998), WO 98/34915 (published Aug. 13, 1998), WO 98/33768 (published Aug. 6, 1998), WO 98/30566 (published Jul. 16, 1998), European Patent Publication 606,046 (published Jul. 13, 1994), European Patent Publication 931, 788 (published Jul. 28, 1999), WO 90/05719 (published May 31, 1990), WO 99/52910 (published Oct. 21, 1999), WO 99/52889 (published Oct. 21, 1999), WO 99/29667 (published Jun. 17, 1999), PCT International Application No. PCT/IB98/01113 (filed Jul. 21, 1998), European Patent Application No. 99302232.1 (filed Mar. 25, 1999), Great Britain Patent Application No. 9912961.1 (filed Jun. 3, 1999), U.S. Provisional Application No. 60/148,464 (filed Aug. 12, 1999), U.S. Pat. No. 5,863,949 (issued Jan. 26, 1999), U.S. Pat. No. 5,861,510 (issued Jan. 19, 1999), and European Patent Publication 780,386 (published Jun. 25, 1997), all of which are incorporated herein in their entireties by reference. Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-I. More preferred, are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (i.e., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-11, MMP-12, and MMP-13). Some specific examples of MMP inhibitors useful in the present invention are AG-3340, RO 32-3555, and RS 13-0830. The invention also relates to a method of and to a pharmaceutical composition of treating a cardiovascular disease in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, or an isotopically-labeled derivative thereof, and an amount of one or more therapeutic agents use for the treatment of cardiovascular diseases. Examples for use in cardiovascular disease applications are anti-thrombotic agents, e.g., prostacyclin and salicylates, thrombolytic agents, e.g., streptokinase, urokinase, tissue plasminogen activator (TPA) and anisoylated plasminogen-streptokinase activator complex (APSAC), anti-platelets agents, e.g., acetyl-salicylic acid (ASA) and clopidrogel, vasodilating agents, e.g., nitrates, calcium channel blocking drugs, antiproliferative agents, e.g., colchicine and alkylating agents, intercalating agents, growth modulating factors such as interleukins, transformation growth factor-beta and congeners of platelet derived growth factor, monoclonal antibodies directed against growth factors, anti-inflammatory agents, both steroidal and non-steroidal, and other agents that can modulate vessel tone, function, arteriosclerosis, and the healing response to vessel or organ injury post intervention. Antibiotics can also be included in combinations or coatings comprised by the invention. Moreover, a coating can be used to effect therapeutic delivery focally within the vessel wall. By incorporation of the active agent in a swellable polymer, the active agent will be released upon swelling of the polymer. Other exemplary therapeutic agents useful for a combination therapy include but are not limited to agents as described above, radiation therapy, hormone antagonists, hormones and their releasing factors, thyroid and antithyroid drugs, estrogens and progestins, androgens, adrenocorticotropic hormone; adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of adrenocortical hormones, insulin, oral hypoglycemic agents, and the pharmacology of the endocrine pancreas, agents affecting calcification and bone turnover: calcium, phosphate, parathyroid hormone, vitamin D, calcitonin, vitamins such as water-soluble vitamins, vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K, and E, growth factors, cytokines, chemokines, muscarinic receptor agonists and antagonists; anticholinesterase agents; agents acting at the neuromuscular junction and/or autonomic ganglia; catecholamines, sympathomimetic drugs, and adrenergic receptor agonists or antagonists; and 5-hydroxytryptamine (5-HT, serotonin) receptor agonists and antagonists. Therapeutic agents can also include agents for pain and inflammation such as histamine and histamine antagonists, bradykinin and bradykinin antagonists, 5-hydroxytryptamine (serotonin), lipid substances that are generated by biotransformation of the products of the selective hydrolysis of membrane phospholipids, eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, nonsteroidal anti-inflammatory agents, analgesic-antipyretic agents, agents that inhibit the synthesis of prostaglandins and thromboxanes, selective inhibitors of the inducible cyclooxygenase, selective inhibitors of the inducible cyclooxygenase-2, autacoids, paracrine hormones, somatostatin, gastrin, cytokines that mediate interactions involved in humoral and cellular immune responses, lipid-derived autacoids, eicosanoids, β-adrenergic agonists, ipratropium, glucocorticoids, methylxanthines, sodium channel blockers, opioid receptor agonists, calcium channel blockers, membrane stabilizers and leukotriene inhibitors. Additional therapeutic agents contemplated herein include diuretics, vasopressin, agents affecting the renal conservation of water, rennin, angiotensin, agents useful in the treatment of myocardial ischemia, anti-hypertensive agents, angiotensin converting enzyme inhibitors, β-adrenergic receptor antagonists, agents for the treatment of hypercholesterolemia, and agents for the treatment of dyslipidemia. Other therapeutic agents contemplated include drugs used for control of gastric acidity, agents for the treatment of peptic ulcers, agents for the treatment of gastroesophageal reflux disease, prokinetic agents, antiemetics, agents used in irritable bowel syndrome, agents used for diarrhea, agents used for constipation, agents used for inflammatory bowel disease, agents used for biliary disease, agents used for pancreatic disease. Therapeutic agents used to treat protozoan infections, drugs used to treat Malaria, Amebiasis, Giardiasis, Trichomoniasis, Trypanosomiasis, and/or Leishmaniasis, and/or drugs used in the chemotherapy of helminthiasis. Other therapeutic agents include antimicrobial agents, sulfonamides, trimethoprim-sulfamethoxazole quinolones, and agents for urinary tract infections, penicillins, cephalosporins, and other, β-Lactam antibiotics, an agent comprising an aminoglycoside, protein synthesis inhibitors, drugs used in the chemotherapy oftuberculosis, Mycobacterium aviumcomplex disease, and leprosy, antifungal agents, antiviral agents including nonretroviral agents and antiretroviral agents. Examples of therapeutic antibodies that can be combined with a subject compound include but are not limited to anti-receptor tyrosine kinase antibodies (cetuximab, panitumumab, trastuzumab), anti CD20 antibodies (rituximab, tositumomab), and other antibodies such as alemtuzumab, bevacizumab, and gemtuzumab. Moreover, therapeutic agents used for immunomodulation, such as immunomodulators, immunosuppressive agents, tolerogens, and immunostimulants are contemplated by the methods herein. In addition, therapeutic agents acting on the blood and the blood-forming organs, hematopoietic agents, growth factors, minerals, and vitamins, anticoagulant, thrombolytic, and antiplatelet drugs. Further therapeutic agents that can be combined with a subject compound may be found in Goodman and Gilman's “The Pharmacological Basis of Therapeutics” Tenth Edition edited by Hardman, Limbird and Gilman or the Physician's Desk Reference, both of which are incorporated herein by reference in their entirety. The compounds described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the compounds of the invention will be co-administered with other agents as described above. When used in combination therapy, the compounds described herein may be administered with the second agent simultaneously or separately. This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described above can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the present invention and any of the agents described above can be simultaneously administered, wherein both the agents are present in separate formulations. In another alternative, a compound of the present invention can be administered just followed by and any of the agents described above, or vice versa. In the separate administration protocol, a compound of the present invention and any of the agents described above may be administered a few minutes apart, or a few hours apart, or a few days apart. The methods in accordance with the invention may include administering a PI3-kinase δ selective inhibitor with one or more other agents that either enhance the activity of the inhibitor or compliment its activity or use in treatment. Such additional factors and/or agents may produce an augmented or even synergistic effect when administered with a PI3-kinase δ selective inhibitor, or minimize side effects. In one embodiment, the methods of the invention may include administering formulations comprising a PI3-kinase δ selective inhibitor of the invention with a particular cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent before, during, or after administration of the PI3-kinase δ selective inhibitor. One of ordinary skill can easily determine if a particular cytokine, lymphokine, hematopoietic factor, thrombolytic of anti-thrombotic factor, and/or anti-inflammatory agent enhances or compliments the activity or use of the PI3-kinase δ selective inhibitors in treatment. More specifically, and without limitation, the methods of the invention may comprise administering a PI3-kinase δ selective inhibitor with one or more of TNF, IL-1, IL-2, IL-3, IL4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IFN, G-CSF, Meg-CSF, GM-CSF, thrombopoietin, stem cell factor, and erythropoietin. Compositions in accordance with the invention may also include other known angiopoietins such as Ang-2, Ang4, and Ang-Y, growth factors such as bone morphogenic protein-1, bone morphogenic protein-2, bone morphogenic protein-3, bone morphogenic protein-4, bone morphogenic protein-5, bone morphogenic protein-6, bone morphogenic protein-7, bone morphogenic protein-8, bone morphogenic protein-9, bone morphogenic protein-10, bone morphogenic protein-11, bone morphogenic protein-12, bone morphogenic protein-13, bone morphogenic protein-14, bone morphogenic protein-15, bone morphogenic protein receptor IA, bone morphogenic protein receptor IB, brain derived neurotrophic factor, ciliary neutrophic factor, ciliary neutrophic factor receptor a, cytokine-induced neutrophil chemotactic factor 1, cytokine-induced neutrophil chemotactic factor 2 alpha, cytokine-induced neutrophil chemotactic factor 2 beta, beta endothelial cell growth factor, endothelin 1, epidermal growth factor, epithelial-derived neutrophil attractant, fibroblast growth factor 4, fibroblast growth factor 5, fibroblast growth factor 6, fibroblast growth factor 7, fibroblast growth factor 8, fibroblast growth factor 8b, fibroblast growth factor 8c, fibroblast growth factor 9, fibroblast growth factor 10, fibroblast growth factor acidic, fibroblast growth factor basic, glial cell line-derived neutrophic factor receptor al, glial cell line-derived neutrophic factor receptor a2, growth related protein, growth related protein a, growth related protein .beta., growth related protein .gamma., heparin binding epidermal growth factor, hepatocyte growth factor, hepatocyte growth factor receptor, insulin-like growth factor I, insulin-like growth factor receptor, insulin-like growth factor II, insulin-like growth factor binding protein, keratinocyte growth factor, leukemia inhibitory factor, leukemia inhibitory factor receptor alpha, nerve growth factor, nerve growth factor receptor, neurotrophin-3, neurptrophin-4, placenta growth factor, placenta growth factor 2, platelet derived endothelial cell growth factor, platelet derived growth factor, platelet derived growth factor A chain, platelet derived growth factor AA, platelet derived growth factor AB, platelet derived growth factor B chain, platelet derived growth factor BB, platelet derived growth factor receptor a, platelet derived growth factor receptor beta, pre-B cell growth stimulating factor, stem cell factor, stem cell factor receptor, transforming growth factor alpha, transforming growth factor beta, transforming growth factor beta 1, transforming growth factor beta 1.2, transforming growth factor beta 2, transforming growth factor beta 3, transforming growth factor beta 5, latent transforming growth factor beta 1, transforming growth factor beta binding protein I, transforming growth factor beta binding protein II, transforming growth factor beta binding protein III, tumor necrosis factor receptor type I, tumor necrosis factor receptor type II, urokinase-type plasminogen activator receptor, and chimeric proteins and biologically or immunologically active fragments thereof. The following general methodology described herein provides the manner and process of making and using the compound of the present invention and are illustrative rather than limiting. Further modification of provided methodology and additionally new methods may also be devised in order to achieve and serve the purpose of the invention. Accordingly, it should be understood that there may be other embodiments which fall within the spirit and scope of the invention as defined by the specification hereto. Representative compounds of the present invention include those specified above in Table 1 and pharmaceutically acceptable salts thereof. The present invention should not be construed to be limited to them. General Method of Preparation of Compounds of the Invention The compounds of the present invention may be prepared by the following processes. Unless otherwise indicated, the variables (e.g., R, R1, R2, L1, Cy1and Cy2) when used in the below formulae are to be understood to present those groups described above in relation to formula (I). These methods can similarly be applied to other compounds of formula IA, IA-I, IA-II, IA-III and/or IA-IV. Scheme 1: This scheme provides a general process for synthesis of a compound of formula (I) wherein all the variables R, R1, R2, L1, Cy1and Cy2are as described above in relation to formula (I) Compound of formula (1) wherein Ra is Hydrogen or alkyl can be converted to compound of formula (3) by reacting with a compound of formula (2) wherein LG is a leaving group such as a halogen or an acyl group in the presence of a lewis acid such as aluminium chloride or boron trifluoride. Compound of formula (3) can be converted to Compound of formula (5) by Kostanecki acylation, i.e., by treating with an anhydride of formula (4), wherein R1and R2is hydrogen or substituted or unsubstituted C1-6alkyl in the presence of a base. (See Von Kostanecki, S., Rozycki, A., in Ber. 1901, 34, 102 and by Baker, W. in J. Chem. Soc., 1933, 1381). Compound of formula (5) can then be converted to a compound of formula (6) using a suitable halogenating condition that is known to those skilled in the art. For example, by using bromine in a polar solvent such as acetic acid or N,N-dimethyl formamide or by using a N-halosuccinimide in the presence of a suitable radical initiator such as azabis(isobutyronitrile) or benzoyl peroxide. Compounds of formula (6) can then be reacted with a compound of formula Cy2-L1-H in the presence of a suitable inorganic base such as potassium carbonate or sodium hydride or an organic base such as triethylamine or N,N-diisopropylethylamine to afford the desired compound of formula (I) wherein R1& R2are hydrogen or C1-C6alkyl, Cy1is monocyclic or bicyclic substituted or unsubstituted aryl and L1, R, and Cy2are the same as described above in relation to formula (I). Scheme 1A: This scheme provides a general process for synthesis of a compound of formula (I) wherein Cy1is Cy2is X is CRaor N and all the variables R, R1, R2, L1, and Raare as described above in relation to formula (I). By starting with a suitable anisole derivative (1a) and a phenylacetic acid derivative (2a), compounds of formula (6a) can be synthesised as described in scheme 1 for synthesis of compound of formula (6). Compound of formula (6a) can be reacted with compound of formula (7) wherein X is chosen from CH or N and different occurrence of X can be same or different and Y is chosen from N, CH, C-Hal or C-Ar or C-Het in the presence of a base to afford the desired compound of formula (I) wherein Cy1is Cy2is X is CRaor N and all the variables R, R1, R2, L1, and Raare as described above in relation to formula (I). Using similar methodologies as described above in Scheme 1 & 1A with certain modifications as known to those skilled in the art can be used to synthesize compounds of formula IA-I and/or IA-II wherein the variables are to be understood to present those groups described above in relation to formula IA-I, IA-II and/or IA-IV using suitable intermediates and reagents For example as illustrated below Scheme 1B: This scheme provides a method for preparation of compound of formula IA-II wherein R1& R2are hydrogen or substituted or unsubstituted C1-6alkyl, R3 is substituted or unsubstituted aryl or heteroaryl, Cy1is monocyclic substituted or unsubstituted aryl and R is the same as described above in relation to formula (I) As illustrated in scheme 1B, compound of formula (Ia) wherein Y═C-Hal, i.e., compound of formula (Ib) can be further subjected to a Suzuki reaction to give compound of formula (IA-IIa) wherein R3is substituted or unsubstituted aryl or heteroaryl. Thus, compound of formula (Ib) can be reacted with a boronic acid or its ester of formula (8), wherein ring R3is an substituted or unsubstituted aryl or heteroomatic or heteroaromatic ring, in the presence of a suitable palladium catalyst such as tetrakis(triphenylphosphine)palladium (0) or [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (II) in the presence of a base such as an alkali metal carbonate to afford compound of formula (IA-IIa). Alternately under Sonogashira reaction conditions, compound of formula (Ib) can be reacted with a compound of formula (9) wherein Rais the same as described above in relation to formula (I)), in the presence of a palladium catalyst, to give compound of formula (IA-IIb) wherein R3is substituted or unsubstituted alkynyl. The Suzuki reaction and Sonogashira reaction can be performed under standard thermal conditions or optionally may also be assisted by microwave irradiation. Scheme 2: This scheme provides a method for preparation of compound of formula I wherein R1& R2are hydrogen or substituted or unsubstituted C1-6alkyl, Cy1is monocyclic substituted or unsubstituted aryl and L1, R, and Cy2are the same as described above in relation to formula (I) Compound of formula (3) can be converted to compound of formula (6b) by reacting with a compound of formula (4b) wherein L1is a heteroatom containing functional group and PG is a protecting group in the presence of an ester coupling reagent such as 2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate (HATU) or 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate (HBTU). Deprotection of compound of formula (6b) can give a compound of formula (6c). Compound of formula (6c) can then be reacted with a compound of formula Cy2-Lg wherein Lg is a good leaving group such as Halogen in the presence of a suitable base such as potassium carbonate or sodium hydride to provide the desired compounds of formula (I) wherein R1& R2are hydrogen or substituted or unsubstituted C1-6alkyl, Cy1is monocyclic substituted or unsubstituted aryl and L1, R, and Cy2are the same as described above in relation to formula (I) Scheme 2A: This scheme provides a method for preparation of compound of formula IA-IIIa wherein R1& R2are hydrogen or substituted or unsubstituted C1-6alkyl, Cy1is substituted or unsubstituted Phenyl, Cy2is L1is NH and R, n and Cy2are the same as described above in relation to formula (IA-III) The compound of formula (3a) can be reacted with an N-protected amino acid of formula (4b1) in the presence of an ester coupling reagent such as 2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate (HATU) or 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate (HBTU) to give compound of formula (6b1). The amine protecting group of (6b1) can be removed to give compound of formula (6c1). Compound of formula (6c1) upon reaction with compound of formula (7a) can give compound of formula (IA-IIIa) wherein R1& R2are hydrogen or substituted or unsubstituted C1-6alkyl, Cy1is substituted or unsubstituted Phenyl, Cy2is L1is NH and R, n and Cy2are the same as described above in relation to formula (IA-III). Optionally the coupling of (6c1) with a compound of formula (7a) may be performed in the absence of a base with the assistance of microwave irradiation. Using similar methodologies as described above in Scheme 2 & 2A with certain modifications as known to those skilled in the art can be used to synthesize compounds of formula IA-III. wherein the variables are to be understood to present those groups described above in relation to formula IA-III and/or IA-IV using suitable intermediates and reagents For example as illustrated below EXPERIMENTAL Unless otherwise mentioned, work-up implies distribution of reaction mixture between the aqueous and organic phases indicated within parenthesis, separation and drying over Na2SO4of the organic layer and evaporating the solvent to give a residue. RT implies ambient temperature (25-28° C.). The terms “solvent,” “organic solvent,” or “inert solvent” each mean a solvent inert under the conditions of the reaction being described in conjunction therewith including, for example, benzene, toluene, acetonitrile, tetrahydrofuran (“THF”), dimethylformamide (“DMF”), chloroform, methylene chloride (or dichloromethane), diethyl ether, methanol, N-methylpyrrolidone (“NMP”), pyridine and the like. Unless specified to the contrary, the solvents used in the reactions described herein are inert organic solvents. Unless specified to the contrary, for each gram of the limiting reagent, one cc (or ml) of solvent constitutes a volume equivalent. Isolation and purification of the chemical entities and intermediates described herein can be effected, if desired, by any suitable separation or purification procedure such as, for example, filtration, extraction, crystallization, column chromatography, thin-layer chromatography or thick-layer chromatography, or a combination of these procedures. Specific illustrations of suitable separation and isolation procedures can be had by reference to the examples herein below. However, other equivalent separation or isolation procedures can also be used. Unless otherwise stated, purification implies column chromatography using silica gel as the stationary phase and a mixture of petroleum ether (boiling at 60-80° C.) and ethyl acetate or dichloromethane and methanol of suitable polarity as the mobile phases. When desired, the (R)- and (S)-isomers of the compounds of the present invention, if present, may be resolved by methods known to those skilled in the art, for example by formation of diastereoisomeric salts or complexes which may be separated, for example, by crystallization; via formation of diastereoisomeric derivatives which may be separated, for example, by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic oxidation or reduction, followed by separation of the modified and unmodified enantiomers; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support, such as silica with a bound chiral ligand or in the presence of a chiral solvent. Alternatively, a specific enantiomer may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer to the other by asymmetric transformation. The compounds described herein can be optionally contacted with a pharmaceutically acceptable acid to form the corresponding acid addition salts. Many of the optionally substituted starting compounds and other reactants are commercially available, e.g., from Sigma Aldrich Chemical Company, Alfa Aesar ( ) or can be readily prepared by those skilled in the art using commonly employed synthetic methodology. For instance—various boronic acids which are used can be obtained commercially from various sources. The compounds of the invention can generally be synthesized by an appropriate combination of generally well known synthetic methods. Techniques useful in synthesizing these chemical entities are both readily apparent and accessible to those of skill in the relevant art, based on the instant disclosure. The compounds of the invention can be synthesized by an appropriate combination of known synthetic methods in the art. The discussion below is offered to illustrate certain of the diverse methods available for use in making the compounds of the invention and is not intended to limit the scope of reactions or reaction sequences that can be used in preparing the compounds of the present invention. The examples and preparations provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations. In the following examples molecules with a single chiral center, unless otherwise noted, exist as a racemic mixture. Those molecules with two or more chiral centers, unless otherwise noted, exist as a racemic mixture of diastereomers. Single enantiomers/diastereomers may be obtained by methods known to those skilled in the art. Intermediate 1: 1-(5-Bromo-2-hydroxyphenyl)-2-phenylethanone Phenylacetic acid (1.09 g, 8.0 mmoles) was dissolved in 5 ml dichloromethane. To this mixture, oxalylchloride (1.01 g, 8.0 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 5 ml dichloromethane. To this mixture, 4-bromoanisole (1 g, 5.34 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(1.06 g, 8.0 mmoles) was added and the reaction mixture was warmed to RT and stirred overnight. The reaction mixture was quenched by the addition of 2N HCl and extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography to afford the title compound as white solid (1 g, 66% yield). MP: 83-86° C.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 11.56 (s, 1H), 8.01 (d, J=2.2 Hz, 1H), 7.64 (dd, J=8.8, 2.5 Hz, 1H), 7.32 (t, 2H), 7.29 (m, 3H), 6.96 (d, J=8.8 Hz, 1H), 4.43 (s, 2H). Intermediate 2: 6-Bromo-2-methyl-3-phenyl-4H-chromen-4-one Intermediate 1 (8.9 g, 30.56 mmoles) was taken in a RB flask and to this acetic anhydride (59 ml) and sodium acetate (17.5 g, 213 mmoles) were added and the mixture was refluxed for 12 h. After cooling to RT, the reaction mixture was quenched by the addition of ice cold water. The solid formed was filtered and washed with water. The product was dried under vacuum to afford the title compound as white solid (9.4 g, 97% yield). MP: 119-121° C.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.35 (d, J=2.4 Hz, 1H), 7.75 (dd, J=11.3, 2.4 Hz, 1H), 7.46 (t, 2H), 7.39 (d, J=7.2 Hz, 1H), 7.36 (d, J=8.8 Hz, 1H), 7.28 (m, 2H), 2.32 (s, 3H). Intermediate 3: 6-Bromo-2-(bromomethyl)-3-phenyl-4H-chromen-4-one To a solution of Intermediate 2 (4.5 g, 14.27 mmoles) in carbon tetrachloride (60 ml) N-bromosuccinimide (2.5 g, 14.27 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (45 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was recrystallised from ethyl acetate:petroleum ether (5:95) to afford the title compound as off white solid (3.3 g, 59% yield). MP: 172-175° C.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.35 (d, J=2.4 Hz, 1H), 7.80 (dd, J=8.8, 2.4 Hz, 1H), 7.50-7.36 (m, 6H), 4.23 (s, 2H). Intermediate 4: 2-Methyl-3-phenyl-4H-chromen-4-one To a solution of intermediate 2 (3 g, 9.51 mmoles) in ethanol (30 ml), ammonium formate (6 g, 95.18 mmoles) and palladium on carbon (10%, 300 mg) were added and the solution was refluxed for 2 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as off-white solid (1.98 g, 86% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.17 (dd, J=7.9, 1.4 Hz, 1H), 7.61 (dt, J=8.5, 1.5 Hz, 1H), 7.38-7.28 (m, 5H), 7.25 (m, 2H), 2.25 (s, 3H). Intermediate 5: 2-(Bromomethyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 4 (1.9 g, 8.07 mmoles) in carbon tetrachloride (30 ml) N-bromosuccinimide (1.43 g, 8.07 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (20 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as off white solid (1.62 g, 65% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.06 (d, J=7.6 Hz, 1H), 7.87 (t, J=7.7 Hz, 1H), 7.71 (d, J=8.5 Hz, 1H), 7.53-7.41 (m, 4H), 7.35 (d, J=6.8 Hz, 2H), 4.37 (s, 2H). Intermediate 6: 1-(5-Bromo-2-hydroxyphenyl)-2-(4-fluorophenyl)ethanone 4-Fluoro phenylacetic acid (12.3 g, 79.79 mmoles) was dissolved in 30 ml dichloromethane. To this mixture, oxalylchloride (10.17 g, 79.79 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 30 ml dichloromethane. To this mixture, 4-bromoanisole (10 g, 53.47 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(10.6 g, 79.79 mmoles) was added and the reaction mixture was warmed to RT and stirred overnight. The reaction mixture was quenched by the addition of 2N HCl and extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as white solid (6.1 g, 37% yield.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 12.05 (s, 1H), 7.96 (d, J=2.3 Hz, 1H), 7.58 (dd, J=8.9, 2.4 Hz, 1H), 7.24 (dt, J=5.4, 1.9 Hz, 2H), 7.09 (dt, J=8.6, 2.1 Hz, 2H), 6.79 (d, J=8.7 Hz, 1H), 4.27 (s, 2H). Intermediate 7: 6-Bromo-3-(4-fluorophenyl)-2-methyl-4H-chromen-4-one Intermediate 6 (6.1 g, 19.73 mmoles) was taken in a RB flask and to this acetic anhydride (40 ml) and sodium acetate (11.3 g, 137.75 mmoles) were added and the mixture was refluxed for 12 h. After cooling to RT, the reaction mixture was quenched by the addition of ice cold water. The solid formed was filtered and washed with water. The product was dried under vacuum to afford the title compound as white solid (4.1 g, 63% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.35 (d, J=1.9 Hz, 1H), 7.77 (dd, J=8.8, 1.9 Hz, 1H), 7.37 (d, J=8.9 Hz, 1H), 7.27 (t, J=5.7 Hz, 2H), 7.17 (t, J=8.6 Hz, 2H), 2.33 (s, 3H). Intermediate 8: 6-Bromo-2-(bromomethyl)-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 7 (2 g, 6.00 mmoles) in carbon tetrachloride (20 ml) N-bromo-succinimide (1.06 g, 5.95 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (20 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as off white solid (1.20 g, 50% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.35 (d, J=2.4 Hz, 1H), 7.81 (dd, J=8.9, 2.4 Hz, 1H), 7.43 (d, J=8.9 Hz, 1H), 7.38 (dt, J=5.4, 2.0 Hz, 2H), 7.20 (t, J=8.6 Hz, 2H), 4.22 (s, 2H). Intermediate 9: 3-(4-Fluorophenyl)-2-methyl-4H-chromen-4-one To a solution of intermediate 7 (1.5 g, 4.50 mmoles) in ethanol (15 ml), ammonium formate (2.8 g, 45.02 mmole) and palladium on carbon (10%, 15 mg) were added and the solution was refluxed for 4 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as white solid (0.8 g, 72% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.71 (d, J=7.8 Hz, 1H), 7.69 (t, J=7.35 Hz, 1H), 7.47 (d, J=8.4 Hz, 1H), 7.42 (t, J=7.4 Hz, 1H), 7.29 (t, J=9.5 Hz, 2H), 7.16 (t, J=8.5 Hz, 2H), 2.33 (s, 3H). Intermediate 10: 2-(Bromomethyl)-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 9 (0.80 g, 3.146 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.560 g, 3.146 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (8 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as off white solid (0.7 g, 67% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.23 (dd, J=7.9, 1.3 Hz, 1H), 7.74 (dt, J=8.6, 1.5 Hz, 1H), 7.53 (d, J=8.3 Hz, 1H), 7.45 (m, 3H), 7.19 (t, J=8.7 Hz, 2H), 4.24 (s, 2H). Intermediate 11: 1-(5-bromo-2-hydroxyphenyl)-2-o-tolylethanone 2-Methylphenylacetic acid (9.60 g, 64.15 mmoles) was dissolved in 10 ml dichloromethane. To this mixture, oxalylchloride (7 ml, 80.19 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 100 ml dichloromethane. To this mixture, 4-bromoanisole (10 g, 53.47 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(10.6 g, 80.19 mmoles) was added and the reaction mixture was warmed to RT and stirred for 24 h. The reaction mixture was quenched by the addition of 2N HCl and extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as white solid (5.5 g, 33% yield.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 11.52 (s, 1H), 8.02 (d, J=2.4 Hz, 1H), 7.65 (dd, J=8.8, 2.5 Hz, 1H), 7.16 (m, 4H), 6.97 (d, J=8.9 Hz, 1H), 4.47 (s, 2H), 2.14 (s, 3H). Intermediate 12: 6-bromo-2-methyl-3-o-tolyl-4H-chromen-4-one Intermediate 11 (5.5 g, 16.38 mmoles) was taken in a RB flask and to this acetic anhydride (50 ml) and sodium acetate (9.40 g, 114.69 mmoles) were added and the mixture was refluxed for 12 h. After cooling to RT, the reaction mixture was quenched by the addition of ice cold water. The solid formed was filtered and washed with water. The product was dried under vacuum to afford the title compound as white solid (1.8 g, 30% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.35 (d, J=1.7 Hz, 1H), 7.75 (d, J=6.7 Hz, 1H), 7.37 (d, J=8.8 Hz, 1H), 7.35-7.26 (m, 3H), 7.09 (d, J=6.9 Hz, 1H), 2.20 (s, 3H). 2.15 (s, 3H). Intermediate 13: 6-Bromo-2-(bromomethyl)-3-o-tolyl-4H-chromen-4-one To a solution of intermediate 12 (0.20 g, 0.607 mmoles) in acetic acid (3 ml) bromine (0.03 ml, 1.21 mmoles) was added at 0° C. The reaction mixture heated to 60° C. After 3 h, the reaction mixture was cooled to RT, quenched by the addition of water. The precipitate formed was filtered and dried under reduced pressure to afford the title compound as off white solid (0.176 g, 71% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.35 (d, J=2.2 Hz, 1H), 7.87 (dd, J=8.9, 2.3 Hz, 1H), 7.45 (d, J=8.9 Hz, 1H), 7.39 (m, 3H), 7.17 (d, J=7.3 Hz), 7.12 (d, J=7.5 Hz) (total 1H), 4.20 (d, J=10.8 Hz), 4.08 (d, J=10.7 Hz) (total, 2H), 2.17 (s, 3H). Intermediate 14: 6-Bromo-2-ethyl-3-phenyl-4H-chromen-4-one Intermediate 1 (2.0 g, 6.86 mmoles) was taken in a RB flask and to this triethylamine (16 ml) and propionic anhydride (2.80 g, 21.50 mmoles) were added and the mixture was refluxed for 22 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.78 g, 31% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.10 (d, J=2.4 Hz, 1H), 7.97 (dd, J=8.9, 2.4 Hz, 1H), 7.68 (d, J=8.9 Hz, 1H), 7.46 (m, 3H), 7.27 (d, J=6.9 Hz, 2H), 2.55 (q, J=7.5 Hz, 2H), 1.19 (t, J=7.5 Hz, 3H). Intermediate 15: 6-Bromo-2-(1-bromoethyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 14 (1.0 g, 3.03 mmoles) in carbon tetrachloride (25 ml) N-bromosuccinimide (0.540 g, 3.03 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (5 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as off white solid (0.6 g, 50% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.11 (d, J=2.5 Hz, 1H), 8.04 (dd, J=8.9, 2.5 Hz, 1H), 7.78 (d, J=9.0 Hz, 1H), 7.51 (m, 3H), 7.32 (dd, J=8.1, 1.7 Hz, 2H), 4.97 (q, J=6.8 Hz, 1H), 1.96 (d, J=6.8 Hz, 3H). Intermediate 16: (S)-tert-butyl 1-(6-bromo-4-oxo-3-phenyl-4H-chromen-2-yl)ethylcarbamate To a solution of intermediate 1 (5 g, 17.17 mmoles) in dichloromethane (50 ml), triethylamine (5.2 g, 51.52 mmoles) was added followed by L-N-Boc-Alanine (3.5 g, 18.89 mmoles). To this mixture HATU (13 g, 34.34 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as yellow solid (1.6 g, 21% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.10 (d, J=2.4 Hz, 1H), 7.99 (dd, J=8.9, 2.5 Hz, 1H), 7.62 (d, J=8.9 Hz, 1H), 7.53 (d, J=6.8 Hz, 1H), 7.47 (m, 3H), 7.29 (d, J=7.0 Hz, 2H), 4.49 (q, J=6.9 Hz, 1H), 1.33 (s, 9H), 1.29 (d, J=7.1 Hz, 3H). Intermediate 17: (S)-2-(1-aminoethyl)-6-bromo-3-phenyl-4H-chromen-4-one To a solution of intermediate 16 (0.81 g, 1.821 mmoles) in dichloromethane (10 ml), trifluoroacetic acid (1.4 ml, 18.21 mmoles) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as yellow solid (0.675 g).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.10 (d, J=2.4 Hz, 1H), 8.00 (dd, J=8.9, 2.5 Hz, 1H), 7.69 (d, J=8.9 Hz, 1H), 7.46 (m, 4H), 7.30 (d, J=7.0 Hz, 2H), 7.28 (m, 1H), 3.78 (q, J=6.7 Hz, 1H), 1.29 (d, J=6.7 Hz, 3H). Intermediate 18: tert-Butyl(6-bromo-4-oxo-3-phenyl-4H-chromen-2-yl)methylcarbamate To a solution of intermediate 1 (2 g, 6.86 mmoles) in dichloromethane (20 ml), triethylamine (2.08 g, 51.52 mmoles) was added followed by N-Boc-Glycine (1.3 g, 7.55 mmoles). To this mixture HATU (5.2 g, 13.67 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as yellow solid (1.0 g, 33% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.12 (d, J=2.3 Hz, 1H), 7.99 (dd, J=8.9, 2.5 Hz, 1H), 7.59 (d, J=8.9 Hz, 1H), 7.476 (m, 4H), 7.31 (d, J=6.3 Hz, 2H), 4.06 (d, J=5.6 Hz, 2H), 1.37 (s, 9H). Intermediate 19: 2-(Amino methyl)-6-bromo-3-phenyl-4H-chromen-4-one To a solution of intermediate 18 (0.440 g, 1.02 mmoles) in dichloromethane (5 ml), trifluoroacetic acid (3 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as brown liquid (0.400 g). The crude product was taken for next step. Intermediate 20: 1-(2-Hydroxy-5-methoxyphenyl)-2-phenylethanone Phenylacetic acid (7.39 g, 54.28 mmoles) was dissolved in 50 ml dichloromethane. To this mixture, oxalylchloride (4.74 ml, 54.28 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 30 ml dichloromethane. To this mixture, 4-methoxyanisole (10 g, 53.47 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(9.63 g, 72.37 mmoles) was added and the reaction mixture was warmed to RT and stirred for 12 h. The reaction mixture was quenched by the addition of 2N HCl and extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as yellow liquid (4.3 g, 49% yield.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 11.30 (s, 1H), 7.42 (d, J=3.0 Hz, 1H), 7.33-7.21 (m, 5H), 7.17 (dd, J=9.0, 3.1 Hz, 1H), 6.92 (d, J=9.0 Hz, 1H), 4.43 (s, 2H), 3.74 (s, 3H). Intermediate 21: 6-Methoxy-2-methyl-3-phenyl-4H-chromen-4-one Intermediate 20 (4 g, 16.51 mmoles) was taken in a RB flask and to this acetic anhydride (40 ml) and sodium acetate (9.48 g, 115.57 mmoles) were added and the mixture was refluxed for 12 h. After cooling to RT, the reaction mixture was quenched by the addition of ice cold water. The solid formed was filtered and washed with water. The product was dried under vacuum to afford the title compound as yellow solid (3 g, 68% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 7.60 (d, J=3.0 Hz, 1H), 7.45 (t, J=7.1 Hz, 2H), 7.37 (m, 2H), 7.29 (m, 3H), 3.89 (s, 3H). 2.31 (s, 3H). Intermediate 22: 2-(Bromomethyl)-6-methoxy-3-phenyl-4H-chromen-4-one To a solution of intermediate 21 (2.0 g, 7.501 mmoles) in carbon tetrachloride (25 ml) N-bromosuccinimide (1.30 g, 7.510 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (25 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off white solid (2.6 g).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 7.68 (d, J=9.1 Hz, 1H), 7.53 (m, 5H), 7.34 (d, J=6.7, 2H), 4.36 (s, 2H), 3.85 (s, 3H). Intermediate 23: 1-(5-Bromo-2-hydroxyphenyl)-2-(2-fluorophenyl)ethanone 2-Fluorophenylacetic acid (2.96 g, 19.24 mmoles) was dissolved in 50 ml dichloromethane. To this mixture, oxalylchloride (2.1 ml, 24.05 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 30 ml dichloromethane. To this mixture, 4-bromoanisole (3.0 g, 16.03 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(3.21 g, 24.05 mmoles) was added and the reaction mixture was warmed to RT and stirred for 12 h. The reaction mixture was quenched by the addition of 2N HCl and extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (4.0 g, 81% yield.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 11.97 (s, 1H), 8.01 (d, J=1.7 Hz, 1H), 7.58 (dd, J=8.8, 2.3.1 Hz, 1H), 7.35 (m, 1H), 7.23 (d, J=7.3 Hz, 1H), 7.17 (m, 2H), 6.92 (d, J=8.9 Hz, 1H), 4.33 (s, 2H). Intermediate 24: 6-Bromo-2-ethyl-3-(2-fluorophenyl)-4H-chromen-4-one Intermediate 23 (1.1 g, 3.55 mmoles) was taken in a RB flask and to this triethylamine (10 ml) and propionic anhydride (1.44 g, 11.13 mmoles) were added and the mixture was refluxed for 22 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.800 g, 65% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.10 (d, J=2.5 Hz, 1H), 8.00 (dd, J=9.0, 2.5 Hz, 1H), 7.71 (d, J=9.0 Hz, 1H), 7.51 (m, 1H), 7.36 (m, 3H), 2.54 (m, 2H), 1.19 (t, J=7.6 Hz, 3H). Intermediate 25: 6-Bromo-2-(1-bromoethyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 24 (0.620 g, 1.785 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.317 g, 1.785 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (15 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off white solid consisting of two atrop-isomers (0.625 g).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.13 (t, J=2.3 Hz, 1H), [8.07 (dd, J=2.4, 1.0 Hz), 8.04 (dd, J=2.5, 1.1 Hz), 1H], 7.81 (dd, J=8.8, 1.6 Hz, 1H), 7.57 (m, 1H), 7.39 (m, 3H), [4.99 (q, J=6.8 Hz), 4.93 (q, J=6.8 Hz), 1H], [1.99 (q, J=6.8 Hz), 1.44 (q, J=6.8 Hz), 3H]. Intermediate 26: 6-Bromo-3-(2-fluorophenyl)-2-methyl-4H-chromen-4-one Intermediate 23 (5 g, 16.17 mmoles) was taken in a RB flask and to this acetic anhydride (40 ml) and sodium acetate (9.2 g, 82.03 mmoles) were added and the mixture was refluxed for 12 h. After cooling to RT, the reaction mixture was quenched by the addition of ice cold water. The solid formed was filtered and washed with water. The product was dried under vacuum to afford the title compound as off-white solid (3.81 g, 71% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.34 (d, J=2.3 Hz, 1H), 7.76 (dd, J=8.8, 2.2 Hz, 1H), 7.41 (m, 2H), 7.24 (m, 2H), 7.18 (t, J=8.9 Hz, 1H), 2.30 (s, 3H). Intermediate 27: 6-Bromo-2-(bromomethyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 26 (2.0 g, 6.00 mmoles) in carbon tetrachloride (20 ml) N-bromosuccinimide (1.0 g, 6.00 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (25 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off white solid (1.86 g).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.34 (d, J=2.3 Hz, 1H), 7.82 (dd, J=8.9, 2.3 Hz, 1H), 7.44 (d, J=8.8 Hz, 1H), 7.38 (t, J=6.2 Hz, 1H), 7.29 (m, 2H), [4.22 (d, J=11.0 Hz), 4.17 (d, J=11.1 Hz), 2H]. Intermediate 28: 2-Ethyl-3-phenyl-4H-chromen-4-one To a solution of intermediate 24 (1.0 g, 3.03 mmoles) in ethanol (10 ml), ammonium formate (1.9 g, 30.14 mmoles) and palladium on carbon (10%, 100 mg) were added and the solution was refluxed for 4 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as white solid (0.50 g, 66% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.24 (dd, J=7.9, 1.4 Hz, 1H), 7.68 (dt, J=8.6, 1.6 Hz, 1H), 7.48-7.35 (m, 5H), 7.28 (dd, J=8.3, 1.4 Hz, 2H), 2.62 (q, J=7.5 Hz, 2H), 1.28 (t, J=7.5 Hz, 3H). Intermediate 29: 2-(1-Bromoethyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 28 (0.550 g, 2.20 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.392 g, 2.20 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (5 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as yellow solid (0.680 g, 94% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.24 (dd, J=8.0, 1.7 Hz, 1H), 7.74 (dt, J=7.2, 1.6 Hz, 1H), 7.57 (d, J=8.0 Hz, 1H), 7.49-7.26 (m, 6H), 4.99 (q, J=6.9 Hz, 1H), 1.99 (d, J=6.9 Hz, 3H). Intermediate 30: 6-Bromo-3-phenyl-2-propyl-4H-chromen-4-one Intermediate 1 (3.0 g, 10.30 mmoles) was taken in a RB flask and to this triethylamine (30 ml) and butyric anhydride (5.12 g, 32.37 mmoles) were added and the mixture was refluxed for 22 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (2.0 g, 56% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.10 (d, J=2.4 Hz, 1H), 7.97 (dd, J=8.9, 2.5 Hz, 1H), 7.68 (d, J=8.9 Hz, 1H), 7.46 (m, 3H), 7.26 (dd, J=8.2, 1.3 Hz, 2H), 2.49 (t, J=1.6 Hz, 2H), 1.66 (m, 2H), 0.84 (t, J=7.4 Hz, 3H). Intermediate 31: 3-Phenyl-2-propyl-4H-chromen-4-one To a solution of intermediate 30 (1.5 g, 4.37 mmoles) in ethanol (15 ml), ammonium formate (2.7 g, 43.70 mmoles) and palladium on carbon (10%, 100 mg) were added and the solution was refluxed for 2 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as white solid (0.43 g, % yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.24 (dd, J=7.9, 1.5 Hz, 1H), 7.68 (dt, J=7.2, 1.6 Hz, 1H), 7.46-7.35 (m, 5H), 7.27 (dd, J=7.2, 1.5 Hz, 2H), 2.57 (t, J=7.6 Hz, 2H), 1.78 (m, 2H0, 0.93 (t, J=7.4 Hz, 3H). Intermediate 32: 2-(1-Bromopropyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 31 (0.900 g, 3.40 mmoles) in carbon tetrachloride (15 ml) N-bromosuccinimide (0.606 g, 3.40 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (9 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as yellow solid (0.880 g, 75% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.24 (dd, J=8.0, 1.6 Hz, 1H), 7.74 (dt, J=7.2, 1.7 Hz, 1H), 7.55 (d, J=8.3 Hz, 1H), 7.49-7.20 (m, 6H), 4.71 (t, J=7.6 Hz, 1H), 2.33 (m, 2H), 0.97 (d, J=7.4 Hz, 3H). Intermediate 33: 1-(5-Bromo-2-hydroxyphenyl)-2-(3-fluorophenyl)ethanone 3-Fluorophenylacetic acid (4.90 g, 32.07 mmoles) was dissolved in 50 ml dichloromethane. To this mixture, oxalylchloride (3.5 ml, 40.08 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 50 ml dichloromethane. To this mixture, 4-bromoanisole (5.0 g, 26.72 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(5.3 g, 40.08 mmoles) was added and the reaction mixture was warmed to RT and stirred for 12 h. The reaction mixture was quenched by the addition of 2N HCl, extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (6.6 g, 80% yield.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 12.02 (s, 1H), 7.94 (d, J=2.4 Hz, 1H), 7.57 (dd, J=8.9, 2.4.1 Hz, 1H), 7.36 (m, 1H), 7.04 (m, 3H), 6.90 (d, J=8.9 Hz, 1H), 4.28 (s, 2H). Intermediate 34: 6-Bromo-2-ethyl-3-(3-fluorophenyl)-4H-chromen-4-one Intermediate 33 (3.0 g, 9.70 mmoles) was taken in a RB flask and to this triethylamine (30 ml) and propionic anhydride (3.94 g, 30.37 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (1.30 g, 39% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.10 (d, J=2.3 Hz, 1H), 7.99 (dd, J=8.9, 2.4 Hz, 1H), 7.69 (d, J=8.9 Hz, 1H), 7.51 (q, J=7.9 Hz, 1H), 7.25 (dt, J=10.8, 2.4 Hz, 1H), 7.15 (t, J=12.2 Hz, 2H), 2.57 (q, J=7.6 Hz, 2H), 1.20 (t, J=7.5 Hz, 3H). Intermediate 35: 2-Ethyl-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 34 (1.0 g, 2.88 mmoles) in ethanol (10 ml), ammonium formate (1.81 g, 28.80 mmoles) and palladium on carbon (10%, 80 mg) were added and the solution was refluxed for 2 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the crude title compound as colourless oil (0.792 g).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.05 (dd, J=7.9, 1.3 Hz, 1H), 7.83 (dt, J=8.6, 1.6 Hz, 1H), 7.67 (d, J=8.3 Hz, 1H), 7.50 (m, 2H), 7.24 (dt, J=8.8, 2.5 Hz, 1H), 7.15 (t, J=12.3 Hz, 2H), 2.55 (q, J=7.6 Hz, 2H), 1.20 (t, J=7.6 Hz, 3H). Intermediate 36: 2-(1-Bromoethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 35 (0.700 g, 2.60 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.464 g, 2.60 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (10 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (0.820 g, 91% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.06 (dd, J=7.9, 1.1 Hz, 1H), 7.89 (dt, J=8.4, 1.3 Hz, 1H), 7.77 (d, J=8.3 Hz, 1H), 7.56 (d, J=7.5 Hz, 1H), 7.52 (d, J=7.7 Hz, 1H), 7.31 (dt, J=8.6, 2.1 Hz, 1H), 7.19 (t, J=9.0 Hz, 2H), 5.02 (q, J=6.8 Hz, 1H), 1.97 (d, J=6.8 Hz, 3H). Intermediate 37: 3-(2-Fluorophenyl)-2-methyl-4H-chromen-4-one To a solution of intermediate 26 (0.5 g, 1.50 mmoles) in ethanol (5 ml), ammonium formate (0.945 g, 15.0 mmoles) and palladium on carbon (10%, 40 mg) were added and the solution was refluxed for 2 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as white solid (0.302 g, 79% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.05 (dd, J=7.9, 1.5 Hz, 1H), 7.84 (m, 1H), 7.67 (d, J=8.3 Hz, 1H), 7.51 (m, 2H), 7.37 (dt, J=7.3, 1.7 Hz, 1H), 7.29 (m, 2H), 2.26 (s, 3H). Intermediate 38: 2-(Bromomethyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 37 (0.300 g, 1.17 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.210 g, 1.17 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (15 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (0.281 g, 71% yield). Intermediate 39: 2-Ethyl-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 24 (0.770 g, 2.21 mmoles) in ethanol (10 ml), ammonium formate (1.39 g, 22.18 mmoles) and palladium on carbon (10%, 60 mg) were added and the solution was refluxed for 2 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as white solid (0.560 g, 94% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.05 (dd, J=7.9, 1.5 Hz, 1H), 7.85 (dt, J=7.3, 1.7 Hz, 1H), 7.69 (d, J=8.3 Hz, 1H), 7.52 (m, 2H), 7.36 (m, 2H), 2.52 (m, 2H), 1.19 (t, J=7.5 Hz, 3H). Intermediate 40: 2-(1-Bromoethyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 39 (0.600 g, 2.27 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.404 g, 2.27 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (15 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (0.420 g, 53% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.07 (dd, J=7.9, 1.3 Hz, 1H), 7.92 (dt, J=8.4, 1.3 Hz, 1H), 7.79 (d, J=8.4 Hz, 1H), 7.56 (m, 2H), 7.41 (m, 3H), [4.99 (q, J=6.8 Hz), 4.93 (q, J=6.7 Hz), 1H], [2.00 (d, J=6.8 Hz), 1.95 (d, J=6.8 Hz), 3H]. Intermediate 41: 6-Bromo-3-(2-fluorophenyl)-2-propyl-4H-chromen-4-one Intermediate 23 (2.0 g, 6.46 mmoles) was taken in a RB flask and to this triethylamine (20 ml) and butyric anhydride (3.19 g, 20.25 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless liquid (1.60 g, 69% yield). Intermediate 42: 3-(2-Fluorophenyl)-2-propyl-4H-chromen-4-one To a solution of intermediate 41 (1.60 g, 4.43 mmoles) in ethanol (15 ml), ammonium formate (2.79 g, 63.03 mmoles) and palladium on carbon (10%, 130 mg) were added and the solution was refluxed for 2 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as brown liquid (1.0 g, 81% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.05 (dd, J=7.9, 1.4 Hz, 1H), 7.84 (dt, J=8.5, 1.5 Hz, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.51 (q, J=7.7 Hz, 2H), 7.34 (m, 3H), 2.49 (m, 2H), 1.68 (q, J=7.4 Hz, 2H), 1.17 (t, J=7.4 Hz, 3H) Intermediate 43: 2-(1-Bromopropyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 42 (1.00 g, 3.59 mmoles) in carbon tetrachloride (20 ml) N-bromosuccinimide (0.639 g, 3.59 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (15 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (0.700 g, 54% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.07 (d, J=7.9 Hz, 1H), 7.91 (t, J=7.9 Hz, 1H), 7.78 (dd, J=8.3, 2.0 Hz, 1H), 7.56 (t, J=7.6 Hz, 2H), 7.36 (m, 3H), [4.69 (t, J=7.6 Hz), 4.64 (t, J=7.5 Hz), 1H], 2.38 (m, 2H), [0.97 (t, J=7.3 Hz), 0.88 (t, J=7.2 Hz), 3H]. Intermediate 44: 6-Bromo-3-(3-fluorophenyl)-2-propyl-4H-chromen-4-one Intermediate 33 (3.0 g, 9.70 mmoles) was taken in a RB flask and to this triethylamine (3 ml) and butyric anhydride (4.55 g, 30.37 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless liquid (0.794 g, 23% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.10 (d, J=2.5 Hz, 1H), 7.98 (dd, J=8.9, 2.5 Hz, 1H), 7.69 (d, J=8.9 Hz, 1H), 7.51 (q, J=8.0 Hz, 1H), 7.26 (dt, J=8.7, 2.5 Hz, 1H), 7.14 (dt, J=9.9, 2.3 Hz, 2H), 2.55 (m, 2H), 1.68 (q, J=7.5 Hz, 2H), 0.85 (t, J=7.5 Hz, 3H). Intermediate 45: 3-(3-Fluorophenyl)-2-propyl-4H-chromen-4-one To a solution of intermediate 44 (0.750 g, 2.07 mmoles) in ethanol (10 ml), ammonium formate (1.30 g, 20.76 mmoles) and palladium on carbon (10%, 80 mg) were added and the solution was refluxed for 2 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as colourless liquid (0.51 g, 87% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.05 (dd, J=8.0, 1.3 Hz, 1H), 7.83 (dt, J=8.4, 1.3 Hz, 1H), 7.66 (d, J=8.4 Hz, 1H), 7.51 (m, 2H), 7.24 (dt, J=8.9, 2.5 Hz, 1H), 7.14 (t, J=8.1 Hz, 2H), 2.53 (m, 2H), 1.69 (m, 2H), 0.85 (t, J=7.3 Hz, 3H). Intermediate 46: 2-(1-Bromopropyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 45 (0.48 g, 1.70 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.302 g, 1.70 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (10 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (0.540 g, 88% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.07 (dd, J=7.9, 1.5 Hz, 1H), 7.89 (dt, J=8.5, 1.5 Hz, 1H), 7.75 (d, J=8.4 Hz, 1H), 7.57 (q, J=8.0 Hz, 2H), 7.32 (dt, J=8.6, 2.5 Hz, 1H), 7.17 (dt, J=8.4, 2.3 Hz, 2H), 4.70 (t, J=7.5 Hz, 1H), 2.34 (m, 1H), 2.20 (m, 1H), 0.92 (t, J=7.2 Hz, 3H). Intermediate 47: 6-Bromo-3-(4-fluorophenyl)-2-propyl-4H-chromen-4-one Intermediate 6 (3.0 g, 9.70 mmoles) was taken in a RB flask and to this triethylamine (30 ml) and butyric anhydride (4.55 g, 30.37 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless liquid (2.55 g, 71% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.33 (d, J=2.3 Hz, 1H), 7.76 (dd, J=8.8, 2.3 Hz, 1H), 7.36 (d, J=8.9 Hz, 1H), 7.23 (dd, J=8.7, 5.6 Hz, 2H), 7.15 (t, J=8.7 Hz, 2H), 2.55 (t, J=7.5 Hz, 2H), 1.77 (m, 2H), 0.93 (t, J=7.4 Hz, 3H). Intermediate 48: 3-(4-Fuorophenyl)-2-propyl-4H-chromen-4-one To a solution of intermediate 47 (1.00 g, 2.76 mmoles) in ethanol (10 ml), ammonium formate (1.70 g, 27.60 mmoles) and palladium on carbon (10%, 80 mg) were added and the solution was refluxed for 1 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as colourless liquid (0.750 g, 96% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.23 (dd, J=7.9, 1.4 Hz, 1H), 7.69 (dt, J=8.5, 1.5 Hz, 1H), 7.47 (d, J=8.4 Hz, 1H), 7.41 (t, J=7.8 Hz, 1H), 7.25 (m, 2H), 7.15 (t, J=8.7 Hz, 2H), 2.56 (t, J=7.5 Hz, 2H), 1.78 (m, 2H), 0.94 (t, J=7.3 Hz, 3H). Intermediate 49: 2-(1-Bromopropyl)-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 48 (0.700 g, 2.47 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.441 g, 2.47 mmoles) was added and heated to 80° C. Azobisisobutyronitrile 7 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (1.1 g).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.23 (dd, J=8.0, 1.2 Hz, 1H), 7.74 (dt, J=8.4, 1.3 Hz, 1H), 7.55 (d, J=8.4 Hz, 1H), 7.45 (t, J=7.4 Hz, 1H), 7.35 (m, 2H), 7.19 (t, J=8.7 Hz, 2H), 4.68 (t, J=7.7 Hz, 1H), 2.31 (m, 2H), 0.97 (t, J=7.3 Hz, 3H). Intermediate 50: 1-(5-Fluoro-2-hydroxyphenyl)-2-phenylethanone Phenylacetic acid (8.09 g, 59.46 mmoles) was dissolved in 15 ml dichloromethane. To this mixture, oxalylchloride (5.2 ml, 59.46 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 15 ml dichloromethane. To this mixture, 4-fluoroanisole (5.0 g, 39.64 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(7.92 g, 59.46 mmoles) was added and the reaction mixture was warmed to RT and stirred for 12 h. The reaction mixture was quenched by the addition of 2N HCl, extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (5.1 g, 56% yield.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 11.43 (s, 1H), 7.77 (dd, J=9.5, 3.2 Hz, 1H), 7.42 (dt, J=8.7, 3.2 Hz, 1H), 7.33 (t, J=7.3 Hz, 2H), 7.26 (m, 3H), 7.01 (q, J=4.6 Hz, 1H), 4.42 (s, 2H). Intermediate 51: 6-Fluoro-3-phenyl-2-propyl-4H-chromen-4-one Intermediate 50 (1.6 g, 6.94 mmoles) was taken in a RB flask and to this triethylamine (16 ml) and butyric anhydride (3.43 g, 21.72 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless liquid (1.40 g, 71% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.79 (dd, J=10.2, 4.3 Hz, 1H), 7.73 (dt, J=6.4, 3.1 Hz, 2H), 7.46 (t, J=6.9 Hz, 2H), 7.42 (m, 1H), 7.26 (dd, J=8.3, 1.5 Hz, 2H), 2.52 (t, J=7.4 Hz, 2H), 1.70 (m, 2H), 0.84 (t, J=7.4 Hz, 3H). Intermediate 52: 2-(1-Bromopropyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of intermediate 51 (1.30 g, 4.60 mmoles) in carbon tetrachloride (20 ml) N-bromosuccinimide (0.818 g, 4.60 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (10 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (1.40 g, 84% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.88 (dd, J=9.2, 4.3 Hz, 1H), 7.80-7.71 (m, 2H), 7.52-7.42 (m, 3H), 7.29 (d, J=6.8 Hz, 2H), 4.68 (t, J=7.6 Hz, 1H), 2.34-2.15 (m, 2H), 0.91 (t, J=7.3 Hz, 3H). Intermediate 53: 6-Bromo-2-ethyl-3-(4-fluorophenyl)-4H-chromen-4-one Intermediate 6 (3.0 g, 9.70 mmoles) was taken in a RB flask and to this triethylamine (30 ml) and propionic anhydride (3.94 g, 30.37 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (1.60 g, 47% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.33 (d, J=2.4 Hz, 1H), 7.76 (dd, J=8.8, 2.4 Hz, 1H), 7.38 (d, J=8.8 Hz, 1H), 7.24 (dd, J=5.5, 2.0 Hz, 2H), 7.16 (dt, J=11.4, 2.8 Hz, 2H), 2.61 (q, J=7.6 Hz, 2H), 1.27 (t, J=7.5 Hz, 3H). Intermediate 54: 2-Ethyl-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 53 (1.00 g, 2.88 mmoles) in ethanol (10 ml), ammonium formate (1.70 g, 27.60 mmoles) and palladium on carbon (10%, 80 mg) were added and the solution was refluxed for 1 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as colourless liquid (0.640 g, 83% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.24 (dd, J=8.0, 1.5 Hz, 1H), 7.69 (dt, J=8.6, 1.7 Hz, 1H), 7.48 (d, J=8.3 Hz, 1H), 7.41 (t, J=7.9 Hz, 1H), 7.24 (m, 2H), 7.15 (t, J=8.7 Hz, 1H), 2.62 (q, J=7.6 Hz, 2H), 1.28 (t, J=7.6 Hz, 3H). Intermediate 55: 2-(1-Bromoethyl)-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 54 (0.600 g, 2.23 mmoles) in carbon tetrachloride (15 ml) N-bromosuccinimide (0.398 g, 2.23 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (10 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (1.10 g) which is taken as such for next step. Intermediate 56: 2-Ethyl-6-fluoro-3-phenyl-4H-chromen-4-one Intermediate 50 (3.0 g, 13.63 mmoles) was taken in a RB flask and to this triethylamine (30 ml) and propionic anhydride (5.30 g, 40.78 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (2.27 g, 65% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.79 (dd, J=7.1, 4.4 Hz, 1H), 7.73 (dt, J=7.7, 3.1 Hz, 2H), 7.46 (t, J=8.2 Hz, 2H), 7.40 (m, 1H), 7.27 (dd, J=8.2, 1.4 Hz, 2H), 2.55 (q, J=7.5 Hz, 2H), 1.19 (t, J=7.6 Hz, 3H). Intermediate 57: 2-(1-Bromoethyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of intermediate 56 (1.0 g, 3.72 mmoles) in carbon tetrachloride (20 ml) N-bromosuccinimide (0.662 g, 3.72 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (10 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (1.37 g).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.89 (dd, J=9.2, 4.3 Hz, 1H), 7.79 (dt, J=8.3, 3.2 Hz, 2H), 7.73 (dd, J=8.3.3.1 Hz, 2H), 7.51-7.42 (m, 3H), 7.32 (d, J=6.6 Hz, 2H), 4.97 (q, J=6.8 Hz, 1H), 1.96 (d, J=6.8 Hz, 3H). Intermediate 58: 3-(3-Methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-Iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (0.522 g, 2.0 mmoles) in DMF (10 ml), ethanol (5 ml) and water (5 ml), 3-methoxyphenylboronic acid (0.395 g, 2.59 mmoles) and sodium carbonate (1.05 g, 10 mmoles) were added and the system is degassed for 30 min Palladium tetrakis triphenylphosphine (0.455 g, 0.39 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture neutralised with 1.5N HCl, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.130 g, 27% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.57 (s, 1H), 8.20 (s, 1H), 7.46 (t, J=7.8 Hz, 1H), 7.23 (d, J=7.5 Hz, 1H), 7.18 (d, J=2.4 Hz, 1H), 7.04 (dd, J=8.0, 1.8 Hz, 1H), 3.81 (s, 3H). Intermediate 59: 1-(2-Hydroxyphenyl)-2-phenylethanone To a solution of intermediate 1 (1.00 g, 3.43 mmoles) in ethanol (10 ml), ammonium formate (2.16 g, 34.34 mmoles) and palladium on carbon (10%, 100 mg) were added and the solution was refluxed for 1 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as colourless liquid (0.560 g, 77% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 11.80 (s, 1H), 8.02 (dd, J=5.7, 1.7 Hz, 1H), 7.54 (dt, J=8.6, 1.7 Hz, 1H), 7.33 (m, 5H), 6.98 (m, 2H), 4.43 (s, 2H). Intermediate 60: 6-Bromo-2-ethyl-3-o-tolyl-4H-chromen-4-one Intermediate 11 (3.0 g, 9.83 mmoles) was taken in a RB flask and to this triethylamine (25 ml) and propionic anhydride (4.00 g, 30.76 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless liquid (0.700 g, 20% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.73 (d, J=2.5 Hz, 1H), 7.63 (dd, J=8.7, 2.5 Hz, 1H), 7.34 (t, J=4.8 Hz, 1H), 7.22-7.14 (m, 4H), 2.63 (q, J=7.5 Hz, 2H), 0.94 (t, J=7.5 Hz, 3H). Intermediate 61: 2-Ethyl-3-o-tolyl-4H-chromen-4-one To a solution of intermediate 60 (0.950 g, 2.76 mmoles) in ethanol (15 ml), ammonium formate (1.73 g, 27.60 mmoles) and palladium on carbon (10%, 80 mg) were added and the solution was refluxed for 1 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as colourless liquid (0.620 g, 85% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.05 (dd, J=7.9, 1.3 Hz, 1H), 7.83 (dt, J=8.5, 1.5 Hz, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.50 (t, J=7.5 Hz, 1H), 7.30 (d, J=4.3 Hz, 2H), 7.26 (m, 1H), 7.13 (d, J=7.2 Hz, 1H), 2.46 (m, 2H), 1.15 (t, J=7.6 Hz, 3H). Intermediate 62: 2-(2-Fluorophenyl)-1-(2-hydroxyphenyl)ethanone To a solution of intermediate 23 (9.0 g, 29.13 mmoles) in ethanol (90 ml), ammonium formate (18.3 g, 291.13 mmoles) and palladium on carbon (10%, 0.50 g) were added and the solution was refluxed for 1 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless solid (3.5 g, 52% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 12.08 (s, 1H), 7.90 (d, J=7.0 Hz, 1H), 7.51 (dt, J=7.2, 1.4 Hz, 1H), 7.31-7.23 (m, 2H), 7.15-7.08 (m, 2H), 7.01 (d, J=8.4 Hz, 1H), 6.96 (t, J=8.0 Hz, 1H), 4.36 (s, 2H). Intermediate 63: tert-butyl(3-(2-fluorophenyl)-4-oxo-4H-chromen-2-yl) methyl carbamate To a solution of intermediate 62 (2 g, 8.68 mmoles) in dichloromethane (20 ml), triethylamine (2.6 g, 26.06 mmoles) was added followed by N-Boc-Glycine (1.8 g, 10.27 mmoles). To this mixture HATU (6.6 g, 17.37 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as yellow solid (0.72 g, 23% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.06 (d, J=6.7 Hz, 1H), 7.87 (dt, J=7.0, 1.6 Hz, 1H), 7.62 (d, J=8.5 Hz, 1H), 7.53 (t, J=7.4 Hz, 1H), 7.48-7.35 (m, 3H), 7.30 (m, 2H), 4.04 (d, J=5.9 Hz, 2H), 1.36 (s, 9H). Intermediate 64: 2-(Amino methyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 63 (0.700 g, 1.89 mmoles) in dichloromethane (10 ml), trifluoroacetic acid (3 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as brown liquid (0.440 g, 86%).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.06 (d, J=7.9 Hz, 1H), 7.87 (dt, J=8.5, 1.3 Hz, 1H), 7.70 (d, J=8.4 Hz, 1H), 7.52 (m, 2H), 7.40 (t, J=7.2 Hz, 1H), 7.31 (m, 2H), 3.51 (s, 2H). Intermediate 65: 2-(3-Fluorophenyl)-1-(2-hydroxyphenyl)ethanone To a solution of intermediate 33 (11.0 g, 35.58 mmoles) in ethanol (110 ml), ammonium formate (22.4 g, 355.83 mmoles) and palladium on carbon (10%, 0.550 g) were added and the solution was refluxed for 1 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless solid (5.6 g, 70% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 11.68 (s, 1H), 8.00 (dd, J=8.3, 1.6 Hz, 1H), 7.54 (dt, J=8.5, 1.6 Hz, 1H), 7.38 (m, 1H), 7.14-7.04 (m, 3H), 6.99 (m, 2H), 4.48 (s, 2H). Intermediate 66: 1-(5-Fluoro-2-hydroxyphenyl)-2-(2-fluorophenyl)ethanone 2-Fluorophenylacetic acid (2.0 g, 13.14 mmoles) was dissolved in 20 ml dichloromethane. To this mixture, oxalylchloride (1.66 g, 13.14 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 20 ml dichloromethane. To this mixture, 4-fluoroanisole (1.10 g, 8.76 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(1.75 g, 13.14 mmoles) was added and the reaction mixture was warmed to RT and stirred for 12 h. The reaction mixture was quenched by the addition of 2N HCl, extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (1.17 g, 54% yield.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 11.25 (s, 1H), 7.73 (dd, J=9.5, 3.2 Hz, 1H), 7.43 (dt, J=8.8, 3.1 Hz, 1H), 7.35 (d, J=6.2 Hz, 1H), 7.31 (d, J=7.2 Hz, 1H), 7.19 (d, J=8.2 Hz, 1H), 7.03 (dd, J=9.1, 4.6 Hz, 1H), 4.50 (s, 2H). Intermediate 67: 2-Ethyl-6-fluoro-3-(2-fluorophenyl)-4H-chromen-4-one Intermediate 66 (1.1 g, 4.43 mmoles) was taken in a RB flask and to this triethylamine (10 ml) and propionic anhydride (1.80 g, 13.86 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.800 g, 63% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.82 (dd, J=9.0, 4.4 Hz, 1H), 7.75 (m, 2H), 7.50 (m, 1H), 7.37-7.28 (m, 3H), 2.56 (m, 2H), 1.19 (t, J=7.6 Hz, 3H). Intermediate 68: 2-(1-bromoethyl)-6-fluoro-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 67 (0.790 g, 2.75 mmoles) in carbon tetrachloride (15 ml) N-bromosuccinimide (0.491 g, 2.75 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (10 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as yellow solid (0.824 g).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ [7.93 (d, J=4.3 Hz) 7.91 (d, J=4.2 Hz), 1H], 7.83 (dt, J=8.2, 3.1 Hz, 1H), 7.75 (m, 1H), 7.56 (m, 1H), 7.41 (m, 3H), [5.00 (q, J=6.9 Hz), 4.93 (q, J=6.9 Hz, 1H], [1.99 (d, J=6.9 Hz), 1.95 (d, J=6.8 Hz), 3H). Intermediate 69: 1-(5-bromo-2-hydroxyphenyl)-2-(3,5-difluorophenyl)ethanone 3,5-Difluorophenylacetic acid (5.0 g, 29.0 mmoles) was dissolved in 50 ml dichloromethane. To this mixture, oxalylchloride (3.8 ml, 43.57 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 50 ml dichloromethane. To this mixture, 4-bromooanisole (5.42 g, 29.0 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(5.80 g, 47.57 mmoles) was added and the reaction mixture was warmed to RT and stirred for 12 h. The reaction mixture was quenched by the addition of 2N HCl, extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (7.21 g, 77% yield.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 11.44 (s, 1H), 7.98 (d, J=2.5 Hz, 1H), 7.65 (dd, J=8.9, 2.6 Hz, 1H), 7.13 (tt, J=9.1, 2.4 Hz, 1H), 7.02 (m, 3H), 4.50 (s, 2H). Intermediate 70: 2-(3,5-Difluorophenyl)-1-(2-hydroxyphenyl)ethanone To a solution of intermediate 69 (7.20 g, 22.01 mmoles) in ethanol (70 ml), ammonium formate (13.8 g, 220.17 mmoles) and palladium on carbon (10%, 0.250 g) were added and the solution was refluxed for 1 h. The solution was filtered through celite, diluted with ethyl acetate, washed with 10% sodium bicarbonate solution (100 ml), dried over sodium sulphate and concentrated to afford the title compound as yellow solid (4.1 g, 76% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 11.58 (s, 1H), 7.97 (dd, J=8.3, 1.6 Hz, 1H), 7.55 (dt, J=8.5, 1.5 Hz, 1H), 7.14 (tt, J=7.5, 2.2 Hz, 1H), 7.03-6.96 (m, 4H), 4.52 (s, 2H). Intermediate 71: 3-(3,5-Difluorophenyl)-2-ethyl-4H-chromen-4-one Intermediate 70 (2.0 g, 8.08 mmoles) was taken in a RB flask and to this triethylamine (20 ml) and propionic anhydride (3.26 g, 25.2 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-colourless liquid (1.65 g, 72% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.05 (dd, J=7.9, 1.2 Hz, 1H), 7.84 (dt, J=8.5, 1.4 Hz, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.51 (t, J=7.8 Hz, 1H), 7.30 (tt, J=7.2, 2.2 Hz, 1H), 7.07 (d, J=6.1 Hz, 2H), 2.58 (q, J=7.5 Hz, 1H), 1.21 (t, J=7.6 Hz, 3H). Intermediate 72: 2-(1-Bromoethyl)-3-(3,5-difluorophenyl)-4H-chromen-4-one To a solution of intermediate 71 (1.60 g, 5.58 mmoles) in carbon tetrachloride (20 ml) N-bromosuccinimide (0.994 g, 5.58 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (30 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as brown solid (1.95 g, 96% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.06 (dd, J=7.9, 1.5 Hz, 1H), 7.90 (dt, J=8.6, 1.6 Hz, 1H), 7.78 (d, J=8.3 Hz, 1H), 7.55 (t, J=7.3 Hz, 1H), 7.38 (tt, J=9.5, 2.3 Hz, 1H), 7.10 (dd, J=8.3, 2.2 Hz, 2H), 5.05 (q, J=6.8 Hz, 1H), 1.97 (d, J=6.8 Hz, 3H). Intermediate 73: 1-(5-Fluoro-2-hydroxyphenyl)-2-(3-fluorophenyl)ethanone 3-Fluorophenylacetic acid (7.33 g, 47.56 mmoles) was dissolved in 25 ml dichloromethane. To this mixture, oxalylchloride (7.54 g, 59.46 mmoles) and DMF (3 drops) were added at 0° C. and stirred for 30 min. The solvent was evaporated and dissolved in 25 ml dichloromethane. To this mixture, 4-fluoroanisole (5.00 g, 39.64 mmoles) was added and cooled to 0° C. At 0° C. AlCl3(7.95 g, 59.46 mmoles) was added and the reaction mixture was warmed to RT and stirred for 12 h. The reaction mixture was quenched by the addition of 2N HCl, extracted with ethyl acetate, dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless solid (4.5 g, 45% yield.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 11.34 (s, 1H), 7.75 (dd, J=9.4, 3.1 Hz, 1H), 7.42 (m, 2H), 7.12 (m, 3H), 7.05 (dd, J=9.0, 4.5 Hz, 1H), 4.47 (s, 2H). Intermediate 74: 2-Ethyl-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one Intermediate 73 (3.00 g, 12.08 mmoles) was taken in a RB flask and to this triethylamine (25 ml) and propionic anhydride (4.92 g, 37.82 mmoles) were added and the mixture was refluxed for 24 h. After cooling to RT, the reaction mixture was acidified by the addition of 1N HCl solution, extracted with ethyl acetate, washed with sodium bicarbonate solution, dried with sodium sulphate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-yellow solid (1.80 g, 52% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.80 (m, 1H), 7.76 (m, 2H), 7.51 (dd, J=8.0, 6.4 Hz), 7.22 (m, 1H), 7.18 (m, 2H), 2.56 (q, J=7.6 Hz, 2H), 1.20 (t, J=7.6 Hz, 3H). Intermediate 75: 2-(1-Bromoethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 74 (1.80 g, 6.28 mmoles) in carbon tetrachloride (20 ml) N-bromosuccinimide (1.11 g, 6.28 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (10 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as yellow solid (1.25 g, 55% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.91 (dd, J=9.2, 4.3 Hz, 1H), 7.81 (dt, J=8.2, 2.8 Hz, 1H), 7.74 (dd, J=8.3, 3.1 Hz, 1H), 7.57 (m, 1H), 7.32 (dt, J=8.5, 2.4 Hz, 1H), 7.19 (m, 2H), 5.00 (q, J=6.8 Hz, 1H), 1.97 (d, J=6.8 Hz, 3H). Intermediate 76: 3-(3-fluorophenyl)-2-methyl-4H-chromen-4-one Intermediate 65 (1.50 g, 6.51 mmoles) was taken in a RB flask and to this acetic anhydride (15 ml) and sodium acetate (3.74 g, 45.60 mmoles) were added and the mixture was refluxed for 12 h. After cooling to RT, the reaction mixture was quenched by the addition of ice cold water. The solid formed was filtered and washed with water. The product was dried under vacuum to afford the title compound as colourless solid (1.1 g, 68% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.05 (dd, J=7.9, 1.6 Hz, 1H), 7.83 (m, 1H), 7.66 (d, J=8.1 Hz, 1H), 7.50 (m, 2H), 7.24-7.13 (m, 3H), 2.29 (s, 3H). Intermediate 77: 2-(bromomethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 76 (1.00 g, 3.99 mmoles) in carbon tetrachloride (10 ml) N-bromosuccinimide (0.711 g, 3.99 mmoles) was added and heated to 80° C. Azobisisobutyronitrile (10 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as off-white solid (0.990 g, 74% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.07 (dd, J=8.1, 1.6 Hz, 1H), 7.89 (m, 1H), 7.73 (d, J=8.3 Hz, 1H), 7.56 (m, 2H), 7.32 (dt, J=8.4, 2.3 Hz, 1H), 7.23 (m, 2H), 4.40 (s, 2H). Intermediate 78: 3-(3-fluorophenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.50 g, 5.74 mmoles) in DMF (12 ml), ethanol (7 ml) and water (7 ml), 3-Fluorophenyl boroinc acid (1.6 g, 11.49 mmoles) and sodium carbonate (3.0 g, 28.73 mmoles) were added and the system is degassed for 30 min. Palladium tetrakis triphenylphosphine (1.90 g, 1.72 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture neutralised with 1.5N HCl, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.240 g, 18% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.66 (s, 1H), 8.21 (s, 1H), 7.59 (m, 1H), 7.50 (d, J=7.6, 1.2 Hz, 1H), 7.45 (m, 1H), 7.31 (m, 1H). Intermediate 79: 3-(3-fluoro-5-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (0.700 g, 2.68 mmoles) in DMF (10 ml), ethanol (5 ml) and water (5 ml), 3-Fluoro-5-methoxyphenyl boroinc acid (0.592 g, 3.48 mmoles) and sodium carbonate (1.42 g, 13.40 mmoles) were added and the system is degassed for 30 min Palladium tetrakis triphenylphosphine (0.588 g, 0.509 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.260 g, 37% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.64 (s, 1H), 8.21 (s, 1H), 7.03 (m, 2H), 6.93 (td, J=11.1, 2.3 Hz, 1H), 3.83 (s, 3H). Intermediate 80: 3-(4-Fluoro-3-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (0.500 g, 1.91 mmoles) in DMF (8 ml), ethanol (4 ml) and water (4 ml), 4-Fluoro-3-methoxyphenyl boroinc acid (0.423 g, 2.49 mmoles) and sodium carbonate (1.01 g, 9.57 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.436 g, 0.377 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.240 g, 48% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.64 (s, 1H), 8.20 (s, 1H), 8.08 (s, 1H), 7.54 (d, J=9.3 Hz, 1H), 7.34 (d, J=8.3 Hz, 1H), 3.82 (s, 3H). Intermediate 81: 3-(3-fluoro-4-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.00 g, 3.83 mmoles) in DMF (12 ml), ethanol (7 ml) and water (7 ml), 3-Fluoro-4-methoxyphenyl boroinc acid (0.781 g, 4.59 mmoles) and sodium carbonate (2.03 g, 19.15 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.872 g, 0.754 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.136 g, 14% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.53 (s, 1H), 8.19 (s, 1H), 7.45 (m, 2H), 7.33 (t, J=8.6 Hz, 1H), 3.89 (s, 3H). Intermediate 82: 3-(3-chloro-5-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (0.700 g, 2.68 mmoles) in DMF (10 ml), ethanol (6 ml) and water (6 ml), 3-chloro-5-methoxyphenyl boroinc acid (0.600 g, 3.21 mmoles) and sodium carbonate (1.40, 13.40 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.610 g, 0.528 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.198 g, 27% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.66 (s, 1H), 8.21 (s, 1H), 7.24 (t, J=1.6 Hz, 1H), 7.13 (d, J=1.2 Hz, 1H), 7.11 (t, J=2.1 Hz, 1H), 3.83 (s, 3H). Intermediate 83: 3-(3-(trifluoromethoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.00 g, 3.83 mmoles) in DMF (14 ml), ethanol (7 ml) and water (7 ml), 3-trifluoromethoxyphenyl boroinc acid (1.025 g, 4.97 mmoles) and sodium carbonate (2.02 g, 19.15 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.871 g, 0.754 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.465 g, 41% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.71 (s, 1H), 8.22 (s, 1H), 7.70 (m, 2H), 7.59 (s, 1H), 7.46 (td, J=7.9, 1.4 Hz, 1H). Intermediate 84: 3-(4-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.00 g, 3.83 mmoles) in DMF (14 ml), ethanol (7 ml) and water (7 ml), 4-methoxyphenyl boroinc acid (0.873 g, 5.746 mmoles) and sodium carbonate (2.03 g, 19.15 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.871 g, 0.754 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.250 g, 27% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.46 (s, 1H), 8.19 (s, 1H), 7.59 (td, J=9.5, 2.8 Hz, 2H), 7.11 (td, J=11.6, 2.6, 2H), 3.81 (s, 3H). Intermediate 85: 3-(4-fluoro-2-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.00 g, 3.83 mmoles) in DMF (14 ml), ethanol (7 ml) and water (7 ml), 4-fluoro-2-methoxyphenyl boroinc acid (0.846 g, 4.979 mmoles) and sodium carbonate (2.06 g, 19.15 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.754 g, 0.652 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.350 g, 35% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.46 (s, 1H), 8.14 (s, 1H), 7.40 (t, J=8.4 Hz, 1H), 7.09 (dd, J=11.5, 2.9 Hz, 1H), 6.91 (dt, J=8.4, 2.4 Hz 1H), 3.78 (s, 3H). Intermediate 86: 3-(4-chloro-3-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (0.430 g, 1.65 mmoles) in DMF (3.6 ml), ethanol (1.8 ml) and water (1.8 ml), 4-chloro-3-methoxyphenyl boroinc acid (0.400 g, 2.145 mmoles) and sodium carbonate (0.873 g, 19.15 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.374 g, 0.313 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as green solid (0.060 g, 10% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.62 (s, 1H), 8.20 (s, 1H), 7.56 (d, J=8.1 Hz, 1H), 7.34 (s, 1H), 7.23 (d, J=8.1 Hz 1H), 3.91 (s, 3H). Intermediate 87: 3-(2-chloro-5-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.0770 g, 4.12 mmoles) in DMF (10 ml), ethanol (5 ml) and water (5 ml), 2-chloro-5-methoxyphenyl boroinc acid (1.00 g, 5.364 mmoles) and sodium carbonate (2.186 g, 20.63 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.905 g, 0.783 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as green solid (0.090 g, 16% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.61 (s, 1H), 8.19 (s, 1H), 7.51 (d, J=8.9 Hz, 1H), 7.09 (d, J=8.9 Hz 1H), 7.06 (d, J=2.6 Hz 1H), 3.78 (s, 3H). Intermediate 88: 3-(3,4-dimethoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.00 g, 3.83 mmoles) in DMF (10 ml), ethanol (5 ml) and water (5 ml), 3,4-dimethoxyphenyl boroinc acid (1.04 g, 5.746 mmoles) and sodium carbonate (2.03 g, 19.15 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.872 g, 0.754 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.220 g, 21% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.46 (s, 1H), 8.19 (s, 1H), 7.20 (s, 1H), 7.19 (d, J=9.3 Hz, 1H), 7.11 (d, J=8.1 Hz 1H), 3.81 (s, 6H). Intermediate 89: 6-fluoro-2-methyl-3-phenyl-4H-chromen-4-one Intermediate 50 (50 g, 0.217 moles) was taken in a RB flask and to this acetic anhydride (424 ml) and sodium acetate (124 g, 1.51 moles) were added and the mixture was refluxed for 12 h. After cooling to RT, the reaction mixture was quenched by the addition of ice cold water. The solid formed was filtered and washed with water. The product was dried under vacuum to afford the title compound as colourless solid (44 g, 80% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 7.87 (dd, J=8.3, 3.0 Hz, 1H), 7.47-7.35 (m, 5H), 7.29 (m, 2H), 2.32 (s, 3H). Intermediate 90: 2-(bromomethyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of intermediate 89 (44 gg, 0.16 moles) in carbon tetrachloride (400 ml) N-bromosuccinimide (29.1 g, 0.16 moles) was added and heated to 80° C. Azobisisobutyronitrile (500 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as pale yellow solid (40.2 g, 75% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.87 (dd, J=8.1, 3.0 Hz, 1H), 7.55 (dd, J=9.1, 4.2 Hz, 1H), 7.50-7.37 (m, 6H), 4.24 (s, 2H). Intermediate 91: 6-fluoro-3-(3-fluorophenyl)-2-methyl-4H-chromen-4-one Intermediate 73 (24 g, 0.096 moles) was taken in a RB flask and to this acetic anhydride (230 ml) and sodium acetate (55.2 g, 0.673 moles) were added and the mixture was refluxed for 12 h. After cooling to RT, the reaction mixture was quenched by the addition of ice cold water. The solid formed was filtered and washed with water. The product was dried under vacuum to afford the title compound as brown solid (26 g, quant. yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 7.87 (dd, J=8.2, 3.0 Hz, 1H), 7.48-7.36 (m, 3H), 7.10-6.99 (m, 3H), 2.33 (s, 3H). Intermediate 92: 2-(bromomethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 91 (39 g, 0.143 moles) in carbon tetrachloride (400 ml) N-bromosuccinimide (25.5 g, 0.143 moles) was added and heated to 80° C. Azobisisobutyronitrile (500 mg) was added to the reaction mixture at 80° C. After 12 h, the reaction mixture was cooled to RT, diluted with dichloromethane and washed with water. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the crude title compound as pale brown solid (27 g, 54% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.87 (dd, J=8.1, 3.0 Hz, 1H), 7.69 (dd, J=9.2, 5.1 Hz, 1H), 7.49 (m, 2H), 7.18-7.10 (m, 3H), 4.23 (s, 2H). Intermediate 93: 1-(4-bromo-2-fluorophenyl)ethanol To a ice-cold solution of methyl magnesium iodide prepared from magnesium (1.7 g, 73.88 mmoles) and methyl iodide (4.58 ml, 73.88 mmoles) in diethyl ether (50 ml), 4-bromo-2-fluorobenzaldehyde (5 g, 24.62 mmoles) in diethyl ether (10 ml) was added and warmed to room temperature. After 12 h, the reaction mixture was cooled to 0° C., quenched with dilute HCl and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as red colour liquid (5 g, 94% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 7.40 (t, J=8.2 Hz, 1H), 7.30 (dd, J=8.3, 1.7 Hz, 1H), 7.21 (dd, J=9.9, 1.9 Hz, 1H), 5.17 (q, J=6.4 Hz, 1H), 1.49 (d, J=6.5 Hz, 3H). Intermediate 94: 1-(4-bromo-2-fluorophenyl)ethanone To a solution of intermediate 93 (5.0 g, 22.82 mmoles) in DMF (25 ml), pyridinium dichromate (12.8 g, 34.23 mmoles) was added at room temperature. After 12 h, the reaction mixture was quenched with water, diluted with ethyl acetate. And filtered through celite. The organic layer was washed with brine solution and dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as red colour liquid (4.1 g, 84% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.76 (t, J=8.3 Hz, 1H), 7.73 (dd, J=10.8, 1.8 Hz, 1H), 7.55 (dd, J=5.2, 1.8 Hz, 1H), 2.55 (s, 3H). Intermediate 95: 6-bromo-3-methyl-1H-indazole To a solution of intermediate 94 (3.7 g, 17.04 mmoles) in 1,2-ethanediol (25 ml), hydrazine hydrate (1.65 ml, 34.09 mmoles) was added at room temperature and heated to 165° C. After 12 h, the reaction mixture cooled to room temperature, quenched with water and solid precipitated was filtered and dried under vacuum to afford the title compound as colourless solid (2.5 g, 72% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.74 (s, 1H), 7.67 (d, J=5.8 Hz, 1H), 7.65 (s, 1H), 7.19 (dd, J=8.6, 1.4 Hz, 1H), 2.46 (s, 3H). Intermediate 96: tert-butyl 6-bromo-3-methyl-1H-indazole-1-carboxylate To a solution of intermediate 95 (10.0 g, 47.39 mmoles) in acetonitrile (100 ml) cooled to 20° C., Boc-anhydride (10.3 g, 34.09 mmoles) was added followed by DMAP (0.579 g, 4.73 mmoles) and triethylamine (4.7 g, 47.39 mmoles) and the reaction mixture was stirred at room temperature. After 12 h, the reaction mixture was concentrated and quenched with water and solid precipitated was filtered and dried under vacuum to afford the title compound as colourless solid (10.3 g, 70% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.19 (d, J=1.2 Hz, 1H), 7.81 (d, J=8.4 Hz, 1H), 7.54 (dd, J=8.5, 1.7 Hz, 1H), 2.50 (s, 3H), 1.62 (s, 9H). Intermediate 97: 3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole To a solution of intermediate 95 (1.0 g, 4.73 mmoles) in Dioxan 16 ml), bis(pinacaloto)diboron (1.3 g, 5.21 mmoles) and potassium acetate (0.930 g, 9.47 mmoles) were added and the system is degassed for 30 min Bis(diphenylphosphinoferrocene)dichloro palladium·CH2Cl2(0.387 g, 0.473 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (1.1 g, 91% yield) which is used as such for the next step. Intermediate 98: tert-butyl 3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole-1-carboxylate To a solution of intermediate 96 (2.70 g, 8.67 mmoles) in Dioxan (44 ml), bis(pinacaloto)diboron (2.4 g, 9.54 mmoles) and potassium acetate (1.70 g, 17.35 mmoles) were added and the system is degassed for 30 min. Bis(diphenylphosphinoferrocene)dichloro palladium·CH2Cl2(0.354 g, 0.433 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (2.70 g, 87% yield).).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.46 (s, 1H), 7.82 (d, J=7.9 Hz, 1H), 7.61 (d, J=8.0 Hz, 1H), 2.51 (s, 3H), 1.62 (s, 9H). Intermediate 99: 1-(4-bromo-2-fluorophenyl)propan-1-ol To a ice-cold solution of ethyl magnesium iodide prepared from magnesium (2.39 g, 98.51 mmoles) and ethyl iodide (7.88 ml, 98.51 mmoles) in diethyl ether (50 ml), 4-bromo-2-fluorobenzaldehyde (5 g, 24.62 mmoles) in diethyl ether (10 ml) was added and warmed to room temperature. After 12 h, the reaction mixture was cooled to 0° C., quenched with dilute HCl and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as red colour liquid (5.8 g, 99% yield) which is used as such for step. Intermediate 100: 1-(4-bromo-2-fluorophenyl)propan-1-one To a solution of intermediate 99 (5.8 g, 24.89 mmoles) in DMF (30 ml), pyridinium dichromate (14.04 g, 37.33 mmoles) was added at room temperature. After 12 h, the reaction mixture was quenched with water, diluted with ethyl acetate and filtered through celite. The organic layer was washed with brine solution and dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as colourless liquid (4.4 g, 76% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.78 (t, J=8.1 Hz, 1H), 7.38 (m, 2H), 2.55 (m, 2H), 1.21 (t, J=7.1 Hz, 3H). Intermediate 101: 6-bromo-3-ethyl-1H-indazole To a solution of intermediate 100 (4.3 g, 18.53 mmoles) in DMSO (4.5 ml), hydrazine hydrate 17.3 ml, 357.7 mmoles) was added at room temperature and heated to 130° C. After 22 h, the reaction mixture cooled to room temperature, quenched with water and solid precipitated was filtered and dried under vacuum to afford the title compound as colourless solid (3.8 g, 91% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.73 (s, 1H), 7.70 (d, J=8.6 Hz, 1H), 7.66 (d, J=1.1 Hz, 1H), 7.18 (dd, J=8.5, 1.5 Hz, 1H), 2.92 (q, J=7.6 Hz, 2H), 1.30 (t, J=7.6 Hz, 3H). Intermediate 102: tert-butyl 6-bromo-3-ethyl-1H-indazole-1-carboxylate To a solution of intermediate 101 (3.0 g, 13.32 mmoles) in acetonitrile (30 ml) cooled to 20° C., Boc-anhydride (5.81 g, 26.65 mmoles) was added followed by DMAP (0.162 g, 1.33 mmoles) and triethylamine (1.34 g, 13.32 mmoles) and the reaction mixture was stirred at room temperature. After 12 h, the reaction mixture was concentrated and quenched with water and solid precipitated was filtered and dried under vacuum to afford the title compound as colourless solid (4.04 g, 93% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.31 (s, 1H), 7.54 (d, J=8.4 Hz, 1H),), 7.42 (dd, J=8.4, 1.3 Hz, 1H), 2.99 (q, J=7.6 Hz, 2H), 1.71 (s, 9H), 1.42 (t, J=7.6 Hz, 3H). Intermediate 103: tert-butyl 3-ethyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole-1-carboxylate To a solution of intermediate 102 (1.50 g, 4.61 mmoles) in Dioxan (24 ml), bis(pinacaloto)diboron (1.40 g, 5.53 mmoles) and potassium acetate (0.9050 g, 9.22 mmoles) were added and the system is degassed for 30 min Bis(diphenylphosphinoferrocene)dichloro palladium·CH2Cl2(0.188 g, 0.230 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (1.46 g, 85% yield).).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.47 (s, 1H), 7.86 (d, J=7.9 Hz, 1H),), 7.60 (d, J=8.0 Hz, 1H), 2.98 (q, J=7.6 Hz, 2H), 1.62 (s, 9H), 1.31 (s, 12H), 1.30 (t, J=7.6 Hz, 3H). Intermediate 104: 6-bromo-3-hydroxy-3-methylindolin-2-one To a ice-cold solution of methyl magnesium iodide prepared from magnesium (1.7 g, 70.78 mmoles) and methyl iodide (4.40 ml, 70.78 mmoles) in diethyl ether (60 ml), 6-bromoisatin (4 g, 17.69 mmoles) in THF (120 ml) was added and warmed to room temperature. After 12 h, the reaction mixture was cooled to 0° C., quenched with dilute HCl and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as brown solid (4.2 g, 93% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 10.34 (s, 1H), 7.23 (t, J=7.9 Hz, 1H), 7.14 (dd, J=7.9, 1.7 Hz, 1H), 6.93 (d, J=1.6 Hz, 1H), 5.92 (s, 1H), 1.33 (s, 3H). Intermediate 105: 6-bromo-3-methyl-1H-indole To a solution of intermediate 104 (3.0 g, 12.48 mmoles) in THF (120 ml) cooled to 0° C., boron-dimethylsulfide (2M in THF, 62.44 mmoles) was added and heated to 50° C. After 12 h, the reaction mixture was cooled to 0° C., quenched with methanol and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (1.15 g, 44% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 10.85 (s, 1H), 7.48 (d, J=1.8 Hz, 1H), 7.42 (d, J=8.4 Hz, 1H), 7.12 (t, J=1.1 Hz, 1H), 7.09 (dd, J=8.4, 1.8 Hz, 1H), 2.22 (s, 3H). Intermediate 106: 3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole To a solution of intermediate 105 (1.10 g, 5.23 mmoles) in Dioxan (33 ml), bis(pinacaloto)diboron (1.60 g, 6.28 mmoles) and potassium acetate (1.54 g, 15.70 mmoles) were added and the system is degassed for 30 min Bis(diphenylphosphinoferrocene)dichloro palladium·CH2Cl2(0.128 g, 0.157 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.651 g, 48% yield).).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 10.81 (s, 1H), 7.68 (s, 1H), 7.45 (d, J=7.9 Hz, 1H),), 7.28 (d, J=7.9 Hz, 1H), 7.19 (s, 1H), 2.23 (s, 3H), 1.28 (s, 12H). Intermediate 107: 3-(2,3-dihydrobenzofuran-5-yl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (0.70 g, 2.68 mmoles) in DMF (10 ml), ethanol (6 ml) and water (6 ml), 2,3-dihydrobenzofuran-5-boronic acid (0.527 g, 3.21 mmoles) and sodium carbonate (0.852 g, 8.04 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.610 g, 0.528 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.198 g, 29% yield1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.42 (s, 1H), 8.18 (s, 1H), 7.48 (s, 1H), 7.36 (d, J=8.1 Hz, 1H), 6.90 (d, J=8.2 Hz, 1H), 4.61 (d, J=8.7 Hz, 2H), 3.27 (d, J=8.7 Hz, 2H). Intermediate 108: tert-butyl 6-bromo-2-methyl-1H-benzo[d]imidazole-1-carboxylate To a solution of 6-bromo-2-methylbenzimidazole (1.00 g, 4.737 mmoles) in dichloromethane (20 ml) cooled to 20° C., Boc-anhydride (1.034 g, 4.737 mmoles) was added followed by DMAP (0.057 g, 0.473 mmoles) and triethylamine (0.479 g, 4.73 mmoles) and the reaction mixture was stirred at room temperature. After 12 h, the reaction mixture was concentrated and quenched with water and solid precipitated was filtered and dried under vacuum to afford the title compound as colourless solid as a mixture of two regioisomers (1.22 g, 83% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.00 (d, J=1.9 Hz, 0.53H), 7.80 (d, J=7.5 Hz, 0.47H), 7.78 (s, 0.47H), 7.55 (d, J=8.5 Hz, 0.53H), 7.47 (m, 1H), 2.69 (s, 1.4H), 2.68 (s, 1.6H), 1.63 (s, 9H). Intermediate 109: tert-butyl 2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-benzo[d]imidazole-1-carboxylate To a solution of intermediate 108 (0.500 g, 1.606 mmoles) in Dioxan (24 ml), bis(pinacaloto)diboron (0.489 g, 1.928 mmoles) and potassium acetate (0.946 g, 9.64 mmoles) were added and the system is degassed for 30 min. Bis(diphenylphosphinoferrocene)dichloro palladium·CH2Cl2(0.196 g, 0.241 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as brown solid as a mixture of two regioisomers (0.324 g, 56% yield).).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.42 (s, 0.65H), 8.15 (s, 0.35H), 7.92 (d, J=8.3 Hz, 0.35H), 7.78 (d, J=8.1 Hz, 1H), 7.69 (d, J=7.9 Hz, 0.65H), 2.88 (s, 3H), 1.72 (s, 5.85H), 1.71 (s, 3.15H), 1.35 (s, 12H). Intermediate 110: 4-bromo-2,6-difluorophenol To a solution of 2,6-Difluorophenol (10.0 g, 76.86 mmoles) in DMF (60 mll), N-bromosuccinimide (13.68 g, 76.86 mmoles) was added at 0° C. and stirred at RT for 20 h. The reaction mixture was concentrated, diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as light yellow liquid (15.1 g, 93% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.49 (s, 1H), 7.35 (d, J=6.2 Hz, 2H). Intermediate 111: 5-bromo-1,3-difluoro-2-methoxybenzene To a solution of intermediate 110 (15.0 g, 71.73 mmoles) in acetone (60 mll), potassium carbonate (29.75 g, 215.32 mmoles) was added at 0° C. followed by methyl iodide (22 ml, 358.86 mmoles) and stirred at RT for 22 h. The reaction mixture was concentrated, diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as light yellow liquid (11 g, 68% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 7.08 (d, J=7.8 Hz, 2H). Intermediate 112: 2-(3,5-difluoro-4-methoxyphenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane To a solution of intermediate 111 (2.0 g, 8.968 mmoles) in Dioxan (40 ml), bis(pinacaloto)diboron (2.73 g, 10.76 mmoles) and potassium acetate (2.64 g, 26.90 mmoles) were added and the system is degassed for 30 min Bis(diphenylphosphinoferrocene)dichloro palladium·CH2Cl2(0.219 g, 0.269 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as yellow liquid (2.2 g, 90% yield).).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ δ 7.318 (d, J=8.7 Hz, 2H), 4.02 (s, 3H), 1.32 (s, 12H). Intermediate 113: 3-(3,5-difluoro-4-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.0 g, 3.83 mmoles) in DMF (10 ml), ethanol (5 ml) and water (5 ml), intermediate 112 (1.55 g, 5.74 mmoles) and sodium carbonate (1.21 g, 11.49 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.221 g, 0.19 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.210 g, 19% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.66 (s, 1H), 8.20 (s, 1H), 7.36 (d, J=8.9 Hz, 2H), 6.96 (br s, 2H), 3.97 (s, 3H). Intermediate 114: 6-bromo-1,3-dimethyl-1H-indazole(a) and 6-bromo-2,3-dimethyl-2H-indazole(b) To a solution of intermediate 95 (2 g, 9.47 mmoles) in THF (30 ml) cooled to 0° C., sodium hydride (0.454 g, 60% in paraffin oil, 11.37 mmoles) was added and stirred for 10 min. Methyl iodide (2.0 gl, 14.21 mmoles) was added warmed to room temperature. After 12 h, the reaction mixture cooled to room temperature, quenched with water, extracted with ethyl acetate and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as colourless solid. Fraction I (114a, 0.90 g, 43% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.87 (d, J=1.0 Hz, 1H), 7.64 (d, J=9.5 Hz, 1H), 7.20 (dd, J=9.5, 1.5 Hz, 1H), 3.92 (s, 3H), 2.44 (s, 3H). Fraction II (114b, 0.80 g, 38% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 7.72 (d, J=1.3 Hz, 1H), 7.65 (d, J=8.8 Hz, 1H), 7.20 (dd, J=8.8, 1.6 Hz, 1H), 4.01 (s, 3H), 2.58 (s, 3H). Intermediate 115: 1,3-dimethyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole To a solution of intermediate 114a (0.90 g, 4.00 mmoles) in Dioxan (14 ml), bis(pinacaloto)diboron (1.1 g, 4.4 mmoles) and potassium acetate (0.785 g, 8.0 mmoles) were added and the system is degassed for 30 min. Bis(diphenylphosphinoferrocene)dichloro palladium·CH2Cl2(0.163 g, 0.200 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.85 g, 78% yield).).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 7.84 (s, 1H), 7.65 (d, J=8.0, 0.7 Hz, 1H), 7.53 (d, J=8.1 Hz, 1H), 4.03 (s, 3H), 2.56 (s, 3H), 1.38 (s, 12H). Intermediate 116: 2,3-dimethyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H-indazole To a solution of intermediate 114b (0.80 g, 3.55 mmoles) in Dioxan (14 ml), bis(pinacaloto)diboron (0.992 g, 3.90 mmoles) and potassium acetate (0.697 g, 7.10 mmoles) were added and the system is degassed for 30 min. Bis(diphenylphosphinoferrocene)dichloro palladium·CH2Cl2(0.145 g, 0.177 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.80 g, 83% yield).). (δ ppm, DMSO-d6, 400 MHz): δ 7.85 (s, 1H), 7.62 (dd, J=8.3, 0.8 Hz, 1H), 7.19 (d, J=8.4 Hz, 1H), 4.05 (s, 3H), 2.58 (s, 3H), 1.29 (s, 12H). Example 1 2-[(6-Amino-9H-purin-9-yl) methyl]-6-bromo-3-phenyl-4H-chromen-4-one To a solution of Adenine (0.685 g, 5.07 mmoles) in DMF (10 ml), potassium carbonate (0.701 g, 5.07 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 3 (1 g, 2.53 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.496 g, 43% yield). MP: 207-209° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.11 (d, J=2.4 Hz, 1H), 8.09 (d, J=10.4 Hz, 2H), 7.92 (dd, J=9.0, 2.4 Hz, 1H), 7.48-7.39 (m, 6H), 7.21 (s, 2H), 5.33 (s, 2H). Mass: 448.20 (M+). Example 2 6-Bromo-2-(morpholinomethyl)-3-phenyl-4H-chromen-4-one To a solution of Intermediate 3 (0.30 g, 0.761 mmoles) in THF (2 ml), was added morpholine (0.066 g, 0.761 mmoles) at RT and refluxed for 12 h. The reaction mixture was cooled, diluted with aqueous bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.40 g, 79% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.12 (d, J=2.2 Hz, 1H), 7.98 (dd, J=8.8, 2.3 Hz, 1H), 7.72 (d, J=8.9 Hz, 1H), 7.45-7.39 (m, 3H), 7.29 (d, J=7.0 Hz, 2H), 3.50 (t, J=4.2 Hz, 4H), 3.40 (s, 2H), 2.32 (br S, 4H). Example 2a 6-Bromo-2-(morpholinomethyl)-3-phenyl-4H-chromen-4-one hydrochloride To a solution of Example 2 (0.10 g, 0.249 mmoles) in THF (2 ml), was added hydrochloric acid in diethyl ether (2 ml) at 0° C. and stirred for 30 min. The precipitate formed was filtered, washed with pentane and dried to afford the title compound as pale yellow solid (0.110 g, 99% yield. MP: 229-230° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.13 (s, 1H), 8.06 (d, J=8.7 Hz, 1H), 7.77 (d, J=8.8 Hz, 1H), 7.48 (m, 3H), 7.32 (d, J=6.9 Hz, 2H), 4.35 (br s, 2H), 3.80 (br s, 4H), 3.59 (s, 2H), 3.25 (br s, 2H). Mass: 402.04 (M++1−HCl). Example 3 2-[(6-Amino-9H-purin-9-yl)methyl]-3-phenyl-4H-chromen-4-one To a solution of Example 1 (0.1 g, 0.22 mmoles) in methanol (10 ml), palladium on carbon (10 mg) was added and the solution was hydrogenated at RT under 5 kg/cm2pressure of hydrogen for 3 h. The solution was filtered through celite and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale yellow solid (0.030 g, 37% yield). MP: 173-175° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.10 (d, J=12.5 Hz, 1H), 8.05 (d, J=8.0 Hz, 1H), 7.77 (t, J=7.7 Hz, 1H), 7.48-7.41 (m, 6H), 7.22 (s, 2H), 5.34 (s, 2H). Mass: 370.05 (M++1). Example 4 2-(Morpholinomethyl)-3-phenyl-4H-chromen-4-one To a solution of Example 2 (0.1 g, 0.249 mmoles) in methanol (10 ml), palladium on carbon 20 mg) was added and the solution was hydrogenated at RT under 5 kg/cm2pressure of hydrogen for 4 h. The solution was filtered through celite and concentrated. The crude product was purified by column chromatography with ethyl acetate: petroleum ether to afford the title compound as brown solid (0.080 g, 87% yield).:1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.08 (d, J=7.8 Hz, 1H), 7.90 (t, J=7.4 Hz, 1H), 7.74 (d, J=8.4 Hz, 1H), 7.55 (t, J=7.4 Hz, 1H), 7.49 (m, 3H), 7.31 (d, J=6.5 Hz, 2H), 3.72 (br s, 4H), 3.42 (br s, 6H). Example 4a 2-(Morpholinomethyl)-3-phenyl-4H-chromen-4-one hydrochloride To a solution of Example 4 (0.065 g, 0.202 mmoles) in THF (2 ml), was added hydrochloric acid in diethyl ether (2 ml) at 0° C. and stirred for 30 min. The precipitate formed was filtered, washed with pentane and dried to afford the title compound as off-white solid (0.043 g, 60% yield. MP: 208-209° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 11.42 (br s, 1H), 8.08 (d, J=7.8 Hz, 1H), 7.90 (t, J=8.1 Hz, 1H), 7.79 (d, J=8.4 Hz, 1H), 7.55 (t, J=7.5 Hz, 1H), 7.49-7.44 (m, 3H), 7.33 (d, J=7.3 Hz, 2H), 4.24 (br s, 2H), 3.81 (br s, 5H), 3.08 (br s, 3H). 322.10 (M++1−HCl). Example 5 2-[(1H-Benzo[d]imidazol-1-yl) methyl]-6-bromo-3-phenyl-4H-chromen-4-one To a solution of intermediate 3 (0.10 g, 0.258 mmoles) in THF (2 ml), was added benzimidazole (0.059 g, 0.507 mmoles) at RT and refluxed for 2 h. The reaction mixture was cooled, diluted with aqueous bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as pale yellow solid (0.040 g, 40% yield). MP: 192-197° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.15 (s, 1H), 8.10 (d, J=2.3 Hz, 1H), 7.92 (dd, J=8.9, 2.3 Hz, 1H), 7.63 (m, 1H), 7.54 (m, 4H), 7.41 (d, J=6.8 Hz, 2H), 7.18 (m, 3H), 5.43 (s, 2H). 432.77 (M++1). Example 6 6-Bromo-2-[(4-methyl-1H-benzo[d]imidazol-1-yl) methyl]-3-phenyl-4H-chromen-4-one To a solution of intermediate 3 (0.10 g, 0.258 mmoles) in THF (2 ml), was added 4-methylbenzimidazole (0.066 g, 0.507 mmoles) at RT and refluxed for 2 h. The reaction mixture was cooled, diluted with aqueous bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as pale yellow solid (0.040 g, 35% yield). MP: 176-179° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.10 (s, 1H), 8.09 (d, J=2.3 Hz, 1H), 7.92 (dd, J=9.0, 2.5 Hz, 1H), 7.55 (m, 4H), 7.41 (d, J=6.8 Hz, 2H), 7.08 (t, J=7.5 Hz, 1H), 6.98 (m, 2H), 5.43 (s, 2H), 2.49 (s, 3H). Mass: 445.13 (M+). Example 7 2-[(1H-benzo[d]imidazol-1-yl) methyl]-3-phenyl-4H-chromen-4-one To a solution of intermediate 5 (0.10 g, 0.317 mmoles) in dioxin (2 ml), was added benzimidazole (0.074 g, 0.634 mmoles) at RT and refluxed for 12 h. The reaction mixture was cooled, diluted with aqueous bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol: dichloromethane to afford the title compound as yellow solid (0.050 g, 44% yield). MP: 186-191° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.16 (s, 1H), 8.04 (d, J=7.7 Hz, 1H), 7.78 (t, J=8.3 Hz, 1H), 7.64 (d, J=5.5 Hz, 1H), 7.54-7.42 (m, 7H), 7.18 (s, 3H), 5.43 (s, 2H). Mass: 352.83 (M+). Example 8 2-[(4-methyl-1H-benzo[d]imidazol-1-yl) methyl]-3-phenyl-4H-chromen-4-one To a solution of intermediate 5 (0.10 g, 0.317 mmoles) in dioxan (2 ml), was added 4-methylbenzimidazole (0.083 g, 0.634 mmoles) at RT and refluxed for 12 h. The reaction mixture was cooled, diluted with aqueous bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol: dichloromethane to afford the title compound as yellow solid (0.060 g, 51% yield). MP: 204-208° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.11 (s, 1H), 8.04 (d, J=7.6 Hz, 1H), 7.77 (t, J=7.6 Hz, 1H), 7.55 (m, 7H), 7.08 (t, J=8.0 Hz, 1H), 6.99 (d, J=7.6 Hz, 2H), 5.40 (s, 2H), 2.48 (s, 3H). Mass: 367.25 (M++1). Example 9 2-[(6-Chloro-9H-purin-9-yl) methyl]-3-phenyl-4H-chromen-4-one To a solution of 6-Chloropurine (0.146 g, 0.951 mmoles) in DMF (3 ml), potassium carbonate (0.131 g, 0.951 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 5 (0.150 g, 0.475 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brownish yellow (0.053 g, 28% yield). MP: 187-190° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.71 (s, 1H), 8.67 (s, 1H), 8.05 (d, J=7.0 Hz, 1H), 7.79 (dt, J=8.1, 1.5 Hz, 1H), 7.50 (t, J=7.9 Hz, 2H), 7.43 (m, 5H), 5.53 (s, 2H). 389.09 (M++1). Example 10 6-Bromo-2-[(6-chloro-9H-purin-9-yl) methyl]-3-phenyl-4H-chromen-4-one To a solution of 6-Chloropurine (0.117 g, 0.761 mmoles) in DMF (3 ml), potassium carbonate (0.105 g, 0.761 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 3 (0.150 g, 0.380 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brownish yellow (0.041 g, 22% yield). MP: 234-236° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.71 (s, 1H), 8.67 (s, 1H), 8.11 (d, J=2.4 Hz, 1H), 7.94 (d, J=9.0 Hz, 1H), 7.53 (d, J=8.8 Hz, 1H), 7.41 (m, 5H), 5.52 (s, 2H). Mass: 466.79 (M+−1). Example 11 2-((9H-Purin-6-ylthio) methyl)-3-phenyl-4H-chromen-4-one To a solution of 6-Mercaptopurine (0.162 g, 0.951 mmoles) in DMF (3 ml), potassium carbonate (0.131 g, 0.951 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 5 (0.150 g, 0.475 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.061 g, 33% yield). MP: 208-209° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 13.56 (s, 1H), 8.52 (s, 1H), 8.44 (s, 1H), 8.05 (d, J=7.9 Hz, 1H), 7.81 (t, J=7.2 Hz, 1H), 7.58 (d, J=8.4 Hz, 1H), 7.50 (m 4H), 7.39 (d, J=6.8 Hz, 2H), 4.62 (s, 2H). Mass: 386.78 (M+). Example 12 2-[(1H-Imidazol-1-yl) methyl]-3-phenyl-4H-chromen-4-one To a solution of intermediate 5 (0.10 g, 0.317 mmoles) in dioxan (2 ml), was added imidazole (0.043 g, 0.634 mmoles) at RT and refluxed for 12 h. The reaction mixture was cooled, diluted with aqueous bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brownish yellow solid (0.040 g, 41% yield). MP: 168-171° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.06 (dd, J=7.9, 1.3 Hz, 1H), 7.83 (dt, J=7.9, 1.5 Hz, 1H), 7.61 (s, 1H), 7.58 (d, J=8.5 Hz, 1H), 7.51 (m, 4H), 7.36 (dd, J=8.0 Hz, 2H), 7.12 (s, 1H), 6.90 (s, 1H), 5.10 (s, 2H). Mass: 303.29 (M++1). Example 13 2-[(9H-Purin-6-ylthio) methyl]-6-bromo-3-phenyl-4H-chromen-4-one To a solution of 6-Mercaptopurine (0.097 g, 0.570 mmoles) in DMF (5 ml), potassium carbonate (0.079 g, 0.570 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 3 (0.150 g, 0.380 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as grey solid (0.050 g, 28% yield). MP: 214-218° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 13.54 (s, 1H), 8.51 (s, 1H), 8.43 (s, 1H), 8.10 (d, J=2.2 Hz, 1H), 7.95 (dd, J=8.9, 2.3 Hz, 1H), 7.59 (d, J=9.0 Hz, 1H), 7.45 (m 3H), 7.34 (d, J=6.5 Hz, 2H), 4.62 (s, 2H). Mass: 465.11 (M+). Example 14 2-((4-Amino-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-6-bromo-3-phenyl-4H-chromen-4-one To a solution of 4-Aminopyrazalo[3,4-d]pyramiding (0.102 g, 0.761 mmoles) in DMF (3 ml), potassium carbonate (0.105 g, 0.761 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 3 (0.150 g, 0.380 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol: dichloromethane to afford the title compound as brownish yellow solid (0.031 g, 18% yield). MP: 236-240° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.16 (s, 1H), 8.11 (s, 1H), 8.10 (s, 1H), 7.89 (dd, J=8.8, 2.2 Hz, 1H), 7.72 (br s, 2H), 7.40 (m 6H), 5.41 (s, 2H). Mass: 449.78 (M++1). Example 15 2-[(6-Amino-9H-purin-9-yl) methyl]-6-bromo-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of Adenine (0.0983 g, 0.727 mmoles) in DMF (5 ml), potassium carbonate (0.125 g, 0.727 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 8 (0.150 g, 0.364 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.030 g, 18% yield). MP: 238-242° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.10 (s, 2H), 8.06 (s, 1H), 7.93 (dd, J=8.9, 2.2 Hz, 1H), 7.50 (d, J=8.9 Hz, 1H), 7.45 (t, J=8.2 Hz, 2H), 7.29 (t, J=8.8 Hz, 2H), 7.22 (s, 2H), 5.34 (s, 2H). Mass: 466.11 (M+). Example 16 2-[(6-Amino-9H-purin-9-yl) methyl]-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of Adenine (0.121 g, 0.899 mmoles) in DMF (5 ml), potassium carbonate (0.155 g, 0.899 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 10 (0.150 g, 0.450 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.040 g, 22% yield). MP: 212-216° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.11 (s, 2H), 8.07 (s, 1H), 8.05 (d, J=8.2 Hz, 1H), 7.78 (t, J=8.4 Hz, 1H), 7.50 (m, 4H), 7.29 (m, 4H), 5.34 (s, 2H). Mass: 388.21 (M+1). Example 17 6-Bromo-3-(4-fluorophenyl)-2-(morpholinomethyl)-4H-chromen-4-one To a solution of intermediate 8 (0.150 g, 0.364 mmoles) in THF (5 ml), was added morpholine (0.0634 g, 0.728 mmoles) at RT and refluxed for 4 h. The reaction mixture was cooled, diluted with aqueous bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.50 g, 32% yield).1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.35 (s, 1H), 7.81 (d, J=7.7 Hz, 1H), 7.39 (m, 3H), 7.18 (t, J=7.7 Hz, 2H), 3.80 (br st, 6H), 2.64 (br s, 4H). Example 17a 6-Bromo-3-(4-fluorophenyl)-2-(morpholinomethyl)-4H-chromen-4-one hydrochloride To a solution of Example 17 (0.050 g, 0.1192 mmoles) in THF (2 ml), was added hydrochloric acid in diethyl ether (2 ml) at 0° C. and stirred for 30 min. The precipitate formed was filtered, washed with pentane and dried to afford the title compound as yellow solid (0.030 g, 55% yield. MP: 232-236° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.13 (d, J=2.3 Hz, 1H), 8.06 (d, J=8.9 Hz, 1H), 7.80 (d, J=9.0 Hz, 1H), 7.38 (m, 4H), 4.24 (br s, 2H), 3.83 (br s, 4H), 3.62 (br s, 2H), 3.08 (br s, 2H). Mass: 419.75 (M+1−HCl). Example 18 3-(4-fluorophenyl)-2-(morpholinomethyl)-4H-chromen-4-one To a solution of intermediate 10 (0.150 g, 0.450 mmoles) in dioxan (5 ml), was added morpholine (0.0784 g, 0.90 mmoles) at RT and refluxed for 12 h. The reaction mixture was cooled, diluted with aqueous bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as off-white solid (0.80 g, 52% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.06 (dd, J=7.9.1.0 Hz, 1H), 7.84 (dt, J=8.3, 1.2 Hz, 1H), 7.70 (d, J=8.4 Hz, 1H), 7.51 (t, J=7.6 Hz, 1H), 7.36 (dt, J=6.0, 2.9 Hz, 2H), 7.28 (t, J=8.9 Hz, 2H), 3.50 (br s, 4H), 3.39 (br s, 2H), 2.49 (br s, 4H). Example 18a 3-(4-fluorophenyl)-2-(morpholinomethyl)-4H-chromen-4-one hydrochloride To a solution of Example 18 (0.080 g, 0.235 mmoles) in THF (2 ml), was added hydrochloric acid in diethyl ether (2 ml) at 0° C. and stirred for 30 min. The precipitate formed was filtered, washed with pentane and dried to afford the title compound as yellow solid (0.080 g, 90% yield. MP: 225-229° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.08 (d, J=6.6 Hz, 1H), 7.95 (t, J=7.3 Hz, 1H), 7.78 (d, J=8.2 Hz, 1H), 7.55 (t, J=7.6 Hz, 1H), 7.38 (m, 4H), 4.30 (br s, 2H), 3.88 (br s, 6H), 3.12 (br s, 2H).). Mass: 340.09 (M+1−HCl). Example 19 2-[(6-Amino-9H-purin-9-yl) methyl]-6-bromo-3-o-tolyl-4H-chromen-4-one To a solution of Adenine (0.099 g, 0.735 mmoles) in DMF (3 ml), potassium carbonate (0.101 g, 0.735 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 13 (0.150 g, 0.367 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.046 g, 27% yield). MP: 252-255° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.11 (d, J=2.3 Hz, 1H), 8.03 (s, 1H), 7.97 (s, 1H), 7.94 (dd, J=8.9, 2.4 Hz, 1H), 7.54 (d, J=8.8 Hz, 1H), 7.31-7.22 (m, 6H), 5.22 (s, 2H), 2.00 (s, 3H).). Mass: 463.85 (M+1). Example 20 7-[(6-Bromo-4-oxo-3-phenyl-4H-chromen-2-yl) methyl]-1, 3-dimethyl-1H-purine-2,6(3H,7H)-dione To a solution of Theophylline (0.137 g, 0.761 mmoles) in DMF (3 ml), potassium carbonate (0.105 g, 0.761 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 3 (0.150 g, 0.380 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the title compound as brown solid (0.040 g, 21% yield). MP: 253-255° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.11 (d, J=2.4 Hz, 1H), 8.03 (s, 1H), 7.94 (dd, J=8.9, 2.4 Hz, 1H), 7.52 (d, J=9.1 Hz, 1H), 7.42 (m, 3H), 7.31 (d, J=6.6 Hz, 1H), 5.51 (s, 2H), 3.13 (s, 6H).). Mass: 492.69 (M+). Example 21 2-(1-(6-Amino-9H-purin-9-yl) ethyl)-6-bromo-3-phenyl-4H-chromen-4-one To a solution of Adenine (0.266 g, 1.969 mmoles) in DMF (10 ml), potassium carbonate (0.272 g, 1.969 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 15 (0.400 g, 0.984 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.200 g, 44% yield). MP: 230-231° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.45 (s, 1H), 8.08 (d, J=2.4 Hz, 1H), 8.02 (s, 1H), 7.99 (dd, J=8.9, 2.4 Hz, 1H), 7.68 (d, J=8.9 Hz, 1H), 7.47 (m, 3H), 7.35 (d, J=6.5 Hz, 1H), 7.20 (s, 2H), 5.69 (q, J=7.2 Hz, 1H), 1.88 (d, J=7.2 Hz, 3H).). Mass: 463.92 (M+1). Example 22 2-(1-(9H-Purin-6-ylthio) ethyl)-6-bromo-3-phenyl-4H-chromen-4-one To a solution of 6-Mercaptopurine (0.251 g, 1.477 mmoles) in DMF (10 ml), potassium carbonate (0.255 g, 1.846 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 15 (0.300 g, 0.738 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light green solid (0.130 g, 37% yield). MP: 234-237° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 13.54 (s, 1H), 8.40 (s, 1H), 8.37 (s, 1H), 8.10 (d, J=2.5 Hz, 1H), 7.99 (dd, J=8.8, 2.5 Hz, 1H), 7.77 (d, J=8.9 Hz, 1H), 7.39 (m, 4H), 7.26 (s, 2H), 5.47 (q, J=7.2 Hz, 1H), 1.79 (d, J=7.1 Hz, 3H). Mass: 478.83 (M+). Example 23 2-(1-(6-Amino-9H-purin-9-yl) ethyl)-3-phenyl-4H-chromen-4-one To a solution of example 21 (0.080 g, 0.173 mmoles) in methanol (10 ml), palladium on carbon (10% 16 mg) was added and the solution was hydrogenated at RT under 5 kg/cm2pressure of hydrogen for 24 h. The solution was filtered through celite and concentrated. The crude product was purified by column chromatography with methanol: dichloromethane to afford the title compound as light yellow solid (0.025 g, 38% yield). MP: 254-257° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.46 (s, 1H), 8.03 (s, 1H), 8.01 (d, J=1.6 Hz, 1H), 7.83 (dt, J=7.3, 1.7 Hz, 1H), 7.65 (d, J=8.2 Hz, 1H), 7.50 (m, 4H), 7.37 (dd, J=8.1, 1.7 Hz, 1H), 7.22 (s, 2H), 5.67 (q, J=7.3 Hz, 1H), 1.89 (d, J=7.2 Hz, 3H).). Mass: 384.19 (M+1). Example 24 (S)-2-(1-(9H-purin-6-ylamino) ethyl)-6-bromo-3-phenyl-4H-chromen-4-one To a solution of intermediate 17 (0.20 g, 0.581 mmoles) in tert-butanol (6 ml), N,N-diisopropylethyl amine (0.2 ml, 1.162 mmoles) and 6-bromopurine (0.087 g, 0.435 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as yellow solid (0.065 g, 24% yield). MP: 151-154° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.94 (s, 1H), 8.09 (br s, 3H), 7.94 (d, J=7.9 Hz, 1H), 7.59 (d, J=8.7 Hz, 1H), 7.42 (m, 6H), 5.22 (br t, 1H), 1.82 (d, J=6.4 Hz, 3H). Mass: 463.99 (M+1). Example 25 2-((9H-purin-6-ylamino) methyl)-6-bromo-3-phenyl-4H-chromen-4-one To a solution of intermediate 19 (0.20 g, 0.605 mmoles) in tert-butanol (4 ml), N,N-diisopropylethylamine (0.2 ml, 1.211 mmoles) and 6-bromopurine (0.096 g, 0.484 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as yellow solid (0.065 g, 24% yield). MP: 151-154° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.90 (s, 1H), 8.20 (ms, 4H), 7.91 (dd, J=9.0, 2.5 Hz, 1H), 7.49-7.35 (m, 6H), 4.64 (br s, 2H). Mass: 448.17 (M+). Example 26 2-(1-(4-amino-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-6-bromo-3-phenyl-4H-chromen-4-one To a solution of 4-Aminopyrazalo[3,4-d]pyrimidine (0.299 g, 2.215 mmoles) in DMF (10 ml), potassium carbonate (0.382 g, 2.769 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 15 (0.450 g, 1.107 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.80 g, 16% yield). MP: 239-240° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.10 (d, J=2.5 Hz, 1H), 8.09 (s, 1H), 8.00 (s, 1H), 7.97 (dd, J=8.9, 2.4 Hz, 1H), 7.69 (br s, 2H), 7.60 (d, J=9.0 Hz, 1H), 7.31 (br s, 3H), 7.12 (br s, 2H), 5.83 (q, J=7.1 Hz, 1H), 1.83 (d, J=7.0 Hz, 3H). Mass: 461.96 (M+). Example 27 2-((6-Amino-9H-purin-9-yl) methyl)-6-methoxy-3-phenyl-4H-chromen-4-one To a solution of adenine (0.234 g, 1.738 mmoles) in DMF (6 ml), potassium carbonate (0.240 g, 1.738 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 22 (0.300 g, 0.869 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale yellow solid (0.052 g, 15% yield). MP: 197-198° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.08 (s, 1H), 8.06 (s, 1H), 7.47 (m, 7H), 7.35 (dd, J=9.0, 3.1 Hz, 1H), 7.19 (s, 2H), 5.32 (s, 2H), 3.83 (s, 3H). Mass: 400.03 (M++1). Example 28 2-(1-(6-Amino-9H-purin-9-yl) ethyl)-6-bromo-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.190 g, 1.408 mmoles) in DMF (6 ml), potassium carbonate (0.194 g, 1.408 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 25 (0.300 g, 0.704 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid consisting of a mixture of two atrop-isomers (0.082 g, 24% yield). MP: 256-258° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ [8.47 (s), 8.38 (s), 1H], 8.09 (d, J=2.5 Hz, 1H), [8.05 (dd, J=9.0, 3.0 Hz), 8.00 (dd, J=9.0, 2.5 Hz), 1H], [8.01 (s), 7.91 (s), 1H], [7.81 (d, J=9.0 Hz), 7.69 (d, J=8.9 Hz), 1H], 7.50 (m, 2H), 7.34 (m, 2H), [7.22 (s), 7.16 (s), 2H], [5.71 (q), J=7.0 Hz), 5.64 (q, J=7.2 Hz), 1H], 1.96 (d, J=7.2 Hz), 1.86 (d, J=7.2 Hz), 3H]. Mass: 481.73 (M+1). Example 29 2-((6-Amino-9H-purin-9-yl) methyl)-6-bromo-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.131 g, 0.970 mmoles) in DMF (4 ml), potassium carbonate (0.133 g, 0.970 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 27 (0.200 g, 0.485 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.031 g, 14% yield). MP: 231-233° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.11 (d, J=2.5 Hz, 1H), 8.08 (s, 1H), 8.04 (s, 1H), 7.96 (dd, J=8.9, 2.5 Hz, 1H), 7.54 (d, J=9.0 Hz, 1H), 7.49 (d, J=3.5 Hz, 1H), 7.30 (m, 4H), [5.42 (d, J=16.5 Hz), 5.30 (d, J=16.5 Hz) 2H]. Mass: 466.23 (M+). Example 30 2-(1-(4-Amino-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-phenyl-4H-chromen-4-one To a solution of 4-Aminopyrazalo[3,4-d]pyrimidine (0.279 g, 2.58 mmoles) in DMF (7 ml), potassium carbonate (0.357 g, 2.58 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 29 (0.340 g, 1.03 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.80 g, 16% yield). MP: 226-227° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.09 (s, 1H), 8.04 (dd, J=7.9, 1.5 Hz, 1H), 8.01 (s, 1H), 7.82 (dt, J=8.6, 1.6 Hz, 1H), 7.58 (d, J=8.4 Hz, 2H), 7.51 (t, J=7.4 Hz, 2H). 7.31 (br s, 3H), 5.83 (q, J=7.0 Hz, 1H), 1.84 (d, J=7.0 Hz, 3H). Mass: 383.40 (M+). Example 31 2-(1-(6-Amino-9H-purin-9-yl) propyl)-3-phenyl-4H-chromen-4-one To a solution of adenine (0.190 g, 1.408 mmoles) in DMF (6 ml), potassium carbonate (0.194 g, 1.408 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 32 (0.300 g, 0.704 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.082 g, 24% yield). MP: 223-225° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.54 (s, 1H), 8.04 (s, 1H), 8.03 (dd, J=7.9, 1.5 Hz, 1H), 7.86 (dt, J=7.1, 1.6 Hz), 7.78 (d, J=7.9 Hz, 1H), 7.51 (m, 4H), 7.33 (dd, J=7.8, 1.6 Hz, 2H), 7.23 (s, 2H), 5.52 (t, J=7.3 Hz, 1H), 2.49 (m, 2H), 0.74 (t, J=7.3 Hz, 3H). Mass: 398.12 (M+1). Example 32 2-(1-(6-Amino-9H-purin-9-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.233 g, 1.728 mmoles) in DMF (6 ml), potassium carbonate (0.238 g, 1.728 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 36 (0.300 g, 0.864 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.200 g, 57% yield). MP: 155-158° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.46 (s, 1H), 8.02 (s, 1H), 8.02 (dd, J=7.7, 1.4 Hz, 1H), 7.84 (dt, J=8.6, 1.5 Hz, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.51 (m, 2H), 7.27-7.19 (m, 5H), 5.70 (q, J=7.2 Hz, 1H), 1.90 (d, J=7.2 Hz, 3H). Mass: 402.25 (M+1). Example 33 2-((6-Amino-9H-purin-9-yl)methyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.227 g, 1.68 mmoles) in DMF (5 ml), potassium carbonate (0.232 g, 1.68 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 38 (0.280 g, 0.840 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.046 g, 13% yield). MP: 202-205° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.08 (s, 1H), 8.04 (s, 1H), 8.05 (dd, J=5.0, 1.8 Hz, 1H), 7.81 (dt, J=8.5, 1.7 Hz, 1H), 7.53-7.441 (m, 4H), 7.30 (d, J=6.6 Hz, 1H), 7.26 (d, J=6.6 Hz, 1H), 7.22 (s, 2H), 15.43 (d, J=16.4 Hz), 5.30 (d, J=16.4 Hz), 2H]. Mass: 387.83 (M+). Example 34 2-(1-(6-Amino-9H-purin-9-yl)ethyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.179 g, 1.32 mmoles) in DMF (5 ml), potassium carbonate (0.183 g, 1.68 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 40 (0.230 g, 0.662 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.080 g, 30% yield). MP: 247-250° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ [8.48 (s), 8.39 (s), 1H], [8.05 (s), 7.91 (s), 1H], 8.03 (d, J=7.8 Hz, 1H), 7.86 (m, 2H), 7.53 (m, 3H), 7.36-7.18 (m, 4H), 5.68 (q, J=7.3 Hz, 1H), [1.97 (d, J=7.2 Hz), 1.87 (d, J=7.1 Hz), 3H]. Mass: 402.32 (M+1). Example 35 2-(1-(6-Amino-9H-purin-9-yl)propyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.524 g, 3.87 mmoles) in DMF (5 ml), potassium carbonate (0.535 g, 3.87 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 43 (0.700 g, 1.93 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.060 g, 7% yield). MP: 160-163° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ [8.57 (s), 8.45 (s), 1H], [8.08 (s), 7.92 (s), 1H], 8.03 (d, J=8.0 Hz, 1H), 7.89 (m, 2H), 7.54 (m, 3H), 7.35-7.17 (m, 4H), [5.48 (t, J=7.9 Hz), 5.46 (t, J=7.0 Hz), 1H], 2.48 (m, 2H), [0.82 (t, J=7.4 Hz), 0.75 (t, J=7.3 Hz), 3H]. Mass: 416.04 (M+1). Example 36 2-(1-(6-amino-9H-purin-9-yl)propyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.404 g, 2.99 mmoles) in DMF (12 ml), potassium carbonate (0.413 g, 2.99 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 46 (0.540 g, 1.49 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brownish yellow solid (0.115 g, 19% yield). MP: 102-107° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.54 (s, 1H), 8.03 (s, 1H), 8.01 (d, J=10.1 Hz, 1H), 7.87 (t, J=8.4 Hz, 1H), 7.79 (d, J=8.4 Hz, 1H), 7.52 (t, J=7.6 Hz, 2H), 7.28 (m, 3H), 7.18 (d, J=7.4 Hz, 2H), 5.51 (t, J=7.9 Hz, 1H), 2.39 (m, 2H), 0.76 (t, J=7.3 Hz, 3H). Mass: 415.97 (M+). Example 37 2-(1-(6-Amino-9H-purin-9-yl)propyl)-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.389 g, 2.87 mmoles) in DMF (12 ml), potassium carbonate (0.497 g, 2.87 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 49 (0.520 g, 1.43 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.55 g, 9% yield). MP: 223-227° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.54 (s, 1H), 8.05 (s, 1H), 8.03 (dd, J=8.0, 1.6 Hz, 1H), 7.86 (dt, J=7.1, 1.6 Hz, 1H), 7.78 (d, J=7.8 Hz, 1H), 7.51 (dt, J=8.0, 1.1 Hz, 1H), 7.38 (t, J=8.1 Hz, 2H), 7.30 (t, J=8.8 Hz, 2H), 7.23 (s, 2H), 5.50 (t, J=7.7 Hz, 1H), 2.39 (m, 2H), 0.76 (t, J=7.3 Hz, 3H). Mass: 416.11 (M+1). Example 38 2-(1-(6-amino-9H-purin-9-yl)propyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of adenine (0.374 g, 2.76 mmoles) in DMF (10 ml), potassium carbonate (0.382 g, 2.76 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 52 (0.500 g, 1.38 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.110 g, 19% yield). MP: 266-272° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.54 (s, 1H), 8.04 (s, 1H), 7.92 (dd, J=9.3, 4.3 Hz, 1H), 7.78 (dt, J=8.6, 3.2 Hz, 1H), 7.70 (dd, J=8.3, 5.3 Hz, 1H), 7.46 (m, 3H), 7.32 (d, J=6.4 Hz, 2H), 7.21 (s, 2H), 5.53 (t, J=7.7 Hz, 1H), 2.39 (m, 2H), 0.74 (t, J=7.3 Hz, 3H). Mass: 416.11 (M+1). Example 39 2-(1-(6-Amino-9H-purin-9-yl) ethyl)-3-(4-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.412 g, 3.05 mmoles) in DMF (10 ml), potassium carbonate (0.527 g, 3.81 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 55 (0.530 g, 1.52 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.050 g, 8% yield). MP: 210-212° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.46 (s, 1H), 8.03 (s, 1H), 8.02 (dd, J=8.1, 1.5 Hz, 1H), 7.83 (dt, J=7.1, 1.5 Hz, 1H), 7.67 (d, J=8.3 Hz, 1H), 7.50 (t, J=7.7 Hz, 1H), 7.41 (d, J=8.6 Hz, 1H), 7.39 (d, J=8.4 Hz, 1H), 7.30 (t, J=8.9 Hz, 2H), 7.23 (s, 1H), 5.68 (q, J=6.9 Hz, 1H), 1.90 (d, J=7.2 Hz, 3H). Mass: 402.32 (M+1). Example 40 2-(1-(6-Amino-9H-purin-9-yl)ethyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of adenine (0.389 g, 2.88 mmoles) in DMF (12 ml), potassium carbonate (0.398 g, 2.88 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 57 (0.500 g, 1.44 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.210 g, 36% yield). MP: 264-269° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.46 (s, 1H), 8.02 (s, 1H), 7.80 (dd, J=9.1, 4.4 Hz, 1H), 7.74 (m, 2H), 7.48 (m, 3H), 7.36 (dd, J=8.0, 1.7 Hz, 2H), 7.21 (s, 1H), 5.68 (q, J=7.2 Hz, 1H), 1.88 (d, J=7.2 Hz, 3H). Mass: 402.11 (M+1). Example 41 2-(1-(4-Amino-3-(3-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 58 (0.498 g, 2.06 mmoles) in DMF (5 ml), potassium carbonate (0.356 g, 2.50 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 29 (0.340 g, 1.03 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.160 g, 32% yield). MP: 176-178° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.09 (s, 1H), 8.04 (d, J=8.0 Hz, 1H), 7.83 (t, J=7.0 Hz, 1H), 7.63 (d, J=6.5 Hz, 2H), 7.51 (t, J=7.3 Hz, 1H), 7.46 (t, J=8.1 Hz, 1H), 7.33 (m, 3H), 7.12 (m, 4H), 7.06 (dd, J=8.2, 2.3 Hz, 1H), 5.98 (q, J=6.7 Hz, 1H), 3.81 (s, 3H), 1.90 (d, J=7.0 Hz, 3H). Mass: 490.10 (M+1). Example 42 2-(1-(4-Amino-3-(3-hydroxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-phenyl-4H-chromen-4-one To a solution of example 41 (0.130 g, 0.265 mmoles) in dichloromethane (26 ml), BBr3(1M in dichloromethane, 2.6 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the title compound as light yellow solid (0.070 g, 56% yield). MP: 212-216° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.78 (s, 1H), 8.24 (d, J=7.5 Hz, 1H), 8.05 (d, J=7.9 Hz, 1H), 7.85 (t, J=8.4 Hz, 1H), 7.65 (d, J=8.6 Hz, 1H), 7.53 (t, J=7.7 Hz, 1H), 7.36-7.02 (m, 9H), 6.90 (d, J=8.2 Hz, 1H), 6.03 (q, J=6.9 Hz, 1H), 1.91 (d, J=7.3 Hz, 3H). Mass: 476.17 (M+1). Example 43 2-((9H-purin-6-ylamino) methyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 59 (1.50 g, 7.06 mmoles) in dichloromethane (15 ml), triethylamine (2.9 ml, 21.20 mmoles) was added followed by N-Boc-Glycine (1.3 g, 7.77 mmoles). To this mixture HATU (5.3 g, 14.13 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (1.12 g). To a solution of this intermediate (0.60 g) in dichloromethane (10 ml), trifluoroacetic acid (2.5 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (0.38 g). To a solution of this amine intermediate (0.37 g, 1.47 mmoles) in tert-butanol (6 ml), N,N-diisopropylethylamine (0.5 ml, 2.94 mmoles) and 6-chloropurine (0.226 g, 1.47 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as brown solid (0.131 g, 24% yield). MP: 155-158° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.96 (s, 1H), 8.14-8.040 (m, 4H), 7.77 (t, J=8.2 Hz, 1H), 7.48-7.36 (m, 7H), 4.60 (br s, 2H). Mass: 369.91 (M+). Example 44 2-(1-(6-Amino-9H-purin-9-yl) ethyl)-3-o-tolyl-4H-chromen-4-one To a solution of intermediate 61 (0.610 g, 2.30 mmoles) in acetic acid (8 ml) bromine (0.23 ml, 4.61 mmoles) was added at 0° C. The reaction mixture heated to 60° C. After 6 h, the reaction mixture was cooled to RT, quenched by the addition of water. The precipitate formed was filtered and dried under reduced pressure to afford the bromo intermediate (0.700 g). This intermediate (0.650 g, 1.88 mmoles) was added to a solution of adenine (0.510 g, 3.77 mmoles) and potassium carbonate (0.521 g, 3.77 mmoles) in DMF (15 ml). After 12 h, the reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid as a atrope isomers (0.030 g, 4% yield). MP: 202-205° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.42 (d, J=3.5 Hz, 1H), [8.07 (s), 7.95 (s), 1H], 8.04 (t, J=5.6 Hz, 1H), 7.84 (q, J=7.2 Hz, 1H), [7.70 (d, J=8.2 Hz), 7.68 (d, J=8.1 Hz), 1H], 7.51 (t, J=7.6 Hz, 1H), 7.35-7.20 (m, 6H), 5.56 (m, 1H), [2.09 (s), 1.90 (s), 3H], [1.95 (d, J=7.1 Hz), 1.84 d, J=7.3 Hz), 3H]. Mass: 397.77 (M+). Example 45 2-((9H-purin-6-ylamino) methyl)-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 64 (0.330 g, 1.22 mmoles) in tert-butanol (4 ml), N,N-diisopropylethylamine (0.42 ml, 2.45 mmoles) and 6-bromopurine (0.195 g, 0.980 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as yellow solid (0.040 g, 8% yield). MP: 143-147° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.90 (s, 1H), 8.20 (br s, 1H), 8.10 (s, 1H), 8.09 (s, 1H), 8.05 (dd, J=7.9, 1.4 Hz, 1H), 7.79 (dt, J=8.6, 1.5 Hz, 1H), 7.51-7.41 (m, 4H), 7.28 (m, 2H), 4.64 (br s, 2H). Mass: 387.90 (M+). Example 46 2-((9H-purin-6-ylamino) methyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 65 (1.50 g, 6.51 mmoles) in dichloromethane (15 ml), triethylamine (2.7 ml, 19.54 mmoles) was added followed by N-Boc-Glycine (1.3 g, 7.81 mmoles). To this mixture HATU (4.9 g, 13.03 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (0.80 g). To a solution of this intermediate (0.80 g) in dichloromethane (10 ml), trifluoroacetic acid (1.5 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (0.471 g). To a solution of this amine intermediate (0.30 g, 1.14 mmoles) in tert-butanol (6 ml), N,N-diisopropylethylamine (0.5 ml, 2.94 mmoles) and 6-bromopurine (0.177 g, 0.891 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as brown solid (0.235 g, 55% yield). MP: 211-214° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.97 (s, 1H), 8.20 (br s, 1H), 8.14 (s, 1H), 8.11 (s, 1H), 8.06 (dd, J=7.9, 1.4 Hz, 1H), 7.78 (dt J=8.4, 1.3 Hz, 1H), 7.49 (m, 3H), 7.27-7.17 (m, 3H), 4.10 (q, J=5.3 Hz, 1H), 3.16 (d, J=5.0 Hz, 2H). Mass: 387.90 (M+). Example 47 (S)-2-(1-(9H-purin-6-ylamino) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 65 (2.0 g, 8.68 mmoles) in dichloromethane (20 ml), triethylamine (3.6 ml, 26.06 mmoles) was added followed by N-Boc-Alanine (1.97 g, 10.42 mmoles). To this mixture HATU (6.6 g, 17.37 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (1.70 g). To a solution of this intermediate (1.7 g) in dichloromethane (20 ml), trifluoroacetic acid (3 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (0.641 g). To a solution of this amine intermediate (0.30 g, 1.05 mmoles) in tert-butanol (6 ml), N,N-diisopropylethylamine (0.36 ml, 2.17 mmoles) and 6-bromopurine (0.168 g, 0.847 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as off-white solid (0.041 g, 10% yield). MP: 135-138° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.95 (s, 1H), 8.15 (t, J=6.8 Hz, 1H), 8.11 (s, 1H), 8.08 (s, 1H), 8.03 (d, J=7.8 Hz, 1H), 7.81 (t, J=7.3 Hz, 1H), 7.60 (d, J=8.3 Hz, 1H), 7.49 (t, J=7.3 Hz, 2H), 7.25 (m, 3H), 5.19 (br m, 1H), 1.56 (d, J=6.9 Hz, 3H). Mass: 402.18 (M++1). Example 48 2-(1-(6-amino-9H-purin-9-yl) ethyl)-6-fluoro-3-(2-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.443 g, 3.28 mmoles) in DMF (10 ml), potassium carbonate (0.453 g, 3.28 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 68 (0.600 g, 1.64 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid consisting of a mixture of two atrop-isomers (0.082 g, 24% yield). MP: 245-248° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ [8.49 (s), 8.39 (s), 1H], [8.05 (s), 7.91 (s), 1H], 7.92 (m, 1H), 7.81 (m, 2H), 7.52 (m, 2H), 7.36 (m, 4H), [5.69 (q, J=7.2 Hz), 5.64 (q, J=7.2 Hz), 1H], 1.96 (d, J=7.1 Hz), 1.86 (d, J=7.2 Hz), 3H]. Mass: 419.82 (M+). Example 49 2-(1-(6-Amino-9H-purin-9-yl) ethyl)-3-(3,5-difluorophenyl)-4H-chromen-4-one To a solution of adenine (0.370 g, 2.73 mmoles) in DMF (8 ml), potassium carbonate (0.378 g, 2.73 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 72 (0.500 g, 1.36 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid (0.121 g, 21% yield). MP: 267-269° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.45 (s, 1H), 8.02 (s, 1H), 8.01 (d, J=5.9 Hz, 1H), 7.85 (t, J=8.5 Hz, 1H), 7.70 (d, J=8.4 Hz, 1H), 7.52 (t, J=7.7 Hz, 1H), 7.30 (t, J=9.4 Hz, 1H), 7.23 (s, 2H), 7.11 (d, J=7.6 Hz, 2H), 5.70 (q, J=7.2 Hz, 1H), 1.91 (d, J=7.1 Hz, 3H). Mass: 419.82 (M+). Example 50 2-(1-(6-amino-9H-purin-9-yl) ethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of adenine (0.370 g, 2.73 mmoles) in DMF (8 ml), potassium carbonate (0.378 g, 2.73 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 75 (0.500 g, 1.36 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid (0.150 g, 26% yield). MP: 252-255° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.46 (s, 1H), 8.02 (s, 1H), 7.82 (dd, J=9.2, 4.4 Hz, 1H), 7.76 (dd, J=8.0, 3.0 Hz, 1H), 7.72 (td, J=6.8, 3.6 Hz, 1H), 7.51 (q, J=7.8 Hz, 1H), 7.28-7.18 (m, 5H), 5.70 (q, J=7.0 Hz, 1H), 1.89 (d, J=7.2 Hz, 3H). Mass: 420.03 (M++1). Example 51 2-(1-(4-amino-3-(3-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 58 (0.484 g, 2.01 mmoles) in DMF (6 ml), potassium carbonate (0.345 g, 2.50 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 36 (0.350 g, 1.00 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.302 g, 59% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.07 (s, 1H), 8.04 (dd, J=7.9, 1.4 Hz, 1H), 8.02 (dt, J=6.9, 1.4 Hz, 1H), 7.67 (d, J=8.4 Hz, 1H), 7.53 (t, J=7.9 Hz, 1H), 7.46 (t, J=7.9 Hz, 1H), 7.31 (br s, 1H), 7.19 (d, J=7.7 Hz, 1H), 7.10 (t, J=2.1 Hz, 1H), 7.07 (dt, J=8.6, 4.0 Hz, 2H), 6.90 (br s, 2H), 6.05 (q, J=6.9 Hz, 1H), 3.80 (s, 3H), 1.90 (d, J=7.1 Hz, 3H). Example 51a 2-(1-(4-amino-3-(3-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 51 (0.150 g, 0.290 mmoles) in dichloromethane (25 ml), BBr3(1M in dichloromethane, 1.5 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as grey colour solid (0.110 g, 75% yield). MP: 282-285° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.69 (s, 1H), 8.06 (s, 1H), 8.05 (dd, J=8.0, 1.6 Hz, 1H), 7.86 (dt, J=7.2, 1.6 Hz, 1H), 7.68 (t, J=8.2 Hz, 1H), 7.53 (dt, J=8.0, 0.9 Hz, 1H), 7.34 (t, J=8.0 Hz, 1H), 7.29 (br s, 1H), 7.06-6.84 (m, 6H), 6.03 (q, J=7.1 Hz, 1H), 1.89 (d, J=7.1 Hz, 3H). Mass: 493.95 (M+). Example 52 2-((4-Amino-3-(3-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 58 (0.765 g, 3.17 mmoles) in DMF (7 ml), potassium carbonate (0.548 g, 3.96 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 5 (0.500 g, 1.58 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.280 g, 37% yield). MP: 111-115° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.23 (s, 1H), 8.05 (dd, J=8.0, 1.4 Hz, 1H), 7.77 (dt, J=8.5, 1.5 Hz, 1H), 7.49-7.31 (m, 8H), 7.20 (d, J=7.6 Hz, 1H), 7.12 (s, 1H), 7.04 (dd, J=8.0, 2.1 Hz, 1H), 5.51 (s, 2H), 3.80 (s, 3H). Mass: 475.89 (M+). Example 53 2-((4-Amino-3-(3-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of example 52 (0.150 g, 0.315 mmoles) in dichloromethane (30 ml), BBr3(1M in dichloromethane, 1.5 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.040 g, 27% yield). MP: 154-158° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.69 (s, 1H), 8.22 (s, 1H), 8.06 (dd, J=7.8, 1.2 Hz, 1H), 7.49 (t, J=7.3 Hz, 1H), 7.44 (d, J=8.5 Hz, 1H), 7.37-7.29 (m, 6H), 7.03 (d, J=7.9 Hz, 2H), 6.86 (dd, J=8.3, 1.6 Hz, 1H), 5.49 (s, 2H). Mass: 462.03 (M++1). Example 54 2-((4-amino-3-(3-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 58 (0.278 g, 1.15 mmoles) in DMF (6 ml), potassium carbonate (0.363 g, 2.62 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 77 (0.350 g, 1.05 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid (0.220 g, 40% yield). MP: 175-178° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.21 (s, 1H), 8.05 (dd, J=8.0, 1.7 Hz, 1H), 7.80 (m, 1H), 7.51 (m, 2H), 7.45 (t, J=8.0 Hz, 1H), 7.39 (m, 1H), 7.18-7.08 (m, 5H), 7.04 (dd, J=8.3, 2.0 Hz, 1H), 5.54 (s, 2H), 3.80 (s, 3H). Mass: 493.81 (M+). Example 55 2-((4-amino-3-(3-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of example 54 (0.200 g, 0.383 mmoles) in dichloromethane (30 ml), BBr3(1M in dichloromethane, 2.0 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.070 g, 36% yield). MP: 280-283° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.69 (s, 1H), 8.20 (s, 1H), 8.06 (dd, J=8.2, 1.7 Hz, 1H), 7.80 (m, 1H), 7.51 (m, 2H), 7.39 (m, 2H), 7.17 (m, 2H), 7.11 (dt, J=8.7, 2.2 Hz, 1H), 7.02 (d, J=8.6 Hz, 1H), 7.00 (s, 1H), 6.86 (dd, J=7.7, 1.8 Hz, 1H), 5.53 (s, 2H). Mass: 479.88 (M+). Example 56 (R)-2-(1-(9H-purin-6-ylamino) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 65 (1.00 g, 4.34 mmoles) in dichloromethane (15 ml), triethylamine (1.8 ml, 13.02 mmoles) was added followed by N-Boc-D-Alanine (0.986 g, 5.21 mmoles). To this mixture HATU (3.3 g, 8.68 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (1.70 g). To a solution of this intermediate (0.8 g) in dichloromethane (10 ml), trifluoroacetic acid (3 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (0.410 g). To a solution of this amine intermediate (0.41 g, 1.52 mmoles) in tert-butanol (7 ml), N,N-diisopropylethylamine (0.53 ml, 3.04 mmoles) and 6-bromopurine (0.242 g, 1.21 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as off-white solid (0.130 g, 21% yield). MP: 274-276° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.96 (s, 1H), 8.14-8.01 (m, 4H), 8.11 (s, 1H), 7.81 (dt, J=8.5, 1.5 Hz, 1H), 7.60 (d, J=8.4 Hz, 1H), 7.49 (m, 2H), 7.25-7.19 (m, 3H), 5.18 (br m, 1H), 1.56 (d, J=7.0 Hz, 3H). Mass: 402.04 (M++1). Example 57 (S)-2-(1-(9H-purin-6-ylamino) ethyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of intermediate 50 (2.50 g, 10.85 mmoles) in dichloromethane (25 ml), triethylamine (4.5 ml, 32.57 mmoles) was added followed by N-Boc-L-Alanine (2.46 g, 13.03 mmoles). To this mixture HATU (8.25 g, 21.71 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (1.45 g). To a solution of this intermediate (1.40 g) in dichloromethane (20 ml), trifluoroacetic acid (1.4 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (0.850 g). To a solution of this amine intermediate (0.450 g, 1.52 mmoles) in tert-butanol (7 ml), N,N-diisopropylethylamine (0.55 ml, 3.17 mmoles) and 6-chloropurine (0.194 g, 1.27 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as yellow solid (0.100 g, 15% yield). MP: 196-198° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.95 (s, 1H), 8.11-(m, 3H), 7.69 (m, 3H), 7.42 (m, 5H), 5.20 (br m, 1H), 1.54 (d, J=6.7 Hz, 3H). Mass: 402.18 (M++1). Example 57a 2-((4-Amino-3-iodo-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.404 g, 5.36 mmoles) in DMF (28 ml), potassium carbonate (1.85 g, 13.4 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 5 (2.11 g, 6.70 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (1.10 g, 41% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.18 (s, 1H), 8.06 (dd, J=8.0, 1.6 Hz, 1H), 7.77 (m, 1H), 7.50 (dt, J=8.0, 0.9 Hz, 1H), 7.41-7.30 (m, 6H), 5.44 (s, 2H). Example 57b 2-(1-(4-amino-3-iodo-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-phenyl-4H-chromen-4-one To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (6.0 g, 23 mmoles) in DMF (110 ml), potassium carbonate (7.94 g, 57.2 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 29 (9.5 g, 28.76 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (2.0 g, 17% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.12 (dd, J=7.9, 1.6 Hz, 1H), 8.10 (s, 1H), 7.91 (m, 1H), 7.68 (d, J=8.2 Hz, 1H), 7.60 (dt, J=7.9, 0.9 Hz, 1H), 7.36 (m, 3H), 7.18 (m, 2H), 5.93 (q, J=7.1 Hz, 1H), 1.91 (d, J=7.1 Hz, 3H). Example 57c 2-(1-(4-amino-3-iodo-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.30 g, 5.299 mmoles) in DMF (23 ml), potassium carbonate (1.80 g, 13.24 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (2.3 g, 6.62 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.800 g, 24% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.04 (d, J=1.6 Hz, 1H), 8.02 (s, 1H), 7.94 (s, 1H), 7.86 (dt, J=8.0, 1.5 Hz, 1H), 7.66 (d, J=8.4 Hz, 1H), 7.53 (t, J=7.5 Hz, 1H), 7.29 (m, 1H), 7.09 (dt, J=7.7, 2.4 Hz, 1H), 6.88 (m, 1H), 5.93 (q, J=7.0 Hz, 1H), 1.83 (d, J=7.1 Hz, 3H). Example 57d 2-((4-amino-3-iodo-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.0 g, 3.01 mmoles) in DMF (5 ml), N,N-Diisopropylethylamine (0.5 ml, 6.02 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 90 (1.3 g, 5.11 mmoles) was added and stirred for 16 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.351 g, 23% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.17 (s, 1H), 7.76-7.63 (m, 2H), 7.55 (dd, J=9.1, 4.2 Hz, 1H), 7.39-7.28 (m, 5H), 5.44 (s, 2H). Example 57e 2-(1-(4-amino-3-iodo-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (12.8 g, 49.03 mmoles) in DMF (50 ml), cesium carbonate (18.7 g, 57.62 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 57 (10 g, 28.81 mmoles) was added and stirred for 17 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (3.8 g, 25% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.02 (s, 1H), 7.72 (m, 3H), 7.28 (m, 3H), 7.09 (br s, 2H), 5.86 (q, J=7.1 Hz, 1H), 1.82 (d, J=7.0 Hz, 3H). Example 57f 2-(1-(4-amino-3-iodo-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (10.9 g, 41.90 mmoles) in DMF (45 ml), cesium carbonate (16.0 g, 49.30 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 75 (9.0 g, 24.65 mmoles) was added and stirred for 17 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol: dichloromethane to afford the title compound as light yellow solid (3.2 g, 24% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.01 (s, 1H), 7.81-7.69 (m, 3H), 7.28 (s, 1H), 7.08 (dt, J=8.5, 1.8 Hz, 1H), 6.88 (br s, 2H), 5.93 (q, J=7.0 Hz, 1H) 1.83 (d, J=7.0 Hz, 3H). Example 57g 2-(1-(4-amino-3-iodo-1H-pyrazolo [3,4-d] pyrimidin-1-yl) propyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.44 g, 5.52 mmoles) in DMF (20 ml), potassium carbonate (0.763 g, 5.52 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 46 (1.0 g, 2.76 mmoles) was added and stirred for 17 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.440 g, 29% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.04 (dd, J=7.9, 1.4 Hz, 1H), 8.01 (s, 1H), 7.87 (m, 1H) 7.68 (d, J=8.5 Hz, 1H), 7.53 (t, J=7.2 Hz, 1H), 7.29 (br s, 1H), 7.09 (dt, J=8.9, 1.6 Hz, 1H), 6.88 (m, 2H), 5.72 (J=7.5 Hz, 1H), 2.42 (quintet, J=7.4 Hz, 2H), 0.75 (t, J=7.3 Hz, 3H). Example 58 2-((4-amino-3-(pyridin-3-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57a (0.250 g, 0.50 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), 3-pyridinylboronic acid (0.080 g, 0.65 mmoles) and sodium carbonate (0.264 g, 2.5 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.109 g, 0.095 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.030 g, 13% yield). MP: 253-255° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.78 (d, J=1.7 Hz, 1H), 8.65 (dd, J=4.7, 1.3 Hz, 1H), 8.24 (s, 1H), 8.05 (dd, J=7.9, 1.6 Hz, 1H), 8.00 (td, J=7.9, 1.9 Hz, 1H), 7.77 (d, J=7.2, 1.7 Hz, 1H), 7.54-7.43 (m, 3H), 7.37-7.30 (m, 5H), 7.12 (br s, 2H), 5.54 (s, 2H). Mass: 447.19 (M++1). Example 59 2-((4-amino-3-(3-hydroxyprop-1-ynyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57a (0.180 g, 0.363 mmoles) in THF (5 ml), propargyl alcohol (0.051 g, 0.436 mmoles) diisopropylamine (0.31 ml, 1.81 mmoles), copper(I) iodide (7 mg, 0.036 mmoles) and) Tetrakis triphenylphosphine Palladium (0.042 g, 0.0363 mmoles) were added and the system is degassed for 30 min and heated to reflux for 4 h. The reaction mixture filtered through celite pad and washed with ethyl acetate. The filtrate was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.118 g, 77% yield). MP: 171-173° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.21 (s, 1H), 8.06 (dd, J=7.9, 1.6 Hz, 1H), 7.77 (m, 1H), 7.50 (dt, J=8.0, 0.9 Hz, 1H), 7.40-7.33 (m, 6H), 5.43 (s, 2H), 4.33 (d, J=6.1z, 2H). Mass: 423.88 (M+). Example 60 2-((4-amino-3-(1H-pyrazol-4-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57a (0.500 g, 1.00 mmoles) in DMF (7 ml), ethanol (4 ml) and water (4 ml), N-Boc-Pyrazole-4-boronic acid pinacol ester (0.445 g, 1.51 mmoles) and sodium carbonate (0.534 g, 5.04 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.229 g, 0.198 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.131 g, 29% yield). MP: 235-237° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.20 (s, 1H), 8.20 (s, 1H), 8.10 (s, 1H), 8.05 (dd, J=8.0, 1.7 Hz, 1H), 7.78 (s, 1H), 7.76 (m, 1H), 7.49 (dt, J=8.0, 0.8 Hz, 1H), 7.39-7.31 (m, 6H), 5.45 (s, 2H). Mass: 436.20 (M++1). Example 61 2-((4-amino-3-(3-(hydroxymethyl) phenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57a (0.250 g, 0.50 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), 3-hydroxymethylphenylboronic acid (0.115 g, 0.757 mmoles) and sodium carbonate (0.267 g, 2.53 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.115 g, 0.099 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.116 g, 44% yield). MP: 219-223° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.23 (s, 1H), 8.05 (dd, J=8.0, 1.6 Hz, 1H), 7.77 (m, 1H), 7.58 (s, 1H), 7.50 (m, 3H), 7.44 (d, J=8.5 Hz, 1H), 7.41-7.31 (m, 6H), 5.52 (s, 2H), 5.27 (t, J=5.8 Hz, 1H), 4.57 (d, J=5.7 Hz, 2H). Mass: 476.31 (M++1). Example 62 2-((4-amino-3-(1H-indazol-4-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57a (0.500 g, 1.00 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), 4-Indazoleboronic acid pinacol ester (0.491 g, 2.00 mmoles) and sodium carbonate (0.533 g, 5.02 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.229 g, 0.197 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.040 g, 8% yield). MP: 248-252° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.24 (s, 1H), 8.27 (s, 1H), 8.07 (dd, J=7.9, 1.6 Hz, 1H), 8.01 (s, 1H), 7.78 (m, 1H), 7.63 (d, J=8.4 Hz, 1H), 7.51-7.32 (m, 10H), 7.14 (br s, 1H), 5.56 (s, 1H). Mass: 486.04 (M++1). Example 63 2-((4-amino-3-(3-fluorophenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 78 (0.150 g, 0.654 mmoles) in DMF (5 ml), potassium carbonate (0.180 g, 1.30 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 5 (0.413 g, 1.30 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.130 g, 43% yield). MP: 244-247° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.23 (s, 1H), 8.05 (dd, J=7.9, 1.6 Hz, 1H), 7.77 (m, 1H), 7.58 (m, 1H), 7.49-7.17 (m, 10H), 5.52 (s, 2H). Mass: 463.92 (M+). Example 64 2-((4-amino-3-(3-hydroxypropyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of example 59 (0.170 g, 0.401 mmoles) in methanol (4 ml), palladium on charcoal 1 (10%, 0.050 g) was added and hydrogenated at 5 kg/cm2for 48 h. The reaction mixture filtered through celite pad and washed with methanol. The filtrate was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.072 g, 42% yield). MP: 182-184° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.11 (s, 1H), 8.05 (dd, J=8.0, 1.6 Hz, 1H), 7.76 (m, 1H), 7.49 (t, J=7.1 Hz, 1H), 7.39-7.20 (m, 8H), 4.62 (t, J=4.6 Hz, 1H), 3.45 (q, J=6.1 Hz, 2H), 2.92 (t, J=7.4 Hz, 2H), 1.78 (m, 2H). Mass: 427.87 (M+). Example 65 N-(3-(4-amino-1-((4-oxo-3-phenyl-4H-chromen-2-yl) methyl)-1H-pyrazolo [3,4-d] pyrimidin-3-yl) phenyl) acetamide To a solution of Example 57a (0.250 g, 0.50 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), 3-Acetamidophenyl boronic acid (0.116 g, 0.65 mmoles) and sodium carbonate (0.264 g, 2.50 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.109 g, 0.095 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.080 g, 23% yield). MP: 122-123° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 10.13 (s, 1H), 8.06 (dd, J=7.7, 1.4 Hz, 1H), 7.90 (s, 1H), 7.77 (m, 1H), 7.57-7.47 (m, 3H), 7.48 (m, 3H), 7.37-7.29 (m, 6H), 5.52 (s, 2H), 2.05 (s, 3H). Mass: 503.05 (M++1). Example 66 2-((4-amino-3-(3-fluoro-5-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 79 (0.150 g, 0.58 mmoles) in DMF (5 ml), potassium carbonate (0.160 g, 1.16 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 5 (0.366 g, 1.16 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.120 g, 42% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.23 (s, 1H), 8.05 (dd, J=8.1, 1.5 Hz, 1H), 7.77 (m, 1H), 7.49 (dt, J=8.1, 0.9 Hz, 1H), 7.44 (d, J=8.4 Hz, 1H), 7.38-7.30 (m, 5H), 6.98 (m, 2H), 6.96 (dt, J=7.9, 2.3 Hz, 1H), 5.51 (s, 2H), 3.81 (s, 3H). Example 66a 2-((4-amino-3-(3-fluoro-5-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 66 (0.100 g, 0.202 mmoles) in dichloromethane (15 ml), BBr3(1M in dichloromethane, 1.0 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.035 g, 36% yield). MP: 260-262° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.16 (s, 1H), 8.22 (s, 1H), 8.06 (dd, J=7.9, 1.5 Hz, 1H), 7.78 (m, 1H), 7.50 (dt, J=8.0, 1.0 Hz, 1H), 7.44 (d, J=8.5 Hz, 1H), 7.37-7.31 (m, 5H), 6.86 (t, J=1.5 Hz, 1H), 6.82 (dt, J=7.6, 2.3 Hz, 1H), 6.65 (td, J=10.9, 2.3 Hz, 1H), 5.50 (s, 2H). Mass: 480.02 (M++1). Example 67 2-((4-amino-3-(3-fluoro-5-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl)methyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 79 (0.150 g, 0.58 mmoles) in DMF (5 ml), potassium carbonate (0.160 g, 1.16 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 77 (0.366 g, 1.16 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.120 g, 42% yield). MP: 115-117° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.21 (s, 1H), 8.06 (dd, J=8.3, 1.7 Hz, 1H), 7.80 (m, 1H), 7.51 (m, 2H), 7.39 (q, J=8.0 Hz, 1H), 7.18 (m, 3H), 6.97 (m, 3H), 5.54 (s, 2H), 3.82 (s, 3H). Mass: 511.80 (M+). Example 68 2-((4-amino-3-(3-fluoro-5-hydroxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of example 67 (0.080 g, 0.156 mmoles) in dichloromethane (15 ml), BBr3(1M in dichloromethane, 0.8 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.035 g, 45% yield). MP: 235-237° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.17 (s, 1H), 8.20 (s, 1H), 8.06 (dd, J=8.2, 1.6 Hz, 1H), 7.80 (m, 1H), 7.51 (m, 2H), 7.38 (q, J=7.8 Hz, 1H), 7.17-7.07 (m, 3H), 6.84 (t, J=1.7 Hz, 1H), 6.81 (td, J=79.3, 2.1 Hz, 1H), 6.66 (td, J=10.2, 2.2 Hz, 1H), 5.53 (s, 2H). Mass: 497.87 (M+). Example 69 2-(1-(4-amino-3-(1H-pyrazol-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57b (0.400 g, 0.78 mmoles) in DMF (8 ml), ethanol (4 ml) and water (4 ml), N-Boc-pyrazole-4-boronic acid pinacol ester (0.344 g, 1.17 mmoles) and sodium carbonate (0.413 g, 3.9 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine. Palladium (0.171 g, 0.148 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.070 g, 19% yield). MP: 214-217° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.20 (s, 1H), 8.10 (s, 1H), 8.05 (s, 1H), 8.03 (dd, J=8.0, 1.6 Hz, 1H), 7.82 (m, 2H), 7.61 (d, J=8.0 Hz, 1H), 7.51 (dt, J=8.0, 0.9 Hz, 1H), (m, 3H), 7.31-6.87 (m, 5H), 5.92 (q, J=7.1 Hz, 1H), 1.87 (d, J=7.1 Hz, 3H). Mass: 449.852 (M+). Example 70 2-(1-(4-amino-3-(1H-indazol-6-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57b (0.500 g, 0.98 mmoles) in DMF (10 ml), ethanol (4 ml) and water (4 ml), 6-Indazoleboronic acid pinacol ester (0.478 g, 1.96 mmoles) and sodium carbonate (0.519 g, 4.90 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.214 g, 0.185 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.050 g, 10% yield). MP: 176-178° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.18 (s, 1H), 8.14 (s, 1H), 8.09 (s, 1H), 8.05 (dd, J=8.0, 1.6 Hz, 1H), 7.91 (d, J=8.3 Hz, 1H), 7.83 (m, 1H), 7.73 (s, 1H), 7.63 (d, J=8.3 Hz, 1H), 7.52 (dt, J=7.9, 0.8 Hz, 1H), 7.41 (dd, J=8.3, 1.2 Hz, 1H), 7.31-7.16 (m, 5H), 6.01 (q, J=6.9 Hz, 1H), 1.92 (d, J=7.1 Hz, 3H). Mass: 500.04 (M++1). Example 71 2-(1-(4-amino-3-(3-hydroxy-3-methylbut-1-ynyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57b (0.500 g, 0.981 mmoles) in THF (14 ml), 2-Methyl-3-butyn-2-ol (0.1 ml, 1.178 mmoles) diisopropylamine (0.70 ml, 4.90 mmoles), copper(I) iodide (18.6 mg, 0.098 mmoles) and) Tetrakistriphenylphosphine Palladium (0.113 g, 0.098 mmoles) were added and the system is degassed for 30 min. and heated to reflux for 4 h. The reaction mixture filtered through celite pad and washed with ethyl acetate. The filtrate was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.311 g, 68% yield). MP: 109-113° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.05 (m, 3H), 7.83 (dt, J=8.6, 1.5 Hz, 1H), 7.61 (d, J=8.4 Hz, 1H), 7.52 (t, J=7.2 Hz, 1H), 7.30-7.11 (m, 4H), 5.84 (q, J=7.1z, 1H) 5.74 (s, 1H), 1.82 (d, J=7.0 Hz, 3H), 1.46 (s, 6H). Mass: 466.09 (M++1). Example 72 2-(1-(4-amino-3-(1H-pyrazol-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.758 mmoles) in DMF (5.3 ml), ethanol (2.7 ml) and water (2.7 ml), N-Boc-pyrazole-4-boronic acid pinacol ester (0.334 g, 1.137 mmoles) and sodium carbonate (0.401 g, 3.79 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.172 g, 0.149 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.040 g, 11% yield). MP: 223-226° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.22 (s, 1H), 8.03 (m, 2H), 7.85 (m, 2H), 7.68 (d, J=8.3 Hz, 1H), 7.52 (t, J=7.7 Hz, 1H), 7.25 (m, 1H), 7.07-6.93 (m, 3H), 5.92 (q, J=6.9 Hz, 1H), 1.87 (d, J=7.0 Hz, 3H). Mass: 467.84 (M+). Example 73 (S)-2-(1-(9H-purin-6-ylamino)ethyl)-3-phenyl-4H-chromen-4-one To a solution of intermediate 59 (2.0 g, 9.42 mmoles) in dichloromethane (20 ml), triethylamine (3.9 ml, 28.26 mmoles) was added followed by N-Boc-Alanine (1.90 g, 10.42 mmoles). To this mixture HATU (6.6 g, 17.37 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (1.70 g). To a solution of this intermediate (1.7 g) in dichloromethane (20 ml), trifluoroacetic acid (3 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (0.641 g). To a solution of this amine intermediate (0.30 g, 1.05 mmoles) in tert-butanol (6 ml), N,N-diisopropylethylamine (0.36 ml, 2.17 mmoles) and 6-bromopurine (0.168 g, 0.847 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as off-white solid (0.041 g, 10% yield). MP: 135-138° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.95 (s, 1H), 8.15 (t, J=6.8 Hz, 1H), 8.11 (s, 1H), 8.08 (s, 1H), 8.03 (d, J=7.8 Hz, 1H), 7.81 (t, J=7.3 Hz, 1H), 7.60 (d, J=8.3 Hz, 1H), 7.49 (t, J=7.3 Hz, 2H), 7.25 (m, 3H), 5.19 (br m, 1H), 1.56 (d, J=6.9 Hz, 3H). Mass: 384.12 (M++1). Example 74 (S)-2-(1-(9H-purin-6-ylamino)ethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 73 (2.0 g, 8.05 mmoles) in dichloromethane (20 ml), triethylamine (3.3 ml, 24.17 mmoles) was added followed by N-Boc-L-Alanine (1.82 g, 9.66 mmoles). To this mixture HATU (6.12 g, 16.11 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (2.15 g). To a solution of this intermediate (2.1 g) in dichloromethane (20 ml), trifluoroacetic acid (4 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (0.700 g). To a solution of this amine intermediate (0.450 g, 1.49 mmoles) in tert-butanol (7 ml), N,N-diisopropylethylamine (0.52 ml, 2.98 mmoles) and 6-chloropurine (0.184 g, 1.194 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as off-white solid (0.060 g, 12% yield). MP: 203-206° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.96 (s, 1H), 8.15 (m, 2H), 8.08 (s, 1H), 7.70 (m, 3H), 7.49 (q, J=7.3 Hz, 1H), 7.24 (m, 3H), 5.18 (br m, 1H), 1.55 (d, J=7.1 Hz, 3H). Mass: 420.17 (M++1). Example 75 2-((4-amino-3-(1H-indazol-6-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57a (0.700 g, 1.40 mmoles) in DMF (7 ml), ethanol (3.2 ml) and water (3.2 ml), 6-Indazoleboronic acid pinacol ester (0.687 g, 2.81 mmoles) and sodium carbonate (0.745 g, 7.03 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.320 g, 0.277 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.020 g, 3% yield). MP: 140-143° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.18 (s, 1H), 8.24 (s, 1H), 8.13 (s, 1H), 8.06 (dd, J=7.9, 1.6 Hz, 1H), 7.78 (m, 2H), 7.49-7.30 (m, 7H), 6.89 (q, J=7.7 Hz, 1H), 5.53 (s, 2H). Mass: 485.76 (M++1). Example 76 2-(1-(4-amino-3-(3-fluoro-5-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 79 (0.160 g, 0.617 mmoles) in DMF (6 ml), potassium carbonate (0.171 g, 1.16 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.429 g, 1.23 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.160 g, 49% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.08 (s, 1H), 8.04 (dd, J=8.0, 1.6 Hz, 1H), 7.85 (m, 1H), 7.68 (d, J=8.2 Hz, 1H), 7.53 (dt, J=7.9, 0.9 Hz, 1H), 7.31 (br s, 1H), 7.07 (dt, J=8.6, 2.1 Hz, 1H), 6.97 (m, 5H), 6.03 (q, J=7.1 Hz, 1H), 3.82 (s, 3H), 1.90 (d, J=7.0 Hz, 3H). Example 76a 2-(1-(4-amino-3-(3-fluoro-5-hydroxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 76 (0.160 g, 0.304 mmoles) in dichloromethane (25 ml), BBr3(1M in dichloromethane, 1.6 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.080 g, 51% yield). MP: 271-273° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.17 (s, 1H), 8.06 (s, 1H), 8.05 (dd, J=7.9, 1.4 Hz, 1H), 7.86 (dt, J=8.5, 1.5 Hz, 1H), 7.68 (d, J=8.3 Hz, 1H), 7.53 (t, J=7.9 Hz, 1H), 7.28 (br s, 1H), 7.05 (dt, J=6.8, 2.0 Hz, 1H), 6.91 (br s, 2H), 6.86 (s, 1H), 6.79 (d, J=9.4 Hz, 1H), 6.66 (td, J=10.3, 2.1 Hz, 1H), 6.05 (q, J=6.7 Hz, 1H), 1.88 (d, J=7.1 Hz, 3H). Mass: 511.80 (M+). Example 77 2-(1-(4-amino-3-(1H-indazol-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.350 g, 1.00 mmoles) in DMF (8 ml), ethanol (4 ml) and water (4 ml), 4-Indazoleboronic acid pinacol ester (0.322 g, 1.32 mmoles) and sodium carbonate (0.349 g, 3.3 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine Palladium (0.150 g, 0.130 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.045 g, 13% yield). MP: 231-233° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.25 (s, 1H), 8.10 (s, 1H), 8.06 (m, 2H), 7.86 (m, 1H), 7.66 (t, J=9.0 Hz, 2H), 7.54 (m, 2H), 7.33 (t, J=6.7 Hz, 2H), 7.11-7.06 (m, 3H), 6.07 (q, J=7.1 Hz, 1H), 1.94 (d, J=7.0 Hz, 3H). Mass: 517.96 (M+). Example 78 2-(1-(4-amino-3-(3,5-dimethyl-1H-pyrazol-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.350 g, 0.661 mmoles) in DMF (6 ml), ethanol (3 ml) and water (3 ml), 3,5-Dimethylpyrazole-4-boronic acid pinacol ester (0.191 g, 0.859 mmoles) and sodium carbonate (0.350 g, 3.30 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.150 g, 0.130 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.025 g, 7% yield). MP: 240-243° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.44 (s, 1H), 8.04 (dd, J=8.0, 1.6 Hz, 1H), 8.01 (s, 1H), 7.85 (s, 1H), 7.61 (d, J=8.3 Hz, 1H), 7.52 (dt, J=7.9, 0.7 Hz, 1H), 7.33 (br m, 1H), 7.12-6.95 (m, 3H), 5.97 (q, J=7.0 Hz, 1H), 2.09 (s, 6H), 1.86 (d, J=7.0 Hz, 3H). Mass: 495.84 (M+). Example 79 2-(1-(4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.758 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 3-Methylindazole-6-boronic acid pinacol ester 97 (0.391 g, 1.517 mmoles) and sodium carbonate (0.401 g, 3.79 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.172 g, 0.149 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.095 g, 23% yield). MP: 214-217° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.08 (s, 1H), 8.05 (dd, J=7.9, 1.4 Hz, 1H), 7.86 (m, 2H), 7.68 (d, J=8.3 Hz, 1H), 7.62 (s, 1H), 7.53 (t, J=7.3 Hz, 1H), 7.33 (d, J=8.5 Hz, 1H), 7.31 (br s, 1H), 7.07 (dt, J=8.9, 2.1 Hz, 1H), 6.93 (m, 2H), 6.07 (q, J=6.7 Hz, 1H), 2.51 (s, 3H), 1.91 (d, J=7.0 Hz, 3H). Mass: 532.03 (M++1). Example 80 2-(1-(4-amino-3-(1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.500 g, 0.758 mmoles) in DMF (4.5 ml), ethanol (2.3 ml) and water (2.3 ml), Indazole-6-boronic acid pinacol ester (0.462 g, 1.89 mmoles) and sodium carbonate (0.502 g, 4.74 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.215 g, 0.186 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.080 g, 16% yield). MP: 206-208° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.19 (s, 1H), 8.14 (s, 1H), 8.08 (s, 1H), 8.05 (dd, J=7.9, 1.5 Hz, 1H), 7.90 (d, J=8.3 Hz, 1H), 7.86 (m, 1H), 7.71 (s, 1H), 7.69 (d, J=8.4 Hz, 1H), 7.53 (t, J=7.1 Hz, 1H), 7.39 (dd, J=8.2, 1.1 Hz, 1H), 7.30 (m, 2H), 7.07 (dt, J=8.7, 2.6 Hz, 1H), 6.92 (br m, 2H), 6.06 (q, J=7.1 Hz, 1H), 1.91 (d, J=7.0 Hz, 3H). Mass: 517.96 (M+). Example 81 2-(1-(4-amino-3-(2-(hydroxymethyl) phenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.300 g, 0.568 mmoles) in DMF (3 ml), ethanol (1.5 ml) and water (1.5 ml), 2-Hydroxymethylphenylboronic acid (0.173 g, 1.137 mmoles) and sodium carbonate (0.301 g, 2.844 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.129 g, 0.112 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.090 g, 31% yield). MP: 185-189° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.09 (s, 1H), 8.04 (dd, J=7.9, 1.4 Hz, 1H), 7.84 (m, 1H), 7.66-7.35 (m, 10H), 7.17 (dt, J=10.8, 1.4 Hz, 1H), 7.04 (m, 1H), 6.01 (q, J=6.7 Hz, 1H), 5.13 (t, J=5.7 Hz, 1H), 4.54 (m, 2H), 1.87 (d, J=7.1 Hz, 3H). Mass: 508.16 (M++1). Example 82 2-(1-(4-amino-3-(4-fluoro-3-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 80 (0.120 g, 0.617 mmoles) in DMF (6 ml), potassium carbonate (0.128 g, 0.925 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.323 g, 1.23 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.075 g, 31% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.07 (s, 1H), 8.04 (d, J=7.0 Hz, 1H), 7.85 (t, J=7.1 Hz, 1H), 7.68 (d, J=8.5 Hz, 1H), 7.61 (m, 1H), 7.53 (t, J=7.1 Hz, 1H), 7.36 (m, 2H), 7.16 (m, 1H), 7.07 (t, J=6.7 Hz, 1H), 6.93 (br s, 2H), 6.03 (q, J=7.0 Hz, 1H), 3.88 (s, 3H), 1.90 (d, J=7.0 Hz, 3H). Example 82a 2-(1-(4-amino-3-(4-fluoro-3-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 82 (0.075 g, 0.142 mmoles) in dichloromethane (15 ml), BBr3(1M in dichloromethane, 1 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N Hal solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale green solid (0.040 g, 55% yield). MP: 241-244° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.15 (s, 1H), 8.05 (s, 1H), 8.05 (dd, J=8.6, 1.5 Hz, 1H), 7.86 (m, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.53 (t, J=7.4 Hz, 1H), 7.28 (m, 2H), 7.20 (dd, J=8.5, 1.9 Hz, 1H), 7.05 (m, 4H), 6.04 (q, J=7.1 Hz, 1H), 1.88 (d, J=7.1 Hz, 3H). Mass: 511.94 (M+). Example 83 2-(1-(4-amino-3-(3-hydroxyprop-1-ynyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example—57c (0.400 g, 0.755 mmoles) in THF (10 ml), propargyl alcohol (0.051 g, 0.906 mmoles) diisopropylamine (0.53 ml, 3.77 mmoles), copper (I) iodide (14 mg, 0.075 mmoles) and) Tetrakis triphenylphosphine Palladium (0.087 g, 0.075 mmoles) were added and the system is degassed for 30 min and heated to reflux for 4 h. The reaction mixture filtered through celite pad and washed with ethyl acetate. The filtrate was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.106 g, 23% yield). MP: 171-173° C.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 11.36 (s, 1H), 8.19 (dd, J=7.9, 1.2 Hz, 1H), 7.70 (dt, J=8.6, 1.5 Hz, 1H), 7.47 (d, J=8.4 Hz, 1H), 7.42 (t, J=7.5 Hz, 1H), 7.38 (m, 2H), 7.07 (t, J=8.2 Hz, 1H), 6.99 (m, 2H), 6.00 (q, J=7.0 Hz, 1H), 4.55 (s, 2H), 1.97 (d, J=7.1 Hz, 1H). Mass: 456.08 (M++1). Example 84 2-(1-(4-amino-3-(3-fluoro-4-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 81 (0.130 g, 0.50 mmoles) in DMF (4 ml), potassium carbonate (0.139 g, 1.00 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.350 g, 1.00 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.163 g, 60% yield). MP: 222-224° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.06 (s, 1H), 8.04 (dd, J=7.9, 1.5 Hz, 1H), 7.85 (m, 1H), 7.68 (dd, J=8.4 Hz, 1H), 7.52 (t, J=7.4 Hz, 1H), 7.37-7.28 (m, 4H), 7.07 (dt, J=8.9, 2.4 Hz, 1H), 6.93 (br s, 2H), 6.05 (q, J=7.1 Hz, 1H), 3.89 (s, 3H), 1.89 (d, J=7.0 Hz, 3H). Mass: 525.94 (M+). Example 85 2-(1-(4-amino-3-(3-fluoro-4-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of example 84 (0.100 g, 0.190 mmoles) in dichloromethane (4 ml), BBr3(1M in dichloromethane, 1 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale green solid (0.061 g, 63% yield). MP: 244-247° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.19 (s, 1H), 8.04 (s, 1H), 8.04 (dd, J=8.0, 1.4 Hz, 1H), 7.85 (m, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.52 (t, J=7.2 Hz, 1H), 7.33 (m, 2H), 7.24 (dd, J=8.2, 1.4 Hz, 1H), 7.09-6.91 (m, 4H), 6.00 (q, J=7.0 Hz, 1H), 1.88 (d, J=7.0 Hz, 1H). Mass: 511.94 (M+). Example 86 2-(1-(4-amino-3-(3-chloro-5-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 82 (0.100 g, 0.362 mmoles) in DMF (4 ml), potassium carbonate (0.100 g, 0.725 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.252 g, 0.725 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.132 g, 67% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.08 (s, 1H), 8.04 (d, J=6.8 Hz, 1H), 7.85 (m, 1H), 7.67 (d, J=8.4 Hz, 1H), 7.52 (t, J=7.5 Hz, 1H), 7.28 (br s, 1H), 7.17 (s, 1H), 7.12 (s, 1H), 7.05-6.94 (m, 4H), 6.03 (q, J=7.0 Hz, 1H), 3.82 (s, 3H), 1.90 (d, J=7.0 Hz, 3H). Example 86a 2-(1-(4-amino-3-(3-chloro-5-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 86 (0.100 g, 0.184 mmoles) in dichloromethane (4 ml), BBr3(1M in dichloromethane, 1 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale green solid (0.032 g, 33% yield). MP: 122-124° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.19 (s, 1H), 8.06 (s, 1H), 8.04 (dd, J=7.9, 1.5 Hz, 1H), 7.86 (m, 1H), 7.67 (d, J=8.3 Hz, 1H), 7.53 (t, J=7.1 Hz, 1H), 7.28 (br s, 1H), 7.06-6.87 (m, 6H), 6.03 (q, J=6.9 Hz, 1H), 1.88 (d, J=7.1 Hz, 1H). Mass: 528.11 (M++1). Example 87 2-(1-(4-amino-3-(3-(trifluoromethoxy) phenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 83 (0.200 g, 0.677 mmoles) in DMF (8 ml), potassium carbonate (0.187 g, 1.354 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.472 g, 1.354 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as Off-white solid (0.058 g, 15% yield). MP: 155-157° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.09 (s, 1H), 8.04 (dd, J=6.7, 1.3 Hz, 1H), 7.86 (m, 1H), 7.68-7.45 (m, 8H), 7.28 (br s, 1H), 7.03-6.91 (m, 3H), 6.06 (q, J=7.2 Hz, 1H), 1.90 (d, J=7.1 Hz, 3H). Mass: 562.13 (M++1). Example 88 2-(1-(4-amino-3-(4-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 84 (0.200 g, 0.829 mmoles) in DMF (4 ml), potassium carbonate (0.229 g, 1.658 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.576 g, 1.658 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.180 g, 43% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.05 (m, 2H), 7.85 (m, 1H), 7.68 (dd, J=8.4, 5.7 Hz, 1H), 7.54 (m, 3H), 7.28 (br s, 1H), 7.09-6.90 (m, 5H), 6.01 (q, J=7.0 Hz, 1H), 3.82 (s, 3H), 1.89 (d, J=7.1 Hz, 3H). Example 88a 2-(1-(4-amino-3-(4-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 88 (0.150 g, 0.295 mmoles) in dichloromethane (4 ml), BBr3(1M in dichloromethane, 1.5 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.048 g, 33% yield). MP: 244-247° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.79 (s, 1H), 8.04 (s, 1H), 8.04 (dd, J=8.5, 1.4 Hz, 1H), 7.85 (m, 1H), 7.68 (d, J=8.5 Hz, 1H), 7.53 (t, J=7.6 Hz, 1H), 7.42 (d, J=8.5 Hz, 2H), 7.28 (br s, 1H), 7.06 (t, J=8.5 Hz, 1H), 6.91 (d, J=8.5 Hz, 2H), 6.91 (br s, 2H), 6.00 (q, J=7.1 Hz, 1H), 1.88 (d, J=7.0 Hz, 3H). Mass: 492.69 (M++1). Example 89 2-((6-amino-9H-purin-9-yl)methyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Adenine (0.243 g, 1.80 mmoles) in DMF (5 ml), potassium carbonate (0.248 g, 1.80 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 77 (0.300 g, 0.900 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.080 g, 23% yield). MP: 224-227° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.12 (s, 1H), 8.07 (s, 1H), 8.05 (dd, J=7.7, 1.2 Hz, 1H), 7.79 (m, 1H), 7.55 (m, 3H), 7.28-7.21 (m, 5H), 5.36 (s, 2H). Mass: 388.04 (M++1). Example 90 2-(1-(4-amino-3-(4-fluoro-2-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 85 (0.120 g, 0.462 mmoles) in DMF (6 ml), potassium carbonate (0.127 g, 0.924 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.321 g, 0.924 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.080 g, 33% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.04 (d, J=6.6 Hz, 1H), 8.00 (s, 1H), 7.85 (t, J=8.7 Hz, 1H), 7.66-7.49 (m, 4H), 7.38 (t, J=7.3 Hz, 1H), 7.29 (br s, 1H), 7.08-6.85 (m, 5H), 5.99 (q, J=7.0 Hz, 1H), 3.77 (s, 3H), 1.87 (d, J=7.1 Hz, 3H). Example 90a 2-(1-(4-amino-3-(4-fluoro-2-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 90 (0.080 g, 0.152 mmoles) in dichloromethane (4 ml), BBr3(1M in dichloromethane, 0.8 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.027 g, 35% yield). MP: 235-237° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.66 (s, 1H), 8.04 (d, J=9.8 Hz, 1H), 8.02 (s, 1H), 7.84 (t, J=7.0 Hz, 1H), 7.66 (d, J=8.3 Hz, 1H), 7.52 (t, J=7.9 Hz, 1H), 7.35 (t, J=7.2 Hz, 2H), 7.10 (t, J=8.4 Hz, 1H), 6.96 (br s, 2H), 6.79 (m, 2H), 5.98 (q, J=7.0 Hz, 1H), 1.88 (d, J=7.1 Hz, 3H). Mass: 512.22 (M++1). Example 91 2-((4-amino-3-(3-aminophenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57a (0.400 g, 0.804 mmoles) in DMF (10 ml), ethanol (5 ml) and water (5 ml), 3-Acetamidophenyl boronic acid (0.187 g, 1.045 mmoles) and sodium carbonate (0.426 g, 4.02 mmoles) were added and the system is degassed for 30 min. Palladium tetrakis triphenylphosphine (0.183 g, 0.158 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. To the concentrate ethanol (5 ml) and Con·HCl (0.5 ml) were added and refluxed for 2 h. The reaction mixture was basified with sodium carbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.140 g, 38% yield). MP: 157-159° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.21 (s, 1H), 8.06 (dd, J=7.8, 1.3 Hz, 1H), 7.77 (dt, J=8.6, 1.5 Hz, 1H), 7.49 (t, J=7.4 Hz, 1H), 7.37-7.29 (m, 5H), 7.17 (t, J=7.7 Hz, 1H), 6.84 (s, 1H), 6.71 (d, J=7.5 Hz, 1H), 6.65 (d, J=7.9 Hz, 1H), 5.51 (s, 2H), 5.34 (s, 2H). Mass: 460.84 (M+). Example 92 2-((4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57a (0.462 g, 0.930 mmoles) in DMF (6 ml), ethanol (3 ml) and water (3 ml), N-Boc-3-methylindazole-6-boronic acid pinacol ester 98 (0.500 g, 1.39 mmoles) and sodium carbonate (0.295 g, 2.79 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.057 g, 0.046 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.120 g, 26% yield). MP: 2924-295° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.74 (s, 1H), 8.24 (s, 2H), 8.06 (dd, J=7.9, 1.5 Hz, 1H), 7.84 (d, J=8.3 Hz, 1H), 7.75 (m, 1H), 7.63 (s, 1H), 7.49 (t, J=7.3 Hz, 1H), 7.45 (d, J=8.3 Hz, 1H), 7.38-7.32 (m, 6H), 5.53 (s, 2H), 2.51 (s, 3H). Mass: 499.90 (M+). Example 93 2-(1-(4-amino-3-(2-aminopyrimidin-5-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.350 g, 0.663 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 2-aminopyrimidine-5-boronic acid (0.184 g, 1.327 mmoles) and sodium carbonate (0.351 g, 3.318 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.151 g, 0.130 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.045 g, 14% yield). MP: 264-268° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.38 (s, 2H), 8.05 (s, 1H), 8.03 (d, J=7.9, Hz, 1H), 7.85 (m, 1H), 7.68 (d, J=8.3 Hz, 1H), 7.52 (t, J=7.3 Hz, 1H), 7.29 (br s, 1H), 7.07-6.93 (m, 5H), 5.99 (q, J=7.0 Hz, 1H), 1.88 (d, J=7.0 Hz, 3H). Mass: 494.86 (M+). Example 94 2-(1-(4-amino-3-(1H-indol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.350 g, 0.663 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 6-indoleboronic acid pinacol ester (0.213 g, 1.327 mmoles) and sodium carbonate (0.351 g, 3.318 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.151 g, 0.130 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.050 g, 15% yield). MP: 222-225° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 11.27 (s, 1H), 8.06 (s, 1H), 8.05 (dd, J=8.0, 1.6 Hz, 1H), 7.86 (m, 1H), 7.69 (d, J=7.2 Hz, 2H), 7.62 (s, 1H), 7.53 (t, J=8.1 Hz, 1H), 7.45 (t, J=2.8 Hz, 1H), 7.28 (m, 2H), 7.06-6.89 (m, 3H), 6.50 (s, 1H), 6.04 (q, J=7.1 Hz, 1H), 1.91 (d, J=7.1 Hz, 3H). Mass: 516.84 (M+). Example 95 2-(1-(4-amino-3-(4-chloro-3-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 86 (0.90 g, 0.3262 mmoles) in DMF (3 ml), potassium carbonate (0.090 g, 0.653 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.227 g, 0.653 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as green solid (0.055 g, 31% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.08 (s, 1H), 8.04 (dd, J=8.0, 1.6 Hz, 1H), 7.85 (m, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.55 (m, 2H), 7.29 (br s, 1H), 7.25 (d, J=1.7 Hz, 1H), 7.19 (dd, J=8.1, 1.8 Hz, 1H), 7.08 (dt, J=8.8, 2.4 Hz, 1H), 6.92 (br s, 2H), 6.02 (q, J=7.0 Hz, 1H), 3.90 (s, 3H), 1.90 (d, J=7.1 Hz, 3H). Example 95a 2-(1-(4-amino-3-(4-chloro-3-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 95 (0.055 g, 0.1012 mmoles) in dichloromethane (4 ml), BBr3(1M in dichloromethane, 0.5 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale green solid (0.025 g, 86% yield). MP: 134-136° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.50 (s, 1H), 8.18 (s, 1H), 8.05 (d, J=8.0 Hz, 1H), 7.87 (t, J=7.0 Hz, 1H), 7.64 (d, J=8.4 Hz, 1H), 7.54 (t, J=7.6 Hz, 1H), 7.50 (d, J=8.0 Hz, 2H), 7.29 (br s, 1H), 7.21 (d, J=1.6 Hz, 1H), 7.07-6.93 (m, 4H), 6.07 (q, J=6.9 Hz, 1H), 1.90 (d, J=7.0 Hz, 3H). Mass: 527.76 (M+). Example 96 2-(1-(4-amino-3-(2-chloro-5-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 87 (0.060 g, 0.217 mmoles) in DMF (2 ml), potassium carbonate (0.060 g, 0.435 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.151 g, 0.435 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as green solid (0.030 g, 30% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.05 (s, 1H), 8.04 (dd, J=7.9, 1.3 Hz, 1H), 7.85 (m, 1H), 7.63 (d, J=8.1 Hz, 2H), 7.55 (m, 2H), 7.32 (br s, 1H), 7.18 (m, 2H), 7.00 (d, J=3.0 Hz, 1H), 6.99 (br s, 1H), 6.02 (q, J=7.0 Hz, 1H), 3.90 (s, 3H), 1.90 (d, J=7.1 Hz, 3H). Example 96a 2-(1-(4-amino-3-(2-chloro-5-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 96 (0.030 g, 0.055 mmoles) in dichloromethane (3 ml), BBr3(1M in dichloromethane, 0.27 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated to afford the title compound as pale green solid (0.018 g, 62% yield). MP: 192-195° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.95 (s, 1H), 8.15 (s, 1H), 8.05 (dd, J=7.9, 1.2 Hz, 1H), 7.86 (m, 1H), 7.65-7.49 (m, 4H), 7.39 (d, J=8.7 Hz, 1H), 7.35 (br s, 1H), 7.11 (t, J=7.5 Hz, 1H), 6.97 (dd, J=7.6, 3.2 Hz, 1H), 6.86 (d, J=2.8 Hz, 1H), 6.07 (q, J=6.9 Hz, 1H), 1.88 (d, J=7.0 Hz, 3H). Mass: 527.90 (M+). Example 97 2-(1-(4-amino-3-(3, 4-dimethoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 88 (0.220 g, 0.808 mmoles) in DMF (8 ml), potassium carbonate (0.223 g, 1.61 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.562 g, 1.61 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.163 g, 60% yield). MP: 232-235° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.05 (s, 1H), 8.04 (dd, J=8.0, 1.5 Hz, 1H), 7.85 (m, 1H), 7.68 (dd, J=8.4 Hz, 1H), 7.52 (t, J=7.2 Hz, 1H), 7.29 (br s, 1H), 7.13-6.93 (m, 6H), 6.01 (q, J=7.1 Hz, 1H), 3.80 (s, 6H), 1.90 (d, J=7.1 Hz, 3H). Mass: 538.05 (M++1). Example 98 2-(1-(4-amino-3-(3, 4-dihydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of example 97 (0.180 g, 0.0.335 mmoles) in dichloromethane (10 ml), BBr3(1M in dichloromethane, 1.8 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated to afford the title compound as off-white pale solid (0.040 g, 24% yield). MP: 193-195° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.27 (s, 1H), 9.22 (s, 1H), 8.05 (dd, J=7.3, 1.4 Hz, 1H), 8.03 (s, 1H), 7.86 (m, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.53 (t, J=7.7 Hz, 1H), 7.35 (s, 1H), 7.27 (br s, 1H), 7.05-6.86 (m, 5H), 6.02 (q, J=7.0 Hz, 1H), 1.87 (d, J=7.0 Hz, 3H). Mass: 509.84 (M+). Example 99 2-((4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.477 g, 0.930 mmoles) in DMF (5.3 ml), ethanol (2.6 ml) and water (2.6 ml), N-Boc-3-methyl-6-indazoleboronic acid pinacol ester 98 (0.500 g, 1.395 mmoles) and sodium carbonate (0.295 g, 3.318 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.053 g, 0.046 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.100 g, 20% yield). MP: 246-248° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.22 (s, 1H), 8.06 (dd, J=8.5, 1.8 Hz, 1H), 7.84 (d, J=8.3 Hz, 1H), 7.80 (m, 1H), 7.62 (s, 1H), 7.51 (d, J=8.2 Hz, 2H), 7.39-7.31 (m, 2H), 7.18 (m, 2H), 7.12 (dt, J=8.3, 2.6 Hz, 1H), 5.56 (s, 2H), 2.51 (s, 3H). Mass: 517.51 (M+). Example 100 2-(1-(4-amino-3-(1H-indol-5-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.350 g, 0.663 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 5-indoleboronic acid pinacol ester (0.213 g, 1.327 mmoles) and sodium carbonate (0.351 g, 3.318 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.151 g, 0.130 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.044 g, 13% yield). MP: 197-199° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 11.30 (s, 1H), 8.06 (s, 1H), 8.05 (dd, J=7.9, 1.2 Hz, 1H), 7.85 (m, 1H), 7.77 (s, 1H), 7.69 (d, J=8.4 Hz, 1H), 7.55 (m, 2H), 7.44 (t, J=2.8 Hz, 1H), 7.35 (m, 2H), 7.09-6.94 (m, 3H), 6.54 (m, 1H), 6.05 (q, J=7.0 Hz, 1H), 1.91 (d, J=7.0 Hz, 3H). Mass: 516.91 (M+). Example 101 2-(1-(4-Amino-3-(3-methyl-1H-indol-5-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.757 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), 3-methyl-5-indoleboronic acid pinacol ester (0.292 g, 1.136 mmoles) and sodium carbonate (0.240 g, 2.272 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.040 g, 13% yield). MP: 171-173° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 10.96 (s, 1H), 8.06 (s, 1H), 8.04 (dd, J=7.9, 1.4 Hz, 1H), 7.85 (m, 1H), 7.68 (d, J=8.1 Hz, 2H), 7.52 (d, J=7.2 Hz, 1H), 7.49 (d, J=8.3 Hz, 1H), 7.32 (dd, J=8.2, 1.4 Hz, 2H), 7.20 (s, 1H), 7.08 (dt, J=11.2, 2.7 Hz, 1H), 6.93 (br s, 2H), 6.04 (q, J=7.0 Hz, 1H), 2.28 (s, 3H), 1.92 (d, J=7.0 Hz, 3H). Mass: 530.98 (M+). Example 102 tert-butyl (5-(4-amino-1-(1-(3-(3-fluorophenyl)-4-oxo-4H-chromen-2-yl) ethyl)-1H-pyrazolo [3,4-d] pyrimidin-3-yl) thiophen-2-yl) methylcarbamate To a solution of Example 57c (0.300 g, 0.566 mmoles) in dioxane (4 ml), 2-N-Boc-aminomethylthiophene-5-boronic acid (0.186 g, 0.725 mmoles) and potassium acetate (0.168 g, 1.887 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.052 g, 0.045 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture filtered through celite and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.070 g, 20% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.06 (s, 1H), 8.05 (dd, J=8.0, 1.6 Hz, 1H), 7.85 (m, 1H), 7.65 (d, J=8.5 Hz, 1H), 7.55 (m, 3H), 7.32-7.22 (m, 3H), 7.12 (m, 2H), 6.98 (d, J=3.5 Hz, 1H), 6.92 (br s, 1H), 5.99 (q, J=7.1 Hz, 1H), 4.29 (d, J=6.1 Hz, 2H), 1.87 (d, J=7.0 Hz, 3H). Example 102a 2-(1-(4-amino-3-(5-(aminomethyl) thiophen-2-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 102 (0.070 g, 0.114 mmoles) in dichloromethane (3 ml), TFA (0.1 ml) was added under nitrogen atmosphere stirred at room temperature. After 3 h, the reaction mixture was concentrated, neutralised with sodium bicarbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.030 g, 51% yield). MP: 275-278° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.06 (s, 1H), 8.05 (dd, J=7.9, 1.5 Hz, 1H), 7.85 (m, 1H), 7.66 (d, J=8.3 Hz, 1H), 7.53 (t, J=7.2 Hz, 1H), 7.28 (m, 2H), 7.09 (d, J=3.5 Hz, 1H), 7.05 (dt, J=8.7, 2.4 Hz, 1H), 6.92 (br s, 2H), 6.02 (q, J=7.1 Hz, 1H), 4.03 (s, 2H), 1.87 (d, J=7.1 Hz, 3H). Mass: 513.27 (M++1). Example 103 2-((4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of Example 57d (0.300 g, 0.584 mmoles) in DMF (3 ml), ethanol (1.5 ml) and water (1.5 ml), N-Boc-3-methyl-6-indazoleboronic acid pinacol ester 98 (0.314 g, 0.877 mmoles) and sodium carbonate (0.185 g, 1.754 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.033 g, 0.029 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.012 g, 4% yield). MP: 277-279° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.23 (s, 1H), 7.84 (d, J=8.2 Hz, 1H), 7.74 (dd, J=8.2, 3.0 Hz, 1H), 7.66 (m, 2H), 7.59 (dd, J=9.2, 4.2 Hz, 1H), 7.38-7.32 (m, 6H), 6.54 (s, 2H), 2.51 (s, 3H). Mass: 518.17 (M++1). Example 104 2-(1-(4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-phenyl-4H-chromen-4-one To a solution of Example 57b (0.350 g, 0.684 mmoles) in DMF (3.5 ml), ethanol (1.7 ml) and water (1.7 ml), 3-methyl-6-indazoleboronic acid pinacol ester 97 (0.353 g, 1.369 mmoles) and sodium carbonate (0.217 g, 2.05 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.040 g, 0.034 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.073 g, 21% yield). MP: 249-252° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.09 (s, 1H), 8.05 (dd, J=8.0, 1.6 Hz, 1H), 7.85 (d, J=8.2 Hz, 1H), 7.81 (m, 1H), 7.64 (s, 1H), 7.62 (d, J=8.4 Hz, 1H), 7.51 (t, J=7.3 Hz, 1H), 7.36 (dd, J=9.3, 1.0 Hz, 1H), 7.29 (m, 3H), 7.15 (br s, 2H), 6.01 (q, J=7.0 Hz, 1H), 2.52 (s, 3H), 1.92 (d, J=7.0 Hz, 3H). Mass: 514.18 (M++1). Example 105 2-(1-(4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of Example 57e (0.400 g, 0.758 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 3-methyl-6-indazoleboronic acid pinacol ester 97 (0.391 g, 1.517 mmoles) and sodium carbonate (0.241 g, 2.27 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.044 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.065 g, 15% yield). MP: 253-255° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.08 (s, 1H), 7.85 (d, J=8.3 Hz, 1H), 7.75 (m, 3H), 7.63 (s, 1H), 7.35 (dd, J=8.4, 1.2 Hz, 1H), 7.28 (m, 3H), 7.14 (br s, 2H), 6.00 (q, J=7.1 Hz, 1H), 2.52 (s, 3H), 1.91 (d, J=7.1 Hz, 3H). Mass: 532.03 (M++1). Example 106 2-(1-(4-amino-3-(3-methyl-1H-indazol-5-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.350 g, 0.663 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), N-Boc-3-methyl, 5-indazoleboronic acid pinacol ester (0.356 g, 0.994 mmoles) and sodium carbonate (0.210 g, 0.98 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.038 g, 0.033 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale brown solid (0.050 g, 14% yield). MP: 254-256° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.79 (s, 1H), 8.07 (s, 1H), 8.04 (dd, J=8.0, 1.6 Hz, 1H), 7.87 (s, 1H), 7.85 (m, 1H), 7.69 (d, J=8.3 Hz, 1H), 7.60-7.49 (m, 3H), 7.29 (br s, 1H), 7.07 (dt, J=8.6, 2.3 Hz, 1H), 6.93 (br s, 2H), 6.05 (q, J=7.1 Hz, 1H), 2.51 (s, 3H), 1.91 (d, J=7.0 Hz, 3H). Mass: 532.03 (M++1). Example 107 N-(4-(4-amino-1-(1-(3-(3-fluorophenyl)-4-oxo-4H-chromen-2-yl) ethyl)-1H-pyrazolo [3,4-d] pyrimidin-3-yl) phenyl) acetamide To a solution of Example 57c (0.350 g, 0.663 mmoles) in DMF (3.5 ml), ethanol (1.75 ml) and water (1.75 ml), 4-Acetamidophenyl boronic acid (0.237 g, 1.32 mmoles) and sodium carbonate (0.211 g, 1.99 mmoles) were added and the system is degassed for 30 min Palladium tetrakis triphenylphosphine (0.038 g, 0.033 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.080 g, 24% yield).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 10.12 (s, 1H), 8.06 (s, 1H), 8.04 (dd, J=8.0, 1.4 Hz, 1H), 7.85 (m, 1H), 7.74 (d, J=8.5 Hz, 2H), 7.68 (d, J=8.2 Hz, 1H), 7.58 (m, 3H), 7.32 (m, 1H), 7.06 (dt, J=8.2, 2.4 Hz, 1H), 6.82 (m, 2H), 6.02 (q, J=7.0 Hz, 1H), 2.06 (s, 3H), 1.89 (d, J=7.1 Hz, 3H). Example 107a 2-(1-(4-amino-3-(4-aminophenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 107 (0.080 g, 0.149 mmoles) in ethanol (5 ml), Con·HCl (0.5 ml) was added and refluxed for 2 h. The reaction mixture was basified with sodium carbonate solution and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.020 g, 27% yield). MP: 91-94° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.04 (dd, J=8.3, 1.5 Hz, 1H), 8.02 (s, 1H), 7.85 (m, 1H), 7.67 (d, J=8.4 Hz, 1H), 7.53 (t, J=7.6 Hz, 2H), 7.29 (m, 3H), 7.06 (dt, J=8.7, 2.3 Hz, 1H), 6.91 (br s, 1H), 6.68 (d, J=8.4 Hz, 2H), 6.00 (q, J=7.0 Hz, 1H), 5.42 (s, 2H), 1.87 (d, J=7.0 Hz, 3H). Mass: 492.83 (M+). Example 108 2-(1-(4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57f (0.400 g, 0.733 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), N-Boc-3-methyl-6-indazoleboronic acid pinacol ester 98 (0.393 g, 1.099 mmoles) and sodium carbonate (0.233 g, 2.19 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.045 g, 11% yield). MP: 234-236° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.06 (s, 1H), 7.86-7.70 (m, 4H), 7.61 (s, 1H), 7.33 (m, 2H), 7.06 (dt, J=8.9, 2.5 Hz, 1H), 6.87 (m, 2H), 6.07 (q, J=7.0 Hz, 1H), 2.48 (s, 3H), 1.91 (d, J=7.1 Hz, 3H). Mass: 549.95 (M+). Example 109 2-(1-(4-amino-3-(2,3-dihydrobenzofuran-5-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 107 (0.100 g, 0.394 mmoles) in DMF (4 ml), potassium carbonate (0.109 g, 0.789 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 36 (0.217 g, 0.789 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.085 g, 41% yield). MP: 238-241° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.04 (s, 1H), 8.02 (d, J=6.0 Hz, 1H), 7.83 (m, 1H), 7.68 (d, J=8.3 Hz, 1H), 7.53 (t, J=7.7 Hz, 1H), 7.44 (s, 1H), 7.31 (m, 3H), 7.05 (t, J=8.9 Hz, 1H), 6.90 (m, 2H), 6.01 (q, J=7.0 Hz, 1H), 4.60 (t, J=8.7 Hz, 2H), 3.27 (t, J=8.6 Hz, 2H), 1.88 (d, J=7.0 Hz, 3H), Mass: 520.00 (M+). Example 110 2-(1-(4-amino-3-(3-ethyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.758 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), N-Boc-3-ethyl-6-indazoleboronic acid pinacol ester 103 (0.423 g, 1.137 mmoles) and sodium carbonate (0.241 g, 2.27 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.060 g, 15% yield). MP: 270-273° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.08 (s, 1H), 8.05 (dd, J=7.9, 1.4 Hz, 1H), 7.88 (m, 2H), 7.68 (d, J=8.4 Hz, 1H), 7.63 (s, 1H), 7.53 (d, J=7.2 Hz, 1H), 7.33 (d, J=8.3 Hz, 1H), 7.29 (br s, 1H), 7.07 (dt, J=8.9, 1.4 Hz, 1H), 6.95 (br s, 2H), 6.07 (q, J=6.9 Hz, 1H), 2.98 (q, J=7.5 Hz, 2H), 1.92 (d, J=7.1 Hz, 3H), 1.34 (t, J=7.6 Hz, 3H), Mass: 546.04 (M+). Example 111 2-(1-(4-amino-3-(3-methyl-1H-indol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.758 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 3-methyl-6-indoleboronic acid pinacol ester 106 (0.390 g, 1.517 mmoles) and sodium carbonate (0.241 g, 2.27 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.040 g, 10% yield). MP: 269-272° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 10.91 (s, 1H), 8.06 (s, 1H), 8.05 (d, J=7.8 Hz, 1H), 7.85 (t, J=7.2 Hz, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.63 (d, J=7.1 Hz, 1H), 7.56 (s, 1H), 7.53 (t, J=7.8 Hz, 1H), 7.25 (d, J=8.1 Hz, 1H), 7.28 (br s, 1H), 7.21 (s, 1H), 7.06 (dt, J=9.0, 2.8 Hz, 1H), 6.98 (br s, 2H), 6.04 (q, J=7.0 Hz, 1H), 2.28 (s, 3H), 1.91 (d, J=7.0 Hz, 3H). Mass: 530.99 (M+). Example 112 2-(1-(4-amino-3-(2-methoxypyrimidin-5-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.758 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 2-methoxypyrimidine-5-boronic acid (0.233 g, 1.517 mmoles) and sodium carbonate (0.241 g, 2.27 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.200 g, 51% yield). MP: 224-227° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.72 (s, 2H), 8.09 (s, 1H), 8.04 (dd, J=7.9, 1.4 Hz, 1H), 7.84 (m, 1H), 7.69 (d, J=8.3 Hz, 1H), 7.52 (t, J=7.8 Hz, 1H), 7.32 (m, 1H), 7.12-6.95 m, 5H), 6.03 (q, J=7.1 Hz, 1H), 1.90 (d, J=7.0 Hz, 3H). Mass: 509.99 (M+). Example 113 4-(4-amino-1-(1-(3-(3-fluorophenyl)-4-oxo-4H-chromen-2-yl) ethyl)-1H-pyrazolo [3,4-d] pyrimidin-3-yl) thiophene-2-carbaldehyde To a solution of Example 57c (0.350 g, 0.663 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), 2-formyl-4-thiopheneboronic acid (0.155 g, 0.995 mmoles) and sodium carbonate (0.210 g, 1.98 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.038 g, 0.033 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid (0.065 g, 19% yield). MP: 192-195° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 10.01 (s, 1H), 8.30 (s, 1H), 8.24 (s, 1H), 8.07 (s, 1H), 8.05 (dd, J=7.9, 1.4 Hz, 1H), 7.85 (m, 1H), 7.69 (d, J=8.4 Hz, 1H), 7.53 (t, J=7.8 Hz, 1H), 7.28 (br s, 1H), 7.06 (t, J=8.8 Hz, 1H), 6.93 (br s, 2H), 6.04 (q, J=7.0 Hz, 1H), 1.89 (d, J=7.0 Hz, 3H). Mass: 511.95 (M+). Example 114 2-(1-(4-amino-3-(5-(hydroxymethyl) thiophen-3-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.300 g, 0.568 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 2-hydroxymethyl-4-thiopheneboronic acid (0.133 g, 0.853 mmoles) and sodium carbonate (0.180 g, 1.70 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.033 g, 0.028 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid (0.042 g, 14% yield). MP: 154-156° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.05 (s, 1H), 8.04 (dd, J=7.9, 1.4 Hz, 1H), 7.85 (m, 1H), 7.67 (d, J=6.4 Hz, 1H), 7.66 (s, 1H), 7.53 (t, J=7.2 Hz, 1H), 7.29 (br s, 1H), 7.20 (s, 1H), 7.06 (dt, J=8.8, 2.1 Hz, 1H), 6.98 (br s, 2H), 6.02 (q, J=6.9 Hz, 1H), 5.54 (t, J=5.8 Hz, 1H), 4.68 (d, J=5.7 Hz, 2H), 1.88 (d, J=7.0 Hz, 3H). Mass: 514.19 (M++1). Example 115 2-(1-(4-amino-3-(2-methyl-1H-benzo[d]imidazol-5-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.758 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), intermediate 109 (0.407 g, 1.137 mmoles) and sodium carbonate (0.241 g, 2.274 mmoles) were added and the system is degassed for 30 min Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid (0.025 g, 6% yield). MP: 154-156° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.34 (s, 1H), 8.07 (s, 1H), 8.05 (dd, J=7.9, 1.3 Hz, 1H), 7.83 (m, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.62 (m, 2H), 7.53 (t, J=7.3 Hz, 1H), 7.38-7.30 (m, 3H), 7.05 (dt, J=8.5, 1.9 Hz, 1H), 6.93 (br s, 1H), 6.05 (q, J=6.9 Hz, 1H), 2.50 (s, 3H), 1.91 (d, J=7.0 Hz, 3H). Mass: 531.97 (M+). Example 116 2-(1-(4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) propyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57 g (0.400 g, 0.738 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), N-Boc-3-methyl-6-indazoleboronic acid pinacol ester 98 (0.397 g, 1.108 mmoles) and sodium carbonate (0.157 g, 1.47 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.023 g, 6% yield). MP: 268-270° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.07 (s, 1H), 8.04 (dd, J=7.9, 1.5 Hz, 1H), 7.86 (m, 2H), 7.69 (d, J=8.2 Hz, 1H), 7.64 (s, 1H), 7.53 (t, J=7.9 Hz, 1H), 7.35 (dd, J=8.2, 1.4 Hz, 1H), 7.33 (br s, 1H), 7.09 (dt, J=8.9, 2.2 Hz, 1H), 6.90 (br s, 2H), 5.85 (t, J=6.1 Hz, 1H), 2.51 (s, 3H), 2.50 (m, 2H), 0.82 (t, J=7.3 Hz, 3H). Mass: 545.96 (M+). Example 117 2-(1-(4-amino-3-(3-methyl-1H-indol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57b (0.290 g, 0.583 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), 3-methyl-6-indoleboronic acid pinacol ester 106 (0.299 g, 1.163 mmoles) and sodium carbonate (0.185 g, 1.749 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.033 g, 0.029 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale brown solid (0.014 g, 5% yield). MP: 262-265° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 10.92 (s, 1H), 8.07 (s, 1H), 8.04 (d, J=7.9 Hz, 1H), 7.81 (m, 1H), 7.64 (d, J=7.3 Hz, 1H), 7.57 (s, 1H), 7.51 (t, J=7.6 Hz, 1H), 7.35-7.10 (m, 7H), 5.97 (q, J=7.0 Hz, 1H), 2.28 (s, 3H), 1.91 (d, J=7.0 Hz, 3H). Mass: 512.99 (M+). Example 118 2-((6-amino-9H-purin-9-yl) methyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of Adenine (0.162 g, 1.20 mmoles) in DMF (3.5 ml), potassium carbonate (0.165 g, 1.20 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 90 (0.200 g, 0.600 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.040 g, 17% yield). MP: 207-209° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.09 (s, 1H), 8.07 (s, 1H), 7.73 (dd, J=8.4, 3.1 Hz, 1H), 7.66 (dt, J=8.1, 3.1 Hz, 1H), 7.59 (dd, J=9.1, 4.3 Hz, 1H), 7.45-7.40 (m, 5H), 7.22 (s, 2H), 5.34 (s, 2H). Mass: 388.18 (M++1). Example 119 2-((6-amino-9H-purin-9-yl) methyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Adenine (0.153 g, 1.13 mmoles) in DMF (3.5 ml), potassium carbonate (0.156 g, 1.13 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 92 (0.200 g, 0.567 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as green solid (0.020 g, 9% yield). MP: 180-183° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.11 (s, 1H), 8.07 (s, 1H), 7.73-7.65 (m, 2H), 7.62 (dd, J=9.2, 4.4 Hz, 1H), 7.50 (q, J=7.9 Hz, 1H), 7.26 (m, 5H), 5.36 (s, 2H). Mass: 406.10 (M++1). Example 120 2-((4-amino-3-(3-fluoro-5-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 79 (0.110 g, 0.424 mmoles) in DMF (3 ml), N,N-diisopropylethylamine (0.109 g, 0.848 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 92 (0.298 g, 0.848 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.075 g, 33% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.20 (s, 1H), 7.73-7.61 (m, 3H), 7.38 (q, J=7.6 Hz, 1H), 7.17 (m, 3H), 6.95 (m, 3H), 5.55 (s, 2H), 3.82 (s, 3H). Mass: 515.93 (M+). Example 120a 2-((4-amino-3-(3-fluoro-5-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 120 (0.075 g, 0.140 mmoles) in dichloromethane (10 ml), BBr3(1M in dichloromethane, 1.0 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light brown solid (0.023 g, 31% yield). MP: 127-129° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.18 (s, 1H), 8.19 (s, 1H), 7.74-7.61 (m, 3H), 7.38 (q, J=7.8 Hz, 1H), 7.15 (m, 3H), 6.84 (s, 1H), 6.81 (d, J=8.8 Hz, 1H), 6.65 (d, J=10.8 Hz, 1H), 5.54 (s, 2H). Mass: 515.54 (M+). Example 121 2-(1-(4-amino-3-(3-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 58 (0.254 g, 1.054 mmoles) in DMF (6 ml), potassium carbonate (0.331 g, 2.39 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 75 (0.350 g, 0.958 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.210 g, 42% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.07 (s, 1H), 7.82 (dd, J=9.2, 4.4 Hz, 1H), 7.76 (dd, J=8.0, 3.1 Hz, 1H), 7.72 (dd, J=8.2, 2.7 Hz, 1H), 7.46 (t, J=7.9 Hz, 1H), 7.28 (br s, 1H), 7.18 (d, J=7.7 Hz, 1H), 7.10 (t, J=2.4 Hz, 1H), 7.07 (m, 2H), 6.92 (m, 2H), 6.04 (q, J=7.0 Hz, 1H), 3.80 (s, 3H) 1.89 (d, J=7.1 Hz, 3H). Example 121a 2-(1-(4-amino-3-(3-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 121 (0.180 g, 0.324 mmoles) in dichloromethane (15 ml), BBr3(1M in dichloromethane, 1.6 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as grey solid (0.045 g, 27% yield). MP: 193-196° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.74 (s, 1H), 8.17 (s, 1H), 7.83-7.70 (m, 4H), 7.63 (m, 1H), 7.35 (t, J=8.2 Hz, 1H), 7.31 (m, 1H), 7.12 (m, 4H), 6.99 (m, 2H), 6.08 (q, J=6.8 Hz, 1H), 1.90 (d, J=7.0 Hz, 3H). Mass: 511.87 (M+). Example 122 2-((9H-purin-6-ylamino) methyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 73 (3.0 g, 12.03 mmoles) in dichloromethane (30 ml), triethylamine (5.0 ml, 36.11 mmoles) was added followed by N-Boc-Glycine (2.53 g, 14.44 mmoles). To this mixture HATU (9.15 g, 24.07 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (4 g). To a solution of this intermediate (4.0 g), trifluoroacetic acid (4 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (2.5 g). To a solution of this amine intermediate (0.500 g, 1.74 mmoles) in tert-butanol (8 ml), N,N-diisopropylethylamine (0.6 ml, 2.94 mmoles) and 6-chloropurine (0.268 g, 1.74 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as pale brown solid (0.090 g, 13% yield). MP: 229-232° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.97 (s, 1H), 8.15 (m, 1H), 8.13 (s, 1H), 8.11 (s, 1H), 7.73 (dd, J=8.4, 3.1 Hz, 1H), 7.68 (m, 2H), 7.46 (q, J=6.4 Hz), 7.26-7.20 (m, 3H), 4.60 (br s, 2H). Mass: 406.17 (M++1). Example 123 2-((9H-purin-6-ylamino) methyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of intermediate 50 (3.0 g, 13.03 mmoles) in dichloromethane (30 ml), triethylamine (5.4 ml, 39.09 mmoles) was added followed by N-Boc-Glycine (2.73 g, 15.63 mmoles). To this mixture HATU (9.90 g, 26.08 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (2.5 g). To a solution of this intermediate (2.5 g), trifluoroacetic acid (3 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (1.7 g). To a solution of this amine intermediate (0.500 g, 1.85 mmoles) in tert-butanol (8 ml), N, N-diisopropylethylamine (0.64 ml, 3.71 mmoles) and 6-chloropurine (0.286 g, 1.85 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as pale brown solid (0.070 g, 10% yield). MP: 183-186° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.96 (s, 1H), 8.16 (m, 1H), 8.14 (s, 1H), 8.11 (s, 1H), 7.73 (dd, J=8.4, 3.1 Hz, 1H), 7.67 (m, 2H), 7.45-7.35 m, 5H), 4.59 (br s, 2H). Mass: 388.25 (M++1). Example 124 (R)-2-(1-(9H-purin-6-ylamino) ethyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 73 (3.0 g, 12.03 mmoles) in dichloromethane (30 ml), triethylamine (5.0 ml, 36.11 mmoles) was added followed by N-Boc-D-Alanine (2.70 g, 14.44 mmoles). To this mixture HATU (9.15 g, 24.07 mmoles) was added and stirred at RT for 12 h. The reaction mixture was quenched by the addition of water and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate:petroleum ether to afford the isoflavone intermediate (1.8 g). To a solution of this intermediate (1.8 g), trifluoroacetic acid (1.8 ml) was added and stirred at RT for 2 h. The reaction mixture was concentrated, basified with sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure to afford the amine intermediate (1.1 g). To a solution of this amine intermediate (1.0 g, 3.31 mmoles) in tert-butanol (20 ml), N,N-diisopropylethylamine (1.15 ml, 6.63 mmoles) and 6-chloropurine (0.384 g, 2.48 mmoles) were added and refluxed for 24 h. The reaction mixture was concentrated, diluted with water, extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:ethyl acetate to afford the title compound as pale brown solid (0.100 g, 7% yield). MP: 194-197° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 12.96 (s, 1H), 8.14 (m, 3H), 7.70 (m, 3H), 7.49 (q, J=7.3 Hz, 1H), 7.25 (m, 3H), 5.20 (br s, 1H), 1.55 (d, J=6.9 Hz, 3H). Mass: 419.96 (M+). Example 125 2-((4-amino-3-(1H-pyrazol-4-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-6-fluoro-3-phenyl-4H-chromen-4-one To a solution of intermediate 57d (0.400 g, 0.77 mmoles) in DMF (5 ml), ethanol (2.5 ml) and water (2.5 ml), N-Boc-pyrazole-4-boronic acid pinacol ester (0.344 g, 1.16 mmoles) and sodium carbonate (0.165 g, 1.16 mmoles) were added and the system is degassed for 30 min. Tetrakis triphenylphosphine. Palladium (0.027 g, 0.023 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.120 g, 34% yield). MP: 211-214° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 13.19 (s, 1H), 8.19 (s, 1H), 8.10 (s, 1H), 7.79 (s, 1H), 7.73 (dd, J=8.3, 3.1 Hz, 1H), 7.71 (dt, J=8.7.5, 3.1 Hz, 1H), 7.54 (dd, J=9.3, 4.3 Hz, 1H), 7.40-7.20 (m, 5H), 6.92 (br s, 2H), 5.46 (s, 2H). Mass: 454.26 (M+). Example 126 2-(1-(4-amino-3-(3,5-difluoro-4-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 113 (0.110 g, 0.396 mmoles) in DMF (10 ml), cesium carbonate (0.258 g, 0.792 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 36 (0.275 g, 0.792 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as yellow solid (0.122 g, 56% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.07 (s, 1H), 8.04 (dd, J=7.9, 1.5 Hz, 1H), 7.85 (m, 1H), 7.68 (dd, J=8.3 Hz, 1H), 7.63 (m, 1H), 7.56 (m, 2H), 7.35 (s, 1H), 7.30 (d, J=8.7 Hz, 2H), 7.07 (dt, J=8.7, 2.3 Hz, 1H), 6.93 (br s, 2H), 6.04 (q, J=6.9 Hz, 1H), 3.97 (s, 3H), 1.88 (d, J=7.0 Hz, 3H). Example 126a 2-(1-(4-amino-3-(3,5-difluoro-4-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of example 126 (0.122 g, 0.224 mmoles) in dichloromethane (10 ml), BBr3(1M in dichloromethane, 1.2 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated to afford the title compound as brown solid (0.086 g, 72% yield). MP: 253-257° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 9.64 (s, 1H), 8.05 (s, 1H), 8.04 (dd, J=8.0, 1.4 Hz, 1H), 7.83 (m, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.52 (t, J=5.3 Hz, 1H), 7.30 (m, 1H), 7.20 (d, J=8.7 Hz, 2H), 7.06 (dt, J=8.7, 2.2 Hz, 1H), 6.98 (br s, 2H), 6.00 (q, J=7.0 Hz, 1H), 1.88 (d, J=7.0 Hz, 3H). Mass: 5530.14 (M++1). Example 127 2-((4-amino-3-(3,5-difluoro-4-methoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of intermediate 113 (0.080 g, 0.288 mmoles) in DMF (3 ml), N, N-diisopropylethylamine (0.074 g, 0.577 mmoles) was added and stirred at RT for 10 min. To this mixture, intermediate 92 (0.203 g, 0.577 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.109 g, 68% yield).1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 8.20 (s, 1H), 7.73-7.52 (m, 4H), 7.38 (m, 1H), 7.30 (d, J=8.8 Hz, 2H), 7.16-7.07 (m, 4H), 5.53 (s, 2H), 3.96 (s, 3H). Example 127a 2-((4-amino-3-(3,5-difluoro-4-hydroxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) methyl)-6-fluoro-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of example 127 (0.099 g, 0.180 mmoles) in dichloromethane (10 ml), BBr3(1M in dichloromethane, 0.99 ml) was added at 0° C. and the reaction mixture was warmed to RT and then stirred for 12 h. The reaction mixture was quenched with 1.5N HCl solution and extracted with dichloromethane. The organic layer was dried over sodium sulphate and concentrated to afford the title compound as brown solid (0.022 g, 23% yield). MP: 274-278° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 10.20 (s, 1H), δ 8.18 (s, 1H), 7.72-7.60 (m, 4H), 7.38 (m, 1H), 7.20 (d, J=8.7 Hz, 2H), 7.17-7.10 (m, 4H), 5.51 (s, 2H). Mass: 534.06 (M++1). Example 128 (+)-2-(1-(4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one Example 129 (−)-2-(1-(4-amino-3-(3-methyl-1H-indazol-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The two enantiomerically pure isomers were obtained by preparative chiral hplc separation from example 79 on a CHIRALPAK IA column (250×20 mm; 5μ) using dichloromethane:acetonitrile:methanol (90:08:02, v/v/v) as the mobile phase. (+)-Isomer: Off-white solid, e.e. 99.68%. Rt: 5.55 min (CHIRALPAK IA, conditions as above). MP: 158-161° C. [α]25D196.56 (c=0.40, CH2Cl2).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.74 (s, 1H), 8.08 (s, 1H), 8.05 (dd, J=7.9, 1.4 Hz, 1H), 7.86 (m, 2H), 7.68 (d, J=8.4 Hz, 1H), 7.62 (s, 1H), 7.53 (t, J=7.8 Hz, 1H), 7.34 (d, J=8.3 Hz, 1H), 7.31 (m, 1H), 7.07 (dt, J=8.8, 2.3 Hz, 1H), 6.93 (br s, 2H), 6.07 (q, J=7.0 Hz, 1H), 2.51 (s, 3H), 1.92 (d, J=7.1 Hz, 3H). Mass: 532.39 (M++1). (−)-Isomer: Off-white solid, e.e.98.33%. Rt: 7.39 min (CHIRALPAK IA, conditions as above). MP: 157-160° C. [α]25D−191.54 (c=0.40, CH2Cl2).1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 12.75 (s, 1H), 8.08 (s, 1H), 8.05 (dd, J=7.9, 1.4 Hz, 1H), 7.85 (m, 2H), 7.68 (d, J=8.4 Hz, 1H), 7.62 (s, 1H), 7.53 (t, J=7.9 Hz, 1H), 7.34 (dd, J=8.3, 1.1 Hz, 1H), 7.31 (m, 1H), 7.07 (dt, J=8.6, 2.1 Hz, 1H), 6.94 (br s, 2H), 6.07 (q, J=6.9 Hz, 1H), 2.51 (s, 3H), 1.92 (d, J=7.1 Hz, 3H). Mass: 532.39 (M++1). Example 130 2-(1-(4-amino-3-(3,5-dimethoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of example 57c (100 mg, 0.190 mmol) in DME (1 ml), and water (0.5 ml), 3,5-dimethoxy phenyl boronic acid (0.209 mmol) and sodium carbonate (40 mg, 0.380 mmol) were added and the system was degassed for 5 min 1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (27.8 mg, 0.038 mmol) was added under nitrogen atmosphere and the mixture was heated to 90° C. at a microwave reactor for 15 min. LC-MS analysis indicated the total consumption of example 57c, then ethyl acetate (2 ml) and water (0.5 ml) was added. The two phases were separated and the aqueous layer was extracted by ethyl acetate (1 ml). The combined organic layer was dried over Na2SO4, filtered and evaporated to dryness. The residue was purified by preparative TLC using mixture of ethyl acetate:petroleum ether in 2:1 ratio as an eluent to afford the desired compound. Brown solid (23.4 mg, 23%). MP: 224-227° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.21 (dd, J=8.0, 1.5 Hz, 1H), 7.69 (m, 1H), 7.48 (d, J=8.4 Hz, 1H), 7.42 (t, J=8.0 Hz, 1H), 7.32 (m, 1H), 7.04 (m, 3H), 6.79 (d, J=2.3 Hz, 2H), 6.56 (t, J=2.1 Hz, 1H), 6.11 (q, J=7.2 Hz, 1H), 5.58 (s, 2H), 3.85 (s, 6H), 2.02 (d, J=7.1 Hz, 3H). Mass: 537.8 (M+). Example 131 2-(1-(4-amino-3-(4-methoxy-3, 5-dimethylphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 3, 5-dimethyl-4-methoxyphenyl boronic acid (0.209 mmol). Brown solid (20 mg, 20%). MP: 234-236° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.22 (s, 1H), 8.21 (dd, J=8.0, 1.5 Hz, 1H), 7.69 (m, 1H), 7.48 (d, J=8.4 Hz, 1H), 7.42 (t, J=8.0 Hz, 1H), 7.30 (s, 2H), 7.29 (m, 1H), 7.02-6.95 (m, 3H), 6.10 (q, J=7.1 Hz, 1H), 5.43 (s, 2H), 3.77 (s, 3H), 2.36 (s, 6H), 2.01 (d, J=7.1 Hz, 3H). Mass: 535.9 (M+). Example 132 2-(1-(4-amino-3-(2-fluoro-5-isopropoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 2-Fluoro-5-isopropoxyphenyl boronic acid (0.209 mmol). Brown solid (50.6 mg, 48%). MP: 198-201° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.21 (dd, J=7.9, 1.5 Hz, 1H), 7.68 (m, 1H), 7.47 (d, J=8.0 Hz, 1H), 7.41 (dd, J=8.0, 0.9 Hz, 1H), 7.34 (m, 1H), 7.18 (t, J=9.9 Hz, 1H), 7.07-6.96 (m, 5H), 6.13 (q, J=7.1 Hz, 1H), 5.32 (s, 2H), 4.53 (quintet, J=6.0 Hz, 1H), 2.01 (d, J=7.1 Hz, 3H), 1.35 (d, J=6.0 Hz, 6H). Mass: 553.8 (M+). Example 133 2-(1-(4-amino-3-(2,3-dihydrobenzo[b] [1,4] dioxin-6-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid (or boronic acid pinacol ester) was replaced by -2, 3-dihydrobenzo[b] [1,4] dioxin-6-ylboronic acid (0.209 mmol) Off-white solid (22 mg, 22%). MP: 225-226° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.21 (s, 1H), 8.19 (dd, J=8.1, 1.2 Hz, 1H), 7.69 (m, 1H), 7.48 (d, J=8.4 Hz, 1H), 7.42 (dt, J=8.1, 1.2 Hz, 1H), 7.31 (m, 1H), 7.19 (d, J=2.1 Hz, 1H), 7.15 (dd, J=8.4, 2.1 Hz, 1H), 7.03-6.95 (m, 4H), 6.09 (q, J=7.2 Hz, 1H), 5.58 (s, 2H), 4.31 (s, 4H), 2.00 (d, J=7.2 Hz, 3H). Mass: 535.8 (M+). Example 134 2-(1-(4-amino-3-(1-benzyl-1H-pyrazol-4-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid (or boronic acid pinacol ester) was replaced by -1-benzylpyrazole-4-boronic acid pinacol ester (0.209 mmol) Brown solid (35 mg, 33%). MP: 140-142° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.21 (s, 1H), 8.21 (dd, J=8.2, 1.5 Hz, 1H), 7.84 (s, 1H), 7.73 (s, 1H), 7.67 (m, 1H), 7.47 (d, J=8.2 Hz, 1H), 7.40-7.32 (m, 7H), 6.98 (m, 3H), 6.05 (q, J=7.2 Hz, 1H), 5.41 (s, 2H), 5.38 (s, 2H), 1.98 (d, J=7.1 Hz, 3H). Mass: 557.8 (M+). Example 135 2-(1-(4-amino-3-(2-methylpyridin-4-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 2-methylpyridine-4-boronic acid (0.209 mmol) Off-white solid (30 mg, 32%). MP: 266-268° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ), 8.68 (d, J=5.4 Hz, 1H), 8.27 (s, 1H), 8.22 (dd, J=7.9, 1.5 Hz, 1H), 7.70 (m, 1H), 7.49-7.32 m, 5H), 7.04-6.92 (m, 3H), 6.13 (q, J=7.2 Hz, 1H), 5.47 (s, 2H), 2.67 (s, 3H), 2.02 (d, J=7.2 Hz, 3H). Mass: 492.8 (M+). Example 136 2-(1-(4-amino-3-(3, 4-dihydro-2H-benzo[b] [1,4] dioxepin-7-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 3, 4-dihydro-2H-benzo[b] [1,4] dioxepin-7-ylboronic acid (0.209 mmol) Brown solid (15 mg, 14%). MP: 234-237° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.22 (s, 1H), 8.19 (dd, J=7.5, 1.8 Hz, 1H), 7.69 (m, 1H), 7.48 (d, J=8.4 Hz, 1H), 7.43 (dt, J=7.8, 0.9 Hz, 1H), 7.30 (d, J=2.1 Hz, 1H), 7.28 (m, 2H), 7.23 (d, J=2.1 Hz, 1H), 7.20 (m, 3H), 6.10 (q, J=7.2 Hz, 1H), 5.62 (s, 2H), 4.31 (d, J=5.7 Hz, 4H), 2.27 (t, J=5.7 Hz, 2H), 2.01 (d, J=7.2 Hz, 3H). Mass: 549.5 (M+). Example 137 2-(1-(4-amino-3-(6-morpholinopyridin-3-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 6-morpholinopyridin-3-ylboronic acid (0.209 mmol) Brown solid (36 mg, 34%). MP: 269-271° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.49 (d, J=2.1 Hz, 1H), 8.24 (s, 1H), 8.21 (dd, J=8.0, 1.5 Hz, 1H), 7.82 (dd, J=8.8, 2.4 Hz, 1H), 7.69 (m, 1H), 7.48 (d, J=8.4 Hz, 1H), 7.42 (t, J=8.0 Hz, 1H), 7.30 (m, 1H), 7.02-6.91 (m, 3H), 6.77 (d, J=8.8 Hz, 1H), 6.12 (q, J=7.2 Hz, 1H), 5.41 (s, 2H), 3.86 (t, J=4.6 Hz, 4H), 3.61 (t, J=5.0 Hz, 4H), 2.01 (d, J=7.1 Hz, 3H). Mass: 563.8 (M+). Example 138 2-(1-(4-amino-3-(dibenzo [b, d] furan-4-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by dibenzo[b,d]furan-4-ylboronic acid (0.209 mmol) Brown solid (52.6 mg, 49%). MP: 238-240° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.28 (s, 1H), 8.23 (d, J=6.7 Hz, 1H), 8.10 (d, J=7.1 Hz, 1H), 8.03 (d, J=7.6 Hz, 1H), 7.71 (m, 2H), 7.54-7.49 (m, 4H), 7.46 (t, J=7.6 Hz, 2H), 7.34 (m, 1H), 7.11 (d, J=7.6 Hz, 1H), 7.06 (m, 2H), 6.20 (q, J=7.1 Hz, 1H), 5.29 (s, 2H), 2.07 (d, J=7.1 Hz, 3H). Mass: 567.8 (M+). Example 139 2-(1-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 4-phenoxyphenylboronic acid (0.209 mmol) Brown solid (61.9 mg, 57%). MP: 218-220° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.22 (dd, J=7.9, 1.5 Hz, 1H), 7.69 (m, 3H), 7.42 (d, J=8.2 Hz, 1H), 7.41 (m, 3H), 7.32 (m, 1H), 7.19-7.13 (m, 3H), 7.08-6.92 (m, 5H), 6.11 (q, J=7.1 Hz, 1H), 5.39 (s, 2H), 2.02 (d, J=7.2 Hz, 3H). Mass: 569.8 (M+). Example 140 2-(1-(4-amino-3-(4-(benzyloxy)-3-chlorophenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 4-(benzyloxy)-3-chlorophenylboronic acid (0.209 mmol) Brown solid (58 mg, 49%). MP: 214-216° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.22 (dd, J=7.9, 1.5 Hz, 1H), 7.74 (d, J=2.1 Hz, 1H), 7.69 (m, 1H), 7.49-7.31 (m, 9H), 7.12 (d, J=8.5 Hz, 1H), 7.03 (m, 2H), 6.94 (d, J=9.3 Hz, 1H), 6.10 (q, J=7.2 Hz, 1H), 5.38 (s, 2H), 5.24 (s, 2H), 2.00 (d, J=7.1 Hz, 3H). Mass: 618.8 (M+). Example 141 2-(1-(4-amino-3-(3-chloro-4-isopropoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3-chloro-4-isopropoxyphenylboronic acid (0.209 mmol) Brown solid (52.8 mg, 49%). MP: 198-200° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 7.70 (m, 2H), 7.51-7.47 (m, 2H), 7.42 (dt, J=8.0, 0.9 Hz, 1H), 7.30 (m, 1H), 7.09 (d, J=7.5 Hz, 1H), 7.03-6.91 (m, 3H), 6.12 (q, J=7.1 Hz, 1H), 5.41 (s, 2H), 4.67 (quintet, J=6.2 Hz, 1H), 2.01 (d, J=7.1 Hz, 3H), 1.44 (d, J=6.0 Hz, 6H). Mass: 570.8 (M+). Example 142 2-(1-(4-amino-3-(3-(dimethylamino) phenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3-(dimethylamino)phenylboronic acid (0.209 mmol) Brown solid (60 mg, 60%). MP: 218-220° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.23 (s, 1H), 8.21 (dd, J=8.0, 1.5 Hz, 1H), 7.68 (m, 1H), 7.48 (d, J=8.2 Hz, 1H), 7.41 (m, 2H), 7.35 (m, 1H), 7.05 (d, J=7.6 Hz, 1H), 7.01-6.95 (m, 4H), 6.83 (dd, J=8.7, 2.1 Hz, 1H), 6.11 (q, J=7.1 Hz, 1H), 5.52 (s, 2H), 3.01 (s, 6H), 2.02 (d, J=7.1 Hz, 3H). Mass: 520.8 (M+). Example 143 2-(1-(4-amino-3-(4-ethoxy-3-fluorophenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic was replaced by 4-ethoxy-3-fluorophenylboronic acid (0.209 mmol) Brown solid (47.5 mg, 46%). MP: 216-218° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.22 (dd, J=8.1, 1.5 Hz, 1H), 7.68 (m, 1H), 7.49-7.35 m, 5H), 7.13 (t, J=8.4 Hz, 1H), 7.07 (m, 3H), 6.10 (q, J=7.2 Hz, 1H), 5.50 (s, 2H), 4.19 (q, J=7.2 Hz, 2H), 2.01 (d, J=7.2 Hz, 3H), 1.52 (t, J=7.2 Hz, 3H). Mass: 539.8 (M+). MS DATA Example 144 2-(1-(4-amino-3-(4-isopropoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 4-isopropoxyphenylboronic acid (0.209 mmol) Brown solid (23.2 mg, 23%). MP: 224-226° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.22 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 7.67 (m, 1H), 7.58 (dd, J=6.7, 1.9 Hz, 2H), 7.49 (d, J=8.3 Hz, 1H), 7.42 (dt, J=8.0, 1.0 Hz, 1H), 7.30 (m, 1H), 7.04-6.98 (m, 5H), 6.12 (q, J=7.1 Hz, 1H), 5.41 (s, 2H), 4.65 (quintet, J=6.1 Hz, 1H), 2.01 (d, J=7.1 Hz, 3H), 1.38 (d, J=6.0 Hz, 6H). Mass: 535.8 (M+). Example 145 2-(1-(4-amino-3-(4-(trifluoromethoxy) phenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 4-(trifluoromethoxy)phenylboronic acid (0.209 mmol) Brown solid (46.6 mg, 48%). MP: 224-226° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.26 (s, 1H), 8.22 (dd, J=7.9, 1.5 Hz, 1H), 7.74 (d, J=7.6 Hz, 2H), 7.70 (m, 1H), 7.48 (d, J=8.3 Hz, 1H), 7.42 (m, 1H), 7.40 (d, J=8.1 Hz, 2H), 7.33 (m, 1H), 7.04 (m, 2H), 6.93 (d, J=7.9 Hz, 2H), 6.12 (q, J=7.2 Hz, 1H), 5.39 (s, 2H), 2.02 (d, J=7.2 Hz, 3H). Mass: 561.8 (M+). Example 146 2-(1-(3-(4-acetylphenyl)-4-amino-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 4-acetylphenylboronic acid (0.209 mmol) Off-white solid (20 mg, 20%). MP: 218-221° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.26 (s, 1H), 8.22 (dd, J=7.9, 1.5 Hz, 1H), 8.13 (d, J=8.3 Hz, 2H), 7.82 (d, J=8.4 Hz, 2H), 7.70 (m, 1H), 7.48 (d, J=8.2 Hz, 1H), 7.43 (dt, J=8.0, 0.9 Hz, 1H), 7.31 (m, 1H), 7.04-6.92 (m, 3H), 6.13 (q, J=7.1 Hz, 1H), 5.47 (s, 2H), 2.67 (s, 3H), 2.03 (d, J=7.2 Hz, 3H). Mass: 519.8 (M+). Example 147 2-(1-(4-amino-3-(4-(benzyloxy) phenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 4-(benzyloxy)phenylboronic acid (0.209 mmol) Off-white solid (68.2 mg, 61%). MP: 176-178° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.22 (s, 1H), 8.22 (dd, J=9.0, 1.6 Hz, 1H), 7.69 (m, 1H), 7.48-7.23 (m, 11H), 7.12-6.92 (m, 4H), 6.12 (q, J=7.1 Hz, 1H), 5.37 (s, 2H), 5.16 (s, 2H), 2.01 (d, J=7.1 Hz, 3H). Mass: 583.9 (M+). Example 148 2-(1-(4-amino-3-(4-(dimethylamino) phenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 4-(dimethylamino)phenylboronic acid (0.209 mmol) Brown solid (12.6 mg, 13%). MP: 214-217° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.21 (dd, J=7.8, 1.6 Hz, 1H), 8.21 (s, 1H), 7.69 (m, 1H), 7.54-7.48 (m, 3H), 7.41 (dt, J=8.0, 0.9 Hz, 1H), 7.31 (m, 1H), 7.02-6.95 (m, 3H), 6.84 (d, J=8.8 Hz, 2H), 6.09 (q, J=7.1 Hz, 1H), 5.47 (s, 2H), 3.02 (s, 6H), 2.01 (d, J=7.2 Hz, 3H). Mass: 520.89 (M+). Example 149 2-(1-(4-amino-3-(4-(methylsulfonyl) phenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 4-(methylsulfonyl) phenylboronic acid (0.209 mmol). Off-white solid (48.9 mg, 46%). MP: 259-262° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.27 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 8.14 (d, J=8.5 Hz, 2H), 7.93 (d, J=8.5 Hz, 2H), 7.69 (m, 1H), 7.47 (d, J=8.2 Hz, 1H), 7.43 (dt, J=8.0, 1.0 Hz, 1H), 7.32 (m, 1H), 7.03-6.90 (m, 3H), 6.16 (q, J=7.1 Hz, 1H), 5.56 (s, 2H), 3.12 (s, 3H), 2.02 (d, J=7.1 Hz, 3H). Mass: 555.8 (M+). Example 150 2-(1-(4-amino-3-(3-ethoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 3-ethoxyphenylboronic acid (0.209 mmol) Off-white solid (42.6 mg, 43%). MP: 162-165° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.15 (m, 2H), 7.38 (t, J=7.5 Hz, 1H), 7.18 (m, 3H), 7.11 (m, 3H), 6.95 (m, 4H), 6.04 (q, J=7.0 Hz, 1H), 5.63 (s, 2H), 4.03 (q, J=7.2 Hz, 2H), 1.95 (d, J=6.9 Hz, 3H), 1.39 (t, J=7.2 Hz, 3H). Mass 521.8 (M+). Example 151 2-(1-(4-amino-3-(benzo[b]thiophen-2-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by benzo[b]thiophen-2-ylboronic acid (0.209 mmol) Brown solid (25 mg, 24%). MP: 242-245° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.30-8.20 (m, 2H), 7.91 (m, 2H), 7.69 (m, 2H), 7.50-7.25 (m, 5H), 7.07 (m, 3H), 6.12 (q, J=7.1 Hz, 1H), 5.77 (s, 2H), 2.04 (d, J=7.2 Hz, 3H). Mass 533.8 (M+). Example 152 2-(1-(4-amino-3-(5-chlorothiophen-2-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic acid was replaced by 5-chlorothiophen-2-ylboronic acid (0.209 mmol) Brown solid (14.5 mg, 15%). MP: 226-229° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.25 (s, 1H), 8.21 (dd, J=8.0, 1.5 Hz, 1H), 7.70 (m, 1H), 7.48 (d, J=8.2z, 1H), 7.42 (dt, J=8.0, 1.1 Hz, 1H), 7.34 (m, 1H), 7.16 (dt, J=3.8 Hz, 1H), 7.04 (m, 3H), 6.96 (d, J=9.3 Hz, 1H), 6.08 (q, J=7.1 Hz, 1H), 5.62 (s, 2H), 2.00 (d, J=7.1 Hz, 3H). Mass: 517.88 (M+) Example 153 2-(1-(4-amino-3-(3,5-dimethylisoxazol-4-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3,5-dimethylisoxazol-4-ylboronic acid (0.209 mmol) Brown solid (23.1 mg, 24%). MP: 218-222° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.27 (s, 1H), 8.22 (dd, J=8.5, 1.6 Hz, 1H), 7.69 (m, 1H), 7.42 (m, 3H), 7.11-6.99 (m, 3H), 6.12 (q, J=7.2 Hz, 1H), 5.21 (s, 2H), 2.44 (s, 3H), 2.29 (s, 3H), 1.99 (d, J=7.2 Hz, 3H). Mass: 496.9 (M+). Example 154 2-(1-(4-amino-3-(3-propoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic was replaced by 3-propoxyphenylboronic acid (0.209 mmol) Brown solid (65.4 mg, 64%). MP: 178-182° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.22 (dd, J=8.0, 1.6 Hz, 1H), 7.69 (m, 1H), 7.48-7.38 (m, 3H), 7.31 (m, 1H), 7.23 (m, 2H), 7.04-6.93 (m, 4H), 6.13 (q, J=7.2 Hz, 1H), 5.47 (s, 2H), 4.00 (t, J=6.6 Hz, 2H), 2.02 (d, J=7.1 Hz, 3H), 1.86 (m, 2H), 1.07 (t, J=7.4 Hz, 3H). Mass: 535.8 (M+). Example 155 2-(1-(4-amino-3-(furan-2-yl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by furan-2-ylboronic acid (0.209 mmol) Brown solid (24.6 mg, 28%). MP: 234-236° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.22 (s, 1H), 8.19 (dd, J=8.3, 1.7 Hz, 1H), 7.68 (m, 1H), 7.59 (d, J=1.5 Hz, 1H), 7.49 (t, J=6.8 Hz, 1H), 7.40 (t, J=7.4 Hz, 1H), 7.31 (m, 1H), 6.99-6.96 (m, 4H), 6.61 (q, J=1.7 Hz, 1H), 6.07 (q, J=7.2 Hz, 1H), 1.99 (d, J=7.2 Hz, 3H. Mass: 467.9 (M+). Example 156 2-(1-(4-amino-3-(4-ethoxyphenyl)-1H-pyrazolo [3,4-d] pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3, 5-dimethoxy phenyl boronic was replaced by 4-ethoxyphenylboronic acid (0.209 mmol). Brown solid (53.4 mg, 54%). MP: 229-232° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.22 (s, 1H), 8.22 (d, J=7.8 Hz, 1H), 7.68 (m, 1H), 7.59 (d, J=8.7 Hz, 2H), 7.49 (d, J=8.4 Hz, 1H), 7.40 (m, 2H), 7.06 m, 5H), 6.11 (q, J=7.2 Hz, 1H), 5.62 (s, 2H), 4.11 (q, J=7.2 Hz, 2H), 2.02 (d, J=7.2 Hz, 3H), 1.48 (t, J=7.2 Hz, 3H). Mass: 521.9 (M+). Example 157 2-(1-(4-amino-3-(3-chloro-4-methoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid (or boronic acid pinacol ester) was replaced by 3-chloro-4-methoxyphenylboronic acid (0.209 mmol) Brown solid (30 mg, 29%). MP: 246-249° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 7.72-7.65 (m, 2H), 7.55 (dd, J=8.4, 2.1 Hz, 1H), 7.49 (d, J=8.3 Hz, 1H), 7.42 (dt, J=8.0, 0.9 Hz, 1H), 7.32 (m, 1H), 7.09 (d, J=8.5 Hz, 1H), 7.04 (m, 2H), 6.93 (d, J=8.1 Hz, 1H), 6.12 (q, J=7.3 Hz, 1H), 5.38 (s, 2H), 3.98 (s, 3H), 2.01 (d, J=7.1 Hz, 3H). Mass: 541.8 M+). Example 158 2-(1-(4-amino-3-(3-fluoro-4-isopropoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3-fluoro-4-isopropoxyphenylboronic acid (0.209 mmol) Brown solid (23 mg, 22%). MP: 218-221° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 7.70 (m, 1H), 7.49 (d, J=8.2 Hz, 1H), 7.44-7.30 (m, 4H), 7.14 (d, J=8.4 Hz, 1H), 7.03-6.91 (m, 3H), 6.12 (q, J=7.0 Hz, 1H), 5.43 (s, 2H), 4.66 (quintet, J=6.2 Hz, 1H), 2.00 (d, J=7.1 Hz, 3H) 1.42 (d, J=6.1 Hz, 6H). Mass: 553.8 (M+). Example 159 2-(1-(4-amino-3-(6-fluoropyridin-3-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 6-fluoropyridin-3-ylboronic acid (0.209 mmol) Brown solid (56.6 mg, 60%). MP: 203-206° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.57 (d, J=1.9 Hz, 1H), 8.28 (s, 1H), 8.22 (dd, J=7.9, 1.4 Hz, 1H), 8.16 (dt, J=7.9, 2.4 Hz, 1H), 7.70 (m, 1H), 7.47 (d, J=8.3 Hz, 1H), 7.43 (t, J=7.9 Hz, 1H), 7.34 (m, 1H), 7.15 (dd, J=8.4, 2.8 Hz, 1H), 7.04 (m, 2H), 6.93 (d, J=7.8 Hz, 1H), 6.13 (q, J=7.1 Hz, 1H), 5.38 (s, 2H), 2.02 (d, J=7.1 Hz, 3H). Mass: 496.9 (M+). Example 160 2-(1-(4-amino-3-(pyrimidin-5-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic was replaced by pyrimidin-5-ylboronic acid (0.209 mmol) Brown solid (34 mg, 37%). MP: 207-211° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 9.35 (s, 1H), 9.11 (s, 2H), 8.32 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 7.69 (m, 1H), 7.48 (d, J=8.4 Hz, 1H), 7.43 (t, J=8.0 Hz, 1H), 7.35 (m, 1H), 7.06 (m, 2H), 6.95 (d, J=9.9 Hz, 1H), 6.15 (q, J=7.1 Hz, 1H), 5.31 (s, 2H), 2.03 (d, J=7.1 Hz, 3H). Mass: 479.9 (M+). Example 161 2-(1-(4-amino-3-(3-(methoxymethyl)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3-(methoxymethyl)phenylboronic acid (0.209 mmol) Brown solid (60.5 mg, 61%). MP: 167-170° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.27 (m, 2H), 7.69-7.23 (m, 8H), 7.04-6.94 (m, 3H), 6.12 (q, J=7.0 Hz, 1H), 5.41 (s, 2H), 3.48 (s, 3H), 2.02 (d, J=7.2 Hz, 3H). Mass: 521.9 (M+). Example 162 2-(1-(4-amino-3-(6-hydroxynaphthalen-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 6-hydroxynaphthalen-2-ylboronic acid (0.209 mmol) Brown solid (32 mg, 31%). MP: 281-285° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.26 (s, 1H), 8.22 (dd, J=9.1, 1.4 Hz, 1H), 8.04 (s, 1H), 7.81 (d, J=8.5 Hz, 2H), 7.72-7.65 (m, 2H), 7.51 (d, J=8.4 Hz, 1H), 7.42 (dt, J=8.0, 0.9 Hz, 1H), 7.32 (m, 1H), 7.19 (m, 2H), 7.05 (d, J=7.5 Hz, 1H), 6.16 (q, J=7.1 Hz, 1H), 5.46 (s, 2H), 2.05 (d, J=7.1 Hz, 3H). Mass: 543.8 (M+). Example 163 2-(1-(4-amino-3-(3-isopropoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3-isopropoxyphenylboronic acid (0.209 mmol) Off-white solid (65 mg, 64%). MP: 153-157° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.24 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 7.69 (m, 1H), 7.48 (d, J=8.2 Hz, 1H), 7.44 (m, 2H), 7.30 (m, 1H), 7.21 (m, 2H), 7.04-6.93 (m, 4H), 6.11 (q, J=7.1 Hz, 1H), 5.46 (s, 2H), 4.63 (quintet, J=6.1 Hz, 1H), 2.02 (d, J=7.1 Hz, 3H), 1.37 (d, J=6.1 Hz, 6H). Mass: 535.9 (M+). Example 164 2-(1-(4-amino-3-(1-methyl-1H-pyrazol-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid (or boronic acid pinacol ester) was replaced by 1-methyl-1H-pyrazol-4-ylboronic acid pinacol ester (0.209 mmol) Yellow semi solid (30 mg, 33%).1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.21 (dd, J=8.0, 1.4 Hz, 1H), 8.18 (s, 1H), 7.78 (s, 1H), 7.73 (s, 1H), 7.69 (m, 1H), 7.48 (d, J=8.1 Hz, 1H), 7.42 (dt, J=7.1, 1.8 Hz, 1H), 7.32 (m, 1H), 7.00-6.93 (m, 3H), 6.06 (q, J=7.1 Hz, 1H), 5.54 (d, J=1.3 Hz, 2H), 4.00 (s, 3H), 2.01 (d, J=7.2 Hz, 3H). Mass: 481.9 (M+). Example 165 6-Fluoro-3-(3-fluorophenyl)-2-(1-(4-methoxyphenylamino)ethyl)-4H-chromen-4-one To a solution of 4-methoxyaniline (0.201 g, 1.637 mmoles) in DMF (5 ml), N,N-diisopropylethylamine (0.158 g, 1.22 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 75 (0.300 g, 0.818 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with ethyl acetate 1:petroleum ether to afford the title compound as brown solid (0.105 g, 31% yield). MP: 152-156° C.1H-NMR (δ ppm, DMSO-D6, 400 MHz): δ 7.73-7.66 (m, 3H), 7.58 (q, J=7.7 Hz, 1H), 7.35 (m, 1H), 7.13 (d, J=7.6 Hz, 2H), 6.61 (d, J=8.9 Hz, 2H), 6.34 (d, J=8.9 Hz, 2H), 5.72 (d, J=9.0 Hz, 1H), 4.22 (q, J=6.9 Hz, 1H), 3.56 (s, 3H), 1.55 (d, J=6.8 Hz, 3H). Mass: 408.27 (M++1). Example 166 2-(1-(4-Chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of 6-chloro-7-deazapurine (0.100 g, 0.651 mmoles) in DMF (4 ml), cesium carbonate (0.424 g, 1.302 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 36 (0.452 g, 1.302 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.105 g, 38% yield). MP: 71-75° C.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.53 (s, 1H), 8.21 (dd, J=7.9, 1.4 Hz, 1H), 7.71 (m, 1H), 7.61 (d, J=3.7 Hz, 1H), 7.44-7.36 (m, 3H), 7.17-7.06 (m, 3H), 6.69 (d, J=3.7 Hz, 1H), 6.14 (q, J=7.2 Hz, 1H), 1.90 (d, J=7.2 Hz, 3H). Mass: 420.10 (M+). Example 167 2-(1-(4-Chloro-1H-pyrazolo[3,4-d]pyridin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of 4-chloro-1H-pyrazolo[3,4-b]pyridine (0.100 g, 0.711 mmoles) in DMF (3 ml), cesium carbonate (0.463 g, 1.422 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 36 (0.495 g, 1.422 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as pale yellow solid (0.080 g, 27% yield). MP: 173-176° C.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.29 (d, J=5.0 Hz, 1H), 8.20 (dd, J=8.0, 1.5 Hz, 1H), 8.11 (s, 1H), 7.68 (m, 1H), 7.47 (d, J=8.5 Hz, 1H), 7.41-7.32 (m, 2H), 7.12 (d, J=5.0 Hz, 1H), 7.03-6.95 (m, 3H), 6.20 (q, J=7.2 Hz, 1H), 2.02 (d, J=7.2 Hz, 3H). Mass: 419.96 (M++1). Example 168 2-(1-(4-Chloro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of 4-chloro-1H-pyrazolo[3,4-d]pyrimidine (0.100 g, 0.745 mmoles) in DMF (3 ml), cesium carbonate (0.485 g, 1.49 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 36 (0.517 g, 1.49 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as brown solid (0.040 g, 13% yield). MP: 197-201° C.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.21 (s, 1H), 8.19 (d, J=1.4 Hz, 1H), 7.96 (s, 1H), 7.66 (m, 1H), 7.50 (d, J=8.4 Hz, 1H), 7.41 (t, J=7.2 Hz, 1H), 7.327 m, 1H), 7.03 (m, 2H), 6.90 (m, 1H), 6.05 (q, J=7.1 Hz, 1H), 1.95 (d, J=7.1 Hz, 3H). Mass: 419.87 (M++1). Example 169 2-(1-(4-Chloro-5H-pyrrolo[3,2-d]pyrimidin-5-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of 4-chloro-5H-pyrrolo[3,2-d]pyrimidine (0.100 g, 0.653 mmoles) in DMF (4 ml), cesium carbonate (0.425 g, 1.30 mmoles) was added and stirred at RT for 10 min. To this mixture intermediate 36 (0.455 g, 1.30 mmoles) was added and stirred for 12 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as light yellow solid (0.080 g, 29% yield). MP: 166-168° C.1H-NMR (δ ppm, CDCl3, 400 MHz): δ 8.66 (s, 1H), 8.24 (dd, J=7.9, 1.4 Hz, 1H), 7.86 (d, J=3.4 Hz, 1H), 7.78 (m, 1H), 7.56 (d, J=8.4 Hz, 1H), 7.48 (t, J=7.7 Hz, 1H), 7.30 (m, 2H), 7.09 (dt, J=8.5, 2.0 Hz, 1H), 6.80 (d, J=3.4 Hz, 1H), 6.74 (m, 1H), 6.50 (q, J=7.1 Hz, 1H), 1.99 (d, J=7.1 Hz, 3H). Mass: 419.89 (M+). Example 170 2-(1-(4-amino-3-(1,3-dimethyl-1H-indazol-6-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.758 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), intermediate 115 (0.309 g, 1.137 mmoles) and sodium carbonate (0.241 g, 2.27 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.071 g, 17% yield). MP: 270-272° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.08 (s, 1H), 8.04 (d, J=6.8 Hz, 1H), 7.84 (m, 2H), 7.69 (s, 1H), 7.69 (d, J=10.8 Hz, 1H), 7.53 (t, J=7.5 Hz, 1H), 7.33 (d, J=8.3 Hz, 2H), 7.03-6.95 (m, 3H), 6.06 (q, J=7.1 Hz, 1H), 3.99 (s, 3H), 2.48 (s, 3H), 1.92 (d, J=7.0 Hz, 3H). Mass: 546.24 (M++1). Example 171 2-(1-(4-amino-3-(2,3-dimethyl-2H-indazol-6-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one To a solution of Example 57c (0.400 g, 0.758 mmoles) in DMF (4 ml), ethanol (2 ml) and water (2 ml), intermediate 116 (0.309 g, 1.137 mmoles) and sodium carbonate (0.241 g, 2.27 mmoles) were added and the system is degassed for 30 min. Tetrakistriphenylphosphine Palladium (0.043 g, 0.037 mmoles) was added under nitrogen atmosphere and heated to 80° C. After 12 h, the reaction mixture was celite filtered, concentrated and extracted with ethyl acetate. The organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by column chromatography with methanol:dichloromethane to afford the title compound as off-white solid (0.100 g, 24% yield). MP: 269-274° C.1H-NMR (δ ppm, DMSO-d6, 400 MHz): δ 8.08 (s, 1H), 8.04 (d, J=8.0, 1.5 Hz, 1H), 7.86 (m, 2H), 7.68 (d, J=4.2 Hz, 2H), 7.53 (t, J=8.8 Hz, 1H), 7.35 (m, 1H), 7.24 (m, 1H), 7.07-6.84 (m, 3H), 6.06 (q, J=6.9 Hz, 1H), 4.07 (s, 3H), 2.64 (s, 3H), 1.92 (d, J=7.1 Hz, 3H). Mass: 546.03 (M++1). Example 172 2-(1-(4-amino-3-(6-methoxynaphthalen-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 6-methoxynaphthalen-2-ylboronic acid (0.209 mmol). Brown solid (44.2 mg, 42%). MP: 285-287° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.26 (s, 1H), 8.22 (dd, J=7.9, 1.5 Hz, 1H), 8.06 (s, 1H), 7.91 (d, J=8.5 Hz, 1H), 7.84 (d, J=7.9 Hz, 1H), 7.76 (dd, J=8.4, 1.7 Hz, 1H), 7.69 (m, 1H), 7.50 (d, J=8.2 Hz, 1H), 7.42 (dt, J=8.0, 0.9 Hz, 1H), 7.31 (m, 1H), 7.24 (m, 2H), 7.06-6.95 (m, 3H), 6.16 (q, J=7.1 Hz, 1H), 5.44 (s, 2H), 3.96 (s, 3H), 2.05 (d, J=7.1 Hz, 3H). Mass: 558.3 (M++1). Example 173 2-(1-(4-amino-3-(benzo[b]thiophen-3-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by benzo[b]thiophen-3-ylboronic acid (0.209 mmol). Brown solid (22.4 mg, 22%). MP: 226-229° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.28 (s, 1H), 8.23 (dd, J=7.9, 1.4 Hz, 1H), 8.01 (d, J=7.6 Hz, 1H), 7.99 (d, J=9.1 Hz, 1H), 7.74 (s, 1H), 7.70 (m, 1H), 7.48-7.37 (m, 6H), 7.11-6.99 (m, 2H), 6.19 (q, J=7.1 Hz, 1H), 5.35 (s, 2H), 2.05 (d, J=7.1 Hz, 3H). Mass: 534.3 (M++1). Example 174 2-(1-(4-amino-3-(2,4-dimethoxypyrimidin-5-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 2,4-dimethoxypyrimidin-5-ylboronic acid (0.209 mmol). Brown solid (28.2 mg, 26%). MP: 286-290° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.25 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 8.05 (s, 1H), 7.87 (d, J=8.5 Hz, 1H), 7.83 (d, J=8.9 Hz, 1H), 7.75-7.65 (m, 2H), 7.50 (d, J=8.2 Hz, 1H), 7.42 (dt, J=8.1, 1.0 Hz, 1H), 7.35 (m, 1H), 7.24 (m, 1H), 7.05-6.75 (m, 4H), 6.14 (q, J=7.2 Hz, 1H), 5.46 (s, 2H), 4.21 (q, J=6.9 Hz, 2H), 2.04 (d, J=7.1 Hz, 3H), 1.52 (t, J+6.9 Hz, 3H). Mass: 572.3 (M++1). Example 175 2-(1-(4-amino-3-(6-ethoxynaphthalen-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 6-ethoxynaphthalen-2-ylboronic acid (0.209 mmol). Brown solid (28.2 mg, 26%). MP: 286-290° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.25 (s, 1H), 8.22 (dd, J=8.0, 1.5 Hz, 1H), 8.05 (s, 1H), 7.87 (d, J=8.5 Hz, 1H), 7.83 (d, J=8.9 Hz, 1H), 7.75-7.65 (m, 2H), 7.50 (d, J=8.2 Hz, 1H), 7.42 (dt, J=8.1, 1.0 Hz, 1H), 7.35 (m, 1H), 7.24 (m, 1H), 7.05-6.75 (m, 4H), 6.14 (q, J=7.2 Hz, 1H), 5.46 (s, 2H), 4.21 (q, J=6.9 Hz, 2H), 2.04 (d, J=7.1 Hz, 3H), 1.52 (t, J+6.9 Hz, 3H). Mass: 572.3 (M++1). Example 176 3-(4-amino-1-(1-(3-(3-fluorophenyl)-4-oxo-4H-chromen-2-yl)ethyl)-1H-pyrazolo[3,4-d] pyrimidin-3-yl)-N-cyclopropylbenzamide The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3-(cyclopropylcarbamoyl) phenylboronic acid (0.209 mmol). Reddish brown solid (47 mg, 44%). MP: 127-132° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.26 (s, 1H), 8.21 (dd, J=7.9, 1.4 Hz, 1H), 8.05 (s, 1H), 7.84 (d, J=7.9 Hz, 1H), 7.69-7.47 (m, 4H), 7.42 (t, J=7.1 Hz, 1H), 7.32 (m, 1H), 7.03-6.93 (m, 3H), 6.32 (s, 1H), 6.13 (q, J=7.1 Hz, 1H), 5.37 (s, 2H), 2.95 (m, 1H), 2.02 (d, J=7.1 Hz, 3H), 0.89 (m, 2H), 0.66 (m, 2H). Mass: 561.3 (M++1). Example 177 2-(1-(4-amino-3-(3-(morpholine-4-carbonyl)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3-(morpholine-4-carbonyl)phenylboronic acid (0.209 mmol). Brown solid (30 mg, 26%). MP: 104-106° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.25 (s, 1H), 8.22 (dd, J=7.9, 1.5 Hz, 1H), 7.77-7.32 (m, 9H), 7.01 (m, 2H), 6.12 (q, J=7.2 Hz, 1H), 5.44 (s, 2H), 3.78-3.55 (m, 8H), 2.01 (d, J=7.1 Hz, 3H). Mass: 591.3 (M++1). Example 178 2-(1-(4-amino-3-(3-(difluoromethoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)ethyl)-3-(3-fluorophenyl)-4H-chromen-4-one The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 3-(difluoromethoxy)phenylboronic acid (0.209 mmol). Brown solid (56 mg, 54%). MP: 176-179° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 8.26 (s, 1H), 8.22 (dd, J=8.0, 1.4 Hz, 1H), 7.70 (m, 1H), 7.55-7.32 (m, 7H), 7.05-6.93 (m, 3H), 6.12 (q, J=7.2 Hz, 1H), 5.42 (s, 2H), 2.02 (d, J=7.1 Hz, 3H). Mass: 544.3 (M++1). Example 179 5-(4-amino-1-(1-(3-(3-fluorophenyl)-4-oxo-4H-chromen-2-yl)ethyl)-1H-pyrazolo[3,4-d]pyrimidin-3-yl)furan-2-carbaldehyde The compound was prepared as per the procedure provided above for Example 130 wherein the 3,5-dimethoxy phenyl boronic acid was replaced by 5-formylfuran-2-ylboronic acid (0.209 mmol). Yellow solid (25 mg, 27%). MP: 215-217° C.1H-NMR (δ ppm, CDCl3, 300 MHz): δ 9.65 (s, 1H), 8.24 (s, 1H), 8.21 (dd, J=7.8 Hz, 1H), 7.69 (m, 1H), 7.45 (m, 3H), 7.32 (m, 1H), 7.17 (d, J=3.7 Hz, 1H), 7.04 (m, 3H), 6.10 (q, J=7.2 Hz, 1H), 1.99 (d, J=7.1 Hz, 3H). Mass: 496.3 (M++1). Biological Assay The pharmacological properties of the compounds of this invention may be confirmed by a number of pharmacological assays. The pharmacological assays which can be been carried out with the compounds according to the invention and/or their pharmaceutically acceptable salts is exemplified below. Assay 1: Fluorescent Determination of PI3Kinase Kinase Enzyme Activity Phosphoinositide 3 kinases (PI3K) belong to a class of lipid kinases that play a critical role in the regulation of several key cellular processes. The PI3K are capable of phosphorylating the 3-hydroxy position of phosphoinositols thereby generating second messengers involved in downstream signalling events. The homogenous time resolved fluorescence (HTRF) assay allows detection of 3,4,5-triphosphate (PIPS) formed as a result of phosphorylation of phosphotidylinositol 4,5-biphosphate (PIP2) by PI3K isoforms such as α, β, γ or δ. PI3K isoform activity for α, β, γ or δ was determined using a PI3K human HTRF™ Assay Kit (Millipore, Billerica, MA) with modifications. All incubations were carried out at room temperature. Briefly, 0.5 μl of 40× inhibitor (in 100% DMSO) or 100% DMSO were added to each well of a 384-well black plate (Greiner Bio-One, Monroe, NC) containing 14.5 μl 1× reaction buffer/PIP2 (10 mM MgCl2, 5 mM DTT, 1.38 μM PIP2) mix with or without enzyme and incubated for 10 mM After the initial incubation, 5 μl/well of 400 μM ATP was added and incubated for an additional 30 minutes. Reaction was terminated by adding 5 μl/well stop solution (Millipore, Billerica, MA). Five microliters of detection mix (Millipore, Billerica, MA) were then added to each well and was incubated for 6-18 h in the dark. HRTF ratio was measured on a microplate reader (BMG Labtech., Germany) at an excitation wavelength of 337 nm and emission wavelengths of 665 and 620 nm with an integration time of 400 μsec. Data were analyzed using Graphpad Prism (Graphpad software; San Diego CA) for IC50determination. Percent inhibition was calculated based on the values for the blank and enzyme controls. The results are provided below in Table 2 & 3. Assay 2: Selectivity for PI3Kδ in Isoform Specific Cell-Based Assays Specificity of test compounds towards PI3Kδ can be confirmed using isoform-specific cell based assays as outlined below:PI3Kα: NIH-3T3 cells were seeded at a concentration of 0.5×106cells per well in a 6-well tissue culture plate and incubated overnight. Complete medium was replaced with serum-free media the following day and compounds at the desired concentrations are to be added. After 15 min, 20 ng/ml PDGF was added and incubated for an additional 10 min. Cells were then lysed and AKT phosphorylation was determined by Western Blotting. Intensity of pAKT bands were normalized based on Actin and Data were analysed using Graphpad Prism (Graphpad software; San Diego CA) and % inhibition due to the test compound compared to the control was calculated accordingly.PI3Kβ: NIH-3T3 cells were seeded at a concentration of 0.5×106cells per well in a 6-well tissue culture plate and incubated overnight. Complete medium was replaced with serum-free media the following day and compounds at the desired concentrations were added. After 15 min 5 μM LPA was added and incubated for an additional 5 min. Cells were lysed and AKT phosphorylation was determined by Western Blotting. Intensity of pAKT bands were normalized based on Actin and Data were analysed using Graphpad Prism (Graphpad software; San Diego CA) and % inhibition due to the test compound compared to the control was calculated accordingly.PI3Kγ: RAW cells were seeded at a concentration of 1×106cells per well in a 6-well tissue culture plate and incubated overnight. Complete medium was replaced with serum-free media the following day and compounds at the desired concentrations were added. After 15 min, 50 ng/ml c5a was added and incubated for an additional 10 min. Cells were lysed and AKT phosphorylation was determined by Western Blotting. Intensity of pAKT bands were normalized based on Actin and data were analysed using Graphpad Prism (Graphpad software; San Diego CA) and % inhibition due to the test compound compared to the control was calculated accordingly.PI3Kδ: Compound specificity towards PI3Kδ was determined in an IgM-induced B cell proliferation assay. Briefly, T-cells were rosetted from human whole blood using sheep RBC and B-cells were separated on a Ficoll-Hypaque gradient. Purified B-cells were seeded at a concentration of 0.1×106cells per well in a 96-well tissue culture plate and incubated with desired concentrations of the test compound for 30 min. Cells were stimulated with 5 μg/ml purified goat anti-human IgM. Growth was assessed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) dye reduction test at 0 h (prior to the addition of the test compound) and 48 h after the addition of test compound. Absorbance was read on a Fluostar Optima (BMG Labtech, Germany) at a wave length of 450 nm. Data were analysed using Graphpad Prism (Graphpad software; San Diego CA) and % inhibition due to the test compound compared to the control was calculated accordingly. Compounds of the present invention when tested at 1 μM did not show any significant inhibition of Pi3kα isoform. TABLE 2P110 δ/Pi3K δCompound% inhibition (1 μM)IC 50 nMExample 1+AExample 2a−−Example 3+−Example 4a+−Example 5−−Example 6+−Example 7+−Example 8−−Example 9+−Example 10−−Example 11+−Example 12+−Example 13+−Example 14+−Example 15+−Example 16+−Example 17a−−Example 18a+−Example 19−−Example 20+−Example 21+−Example 22+−Example 23++AExample 24+−Example 25+−Example 26+−Example 27+−Example 28+−Example 29+−Example 30+−Example 31++BExample 32++AExample 33+−Example 34++−Example 35+−Example 36+−Example 37+−Example 38+−Example 39+−Example 40++−Example 41++−Example 42++AExample 43+−Example 44++−Example 45+−Example 46+−Example 47++AExample 48+−Example 49+−Example 50++−Example 51a++AExample 52+Example 53++AExample 54+−Example 55++AExample 56+−Example 57++AExample 58+−Example 59++BExample 60++AExample 61+−Example 62+−Example 63+−Example 64+−Example 65+−Example 66a++AExample 67+−Example 68++AExample 69++−Example 70++−Example 71+−Example 72++AExample 73++−Example 74++AExample 75++−Example 76a++−Example 77++−+ is less than equal to 50% inhibition at 1 μM;++ is less than equal to 100% inhibition but more than equal to 50% at 1 μM;A represents an IC 50 value of less than equal to 250 nM;B represents an IC50 value of 250-500 nM;C represents an IC50 value of greater than 500 nM. TABLE 2P110 δ/Pi3K δCompound% inhibition*IC 50 nMExample 78+−Example 79++AExample 80+−Example 81+−Example 82++−Example 83++−Example 84++AExample 85++−Example 86a++−Example 87++Example 88a++AExample 89+Example 90+Example 91+Example 92++Example 93++Example 94++Example 95a++AExample 96a++Example 97++Example 98++AExample 99++AExample 100++Example 101+Example 102a+Example 103++AExample 104++AExample 105++AExample 106+Example 107a+Example 108++AExample 109++Example 110++Example 111++Example 112+Example 113++Example 114++Example 115++AExample 116++−Example 117++Example 118+Example 119+Example 120a++Example 121a++Example 122+Example 123+Example 124+Example 125+Example 126a+Example 127a+Example 128−AExample 129−CExample 130+Example 131+Example 132+Example 133+Example 134+Example 135+Example 136+Example 137+Example 138−Example 139+Example 140+Example 141++AExample 142+Example 143+Example 144++AExample 145+Example 146++Example 147+Example 148++Example 149+Example 150+Example 151++AExample 152+Example 153+Example 154+Example 155++Example 156+Example 157+Example 158++AExample 159+Example 160+Example 161+Example 162++Example 163+Example 164+Example 165+Example 166+Example 167+Example 168+Example 169+Example 170++Example 171+Example 172+Example 173++Example 174−Example 175−Example 176+Example 177+Example 178++Example 179+++ is less than equal to 50% inhibition at 1 μM;++ is less than equal to 100% inhibition but more than equal to 50% at 1 μM;A represents an IC 50 value of less than equal to 250 nM;B represents an IC50 value of 250-500 nM;C represents an IC50 value of greater than 500 nM;*@ 0.3 uM. Table 3 TABLE 3% inhibition (1 μM)CompoundP110αP110βP110γExample 1++−Example 2−−−Example 3−−−Example 4−−−Example 5−−−Example 6−−−Example 7−−−Example 8−−−Example 9−−−Example 10++−Example 11−−−Example 12−−−Example 13−−−Example 14−−−Example 15−−Example 16−−−Example 17−+−Example 18−−−Example 19−−−Example 20−−−Example 21−−−Example 22−−−Example 23++−Example 24−−−Example 25−−−Example 26−−−Example 27−−−Example 28−−−Example 29−−−Example 30−−−Example 31−−−Example 32+++Example 33−−−Example 34−−−Example 35−−−Example 36−−−Example 37−−−Example 38−−−Example 39−−−Example 40−−−Example 41−−−Example 42++++Example 43−−−Example 44−−−Example 45−−−Example 46−−−Example 47+++++Example 48−−−Example 49−−−Example 50−−−Example 51+++++Example 52−−−Example 53+++++Example 54−−−Example 55+++++Example 56−−−Example 57+++−Example 58−−−Example 59−−−Example 60−−−Example 61−−−Example 62−−−Example 63−−−Example 64−−−Example 65−−−Example 66−−−Example 67−−−Example 68++++Example 69−−−Example 70−−−Example 71−−−Example 72−−−Example 73−−−Example 74−++++Example 75−−−Example 76−−−Example 77−−−Example 78−−−Example 79+++++Example 80−−−Example 81−−−Example 82−−−Example 83−−−Example 84++−Example 85++++−Example 86Example 87Example 88Example 89Example 90Example 91Example 92Example 93Example 94Example 95Example 96Example 97Example 98Example 99Example 100Example 101Example 102Example 103Example 104Example 105−++++++Example 106−+++Example 107Example 108−+++++Example 109++++Example 110−++++Example 111+++++Example 112++−Example 113−+−Example 114−+−Example 115++−Example 116++−Example 117−+−Example 118+++−Example 119++−Example 120++−Example 121Example 122−+++−Example 123−+−Example 124++−Example 125++−Example 126−+−Example 127−+++−Example 128++++Example 129−++−Example 130++−Example 131+++−Example 132Example 133++++−Example 134−++−Example 135−++−Example 136−++−Example 137−++−Example 138−++−Example 139−++++Example 140−++++Example 141−+++++Example 142−+++Example 143−+++Example 144−+++−Example 145−++−Example 146+−Example 147++−Example 148+++−Example 149++−Example 150++Example 151++Example 152+Example 153+Example 154+Example 155+Example 156++Example 157−Example 158Example 159Example 160Example 161Example 162Example 163Example 164Example 165Example 166Example 167Example 168Example 169Example 170Example 171Example 172Example 173Example 174Example 175Example 176Example 177Example 178Example 179+ is less than equal to 25% inhibition at 1 μM;++ is less than equal to 50% inhibition but more than equal to 25% at 1 μM ;+++ is less than equal to 100% inhibition but more than equal to 50% at 1 μM; Assay 3: In Vitro Cell Proliferation Assay in Leukemic Cell Lines Growth inhibition assays were carried out using 10% FBS supplemented media. Cells were seeded at a concentration of 5000-20,000 cells/well in a 96-well plate. Test compound at a concentration range from 0.01 to 10000 nM were added after 24 h. Growth was assessed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) dye reduction test at 0 h (prior to the addition of the test compound) and 48 h after the addition of test compound. Absorbance was read on a Fluostar Optima (BMG Labtech, Germany) at a wave length of 450 nm. Data were analysed using GraphPad Prism and % inhibition due to the test compound compared to the control was calculated accordingly. Exemplary compounds of the present invention when tested @1 uM in THP-1; DLBCL; HL-60; MDA-MB-468; RPMI8226 and TOLEDO cell lines showed a 20 to 80% inhibition. Assay 4: Determination of Cytotoxicity in Leukemic Cell Lines Cytotoxicity of test compounds was determined using a lactate dehydrogenase assay kit (Cayman Chemicals, MI) as per the manufacturer's instructions with some minor modifications. Briefly, 20,000 cells/well in complete RPMI-1640 media were seeded in a 96-well tissue culture plate and incubated overnight at 37° C. and 5% CO2. Inhibitors were added to the wells in triplicate at the desired concentrations. Doxorubicin and/or 1% Triton-X were used as a positive control. After 48 h, media was removed and assayed for lactate dehydrogenase in a colorimetric assay. Optical density was measured on a microplate reader (BMG Labtech., Germany) at 490 nM. Data were analyzed using Graphpad Prism (Graphpad software; San Diego CA). Results: Exemplary compounds of the present invention were found to be non-toxic when tested @10 uM. Assay 5: Inhibition of PI3Kδ Signalling in Basophils from Human Whole Blood PI3Kδ signalling in basophils manifested by an alteration of anti-FcεR1 induced CD63 expression is a useful pharmacodynamic marker determined using the Flow2CAST® kit (Buhlmann Laboratories, Switzerland). Briefly, it involves the following steps:Mix the anti-coagulated blood sample by inverting the venipuncture tube several timesPrepare fresh and pyrogen-free 3.5 ml polypropylene or polystyrene tubes suitable for Flow Cytometry measurementsAdd 49 μl of patient's whole blood to each tube.Add 1 μl of 10% DMSO (background) or compound (10% DMSO) to the assigned tubes and mix gently. Incubate at room temperature for 15 minPipet 50 μl of the Stimulation buffer (background) or anti-FIERI Ab to each tubeAdd 100 μl of Stimulation Buffer to each tubeMix gently. Add 20 μl Staining Reagent (1:1 mix of FITC-CD63 and PE-CCR3) to each tubeMix gently, cover the tubes and incubate for 15 minutes at 37° C. in a water bath. (using an incubator will take about 10 minutes longer incubation time due to less efficient heat transfer)Add 2 ml pre-warmed (18-28° C.) Lysing Reagent to each tube, mix gentlyIncubate for 5-10 minutes at 18-28° C.Centrifuge the tubes for 5 minutes at 500×gDecant the supernatant by using blotting paperResuspend the cell pellet with 300-800 μl of Wash BufferVortex gently and acquire the data on the flow cytometer within the same day.Percent CD63 positive cells within the gated basophil population are to be determined in different treatment groups and normalized to vehicle control. Assay 6: Inhibition of Apoptosis in Leukemic Cell Lines Apoptosis in leukemic cells was determined using an in-situ Caspase 3 kit (Millipore, US) as outlined below:Seed leukemic cells—at a density of 1×106cells/well in a 6 well plateAdd test compound/DMSO at desired concentrationsIncubate the plate for 24 hrs at 37° C. in 5% CO2incubatorCollect cells in a 2 ml centrifuge tubeAdd 1.6 μL of freshly prepared 5× FLICA reagent and mix cells by slightly flicking the tubesIncubate tubes for 1 hour at 37° C. under 5% CO2Add 2 ml of 1× wash buffer to each tube and mixCentrifuge cells at <400×g for 5 minutes at room temperature.Carefully remove and discard supernatant, and gently vortex cell pellet to disrupt any cell-to-cell clumping.Resuspend cell pellet in 300 ul of 1× wash bufferPlace 100 μL of each cell suspension into each of two wells of a black microtiter plate. Avoid creation of bubbles.Read absorbance of each microwell using an excitation wavelength of 490 nm and an emission wavelength of 520 nmPercent increase in caspase-3 activity manifested by an increase in fluorescence compared to the control blank is to be calculated. Although the invention herein has been described with reference to particular embodiments, it is to be understood that these embodiments are merely illustrative of the principles and applications of the present invention. It is therefore to be understood that numerous modifications may be made to the illustrative embodiments and that other arrangements may be devised without departing from the spirit and scope of the present invention as described above. It is intended that the appended claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby. All publications and patent and/or patent applications cited in this application are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated herein by reference.
437,901
11858908
DETAILED DESCRIPTION OF THE INVENTION I. Definitions The following terms are intended to have the meanings presented therewith below and are useful in understanding the description and intended scope of the present invention. When describing the invention, which may include compounds, pharmaceutical compositions containing such compounds and methods of using such compounds and compositions, the following terms, if present, have the following meanings unless otherwise indicated. It should also be understood that when described herein any of the moieties defined forth below may be substituted with a variety of substituents, and that the respective definitions are intended to include such substituted moieties within their scope as set out below. Unless otherwise stated, the term ‘substituted’ is to be defined as set out below. It should be further understood that the terms “groups” and “radicals” can be considered interchangeable when used herein. The articles “a” and “an” may be used herein to refer to one or to more than one (i.e., at least one) of the grammatical objects of the article. By way of example “an analogue” means one analogue or more than one analogue. As used herein, the term “pharmaceutical composition” means a mixture comprising a pharmaceutically acceptable active ingredient, in combination with suitable pharmaceutically acceptable excipients. In one embodiment the pharmaceutically acceptable ingredient is a pharmaceutically acceptable acid addition salt of the compound of formula I, or a solvate or hydrate of this acid addition salt. Pharmaceutical excipients are substances other than the pharmaceutically acceptable active ingredient which have been appropriately evaluated for safety and which are intentionally included in an oral solid dosage form. For example, excipients can aid in the processing of the drug delivery system during its manufacture, protect, support or enhance stability, bioavailability or patient acceptability, assist in product identification, or enhance any other attribute of the overall safety, effectiveness or delivery of the drug during storage or use. Examples of excipients include, for example but without limitation inert solid diluents (bulking agent e.g., lactose), binders (e.g., starch), glidants (e.g., colloidal silica), lubricants (e.g., non-ionic lubricants such as vegetable oils), disintegrants (e.g., starch, polivinylpyrrolidone), coating better polymers (e.g., hydroxypropyl methylcellulose), colorants (e.g., iron oxide), and/or surfactants (e.g., non-ionic surfactants). As used herein, the term “pharmaceutical formulation” means a composition in which different chemical substances, including the active drug, are combined to produce a final medicinal product. Examples of formulation include enteral formulations (tablets, capsules), parenteral formulations (liquids, lyophilized powders), or topical formulations (cutaneous, inhalable). “Pharmaceutically acceptable” means approved or approvable by a regulatory agency of the Federal or a state government or the corresponding agency in countries other than the United States, or that is listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly, in humans. “Pharmaceutically acceptable salt” refers to a salt of a compound of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or derivatives thereof that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. In particular, such salts are non-toxic may be inorganic or organic acid addition salts and base addition salts. Specifically, such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g. an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine and the like. Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the compound contains a basic functionality, salts of non-toxic organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like. More particularly, such salts are formed with hydrobromic acid, hydrochloric acid, sulfuric acid, toluenesulfonic acid, benzenesulfonic acid, oxalic acid, maleic acid, naphthalene-2-sulfonic acid, naphthalene-1,5-disulfonic acid, 1-2-ethane disulfonic acid, methanesulfonic acid, 2-hydroxy ethanesulfonic acid, phosphoric acid, ethane sulfonic acid, malonic acid, 2-5-dihydroxybenzoic acid, or L-Tartaric acid; and (3) salts formed when a proton is removed from the parent compound, such that an anion of the compound is formed that can pair with a suitable cation to form the salt. The term “pharmaceutically acceptable cation” refers to an acceptable cationic counter-ion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium cations, and the like. “Pharmaceutically acceptable vehicle” refers to a diluent, adjuvant, excipient or carrier with which a compound described herein is administered. “Solvate” refers to forms of the compound that are associated with a solvent, usually by a solvolysis reaction. This physical association includes hydrogen bonding. Conventional solvents include water, ethanol, acetic acid and the like. The compounds described herein may be prepared e.g. in crystalline form and may be solvated or hydrated. Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates. In certain instances, the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. ‘Solvate’ encompasses both solution-phase and isolable solvates. Representative solvates include hydrates, ethanolates and methanolates. The terms “inert solid diluent” or “solid diluent” or “diluents” refer to materials used to produce appropriate dosage form size, performance and processing properties for tablets and/or capsules. An inert solid diluent can be also referred to as filler or filler material. Particular examples of diluents include cellulose powdered, silicified microcrystalline cellulose acetate, compressible sugar, confectioner's sugar, corn starch and pregelatinized starch, dextrates, dextrin, dextrose, erythritol, ethylcellulose, fructose, fumaric acid, glyceryl palmitostearate, inhalation lactose, isomalt, kaolin, lactitol, lactose, anhydrous, monohydrate and corn starch, spray dried monohydrate and microcrystalline cellulose, maltodextrin, maltose, mannitol, medium-chain triglycerides, microcrystalline cellulose, polydextrose, polymethacrylates, simethicone, sorbitol, pregelatinized starch, sterilizable maize, sucrose, sugar spheres, sulfobutylether β-cyclodextrin, talc, tragacanth, trehalose, or xylitol. More particular examples of diluents include cellulose powdered, silicified microcrystalline cellulose acetate, compressible sugar, corn starch and pregelatinized starch, dextrose, fructose, glyceryl palmitostearate, anhydrous, monohydrate and corn starch, spray dried monohydrate and microcrystalline cellulose, maltodextrin, maltose, mannitol, medium-chain triglycerides, microcrystalline cellulose, polydextrose, sorbitol, starch, pregelatinized, sucrose, sugar spheres, trehalose, or xylitol. “Lubricant” refers to materials that prevent or reduce ingredients from clumping together and from sticking to the tablet punches or capsule filling machine. Lubricants also ensure that tablet formation and ejection can occur with low friction between the solid and die wall. Particular examples of lubricants include canola oil, hydrogenated castor oil, cottonseed oil, glyceryl behenate, glyceryl monostearate, glyceryl palmitostearate, medium-chain triglycerides, mineral oil, light mineral oil, octyldodecanol, poloxamer, polyethylene glycol, polyoxyethylene stearates, polyvinyl alcohol, starch, or hydrogenated vegetable oil. More particular examples of diluents include glyceryl behenate, glyceryl monostearate, or hydrogenated vegetable oil. “Disintegrant” refers to material that dissolve when wet causing the tablet to break apart in the digestive tract, releasing the active ingredients for absorption. They ensure that when the tablet is in contact with water, it rapidly breaks down into smaller fragments, facilitating dissolution. Particular examples of disintegrants include alginic acid, powdered cellulose, chitosan, colloidal silicon dioxide, corn starch and pregelatinized starch, crospovidone, glycine, guar gum, low-substituted hydroxypropyl cellulose, methylcellulose, microcrystalline cellulose, or povidone. The term “colorant” describes an agent that imparts color to a formulation. Particular examples of colorants include iron oxide, or synthetic organic dyes (US Food and Drug administration, Code of Federal Regulations, Title 21 CFR Part73, Subpart B). The term “plasticizing agent” or “plasticizer” refers to an agent that is added to promote flexibility of films or coatings. Particular examples of plasticizing agent include polyethylene glycols or propylene glycol. The term “pigment” used herein refers to an insoluble coloring agent. The term “film-coating agent” or ‘coating agent’ or ‘coating material’ refers to an agent that is used to produce a cosmetic or functional layer on the outer surface of a dosage form. Particular examples of film-coating agent include glucose syrup, maltodextrin, alginates, or carrageenan. “Glidant” refers to materials that are used to promote powder flow by reducing interparticle friction and cohesion. These are used in combination with lubricants as they have no ability to reduce die wall friction. Particular examples of glidants include powdered cellulose, colloidal silicon dioxide, hydrophobic colloidal silica, silicon dioxide, or talc. More particular examples of glidants include colloidal silicon dioxide, hydrophobic colloidal silica, silicon dioxide, or talc. “Flavoring agents” refers to material that can be used to mask unpleasant tasting active ingredients and improve the acceptance that the patient will complete a course of medication. Flavorings may be natural (e.g., fruit extract) or artificial. Non limiting examples of flavoring agents include mint, cherry, anise, peach, apricot, licorice, raspberry, or vanilla. The term “subject” includes mammals such a human, a dog, a cat, a rat, a monkey, rabbits, guinea pigs, etc. The terms “human”, “patient” and “subject” are used interchangeably herein. “Effective amount” means the amount of a compound described herein that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease. The ‘effective amount’ can vary depending on the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated. “Preventing” or “prevention” refers to a reduction in risk of acquiring or developing a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to a disease-causing agent, or predisposed to the disease in advance of disease onset). The term “prophylaxis” is related to “prevention”, and refers to a measure or procedure the purpose of which is to prevent, rather than to treat or cure a disease. Non-limiting examples of prophylactic measures may include the administration of vaccines; the administration of low molecular weight heparin to hospital patients at risk for thrombosis due, for example, to immobilization; and the administration of an anti-malarial agent such as chloroquine, in advance of a visit to a geographical region where malaria is endemic or the risk of contracting malaria is high. “Treating” or “treatment” of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting the disease or reducing the manifestation, extent or severity of at least one of the clinical symptoms thereof). In another embodiment “treating” or “treatment” refers to ameliorating at least one physical parameter, which may not be discernible by the subject. In yet another embodiment, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In a further embodiment, “treating” or “treatment” relates to slowing the progression of the disease. As used herein, the term “isotopic variant” refers to a compound that contains unnatural proportions of isotopes at one or more of the atoms that constitute such compound. For example, an “isotopic variant” of a compound can contain one or more non-radioactive isotopes, such as for example, deuterium (2H or D), carbon-13 (13C), nitrogen-15 (15N), or the like. It will be understood that, in a compound where such isotopic substitution is made, the following atoms, where present, may vary, so that for example, any hydrogen may be2H/D, any carbon may be13C, or any nitrogen may be15N, and that the presence and placement of such atoms may be determined within the skill of the art. Likewise, the compounds described herein may include isotopic variants with radioisotopes, in the instance for example, where the resulting compounds may be used for drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e.,3H, and carbon-14, i.e.14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Further, compounds may be prepared that are substituted with positron emitting isotopes, such as11C,18F,15O and13N, and would be useful in Positron, and 13 Emission Topography (PET) studies for examining substrate receptor occupancy. All isotopic variants of the compounds provided herein, radioactive or not, are intended to be encompassed within the scope of the compound. “Tautomers” refer to compounds that are interchangeable forms of a particular compound structure, and that vary in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of π electrons and an atom (usually H). For example, enols and ketones are tautomers because they are rapidly interconverted by treatment with either acid or base. Another example of tautomerism is the aci- and nitro-forms of phenylnitromethane, that are likewise formed by treatment with acid or base. Tautomeric forms may be relevant to the attainment of the optimal chemical reactivity and biological activity of a compound of interest. The term “alkyl” as used herein, whether used alone or as part of another group, refers to a substituted or unsubstituted aliphatic hydrocarbon chain and includes, but is not limited to, straight and branched chains containing from 1 to 20 carbon atoms, preferably from 2 to 20, from 1 to 10, from 2 to 10, from 1 to 8, from 2 to 8, from 1 to 6, from 2 to 6, from 1 to 4, from 2 to 4, from 1 to 3 carbon atoms, unless explicitly specified otherwise. Illustrative alkyl groups can include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, t-butyl, isobutyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl), hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl, 2,2,4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2-methyl-1-pentyl, 2,2-dimethyl-1-propyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, and the like. The term “alkenyl” as used herein, whether used alone or as part of another group, refers to a substituted or unsubstituted aliphatic hydrocarbon chain and includes, but is not limited to, straight and branched chains having 2 to 8 carbon atoms and containing at least one carbon-carbon double bond. The term “alkynyl” as used herein, whether used alone or as part of another group, refers to a substituted or unsubstituted aliphatic hydrocarbon chain and includes, but is not limited to, straight and branched chains having 1 to 6 carbon atoms and containing at least one carbon-carbon triple bond. The term “alkoxy” as used herein, whether used alone or as part of another group, refers to alkyl-O— wherein alkyl is hereinbefore defined. The term “cycloalkyl” as used herein, whether used alone or as part of another group, refers to a monocyclic, bicyclic, tricyclic, fused, bridged or spiro monovalent saturated hydrocarbon moiety, wherein the carbon atoms are located inside or outside of the ring system. Any suitable ring position of the cycloalkyl moiety may be covalently linked to the defined chemical structures. Illustrative cycloalkyl groups can include, but are not limited to, cyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexylmethyl, cyclohexylethyl, cycloheptyl, norbornyl, adamantly, spiro[4,5]decanyl, and homologs, isomers and the alike. The term “aryl” as used herein, whether used alone or as part of another group, refers to an aromatic carbocyclic ring system having 6 to 30 carbon atoms, preferably 6 to 10 carbon atoms, optionally substituted with 1 to 3 substituents independently selected from halogen, nitro cyano, hydroxy, alkyl, alkenyl, alkoxy, cycloalkyl, amino, alkylamino, dialkylamino, carboxy, alkoxycarbonyl, haloalkyl, and phenyl. The term “phenyl” as used herein, whether used alone or as part of another group, refers to a substituted or unsubstituted phenyl group. The term “heteroaryl” as used herein, whether used alone or as part of another group, refers to a 3 to 30 membered aryl heterocyclic ring, which contains from 1 to 4 heteroatoms selected from the group consisting of O, N, Si, P and S atoms in the ring and may be fused with a carbocyclic or heterocyclic ring at any possible position. The term “heterocycloalkyl” as used herein, whether used alone or as part of another group, refers to a 5 to 7 membered saturated ring containing carbon atoms and from 1 to 2 heteroatoms selected from the group consisting of O, N and S atoms. The term “halogen or halo” as used herein, refers to fluoro, chloro, bromo or iodo. The term “haloalkyl” as used herein, whether used alone or as part of another group, refers to an alkyl as hereinbefore defined, independently substituted with 1 to 3, F, Cl, Br or I. “Substituted,” as used herein, refers to all permissible substituents of the functional groups described herein. In the broadest sense, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Illustrative sub stituents include, but are not limited to, halogens, hydroxyl groups, or any other organic groupings containing any number of carbon atoms, preferably 1-14 carbon atoms, and optionally include one or more heteroatoms such as oxygen, sulfur, or nitrogen grouping in linear, branched, or cyclic structural formats. Representative substituents include a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted heterocyclyl, a substituted or unsubstituted phenyl, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted polyaryl, a substituted or unsubstituted polyheteroaryl, a substituted or unsubstituted aralkyl, a halogen, a hydroxyl, an alkoxy, a phenoxy, an aroxy, a silyl, a thiol, an alkylthio, a substituted alkylthio, a phenylthio, an arylthio, a cyano, an isocyano, a nitro, a substituted or unsubstituted carbonyl, a carboxyl, an amino, an amido, an oxo, a sulfinyl, a sulfonyl, a sulfonic acid, a phosphonium, a phosphanyl, a phosphoryl, a phosphonyl, an amino acid. Such a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted heterocyclyl, a substituted or unsubstituted phenyl, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted polyaryl, a substituted or unsubstituted polyheteroaryl, a substituted or unsubstituted aralkyl, a halogen, a hydroxyl, an alkoxy, a phenoxy, an aroxy, a silyl, a thiol, an alkylthio, a substituted alkylthio, a phenylthio, an arylthio, a cyano, an isocyano, a nitro, a substituted or unsubstituted carbonyl, a carboxyl, an amino, an amido, an oxo, a sulfinyl, a sulfonyl, a sulfonic acid, a phosphonium, a phosphanyl, a phosphoryl, a phosphonyl, and an amino acid can be further substituted. The disclosed compounds and substituent groups, can, independently, possess two or more of the groups listed above. For example, if the compound or substituent group is a straight chain alkyl group, one of the hydrogen atoms of the alkyl group can be substituted with a hydroxyl group, an alkoxy group, etc. Depending upon the groups that are selected, a first group can be incorporated within second group or, alternatively, the first group can be pendant (i.e., attached) to the second group. For example, with the phrase “an alkyl group comprising an ester group,” the ester group can be incorporated within the backbone of the alkyl group. Alternatively, the ester can be attached to the backbone of the alkyl group. The nature of the group(s) that is (are) selected will determine if the first group is embedded or attached to the second group. The term “small molecule inhibitors or antagonists” as used herein, refers to inhibitors or antagonists that have a molecular weight in a range from 0.1 kDa to 1 kDa. The term “about” as used herein, refers that the numerical value is approximate and small variations would not significantly affect the practice of the disclosed embodiments. Where a numerical limitation is used, unless indicated otherwise by the context, “about” means the numerical value can vary by ±10% and remain within the scope of the disclosed embodiments. Additionally, in phrase “about X to Y,” is the same as “about X to about Y,” that is the term “about” modifies both “X” and “Y.” The term “compound” as used herein, refers to salts, solvates, complexes, isomers, stereoisomers, diastereoisomers, tautomers, and isotopes of the compound or any combination thereof. The term “comprising” (and any form of comprising, such as “comprise”, “comprises”, and “comprised”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”), or “containing” (and any form of containing, such as “contains” and “contain”), are used in their inclusive, open-ended, and non-limiting sense. The term “racemic” as used herein refers to a mixture of the (+) and (−) enantiomers of a compound wherein the (+) and (−) enantiomers are present in approximately a 1:1 ratio. The terms “substantially optically pure,” “optically pure,” and “optically pure enantiomers,” as used herein, mean that the composition contains greater than about 90% of a single stereoisomer by weight, preferably greater than about 95% of the desired enantiomer by weight, and more preferably greater than about 99% of the desired enantiomer by weight, based upon the total weight. The term “enantiomer” refers to a stereoisomer that is a non-superimposable mirror image of each other. A diastereomer is a stereoisomer with two or more stereocenters, and the isomers are not mirror images of each other. A hallmark of aging is the progressive decline in skeletal muscle function, characterized by reduced force generating capacity and loss of muscle mass. These phenomena, referred to collectively as sarcopenia, are common in aged humans and animal models. Moreover, age-dependent deterioration of muscle function is not restricted to mammals as it is also observed in the nematodeCaenorhabditis elegans(C. elegans). It is estimated that sarcopenia affects as many as 45% of the population over the age of 60, leading to profound loss of function in the elderly. Indeed, loss of muscular strength is highly predictive of frailty, and disability, all of which cause mortality with increased age. Much attention is focused on understanding how to reverse muscle wasting, and significant advances in this field have led to early clinical trials targeting muscle growth, but there are no established treatments for age-related loss of muscle mass at this time. In contrast, improving specific force production, which is also significantly reduced in aged muscle, has received less attention. The loss of specific force suggests that the calcium-(Ca2+) dependent process known as excitation-contraction (EC) coupling may be impaired in aged muscle. During EC coupling in skeletal muscle, muscle membrane depolarization activates voltage sensing channels in the transverse tubules (Cav1.1) which in turn activate the sarcoplasmic reticulum (SR) Ca2+release channel, also known in skeletal muscle as the ryanodine receptor 1 (RyR1). The release of SR Ca2+via RyR1 raises cytoplasmic [Ca2+]cytleading to activation of actin-myosin cross-bridging and shortening of the sarcomere, manifesting as muscle contraction. Impaired Ca2+handling is associated with contractile dysfunction in heart failure and muscular dystrophy, and sarcopenic skeletal muscle is reported to have decreased SR Ca2+release. Thus, proper Ca2+handling in muscle plays a key role in normal EC coupling and specific force production. Cysteine nitrosylation (SNO) and carbonyl modifications of proteins are emerging as important cellular mediators for RyR function and Ca2+signaling. Excessive SNO-modification of RyR1 disrupts the interaction between RyR1 and calstabin1 (also known as FKBP12 in skeletal muscle). Loss of the RyR1/Calstabin1 interaction results in channels that leak SR Ca2+. This leak leads to reduced SR Ca2+release and muscle function. Currently, the primary treatment for sarcopenia is exercise. Specifically, resistance training or strength training—exercises that increase muscle strength and endurance with weights or resistance bands—are shown to be beneficial for both the prevention and treatment of sarcopenia. Resistance training is reported to positively influence the neuromuscular system, hormone concentrations, and protein synthesis rate. Research show that an exercise program of progressive resistance training can increase protein synthesis rates in the elderly in as little as two weeks. While this is possible for patients who are otherwise generally in good health and capable of conducting such exercise, it is not possible for a certain segment of the population to continually and properly follow an exercise regimen. Current interventions for sarcopenia are focused on ways to increase muscle mass and/or reduce wasting of the aged muscle. This focus includes therapeutic regimens that utilize anabolic pathways such as testosterone, growth hormone, and insulin-like growth factor-1 signaling. Some trials with these anabolic regimens demonstrate modest increase in muscle growth but no increase in muscle strength or power. Inhibition of the endogenous negative regulator of myogenesis, myostatin (growth differentiation factor 8), has emerged as an attractive target for combating muscle weakness diseases as mutations in myostatin that inactivate or reduce its function lead to a dramatic increase of muscle girth in mice, dogs, and cattle. However, muscular dystrophy patients treated with an anti-myostatin recombinant human antibody, which inactivates the function of myostatin, failed to improve muscle power. Interestingly, the muscular dystrophy mouse model associated with dystrophinopathy. It will be appreciated that compounds described herein may be metabolized to yield biologically active metabolites. II. Inhibitors of IDO1 In some aspects, the IDO1 small molecule inhibitors or antagonists can be a compound of Formula Ia: wherein:X, X′, P, P′, Q, Q′, G, G′, J, J′, E, and E′ are independently C, N, O or S;R1-R3, R6-R9, and R13-R17are independently absent, H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted C3-10cycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl, or two neighboring R groups together form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N;L1and L2are independently a linker, optionally the linker is a substituted or unsubstituted C1-10alkyl or substituted or unsubstituted C1-10alkoxy; andY is H, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted aralkyl (e.g. a benzyl);or an enantiomer, tautomer, stereoisomers, solvate, zwitterion, polymorph, prodrug, or a pharmaceutically acceptable salt thereof. In some aspects of Formula Ia, L1and L2are independently wherein m is an integer from 0 to 10; R4and each occurrence of R5is independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a carbonyl (e.g., an aldehyde, a ketone, a carboxylic acid, a carboxylate ester, etc.), an amino group, an amide group, a haloalkyl, a nitro group, a nitrile group, a cyanate, an isocyanate, or r is an integer from 1 to 6, from 1 to 5, from 1 to 4, from 1 to 3, from 1 to 2, or 1, R18is a carbonyl (e.g., an aldehyde (—COH), a ketone, a carboxylic acid (—COOH), a carboxylate ester (—COOR10, etc.), a hydroxyl, an alkoxy (—OR10), a halogen, an amino group (e.g. a primary amino NH2, a secondary amino NHR11, or a tertiary amino NR11R12), an amide group (—CONR11R12), a nitro group, a nitrile group, R19and R20are independently H or a substituted or unsubstituted C1-10alkyl, and R10, R11and R12are independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl. In some aspects, R19and R20are H. In some aspects, R18is a carbonyl (e.g., an aldehyde (—COH), a ketone, a carboxylic acid (—COOH), a carboxylate ester (—COOR10, etc.), a hydroxyl, an alkoxy (—OR10), a halogen, or an amino group (e.g. a primary amino NH2, a secondary amino NHR11, or a tertiary amino NR11R12). In some aspects, the IDO1 small molecule inhibitor or antagonist contains one or more stereocenters. For example, the stereocenter is one or both of the C* in the linker L1and L2having the structure of In some forms of Formula Ia, X, X′, P, P′, Q, Q′, G, G′, J, J′, E, and E′ are independently C or N. In some aspects, the IDO1 small molecule inhibitors or antagonists can be a compound of Formula Ib: wherein:X, P, Q, G, J, and E are independently C, N, O or S;R1-R3, R6-R9, R13, R14, and R16are independently absent, H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted C3-10cycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl, or two neighboring R groups together form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N;R4and each occurrence of R5is independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a carbonyl (e.g., an aldehyde, a ketone, a carboxylic acid, a carboxylate ester, etc.), an amino group, an amide group, a haloalkyl, a nitro group, a nitrile group, a cyanate, an isocyanate, or r is an integer from 1 to 6, from 1 to 5, from 1 to 4, from 1 to 3, from 1 to 2, or 1, R18is a carbonyl (e.g., an aldehyde (—COH), a ketone, a carboxylic acid (—COOH), a carboxylate ester (—COOR10, etc.), a hydroxyl, an alkoxy (—OR10), a halogen, an amino group (e.g. a primary amino NH2, a secondary amino NHR11, or a tertiary amino NR11R12), an amide group (—CONR11R12), a nitro group, a nitrile group, R19and R20are independently H or a substituted or unsubstituted C1-10alkyl, and R10, R11and R12are independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl;Y is H, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted aralkyl (e.g. a benzyl);m is an integer from 0 to 10, from 0 to 8, or from 0 to 6, such as 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; andn is an integer from 1 to 6, such as 1, 2, 3, 4, 5 or 6;or an enantiomer, tautomer, stereoisomers, solvate, zwitterion, polymorph, prodrug, or a pharmaceutically acceptable salt thereof. In some forms of Formula Ib, X, P, Q, G, J, and E are independently C or N. In one aspect, the IDO1 small molecule inhibitors or antagonists can be a compound of Formula Ic: wherein:X is C, N, O or S;R1-R3, R6-R9, R13, R14, and R16are independently absent, H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted C3-10cycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl, or two neighboring R groups together form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N;R4and each occurrence of R5is independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a carbonyl (e.g., an aldehyde, a ketone, a carboxylic acid, a carboxylate ester, etc.), an amino group, an amide group, a haloalkyl, a nitro group, a nitrile group, a cyanate, an isocyanate, or r is an integer from 1 to 6, from 1 to 5, from 1 to 4, from 1 to 3, from 1 to 2, or 1, R18is a carbonyl (e.g., an aldehyde (—COH), a ketone, a carboxylic acid (—COOH), a carboxylate ester (—COOR10, etc.), a hydroxyl, an alkoxy (—OR10), a halogen, an amino group (e.g. a primary amino NH2, a secondary amino NHR11, or a tertiary amino NR11R12), an amide group (—CONR11R12), a nitro group, a nitrile group, R19and R20are independently H or a substituted or unsubstituted C1-10alkyl, and R10, R11and R12are independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl;Y is H, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted aralkyl (e.g. a benzyl);m is an integer from 0 to 10, from 0 to 8, or from 0 to 6, such as 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; andn is an integer from 1 to 6, such as 1, 2, 3, 4, 5 or 6;or an enantiomer, tautomer, stereoisomers, solvate, zwitterion, polymorph, prodrug, or a pharmaceutically acceptable salt thereof. In some aspects of Formula Ib and Formula Ic, R4and each occurrence of R5are independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, COOH, —COOR10, —CONH2, —NCO, —CHO, —CN, NO2, NH2, NHR11, NR11R12, or r is an integer from 1 to 6, R18is —COOH, —COOR10, —OH, —OR10, —NH2, NHR11, NR11R12, halogen, and R10-R12, are independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl. In some aspects of Formula Ib and Formula Ic, R4and each occurrence of R5are independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, COOH, —COOR10, —CH2COOH; —CH2COOR10, CH2OH, —CH2OR10, —CONH2, —CH2NH2, CH2NHR11, —CH2N11R12, NCO, —CH2-halogen, —CHO, —CN, NO2, NH2, NHR11, NR11R12, and R10-R12are independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl. In some aspects of Formula Ib and/or Formula Ic, R2and R3together and/or R13and R14together form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N, and/or R16and R6together, R6and R7together, R7and R8together, and/or R8and R9together, form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N. In some aspects of Formula Ib and/or Formula Ic, R2and R3together form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N. In some aspects of Formula Ib and/or Formula Ic, R6and R7together or R7and R8together form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N. In some aspects of Formula Ib and/or Formula Ic, R2and R3together form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N, and/or R6and R7together or R7and R8together form a substituted or unsubstituted 3-6 membered carbocycle or substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N. One aspect provides a compound of Formula I: whereinX is each independently C, CH or N;R1is independently absent, H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted C3-10cycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl;R2and R3together form a substituted or unsubstituted 3-6 membered carbocycle or a substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N; orR2and R3are each independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl or a substituted or unsubstituted heteroaryl;R4and each occurrence of R5is independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a carbonyl (e.g., an aldehyde, a ketone, a carboxylic acid, a carboxylate ester, etc.), an amino group, an amide group, a haloalkyl, a nitro group, a nitrile group, a cyanate, an isocyanate, or r is an integer from 1 to 6, from 1 to 5, from 1 to 4, from 1 to 3, from 1 to 2, or 1, R18is a carbonyl (e.g., an aldehyde (—COH), a ketone, a carboxylic acid (—COOH), a carboxylate ester (—COOR10, etc.), a hydroxyl, an alkoxy (—OR10), a halogen, an amino group (e.g. a primary amino NH2, a secondary amino NHR11, or a tertiary amino NR11R12), an amide group (—CONR11R12), a nitro group, a nitrile group, R19and R20are independently H or a substituted or unsubstituted C1-10alkyl, and R10, R11and R12are independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl;R6and R7together may form a substituted or unsubstituted 3-6 membered carbocycle or a substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N; orR7and R8together may form a substituted or unsubstituted 3-6 membered carbocycle or a substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N; orR8and R9together may form a substituted or unsubstituted 3-6 membered carbocycle or a substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N; orR6, R7, R8and R9are each independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl or a substituted or unsubstituted heteroaryl; m is an integer from 0 to 10, from 0 to 8, or from 0 to 6, such as 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;n is an integer from 1 to 6, such as 1, 2, 3, 4, 5 or 6; andY is H, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted aralkyl (e.g. a benzyl);or an enantiomer, tautomer, stereoisomers, solvate, zwitterion, polymorph, prodrug, or a pharmaceutically acceptable salt thereof. In some aspects of Formula I, R4and each occurrence of R5are independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, COOH, —COOR10, —CONH2, —NCO, —CHO, —CN, NO2, NH2, NHR11, NR11R12, or r is an integer from 1 to 6, R18is —COOH, —COOR10, —OH, —OR10, —NH2, NHR11, NR11R12, halogen, and R10-R12, are independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl. In some aspects of Formula I, R4and each occurrence of R5are independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, COOH, —COOR10, CH2COOH; —CH2COOR10, CH2OH, —CH2OR10, —CONH2, —CH2NH2, CH2NHR11, —CH2N11R12, NCO, —CH2-halogen, —CHO, —CN, NO2, NH2, NHR11, NR11R12, and R10-R12are independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl. In some aspects of Formula Ib, Formula Ic, and/or Formula I, R6and R7together or R7and R8together form a substituted or unsubstituted 4-6 membered heterocyclyl containing one or more heteroatom selected from the group consisting of O, S and N. In some aspects of Formula Ib, Formula Ic, and/or Formula I, R6and R7together or R7and R8together form a substituted or unsubstituted 4-6 membered heterocyclyl containing one or more O and/or one or more N. In some aspects of Formula Ib, Formula Ic, and/or Formula I, R6and R7together or R7and R8together form a substituted or unsubstituted 5 membered heterocyclyl containing one or more O, such as two O. In other embodiments, R1is H, a substituted or unsubstituted C1-10alkyl (e.g. an unsubstituted C1-10alkyl), a substituted or unsubstituted C1-10alkoxy, or halogen. In another embodiments, R2and R3are H, a substituted or unsubstituted C1-10alkyl (e.g. an unsubstituted C1-10alkyl), a substituted or unsubstituted C1-10alkoxy, or R2and R3together form a substituted or unsubstituted 6 membered aryl ring. In some embodiment, R4and each occurrence of R5are independently H, —COOH, —COOR10, or r is an integer from 1 to 6, R18is —COOH or —COOR10, and R10is independently H or a substituted or unsubstituted C1-10alkyl, such as an unsubstituted C1-10alkyl. In some embodiment, R4and each occurrence of R5are independently H, COOH or CH2COOH. In other embodiment, R6and R7together form a substituted or unsubstituted 5 membered heterocyclyl containing one or more O, such as an unsubstituted 5 membered heterocyclyl containing one or more O. In another embodiment, R7and R8together form a substituted or unsubstituted 5 membered heterocyclyl containing one or more O, such as an unsubstituted 5 membered heterocyclyl containing one or more O. In further embodiment, R8and R9together form a substituted or unsubstituted 5 membered heterocyclyl containing one or more O, such as an unsubstituted 5 membered heterocyclyl containing one or more O. In other embodiment, X is CH or N. In one aspect, the IDO1 small molecule inhibitors or antagonists can be a compound of Formula IIa or Formula IIb: whereinX is each independently C or N;R1is independently absent, H, OH, halogen, an unsubstituted C1-10alkyl, or an unsubstituted C1-10alkoxy;R2and R3together form a substituted or unsubstituted 3-6 membered carbocycle (e.g. an aromatic or a saturated carbocycle), or R2and R3are each independently H, OH, halogen, an unsubstituted C1-10alkyl, or an unsubstituted C1-10alkoxy;R4and R5are independently H, OH, halogen, a substituted or unsubstituted C1-10alkyl, a substituted or unsubstituted C1-10alkoxy, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, —COOH, —COOR10, —CH2COOH; —CH2COOR10, —CH2OH, —CH2OR10, —CONH2, —CH2NH2, —CH2NHR11, —CH2N11R12, —NCO, —CH2-halogen, —CHO, —CN, —NO2, —NH2, —NHR11, —NR11R12, R10, R11and R12are each independently a substituted or unsubstituted C1-10alkyl (e.g. an unsubstituted C1-10alkyl);m is an integer from 0 to 10, from 0 to 8, or from 0 to 6, such as 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; andn is an integer from 1 to 6, such as 1, 2, 3, 4, 5 or 6;or an enantiomer, tautomer, stereoisomers, solvate, zwitterion, polymorph, prodrug, or a pharmaceutically acceptable salt thereof. In some aspects of Formula Ia, Ib, Formula Ic, Formula I, Formula IIa, and/or Formula IIb, the compound contains one or more stereocenters on the carbon(s) attached to R4and/or R5. The compounds of any one of Formula Ib, Formula Ic, Formula I, Formula IIa, and Formula IIb may contain one or more chiral centers or may otherwise be capable of existing as multiple stereoisomers. These may be pure (single) stereoisomers or mixtures of stereoisomers, such as enantiomers, diastereomers, and enantiomerically or diastereomerically enriched mixtures. The compounds may be capable of existing as geometric isomers. Accordingly, it is to be understood that the compounds can be pure geometric isomers or mixtures of geometric isomers. In some aspects, for any one of Formula Ia, Ib, Ic, I, IIa, and IIb, the C1-10alkyl, C1-10alkoxy, C3-10cycloalkyl, aryl, and/or heteroaryl are unsubstituted C1-10alkyl, C1-10alkoxy, C3-10cycloalkyl, aryl, and/or heteroaryl. For any one of Formula Ia, Ib, Ic, I, IIa, and IIb, the C3-10cycloalkyl can be either monocyclic or polycyclic. For any one of Formula Ia, Ib, Ic, I, IIa, and IIb, the alkyl can be a linear substituted or unsubstituted alkyl or a branched substituted or unsubstituted alkyl, such as a linear unsubstituted alkyl or a branched unsubstituted alkyl. Exemplary alkyl include a linear substituted or unsubstituted C1-C10alkyl, a branched substituted or unsubstituted C4-C10alkyl, a linear substituted or unsubstituted C1-C6alkyl, a branched substituted or unsubstituted C4-C6alkyl, a linear substituted or unsubstituted C1-C4alkyl, such as a linear substituted or unsubstituted C1-C10, C1-C9, C1-C8, C1-C7, C1-C6, C1-C5, C1-C4, C1-C3, or C1-C2alkyl group or a branched substituted or unsubstituted C3-C9, C3-C9, C3-C8, C3-C7, C3-C6, C3-C5, or C3-C4alkyl group. For any of Formulae Ia, Ib, Ic, I, IIa, and IIb, the aryl group can be a C5-C30aryl, a C5-C20aryl, a C5-C12aryl, a C5-C11aryl, a C5-C9aryl, a C6-C20aryl, a C6-C12aryl, a C6-C11aryl, or a C6-C9aryl; and the heteroaryl can be a C5-C30heteroaryl, a C5-C20heteroaryl, a C5-C12heteroaryl, a C5-C11heteroaryl, a C5-C9heteroaryl, a C6-C30heteroaryl, a C6-C20heteroaryl, a C6-C12heteroaryl, a C6-C11heteroaryl, or a C6-C9heteroaryl. For any of Formulae Ia, Ib, Ic, I, IIa, and IIb, the aryl group can be a polyaryl group, such as a C10-C30polyaryl, a C10-C20polyaryl, a C10-C12polyaryl, a C10-C11polyaryl, or a C12-C20polyaryl. For any of Formulae Ia, Ib, Ic, I, IIa, and IIb, the heteroaryl group can be a polyheteroaryl, such as a C10-C30polyheteroaryl, a C10-C20polyheteroaryl, a C10-C12polyheteroaryl, a C10-C11polyheteroaryl, or a C12-C20polyheteroaryl. In some embodiments, the compound is selected from the group consisting of or a pharmaceutically acceptable salt thereof. When the compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb, and the exemplary compounds described above are in the form of pharmaceutically acceptable salts, the compounds can be the anion or the cation of the salt. In some aspects, when in the form of a pharmaceutically acceptable salt, the compound is the anion of the salt. Anionic forms of the compounds can be formed by dissociation of one or more functional group of the compound. For example, an anionic form of the compound is formed by dissociation of a hydroxyl and/or carboxylic acid group of the compound, such that a —O−and/or —COO−is formed. Anionic forms of the compounds can pair with any suitable cations to form the salt, such as ammonium, iminium, metal cations, etc. In some aspects, when in the form of a pharmaceutically acceptable salt, the compound is the cation of the salt. Cationic forms of the compounds can be formed by adding a proton on one or more functional group and/or atoms of the compound. For example, a cationic form of the compound is formed by adding a proton on an amino and/or imino group of the compound, such that an ammonium and/or iminium is formed. Cationic forms of the compounds may also be formed by removing one or more electrons from an atom (e.g. oxygen) of the compound. Cationic forms of the compounds can pair with any suitable anions to form the salt, such as halide ions, phosphate, sulfate, etc. Another embodiment provides a process for preparing a compound of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb. In some embodiments the compound is selected from the group consisting of or a pharmaceutically acceptable salt thereof. Generally, the compounds described herein can be synthesized by performing a coupling reaction between a first building block containing acyl halide or carboxylic acid and a second building block containing amines. Selecting suitable reaction conditions (e.g. reaction temperature, reaction time period, pressure, activating agent, solvent, etc.) for the coupling reaction is known. For example, synthesis of the compounds described herein includes conjugating equimolar quantities of a first building block containing carboxylic acid and a second building block containing amine using isobutyl chloroformate as an activating agent for the carboxylic acid of the first building block. The reaction mixture was then taken to dryness and the product was isolated using column chromatography. Protecting groups for either building blocks may be used as needed and when used, a deprotection step takes place after the coupling reaction has been completed. Selecting and adding a suitable protecting group for the building blocks and performing deprotection to remove the protecting group are known. An exemplary coupling reaction for synthesizing the compound of Formula Ia is shown below: wherein X, X′, P, P′, Q, Q′, G, G′, J, J′, E, and E′; R1-R3, R6-R9, and R13-R17; L1and L2; and Y are as defined above for Formula Ia; and Z is a halogen (e.g. Cl, Br, or I) or OH. An exemplary coupling reaction for synthesizing the compound of Formula Ib is shown below: wherein X, P, Q, G, J, and E; R1-R9, R13, R14, and R16; Y; m; and n are as defined above for Formula Ib; and Z is a halogen (e.g. Cl, Br, or I) or OH. An exemplary coupling reaction for synthesizing the compound of Formula Ic is shown below: wherein X; R1-R9, R13, R14, and R16; Y; m; and n are as defined above for Formula Ic; and Z is a halogen (e.g. Cl, Br, or I) or OH. An exemplary coupling reaction for synthesizing the compound of Formula I is shown below: wherein X; R1-R9; Y; m; and n are as defined above for Formula I; and Z is a halogen (e.g. Cl, Br, or I) or OH. An exemplary coupling reaction for synthesizing the compound of Formula Iia or Formula Iib is shown below: wherein X; R1-R5; m; and n are as defined above for Formula I; and Z is a halogen (e.g. Cl, Br, or I) or OH. III. Pharmaceutical Formulations The compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb and combinations thereof can be formulated into a pharmaceutical composition. In some embodiments, the compounds contained in the pharmaceutical composition are selected from the group consisting of or a pharmaceutically acceptable salt thereof. The disclosed pharmaceutical compositions can be for formulated for administration by parenteral (intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection), enteral, transdermal (either passively or using iontophoresis or electroporation), or transmucosal (nasal, pulmonary, vaginal, rectal, or sublingual) routes of administration or using bioerodible inserts and can be formulated in dosage forms appropriate for each route of administration. The compositions can be administered systemically. In one embodiment, the compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb can be formulated for immediate release, extended release, or modified release. A delayed release dosage form is one that releases a drug (or drugs) at a time other than promptly after administration. An extended release dosage form is one that allows at least a twofold reduction in dosing frequency as compared to that drug presented as a conventional dosage form (e.g., as a solution or prompt drug-releasing, conventional solid dosage form). A modified release dosage form is one for which the drug release characteristics of time course and/or location are chosen to accomplish therapeutic or convenience objectives not offered by conventional dosage forms such as solutions, ointments, or promptly dissolving dosage forms. Delayed release and extended release dosage forms and their combinations are types of modified release dosage forms. In some embodiments, the disclosed formulations are prepared using a pharmaceutically acceptable “carrier” composed of materials that are considered safe and effective and may be administered to an individual without causing undesirable biological side effects or unwanted interactions. The “carrier” is all components present in the pharmaceutical formulation other than the active ingredient or ingredients. The term “carrier” includes, but is not limited to, diluents, binders, lubricants, disintegrators, fillers, and coating compositions. “Carrier” also includes all components of the coating composition which may include plasticizers, pigments, colorants, stabilizing agents, and glidants. The delayed release dosage formulations may be prepared as described in references such as “Pharmaceutical dosage form tablets”, eds. Liberman et. Al. (New York, Marcel Dekker, Inc., 1989), “Remington—The science and practice of pharmacy”, 20thed., Lippincott Williams & Wilkins, Baltimore, MD, 2000, and “Pharmaceutical dosage forms and drug delivery systems”, 6thEdition, Ansel et. al., (Media, PA: Williams and Wilkins, 1995) which provides information on carriers, materials, equipment and process for preparing tablets and capsules and delayed release dosage forms of tablets, capsules, and granules. In one embodiment, the compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb can be administered to a subject with or without the aid of a delivery vehicle. Appropriate delivery vehicles for the compounds are known in the art and can be selected to suit the particular active agent. For example, in some embodiments, the active agent(s) is/are incorporated into or encapsulated by, or bound to, a nanoparticle, microparticle, micelle, synthetic lipoprotein particle, or carbon nanotube. For example, the compositions can be incorporated into a vehicle such as polymeric microparticles which provide controlled release of the compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb. In some embodiments, release of the compounds according to any one of Formulae Ia, Ib, Ic, I, IIa, and Iib is controlled by diffusion of the compounds out of the microparticles and/or degradation of the polymeric particles by hydrolysis and/or enzymatic degradation. Suitable polymers include ethylcellulose and other natural or synthetic cellulose derivatives. Polymers which are slowly soluble and form a gel in an aqueous environment, such as hydroxypropyl methylcellulose or polyethylene oxide, may also be suitable as materials for drug containing microparticles or particles. Other polymers include, but are not limited to, polyanhydrides, poly (ester anhydrides), polyhydroxy acids, such as polylactide (PLA), polyglycolide (PGA), poly(lactide-co-glycolide) (PLGA), poly-3-hydroxybut rate (PHB) and copolymers thereof, poly-4-hydroxybutyrate (P4HB) and copolymers thereof, polycaprolactone and copolymers thereof, and combinations thereof. In some embodiments, both agents are incorporated into the same particles and are formulated for release at different times and/or over different time periods. For example, in some embodiments, one of the agents is released entirely from the particles before release of the second agent begins. In other embodiments, release of the first agent begins followed by release of the second agent before all of the first agent is released. In still other embodiments, both agents are released at the same time over the same period of time or over different periods of time. A. Formulations for Parenteral Administration In one embodiment, compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb and pharmaceutical compositions thereof can be administered in an aqueous solution, by parenteral injection. The formulation may also be in the form of a suspension or emulsion. In general, pharmaceutical compositions are provided including effective amounts of the compound(s) and optionally include pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers. Such compositions include diluents sterile water, buffered saline of various buffer content (e.g., Tris-HCl, acetate, phosphate), pH and ionic strength; and optionally, additives such as detergents and solubilizing agents (e.g., TWEEN® 20, TWEEN® 80 also referred to as POLYSORBATE® 20 or 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), and preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol). Examples of non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate. The formulations may be lyophilized and redissolved/resuspended immediately before use. The formulation may be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. B. Oral Immediate Release Formulations Another embodiment provides suitable oral dosage forms containing of the compounds of Formula I that include but are not limited to tablets, capsules, solutions, suspensions, syrups, and lozenges. Tablets can be made using compression or molding techniques well known in the art. Gelatin or non-gelatin capsules can be prepared as hard or soft capsule shells, which can encapsulate liquid, solid, and semi-solid fill materials, using techniques well known in the art. Examples of suitable coating materials include, but are not limited to, cellulose polymers such as cellulose acetate phthalate, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate and hydroxypropyl methylcellulose acetate succinate; polyvinyl acetate phthalate, acrylic acid polymers and copolymers, and methacrylic resins that are commercially available under the trade name Eudragit® (Roth Pharma, Westerstadt, Germany), Zein, shellac, and polysaccharides. Additionally, the coating material may contain conventional carriers such as plasticizers, pigments, colorants, glidants, stabilization agents, pore formers and surfactants. Optional pharmaceutically acceptable excipients present in the drug-containing tablets, beads, granules or particles include, but are not limited to, diluents, binders, lubricants, disintegrants, colorants, stabilizers, and surfactants. Diluents, also termed “fillers,” are typically necessary to increase the bulk of a solid dosage form so that a practical size is provided for compression of tablets or formation of beads and granules. Suitable diluents include, but are not limited to, dicalcium phosphate dihydrate, calcium sulfate, lactose, sucrose, mannitol, sorbitol, cellulose, microcrystalline cellulose, kaolin, sodium chloride, dry starch, hydrolyzed starches, pregelatinized starch, silicone dioxide, titanium oxide, magnesium aluminum silicate and powder sugar. In some embodiments, binders are used to impart cohesive qualities to a solid dosage formulation, and thus ensure that a tablet or bead or granule remains intact after the formation of the dosage forms. Suitable binder materials include, but are not limited to, starch, pregelatinized starch, gelatin, sugars (including sucrose, glucose, dextrose, lactose and sorbitol), polyethylene glycol, waxes, natural and synthetic gums such as acacia, tragacanth, sodium alginate, cellulose, including hydorxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, and veegum, and synthetic polymers such as acrylic acid and methacrylic acid copolymers, methacrylic acid copolymers, methyl methacrylate copolymers, aminoalkyl methacrylate copolymers, polyacrylic acid/polymethacrylic acid and polyvinylpyrrolidone. In some embodiments, lubricants are used to facilitate tablet manufacture. Examples of suitable lubricants include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, glycerol behenate, polyethylene glycol, talc, and mineral oil. Disintegrants are used to facilitate dosage form disintegration or “breakup” after administration, and generally include, but are not limited to, starch, sodium starch glycolate, sodium carboxymethyl starch, sodium carboxymethylcellulose, hydroxypropyl cellulose, pregelatinized starch, clays, cellulose, alginine, gums or cross linked polymers, such as cross-linked PVP (Polyplasdone XL from GAF Chemical Corp). In some embodiments, stabilizers are used to inhibit or retard drug decomposition reactions which include, by way of example, oxidative reactions. Some embodiments include surfactants. The surfactants may be anionic, cationic, amphoteric or nonionic surface active agents. Suitable anionic surfactants include, but are not limited to, those containing carboxylate, sulfonate and sulfate ions. Examples of anionic surfactants include sodium, potassium, ammonium of long chain alkyl sulfonates and alkyl aryl sulfonates such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium bis-(2-ethylthioxyl)-sulfosuccinate; and alkyl sulfates such as sodium lauryl sulfate. Cationic surfactants include, but are not limited to, quaternary ammonium compounds such as benzalkonium chloride, benzethonium chloride, cetrimonium bromide, stearyl dimethylbenzyl ammonium chloride, polyoxyethylene and coconut amine. Examples of nonionic surfactants include ethylene glycol monostearate, propylene glycol myristate, glyceryl monostearate, glyceryl stearate, polyglyceryl-4-oleate, sorbitan acylate, sucrose acylate, PEG-150 laurate, PEG-400 monolaurate, polyoxyethylene monolaurate, polysorbates, polyoxyethylene octylphenylether, PEG-1000 cetyl ether, polyoxyethylene tridecyl ether, polypropylene glycol butyl ether, POLOXAMER® 401, stearoyl monoisopropanolamide, and polyoxyethylene hydrogenated tallow amide. Examples of amphoteric surfactants include sodium N-dodecyl-.beta.-alanine, sodium N-lauryl-.beta.-iminodipropionate, myristoamphoacetate, lauryl betaine and lauryl sulfobetaine. If desired, the tablets, beads granules or particles may also contain minor amount of nontoxic auxiliary substances such as wetting or emulsifying agents, dyes, pH buffering agents, and preservatives. C. Extended Release Dosage Forms One embodiment provides extended release formulations of compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb that are generally prepared as diffusion or osmotic systems, for example, as described in “Remington—The science and practice of pharmacy” (20thed., Lippincott Williams & Wilkins, Baltimore, MD, 2000). A diffusion system typically consists of two types of devices, reservoir and matrix, and is well known and described in the art. The matrix devices are generally prepared by compressing the drug with a slowly dissolving polymer carrier into a tablet form. The three major types of materials used in the preparation of matrix devices are insoluble plastics, hydrophilic polymers, and fatty compounds. Plastic matrices include, but not limited to, methyl acrylate-methyl methacrylate, polyvinyl chloride, and polyethylene. Hydrophilic polymers include, but are not limited to, methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and carbopol 934, polyethylene oxides. Fatty compounds include, but are not limited to, various waxes such as carnauba wax and glyceryl tristearate. Alternatively, extended release formulations of the compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb can be prepared using osmotic systems or by applying a semi-permeable coating to the dosage form. In the latter case, the desired drug release profile can be achieved by combining low permeable and high permeable coating materials in suitable proportion. The devices with different drug release mechanisms described above could be combined in a final dosage form comprising single or multiple units. Examples of multiple units include multilayer tablets, capsules containing tablets, beads, granules, etc. An immediate release portion can be added to the extended release system by means of either applying an immediate release layer on top of the extended release core using coating or compression process or in a multiple unit system such as a capsule containing extended and immediate release beads. Extended release tablets containing hydrophilic polymers are prepared by techniques commonly known in the art such as direct compression, wet granulation, or dry granulation processes. Their formulations usually incorporate polymers, diluents, binders, and lubricants as well as the active pharmaceutical ingredient. The usual diluents include inert powdered substances such as any of many different kinds of starch, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders. Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts such as sodium chloride and powdered sugar. Powdered cellulose derivatives are also useful. Typical tablet binders include substances such as starch, gelatin and sugars such as lactose, fructose, and glucose. Natural and synthetic gums, including acacia, alginates, methylcellulose, and polyvinylpyrrolidine can also be used. Polyethylene glycol, hydrophilic polymers, ethylcellulose and waxes can also serve as binders. A lubricant is necessary in a tablet formulation to prevent the tablet and punches from sticking in the die. The lubricant is chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils. Extended release tablets containing wax materials are generally prepared using methods known in the art such as a direct blend method, a congealing method, and an aqueous dispersion method. In a congealing method, the drug is mixed with a wax material and either spray-congealed or congealed and screened and processed. D. Delayed Release Dosage Forms In some embodiments delayed release formulations of compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb are created by coating a solid dosage form with a film of a polymer which is insoluble in the acid environment of the stomach, and soluble in the neutral environment of small intestines. The delayed release dosage units can be prepared, for example, by coating a drug or a drug-containing composition with a selected coating material. The drug-containing composition may be, e.g., a tablet for incorporation into a capsule, a tablet for use as an inner core in a “coated core” dosage form, or a plurality of drug-containing beads, particles or granules, for incorporation into either a tablet or capsule. Preferred coating materials include bioerodible, gradually hydrolyzable, gradually water-soluble, and/or enzymatically degradable polymers, and may be conventional “enteric” polymers. Enteric polymers, as will be appreciated by those skilled in the art, become soluble in the higher pH environment of the lower gastrointestinal tract or slowly erode as the dosage form passes through the gastrointestinal tract, while enzymatically degradable polymers are degraded by bacterial enzymes present in the lower gastrointestinal tract, particularly in the colon. Suitable coating materials for effecting delayed release include, but are not limited to, cellulosic polymers such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxymethyl cellulose, hydroxypropyl methyl cellulose, hydroxypropyl methyl cellulose acetate succinate, hydroxypropylmethyl cellulose phthalate, methylcellulose, ethyl cellulose, cellulose acetate, cellulose acetate phthalate, cellulose acetate trimellitate and carboxymethylcellulose sodium; acrylic acid polymers and copolymers, preferably formed from acrylic acid, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate, and other methacrylic resins that are commercially available under the tradename EUDRAGIT®. (Rohm Pharma; Westerstadt, Germany), including EUDRAGIT®. L30D-55 and L100-55 (soluble at pH 5.5 and above), EUDRAGIT®. L-100 (soluble at pH 6.0 and above), EUDRAGIT®. S (soluble at pH 7.0 and above, as a result of a higher degree of esterification), and EUDRAGITS®. NE, RL and RS (water-insoluble polymers having different degrees of permeability and expandability); vinyl polymers and copolymers such as polyvinyl pyrrolidone, vinyl acetate, vinylacetate phthalate, vinylacetate crotonic acid copolymer, and ethylene-vinyl acetate copolymer; enzymatically degradable polymers such as azo polymers, pectin, chitosan, amylose and guar gum; zein and shellac. Combinations of different coating materials may also be used. Multi-layer coatings using different polymers may also be applied. The preferred coating weights for particular coating materials may be readily determined by those skilled in the art by evaluating individual release profiles for tablets, beads and granules prepared with different quantities of various coating materials. It is the combination of materials, method and form of application that produce the desired release characteristics, which one can determine only from the clinical studies. The coating composition may include conventional additives, such as plasticizers, pigments, colorants, stabilizing agents, glidants, etc. A plasticizer is normally present to reduce the fragility of the coating, and will generally represent about 10 wt. % to 50 wt. % relative to the dry weight of the polymer. Examples of typical plasticizers include polyethylene glycol, propylene glycol, triacetin, dimethyl phthalate, diethyl phthalate, dibutyl phthalate, dibutyl sebacate, triethyl citrate, tributyl citrate, triethyl acetyl citrate, castor oil and acetylated monoglycerides. A stabilizing agent is preferably used to stabilize particles in the dispersion. Typical stabilizing agents are nonionic emulsifiers such as sorbitan esters, polysorbates and polyvinylpyrrolidone. Glidants are recommended to reduce sticking effects during film formation and drying, and will generally represent approximately 25 wt. % to 100 wt. % of the polymer weight in the coating solution. One effective glidant is talc. Other glidants such as magnesium stearate and glycerol monostearates may also be used. Pigments such as titanium dioxide may also be used. Small quantities of an anti-foaming agent, such as a silicone (e.g., simethicone), may also be added to the coating composition. E. Formulations for Mucosal and Pulmonary Administration The compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb and compositions thereof can be formulated for pulmonary or mucosal administration. The administration can include delivery of the composition to the lungs, nasal, oral (sublingual, buccal), vaginal, or rectal mucosa. In a particular embodiment, the composition is formulated for and delivered to the subject sublingually. In one embodiment, the compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb are formulated for pulmonary delivery, such as intranasal administration or oral inhalation. The respiratory tract is the structure involved in the exchange of gases between the atmosphere and the blood stream. The lungs are branching structures ultimately ending with the alveoli where the exchange of gases occurs. The alveolar surface area is the largest in the respiratory system and is where drug absorption occurs. The alveoli are covered by a thin epithelium without cilia or a mucus blanket and secrete surfactant phospholipids. The respiratory tract encompasses the upper airways, including the oropharynx and larynx, followed by the lower airways, which include the trachea followed by bifurcations into the bronchi and bronchioli. The upper and lower airways are called the conducting airways. The terminal bronchioli then divide into respiratory bronchiole, which then lead to the ultimate respiratory zone, the alveoli, or deep lung. The deep lung, or alveoli, is the primary target of inhaled therapeutic aerosols for systemic drug delivery. One embodiment provides for nasal delivery for administration of the compounds of formula I. The compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb can be formulated as an aerosol. The term aerosol refers to any preparation of a fine mist of particles, which can be in solution or a suspension, whether or not it is produced using a propellant. Aerosols can be produced using standard techniques, such as ultrasonication or high-pressure treatment. Carriers for pulmonary formulations can be divided into those for dry powder formulations and for administration as solutions. Aerosols for the delivery of therapeutic agents to the respiratory tract are known in the art. For administration via the upper respiratory tract, the formulation can be formulated into a solution, e.g., water or isotonic saline, buffered or un-buffered, or as a suspension, for intranasal administration as drops or as a spray. Preferably, such solutions or suspensions are isotonic relative to nasal secretions and of about the same pH, ranging e.g., from about pH 4.0 to about pH 7.4 or, from pH 6.0 to pH 7.0. Buffers should be physiologically compatible and include, simply by way of example, phosphate buffers. For example, a representative nasal decongestant is described as being buffered to a pH of about 6.2. One skilled in the art can readily determine a suitable saline content and pH for an innocuous aqueous solution for nasal and/or upper respiratory administration. Preferably, the aqueous solution is water, physiologically acceptable aqueous solutions containing salts and/or buffers, such as phosphate buffered saline (PBS), or any other aqueous solution acceptable for administration to an animal or human. Such solutions are well known to a person skilled in the art and include, but are not limited to, distilled water, de-ionized water, pure or ultrapure water, saline, phosphate-buffered saline (PBS). Other suitable aqueous vehicles include, but are not limited to, Ringer's solution and isotonic sodium chloride. Aqueous suspensions may include suspending agents such as cellulose derivatives, sodium alginate, polyvinyl-pyrrolidone and gum tragacanth, and a wetting agent such as lecithin. Suitable preservatives for aqueous suspensions include ethyl and n-propyl p-hydroxybenzoate. In another embodiment, solvents that are low toxicity organic (i.e. nonaqueous) class 3 residual solvents, such as ethanol, acetone, ethyl acetate, tetrahydrofuran, ethyl ether, and propanol may be used for the formulations. The solvent is selected based on its ability to readily aerosolize the formulation. The solvent should not detrimentally react with the compounds. An appropriate solvent should be used that dissolves the compounds or forms a suspension of the compounds. The solvent should be sufficiently volatile to enable formation of an aerosol of the solution or suspension. Additional solvents or aerosolizing agents, such as freons, can be added as desired to increase the volatility of the solution or suspension. In one embodiment, compositions may contain minor amounts of polymers, surfactants, or other excipients well known to those of the art. In this context, “minor amounts” means no excipients are present that might affect or mediate uptake of the compounds in the lungs and that the excipients that are present are present in amount that do not adversely affect uptake of compounds in the lungs. Dry lipid powders can be directly dispersed in ethanol because of their hydrophobic character. For lipids stored in organic solvents such as chloroform, the desired quantity of solution is placed in a vial, and the chloroform is evaporated under a stream of nitrogen to form a dry thin film on the surface of a glass vial. The film swells easily when reconstituted with ethanol. To fully disperse the lipid molecules in the organic solvent, the suspension is sonicated. Nonaqueous suspensions of lipids can also be prepared in absolute ethanol using a reusable PARI LC Jet+ nebulizer (PARI Respiratory Equipment, Monterey, CA). Dry powder formulations (“DPFs”) with large particle size have improved flowability characteristics, such as less aggregation, easier aerosolization, and potentially less phagocytosis. Dry powder aerosols for inhalation therapy are generally produced with mean diameters primarily in the range of less than 5 microns, although a preferred range is between one and ten microns in aerodynamic diameter. Large “carrier” particles (containing no drug) have been co-delivered with therapeutic aerosols to aid in achieving efficient aerosolization among other possible benefits. Polymeric particles may be prepared using single and double emulsion solvent evaporation, spray drying, solvent extraction, solvent evaporation, phase separation, simple and complex coacervation, interfacial polymerization, and other methods well known to those of ordinary skill in the art. Particles may be made using methods for making microspheres or microcapsules known in the art. The preferred methods of manufacture are by spray drying and freeze drying, which entails using a solution containing the surfactant, spraying to form droplets of the desired size, and removing the solvent. The particles may be fabricated with the appropriate material, surface roughness, diameter and tap density for localized delivery to selected regions of the respiratory tract such as the deep lung or upper airways. For example, higher density or larger particles may be used for upper airway delivery. Similarly, a mixture of different sized particles, provided with the same or different active agents may be administered to target different regions of the lung in one administration. F. Topical and Transdermal Formulations Transdermal formulations containing the compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb may also be prepared. These will typically be gels, ointments, lotions, sprays, or patches, all of which can be prepared using standard technology. Transdermal formulations can include penetration enhancers. An “oil” is a composition containing at least 95% wt of a lipophilic substance. Examples of lipophilic substances include but are not limited to naturally occurring and synthetic oils, fats, fatty acids, lecithins, triglycerides and combinations thereof. A “continuous phase” refers to the liquid in which solids are suspended or droplets of another liquid are dispersed, and is sometimes called the external phase. This also refers to the fluid phase of a colloid within which solid or fluid particles are distributed. If the continuous phase is water (or another hydrophilic solvent), water-soluble or hydrophilic drugs will dissolve in the continuous phase (as opposed to being dispersed). In a multiphase formulation (e.g., an emulsion), the discreet phase is suspended or dispersed in the continuous phase. An “emulsion” is a composition containing a mixture of non-miscible components homogenously blended together. In particular embodiments, the non-miscible components include a lipophilic component and an aqueous component. An emulsion is a preparation of one liquid distributed in small globules throughout the body of a second liquid. The dispersed liquid is the discontinuous phase, and the dispersion medium is the continuous phase. When oil is the dispersed liquid and an aqueous solution is the continuous phase, it is known as an oil-in-water emulsion, whereas when water or aqueous solution is the dispersed phase and oil or oleaginous substance is the continuous phase, it is known as a water-in-oil emulsion. Either or both of the oil phase and the aqueous phase may contain one or more surfactants, emulsifiers, emulsion stabilizers, buffers, and other excipients. Preferred excipients include surfactants, especially non-ionic surfactants; emulsifying agents, especially emulsifying waxes; and liquid non-volatile non-aqueous materials, particularly glycols such as propylene glycol. The oil phase may contain other oily pharmaceutically approved excipients. For example, materials such as hydroxylated castor oil or sesame oil may be used in the oil phase as surfactants or emulsifiers. “Emollients” are an externally applied agent that softens or soothes skin and are generally known in the art and listed in compendia, such as the “Handbook of Pharmaceutical Excipients”, 4thEd., Pharmaceutical Press, 2003. These include, without limitation, almond oil, castor oil, ceratonia extract, cetostearoyl alcohol, cetyl alcohol, cetyl esters wax, cholesterol, cottonseed oil, cyclomethicone, ethylene glycol palmitostearate, glycerin, glycerin monostearate, glyceryl monooleate, isopropyl myristate, isopropyl palmitate, lanolin, lecithin, light mineral oil, medium-chain triglycerides, mineral oil and lanolin alcohols, petrolatum, petrolatum and lanolin alcohols, soybean oil, starch, stearyl alcohol, sunflower oil, xylitol and combinations thereof. In one embodiment, the emollients are ethylhexylstearate and ethylhexyl palmitate. “Surfactants” are surface-active agents that lower surface tension and thereby increase the emulsifying, foaming, dispersing, spreading and wetting properties of a product. Suitable non-ionic surfactants include emulsifying wax, glyceryl monooleate, polyoxyethylene alkyl ethers, polyoxyethylene castor oil derivatives, polysorbate, sorbitan esters, benzyl alcohol, benzyl benzoate, cyclodextrins, glycerin monostearate, poloxamer, povidone and combinations thereof. In one embodiment, the non-ionic surfactant is stearyl alcohol. “Emulsifiers” are surface active substances which promote the suspension of one liquid in another and promote the formation of a stable mixture, or emulsion, of oil and water. Common emulsifiers are: metallic soaps, certain animal and vegetable oils, and various polar compounds. Suitable emulsifiers include acacia, anionic emulsifying wax, calcium stearate, carbomers, cetostearyl alcohol, cetyl alcohol, cholesterol, diethanolamine, ethylene glycol palmitostearate, glycerin monostearate, glyceryl monooleate, hydroxpropyl cellulose, hypromellose, lanolin, hydrous, lanolin alcohols, lecithin, medium-chain triglycerides, methylcellulose, mineral oil and lanolin alcohols, monobasic sodium phosphate, monoethanolamine, nonionic emulsifying wax, oleic acid, poloxamer, poloxamers, polyoxyethylene alkyl ethers, polyoxyethylene castor oil derivatives, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene stearates, propylene glycol alginate, self-emulsifying glyceryl monostearate, sodium citrate dehydrate, sodium lauryl sulfate, sorbitan esters, stearic acid, sunflower oil, tragacanth, triethanolamine, xanthan gum and combinations thereof. In one embodiment, the emulsifier is glycerol stearate. A “lotion” is a low- to medium-viscosity liquid formulation. A lotion can contain finely powdered substances that are in soluble in the dispersion medium through the use of suspending agents and dispersing agents. Alternatively, lotions can have as the dispersed phase liquid substances that are immiscible with the vehicle and are usually dispersed by means of emulsifying agents or other suitable stabilizers. In one embodiment, the lotion is in the form of an emulsion having a viscosity of between 100 and 1000 centistokes. The fluidity of lotions permits rapid and uniform application over a wide surface area. Lotions are typically intended to dry on the skin leaving a thin coat of their medicinal components on the skin's surface. A “cream” is a viscous liquid or semi-solid emulsion of either the “oil-in-water” or “water-in-oil type”. Creams may contain emulsifying agents and/or other stabilizing agents. In one embodiment, the formulation is in the form of a cream having a viscosity of greater than 1000 centistokes, typically in the range of 20,000-50,000 centistokes. Creams are often time preferred over ointments as they are generally easier to spread and easier to remove. An emulsion is a preparation of one liquid distributed in small globules throughout the body of a second liquid. The dispersed liquid is the discontinuous phase, and the dispersion medium is the continuous phase. When oil is the dispersed liquid and an aqueous solution is the continuous phase, it is known as an oil-in-water emulsion, whereas when water or aqueous solution is the dispersed phase and oil or oleaginous substance is the continuous phase, it is known as a water-in-oil emulsion. The oil phase may consist at least in part of a propellant, such as an HFA propellant. Either or both of the oil phase and the aqueous phase may contain one or more surfactants, emulsifiers, emulsion stabilizers, buffers, and other excipients. Preferred excipients include surfactants, especially non-ionic surfactants; emulsifying agents, especially emulsifying waxes; and liquid non-volatile non-aqueous materials, particularly glycols such as propylene glycol. The oil phase may contain other oily pharmaceutically approved excipients. For example, materials such as hydroxylated castor oil or sesame oil may be used in the oil phase as surfactants or emulsifiers. A sub-set of emulsions are the self-emulsifying systems. These drug delivery systems are typically capsules (hard shell or soft shell) comprised of the drug dispersed or dissolved in a mixture of surfactant(s) and lipophillic liquids such as oils or other water immiscible liquids. When the capsule is exposed to an aqueous environment and the outer gelatin shell dissolves, contact between the aqueous medium and the capsule contents instantly generates very small emulsion droplets. These typically are in the size range of micelles or nanoparticles. No mixing force is required to generate the emulsion as is typically the case in emulsion formulation processes. The basic difference between a cream and a lotion is the viscosity, which is dependent on the amount/use of various oils and the percentage of water used to prepare the formulations. Creams are typically thicker than lotions, may have various uses and often one uses more varied oils/butters, depending upon the desired effect upon the skin. In a cream formulation, the water-base percentage is about 60-75% and the oil-base is about 20-30% of the total, with the other percentages being the emulsifier agent, preservatives and additives for a total of 100%. An “ointment” is a semisolid preparation containing an ointment base and optionally one or more active agents. Examples of suitable ointment bases include hydrocarbon bases (e.g., petrolatum, white petrolatum, yellow ointment, and mineral oil); absorption bases (hydrophilic petrolatum, anhydrous lanolin, lanolin, and cold cream); water-removable bases (e.g., hydrophilic ointment), and water-soluble bases (e.g., polyethylene glycol ointments). Pastes typically differ from ointments in that they contain a larger percentage of solids. Pastes are typically more absorptive and less greasy that ointments prepared with the same components. A “gel” is a semisolid system containing dispersions of small or large molecules in a liquid vehicle that is rendered semisolid by the action of a thickening agent or polymeric material dissolved or suspended in the liquid vehicle. The liquid may include a lipophilic component, an aqueous component or both. Some emulsions may be gels or otherwise include a gel component. Some gels, however, are not emulsions because they do not contain a homogenized blend of immiscible components. Suitable gelling agents include, but are not limited to, modified celluloses, such as hydroxypropyl cellulose and hydroxyethyl cellulose; Carbopol homopolymers and copolymers; and combinations thereof. Suitable solvents in the liquid vehicle include, but are not limited to, diglycol monoethyl ether; alklene glycols, such as propylene glycol; dimethyl isosorbide; alcohols, such as isopropyl alcohol and ethanol. The solvents are typically selected for their ability to dissolve the drug. Other additives, which improve the skin feel and/or emolliency of the formulation, may also be incorporated. Examples of such additives include, but are not limited, isopropyl myristate, ethyl acetate, C12-C15 alkyl benzoates, mineral oil, squalane, cyclomethicone, capric/caprylic triglycerides, and combinations thereof. Foams consist of an emulsion in combination with a gaseous propellant. The gaseous propellant consists primarily of hydrofluoroalkanes (HFAs). Suitable propellants include HFAs such as 1,1,1,2-tetrafluoroethane (HFA 134a) and 1,1,1,2,3,3,3-heptafluoropropane (HFA 227), but mixtures and admixtures of these and other HFAs that are currently approved or may become approved for medical use are suitable. The propellants preferably are not hydrocarbon propellant gases which can produce flammable or explosive vapors during spraying. Furthermore, the compositions preferably contain no volatile alcohols, which can produce flammable or explosive vapors during use. Buffers are used to control pH of a composition. Preferably, the buffers buffer the composition from a pH of about 4 to a pH of about 7.5, more preferably from a pH of about 4 to a pH of about 7, and most preferably from a pH of about 5 to a pH of about 7. In a preferred embodiment, the buffer is triethanolamine. Preservatives can be used to prevent the growth of fungi and microorganisms. Suitable antifungal and antimicrobial agents include, but are not limited to, benzoic acid, butylparaben, ethyl paraben, methyl paraben, propylparaben, sodium benzoate, sodium propionate, benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, and thimerosal. Additional agents that can be added to the formulation include penetration enhancers. In some embodiments, the penetration enhancer increases the solubility of the drug, improves transdermal delivery of the drug across the skin, in particular across the stratum corneum, or a combination thereof. Some penetration enhancers cause dermal irritation, dermal toxicity and dermal allergies. However, the more commonly used ones include urea, (carbonyldiamide), imidurea, N, N-diethylformamide, N-methyl-2-pyrrolidone, 1-dodecal-azacyclopheptane-2-one, calcium thioglycate, 2-pyrrolidone, N,N-diethyl-m-toluamide, oleic acid and its ester derivatives, such as methyl, ethyl, propyl, isopropyl, butyl, vinyl and glycerylmonooleate, sorbitan esters, such as sorbitan monolaurate and sorbitan monooleate, other fatty acid esters such as isopropyl laurate, isopropyl myristate, isopropyl palmitate, diisopropyl adipate, propylene glycol monolaurate, propylene glycol monooleatea and non-ionic detergents such as BRIJ® 76 (stearyl poly(10 oxyethylene ether), BRIJ® 78 (stearyl poly(20)oxyethylene ether), BRIJ® 96 (oleyl poly(10)oxyethylene ether), and BRIJ® 721 (stearyl poly (21) oxyethylene ether) (ICI Americas Inc. Corp.). Chemical penetrations and methods of increasing transdermal drug delivery are described in Inayat, et al.,Tropical Journal of Pharmaceutical Research,8 (2):173-179 (2009) and Fox, et al.,Molecules,16:10507-10540 (2011). In some embodiments, the penetration enhancer is, or includes, an alcohol such ethanol, or others disclosed herein or known in the art. Delivery of drugs by the transdermal route has been known for many years. Advantages of a transdermal drug delivery compared to other types of medication delivery such as oral, intravenous, intramuscular, etc., include avoidance of hepatic first pass metabolism, ability to discontinue administration by removal of the system, the ability to control drug delivery for a longer time than the usual gastrointestinal transit of oral dosage form, and the ability to modify the properties of the biological barrier to absorption. Controlled release transdermal devices rely for their effect on delivery of a known flux of drug to the skin for a prolonged period of time, generally a day, several days, or a week. Two mechanisms are used to regulate the drug flux: either the drug is contained within a drug reservoir, which is separated from the skin of the wearer by a synthetic membrane, through which the drug diffuses; or the drug is held dissolved or suspended in a polymer matrix, through which the drug diffuses to the skin. Devices incorporating a reservoir will deliver a steady drug flux across the membrane as long as excess undissolved drug remains in the reservoir; matrix or monolithic devices are typically characterized by a falling drug flux with time, as the matrix layers closer to the skin are depleted of drug. Usually, reservoir patches include a porous membrane covering the reservoir of medication which can control release, while heat melting thin layers of medication embedded in the polymer matrix (e.g., the adhesive layer), can control release of drug from matrix or monolithic devices. Accordingly, the active agent can be released from a patch in a controlled fashion without necessarily being in a controlled release formulation. Patches can include a liner which protects the patch during storage and is removed prior to use; drug or drug solution in direct contact with release liner; adhesive which serves to adhere the components of the patch together along with adhering the patch to the skin; one or more membranes, which can separate other layers, control the release of the drug from the reservoir and multi-layer patches, etc., and backing which protects the patch from the outer environment. Common types of transdermal patches include, but are not limited to, single-layer drug-in-adhesive patches, wherein the adhesive layer contains the drug and serves to adhere the various layers of the patch together, along with the entire system to the skin, but is also responsible for the releasing of the drug; multi-layer drug-in-adhesive, wherein which is similar to a single-layer drug-in-adhesive patch, but contains multiple layers, for example, a layer for immediate release of the drug and another layer for control release of drug from the reservoir; reservoir patches wherein the drug layer is a liquid compartment containing a drug solution or suspension separated by the adhesive layer; matrix patches, wherein a drug layer of a semisolid matrix containing a drug solution or suspension which is surrounded and partially overlaid by the adhesive layer; and vapor patches, wherein an adhesive layer not only serves to adhere the various layers together but also to release vapor. Methods for making transdermal patches are described in U.S. Pat. Nos. 6,461,644, 6,676,961, 5,985,311, and 5,948,433. In some embodiments, the composition is formulated for transdermal delivery and administered using a transdermal patch. In some embodiments, the formulation, the patch, or both are designed for extended release of the curcumin conjugate. Exemplary symptoms, pharmacologic, and physiologic effects are discussed in more detail below. G. Methods of Manufacture As will be appreciated by those skilled in the art and as described in the pertinent texts and literature, a number of methods are available for preparing formulations containing the compounds of Formula I including but not limited to tablets, beads, granules, microparticle, or nanparticles that provide a variety of drug release profiles. Such methods include, but are not limited to, the following: coating a drug or drug-containing composition with an appropriate coating material, typically although not necessarily incorporating a polymeric material, increasing drug particle size, placing the drug within a matrix, and forming complexes of the drug with a suitable complexing agent. The delayed release dosage units may be coated with the delayed release polymer coating using conventional techniques, e.g., using a conventional coating pan, an airless spray technique, fluidized bed coating equipment (with or without a Wurster insert). For detailed information concerning materials, equipment and processes for preparing tablets and delayed release dosage forms, see Pharmaceutical Dosage Forms: Tablets, eds. Lieberman et al. (New York: Marcel Dekker, Inc., 1989), and Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 6.sup.th Ed. (Media, PA: Williams & Wilkins, 1995). An exemplary method for preparing extended release tablets includes compressing a drug-containing blend, e.g., blend of drug-containing granules, prepared using a direct blend, wet-granulation, or dry-granulation process. Extended release tablets may also be molded rather than compressed, starting with a moist material containing a suitable water-soluble lubricant. However, tablets are preferably manufactured using compression rather than molding. A preferred method for forming extended release drug-containing blend is to mix drug particles directly with one or more excipients such as diluents (or fillers), binders, disintegrants, lubricants, glidants, and colorants. As an alternative to direct blending, a drug-containing blend may be prepared by using wet-granulation or dry-granulation processes. Beads containing the active agent may also be prepared by any one of a number of conventional techniques, typically starting from a fluid dispersion. For example, a typical method for preparing drug-containing beads involves dispersing or dissolving the active agent in a coating suspension or solution containing pharmaceutical excipients such as polyvinylpyrrolidone, methylcellulose, talc, metallic stearates, silicone dioxide, plasticizers or the like. The admixture is used to coat a bead core such as a sugar sphere (or so-called “non-pareil”) having a size of approximately 60 to 20 mesh. An alternative procedure for preparing drug beads is by blending drug with one or more pharmaceutically acceptable excipients, such as microcrystalline cellulose, lactose, cellulose, polyvinyl pyrrolidone, talc, magnesium stearate, a disintegrant, etc., extruding the blend, spheronizing the extrudate, drying and optionally coating to form the immediate release beads. IV. Methods of Use The compounds of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or N-(2-benzo[1,3]dioxol-5-yl-ethyl)-2-(4-methyl-benzyl)-succinamic acid and pharmaceutical compositions thereof are useful for the treatment of an IDO1-related disease, disorder or condition in a subject in need thereof. Generally, the method includes administering to the subject a therapeutically effective amount of the compound disclosed herein or N-(2-benzo[1,3]dioxol-5-yl-ethyl)-2-(4-methyl-benzyl)-succinamic acid or a pharmaceutical composition thereof to treat the IDO1-related disease, disorder or condition. The subject can be a mammal, such as humans, dogs, cats, mice, rats, monkeys, rabbits, guinea pigs, or agricultural animals such as cattle, sheep, pigs, etc. In some embodiments, the IDO1-related disease, disorder or condition treated using the methods disclosed herein is age related muscle loss or sarcopenia, wherein the compound or pharmaceutical composition thereof is administered in an effective amount to treat or prevent the age-related muscle loss or sarcopenia. In some embodiments, the IDO1-related disease, disorder or condition treated using the method disclosed herein is muscle loss related to systemic inflammation (e.g. cancer and/or HIV-induced muscle wasting), wherein the compound or pharmaceutical composition thereof is administered in an effective amount to treat or prevent the muscle loss related to systemic inflammation. For example, the IDO1-related disease, disorder or condition treated using the method disclosed herein is cancer-induced muscle wasting (cachexia) wherein the compound or pharmaceutical composition thereof is administered in an effective amount to treat or prevent the muscle wasting. For example, the IDO1-related disease, disorder or condition treated using the method disclosed herein is HIV-induced muscle wasting (including HIV before/under/after treatment with antiretroviral therapy), wherein the compound or pharmaceutical composition thereof is administered in an effective amount to treat or prevent the muscle wasting. In some embodiments, a compound is administered in an effective amount to treat an age-related disorder using the method disclosed herein. One disorder of particular importance is frailty. Frailty is a syndrome that can be characterized by loss of reserve, feebleness, vulnerability, and failure of homeostasis (Chan,The Hong Kong Medical Diary,13 (9):7-9 (2008)). The loss of reserve and resilience is part of a feed-forward loop inviting associated comorbidities leading to further decreasing reserve. It is believed that declines in the molecular, cellular and physiological systems of the aging body are the underlying mechanisms associated with the reduction in the effectiveness of muscle and bone as well as declines in the circulatory, hormonal, and immune systems that are typical of frail individuals (Fried, et al.,Journal of Gerontology: Medical Sciences,56A (3):M146-156 (2001), and Chan,The Hong Kong Medical Diary,13 (9):7-9 (2008)). Frailty can be the consequence of one or more additional underlying diseases, for example cachexia, immobilization, aging, chronic disease, or cancer. Although frailty, like many other age-related diseases, is often associated with chronological age, not all elderly individuals are frail and not all frail individuals are elderly. Frail individuals are typically at an increased risk of disability and death from minor internal stresses such as anxiety and depression, or external stresses such physical strain, infections, heat, and cold. For example, individuals suffering from frailty can exhibit one or more symptoms including sarcopenia, unintentional non-muscle weight loss greater than 10 lbs per week, decreased grip strength, low energy expenditure, weakness, fatigue, and decreased walking time. These factors can contribute to a progressive increase in disability, dependency, the need for long term care, and mortality in frail individuals over time (Chan,The Hong Kong Medical Diary,13 (9):7-9 (2008)). In some embodiments, the subject suffers from a disease or condition such as muscle atrophy, muscular dystrophy, sarcopenia, frailty, or combinations thereof. The disclosed compositions and methods can be used to treat or prevent muscle atrophy, muscular dystrophy, sarcopenia, frailty, combinations thereof, or one or more symptoms or comorbidities thereof, wherein the compound is administered in an effective amount to treat or prevent one or more of these diseases or conditions. The muscle atrophy or sarcopenia can result from cachexia, immobilization, aging, chronic disease, cancer, or combinations thereof. The disclosed compositions and methods can also be used to treat or prevent muscle loss related to systemic inflammation, such as muscle wasting (cachexia) induced by cancer and HIV (including HIV being treated with antiretroviral therapy). Another embodiment provides methods for inhibiting IDO1 in a subject in need thereof by contacting the subject's cells expressing IDO1 with an effective amount of a compound of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or N-(2-benzo[1,3]dioxol-5-yl-ethyl)-2-(4-methyl-benzyl)-succinamic acid or a pharmaceutical composition thereof to inhibit IDO1 in the subject. In some aspects, the methods for inhibiting IDO1 in a subject in need thereof includes contacting the subject's cells expressing IDO1 with an effective amount of a compound of any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or a pharmaceutical composition thereof to inhibit IDO1 in the subject. In some aspects, the methods for inhibiting IDO1 in a subject in need thereof includes contacting the subject's cells expressing IDO1 with an effective amount of N-(2-benzo[1,3]dioxol-5-yl-ethyl)-2-(4-methyl-benzyl)-succinamic acid or a pharmaceutical composition thereof to inhibit IDO1 in the subject. In one embodiment, the compound used in the method for inhibiting IDO1 in the subject in need thereof is selected from the group consisting of or a pharmaceutically acceptable salt thereof. Another embodiment provides a method for treating a sarcopenia in a subject in need thereof by administering to the subject a therapeutically effective amount of the compound according to any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or a pharmaceutical composition thereof to treat sarcopenia. Sarcopenia typically refers to the loss of skeletal muscle mass associated with advancing age (Cruz-Jentoft, A. et al.,Age and Aging,39:412-423 (2010); Lang, T. et al.,Osteoporosis Int,21:543-559 (2010)). Loss of skeletal muscle mass can also be unrelated to age. For example, loss of skeletal muscle mass occurs in subjects with cachexia. In some embodiments the compound is selected from the group consist of still another embodiment provides a method for inhibiting or reducing the production of kynurenine in a subject in need thereof comprising administering to the subject an effective amount of a compound according to any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or a pharmaceutically acceptable salt or pharmaceutical composition thereof to inhibit or reduce the production of kynurenine in the subject. In some aspects, using the method disclosed herein, an effective amount of the compounds according to any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or a pharmaceutically acceptable salt or pharmaceutical composition thereof is administered to lower kynurenine or other downstream tryptophan metabolite levels. In some embodiments the compound used in the method for inhibiting or reducing the production of kynurenine in the subject in need thereof is selected from the group consisting of or a pharmaceutically acceptable salt thereof. Another embodiment provides a method for inhibiting or reducing the production of kynurenine in a subject in need thereof comprising administering to the subject an effective amount of a compound according to any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or N-(2-benzo[1,3]dioxol-5-yl-ethyl)-2-(4-methyl-benzyl)-succinamic acid or a pharmaceutically acceptable salt or pharmaceutical composition thereof to inhibit or reduce the production of kynurenine in the subject. In some aspects, the method for inhibiting or reducing the production of kynurenine in a subject in need thereof comprising administering to the subject an effective amount of a compound according to any one of Formulae Ia, Ib, Ic, I, IIa, and IIb or a pharmaceutically acceptable salt or pharmaceutical composition thereof to inhibit or reduce the production of kynurenine in the subject. In some embodiments the compound used in the method for inhibiting or reducing the production of kynurenine in the subject in need thereof is selected from the group consisting of or a pharmaceutically acceptable salt thereof. In some embodiments, the disclosed compounds are administered in combination with one or more additional therapeutic agents. The combination of active agents can be administered in the same or different admixture. The combination of active agents can be administered at concurrently or sequential. Current treatments for many age-related disorders, including frailty are generally limited to treating the physical symptoms of the disease. For example, frail individuals may be encouraged to increase their amount of exercise and dietary intake, which can induce weight gain, increase mobility, enhance physical performance, improve gait, improve balance, increase bone mineral density, and increase general well-being (Espinoza and Walston,Cleveland Clinic Journal of Medicine,72 (12):1105-1112 (2005)). Pharmaceutical treatments can include agents to improve appetite, analgesics, or hormone replacement therapy. Many of these traditional remedies are insufficient alone because they are limited to managing symptoms of the disease (such as pain), their efficacy is low, or they require the help or service of a caregiver (for example exercise or physical therapy). The compounds and pharmaceutical compositions thereof can be administered, for example, parenterally (e.g., intramuscular, intraperitoneal, intravenous (IV) or subcutaneous) injection or infusion enterally (e.g., orally), or topically. Topical administration can include application to the lungs, nasal, oral (sublingual, buccal), vaginal, or rectal mucosa. In some embodiments, the compositions are administered in combination with transdermal or mucosal transport elements. In some embodiments, the composition is administered for e.g., days, weeks, months or years. In some embodiments, the composition is administered indefinitely, (e.g., as a nutraceutical with no duration limit). In some embodiments, the composition is administered daily. EXAMPLES Example I: Receptor-Based Virtual Screening Methods and Materials Methods for detecting kynurenine levels in cell culture media following IDO1 inhibitor treatment are as follows. The initial step in Phase I study was to develop an assay that could be utilized to measure kynurenine levels in vitro as a strategy to identify effective IDO1 inhibitors. Primary human myoblast cells (Gibco A12555) were treated with interferon gamma (100 ng/ml human IFN-gamma; R&D Systems cat. 285-IF) to increase IDO1 activity and then the cells were treated with various molecules previously been reported to suppress IDO1 activity. These inhibitors (10 μg/mL) included a known inhibitor of IDO1, indoximod (1-methyl-D-tryptophan), as well as a number of other compounds previously published as inhibiting IDO1, such as brassinin and rosmarinic acid. Kynurenine levels were measured in conditioned media 24 hrs after treatment and normalized to protein concentration of lysate in each well. Kynurenine levels were examined using ELISA performed at the Georgia Cancer Center and using LC/MS performed at the Medical College of Georgia (MCG) Proteomics Core Facility. Detection of kynurenine in samples was also performed using competitive ELISA kit (IBL America Cat #IB89190) according to manufacturer's instructions. In brief, 10 ul of samples and standards were mixed with 250 ul of acylation buffer and 25ul of acylation reagent and incubated for 90 min at 37° C. 20 ul of acylation reagent was used for 0/N incubation with Kynurenine Antiserum in Kynurenine Microtiter Strips. The next day, wells were washed and incubated for 30 min with 100 ul of goat anti-rabbit conjugated with perodixase. After washing, wells were incubated for 25 min with 100 ul of substrate followed by 100 ul stop solution. Absorbance at 450 nm was read using microplate reader. Standard curves and kynurenine concentration were calculated using R package installed on R 3.5.2. Limit of detection was calculated as the average of background samples minus 3×SD. Assay and data calculations were performed at Immune Monitoring Shared Resource (Augusta University). The data indicate that the two approaches (i.e. ELISA and LC/MS) yield similar results and are significantly correlated (P<0.01, r=0.94). ELISA assay was used for subsequent analyses as it was most effective for processing large numbers of samples. Several molecules were screened to identify compounds that block IDO1 in primary human skeletal muscles cells and in vascular smooth muscle cells. Cells were treated with inhibitors as described above and kynurenine levels were analyzed using ELISA assay. Results Kynurenine levels were examined using ELISA performed at the Georgia Cancer Center and using LC/MS performed at the Medical College of Georgia (MCG) Proteomics Core Facility to determine the most effective outcome measure for identifying IDO1 activity and inhibition. As shown inFIGS.1A and1B, the results demonstrate that the two approaches yield similar results and are significantly correlated (P<0.01, r=0.94). The two bars inFIG.1Arepresent replicates whereas the bars inFIG.1Brepresent data from the LC/MS approach. Regardless which method was used, the outcomes were similar. ELISA assay was used for subsequent analyses as it was most effective for processing large numbers of samples. A known inhibitor of IDO1, Indoximod (1-methyl-D-tryptophan), as well as a number of other naturally occurring compounds previously published as inhibiting IDO1 (e.g., ebselin, brassinin, etc) were screened along with rosmarinic acid and its succinamic acid analog. Primary human skeletal muscle cells (FIG.2A) or vascular smooth muscle cells (FIG.2B) (Gibco) were treated with 100 ng/ml human IFN-gamma (R&D Systems cat. 285-IF) to increase IDO1 activity in the presence of the various inhibitors (10 μg/mL). Kynurenine levels were measured in conditioned media 24 hrs after treatment and normalized to protein concentration of lysate in each well. As shown inFIGS.1A and1B, the results demonstrate that Indoximod is a weak inhibitor of IDO1 in muscle cells, whereas several compounds, such as brassinin and the succinamic acid analog, significantly reduced KYN levels by more than 50% compared to cells exposed to IFN gamma alone. Example II: Chemical Synthesis Compounds described herein, salts and prodrugs thereof can be synthesized by conjugating equimolar quantities of a corresponding carboxylic acid and amine using isobutyl chloroformate as an activating agent for carboxylic acid. Reaction mixture was then taken to dryness and the product was isolated via column chromatography. The structures of exemplary compounds1,2,3, and7are shown below. Example III: Molecular Docking Studies Methods and Materials Molecular docking analysis was carried out on the pocket sites as discussed by Meng, et al.,Curr. Comput. Aided Drug Des.,7 (2):146-157 (2011). Docking was carried out to allow a hybrid approach to calculate the free binding energies and binding affinity using MMGBSA approach. Inter- and Intra-ligand clashes were analyzed and torsions were analyzed to check the possibility of presence of dihedrals in CSD dataset for validation purpose. 2D depiction of the interactions were generated using PoseView. Results Molecular Docking Data The crystal structures of indoleamine 2,3-dioxygenagse 1 (IDO1) complexed with Amg-1 Reference Ligands: SA, RA and PKJ, respectively, and with exemplary compounds disclosed herein, respectively, were obtained (FIGS.3A-3D). While in the foregoing specification this invention has been described in relation to certain embodiments thereof, and many details have been put forth for the purpose of illustration, it will be apparent to those skilled in the art that the invention is susceptible to additional embodiments and that certain of the details described herein can be varied considerably without departing from the basic principles of the invention. All references cited herein are incorporated by reference in their entirety. The present invention may be embodied in other specific forms without departing from the spirit or essential attributes thereof and, accordingly, reference should be made to the appended claims, rather than to the foregoing specification, as indicating the scope of the invention.
118,606
11858909
DEFINITIONS The following are definitions of terms used in the present specification. The initial definition provided for a group or term herein applies to that group or term throughout the present specification individually or as part of another group, unless otherwise indicated. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. As used here, “etoricoxib” refers to 5-chloro-6′-methyl-3-[4-methylsulfonyl)pheny]-2,3′-bipyridine, or a pharmaceutically acceptable salt or solvate thereof. Etoricoxib has the chemical structure: Salts of etoricoxib are described in International Patent Application Publication No. WO 2012/004677 A1 to Actavis Group PTC EHF/Khunt, the contents of which are incorporated herein by reference in their entirety. Polymorphs of etoricoxib are described in International Patent Application Publication No. WO 2005/085199 A1 to Cadila HealthCare Ltd./Lohray, the contents of which are incorporated herein by reference in their entirety. As used herein, “d3-etoricoxib” refers to the compound 5-chloro-6′-(methyl-d3)-3-(4-(methylsulfonyl) phenyl)-2,3′-bipyridine, having the structural formula: As used herein, the term “salt” refers to any and all salts, and encompasses pharmaceutically acceptable salts. Salts include ionic compounds that result from the neutralization reaction of an acid and a base. A salt is composed of one or more cations (positively charged ions) and one or more anions (negative ions) so that the salt is electrically neutral (without a net charge). Salts of the compounds of this invention include those derived from inorganic and organic acids and bases. Examples of acid addition salts are salts of an amino group formed with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid, or with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid or by using other methods known in the art such as ion exchange. Other salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, hippurate, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1-4alkyl)4salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further salts include ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate. The term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid or with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid or by using other methods known in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium, and N+(C1-4alkyl)4−salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate. The term “solvate” refers to forms of the compound, or a salt thereof, that are associated with a solvent, usually by a solvolysis reaction. In some embodiments, this physical association includes hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, diethyl ether, and the like. In some embodiments, the compounds disclosed herein are prepared, e.g., in crystalline form, and are solvated. Suitable solvates include pharmaceutically acceptable solvates and further include both stoichiometric solvates and non-stoichiometric solvates. In certain instances, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated in the crystal lattice of a crystalline solid. “Solvate” encompasses both solution-phase and isolatable solvates. Representative solvates include hydrates, ethanolates, and methanolates. The term “stoichiometric solvate” refers to a solvate, which comprises a compound (e.g., a compound disclosed herein) and a solvent, wherein the solvent molecules are an integral part of the crystal lattice, in which they interact strongly with the compound and each other. The removal of the solvent molecules will cause instability of the crystal network, which subsequently collapses into an amorphous phase or recrystallizes as a new crystalline form with reduced solvent content. The term “non-stoichiometric solvate” refers to a solvate, which comprises a compound (e.g., a compound disclosed herein) and a solvent. In some embodiments, the solvent content varies without major changes in the crystal structure. The amount of solvent in the crystal lattice only depends on the partial pressure of solvent in the surrounding atmosphere. In the fully solvated state, non-stoichiometric solvates may, but not necessarily have to, show an integer molar ratio of solvent to the compound. In some embodiments, during drying of a non-stoichiometric solvate, a portion of the solvent is removed without significantly disturbing the crystal network, and the resulting solvate can subsequently be resolvated to give the initial crystalline form. Unlike stoichiometric solvates, the desolvation and resolvation of non-stoichiometric solvates is not accompanied by a phase transition, and all solvation states represent the same crystal form. The term “hydrate” refers to a compound that is associated with water. Typically, the number of the water molecules contained in a hydrate of a compound is in a definite ratio to the number of the compound molecules in the hydrate. Therefore, a hydrate of a compound may be represented, for example, by the general formula R·x H2O, wherein R is the compound, and x is a number greater than 0. In some embodiments, a given compound forms more than one type of hydrate, including, e.g., monohydrates (x is 1), lower hydrates (x is a number greater than 0 and smaller than 1, e.g., hemihydrates (R·0.5 H2O)), and polyhydrates (x is a number greater than 1, e.g., dihydrates (R·2 H2O) and hexahydrates (R·6 H2O)). The term “crystalline” or “crystalline form” refers to a solid form substantially exhibiting long-range three-dimensional order. In certain embodiments, a crystalline form of a solid is a solid form that is substantially not amorphous. In certain embodiments, the X-ray powder diffraction (XRPD) pattern of a crystalline form includes one or more sharply defined peaks. In some embodiments, the composition comprises one or more crystalline forms. The term “amorphous” or “amorphous form” refers to a form of a solid (“solid form”), the form substantially lacking three-dimensional order. In certain embodiments, an amorphous form of a solid is a solid form that is substantially not crystalline. In certain embodiments, the X-ray powder diffraction (XRPD) pattern of an amorphous form includes a wide scattering band with a peak at 2θ of, e.g., between 20 and 70°, inclusive, using CuKα radiation. In certain embodiments, the XRPD pattern of an amorphous form further includes one or more peaks attributed to crystalline structures. In certain embodiments, the maximum intensity of any one of the one or more peaks attributed to crystalline structures observed at a 2θ of between 20 and 70°, inclusive, is not more than 300-fold, not more than 100-fold, not more than 30-fold, not more than 10-fold, or not more than 3-fold of the maximum intensity of the wide scattering band. In certain embodiments, the XRPD pattern of an amorphous form includes no peaks attributed to crystalline structures. The term “co-crystal” refers to a crystalline structure comprising at least two different components (e.g., a compound disclosed herein and an acid), wherein each of the components is independently an atom, ion, or molecule. In certain embodiments, none of the components is a solvent. In certain embodiments, at least one of the components is a solvent. A co-crystal of a compound disclosed herein and an acid is different from a salt formed from a compound disclosed herein and the acid. In the salt, a compound disclosed herein is complexed with the acid in a way that proton transfer (e.g., a complete proton transfer) from the acid to a compound disclosed herein easily occurs at room temperature. In the co-crystal, however, a compound disclosed herein is complexed with the acid in a way that proton transfer from the acid to a compound disclosed herein does not easily occur at room temperature. In certain embodiments, in the co-crystal, there is no proton transfer from the acid to a compound disclosed herein. In certain embodiments, in the co-crystal, there is partial proton transfer from the acid to a compound disclosed herein. In some embodiments, co-crystals improve the properties (e.g., solubility, stability, and ease of formulation) of a compound disclosed herein. The term “polymorph” refers to a crystalline form of a compound (or a salt, hydrate, or solvate thereof). All polymorphs have the same elemental composition. Different crystalline forms usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and solubility. In some embodiments, recrystallization solvent, rate of crystallization, storage temperature, and other factors cause one crystal form to dominate. In some embodiments, various polymorphs of a compound are prepared by crystallization under different conditions. The term “prodrug” as employed herein denotes a compound that, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of the present subject matter, or a salt and/or solvate thereof. The term “isotopes” refers to variants of a particular chemical element such that, while all isotopes of a given element share the same number of protons in each atom of the element, those isotopes differ in the number of neutrons. Deuterium is a stable isotope of hydrogen with a nucleus consisting of one proton and one neutron. The term “alkyl” refers to a radical of a straight-chain or branched saturated hydrocarbon group having from 1 to 20 carbon atoms (“C1-20alkyl”). In some embodiments, an alkyl group has 1 to 12 carbon atoms (“C1-12alkyl”). In some embodiments, an alkyl group has 1 to 10 carbon atoms (“C1-10alkyl”). In some embodiments, an alkyl group has 1 to 9 carbon atoms (“C1-9alkyl”). In some embodiments, an alkyl group has 1 to 8 carbon atoms (“C1-8alkyl”). In some embodiments, an alkyl group has 1 to 7 carbon atoms (“C1-7alkyl”). In some embodiments, an alkyl group has 1 to 6 carbon atoms (“C1-6alkyl”). In some embodiments, an alkyl group has 1 to 5 carbon atoms (“C1-5alkyl”). In some embodiments, an alkyl group has 1 to 4 carbon atoms (“C1-4alkyl”). In some embodiments, an alkyl group has 1 to 3 carbon atoms (“C1-3alkyl”). In some embodiments, an alkyl group has 1 to 2 carbon atoms (“C1-2alkyl”). In some embodiments, an alkyl group has 1 carbon atom (“C1alkyl”). In some embodiments, an alkyl group has 2 to 6 carbon atoms (“C2-6alkyl”). Examples of C1-6alkyl groups include methyl (C1), ethyl (C2), propyl (C3) (e.g., n-propyl, isopropyl), butyl (C4) (e.g., n-butyl, tert-butyl, sec-butyl, isobutyl), pentyl (C5) (e.g., n-pentyl, 3-pentanyl, amyl, neopentyl, 3-methyl-2-butanyl, tert-amyl), and hexyl (C6) (e.g., n-hexyl). Additional examples of alkyl groups include n-heptyl (C7), n-octyl (C8), n-dodecyl (C12), and the like. Unless otherwise specified, each instance of an alkyl group is independently unsubstituted (an “unsubstituted alkyl”) or substituted (a “substituted alkyl”) with one or more substituents (e.g., halogen, such as F). In certain embodiments, the alkyl group is an unsubstituted C1-12alkyl (such as unsubstituted C1-6alkyl, e.g., —CH3(Me), unsubstituted ethyl (Et), unsubstituted propyl (Pr, e.g., unsubstituted n-propyl (n-Pr), unsubstituted isopropyl (i-Pr)), unsubstituted butyl (Bu, e.g., unsubstituted n-butyl (n-Bu), unsubstituted tert-butyl (tert-Bu or t-Bu), unsubstituted sec-butyl (sec-Bu or s-Bu), unsubstituted isobutyl (i-Bu)). In certain embodiments, the alkyl group is a substituted C1-12alkyl (such as substituted C1-6alkyl, e.g., —CH2F, —CHF2, —CF3, —CH2CH2F, —CH2CHF2, —CH2CF3, or benzyl (Bn)). A group is optionally substituted unless expressly provided otherwise. The term “optionally substituted” refers to being substituted or unsubstituted. In certain embodiments, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl groups are optionally substituted. “Optionally substituted” refers to a group which is substituted or unsubstituted (e.g., “substituted” or “unsubstituted” alkyl, “substituted” or “unsubstituted” alkenyl, “substituted” or “unsubstituted” alkynyl, “substituted” or “unsubstituted” heteroalkyl, “substituted” or “unsubstituted” heteroalkenyl, “substituted” or “unsubstituted” heteroalkynyl, “substituted” or “unsubstituted” carbocyclyl, “substituted” or “unsubstituted” heterocyclyl, “substituted” or “unsubstituted” aryl or “substituted” or “unsubstituted” heteroaryl group). In general, the term “substituted” means that at least one hydrogen present on a group is replaced with a permissible substituent, e.g., a substituent which upon substitution results in a stable compound, e.g., a compound which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, or other reaction. Unless otherwise indicated, a “substituted” group has a substituent at one or more substitutable positions of the group, and when more than one position in any given structure is substituted, the substituent is either the same or different at each position. The term “substituted” is contemplated to include substitution with all permissible substituents of organic compounds, and includes any of the substituents described herein that results in the formation of a stable compound. The present invention contemplates any and all such combinations in order to arrive at a stable compound. For purposes of this invention, heteroatoms such as nitrogen may have hydrogen substituents and/or any suitable substituent as described herein which satisfy the valencies of the heteroatoms and results in the formation of a stable moiety. The invention is not limited in any manner by the exemplary substituents described herein. Exemplary carbon atom substituents include halogen, —CN, —NO2, —N3, —SO2H, —SO3H, —OH, —ORaa, —ON(Rbb)2, —N(Rbb)2, —N(Rbb)3+X−, —N(ORcc)Rbb, —SH, —SRaa, —SSRcc, —C(═O)Raa, —CO2H, —CHO, —C(ORcc)2, —CO2Raa, —OC(═O)Raa, —OCO2Raa, —C(═O)N(Rbb)2, —OC(═O)N(Rbb)2, —NRbbC(═O)Raa, —NRbbCO2Raa, —NRbbC(═O)N(Rbb)2, —C(═NRbb)Raa, —C(═NRbb)ORaa, —OC(═NRbb)Raa, —OC(═NRbb)ORaa, —C(═NRbb)N(Rbb)2, —OC(═NRbb)N(Rbb)2, —NRbbC(═NRbb)N(Rbb)2, —C(═O)NRbbSO2Raa, —NRbbSO2Raa, —SO2N(Rbb)2, —SO2Raa, —SO2ORaa, —OSO2Raa, —S(═O)Raa, —OS(═O)Raa, —Si(Raa)3, —OSi(Raa)3—C(═S)N(Rbb)2, —C(═O)SRaa, —C(═S)SRaa, —SC(═S)SRaa, —SC(═O)SRaa, —OC(═O)SRaa, —SC(═O)ORaa, —SC(═O)Raa, —P(═O)(Raa)2, —P(═O)(ORcc)2, —OP(═O)(Raa)2, —OP(═O)(ORcc)2, —P(═O)(N(Rbb)2)2, —OP(═O)(N(Rbb)2)2, —NRbbP(═O)(Raa)2, —NRbbP(═O)(ORcc)2, —NRbbP(═O)(N(Rbb)2)2, —P(Rcc)2, —P(ORcc)2, —P(Rcc)3+X−, —P(ORcc)3+X−, —P(Rcc)4, —P(ORcc)4, —OP(Rcc)2, —OP(Rcc)3+X−, —OP(ORcc)2, —OP(ORcc)3+X−, —OP(Rcc)4, —OP(ORcc)4, —B(Raa)2, —B(ORcc)2, —BRaa(ORcc), C1-20alkyl, C1-20perhaloalkyl, C1-20alkenyl, C1-20alkynyl, heteroC1-20alkyl, heteroC1-20alkenyl, heteroC1-20alkynyl, C3-10carbocyclyl, 3-14 membered heterocyclyl, C6-14aryl, and 5-14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rddgroups; wherein X−is a counterion;or two geminal hydrogens on a carbon atom are replaced with the group ═O, ═S, ═NN(Rbb)2, ═NNRbbC(═O)Raa, ═NNRbbC(═O)ORaa, ═NNRbbS(═O)2Raa, ═NRbb, or ═NORcc;wherein:each instance of Raais, independently, selected from C1-20alkyl, C1-20perhaloalkyl, C1-20alkenyl, C1-20alkynyl, heteroC1-20alkyl, heteroC1-20alkenyl, heteroC1-20alkynyl, C3-10carbocyclyl, 3-14 membered heterocyclyl, C6-14aryl, and 5-14 membered heteroaryl, or two Raagroups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each of the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rddgroups;each instance of Rbbis, independently, selected from hydrogen, —OH, —ORaa, —N(Rcc)2, —CN, —C(═O)Raa, —C(═O)N(Rcc)2, —CO2Raa, —SO2Raa, —C(═NRcc)ORaa, —C(═NRcc)N(Rcc)2, —SO2N(Rcc)2, —SO2Rcc, —SO2ORcc, —SORaa, —C(═S)N(Rcc)2, —C(═O)SRcc, —C(═S)SRcc, —P(═O)(Raa)2, —P(═O)(ORcc)2, —P(═O)(N(Rcc)2)2, C1-20alkyl, C1-20perhaloalkyl, C1-20alkenyl, C1-20alkynyl, heteroC1-20alkyl, heteroC1-20alkenyl, heteroC1-20alkynyl, C3-10carbocyclyl, 3-14 membered heterocyclyl, C6-14aryl, and 5-14 membered heteroaryl, or two Rbbgroups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rddgroups;each instance of Rccis, independently, selected from hydrogen, C1-20alkyl, C1-20perhaloalkyl, C1-20alkenyl, C1-20alkynyl, heteroC1-20alkyl, heteroC1-20alkenyl, heteroC1-20alkynyl, C3-10carbocyclyl, 3-14 membered heterocyclyl, C6-14aryl, and 5-14 membered heteroaryl, or two Rccgroups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rddgroups;each instance of Rddis, independently, selected from halogen, —CN, —NO2, —N3, —SO2H, —SO3H, —OH, —ORee, —ON(Rff)2, —N(Rff)2, —N(Rff)3+X−, —N(ORee)Rff, —SH, —SRee, —SSRee, —C(═O)Ree, —CO2H, —CO2Ree, —OC(═O)Ree, —OCO2Ree, —C(═O)N(Rff)2, —OC(═O)N(Rff)2, —NRffC(═O)Ree, —NRffCO2Ree, —NRffC(═O)N(Rff)2, —C(═NRff)ORee, —OC(═NRff)Ree, —OC(═NRff)ORee, —C(═NRff)N(Rff)2, —OC(═NRff)N(Rff)2, —NRffC(═NRff)N(Rff)2, —NRffSO2Ree, —SO2N(Rff)2, —SO2Ree, —SO2ORee, —OSO2Ree, —S(═O)Ree, —Si(Ree)3, —OSi(Ree)3, —C(═S)N(Rff)2, —C(═O)SRee, —C(═S)SRee, —SC(═S)SRee, —P(═O)(ORee)2, —P(═O)(Ree)2, —OP(═O)(Ree)2, —OP(═O)(ORee)2, C1-10alkyl, C1-10perhaloalkyl, C1-10alkenyl, C1-10alkynyl, heteroC1-10alkyl, heteroC1-10alkenyl, heteroC1-10alkynyl, C3-10carbocyclyl, 3-10 membered heterocyclyl, C6-10aryl, and 5-10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rgggroups, or two geminal Rddsubstituents are joined to form ═O or ═S; wherein X−is a counterion;each instance of Reeis, independently, selected from C1-10alkyl, C1-10perhaloalkyl, C1-10alkenyl, C1-10alkynyl, heteroC1-10alkyl, heteroC1-10alkenyl, heteroC1-10alkynyl, C3-10carbocyclyl, C6-10aryl, 3-10 membered heterocyclyl, and 3-10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rgggroups;each instance of Rffis, independently, selected from hydrogen, C1-10alkyl, C1-10perhaloalkyl, C1-10alkenyl, C1-10alkynyl, heteroC1-10alkyl, heteroC1-10alkenyl, heteroC1-10alkynyl, C3-10carbocyclyl, 3-10 membered heterocyclyl, C6-10aryl, and 5-10 membered heteroaryl, or two Rffgroups are joined to form a 3-10 membered heterocyclyl or 5-10 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rgggroups;each instance of Rggis, independently, halogen, —CN, —NO2, —N3, —SO2H, —SO3H, —OH, —OC1-6alkyl, —ON(C1-6alkyl), —N(C1-6alkyl)2, —N(C1-6alkyl)3+X−, —NH(C1-6alkyl)2+X−, —NH2(C1-6alkyl)+X−, —NH3+X−, —N(OC1-6alkyl)(C1-6alkyl), —N(OH)(C1-6alkyl), —NH(OH), —SH, —SC1-6alkyl, —SS(C1-6alkyl), —C(═O)(C1-6alkyl), —CO2H, —CO2(C1-6alkyl), —OC(═O)(C1-6alkyl), —OCO2(C1-6alkyl), —C(═O)NH2, —C(═O)N(C1-6alkyl)2, —OC(═O)NH(C1-6alkyl), —NHC(═O)(C1-6alkyl), —N(C1-6alkyl)C(═O)(C1-6alkyl), —NHCO2(C1-6alkyl), —NHC(═O)N(C1-6alkyl)2, —NHC(═O)NH(C1-6alkyl), —NHC(═O)NH2, —C(═NH)O(C1-6alkyl), —OC(═NH)(C1-6alkyl), —OC(═NH)OC1-6alkyl, —C(═NH)N(C1-6alkyl)2, —C(═NH)NH(C1-6alkyl), —C(═NH)NH2, —OC(═NH)N(C1-6alkyl)2, —OC(NH)NH(C1-6alkyl), —OC(NH)NH2, —NHC(NH)N(C1-6alkyl)2, —NHC(═NH)NH2, —NHSO2(C1-6alkyl), —SO2N(C1-6alkyl)2, —SO2NH(C1-6alkyl), —SO2NH2, —SO2C1-6alkyl, —SO2OC1-6alkyl, —OSO2C1-6alkyl, —SOC1-6alkyl, —Si(C1-6alkyl)3, —OSi(C1-6alkyl)3—C(═S)N(C1-6alkyl)2, C(═S)NH(C1-6alkyl), C(═S)NH2, —C(═O)S(C1-6alkyl), —C(═S)SC1-6alkyl, —SC(═S)SC1-6alkyl, —P(═O)(OC1-6alkyl)2, —P(═O)(C1-6alkyl), —OP(═O)(C1-6alkyl), —OP(═O)(OC1-6alkyl)2, C1-10alkyl, C1-10perhaloalkyl, C1-10alkenyl, C1-10alkynyl, heteroC1-10alkyl, heteroC1-10alkenyl, heteroC1-10alkynyl, C3-10carbocyclyl, C6-10aryl, 3-10 membered heterocyclyl, or 5-10 membered heteroaryl; or two geminal Rggsubstituents can be joined to form ═O or ═S; andeach X−is a counterion. In certain embodiments, each carbon atom substituent is independently halogen, substituted (e.g., substituted with one or more halogen) or unsubstituted C1-6alkyl, —ORaa, —SRaa, —N(Rbb)2, —CN, —SCN, —NO2, —C(═O)Raa, —CO2Raa, —C(═O)N(Rbb)2, —OC(═O)Raa, —OCO2Raa, —OC(═O)N(Rbb)2, —NRbbC(═O)Raa, —NRbbCO2Raa, or —NRbbC(═O)N(Rbb)2. In certain embodiments, each carbon atom substituent is independently halogen, substituted (e.g., substituted with one or more halogen) or unsubstituted C1-10alkyl, —ORaa, —SRaa, —N(Rbb)2, —CN, —SCN, —NO2, —C(═O)Raa, —CO2Raa, —C(═O)N(Rbb)2, —OC(═O)Raa, —OCO2Raa, —OC(═O)N(Rbb)2, —NRbbC(═O)Raa, —NRbbCO2Raa, or —NRbbC(═O)N(Rbb)2, wherein Raais hydrogen, substituted (e.g., substituted with one or more halogen) or unsubstituted C1-10alkyl, an oxygen protecting group (e.g., silyl, TBDPS, TBDMS, TIPS, TES, TMS, MOM, THP, t-Bu, Bn, allyl, acetyl, pivaloyl, or benzoyl) when attached to an oxygen atom, or a sulfur protecting group (e.g., acetamidomethyl, t-Bu, 3-nitro-2-pyridine sulfenyl, 2-pyridine-sulfenyl, or triphenylmethyl) when attached to a sulfur atom; and each Rbbis independently hydrogen, substituted (e.g., substituted with one or more halogen) or unsubstituted C1-10alkyl, or a nitrogen protecting group (e.g., Bn, Boc, Cbz, Fmoc, trifluoroacetyl, triphenylmethyl, acetyl, or Ts). In certain embodiments, each carbon atom substituent is independently halogen, substituted (e.g., substituted with one or more halogen) or unsubstituted C1-6alkyl, —ORaa, —SRaa, —N(Rbb)2, —CN, —SCN, or —NO2. In certain embodiments, each carbon atom substituent is independently halogen, substituted (e.g., substituted with one or more halogen moieties) or unsubstituted C1-10alkyl, —ORaa, —SRaa, —N(Rbb)2, —CN, —SCN, or —NO2, wherein Raais hydrogen, substituted (e.g., substituted with one or more halogen) or unsubstituted C1-10alkyl, an oxygen protecting group (e.g., silyl, TBDPS, TBDMS, TIPS, TES, TMS, MOM, THP, t-Bu, Bn, allyl, acetyl, pivaloyl, or benzoyl) when attached to an oxygen atom, or a sulfur protecting group (e.g., acetamidomethyl, t-Bu, 3-nitro-2-pyridine sulfenyl, 2-pyridine-sulfenyl, or triphenylmethyl) when attached to a sulfur atom; and each Rbbis independently hydrogen, substituted (e.g., substituted with one or more halogen) or unsubstituted C1-10alkyl, or a nitrogen protecting group (e.g., Bn, Boc, Cbz, Fmoc, trifluoroacetyl, triphenylmethyl, acetyl, or Ts). As used herein, the “purity” of a compound refers to the amount of the compound in a composition relative to the total amount of the composition. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 80% to 100%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%. In certain embodiments, embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 80%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 81%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 82%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 83%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 84%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 85%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 86%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 87%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 88%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 89%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 90%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 91%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 92%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 93%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 94%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 95%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 96%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 97%. In certain embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 98%. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has a purity of at least 99%. In some embodiments, the purity of compounds and compositions disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib) is determined as a percent (%) area basis. In some embodiments, purity is quantified by analytical chromatography. In some embodiments, purity is quantified by HPLC, UHPLC, or UPLC. In certain embodiments, purity is quantified by HPLC. As used herein, the term “isotopic purity” refers to the percentage of molecules of an isotopically enriched compound (e.g., incorporating one or more heavy atoms, e.g., deuterium) present relative to the total number of molecules of all isotopes of the compound. For example, the isotopic purity of d3-etoricoxib as recited herein refers to the percentage of molecules of d3-etoricoxib present relative to the total number of molecules of etoricoxib isotopes. In some embodiments, a compound disclosed herein has an isotopic purity of at least 50.0%, 60.0%, 70.0%, 75.0%, 80.0%, 85.0%, 90.0%, 95.0%, 97.0%, 98.0%, 99.0%, 99.5%, 99.7%, 99.8%, or 99.9%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 50.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 60.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 70.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 75.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 80.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 85.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 90.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 95.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 97.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 98.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 99.0%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 99.5%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 99.7%. In some embodiments, a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 99.9%. More specifically, a “compound” may be considered to include more than a single molecule. For example, in some embodiments, the compound is present in an amount measured in micrograms, milligrams, grams, or kilograms, and as such comprises a large number of individual molecules. As used herein, the terms “isotopic enrichment” or “isotopically enriched” refer to a compound which comprises a greater percentage of one or more heavy atoms (e.g., deuterium) than that which would occur naturally, i.e., as a result of natural abundance. The terms “isotopic enrichment” or “isotopically enriched” may also refer to a particular site (or particular sites) on a molecule which comprise a greater percentage of isotopic atoms (e.g., deuterium) at that site or sites of the molecule than that which would occur naturally, i.e., as a result of natural abundance. In some embodiments, isotopically-enriched etoricoxib disclosed herein comprises a greater percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib than would occur as a result of the natural abundance of deuterium (i.e., greater than approximately 0.0115% to 0.0156% relative to the total number of hydrogen isotopes). In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 0.05%, at least 0.1%, at least 0.5%, at least 1.0, at least 2.0%, at least 3.0%, at least 4.0%, at least 5.0%, at least 10.0%, at least 20.0%, at least 30.0%, at least 40.0%, at least 50.0%, at least 60.0%, at least 70.0%, at least 75.0%, at least 80.0%, at least 85.0%, at least 90.0%, at least 94.0%, at least 95.0%, at least 96.0%, at least 97.0%, at least 98.0%, at least 99.0%, at least 99.5%, at least 99.7%, at least 99.8%, at least 99.9%, or at least 100%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 0.05%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 0.1%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 0.5%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 1.0. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 2.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 3.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 4.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 5.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 10.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 20.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 30.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 40.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 50.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 60.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 70.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 75.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 80.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 85.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 90.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 94.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 95.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 96.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 97.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 98.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 99.0%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 99.5%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 99.7%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 99.8%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 99.9%. In some embodiments, the percentage of the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is isotopically enriched by at least 100%. In some embodiments, the compound of Formula (I) is isotopically enriched at one or more of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15. In certain embodiments, the compound of Formula (I) is isotopically enriched at one or more of R13, R14, and R15. In some embodiments, the compound of Formula (I) is isotopically enriched at each of R13, R14, and R15. As used herein, the term “the percentage of deuterium” refers to the percentage of hydrogen atoms replaced by deuterium atoms in a compound disclosed herein. The term “total amount of etoricoxib” refers to the combined total amount of deuterated etoricoxib and non-isotopically enriched etoricoxib in a given composition. The terms “composition” and “formulation” are used interchangeably. A “subject” to which administration is contemplated refers to a human (i.e., male or female of any age group, e.g., pediatric subject (e.g., infant, child, or adolescent) or adult subject (e.g., young adult, middle-aged adult, or senior adult)) or non-human animal. In certain embodiments, the non-human animal is a mammal (e.g., primate (e.g., cynomolgus monkey or rhesus monkey), commercially relevant mammal (e.g., cattle, pig, horse, sheep, goat, cat, or dog), or bird (e.g., commercially relevant bird, such as chicken, duck, goose, or turkey)). In certain embodiments, the non-human animal is a fish, reptile, or amphibian. In some embodiments, the non-human animal is a male or female at any stage of development. In some embodiments, the non-human animal is a transgenic animal or genetically engineered animal. As used herein, the term “patient” refers to a subject that is a human or animal. In some embodiments, the patient is a healthy patient who is in a generally healthy condition. In some embodiments, the patient is a health patient who is in a generally healthy condition and is eligible to participate as a healthy volunteer in a Phase 1 pharmacokinetic study of a compound or pharmaceutical composition disclosed herein. The term “biological sample” refers to any sample including tissue samples (such as tissue sections and needle biopsies of a tissue); cell samples (e.g., cytological smears (such as Pap or blood smears) or samples of cells obtained by microdissection); samples of whole organisms (such as samples of yeasts or bacteria); or cell fractions, fragments or organelles (such as obtained by lysing cells and separating the components thereof by centrifugation or otherwise). Other examples of biological samples include blood, serum, urine, semen, fecal matter, cerebrospinal fluid, interstitial fluid, mucous, tears, sweat, pus, biopsied tissue (e.g., obtained by a surgical biopsy or needle biopsy), nipple aspirates, milk, vaginal fluid, saliva, swabs (such as buccal swabs), or any material containing biomolecules that is derived from a first biological sample. The term “administer,” “administering,” or “administration” refers to implanting, absorbing, ingesting, injecting, inhaling, or otherwise introducing a compound disclosed herein, or a composition thereof, in or on a subject. The terms “treatment,” “treat,” and “treating” refer to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease disclosed herein. In some embodiments, treatment is administered after one or more signs or symptoms of the disease have developed or have been observed. In other embodiments, treatment is administered in the absence of signs or symptoms of the disease. For example, in some embodiments, treatment is administered to a susceptible subject prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of exposure to a pathogen). In some embodiments, treatment is continued after symptoms have resolved, for example, to delay or prevent recurrence of a disease or condition. The terms “condition,” “disease,” and “disorder” are used interchangeably. An “effective amount” of a compound disclosed herein refers to an amount sufficient to elicit the desired biological response. In some embodiments, an effective amount of a compound disclosed herein varies depending on such factors as the desired biological endpoint, severity of side effects, disease, or disorder, the identity, pharmacokinetics, and pharmacodynamics of the particular compound, the condition being treated, the mode, route, and desired or required frequency of administration, the species, age and health or general condition of the subject. In certain embodiments, an effective amount is a therapeutically effective amount. In certain embodiments, an effective amount is a prophylactic treatment. In certain embodiments, an effective amount is the amount of a compound disclosed herein in a single dose. In certain embodiments, an effective amount is the combined amounts of a compound disclosed herein in multiple doses. In certain embodiments, the desired dosage is delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage is delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations). A “therapeutically effective amount” of a compound disclosed herein is an amount sufficient to provide a therapeutic benefit in the treatment of a condition or to delay or minimize one or more symptoms associated with the condition. A therapeutically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other therapies, which provides a therapeutic benefit in the treatment of the condition. The term “therapeutically effective amount” can encompass an amount that improves overall therapy, reduces or avoids symptoms, signs, or causes of the condition, and/or enhances the therapeutic efficacy of another therapeutic agent. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting COX activity. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting COX-1 activity. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting COX-2 activity. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting COX-3 activity. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting one or more of COX-1 activity, COX-2 activity, and/or COX-3 activity. In certain embodiments, a therapeutically effective amount is an amount sufficient for treating pain, fever, or inflammation. In certain embodiments, a therapeutically effective amount is an amount sufficient for treating pain, fever, or inflammation. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting COX activity and treating pain, fever, or inflammation. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting COX-1 activity and treating pain, fever, or inflammation. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting COX-2 activity and treating pain, fever, or inflammation. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting COX-3 activity and treating pain, fever, or inflammation. In certain embodiments, a therapeutically effective amount is an amount sufficient for inhibiting one or more of COX-1 activity, COX-2 activity, and/or COX-3 activity and treating pain, fever, or inflammation. A “prophylactically effective amount” of a compound disclosed herein is an amount sufficient to prevent a condition, or one or more symptoms associated with the condition or prevent its recurrence. A prophylactically effective amount of a compound means an amount of a therapeutic agent, alone or in combination with other agents, which provides a prophylactic benefit in the prevention of the condition. The term “prophylactically effective amount” can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of another prophylactic agent. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting COX activity. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting COX-1 activity. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting COX-2 activity. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting COX-3 activity. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting one or more of COX-1 activity, COX-2 activity, and/or COX-3 activity. In certain embodiments, a prophylactically effective amount is an amount sufficient for preventing pain, fever, or inflammation. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting COX activity and preventing pain, fever, or inflammation. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting COX-1 activity and preventing pain, fever, or inflammation. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting COX-2 activity and preventing pain, fever, or inflammation. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting COX-3 activity and preventing pain, fever, or inflammation. In certain embodiments, a prophylactically effective amount is an amount sufficient for inhibiting one or more of COX-1 activity, COX-2 activity, and/or COX-3 activity and preventing pain, fever, or inflammation. The phrase “same or equivalent amount,” as used herein refers to amounts as measured by mass or by moles, respectively. As used herein the term “inhibit” or “inhibition” in the context of enzymes, for example, in the context of COX or COX-2, refers to a reduction in the activity of the enzyme. In some embodiments, the term refers to a reduction of the level of enzyme activity, e.g., COX activity or COX-2 activity, to a level that is statistically significantly lower than an initial level, which may, for example, be a baseline level of enzyme activity. In some embodiments, the term refers to a reduction of the level of enzyme activity, e.g., COX activity or COX-2 activity, to a level that is less than 75%, less than 50%, less than 40%, less than 30%, less than 25%, less than 20%, less than 10%, less than 9%, less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, less than 3%, less than 2%, less than 1%, less than 0.5%, less than 0.1%, less than 0.01%, less than 0.001%, or less than 0.0001% of an initial level, which may, for example, be a baseline level of enzyme activity. As used herein, the term “mean” refers to a geometric mean unless expressly indicated otherwise. The pharmacokinetic parameters such as “mean Cmax” or “mean AUC” refers to the geometric mean value of a Cmaxor an AUC, unless expressly indicated otherwise (e.g., an arithmetic mean). The term “inflammation” refers to the biological response of cells, tissues to harmful stimuli, such as pathogens, damaged cells, toxic molecules, or irritants. Inflammation takes on many forms and includes, but is not limited to, acute, adhesive, atrophic, catarrhal, chronic, cirrhotic, diffuse, disseminated, exudative, fibrinous, fibrosing, focal, granulomatous, hyperplastic, hypertrophic, interstitial, metastatic, necrotic, obliterative, parenchymatous, plastic, productive, proliferous, pseudomembranous, purulent, sclerosing, seroplastic, serous, simple, specific, subacute, suppurative, toxic, traumatic, and/or ulcerative inflammation. The terms “inflammatory disease” and “inflammatory condition” are used interchangeably herein, and refer to a disease or condition caused by, resulting from, or resulting in inflammation Inflammatory diseases and conditions include those diseases, disorders or conditions that are characterized by signs of pain (dolor, from the generation of noxious substances and the stimulation of nerves), heat (calor, from vasodilatation), redness (rubor, from vasodilatation and increased blood flow), swelling (tumor, from excessive inflow or restricted outflow of fluid), and/or loss of function (functio laesa, partial or complete, temporary or permanent). In some embodiments, the term “inflammatory disease” refers to a dysregulated inflammatory reaction that causes an exaggerated response by macrophages, granulocytes, and/or T-lymphocytes leading to abnormal tissue damage and/or cell death. In some embodiments, an inflammatory disease is either an acute or chronic inflammatory condition and can result from infections or non-infectious causes. Inflammatory diseases include, without limitation, atherosclerosis, arteriosclerosis, autoimmune disorders, multiple sclerosis, systemic lupus erythematosus, polymyalgia rheumatica (PMR), gouty arthritis, degenerative joint disease, degenerative arthritis, tendonitis, bursitis, psoriasis, cystic fibrosis, hemophilic arthropathy, arthrosteitis, rheumatoid arthritis, inflammatory arthritis, Sjogren's syndrome, giant cell arteritis, progressive systemic sclerosis (scleroderma), ankylosing spondylitis, myositis, polymyositis, dermatomyositis, pemphigus, pemphigoid, diabetes (e.g., Type I), myasthenia gravis, Hashimoto's thyroiditis, Graves' disease, Goodpasture's disease, mixed connective tissue disease, sclerosing cholangitis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, pernicious anemia, inflammatory dermatoses, usual interstitial pneumonitis (UIP), asbestosis, silicosis, bronchiectasis, berylliosis, talcosis, pneumoconiosis, sarcoidosis, desquamative interstitial pneumonia, lymphoid interstitial pneumonia, giant cell interstitial pneumonia, cellular interstitial pneumonia, extrinsic allergic alveolitis, Wegener's granulomatosis and related forms of angiitis (temporal arteritis and polyarteritis nodosa), inflammatory dermatoses, hepatitis, delayed-type hypersensitivity reactions (e.g., poison ivy dermatitis), pneumonia, respiratory tract inflammation, Adult Respiratory Distress Syndrome (ARDS), encephalitis, immediate hypersensitivity reactions, asthma, hayfever, allergies, acute anaphylaxis, rheumatic fever, glomerulonephritis, pyelonephritis, cellulitis, cystitis, chronic cholecystitis, ischemia (ischemic injury), reperfusion injury, allograft rejection, host-versus-graft rejection, appendicitis, arteritis, blepharitis, bronchiolitis, bronchitis, cervicitis, cholangitis, chorioamnionitis, conjunctivitis, dacryoadenitis, dermatomyositis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, gingivitis, ileitis, iritis, laryngitis, myelitis, myocarditis, nephritis, omphalitis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, pharyngitis, pleuritis, phlebitis, pneumonitis, proctitis, prostatitis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, testitis, tonsillitis, urethritis, urocystitis, uveitis, vaginitis, vasculitis, vulvitis, vulvovaginitis, angitis, chronic bronchitis, osteomyelitis, optic neuritis, temporal arteritis, transverse myelitis, necrotizing fasciitis, and necrotizing enterocolitis. An ocular inflammatory disease includes, but is not limited to, post-surgical inflammation. Additional exemplary inflammatory conditions include, but are not limited to, inflammation associated with acne, anemia (e.g., aplastic anemia, hemolytic autoimmune anemia), asthma, arteritis (e.g., polyarteritis, temporal arteritis, periarteritis nodosa, Takayasu's arteritis), arthritis (e.g., crystalline arthritis, osteoarthritis, psoriatic arthritis, gouty arthritis, reactive arthritis, rheumatoid arthritis and Reiter's arthritis), ankylosing spondylitis, amylosis, amyotrophic lateral sclerosis, autoimmune diseases, allergies or allergic reactions, atherosclerosis, bronchitis, bursitis, chronic prostatitis, conjunctivitis, Chagas disease, chronic obstructive pulmonary disease, cermatomyositis, diverticulitis, diabetes (e.g., type I diabetes mellitus, Type II diabetes mellitus), a skin condition (e.g., psoriasis, eczema, burns, dermatitis, pruritus (itch)), endometriosis, Guillain-Barre syndrome, infection, ischemic heart disease, Kawasaki disease, glomerulonephritis, gingivitis, hypersensitivity, headaches (e.g., migraine headaches, tension headaches), ileus (e.g., postoperative ileus and ileus during sepsis), idiopathic thrombocytopenic purpura, interstitial cystitis (painful bladder syndrome), gastrointestinal disorder (e.g., selected from peptic ulcers, regional enteritis, diverticulitis, gastrointestinal bleeding, eosinophilic gastrointestinal disorders (e.g., eosinophilic esophagitis, eosinophilic gastritis, eosinophilic gastroenteritis, eosinophilic colitis), gastritis, diarrhea, gastroesophageal reflux disease (GORD, or its synonym GERD), inflammatory bowel disease (IBD) (e.g., Crohn's disease, ulcerative colitis, collagenous colitis, lymphocytic colitis, ischemic colitis, diversion colitis, Behcet's syndrome, indeterminate colitis) and inflammatory bowel syndrome (IBS)), lupus, multiple sclerosis, morphea, myasthenia gravis, myocardial ischemia, nephrotic syndrome, pemphigus vulgaris, pernicious aneemia, peptic ulcers, polymyositis, primary biliary cirrhosis, neuroinflammation associated with brain disorders (e.g., Parkinson's disease, Huntington's disease, and Alzheimer's disease), prostatitis, chronic inflammation associated with cranial radiation injury, pelvic inflammatory disease, reperfusion injury, regional enteritis, rheumatic fever, systemic lupus erythematosus, scleroderma, scleroderma, sarcoidosis, spondyloarthopathies, Sjogren's syndrome, thyroiditis, transplantation rejection, tendonitis, trauma or injury (e.g., frostbite, chemical irritants, toxins, scarring, burns, physical injury), vasculitis, vitiligo and Wegener's granulomatosis. In certain embodiments, the inflammatory disorder is selected from arthritis (e.g., rheumatoid arthritis), inflammatory bowel disease, inflammatory bowel syndrome, asthma, psoriasis, endometriosis, interstitial cystitis and prostatistis. In certain embodiments, the inflammatory condition is an acute inflammatory condition (e.g., for example, inflammation resulting from infection). In certain embodiments, the inflammatory condition is a chronic inflammatory condition (e.g., conditions resulting from asthma, arthritis and inflammatory bowel disease). In some embodiments, the compounds are useful in treating inflammation associated with trauma and non-inflammatory myalgia. In some embodiments, the compounds disclosed herein are useful in treating inflammation associated with cancer. The term “pain” includes, but is not limited to, neuropathic pain (e.g., peripheral neuropathic pain), central pain, deafferentiation pain, short-term pain, chronic pain (e.g., chronic nociceptive pain, and other forms of chronic pain such as post-operative pain, e.g., pain arising after hip, knee, or other replacement surgery), pre-operative pain, stimulus of nociceptive receptors (nociceptive pain), acute pain (including phantom and transient acute pain), noninflammatory pain, inflammatory pain, pain associated with cancer, wound pain, burn pain, post-operative pain, pain associated with medical procedures, pain resulting from pruritus, painful bladder syndrome, pain associated with premenstrual dysphoric disorder and/or premenstrual syndrome, dysmenorrhea, post-partum pain, pain associated with endometriosis, pain associated with chronic fatigue syndrome, pain associated with pre-term labor, pain associated with withdrawal symptoms from drug addiction, joint pain, arthritic pain (e.g., pain associated with crystalline arthritis, osteoarthritis, psoriatic arthritis, acute gouty arthritis, reactive arthritis, rheumatoid arthritis or Reiter's arthritis), lumbosacral pain, musculoskeletal pain, headache, migraine, muscle ache, lower back pain, neck pain, toothache, dental pain, maxillofacial pain, visceral pain and the like. In some embodiments, the post-operative pain or pain associated with medical procedures (iatrogenic pain) is associated with anorectal, pelvic floor, or urogynecologic procedures (vaginal/perineal approach) (e.g., colon resection, hemorrhoidectomy, vaginal hysterectomy), breast procedures (e.g., lumpectomy, mastectomy, reconstruction, reduction), dental surgery (e.g., third molar extraction), extremity trauma requiring surgery (e.g., amputation, open reduction, internal fixation), orthopedic surgery or procedures, joint replacement (e.g. total hip arthroplasty (THA), total knee arthroplasty (TKA)), laparoscopic abdominal procedures (e.g., appendectomy, bariatric surgery, cholecystectomy, colectomy, hysterectomy, prostatectomy), open abdominal procedures (e.g., appendectomy, cholecystectomy, colectomy, hysterectomy), laparoscopic or open abdominal wall procedures (e.g., femoral hernia, incisional hernia, inguinal hernia), obstetric procedures (e.g., cesarean delivery, vaginal delivery), oropharyngeal procedures (e.g., tonsillectomy), spine procedures (e.g., fusion in both adults and children, laminectomy), procedures related to sport-related injuries (e.g., ACL repair and reconstruction, joint arthroscopy, rotator cuff repair), or thoracic procedures (e.g., thoracoscopy, repair of pectus excavatum in children (Nuss procedure)). As disclosed herein, pain can comprise mixtures of various types of pain provided above and herein (e.g., nociceptive pain, inflammatory pain, neuropathic pain, etc.). In some embodiments, a particular pain can dominate. In other embodiments, the pain comprises two or more types of pains without one dominating. A skilled clinician can determine the dosage to achieve a therapeutically effective amount for a particular subject based on the pain. In certain embodiments, the pain is inflammatory pain. In certain embodiments, the pain (e.g., inflammatory pain) is associated with inflammation or an inflammatory condition. In certain embodiments, the pain is non-inflammatory pain. The types of non-inflammatory pain include, without limitation, peripheral neuropathic pain (e.g., pain caused by a lesion or dysfunction in the peripheral nervous system), central pain (e.g., pain caused by a lesion or dysfunction of the central nervous system), deafferentation pain (e.g., pain due to loss of sensory input to the central nervous system), chronic nociceptive pain (e.g., certain types of cancer pain), noxious stimulus of nociceptive receptors (e.g., pain felt in response to tissue damage or impending tissue damage), phantom pain (e.g., pain felt in a part of the body that no longer exists, such as a limb that has been amputated), pain felt by psychiatric subjects (e.g., pain where no physical cause exists), and wandering pain (e.g., wherein the pain repeatedly changes location in the body). In addition, there is pain associated with normally non-painful sensations such as “pins and needles” (paraesthesias and dysesthesias), increased sensitivity to touch (hyperesthesia), painful sensation following innocuous stimulation (dynamic, static or thermal allodynia), increased sensitivity to noxious stimuli (thermal, cold, mechanical hyperalgesia), continuing pain sensation after removal of the stimulation (hyperpathia) or an absence of or deficit in selective sensory pathways (hypoalgesia). In some embodiments, pain is measured through any clinically-validated pain assessment measurement. In some embodiments, pain is measured through the Pain Intensity Numerical Rating Scale. In some embodiments, pain is measured by use of a visual analogue scale (“VAS,” such as a 10 cm scale with 0 representing no pain and 10 representing maximal pain). The term “fever” refers to an elevated body temperature that is approximately 37.7° C. (99.9° F.) or greater for a human subject. In some embodiments, the fever is an infectious fever caused by or associated with an infectious cause, disorder or disease or a non-infectious fever not caused by or associated with an infectious cause, disorder or disease. Common causes of infectious fevers include, but are not limited to, upper and lower respiratory tract infections, gastrointestinal infections, urinary tract infection, and skin infections. Non-limiting pathogens associated with infectious diseases include viruses, bacteria, fungi, yeast, protozoans, nematodes, and other parasites. In some embodiments, a non-infectious fever is associated with an inflammatory disorder or disease, or cancer, or is caused by a drug, an immunization, heat exhaustion, sunburn, and the like. In some embodiments, the fever is associated with a viral infection (e.g., influenza, the common cold, COVID-19), dengue fever, or rheumatic fever. Unless otherwise required by context, singular terms shall include pluralities, and plural terms shall include the singular. The singular forms “a”, “an” and “the” include plural reference unless the context clearly dictates otherwise. The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.” When a range of values (“range”) is listed, it encompasses each value and sub-range within the range. A range is inclusive of the values at the two ends of the range unless otherwise provided. It will be understood that when a range is recited in the application, the ends of the range are specifically disclosed as if specifically recited. For example, a range of about 19% to about 99% specifically include a disclosure separately of 19% and separately of 99%. Other than in the examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term “about.” “About” and “approximately” shall generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, or more typically, within 5%, 4%, 3%, 2%, or 1% of a given value or range of values. DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS Disclosed herein are deuterated forms of etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, compositions and kits comprising the same, and methods of using the same. United States Patent Application Publication No. US 2009/0076087 A1 to Czarnik/Protia, LLC generically and prophetically discloses “deuterium-enriched etoricoxib.” However, the inventors unexpectedly discovered that certain deuterated forms of etoricoxib display improved properties, including improved pharmacokinetic properties. For example, certain deuterated forms of etoricoxib disclosed herein possess improved maximum serum concentration (Cmax), total systemic exposure (AUC), time of maximal plasma concentration (Tmax), and/or half-life (t1/2) that are distinct from those demonstrated by previously known etoricoxib compositions. Without wishing to be bound by theory, the inventors posit that these deuterated forms of etoricoxib allow for advantages such as reduced dosages, reduced dosage intervals, improved therapeutic windows, modified release, reduced formation of one or more metabolites of etoricoxib, and/or reduced side effects, as compared to previously known etoricoxib compositions. Compounds In one aspect, the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof; wherein: each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium; provided that:(1) at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium; and(2) the compound is not of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In another aspect, the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof; wherein: each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium; provided that:(1) at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium;(2) at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is hydrogen;(3) when each of R1, R2, R3, R8, R9, R10, R11, R12, R13, R14, and R15is hydrogen, at least one of R4, R5, R6, and R7is hydrogen;(4) when each of R1, R2, R3, R4, R5, R6, R7, R10, R11, R12, R13, R14, and R15is hydrogen, at least one of R8and R9is hydrogen; and(5) when each of R1, R2, R3, R4, R5, R6, R7, R8, and R9is hydrogen, at least one of R10, R11, R12, R13, R14, and R15is hydrogen. In some embodiments, the compound of Formula (I) is of Formula (I-A): or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, at least one of R4, R5, R6, and R7is hydrogen. In some embodiments, at least one of R8and R9is hydrogen. In some embodiments, when each of R13, R14, and R15is deuterium, at least one of R10, R11, and R12is hydrogen. In some embodiments, when each of R10, R11, and R12is deuterium, at least one of R13, R14, and R15is hydrogen. In some embodiments, at least one of R4, R5, R6, and R7is hydrogen, at least one of R8and R9is hydrogen, when each of R13, R14, and R15is deuterium, at least one of R10, R11, and R12is hydrogen, and when each of R10, R11, and R12is deuterium, at least one of R13, R14, and R15is hydrogen. In some embodiments, at least one of R1, R2, and R3is deuterium. In some embodiments, at least two of R1, R2, and R3are deuterium. In some embodiments, each of R1, R2, and R3is deuterium. In some embodiments, R1is deuterium. In some embodiments R2is deuterium. In some embodiments R3is deuterium. In some embodiments R1and R2are deuterium. In some embodiments, R1and R3are deuterium. In some embodiments, R2and R3are deuterium. In some embodiments, each of R1, R2, and R3is hydrogen. In some embodiments, at least one of R4, R5, R6, and R7is deuterium. In some embodiments, at least two of R4, R5, R6and R7are deuterium. In some embodiments, three of R4, R5, R6, and R7are deuterium. In some embodiments, R4is deuterium. In some embodiments, R5is deuterium. In some embodiments, R6is deuterium. In some embodiments, R7is deuterium. In some embodiments, R4and R5are deuterium. In some embodiments, R4and R6are deuterium. In some embodiments, R4and R7are deuterium. In some embodiments, R5and R6are deuterium. In some embodiments, R5and R7are deuterium. In some embodiments, R6and R7are deuterium. In some embodiments, R4, R5, and R6are deuterium. In some embodiments, R4, R5, and R7are deuterium. In some embodiments, R4, R6, and R7are deuterium. In some embodiments, R5, R6, and R7are deuterium. In some embodiments, each of R4, R5, R6, and R7is hydrogen. In some embodiments, one of R8and R9is deuterium. In some embodiments, R8is deuterium. In some embodiments, R9is deuterium. In some embodiments, each of R8and R9is hydrogen. In some embodiments, at least one of R10, R11, and R12is deuterium. In some embodiments, at least two of R10, R11, and R12are deuterium. In some embodiments, each of R10, R11, and R12is deuterium. In some embodiments, R10is deuterium. In some embodiments, R11is deuterium. In some embodiments, R12is deuterium. In some embodiments, R10and R11are deuterium. In some embodiments, R10and R12are deuterium. In some embodiments, R11and R12are deuterium. In some embodiments, each of R10, R11, and R12is hydrogen. In some embodiments, at least one of R13, R14, and R15is deuterium. In some embodiments, at least two of R13, R14, and R15are deuterium. In some embodiments, each of R13, R14, and R15is deuterium. In some embodiments, R13is deuterium. In some embodiments, R14is deuterium. In some embodiments, R15is deuterium. In some embodiments, R13and R14are deuterium. In some embodiments, R13and R15are deuterium. In some embodiments, R14and R15are deuterium. In some embodiments, each of R13, R14, and R15is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R11is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R12is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10and R11are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10and R12are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R11and R12are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10, R11, and R12are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R14is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R15is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13and R14are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13and R15are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R14and R15are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13, R14, and R15are hydrogen. In some embodiments, each of R1, R2, R3, R13, R14, and R15is deuterium. In some embodiments, each of R1, R2, R3, R4, R5, R6, R7, R13, R14, and R15is deuterium. In some embodiments, each of R4, R5, R6, R7, R13, R14, and R15is deuterium. In some embodiments, each of R8, R9, R13, R14, and R15is deuterium. In some embodiments, each of R1, R2, R3, R8, R9, R13, R14, and R15is deuterium. In some embodiments, the compound is not of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the compound is not of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the compound is not of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the compound is not of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the compound is not of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In another aspect, the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In another aspect, the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt thereof. In another aspect, the present disclosure provides a compound of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In another aspect, the present disclosure provides a compound of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In another aspect, the present disclosure provides a compound of the formula: or a pharmaceutically acceptable salt thereof. In some embodiments, the salt of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib is an oxalate, succinate, fumarate, besylate, hydrobromide, hydrochloride, glutamate, sulfamate, benzoate, cinnamate, salicylate, or tosylate salt. In some embodiments, the salt is an oxalate salt. In some embodiments, the salt is a succinate salt. In some embodiments, the salt is a fumarate salt. In some embodiments, the salt is a besylate salt. In some embodiments, the salt is a hydrobromide salt. In some embodiments, the salt is a hydrochloride salt. In some embodiments, the salt is a glutamate salt. In some embodiments, the salt is a sulfamate salt. In some embodiments, the salt is a benzoate salt. In some embodiments, the salt is a cinnamate salt. In some embodiments, the salt is a salicylate salt. In some embodiments, the salt is a tosylate salt. In some embodiments, the metabolite of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, comprises a hydroxyl group in place of a hydrogen or deuterium. In some embodiments, the metabolite substitutes a hydroxyl group for hydrogen or deuterium at R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, or R15. In some embodiments, the metabolite substitutes a hydroxyl group for hydrogen or deuterium at one or more of R13, R14, and R15. In some embodiments, the metabolite substitutes a hydroxyl group for one hydrogen or deuterium at R13, R14, and R15. In some embodiments, the metabolite substitutes a hydroxyl group for hydrogen or deuterium at two of R13, R14, and R15. In some embodiments, the metabolite substitutes a hydroxyl group for hydrogen or deuterium at each of R13, R14, and R15. In some embodiments, the metabolite of a compound of Formula (I) has the structure: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, achieves a higher geometric mean Cmaxplasma concentration following administration of a single dose of the compound, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or the pharmaceutical composition, to a population of patients compared to a control composition comprising non-isotopically enriched etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition comprising etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, and a pharmaceutically acceptable excipient. In some embodiments, the patients are healthy patients. In some embodiments, the geometric mean Cmaxplasma concentration is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 5% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 10% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 15% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 20% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 25% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 30% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 35% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 40% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 45% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 50% higher. In some embodiments, the compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, achieves a higher geometric mean plasma AUC following administration of a single dose of the compound, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or the pharmaceutical composition, to a population of patients compared to a control composition comprising non-isotopically enriched etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition comprising etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, and a pharmaceutically acceptable excipient. In some embodiments, the patients are healthy patients. In some embodiments, the geometric mean plasma AUC is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% higher. In some embodiments, the geometric mean plasma AUC is at least 5% higher. In some embodiments, the geometric mean plasma AUC is at least 10% higher. In some embodiments, the geometric mean plasma AUC is at least 15% higher. In some embodiments, the geometric mean plasma AUC is at least 20% higher. In some embodiments, the geometric mean plasma AUC is at least 25% higher. In some embodiments, the geometric mean plasma AUC is at least 30% higher. In some embodiments, the geometric mean plasma AUC is at least 35% higher. In some embodiments, the geometric mean plasma AUC is at least 40% higher. In some embodiments, the geometric mean plasma AUC is at least 45% higher. In some embodiments, the geometric mean plasma AUC is at least 50% higher. In some embodiments, the geometric mean plasma AUC is the geometric mean plasma AUC0-24. In some embodiments, the geometric mean plasma AUC is the geometric mean plasma AUC0-∞. In another aspect, the present disclosure provides a compound of Formula (II): or a salt, hydrate, solvate, polymorph, or co-crystal thereof; wherein each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium, provided that at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium. In another aspect, the present disclosure provides a compound of Formula (II): or a salt thereof; wherein each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium, provided that at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium. In another aspect, the present disclosure provides a compound of the formula: or a salt, hydrate, solvate, polymorph, or co-crystal thereof. In another aspect, the present disclosure provides a compound of the formula: or a salt thereof. In some embodiments, at least one of R4, R5, R6, and R7is hydrogen. In some embodiments, at least one of R8and R9is hydrogen. In some embodiments, when each of R13, R14, and R15is deuterium, at least one of R10, R11, and R12is hydrogen. In some embodiments, when each of R10, R11, and R12is deuterium, at least one of R13, R14, and R15is hydrogen. In some embodiments, at least one of R4, R5, R6, and R7is hydrogen, at least one of R8and R9is hydrogen, when each of R13, R14, and R15is deuterium, at least one of R10, R11, and R12is hydrogen, and when each of R10, R11, and R12is deuterium, at least one of R13, R14, and R15is hydrogen. In some embodiments, at least one of R1, R2, and R3is deuterium. In some embodiments, at least two of R1, R2, and R3are deuterium. In some embodiments, each of R1, R2, and R3is deuterium. In some embodiments, R1is deuterium. In some embodiments R2is deuterium. In some embodiments R3is deuterium. In some embodiments R1and R2are deuterium. In some embodiments, R1and R3are deuterium. In some embodiments, R2and R3are deuterium. In some embodiments, each of R1, R2, and R3is hydrogen. In some embodiments, at least one of R4, R5, R6, and R7is deuterium. In some embodiments, at least two of R4, R5, R6and R7are deuterium. In some embodiments, three of R4, R5, R6, and R7are deuterium. In some embodiments, R4is deuterium. In some embodiments, R5is deuterium. In some embodiments, R6is deuterium. In some embodiments, R7is deuterium. In some embodiments, R4and R5are deuterium. In some embodiments, R4and R6are deuterium. In some embodiments, R4and R7are deuterium. In some embodiments, R5and R6are deuterium. In some embodiments, R5and R7are deuterium. In some embodiments, R6and R7are deuterium. In some embodiments, R4, R5, and R6are deuterium. In some embodiments, R4, R5, and R7are deuterium. In some embodiments, R4, R6, and R7are deuterium. In some embodiments, R5, R6, and R7are deuterium. In some embodiments, each of R4, R5, R6, and R7is hydrogen. In some embodiments, one of R8and R9is deuterium. In some embodiments, R8is deuterium. In some embodiments, R9is deuterium. In some embodiments, each of R8and R9is hydrogen. In some embodiments, at least one of R10, R11, and R12is deuterium. In some embodiments, at least two of R10, R11, and R12are deuterium. In some embodiments, each of R10, R11, and R12is deuterium. In some embodiments, R10is deuterium. In some embodiments, R11is deuterium. In some embodiments, R12is deuterium. In some embodiments, R10and R11are deuterium. In some embodiments, R10and R12are deuterium. In some embodiments, R11and R12are deuterium. In some embodiments, each of R10, R11, and R12is hydrogen. In some embodiments, at least one of R13, R14, and R15is deuterium. In some embodiments, at least two of R13, R14, and R15are deuterium. In some embodiments, each of R13, R14, and R15is deuterium. In some embodiments, R13is deuterium. In some embodiments, R14is deuterium. In some embodiments, R15is deuterium. In some embodiments, R13and R14are deuterium. In some embodiments, R13and R15are deuterium. In some embodiments, R14and R15are deuterium. In some embodiments, each of R13, R14, and R15is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R11is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R12is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10and R11are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10and R12are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R11and R12are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10, R11, and R12are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R14is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R15is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13and R14are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13and R15are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R14and R15are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13, R14, and R15are hydrogen. In some embodiments, each of R1, R2, R3, R13, R14, and R15is deuterium. In some embodiments, each of R1, R2, R3, R4, R5, R6, R7, R13, R14, and R15is deuterium. In some embodiments, each of R4, R5, R6, R7, R13, R14, and R15is deuterium. In some embodiments, each of R8, R9, R13, R14, and R15is deuterium. In some embodiments, each of R1, R2, R3, R8, R9, R13, R14, and R15is deuterium. In another aspect, the present disclosure provides a compound of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In another aspect, the present disclosure provides a compound of the formula: or a pharmaceutically acceptable salt thereof. In another aspect, the present disclosure provides a compound of Formula (III): or a salt, hydrate, solvate, polymorph, or co-crystal thereof; wherein each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium, provided that at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium. In another aspect, the present disclosure provides a compound of Formula (III): or a salt thereof; wherein each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium, provided that at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium. In some embodiments, at least one of R4, R5, R6, and R7is hydrogen. In some embodiments, at least one of R8and R9is hydrogen. In some embodiments, when each of R13, R14, and R15is deuterium, at least one of R10, R11, and R12is hydrogen. In some embodiments, when each of R10, R11, and R12is deuterium, at least one of R13, R14, and R15is hydrogen. In some embodiments, at least one of R4, R5, R6, and R7is hydrogen, at least one of R8and R9is hydrogen, when each of R13, R14, and R15is deuterium, at least one of R10, R11, and R12is hydrogen, and when each of R10, R11, and R12is deuterium, at least one of R13, R14, and R15is hydrogen. In some embodiments, at least one of R1, R2, and R3is deuterium. In some embodiments, at least two of R1, R2, and R3are deuterium. In some embodiments, each of R1, R2, and R3is deuterium. In some embodiments, R1is deuterium. In some embodiments R2is deuterium. In some embodiments R3is deuterium. In some embodiments R1and R2are deuterium. In some embodiments, R1and R3are deuterium. In some embodiments, R2and R3are deuterium. In some embodiments, each of R1, R2, and R3is hydrogen. In some embodiments, at least one of R4, R5, R6, and R7is deuterium. In some embodiments, at least two of R4, R5, R6and R7are deuterium. In some embodiments, three of R4, R5, R6, and R7are deuterium. In some embodiments, R4is deuterium. In some embodiments, R5is deuterium. In some embodiments, R6is deuterium. In some embodiments, R7is deuterium. In some embodiments, R4and R5are deuterium. In some embodiments, R4and R6are deuterium. In some embodiments, R4and R7are deuterium. In some embodiments, R5and R6are deuterium. In some embodiments, R5and R7are deuterium. In some embodiments, R6and R7are deuterium. In some embodiments, R4, R5, and R6are deuterium. In some embodiments, R4, R5, and R7are deuterium. In some embodiments, R4, R6, and R7are deuterium. In some embodiments, R5, R6, and R7are deuterium. In some embodiments, each of R4, R5, R6, and R7is hydrogen. In some embodiments, one of R8and R9is deuterium. In some embodiments, R8is deuterium. In some embodiments, R9is deuterium. In some embodiments, each of R8and R9is hydrogen. In some embodiments, at least one of R10, R11, and R12is deuterium. In some embodiments, at least two of R10, R11, and R12are deuterium. In some embodiments, each of R10, R11, and R12is deuterium. In some embodiments, R10is deuterium. In some embodiments, R11is deuterium. In some embodiments, R12is deuterium. In some embodiments, R10and R11are deuterium. In some embodiments, R10and R12are deuterium. In some embodiments, R11and R12are deuterium. In some embodiments, each of R10, R11, and R12is hydrogen. In some embodiments, at least one of R13, R14, and R15is deuterium. In some embodiments, at least two of R13, R14, and R15are deuterium. In some embodiments, each of R13, R14, and R15is deuterium. In some embodiments, R13is deuterium. In some embodiments, R14is deuterium. In some embodiments, R15is deuterium. In some embodiments, R13and R14are deuterium. In some embodiments, R13and R15are deuterium. In some embodiments, R14and R15are deuterium. In some embodiments, each of R13, R14, and R15is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R11is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R12is hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10and R11are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10and R12are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R11and R12are hydrogen. In some embodiments, each of R13, R14, and R15is deuterium, and R10, R11, and R12are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R14is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R15is hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13and R14are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13and R15are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R14and R15are hydrogen. In some embodiments, each of R10, R11, and R12is deuterium, and R13, R14, and R15are hydrogen. In some embodiments, each of R1, R2, R3, R13, R14, and R15is deuterium. In some embodiments, each of R1, R2, R3, R4, R5, R6, R7, R13, R14, and R15is deuterium. In some embodiments, each of R4, R5, R6, R7, R13, R14, and R15is deuterium. In some embodiments, each of R8, R9, R13, R14, and R15is deuterium. In some embodiments, each of R1, R2, R3, R8, R9, R13, R14, and R15is deuterium. In another aspect, the present disclosure provides a compound of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In another aspect, the present disclosure provides a compound of the formula: or a pharmaceutically acceptable salt thereof. Pharmaceutical Compositions, Kits, and Administration In another aspect, the present disclosure provides a pharmaceutical composition comprising a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof; and a pharmaceutically acceptable excipient. In another aspect, the present disclosure provides a pharmaceutical composition comprising a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof; and a pharmaceutically acceptable excipient. In another aspect, the present disclosure provides a pharmaceutical composition comprising a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable excipient. In another aspect, the present disclosure provides a pharmaceutical composition comprising a compound of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof; and a pharmaceutically acceptable excipient. In another aspect, the present disclosure provides a pharmaceutical composition comprising a compound of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof; and a pharmaceutically acceptable excipient. In another aspect, the present disclosure provides a pharmaceutical composition comprising a compound of the formula: or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable excipient. In another aspect, the present disclosure provides a kit comprising a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof; or a pharmaceutical composition thereof; and instructions for using the compound, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or the pharmaceutical composition. In another aspect, the present disclosure provides a kit comprising a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof; or a pharmaceutical composition thereof; and instructions for using the compound, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or the pharmaceutical composition. In another aspect, the present disclosure provides a kit comprising a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt thereof; or a pharmaceutical composition thereof; and instructions for using the compound, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition. In another aspect, the present disclosure provides a kit comprising a compound of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof; or a pharmaceutical composition thereof; and instructions for using the compound, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or the pharmaceutical composition. In another aspect, the present disclosure provides a kit comprising a compound of the formula: or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof; or a pharmaceutical composition thereof; and instructions for using the compound, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or the pharmaceutical composition. In another aspect, the present disclosure provides a kit comprising a compound of the formula: or a pharmaceutically acceptable salt thereof; or a pharmaceutical composition thereof; and instructions for using the compound, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition. In some embodiments, the pharmaceutical composition is an oral dosage form. In some embodiments, the pharmaceutical composition is a parenteral, topical, buccal, ophthalmic, rectal, transdermal, or vaginal dosage form. In some embodiments, the pharmaceutical composition is a parenteral dosage form. In some embodiments, the pharmaceutical composition is a topical dosage form. In some embodiments the pharmaceutical composition is a buccal dosage form. In some embodiments, the pharmaceutical composition is an ophthalmic dosage form. In some embodiments, the pharmaceutical composition is a rectal dosage form. In some embodiments, the pharmaceutical composition is a transdermal dosage form. In some embodiments, the pharmaceutical composition is a vaginal dosage form. In some embodiments, the pharmaceutical composition is an injectable dosage form. In some embodiments, the injectable dosage form is an intravenous dosage form. In some embodiments, the pharmaceutical composition is a solid dosage formulation. In some embodiments, the solid dosage formulation is a tablet, capsule, granule, powder, sachet, or chewable dosage form. In some embodiments, the pharmaceutical composition is a liquid dosage formulation. In some embodiments, the pharmaceutical composition is a solution. In some embodiments, pharmaceutical composition is a suspension. In some embodiments, the pharmaceutical composition is a syrup. In some embodiments, the pharmaceutical composition achieves a higher geometric mean Cmaxplasma concentration following administration of a single dose of the compound, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or the pharmaceutical composition, to a population of patients compared to a control composition comprising non-isotopically enriched etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition comprising etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, and a pharmaceutically acceptable excipient. In some embodiments, the patients are healthy patients. In some embodiments, the geometric mean Cmaxplasma concentration is at least 5% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 10% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 15% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 20% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 25% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 30% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 35% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 40% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 45% higher. In some embodiments, the geometric mean Cmaxplasma concentration is at least 50% higher. In some embodiments, the pharmaceutical composition achieves a higher geometric mean plasma AUC following administration of a single dose of the compound, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or the pharmaceutical composition, to a population of patients compared to a control composition comprising non-isotopically etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition comprising etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, and a pharmaceutically acceptable excipient. In some embodiments, the patients are healthy patients. In some embodiments, the geometric mean plasma AUC is at least 5% higher. In some embodiments, the geometric mean plasma AUC is at least 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% higher. In some embodiments, the geometric mean plasma AUC is at least 10% higher. In some embodiments, the geometric mean plasma AUC is at least 15% higher. In some embodiments, the geometric mean plasma AUC is at least 20% higher. In some embodiments, the geometric mean plasma AUC is at least 25% higher. In some embodiments, the geometric mean plasma AUC is at least 30% higher. In some embodiments, the geometric mean plasma AUC is at least 35% higher. In some embodiments, the geometric mean plasma AUC is at least 40% higher. In some embodiments, the geometric mean plasma AUC is at least 45% higher. In some embodiments, the geometric mean plasma AUC is at least 50% higher. In some embodiments, the geometric mean plasma AUC is the geometric mean plasma AUC0-24. In some embodiments, the geometric mean plasma AUC is the geometric mean plasma AUC0-∞. Pharmaceutical compositions disclosed herein can be prepared by any method known in the art of pharmaceutics. In general, such preparatory methods include bringing the compound disclosed herein (i.e., the “active ingredient”) into association with a carrier or excipient, and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping, and/or packaging the product into a desired single- or multi-dose unit. In some embodiments, pharmaceutical compositions are prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. A “unit dose” is a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage, such as one-half or one-third of such a dosage. Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition disclosed herein will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. In some embodiments, the composition comprises between 0.1% and 100% (w/w) active ingredient. Pharmaceutically acceptable excipients used in the manufacture of the disclosed pharmaceutical compositions include inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. In some embodiments, excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and perfuming agents are present in the composition. Exemplary diluents include calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, and mixtures thereof. Exemplary granulating and/or dispersing agents include potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose, and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, and mixtures thereof. Exemplary surface active agents and/or emulsifiers include natural emulsifiers (e.g., acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g., bentonite (aluminum silicate) and Veegum (magnesium aluminum silicate)), long chain amino acid derivatives, high molecular weight alcohols (e.g., stearyl alcohol, cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g., carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g., carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g., polyoxyethylene sorbitan monolaurate (Tween® 20), polyoxyethylene sorbitan (Tween® 60), polyoxyethylene sorbitan monooleate (Tween® 80), sorbitan monopalmitate (Span® 40), sorbitan monostearate (Span® 60), sorbitan tristearate (Span® 65), glyceryl monooleate, sorbitan monooleate (Span® 80), polyoxyethylene esters (e.g., polyoxyethylene monostearate (Myrj® 45), polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and Solutol®), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g., Cremophor®), polyoxyethylene ethers, (e.g., polyoxyethylene lauryl ether (Brij® 30)), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic® F-68, poloxamer P-188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, and/or mixtures thereof. Exemplary binding agents include starch (e.g., cornstarch and starch paste), gelatin, sugars (e.g., sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol, etc.), natural and synthetic gums (e.g., acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (Veegum®), and larch arabogalactan), alginates, polyethylene oxide, polyethylene glycol, inorganic calcium salts, silicic acid, polymethacrylates, waxes, water, alcohol, and/or mixtures thereof. Exemplary preservatives include antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, antiprotozoan preservatives, alcohol preservatives, acidic preservatives, and other preservatives. In certain embodiments, the preservative is an antioxidant. In other embodiments, the preservative is a chelating agent. Exemplary antioxidants include alpha tocopherol, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and sodium sulfite. Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA) and salts and hydrates thereof (e.g., sodium edetate, disodium edetate, trisodium edetate, calcium disodium edetate, dipotassium edetate, and the like), citric acid and salts and hydrates thereof (e.g., citric acid monohydrate), fumaric acid and salts and hydrates thereof, malic acid and salts and hydrates thereof, phosphoric acid and salts and hydrates thereof, and tartaric acid and salts and hydrates thereof. Exemplary antimicrobial preservatives include benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and thimerosal. Exemplary antifungal preservatives include butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and sorbic acid. Exemplary alcohol preservatives include ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and phenylethyl alcohol. Exemplary acidic preservatives include vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and phytic acid. Other preservatives include tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, Glydant® Plus, Phenonip®, methylparaben, Germall® 115, Germaben® II, Neolone®, Kathon®, and Euxyl®. Exemplary buffering agents include citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, D-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline, Ringer's solution, ethyl alcohol, and mixtures thereof. Exemplary lubricating agents include magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, and mixtures thereof. Exemplary natural oils include almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean, sunflower, tea tree, thistle, tsubaki, vetiver, walnut, and wheat germ oils. Exemplary synthetic oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and mixtures thereof. Liquid dosage forms for oral and parenteral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In some embodiments, in addition to the active ingredients, the liquid dosage forms comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (e.g., cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. In certain embodiments for parenteral administration, the conjugates disclosed herein are mixed with solubilizing agents such as Cremophor®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and mixtures thereof. In some embodiments, injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions are formulated according to the known art using suitable dispersing or wetting agents and suspending agents. In some embodiments, the sterile injectable preparation is a sterile injectable solution, suspension, or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. In some embodiments, the acceptable vehicles and solvents that are employed include water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. In some embodiments, any bland fixed oil is employed for this purpose, including synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. In some embodiments, the injectable formulations are sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions. In some embodiments, such sterilized injectable formulations are dissolved or dispersed in sterile water or other sterile injectable medium prior to use. In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. In some embodiments, this is accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. In some embodiments, the rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, depends upon crystal size and crystalline form. Alternatively, in some embodiments, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Compositions for rectal or vaginal administration are typically suppositories. In some embodiments, compositions for rectal or vaginal administrations are suppositories prepared by mixing the compounds disclosed herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol, or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or (a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, (c) humectants such as glycerol, (d) disintegrating agents such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, (e) solution retarding agents such as paraffin, (f) absorption accelerators such as quaternary ammonium compounds, (g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, (h) absorbents such as kaolin and bentonite clay, and (i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In some embodiments, in the case of capsules, tablets, and pills, the dosage form includes a buffering agent. In some embodiments, solid compositions of a similar type are employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. In some embodiments, the solid dosage forms of tablets, dragees, capsules, pills, and granules are prepared with coatings and shells such as enteric coatings and other coatings well known in the art of pharmacology. In some embodiments, the solid dosage forms optionally comprise opacifying agents. In some embodiments, the solid dosage forms are of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. In some embodiments, encapsulating compositions include polymeric substances and waxes. In some embodiments, solid compositions of a similar type are employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. In some embodiments, the active ingredient is in a micro-encapsulated form with one or more excipients as noted above. In some embodiments, the solid dosage forms of tablets, dragees, capsules, pills, and granules are prepared with coatings and shells such as enteric coatings, release controlling coatings, and other coatings well known in the pharmaceutical formulating art. In some embodiments, in such solid dosage forms the active ingredient is admixed with at least one inert diluent such as sucrose, lactose, or starch. In some embodiments, such dosage forms comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In some embodiments, in the case of capsules, tablets and pills, the dosage forms comprise buffering agents. In some embodiments, the dosage forms optionally comprise opacifying agents and are of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. In some embodiments, the encapsulating agents include polymeric substances and waxes. In some embodiments, dosage forms for topical and/or transdermal administration of a compound disclosed herein include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants, and/or patches. Generally, the active ingredient is admixed under sterile conditions with a pharmaceutically acceptable carrier or excipient and/or any needed preservatives and/or buffers as required. Additionally, the present disclosure contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of an active ingredient to the body. In some embodiments, such dosage forms are prepared, for example, by dissolving and/or dispensing the active ingredient in the proper medium. In some embodiments, alternatively or additionally, the rate is controlled by either providing a rate controlling membrane and/or by dispersing the active ingredient in a polymer matrix and/or gel. Suitable devices for use in delivering intradermal pharmaceutical compositions disclosed herein include short needle devices. In some embodiments, intradermal compositions are administered by devices which limit the effective penetration length of a needle into the skin. In some embodiments, alternatively or additionally, conventional syringes are used in the classical mantoux method of intradermal administration. Jet injection devices which deliver liquid formulations to the dermis via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable. Ballistic powder/particle delivery devices which use compressed gas to accelerate the compound in powder form through the outer layers of the skin to the dermis are suitable. Formulations suitable for topical administration include, but are not limited to, liquid and/or semi-liquid preparations such as liniments, lotions, oil-in-water and/or water-in-oil emulsions such as creams, ointments, and/or pastes, and/or solutions and/or suspensions. Topically administrable formulations may, for example, comprise from about 1% to about 10% (w/w) active ingredient. In some embodiments, the concentration of the active ingredient is as high as the solubility limit of the active ingredient in the solvent. In some embodiments, formulations for topical administration further comprise one or more of the additional ingredients disclosed herein. In some embodiments, a pharmaceutical composition disclosed herein is prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity. In some embodiments, such a formulation comprises dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 to about 7 nanometers, or from about 1 to about 6 nanometers. Such compositions are conveniently in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant is directed to disperse the powder and/or using a self-propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container. Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nanometers and at least 95% of the particles by number have a diameter less than 7 nanometers. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nanometer and at least 90% of the particles by number have a diameter less than 6 nanometers. In some embodiments, dry powder compositions include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form. Low boiling propellants generally include liquid propellants having a boiling point of below 65° F. at atmospheric pressure. In some embodiments, the propellant constitutes 50 to 99.9% (w/w) of the composition, and the active ingredient constitutes 0.1 to 20% (w/w) of the composition. In some embodiments, the propellant further comprises additional ingredients such as a liquid non-ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient). In some embodiments, pharmaceutical compositions disclosed herein formulated for pulmonary delivery provide the active ingredient in the form of droplets of a solution and/or suspension. In some embodiments, such formulations are prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising the active ingredient, and may conveniently be administered using any nebulization and/or atomization device. In some embodiments, such formulations further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as methylhydroxybenzoate. In some embodiments, the droplets provided by this route of administration have an average diameter in the range from about 0.1 to about 200 nanometers. Formulations disclosed herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition disclosed herein. Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 to 500 micrometers. Such a formulation is administered by rapid inhalation through the nasal passage from a container of the powder held close to the nares. For example, in some embodiments, formulations for nasal administration comprise from about as little as 0.1% (w/w) to as much as 100% (w/w) of the active ingredient. In some embodiments, formulations for nasal administration further comprise one or more of the additional ingredients disclosed herein. In some embodiments, a pharmaceutical composition disclosed herein is prepared, packaged, and/or sold in a formulation for buccal administration. For example, in some embodiments, such formulations are in the form of tablets and/or lozenges made using conventional methods, and contain, for example, 0.1 to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients disclosed herein. Alternately, in some embodiments, formulations for buccal administration comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising the active ingredient. In some embodiments, such powdered, aerosolized, and/or aerosolized formulations, when dispersed, have an average particle and/or droplet size in the range from about 0.1 to about 200 nanometers, and further comprise one or more of the additional ingredients disclosed herein. In some embodiments, a pharmaceutical composition disclosed herein is prepared, packaged, and/or sold in a formulation for ophthalmic administration. Such formulations may, for example, be in the form of eye drops including, for example, a 0.1-1.0% (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid carrier or excipient. In some embodiments, such drops further comprise buffering agents, salts, and/or one or more other of the additional ingredients disclosed herein. Other opthalmically-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a liposomal preparation. Ear drops and/or eye drops are also contemplated as being within the scope of this disclosure. Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with ordinary experimentation. Compounds provided herein are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions disclosed herein will be decided by a physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the disease being treated and the severity of the disorder; the activity of the specific active ingredient employed; the specific composition employed; the age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific active ingredient employed; the duration of the treatment; drugs used in combination or coincidental with the specific active ingredient employed; and like factors well known in the medical arts. In some embodiments, the compounds and compositions disclosed herein are administered by any route, including enteral (e.g., oral), parenteral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal, buccal, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol. Specifically contemplated routes are oral administration, intravenous administration (e.g., systemic intravenous injection), regional administration via blood and/or lymph supply, and/or direct administration to an affected site. In general, the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), and/or the condition of the subject (e.g., whether the subject is able to tolerate oral administration). The exact amount of a compound required to achieve an effective amount will vary from subject to subject, depending, for example, on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular compound, mode of administration, and the like. In some embodiments, an effective amount is included in a single dose (e.g., single oral dose) or multiple doses (e.g., multiple oral doses). In certain embodiments, when multiple doses are administered to a subject or applied to a tissue or cell, any two doses of the multiple doses include different or substantially the same amounts of a compound disclosed herein. In certain embodiments, when multiple doses are administered to a subject or applied to a tissue or cell, the frequency of administering the multiple doses to the subject or applying the multiple doses to the tissue or cell is three doses a day, two doses a day, one dose a day, one dose every other day, one dose every third day, one dose every week, one dose every two weeks, one dose every three weeks, or one dose every four weeks. In certain embodiments, the frequency of administering the multiple doses to the subject or applying the multiple doses to the tissue or cell is one dose per day. In certain embodiments, the frequency of administering the multiple doses to the subject or applying the multiple doses to the tissue or cell is two doses per day. In certain embodiments, the frequency of administering the multiple doses to the subject or applying the multiple doses to the tissue or cell is three doses per day. In certain embodiments, when multiple doses are administered to a subject or applied to a tissue or cell, the duration between the first dose and last dose of the multiple doses is one day, two days, four days, one week, two weeks, three weeks, one month, two months, three months, four months, six months, nine months, one year, two years, three years, four years, five years, seven years, ten years, fifteen years, twenty years, or the lifetime of the subject, tissue, or cell. In certain embodiments, the duration between the first dose and last dose of the multiple doses is three months, six months, or one year. In certain embodiments, the duration between the first dose and last dose of the multiple doses is the lifetime of the subject, tissue, or cell. In some embodiments, a first dose (loading dose) of the compound is provided, followed by a second, lower dose (maintenance dose) of the compound. In some embodiments, the maintenance dose is administered for up to 3 days, up to 5 days, up to 7 days, up to 10 days, up to 14 days, or chronically thereafter. Dose ranges as disclosed herein provide guidance for the administration of the disclosed pharmaceutical compositions to an adult. In some embodiments, the amount to be administered to, for example, a child or an adolescent is determined by a medical practitioner or person skilled in the art and is lower or the same as that administered to an adult. In some embodiments, the pharmaceutical composition comprises about 0.1 mg to about 1,000 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg to about 500 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg to about 1,000 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg to about 500 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg to about 300 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 5 mg to about 250 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg to about 200 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 5 mg to about 150 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg to about 140 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 5 mg to about 130 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg to about 120 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 5 mg, 7.5 mg, 10 mg, 12.5 mg, 15 mg, 17.5 mg, 20 mg, 22.5 mg, 25 mg, 27.5 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 100 mg, 110 mg, 120 mg, 140 mg, 160 mg, 180 mg, 200 mg, 220 mg, 240 mg, 260 mg, 280 mg, or 300 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 7.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 10 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 12.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 15 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 17.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 20 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 22.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 25 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 27.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 30 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 35 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 40 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 45 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 50 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 55 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 60 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 65 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 70 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 75 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 80 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 85 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 90 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 100 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 110 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 120 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 140 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 160 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 180 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 200 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 220 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 240 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 260 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 280 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 300 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg to about 1,000 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg to about 500 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg to about 1,000 mg of a compound of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg to about 500 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg to about 300 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 5 mg to about 250 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg to about 200 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 5 mg to about 150 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg to about 140 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 5 mg to about 130 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg to about 120 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 5 mg, 7.5 mg, 10 mg, 12.5 mg, 15 mg, 17.5 mg, 20 mg, 22.5 mg, 25 mg, 27.5 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 100 mg, 110 mg, 120 mg, 140 mg, 160 mg, 180 mg, 200 mg, 220 mg, 240 mg, 260 mg, 280 mg, or 300 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 7.5 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 10 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 12.5 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 15 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 17.5 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 20 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 22.5 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 25 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 27.5 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 30 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 35 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 40 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 45 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 50 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 55 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 60 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 65 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 70 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 75 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 80 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 85 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 90 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 100 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 110 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 120 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 140 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 160 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 180 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 200 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 220 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 240 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 260 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 280 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 300 mg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/mL to about 25 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/mL to about 10 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/mL to about 25 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/mL to about 10 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/mL to about 2 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/mL to about 5 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg/mL to about 10 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 10 mg/mL to about 15 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 15 mg/mL to about 20 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 20 mg/mL to about 25 mg/mL of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/mL, 0.5 mg/mL, 1 mg/mL, 2 mg/mL, 5 mg/mL, 7.5 mg/mL, 10 mg/mL, 12.5 mg/mL, 15 mg/mL, 17.5 mg/mL, 20 mg/mL, 22.5 mg/mL, or 25 mg/mL. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/mL. In some embodiments, the pharmaceutical composition comprises about 0.5 mg/mL. In some embodiments, the pharmaceutical composition comprises about 1 mg/mL. In some embodiments, the pharmaceutical composition comprises about 2 mg/mL. In some embodiments, the pharmaceutical composition comprises about 5 mg/mL. In some embodiments, the pharmaceutical composition comprises about 7.5 mg/mL. In some embodiments, the pharmaceutical composition comprises about 10 mg/mL. In some embodiments, the pharmaceutical composition comprises about 12.5 mg/mL. In some embodiments, the pharmaceutical composition comprises about 15 mg/mL. In some embodiments, the pharmaceutical composition comprises about 17.5 mg/mL. In some embodiments, the pharmaceutical composition comprises about 20 mg/mL. In some embodiments, the pharmaceutical composition comprises about 22.5 mg/mL. In some embodiments, the pharmaceutical composition comprises about 25 mg/mL. In certain embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 25 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 20 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 15 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 10 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 5 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 25 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 20 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 15 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 10 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 5 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.1 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, about 10 mg/kg, about 11 mg/kg, about 12 mg/kg, about 13 mg/kg, about 14 mg/kg, about 15 mg/kg, about 16 mg/kg, about 17 mg/kg, about 18 mg/kg, about 19 mg/kg, about 20 mg/kg, or about 25 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.5 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 2 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 3 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 4 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 6 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 7 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 8 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 9 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 10 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 11 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 12 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 13 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 14 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 15 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 16 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 17 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 18 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 19 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 20 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 25 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 25 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 20 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 15 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 10 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.1 mg/kg to about 5 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 25 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 20 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 15 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 10 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/kg to about 5 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.1 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, about 10 mg/kg, about 11 mg/kg, about 12 mg/kg, about 13 mg/kg, about 14 mg/kg, about 15 mg/kg, about 16 mg/kg, about 17 mg/kg, about 18 mg/kg, about 19 mg/kg, about 20 mg/kg, or about 25 mg/kg of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 0.1 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 0.5 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 1 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 2 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 3 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 4 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 5 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 6 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 7 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 8 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 9 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 10 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 11 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 12 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 13 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 14 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 15 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 16 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 17 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 18 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 19 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the pharmaceutical composition comprises about 20 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises about 25 mg/kg, of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the pharmaceutical composition comprises d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, wherein d3-etoricoxib has an isotopic purity of at least 50.0%, 60.0%, 70.0%, 75.0%, 80.0%, 85.0%, 90.0%, 95.0%, 97.0%, 98.0%, 99.0%, 99.5%, 99.7%, 99.8%, or 99.9%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 50.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 60.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 70.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 75.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 80.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 85.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 90.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 95.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 97.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 98.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 99.0%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 99.5%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 99.7%. In some embodiments, d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, has an isotopic purity of at least 99.9%. In some embodiments, the pharmaceutical composition comprises a compound that is isotopically enriched with deuterium at R13, R14, and R15by at least 50.0%, at least 60.0%, at least 70.0%, at least 75.0%, at least 80.0%, at least 85.0%, at least 90.0%, at least 94.0%, at least 95.0%, at least 96.0%, at least 97.0%, at least 98.0%, at least 99.0%, at least 99.5%, at least 99.7%, at least 99.8%, at least 99.9%, or at least 100%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 50.0%. In some embodiments, the compound is isotopically enriched at with deuterium R13, R14, and R15by at least 60.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 70.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 80.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 90.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 94.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 95.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 96.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 97.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 98.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 99.0%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 99.5%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 99.7%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 99.8%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 99.9%. In some embodiments, the compound is isotopically enriched with deuterium at R13, R14, and R15by at least 100%. In some embodiments, a compound or composition, as disclosed herein, is administered in combination with one or more additional pharmaceutical agents (e.g., therapeutically and/or prophylactically active agents). In some embodiments, the compounds or compositions are administered in combination with additional pharmaceutical agents that improve their activity (e.g., activity (e.g., potency and/or efficacy) in treating a disease in a subject in need thereof, in preventing a disease in a subject in need thereof, in reducing the risk to develop a disease in a subject in need thereof, and/or in inhibiting the activity of a protein kinase in a subject or cell), improve bioavailability, improve safety, reduce drug resistance, reduce and/or modify metabolism, inhibit excretion, and/or modify distribution in a subject or cell. It will also be appreciated that the therapy employed may achieve a desired effect for the same disorder, and/or it may achieve different effects. In certain embodiments, a pharmaceutical composition disclosed herein including a compound disclosed herein and an additional pharmaceutical agent shows a synergistic effect that is absent in a pharmaceutical composition including one of the compound and the additional pharmaceutical agent, but not both. In some embodiments, the additional pharmaceutical agent achieves a desired effect for the same disorder. In some embodiments, the additional pharmaceutical agent achieves different effects. In some embodiments, the pharmaceutical composition further comprises one or more additional agents. In some embodiments, the compound or composition is administered concurrently with, prior to, or subsequent to one or more additional pharmaceutical agents. For example, the one or more additional pharmaceutical agents are useful as combination therapies. Pharmaceutical agents include therapeutically active agents. Pharmaceutical agents also include prophylactically active agents. Pharmaceutical agents include small organic molecules such as drug compounds (e.g., compounds approved for human or veterinary use by the U.S. Food and Drug Administration as provided in the Code of Federal Regulations (CFR)), peptides, proteins, carbohydrates, monosaccharides, oligosaccharides, polysaccharides, nucleoproteins, mucoproteins, lipoproteins, synthetic polypeptides or proteins, small molecules linked to proteins, glycoproteins, steroids, nucleic acids, DNAs, RNAs, nucleotides, nucleosides, oligonucleotides, antisense oligonucleotides, lipids, hormones, vitamins, and cells. The additional pharmaceutical agents include, but are not limited to, anti-proliferative agents, anti-cancer agents, anti-angiogenesis agents, steroidal or non-steroidal anti-inflammatory agents, immunosuppressants, anti-bacterial agents, anti-viral agents, cardiovascular agents, cholesterol-lowering agents, anti-diabetic agents, anti-allergic agents, contraceptive agents, pain-relieving agents, anesthetics, anti-coagulants, inhibitors of an enzyme, steroidal agents, steroidal or antihistamine, antigens, vaccines, antibodies, decongestant, sedatives, opioids, analgesics, anti-pyretics, hormones, and prostaglandins. In some embodiments, each additional pharmaceutical agent is administered at a dose and/or on a time schedule determined for that pharmaceutical agent. In some embodiments, the additional pharmaceutical agents are administered together with each other and/or with the compound or composition disclosed herein in a single dose or composition or administered separately in different doses or compositions. The particular combination to employ in a regimen will take into account compatibility of the compound disclosed herein with the additional pharmaceutical agent(s) and/or the desired therapeutic and/or prophylactic effect to be achieved. In general, it is expected that the additional pharmaceutical agent(s) in combination be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually. In certain embodiments, the additional agent is ergotamine, an anti-inflammatory agent, a steroid, a barbiturate, an opioid analgesic, caffeine, or a combination thereof. In some embodiments, the additional agent is ergotamine. In certain embodiments, the additional agent is an anti-inflammatory agent. In some embodiments, the anti-inflammatory agent is one or more of a cyclooxygenase-2 (COX-2) inhibitor, a cyclooxygenase-3 (COX-3) inhibitor, a non-steroidal anti-inflammatory drug (NSAID), or a combination thereof. In certain embodiments, the anti-inflammatory agent is a cyclooxygenase-2 (COX-2) inhibitor. In some embodiments, the COX-2 inhibitor is celecoxib, valdecoxib, rofecoxib, or a combination thereof. In certain embodiments, the anti-inflammatory agent is a cyclooxygenase-3 (COX-3) inhibitor. In some embodiments, the COX-3 inhibitor is acetaminophen, phenacetin, antipyrine, dipyrone, or a combination thereof. In certain embodiments, the anti-inflammatory agent is a non-steroidal anti-inflammatory drug (NSAID). In some embodiments, the NSAID is ibuprofen, naproxen, sulindac, ketoprofen, tolmetin, etodolac, fenoprofen, diclofenac, flurbiprofen, piroxicam, ketorolac, indomethacin, nabumetone, oxaprozin, mefanamic acid, diflunisal, or a combination thereof. In certain embodiments, the additional agent is a steroid. In some embodiments, the additional agent is a barbiturate. In certain embodiments, the barbiturate is secobarbital, mephobarbital, pentobarbital, butabarbital, phenobarbital, amobarbital, or a combination thereof. In some embodiments, the additional agent is an opioid analgesic. In certain embodiments, the opioid analgesic is codeine, fentanyl, hydrocodone, hydromorphone, meperidine, methadone, morphine, oxycodone, or a combination thereof. Also encompassed by the disclosure are kits (e.g., pharmaceutical packs). In some embodiments, the kits disclosed herein comprise a pharmaceutical composition or compound disclosed herein and a container (e.g., a vial, ampule, bottle, syringe, and/or dispenser package, or other suitable container). In some embodiments, the disclosed kits optionally further include a second container comprising a pharmaceutical excipient for dilution or suspension of a pharmaceutical composition or compound disclosed herein. In some embodiments, the pharmaceutical composition or compound disclosed herein provided in the first container and the second container are combined to form one unit dosage form. Thus, in one embodiment, disclosed herein are kits including a first container comprising a compound or pharmaceutical composition disclosed herein. In certain embodiments, the kits are useful for treating pain, fever, or inflammation in a subject in need thereof. In certain embodiments, the kits are useful for preventing pain, fever, or inflammation in a subject in need thereof. In certain embodiments, the kits are useful for reducing the risk of developing pain, fever, or inflammation in a subject in need thereof. In certain embodiments, the kits are useful for inhibiting the activity of COX-2 in a subject or cell. In certain embodiments, a kit disclosed herein further includes instructions for using the kit. In some embodiments, a kit disclosed herein also includes information as required by a regulatory agency such as the U.S. Food and Drug Administration (FDA). In certain embodiments, the information included in the kits is prescribing information. In certain embodiments, the kits and instructions provide for treating pain, fever, or inflammation in a subject in need thereof. In certain embodiments, the kits and instructions provide for preventing pain, fever, or inflammation in a subject in need thereof. In certain embodiments, the kits and instructions provide for reducing the risk of developing pain, fever, or inflammation in a subject in need thereof. In certain embodiments, the kits and instructions provide for inhibiting the activity of COX-2 in a subject or cell. In some embodiments, a kit disclosed herein includes one or more additional pharmaceutical agents disclosed herein as a separate composition. Methods of Use In another aspect, the present disclosure provides a method for treating pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides a method for preventing pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a prophylactically effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in therapy. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in vitro comprising contacting a cell, tissue, or biological sample with a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In yet another aspect, the present disclosure provides a method of inhibiting COX-2 activity in vitro comprising contacting a cell, tissue, or biological sample with a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in vitro. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in vitro. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in a subject in need thereof comprising administering to the subject an effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a method of inhibiting COX-2 activity in a subject in need thereof comprising administering to the subject an effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides a method for treating pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides a method for preventing pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a prophylactically effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in therapy. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in vitro comprising contacting a cell, tissue, or biological sample with a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In yet another aspect, the present disclosure provides a method of inhibiting COX-2 activity in vitro comprising contacting a cell, tissue, or biological sample with a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in vitro. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in vitro. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in a subject in need thereof comprising administering to the subject an effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a method of inhibiting COX-2 activity in a subject in need thereof comprising administering to the subject an effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides a method for treating pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides a method for preventing pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a prophylactically effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in therapy. In yet another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in vitro comprising contacting a cell, tissue, or biological sample with a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In yet another aspect, the present disclosure provides a method of inhibiting COX-2 activity in vitro comprising contacting a cell, tissue, or biological sample with a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in vitro. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in vitro. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in a subject in need thereof comprising administering to the subject an effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a method of inhibiting COX-2 activity in a subject in need thereof comprising administering to the subject an effective amount of a compound disclosed herein (e.g., a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides the use of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides a method for treating pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a therapeutically effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides a method for preventing pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a prophylactically effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in therapy. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in vitro comprising contacting a cell, tissue, or biological sample with d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In yet another aspect, the present disclosure provides a method of inhibiting COX-2 activity in vitro comprising contacting a cell, tissue, or biological sample with d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in vitro. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in vitro. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in a subject in need thereof comprising administering to the subject an effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a method of inhibiting COX-2 activity in a subject in need thereof comprising administering to the subject an effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides a method for treating pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a therapeutically effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides a method for preventing pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a prophylactically effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in therapy. In yet another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in vitro comprising contacting a cell, tissue, or biological sample with d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In yet another aspect, the present disclosure provides a method of inhibiting COX-2 activity in vitro comprising contacting a cell, tissue, or biological sample with d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in vitro. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in vitro. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in a subject in need thereof comprising administering to the subject an effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a method of inhibiting COX-2 activity in a subject in need thereof comprising administering to the subject an effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides a method for treating pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a therapeutically effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides a method for preventing pain, fever, or inflammation in a subject in need thereof comprising administering to the subject a prophylactically effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in treating pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in preventing pain, fever, or inflammation in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in therapy. In yet another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in vitro comprising contacting a cell, tissue, or biological sample with d3-etoricoxib, or a pharmaceutically acceptable salt thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In yet another aspect, the present disclosure provides a method of inhibiting COX-2 activity in vitro comprising contacting a cell, tissue, or biological sample with d3-etoricoxib, or a pharmaceutically acceptable salt thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in vitro. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in vitro. In another aspect, the present disclosure provides a method of inhibiting cyclooxygenase (COX) activity in a subject in need thereof comprising administering to the subject an effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides a method of inhibiting COX-2 activity in a subject in need thereof comprising administering to the subject an effective amount of d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides the use of d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for inhibiting COX-2 activity in a subject in need thereof. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in inhibiting cyclooxygenase (COX) activity in a subject in need thereof. In some embodiments, the cyclooxygenase is one or more of COX-1, COX-2, or COX-3. In some embodiments, the cyclooxygenase is COX-1. In some embodiments, the cyclooxygenase is COX-2. In some embodiments, the cyclooxygenase is COX-3. In another aspect, the present disclosure provides d3-etoricoxib, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in inhibiting COX-2 activity in a subject in need thereof. In some embodiments, the pain, fever, or inflammation is associated with one or more of a viral infection, the common cold, bursitis, myositis, synovitis, arthritis, a degenerative joint disease, a burn, hemophilic arthropathy, migraine, and rheumatic fever. In some embodiments, the viral infection is influenza or COVID-19. In some embodiments, the degenerative joint disease is osteoarthritis. In some embodiments, the pain is one or more of acute pain, post-operative pain, chronic musculoskeletal pain, or short-term pain. In some embodiments, the pain is one or more of low back pain, chronic low back pain, neck pain, dysmenorrhea, headache, toothache, a muscle sprain, a muscle strain, neuralgia, and a burn. In some embodiments, the short-term pain is cramp-like pain. In some embodiments, the cramp-like pain occurs before or during a menstrual period. In some embodiments, the post-operative pain is due to dental surgery. In some embodiments, the pain, fever, or inflammation is associated with one or more of osteoarthritis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and gout. In some embodiments, the pain is acute pain. In some embodiments, the acute pain is caused by one or more of acute injury, trauma, illness, or surgery (for example, open chest surgery (including open heart surgery or bypass surgery)). Acute pain also includes, but is not limited to, one or more of headache, postoperative pain, pain caused by kidney stones, pain in the gall bladder, pain caused by stones in the gall bladder, birth pain, rheumatic pain, dental pain including toothache, pain due to sports injuries, carpal tunnel syndrome, burns, musculoskeletal sprains and strains, muscle tendon deformities, neck and shoulder pain, dyspepsia, gastric ulcer, duodenal ulcer, dysmenorrhea, as well as endometriosis. In some embodiments, the pain is post-operative pain. In some embodiments, the post-operative pain is associated with anorectal, pelvic floor, and urogynecologic procedures (vaginal/perineal approach) (e.g., colon resection, hemorrhoidectomy, vaginal hysterectomy). In some embodiments, the pain is associated with obstetric procedures (e.g., cesarean delivery, vaginal delivery). In some embodiments, the pain is associated with breast procedures (e.g., lumpectomy, mastectomy, reconstruction, reduction). In some embodiments, the pain is associated with thoracic procedures (e.g., thoracoscopy, repair of pectus excavatum in children (Nuss procedure)). In some embodiments, the pain is associated with extremity trauma requiring surgery (e.g., amputation, open reduction, and internal fixation). In some embodiments, the pain is associated with spine procedures (e.g., fusion in both adults and children, laminectomy). In some embodiments, the pain is associated with a procedure related to a sport-related injury (e.g., ACL repair and reconstruction, joint arthroscopy, rotator cuff repair). In some embodiments, the pain is associated with joint replacement (e.g., total hip arthroplasty (THA), total knee arthroplasty (TKA)). In some embodiments, the pain is associated with laparoscopic or open abdominal wall procedures (e.g., femoral hernia, incisional hernia, inguinal hernia). In some embodiments, the pain is associated with laparoscopic abdominal procedures (e.g., appendectomy, bariatric surgery, cholecystectomy, colectomy, hysterectomy, prostatectomy). In some embodiments, the pain is associated with open abdominal procedures (e.g., appendectomy, cholecystectomy, colectomy, hysterectomy); see also Laparoscopic abdominal procedures. In some embodiments, the pain is dental pain. In some embodiments, the pain is associated with dental surgeries (e.g., third molar extraction). In some embodiments, the pain is associated with oropharyngeal procedures (e.g., tonsillectomy). In some embodiments, the pain is cancer pain. In some embodiments, the pain is associated dysmenorrhea. In some embodiments, the pain is associated with endometriosis. In some embodiments, the pain is associated with migraines. In some embodiments, the migraine is associated with von Willebrand's disease or in a patient having von Willebrand's disease. In some embodiments, the pain is associated with acute gouty arthritis. In some embodiments, the pain is postpartum pain. In some embodiments, the postpartum pain is associated with postpartum depression. In some embodiments, the method reduces the risk of developing postpartum depression. In some embodiments, provided herein is a method for treating postpartum depression in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof. In some embodiments, the fever is dengue fever. In some embodiments, the inflammation is associated with acute gouty arthritis. In some embodiments, the inflammation is associated with endometriosis. In some embodiments, the methods disclosed herein avoid or reduce the incidence of one or more side effects or adverse reactions associated with non-isotopically enriched etoricoxib at an equivalent dose. In some embodiments, the methods disclosed herein avoid or reduce the incidence of one or more side effects or adverse reactions associated with non-isotopically enriched etoricoxib at an equivalent dose when administered by the same route of administration. In some embodiments, the method of treating pain, fever, or inflammation reduces side effects associated with the administration of non-isotopically enriched etoricoxib at an equivalent dose. In some embodiments, the method of treating pain, fever, or inflammation reduces side effects associated with the administration of non-isotopically enriched etoricoxib at an equivalent dose when administered by the same route of administration. In some embodiments, the side effect is indigestion, abdominal pain, melena, tiredness, dizziness, constipation, diarrhea, swelling (e.g., swollen ankles), heart palpitations, shortness of breath, bruising, headache, flu-like symptoms, high blood pressure, chest pains, jaundice, or liver problems. In some embodiments, the subject is a mammal. In some embodiments, the subject is a human. In some embodiments, the subject is at least 16 years of age. In some embodiments, the subject is at most 65 years of age. In some embodiments, the therapeutically effective amount is administered to the subject once daily. In some embodiments, the therapeutically effective amount is administered to the subject twice daily. In some embodiments, the therapeutically effective amount is administered to the subject three times daily. In some embodiments, the method further comprises administering a loading dose of the compound and a maintenance dose of the compound. In some embodiments, the therapeutically effective amount is administered with food. In some embodiments, the therapeutically effective amount is administered under fasted conditions. In some embodiments, about 10 mg, about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, or about 900 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 10 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 25 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 50 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 75 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 100 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 150 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 200 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 250 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 300 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 350 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 400 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 450 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 500 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 550 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 600 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 650 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 700 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 750 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 800 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 850 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, about 900 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, or a pharmaceutical composition thereof, is administered to the subject per day. In some embodiments, the method further comprises administering to the subject ergotamine, an anti-inflammatory agent, a steroid, a barbiturate, an opioid analgesic, caffeine, or a combination thereof. In some embodiments, the anti-inflammatory agent is a cyclooxygenase-2 (COX-2) inhibitor, a cyclooxygenase-3 (COX-3) inhibitor, a non-steroidal anti-inflammatory drug (NSAID), or a combination thereof. In some embodiments, the NSAID is ibuprofen, naproxen, sulindac, ketoprofen, tolmetin, etodolac, fenoprofen, diclofenac, flurbiprofen, piroxicam, ketorolac, indomethacin, nabumetone, oxaprozin, mefanamic acid, diflunisal, or a combination thereof. In some embodiments, the opioid analgesic is codeine, fentanyl, hydrocodone, hydromorphone, meperidine, methadone, morphine, oxycodone, or a combination thereof. In some embodiments, the barbiturate is secobarbital, mephobarbital, pentobarbital, butabarbital, phenobarbital, amobarbital, or a combination thereof. In some embodiments, the COX-2 inhibitor is celecoxib, valdecoxib, rofecoxib, or a combination thereof. In some embodiments, the COX-3 inhibitor is acetaminophen, phenacetin, antipyrine, dipyrone, or a combination thereof. In some embodiments, the therapeutically effective amount is about 5 mg to about 300 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 5 mg to about 250 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 5 mg to about 200 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 5 mg to about 150 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 5 mg to about 140 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 5 mg to about 130 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 5 mg to about 120 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 5 mg, 7.5 mg, 10 mg, 12.5 mg, 15 mg, 17.5 mg, 20 mg, 22.5 mg, 25 mg, 27.5 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 100 mg, 110 mg, 120 mg, 140 mg, 160 mg, 180 mg, 200 mg, 220 mg, 240 mg, 260 mg, 280 mg, or 300 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 7.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 10 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 12.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 15 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 17.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 20 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 22.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 25 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 27.5 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 30 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 35 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 40 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 45 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 50 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 55 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments the therapeutically effective amount is about 60 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 65 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 70 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 75 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 80 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 85 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 90 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 100 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 110 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 120 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 140 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 160 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 180 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 200 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 220 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 240 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 260 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 280 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 300 mg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 0.1 mg/kg to about 25 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 0.1 mg/kg to about 20 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 0.1 mg/kg to about 15 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 0.1 mg/kg to about 10 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 0.1 mg/kg to about 5 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 1 mg/kg to about 25 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 1 mg/kg to about 20 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 1 mg/kg to about 15 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 1 mg/kg to about 10 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 1 mg/kg to about 5 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 0.1 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, about 10 mg/kg, about 11 mg/kg, about 12 mg/kg, about 13 mg/kg, about 14 mg/kg, about 15 mg/kg, about 16 mg/kg, about 17 mg/kg, about 18 mg/kg, about 19 mg/kg, about 20 mg/kg, or about 25 mg/kg of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 0.1 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 0.5 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 1 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 2 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 3 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 4 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 5 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 6 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 7 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 8 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 9 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 10 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 11 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 12 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 13 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 14 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 15 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 16 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 17 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 18 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 19 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In certain embodiments, the therapeutically effective amount is about 20 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. In some embodiments, the therapeutically effective amount is about 25 mg/kg, of a compound of any one of Formulae (I), (II), or (III), or d3-etoricoxib, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof. Methods of Preparation In another aspect, the present disclosure provides a method of preparing a compound of Formula (I): or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, the method comprising contacting a compound of Formula (III): or a salt, hydrate, solvate, polymorph, or co-crystal thereof with a source of nitrogen to produce the compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof, wherein: each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium; provided that at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium. In another aspect, the present disclosure provides a method of preparing a compound of Formula (I): or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, the method comprising:(1) contacting a compound of Formula (II): or a salt, hydrate, solvate, polymorph, or co-crystal thereof with one or more acids to produce a compound of Formula (III): or a salt, hydrate, solvate, polymorph, or co-crystal thereof; and(2) contacting the compound of Formula (III), or a salt, hydrate, solvate, polymorph, or co-crystal thereof with a source of nitrogen to produce the compound Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof; wherein each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium; provided that at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium. In another aspect, the present disclosure provides a method of preparing a compound of Formula (I): or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, the method comprising:(1) contacting a compound of Formula (A): or a salt, hydrate, solvate, polymorph, or co-crystal thereof with one or more bases and a compound of Formula (B): or a salt, hydrate, solvate, polymorph, or co-crystal thereof to produce a compound of Formula (II): or a salt, hydrate, solvate, polymorph, or co-crystal thereof;(2) contacting the compound of Formula (II), or a salt, hydrate, solvate, polymorph, or co-crystal thereof with one or more acids to produce a compound of Formula (III): or a salt, hydrate, solvate, polymorph, or co-crystal thereof; and(3) contacting the compound of Formula (III) with a source of nitrogen to produce the compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof; wherein: each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium, provided that at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium; each of RB1, RB2, RB3, and RB4is independently optionally substituted alkyl; and X−is an anion. In another aspect, the present disclosure provides a method of preparing a compound of Formula (I): or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, co-crystal, metabolite, or prodrug thereof, the method comprising:(1) contacting a compound of Formula (C): or a salt, hydrate, solvate, polymorph, or co-crystal thereof with a compound of Formula (D): or a salt, hydrate, solvate, polymorph, or co-crystal thereof to produce a compound of Formula (A): or a salt, hydrate, solvate, polymorph, or co-crystal thereof;(2) contacting the compound of Formula (A), or a salt, hydrate, solvate, polymorph, or co-crystal thereof with one or more bases and a compound of Formula (B): or a salt, hydrate, solvate, polymorph, or co-crystal thereof to produce a compound of Formula (II): or a salt, hydrate, solvate, polymorph, or co-crystal thereof;(3) contacting the compound of Formula (II), or a salt, hydrate, solvate, polymorph, or co-crystal thereof with one or more acids to produce a compound of Formula (III): or a salt, hydrate, solvate, polymorph, or co-crystal thereof; and(4) contacting the compound of Formula (III) with a source of nitrogen to produce the compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or co-crystal thereof; wherein: each of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is independently hydrogen or deuterium, provided that at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, and R15is deuterium; each of RB1, RB2, RB3, and RB4is independently optionally substituted alkyl; X−is an anion; and Z is halogen. In some embodiments, each of RB1, RB2, RB3, and RB4is independently optionally substituted alkyl. In some embodiments, each of RB1, RB2, RB3, and RB4is independently optionally substituted C1-6alkyl. In some embodiments, each of RB1, RB2, RB3, and RB4is independently substituted C1-6alkyl. In some embodiments, each of RB1, RB2, RB3, and RB4is independently unsubstituted C1-6alkyl. In some embodiments, each of RB1, RB2, RB3, and RB4is independently methyl, ethyl, propyl, or butyl. In some embodiments, each of RB1, RB2, RB3, and RB4is independently methyl, ethyl, or propyl. In some embodiments, each of RB1, RB2, RB3, and RB4is independently methyl or ethyl. In some embodiments, each of RB1, RB2, RB3, and RB4is methyl. In some embodiments, X−is a non-coordinating anion. In some embodiments, X−is −PF6. In some embodiments, X−is tetrafluoroborate. In some embodiments, X−is perchlorate. In some embodiments, Z is Cl, Br, or I. In some embodiments, Z is Cl. In some embodiments, Z is Br. In some embodiments, Z is I. In some embodiments, the step of contacting a compound of Formula (C) with a compound of Formula (D) is transition metal-catalyzed. In some embodiments, the step of contacting a compound of Formula (C) with a compound of Formula (D) is palladium-catalyzed. In some embodiments, the step of contacting a compound of Formula (C) with a compound of Formula (D) proceeds via enolate arylation. In some embodiments, the step of contacting a compound of Formula (C) with a compound of Formula (D) proceeds via transition metal-catalyzed enolate arylation. In some embodiments, the step of contacting a compound of Formula (C) with a compound of Formula (D) proceeds via palladium-catalyzed enolate arylation. In some embodiments, the base is a strong base. In some embodiments, the base is a non-nucleophilic base. In some embodiments, the base is tert-butoxide salt. In some embodiments, the base is potassium tert-butoxide. In some embodiments, the base is sodium tert-butoxide. In some embodiments, the base is lithium diisopropylamide (LDA). In some embodiments, the acid is an organic acid. In some embodiments, the acid is trifluoroacetic acid. In some embodiments, the acid is acetic acid. In some embodiments, the one or more acids are acetic acid and trifluoroacetic acid. In some embodiments, one or more steps are performed in organic solvent. In some embodiments, one or more steps are performed in polar aprotic organic solvent. In some embodiments, one or more steps are performed in tetrahydrofuran. In some embodiments, the step of contacting a compound of Formula (A) with one or more bases is performed in tetrahydrofuran. In some embodiments, the source of nitrogen is ammonia. In some embodiments, the source of nitrogen is an ammonium salt. In some embodiments, the source of nitrogen is ammonium hydroxide. In some embodiments, the source of nitrogen is ammonium acetate. EXAMPLES In order that the present disclosure may be more fully understood, the following examples are set forth. The synthetic and biological examples disclosed in this application are offered to illustrate the compounds, pharmaceutical compositions, and methods disclosed herein and are not to be construed in any way as limiting in their scope. Abbreviations AUC0-∞Area under the concentration-time curve from time zero extrapolated to infinityAUC0-tArea under the concentration-time curve from time zero to time tCmaxMaximum concentration observedHPLC High Performance/Pressure Liquid ChromatographyLC/MS Liquid Chromatography Tandem Mass Spectrometrymin Minute(s)No. NumberSD Standard deviationt1/2Terminal half-lifeTmaxTime of observed maximum concentration Example 1 Synthesis of d3-Etoricoxib d3-Etoricoxib was synthesized according to the following synthetic scheme: Example 2 d3-Etoricoxib 50 mg Batch Analysis d3-Etoricoxib was prepared in a 50 mg batch (batch no. 0605175). HPLC (65:35:10 mM MeOH:H2O:NH4OAc, 1 mL/min, 210 nm): 94.1% purity, Rtof 7.53 minutes (FIG.1). LC/MS (MM-ES+APCl, Dual pos): m/z 362.1 (M+1), d0 detected at 0.53% (FIGS.2and3).1H NMR (methanol-d4, 400 MHz): δ8.71 (d, 1H, J=2.3 Hz), 8.27 (d, 1H, J=1.8 Hz), 8.00 (d, 1H, J=2.3 Hz), 7.9-7.9 (m, 2H, J=8.3 Hz), 7.67 (dd, 1H, J=2.3, 8.3 Hz), 7.4-7.5 (m, 2H, J=8.3 Hz), 7.23 (d, 1H, J=7.8 Hz), 3.1-3.1 (m, 3H). TLC (80:20 ethyl acetate:heptane): single spot, Rf0.21, visualized with UV (FIG.4). Example 3 d3-Etoricoxib 100 mg Batch Analysis d3-Etoricoxib was prepared in a 100 mg batch (batch no. 0605176). HPLC (65:35:10 mM MeOH:H2O:NH4OAc, 1 mL/min, 210 nm): 96.2% purity, Rtof 7.55 minutes (FIG.5). LC/MS (MM-ES+APCl, Dual pos): m/z 362.1 (M+1), d0 detected at 3.33% (FIGS.6and7).1H NMR (methanol-d4, 400 MHz): δ8.71 (d, 1H, J=2.3 Hz), 8.27 (d, 1H, J=2.3 Hz), 8.00 (d, 1H, J=2.3 Hz), 7.91 (d, 2H, J=8.7 Hz), 7.67 (dd, 1H, J=2.3, 8.3 Hz), 7.50 (m, 2H, J=8.0 Hz), 7.23 (d, 1H, J=8.3 Hz), 3.1-3.1 (m, 3H). TLC (80:20 ethyl acetate:heptane): single spot, Rf0.21, visualized with UV (FIG.8). Example 4 Single Dose Crossover Study A single dose crossover study was conducted to compare the pharmacokinetics of d3-etoricoxib and non-isotopically enriched etoricoxib in dogs. Four male dogs were dosed, and animals received each of the following treatments sequentially, with a minimum of four days washout between doses. The doses are summarized in Table 1. TABLE 1Doses of d3-etoricoxib and etoricoxib administeredto four male dogs in a crossover study.StudyDoseRoute ofVehicle/DayLevelCompoundAdministrationPresentation12.00 mg/kgd3-etoricoxiboral (gavage)1% methylcellulosein water81.98 mg/kgetoricoxiboral (gavage)1% methylcellulosein water Blood samples were taken at 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post-dose. Plasma was analyzed for d3-etoricoxib or non-isotopically enriched etoricoxib concentration. Results are summarized in Table 2. TABLE 2Pharmacokinetic parameters of d3-etoricoxib and etoricoxibadministered to four male dogs in a crossover study.Cmax(ng/mL)AUC0.5-24 h(ng*h/mL)Animald3-etoricoxibetoricoxibd3-etoricoxibetoricoxib1001628259516342100279142610246061003593670126411311004317206658336Mean (SD)604 (373)390 (209)865 (341)582 (197)Average %+54.9%N/A+48.6%N/Adelta d3-etoricoxib vsetoricoxib A comparison of the mean plasma concentration (ng/mL) of etoricoxib and d3-etoricoxib over 24 hours following administration of etoricoxib (1.98 mg/kg) or d3-etoricoxib (2.00 mg/kg) to a population of four male dogs is shown inFIG.9. Individual mean plasma concentrations (ng/mL) of etoricoxib and d3-etoricoxib over 24 hours following administration of etoricoxib (1.98 mg/kg) or d3-etoricoxib (2.00 mg/kg) to each of four male dogs is shown inFIGS.10A-D. These results demonstrate that, at equivalent doses, d3-etoricoxib is substantially resistant to metabolism in dogs relative to non-isotopically enriched etoricoxib and that this separation occurs almost immediately, resulting in not only higher total exposure (AUC) to d3-etoricoxib, but also a higher Cmax, relative to non-isotopically enriched etoricoxib. Example 5 In Vitro Metabolic Studies of Etoricoxib and d3-Etoricoxib with Human Liver Microsomes The metabolic stability of non-isotopically enriched etoricoxib and d3-etoricoxib, with human liver microsomes was evaluated. Assay conditions are summarized in Table 3. TABLE 3Metabolic Stability with Human LiverMicrosomes - Assay ConditionsStudy species:HumanIncubation volume:300 μL, pH 7.4 in phosphatebuffer, 2 mM MgCl2Protein content:0.5 mg/mLCofactors & concentrations:NADPH (1 mM) and UDPGA (1 mM)DMSO content:0.5%Preincubation time:10 min at 37° C.Incubation times:0, 10, 20, 40, 60 minReplicates:2 with cofactors, 1 without cofactorReaction started by:Addition of study compoundTermination of incubations:2-fold volume of 75% acetonitrileStorage of samples:Immediate analysisTest compound concentration:1 μM Etoricoxib is extensively metabolized in humans with <1% of a dose recovered in urine as the parent drug. The primary pathway for metabolism of etoricoxib in humans is through the production of the methylhydroxy metabolite of etoricoxib (“M1”). Metabolism of etoricoxib involves conversion primarily to the “M1” derivative, mainly (ca. 60%) by CYP3A4, with less contribution by CYPs 1A2, 2C9, 2C19 and 2D6 (ca. 40% collectively). See, e.g., Arcoxia® (etoricoxib) Australian Product Information, available at http://apps.medicines.org.au/files/mkparcox.pdf (last visited Apr. 9, 2021), the contents of which are herein incorporated by reference in their entirety. Production of the “M1” metabolite and d3-etoricoxib (“d2-M1”) was evaluated in this experiment throughout the incubation period. FIGS.11-12show peak area and relative percentage of parent corresponding to the “M1” and “d2-M1” metabolites formed from non-isotopically enriched etoricoxib and d3-etoricoxib over 60 minutes time on incubation with human liver microsomes in the microsomal stability assay, respectively. Under these conditions, approximately three-fold less of the “d2-M1” metabolite was formed from d3-etoricoxib than of “M1” formed from etoricoxib in this experiment, supporting the result demonstrated in dogs that d3-etoricoxib is resistant to the primary metabolic pathway in humans and dogs of conversion from parent to “d2-M1” relative to non-isotopically enriched etoricoxib and its respective conversion to “M1.” Example 6 In Vitro Metabolic Studies of Etoricoxib, d3-Etoricoxib, and d4-Etoricoxib in CYP3A4 Incubations The metabolic stability of non-isotopically enriched etoricoxib, d3-etoricoxib, and an alternate deuterated form of etoricoxib, d4-etoricoxib (5-chloro-6′-methyl-3-(4-(methylsulfonyl)phenyl-2,3,5,6-d4)-2,3′-bipyridine, CAS No.: 1131345-14-6), in CYP3A4 incubations was evaluated. d4-Etoricoxib has the chemical structure: Assay conditions are summarized in Table 4. TABLE 4Metabolic Stability of Etoricoxib, d3-Etoricoxib, and d4-Etoricoxibin CYP3A4 Incubations - Optimization Assay ConditionsStudy species:Human recombinant CYP enzyme 3A4Buffer:0.1M phosphate buffer pH 7.4, 2 mM MgCl2Spiking solvent:50% DMSO (1/100 to incubation)Test compound1 μMconcentration:Protein content:100 μmol/mLCofactors:NADPH (1 mM)Incubation time:0, 10, 20, 30, and 40 min at 37° CReplicates:2Termination of2-fold volume of cold 75% acetonitrileincubations:Storage of samples:Immediate analysis Human recombinant CYP3A4 (Corning, item #456202, lot #9322001) was chosen for the assay conditions as CYP3A4 is the principle enzyme responsible for the metabolism of etoricoxib in humans. Formation of the methylhydroxy metabolites formed from etoricoxib, d3-etoricoxib, and d4-etoricoxib was monitored over time (Table 5,FIGS.13and14). TABLE 5Formation of M1 Metabolites Derived from Etoricoxib, d3-Etoricoxib,and d4-Etoricoxib in CYP3A4 Incubations (100 pmol/mL)IncubationLC/MS Peak Area% of Parent at BaselineTimed3-d4-d3-d4-(min)EtoricoxibEtoricoxibEtoricoxibEtoricoxibEtoricoxibEtoricoxib0——————102,803,382188,9082,160,6561006.777.1206,011,436475,8885,037,1921007.983.8308,373,189717,7177,045,4361008.684.14010,079,114777,4647,765,6021007.777.0 FIG.13shows peak area corresponding to the “M1,” “d2-M1,” and “d4-M1” metabolites formed from etoricoxib, d3-etoricoxib, and d4-etoricoxib over time on incubation with certain human recombinant CYP3A4 alleles.FIG.14shows percent total of “M1,” “d2-M1,” and “d4-M1” metabolites formed from etoricoxib, d3-etoricoxib, and d4-etoricoxib over time on incubation with certain human recombinant CYP3A4 alleles. Formation over time of “M1” formed from etoricoxib was similar to that of “d4-M1” formed from d4-etoricoxib. Formation of “d2-M1” formed from d3-etoricoxib was about six-fold lower than formation of “M1” formed from etoricoxib or “d4-M1” formed from d4-etoricoxib. Thus, despite containing less deuterium than d4-etoricoxib, d3-etoricoxib demonstrated a substantially different susceptibility to metabolism by CYP3A4, while d4-etoricoxib exhibited susceptibility to metabolism by CYP3A4 almost identical to non-isotopically enriched etoricoxib. These results demonstrate that deuteration, in and of itself, does not necessarily affect the metabolism of a particular molecule. The location of deuteration can greatly affect metabolic outcome and requires experimentation to determine what, if any, effects may occur EQUIVALENTS AND SCOPE In the claims articles such as “a,” “an,” and “the” may mean one or more than one unless indicated to the contrary or otherwise evident from the context. Claims or descriptions that include “or” between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The invention includes embodiments in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process. Furthermore, the invention encompasses all variations, combinations, and permutations in which one or more limitations, elements, clauses, and descriptive terms from one or more of the listed claims is introduced into another claim. For example, any claim that is dependent on another claim can be modified to include one or more limitations found in any other claim that is dependent on the same base claim. Where elements are presented as lists, e.g., in Markush group format, each subgroup of the elements is also disclosed, and any element(s) can be removed from the group. It should it be understood that, in general, where the invention, or aspects of the invention, is/are referred to as comprising particular elements and/or features, certain embodiments of the invention or aspects of the invention consist, or consist essentially of, such elements and/or features. For purposes of simplicity, those embodiments have not been specifically set forth in haec verba herein. It is also noted that the terms “comprising” and “containing” are intended to be open and permits the inclusion of additional elements or steps. Where ranges are given, endpoints are included. Furthermore, unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or sub-range within the stated ranges in different embodiments of the invention, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise. Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation many equivalents to the specific embodiments disclosed herein. The scope of the present embodiments disclosed herein is not intended to be limited to the above Description, but rather is as set forth in the appended claims. Those of ordinary skill in the art will appreciate that various changes and modifications to this description may be made without departing from the spirit or scope of the present invention, as defined in the following claims.
291,338
11858910
DETAILED DESCRIPTION OF THE INVENTION Definitions and General Terminology Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structures and formulas. The invention is intended to cover all alternatives, modifications, and equivalents which may be included within the scope of the present invention as defined by the claims. One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated literature, patents, and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls. It is further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination. As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, and the Handbook of Chemistry and Physics, 75th Ed. 1994. Additionally, general principles of organic chemistry are described in “Organic Chemistry”, Thomas Sorrell, University Science Books, Sausalito: 1999, and “March's Advanced Organic Chemistry” by Michael B. Smith and Jerry March, John Wiley & Sons, New York: 2007, the entire contents of which are hereby incorporated by reference. The grammatical articles “a”, “an” and “the”, as used herein, are intended to include “at least one” or “one or more” unless otherwise indicated herein or clearly contradicted by the context. Thus, the articles are used herein to refer to one or more than one (i.e., at least one) of the grammatical objects of the article. By way of example, “a component” means one or more components, and thus, possibly, more than one component is contemplated and may be employed or used in an implementation of the described embodiments. “Stereoisomers” refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include enantiomer, diastereomers, conformer (rotamer), geometric (cis/trans) isomer, atropisomer, etc. “Chiral” refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner. “Enantiomers” refer to two stereoisomers of a compound which are non-superimposable mirror images of one another. “Racemate” or “racemic mixture” refers to an equimolar mixture of two enantiomers lacking optical activity. “Diastereomer” refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boling points, spectral properties or reactivities. A mixture of diastereomers may be separated under high resolution analytical procedures such as electrophoresis and chromatography such as HPLC. Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., “Stereochemistry of Organic Compounds”, John Wiley & Sons, Inc., New York, 1994. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and l or (+) and (−) are employed to designate the sign of rotation of plane-polarized light by the compound, with (−) or l meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. A specific stereoisomer may be referred to as an enantiomer, and a mixture of such stereoisomers is called an enantiomeric mixture. A mixture of enantiomers with a ratio of 50:50 is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. Any asymmetric atom (e.g., carbon or the like) of the compound(s) disclosed herein can be present in racemic or enantiomerically enriched, for example the (R)-, (S)- or (R, S)-configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R)- or (S)-configuration. Depending on the choice of the starting materials and procedures, the compounds can be present in the form of one of the possible stereoisomers or as mixtures thereof, such as racemates and diastereoisomer mixtures, depending on the number of asymmetric carbon atoms. Optically active (R)- and (S)-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituent may be E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis- or trans-configuration. Any resulting mixtures of stereoisomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric isomers, enantiomers, diastereomers, for example, by chromatography and/or fractional crystallization. Any resulting racemates of final products or intermediates can be resolved into the optical antipodes by methods known to those skilled in the art, e.g., by separation of the diastereomeric salts thereof. Racemic products can also be resolved by chiral chromatography, e.g., high performance liquid chromatography (HPLC) using a chiral adsorbent. Preferred enantiomers can also be prepared by asymmetric syntheses. See, for example, Jacques, et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981);Principles of Asymmetric Synthesis(2ndEd. Robert E. Gawley, Jeffrey Aubé, Elsevier, Oxford, UK, 2012); Eliel, E. L. Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); Wilen, S. H. Tables of Resolving Agents and Optical Resolutions p. 268 (E. L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972); Chiral Separation Techniques: A Practical Approach (Subramanian, G. Ed., Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, Germany, 2007). The term “tautomer” or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier. Where tautomerization is possible (e.g., in solution), a chemical equilibrium of tautomers can be reached. For example, protontautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. The term “pharmaceutically acceptable,” as used herein, refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and are effective for their intended use. The terms “optionally substituted with . . . ” and “unsubstituted or substituted with” can be used interchangeably, i.e., the structure is unsubstituted or substituted with one or more of the substituents described in the present invention, the substituents disclosed herein include, but are not limited to, D, F, Cl, Br, I, N3, —CN, —NO2, —NH2, —OH, —SH, —COOH, —CONH2, —C(═O)NHCH3, —C(═O)N(CH3)2, —C(═O)-alkyl, —C(═O)-alkoxy, alkyl, alkenyl, alkynyl, haloalkyl, alkoxy, haloalkoxy, alkylthio, alkylamino, hydroxy-substituted alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and so on. In general, the term “substituted” refers to the replacement of one or more hydrogen radicals in a given structure or radical with a specified substituent. Unless otherwise indicated, a substituent may substitute at any substitutable position of a radical. When more than one positions of a given structure can be substituted with one or more specified substituents, the substituents may be either the same or different at each position. Furthermore, what need to be explained is that the phrase “each . . . is independently” and “each of . . . and . . . is independently”, unless otherwise stated, should be broadly understood. The specific options expressed by the same symbol are independent of each other in different groups; or the specific options expressed by the same symbol are independent of each other in same groups. As used herein, the term “subject” refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human. As used herein, “patient” refers to a human (including adults and children) or other animal. In one embodiment, “patient” refers to a human. The term “comprise” is an open expression, it means comprising the contents disclosed herein, but don't exclude other contents. At various places in the present specification, substituents of compounds disclosed herein are disclosed in groups or in ranges. It is specifically intended that the invention include each and every individual subcombination of the members of such groups and ranges. For example, the term “C1-C6alkyl” is specifically intended to individually disclose methyl, ethyl, C3alkyl, C4alkyl, C5alkyl, and C6alkyl. At various places in the present specification, linking substituents are described. Where the structure clearly requires a linking group, the Markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the Markush group definition for that variable lists “alkyl” or “aryl” then it is understood that the “alkyl” or “aryl” represents a linking alkylene group or arylene group, respectively. The term “D” or “2H” refers to a single deuterium atom. The terms “halogen” and “halo” can be used interchangeably, which refer to Fluoro (F), Chloro (Cl), Bromo (Br), or Iodo (I). The term “heteroatom” refers to oxygen, sulfur, nitrogen, phosphorus and silicon, including any oxidized form of nitrogen, sulfur, or phosphorus; primary, secondary, tertiary amines and quaternary ammonium salts forms; or a substitutable nitrogen of a heterocyclic ring, for example, N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR (as in N-substituted pyrrolidinyl, wherein R is the substituent described herein). The term “alkyl” or “alkyl group” refers to a saturated linear or branched-chain monovalent hydrocarbon group of 1-20 carbon atoms, wherein the alkyl group is optionally substituted with one or more substituents described herein. In one embodiment, the alkyl group contains 1-6 carbon atoms. In other embodiment, the alkyl group contains 1-4 carbon atoms. In still other embodiment, the alkyl group contains 1-3 carbon atoms. Examples of the alkyl group include, but are not limited to, methyl (Me, —CH3), ethyl (Et, —CH2CH3), n-propyl (n-Pr, —CH2CH2CH3), i-propyl (i-Pr, —CH(CH3)2), n-butyl (n-Bu, —CH2CH2CH2CH3), i-butyl(i-Bu, —CH2CH(CH3)2), s-butyl (s-Bu, —CH(CH3)CH2CH3), t-butyl (t-Bu, —C(CH3)3), and the like. The term “alkenyl” refers to linear or branched-chain monovalent hydrocarbon radical of 2 to 12 carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon sp2double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having “cis” and “trans” orientations, or alternatively, “E” and “Z” orientations. In some embodiments, the alkenyl contains 2 to 8 carbon atoms. In other embodiments, the alkenyl contains 2 to 6 carbon atoms. In still other embodiments, the alkenyl contains 2 to 4 carbon atoms. Some non-limiting examples of the alkenyl group include ethenyl or vinyl (—CH═CH2), allyl (—CH2CH═CH2), 1-propenyl (i.e., propenyl, —CH═CH—CH3), and the like. The term “alkynyl” refers to a linear or branched-chain monovalent hydrocarbon radical of 2 to 12 carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon sp triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. In some embodiments, the alkynyl contains 2 to 8 carbon atoms. In other embodiments, the alkynyl contains 2 to 6 carbon atoms. In still other embodiments, the alkynyl contains 2 to 4 carbon atoms. Examples of such groups include, but are not limited to, ethynyl (—C≡CH), propargyl (—CH2C≡CH), 1-propynyl (i.e., propynyl, —C≡C—CH3), and the like. The term “alkoxy” refers to an alkyl group, as previously defined, attached to the parent molecular moiety via an oxygen atom. Unless otherwise specified, the alkoxy group contains 1-12 carbon atoms. In one embodiment, the alkoxy group contains 1-6 carbon atoms. In other embodiment, the alkoxy group contains 1-4 carbon atoms. In still other embodiment, the alkoxy group contains 1-3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents disclosed herein. Examples of the alkoxy group include, but are not limited to, methoxy (MeO, —OCH3), ethoxy (EtO, —OCH2CH3), 1-propoxy (n-propyl-oxy, n-PrO, n-propoxy, —OCH2CH2CH3), 2-propoxy (i-propyl-oxy, i-PrO, i-propoxy, —OCH(CH3)2), 1-butoxy (n-BuO, n-butoxy, —OCH2CH2CH2CH3), 2-methyl-1-propoxy (i-BuO, i-butoxy, —OCH2CH(CH3)2), 2-butoxy (s-BuO, s-butoxy, —OCH(CH3)CH2CH3), 2-methyl-2-propoxy (t-BuO, t-butoxy, —OC(CH3)3), and the like. The term “alkylthio” refers to an alkyl group, as previously defined, attached to the parent molecular moiety via a sulfur atom. Unless otherwise specified, the alkylthio group contains 1-12 carbon atoms. In one embodiment, the alkylthio group contains 1-6 carbon atoms. In other embodiment, the alkylthio group contains 1-4 carbon atoms. In still other embodiment, the alkylthio group contains 1-3 carbon atoms. The alkylthio group may be optionally substituted with one or more substituents disclosed herein. Examples of the alkylthio group include, but are not limited to, methylthio (MeS, —SCH3), ethylthio (EtS, —SCH2CH3), 1-propylthio (n-PrS, n-propylthio, —SCH2CH2CH3), 2-propylthio (i-PrS, i-propylthio, —SCH(CH3)2), 1-butylthio (n-BuS, n-butylthio, —SCH2CH2CH2CH3), 2-methyl-1-propylthio (i-BuS, i-butylthio, —SCH2CH(CH3)2), 2-butylthio (s-BuS, s-butylthio, —SCH(CH3)CH2CH3), 2-methyl-2-propylthio (t-BuS, t-butylthio, —SC(CH3)3), and the like. The term “alkylamino” embraces “N-alkylamino” and “N,N-dialkylamino”, that is an amino group is independently substituted with one or two alkyl radicals and wherein the alkyl group is as defined herein. Suitable alkylamino radical may be monoalkylamino or dialkylamino. Examples of the alkylamino radical include, but are not limited to, N-methylamino (methylamino), —N-ethylamino (ethylamino), —N,N-dimethylamino (dimethylamino), N,N-diethylamino (diethylamino), and the like. And wherein the alkylamino radical is optionally substituted with one or more substituents described herein. The term “hydroxy-substituted alkyl” refers to an alkyl group substituted with one or more hydroxy groups, wherein the alkyl is as defined herein. Examples of such group include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxy-1-propyl, 3-hydroxy-1-propyl, 2,3-dihydroxypropyl, and the like. The term “haloalkyl” refers to an alkyl group substituted with one or more halo groups, wherein the alkyl is as defined herein. Examples of such group include, but are not limited to, —CHF2, —CF3, —CHFCH2F, —CF2CHF2, —CH2CHF2, —CH2CF3, —CHFCH3, —CH2CH2F, —CF2CH3, —CH2CF2CHF2, and the like. In some embodiments, C1-C6haloalkyl include fluoro substituted C1-C6alkyl; in the other embodiments, C1-C4haloalkyl include fluoro substituted C1-C4alkyl; in still other embodiments, C1-C2haloalkyl include fluoro substituted C1-C2alkyl. The term “haloalkoxy” refers to an alkoxy group substituted with one or more halo groups, wherein the alkoxy is as defined herein. Examples of such group include, but are not limited to, —OCHF2, —OCF3, —OCHFCH2F, —OCF2CHF2, —OCH2CHF2, —OCH2CF3, —OCHFCH3, —OCH2CH2F, —OCF2CH3, —OCH2CF2CHF2, and the like. In some embodiments, C1-C6haloalkoxy include fluoro substituted C1-C6alkoxy; in other embodiments, C1-C4haloalkoxy include fluoro substituted C1-C4alkoxy; in still other embodiments, C1-C2haloalkoxy include fluoro substituted C1-C2alkoxy. The term “consisting of n atoms” or “n-membered”, used interchangeably herein, where n is an integer typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n. For example, “5-10 membered heteroaryl” refers to a heteroaryl consisting of 5, 6, 7, 8, 9 or 10 ring atoms. As another example, piperidinyl is a heterocyclyl consisting of 6 ring atoms or a 6 membered heterocyclyl, and pyridyl is a heteroaryl consisting of 6 ring atoms or a 6 membered heteroaryl. The term “carbocyclyl”, “carbocycle” or “carbocyclic ring” refers to a monovalent or multivalent, nonaromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms as a monocyclic, bicyclic or tricyclic ring system. A carbobicyclyl group includes a spiro carbobicyclyl group or a fused carbobicyclyl group. Suitable carbocyclyl groups include, but are not limited to, cycloalkyl, cycloalkenyl and cycloalkynyl. Further examples of the carbocyclyl group include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like. and wherein the carbocyclyl group is optionally substituted with one or more substituents described herein. The term “cycloalkyl” refers to a monovalent or multivalent saturated ring having 3 to 12 carbon atoms as a monocyclic, bicyclic, or tricyclic ring system, wherein the bicyclic or tricyclic ring system may include fused ring, bridged ring and spiro ring. In some embodiments, the cycloalkyl group contains 3 to 10 carbon atoms. In other embodiments, the cycloalkyl group contains 3 to 8 carbon atoms. In still other embodiments, the cycloalkyl group contains 3 to 6 carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. The cycloalkyl radical is optionally substituted with one or more substituents described herein. The term “heterocycle”, “heterocyclyl”, or “heterocyclic ring” as used interchangeably herein refers to a saturated or partially unsaturated and nonaromatic monocyclic, bicyclic or tricyclic ring system containing 3-12 ring atoms, wherein the bicyclic or tricyclic ring system may include a fused ring, bridged ring and spiro ring. Wherein one or more atoms on the ring each are independently replaced by heteroatom, the heteroatom is as defined herein. In some embodiments, the heterocyclyl group is a monocyclic heterocyclyl having 3-8 ring members (e.g., 2 to 6 carbon atoms and 1 to 3 heteroatoms selected from N, O, P and S, wherein the S or P is optionally substituted with one or more oxo to provide the group SO or SO2, PO or PO2); in other embodiments, the heterocyclyl group is a monocyclic heterocyclyl having 3-6 ring members (e.g., 2 to 5 carbon atoms and 1 to 3 heteroatoms selected from N, O, P and S, wherein the S or P is optionally substituted with one or more oxo to provide the group SO or SO2, PO or PO2); in still other embodiments, the heterocyclyl group is a bicyclic heterocyclyl having 7-12 ring members (e.g., 4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P and S, wherein the S or P is optionally substituted with one or more oxo to provide the group SO or SO2, PO or PO2); and wherein the heterocyclyl group is optionally substituted with one or more substituents described herein. The heterocyclyl may be a carbon atom radical or heteroatom radical. Of which a —CH2— group can be optionally replaced by a —C(═O)— group. Ring sulfur atoms may be optionally oxidized to form S-oxides, and ring nitrogen atoms may be optionally oxidized to form N-oxides. Some non-limiting examples of the heterocyclyl group include oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1,3-dioxolanyl, dithiolanyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thioxanyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-oxa-5-azabicyclo[2.2.1]hept-5-yl, and the like. Some non-limiting examples of heterocyclyl wherein —CH2— group is replaced by —C(═O)— moiety include 2-oxopyrrolidinyl, oxo-1,3-thiazolidinyl, 2-piperidinonyl, 3,5-dioxopiperidinyl, pyrimidinedioneyl, and the like. Some non-limiting examples of oxidized ring sulfur atoms of the heterocyclyl group include sulfolanyl, 1,1-dioxo-thiomorpholinyl, and the like. The heterocyclyl group is optionally substituted with one or more substituents described herein. The term “aryl” refers to monocyclic, bicyclic and tricyclic carbocyclic ring systems having a total of six to fourteen ring members, or six to twelve ring members, or six to ten ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3 to 7 ring members. The aryl group is generally, but not necessarily bonded to the parent molecule through an aromatic ring of the aryl group. The term “aryl” and “aromatic ring” can be used interchangeably herein. Examples of the aryl group may include phenyl, indenyl, naphthyl, and anthryl. The aryl radical is optionally substituted with one or more substituents described herein. The term “heteroaryl” refers to monocyclic, bicyclic and tricyclic ring systems having a total of five to twelve ring members, or five to ten ring members, or five to six ring members, wherein at least one ring in the system is aromatic, and in which at least one ring system contains one or more heteroatoms, and wherein each ring system contains a 5 to 7 members ring. The heteroaryl group is generally, but not necessarily bonded to the parent molecule through an aromatic ring of the heteroaryl group. The term “heteroaryl” may be used interchangeably with the term “heteroaryl ring”, “aromatic heterocyclic” or the term “heteroaromatic compound”. The heteroaryl group is optionally substituted with one or more substituents disclosed herein. In one embodiment, a 5-10 membered heteroaryl comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. Some non-limiting examples of heteroaryl rings include 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, 1,2,3-oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3-triazolyl, 1,2,3-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, pyrazinyl, 1,3,5-triazinyl, and the following bicycles: benzoimidazolyl, benzofuryl, benzothiophenyl, indolyl (e.g., 2-indolyl), purinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl or 4-isoquinolinyl), imidazo[1,2-a]pyridyl, pyrazolo[1,5-a]pyridyl, pyrazolo[1,5-a]pyrimidyl, imidazo[1,2-b]pyridazinyl, [1,2,4]triazolo[4,3-b]pyridazinyl, [1,2,4]triazolo[1,5-a]pyrimidinyl, or [1,2,4]triazolo[1,5-a]pyridyl, and so on. As described herein, a ring system (such as formula f) forming by a bond drawn from a substituent R7to the center of piperidine ring represents that the substituent R7may be substituted at any substitutable position on the piperidine ring, such as formula f1-4. The sentence “R1is optionally substituted with 1, 2, 3, 4 or 5 substituents independently selected from R1a, R1b, R1c, R1dand R1e” means that R1may be unsubstituted, or R1may be substituted with 1, 2, 3, 4 or 5 substituents independently selected from R1a, R1b, R1c, R1dand R1e, wherein R1a, R1b, R1c, R1dand R1ehave the definition described herein, which may be the same group, also may be independently different group. The term “protecting group” or “PG” refers to a substituent that is commonly employed to block or protect a particular functionality while reacting with other functional groups on the compound. For example, an “amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC, Boc), benzyloxycarbonyl (CBZ, Cbz) and 9-fluorenylmethylenoxycarbonyl (Fmoc). Similarly, a “hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Some non-limiting examples of suitable hydroxy-protecting groups include trialkylsilyl, acetyl, benzoyl, and benzyl. A “carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Common carboxy-protecting groups include —CH2CH2SO2Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl) ethyl, 2-(p-nitrophenylsulfonyl)ethyl, 2-(diphenylphosphino)ethyl, nitroethyl and the like. For a general description of protecting groups and their use, see Greene et al.,Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991 and Kocienski et al.,Protecting Groups, Thieme, Stuttgart, 2005. The term “prodrug” refers to a compound that is transformed in vivo into a compound of Formula (I), (II), (III), (IVa), (IVb), (IVc) or (V). Such a transformation can be affected, for example, by hydrolysis of the prodrug form in blood or enzymatic transformation to the parent form in blood or tissue. Prodrugs of the compounds disclosed herein may be, for example, esters. Some common esters which have been utilized as prodrugs are phenyl esters, aliphatic (C1-24) esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound disclosed herein that contains a hydroxy group may be acylated at this position in its prodrug form. Other prodrug forms include phosphates, such as, those phosphate compounds derived from the phosphonation of a hydroxy group on the parent compound. A “metabolite” is a product produced through metabolism in the body of a specified compound or salt thereof. The metabolites of a compound may be identified using routine techniques known in the art and their activities may be determined using tests such as those described herein. Such products may result for example from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzyme cleavage, and the like, of the administered compound. Accordingly, the invention includes metabolites of compounds disclosed herein, including metabolites produced by contacting a compound disclosed herein with a mammal for a sufficient time period. A “pharmaceutically acceptable salts” refers to organic or inorganic salts of a compound disclosed herein. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66: 1-19, which is incorporated herein by reference. Some non-limiting examples of pharmaceutically acceptable and nontoxic salts include salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid and malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphanic acid salt, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, laurylsulfate, malate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, stearate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1-4alkyl)4salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil soluble or dispersable products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, C1-8sulfonate or aryl sulfonate. The term “solvate” refers to an association or complex of one or more solvent molecules and a compound disclosed herein. Some non-limiting examples of the solvent that form solvates include water, isopropanol, ethanol, methanol, dimethylsulfoxide (DMSO), ethyl acetate, acetic acid, ethanolamine or a combination thereof. The term “hydrate” refers to the complex where the solvent molecule is water. The term “hydrate” can be used when said solvent is water. In one embodiment, one solvent molecule is associated with one molecule of the compounds disclosed herein, such as a hydrate. In another embodiment, more than one solvent molecule may be associated with one molecule of the compounds disclosed herein, such as a dihydrate. In still another embodiment, less than one solvent molecule may be associated with one molecule of the compounds disclosed herein, such as a hemihydrate. Furthermore, all the hydrates of the invention retain the biological effectiveness of the non-hydrate form of the compounds disclosed herein. The term “treat”, “treating” or “treatment” of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment “treat”, “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient. In yet another embodiment, “treat”, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In yet another embodiment, “treat”, “treating” or “treatment” refers to preventing or delaying the onset or development or exacerbation of the disease or disorder. The term “preventing” or “prevention” refers to a reduction in risk of acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease). Unless otherwise stated, all suitable isotopic variations, stereoisomers, tautomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs of the compounds disclosed herein are within the scope of the invention. All stereoisomers of the structure disclosed herein are considered within the scope of the invention whether the stereochemistry of the structure is indicated or not, and which are interpreted as disclosed compounds of the invention and included in the invention. When the stereochemistry of a structure is indicated by solid wedge or dash line, the stereoisomer of the structure is definite. N-oxides of the compound disclosed herein are also included in the invention. N-oxides of the compound of the invention can be prepared by oxidizing corresponding nitrogen-containing alkaline substances with common oxidants (hydrogen peroxide) under a rising temperature in the presence of an acid, such as acetic acid, or by reacting with peracid in a suitable solvent, e.g., by reacting with peracetic acid in dichloromethane, ethyl acetate or methyl acetate, or by reacting with 3-chloroperoxybenzoic acid in chloroform or dichloromethane. The compound of Formula (I), (II), (III), (IVa), (IVb), (IVc) or (V) can be exist in salt forms. In one embodiment, the salt is a pharmaceutically acceptable salt thereof. The phrase “pharmaceutically acceptable” refers to that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising a formulation, and/or the mammal being treated therewith. In other embodiments, the salt may not be a pharmaceutically acceptable salt, may be an intermediate used for preparing and/or purifying the compound of Formula (I), (II), (III), (IVa), (IVb), (IVc) or (V) and/or isolating an enantiomer from the compound of Formula (I), (II), (III), (IVa), (IVb), (IVc) or (V). The pharmaceutically acceptable salts of the present invention can be synthesized from a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable. Lists of additional suitable salts can be found, e.g., in “Remington's Pharmaceutical Sciences”,20th ed., Mack Publishing Company, Easton, Pa., (1985); and in “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002). Any formula given herein is also intended to represent isotopically unenriched forms as well as isotopically enriched forms of the compounds. Isotopically enriched compounds have the structure depicted by the general formula given herein, except that one or more atoms are replaced by atom(s) having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, and chlorine, such as2H (deuterium, D),3H,11C,13C,14C,15N,17O,18O,18F,31P,32P,35S,36Cl,125I, respectively. In other aspect, provided herein is an intermediate for preparing the compound of Formula (I), (II), (III), (IVa), (IVb), (IVc) or (V). In other aspect, provided herein is a pharmaceutical composition comprising the compound disclosed herein. In some embodiments, the pharmaceutical composition disclosed herein further comprises at least one of pharmaceutically acceptable carrier, excipient, adjuvant, solvent or a combination thereof. In other embodiment, the pharmaceutical composition can be liquid, solid, semi-solid, gel or spray. DESCRIPTION OF COMPOUNDS OF THE INVENTION The pyridinylmethylenepiperidine derivatives, pharmaceutically acceptable salts, pharmaceutical formulations and compositions thereof disclosed herein can be used to activate 5-HT1Freceptors and inhibit neuronal protein extravasation, and have potential therapeutic use for 5-HT1Freceptor-related diseases, especially migraine. The present invention further describes the synthetic method of the compound. The compounds of the invention show good bioactivity. In one aspect, provided herein is a compound having Formula (I) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each R1, R2, R3, R4, R5, R6, R7, R8and L is as defined herein. In some embodiments, L is —C(═O)—, —C(═S)— or —S(═O)2—. In some embodiments, R1is C1-C6alkyl, C3-C8cycloalkyl, 3-8 membered heterocyclyl, C6-C10aryl or 5-10 membered heteroaryl, wherein R1is optionally substituted with 1, 2, 3, 4 or 5 substituents independently selected from R1a, R1b, R1c, R1dand R1e; wherein R1a, R1b, R1c, R1dand R1ehave the definition described herein. In some embodiments, each R1a, R1b, R1c, R1dand R1eis independently H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —SH, —COOH, —C(═O)NH2, —C(═O)NHCH3, —C(═O)N(CH3)2, —C(═O)—(C1-C6alkyl), —C(═O)—(C1-C6alkoxy), C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6alkylthio, C1-C6alkylamino, hydroxy-substituted C1-C6alkyl, C3-C8cycloalkyl, 3-8 membered heterocyclyl, C6-C10aryl or 5-10 membered heteroaryl. In some embodiments, R2is H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy or hydroxy-substituted C1-C6alkyl. In some embodiments, each of R3, R4and R5is independently H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy or hydroxy-substituted C1-C6alkyl. In some embodiments, R6is H, F, Cl, Br or I. In some embodiments, R7is H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy or hydroxy-substituted C1-C6alkyl. In some embodiments, R8is H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —SH, —COOH, —C(═O)NH2, —C(═O)NHCH3, —C(═O)N(CH3)2, —C(═O)—(C1-C6alkyl), —C(═O)—(C1-C6alkoxy), C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6alkylthio, C1-C6alkylamino, hydroxy-substituted C1-C6alkyl, C3-C8cycloalkyl, 3-8 membered heterocyclyl, C6-C10aryl or 5-10 membered heteroaryl. In some embodiments, R1is C1-C4alkyl, C3-C6cycloalkyl, 3-6 membered heterocyclyl, C6-C10aryl or 5-10 membered heteroaryl, wherein R1is optionally substituted with 1, 2, 3, 4 or 5 substituents independently selected from R1a, R1b, R1c, R1dand R1e; wherein R1a, R1b, R1c, R1dand R1ehave the definition described herein. In other embodiments, wherein R1is phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furyl, thienyl, thiazolyl, oxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, indolyl or quinolyl, wherein R1is optionally substituted with 1, 2, 3, 4 or 5 substituents independently selected from R1a, R1b, R1c, R1dand R1e; wherein R1a, R1b, R1c, R1dand R1ehave the definition described herein. In some embodiments, each R1a, R1b, R1c, R1dand R1eis independently H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —SH, —COOH, —C(═O)NH2, —C(═O)NHCH3, —C(═O)N(CH3)2, —C(═O)—(C1-C4alkyl), —C(═O)—(C1-C4alkoxy), C1-C4alkyl, C2-C4alkenyl, C2-C4alkynyl, C1-C4haloalkyl, C1-C4alkoxy, C1-C4haloalkoxy, C1-C4alkylthio, C1-C4alkylamino, hydroxy-substituted C1-C4alkyl, C3-C6cycloalkyl, 3-6 membered heterocyclyl, C6-C10aryl or 5-10 membered heteroaryl. In other embodiments, each R1a, R1b, R1c, R1dand R1eis independently H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —SH, —COOH, —C(═O)NH2, —C(═O)NHCH3, —C(═O)N(CH3)2, —C(═O)—CH3, —C(═O)—OCH3, methyl, ethyl, n-propyl, i-propyl, allyl, propenyl, propargyl, propinyl, —CHF2, —CF3, —CHFCH2F, —CF2CHF2, —CH2CF3, —CH2CF2CHF2, methoxy, ethoxy, n-propoxy, i-propoxy, —OCHF2, —OCF3, —OCHFCH2F, —OCF2CHF2, —OCH2CF3, —OCH2CF2CHF2, methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, 2-hydroxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuranyl, piperidyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, thienyl, thiazolyl, oxazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazyl, benzimidazolyl, indolyl or quinolyl. In some embodiments, R2is H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, C1-C4haloalkoxy or hydroxy-substituted C1-C4alkyl. In other embodiments, R2is H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, methyl, ethyl, n-propyl, i-propyl, —CF3, —CH2CF3, methoxy, ethoxy, n-propoxy or i-propoxy. In some embodiments, each of R3, R4and R5is independently H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, C1-C4haloalkoxy or hydroxy-substituted C1-C4alkyl. In other embodiments, each of R3, R4and R5is independently H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, methyl, ethyl, n-propyl, i-propyl, —CF3, —CH2CF3, methoxy, ethoxy, n-propoxy or i-propoxy. In some embodiments, R7is H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, C1-C4haloalkoxy or hydroxy-substituted C1-C4alkyl. In other embodiments, R7is H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —COOH, —C(═O)NH2, methyl, ethyl, n-propyl, i-propyl, —CF3, —CH2CF3, methoxy, ethoxy, n-propoxy or i-propoxy. In some embodiments, R8is H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —SH, —COOH, —C(═O)NH2, —C(═O)NHCH3, —C(═O)N(CH3)2, —C(═O)—(C1-C4alkyl), —C(═O)—(C1-C4alkoxy), C1-C4alkyl, C2-C4alkenyl, C2-C4alkynyl, C1-C4haloalkyl, C1-C4alkoxy, C1-C4haloalkoxy, C1-C4alkylthio, C1-C4alkylamino, hydroxy-substituted C1-C4alkyl, C3-C6cycloalkyl, 3-6 membered heterocyclyl, C6-C10aryl or 5-10 membered heteroaryl. In other embodiments, wherein R8is independently H, F, Cl, Br, I, —CN, —NO2, —NH2, —OH, —SH, —COOH, —C(═O)NH2, —C(═O)NHCH3, —C(═O)N(CH3)2, —C(═O)—CH3, —C(═O)—OCH3, methyl, ethyl, n-propyl, i-propyl, allyl, propenyl, propargyl, propinyl, —CHF2, —CF3, —CHFCH2F, —CF2CHF2, —CH2CHF2, —CH2CF3, —CH2CF2CHF2, methoxy, ethoxy, n-propoxy, i-propoxy, —OCHF2, —OCF3, —OCHFCH2F, —OCF2CHF2, —OCH2CF3, —OCH2CF2CHF2, methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, 2-hydroxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuranyl, piperidyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, thienyl, thiazolyl, oxazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazyl, benzimidazolyl, indolyl or quinolyl. In some embodiments, provided herein is a compound having Formula (II) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each R1, R2, R3, R4, R5, R6, R7and R8is as defined herein. In other embodiments, provided herein is a compound having Formula (III) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each R1a, R1b, R1c, R1d, R1e, R2, R3, R4, R5, R6, R7and R8is as defined herein. In yet other embodiments, provided herein is a compound having Formula (IVa) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each R1b, R1c, R1d, R1e, R2, R3, R4, R5, R6, R7and R8is as defined herein. In yet other embodiments, provided herein is a compound having Formula (IVb) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each R1a, R1b, R1c, R1d, R1e, R2, R3, R4, R5, R6, R7and R8is as defined herein. In yet other embodiments, provided herein is a compound having Formula (IVc) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each R1a, R1b, R1d, R1e, R2, R3, R4, R5, R6, R7and R8is as defined herein. In yet other embodiments, provided herein is a compound having Formula (V) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each R1b, R1c, R1d, R2, R3, R4, R5, R6, R7and R8is as defined herein. In some embodiments, the compound disclosed herein has one of the following structures or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, but is in no way limited to: In other aspect, provided herein is a pharmaceutical composition comprising the compound of Formula (I), (II), (III), (IVa), (IVb), (IVc) or (V). In some embodiments, the pharmaceutical composition disclosed herein further comprises a pharmaceutically acceptable excipient, a carrier, an adjuvant or a combination thereof. In other aspect, the present invention relates to use of the compound represented by formula (I), (II), (III), (IVa), (IVb), (IVc) or (V) or the pharmaceutical composition in the manufacture of a medicament for preventing, treating or lessening a 5-HT1Freceptor-related disease in a patient. In some embodiments, wherein the 5-HT1Freceptor-related disease is migraine, general pain, trigeminal neuralgia, dental pain or temporomandibular joint dysfunction pain, autism, obsession, phobia, depression, social phobia, anxiety, general anxiety disorder, disorders of sleep, post-traumatic syndrome, chronic fatigue syndrome, premenstrual syndrome or late luteal phase syndrome, borderline personality disorder, disruptive behavior disorders, impulse control disorders, attention deficit hyperactivity disorder, alcoholism, tobacco abuse, mutism, trichotillomania, bulimia, anorexia nervosa, premature ejaculation, erectile dysfunction, memory loss or dementia. In other embodiments, the 5-HT1Freceptor-related disease is migraine. In yet other aspect, the present invention relates to use of the compound represented by formula (I), (II), (III), (IVa), (IVb), (IVc) or (V) or the pharmaceutical composition in the manufacture of a medicament for activating 5-HT1Freceptor. In other aspect, provided herein is a method of preparing, separating or purifying the compound of Formula (I), (II), (III), (IVa), (IVb), (IVc) or (V). Pharmaceutical Composition of the Compound of the Invention and Preparations and Administration The invention provides a pharmaceutical composition containing a compound of formula (I), (II), (III), (IVa), (IVb), (IVc) or (V) or an independent stereoisomer thereof, a racemic mixture or non-racemic mixture of the stereoisomer thereof, or a pharmaceutically acceptable salt or solvent thereof. In one embodiment of the invention, the pharmaceutical composition further comprises at least one pharmaceutically acceptable carrier, adjuvant or excipient, and optionally other treating and/or preventing ingredients. The dosage form of the compound used in the method of the invention can be determined by the selected compound, the type of pharmacokinetic distribution required by the route of administration and the state of the patient. A formulation suitable for oral, sublingual, intranasal or injection administration is prepared according to a well-known method in the pharmaceutical field, and the formulation contains at least one active compound. See, for example, REMINGTON'S PHARMACEUTICAL SCIENCES (16th ed. 1980). Generally speaking, the formulation of the present invention includes an active ingredient (a compound of formula (I), (II), (III), (IVa), (IVb), (IVc) or (V)), and is usually mixed with excipients, diluted with excipients or encapsulated in carriers that may be in the form of capsules, small capsules, paper or other containers. When an excipient is used as a diluent, it may be a solid, semi-solid or liquid material, acting as an excipient, carrier or medium for the active component. Therefore, the formulations may be tablets, pills, powders, lozenges, sachets, flat capsules, elixir, suspension, emulsion, solution, syrup, aerosol (solid or liquid medium), ointments containing an active compound up to 10 wt %, soft and hard capsules, gelatin, suppository, sterile injection and aseptic packaging powder. Before mixing with other components in preparation process, the active compound may need to be ground to provide appropriate particle size. If the active compound is insoluble, it is usually ground to a size of less than 200 mesh. If the active compound is basically water-soluble, its particle size is adjusted by grinding, so that the compound has an uniform particle size distribution in the formulation, for example, about 40 meshes. In one embodiment of the present invention, the particle size is about 0.1-100 μm. A suitable carrier, adjuvant or excipient is well known for the technical personnel in the field and was described in detail in Ansel H. C. et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems (2004) Lippincott, Williams & Wilkins, Philadelphia; Gennaro A. R. et al., Remington: The Science and Practice of Pharmacy (2000) Lippincott, Williams & Wilkins, Philadelphia; and Rowe R. C., Handbook of Pharmaceutical Excipients (2005) Pharmaceutical Press, Chicago. “Pharmaceutically acceptable excipient” as used herein means a pharmaceutically acceptable material, composition or vehicle involved in giving form or consistency to the pharmaceutical composition. Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled, such that interactions which would substantially reduce the efficacy of the compound of the invention when administered to a patient and/or would result in pharmaceutically unacceptable compositions are avoided. In addition, each excipient must be of sufficiently high purity to render it pharmaceutically acceptable. Suitable pharmaceutically acceptable excipients will vary depending upon the particular dosage form chosen. In addition, suitable pharmaceutically acceptable excipients may be chosen for a particular function that they may serve in the composition. For example, certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms. Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms. Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the carrying or transporting the compound of the present invention once administered to the patient from one organ, or portion of the body, to another organ, or portion of the body. Certain pharmaceutically acceptable excipients may be chosen for their ability to enhance patient compliance. Examples of suitable excipients include lactose, glucose, sucrose, sorbitol, mannitol, starch, Arabic gum, calcium phosphate, alginate, xanthate, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup and methyl cellulose. Suitable pharmaceutically acceptable excipients further include the following types: diluents, fillers, binders, disintegrants, lubricants (such as talc powder, magnesium stearate and mineral oil), glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweetners, flavoring agents, flavor masking agents, coloring agents, anticaking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives (such as methyl hydroxybenzoate and propyl hydroxybenzoate), stabilizers, surfactants, and buffering agents. The skilled artisan will appreciate that certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the formulation and what other ingredients are present in the formulation. The compounds of the invention can be prepared by known methods in the field so that the active components will be released rapidly, continuously or controllably after administration in patients. Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically acceptable excipients in appropriate amounts for use in the invention. In addition, there are a number of resources that are available to the skilled artisan which describe pharmaceutically acceptable excipients and may be useful in selecting suitable pharmaceutically acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press). Pharmaceutically acceptable carriers may be solid or liquid carriers for the preparation of pharmaceutical compositions using compounds described in the present invention. Solid formulations include powders, tablets, dispersible granules, capsules, cachets, and suppositories. Powders and tablets may contain about 5% to about 95% active ingredients. Suitable solid carriers are known in the field, such as magnesium carbonate, magnesium stearate, talc powder, sugar or lactose. Tablets, powders, flat capsules and capsules may be used as solid dosage forms suitable for oral administration. Examples of medicinal carriers and methods for preparing various compositions can be obtained as follows: A. Gennaro (ed.), Remington's Pharmaceutical Sciences, 18thed., 1990, Mack Publishing Company Co., Easton, Pennsylvania In Remington: The Science and Practice of Pharmacy, 21st edition, 2005, ed. D. B. Troy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York disclosed various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof, the contents of each of which is incorporated by reference herein. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention. The pharmaceutical compositions of the invention are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company). Therefore, another aspect of the present invention is related to a method for preparing a pharmaceutical composition, wherein the pharmaceutical composition contains the compound disclosed herein and pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof, and the method comprises mixing various ingredients. The pharmaceutical composition containing the compound disclosed herein can be prepared at for example environment temperature and under barometric pressure. The compound of the invention will typically be formulated into a dosage form adapted for administration to the patient by the desired route of administration. For example, dosage forms include those adapted for (1) oral administration such as tablets, capsules, caplets, pills, troches, powders, syrups, elixers, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols, solutions, and dry powders; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels. It will also be appreciated that certain of the compounds of present invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative. According to the present invention, a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need thereof is capable of providing, directly or indirectly, a compound as described herein, or a metabolite or residue thereof. In one embodiment, the compounds disclosed herein can be prepared to oral administration. In the other embodiment, the compounds disclosed herein can be prepared to inhalation. In the still other embodiment, the compounds disclosed herein can be prepared to nasal administration. In the yet other embodiment, the compounds disclosed herein can be prepared to transdermal administration. In the still yet other embodiments, the compounds disclosed herein can be prepared to topical administration. The pharmaceutical compositions provided herein may be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, enteric-coating tablets, sugar-coated, or film-coated tablets. Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenylsalicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets. The tablet dosage forms may be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges. The pharmaceutical compositions provided herein may be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as the dry-filled capsule (DFC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient. The pharmaceutical compositions provided herein may be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups. An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil Emulsions may include a pharmaceutically acceptable non-aqueous liquids or solvent, emulsifying agent, and preservative. Suspensions may include a pharmaceutically acceptable suspending agent and preservative. Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl) acetal of a lower alkyl aldehyde, e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxy groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, and hydroalcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative. For a liquid dosage form, for example, a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration. The pharmaceutical composition of the invention can be prepared to a dosage form adapted for administration to a patient by inhalation, for example as a dry powder, an aerosol, a suspension, or a solution composition. In one embodiment, the pharmaceutical composition disclosed in the invention is directed to a dosage form adapted for administration to a patient by inhalation as a dry powder. In one embodiment, the pharmaceutical composition disclosed in the invention is directed to a dosage form adapted for administration to a patient by inhalation as a nebulizer. Dry powder compositions for delivery to the lung by inhalation typically comprise a compound disclosed herein as a finely divided powder together with one or more pharmaceutically acceptable excipients as finely divided powders. Pharmaceutically acceptable excipients particularly suited for use in dry powders are known to those skilled in the art and include lactose, starch, mannitol, and mono-, di-, and polysaccharides. The finely divided powder may be prepared by, for example, micronisation and milling. Generally, the size-reduced (e.g., micronised) compound can be defined by a D50value of about 1 to about 10 microns (for example as measured using laser diffraction). Pharmaceutical compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the patient for a prolonged period of time. For example, the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318 (1986). Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. Ointments, creams and gels, may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents. Such bases may thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol Thickening agents and gelling agents which may be used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents. The compounds disclosed herein can also be coupled to soluble polymers as targeted medicament carriers. Such polymers may encompass polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidophenol, polyhydroxyethylaspartamidophenol or polyethylene oxide polylysine substituted by palmitoyl radicals. The compounds may furthermore be coupled to a class of biodegradable polymers which are suitable for achieving controlled release of a medicament, for example polylactic acid, poly-ε-caprolactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihydroxypyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels. The pharmaceutical compositions provided herein may be administered parenterally by injection, infusion, or implantation, for local or systemic administration. Parenteral administration, as used herein, includes intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, and subcutaneous administration. The pharmaceutical compositions provided herein may be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection. Such dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see, Remington: The Science and Practice of Pharmacy, supra). The pharmaceutical compositions intended for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases. The pharmaceutical compositions provided herein can be administered by rectal in suppository form, in which the drug was mixed with suitable non-irritating excipients such as cocoa oil and glycerol ester synthesized by polyethylene glycol, and the mixture was solid at room temperature and can be released when liquefied or dissolved in the rectal cavity. Because of individual differences, the severity of symptoms between individuals will have great difference, and every drug has its unique therapeutic properties. Therefore, the exact way of administration, dosage form and treatment plan for each individual should be determined by a practicing physician. The pharmaceutical compositions provided herein may be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms. While it is possible to administer a compound employed in the methods of this invention directly without any formulation, the compounds are usually administered in the form of pharmaceutical formulations comprising a pharmaceutically acceptable excipient and at least one active ingredient. These formulations can be administered by a variety of routes including oral, buccal, rectal, intranasal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. Many of the compounds employed in the methods of this invention are effective as both injectable and oral compositions. In order to administer transdermally, a transdermal delivery device (“patch”) is needed. The transdermal patch can be used to continuously or intermittently deliver the controllable amount of the compound of the invention. The structure and application of transdermal patches for drug delivery are well known in the field. See, e.g., U.S. Pat. No. 5,023,252. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. Frequently, it will be desirable or necessary to introduce the pharmaceutical composition to the brain, either directly or indirectly. Direct techniques usually involve placement of a drug delivery catheter into the host's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of biological factors to specific anatomical regions of the body, is described in U.S. Pat. No. 5,011,472. The delivery of hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic solutions which can transiently open the blood-brain barrier. In one preferred embodiment of the present invention, there is provided a pharmaceutical formulation comprising at least one active compound as described above in a formulation adapted for buccal and/or sublingual, or nasal administration. This embodiment provides administration of the active compound in a manner that avoids gastric complications, such as first bypassing gastric system metabolism and/or first undergoing liver metabolism. This administration route may also reduce adsorption times, providing more rapid onset of therapeutic benefit. The compounds of the present invention may provide particularly favorable solubility distributions to facilitate sublingual/buccal formulations. Such formulations typically require relatively high concentrations of active ingredients to deliver sufficient amounts of active ingredients to the limited surface area of the sublingual/buccal mucosa for the relatively short durations of contact with sublingual/buccal mucosal surface, to allow the absorption of the active ingredient. Thus, the very high activity of the compounds of the present invention and their high solubilities facilitate their suitability for sublingual/buccal formulation. The term “therapeutically effective amount,” as used herein, refers to the total amount of each active component that is sufficient to show an useful treatment effect. For example, the drug amount of administration or balance in the body sufficient to treat, cure, or alleviate symptoms of a disease. The effective amount required for a special treatment depends on a variety of factors, including diseases, the severity of the disease, the activity of the used specific drug, the mode of administration, the clearance rate of the specific drug, the duration of therapy, the combination of drugs, age, weight, gender, diet and patient's health, and so on. The description of other factors that need to be considered for “therapeutically effective amount” in this field can be found in Gilman et al., eds., Goodman And Gilman's: The Pharmacological Bases of Therapeutics, 8thed., Pergamon Press, 1990; Remington's Pharmaceutical Sciences, 17thed., Mack Publishing Company, Easton, Pa., 1990. A compound of formula (I) is preferably formulated in an unit dosage form, each dosage containing from about 0.001 to about 100 mg, more usually about 1.0 to about 30 mg, of the active ingredient. The term “unit dosage form” refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient as described above. The compounds are generally effective over a wide dosage range. For example, dosages per day normally fall within the range of about 0.0001 to about 30 mg/kg of body weight. In the treatment of adult humans, the range of about 0.1 to about 15 mg/kg/day, in single or divided dose, is especially preferred. However, it will be understood that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound or compounds administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms, and therefore the above dosage ranges are not intended to limit the scope of the invention in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, provided that such larger doses are first divided into several smaller doses for administration throughout the day. The term “administration” refers to provision of a therapeutically effective amount of medicine to an individual by oral, sublingual, intravenous, subcutaneous, percutaneous, intramuscular, intradermal, intrathecal, epidural, intraocular, intracranial, inhalation, rectal, vagina, etc. The pharmaceutical dosage forms include plaster, lotion, tablet, capsule, pill, dispersible powder, granule, suppository, sublimed preparation, lozenge, injection, aseptic solution or non-aqueous solution, suspension, emulsion, paster, etc. An active component is complexed with a non-toxic pharmaceutically acceptable carrier (such as glucose, lactose, gum arabic, gelatin, mannitol, starch paste, magnesium trisilicate, talcum powder, corn starch, keratin, silica gel, potato starch, urea, dextran, etc.). The preferred route of administration varies with clinical characteristics. Dose changes must depend on situation of patients receiving treatment. Doctors will determine the appropriate dose according to individual status of patients. The therapeutically effective amount per unit dose depends on body weight, physiological function and the selected vaccination program. An amount of compound per unit dose refers to the weight of the compound per each administration, excluding weight of carriers (the drug formulation contains carriers). The pharmaceutical compositions provided herein may be formulated for single or multiple dosage administration. The single dosage formulations are packaged in an ampoule, a vial, or a syringe. The multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art. The pharmaceutical compositions provided herein may be co-formulated with other active ingredients which do not impair the desired therapeutic action, or with substances that supplement the desired action. In one embodiment, the therapeutic methods disclosed herein comprise administrating to a patient in need of the treatment a safe and effective amount of the compound of the invention or the pharmaceutical composition containing the compound of the invention. Each example disclosed herein comprises the method of treating the diseases comprising administrating to a patient in need of the treatment a safe and effective amount of the compound of the invention or the pharmaceutical composition containing the compound of the invention. In one embodiment, the compound of the invention or the pharmaceutical composition thereof may be administered by any suitable route of administration, including both systemic administration and topical administration. Systemic administration includes oral administration, parenteral administration, transdermal administration and rectal administration. Parenteral administration refers to routes of administration other than enteral or transdermal, and is typically by injection or infusion. Parenteral administration includes intravenous, intramuscular, and subcutaneous injection or infusion. Topical administration includes application to the skin as well as intraocular, otic, intravaginal, inhaled and intranasal administration. In one embodiment, the compound of the invention or the pharmaceutical composition thereof may be administered orally. In another embodiment, the compound of the invention or the pharmaceutical composition thereof may be administered by inhalation. In still one embodiment, the compound of the invention or the pharmaceutical composition thereof may be administered intranasally. In one embodiment, the compound of the invention or the pharmaceutical composition thereof may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered once, twice, three, or four times per day. In one embodiment, a dose is administered once per day. In a further embodiment, a dose is administered twice per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for the compound of the invention or the pharmaceutical composition thereof depend on the pharmacokinetic properties of that compound, such as absorption, distribution, and half-life, which can be determined by the skilled artisan. In addition, suitable dosing regimens, including the duration of implementation of such regimens, for the compound of the invention or the pharmaceutical composition thereof depend on the disorder being treated, the severity of the disorder being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and the like within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change. The compounds of the present invention may be administered either simultaneously with, or before or after, one or more other therapeutic agents. The compounds of the present invention may be administered separately, by the same or different route of administration, or together in the same pharmaceutical composition as the other agents. This is chosen by the technical personnel in the field according to the actual conditions of the patient's health, age, weight and so on. If formulated as a fixed dose, such combination products employ the compounds of this invention within the dosage range described herein and the other pharmaceutically active agent within its dosage range. Accordingly, in an aspect, this invention includes combinations comprising an amount of at least one compound of the invention, or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, and an effective amount of one or more additional agents described above. Additionally, the compounds of the invention may be administered as prodrugs. As used herein, a “prodrug” of a compound of the invention is a functional derivative of the compound which, upon administration to a patient, eventually liberates the compound of the invention in vivo. Administration of a compound of the invention as a prodrug may enable the skilled artisan to do one or more of the following: (a) modify the onset of action of the compound in vivo; (b) modify the duration of action of the compound in vivo; (c) modify the transportation or distribution of the compound in vivo; (d) modify the solubility of the compound in vivo; and (e) overcome a side effect or other difficulty encountered with the compound. Typical functional derivatives used to prepare prodrugs include modifications of the compound that are chemically or enzymatically cleaved in vivo. Such modifications, which include the preparation of phosphates, amides, esters, thioesters, carbonates, and carbamates, are well known to those skilled in the art. Use of the Compounds and Pharmaceutical Compositions The compounds and pharmaceutical compositions provided by the invention can be used to prepare a medicament for activating 5-HT1Freceptors, and also to prepare a medicament for preventing, treating or alleviating a 5-HT1Freceptor-related disease, especially migraine. Specifically, the amount of the compound or the compound of the pharmaceutical composition of the present invention can effectively, detectably and selectively activate 5-HT1Freceptor. Specifically, the amount of the compound or the compound of the pharmaceutical composition of the present invention can effectively, detectably and selectively inhibit neuronal protein extravasation. Compounds disclosed herein would be useful for, but are in no way limited to, the prevention or treatment or alleviation of a 5-HT1Freceptor-related disease in a patient by administering to the patient a compound or a composition disclosed herein in an effective amount. The 5-HT1Freceptor-related diseases further include, but are not limited to, migraine, general pain, trigeminal neuralgia, dental pain or temporomandibular joint dysfunction pain, autism, obsession, phobia, depression, social phobia, anxiety, general anxiety disorder, disorders of sleep, post-traumatic syndrome, chronic fatigue syndrome, premenstrual syndrome or late luteal phase syndrome, borderline personality disorder, disruptive behavior disorders, impulse control disorders, attention deficit hyperactivity disorder, alcoholism, tobacco abuse, mutism, trichotillomania, bulimia, anorexia nervosa, premature ejaculation, erectile dysfunction, memory loss and dementia. Besides being useful for human treatment, the compounds and pharmaceutical compositions of the present invention are also useful for veterinary treatment of animals such as companion animals, exotic animals and farm animals. In other embodiments, the animals disclosed herein include horses, dogs, and cats. As used herein, the compounds disclosed herein include the pharmaceutically acceptable derivatives thereof. General Synthetic Procedures The following examples are provided so that the invention might be more fully understood. However, it should be understood that these embodiments merely provide a method of practicing the present invention, and the present invention is not limited to these embodiments. Generally, the compounds disclosed herein may be prepared by methods described herein, wherein the substituents are as defined for Formula (I), (II), (III), (IVa), (IVb), (IVc) or (V) above, except where further noted. The following non-limiting schemes and examples are presented to further exemplify the invention. Persons skilled in the art will recognize that the chemical reactions described may be readily adapted to prepare a number of other compounds disclosed herein, and alternative methods for preparing the compounds disclosed herein are deemed to be within the scope disclosed herein. For example, the synthesis of non-exemplified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known reaction conditions in the art will be recognized as having applicability for preparing other compounds disclosed herein. In the examples described below, unless otherwise indicated all temperatures are set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company, and were used without further purification unless otherwise indicated. Common solvents were purchased from commercial suppliers such as Shantou XiLong Chemical Factory, Guangdong Guanghua Reagent Chemical Factory Co. Ltd., Guangzhou Reagent Chemical Factory, Tianjin YuYu Fine Chemical Ltd., Tianjin Fuchen Chemical Reagent Factory, Wuhan XinHuaYuan Technology Development Co. Ltd., Qingdao Tenglong Reagent Chemical Ltd., and Qingdao Ocean Chemical Factory. Anhydrous THF, dioxane, toluene, and ether were obtained by refluxing the solvent with sodium. Anhydrous CH2Cl2and CHCl3were obtained by refluxing the solvent with CaH2. EtOAc, PE, hexane, DMAC and DMF were treated with anhydrous sodium sulfate prior to use. The reactions set forth below were done generally under a positive pressure of nitrogen or argon or with a drying tube (unless otherwise stated) in anhydrous solvents, and the reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried. Column chromatography was conducted using a silica gel column. Silica gel (300-400 mesh) was purchased from Qingdao Ocean Chemical Factory. 1H NMR spectra were recorded by Bruker 400 MHz or 600 MHz NMR spectrometer.1H NMR spectra were obtained by using CDCl3, DMSO-d6, CD3OD or acetone-d6solutions (in ppm), with TMS (0 ppm) or chloroform (7.26 ppm) as the reference standard. When peak multiplicities are reported, the following abbreviations are used: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broadened), brs (broadened singlet), dd (doublet of doublets), ddd (doublet of doublet of doublets), dt (doublet of triplets), td (triplet of doublets), tt (triplet of triplets). Coupling constants J, when given, were reported in Hertz (Hz). Low resolution mass spectrum (MS) data measurement condition: Agilent 6120 Quadrupole HPLC-M (column type: Zorbax SB-C18, 2.1×30 mm, 3.5 micron, 6 min, flow rate 0.6 mL/min. Mobile phase: in the proportion of 5%-95% (CH3CN containing 0.1% of formic acid) in (H2O containing 0.1% of formic acid), using electrospray ionization (ESI), UV detection, at 210 nm/254 nm. Pure compound was detected by Agilent 1260 pre-HPLC or Calesep pump 250 pre-HPLC (NOVASEP 50/80 mm DAC) with UV detection at 210 nm/254 nm. The following abbreviations are used throughout the specification: CH2Cl2, DCMdichloromethaneCDCl3deuterochloroformDMSOdimethyl sulfoxideDMSO-d6deuterated dimethyl sulfoxideEtOAc, EAethyl acetateCH3OH, MeOHmethanolCD3ODdeuterated methanolnMnanomole per literμMmicromole per litermMmillimole per literMmole per literngnanogramμgmicrogrammeBoc, BOCtert-butoxycarbonylmgmilligramggrammL, mlmilliliterμL, μlmicroliternL, nlnanoliterminminutehhourPEpetroleum ether (60-90° C.)RT, rt, r.t.room temperatureEDTA-K2dipotassium ethylenediaminetetraacetateSolutolpolyoxyl-15-hydroxystearateMTBEtert-butyl methyl etherPEG400polyethylene glycol 400EtCH3CH2, ethyl The following synthetic schemes describe the steps for preparing the compounds disclosed herein, unless otherwise specified, wherein each R1a, R1b, R1c, R1dand R1eis as defined herein. Wherein X is CR1aor N; R0is alkyl, cycloalkyl or haloalkyl. A compound of formula (8) can be prepared through the following process: a compound of formula (1) can react with triethyl phosphite to afford a compound of formula (2); and then the compound of formula (2) can be condensed with a compound of formula (3) to get a compound of formula (4). The compound of formula (4) can react with a compound of formula (5) to get a compound of formula (6). The compound of formula (6) can convert to a compound of formula (7) through de-protection; the compound of formula (7) can convert to a compound of formula (8) by Borch reductive amination with corresponding aldehyde or ketal in the presence of a reductant (e.g., sodium cyanoborohydride). Wherein X is CR1aor N; R0is alkyl, cycloalkyl or haloalkyl. A compound of formula (15) can be prepared through the following process: a compound of formula (9) can react with diethyl phosphite to afford a compound of formula (10); the compound of formula (10) can be fluorinated to get a compound of formula (11); and then the compound of formula (11) can be condensed with a compound of formula (3) to get a compound of formula (12). The compound of formula (12) can react with a compound of formula (5) to get a compound of formula (13). The compound of formula (13) can convert to a compound of formula (14) through de-protection; the compound of formula (14) can convert to a compound of formula (15) by Borch reductive amination with corresponding aldehyde or ketal in the presence of a reductant (e.g., sodium cyanoborohydride). Wherein X is CR1aor N; R0is alkyl, cycloalkyl or haloalkyl. A compound of formula (8) can be prepared through the following process: a compound of formula (4) can convert to a compound of formula (16) through de-protection; the compound of formula (16) can convert to a compound of formula (17) by nucleophilic substitution reaction with corresponding alkyl derivative or cycloalkyl derivative; and then the compound of formula (17) can be condensed with a compound of formula (5) to get a compound of formula (8). Wherein X is CR1aor N; R0is alkyl, cycloalkyl or haloalkyl. A compound of formula (15) can be prepared through the following process: a compound of formula (12) can convert to a compound of formula (18) through de-protection; the compound of formula (18) can convert to a compound of formula (19) by nucleophilic substitution reaction with corresponding alkyl derivative or naphthenic derivative; and then the compound of formula (19) can be condensed with a compound of formula (5) to get a compound of formula (15). The following examples are provided to further illustrate the compounds, pharmaceutical compositions and their applications thereof. EXAMPLE Example 1: Synthesis of 4-fluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide Step 1) Synthesis of diethyl((6-bromopyridin-2-yl)methyl)phosphonate 2-Bromo-6-(bromomethyl)pyridine (5.0 g, 19.9 mmol) and triethyl phosphite (6.0 mL, 35.0 mmol) were added into a 100 mL single-neck round bottom flask, and the mixture was stirred at 140° C. for 12 hours. After the reaction was completed, the mixture was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate (v/v)=5/1) to get the title compound as light yellow oil (4.91 g, 80.0%). MS (ESI, pos. ion) m/z: 308.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.49 (t, J=7.6 Hz, 1H), 7.37-7.33 (m, 2H), 4.13-4.04 (m, 4H), 3.37 (d, J=22.0 Hz, 2H), 1.28 (t, J=7.2 Hz, 6H). Step 2) Synthesis of tert-butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate Under 0° C., diethyl((6-bromopyridin-2-yl)methyl)phosphonate (446 mg, 1.45 mmol), tert-butyl 4-oxopiperidine-1-carboxylate (0.6 g, 3.0 mmol) and tetrahydrofuran (10 mL) were added into a 100 mL single-neck round bottom flask, and then sodium hydride (70 mg, 1.75 mmol) was added. After the mixture was stirred for 15 min, the mixture was further stirred at 25° C. for 5 hours. After the reaction was stopped stirring, water (20 mL) was added, and the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate (v/v)=5/1) to get the title compound as colorless oil (500 mg, 97.8%). MS (ESI, pos. ion) m/z: 353.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.46 (t, J=8.0 Hz, 1H), 7.28 (d, J=6.8 Hz, 1H), 7.08 (d, J=7.6 Hz, 1H), 6.29 (s, 1H), 3.55-3.50 (m, 2H), 3.49-3.44 (m, 2H), 2.85 (brs, 2H), 2.35 (t, J=5.6 Hz, 2H), 1.48 (s, 9H). Step 3) Synthesis of tert-butyl 4-((6-(4-fluorobenzamide)pyridin-2-yl)methylene)piperidine-1-carboxylate tert-Butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate (500 mg, 1.42 mmol), 4-fluorobenzamide (348 mg, 2.50 mmol), potassium carbonate (1.38 g, 9.98 mmol), cuprous iodide (280 mg, 1.47 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (122 mg, 0.86 mmol), water (1.3 mL) and toluene (10 mL) were added into a 100 mL single-neck round bottom flask, and the mixture was stirred at 115° C. under N2protection for 12 hours. After the reaction was stopped stirring, water (20 mL) was added, and the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (1.5 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate (v/v)=4/1) to get the title compound as light yellow oil (420 mg, 72.1%). MS (ESI, pos. ion) m/z: 412.3 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.46 (s, 1H), 8.15 (d, J=8.4 Hz, 1H), 7.95-7.88 (m, 2H), 7.69 (t, J=8.0 Hz, 1H), 7.16 (t, J=8.4 Hz, 2H), 6.94 (d, J=7.6 Hz, 1H), 6.27 (s, 1H), 3.52 (t, J=5.6 Hz, 2H), 3.43 (t, J=5.6 Hz, 2H), 2.77 (t, J=5.6 Hz, 2H), 2.35 (t, J=5.6 Hz, 2H), 1.46 (s, 9H). Step 4) Synthesis of 4-fluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide Under 25° C., tert-butyl 4-((6-(4-fluorobenzamide)pyridin-2-yl)methylene)piperidine-1-carboxylate (410 mg, 1.0 mmol) and methanol (5 mL) were added into a 50 mL single-neck round bottom flask, and then hydrogen chloride ethyl acetate solution (2 M, 2 mL) was added. The mixture was further stirred for 2 hours. After the reaction was stopped stirring, the mixture was concentrated using a rotary evaporator under reduced pressure, and then saturated sodium bicarbonate aqueous solution (20 mL) was added. The resulting mixture was extracted with dichloromethane (20 mL) and then was partitioned. The organic layer was dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol (v/v)=20/1) to get the title compound as a light yellow solid (200 mg, 64.9%). MS (ESI, pos. ion) m/z: 312.1 [M+H]+; 1H NMR (400 MHz, CD3OD) δ (ppm) 8.03-7.94 (m, 3H), 7.75 (t, J=8.0 Hz, 1H), 7.23 (t, J=8.8 Hz, 2H), 7.03 (d, J=7.6 Hz, 1H), 6.42 (s, 1H), 3.36-3.18 (m, 8H). Step 5) Synthesis of 4-fluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide 4-Fluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide (200 mg, 0.64 mmol) and methanol (5 mL) were added into a 50 mL single-neck round bottom flask, and two drops of acetic acid were added. Under 0° C., formaldehyde (40%, 0.17 mL, 2.2 mmol) was added, and then sodium cyanoborohydride (122 mg, 1.93 mmol) was added into the reaction mixture in portions. After stirring for 10 min, the mixture was warmed to 25° C. and further stirred for 5 hours. The reaction was quenched with saturated sodium bicarbonate aqueous solution (10 mL), then the resulting mixture was extracted with dichloromethane (20 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure, and the residue was purified by column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a white solid (163 mg, 78.0%). MS (ESI, pos. ion) m/z: 326.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.18 (d, J=8.4 Hz, 1H), 7.97 (dd, J=8.4, 5.2 Hz, 2H), 7.71 (t, J=8.0 Hz, 1H), 7.17 (t, J=8.4 Hz, 2H), 6.91 (d, J=7.6 Hz, 1H), 6.33 (s, 1H), 3.32 (brs, 2H), 3.25-3.04 (m, 6H), 2.77 (s, 3H). Example 2: Synthesis of 4-chloro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide Step 1) Synthesis of tert-butyl 4-((6-(4-chlorobenzamide)pyridin-2-yl)methylene)piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate (500 mg, 1.42 mmol), 4-chlorobenzamide (344 mg, 2.21 mmol), potassium carbonate (1.38 g, 9.98 mmol), cuprous iodide (280 mg, 1.47 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (122 mg, 0.86 mmol), water (1.3 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=4/1) to get the title compound as a white solid (420 mg, 69.3%). MS (ESI, pos. ion) m/z: 428.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.43 (s, 1H), 8.16 (d, J=8.0 Hz, 1H), 7.85 (d, J=8.4 Hz, 2H), 7.70 (t, J=8.0 Hz, 1H), 7.47 (d, J=8.8 Hz, 2H), 6.95 (d, J=7.6 Hz, 1H), 6.29 (s, 1H), 3.53 (t, J=5.6 Hz, 2H), 3.45 (t, J=5.6 Hz, 2H), 2.78 (t, J=5.6 Hz, 2H), 2.36 (t, J=5.6 Hz, 2H), 1.49 (s, 9H). Step 2) Synthesis of 4-chloro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-(4-chlorobenzamide)pyridin-2-yl)methylene)piperidine-1-carboxylate (420 mg, 0.98 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) was reacted in methanol (5 mL) to prepared it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (220 mg, 68.6%). MS (ESI, pos. ion) m/z: 328.2 [M+H]+; 1H NMR (400 MHz, CD3OD) δ (ppm) 7.97 (d, J=8.4 Hz, 1H), 7.92 (d, J=8.8 Hz, 2H), 7.73 (t, J=8.0 Hz, 1H), 7.50 (d, J=8.4 Hz, 2H), 7.01 (d, J=7.6 Hz, 1H), 6.30 (s, 1H), 3.06-2.98 (m, 4H), 2.96 (d, J=5.6 Hz, 2H), 2.43 (t, J=5.6 Hz, 2H). Step 3) Synthesis of 4-chloro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 4-chloro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide (220 mg, 0.67 mmol), sodium cyanoborohydride (131 mg, 2.18 mmol) and formaldehyde (40%, 0.1 mL, 1.38 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=30/1) to give the title compound as a white solid (228 mg, 99.4%). MS (ESI, pos. ion) m/z: 342.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.18 (d, J=8.0 Hz, 1H), 7.89 (d, J=8.4 Hz, 2H), 7.71 (t, J=8.0 Hz, 1H), 7.47 (d, J=8.4 Hz, 2H), 6.93 (d, J=7.6 Hz, 1H), 6.31 (s, 1H), 3.18 (br, 2H), 3.01-2.89 (m, 4H), 2.68-2.62 (m, 2H), 2.61 (s, 3H). Example 3: Synthesis of 2,4-difluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridine-2-yl)benzamide Step 1) Synthesis of tert-butyl 4-((6-(2,4-difluorobenzamide)pyridin-2-yl)methylene)piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate (500 mg, 1.42 mmol), 2,4-difluorobenzamide (363 mg, 2.31 mmol), potassium carbonate (1.38 g, 9.98 mmol), cuprous iodide (280 mg, 1.47 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (122 mg, 0.86 mmol), water (1.3 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=4/1) to get the title compound as a white solid (501 mg, 82.2%). MS (ESI, pos. ion) m/z: 430.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.89 (d, J=13.2 Hz, 1H), 8.23-8.12 (m, 2H), 7.69 (t, J=8.0 Hz, 1H), 7.08-7.02 (m, 1H), 6.99-6.91 (m, 2H), 6.30 (s, 1H), 3.54 (t, J=5.6 Hz, 2H), 3.47 (t, J=5.6 Hz, 2H), 2.85 (t, J=5.6 Hz, 2H), 2.37 (t, J=5.6 Hz, 2H), 1.48 (s, 9H). Step 2) Synthesis of 2,4-difluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-(2,4-fluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (481 mg, 1.12 mmol) and hydrogen chloride ethyl acetate solution (2 M, 4 mL) was reacted in methanol (5 mL) to prepared it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (315 mg, 85.4%). MS (ESI, pos. ion) m/z: 330.0 [M+H]+; 1H NMR (400 MHz, CD3OD) δ (ppm) 8.02 (d, J=8.0 Hz, 1H), 7.91-7.83 (m, 1H), 7.76 (t, J=8.0 Hz, 1H), 7.17-7.08 (m, 2H), 7.04 (d, J=7.6 Hz, 1H), 6.39 (s, 1H), 3.27-3.23 (m, 2H), 3.22-3.18 (m, 2H), 3.17-3.11 (m, 2H), 2.55 (t, J=5.6 Hz, 2H). Step 3) Synthesis of 2,4-difluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 2,4-difluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide (295 mg, 0.90 mmol), sodium cyanoborohydride (177 mg, 2.81 mmol) and formaldehyde (40%, 0.13 mL, 1.82 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=30/1) to give the title compound as a white solid (136 mg, 44.2%). MS (ESI, pos. ion) m/z: 344.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.21-8.10 (m, 2H), 7.71 (t, J=8.0 Hz, 1H), 7.09-7.01 (m, 1H), 6.99-6.90 (m, 2H), 6.33 (s, 1H), 3.26 (br, 2H), 3.13-2.99 (m, 6H), 2.67 (s, 3H). Example 4: Synthesis of 4-chloro-2-fluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl) pyridine-2-yl)benzamide Step 1) Synthesis of tert-butyl 4-((6-(4-chloro-2-fluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate (500 mg, 1.42 mmol), 4-chloro-2-fluoro-benzamide (344 mg, 1.98 mmol), potassium carbonate (1.38 g, 9.98 mmol), cuprous iodide (280 mg, 1.47 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (122 mg, 0.86 mmol), water (1.3 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=4/1) to get the title compound as a light yellow solid (470 mg, 74.5%). MS (ESI, pos. ion) m/z: 446.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.90 (d, J=12.8 Hz, 1H), 8.15 (d, J=8.4 Hz, 1H), 8.11 (t, J=8.8 Hz, 1H), 7.70 (t, J=8.0 Hz, 1H), 7.32 (dd, J=8.4, 1.6 Hz, 1H), 7.23 (d, J=1.6 Hz, 1H), 6.96 (d, J=7.6 Hz, 1H), 6.30 (s, 1H), 3.54 (t, J=5.6 Hz, 2H), 3.47 (t, J=5.6 Hz, 2H), 2.85 (t, J=5.6 Hz, 2H), 2.37 (t, J=5.6 Hz, 2H), 1.48 (s, 9H). Step 2) Synthesis of 4-chloro-2-fluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-(4-chloro-2-fluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (453 mg, 1.02 mmol) and hydrogen chloride ethyl acetate solution (2 M, 4 mL) was reacted in methanol (5 mL) to prepared it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (345 mg, 98.2%). MS (ESI, pos. ion) m/z: 346.2 [M+H]+; 1H NMR (600 MHz, CD3OD) δ (ppm) 8.00 (d, J=8.0 Hz, 1H), 7.81-7.75 (m, 1H), 7.75-7.69 (m, 1H), 7.37-7.30 (m, 2H), 7.03-6.98 (m, 1H), 6.31 (s, 1H), 3.15-3.03 (m, 6H), 2.48 (brs, 2H). Step 3) Synthesis of 4-chloro-2-fluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 4-chloro-2-fluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl) benzamide (336 mg, 0.97 mmol), sodium cyanoborohydride (147 mg, 2.34 mmol) and formaldehyde (40%, 0.19 mL, 2.72 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=30/1) to give the title compound as a white solid (182 mg, 52.1%). MS (ESI, pos. ion) m/z: 360.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.15 (d, J=8.0 Hz, 1H), 8.07 (t, J=8.4 Hz, 1H), 7.69 (t, J=8.0 Hz, 1H), 7.30 (dd, J=8.4, 1.6 Hz, 1H), 7.23 (dd, J=11.6, 1.6 Hz, 1H), 6.93 (d, J=7.6 Hz, 1H), 6.29 (s, 1H), 3.20 (t, J=5.6 Hz, 2H), 2.98-2.93 (m, 2H), 2.88 (t, J=5.6 Hz, 2H), 2.68-2.63 (m, 2H), 2.58 (s, 3H). Example 5: Synthesis of 2,4,6-trifluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridine-2-yl)benzamide Step 1) Synthesis of tert-butyl 4-((6-(2,4,6-trifluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate (500 mg, 1.42 mmol), 2,4,6-trifluorobenzamide (390 mg, 2.23 mmol), potassium carbonate (1.38 g, 9.98 mmol), cuprous iodide (280 mg, 1.47 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (122 mg, 0.86 mmol), water (1.3 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=4/1) to get the title compound as a light yellow solid (241 mg, 38%). MS (ESI, pos. ion) m/z: 448.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.15 (d, J=8.4 Hz, 1H), 7.74-7.69 (m, 1H), 6.96 (d, J=7.6 Hz, 1H), 6.77 (t, J=8.0 Hz, 2H), 6.24 (s, 1H), 3.56-3.50 (m, 2H), 3.52 (t, J=5.6 Hz, 2H), 2.80 (t, J=5.6 Hz, 2H), 2.36 (t, J=5.6 Hz, 2H), 1.47 (s, 9H). Step 2) Synthesis of 2,4,6-trifluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-(2,4,6-trifluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (825 mg, 1.84 mmol) and hydrogen chloride ethyl acetate solution (2 M, 4 mL) was reacted in methanol (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (490 mg, 76.5%). MS (ESI, pos. ion) m/z: 348.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.19 (d, J=8.2 Hz, 1H), 7.78-7.71 (m, 1H), 7.05 (d, J=7.6 Hz, 1H), 7.01-6.75 (m, 2H), 6.29 (s, 1H), 3.31-3.12 (m, 4H), 2.70 (brs, 2H). Step 3) Synthesis of 2,4,6-trifluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 2,4,6-trifluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)benzamide (471 mg, 1.36 mmol), sodium cyanoborohydride (246 mg, 3.9 mmol) and formaldehyde (40%, 0.19 mL, 2.72 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=30/1) to give the title compound as a white solid (427 mg, 87.1%). MS (ESI, pos. ion) m/z: 362.3 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.15 (d, J=8.4 Hz, 1H), 7.71 (t, J=8.0 Hz, 1H), 6.92 (d, J=7.2 Hz, 1H), 6.75 (t, J=8.4 Hz, 2H), 6.27 (s, 1H), 3.06-2.96 (m, 6H), 2.76-2.70 (m, 2H), 2.67 (s, 3H). Example 6: Synthesis of 5-fluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) picolinamide Step 1) Synthesis of tert-butyl 4-((6-(5-fluoropicolinamide)pyridin-2-yl)methylene)piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate (500 mg, 1.42 mmol), 5-fluoro-picolinamide (439 mg, 3.13 mmol), potassium carbonate (1.38 g, 9.98 mmol), cuprous iodide (280 mg, 1.47 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (122 mg, 0.86 mmol), water (1.3 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=4/1) to get the title compound as a light yellow solid (372 mg, 63.4%). MS (ESI, pos. ion) m/z: 413.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 10.23 (s, 1H), 8.48 (d, J=2.4 Hz, 1H), 8.33 (dd, J=8.4, 4.4 Hz, 1H), 8.20 (d, J=8.4 Hz, 1H), 7.70 (t, J=8.0 Hz, 1H), 7.59 (td, J=8.4, 2.8 Hz, 1H), 6.95 (d, J=7.6 Hz, 1H), 6.32 (s, 1H), 3.54 (t, J=5.6 Hz, 2H), 3.48 (t, J=5.6 Hz, 2H), 2.87 (t, J=5.6 Hz, 2H), 2.37 (t, J=5.2 Hz, 2H), 1.48 (s, 9H). Step 2) Synthesis of 5-fluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)picolinamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-(5-fluoropicolinamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (355 mg, 0.86 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) was reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (252 mg, 93.8%). MS (ESI, pos. ion) m/z: 313.1 [M+H]+; 1H NMR (600 MHz, CDCl3) δ (ppm) 8.52 (dd, J=8.4, 4.2 Hz, 1H), 8.42 (d, J=8.4 Hz, 1H), 7.91 (t, J=7.8 Hz, 1H), 7.79 (td, J=8.4, 3.0 Hz, 1H), 7.45 (d, J=1.2 Hz, 1H), 7.11 (d, J=7.8 Hz, 1H), 6.55 (s, 1H), 3.62-3.58 (m, 2H), 3.52 (t, J=5.4 Hz, 2H), 3.49-3.45 (m, 2H), 2.94 (t, J=5.4 Hz, 2H). Step 3) Synthesis of 5-fluoro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) picolinamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 5-fluoro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)picolinamide (242 mg, 0.77 mmol), sodium cyanoborohydride (147 mg, 2.34 mmol) and formaldehyde (40%, 0.16 mL, 2.31 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a white solid (221 mg, 87.4%). MS (ESI, pos. ion) m/z: 327.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.48 (d, J=2.8 Hz, 1H), 8.31 (dd, J=8.7, 4.5 Hz, 1H), 8.21 (d, J=8.4 Hz, 1H), 7.70 (t, J=8.0 Hz, 1H), 7.59 (td, J=8.4, 2.8 Hz, 1H), 6.91 (d, J=7.6 Hz, 1H), 6.35 (s, 1H), 3.49 (brs, 2H), 3.25-3.13 (m, 4H), 2.81 (brs, 2H), 2.79 (s, 3H). Example 7: Synthesis of 5-chloro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) picolinamide Step 1) Synthesis of tert-butyl 4-((6-(5-chloropicolinamide)pyridin-2-yl)methylene)piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate (500 mg, 1.42 mmol), 5-chloro-picolinamide (451 mg, 2.88 mmol), potassium carbonate (1.38 g, 9.98 mmol), cuprous iodide (280 mg, 1.47 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (122 mg, 0.86 mmol), water (1.3 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=4/1) to get the title compound as a light yellow solid (200 mg, 32.9%). MS (ESI, pos. ion) m/z: 429.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 10.25 (s, 1H), 8.59 (d, J=1.6 Hz, 1H), 8.26-8.20 (m, 2H), 7.88 (dd, J=8.4, 2.4 Hz, 1H), 7.71 (t, J=8.0 Hz, 1H), 6.95 (d, J=7.2 Hz, 1H), 6.32 (s, 1H), 3.57-3.51 (m, 2H), 3.48 (t, J=5.6 Hz, 2H), 2.86 (t, J=5.6 Hz, 2H), 2.41-2.34 (m, 2H), 1.48 (s, 9H). Step 2) Synthesis of 5-chloro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)picolinamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-(5-chloro-picolinamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (188 mg, 0.44 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) was reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as alight yellow solid (139 mg, 96.5%). MS (ESI, pos. ion) m/z: 329.1 [M+H]+; 1H NMR (600 MHz, CDCl3) δ (ppm) 10.22 (s, 1H), 8.61 (d, J=2.4 Hz, 1H), 8.25 (d, J=8.4 Hz, 2H), 7.89 (dd, J=8.4, 2.4 Hz, 1H), 7.73 (t, J=7.8 Hz, 1H), 6.94 (d, J=7.8 Hz, 1H), 6.38 (s, 1H), 3.42 (t, J=5.4 Hz, 2H), 3.36 (t, J=5.4 Hz, 2H), 3.31 (t, J=5.4 Hz, 2H), 2.77 (t, J=5.4 Hz, 2H). Step 3) Synthesis of 5-chloro-N-(6-((1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) picolinamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 5-chloro-N-(6-(piperidin-4-ylidenemethyl)pyridin-2-yl)picolinamide (134 mg, 0.41 mmol), sodium cyanoborohydride (79 mg, 1.25 mmol) and formaldehyde (40%, 0.08 mL, 1.2 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a white solid (108 mg, 77.3%). MS (ESI, pos. ion) m/z: 343.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.59 (d, J=2.0 Hz, 1H), 8.22 (t, J=8.8 Hz, 2H), 7.87 (dd, J=8.4, 2.4 Hz, 1H), 7.70 (t, J=8.0 Hz, 1H), 6.93 (d, J=7.6 Hz, 1H), 6.32 (s, 1H), 3.21 (t, J=5.6 Hz, 2H), 2.92 (t, J=5.6 Hz, 2H), 2.86 (t, J=5.6 Hz, 2H), 2.64 (t, J=5.6 Hz, 2H), 2.56 (s, 3H). Example 8: Synthesis of 4-fluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl)pyridine-2-yl)benzamide Step 1) Synthesis of diethyl((6-bromopyridin-2-yl)(hydroxy)methyl)phosphonate 6-Bromopyridylaldehyde (4.0 g, 21.5 mmol) and diethyl phosphite (3.6 mL, 28 mmol) were added into a 100 mL single-neck round bottom flask, and then ethanol (20 mL) and triethylamine (1.5 mL, 11 mmol) were added. The mixture was stirred at 65° C. for 2 hours. After the reaction was completed, the mixture was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate (v/v)=2/1) to get the title compound as a white solid (6.2 g, 89%). MS (ESI, pos. ion) m/z: 324.1 [M+H]+; 1H NMR (600 MHz, CDCl3) δ (ppm) 7.56 (t, J=7.7 Hz, 1H), 7.51 (dd, J=7.5, 1.8 Hz, 1H), 7.41 (d, J=7.6 Hz, 1H), 5.07 (dd, J=12.5, 5.2 Hz, 1H), 4.18-4.13 (m, 2H), 4.08-4.02 (m, 2H), 1.30 (t, J=7.1 Hz, 3H), 1.22 (t, J=7.1 Hz, 3H). Step 2) Synthesis of diethyl ((6-bromopyridin-2-yl)fluoromethyl)phosphonate Under 0° C., diethyl((6-bromopyridin-2-yl)(hydroxy)methyl)phosphonate (3.72 g, 11.5 mmol), pyridine hydrofluoride (1.2 mL) and dichloromethane (30 mL) were added into a 100 mL single-neck round bottom flask, and then diethylaminosulphur trifluoride (2.0 mL, 14.9 mmol) was added dropwise. The mixture was further stirred at 25° C. for 1 hour. After the mixture was stopped stirring, the reaction was quenched with saturated sodium bicarbonate aqueous solution (50 mL). The resulting mixture was separated. The organic layers were collected and dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate (v/v)=2/1) to get the title compound as light yellow oil (1.56 g, 41.7%). MS (ESI, pos. ion) m/z: 326.1 [M+H]+; 1H NMR (600 MHz, CDCl3) δ (ppm) 7.62 (t, J=7.8 Hz, 1H), 7.54 (dd, J=7.6, 1.3 Hz, 1H), 7.46 (d, J=7.9 Hz, 1H), 5.73 (dd, J=44.8, 9.1 Hz, 1H), 4.22 (q, J=7.2 Hz, 2H), 4.14-4.11 (m, 2H), 1.35-1.32 (m, 3H), 1.28 (t, J=7.1 Hz, 3H). Step 3) Synthesis of tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate Under 0° C., diethyl ((6-bromopyridin-2-yl)fluoromethyl)phosphonate (927 mg, 2.84 mmol), tert-butyl 4-oxopiperidine-1-carboxylate (0.8 g, 4.0 mmol) and tetrahydrofuran (10 mL) were added into a 100 mL single-neck round bottom flask, and then sodium hydride (160 mg, 4.0 mmol) was added. The mixture was stirred for 15 min, then the mixture was further stirred at 25° C. for 8 hours. After the mixture was stopped stirring, water (20 mL) was added, then the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate (v/v)=10/1) to get the title compound as a white solid (950 mg, 90%). MS (ESI, pos. ion) m/z: 271.1 [M+H-100]+; 1H NMR (600 MHz, CDCl3) δ (ppm) 7.60 (t, J=7.8 Hz, 1H), 7.47 (d, J=7.6 Hz, 1H), 7.40 (d, J=7.9 Hz, 1H), 3.53 (dd, J=13.1, 6.9 Hz, 4H), 2.98 (br, 2H), 2.56 (br, 2H), 1.51 (s, 9H). Step 4) Synthesis of tert-butyl 4-(fluoro(6-(4-fluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (360 mg, 0.97 mmol), 4-fluorobenzamide (300 mg, 2.2 mmol), potassium carbonate (0.95 g, 6.8 mmol), cuprous iodide (410 mg, 2.2 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.16 mL, 0.94 mmol), water (0.87 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=5/1) to get the title compound as a white solid (330 mg, 79%). MS (ESI, pos. ion) m/z: 430.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.42 (s, 1H), 8.31 (d, J=8.3 Hz, 1H), 7.97 (dd, J=8.6, 5.2 Hz, 2H), 7.83 (t, J=8.0 Hz, 1H), 7.30 (s, 1H), 7.22 (t, J=8.5 Hz, 2H), 3.55 (t, J=5.7 Hz, 2H), 3.52-3.47 (m, 2H), 2.81 (t, J=5.3 Hz, 2H), 2.56 (br, 2H), 1.50 (s, 9H). Step 5) Synthesis of 4-fluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-(fluoro(6-(4-fluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (330 mg, 0.77 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) was reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as alight yellow solid (210 mg, 83%). MS (ESI, pos. ion) m/z: 330.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.49 (s, 1H), 8.30 (d, J=8.3 Hz, 1H), 7.97 (dd, J=8.7, 5.2 Hz, 2H), 7.82 (t, J=8.0 Hz, 1H), 7.27 (d, J=7.3 Hz, 1H), 7.21 (t, J=8.5 Hz, 2H), 3.01 (brs, 2H), 2.95 (brs, 2H), 2.77 (br, 2H), 2.55 (brs, 2H). Step 6) Synthesis of 4-fluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 4-fluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) benzamide (205 mg, 0.62 mmol), sodium cyanoborohydride (120 mg, 1.9 mmol) and formaldehyde (40%, 0.46 mL, 6.2 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as an off-white solid (150 mg, 70.2%). MS (ESI, pos. ion) m/z: 344.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.45 (s, 1H), 8.30 (d, J=8.3 Hz, 1H), 7.97 (dd, J=8.7, 5.2 Hz, 2H), 7.82 (t, J=8.0 Hz, 1H), 7.29 (s, 1H), 7.27 (s, 1H), 7.22 (t, J=8.5 Hz, 2H), 2.85 (t, J=5.3 Hz, 2H), 2.64 (t, J=4.6 Hz, 2H), 2.54 (t, J=5.6 Hz, 2H), 2.47 (t, J=5.3 Hz, 2H), 2.33 (s, 3H). Example 9: Synthesis of 4-chloro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl)pyridine-2-yl)benzamide Step 1) Synthesis of tert-butyl 4-(fluoro(6-(4-chlorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (360 mg, 0.97 mmol), 4-chlorobenzamide (300 mg, 1.9 mmol), potassium carbonate (0.95 g, 6.8 mmol), cuprous iodide (410 mg, 2.2 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.16 mL, 0.94 mmol), water (0.87 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=5/1) to get the title compound as a white solid (220 mg, 51%). MS (ESI, pos. ion) m/z: 446.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.44 (s, 1H), 8.32 (d, J=8.3 Hz, 1H), 7.89 (d, J=8.5 Hz, 2H), 7.84 (t, J=8.0 Hz, 1H), 7.52 (d, J=8.5 Hz, 2H), 7.29 (d, J=8.4 Hz, 1H), 3.55 (t, J=5.7 Hz, 2H), 3.50 (t, J=5.3 Hz, 2H), 2.81 (t, J=5.3 Hz, 2H), 2.57 (brs, 2H), 1.51 (s, 9H). Step 2) Synthesis of 4-chloro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-(fluoro(6-(4-chlorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (200 mg, 0.45 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) was reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as alight yellow solid (140 mg, 90%). MS (ESI, pos. ion) m/z: 346.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.49 (s, 1H), 8.30 (d, J=8.3 Hz, 1H), 7.89 (d, J=8.5 Hz, 2H), 7.83 (t, J=8.0 Hz, 1H), 7.51 (d, J=8.5 Hz, 2H), 7.28 (s, 1H), 3.02 (brs, 2H), 2.97 (brs, 2H), 2.76 (brs, 2H), 2.55 (brs, 2H). Step 3) Synthesis of 4-chloro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 4-chloro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) benzamide (150 mg, 0.43 mmol), sodium cyanoborohydride (90 mg, 1.43 mmol) and formaldehyde (40%, 0.32 mL, 4.3 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (106 mg, 67.9%). MS (ESI, pos. ion) m/z: 360.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.49 (s, 1H), 8.31 (d, J=8.3 Hz, 1H), 7.89 (d, J=8.4 Hz, 2H), 7.83 (t, J=8.0 Hz, 1H), 7.51 (d, J=8.4 Hz, 2H), 7.29 (d, J=3.8 Hz, 1H), 2.85 (t, J=5.2 Hz, 2H), 2.64 (d, J=4.6 Hz, 2H), 2.57-2.52 (m, 2H), 2.48 (t, J=5.1 Hz, 2H), 2.34 (s, 3H). Example 10: Synthesis of 2,4-difluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl) pyridine-2-yl)benzamide Step 1) Synthesis of tert-butyl 4-(fluoro(6-(2,4-difluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (1.0 g, 2.69 mmol), 2,4-difluorobenzamide (700 mg, 4.46 mmol), potassium carbonate (2.6 g, 18.8 mmol), cuprous iodide (400 mg, 2.1 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.3 mL, 1.76 mmol), water (2.5 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=10/1) to get the title compound as a white solid (802 mg, 66%). MS (ESI, pos. ion) m/z: 448.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.89 (d, J=14.4 Hz, 1H), 8.27 (d, J=8.3 Hz, 1H), 8.20 (dd, J=15.5, 9.0 Hz, 1H), 7.81 (t, J=8.0 Hz, 1H), 7.30 (d, J=7.4 Hz, 1H), 7.26 (s, 1H), 7.11-7.02 (m, 1H), 7.00-6.93 (m, 1H), 3.54-3.49 (m, 4H), 2.88 (d, J=5.4 Hz, 2H), 2.54 (brs, 2H), 1.57 (s, 9H). Step 2) Synthesis of 2,4-difluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-(fluoro(6-(2,4-difluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (800 mg, 1.79 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) were reacted in methanol (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as alight yellow solid (310 mg, 49.9%). MS (ESI, pos. ion) m/z: 348.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.91 (d, J=0.8 Hz, 1H), 8.31-8.24 (m, 1H), 8.23-8.15 (m, 1H), 7.85-7.76 (m, 1H), 7.31 (d, J=8.0 Hz, 1H), 7.07 (t, J=8.3 Hz, 1H), 7.01-6.91 (m, 1H), 3.05-2.97 (m, 4H), 2.90 (br, 2H), 2.59 (br, 2H). Step 3) Synthesis of 2,4-difluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 2,4-difluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) benzamide (300 mg, 0.86 mmol), sodium cyanoborohydride (200 mg, 3.17 mmol) and formaldehyde (40%, 0.64 mL, 8.6 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (240 mg, 76.9%). MS (ESI, pos. ion) m/z: 362.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.89 (d, J=13.2 Hz, 1H), 8.26 (d, J=8.3 Hz, 1H), 8.19 (dd, J=15.5, 8.9 Hz, 1H), 7.79 (t, J=8.0 Hz, 1H), 7.29 (d, J=7.8 Hz, 1H), 7.10-7.03 (m, 1H), 7.00-6.91 (m, 1H), 2.92 (t, J=5.3 Hz, 2H), 2.65-2.58 (m, 2H), 2.55-2.50 (m, 2H), 2.48 (t, J=5.2 Hz, 2H), 2.32 (s, 3H). Example 11: Synthesis of 4-chloro-2-fluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene) methyl)pyridin-2-yl)benzamide Step 1) Synthesis of tert-butyl 4-(fluoro(6-(4-chloro-2-fluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (0.6 g, 1.62 mmol), 4-chloro-2-fluorobenzamide (400 mg, 2.3 mmol), potassium carbonate (2.0 g, 14.5 mmol), cuprous iodide (200 mg, 1.05 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.2 mL, 1.17 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=10/1) to get the title compound as a white solid (543 mg, 72.2%). MS (ESI, pos. ion) m/z: 464.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.90 (d, J=13.7 Hz, 1H), 8.27 (d, J=8.3 Hz, 1H), 8.11 (t, J=8.5 Hz, 1H), 7.81 (t, J=8.0 Hz, 1H), 7.36-7.29 (m, 2H), 7.30-7.27 (m, 1H), 3.56-3.46 (m, 4H), 2.87 (t, J=5.3 Hz, 2H), 2.54 (s, 2H), 1.49 (s, 9H). Step 2) Synthesis of 4-chloro-2-fluoro-N-(6-(fluoro(piperidine-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-(fluoro(6-(4-chloro-2-fluorobenzamide)pyridin-2-yl) methylene)piperidine-1-carboxylate (520 mg, 1.12 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) was reacted in methanol (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a white slime-like substance (340 mg, 83.3%). MS (ESI, pos. ion) m/z: 364.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.92 (d, J=13.4 Hz, 1H), 8.29 (dd, J=8.3, 3.7 Hz, 1H), 8.13 (td, J=8.5, 2.5 Hz, 1H), 7.83 (td, J=8.0, 3.0 Hz, 1H), 7.37-7.33 (m, 1H), 7.33-7.25 (m, 2H), 3.57-3.51 (m, 2H), 3.01 (br, 2H), 2.89 (brs, 2H), 2.56 (brs, 2H). Step 3) Synthesis of 4-chloro-2-fluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl) pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 4-chloro-2-fluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) benzamide (310 mg, 0.85 mmol), sodium cyanoborohydride (200 mg, 3.17 mmol) and formaldehyde (40%, 0.64 mL, 8.6 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (272 mg, 84.5%). MS (ESI, pos. ion) m/z: 378.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.92 (d, J=13.3 Hz, 1H), 8.28 (d, J=8.3 Hz, 1H), 8.13 (t, J=8.5 Hz, 1H), 7.82 (t, J=8.0 Hz, 1H), 7.35 (d, J=8.6 Hz, 1H), 7.31 (d, J=7.7 Hz, 1H), 7.27 (d, J=10.2 Hz, 1H), 2.94 (t, J=5.5 Hz, 2H), 2.66-2.60 (m, 2H), 2.57-2.51 (m, 2H), 2.49 (t, J=5.4 Hz, 2H), 2.34 (s, 3H). Example 12: Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl) pyridine-2-yl)benzamide Step 1) Synthesis of tert-butyl 4-(fluoro(6-(2,4,6-trifluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (0.6 g, 1.62 mmol), 2,4,6-trifluorobenzamide (400 mg, 2.28 mmol), potassium carbonate (2.0 g, 14.5 mmol), cuprous iodide (200 mg, 1.05 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.2 mL, 1.17 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=10/1) to get the title compound as a white solid (330 mg, 43.7%). MS (ESI, pos. ion) m/z: 466.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.27 (d, J=8.2 Hz, 1H), 7.82 (t, J=8.0 Hz, 1H), 7.30 (d, J=7.7 Hz, 1H), 7.26 (s, 1H), 6.78 (t, J=8.2 Hz, 2H), 3.55-3.48 (m, 2H), 3.45 (t, J=5.4 Hz, 2H), 2.81 (t, J=5.3 Hz, 2H), 2.52 (br, 2H), 1.47 (s, 9H). Step 2) Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-(fluoro(6-(2,4,6-trifluorobenzamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (330 mg, 0.71 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (151 mg, 57.9%). MS (ESI, pos. ion) m/z: 366.1 [M+H]+; 1H NMR (600 MHz, CDCl3) δ (ppm) 8.52 (s, 1H), 8.26 (d, J=8.2 Hz, 1H), 7.81 (t, J=8.0 Hz, 1H), 7.28 (d, J=7.7 Hz, 1H), 6.76 (t, J=8.2 Hz, 2H), 2.98 (br, 2H), 2.91 (br, 2H), 2.79 (br, 2H), 2.52 (br, 2H). Step 3) Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl) pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 2,4,6-trifluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) benzamide (150 mg, 0.41 mmol), sodium cyanoborohydride (100 mg, 1.58 mmol) and formaldehyde (40%, 0.32 mL, 4.3 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (101 mg, 64.2%). MS (ESI, pos. ion) m/z: 380.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.66 (s, 1H), 8.27 (d, J=8.2 Hz, 1H), 7.81 (t, J=8.0 Hz, 1H), 7.28 (d, J=6.4 Hz, 1H), 6.73 (t, J=8.3 Hz, 2H), 2.84 (t, J=5.1 Hz, 2H), 2.59 (br, 2H), 2.54-2.49 (m, 2H), 2.42 (t, J=5.2 Hz, 2H), 2.30 (s, 3H). Example 13: Synthesis of 5-fluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl)picolinamide Step 1) Synthesis of tert-butyl 4-(fluoro(6-(5-fluoropicolinamide)pyridin-2-yl)methylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (0.31 g, 0.84 mmol), 5-fluoropyridine-2-formamide (250 mg, 1.80 mmol), potassium carbonate (0.82 g, 5.9 mmol), cuprous iodide (350 mg, 1.8 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.14 mL, 0.82 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=10/1) to get the title compound as light yellow oil (0.26 g, 72%). MS (ESI, pos. ion) m/z: 431.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 10.26 (s, 1H), 8.53 (d, J=2.6 Hz, 1H), 8.39-8.34 (m, 2H), 7.83 (t, J=8.0 Hz, 1H), 7.63 (td, J=8.3, 2.7 Hz, 1H), 7.31 (d, J=7.6 Hz, 1H), 3.58-3.50 (m, 4H), 2.93 (t, J=5.3 Hz, 2H), 2.60-2.55 (m, 2H), 1.51 (s, 9H). Step 2) Synthesis of 5-fluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl)picolinamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-(fluoro(6-(5-fluoropicolinamide)pyridin-2-yl)methylene) piperidine-1-carboxylate (250 mg, 0.58 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (0.12 g, 62.5%). MS (ESI, pos. ion) m/z: 331.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 10.24 (s, 1H), 8.52 (d, J=2.3 Hz, 1H), 8.35 (dd, J=11.0, 6.1 Hz, 2H), 7.82 (t, J=7.9 Hz, 1H), 7.66-7.59 (m, 1H), 7.30 (s, 1H), 3.06 (br, 6H), 2.80 (s, 1H), 2.65 (brs, 2H). Step 3) Synthesis of 5-fluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) picolinamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 5-fluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) picolinamide (0.22 g, 0.67 mmol), sodium cyanoborohydride (130 mg, 2.0 mmol) and formaldehyde (40%, 0.49 mL, 6.6 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=50/1) to give the title compound as an off-white solid (0.12 g, 52.3%). MS (ESI, pos. ion) m/z: 345.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 10.28 (d, J=11.6 Hz, 1H), 8.52 (d, J=2.6 Hz, 1H), 8.40-8.32 (m, 2H), 7.82 (t, J=8.0 Hz, 1H), 7.63 (td, J=8.3, 2.7 Hz, 1H), 7.31 (s, 1H), 2.97 (t, J=5.3 Hz, 2H), 2.64 (d, J=4.7 Hz, 2H), 2.60-2.46 (m, 4H), 2.35 (s, 3H). Example 14: Synthesis of 5-chloro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl) pyridin-2-yl)picolinamide Step 1) Synthesis of tert-butyl 4-((6-(5-chloropicolinamide)pyridin-2-yl)fluoromethylene) piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (0.33 g, 0.89 mmol), 5-chloropyridine-2-formamide (280 mg, 2.2 mmol), potassium carbonate (0.87 g, 6.2 mmol), cuprous iodide (500 mg, 2.6 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.15 mL, 0.88 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=10/1) to get the title compound as a white solid (0.29 g, 73%). MS (ESI, pos. ion) m/z: 447.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 10.27 (s, 1H), 8.64 (d, J=1.8 Hz, 1H), 8.36 (d, J=8.3 Hz, 1H), 8.28 (d, J=8.4 Hz, 1H), 7.92 (dd, J=8.4, 2.2 Hz, 1H), 7.84 (t, J=8.0 Hz, 1H), 7.31 (d, J=7.7 Hz, 1H), 3.58-3.52 (m, 4H), 2.93 (t, J=5.4 Hz, 2H), 2.58 (brs, 2H), 1.52 (s, 9H). Step 2) Synthesis of 5-chloro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl)picolinamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-(5-chloropicolinamide)pyridin-2-yl)fluoromethylene) piperidine-1-carboxylate (100 mg, 0.22 mmol) and hydrogen chloride ethyl acetate solution (2 M, 2 mL) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (0.07 g, 90.2%). MS (ESI, pos. ion) m/z: 347.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 10.28 (s, 1H), 8.63 (d, J=1.9 Hz, 1H), 8.35 (d, J=8.3 Hz, 1H), 8.28 (d, J=8.3 Hz, 1H), 7.91 (dd, J=8.4, 2.3 Hz, 1H), 7.83 (t, J=8.0 Hz, 1H), 7.31 (s, 1H), 3.05-2.93 (m, 4H), 2.87 (s, 2H), 2.56 (s, 2H). Step 3) Synthesis of 5-chloro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl)pyridin-2-yl) picolinamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 5-chloro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) picolinamide (0.2 g, 0.58 mmol), sodium cyanoborohydride (110 mg, 1.7 mmol) and formaldehyde (40%, 0.43 mL, 5.8 mmol) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=50/1) to give the title compound as an off-white solid (0.17 g, 80.7%). MS (ESI, pos. ion) m/z: 361.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 10.27 (s, 1H), 8.63 (d, J=2.0 Hz, 1H), 8.34 (d, J=8.3 Hz, 1H), 8.27 (d, J=8.4 Hz, 1H), 7.91 (dd, J=8.4, 2.2 Hz, 1H), 7.82 (t, J=8.0 Hz, 1H), 7.29 (d, J=7.5 Hz, 1H), 2.96 (t, J=5.2 Hz, 2H), 2.64 (t, J=4.6 Hz, 2H), 2.57-2.48 (m, 4H), 2.34 (s, 3H). Example 15: Synthesis of 2,4,6-trifluoro-N-(6-((1-ethylpiperidin-4-ylidene)methyl) pyridin-2-yl)benzamide Step 1) Synthesis of 2-bromo-6-(piperidin-4-ylidenemethyl)pyridine The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)methylene)piperidine-1-carboxylate (5.0 g, 14.16 mmol) and hydrogen chloride ethyl acetate solution (2 M, 20 mL) were reacted in methanol (20 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (3.5 g, 98%). MS (ESI, pos. ion) m/z: 253.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.68 (t, J=7.6 Hz, 1H), 7.41 (d, J=7.6 Hz, 1H), 7.28 (d, J=7.6 Hz, 1H), 6.25 (s, 1H), 2.91-2.86 (m, 2H), 2.83 (brs, 4H), 2.30 (t, J=5.6 Hz, 2H). Step 2) Synthesis of 2-bromo-6-((1-ethylpiperidin-4-ylidene)methyl)pyridine Under 25° C., 2-bromo-6-(piperidin-4-ylidenemethyl)pyridine (800 mg, 3.2 mmol), N,N-diisopropylethylamine (1.0 mL, 6.05 mmol) and acetonitrile (10 mL) were added into a 100 mL single-neck round bottom flask, and then iodoethane (0.38 mL, 4.75 mmol) was added. The mixture was stirred for 16 hours. After the mixture was stopped stirring, water (20 mL) was added, and the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol (v/v)=20/1) to get the title compound as light yellow oil (0.87 g, 96.7%). MS (ESI, pos. ion) m/z: 281.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.49 (t, J=7.6 Hz, 1H), 7.28 (d, J=7.6 Hz, 1H), 7.07 (d, J=7.6 Hz, 1H), 6.30 (s, 1H), 3.46 (brs, 2H), 3.23-3.16 (m, 4H), 3.07 (q, J=7.2 Hz, 2H), 2.85 (t, J=5.2 Hz, 2H), 1.46 (t, J=7.2 Hz, 3H). Step 3) Synthesis of 2,4,6-trifluoro-N-(6-((1-ethylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., 2-bromo-6-((1-ethylpiperidin-4-ylidene)methyl)pyridine (0.2 g, 1.1 mmol), 2,4,6-trifluoro-benzamide (351 mg, 2.0 mmol), potassium carbonate (1.1 g, 7.98 mmol), cuprous iodide (220 mg, 1.2 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.2 mL, 1.17 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methano (v/v)=20/1) to get the title compound as alight yellow solid (220 mg, 51.3%). MS (ESI, pos. ion) m/z: 376.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ (ppm) 11.14 (s, 1H), 7.96 (d, J=8.0 Hz, 1H), 7.83 (t, J=8.0 Hz, 1H), 7.37 (t, J=8.4 Hz, 2H), 7.09 (d, J=7.6 Hz, 1H), 6.38 (s, 1H), 3.41 (brs, 2H), 3.22-3.13 (m, 4H), 3.08-3.01 (m, 2H), 2.87-2.82 (m, 2H), 1.19 (t, J=7.2 Hz, 3H). Example 16: Synthesis of N-(6-((1-cyclopropylpiperidin-4-ylidene)methyl)pyridin-2-yl)-2,4,6-trifluorobenzamide Step 1) Synthesis of 2-bromo-6-((1-cyclopropylpiperidin-4-ylidene)methyl)pyridine The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 2-bromo-6-(piperidin-4-ylidenemethyl)pyridine (800 mg, 3.2 mmol), sodium cyanoborohydride (600 mg, 9.48 mmol), 1-ethoxy-1-trimethylsiloxycyclopropane (1.0 mL, 5.0 mmol) and acetic acid (0.1 mL) were reacted in methanol (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=100/1) to give the title compound as a light yellow solid (110 mg, 11.9%). MS (ESI, pos. ion) m/z: 293.1 [M+H]+; Step 2) Synthesis of N-(6-((1-cyclopropylpiperidin-4ylidene)methyl)pyridin-2-yl)-2,4,6-trifluorobenzamide The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., 2-bromo-6-((1-cyclopropylpiperidin-4-ylidene)methyl)pyridine (0.41 g, 1.4 mmol), 2,4,6-trifluoro-benzamide (200 mg, 1.1 mmol), potassium carbonate (1.1 g, 7.98 mmol), cuprous iodide (220 mg, 1.2 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.2 mL, 1.17 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methano (v/v)=20/1) to get the title compound as a light yellow solid (139 mg, 31.4%). MS (ESI, pos. ion) m/z: 388.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.94 (s, 1H), 8.16 (d, J=8.0 Hz, 1H), 7.70 (t, J=8.0 Hz, 1H), 6.92 (d, J=7.6 Hz, 1H), 6.69 (t, J=8.0 Hz, 2H), 6.06 (s, 1H), 2.75 (t, J=5.6 Hz, 2H), 2.71 (t, J=5.6 Hz, 2H), 2.57 (t, J=5.6 Hz, 2H), 2.35 (t, J=5.6 Hz, 2H), 1.61-1.53 (m, 1H), 0.43 (m, 4H). Example 17: Synthesis of N-(6-((1-(2,2-difluoroethyl)piperidin-4-ylidene)methyl)pyridin-2-yl)-2,4,6-trifluorobenzamide Step 1) Synthesis of 2-bromo-6-((1-(2,2-difluoroethyl)piperidin-4-ylidene)methyl)pyridine 2-Bromo-6-(piperidin-4-ylidenemethyl)pyridine (600 mg, 2.4 mmol), 2,2-difluoroethyl 4-methylbenzenesulfonate (900 mg, 3.8 mmol), sodium iodide (400 mg, 2.7 mmol), potassium carbonate (370 mg, 2.7 mmol) and N,N-dimethylformamide (10 mL) were added into a 100 mL single-neck round bottom flask. The mixture was stirred at 70° C. in an oil bath for 24 hours. After the mixture was stopped stirring, water (40 mL) was added, and the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol (v/v)=100/1) to get the title compound as light yellow oil (0.74 g, 98.4%). MS (ESI, pos. ion) m/z: 317.0 [M+H]+; Step 2) Synthesis of N-(6-((1-(2,2-difluoroethyl)piperidin-4-ylidene)methyl)pyridin-2-yl)-2,4,6-trifluorobenzamide The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., 2-bromo-6-((1-(2,2-difluoroethyl)piperidin-4-ylidene)methyl)pyridine (0.74 g, 2.3 mmol), 2,4,6-trifluoro-benzamide (200 mg, 1.1 mmol), potassium carbonate (1.1 g, 7.98 mmol), cuprous iodide (220 mg, 1.2 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.2 mL, 1.17 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methano (v/v)=50/1) to get the title compound as a light yellow solid (103 mg, 21.9%). MS (ESI, pos. ion) m/z: 412.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.61 (s, 1H), 8.15 (d, J=8.0 Hz, 1H), 7.70 (t, J=8.0 Hz, 1H), 6.93 (d, J=7.6 Hz, 1H), 6.73 (t, J=8.4 Hz, 2H), 6.13 (s, 1H), 5.88 (tt, J=56.0, 4.4 Hz, 1H), 2.83 (t, J=5.6 Hz, 2H), 2.76 (dd, J=15.2, 4.4 Hz, 2H), 2.69 (dd, J=8.8, 4.8 Hz, 2H), 2.57 (t, J=5.6 Hz, 2H), 2.40 (t, J=5.6 Hz, 2H). Example 18: Synthesis of N-(6-((1-(2,2,2-trifluoroethyl)piperidin-4-ylidene)methyl) pyridin-2-yl)-2,4,6-trifluorobenzamide Step 1) Synthesis of 2-bromo-6-((1-(2,2,2-trifluoroethyl)piperidin-4-ylidene)methyl)pyridine Under 25° C., 2-bromo-6-(piperidin-4-ylidenemethyl)pyridine (500 mg, 2.0 mmol), N,N-diisopropylethylamine (1.0 mL, 6.1 mmol), 2,2,2-trifluoroethyltrifluoromethanesulfonate (0.7 mL, 3.9 mmol) and dichloromethane (10 mL) were added into a 100 mL single-neck round bottom flask, and the mixture was stirred for 16 hours. After the mixture was stopped stirring, water (30 mL) was added, and the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol (v/v)=100/1) to get the title compound as light yellow oil (0.61 g, 92.1%). MS (ESI, pos. ion) m/z: 335.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.46 (t, J=7.6 Hz, 1H), 7.26 (d, J=7.6 Hz, 1H), 7.07 (d, J=7.6 Hz, 1H), 6.23 (s, 1H), 3.01 (q, J=9.6 Hz, 2H), 2.90 (t, J=5.6 Hz, 2H), 2.79 (t, J=5.6 Hz, 2H), 2.72 (t, J=5.6 Hz, 2H), 2.41 (t, J=5.6 Hz, 2H). Step 2) Synthesis of N-(6-((1-(2,2,2-trifluoroethyl)piperidin-4-ylidene)methyl)pyridin-2-yl)-2,4,6-trifluorobenzamide The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., 2-bromo-6-((1-(2,2,2-trifluoroethyl)piperidin-4-ylidene)methyl) pyridine (0.6 g, 1.8 mmol), 2,4,6-trifluoro-benzamide (200 mg, 1.1 mmol), potassium carbonate (1.1 g, 7.98 mmol), cuprous iodide (220 mg, 1.2 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.2 mL, 1.17 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methano (v/v)=50/1) to get the title compound as a light yellow solid (277 mg, 56.5%). MS (ESI, pos. ion) m/z: 430.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.95 (d, J=8.0 Hz, 1H), 7.79 (t, J=8.0 Hz, 1H), 7.34 (t, J=8.8 Hz, 2H), 7.04 (d, J=7.6 Hz, 1H), 6.22 (s, 1H), 3.20 (q, J=10.4 Hz, 2H), 2.95 (brs, 2H), 2.74 (t, J=5.2 Hz, 2H), 2.66 (t, J=4.8 Hz, 2H), 2.32 (brs, 2H). Example 19: Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(1-ethylpiperidin-4-ylidene)methyl) pyridin-2-yl)benzamide Step 1) Synthesis of 2-bromo-6-(fluoro(piperidin-4-ylidene)methyl)pyridine The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (1.6 g, 4.3 mmol) and methanesulfonic acid (0.84 mL, 13.0 mmol) were reacted in dichloromethane (15 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a yellow solid (1.15 g, 95.8%). MS (ESI, pos. ion) m/z: 271.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.59 (t, J=7.8 Hz, 1H), 7.46 (d, J=7.8 Hz, 1H), 7.38 (d, J=7.9 Hz, 1H), 3.01-2.90 (m, 6H), 2.53 (dd, J=7.6, 3.4 Hz, 2H). Step 2) Synthesis of 2-bromo-6-((1-ethylpiperidin-4-ylidene)fluoromethyl)pyridine Under 25° C., 2-bromo-6-(fluoro(piperidin-4-ylidene)methyl)pyridine (0.38 g, 1.4 mmol), N,N-diisopropylethylamine (0.69 mL, 4.2 mmol) and acetonitrile (5 mL) were added into a 100 mL single-neck round bottom flask, and then iodoethane (0.44 g, 2.8 mmol) was added. The mixture was stirred for 5 hours. After the mixture was stopped stirring, water (20 mL) was added, and the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol (v/v)=20/1) to get the title compound as a light yellow solid (0.4 g, 95.4%). MS (ESI, pos. ion) m/z: 299.0 [M+H]+; 1H NMR (600 MHz, CDCl3) δ (ppm) 7.60 (t, J=7.8 Hz, 1H), 7.47 (d, J=7.8 Hz, 1H), 7.40 (d, J=7.9 Hz, 1H), 3.21 (brs, 2H), 2.76 (brs, 6H), 2.67-2.62 (m, 2H), 1.26 (t, J=7.2 Hz, 3H). Step 3) Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(1-ethylpiperidin-4-ylidene)methyl)pyridin-2-yl) benzamide The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., 2-bromo-6-((1-ethylpiperidin-4-ylidene)fluoromethyl)pyridine (0.38 g, 1.27 mmol), 2,4,6-trifluoro-benzamide (330 mg, 1.9 mmol), potassium carbonate (0.53 g, 3.8 mmol), cuprous iodide (240 mg, 1.3 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.21 mL, 1.3 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methano (v/v)=20/1) to get the title compound as a light yellow solid (0.2 g, 40%). MS (ESI, pos. ion) m/z: 394.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.79 (s, 1H), 8.30 (d, J=8.2 Hz, 1H), 7.83 (t, J=8.0 Hz, 1H), 7.28 (d, J=6.8 Hz, 1H), 6.73 (t, J=8.4 Hz, 2H), 2.84 (t, J=4.9 Hz, 2H), 2.59 (br, 2H), 2.56 (br, 2H), 2.44 (br, 4H), 1.11 (t, J=7.1 Hz, 3H). Example 20: Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(1-cyclopropylpiperidin-4-ylidene) methyl)pyridin-2-yl)benzamide Step 1) Synthesis of 2-bromo-6-((1-cyclopropylpiperidin-4-ylidene)fluoromethyl)pyridine The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 2-bromo-6-(fluoro(piperidin-4-ylidene)methyl)pyridine (1.1 g, 4.06 mmol), sodium cyanoborohydride (1.04 g, 16.2 mmol), 1-ethoxy-1-trimethylsiloxycyclopropane (2.44 mL, 12.2 mmol) and acetic acid (0.7 mL) were reacted in methanol (15 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=100/1) to give the title compound as a white solid (1.2 g, 95.0%). MS (ESI, pos. ion) m/z: 311.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.57 (t, J=7.8 Hz, 1H), 7.45 (d, J=7.7 Hz, 1H), 7.37 (d, J=7.9 Hz, 1H), 2.96 (t, J=5.2 Hz, 2H), 2.74-2.69 (m, 4H), 2.57-2.54 (m, 2H), 1.65-1.59 (m, 1H), 0.48 (d, J=6.6 Hz, 4H). Step 2) Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(1-cyclopropylpiperidin-4-ylidene)methyl) pyridin-2-yl)benzamide The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., 2-bromo-6-((1-cyclopropylpiperidin-4-ylidene)fluoromethyl)pyridine (1.2 g, 3.9 mmol), 2,4,6-trifluoro-benzamide (1.0 g, 5.7 mmol), potassium carbonate (1.6 g, 11.0 mmol), cuprous iodide (0.73 g, 3.8 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.63 mL, 3.9 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=5/1) to get the title compound as a white solid (0.99 g, 63.0%). MS (ESI, pos. ion) m/z: 406.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.50 (s, 1H), 8.29 (d, J=8.2 Hz, 1H), 7.83 (t, J=8.0 Hz, 1H), 7.30 (d, J=8.4 Hz, 1H), 6.78 (t, J=8.3 Hz, 2H), 2.80 (t, J=5.2 Hz, 2H), 2.72 (t, J=5.6 Hz, 2H), 2.64 (t, J=5.3 Hz, 2H), 2.55 (t, J=4.7 Hz, 2H), 1.61-1.58 (m, 1H), 0.49-0.42 (m, 4H). Example 21: Synthesis of N-(6-((1-(2,2-difluoroethyl)piperidin-4-ylidene)fluoromethyl) pyridin-2-yl)-2,4,6-trifluorobenzamide Step 1) Synthesis of 2-bromo-6-((1-(2,2-difluoroethyl)piperidin-4-ylidene)fluoromethyl)pyridine 2-Bromo-6-(fluoro(piperidin-4-ylidene)methyl)pyridine (1.0 g, 3.69 mmol), 2,2-difluoroethyl 4-methylbenzenesulfonate (1.31 g, 5.55 mmol), sodium iodide (550 mg, 3.7 mmol), potassium carbonate (770 mg, 5.5 mmol) and acetonitrile (15 mL) were added into a 100 mL single-neck round bottom flask, and the mixture was stirred at 90° C. in an oil bath for 12 hours. After the mixture was stopped stirring, water (40 mL) was added, and the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate (v/v)=40/1) to get the title compound as yellow oil (1.1 g, 89%). MS (ESI, pos. ion) m/z: 335.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.59 (t, J=7.8 Hz, 1H), 7.40 (t, J=6.9 Hz, 2H), 4.19 (td, J=12.7, 4.1 Hz, 1H), 3.03 (t, J=5.2 Hz, 2H), 2.79 (td, J=15.0, 4.3 Hz, 2H), 2.72-2.67 (m, 4H), 2.63-2.57 (m, 2H). Step 2) Synthesis of N-(6-((1-(2,2-difluoroethyl)piperidin-4-ylidene)fluoromethyl)pyridin-2-yl)-2,4,6-trifluorobenzamide The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., 2-bromo-6-((1-(2,2-difluoroethyl)piperidin-4-ylidene)fluoromethyl) pyridine (1.05 g, 3.13 mmol), 2,4,6-trifluoro-benzamide (0.83 g, 4.7 mmol), potassium carbonate (1.31 g, 9.38 mmol), cuprous iodide (0.6 g, 3.2 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.51 mL, 3.1 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=5/1) to get the title compound as a white solid (0.92 g, 68%). MS (ESI, pos. ion) m/z: 430.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.39 (s, 1H), 8.29 (d, J=8.2 Hz, 1H), 7.84 (t, J=8.0 Hz, 1H), 7.31 (d, J=7.8 Hz, 1H), 6.80 (t, J=8.2 Hz, 2H), 5.92 (tt, J=55.9, 4.2 Hz, 1H), 2.86 (t, J=5.1 Hz, 2H), 2.77 (td, J=15.0, 4.3 Hz, 2H), 2.72-2.67 (m, 2H), 2.64-2.59 (m, 4H). Example 22: Synthesis of N-(6-((1-(2,2,2-trifluoroethyl)piperidin-4-ylidene)fluoromethyl) pyridin-2-yl)-2,4,6-trifluorobenzamide Step 1) Synthesis of 2-bromo-6-(fluoro(1-(2,2,2-trifluoroethyl)piperidin-4-ylidene)methyl) pyridine Under 25° C., 2-bromo-6-(fluoro(piperidin-4-ylidene)methyl)pyridine (250 mg, 0.92 mmol), N,N-diisopropylethylamine (1.22 mL, 7.36 mmol), 2,2,2-trifluoroethyltrifluoromethanesulfonate (0.8 mL, 4.4 mmol) and dichloromethane (5 mL) were added into a 100 mL single-neck round bottom flask, and the mixture was stirred for 16 hours. After the mixture was stopped stirring, water (30 mL) was added, and the resulting mixture was extracted with dichloromethane (30 mL×2). The combined organic layers were dried over anhydrous sodium sulfate (2 g) and filtered. The filtrate was concentrated using a rotary evaporator under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate (v/v)=50/1) to get the title compound as colorless oil (0.27 g, 82.9%). MS (ESI, pos. ion) m/z: 353.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.59 (t, J=7.8 Hz, 1H), 7.46 (d, J=7.7 Hz, 1H), 7.39 (d, J=7.9 Hz, 1H), 3.08-2.96 (m, 4H), 2.82-2.77 (m, 4H), 2.65-2.54 (m, 2H). Step 2) Synthesis of N-(6-((1-(2,2,2-trifluoroethyl)piperidin-4-ylidene)fluoromethyl)pyridin-2-yl)-2,4,6-trifluorobenzamide The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., 2-bromo-6-(fluoro(1-(2,2,2-trifluoroethyl)piperidin-4-ylidene)methyl) pyridine (0.5 g, 1.42 mmol), 2,4,6-trifluoro-benzamide (0.49 g, 2.8 mmol), potassium carbonate (0.6 g, 4.3 mmol), cuprous iodide (0.4 g, 2.1 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.23 mL, 1.4 mmol), water (1.0 mL) were reacted in toluene (10 mL) to prepare it. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=5/1) to get the title compound as a white solid (0.35 g, 55.3%). MS (ESI, pos. ion) m/z: 448.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 8.51 (s, 1H), 8.30 (d, J=8.2 Hz, 1H), 7.84 (t, J=8.0 Hz, 1H), 7.30 (d, J=7.9 Hz, 1H), 6.78 (t, J=8.2 Hz, 2H), 3.01 (q, J=9.6 Hz, 2H), 2.86 (t, J=5.0 Hz, 2H), 2.79 (t, J=5.6 Hz, 2H), 2.70 (t, J=5.2 Hz, 2H), 2.64-2.54 (m, 2H). Example 23: Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl) pyridin-2-yl)benzenesulfonamide Step 1) Synthesis of tert-butyl 4-(fluoro(6-(2,4,6-trifluorobenzenesulfonamide)pyridin-2-yl) methylene)piperidine-1-carboxylate The title compound of this step was prepared by referring to the method described in step 3 of example 1, i.e., tert-butyl 4-((6-bromopyridin-2-yl)fluoromethylene)piperidine-1-carboxylate (0.2 g, 0.54 mmol), 2,4,6-trifluorobenzenesulfonamide (170 mg, 0.81 mmol), potassium carbonate (0.23 g, 1.6 mmol), cuprous iodide (100 mg, 0.53 mmol), (1R,2R)—N1,N2-dimethylcyclohexane-1,2-diamine (0.09 mL, 0.6 mmol), water (0.5 mL) were reacted in toluene (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/ethyl acetate (v/v)=10/1) to get the title compound as yellow oil (0.24 g, 88.8%). MS (ESI, pos. ion) m/z: 446.2 [M+H-56]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.72 (t, J=8.0 Hz, 1H), 7.24 (t, J=9.2 Hz, 2H), 6.78 (t, J=8.6 Hz, 2H), 3.55-3.51 (m, 2H), 3.49-3.45 (m, 2H), 2.80 (t, J=5.1 Hz, 2H), 2.52 (brs, 2H), 1.51 (s, 9H). Step 2) Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(piperidine-4-ylidene)methyl)pyridin-2-yl) benzenesulfonamide The title compound of this step was prepared by referring to the method described in step 4 of example 1, i.e., tert-butyl 4-(fluoro(6-(2,4,6-trifluorobenzenesulfonamide)pyridin-2-yl) methylene)piperidine-1-carboxylate (0.23 g, 0.46 mmol) and methanesulfonic acid (0.13 g, 1.4 mmol) were reacted in dichloromethane (5 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=20/1) to give the title compound as a light yellow solid (0.11 g, 59.7%). MS (ESI, pos. ion) m/z: 402.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.54-7.47 (m, 1H), 7.22 (s, 1H), 7.14 (d, J=8.4 Hz, 1H), 6.85 (d, J=7.7 Hz, 1H), 6.70 (t, J=8.8 Hz, 1H), 2.82-2.75 (m, 4H), 2.51 (br, 4H). Step 3) Synthesis of 2,4,6-trifluoro-N-(6-(fluoro(1-methylpiperidin-4-ylidene)methyl) pyridin-2-yl)benzenesulfonamide The title compound of this step was prepared by referring to the method described in step 5 of example 1, i.e., 2,4,6-trifluoro-N-(6-(fluoro(piperidin-4-ylidene)methyl)pyridin-2-yl) benzenesulfonamide (0.09 g, 0.22 mmol), sodium cyanoborohydride (50 mg, 0.8 mmol) and formaldehyde (40%, 0.17 mL, 2.3 mmol) were reacted in methanol (4 mL) to prepare it. The crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v)=50/1) to give the title compound as a light yellow solid (0.085 g, 91.2%). MS (ESI, pos. ion) m/z: 416.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ (ppm) 7.64 (t, J=8.0 Hz, 1H), 7.21 (d, J=8.3 Hz, 1H), 7.10 (d, J=7.5 Hz, 1H), 6.76 (t, J=8.6 Hz, 2H), 3.07 (t, J=4.5 Hz, 2H), 2.97 (t, J=5.4 Hz, 2H), 2.92 (t, J=4.9 Hz, 2H), 2.78 (t, J=5.0 Hz, 2H), 2.62 (s, 3H). Biological Assay Example A: Evaluation of the Activating Effect of the Compound of the Invention on Human 5-HT1FReceptor Transfected by CHO-K1 Cells Experimental purposes: To evaluate the activating effect of the compound on CHO-K1 cells transfected human 5-HT1Freceptor by using HitHunter cAMP detection kit. Experimental brief process: CHO-K1 cells were cultured in a 384 microporous plate. The volume of cell culture medium (Assay Complete™ Cell Plating Reagent, DIscoverX) was 20 μL and the cell density was 10,000/well. The cells were cultured overnight at 37° C. and 5% CO2. Then the culture medium was removed, and 15 μL of cAMP Assay Buffer (DIscoverX) was added into each well, and then 5 μL of test sample containing 4× sample (test compound or 5-HT) and 4× forskolin (final concentration of forskolin is 15 μM) was added. The microporous plate was placed at 37° C. for 30 minutes. Then 5 μL of cAMP Antibody Reagent (DiscoverX) and 20 μL of cAMP Working Detection Solution (DiscoverX) were added and incubated in the dark for 1 hour, and then 20 μL of cAMP Solution A was added and incubated in the dark for 3 hours. The microporous plate was placed in the Perkin Elmer Envision™ to read the intensity of the optical signal. The activation rate was calculated by the intensity of the optical signal obtained from testing the compounds with different concentrations ((1−(Y/Z))*100%=the activation rate, wherein, Y represents the intensity of the optical signal adding test sample, and Z represents the intensity of the optical signal adding only forskolin). The dose-effect curve of the compounds was calculated by Prism software, and the concentration of the agonist with half maximum response was calculated and expressed by EC50value. The results were shown in Table A. 5-HT was used as a positive control drug to ensure the normal experimental system in this experiment. TABLE ATest results of the activating effect of thecompound of the invention on human5-HT1Freceptor transfected by CHO-K1cellsExample No.EC50(nM)Example 35.9Example 51.9Example 103.0Example 121.5Example 196.8 The experimental results show that the compound of the invention has strong activation activity on 5-HT1Freceptor. Example B: Pharmacokinetic Evaluation after Administering a Certain Amount of the Compound of the Invention by Intravenous or Gavage to Rats and Dogs The inventors had evaluated the pharmacokinetics of the compounds of the invention in rats and dogs. Wherein animal information was detailed in Table 1. TABLE 1The animal subject information of the inventionGermlineGradeSexWeightAgeSourceSD ratsSPFmale180-350 g6-11Hunan SJAweeksLaboratoryAnimal Co., Ltd.BeagleConventionalmale8-12 kg6-12Beijing MarshalldogsmonthsBiotechnologyCo., Ltd. Test Method: The compound was administered to the animal subjects in the form of 5% DMSO+60% PEG400+35% Saline solution or 10% DMSO+10% Kolliphor HS15+30% PEG400+50% Saline solution. The animals were fasted for 12 hours before administration, but drinking water freely. For the group of intravenous administration, the administration dose is 0.5 mg/kg or 1 mg/kg, and vein blood samples (0.15 mL) were collected at the time points of 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24 h (dogs) or 0.083, 0.25, 0.5, 1.0, 2.0, 5.0, 7.0 and 24 h (rats) after drug administration. EDTA-K2as anticoagulant was pre-added into the blood vessel. The plasma solutions were collected by centrifuging each blood sample at 12,000 rpm for 2 minutes and kept at −20° C. or −70° C. For the group of gavage administration, the administration dose is 2.5 mg/kg or 5 mg/kg, and vein blood samples (0.15 mL) were collected at the time points of 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24 h (dogs) or 0.25, 0.5, 1.0, 2.0, 5.0, 7.0 and 24 h (rats) after drug administration. EDTA-K2as anticoagulant was pre-added into the blood vessel. The plasma solutions were collected by centrifuging each blood sample at 12,000 rpm for 2 minutes and kept at −20° C. or −70° C. After processing the above plasma sample (the frozen plasma was melted at room temperature and then whirled for 15 seconds. 10 to 20 μL of the plasma was taken out, and whirled with 120 to 150 μL of an acetonitrile solution containing an internal standard for 5 minutes. The mixture was centrifuged for 5 minutes at 4,000 rpm. 100 μL of the supernatant was taken out and mixed with 120 to 150 μL of methanol-water (v/v=1/1)), the concentration of compounds in plasma was analyzed by LC-MS/MS. The analysis results show that the compound of the invention has good pharmacokinetic properties in rats, dogs. The compound of the invention has better drug properties and better clinical application prospects. Wherein the pharmacokinetic parameters of examples 12 and 19 in rats were detailed in Table B1; the pharmacokinetic parameters of examples 12 and 19 in dogs were detailed in Table B2. TABLE B1Pharmacokinetic parameters of the compounds of the invention in ratsTestDoseTmaxCmaxAUClastAUCINFMRTINFT1/2ClVssFGroupssubstance(mg/kg)(h)(ng/mL)(h * ng/mL)(h * ng/mL)(h)(h)(mL/min/kg)(L/kg)(%)i.v groupExample 1210.08380.51111141.661.3814614.6NDExample 1910.567.72402592.952.164.411.4NDi.g groupExample 125271.82963544.052.31NDND62Example 1953.67169126013205.013.06NDND105ND means no test data. The results of Table B1 show that the compounds of the invention have good pharmacokinetic properties in rats. TABLE B2Pharmacokinetic parameters of the compounds of the invention in dogsTestDoseTmaxCmaxAUClastAUCINFMRTINFT1/2ClVssFGroupssubstance(mg/kg)(h)(ng/mL)(h * ng/mL)(h * ng/mL)(h)(h)(mL/min/kg)(L/kg)(%)i.v groupExample 1210.251706526604.113.5225.26.22NDExample 190.50.0834426326341.391.0613.11.1NDi.g groupExample 1251.6782.86917055.984.28NDND21.4Example 192.511263563923.261.96NDND12.4ND means no test data. The results of Table B2 show that the compounds of the invention have good pharmacokinetic properties in dogs. Reference throughout this specification to “an embodiment”, “some embodiments”, “one embodiment”, “another example”, “an example”, “a specific example”, or “some examples” means that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the present disclosure. Thus, the appearances of the phrases such as “in some embodiments”, “in one embodiment”, “in an embodiment”, “in another example”, “in an example”, “in a specific example”, or “in some examples” in various places throughout this specification are not necessarily referring to the same embodiment or example of the present disclosure. Furthermore, the particular features, structures, materials, or characteristics may be combined in any suitable manner in one or more embodiments or examples. In addition, those skilled in the art can integrate and combine different embodiments, examples or the features of them as long as they are not contradictory to one another. Although explanatory embodiments have been shown and described, it would be appreciated by those skilled in the art that the above embodiments cannot be construed to limit the present disclosure, and changes, alternatives, and modifications can be made in the embodiments without departing from spirit, principles and scope of the present disclosure.
166,821
11858911
DETAILED DESCRIPTION Hereinafter, example embodiments will be described in detail, and may be easily performed by a person having an ordinary skill in the related art. However, this disclosure may be embodied in many different forms and is not construed as limited to the example embodiments set forth herein. It should be understood that, although the terms first, second, third, etc. may be used herein to describe various elements, components, regions, layers and/or sections, these elements, components, regions, layers, and/or sections should not be limited by these terms. These terms are only used to distinguish one element, component, region, layer, or section from another region, layer, or section. Thus, a first element, component, region, layer, or section discussed below could be termed a second element, component, region, layer, or section without departing from the teachings of example embodiments. Spatially relative terms (e.g., “beneath,” “below,” “lower,” “above,” “upper,” and the like) may be used herein for ease of description to describe one element or feature's relationship to another element(s) or feature(s) as illustrated in the figures. It should be understood that the spatially relative terms are intended to encompass different orientations of the device in use or operation in addition to the orientation depicted in the figures. For example, if the device in the figures is turned over, elements described as “below” or “beneath” other elements or features would then be oriented “above” the other elements or features. Thus, the term “below” may encompass both an orientation of above and below. The device may be otherwise oriented (rotated 90 degrees or at other orientations) and the spatially relative descriptors used herein interpreted accordingly. The terminology used herein is for the purpose of describing various embodiments only and is not intended to be limiting of example embodiments. As used herein, the singular forms “a,” “an,” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will be further understood that the terms “includes,” “including,” “comprises,” and/or “comprising,” when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. Example embodiments are described herein with reference to cross-sectional illustrations that are schematic illustrations of idealized embodiments (and intermediate structures) of example embodiments. As such, variations from the shapes of the illustrations as a result, for example, of manufacturing techniques and/or tolerances, are to be expected. Thus, example embodiments should not be construed as limited to the shapes of regions illustrated herein but are to include deviations in shapes that result, for example, from manufacturing. Unless otherwise defined, all terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skill in the art to which example embodiments belong. It will be further understood that terms, including those defined in commonly used dictionaries, should be interpreted as having a meaning that is consistent with their meaning in the context of the relevant art and will not be interpreted in an idealized or overly formal sense unless expressly so defined herein. As used herein, when a definition is not otherwise provided, “infrared light” refers to “near infrared light (NIR)” in a region of about 700 nm to about 1400 nm. When the terms “about” or “substantially” are used in this specification in connection with a numerical value, it is intended that the associated numerical value include a tolerance of ±10% around the stated numerical value. When ranges are specified, the range includes all values therebetween such as increments of 0.1%. As used herein, when a definition is not otherwise provided, “substituted” refers to replacement of hydrogen of a compound or a functional group by a substituent selected from a halogen (F, Br, Cl, or I), a hydroxy group, a nitro group, a cyano group, an amino group, an azido group, an amidino group, a hydrazino group, a hydrazono group, a carbonyl group, a carbamyl group, a thiol group, an ester group, a carboxyl group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid or a salt thereof, a C1 to C20 alkyl group, for example a C1 to C10 alkyl group, a C1 to C20 alkoxy group, for example a C1 to C10 alkoxy group, a C2 to C20 alkenyl group, a C2 to C20 alkynyl group, a C6 to C30 aryl group, a C7 to C30 arylalkyl group, a C3 to C20 heteroaryl group, a C3 to C20 heteroarylalkyl group, a C3 to C30 cycloalkyl group, a C3 to C15 cycloalkenyl group, a C6 to C15 cycloalkynyl group, a C2 to C20 heterocycloalkyl group, and a combination thereof. In addition, “substituted” in an aromatic ring group refers to replacement of —CH2— in the ring by —NR— (wherein R is selected from hydrogen, a halogen, a C1 to C10 alkyl group, a C1 to C10 alkoxy group, a C6 to C30 aryl group, and a C3 to C20 heteroaryl group), —O—, —S—, or —Se— or replacement of —CH═ in the ring by —N═. As used herein, when a definition is not otherwise provided, “hetero” refers to inclusion of one to three heteroatoms selected from N, O, S, P, and Si. As used herein, when a definition is not otherwise provided, “halogen” refers to F, Br, Cl, or I. Singular terms in the present disclosure may include a plurality of objects unless one object is precisely indicated. All numerical ranges of the present disclosure include all numbers and ranges within set forth numerical ranges. In addition, numerical ranges and parameters indicating a broad scope of this disclosure are approximate values but the numerical values set forth in the Examples section are reported as precisely as possible. However, it should be understood that such numerical values inherently contain certain errors resulting from measuring equipment and/or a measuring technique. According to some example embodiments, a squarylium compound represented by Chemical Formula 1 is provided. In Chemical Formula 1, X1and X2are the same or different and are independently selected from a functional group represented by Chemical Formula 1A, a functional group represented by Chemical Formula 1B, a functional group represented by Chemical Formula 1C, and a functional group represented by Chemical Formula 1D, provided that at least one of X1and X2is a functional group represented by Chemical Formula 1A or a functional group represented by Chemical Formula 1B, wherein, in Chemical Formula 1A,Y1and Y2are independently N or NR16,R11and R12are independently one compound of a first set of compounds, where the first set of compounds includes hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, or R11and R12are linked with each other to collectively comprise a fused ring with a quinazoline ring,R13is one compound of a second set of compounds, the second set of compounds including a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 heteroaryl group, a substituted or unsubstituted C6 to C20 arylamine group, and a substituted or unsubstituted C3 to C30 heteroarylamine group, andR14, R15, and R16are independently selected from (“independently one of”) hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group or R14and R15are linked with each other to provide a fused ring with a quinazoline ring, wherein, in Chemical Formula 1B,m is 1 or 2,Z1and Z2are independently hydrogen or a hydroxyl group,R21to R26are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, andR27and R28are independently selected from a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group or R27and R28are optionally linked with each other to provide (“collectively comprise”) an N-containing aromatic ring group or an N-containing alicyclic cyclic group, wherein, in Chemical Formula 1C,n is 1 or 2,R31and R32are linked with each other to provide an aromatic ring group or an alicyclic cyclic group,R33and R34are linked with each other to provide an aromatic ring group or an alicyclic cyclic group, andR35to R38are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, wherein, in Chemical Formula 1 D,k is 0 or 1,Z3and Z4are independently hydrogen or a hydroxyl group,R41and R42are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, andR43and R44are independently selected from a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group or R43and R44are optionally linked with each other to provide an N-containing aromatic ring group or an N-containing alicyclic cyclic group. The squarylium compound represented by Chemical Formula 1 may be a compound represented by Chemical Formula 1′. The squarylium compound represented by Chemical Formula 1 includes an electron donating group selected from the functional group represented by Chemical Formula 1A, the functional group represented by Chemical Formula 1B, the functional group represented by Chemical Formula 10, and the functional group represented by Chemical Formula 1D in the center of the squarylium nucleus and at least one of X1and X2includes the functional group represented by Chemical Formula 1A or the functional group represented by Chemical Formula 1B, and thereby the squarylium compound has excellent absorbance in an infrared/near infrared wavelength spectrum of light. The squarylium compound may have (“be configured to have”) a maximum absorption wavelength (λmax) in a range of greater than or equal to about 700 nm and less than or equal to about 1300 nm, for example greater than or equal to about 710 nm and less than or equal to about 1200 nm and a full width at half maximum (FWHM) of greater than or equal to about 50 nm and less than or equal to about 150 nm, for example greater than or equal to about 50 nm and less than or equal to about 130 nm, based on the squarylium compound being in a thin film state (e.g., being included in a thin film). The squarylium compound represented by Chemical Formula 1 has high absorbance in an infrared/near infrared wavelength spectrum of light and high transmittance in a visible ray region and thereby high selective absorbance in an infrared/near infrared wavelength spectrum of light. That is, the squarylium compound may be associated with a maximum absorption coefficient in an infrared ray (IR) wavelength spectrum of light (ANIR) and a maximum absorption coefficient in a visible wavelength spectrum of light (AVIS) that satisfy Relationship Equation 1. ANIR/AVIS≥8  [Relationship Equation 1] In Relationship Equation 1, ANIRis a maximum absorption coefficient in an infrared ray (IR) region and AVISis a maximum absorption coefficient in a visible wavelength spectrum of light. A ratio (ANIR/AVIS) of the maximum absorption coefficient in the infrared ray (IR) and the maximum absorption coefficient in the visible wavelength spectrum of light may be greater than or equal to about 9, for example about 10 to about 550 or about 15 to about 550. When the ratio (ANIR/AVIS) of the absorption coefficients is within the ranges, selective absorbance for light in an infrared/near infrared wavelength spectrum of light is improved. Transmittance in a visible wavelength spectrum of light of the squarylium compound represented by Chemical Formula 1 may be greater than or equal to about 80%, for example greater than or equal to about 90%, and particularly transmittance in a blue wavelength spectrum of light of about 300 nm to about 450 nm may be greater than or equal to about 80%, for example greater than or equal to about 90%. In addition, a molar extinction coefficient in an infrared/near infrared wavelength spectrum of light may be greater than or equal to about 7×104M−1cm−1, for example greater than or equal to about 7.5×104M−1cm−1, or greater than or equal to about 8×104M−1cm−1. If absorbance in a blue wavelength spectrum of light of about 300 nm to about 450 nm is high despite high absorbance in an infrared ray (IR) region, efficiency may be deteriorated. From this view, the squarylium compound represented by Chemical Formula 1 has low absorbance (i.e., high transmittance) in a blue wavelength spectrum of light of about 300 nm to about 450 nm and high absorbance in an infrared ray (IR) region, and thus efficiency (e.g., external quantum efficiency) of an electronic device may be improved based on including the squarylium compound represented by Chemical Formula 1. R11and R12represented by Chemical Formula 1A are linked with each other to provide a C6 or C7 aromatic ring that is fused with a fused ring (e.g., a quinazoline ring) of a 6-membered aromatic ring including Y1and Y2and a benzene ring and the aromatic ring may not include a heteroatom. In this case, a structure where three aromatic rings are fused structure is provided. Chemical Formula 1A may be represented by Chemical Formula 1A-1. In Chemical Formula 1A-1,Y1and Y2are independently N or NR16,R16, R17, R18, and R19are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,R13is a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 heteroaryl group, a substituted or unsubstituted C6 to C20 arylamine group, and a substituted or unsubstituted C3 to C30 heteroarylamine group, andR14and R15are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group or R14and R15are linked with each other to provide a ring fused with a quinazoline ring. R13represented by Chemical Formula 1A or Chemical Formula 1A-1 may be selected from a substituted or unsubstituted phenyl group, a substituted or unsubstituted naphthyl group, a substituted or unsubstituted anthracenyl group, a substituted or unsubstituted phenanthrenyl group, a substituted or unsubstituted pyrenyl group, a substituted or unsubstituted chrysenyl group, a substituted or unsubstituted fluorenyl group, and a substituted or unsubstituted perylenyl group. R13represented by Chemical Formula 1A or Chemical Formula 1A-1 may be selected from a substituted or unsubstituted pyridyl group, a substituted or unsubstituted pyrazinyl group, a substituted or unsubstituted pyrimidinyl group, a substituted or unsubstituted pyridazinyl group, a substituted or unsubstituted quinolyl group, a substituted or unsubstituted isoquinolyl group, a substituted or unsubstituted phthalazinyl group, a substituted or unsubstituted quinazolinyl group, a substituted or unsubstituted quinoxalinyl group, a substituted or unsubstituted naphthyridinyl group, a substituted or unsubstituted cinnolinyl group, a substituted or unsubstituted pyrrolyl group, a substituted or unsubstituted pyrazolyl group, a substituted or unsubstituted imidazolyl group, a substituted or unsubstituted triazolyl group, a substituted or unsubstituted tetrazolyl group, a substituted or unsubstituted thienyl group, a substituted or unsubstituted thiazolyl group, a substituted or unsubstituted oxazolyl group, a substituted or unsubstituted indolyl group, a substituted or unsubstituted isoindolyl group, a substituted or unsubstituted indazolyl group, a substituted or unsubstituted benzoimidazolyl group, a substituted or unsubstituted benzotriazolyl group, a substituted or unsubstituted benzothiazolyl group, a substituted or unsubstituted benzooxazolyl group, a substituted or unsubstituted carbazole group, a substituted or unsubstituted phenazinyl group, and a substituted or unsubstituted acridinyl group. R13represented by Chemical Formula 1A or Chemical Formula 1A-1 may be one functional group of a plurality of functional groups represented by Chemical Formula 2. In Chemical Formula 2,hydrogen of each aromatic ring may be replaced by a substituent selected from a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, andeach position of a plurality of aromatic rings of the plurality of functional groups that is not indicated by an asterisk (*) may be a binding position at R13of Chemical Formula 1A. R13represented by Chemical Formula 1A or Chemical Formula 1A-1 may be a substituted or unsubstituted pyrrolidinyl group, a substituted or unsubstituted piperidinyl group, a substituted or unsubstituted piperazinyl group, a substituted or unsubstituted morpholinyl group, a substituted or unsubstituted thiomorpholinyl group, a substituted or unsubstituted tetrahydropyridyl group, a substituted or unsubstituted tetrahydroquinolinyl group, a substituted or unsubstituted tetrahydroisoquinolinyl group, a substituted or unsubstituted tetrahydrofuryl group, a substituted or unsubstituted tetrahydropyranyl group, a substituted or unsubstituted dihydrobenzofuranyl group, a substituted or unsubstituted indolinyl group, a substituted or unsubstituted isoindolinyl group, and a substituted or unsubstituted tetrahydrocarbazolyl group which are represented by Chemical Formula 3. In Chemical Formula 3,Raand Rbare independently selected from a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, hydrogen of each aromatic ring or alicyclic ring may be replaced by a substituent selected from a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, andany position of aromatic rings or alicyclic rings of functional groups that are not indicated by asterisk (*) may be a binding position at R13represented by Chemical Formula 1A or 1A-1. In R13represented by Chemical Formula 1A or Chemical Formula 1A-1, the substituted or unsubstituted C6 to C20 arylamine group and the substituted or unsubstituted C3 to C30 heteroarylamine group may be represented by —NRxRywherein Rxand Ryare independently selected from a substituted or unsubstituted C6 to C30 aryl group and a substituted or unsubstituted C3 to C20 heteroaryl group. In addition, the functional group represented by Chemical Formula 1B may improve selective absorbance in an infrared/near infrared wavelength spectrum of light by further including a phenylene ring in front of an amine group. The functional group represented by Chemical Formula 1B may be one functional group of a plurality of functional groups represented by Chemical Formula 1B-1, Chemical Formula 1B-2, and Chemical Formula 1B-3. In Chemical Formula 1B-1,m is 1 or 2,Z1and Z2are independently hydrogen or a hydroxyl group,R21to R26are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,Raand Rbare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, anda and b are independently an integer that is inclusively between 0 to 5. In Chemical Formula 1B-2,m is 1 or 2,Z1and Z2are independently hydrogen or a hydroxyl group,R21to R26are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,Raand Rbare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, anda and b are independently an integer that is inclusively between 0 to 4. In Chemical Formula 1B-3,m is 1 or 2,Z1and Z2are independently hydrogen or a hydroxyl group,R21to R26are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,Raand Rbare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,Y is selected from NRc, O, S, Se, and Te (wherein Rcis selected from hydrogen and a substituted or unsubstituted C1 to C6 alkyl group), and a and b are independently an integer that is inclusively between 0 to 4. Chemical Formula 10 may be selected from functional groups (“one functional group of a plurality of functional groups”) represented by Chemical Formulae 1C-1 and 1C-2. In Chemical Formula 1C-1 and Chemical Formula 1C-2,Ra, Rb, Re, and Rdare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,a and b are independently an integer that is inclusively between 0 to 6,c is an integer that is inclusively between 0 to 2, ande is an integer that is inclusively between 0 to 3. Chemical Formula 1D may be selected from functional groups represented by Chemical Formula 1D-1, Chemical Formula 1D-2, and Chemical Formula 1D-3. In Chemical Formula 1D-1,k is 0 or 1,Z3and Z4are independently hydrogen or a hydroxyl group,R41and R42are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,Raand Rbare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, anda and b are independently an integer that is inclusively between 0 to 5. In Chemical Formula 1D-2 and Chemical Formula 1D-3,k is 0 or 1,Z3and Z4are independently hydrogen or a hydroxyl group,R41and R42are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,Raand Rbare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group,Y is selected from NRc, O, S, Se, and Te (wherein Rcis selected from hydrogen and a substituted or unsubstituted C1 to C6 alkyl group), and a and b are independently an integer that is inclusively between 0 to 4. The squarylium compound may be a particular compound represented by one chemical formula represented by Chemical Formula 4-1, Chemical Formula 4-2, Chemical Formula 4-3, Chemical Formula 4-4, Chemical Formula 4-5, Chemical Formula 4-6, Chemical Formula 4-7, Chemical Formula 4-8, Chemical Formula 4-9, and Chemical Formula 4-10. In Chemical Formula 4-1,R14, R15, R16, R17, R18, R19, R14′, R15′, R16′, R17′, R18′, R19′, Rp, and Rp′are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, and p and p′ are independently an integer that is inclusively between 0 to 4. In Chemical Formula 4-2,R14, R15, R16, R17, R18, R19, R14′, R15′, R16′, R17′, R18′, R19′, Rp, Rp′, Rq, and Rq′are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, p and p′ are independently an integer that is inclusively between 0 to 4, and q and q′ are independently an integer that is inclusively between 0 to 2. In Chemical Formula 4-3,R14, R15, R16, R17, R18, R19, R14′, R15′, R16′, R17′, R18′, R19′, Rp, Rp′, Rq, and Rq′are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, and p and p′ are independently an integer that is inclusively between 0 to 4. In Chemical Formula 4-4,R14, R15, R16, R17, R18, R19, Rp, Rr, and Rsare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, p is an integer that is inclusively between 0 to 4, and r and s are independently an integer that is inclusively between 0 to 5. In Chemical Formula 4-5,R14, R15, R16, R17, R18, R19, Rp, Rq, Rr, and Rsare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, p is an integer that is inclusively between 0 to 4, q is an integer that is inclusively between 0 to 2, and r and s are independently an integer that is inclusively between 0 to 5. In Chemical Formula 4-6,R14, R15, R16, R17, R18, R19, Rp, Rq, Rr, and Rsare independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, p is an integer that is inclusively between 0 to 4, q is an integer that is inclusively between 0 to 2, and r and s are independently an integer that is inclusively between 0 to 5. In Chemical Formula 4-7,R14, R15, R16, R17, R18, R19, Rp, Rq, Rr, Rs, and Rware independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, p and w are independently an integer that is inclusively between 0 to 4, and r and s are independently an integer that is inclusively between 0 to 5. In Chemical Formula 4-8,R14, R15, R16, R17, R18, R19, Rp, Rq, Rr, Rs, and Rware independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, p and w are independently an integer that is inclusively between 0 to 4, q is an integer that is inclusively between 0 to 2, and r and s are independently an integer that is inclusively between 0 to 5. In Chemical Formula 4-9,R14, R15, R16, R17, R18, R19, Rp, Rq, Rr, Rs, and Rware independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, p and w are independently an integer that is inclusively between 0 to 4, and r and s are independently an integer that is inclusively between 0 to 5. In Chemical Formula 4-10,Rr, Rs, Rw, Rr′, Rs′, and Rw′are independently selected from hydrogen, a halogen, a cyano group, a nitro group, a hydroxyl group, a carboxyl group, an ester group, a substituted or unsubstituted C1 to C20 alkyl group, a substituted or unsubstituted C1 to C20 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C20 cycloalkyl group, a substituted or unsubstituted C3 to C20 heteroaryl group, and a substituted or unsubstituted C2 to C20 heterocycloalkyl group, w and w′ are independently an integer that is inclusively between 0 to 4, and r, s, r′, and s′ are independently an integer that is inclusively between 0 to 5. As described above, the squarylium compound has improved selective light absorbance in an infrared/near infrared wavelength spectrum of light and thus may be applied to an infrared cut film. Hereinafter, an infrared cut film including the squarylium compound represented by Chemical Formula 1 is described. The infrared cut film may be manufactured by coating a composition including the squarylium compound represented by Chemical Formula 1 and an organic solvent on a transparent substrate and removing the organic solvent to manufacture a film. The organic solvent may include for example ethers such as dimethoxyethane, methoxyethoxyethane, tetrahydrofuran, dioxane, and the like, ketones such as acetone, methylethylketone, methylisobutylketone, cyclohexanone, and the like, aromatic hydrocarbons such as benzene, toluene, xylene, monochlorobenzene, and the like, which may be used in an amount of about 10 parts by weight to about 3000 parts by weight based on 1 part by weight of the squarylium compound represented by Chemical Formula 1. The composition may further include a binder, and the binder may be for example a polyester-based resin, a polycarbonate-based resin, a polyacrylic acid-based resin, a polystyrene-based resin, a polyvinyl chloride-based resin, a polyvinyl acetate-based resin, and the like. The binder may be used in an amount of about 10 parts by weight to about 500 parts by weight based on 1 part by weight of the squarylium compound represented by Chemical Formula 1. The composition including the squarylium compound represented by Chemical Formula 1 may be coated on the transparent substrate using a known method such as a bar coat method, a spray method, a roll coating method, a dipping method, and the like. In addition, the infrared cut film may be manufactured by dispersing the squarylium compound represented by Chemical Formula 1 in a resin, molding the resultant, and making it into a film. The resin may be selected from a polyester-based resin, a polycarbonate-based resin, a polyacrylic acid-based resin, a polystyrene-based resin, a polyvinyl chloride-based resin, and a polyvinyl acetate-based resin. The infrared cut film may be manufactured by further adhering a transparent substrate to a surface of a base film that is manufactured as above, as needed. The transparent substrate is not particularly as long as it is transparent resin or glass having low absorption and scattering properties. The resin may be for example a polyester-based resin, a polycarbonate-based resin, a polyacrylic acid-based resin, a polystyrene-based resin, a polyvinyl chloride-based resin, a polyvinyl acetate-based resin, and the like. The infrared cut film may be used as an infrared cut filter due to infrared ray/near infrared ray cutting performance. The infrared cut filter may include an infrared cut film including the squarylium compound represented by Chemical Formula 1 and an infrared light reflection layer disposed on the infrared cut film as needed. An infrared cut filter having such a structure is described referring toFIGS.1and2. In the drawings, the thickness of layers, films, panels, regions, etc., are exaggerated for clarity. Like reference numerals designate like elements throughout the disclosure. It will be understood that when an element such as a layer, film, region, or substrate is referred to as being “on” another element, it can be directly on the other element or intervening elements may also be present. In contrast, when an element is referred to as being “directly on” another element, there are no intervening elements present. FIG.1is a schematic cross-sectional view of an infrared cut filter according to some example embodiments. Referring toFIG.1, an infrared cut filter1according to some example embodiments includes an infrared cut film11including the squarylium compound represented by Chemical Formula 1 and an infrared light reflection layer13disposed on the infrared cut film11. The infrared cut film11may be manufactured by dispersing the squarylium compound represented by Chemical Formula 1 in a resin, molding the resultant, and making it into a film as described above. The infrared cut film11may be a glass substrate including the squarylium compound represented by Chemical Formula 1. The glass substrate may further include copper oxide. The infrared light reflection layer13may be a thin film including an inorganic particulate and may be a deposition film of an inorganic particulate or a metal deposition film. The inorganic particulate may be at least one particulate of silica (SiO2), titania (TiO2) alumina (Al2O3), zirconia, tantalum pentoxide, niobium pentoxide, lanthanum oxide, yttrium oxide, zinc oxide, zinc sulfide, indium oxide, tin oxide, lanthanum fluoride, magnesium fluoride, sodium hexafluoroaluminate, and the like. Examples of the metal deposition film may be an aluminum deposition film. The deposition film may be for example formed by depositing an inorganic particulate or a metal using a CVD method, a sputtering method, a vacuum deposition method, an ion-assist deposition method, an ion plating method, and the like on the infrared cut film11, but is not limited thereto. The infrared light reflection layer13may be a thin film that may be formed based on codepositing different kinds of inorganic particulates or a multi-layered thin film including deposited different kinds of inorganic particulates. For example, a first deposition film of an inorganic particulate that is at least one particulate of titania (TiO2), zirconia, and a combination thereof is formed on the infrared cut film11and a second deposition film of an inorganic particulate selected from silica (SiO2), alumina, and a combination thereof may be formed thereon. Such inorganic particulate deposition films of the infrared light reflection layer13may include 5 to 30 repeated layers. A thickness of the infrared cut film11may be about 50 μm to about 200 μm, for example about 55 μm to about 190 μm or about 60 μm to about 180 μm and a thickness of the infrared light reflection layer13may be about 0.1 μm to about 20 μm, for example about 0.5 μm to about 10 μm, or about 0.7 μm to about 5 μm. Within the ranges, infrared light cutting performance may be improved and mechanical strength may also be ensured. FIG.2is a schematic cross-sectional view of an infrared cut filter according to some example embodiments. Referring toFIG.2, an infrared cut filter2according to some example embodiments includes an infrared cut film21including the squarylium compound represented by Chemical Formula 1 and a first infrared light reflection layer23and a second infrared light reflection layer25disposed on both surfaces of the infrared cut film21. The infrared cut film21may be manufactured by dispersing the squarylium compound represented by Chemical Formula 1 in a resin, molding the resultant, and making it into a film as described above. The infrared cut film21may be a glass substrate including the squarylium compound represented by Chemical Formula 1. The glass substrate may further include copper oxide. The first infrared light reflection layer23and the second infrared light reflection layer25may be a thin film including an inorganic particulate and may be a deposition film of an inorganic particulate or a metal deposition film. The inorganic particulate may be silica (SiO2), titania (TiO2) alumina (Al2O3), zirconia, tantalum pentoxide, niobium pentoxide, lanthanum oxide, yttrium oxide, zinc oxide, zinc sulfide, indium oxide, tin oxide, lanthanum fluoride, magnesium fluoride, sodium hexafluoroaluminate, and the like. Examples of the metal deposition film may be an aluminum deposition film. The deposition film may be for example formed by depositing an inorganic particulate or a metal using a CVD method, a sputtering method, a vacuum deposition method, an ion-assist deposition method, an ion plating method, and the like on the infrared cut film21, but is not limited thereto. The first infrared light reflection layer23and the second infrared light reflection layer25reflect light in an infrared ray wavelength spectrum of light effectively, and thereby optical distortion by light in an infrared ray wavelength spectrum of light may be effectively reduced or prevented. The first infrared light reflection layer23and the second infrared light reflection layer25may reflect light in a part of a near infrared wavelength spectrum of light, a mid-infrared wavelength spectrum of light, and a far-infrared wavelength spectrum of light, for example light in a wavelength spectrum of light of about 700 nm to 3 μm. The first infrared light reflection layer23and the second infrared light reflection layer25is not particularly limited as long as it reflects light in an infrared ray wavelength spectrum of light, and may be for example a high refractive reflective layer, a reflective layer including a nano particle having a high refractive index or a multilayer including a plurality of layers having different refractive indexes, but is not limited thereto. The first infrared light reflection layer23and the second infrared light reflection layer25may be a thin film obtained by codepositing different kinds of inorganic particulates or a multi-layered thin film obtained by depositing different kinds of inorganic particulates. For example, the first infrared light reflection layer23and the second infrared light reflection layer25may include a first layer and a second layer consisting of each material having a different refractive index and may include a multilayer including the first layer and the second layer that are alternately and repeatedly stacked. Each of the first layer and the second layer may be for example a dielectric layer including oxide layer, a nitride layer, an oxynitride layer, a sulfide layer, or a combination thereof, and for example the first layer may have a refractive index of less than about 1.7 and the second layer may have a refractive index of greater than or equal to about 1.7. Within the ranges, for example the first layer may have a refractive index of greater than or equal to about 1.1 and less than 1.7 and the second layer may have a refractive index of about 1.7 to about 2.7, or within the ranges, for example the first layer may have a refractive index of about 1.2 to about 1.6 and the second layer may have a refractive index of about 1.8 to about 2.5. The first layer and the second layer may include a material having the refractive indexes without a particular limit, and the first layer may include for example silicon oxide, aluminum oxide, or a combination thereof and the second layer may include titanium oxide, zinc oxide, indium oxide, zirconium oxide, or a combination thereof. The first layer and the second layer may have for example five layers to eighty layers, for example five layers to fifty layers. Each thickness of the first layer and the second layer may be determined according to a refractive index and a reflection wavelength of each layer and for example each first layer may have a thickness of about 10 nm to about 700 nm and each second layer may have a thickness of about 30 nm to about 600 nm. Thicknesses of the first layer and the second layer may be the same or different. A thickness of the infrared cut film21may be about 50 μm to about 200 μm, for example about 55 μm to about 190 μm or about 60 μm to about 180 μm and each thickness of the first infrared light reflection layer23and the second infrared light reflection layer25may be about 0.1 μm to about 20 μm, for example about 0.5 μm to about 10 μm, or about 0.7 μm to about 5 μm. Within the ranges, infrared light cutting performance may be improved and mechanical strength may also be ensured. As described above, the squarylium compound may be applied to various electronic devices due to improved selective light absorbance in an infrared/near infrared wavelength spectrum of light. The electronic devices may be for example an image sensor, a liquid crystal display, a plasma display, an organic electroluminescence display, a laser display, a solar cell, a bio sensor, an illumination, and the like. Particularly, a compound having improved absorbance in an infrared/near infrared wavelength spectrum of light such as the squarylium compound may improve spectral sensitivity at a low illumination, and may be usefully used in an iris identification sensor, a night vision device, and the like. Hereinafter, an image sensor as an example of an electronic device id described with reference to the drawings. An image sensor according to some example embodiments includes a first photo-sensing device configured to sense light in a blue wavelength spectrum of light,a second photo-sensing device configured to sense light in a red wavelength spectrum of light,a third photo-sensing device configured to sense light in a green wavelength spectrum of light, anda fourth photo-sensing device configured to sense light in an infrared/near infrared wavelength spectrum of light,wherein the fourth photo-sensing device may include the squarylium compound. The first photo-sensing device to the fourth photo-sensing device may be arranged (may extend) adjacently and parallel or perpendicular, to each other collectively in the form of a single group. The first photo-sensing device to the fourth photo-sensing device may each be an inorganic photodiode or an organic photodiode and at least one of the first photo-sensing device to the fourth photo-sensing device may be an organic photodiode. The inorganic photodiode may be for example a silicon photodiode, but is not limited thereto. The organic photodiode may be an organic photoelectric device including a pair of light-transmitting electrodes facing each other and a photoactive layer disposed between them and including an organic light-absorbing material. One of the pair of light-transmitting electrodes may be an anode and the other may be a cathode. The light-transmitting electrodes may be made of, for example, a transparent conductor such as indium tin oxide (ITO), indium zinc oxide (IZO), zinc oxide (ZnO), tin oxide (SnO), aluminum tin oxide (AITO), and fluorine-doped tin oxide (FTO), or may be a metal thin layer having a thin thickness of several nanometers or several tens of nanometers or a metal thin layer having a thin thickness of several nanometers to several tens of nanometers doped with a metal oxide. The photoactive layer is a layer including a p-type semiconductor material and an n-type semiconductor material to provide a pn junction, which is a layer producing excitons by receiving light from outside and then separating holes and electrons from the produced excitons. The photoactive layer may include an intrinsic layer including both the p-type semiconductor and the n-type semiconductor and may be formed according to a method of, for example, co-deposition and the like. In addition, the photoactive layer may further include at least one selected from a p-type layer and an n-type layer besides the intrinsic layer, wherein the p-type layer may include a p-type semiconductor material, and the n-type layer may include an n-type semiconductor material. The kind of the p-type semiconductor material and the n-type semiconductor material may be determined according to the absorption wavelength. At least one of charge auxiliary layers may be further included between the light-transmitting electrode and the photoactive layer. The charge auxiliary layer may further facilitate the movement of holes and electrons separated from the photoactive layer to enhance efficiency, and may be at least one selected from, for example, a hole injection layer (HIL) facilitating hole injection, a hole transport layer (HTL) facilitating hole transportation, an electron blocking layer (EBL) blocking electron transportation, an electron injection layer (EIL) facilitating electron injection, an electron transport layer (ETL) facilitating electron transportation, and a hole blocking layer (HBL) blocking hole transportation. The hole transport layer (HTL) may include, for example one selected from poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate) (PEDOT:PSS), polyarylamine, poly(N-vinylcarbazole), polyaniline, polypyrrole, N,N,N′,N′-tetrakis(4-methoxyphenyl)-benzidine (TPD), 4-bis[N-(1-naphthyl)-N-phenyl-amino]biphenyl (α-NPD), m-MTDATA, 4,4′,4″-tris(N-carbazolyl)-triphenylamine (TCTA), tungsten oxide (WOx, 0<x≤3), molybdenum oxide (MOx, 1<x≤3), vanadium oxide (V2O5), rhenium oxide, nickel oxide (NiOx, 1<x≤4), copper oxide, titanium oxide, molybdenum sulfide, and a combination thereof, but is not limited thereto. The electron blocking layer (EBL) may include one selected from, for example, poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate) (PEDOT:PSS), polyarylamine, poly(N-vinylcarbazole), polyaniline, polypyrrole, N,N,N′,N′-tetrakis(4-methoxyphenyl)-benzidine (TPD), 4-bis[N-(1-naphthyl)-N-phenyl-amino]biphenyl (α-NPD), m-MTDATA, 4,4′,4″-tris(N-carbazolyl)-triphenylamine (TCTA), and a combination thereof, but is not limited thereto. The electron transport layer (ETL) may include one selected from, for example, 1,4,5,8-naphthalene-tetracarboxylic dianhydride (NTCDA), bathocuproine (BCP), LiF, Alq3, Gaq3, Inq3, Znq2, Zn(BTZ)2, BeBq2, aluminum (Al), magnesium (Mg), molybdenum (Mo), aluminum oxide, magnesium oxide, molybdenum oxide, and a combination thereof, but is not limited thereto. The hole blocking layer (HBL) may include one selected from, for example 1,4,5,8-naphthalene-tetracarboxylic dianhydride (NTCDA), dicyanovinyl-terthiophene (DCV3T), bathocuproine (BCP), LiF, Alq3, Gaq3, Inq3, Znq2, Zn(BTZ)2, BeBq2, and a combination thereof, but is not limited thereto. The organic photoelectric device may produce excitons at the inside thereof when light in a predetermined region is adsorbed in the photoactive layer by entering light from one light-transmitting electrode side. The excitons are separated into holes and electrons in the photoactive layer, and the separated holes may be transported into an anode side and the separated electrons may be transported into a cathode side to flow current through the organic photoelectric device. In an image sensor according to some example embodiments, the first photo-sensing device, the second photo-sensing device, the third photo-sensing device, and the fourth photo-sensing device may be an inorganic photodiode. FIG.3is a schematic cross-sectional view of an image sensor according to some example embodiments. Referring toFIG.3, an image sensor100includes a semiconductor substrate110integrated with a blue photodiode50B, a green photodiode50G, a red photodiode50R, an infrared light/near infrared light diode (infrared photodiode)501R, and a transmission transistor (not shown), a lower insulation layer65, a color filter layer70, an upper insulation layer85, and an infrared cut filter701R. The semiconductor substrate110may be a silicon substrate and may be integrated with the blue photodiode50B, the green photodiode50G, the red photodiode50R, the infrared light/near infrared light diode501R, and the transmission transistor (not shown). The blue photodiode50B, the green photodiode50G, the red photodiode50R may be respectively integrated in each of a blue pixel, a green pixel, and a red pixel. The blue photodiode50B, the green photodiode50G, the red photodiode50R, and the infrared light/near infrared light diode501R may sense light, and the sensed information may be transferred by a transport transistor. The transmission transistor may transfer photocharges generated by the photodiode to a driving transistor (not shown). Metal wires (not shown) and pads (not shown) are formed on the semiconductor substrate110. In order to decrease signal delay, the metal wires and pads may be made of a metal having low resistivity, for example, aluminum (Al), copper (Cu), silver (Ag), and alloys thereof, but are not limited thereto. The lower insulation layer65may be formed on the metal wires and pads. The lower insulation layer65may be made of an inorganic insulation material such as a silicon oxide and/or a silicon nitride, or a low dielectric constant (low K) material such as SiC, SiCOH, SiCO, and SiOF. The color filter layer70formed on the lower insulation layer65includes a blue filter70B formed in a blue pixel, a green filter70G formed in a green pixel, and a red filter70R formed in a red pixel. The upper insulation layer85is formed on the color filter layer70. The upper insulation layer85removes steps caused by the color filter layer70, and planarize it. The upper insulation layer85and the lower insulation layer65may include a contact hole (not shown) to expose pads. The infrared cut filter701R is formed on the upper insulation layer85. The infrared cut filter701R includes the squarylium compound represented by Chemical Formula 1. The infrared cut film (IR) may selectively absorb light in an infrared ray (particularly, near infrared ray) region of greater than or equal to about 700 nm and less than or equal to about 1300 nm without absorption in a visible wavelength spectrum of light. As described above, the color filter layer70including the color filters70B,70G, and70R absorbing light in a visible ray region and the infrared cut filter701R are vertically stacked and thereby an area absorbing infrared light may be enlarged and absorption efficiency may be increased. A focusing lens (not shown) may be further formed on the infrared cut filter701R. The focusing lens may control a direction of incident light and gather the light in one region. The focusing lens may have a shape of, for example, a cylinder or a hemisphere, but is not limited thereto. As shown inFIG.4, an image sensor200includes a focusing lens90formed on the upper insulation layer85and the infrared cut filter701R formed on the focusing lens90. In an image sensor according to some example embodiments, the infrared cut filter701R may be disposed only on an infrared light/near infrared light diode.FIG.5is a schematic cross-sectional view showing such an image sensor30. In an image sensor according to some example embodiments, the first photo-sensing device (“photodiode”), the second photo-sensing device, and the third photo-sensing device may be an organic photodiode and the fourth photo-sensing device may be an inorganic photodiode. FIG.6is a schematic cross-sectional view showing an image sensor according to some example embodiments. Referring toFIG.6, an image sensor400according to some example embodiments includes a semiconductor substrate110integrated with an infrared photodiode501R, a blue charge storage55B, a green charge storage55G, a red charge storage55R, and a transmission transistor (not shown), a lower insulation layer65, an upper insulation layer85, a blue photo-sensing device100B, a green photo-sensing device1000, and a red photo-sensing device100R. The semiconductor substrate110may be a silicon substrate, and may be integrated with the infrared photodiode501R, the blue charge storage55B, the green charge storage55G, the red charge storage55R, and the transmission transistor (not shown). The blue charge storage55B, the green charge storage55G, and the red charge storage55R may be respectively integrated in each of a blue pixel, a green pixel, and a red pixel. The infrared photodiode501R may absorb light in an infrared ray (particularly, a near infrared ray) region and the sensed information may be transferred by a transport transistor. Charges absorbed in the blue photo-sensing device100B, the green photo-sensing device1000, and the red photo-sensing device100R are collected in the blue charge storage55B, the green charge storage55G, and the red charge storage55R which are electrically connected to each of the blue photo-sensing device100B, the green photo-sensing device1000, and the red photo-sensing device100R. Metal wires (not shown) and pads (not shown) are formed on the semiconductor substrate110. In order to decrease signal delay, the metal wires and pads may be made of a metal having low resistivity, for example, aluminum (Al), copper (Cu), silver (Ag), and alloys thereof, but are not limited thereto. The lower insulation layer65may be formed on the metal wires and pads. The lower insulation layer65may be made of an inorganic insulation material such as a silicon oxide and/or a silicon nitride, or a low dielectric constant (low K) material such as SiC, SiCOH, SiCO, and SiOF. The blue photo-sensing device100B, the green photo-sensing device1000, and the red photo-sensing device100R are formed on the lower insulation layer65. The blue photo-sensing device100B includes a lower electrode10B, an upper electrode20B, and a photoactive layer30B selectively absorbing light in a blue wavelength spectrum of light, the green photo-sensing device100G includes a lower electrode10G, an upper electrode20G and a photoactive layer30G selectively absorbing light in a green wavelength spectrum of light, and the red photo-sensing device100R includes a lower electrode10R, an upper electrode20R, and a photoactive layer30R selectively absorbing light in a red wavelength spectrum of light. The lower electrodes10B,10G, and10R and the upper electrodes20B,20G, and20R may be light-transmitting electrodes and may be made of, for example, a transparent conductor such as indium tin oxide (ITO), indium zinc oxide (IZO), zinc oxide (ZnO), tin oxide (SnO), aluminum tin oxide (AITO), and fluorine-doped tin oxide (FTO), or may be a metal thin layer having a thin thickness of several nanometers or several tens of nanometers or a metal thin layer having a thin thickness of several nanometers to several tens of nanometers doped with a metal oxide. The photoactive layers30B,30G, and30R may include a p-type semiconductor material and an n-type semiconductor material. The photoactive layer30B of the blue photo-sensing device100B may include a p-type semiconductor material selectively absorbing light in a blue wavelength spectrum of light and an n-type semiconductor material selectively absorbing light in a blue wavelength spectrum of light, the photoactive layer30G of the green photo-sensing device100G may include a p-type semiconductor material selectively absorbing light in a green wavelength spectrum of light and an n-type semiconductor material selectively absorbing light in a green wavelength spectrum of light, and the photoactive layer30R of the red photo-sensing device100R may include a p-type semiconductor material selectively absorbing light in a red wavelength spectrum of light and an n-type semiconductor material selectively absorbing light in a red wavelength spectrum of light. The upper insulation layer85is formed on the lower insulation layer65. The upper insulation layer85is disposed on the infrared photodiode501R and may reduce steps with the blue photo-sensing device100B, the green photo-sensing device1000, and the red photo-sensing device100R. The infrared cut filter70IR is disposed on the blue photo-sensing device1006, the green photo-sensing device1000, the red photo-sensing device100R, and the upper insulation layer85. The infrared cut filter701R includes the squarylium compound represented by Chemical Formula 1. The infrared cut film (IR) may selectively absorb light in an infrared ray (particularly, near infrared ray) region of greater than or equal to about 700 nm and less than or equal to about 1300 nm without absorption in a visible wavelength spectrum of light. As shown inFIG.6, the infrared cut filter701R is formed on an entire surface of a blue pixel, a green pixel, and a red pixel, and thereby an area absorbing infrared light may be enlarged and absorption efficiency may be increased. The blue photo-sensing device1006, the green photo-sensing device1000, and the red photo-sensing device100R may be vertically stacked. In this way, the area of the image sensor may be decreased and down-sizing of the image sensor may be implemented by stacking the photo-sensing devices1006,1000, and100R vertically. A stacking order of the photo-sensing devices1006,1000, and100R are not particularly limited. A focusing lens (not shown) may be further formed on the infrared ray filter701R. The focusing lens may control a direction of incident light and gather the light in one region. The focusing lens may have a shape of, for example, a cylinder or a hemisphere, but is not limited thereto. In addition, the image sensor400may include a focusing lens formed on the blue photo-sensing device100B, the green photo-sensing device1000, the red photo-sensing device100R, and the upper insulation layer85, and the infrared ray filter70IR formed on the focusing lens. The infrared ray filter701R ofFIG.6may be formed at a position corresponding to the infrared light/near infrared light diode501R as shown inFIG.5. In an image sensor according to some example embodiments, the third photo-sensing device may be an organic photodiode and the first photo-sensing device, the second photo-sensing device, and the fourth photo-sensing device may be an inorganic photodiode. FIG.7is a schematic cross-sectional view showing an image sensor according to some example embodiments. Referring toFIG.7, an image sensor50according to some example embodiments includes a semiconductor substrate110integrated with a blue photodiode50B, a red photodiode50R, a green charge storage55G, an infrared light/near infrared light diode50IR, and a transmission transistor (not shown), a lower insulation layer65, color filter layers (“color filters”)70B and70R, a first upper insulation layer85a, a green photo-sensing device1000, a second upper insulation layer85b, and an infrared ray filter70IR. The semiconductor substrate110may be a silicon substrate and may be integrated with the blue photodiode50B, the red photodiode50R, the green charge storage55G, the infrared light/near infrared light diode50IR, and the transmission transistor (not shown). The blue photodiode50B and the transmission transistor may be integrated in each of a blue pixel, the red photodiode50R and the transmission transistor integrated in each of a red pixel, and the green charge storage55G and the transmission transistor integrated in each of a green pixel. Metal wires (not shown) and pads (not shown) are formed on the semiconductor substrate110. In order to decrease signal delay, the metal wires and pads may be made of a metal having low resistivity, for example, aluminum (Al), copper (Cu), silver (Ag), and alloys thereof, but are not limited thereto. However, the image sensor is not limited to the structure and the metal wires and pads may be disposed under the blue photodiode50B, the red photodiode50R, the green charge storage55G, and the infrared light/near infrared light diode501R. The lower insulation layer65may be formed on the metal wires and pads. The lower insulation layer65may be made of an inorganic insulation material such as a silicon oxide and/or a silicon nitride, or a low dielectric constant (low K) material such as SiC, SiCOH, SiCO, and SiOF. Color filters70B and70R may be formed on the lower insulation layer65. The color filter70B of the blue pixel adsorbs light in the blue wavelength spectrum of light and transfers it to the blue photo-sensing device50B, and the color filter70R of the red pixel adsorbs light in the red wavelength spectrum of light and transfers it to the red photo-sensing device50R. The green pixel does not include a color filter. The first upper insulation layer85ais formed on the color filters70B and70R. The first upper insulation layer85aremoves steps caused by the color filter70B and70R, and planarizes it. The green photo-sensing device100G and the second upper insulation layer85bare formed on the first upper insulation layer85a. The green photo-sensing device100G includes light-transmitting electrodes10G and20G and a photoactive layer30G. One of the light-transmitting electrodes10G and20G may be an anode and the other may be a cathode. The light-transmitting electrodes10G and20G may be made of, for example, a transparent conductor such as indium tin oxide (ITO), indium zinc oxide (IZO), zinc oxide (ZnO), tin oxide (SnO), aluminum tin oxide (AITO), and fluorine-doped tin oxide (FTO), or may be a metal thin layer having a thin thickness of several nanometers or several tens of nanometers or a metal thin layer having a thin thickness of several nanometers to several tens of nanometers doped with a metal oxide. The photoactive layer30G selectively absorbs light in a green wavelength spectrum of light and passes light in other wavelength spectrum of lights except the green wavelength spectrum of light, which are the blue wavelength spectrum of light and the red wavelength spectrum of light. The photoactive layer30G may include a p-type semiconductor compound selectively adsorbing light in the green wavelength spectrum of light and an n-type semiconductor compound selectively adsorbing light in the green wavelength spectrum of light, and the p-type semiconductor compound and the n-type semiconductor compound may provide a pn junction. The photoactive layer30G selectively adsorbs light in the green wavelength spectrum of light and produces excitons, and then the produced excitons are separated into holes and electrons to impart the photoelectric effects. The photoactive layer30G may be substituted for a color filter of the green pixel. Each of the p-type semiconductor material and the n-type semiconductor material may have an energy bandgap of, for example, about 2.0 to about 2.5 eV, and the p-type semiconductor material and the n-type semiconductor material may have a LUMO difference of, for example, about 0.2 to about 0.7 eV. The p-type semiconductor material may be, for example, quinacridone or a derivative thereof, and the n-type semiconductor material may be, for example, a cyanovinyl group-containing a thiophene derivative, but they are not limited thereto. The green photo-sensing device100G may produce excitons at the inside when light enters from the upper electrode20G, and the photoactive layer30G absorbs light in the green wavelength spectrum of light. Excitons are separated into holes and electrons in the photoactive layer30G, and the separated holes are moved to the anode side, which is one of the lower electrode10G and the upper electrode20G, and the separated electrons are moved to a cathode which is the other of the lower electrode10G and the upper electrode20G, so as to flow a current. The separated electrons or holes may be collected in the charge storage55G. Light in other wavelength spectrum of lights except the green wavelength spectrum of light may pass through the green photo-sensing device100G and the color filters70B and70R, and may be sensed by the blue photo-sensing device50B or the red photo-sensing device50R. The photoactive layer30G may be formed on an entire surface of the blue pixel (B), the red pixel (R), and the green pixel (G), such that the light absorption area is increased to accomplish the high light-absorptive efficiency. The second upper insulation layer85bmay be disposed on the infrared photodiode501R and may reduce steps with the green photo-sensing device100G. The infrared cut filter701R is formed on the green photo-sensing device100G and the upper insulation layer80b. The infrared cut filter701R includes the squarylium compound represented by Chemical Formula 1. The infrared cut film (IR) may selectively absorb light in an infrared ray (particularly, near infrared ray) region of greater than or equal to about 700 nm and less than or equal to about 1300 nm without absorption in a visible wavelength spectrum of light. A focusing lens (not shown) may be further formed on the infrared ray filter701R. The focusing lens may control a direction of incident light and gather the light in one region. The focusing lens may have a shape of, for example, a cylinder or a hemisphere, but is not limited thereto. In addition, the image sensor50may include a focusing lens formed on the green photo-sensing device1000and the second upper insulation layer85band the infrared cut filter701R formed on the focusing lens. In some example embodiments, for better understanding and ease of description, the structure in which the green photo-sensing device100G is stacked is exemplified, but it is not limited thereto. The structure may be stacked with the red photo-sensing device100R or the blue photo-sensing device100B instead of the green photo-sensing device100G. As described above, an area of the image sensor may be decreased and down-sizing of the image sensor may be implemented by vertically stacking a color filter layer including color filters absorbing light in a blue wavelength spectrum of light and light in a red wavelength spectrum of light of a visible ray region, a green photo-sensing device absorbing light in a green wavelength spectrum of light, and an infrared cut filter absorbing light in an infrared light. In addition, a photo-sensing device selectively absorbing light in a green wavelength spectrum of light and an infrared cut filter are formed on an entire surface of an image sensor and an area absorbing light may be enlarged and absorption efficiency may be increased. In an image sensor according to some example embodiments, the first photo-sensing device, the second photo-sensing device, the third photo-sensing device, and the fourth photo-sensing device may be an organic photodiode. FIG.8is a schematic cross-sectional view showing an image sensor according to some example embodiments. Referring toFIG.8, an image sensor600according to some example embodiments includes a semiconductor substrate110integrated with an infrared light/near infrared light charge storage551R, a blue charge storage55B, a green charge storage55G, a red charge storage55R, and a transmission transistor (not shown), a lower insulation layer65, a blue photo-sensing device100B, a green photo-sensing device1000, a red photo-sensing device100R and an infrared/near infrared photo-sensing device100IR. The semiconductor substrate110may be a silicon substrate and may be integrated with the infrared light/near infrared light charge storage551R, the blue charge storage55B, the green charge storage55G, the red charge storage55R, and the transmission transistor (not shown). The blue charge storage55B, the green charge storage55G, and the red charge storage55R may be respectively integrated in each of a blue pixel, a green pixel, and a red pixel. Charges absorbed in the infrared/near infrared photo-sensing device100IR, the blue photo-sensing device100B, the green photo-sensing device1000, and the red photo-sensing device100R are collected in in the infrared light/near infrared light charge storage551R, the blue charge storage55B, the green charge storage55G, and the red charge storage55R which are electrically connected to each of the infrared/near infrared photo-sensing device100IR, the blue photo-sensing device100B, the green photo-sensing device1000, and the red photo-sensing device100R. Metal wires (not shown) and pads (not shown) are formed on the semiconductor substrate110. In order to decrease signal delay, the metal wires and pads may be made of a metal having low resistivity, for example, aluminum (Al), copper (Cu), silver (Ag), and alloys thereof, but are not limited thereto. The lower insulation layer65may be formed on the metal wires and pads. The lower insulation layer65may be made of an inorganic insulation material such as a silicon oxide and/or a silicon nitride, or a low dielectric constant (low K) material such as SiC, SiCOH, SiCO, and SiOF. The blue photo-sensing device100B, the green photo-sensing device1000, the red photo-sensing device100R, and the infrared/near infrared photo-sensing device100IR are formed on the lower insulation layer65. The blue photo-sensing device100B includes a lower electrode10B, an upper electrode20B, and a photoactive layer30B selectively absorbing light in a blue wavelength spectrum of light, the green photo-sensing device100G includes a lower electrode10G, an upper electrode20G and a photoactive layer30G selectively absorbing light in a green wavelength spectrum of light, the red photo-sensing device100R includes a lower electrode10R, an upper electrode20R, and a photoactive layer30R selectively absorbing light in a red wavelength spectrum of light, and the infrared/near infrared photo-sensing device100IR includes a lower electrode10IR, an upper electrode201R, and a photoactive layer301R selectively absorbing light in an infrared/near infrared wavelength spectrum of light. The lower electrodes10B,10G,10R, and10IR and the upper electrodes20B,20G,20R, and201R may be light-transmitting electrodes and may be made of, for example, a transparent conductor such as indium tin oxide (ITO), indium zinc oxide (IZO), zinc oxide (ZnO), tin oxide (SnO), aluminum tin oxide (AITO), and fluorine-doped tin oxide (FTO), or may be a metal thin layer having a thin thickness of several nanometers or several tens of nanometers or a metal thin layer having a thin thickness of several nanometers to several tens of nanometers doped with a metal oxide. The photoactive layers30B,30G,30R, and301R may include a p-type semiconductor material and an n-type semiconductor material. The photoactive layer30B of the blue photo-sensing device100B may include a p-type semiconductor material selectively absorbing light in a blue wavelength spectrum of light and an n-type semiconductor material selectively absorbing light in a blue wavelength spectrum of light, the photoactive layer30G of the green photo-sensing device100G may include a p-type semiconductor material selectively absorbing light in a green wavelength spectrum of light and an n-type semiconductor material selectively absorbing light in a green wavelength spectrum of light, the photoactive layer30R of the red photo-sensing device100R may include a p-type semiconductor material selectively absorbing light in a red wavelength spectrum of light and an n-type semiconductor material selectively absorbing light in a red wavelength spectrum of light, and the photoactive layer301R of the infrared/near infrared photo-sensing device100IR may include a p-type semiconductor material selectively absorbing light in an infrared wavelength spectrum of light and an n-type semiconductor material selectively absorbing light in an infrared wavelength spectrum of light. The photoactive layer301R of the infrared/near infrared photo-sensing device100IR uses the squarylium compound represented by Chemical Formula 1 as a p-type semiconductor material and sub-phthalocyanine or a sub-phthalocyanine derivative, fullerene or a fullerene derivative, thiophene or a thiophene derivative, or a combination thereof as an n-type semiconductor material. The fullerene may include C60, C70, C76, C78, C80, C82, C84, C90, C96, C240, C540, a mixture thereof, a fullerene nanotube, and the like. The fullerene derivative may refer to compounds of these fullerenes having a substituent attached thereto. The fullerene derivative may include a substituent such as alkyl group, aryl group, or a heterocyclic group. Examples of the aryl groups and heterocyclic groups may be are a benzene ring, a naphthalene ring, an anthracene ring, a phenanthrene ring, a fluorene ring, a triphenylene ring, a naphthacene ring, a biphenyl ring, a pyrrole ring, a furan ring, a thiophene ring, an imidazole ring, an oxazole ring, a thiazole ring, a pyridine ring, a pyrazine ring, a pyrimidine ring, a pyridazine ring, an indolizine ring, an indole ring, a benzofuran ring, a benzothiophene ring, an isobenzofuran ring, a benzimidazole ring, an imidazopyridine ring, a quinolizidine ring, a quinoline ring, a phthalazine ring, a naphthyridine ring, a quinoxaline ring, a quinoxazoline ring, an isoquinoline ring, a carbazole ring, a phenanthridine ring, an acridine ring, a phenanthroline ring, a thianthrene ring, a chromene ring, an xanthene ring, a phenoxathin ring, a phenothiazine ring, or a phenazine ring. The infrared/near infrared photo-sensing device100IR may selectively absorb light in an infrared ray (particularly, near infrared ray) region of greater than or equal to about 700 nm and less than or equal to about 1300 nm without absorption in a visible wavelength spectrum of light. FIGS.9and10are schematic cross-sectional views of an image sensor according to some example embodiments. Referring toFIG.9, an image sensor700includes a semiconductor substrate110integrated with an infrared light/near infrared light charge storage551R, a blue charge storage55B, a green charge storage55G, a red charge storage55R, and a transmission transistor (not shown), a lower insulation layer65, a blue photo-sensing device100B, a green photo-sensing device1000, a red photo-sensing device100R, and an infrared/near infrared photo-sensing device100IR. The infrared/near infrared photo-sensing device100IR is formed on an entire surface of the blue photo-sensing device100B, the green photo-sensing device1000, and the red photo-sensing device100R. Other structures are the same as the image sensor ofFIG.8. In the structure ofFIG.9, the infrared/near infrared photo-sensing device100IR may be disposed on the lower insulation layer65and the blue photo-sensing device100B, the green photo-sensing device1000, and the red photo-sensing device100R may be disposed thereon. An image sensor having such a structure is shown inFIG.10. The infrared/near infrared photo-sensing device100IR may selectively absorb light in an infrared ray (particularly, near infrared ray) region of greater than or equal to about 700 nm and less than or equal to about 1300 nm without absorption in a visible wavelength spectrum of light and may improve efficiency due to a large absorption area. FIG.11is a schematic cross-sectional view of an image sensor according to some example embodiments. Referring toFIG.11, an image sensor900includes a semiconductor substrate110integrated with a blue charge storage55B, a green charge storage55G, a red charge storage55R, and a transmission transistor (not shown); a lower insulation layer65, a color filter layer (70) and a upper insulation layer85on the semiconductor substrate110; and an infrared/near infrared photo-sensing device100IR under the semiconductor substrate110. FIG.12is a schematic cross-sectional view of an image sensor according to some example embodiments. Referring toFIG.12, an image sensor1000includes a semiconductor substrate110integrated with a blue photodiode50B, a red photodiode50R, a green photodiode50G, an infrared light/near infrared light charge storage55IR, and a transmission transistor (not shown); a lower insulation layer65; a blue filter70B, a green filter70G; a red filter70R; a upper insulation layer85a; and an infrared/near infrared photo-sensing device100IR. FIG.13is a schematic cross-sectional view of an image sensor according to some example embodiments. Referring toFIG.13, an image sensor1100includes a semiconductor substrate110integrated with an infrared light/near infrared light charge storage551R, a blue storage55B, a green storage55G, a red storage55R and a transmission transistor (not shown); a lower insulation layer65; a blue photo-sensing device100B, a green photo-sensing device1000, a red photo-sensing device100R, an infrared/near infrared photo-sensing device100IR, a blue filter70B, a green filter70G, and a red filter70R. FIG.14is a schematic cross-sectional view of an image sensor according to some example embodiments. Referring toFIG.14, an image sensor1200includes a semiconductor substrate110integrated with an infrared light/near infrared light charge storage551R, a blue storage55B, a green storage55G, a red storage55R and a transmission transistor (not shown); a lower insulation layer65; a blue photo-sensing device100B, a green photo-sensing device1000, a red photo-sensing device100R, an infrared/near infrared photo-sensing device100IR, a blue filter70B, a green filter70G, and a red filter70R. FIG.15is a schematic cross-sectional view of an image sensor according to some example embodiments. Referring toFIG.15, an image sensor1300includes a semiconductor substrate110integrated with an infrared light/near infrared light charge storage551R, a blue storage55B, a green storage55G, a red storage55R and a transmission transistor (not shown); a lower insulation layer65; a blue filter70B, a red filter70R; a upper insulation layers85aand85b; a green photo-sensing device1000; and an infrared/near infrared photo-sensing device100IR. FIG.16is a schematic cross-sectional view of an image sensor according to some example embodiments. In the image sensor1400ofFIG.16, the blue photodiode50B and the red photodiode50R are stacked perpendicularly, differing from the image sensor1300ofFIG.15. FIG.17is a schematic cross-sectional view of an image sensor according to some example embodiments. Referring toFIG.17, an image sensor1500includes a semiconductor substrate110integrated with an infrared light/near infrared light charge storage551R, a blue storage55B, a green storage55G, a red storage55R and a transmission transistor (not shown); a lower insulation layer65; a blue filter70B, a red filter70R; a upper insulation layers85aand85b; an infrared/near infrared photo-sensing device100IR; and a green photo-sensing device100G. FIG.18is a schematic cross-sectional view of an image sensor according to some example embodiments. In the image sensor1600ofFIG.18, the blue photodiode50B and the red photodiode50R are stacked perpendicularly, differing from the image sensor1500ofFIG.17. The image sensor may be applied to various electronic devices, for example, a mobile phone, a digital camera, and the like but is not limited thereto. FIG.19is a block diagram of a digital camera including an image sensor according to some example embodiments. Referring toFIG.19, a digital camera1900includes a lens1010, an image sensor1020, a motor unit1030, and an engine unit1040. The image sensor1020may be one of image sensors according to embodiments shown inFIGS.2to18. The lens1010concentrates incident light on the image sensor1020. The image sensor1020generates RGB data for received light through the lens1010. In some embodiments, the image sensor1020may interface with the engine unit1040. The motor unit1030may adjust the focus of the lens1010or perform shuttering in response to a control signal received from the engine unit1040. The engine unit1040may control the image sensor1020and the motor unit1030. The engine unit1040may be connected to a host/application1050. Hereinafter, some example embodiments are illustrated in more detail with reference to examples. However, it will be understood that these are examples, and the present disclosure is not limited thereto. Synthesis Example I Synthesis Example 1 2.86 g (20 mmol) of 2-methylquinoline (Compound (1)) and 2.66 g (24 mmol) of selenium dioxide are added to 50 ml of dioxane and reacted at 120° C. for 5 hours to obtain Compound (2). Non-reacted reactants are filtered, 3.164 g (20 mmol) of 1,8-diaminonaphthalene and 0.5 ml of acetic acid are added thereto, and further reaction is performed for 3 hours to obtain a red powder. The powder is filtered and the resultant is washed with dioxane and acetone several times and purified with a column chromatography to obtain 99.5% of Compound (3). 1.16 g (5 mmol) of 3,4-dihydroxycyclobutane-1,2-dione (Compound (4)) and 2.95 g (10 mmol) of Compound (3) are reacted in 40 ml of a toluene/butanol (a volume ratio of 1:1) solution at 140° C. for 12 hours, and the obtained product is filtered and purified with a column chromatography to obtain final Compound A with a purity of 99.8%. Absorbance of Compound A of Synthesis Example 1 is measured and shown inFIG.20. FIG.20is a graph showing absorbance depending on a wavelength of Compound A obtained in Synthesis Example 1. Referring toFIG.20, Compound A has a maximum absorption wavelength at about 854 nm and high infrared light wavelength selectivity in about 850 nm to about 900 nm. Synthesis Examples 2 to 15 Compounds B to O are synthesized by the same method as Synthesis Example 1 except for using each reactant of Table 1 instead of 2-methylquinoline. TABLE 1SynthesisCom-ExamplepoundReactantFinal compounds1A2B3C4D5E6F7G8H9I10J11K12L13M14N15O Evaluation I Maximum absorption wavelengths (λmax) and extinction coefficients of Compounds A to O of Synthesis Examples 1 to 15 are measured. The maximum absorption wavelengths (λmax) and absorbances are measured by dissolving each compound in dichloromethane to prepare a solution and measuring with a UV-Vis spectrometer. The results are shown in Table 2. TABLE 2Synthesis ExamplesCompoundsλmax(nm)ANIR/AVIS1A85422.222B75716.033C76838.464D75717.865E758526.316F78128.577G91421.748H77811.369I80712.3510J71543.4811K72017.5412L7269.0913M71541.6714N76373.7215O71262.5* ANIR: maximum absorption coefficient in an infrared ray (IR) region* AVIS:maximum absorption coefficient in a visible wavelength spectrum of light Referring to Table 2, compounds of Synthesis Examples 1 to 15 have a maximum absorption wavelength of greater than or equal to 700 nm and ANIR/AVISof greater than or equal to 8 which indicates high infrared light absorption selectivity. Synthesis Example II Synthesis Example 16 3,4-diisopropoxycyclobut-3-2n2-1,2-dione (Compound (1)) is reacted with diphenylamine in a solution including thick hydrochloric acid in propylalcohol and the resultant refluxed for 3 hours to obtain Compound (2). Compound (5) is synthesized in the same process as in Reaction Scheme A except for using 2-methylpyridine (Compound (3)) instead of 2-methylquinoline. Subsequently, 1.33 g (5 mmol) of 3-(diphenylamino)-4-hydroxycyclobut-3-ene-1,2-dione and 1.23 g (5 mmol) of Compound (5) (2-(pyridine-2-yl)-1H-perimidine) are reacted in 40 ml of toluene/butanol (a volume ratio of 1:1) solvent at 140° C. for 12 hours, and the obtained product is filtered and purified with column chromatography to obtain a final compound, Compound Q with a purity of 99.7%. Absorbance of the obtained Compound Q is measured and shown inFIG.21. FIG.21is a graph showing absorbance depending on a wavelength of Compound Q obtained in Synthesis Example 16. Referring toFIG.21, Compound Q has (“is configured to have”) a maximum absorption wavelength at about 800 nm and high infrared light wavelength selectivity in about 750 nm to 850 nm. Synthesis Example 17 Compound (1) (4′-bromo-3,5-dimethoxy-1,1′-bipheny) and diphenylamine are reacted in 50 ml of toluene in the presence of a catalyst Pd2(dba)3and t-BuONa at 120° C. for 8 hours to obtain Compound (2), and obtained Compound (2) is reacted with BBr3under CH2Cl2to obtain Compound (3). Subsequently, 3.53 g (10 mmol) of Compound (3) and 580.35 mg (5 mmol) of Compound (4) are reacted in 40 ml of a toluene/butanol (a volume ratio of 1:1) solvent for 140° C. for 12 hours, and the obtained product is filtered and purified with a column chromatography to obtain a final compound, Compound R with a purity of 99.7%. Absorbance of Compound R is measured and shown inFIG.22. FIG.22is a graph showing absorbance depending on a wavelength of Compound R obtained in Synthesis Example 17. Referring toFIG.22, Compound R has a maximum absorption wavelength at about 800 nm and high infrared light wavelength selectivity in about 750 nm to 850 nm. Comparative Synthesis Example 1 Compound (1) (1-bromo-3,5-dimethoxybenzene) and diphenylamine are reacted in 50 ml of toluene in the presence of a catalyst, Pd2(dba)3and t-BuONa at 120° C. for 8 hours to obtain Compound (2), and obtained Compound (2) is reacted with BBr3under CH2Cl2to obtain Compound (3). Subsequently, 2.77 g (10 mmol) of Compound (3) and 580.35 mg (5 mmol) of Compound (4) are reacted in 40 ml of toluene/butanol (a volume ratio of 1:1) solvent for 140° C. for 12 hours, and the obtained product is filtered and purified with a column chromatography to obtain a final compound, Compound T with a purity of 99.7%. Absorbance of the compound T obtained in Comparative Synthesis Example 1 is measured and shown inFIG.23. FIG.23is a graph showing absorbance depending on a wavelength of Compound T obtained in Comparative Synthesis Example 1. Referring toFIG.23, Compound T has a maximum absorption wavelength at about 650 nm and shows main absorption in a visible ray region. Comparative Synthesis Example 2 Absorbance of Compound U is measured and shown inFIG.24. FIG.24is a graph showing absorbance depending on a wavelength of Compound U obtained in Comparative Synthesis Example 2. Referring toFIG.24, Compound U shows light absorption characteristics in a broad range of about 650 nm to 840 nm. Comparative Synthesis Example 3 Absorbance of Compound V disclosed in US 2008-0230123 A1 is shown inFIG.25. FIG.25is a graph showing absorbance depending on a wavelength of Compound V obtained in Comparative Synthesis Example 3. Referring toFIG.25, Compound V shows light absorption characteristics in broad ranges of about 300 nm to 450 nm and about 550 nm to 800 nm and low ANIR/AVISratio of 0.65. Comparative Synthesis Example 4 Absorbance of Compound W disclosed in US 2008-0230123 A1 is shown inFIG.26. FIG.26is a graph showing absorbance depending on a wavelength of Compound W obtained in Comparative Synthesis Example 4. Referring toFIG.26, Compound W shows light absorption characteristics in broad ranges of about 300 nm to 450 nm and about 650 nm to 800 nm and low ANIR/AVISratio. While this disclosure has been described in connection with what is presently considered to be practical example embodiments, it is to be understood that the inventive concepts are not limited to the disclosed embodiments, but, on the contrary, is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.
95,746
11858912
DETAILED DESCRIPTION OF THE DISCLOSURE Definitions Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein, unless incompatible therewith. All of the features disclosed in this specification (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or process so disclosed, may be combined in any combination, except combinations where at least some of such features and/or steps are mutually exclusive. The invention is not restricted to the details of any foregoing embodiments. The invention extends to any novel one, or any novel combination, of the features disclosed in this specification (including any accompanying claims, abstract and drawings), or to any novel one, or any novel combination, of the steps of any method or process so disclosed. The following definitions are provided to facilitate understanding of certain terms used frequently herein and are not meant to limit the scope of the present disclosure. All references referred to herein are incorporated by reference in their entirety. The term “alkyl” refers to a mono- or multivalent, e.g., a mono- or bivalent, linear or branched saturated hydrocarbon group of 1 to 6 carbon atoms (“C1-C6-alkyl”), e.g., 1, 2, 3, 4, 5, or 6 carbon atoms. In some embodiments, the alkyl group contains 1 to 3 carbon atoms, e.g., 1, 2 or 3 carbon atoms. Some non-limiting examples of alkyl include methyl, ethyl, propyl, 2-propyl (isopropyl), n-butyl, iso-butyl, sec-butyl, tert-butyl, and 2,2-dimethylpropyl. Particularly preferred, yet non-limiting examples of alkyl include methyl and ethyl. The term “alkyldiyl” as used herein refers to a saturated linear or branched-chain divalent hydrocarbon radical of about one to to six carbon atoms (C1-C6). Examples of alkyldiyl groups include, but are not limited to, methylene (—CH2—), ethylene (—CH2CH2—), propylene (—CH2CH2CH2—), and the like. An alkyldiyl group may also be referred to as an “alkylene” group. The term “alkoxy” refers to an alkyl group, as previously defined, attached to the parent molecular moiety via an oxygen atom. Unless otherwise specified, the alkoxy group contains 1 to 6 carbon atoms (“C1-C6-alkoxy”). In some preferred embodiments, the alkoxy group contains contains 1 to 4 carbon atoms. In still other embodiments, the alkoxy group contains 1 to 3 carbon atoms. Some non-limiting examples of alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy and tert-butoxy. A particularly preferred, yet non-limiting example of alkoxy is methoxy. The term “halogen” or “halo” refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I). Preferably, the term “halogen” or “halo” refers to fluoro (F), chloro (Cl) or bromo (Br). Particularly preferred, yet non-limiting examples of “halogen” or “halo” are fluoro (F) and chloro (Cl). The term “cycloalkyl” as used herein refers to a saturated or partly unsaturated monocyclic or bicyclic hydrocarbon group of 3 to 10 ring carbon atoms (“C3-C10-cycloalkyl”). In some preferred embodiments, the cycloalkyl group is a saturated monocyclic hydrocarbon group of 3 to 8 ring carbon atoms. “Bicyclic cycloalkyl” refers to cycloalkyl moieties consisting of two saturated carbocycles having two carbon atoms in common, i.e., the bridge separating the two rings is either a single bond or a chain of one or two ring atoms, and to spirocyclic moieties, i.e., the two rings are connected via one common ring atom. Preferably, the cycloalkyl group is a saturated monocyclic hydrocarbon group of 3 to 6 ring carbon atoms, e.g., of 3, 4, 5 or 6 carbon atoms. Some non-limiting examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and spiro[2.3]hexan-5-yl. The term “aminoalkoxy” refers to an alkoxy group, wherein at least one of the hydrogen atoms of the alkoxy group has been replaced by an amino group. Preferably, “aminoalkoxy” refers to an alkoxy group wherein 1, 2 or 3 hydrogen atoms of the alkoxy group have been replaced by an amino group. Preferred, yet non-limiting examples of aminoalkoxy are aminomethoxy and 1-aminoethoxy. The term “heterocyclyl” refers to a saturated or partly unsaturated mono- or bicyclic, preferably monocyclic ring system of 3 to 14 ring atoms, preferably 3 to 10 ring atoms, more preferably 3 to 8 ring atoms wherein 1, 2, or 3 of said ring atoms are heteroatoms selected from N, O and S, the remaining ring atoms being carbon. Preferably, 1 to 2 of said ring atoms are selected from N and O, the remaining ring atoms being carbon. “Bicyclic heterocyclyl” refers to heterocyclic moieties consisting of two cycles having two ring atoms in common, i.e., the bridge separating the two rings is either a single bond or a chain of one or two ring atoms, and to spirocyclic moieties, i.e., the two rings are connected via one common ring atom. Some non-limiting examples of heterocyclyl groups include azetidin-3-yl; azetidin-2-yl; oxetan-3-yl; oxetan-2-yl; piperidyl; piperazinyl; pyrrolidinyl; 2-oxopyrrolidin-1-yl; 2-oxopyrrolidin-3-yl; 5-oxopyrrolidin-2-yl; 5-oxopyrrolidin-3-yl; 2-oxo-1-piperidyl; 2-oxo-3-piperidyl; 2-oxo-4-piperidyl; 6-oxo-2-piperidyl; 6-oxo-3-piperidyl; 1-piperidinyl; 2-piperidinyl; 3-piperidinyl; 4-piperidinyl; morpholino; morpholin-2-yl; morpholin-3-yl; pyrrolidinyl (e.g., pyrrolidin-3-yl); 3-azabicyclo[3.1.0]hexan-6-yl; 2,5-diazabicyclo[2.2.1]heptan-2-yl; 2-azaspiro[3.3]heptan-2-yl; 2,6-diazaspiro[3.3]heptan-2-yl; 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl, and 3-aza-6-azoniaspiro[5.5]undecane. The term “heterocyclylalkyl” refers to a heterocyclyl group that is bound to the parent moiety via an alkylene group. A preferred, yet non-limiting example of a heterocyclylalkyl group is azetidin-3-ylmethyl. The term “aryl” refers to a monocyclic, bicyclic, or tricyclic carbocyclic ring system having a total of 6 to 10 ring members (“C6-C10-aryl”) and wherein at least one ring in the system is aromatic. A particularly preferred, yet non-limiting example of aryl is phenyl. The term “heteroaryl” refers to a mono- or multivalent, monocyclic or bicyclic, preferably bicyclic ring system having a total of 5 to 14 ring members, preferably, 5 to 12 ring members, and more preferably 5 to 10 ring members, wherein at least one ring in the system is aromatic, and at least one ring in the system contains one or more heteroatoms. Preferably, “heteroaryl” refers to a 5-10 membered heteroaryl comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. Most preferably, “heteroaryl” refers to a 5-10 membered heteroaryl comprising 1 to 2 heteroatoms independently selected from O and N. Some non-limiting examples of heteroaryl include 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridazin-3-yl, pyridazin-4-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyrimidin-6-yl, indol-1-yl, 1H-indol-2-yl, 1H-indol-3-yl, 1H-indol-4-yl, 1H-indol-5-yl, 1H-indol-6-yl, 1H-indol-7-yl, 1,2-benzoxazol-3-yl, 1,2-benzoxazol-4-yl, 1,2-benzoxazol-5-yl, 1,2-benzoxazol-6-yl, 1,2-benzoxazol-7-yl, 1H-indazol-3-yl, 1H-indazol-4-yl, 1H-indazol-5-yl, 1H-indazol-6-yl, 1H-indazol-7-yl, pyrazol-1-yl, 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl, imidazol-1-yl, 1H-imidazol-2-yl, 1H-imidazol-4-yl, 1H-imidazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, thiazol-4-yl, 1,2,4-oxadiazol-3-yl, 1H-triazol-5-yl, 1H-triazol-4-yl, and triazol-1-yl. Most preferably, “heteroaryl” refers to pyridyl and pyrimidinyl. The term “hydroxy” refers to an —OH group. The term “carboxy” refers to a group —C(O)2H, i.e. a carboxylic acid group. The term “oxo” refers to an oxygen atom that is bound to the parent moiety via a double bond (═O). The term “amino” refers to an —NH2group. The term “cyano” refers to a —CN (nitrile) group. The term “carbamoyl” refers to a —C(O)NH2group. The term “carbonyl” refers to a carbon radical having two of the four covalent bonds shared with an oxygen atom (C═O). The term “haloalkyl” refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by a halogen atom, preferably fluoro. Preferably, “haloalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms of the alkyl group have been replaced by a halogen atom, most preferably fluoro. Non-limiting examples of haloalkyl are fluoromethyl, difluoromethyl, trifluoromethyl, trifluoroethyl, 2-fluoroethyl, and 2,2-difluoroethyl. A particularly preferred, yet non-limiting example of haloalkyl is trifluoromethyl. The term “cyanoalkyl” refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by cyano group. Preferably, “cyanoalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms of the alkyl group have been replaced by a cyano group. Most preferably, “cyanoalkyl” refers to an alkyl group wherein 1 hydrogen atom of the alkyl group has been replaced by a cyano group. A preferred, yet non-limiting example of cyanoalkyl is cyanomethyl. The term “alkoxyalkyl” refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by an alkoxy group. Preferably, “alkoxyalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms of the alkyl group have been replaced by an alkoxy group. Most preferably, “alkoxyalkyl” refers to an alkyl group wherein 1 hydrogen atom of the alkyl group has been replaced by an alkoxy group. A preferred, yet non-limiting example of alkoxyalkyl is 2-methoxyethyl. The term “haloalkoxy” refers to an alkoxy group, wherein at least one of the hydrogen atoms of the alkoxy group has been replaced by a halogen atom, preferably fluoro. Preferably, “haloalkoxy” refers to an alkoxy group wherein 1, 2 or 3 hydrogen atoms of the alkoxy group have been replaced by a halogen atom, most preferably fluoro. Particularly preferred, yet non-limiting examples of haloalkoxy are fluoromethoxy (FCH2O—), difluoromethoxy (F2CHO—), and trifluoromethoxy (F3CO—). The term “carbamoylalkyl” refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by a carbamoyl group. Preferably, “carbamoylalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms of the alkyl group have been replaced by a carbamoyl group. Most preferably, “carbamoylalkyl” refers to an alkyl group wherein 1 hydrogen atom of the alkyl group has been replaced by a carbamoyl group. A preferred, yet non-limiting example of a carbamoylalkyl group is 2-amino-2-oxo-ethyl. The term “carboxyalkyl” refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by a carboxy group. Preferably, “carboxyalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms of the alkyl group have been replaced by a carboxy group. Most preferably, “carboxyalkyl” refers to an alkyl group wherein 1 hydrogen atom of the alkyl group has been replaced by a carboxy group. A preferred, yet non-limiting example of a carboxyalkyl group is carboxymethyl. The term “hydroxyalkyl” refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by a hydroxy group. Preferably, “hydroxyalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms, most preferably 1 hydrogen atom of the alkyl group have been replaced by a hydroxy group. Preferred, yet non-limiting examples of hydroxyalkyl are hydroxymethyl, hydroxyethyl (e.g. 2-hydroxyethyl), and 3-hydroxy-3-methyl-butyl. The term “pharmaceutically acceptable salt” refers to those salts which retain the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable. The salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, in particular hydrochloric acid, and organic acids such as formic acid, acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, lactic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcystein and the like. In addition these salts may be prepared by addition of an inorganic base or an organic base to the free acid. Salts derived from an inorganic base include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium salts and the like. Salts derived from organic bases include, but are not limited to salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, lysine, arginine, N-ethylpiperidine, piperidine, polyimine resins and the like. Particular pharmaceutically acceptable salts of compounds of formula (I) are hydrochlorides, fumarates, formates, lactates (in particular derived from L-(+)-lactic acid), tartrates (in particular derived from L-(+)-tartaric acid) and trifluoroacetates. The compounds of formula (I) can contain several asymmetric centers and can be present in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereioisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates. According to the Cahn-Ingold-Prelog Convention, the asymmetric carbon atom can be of the “R” or “S” configuration. The term “treatment” as used herein includes: (1) inhibiting the state, disorder or condition (e.g. arresting, reducing or delaying the development of the disease, or a relapse thereof in case of maintenance treatment, of at least one clinical or subclinical symptom thereof); and/or (2) relieving the condition (i.e., causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms). The benefit to a patient to be treated is either statistically significant or at least perceptible to the patient or to the physician. However, it will be appreciated that when a medicament is administered to a patient to treat a disease, the outcome may not always be effective treatment. The term “mammal” as used herein includes both humans and non-humans and includes but is not limited to humans, non-human primates, canines, felines, murines, bovines, equines, and porcines. In a particularly preferred embodiment, the term “mammal” refers to humans. The term “nosocomial infection” refers to a hospital-acquired infection (HAI), which is an infection that is acquired in a hospital or other health care facility. To emphasize both hospital and nonhospital settings, it is sometimes instead called a health care-associated infection (HAI or HCAI). Such an infection can be acquired in hospitals, nursing homes, rehabilitation facilities, outpatient clinics, or other clinical settings. Compounds of the Invention In a first aspect, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein:X is N or C—R5;R1and R2, taken together with the nitrogen atom to which they are attached, form a group orR1is a group and R2is hydrogen;R3is halogen or C1-C6-alkyl;R4is selected from hydrogen, halogen, C1-C6-alkyl, and C1-C6-alkoxy;R5is selected from hydrogen, halogen, and C1-C6-alkyl;R6is selected from hydrogen, C1-C6-alkyl, carbamoyl-C1-C6-alkyl-NH—, amino, halogen, hydroxy-C1-C6-alkyl, halo-C1-C6-alkyl, and a group R7is selected from hydrogen, C1-C6-alkyl, C1-C6-alkoxy-C1-C6-alkyl-, hydroxy-C1-C6-alkyl, halo-C1-C6-alkyl, carbamoyl-C1-C6-alkyl, C1-C6-alkyl-NH—C(O)—C1-C6-alkyl-, C1-C6-alkyl-NH—C(O)—NH—C1-C6-alkyl-, cyano-C1-C6-alkyl, C1-C6-alkyl-SO2—C1-C6-alkyl-, halo-C1-C6-alkoxy-C1-C6-alkyl-, amino-C1-C6-alkoxy-C1-C6-alkyl-, and a group and R8is selected from hydrogen, halogen, C1-C6-alkyl, halo-C1-C6-alkyl, and C1-C6-alkoxy-C1-C6-alkyl; orR7and R8, taken together with the atoms to which they are attached, form a 3- to 14-membered heterocycle;R9and R10are each independently hydrogen or halogen;RA1is selected from hydrogen, (C1-C6-alkyl)2N—C1-C6-alkyl-C(O)—, (RA6)3N+—C1-C6-alkyl-C(O)—, and a group R RA1is selected from hydrogen, hydroxy, amino, C1-C6-alkyl, halo-C1-C6-alkyl, hydoxy-C1-C6-alkyl, amino-C1-C6-alkyl, carbamoyl-C1-C6-alkyl, C1-C6-alkoxy-C1-C6-alkyl, H2N—SO2—C1-C6-alkyl-, H2N—NH—C(O)—C1-C6-alkyl-, C1-C6-alkoxy, oxo, carbamoyl, and a group RA2is selected from hydrogen, hydroxy, amino, C1-C6-alkyl, carboxy-C1-C6-alkyl, and carbamoyl-C1-C6-alkyl;RA3, RA4, RA5, RC2, and RC3are each independently selected from hydrogen and C1-C6-alkyl;each RA6is independently selected from C1-C6-alkyl, amino-C1-C6-alkyl, C1-C6-alkyl-NH—C1-C6-alkyl-, (C1-C6-alkyl)2N— C1-C6-alkyl-, carboxy-C1-C6-alkyl, and (3- to 14-membered heterocyclyl)-C1-C6-alkyl-;RA7is selected from hydrogen and C1-C6-alkyl;RB1is selected from hydrogen, halogen, cyano, amino, oxo, C1-C6-alkyl, C1-C6-alkoxy, and 3- to 14-membered heterocyclyl;RB2is selected from hydrogen, halogen, and C1-C6-alkyl;RCis selected from hydrogen, (C1-C6-alkyl)2N—C1-C6-alkyl-C(O)—, (C1-C6-alkyl)3N+—C1-C6-alkyl-C(O)—, and a group RC1is hydroxy;REis selected from C1-C6-alkyl, C1-C6-alkoxy, and halogen;LAis selected from —C1-C6-alkyldiyl-, carbonyl, —C(O)—NH—, —NH—C(O)—, —C(O)—N(C1-C6-alkyl)-, —N(C1-C6-alkyl)-C(O)—, —C1-C6-alkyldiyl-NH—C(O)—, —SO2—NH—, —NH—SO2—, —C1-C6-alkyldiyl-C(O)—, and —C(O)—C1-C6-alkyldiyl-C(O)—;LBis selected from a covalent bond, —C1-C6-alkyldiyl-, —NH—C(O)—C1-C6-alkyldiyl-, —C(O)—NH—C1-C6-alkyldiyl-, —NH—C(O)—NH—C1-C6-alkyldiyl-, —C(O)—C1-C6-alkyldiyl-, and —SO2—NH—C1-C6-alkyldiyl-;LCand LEare each independently a covalent bond or —C1-C6-alkyldiyl-;LC1is —NH—C(O)— or carbonyl;A, C, and C1 are each independently a 3- to 14-membered heterocyclyl;A2 is selected from 3- to 14-membered heterocyclyl and C6-C10-aryl;A1 is selected from 3- to 14-membered heterocyclyl, 5- to 14-membered heteroaryl, and C3-C10-cycloalkyl;B is selected from 5- to 14-membered heteroaryl, 3- to 14-membered heterocyclyl, C3-C10-cycloalkyl, and C6-C10-aryl;D is a 5- to 14-membered heteroaryl;E is selected from 5- to 14-membered heteroaryl, 3- to 14-membered heterocyclyl, and C6-C10-aryl; anda wavy line represents the point of attachment of the respective R group to the remainder of formula (I). In one embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein:X is N or C—R5;R1and R2, taken together with the nitrogen atom to which they are attached, form a group orR1is a group and R2is hydrogen;R3is halogen or C1-C6-alkyl;R4is selected from hydrogen, halogen, C1-C6-alkyl, and C1-C6-alkoxy;R5is selected from hydrogen, halogen, and C1-C6-alkyl;R6is selected from hydrogen, C1-C6-alkyl, carbamoyl-C1-C6-alkyl-NH—, amino, halogen, hydroxy-C1-C6-alkyl, halo-C1-C6-alkyl, and a group R7is selected from hydrogen, C1-C6-alkyl, C1-C6-alkoxy-C1-C6-alkyl-, hydroxy-C1-C6-alkyl, halo-C1-C6-alkyl, carbamoyl-C1-C6-alkyl, C1-C6-alkyl-NH—C(O)—C1-C6-alkyl-, C1-C6-alkyl-NH—C(O)—NH—C1-C6-alkyl-, cyano-C1-C6-alkyl, C1-C6-alkyl-SO2—C1-C6-alkyl-, halo-C1-C6-alkoxy-C1-C6-alkyl-, amino-C1-C6-alkoxy-C1-C6-alkyl-, and a group and R8is selected from hydrogen, halogen, C1-C6-alkyl, halo-C1-C6-alkyl, and C1-C6-alkoxy-C1-C6-alkyl; orR7and R8, taken together with the atoms to which they are attached, form a 3- to 14-membered heterocycle;R9and R10are each independently hydrogen or halogen;RA1is selected from hydrogen, (C1-C6-alkyl)2N—C1-C6-alkyl-C(O)—, (RA6)3N+—C1-C6-alkyl-C(O)—, and a group RA1is selected from hydrogen, hydroxy, amino, C1-C6-alkyl, halo-C1-C6-alkyl, hydoxy-C1-C6-alkyl, amino-C1-C6-alkyl, carbamoyl-C1-C6-alkyl, C1-C6-alkoxy-C1-C6-alkyl, H2N—SO2—C1-C6-alkyl-, H2N—NH—C(O)—C1-C6-alkyl-, C1-C6-alkoxy, oxo, carbamoyl, and a group RA2is selected from hydrogen, hydroxy, amino, C1-C6-alkyl, carboxy-C1-C6-alkyl, and carbamoyl-C1-C6-alkyl;RA3, RA4, RA5, RC2, and RC3are each independently selected from hydrogen and C1-C6-alkyl;each RA6is independently selected from C1-C6-alkyl, amino-C1-C6-alkyl, carboxy-C1-C6-alkyl, and (3- to 14-membered heterocyclyl)-C1-C6-alkyl-;RB1is selected from hydrogen, halogen, cyano, amino, oxo, C1-C6-alkyl, and C1-C6-alkoxy;RB2is selected from hydrogen, halogen, and C1-C6-alkyl;RCis selected from hydrogen, (C1-C6-alkyl)2N—C1-C6-alkyl-C(O)—, (C1-C6-alkyl)3N+—C1-C6-alkyl-C(O)—, and a group RC1is hydroxy;REis selected from C1-C6-alkyl, C1-C6-alkoxy, and halogen;LAis selected from —C1-C6-alkyldiyl-, carbonyl, —C(O)—NH—, and —C1-C6-alkyldiyl-C(O)—;LBis selected from a covalent bond, —C1-C6-alkyldiyl-, —NH—C(O)—C1-C6-alkyldiyl-, —C(O)—NH—C1-C6-alkyldiyl-, —NH—C(O)—NH—C1-C6-alkyldiyl-, —C(O)—C1-C6-alkyldiyl-, and —SO2—NH—C1-C6-alkyldiyl-;LCand LEare each independently a covalent bond or —C1-C6-alkyldiyl-;LC1is —NH—C(O)— or carbonyl;A, A2, C, and C1 are each independently a 3- to 14-membered heterocyclyl;A1 is selected from 3- to 14-membered heterocyclyl and C3-C10-cycloalkyl;B is selected from 3- to 14-membered heteroaryl, 3- to 14-membered heterocyclyl, C3-C10-cycloalkyl, and C6-C10-aryl;D is a 3- to 14-membered heteroaryl;E is selected from 3- to 14-membered heteroaryl, 3- to 14-membered heterocyclyl, and C6-C10-aryl; anda wavy line represents the point of attachment of the respective R group to the remainder of formula (I). In one embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein:R1and R2, taken together with the nitrogen atom to which they are attached, form a group RAis selected from (RA6)3N+—C1-C6-alkyl-C(O)—, and a group RA1is selected from hydroxy, C1-C6-alkyl, carbamoyl-C1-C6-alkyl, and a group RA2is selected from C1-C6-alkyl, carboxy-C1-C6-alkyl, and carbamoyl-C1-C6-alkyl;RA3is selected from hydrogen and C1-C6-alkyl;RA4and RA5are hydrogen;each RA6is independently selected from amino-C1-C6-alkyl and carboxy-C1-C6-alkyl;LAis carbonyl; andA, A1 and A2 are each independently a 3- to 14-membered heterocyclyl. In a preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein:R1and R2, taken together with the nitrogen atom to which they are attached, form a group or a group RAis selected from (RA6)3N+—(CH2)3—C(O)—, and a group RA1is selected from hydroxy, methyl, 2-amino-2-oxo-ethyl, and a group RA2is selected from methyl, carboxymethyl, and 2-amino-2-oxo-ethyl;RA3is selected from hydrogen and methyl;RA4and RA5are hydrogen;each RA6is independently selected from aminopropyl and carboxymethyl;LAis carbonyl;A1 is selected from pyrrolidinyl, piperazinyl, piperidinyl, and 3-azabicyclo[3.1.0]hexan-6-yl; andA2 is azetidinyl. In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R1and R2, taken together with the nitrogen atom to which they are attached, form a group or a group wherein RAis as defined herein. In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein LAis selected from —C1-C6-alkyldiyl-, carbonyl, —C(O)—NH—, —NH—C(O)—, —C(O)—N(C1-C6-alkyl)-, —C1-C6-alkyldiyl-NH—C(O)—, —SO2—NH—, —NH—SO2—, —C1-C6-alkyldiyl-C(O)—, and —C(O)—C1-C6-alkyldiyl-C(O)—. In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein LAis selected from —C1-C6-alkyldiyl-, carbonyl, —C(O)—NH—, —SO2—NH—, and —C1-C6-alkyldiyl-C(O)—. In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:R1is a group R2is hydrogen;RCis selected from hydrogen, (C1-C6-alkyl)2N—C1-C6-alkyl-C(O)—, (C1-C6-alkyl)3N+—C1-C6-alkyl-C(O)—, and a group RC1is hydroxy;RC2and RC3are each independently selected from hydrogen and C1-C6-alkyl;LCis a covalent bond or —C1-C6-alkyldiyl-;LC1is —NH—C(O)— or carbonyl; andC and C1 are each independently a 3- to 14-membered heterocyclyl. In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:R1is a group R2is hydrogen;RCis selected from hydrogen, (C1-C6-alkyl)2N—C1-C6-alkyl-C(O)—, (C1-C6-alkyl)3N+—C1-C6-alkyl-C(O)—, and a group RC1is hydroxy;RC2and RC3are each independently selected from hydrogen and C1-C6-alkyl;LC1is —NH—C(O)— or carbonyl; andC1 is a 3- to 14-membered heterocyclyl. In a further preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, whereinR1and R2, taken together with the nitrogen atom to which they are attached, form a group which is wherein RAis as defined herein. In a further preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, whereinR1and R2, taken together with the nitrogen atom to which they are attached, form a group which is wherein RAis as defined herein. In one embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein R3is halogen. In a preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein R3is chloro or fluoro. In a particularly preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein R3is chloro. In one embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein R3is C1-C6-alkyl. In a preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein R3is methyl. In one embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R4and R5are both halogen; andR9is hydrogen. In a preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R4is fluoro or chloro;R5is fluoro; andR9is hydrogen. In one embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R4is selected from halogen and C1-C6-alkyl;R5is halogen; andR9is hydrogen. In a preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R4is selected from methyl, fluoro and chloro;R5is fluoro; andR9is hydrogen. In one embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein: R6is selected from hydrogen, C1-C6-alkyl, and a group;R7is selected from hydrogen, C1-C6-alkoxy-C1-C6-alkyl-, and a group R8is selected from hydrogen, halogen, C1-C6-alkyl, and halo-C1-C6-alkyl;RB1is selected from hydrogen and halogen;RB2is hydrogen;REis halogen;LBis selected from —C1-C6-alkyldiyl- and —NH—C(O)—C1-C6-alkyldiyl-;LEis a covalent bond;B and D are each independently a 3- to 14-membered heteroaryl; andE is C6-C10-aryl. In a preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein:R6is selected from hydrogen, methyl, and a group R7is selected from hydrogen, 2-methoxyethyl, and a group R8is selected from hydrogen, chloro, methyl, and difluoromethyl;RB1is selected from hydrogen and fluoro;RB2is hydrogen;REis fluoro;LBis selected from —CH2— and —NH—C(O)—CH2—;LEis a covalent bond;B is selected from pyridyl and pyridazinyl;D is pyrazolyl; andE is phenyl. In a preferred embodiment, the present invention provides a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, wherein R10is hydrogen. In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the group is a group R In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R1and R2, taken together with the nitrogen atom to which they are attached, form a group R3, R4, R5and REare each independently halogen;R6is selected from hydrogen, C1-C6-alkyl, and a group R7is selected from hydrogen, C1-C6-alkoxy-C1-C6-alkyl-, and a group R8is selected from hydrogen, halogen, C1-C6-alkyl, and halo-C1-C6-alkyl;R9, R10, RA4, RA5, and RB2are hydrogen;RA1is selected from (RA6)3N+—C1-C6-alkyl-C(O)—, and a group RA1is selected from hydroxy, C1-C6-alkyl, carbamoyl-C1-C6-alkyl, and a group RA2is selected from C1-C6-alkyl, carboxy-C1-C6-alkyl, and carbamoyl-C1-C6-alkyl;RA3is selected from hydrogen and C1-C6-alkyl;each RA6is independently selected from amino-C1-C6-alkyl and carboxy-C1-C6-alkyl;RB1is selected from hydrogen and halogen;LAis carbonyl;LBis selected from —C1-C6-alkyldiyl- and —NH—C(O)—C1-C6-alkyldiyl-;LEis a covalent bond;A, A1 and A2 are each independently a 3- to 14-membered heterocyclyl;B and D are each independently a 3- to 14-membered heteroaryl; andE is C6-C10-aryl. In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R1and R2, taken together with the nitrogen atom to which they are attached, form a group or a group; R3is chloro,R4is fluoro or chloro;R5and REare fluoro;R6is selected from hydrogen, methyl, and a group R7is selected from hydrogen, 2-methoxyethyl, and a group R8is selected from hydrogen, chloro, methyl, and difluoromethyl;R9, R10, RA4, RA5, and RB2are hydrogen;RA1is selected from (RA6)3N+—(CH2)3—C(O)—, and a group RA1is selected from hydroxy, methyl, 2-amino-2-oxo-ethyl, and a group RA2is selected from methyl, carboxymethyl, and 2-amino-2-oxo-ethyl;RA3is selected from hydrogen and methyl;each RA6is independently selected from aminopropyl and carboxymethyl;RB1is selected from hydrogen and fluoro;LAis carbonyl;LBis selected from —CH2— and —NH—C(O)—CH2—;LEis a covalent bond;A1 is selected from pyrrolidinyl, piperazinyl, piperidinyl, and 3-azabicyclo[3.1.0]hexan-6-yl;A2 is azetidinyl;B is selected from pyridyl and pyridazinyl;D is pyrazolyl; andE is phenyl. In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R1and R2, taken together with the nitrogen atom to which they are attached, form a group R3, R4, and R5are each independently halogen;R7is C1-C6-alkoxy-C1-C6-alkyl-;R8is C1-C6-alkyl;R6, R9, R10, RA3, RA4, and RA5are hydrogen;RAis a group RA1is a group RA2is carboxy-C1-C6-alkyl;LA1is carbonyl;A, A1 and A2 are each independently a 3- to 14-membered heterocyclyl; andD is a 3- to 14-membered heteroaryl. In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R1and R2, taken together with the nitrogen atom to which they are attached, form a group R3is chloro;R4and R5are both fluoro;R7is 2-methoxyethyl;R8is methyl;R6, R9, R10, RA3, RA4, and RA5are hydrogen;RAis a group RA1is a group RA2is carboxymethyl;LAis carbonyl;A1 is piperidyl;A2 is azetidinyl; andD is a pyrazolyl. In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R1and R2, taken together with the nitrogen atom to which they are attached, form a group R3and R5are each independently halogen;R4is C1-C6-alkyl;R7is C1-C6-alkoxy-C1-C6-alkyl-;R8is C1-C6-alkyl;R6, R9, R10, RA3, RA4, and RA5are hydrogen;RAis a group RA1is a group R RA2is carboxy-C1-C6-alkyl;LAis carbonyl;A, A1 and A2 are each independently a 3- to 14-membered heterocyclyl; andD is a 3- to 14-membered heteroaryl. In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:X is C—R5;R1and R2, taken together with the nitrogen atom to which they are attached, form a group R3is chloro;R4is methyl;R5is fluoro;R7is 2-methoxyethyl;R8is methyl;R6, R9, R10, RA3, RA4, and RA5are hydrogen;RAis a group RA1is a group RA2is carboxymethyl;LAis carbonyl;A1 is piperidyl;A2 is azetidinyl; andD is a pyrazolyl. In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is a compound of formula (II): wherein:X, D, R3, R4, and R6to R10are as defined herein;Y is CH or N, most preferably N; andRYis selected from hydrogen, wherein a wavy line indicates the point of attachment of R to Y. In one embodiment, the present invention provides a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) or (II) is a compound of formula (III): wherein:X, R1to R4, R9and R10are as defined herein; andRZis selected from: wherein a wavy line indicates the point of attachment of RZto the remainder of formula (I). In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is selected from:N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-[(3S)-1,1-dimethylpyrrolidin-1-ium-3-yl]acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(4-hydroxy-1,1-dimethyl-piperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(3-hydroxy-1,1-dimethyl-piperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;[2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-2-oxo-ethyl]-trimethyl-ammonium;[2-[4-[[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]methyl]-1-piperidyl]-2-oxo-ethyl]-trimethyl-ammonium;N-[3-chloro-4-[4-(4,4-dimethylpiperazin-4-ium-1-carbonyl)piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,3S)-3-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S)-4-(hydroxymethyl)-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(4-methoxy-1,1-dimethyl-piperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2-hydroxyethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(azetidin-3-ylmethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[4-[1-(2,2-difluoroethyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2-hydroxyethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2-hydroxyethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2-methoxyethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2-hydroxyethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;2-[4-[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperidine-4-carbonyl]-1-methyl-piperazin-1-ium-1-yl]acetic acid;2-[1-(2-amino-2-oxo-ethyl)-4-[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperidine-4-carbonyl]piperazin-1-ium-1-yl]acetic acid;N-[4-[4-[4-(2-amino-2-oxo-ethyl)-4-methyl-piperazin-4-ium-1-carbonyl]piperidine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-4-hydroxy-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2-hydrazino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;N-[3-chloro-4-[4-[1-[(1,1-dimethylazetidin-1-ium-3-yl)methyl]-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; diformate;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;N-[4-[4-[1-(azetidin-3-ylmethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;2-[1-(azetidin-3-ylmethyl)-4-[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperidine-4-carbonyl]piperazin-1-ium-1-yl]acetic acid;N-[4-[4-[4-(azetidin-3-ylmethyl)-4-methyl-piperazin-4-ium-1-carbonyl]piperidine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[4-(2-amino-2-oxo-ethyl)-4-(azetidin-3-ylmethyl)piperazin-4-ium-1-carbonyl]piperidine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(1R,5S)-3,3-dimethyl-3-azoniabicyclo[3.1.0]hexane-6-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(1R,5S)-3,3-dimethyl-3-azoniabicyclo[3.1.0]hexane-6-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1-(3-hydroxypropyl)-1-methyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; iodide;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[3-chloro-2-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;N-[3-chloro-4-[4-[1-[(1,1-dimethylazetidin-1-ium-3-yl)methyl]piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2,2-difluoroethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2-sulfamoylethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-methyl-1-(2-sulfamoylethyl)piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[1-(2-hydroxyethyl)-1-methyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1-(2-hydroxyethyl)-1-methyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2-chloro-3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperazin-1-ium-1-yl]acetic acid;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[5-chloro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;N-[3-chloro-4-[4-[2-(3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[2-(3-amino-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[2-(3-amino-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[2-[(3S,4S)-3-amino-4-methoxy-1-methyl-pyrrolidin-1-ium-1-yl]acetyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[2-[3-(aminomethyl)-3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl]acetyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[2-(3-carbamoyl-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-[(3R)-3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl]acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-[(3S)-3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl]acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-[(3R,4R)-3,4-dihydroxy-1-methyl-pyrrolidin-1-ium-1-yl]acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-[(3R,4R)-3,4-dihydroxy-1-methyl-pyrrolidin-1-ium-1-yl]acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[2-[(3R,4R)-3-amino-4-methoxy-1-methyl-pyrrolidin-1-ium-1-yl]acetyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(1-methylpyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,3S)-3-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(3-hydroxy-1,1-dimethyl-piperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(3-hydroxy-1,1-dimethyl-piperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[3-(hydroxymethyl)-4,4-dimethyl-piperazin-4-ium-1-carbonyl]piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2R,4S)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2R,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4S)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(3-methoxypropyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(3-methoxypropyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(3S)-4,4-dimethylmorpholin-4-ium-3-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(3-methoxypropyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(4,4-dimethyl-2-oxo-piperazin-4-ium-1-yl)methyl]piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(1-methylpyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(4-hydroxy-1,1-dimethyl-piperidin-1-ium-4-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-[4-hydroxy-4-(hydroxymethyl)-1-methyl-piperidin-1-ium-1-yl]acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-(4-amino-1,1-dimethyl-piperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(4,4-dimethyl-2-oxo-piperazin-4-ium-1-yl)methyl]piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-N-[4-[4-(3-hydroxypiperidine-4-carbonyl)piperazine-1-carbonyl]-3-methyl-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]amino]piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(4-hydroxypiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(3-hydroxypiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S)-5-oxopyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(2-oxopiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(2-pyrrolidin-1-ylacetyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(pyrrolidine-2-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-(3-aminobicyclo[1.1.1]pentane-1-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(dimethylamino)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[[1-[2-(dimethylamino)acetyl]-4-piperidyl]methylcarbamoyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(dimethylamino)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;[2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-2-oxo-ethyl]-trimethyl-ammonium;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(1,1-dimethylpiperidin-1-ium-4-yl)sulfonylamino]piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[[(1R,5S)-3-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]-3-azabicyclo[3.1.0]hexan-6-yl]carbamoyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[[(1S,5R)-3-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]-3-azabicyclo[3.1.0]hexan-6-yl]carbamoyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;bis(3-aminopropyl)-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium;bis(azetidin-3-ylmethyl)-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(4-methoxypiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-(2,2-difluoroethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-[(3S)-pyrrolidin-3-yl]acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-3-methyl-phenyl]-1-methyl-imidazole-2-carboxamide;5-[2-chloro-3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-N-[4-[4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]-3-methyl-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-(4-piperidylmethylcarbamoyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[3-fluoro-4-[3-(trifluoromethyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-(methoxymethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[2-(dimethylamino)acetyl]piperazine-1-carbonyl]-3-methyl-phenyl]-5-[4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(dimethylamino)acetyl]piperazine-1-carbonyl]phenyl]-1-methyl-5-[4-(3-methyl-1H-pyrazol-4-yl)phenyl]imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(dimethylamino)acetyl]piperazine-1-carbonyl]phenyl]-5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-fluoro-6-[5-(4-methoxyphenyl)-1H-pyrazol-4-yl]-3-pyridyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-fluoro-6-(1H-pyrazol-4-yl)-3-pyridyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(1-methylpyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-N-[(3R,4R)-4-hydroxypyrrolidin-3-yl]-3-azabicyclo[3.1.0]hexane-3-carboxamide;N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-5-[4-(1H-pyrazol-4-yl)phenyl]imidazole-2-carboxamide;5-[4-[1-(3-amino-3-oxo-propyl)-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-chloro-3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(3-hydroxy-3-methyl-butyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-(methylamino)-2-oxo-ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(3-hydroxy-2-methyl-propyl)-3-(trifluoromethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[(2S)-2,3-dihydroxypropyl]-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-isopropyl-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[5-(difluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-N-[4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]-3-methyl-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(3-hydroxypropyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(3-hydroxypropyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-methoxy-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[5-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(4-hydroxybutyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-chloro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[3-fluoro-2-methoxy-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[2-(4-hydroxy-1,1-dimethyl-piperidin-1-ium-4-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-(hydroxymethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[3-amino-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-phenyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; chloride;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[5-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methylsulfonylethyl)-3-(trifluoromethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-hydroxypropyl)-3-(trifluoromethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-(4-amino-4-oxo-butyl)-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-(2-amino-2-oxo-ethyl)-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-(trifluoromethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-morpholinoethyl)-5-(trifluoromethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(3-methoxypropyl)-3-(trifluoromethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-(tetrahydropyran-4-ylmethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-(5-amino-2-pyridyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide; chloride;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-(2-pyridylmethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-(1H-pyrazol-4-yl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-[(5-amino-2-pyridyl)methyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide; chloride;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-(2-methylpyrazol-3-yl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-(3-fluorophenyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-(2-oxo-1-pyridyl)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-(1-methylpyrazol-4-yl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-(1H-pyrazol-4-yl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-phenyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-(2-pyridyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;[2-[4-[2-chloro-4-[[1-methyl-5-[4-(3-methyl-1H-pyrazol-4-yl)phenyl]imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-2-oxo-ethyl]-trimethyl-ammonium;[2-[4-[2-chloro-4-[[5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-2-oxo-ethyl]-trimethyl-ammonium;4-chloro-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(6-methoxy-2-pyridyl)ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[(6-methoxy-2-pyridyl)methyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-(1H-triazol-4-ylmethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-3-pyridyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-(3-cyanopropyl)-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-fluoro-6-[5-(4-methoxyphenyl)-1H-pyrazol-4-yl]-3-pyridyl]-1-methyl-imidazole-2-carboxamide;(1R,5S)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-N-[(3R,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-3-yl]-3-azabicyclo[3.1.0]hexane-3-carboxamide;5-[4-[5-[(3-amino-3-oxo-propyl)amino]-2-pyridyl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-(5-amino-3-methyl-2-pyridyl)-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-(5-amino-2-pyridyl)-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-N-[(3R,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-3-yl]-3-azabicyclo[3.1.0]hexane-3-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-fluoro-6-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]-3-pyridyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(isopropylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-[2-(tert-butylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-[2-(1-bicyclo[1.1.1]pentanylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[(3-cyano-1-bicyclo[1.1.1]pentanyl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-(2-anilino-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(2-fluoroanilino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(2-methoxyanilino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(4-fluoroanilino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-(cyclohexylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(thiazol-2-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(1H-pyrazol-4-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(1-methylpyrazol-4-yl)amino]-2-oxo-ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(3-fluoroanilino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; chloride;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(2-pyridylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(1-methylpyridin-1-ium-3-yl)amino]-2-oxo-ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; diformate;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(pyrimidin-2-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(4-pyridylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; chloride;5-[4-[1-[2-(tert-butylamino)-2-oxo-ethyl]-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-methyl-1-[2-oxo-2-(2-pyridylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(tetrahydrofuran-3-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(tetrahydropyran-2-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(tetrahydropyran-3-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(pyridazin-3-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[(1,1-dimethylpiperidin-1-ium-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide; diformate;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-methyl-1-[2-oxo-2-(pyridazin-3-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[(4,4-difluorocyclohexyl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(3-pyridylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-[(5-methoxy-2-pyridyl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(1-piperidyl)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[4-(4,4-dimethylpiperazin-4-ium-1-carbonyl)anilino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-[[(1S,2S)-2-methoxycyclohexyl]amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-oxo-2-(pyridazin-4-ylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[(6-cyano-3-pyridyl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-5-methyl-pyrazol-1-yl]ethyl]pyridine-2-carboxamide;N-[2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazol-1-yl]ethyl]pyridine-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-[(4-fluorophenyl)sulfonylamino]ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-[2-(tert-butylcarbamoylamino)ethyl]-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-[2-(tert-butylcarbamoylamino)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-methyl-1-[2-(2-pyridylcarbamoylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-(2-pyridylcarbamoylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-(pyrrolidine-1-carbonylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(4-methoxyphenyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(3-methoxyphenyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-methoxyphenyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(5-thiazol-4-yl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(5-tetrahydropyran-4-yl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[5-(3,6-dihydro-2H-pyran-4-yl)-1H-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(azetidin-3-ylmethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide;5-[4-[1-[2-(2-aminoethoxy)ethyl]-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-1λ5-azolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-[(2,2-difluorocyclopropyl)methyl]-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-(3-benzyl-3-aza-6-azoniaspiro[5.5]undecane-9-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methyl-3-pyridyl)carbamoylamino]ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(4-oxo-3-aza-6-azoniaspiro[5.5]undecane-9-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[3-fluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;bis(4-aminobutyl)-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]-3-methyl-piperazino]-4-keto-butyl]ammonium;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methylpyridazin-3-yl)amino]-2-oxo-ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-[(6-methoxypyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-morpholinopyridazin-3-yl)amino]-2-oxo-ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;2-[2-(3-aminopropyl)-4-[3-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotoyl]amino]propyl]pyridin-1-ium-1-yl]acetic acid;azetidin-3-ylmethyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formic acid;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;3-aminopropyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;carboxymethyl-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-bis[2-(dimethylamino)ethyl]ammonium;cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetate;bis(3-aminopropyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide;3-aminopropyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-4-keto-butyl]-methyl-ammonium 0.1:1 2,2,2-trifluoroacetic acid;bis(4-aminobutyl)-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium;cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;trans 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;2-[1-(azetidin-3-ylmethyl)-4-[5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-pentanoyl]piperazin-1-ium-1-yl]acetic acid;2-[1-(3-aminopropyl)-4-[5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-pentanoyl]piperazin-1-ium-1-yl]acetic acid;2-[1-(azetidin-3-ylmethyl)-4-[(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-3-azabicyclo[3.1.0]hexane-3-carbonyl]piperidin-1-ium-1-yl]acetic acid;2-[1-(azetidin-3-ylmethyl)-4-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]-4-piperidyl]-methyl-carbamoyl]piperidin-1-ium-1-yl]acetic acid;azetidin-3-ylmethyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium;cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methylpyrazol-4-yl]phenyl]-1-methylimidazole-2-carbonyl]amino]-2-methylbenzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-methyl-benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; andcis 2-[1-(azetidin-3-ylmethyl)-4-[4-[4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methylpyrazol-4-yl]-2-methylphenyl]-1-methylimidazole-2-carbonyl]amino]-2-methylbenzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid. In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is selected from:N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-(4,4-dimethylpiperazin-4-ium-1-carbonyl)piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[4-[4-[4-(2-amino-2-oxo-ethyl)-4-(azetidin-3-ylmethyl)piperazin-4-ium-1-carbonyl]piperidine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(1R,5S)-3,3-dimethyl-3-azoniabicyclo[3.1.0]hexane-6-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid;N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;N-[3-chloro-4-[4-[(2R,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide;bis(3-aminopropyl)-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium;N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2-chloro-3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; andN-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[5-(difluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide. In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide. In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid, in particular cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid formate. In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is N-[3-chloro-4-[4-[(2R,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide. In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide. In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid. In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein said compound of formula (I) is N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide. In one embodiment, the present invention provides pharmaceutically acceptable salts of the compounds of formula (I) as described herein, especially pharmaceutically acceptable salts selected from hydrochlorides, fumarates, lactates (in particular derived from L-(+)-lactic acid), tartrates (in particular derived from L-(+)-tartaric acid) and trifluoroacetates. In yet a further particular embodiment, the present invention provides compounds according to formula (I) as described herein (i.e., as “free bases” or “free acids”, respectively). In some embodiments, the compounds of formula (I) are isotopically-labeled by having one or more atoms therein replaced by an atom having a different atomic mass or mass number. Such isotopically-labeled (i.e., radiolabeled) compounds of formula (I) are considered to be within the scope of this disclosure. Examples of isotopes that can be incorporated into the compounds of formula (I) include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, chlorine, and iodine, such as, but not limited to,2H,3H,11C,13C,14C,13N,15N,15O,17O,18O,31P,32P,35S,18F,36Cl,123I, and125I, respectively. Certain isotopically-labeled compounds of formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e.3H, and carbon-14, i.e.,14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. For example, a compound of formula (I) can be enriched with 1, 2, 5, 10, 25, 50, 75, 90, 95, or 99 percent of a given isotope. Substitution with heavier isotopes such as deuterium, i.e.2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements. Substitution with positron emitting isotopes, such as11C,18F,15O and13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed. Processes of Manufacturing The preparation of compounds of formula (I) of the present invention may be carried out in sequential or convergent synthetic routes. Syntheses of the compounds of the invention are shown in the following schemes. The skills required for carrying out the reactions and purifications of the resulting products are known to those skilled in the art. The substituents and indices used in the following description of the processes have the significance given herein before unless indicated to the contrary. In more detail, the compounds of formula (I) can be manufactured by the methods given below, by the methods given in the examples or by analogous methods. Appropriate reaction conditions for the individual reaction steps are known to a person skilled in the art. Also, for reaction conditions described in literature affecting the described reactions see for example:Comprehensive Organic Transformations: A Guide to Functional Group Preparations,3rd Edition, Richard C. Larock. John Wiley & Sons, New York, NY. 2018). We find it convenient to carry out the reactions in the presence or absence of a solvent. There is no particular restriction on the nature of the solvent to be employed, provided that it has no adverse effect on the reaction or the reagents involved and that it can dissolve the reagents, at least to some extent. The described reactions can take place over a wide range of temperatures, and the precise reaction temperature is not critical to the invention. It is convenient to carry out the described reactions in a temperature range between −78° C. to reflux temperature. The time required for the reaction may also vary widely, depending on many factors, notably the reaction temperature and the nature of the reagents. However, a period of from 0.5 h to several days will usually suffice to yield the described intermediates and compounds. The reaction sequence is not limited to the one displayed in the schemes, however, depending on the starting materials and their respective reactivity the sequence of reaction steps can be freely altered. Starting materials are either commercially available or can be prepared by methods analogous to the methods given below, by methods described in references cited in the description or in the examples, or by methods known in the art. All substituents, in particular, X, D, and R1to R9are as defined above and in the claims, unless otherwise indicated. Furthermore, and unless explicitly otherwise stated, all reactions, reaction conditions, abbreviations and symbols have the meanings well known to a person of ordinary skill in organic chemistry. Intermediate A can be prepared according to Scheme 1. Protection of substituted 4-nitrobenzoic acid A, e.g. with (Boc)2O, gives compound B. Reduction of the nitro group of compound B, for example using the well-known ammonium chloride/iron system at room temperature, yields amine C. Coupling of carboxylic acid D and amine C in the presence of a condensing agent, such as HATU/DIPEA in a solvent, such as DMSO, affords Intermediate A. Intermediates B, C, D can be prepared according to Scheme 2. Hydrolysis the Intermediate A gives carboxylic acid E, which can be coupled with diverse amines in the presence of a condensing agent, such as HATU/DIPEA in a solvent, such as DMSO, to afford Intermediates B, C, and D. Intermediate G can be prepared according to Scheme 3. Thus, Suzuki coupling of Intermediate E with boronic acid esters F, e.g. using palladium catalysts and phosphine ligands, affords Intermediate F. Intermediate F is further reacted with Bis(pinacolato)diboron using palladium catalysts and phosphine ligands to afford Intermediate G (in some cases, the bronic acid ester will directly hydrolyse to the bronic acid in the reaction system). Intermediate H-K, and M-Q can be prepared according to Scheme 4. Thus, Suzuki coupling of Intermediate B, C, D with bronic acid ester Intermediate G in the presence of a palladium catalyst and a phosphine ligand affords Intermediate H-K, and M-Q. In addition to the procedure outlined in Scheme 4, Intermediate H-K, and M-Q also can be prepared according to Scheme 5. Thus, Suzuki coupling of Intermediate A with boronic acid ester Intermediate G in the presence of a palladium catalyst and a phosphine ligand affords compound G. Hydrolysis of compound G yields carboxylic acid Intermediate L, which is subsequently coupled with diverse amines in the presence of a condensing agent, such as HATU/DIPEA in a solvent, such as DMSO, to afford Intermediate H-K, and M-Q. The Examples can be prepared according to Scheme 6. Thus, removal of PG2(in case PG2is not hydrogen) from Intermediates H-K, and M-Q affords compound H. Subsequent removal of PG1(in case PG1is not hydrogen) finally affords Examples. The order of deprotection steps can also be reversed, going via compound I. In some cases, the final Examples are achieved by alkylation of certain intermediates, e.g. methylation using Mel in the presence of DIPEA in acetonitrile at room temperature. The removal of the protective groups PG1and PG2can occur before or after the alkylation step, based on the requirements of the substitution pattern. In one aspect, the present invention provides a process of manufacturing the compounds of formula (I) described herein, wherein said process is as described in any one of Schemes 1 to 6 above. In a further aspect, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, when manufactured according to the processes disclosed herein. Using the Compounds As illustrated in the experimental section, the compounds of formula (I) and their pharmaceutically acceptable salts possess valuable pharmacological properties for the treatment or prevention of infections and resulting diseases, particularly bacteremia, pneumonia, meningitis, urinary tract infection, and wound infection, caused by pathogens, particularly by bacteria, more particularly byAcinetobacterspecies, most particularly byAcinetobacter baumannii. The compounds of formula (I) and their pharmaceutically acceptable salts exhibit activity as antibiotics, particularly as antibiotics againstAcinetobacterspecies, more particularly as antibiotics againstAcinetobacter baumannii, most particularly as pathogen-specific antibiotics againstAcinetobacter baumannii. The compounds of formula (I) and their pharmaceutically acceptable salts can be used as antibiotics, i.e. as antibacterial pharmaceutical ingredients suitable in the treatment and prevention of bacterial infections, particularly in the treatment and prevention of bacterial infections caused byAcinetobacterspecies, more particularly in the treatment and prevention of bacterial infections caused byAcinetobacter baumannii. The compounds of the present invention can be used, either alone or in combination with other drugs, for the treatment or prevention of infections and resulting diseases, particularly bacteremia, pneumonia, meningitis, urinary tract infection, and wound infection, caused by pathogens, particularly by bacteria, more particularly caused byAcinetobacterspecies, most particularly byAcinetobacter baumannii. In one aspect, the present invention provides compounds of formula (I) or their pharmaceutically acceptable salts as described herein for use as therapeutically active substances. In a further aspect, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for use as antibiotic. In a further aspect, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of nosocomial infections and resulting diseases. In a particular embodiment, said nosocomial infections and resulting diseases are selected from bacteremia, pneumonia, meningitis, urinary tract infection and wound infection, or a combination thereof. In a further aspect, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of infections and resulting diseases caused by Gram-negative bacteria. In a particular embodiment, said infections and resulting diseases caused by Gram-negative bacteria are selected from bacteremia, pneumonia, meningitis, urinary tract infection and wound infection, or a combination thereof. In a further aspect, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of infections and resulting diseases caused byEnterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacterspecies orE. coli, or a combination thereof. In a further aspect, the present invention provides a method for the treatment or prevention of infections and resulting diseases caused byEnterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacterspecies orE. coli, or a combination thereof, which method comprises administering a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, to a mammal. In a further aspect, the present invention provides the use of a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, as an antibiotic. In a further aspect, the present invention provides the use of a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for the treatment or prevention of infections and resulting diseases caused byEnterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacterspecies orE. coli, or a combination thereof. In a further aspect, the present invention provides the use of a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for the preparation of medicaments useful for the treatment or prevention of infections and resulting diseases caused byEnterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacterspecies orE. coli, or a combination thereof. In a particular embodiment, said infections and resulting diseases caused byEnterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacterspecies orE. coli, or a combination thereof, are selected from bacteremia, pneumonia, meningitis, urinary tract infection and wound infection, or a combination thereof. In a further aspect, the present invention provides compounds of formula (I) or their pharmaceutically acceptable salts as defined above for use in the treatment or prevention of infections and resulting diseases, particularly bacteremia, pneumonia, meningitis, urinary tract infection, and wound infection, caused by pathogens, particularly by bacteria, more particularly caused byAcinetobacterspecies, most particularly byAcinetobacter baumannii. In a further aspect, the present invention provides a method for the treatment or prevention of infections and resulting diseases, particularly bacteremia, pneumonia, meningitis, urinary tract infection, and wound infection, caused by pathogens, particularly by bacteria, more particularly caused byAcinetobacterspecies, most particularly byAcinetobacter baumannii, which method comprises administering a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined above to a mammal. In a further aspect, the present invention provides the use of compounds of formula (I) or their pharmaceutically acceptable salts as defined above for the treatment or prevention of infections and resulting diseases, particularly bacteremia, pneumonia, meningitis, urinary tract infection, and wound infection, caused by pathogens, particularly by bacteria, more particularly caused byAcinetobacterspecies, most particularly byAcinetobacter baumannii. In a further aspect, the present invention provides the use of compounds of formula (I) or their pharmaceutically acceptable salts as defined above for the preparation of medicaments for the treatment or prevention of infections and resulting diseases, particularly bacteremia, pneumonia, meningitis, urinary tract infection, and wound infection, caused by pathogens, particularly by bacteria, more particularly caused byAcinetobacterspecies, most particularly byAcinetobacter baumannii. Such medicaments comprise compounds of formula (I) or their pharmaceutically acceptable salts as defined above. Pharmaceutical Compositions and Administration In one aspect, the present invention provides pharmaceutical compositions comprising compounds of formula (I) or their pharmaceutically acceptable salts as defined above and one or more pharmaceutically acceptable excipients. Exemplary pharmaceutical compositions are described in Examples 1-4. In a further aspect, the present invention relates to pharmaceutical compositions comprising compounds of formula (I) or their pharmaceutically acceptable salts as defined above and one or more pharmaceutically acceptable excipients for the treatment or prevention of infections and resulting diseases, particularly bacteremia, pneumonia, meningitis, urinary tract infection, and wound infection, caused by pathogens, particularly by bacteria, more particularly caused byAcinetobacterspecies, most particularly byAcinetobacter baumannii. The compounds of formula (I) and their pharmaceutically acceptable salts can be used as medicaments (e.g. in the form of pharmaceutical preparations). The pharmaceutical preparations can be administered internally, such as orally (e.g. in the form of tablets, coated tablets, dragées, hard and soft gelatin capsules, solutions, emulsions or suspensions), nasally (e.g. in the form of nasal sprays) or rectally (e.g. in the form of suppositories). However, the administration can also be effected parentally, such as intramuscularly or intravenously (e.g. in the form of injection solutions or infusion solutions). The compounds of formula (I) and their pharmaceutically acceptable salts can be processed with pharmaceutically inert, inorganic or organic excipients for the production of tablets, coated tablets, dragées and hard gelatin capsules. Lactose, corn starch or derivatives thereof, talc, stearic acid or its salts etc. can be used, for example, as such excipients for tablets, dragées and hard gelatin capsules. Suitable excipients for soft gelatin capsules are, for example, vegetable oils, waxes, fats, semi-solid substances and liquid polyols, etc. Suitable excipients for the production of solutions and syrups are, for example, water, polyols, saccharose, invert sugar, glucose, etc. Suitable excipients for injection solutions are, for example, water, alcohols, polyols, glycerol, vegetable oils, etc. Suitable excipients for suppositories are, for example, natural or hardened oils, waxes, fats, semi-solid or liquid polyols, etc. Moreover, the pharmaceutical preparations can contain preservatives, solubilizers, viscosity-increasing substances, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They can also contain still other therapeutically valuable substances. The dosage can vary in wide limits and will, of course, be fitted to the individual requirements in each particular case. In general, in the case of oral administration a daily dosage of about 0.1 mg to 20 mg per kg body weight, preferably about 0.5 mg to 4 mg per kg body weight (e.g. about 300 mg per person), divided into preferably 1-3 individual doses, which can consist, for example, of the same amounts, should be appropriate. It will, however, be clear that the upper limit given herein can be exceeded when this is shown to be indicated. Co-Administration of Compounds of Formula (I) and Other Agents The compounds of formula (I) or salts thereof or a compound disclosed herein or a pharmaceutically acceptable salt thereof may be employed alone or in combination with other agents for treatment. For example, the second agent of the pharmaceutical combination formulation or dosing regimen may have complementary activities to the compound of formula (I) such that they do not adversely affect each other. The compounds may be administered together in a unitary pharmaceutical composition or separately. In one embodiment a compound or a pharmaceutically acceptable salt can be co-administered with an antibiotic, in particular with an antibiotic for the treatment or prevention of infections and resulting diseases caused byEnterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacterspecies orE. coli, or a combination thereof. The term “co-administering” refers to either simultaneous administration, or any manner of separate sequential administration, of a compound of formula (I) or a salt thereof or a compound disclosed herein or a pharmaceutically acceptable salt thereof and a further active pharmaceutical ingredient or ingredients, including antibiotic agents. If the administration is not simultaneous, the compounds are administered in a close time proximity to each other. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered intravenously and another compound may be administered orally. Typically, any agent that has antimicrobial activity may be co-administered. Particular examples of such agents are Carbapenems (meropenem), Fluoroquinolone (Ciprofloxacin), Aminoglycoside (amikacin), Tetracyclines (tigecycline), Colistin, Sulbactam, Sulbactam+Durlobactam, Cefiderocol (Fetroja), macrocyclic peptides as exemplified e.g. in WO 2017072062 A1, WO 2019185572 A1 and WO 2019206853 A1, and Macrolides (erythromycin). In one aspect, the present invention provides a pharmaceutical composition described herein, further comprising an additional therapeutic agent. In one embodiment, said additional therapeutic agent is an antibiotic agent. In one embodiment, said additional therapeutic agent is an antibiotic agent that is useful for the treatment or prevention of infections and resulting diseases caused byEnterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacterspecies orE. coli, or a combination thereof. In one embodiment, said additional therapeutic agent is an antibiotic agent selected from Carbapenems (meropenem), Fluoroquinolone (Ciprofloxacin), Aminoglycoside (amikacin), Tetracyclines (tigecycline), Colistin, Sulbactam, Sulbactam+Durlobactam, Cefiderocol (Fetroja), macrocyclic peptides as exemplified in WO 2017072062 A1, WO 2019185572 A1 and WO 2019206853 A1, and Macrolides (erythromycin). EXAMPLES The invention will be more fully understood by reference to the following examples. The claims should not, however, be construed as limited to the scope of the examples. In case the preparative examples are obtained as a mixture of enantiomers, the pure enantiomers can be separated by methods described herein or by methods known to the man skilled in the art, such as e.g., chiral chromatography (e.g., chiral SFC) or crystallization. All reaction examples and intermediates were prepared under an argon atmosphere if not specified otherwise. Abbreviations used herein are as follows:ACN or MeCN acetonitrileBINAP 2,2′-Bis(diphenylphosphino)-1,1′-binaphthaleneCFU colony-forming unitd dayDCM dichloromethaneDIPEA N,N-diisopropylethylamineEtOAc or EA ethyl acetateFA formic acidh(s) or hr(s) hour(s)HATU: 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphateHPLC: high performance liquid chromatographyHPLC-UV: high performance liquid chromatography with ultraviolet detectorIC50 half maximal inhibitory concentrationIC90 90% inhibitory concentrationPE petroleum etherPdCl2(DPPF) [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II)Pd2(dba)3Tris(dibenzylideneacetone)dipalladium(0)PG Protecting groupPrecat precatalystprep-HPLC preparative high performance liquid chromatographyRBF Round bottom flaskrt room temperaturesat saturatedSEM 2-methoxyethyl(trimethyl)silaneFA Formic acidTFA Trifluoroacetic Acidwt weightX-PHOS 2-Dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl Intermediate A1 tert-butyl 4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoate Step 1: tert-butyl 2-chloro-4-nitro-benzoate To a mixture of 2-chloro-4-nitro-benzoic acid (15.0 g, 74.42 mmol), N,N-dimethylpyridin-4-amine (2.73 g, 22.33 mmol) and N,N-diethylethanamine (31.12 mL, 223.26 mmol) in THE (80 mL) was added a solution of tert-butoxycarbonyl tert-butyl carbonate (24.36 g, 111.63 mmol) in THE (20 mL) at −10° C. The resulting mixture was warmed to 25° C. and stirred for another 14 h. The mixture was concentrated. The residue was treated with EA (50 mL) and H2O (50 mL). The mixture was extracted with EA. The combined organic layers were concentrated. The crude was then purified by flash column chromatography to afford tert-butyl 2-chloro-4-nitro-benzoate (18.8 g) as a colorless solid. Step 2: tert-butyl 4-amino-2-chloro-benzoate To a mixture of tert-butyl 2-chloro-4-nitro-benzoate (18.8 g, 72.96 mmol) and Ammonium chloride (19.51 g, 364.81 mmol) in ethanol (200 mL) and water (200 mL) was added Iron (20.37 g, 364.81 mmol). The mixture was stirred at 25° C. for 14 h. The mixture was filtered by Celite. The filtrate was concentrated to remove ethanol. The mixture was extracted with EA. The combined organic layers were dried over anhydrous Na2SO4and concentrated to afford tert-butyl 4-amino-2-chloro-benzoate (16.31 g) as a light yellow solid. MS [M+H]+: 228.1. Step 3: tert-butyl 4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoate A mixture of 5-bromo-1-methyl-imidazole-2-carboxylic acid hydrochloride (7.0 g, 28.99 mmol), tert-butyl 4-amino-2-chloro-benzoate (6.0 g, 26.35 mmol), HATU (13.23 g, 34.79 mmol) and DIPEA (16.16 mL, 92.77 mmol) in DMF (15 mL) was stirred at 25° C. for 3 h. The mixture was added water (10 mL) and extracted with EA. The combined organic layers were concentrated. The crude was purified by FCC to afford tert-butyl 4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoate (8 g, 19.29 mmol) as a white solid. MS [M+H]+: 414.0. The following Intermediates were prepared in analogy to Intermediate A1. MSESIStartingEx#NameStructure[M + H]+MaterialIntermediate A2tert-butyl 4-[(5-bromo- 1-methyl-imidazole-2- carbonyl)amino]-2- methyl-benzoate394.22-methyl-4- nitro-benzoic acid and tert- butoxycarbonyl tert-butyl carbonate Intermediate B1 5-bromo-N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-1-methyl-imidazole-2-carboxamide Step 1: 4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoic acid In a 250 mL round-bottomed flask, tert-butyl 4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoate (5 g, 12.1 mmol) was combined with CH2Cl2(30 mL) to give a light brown solution. TFA (41.2 g, 27.9 mL, 362 mmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum to afford 4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoic acid (4.32 g). MS [M+H]+: 359.8. Step 2: tert-butyl 4-(4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoyl)piperazine-2-carboxylate In a 100 mL round-bottomed flask, 4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoic acid (2 g, 5.58 mmol), tert-butyl piperazine-1-carboxylate (1.19 g, 6.41 mmol) and DIPA (2.16 g, 16.7 mmol) were combined with DMF (15 mL) to give a colorless solution. HATU (2.76 g, 7.25 mmol) was added. The reaction was stirred at room temperature for 1 h. The reaction mixture was poured into 150 mL H2N and extracted with EtOAc (3×75 mL). The organic layers were combined, washed with sat. NaCl (1×75 mL). The organic layers were dried over Na2SO4and concentrated in vacuum to afford tert-butyl 4-(4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoyl)piperazine-1-carboxylate (2.94 g). MS [M+H]+: 527.9. The following Intermediates were prepared in analogy to Intermediate B1. MSStartingEx#NameStructureESI [M + H]+MaterialIntermediate B2tert-butyl 4-[4-[(5- bromo-1-methyl- imidazole-2- carbonyl)amino]-2- methyl- benzoyl]piperazine-1- carboxylate506.1Intermediate A2; tert-butyl piperazine-1- carboxylateIntermediate B3tert-butyl N-[1-[4-[(5- bromo-1-methyl- imidazole-2- carbonyl)amino]-2- chloro-benzoyl]-4- piperidyl]carbamate540.1Intermediate A1; tert-butyl N-(4- piperidyl) carbamateIntermediate B4tert-butyl 1-[4-[(5- bromo-1-methyl- imidazole-2- carbonyl)amino]-2- chloro- benzoyl]piperidine-4- carboxylate525.1Intermediate A1; tert-butyl piperidine-4- carboxylateIntermediate B5tert-butyl 4-[[[4-[(5- bromo-1-methyl- imidazole-2- carbonyl)amino]-2- chloro- benzoyl]amino]methyl] piperidine-1- carboxylate554.1Intermediate A1; tert-butyl 4- (aminomethyl) piperidine-1- carboxylateIntermediate B6tert-butyl (1S,5R)-6- [[4-[(5-bromo-1- methyl-imidazole-2- carbonyl)amino]-2- chloro- benzoyl]amino]-3- azabicyclo[3.1.0] hexane-3-carboxylate538.8Intermediate A1; tert-butyl rac-(1R,5S)-6- amino-3- azabicyclo[3.1.0] hexane-3- carboxylate Intermediate C1 5-bromo-N-(3-chloro-4-(piperazine-1-carbonyl)phenyl)-1-methyl-1h-imidazole-2-carboxamide In a 100 mL round-bottomed flask, tert-butyl 4-(4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoyl)piperazine-1-carboxylate (2.94 g, 5.58 mmol) was combined with THE (20 mL) to give a light brown solution. HCl (in water) (11.6 mL, 140 mmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford 5-bromo-N-(3-chloro-4-(piperazine-1-carbonyl)phenyl)-1-methyl-1h-imidazole-2-carboxamide (2.38 g). MS [M+H]+: 427.8. The following Intermediates were prepared in analogy to Intermediate C1 MSStartingEx#NameStructureESI [M + H]+MaterialIntermediate C25-bromo-1-methyl-N- [3-methyl-4- (piperazine-1- carbonyl)phenyl] imidazole-2- carboxamide406.1Intermediate B2 and HClInterme diate C31-[4-[(5-bromo-1- methyl-imidazole-2- carbonyl)amino]-2- chloro- benzoyl]piperidine-4- carboxylic acid469.1Intermediate B4 and HClIntermediate C4N-[4-[[(1S,5R)-3- azabicyclo[3.1.0]hexan- 6-yl]carbamoyl]-3- chloro-phenyl]-5- bromo-1-methyl- imidazole-2- carboxamide438.7Intermediate B6 and HCl Intermediate D1 tert-butyl 4-(4-(4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoyl)piperazine-1-carbonyl)piperidine-1-carboxylate In a 100 mL round-bottomed flask, 5-bromo-N-(3-chloro-4-(piperazine-1-carbonyl)phenyl)-1-methyl-1h-imidazole-2-carboxamide (2.38 g, 5.58 mmol), 1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (2.05 g, 8.92 mmol) and DIPEA (2.16 g, 16.7 mmol) were combined with DMF (15 mL) to give a light brown solution. HATU (3.39 g, 8.92 mmol) was added. The reaction was stirred at room temperature for 1 h. The reaction mixture was poured into 150 mL H2O and extracted with EtOAc (3×50 mL). The organic layers were combined, washed with sat NaCl (1×75 mL). The organic layers were dried over Na2SO4and concentrated in vacuum to afford tert-butyl 4-(4-(4-(5-bromo-1-methyl-1h-imidazole-2-carboxamido)-2-chlorobenzoyl)piperazine-1-carbonyl)piperidine-1-carboxylate (3.56 g). MS [M+H]+: 638.9. The following intermediates were prepared in analogy to Intermediate D1. MSESIStartingEx#NameStructure[M + H]+MaterialIntermediate D2tert-butyl (3S)-3-[2-[4- [4-[(5-bromo-1-methyl - imidazole-2- carbonyl)amino]-2- chloro- benzoyl]piperazin-1-yl]- 2-oxo-ethyl]pyrrolidine- 1-carboxylate637.9Intermediate C1 and 2- [(3 S)-1-tert- butoxycarbonyl- pyrrolidin- 3-yl] acetic acidIntermediate D35-bromo-N-[3-chloro-4- [4-[2- (dimethylamino)acetyl] piperazine-1- carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide511.2Intermediate C1 and 2- (dimethylamino) acetic acidIntermediate D4tert-butyl (2S,4R)-2-[4- [4-[(5-bromo-1-methyl- imidazole-2- carbonyl)amino]-2- methyl- benzoyl]piperazine-1- carbonyl]-4-hydroxy- pyrrolidine-1-carboxylate619.1Intermediate C2 and (2S,4R)-1- tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acidIntermediate D55-bromo-N-[4-[4-[2- (dimethylamino)acetyl] piperazine-1-carbonyl]-3- methyl-phenyl]-1- methyl-imidazole-2- carboxamide491.2Intermediate C2 and 2- (dimethylamino) acetic acidIntermediate D6tert-butyl (2S,4R)-2-[4- [4-[(5-bromo-1-methyl- imidazole-2- carbonyl)amino]-2- chloro- benzoyl]piperazine-1- carbonyl] -4-hydroxy- pyrrolidine-1-carboxylate639.2Intermediate C1 and (2S,4R)-1- tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acidIntermediate D7tert-butyl 4-[1-[4-[(5- bromo-1-methyl- imidazole-2- carbonyl)amino]-2- chloro- benzoyl]piperidine-4- carbonyl]piperazine-l- carboxylate637.0Intermediate C3 and 1-Boc- piperazineIntermediate D8tert-butyl 4-[2-[4-[4-[(5- bromo-1-methyl- imidazole-2- carbonyl)amino]-2- chloro- benzoyl]piperazin-1-yl]- 2-oxo-ethyl]-4-hydroxy- piperidine-1-carboxylate667.2Intermediate C1 and2-(1- tert- butoxycarbonyl- 4-hydroxy-4- piperidyl) acetic acidIntermediate D95-bromo-N-[3-chloro-4- [4-(1-methylpiperidine- 4-carbonyl)piperazine-1- carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide551.2Intermediate C1 and 1- methylpiperidine- 4-carboxylic acid Intermediate D10 tert-butyl 4-[4-[4-[(5-bromo-4-chloro-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate Step 1: 5-bromo-4-chloro-1-methyl-imidazole 4-chloro-1-methyl-imidazole (466 mg, 4. mmol) was dissolved in N,N-dimethylformamide (8 mL), NBS (498.13 mg, 2.8 mmol) was added at rt. The mixture was stirred at room temperature for 1 h. The reaction mixture was diluted with sat. NaHCO3-solution (20 mL) and extracted two times with EtOAc (30 mL). The organic layers were washed with brine (30 mL), dried over Na2SO4and concentrated to dryness. The crude product was directly used to the next step, to afford 5-bromo-4-chloro-1-methyl-imidazole (284 mg, 36.34%) as light brown solid. MS [M+H]+: 196.9. Step 2: 5-bromo-4-chloro-1-methyl-imidazole-2-carboxylic acid methyl ester 5-bromo-4-chloro-1-methyl-imidazole (284 mg, 1.45 mmol) was dissolved in tetrahydrofuran (5 mL), 2 M lithium diisopropylamide (871.88 uL, 1.74 mmol) was added at −78° C., The reaction was stirred at −78° C. for 30 mins, methyl chloroformate (164.79 mg, 1.74 mmol) was added at −78° C. The mixture was warmed to room temperature with stirring for 1 h. The reaction mixture was diluted with water (20 mL) and extracted two times with EtOAc (20 mL). The organic layers were washed with brine (20 mL), dried over Na2SO4and concentrated to dryness. The crude product was directly used to the next step, to afford 5-bromo-4-chloro-1-methyl-imidazole-2-carboxylic acid methyl ester (360 mg) as light brown oil. MS [M+H]+: 254.9. Step 3: 5-bromo-4-chloro-1-methyl-imidazole-2-carboxylic acid 5-bromo-4-chloro-1-methyl-imidazole-2-carboxylic acid methyl ester (360 mg, 1.42 mmol) was dissolved in methanol (9 mL) and water (3 mL), NaOH (284.05 mg, 7.1 mmol) was added at rt. The mixture was stirred at room temperature for 1 h. The PH of the reaction mixture was adjusted to 6. The reaction was concentrated to dryness. The crude product was directly used to the next step, to afford 5-bromo-4-chloro-1-methyl-imidazole-2-carboxylic acid (340 mg) as light yellow solid. MS [M+H]+: 240.9. Step 4: 4-(1-tert-butoxycarbonylisonipecotoyl)piperazine-1-carboxylic acid benzyl ester 1-tert-butoxycarbonylisonipecotic acid (2.5 g, 10.9 mmol) was dissolved in N,N-dimethylformamide (31.25 mL), benzyl 1-piperazinecarboxylate (2.64 g, 11.99 mmol), HATU (4.98 g, 13.09 mmol) and DIEA (2.82 g, 21.81 mmol) were added at rt. The mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with water (30 mL) and extracted two times with EtOAc (30 mL). The organic layers were washed with brine (30 mL), dried over Na2SO4and concentrated to dryness. The crude material was purified by flash chromatography on silica gel (5% MeOH in DCM). to afford 4-(1-tert-butoxycarbonylisonipecotoyl)piperazine-1-carboxylic acid benzyl ester (3 g, 63.76%) as colorless oil. MS [M+H]+: 454.2. Step 5: 4-(piperazine-1-carbonyl)piperidine-1-carboxylic acid tert-butyl ester 4-(1-tert-butoxycarbonylisonipecotoyl)piperazine-1-carboxylic acid benzyl ester (3 g, 6.95 mmol) was dissolved in methanol (50 mL), palladium hydroxide on carbon (97.63 mg, 0.695 mmol) was added at rt. The mixture was purge from the H2ballon three times, and stirred at room temperature for 15 h. The reaction mixture was filtered, the filtrate was concentrated to dryness. The crude product was directly used to the next step to afford 4-(piperazine-1-carbonyl)piperidine-1-carboxylic acid tert-butyl ester (2.07 g) as colorless oil. MS [M+H]+: 298.2. Step 6: 4-[4-(4-amino-2-chloro-benzoyl)piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester 4-(piperazine-1-carbonyl)piperidine-1-carboxylic acid tert-butyl ester (1 g, 3.36 mmol) was dissolved in N,N-dimethylformamide (7 mL), 4-amino-2-chloro-benzoic acid (576.95 mg, 3.36 mmol), HATU (1.53 g, 4.04 mmol) and DIEA (869.16 mg, 1.17 mL, 6.73 mmol) were added at rt. The mixture was stirred at room temperature for 1 h. The reaction mixture was diluted with water (40 mL) and extracted two times with EtOAc (30 mL). The organic layers were washed with brine (30 mL), dried over Na2SO4and concentrated to dryness. The crude material was purified by flash chromatography on silica gel (10% MeOH in DCM). to afford 4-[4-(4-amino-2-chloro-benzoyl)piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester (1.09 g) as white solid. MS [M+H]+: 351.2. Step 7: 4-[4-[4-[(5-bromo-4-chloro-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester 4-[4-(4-amino-2-chloro-benzoyl)piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester (400 mg, 0.887 mmol) was dissolved in N,N-dimethylformamide (5 mL), 5-bromo-4-chloro-1-methyl-imidazole-2-carboxylic acid (212.39 mg, 0.887 mmol), HATU (472.17 mg, 1.24 mmol) and DIEA (229.27 mg, 1.77 mmol) were added at rt. The mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with water (30 mL) and extracted two times with EtOAc (30 mL). The organic layers were washed with brine (30 mL), dried over Na2SO4and concentrated to dryness. The crude material was purified by flash chromatography on silica gel (10% MeOH in DCM). to afford 4-[4-[4-[(5-bromo-4-chloro-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester (277 mg) as light brown solid. MS [M+H]+: 573.1. Intermediate D11 tert-butyl (3R,4R)-3-[[(1S,5R)-6-[[4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]amino]-3-azabicyclo[3.1.0]hexane-3-carbonyl]amino]-4-hydroxy-pyrrolidine-1-carboxylate In a 25 mL round bottomed-flask equipped with a magnetic stirrer-bar, a N2-balloon and a spetum-cap, tert-butyl (3R,4R)-3-amino-4-hydroxypyrrolidine-1-carboxylate (253 mg, 1.25 mmol) was dissolved in DMF (1 mL). TEA (211 mg, 2.08 mmol) and CDI (169 mg, 1.04 mmol) were added to the clear solution and stirred at RT. After 15 min, N-(4-(((1R,5S,6s)-3-azabicyclo[3.1.0]hexan-6-yl)carbamoyl)-3-chlorophenyl)-5-bromo-1-methyl-1H-imidazole-2-carboxamide hydrochloride (197.8 mg, 416 μmol) was added and the resulting light brownish reaction solution was stirred for 1.5 h. Water (7 mL) was added to the reaction mixture, but the product did not precipitate out. The aqueous layer was extracted with EA (2×10 mL). The organic layers were washed with LiCl-solution (5% in water) (10 mL each organic layer) and with sat.-NaCl solution (1×10 mL), dried over Na2SO4, filtered off and concentrated in vacuum at 40° C. Crude product purified by silica gel chromatography, to yield tert-butyl (3R,4R)-3-((1R,5S)-6-(4-(5-bromo-1-methyl-1H-imidazole-2-carboxamido)-2-chlorobenzamido)-3-azabicyclo[3.1.0]hexane-3-carboxamido)-4-hydroxypyrrolidine-1-carboxylate (216 mg). [M+H]+: 668.3. The following intermediates were prepared in analogy to Intermediate D11. MSESIStartingEx#NameStructure[M + H]+MaterialIntermediate D11tert-butyl 4-[4-[4-[(5- bromo-1-methyl - imidazole-2- carbonyl)amino]-2- chloro- benzoyl]piperazine-1- carbonyl]piperazine-1- carboxylate638.9Intermediate C1 and CDI; tert-butyl piperazine-1- carboxylate Intermediate E1 and Intermediate E2 1-(2-methoxyethyl)-3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (Intermediate E1) 1-(2-methoxyethyl)-5-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (Intermediate E2) To a 25 mL microwave vial was added 3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (2 g, 9.61 mmol), 1-bromo-2-methoxyethane (1.74 g, 12.5 mmol), K2C3(1.73 g, 12.5 mmol) and potassium iodide (319 mg, 1.92 mmol) in DMF (15 mL). The vial was capped and heated in the microwave at 100° C. for 15 h. The reaction mixture was filtered through glass fiber paper. The filtrate was concentrated in vacuum. The crude material was purified by flash chromatography to afford 2 g of crude product, The crude product was purified by preparative HPLC to afford 1-(2-methoxyethyl)-3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (550 mg)(Intermediate E1) and 1-(2-methoxyethyl)-5-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (316 mg))(Intermediate E2). MS [M+H]+: 267.1. The following intermediates were prepared in analogy to Intermediate E1. MSESIStartingEx#NameStructure[M + H]+MaterialIntermediate E31-(2-methoxyethyl)-3,5- dimethyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazole281.1Intermediate SM1 and 1-bromo-2- methoxyethaneIntermediate E4trimethyl-[2-[[3-methyl- 4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazol-1- yl]methoxy]ethyl]silane339.0Intermediate SM1 and 2- (chloro methoxy)ethyl- trimethyl- silaneIntermediate E52-[[3,5-dimethyl-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazol-1- yl]methoxy]ethyl- trimethyl-silane353.2Intermediate SM1 and 2-(chloro methoxy)ethyl- trimethyl- silaneIntermediate E62-methyl-4-[5-methyl-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazol-1-yl]butan-2- ol295.2Intermediate SM1 and 4- bromo-2- methyl-butan- 2-olIntermediate E7N-methyl-2-[3-methyl-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazol-1- yl]acetamide280.2Intermediate SM1 and 2- iodo-N- methyl- acetamideIntermediate E8methyl 2-[3-methyl-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazol-1-yl]acetate281.4Intermediate SM1 and methyl 2- bromoacetateIntermediate E9tert-butyl-dimethyl-[2- [3-methyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazol-1- yl]ethoxy]silane367.3Intermediate SM1 and (2- bromo ethoxy)(tert- butyl) dimethylsilaneIntermediate E10tert-butyl-dimethyl-[2- [5-methyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazol-1- yl]ethoxy]silane367.3Intermediate SM1 and (2- bromo ethoxy)(tert- butyl) dimethylsilaneIntermediate E111-(3-methoxypropyl)-3- methyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazole281.1Intermediate SM1 and 1-bromo-3- methoxypropaneIntermediate E121-(3-methoxypropyl)-5- methyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazole281.1Intermediate SM1 and 1-bromo-3- methoxypropaneIntermediate E13tert-butyl-dimethyl-[3- [3-methyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazol-1- yl]propoxy]silane381.3Intermediate SM1 and(3- bromo propoxy)(tert- butyl) dimethylsilaneIntermediate E14tert-butyl-dimethyl-[3- [5-methyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazol-1- yl]propoxy]silane381.4Intermediate SM1 and(3- bromo propoxy)(tert- butyl) dimethylsilaneIntermediate E15methyl 2-[5-methyl-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazol-1-yl]acetate281.4Intermediate SM1 and methyl 2- bromoacetateIntermediate E163-methyl-1- (tetrahydropyran-4- ylmethyl)-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazole306.9Intermediate SM1 and bromomethyl cyclohexaneIntermediate E172-[[3-methyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazol-1- yl]methyl]pyridine299.2Intermediate SM1 and bromomethyl benzeneIntermediate E212-methoxy-6-[[3-methyl- 4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazol-1- yl]methyl]pyridine329.2Intermediate SM1 and 2- (bromomethyl)- 6-methoxy- pyridineIntermediate E224-[3,5-dimethyl-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazol-1- yl]butanenitrile289.23,5-dimethyl- 4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)-1H- pyrazole and 4-bromobutane nitrileIntermediate E231-(2-methoxyethyl)-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazole253.14-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)-1H- pyrazole and 1-bromo- 2-methoxy- ethaneIntermediate E24tert-butyl-[2-[3,5- dimethyl-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazol-1-yl]ethoxy]- dimethyl-silane381.33,5-dimethyl- 4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)-1H- pyrazole and (2-bromo ethoxy)(tert- butyl) dimethylsilaneIntermediate E251-[(2,2- difluorocyclopropyl) methyl]-3,5-dimethyl-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)pyrazole313.23,5-dimethyl- 4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)-1H- pyrazole and 2- (bromomethyl)- 1,1-difluoro- cyclopropane Intermediate E18 5-ethyl-1-(2-methoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole A solution of 4-bromo-5-ethyl-1-(2-methoxyethyl)-1H-pyrazole (0.47 g, 2.0 mmol) and 2-isopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (0.56 g, 3.0 mmol) were dissolved in THE (8.0 ml). The solution was cooled to −78° C. under argon atmosphere. N-butyllithium (2.0 mL, 3.0 mol) was then added dropwise to the solution. The resulting solution was stirred for 60 min at this temperature, and then the temperature was raise to room temperature gradually. The reaction mixture was quenched by methanol at 0° C. and evaporation of the solvent gave a crude product, which was purified by flash chromatography on silica gel to give 5-ethyl-1-(2-methoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (706 mg). MS [M+H]+: 281.1. Intermediate E19 tert-butyl-dimethyl-[4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(2-trimethylsilylethoxymethyl)pyrazol-3-yl]butoxy]silane Step 1: 2-[(4-bromopyrazol-1-yl)methoxy]ethyl-trimethyl-silane 4-bromo-1H-pyrazole (7.35 g, 50 mmol) and DIPEA (13.0 g, 100 mmol) were dissolved in anhydrous CH2Cl2(50 ml). The solution was cooled to 0° C. and (2-(chloromethoxy)ethyl)trimethylsilane (10 g, 60 mmol) was then added dropwise. The mixture was warmed to room temperature and then stirred for 12.0 h. The mixture was poured into water and the aqueous solution was extracted with EtOAc (2×150 ml). The organic layers were combined and washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a red oil which was purified by flash chromatography on silica gel to afford 2-[(4-bromopyrazol-1-yl)methoxy]ethyl-trimethyl-silane (8.6 g). MS [M+H]+: 277.1. Step 2: 2-[[4-bromo-5-[4-[tert-butyl(dimethyl)silyl]oxybutyl]pyrazol-1-yl]methoxy]ethyl-trimethyl-silane To a solution of 2-[(4-bromopyrazol-1-yl)methoxy]ethyl-trimethyl-silane (1.4 g, 5.0 mmol) in anhydrous THF (15 mL) was added dropwise LDA (2.0 M in THF) (5.0 mmol) at −78° C. under argon. The resulting mixture was stirred for 1.0 h at −78° C., tert-butyl-(4-iodobutoxy)-dimethyl-silane (2.4 g, 7.5 mmol) was added. The reaction was stirred at −78° C. for 30 min. the reaction was warmed to room temperature with stirring. The reaction was quenched by saturated aqueous solution of ammonium chloride and followed by extracted with EtOAc (2×50 mL). The organic layers were combined, washed with water and brine, dried over anhydrous sodium sulfate and concentrated in vacuum. The crude material was purified by flash chromatography on silica gel to give 2-[[4-bromo-5-[4-[tert-butyl(dimethyl)silyl]oxybutyl]pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (1.2 g). MS [M+H]+: 463.3. Step 3: tert-butyl-dimethyl-[4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(2-trimethylsilylethoxymethyl)pyrazol-3-yl]butoxy]silane To a solution of 2-[[4-bromo-5-[4-[tert-butyl(dimethyl)silyl]oxybutyl]pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (0.46 g, 1.0 mmol) and 2-isopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (0.28 g, 1.5 mmol) in anhydrous THE (15 mL) was added dropwise n-BuLi (1.6 M in THF) (1.3 mL, 2.0 mmol) at −78° C. under argon. The resulting mixture was stirred for 1.0 h at −78° C. and then the reaction was warmed to room temperature, stirred overnight. The reaction was quenched by saturated aqueous solution of ammonium chloride and followed by extracted by EtOAc (2×50 mL). The organic layers were combined, washed with water and brine, dried over anhydrous sodium sulfate and concentrated in vacuum. The crude material was purified by flash chromatography on silica gel to give tert-butyl-dimethyl-[4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(2-trimethylsilylethoxymethyl)pyrazol-3-yl]butoxy]silane the title compound. (400.0 mg). MS [M+H]+: 511.0. Intermediate E20 1-(2-methoxyethyl)-5-(methoxymethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole Step 1: 4-bromo-1-(2-methoxyethyl)pyrazole To a solution of 4-bromo-1H-pyrazole (5.88 g, 40 mmol) in anhydrous DMF (25 ml) was added NaH (2.4 g, 60 mmol) and then the mixture stirred at 0° C. for 1 h. 1-bromo-2-methoxyethane (8.34 g, 60 mmol) was added in batches to the mixture and then stirred for extra 2.0 h at room temperature. The reaction mixture was quenched by water at 0° C. and then acidified with 1N HCl to PH=7-8, the aqueous solution was extracted with EtOAc, the combined extracts were concentrated in vacuum. The crude material was purified by flash chromatography on silica gel to afford 4-bromo-1-(2-methoxyethyl)pyrazole (7.2 g). Step 2: 4-bromo-2-(2-methoxyethyl)pyrazole-3-carbaldehyde To a solution of 4-bromo-1-(2-methoxyethyl)pyrazole (3.1 g, 15 mmol) in anhydrous THE (20 mL) was added dropwise LDA (22.5 mmol) at −78° C. under argon. The resulting mixture was stirred for 1.0 h at −78° C. and then DMF (1.65 g, 22.5 mmol) was added dropwise into the mixture and stirred for extra 8.0 h at room temperature. The reaction mixture was quenched by water at 0° C. and then acidified with 1N HCl to PH=7-8. The aqueous solution was extracted with EtOAc, the combined extracts were concentrated in vacuum. The crude material was purified by flash chromatography on silica gel to afford afford 4-bromo-2-(2-methoxyethyl)pyrazole-3-carbaldehyde (3.0 g). MS [M+H]+: 232.9. Step 3: [4-bromo-2-(2-methoxyethyl)pyrazol-3-yl]methanol To a solution of 4-bromo-2-(2-methoxyethyl)pyrazole-3-carbaldehyde (3.5 g, 15 mmol) in anhydrous THF (65 mL) was added dropwise borane (1.0 M in THF) (22.5 mmol) at −78° C. under argon. The resulting mixture was stirred for 2.0 h at −78° C. and then stirred for extra 5.0 h at room temperature. The reaction mixture was quenched by water at 0° C. and then extracted with EtOAc (75 mL×3), the combined extracts were concentrated in vacuum. The crude material was purified by flash chromatography on silica gel to afford [4-bromo-2-(2-methoxyethyl)pyrazol-3-yl]methanol (2.8 g). MS [M+H]+: 235.0. Step 4: 4-bromo-1-(2-methoxyethyl)-5-(methoxymethyl)pyrazole To a solution of [4-bromo-2-(2-methoxyethyl)pyrazol-3-yl]methanol (1.2 g, 5.0 mmol) in anhydrous THF (25 mL) was added NaH (300 mg, 7.5 mmol) at 0° C. and then the suspension was stirred for 1 h. Iodomethane (1.1 g, 7.5 mmol) was added into the mixture, stirred for extra 2.0 h at room temperature. The mixture was quenched by water and then poured into water (50 mL) and the aqueous solution was extracted with EtOAC (100 mL×2). The organic layers were combined and washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a red oil, the residue was purified by flash chromatography on silica gel to afford 4-bromo-1-(2-methoxyethyl)-5-(methoxymethyl)pyrazole (0.86 g). MS [M+H]+: 249.0. Step 5: 1-(2-methoxyethyl)-5-(methoxymethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole A solution of 4-bromo-1-(2-methoxyethyl)-5-(methoxymethyl)pyrazole (2.5 g, 10 mmol) and 2-isopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (2.3 g, 12 mmol) were dissolved in THE (20 mL). The solution was cooled to −78° C. under argon atmosphere, n-butyllithium (7.5 mL, 12 mmol) was then added dropwise to the solution. The resulting solution was stirred for 1.0 h at this temperature, and then the temperature was raised to room temperature. The reaction mixture was quenched by methanol at 0° C. Evaporation of the solvent gave a crude product, which was purified by flash chromatography on silica gel to give 1-(2-methoxyethyl)-5-(methoxymethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (1.0 g). MS [M+H]+: 297.1. Intermediate F1 and Intermediate F2 4-(4-bromo-2,3-difluoro-phenyl)-1-(2,2-difluoroethyl)-3-methyl-pyrazole (Intermediate F1) 4-(4-bromo-2,3-difluoro-phenyl)-1-(2,2-difluoroethyl)-3-methyl-pyrazole (Intermediate F2) Step 1: 4-(4-bromo-2,3-difluoro-phenyl)-3-methyl-1H-pyrazole To a 25 mL microwave vial was added 3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (1.7 g, 8.15 mmol), 1-bromo-2,3-difluoro-4-iodobenzene (2 g, 6.27 mmol), Na2CO3(1.99 g, 18.8 mmol) and PdCl2(dppf)-CH2Cl2adduct (459 mg, 627 μmol) in Dioxane (50 mL)/Water (5 mL). The vial was capped and heated in the microwave at 100° C. for 15 h under N2. The crude reaction mixture was concentrated in vacuum. The crude material was purified by flash chromatography to afford 4-(4-bromo-2,3-difluorophenyl)-3-methyl-1H-pyrazole (1.7 g). MS [M+H]+: 275.0. Step 2: 4-(4-bromo-2,3-difluoro-phenyl)-1-(2,2-difluoroethyl)-3-methyl-pyrazole and 4-(4-bromo-2,3-difluoro-phenyl)-1-(2,2-difluoroethyl)-3-methyl-pyrazole In a 50 mL round-bottomed flask, 4-(4-bromo-2,3-difluorophenyl)-3-methyl-1H-pyrazole (1 g, 3.66 mmol), K2CO3(759 mg, 5.49 mmol) and 1,1-difluoro-2-iodoethane (914 mg, 4.76 mmol) were combined with DMF (10 mL) to give a light yellow solution. The reaction mixture was heated to 100° C. and stirred for 15 h. The reaction mixture was filtered through glass fiber paper. The filtrate was concentrated in vacuum. The crude material was purified by flash chromatography to afford 400 mg the mixture product. The mixture was purified by preparative chiral-HPLC to obtain 4-(4-bromo-2,3-difluoro-phenyl)-1-(2,2-difluoroethyl)-3-methyl-pyrazole (96.7 mg) and 4-(4-bromo-2,3-difluoro-phenyl)-1-(2,2-difluoroethyl)-5-methyl-pyrazole (60 mg). MS [M+H]+: 337.1. Intermediate F3 4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)-1H-pyrazole To a solution of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3-(trifluoromethyl)-1H-pyrazole (5 g, 19.1 mmol), 1-bromo-2,3-difluoro-4-iodobenzene (6.08 g, 19.1 mmol), sodium carbonate (6.07 g, 57.2 mmol) and PdCl2(dppf)-CH2Cl2adduct (1.56 g, 1.91 mmol) in dioxane (90 mL) and water (9. mL). The resultant mixture was heated at 100° C. for 10 h under N2. The crude reaction mixture was concentrated in vacuum. The residue was purified by flash chromatography to afford 4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)-1H-pyrazole (3.9 g). MS [M+H]+: 327.0. The following example was prepared in analogy to Intermediate F3. MSESIStartingEx#NameStructure[M + H]+MaterialIntermediate F44-(4-bromo-2- fluoro-phenyl)-3- (trifluoromethyl)- 1H-pyrazole309.14-bromo-2- fluoro-1- iodobenzene and 4- (4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)-3- (trifluoromethyl)- 1H-pyrazole Intermediate G1 2-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane Step 1: 2-[[4-(4-bromo-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane In a 100 mL round-bottomed flask, 4-(4-bromo-2,3-difluorophenyl)-3-methyl-1H-pyrazole (1.7 g, 6.23 mmol) and DIPEA (1.21 g, 9.34 mmol) were combined with THE (30 mL) to give a light brown solution. SEM-Cl (1.56 g, 1.66 mL) was added. The reaction was stirred at room temperature for 1 h. The reaction mixture was poured into 50 mL H2O and extracted with EtOAc (3×30 ml). The organic layers were combined, washed with sat NaCl (1×25 mL), The organic layers were dried over Na2SO4and concentrated in vacuum to afford 2-[[4-(4-bromo-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane(1.51 g). MS [M+H]+: 405.1. Step 2: 2-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane In a 250 mL round-bottomed flask, bis(pinacolato)diboron (1.44 g, 5.65 mmol), 2-[4-(4-bromo-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]ethoxymethyl-trimethyl-silane(1.52 g, 3.77 mmol), PdCl2(dppf)-CH2Cl2adduct (276 mg, 377 μmol) and potassium acetate (1.11 g, 11.3 mmol) were combined with Dioxane (60 mL) to give a dark red solution. The reaction mixture was heated to 80° C. and stirred for 15 h under N2. The crude reaction mixture was concentrated in vacuum. The reaction mixture was poured into 50 mL H2O and extracted with EtOAc (3×50 mL). The organic layers were combined, washed with sat NaCl (1×50 mL), The organic layers were dried over Na2SO4and concentrated in vacuum. The crude material was purified by flash chromatography to afford 2-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (1 g). Intermediate G2 4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-(2-methoxyethyl)-3-methyl-pyrazole Step 1: 4-(4-bromo-2,3-difluoro-phenyl)-1-(2-methoxyethyl)-3-methyl-pyrazole In a 50 mL round-bottomed flask, 1-(2-methoxyethyl)-3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (459 mg, 1.72 mmol), 1-bromo-2,3-difluoro-4-iodobenzene (500 mg, 1.57 mmol), PdCl2(dppf)-CH2Cl2adduct (115 mg, 157 μmol) and Na2CO3(499 mg, 4.7 mmol) were combined with Dioxane (10 mL)/Water (1 mL) to give a dark red solution. The reaction mixture was heated to 100° C. and stirred for 15 h under N2. The reaction mixture was filtered through glass fiber paper. The filtrate was concentrated in vacuum. The crude material was purified by flash chromatography to afford 4-(4-bromo-2,3-difluoro-phenyl)-1-(2-methoxyethyl)-3-methyl-pyrazole (310 mg). MS [M+H]+: 333.1. Step 2: 4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-(2-methoxyethyl)-3-methyl-pyrazole In a 50 mL round-bottomed flask, 4-(4-bromo-2,3-difluoro-phenyl)-1-(2-methoxyethyl)-3-methyl-pyrazole (310 mg, 936 μmol), 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (357 mg, 1.4 mmol), PdCl2(dppf)-CH2Cl2adduct (68.5 mg, 93.6 μmol) and potassium acetate (276 mg, 2.81 mmol) were combined with dioxane (10 mL) to give a dark red solution. The reaction mixture was heated to 100° C. and stirred for 15 h under N2. The reaction mixture was filtered through glass fiber paper. The crude reaction mixture was concentrated in vacuum. The crude material was purified by flash chromatography to afford 4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-(2-methoxyethyl)-3-methyl-pyrazole (350 mg). MS [M+H]+: 379.1. The following intermediates were prepared in analogy to Intermediate G2. MSESIStartingEx #NameStructure[M + H]+MaterialInter- mediate G34-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole379.3Intermediate E2; 1-bromo- 2,3-difluoro- 4- iodobenzene and bis (pinacolato) diboronInter- mediate G41-(2,2-difluoroethyl)- 4-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-5-methyl- pyrazole385.2Intermediate F2 and bis (pinacolato) diboronInter- mediate G51-(2,2-difluoroethyl)- 4-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-3-methyl- pyrazole385.2Intermediate F1 and bis (pinacolato) diboronInter- mediate G64-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-3,5- dimethyl-pyrazole393.2Intermediate E3; 1-bromo- 2,3-difluoro- 4-iodobenze and bis (pinacolato) diboronInter- mediate G71-(2-methoxyethyl)- 3,5-dimethyl-4-[4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]pyrazole357.3Intermediate E3; 1-bromo- 4-iodo- benzene and bis (pinacolato) diboronInter- mediate G8trimethyl-[2-[[3- methyl-4-[4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]pyrazol-1- yl]methoxy]ethyl] silane415.1Intermediate E4; 1-bromo- 4-iodo- benzene and bis (pinacolato) diboronInter- mediate G92-[[3,5-dimethyl-4-[4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]pyrazol-1- yl]methoxy]ethyl- trimethyl-silane429.4Intermediate E5; 1-bromo- 4-iodo- benzene and bis (pinacolato) diboronInter- mediate G104-[3-fluoro-2-methyl- 4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole375.8Intermediate E2; 1-bromo- 2-fluoro-4- iodo-3- methyl- benzene and bis (pinacolato) diboronInter- mediate G114-[3-chloro-2-fluoro- 4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole395.7Intermediate E2; 1-bromo- 2-chloro-3- fluoro-4-iodo- benzene and bis (pinacolato) diboronInter- mediate G121-(2-methoxyethyl)-5- methyl-4-[4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]pyrazole343.9Intermediate E2; 1-bromo- 4-iodo- benzene and bis (pinacolato) diboronInter- mediate G134-[2-fluoro-3-methyl- 4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole375.6Intermediate E2; 1-bromo- 3-fluoro-4- iodo-2- methyl- benzene and bis (pinacolato) diboronInter- mediate G144-[4-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-5-methyl- pyrazol-1-yl]-2- methyl-butan-2-ol407.3Intermediate E6; 1-bromo- 2,3-difluoro- 4-iodobenze and bis (pinacolato) diboronInter- mediate G152-[4-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-3-methyl- pyrazol-1-yl]-N- methyl-acetamide392.2Intermediate E7; 1-bromo- 2,3-difluoro- 4-iodobenze and bis (pinacolato) diboronInter- mediate G16methyl 2-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3-methyl- pyrazol-1-yl]acetate393.4Intermediate E8; 1-bromo- 2,3-difluoro- 4-iodobenze and bis (pinacolato) diboronInter- mediate G174-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(3- methoxypropyl)-3- methyl-pyrazole393.1Intermediate E11; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G184-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(3- methoxypropyl)-5- methyl-pyrazole393.2Intermediate E12; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G19tert-butyl-[3-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3-methyl- pyrazol-1- yl]propoxy]-dimethyl- silane493.3Intermediate E13; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G20tert-butyl-[3-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5-methyl- pyrazol-1- yl]propoxy]-dimethyl- silane493.3Intermediate E14; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G211-(2-methoxyethyl)-4- [3-methoxy-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-5-methyl- pyrazole373.0Intermediate E2; 1-bromo- 4-iodo-2- methoxy- benzene and bis (pinacolato) diboronInter- mediate G221-(2-methoxyethyl)-5- methyl-4-[3-methyl-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]pyrazole357.0Intermediate E2; 1-bromo- 4-iodo-2- methyl- benzene and bis (pinacolato) diboronInter- mediate G234-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-5-ethyl-1- (2-methoxyethyl) pyrazole393.1Intermediate E18; 1- bromo-2,3- difluoro-4- iodobenzene and bis (pinacolato) diboronInter- mediate G24tert-butyl-[4-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-2-(2- trimethylsilylethoxy- methyl)pyrazol-3- yl]butoxy]-dimethyl- silane623.4Intermediate E19; 1- bromo-2,3- difluoro-4- iodobenzene and bis (pinacolato) diboronInter- mediate G254-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- (methoxymethyl) pyrazole409.1Intermediate E20; 1- bromo-2,3- difluoro-4- iodobenzene and bis (pinacolato) diboronInter- mediate G26tert-butyl-[2-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5-methyl- pyrazol-1-yl]ethoxy]- dimethyl-silane479.4Intermediate E10; 1- bromo-2,3- difluoro-4- iodobenzene and bis (pinacolato) diboronInter- mediate G27tert-butyl-[2-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3-methyl- pyrazol-1-yl]ethoxy]- dimethyl-silane479.4Intermediate E9; 1-bromo- 2,3-difluoro- 4- iodobenzene and bis (pinacolato) diboronInter- mediate G284-[3-chloro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole377.8Intermediate E2; 1-bromo- 2-chloro-4- iodo-benzene and bis (pinacolato) diboronInter- mediate G294-[2-fluoro-3- methoxy-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole391.2Intermediate E2; 1-bromo- 3-fluoro-4- iodo-2- methoxy- benzene and bis (pinacolato) diboronInter- mediate G30methyl 2-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5-methyl- pyrazol-1-yl]acetate393.4Intermediate E15; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G314-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-3-methyl- 1-(tetrahydropyran-4- ylmethyl)pyrazole419.0Intermediate E16; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G322-[[4-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-3-methyl- pyrazol-1- yl]methyl]pyridine330.0Intermediate E17; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G77[2,3-difluoro-4-[1-[(6- methoxy-2- pyridyl)methyl]-3- methyl-pyrazol-4- yl]phenyl]boronic acid360.1Intermediate E21; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G824-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-methyl- pyrazole321.11-methyl-4- (4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)pyrazole; 1-bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G834-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)pyrazole365.2Intermediate E23; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G892-[[4-[2,3-difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-3,5- dimethyl-pyrazol-1- yl]methoxy]ethyl- trimethyl-silane465.43,5-dimethyl- 4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)-1H- pyrazole; 1- bromo-2,3- difluoro-4- iodobenze and bis (pinacolato) diboronInter- mediate G90trimethyl-[2-[[4-[4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]pyrazol-1- yl]methoxy]ethyl]silane401.4trimethyl-[2- [[4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)pyrazol- 1-yl] methoxy] ethyl]silane; 1-bromo-4- iodo-benzene and bis (pinacolato) diboronInter- mediate G924-[2-chloro-5-methyl- 4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole391.2Intermediate E2; 1-bromo- 5-chloro-4- iodo-2- methyl- benzene and bis (pinacolato) diboronInter- mediate G934-[2-chloro-3-fluoro- 4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole395.6Intermediate E2; 1-bromo- 3-chloro-2- fluoro-4-iodo- benzene and bis (pinacolato) diboron Intermediate G33 1-[2-(difluoromethoxy)ethyl]-4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazole Step 1: 2-[4-(4-bromo-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]ethoxy-tert-butyl-dimethyl-silane To a solution of 1-bromo-2,3-difluoro-4-iodobenzene (1000 mg, 3.14 mmol) in the mixture solvent of dioxane (10 mL) and Water (2 mL) was added sodium carbonate (665 mg, 6.27 mmol), 1-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (1.15 g, 3.14 mmol) and PdCl2(dppf)-CH2Cl2adduct (256 mg, 314 μmol). The reaction was stirred for 3 h at 130° C. under microwave irritation and atmosphere of argon. The mixture was filtered and the filtrate was concentrated in vacuum. The residue was purified by column chromatography to give 2-[4-(4-bromo-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]ethoxy-tert-butyl-dimethyl-silane (1 g). MS [M+H]+: 431.1. Step 2: 2-[4-(4-bromo-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]ethanol To a solution of 4-(4-bromo-2,3-difluorophenyl)-1-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methyl-1H-pyrazole (1 g, 2.32 mmol) in THE (10 mL) was added TBAF (6.95 mL, 6.95 mmol), the reaction was stirred for 1 h at room temperature. The reaction mixture was washed with brine (20 mL) and extracted in DCM (30 mL). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuum. The residue was purified by column chromatography to give 2-[4-(4-bromo-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]ethanol (500 mg). MS [M+H]+: 317.0. Step 3: 4-(4-bromo-2,3-difluoro-phenyl)-1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazole To a solution of 2-(4-(4-bromo-2,3-difluorophenyl)-3-methyl-1H-pyrazol-1-yl)ethan-1-ol (450 mg, 1.42 mmol) in Acetonitrile (5 mL) was added copper (I) iodide (54 mg, 284 μmol), the reaction was heated to 60° C., then the solution of 2,2-difluoro-2-(fluorosulfonyl)acetic acid (505 mg, 2.84 mmol) in Acetonitrile (5 mL) was added dropwise over 5 min. The reaction was stirred for another 30 min. The reaction was cooled to room temperature and the mixture was concentrated in vacuum. The residue was purified by column chromatography to give 4-(4-bromo-2,3-difluoro-phenyl)-1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazole (200 mg). MS [M+H]+: 367.0. Step 4: 1-[2-(difluoromethoxy)ethyl]-4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazole To a solution of 4-(4-bromo-2,3-difluorophenyl)-1-(2-(difluoromethoxy)ethyl)-3-methyl-1H-pyrazole (200 mg, 545 μmol) in Dioxane (3 mL) was added potassium acetate (107 mg, 1.09 mmol), PdCl2(dppf)-CH2Cl2adduct (44.5 mg, 54.5 μmol) and bis(pinacolato)diboron (138 mg, 545 μmol), the reaction was stirred for 15 h at 80° C. under atmosphere of argon. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuum and the residue was purified by column chromatography to give 1-[2-(difluoromethoxy)ethyl]-4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazole (150 mg). MS [M+H]+: 415.2. The following intermediates were prepared in analogy to Intermediate G33. MSESIStartingEx #NameStructure[M + H]+MaterialInter- mediate G341-[2- (difluoromethoxy) ethyl]-4-[2,3- difluoro-4- (4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl]-5-methyl- pyrazole415.2Intermediate E10; 1- bromo-2,3- difluoro-4- iodobenze; TBAF; 2,2- difluoro-2- (fluorosulfonyl) acetic acid and bis(pinacolato) diboron Intermediate G35 4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-isopropyl-1-(2-methoxyethyl)pyrazole Step 1: 4-bromo-3-isopropyl-1-(2-methoxyethyl)pyrazole To a solution of 4-bromo-3-isopropyl-1H-pyrazole (1000 mg, 5.29 mmol) in acetonitrile (10 mL) was added 1-bromo-2-methoxy-ethane (735.2 mg, 5.29 mmol) and cesium carbonate (3.45 g, 10.58 mmol), the reaction was stirred for 8 h at 100° C. The reaction was mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuum and the residue was purified by Chiral HPLC to give 4-bromo-3-isopropyl-1-(2-methoxyethyl)pyrazole (1.2 g). MS [M+H]+: 247.0. Step 2: 2,3-difluoro-4-[3-isopropyl-1-(2-methoxyethyl)pyrazol-4-yl]phenol To a solution of 4-bromo-3-isopropyl-1-(2-methoxyethyl)pyrazole (900 mg, 3.64 mmol) in the mixture solvent of 1,4-dioxane (12 mL) and water (2.4 mL) was added (2,3-difluoro-4-hydroxy-phenyl)boronic acid (1.27 g, 7.28 mmol), tetrakis(triphenylphosphine)palladium (420.83 mg, 0.364 mmol) and sodium carbonate (1.16 g, 10.93 mmol), the reaction was stirred for 3 h at 100° C. under atmosphere of nitrogen. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuum. The residue was purified by column chromatography to give 2,3-difluoro-4-[3-isopropyl-1-(2-methoxyethyl)pyrazol-4-yl]phenol (210 mg). MS [M+H]+: 297.1. Step 3: [2,3-difluoro-4-[3-isopropyl-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]trifluoromethanesulfonate To a solution of 2,3-difluoro-4-[3-isopropyl-1-(2-methoxyethyl)pyrazol-4-yl]phenol (100 mg, 0.337 mmol) in N,N-dimethylformamide (3 mL) was added 1,1,1-trifluoro-N-phenyl-N-triflyl-methanesulfonamide (144.68 mg, 0.405 mmol), triethylamine (68.3 mg, 0.675 mmol) and n-(4-pyridyl)dimethylamine (4.12 mg, 0.034 mmol), the reaction was stirred for 1 h at room temperature. The reaction mixture was concentrated in vacuum. The residue was purified by flash column chromatography to give [2,3-difluoro-4-[3-isopropyl-1-(2-methoxyethyl)pyrazol-4-yl]phenyl] trifluoromethanesulfonate (135 mg). MS [M+H]+: 429.1. Step 4: 4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-isopropyl-1-(2-methoxyethyl)pyrazole To a solution of trifluoromethanesulfonic acid [2,3-difluoro-4-[3-isopropyl-1-(2-methoxyethyl)pyrazol-4-yl]phenyl] ester (80 mg, 0.187 mmol) in anhydrous 1,4-dioxane (3 mL) was added 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (47.42 mg, 0.187 mmol), potassium acetate (36.66 mg, 0.374 mmol) and [1,1′-bis(diphenylphosphino)ferrocene]palladium(ii) dichloride dichloromethane adduct (15.25 mg, 0.019 mmol), the reaction was stirred for 5 h at 100° C. under atmosphere of nitrogen. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuum. The residue was purified by column chromatography to give 4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-isopropyl-1-(2-methoxyethyl)pyrazole (60 mg). MS [M+H]+: 407.2. The following intermediates were prepared in analogy to Intermediate G35. MSESIStartingEx #NameStructure[M + H]+MaterialInter- mediate G365- (difluoromethyl)- 4-[2,3-difluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan- 2-yl)phenyl]-1-(2- methoxyethyl) pyrazole415.2Intermediate R4; 1,1,1- trifluoro-N- phenyl-N- triflyl- methane- sulfonamide and bis (pinacolato) diboronInter- mediate G374-[2,3-difluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan- 2-yl)phenyl]-3- fluoro-1-(2- methoxyethyl) pyrazole383.24-bromo-3- fluoro-1H- pyrazole; 1- bromo-2- methoxy- ethane; 1,1,1- trifluoro-N- phenyl-N- triflyl- methane- sulfonamide and bis (pinacolato) diboron Intermediate G38 3-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazol-1-yl]propanamide Step 1: 3-[4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)pyrazol-1-yl]propanamide 4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazole (500 mg, 1.53 mmol), 3-bromopropanamide (279 mg, 1.83 mmol) and potassium carbonate (634 mg, 4.59 mmol) were heated in anhydrous acetonitrile (7.64 mL) at 60° C. for 18 h. The mixture was cooled to room temperature, and 100-200 mesh silica gel was added to absorb the material. The loaded sample was purified by flash chromatography to afford the final compound as yellow oil (550 mg). MS [M+H]+: 398.0. Step 2: 3-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazol-1-yl]propanamide To a solution of 3-(4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)propanamide (590 mg, 1.48 mmol) in dioxane (14.8 mL) was added 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (452 mg, 1.78 mmol), potassium acetate (436 mg, 4.45 mmol) and PdCl2(dppf)-CH2Cl2(122 mg, 148 μmol), the reaction was stirred for 18 hours at 100° C. under atmosphere of argon. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuum and the residue was purified by column chromatography to afford the product as yellow solid (390 mg). MS [M+H]+: 446.2. The following examples were prepared in analogy to Intermediate G38. MSESIStartingEx #NameStructure[M + H]+MaterialInter- mediate G394-[2,3-difluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-1-(2- methylsulfonylethyl)- 3-(trifluoromethyl) pyrazole481.2Intermediate F3 and 1- methylsulfonylethyleneInter- mediate G404-[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3- (trifluoromethyl) pyrazol-1- yl]butanamide460.2Intermediate F3 and 4- bromobutanamideInter- mediate G412-[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3- (trifluoromethyl) pyrazol-1- yl]acetamide432.1Intermediate F3 and 2- iodoacetamideInter- mediate G424-[2,3-difluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-3- (trifluoromethyl) pyrazole433.2Intermediate F3 and 1- bromo-2- methoxy- ethaneInter- mediate G434-[2-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5- (trifluoromethyl) pyrazol-1- yl]ethyl]morpholine488.2Intermediate F3 and 4-(2- bromoethyl) morpholineInter- mediate G444-[2,3-difluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-1-(3- methoxypropyl)-3- (trifluoromethyl) pyrazole447.2Intermediate F3 and 1- bromo-3- methoxy- propaneInter- mediate G454-[2,3-difluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-1- [[(4S)-2,2- dimethyl-1,3- dioxolan-4- yl]methyl]-3- (trifluoromethyl) pyrazole489.2Intermediate F3 and (S)- (2,2- dimethyl-1,3- dioxolan-4- yl)methyl 4- methylbenzenesulfonateInter- mediate G462-[[4-[2-fluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3- (trifluoromethyl) pyrazol-1- yl]methoxy]ethyl- trimethyl-silane487.2Intermediate F4 and 2- (Trimethylsilyl) ethoxymethyl chloride Intermediate G47 1-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazol-1-yl]propan-2-ol Step 1: 1-[4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)pyrazol-1-yl]propan-2-one 4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazole (110 mg, 336 μmol), 1-bromopropan-2-one (55.3 mg, 404 μmol) and potassium carbonate (139 mg, 1.01 mmol) were stirred in anhydrous acetonitrile (3.36 mL) at room temperature for 30 min. 100-200 mesh silica gel was added to absorb the material. The loaded sample was purified by flash chromatography to afford the final compound as light yellow oil (120 mg). MS [M+H]+: 383.0. Step 2: 1-[4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)pyrazol-1-yl]propan-2-ol 1-(4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)propan-2-one (120 mg, 313 μmol) was dissolved in MeOH. The solution was cooled to 0° C. To this solution was added sodium tetrahydroborate (11.8 mg, 313 μmol), and the resulting mixture was stirred for 1 h at the same temperature. 100-200 mesh silica gel was added to absorb the material; the loaded sample was then purified by flash chromatography to afford the final compound as light yellow oil (110 mg). MS [M+H]+: 385.0. Step 3: 1-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazol-1-yl]propan-2-ol To a solution of 1-(4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)propan-2-ol (120 mg, 312 μmol) in dioxane (3.12 mL) was added 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (94.9 mg, 374 μmol), potassium acetate (91.7 mg, 935 μmol) and PdCl2(dppf)-CH2Cl2(25.5 mg, 31.2 μmol), the reaction was stirred for 18 hours at 100° C. under atmosphere of argon. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuum and the residue was purified by column chromatography to afford the final compound as light brown oil (90 mg). MS [M+H]+: 433.2. Intermediate G48 1-[2-(difluoromethoxy)ethyl]-4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazole Step 1: 2-[4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)pyrazol-1-yl]ethanol To a solution of 4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazole (600 mg, 1.83 mmol) in DMF (15 mL) was added 2-iodoethan-1-ol (315 mg, 1.83 mmol) and potassium carbonate (761 mg, 5.5 mmol), the reaction was stirred for 3 hours at 90° C. The reaction mixture was cooled to room temperature and washed with brine, extracted in DCM. The organic layer was dried over anhydrous Na2SO4and concentrated in vacuum. The residue was purified by column chromatography to give 2-[4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)pyrazol-1-yl]ethanol (640 mg). MS [M+H]+: 371.1. Step 2: 4-(4-bromo-2,3-difluoro-phenyl)-1-[2-(difluoromethoxy)ethyl]-3-(trifluoromethyl)pyrazole To a solution of 2-(4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)ethan-1-ol (500 mg, 1.35 mmol) in Acetonitrile (5 mL) was added copper (I) iodide (51.3 mg, 269 μmol), the reaction was heated to 60° C., then the solution of 2,2-difluoro-2-(fluorosulfonyl)acetic acid (480 mg, 2.69 mmol) in Acetonitrile (5 mL) was added dropwise over 5 min. The reaction was stirred for another 30 min. The reaction was cooled to room temperature and the mixture was concentrated in vacuum. The residue was purified by f column chromatgraphy to give 4-(4-bromo-2,3-difluoro-phenyl)-1-[2-(difluoromethoxy)ethyl]-3-(trifluoromethyl)pyrazole (260 mg). MS [M+H]+: 421.1. Step 3: 1-[2-(difluoromethoxy)ethyl]-4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazole To a solution of 4-(4-bromo-2,3-difluorophenyl)-1-(2-(difluoromethoxy)ethyl)-3-(trifluoromethyl)-1H-pyrazole (260 mg, 617 μmol) in Dioxane (3 mL) was added potassium acetate (121 mg, 1.23 mmol), PdCl2(dppf)-CH2Cl2(50.4 mg, 61.7 μmol) and bis(pinacolato)diboron (157 mg, 617 μmol), the reaction was stirred for 15 h at 80° C. under atmosphere of argon. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuum and the residue was purified by column chromatography to give 1-[2-(difluoromethoxy)ethyl]-4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazole (205 mg). MS [M+H]+: 469.1. Intermediate G49 3-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazol-1-yl]-2-methyl-propan-1-ol Step 1: 4-(4-bromo-2,3-difluoro-phenyl)-1-(2-methylallyl)-3-(trifluoromethyl)pyrazole To a solution of 4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazole (600 mg, 1.83 mmol) in DMF (5 mL) was added 3-iodo-2-methylprop-1-ene (334 mg, 1.83 mmol) and potassium carbonate (761 mg, 5.5 mmol), the reaction was stirred for 3 h at 90° C. The reaction mixture was cooled to room temperature and washed with brine, extracted in DCM. The organic layer was dried over anhydrous Na2SO4and concentrated in vacuum. The crude product was purified by column chromatography to give 4-(4-bromo-2,3-difluoro-phenyl)-1-(2-methylallyl)-3-(trifluoromethyl)pyrazole (400 mg). MS [M+H]+: 381.1. Step 2: 3-[4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)pyrazol-1-yl]-2-methyl-propan-1-ol To a solution of 4-(4-bromo-2,3-difluorophenyl)-1-(2-methylallyl)-3-(trifluoromethyl)-1H-pyrazole (400 mg, 1.05 mmol) in THE (5 mL) was added Borane tetrahydrofuran complex solution (2.1 mL, 2.1 mmol) drop wise at room temperature under atmosphere of nitrogen. The reaction was stirred for 4 h, then water (0.5 mL) was added dropwise, followed by sodium hydroxide (2.1 mL, 3 mol/L) and hydrogen peroxide (2.1 mL, 30%). The reaction was stirred for another 4 h. The reaction mixture was concentrated in vacuum and the residue was purified by column chromatography to give 3-[4-(4-bromo-2,3-difluoro-phenyl)-3-(trifluoromethyl)pyrazol-1-yl]-2-methyl-propan-1-ol (320 mg). MS [M+H]+: 399.1. Step 3: 3-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazol-1-yl]-2-methyl-propan-1-ol To a solution of 3-(4-(4-bromo-2,3-difluorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)-2-methylpropan-1-ol (400 mg, 1 mmol) in dioxane (5 mL) was added potassium acetate (197 mg, 2 mmol), PdCl2(dppf)-CH2Cl2(81.8 mg, 100 μmol) and bis(pinacolato)diboron (254 mg, 1 mmol), the reaction was stirred for 15 h at 80° C. under atmosphere of argon. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuum and the residue was purified by column chromatography to give 3-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-(trifluoromethyl)pyrazol-1-yl]-2-methyl-propan-1-ol (260 mg). MS [M+H]+: 447.1. Intermediate G50 [4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid Step 1: 1-(4-benzyloxy-2,3-difluoro-phenyl)butan-2-one To a solution of 2-(4-benzyloxy-2,3-difluoro-phenyl)-N-methoxy-N-methyl-acetamide (6.7 g, 20.85 mmol) in THE (50.0 mL) was added ethylmagnesium bromide in Et2O (3M) (10.43 mL, 31.28 mmol) slowly at −40° C. under N2. This reaction mixture was stirred at −10° C. for 2 h. This reaction was quenched by NH4Cl (50.0 mL) and was extracted by EtOAc (50.0 mL×2). The combined organic layers were dried over Na2SO4and concentrated to get the crude product. This crude product was purified by silica gel chromatography (PE:EtOAc=8:1) to get 1-(4-benzyloxy-2,3-difluoro-phenyl)butan-2-one (1.4 g) as yellow solid. Step 2: 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-ethyl-1-(2-methoxyethyl)pyrazole To a mixture of 1-(4-benzyloxy-2,3-difluoro-phenyl)butan-2-one (700.0 mg, 2.41 mmol) and molecular sieves 4A (500.0 mg) in toluene (8.0 mL) was added 2-methoxyethylhydrazine (521.53 mg, 5.79 mmol) in one portion. The reaction mixture was stirred at 100° C. for 3h. The mixture was filtered and concentrated under reduced pressure affording the residue. The mixture of residue and N,N-dimethylformamide dimethyl acetal (7.07 mL, 86.81 mmol) was stirred at 100° C. for 16 h. This reaction was quenched by H2O (10.0 mL) and was extracted by EtOAc (10.0 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product. The crude product was purified by silica gel chromatography (PE:EtOAc=5:1) to get 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-ethyl-1-(2-methoxyethyl)pyrazole (630.0 mg) as yellow oil. MS [M+H]+: 373.2. Step 3: 4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol To a solution of 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-ethyl-1-(2-methoxyethyl)pyrazole (630.0 mg, 1.69 mmol) in methanol (6 mL) was added palladium on carbon (180.03 mg) in one portion under N2. This mixture was degassed and purged with N2for 3 times. Then H2(15 psi) was introduced into this system. The reaction mixture was stirred at 20° C. for 16 h under H2atmosphere. The mixture was filtered and concentrated under reduced pressure affording the crude product 4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol (470.0 mg) as a black oil. MS [M+H]+: 283.2. Step 4: [4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]trifluoromethanesulfonate A mixture of 4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol (220.0 mg, 0.78 mmol) and pyridine (0.09 mL, 1.17 mmol) in DCM (5 mL) was degassed and purged with N2for 3 times. Then trifluoromethanesulfonic anhydride (0.15 mL, 0.94 mmol) was added dropwise into the mixture at 0° C. The reaction mixture was stirred at 20° C. for 2 h under N2atmosphere. This reaction was quenched by NaHCO3(10 mL) and was extracted by DCM (10 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product [4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate (390.0 mg) as red oil. MS [M+H]+: 415.1. Step 5: [4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid A mixture of bis(pinacolato)diboron (478.03 mg, 1.88 mmol), [4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate (390.0 mg, 0.94 mmol), potassium acetate (0.15 mL, 2.35 mmol) and X-PHOS (44.87 mg, 0.09 mmol) in 1,4-dioxane (5 mL) was degassed and purged with N2for 3 times. Then tris(dibenzylideneacetone)dipalladium (0) (43.1 mg, 0.05 mmol) was added into the mixture. The reaction mixture was stirred at 100° C. for 2 h under N2atmosphere. This reaction was extracted by EtOAc (10 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product. The crude product was purified by preparative HPLC (TFA) to get [4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid (150.0 mg, 0.480 mmol, 49.46% yield) as brown oil. MS [M+H]+: 311.2. Intermediate G51 [4-[3-ethyl-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid Step 1: methyl 4-bromo-1-(2-methoxyethyl)pyrazole-3-carboxylate A mixture of methyl 4-bromo-1H-pyrazole-3-carboxylate (2.5 g, 12.19 mmol) and potassium carbonate (2.53 g, 18.29 mmol) in ACN (10 mL) was degassed and purged with N2for 3 times. Then 1-bromo-2-methoxy-ethane (3.44 mL, 36.58 mmol) was added into the mixture. The reaction mixture was stirred at 80° C. for 2 h under N2atmosphere. The mixture was filtered and concentrated under reduced pressure affording the crude product. The crude product (5 batches) was purified by Prep-HPLC to get methyl 4-bromo-1-(2-methoxyethyl)pyrazole-3-carboxylate (8.0 g). MS [M+H]+: 263.0. Step 2: [4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]methanol To a solution of methyl 4-bromo-1-(2-methoxyethyl)pyrazole-3-carboxylate (2.0 g, 7.6 mmol) in THE (20.0 mL) was added lithium borohydride (5.7 mL, 11.4 mmol) slowly at −40° C. under N2. This reaction mixture was stirred at 20° C. for 16 h. This reaction was quenched by HCl (1 M, 20 mL) and was extracted by EtOAc (20 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product [4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]methanol (1.6 g). MS [M+H]+: 235.0. Step 3: [4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane To a solution of [4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]methanol (1.2 g, 5.1 mmol) in DMF (10.0 mL) was added imidazole (0.48 mL, 7.15 mmol) and tert-butyldimethylchlorosilane (1.08 g, 7.15 mmol) in one portion. This reaction mixture was stirred at 20° C. for 16 h. This reaction was quenched by brine (10 mL) and was extracted by EtOAc (20 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product. The crude product was purified by silica gel chromatography (PE:EtOAc=5:1) to get [4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane (1.36 g). MS [M+H]+: 349.1. Step 4: [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane A mixture of 2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenol (1.31 g, 3.78 mmol), [4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane (1.36 g, 3.78 mmol) and potassium carbonate (1.04 g, 7.55 mmol) in 1,4-dioxane (10 mL) and water (1 mL) was degassed and purged with N2for 3 times. Then [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (276.36 mg, 0.38 mmol) was added into the mixture. The reaction mixture was stirred at 100° C. for 16 h under N2atmosphere. The mixture was filtered and concentrated under reduced pressure affording the crude product. The crude product was purified by silica gel chromatography (PE:EtOAc=5:1) to get [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane (1.17 g). MS [M+H]+: 489.2. Step 5: 4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol To a solution of [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane (1.0 g, 2.05 mmol) in methanol (10 mL) was added palladium on carbon (217.79 mg) in one portion under N2. This mixture was degassed and purged with N2for 3 times. Then H2(15 psi) was introduced into this system. The reaction mixture was stirred at 20° C. for 2 h under H2atmosphere. The mixture was filtered and concentrated under reduced pressure affording the crude product. The crude product was purified by silica gel chromatography (PE:EtOAc=2:1) to get 4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol (480.0 mg). MS [M+H]+: 399.2. Step 6: [4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate A mixture of 4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol (250.0 mg, 0.63 mmol) and pyridine (0.08 mL, 0.94 mmol) in DCM (5 mL) was degassed and purged with N2for 3 times. Then trifluoromethanesulfonic anhydride (0.12 mL, 0.75 mmol) was added dropwise into the mixture at 0° C. The reaction mixture was stirred at 20° C. for 2 h under N2atmosphere. This reaction was quenched by NaHCO3(10 mL) and was extracted by DCM (10 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product [4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate (370.0 mg). MS [M+H]+: 531.2. Step 7: [4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid A mixture of [4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate (330.0 mg, 0.62 mmol), bis(pinacolato) diboron (315.88 mg, 1.24 mmol), potassium acetate (0.1 mL, 1.55 mmol) and X-PHOS (29.65 mg, 0.06 mmol) in 1,4-dioxane (8 mL) was degassed and purged with N2for 3 times. Then tris(dibenzylideneacetone)dipalladium (0) (28.48 mg, 0.03 mmol) was added into the mixture. The reaction mixture was stirred at 100° C. for 3 h under N2atmosphere. This reaction was extracted by EtOAc (10 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product. The crude product was purified by preparative HPLC (TFA) to get [4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid (210.0 mg). MS [M+H]+: 427.3. Intermediate G52 [2,3-difluoro-4-[1-(2-methoxyethyl)-3-(methylamino)pyrazol-4-yl]phenyl]boronic acid Step 1: 4-bromo-1-(2-methoxyethyl)pyrazole-3-carboxylic acid To a solution of methyl 4-bromo-1-(2-methoxyethyl)pyrazole-3-carboxylate (1.0 g, 3.8 mmol) in THE (10.0 mL), methanol (10.0 mL) and water (2.5 mL) was added lithium hydroxide monohydrate (638.0 mg, 15.2 mmol) in one portion. This reaction mixture was stirred at 25° C. for 4 h. HCl (1 M) was added into this mixture to make pH<3. This mixture was extracted by EtOAc (30 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get 4-bromo-1-(2-methoxyethyl)pyrazole-3-carboxylic acid (900.0 mg), The crude product would be used in the next step directly without further purification. MS [M+H]+: 249.0. Step 2: tert-butyl N-[4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]carbamate To a solution of 4-bromo-1-(2-methoxyethyl)pyrazole-3-carboxylic acid (900.0 mg, 3.61 mmol) and triethylamine (1.01 mL, 7.23 mmol) in tert-butanol (20 mL) was added diphenylphosphonic azide (1.56 mL, 7.23 mmol) in one portion. This reaction mixture was stirred at 80° C. for 4 h. This reaction mixture was concentrated to get the residue. The residue was diluted with EtOAc (30 mL) and washed by saturated aqueous Na2CO3(5 mL×2). The organic layer was dried over Na2SO4and concentrated to get the crude product. This crude product was purified by silica gel chromatography (PE:EtOAc=1:1˜1:2) to get tert-butyl N-[4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]carbamate (1.0 g). MS [M+H]+: 320.0. Step 3: tert-butyl N-[4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]carbamate To a solution of tert-butyl N-[4-bromo-1-(2-methoxyethyl)pyrazol-3-yl]carbamate (1.0 g, 3.12 mmol), 2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenol (1.08 g, 3.12 mmol) and potassium carbonate (0.86 g, 6.25 mmol) in 1,4-dioxane (20 mL) and water (2 mL) was added [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (228.53 mg, 0.31 mmol) in one portion under N2. This reaction mixture was stirred at 100° C. for 16 h. This reaction mixture was filtered, and the filtrate was concentrated to get the residue. This residue was diluted with EtOAc (50 mL) and was washed by brine (10 mL×2). The organic layer was dried over Na2SO4and concentrated to get the crude product. This crude product was purified by silica gel chromatography (PE:EtOAc=5:1˜1:1) to get tert-butyl N-[4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]carbamate (1.1 g). MS [M+H]+: 460.1. Step 4: tert-butyl N-[4-(2,3-difluoro-4-hydroxy-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]carbamate To a solution of tert-butyl N-[4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]carbamate (800.0 mg, 1.74 mmol) in methanol (10 mL) was added palladium on carbon (185.28 mg) in one portion under N2. Then H2(15 psi) was introduced into this system. The reaction mixture was stirred at 25° C. for 16 h. This reaction mixture was filtered, and the filtrate was concentrated to get tert-butyl N-[4-(2,3-difluoro-4-hydroxy-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]carbamate (600.0 mg), the crude product would be used in the next step directly without further purification. MS [M+H]+: 370.0. Step 5: [4-[3-(tert-butoxycarbonylamino)-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate To a solution of tert-butyl N-[4-(2,3-difluoro-4-hydroxy-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]carbamate (600.0 mg, 1.62 mmol) and pyridine (0.2 mL, 2.44 mmol) in DCM (10 mL) was added trifluoromethanesulfonic anhydride (0.32 mL, 1.95 mmol) in one portion at 0° C. Then this reaction mixture was warmed to 25° C. and stirred for 1 h. This reaction was quenched by saturated aqueous NaHCO3(10 mL) and extracted by DCM (10 mL×2). The combined organic layers were dried over Na2SO4and concentrated to get [4-[3-(tert-butoxycarbonylamino)-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate (800.0 mg), MS [M+H]+: 502.0. Step 6: [4-[3-(tert-butoxycarbonylamino)-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid To a solution of bis(pinacolato)diboron (607.73 mg, 2.39 mmol), [4-[3-(tert-butoxycarbonylamino)-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]trifluoromethanesulfonate (800.0 mg, 1.6 mmol) and potassium acetate (313.17 mg, 3.19 mmol) in 1,4-dioxane (10.0 mL) was added tris(dibenzylideneacetone)dipalladium (0) (146.1 mg, 0.16 mmol) and X-PHOS (76.06 mg, 0.16 mmol) in one portion under N2. This reaction mixture was stirred at 100° C. for 2 h. This reaction mixture was filtered, and the filtrate was concentrated to get the crude product. This crude product was purified by Prep-HPLC (TFA) to get [4-[3-(tert-butoxycarbonylamino)-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid (190.0 mg). MS [M+H]+: 342.1. Intermediate G53 2-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-phenyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane Step 1: 3-phenyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazole To a solution of 3-phenyl-1H-pyrazole (1.0 g, 6.94 mmol) in DMF (20 mL) was added sodium hydride (416.2 mg, 10.4 mmol) at 0° C. The mixture was stirred at 0° C. for 1 h. Then 2-(trimethylsilyl)ethoxymethyl chloride (1.6 mL, 9.02 mmol) was added and the mixture was stirred at 0° C. for 12 h. The reaction mixture was poured into water (100 mL) and extracted with EtOAc (100 mL×3), and the organics washed with water (50 mL×2) then saturated brine solution (50 mL×1). The organics were then separated and dried (MgSO4) before concentration to dryness. The crude was then purified by flash column and dried by lyophilization to give trimethyl-[2-[(3-phenylpyrazol-1-yl)methoxy]ethyl]silane (1.85 g). MS [M+H]+: 275.4. Step 2: 4-bromo-3-phenyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazole To a solution of trimethyl-[2-[(3-phenylpyrazol-1-yl)methoxy]ethyl]silane (500.0 mg, 1.82 mmol) in DMF (10 mL) was added N-bromosuccinimide (0.45 mL, 2.37 mmol) at 20° C. The mixture was stirred at 20° C. for 1 h. The reaction was taken up in EtOAc (50 mL) and the organics washed with water (50 mL×2) then saturated brine solution (50 mL×1). The organics were then separated and dried (MgSO4) before concentration to dryness to give 2-[(4-bromo-3-phenyl-pyrazol-1-yl)methoxy]ethyl-trimethyl-silane (300 mg). MS [M+H]+: 353.1. Step 3: 4-(4-(benzyloxy)-2,3-difluorophenyl)-3-phenyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazole To a solution of 2-[(4-bromo-3-phenyl-pyrazol-1-yl)methoxy]ethyl-trimethyl-silane (1.0 g, 2.83 mmol) and 2-(4-benzyloxy-2,3-difluoro-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.1 g, 3.11 mmol) in 1,4-dioxane (20 mL) and water (2 mL) was added potassium carbonate (782.3 mg, 5.66 mmol) and Pd(dppf)Cl2(206.9 mg, 0.28 mmol) under argon in glove box. The mixture was stirred at 90° C. for 2 h. The reaction mixture was filtered and the filtrate was concentrated in vacuum to give a residue, which was purified by flash column and dried by lyophilization to give 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-phenyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (1 g). MS [M+H]+: 493.2. Step 4: 4-(4-(benzyloxy)-2,3-difluorophenyl)-3-phenyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazole To a solution of 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-phenyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (1.0 g, 2.03 mmol) in methanol (20.0 mL) was added Pd/C (1.0 g) under nitrogen. Then the mixture was stirred under hydrogen at 20° C. for 2 h. The reaction mixture was filtered and the filtrate was concentrated in vacuum to give 2,3-difluoro-4-[3-phenyl-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenol (800.0 mg). MS [M+H]+: 403.1. Step 5: 2,3-difluoro-4-(3-phenyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)phenyl trifluoromethanesulfonate To a solution of 2,3-difluoro-4-[3-phenyl-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenol (350.0 mg, 0.87 mmol) in pyridine (10.0 mL) was added trifluoromethanesulfonic anhydride (490.7 mg, 1.74 mmol) at 0° C. The mixture was stirred at 0° C. for 1 h. The reaction was taken up in EtOAc (50 mL) and the organics washed with water (50 mL×2) then saturated brine solution (50 mL×1). The organics were then separated and dried (MgSO4) before concentration to dryness to give [2,3-difluoro-4-[3-phenyl-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl] trifluoromethanesulfonate (400 mg). MS [M+H]+: 535.1. Step 6: 4-(2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3-phenyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazole To a solution of [2,3-difluoro-4-[3-phenyl-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]trifluoromethanesulfonate (400.0 mg, 0.75 mmol) and bis(pinacolato)diboron (228.0 mg, 0.90 mmol) in 1,4-dioxane (10 mL) was added Pd(dppf)Cl2(54.7 mg, 0.07 mmol) and potassium acetate (110.1 mg, 1.12 mmol) under argon in glove box. The mixture was stirred at 100° C. for 1 h. The reaction mixture was filtered and the filtrate was concentrated in vacuum to give a residue, which was purified by flash column and dried by lyophilization to give 2-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-phenyl-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (250.0 mg, 0.49 mmol, 65.2% yield) as a yellow solid. MS [M+H]+: 513.2. Intermediate G54 3-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole Step 1: 3-bromo-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole To a solution of 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole (500.0 mg, 4.62 mmol) in DCM (5.0 mL) was added N-bromosuccinimide (905.2 mg, 5.09 mmol). The mixture was stirred at 20° C. for 12 h under N2. The reaction mixture was quenched by water (10 mL), extracted with DCM (20 mL×3). The combined organic layers were washed brine (20 mL), dried (Na2SO4) and concentrated to give crude product 3-bromo-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole (850.0 mg). MS [M+H]+: 187.0. Step 2: 3-(4-benzyloxy-2,3-difluoro-phenyl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole A mixture of cpd 3-bromo-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole (0.8 g, 4.28 mmol), 2-(4-benzyloxy-2,3-difluoro-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.6 g, 4.70 mmol), K2CO3(1.2 g, 8.55 mmol) and 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (349.0 mg, 0.43 mmol) in a flask. The flask was degassed and purged with N2gas for four times. 1,4-dioxane (5 mL) and water (1 mL) was added by injector to the mixture. The mixture was stirred at 90° C. for 2 h under N2. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to remove the solvent, and the crude was purified by chromatography column flash and concentrated to give 3-(4-benzyloxy-2,3-difluoro-phenyl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole (900.0 mg). MS [M+H]+: 327.1. Step 3: 4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenol To a solution of 3-(4-benzyloxy-2,3-difluoro-phenyl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole (900.0 mg, 2.76 mmol) in THE (20 mL) was added Pd/C (500.0 mg, 2.76 mmol) under N2. The suspension was degassed under vacuum and purged with H2several times at 20° C. for 2 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give 4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenol (600.0 mg) as orange oil. MS [M+H]+: 237.0. Step 4: [4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenyl]trifluoromethanesulfonate A solution of 4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenol (600.0 mg, 2.54 mmol) in Pyridine (6.0 mL) was added trifluoromethanesulfonic anhydride (1.3 mL, 5.08 mmol) under 0° C., The reaction was stirred at 20° C. for 1 h. The reaction was added water (20 mL), extracted with EtOAc (50 mL×3). The combined organic layers were washed by brine (30 mL×2), dried (Na2SO4) and concentrated to give crude product [4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenyl] trifluoromethanesulfonate (900.0 mg). MS [M+H]+: 369.0. Step 5: [4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenyl]boronic acid A mixture of bis(pinacolato)diboron (703.3 mg, 2.77 mmol), 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (188.3 mg, 0.23 mmol), [4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenyl]trifluoromethanesulfonate (850.0 mg, 2.31 mmol) and potassium acetate (453.0 mg, 4.62 mmol) in a flask. 1,4-dioxane (8 mL) was added by injector to the mixture. The flask was degassed and purged with N2gas for four times. The mixture was stirred at 100° C. for 2 h under N2atmosphere. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to remove the solvent, then the product was purified by reversed-phase chromatography (FA) to give [4-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2,3-difluoro-phenyl]boronic acid (210.0 mg). MS [M+H]+: 265.0. Intermediate G55 [2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]boronic acid Step 1: [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane To a solution of 4-[3-[[tert-butyl(dimethyl)silyl]oxymethyl]-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol (200.0 mg, 0.5 mmol) in DMF (5 mL) was added potassium carbonate (138.7 mg, 1.0 mmol) and benzyl bromide (0.07 mL, 0.6 mmol). The reaction mixture was stirred at 20° C. for 4 h. The mixture was filtered and extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, and concentrated under reduced pressure to afford the crude product. The crude product was purified by TLC (PE:EtOAc=3:1) to get [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane (220.0 mg). MS [M+H]+: 489.2. Step 2: [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methanol To a solution of compound [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methoxy-tert-butyl-dimethyl-silane (220.0 mg, 0.45 mmol) in DCM (5 mL) was added hydrochloric acid in MeOH (4M) (1.8 mL, 7.2 mmol). The reaction mixture was stirred at 20° C. for 2 h. The mixture was concentrated under reduced pressure to afford the crude product [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methanol (170.0 mg). MS [M+H]+: 375.2. Step 3: 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-(fluoromethyl)-1-(2-methoxyethyl)pyrazole To a solution of [4-(4-benzyloxy-2,3-difluoro-phenyl)-1-(2-methoxyethyl)pyrazol-3-yl]methanol (150.0 mg, 0.4 mmol) in DCM (5 mL) was added diethylaminosulfur trifluoride (0.21 mL, 1.6 mmol) slowly at −60° C. under N2. This reaction mixture was stirred at −60° C. for 1 h. This reaction was quenched by NaHCO3(20 mL) and was extracted by EtOAc (10 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product. The crude product was purified by TLC (PE:EtOAc=1:1) to get 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-(fluoromethyl)-1-(2-methoxyethyl)pyrazole (130.0 mg). MS [M+H]+: 377.1. Step 4: 2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenol To a solution of 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-(fluoromethyl)-1-(2-methoxyethyl)pyrazole (130.0 mg, 0.35 mmol) in methanol (10 mL) was added palladium on carbon (0.04 mL, 0.03 mmol) in one portion under N2. This mixture was degassed and purged with N2for three times. Then H2(15 psi) was introduced into this system. The reaction mixture was stirred at 20° C. for 2 h under H2atmosphere. The mixture was filtered and concentrated under reduced pressure to afford the crude product. The crude product was purified by TLC (PE:EtOAc=1:1) to get 2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenol (90.0 mg). MS [M+H]+: 287.1. Step 5: [2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]trifluoromethanesulfonate A mixture of 2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenol (90.0 mg, 0.31 mmol) and pyridine (0.05 mL, 0.63 mmol) in DCM (5.0 mL) was degassed and purged with N2for three times. Then trifluoromethanesulfonic anhydride (0.07 mL, 0.44 mmol) was added into the mixture at 0° C. The reaction mixture was stirred at 20° C. for 2 h under N2atmosphere. This reaction was quenched by NaHCO3(10 mL) and was extracted by DCM (10 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product [2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenyl] trifluoromethanesulfonate (130.0 mg). MS [M+H]+: 419.1. Step 6: [2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]boronic acid A mixture of [2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]trifluoromethanesulfonate (130.0 mg, 0.31 mmol), bis(pinacolato)diboron (157.8 mg, 0.62 mmol), potassium acetate (76.3 mg, 0.78 mmol) and X-PHOS (14.8 mg, 0.03 mmol) in 1,4-dioxane (5.0 mL) was degassed and purged with N2for three times. Then tris(dibenzylideneacetone)dipalladium (28.46 mg, 0.03 mmol) was added into the mixture. The reaction mixture was stirred at 100° C. for 2 h under N2atmosphere. This reaction was filtered and concentrated to get the crude product. The crude product was purified by TLC (PE:EtOAc=3:1) to get [2,3-difluoro-4-[3-(fluoromethyl)-1-(2-methoxyethyl)pyrazol-4-yl]phenyl]boronic acid (70.0 mg). MS [M+H]+: 315.1. Intermediate G56 [4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid Step 1: 3-chloro-1-(2-methoxyethyl)pyrazole A mixture of 3-chloro-1H-pyrazole (4.5 g, 43.89 mmol) and potassium carbonate (9.1 g, 65.84 mmol) in ACN (100 mL) was degassed and purged with N2for three times. Then 1-bromo-2-methoxy-ethane (12.37 mL, 131.68 mmol) was added into the mixture. The reaction mixture was stirred at 80° C. for 16 h under N2atmosphere. The mixture was filtered and concentrated under reduced pressure to afford the crude product. The crude product was extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, and concentrated under reduced pressure to afford the crude product. The crude product was purified by silica gel chromatography, to get 3-chloro-1-(2-methoxyethyl)pyrazole (6.5 g) as yellow oil. MS [M+H]+: 161.1. Step 2: 4-bromo-3-chloro-1-(2-methoxyethyl)pyrazole To a solution of 3-chloro-1-(2-methoxyethyl)pyrazole (1.0 g, 6.23 mmol) in ACN (10 mL) was added N-bromosuccinimide (1.22 g, 6.85 mmol). The reaction mixture was stirred at 20° C. for 6 h. This reaction was quenched by Na2SO3(40 mL) and was extracted by EtOAc (40 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product 4-bromo-3-chloro-1-(2-methoxyethyl)pyrazole (1.8 g). MS ([M+H]+/[M+2+H]+): 239.0/241.0. Step 3: 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-chloro-1-(2-methoxyethyl)pyrazole A mixture of 4-bromo-3-chloro-1-(2-methoxyethyl)pyrazole (1.8 g, 7.52 mmol), 2-(4-benzyloxy-2,3-difluoro-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (3.9 g, 11.27 mmol) and potassium carbonate (2.08 g, 15.03 mmol) in 1,4-dioxane (20 mL) and water (2 mL) was degassed and purged with N2for three times. Then [1,1′-bis(diphenylphosphino) ferrocene]dichloropalladium(II) (274.97 mg, 0.38 mmol) was added dropwise into the mixture. The reaction mixture was stirred at 80° C. for 9 h under N2atmosphere. The mixture was filtered and concentrated under reduced pressure to afford the crude product. The crude product was purified by silica gel chromatography (PE:EtOAc=10:1˜2:1) to get 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-chloro-1-(2-methoxyethyl)pyrazole (1.7 g). MS [M+H]+: 379.1. Step 4: 4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol To a solution of 4-(4-benzyloxy-2,3-difluoro-phenyl)-3-chloro-1-(2-methoxyethyl)pyrazole (3.4 g, 8.98 mmol) in methanol (30 mL) was added platinum(IV) oxide (101.91 mg, 0.45 mmol) in one portion under Ar2. This mixture was degassed and purged with Ar2for 3 times. Then H2(15 psi) was introduced into this system. The reaction mixture was stirred at 20° C. for 6 h under H2atmosphere. This reaction was filtered carefully and concentrated to get the crude product. The crude product was purified by silica gel chromatography (PE:EtOAc=4:1˜1:1) to get 4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol (2.4 g). MS [M+H]+: 289.1. Step 5: [4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]trifluoromethanesulfonate A mixture of 4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenol (500.0 mg, 1.73 mmol) and pyridine (0.28 mL, 3.46 mmol) in DCM (5 mL) was degassed and purged with N2for 3 times. Then trifluoromethanesulfonic anhydride (0.4 mL, 2.42 mmol) was added into the mixture at 0° C. The reaction mixture was stirred at 20° C. for 2 h under N2atmosphere. This reaction was quenched by NaHCO3(10 mL) and was extracted by DCM (10 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product [4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate (750.0 mg). MS [M+H]+: 421.0. Step 6: [4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid A mixture of [4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]trifluoromethanesulfonate (900.0 mg, 2.14 mmol), bis(pinacolato)diboron (1.09 g, 4.28 mmol), potassium acetate (524.84 mg, 5.35 mmol) and X-PHOS (50.99 mg, 0.11 mmol) in 1,4-dioxane (5 mL) was degassed and purged with N2for 3 times. Then tris(dibenzylideneacetone)dipalladium (0) (97.94 mg, 0.11 mmol) was added into the mixture. The reaction mixture was stirred at 90° C. for 2 h under N2atmosphere. This reaction was filtered and concentrated to get the crude product. The crude product was purified by TLC (PE:EA=3:1) to get [4-[3-chloro-1-(2-methoxyethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]boronic acid (450.0 mg). MS [M+H]+: 317.0. The following examples were prepared in analogy to Intermediate G54. MSESIStartingEx #NameStructure[M + H]+MaterialInter- mediate G57[4-[5-chloro-1-(2- methoxyethyl) pyrazol-4-yl]-2,3- difluoro- phenyl]boronic acid481.23-chloro-1H- pyrazole; 1- bromo-2- methoxy- ethane, 2-(4- benzyloxy-2,3- difluoro- phenyl)- 4,4,5,5- tetramethyl- 1,3,2- dioxaborolane Intermediate G58 [2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]boronic acid Step 1: 2-(4-bromo-1H-pyrazol-5-yl)pyridine To a solution of 2-(1H-pyrazol-5-yl)pyridine (3.0 g, 20.7 mmol) in acetic acid (20 mL) was added and bromine (3.6 g, 22.7 mmol), the mixture was stirred at 20° C. for 0.5 h. The mixture was concentrated, the residue was diluted with 20 mL of water, the solution was added to the solution of NaOH in water (30 mL, 1 M), the precipitate was filtered off and dried in vacuum to give 2-(4-bromo-1H-pyrazol-5-yl)pyridine (4.6 g). MS [M+H]+: 223.9. Step 2: 2-(4-bromo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-5-yl)pyridine To a solution of 2-(4-bromo-1H-pyrazol-5-yl)pyridine (4.5 g, 20.08 mmol) in ACN (50.0 mL) was added cesium carbonate (13.1 g, 40.17 mmol) and 2-(trimethylsilyl)ethoxymethyl chloride (4.27 mL, 24.1 mmol), the mixture was stirred at 70° C. for 1 h. The mixture was filtered off, the filtrate was concentrated, the residue was purified by prep-HPLC to give 2-[[4-bromo-5-(2-pyridyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (3.82 g). MS [M+H]+: 354.0. Step 3: 2-(4-(4-(benzyloxy)-2,3-difluorophenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-5-yl)pyridine To a solution of 2-[[4-bromo-5-(2-pyridyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (1.0 g, 2.82 mmol) in 1,4-dioxane (10 mL) was added 2-(4-benzyloxy-2,3-difluoro-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.2 g, 3.39 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (206.5 mg, 0.28 mmol), potassium carbonate (780.2 mg, 5.64 mmol) and dioxane (10 mL)/water (1 mL) in glove box, the mixture was stirred at 100° C. for 16 h under Ar2. The mixture was diluted with 10 mL of ethyl acetate and filtered off, the filtrated was concentrated, the residue was purified by Flash-HPLC to give 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-5-(2-pyridyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (520 mg). MS [M+H]+: 494.2. Step 4: 2,3-difluoro-4-(5-(pyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)phenol To a solution of 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-5-(2-pyridyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (470.0 mg, 0.95 mmol) in Methanol (10 mL) was added Pd/C (80.0 mg) under N2, then the mixture was flashed with H2and stirred with a H2balloon at 20° C. for 2 h. The mixture was filtered off, the filtrate was concentrated to give 2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenol (290.0 mg). MS [M+H]+: 404.1. Step 5: 2,3-difluoro-4-(5-(pyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)phenyl trifluoromethanesulfonate To solution of 2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenol (290.0 mg, 0.72 mmol) in pyridine (4.0 mL) was added trifluoromethanesulfonic anhydride (405.5 mg, 1.44 mmol) at 0° C. under N2. The mixture was stirred at 20° C. for 0.5 h. the mixture was diluted with 50 mL of ethyl acetate, washed with 20 mL of water and 20 mL of brine, the organic layer was dried over Na2SO4and concentrated to give [2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl] trifluoromethanesulfonate (384.0 mg). MS [M+H]+: 536.1. Step 6: (2,3-difluoro-4-(5-(pyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)phenyl)boronic acid To a solution of [2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl] trifluoromethanesulfonate (350.0 mg, 0.65 mmol) in 1,4-dioxane (2.0 mL) was added potassium acetate (0.08 mL, 1.31 mmol), bis(pinacolato)diboron (331.9 mg, 1.31 mmol) and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (47.8 mg, 0.07 mmol) in glove box. The mixture was stirred at 100° C. for 8 h. The mixture was filtered off, the filtrate was concentrated, the residue was purified by Prep-HPLC to give [2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]boronic acid (500.0 mg). MS [M+H]+: 432.1. Intermediate G59 2-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane Step 1: 2-[(4-bromopyrazol-1-yl)methoxy]ethyl-trimethyl-silane To a solution of 4-bromo-1H-pyrazole (20.0 g, 136.08 mmol) in THE (500 mL) was added sodium hydride, 60% in oil (6.5 g, 163.3 mmol) slowly at 0° C. After addition, this reaction mixture was stirred at 0° C. for 1 h. Then 2-(trimethylsilyl)ethoxymethyl chloride (36.1 mL, 204.12 mmol) was added into this mixture slowly at 0° C. This reaction mixture was warmed to 20° C. and stirred for 15 h. This reaction was quenched by saturated aqueous NH4Cl (50 mL) and extracted by EtOAc (50 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get 2-[(4-bromopyrazol-1-yl)methoxy]ethyl-trimethyl-silane (32.0 g). Step 2: 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane To a solution of 2-(4-benzyloxy-2,3-difluoro-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (12.5 g, 36.07 mmol), 2-[(4-bromopyrazol-1-yl)methoxy]ethyl-trimethyl-silane (10.0 g, 36.07 mmol) and potassium carbonate (7.5 g, 54.11 mmol) in 1,4-dioxane (200 mL) and water (20 mL) was added [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (1.3 g, 1.8 mmol) in one portion under N2. This reaction mixture was stirred at 100° C. for 4 h. and the filtrate was concentrated to get the residue. This residue was diluted with EtOAc (200 mL) and was washed by brine (50 mL×2). The organic layer was dried over Na2SO4and concentrated to get the crude product. This crude product was purified by silica gel chromatography (PE:EtOAc=5:1˜1:1) to get 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (10.0 g). MS [M+H]+: 417.2. Step 3: 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-5-bromo-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane To a solution of 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (9.0 g, 21.61 mmol) in DMF (100 mL) was added N-bromosuccinimide (4.6 g, 25.93 mmol) in one portion. This reaction mixture was stirred at 80° C. for 16 h. This reaction was quenched by saturated aqueous Na2SO3(100 mL) and extracted by EtOAc (50 mL×3). The combined organic layers were dried over Na2SO4and concentrated to get the crude product. This crude product was purified by Prep-HPLC to get 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-5-bromo-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (3.0 g). MS [M+H]+: 496.9. Step 4: 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane To a solution of 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-5-bromo-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (184.0 mg, 1.21 mmol), compound 5 (400.0 mg, 0.81 mmol) and potassium carbonate (223.2 mg, 1.61 mmol) in 1,4-dioxane (10 mL) and water (1 mL) was added [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (59.1 mg, 0.08 mmol) in one portion under N2. This reaction mixture was stirred at 100° C. for 4 h. This reaction mixture was filtered, and the filtrate was concentrated to get the residue. This residue was diluted with EtOAc (50 mL) and was washed by brine (10 mL×2). The organic phase was dried over Na2SO4and concentrated to get the crude product. This crude product was purified by silica gel chromatography to get 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (330.0 mg). MS [M+H]+: 523.2. Step 5: 2,3-difluoro-4-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenol To a solution of 2-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (280.0 mg, 0.54 mmol) in methanol (20 mL) was added palladium on carbon (10%, 57.0 mg) in one portion under N2. Then H2(15 psi) was introduced into this system. This reaction mixture was stirred at 20° C. for 16 h. This reaction mixture was filtered, and the filtrate was concentrated to get 2,3-difluoro-4-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenol (230.0 mg, 0.53 mmol), which would be used in the next step directly without further purification. MS [M+H]+: 433.2. Step 6: [2,3-difluoro-4-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl] trifluoromethanesulfonate To a solution of 2,3-difluoro-4-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl) pyrazol-4-yl]phenol (230.0 mg, 0.53 mmol) and pyridine (0.06 mL, 0.8 mmol) in DCM (10 mL) was added trifluoromethanesulfonic anhydride (0.11 mL, 0.64 mmol) in one portion at 0° C. Then reaction mixture was warmed to 20° C. and stirred for 2 h. This reaction was quenched by saturated aqueous Na2CO3(10 mL) and extracted by DCM (10 mL×2). The combined organic layers were dried over Na2SO4and concentrated to get [2,3-difluoro-4-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl] trifluoromethanesulfonate (320.0 mg). MS [M+H]+: 565.0. Step 7: 2-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane To a solution of [2,3-difluoro-4-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl) pyrazol-4-yl]phenyl] trifluoromethanesulfonate (320.0 mg, 0.57 mmol), bis(pinacolato)diboron (287.8 mg, 1.13 mmol) and potassium acetate (139.06 mg, 1.42 mmol) in 1,4-dioxane (10 mL) was added X-PHOS (27.0 mg, 0.06 mmol) and tris(dibenzylideneacetone)dipalladium (0) (51.9 mg, 0.06 mmol) in one portion under N2. This reaction mixture was stirred at 100° C. for 2 h. This reaction mixture was filtered, and the filtrate was concentrated to get the crude product. This crude product was purified by silica gel chromatography to get 2-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (250.0 mg, 0.46 mmol). MS [M+H]+: 543.3. The following examples were prepared in analogy to Intermediate G59 MSESIStartingEx #NameStructure[M + H]+MaterialInter- mediate G602-[[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5-(3- methoxyphenyl) pyrazol-1- yl]methoxy]ethyl- trimethyl-silane543.22-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1-yl] methoxy]ethyl- trimethyl- silane and (3- methoxyphenyl) boronic acidInter- mediate G612-[[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5-(2- methoxyphenyl) pyrazol-1- yl]methoxy]ethyl- trimethyl-silane543.32-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1- yl]methoxy] ethyl-trimethyl- silane and (2- methoxyphenyl) boronic acidInter- mediate G622-[[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5- thiazol-4-yl- pyrazol-1- yl]methoxy]ethyl- trimethyl-silane520.22-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1-yl] methoxy]ethyl- trimethyl- silane and tributyl(thiazol- 4-yl)stannaneInter- mediate G632-[[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5- tetrahydropyran-4- yl-pyrazol-1- yl]methoxy]ethyl- trimethyl-silane521.32-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1-yl] methoxy]ethyl- trimethyl- silane and Intermediate R9Inter- mediate G642-[[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5-(3,6- dihydro-2H-pyran- 4-yl)pyrazol-1- yl]methoxy]ethyl- trimethyl-silane519.32-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1-yl] methoxy]ethyl- trimethyl- silane and Intermediate R9Inter- mediate G652-[[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3-(2- methylpyrazol-3- yl)pyrazol-1- yl]methoxy]ethyl- trimethyl-silane517.72-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1- yl]methoxy] ethyl-trimethyl- silane and 1- methyl-5- (4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)pyrazoleInter- mediate G662-[[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3-(3- fluorophenyl) pyrazol-1- yl]methoxy]ethyl- trimethyl-silane531.42-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1- yl]methoxy] ethyl-trimethyl- silane and (3- fluorophenyl) boronic acidInter- mediate G67[2,3-difluoro-4-[3- (1-methylpyrazol- 4-yl)-1-(2- trimethylsilylethoxy- methyl)pyrazol-4- yl]phenyl]boronic acid435.22-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1- yl]methoxy] ethyl-trimethyl- silane and 1- methyl-4- (4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)pyrazoleInter- mediate G68[2,3-difluoro-4-[1- (2- trimethylsilylethoxy- methyl)-3-[1-(2- trimethylsilylethoxy- methyl)pyrazol-4- yl]pyrazol-4- yl]phenyl]boronic acid644.42-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1- yl]methoxy] ethyl-trimethyl- silane and trimethyl-[2- [[4-(4,4,5,5- tetramethyl- 1,3,2-dioxa- borolan-2- yl)pyrazol-1- yl]methoxy] ethyl]silaneInter- mediate G69[2,3-difluoro-4-[1- (2-methoxyethyl)- 3-phenyl-pyrazol- 4- yl]phenyl]boronic acid359.02-[[4-(4- benzyloxy- 2,3-difluoro- phenyl)-5- bromo- pyrazol-1- yl]methoxy] ethyl-trimethyl- silane and phenylboronic acid Intermediate G70 tert-butyl N-[6-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]-3-pyridyl]carbamate Step 1: 2-(4-bromo-3-methyl-1H-pyrazol-1-yl)-5-nitropyridine To a solution of 4-bromo-3-methyl-1H-pyrazole (2.5 g, 15.53 mmol) in DMF (100 mL) was added sodium hydride (750.0 mg, 18.75 mmol) at 0° C. portionwise under N2, after the addition was complete, the mixture was stirred at 0° C. for 0.5 h. Then 2-chloro-5-nitropyridine (2.5 g, 15.77 mmol) was added to the above mixture at 0° C. and the resultant mixture was warmed to 20° C. gradually and stirred for 11.5 h. Then the mixture was quenched with saturated NH4Cl (800 mL), filtered, the filter cake was washed with ACN (50 mL), dried under vacuum to afford 2-(4-bromo-3-methyl-pyrazol-1-yl)-5-nitro-pyridine (3.8 g). MS [M+H]+: 283.0. Step 2: 6-(4-bromo-3-methyl-1H-pyrazol-1-yl)pyridin-3-amine To a solution of 2-(4-bromo-3-methyl-pyrazol-1-yl)-5-nitro-pyridine (3.8 g, 13.42 mmol) and ammonium chloride (8.6 g, 161.09 mmol) in ethanol (70 mL) and water (20 mL) was added Fe powder (2.5 g, 44.29 mmol) at 20° C. under N2, the resultant mixture was stirred at 80° C. for 3 h under N2. TLC (PE:EA=1:1) indicated the desired product was formed. After the reaction mixture was cooled down to 20° C., the mixture was added saturated NaHCO3(60 mL), extracted with brine (150 mL) and EtOAc (120 mL×3). The combined organics were dried over Na2SO4, filtered, concentrated in vacuo to afford 6-(4-bromo-3-methyl-pyrazol-1-yl)pyridin-3-amine (3.2 g). Step 3: tert-butyl (6-(4-bromo-3-methyl-1H-pyrazol-1-yl)pyridin-3-yl)carbamate(4) To a solution of afford 6-(4-bromo-3-methyl-pyrazol-1-yl)pyridin-3-amine (3.2 g, 12.64 mmol) and Boc2O (5.5 g, 25.30 mmol) in methanol (50 mL) was added triethylamine (3.5 mL, 25.29 mmol), the resultant mixture was stirred at 20° C. for 14 h. Then the mixture was concentrated in vacuum to give a residue, which was purified by HPLC and evaporated under vacuum to afford tert-butyl N-[6-(4-bromo-3-methyl-pyrazol-1-yl)-3-pyridyl]carbamate (2.8 g). MS [M+H]+: 353.0. Step 4: tert-butyl (6-(4-(4-(benzyloxy)-2,3-difluorophenyl)-3-methyl-1H-pyrazol-1-yl)pyridin-3-yl)carbamate To a solution of tert-butyl N-[6-(4-bromo-3-methyl-pyrazol-1-yl)-3-pyridyl]carbamate (1.0 g, 2.83 mmol), 2-(4-benzyloxy-2,3-difluoro-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.5 g, 4.33 mmol) and potassium carbonate (800.0 mg, 5.79 mmol) in 1,4-dioxane (25 mL) and water (5.0 mL) was added Pd(dppf)Cl2(207.0 mg, 0.28 mmol) under N2at 20° C., then the mixture was stirred at 85° C. for 15 h under N2. After the mixture was cooled down to 20° C., the mixture was filtered through celite, the filtrate was extracted with brine (200 mL) and EtOAc (100 mL×3). The combined organics were dried over Na2SO4, filtered, concentrated in vacuum to give a residue, which was purified by HPLC and evaporated under vacuum to afford tert-butyl N-[6-[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]-3-pyridyl]carbamate (1.1 g) as yellow solid. MS [M+H]+: 493.1. Step 5: tert-butyl (6-(4-(2,3-difluoro-4-hydroxyphenyl)-3-methyl-1H-pyrazol-1-yl)pyridin-3-yl)carbamate To a solution of tert-butyl N-[6-[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]-3-pyridyl]carbamate (500.0 mg, 1.02 mmol) in methanol (15 mL) and ethyl acetate (15 mL) was added Pd/C (500.0 mg) at 20° C. under H2, the resultant mixture was stirred at 20° C. for 12 h under a balloon of H2. Then the mixture was filtered through celite, the filtrate was concentrated in vacuum to afford tert-butyl N-[6-[4-(2,3-difluoro-4-hydroxy-phenyl)-3-methyl-pyrazol-1-yl]-3-pyridyl]carbamate (325.0 mg). MS [M+H]+: 403.1. Step 6: 4-(1-(5-((tert-butoxycarbonyl)amino)pyridin-2-yl)-3-methyl-1H-pyrazol-4-yl)-2,3-difluorophenyl trifluoromethanesulfonate To a solution of tert-butyl N-[6-[4-(2,3-difluoro-4-hydroxy-phenyl)-3-methyl-pyrazol-1-yl]-3-pyridyl]carbamate (325.0 mg, 0.81 mmol) and pyridine (0.4 mL, 4.95 mmol) in DCM (15 mL) was added trifluoromethanesulfonic anhydride (290.0 mg, 1.03 mmol) at 20° C., the resultant mixture was stirred at 20° C. for 2 h. Then the mixture was extracted with saturated NH4Cl (70.0 mL) and EtOAc (60 mL×3). The combined organics were dried over Na2SO4, filtered, concentrated in vacuo to afford [4-[1-[5-(tert-butoxycarbonylamino)-2-pyridyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate (390.0 mg). MS [M+H]+: 534.9. Step 7: tert-butyl (6-(4-(2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3-methyl-1H-pyrazol-1-yl)pyridin-3-yl)carbamate To a solution of [4-[1-[5-(tert-butoxycarbonylamino)-2-pyridyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl] trifluoromethanesulfonate (300.0 mg, 0.56 mmol), bis(pinacolato)diboron (280.0 mg, 1.10 mmol) and potassium acetate (0.07 mL, 1.12 mmol) in 1,4-dioxane (8.0 mL) was added Pd(dppf)Cl2(42.0 mg, 0.06 mmol) at 20° C. under N2, the resultant mixture was stirred at 100° C. for 14 h under N2. After the mixture was cooled down, filtered through celite, the filtrate was concentrated in vacuum to give a residue, which was purified by prep-HPLC (FA) and concentrated under vacuum to afford tert-butyl N-[6-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]-3-pyridyl]carbamate (160.0 mg). MS [M+H]+: 513.2. The following examples were prepared in analogy to Intermediate G70. MSESIStartingEx #NameStructure[M + H]+MaterialInter- mediate G712-[[4-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3- methyl-pyrazol-1- yl]pyrazol-1- yl]methoxy]ethyl- trimethyl-silane517.14-bromo-3- methyl-1H- pyrazole and 2-[(4- iodopyrazol- 1-yl) methoxy]ethyl- trimethyl- silane; bis(pinacolato) diboronInter- mediate G72[4-[1-[[5-(tert- butoxycarbonyl- amino)-2- pyridyl]methyl]-3- methyl-pyrazol-4- yl]-2,3-difluoro- phenyl]boronic acid445.24-bromo-3- methyl-1H- pyrazole and tert-butyl N- [6- (bromomethyl)- 3-pyridyl] carbamate; bis(pinacolato) diboronInter- mediate G73[2,3-difluoro-4-[3- methyl-1-[2-(2- oxo-1- pyridyl)ethyl] pyrazol-4- yl]phenyl]boronic acid360.14-bromo-3- methyl-1H- pyrazole, piperidin-2- one and 1,2- dibromoethane; bis(pinacolato) diboronInter- mediate G742-[4-[2,3-difluoro- 4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3- methyl-pyrazol-1- yl]pyridine398.24-bromo-3- methyl-1H- pyrazole and 2- chloropyridine; bis(pinacolato) diboron Intermediate G75 2-[[4-[6-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl]-3-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane Step 1: 6-bromo-2-fluoropyridin-3-ol To a solution of 2-fluoropyridin-3-ol (2.0 g, 17.69 mmol) in acetic acid (20.0 mL) was added sodium acetate (1.3 mL, 17.69 mmol), after the solid was dissolved, bromine (2.8 g, 17.69 mmol) was added by drops at 0° C. The mixture was stirred at 20° C. for 3 h. 1 g of sodium sulfite was added to the reaction mixture, then the mixture was concentrated. The residue was diluted with 20 mL of water and neutralized with 2 M NaOH solution, the solid was filtered off and dried in vacuum to give 6-bromo-2-fluoro-pyridin-3-ol (1.8 g). MS [M+H]+: 191.9. Step 2: 3-(benzyloxy)-6-bromo-2-fluoropyridine To a solution of 6-bromo-2-fluoro-pyridin-3-ol (1.5 g, 7.81 mmol) in DMF (20.0 mL) was added NaH (375.0 mg, 9.38 mmol) at 0° C., the mixture was stirred at 0° C. for 0.5 h, then benzyl bromide (0.9 mL, 7.81 mmol) was added, the mixture was stirred at 20° C. for 16 h. The mixture was quenched with 30 mL of NH4Cl water solution at 0° C., then the mixture was extracted with ethyl acetate (30 mL×3), the organic layer was concentrated, the residue was purified by Prep-HPLC to give 3-benzyloxy-6-bromo-2-fluoro-pyridine (1.7 g). MS [M+H]+: 281.9. Step 3: 3-(benzyloxy)-2-fluoro-6-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)pyridine To a solution of 3-benzyloxy-6-bromo-2-fluoro-pyridine (1.5 g, 5.32 mmol) in 1,4-dioxane (60.0 mL) and water (6.0 mL) was added trimethyl-[2-[[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazol-1-yl]methoxy]ethyl]silane (3.8 g, 5.85 mmol), [1,1′-bis(diphenylphosphino) ferrocene]dichloropalladium(II) (389.1 mg, 0.53 mmol), K2CO3(1.1 g, 10.63 mmol) in glovebox, the mixture was stirred at 60° C. for 16 h under Ar2. The mixture was filtered off, the filtrate was concentrated, the residue was purified by silica gel column to give 2-[[4-(5-benzyloxy-6-fluoro-2-pyridyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (1.55 g) as a yellow solid. MS [M+H]+: 400.1. Step 4: 3-(benzyloxy)-6-(5-bromo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)-2-fluoropyridine To a solution of 2-[[4-(5-benzyloxy-6-fluoro-2-pyridyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (1.5 g, 3.75 mmol) in DMF (15 mL) was added NBS (1.34 g, 7.51 mmol), the mixture was stirred at 60° C. for 16 h. The mixture was purified by Prep-HPLC to give 2-[[4-(5-benzyloxy-6-fluoro-2-pyridyl)-5-bromo-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (800.0 mg). MS [M+H]+: 478.1. Step 5: 3-(benzyloxy)-2-fluoro-6-(5-(4-methoxyphenyl)-1-((2-(trimethylsilyl)ethoxy)-methyl)-1H-pyrazol-4-yl)pyridine To a solution of (4-methoxyphenyl)boronic acid (297.3 mg, 1.96 mmol) in 1,4-dioxane (1.0 mL) and water (0.1 mL) was added 2-[[4-(5-benzyloxy-6-fluoro-2-pyridyl)-5-bromo-pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (780.0 mg, 1.63 mmol), [1,1′-bis(diphenylphosphino) ferrocene]dichloropalladium(II) (119.3 mg, 0.16 mmol), K2CO3(518.4 mg, 4.89 mmol) in glovebox, the mixture was stirred at 60° C. for 4 h under Ar2. The mixture was filtered off, the filtrate was concentrated, the residue was purified by silica gel column to give 2-[[4-(5-benzyloxy-6-fluoro-2-pyridyl)-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (550.0 mg). MS [M+H]+: 506.2. Step 6: 2-fluoro-6-(5-(4-methoxyphenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)pyridin-3-ol A mixture of 2-[[4-(5-benzyloxy-6-fluoro-2-pyridyl)-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (550.0 mg, 1.09 mmol) in methanol (100 mL) was added Pd/C (50.0 mg) under N2, then the mixture flushed with H2and stirred with a H2balloon for 6 h at 15° C. The mixture was filtered off, the filtrate was concentrated to give 2-fluoro-6-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]pyridin-3-ol (450.0 mg). MS [M+H]+: 416.1. Step 7: tert-butyl 2-fluoro-6-(5-(4-methoxyphenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)pyridin-3-yl trifluoromethanesulfonate A mixture of 2-fluoro-6-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]pyridin-3-ol (450.0 mg, 1.08 mmol) in pyridine (15 mL) was added trifluoromethanesulfonic anhydride (611.1 mg, 2.17 mmol) at 0° C., then the mixture was stirred at 15° C. for 1 h. The mixture was diluted with 50 mL of ethyl acetate, washed with 30 mL of water and 30 mL of brine. The organic layer was dried over Na2SO4and concentrated to give [2-fluoro-6-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]-3-pyridyl]trifluoromethanesulfonate (580.0 mg). MS [M+H]+: 548.1. Step 8: 2-fluoro-6-(5-(4-methoxyphenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine To a solution of [2-fluoro-6-[5-(4-methoxyphenyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]-3-pyridyl] trifluoromethanesulfonate (560.0 mg, 1.02 mmol) in 1,4-dioxane (1.0 mL) was added bis(pinacolato)diboron (389.5 mg, 1.53 mmol), [1,1′-bis(diphenylphosphino)ferrocene]-dichloropalladium(II) (74.8 mg, 0.10 mmol) and potassium acetate (300.9 mg, 3.07 mmol) in glovebox, the mixture was stirred at 90° C. for 16 h. The mixture was filtered off, the filtrate was concentrated, the residue was purified by Prep-HPLC to give 2-[[4-[6-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl]-5-(4-methoxyphenyl)pyrazol-1-yl]methoxy]ethyl-trimethyl-silane (530.0 mg). MS [M+H]+: 526.3. Intermediate G76 2-[2-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]ethyl]-6-methoxy-pyridine Step 1: 2-methoxy-6-vinylpyridine A mixture of 2-bromo-6-methoxy-pyridine (6.5 mL, 53.19 mmol) and potassium; trifluoro(vinyl)boranuide (8.6 g, 63.82 mmol) in 1,4-dioxane (120 mL) was added [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (0.8 g, 1.06 mmol), Potassium carbonate (20.6 g, 149.31 mmol) and Water (12 mL) in glove box. The mixture was stirred at 100° C. for 16 h under Ar. The mixture was distilled in vacuum, the collector was cooled with dry ice, the distilment was added 20 mL of HCl/1,4-dioxane (4 M), stirred at 60° C. for 2 h, then the mixture was concentrated to give 2-methoxy-6-vinyl-pyridine; hydrochloride (2.6 g) as a yellow gum. MS [M+H]+: 136.1. Step 2: 2-(2-(4-bromo-3-methyl-1H-pyrazol-1-yl)ethyl)-6-methoxypyridine A mixture of 2-methoxy-6-vinyl-pyridine; hydrochloride (2.5 g, 18.5 mmol) in DMSO (20.0 mL) was added Potassium carbonate (5.1 g, 36.99 mmol) and 4-bromo-3-methylpyrazole (3.6 g, 22.20 mmol) the mixture was stirred at 80° C. for 12 h. The mixture was filtered off, the filtrate was purified by Prep-HPLC (TFA as additive) to give 2-[2-(4-bromo-3-methyl-pyrazol-1-yl) ethyl]-6-methoxy-pyridine (2.0 g) as a yellow oil. MS [M+H]+: 296.0. Step 3: 2-(2-(4-(4-(benzyloxy)-2,3-difluorophenyl)-3-methyl-1H-pyrazol-1-yl)ethyl)-6-methoxypyridine To a solution of 2-[2-(4-bromo-3-methyl-pyrazol-1-yl) ethyl]-6-methoxy-pyridine (2.2 g, 6.36 mmol), Pd(dppf)2Cl2(0.9 g, 0.70 mmol), 2-(4-benzyloxy-2,3-difluoro-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.8 g, 6.08 mmol) and K2CO3(1.8 g, 13.02 mmol) in 1,4-dioxane (10 mL) and water (3 mL) was stirred at 80° C. for 12 h. The reaction mixture was dried by vacuum, the residue was purified by column and dried by vacuum to obtain 2-[2-[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]ethyl]-6-methoxy-pyridine (1.7 g, 3.81 mmol, 62.6% yield) as off-white solid. MS [M+H]+: 435.9. Step 4: 2,3-difluoro-4-(1-(2-(6-methoxypyridin-2-yl)ethyl)-3-methyl-1H-pyrazol-4-yl)phenol A solution of 2-[2-[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]ethyl]-6-methoxy-pyridine (2.1 g, 4.82 mmol) and Pd/C (200.0 mg) in methanol (0.2 mL) was stirred at 25° C. for 2 h. It was filtered and dried by vacuum to get 2,3-difluoro-4-[1-[2-(6-methoxy-2-pyridyl)ethyl]-3-methyl-pyrazol-4-yl]phenol (560.0 mg) as off-white solid. MS [M+H]+: 345.8. Step 5: 2,3-difluoro-4-(1-(2-(6-methoxypyridin-2-yl)ethyl)-3-methyl-1H-pyrazol-4-yl)phenyl trifluoromethanesulfonate A mixture of 2,3-difluoro-4-[1-[2-(6-methoxy-2-pyridyl)ethyl]-3-methyl-pyrazol-4-yl]phenol (500.0 mg, 1.45 mmol) and pyridine (0.5 mL) in DCM (2.0 mL) was added trifluoromethanesulfonic anhydride (408.5 mg, 1.45 mmol) at 0° C., it was stirred at 0° C. for 2 h. The reaction mixture was poured into ice-water (50 mL), extracted by EA (50 mL), and dried by Na2SO4, the organic phase was concentrated under reduced pressure to obtain [2,3-difluoro-4-[1-[2-(6-methoxy-2-pyridyl)ethyl]-3-methyl-pyrazol-4-yl]phenyl] trifluoromethanesulfonate (803.0 mg, 1.68 mmol, 116.2% yield) as light yellow gum. MS [M+H]+: 478.1. Step 6: 2-(2-(4-(2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3-methyl-1H-pyrazol-1-yl)ethyl)-6-methoxypyridine A mixture of [2,3-difluoro-4-[1-[2-(6-methoxy-2-pyridyl)ethyl]-3-methyl-pyrazol-4-yl]phenyl]trifluoromethanesulfonate (600.0 mg, 2.36 mmol), bis(pinacolato)diboron (800.0 mg, 1.68 mmol), Pd(dppf)Cl2(200.0 mg, 0.25 mmol) and AcOK (400.0 mg, 4.08 mmol) in 1,4-dioxane (0.3 mL) was stirred at 80° C. for 2 h. The reaction mixture was Prep-HPLC (FA) to obtain 2-[2-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]ethyl]-6-methoxy-pyridine (402.0 mg). MS [M+H]+: 373.9. Intermediate G78 2-[[4-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane Step 1: 4-bromo-3-methyl-1-prop-2-ynyl-pyrazole A mixture of propargyl bromide (11.1 g, 93.17 mmol) in NMP (100.0 mL) was added 4-bromo-3-methyl-1H-pyrazole (10.0 g, 62.11 mmol) and potassium carbonate (17.2 g, 124.22 mmol), The mixture was stirred at 100° C. for 16 h. This reaction mixture was poured into water (300 mL) and EtOAc (200 mL) and was washed by brine (200 mL×2). The organic layer was dried over Na2SO4and concentrated to give crude product which was further purified by prep-HPLC (FA) to get the mixture of 4-bromo-3-methyl-1-prop-2-ynyl-pyrazole (2) (8.0 g). MS [M+H]+: 198.7. Step 2: 4-[(4-bromo-3-methyl-pyrazol-1-yl)methyl]-1H-triazole A mixture of 4-bromo-3-methyl-1-prop-2-ynyl-pyrazole (5.0 g, 25.12 mmol) in azidotrimethylsilane (33.3 mL, 251.19 mmol) was stirred at 100° C. for 16 h. This reaction mixture was diluted with EtOAc (100 mL) and was washed by saturated aqueous Na2CO3(200 mL×2). The organic layer was dried over Na2SO4and concentrated to give crude product which was further purified by prep-HPLC (FA) to give a mixture of isomers which was further purified by chiral separation to get 4-[(4-bromo-3-methyl-pyrazol-1-yl)methyl]-1H-triazole (1.7 g). MS [M+H]+: 242.0. Step 3: 2-[[4-[(4-bromo-3-methyl-pyrazol-1-yl)methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane To a solution of 4-[(4-bromo-3-methyl-pyrazol-1-yl)methyl]-1H-triazole (1.6 g, 6.61 mmol) in DMF (20 mL) was added NaH (396.6 mg, 9.91 mmol) and 2-(trimethylsilyl) ethoxymethyl chloride (1.2 mL, 6.94 mmol) at 0° C., The mixture was stirred at 25° C. for 12 h under N2. The reaction mixture was quenched by (30 mL) NH4Cl (aq), extracted with EtOAc (80 mL×3), the organic phase was washed with brine (50 mL×2), dried over anhydrous Na2SO4and concentrated in vacuum to give 2-[[4-[(4-bromo-3-methyl-pyrazol-1-yl)methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane (2200.0 mg). MS [M+H]+: 372.0. Step 4: 2-[[4-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane A mixture of 2-[[4-[(4-bromo-3-methyl-pyrazol-1-yl)methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane (1300.0 mg, 3.49 mmol), 2-(4-benzyloxy-2,3-difluoro-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1208.7 mg, 3.49 mmol), Na2CO3(740.1 mg, 6.98 mmol) and Pd(dppf)Cl2(307.6 mg, 0.35 mmol) in a flask. The flask was degassed and purged with N2gas for four times. 1,4-dioxane (15.0 mL) and water (2.0 mL) was added by injector to the mixture. The mixture was stirred at 80° C. for 2 h under N2. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to remove the solvent, then the product was purified by chromatography column flash (petroleum/EtOAc=20/1˜1/1) and concentrated to give 2-[[4-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane (1.7 g). MS [M+H]+: 512.2. Step 5: 2,3-difluoro-4-[3-methyl-1-[[1-(2-trimethylsilylethoxymethyl)triazol-4-yl]methyl]pyrazol-4-yl]phenol To a solution of 2-[[4-[[4-(4-benzyloxy-2,3-difluoro-phenyl)-3-methyl-pyrazol-1-yl]methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane (1.65 g, 3.22 mmol) in methanol (20.0 mL) was added Pd/C (300.0 mg) under N2. The suspension was degassed under vacuum and purged with H2several times at 20° C. for 2 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give 2,3-difluoro-4-[3-methyl-1-[[1-(2-trimethylsilylethoxymethyl)triazol-4-yl]methyl]pyrazol-4-yl]phenol (1.35 g). MS [M+H]+: 422.1. Step 6: [2,3-difluoro-4-[3-methyl-1-[[1-(2-trimethylsilylethoxymethyl)triazol-4-yl]methyl]pyrazol-4-yl]phenyl] trifluoromethanesulfonate A solution of 2,3-difluoro-4-[3-methyl-1-[[1-(2-trimethylsilylethoxymethyl)triazol-4-yl]methyl]pyrazol-4-yl]phenol (1.3 g, 3.08 mmol) in pyridine (15.0 mL) was added trifluoromethanesulfonic anhydride (1.0 mL, 6.17 mmol) under 0° C., The reaction was stirred at 20° C. for 2 h. The mixture was poured into ice water (20 mL) and extracted with EtOAc (50 mL×2). The mixture was combined and washed with brine (20 mL). The organic layer was dried and concentrated under vacuum to give [2,3-difluoro-4-[3-methyl-1-[[1-(2-trimethylsilylethoxymethyl)triazol-4-yl]methyl]pyrazol-4-yl]phenyl] trifluoromethanesulfonate (1700.0 mg). MS [M+H]+: 554.0. Step 7: 2-[[4-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane A mixture of 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (908.3 mg, 3.58 mmol), Pd(dppf)Cl2(243.2 mg, 0.30 mmol), [2,3-difluoro-4-[3-methyl-1-[[1-(2-trimethylsilylethoxymethyl)triazol-4-yl]methyl]pyrazol-4-yl]phenyl]trifluoromethanesulfonate (1650.0 mg, 2.98 mmol) and potassium acetate (585.1 mg, 5.96 mmol) in a flask. 1,4-dioxane (20 mL) was added by injector to the mixture. The flask was degassed and purged with N2gas for four times. The mixture was stirred at 80° C. for 2 h under N2atmosphere. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to remove the solvent, then the product was purified by reversed-phase chromatography (FA as additive) to give 2-[[4-[[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]methyl]triazol-1-yl]methoxy]ethyl-trimethyl-silane (800.0 mg). MS [M+H]+: 532.3. Intermediate G79 [2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-3-pyridyl]boronic acid Step 1: 6-bromo-2-fluoropyridin-3-ol To a solution of 2-fluoropyridin-3-ol (2.5 g, 22.11 mmol) in acetic acid (25 mL) was added sodium acetate (1.67 mL, 22.11 mmol), after the solid was dissolved, bromine (3.5 g, 22.11 mmol) was added by drops at 0° C., then the mixture was stirred at 20° C. for 3 h. The reaction mixture was poured into ice-water, it was extracted by EtOAc (2×200 mL). The reaction mixture was washed by 100 mL solution of sodium sulfite in water, and then organic layer was dried in vacuum to give 6-bromo-2-fluoro-pyridin-3-ol (3.15 g). MS [M+H]+: 191.6. Step 2: 3-(benzyloxy)-6-bromo-2-fluoropyridine A solution of 6-bromo-2-fluoro-pyridin-3-ol (3.1 g, 16.15 mmol, 1.0 eq), benzyl bromide (1.95 mL, 16.37 mmol, 1.0 eq) and K2CO3(3.1 g, 22.43 mmol, 1.39 eq) in ACN (30 mL) was stirred at 25° C. for 12 h. The reaction mixture was filtered and washed by EtOAc (100 mL), then the filter was washed by brine (2×200 mL), it was dried by vacuum to obtain 3-benzyloxy-6-bromo-2-fluoro-pyridine (4.1 g). MS [M+H]+: 281.6. Step 3: 3-(benzyloxy)-2-fluoro-6-(1-(2-methoxyethyl)-5-methyl-1H-pyrazol-4-yl)pyridine To a solution of 3-benzyloxy-6-bromo-2-fluoro-pyridine (1.0 g, 3.54 mmol), 1-(2-methoxyethyl)-5-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (1.0 g, 3.76 mmol) in 1,4-dioxane (10 mL) and water (3 mL) was added Pd(dppf)Cl2(300.0 mg, 0.37 mmol) and Na2CO3(0.8 g, 7.55 mmol). The reaction mixture was stirred at 80° C. for 2 h. The reaction mixture was purified by Pre-HPLC (FA as additive) and dried by vacuum to obtain 3-benzyloxy-2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]pyridine (340 mg). MS [M+H]+: 342.1. Step 4: 2-fluoro-6-(1-(2-methoxyethyl)-5-methyl-1H-pyrazol-4-yl)pyridin-3-ol A solution of 3-benzyloxy-2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]pyridine (320.0 mg, 0.94 mmol) and Pd/C (100.0 mg) in methanol (0.5 mL) was stirred at 25° C. for 12 h under H2balloon. The reaction mixture was filtered and it was dried by vacuum to obtain 2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]pyridin-3-ol (236.0 mg). MS [M+H]+: 251.8. Step 5: 2-fluoro-6-(1-(2-methoxyethyl)-5-methyl-1H-pyrazol-4-yl)pyridin-3-yl trifluoromethanesulfonate A mixture of 2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]pyridin-3-ol (300.0 mg, 1.19 mmol) in pyridine (1.0 mL) was added trifluoromethanesulfonic anhydride (336.8 mg, 1.19 mmol) at 0° C., The reaction mixture was stirred at 0° C. for 2 h. This reaction mixture was poured into ice-water (50 mL), it was extracted by EA (50 mL), organic layer was dried over Na2SO4, filtered and then evaporated to give [2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-3-pyridyl]trifluoromethanesulfonate (320.0 mg). MS [M+H]+: 383.8. Step 6: [2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-3-pyridyl]boronic acid A mixture of 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (260.0 mg, 1.02 mmol), [2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-3-pyridyl]trifluoromethanesulfonate (320.0 mg, 0.83 mmol), Pd(dppf)Cl2(96.0 mg, 0.12 mmol) and KOAc (200.0 mg, 2.06 mmol) in 1,4-dioxane (5 mL) was stirred at 80° C. for 2 h. The reaction mixture was purified by Pre-HPLC (FA) and dried by vacuum to obtain [2-fluoro-6-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-3-pyridyl]boronic acid (126.0 mg). MS [M+H]+: 279.8. The following examples were prepared in analogy to Intermediate G79. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate G812-[[4-[6-fluoro-5- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2-yl)- 2-pyridyl]pyrazol- 1- yl]methoxy]ethyl- trimethyl-silane338.22- fluoropyridin- 3-ol and trimethyl-[2- [[4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)pyrazol- 1- yl]methoxy] ethyl]silane;bis(pinacolato)diboronIntermediate G912-fluoro-6-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol- 4-yl]-3-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)pyridine293.82- fluoropyridin- 3-ol and 1-(2- methoxyethyl)- 3,5- dimethyl-4- (4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)pyrazole; bis(pinacolato) diboron Intermediate G86 Step 1: (6-bromo-3-pyridyl)-[3-[tert-butyl(dimethyl)silyl]oxypropyl]amine (6-bromo-3-pyridyl)amine (2.45 g, 14.16 mmol), 3-[tert-butyl(dimethyl)silyl]oxypropion-aldehyde (2.67 g, 14.16 mmol) and acetic acid (170.07 mg, 2.83 mmol) were dissolved in dichloromethane (50 mL). To this solution was added sodium triacetoxyborohydride (3.6 g, 16.99 mmol) portionwise. The mixture was stirred at rt for 1 h after addition. The mixture was poured into 100 mL water and extracted with DCM (50 mL×2). The extracts were combined, washed with brine and dried over sodium sulfate. The solvent was removed in vacuum and the residue was purified by flash chromatography (silica gel; EtOAc:PE=0:1 to 1:1). To afford (6-bromo-3-pyridyl)-[3-[tert-butyl(dimethyl)silyl]oxypropyl]amine (3 g). MS [M+H]+: 345.6. Step 2: N-(6-bromo-3-pyridyl)-N-(3-hydroxypropyl)carbamic acid 9H-fluoren-9-ylmethyl ester (6-bromo-3-pyridyl)-[3-[tert-butyl(dimethyl)silyl]oxypropyl]amine (3.00 g, 8.69 mmol) was dissolved in 5 mL toluene and this solution was added dropwise to a solution of chlorocarbonic acid 9H-fluoren-9-ylmethyl ester (2.25 g, 8.69 mmol) in anhydrous toluene, extra dry (20 mL) at 0° C. After addition, the mixture was stirred at 0° C. for 1 h and then at rt for another 1 h. yellow precipitate formed. The mixture was left overnight. The solvent was removed in vacuum, and the residue was purified by flash chromatography (silica gel; EtOAc:PE=0:1 to 1:0). To afford N-(6-bromo-3-pyridyl)-N-(3-hydroxypropyl)carbamic acid 9H-fluoren-9-ylmethyl ester (2 g). MS [M+H]+: 453.1. Step 3: 3-[(6-bromo-3-pyridyl)-(9H-fluoren-9-ylmethoxycarbonyl)amino]propionic acid iodobenzene diacetate (703.41 mg, 2.18 mmol), TEMPO (62.44 mg, 0.397 mmol) and N-(6-bromo-3-pyridyl)-N-(3-hydroxypropyl)carbamic acid 9H-fluoren-9-ylmethyl ester (900 mg, 1.99 mmol) were combined in a reaction vessel, and to this mixture was added acetonitrile (10.99 mL) and water (5.99 mL). The reaction mixtures were stirred for 3 h before another batch of iodobenzene diacetate (703.41 mg, 2.18 mmol) was added. The stirring was continued 18 h. The solvent was removed in vacuo and the residue was purified by flash chromatography (silica gel; EtOAc:PE=0:1 to 1:0), to afford 3-[(6-bromo-3-pyridyl)-(9H-fluoren-9-ylmethoxycarbonyl)amino]propionic acid (778 mg). MS [M+H]+: 467.1. Step 4: N-(3-amino-3-keto-propyl)-N-(6-bromo-3-pyridyl)carbamic acid 9H-fluoren-9-ylmethyl ester 3-[(6-bromo-3-pyridyl)-(9H-fluoren-9-ylmethoxycarbonyl)amino]propionic acid (770 mg, 1.65 mmol), ammonium chloride (176.27 mg, 3.3 mmol) and DIEA (1.06 g, 8.24 mmol) were stirred in N,N-dimethylacetamide (27.11 mL) for 1 min. HATU (751.82 mg, 1.98 mmol) was added to the mixture, and the resulting solution was stirred at 25° C. for 1 h. The mixture was poured into 100 mL water and extracted with EtOAc (50 mL×3). The extracts were combined, washed with 50 mL brine, dried over sodium sulfate and concentrated in vacuum. The residue was purified by flash chromatography (silica gel; EtOAc:PE=0:1 to 1:0), to afford N-(3-amino-3-keto-propyl)-N-(6-bromo-3-pyridyl)carbamic acid 9H-fluoren-9-ylmethyl ester (725 mg). MS [M+H]+: 466.2. Step 5: N-(3-amino-3-keto-propyl)-N-[6-(2,3-difluoro-4-hydroxy-phenyl)-3-pyridyl]carbamic acid 9H-fluoren-9-ylmethyl ester In a microwave tube were placed N-(3-amino-3-keto-propyl)-N-(6-bromo-3-pyridyl)carbamic acid 9H-fluoren-9-ylmethyl ester (400 mg, 0.858 mmol), palladiumtetrakis (99.12 mg, 0.086 mmol), Na2CO3(272.74 mg, 2.57 mmol) and (2,3-difluoro-4-hydroxy-phenyl)boronic acid (223.76 mg, 1.29 mmol) in water (0.351 mL) and 1,4-dioxane (3.51 mL). The vial was sealed with a rubber septum, evacuated and backfilled with nitrogen for 5 times. The mixture was then heated at 100° C. for 0.5 h. The mixture was cooled to rt, 100-200 mesh silica gel was added to absorb the material. The loaded sample was purified by flash chromatography (silica gel; MeOH:DCM=0:1 to 1:10), to afford N-(3-amino-3-keto-propyl)-N-[6-(2,3-difluoro-4-hydroxy-phenyl)-3-pyridyl]carbamic acid 9H-fluoren-9-ylmethyl ester (166 mg). MS [M+H]+: 516.8. Step 6: trifluoromethanesulfonic acid [4-[5-[(3-amino-3-keto-propyl)-(9H-fluoren-9-ylmethoxycarbonyl)amino]-2-pyridyl]-2,3-difluoro-phenyl] ester N-(3-amino-3-keto-propyl)-N-[6-(2,3-difluoro-4-hydroxy-phenyl)-3-pyridyl]carbamic acid 9H-fluoren-9-ylmethyl ester (165 mg, 0.320 mmol) and pyridine (75.95 mg, 0.960 mmol) were dissolved in anhydrous dichloromethane, extra dry (3.3 mL). Tf2O (108.37 mg, 0.384 mmol) was added dropwise at 0° C. The solution was stirred at the same temperature for 1 h. 100-200 mesh silica gel was added to absorb the material. The loaded sample was purified by flash chromatography (silica gel; EtOAc:PE=0:1 to 1:0), to afford trifluoromethanesulfonic acid [4-[5-[(3-amino-3-keto-propyl)-(9H-fluoren-9-ylmethoxycarbonyl)amino]-2-pyridyl]-2,3-difluoro-phenyl] ester (168 mg). MS [M+H]+: 648.3. Step 7: N-(3-amino-3-keto-propyl)-N-[6-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-pyridyl]carbamic acid 9H-fluoren-9-ylmethyl ester trifluoromethanesulfonic acid [4-[5-[(3-amino-3-keto-propyl)-(9H-fluoren-9-ylmethoxycarbonyl)amino]-2-pyridyl]-2,3-difluoro-phenyl] ester (165 mg, 0.255 mmol), 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (21.06 mg, 0.025 mmol),bis(pinacolato)diboron (77.64 mg, 0.306 mmol) and potassium acetate (75.02 mg, 0.764 mmol) were placed in 1,4-dioxane, extra dry (3.61 mL) in a microwave tube. The tube was sealed with a rubber septum, evacuated and backfilled with nitrogen for 5 times. The mixture was heated at 100° C. for 1 h. The mixture was cooled to rt and absorbed to 100-200 mesh silica gel. The loaded sample was purified by flash chromatography (silica gel; MeOH:DCM=0:1 to 1:10), to afford N-(3-amino-3-keto-propyl)-N-[6-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-pyridyl]carbamic acid 9H-fluoren-9-ylmethyl ester (90 mg). MS [M+H]+: 648.3. The following examples were prepared in analogy to Intermediate G86. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate G879H-fluoren-9- ylmethyl N-[6-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-5- methyl-3- pyridyl]carbamate569.16-bromo-5- methyl- pyridin-3- amine and 9H- fluoren-9- ylmethyl carbonochloridate; (2,3- difluoro-4- hydroxy- phenyl)boronic acid; bis(pinacolato) diboronIntermediate G889H-fluoren-9- ylmethyl N-[6-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3- pyridyl]carbamate555.26- bromopyridin- 3-amine and 9H-fluoren-9- ylmethyl carbonochloridate; (2,3- difluoro-4- hydroxy- phenyl)boronic acid; bis(pinacolato) diboron Intermediate H1 N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide Step 1: tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate To a 25 mL microwave vial was added tert-butyl 4-[4-[4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (180 mg, 282 μmol), 4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-(2-methoxyethyl)-3-methyl-pyrazole (160 mg, 423 μmol), 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride (18.4 mg, 28.2 μmol) and Na2CO3(89.7 mg, 846 μmol) in 1,4-Dioxane (15 mL)/Water (1.5 mL). The vial was capped and heated in the microwave at 100° C. for 3 h under N2. The reaction mixture was filtered through glass fiber paper. The filtrate was concentrated in vacuum. The crude material was purified by flash chromatography to afford tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (150 mg). MS [M+H]+: 809.5. Step 2: N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide In a 50 mL round-bottomed flask, tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (150 mg, 185 μmol) was combined with THE (2 mL) to give a light brown solution. HCl water solution (1.24 mL, 14.8 mmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (131 mg). MS [M+H]+: 709.3. The following intermediates were prepared in analogy to Intermediate H1. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate H2N-[3-chloro-4-[4- (piperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide709.3Intermediate D1 and Intermediate G3; HClIntermediate H3N-[3-chloro-4-[4- (piperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-(2,2- difluoroethyl)-3- methyl-pyrazol-4-yl]- 2,3-difluoro-phenyl]- 1-methyl-imidazole-2- carboxamide715.8Intermediate D1 and Intermediate G5; HClIntermediate H4N-[3-chloro-4-[4- (piperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide723.7Intermediate D1 and Intermediate G6; HClIntermediate H5N-[3-chloro-4-[4- (piperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide687.3Intermediate D1 and Intermediate G7; HClIntermediate H6N-[3-chloro-4-[4- (piperazine-1- carbonyl)piperidine-1- carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide709.6Intermediate D7 and Intermediate G3; HClIntermediate H75-[2-chloro-3-fluoro- 4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-N-[3- chloro-4-(piperazine- 1-carbonyl)phenyl]-1- methyl-imidazole-2- carboxamide613.2Intermediate A1 and Intermediate G11; HClIntermediate H8N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide597.3Intermediate A1 and Intermediate G3; HClIntermediate H9N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]-5- [3-fluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4-yl]- 2-methyl-phenyl]-1- methyl-imidazole-2- carboxamide593.3Intermediate A1 and Intermediate G13; HClIntermediate H105-[5-chloro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4-yl]- 2-methyl-phenyl]-N- [3-chloro-4-[4- (piperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide721.7Intermediate D1 and Intermediate G92; HClIntermediate H115-[3-chloro-2-fluoro- 4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-N-[3- chloro-4-(piperazine- 1-carbonyl)phenyl]-1- methyl-imidazole-2- carboxamide613.2Intermediate A1 and Intermediate G93; HCl Intermediate I1 N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide Step 1: tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate In a 100 mL round-bottomed flask, tert-butyl 4-[4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]piperazine-1-carboxylate (150 mg, 285 μmol), 4-(2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(2-methoxyethyl)-5-methyl-1H-pyrazole (151 mg, 399 μmol), 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride (18.6 mg, 28.5 μmol) and Na2CO3(90.5 mg, 854 μmol) were combined with dioxane (10 mL)/Water (1 mL) to give a dark red solution. The reaction mixture was heated to 100° C. and stirred for 15 h under N2. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (160 mg). MS [M+H]+: 698.2. Step 2: N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide In a 50 mL round-bottomed flask, tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (160 mg, 229 μmol) was combined with THE (2 mL) to give a light brown solution. HCl water solution (1.15 ml, 13.8 mmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step, To afford N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (137 mg). MS [M+H]+: 598.1. The following intermediates were prepared in analogy to Intermediate I1. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate I2N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide598.1Intermediate B1 and Intermediate G2; HClIntermediate I3N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide612.2Intermediate B1 and Intermediate G6; HClIntermediate I45-[2,3-difluoro-4-[1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl-N- [3-methyl-4- (piperazine-1- carbonyl)phenyl]imidazole- 2-carboxamide578.3Intermediate B2 and Intermediate G2; HClIntermediate I5N-[4-(4- aminopiperidine-1- carbonyl)-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)- 3-methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide612.3Intermediate B3 and Intermediate G2; HClIntermediate I61-[2-chloro-4-[[5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperidine-4- carboxylic acid641.2Intermediate B4 and Intermediate G3; TFAIntermediate I7N-[3-chloro-4-(4- piperidylmethylcarbamoyl) phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide626.4Intermediate B5 and Intermediate G3; HClIntermediate I8N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]-5- [2,3-difluoro-4-[1-(3- methoxypropyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide612.3Intermediate B1 and Intermediate G17; HClIntermediate I9N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]-5- [2,3-difluoro-4-[1-(3- methoxypropyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide612.3Intermediate B1 and Intermediate G18; HClIntermediate I10N-[4-(4- aminopiperidine-1- carbonyl)-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)- 5-methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide612.3Intermediate B3 and Intermediate G3; HClIntermediate I11N-[4-[[(1R,5S)-3- azabicyclo[3.1.0]hexan- 6-yl]carbamoyl]-3- chloro-phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide624.0Intermediate B6 and Intermediate G6; HClIntermediate I12N-[4-[[(1S,5R)-3- azabicyclo[3.1.0]hexan- 6-yl]carbamoyl]-3- chloro-phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide610.0Intermediate B6 and Intermediate G3; HCl Intermediate J1 N-[3-chloro-4-[4-[2-(dimethylamino)acetyl]piperazine-1-carbonyl]phenyl]-1-methyl-5-[4-[3-methyl-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]imidazole-2-carboxamide Under N2protection, a mixture of trimethyl-[2-[[3-methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]pyrazol-1-yl]methoxy]ethyl]silane (323.9 mg, 0.782 mmol), 5-bromo-N-[3-chloro-4-[4-[2-(dimethylamino)acetyl]piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide (400 mg, 782 μmol), Na2CO3(249 mg, 2.34 mmol) and 1,1-bis(di-tert-butylphosphino)ferrocene palladium dichloride (25.5 mg, 39.1 μmol) in 1,4-dioxane (7 mL) and water (0.7 mL) was heated at 95° C. for 15 h. Then the mixture was filtered and concentrated in vacuum. The crude product was purified by flash column to afford N-[3-chloro-4-[4-[2-(dimethylamino)acetyl]piperazine-1-carbonyl]phenyl]-1-methyl-5-[4-[3-methyl-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]imidazole-2-carboxamide (50 mg) as a yellow solid. MS [M+H]+: 719.2. The following examples were prepared in analogy to Intermediate J1. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate J2N-[3-chloro-4-[4- [2- (dimethylamino) acetyl]piperazine-1- carbonyl]phenyl]- 5-[4-[3,5-dimethyl- 1-(2- trimethylsilylethoxy methyl)pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide733.5Intermediate D3 and Intermediate G9 Intermediate K1 N-[3-chloro-4-[[4-(2,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]l-methyl-imidazole-2-carboxamide Step 1: tert-butyl (2S,4R)-2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-4-hydroxy-pyrrolidine-1-carboxylate At room temperature, a mixture of N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (350 mg, 585 μmol), (2S,4R)-1-tert-butoxycarbonyl-4-hydroxy-pyrrolidine-2-carboxylic acid (176 mg, 761 μmol), HATU (334 mg, 878 μmol) and DIPEA (227 mg, 1.76 mmol) in DMF (5 mL) was stirred for 16 h. Then the mixture was poured into water. The water layer was extracted with DCM. The combined organic layers were washed with water and concentrated in vacuum. The residue was purified by flash column to afford tert-butyl (2S,4R)-2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-4-hydroxy-pyrrolidine-1-carboxylate (310 mg) as a brown solid. MS [M+H]+: 811.2. Step 2: N-[3-chloro-4-[4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide At room temperature, a solution of tert-butyl (2S,4R)-2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-4-hydroxy-pyrrolidine-1-carboxylate (310 mg, 382 μmol) in DCM (10 mL) and TFA (5 mL) was stirred for 1 h. Then the mixture was concentrated in vacuum. The residue was basified by NH3·H2O to PH 8-9. The water layer was extracted with DCM. The organic layer was dried over anhydrous Na2SO4and concentrated in vacuum to afford the crude product N-[3-chloro-4-[4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (240 mg) as a brown oil. MS [M+H]+: 711.3. The following examples were prepared in analogy to Intermediate K1. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate K2N-[3-chloro-4-[4- (4- hydroxypiperidine- 4- carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro-4- [1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide725.2Intermediate I1 and 1-(tert- butoxycarbonyl)- 4- hydroxypiperidine- 4- carboxylic acid and HClIntermediate K3N-[3-chloro-4-[4- (3- hydroxypiperidine- 4- carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro-4- [1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide725.2Intermediate I1 and 1-(tert- butoxycarbonyl)- 3- hydroxypiperidine- 4- carboxylic acid and HClIntermediate K4N-[3-chloro-4-[4- [2- (dimethylamino) acetyl]piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro-4- [1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide683.4Intermediate I1 and dimethylglycineIntermediate K5N-[3-chloro-4-[[1- [2- (dimethylamino) acetyl]-4- piperidyl] methylcarbamoyl] phenyl]-5- [2,3-difluoro-4-[1- (2-methoxyethyl)- 5-methyl-pyrazol- 4-yl]phenyl]-1- methyl-imidazole- 2-carboxamide711.3Intermediate I7 and dimethylglycineIntermediate K6N-[3-chloro-4-[4- [(2S,3S)-3- hydroxypyrrolidine- 2- carbonyl]piperazine- 1- carbonyl]phenyl]- 5-[2,3-difluoro-4- [1-(2- methoxyethyl)-3,5- dimethyl-pyrazol- 4-yl]phenyl]-1- methyl-imidazole- 2-carboxamide725.2Intermediate I3 and rac- (2S,3S)-1- tert- butoxycarbonyl- 3-hydroxy- pyrrolidine-2- carboxylic acid and HClIntermediate K7N-[3-chloro-4-[4- (4- hydroxypiperidine- 4- carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro-4- [1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide725.2Intermediate I2 and 1-(tert- butoxy carbonyl)-4- hydroxy piperidine-4- carboxylic acid and HClIntermediate K8N-[3-chloro-4-[4- [(2S)-4- (hydroxymethyl) pyrrolidine-2- carbonyl] piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro-4- [1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide725.2Intermediate I2 and Intermediate R6 and HClIntermediate K9N-[3-chloro-4-[4- (piperidine-4- carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro-4- (3-methyl-1H- pyrazol-4- yl)phenyl]-1- methyl-imidazole- 2-carboxamide651.2Intermediate M1 and 1- tert- butoxycarbonyl piperidine- 4-carboxylic acid; HClIntermediate K10N-[3-chloro-4-[4- [(2S,4R)-4- hydroxypyrrolidine- 2-carbonyl] piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro-4- (3-methyl-1H- pyrazol-4- yl)phenyl]-1- methyl-imidazole- 2-carboxamide653.2Intermediate M1 and intermediate R3; HClIntermediate K11N-[3-chloro-4-[4- (pyrrolidine-2- carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro-4- (3-methyl-1H- pyrazol-4- yl)phenyl]-1- methyl-imidazole- 2-carboxamide637.2Intermediate M1 and 1- tert- butoxycarbonyl pyrrolidine- 2-carboxylic acid; HCl Intermediate L1 2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]l-methyl-imidazole-2-carbonyl]amino]benzoic acid Step 1: tert-butyl 2-chloro-4-[[5-[2,3-difluoro-4-[3-methyl-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoate In a round bottomed flask were placed tert-butyl 4-(5-bromo-1-methyl-1H-imidazole-2-carboxamido)-2-chlorobenzoate (3.3 g, 7.96 mmol), 4-(2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazole (4.29 g, 9.52 mmol), PdCl2(dtbpf) (259 mg, 398 μmol) and potassium carbonate (3.3 g, 23.9 mmol). Water (7.23 mL) and dioxane (72.3 mL) were added, and the flask was evacuated and backfilled with argon for 5 times. The mixture was heated at 100° C. for 18h. The reaction was cooled to room temperature and poured into 100 mL water. The aqueous phase was extracted with EtOAc (50 mL×3). The organic layers were combined, washed with brine (50 mL), dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash chromatography to afford the product (2.46 g). MS [M+H]+: 658.2. Step 2: 2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoic acid tert-butyl 2-chloro-4-(5-(2,3-difluoro-4-(3-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamido)benzoate (2.45 g, 3.72 mmol) was stirred in TFA (3.72 mL) and CH2Cl2(14.9 mL) for 5h. The solvent was removed in vacuum and the residue was azeotroped with toluene (50 mL×3) to give the product which was used next without further purification (1.74 g). MS [M+H]+: 472.2. The following examples were prepared in analogy to Intermediate L1. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate L22-chloro-4-[[5- [2,3-difluoro-4-[1- (2-methoxyethyl)- 5-methyl-pyrazol- 4-yl]phenyl]-1- methyl-imidazole- 2-carbonyl]- amino]benzoic acid530.2Intermediate A1; Intermediate G3 and TFAIntermediate L32-chloro-4-[[5- [2,3-difluoro-4-[1- (2-methoxyethyl)- 3-methyl-pyrazol- 4-yl]phenyl]-1- methyl-imidazole- 2-carbonyl]- amino]benzoic acid530.2Intermediate A1; Intermediate G2 and TFAIntermediate L42-chloro-4-[[5-[4- (3,5-dimethyl-1H- pyrazol-4-yl)-2,3- difluoro-phenyl]-1- methyl-imidazole- 2- carbonyl]amino] benzoic acid486.1Intermediate A1; Intermediate G89 and TFA Intermediate M1 N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride Step 1: tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate 1-((dimethylamino)(dimethyliminio)methyl)-1H-[1,2,3]triazolo[4,5-b]pyridine 3-oxide hexafluorophosphate(V) (96.7 mg, 254 μmol) was added to a solution of N-ethyl-N-isopropylpropan-2-amine (137 mg, 1.06 mmol), 2-chloro-4-(5-(2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamido)benzoic acid (100 mg, 212 μmol) and tert-butyl piperazine-1-carboxylate (47.4 mg, 254 μmol) in DMA (4.24 mL) and stirred for 2h at room temperature. The mixture was poured into 100 mL water and extracted with EtOAc (50 mL×4). The organic layer was washed with 50 mL brine, dried over sodium sulfate, and concentrated in vacuum. The residue was purified by flash chromatography to afford the product (105 mg). MS [M+H]+: 640.2. Step 2: N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride tert-butyl 4-(2-chloro-4-(5-(2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamido)benzoyl)piperazine-1-carboxylate (1.38 g, 2.16 mmol) was dissolved in 4M HCl/MeOH (10.8 mL) solution. The solution was stirred at room temperature for 1h. The solvent was removed in vacuum and the residue was azeotroped with toluene (50 mL×3) to give the product, which was used in the next step without purification (1.17 g). MS [M+H]+: 540.2. The following examples were prepared in analogy to Intermediate M1. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate M2N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]- 5-[4-(3,5-dimethyl- 1H-pyrazol-4-yl)- 2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide554.2Intermediate L4 and tert- butyl piperazine-1- carboxylate and HCl Intermediate N1 and Intermediate N2 tert-butyl (1R,5S)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (Intermediate N1), and tert-butyl (1R,5S)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (Intermediate N2) Step 1: tert-butyl (1S,5R)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate In a 100 mL round-bottomed flask, (1R,5S,6r)-3-(tert-butoxycarbonyl)-3-azabicyclo[3.1.0]hexane-6-carboxylic acid (248 mg, 1.09 mmol), N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide (491 mg, 909 μmol), HATU (415 mg, 1.09 mmol) and DIPEA (235 mg, 1.82 mmol) were combined with DMF (6 mL) to give a light brown solution. The reaction was stirred at room temperature for 1 h. The reaction mixture was poured into 25 mL H2O and extracted with EtOAc (3×25 mL). The organic layers were combined, washed with sat NaCl (1×25 mL), The organic layers were dried over Na2SO4and concentrated in vacuum. The crude material was purified by flash chromatography to afford tert-butyl (1S,5R)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (520 mg). MS [M+H]+: 749.3. Step 2: tert-butyl (1R,5S)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (Intermediate N1), and tert-butyl (1R,5S)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (Intermediate N2) To a 5 mL microwave vial was added tert-butyl (1S,5R)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (330 mg, 440 μmol), 1-bromo-2-methoxyethane (91.8 mg, 661 μmol) and K2CO3(122 mg, 881 μmol) in DMF (3 mL). The vial was capped and heated in the microwave at 70° C. for 1 h. The reaction mixture was poured into 50 mL H2O and extracted with EtOAc (3×25 mL). The organic layers were combined, washed with sat NaCl (1×25 mL), The organic layers were dried over Na2SO4and concentrated in vacuum. The crude material was purified by flash chromatography to afford 220 mg crude product, The crude product was purified by preparative chiral-HPLC to afford tert-butyl (1R,5S)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (60 mg) and tert-butyl (1R,5S)-6-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (36 mg). MS [M+H]+: 807.4. Intermediate O1 2-[4-[4-[2-[[4-[4-(1-tert-butoxycarbonylpiperidine-4-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazol-1-yl]acetic acid Step 1: tert-butyl 4-[4-[2-chloro-4-[[5-[4-[1-(2-ethoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate A mixture of tert-butyl 4-[4-[4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (4554.2 mg, 7.14 mmol), ethyl 2-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3-methyl-pyrazol-1-yl]acetate (2900.0 mg, 7.14 mmol), Na2CO3(1513.3 mg, 14.28 mmol) and [1,1-Bis(di-tert-butylphosphino)ferrocene]palladium(II) Dichloride (943.4 mg, 1.07 mmol) in a flask. The flask was degassed and purged with N2gas for four times. 1,4-dioxane (30 mL) and water (6 mL) was added by injector to the mixture. The mixture was stirred at 90° C. for 2 h under N2. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to remove the solvent, then the product was purified by reversed-phase chromatography and dried by lyophilization to give tert-butyl 4-[4-[2-chloro-4-[[5-[4-[1-(2-ethoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (2750.0 mg). MS [M+H]+: 837.3. 2-[4-[4-[2-[[4-[4-(1-tert-butoxycarbonylpiperidine-4-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazol-1-yl]acetic acid To a solution of tert-butyl 4-[4-[2-chloro-4-[[5-[4-[1-(2-ethoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (2740.0 mg, 3.27 mmol) in Methanol (30.0 mL) and water (10.0 mL) was added LiOH (235.1 mg, 9.82 mmol). The mixture was stirred at 20° C. for 12 h under N2. The reaction mixture was quenched by water (50 mL), extracted with EtOAc (100×3 mL). The combined organic layers were washed brine (50 mL), dried (Na2SO4) and concentrated to give 2-[4-[4-[2-[[4-[4-(1-tert-butoxycarbonylpiperidine-4-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl] carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazol-1-yl]acetic acid (2500.0 mg). MS [M+H]+: 809.3. The following examples were prepared in analogy to Intermediate O1. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate O22-[4-[4-[2-[[4-[4- (1-tert- butoxycarbonyl piperidine-4- carbonyl)piperazine- 1-carbonyl]-3- chloro- phenyl]carbamoyl]- 3-methyl-imidazol- 4-yl]-2,3-difluoro- phenyl]-5-methyl- pyrazol-1-yl]acetic acid809.6Intermediate D1; Intermediate G30 and LiOH Intermediate P1 N-[4-[4-[1-(2-amino-2-oxo-ethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-bromo-1-methyl-imidazole-2-carboxamide Step 1: 5-bromo-N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide To a solution of tert-butyl 4-(4-(4-(5-bromo-1-methyl-1H-imidazole-2-carboxamido)-2-chlorobenzoyl)piperazine-1-carbonyl)piperidine-1-carboxylate (1.9 g, 3.0 mmol) in DCM (25 mL) and was TFA (5 mL) then the resultant mixture was stirred for 1.0 h at room temperature. The mixture was basified with aqueous ammonia to pH=9-10 and then the mixture was poured into water (50 mL) and then extracted with dichloromethane/isopropanol (100/10 mL), the organic layer was concentrated in vacuum to give 5-bromo-N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide (1.2 g), which was used in next step without purification. MS [M+H]+: 537.3. Step 2: N-[4-[4-[1-(2-amino-2-oxo-ethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-bromo-1-methyl-imidazole-2-carboxamide To a solution of 5-bromo-N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide (2.15 g, 4.0 mmol) and DIPEA (1.55 g, 12 mmol) in acetonitrile (25 ml) was added 2-iodoacetamide (888 mg, 4.8 mmol) at room temperature, and then the resultant mixture was stirred overnight. The mixture was poured into water (50 mL) and then extracted with dichloromethane/isopropanol (100/10 mL), the organic layer was concentrated to give a red oil, which was purified by flash chromatography on silica gel to afford N-[4-[4-[1-(2-amino-2-oxo-ethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-bromo-1-methyl-imidazole-2-carboxamide (1.8 g). MS [M+H]+: 594.2. Intermediate Q1 tert-butyl 4-[4-[4-[[5-[4-[1-(2-aminoethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate Step 1: tert-butyl 4-[4-[4-[[5-[4-[1-[2-[tert-butyl(dimethyl)silyl]oxyethyl]-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate 4-[4-[4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester (1.5 g, 2.35 mmol) was dissolved in 1,4-dioxane (15 mL) and water (1.5 mL). tert-butyl-[2-[4-[2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5-methyl-pyrazol-1-yl]ethoxy]-dimethyl-silane (1.46 g, 3.06 mmol), 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride (153.24 mg, 0.235 mmol) and Na2CO3(747.64 mg, 7.05 mmol) were added at rt. The mixture was stirred at 100° C. for 15 h under N2. The reaction was filtered, the filtrate was concentrated under vacuum, The crude compound was purified by flash chromatography on silica gel, to afford 4-[4-[4-[[5-[4-[1-[2-[tert-butyl(dimethyl)silyl]oxyethyl]-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester (1.2 g). MS [M+H]+: 910.1. Step 2: tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-hydroxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate tert-butyl 4-[4-[4-[[5-[4-[1-[2-[tert-butyl(dimethyl)silyl]oxyethyl]-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (1.2 g, 1.32 mmol) was dissolved in N,N-dimethylformamide (20 mL) and water (5 mL), ammonium fluoride ((NH4)F) (977.37 mg, 26.39 mmol) was added at rt. The reaction was stirred at 60° C. for 1 h. The reaction mixture was diluted with water (80 mL) and extracted two times with EtOAc (40 mL). The organic layers were washed with brine (30 mL), dried over Na2SO4and concentrated to dryness. The crude product was directly used to the next step, to afford tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-hydroxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (1.04 g). MS [M+H]+: 795.9. Step 3: tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[5-methyl-1-(2-methylsulfonyloxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-hydroxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (1 g, 1.26 mmol) was dissolved in dichloromethane (30 mL), Methanesulfonic anhydride (328.57 mg, 1.89 mmol) and DIEA (325.02 mg, 2.51 mmol) were added at rt. The mixture was stirred at room temperature for 3 h. The reaction was concentrated under vacuum, The crude material was purified by flash chromatography on silica gel to afford tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[5-methyl-1-(2-methylsulfonyloxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (580 mg). MS [M+H]+: 873.8. Step 4: tert-butyl 4-[4-[4-[[5-[4-[1-(2-aminoethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[5-methyl-1-(2-methylsulfonyloxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (580 mg, 0.664 mmol) was dissolved in methanol (10 mL), 7 M ammonia (3.73 g, 33.21 mmol) was added at rt. The mixture was stirred at 80° C. for 15 h. The reaction was concentrated under vacuum, the crude product was directly used to the next step, to afford tert-butyl 4-[4-[4-[[5-[4-[1-(2-aminoethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (527 mg). MS [M+H]+: 794.8. The following examples were prepared in analogy to Intermediate Q1. MSESIStartingEx #NameStructure[M + H]+MaterialIntermediate Q2tert-butyl 4-[4-[4- [[5-[4-[1-(2- aminoethyl)-3- methyl-pyrazol-4- yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carbonyl]amino]-2- chloro- benzoyl]piperazine- 1- carbonyl]piperidine- 1-carboxylate530.2Intermediate D1 and Intermediate G27; ammonium fluoride; Methanesulfonic anhydride; ammonia Intermediate R1 O1-tert-butyl O2-methyl (2S,4R)-4-[tert-butyl(diphenyl)silyl]oxypyrrolidine-1,2-dicarboxylate 1-(tert-butyl) 2-methyl (2S,4R)-4-hydroxypyrrolidine-1,2-dicarboxylate (500 mg, 2.04 mmol), tert-butylchlorodiphenylsilane (672 mg, 2.45 mmol), 1H-imidazole (278 mg, 4.08 mmol) and N,N-dimethylpyridin-4-amine (24.9 mg, 204 μmol) were stirred in CH2Cl2(10.2 mL) for 18 h. The reaction mixture was quenched with a saturated aqueous Na2CO3solution until pH=10. The aqueous layer was extracted with CH2Cl2and the combined organic phases were dried over Na2SO4and filtered. The solvent was removed in vacuum to afford an oil, which was purified by flash column chromatography on silica gel to afford the product (902 mg). MS [M+H]+: 484.2. Intermediate R2 (2S,4R)-4-[tert-butyl(diphenyl)silyl]oxy-1-methyl-pyrrolidine-2-carboxylic acid Step 1: methyl (2S,4R)-4-[tert-butyl(diphenyl)silyl]oxypyrrolidine-2-carboxylate O1-tert-butyl O2-methyl (2S,4R)-4-[tert-butyl(diphenyl)silyl]oxypyrrolidine-1,2-dicarboxylate (700 mg, 1.45 mmol) was dissolved in 5 mL 20% TFA/DCM solution and stirred at room temperature for 1 h. The solvent was removed in vacuum. The residue was neutralized with 100 mL sat. K2CO3, extracted with MeOH/DCM (50 mL×4, v/v=1:10), dried over sodium sulfate and concentrated in vacuo to afford the product (545 mg). MS [M+H]+: 384.2. Step 2: methyl (2S,4R)-4-[tert-butyl(diphenyl)silyl]oxy-1-methyl-pyrrolidine-2-carboxylate methyl (2S,4R)-4-((tert-butyldiphenylsilyl)oxy)pyrrolidine-2-carboxylate (270 mg, 704 μmol) and formaldehyde (85.7 mg, 1.06 mmol) (37% aqueous solution) was dissolved in 1,2-dichloroethane (3.52 mL) and stirred at room temperature for 30 min before adding of NaBH3CN (66.4 mg, 1.06 mmol). The mixture was then continued for 3 h. The mixture was poured into 100 mL water and extracted with DCM (50 mL×3). The organic layers were combined, washed with 50 mL brine, dried over sodium sulfate and concentrated in vacuum. The residue was purified by flash chromatography to afford the product (230 mg) as colorless oil. MS [M+H]+: 398.2. Step 3: (2S,4R)-4-[tert-butyl(diphenyl)silyl]oxy-1-methyl-pyrrolidine-2-carboxylic acid methyl (2S,4R)-4-[tert-butyl(diphenyl)silyl]oxy-1-methyl-pyrrolidine-2-carboxylate (230 mg) was dissolved in 5 mL MeOH and 1 mL water. To this solution was added 100 mg LiOH. The resulting mixture was stirred at room temperature for 48 h. The solution was poured into 100 mL 1N HCl and extracted with DCM (50 mL×4). The organic layers were combined, washed with 50 mL brine, dried over sodium sulfate and concentrated in vacuum. The crude product (200 mg) was used in the next step without purification. MS [M+H]+: 384.2. Intermediate R3 (2S,4R)-1-tert-butoxycarbonyl-4-[tert-butyl(diphenyl)silyl]oxy-pyrrolidine-2-carboxylic acid 1-(tert-butyl) 2-methyl (2S,4R)-4-((tert-butyldiphenylsilyl)oxy)pyrrolidine-1,2-dicarboxylate (200 mg, 413 μmol) and lithium hydroxide (99 mg, 4.13 mmol) were stirred in water (345 μL) and MeOH (1.72 mL) for 18 h. After completion, the mixture was poured into 100 mL 1M HCl. The aqueous phase was extracted with DCM (50 mL×3). The organic layers were combined, washed with 50 mL brine, dried over sodium sulfate and concentrated in vacuum. The crude product was used in the coming step without purification. MS [M+H]+: 470.3. Intermediate R4 5-(difluoromethyl)-4-iodo-1-(2-methoxyethyl)pyrazole To a solution of 4-iodo-2-(2-methoxyethyl)pyrazole-3-carbaldehyde (1000 mg, 3.57 mmol) in dichloromethane (10 mL) was added last (748.22 mg, 613.3 uL, 4.64 mmol) slowly at 0° C., the reaction was gradually warmed to room temperature and stirred for 18 h. The reaction was quenched with water. The reaction mixture was washed with brine and extracted in DCM. The organic layer was dried over anhydrous Na2SO4and concentrated in vacuum The residue was purified by column chromatography to give 5-(difluoromethyl)-4-iodo-1-(2-methoxyethyl)pyrazole (800 mg). MS [M+H]+: 303.0. Intermediate R5 4-methyl-1-(4-piperidylmethyl)piperazin-2-one Step 1: tert-butyl 4-[(4-methyl-2-oxo-piperazin-1-yl)methyl]piperidine-1-carboxylate sodium hydride (275 mg, 6.84 mmol) was added to a solution of 4-methylpiperazin-2-one (650 mg, 5.7 mmol) in anhydrous DMF (15 ml). After 30 min, tert-butyl 4-(bromomethyl)piperidine-1-carboxylate (1.60 g, 5.7 mmol) was added and the mixture was stirred for 12 h at room temperature. The mixture was poured into water (80 mL) and extracted with EtOAc (50 mL×3). The organic layers were combined, washed with water and brine, dried over sodium sulfate and concentrated in vacuum to give tert-butyl 4-[(4-methyl-2-oxo-piperazin-1-yl)methyl]piperidine-1-carboxylate (1.2 g), which was directly used in the next step without further purification. MS [M+H]+: 312.2. Step 2: 4-methyl-1-(4-piperidylmethyl)piperazin-2-one To a solution of tert-butyl 4-((4-methyl-2-oxopiperazin-1-yl)methyl)piperidine-1-carboxylate (1.2 g, 3.8 mmol) in DCM (10 mL) and was added TFA (3 mL) then the resultant mixture was stirred for 1.0 h at room temperature. The solvent was concentrated under reduced pressure to give 4-methyl-1-(4-piperidylmethyl)piperazin-2-one (600 mg). MS [M+H]+: 212.2. Intermediate R6 rac-(2S)-1-tert-butoxycarbonyl-4-(hydroxymethyl)pyrrolidine-2-carboxylic acid Step 1: O1-tert-butyl O2-methyl rac-(2S)-4-(hydroxymethyl)pyrrolidine-1,2-dicarboxylate To a solution of O1-tert-butyl O2-methyl (2S)-4-methylenepyrrolidine-1,2-dicarboxylate (1.0 g, 4.14 mmol) in THF (20.0 mL) was added 9-borabicyclo[3.3.1]nonane (0.5 M in THF) (9.95 mL, 4.97 mmol) slowly under N2at 0° C. After addition, this reaction mixture was warmed to 20° C. and stirred for 4 h. Then sodium hydroxide (2 M) (5.18 mL, 10.36 mmol) and hydrogen peroxide (1.46 g, 12.85 mmol) were added into this mixture slowly at 0° C. This reaction mixture was stirred at 0° C. for 1 h. Then the reaction mixture was warmed to 20° C. and stirred for further 2 h. This reaction was quenched by saturated aqueous Na2SO3(50 mL) and extracted by EtOAc (20 mL×2). The combined organic layers were dried over Na2SO4and concentrated to get the crude product. This crude product was purified by silica gel chromatography (PE:EtOAc=1:1) to get O1-tert-butyl O2-methyl rac-(2S)-4-(hydroxymethyl)pyrrolidine-1,2-dicarboxylate (500.0 mg) as colorless oil. MS [M+H]+: 260.2. Step 2: rac-(2S)-1-tert-butoxycarbonyl-4-(hydroxymethyl)pyrrolidine-2-carboxylic acid To a solution of rac-(2S)-4-(hydroxymethyl)pyrrolidine-1,2-dicarboxylate (400.0 mg, 1.54 mmol) in methanol (5.0 mL), THE (5 mL) and water (1 mL) was added lithium hydroxide monohydrate (97.1 mg, 2.31 mmol) in one portion. This reaction mixture was stirred at 25° C. for 1 h. This reaction mixture was concentrated to get the crude product. This crude product was purified by Prep-HPLC (FA) to get rac-(2S)-1-tert-butoxycarbonyl-4-(hydroxymethyl)pyrrolidine-2-carboxylic acid (160.0 mg, 0.65 mmol, 34.34% yield) as yellow oil. MS [M+H]+: 146.0. Intermediate R7 2-[4-hydroxy-4-(hydroxymethyl)-1-piperidyl]-1-piperazin-1-yl-ethanone; 2,2,2-trifluoroacetic acid Step 1: tert-butyl 4-(2-bromoacetyl)piperazine-1-carboxylate To a solution of tert-butyl piperazine-1-carboxylate (500.0 mg, 2.68 mmol) and in DCM (10.0 mL) was added 2-bromoacetyl chloride (464.78 mg, 2.95 mmol) slowly at 0° C. After addition, the reaction mixture was stirred at 0° C. for 2 h. This reaction was quenched by saturated aqueous NaHCO3(20 mL) and extracted by DCM (10 mL×2). The combined organic layers were dried over Na2SO4and concentrated to get tert-butyl 4-(2-bromoacetyl)piperazine-1-carboxylate (800.0 mg), the crude product would be used in the next step directly without further purification. MS [M+H-C4H8]+: 251.0. Step 2: tert-butyl 4-[2-[4-hydroxy-4-(hydroxymethyl)-1-piperidyl]acetyl]piperazine-1-carboxylate To a solution of 4-(hydroxymethyl)piperidin-4-ol (100.0 mg, 0.76 mmol) and potassium carbonate (210.73 mg, 1.52 mmol) in ACN (5.0 mL) was added tert-butyl 4-(2-bromoacetyl)piperazine-1-carboxylate (100.0 mg, 0.76 mmol) and potassium carbonate (351.28 mg, 1.14 mmol) in one portion. The reaction was stirred at room temperature for 1 h. This reaction mixture was filtered, and the filtrate was concentrated to get the crude product. This crude product was purified by Prep-HPLC (FA). The solution of desired product in ACN/H2O was lyophilized to get tert-butyl 4-[2-[4-hydroxy-4-(hydroxymethyl)-1-piperidyl]acetyl]piperazine-1-carboxylate (180.0 mg). MS [M+H]+: 358.1. Step 3: 2-[4-hydroxy-4-(hydroxymethyl)-1-piperidyl]-1-piperazin-1-yl-ethanone; 2,2,2-trifluoroacetic acid To a solution of tert-butyl 4-[2-[4-hydroxy-4-(hydroxymethyl)-1-piperidyl]acetyl]piperazine-1-carboxylate (180.0 mg, 0.5 mmol) in DCM (5 mL) was added trifluoroacetic acid (0.5 mL, 6.49 mmol) in one portion. This reaction mixture was stirred at 25° C. for 1 h. This reaction mixture was concentrated to get 2-[4-hydroxy-4-(hydroxymethyl)-1-piperidyl]-1-piperazin-1-yl-ethanone; 2,2,2-trifluoroacetic acid (190.0 mg), The crude product would be used in the next step directly without further purification. MS [M+H]+: 258.1. Intermediate R8 tetrahydropyran-3-amine Step 1: tetrahydropyran-3-one oxime To a solution of tetrahydropyran-3-one (400.0 mg, 4.0 mmol) and potassium carbonate (828.2 mg, 5.99 mmol) in ACN (5 mL) was added hydroxylamine hydrochloride (555.3 mg, 7.99 mmol). The reaction mixture was stirred at 15° C. for 2 h. This reaction mixture was filtered and concentrated under reduced pressure to afford the crude product. The crude product was purified by silica gel chromatography to get tetrahydropyran-3-one oxime (400.0 mg). Step 2: tetrahydropyran-3-amine To a solution of tetrahydropyran-3-one oxime (350.0 mg, 3.04 mmol) in methanol (5.0 mL) was added palladium on carbon (161.7 mg) in one portion under N2. This mixture was degassed and purged with N2for 3 times. Then H2(15 psi) was introduced into this system. The reaction mixture was stirred at 30° C. for 6 h under H2atmosphere. This reaction was filtered carefully and concentrated to get the crude product tetrahydropyran-3-amine (150.0 mg). Intermediate R9 4-methyl-N-(tetrahydropyran-4-ylideneamino)benzenesulfonamide To a solution of tetrahydropyran-4-one (2.0 g, 19.98 mmol) in methanol (30.0 mL) was added 4-methylbenzenesulfonhydrazide (3.7 g, 19.98 mmol) the mixture was stirred at 20° C. for 3 h. The mixture was concentrated to give 4-methyl-N-(tetrahydropyran-4-ylideneamino)benzenesulfonamide (5.3 g, 19.75 mmol) as a white solid. MS [M+H]+: 269.0. Intermediate R10 4-(tert-butoxycarbonylamino)-1,1-dimethyl-piperidin-1-ium-4-carboxylic acid; chloride To a solution of 4-((tert-butoxycarbonyl)amino)piperidine-4-carboxylic acid (300 mg, 1.23 mmol) in MeOH (5 mL) was added sodium hydroxide (147 mg, 3.68 mmol) and methyl iodide (872 mg, 6.14 mmol), the reaction was stirred for two hours at room temperature. The reaction mixture was acidified with 3N HCl and concentrated in vacuum to give 4-(tert-butoxycarbonylamino)-1,1-dimethyl-piperidin-1-ium-4-carboxylic acid; chloride, the crude product was directly used for the next step without further purification. MS [M]+: 273.2. Intermediate R11 (2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)-bis[3-(methylamino)propyl]ammonium; bromide Step 1: benzyl 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoate To a solution of 3-(BOC-amino)propyl bromide (4.56 g, 19.16 mmol) in ACN (30 mL) was added benzyl 4-aminobutanoate; hydrochloride (2.0 g, 8.71 mmol) at 10° C., then the mixture was stirred at 70° C. for 16 h. The solution was concentrated, the residue was purified by reversed phase-HPLC (FA) to afford benzyl 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoate (2.2 g). MS [M+H]+: 508.4. Step 2: (4-benzyloxy-4-oxo-butyl)-bis[3-(tert-butoxycarbonylamino)propyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; bromide To a solution of tert-butyl bromoacetate (1.27 mL, 7.88 mmol) in ACN (20 mL) was added benzyl 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoate (2.0 g, 3.94 mmol) at 10° C., then the mixture was stirred at 80° C. for 16 h. The solution was concentrated, the residue was purified by reversed phase-HPLC (TFA) to afford (4-benzyloxy-4-oxo-butyl)-bis[3-(tert-butoxycarbonylamino)propyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; bromide (1 g) as colorless oil. MS [M]+: 622.5. Step 3: bis[3-(tert-butoxycarbonylamino)propyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide A solution of palladium on activated carbon (151.44 mg, 0.140 mmol) in Methanol (10 mL) was added (4-benzyloxy-4-oxo-butyl)-bis[3-(tert-butoxycarbonylamino)propyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; bromide (1.0 g, 1.42 mmol) under N2, then the mixture was stirred under H2at 10° C. for 16 h. The starting materials were consumed completed. The mixture was filtered and concentrated to afford bis[3-(tert-butoxycarbonylamino)propyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide (700 mg). MS [M]+: 532.4. The following examples were prepared in analogy to Intermediate R11. MSESIStartingEx#NameStructure[M + H]+MaterialIntermediate R12bis[(1-tert-butoxy- carbonylazetidin- 3-yl)methyl]- (2-tert-butoxy-2- oxo-ethyl)-(3- carboxypropyl) ammonium; bromide556.4benzyl 4- aminobutanoate and tert- butyl 3- (bromomethyl) azetidine-1- carboxylate; tert-butyl bromoacetate Intermediate R13 [(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-yl]-piperazin-1-yl-methanone Step 1: benzyl 4-[(2S,4R)-1-tert-butoxycarbonyl-4-hydroxy-pyrrolidine-2-carbonyl]piperazine-1-carboxylate To a mixture of 1-CBZ-piperazine (5.0 g, 22.7 mmol) in THE (25 mL) and DMF (25 mL) was added N,N-diisopropylethylamine (17.57 g, 136.2 mmol) and BOC-HYP-OH (5.25 g, 22.7 mmol). Then the mixture was added propylphosphonic anhydride (18.78 g, 29.51 mmol) after 10 min. The reaction mixture was stirred at 25° C. for 16 h. The solution was extracted with water (50 mL) and EA (100 mL) and washed with sat. aq NaCl (50 mL), dried by anhydrous Na2SO4. The crude was purified by prep-HPLC (FA) to obtain the title compound (9.8 g). MS [M+H]+: 434.5. Step 2: benzyl 4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carboxylate To a mixture of benzyl 4-[rac-(2S,4R)-1-tert-butoxycarbonyl-4-hydroxy-pyrrolidine-2-carbonyl]piperazine-1-carboxylate (9.84 g, 22.7 mmol) in methanol (20 mL) was added HCl/dioxane (20 mL). The reaction mixture was stirred at 25° C. for 16 h. The solution was not purified and used next step directly to obtain the title compound (7.56 g). MS [M+H]+: 334.5. Step 3: benzyl 4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carboxylate To a mixture of benzyl 4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carboxylate (7.56 g, 22.68 mmol) in MeCN (70 mL) and water (7 mL) was added iodomethane (32.19 g, 226.77 mmol) and triethylamine (63.21 mL, 453.54 mmol). The reaction mixture was stirred at 25° C. for 16 h. The mixture was concentrated to remove solvent, purified by prep-HPLC (0.1% FA) to afford the title compound (4 g). MS [M]+: 362.4. Step 4: piperazin-1-yl-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-yl]methanone To a mixture of benzyl 4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carboxylate (1600.0 mg, 4.41 mmol) in methanol (20 mL) and were added ammonium hydroxide (2.0 mL, 4.41 mmol) and wet palladium 10% on activated carbon (0.140 mmol, 0.030 eq). The reaction mixture was stirred at 25° C. for 16 h under hydrogen at 15 psi. The mixture was filtered and filtrate was concentrated to remove solvent, to afford the title compound (1 g). MS [M]+: 228.2. Intermediate R14 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylate Step 1: benzyl 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]piperidine-4-carboxylate To a solution of benzyl piperidine-4-carboxylate hydrochloride (500.0 mg, 1.96 mmol) and potassium carbonate (811 mg, 5.87 mmol) in DMF (10 mL) was added 1-BOC-3-(bromomethyl)azetidine (587 mg, 2.35 mmol). The mixture was stirred at 25° C. for 16 h. Then the mixture was stirred at 50° C. for another 16 h. The mixture was diluted with EtOAc (100 mL) and then washed with brine (30 mL×3). The organic layer was dried over sodium sulfate, filtered and the filtrate was concentrated under vacuum. The residue was purified by column chromatography (PE:EA=2:1˜0:1) to give the title compound (710 mg, 1.83 mmol, 74.17% yield) as light yellow gum. MS [M+H]+: 389.1. Step 2: benzyl 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylate; formate To a solution of benzyl 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]piperidine-4-carboxylate (4.7 g, 12.1 mmol) and sodium iodide (181 mg, 1.21 mmol) in DMF (50 mL) was added tert-butyl bromoacetate (4.72 g, 24.2 mmol) and N,N-diisopropylethylamine (6.32 mL, 36.29 mmol). The mixture was stirred at 60° C. for 16 h. The mixture was concentrated under vacuum. The residue was purified twice by prep-HPLC (FA condition) to give the title compound (5 g, 9.93 mmol, 82.06% yield) as a yellow solid. MS [M]+: 503.2. Step 3: 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylate To a solution of benzyl 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylate formate (4.7 g, 8.57 mmol) in methanol (150 mL) was added palladium on charcoal (400 mg, 10% purity) and palladium hydroxide on charcoal (400.0 mg, 10% wt) under nitrogen atmosphere. The mixture was degassed and then stirred at 15° C. for 4 h under hydrogen (760 mmHg). The mixture was filtered through celite pad, the solid was washed with MeOH (20 mL×4). The combined filtrate was concentrated under vacuum. The solid was dissolved in water (100 mL) and then lyophilized to give 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylate (3.5 g, 8.48 mmol, 93.86% yield) as a white solid. MS [M]+: 413.2. Intermediate R15 Tert-butyl 2-[1-[3-(tert-butoxycarbonylamino)propyl]piperazin-1-ium-1-yl]acetate; formate Step 1: benzyl 4-[3-(tert-butoxycarbonylamino)propyl]piperazine-1-carboxylate To a solution of 1-CBZ-piperazine (5.0 g, 22.7 mmol) in MeCN (100 mL) was added triethylamine (3.16 mL, 22.7 mmol) and 3-(BOC-amino)propyl bromide (5.68 g, 23.83 mmol), then the mixture was stirred at 25° C. for 16 h. The mixture was concentrated in vacuum and purified by silica gel column (PE/EA=100:1˜1:2) to obtain the title compound (5.2 g, 13.78 mmol, 60.69% yield) as light brown solid. MS [M+H]+: 378.3. Step 2: benzyl 4-[3-(tert-butoxycarbonylamino)propyl]-4-(2-tert-butoxy-2-oxo-ethyl)piperazin-4-ium-1-carboxylate; formate To a solution of benzyl 4-[3-(tert-butoxycarbonylamino)propyl]piperazine-1-carboxylate (5.2 g, 13.78 mmol) in MeCN (100 mL) was added triethylamine (1.92 mL, 13.78 mmol) and tert-butyl bromoacetate (5.37 g, 27.55 mmol), then the mixture was stirred at 50° C. for 16 h. The mixture was concentrated in vacuum and purified by prep-HPLC (0.1% FA)-MeOH to obtain the title compound (4 g, 59% yield) as light yellow solid. MS [M+H]+: 492.4. Step 3: tert-butyl 2-[1-[3-(tert-butoxycarbonylamino)propyl]piperazin-1-ium-1-yl]acetate; formate To a solution of benzyl 4-[3-(tert-butoxycarbonylamino)propyl]-4-(2-tert-butoxy-2-oxo-ethyl)piperazin-4-ium-1-carboxylate formate (4.0 g, 8.12 mmol) in THF (40 mL) was added 10% palladium on charcoal (400 mg) and the reaction stirred under hydrogen atmosphere at 25° C. for 16 h. The mixture was concentrated in vacuum and purified by prep-HPLC (0.1% FA)-ACN to obtain tert-butyl 2-[1-[3-(tert-butoxycarbonylamino)propyl]piperazin-1-ium-1-yl]acetate; formate (1.5 g, 4.18 mmol, 51.53% yield) as white solid. MS [M]+: 358.3. Example A1 N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate In a 50 mL round-bottomed flask, afford N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (131 mg, 185 μmol), Mel (105 mg, 739 μmol) and DIPEA (95.5 mg, 739 μmol) were combined with MeCN (6 mL) to give a light brown solution. The reaction was stirred at room temperature for 15 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by preparative HPLC to afford N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (80 mg). MS [M]+: 737.3. The following examples were prepared in analogy to Examples A1. MSESIStartingEx#NameStructure[M]+MaterialExample A2N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate751.3Intermediate H4 and iodomethaneExample A3N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[4- [1-(2-methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate715.2Intermediate H5 and iodomethaneExample A4N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate737.4Intermediate H2 and iodomethaneExample A5N-[3-chloro-4-[4-[2- [(3S)-1,1- dimethylpyrrolidin-1-ium-3- yl]acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate737.2Example 12 and iodomethaneExample A6N-[3-chloro-4-[4-(4- hydroxy-1,1-dimethyl- piperidin-1-ium-4- carbonyl)piperazne-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate753.3Intermediate K2 and iodomethaneExample A7N-[3-chloro-4-[4-(3- hydroxy-1,1-dimethyl- piperidin-1-ium-4- carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate753.3Intermediate K3 and iodomethaneExample A8[2-[4-[2-chloro-4-[[5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carbonyl]amino]benzoyl] piperazin-1-yl]-2-oxo- ethyl]-trimethyl- ammonium; 2,2,2- trifluoroacetate697.3Intermediate K4 and iodomethaneExample A9[2-[4-[[[2-chloro-4-[[5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carbonyl]amino]benzoyl] amino]methyl]-1- piperidyl]-2-oxo-ethyl]- trimethyl-ammonium; 2,2,2-trifluoroacetate725.3Intermediate K5 and iodomethaneExample A10N-[3-chloro-4-[4-(4,4- dimethylpiperazin-4- ium-1- carbonyl)piperidine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate737.1Intermediate H6 and iodomethaneExample A11N-[3-chloro-4-[4- [(2S,3S)-3-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate753.4Intermediate K6 and iodomethaneExample A12N-[3-chloro-4-[4-[(2S)- 4-(hydroxymethyl)-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate753.4Intermediate K8 and iodomethaneExample A13N-[3-chloro-4-[4-(4- methoxy-1,1-dimethyl- piperidin-1-ium-4- carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate767.3Example D44 and iodomethane Example B1 N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate Step 1: N-[3-chloro-4-[4-(1-methylpiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide In a 100 mL round-bottomed flask, afford N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (350 mg, 494 μmol), formaldehyde (74.1 mg, 2.47 mmol) and NaBH3CN (155 mg, 2.47 mmol) were combined with MeOH (12 mL) to give a light brown solution. The reaction mixture was heated to 50° C. and stirred for 1 h. The crude reaction mixture was concentrated in vacuum. The reaction mixture was poured into 25 mL sat NaHCO3and extracted with EtOAc (3×25 mL). The organic layers were combined, washed with sat NaCl (1×25 mL), The organic layers were dried over Na2SO4and concentrated in vacuum to afford N-[3-chloro-4-[4-(1-methylpiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (357 mg). MS [M+H]+: 723.4. Step 2: N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate In a 50 mL round-bottomed flask, N-[3-chloro-4-[4-(1-methylpiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (89 mg, 123 μmol), 2-iodoacetamide (45.5 mg, 246 μmol) and DIPEA (79.5 mg, 615 μmol) were combined with MeCN (5 mL) to give a light brown solution. The reaction was stirred at room temperature for 15 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by preparative HPLC to afford N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (22.6 mg). MS [M]+: 780.3. Example B2 & Example B3 N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (Example B2) N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (Example B3) N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate was purified by preparative chial-HPLC. To afford N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (42.6 mg) and N-[4-[4-[1-(2-amino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (38 mg). MS [M]+: 780.2. The following examples were prepared in analogy to Examples B1. MSESIStartingEx#NameStructure[M + H]+MaterialExample B4N-[3-chloro-4-[4-[1-(2- hydroxyethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate767.4Intermediate H1; formaldehyde and 2- bromoethanolExample B5N-[4-[4-[1-(azetidin-3- ylmethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid; formate792.2Intermediate H1; formaldehyde and tert-butyl 3- (iodomethyl) azetidine-1- carboxylateExample B6N-[4-[4-[1-(2-amino-2- oxo-ethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate780.3Intermediate H2; formaldehyde and 2- iodoacetamideExample B7N-[4-[4-[1-(2-amino-2- oxo-ethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[4-[1-(2,2- difluoroethyl)-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-1- methyl-imidazole-2- carboxamide; formate786.3Intermediate H3; formaldehyde and 2- iodoacetamideExample B8N-[3-chloro-4-[4-[1-(2- hydroxyethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate781.3Intermediate H4; formaldehyde and 2- bromoethanolExample B9N-[3-chloro-4-[4-[1-(2- methoxyethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate795.3Intermediate H4; formaldehyde and 1-bromo- 2-methoxy- ethaneExample B10N-[4-[4-[1-(2-amino-2- oxo-ethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate758.3Intermediate H5; formaldehyde and 2- iodoacetamideExample B11N-[3-chloro-4-[4-[1-(2- hydroxyethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]phenyl]-5-[4- [1-(2-methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate745.4Intermediate H5; formaldehyde and 2- bromoethanolExample B12N-[3-chloro-4-[4-[1-(2- hydroxyethyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate767.4Intermediate H2; formaldehyde and 2- bromoethanolExample B132-[4-[1-[2-chloro-4-[[5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carbonyl]amino]benzoyl] piperidine-4-carbonyl]-1- methyl-piperazin-1-ium- l-yl]acetic acid; 2,2,2- trifluoroacetate781.3Intermediate H6; formaldehyde and tert-butyl 2- bromoacetateExample B142-[1-(2-amino-2-oxo- ethyl)-4-[1-[2-chIoro-4- [[5-[2,3-difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carbonyl]amino]benzoyl] piperidine-4- carbonyl]piperazin-1- ium-1-yl]acetic acid; 2,2,2-trifluoroacetate824.3Intermediate H6; 2- iodoacetamide and tert-butyl 2- bromoacetateExample B15N-[4-[4-[4-(2-amino-2- oxo-ethyl)-4-methyl- piperazin-4-ium-1- carbonyl]piperidine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate780.4Intermediate H6; formaldehyde and 2- iodoacetamideExample B16N-[4-[4-[1-(2-amino-2- oxo-ethyl)-4-hydroxy-1- methyl-piperidin-1-ium- 4-carbonyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate796.4Intermediate K7; formaldehyde and 2- iodoacetamide Example B17 N-[3-chloro-4-[4-[1-(2-hydrazino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formic acid; formate Step 1: N-[3-chloro-4-[4-(1-methylpiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (420 mg, 0.592 mmol) was dissolved in methanol (10 mL), formaldehyde (88.92 mg, 2.96 mmol) and sodium cyanoborohydride (186.08 mg, 2.96 mmol) were added at rt. The mixture was stirred at 50° C. for 2 h. The reaction mixture was concentrated and diluted with water (20 mL) and extracted two times with EtOAc (20 mL). The organic layers were washed with brine (20 mL), dried over Na2SO4and concentrated to dryness. The crude product was directly used to the next step to afford N-[3-chloro-4-[4-(1-methylisonipecotoyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (400 mg). MS [M+H]+: 723.6. Step 2: tert-butyl 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetate N-[3-chloro-4-[4-(1-methylisonipecotoyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (100 mg, 0.138 mmol) was dissolved in acetonitrile (10 mL), and tert-butyl bromoacetate (107.88 mg, 0.553 mmol) and DIEA (71.48 mg, 0.553 mmol) were added at rt. The mixture was stirred at room temperature for 1 h. The reaction was concentrated under vacuum, the crude product was directly used to the next step to afford tert-butyl 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetate (115 mg). MS [M]+: 837.7. Step 3: 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetic acid tert-butyl 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetate (115 mg, 0.137 mmol) was dissolved in tetrahydrofuran (2 mL) and 4 M HCl (in dioxane) (2.06 g, 6.86 mmol) was added at rt. The mixture was stirred at room temperature for 2 h. The reaction was concentrated under vacuum, the crude product was directly used to the next step to afford 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetic acid (107 mg). MS [M]+: 781.7. Step 4: tert-butyl N-[[2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetyl]amino]carbamate 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetic acid (107 mg, 0.137 mmol) was dissolved in N,N-dimethylformamide (3 mL), N-aminocarbamic acid tert-butyl ester (126.54 mg, 0.957 mmol), DIEA (35.36 mg, 0.274 mmol) and HATU (364.06 mg, 0.957 mmol) were added at rt. The mixture was stirred at room temperature for 1 h. The reaction was directly used to the next step to afford tert-butyl N-[[2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetyl]amino]carbamate (122.61 mg) as light brown solid. MS [M]+: 895.8. Step 5: N-[3-chloro-4-[4-[1-(2-hydrazino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formic acid; formate N-[[2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-methyl-piperidin-1-ium-1-yl]acetyl]amino]carbamate (122.61 mg, 0.137 mmol) was dissolved in N,N-dimethylformamide (2 mL), and TFA (779.79 mg, 6.84 mmol) was added at rt. The mixture was stirred at room temperature for 1h. The reaction was concentrated under vacuum, the crude product was purified by HPLC to afford N-[3-chloro-4-[4-[1-(2-hydrazino-2-oxo-ethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formic acid; formate (12.7 mg, 10.25%) as white powder. MS [M]+: 795.7. Example B18 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl] piperidin-1-ium-1-yl]acetic acid; formic acid; formate Step 1: tert-butyl 3-[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]methyl]azetidine-1-carboxylate In a 100 mL round-bottomed flask, N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (175 mg, 247 μmol), tert-butyl 3-formylazetidine-1-carboxylate (91.4 mg, 494 μmol) and NaBH3CN (77.5 mg, 1.23 mmol) were combined with MeOH (12 mL) to give a light brown solution. The reaction mixture was heated to 50° C. and stirred for 1 h. The crude reaction mixture was concentrated in vacuum. The reaction mixture was poured into 25 mL sat NaHCO3and extracted with EtOAc (3×25 mL). The organic layers were combined, washed with sat NaCl (1×25 mL), The organic layers were dried over Na2SO4and concentrated in vacuum to afford tert-butyl 3-[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]methyl]azetidine-1-carboxylate (217 mg). MS [M]+: 879.0. Step 2: tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]methyl]azetidine-1-carboxylate; bromide In a 50 mL round-bottomed flask, tert-butyl 3-[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]methyl]azetidine-1-carboxylate (108 mg, 123 μmol), tert-butyl 2-bromoacetate (36 mg, 184 μmol) and DIPEA (31.8 mg, 42.9 μl, 246 μmol) were combined with MeCN (3 mL) to give a light brown solution. The reaction mixture was heated to 40° C. and stirred for 15 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]methyl]azetidine-1-carboxylate; bromide (122 mg). MS [M]+: 993.1. Step 3: 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formic acid; formate In a 50 mL round-bottomed flask, tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]methyl]azetidine-1-carboxylate; bromide (122 mg, 123 μmol) was combined with THE (2 mL) to give a light brown solution. HCl water solution (1.53 ml, 18.4 mmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by preparative HPLC to afford 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formic acid; formate (23.7 mg). MS [M]+: 836.7. The following examples were prepared in analogy to Examples B18. MSESIStartingEx#NameStructure[M + H]+MaterialExample B19N-[3-chloro-4-[4-[1- [(1,1-dimethylazetidin-1- ium-3-yl)methyl]-1- methyl-piperidin-1-ium- 4-carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; diformate778.4Intermediate H1; tert-butyl 3- formylazetidine- 1-carboxylate and iodomethaneExample B202-[1-(azetidin-3- ylmethyl)-4-[4-[2-chloro- 4-[[5-[2,3-difluoro-4-[1- (2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperazine-1- carbonyl]piperidin-1- ium-1-yl]acetic acid; formic acid; formate836.7Intermediate H2; tert-butyl 3- formylazetidine- 1-carboxylate and tert-butyl 2-bromoacetateExample B21N-[4-[4-[1-(azetidin-3- yl methyl)-1-methyl- piperidin-1-ium-4- carbonyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid; formate792.1Intermediate H2; tert-butyl 3- formylazetidine- 1-carboxylate and iodomethaneExample B222-[1-(azetidin-3- ylmethyl)-4-[1-[2-chloro- 4-[[5-[2,3-difluoro-4-[1- (2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperidine-4- carbonyl]piperazin-1- ium-1-yl]acetic acid; formate836.2Intermediate H6; tert-butyl 3- formylazetidine- 1-carboxylate and tert-butyl 2-bromoacetateExample B23N-[4-[4-[4-(azetidin-3- ylmethyl)-4-methyl- piperazin-4-ium-1- carbonyl]piperidine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate792.2Intermediate H6; tert-butyl 3- formylazetidine- 1-carboxylate and iodomethaneExample B24N-[4-[4-[4-(2-amino-2- oxo-ethyl)-4-(azetidin-3- ylmethyl)piperazin-4- ium-1- carbonyl]piperidine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetate; 2,2,2- trifluoroacetic acid835.2Intermediate H6; tert-butyl 3- formylazetidine- 1-carboxylate and 2- iodoacetamideExample B25N-[3-chloro-4-[4- [(1R,5S)-3,3-dimethyl-3- azoniabicyclo[3.10]hexane- 6-carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate735.4Intermediate N1; HCl and iodomethaneExample B26N-[3-chloro-4-[4- [(1R,5S)-3,3-dimethyl-3- azoniabicyclo[3.10]hexane- 6-carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate735.4Intermediate N2; HCl and iodomethaneExample B27N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy-1-(3- hydroxypropyl)-1- methyl-pyrrolidin-1-ium- 2-carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; iodide783.3Intermediate K1; 3-[tert- butyl(dimethyl) silyl]oxypropanal and iodomethaneExample B402-[1-(azetidin-3- ylmethyl)-4-[4-[2-chloro- 4-[[5-[5-chloro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]-2-methyl- phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperazine-1- carbonyl]piperidin-1- ium-1-yl]acetic acid; formate848.3Intermediate H10; tert-butyl 3- formylazetidine- 1-carboxylate and tert-butyl 2-bromoacetate Example B28 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[3-chloro-2-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formate 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-(2-tert-butoxy-2-keto-ethyl)piperidin-1-ium-4-carboxylate (28 mg, 0.068 mmol, Intermediate R14) was dissolved in dichloromethane, extra dry (1 mL) (purged with Ar) and 5-[3-chloro-2-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-1-methyl-imidazole-2-carboxamide. 1:1 hydrogen chloride (30 mg, 0.045 mmol), Intermediate H11 was added. DIEA (34.78 mg, 47 uL, 0.269 mmol) and (7-azabenzotriazol-1-yloxy)tripyrrolidino-phosphonium hexafluorophosphate (33 mg, 0.063 mmol) were added to the reaction mixture which was then stirred at rt for 1.5 h under Ar. 4 M HCl (1.2 g, 1 mL, 4 mmol) in dioxane was added (slowly, reaction is slightly exothermic) and the mixture was stirred at rt for 30 min. The volatiles were removed under reduced pressure to give a colourless solid (in some brownish oil). The crude reaction mixture was purified by prepHPLC and lyophilized to get 10 mg of a colourless foam, purity (LC/MS, UV) 96%, yield 24%. MS: [M]+: 852.3. The following examples were prepared in analogy to Examples B28. MSESIStartingEx#NameStructure[M + H]+MaterialExample B372-[1-(azetidin-3- ylmethyl)-4-[4-[2-chloro- 4-[[5-[2-chloro-3-fluoro- 4-[1-(2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperazine-1- carbonyl]piperidin-1- ium-1-yl]acetic acid; formate852.3Intermediate H7 and Intermediate R14Example B392-[1-(azetidin-3- ylmethyl)-4-[4-[2-chloro- 4-[[5-[3-fluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]-2-methyl- phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperazine-1- carbonyl]piperidin-1- ium-1-yl]acetic acid; formate832.4Intermediate H9 and Intermediate R14 Example B29 & Example B30 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formic acid; formate (Example B29) 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formic acid; formate (Example B30) 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formic acid; formate (80 mg) was purified by preparative chial-HPLC to afford 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formic acid; formate (19.4 mg) and 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formic acid; formate (28.3 mg). MS [M]m: 836.0. Example B31 N-[3-chloro-4-[4-[1-[(1,1-dimethylazetidin-1-ium-3-yl)methyl]piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide Step 1: 3-(iodomethyl)-1,1-dimethyl-azetidin-1-ium In a 50 mL round-bottomed flask, 3-(iodomethyl)azetidine (126 mg, 640 μmol), Mel (454 mg, 3.2 mmol) and DIPEA (413 mg, 3.2 mmol) were combined with MeCN (6 mL) to give a light brown solution. The reaction was stirred at room temperature for 2 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford 3-(iodomethyl)-1,1-dimethylazetidin-1-ium (145 mg). MS [M]+: 225.9. Step 2: N-[3-chloro-4-[4-[1-[(1,1-dimethylazetidin-1-ium-3-yl)methyl]piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide In a 50 mL round-bottomed flask, N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (70 mg, 98.7 μmol), 3-(iodomethyl)-1,1-dimethyl-1l4-azetidine (22.3 mg, 98.7 μmol) and DIPEA (63.8 mg, 494 μmol) were combined with MeCN (6 mL) to give a light brown solution. The reaction mixture was heated to 50° C. and stirred for 15 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by preparative HPLC to afford Product 1 (17.8 mg). MS [M]+: 806.7. Example B32 N-[3-chloro-4-[4-[1-(2,2-difluoroethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate Step 1: N-[3-chloro-4-[4-[1-(2,2-difluoroethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide To a solution of N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (300 mg, 423 μmol) and DIPEA (164 mg, 1.27 mmol) in DCM (2 mL) was added slowly a solution of 2,2-difluoroethyl trifluoromethanesulfonate (272 mg, 1.27 mmol) in DCM (1 mL) at 0° C. The ice bath was removed after 1 h and the mixture was stirred for another 16 h. Then the solution was concentrated in vacuum to afford the crude product N-[3-chloro-4-[4-[1-(2,2-difluoroethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide. MS [M+H]+: 773.0. Step 2: N-[3-chloro-4-[4-[1-(2,2-difluoroethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate A mixture of N-[3-chloro-4-[4-[1-(2,2-difluoroethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (150 mg, 194 μmol), iodomethane (551 mg, 3.88 mmol) and DIPEA (501 mg, 3.88 mmol) in acetonitrile (5 mL) was stirred at room temperature for 48 h. Then the mixture was concentrated in vacuum and the residue was purified by Prep-HPLC to afford N-[3-chloro-4-[4-[1-(2,2-difluoroethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate (46 mg). MS [M]+: 787.2. Example B33 N-[3-chloro-4-[4-[1-(2-sulfamoylethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide Step 1: 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]ethanesulfonyl fluoride A mixture of N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (150 mg, 212 μmol) and ethenesulfonyl fluoride (27.9 mg, 254 μmol) in DMF (2 mL) was stirred at room temperature for 3 h. Then the mixture was heated at 50° C. for 1 h. The mixture was concentrated in vacuum to afford the crude material 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]ethanesulfonyl fluoride. The crude material was used into next step reaction without further purification. MS [M+H]*. 819.4. Step 2: N-[3-chloro-4-[4-[1-(2-sulfamoylethyl)piperidine-4-carbonyl]piperazine-1-carbonyl] phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl] phenyl]-1-methyl-imidazole-2-carboxamide At room temperature, 2-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]ethanesulfonyl fluoride was dissolved in DMF (2 mL). Then 7 M NH3 (3 mL) in methanol solution was added. The mixture was stirred for 16 h. Then the mixture was concentrated in vacuum. The crude material was purified by Prep-HPLC to afford N-[3-chloro-4-[4-[1-(2-sulfamoylethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (10 mg). MS [M+H]+: 816.1. Example B34 N-[3-chloro-4-[4-[1-methyl-1-(2-sulfamoylethyl)piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate At room temperature, iodomethane (2 mL) was added into a solution of N-[3-chloro-4-[4-[1-(2-sulfamoylethyl)piperidine-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (150 mg, 184 μmol) in DMF (2 mL). The mixture was stirred for 16 h. Then the mixture was purified by Prep-HPLC to afford N-[3-chloro-4-[4-[1-methyl-1-(2-sulfamoylethyl)piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (6 mg). MS [M]+: 830.4. Example B35 N-[3-chloro-4-[4-[1-(2-hydroxyethyl)-1-methyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate Step 1: N-[4-[4-[1-[2-[tert-butyl(dimethyl)silyl]oxyethyl]pyrrolidine-2-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide N-(3-chloro-4-(4-prolylpiperazine-1-carbonyl)phenyl)-5-(2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamide (83 mg, 130 μmol) and 2-((tert-butyldimethylsilyl)oxy)acetaldehyde (34.1 mg, 195 μmol) were stirred in dry THE (2.61 mL) before adding of NaBH(OAc)3(41.4 mg, 195 μmol). The resulting mixture was stirred at room temperature for 1 h. Silica gel was was added to absorb the material. The solid sample was purified by flash chromatography to afford the product (59 mg). MS [M]+: 795.4. Step 2: N-[3-chloro-4-[4-[1-(2-hydroxyethyl)-1-methyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate N-(4-(4-((2-((tert-butyldimethylsilyl)oxy)ethyl)prolyl)piperazine-1-carbonyl)-3-chlorophenyl)-5-(2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamide (59 mg, 74.2 μmol) and iodomethane (52.6 mg, 23.1 μL, 371 μmol) were stirred in MeCN (1.48 mL) for 5h. The solvent was removed in vacuum and the residue was treated with tetrabutylammonium fluoride (223 μL, 223 μmol, 1M in THF) at room temperature. After completion, the product was purified directly by prep. HPLC to afford the product (59 mg) as white powder. MS [M]+: 695.2. The following examples were prepared in analogy to Example B35. MSESIStartingEx#NameStructure[M]+MaterialExample B36N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy-1- (2-hydroxyethyl)-1- methyl-pyrrolidin-1- ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-(3- methyl-1H-pyrazol-4- yl)phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetate711.4Intermediate K10 and tert- butyl-(2- iodoethoxy)- dimethyl- silane; TBAF; iodomethane Example B38 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperazin-1-ium-1-yl]acetic acid; formate Step 1: tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperazin-1-ium-1-yl]methyl]azetidine-1-carboxylate; formate To a mixture of tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)piperazin-1-ium-1-yl]methyl]azetidine-1-carboxylate (100.0 mg, 0.270 mmol) Intermediate R15 and bis(trichloromethyl)carbonate (27.23 mg, 0.090 mmol) in DCM (2 mL) was stirred at 20° C. for 1 h. Then, N,N-diisopropylethylamine (0.1 mL, 0.590 mmol) and N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (161.41 mg, 0.270 mmol) in DCM (1 mL) was added to the above solution. The reaction mixture was stirred at 20° C. for 2 h. The reaction mixture was concentrated in vacuum to give the residue, which was purified by prep-HPLC (FA) to afford the title compound (100 mg, 0.100 mmol, 37.25% yield) as yellow solid. MS [M−56]+: 937.6. Step 2: 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperazin-1-ium-1-yl]acetic acid; formate To a solution of tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperazin-1-ium-1-yl]methyl]azetidine-1-carboxylate; formate (49.33 mg, 0.050 mmol, 1 eq) in DCM (1 mL) was added trifluoroacetic acid (1.0 mL, 12.98 mmol, 273.51 eq). The reaction mixture was stirred at 20° C. for 16 h. The reaction mixture was concentrated in vacuo. The residue was then purified by prep-HPLC (FA) to afford the title compound (19.3 mg, 0.020 mmol, 45.03% yield) as yellow solid. MS 837.2, [M]+, ESI+ Example C1 N-[3-chloro-4-[4-[2-(3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate Step 1: N-[3-chloro-4-[4-(2-chloroacetyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide In a 50 mL round-bottomed flask, N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (137 mg, 229 μmol) and DIPEA (88.8 mg, 687 μmol) were combined with MeCN (5 mL) to give a light yellow solution. 2-chloroacetyl chloride (51.7 mg, 458 μmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by flash chromatography to afford N-[3-chloro-4-[4-(2-chloroacetyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (150 mg). MS [M+H]+: 674.2. Step 2: N-[3-chloro-4-[4-[2-(3-hydroxypyrrolidin-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide To a 5 mL microwave vial was added N-[3-chloro-4-[4-(2-chloroacetyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (70 mg, 104 μmol), pyrrolidin-3-ol (18.1 mg, 208 μmol) and DIPEA (40.2 mg, 311 μmol) in MeCN (3 mL). The vial was capped and heated in the microwave at 70° C. for 30 min. The reaction was directly used the next step to afford N-[3-chloro-4-[4-[2-(3-hydroxypyrrolidin-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (75.3 mg). MS [M+H]+: 725.2. Step 3: N-[3-chloro-4-[4-[2-(3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate In a 50 mL round-bottomed flask, N-[3-chloro-4-[4-[2-(3-hydroxypyrrolidin-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (75.3 mg, 104 μmol), Mel (73.7 mg, 519 μmol) and DIPEA (67.1 mg, 519 μmol) were combined with MeCN (3 mL) to give a light yellow solution. The reaction mixture was heated to 40° C. and stirred for 2 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by preparative HPLC to afford N-[3-chloro-4-[4-[2-(3-hydroxy-1-methyl-pyrrolidin-1-ium-1-yl)acetyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (16.6 mg). MS [M]+: 739.3. The following examples were prepared in analogy to Examples C1. MSESIStartingEx#NameStructure[M]+MaterialExample C2N-[4-[4-[2-(3-amino-1- methyl-pyrrolidin-1-ium- 1-yl)acetyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid; formate738.4Intermediate I1; 2-chloroacetyl chloride; tert-butyl N-pyrrolidin-3- ylcarbamate and iodomethaneExample C3N-[4-[4-[2-(3-amino-1- methyl-pyrrolidin-1-ium- 1-yl)acetyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid; formate738.3Intermediate I2; 2-chloroacetyl chloride;tert-butyl N-pyrrolidin-3- ylcarbamate and iodomethaneExample C4N-[3-chloro-4-[4-[2-(3- hydroxy-1-methyl- pyrrolidin-1-ium-1- yl)acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide: formate739.2Intermediate I2; 2-chloroacetyl chloride; pyrrolidin- 3-ol and iodomethaneExample C5N-[4-[4-[2-[(3S,4S)-3- amino-4-methoxy-1- methyl-pyrrolidin-1-ium- 1-yl]acetyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid; formate768.0Intermediate I2; 2-chloroacetyl chloride; tert-butyl N-[(3S,4S)-4- methoxypyrrolidin- 3-yl]carbamate and iodomethaneExample C6N-[4-[4-[2-[3- (aminomethyl)-3- hydroxy-1-methyl- pyrrolidin-1-ium-1- yl]acetyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid; formate768.1Intermediate I2; 2-chloroacetyl chloride; tert-butyl N-[(3- hydroxypyrrolidin- 3- yl)methyl]carbamate and iodomethaneExample C7N-[4-[4-[2-(3-carbamoyl- 1-methyl-pyrrolidin-1- ium-1- yl)acetyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate766.4Intermediate I2; 2-chloroacetyl chloride; pyrrolidine- 3-carboxamide and iodomethaneExample C8N-[3-chloro-4-[4-[2- [(3R)-3-hydroxy-1- methyl-pyrrolidin-1-ium- 1-yl]acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate739.2Intermediate I2; 2-chloroacetyl chloride; rac-(3R)- pyrrolidin-3-ol and iodomethaneExample C9N-[3-chloro-4-[4-[2- [(3S)-3-hydroxy-1- methyl-pyrrolidin-1-ium- 1-yl]acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate739.2Intermediate I2; 2-chloroacetyl chloride; rac-(3S)- pyrrolidin-3-ol and iodomethaneExample C10N-[3-chloro-4-[4-[2- [(3R,4R)-3,4-dihydroxy- 1-methyl-pyrrolidin-1- ium-1- yl]acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate755.1Intermediate I2; 2-chloroacetyl chloride; rac- (3R,4R)-pyrrolidine- 3,4-diol and iodomethaneExample C11N-[3-chloro-4-[4-[2-(3- hydroxy-1-methyl- pyrrolidin-1-ium-1- yl)acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate753.1Intermediate I3; 2-chloroacetyl chloride; pyrrolidin- 3-ol and iodomethaneExample C12N-[3-chloro-4-[4-[2- [(3R,4R)-3,4-dihydroxy- 1-methyl-pyrrolidin-1- ium-1- yl]acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate769.2Intermediate I3; 2-chloroacetyl chloride; rac- (3R,4R)- pyrrolidine- 3,4-diol and iodomethaneExample C13N-[4-[4-[2-[(3R,4R)-3- amino-4-methoxy-1- methyl-pyrrolidin-1-ium- 1-yl]acetyl]piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)- 3,5-dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid; formate782.1Intermediate I3; 2-chloroacetyl chloride; rac- (3R,4R)-4- methoxypyrrolidin- 3-amine and iodomethaneExample C14N-[3-chloro-4-[4-[2-(1- methylpyrrolidin-1-ium- 1-yl)acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; 2,2,2- trifluoroacetate723.3Intermediate I-1; 2-(pyrrolidin-1- yl)acetic acid and iodomethane Example D1 N-[3-chloro-4-[4-[(2S,3S)-3-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate Step 1: tert-butyl (2S,3S)-2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-hydroxy-pyrrolidine-1-carboxylate In a 50 mL round-bottomed flask, (2S,3S)-1-tert-butoxycarbonyl-3-hydroxy-pyrrolidine-2-carboxylic acid (40.2 mg, 174 μmol), N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (80 mg, 134 μmol), HATU (66.1 mg, 174 μmol) and DIPEA (17.3 mg, 134 μmol) were combined with DMF (3 mL) to give a light brown solution. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford tert-butyl (2S,3S)-2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-hydroxy-pyrrolidine-1-carboxylate (100 mg). MS [M+H]+: 811.3. Step 2: N-[3-chloro-4-[4-[(2S,3S)-3-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide In a 50 mL round-bottomed flask, tert-butyl (2S,3S)-2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]-piperazine-1-carbonyl]-3-hydroxy-pyrrolidine-1-carboxylate (100 mg, 123 μmol) was combined with THE (2 mL) to give a light brown solution. HCl (1.03 ml, 12.3 mmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford N-[3-chloro-4-[4-[(2S,3S)-3-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (87.7 mg). MS [M+H]+: 711.2. Step 3: N-[3-chloro-4-[4-[(2S,3S)-3-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate In a 50 mL round-bottomed flask, tert-butyl (2S,3S)-2-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-3-hydroxy-pyrrolidine-1-carboxylate (87 mg, 122 μmol), Mel (86.8 mg, 612 μmol) and DIPEA (79.1 mg, 612 μmol) were combined with MeCN (5 mL) to give a light red solution. The reaction mixture was heated to 40° C. and stirred for 15 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by preparative HPLC to afford N-[3-chloro-4-[4-[(2S,3S)-3-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (16.1 mg). MS [M]+: 739.1. The following examples were prepared in analogy to Examples D1. MSESIStartingEx#NameStructure[M]+MaterialExample D2N-[3-chloro-4-[4-(3- hydroxy-1,1-dimethyl- piperidin-1-ium-4- carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate753.2Intermediate I2; 1-tert- butoxycarbonyl- 3-hydroxy- piperidine-4- carboxylic acid; iodomethaneExample D3N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate753.4Intermediate I3; rac-(2S,4R)- 1-tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; iodomethaneExample D4N-[3-chloro-4-[4-(3- hydroxy-1,1-dimethyl- piperidin-1-ium-4- carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3 difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate767.3Intermediate I3; 1-tert- butoxycarbonyl- 3-hydroxy- piperidine-4- carboxylic acid; iodomethaneExample D6N-[3-chloro-4-[4-[3- (hydroxymethyl)-4,4- dimethyl-piperazin-4- ium-1- carbonyl]piperidine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate767.2Intermediate I6; tert-butyl 2- (hydroxymethyl) piperazine-1- carboxylate; iodomethaneExample D7N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate739.2Intermediate I2; (2S,4R)-1- tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; iodomethaneExample D8N-[3-chloro-4-[4- [(2R,4S)-4-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate739.2Intermediate I2; (2R,4S)-1- tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; iodomethaneExample D9N-[3-chloro-4-[4- [(2R,4R)-4-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate739.2Intermediate I2; (2R,4R)- 1-tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; iodomethaneExample D10N-[3-chloro-4-[4- [(2S,4S)-4-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate739.2Intermediate I2; (2S,4S)-1- tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; iodomethaneExample D11N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(3- methoxypropyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate753.1Intermediate I8; (2S,4R)-1- tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; iodomethaneExample D12N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(3- methoxypropyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate753.4Intermediate I9; (2S,4R)-1- tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; iodomethaneExample D13N-[3-chloro-4-[4-[(3S)- 4,4-dimethylmorpholin- 4-ium-3- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(3- methoxypropyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate753.4Intermediate I2; (S)-4-(tert- butoxycarbonyl) morpholine-3- carboxylic acid; iodomethaneExample D14N-[3-chloro-4-[4-[(4,4- dimethyl-2-oxo- piperazin-4-ium-1- yl)methyl]piperidine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; 2,2,2- trifluoroacetate737.2Intermediate L2; intermediate R5; iodomethaneExample D15N-[3-chloro-4-[4-[2-(1- methylpyrrolidin-1-ium- 1-yl)acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-(3-methyl-1H- pyrazol-4-yl)phenyl]-1- methyl-imidazole-2- carboxamide; formate665.2Intermediate M1; 2- (pyrrolidin-1- yl)acetic acid; iodomethaneExample D16N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-(3-methyl-1H- pyrazol-4-yl)phenyl]-1- methyl-imidazoIe-2- carboxamide; formate679.3Intermediate M1 and 1-tert- butoxycarbonyl- piperidine-4- carboxylic acid; then iodomethaneExample D17N-[3-chloro-4-[4-[2-(4- hydroxy-1,1-dimethyl- piperidin-1-ium-4- yl)acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate767.3Intermediate I2; 2-(1-tert- butoxycarbonyl- 4-hydroxy-4- piperidyl)acetic acid; iodomethaneExample D18N-[3-chloro-4-[4-[2-[4- hydroxy-4- (hydroxymethyl)-1- methyl-piperidin-1-ium- l-yl]acetyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-3-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate783.4Intermediate L3; Intermediate R7; iodomethaneExample D19N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy-1,1- dimethyl-pyrrolidin-1- ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-(3-methyl-1H- pyrazol-4-yl)phenyl]-1- methyl-imidazole-2- carboxamide; formate681.3Intermediate M1 and Intermediate R2; HCl; iodomethaneExample D20N-[4-[4-(4-amino-1,1- dimethyl-piperidin-1- ium-4- carbonyl)piperazine-1- carbonyl]-3-chloro- phenyl]-5-[2,3-difluoro- 4-[1-(2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate752.3Intermediate I1 and Intermediate R10; HClExample D21N-[3-chloro-4-[4-[(4,4- dimethyl-2-oxo- piperazin-4-ium-1- yl)methyl]piperidine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-(3-methyl-1H- pyrazol-4-yl)phenyl]-1- methyl-imidazole-2- carboxamide; formate679.3Intermediate L1; intermediate R5; iodomethaneExample D35[2-[4-[2-chloro-4-[[5- [2,3-difluoro-4-(3- methyl-1H-pyrazol-4- yl)phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperazin-1-yl]-2-oxo- ethyl]-trimethyl- ammonium; formate639.4Example D34 and iodomethaneExample D37N-[3-chloro-4-[4-[(1,1- dimethylpiperidin-1-ium-4- yl)sulfonylamino]piperidine- 1-carbonyl]phenyl]- 5-[2,3-difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate787.3Intermediate I10 and tert- butyl 4- chlorosulfonyl- piperidine-1- carboxylate; HCl; iodomethaneExample D38N-[3-chloro-4-[[(1R,5S)- 3-[(2S,4R)-4-hydroxy- 1,1-dimethyl-pyrrolidin- 1-ium-2-carbonyl]-3- azabicyclo[3.10]hexan- 6-yl]carbamoyl]phenyl]- 5-[2,3-difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate765.3Intermediate I11 and (2S,4R)-1-tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; HCl; iodomethaneExample D40N-[3-chloro-4-[[(1S,5R)- 3-[(2S,4R)-4-hydroxy- 1,1-dimethyl-pyrrolidin- 1-ium-2-carbonyl]-3- azabicyclo[3.10]hexan- 6-yl]carbamoyl]phenyl]- 5-[2,3-difluoro-4-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate751.3Intermediate I12 and (2S,4R)-1-tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; HCl; iodomethaneExample D41bis(3-aminopropyl)- (carboxymethyl)-[4-[4- [2-chloro-4-[[5-[2,3- difluoro-4-(3-methyl-1H- pyrazol-4-yl)phenyl]-1- methyl-imidazole-2- carbonyl]amino]benzoyl] piperazin-1-yl]-4-oxo- butyl]ammonium; formic acid; formate797.4Intermediate M1 and Intermediate R11: HClExample D42bis(azetidin-3-ylmethyl)- (carboxymethyl)-[4-[4- [2-chloro-4-[[5-[2,3- difluoro-4-(3-methyl-1H- pyrazol-4-yl)phenyl]-1- methyl-imidazoIe-2- carbonyl]amino]benzoyl] piperazin-1-yl]-4-oxo- butyl]ammonium; formic acid; formate821.5Intermediate M1 and Intermediate R12: HCl Example D22 5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-N-[4-[4-(3-hydroxypiperidine-4-carbonyl)piperazine-1-carbonyl]-3-methyl-phenyl]-1-methyl-imidazole-2-carboxamide; formic acid At room temperature, a mixture of 5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-N-[3-methyl-4-(piperazine-1-carbonyl)phenyl]imidazole-2-carboxamide (300 mg, 519 μmol), 1-tert-butoxycarbonyl-3-hydroxy-piperidine-4-carboxylic acid (191 mg, 779 μmol), HATU (296 mg, 779 μmol) and DIPEA (201 mg, 1.56 mmol) in DMIF (2 mL) was stirred for 16 h. Then the mixture was poured into water. The water layer was extracted with DCM. The combined organic layers were washed with water, dried over anhydrous Na2SO4and concentrated in vacuum. The crude product tert-butyl 4-[4-[4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-methyl-benzoyl]piperazine-1-carbonyl]-3-hydroxy-piperidine-1-carboxylate was used into next step reaction directly. MS [M+H]+: 805.5. Step 2: 5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-N-[4-[4-(3-hydroxypiperidine-4-carbonyl)piperazine-1-carbonyl]-3-methyl-phenyl]-1-methyl-imidazole-2-carboxamide; formic acid At room temperature, a solution of tert-butyl 4-[4-[4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-methyl-benzoyl]piperazine-1-carbonyl]-3-hydroxy-piperidine-1-carboxylate (400 mg, 497 μmol) in DCM (10 mL) and TFA (5 mL) was stirred for 1 h. Then the mixture was concentrated in vacuum. The residue was basified by NH3·H2O to PH 8-9. The water layer was extracted with DCM. The organic layer was dried over anhydrous Na2SO4and concentrated in vacuum. The residue was purified by Prep-HPLC to afford 5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-N-[4-[4-(3-hydroxypiperidine-4-carbonyl)piperazine-1-carbonyl]-3-methyl-phenyl]-1-methyl-imidazole-2-carboxamide; formic acid. MS [M+H]+: 705.4. The following examples were prepared in analogy to Example D22. MSESIStartingEx#NameStructure[M + H]+MaterialExample D23N-[3-chloro-4-[4- [[(2S,4R)-4- hydroxypyrrolidine-2- carbonyl]amino] piperidine-1- carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid725.3Intermediate I5 and (2S,4R)-1- tert- butoxy- carbonyl-4- hydroxy- pyrrolidine-2- carboxylic acid; HClExample D24N-[3-chloro-4-[4-(4- hydroxypiperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid725.2Intermediate I1 and 1-(tert- butoxy- carbonyl)-4- hydroxy- piperidine-4- carboxylic acid; HClExample D25N-[3-chloro-4-[4-(3- hydroxypiperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid725.2Intermediate I1 and 1-(tert- butoxy- carbonyl)-3- hydroxy- piperidine-4- carboxylic acid; HClExample D26N-[3-chloro-4-[4- [(2S)-5- oxopyrrolidine-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetic acid709.5Intermediate I1 and (S)-5- oxopyrrolidine-2- carboxylic acidExample D27N-[3-chloro-4-[4-(2- oxopiperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetic acid723.3Intermediate I1 and 2- oxopiperidine-4- carboxylic acidExample D28N-[3-chloro-4-[4-(2- pyrrolidin-1- ylacetyl)piperazine-1- carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetic acid709.2Intermediate I1 and 2- (pyrrolidin-1- yl)acetic acidExample D29N-[3-chloro-4-[4- (pyrrolidine-2- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetic acid695.3Intermediate I1 and (tert- butoxy- carbonyl)proline; HClExample D30N-[4-[4-(3- aminobicyclo[1.1.1] pentane-1- carbonyl)piperazine- 1-carbonyl]-3-chloro- phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetic acid707.8Intermediate I1 and 3-(tert- butoxycarbonyl- amino)bicyclo [1.1.1]pentane- 1-carboxylic acid; HClExample D31N-[3-chloro-4-[4- [(2S,4R)-4- hydroxypyrrolidine-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formic acid711.3Intermediate I2 and (2S,4R)-1- tert- butoxycarbonyl- 4-hydroxy- pyrrolidine-2- carboxylic acid; HClExample D32N-[3-chloro-4-[4-[2- (dimethylamino)acetyl] piperazine-1- carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetic acid683.4Intermediate I1 and dimethylglycineExample D33N-[3-chloro-4-[[1-[2- (dimethyl- amino)acetyl]-4- piperidyl]methyl- carbamoyl]phenyl]-5- [2,3- difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetic acid711.3Intermediate I7 and dimethylglycineExample D34N-[3-chloro-4-[4-[2- (dimethylamino)acetyl] piperazine-1- carbonyl]phenyl]-5- [2,3-difluoro-4-(3- methyl-1H-pyrazol-4- yl)phenyl]-1-methyl- imidazole-2- carboxamide; formic acid625.2Intermediate M1 and dimethylglycineExample D36N-[3-chloro-4-[4- [(2S,4R)-4- hydroxypyrrolidine-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-(3- methyl-1H-pyrazol-4- yl)phenyl]-1-methyl- imidazole-2- carboxamide653.2Intermediate M1; Intermediate R3, and TFAExample D44N-[3-chloro-4-[4-(4- methoxypiperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide739.2Intermediate I1 and 1-(tert- butoxy- carbonyl)-4- methoxy- piperidine-4- carboxylic acid and HCl Example D43 N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide; formate To a mixture of 2-chloro-4-[[5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoic acid (105.0 mg, 0.220 mmol) in THE (5 mL) and DMF (5 mL) was added [(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-yl]-piperazin-1-yl-methanone (49.34 mg, 0.220 mmol), N,N-diisopropylethylamine (0.4 mL, 2.3 mmol) and propylphosphonic anhydride (438.99 mg, 1.38 mmol). The reaction mixture was stirred at 25° C. for 16 h. The mixture was concentrated to remove solvent, purified by prep-HPLC (0.1% FA)-ACN to afford N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide; formate (2.5 mg). MS [M]+: 695.3. Example E1 N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-(2,2-difluoroethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide Step 1: tert-butyl 4-[4-[2-chloro-4-[[5-[4-[1-(2,2-difluoroethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate To a 25 mL microwave vial was added tert-butyl 4-[4-[4-[(5-bromo-1-methyl-imidazole-2-carbonyl)amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (48 mg, 75.2 μmol), 4-(2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(2,2-difluoroethyl)-5-methyl-1H-pyrazole (31.8 mg, 82.8 μmol), 4-(2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(2,2-difluoroethyl)-5-methyl-1H-pyrazole (31.8 mg, 82.8 μmol) and Na2CO3(23.9 mg, 226 μmol) in 1,4-Dioxane (10 mL)/Water (1 mL). The vial was capped and heated in the microwave at 100° C. for 3 h under N2. The crude reaction mixture was concentrated in vacuum. The crude material was purified by flash chromatography to afford tert-butyl 4-[4-[2-chloro-4-[[5-[4-[1-(2,2-difluoroethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (26 mg). MS [M−100]+: 715.2. Step 2: N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-(2,2-difluoroethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide In a 50 mL round-bottomed flask, tert-butyl 4-(4-(2-chloro-4-(5-(4-(1-(2,2-difluoroethyl)-5-methyl-1H-pyrazol-4-yl)-2,3-difluorophenyl)-1-methyl-1H-imidazole-2-carboxamido)benzoyl)piperazine-1-carbonyl)piperidine-1-carboxylate (26 mg, 31.9 μmol) was combined with THE (2 mL) to give a light brown solution. HCl water solution (797 μL, 9.57 mmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by preparative HPLC to afford N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-(2,2-difluoroethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide (1.4 mg). MS [M+H]+: 715.4. The following examples were prepared in analogy to Examples E1. MSESIStartingEx#NameStructure[M + H]+MaterialExample E2N-[3-chloro-4- [4-[2-[(3S)- pyrrolidin-3-yl] acetyl]piperazine- 1-carbonyl]phenyl]- 5-[2,3-difluoro- 4-[1-(2-methoxy- ethyl)-5-methyl- pyrazol-4- yl]phenyl]- 1-methyl- imidazole-2- carboxamide; formic acid709.3Intermediate D2; Intermediate G3 and HClExample E3N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2-fluoro- 4-[1-(2-methoxy- ethyl)-5-methyl- pyrazol-4-yl]- 3-methyl- phenyl]-1- methyl-imidazole- 2-carboxamide; formic acid705.4Intermediate D1; Intermediate G10 and HClExample E45-[2-chloro- 3-fluoro-4- [1-(2-methoxy- ethyl)-5-methyl- pyrazol-4-yl] phenyl]-N-[3- chloro-4-[4- (piperidine-4- carbonyl) piperazine-1- carbonyl]phenyl]- 1-methyl- imidazole-2- carboxamide; formic acid725.2Intermediate D1; Intermediate G11 and HClExample E5N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[3-fluoro- 4-[1-(2-methoxy- ethyl)-5-methyl- pyrazol-4-yl]- 2-methyl- phenyl]-1-methyl- imidazole-2- carboxamide; formic acid705.4Intermediate D1; Intermediate G13 and HClExample E6N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[4-[1-[2- (difluoro- methoxy)ethyl]- 3-(trifluoro- methyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formic acid799.2Intermediate D1; Intermediate G46 and TFAExample E7N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]- 5-[2,3-difluoro- 4-[1-(2-methoxy- ethyl)-5-methyl- pyrazol-4-yl] phenyl]-1- methyl-imidazole- 2-carboxamide; 2,2,2- trifluoroacetic acid598.4Intermediate B1; Intermediate G3 and HClExample E85-[2,3-difluoro- 4-[1-(2-methoxy- ethyl)-5-methyl- pyrazol-4-yl] phenyl]-N- [4-[4-[(2S,4R)- 4-hydroxy- pyrrolidine-2- carbonyl] piperazine-1- carbonyl]-3-methyl- phenyl]-1-methyl- imidazole-2- carboxamide691.4Intermediate D4; Intermediate G3 and HClExample E9N-[3-chloro-4- [4-[(2S,4R)-4- hydroxy- pyrrolidine-2- carbonyl] piperazine-1- carbonyl]phenyl]- 5-[2,3- difluoro-4-[1-(2- methoxyethyl)- 5-methyl- pyrazol-4-yl] phenyl]-1- methyl-imidazole- 2-carboxamide; 2,2,2- trifluoroacetic acid711.3Intermediate D6; Intermediate G3 and HClExample E10N-[3-chloro-4- (4-piperidyl- methyl-carbamoyl) phenyl]-5-[2,3- difluoro-4-[1-(2- methoxyethyl)- 5-methyl- pyrazol-4-yl] phenyl]-1- methyl-imidazole- 2-carboxamide; 2,2,2- trifluoroacetic acid626.4Intermediate B5; Intermediate G3 and HClExample E11N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3- difluoro-4-[1-(2- methoxyethyl)- 5-methyl- pyrazol-4-yl] phenyl]-1- methyl-imidazole- 2-carboxamide; 2,2,2- trifluoroacetic acid709.3Intermediate D1; Intermediate G3 and HClExample E12N-[4-[4-[1-(2- amino-2-oxo-ethyl) piperidine-4- carbonyl] piperazine-1- carbonyl]-3-chloro- phenyl]-5-[3- fluoro-4-[3- (trifluoromethyl)- 1H-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide; 2,2,2- trifluoroacetic acid744.2Intermediate G46; Intermediate P1 and HClExample E13N-[3-chloro-4-[4- (piperidine-4- carbonyl)piperazine- 1-carbonyl]phenyl]- 5-[2,3-difluoro- 4-[1-(2-methoxy- ethyl)-5-(methoxy- methyl)pyrazol- 4-yl]phenyl]- 1-methyl- imidazole-2- carboxamide; formic acid739.3Intermediate D1; Intermediate G25 and HClExample E14N-[4-[4-[2- (dimethylamino) acetyl] piperazine-1- carbonyl]-3- methyl-phenyl]- 5-[4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide627.4Intermediate D5; Intermediate G12Example E15N-[3-chloro-4- [4-[2-(dimethyl- amino)acetyl] piperazine-1- carbonyl]phenyl]- 1-methyl-5-[4- (3-methyl-1H- pyrazol-4-yl) phenyl]imidazole- 2-carboxamide; formic acid589.4Intermediate J1; TFAExample E16N-[3-chloro-4- [4-[2-(dimethyl- amino)acetyl] piperazine-1- carbonyl]phenyl]- 5-[4-(3,5- dimethyl-1H- pyrazol-4- yl)phenyl]-1- methyl-imidazole- 2-carboxamide603.4Intermediate J2 and TFAExample E17N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2-fluoro- 6-[5-(4-methoxy- phenyl)-1H- pyrazol-4-yl]- 3-pyridyl]- 1-methyl- imidazole-2- carboxamide; formic acid726.3Intermediate D1 and Intermediate G75; HClExample E18N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2-fluoro- 6-(1H-pyrazol-4- yl)-3-pyridyl]- 1-methyl- imidazole-2- carboxamide620.2Intermediate D1 and Intermediate G81; HClExample E19N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro- 4-(3-methyl-1H- pyrazol-4- yl)phenyl]-1- methyl-imidazole- 2-carboxamide651.1Intermediate D1 and Intermediate G1; HClExample E20N-[3-chloro- 4-[4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3-difluoro- 4-(1-methyl- pyrazol-4- yl)phenyl]-1- methyl-imidazole- 2-carboxamide651.1Intermediate D1 and Intermediate G82; HClExample E21N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 5-[2,3- difluoro-4-[1-(2- methoxyethyl) pyrazol-4-yl] phenyl]-1-methyl- imidazole-2- carboxamide695.2Intermediate D1 and Intermediate G83; HClExample E22(1S,5R)-6-[[2- chloro-4-[[5- [2,3-difluoro-4- [1-(2-methoxy- ethyl)-5-methyl- pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carbonyl] amino]benzoyl] amino]-N- [(3R,4R)-4- hydroxypyrrolidin- 3-yl]-3- azabicyclo[3.1.0] hexane-3- carboxamide738.2Intermediate D11 and Intermediate G3; HClExample E23N-[3-chloro-4- [4-(piperidine- 4-carbonyl) piperazine-1- carbonyl]phenyl]- 1-methyl-5-[4- (1H-pyrazol-4- yl)phenyl] imidazole-2- carboxamide601.2Intermediate D1 and Intermediate G90; HCl Example F1 5-[4-[1-(3-amino-3-oxo-propyl)-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide; formate Step 1: tert-butyl 4-[4-[4-[[5-[4-[1-(3-amino-3-oxo-propyl)-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate 3-(4-(2,3-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)propanamide (100 mg, 225 μmol), tert-butyl 4-(4-(4-(5-bromo-1-methyl-1H-imidazole-2-carboxamido)-2-chlorobenzoyl)piperazine-1-carbonyl)piperidine-1-carboxylate (129 mg, 202 μmol), sodium carbonate (71.4 mg, 674 μmol) and Pd-118 (29.3 mg, 44.9 μmol) were placed in water (408 μL) and dioxane (4.08 ml) in microwave tube. The tube was evacuated and backfilled with argon for 5 times. The mixture was then heated at 100° C. for 1 h. The mixture was cooled to room temperature, 100-200 mesh silica gel was added to absorb the material. The loaded sample was purified by flash chromatography to afford the product (98 mg). MS [M+H]+: 876.3. Step 2: 5-[4-[1-(3-amino-3-oxo-propyl)-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide tert-butyl 4-(4-(4-(5-(4-(1-(3-amino-3-oxopropyl)-3-(trifluoromethyl)-1H-pyrazol-4-yl)-2,3-difluorophenyl)-1-methyl-1H-imidazole-2-carboxamido)-2-chlorobenzoyl)piperazine-1-carbonyl)piperidine-1-carboxylate (98 mg, 112 μmol) was dissolved in 2 mL 20% TFA/DCM solution and the resulting solution was stirred for 1 h at room temperature. After completion, the solvent was removed in vacuo to afford the product which was used without purification (70 mg). MS [M+H]+: 776.2. Step 3: 5-[4-[1-(3-amino-3-oxo-propyl)-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide; formate 5-[4-[1-(3-amino-3-oxo-propyl)-3-(trifluoromethyl)pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide (70 mg) was dissolved in DMA (2.24 mL), to this solution was added N-ethyl-N-isopropylpropan-2-amine (72.3 mg, 559 μmol) and iodomethane (31.7 mg, 224 μmol). The solution was stirred at rt for 2 h. After completion, the product was purified by preparative HPLC directly to afford the product (28.9 mg). MS [M]+: 804.3. The following examples were prepared in analogy to Example F1. MSESIStartingEx#NameStructure[M]+MaterialExample F2N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2-chloro-3-fluoro-4- [1-(2-methoxyethyl)- 5-methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate753.3Intermediate D1 and Intermediate G11; HCl; iodomethane.Example F3N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [3-fluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4-yl]- 2-methyl-phenyl]-1- methyl-imidazole-2- carboxamide; formate733.4Intermediate D1 and Intermediate G13; HCl; iodomethane.Example F4N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(3- hydroxy-3-methyl- butyl)-5-methyl- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate767.3Intermediate D6 and Intermediate G14; HCl; iodomethane.Example F5N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2- (methylamino)-2-oxo- ethyl]pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate752.3Intermediate D6 and Intermediate G15; HCl; iodomethane.Example F6N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2- (difluoromethoxy) ethyl]-3-(trifluoro- methyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate827.2Intermediate D1 and Intermediate G48; TFA; iodomethane.Example F7N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(3- hydroxy-2-methyl- propyl)-3-(trifluoro- methyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate805.3Intermediate D1 and Intermediate G49; TFA; iodomethane.Example F8N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-[(2S)-2,3- dihydroxypropyl]-3- (trifluoro- methyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate807.3Intermediate D1 and Intermediate G45; HCl; iodomethane.Example F9N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2-(difluoro- methoxy)ethyl]- 3-methyl-pyrazol-4- yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate775.3Intermediate D6 and Intermediate G33; HCl; iodomethane.Example F10N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2-(difluoro- methoxy)ethyl]- 5-methyl-pyrazol-4- yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate775.3Intermediate D6 and Intermediate G34; HCl; iodomethane.Example F11N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- isopropyl-1-(2- methoxyethyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate767.3Intermediate D6 and Intermediate G35; HCl; iodomethane.Example F12N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [4-[5- (difluoromethyl)-1-(2- methoxyethyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate775.3Intermediate D6 and Intermediate G36; HCl; iodomethane.Example F145-[2,3-difluoro-4-[1- (2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-N-[4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]-3-methyl- phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetate719.4Intermediate D4 and Intermediate G3; HCl; iodomethane.Example F15N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; 2,2,2- trifluoroacetate739.4Intermediate D6 and Intermediate G3; HCl; iodomethane.Example F16N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(3- hydroxypropyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate739.3Intermediate D6 and Intermediate G19; HCl; iodomethane.Example F17N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(3- hydroxypropyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate739.3Intermediate D6 and Intermediate G20; HCl; iodomethane.Example F18N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2-methoxy-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate731.2Intermediate D1 and Intermediate G21; HCl; iodomethane.Example F19N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4-yl]- 2-methyl-phenyl]-1- methyl-imidazole-2- carboxamide; formate715.2Intermediate D1 and Intermediate G22; HCl; iodomethane.Example F20N-[3-chloro-4-[4- [(2S,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine- 1-carbonyl]phenyl]-5- [4-[5-ethyl-1-(2- methoxyethyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate753.3Intermediate D6 and Intermediate G23; HCl; iodomethane.Example F21N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[5-(4- hydroxybutyl)-1H- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate737.2Intermediate D1 and Intermediate G24; HCl; iodomethane.Example F22N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2-chloro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate737.3Intermediate D1 and Intermediate G28; HCl; iodomethane.Example F23N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [3-fluoro-2-methoxy- 4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate749.5Intermediate D1 and Intermediate G29; HCl; iodomethane.Example F24N-[3-chloro-4-[4-[2- (4-hydroxy-1,1- dimethyl-piperidin-1- ium-4- yl)acetyl]piperazine- 1-carbonyl]phenyl]-5- [4-[3-ethyl-1-(2- methoxyethyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate781.4Intermediate D8 and Intermediate G50; HCl; iodomethane.Example F25N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- (hydroxymethyl)-1-(2- methoxyethyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate753.6Intermediate D1 and Intermediate G51; HCl; iodomethane.Example F265-[4-[3-amino-1-(2- methoxyethyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-N-[3-chloro- 4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate738.4Intermediate D1 and Intermediate G52; HCl; iodomethane.Example F27N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-(3- phenyl-1H-pyrazol-4- yl)phenyl]-1-methyl- imidazole-2- carboxamide; formate741.5Intermediate D1 and Intermediate G53; HCl; iodomethane.Example F28N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-(5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol- 3-yl)-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate705.1Intermediate D1 and Intermediate G54; HCl; Iodomethane.Example F29N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- (fluoromethyl)-1-(2- methoxyethyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; chloride755.3Intermediate D1 and Intermediate G55; HCl; Iodomethane.Example F30N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[3-chloro-1-(2- methoxyethyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate757.3Intermediate D1 and Intermediate G56; HCl; Iodomethane.Example F31N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[5-chloro-1-(2- methoxyethyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate757.3Intermediate D1 and Intermediate G57; iodomethane.Example F32N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methylsulfonylethyl)- 3-(trifluoro- methyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate839.2Intermediate D1 and intermediate G39; then TFA and iodomethane.Example F33N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- hydroxypropyl)-3- (trifluoromethyl) pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate791.3Intermediate D1 and intermediate G47; then TFA and iodomethane.Example F345-[4-[1-(4-amino-4- oxo-butyl)-3-(trifluoro- methyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-N-[3-chloro- 4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate818.3Intermediate D1 and intermediate G40; then TFA and iodomethane.Example F355-[4-[1-(2-amino-2- oxo-ethyl)-3- (trifluoromethyl)pyrazol- 4-yl]-2,3-difluoro- phenyl]-N-[3-chloro- 4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate790.4Intermediate D1 and intermediate G41; then TFA and iodomethane.Example F36N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-3- (trifluoromethyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate791.2Intermediate D1 and intermediate G42; then TFA and iodomethane.Example F37N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- morpholinoethyl)-5- (trifluoromethyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate846.3Intermediate D1 and intermediate G43; then TFA and iodomethane.Example F38N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(3- methoxypropyl)-3- (trifluoromethyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate805.4Intermediate D1 and intermediate G44; then TFA and iodomethane.Example F39N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1- (tetrahydropyran-4- ylmethyl)pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate777.1Intermediate D1 and intermediate G31; TFA; iodomethane.Example F405-[4-[1-(5-amino-2- pyridyl)-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-N- [3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; chloride771.2Intermediate D1 and intermediate G70; HCl; iodomethane.Example F41N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-(2- pyridylmethyl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate770.3Intermediate D1 and intermediate G32; HCl; iodomethaneExample F42N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-(1H- pyrazol-4-yl)pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate745.3Intermediate D1 and intermediate G71; HCl; iodomethaneExample F435-[4-[1-[(5-amino-2- pyridyl)methyl]-3- methyl-pyrazol-4-yl]- 2,3-difluoro-phenyl]- N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; chloride785.5Intermediate D1 and intermediate G72; HCl; iodomethaneExample F44N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3-(2- methylpyrazol-3-yl)- 1H-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate745.4Intermediate D1 and intermediate G65; HCl; iodomethaneExample F45N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3-(3- fluorophenyl)-1H- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate759.3Intermediate D1 and intermediate G66; HCl; iodomethaneExample F46N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-(2-oxo-1- pyridyl)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate800.6Intermediate D1 and intermediate G73; HCl; iodomethaneExample F47N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3-(1- methylpyrazol-4-yl)- 1H-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate745.2Intermediate D1 and intermediate G67; HCl; iodomethaneExample F48N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- (1H-pyrazol-4-yl)-1H- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate731.2Intermediate D1 and intermediate G68; HCl; iodomethaneExample F49N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-3- phenyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate799.4Intermediate D1 and intermediate G69; HCl; iodomethaneExample F50N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-(2- pyridyl)pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate756.2Intermediate D1 and intermediate G74; HCl; iodomethaneExample F51[2-[4-[2-chloro-4-[[1- methyl-5-[4-(3- methyl-1H-pyrazol-4- yl)phenyl]imidazole-2- carbonyl]amino]benzoyl] piperazin-1-yl]-2- oxo-ethyl]-trimethyl- ammonium; formate603.4Intermediate D3 and intermediate G8; HCl; iodomethaneExample F52[2-[4-[2-chloro-4-[[5- [4-(3,5-dimethyl-1H- pyrazol-4-yl)phenyl]- 1-methyl-imidazole-2- carbonyl]amino]benzoyl] piperazin-1-yl]-2- oxo-ethyl]-trimethyl- ammonium; 2,2,2- trifluoroacetate617.4Intermediate D3 and intermediate G9; HCl; iodomethaneExample F534-chloro-N-[3-chloro- 4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate773.4Intermediate D10 and intermediate G3; HCl; iodomethaneExample F54N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[2- (6-methoxy-2- pyridyl)ethyl]-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate814.3Intermediate D1 and intermediate G76; HCl; iodomethaneExample F55N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[(6- methoxy-2- pyridyl)methyl]-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate800.3Intermediate D1 and intermediate G77; HCl; iodomethaneExample F56N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-(1H-triazol- 4-ylmethyl)pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate760.3Intermediate D1 and intermediate G78; HCl; iodomethaneExample F57N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2-fluoro-6-[1-(2- methoxyethyl)-5- methyl-pyrazol-4-yl]- 3-pyridyl]-1-methyl- imidazole-2- carboxamide; formate720.2Intermediate D1 and intermediate G79; HCl; iodomethaneExample F59N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2-fluoro-6-[5-(4- methoxyphenyl)-1H- pyrazol-4-yl]-3- pyridyl]-1-methyl- imidazole-2- carboxamide; formate754.3Intermediate D1 and intermediate G75; HCl; iodomethaneExample F635-[4-[5-[(3-amino-3- oxo-propyl)amino]-2- pyridyl]-2,3-difluoro- phenyl]-N-[3-chloro- 4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate762.3Intermediate D1 and intermediate G86; HCl; iodomethane; piperidineExample F645-[4-(5-amino-3- methyl-2-pyridyl)-2,3- difluoro-phenyl]-N- [3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate705.6Intermediate D1 and intermediate G87; HCl; iodomethane; piperidineExample F655-[4-(5-amino-2- pyridyl)-2,3-difluoro- phenyl]-N-[3-chloro- 4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate691.3Intermediate D1 and intermediate G88; HCl; iodomethane; piperidineExample F66(1S,5R)-6-[[2-chloro- 4-[[5-[2,3-difluoro-4- [1-(2-methoxyethyl)- 5-methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] amino]-N- [(3R,4R)-4-hydroxy- 1,1-dimethyl- pyrrolidin-1-ium-3- yl]-3- azabicyclo[3.1.0]hexane- 3-carboxamide; formate766.3Intermediate D11 and Intermediate G3; HCl; iodomethaneExample F67N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2-fluoro-6-[1-(2- methoxyethyl)-3,5- dimethyl-pyrazol-4- yl]-3-pyridyl]-1- methyl-imidazole-2- carboxamide; formate734.3Intermediate D1 and intermediate G91; HCl; iodomethane Example F58 N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-(3-cyanopropyl)-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide; formate Step 1: tert-butyl (2S,4R)-2-[4-[2-chloro-4-[[5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-4-hydroxy-pyrrolidine-1-carboxylate To a mixture of N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide (1.0 g, 1.44 mmol) and BOC-HYP-OH (400.73 mg, 1.73 mmol) in THE (10 mL) was added N,N-diisopropylethylamine (0.56 g, 4.33 mmol) and PROPYLPHOSPHONIC ANHYDRIDE (1.19 g, 1.88 mmol). The reaction mixture was stirred at 25° C. for 4 h. The solution was extracted with water (10 mL) and EA (20 mL) and washed with sat.aq NaCl (10 mL), dried by anhydrous Na2SO4. The crude was purified by prep-HPLC (FA) to obtain the title compound (600 mg). MS [M+H]+: 767.1. Step 2: tert-butyl (2S,4R)-2-[4-[2-chloro-4-[[5-[4-[1-(3-cyanopropyl)-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-4-hydroxy-pyrrolidine-1-carboxylate To a mixture of tert-butyl (2S,4R)-2-[4-[2-chloro-4-[[5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-4-hydroxy-pyrrolidine-1-carboxylate (100.0 mg, 0.130 mmol) in DMF (3 mL) was added 4-bromo butyronitrile (28.94 mg, 0.200 mmol) and cesium carbonate (127.4 mg, 0.390 mmol). The reaction mixture was stirred at 80° C. for 16 h. The solution was purified by prep-HPLC (FA) directly to obtain the title compound (50 mg). MS [M+H]+: 834.3. Step 3: N-[3-chloro-4-[4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-(3-cyanopropyl)-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide To a mixture of tert-butyl (2S,4R)-2-[4-[2-chloro-4-[[5-[4-[1-(3-cyanopropyl)-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-4-hydroxy-pyrrolidine-1-carboxylate (30.0 mg, 0.040 mmol) in DCM (2 mL) was added TFA (2.0 mL, 0.110 mmol). The reaction mixture was stirred at 25° C. for 16 h. The solution was concentrated in vacuum directly to obtain the title compound (26.4 mg). MS [M+H]+: 734.1. Step 4: N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-(3-cyanopropyl)-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide; formate To a mixture of N-[3-chloro-4-[4-[(2S,4R)-4-hydroxypyrrolidine-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-[1-(3-cyanopropyl)-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide (26.4 mg, 0.040 mmol) in MeCN (2 mL) and water (0.200 mL) was added TEA (0.1 mL, 0.110 mmol) and iodomethane (0.1 mL, 0.040 mmol). The reaction mixture was stirred at 25° C. for 16 h. The solution was concentrated in vacuum and the crude was purified by prep-HPLC (FA) to obtain the title compound (16.9 mg). MS [M]+: 762.2. Example F60 rac-(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-N-[(3R,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-3-yl]-3-azabicyclo[3.1.0]hexane-3-carboxamide; formate Step 1: rac-tert-butyl (3R,4R)-3-[[(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-3-azabicyclo[3.1.0]hexane-3-carbonyl]amino]-4-hydroxy-pyrrolidine-1-carboxylate In a 5 mL sealed, tert-butyl (trans)-3-amino-4-hydroxypyrrolidine-1-carboxylate (45.9 mg, 227 μmol) was combined with DMF (0.5 ml), TEA (38.3 mg, 52.8 μl, 378 μmol) and CDI (30.7 mg, 189 μmol) were added and the reaction mixture was stirred at RT for 20 min. Then N-(4-(((exo)-3-azabicyclo[3.1.0]hexan-6-yl)carbamoyl)-3-chlorophenyl)-5-(2,3-difluoro-4-(1-(2-methoxyethyl)-3,5-dimethyl-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamide hydrochloride (50 mg, 75.7 μmol) was added and the reaction was stirred at RT. The product was submitted for purification by prep HPLC. To afford the title compound (15.5 mg). MS [M+H]+: 852.3. Step 2: rac-tert-butyl (3R,4R)-3-[[(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-3-azabicyclo[3.1.0]hexane-3-carbonyl]amino]-4-hydroxy-pyrrolidine-1-carboxylate In a 5 mL round-bottomed flask, rac-tert-butyl (3R,4R)-3-[[(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-3-azabicyclo[3.1.0]hexane-3-carbonyl]amino]-4-hydroxy-pyrrolidine-1-carboxylate (15 mg, 17.6 μmol) was combined with DCM (150 μL) to give a light brown solution. HCl 4 M in dioxane (22 μL, 88 μmol) was added and the reaction mixture was stirred at RT. The reaction mixture was concentrated to dryness then lyophilized. To afford the title compound (14.1 mg). MS [M+H]+ 752.3. Step 3: rac-(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-N-[(3R,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-3-yl]-3-azabicyclo[3.1.0]hexane-3-carboxamide; formate In a 5 mL round-bottomed flask, rac-tert-butyl (3R,4R)-3-[[(1S,5R)-6-[[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]amino]-3-azabicyclo[3.1.0]hexane-3-carbonyl]amino]-4-hydroxy-pyrrolidine-1-carboxylate (14 mg, 17.8 μmol) was combined with acetonitrile (300 μL) to give a light brown suspension. DIPEA (6.88 mg, 9.3 μl, 53.3 μmol) and Mel (6.05 mg, 42.6 μmol) were added and the reaction mixture was stirred overnight at RT. The reaction mixture was concentrated to dryness and submitted for purification to prep HPLC. Finally the product was lyophilized to afford the title compound (6.8 mg). MS [M]*. 780.3. Example G1 N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(isopropylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate Step 1: tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-(isopropylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate 2-[4-[4-[2-[[4-[4-(1-tert-butoxycarbonylisonipecotoyl)piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazol-1-yl]acetic acid (150 mg, 0.185 mmol), DIPEA (71.87 mg, 0.556 mmol) and isopropylamine (16.43 mg, 0.278 mmol) were dissolved in acetonitrile (1.85 mL). To this stirred solution was added HATU (84.57 mg, 0.222 mmol) in one portion. The resulting yellow solution was stirred at room temperature for 30 min. The solvent was removed in vacuo, and the residue was purified by flash chromatography to afford the product (141 mg). MS [M+H]+: 850.8. Step 2: N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(isopropylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-(isopropylamino)-2-keto-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester (141 mg, 0.166 mmol) was stirred in 5 mL 1.5 N HCl/MeOH solution at room temperature for 5 h. The solvent was removed in vacuum, and the crude product (140 mg) was used without purification. MS [M+H]+: 750.6. Step 3: N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(isopropylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(isopropylamino)-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (140 mg) was dissolved in N,N-dimethylacetamide (2 mL). To this solution was added DIPEA (214.3 mg, 1.66 mmol) and iodomethane (70.61 mg, 0.497 mmol). The solution was stirred at room temperature for 1 h. The product was purified by preparative HPLC directly to afford the product as amorphous powder (56 mg). MS [M]+: 778.5. The following examples were prepared in analogy to Example G1. MSESIStartingEx#NameStructure[M]+MaterialExample G25-[4-[1-[2-(tert- butylamino)-2-oxo- ethyl]-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-N- [3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate792.7Intermediate O1 and 2- methylpropan- 2-amine; HCl; iodomethane.Example G35-[4-[1-[2-(1- bicyclo[1.1.1]pentanyl amino)-2-oxo-ethyl]- 3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-N- [3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate802.4Intermediate O1 and bicyclo[1.1.1] pentan-1- amine; HCl; iodomethane.Example G4N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2-[(3-cyano-1- bicyclo[1.1.1]pentanyl) amino]-2-oxo-ethyl]- 3-methyl-pyrazol-4- yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide; formate827.5Intermediate O1 and 3- aminobicyclo [1.1.1]pentane- 1-carbonitrile; HCl; iodomethane.Example G55-[4-[1-(2-anilino-2- oxo-ethyl)-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-N- [3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate812.6Intermediate O1 and aniline; HCl; iodomethane.Example G6N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[2- (2-fluoroanilino)-2- oxo-ethyl]-3-methyl- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate830.4Intermediate O1 and 2- fluoroaniline; HCl; iodomethane.Example G7N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[2- (2-methoxyanilino)-2- oxo-ethyl]-3-methyl- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate842.4Intermediate O1 and 2- methoxyaniline; HCl; iodomethaneExample G8N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[2- (4-fluoroanilino)-2- oxo-ethyl]-3-methyl- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate830.4Intermediate O1 and 4- fluoroaniline; HCl; iodomethaneExample G9N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2- (cyclohexylamino)-2- oxo-ethyl]-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-1- methyl-imidazole-2- carboxamide; formate818.4Intermediate O1 and cyclo- hexanamine; HCl; iodomethaneExample G10N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2- (thiazol-2- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate819.3Intermediate O1 and thiazol-2- amine; HCl; iodomethaneExample G11N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2- (1H-pyrazol-4- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate802.3Intermediate O1 and 1H- pyrazol-4- amine; HCl; iodomethaneExample G12N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-[(1- methylpyrazol-4- yl)amino]-2-oxo- ethyl]pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate816.3Intermediate O1 and 1- methylpyrazol- 4-amine; HCl; iodomethaneExample G13N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[2- (3-fluoroanilino)-2- oxo-ethyl]-3-methyl- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; chloride830.4Intermediate O1 and 3- fluoroaniline; HCl; iodomethaneExample G14N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2-(2- pyridylamino)ethyl] pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate813.4Intermediate O1 and pyridin-2- amine; HCl; iodomethaneExample G15N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-[(1- methylpyridin-1-ium- 3-yl)amino]-2-oxo- ethyl]pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; diformate827.4Intermediate O1 and pyridin-3- amine; HCl; iodomethaneExample G16N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2- (pyrimidin-2- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate814.3Intermediate O1 and pyrimidin-2- amine; HCl; iodomethaneExample G17N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2-(4- pyridylamino)ethyl] pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; chloride813.2Intermediate O1 and pyridin-4- amine; HCl; iodomethaneExample G185-[4-[1-[2-(tert- butylamino)-2-oxo- ethyl]-5-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-N- [3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl] phenyl]-1- methyl-imidazole-2- carboxamide; formate792.3Intermediate O2 and 2- methylpropan- 2-amine; HCl; iodomethaneExample G19N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[5- methyl-1-[2-oxo-2-(2- pyridylamino)ethyl] pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate813.3Intermediate O2 and pyridin-2- amine; HCl; iodomethaneExample G20N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2- (tetrahydrofuran-3- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate806.6Intermediate O1 and tetrahydrofuran- 3-amine; HCl; iodomethaneExample G21N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2- (tetrahydropyran-2- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate820.3Intermediate O1 and tetrahydropyran- 2-amine; HCl; iodomethaneExample G22N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2- (tetrahydropyran-3- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate820.3Intermediate O1 and intermediate R8; HCl; iodomethaneExample G23N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2- (pyridazin-3- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate814.5Intermediate O1 and pyridazin-3- amine; HCl; iodomethaneExample G24N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2-[0,1- dimethylpiperidin-1- ium-3-yl)amino]-2- oxo-ethyl]-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-1- methyl-imidazole-2- carboxamide; diformate424.6Intermediate O1 and benzyl 3- aminopiperidine- 1-carboxylate; HCl; iodomethaneExample G25N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[5- methyl-1-[2-oxo-2- (pyridazin-3- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate814.0Intermediate O2 and pyridazin-3- amine; HCl; iodomethaneExample G26N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2-[(4,4- difluorocyclohexyl) amino]-2-oxo-ethyl]-3- methyl-pyrazol-4-yl]- 2,3-difluoro-phenyl]- 1-methyl-imidazole-2- carboxamide; formate854.4Intermediate O1 and 4,4- difluorocyclo hexanamine; HCl; iodomethaneExample G27N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- pyridylamino)ethyl] pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate813.6Intermediate O1 and pyridin-3- amine; HCl; iodomethaneExample G28N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[2- [(5-methoxy-2- pyridyl)amino]-2-oxo- ethyl]-3-methyl- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate845.1Intermediate O1 and 5- methoxypyridin- 2-amine; HCl; iodomethaneExample G29N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2-(1- piperidyl)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate804.4Intermediate O1 and piperidine; HCl; iodomethaneExample G30N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2-[4-(4,4- dimethylpiperazin-4- ium-1- carbonyl)anilino]-2- oxo-ethyl]-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-1- methyl-imidazole-2- carboxamide; diformate477.1Intermediate O1 and (4- aminophenyl)- (4- methylpiperazin- 1- yl)methanone; HCl; iodomethaneExample G31N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[2- [(6-fluoropyridazin-3- yl)amino]-2-oxo- ethyl]-3-methyl- pyrazol-4-yl]phenyl]- 1-methyl-imidazole-2- carboxamide; formate832.3Intermediate O1 and 6- fluoropyridazin- 3-amine; HCl; iodomethaneExample G32N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[1-[2- [[(1S,2S)-2- methoxycyelohexyl] amino]-2-oxo-ethyl]-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate848.5Intermediate O1 and rac- (1S,2S)-2- methoxycyclo hexanamine; HCl; iodomethaneExample G33N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [2,3-difluoro-4-[3- methyl-1-[2-oxo-2- (pyridazin-4- ylamino)ethyl]pyrazol- 4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate814.5Intermediate O1 and pyridazin-4- amine; HCl; iodomethaneExample G34N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]-5- [4-[1-[2-[(6-cyano-3- pyridyl)amino]-2-oxo- ethyl]-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-1- methyl-imidazole-2- carboxamide; formate838.4Intermediate O1 and pyridazin-4- amine; HCl; iodomethane Example H1 N-[2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-5-methyl-pyrazol-1-yl]ethyl]pyridine-2-carboxamide; formate Step 1: tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[5-methyl-1-[2-(pyridine-2-carbonylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate tert-butyl 4-[4-[4-[[5-[4-[1-(2-aminoethyl)-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (100 mg, 0.126 mmol) was dissolved in dichloromethane (5 mL), 2-picolinic acid (18.6 mg, 0.151 mmol), HATU (57.44 mg, 0.151 mmol) and DIEA (32.54 mg, 0.252 mmol) were added at rt. The mixture was stirred at room temperature for 1 h. The reaction mixture was diluted with water (30 mL) and extracted two times with DCM (25 mL). The organic layers were washed with brine (20 mL), dried over Na2SO4and concentrated to dryness. the crude product was directly used to the next step to afford tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[5-methyl-1-[2-(pyridine-2-carbonylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (113 mg). MS [M+H]+: 899.7. Step 2: N-[2-[4-[4-[2-[[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-5-methyl-pyrazol-1-yl]ethyl]pyridine-2-carboxamide tert-butyl 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[5-methyl-1-[2-(pyridine-2-carbonylamino)ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylate (113 mg, 0.126 mmol) was dissolved in tetrahydrofuran (2 mL) and 12 M HCl (in water) (628.21 mg, 6.28 mmol) was added at rt. The mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated under vacuum, the crude product was directly used to the next step, to afford N-[2-[4-[4-[2-[[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-5-methyl-pyrazol-1-yl]ethyl]pyridine-2-carboxamide (100 mg) as light brown solid. MS [M+H]+: 799.9. Step 3: N-[2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-5-methyl-pyrazol-1-yl]ethyl]pyridine-2-carboxamide; formate N-[2-[4-[4-[2-[[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-5-methyl-pyrazol-1-yl]ethyl]pyridine-2-carboxamide (100 mg, 0.125 mmol) was dissolved in acetonitrile (6 mL), iodomethane (53.28 mg, 0.375 mmol) and DIEA (48.51 mg, 0.375 mmol) were added at rt. The mixture was stirred at room temperature for 1 h. The reaction was concentrated under vacuum, the crude product was purified by HPLC to afford N-[2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-5-methyl-pyrazol-1-yl]ethyl]pyridine-2-carboxamide; formate (56.6 mg) as white powder. MS [M]+: 828.1. The following examples were prepared in analogy to Examples H1. MSESIStartingEx#NameStructure[M]+MaterialExample H2N-[2-[4-[4-[2-[[3-chloro- 4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl] carbamoyl]- 3-methyl-imidazol- 4-yl]-2,3-difluoro- phenyl]-3-methyl- pyrazol-1- yl]ethyl]pyridine-2- carboxamide; formate827.7Inter- mediate Q2 and 2- picolinic acid; HCl iodomethaneExample H3N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]- 5-[2,3- difluoro-4-[1-[2-[(4- fluorophenyl) sulfonylamino] ethyl]-3-methyl- pyrazol-4-yl]phenyl]-1- methyl -imidazole-2- carboxamide; formate881.3Inter- mediate Q2; 4- fluoro- benzene sulfonyl chloride; HCI iodomethaneExample H45-[4-[1-[2-(tert- butylcarbamoylamino) ethyl]-5-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]- N-[3-chloro-4-[4-(1,1- dimethylpiperidin- 1-ium-4- carbonyl)piperazine-1- carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate822.2Inter- mediate Q1; 2- isocyanato- 2- methyl- propane; HCI iodomethaneExample H55-[4-[1-[2-(tert- butylcarbamoylamino) ethyl]-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]- N-[3-chloro-4-[4-(1,1- dimethylpiperidin- 1-ium-4- carbonyl)piperazine-1- carbonyl]phenyl]-1- methyl-imidazole-2- carboxamide; formate822.2Inter- mediate Q2; 2- isocyanato- 2-methyl- propane; HCI iodomethaneExample H6N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]- 5-[2,3- difluoro-4-[5-methyl-1- [2-(2- pyridylcarbamoylamino) ethyl]pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate842.7Inter- mediate Q1; 2- isocyanato- pyridine; HCI iodomethaneExample H7N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]- 5-[2,3- difluoro-4-[3-methyl-1- [2-(2- pyridylcarbamoylamino) ethyl]pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate842.3Inter- mediate Q2; 2- isocyanato- pyridine; HCI iodomethaneExample H8N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1- ium-4- carbonyl)piperazine- 1-carbonyl]phenyl]- 5-[2,3- difluoro-4-[3-methyl-1- [2-(pyrrolidine-1- carbonylamino)ethyl] pyrazol-4-yl] phenyl]-1- methyl-imidazole-2- carboxamide; formate819.5Inter- mediate Q2; pyrrolidine- 1-carbonyl chloride; HCI iodomethane Example 11 N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate Step 1: N-(3-chloro-4-(4-(1-methylpiperidine-4-carbonyl)piperazine-1-carbonyl)phenyl)-5-(2,3-difluoro-4-(5-(pyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamide To a solution of 5-bromo-N-[3-chloro-4-[4-(1-methylpiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide (80.0 mg, 0.14 mmol) in 1,4-dioxane (1.0 mL) was added [2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]boronic acid (75.0 mg, 0.17 mmol), sodium carbonate (46.1 mg, 0.43 mmol), [1,1-Bis(di-tert-butylphosphino)ferrocene]palladium(II) Dichloride (19.2 mg, 0.02 mmol) and water (0.1 mL) in glove box, the mixture was stirred at 100° C. for 2 h. The mixture was diluted with 10 mL of DCM and filtered off, the filtrate was concentrated, the residue was purified by Prep-HPLC (FA as additive) to give N-[3-chloro-4-[4-(1-methylpiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (45 mg). MS [M+H]+: 858.4. Step 2: N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; iodide To a solution of N-[3-chloro-4-[4-(1-methylpiperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (39.0 mg, 0.05 mmol) in DMF (1 mL) was added N,N-diisopropylethylamine (0.03 mL, 0.18 mmol) and iodomethane (25.8 mg, 0.18 mmol), the mixture was stirred at 25° C. for 0.5 h. The mixture was purified by Flash-HPLC and lyophilized to give N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; iodide (35.0 mg). MS[M]+: 872.5. Step 3: N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate The mixture of N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1-(2-trimethylsilylethoxymethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; iodide (35.0 mg, 0.04 mmol) and HCl/MeOH (4.0 mL) was stirred at 25° C. for 16 h. The mixture was concentrated at 20° C. in vacuum, the residue was purified by Prep-HPLC (FA as additive) and lyophilized to give N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[5-(2-pyridyl)-1H-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (10.1 mg). MS [M]+: 742.4. The following examples were prepared in analogy to Examples I1. MSESIStartingEx#NameStructure[M]+MaterialExam- ple I2N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[5-(4- methoxyphenyl)-1H- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate771.2Intermediate D9 and Intermediate G59; iodomethane; HClExam- ple I3N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[5-(3- methoxyphenyl)-1H- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate771.2Intermediate D9 and Intermediate G60; iodomethane; HClExam- ple I4N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[5-(2- methoxyphenyl)-1H- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carboxamide; formate771.2Intermediate D9 and Intermediate G61; iodomethane; HClExam- ple I5N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-(5-thiazol-4- yl-1H-pyrazol-4- yl)phenyl]-1-methyl- imidazole-2- carboxamide; formate748.1Intermediate D9 and Intermediate G62; iodomethane; HClExam- ple I6N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-(5- tetrahydropyran-4-yl-1H- pyrazol-4-yl)phenyl]-1- methyl-imidazole-2- carboxamide; formate749.3Intermediate D9 and Intermediate G63; iodomethane; HClExam- ple I7N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[4- [5-(3,6-dihydro-2H- pyran-4-yl)-1H-pyrazol- 4-yl]-2,3-difluoro- phenyl]-1-methyl- imidazole-2- carboxamide;formate747.0Intermediate D9 and Intermediate G64; iodomethane; HCl Example J1 N-[4-[4-[1-(azetidin-3-ylmethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide; formate Step 1: tert-butyl 3-[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]methyl]azetidine-1-carboxylate In a 50 mL round-bottomed flask, N-[3-chloro-4-[4-(piperidine-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide (88 mg, 135 μmol), tert-butyl 3-formylazetidine-1-carboxylate (50.1 mg, 270 μmol) and NaBH3CN (25.5 mg, 405 μmol) were combined with MeOH (5 mL) to give a light brown solution. The reaction mixture was heated to 45° C. and stirred for 3 h. The crude reaction mixture was concentrated in vacuum. The reaction mixture was poured into 25 mL sat NaHCO3and extracted with EtOAc (3×25 mL). The organic layers were combined, washed with sat NaCl (1×25 mL), The organic layers were dried over Na2SO4and concentrated in vacuum to afford tert-butyl 3-[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]methyl]azetidine-1-carboxylate (111 mg). MS [M+H]+: 821.1. Step 2: tert-butyl 4-[4-[2-[[4-[4-[1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]piperidine-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazole-1-carboxylate In a 100 mL round-bottomed flask, tert-butyl 3-[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]-1-piperidyl]methyl]azetidine-1-carboxylate (110 mg, 134 μmol), Boc2O (43.9 mg, 201 μmol) and TEA (20.4 mg, 201 μmol) were combined with DCM (5 mL) to give a light brown solution. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford tert-butyl 4-[4-[2-[[4-[4-[1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]piperidine-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazole-1-carboxylate (123 mg, 134 μmol). MS [M+H]m: 920.8. Step 3: tert-butyl 4-[4-[2-[[4-[4-[1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazole-1-carboxylate; iodide In a 100 mL round-bottomed flask, tert-butyl 4-[4-[2-[[4-[4-[1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]piperidine-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazole-1-carboxylate (123 mg, 134 μmol), Mel (94.8 mg, 668 μmol) and DIPEA (86.4 mg, 668 μmol) were combined with MeCN (5 mL) to give a light brown solution. The reaction was stirred at room temperature for 15 h. The crude reaction mixture was concentrated in vacuum. The crude product was directly used to the next step to afford tert-butyl 4-[4-[2-[[4-[4-[1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazole-1-carboxylate; iodide (125 mg). MS [M]+: 935.1. Step 4: N-[4-[4-[1-(azetidin-3-ylmethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate In a 100 mL round-bottomed flask, tert-butyl 4-[4-[2-[[4-[4-[1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazole-1-carboxylate; iodide (120 mg, 128 μmol) was combined with DCM (3 mL) to give a light brown solution. 2,2,2-trifluoroacetic acid (1.46 g, 12.8 mmol) was added. The reaction was stirred at room temperature for 1 h. The crude reaction mixture was concentrated in vacuum. The crude material was purified by preparative HPLC to afford N-[4-[4-[1-(azetidin-3-ylmethyl)-1-methyl-piperidin-1-ium-4-carbonyl]piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate (17.4 mg). MS [M]+: 734.9. Example K1 5-[4-[1-[2-(2-aminoethoxy)ethyl]-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate Step 1: N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(2-hydrazinoethoxy)ethyl]-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate To a mixture of N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[4-(3,5-dimethyl-1H-pyrazol-4-yl)-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide; formate (100.0 mg, 0.140 mmol) in DMF (10 mL) was added tert-butyl N-[2-(2-bromoethoxy)ethyl]carbamate (385.18 mg, 1.44 mmol), and sodium borohydride (108.69 mg, 2.87 mmol). The reaction mixture was stirred at 80° C. for 16 h. The mixture was concentrated to remove solvent and purified by prep-HPLC (0.1% FA)-ACN to afford N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(2-hydrazinoethoxy)ethyl]-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (60 mg). MS [M]+: 882.3. Step 2: 5-[4-[1-[2-(2-aminoethoxy)ethyl]-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-[2-(2-hydrazinoethoxy)ethyl]-3,5-dimethyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (60.0 mg, 0.070 mmol) was added into HCl/dioxane (5.0 mL, 0.070 mmol). The reaction mixture was stirred at 25° C. for 2 h. The mixture was concentrated to get crude product and purified by prep-HPLC (0.1% TFA)-ACN to obtain 5-[4-[1-[2-(2-aminoethoxy)ethyl]-3,5-dimethyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate (13.3 mg). MS [M]+: 782.5. The following examples were prepared in analogy to Examples K1. MSESIStartingEx#NameStructure[M]+MaterialExample K2N-[3-chloro-4-[4-[(4R)- 4-hydroxy-1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[4- [1-2- (difluoromethoxy)ethyl]- 3,5-dimethyl-pyrazol-4- yl]-2,3-difluoro-phenyl]- 1-methyl-imidazole-2- carboxamide; formate789.3Example D43 and 1-bromo-2- (difluoromethoxy) ethaneExample K3N-[3-chloro-4-[4-[(4R)- 4-hydroxy-1,1-dimethyl- pyrrolidin-1-ium-2- carbonyl]piperazine-1- carbonyl]phenyl]-5-[4- [1-2,2- difluorocyclopropyl) methyl]-3,5-dimethyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-1- methyl-imidazole-2- carboxamide; formate785.3Example D43 and 2- (bromomethyl)- 1,1- difluoro- cyclopropane Intermediate G94 tert-butyl 2-[3-methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]pyrazol-1-yl]acetate Step 1: tert-butyl 2-[4-(4-bromophenyl)-3-methyl-pyrazol-1-yl]acetate A mixture of 4-bromophenylboronic acid (6.23 g, 31.04 mmol), tert-butyl 2-(4-iodo-3-methyl-pyrazol-1-yl)acetate (10.0 g, 31.04 mmol)(isomer mixture), sodium carbonate (6.58 g, 62.08 mmol) and Pd(dppf)Cl2 (2.27 g, 3.1 mmol) in 1,4-dioxane (200 mL)/Water (20 mL) was stirred under N2 at 85° C. for 16 h. The reaction mixture was filtered and the filtrate was concentrated in vacuum, purified by silica column (PE:EA=5:1) to afford tert-butyl 2-[4-(4-bromophenyl)-3-methyl-pyrazol-1-yl] acetate (3.3 g, 9.4 mmol, 30.27% yield)(isomer mixture) as yellow oil. MS [(M+2+H)+]: 353.0. Step 2: tert-butyl 2-[3-methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]pyrazol-1-yl]acetate A mixture of tert-butyl 2-[4-(4-bromophenyl)-3-methyl-pyrazol-1-yl]acetate (3.3 g, 9.4 mmol)(isomer mixture), potassium acetate (1.84 g, 18.79 mmol), bis(pinacolato)diboron (7157.49 mg, 28.19 mmol) and Pd(dppf)Cl2 (0.69 g, 0.94 mmol) in 1,4-dioxane (50 mL) was evacuated and backfilled with N2 (3×), then the mixture was stirred under N2 at 95° C. for 16 h. The mixture was cooled to room temperature, filtered and concentrated in vacuum to afford a residue. The residue was purified by silica column (PE:EA=10:1 to 1:1) to afford tert-butyl 2-[3-methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]pyrazol-1-yl]acetate (3.7 g, 9.29 mmol, 98.87% yield)(isomer mixture) as yellow solid. MS [(M+H)+]: 399.3. The following examples were prepared in analogy to Intermediate G94. MSStartingEx#NameStructure[M + H]+MaterialIntermediate G95tert-butyl 2-[4-[2- fluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3- methyl-pyrazol-1- yl]acetate417.24-bromo-2- fluorobenzene boronic acidIntermediate G96tert-butyl 2-[4-[2,3- difluoro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-3- methyl-pyrazol-1- yl]acetate435.14-bromo-2,3- difluorophenylboronic acid Intermediate G97 Step 1: 1-[2-(difluoromethoxy)ethyl]-4-iodo-5-methyl-pyrazole To a mixture of 4-iodo-3-methyl-1H-pyrazole (13.0 g, 62.5 mmol) and 2-(difluoromethoxy)ethyl 4-methylbenzenesulfonate (14.98 g, 56.25 mmol) in DMF (150 mL) was added potassium carbonate (21.59 g, 156.25 mmol) at 25° C. Then the mixture was stirred at 65° C. for 16 h. The reaction mixture was pour into water (150 mL), extracted with EA (150 mL×2). The combined organic layer was washed with brine (200 mL), dry over sodium sulfate, concentrated in vacuum to give the residue, which was purified by column (PE:EA=10:1 to 3:1) to afford 1-[2-(difluoromethoxy)ethyl]-4-iodo-5-methyl-pyrazole (12.0 g, 39.72 mmol, 31.78% yield)(isomer mixture) as yellow oil. MS [(M+H)+]: 303.0. Step 2: 4-(4-bromo-2-fluoro-3-methyl-phenyl)-1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazole To a mixture of 1-[2-(difluoromethoxy)ethyl]-4-iodo-5-methyl-pyrazole (12.98 g, 42.95 mmol)(isomer mixture), (4-bromo-2-fluoro-3-methyl-phenyl)boronic acid (10.0 g, 42.95 mmol) and sodium carbonate (11.38 g, 107.37 mmol) in 1,4-dioxane (200 mL)/Water (20 mL) was added Pd(dppf)Cl2 (3.14 g, 4.30 mmol) at 25° C., the mixture was evacuated, backfilled with N2 (3×) and stirred at 60° C. for 4 h under N2. The reaction mixture was filtered, the filtrate was concentrated in vacuum to give the residue, which was purified by column (PE:EA=10:1 to 2:1) to afford 4-(4-bromo-2-fluoro-3-methyl-phenyl)-1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazole (12.0 g, 33.04 mmol, 76.94% yield)(isomer mixture) as yellow oil. MS [(M+H)+]: 364.9. Step 3: 1-[2-(difluoromethoxy)ethyl]-4-[2-fluoro-3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5-methyl-pyrazole A stirred mixture of 4-(4-bromo-2-fluoro-3-methyl-phenyl)-1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazole (12.0 g, 33.04 mmol)(isomer mixture), Pd(dppf)Cl2 (2.42 g, 3.3 mmol), bis(pinacolato)diboron (12.59 g, 49.56 mmol) and potassium acetate (5.16 mL, 82.61 mmol) in 1,4-dioxane (200 mL) was evacuated and backfilled with N2 (3×) at 25° C. Then the mixture was stirred at 95° C. for 16 h under N2. The reaction mixture was cooled to room temperature, filtered, the filtrate was concentrated in vacuum to give the residue, which was purified by column (PE:EA=10:1 to 3:1) to afford 1-[2-(difluoromethoxy)ethyl]-4-[2-fluoro-3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5-methyl-pyrazole (10.0 g, 24.38 mmol, 73.77% yield)(isomer mixture) as yellow solid. MS [(M+H)+]: 410.9. The following examples were prepared in analogy to Intermediate G2. MSStartingEx#NameStructure[M + H]+MaterialIntermediate G984-[2-fluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-5- methyl-pyrazole361.31-bromo-3- fluoro-4- iodobenzene and Intermediate E1Intermediate G994-[2-fluoro-4- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2- yl)phenyl]-1-(2- methoxyethyl)-3- methyl-pyrazole361.31-bromo-3- fluoro-4- iodobenzene and Intermediate E2 The following example were prepared in analogy to Intermediate H1. MSStartingEx#NameStructure[M − H]−MaterialIntermediate H12N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]- 5-[3-fluoro-4-[1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide hydrochloride578.3Intermediate Bl and Intermediate G98Intermediate H13N-[3-chloro-4- (piperazine-1- carbonyl)phenyl]- 5-[3-fluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carboxamide hydrochloride578.4Intermediate B1 and Intermediate G99Intermediate H145-[2,3-difluoro-4- [1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1- methyl-N-[3- methyl-4- (piperazine-1- carbonyl)phenyl] imidazole-2- carboxamide hydrochloride578.3Intermediate B2 and Intermediate G2Intermediate H155-[3-fluoro-4-[1-(2- methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1- methyl-N-[3- methyl-4- (piperazine-1- carbonyl)phenyl] imidazole-2- carboxamide hydrochloride560.5Intermediate B2 and Intermediate G98Intermediate H165-[3-fluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]-2-methyl- phenyl]-1-methyl- N-[3-methyl-4- (piperazine-1- carbonyl)phenyl] imidazole-2- carboxamide hydrochloride574.5Intermediate B2 and Intermediate G13 Intermediate I13 N-[3-chloro-4-[4-(methylamino)piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formic acid To 188 mg of 2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoic acid (23.5 mg, 0.0338 mmol) dissolved in 1 ml DMIF was added HATU (15.4 mg, 0.041 mmol, 1.200 eq) and Et3N (24 uL, 0.169 mmol, 5.000 eq). The mixture was stirred at room temperature for 10 mi and then N-methyl-N-(4-piperidyl)carbamic acid tert-butyl ester (9.4 mg, 0.0439 mmol, 1.300 eq) was added and the mixture was stirred at room temperature over night. The mixture was then evaporated to dryness, dissolved in 1 ml DCM and treated with an excess of 4N HCl in dioxan (250 uL, 1.01 mmol, 30.000 eq) over night at RT. The mixtures was then evaporated to dryness and directly purified by preparative HPLC to afford the title compound (2 mg. 8.8% yield). MS [(M−H)−]: 624.3. The following example was prepared in analogy to Intermediate I13. MSStartingEx#NameStructure[M + H]+MaterialIntermediate I14tert-butyl 4-[4-[[5- [4-[1-(2-tert- butoxy-2-oxo- ethyl)-3-methyl- pyrazol-4-yl]-3- fluoro-phenyl]-1- methyl-imidazole- 2-carbonyl]amino]- 2-chloro- benzoyl]piperazine- 1-carboxylate612.53-methylpiperazine- 1-carboxylic acid tert-butyl ester Intermediate O4 Step 1: ethyl 5-[4-[1-(2-tert-butoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxylate A mixture of tert-butyl 2-[3-methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]pyrazol-1-yl]acetate (3.7 g, 9.29 mmol)(isomer mixture), ethyl 5-bromo-1-methyl-imidazole-2-carboxylate (3.25 g, 13.93 mmol), potassium carbonate (2.57 g, 18.58 mmol), BrettPhos Pd G3 (843.03 mg, 0.93 mmol) in 1,4-dioxane (50 mL)/Water (5 mL) was stirred under N2 at 90° C. for 16 h. The mixture was concentrated in vacuum and purified by silica column (PE:EA=2:1) to afford ethyl 5-[4-[1-(2-tert-butoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxylate (1.2 g, 2.83 mmol, 30.43% yield)(isomer mixture) as yellow oil. MS [(M+H)+]: 425.1. Step 2: tert-butyl 4-[4-[[5-[4-[1-(2-tert-butoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carboxylate To a solution of tert-butyl 4-(4-amino-2-chloro-benzoyl)piperazine-1-carboxylate (800.54 mg, 2.36 mmol) and ethyl 5-[4-[1-(2-tert-butoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxylate (1.0 g, 2.36 mmol)(isomer mixture) in THE (20 mL) was added dropwise potassium bis(trimethylsilyl) amide (3.53 mL, 3.53 mmol, 1M in hexane) slowly at −35° C. and stirred for 0.5 h under N2. The solution was poured into sat.NH4Cl (100 ml) and extracted with EA (50 ml×2), washed with brine (100 ml), dried by anhydrous Na2SO4, then concentrated in vacuum to afford a residue. The residue was purified by silica column (55% EtOAc in PE) to afford the isomer mixture. The isomer mixture was purified by SFC [Column: Chiralpak IG-3 50×4.6 mm I.D., 3 um Mobile phase: Phase A for CO2, and Phase B for MeOH+ACN (0.05% DEA); Gradient elution: 60% Methanol+ACN (0.05% DEA) in CO2 Flow rate: 3 mL/min; Detector: PDA Column Temp: 35° C.; Back Pressure: 100 Bar] to afford tert-butyl 4-[4-[[5-[4-[1-(2-tert-butoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl] piperazine-1-carboxylate (470.0 mg, 0.65 mmol, 27.78% yield) as light yellow solid MS (ESI+) [(M+H)+]: 718.3. The following example were prepared in analogy to Intermediate O4. MSStartingEx#NameStructure[M + H]+MaterialIntermediate O5tert-butyl 4-[4-[[5- [4-[1-(2-tert- butoxy-2-oxo- ethyl)-3-methyl- pyrazol-4-yl]-3- fluoro-phenyl]-1- methyl-imidazole- 2-carbonyl]amino]- 2-chloro- benzoyl]piperazine- 1-carboxylate736.2Intermediate G95Intermediate O6tert-butyl 4-[4-[[5- [4-[1-(2-tert- butoxy-2-oxo- ethyl)-3-methyl- pyrazol-4-yl]-2,3- difluoro-phenyl]-1- methyl-imidazole- 2-carbonyl]amino]- 2-chloro- benzoyl]piperazine- 1-carboxylate754.4Intermediate G96Intermediate O7tert-butyl 4-[2- chloro-4-[[5-[4-[1- [2- (difluoromethoxy) ethyl]-5-methyl- pyrazol-4-yl]-3- fluoro-2-methyl- phenyl]-1-methyl- imidazole-2- carbonyl]amino] benzoyl]piperazine-1-carboxylate730.0Intermediate G97 Intermediates R16 and R16′ cis-1-(2-(tert-butoxy)-2-oxoethyl)-1-((1-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-4-carboxypiperidin-1-ium; 2,2,2-trifluoroacetate trans-1-(2-(tert-butoxy)-2-oxoethyl)-1-((1-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-4-carboxypiperidin-1-ium; 2,2,2-trifluoroacetate Intermediate R14 was purified by SFC (Column: DAICEL CHIRALCEL OD (250 mm×30 mm, 10 um); condition: 0.1% NH3H2O MEOH; 20% B; Gradient Time(min): 6.5 min; FlowRate(ml/min): 50 mL/min) to give cis-1-(2-(tert-butoxy)-2-oxoethyl)-1-((1-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-4-carboxypiperidin-1-ium; 2,2,2-trifluoroacetate (4.6 g, 11.15 mmol, 28.65% yield) as a white solid, MS [(M)+]: 413.0 and trans-1-(2-(tert-butoxy)-2-oxoethyl)-1-((1-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-4-carboxypiperidin-1-ium; 2,2,2-trifluoroacetate (4.6 g, 11.15 mmol, 28.65% yield) as a white solid MS [(M)+]: 413.0. Intermediate R17 (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)-methyl-ammonium; 2,2,2-trifluoroacetate Step 1: tert-butyl 3-[[(4-benzyloxy-4-oxo-butyl)amino]methyl]azetidine-1-carboxylate To a mixture of benzyl 4-aminobutanoate; hydrochloride (4.5 g, 19.59 mmol) and potassium carbonate (8.12 g, 58.77 mmol), sodium iodide (146.83 mg, 0.98 mmol) in ACN (45 mL) was added 1-BOC-3-(bromomethyl)azetidine (6.37 g, 25.47 mmol) at 20° C. Then the mixture was stirred at 50° C. for 16 h. The mixture was filtered, the solid was washed with ACN (20 mL×3). The combined filtrate was concentrated under vacuum to give tert-butyl 3-[[(4-benzyloxy-4-oxo-butyl)amino]methyl]azetidine-1-carboxylate (12.0 g, 33.11 mmol, 99.71% yield) as a light yellow oil. MS [(M+H)+]: 363.2. Step 2: tert-butyl 3-[[(4-benzyloxy-4-oxo-butyl)-methyl-amino]methyl]azetidine-1-carboxylate To a solution of tert-butyl 3-[[(4-benzyloxy-4-oxo-butyl)amino]methyl]azetidine-1-carboxylate (12.0 g, 33.11 mmol) in MeCN (100 mL) was added acetic acid (397.63 mg, 6.62 mmol) and formaldehyde (13.44 g, 165.54 mmol, 37% in water) at 30° C. and stirred for 2 h. Sodium cyan borohydride (4.16 g, 66.21 mmol) was added into the mixture in five portions at 30° C. and stirred at 30° C. for another 16 h. The mixture was concentrated under vacuum to afford a residue. The residue was dissolved in ACN (100 mL), filtered through the celite pad, The filtrate was concentrated under vacuum and purified by reversed phase-HPLC (water (0.1% FA)-ACN) and lyophilized to give tert-butyl 3-[[(4-benzyloxy-4-oxo-butyl)-methyl-amino]methyl]azetidine-1-carboxylate (1.9 g, 5.05 mmol, 15.24% yield) as light red oil. MS [(M+Na)+]: 399.2. Step 3: (4-benzyloxy-4-oxo-butyl)-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-(2-tert-butoxy-2-oxo-ethyl)-methyl-ammonium; 2,2,2-trifluoroacetate To a solution of tert-butyl 3-[[(4-benzyloxy-4-oxo-butyl)-methyl-amino]methyl]azetidine-1-carboxylate (1.7 g, 4.52 mmol) and DIEA (1.57 mL, 9.03 mmol) in ACN (20 mL) was added sodium iodide (67.68 mg, 0.45 mmol) and tert-butyl bromoacetate (1.32 g, 6.77 mmol). The mixture was stirred at 50° C. for 16 h. The mixture was concentrated under vacuum to afford a residue, which was purified by revered phase-HPLC (Owater (0.1% TFA)-ACN), lyophilized to give (4-benzyloxy-4-oxo-butyl)-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-(2-tert-butoxy-2-oxo-ethyl)-methyl-ammonium; 2,2,2-trifluoroacetate (2.3 g, 3.8 mmol, 81.35% yield) as light yellow gum. MS [(M)+]: 491.4. Step 4: (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)-methyl-ammonium; 2,2,2-trifluoroacetate To a solution of (4-benzyloxy-4-oxo-butyl)-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-(2-tert-butoxy-2-oxo-ethyl)-methyl-ammonium; 2,2,2-trifluoroacetate (2.3 g, 3.8 mmol) in Methanol (30 mL) was added palladium on charcoal (460 mg, 0.43 mmol) and palladium hydroxide on charcoal (460.0 mg, 0.33 mmol) under N2. The mixture was degassed and then stirred at 30° C. for 3 h under hydrogen (760 mmHg). The mixture was filtered through celite pad, the solid was washed with MeOH (10 mL×3). The filtrate was concentrated under vacuum to give (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)-methyl-ammonium; 2,2,2-trifluoroacetate (1.87 g, 3.63 mmol, 95.55% yield). MS [(M)+]: 401.1. The following example was prepared in analogy to Intermediate R17. MSStartingEx#NameStructure[M + H]+MaterialIntermediate R183-(tert-butoxycarbonyl- amino)propyl-(2-tert- butoxy-2-oxo-ethyl)- (3-carboxypropyl)- methyl-ammonium; bromide389.3benzyl 4- aminobutanoate; hydrochloride and 3-(Boc- amino)propyl bromide Intermediate R19 tert-butyl N-[3-[4-(3-aminopropyl)-2-pyridyl]propyl]carbamate; hydrochloride Step 1: benzyl N-prop-2-ynylcarbamate To a mixture of propargylamine (5.0 g, 90.78 mmol, 1.0 eq) and sodium hydrogen carbonate (38.13 g, 453.89 mmol, 5.0 eq) in EtOAc (100 mL)/water (100 mL) was added dropwise benzyl chloroformate (14.25 mL, 99.85 mmol, 1.1 eq) at 0° C., then the mixture was stirred for 1 h at 0° C. The solution was poured into water (50 mL) and extracted with EtOAc, washed with brine, dried over Na2SO4, concentrated in vacuo and purified by flash chromatography to afford benzyl N-prop-2-ynylcarbamate (15.4 g, 81.39 mmol, 89.66% yield) as light yellow oil. MS [(M+H)+]: 189.9 Step 2: benzyl N-[3-(2-bromo-4-pyridyl)prop-2-ynyl]carbamate A mixture of benzyl N-prop-2-ynylcarbamate (6.0 g, 31.7 mmol, 1.0 eq), copper(I) iodide (0.32 mL, 9.51 mmol, 0.3 eq) and tetrakis(triphenylphosphine)palladium(0) (1.83 g, 1.59 mmol, 0.05 eq) in toluene (20 mL) and was added 2-bromo-4-iodopyridine (9.0 g, 31.7 mmol, 1.0 eq), tetrabutylammonium fluoride in THE (31.7 mL, 31.7 mmol, 1.0 eq) and triethylamine (13.26 mL, 95.11 mmol, 3.0 eq) under N2, then the mixture was stirred at 25° C. for 16 h. The reaction was no further work-up and purified by flash chromatography to obtain benzyl N-[3-(2-bromo-4-pyridyl)prop-2-ynyl]carbamate (9.1 g, 26.36 mmol, 83.16% yield) as brown oil. MS [(M+H)+]: 345.0 Step 3: tert-butyl N-[3-[4-[3-(benzyloxycarbonylamino)prop-1-ynyl]-2-pyridyl]prop-2-ynyl]carbamate To a mixture of N—BOC-propargylamine (4.09 g, 26.36 mmol, 1.0 eq), copper(I) iodide (0.16 mL, 4.78 mmol, 0.18 eq) and bis(triphenylphosphine)palladium(II) chloride (0.93 g, 1.32 mmol, 0.05 eq) in DMF (10 mL) was added benzyl N-[3-(2-bromo-4-pyridyl)prop-2-ynyl]carbamate (9.1 g, 26.36 mmol, 1.0 eq) and triethylamine (91.0 mL, 652.89 mmol, 24.77 eq) under N2, then the mixture was stirred at 50° C. for 16 h. The reaction was concentrated under vacuum and purified by flash chromatography to obtain tert-butyl N-[3-[4-[3-(benzyloxycarbonylamino)prop-1-ynyl]-2-pyridyl]prop-2-ynyl]carbamate (9.4 g, 22.41 mmol, 85.0% yield) as brown oil. MS [(M+H)+]: 420.2 Step 4: tert-butyl N-[3-[4-(3-aminopropyl)-2-pyridyl]propyl]carbamate; hydrochloride To a mixture of tert-butyl N-[3-[4-[3-(benzyloxycarbonylamino)prop-1-ynyl]-2-pyridyl]prop-2-ynyl]carbamate (4.0 g, 9.54 mmol, 1.0 eq) and ammonium hydroxide (1.0 mL, 9.54 mmol, 1.0 eq) in methanol (50 mL) was added wet palladium 10% on activated carbon (400.0 mg, 9.54 mmol, 1.0 eq) at 25° C. The mixture was degassed and purged with H2 for 3 times. Then the mixture was stirred under hydrogen for 48 h. The reaction was filtered through a pad of celite and the filtrate was concentrated in vacuo. The residue was purified by prep-HPLC and lyophilized to obtain the white solid, then dissolved in HCl solution (5%, 100 mL) and lyophilized to afford tert-butyl N-[3-[4-(3-aminopropyl)-2-pyridyl]propyl]carbamate; hydrochloride (2090.86 mg, 6.34 mmol, 64.54% yield) as light yellow oil. MS [(M+H)+]: 294.2 Intermediate R19 bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide Step 1: benzyl 4-[bis[4-(tert-butoxycarbonylamino)butyl]amino]butanoate To a mixture of 4-(tert-butoxycarbonylamino)butyl 4-methylbenzenesulfonate (4934.05 mg, 14.37 mmol) and benzyl 4-aminobutanoate; hydrochloride (1.5 g, 6.53 mmol) in ACN (60 mL) was added potassium carbonate (2707.56 mg, 19.59 mmol). The reaction mixture was stirred at 50° C. for 16 h. The reaction mixture was filtered, the filtrate was concentrated in vacuum to give the residue, which was purified by reversed phase-HPLC and lyophilized to afford benzyl 4-[bis[4-(tert-butoxycarbonylamino)butyl]amino]butanoate (2000.0 mg, 3.25 mmol, 57.17% yield) as yellow oil. MS [(M+H)+]: 536.4. Step 2: (4-benzyloxy-4-oxo-butyl)-bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; bromide To a mixture of benzyl 4-[bis[4-(tert-butoxycarbonylamino)butyl]amino]butanoate (2000.0 mg, 3.25 mmol) and N-ethyl-N-isopropylpropan-2-amine (839.74 mg, 6.5 mmol) in ACN (10 mL) was added and tert-butyl bromoacetate (633.67 mg, 3.25 mmol) at 20° C. The reaction mixture was stirred at 50° C. for 16 h. The reaction mixture was filtered, the filtrate was concentrated in vacuum to give the residue, which was purified by reversed phase-HPLC and lyophilized to afford (4-benzyloxy-4-oxo-butyl)-bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; bromide (400.0 mg, 0.55 mmol, 18.92% yield) as yellow solid. MS [(M)+]: 650.4. Step 3: bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide To a solution of (4-benzyloxy-4-oxo-butyl)-bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; bromide (500.0 mg, 0.68 mmol) in Methanol (20 mL) was added wet palladium on active carbon (50.0 mg, 10%) at 30° C. under N2, then the reaction mixture was stirred at 30° C. for 16h under H2 (balloon, 15 psi). The reaction mixture was filtered, the filtrate was concentrated in vacuum to give bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide (250.0 mg, 0.39 mmol, 57.03% yield) as colorless oil. MS [(M)+]: 560.8. Intermediate R20 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoic acid Step 1: benzyl 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoate To a stirred mixture of benzyl 4-aminobutanoate; hydrochloride (4.5 g, 19.59 mmol) and potassium carbonate (5.42 g, 39.18 mmol) in ACN (100 mL) was added 3-(BOC-amino)propyl bromide (11.66 g, 48.98 mmol) at 20° C., then the mixture was stirred at 60° C. for 16 h. The solution was cooled to room temperature and filtered. The filtrate was concentrated in vacuum, purified by reversed phase-HPLC and lyophilized to afford benzyl 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoate (3.5 g, 6.89 mmol, 35.19% yield) as colorless oil. MS [(M+H)+]: 508.1. Step 2: 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoic acid To a solution of benzyl 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoate (1.3 g, 2.56 mmol) in Methanol (50 mL) was added wet palladium on active carbon (0.27 g, 0.26 mmol, 10%) at 20° C. under N2, then the mixture was stirred under H2 (balloon) at 20° C. for 16 h. The solution was filtered with diatomite and concentrated to afford 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoic acid (706.0 mg, 1.69 mmol, 66.03% yield) as colorless oil. MS [(M+H)+]: 418.3. Intermediate R21 bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide Step 1: benzyl 4-[bis[2-(tert-butoxycarbonylamino)ethyl]amino]butanoate To a solution of benzyl 4-aminobutanoate; hydrochloride (11.0 g, 47.89 mmol) in ACN (100 mL) was added tert-butyl N-(2-bromoethyl)carbamate (32.2 g, 143.67 mmol) and DIEA (33.36 mL, 191.55 mmol) at 10° C., then the mixture was stirred at 50° C. for 48 h. The solution was concentrated, the residue was purified by reversed phase-HPLC and lyophilized to afford benzyl 4-[bis[2-(tert-butoxycarbonylamino)ethyl]amino]butanoate (6.5 g, 13.55 mmol, 28.3% yield) as light yellow oil. MS [(M+H)+]: 480.3. Step 2: (4-benzyloxy-4-oxo-butyl)-bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; bromide To a mixture of benzyl 4-[bis[2-(tert-butoxycarbonylamino)ethyl]amino]butanoate (5.0 g, 10.43 mmol) in ACN (50 mL) was added tert-butyl bromoacetate (12.2 g, 62.55 mmol) and DIEA (5.45 mL, 31.28 mmol) at 10° C. The reaction mixture was stirred at 60° C. for 16 h. The mixture was concentrated under vacuum and the residue was purified by reversed phase-HPLC and lyophilized to afford (4-benzyloxy-4-oxo-butyl)-bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; bromide (5.5 g, 8.17 mmol, 78.33% yield) as white solid. MS [(M)+]: 594.3. Step 3: bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide To a solution of (4-benzyloxy-4-oxo-butyl)-bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)ammonium; formate (2.5 g, 3.91 mmol) in Methanol (50 mL) was added wet palladium on active carbon (250 mg, 10%) under N2. Then the mixture was stirred under Hydrogen (15 Psi) at 20° C. for 16 h. The mixture was filtered with diatomite and the filtrate was concentrated to afford bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide (1.2 g, 2.05 mmol, 52.54% yield) as white solid. MS [(M)+]: 504.4. Intermediate R22 tert-butyl 2-[1-[3-(tert-butoxycarbonylamino)propyl]piperazin-1-ium-1-yl]acetate; formate Step 1: benzyl 4-[3-(tert-butoxycarbonylamino)propyl]piperazine-1-carboxylate To a solution of 1-CBZ-piperazine (5.0 g, 22.7 mmol, 1.0 eq) in MeCN (100 mL) was added triethylamine (3.16 mL, 22.7 mmol, 1.0 eq) and 3-(BOC-amino)propyl bromide (5.68 g, 23.83 mmol, 1.05 eq), then the mixture was stirred at 25° C. for 16 h. The mixture was concentrated in vacuo and purified by flash chromatography to obtain benzyl 4-[3-(tert-butoxycarbonylamino)propyl]piperazine-1-carboxylate (5.2 g, 13.78 mmol, 60.69% yield) as light brown solid. MS [(M+H)+]: 378.3 Step 2: benzyl 4-[3-(tert-butoxycarbonylamino)propyl]piperazine-1-carboxylate To a solution of benzyl 4-[3-(tert-butoxycarbonylamino)propyl]piperazine-1-carboxylate (5.2 g, 13.78 mmol, 1 eq) in MeCN (100 mL) was added triethylamine (1.92 mL, 13.78 mmol, 1 eq) and tert-butyl bromoacetate (5.37 g, 27.55 mmol, 2 eq), then the mixture was stirred at 50° C. for 16 h. The mixture was concentrated in vacuo and purified by prep-HPLC to obtain benzyl 4-[3-(tert-butoxycarbonylamino)propyl]-4-(2-tert-butoxy-2-oxo-ethyl)piperazin-4-ium-1-carboxylate; formate (4.0 g, 7.44 mmol, 58.94% yield) as light yellow oil. MS [(M+]: 492.4 Step 3: tert-butyl 2-[1-[3-(tert-butoxycarbonylamino)propyl]piperazin-1-ium-1-yl]acetate; formate To a solution of benzyl 4-[3-(tert-butoxycarbonylamino)propyl]-4-(2-tert-butoxy-2-oxo-ethyl)piperazin-4-ium-1-carboxylate (4.0 g, 8.12 mmol, 1.0 eq) in THE (40 mL) with palladium (0.39 mL, 0.38 mmol, 0.05 eq) was treated under Hydrogen (16.24 mg, 8.12 mmol, 1.0 eq) at 25° C. for 16 h. The cat. was filtered and washed. The filtrate was concentrated in vacuo and purified by prep-HPLC to obtain tert-butyl 2-[1-[3-(tert-butoxycarbonylamino)propyl]piperazin-1-ium-1-yl]acetate (1.5 g, 3.72 mmol, 51.53% yield)tert-butyl 2-[1-[3-(tert-butoxycarbonylamino)propyl]piperazin-1-ium-1-yl]acetate; formate (1.5 g, 3.72 mmol, 51.53% yield) as white solid. MS [(M+]: 358.3 The following example was prepared in analogy to Intermediate R22. MSStartingEx#NameStructure[M+]MaterialIntermediate R23tert-butyl 3-[[1-(2- tert-butoxy-2-oxo- ethyl)piperazin-1- ium-1- yl]methyl]azetidine- 1-carboxylate; formate370.61-BOC-3- (bromomethyl) azetidine Intermediate R24 1-[2-(tert-butoxycarbonylamino)ethyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylic acid; 2,2,2-trifluoroacetate Step 1: benzyl 1-[2-(tert-butoxycarbonylamino)ethyl]piperidine-4-carboxylate To a solution of benzyl piperidine-4-carboxylate; hydrochloride (6.0 g, 23.46 mmol, 1.0 eq) and potassium carbonate (6.48 g, 46.92 mmol) in DMF (120 mL) was added tert-butyl N-(2-bromoethyl)carbamate (6.31 g, 28.15 mmol) at 20° C. and stirred at 20° C. for 16 h. The mixture was poured into water (200 mL) and then extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine (80 mL×3), dried over sodium sulfate, filtered, the filtrate was concentrated under vacuum to give benzyl 1-[2-(tert-butoxycarbonylamino)ethyl]piperidine-4-carboxylate (9.23 g, 25.46 mmol, 97.69% yield) as a light yellow oil. MS [(M+H)+]: 363.2. Step 2: benzyl 1-[2-(tert-butoxycarbonylamino)ethyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylate; 2,2,2-trifluoroacetate To a solution of benzyl 1-[2-(tert-butoxycarbonylamino)ethyl]piperidine-4-carboxylate (9.23 g, 25.46 mmol), NaI (381.7 mg, 2.55 mmol) and DIEA (6.65 mL, 38.2 mmol) in DMF (90 mL) was added tert-butyl bromoacetate (5.96 g, 30.56 mmol) at 20° C. and stirred at 20° C. for 16 h. The mixture was concentrated under vacuum, the residue was purified by reversed phase-HPLC (water (0.1% TFA)-ACN) and lyophilized to give benzyl 1-[2-(tert-butoxycarbonylamino)ethyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylate; 2,2,2-trifluoroacetate (5.7 g, 9.65 mmol, 46.87% yield) as a light yellow gum. MS [(M)+]: 477.2. Step 3: 1-[2-(tert-butoxycarbonylamino)ethyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylic acid; 2,2,2-trifluoroacetate To a solution of benzyl 1-[2-(tert-butoxycarbonylamino)ethyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylate; 2,2,2-trifluoroacetate (6.43 g, 10.89 mmol) in methanol (120 mL) was added palladium hydroxide on charcoal (1528.98 mg, 1.09 mmol, 10%) and palladium on charcoal (1.13 mL, 1.09 mmol, 10%) under N2. The mixture was degassed and then stirred under hydrogen (760 mmHg) at 25° C. for 4 h. The mixture was filtered through the celite pad, the solid was washed with MeOH (20 mL×4). The combined filtrate was concentrated under vacuum to give 1-[2-(tert-butoxycarbonylamino)ethyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylic acid; 2,2,2-trifluoroacetate (4.1 g, 8.19 mmol, 67.71% yield) as a white solid. MS [(M)+]: 387.2. Example L1 N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate Example L1 Step 1: tert-butyl 2-[4-[4-[2-[[3-chloro-4-(piperazine-1-carbonyl)phenyl]carbamoyl]-3-methyl-imidazol-4-yl]phenyl]-3-methyl-pyrazol-1-yl]acetate; formic acid To a solution of tert-butyl 4-[4-[[5-[4-[1-(2-tert-butoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carboxylate (200.0 mg, 0.28 mmol) in DCM (5 mL) was added TFA (0.25 mL, 3.24 mmol) at 25° C. and for 16 h. The mixture was concentrated in vacuum and purified by reversed phase HPLC (water (0.1% FA)-ACN) and lyophilized to afford tert-butyl 2-[4-[4-[2-[[3-chloro-4-(piperazine-1-carbonyl) phenyl] carbamoyl]-3-methyl-imidazol-4-yl] phenyl]-3-methyl-pyrazol-1-yl] acetate; formic acid (70.0 mg, 0.11 mmol, 40.67% yield) as white solid. MS [(M+H)+]: 618.3 Step 2: tert-butyl 2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]phenyl]-3-methyl-pyrazol-1-yl]acetate; formate To a solution of tert-butyl 2-[4-[4-[2-[[3-chloro-4-(piperazine-1-carbonyl)phenyl]carbamoyl]-3-methyl-imidazol-4-yl]phenyl]-3-methyl-pyrazol-1-yl]acetate; formic acid (70.0 mg, 0.11 mmol), 1,1-dimethylpiperidin-1-ium-4-carboxylic acid; iodide (60.1 mg, 0.21 mmol), DIEA (0.09 mL, 0.53 mmol) in DMF (2 mL) was added HATU (60.11 mg, 0.16 mmol) at 0° C. and stirred at 0° C. for 0.5 h. The mixture was purified by reversed phase-HPLC (water (0.1% FA)-ACN) and lyophilized to afford tert-butyl 2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]phenyl]-3-methyl-pyrazol-1-yl]acetate; formate (60.0 mg, 0.08 mmol, 75.07% yield) as yellow solid. MS [(M)+]: 757.4. Step 3: 2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]phenyl]-3-methyl-pyrazol-1-yl]acetic acid; chloride To a solution of tert-butyl 2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]phenyl]-3-methyl-pyrazol-1-yl]acetate; formate (60.0 mg, 0.07 mmol) in DCM (2 mL) was added TFA (2.0 mL, 25.96 mmol) at 25° C. and stirred for 0.5 h. The mixture was concentrated in vacuum and purified by reversed phase HPLC (water (0.1% HCl)-ACN), lyophilized to afford 2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]phenyl]-3-methyl-pyrazol-1-yl]acetic acid; chloride (40.0 mg, 0.05 mmol, 76.27% yield) as white solid. MS [(M)+]: 701.4. Step 4: N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate To a solution of 2-[4-[4-[2-[[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]carbamoyl]-3-methyl-imidazol-4-yl]phenyl]-3-methyl-pyrazol-1-yl]acetic acid; chloride (70.0 mg, 0.09 mmol) in DMF (2 mL) was added DIEA (0.05 mL, 0.28 mmol) and 2-chloro-1-methylpyridinium iodide (36.36 mg, 0.14 mmol) at 25° C. and stirred for 20 min. Then 6-fluoropyridazin-3-amine (16.1 mg, 0.14 mmol) was added to the solution and stirred at 80° C. for 8 h. The mixture was purified by Prep-HPLC (Phenomenex Luna C18 75×30 mm×3 urn, water (0.225 FA)-ACN, B=190%-39%; FowRat: 25 ml/min) to afford N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; formate (28.9 mg, 0.03 mmol, 35.07% yield) as yellow solid. MS [(M)+]: 796.0. The following examples were prepared in analogy to Examples L1. MSStartingEx#NameStructure[M]+MaterialExample L2N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[3- fluoro-4-[1-[2-[(6- fluoropyridazin-3- yl)amino]-2-oxo-ethyl]- 3-methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate814.2Intermediate O5 and 6- fluoropyridazin- 3-amineExample L3N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[3-methyl-1- [2-[(6-methylpyridazin- 3-yl)amino]-2-oxo- ethyl]pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate828.4Intermediate O6 and 3- amino-6- methylpyridazineExample L4N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[1-[2-[(6- methoxypyridazin-3- yl)amino]-2-oxo-ethyl]- 3-methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate844.4Intermediate O6 and 3- amino-6- methoxypyridazineExample L5N-[3-chloro-4-[4-(1,1- dimethylpiperidin-1-ium- 4-carbonyl)piperazine-1- carbonyl]phenyl]-5-[2,3- difluoro-4-[3-methyl-1- [2-[(6- morpholinopyridazin-3- yl)amino]-2-oxo- ethyl]pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carboxamide; formate899.2Intermediate O6 and 6- morpholinopyridazin- 3-amine Example L6 cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formate Step 1: 2-[4-[4-[2-[[4-(4-tert-butoxycarbonylpiperazine-1-carbonyl)-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazol-1-yl]acetic acid To a solution of tert-butyl 4-[4-[[5-[4-[1-(2-tert-butoxy-2-oxo-ethyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carboxylate (300.0 mg, 0.4 mmol) in Water (3 mL) and THF (3 mL) was added sodium hydroxide (47.7 mg, 1.19 mmol). The mixture was stirred at 10° C. and for 16 h. The reaction mixture was pour into water (10 mL), acidified with aq. HCl (1N) to pH=6, extracted with EA (10 mL×2). The combined organic layer was washed with brine, dried over sodium sulfate, concentrated in vacuum to afford 2-[4-[4-[2-[[4-(4-tert-butoxycarbonylpiperazine-1-carbonyl)-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazol-1-yl]acetic acid (220.0 mg, 79.2% yield) as off-white solid. MS [(M+H)+]: 698.3. Step 2: tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate To a solution of 2-[4-[4-[2-[[4-(4-tert-butoxycarbonylpiperazine-1-carbonyl)-3-chloro-phenyl]carbamoyl]-3-methyl-imidazol-4-yl]-2,3-difluoro-phenyl]-3-methyl-pyrazol-1-yl]acetic acid (220.0 mg, 0.32 mmol) and 6-fluoropyridazin-3-amine (142.55 mg, 1.26 mmol) in DMF (3 mL) was added DIEA (0.16 mL, 0.95 mmol) and BopCl (120.33 mg, 0.47 mmol). The mixture was stirred at 30° C. and for 1 h. The mixture was purified by reversed phase-HPLC (water (0.1% FA)-ACN, B=10%-60%) and lyophilized to afford tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (200.0 mg, 0.25 mmol, 69.1% yield) as light yellow solid. MS [(M+H)+]: 793.2 Step 3: N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide To a solution of tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (200.0 mg, 0.25 mmol) in DCM (2 mL) was added TFA (0.4 mL, 5.19 mmol) at 30° C. and stirred at 30° C. for 3 h. The reaction mixture was pour into water, basified with aq.NaHCO3to pH=8, extracted with EA (10 mL×2). The combined organic layer was washed with brine, dry over sodium sulfate, concentrated in vacuum to afford N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (120.0 mg, 0.17 mmol, 63.3% yield) as light yellow solid. MS [(M+H)+]: 693.2. Step 4: tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]methyl]azetidine-1-carboxylate; formic acid To a mixture of N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (80.0 mg, 0.12 mmol) and (1s,4s)-1-(2-(tert-butoxy)-2-oxoethyl)-1-((1-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-4-carboxypiperidin-1-ium; 2,2,2-trifluoroacetate (57.28 mg, 0.14 mmol) in DMF (1 mL) was added Bop-C1 (29.32 mg, 0.12 mmol) and DIEA (0.06 mL, 0.35 mmol). The reaction mixture was stirred at 30° C. for 1 h. The reaction mixture was purified by reversed phase HPLC (water (0.1% FA)-ACN, B=60%-90%) and lyophilized to give tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-[2-[(6-fluoropyridazin-3-yl)amino]-2-oxo-ethyl]-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]methyl]azetidine-1-carboxylate; formic acid (40.0 mg, 0.04 mmol, 25.5% yield) as light yellow solid. MS [(M)+]: 1087.5. Example M1 N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methyl-3-pyridyl)carbamoylamino]ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide formate Step 1: tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate 3-(((ethylimino)methylene)amino)-N,N-dimethylpropan-1-amine hydrochloride (4.61 g, 24 mmol, Eq: 1.05) and N-ethyl-N-isopropylpropan-2-amine (14.8 g, 20 ml, 114 mmol, Eq: 5) were added to a solution of 2-chloro-4-(5-(2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamido)benzoic acid (10.8 g, 22.9 mmol, Eq: 1) and 1H-benzo[d][1,2,3]triazol-1-ol (3.25 g, 24 mmol, Eq: 1.05) in DMA (57.2 ml) and stirred for 18 hours at room temperature. The mixture was poured into 100 mL and extracted with EtOAc (50 mL×4). The organic layer was washed with 50 mL with brine, dried over sodium sulfate, and concentrated in vacuo. The residue was used in the next step without purification. Step 2: 4-[4-[[5-[4-[1-[2-[tert-butyl(dimethyl)silyl]oxyethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carboxylic acid tert-butyl ester 4-[2-chloro-4-[[5-[2,3-difluoro-4-(5-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylic acid tert-butyl ester (1.9 g, 2.88 mmol, 1.000 eq) was dissolved in acetonitrile (60 mL). cesium carbonate (1.41 g, 4.32 mmol, 1.500 eq) was added followed by tert-butyl(2-iodoethoxy)dimethylsilane (1.07 g, 3.75 mmol, 1.300 eq). The mixture was heated at 70° C. over night. The cesium carbonate was filtered off. The mixture was purified by flash chromatography (100% heptane to 100% EtOAc). The mixture of regioisomers was separated by Prep-SFC chiral. The structures of regioisomeres were elucitated by NMR. The title compound (814 mg, 32.9%) was isolated as colorless oil. MS [(M+H-Buten)+]: 742.2. Step 3: 4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-hydroxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylic acid tert-butyl ester 4-[4-[[5-[4-[1-[2-[tert-butyl(dimethyl)silyl]oxyethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carboxylic acid tert-butyl ester (814 mg, 1.02 mmol, 1.000 eq) was dissolved in N,N-dimethylformamide (4 mL) and water (1 mL). Ammonium fluoride (377.61 mg, 10.2 mmol, 10.000 eq) was added at room temperature. The mixture was stirred at 60° C. over two days. The reaction mixture was poured into 100 mL brine and extracted two times with 100 mL EtOAc. The organic layers were combined, dried with Na2SO4, filtered and evaporated to dryness to afford the title compound (501.6 mg, 69.76%) as colorless waxy solid. MS [(M+H)+]: 684.3. Step 4: 4-[2-chloro-4-[[5-[2,3-difluoro-4-[3-methyl-1-(2-methylsulfonyloxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylic acid tert-butyl ester To 4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-hydroxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylic acid tert-butyl ester (468 mg, 0.664 mmol, 1.000 eq) dissolved in dichloromethane (30 mL) was added methanesulfonic anhydride (173.39 mg, 188.46 uL, 0.995 mmol, 1.500 eq) and DIEA (171.52 mg, 231.78 uL, 1.33 mmol, 2.000 eq). The mixture was stirred at room temperature for 3h. The reaction mixture was poured into 30 mL brine and extracted three times with 30 mL DCM. The organic layers were combined, dried over MgSO4and concentrated to dryness. The title compound was obtained as a light brown solid with an assumed purity of 85% and was used without further purification. MS [(M+H)+]: 762.4. Step 5: 4-[4-[[5-[4-[1-(2-aminoethyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carboxylic acid tert-butyl ester 4-[2-chloro-4-[[5-[2,3-difluoro-4-[3-methyl-1-(2-methylsulfonyloxyethyl)pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylic acid tert-butyl ester (585 mg, 0.652 mmol, 1.000 eq) was treated with 2 M ammonia solution (2M in iPrOH) (12.61 g, 16.31 mL, 32.62 mmol, 50.000 eq) at 70° C. over 2 days. The title compound was obtained as a light brown gum with an assumed purity of 89% and was used without further purification. MS [(M+H)+]: 683.4 Step 6: tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methyl-3-pyridyl)carbamoylamino]ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate 4-[4-[[5-[4-[1-(2-aminoethyl)-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]-2-chloro-benzoyl]piperazine-1-carboxylic acid tert-butyl ester (33.7 mg, 0.044 mmol, 1.000 eq) was dissolved in N,N-dimethylformamide (1.5 mL). 5-isocyanato-2-methyl-pyridine (8.83 mg, 0.066 mmol, 1.500 eq) and DIEA (28.37 mg, 38.34 uL, 0.220 mmol, 5.000 eq) were added and the mixture was stirred over night at room temperature. The mixture was concentrated to dryness and used crude for next step. Step 7: N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methyl-3-pyridyl)carbamoylamino]ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide Crude tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methyl-3-pyridyl)carbamoylamino]ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate was treated with an excess of HCl 4N in dioxan (30 eq) at room temperature over night. The crude mixture was concentrated and purified by Prep-HPLC to afford the title compound (20 mg, 59.7%) as white amorph freeze-dried solid. MS [(M+H)+]: 717.2. Step 8: N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methyl-3-pyridyl)carbamoylamino]ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide formate N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methyl-3-pyridyl)carbamoylamino]ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (20 mg, 0.028 mmol, 1.000 eq) was dissolved in dichloromethane (1 mL). 1,1-dimethylpiperidin-1-ium-4-carboxylic acid iodide (24 mg, 0.069 mmol, 2.500 eq), DIEA (36.04 mg, 48.7 uL, 0.279 mmol, 10.000 eq) and propylphosphonic anhydride (50% solution) (53.24 mg, 0.084 mmol, 3.000 eq) were added and the mixture was stirred at room temperature over night. The crude mixture was concentrated, dissolved in MeOH and purified by Prep-HPLC to afford N-[3-chloro-4-[4-(1,1-dimethylpiperidin-1-ium-4-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[3-methyl-1-[2-[(6-methyl-3-pyridyl)carbamoylamino]ethyl]pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide formate (1.2 mg, 4.5% yield) as white amorph freeze-dried solid. MS [M+]: 856.5. Example N1 azetidin-3-ylmethyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formic acid; formate Step 1: N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride To a mixture of tert-butyl 4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (100.0 mg, 0.14 mmol) in 1,4-Dioxane (2 mL) was added HCl (0.5 mL, 2.0 mmol, 4M in 1,4-dioxane) at 30° C. and stirred at 30° C. for 4 h. The reaction mixture was concentrated in vacuum to afford crude N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride (90.0 mg, 0.14 mmol, 98.6% yield) as light yellow solid. MS [(M+H)+]: 630.0. Step 2: (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; 2,2,2-trifluoroacetate To a solution of N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride (90.0 mg, 0.14 mmol), (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)-methyl-ammonium; 2,2,2-trifluoroacetate (115.79 mg, 0.23 mmol) and DIEA (0.1 mL, 0.6 mmol) in DMF (0.5 mL) was added HATU (85.57 mg, 0.23 mmol) at 0° C. The mixture was stirred at 0° C. for 0.5 h. The mixture was quenched with water (1 drop) and then concentrated under vacuum to afford a residue, which was purified by reversed phase and lyophilized to give (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; 2,2,2-trifluoroacetate (70 mg, 0.06 mmol, 41.4% yield) as a light yellow solid. MS [(M)+]: 1012.4. Step 3: azetidin-3-ylmethyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formic acid; formate To a solution of (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; 2,2,2-trifluoroacetate (65.0 mg, 0.06 mmol) in DCM (0.5 mL) was added TFA (2.6 mL, 33.75 mmol) at 30° C. and stirred for 16h. The mixture was concentrated under vacuum, the residue was purified by prep-HPLC (Column: Unisil 3-100 C18 Ultra 150×50 mm×3 um; Condition: water (0.225% FA)-ACN; Begin B=16%, End B=36%; Gradient Time(min) 10 min) and lyophilized to give azetidin-3-ylmethyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-3-fluoro-2-methyl-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formic acid; formate (33.1 mg, 0.03 mmol, 57.11% yield) as a white solid. MS [(M)+]: 856.1. The following examples were prepared in analogy to Example N1. MSStartingEx#NameStructure[M]+MaterialExam- ple N22-[1-(azetidin-3- ylmethyl)-4-[4- [2-chloro- 4-[[5-[4-[1-[2- (difluoromethoxy) ethyl]-5-methyl- pyrazol-4-yl]- 3-fluoro-2-methyl- phenyl]-1-methyl- imidazole-2- carbonyl]amino] benzoyl] piperazine-1- carbonyl]piperidin- 1-ium-1- yl]acetic acid; formate868.1Inter- mediate O7 and inter- mediate R14Exam- ple N33-aminopropyl- (carboxymethyl)- [4-[4- [2-chloro-4- [[5-[4-[1-[2- (difluoromethoxy) ethyl]- 5-methyl-pyrazol- 4-yl]-3-fluoro-2- methyl-phenyl]- 1-methyl- imidazole-2- carbonyl]amino] benzoyl] piperazin-1-yl]-4- oxo-butyl]-methyl- ammonium; formate844.6Inter- mediate O7 and inter- mediate R18 Example N4 cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetate Step 1: tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-car bonyl]amino]benzoyl]piperazine-1-carboxylate 3-(((ethylimino)methylene)amino)-N,N-dimethylpropan-1-amine hydrochloride (4.61 g, 24 mmol, Eq: 1.05) and N-ethyl-N-isopropylpropan-2-amine (14.8 g, 20 ml, 114 mmol, Eq: 5) were added to a solution of 2-chloro-4-(5-(2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl)-1-methyl-1H-imidazole-2-carboxamido)benzoic acid (10.8 g, 22.9 mmol, Eq: 1) and 1H-benzo[d][1,2,3]triazol-1-ol (3.25 g, 24 mmol, Eq: 1.05) in DMA (57.2 ml) and stirred for 18 hours at room temperature. The mixture was poured into 100 mL and extracted with EtOAc (50 mL×4). The organic layer was washed with 50 mL with brine, dried over sodium sulfate, and concentrated in vacuo. The residue was used in the next step without purification. Step 2: tert-butyl 4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate To a mixture of tert-butyl 4-[2-chloro-4-[[5-[2,3-difluoro-4-(3-methyl-1H-pyrazol-4-yl)phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (700.0 mg, 1.09 mmol, 1.0 eq) and 2-(difluoromethoxy)ethyl 4-methylbenzenesulfonate (349.42 mg, 1.31 mmol, 1.2 eq) in DMF (10 mL) was added potassium carbonate (453.43 mg, 3.28 mmol, 3.0 eq). The reaction mixture was stirred at 60° C. for 16 h. The reaction mixture was filtered, the filtrate was purified by prep-HPLC (FA) to afford a mixture of isomers. The mixture was further purified by SFC to afford tert-butyl 4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (125.0 mg, 0.17 mmol, 15.57% yield) as yellow solid and the title compound tert-butyl 4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-5-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (125.0 mg, 0.17 mmol, 15.57% yield) as yellow solid. MS [(M+H)+]: 734.3. Step 3: N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride A mixture of tert-butyl 4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carboxylate (125.0 mg, 0.17 mmol, 1.0 eq) in HCl (4N in dioxane) (5.0 mL, 5.0 mmol, 29.37 eq) was stirred at 30° C. for 15 h. The reaction mixture was concentrated in vacuum to afford N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride (100.0 mg, 0.15 mmol, 92.63% yield) as yellow solid. MS [(M+H)+]: 634.1. Step 4: tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]methyl]azetidine-1-carboxylate To a mixture of 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylic acid (156.54 mg, 0.38 mmol, 1.2 eq) and N-ethyl-N-isopropylpropan-2-amine (0.16 mL, 0.95 mmol, 3.0 eq) in DMF (3 mL) was added 2-chloro-1-methylpyridin-1-ium iodide (96.71 mg, 0.38 mmol, 1.2 eq). The reaction mixture was stirred at 30° C. for 0.5 h. Then, N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carboxamide (200.0 mg, 0.32 mmol, 1.0 eq) was added to the above solution. The reaction mixture was stirred at 30° C. for another 3.5 h. The reaction mixture was purified by prep-HPLC (FA) to afford tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]methyl]azetidine-1-carboxylate (170.0 mg, 0.17 mmol, 52.35% yield) as yellow solid. MS [(M+H)+]: 1028.5. Step 5: cis 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetate A solution of tert-butyl 3-[[1-(2-tert-butoxy-2-oxo-ethyl)-4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]methyl]azetidine-1-carboxylate (150.0 mg, 0.15 mmol, 1.0 eq) in DCM (2 mL) was added trifluoroacetic acid (2.0 mL, 25.96 mmol, 178.18 eq). The reaction mixture was stirred at 30° C. for 16 h. The reaction mixture was concentrated in vacuum to give the residue, which was purified by prep-HPLC (FA) to afford 2-[1-(azetidin-3-ylmethyl)-4-[4-[2-chloro-4-[[5-[4-[1-[2-(difluoromethoxy)ethyl]-3-methyl-pyrazol-4-yl]-2,3-difluoro-phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetic acid; formate (82.3 mg, 0.09 mmol, 58.8% yield) as white solid. Cis and trans isomers were separated by SFC chiral to afford the cis title compound (15.2 mg, 42.57%) as white powder. MS [(M+H)+]: 872.3 Example O1 2-[1-(azetidin-3-ylmethyl)-4-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]-4-piperidyl]-methyl-carbamoyl]piperidin-1-ium-1-yl]acetic acid formate To N-[3-chloro-4-[4-(methylamino)piperidine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (8.7 mg, 0.014 mmol, 1 eq) dissolved in dichloromethane (2 mL), was added PyAOP reagent (9.49 mg, 0.018 mmol, 1.300 eq) and DIEA (7.24 mg, 9.8 uL, 0.056 mmol, 4.000 eq) followed by 1-[(1-tert-butoxycarbonylazetidin-3-yl)methyl]-1-(2-tert-butoxy-2-keto-ethyl)piperidin-1-ium-4-carboxylic acid; 2,2,2-trifluoroacetate (11.8 mg, 0.0224 mmol, 1.6 eq). The mixture was stirred at room temperature over night. The mixture was then treated with an excess of HCl 4N in dioxane (15.31 mg, 12.76 uL, 0.420 mmol, 30.000 eq) over night at room temperature The mixture was then concentrated and directly purified by preparative HPLC to afford the title compound (1.7 mg, 13.3% yield). MS [M-]: 862.5. The following examples were prepared in analogy to Example O1. StartingEx#NameStructureMSMaterialExam- ple O22-[1-(azetidin-3- ylmethyl)-4- [(1S,5R) 6-[[2-chloro-4-[[5- [2,3-difluoro-4-[1- (2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1- methyl- imidazole-2- carbonyl]amino] benzoyl]amino]-3- azabicyclo[3.1.0] hexane-3- carbonyl]piperidin- 1-ium-1-yl]aceticMS [M]− 846.4Inter- mediate I12 and Inter- mediate R14acid formateExam- ple O3bis(4-aminobutyl)- (carboxymethyl)-[4- [4-[2-chloro-4-[[5- [2,3-difluoro-4-[1- (2-methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1- methyl- imidazole-2- carbonyl]amino] benzoyl]-3- methyl- piperazino]-4- keto-butyl] ammonium formateMS [M]−895.9Inter- mediate 114 and Inter- mediate R19Exam- ple O4cis 2-[1-(azetidin-3- ylmethyl)-4-[4-[2- chloro-4-[[5-[3- fluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol- 4-yl]-2-methyl- phenyl]-1-methyl- imidazole-2- carbonyl]amino] benzoyl] piperazine-1- carbonyl]piperidin- 1-ium-1-yl]acetic acid formateMS [M]−830.3Inter- mediate H9 and Inter- mediate R16Exam- ple O53-aminopropyl- (carboxymethyl)- [4-[4-[2-chloro-4- [[5-[3- fluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]-2-methyl- phenyl]-1-methyl- imidazole-2- carbonyl]amino] benzoyl] piperazino]- 4-keto-butyl]- methyl-ammonium. 1:12,2,2- trifluoroacetic acid; 2,2,2-MS [M]−806.7Inter- mediate H9 and Inter- mediate R18trifluoroacetateExam- ple O62-[1-(azetidin-3- ylmethyl)-4-[4-[2- chloro-4-[[5-[3- fluoro-4-[1-(2- methoxyethyl)-5- methyl-pyrazol-4- yl]phenyl]-1- methyl-imidazole- 2-carbonyl]amino] benzoyl]piperazine- 1-carbonyl] piperidin-1- ium-1-yl]acetic acid; 2,2,2- trifluoroacetateMS [M]−818.3Inter- mediate H12 and Inter- mediate R14Exam- ple O7cis 2-[1-(azetidin- 3-ylmethyl)-4-[4- [2-chloro-4-[[5- [3-fluoro-4-[1- (2-methoxyethyl)- 3-methyl-pyrazol- 4-yl]phenyl]-1- methyl- imidazole-2- carbonyl]amino] benzoyl] piperazine- 1-carbonyl] piperidin-1- ium-1-yl]acetic acid; 2,2,2- trifluoroacetateMS [M]+818.8Inter- mediate H13 and Inter- mediate R16Exam- ple O8cis 2-[1-(azetidin- 3-ylmethyl)-4-[4- [4-[[5- [2,3-difluoro-4- [1-(2- methoxyethyl)- 3-methylpyrazol- 4-yl]phenyl]-1- methylimidazole-2- carbonyl]amino]-2- methylbenzoyl] piperazine-1- carbonyl]piperidin- 1-ium-1-yl]acetic acid formateMS [M]+816.4Inter- mediate H14 and Inter- mediate R16Exam- ple O9cis 2-[1- (azetidin-3- ylmethyl)-4-[4-[4- [[5-[3-fluoro-4-[1- (2-methoxyethyl)- 3-methyl-pyrazol- 4-yl]phenyl]-1- methyl- imidazole-2- carbonyl]amino]-2- methyl- benzoyl] piperazine- 1-carbonyl] piperidin-1- ium-1-yl]acetic acid formateMS [M]+798.6Inter- mediate H15 and Inter- mediate R16Exam- ple O10cis 2-[1- (azetidin-3- ylmethyl)-4-[4-[4- [[5-[3-fluoro-4- [1-(2- methoxyethyl)-5- methylpyrazol- 4-yl]- 2-methylphenyl]-1- methylimidazole-2- carbonyl]amino]-2- methylbenzoyl] piperazine-1- carbonyl] piperidin- 1-ium-1-yl]acetic acid; formateMS [M]+812.6Inter- mediate H16 and Inter- mediate R16 Example O11 N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate Step 1: (2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carboxylic acid trans-4-hydroxy-L-proline (8 g, 61.01 mmol, 1 eq) is dispensed in methanol (50 mL). 1,3-diisopropyl-2-methyl-isourea (10.62 g, 67.11 mmol, 1.1 eq) was added and the suspension is stirred at 22° C. for 18 hr. Motherliquor is evaporated to dryness to afford crude (2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carboxylic acid (15.011 g, 99.8% yield, purity 65%) which was used crude for next step. MS [M+]: 160.1 Step 2: N-[3-chloro-4-[4-[(2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carbonyl]piperazine-1-carbonyl]phenyl]-5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate (2S,4R)-4-hydroxy-1,1-dimethyl-pyrrolidin-1-ium-2-carboxylic acid (13.9 mg, 0.039 mmol, 1.179 eq) was dissolved in N,N-dimethylformamide (1 mL), DIEA (12.58 mg, 17 uL, 0.097 mmol, 2.937 eq) and HATU (15 mg, 0.039 mmol, 1.190 eq) were added and the mixture was stirred for 2 min before N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[3-fluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]-2-methyl-phenyl]-1-methyl-imidazole-2-carboxamide 0.1:1 2,2,2-trifluoroacetic acid (24.7 mg, 0.033 mmol, 1.000 eq) was added. The reaction mixture was stirred at room temperature for 1.5 h. an excess of 4 M HCl in dioxane (1.03 g, 855.73 uL, 3.42 mmol, 42.82 eq) was added and the mixture was stirring for 5 h at room temperature. The crude reaction mixture was directly purified by preparative HPLC and lyophilized to afford the title compound (14.6 mg, 52% yield) a colourless lyoph powder. MS [M+]: 735.5. Example O12 2-[2-(3-aminopropyl)-4-[3-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotoyl]amino]-propyl]pyridin-1-ium-1-yl]acetic acid; formate Step 1: N-[3-[4-[3-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotoyl]amino]propyl]-2-pyridyl]propyl]carbamic acid tert-butyl ester To a solution of 1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotic acid (105.7 mg, 0.165 mmol, 1.000 eq) and N-[3-[4-(3-aminopropyl)-2-pyridyl]propyl]carbamic acid tert-butyl ester; hydrochloride (92 mg, 0.279 mmol, 1.691 eq) in N,N-dimethylformamide (1 mL) was added DIPEA (127.86 mg, 172.78 uL, 0.989 mmol, 6.000 eq) and HATU (81.5 mg, 0.214 mmol, 1.300 eq), the reaction mixture was stirred at room temperature overnight. The solvent was removed in vacuo and the residue purified via preparative HPLC to afford the title compound (95.7 mg, 63.33% yield) as off-white lyoph powder. MS [(M+H)+]: 916.9. Step 2: 2-[2-[3-(tert-butoxycarbonylamino)propyl]-4-[3-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotoyl]amino]propyl]pyridin-1-ium-1-yl]acetic acid tert-butyl ester To a solution of N-[3-[4-[3-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotoyl]amino]-propyl]-2-pyridyl]propyl]carbamic acid tert-butyl ester (95.7 mg, 0.104 mmol, 1.000 eq) in acetonitrile (2 mL) was added tert-butyl bromoacetate (162.94 mg, 123.44 uL, 0.835 mmol, 8.000 eq) and DIPEA (107.97 mg, 145.9 uL, 0.835 mmol, 8.000 eq), and the reaction mixture stirred at 50° C. overnight. The solvent was removed in vacuo to afford crude 2-[2-[3-(tert-butoxycarbonylamino)propyl]-4-[3-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotoyl]-amino]propyl]pyridin-1-ium-1-yl]acetic acid tert-butyl ester (243.8 mg, 99.58%) as yellow viscous oil, which was used without further purification. MS [M+]: 1030.9. Step 3: 2-[2-(3-aminopropyl)-4-[3-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotoyl]-amino]propyl]pyridin-1-ium-1-yl]acetic acid; formate To a solution of 2-[2-[3-(tert-butoxycarbonylamino)propyl]-4-[3-[[1-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]isonipecotoyl]amino]propyl]pyridin-1-ium-1-yl]acetic acid tert-butyl ester (243.8 mg, 0.104 mmol, 1.000 eq) in dichloromethane (2 mL) was added 4 M HCl in Dioxane (249.57 mg, 207.97 uL, 0.832 mmol, 8.000 eq) and the reaction mixture was stirred at room temperature overnight. The solvent was removed in vacuo and the residue purified via preparative HPLC to afford the title compound as light yellow lyoph powder. MS [M+]: 874.8. Example P1 azetidin-3-ylmethyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formate Step 1: tert-butyl N-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]carbamate To a stirred mixture of N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (633.0 mg, 1.0 mmol), DIEA (0.52 mL, 3.01 mmol) and BOC-gamma-abu-OH (0.2 g, 1.0 mmol) in DMF (10 mL) was added HATU (0.76 g, 2.01 mmol) at 15° C. The reaction mixture was stirred at 15° C. for 1 h. The mixture was poured into water, extracted with EtOAc, washed with brine, dried over Na2SO4and concentrated in vacuum to afford crude tert-butyl N-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]carbamate (500.0 mg, 0.64 mmol, 63.63% yield) as black oil. MS [(M+H)+]: 783.5. Step 2: N-[4-[4-(4-aminobutanoyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide A solution of tert-butyl N-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]carbamate (500.0 mg, 0.64 mmol) in hydrochloric acid (6.0 mL, 4 M in 1,4-dioxane) was stirred at 15° C. for 1 h. The mixture was concentrated in vacuum, the residue was purified by reversed phase HPLC and lyophilized to afford N-[4-[4-(4-aminobutanoyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (320.0 mg, 0.47 mmol, 73.38% yield) as white solid MS [(M+H)+]: 683.1. Step 3: tert-butyl 3-[[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]methyl]azetidine-1-carboxylate To a solution of N-[4-[4-(4-aminobutanoyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (110.0 mg, 0.16 mmol) in ACN (5 mL) was added 1-BOC-3-(bromomethyl)azetidine (40.28 mg, 0.16 mmol), sodium iodide (24.14 mg, 0.16 mmol) and potassium carbonate (33.38 mg, 0.24 mmol, 1.5 eq) at 20° C. Then the reaction mixture was heated to 50° C. and stirred for 16 h. The mixture was filtered and the filtrate was concentrated in vacuum. The residue was purified by Prep-HPLC, lyophilized to afford tert-butyl 3-[[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]methyl]azetidine-1-carboxylate (40.0 mg, 0.05 mmol, 29.14% yield) as white solid. MS [(M+H)+]: 852.2. Step 4: tert-butyl 3-[[(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]methyl]azetidine-1-carboxylate To a solution of tert-butyl 3-[[[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]methyl]azetidine-1-carboxylate (40.0 mg, 0.05 mmol) and TEA (0.02 mL, 0.14 mmol) in ACN (5 mL) was added tert-butyl bromoacetate (0.01 mL, 0.06 mmol) at 20° C., then the solution was stirred at 35° C. for 16 h. The reaction mixture was concentrated, purified by reversed phase-HPLC and concentrated to afford tert-butyl 3-[[(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]methyl]azetidine-1-carboxylate (40.0 mg, 0.04 mmol, 88.19% yield) as yellow oil. MS [(M+H)+]: 966.2. Step 5: (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; iodide To a solution of tert-butyl 3-[[(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]methyl]azetidine-1-carboxylate (40.0 mg, 0.04 mmol) and TEA (0.02 mL, 0.12 mmol) in ACN (2 mL) was added iodomethane (0.03 mL, 0.41 mmol) at 15° C., then the solution was stirred at 15° C. for 2 h. The reaction mixture was concentrated in vacuum, the residue was purified by reversed phase HPLC and lyophilized to afford (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; iodide (30.0 mg, 0.03 mmol, 98.47% yield) as white solid. MS [(M)+]: 980.2. Step 6: azetidin-3-ylmethyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formate A solution of (1-tert-butoxycarbonylazetidin-3-yl)methyl-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; iodide (30.0 mg, 0.03 mmol) in DCM (1 mL) was added TFA (1.0 mL, 12.98 mmol) at 15° C. and stirred at 15° C. for 16 h. The reaction mixture was concentrated in vacuum, the residue was purified by Prep-HPLC and lyophilized to afford azetidin-3-ylmethyl-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formate (13.7 mg, 0.02 mmol) as white solid. MS [(M)+]: 824.1. Example P2 bis(4-aminobutyl)-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate Step 1: bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate To a mixture of bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide (128.55 mg, 0.2 mmol) and DIEA (64.83 mg, 0.5 mmol) in DMF (2 mL) was added CMPI (51.26 mg, 0.2 mmol) at 30° C. and stirred at 30° C. for 0.5 h. Then, N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (100.0 mg, 0.17 mmol) was added to the above solution and stirred at 30° C. for another 3.5 h. The reaction mixture was purified by reversed phase-HPLC (water (0.1% FA)-ACN, B=85%˜90%) and lyophilized to afford bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate (80.0 mg, 0.07 mmol, 41.94% yield) as yellow solid. MS [(M)+]: 1139.5. Step 2: bis(4-aminobutyl)-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate To a mixture of bis[4-(tert-butoxycarbonylamino)butyl]-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate (60.0 mg, 0.05 mmol) in DCM (2 mL) was added TFA (2.0 mL, 25.96 mmol) at 30° C. and stirred for 16 h. The reaction mixture was concentrated in vacuum to give the residue, which was purified by prep-HPLC (Phenomenex Synergi C18 150×25 mm×10 um, water (0.225% FA)-ACN, B=9%-39% FlowRat: 25 ml/min) to afford bis(4-aminobutyl)-(carboxymethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate (30.0 mg, 0.03 mmol) as white solid. MS [(M)+]: 883.5. Example P3 bis(3-aminopropyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formate Step 1: tert-butyl N-[3-[3-(tert-butoxycarbonylamino)propyl-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]propyl]carbamate To a stirred mixture of N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (100.0 mg, 0.17 mmol), DIEA (0.09 mL, 0.5 mmol) and 4-[bis[3-(tert-butoxycarbonylamino)propyl]amino]butanoic acid (69.82 mg, 0.17 mmol) in DMF (3 mL) was added HATU (0.13 g, 0.33 mmol) at 15° C. Then the solution was stirred at 15° C. for 12 h. The mixture was poured into water (40 ml), extracted with EtOAc (40 ml), washed with brine (30 ml), dried over Na2SO4and concentrated in vacuum to afford a residue. The residue was purified by reversed phase HPLC and lyophilized to afford tert-butyl N-[3-[3-(tert-butoxycarbonylamino)propyl-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]propyl]carbamate (60.0 mg, 0.06 mmol, 35.97% yield) as white solid. MS [(M+H)+]: 997.2. Step 2: bis[3-(tert-butoxycarbonylamino)propyl]-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; iodide To a solution of tert-butyl N-[3-[3-(tert-butoxycarbonylamino)propyl-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]amino]propyl]carbamate (60.0 mg, 0.06 mmol) and TEA (0.03 mL, 0.18 mmol) in ACN (3 mL) was added iodomethane (0.04 mL, 0.6 mmol) at 15° C. and stirred for 16 h. The reaction mixture was concentrated in vacuum, purified by reversed phase and lyophilized to afford bis[3-(tert-butoxycarbonylamino)propyl]-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; iodide (25.0 mg, 0.02 mmol, 36.48% yield) as white solid. MS [(M)+]: 1011.3. Step 3: bis(3-aminopropyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formate To a solution of bis[3-(tert-butoxycarbonylamino)propyl]-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; iodide (50.0 mg, 0.04 mmol) in DCM (3 mL) was added TFA (3.0 mL, 38.94 mmol), the solution was stirred at 25° C. for 2 h. The reaction mixture was concentrated in vacuum and purified by Prep-HPLC, lyophilized to afford bis(3-aminopropyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-methyl-ammonium; formate (15.7 mg, 0.02 mmol) as white solid. MS [(M)+]: 811.5. Example P4 carboxymethyl-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-bis[2-(dimethylamino)ethyl]ammonium; 2,2,2-trifluoroacetate Step 1: bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate To a solution of N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (200.0 mg, 0.33 mmol), DIEA (0.17 mL, 1.0 mmol) and bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)-(3-carboxypropyl)ammonium; bromide (195.49 mg, 0.33 mmol) in DMF (5 mL) was added HATU (0.25 g, 0.67 mmol) at 15° C. Then the solution was stirred at 15° C. for 16 h. The mixture was purified by reversed phase-HPLC and lyophilized to afford bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate (300.0 mg, 0.27 mmol, 82.7% yield) as light brown solid. MS [(M)+]: 1083.5. Step 2: (2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-bis[2-(dimethylamino)ethyl]-ammonium; formate To a solution of bis[2-(tert-butoxycarbonylamino)ethyl]-(2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]ammonium; formate (245.0 mg, 0.22 mmol) and formaldehyde (1.0 mL, 37% in water) in DCM (4 mL) was added TFA (1.0 mL, 12.98 mmol) and stirred at 20° C. for 16 h. NaBH(OAc)3(229.82 mg, 1.08 mmol) was added to the solution and stirred at 20° C. for 1 h. The reaction mixture was concentrated in vacuum and the residue was purified by reversed phase-HPLC and lyophilized to obtain (2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-bis[2-(dimethylamino)ethyl]ammonium; formate (60.0 mg, 0.06 mmol, 28.07% yield) as light brown solid. MS [(M)+]: 939.6. Step 3: carboxymethyl-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-bis[2-(dimethylamino)ethyl]ammonium; 2,2,2-trifluoroacetate To a solution of (2-tert-butoxy-2-oxo-ethyl)-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-bis[2-(dimethylamino)ethyl]ammonium; formate (60.0 mg, 0.06 mmol) in DCM (1 mL) was added TFA (1.0 mL, 12.98 mmol) and stirred at 20° C. for 16 h. The reaction mixture was concentrated in vacuum and the residue was purified by prep-HPLC to obtain carboxymethyl-[4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazin-1-yl]-4-oxo-butyl]-bis[2-(dimethylamino)ethyl]ammonium; 2,2,2-trifluoroacetate (11.86 mg, 0.01 mmol) as white solid. MS [(M)+]: 883.6. Example P5 2-[1-(3-aminopropyl)-4-[5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-pentanoyl]piperazin-1-ium-1-yl]acetic acid; formate Step 1: 5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-valeric acid methyl ester 5-keto-5-methoxy-valeric acid (35.93 mg, 0.246 mmol, 1.300 eq), HATU (107.87 mg, 0.284 mmol, 1.500 eq) and DIEA (122.21 mg, 165.15 uL, 0.946 mmol, 5.000 eq) are dissolved in N,N-dimethylformamide (1.2 mL). The mixture is stirred for 10 min. N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride (120 mg, 0.189 mmol, 1.000 eq) is then added and the mixture was stirred at room temperature over night. The mixture was poured into 50 mL water and extracted 2× with EtOAc. The organic phases were combined and evaporated to dryness to give 5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-valeric acid methyl ester 0.1:1 hydrogen chloride (299.5 mg, 99.67%) as light brown solid and used crude for next step. [(M+H)+]: 726.3. Step 2: 5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-valeric acid 5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-valeric acid methyl ester (299.5 mg, 0.189 mmol, 1.000 eq) was dissolved in methanol (5 mL) and water (2 mL). lithium hydroxide monohydrate (79.1 mg, 1.89 mmol, 10.000 eq) was added and the reaction is stirred at 22° C. for 18 hr. The clear solution was acidified with 4M aq. HCl solution. The product is extracted two times with ethylacetate. Both organic layers were combined, dried with sodium sulfate, filtered and evaporated to dryness. The crude product purified via prep HPLC to afford after lyophilization 5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-valeric acid (50.6 mg, 37.32%) as white solid. [(M+H)+]: 712.2. Step 3: 2-[1-(3-aminopropyl)-4-[5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-pentanoyl]piperazin-1-ium-1-yl]acetic acid; formate To a solution of 5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazino]-5-keto-valeric acid (12.5 mg, 0.017 mmol, 1.000 eq) and 2-[1-[3-(tert-butoxycarbonylamino)propyl]piperazin-1-ium-1-yl]acetic acid tert-butyl ester; formate (10.37 mg, 0.024 mmol, 1.477 eq) in dichloromethane (1.04 mL) was added DIEA (10.68 mg, 14.44 uL, 0.083 mmol, 5.000 eq) and then PyAOP reagent (11.2 mg, 0.021 mmol, 1.300 eq). The reaction mixture was stirred at room temperature for 2 hr. Boc and t-butyl were then deprotected with 4N HCl in dioxane (247.96 mg, 206.64 uL, 0.827 mmol, 50.000 eq) over night at room temperature. Volatiles were removed in vacuo and the residue was purified via prep HPLC to yield the desired compound 2-[1-(3-aminopropyl)-4-[5-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-5-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl] amino]benzoyl]piperazino]-5-keto-pentanoyl]piperazin-1-ium-1-yl] acetic acid; formate (5.8 mg, 37.27%) as white powder. MS [M-]: 940.5. The following example was prepared in analogy to Example P5. MSStartingEx#NameStructure[M]MaterialExample P62-[1-(azetidin-3- ylmethyl)-4-[5-[4-[2- chloro-4-[[5-[2,3- difluoro-4-[1-(2- methoxyethyl)-5-methyl- pyrazol-4-yl]phenyl]-1- methyl-imidazole-2- carbonyl]amino]benzoyl] piperazino]-5-keto- pentanoyl]piperazin-1- ium-1-yl]acetic acid; 1:1 formate852.5Intermediate R23 Example Q1 N-[3-chloro-4-[4-(4-oxo-3-aza-6-azoniaspiro[5.5]undecane-9-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate Step 1 tert-butyl 2-[1-[2-(tert-butoxycarbonylamino)ethyl]-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetate; 2,2,2-trifluoroacetate To a solution of N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride (200.0 mg, 0.32 mmol), DIEA (0.16 mL, 0.95 mmol) and 1-[2-(tert-butoxycarbonylamino)ethyl]-1-(2-tert-butoxy-2-oxo-ethyl)piperidin-1-ium-4-carboxylic acid; 2,2,2-trifluoroacetate (205.09 mg, 0.41 mmol) in DMF (4 mL) was added HATU (179.78 mg, 0.47 mmol, 1.5 eq) at 0° C. and stirred at 0° C. for 1 h. The solution was purified by reversed phase-HPLC and lyophilized to afford tert-butyl 2-[1-[2-(tert-butoxycarbonylamino)ethyl]-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetate; 2,2,2-trifluoroacetate (251.0 mg, 0.23 mmol, 73.7% yield) as white solid. MS [(M)+]: 966.6. Step 2: N-[3-chloro-4-[4-(4-oxo-3-aza-6-azoniaspiro[5.5]undecane-9-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate To a solution of tert-butyl 2-[1-[2-(tert-butoxycarbonylamino)ethyl]-4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidin-1-ium-1-yl]acetate; 2,2,2-trifluoroacetate (51.0 mg, 0.05 mmol) in DCM (2 mL) was added TFA (4.0 mL, 51.92 mmol) in one portion at 10° C. and stirred at 10° C. for 16 h. The solution was concentrated, purified by Prep-HPLC and lyophilized to afford N-[3-chloro-4-[4-(4-oxo-3-aza-6-azoniaspiro[5.5]undeACNe-9-carbonyl)piperazine-1-carbonyl]phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; 2,2,2-trifluoroacetate (17.4 mg, 0.02 mmol) as white solid. MS [(M)+]: 792.3. Example Q2 N-[4-[4-(3-benzyl-3-aza-6-azoniaspiro[5.5]undecane-9-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide Step 1: 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester To a solution of 1-tert-butoxycarbonylisonipecotic acid (20.93 mg, 91.3 umol, 1.200 eq), N-[3-chloro-4-(piperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (50 mg, 76.08 umol, 1.000 eq), HATU (43.39 mg, 114.12 umol, 1.500 eq) and N,N-dimethylformamide, extra dry (400 uL) was added DIEA (29.5 mg, 39.86 uL, 228.25 umol, 3.000 eq) and the reaction mixture was stirred at room temperature for 2h. The reaction mixture was diluted in EtOAc, and then washed with water and brine, filtered over MgSO4 and then concentrated under vacuo. The crude material was purified by flash chromatography to yield 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester (41.8 mg, 67.89%) as yellow waxy solid. MS [(M+H)+]: 809.4 Step 2: N-[3-chloro-4-(4-isonipecotoylpiperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide; hydrochloride 4-[4-[2-chloro-4-[[5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carbonyl]amino]benzoyl]piperazine-1-carbonyl]piperidine-1-carboxylic acid tert-butyl ester (41.8 mg, 0.052 mmol, 1.000 eq) was treated with an excess of 4 M HCl in dioxane (129.12 uL, 516.5 umol, 10.000 eq) in 1,4-dioxane (1.0 mL) and was stirred at room temperature over 4 days. Diethyl ether was added so the product was crash out. The solvent was evaporated to yield N-[3-chloro-4-(4-isonipecotoylpiperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide hydrochloride (40.8 mg, 100%) as white solid. MS [(M+H)+]: 709.3. Step 3: N-[4-[4-(3-benzyl-3-aza-6-azoniaspiro[5.5]undecane-9-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide Benzyl-bis(2-bromoethyl)amine (9.47 mg, 6.06 uL, 29.5 umol, 1.100 eq), N-[3-chloro-4-(4-isonipecotoylpiperazine-1-carbonyl)phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (20 mg, 26.82 umol, 1.000 eq), potassium carbonate (12.23 mg, 88.51 umol, 3.300 eq) and potassium iodide (890.51 ug, 5.36 umol, 0.200 eq) were combined in N,N-dimethylformamide (500 uL). The reaction mixture was then stirred at room temperature over night. The mixture was purified over prep HPLC to afford N-[4-[4-(3-benzyl-3-aza-6-azoniaspiro[5.5]undecane-9-carbonyl)piperazine-1-carbonyl]-3-chloro-phenyl]-5-[2,3-difluoro-4-[1-(2-methoxyethyl)-3-methyl-pyrazol-4-yl]phenyl]-1-methyl-imidazole-2-carboxamide (5.1 mg, 20.56%) as colorless solid. MS [(M)+]: 868.5. The following examples were prepared in analogy to Example O1. MSStartingEx#NameStructure[M]+MaterialExample Q3trans 2-[1-(azetidin-3- ylmethyl)-4-[4-[2-chloro- 4-[[5-[2,3-difluoro-4-[1- (2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperazine-1- carbonyl]piperidin-1- ium-1-yl]acetic acid; formate836.6Intermediate I2 and Intermediate R16’Example Q4cis 2-[1-(azetidin-3- ylmethyl)-4-[4-[2-chloro- 4-[[5-[2,3-difluoro-4-[1- (2-methoxyethyl)-3- methyl-pyrazol-4- yl]phenyl]-1-methyl- imidazole-2- carbonyl]amino]benzoyl] piperazine-1- carbonyl]piperidin-1- ium-1-yl]acetic acid formate;836.6Intermediate I2 and Intermediate R16 Assay Procedures Antimicrobial Susceptibility Testing: 90% Growth Inhibitory Concentration (IC90) Determination The in vitro antimicrobial activity of the compounds was determined according to the following procedure: The assay used a 10-points Iso-Sensitest broth medium to measure quantitatively the in vitro activity of the compounds againstAcinetobacter baumanniiATCC17961. Stock compounds in DMSO were serially twofold diluted (e.g. range from 50 to 0.097 μM final concentration) in 384 wells microtiter plates and inoculated with 49 μl the bacterial suspension in Iso-Sensitest medium to have a final cell concentration of ˜5×10(5)CFU/ml in a final volume/well of 50 ul/well. Microtiter plates were incubated at 35±2° C. Bacterial cell growth was determined with the measurement of optical density at λ=600 nm each 20 minutes over a time course of 16h. Growth inhibition was calculated during the logarithmic growth of the bacterial cells with determination of the concentration inhibiting 50% (IC50) and 90% (IC90) of the growth. Table 1 provides the 90% growth inhibitory concentrations (IC90) in micromoles per liter of the compounds of present invention obtained against the strainAcinetobacter baumanniiATCC17961. Particular compounds of the present invention exhibit an IC90 (Acinetobacter baumanniiATCC17961)≤25 μmol/l. More particular compounds of the present invention exhibit an IC90 (Acinetobacter baumanniiATCC17961)≤5 μmol/l. Most particular compounds of the present invention exhibit an IC90 (Acinetobacter baumanniiATCC17961)≤1 μmol/l. TABLE 1ATCC17961IC90Example[μM]Example A10.12Example A20.23Example A30.23Example A40.11Example A50.055Example A60.058Example A70.055Example A80.053Example A90.16Example A100.098Example A110.36Example A120.21Example A130.11Example B10.19Example B20.12Example B30.13Example B40.23Example B50.18Example B60.12Example B70.17Example B80.28Example B90.51Example B100.2Example B110.17Example B120.11Example B130.81Example B140.68Example B150.17Example B160.3Example B170.26Example B180.43Example B190.057Example B200.22Example B210.1Example B220.33Example B230.098Example B240.095Example B250.27Example B260.064Example B270.24Example B280.23Example B290.34Example B300.19Example B310.071Example B320.058Example B330.82Example B34/Example B350.13Example B360.11Example B370.2Example B380.3Example B390.25Example B401.8Example C10.045Example C20.041Example C30.1Example C40.16Example C50.24Example C60.14Example C70.14Example C80.21Example C90.26Example C100.28Example C110.32Example C120.153Example C13/Example C140.082Example D10.28Example D20.21Example D30.39Example D40.178Example D60.085Example D70.15Example D80.3Example D90.3Example D100.21Example D110.27Example D120.2Example D130.34Example D140.11Example D150.1Example D160.15Example D170.26Example D180.058Example D19/Example D200.064Example D210.2Example D220.199Example D230.68Example D240.073Example D250.051Example D260.19Example D270.23Example D280.03Example D29<0.02Example D300.24Example D310.22Example D320.052Example D330.21Example D340.11Example D350.17Example D360.088Example D370.15Example D380.15Example D400.077Example D410.061Example D420.06Example D430.81Example D440.16Example E1/Example E20.048Example E30.49Example E4/Example E50.18Example E60.95Example E70.21Example E80.2Example E90.065Example E100.17Example E110.061Example E120.15Example E130.38Example E140.27Example E150.18Example E160.23Example E170.71Example E180.82Example E190.13Example E200.53Example E210.29Example E220.059Example E230.14Example F10.33Example F20.065Example F30.12Example F40.83Example F50.28Example F60.57Example F70.52Example F8/Example F90.16Example F100.075Example F110.8Example F120.17Example F140.1Example F150.055Example F160.24Example F170.27Example F181.1Example F190.33Example F200.18Example F21/Example F220.17Example F230.75Example F240.59Example F251.2Example F261.4Example F270.4Example F280.27Example F290.28Example F300.097Example F310.067Example F320.58Example F330.36Example F340.23Example F35/Example F360.58Example F370.69Example F380.76Example F390.34Example F400.61Example F410.15Example F420.21Example F430.57Example F440.73Example F450.31Example F461.1Example F470.53Example F480.47Example F491.52Example F501.1Example F510.21Example F520.27Example F530.11Example F540.59Example F550.27Example F560.33Example F570.88Example F580.48Example F590.75Example F600.15Example K20.36Example K30.3Example F630.56Example F640.25Example F650.37Example F660.029Example F671.1Example G10.29Example G20.32Example G30.1Example G40.19Example G5/Example G60.14Example G70.55Example G80.084Example G90.21Example G100.12Example G110.71Example G120.45Example G130.13Example G140.089Example G150.3Example G161.1Example G170.72Example G180.51Example G19/Example G20/Example G21/Example G220.67Example G230.27Example G240.508Example G251Example G260.1Example G270.12Example G280.088Example G290.33Example G301Example G310.14Example G320.185Example G330.44Example G340.068Example H10.15Example H20.19Example H30.18Example H40.16Example H50.17Example H60.17Example H70.19Example H80.54Example I10.432Example I20.608Example I30.398Example I41.5Example I5/Example I60.82Example I70.76Example J10.022Example K10.6Example L10.206Example L20.168Example L30.264Example L40.106Example L50.374Example L60.197Example M10.152Example N10.189Example N20.183Example N30.352Example N40.108Example O10.354Example O20.362Example O30.113Example O40.118Example O50.200Example O60.134Example O70.140Example O80.245Example O90.284Example O100.231Example O110.156Example O120.339Example P10.191Example P20.066Example P30.293Example P40.657Example P50.295Example P60.310Example Q10.165Example Q20.705Example Q30.359Example Q40.195 Minimal Inhibitory Concentration Protocol (MIC) Assay: Table 2 provides the in vitro potency of the compounds of present invention obtained against the strainAcinetobacter baumanniiATCC17978, which was assessed by an MIC (Minimal Inhibitory Concentration) assay as follows. Test compounds were prepared from 10 mM DMSO stock solutions. The top dose was diluted from 10 mM to 2.5 mM by DMSO, followed by serial 2-fold 11 points dilutions in DMSO in a master plate (Greiner, Cat No: 651201). 2 μL compounds were transferred from the master plate into a new 96-well assay plate (Costar, 3599). The growth medium Caution-Adjusted Mueller Hinton Broth (CAMHIB) was prepared by adding 22 g powder (BD, 212322) in 1 L purified water, autoclaved, and supplemented with sterilized CaCl2) (20 mg per liter) and MgCl2(10 mg per liter). Vials of each of the test microorganisms were maintained frozen in the vapor phase of a liquid nitrogen freezer. Took out the bacterial strain ATCC 17978 from liquid nitrogen freezer, thawed it at room temperature, and diluted the bacterial in the CAMHB medium to achieve a final inoculum of 2×105CFU/mL. 98 μL of the adjusted bacteria suspension was dispensed to the assay plate and pipetted 5 times. Then the assay plates were incubated for 20 hours at 35±2° C. in ambient air with humidity. Following incubation, MIC (g/mL) value, the lowest concentration of drug that inhibits visible growth of the microorganism, was recorded by visual judgment of bacterial growth through magnification mirror of MIC reader, and the assay plates were photographed with Qcount system as image raw data. Meanwhile, the D600 of assay plates was recorded with SpectraMax Plus384 as GD raw data. TABLE 2MIC AB.ATCC17978Example[μM]Example B340.65Example C121.20Example C131.22Example D041.56Example D181.23Example D191.07Example D221.10Example D290.45Example E010.56Example E040.57Example F81.41Example F212.31Example F294.73Example F301.19Example F351.24Example F492.50Example F530.60Example G50.32Example G190.32Example G202.52Example G212.57Example G243.85Example G320.66Example I12.32Example I22.41Example I31.81Example I50.58 Example 1 A compound of formula (I) can be used in a manner known per se as the active ingredient for the production of tablets of the following composition: Per tablet Active ingredient200 mgMicrocrystalline cellulose155 mgCorn starch25 mgTalc25 mgHydroxypropylmethylcellulose20 mg425 mg Example 2 A compound of formula (I) can be used in a manner known per se as the active ingredient for the production of capsules of the following composition: Per capsule Active ingredient100.0 mgCorn starch20.0 mgLactose95.0 mgTalc4.5 mgMagnesium stearate0.5 mg220.0 mg Example 3 A compound of formula (I) can be used in a manner known per se as the active ingredient for the production of an infusion solution of the following composition: Active ingredient100 mgLactic acid 90%100 mgNaOH q.s. or HCl q.s. for adjustment to pH 4.0Sodium chloride q.s. or glucose q.s. for adjustment of the osmolality to 290 mOsm/kgWater for injection (WFI) ad 100 ml Example 4 A compound of formula (I) can be used in a manner known per se as the active ingredient for the production of an infusion solution of the following composition: Active ingredient100 mgHy droxypropyl-beta-cyclodextrin10 gNaOH q.s. or HCl q.s. for adjustment to pH 7.4Sodium chloride q.s. or glucose q.s. for adjustment of the osmolality to 290 mOsm/kgWater for injection (WFI) ad 100 ml
539,468
11858913
DETAILED DESCRIPTION Definitions Various embodiments are described hereinafter. It should be noted that the specific embodiments are not intended as an exhaustive description or as a limitation to the broader aspects discussed herein. One aspect described in conjunction with a particular embodiment is not necessarily limited to that embodiment and can be practiced with any other embodiment(s). As used herein, “about” will be understood by persons of ordinary skill in the art and will vary to some extent depending upon the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art, given the context in which it is used, “about” will mean up to plus or minus 10% of the particular term. The use of the terms “a” and “an” and “the” and similar referents in the context of describing the elements (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the embodiments and does not pose a limitation on the scope of the claims unless otherwise stated. No language in the specification should be construed as indicating any non-claimed element as essential. In the definition of chemical substituents, each of Rxand Ryis independently hydrogen, alkyl, carbocyclic ring, heterocyclic ring, aryl, or heteroaryl, all of which, except hydrogen, are optionally substituted. Unless otherwise indicated, the abbreviations “TR” and “THR” refer to thyroid hormone receptors. As used herein, “pharmaceutically acceptable salt” refers to a salt of a compound that does not cause significant irritation to a patient to which it is administered and does not abrogate the biological activity and properties of the compound. Pharmaceutical salts can be obtained by reaction of a compound disclosed herein with an acid or base. Base-formed salts include, without limitation, ammonium salt (NH4+); alkali metal, such as, without limitation, sodium or potassium, salts; alkaline earth, such as, without limitation, calcium or magnesium, salts; salts of organic bases such as, without limitation, dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine; and salts with the amino group of amino acids such as, without limitation, arginine and lysine. Useful acid-based salts include, without limitation, hydrochlorides, hydrobromides, sulfates, nitrates, phosphates, methane-sulfonates, ethanesulfonates, p-toluenesulfonates and salicylates. As used herein, “pharmaceutically acceptable ester” refers to an ester of a compound that does not cause significant irritation to a patient to which it is administered. The ester is metabolized in the body to result in the parent compound, e.g., the claimed compound. Accordingly, the ester does not abrogate the biological activity and properties of the compound. Pharmaceutical esters can be obtained by reaction of a compound disclosed herein with an alcohol. Methyl, ethyl, and isopropyl esters are some of the common esters to be prepared. Other esters suitable are well-known to those skilled in the art (see, for example Wuts, P. G. M., Greene's Protective Groups in Organic Synthesis, 5thEd., John Wiley & Sons, New York, N.Y., 2014, which is incorporated herein by reference in its entirety). Where the compounds disclosed herein have at least one chiral center, they may exist as a racemate or as individual enantiomers. It should be noted that all such isomers and mixtures thereof are included in the scope of the present disclosure. Thus, the illustration of a chiral center without a designation of R or S signifies that the scope of the disclosure includes the R isomer, the S isomer, the racemic mixture of the isomers, or mixtures where one isomer is present in greater abundance than the other. Where the processes for the preparation of the compounds disclosed herein give rise to mixtures of stereoisomers, such isomers may be separated by conventional techniques such as preparative chiral chromatography. The compounds may be prepared in racemic form or individual enantiomers may be prepared by stereoselective synthesis or by resolution. The compounds may be resolved into their component enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (−)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-1-tartaric acid, followed by fractional crystallization and regeneration of the free base. The compounds may also be resolved by formation of diastereomeric esters or amides followed by chromatographic separation and removal of the chiral auxiliary. Unless otherwise indicated, when a substituent is deemed to be “optionally substituted” it is meant that the substituent is a group that may be substituted with one or more (e.g., 1 or 2, or 1 to 3, or 1 to 4 or 1 to 5, or 1 to 6) group(s) individually and independently selected, without limitation, from alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, O-carbamoyl, N-carbamoyl, O-thiocarbamoyl, N-thiocarbamoyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, is O-cyanato, thiocyanato, isothiocyanato, nitro, silyl, trihalomethanesulfonyl, and amino (e.g., —NRxRy), including mono- and di-substituted amino groups, and the protected derivatives thereof. The protecting groups that may form the protective derivatives of the above substituents are known to those of skill in the art and may be found in references such as Wuts, above. As used herein, a “carbocyclic ring” is an aromatic or non-aromatic ring structure in which all the atoms in the ring are carbon atoms. As such, the ring structure may be fully saturated, fully unsaturated, or partially saturated. If any of the atoms in the ring is anything other than a carbon atom, then the ring is a “heterocyclic ring.” Examples of atoms that are within a ring include sulfur, oxygen, and nitrogen. A carbocyclic ring or a heterocyclic ring may be polycyclic, e.g., a fused ring system, a spirocyclic ring system, or a bridged ring system. These polycyclic rings include, for example, adamantyl, norbornyl (i.e., bicyclo[2.2.1]heptanyl), norbornenyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Additional non-limiting examples include bicyclic rings such as but not limited to: As used herein, “aryl” refers to a carbocyclic (all carbon) ring that has a fully delocalized pi-electron system. The “aryl” group can be made up of two or more fused rings (rings that share two adjacent carbon atoms). When the aryl is a fused ring system, then the ring that is connected to the rest of the molecule has a fully delocalized pi-electron system. The other ring(s) in the fused ring system may or may not have a fully delocalized pi-electron system. Further, the other ring(s) may or may not contain one or more heteroatoms (e.g., O, N, or S). Examples of aryl groups include, without limitation, the radicals of benzene, naphthalene and azulene. Additional non-limiting examples include: As used herein, “heteroaryl” refers to a ring that has a fully delocalized pi-electron system and contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur in the ring. The “heteroaryl” group can be made up of two or more fused rings (rings that share two adjacent carbon atoms). When the heteroaryl is a fused ring system, then the ring that is connected to the rest of the molecule has a fully delocalized pi-electron system. The other ring(s) in the fused ring system may or may not have a fully delocalized pi-electron system. Examples of heteroaryl rings include, without limitation, furan, thiophene, phthalazinone, pyrrole, oxazole, thiazole, imidazole, pyrazole, isoxazole, isothiazole, triazole, thiadiazole, pyridine, pyridazine, pyrimidine, pyrazine and triazine. Wherever “hetero” is used it is intended to mean a group as specified, such as an alkyl or an aryl group, where at least one carbon atom has been replaced with a heteroatom selected from nitrogen, oxygen and sulfur. As used herein, “alkyl” refers to a straight or branched chain fully saturated (no double or triple bonds) hydrocarbon group. An alkyl group of the presently disclosed compounds may comprise from 1 to 20 carbon atoms. An alkyl group herein may also be of medium size having 1 to 10 carbon atoms. An alkyl group herein may also be a lower alkyl having 1 to 5 carbon atoms or 1 to 6 carbon atoms. Examples of alkyl groups include, without limitation, methyl, ethyl, n-propyl, isopropyl, n-butyl, i-butyl, sec-butyl, t-butyl, amyl, t-amyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl and dodecyl. An alkyl group of the presently disclosed compounds may be substituted or unsubstituted. When substituted, the substituent group(s) can be one or more group(s) independently selected from cycloalkyl, aryl, heteroaryl, heterocycloalkyl, hydroxy, protected hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamoyl, N-carbamoyl, O-thiocarbamoyl, N-thiocarbamoyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, trihalomethanesulfonyl, amino (e.g., —NRxRy) and protected amino. As used herein, “alkenyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. An alkenyl group of the presently disclosed compounds may be unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above regarding alkyl group substitution, or with regard to optional substitution. As used herein, “alkynyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. An alkynyl group of the presently disclosed compounds may be unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above regarding alkyl group substitution, or with regard to optional substitution. As used herein, “acyl” refers to an “RxC(═O)—” group. As used herein, “cycloalkyl” refers to a completely saturated (no double bonds) hydrocarbon ring. In some embodiments, cycloalkyl refers to a hydrocarbon ring containing no double bonds or one or more double bonds provided that they do not form a fully delocalized pi-electron system in the ring. Cycloalkyl groups of the presently disclosed compounds may range from C3to C8. A cycloalkyl group may be unsubstituted or substituted. If substituted, the substituent(s) may be selected from those indicated above regarding substitution of an alkyl group. The “cycloalkyl” group can be made up of two or more fused rings (rings that share two adjacent carbon atoms). When the cycloalkyl is a fused ring system, then the ring that is connected to the rest of the molecule is a cycloalkyl as defined above. The other ring(s) in the fused ring system may be a cycloalkyl, a cycloalkenyl, an aryl, a heteroaryl, or a heterocycloalkyl. As used herein, “cycloalkenyl” refers to a cycloalkyl group that contains one or more double bonds in the ring although, if there is more than one, they cannot form a fully delocalized pi-electron system in the ring (otherwise the group would be “aryl,” as defined herein). A cycloalkenyl group of the presently disclosed compounds may unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above regarding alkyl group substitution. The “cycloalkenyl” group can be made up of two or more fused rings (rings that share two adjacent carbon atoms). When the cycloalkenyl is a fused ring system, then the ring that is connected to the rest of the molecule is a cycloalkenyl as defined above. The other ring(s) in the fused ring system may be a cycloalkyl, a cycloalkenyl, an aryl, a heteroaryl, or a heterocycloalkyl. The term “alkylene” refers to an alkyl group, as defined herein, which is a biradical and is connected to two other moieties. Thus, methylene (—CH2—), ethylene (—CH2CH2—), propylene (—CH2CH2CH2—), isopropylene (IUPAC: (methyl)ethylene) (—CH2—CH(CH3)—), and isobutylene (IUPAC: 2-(methyl)propylene) (—CH2—CH(CH3)—CH2—) are examples, without limitation, of an alkylene group. Similarly, the term “cycloalkylene” refers to a cycloalkyl group, as defined here, which binds in an analogous way to two other moieties. If the alkyl and cycloalkyl groups contain unsaturated carbons, the terms “alkenylene” and “cycloalkenylene” are used. As used herein, “heterocycloalkyl,” “heteroalicyclic,” or “heteroali-cyclyl” refers to a ring having in the ring system one or more heteroatoms independently selected from nitrogen, oxygen and sulfur. The ring may also contain one or more double bonds provided that they do not form a fully delocalized pi-electron system in the rings. The ring defined herein can be a stable 3- to 18-membered ring that consists of carbon atoms and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur. Heterocycloalkyl, groups of the presently disclosed compounds may be unsubstituted or substituted. When substituted, the substituent(s) may be one or more groups independently selected from the group consisting of halogen, hydroxy, protected hydroxy, cyano, nitro, alkyl, alkoxy, acyl, acyloxy, carboxy, protected carboxy, amino, protected amino, carboxamide, protected carboxamide, alkylsulfonamido and trifluoromethane-sulfonamido. The “heterocycloalkyl” group can be made up of two or more fused rings (rings that share two adjacent carbon atoms). When the heterocycloalkyl is a fused ring system, then the ring that is connected to the rest of the molecule is a heterocycloalkyl as defined above. The other ring(s) in the fused ring system may be a cycloalkyl, a cycloalkenyl, an aryl, a heteroaryl, or a heterocycloalkyl. As used herein, “aralkyl” refers to an alkylene substituted with an aryl group. As used herein, “carbocyclic alkyl” or “(carbocyclic)alkyl” refers to an alkylene substituted with a carbocyclic group. As used herein, “heterocyclicalkyl” or (heterocyclic)alkyl” refers to an alkylene substituted with a heterocyclic group. Similarly, “(heterocycloalkyl)alkyl” refers to an alkylene substituted with a heterocycloalkyl group. As used herein, “heteroarylalkyl” or “(heteroaryl)alkyl” refers to an alkylene substituted with a heteroaryl group. An “O-carboxy” group refers to a “RxC(═O)O—” group. A “C-carboxy” group refers to a “—C(═O)ORx” group. An “acetyl” group refers to a CH3C(═O)— group. A “C-amido” group refers to a “—C(═O)NRxRy” group. An “N-amido” group refers to a “RyC(═O)NRx—” group. The term “perhaloalkyl” refers to an alkyl group in which all the hydrogen atoms are replaced by halogen atoms. Any unsubstituted or monosubstituted amine group on a compound herein can be converted to an amide, any hydroxy group can be converted to an ester and any carboxyl group can be converted to either an amide or ester using techniques well-known to those skilled in the art (see, for example Wuts, above). It is understood that, in any compound of the presently disclosed compounds having one or more chiral centers, if an absolute stereochemistry is not expressly indicated, then each center may independently be R or S or a mixture thereof. In addition, it is understood that, in any compound of the presently disclosed compounds having one or more double bond(s) generating geometrical isomers that can be defined as E or Z each double bond may independently be E or Z, or a mixture thereof. It is understood that the disclosure of a compound herein inherently includes the disclosure of a tautomer thereof, if applicable. For instance, the disclosure of: also includes the disclosure of: and vice versa, even if only one of the two structures is disclosed. Throughout the present disclosure, when a compound is illustrated or named, it is understood that the isotopically enriched analogs of the compound are also contemplated. For example, a compound may have a deuterium incorporated instead of a hydrogen, or a carbon-13 instead of carbon with natural isotopic distribution. The isotopic enrichment may be in one location on the compound, i.e., only one hydrogen is replaced by a deuterium, or in more than one location. The present disclosure also encompasses compounds where all the similar atoms are replaced by their less common isotope, for example, a perdeutero compound where all the hydrogen atoms are replaced by a deuterium. The isotopically enriched compounds are useful when obtaining NMR spectra or when making use of an isotope effect in managing the kinetics of the reaction the compound undergoing. The term “pharmaceutical composition” refers to a mixture of one or more compounds disclosed herein with other chemical components, such as diluents or carriers. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, injection, aerosol, parenteral, and topical administration. Pharmaceutical compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. The term “carrier” defines a chemical compound that facilitates the incorporation of a compound into cells or tissues. For example, dimethyl sulfoxide (DMSO) is a commonly utilized carrier as it facilitates the uptake of many organic compounds into the cells or tissues of an organism. The term “diluent” defines chemical compounds diluted in water that will dissolve the compound of interest as well as stabilize the biologically active form of the compound. Salts dissolved in buffered solutions are utilized as diluents in the art. One commonly used buffered solution is phosphate buffered saline because it mimics the salt conditions of human blood. Since buffer salts can control the pH of a solution at low concentrations, a buffered diluent rarely modifies the biological activity of a compound. In certain embodiments, the same substance can act as a carrier, diluent, or excipient, or have any of the two roles, or have all three roles. Thus, a single additive to the pharmaceutical composition can have multiple functions. The term “pharmaceutically acceptable” defines a carrier or diluent that does not abrogate the biological activity and properties of the compound. Compounds In one aspect, provided herein are compounds of Formula I: or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, whereinR1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; orR1and R2together with the carbon atoms to which they are attached form a polycyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; R3and R4are each independently selected from halogen, —CN, optionally substituted C1-C3alkyl, optionally substituted C1-C2alkoxy, optionally substituted C2-C3alkenyl, and cyclopropyl;R5is selected from: R6is H or C1-C3alkyl;R7is H or C1-C3alkyl optionally substituted with 1-5 halogens;R8is selected from H, halogen, —CN, optionally substituted C1-C3alkyl, and optionally substituted C1-C2alkoxy; orR3and R8together with the carbon atoms to which they are attached form a 4-, 5-, or 6-membered partially unsaturated carbocyclic ring; a 4-, 5-, or 6-membered partially unsaturated heterocyclic ring; a C6-C10aryl ring; or a 5- or 6-membered heteroaryl ring;Q is selected from N, CH, and CF; andX is O or CH2;wherein when R1and R2together with the carbon atoms to which they are attached form a C6aromatic monocyclic ring, R5is selected from: wherein 0 to 10 hydrogen atoms that are attached to one or more carbon atoms are replaced with deuterium atom(s). In some embodiments, R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, R1and R2together with the carbon atoms to which they are attached form a C5monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, R1and R2together with the carbon atoms to which they are attached form a C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R1and R2together with the carbon atoms to which they are attached form a C5monocyclic ring optionally substituted with C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R1and R2together with the carbon atoms to which they are attached form a C6monocyclic ring optionally substituted with C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, the monocyclic ring is not aromatic. In some embodiments, R1and R2together with the carbon atoms to which they are attached form a polycyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R1and R2together with the carbon atoms to which they are attached form a polycyclic ring optionally substituted with 1 to 3 substituents independently selected from C1-C6alkyl. In some embodiments, the polycyclic ring is a spirocyclic ring system. In some embodiments, the polycyclic ring is a fused ring system. In some embodiments, the polycyclic ring is a bridged ring system. In some embodiments, R3and R4are each independently selected from halogen; —CN; C1-C3alkyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkoxy; C1-C2alkoxy optionally substituted with 1 to 3 substituents independently selected from halogen; and C2-C3alkenyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkoxy; and cyclopropyl. In some embodiments, R3and R4are each independently selected from C1; —CN; C1-C3alkyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkoxy; C1-C2alkoxy optionally substituted with 1 to 3 substituents independently selected from halogen; and C2-C3alkenyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkoxy; and cyclopropyl. In some embodiments, R3and R4are each independently selected from halogen and C1-C3alkyl. In some embodiments, R3and R4are both halogen. In some embodiments, R3and R4are both C1. In some embodiments, R3and R4are both methyl. In some embodiments, R5is selected from: In some embodiments, R5is In some embodiments, R5is In some embodiments, R5is and R6is H. In some embodiments, R5is and R6is C1-C3alkyl. In some embodiments, R5is In some embodiments, R5is In some embodiments, R5is In some embodiments, R5is In some embodiments, Q is CH, CD, or CF. In some embodiments, Q is CH. In some embodiments, Q is CD. In some embodiments, Q is CF. In some embodiments, Q is N. In some embodiments, X is CH2. In some embodiments, X is O. In some embodiments, R7is H. In some embodiments, R7is C1-C3alkyl. In some embodiments, R8is selected from H; halogen; —CN; C1-C3alkyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C2alkoxy; and C1-C2alkoxy optionally substituted with 1 to 3 substituents independently selected from halogen. In some embodiments, R8is hydrogen or C1-C3alkyl. In some embodiments, R8is hydrogen. In some embodiments, R3and R8together with the carbon atoms to which they are attached form a 4-, 5-, or 6-membered partially unsaturated carbocyclic ring; a 4-, 5-, or 6-membered partially unsaturated heterocyclic ring; a C6-C10aryl ring; or a 5- or 6-membered heteroaryl ring. In some embodiments, R3and R8together with the carbon atoms to which they are attached form a 4-, 5-, or 6-membered partially unsaturated carbocyclic ring. In some embodiments, R3and R8together with the carbon atoms to which they are attached form a 4-, 5-, or 6-membered partially unsaturated heterocyclic ring. In some embodiments, R3and R8together with the carbon atoms to which they are attached form a C6-C10aryl ring. In some embodiments, R3and R8together with the carbon atoms to which they are attached form a 5- or 6-membered heteroaryl ring. In some embodiments, the compound of Formula I has the chemical structure of: In some embodiments, the compound of Formula I has the chemical structure of: In some embodiments, the compound of Formula I has the chemical structure of: In some embodiments, the compound of Formula I has the chemical structure of: In some embodiments, the compound of Formula I has the chemical structure of: In some embodiments, the compound of Formula I has the chemical structure of: wherein each R9is independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; and n is 0, 1, 2, 3, or 4. In some embodiments, the compound of Formula I has the chemical structure of: wherein each R9is independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; and n is 0, 1, 2, 3, or 4. In some embodiments, the compound of Formula I has the chemical structure of: wherein each R9is independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; and n is 0, 1, 2, 3, or 4. In some embodiments, the compound of Formula I has the chemical structure of: wherein R9aand R9bare each independently selected from H, halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; or R9aand R9btogether with the carbon atom to which they are attached form a C3-C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, the compound of Formula I has the chemical structure of: wherein R9aand R9bare each independently selected from H, halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; or R9aand R9btogether with the carbon atom to which they are attached form a C3-C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, the compound of Formula I has the chemical structure of: wherein R9aand R9bare each independently selected from H, halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; or R9aand R9btogether with the carbon atom to which they are attached form a C3-C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, the compound of Formula I has the chemical structure of: wherein each R9is independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; and n is 0, 1, 2, 3, or 4. In some embodiments, the compound of Formula I has the chemical structure of: wherein each R9is independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; and n is 0, 1, 2, 3, or 4. In some embodiments, the compound of Formula I has the chemical structure of: wherein each R9is independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; and n is 0, 1, 2, 3, or 4. In some embodiments, the compound of Formula I has the chemical structure of: wherein R9aand R9bare each independently selected from H, halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; or R9aand R9btogether with the carbon atom to which they are attached form a C3-C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, the compound of Formula I has the chemical structure of: wherein R9aand R9bare each independently selected from H, halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; or R9aand R9btogether with the carbon atom to which they are attached form a C3-C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, the compound of Formula I has the chemical structure of: wherein R9aand R9bare each independently selected from H, halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; or R9aand R9btogether with the carbon atom to which they are attached form a C3-C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, R9is selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R9is halogen. In some embodiments, R9is C1-C6alkyl. In some embodiments, R9is methyl. In some embodiments, R9is ethyl. In some embodiments, R9is C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R9is cyclopropyl optionally substituted with 1-3 halogens. In some embodiments, R9is cyclopropyl substituted with 2 fluorines. In some embodiments, R9is cyclobutyl optionally substituted with 1-3 halogens. In some embodiments, R9is cyclobutyl substituted with two fluorines. In some embodiments, R9ais selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R9ais halogen. In some embodiments, R9ais C1-C6alkyl. In some embodiments, R9ais methyl. In some embodiments, R9ais ethyl. In some embodiments, R9ais C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R9ais cyclopropyl optionally substituted with 1-3 halogens. In some embodiments, R9ais cyclopropyl substituted with 2 fluorines. In some embodiments, R9ais cyclobutyl optionally substituted with 1-3 halogens. In some embodiments, R9ais cyclobutyl substituted with two fluorines. In some embodiments, R9bis selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R9bis halogen. In some embodiments, R9bis C1-C6alkyl. In some embodiments, R9bis methyl. In some embodiments, R9bis ethyl. In some embodiments, R9bis C3-C5cycloalkyl optionally substituted with 1-3 halogens. In some embodiments, R9bis cyclopropyl optionally substituted with 1-3 halogens. In some embodiments, R9bis cyclopropyl substituted with 2 fluorines. In some embodiments, R9bis cyclobutyl optionally substituted with 1-3 halogens. In some embodiments, R9bis cyclobutyl substituted with two fluorines. In some embodiments, R9aand R9btogether with the carbon atom to which they are attached form a C3-C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, R9aand R9btogether with the carbon atom to which they are attached form a C3monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, R9aand R9btogether with the carbon atom to which they are attached form a C4monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, R9aand R9btogether with the carbon atom to which they are attached form a C5monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In some embodiments, R9aand R9btogether with the carbon atom to which they are attached form a C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. In another aspect, disclosed herein is a compound selected from the group consisting of: (R)-6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; (S)-6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(4-chloro-5-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)bicyclo[4.2.0]octa-1,3,5-trien-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2,6-d2)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(5-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)-4-methylbicyclo[4.2.0]octa-1,3,5-trien-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((7-ethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((7-(3,3-difluorocyclobutyl)-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((4′-oxo-3′,4′,6′,7′-tetrahydrospiro[cyclopentane-1,5′-cyclopenta[d]pyridazin]-1′-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,6,7-tetrahydrospiro[cyclopenta[d]pyridazine-5,1′-cyclopropan]-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((4′-oxo-3′,4′,6′,7′-tetrahydrospiro[cyclobutane-1,5′-cyclopenta[d]pyridazin]-1′-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((5-methyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((5-ethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((5-cyclopropyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((5,5-dimethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((5,5-diethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((5-oxo-3,4-diazabicyclo[4.2.0]octa-1(6),2-dien-2-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((7,7-dimethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((7,7-diethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,4b,5,5a,6-hexahydrocyclopropa[3,4]cyclopenta[1,2-d]pyridazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((5-methyl-4-oxo-3,4,5,6,7,8-hexahydro-5,8-ethanophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((9-methyl-1-oxo-2,5,6,7,8,9-hexahydro-1H-cyclohepta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2-d)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-4-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2-d)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(3,5-dichloro-2-fluoro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; 6-amino-2-(4-chloro-6-methyl-5-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)pyridin-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione; and 6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,5,6,7,8-hexahydro-5,8-methanophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof. Synthesis of the Compounds The presently disclosed compounds were synthesized using the general synthetic procedures set forth in Schemes below. The carrying out of each individual illustrated step is within the skill of an ordinary artisan, who also knows how to modify the synthetic procedures of the below schemes to synthesize the full scope of the compounds disclosed herein. The synthetic procedure for individual compounds is provided in the Examples section, below. In Scheme 1, ketones may be condensed with a secondary amine (for example pyrrolidine) generating compounds of formula A1 containing a double bond suitable for the inverse electron demand Diels Alder reaction with dichloro-1,2,4,5-tetrazine to afford cyclized products of formula A2. For examples of the inverse electron demand Diels Alder reaction, see, e.g., J. Am. Chem. Soc. 2011, 133, 12285-12292, Org. Lett. 2014, 16, 5084-5087, and Tetrahedron Letters, Vol. 38, No. 22, pp. 3805-3808, 1997. Reaction of A2 and A3 with the aid of CuI and base (e.g. K2CO3) in a polar aprotic solvent (e.g. DMSO) affords compounds of formula A4. Hydrolysis of the chloropyridazine (for example with NaOAc, acetic acid at elevated temperature with a basic, aqueous workup) afford compounds A5, where the separation of potential regioisomers is facilitated. Scheme 2 describes the synthesis of compounds of Formula B3. Compounds of formula B1 (X=halogen), may be coupled with the phenolic compounds of formula B1a under a copper mediated coupling reaction in a polar aprotic solvent with base (e.g., K2CO3) at elevated temperature to afford intermediates of type B2. Alternatively, the coupling may take place with a Pd catalyst in an appropriate solvent with base. Subsequent deprotection of the protecting groups (Pg) leads to the formation of compounds of B3. As described in Scheme 3, an aromatic amine compound of Formula C1 is converted to an aza-uracil compound of Formula C2, first by generating the corresponding diazonium salt, followed by reaction with an N-(2-cyanoacetyl)-carbamate, and then cyclization, results in the formation of a compound of Formula C2. Subsequent hydrolysis of the nitrile of Formula C2 to a carboxylic acid compound of Formula E3 using described conditions. The compounds of Formula C4 can be afforded from the acid compounds C3 proceeding through an acyl azide intermediate and subsequent Curtius rearrangement. Subsequent deprotection (of the protecting group Pg) of the compound of Formula C4, (for example if Pg is boc, using HCl or TFA), leads to compounds of formula C5. Scheme 4 describes the synthesis of a compound of Formula D4. A transmetalation reaction of a compound of Formula D1 (e.g., X is Br or I) is followed by an addition to the aldehyde of general formula D1a affording the alcohol compound of Formula D2, which is then reduced to a compound of Formula D3. Deprotection of Pg of the compound of Formula D3 results in the formation of a compound of Formula D4. Scheme 5 describes boronic acid compounds of Formula E1 are coupled to bromide compounds of formula Ela under typical Suzuki-Miyaura cross-coupling reaction conditions to afford compounds of formula E2. R7in this scheme may be a protecting group (Pg). Scheme 6 depicts the synthesis of a compound of formula F2 from a compound of formula F1 in a Suzuki-Miyaura type coupling reaction with 4,4,5,5-tetramethyl-2-[(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)methyl]-1,3,2-dioxaborolane. An appropriate protecting group (Pg), or groups, may be required. Scheme 7 describes the synthesis of a compound of Formula H6. The halogen (X) of nitrobenzene compounds H1 may be displaced by a cyanoacetate (e.g., tert-butyl 2-cyanoacetate) to afford compounds H2. Removal of the ester group and concomitant reduction of the nitro group could take place under conditions such as: SnCl2, HCl, in ethanol at elevated temperature to afford H3. Under basic conditions H3 is reacted with compounds of formula H4 to yield compounds of formula H5. The CN group is subsequently removed under acidic conditions and elevated temperature to afford compounds of the formula H6. A compound of Formula K1 (Scheme 8) can be formed by reacting the aromatic amine of formula C1 with ethyl 2-chloro-2-oxoacetate in the presence of an organic base, in an appropriate organic solvent. Standard hydrolysis conditions are performed to afford compounds of formula K2. Compounds of the formula C1 may also be reacted with 5-oxo-4,5-dihydro-1,2,4-oxadiazole-3-carbonyl chloride in a non-polar organic solvent in the presence of a base to afford compounds of the formula K3. Alternatively, compounds of the formula C1 may also be reacted with 5-oxo-4,5-dihydro-1,2,4-oxadiazole-3-carboxylic acid in a polar aprotic solvent in the presence of a base and coupling agent to afford compounds of the formula K3. Compounds of the formula K4 may be produced by reaction of compounds of formula C1 with 5-oxo-4,5-dihydro-1,2,4-oxadiazole-3-carbaldehyde under standard reductive amination conditions. Alternatively, compounds of the formula K4 may be produced by reaction of compounds of formula C1 with 3-(bromomethyl)-1,2,4-oxadiazol-5(4H)-one (or, for example, 3-(chloromethyl)-1,2,4-oxadiazol-5(4H)-one) under basic conditions with optional heating. Scheme 9 depicts the synthesis of compounds of formula M3. The aryl halide compounds M1 are reacted with methyl propiolate under typical Sonogoshira conditions to afford compounds of formula M2. Cyclization towards heterocycles of the formula M3 occurs under conditions described in the literature (e.g., J. Med. Chem. 2002, 45, 9, 1785-1798; J. Med. Chem. 2013, 56, 5, 1894-1907). Compounds of formula M3 can alternatively be generated via other described methods (e.g., J. Org. Chem. 2000, 65, 4, 1003-1007; J. Med. Chem. 1989, 32, 9, 2116-2128). Scheme 10 describes the synthesis of compounds of formula M12. Aryl cyanide compounds M10 can be transformed to compounds of formula M11 via the addition of hydroxylamine. Further conversion to compounds of formula M12 proceeds via the addition of carbonyl diimidazole with base in an appropriate solvent, often at elevated temperature (see, e.g., Molecular Pharmaceutics, 16(4), 1489-1497; 2019). Compounds of the formula M10 may be synthesized from the aryl halides M1 via several described methods using either a copper catalyst (known as the Rosenmund-von Braun reaction) or alternatively using a Pd catalyst (see, e.g., J. Am. Chem. Soc., 2011, 133, 10999-11005). The aryl cyanides of formula M10 may also be generated from the amine using the Sandmeyer reaction. Pharmaceutical Compositions In another aspect, disclosed herein are pharmaceutical compositions comprising, consisting essentially of, or consisting of a compound as described herein, and at least one pharmaceutically acceptable excipient. In another aspect, disclosed herein are pharmaceutical compositions comprising, consisting essentially of, or consisting of a compound of Formula I, as described herein, and at least one pharmaceutically acceptable excipient. The pharmaceutical composition disclosed herein may comprise a pharmaceutically acceptable carrier, such as diluents, disintegrants, sweetening agents, glidants, or flavoring agents and may be formulated into an oral dosage form such as tablets, capsules, powders, granules, suspensions, emulsions, or syrups; or a parenteral dosage form such as liquids for external use, suspensions for external use, emulsions for external use, gels (ointments or the like), inhaling agents, spraying agents, injections, etc. Said dosage forms may be formulated in various forms, e.g., a dosage form for single administration or for multiple administrations. The pharmaceutical composition disclosed herein may include excipients such as lactose, corn starch, or the like, glidants such as magnesium stearate, etc., emulsifying agents, suspending agents, stabilizers, and isotonic agents, etc. If desired, a sweetening agent and/or a flavoring agent may be added. Exemplary excipients include, without limitation, polyethylene glycol (PEG), hydrogenated castor oil (HCO), cremophors, carbohydrates, starches (e.g., corn starch), inorganic salts, antimicrobial agents, antioxidants, binders/fillers, surfactants, lubricants (e.g., calcium or magnesium stearate), glidants such as talc, disintegrants, diluents, buffers, acids, bases, film coats, combinations thereof, and the like. Specific carbohydrate excipients include, for example: monosaccharides, such as fructose, maltose, galactose, glucose, D-mannose, sorbose, and the like; disaccharides, such as lactose, sucrose, trehalose, cellobiose, and the like; polysaccharides, such as raffinose, melezitose, maltodextrins, dextrans, starches, and the like; and alditols, such as mannitol, xylitol, maltitol, lactitol, xylitol, sorbitol (glucitol), pyranosyl sorbitol, myoinositol, and the like. Inorganic salt or buffers include, but are not limited to, citric acid, sodium chloride, potassium chloride, sodium sulfate, potassium nitrate, sodium phosphate monobasic, sodium phosphate dibasic, and combinations thereof. Suitable antioxidants for use in the present disclosure include, for example, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorous acid, monothioglycerol, propyl gallate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, and combinations thereof. Additional exemplary excipients include surfactants such as polysorbates, e.g., “Tween 20” and “Tween 80,” and pluronics such as F68 and F88 (both of which are available from BASF, Mount Olive, N.J.), sorbitan esters, lipids (e.g., phospholipids such as lecithin and other phosphatidylcholines, and phosphatidylethanolamines), fatty acids and fatty esters, steroids such as cholesterol, and chelating agents, such as EDTA, zinc and other such suitable cations. Further, a composition disclosed herein may optionally include one or more acids or bases. Non-limiting examples of acids that can be used include those acids selected from the group consisting of hydrochloric acid, acetic acid, phosphoric acid, citric acid, malic acid, lactic acid, formic acid, trichloroacetic acid, nitric acid, perchloric acid, phosphoric acid, sulfuric acid, fumaric acid, and combinations thereof. Non-limiting examples of suitable bases include bases selected from the group consisting of sodium hydroxide, sodium acetate, ammonium hydroxide, potassium hydroxide, ammonium acetate, potassium acetate, sodium phosphate, potassium phosphate, sodium citrate, sodium formate, sodium sulfate, potassium sulfate, potassium fumerate, and combinations thereof. The amount of any individual excipient in the composition will vary depending on the role of the excipient, the dosage requirements of the active agent components, and particular needs of the composition. Generally, however, the excipient will be present in the composition in an amount of about 1% to about 99% by weight, preferably from about 5% to about 98% by weight, more preferably from about 15 to about 95% by weight of the excipient. In general, the amount of excipient present in a composition of the disclosure is selected from the following: at least about 2%, 5%0, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or even 95% by weight. The pharmaceutical compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s). Techniques for formulation and administration of the compounds of the instant application may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., 18th edition, 1990. Suitable routes of administration may, for example, include oral, transdermal, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as inhalation, intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections. The pharmaceutical compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes. These pharmaceutical compositions, then, may be formulated in a conventional manner using one or more known physiologically acceptable carriers comprising excipients and/or auxiliaries, which facilitate processing of the active compounds into preparations that can be used pharmaceutically. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences, above. Pharmaceutical compositions suitable for use in the presently disclosed formulations include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. In some embodiments, a therapeutically effective amount means an amount of compound effective to alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Although the exact dosage can be determined on a drug-by-drug basis, in most cases, some generalizations regarding the dosage can be made. The daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.001 mg and 1000 mg of each ingredient, preferably between 0.01 mg and 500 mg, for example 1 to 200 mg or each active ingredient of the pharmaceutical compositions disclosed herein or a pharmaceutically acceptable salt thereof calculated as the free base or free acid, the composition being administered 1 to 4 times per day or per week. Alternatively, the compositions disclosed herein may be administered by continuous such as sustained, delayed, or extended release, preferably at a dose of each ingredient up to 500 mg per day. Thus, the total daily dosage by oral administration of each ingredient will typically be in the range 0.1 mg to 2000 mg. Methods of Treatment In another aspect, disclosed herein are methods of treating a thyroid hormone receptor related disorder in a patient, the method comprising, consisting essentially of, or consisting of the steps of identifying a patient in need of treatment for the thyroid hormone receptor related disorder, and administering to the patient, or contacting the patient with, a compound as described herein. In another aspect, disclosed herein are methods of treating a thyroid hormone receptor related disorder in a patient, the method comprising, consisting essentially of, or consisting of the steps of identifying a patient in need of treatment for the thyroid hormone receptor related disorder, and administering to the patient, or contacting the patient with, a compound of Formula I, as described herein. In some embodiments, a health care professional, such as a physician, physician's assistant, nurse practitioner, or the like, identifies an individual as being in need of treatment for the thyroid hormone receptor related disorder, and/or a candidate for treatment with a compound disclosed herein. The identification may be based on medical test results, non-responsiveness to other, first-line therapies, the specific nature of the particular liver disorder, or the like. In some embodiments, the thyroid hormone receptor related disorder is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. In another aspect, disclosed herein are methods of treating a disorder or disease in a subject in need thereof, the method comprising, consisting essentially of, or consisting of administering to the subject a therapeutically effective amount of a compound or composition disclosed herein, wherein the disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. In another aspect, disclosed herein are methods of treating NASH in a subject in need thereof, the method comprising, consisting essentially of, or consisting of administering to the subject a therapeutically effective amount of a compound or composition disclosed herein. In another aspect, disclosed herein are methods of treating obesity in a subject in need thereof, the method comprising, consisting essentially of, or consisting of administering to the subject a therapeutically effective amount of a compound or composition disclosed herein. In another aspect, disclosed herein are methods of treating hyperlipidemia in a subject in need thereof, the method comprising, consisting essentially of, or consisting of administering to the subject a therapeutically effective amount of a compound or composition disclosed herein. In another aspect, disclosed herein are methods of treating hypercholesterolemia in a subject in need thereof, the method comprising, consisting essentially of, or consisting of administering to the subject a therapeutically effective amount of a compound or composition disclosed herein. In another aspect, disclosed herein are methods of treating diabetes in a subject in need thereof, the method comprising, consisting essentially of, or consisting of administering to the subject a therapeutically effective amount of a compound or composition disclosed herein. In another aspect, disclosed herein are methods of treating liver steatosis in a subject in need thereof, the method comprising, consisting essentially of, or consisting of administering to the subject a therapeutically effective amount of a compound or composition disclosed herein. In some embodiments, the compound of Formula I, as described herein, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition; is administered in combination with a KHK inhibitor, an FXR agonist, a SSAO inhibitor, a FASN inhibitor, or a SCD1 modulator. In some embodiments, the KHK inhibitor is PF-06835919; the FXR agonist is TERN-101 (LY2562175), Tropifexor, obeticholic acid (OCA), or ASC42; the SSAO inhibitor is TERN-201; the FASN inhibitor is ASC40; and the SCD1 modulator is aramchol. In another aspect, disclosed herein are methods of selectively modulating the activity of a thyroid hormone receptor beta (THR-β) comprising, consisting essentially of, or consisting of contacting a compound as described herein, with a thyroid hormone receptor. In some embodiments, the contacting is in vitro or ex vivo, whereas in other embodiments, the contacting is in vivo. In another aspect, disclosed herein are methods of selectively modulating the activity of a thyroid hormone receptor beta (THR-β) comprising, consisting essentially of, or consisting of contacting a compound of Formula I, as described herein, with a thyroid hormone receptor. In some embodiments, the contacting is in vitro or ex vivo, whereas in other embodiments, the contacting is in vivo. In another aspect, disclosed herein are methods of selectively modulating the activity of a thyroid hormone receptor beta (THR-β) comprising, consisting essentially of, or consisting of contacting a composition described herein, with a thyroid hormone receptor. In some embodiments, the contacting is in vitro or ex vivo, whereas in other embodiments, the contacting is in vivo. EXAMPLES Example 1. Preparation of Compounds 1a and 1b A solution of 2-methylcyclopentan-1-one (6.00 g, 61.1 mmol) pyrrolidine (6.52 g, 91.7 mmol) and TsOH (1.16 g, 6.11 mmol) in toluene (70 mL) was refluxed for overnight under N2. The resulting solution was concentrated under reduced pressure to provide 6 g of 1-(5-methylcyclopent-1-en-1-yl)pyrrolidine as a white solid. LC-MS (ESI, m/z): 152 [M+H]+. The crude product was used in the next step without further purification. A solution of dichloro-1,2,4,5-tetrazine (3.00 g, 19.9 mmol) and 1-(5-methylcyclopent-1-en-1-yl)pyrrolidine (6.01 g, 39.7 mmol) in toluene (100 mL) was stirred for overnight at 80° C. under N2. The resulting mixture was concentrated under reduced pressure. The residue was chromatographed on a silica gel column with petroleum ether/ethyl acetate (19/1) to provide 1.5 g (yield 37%) of 1,4-dichloro-5-methyl-5H,6H,7H-cyclopenta[d]pyridazine as a pink solid. LC-MS (ESI, m/z): 203 [M+H]+. To a solution of 1,4-dichloro-5-methyl-5H,6H,7H-cyclopenta[d]pyridazine (1.00 g, 4.92 mmol) in dimethyl sulfoxide (20 mL) were added 4-amino-2,6-dichlorophenol (964 mg, 5.42 mmol), K2CO3(1.36 g, 9.85 mmol), CuI (468 mg, 2.46 mmol) in portions. The resulting mixture was stirred overnight at 90° C. for 16 h under nitrogen atmosphere and quenched with water (60 mL). The resulting mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, the solids were removed by filtration and the filtrate was concentrated under reduced pressure to provide 1 g of 3,5-dichloro-4-([4-chloro-5-methyl-5H,6H,7H-cyclopenta[d]pyridazin-1-yl]oxy)aniline as a brown solid. LC-MS (ESI, m/z): 344 [M+H]+. The crude product was used in the next step without further purification. To a solution of 3,5-dichloro-4-({4-chloro-5-methyl-5H,6H,7H-cyclopenta[d]pyridazin-1-yl}oxy)aniline (850 mg, 2.45 mmol) in acetic acid (10 mL) was added sodium acetate (1.01 g, 12.3 mmol). The mixture was stirred overnight at 100° C. The resulting mixture was concentrated and diluted with water (50 mL). The mixture was adjusted to pH 9 with NaOH (aq., 1 M). The resulting solution was extracted with ethyl acetate (3×50 mL) and the combined organic layers were concentrated under reduced pressure then dissolved in CH3OH (5 mL) and NaOH (834 mg, 20.9 mmol) in water (5 mL). The resulting mixture was stirred overnight at 120° C. The resulting mixture was concentrated and extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, the solids were removed by filtration and the filtrate was concentrated under reduced pressure. The residue was chromatographed on a silica gel column with petroleum ether/ethyl acetate to provide 250 mg (yield 31%) of 4-(4-amino-2,6-dichlorophenoxy)-7-methyl-2H,5H,6H,7H-cyclopenta[d]pyridazin-1-one as a brown solid.1H NMR (300 MHz, DMSO-d6) δ 12.01 (s, 1H), 6.62 (s, 2H), 5.59 (s, 2H), 3.19-3.29 (m, 1H), 2.78-2.99 (m, 2H), 2.29-2.39 (m, 1H), 1.61-1.72 (m, 1H), 1.23 (d, J=6.0 Hz, 3H). LC-MS (ESI, m/z): 326 [M+H]+. To a solution of 4-(4-amino-2,6-dichlorophenoxy)-7-methyl-2H,5H,6H,7H-cyclopenta[d]pyridazin-1-one (200 mg, 0.613 mmol), water (12 mL), HCl (conc., 5 mL) and acetic acid (15 mL) was added sodium nitrite (84.6 mg, 1.22 mmol) in water (5 mL) dropwise at 0° C. After the addition, the reaction was stirred at 0° C. for 45 min. Then the reaction mixture was added to a solution of ethyl N-(2-cyanoacetyl)carbamate (143 mg, 0.919 mmol) in water (12 mL) and pyridine (5 mL) at 0° C. quickly. The resulting mixture was stirred at 0° C. for 1 h and filtered. The filter cake was washed with water (30 mL) and petroleum ether (30 mL), dried under the IR lamp to provide 190 mg of ethyl (2-cyano-2-(2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)hydrazineylidene)acetyl)carbamate as an orange solid. LC-MS (ESI, m/z): 493 [M+H]+. The crude product was used in the next step without further purification. To a solution of ethyl (2-cyano-2-(2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)hydrazineylidene)acetyl)carbamate (190 mg, 0.385 mmol) in DMA (8 mL) was added K2CO3(189 mg, 1.93 mmol). The reaction was stirred at 120° C. for 2 h and cooled to room temperature. The resulting mixture was quenched with water (30 mL) and extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to provide 180 mg of 2-[3,5-dichloro-4-({5-methyl-4-oxo-3H,5H,6H,7H-cyclopenta[d]pyridazin-1-yl}oxy)phenyl]-3,5-dioxo-4H-1,2,4-triazine-6-carbonitrile as a red solid. LC-MS (ESI, m/z): 447 [M+H]+. The crude product was used in the next step without further purification. A solution of 2-[3,5-dichloro-4-({5-methyl-4-oxo-3H,5H,6H,7H-cyclopenta[d]pyridazin-1-yl}oxy)phenyl]-3,5-dioxo-4H-1,2,4-triazine-6-carbonitrile (180 mg, 0.402 mmol) in HCl (2 mL) was added acetic acid (4 mL). The resulting mixture was stirred 4 h at 100° C. and concentrated reduced pressure. The residue was diluted with NaHCO3(aq., 20 mL), the resulting mixture was extracted with ethyl acetate (2×15 mL) and the organic layers were discarded. The pH value of the aqueous layers was adjusted to 6 with HCl (conc.). The resulting solution was extracted with ethyl acetate (3×15 mL). The precipitate was generated, isolated by filtration, and dried under reduced pressure to provide 100 mg of 2-[3,5-dichloro-4-({5-methyl-4-oxo-3H,5H,6H,7H-cyclopenta[d]pyridazin-1-yl}oxy)phenyl]-3,5-dioxo-4H-1,2,4-triazine-6-carboxylic acid as a brown solid. LC-MS (ESI, m/z): 466 [M+H]+. The crude product was used in the next step without further purification. To a solution of 2-[3,5-dichloro-4-({5-methyl-4-oxo-3H,5H,6H,7H-cyclopenta[d]pyridazin-1-yl}oxy)phenyl]-3,5-dioxo-4H-1,2,4-triazine-6-carboxylic acid (100 mg, 0.214 mmol) in t-butanol (5 mL) was added triethylamine (108 mg, 1.07 mmol) and diphenylphosphoryl azide (DPPA, 177 mg, 0.642 mmol). The reaction was stirred at 85° C. overnight then concentrated under reduced pressure. The residue was dissolved in ethyl acetate (40 mL) and washed with NaHCO3(aq., 40 mL) and brine (40 mL), dried over anhydrous Na2SO4. The solids were removed by filtration, the filtrate was concentrated under reduced pressure to provide 80 mg of t-butyl N-{2-[3,5-dichloro-4-({5-methyl-4-oxo-3H,5H,6H,7H-cyclopenta[d]pyridazin-1-yl}oxy)phenyl]-3,5-dioxo-4H-1,2,4-triazin-6-yl}carbamate as a brown solid. LC-MS (ESI, m/z): 537 [M+H]+. The crude product was used in the next step without further purification. To a solution of t-butyl N-{2-[3,5-dichloro-4-({5-methyl-4-oxo-3H,5H,6H,7H-cyclopenta[d]pyridazin-1-yl}oxy)phenyl]-3,5-dioxo-4H-1,2,4-triazin-6-yl}carbamate (80.0 mg, 14.9 mmol) in CH2Cl2(2 mL) was added trifluoroacetic acid (2 mL). The reaction was stirred at room temperature for 4 h and concentrated under reduced pressure. The crude product (80 mg) was purified by reverse phase prep LC. Column: XBridge Prep Phenyl OBD Column, 5 mm, 19×250 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1% NH3·H2O), Mobile Phase B: CH3CN; Flow rate: 25 mL/min; Gradient: 20% B to 42% B in 7 min; 210 nm; to provide 24 mg (yield 36%) of 6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione. LC-MS (ESI, m/z): 437 [M+H]+. The enantiomers were purified by prep-chiral chromatography (Column: CHIRALPAK IA, 2×25 cm, 5 mm; Mobile Phase A: HEX: DCM=3:1 (0.1% DEA). HPLC, Mobile Phase B: EtOH. Flow rate: 20 mL/min; Gradient: 20% B to 20% B in 30 min; collected at 220/254 nm; Rt 1: 11.841 min; Rt 2: 14.619 min; Injection Volume: 0.5 mL).1H NMR (400 MHz, DMSO-d6) δ 7.88 (s, 2H), 6.33 (s, 2H), 3.10-3.22 (m, 1H), 2.88-3.09 (m, 2H), 2.31-2.43 (m, 1H), 1.64-1.84 (m, 1H), 1.26 (d, J=8.0 Hz, 3H). The first eluting enantiomer afforded compound 1a, (6 mg, 30% yield) of a white solid. LC-MS Method A, Rt: 0.629 min (ESI, m/z): 437 [M+H]+. The second eluting enantiomer afforded compound 1b (4.9 mg, 29% yield) of a white solid. LC-MS Method A, Rt: 0.621 min (ESI, m/z): 437 [M+H]+. The absolute stereochemistry of the enantiomers was not established. Compounds 2a/2b and 3a/3b can be synthesized in a similar manner as that described for compounds 1a/1b. LC-MS Methods LC MethodCol TRunnameInstrumentColumnMobile phaseGradientFlow(° C.)timeAShimadzuKinetexA: Water/6.5 mMFrom 90% A to1.2402 minLCMS-2020EVO C18NH4HCO3+ NH45% A in 1.19 min,mL/min(2.6 μm,OH (pH = 10)held for 0.6 min,3.0 × 30B: CH3CNto 90% A in 0.02mm)min, held for0.18 min Biological Assays THR Biochemical Assay (Assay 1) The TR-FRET thyroid receptor beta coactivator assay was used with slight, optimized modifications of the manufacturer's protocol (Invitrogen). The assay uses a terbium-labeled anti-GST antibody, a glutathione-S-transferase (GST) tagged human thyroid receptor, beta or alpha, ligand-binding domain (LBD), and a fluorescein labeled SRC2-2 coactivator peptide. The antibody interacts with the LBD, where the agonist also binds, resulting in increased affinity for the SRC2-2 coactivator peptide causing energy transfer of the acceptor fluorophore and a FRET emission shift from 495 to 520 nm. The energy transfer was detected as an increase in the fluorescence emission of the fluorescein acceptor, and a decrease in the fluorescence emission of the terbium donor. The assay was performed in a 384-well black plate in a final volume of 20 μL. Serial dilution of various test agonists was performed in DMSO (1% final DMSO concentration) and added to the test plate. Thyroid receptor beta LBD was added to the plate at a final concentration of 1 nM, followed by the mixture of the fluorescein labeled SRC2-2 coactivator peptide, and the terbium-labeled anti-GST antibody at final concentrations of 200 nM and 2 nM respectively. The assay was incubated for 1 hr at rt protected from light. The TR-FRET was then measured on a Victor multilabel reader (Perkin Elmer) using an excitation wavelength of 340 nm with emission filters of 495 nm and 520 nm. The assay was quantified by expressing a ratio (520:495) of the intensities, and the resulting activation curves; EC50values were generated using a sigmoidal dose response (variable slope) equation in GraphPad™ Prism 8.0. Compounds of Formula (I) are active as THR-beta agonists as shown in Table 1, where: for Assay 1: ‘A’ indicates an EC50<50 nM, ‘B’ indicates an EC50of >50 nM and <250 nM, ‘C’ indicates an EC50>250 nM and <1000 nM, ‘D’ indicates an EC50>1000 nM and <25000 nM, and ‘E’ indicates an EC50>25000 nM. TABLE 1Compound numberAssay 11aA1bB Diet-Induced Obese (DIO) Mouse Model of NASH C57BL/6J mice are fed a high-fat diet for 10 weeks to induce obesity and injected intraperitoneally twice weekly with carbon tetrachloride (CCl4) for an additional 4 weeks to induce fibrosis. Mice fed a normal chow diet are used as healthy controls. Concomitant with CCl4dosing, mice are treated with vehicle or with a compound disclosed herein, administered by oral gavage once daily for 28 days. Drug exposure is measured in a separate experiment in lean male C57BL/6J mice. Livers of mice in the NASH study are harvested and evaluated for liver steatosis and fibrosis by histology and whole transcriptome analysis in the liver using RNA sequencing. Target engagement is confirmed by monitoring expression of TRβ-regulated genes. Human Clinical Study: NASH In a randomized, double-blind, placebo-controlled study, adult patients (with biopsy confirmed NASH (fibrosis stages 1-3) and hepatic fat fraction of at least 10% at baseline when assessed by MRI-proton density fat fraction (MRI-PDFF) are administered a compound disclosed herein or placebo. Serial hepatic fat measurements are obtained at weeks 12 and 36, and a second liver biopsy is obtained at week 36. The primary endpoint is relative change in MRI-PDFF assessed hepatic fat compared with placebo at week 12 in patients who have both a baseline and week 12 MRI-PDFF. REFERENCES 1. Younossi, Z M, Koenig, A B, Abdelatif, D, Fazel, Y, Henry, L, Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology, 2016, 64(1):73e84.2. Gastroenterology. 2012 June; 142(7): 1592-609. doi: 10.1053/j.gastro.2012.04.001. Epub 2012 May 15.3. Serfaty, L., Lemoine, M. Definition and natural history of metabolic steatosis: clinical aspects of NAFLD, NASH and cirrhosis. Diabetes and Metabolism, 2008, 34 (6 Pt 2):634e637.4. Hepatology. 2012 October; 56(4): 1580-1584. doi: 10.1002/hep.260315. Dulai, P S, Singh, S, Patel, J, Soni, M, Prokop, L J, Younossi, Z, et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: systematic review and meta-analysis. Hepatology, 2017, 65(5):1557e1565.6. Younossi, Z M, Loomba, R, Rinella, M E, Bugianesi, E, Marchesini, G, Neuschwander-Tetri, B A, et al. Current and future therapeutic regimens for non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Hepatology, 2018, 68(1):349e360.7. Harvey C B, Williams G R. Mechanism of thyroid hormone action. Thyroid, 2002 June; 12(6):441-6.8. Bookout A L, Jeong Y, Downes M, Yu R T, Evans R M, Mangelsdorf D J. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell, 2006, 126:789-7999. Flamant F, Baxter J D, Forrest D, Refetoff S, Samuels H H, Scanlan T S, Vennstrom B, Samarut J. International union of pharmacology. LIX. The pharmacology and classification of the nuclear receptor superfamily: thyroid hormone receptors. Pharmacol. Rev., 2006, 58:705-71110. Haning H, Woltering M, Mueller U, Schmidt G, Schmeck C, Voehringer V, Kretschmer A, Pernerstorfer J. Bioorg. Med Chem Lett., 2005 Apr. 1, 15(7): 1835-40. Novel heterocyclic thyromimetics.11. Hirano T, Kagechika H. Thyromimetics: a review of recent reports and patents (2004-2009). Expert Opin Ther Pat., 2010 February; 20(2):213-28. doi: 10.1517/13543770903567069.12. Kowalik M A, Columbano A, Perra A. Thyroid Hormones, Thyromimetics and Their Metabolites in the Treatment of Liver Disease. Front Endocrinol (Lausanne), 2018 Jul. 10; 9:382. doi: 10.3389/fendo.2018.00382. eCollection 2018.13. Erion M D, Cable E E, Ito B R, Jiang H, Fujitaki J M, Finn P D, Zhang B H, Hou J, Boyer S H, van Poelje P D, Linemeyer D L. Targeting thyroid hormone receptor-beta agonists to the liver reduces cholesterol and triglycerides and improves the therapeutic index. Proc Natl Acad Sci USA., 2007 Sep. 25; 104(39):15490-5. Epub 2007 Sep. 18.14. Hartley M D, Kirkemo L L, Banerji T, Scanlan T S. A Thyroid Hormone-Based Strategy for Correcting the Biochemical Abnormality in X-Linked Adrenoleukodystrophy. Endocrinology 2017, 158(5), p 1328-1338. doi: 10.1210/en.2016-1842.15. Milanesi A, Brent G A. Beam Me In: Thyroid Hormone Analog Targets Alternative Transporter in Mouse Model of X-Linked Adrenoleukodystrophy. Endocrinology 2017, 158, p 1116-1119. doi: 10.1210/en.2017-00206. EMBODIMENTS Embodiment P1. A compound of Formula I: or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, whereinR1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; orR1and R2together with the carbon atoms to which they are attached form a polycyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens;R3and R4are each independently selected from halogen, —CN, optionally substituted C1-C3alkyl, optionally substituted C1-C2alkoxy, optionally substituted C2-C3alkenyl, and cyclopropyl;R5is selected from: R6is H or C1-C3alkyl;R7is H or C1-C3alkyl;R8is selected from H, halogen, —CN, optionally substituted C1-C3alkyl, and optionally substituted C1-C2alkoxy; orR3and R8together with the carbon atoms to which they are attached form a 4-, 5-, or 6-membered partially unsaturated carbocyclic ring; a 4-, 5-, or 6-membered partially unsaturated heterocyclic ring; a C6-C10aryl ring; or a 5- or 6-membered heteroaryl ring;Q is selected from N, CH, and CF; andX is O or CH2;wherein 0 to 10 hydrogen atoms that are attached to one or more carbon atoms are replaced with deuterium atom(s). Embodiment P2. The compound of embodiment P1, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. Embodiment P3. The compound of embodiment P1 or embodiment P2, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. Embodiment P4. The compound of embodiment P1 or embodiment P2, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with C3-C5cycloalkyl optionally substituted with 1-3 halogens. Embodiment P5. The compound of embodiment P1, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a polycyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. Embodiment P6. The compound of any one of embodiments P1-P5, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R4are each independently selected from halogen; —CN; C1-C3alkyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkoxy; C1-C2alkoxy optionally substituted with 1 to 3 substituents independently selected from halogen; and C2-C3alkenyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkoxy; and cyclopropyl. Embodiment P7. The compound of any one of embodiments P1-P6, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R4are each independently selected from halogen and C1-C3alkyl. Embodiment P8. The compound of any one of embodiments P1-P7, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R4are both halogen. Embodiment P9. The compound of any one of embodiments P1-P7, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R4are both methyl. Embodiment P10. The compound of any one of embodiments P1-P9, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R8is selected from H; halogen; —CN; C1-C3alkyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C2alkoxy; and C1-C2alkoxy optionally substituted with 1 to 3 substituents independently selected from halogen. Embodiment P11. The compound of any one of embodiments P1-P5, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R8together with the carbon atoms to which they are attached form a 4-, 5-, or 6-membered partially unsaturated carbocyclic ring; a 4-, 5-, or 6-membered partially unsaturated heterocyclic ring; a C6-C10aryl ring; or a 5- or 6-membered heteroaryl ring. Embodiment P12. The compound of any one of embodiments P1-P11, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R5is Embodiment P13. The compound of any one of embodiments P1-P12, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein X is CH2. Embodiment P14. The compound of any one of embodiments P1-P12, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein X is O. Embodiment P15. The compound of any one of embodiments P1-P14, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R7is H. Embodiment P16. The compound of any one of embodiments P1-P14, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R7is C1-C3alkyl. Embodiment P17. The compound of any one of embodiments P1-P16, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein Q is CH. Embodiment P18. The compound of any one of embodiments P1-P16, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein Q is N. Embodiment P19. A compound selected from the group consisting of:(R)-6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;(S)-6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(4-chloro-5-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)bicyclo[4.2.0]octa-1,3,5-trien-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2,6-d2)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(5-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)-4-methylbicyclo[4.2.0]octa-1,3,5-trien-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-ethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-(3,3-difluorocyclobutyl)-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4′-oxo-3′,4′,6′,7′-tetrahydrospiro[cyclopentane-1,5′-cyclopenta[d]pyridazin]-1′-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,6,7-tetrahydrospiro[cyclopenta[d]pyridazine-5,1′-cyclopropan]-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4′-oxo-3′,4′,6′,7′-tetrahydrospiro[cyclobutane-1,5′-cyclopenta[d]pyridazin]-1′-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-methyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-ethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-cyclopropyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5,5-dimethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5,5-diethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-oxo-3,4-diazabicyclo[4.2.0]octa-1(6),2-dien-2-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7,7-dimethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7,7-diethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,4b,5,5a,6-hexahydrocyclopropa[3,4]cyclopenta[1,2-d]pyridazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-methyl-4-oxo-3,4,5,6,7,8-hexahydro-5,8-ethanophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((9-methyl-1-oxo-2,5,6,7,8,9-hexahydro-1H-cyclohepta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2-d)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2-d)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-2-fluoro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(4-chloro-6-methyl-5-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)pyridin-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione; and6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,5,6,7,8-hexahydro-5,8-methanophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof. Embodiment P20. A pharmaceutical composition comprising the compound of any one of embodiments P1-P19, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient. Embodiment P21. A method of treating a disorder or disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of embodiments P1-P19, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of embodiment P20, wherein the disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment P22. Use of the compound of any one of embodiments P1-P19, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment P23. A compound of any one of embodiments P1-P19, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, for use in treating a disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment P24. A composition of embodiment P20 for use in treating a disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment P25. A method of treating a thyroid hormone receptor related disorder in a patient, the method comprising the steps of: identifying a patient in need of treatment for the thyroid hormone receptor related disorder, and administering to the patient, or contacting the patient with, the compound of any one of embodiments P1-P19, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of embodiment P20. Embodiment P26. The method of embodiment P25, wherein the thyroid hormone receptor related disorder is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment P27. A method of selectively modulating the activity of a thyroid hormone receptor beta (THR-β) comprising contacting the compound of any one of embodiments P1-P19, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, with the thyroid hormone receptor. Embodiment P28. The method of embodiment P27, wherein the contacting is in vitro or ex vivo. Embodiment P29. The method of embodiment P27, wherein the contacting is in vivo. Embodiment P30. A compound of any one of embodiments P1-P19, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, for use in selectively modulating the activity of a thyroid hormone receptor beta (THR-β). Embodiment P31. A composition of embodiment P20 for use in selectively modulating the activity of a thyroid hormone receptor beta (THR-β). ADDITIONAL EMBODIMENTS Embodiment 1. A compound of Formula I: or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, whereinR1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens; orR1and R2together with the carbon atoms to which they are attached form a polycyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens;R3and R4are each independently selected from halogen, —CN, optionally substituted C1-C3alkyl, optionally substituted C1-C2alkoxy, optionally substituted C2-C3alkenyl, and cyclopropyl;R5is selected from: R6is H or C1-C3alkyl;R7is H or C1-C3alkyl;R8is selected from H, halogen, —CN, optionally substituted C1-C3alkyl, and optionally substituted C1-C2alkoxy; orR3and R8together with the carbon atoms to which they are attached form a 4-, 5-, or 6-membered partially unsaturated carbocyclic ring; a 4-, 5-, or 6-membered partially unsaturated heterocyclic ring; a C6-C10aryl ring; or a 5- or 6-membered heteroaryl ring;Q is selected from N, CH, and CF; andX is O or CH2;wherein when R1and R2together with the carbon atoms to which they are attached form a C6aromatic monocyclic ring, R5is selected from: andwherein 0 to 10 hydrogen atoms that are attached to one or more carbon atoms are replaced with deuterium atom(s). Embodiment 2. The compound of embodiment 1, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. Embodiment 3. The compound of embodiment 1 or embodiment 2, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. Embodiment 4. The compound of embodiment 1 or embodiment 2, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C5monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. Embodiment 5. The compound of embodiment 1 or embodiment 2, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C6monocyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkyl. Embodiment 6. The compound of embodiment 1 or embodiment 2, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C4-C7monocyclic ring optionally substituted with C3-C5cycloalkyl optionally substituted with 1-3 halogens. Embodiment 7. The compound of embodiment 1 or embodiment 2, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C5monocyclic ring optionally substituted with C3-C5cycloalkyl optionally substituted with 1-3 halogens. Embodiment 8. The compound of embodiment 1 or embodiment 2, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a C6monocyclic ring optionally substituted with C3-C5cycloalkyl optionally substituted with 1-3 halogens. Embodiment 9. The compound of any one of embodiments 1-8, wherein the monocyclic ring is not aromatic. Embodiment 10. The compound of embodiment 1, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R1and R2together with the carbon atoms to which they are attached form a polycyclic ring optionally substituted with 1 to 3 substituents independently selected from halogen, C1-C6alkyl, and C3-C5cycloalkyl optionally substituted with 1-3 halogens. Embodiment 11. The compound of any one of embodiments 1-10, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R4are each independently selected from halogen; —CN; C1-C3alkyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkoxy; C1-C2alkoxy optionally substituted with 1 to 3 substituents independently selected from halogen; and C2-C3alkenyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C6alkoxy; and cyclopropyl. Embodiment 12. The compound of any one of embodiments 1-11, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R4are each independently selected from halogen and C1-C3alkyl. Embodiment 13. The compound of any one of embodiments 1-12, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R4are both halogen. Embodiment 14. The compound of any one of embodiments 1-12, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R4are both methyl. Embodiment 15. The compound of any one of embodiments 1-14, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R8is selected from H; halogen; —CN; C1-C3alkyl optionally substituted with 1 to 3 substituents independently selected from halogen and C1-C2alkoxy; and C1-C2alkoxy optionally substituted with 1 to 3 substituents independently selected from halogen. Embodiment 16. The compound of any one of embodiments 1-14, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R8is H. Embodiment 17. The compound of any one of embodiments 1-10, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3and R8together with the carbon atoms to which they are attached form a 4-, 5-, or 6-membered partially unsaturated carbocyclic ring; a 4-, 5-, or 6-membered partially unsaturated heterocyclic ring; a C6-C10aryl ring; or a 5- or 6-membered heteroaryl ring. Embodiment 18. The compound of any one of embodiments 1-16, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R5is Embodiment 19. The compound of any one of embodiments 1-18, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein X is CH2. Embodiment 20. The compound of any one of embodiments 1-18, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein X is O. Embodiment 21. The compound of any one of embodiments 1-20, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R7is H. Embodiment 22. The compound of any one of embodiments 1-20, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein R7is C1-C3alkyl. Embodiment 23. The compound of any one of embodiments 1-22, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein Q is CH. Embodiment 24. The compound of any one of embodiments 1-22, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein Q is N. Embodiment 25. A compound selected from the group consisting of:(R)-6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;(S)-6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(4-chloro-5-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)bicyclo[4.2.0]octa-1,3,5-trien-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2,6-d2)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(5-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)-4-methylbicyclo[4.2.0]octa-1,3,5-trien-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-ethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-(3,3-difluorocyclobutyl)-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4′-oxo-3′,4′,6′,7′-tetrahydrospiro[cyclopentane-1,5′-cyclopenta[d]pyridazin]-1′-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,6,7-tetrahydrospiro[cyclopenta[d]pyridazine-5,1′-cyclopropan]-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4′-oxo-3′,4′,6′,7′-tetrahydrospiro[cyclobutane-1,5′-cyclopenta[d]pyridazin]-1′-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-methyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-ethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-cyclopropyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5,5-dimethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5,5-diethyl-4-oxo-3,4,5,6,7,8-hexahydrophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-oxo-3,4-diazabicyclo[4.2.0]octa-1(6),2-dien-2-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7,7-dimethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7,7-diethyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,4b,5,5a,6-hexahydrocyclopropa[3,4]cyclopenta[1,2-d]pyridazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((5-methyl-4-oxo-3,4,5,6,7,8-hexahydro-5,8-ethanophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((9-methyl-1-oxo-2,5,6,7,8,9-hexahydro-1H-cyclohepta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2-d)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-4-((7-cyclopropyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl-2-d)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(3,5-dichloro-2-fluoro-4-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione;6-amino-2-(4-chloro-6-methyl-5-((7-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[d]pyridazin-4-yl)oxy)pyridin-2-yl)-1,2,4-triazine-3,5(2H,4H)-dione; and6-amino-2-(3,5-dichloro-4-((4-oxo-3,4,5,6,7,8-hexahydro-5,8-methanophthalazin-1-yl)oxy)phenyl)-1,2,4-triazine-3,5(2H,4H)-dione; or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof. Embodiment 26. A pharmaceutical composition comprising the compound of any one of embodiments 1-25, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient. Embodiment 27. A method of treating a disorder or disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of embodiments 1-25, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of embodiment 26, wherein the disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment 28. Use of the compound of any one of embodiments 1-25, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment 29. A compound of any one of embodiments 1-25, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, for use in treating a disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment 30. A composition of embodiment 29 for use in treating a disorder or disease is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment 31. A method of treating a thyroid hormone receptor related disorder in a patient, the method comprising the steps of:identifying a patient in need of treatment for the thyroid hormone receptor related disorder, andadministering to the patient, or contacting the patient with, the compound of any one of embodiments 1-25, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of embodiment 26. Embodiment 32. The method of embodiment 31, wherein the thyroid hormone receptor related disorder is selected from non-alcoholic steatohepatitis (NASH), obesity, hyperlipidemia, hypercholesterolemia, diabetes, liver steatosis, atherosclerosis, cardiovascular diseases, hypothyroidism, and thyroid cancer. Embodiment 33. A method of selectively modulating the activity of a thyroid hormone receptor beta (THR-β) comprising contacting the compound of any one of embodiments 1-24, or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, with the thyroid hormone receptor. Embodiment 34. The method of embodiment 33, wherein the contacting is in vitro or ex vivo. Embodiment 35. The method of embodiment 33, wherein the contacting is in vivo. Embodiment 36. A compound of any one of embodiments 1-25, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof, for use in selectively modulating the activity of a thyroid hormone receptor beta (THR-β). Embodiment 37. A composition of embodiment 26 for use in selectively modulating the activity of a thyroid hormone receptor beta (THR-β). Embodiment 38. The method of embodiments 27, 31 or 32, wherein the compound of any one of embodiments 1-25 or the stereoisomer or the tautomer thereof, or the pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of embodiment 26, is administered in combination with a KHK inhibitor, an FXR agonist, a SSAO inhibitor, a FASN inhibitor, or a SCD1 modulator. Embodiment 39. The method of embodiment 38, the KHK inhibitor is PF-06835919; the FXR agonist is TERN-101 (LY2562175), Tropifexor, obeticholic acid (OCA), or ASC42; the SSAO inhibitor is TERN-201; the FASN inhibitor is ASC40; and the SCD1 modulator is aramchol. While certain embodiments have been illustrated and described, it should be understood that changes and modifications can be made therein in accordance with ordinary skill in the art without departing from the technology in its broader aspects as defined in the following claims. The embodiments, illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms “comprising,” “including,” “containing,” etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the claimed technology. Additionally, the phrase “consisting essentially of” will be understood to include those elements specifically recited and those additional elements that do not materially affect the basic and novel characteristics of the claimed technology. The phrase “consisting of” excludes any element not specified. The present disclosure is not to be limited in terms of the particular embodiments described in this application. Many modifications and variations can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods and compositions within the scope of the disclosure, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. The present disclosure is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. It is to be understood that this disclosure is not limited to particular methods, reagents, compounds, or compositions, which can of course vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group. As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like, include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. All publications, patent applications, issued patents, and other documents referred to in this specification are herein incorporated by reference as if each individual publication, patent application, issued patent, or other document was specifically and individually indicated to be incorporated by reference in its entirety. Definitions that are contained in text incorporated by reference are excluded to the extent that they contradict definitions in this disclosure.
107,533
11858914
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS Example 1: Chemical Synthesis of Compound 3a-3i The synthesis route was as follows: The general process was as follows: 1a-1i was used as a raw material to undergo a nucleophilic substitution reaction with 6-aminoindazole to generate an intermediate 2a-2i. 4-(4-methylpiperazine)aniline and the obtained intermediate 2a-2i underwent a nucleophilic substitution reaction to generate a final product 3a-3i. Taking the synthesis of a compound 3a for example, a specific process was as follows:2,4-dichloro-5-fluoropyrimidine (333.94 mg, 2 mmol) and N, N-diisopropylethylamine (DIPEA) (516.96 mg, 4 mmol) were dissolved in DMF (4 mL) and cooled to 0° C. Then 6-aminoindazole (266.3 mg, 2 mmol) dissolved in DMF (2 mL) was added dropwise to the mixed solution. The reaction mixture was stirred at 0° C. for about 1 hour. Next, the ice bath was removed, the reaction mixture was stirred at room temperature, and the reaction was monitored by TLC. The resulting mixture was extracted with ethyl acetate (3×25 mL), washed with saturated salt solution, dried and concentrated by anhydrous Na2SO4, and then a product 2a was obtained by silica gel column chromatography. Compound 2a (263.66 mg, 1 mmol) and 4-(4-methylpiperazine)aniline (191.27 mg, 1 mmol) were dissolved in methanol (4 mL), then added with trifluoroacetic acid (TFA) (148.56 v L, 2 mmol), heated to 80° C., and the reaction was monitored by TLC. After the reaction was completed, the mixture was cooled to room temperature, the resulting mixture was adjusted to be neutral with saturated sodium bicarbonate, extracted with ethyl acetate (3×25 mL), washed with saturated salt solution, dried and concentrated by anhydrous Na2SO4, and a final product 3a was obtained by silica gel column chromatography. Characterization data of compound 3a-3i was as follows: 5-fluoro-N4-(1H-indazol-6-yl)-N2-(4-(4-methylpiperazine-1-yl) p henyl)pyrimidine-2,4-diamine (3a), yellow solid, yield: 26.9%.1H NMR (500 MHz, DMSO-d6) δ 12.93 (s, 1H), 9.41 (s, 1H), 8.99 (s, 1H), 8.08 (d, J=3.5 Hz, 1H), 8.01 (s, 1H), 7.91 (s, 1H), 7.69 (d, J=8.5 Hz, 1H), 7.52 (d, J=8.5 Hz, 3H), 6.81 (d, J=8.5 Hz, 2H), 3.06 (s, 4H), 2.57 (s, 4H), 2.30 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 170.28, 155.87, 149.81, 149.73, 145.77, 140.26, 139.40, 137.05, 133.30, 133.15, 120.02, 119.33, 116.55, 115.88, 101.58, 54.61, 48.85, 14.05; ESI-MS m/z: 419.2 (M+H)+. 5-chloro-N4-(1H-indazol-6-yl)-N2-(4-(4-methylpiperazine-1-yl) p henyl)pyrimidine-2,4-diamine (3b)1H NMR (400 MHz, DMSO-d6) δ 9.06 (s, 1H), 8.89 (s, 1H), 8.10 (s, 1H), 8.04 (s, 1H), 7.76 (s, 1H), 7.71 (d, J=8.6 Hz, 1H), 7.42 (d, J=8.8 Hz, 2H), 7.36 (d, J=8.6 Hz, 1H), 6.69 (d, J=8.8 Hz, 2H), 5.76 (s, 1H), 2.99 (s, 4H), 2.44 (s, 4H), 2.21 (s, 3H).13C NMR (100 MHz, DMSO-d6) δ 158.40, 156.64, 155.15, 146.47, 137.26, 133.78, 133.03, 120.79, 120.44, 120.34, 118.62, 116.20, 104.46, 104.00, 103.67, 55.17, 49.34, 46.27. N4-(1H-indazol-6-yl)-N2-(4-(4-methylpiperazine-1-yl) phenyl)-5-(trifluoromethyl)pyrimidine-2,4-diamine (3c), white Solid, yield: 16.5%.1H NMR (400 MHz, DMSO-d6) δ 9.70 (s, 1H), 8.63 (s, 1H), 8.33 (s, 1H), 7.92 (s, 1H), 7.72 (dd, J=5.6, 3.2 Hz, 1H), 7.68-7.65 (m, 1H), 7.48 (d, J=8.8 Hz, 1H), 7.29 (d, J=8.8 Hz, 3H), 7.01 (d, J=8.8 Hz, 2H), 3.18-3.14 (m, 4H), 2.51 (d, J=1.6 Hz, 4H), 2.24 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 166.94, 160.70, 155.18, 148.42, 140.43, 138.08, 133.18, 131.68, 131.47, 129.73, 128.62, 127.55, 126.84, 119.81, 118.54, 115.35, 99.13, 54.50, 48.25, 45.65; ESI-MS m/z: 469.2 (M+H)+. N4-(1H-indazol-6-yl)-5-methyl-N2-(4-(4-methylpiperazine-1-yl) p henyl)pyrimidine-2,4-diamine (3d), white solid, yield: 31.5%.1H NMR (500 MHz, DMSO-d6) δ 12.89 (s, 1H), 8.74 (s, 1H), 8.36 (s, 1H), 8.00 (s, 1H), 7.86 (s, 1H), 7.82 (s, 1H), 7.68 (d, J=9.0 Hz, 1H), 7.51 (d, J=9.0 Hz, 2H), 7.42 (d, J=9.0 Hz, 1H), 6.72 (d, J=9.0 Hz, 2H), 2.99 (d, J=4.0 Hz, 4H), 2.46 (d, J=4.5 Hz, 4H), 2.23 (s, 3H), 2.12 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.36, 158.45, 155.73, 145.35, 140.38, 138.06, 133.62, 133.23, 119.71, 119.18, 117.52, 115.93, 105.03, 102.29, 54.64, 48.98, 45.61, 13.49; ESI-MS m/z: 415.2 (M+H)+. N4-(1H-indazol-6-yl)-5-methoxy-N2-(4-(4-methylpiperazine-1-yl) phenyl)pyrimidine-2,4-diamine (3e)1H NMR (400 MHz, DMSO-d6) δ 8.77 (s, 1H), 8.65 (s, 1H), 7.96 (s, 2H), 7.83 (s, 1H), 7.65 (d, J=8.7 Hz, 1H), 7.55-7.53 (m, 3H), 6.79 (d, J=9.0 Hz, 2H), 3.86 (s, 3H), 3.01 (s, 4H), 2.45 (s, 4H), 2.22 (s, 3H).13C NMR (100 MHz, DMSO-d6) δ 154.60, 152.26, 145.77, 138.16, 137.40, 134.87, 134.48, 133.77, 120.40, 119.79, 119.69, 119.52, 117.10, 116.54, 101.56, 57.42, 55.25, 49.61, 46.27. Ethyl-4-((1H-indazol-6-yl)amino)-2-((4-(4-methylpiperazine-1-yl)phenyl)amino)pyrimidine-5-carboxylate (3f), yellow solid, yield: 95.1%.1H NMR (400 MHz, DMSO-d6) δ 13.05 (s, 1H), 10.34 (s, 1H), 9.86 (s, 1H), 8.73 (s, 1H), 8.08 (s, 1H), 7.76 (d, J=8.4 Hz, 1H), 7.53 (d, J=5.6 Hz, 2H), 7.29 (d, J=7.3 Hz, 1H), 6.81 (s, 2H), 4.34 (q, J=6.8 Hz, 2H), 3.17 (s, 4H), 2.75 (s, 4H), 2.43 (s, 3H), 1.37 (t, J=7.0 Hz, 3H).13C NMR (100 MHz, DMSO-d6) δ 166.39, 160.64, 160.44, 153.10, 146.23, 140.24, 136.11, 136.03, 133.47, 133.43, 131.57, 121.48, 120.57, 115.83, 60.36, 53.65, 47.55, 44.23, 14.16; ESI-MS m/z: 473.2 (M+H)+. N4-(1H-indazol-6-yl)-N2-(4-(4-methylpiperazine-1-yl) phenyl)-6-(trifluoromethyl)pyrimidine-2,4-diamine (3g), yellow solid, yield: 50.7%.1H NMR (500 MHz, DMSO-d6) δ 13.07 (s, 1H), 9.47 (s, 1H), 8.81 (s, 1H), 8.31 (s, 1H), 8.08 (s, 1H), 7.74 (d, J=8.5 Hz, 1H), 7.57 (s, 1H), 7.26 (s, 2H), 7.17 (s, 1H), 6.43 (s, 2H), 2.93 (s, 4H), 2.41 (s, 4H), 2.20 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 160.52, 157.77, 155.55, 155.51, 146.34, 140.18, 136.35, 133.20, 131.54, 126.00, 123.86, 120.76, 120.73, 120.52, 119.94, 115.31, 54.58, 48.62, 45.70; ESI-MS m/z: 469.2 (M+H)+. N4-(1H-indazol-6-yl) amino)-6-methyl-N2-(4-(4-methylpiperazine-1-yl)phenyl)pyrimidine-2,4-diamine (3h), yellow liquid, yield: 29.3%.1H NMR (500 MHz, DMSO-d6) δ 12.76 (s, 1H), 9.24 (s, 1H), 8.85 (s, 1H), 7.96 (s, 1H), 7.82 (s, 1H), 7.64 (d, J=8.5 Hz, 1H), 7.60 (d, J=8.5 Hz, 2H), 7.32 (d, J=8.5 Hz, 1H), 6.84 (d, J=8.0 Hz, 2H), 6.07 (s, 1H), 3.05 (s, 4H), 2.50 (s, 4H), 2.24 (s, 3H), 2.20 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 164.46, 161.29, 159.59, 146.55, 140.88, 139.50, 133.04, 132.11, 121.71, 120.21, 119.76, 117.86, 116.03, 115.10, 97.80, 54.45, 48.48, 45.62, 23.61; ESI-MS m/z: 415.2 (M+H)+. N4-(1H-indazol-6-yl) amino)-6-methyl-N2-(4-(4-methylpiperazine-1-yl)phenyl)thiophene[2,3-d]pyrimidine-2,4-diamine (3i),1H NMR (400 MHz, DMSO-d6) δ 9.51 (s, 1H), 8.86 (s, 1H), 8.05 (d, J=5.4 Hz, 1H), 8.03 (s, 1H), 7.93 (s, 1H), 7.71 (d, J=8.7 Hz, 1H), 7.62 (d, J=9.0 Hz, 2H), 7.54-7.49 (m, 1H), 7.18 (d, J=5.4 Hz, 1H), 6.80 (d, J=9.0 Hz, 2H), 3.03 (s, 4H), 2.44 (s, 4H), 2.20 (S, 3H).13C NMR (100 MHz, DMSO-d6) δ 162.66, 158.62, 155.98, 146.08, 140.80, 137.97, 134.18, 134.11, 133.81, 123.85, 120.58, 120.51, 120.40, 119.98, 117.83, 116.43, 107.67, 103.16, 55.24, 49.54, 46.29. Example 2: Chemical Synthesis of Compound 5a-5j The synthesis route was as follows: The general process was as follows: 2,4-dichloro-5-methylpyrimidine (1d) underwent a nucleophilic substitution reaction with various substituted amines to generate an intermediate 4a-4j. 4-(4-methylpiperazine)aniline and the obtained intermediate 4a-4j underwent a nucleophilic substitution reaction to generate a final product 5a-5j. Taking the synthesis of a compound 5a for example, a specific process was as follows: N, N-diisopropylethylamine (DIPEA) (516.96 mg, 4 mmol) was dissolved in DMF (4 mL) and cooled to 0° C. Then 5-aminoindole (266.3 mg, 2 mmol) dissolved in DMF (2 mL) was added dropwise to the mixed solution. The reaction mixture was stirred at 0° C. for about 1 hour. Next, the ice bath was removed, the reaction mixture was stirred at room temperature, and the reaction was monitored by TLC. After the reaction was completed, the resulting mixture was extracted with ethyl acetate (3×25 mL), washed with saturated salt solution, dried and concentrated by anhydrous Na2SO4, and an intermediate 4a was obtained by silica gel column chromatography. The intermediate 4a (258.7 mg, 1 mmol) and 4-(4-methylpiperazine)aniline (191.27 mg, 1 mmol) were dissolved in methanol (4 mL), then added with TFA (148.56 μl L, 2 mmol), heated to 80° C., and the reaction was monitored by TLC. After the reaction was completed, the mixture was cooled to room temperature, the resulting mixture was adjusted to be neutral with saturated sodium bicarbonate, extracted with ethyl acetate (3×25 mL), washed with saturated salt solution, dried and concentrated by anhydrous Na2SO4, and a final product 5a was obtained by silica gel column chromatography. A structure and characterization data of the obtained compound 5a-5k were as follows: N4-(1H-indazol-5-yl) amino)-5-methyl-N2-(4-(4-methylpiperazine-1-yl)phenyl)pyrimidine-2,4-diamine (5a), pink liquid, yield: 41.91%.1H NMR (500 MHz, DMSO-d6) δ 11.00 (s, 1H), 8.54 (s, 1H), 8.09 (s, 1H), 7.83 (s, 1H), 7.76 (s, 1H), 7.47 (d, J=9.0 Hz, 2H), 7.39-7.30 (m, 2H), 7.25 (d, J=8.5 Hz, 1H), 6.65 (d, J=9.0 Hz, 2H), 6.39 (s, 1H), 2.96 (d, J=4.5 Hz, 4H), 2.47-2.38 (m, 4H), 2.21 (s, 3H), 2.09 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.95, 158.57, 154.90, 145.17, 133.84, 133.01, 131.42, 127.55, 125.44, 119.65, 118.64, 115.98, 114.77, 110.70, 104.30, 101.05, 54.74, 49.16, 45.75, 13.52; ESI-MS m/z: 414.2 (M+H)+. N4-(1H-indazol-4-yl) amino)-5-methyl-N2-(4-(4-methylpiperazine-1-yl)phenyl)pyrimidine-2,4-diamine (5b), gray solid, yield: 74.49%.1H NMR (400 MHz, DMSO-d6) δ 11.15 (s, 1H), 8.60 (s, 1H), 8.08 (s, 1H), 7.85 (s, 1H), 7.43-7.35 (m, 3H), 7.31 (t, J=2.8 Hz, 1H), 7.26 (d, J=8.1 Hz, 1H), 7.12 (t, J=8.0 Hz, 1H), 6.65 (d, J=8.8 Hz, 2H), 6.49-6.33 (m, 1H), 3.02-2.96 (m, 4H), 2.50-2.44 (m, 4H), 2.25 (s, 3H), 2.19 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.94, 158.51, 155.20, 145.12, 136.81, 133.78, 131.53, 124.20, 123.06, 120.90, 119.42, 115.74, 114.12, 107.71, 104.64, 99.56, 54.67, 49.10, 45.67, 13.38; ESI-MS m/z: 414.2 (M+H)>. 5-methyl-N4-(1-methyl-1H-indazol-5-yl)-N2-(4-(4-methylpiperazi ne-1-yl)phenyl)pyrimidine-2,4-diamine (5c), gray solid, yield: 52.1%.1H NMR (400 MHz, DMSO-d6) δ 8.63 (s, 1H), 8.18 (s, 1H), 7.90 (d, J=2.0 Hz, 1H), 7.80 (s, 1H), 7.50 (d, J=9.2 Hz, 2H), 7.43 (d, J=8.8 Hz, 1H), 7.33 (dd, J=7.2, 2.8 Hz, 2H), 6.69 (d, J=9.2 Hz, 2H), 6.40 (d, J=2.8 Hz, 1H), 3.83 (s, 3H), 3.04-2.97 (m, 4H), 2.49-2.43 (m, 4H), 2.24 (s, 3H), 2.12 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.88, 158.55, 154.96, 145.17, 133.76, 133.55, 131.65, 129.71, 127.89, 119.73, 118. 59, 115. 82, 114. 93, 109. 00, 104. 29, 100. 22, 54. 73, 49. 02, 45. 75, 32. 51, 13. 52; ESI-MS m/z: 428.6 (M+H)+. N4-(1H-indazol-5-yl) amino)-5-methyl-N2-(4-(4-methylpiperazine-1-yl)phenyl)pyrimidine-2,4-diamine (5d), yellow oil, yield: 63.5%.1H NMR (500 MHz, DMSO-d6) δ 8.80 (s, 1H), 8.76 (d, J=3.5 Hz, 1H), 8.56 (s, 1H), 8.50 (s, 1H), 8.12 (d, J=8.0 Hz, 1H), 8.04 (dd, J=9.0, 2.0 Hz, 1H), 7.96-7.89 (m, 3H), 7.48 (d, J=8.5 Hz, 2H), 6.78 (d, J=9.0 Hz, 1H), 3.06-3.00 (m, 4H), 2.48-2.43 (m, 4H), 2.23 (s, 2H), 2.16 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 158.89, 158.64, 156.12, 148.42, 145.85, 144.42, 138.08, 135.19, 133.13, 128.64, 128.28, 125.73, 121.23, 120.85, 116.54, 115.85, 105.27, 54.73, 48.98, 45.76, 13.48; ESI-MS m/z: 414.3 (M+H)>. 5-methyl-N2-(4-(4-methylpiperazine-1-yl) phenyl)-N4-(benzopyraz ine-6-yl)-pyrimidine-2,4-diamine (5e), yellow oil, yield: 68.8%.1H NMR (500 MHz, DMSO-d6) δ 8.90 (s, 1H), 8.86 (s, 1H), 8.79 (d, J=1.5 Hz, 1H), 8.76 (s, 1H), 8.57 (s, 1H), 8.32 (dd, J=9.0, 2.0 Hz, 1H), 8.00 (d, J=9.0 Hz, 1H), 7.96 (s, 1H), 7.53 (d, J=8.5 Hz, 2H), 6.80 (d, J=8.5 Hz, 2H), 3.02 (s, 4H), 2.45 (s, 4H), 2.22 (s, 3H), 2.20 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 158.69, 158.50, 156.56, 145.72, 145.46, 143.27, 143.18, 141.62, 138.81, 133.13, 128.52, 126.27, 120.34, 116.74, 115.96, 105.74, 54.72, 49.02, 45.76, 13.56; ESI-MS m/z: 427.2 (M+H)+. 5-methyl-N2-(4-(4-methylpiperazine-1-yl) phenyl)-N4-(quinoxalin e-7-yl)-pyrimidine-2,4-diamine (5f), gray solid, yield: 44.6%.1H NMR (500 MHz, DMSO) δ 8.75 (dd, J=6.0, 2.5 Hz, 2H), 8.53 (s, 1H), 8.50 (s, 1H), 8.11 (d, J=7.5 Hz, 1H), 8.02 (d, J=9.0 Hz, 1H), 7.95-7.89 (m, 2H), 7.45 (t, J=7.0 Hz, 3H), 6.78 (d, J=8.5 Hz, 2H), 3.02 (d, J=4.0 Hz, 4H), 2.45 (d, J=4.5 Hz, 4H), 2.22 (s, 3H), 2.15 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 158.90, 158.62, 156.10, 148.45, 145.88, 144.29, 138.04, 135.31, 133.03, 128.55, 128.27, 125.76, 121.29, 120.98, 116.56, 115.86, 105.31, 54.64, 48.89, 45.67, 13.42; ESI-MS m/z: 426.2 (M+H)+. 5-methyl-N2-(4-(4-methylpiperazine-1-yl)phenyl)-N4-(5,6,7,8-te trahydronaphthalene-2-yl)-pyrimidine-2,4-diamine (5g), gray solid, yield: 71.2%.1H NMR (400 MHz, DMSO-d6) δ 8.62 (s, 1H), 7.96 (s, 1H), 7.79 (s, 1H), 7.37 (d, J=9.2 Hz, 2H), 7.23-7.14 (m, 2H), 7.08-7.01 (m, 1H), 6.64 (d, J=8.8 Hz, 2H), 3.03-2.95 (m, 4H), 2.83 (s, 2H), 2.61 (s, 2H), 2.51-2.47 (m, 4H), 2.26 (s, 3H), 2.10 (s, 3H), 1.69 (s, 4H).13C NMR (126 MHz, DMSO-d6) δ 160.32, 158.46, 154.96, 144.92, 137.86, 137.35, 134.04, 133.50, 126.21, 125.11, 124.56, 118.99, 115.78, 104.01, 54.60, 49.08, 45.58, 29.32, 24.52, 22.43, 22.36, 13.28; ESI-MS m/z: 429.3 (M+H)+. N4-(2,3-dihydro-1H-indene-5-yl)-5-methyl-N2-(4-(4-methylpipera zine-1-yl)phenyl)pyrimidine-2,4-diamine (5h), yellow oil, yield: 43.4%.1H NMR (400 MHz, DMSO) δ 8.70 (s, 1H), 8.12 (s, 1H), 7.83 (s, 1H), 7.65 (d, J=1.2 Hz, 1H), 7.51 (d, J=9.2 Hz, 2H), 7.38 (dd, J=8.0, 1.6 Hz, 1H), 7.18 (d, J=8.0 Hz, 1H), 6.79 (d, J=9.2 Hz, 2H), 3.07-3.01 (m, 4H), 2.87 (t, J=7.6 Hz, 4H), 2.50-2.45 (m, 4H), 2.24 (s, 3H), 2.10 (s, 3H), 2.05 (dd, J=14.8, 7.6 Hz, 2H).13C NMR (126 MHz, DMSO) δ 159.24, 158.51, 155.42, 145.43, 143.59, 138.01, 137.82, 133.56, 123.63, 120.23, 120.00, 118.48, 115.84, 104.63, 54.71, 49.08, 45.73, 32.58, 31. 81, 25.29, 13.48; ESI-MS m/z: 415.3 (M+H)+. 5-methyl-N4-(3-methyl-1H-indazol-6-yl)-N2-(4-(4-methylpiperazi ne-1-yl)phenyl)pyrimidine-2,4-diamine (5i), white solid, yield: 35.4%.1H NMR (500 MHz, DMSO-d6) δ 12.48 (s, 1H), 8.73 (s, 1H), 8.37 (s, 1H), 7.85 (s, 1H), 7.80 (s, 1H), 7.59 (d, J=8.0 Hz, 1H), 7.51 (d, J=7.5 Hz, 2H), 7.36 (d, J=8.0 Hz, 1H), 6.72 (d, J=8.0 Hz, 2H), 3.01 (s, 4H), 2.47 (s, 4H), 2.25 (s, 4H), 2.12 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.41, 158.41, 155.63, 145.20, 141.34, 138.09, 133.68, 119.71, 119.10, 118.55, 116.73, 115.95, 105.08, 102.37, 54.45, 48.77, 45.36, 13.52, 11.69; ESI-MS m/z: 429.3 (M+H)+. Tert-butyl (2-(6-((5-methyl-2-((4-(4-methylpiperazine-1-yl)phe nyl) amino)-pyrimidine-4-yl) amino)-2H-indazol-2-yl) ethyl) carbamate (5j), gray solid, yield: 12.7%.1H NMR (500 MHz, DMSO-d6) δ 8.75 (s, 1H), 8.21 (s, 1H), 8.14 (s, 2H), 7.84 (s, 1H), 7.61 (d, J=8.5 Hz, 1H), 7.55 (d, J=8.5 Hz, 1H), 7.28 (d, J=8.0 Hz, 1H), 7.05 (s, 1H), 6.79 (d, J=8.5 Hz, 2H), 4.41 (d, J=6.0 Hz, 2H), 3.49-3.45 (m, 2H), 3.02 (s, 4H), 2.45 (s, 4H), 2.21 (s, 3H), 2.12 (s, 3H), 1.37 (s, 9H).13C NMR (126 MHz, DMSO-d6) δ 159.25, 158.53, 155.58, 155.51, 148.76, 145.43, 137.09, 133.50, 123.78, 119.93, 119.78, 119.28, 118.05, 116.03, 107.06, 105.02, 77.95, 54.71, 52.05, 49.09, 45.73, 40.51, 28.17, 13.52; ESI-MS m/z: 558.3 (M+H)+. 5-methyl-N4-(1-methyl-1H-indazol-6-yl)-N2-(4-(4-methylpiperazi ne-1-yl)phenyl)pyrimidine-2,4-diamine (5k), gray solid, yield: 54.1%.1H NMR (400 MHz, DMSO-d6) δ 8.83 (s, 1H), 8.44 (s, 1H), 8.14 (s, 1H), 7.97 (s, 1H), 7.91 (s, 1H), 7.68 (s, 1H), 7.52 (s, 2H), 7.44 (s, 1H), 6.75 (s, 2H), 3.91 (s, 3H), 3.02 (s, 4H), 2.46 (s, 4H), 2.24 (s, 3H), 2.17 (s, 3H).13C NMR (100 MHz, DMSO-d6) δ 159.17, 158.41, 155.84, 145.55, 140.11, 138.42, 133.40, 132.08, 120.14, 119.46, 117.03, 115.78, 105.23, 100.67, 54.71, 48.97, 45.73, 35.20, 13.54; ESI-MS m/z: 429.2 (M+H)+. Example 3: Chemical Synthesis of Compound 6a-6m The synthesis route of the compound 6a-6m was as follows: The general synthesis route was as follows: 2,4-dichloro-5-methylpyrimidine (1d) reacted with 6-aminoindazole to produce an intermediate 2d, and the intermediate 2d and various substituted amines underwent a nucleophilic substitution reaction to generate a final product 6a-6m. Taking the synthesis of a compound 6a for example, a specific synthesis process was as follows: Compound 2d was prepared according to Example 1. Compound 2d (259.7 mg, 1 mmol) and 4-(4-methylpiperazine)aniline (191.27 mg, 1 mmol) were dissolved in 4 ml of methanol, then added with (TFA (148.56 v L, 2 mmol), heated to 80° C., and the reaction was monitored by TLC. After the reaction was completed, the mixture was cooled to room temperature, the resulting mixture was adjusted to be neutral with saturated sodium bicarbonate, extracted with ethyl acetate (3×25 mL), washed with saturated salt solution, dried and concentrated by anhydrous Na2SO4, and a final product 6a was obtained by silica gel column chromatography. A structure and characterization data of the compound 6a-6m were as follows: N4-(1H-indazol-6-yl)-5-methyl-N2-(4-morphinophenyl) pyrimidine-2,4-diamine (6a), yellow solid, yield: 33.9%.1H NMR (500 MHz, DMSO-d6) δ 12.89 (s, 1H), 8.72 (s, 1H), 8.35 (s, 1H), 8.00 (s, 1H), 7.86 (s, 1H), 7.82 (s, 1H), 7.67 (d, J=8.5 Hz, 1H), 7.50 (d, J=8.5 Hz, 2H), 7.41 (d, J=8.5 Hz, 1H), 6.70 (d, J=9.0 Hz, 2H), 2.89 (d, J=4.5 Hz, 4H), 2. 82 (d, J=4. 5 Hz, 4H), 2. 12 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.35, 158.46, 155.74, 146.12, 140.43, 138.06, 133.52, 133.18, 119.72, 119.17, 117.53, 115.92, 104.99, 102.32, 50.38, 45.62, 13.48; ESI-MS m/z: 402.2 (M+H)+. N4-(1H-indazol-6-yl)-5-methyl-N2-(4-(piperazine-1-yl) phenyl) py rimidine-2,4-diamine (6b), yellow solid, yield: 29.4%.1H NMR (500 MHz, DMSO-d6) δ 12.89 (s, 1H), 8.72 (s, 1H), 8.35 (s, 1H), 8.00 (s, 1H), 7.86 (s, 1H), 7.82 (s, 1H), 7.67 (d, J=8.5 Hz, 1H), 7.50 (d, J=8.5 Hz, 2H), 7.41 (d, J=8.5 Hz, 1H), 6.70 (d, J=9.0 Hz, 2H), 2.89 (d, J=4.5 Hz, 4H), 2.82 (d, J=4.5 Hz, 4H), 2.12 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.35, 158.46, 155.74, 146.12, 140.43, 138.06, 133.52, 133.18, 119.72, 119.17, 117.53, 115.92, 104.99, 102.32, 50.38, 45.62, 13.48; ESI-MS m/z: 402.2 (M+H)>. N2-(4-(4,4-difluoropiperazine-1-yl)phenyl)-N4-(1H-indazol-6-yl)-5-methylpyrimidine-2,4-diamine (6c), gray solid, yield: 61.1%.1H NMR (500 MHz, DMSO-d6) δ 12.98 (s, 1H), 8.79 (s, 1H), 8.44 (s, 1H), 7.99 (s, 1H), 7.92 (s, 1H), 7.86 (s, 1H), 7.66 (d, J=8.5 Hz, 1H), 7.55 (d, J=9.0 Hz, 2H), 7.44 (d, J=8.5 Hz, 1H), 6.79 (d, J=9.0 Hz, 2H), 3.19-3.10 (m, 4H), 2.14 (s, 3H), 2.07-1.99 (n, 4H).13C NMR (126 MHz, DMSO-d6) δ 159.38, 158.36, 155.70, 144.03, 140.58, 140.41, 138.04, 134.15, 133.18, 119.67, 119.14, 117.52, 116.94, 105.19, 102.39, 54.87, 46.85, 33.07, 13.54; ESI-MS m/z: 436.2 (M+H)+. 4-(4-((4-((1H-indazol-6-yl) amino)-5-methylpyrimidine-2-yl) ami no)phenyl)thiomorpholine 1,1-dioxide (6d), gray solid, yield: 81.4%.1H NMR (500 MHz, DMSO-d6) δ 13.00 (s, 1H), 8.83 (s, 1H), 8.45 (s, 1H), 8.01-7.84 (m, 3H), 7.67 (d, J=8.5 Hz, 1H), 7.57 (d, J=8.0 Hz, 2H), 7.43 (d, J=8.0 Hz, 1H), 6.82 (d, J=8.0 Hz, 2H), 3.62 (s, 4H), 3.11 (s, 4H), 2.14 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.39, 158.85, 158.29, 155.68, 142.01, 138.02, 134.28, 133.10, 119.84, 119.67, 119.11, 117.52, 116.69, 105.32, 102.49, 49.91, 47.86, 13.56; ESI-MS m/z: 450.2 (M+H)+. N4-(1H-indazol-6-yl)-N2-(2-methoxy-4-(4-methylpiperazine-1-yl) phenyl)-5-methylpyrimidine-2,4-diamine (6e), gray solid, yield: 26.6%.1H NMR (500 MHz, DMSO-d6) δ 12.92 (s, 1H), 8.45 (s, 1H), 7.98 (s, 1H), 7.91-7.81 (m, 3H), 7.64 (d, J=9.0 Hz, 1H), 7.41 (d, J=8.5 Hz, 1H), 7.26 (s, 1H), 6.61 (s, 1H), 6.31 (d, J=7.5 Hz, 1H), 3.81 (s, 3H), 3.07 (s, 4H), 2.48 (s, 4H), 2.24 (s, 3H), 2.13 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.34, 158.44, 155.70, 149.45, 146.79, 140.43, 138.05, 133.17, 121.81, 120.86, 119.72, 119.07, 117.28, 106.97, 105.48, 101.85, 100.14, 55.60, 54.71, 48.97, 45.74, 13.47; ESI-MS m/z: 445.3 (M+H)+. N4-(1H-indazol-6-yl)-N2-(3-methoxy-4-(4-methylpiperazine-1-yl) phenyl)-5-methylpyrimidine-2,4-diamine (6f), yellow solid, yield: 55.9%.1H NMR (500 MHz, DMSO-d6) δ 12.85 (s, 1H), 8.79 (s, 1H), 8.37 (s, 1H), 7.99 (s, 1H), 7.89 (s, 1H), 7.81 (s, 1H), 7.66 (d, J=8.5 Hz, 1H), 7.40 (d, J=8.5 Hz, 1H), 7.29 (s, 1H), 7. 24 (d, J=9. 5 Hz, 1H), 6. 63 (s, 1H), 3.43 (s, 3H), 2.86 (s, 4H), 2.47 (s, 4H), 2.24 (s, 3H), 2.13 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.28, 158.30, 155.70, 151.91, 140.40, 138.05, 136.65, 134.85, 133.21, 119.78, 119.18, 117.76, 117.43, 110.62, 105.47, 103.75, 102.14, 55.01, 54.84, 50.31, 45.84, 13.50; ESI-MS m/z: 445.2 (M+H)>. N4-(1H-indazol-6-yl)-5-methyl-N2-(4-(4-methylpiperazine-1-yl) p henyl)pyrimidine-2,4-diamine (6g), yellow solid, yield: 29.4%.1H NMR (500 MHz, DMSO-d6) δ 12.93 (s, 1H), 8.93 (s, 1H), 8.55 (s, 1H), 8.01 (s, 1H), 7.84 (d, J=18.5 Hz, 2H), 7.69 (d, J=8.5 Hz, 1H), 7.50 (d, J=9.0 Hz, 2H), 7.41 (d, J=8.0 Hz, 1H), 6.73 (d, J=8.5 Hz, 2H), 3.74-3.67 (m, 4H), 3.01-2.92 (m, 4H), 2.13 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.84, 156.85, 152.41, 146.01, 140.31, 137.43, 133.25, 132.61, 120.52, 119.89, 119.51, 117.78, 115.57, 105.57, 103.16, 66.11, 49.26, 13.41; ESI-MS m/z: 401.2 (M+H)+. N2-(4-(4-ethylpiperazine-1-yl)-2-methoxyphenyl)-N4-(1H-indazol-6-yl)-5-methylpyrimidine-2,4-diamine (6h), white solid, yield: 19.2%.1H NMR (500 MHz, DMSO-d6) δ 12.82 (s, 1H), 8.38 (s, 1H), 7.99 (s, 1H), 7.91-7.82 (m, 3H), 7.65 (d, J=8.5 Hz, 1H), 7.39 (d, J=9.5 Hz, 1H), 7.28 (s, 1H), 6.61 (d, J=2.0 Hz, 1H), 6.31 (d, J=10.0 Hz, 1H), 3.81 (s, 3H), 3.06 (s, 4H), 2.51 (s Hz, 4H), 2.38 (q, J=7.0 Hz, 2H), 2.12 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.34, 158.42, 155.69, 149.45, 146.81, 140.40, 138.03, 133.19, 121.81, 120.86, 119.71, 119.06, 117.28, 106.95, 105.47, 101.86, 100.16, 55.61, 52.37, 51.58, 49.06, 13.47, 11.92; ESI-MS m/z: 459.3 (M+H)+. N2-(4-(4-ethylpiperazine-1-yl)-3-methoxyphenyl)-N4-(1H-indazol-6-yl)-5-methylpyrimidine-2,4-diamine (6i), colorless oil, yield: 61.5%.1H NMR (400 MHz, DMSO-d6) δ 12.90 (s, 1H), 8.82 (s, 1H), 8.42 (s, 1H), 7.99 (s, 1H), 7.89 (s, 1H), 7.85 (s, 1H), 7.64 (d, J=8.4 Hz, 1H), 7.41 (d, J=8.8 Hz, 1H), 7.30 (s, 1H), 7.25 (d, J=8.8 Hz, 1H), 6.63 (d, J=8.8 Hz, 1H), 5.33 (s, 1H), 3.43 (s, 3H), 2.86 (s, 4H), 2.39-2.37 (m, 2H), 2.13 (s, 4H), 1.77 (s, 3H), 1.22 (t, J=6.8 Hz, 1H).13C NMR (126 MHz, DMSO-d6) δ 159.29, 158.27, 155.68, 151.90, 140.42, 138.07, 136.66, 134.84, 133.18, 119.74, 119.12, 117.71, 117.39, 110.60, 105.52, 103.74, 102.17, 52.63, 52.07, 51.67, 50.36, 13.53, 11.87; ESI-MS m/z: 459.3 (M+H)+. N4-(1H-indazol-6-yl)-5-methyl-N2-(4-(4-(4-methylpiperazine-1-y 1)piperidine-1-yl)phenyl)pyrimidine-2,4-diamine (6i), white solid, yield: 73.6%.1H NMR (500 MHz, DMSO-d6) δ 12.89 (s, 1H), 8.71 (s, 1H), 8.35 (s, 1H), 7.99 (s, 1H), 7.84 (d, J=12.5 Hz, 2H), 7.67 (d, J=8.7 Hz, 1H), 7.49 (d, J=8.9 Hz, 2H), 7.42 (d, J=8.6 Hz, 1H), 6.71 (d, J=8.9 Hz, 2H), 3.52 (d, J=12.0 Hz, 2H), 2.54 (s, 1H), 2.30 (s, 4H), 2.26-2.18 (m, 2H), 1.80 (d, J=11.6 Hz, 3H), 1.54-1.42 (n, 3H).13C NMR (126 MHz, DMSO-d6) δ 159.35, 158.46, 155.74, 145.54, 138.07, 133.40, 133.21, 119.72, 119.15, 117.51, 116.40, 105.01, 102.28, 83.88, 79.19, 60.85, 55.15, 49.26, 48.54, 45.74, 27.88, 13.51; ESI-MS m/z: 498.3 (M+H)+. N4-(1H-indazol-6-yl)-N2-(2-methoxy-4-(4-(4-methylpiperazine-1-yl)piperidine-1-yl)phenyl)-5-methylpyrimidine-2,4-diamine (6k). Yellow solid, 68.9% yield.1H NMR (500 MHz, DMSO-d6) δ 12.83 (s, 1H), 8.38 (s, 1H), 7.98 (s, 1H), 7.85 (d, J=7.0 Hz, 3H), 7.64 (d, J=8.5 Hz, 1H), 7.38 (d, J=8.0 Hz, 1H), 7.27 (s, 1H), 6.60 (s, 1H), 6.31 (d, J=7.0 Hz, 1H), 3.80 (s, 3H), 3.61 (d, J=12.0 Hz, 2H), 2.58 (t, J=11.5 Hz, 3H), 2.36-2.20 (m, 4H), 2.13 (d, J=9.0 Hz, 6H), 1.83 (d, J=11.3 Hz, 2H), 1.56-1.42 (m, 2H).13C NMR (126 MHz, DMSO-d6) δ 159.36, 158.47, 155.66, 150.46, 149.64, 146.99, 138.02, 133.30, 121.54, 121.21, 119.78, 119.04, 117.34, 107.47, 105.40, 101.95, 100.62, 60.84, 55.57, 54.98, 49.11, 48.46, 45.63, 27.76, 13.39; ESI-MS m/z: 528.3 (M+H)+. (4-((4-((1H-indazol-6-yl) amino)-5-methylpyrimidine-2-yl) amino) phenyl) (4-methylpiperazine-1-yl)methanone ((6m), colorless liquid, yield: 13.8%.1H NMR (400 MHz, DMSO-d6) δ 9.30 (s, 1H), 8.53 (s, 1H), 7.96 (s, 1H), 7.88 (s, 1H), 7.79 (s, 1H), 7.71 (d, J=8.8 Hz, 2H), 7.64 (d, J=8.4 Hz, 1H), 7.33 (d, J=8.8 Hz, 1H), 7.11 (d, J=8. 8 Hz, 2H), 2. 84 (s, 4H), 2. 44 (s, 4H), 2.10 (s, 3H), 1.70 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 169.22, 159.54, 157.86, 155.63, 142.65, 140.46, 137.79, 133.12, 127.81, 126.85, 119.73, 119.34, 117.88, 117.29, 106.37, 103.04, 54.55, 53.86, 45.59, 13.57; ESI-MS m/z: 473.2 (M+H)+. (4-((4-((1H-indazol-6-yl)amino)-5-methylpyrimidine-2-yl)amino)-N-(2-morpholinoethyl)benzamide (6n), white solid, yield: 60.2%.1H NMR (500 MHz, DMSO-d6) δ 13.02 (s, 1H), 9.33 (s, 1H), 8.59 (s, 1H), 8.24 (s, 1H), 8.15-7.88 (m, 3H), 7.79 (d, J=8.0 Hz, 2H), 7.71 (d, J=8.5 Hz, 1H), 7.63 (d, J=8.0 Hz, 2H), 7.46 (d, J=8.0 Hz, 1H), 3.57 (s, 4H), 2.43 (d, J=13.5 Hz, 6H), 2.17 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 165.92, 159.44, 157.76, 155.56, 143.90, 140.41, 137.84, 133.18, 127.69, 125.85, 119.69, 119.25, 117.63, 116.82, 106.65, 102.50, 66.14, 57.44, 53.26, 36.35, 13.64; ESI-MS m/z: 473.2 (M+H)+. Example 4: Kinase (PDGFRα, PDGFRβ, ABL1 and FLT3) Inhibition Test of Compounds The method adopted in the test was Caliper Mobility Shift Assay, which was a detection platform based on the mobility detection technology of microfluidic chip technology. Test steps: 1.25× kinase reaction buffer (62.5 mmol/L HEPES, pH 7.5; 0.001875% Brij-35; 12.5 mmol/L MgCl2; 2.5 mM DTT) and kinase reaction stop solution (100 mmol/L HEPES, pH 7.5; 0.015% Brij-35; 0.2% Coating Reagent #3) were configured. 10 μL of 2.5× kinase solution (adding kinase in 1.25× kinase reaction buffer) was added into 5 μL of compound solution with 5× concentration (dissolved in DMSO, diluted 10 times with water), incubated at room temperature for 10 minutes, then added with 10 μL of 2.5× substrate peptide solution (adding FAM labeled peptide and ATP in 1.25× kinase reaction buffer), reacted at 28° C. for a specific time, and then added with 25 μL of kinase reaction stop solution. Collected data was tested on Caliper to yield that inhibition ratio to kinase activity=(max−conversion)/(max−min)×100. “max” was DMSO control without adding compound, and “min” was low control. When IC50was determined, each sample was provided with 10 dilutions, each with 2 multiple holes, and repeated for 3 times. The results were shown in Table 1. TABLE 1Chemical structures of the synthesized compounds andinhibitory rates thereof (%) on PDGFR α, PDGFR β, ABL1 and FLT3kinases at a concentration of 100 nMSerialnumberCompoundPDGFR-αPDGFR-βABL1FLT313a87.194.490.487.523b93.795.895.190.233c62.381.084.278.643d94.299.695.290.553e32.955.054.351.263f1.33.54.334.873g6.07.26.914.683h3.221.25.913.893i6.630.723.642.6105a57.378.658.960.3115b66.190.378.860.9125c67.779.258.965.4135d59.184.180.980.4145e57.874.165.569.8155f82.591.090.290.5165h71.888.980.287.1175g78.390.489.285.7185j60.671.171.674.3195k91.196.490.488.7205i94.598.085.293.4216a84.493.898.287.2226b55.068.791.287.3236c48.948.550.674.6246d66.670.359.678.4256e64.863.478.879.5266f94.497.098.097.8276g87.793.889.687.6286h69.380.287.582.6296i97.599.895.398.9306j82.585.487.691.0316k65.076.447.558.4326m66.878.481.085.6336n80.582.481.282.334Pazopanib96.976.775.380.6 TABLE 2IC50values of some active compounds inhibiting PDGFR αand PDGFR β kinasesSerialCompoundIC50(nM)numbernumberPDGFR-αPDGFR-β15k197.325i157.936a174.246b5568.756c8110366d66.670.376e547286f7.42.696g198.8106h7440116i2.71.7126j1512136n2019 TABLE 3IC50values of some compounds inhibiting related kinasesCom-Serialpoundnum-num-IC50(nM)berberFGFR2PDGFR-αPDGFR-βALKJAK1IKK β16d11746391869961926e>100054727.0199>100036j8915122314>100046c5408110347672>100056g81198.8261359566n1022019956.018176i272.71.75.07.769486f617.42.61131>100096h>100074405.8320>1000 The results showed that 10 active compounds could effectively inhibit the PDGFRα and PDGFRβ kinases, but have relatively weak inhibitory activity on other kinases, indicating good kinase selectivity. In particular, the compounds 6g, 6i and 6f had significant inhibitory activities on PDGFRα and PDGFRβ wherein the compound 6i had the strongest inhibitory activity. Example 5: Kinase Selectivity Test of Active Compounds According to the above-mentioned method for measuring kinase inhibitory activity, the inhibitory activity of the active compounds on other multiple kinases was measured to characterize the kinase selectivity of active inhibitors. TABLE 4Selective inhibitory activity of the active compound6i on 16 kinases at a concentration of 100 nMCompounds % inhibition @ 100 nM6i (% inhibition @ 100 nM)%% inhibitionAverageKinaseinhibition 12% inhibitionSDEGFR40.943.642.31.9EGFR73.475.774.51.6L858R/T790M/C797SEGFR T790M57.056.856.90.1FLT178.780.779.71.5JAK191.789.390.51.7FGFR38.113.710.94.0KDR71.571.371.40.2EGFR/T790M/76.677.977.20.9L858REGFR L858R61.762.662.10.6FGFR165.066.665.81.1BTK49.151.350.21.6FGFR283.8582.1183.01.2PDGFR α98.91103.00101.02.9PDGFR β100.98102.53101.81.1SRC90.9893.4792.21.8 The results showed that the active compound 6i could effectively inhibit the PDGFRα and PDGFRβ kinases at the concentration of 100 nM, wherein the inhibition rate could be over 100%, but had relatively weak inhibitory activity on other kinases, indicating good kinase selectivity. Example 6 Inhibition Test of Active Compounds on Osteosarcoma Cells Logarithmic osteosarcoma cells were inoculated into a 96-well plate with a cell concentration of 1,500 cell/well for 6 hours by using MTT assay, and each well was filled with 200 μL of cell suspension. Samples were prepared into solutions with 5 concentration gradients of 0.5, 2.5, 5, 10 and 25 μmol/L, and each sample was equipped with 5 multiple holes. The samples were cultured in an incubator for 48 hours (37° C., 5% CO2), and 20 v L of MTT (3-(4,5-dimethylthiazole-2)-2,5-diphenyltetrazole) were added into each hole. After continuous culture for 4 hours, the culture solution was discarded by suction, and 200 v L of DMSO were added to each well. After shaking and dissolving for 10 minutes, OD values of each well were measured at 490 nM with a multifunctional microplate reader and the inhibition rate was calculated. Calculation method of IC50value: the IC50values of the samples were calculated by curve fitting with GraphPad Prism software. TABLE 5Inhibitory activity (IC50) of active compounds on fourosteosarcoma cells determined by MTT assayMG-63*U2OSSAOS-2MNNG/HOSPazo->100 μM>100 μM>100 μM8.52 ± 0.868panib#μMAxitinib#>100 μM>100 μM35.14 ± 0.2537.47 ± 0.202μMμMImatinib#31.69 ± 0.70327.15 ± 0.60515.01 ± 0.06420.5 ± 0.808μMμMμMμM6f0.841 ± 0.0050.756 ± 0.0010.72 ± 0.0251.36 ± 0.019μMμMμMμM6g0.958 ± 0.0070.552 ± 0.00230.95 ± 0.0011.4 ± 0.003μMμMμM6i0.438 ± 0.0010.418 ± 0.0170.37 ± 0.0021.03 ± 0.08μMμMμM The results showed that three active compounds had strong inhibitory activities on the proliferation of four osteosarcoma cell lines compared with three positive drugs, indicating that these compounds had excellent anti-osteosarcoma effects. Example 7: Adhesion and Metastasis Test of Active Compound 6i Inhibiting Osteosarcoma Cells Human Plasma Fibronectin was pre-incubated in a 96-well plate, and inoculated with osteosarcoma cells subjected to the effects of 0, 0.1, 0.2 and 0.4 μM of 6i, 0.4 μM of Pazopanib, and 0.4 μM of Imatinib for 48 hours, with 5×104cells per well, and each group was provided with 6 multiple wells, and cultured in 5% CO2at 37° C. for 40 minutes, washed with PBS to remove non-adhered cells, fixed with 4% paraformaldehyde at room temperature for 15 minutes and 100 μL, washed with 200 μL of PBS for three times, then the PBS was discarded, and each well was dyed with 50 μL of crystal violet for 5 minutes at room temperature, washed with 200 μL of ultrapure water for three times after dyeing, then the ultrapure water was discarded. Each well was dried, and added with 100 μL of 33% glacial acetic acid and the crystal violet was dissolved by shaking for 10 minutes. The absorbance was measured by a microplate reader at 570 nM, and the adhesion rate (%)=(OD value of tumor cells in the drug-added treated group−Hu FN OD value)/(OD value of untreated tumor cells−Hu FN OD value)×100%. Osteosarcoma cells with a density of 5×104cells (treated with 0, 0.1, 0.2 and 0.4 μM of 6i, 0.4 of μM Pazopanib, and 0.4 μM of Imatinib) after 48 hours were inoculated into a 6-well plate, RMPL 1640 culture medium containing 10% serum was added and placed in an incubator to make the cells adhere to the wall to form monolayer cells, and a “+” cross scratch was made with a 200 μL pipette tip. The cells were washed with sterile PBS for three times, and basal medium containing 2.5 μL of TCS was added respectively. Photographs were taken at 0 hour, 24 hours and 48 hours under an inverted microscope, and migration distances were measured with Imag J software. The results were shown inFIG.1. The results showed that 0.1, 0.2 and 0.4 uM of active compound 6i had strong dose-dependent inhibitory activity on the adhesion (4A) and metastasis (4B) of osteosarcoma cells MG63 and MNNG compared with the positive drugs. Example 8: Inhibitory Activity of Active Compound 6i on Two Osteosarcoma Cells PDGFR Signaling Pathway Related Proteins After pretreating the osteosarcoma cell line with the compound for 48 hours, the culture medium was sucked off, washed with PBS for 3 times, and the total protein was extracted after lysis. A phosphorylation level of the PDGFR signaling pathway related proteins was detected by Western Blot. The results were shown inFIG.2. The results showed that 0.1, 0.2 and 0.4 uM of active compound 6i had strong dose-dependent inhibition of phosphorylation of osteosarcoma cells MG63 and MNNG related signaling pathway proteins compared with positive drugs. Example 9: Inhibitory Activity Test of Active Compounds on Two Human Neovascular Cells The same MTT assay as in Example 6 was used to determine the inhibitory activity of the active compound 6i on two retinal cells. TABLE 6Inhibitory activity (IC50, uM) of active compounds onthree human retinal cells determined by MTT assayCompoundsARPE-19HCMEC-D3HBVP6i17.48 ± 0.370.23 ± 0.0020.55 ± 0.098Sorafenib23.29 ± 0.1436.32 ± 0.739.92 ± 0.598Vorolanib56.23 ± 3.2334.32 ± 0.88518.5 ± 10.520Imatinib40.33 ± 1.66244.89 ± 1.36615.15 ± 1.088 The results showed that the active compound 6i had strong inhibitory activity on human immortalized human brain microvascular endothelial cells (HCMEC/D3) and human cerebrovascular pericytes (HBVP) compared with multiple positive drugs, but had almost no inhibitory activity on human normal retinal cells ARPE-19 at low concentration, which indicated that the active compound 6i could effectively inhibit fundus vascular proliferation, thus alleviating ophthalmic diseases wet age-related macular degeneration or uveitis, and had low toxicity.
34,864
11858915
DETAILED DESCRIPTION OF THE INVENTION As used in the specification and appended claims, unless specified to the contrary, the following terms have the meaning indicated below. As used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “an agent” includes a plurality of such agents, and reference to “the cell” includes reference to one or more cells (or to a plurality of cells) and equivalents thereof known to those skilled in the art, and so forth. When ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and sub-combinations of ranges and specific embodiments therein are intended to be included. The term “about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range, in some instances, will vary between 1% and 15% of the stated number or numerical range. The term “comprising” (and related terms such as “comprise” or “comprises” or “having” or “including”) is not intended to exclude that in other certain embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, described herein, “consist of” or “consist essentially of” the described features. “Administering” when used in conjunction with a therapeutic means to administer a therapeutic systemically or locally, as directly into or onto a target tissue, or to administer a therapeutic to a subject whereby the therapeutic positively impacts the tissue to which it is targeted. Thus, as used herein, the term “administering”, when used in conjunction with a composition described herein, can include, but is not limited to, providing a composition into or onto the target tissue; providing a composition systemically to a subject by, e.g., oral administration whereby the therapeutic reaches the target tissue or cells. “Administering” a composition may be accomplished by injection, topical administration, and oral administration or by other methods alone or in combination with other known techniques. The term “C2-C6alkenyl” as used herein, means an alkyl moiety comprising 2 to 6 carbon atoms having at least one carbon-carbon double bond. The carbon-carbon double bond in such a group may be anywhere along the 2 to 6 carbon atom chain that will result in a stable compound. Examples of such groups include, but are not limited to, ethenyl, propenyl, butenyl, allyl, and pentenyl. The alkenyl may be in either the cis or trans conformation about the double bond(s), and should be understood to include both isomers. Examples of alkenyls include, but are not limited to ethenyl (—CH═CH2), 1-propenyl (—CH2CH═CH2), isopropenyl [—C(CH3)═CH2], butenyl, 1,3-butadienyl and the like. Whenever it appears herein, a numerical range such as “C2-C6alkenyl” means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms, although the present definition also covers the occurrence of the term “alkenyl” where no numerical range is designated. In some embodiments, the alkenyl is a C2-C10alkenyl, a C2-C9alkenyl, a C2-C8alkenyl, a C2-C7alkenyl, a C2-C6alkenyl, a C2-C5alkenyl, a C2-C4alkenyl, a C2-C3alkenyl, or a C2alkenyl. The term “C1-C6alkyl;” as used herein, refers to a straight or branched chain hydrocarbon monoradical, which may be fully saturated or unsaturated, having from one to about ten carbon atoms, or from one to six carbon atoms. Examples of saturated hydrocarbon monoradical include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, neopentyl, tert-amyl and hexyl, and longer alkyl groups, such as heptyl, octyl, and the like. Whenever it appears herein, a numerical range such as “C1-C6alkyl” means that the alkyl group consists of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated. The term “C2-C6alkynyl,” as used herein, means an alkyl moiety comprising from 2 to 6 carbon atoms and having at least one carbon-carbon triple bond. The carbon-carbon triple bond in such a group may be anywhere along the 2 to 6 carbon chain that will result in a stable compound. Examples of such groups include, but are not limited to, ethyne, propyne, 1-butyne, 2-butyne, 1-pentyne, 2-pentyne, 1-hexyne, 2-hexyne, and 3-hexyne, ethynyl, 2-propynyl, 2-butynyl, 1,3-butadiynyl and the like. Whenever it appears herein, a numerical range such as “C2-C6alkynyl” means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms, although the present definition also covers the occurrence of the term “alkynyl” where no numerical range is designated. The term “C6-C10aryl,” as used herein, refers to a radical derived from a hydrocarbon ring system comprising hydrogen, 6 to 10 carbon atoms and at least one aromatic ring. The aryl radical may be a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include fused (when fused with a cycloalkyl or heterocycloalkyl ring, the aryl is bonded through an aromatic ring atom) or bridged ring systems. In some embodiments, the aryl is a 6- to 10-membered aryl. In some embodiments, the aryl is a 6-membered aryl. Aryl radicals include, but are not limited to, aryl radicals derived from the hydrocarbon ring systems of anthrylene, naphthylene, phenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. In some embodiments, the aryl is phenyl. The term “C1-C6aminoalkyl,” as used herein, refers to a C1-C6alkyl radical, as defined above, that is substituted with one or more amino groups. The amino groups in such C1-C6aminoalkyl groups may be unsubstituted, mono-substituted, or disubstituted. Examples of C1-C6aminoalkyl groups include, but are not limited to, —CH2NH2, —CH2N(H)CH3, —CH2N(CH3)2, and the like. The term “C3-C10cycloalkyl” refers to a partially or fully saturated, monocyclic, or polycyclic carbocyclic ring comprising from 3 to 10 carbon atoms, which may include fused (when fused with an aryl or a heteroaryl ring, the cycloalkyl is bonded through a non-aromatic ring atom) or bridged ring systems. Representative cycloalkyls include. In some embodiments, the cycloalkyl is a 3- to 6-membered cycloalkyl. In some embodiments, the cycloalkyl is a 5- to 6-membered cycloalkyl. Monocyclic cycloalkyls include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic cycloalkyls or carbocycles include, for example, adamantyl, norbornyl, decalinyl, bicyclo[3.3.0]octane, bicyclo[4.3.0]nonane, cis-decalin, trans-decalin, bicyclo[2.1.1]hexane, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane, bicyclo[3.2.2]nonane, and bicyclo[3.3.2]decane, and 7,7-dimethyl-bicyclo[2.2.1]heptanyl. Partially saturated cycloalkyls include, for example cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl The term “C1-C6deuteroalkyl.” as used herein, means a C1-C6alkyl group as defined herein wherein one or more hydrogen atoms in the C1-C6alkyl group is replaced with a deuterium atom. The term “C1-C6haloalkyl,” as used herein, refers to a C1-C6alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, fluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. The term “C1-C6hydroxyalkyl,” as used herein, refers to a C1-C6alkyl radical, as defined above, that is substituted with one or more hydroxy groups. The term “animal” as used herein includes, but is not limited to, humans and non-human vertebrates such as wild, domestic and farm animals. As used herein, the terms “subject,” “subject” and “individual” are intended to include living organisms in which certain conditions as described herein can occur. Examples include humans, monkeys, cows, sheep, goats, dogs, cats, mice, rats, and transgenic species thereof. In a preferred embodiment, the subject is a primate. In certain embodiments, the primate or subject is a human. In certain instances, the human is an adult. In certain instances, the human is child. In further instances, the human is under the age of 12 years. In certain instances, the human is elderly. In other instances, the human is 60 years of age or older. Other examples of subjects include experimental animals such as mice, rats, dogs, cats, goats, sheep, pigs, and cows. The experimental animal can be an animal model for a disorder, e.g., a transgenic mouse with hypertensive pathology. The term “Aurora kinase A,” or “AurA,” as used herein, means the human protein known to those of ordinary skill in the art as Aurora kinase A, and that is encoded by the AURKA gene. The term “Aurora kinase B,” or “AurB,” as used herein, means the human protein known to those of ordinary skill in the art as Aurora kinase B, and that is encoded by the AURKB gene. A “cyano” group refers to a —CN group. The term “halo” or “halogen,” as used herein, refers to bromo, chloro, fluoro or iodo. In some embodiments, halogen is fluoro or chloro. In some embodiments, halogen is fluoro. The term “heterocycloalkyl,” as used herein, refers to a 3- to 24-membered partially or fully saturated ring radical comprising 2 to 23 carbon atoms and from one to 8 heteroatoms selected from boron, nitrogen, oxygen, phosphorous, and sulfur. Unless stated otherwise specifically in the specification, the heterocycloalkyl radical may be a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include fused (when fused with an aryl or a heteroaryl ring, the heterocycloalkyl is bonded through a non-aromatic ring atom) or bridged ring systems; and the nitrogen, carbon, or sulfur atoms in the heterocycloalkyl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized. In some embodiments, the heterocycloalkyl is a 3- to 6-membered heterocycloalkyl. In some embodiments, the heterocycloalkyl is a 5- to 6-membered heterocycloalkyl. Examples of such heterocycloalkyl radicals include, but are not limited to, aziridinyl, azetidinyl, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, 1,1-dioxo-thiomorpholinyl, 1,3-dihydroisobenrofuran-1-yl, 3-oxo-1,3-dihydroisobenzofuran-1-yl, methyl-2-oxo-1,3-dioxol-4-yl, and 2-oxo-1,3-dioxol-4-yl. The term heterocycloalkyl also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides, and the oligosaccharides. It is understood that when referring to the number of carbon atoms in a heterocycloalkyl, the number of carbon atoms in the heterocycloalkyl is not the same as the total number of atoms (including the heteroatoms) that make up the heterocycloalkyl (i.e. skeletal atoms of the heterocycloalkyl ring). The term “C1-C6heteroalkyl,” as used herein, means an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g., boron, oxygen, nitrogen (e.g. —NH—, —N(alkyl)-), sulfur, or combinations thereof. A heteroalkyl is attached to the rest of the molecule at a carbon atom of the heteroalkyl. In one aspect, a heteroalkyl is a C1-C6heteroalkyl wherein the heteroalkyl is comprised of 1 to 6 carbon atoms and one or more atoms other than carbon, e.g., oxygen, nitrogen (e.g. —NH—, —N(alkyl)-), sulfur, or combinations thereof wherein the heteroalkyl is attached to the rest of the molecule at a carbon atom of the heteroalkyl. The term “heteroaryl,” as used herein refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from boron, nitrogen, oxygen, phosphorous, and sulfur, and at least one aromatic ring. The heteroaryl radical may be a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include fused (when fused with a cycloalkyl or heterocycloalkyl ring, the heteroaryl is bonded through an aromatic ring atom) or bridged ring systems; and the nitrogen, carbon, or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized. In some embodiments, the heteroaryl is a 5- to 10-membered heteroaryl. In some embodiments, the heteroaryl is a 5- to 6-membered heteroaryl. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridinyl, 1-oxidopyrimidinyl, I-oxidopyrazinyl, 1-oxidopyridazinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e., thienyl). By “pharmaceutically acceptable,” as used herein, is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. The term “pharmaceutical composition” means a composition comprising at least one active ingredient, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human). Those of ordinary skill in the art will understand and appreciate the techniques appropriate for determining whether an active ingredient has a desired efficacious outcome based upon the needs of the artisan. The term “pharmaceutically acceptable salt,” as used herein, means a salt of a compound of the present invention that retains the biological effectiveness of the free acids and bases of the specified derivative and that is not biologically or otherwise undesirable. The term “PLK4,” as used herein, means the human protein known to those of ordinary skill in the art as polo-like kinase 4, and that is encoded by the PLK4 gene The term “oxo,” as used herein, refers to a carbonyl moiety such that alkyl substituted by oxo refers to a ketone group. The term “solvate,” as used herein, means a molecular complex between compounds of the present invention and solvent molecules. Examples of solvates include, but are not limited to, compounds of the invention in combination water, isopropanol, ethanol, methanol, dimethylsulfoxide (DMSO), ethyl acetate, acetic acid, ethanolamine, or mixtures thereof. The term “hydrate” can be used when said solvent is water. It is specifically contemplated that in the present invention one solvent molecule can be associated with one molecule of the compounds of the present invention, such as a hydrate. Furthermore, it is specifically contemplated that in the present invention, more than one solvent molecule may be associated with one molecule of the compounds of the present invention, such as a dihydrate. Additionally, it is specifically contemplated that in the present invention less than one solvent molecule may be associated with one molecule of the compounds of the present invention, such as a hemihydrate. Furthermore, solvates of the present invention are contemplated as solvates of compounds of the present invention that retain the biological effectiveness of the non-hydrate form of the compounds. Where a compound of the invention contains an alkenyl group, geometric cis/trans (or Z/E) isomers are possible. Where the compound contains, for example, a keto or oxime group or an aromatic moiety, tautomeric isomerism (‘tautomerism’) can occur. Examples of tautomerism include keto and enol tautomers. A single compound may exhibit more than one type of isomerism. Included within the scope of the invention are all stereoisomers, geometric isomers, and tautomeric forms of the inventive compounds, including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallization. The term “stereoisomers” refers to compounds that have identical chemical constitution, but differ with regard to the arrangement of their atoms or groups in space. In particular, the term “enantiomers” refers to two stereoisomers of a compound that are non-superimposable mirror images of one another. The terms “racemic” or “racemic mixture,” as used herein, refer to a 1:1 mixture of enantiomers of a particular compound. A mixture of racemates in which one racemate is present in a greater amount than the other racemate in such mixture may be described as “enantiomerically enriched.” The term “diastereomers”, on the other hand, refers to the relationship between a pair of stereoisomers that comprise two or more asymmetric centers and are not mirror images of one another. Designations that are conventional in the art may be used to describe stereoisomers of compounds, or the stereochemistry of a particular asymmetric carbon atom, of the compounds disclosed herein, or mixtures thereof. For example, a single racemate or stereocenter of a compound, may be described as of the (+), the (−), the (R)-, or the (S) configuration. A mixture of racemates may be described by use of the (±) symbol. The compounds of the present invention may have asymmetric carbon atoms. The carbon-carbon bonds of the compounds of the present invention may be depicted herein using a solid line (), a solid wedge () or a dotted wedge (). The use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers (e.g. specific enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of either a solid or dotted wedge to depict bonds to asymmetric carbon atoms is meant to indicate that only the stereoisomer shown is meant to be included. It is possible that compounds of the invention may contain more than one asymmetric carbon atom. In those compounds, the use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers are meant to be included. For example, unless stated otherwise, it is intended that the compounds of the present invention can exist as enantiomers and diastereomers or as racemates and mixtures thereof. The use of a solid line to depict bonds to one or more asymmetric carbon atoms in a compound of the invention and the use of a solid or dotted wedge to depict bonds to other asymmetric carbon atoms in the same compound is meant to indicate that a mixture of diastereomers is present. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate using, for example, chiral high pressure liquid chromatography (HPLC). Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound contains an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to one skilled in the art. Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% isopropanol, typically from 2 to 20%, and from 0 to 5% of an alkylamine, typically 0.1% diethylamine. Concentration of the eluate affords the enriched mixture. Stereoisomeric conglomerates may be separated by conventional techniques known to those skilled in the art. See, e.g. “Stereochemistry of Organic Compounds” by E L Eliel (Wiley, New York, 1994), the disclosure of which is incorporated herein by reference in its entirety. The term “substituted,” as used herein, means that the specified group or moiety bears one or more substituents. The term “unsubstituted” means that the specified group bears no substituents. The term “optionally substituted” means that the specified group is unsubstituted or substituted by one or more substituents. It is to be understood that in the compounds of the present invention when a group is said to be “unsubstituted,” or is “substituted” with fewer groups than would fill the valencies of all the atoms in the compound, the remaining valencies on such a group are filled by hydrogen. For example, if a C6aryl group, also called “phenyl” herein, is substituted with one additional substituent, one of ordinary skill in the art would understand that such a group has 4 open positions left on carbon atoms of the C6aryl group (6 initial positions, minus one to which the remainder of the compound of the present invention is bonded, minus an additional substituent, to leave 4). In such cases, the remaining 4 carbon atoms are each bound to one hydrogen atom to fill their valencies. Similarly, if a C6aryl group in the present compounds is said to be “disubstituted,” one of ordinary skill in the art would understand it to mean that the C6aryl group has 3 carbon atoms remaining that are unsubstituted. Those three unsubstituted carbon atoms are each bound to one hydrogen atom to fill their valencies. In accordance with a convention used in the art, the symbol is used in structural formulas herein to depict the bond that is the point of attachment of the moiety or substituent to the core or backbone structure. In accordance with another convention, in some structural formulae herein the carbon atoms and their bound hydrogen atoms are not explicitly depicted, e.g., represents a methyl group, represents an ethyl group, and represents a cyclopentyl group, etc. if a group, as for example, (R1)nis depicted as “floating” Ring A in the formula: then, unless otherwise defined, the substituent R1may reside on any atom of the ring system, assuming replacement of a depicted, implied, or expressly defined hydrogen from one of the ring atoms, so long as a stable structure is formed. A ring system A may be, for example, but not limited to aryl, heteroaryl, cycloalkyl, cycloheteroalkyl, spirocyclyl or a fused ring system. If a group “R” is depicted as “floating” on a ring system A as shown above, and Ring A contains saturated carbons, then “n” can be more than one, assuming each replaces a currently depicted, implied, or expressly defined hydrogen the ring A; then, unless otherwise defined, where the resulting structure is stable, two R1groups may reside on the same carbon. For example, when R1is a methyl group, there can exist a germinal dimethyl on a carbon of the ring A. In another example, two R1groups on the same carbon, including that carbon, may form a ring, thus creating a spirocyclic ring (a “spirocyclyl group”). It is to be understood that in the compounds of Formulae (I), (Ia), (Ib), (II), and (III) if n is less than the number of substitutable atoms on Ring A, the other substitutable positions on Ring A are bonded to a hydrogen atom. As used herein, the term “therapeutic” means an agent utilized to treat, combat, ameliorate, prevent, or improve an unwanted condition or disease of a subject. A “therapeutically effective amount” or “effective amount” as used herein refers to the amount of active compound or pharmaceutical agent that elicits a biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes one or more of the following: (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease, (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology), and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology). The terms “treat,” “treated,” “treatment,” or “treating” as used herein refers to both therapeutic treatment in some embodiments and prophylactic or preventative measures in other embodiments, wherein the object is to prevent or slow (lessen) an undesired physiological condition, disorder, or disease, or to obtain beneficial or desired clinical results. For the purposes described herein, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of the condition, disorder or disease; stabilization (i.e., not worsening) of the state of the condition, disorder or disease; delay in onset or slowing of the progression of the condition, disorder or disease, amelioration of the condition, disorder or disease state; and remission (whether partial or total), whether detectable or undetectable, or enhancement or improvement of the condition, disorder or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment. A prophylactic benefit of treatment includes prevention of a condition, retarding the progress of a condition, stabilization of a condition, or decreasing the likelihood of occurrence of a condition. As used herein, “treat,” “treated,” “treatment,” or “treating” includes prophylaxis in some embodiments. The term “TRIM37,” as used herein, means the human protein known those of ordinary skill in the art as tripartite motif-containing protein 37, an E3 ubiquitin ligase that is encoded by the TRIM37 gene. The term “CFI-400495” means the compound having the Chemical Abstract Service Registry No. 1338806-73-7, and the structure shown below. The preparation of the compound is described in PCT Application Publication No. WO 2011/123946 and is commercially available for purchase. PLK4 Inhibitor Compounds Provided herein are compounds of Formula (I), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl, heteroaryl, C3-C10cycloalkyl, or heterocycloalkyl;each Rais independently deuterium, halogen, —CN, oxo, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, —P(O)(Ra)2, —P(O)2(Ra)2, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C10alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;or two Raon adjacent atoms are taken together to form a C3-C10cycloalkyl or heterocycloalkyl; each optionally substituted with one or more R1b;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1aon the same atom are taken together to form an oxo;each R1bis independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1bon the same atom are taken together to form an oxo;n is 0, 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;R3is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;each of R4a, R4b, and R4cis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —NRcRd, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R5is hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each R6is independently hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R7is hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, or C1-C6aminoalkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O), NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6alkoxy, C1-C6aminoalkyl, C1-C6alkylamino, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3), —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)?N(CH3)2, —NH2, —NHCH3, —N(CH3), —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C3-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Provided herein are compounds of Formula (I), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl, heteroaryl, C3-C10cycloalkyl, or heterocycloalkyl;each R1is independently deuterium, halogen, —CN, oxo, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C10alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;or two R1on adjacent atoms are taken together to form a C3-C10cycloalkyl or heterocycloalkyl; each optionally substituted with one or more R1b;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1aon the same atom are taken together to form an oxo;each R1bis independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1bon the same atom are taken together to form an oxo;n is 0, 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;R3is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;each of R4a, R4b, and R4cis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —NRcRd, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R5is hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each R6is independently hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R7is hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, or C1-C6aminoalkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)?N(CH3)2, —NH2, —NHCH3, —N(CH3), —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C3-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. In other embodiments are provided compounds of Formula (Ia), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl, heteroaryl, C3-C10cycloalkyl, or heterocycloalkyl;each Rais independently deuterium, halogen, —CN, oxo, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C1-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C1-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;or two R1on adjacent atoms are taken together to form a C3-C10cycloalkyl or heterocycloalkyl; each optionally substituted with one or more R1b;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1aon the same atom are taken together to form an oxo;each R1bis independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1bon the same atom are taken together to form an oxo;n is 0, 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;R3is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;each of R4a, R4b, and R4cis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —NRcRd, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R5is hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each R6is independently hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R7is hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, or C1-C6aminoalkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. In other embodiments are provided compounds of Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl, heteroaryl, C3-C10cycloalkyl, or heterocycloalkyl;each R1is independently deuterium, halogen, —CN, oxo, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;or two R1on adjacent atoms are taken together to form a C3-C10cycloalkyl or heterocycloalkyl; each optionally substituted with one or more R1b;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1aon the same atom are taken together to form an oxo;each R1bis independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1bon the same atom are taken together to form an oxo;n is 0, 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;R3is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;each of R4a, R4b, and R4cis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —NRcRd, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R5is hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each R6is independently hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R7is hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, or C1-C6aminoalkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C6-C10aryl or heteroaryl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C6-C10aryl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is phenyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is heteroaryl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is furanyl, pyrrolyl, thiophenyl, oxazolyl, imidazolyl, thiazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, or pyrimidinyl. In some embodiments are provided compounds of Formula (I). Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl, or 5-pyrazolyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazolyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrazolyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazolyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-pyridinyl, or 6-pyridinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, or 6-pyrazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrimidinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrimidinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrimidinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrimidinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, or 6-pyridazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridazinyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C3-C10cycloalkyl or heterocycloalkyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C3-C10cycloalkyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is heterocycloalkyl. Further provided herein are compounds of Formula (I). Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C1-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, —OC1-C6haloalkyl, —CF3, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, bromo, iodo, —CN, —OH, —ORa, C1-C6alkyl, —OC1-C6haloalkyl, —CF3, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, bromo, —CN, —OH, —OC1-C6alkyl, —OC1-C6haloalkyl, C1-C6alkyl, —CF3, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the —OC1-C6alkyl, C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each Rais independently fluoro, chloro, —CN, —OH, —OCH3, —OCH2CH3, —C1-C6alkyl(OR1a), —CH3, —CH2CH3, iso-propyl, n-propyl, n-butyl, i-butyl, t-butyl, —OCHF2, —OC1-C6hydroxyalkyl, —CF3, cyclopropyl, azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, 1,4-oxazepanyl, or thiazinyl; wherein each of the azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, thiazinyl, and 1,4-oxazepanyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OH, —OCH3, —OCH2CH3, —CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, or 1,4-oxazepanyl; wherein each of the azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, and 1,4-oxazepanyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, azetidinyl, piperidinyl, piperazinyl, or morpholinyl; wherein each of the azetidinyl, piperidinyl, piperazinyl, and morpholinyl, is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, azetidinyl, piperidinyl, piperazinyl, or morpholinyl; wherein each of the azetidinyl, piperidinyl, piperazinyl, and morpholinyl, is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more R1a. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. In some embodiments are provided compounds of Formula (I). Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, —OCHF2, cyclopropyl, or morpholinyl; wherein morpholinyl is optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3—CH3, —OCHF2, cyclopropyl, or morpholinyl; wherein morpholinyl is optionally substituted with one or more —CH3. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 1, 2, or 3. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 1. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 2. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 3. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R2is hydrogen. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R3is hydrogen. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care independently hydrogen or halogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4ais halogen and R4b, and R4care hydrogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare hydrogen and R4bis halogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare hydrogen and R4cis halogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare halogen and R4cis hydrogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare halogen and R4cis hydrogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4band R4care halogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care hydrogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R5is hydrogen. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R6is hydrogen. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is hydrogen or C1-C6alkyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is hydrogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is C1-C6alkyl. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each of R8a, R8b, R8c, and R8dis independently hydrogen, halogen, or —ORa. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each of R8a, R8b, and R8dare hydrogen and R8cis hydrogen, halogen, or —ORa. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis halogen or —ORa. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis halogen. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis fluoro, chloro, bromo, or iodo. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis —ORa. Further provided herein are compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Rais C1-C6alkyl. In some embodiments are provided compounds of Formula (I), Formula (Ia), and Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Rais —CH3. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl or heteroaryl, C3-C10cycloalkyl, and heterocycloalkyl;each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;n is 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen or C1-C6alkyl;R3is hydrogen or C1-C6alkyl;R4a, R4b, and R4care each independently hydrogen, deuterium, or halogen;R7is hydrogen or C1-C6alkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, or —ORa;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C4hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C1-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C6-C10aryl or heteroaryl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C6-C10aryl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is phenyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is heteroaryl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is furanyl, pyrrolyl, thiophenyl, oxazolyl, imidazolyl, thiazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, or pyrimidinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl, or 5-pyrazolyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazolyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrazolyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazolyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-pyridinyl, or 6-pyridinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, or 6-pyrazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrimidinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrimidinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrimidinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrimidinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, or 6-pyridazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridazinyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C3-C10cycloalkyl or heterocycloalkyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C3-C10cycloalkyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is heterocycloalkyl. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, OC1-C6haloalkyl, C1-C6hydroxyalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, bromo, iodo, —CN, —OH, —ORa, C1-C6alkyl, —CF3, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, bromo, —CN, —OH, —ORa, C1-C6alkyl, —CF3, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, —CN, —OH, —OC1-C6alkyl, C1-C6alkyl, —CF3, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the —OC1-C6alkyl, C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, —CN, —OH, —OCH3, —OCH2CH3, —C1-C6alkyl(OR1a), —CH3, —CH2CH3, iso-propyl, n-propyl, n-butyl, i-butyl, t-butyl, —OC1-C6hydroxyalkyl, —CF3, —OCHF2, cyclopropyl, azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, 1,4-oxazepanyl, or thiazinyl; wherein each of the azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, thiazinyl, and 1,4-oxazepanyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OH, —OCH3, —OCH2CH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, or 1,4-oxazepanyl; wherein each of the azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, and 1,4-oxazepanyl is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, azetidinyl, piperidinyl, piperazinyl, or morpholinyl; wherein each of the azetidinyl, piperidinyl, piperazinyl, and morpholinyl, is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH1, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein each of the cyclopropyl, piperidinyl, piperazinyl, and morpholinyl, is optionally and independently substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more R1a. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3, —CH3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3—CH3, —OCHF2, cyclopropyl, or morpholinyl; wherein morpholinyl is optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3, —CH3, cyclopropyl, or morpholinyl; wherein morpholinyl is optionally substituted with one or more —CH3. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 1, 2, or 3. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 1. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 2. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 3. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R2is hydrogen. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R3is hydrogen. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care independently hydrogen or halogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4ais halogen and R4b, and R4care hydrogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4care hydrogen and R4bis halogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare hydrogen and R4cis halogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare halogen and R4cis hydrogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4care halogen and R4bis hydrogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4ais halogen and R4band R4care hydrogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care hydrogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care halogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care fluoro. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is hydrogen or C1-C6alkyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is hydrogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is C1-C6alkyl. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, or —ORa. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each of R8a, R8b, and R8dare hydrogen and R8cis hydrogen, halogen, or —ORa. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis halogen or —ORa. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis halogen. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis fluoro, chloro, bromo, or iodo. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis —ORa. Further provided herein are compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Rais C1-C6alkyl. In some embodiments are provided compounds of Formula (II), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Rais —CH3. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is heteroaryl; each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, 3, 4, 5, 6, 7, or 8; R7is hydrogen or C1-C6alkyl; R8cis halogen or —ORa; andeach Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl, or 5-pyrazolyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazolyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrazolyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazolyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-pyridinyl, or 6-pyridinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, or 6-pyrazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrimidinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrimidinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrimidinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrimidinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, or 6-pyridazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridazinyl. In some embodiments are provided compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridazinyl. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein:Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl;each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C1-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a;each R1ais independently —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6hydroxyalkyl, or C1-C6heteroalkyl;n is 1, 2, or 3;R7is hydrogen;R8cis —ORa; andeach Rais C1-C6alkyl. In some embodiments. Ring A is pyrimidinyl. In other embodiments, Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. Instill further embodiments, Ring A is 2-pyrimidinyl. Further embodiments provide Ring A is 4-pyrimidinyl. In other embodiments, Ring A is 5-pyrimidinyl. In yet other embodiments, Ring A is 6-pyrimidinyl. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein:Ring A is pyridinyl or pyrimidinyl;each R1is independently fluoro, chloro, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a;each R1ais independently —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6hydroxyalkyl, or C1-C6heteroalkyl;n is 1, 2, or 3;R7is hydrogen;R8cis —OCH3; andeach Rais C1-C6alkyl. In some embodiments, Ring A is pyridinyl. In other embodiments, Ring A is 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-pyridinyl, or 6-pyridinyl. In still further embodiments, Ring A is 2-pyridinyl. In some embodiments, Ring A is 3-pyridinyl. In some embodiments, Ring A is 4-pyridinyl. In some embodiments, Ring A is 5-pyridinyl. In some embodiments, Ring A is 6-pyridinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. In some embodiments, Ring A is 2-pyrimidinyl. In some embodiments, Ring A is 4-pyrimidinyl. In some embodiments, Ring A is 5-pyrimindinyl. In some embodiments, Ring A is 6-pyrimindinyl. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein:Ring A is pyridinyl or pyrimidinyl;each R1is independently chloro, —CN, —OH, —OCH3, —OCH2CH3, —CH3, —CH2CH3, —CH(CH3)2, —CF3, —OCHF2, cyclopropyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, 1,1-dioxidothiomorpholinyl, or oxetanyl; wherein morpholinyl, piperidinyl, piperazinyl, azetidinyl, 1,1-dioxidothiomorpholinyl, and oxetanyl are each optionally and independently substituted with one or more R1a;each R1ais independently —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6hydroxyalkyl, or C1-C6heteroalkyl;n is 1, 2, or 3;R7is hydrogen;R8cis —OCH3; andeach Rais C1-C6alkyl. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is heteroaryl; each R1is independently halogen, —CN, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, or 3; R7is hydrogen or C1-C6alkyl; R8cis halogen or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C10alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CHs, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; and each Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CHs, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. In some embodiments, Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. In some embodiments, Ring A is pyrazolyl. In some embodiments, Ring A is pyridinyl. In some embodiments, Ring A is pyrazinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is pyridazinyl. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is heteroaryl; each R1is independently halogen, —OR3, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C10alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, or 3: R7is hydrogen or C1-C6alkyl;R8cis halogen or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C1-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C10alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; and each Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. In some embodiments, Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. In some embodiments. Ring A is pyrazolyl. In some embodiments, Ring A is pyridinyl. In some embodiments, Ring A is pyrazinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is pyridazinyl Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is heteroaryl; each R1is independently halogen, —ORa, —SRa, —S(═O)R3, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, or 3; R7is hydrogen or C1-C6alkyl; R8cis halogen or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH2, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH2, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkenyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; and each Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. In some embodiments, Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. In some embodiments, Ring A is pyrazolyl. In some embodiments, Ring A is pyridinyl. In some embodiments, Ring A is pyrazinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is pyridazinyl. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —S(═O)2(C1-C6alkyl), —S(═O)2N(C1-C6alkyl)2, —OC1-C6alkyl, —C(═O)N(C1-C6alkyl)2, —C(═O)N(H)(C1-C6alkyl), —OC1-C6haloalkyl, C1-C6alkyl, —S(C1-C6alkyl), heterocycloalkyl, or —C(═O)(heterocycloalkyl); n is 1, 2, or 3; R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. In some embodiments, Ring A is phenyl. In some embodiments, Ring A is pyrazolyl. In some embodiments, Ring A is pyridinyl. In some embodiments, Ring A is pyrazinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is pyridazinyl. In some embodiments, R8cis hydrogen. In some embodiments, R8cis halogen. In some embodiments, R8cis fluoro, chloro, bromo, or iodo. In some embodiments, R8cis fluoro. In some embodiments, R8cis CH3. In some embodiments, R8cis or —OCH3. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(CH3), —S(═O)2(CH2CH3)—S(═O)2N(CH3)2, —OCH3, —CH2CHF2, —C(═O)N(CH3)2, —C(═O)N(H)(CH3), —OC1C6haloalkyl, —CH3, —CH2CH3, iso-propyl, n-propyl, —SCH3, azetidinyl, pyrrolidinyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl; n is 1, 2, or 3; R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. In some embodiments, Ring A is phenyl. In some embodiments, Ring A is pyrazolyl. In some embodiments, Ring A is pyridinyl. In some embodiments, Ring A is pyrazinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is pyridazinyl. In some embodiments, R8cis hydrogen. In some embodiments, R8cis halogen. In some embodiments. R8cis fluoro, chloro, bromo, or iodo. In some embodiments, R8cis fluoro. In some embodiments, R8cis CH3. In some embodiments, R8cis or —OCH3. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(CH3), —S(═O)2(CH2CH3), —S(═O)2(i-Pr), —S(═O)2(cyclopropyl), —S(═O)2(C1C6haloalkyl), —S(═O)2N(CH3)2, —S(═O)2NH2, —S(═O)2N(CH3)(H), —OCH3, —OCH3, —OCH2CH3, —CH2CHF2, —C(═O)N(CH3)2, —C(═O)N(H)(CH3), —OC1-C6haloalkyl, —CH3, —CH2CH3, iso-propyl, n-propyl, —SCH3, azetidinyl, pyrrolidinyl, oxazolyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl; n is 1, 2, or 3: R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. In some embodiments, Ring A is phenyl. In some embodiments, Ring A is pyrazolyl. In some embodiments, Ring A is pyridinyl. In some embodiments, Ring A is pyrazinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is pyridazinyl. In some embodiments, R8cis hydrogen. In some embodiments, R8cis halogen. In some embodiments, R8cis fluoro, chloro, bromo, or iodo. In some embodiments, R8cis fluoro. In some embodiments, R8cis CH. In some embodiments, R8cis or —OCH3. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(C1-C6alkyl), —S(═O)2(C3-C10cycloalkyl), —S(═O)2N(C1-C6alkyl)2, —S(═O)?NH2, —S(═O)2N(C1-C6alkyl)(H), —OC1-C6alkyl, —CH2CHF2, —C(═O)N(C1-C6alkyl)2, —C(═O)N(H)(C1-C6alkyl), —OC1C6haloalkyl, —C1-C6alkyl, —SC1-C6alkyl, azetidinyl, pyrrolidinyl, oxazolyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl; n is 1, 2, or 3: R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. In some embodiments, Ring A is phenyl. In some embodiments, Ring A is pyrazolyl. In some embodiments, Ring A is pyridinyl. In some embodiments, Ring A is pyrazinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is pyridazinyl. In some embodiments, R8cis hydrogen. In some embodiments, R8cis halogen. In some embodiments, R8cis fluoro, chloro, bromo, or iodo. In some embodiments, R8cis fluoro. In some embodiments, R8cis CH3. In some embodiments, R8cis or —OCH3. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(C1-C6alkyl), —S(═O)2(C3-C10cycloalkyl), —S(═O)2N(C1-C6alkyl)2, —S(═O)2NH2, —S(═O)2N(C1-C6alkyl)(H), —OC1-C6alkyl, —CH2CHF2, —C(═O)N(C1-C6alkyl)2, —C(═O)N(H)(C1-C6alkyl), —OC1C6haloalkyl, —C1-C6alkyl, —SC1-C6alkyl, azetidinyl, pyrrolidinyl, oxazolyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl, and wherein at least one of R1is —S(═O)2(C1-C6alkyl), —S(═O)2(C3-C10cycloalkyl), —S(═O)2N(C1-C6alkyl)2, —S(═O)2NH2, or —S(═O)2N(C1-C6alkyl)(H); n is 1, 2, or 3; R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. In some embodiments, Ring A is phenyl. In some embodiments, Ring A is pyrazolyl. In some embodiments, Ring A is pyridinyl. In some embodiments, Ring A is pyrazinyl. In some embodiments, Ring A is pyrimidinyl. In some embodiments, Ring A is pyridazinyl. In some embodiments, R8cis hydrogen. In some embodiments, R8cis halogen. In some embodiments, R8cis fluoro, chloro, bromo, or iodo. In some embodiments, R8cis fluoro. In some embodiments, R8cis CH3. In some embodiments, R8cis or —OCH3. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is heterocycloalkyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(C1-C6alkyl), —S(═O)2(C3-C10cycloalkyl), —S(═O)2N(C1-C6alkyl)2, —S(═O)2NH2, —S(═O)2N(C1-C6alkyl)(H), —OC1-C6alkyl, —CH2CHF2, —C(═O)N(C1-C6alkyl)2, —C(═O)N(H)(C1-C6alkyl), —OC1C6haloalkyl, —C1-C6alkyl, —SC1-C6alkyl, azetidinyl, pyrrolidinyl, oxazolyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl; n is 1, 2, or 3: R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. In some embodiments, Ring A is dihydropyridazinyl. In some embodiments, Ring A is 1,6-dihydropyridazin-3-yl. In some embodiments, R8cis hydrogen. In some embodiments, R8cis halogen. In some embodiments, R8cis fluoro, chloro, bromo, or iodo. In some embodiments, R8cis fluoro. In some embodiments, Rcis CH3. In some embodiments, R8cis or —OCH3. Further provided herein are compounds of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is aryl; each R1is independently halogen, —CN, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C10alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, or 3: R7is hydrogen or C1-C6alkyl; R8cis hydrogen, C1-C6alkyl, halogen or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; and each Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CHs, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Further provided herein are compounds of Formula (I) selected from (1R,2S)-5′-methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(5-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloropyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-ethoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-cyclopropylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloropyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1S,2R)-5′-methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2-chloro-5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(piperidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyrazin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(6-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyridin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxypyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyridin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(4-methylpiperazin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(1,3,5-trimethyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-(trifluoromethyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloro-2-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methoxypyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-1-methyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(3-hydroxy-2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[(3S)-3-hydroxy-2,3-dihydro-1-benzofuran-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[(3R)-3-hydroxy-2,3-dihydro-1-benzofuran-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,3-dihydropyrazolo[5,1-b][1,3]oxazol-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-oxo-2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,3-dihydrofuro[2,3-c]pyridin-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[(3S)-3-(hydroxymethyl)-2,3-dihydrofuro[2,3-c]pyridin-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[(3R)-3-(hydroxymethyl)-2,3-dihydrofuro[2,3-c]pyridin-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((6-(3-oxa-8-azabicyclo[3.2.1]octan-8-yl)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-{[6-(2-hydroxyethoxy)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one); (1R,2S)-5′-methoxy-2-(3-{[6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-methyl-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(azetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidine-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[1-(2-hydroxyethyl)-3-methoxy-1H-pyrazol-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-cyclopropyl-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((5-chloro-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (R,2S)-2-(3-{[5-(2-hydroxyethyl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(2-hydroxyethyl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-5-methoxy-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((5-chloro-6-(1,1-dioxidothiomorpholino)-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((2-cyclopropyl-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-6-(morpholin-4-yl)pyrimidine-2-carbonitrile; 4-(1,1-dioxidothiomorpholino)-5-methoxy-6-((6-((1R,2S)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazol-3-yl)amino)pyrimidine-2-carbonitrile; (1R,2S)-2-{3-[(1,3-dimethyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[1-methyl-3-(trifluoromethyl)-1H-pyrazol-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(1-methyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-1-methyl-1H-pyrazole-3-carbonitrile; (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxy-2-methylpyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(2-hydroxyethyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((2-cyclopropyl-6-(1,1-dioxidothiomorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((5-chloro-2-cyclopropyl-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyrazin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-5-methoxy-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(1,3-dimethyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-cyclopropyl-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-[3-({2-cyclopropyl-6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((5-chloro-6-(1,1-dioxidothiomorpholino)-2-isopropylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methyl-1H-pyrazol-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(6-cyclopropyl-3-methoxypyrazin-2-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-cyclopropyl-6-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(3,6-dimethylpyrazin-2-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-2-isopropyl-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(morpholin-4-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(5-methoxy-2-methylpyridin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyridin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methoxy-5-methylpyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxypyridazin-3-yl)amino]-1H-indazol-6-yl)}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(3-cyclopropyl-1-methyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(3-cyclopropyl-1-ethyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(2-hydroxy-2-methylpropyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(morpholin-4-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-2-(morpholin-4-yl)pyridin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloro-2-methylpyridin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloro-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(morpholin-4-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(oxetan-3-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(propan-2-yl)pyridazin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[6-(morpholin-4-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(3-hydroxyazetidin-1-yl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methyl-6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-1′-ethyl-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(azetidin-3-yl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; and (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Further provided herein are compounds of Formula (I) selected from (1R,2S)-2-(3-{[1-(2,2-difluoroethyl)-3-methyl-1H-pyrazol-5-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-1′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[6-(oxetan-3-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-(oxetan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxetan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-1′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2R)-2-{7-fluoro-3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1S,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-4′-fluoro-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1S,2S)-4′-fluoro-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-6′-fluoro-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-fluoro-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-ethoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-fluorospiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-[3-({2-methyl-5-[(propan-2-yl)oxy]pyrimidin-4-yl}amino)-1H-indazol-6-yl]spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-(trifluoromethyl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-(trifluoromethoxy)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1S,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-(trifluoromethoxy)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-cyclopropyl-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-(oxetan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2R)-5′-fluoro-2-{7-fluoro-3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; and (1R,2R)-2-(3-{[5-(difluoromethoxy)-2-methylpyrimidin-4-yl]amino}-7-fluoro-1H-indazol-6-yl)-5′-fluorospiro[cyclopropane-1,3′-indol]-2′(1′H)-one, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Further provided herein are compounds of Formula (I) selected from (R,2R)-2-{5-fluoro-3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyridazin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(cyclopropylmethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(2,2-difluoroethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-[3-(2-methoxy-5-methylanilino)-1H-indazol-6-yl]spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methyl-5-(2,2,2-trifluoroethoxy)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methoxy-6-methylpyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methyl-6-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-ethyl-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methyl-6,7-dihydrofuro[3,2-d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,5-dimethoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(trifluoromethyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2-ethyl-5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(7-methoxyquinolin-6-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methyl-5-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxyquinolin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,5-dimethoxypyridin-3-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2-chlorofuro[3,2-d]pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(pyrrolidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(methanesulfonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-[3-({2-[(3R)-3-fluoropyrrolidin-1-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(3,3-difluoropyrrolidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2-chloro-5-methyl-5H-pyrrolo[3,2-d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidine-2-carbonitrile; (1R,2S)-2-(3-{[2-(3,3-difluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(3-fluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[5-(ethanesulfonyl)-2-methoxyanilino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-methoxy-3-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylbenzamide; 4-methoxy-3-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methylbenzamide; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-5-(propane-2-sulfonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-methoxy-3-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylbenzene-1-sulfonamide; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-5-(morpholine-4-carbonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-carboxamide; (1R,2S)-2-(3-{[2-(dimethylamino)-5-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(morpholine-4-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3,3-difluoroazetidine-1-carbonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(4,4-difluoropiperidine-1-carbonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(4-fluoro-3-{[5-methoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methoxy-5-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(2-oxa-6-azaspiro[3.3]heptane-6-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(4,4-difluoropiperidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-[5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidin-2-yl]-1λ6-thiomorpholine-1,1-dione; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methylpyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(2-oxa-6-azaspiro[3.3]heptan-6-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(methanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methyl-N-(propan-2-yl)pyridine-2-carboxamide; (1R,2S)-2-(3-{[5-ethoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(methanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-carboxamide; 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-4-(morpholine-4-carbonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; Diastereomer 1: (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxolan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; Diastereomer 2: (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxolan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-ethoxy-6-(methanesulfonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-ethoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-carboxamide; 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-sulfonamide; (1R,2S)-2-(3-{[6-(dimethylphosphoryl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 2-fluoro-5-methoxy-4-((6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl)amino)-N,N-dimethylbenzamide; (1R,2S)-2-{3-[5-fluoro-2-methoxy-4-(morpholine-4-carbonyl)anilino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; and (1R,2S)-2-(3-{[3-ethoxy-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one, or a pharmaceutically acceptable salt thereof. Further provided herein are compounds of Formula (I) selected from (1R,2S)-2-(3-{[6-(ethanesulfonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,2-dimethylbenzene-1-sulfonamide; (1R,2S)-2-{3-[(2,5-dimethyl-5,7-dihydrothieno[3,4-d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 2,5-dimethoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)benzene-1-sulfonamide; (1R,2S)-2-(3-{[24dimethylamino)-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-ethoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(2-oxopyrrolidin-1-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-5-(morpholine-4-sulfonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-1,5-naphthyridin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; N,6dimethoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methylpyridine-2-carboxamide; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N′-(propan-2-yl)pyridine-2-carbohydrazide; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-5-(2-oxopyrrolidin-1-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-5-(3-methyl-2-oxoimidazolidin-1-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-sulfonamide; 6-ethoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-sulfonamide; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-5-(oxane-4-sulfonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(dimethylphosphoryl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(2-hydroxypropan-2-yl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (0R,2S)-2-(3-{[6-(methanesulfonyl)-2-methoxy-5-methylpyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(ethanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(dimethylphosphoryl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; N-(cyclopropylmethyl)-6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carboxamide; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-(propan-2-yl)pyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(3-oxa-8-azabicyclo[3.2.1]octane-8-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methyl-6-oxo-1,6-dihydropyridazin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-5-(1,3-oxazol-2-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(3-methoxyazetidine-1-carbonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyrazine-2-carboxamide; 6-ethoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyrazine-2-carboxamide; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N,3-trimethylpyridine-2-carboxamide; (1R,2S)-2-(3-{[6-(methanesulfinyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(methanesulfinyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(methanesulfonyl)-4-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(methanesulfonyl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; N,N-dicyclopropyl-6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carboxamide; N-(2,2-difluoroethyl)-6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carboxamide; (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylpiperidine-1-carbonyl]-2-methoxypyridin-3-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(8-oxa-3-azabicyclo[3.2.1]octane-3-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[3-chloro-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(propane-2-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(dimethylphosphoryl)-4-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; N-(1,3-difluoropropan-2-yl)-6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carboxamide; 6-chloro-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; (1R,2S)-2-{3-[(5-chloro-2-methyl-1,3-benzoxazol-6-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; 3-[6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrazin-2-yl]-1)6-thietane-1,1-dione; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(morpholine-4-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(8-oxa-3-azabicyclo[3.2.1]octane-3-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(diethylphosphoryl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(cyclopropanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(oxane-4-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carbonitrile; (1R,2S)-2-{3-[5-(diethylphosphoryl)-2-methoxyanilino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(ethanesulfonyl)-4-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(azetidine-1-carbonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(morpholine-4-carbonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(6-methoxy-2-methyl-1-oxo-2,3-dihydro-1H-isoindol-5-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-2-methyl-1-oxo-2,3-dihydro-1H-isoindol-5-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(1,2-oxazolidine-2-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(azetidine-1-sulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-methoxy-6-{(6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-(3-methyloxetan-3-yl)pyridine-3-sulfonamide; 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methyl-N-(3-methyloxetan-3-yl)pyridine-3-sulfonamide; (1R,2S)-2-(3-{[5-(I-hydroxyethyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[2-ethoxy-4-(methanesulfonyl)anilino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[3-ethoxy-5-(4-methylpiperazine-1-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(ethanesulfonyl)-3-ethoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-ethoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methylpyridine-3-sulfonamide; (1R,2S)-5′-chloro-2-(3-{[3-ethoxy-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(4-ethoxy-1-methyl-6-oxo-1,6-dihydropyridazin-3-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(2-hydroxypropan-2-yl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[4-ethoxy-6-(methanesulfonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; and (1R,2S)-2-(3-{[5-(difluoromethanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one, or a pharmaceutically acceptable salt thereof. Further provided herein are compounds of Formula (I) selected from those set forth in Table 1A. TABLE 1AExampleNo.StructureName1±-5′-methoxy-2-{3-[(5- methoxypyrimidin-4-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one (racemic mixture)2±-5′-methoxy-2-{3-[(5- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one (racemic mixture)3±-2-{3-[(5-chloropyrimidin-4- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one (racemic mixture)4(1R,2S)-5′-methoxy-2-{3-[(5- methoxypyrimidin-4-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one5(1R,2S)-2-{3-[(5- ethoxypyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one6(1R,2S)-2-{3-[(5- cyclopropylpyrimidin-4- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one7(1R,2S)-2-{3-[(5-chloropyrimidin- 4-yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one8(1S,2R)-5′-methoxy-2-{3-[(5- methoxypyrimidin-4-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one9(1R,2S)-2-(3-{[5-chloro-6- (morpholin-4-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one10(1R,2S)-2-{3-[(2-chloro-5- methoxypyrimidin-4-yl)amino]- 1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one11(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-6-(morpholin-4- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one12(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-6-(piperidin-1- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one13(1R,2S)-5′-methoxy-2-{3-[(3- methoxypyrazin-2-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one14(1R,2S)-5′-methoxy-2-{3-[(6- methoxypyrimidin-4-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one15(1R,2S)-2-{3-[(6,7-dihydro-5H- cyclopenta[d]pyrimidin-4- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one16(1R,2S)-2-{3-[(2,3-dihydro-1- benzofuran-7-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one17(1R,2S)-5′-methoxy-2-{3-[(3- methoxypyridin-2-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one18(1R,2S)-5′-methoxy-2-{3-[(4- methoxypyridin-3-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one19(1R,2S)-5′-methoxy-2-{3-[(3- methoxypyridin-4-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one20(1R,2S)-2-(3-{[5-chloro-6-(4- methylpiperazin-1-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one21(1R,2S)-5′-methoxy-2-{3-[(1,3,5- trimethyl-1H-pyrazol-4-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one22(1R,2S)-5′-methoxy-2-(3-{[5- (trifluoromethyl)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one23(1R,2S)-2-{3-[(5-chloro-2- methoxypyrimidin-4-yl)amino]- 1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one24(1R,2S)-5′-methoxy-2-{3-[(2- methoxypyridin-3-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one25(1R,2S)-5′-methoxy-2-{3-[(3- methoxy-1-methyl-1H-pyrazol-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one26(1R,2S)-2-{3-[(1-benzofuran-7- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one27(1R,2S)-2-{3-[(3-hydroxy-2,3- dihydro-1-benzofuran-7- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one (mixture of diastereomers)28(1R,2S)-2-(3-{[(3S)-3-hydroxy- 2,3-dihydro-1-benzofuran-7- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one29(1R,2S)-2-(3-{[(3R)-3-hydroxy- 2,3-dihydro-1-benzofuran-7- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one30(1R,2S)-2-{3-[(2,3- dihydropyrazolo[5,1- b][1,3]oxazol-7-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one31(1R,2S)-5′-methoxy-2-{3-[(3-oxo- 2,3-dihydro-1-benzofuran-7- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one32(1R,2S)-2-{3-[(2,3- dihydrofuro[2,3-c]pyridin-7- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one33(1R,2S)-2-(3-{[(3S)-3- (hydroxymethyl)-2,3- dihydrofuro[2,3-c]pyridin-7- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one34(1R,2S)-2-(3-{[(3R)-3- (hydroxymethyl)-2,3- dihydrofuro[2,3-c]pyridin-7- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one35(1R,2S)-5′-methoxy-2-(3-{[6-(3- methoxyazetidin-1-yl)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one36(1R,2S)-2-(3-{[6-(3- hydroxyazetidin-1-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one37(1R,2S)-2-(3-((6-(3-oxa-8- azabicyclo[3.2.1]octan-8-yl)-5- methoxypyrimidin-4-yl)amino)- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one38(1R,2S)-2-(3-{[6-(2- hydroxyethoxy)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one39(1R,2S)-2-(3-((6-(1,1- dioxidothiomorpholino)pyrimidin- 4-yl)amino)-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one)40(1R,2S)-5′-methoxy-2-(3-{[6-(1,4- oxazepan-4-yl)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one41(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-2-methyl-6-(morpholin- 4-yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one42(1R,2S)-2-(3-{[6-(azetidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one43(1R,2S)-2-(3-{[6-(3- hydroxyazetidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one44(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-6-(1,4-oxazepan-4- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one45(1R,2S)-2-(3-{[6-(azetidin-1- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one46(1R,2S)-2-(3-{[5-chloro-6-(3- hydroxyazetidin-1-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one47(1R,2S)-2-(3-{[5-chloro-6-(3- methoxyazetidin-1-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one48(1R,2S)-2-(3-{[2-chloro-5- methoxy-6-(morpholin-4- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one49(1R,2S)-2-(3-{[4-chloro-5- methoxy-6-(morpholin-4- yl)pyrimidin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one50(1R,2S)-2-(3-{[1-(2- hydroxyethyl)-3-methoxy-1H- pyrazol-4-yl]amino}-1H-indazol- 6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one51(1R,2S)-2-(3-{[2-cyclopropyl-5- methoxy-6-(morpholin-4- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one52(1R,2S)-2-[3-({6-[(2R,6S)-2,6- dimethylmorpholin-4-yl]-5- methoxypyrimidin-4-yl}amino)- 1H-indazol-6-yl]-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one53(1R,2S)-2-(3-((5-chloro-6-(1,1- dioxidothiomorpholino)pyrimidin- 4-yl)amino)-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one54(1R,2S)-2-(3-((6-(1,1- dioxidothiomorpholino)-5- methoxypyrimidin-4-yl)amino)- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one55(1R,2S)-2-(3-{[5-(2- hydroxyethyl)-3-methoxypyrazin- 2-yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one56(1R,2S)-2-(3-{[6-(2- hydroxyethyl)-3-methoxypyrazin- 2-yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one57(1R,2S)-2-(3-((6-(1,1- dioxidothiomorpholino)-5- methoxy-2-methylpyrimidin-4- yl)amino)-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one58(1R,2S)-2-(3-((5-chloro-6-(1,1- dioxidothiomorpholino)-2- methylpyrimidin-4-yl)amino)-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one59(1R,2S)-2-(3-((2-cyclopropyl-6- (1,1- dioxidothiomorpholino)pyrimidin- 4-yl)amino)-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one60(1R,2S)-2-(3-((6-(1,1- dioxidothiomorpholino)-2- methylpyrimidin-4-yl)amino)-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one615-methoxy-4-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-6-(morpholin-4- yl)pyrimidine-2-carbonitrile624-(1,1-dioxidothiomorpholino)-5- methoxy-6-((6-((1R,2S)-5′- methoxy-2′- oxospiro[cyclopropane-1,3′- indolin]-2-yl)-1H-indazol-3- yl)amino)pyrimidine-2- carbonitrile63(1R,2S)-2-{3-[(1,3-dimethyl-1H- pyrazol-4-yl)amino]-1H-indazol- 6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one64(1R,2S)-5′-methoxy-2-(3-{[1- methyl-3-(trifluoromethyl)-1H- pyrazol-4-yl]amino}-1H-indazol- 6-yl)spiro[cyclopropane-1,3′- indol]-2′(1′H)-one65(1R,2S)-5′-methoxy-2-{3-[(1- methyl-1H-pyrazol-4-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one664-({6-[(1R,2S)-5′-methoxy-2′- oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-1-methyl-1H-pyrazole- 3-carbonitrile67(1R,2S)-2-[3-({6-[(2R,6S)-2,6- dimethylmorpholin-4-yl]-5- methoxy-2-methylpyrimidin-4- yl}amino)-1H-indazol-6-yl]-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one68(1R,2S)-2-(3-{[2-(2- hydroxyethyl)-5-methoxy-6- (morpholin-4-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one69(1R,2S)-2-(3-((2-cyclopropyl-6- (1,1-dioxidothiomorpholino)-5- methoxypyrimidin-4-yl)amino)- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one70(1R,2S)-2-(3-((5-chloro-2- cyclopropyl-6-(1,1- dioxidothiomorpholino)pyrimidin- 4-yl)amino)-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one71(1R,2S)-5′-methoxy-2-{3-[(3- methoxy-6-methylpyrazin-2- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one72(1R,2S)-2-(3-{[5-chloro-6-(3- hydroxyazetidin-1-yl)-2- methylpyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one73(1R,2S)-2-(3-{[6-(3- hydroxyazetidin-1-yl)-5-methoxy- 2-methylpyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one74(1R,2S)-2-{3-[(1,3-dimethyl-1H- pyrazol-5-yl)amino]-1H-indazol- 6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one75(1R,2S)-5′-methoxy-2-{3-[(4- methoxy-1-methyl-1H-pyrazol-5- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one76(1R,2S)-2-(3-{[5-chloro-2- cyclopropyl-6-(3-hydroxyazetidin- 1-yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one77(1R,2S)-2-[3-({2-cyclopropyl-6- [(2R,6S)-2,6-dimethylmorpholin- 4-yl]-5-methoxypyrimidin-4- yl}amino)-1H-indazol-6-yl]-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one78(1R,2S)-2-(3-((5-chloro-6-(1,1- dioxidothiomorpholino)-2- isopropylpyrimidin-4-yl)amino)- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one79(1R,2S)-5′-methoxy-2-{3-[(4- methoxy-1-methyl-1H-pyrazol-3- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one80(1R,2S)-2-{3-[(6-cyclopropyl-3- methoxypyrazin-2-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one81(1R,2S)-2-(3-{[2-cyclopropyl-6- (3-hydroxyazetidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one82(1R,2S)-2-{3-[(3,6- dimethylpyrazin-2-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one83(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-6-(propan-2-yl)pyrazin- 2-yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one84(1R,2S)-2-(3-((6-(1,1- dioxidothiomorpholino)-2- isopropyl-5-methoxypyrimidin-4- yl)amino)-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one85(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-6-(morpholin-4-yl)-2- (propan-2-yl)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one86(1R,2S)-5′-methoxy-2-{3-[(5- methoxy-2-methylpyridin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one87(1R,2S)-5′-methoxy-2-{3-[(5- methoxy-2-methylpyrimidin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one88(1R,2S)-5′-methoxy-2-{3-[(3- methoxy-6-methylpyridin-2- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one89(1R,2S)-5′-methoxy-2-{3-[(2- methoxy-5-methylpyridin-3- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one90(1R,2S)-5′-methoxy-2-{3-[(4- methoxypyridazin-3-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one91(1R,2S)-2-{3-[(3-cyclopropyl-1- methyl-1H-pyrazol-5-yl)amino]- 1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one92(1R,2S)-2-{3-[(3-cyclopropyl-1- ethyl-1H-pyrazol-5-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one93(1R,2S)-2-(3-{[2-(2-hydroxy-2- methylpropyl)-5-methoxy-6- (morpholin-4-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one94(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5-(morpholin-4- yl)pyrazin-2-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one95(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-2-(morpholin-4- yl)pyridin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one96(1R,2S)-2-{3-[(5-chloro-2- methylpyridin-4-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one97(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-2-(morpholin-4- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one98(1R,2S)-2-{3-[(5-chloro-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one99(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-6-(morpholin-4- yl)pyrazin-2-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one100(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-6-(oxetan-3-yl)pyrazin- 2-yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one101(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-6-(propan-2- yl)pyridazin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one102(1R,2S)-5′-methoxy-2-(3-{[6- (morpholin-4-yl)-2-(propan-2- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one103(1R,2S)-2-(3-{[5-chloro-2- (morpholin-4-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one104(1R,2S)-2-(3-{[5-(3- hydroxyazetidin-1-yl)-3- methoxypyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one105(1R,2S)-5′-methoxy-2-(3-{[3- methyl-6-(propan-2-yl)pyrazin-2- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one106(1R,2S)-5′-methoxy-2-(3-{[6- (propan-2-yl)pyrazin-2-yl]amino}- 1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one107(1R,2S)-2-(3-{[5-chloro-6-(3- hydroxyazetidin-1-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxy-1′- methylspiro[cyclopropane-1,3′- indol]-2′(1′H)-one108(1R,2S)-2-(3-{[5-chloro-6-(3- hydroxyazetidin-1-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-1′- ethyl-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one109(1R,2S)-2-(3-{[5- (difluoromethoxy)-6-(morpholin- 4-yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one110(1R,2S)-2-(3-{[6-(azetidin-3-yl)-3- methoxypyrazin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one111(1R,2S)-2-(3-{[6-(3- hydroxyazetidin-1-yl)-2-(propan- 2-yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one112(1R,2S)-2-(3-{[1-(2,2- difluoroethyl)-3-methyl-1H- pyrazol-5-yl]amino}-1H-indazol- 6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one113(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-6-(morpholin-4- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-1′- methylspiro[cyclopropane-1,3′- indol]-2′(1′H)-one114(1R,2S)-2-(3-{[2-(3- hydroxyazetidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one115(1R,2S)-5′-methoxy-2-(3-{[6- (oxetan-3-yl)pyrazin-2-yl]amino}- 1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one116(1R,2S)-2-(3-((5-chloro-2- cyclopropylpyrimidin-4- yl)amino)-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indolin]-2′-one117(1R,2S)-2-(3-{[5-chloro-2- (propan-2-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one118(1R,2S)-2-(3-{[5-chloro-2- (oxetan-3-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one119(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-2-(oxetan-3- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one120(1R,2S)-2-(3-{[5-chloro-2-(3- hydroxyazetidin-1-yl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one121(1R,2S)-2-(3-{[5- (difluoromethoxy)-2- methylpyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one122(1R,2S)-2-{3-[(5-methoxy-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-1′- methylspiro[cyclopropane-1,3′- indol]-2′(1′H)-one123(1R,2R)-2-{7-fluoro-3-[(5- methoxy-2-methylpyrimidin-4- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one124(1R,2S)-2-{3-[(5-methoxy-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one125(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-2-(propan-2- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one126(1R,2S)-2-{3-[(5-methoxy-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- methylspiro[cyclopropane-1,3′- indol]-2′(1′H)-one127(1S,2S)-2-{3-[(5-methoxy-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- methylspiro[cyclopropane-1,3′- indol]-2′(1′H)-one128(1R,2S)-2-(3-{[5- (difluoromethoxy)-2-(propan-2- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one129(1R,2S)-4′-fluoro-2-{3-[(5- methoxy-2-methylpyrimidin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one130(1S,2S)-4′-fluoro-2-{3-[(5- methoxy-2-methylpyrimidin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one131(1R,2S)-6′-fluoro-2-{3-[(5- methoxy-2-methylpyrimidin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one132(1R,2S)-2-(3-{[5-chloro-6-(2-oxa- 6-azaspiro[3.3]heptan-6- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one133(1R,2S)-5′-fluoro-2-{3-[(5- methoxy-2-methylpyrimidin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one134(1R,2S)-2-{3-[(5-ethoxy-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one135(1R,2S)-2-(3-{[5- (difluoromethoxy)-2- methylpyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- fluorospiro[cyclopropane-1,3′- indol]-2′(1′H)-one136(1R,2S)-5′-methoxy-2-[3-({2- methyl-5-[(propan-2- yl)oxy]pyrimidin-4-yl}amino)-1H- indazol-6-yl]spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one137(1R,2S)-2-{3-[(5-methoxy-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- (trifluoromethyl)spiro[cyclo- propane-1,3′-indol]-2′(1′H)-one138(1R,2S)-2-{3-[(5-methoxy-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- (trifluoromethoxy)spiro[cyclo- propane-1,3′-indol]-2′(1′H)-one139(1S,2S)-2-{3-[(5-methoxy-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- (trifluoromethoxy)spiro[cyclo- propane-1,3′-indol]-2′(1′H)-one140(1R,2S)-2-{3-[(5-cyclopropyl-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one141(1R,25)-2-(3-{[5- (difluoromethoxy)-2-(oxetan-3- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one142(1R,2R)-5′-fluoro-2-{7-fluoro-3- [(5-methoxy-2-methylpyrimidin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one143(1R,2R)-2-(3-{[5- (difluoromethoxy)-2- methylpyrimidin-4-yl]amino}-7- fluoro-1H-indazol-6-yl)-5′- fluorospiro[cyclopropane-1,3′- indol]-2′(1′H)-one144(1R,2R)-2-{5-fluoro-3-[(5- methoxy-2-methylpyrimidin-4- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one145(1R,2S)-5′-methoxy-2-{3-[(3- methoxy-6-methylpyridazin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one146(1R,2S)-2-(3-{[5- (cyclopropylmethoxy)-2- methylpyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one147(1R,2S)-2-(3-{[5-(2,2- difluoroethoxy)-2- methylpyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one148(1R,2S)-5′-methoxy-2-[3-(2- methoxy-5-methylanilino)-1H- indazol-6-yl]spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one149(1R,2S)-5′-methoxy-2-(3-{[2- methyl-5-(2,2,2- trifluoroethoxy)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one150(1R,2S)-5′-methoxy-2-{3-[(2- methoxy-6-methylpyridin-3- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one151(1R,2S)-5′-methoxy-2-(3-{[2- methyl-6-(propan-2-yl)pyrimidin- 4-yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one152(1R,2S)-2-{3-[(5-ethyl-2- methylpyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one153(1R,2S)-5′-methoxy-2-{3-[(2- methyl-6,7-dihydrofuro[3,2- d]pyrimidin-4-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one154(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-2-(oxan-4-yl)pyrimidin- 4-yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one155(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-2- (methylsulfanyl)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one156(1R,2S)-2-{3-[(2,5- dimethoxypyrimidin-4-yl)amino]- 1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one157(1R,2S)-2-(3-{[2-(azetidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one158(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5- (trifluoromethyl)pyridin-2- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one159(1R,2S)-5′-methoxy-2-{3-[(2- methyl-6,7-dihydro-5H- cyclopenta[d]pyrimidin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one160(1R,2S)-2-{3-[(2-ethyl-5- methoxypyrimidin-4-yl)amino]- 1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one161(1R,2S)-5′-methoxy-2-{3-[(7- methoxyquinolin-6-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one162(1R,2S)-5′-methoxy-2-(3-{[2- methyl-5- (methylsulfanyl)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one163(1R,2S)-5′-methoxy-2-{3-[(3- methoxyquinolin-2-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one164(1R,2S)-2-{3-[(2,5- dimethoxypyridin-3-yl)amino]- 1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one165(1R,2S)-2-{3-[(2-chlorofuro[3,2- d]pyrimidin-4-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one1666-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 2-carboxamide167(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-2-(pyrrolidin-1- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one168(1R,2S)-2-(3-{[6- (methanesulfonyl)-2- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one169(1R,2S)-2-[3-({2-[(3R)-3- fluoropyrrolidin-1-yl]-5- methoxypyrimidin-4-yl}amino)- 1H-indazol-6-yl]-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one170(1R,2S)-2-(3-{[2-(3,3- difluoropyrrolidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one171(1R,2S)-2-{3-[(2-chloro-5-methyl- 5H-pyrrolo[3,2-d]pyrimidin-4- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one1725-methoxy-4-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)pyrimidine-2- carbonitrile173(1R,2S)-2-(3-{[2-(3,3- difluoroazetidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one174(1R,2S)-2-(3-{[2-(3- fluoroazetidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one175(1R,2S)-2-{3-[5-(ethanesulfonyl)- 2-methoxyanilino]-1H-indazol-6- yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one1764-methoxy-3-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N- dimethylbenzamide1774-methoxy-3-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N-methylbenzamide178(1R,2S)-5′-methoxy-2-{3-[2- methoxy-5-(propane-2- sulfonyl)anilino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one1794-methoxy-3-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylbenzene- 1-sulfonamide180(1R,2S)-5′-methoxy-2-{3-[2- methoxy-5-(morpholine-4- carbonyl)anilino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one1816-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 3-carboxamide182(1R,2S)-2-(3-{[2- (dimethylamino)-5- methylpyrimidin-4-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one183(1R,2S)-5′-methoxy-2-(3-{[2- methoxy-6-(morpholine-4- carbonyl)pyridin-3-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one184(1R,2S)-2-(3-{[6-(3,3- difluoroazetidine-1-carbonyl)-2- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one185(1R,2S)-2-(3-{[6-(4,4- difluoropiperidine-1-carbonyl)-2- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one186(1R,2S)-2-(4-fluoro-3-{[5- methoxy-2- (methylsulfanyl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one187(1R,2S)-5′-methoxy-2-{3-[(2- methoxy-5-methylpyrimidin-4- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one188(1R,2S)-5′-methoxy-2-(3-{[2- methoxy-6-(2-oxa-6- azaspiro[3.3]heptane-6- carbonyl)pyridin-3-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one189(1R,2S)-2-(3-{[2-(4,4- difluoropiperidin-1-yl)-5- methoxypyrimidin-4-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one1904-[5-methoxy-4-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)pyrimidin-2-yl]-1λ6- thiomorpholine-1,1-dione1916-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N-methylpyridine-2- carboxamide192(1R,2S)-5′-methoxy-2-(3-{[5- methoxy-2-(2-oxa-6- azaspiro[3.3]heptan-6- yl)pyrimidin-4-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one193(1R,2S)-2-(3-{[5- (methanesulfonyl)-3- methoxypyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one1946-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N-methyl-N-(propan-2- yl)pyridine-2-carboxamide195(1R,2S)-2-(3-{[5-ethoxy-2- (methylsulfanyl)pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one196(1R,2S)-2-(3-{[6- (methanesulfonyl)-3- methoxypyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one1975-methoxy-6-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 3-carboxamide1985-methoxy-4-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 2-carboxamide199(1R,2S)-5′-methoxy-2-{3-[2- methoxy-4-(morpholine-4- carbonyl)anilino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one200Diastercomer 1: (1R,2S)-5′- methoxy-2-(3-{[5-methoxy-2- (oxolan-3-yl)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one201Diastercomer 2: (1R,2S)-5′- methoxy-2-(3-{[5-methoxy-2- (oxolan-3-yl)pyrimidin-4- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one202(1R,2S)-2-(3-{[2-ethoxy-6- (methanesulfonyl)pyridin-3- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one2035-ethoxy-6-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 3-carboxamide2045-methoxy-6-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 3-sulfonamide205(1R,2S)-2-(3-{[6- (dimethylphosphoryl)-2- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one2062-fluoro-5-methoxy-4-({6- [(1R,2S)-5′-methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N- dimethylbenzamide207(1R,2S)-2-{3-[5-fluoro-2- methoxy-4-(morpholine-4- carbonyl)anilino]-1H-indazol-6- yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one208(1R,2S)-2-{3-[5-fluoro-2- methoxy-4-(morpholine-4- carbonyl)anilino]-1H-indazol-6- yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one209(1R,2S)-2-(3-{[6-(ethanesulfonyl)- 2-methoxypyridin-3-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one2105-methoxy-4-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,2-dimethylbenzene- 1-sulfonamide211(1R,2S)-2-{3-[(2,5-dimethyl-5,7- dihydrothieno[3,4-d]pyrimidin-4- yl)amino]-1H-indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one (single diastereomer of unknown absolute configuration)2122,5-dimethoxy-4-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)benzene-1-sulfonamide213(1R,2S)-2-(3-{[2- (dimethylamino)-6,7-dihydro-5H- cyclopenta[d]pyrimidin-4- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one2146-ethoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 2-carboxamide215(1R,2S)-5′-methoxy-2-(3-{[2- methoxy-6-(2-oxopyrrolidin-1- yl)pyridin-3-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one216(1R,2S)-5′-methoxy-2-{3-[2- methoxy-5-(morpholine-4- sulfonyl)anilino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one217(1R,2S)-5′-methoxy-2-{3-[(3- methoxy-1,5-naphthyridin-2- yl)amino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one218N,6-dimethoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N-methylpyridine-2- carboxamide2196-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N′-(propan-2- yl)pyridine-2-carbohydrazide220(1R,2S)-5′-methoxy-2-(3-{[2- methoxy-5-(2-oxopyrrolidin-1- yl)pyridin-3-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one221(1R,2S)-5′-methoxy-2-(3-{[2- methoxy-5-(3-methyl-2- oxoimidazolidin-1-yl)pyridin-3- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one2226-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 2-sulfonamide2236-ethoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 2-sulfonamide224(1R,2S)-5′-methoxy-2-{3-[2- methoxy-5-(oxane-4- sulfonyl)anilino]-1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one225(1R,2S)-2-(3-{[5- (dimethylphosphoryl)-3- methoxypyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one226(1R,2S)-2-(3-{[5-(2- hydroxypropan-2-yl)-2- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one227(1R,2S)-2-(3-{[6- (methanesulfonyl)-2-methoxy-5- methylpyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one228(1R,2S)-2-(3-{[5-(ethanesulfonyl)- 3-methoxypyridin-2-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one229(1R,2S)-2-(3-{[5- (dimethylphosphoryl)-3- methoxypyrazin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one230N-(cyclopropylmethyl)-6- methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)pyridine-2-carboxamide2316-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N-(propan-2- yl)pyridine-2-carboxamide232(1R,2S)-5′-methoxy-2-(3-{[2- methoxy-6-(3-oxa-8- azabicyclo[3.2.1]octane-8- carbonyl)pyridin-3-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one233(1R,2S)-5′-methoxy-2-{3-[(4- methoxy-1-methyl-6-oxo-1,6- dihydropyridazin-3-yl)amino]-1H- indazol-6-yl}spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one234(1R,2S)-5′-methoxy-2-(3-{[2- methoxy-5-(1,3-oxazol-2- yl)pyridin-3-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one235(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5-(3-methoxyazetidine- 1-carbonyl)pyridin-2-yl]amino}- 1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one2366-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyrazine- 2-carboxamide2376-ethoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyrazine- 2-carboxamide2386-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N,3- trimethylpyridine-2-carboxamide239Diastereomer 1: (1R,2S)-2-(3-{[6- (methanesulfinyl)-2- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one240Diastereomer 2: (1R,2S)-2-(3-{[6- (methanesulfinyl)-2- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one241(1R,2S)-2-(3-{[6- (methanesulfonyl)-4- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one242(1R,2S)-2-(3-{[5- (methanesulfonyl)-3- methoxypyrazin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one243N,N-dicyclopropyl-6-methoxy-5- ({6-[(1R,2S)-5′-methoxy-2′-oxo- 1′,2′-dihydrospiro[cyclopropane- 1,3′-indol]-2-yl]-1H-indazol-3- yl}amino)pyridine-2-carboxamide244N-(2,2-difluoroethyl)-6-methoxy- 5-({6-[(1R,2S)-5′-methoxy-2′-oxo- 1′,2′-dihydrospiro[cyclopropane- 1,3′-indol]-2-yl]-1H-indazol-3- yl}amino)pyridine-2-carboxamide245(1R,2S)-2-[3-({6-[(2R,6S)-2,6- dimethylpiperidine-1-carbonyl]-2- methoxypyridin-3-yl}amino)-1H- indazol-6-yl]-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one246(1R,2S)-5′-methoxy-2-(3-{[2- methoxy-6-(8-oxa-3- azabicyclo[3.2.1]octane-3- carbonyl)pyridin-3-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one247(1R,2S)-2-(3-{[3-chloro-5- (methanesulfonyl)pyridin-2- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one248(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5-(propane-2- sulfonyl)pyridin-2-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one249(1R,2S)-2-(3-{[6- (dimethylphosphoryl)-4- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one250N-(1,3-difluoropropan-2-yl)-6- methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)pyridine-2-carboxamide2516-chloro-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 2-carboxamide252(1R,2S)-2-{3-[(5-chloro-2-methyl- 1,3-benzoxazol-6-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one2534-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N,N-dimethylpyridine- 2-carboxamide2543-[6-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)pyrazin-2-yl]-1λ6- thietane-1,1-dione255(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5-(morpholine-4- sulfonyl)pyridin-2-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one256(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5-(8-oxa-3- azabicyclo[3.2.1]octane-3- sulfonyl)pyridin-2-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one257(1R,2S)-2-(3-{[6- (diethylphosphoryl)-2- methoxypyridin-3-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one258(1R,2S)-2-(3-{[5- (cyclopropanesulfonyl)-3- methoxypyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one259(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5-(oxane-4- sulfonyl)pyridin-2-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one2606-methoxy-5-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)pyridine-2-carbonitrile261(1R,2S)-2-{3-[5- (diethylphosphoryl)-2- methoxyanilino]-1H-indazol-6- yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one262(1R,2S)-2-(3-{[6-(ethanesulfonyl)- 4-methoxypyridin-3-yl]amino}- 1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one263(1R,25)-2-(3-{[5-(azetidine-1- carbonyl)-3-methoxypyridin-2- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one264(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5-(morpholine-4- carbonyl)pyridin-2-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one265(1R,2S)-5′-methoxy-2-{3-[(6- methoxy-2-methyl-1-oxo-2,3- dihydro-1H-isoindol-5-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one266(1R,2S)-5′-methoxy-2-{3-[(4- methoxy-2-methyl-1-oxo-2,3- dihydro-1H-isoindol-5-yl)amino]- 1H-indazol-6- yl}spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one267(1R,2S)-5′-methoxy-2-(3-{[3- methoxy-5-(1,2-oxazolidine-2- sulfonyl)pyridin-2-yl]amino}-1H- indazol-6-yl)spiro[cyclopropane- 1,3′-indol]-2′(1′H)-one268(1R,2S)-2-(3-{[5-(azetidine-1- sulfonyl)-3-methoxypyridin-2- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one2695-methoxy-6-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N-(3-methyloxetan-3- yl)pyridine-3-sulfonamide2705-methoxy-6-({6-[(1R,2S)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N-methyl-N-(3- methyloxetan-3-yl)pyridine-3- sulfonamide271(1R,2S)-2-(3-{[5-(1- hydroxyethyl)-2-methoxypyridin- 3-yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one (mixture of diastereomers)272(1R,2S)-2-{3-[2-ethoxy-4- (methanesulfonyl)anilino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one273(1R,2S)-2-(3-{[3-ethoxy-5-(4- methylpiperazine-1- sulfonyl)pyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one274(1R,2S)-2-(3-{[5-(ethanesulfonyl)- 3-ethoxypyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one2755-ethoxy-6-({6-[(1R,25)-5′- methoxy-2′-oxo-1′,2′- dihydrospiro[cyclopropane-1,3′- indol]-2-yl]-1H-indazol-3- yl}amino)-N-methylpyridine-3- sulfonamide276(1R,2S)-5′-chloro-2-(3-{[3- ethoxy-5- (methanesulfonyl)pyridin-2- yl]amino}-1H-indazol-6- yl)spiro[cyclopropane-1,3′-indol]- 2′(1′H)-one277(1R,2S)-2-{3-[(4-ethoxy-1- methyl-6-oxo-1,6- dihydropyridazin-3-yl)amino]-1H- indazol-6-yl}-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one278(1R,2S)-2-(3-{[6-(2- hydroxypropan-2-yl)-3- methoxypyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one279(1R,2S)-2-(3-{[4-ethoxy-6- (methanesulfonyl)pyridin-3- yl]amino}-1H-indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one280(1R,2S)-2-(3-{[5- (difluoromethanesulfonyl)-3- methoxypyridin-2-yl]amino}-1H- indazol-6-yl)-5′- methoxyspiro[cyclopropane-1,3′- indol]-2′(1′H)-one Further provided herein are pharmaceutical compositions comprising an amount of a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, and one or more pharmaceutically acceptable excipients. Methods of Treatment Further provided herein are methods of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or a pharmaceutical composition disclosed herein comprising a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Provided herein are such methods of treating cancer in a subject, wherein the cancer in the subject is a solid tumor. In some embodiments, the cancer is neuroblastoma, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (CNS), primary CNS lymphoma, spinal axis tumors, brain stem glioma, or pituitary adenoma. In some embodiments, the cancer in the subject expresses polo-like kinase 4 (PLK4). In some embodiments, the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering to the subject a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. In some embodiments, the cancer in the subject exhibits an overexpression of the E3 ubiquitin-protein ligase (TRIM37) protein. In some embodiments, the cancer in the subject exhibits an overexpression of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). In some embodiments, the cancer in the subject exhibits an amplification of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Further provided herein are methods of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein the cancer in the subject has been determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37) prior to administration of the compound to the subject. Further provided herein are methods of treating cancer in a subject in need thereof, wherein the cancer in the subject has been determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37), comprising administering to the subject a therapeutically effective amount of compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Further provided herein are methods of treating cancer in a subject, comprising:a. obtaining a biological sample of the cancer from the subject;b. determining whether the biological sample of the cancer overexpresses the gene that encodes the tripartite motif-containing protein 37 (TRIM37); andc. administering to the subject a therapeutically effective amount of a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, if the biological sample of the cancer is determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Further provided herein are methods of treating cancer in a subject described herein, wherein the cancer is neuroblastoma or breast cancer. Also provided herein are methods of treating cancer in a subject described herein, wherein the cancer is neuroblastoma. Also provided herein are methods of treating cancer in a subject described herein, wherein the cancer is breast cancer. Further provided herein are methods of treating cancer in a subject described herein, wherein a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered to the subject with one or more additional therapeutic agents. In some embodiments, the one or more additional therapeutic agents is selected from one or more mitotic inhibitors, alkylating agents, antimetabolites, antitumor antibiotics, anti-angiogenesis agents, topoisomerase I and II inhibitors, plant alkaloids, hormonal agents and antagonists, growth factor inhibitors, radiation, signal transduction inhibitors, such as inhibitors of protein tyrosine kinases and/or serine/threonine kinases, cell cycle inhibitors, biological response modifiers, enzyme inhibitors, antisense oligonucleotides or oligonucleotide derivatives, cytotoxics, and immuno-oncology agents. Further provided herein are methods of inhibiting polo-like kinase 4 (PLK4) in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or a pharmaceutical composition comprising a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Further provided herein are methods of inhibiting polo-like kinase 4 (PLK4) in a subject having cancer, comprising administering to the subject a therapeutically effective amount of a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or a pharmaceutical composition comprising a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering the compound or the pharmaceutical composition to the subject. Further provided herein are methods of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein the cancer in the subject is acute myeloid leukemia, myelodysplastic syndromes, chronic myelomonocytic leukemia, triple negative breast cancer, advanced breast cancer, metastatic breast cancer, or prostate cancer. In some embodiments, the cancer in the subject is acute myeloid leukemia. In some embodiments, the cancer in the subject is myelodysplastic syndromes. In some embodiments, the cancer in the subject is chronic myelomonocytic leukemia. In some embodiments, the cancer in the subject is triple negative breast cancer. In some embodiments, the cancer in the subject is advanced breast cancer. In some embodiments, the cancer in the subject is metastatic breast cancer. In some embodiments, the cancer in the subject is prostate cancer. Further provided herein are compounds of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or pharmaceutical compositions comprising a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, for use in methods of treating cancer in a subject in need thereof. In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer in the subject is a solid tumor. In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer is neuroblastoma, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (CNS), primary CNS lymphoma, spinal axis tumors, brain stem glioma, or pituitary adenoma. In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer in the subject expresses polo-like kinase 4 (PLK4). In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering the compound or the pharmaceutical composition to the subject. In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer in the subject exhibits an overexpression of the E3 ubiquitin-protein ligase (TRIM37) protein. In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer in the subject exhibits an overexpression of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer in the subject exhibits an amplification of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer in the subject has been determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37) prior to administration of the compound or the pharmaceutical composition to the subject. In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer is neuroblastoma or breast cancer. In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer is neuroblastoma. In some embodiments are provided such compounds or pharmaceutical compositions for such use, wherein the cancer is breast cancer. Further provided herein are compounds of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or pharmaceutical compositions comprising a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, for use in methods of treating cancer in a subject in need thereof wherein the cancer in the subject is acute myeloid leukemia, myelodysplastic syndromes, chronic myelomonocytic leukemia, triple negative breast cancer, advanced breast cancer, metastatic breast cancer, or prostate cancer. In some embodiments, the cancer in the subject is acute myeloid leukemia. In some embodiments, the cancer in the subject is myelodysplastic syndromes. In some embodiments, the cancer in the subject is chronic myelomonocytic leukemia. In some embodiments, the cancer in the subject is triple negative breast cancer. In some embodiments, the cancer in the subject is advanced breast cancer. In some embodiments, the cancer in the subject is metastatic breast cancer. In some embodiments, the cancer in the subject is prostate cancer. Further provided herein are compounds of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or pharmaceutical compositions comprising a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, for use in methods of inhibiting polo-like kinase 4 (PLK4) in a subject having cancer. Further provided herein are uses of a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, in the manufacture of a medicament for the treatment of cancer in a subject in need thereof. In some embodiments are provided such uses, wherein the cancer is neuroblastoma, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (CNS), primary CNS lymphoma, spinal axis tumors, brain stem glioma, or pituitary adenoma. In some embodiments the cancer in the subject expresses polo-like kinase 4 (PLK4). In some embodiments the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering the compound to the subject. In some embodiments the cancer in the subject exhibits an overexpression of the E3 ubiquitin-protein ligase (TRIM37) protein. In some embodiments the cancer in the subject exhibits an overexpression of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). In some embodiments the cancer in the subject exhibits an amplification of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). In some embodiments the cancer in the subject has been determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37) prior to administration of the compound to the subject. In some embodiments the cancer is neuroblastoma or breast cancer. In some embodiments the cancer is neuroblastoma. In some embodiments the cancer is breast cancer. Further provided herein are uses of a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, in the manufacture of a medicament for the treatment of cancer in a subject in need thereof, wherein the cancer in the subject is acute myeloid leukemia, myelodysplastic syndromes, chronic myelomonocytic leukemia, triple negative breast cancer, advanced breast cancer, metastatic breast cancer, or prostate cancer. In some embodiments, the cancer in the subject is acute myeloid leukemia. In some embodiments, the cancer in the subject is myelodysplastic syndromes. In some embodiments, the cancer in the subject is chronic myelomonocytic leukemia. In some embodiments, the cancer in the subject is triple negative breast cancer. In some embodiments, the cancer in the subject is advanced breast cancer. In some embodiments, the cancer in the subject is metastatic breast cancer. In some embodiments, the cancer in the subject is prostate cancer. In some embodiments, compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is used in combination with one or more additional anti-cancer agents. In some embodiments, the anti-cancer agent is mitoxantrone, estramustine, etoposide, vinblastine, carboplatin, vinorelbine, paclitaxel, daunomycin, darubicin, epirubicin, docetaxel, cabazitaxel, or doxorubicin. In some embodiments, the anti-cancer agent is paclitaxel, daunomycin, darubicin, epirubicin, docetaxel, cabazitaxel, or doxorubicin. In certain embodiments, the anti-cancer agent is docetaxel. In some embodiments, one or more additional anti-cancer agents may include, without limitations, surgery, radiation, or chemotherapy. The chemotherapy may be an androgen receptor antagonist, a mitotic inhibitor, an antimetabolite, a platinum-based agent. Examples of androgen receptor antagonist include, without limitations, apalutamide, flutamide, nilutamide, bicalutamide, or enzalutamide. Examples of mitotic inhibitors include, without limitations, a taxane (e.g. paclitaxel, docetaxel, paclitaxel, docetaxel, cabazitaxel, tesetaxel, or nab-paclitaxel. or nab-paclitaxel) or a vinca alkaloid (e.g., vinblastine, vincristine, vindesine, or vinorelbine). Examples of antimetabolites include, without limitations, 5-Fluorouracil, 6-mercaptopurine, capecitabine, cytarabine, floxuridine, fludarabine, gemcitabine, hydroxycarbamide, methotrexate, pemetrexed, or phototrexate. Examples of platinum-based agents include, without limitations, cisplatin, carboplatin, dicycloplatin, eptaplatin, lobaplatin, miriplatin, nedaplatin, oxaliplatin, picoplatin, satraplatin, or triplatin tetranitrate. The additional anti-cancer therapy may comprise an anti-PDL1 agent, an anti-PD1 agent or an anti CTLA-4 agent. The anti-PD-LI agent may comprise atezolizumab, avelumab, durvalumab, MPDL3280A (RG7446), MDX-1105 (BMS-936559) or BMS-935559, MSB0010718C, and MED14736. The anti-PD1 agent may comprise pembrolizumab, nivolumab, cemiplimab, partalizumab (PDR001), camrelizumab (SHR1210), sintilimab (IB1308), tislelizumab (BGB-A317), toripalimab (JS 001), dostarlimab (TSR-042, WBP-285), INCMGA00012 (MGA012), AMP-224, or AMP-514 (MEDI0680). The anti-CTLA agent may comprise ipilimumab, or tremelimumab. Methods of Treatment in Conjunction with Biomarkers Disclosed herein, in some embodiments, are methods of detecting the presence, absence, or level, of a biomarker. Such biomarkers may comprise genetic alterations in the gene encoding for certain proteins such as tripartite motif-containing protein 37 (TRIM37). The presence, absence, or level, of such biomarkers may be measured in a biological sample obtained from a subject, such as a sample of a solid tumor, such as a prostate cancer, or from a sample of a relevant biological fluid, such as a blood sample. In some instances, the methods of detection disclosed herein are useful for predicting a therapeutic response to a therapy described herein (e.g., a PLK4 inhibitor) in, monitor the treatment using the therapy of, and treating with the therapy, a proliferative disease or condition described herein in a subject. In some embodiments, the presence, or an absence, and/or a level of expression of the one or more biomarkers is detected in the sample obtained from a subject by analyzing the genetic material in the sample. In some embodiments, the genetic material is obtained from blood, serum, plasma, sweat, hair, tears, urine, and other techniques known by one of skill in the art. In some embodiments the sample comprises circulating tumor RNA (ctRNA). In some embodiments the sample comprises peripheral blood mononuclear cells (PBMCs). In some cases, the genetic material is obtained from a tumor biopsy or liquid biopsy. In some embodiments, a tumor biopsy comprises a formalin-fixed paraffin embedded biopsy, a fresh frozen biopsy, a fresh biopsy, or a frozen biopsy. In some embodiments, a liquid biopsy comprises PBMCs, circulating tumor RNA, plasma cell-free RNA, or circulating tumor cells (CTCs). Tumor biopsies can undergo additional analytic processing for sample dissociation, cell sorting, and enrichment of cell populations of interest. In some embodiments, methods of detecting a presence, absence, or level of a biomarker in the sample obtained from the subject involve detecting a nucleic acid sequence. In some cases, the nucleic acid sequence comprises deoxyribonucleic acid (DNA), such as in the case of detecting complementary DNA (cDNA) of an mRNA transcript. In some instances, the nucleic acid sequence comprises a denatured DNA molecule or fragment thereof. In some instances, the nucleic acid sequence comprises DNA selected from: genomic DNA, viral DNA, mitochondrial DNA, plasmid DNA, amplified DNA, circular DNA, circulating DNA, cell-free DNA, or exosomal DNA. In some instances, the DNA is single-stranded DNA (ssDNA), double-stranded DNA, denaturing double-stranded DNA, synthetic DNA, and combinations thereof. The circular DNA may be cleaved or fragmented. In some instances, the nucleic acid sequence comprises ribonucleic acid (RNA). In some instances, the nucleic acid sequence comprises fragmented RNA. In some instances, the nucleic acid sequence comprises partially degraded RNA. In some instances, the nucleic acid sequence comprises a microRNA or portion thereof. In some instances, the nucleic acid sequence comprises an RNA molecule or a fragmented RNA molecule (RNA fragments) selected from: a microRNA (miRNA), a pre-miRNA, a pri-miRNA, a mRNA, a pre-mRNA, a viral RNA, a viroid RNA, a virusoid RNA, circular RNA (circRNA), a ribosomal RNA (rRNA), a transfer RNA (tRNA), a pre-tRNA, a long non-coding RNA (lncRNA), a small nuclear RNA (snRNA), a circulating RNA, a cell-free RNA, an exosomal RNA, a vector-expressed RNA, an RNA transcript, a synthetic RNA, and combinations thereof. Disclosed herein, in some embodiments, the biomarker is detected by subjecting a sample obtained from the subject to a nucleic acid-based detection assay. In some instances, the nucleic acid-based detection assay comprises quantitative polymerase chain reaction (qPCR), gel electrophoresis (including for e.g., Northern or Southern blot), immunochemistry, in situ hybridization such as fluorescent in situ hybridization (FISH), cytochemistry, microarray, or sequencing. In some embodiments, the sequencing technique comprises next generation sequencing. In some embodiments, the methods involve a hybridization assay such as fluorogenic qPCR (e.g., TaqMan™, SYBR green, SYBR green I, SYBR green II, SYBR gold, ethidium bromide, methylene blue, Pyronin Y, DAPI, acridine orange, Blue View or phycoerythrin), which involves a nucleic acid amplification reaction with a specific primer pair, and hybridization of the amplified nucleic acid probes comprising a detectable moiety or molecule that is specific to a target nucleic acid sequence. In some instances, a number of amplification cycles for detecting a target nucleic acid in a qPCR assay is about 5 to about 30 cycles. In some instances, the number of amplification cycles for detecting a target nucleic acid is at least about 5 cycles. In some instances, the number of amplification cycles for detecting a target nucleic acid is at most about 30 cycles. In some instances, the number of amplification cycles for detecting a target nucleic acid is about 5 to about 10, about 5 to about 15, about 5 to about 20, about 5 to about 25, about 5 to about 30, about 10 to about 15, about 10 to about 20, about 10 to about 25, about 10 to about 30, about 15 to about 20, about 15 to about 25, about 15 to about 30, about 20 to about 25, about 20 to about 30, or about 25 to about 30 cycles. For TaqMan™ methods, the probe may be a hydrolysable probe comprising a fluorophore and quencher that is hydrolyzed by DNA polymerase when hybridized to a target nucleic acid. In some cases, the presence of a target nucleic acid is determined when the number of amplification cycles to reach a threshold value is less than 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, or 20 cycles. In some instances, hybridization may occur at standard hybridization temperatures, e.g., between about 35° C. and about 65° C. in a standard PCR buffer. An additional exemplary nucleic acid-based detection assay comprises the use of nucleic acid probes conjugated or otherwise immobilized on a bead, multi-well plate, or other substrate, wherein the nucleic acid probes are configured to hybridize with a target nucleic acid sequence. In some instances, the nucleic acid probe is specific to one or more gene products described herein. In some instances, the nucleic acid probe specific to a biomarker comprises a nucleic acid probe sequence sufficiently complementary to the polynucleotide sequence of the biomarker. In some instances, the biomarker comprises a transcribed polynucleotide sequence (e.g., RNA, cDNA). In some embodiments, the nucleic acid probe can be, for example, a full-length cDNA, or a portion thereof, such as an oligonucleotide of at least about 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides in length and sufficient to specifically hybridize under standard hybridization conditions to the target nucleic acid sequence. In some embodiments, the target nucleic acid sequence is immobilized on a solid surface and contacted with a probe, for example by running the isolated target nucleic acid sequence on an agarose gel and transferring the target nucleic acid sequence from the gel to a membrane, such as nitrocellulose. In some embodiments, the probe(s) are immobilized on a solid surface, for example, in an Affymetrix gene chip array, and the probe(s) are contacted with the target nucleic acid sequence. In some embodiments, the term “probe” with regards to nucleic acids, refers to any nucleic acid molecule that is capable of selectively binding to a specifically intended target nucleic acid sequence. In some instances, probes are specifically designed to be labeled, for example, with a radioactive label, a fluorescent label, an enzyme, a chemiluminescent tag, a colorimetric tag, or other labels or tags that are known in the art. In some instances, the fluorescent label comprises a fluorophore. In some instances, the fluorophore is an aromatic or heteroaromatic compound. In some instances, the fluorophore is a pyrene, anthracene, naphthalene, acridine, stilbene, benzoxazole, indole, benzindole, oxazole, thiazole, benzothiazole, canine, carbocyanine, salicylate, anthranilate, xanthenes dye, coumarin. Exemplary xanthene dyes include, e.g., fluorescein and rhodamine dyes. Fluorescein and rhodamine dyes include, but are not limited to 6-carboxyfluorescein (FAM), 2′7′-dimethoxy-4′5′-dichloro-6-carboxyfluorescein (JOE), tetrachlorofluorescein (TET), 6-carboxyrhodamine (R6G), N,N,N; N′-tetramethyl-6-carboxyrhodamine (TAMRA), 6-carboxy-X-rhodamine (ROX). Suitable fluorescent probes also include the naphthylamine dyes that have an amino group in the alpha or beta position. For example, naphthylamino compounds include 1-dimethylaminonaphthyl-5-sulfonate, 1-anilino-8-naphthalene sulfonate, and 2-p-toluidinyl-6-naphthalene sulfonate, 5-(2′-aminoethyl)aminonaphthalene-1-sulfonic acid (EDANS). Exemplary coumarins include, e.g., 3-phenyl-7-isocyanatocoumarin; acridines, such as 9-isothiocyanatoacridine and acridine orange; N-(p-(2-benzoxazolyl)phenyl) maleimide; cyanines, such as, e.g., indodicarbocyanine 3 (Cy3), indodicarbocyanine 5 (Cy5), indodicarbocyanine 5.5 (Cy5.5), 3-(-carboxy-pentyl)-3′-ethyl-5,5′-dimethyloxacarbocyanine (CyA); 1H, 5H, 11H, 15H-Xantheno[2,3, 4-ij: 5,6, 7-i′j′]diquinolizin-18-ium, 9-[2 (or 4)-[[[6-[2,5-dioxo-1-pyrrolidinyl)oxy]-6-oxohexyl]amino]sulfonyl]-4 (or 2)-sulfophenyl]-2,3, 6,7, 12,13, 16,17-octahydro-inner salt (TR or Texas Red); or BODIPY™ dyes. In some cases, the probe comprises FAM as the dye label. In some embodiments, detecting the one or more biomarkers, such as gene products in a predictive response signature (PRS), comprises sequencing genetic material obtained from a sample from the subject. Sequencing can be performed with any appropriate sequencing technology, including but not limited to single-molecule real-time (SMRT) sequencing, Polony sequencing, sequencing by ligation, reversible terminator sequencing, proton detection sequencing, ion semiconductor sequencing, nanopore sequencing, electronic sequencing, pyrosequencing, Maxam-Gilbert sequencing, chain termination (e.g., Sanger) sequencing, +S sequencing, or sequencing by synthesis. Sequencing methods also include next-generation sequencing, e.g., modern sequencing technologies such as Illumina sequencing (e.g., Solexa), Roche 454 sequencing, Ion torrent sequencing, and SOLiD sequencing. In some cases, next-generation sequencing involves high-throughput sequencing methods. Additional sequencing methods available to one of skill in the art may also be employed. In some instances, a number of nucleotides that are sequenced are at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 150, 200, 300, 400, 500, 2000, 4000, 6000, 8000, 10000, 20000, 50000, 100000, or more than 100000 nucleotides. In some instances, the number of nucleotides sequenced is in a range of about 1 to about 100000 nucleotides, about 1 to about 10000 nucleotides, about 1 to about 1000 nucleotides, about 1 to about 500 nucleotides, about 1 to about 300 nucleotides, about 1 to about 200 nucleotides, about 1 to about 100 nucleotides, about 5 to about 100000 nucleotides, about 5 to about 10000 nucleotides, about 5 to about 1000 nucleotides, about 5 to about 500 nucleotides, about 5 to about 300 nucleotides, about 5 to about 200 nucleotides, about 5 to about 100 nucleotides, about 10 to about 100000 nucleotides, about 10 to about 10000 nucleotides, about 10 to about 1000 nucleotides, about 10 to about 500 nucleotides, about 10 to about 300 nucleotides, about 10 to about 200 nucleotides, about 10 to about 100 nucleotides, about 20 to about 100000 nucleotides, about 20 to about 10000 nucleotides, about 20 to about 1000 nucleotides, about 20 to about 500 nucleotides, about 20 to about 300 nucleotides, about 20 to about 200 nucleotides, about 20 to about 100 nucleotides, about 30 to about 100000 nucleotides, about 30 to about 10000 nucleotides, about 30 to about 1000 nucleotides, about 30 to about 500 nucleotides, about 30 to about 300 nucleotides, about 30 to about 200 nucleotides, about 30 to about 100 nucleotides, about 50 to about 100000 nucleotides, about 50 to about 10000 nucleotides, about 50 to about 1000 nucleotides, about 50 to about 500 nucleotides, about 50 to about 300 nucleotides, about 50 to about 200 nucleotides, or about 50 to about 100 nucleotides. Disclosed herein are methods comprising: (a) providing a sample obtained from a subject with a proliferative disease or condition (e.g., cancer); (b) assaying to detect in the sample obtained from the subject a presence or absence of the relevant biomarker; and (c) detecting the presence or absence of the biomarker in the sample using the methods described herein. In some cases, a hybridization assay, such as those described herein, is used to detect the biomarker in the sample. Exemplary probe sequences that are hybridizable to a target nucleic acid sequence (e.g., one or more genes in the biomarker, such as the PRS) comprise at least 10, but no more than 100 contiguous nucleotides comprising the relevant sequence. In some cases, RNA sequencing (RNAseq) is used to detect the one or more biomarkers. Detection of the relevant biomarker, in some cases, involves amplification of the subject's nucleic acid by the polymerase chain reaction (PCR). In some embodiments, the PCR assay involves use of a pair of primers capable of amplifying at least about 10 contiguous nucleobases within a nucleic acid sequence, thereby amplifying the one or more gene products in the biomarker. In fluorogenic quantitative PCR, quantitation is based on amount of fluorescence signals (TaqMan and SYBR green). In some embodiments, the nucleic acid probe is conjugated to a detectable molecule. The detectable molecule may be a fluorophore. The nucleic acid probe may also be conjugated to a quencher. In some embodiments, the assay for detecting the presence or absence of a relevant biomarker comprises reverse-transcribing the relevant mRNA molecule to produce a corresponding complementary DNA (cDNA) molecule). In some embodiments, the assay further comprises contacting the cDNA molecule with a nucleic acid probe comprising a nucleic acid sequence that is complementary to a nucleic acid sequence of the cDNA molecule. In some embodiments, the assay comprises detecting a double-stranded hybridization product between the nucleic acid probe and the cDNA molecule. In some embodiments, the hybridization product is further amplified using a pair of primers. In some embodiments, the primers comprises a first primer with a nucleic acid sequence comprising at least 10 but not more than 50 contiguous nucleic acids within a relevant nucleic acid sequence that binds to a top strand of the double-stranded hybridization product; and a second primer with a nucleic acid sequence comprising at least 10 but not more than 50 contiguous nucleic acids within a nucleic acid sequence that is reverse complement to the relevant nucleic acid sequence that binds to a bottom strand of the double-stranded hybridization product. Disclosed herein, in some embodiments, are methods comprising preparing a complementary DNA (cDNA) library. In some embodiments, the cDNA library is sequenced using suitable sequence methodologies disclosed herein. In some embodiments, the cDNA library is labeled, a plurality of nucleic acid probes is generated, and fixed to an immobile surface (such as a microarray). In some embodiments, the plurality of nucleic acid probes is capable of hybridizing to at least about 10 contiguous nucleotides of the two or more genes in a sample obtained from the subject. In some embodiments, detecting the presence of or absence of a biomarker includes detecting a high or a low level of expression of the two or more genes as compared to a reference level. Disclosed herein, in some embodiments, genetic material is extracted from a sample obtained from a subject, e.g., a sample of blood or serum. In certain embodiments where nucleic acids are extracted, the nucleic acids are extracted using any technique that does not interfere with subsequent analysis. In certain embodiments, this technique uses alcohol precipitation using ethanol, methanol, or isopropyl alcohol. In certain embodiments, this technique uses phenol, chloroform, or any combination thereof. In certain embodiments, this technique uses cesium chloride. In certain embodiments, this technique uses sodium, potassium or ammonium acetate or any other salt commonly used to precipitate DNA. In certain embodiments, this technique utilizes a column or resin based nucleic acid purification scheme such as those commonly sold commercially, one non-limiting example would be the GenElute Bacterial Genomic DNA Kit available from Sigma Aldrich. In certain embodiments, after extraction the nucleic acid is stored in water, Tris buffer, or Tris-EDTA buffer before subsequent analysis. In an exemplary embodiment, the nucleic acid material is extracted in water. In some cases, extraction does not comprise nucleic acid purification. In certain embodiments, RNA may be extracted from cells using RNA extraction techniques including, for example, using acid phenol/guanidine isothiocyanate extraction (RNAzol B; Biogenesis), RNeasy RNA preparation kits (Qiagen) or PAXgene (PreAnalytix, Switzerland). Circulating Tumor DNA (ctDNA) and RNA (ctRNA) In some aspects, circulating tumor DNA (ctDNA) is used to assess the presence of certain DNA molecules and circulating tumor RNA (ctRNA) is used to assess the expression levels of RNA molecules, shed by the tumor into the blood stream. In some embodiments, detection of ctDNA or ctRNA is useful, for example, for detecting and diagnosing a tumor. Because tumor DNA and RNA has acquired multiple genetic mutations, leading to tumor development, ctDNA and ctRNA are not an exact match to the individual's DNA and RNA, respectively. Finding DNA and RNA with genetic differences aids in tumor detection. Diagnosing the type of tumor using ctDNA or ctRNA can reduce the need for getting a sample of the tumor tissue (tumor biopsy), which can be challenging when a tumor is difficult to access, such as a tumor in the brain or lung. In some embodiments, a decrease in the quantity of ctDNA or ctRNA suggests the solid tumor is shrinking and treatment with a compound of Formulae (I), (Ia), (Ib), (II), and (III), or a pharmaceutically acceptable salt thereof is effective. In some embodiments, a lack of ctDNA or ctRNA in the bloodstream indicates that the cancer has not returned after treatment with a compound of Formulae (I), (Ia), (Ib), (II), and (III), or a pharmaceutically acceptable salt thereof. Described herein are methods of assessing genetic alterations by ctDNA or ctRNA genomic profiling. In some embodiments, the genomic profiling is performed after each treatment cycle with a compound of Formulae (I), (Ia), (Ib), (II), and (III), or a pharmaceutically acceptable salt thereof. In some embodiments, the gene mutations indicate that the cancer is becoming resistant to the treatment with a compound of Formulae (I), (Ia), (Ib), (II), and (III), or a pharmaceutically acceptable salt thereof. In some embodiments, the lack of gene mutations indicate that the cancer is not becoming resistant to the treatment with a compound of Formulae (I), (Ia), (Ib), (II), and (III), or a pharmaceutically acceptable salt thereof. The compounds of Formulae (I), (Ia), (Ib), (II), and (III) may be administered as prodrugs. Thus certain derivatives of the compounds, which may have little or no pharmacological activity themselves can, when administered to a mammal, be converted into a compound having the desired activity, for example, by hydrolytic cleavage. Such derivatives are referred to as “prodrugs.” Prodrugs can, for example, be produced by replacing appropriate functionalities present in the compound of Formulae (I), (Ia), (Ib), (II), and (III) with certain moieties known to those skilled in the art. See, e.g. “Pro-drugs as Novel Delivery Systems”, Vol. 14, ACS Symposium Series (T Higuchi and W Stella) and “Bioreversible Carriers in Drug Design”. Pergamon Press, 1987 (ed. E B Roche, American Pharmaceutical Association), the disclosures of which are incorporated herein by reference in their entireties. Some examples of such prodrugs include: an ester moiety in the place of a carboxylic acid functional group; an ether moiety or an amide moiety in place of an alcohol functional group; and an amide moiety in place of a primary or secondary amino functional group. Examples of replacement groups are known to those of skill in the art. See, e.g. “Design of Prodrugs” by H Bundgaard (Elsevier, 1985), the disclosure of which is incorporated herein by reference in its entirety. Salts of the present invention can be prepared according to methods known to those of skill in the art. Examples of salts include, but are not limited to, acetate, acrylate, benzenesulfonate, benzoate (such as chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, and methoxybenzoate), bicarbonate, bisulfate, bisulfite, bitartrate, borate, bromide, butyne-1,4-dioate, calcium edetate, camsylate, carbonate, chloride, caproate, caprylate, clavulanate, citrate, decanoate, dihydrochloride, dihydrogenphosphate, edetate, edisylate, estolate, esylate, ethylsuccinate, formate, fumarate, gluceptate, gluconate, glutamate, glycollate, glycollylarsanilate, heptanoate, hexyne-1,6-dioate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, .gamma.-hydroxybutyrate, iodide, isobutyrate, isothionate, lactate, lactobionate, laurate, malate, maleate, malonate, mandelate, mesylate, metaphosphate, methane-sulfonate, methylsulfate, monohydrogenphosphate, mucate, napsylate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, nitrate, oleate, oxalate, pamoate (embonate), palmitate, pantothenate, phenylacetates, phenylbutyrate, phenylpropionate, phthalate, phosphate/diphosphate, polygalacturonate, propanesulfonate, propionate, propiolate, pyrophosphate, pyrosulfate, salicylate, stearate, subacetate, suberate, succinate, sulfate, sulfonate, sulfite, tannate, tartrate, teoclate, tosylate, triethiodode, and valerate salts. The compounds of Formulae (I), (Ia), (Ib), (II), and (III) that are basic in nature are capable of forming a wide variety of different salts with various inorganic and organic acids. Although such salts must be pharmaceutically acceptable for administration to animals, it is often desirable in practice to initially isolate the compound of the present invention from the reaction mixture as a pharmaceutically unacceptable salt and then simply convert the latter back to the free base compound by treatment with an alkaline reagent and subsequently convert the latter free base to a pharmaceutically acceptable acid addition salt. The acid addition salts of the base compounds of this invention can be prepared by treating the base compound with a substantially equivalent amount of the selected mineral or organic acid in an aqueous solvent medium or in a suitable organic solvent, such as methanol or ethanol. Upon evaporation of the solvent, the desired solid salt is obtained. The desired acid salt can also be precipitated from a solution of the free base in an organic solvent by adding an appropriate mineral or organic acid to the solution. The compounds of Formulae (I), (Ia), (Ib), (II), and (III) that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. Examples of such salts include the alkali metal or alkaline-earth metal salts and particularly, the sodium and potassium salts. These salts are all prepared by conventional techniques. The chemical bases which are used as reagents to prepare the pharmaceutically acceptable base salts of this invention are those which form non-toxic base salts with the acidic compounds of the present invention. Such non-toxic base salts include those derived from such pharmacologically acceptable cations as sodium, potassium calcium and magnesium, etc. These salts can be prepared by treating the corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations, and then evaporating the resulting solution to dryness, preferably under reduced pressure. Alternatively, they may also be prepared by mixing lower alkanolic solutions of the acidic compounds and the desired alkali metal alkoxide together, and then evaporating the resulting solution to dryness in the same manner as before. In either case, stoichiometric quantities of reagents are preferably employed in order to ensure completeness of reaction and maximum yields of the desired final product. If the compound of Formulae (I), (Ia), (Ib), (II), and (III) is a base, the desired salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like. If the compound of Formulae (I), (Ia), (Ib), (II), and (III) is an acid, the desired salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like. Illustrative examples of suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium. If the compound of Formulae (I), (Ia), (Ib), (II), and (III) is a solid, it is understood by those skilled in the art that the compounds or salts thereof may exist in different crystal or polymorphic forms, all of which are intended to be within the scope of the present invention and specified formulas. Also provided herein are isotopically-labeled compounds of Formulae (I), (Ia), (Ib), (II), and (III), wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as2H and3H, carbon, such as11C,12C and14C, chlorine, such as36Cl, fluorine, such as18F, iodine, such as123I and125I, nitrogen, such as13N and15N, oxygen, such as15O,17O and18O, phosphorus, such as32P, and sulfur, such as35S. Certain isotopically-labeled compounds of the invention, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium,3H, and carbon-14,14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium,2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. Substitution with positron emitting isotopes, such as11C,18F,15O and13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of Formulae (I), (Ia), (Ib), (II), and (III) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed. In one aspect, the compositions of compounds of Formulae (I), (Ia), (Ib), (II), and (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, described herein are used for the treatment of cancer in a subject. In one embodiment, such compositions are in the form of suitable dosage forms. Suitable dosage forms include, for example, liquids, suspensions, powders for reconstitution, tablets, pills, sachets, or capsules of hard or soft gelatin (See, e.g., Remington: The Science and Practice of Pharmacy (Gennaro, 21st Ed. Mack Pub. Co., Easton, PA (2005)). The compounds of Formulae (I), (Ia), (Ib), (II), and (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, may be formulated into pharmaceutical compositions as described below in any pharmaceutical form recognizable to the skilled artisan as being suitable. Pharmaceutical compositions of the invention comprise a therapeutically effective amount of at least one compound of the present invention and an inert, pharmaceutically acceptable carrier or diluent. The pharmaceutical carriers employed may be either solid or liquid. Exemplary solid carriers are lactose, sucrose, tale, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid, and the like. Exemplary liquid carriers are syrup, peanut oil, olive oil, water, and the like. Similarly, the inventive compositions may include time-delay or time-release material known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate or the like. Further additives or excipients may be added to achieve the desired formulation properties. For example, a bioavailability enhancer, such as Labrasol™, Gelucire™ or the like, or formulator, such as CMC (carboxymethylcellulose), PG (propyleneglycol), or PEG (polyethyleneglycol), may be added. Gelucire™, a semi-solid vehicle that protects active ingredients from light, moisture, and oxidation, may be added, e.g., when preparing a capsule formulation. If a solid carrier is used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form, or formed into a troche or lozenge. The amount of solid carrier may vary, but generally will be from about 25 mg to about 1 g. If a liquid carrier is used, the preparation may be in the form of syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampoule or vial or non-aqueous liquid suspension. If a semi-solid carrier is used, the preparation may be in the form of hard and soft gelatin capsule formulations. The inventive compositions are prepared in unit-dosage form appropriate for the mode of administration, e.g. parenteral or oral administration. To obtain a stable water-soluble dose form, a salt of a compound of the present invention may be dissolved in an aqueous solution of an organic or inorganic acid, such as a 0.3 M solution of succinic acid or citric acid. If a soluble salt form is not available, the agent may be dissolved in a suitable co-solvent or combinations of co-solvents. Examples of suitable co-solvents include alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from 0 to 60% of the total volume. In an exemplary embodiment, a compound of the present invention is dissolved in DMSO and diluted with water. The composition may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle such as water or isotonic saline or dextrose solution. Proper formulation is dependent upon the route of administration selected. For injection, the agents of the compounds of the present invention may be formulated into aqueous solutions, preferably in physiologically compatible buffers such as Hanks solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. For oral administration, the compounds can be formulated by combining the active compounds with pharmaceutically acceptable carriers known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a subject to be treated. Pharmaceutical preparations for oral use can be obtained using a solid excipient in admixture with the active ingredient (agent), optionally grinding the resulting mixture, and processing the mixture of granules after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include: fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; and cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as crosslinked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, polyvinyl pyrrolidone, Carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active agents. Pharmaceutical preparations that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with fillers such as lactose, binders such as starches, and/or lubricants such as tale or magnesium stearate, and, optionally, stabilizers. In soft capsules, the active agents may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. For administration intranasally or by inhalation, the compounds for use according to the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of gelatin for use in an inhaler or insufflator and the like may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit-dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active agents may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. sterile pyrogen-free water, before use. In addition to the formulations described above, the compounds of the present invention may also be formulated as a depot preparation. Such long-acting formulations may be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion-exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. A pharmaceutical carrier for hydrophobic compounds is a co-solvent system comprising benzyl alcohol, a non-polar surfactant, a water-miscible organic polymer, and an aqueous phase. The co-solvent system may be a VPD co-solvent system. VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the non-polar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. The VPD co-solvent system (VPD: 5 W) contains VPD diluted 1:1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. The proportions of a co-solvent system may be suitably varied without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity non-polar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may be substituted for dextrose. Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide (DMSO) also may be employed, although usually at the cost of greater toxicity due to the toxic nature of DMSO. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed. The pharmaceutical compositions also may comprise suitable solid- or gel-phase carriers or excipients. These carriers and excipients may provide marked improvement in the bioavailability of poorly soluble drugs. Examples of such carriers or excipients include calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Furthermore, additives or excipients such as Gelucire™, Capryol™, Labrafil™, Labrasol™, Lauroglycol™, Plurol™, Peceol™, Transcutol™ and the like may be used. Further, the pharmaceutical composition may be incorporated into a skin patch for delivery of the drug directly onto the skin. It will be appreciated that the actual dosages of the agents of this invention will vary according to the particular agent being used, the particular composition formulated, the mode of administration, and the particular site, host, and disease being treated. Those skilled in the art using conventional dosage-determination tests in view of the experimental data for a given compound may ascertain optimal dosages for a given set of conditions. For oral administration, an exemplary daily dose generally employed will be from about 0.001 to about 1000 mg/kg of body weight, with courses of treatment repeated at appropriate intervals. Furthermore, the pharmaceutically acceptable formulations of the present invention may contain a compound of the present invention, or a salt or solvate thereof, in an amount of about 10 mg to about 2000 mg, or from about 10 mg to about 1500 mg, or from about 10 mg to about 1000 mg, or from about 10 mg to about 750 mg, or from about 10 mg to about 500 mg, or from about 25 mg to about 500 mg, or from about 50 to about 500 mg, or from about 100 mg to about 500 mg. Additionally, the pharmaceutically acceptable formulations of the present invention may contain a compound of the present invention, or a salt or solvate thereof, in an amount from about 0.5 w/w % to about 95 w/w %, or from about 1 w/w % to about 95 w/w %, or from about 1 w/w % to about 75 w/w %, or from about 5 w/w % to about 75 w/w %, or from about 10 w/w % to about 75 w/w %, or from about 10 w/w % to about 50 w/w %. The compounds of the present invention, or salts or solvates thereof, may be administered to a mammal suffering from abnormal cell growth, such as a human, either alone or as part of a pharmaceutically acceptable formulation, once a day, twice a day, three times a day, or four times a day, or even more frequently. Those of ordinary skill in the art will understand that with respect to the compounds Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, the particular pharmaceutical formulation, the dosage, and the number of doses given per day to a mammal requiring such treatment, are all choices within the knowledge of one of ordinary skill in the art and can be determined without undue experimentation. Dosages of compositions described herein can be determined by any suitable method. Maximum tolerated doses (MTD) and maximum response doses (MRD) for the compounds of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, can be determined via established animal and human experimental protocols as well as in the examples described herein. For example, toxicity and therapeutic efficacy of the compounds of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50. The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with minimal toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. Additional relative dosages, represented as a percent of maximal response or of maximum tolerated dose, are readily obtained via the protocols. In some embodiments, the amount of the compounds of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, comprising a formulation that corresponds to such an amount varies depending upon factors such as the particular salt or form, disease condition and its severity, the identity (e.g., age, weight, sex) of the subject or host in need of treatment, but can nevertheless be determined according to the particular circumstances surrounding the case, including. e.g., the specific agent being administered, the liquid formulation type, the condition being treated, and the subject or host being treated. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered in an amount between about 10 mg to 500 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or a pharmaceutically acceptable salt thereof) is administered in an amount between about 100 mg to about 400 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof is administered in an amount between about 150 mg to about 350 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof is administered in an amount between about 150 mg to about 300 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered in an amount between about 160 mg to about 300 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered in an amount of about 160 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered in an amount of about 200 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered in an amount of about 240 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered in an amount of about 280 mg per day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered in an amount of about 320 mg per day. In general, an appropriate dose and treatment regimen provides the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (e.g., an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity. Optimal doses are generally determined using experimental models and/or clinical trials. The optimal dose depends upon the body mass, weight, or blood volume of the subject. In general, an appropriate dose and treatment regimen provides the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (e.g., an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity. Optimal doses are generally determined using experimental models and/or clinical trials. The optimal dose depends upon the body mass, weight, or blood volume of the subject. In certain embodiments wherein the subject's condition does not improve, upon the doctor's discretion the administration of a composition described herein are administered chronically, that is, for an extended period of time, including throughout the duration of the subject's life in order to ameliorate or otherwise control or limit the symptoms of the subject's disease. In other embodiments, administration of a composition continues until complete or partial response of a disease. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered to a subject in need thereof once a day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, is administered to a subject in need thereof twice a day. In some embodiments, a compound of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof is administered to a subject in need thereof three times a day. In some instances, the methods described herein comprise administering the compositions and formulations comprising the compounds of Formulae (I), (Ia), (Ib), (II), (III) in combination with one or more additional therapeutic agents, to the subject or subject in need thereof in multiple cycles repeated on a regular schedule with periods of rest in between each cycle. For example, in some instances, treatment given for one week followed by three weeks of rest is one treatment cycle. The length of a treatment cycle depends on the treatment being given. In some embodiments, the length of a treatment cycle ranges from two to six weeks. In some embodiments, the length of a treatment cycle ranges from three to six weeks. In some embodiments, the length of a treatment cycle ranges from three to four weeks. In some embodiments, the length of a treatment cycle is three weeks (or 21 days). In some embodiments, the length of a treatment cycle is four weeks (28 days). In some embodiments, the length of a treatment cycle is 56 days. In some embodiments, a treatment cycle lasts one, two, three, or four weeks. In some embodiments, a treatment cycle lasts three weeks. In some embodiments, a treatment cycle lasts four weeks. The number of treatment doses scheduled within each cycle also varies depending on the drugs being given. Kits and Articles of Manufacture Disclosed herein, in certain embodiments, are kits and articles of manufacture for use with one or more methods and compositions described herein. Such kits include a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. In one embodiment, the containers are formed from a variety of materials such as glass or plastic. A kit typically includes labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included. In one embodiment, a label is on or associated with the container. In one embodiment, a label is on a container when letters, numbers or other characters forming the label are attached, molded, or etched into the container itself, a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In one embodiment, a label is used to indicate that the contents are to be used for a specific therapeutic application. The label also indicates directions for use of the contents, such as in the methods described herein. In certain embodiments, the pharmaceutical compositions are presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein. The pack, for example, contains metal or plastic foil, such as a blister pack. In one embodiment, the pack or dispenser device is accompanied by instructions for administration. In one embodiment, the pack or dispenser is also accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, is the labeling approved by the U.S. Food and Drug Administration for drugs, or the approved product insert. In one embodiment, compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are also prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Methods of Preparation Compounds of Formulae (I), (Ia), (Ib), (II), or (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, may be prepared using the reaction routes and synthetic schemes described below, employing the techniques available in the art using starting materials that are readily available. The preparation of certain embodiments of the present invention is described in detail in the following examples, but those of ordinary skill in the art will recognize that the preparations described may be readily adapted to prepare other embodiments of the present invention. For example, the synthesis of non-exemplified compounds according to the invention may be performed by modifications apparent to those skilled in the art, e.g. by appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions. Alternatively, other reactions referred to herein or known in the art will be recognized as having adaptability for preparing other compounds of the invention. The compounds of Formula (I) may be prepared from Compounds of Formula (IV), wherein R2, R3, R4a, R4b, R4c, R5, R6, R7, R8a, R8b, R8c, and R8dare as defined herein, by allowing the compounds to react with compounds of Formula (V), wherein A, R1, and n are as defined herein, and wherein LG is a leaving group. LG that may be used include halogens, such as chloro, bromo, and iodo. The reaction of the compounds of Formula (IV) with compounds of Formula (V) may be conducted using methods known to those of ordinary skill in the art. For example, the reaction of the compounds of Formula (IV) with compounds of Formula (V) may be conducted in aprotic solvents, such as acetonitrile, DMF, and the like, protic solvents, such as water or alcohols, mixtures of protic and aprotic solvents, such as mixtures of acetonitrile and water, at temperatures in the range from 25° C. to 200° C., and in the presence of an acid or a base. Compounds of Formula (V) may be prepared by methods disclosed herein and/or by methods known to those of ordinary skill in the art. Alternatively, compounds of Formula (I) may be prepared by allowing compounds of Formula (VI), wherein R3, R4a, R4b, R4c, R5, R6, R7, R8a, R8b, R8c, and R8dare as defined herein, and Hal is a halogen, such as bromo or iodo, by allowing the compounds to react with compounds of Formula (VII), wherein A, R1, R2, and n are as defined herein. Such reactions may be performed in the presence of a catalytic amount of a palladium-containing compound, such as palladium(0) bis(dibenzylideneacetone) (also known as Pd(dba)2), a phosphate ligand, such as 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (also known as Xantphos), a base, and in an aprotic solvent. The base may be selected from an organic base, such as a tertiary amine, for example triethyl amine, or an inorganic base, for example cesium carbonate. The aprotic solvent may be, for example, toluene. The reactions of the compounds of Formula (VI) with the compounds of Formula (VII) may be conducted at temperatures in the range from 25° C. to 200° C., for example such reactions may be conducted in toluene at a temperature of 100° C. The compounds of Formula (VII) are commercially available, or may be prepared by methods known to those having ordinary skill in the art, or by methods similar to those set forth herein. Compounds of Formula (VI) may be prepared by methods known to those having ordinary skill in the art. For example, the compound of Formula (IV) (1R,2S)-2-(3-bromo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one may be prepared according to the scheme set forth below. Other compounds of Formula (VI) may be prepared by methods known to those of skill in the art by modifications apparent to those skilled in the art, e.g. by using different starting materials, appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions. Similarly, (1R,2S)-2-(3-Iodo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one may be prepared by allowing (1R,2S)-2-(1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one to react with iodine in DMF and methanol in the presence of potassium carbonate as set forth below. Compounds of Formula (IV) may be prepared from compounds of Formula (VI) by methods known to those having ordinary skill in the art. For example, tert-butyl (1R,2S)-2-(3-amino-1-(tert-butoxycarbonyl)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate may be prepared from (1R,2S)-2-(3-bromo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one as set forth below. Compounds such as tert-butyl (1R,2S)-2-(3-amino-1-(tert-butoxycarbonyl)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate may be allowed to react with compounds of Formula (V) as described herein, followed by deprotection of the Boc groups using an acid, such as trifluoroacetic acid, to provide compounds of Formula (I). For example, tert-butyl (1R,2S)-2-(3-amino-1-(tert-butoxycarbonyl)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate may be allowed to react with 4-chloro-5-methoxypyrimidine to afford (1R,2S)-5′-methoxy-2-(3-((5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indolin]-2′-one. In the following Preparations and Examples, “Ac” means acetyl, “ACN” and “MeCN” mean acetonitrile, “Me” means methyl, “Et” means ethyl, “Ph” means phenyl, “BOC”, “Boc” or “boc” means N-tert-butoxycarbonyl, “DCM” (CH2Cl2) means methylene chloride, “DIPEA” or “DIEA” means diisopropyl ethyl amine, “DMA” means N,N-dimethylacetamide, “DMF” means N—N-dimethyl formamide, “DMSO” means dimethylsulfoxide, “DPPP” means 1,3-bis(diphenylphosphino)propane, “HOAc” means acetic acid, “IPA” means isopropyl alcohol. “min” means minute. “NMP” means 1-methyl 2-pyrrolidinone, “TEA” means triethyl amine, “TFA” means trifluoroacetic acid, “DCM” means dichloromethane, “EtOAc” and “EA” mean ethyl acetate, “MgSO4” means magnesium sulphate, “Na2SO4” means sodium sulphate, “MeOH” means methanol, “Et2O” means diethyl ether, “EtOH” means ethanol, “H2O” means water, “HCl” means hydrochloric acid, “K2CO3” means potassium carbonate, “THF” means tetrahydrofuran, “DBU” means 1,8-diazabicyclo[5.4.0]undec-7-ene, “LiHMDS” or “LHMDS” means lithium hexamethyldisilazide, “TBME” or “MTBE” means tert-butyl methyl ether, “LDA” means lithium diisopropylamide, “N” means Normal, “M” means molar, “mL” means milliliter, “mmol” means millimoles, “μmol” means micromoles, “eq.” means equivalent, “° C.” means degrees Celsius, “Pa” means pascals, “rt” or “RT” means room temperature, “h” means hours, “satd.” means saturated, “aq” means aqueous, “anhyd.” or “anh.” means anhydrous, “MBTE” means methyl tert-butyl ether, “PE” means petroleum ether, and “TBSCl” means tert-butyldimethylsilyl chloride. EXAMPLES Intermediate 1. (1R,2S)-2-(3-Amino-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. (E)-3-(3-Fluoro-4-isocyanobenzylidene)-5-methoxyindolin-2-one A round bottom flask was charged with 5-methoxyoxindole (5.00 g, 30.6 mmol), 4-cyano-3-fluorobenzaldehyde (4.57 g, 30.6 mmol), piperidine (835 μL, 8.40 mmol) and ethanol (120 mL). The reaction was refluxed for 4 h and was stirred for 16 h at rt. The reaction was cooled to 0° C. and the resulting precipitate was collected by filtration and dried to give the title compound (5.10 g, 57%) as a dark red solid. m/z (ESI, +ve ion)=295.0 [M+H]+. Step B. racemic-2-Fluoro-4-((1R,2S)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)benzonitrile To a solution of trimethylsulfoxonium iodide (4.20 g, 19.1 mmol) in anhydrous DMF (173 mL) under nitrogen was added sodium hydride (60% dispersion in oil) (81.5 mg, 2.04 mmol) at 0° C. The mixture was stirred for 15 minutes after which (E)-3-(3-fluoro-4-isocyanobenzylidene)-5-methoxyindolin-2-one (5.10 g, 17.3 mmol) was added to the solution and the reaction was stirred for 1 h at rt. The solution was quench with satd. aq. ammonium chloride solution and extracted with EtOAc. The organic layer was then washed with brine, dried with anhyd. Na2SO4, filtered and concentrated under vacuum. The crude mixture was purified by column chromatography (10% to 65% EtOAc/heptanes, gradient elution), affording the title compound (1.50 g, 28%) as an orange solid. NOESY NMR experiment confirmed relative stereochemistry. m/z (ESI, +ve ion)=309.0 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 8.14 (s, 1H), 7.63-7.54 (m, 1H), 7.13-7.08 (m, 2H), 6.85 (d, J=8.5 Hz, 1H), 6.67 (dd, J=8.5, 2.5 Hz, 1H), 5.55 (d, J=2.4 Hz, 1H), 3.55 (s, 3H), 3.29 (t, J=8.5 Hz, 1H), 2.26 (dd, J=9.0, 5.0 Hz, 1H), 1.94 (dd, J=8.0, 5.0 Hz, 1H). The corresponding diastereoisomer was found to be less polar and was first to elute in the given conditions. m/z (ESI, +ve ion)=309.0 [M+H]+. 1 H NMR (400 MHz, CDCl3) δ 8.08 (s, 1H), 7.52 (dd, J=7.8, 6.9 Hz, 1H), 7.21 (s, 1H), 7.19 (s, 1H), 6.78 (d, J=1.5 Hz, 2H), 6.54 (s, 1H), 3.81 (s, 3H), 3.07 (t, J=8.7 Hz, 1H), 2.34 (dd, J=8.5, 5.3 Hz, 1H), 2.12 (dd, J=8.9, 5.3 Hz, 1H). Step C. (1R,2S)-2-(3-Amino-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one In a 20 mL vial was dissolved 2-fluoro-4-((1R,2S)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)benzonitrile (20.0 mg, 64.9 μmol) in tert-amyl alcohol (10.0 mL) and hydrazine hydrate solution (50.0 μL, 1.58 mmol) was subsequently added. The reaction was reflux for 16 h. The reaction was cooled to rt and silica was directly added to the mixture and concentrated. The product was purified by column chromatography (0 to 20% MeOH/DCM, gradient elution), affording the title compound (60.0 mg, 58%) as colorless oil. m/z (ESI, +ve ion)=321.1 [M+H]+. Intermediate 2. (1R,2S)-2-(1H-Indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 1-Benzyl-5-methoxyindoline-2,3-dione Benzyl bromide (9.65 mL, 79.7 mmol) was added to a mixture of 5-methoxyisatin (12.0 g, 66.4 mmol) and potassium carbonate (27.5 g, 199 mmol) in acetonitrile (250 mL). The mixture was stirred for 15 h at 80° C. then cooled to rt. The mixture was filtered and the filtrate concentrated. It was diluted with water (300 mL) and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine, then dried (Na2SO4), filtered and concentrated. The resulting solid was triturated with heptane, filtered, and washed with heptane, affording the title compound (18.2 g, quantitative yield) as a solid. m/z (ESI, +ve ion)=268.1 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 7.38-7.27 (m, 5H), 7.15 (d, J=2.7 Hz, 1H), 7.02 (dd, J=8.6, 2.7 Hz, 1H), 6.67 (d, J=8.6 Hz, 1H), 4.90 (s, 2H), 3.77 (s, 3H). Step B. 1-Benzyl-5-methoxyindolin-2-one Hydrazine monohydrate (8.64 mL, 107 mmol) was added to a mixture of 1-benzyl-5-methoxyindoline-2,3-dione (18.2 g, 68.1 mmol) in DMSO (44.1 mL). The mixture was stirred for 5 h at 140° C. then cooled to rt. The mixture was diluted with water (300 mL) and extracted with EtOAc (3×200 mL). The combined organic layers were washed with 1 M H2SO4, brine (twice), then dried (Na2SO4), filtered and concentrated, affording the title compound (14.0 g, 81%) as a dark oil. m/z (ESI, +ve ion)=254.1 [M+H]+. 1 H NMR (400 MHz, CDCl3) δ 7.38-7.22 (m, 5H), 6.90-6.86 (m, 1H), 6.68 (dd, J=8.5, 2.6 Hz, 1H), 6.60 (d, J=8.5 Hz, 1H), 4.89 (s, 2H), 3.75 (s, 3H), 3.61 (s, 2H). Step C. 1-Benzyl-6-bromo-1H-indazole Potassium tert-butoxide (20.5 g, 179 mmol) was added to a mixture of 6-bromo-1H-indazole (30.0 g, 152 mmol) in DMSO (149 mL). The mixture was stirred for 10 min then benzyl chloride (20.8 mL, 179 mmol) was slowly added at 0° C. The mixture was stirred at rt for 3 h then diluted with saturated aqueous NH4Cl (400 mL) and extracted with MTBE (3×200 mL). The combined organic layers were washed with brine twice, then dried (Na2SO4), filtered and concentrated to give crude material as a mixture of 1-benzyl-6-bromo-1H-indazole and 2-benzyl-6-bromo-2H-indazole. Benzyl bromide (37.7 mL, 311 mmol) was added to a mixture of 1-benzyl-6-bromo-1H-indazole and 2-benzyl-6-bromo-2H-indazole (31 g, 108 mmol). The mixture was stirred neat at 150° C. After 6 h, benzyl bromide was removed by distillation under high vacuum (vacuum pump) at 130° C. The residue was triturated in heptanes, then filtered and washed with heptanes. The crude material was put on the high vacuum overnight, affording the tittle compound (20.6 g, 67%) as a solid. m/z (ESI, +ve ion)=287.0 [M+H]+. Step D. 1-Benzyl-6-vinyl-1H-indazole A mixture of 1-benzyl-6-bromo-1H-indazole (6.33 g, 22.0 mmol) and potassium carbonate (9.14 g, 66.1 mmol) in previously degassed (nitrogen bubbled through) DME/water (3:1) (70.0 mL) was purged with nitrogen and nitrogen was further bubbled through the reaction mixture. Vinylboronic acid pinacol ester (4.82 mL, 27.6 mmol) was added, followed by dichlorobis(triphenylphosphine)palladium (II) (774 mg, 1.10 mmol) and the mixture was heated to 80° C. overnight. The mixture was diluted with heptanes and washed with water (3×) and brine. The organic phase was dried with Na2SO4, filtered and concentrated. The crude product was purified by column chromatography (0 to 10% EtOAc/hexanes, gradient elution) to afford the title compound 4D (3.80 g, 74%). m/z (ESI, +ve ion)=235.4 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 8.01 (d, J=0.9 Hz, 1H), 7.73-7.64 (m, 1H), 7.36-7.23 (m, 5H), 7.23-7.16 (m, 2H), 6.80 (dd, J=17.6, 10.9 Hz, 1H), 5.80 (dd, J=17.5, 0.7 Hz, 1H), 5.60 (s, 2H), 5.30 (dd, J=10.9, 0.6 Hz, 1H). Step E. (S)-1-(1-Benzyl-1H-indazol-1-yl)ethane-1,2-diol To a 500 mL flask was added AD-mix-alpha (83.7 g, 59.8 mmol) and t-BuOH/water (1:1) (598 mL) forming a clear biphasic mixture on stirring. The reaction mixture was cooled with an ice bath to 0° C. before adding 1-benzyl-6-vinyl-1H-indazole (14.0 g, 59.8 mmol). The resulting mixture was vigorously stirred at 0° C. and let warm to room temperature with the ice bath slowly warming up. The reaction mixture was stirred for 9 h. The reaction was quench by the portion-wise addition of 92 g of sodium sulfite. The reaction mixture was stirred overnight. The reaction mixture was diluted with brine and DCM and filtered through a pad of Celite. The filtrate was extracted with DCM (4×) and the combined organic layers were dried over MgSO4, filtered and concentrated. The crude product was recrystallized from toluene (80 mL) to afford the title compound (12.2 g, 76%) as a white solid. m/z (ESI, +ve ion)=269.2 [M+H]+. 99.1% ee. Step F. (S)-1-(1-Benzyl-1H-indazol-6-yl)ethane-1,2-diyl dimethanesulfonate A solution of (S)-1-(1-Benzyl-1H-indazol-6-yl)ethane-1,2-diol (12.2 g, 45.5 mmol) and triethylamine (16.0 mL, 114 mmol) in DCM (227 mL) was cooled in an ice bath and treated by slow addition of methanesulfonyl chloride (7.77 mL, 100 mmol) over 15 minutes. The internal temperature increased to a maximum of 11° C. The resulting mixture was stirred at 0° C. After 6 h, LCMS showed 10% mono mesylated product. 0.400 mL of methanesulfonyl chloride and 0.600 mL of triethylamine were added. The mixture was stirred 1 h and upon completion, diluted with DCM (500 mL) and 1 M aqueous HCl (200 mL) at 0° C. The layers were separated and the organic layer was washed with saturated aqueous NaHCO3(2×200 mL), brine (200 mL), then dried (Na2SO4), filtered and concentrated. The crude material was passed through a small pad of Celite eluting with a mixture of DCM/Et2O (1:1). Removal of the solvents gave a white solid. The solid was triturated in Et2O (40 mL) and the precipitate was collected by filtration, affording the title compound (17.5 g, 91%) as white crystalline solid. m/z (ESI, +ve ion)=425.0 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 8.08 (s, 1H), 7.81 (d, J=8.3 Hz, 1H), 7.41 (s, 1H), 7.35-7.27 (m, 3H), 7.18 (dd, J=17.3, 7.5 Hz, 3H), 5.89 (dd, J=8.6.3.2 Hz, 1H), 5.66 (d, J=15.8 Hz, 1H), 5.60 (d, J=15.8 Hz, 1H), 4.53 (dd, J=11.9, 8.6 Hz, 1H), 4.40 (dd, J=11.9, 3.3 Hz, 1H), 3.05 (s, 3H), 2.75 (s, 3H). Step G. (1R,2S)-1′-Benzyl-2-(1-benzyl-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one A solution of (S)-1-(1-benzyl-1H-indazol-6-yl)ethane-1,2-diyl dimethanesulfonate (2.03 g, 8.01 mmol) in dry THF (80 mL) under nitrogen was cooled in an ice bath. Sodium hydride (673 mg, 16.8 mmol) was added portion-wise and the mixture was stirred at 0° C. for 15 min. A solution of 1-benzyl-5-methoxyindolin-2-one (3.40 g, 8.01 mmol) in dry THF (50 mL) was added dropwise with an addition funnel. The reaction mixture was stirred for 3 h at 0° C. The reaction was quench with satd. NH4Cl solution, diluted with water, and extracted with EtOAc (3×). The organic layer was dried with anhyd. MgSO4and concentrated to a crude product. The crude product was triturated with 3:1 hexanes/EtOAc, affording the title compound (2.10 g, 54%) as an orange solid. m/z (ESI, +ve ion)=486.2 [M+H]+. Step H. (1R,2S)-2-(1H-Indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one To a round-bottom flask charged with a stir bar was added (1R,2S)-1′-Benzyl-2-(1-benzyl-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2-one (4.00 g, 8.24 mmol) in THF (118 mL). The solution was cooled to 0° C. and potassium tert-butoxide (23.0 mL, 165 mmol) was added portion wise over 20 min and then DMSO (10.7 mL) was added. Oxygen was bubbled through the solution at 0° C. for 1 h. The reaction was quenched with satd. aqueous NH4Cl at 0° C. and diluted with EtOAc (50 mL). The mixture was washed with satd. aqueous NH4Cl (1×) and extracted with EtOAc (2×). The organic phase was dried over Na2SO4, filtered, and concentrated under vacuum. The crude was triturated in Et2O and recrystallized from ethanol, affording the title compound (2.56 g, 56%). m/z (ESI, +ve ion)=306.4 [M+H]+. Intermediate 3: (1R,2S)-2-(3-Bromo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one In a flask. (1R,2S)-2-(1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (4.49 g, 12.5 mmol) was dissolved in DMF (16.7 mL) and NBS (2.70 g, 15.0 mmol) dissolved in DMF (8.33 mL) was added dropwise at 0° C. The reaction was stirred for 2 h at rt. The reaction was quenched with an aqueous solution of Na2S2O3and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine, dried (Na2SO4), filtered and concentrated. The crude was purified by column chromatography (40 to 100% EtOAc/hexanes, gradient elution), affording the title compound (3.12 g, 65%). m/z (ESI, +ve ion)=384.0, 386.0 [M+H]+. Intermediate 4: Tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate 4-Dimethylaminopyridine (79.8 mg, 640 μmol) was added to a solution of triethylamine (3.61 mL, 25.6 mmol), di-tert-butyl dicarbonate (4.0 mL, 17.3 mmol) and (1R,2S)-2-(3-bromo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (2.46 g, 6.40 mmol) in DCM (24 mL). The solution was stirred at room temperature for 16 h. LCMS showed incomplete conversion. Di-tert-butyl dicarbonate (0.75 mL, 3.2 mmol, 0.5 equiv.) was added and the reaction was stirred another hour. The crude product was purified by column chromatography (0 to 20% EtOAc/heptanes, gradient elution), affording the title compound (3.06 g, 82%) as a yellow foamy solid. m/z (EST, +ve ion)=384.0, 386.0 [M+H-boc]+. Intermediate 5: Tert-butyl 3-amino-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate Step A. Tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl-3-((diphenylmethylene)amino)-1H-indazole-1-carboxylate A microwave vial was charged with tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (1.00 g, 1.71 mmol), cesium carbonate (1.14 g, 3.42 mmol), Pd2(dba)3(157 mg, 171 μmol) and XantPhos (101 mg, 171 μmol). Dry dioxane (17.1 mL) followed by benzophenone imine (310 μL, 1.83 mmol) were added and nitrogen was bubbled through the reaction mixture for 5 min. The vial was sealed, and the reaction mixture was heated to 90° C. for 2 h in an oil bath. A satd. aqueous solution of NaHCO3was added and the reaction mixture was extracted with EtOAc (3×). The combined extracts were then washed with brine, dried with anhyd. Na2SO4, filtered and concentrated under vacuum. The crude product was purified by column chromatography (0 to 30% EtOAc/heptanes, gradient elution), affording the title compound (1.03 g, 88%) as yellow oil. m/z (ESI, +ve ion)=685.4 [M+H]+. Step B. Tert-butyl 3-amino-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate Hydroxylamine hydrochloride (101 mg, 1.46 mmol) and sodium acetate (120 mg, 1.46 mmol) were added to tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-((diphenylmethylene)amino)-1H-indazole-1-carboxylate (1.00 g, 1.46 mmol) in dry MeOH (14.6 mL) at room temperature and the reaction was stirred for 16 h. The solvent was removed under reduced pressure. The crude product was purified by column chromatography (0 to 60% EtOAc/heptanes, gradient elution), affording the title compound (640 mg, 84%) as yellow solid. m/z (EST, +ve ion)=521.0 [M+H]+. 1H NMR (400 MHz, CDCl3) δ8.06 (s, 1H), 7.78 (d, J=8.9 Hz, 1H), 7.41 (d, J=8.2 Hz, 1H), 7.01 (d, J=8.2 Hz, 1H), 6.66 (dd, J=8.9, 2.6 Hz, 1H), 5.55 (d, J=2.3 Hz, 1H), 4.44 (s, 2H), 3.49 (t, J=8.6 Hz, 1H), 3.37 (s, 3H), 2.34 (dd, J=9.2, 4.8 Hz, 1H), 2.14-2.06 (m, 1H), 1.67 (d, J=2.4 Hz, 18H). Example 1. racemic-5′-Methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one In a 4 mL vial was dissolved (1R,2S)-2-(3-amino-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (54.0 mg, 169 μmol) and 4-chloro-5-methoxypyrimidine (29.8 mg, 202 μmol) in acetic acid/water (1:1) (1.00 mL). The reaction was heated to 100° C. for 1 h. The reaction mixture was poured into 5 mL of a solution of aq. NaOH 2 M. The aqueous layer was extracted with EtOAc (3×). The combined organic layers were washed with brine, dried over anhyd. sodium sulfate and concentrated. The product was purified by column chromatography (0 to 10% MeOH/DCM, gradient elution), affording Example 1 (22.9 mg, 32%) as white solid after lyophilization. m/z (EST, +ve ion)=429.2 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.68 (s, 1H), 10.42 (s, 1H), 9.12 (s, 1H), 8.04 (d, J=3.0 Hz, 2H), 7.40 (d, J=7.9 Hz, 2H), 6.89 (d, J=9.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.72 (d, J=2.5 Hz, 1H), 3.94 (s, 3H), 3.33 (s, 3H under water peak), 3.18 (t, J=8.5 Hz, 1H), 2.33 (dd, J=7.8, 4.7 Hz, 1H), 1.98 (dd, J=9.0, 4.7 Hz, 1H). Example 2. racemic-5′-Methoxy-2-{3-[(5-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 2 was prepared using the procedure described in Example 1 from (1R,2S)-2-(3-amino-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (60.0 mg, 187 μmol) and 4-chloro-5-methylpyrimidine (27 mg, 206 μmol). The product was purified by column chromatography (0 to 10/o MeOH/DCM, gradient elution), concentrated, and then lyophilized from MeCN and water to afford Example 2 (7.7 mg, 10%) as white solid. m/z (ESI, +ve ion)=413.2 [M+H]+. 1 H NMR (400 MHz, DMSO) δ12.69 (s, 1H), 10.43 (s, 1H), 9.02 (s, 1H), 8.24 (s, 1H), 8.13 (s, 1H), 7.41 (s, 1H), 7.34 (d, J=8.5 Hz, 1H), 6.89 (dd, J=8.5, 1.0 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.71 (d, J=2.6 Hz, 1H), 3.33 (s, 3H, under water peak), 3.18 (t, J=8.4 Hz, 1H), 2.33 (dd, J=7.9.4.7 Hz, 1H), 2.21 (s, 3H), 1.98 (dd, J=9.0, 4.7 Hz, 1H). Example 3. racemic-2-{3-[(5-Chloropyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 3 was prepared using the procedure described in Example 1 from (1R,2S)-2-(3-Amino-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (60.0 mg, 187 μmol) and 4,5-dichloropyrimidine (31.3 mg, 206 μmol). The product was purified by C18 column chromatography (5 to 40% MeCN/aq. ammonium formate buffer, gradient elution), affording Example 3 (4.8 mg, 6%) as white solid after lyophilization. m/z (EST, +ve ion)=433.1 [M+H]+. 1H NMR (40 MHz, DMSO) δ 12.85 (s, 1H), 10.43 (s, 1H), 9.58 (s, 1H), 8.46 (s, 1H), 8.31 (s, 1H), 7.44 (s, 1H), 7.37 (d, J=8.3 Hz, 1H), 6.95-6.87 (m, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.70 (d, J=2.5 Hz, 1H), 3.33 (s, 3H), 3.19 (t, J=8.4 Hz, 1H), 2.34 (dd, J=8.0, 4.7 Hz, 1H), 1.98 (dd, J=9.0, 4.6 Hz, 1H). Example 4. (1R,2S)-5′-Methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared using the procedure described in Example 1, from tert-butyl 3-amino-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (108 mg, 207 μmol) and 4-chloro-5-methoxypyrimidine (36.7 mg, 249 μmol). The Boc groups are cleaved in situ during the reaction conditions. The product was purified by C18 column chromatography (10% to 30% MeCN/aq. ammonium formate buffer, gradient elution), affording Example 4 (9.6 mg, 11%) as white solid after lyophilization. m/z (ESI, +ve ion)=429.2 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.68 (s, 1H), 10.43 (s, 1H), 9.12 (s, 1H), 8.04 (d, J=3.1 Hz, 2H), 7.44-7.35 (m, 2H), 6.89 (d, J=9.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.72 (d, J=2.5 Hz, 1H), 3.94 (s, 3H), 3.33 (s, 3H under water peak), 3.18 (t, J=8.6 Hz, 1H), 2.33 (dd, J=7.9, 4.7 Hz, 1H), 1.98 (dd, J=9.0, 4.6 Hz, 1H). Example 5. (1R,2S)-5′-Methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. (1R,2S)-2-(3-Iodo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one To an oven-dried flask was added (1R,2S)-2-(1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (4.00 g, 13.1 mmol) followed by DMF (8 mL and Methanol (8 mL. To this suspension was added K2CO3(3.62 g, 26.2 mmol). Finally, molecular iodine (4.32 g, 17.0 mmol) dissolved in DMF (8 mL) and was added dropwise and allowed to stir at rt. After 4 h, the reaction was complete. The mixture was quenched with Na2S2O3in water and stirred for 2 h. Solid was collected by filtration and washed with water. Wet solid was frozen and lyophilized, affording the title compound (4.4 g, 78% yield. Step B. Tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To an oven-dried flask was added 4-dimethylaminopyridine (9.0 mg, 0.07 mmol) followed by (1R,2S)-2-(3-iodo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (637 mg, 1.48 mmol), N-ethyl-N-isopropyl-propan-2-amine (1.0 mL, 5.9 mmol) and MeCN (5.0 mL). The mixture was stirred at room temperature and di-tert-butyl dicarbonate (967 mg, 4.43 mmol) was added, resulted in light yellow homogeneous solution. After 2 h, the reaction mixture was concentrated and the resulting residue was purified by column chromatography (0% to 25%, EtOAc/hexanes, gradient elution) to afford the product as a white foam (51) (822 mg, 88%). Step C. Tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[(5-ethoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a 50 ml round bottom flask were added cesium carbonate (41.3 mg, 0.130 mmol), 5-ethoxypyrimidin-4-amine (9.3 mg, 0.070 mmol), Pd2(dba)3(5.8 mg, 0.010 mmol), tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (40.0 mg, 0.0600 mmol), XantPhos (3.7 mg, 0.010 mmol) and dry toluene (4.2 mL). The reaction mixture was stirred and purged with argon (in balloon) for 10 min to form a green suspension, and then heated to 120° C., resulting in a yellow suspension. The reaction was monitored by LCMS and TLC until the full conversion of the starting materials (approx. 70 min), cooled down to rt, diluted with EtOAc, washed with sat. aq. NaHCO3and dried over Na2SO4. The residue was purified by column chromatography (0% to 90% ethyl acetate/hexane, gradient elution) to provide the title compound (24.0 mg, 59%) as a yellow oil. Step D. (1R,2S)-2-[3-[(5-ethoxypyrimidin-4-yl)amino]-1H-indazol-6-yl]-5′-methoxy-spiro[cyclopropane-1,3′-indoline]-2′-one To a 50 ml round bottom flask containing tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[(5-ethoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (24.0 mg, 0.0400 mmol) in DCM (1.9 mL) was added trifluoroacetic acid (0.15 mL, 1.9 mmol). The reaction mixture was stirred and monitored by LCMS until the full conversion of the starting materials (approx. 3 hrs), diluted with acetonitrile. The resulting brown solution was purified by Prep. HPLC (Gemini C18, 30 to 80% (0.1% TFA in water)/(0.1% TFA in Acetonitrile)) to provide the desired product Example 5 (12.4 mg, 75%) as a yellow film. m/z (ESI, +ve ion) 443.2 (M+H)+.1H NMR (400 MHz, METHANOL-d4) δ ppm 1.58 (t, J=6.94 Hz, 3H) 2.10-2.32 (m, 2H) 3.26-3.30 (m, 3H) 3.32-3.39 (m, 1H) 4.27-4.44 (m, 2H) 5.50-5.61 (m, 1H) 6.55-6.67 (m, 1H) 6.76-6.89 (m, 1H) 6.91-7.04 (m, 1H) 7.44-7.62 (m, 2H) 8.03-8.18 (m, 1H) 8.36-8.49 (m, 1H). Example 6. (1R,2S)-2-{3-[(5-cyclopropylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. Tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[(5-cyclopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (40.0 mg, 0.0600 mmol) and 5-cyclopropylpyrimidin-4-amine (9.0 mg, 0.070 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0˜90%) to provide the title compound (17.0 mg, 42%) as a yellow oil. Step B. (1R,2S)-2-[3-[(5-Cyclopropylpyrimidin-4-yl)amino]-1H-indazol-6-yl]-5′-methoxy-spiro[cyclopropane-1,3′-indoline]-2′-one This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[(5-cyclopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (17.0 mg, 0.0400 mmol) and trifluoroacetic acid (0.10 mL, 1.3 mmol). The resulting brown solution was purified by Prep. HPLC (Gemini C18, 10 to 90% (0.1% TFA in water)/(0.1% TFA in Acetonitrile)) to provide the desired product Example 6 (6.2 mg, 53%) as a colorless film. m/z (ESI, +ve ion) 439.2 (M+H)+.1H NMR (400 MHz, METHANOL-d4) δ ppm 0.85-0.97 (m, 2H) 1.16-1.31 (m, 2H) 1.87-2.04 (m, 1H) 2.16-2.32 (m, 2H) 5.53-5.63 (m, 1H) 6.58-6.69 (m, 1H) 6.79-6.90 (m, 1H) 6.96-7.06 (m, 1H) 7.48-7.63 (m, 2H) 8.15-8.30 (m, 1H) 8.52-8.63 (m, 1H). Example 7. (1R,2S)-2-{3-[(5-Chloropyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. Tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-((5-chloropyrimidin-4-yl)amino)-1H-indazole-1-carboxylate This compound was prepared using the procedure described in Example 5 from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (110 mg, 188 μmol) and 4-amino-5-chloropyrimidine (29.3 mg, 215 μmol). The product was purified by column chromatography (20 to 100/o EtOAc/heptanes, gradient elution), affording the title compound (18.0 mg, 15%). m/z (ESI, +e ion)=633.3 [M+H]+. Step B. (1R,2S)-2-(3-((5-Chloropyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one This compound was prepared using the procedure described in Example 5 from tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-((5-chloropyrimidin-4-yl)amino)-1H-indazole-1-carboxylate (18 mg, 28.4 μmol). The product was purified by C18 column chromatography (10 to 40% MeCN in aq. ammonium formate buffer), affording Example 7 (3.0 mg, 24%) as white solid after lyophilization. m/z (ESI, +ve ion)=433.1 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.85 (s, 1H), 10.43 (s, 1H), 9.58 (s, 1H), 8.46 (s, 1H), 8.31 (s, 1H), 7.44 (s, 1H), 7.37 (d, J=8.4 Hz, 1H), 6.91 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.4, 2.4 Hz, 1H), 5.70 (d, J=2.3 Hz, 1H), 3.33 under water (s, 3H), 3.19 (t, J=8.4 Hz, 1H), 2.34 (dd, J=7.9, 4.6 Hz, 1H), 1.98 (dd, J=9.0, 4.6 Hz, 1H). Example 8. (1S,2R)-5′-Methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. (1S,2R)-5′-methoxy-2-(3-((5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indolin]-2′-one (8A) A vial containing Example 1 (17.0 mg, 39.7 μmol) was submitted to chiral HPLC separation. Separation conditions are: Column: AS-H, 10×250 mm 5 um, Mode: Isocratic, Mobile phase: 60% MeOH-0.1% ammonium hydroxide, 40% supercritical CO2, Flow rate: 10 mL/min, Back pressure: 120 bar, Column Temperature: 40° C., Run time (min): 16). The second peak to elute corresponds to title product while the first peak is the corresponding enantiomer (1R,2S). The solution is concentrated and lyophilized from MeCN and water, affording the title compound (15a) (6.7 mg, 39%) as white solid. m/z (ESI, +ve ion)=429.3 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.67 (s, 1H), 10.41 (s, 1H), 9.10 (s, 1H), 8.04 (d, J=2.1 Hz, 2H), 7.42-7.37 (m, 2H), 6.89 (d, J=9.5 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.72 (d, J=2.5 Hz, 1H), 3.94 (s, 3H), 3.18 (t, J=8.5 Hz, 1H), 2.32 (dd, J=7.9, 4.6 Hz, 1H), 1.98 (dd, J=9.0, 4.7 Hz, 1H). Example 9. (1R,2S)-2-(3-{[5-Chloro-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-Chloro-6-morpholinopyrimidin-4-amine (9A) A reaction vial was charged with 4-amino-5,6-dichloropyrimidine (300 mg, 1.83 mmol) and morpholine (145 μL, 1.65 mmol) in DMSO (3.66 mL). The reaction mixture was heated to 60° C. for 16 h. The reaction mixture was partially concentrated and directly purified by column chromatography (40% to 100% EtOAc/heptanes, gradient elution), affording the title compound (9A) (318 mg, 81%) as white crystals. m/z (ESI, +ve ion)=215.0 [M+H]+. Step B. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-chloro-6-morpholinopyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate A microwave vial was charged with tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (60.0 mg, 103 μmol), 5-chloro-6-morpholinopyrimidin-4-amine (24.2 mg, 113 μmol), cesium carbonate (68.3 mg, 205 μmol), Pd2(dba)3(9.4 mg, 10.3 μmol) and XantPhos (6.0 mg, 10.3 μmol) and was purged with nitrogen. Previously degassed toluene (2.0 mL) was added and nitrogen was bubbled through the reaction mixture for 2 min. The vial was sealed and the reaction mixture was heated to 100° C. for 2 h in an oil bath. The reaction mixture was filtered on a pad of celite using EtOAc and concentrated. The crude product was purified by column chromatography (0% to 10% MeOH/DCM, gradient elution), affording the title compound (49.5 mg, 67%). m/z (ESI, +ve ion)=718.0 [M+H]+. Step C In a flask, tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-chloro-6-morpholinopyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (49.5 mg, 60.7 μmol) was dissolved in DCM (4.40 mL) and trifluoroacetic acid (440 μL, 5.69 mmol) was added. The reaction was stirred at rt for 5 h and then concentrated to dryness. The crude residue was directly purified by C18 column chromatography (10 to 40% MeCN/aq. ammonium formate buffer, gradient elution). The desired fractions were combined and lyophilized, affording Example 9 (12.3 mg, 39%) as white solid. m/z (ESI, +ve ion)=518.2 [M+H]+. 1 H NMR (400 MHz, DMSO) δ 12.72 (s, 1H), 10.43 (s, 1H), 9.16 (s, 1H), 7.98 (s, 1H), 7.41 (s, 1H), 7.35 (d, J=8.3 Hz, 1H), 6.94-6.83 (m, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.5 Hz, 1H), 5.71 (d, J=2.6 Hz, 1H), 3.74-3.67 (m, 4H), 3.51-3.44 (m, 4H), 3.33 (with water peak) (s, 3H), 3.18 (t, J=8.4 Hz, 1H), 2.33 (dd, J=8.0, 4.8 Hz, 1H), 1.98 (dd, J=9.0, 4.7 Hz, 1H). Example 10. (1R,2S)-2-{3-[(2-Chloro-5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one A microwave vial was charged with tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (75.0 mg, 128 μmol), 2-chloro-5-methoxypyrimidin-4-amine (22.6 mg, 137 μmol), cesium carbonate (85.3 mg, 257 μmol). Pd2(dba)3(11.8 mg, 12.8 μmol) and XantPhos (7.6 mg, 12.8 μmol) and was purged with nitrogen. Previously degassed toluene (2.6 mL) was added, and nitrogen was bubbled through the reaction mixture for 2 min. The vial was scaled, and the reaction mixture was heated to 100° C. for 2 h in an oil bath. The reaction mixture was then filtered on a pad of Celite using EtOAc and the crude product was concentrated. The residue was dissolved in DCM (5.00 mL) and trifluoroacetic acid (1.00 mL, 13.0 mmol) was added. The reaction was stirred at rt for 1.5 h. A saturated aqueous solution of sodium bicarbonate was slowly added, and the reaction mixture was transferred to an extraction funnel. The layers were separated, and the aqueous layer was extracted with DCM (3×10 mL). The organic layer was dried with anhyd. Na2SO4, filtered and concentrated under vacuum. The product was purified by C18 column chromatography (10% to 40% MeCN/aq. ammonium formate buffer, gradient elution). The desired fractions were combined and lyophilized, affording Example 10 (4.8 mg, 8.0%) as white solid. m/z (ESI, +ve ion)=463.2 [M+H]+. 1 H NMR (400 MHz, DMSO) δ 12.80 (s, 1H), 10.43 (s, 1H), 9.61 (br s, 1H), 7.94 (s, 1H), 7.46-7.40 (m, 2H), 6.97-6.91 (m, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.57 (dd, J=8.5, 2.6 Hz, 1H), 5.70 (d, J=2.5 Hz, 1H), 3.94 (s, 3H), 3.30 (s, 3H), 3.19 (t, J=8.3 Hz, 1H), 2.33 (dd, J=7.9, 4.6 Hz, 1H), 1.98 (dd, J=9.1, 4.7 Hz, 1H). Example 11. (1R,2S)-5′-Methoxy-2-(3-{[5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one A microwave vial was charged with tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (75.0 mg, 128 μmol), 5-chloro-6-morpholinopyrimidin-4-amine (29.7 mg, 141 μmol), cesium carbonate (85.3 mg, 257 μmol), Pd2(dba)3(11.8 mg, 12.8 μmol) and XantPhos (7.6 mg, 12.8 μmol) and was purged with nitrogen. Previously degassed toluene (2.57 mL) was added and nitrogen was bubbled through the reaction mixture for 2 min. The vial was sealed and the reaction mixture was heated to 100° C. for 2 h in an oil bath. The reaction mixture was filtered on a pad of Celite using EtOAc and the crude product was concentrated. The residue was then dissolved in DCM (5.00 mL) and trifluoroacetic acid (1.00 mL, 13.0 mmol) was added. The reaction was stirred at rt for 1.5 h. A saturated aqueous solution of sodium bicarbonate was slowly added and the reaction mixture was transferred to an extraction funnel. The layers were separated and the aqueous layer was extracted with DCM (3×10 mL). The combined extracts were dried with anhyd. Na2SO4, filtered and concentrated under vacuum. The crude was purified by C18 column chromatography (10% to 40% MeCN/aq. ammonium formate buffer, gradient elution). The desired fractions were combined and lyophilized, affording Example 11 (23.2 mg, 35%) as white solid. m/z (ESI, +ve ion)=514.3 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.58 (s, 1H), 10.42 (s, 1H), 8.88 (s, 1H), 7.80 (s, 1H), 7.47-7.32 (m, 2H), 6.87 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.4, 2.3 Hz, 1H), 5.72 (d, J=2.2 Hz, 1H), 379-3.68 (m, 4H), 3.67 (s, 3H), 3.62-3.53 (m, 4H), 3.33 (s, 3H), 3.18 (t, J=8.4 Hz, 1H), 2.32 (dd, J=7.7, 4.7 Hz, 1H), 1.98 (dd, J=8.9, 4.6 Hz, 1H). Example 12. (1R,2S)-5′-Methoxy-2-(3-{[5-methoxy-6-(piperidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-Methoxy-6-(piperidin-1-yl)pyrimidin-4-amine (12A) A reaction vial was charged with 4-amino-6-chloro-5-methoxypyrimidine (100 mg, 595 μmol) and piperidine (120 μL, 1.19 mmol) in toluene (1.2 mL). The reaction mixture was heated to 105° C. for 16 h. The solution was then concentrated to dryness. The product was purified by column chromatography (50% to 100% EtOAc/heptanes, gradient elution), affording the title compound (87.8 mg, 71%) as white crystals. m/z (ESI, +ve ion)=209.0 [M+H]+. Step B. (1R,2S)-5′-methoxy-2-(3-((5-methoxy-6-(piperidin-1-yl)pyrimidin-4-yl)amino)-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indolin]-2′-one A microwave vial was charged with tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (75.0 mg, 128 μmol), 5-methoxy-6-(piperidin-1-yl)pyrimidin-4-amine (29.4 mg, 141 μmol), cesium carbonate (85.3 mg, 257 μmol), Pd2(dba)3(11.8 mg, 12.8 μmol) and XantPhos (7.6 mg, 12.8 μmol) and was purged with nitrogen. Previously degassed toluene (2.57 mL) was added and nitrogen was bubbled through the reaction mixture for 2 min. The vial was sealed and the reaction mixture was heated to 100° C. for 2 h in an oil bath. The reaction mixture was filtered on a pad of Celite using EtOAc and the crude product was concentrated. The residue was dissolved in DCM (5.00 mL) and trifluoroacetic acid (1.00 mL, 13.0 mmol) was added. The reaction was stirred at rt for 1.5 h. A satd. aq. solution of bicarbonate was added and the reaction mixture was transferred to an extraction funnel. The layers were separated, and the aqueous layer was extracted with DCM (3×10 mL). The combined organic extracts were dried with anhyd. Na2SO4, filtered and concentrated under vacuum. The product was purified by C18 column chromatography (20 to 40% MeCN in aq. ammonium formate buffer). The desired fractions were combined and lyophilized, affording the title compound (23.2 mg, 35%) as white solid. m/z (ESI, +ve ion)=512.3 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.53 (s, 1H), 10.41 (s, 1H), 8.76 (1Hs), 7.76 (s, 1H), 7.42 (d, J=8.4 Hz, 1H), 7.37 (s, 1H), 6.87 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.4, 2.4 Hz, 11H), 5.72 (d, J=2.3 Hz, 1H), 3.65 (s, 3H), 3.62-3.53 (m, 4H), 3.33 (s, 3H under water peak), 3.17 (t, J=8.5 Hz, 1H), 2.31 (dd, J=7.8, 4.7 Hz, 1H), 1.97 (dd, J=8.9, 4.6 Hz, 1H), 1.67-1.54 (m, 6H). Example 13. (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyrazin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared using the procedure described in Example 5, from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (50.0 mg, 85.5 μmol) and 3-methoxypyrazine-2-amine (18.6 mg, 145 μmol). The product was purified by C18 column chromatography (0 to 50% MeCN in water), affording the title compound (13) (9.3 mg, 20%) as yellow solid after lyophilization. m/z (ESI, +ve ion)=429.4 [M+H]+. 1H NMR (500 MHz, DMSO) δ 12.56 (s, 1H), 10.40 (s, 1H), 8.80 (s, 1H), 7.48 (d, J=3.0 Hz, 1H), 7.46 (d, J=3.0 Hz, 1H), 7.42-7.37 (m, 2H), 6.87 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.4, 2.6 Hz, 1H), 5.72 (d, J=2.5 Hz, 1H), 3.98 (s, 3H), 3.33 (s, J=0.9 Hz, 3H), 3.17 (t, J=8.5 Hz, 1H), 2.31 (dd, J=7.9, 4.7 Hz, 1H), 1.98 (dd, J=9.0, 4.7 Hz, 1H). Example 14. (1R,2S)-5′-methoxy-2-{3-[(6-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared according to the procedure described in Example 5, from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (50.0 mg, 85.5 μmol) and 6-methoxypyrimidine-4-amine (16.4 mg, 128 μmol). The product was purified by C18 column chromatography (0 to 50% MeCN in aq. ammonium formate buffer), affording the title compound (19.6 mg, 43%) as white solid after lyophilization. m/z (EST, +ve ion)=429.3 [M+H]+. 1H NMR (500 MHz, DMSO) δ 12.43 (s, 1H), 10.41 (s, 1H), 10.17 (s, 1H), 8.38 (d, J=0.9 Hz, 1H), 7.93 (d, J=8.4 Hz, 1H), 7.35 (s, 1H), 7.20 (s, 1H), 6.91 (dd, J=8.5, 1.0 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.68 (d, J=2.6 Hz, 1H), 3.87 (s, 3H), 3.31 (s, 3H), 3.17 (t, J=8.4 Hz, 1H), 2.32 (dd, J=7.9, 4.7 Hz, 1H), 1.97 (dd, J=9.0, 4.7 Hz, 1H). Example 15. (1R,2S)-2-{3-[(6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[5H,6H,7H-cyclopenta[d]pyrimidin-4-ylamino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a mixture of tert-butyl (R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (240.00 mg, 0.380 mmol, 1.00 equiv) and 5H,6H,7H-cyclopenta[d]pyrimidin-4-amine (61.65 mg, 0.456 mmol, 1.2 equiv) in dry toluene (20.00 mL) were added Cs2CO3(247.67 mg, 0.760 mmol, 2.00 equiv). Pd2(dba)3(34.80 mg, 0.038 mmol, 0.10 equiv) and XantPhos (21.99 mg, 0.038 mmol, 0.10 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The solvent was filtered and washed with EtOAc (10 mL). The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography, eluted with 0-50% EtOAc in PE to give the title compound (80 mg, 31.31%) as light yellow oil. m/z (ESI, +ve ion)=639.35 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.57 (s, 1H), 8.12 (s, 1H), 7.89 (d, J=8.4 Hz, 1H), 7.82 (d, J=8.8 Hz, 1H), 7.12 (d, J=8.4 Hz, 1H), 6.72-6.69 (m, 1H), 5.62 (d, J=2.8 Hz, 1H), 4.17-4.12 (m, 1H), 3.53 (t, J=8.4 Hz, 1H), 3.42 (s, 3H), 3.12 (t, J=8.0 Hz, 2H), 2.94 (t, =7.6 Hz, 2H), 2.41-2.37 (m, 1H), 2.28-2.21 (m, 2H), 2.13-2.10 (m, 1H), 1.70 (s, 18H). Step B. (1R,2S)-2-(3-[5H,6H,7H-cyclopenta[d]pyrimidin-4-ylamino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a mixture of the compound from Step A (60.00 mg, 0.094 mmol, 1.00 equiv) in DCM (1.00 mL) was added TFA (0.20 mL). The mixture was stirred at 25° C. for 12 h, then diluted with DCM (20 mL) and washed with saturated NaHCO3(20 mL). The aqueous layer was extracted with DCM (2×10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue (two batches combined, total 0.125 mmol) was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 13% B to 40% B in 8 min; 254/220 nm; RT1: 7.35 min to give Example 15 (30 mg, 72.10%) as a white solid. m/z (ESI+ve ion)=439.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.65 (s, 1H), 10.42 (s, 1H), 9.22 (s, 1H), 8.24 (s, 1H), 7.43-7.41 (m, 2H), 6.92-6.89 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.58 (m, 1H), 5.72 (d, J=2.4 Hz, 1H), 3.35 (s, 3H), 3.19 (t, J=8.4 Hz, 1H), 2.80 (t, J=7.6 Hz, 2H), 2.73-2.68 (m, 2H), 2.35-2.32 (m, 1H), 2.04-1.96 (m, 3H). Example 16. (1R,2S)-2-{3-[(2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′ 1′H one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-(2,3-dihydro-1-benzofuran-7-ylamino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (200.00 mg, 0.317 mmol, 1.00 equiv) and 2,3-dihydro-1-benzofuran-7-amine (51.37 mg, 0.380 mmol, 1.20 equiv) in dry toluene (5.0 mL) were added Cs2CO3(206.39 mg, 0.634 mmol, 2.00 equiv), Pd2(dba)3(29.00 mg, 0.032 mmol, 0.10 equiv) and XantPhos (18.33 mg, 0.032 mmol, 0.10 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The solvent was removed under reduced pressure. The residue was purified by prep-TLC (rinsed with PE/EA=2/1) to give the title compound (200 mg, 97.88%) as yellow oil. m/z (ESI+ve ion)=639.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.16 (d, J=8.0 Hz, 1H), 7.82-7.80 (m, 1H), 7.56-7.52 (m, 1H), 7.06 (d, J=8.4 Hz, 1H), 6.96-6.88 (m, 2H), 6.70-6.66 (m, 2H), 5.57 (d, J=2.8 Hz, 1H), 4.69-4.65 (m, 2H), 3.56-3.50 (m, 1H), 3.38 (s, 3H), 3.31 (t, J=8.4 Hz, 2H), 2.39-2.36 (m, 1H), 2.14-2.10 (m, 1H), 1.71 (d, J=5.6 Hz, 18H). Step B. (1R,2S)-2-[3-(2,3-dihydro-1-benzofuran-7-ylamino)-1H-indazol-6-yl]-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To the mixture of the compound from Step A (215 mg, 0.336 mmol) in DCM (5 mL) was added TFA (0.5 mL). The mixture was stirred at 25° C. for 12 h, then diluted with DCM (20 mL) and washed with saturated NaHCO3(20 mL) and brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 19×250 mm, 10 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1% NH3·H2O). Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 42% B to 52% B in 8 min; 254 nm; RT1: 7.15 min to give Example 2 (80 mg, 54%) as a white solid. m/z (ESI+ve ion)=439.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 10.41 (s, 1H), 7.89 (d, J=8.8 Hz, 2H), 7.82 (t, J=4.4 Hz, 1H), 7.28 (s, 1H), 6.86-6.83 (m, 1H), 6.76-6.74 (m, 3H), 6.60-6.57 (m, 1H), 5.71 (d, J=2.4 Hz, 1H), 4.60 (t, J=8.8 Hz, 2H), 3.33 (s, 3H), 3.33-3.15 (m, 3H), 2.33-2.30 (m, 1H), 1.99-1.97 (m, 1H). Example 17. (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyridin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared using the procedure described in Example 5, from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (75.0 mg, 128 μmol) and 3-methoxypyridin-2-amine (17.9 mg, 141 μmol). The product was purified by C18 column chromatography (10 to 30% MeCN in aq. ammonium formate buffer), affording the title compound (16.1 mg, 29%) as white solid after lyophilization. m/z (ESI, +ve ion)=428.3 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.42 (s, 1H), 10.42 (s, 1H), 8.16 (s, 1H), 7.51 (dd, J=5.0, 1.3 Hz, 1H), 7.44 (d, J=8.4 Hz, 1H), 7.35 (s, 1H), 7.19 (dd, J=7.9, 1.3 Hz, 1H), 6.85 (d, J=8.7 Hz, 1H), 6.76-6.66 (m, 2H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.74 (d, J=2.6 Hz, 1H), 3.87 (s, 3H), 3.30 (s, 3H, peak under water), 3.17 (t, J=8.4 Hz, 1H), 2.35-2.29 (m, 1H), 1.97 (dd, J=9.0, 4.6 Hz, 1H). Example 18. (1R,2S)-5′-methoxy-2-{3-[(4-methoxypyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared using the procedure described in Example 5, from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (75.0 mg, 128 μmol) and 4-methoxypyridin-3-amine (18.4 mg, 141 μmol). The product was purified by C18 column chromatography (10 to 40% MeCN in aq. ammonium formate buffer), affording the title compound (9.5 mg, 17%) as white solid after lyophilization. m/z (ESI, +ve ion)=428.2 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.13 (s, 1H), 10.43 (s, 1H), 9.18 (s, 1H), 8.02 (d, J=5.3 Hz, 1H), 7.90-7.81 (m, 2H), 7.31 (s, 1H), 7.03 (d, J=5.4 Hz, 1H), 6.87 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.4, 2.5 Hz, 1H), 5.70 (d, J=2.4 Hz, 1H), 3.94 (s, 3H), 3.32 (s, 3H), 3.17 (t, J=8.5 Hz, 1H), 2.32 (dd, J=7.8, 4.8 Hz, 1H), 1.97 (dd, J=9.0, 4.7 Hz, 1H). Example 19. (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyridin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared according to the procedure described in Example 5, from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (50.0 mg, 85.5 μmol) and 3-methoxypyridin-4-amine (16.1 mg, 126 μmol). The product was purified by C18 column chromatography (0 to 30/o MeCN in water), affording the title compound (6.5 mg, 14%) as white solid after lyophilization. m/z (ESI, +ve ion)=428.3 [M+H]+. 1H NMR (500 MHz, DMSO) δ 12.95 (s, 1H), 10.43 (s, 1H), 9.97 (s, 1H), 8.28 (s, 1H), 8.20 (d, J=6.6 Hz, 1H), 7.82 (d, J=6.6 Hz, 1H), 7.77 (d, J=8.4 Hz, 1H), 7.48 (s, 1H), 7.00 (d, J=8.5 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (dd, J=8.5, 2.6 Hz, 1H), 5.70 (d, J=2.6 Hz, 1H), 4.07 (s, 3H), 3.20 (t, J=8.4 Hz, 1H), 2.35 (dd, J=8.0, 4.9 Hz, 1H), 2.00 (dd, J=9.1, 4.7 Hz, 1H). Example 20. (1R,2S)-2-(3-{[5-chloro-6-(4-methylpiperazin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4,5-dichloro-6-(4-methylpiperazin-1-yl)pyrimidine 1-Methylpiperazine (200 μL, 1.77 mmol) was added to a mixture of 4,5,6-trichloropyrimidine (336 mg, 1.77 mmol) and N,N-diisopropylethylamine (930 μL, 5.32 mmol) in NMP (7.0 mL). The reaction was stirred at 80° C. for 15 h. EtOAc and water were added and the reaction mixture was transferred to an extraction funnel. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layers were then washed with brine, dried with anh. Na2SO4, filtered and concentrated under vacuum. The product was purified by column chromatography (0 to 5% MeOH in DCM), affording the title compound (312 mg, 71%) as red oil. m/z (ESI, +ve ion)=247.0 [M+H]+. Step B. (1R,2S)-2-(3-((5-chloro-6-(4-methylpiperazin-1-yl)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one A microwave vial was charged with tert-butyl (1R,2S)-2-(3-amino-1-4tert-butoxycarbonyl)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (75.0 mg, 144 μmol), 4,5-dichloro-6-(4-methylpiperazin-1-yl)pyrimidine (39.2 mg, 158 μmol), cesium carbonate (95.8 mg, 288 μmol), Pd2(dba)3(13.2 mg, 14.4 μmol) and XantPhos (8.5 mg, 14.4 μmol) and was purged with nitrogen. Previously degassed toluene (3.0 mL) was added and nitrogen was bubbled through the reaction mixture for 2 min. The vial was sealed and the reaction mixture was heated to 100° C. for 1.5 h in an oil bath. The reaction mixture was filtered on a pad of Celite using EtOAc and the crude product was concentrated. The residue was dissolved in DCM (5.00 mL) and trifluoroacetic acid (1.00 mL, 13.0 mmol) was added. The reaction was stirred at rt for 1.5 h. A satd. aqueous solution of sodium bicarbonate was added and the reaction mixture was transferred to an extraction funnel. The layers were separated and the aqueous layer was extracted with DCM (3×10 mL). The combined extracts were dried with anhyd. Na2SO4, filtered and concentrated under vacuum. The product was purified by C18 column chromatography (5 to 30% MeCN in aq. ammonium formate buffer). The desired fraction were combined and lyophilized, affording the title compound (37.1 mg, 55%) as yellow solid identified as a formate salt. m/z (ESI, +ve ion)=531.3 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.71 (s, 1H), 10.43 (s, 1H), 9.11 (s, 1H), 7.95 (s, 1H), 7.41 (s, 1H), 7.35 (d, J=8.4 Hz, 1H), 6.89 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.4, 2.2 Hz, 1H), 5.71 (d, J=2.0 Hz, 1H), 3.54-3.45 (m, 4H), 3.33 (s, 3H under water), 3.18 (t, J=8.4 Hz, 1H), 2.47-2.39 (m, 4H), 2.33 (dd, J=7.5, 4.9 Hz, 1H), 2.22 (s, 3H), 1.98 (dd, J=8.9, 4.6 Hz, 1H). Example 21. (1R,2S)-5′-methoxy-2-{3-[(1,3,5-trimethyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared according to the procedure described in Example 5, from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (50.0 mg, 85.5 μmol) and 1,3,5-trimethyl-1H-pyrazol-4-amine (7.3 mg, 58.2 μmol). The product was purified by C18 column chromatography (0 to 30% MeCN in aq. ammonium formate buffer), affording the title compound (8.2 mg, 33%) as white solid after lyophilization. m/z (ESI, +ve ion)=429.1 [M+H]+. 1H NMR (400 MHz, DMSO) δ 11.35 (s, 1H), 10.39 (s, 1H), 7.41 (d, J=8.1 Hz, 1H), 7.22 (s, 1H), 7.14 (s, 1H), 6.75 (s, 1H), 6.73 (s, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.67 (d, J=2.6 Hz, 1H), 3.64 (s, 3H), 3.12 (t, J=8.5 Hz, 1H), 2.25 (dd, J=7.9.4.6 Hz, 1H), 2.06 (d, J=5.9 Hz, 3H), 1.98-1.89 (m, 4H). Example 22. (1R,2S)-5′-methoxy-2-(3-{[5-(trifluoromethyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared according to the procedure described in Example 1, from tert-butyl 3-amino-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (30.9 mg, 59.4 μmol) and 4-chloro-5-(trifluoromethyl)pyrimidine (11.9 mg, 65.3 μmol). The product was purified by C18 column chromatography (15 to 40% MeCN in aq. ammonium formate buffer), affording the title compound (6.9 mg, 25%) as yellow solid after lyophilization. m/z (ESI, +ve ion)=467.1 [M+H]+. 1H NMR (500 MHz, DMSO) δ 12.89 (s, 1H), 10.42 (s, 1H), 8.68 (s, 1H), 8.54 (s, 1H), 7.46 (s, 1H), 7.32 (d, J=8.4 Hz, 1H), 6.92 (dd, J=8.4, 1.0 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.69 (d, J=2.6 Hz, 1H), 3.19 (t, J=8.4 Hz, 1H), 2.33 (dd, J=8.0, 4.7 Hz, 1H), 1.99 (dd, J=9.0, 4.7 Hz, 1H). CH3O signal is in water peak. Example 23. (1R,2S)-2-{3-[(5-chloro-2-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-chloro-2-methoxypyrimidin-4-amine In a flask was dissolved 2,5-dichloropyrimidin-4-amine (300 mg, 1.74 mmol) in MeOH (8.7 mL) to which was added a sodium methoxide solution (30 wt. % in MeOH, 150 μL, 2.09 mmol) and the solution was heated at 70° C. for 2 h (reflux) and cooled back to rt. Some conversion was observed by LCMS but reaction was not complete. Another 0.5 equiv of sodium methoxide solution (60 μL, 869 μmol) was added and the reaction was stirred further at reflux for another 2 h, when it was then quenched with water and extracted with EtOAc. The organic layer was washed with water, dried (Na2SO4) and concentrated. Crude white solid material (6a) was used such as in next step. m/z (ESI, +ve ion)=159.7 [M+H]+. 1H NMR (500 MHz, DMSO) δ 8.19 (s, 1H), 8.13-7.38 (m, 2H), 3.86 (s, 3H). Step B. (1R,2S)-2-(3-((5-chloro-2-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one In a vial were added tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (50.0 mg, 85.5 μmol), cesium carbonate (70.4 mg, 216 μmol), Pd2(dba); (9.8 mg, 10.7 μmol), XantPhos (6.31 mg, 10.9 μmol), 5-chloro-2-methoxypyrimidin-4-amine (41.0 mg, 128 μmol) and toluene (2.50 mL) and the vial degassed (nitrogen bubbled through solvent for 5 min), and scaled and stirred at 100° C. for 2 h in an oil bath. The reaction was then transferred to a flask and concentrated to dryness. The residue was dissolved in DCM (2.50 mL) and trifluoroacetic acid (1.7 mL) and stirred at rt for 1 h when it was subsequently concentrated to dryness. The product was purified by C18 column chromatography (15 to 40% MeCN in aq. ammonium formate buffer). The desired fraction were combined and lyophilized, affording the title compound (9.0 mg, 18%) as white solid. m/z (ESI, +ve ion)=463.1 [M+H]+. 1H NMR (500 MHz, DMSO) δ 12.81 (s, 1H), 10.41 (s, 1H), 9.57 (s, 1H), 8.23 (s, 1H), 7.44-7.40 (m, 2H), 6.91 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.65 (d, J=2.6 Hz, 1H), 3.48 (s, 3H), 3.19 (t, J=8.5 Hz, 1H), 2.32 (dd, J=8.0, 4.7 Hz, 1H), 1.98 (dd, J=9.0, 4.7 Hz, 1H); (Extra MeO signal unseparated from water peak). Example 24. (1R,2S)-5′-methoxy-2-{3-[(2-methoxypyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared using the procedure described in Example 5, from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (75.0 mg, 128 μmol) and 2-methoxypyridin-3-amine (18.6 mg, 145 μmol). The product was purified by C18 column chromatography (10 to 50% MeCN in aq. ammonium formate buffer), affording the title compound (28.0 mg, 51%) as white solid after lyophilization. m/z (ESI, +ve ion)=428.2 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.16 (s, 1H), 10.43 (s, 1H), 8.37 (dd, J=7.8, 1.4 Hz, 1H), 8.02 (s, 1H), 7.91 (d, J=8.4 Hz, 1H), 7.61 (dd, J=4.9, 1.4 Hz, 1H), 7.31 (s, 1H), 6.97-6.85 (m, 2H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.4, 2.5 Hz, 1H), 5.70 (d, J=2.3 Hz, 1H), 3.98 (s, 3H), 3.32 (s, 3H), 3.17 (t, J=8.4 Hz, 1H), 2.32 (dd, J=7.7, 4.8 Hz, 1H), 1.97 (dd, J=8.9, 4.6 Hz, 1H). Example 25. (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-1-methyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one This compound was prepared according to the procedure described in Example 5, from tert-butyl 3-bromo-6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate (50.0 mg, 85.5 μmol) and 3-methoxy-1-methyl-1H-pyrazol-4-amine hydrochloride (15.4 mg, 94.1 μmol). The product was purified by C18 column chromatography (20 to 40% MeCN in aq. ammonium formate buffer), affording the title compound (7.4 mg, 20%) as white solid after lyophilization. m/z (ESI, +ve ion)=431.4 [M+H]+. 1H NMR (500 MHz, DMSO) δ 11.54 (s, 1H), 10.39 (s, 1H), 7.88 (s, 1H), 7.85 (d, J=8.4 Hz, 1H), 7.82 (s, 1H), 7.17 (s, 1H), 6.77 (d, J=8.5 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.57 (dd, J=8.5, 2.6 Hz, 1H), 5.68 (d, J=2.5 Hz, 1H), 3.84 (s, 3H), 3.66 (s, 3H), 3.33 (s, 3H), 3.14 (t, J=8.5 Hz, 1H), 2.27 (dd, J=8.0, 4.6 Hz, 1H), 1.95 (dd, J=9.0.4.7 Hz, 1H). Example 26. (1R,2S)-2-{3-[(1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 7-bromo-2,3-dihydro-1-benzofuran-3-ol NaBH4(23.09 mg, 0.610 mmol, 1.3 equiv) was added to a mixture of 7-bromo-2H-1-benzofuran-3-one (100.00 mg, 0.469 mmol, 1.00 equiv) in MeOH (1.00 mL) at 0° C. The mixture was stirred at 0° C. for 1 h. The solvent was concentrated in vacuo. The residue was diluted with water (20 mL), extracted with EA (3×10 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and the filtrate was concentrated in vacuo to give the title compound (100 mg, 97%) as a light yellow solid. The product showed no signal in LCMS.1H NMR (400 MHz, Chloroform-d) δ 7.48-7.38 (m, 2H), 6.90-6.86 (m, 1H), 5.50-5.48 (m, 1H), 4.70-4.67 (m, 1H), 4.59-4.56 (m, 1H). Step B: [(7-bromo-2,3-dihydro-1-benzofuran-3-yl)oxy](tert-butyl)diphenylsilane To the mixture of 7-bromo-2,3-dihydro-1-benzofuran-3-ol (100.00 mg, 0.465 mmol, 1.00 equiv) and imidazole (63.31 mg, 0.930 mmol, 2 equiv) in DMF (1.00 mL) was added TBDPS-Cl (153.38 mg, 0.558 mmol, 1.2 equiv) at 0° C. under nitrogen atmosphere. The resulting mixture was stirred at 25° C. for 3 h. The reaction was diluted with water (10 mL) and extracted with EA (3×10 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and the filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-10% EA in PE to give the title compound (150 mg, 70.43%) as a colorless oil. The product showed no signal in LCMS.1H NMR (400 MHz, Chloroform-d) δ 7.72-7.66 (m, 4H), 7.52-7.35 (m, 7H), 6.88 (d, J=7.6 Hz, 1H), 6.70 (t, J=8.0 Hz, 1H), 5.54-5.52 (m, 1H), 4.52-4.48 (m, 1H), 4.40-4.36 (m, 1H), 1.08 (s, 9H). Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-([3-[(tert-butyldiphenylsilyl)oxy]-2,3-dihydro-1-benzofuran-7-yl]amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (130.00 mg, 0.250 mmol, 1.00 equiv) and [(7-bromo-2,3-dihydro-1-benzofuran-3-yl)oxy](tert-butyl)diphenylsilane (147.21 mg, 0.325 mmol, 1.30 equiv) in dry Dioxane (4 mL) were added K3PO4(106.01 mg, 0.499 mmol, 2 equiv), Pd2(dba)3(22.87 mg, 0.025 mmol, 0.1 equiv) and XantPhos (14.45 mg, 0.025 mmol, 0.1 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The solvent was removed under reduced pressure. The residue was purified by prep-TLC (rinsed with PE/EA=3/1) to give the title compound (120 mg, 51.11%) as yellow solid. m/z (ESI, +ve ion)=893.25 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.24 (d, J=8.0 Hz, 1H), 8.13-8.10 (m, 1H), 7.83-7.80 (m, 1H), 7.73-7.68 (m, 4H), 7.53-7.40 (m, 6H), 7.06 (d, J=8.0 Hz, 1H), 6.92-6.88 (m, 1H), 6.71-6.66 (m, 3H), 5.57-5.52 (m, 2H), 4.51-4.47 (m, 1H), 4.38-4.34 (m, 1H), 3.53 (t, J=8.4 Hz, 1H), 3.38 (d, J=6.8 Hz, 3H), 2.40-2.36 (m, 1H), 2.12 (s, 1H), 1.73-1.70 (m, 18H), 1.09 (s, 9H) Step D. (1R,2S)-2-[3-([3-[(tert-butyldiphenylsilyl)oxy]-2,3-dihydro-1-benzofuran-7-yl]amino)-1H-indazol-6-yl]-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-([3-[(tert-butyldiphenylsilyl)oxy]-2,3-dihydro-1-benzofuran-7-yl]amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (68.00 mg, 0.076 mmol, 1.00 equiv) in bis(2-aminoethyl)amine (0.50 mL) was stirred at 25° C. for 16 h. The resulting mixture was diluted with EA (20 mL) and washed with brine (20 mL). The organic layer was dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by prep-TLC (rinsed with EA/PE=2/1) to give the title compound (50 mg, 90.04%) as a yellow solid. m/z (ESI, +ve ion)=693.15 [M+H].1H NMR (400 MHz, Chloroform-d) δ 7.81-7.79 (m, 1H), 7.75-7.66 (m, 5H), 7.56-7.54 (m, 1H), 7.49-7.40 (m, 8H), 6.95-6.80 (m, 2H), 6.70-6.61 (m, 1H), 5.58-5.52 (m, 2H), 4.49-4.45 (m, 1H), 4.38-4.32 (m, 1H), 3.47-3.42 (m, 1H), 3.37 (d, J=1.6 Hz 3H), 2.30-2.26 (m, 1H), 2.06-2.04 (m, 1H), 1.10-1.09 (m, 9H). Step E. (1R,2S)-2-[3-(1-benzofuran-7-ylamino)-1H-indazol-6-yl]-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To the mixture of (1R,2S)-2-[3-([3-[(tert-butyldiphenylsilyl)oxy]-2,3-dihydro-1-benzofuran-7-yl]amino)-1H-indazol-6-yl]-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one (9.00 mg, 0.010 mmol, 1.00 equiv) in DCM (0.50 mL, 0.006 mmol, 0.58 equiv) was added TFA (0.05 mL). The mixture was stirred at 25° C. for 12 h. The mixture was diluted with DCM (20 ml) and washed with sat. aq. NaHCO3(20 mL). The organic layer was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1% NH3·H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 35% B to 65% B in 8 min; 254 nm; RT1: 6.73 min. The product-containing fractions were concentrated to give the title compound as a white solid (2.2 mg, 49.52%). m/z (ESI, +ve ion)=437.15 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.76 (d, J=2.0 Hz, 1H), 7.71 (d, J=8.4 Hz, 1H), 7.57-7.55 (m, 1H), 7.37 (s, 1H), 7.16-7.10 (m, 2H), 6.91-6.87 (m, 1H), 6.85-6.83 (m, 2H), 6.65-6.62 (m, 1H), 5.63 (d, J=2.4 Hz, 1H), 3.39-3.37 (m, 1H), 3.32 (s, 3H), 2.26-2.23 (m, 1H), 2.20-2.17 (m, 1H). Example 27. 2-{3-[(3-hydroxy-2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one (Mixture of Diastereomers) To the mixture of (1R,2S)-2-[3-([3-[(tert-butyldiphenylsilyl)oxy]-2,3-dihydro-1-benzofuran-7-yl]amino)-1H-indazol-6-yl]-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one (50.00 mg, 0.069 mmol, 1.00 equiv) in tetraethylene glycol (1.00 mL) was added KF (5.97 mg, 0.104 mmol, 1.50 equiv). The resulting mixture was stirred at 80° C. for 12 h. The mixture was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 38% B in 8 min; 254/220 nm; RT1: 7.63 min. The product-containing fractions were concentrated to give the title compound (15 mg, 45.7%) as a white solid. m/z (ESI, +ve ion)=455.10 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.66-7.64 (m, 2H), 7.34 (s, 1H), 6.99 (d, J=7.2 Hz, 1H), 6.89-6.83 (m, 3H), 6.64-6.61 (m, 1H), 5.61 (s, 1H), 5.39-5.38 (m, 1H), 4.65-4.60 (m, 1H), 4.47-4.44 (m, 1H), 3.38-3.35 (m, 1H), 3.30 (s, 3H), 2.25-2.21 (m, 1H), 2.20-2.16 (m, 1H). Example 28. (1R,2S)-2-(3-{[(3S)-3-hydroxy-2,3-dihydro-1-benzofuran-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 29. (1R,2S)-2-(3-{[(3R)-3-hydroxy-2,3-dihydro-1-benzofuran-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one The racemic mixture was separated by PREP-Chiral-HPLC with the following conditions: CHIRALPAK IF, 2×25 cm, 5 μm; Mobile Phase A: HEX:DCM=3:1 (0.1% DEA)-HPLC, Mobile Phase B: IPA-HPLC; Flow rate: 20 mL/min; Gradient: 20% B to 20% B in 15.5 min; 220/254 nm; RT1: 9.797 min; RT2: 13.157 min. The first product-containing fractions were collected and roto-evaporated in vacuo and lyophilized overnight to give Example 4 (5 mg) as a white solid. m/z (EST, +ve ion)=455.35 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.66-7.64 (m, 2H), 7.34 (s, 1H), 6.99 (d, J=6.8 Hz, 1H), 6.89-6.83 (m, 3H), 6.64-6.61 (m, 1H), 5.61 (d, J=2.4 Hz, 1H), 5.40-5.38 (m, 1H), 4.64-4.60 (m, 1H), 4.48-4.44 (m, 1H), 3.35-3.33 (m, 1H), 3.30 (s, 3H), 2.25-2.22 (m, 1H), 2.20-2.16 (m, 1H). The second product-containing fractions were collected and roto-evaporated in vacuo and lyophilized overnight to give Example 5 (3.9 mg) as a white solid. m/z (ESI, +ve ion)=455.30 [M+H]+. 1H NMR (400 MHz, Methanol-d4) δ 7.67-7.65 (m, 2H), 7.34 (s, 1H), 6.99 (d, J=7.2 Hz, 1H), 6.89-6.83 (m, 3H), 6.64-6.61 (m, 1H), 5.61 (d, J=2.4 Hz, 1H), 5.40-5.38 (m, 1H), 4.65-4.61 (m, 1H), 4.48-4.44 (m, 1H), 3.35-3.33 (m, 1H), 3.30 (s, 3H), 2.24-2.22 (m, 1H), 2.20-2.17 (in, 1H). Example 30. (1R,2S)-2-{3-[(2,3-dihydropyrazolo[5,1-b][1,3]oxazol-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. benzyl (2,3-dihydropyrazolo[5,1-b]oxazol-7-yl)carbamate To an oven-dried flask was added 2,3-dihydropyrazolo[5,1-b]oxazole-7-carboxylic acid (250 mg, 1.62 mmol) followed by 1,4-dioxane (7.0 mL). diisopropylethylamine (0.57 mL, 3.2 mmol), benzyl alcohol (0.84 mL, 8.1 mmol) and diphenyl phosphorazidate (0.42 mL, 1.9 mmol). The mixture was heated at 90° C. overnight during which time the solution turned dark brown. The mixture was concentrated purified by column chromatography (20% to 30% acetone/hexanes, gradient elution) to afford the title compound (220 mg, 52%) as an off-white solid. m/z (ESI, +ve ion)=260.2 [M+H]+. Step B. 2,3-dihydropyrazolo[5,1-b]oxazol-7-amine To a suspension of benzyl (2,3-dihydropyrazolo[5,1-b]oxazol-7-yl)carbamate (220 mg, 0.849 mmol) in ethanol (9.0 mL) was added 10% Pd/C (44 mg). The atmosphere was replaced with argon and then replaced with hydrogen. The reaction mixture was stirred at under a hydrogen atmosphere for 2.5 h. The catalyst was removed by filtering over a pad of celite and rinsed with ethanol. The filtrate was concentrated and purified by column chromatography (0% to 10% methanol/DCM, gradient elution) to afford the title compound (90 mg, 85%) as a red purple solid. m/z (ESI, +ve ion)=126.3 [M+H]+. Step C. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((2,3-dihydropyrazolo[5,1-b]oxazol-7-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To an oven-dried flask was added tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (63.1 mg, 0.100 mmol), 2,3-dihydropyrazolo[5,1-b]oxazol-7-amine (13.8 mg, 0.110 mmol), xantphos (5.8 mg, 0.010 mmol). Pd2dba3(9.2 mg, 0.010 mmol), and toluene (2.0 mL). The mixture was degassed with bubbling argon for 10 min. At this time, Cs2CO3(65.2 mg, 0.200 mmol) was added and the reaction mixture was heated to 100° C. for 2 h. The reaction mixture was cooled was cooled to room temperature and diluted with EtOAc and washed with sat. aqueous NaHCO3. The aqueous layer was extracted an additional three times with EtOAc. Combined organic layers were washed with brine, dried with MgSO4, filtered, and concentrated. The residue was purified by column chromatography (0.5% to 7% methanol/DCM, a gradient elution) to provide the title compound (10 mg, 16%) as a yellow foam. m/z (ESI, +ve ion)=629.2 [M+H]+. Step D To an oven-dried flask was added tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((2,3-dihydropyrazolo[5,1-b]oxazol-7-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (10 mg, 0.016 mmol) followed DCM (0.4 mL) and trifluoroacetic acid (0.12 mL, 1.5 mmol). The reaction mixture was stirred at room temperature for 2.5 h. At this time, the mixture was concentrated and purified by prep HPLC (15% to 45% ACN/H2O, 0.1% TFA modifier, gradient elution) to afford the Example 30 (5.7 mg, 66%) as a white amorphous solid after lyophilization. m/z (ESI, +ve ion)=429.1 [M+H]+.1H NMR (400 MHz, MeOD) δ=7.81 (d, J=8.6 Hz, 1H), 7.52 (s, 1H), 7.38 (d, J=0.8 Hz, 1H), 7.06 (dd, J=1.0, 8.6 Hz, 1H), 6.86 (d, J=8.1 Hz, 1H), 6.67 (dd, J=2.5, 8.6 Hz, 1H), 5.65 (d, J=2.5 Hz, 1H), 5.23-5.17 (m, 2H), 4.38 (t, J=8.0 Hz, 2H), 3.41 (s, 3H), 3.35-3.34 (m, 1H), 2.29-2.16 (m, 2H). Did not observe exchangeable protons. Example 31. (1R,2S)-5′-methoxy-2-{3-[(3-oxo-2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-hydroxy-2,3-dihydro-1-benzofuran-7-yl)amino]indazol-6-yl]-5′-meth oxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((3-((tert-butyldiphenylsilyl)oxy)-2,3-dihydrobenzofuran-7-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (300.00 mg, 0.336 mmol, 1.00 equiv) in THF (5.00 mL) was added TBAF (131.74 mg, 0.504 mmol, 1.50 equiv). After stirred at 25° C. for 2 h. the solvent was removed under reduced pressure. The residue was purified by prep-TLC (rinsed with EA/PE=1/1) to give the title compound (7a) (200 mg, 90.94%) as a light yellow solid. m/z (EST, +ve ion)=655.30 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-oxo-2H-1-benzofuran-7-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-hydroxy-2,3-dihydro-1-benzofuran-7-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (50.00 mg, 0.076 mmol, 1.00 equiv) in DMSO (1.00 mL) was added IBX (42.77 mg, 0.152 mmol, 2.00 equiv). The resulting mixture was stirred at 25° C. for 12 h. The mixture was diluted with EA (50 mL) and washed with brine (30 mL×3). The organic layers were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by prep-TLC (rinsed with EA/PE=1/1) to give the title compound (35 mg, 66.71%) as a light yellow solid. m/z (ESI, +ve ion)=653.30 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.71 (d, J=8.0 Hz, 1H), 8.11 (s, 1H), 7.84-7.82 (m, 1H), 7.58 (d, J=8.4 Hz, 1H), 7.33-7.31 (m, 1H), 7.22-7.19 (m, 1H), 7.12 (d, J=8.4 Hz, 1H), 6.86 (s, 1H), 6.71-6.68 (m, 1H), 5.58 (d, J=2.8 Hz, 1H), 4.77 (s, 2H), 3.59-3.52 (m, 1H), 3.41 (s, 3H), 2.41-2.38 (m, 1H), 2.12 (s, 1H), 1.72 (d, J=9.2 Hz, 18H). Step C. (1R,2S)-5′-methoxy-2-[3-[(3-oxo-2H-1-benzofuran-7-yl)amino]-1H-indazol-6-yl]-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-oxo-2H-1-benzofuran-7-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (25.00 mg, 0.038 mmol, 1.00 equiv) in 1,1,1,3,3,3-hexafluoropropan-2-ol (1.00 mL) was stirred at 50° C. for 12 h. The solvent was removed under reduced pressure and the residue was purified with the following conditions: Column: XBridge Prep OBD C18 Column, 30 Å, 150 mm 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 28% B to 42% B in 8 min; Detector: 254/220 nm; RT1: 7.27 min. The product-containing fractions was concentrated in vacuo to give Example 31 (3.3 mg, 18.85%) as a light yellow solid. m/z (ESI, +ve ion)=453.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.16-8.14 (m, 1H), 7.74 (d, J=8.0 Hz, 1H), 7.38 (s, 1H), 7.16-7.14 (m, 1H), 7.08-7.04 (m, 1H), 6.92 (d, J=8.4 Hz, 1H), 6.84 (d, J=8.8 Hz, 1H), 6.64-6.62 (m, 1H), 5.62 (d, J=2.4 Hz, 1H), 4.82 (s, 2H), 3.39-3.37 (m, 1H), 3.31 (s, 3H), 2.26-2.23 (m, 1H), 2.20-2.17 (m, 1H). Example 32. (1R,2S)-2-{3-[(2,3-dihydrofuro[2,3-c]pyridin-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2H,3H-furo[2,3-c]pyridin-7-amine To a mixture of furo[2,3-c]pyridin-7-amine (400.00 mg, 2.982 mmol, 1.00 equiv) in AcOH (8 mL) was added 10% Pd/C (400.00 mg, 0.376 mmol, 0.13 equiv). The reaction mixture was stirred for 16 h at 1 atm H2atmosphere. The reaction mixture was concentrated under reduced pressure. The residue was dissolved with EA (50 mL) and washed with saturated NaHCO3(3×20 mL). The organic layers were dried over anhydrous Na2SO4, filtered. The filtrate was concentrated under reduced pressure to give the title compound (400 mg, 98.52%) as a yellow oil. m/z (EST, +ve ion)=137.00 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.46 (d, J=5.2 Hz, 1H), 6.53 (d, J=5.2 Hz, 1H), 5.53 (s, 2H), 4.50 (t, J=8.8 Hz, 2H), 3.18-3.08 (m, 2H). Step B. tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl)-3-[2H,3H-furo[2,3-c] pyridin-7-ylamino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (190.00 mg, 0.301 mmol, 1.00 equiv), 2H,3H-furo[2,3-c]pyridin-7-amine (49.16 mg, 0.361 mmol, 1.2 equiv) and Cs2CO3(196.07 mg, 0.602 mmol, 2.00 equiv) in toluene (6.00 mL) were added Pd2(dba)3(27.55 mg, 0.030 mmol, 0.1 equiv) and XantPhos (17.41 mg, 0.030 mmol, 0.10 equiv) under N2atmosphere. The resulting mixture was stirred for 2 h at 90° C. The reaction mixture was filtered. The filtrate was concentrated under reduced pressure. The residue was purified with silica gel chromatography, eluted with 7/o MeOH in DCM to afford the title compound (160 mg, 83.13%) as a yellow oil. m/z (EST, +ve ion)=(40.40 [M+H]+. Step C. (1R,2S)-2-(3-[2H,3H-furo[2,3-c]pyridin-7-ylamino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[2H,3H-furo[2,3-c] pyridin-7-ylamino]indazol-6-yl]-5′-methoxy-2-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (160.00 mg, 0.250 mmol, 1.00 equiv) in DCM (5.00 mL) was added TFA (0.50 mL, 6.732 mmol, 26.91 equiv). The resulting mixture was stirred for 16 h at room temperature. The reaction mixture was concentrated under reduced pressure. The residue was purified by RP flash, eluted with 45% ACN in water (10 mM NH4HCO3) to afford Example 32 (40 mg, 36.39%) as an off-white solid. m/z (ESI, +ve ion)=440.05 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.32 (s, 1H), 10.42 (s, 1H), 8.42 (s, 1H), 7.52 (d, J=5.0 Hz, 1H), 7.42 (d, J=8.4 Hz, 1H), 7.34 (s, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.77-6.73 (m, 2H), 6.59 (dd, J=8.5, 2.6 Hz, 1H), 5.74 (d, J=2.6 Hz, 1H), 4.59 (t, J=8.9 Hz, 2H), 3.3 (s, 3H), 3.22 (t, J=8.9 Hz, 2H), 3.17 (t, J=8.4 Hz, 1H), 2.32 (dd, J=8.0, 4.7 Hz, 1H), 1.98 (dd, J=9.1, 4.6 Hz, 1H). Example 33. (1R,2S)-2-(3-{[(3S)-3-(hydroxymethyl)-2,3-dihydrofuro[2,3-c]pyridin-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 34. (1R,2S)-2-(3-{[(3R)-3-(hydroxymethyl)-2,3-dihydrofuro[2,3-c]pyridin-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 3-[[(tert-butyldimethylsilyl)oxy]methyl]-7-chloro-2H,3H-furo[2,3-c] pyridine To a stirred mixture of [7-chloro-2H,3H-furo[2,3-c]pyridin-3-yl]methanol (500.00 mg, 2.694 mmol, 1.00 equiv) and Imidazole (220.07 mg, 3.233 mmol, 1.20 equiv) in DMF (4.00 mL, 0.055 mmol) were added t-butyldimethylchlorosilane (487.22 mg, 3.233 mmol, 1.20 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was extracted with EtOAc (3×40 mL). The combined organic layers were washed with brine (3×40 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford the crude title product (1.0 g). m/z (ESI, +ve ion)=300.15 [M+H]+. Step B. N-(3-[[(tert-butyldimethylsilyl)oxy]methyl]-2H,3H-furo[2,3-c]pyridin-7-yl)-1,1-diphenylmethanimine To a stirred mixture of 3-[[(tert-butyldimethylsilyl)oxy]methyl]-7-chloro-2H,3H-furo[2,3-c] pyridine (300.00 mg, 1.000 mmol, 1.00 equiv), diphenylmethanimine (36.26 mg, 0.200 mmol, 1.20 equiv) in toluene (5.00 mL, 4.699 mmol, 28.18 equiv) were added t-BuONa (134.60 mg, 1.400 mmol, 1.40 equiv), BINAP (12.46 mg, 0.020 mmol, 0.12 equiv) and Pd2(dba)3(36.64 mg, 0.040 mmol, 0.04 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 90° C. under nitrogen atmosphere. The resulting mixture was extracted with EtOAc (3×40 mL). The combined organic layers were washed with brine (60 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to give 400 mg crude title compound. m/z (ESI, +ve ion)=445.15 [M+H]+. Step C. 3-[[(tert-butyldimethylsilyl)oxy]methyl]-2H,3H-furo[2,3-c]pyridin-7-amine A mixture of N-(3-[[(tert-butyldimethylsilyl)oxy]methyl]-2H,3H-furo[2,3-c]pyridin-7-yl)-1,1-diphenylmethanimine (400.00 mg, 0.900 mmol, 1.00 equiv) and NH2OH (50% in water, 0.50 mL) in MeOH (0.50 mL) and H2O (0.50 mL) was stirred for 1 h at room temperature under nitrogen atmosphere. The resulting mixture was extracted with EtOAc (3×40 mL). The combined organic layers were washed with brine (60 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford the title compound (177 mg, 70.16%) as a white solid. m/z (ESI, +ve ion)=281.25 [M+H]+.1H NMR (400 MHz, DMSO-d) δ 7.47 (d, J=4.8 Hz, 1H), 6.56 (d, J=5.2 Hz, 1H), 5.55 (s, 2H), 4.61-4.51 (m, 1H), 4.35-4.26 (m, 1H), 3.81-3.73 (m, 1H), 3.72-3.64 (m, 11H), 3.63-3.52 (m, 1H), 0.84 (s, 9H), 0 (s, 6H). Step D. tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-[[(tert-butyldimethylsilyl)oxy]methyl]-2H,3H-furo[2,3-c]pyridin-7-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (150.00 mg, 0.238 mmol, 1.00 equiv) and 3-[[(tert-butyldimethylsilyl)oxy]methyl]-2H,3H-furo[2,3-c]pyridin-7-amine (79.94 mg, 0.286 mmol, 1.20 equiv) in toluene (2.30 mL) were added Cs2CO3(154.79 mg, 0.476 mmol, 2.00 equiv), XantPhos (13.74 mg, 0.024 mmol, 0.10 equiv) and Pd2(dba)3·CHCl3(24.59 mg, 0.024 mmol, 0.10 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 85° C. under nitrogen atmosphere then concentrated under vacuum. The resulting mixture was extracted with EtOAc (3×40 mL). The combined organic layers were washed with brine (3×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to give 100 mg of the crude title compound. The crude resulting mixture was used for the next step directly without further purification. m/z (ESI, +ve ion)=784.50 [M+H]+. Step E. (1R,2S)-2-(3-[[3-(hydroxymethyl)-2H,3H-furo[2,3-c]pyridin-7-yl] amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of the crude tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-[[(tert-butyldimethylsilyl)oxy]methyl]-2H,3H-furo[2,3-c]pyridin-7-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100.00 mg, 1 equiv) were added DCM (3.00 mL) and TFA (1.50 mL) at room temperature. The resulting mixture was stirred for 6 h at room temperature, then concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions: Column: Sunfire prep C18 column, 30×150, 5 μm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 5% B to 40% B in 8 min 220 nm; RT1: 7.12 m in to afford the title compound (55 mg, 91.84%) as a white solid. m/z (EST, +ve ion)=470.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.53 (s, 1H), 10.42 (s, 1H), 8.91 (s, 1H), 7.66-7.46 (m, 2H), 7.36 (s, 1H), 6.94-6.79 (m, 2H), 6.75 (d, J=8.4 Hz, 1H), 6.61-6.54 (m, 1H), 5.73 (d, J=2.6 Hz, 1H), 5.04 (s, 1H), 4.77-4.65 (m, 1H), 4.55-4.42 (m, 1H), 3.74-3.53 (m, 3H), 3.22-3.13 (m, 1H), 2.51 (s, 3H), 2.38-2.26 (m, 1H), 2.04-1.92 (m, 1H). Step F: (1R,2S)-2-(3-[[(3S)-3-(hydroxymethyl)-2H,3H-furo[2,3-c] pyridin-7-yl]amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one and (1R,2S)-2-(3-[[(3R)-3-(hydroxymethyl)-2H,3H-furo[2,3-c]pyridin-7-yl]amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one Racemic (1R,2S)-2-(3-[[3-(hydroxymethyl)-2H,3H-furo[2,3-c]pyridin-7-yl] amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one was separated by prep-Chiral-HPLC with the following conditions: Column: Column: CHIRALPAK IG, 2×25 cm, 5 un; Mobile Phase A: Hex (0.2% DEA)-HPLC, Mobile Phase B: EtOH:DCM=1:1-HPLC; Flow rate: 20 mL/min; Gradient: 60% B to 60% B in 12 min; Wavelength: 220/254 nm; RT1(min): 6.603: RT2 (min): 8.571 to afford (first peak) Example 34 (13.9 mg) as a white solid. m/z (EST, +ve ion)=470.15 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.63 (d, J=8.4 Hz, 2H), 7.40 (s, 1H), 6.92 (d, J=7.6 Hz, 2H), 6.83 (d, J=8.4 Hz, 1H), 6.63 (d, J=2.8 Hz, 1H), 6.61 (d, J=2.4 Hz, 1H), 5.66 (d, J=2.4 Hz, 1H), 4.89 (s, 3H), 4.82-4.78 (m, 1H), 4.65-4.63 (m, 1H), 3.85-3.71 (m, 3H), 3.36-3.34 (s, 1H), 2.26-2.22 (m, 1H), 2.19-2.16 (m, 1H). And to afford second peak, the other diastereomer (11.3 mg) as a white solid. m/z (ESI, +ve ion)=470.15 [M+H]+. 1H NMR (400 MHz, Methanol-d4) δ 7.63 (d, J=8.4 Hz, 2H), 7.40 (s, 1H), 6.92 (d, J=7.6 Hz, 2H), 6.83 (d, J=8.4 Hz, 1H), 6.63 (d, J=2.8 Hz, 1H), 6.61 (d, J=2.4 Hz, 1H), 5.66 (d, J=2.4 Hz, 1H), 4.89 (s, 3H), 4.82-4.78 (m, 1H), 4.65-4.63 (m, 1H), 3.85-3.71 (m, 3H), 3.36-3.34 (s, 1H), 2.26-2.22 (m, 1H), 2.19-2.16 (m, 1H). Example 35. (1R,2S)-5′-methoxy-2-(3-{[6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-(3-methoxyazetidin-1-yl)pyrimidin-4-amine) To a mixture of 6-chloropyrimidin-4-amine (516.00 mg, 3.983 mmol, 1.00 equiv) and 3-methoxyazetidine hydrochloride (590.67 mg, 4.800 mmol, 1.20 equiv) in THF (20.00 mL) was added TEA (806.08 mg, 7.966 mmol, 2.00 equiv). The resulting mixture was stirred for 16 h at 60° C. The reaction was concentrated under reduced pressure. The residue was purified by RP flash, eluted with 10% ACN in water (10 mM NH4HCO3) to afford the title compound (200 mg, 27.86%) as a white solid. m/z (ESI, +ve ion)=181.15 [M+H]+.1H NMR (400 MHz, DMSO-dt) δ 7.91 (d, J=1.0 Hz, 1H), 6.23 (s, 2H), 5.23 (d, J=1.0 Hz, 1H), 4.30-4.26 (m, 1H), 4.08-4.04 (m, 2H), 3.69-3.66 (m, 2H), 3.23 (s, 3H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[[6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of 6-(3-methoxyazetidin-1-yl)pyrimidin-4-amine) (35.00 mg, 0.194 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (134.91 mg, 0.214 mmol, 1.1 equiv) in toluene (3.00 mL) were added Cs2CO3(126.56 mg, 0.388 mmol, 2 equiv). Pd2(dba)3(17.78 mg, 0.019 mmol, 0.1 equiv) and XantPhos (11.24 mg, 0.019 mmol, 0.1 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (3×10 mL). The filtrate was removed under reduced pressure and the residue was purified by prep-TLC (rinsed with EA/PE=1/1) to give the title compound (88 mg, 62.95%) as a yellow solid. m/z (ESI, +ve ion)=684.45 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.29 (d, J=1.2 Hz, 1H), 8.13 (s, 1H), 7.81 (d, J=8.8 Hz, 1H), 7.75-7.73 (m, 1H), 7.56-7.54 (m, 1H), 7.13 (d, J=8.4 Hz, 1H), 6.70-6.67 (m, 1H), 5.55 (d, J=2.4 Hz, 1H), 4.41-4.38 (m, 3H), 4.11 (d, J=8.0 Hz, 2H), 3.52 (t, J=8.8 Hz, 1H), 3.40 (d, J=8.8 Hz, 6H), 2.39-2.36 (m, 1H), 2.12 (s, 1H), 1.71 (d, J=4.4 Hz, 18H). Step C. (1R,2S)-5′-methoxy-2-(3-[[6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl] amino]-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[[6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.146 mmol, 1.00 equiv) in TFA (0.50 mL) and DCM (5.00 mL) was stirred at 25° C. for 2 h. The mixture was quenched with saturated aqueous of NaHCO2(20 mL) and extracted with EA (3×20 mL). The combined organic was dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The product was purified with the following conditions: Column: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3). Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 18% B to 35% B in 8 min; Detector: 254 and 220 nm; RT1: 7.43 min. The product-containing fractions were combined and concentrated to give Example 35 (32.3 mg, 45.7%) as a white solid. m/z (ESI, +ve ion)=484.20 [M+H].1H NMR (400 MHz, DMSO-4) δ 12.37 (s, 1H), 10.42 (s, 1H), 9.82 (s, 1H), 8.15 (d, J=0.8 Hz, 1H), 7.95 (d, J=8.4 Hz, 1H), 7.33 (s, 1H), 6.90-6.88 (m, 1H), 6.78-6.74 (m, 2H), 6.60-6.57 (m, 1H), 5.69 (d, J=2.8 Hz, 1H), 4.37-4.31 (m, 1H), 4.19-4.15 (m, 2H), 3.80-3.76 (m, 2H), 3.33 (s, 3H), 3.26 (s, 3H), 3.17 (t, J=8.4 Hz, 1H), 2.35-2.32 (m, 1H), 1.99-1.95 (m, 1H). Example 36. (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 3-[(tert-butyldimethylsilyl)oxy]azetidine To a stirred solution of azetidin-3-ol (1.09 g, 14.912 mmol, 1.00 equiv) and t-butyldimethylchlorosilane (2.25 g, 14.912 mmol, 1.00 equiv) in DCM (10.00 mL) was added DIEA (4.82 g, 37.280 mmol, 2.50 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere, then concentrated under vacuum. The residue was neutralized to pH 8 with saturated NaHCO3. The resulting mixture was extracted with EtOAc (2×100 mL). The combined organic layers were washed with brine (3×40 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to give the title compound (12a) (1.5 g, 53.69%) as a yellow oil.1H NMR (400 MHz, Chloroform-d) δ 4.50-4.53 (m, 1H), 3.47-3.44 (m, 2H), 2.50-2.52 (s, 2H), 0.85-0.87 (m, 9H), 0.02-0.04 (m, 6H). Step B. 6-[3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl]pyrimidin-4-amine To a stirred mixture of 3-[(tert-butyldimethylsilyl)oxy]azetidine (299.37 mg, 1.598 mmol, 1.50 equiv) and 6-chloropyrimidin-4-amine (138.00 mg, 1.065 mmol, 1.00 equiv) in THF (0.50 mL) was added TEA (161.40 mg, 1.598 mmol, 1.50 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 60° C., then cooled down to room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (3/1) to afford the title compound (256 mg, 85.69%) as a yellow solid. m/z (ESI, +ve ion)=281.20 [M+H]+. Step C. (1R,2S)-5′-methoxy-2-(3-[[5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl](methyl)amino]-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To the mixture of 6-[3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl]pyrimidin-4-amine (60.00 mg, 0.214 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (135.10 mg, 0.214 mmol, 1 equiv) in toluene (2.50 mL) were added Cs2CO3(139.41 mg, 0.428 mmol, 2 equiv), Pd2(dba)3(19.59 mg, 0.021 mmol, 0.1 equiv) and XantPhos (12.38 mg, 0.021 mmol, 0.1 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (3×10 mL). The filtrate was removed under reduced pressure and the residue was purified by silica gel column eluted with EA/PE=1/1 to give the title compound (100 mg, 56.64%) as an off-white solid. m/z (ESI, +ve ion)=784.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 10.45 (s, 1H), 8.28-8.24 (m, 2H), 8.04 (s, 1H), 7.68 (d, J=8.8 Hz, 1H), 7.36-7.34 (m, 1H), 7.26 (d, J=1.2 Hz, 1H), 6.74-6.71 (m, 1H), 5.71 (d, J=2.8 Hz, 1H), 4.85-4.80 (m, 1H), 4.31-4.28 (m, 2H), 3.80-3.74 (m, 2H), 3.44-3.40 (m, 1H), 3.35 (s, 3H), 2.48-2.45 (m, 1H), 2.21-2.17 (m, 1H), 1.60 (d, J=6.4 Hz, 18H), 0.88 (s, 9H), 0.08 (s, 6H). Step D. (1R,2S)-2-(3-[[6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of (1R,2S)-5′-methoxy-2-(3-[[5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl](methyl)amino]-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one (100.00 mg, 0.128 mmol, 1.00 equiv) in TFA (0.50 mL) and DCM (2.50 mL) was stirred at 25° C. for 12 h. The solvent was removed under reduced pressure. The residue was further purified with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 10% B to 40% B in 7 min; Detector: 254 & 220 nm; RT1: 7.27 min. The product-containing fractions were combined and concentrated to give Example 36 (33.4 mg, 58.12%) as a white solid. m/z (ESI, +ve ion)=470.35 [M+H].1H NMR (400 MHz, DMSO-d6) δ 12.38 (s, 1H), 10.43 (s, 1H), 9.81 (s, 1H), 8.14 (d, J=0.8 Hz, 1H), 7.94 (d, J=8.4 Hz, 1H), 7.33 (s, 1H), 6.90-6.87 (m, 1H), 6.77-6.73 (m, 2H), 6.59-6.56 (m, 1H), 5.75 (d, J=6.8 Hz, 1H), 5.68 (d, J=2.4 Hz, 1H), 4.58 (s, 1H), 4.20-4.16 (m, 2H), 3.72-3.69 (m, 2H), 3.32 (s, 3H), 3.17 (t, J=8.4 Hz, 1H), 2.35-2.32 (m, 1H), 1.98-1.95 (m, 1H). Example 37. (1R,2S)-2-(3-((6-(3-oxa-8-azabicyclo[3.2.1]octan-8-yl)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 5-(6-chloro-5-methoxy-pyrimidin-4-yl)-2-oxa-5-azabicyclo[2.2.2]octane To a 50 ml round bottom flask containing 4,6-Dichloro-5-methoxypyrimidine (200.0 mg, 1.120 mmol) in DMSO (3.7 mL) were added (1R,5S)-3-Oxa-8-azabicyclo[3.2.1]octane hydrochloride (1:1) (167.2 mg, 1.120 mmol) and N-ethyl-N-isopropylpropan-2-amine (0.50 mL, 2.8 mmol). The reaction mixture was heated to 60° C. and stirred, monitored by LCMS until the full conversion of the starting materials (approx. 80 min). Then the reaction mixture was cooled down to rt, diluted with EtOAc and water, extracted with EtOAc for 3 times. The organic layer was then dried over Na2SO4. The residue was purified by column chromatography (ethyl acetate/hexane=0˜50%) to provide the title compound (228.0 mg, 80%) as a white solid. Step B. tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[5-methoxy-6-(2-oxa-5-azabicyclo[2.2.2]octan-5-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a 50 ml round bottom flask were added cesium carbonate (37.6 mg, 0.115 mmol), tert-butyl (1R,2S)-2-(3-amino-1-tert-butoxycarbonyl-indazol-6-yl)-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (30.0 mg, 0.0600 mmol), Tris(dibenzylideneacetone)dipalladium(0) (5.3 mg, 0.010 mmol), 5-(6-chloro-5-methoxy-pyrimidin-4-yl)-2-oxa-5-azabicyclo[2.2.2]octane (15.4 mg, 0.0605 mmol), Xantphos (3.3 mg, 0.010 mmol) and dry toluene (2.9 mL). The reaction mixture was stirred and purged with argon (in balloon) for 10 min to form a green suspension, and then heated to 120° C., resulting in a yellow suspension. The reaction was monitored by LCMS and TLC until the full conversion of the starting materials (approx. 5 hrs), cooled down to rt, diluted with EtOAc, washed with sat. aq. NaHCO3and dried over Na2SO4. The residue was purified by column chromatography (ethyl acetate/hexane=0˜50%) to provide the title compound (16.0 mg, 38%) as a yellow oil. Step C This compound was prepared from tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[5-methoxy-6-(2-oxa-5-azabicyclo[2.2.2]octan-5-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (16.0 mg, 0.0200 mmol) and trifluoroacetic acid (0.17 mL, 2.2 mmol). The resulting brown solution was purified by Prep. HPLC (Gemini C18, 10 to 90% (0.1% TFA in water)/(0.1% TFA in Acetonitrile)) to provide Example 37 (7.8 mg, 67%) as a colorless film.1H NMR (400 MHz, METHANOL-d4) δ ppm 2.08-2.14 (m, 2H) 2.15-2.27 (m, 5H) 3.34-3.36 (m, 3H) 3.70-3.75 (m, 2H) 3.79-3.84 (m, 5H) 4.98-5.03 (m, 2H) 5.61 (d, J=2.27 Hz, 1H) 6.62 (dd, J=8.59, 2.53 Hz, 1H) 6.83 (d, J=8.84 Hz, 1H) 7.03 (dd, J=8.46, 0.88 Hz, 1H) 7.48-7.51 (m, 1H) 7.87-7.91 (m, 1H) 8.22 (s, 1H); m/z (ESI, +ve ion) 540.2 (M+H)+. Example 38. (1R,2S)-2-(3-{[6-(2-hydroxyethoxy)pyrimidin-4-yl]amino}-1H-indazol-4-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-[2-[(tert-butyldimethylsilyl)oxy]ethoxy] pyrimidin-4-amine To a stirred mixture of 6-chloropyrimidin-4-amine (600.00 mg, 4.631 mmol, 1.00 equiv) and 2-[(tert-butyldimethylsilyl)oxy]ethanol (1224.99 mg, 0.000 mmol, 1.50 equiv) in THF (15.00 mL) was added NaH (222.29 mg, 9.262 mmol, 2.00 equiv, 60/o in mineral oil) in portions at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 100° C. under nitrogen atmosphere, then cooled down to room temperature. The reaction was quenched with sat. NH4Cl (30 mL) at room temperature. The resulting mixture was extracted with DCM (3×20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase. ACN in water, 30% to 70% gradient in 60 min-; detector, UV 254 nm to give the title compound (164 mg, 13.14%) as a pink solid. m/z (ESI, +ve ion)=270.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J=1.0 Hz, 1H), 6.60 (s, 2H), 5.68 (d, J=1.1 Hz, 1H), 4.58-4.07 (m, 2H), 3.86 (m, 2H), 0.86 (d, J=1.4 Hz, 9H), 0.12-0.03 (m, 6H). Step B. tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-[2-[(tert-butyldimethylsilyl)oxy]ethoxy]pyrimidin-4-yl)amino]indazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate To a stirred solution of 6-[2-[(tert-butyldimethylsilyl)oxy]ethoxy] pyrimidin-4-amine (51.15 mg, 1.2 equiv) and tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (100 mg, 1.00 equiv) in toluene (3 mL) were added Pd2(dba), (14.50 mg, 0.1 equiv), XantPhos (9.16 mg, 0.1 equiv) and Cs2CO3(103.26 mg, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was cooled down and filtered, the filter cake was washed with MeOH (2×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (2/1) to afford the title compound (110 mg, 89.86%) as a yellow solid. m/z [ESI+ve ion]=773.35 [M+H]+. Step C. (1R,2S)-2-(3-[[6-(2-hydroxyethoxy)pyrimidin-4-yl]amino]-1H-indazol-6-yl)-5-methoxy-1H-spiro[cyclopropane-1,3-indol]-2-one A mixture of tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-[2-[(tert-butyldimethylsilyl)oxy]ethoxy]pyrimidin-4-yl)amino]indazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (130.00 mg, 0.168 mmol, 1.00 equiv) and TFA (2.00 mL, 0.018 mmol, 0.10 equiv) in DCM (10.00 m L) was stirred for 4 h at room temperature. The mixture was neutralized to pH 7 with saturated NaHCO3. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions Column: YMC-Actus Triart C18 ExRS, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 30% B in 10 min, 30% B; Wavelength: 254 nm; RT1: 8.5 min to afford Example 38 (8.2 mg, 25.94%) as a white solid. m/z (ESI+ve ion)=459.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.42 (s, 1H), 10.42 (s, 1H), 10.19 (s, 1H), 8.36 (d, J=0.8 Hz, 1H), 7.95 (d, J=8.4 Hz, 1H), 7.36 (s, 1H), 7.24 (s, 1H), 6.92 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (m, 1H), 5.69 (d, J=2.4 Hz, 1H), 4.88 (s, 1H), 4.30 (t, J=4.8 Hz, 2H), 3.70 (d, J=4.4 Hz, 2H), 3.30 (s, 3H), 3.20 (s, 1H), 2.33 (s, 1H), 1.98 (m, 1H). Example 39. (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 4-(6-aminopyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione Into a 20 mL vial were added 6-chloropyrimidin-4-amine (300.00 mg, 2.316 mmol, 1.00 equiv) and 1lambda 6-thiomorpholine-1,1-dione (939.11 mg, 6.947 mmol, 3.00 equiv) at room temperature. The resulting mixture was stirred overnight at 80° C. under nitrogen atmosphere. After cooled down to room temperature, the residue was purified by silica gel column chromatography, eluted with PE/EtOAc (3/1) to afford the title compound (110 mg, 19.21%) as a yellow solid. m/z (ESI, +ve ion)=229.10 [M+H]+.1H NMR (400 MHz, DMSO-db) δ 8.01 (d, J=0.9 Hz, 1H), 6.34 (s, 2H), 5.75 (d, J=1.1 Hz, 1H), 4.00-3.88 (m, 4H), 3.15-3.06 (m, 4H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[[6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (100.00 mg, 0.158 mmol, 1.00 equiv) and 4-(6-aminopyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (43.38 mg, 0.190 mmol, 1.20 equiv) in toluene (1.00 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2.00 equiv), Pd2(dba)3(14.50 mg, 0.016 mmol, 0.10 equiv) and XantPhos (9.16 mg, 0.016 mmol, 0.10 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere and then cooled down to room temperature. The resulting mixture was diluted with water (20 mL), extracted with EtOAc (3×40 mL). The combined organic layers were washed with brine (40 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (100 mg, 86.29%) as an off-white solid. m/z (ESI, +ve ion)=732.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 10.57 (s, 1H), 8.36 (d, J=0.9 Hz, 1H), 8.27 (d, J=8.3 Hz, 1H), 8.05 (d, J=14.3 Hz, 1H), 7.77 (s, 1H), 7.68 (d, J=8.9 Hz, 1H), 7.40-7.33 (m, 1H), 6.74-6.69 (m, 1H), 5.70 (d, J=2.7 Hz, 1H), 4.20-3.99 (m, 4H), 3.27-3.08 (m, 8H), 1.60 (d, J=5.2 Hz, 18H), 1.23 (s, 2H). Step C. 4-[6-([6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl]amino)pyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[[6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100.00 mg) in DCM (0.80 mL) and TFA (0.20 mL) was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 32% B in 8 min, 254/220 nm; RT1: 7.05 min to afford Example 39 (15.1 mg) as a white solid m/z (ESI, +ve ion)=532.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.41 (s, 1H), 10.42 (s, 1H), 9.93 (s, 1H), 8.26 (s, 1H), 7.94 (d, J=8.4 Hz, 1H), 7.34 (s, 1H), 7.27 (s, 1H), 6.90 (d, J=7.6 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.69 (d, J=2.8 Hz, 1H), 4.04 (s, 4H), 3.33 (s, 3H), 3.19-3.16 (m, 5H), 2.35-2.32 (m, 1H), 1.99-1.96 (m, 1H). Example 40. (1R,2S)-5′-methoxy-2-(3-{[6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-(1,4-oxazepan-4-yl)pyrimidin-4-amine A mixture of 6-chloropyrimidin-4-amine (516.00 mg, 1 equiv) in 1,4-oxazepane (3.00 mL) was stirred for 16 h at 60° C. The mixture was purified with RP flash, eluted with RP flash, eluted with 10% ACN in water (10 mM NH4HCO3) to afford the title compound (300 mg, 38.78%) as a white solid. m/z (ESI+ve ion)=195.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.92 (d, J=1.0 Hz, 1H), 6.12 (s, 2H), 5.50 (d, J=1.0 Hz, 1H), 3.66 (s, 4H), 3.63-3.55 (m, 4H), 1.88-1.78 (m, 2H). Step B. tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl)-3-[[6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (200.00 mg, 0.317 mmol, 1.00 equiv) and 6-(1,4-oxazepan-4-yl)pyrimidin-4-amine (73.82 mg, 0.380 mmol, 1.2 equiv) in toluene (5.00 mL) were added Cs2C03(206.39 mg, 0.633 mmol, 2 equiv), Pd2(dba)3(29.00 mg, 0.032 mmol, 0.1 equiv) and XantPhos (18.33 mg, 0.032 mmol, 0.1 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (3×10 mL). The filtrate was removed under reduced pressure and the residue was purified by silica gel column eluted with EA/PE=1/1 to give the title compound (80 mg, 34.39%) as a yellow solid. m/z (ESI, +ve ion)=698.50 [M+H].1H NMR (400 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.31-8.27 (m, 2H), 8.04 (s, 1H), 7.72-7.68 (m, 1H), 7.60 (s, 1H), 7.36 (d, J=8.8 Hz, 1H), 6.73 (d, J=9.6 Hz, 1H), 5.71 (s, 1H), 3.89-3.71 (m, 6H), 3.66-3.62 (m, 2H), 3.45-3.43 (m, 1H), 3.35 (s, 3H), 2.34 (s, 1H), 2.22-2.18 (m, 1H), 1.92 (s, 2H), 1.60 (s, 18H). Step C. (1R,2S)-5′-methoxy-2-(3-[[6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino]-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[[6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.00 mg, 0.115 mmol, 1.00 equiv) in TFA (0.50 mL) and DCM (5.00 mL) was stirred at 25° C. for 2 h. The mixture was quenched with saturated aqueous of NaHCO3(20 mL) and extracted with EA (3×20 mL). The combined organics were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The product was purified with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20/o B to 45% B in 8 min; Detector: 254&220 nm; RT1: 5.73 min. The product-containing fractions were combined and concentrated to give Example 40 (19.3 mg, 33.50%) as a white solid. m/z (ESI, +ve ion)=498.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.37 (s, 1H), 10.43 (s, 1H), 9.75 (s, 1H), 8.17 (s, 1H), 7.96 (d, J=8.4 Hz, 1H), 7.33 (s, 1H), 7.12 (s, 1H), 6.90-6.88 (m, 1H), 6.75 (d, J=8.6 Hz, 1H), 6.60-6.57 (m, 1H), 5.69 (d, J=2.4 Hz, 1H), 3.78-3.68 (m, 6H), 3.63 (t, J=5.6 Hz, 2H), 3.33 (s, 3H), 3.18 (t, J=8.0 Hz, 1H), 2.35-2.32 (m, 1H), 1.99-1.96 (m, 1H), 1.90-1.88 (m, 2H). Example 41. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-methyl-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-Hydroxy-5-methoxy-2-methylpyrimidin-4 (3H)-one To an oven-dried flask containing methanol (100 mL) at 0° C. was added sodium tert-butoxide (12.2 g, 127 mmol) in four-portions over 20 minutes. To this solution was added acetimidamide hydrochloride (4.5 g, 50.9 mmol) and dimethyl methoxymalonate (7.0 mL, 51 mmol). The mixture was heated to reflux for 2 h and then cooled to 0° C. The reaction mixture was made acidic with concentrated HCl and the resulting solid was collected by vacuum filtration. The wet material was frozen and lyophilized to afford the title compound (2.4 g, 30%) as a beige solid. m/z (ESI, +ve ion)=157.1 [M+H]+. Step B. 4,6-Dichloro-5-methoxy-2-methylpyrimidine To a microwave tube was added 6-hydroxy-5-methoxy-2-methylpyrimidin-4 (3H)-one (2.40 g, 15.4 mmol), N,N-dimethylaniline (1.94 mL, 15.4 mmol), and phosphoryl chloride (12.7 mL, 135 mmol) and the mixture was heated to 100° C. in a microwave reactor for 1 h. The reaction mixture was filtered and concentrated. The resulting material was purified by column chromatography (0% to 25% EtOAc/hexanes, gradient elution) to afford the title compound (2.6 g, 87%) as a white solid. Step C. 4-(6-Chloro-5-methoxy-2-methylpyrimidin-4-yl)morpholine To an oven-dried flask was added 4,6-Dichloro-5-methoxy-2-methylpyrimidine (1.0 g, 5.18 mmol) followed by ethanol (30 mL). The mixture was cooled to 0° C. morpholine (0.54 mL, 6.2 mmol) was added followed by TEA (1.0 mL, 7.3 mmol), dropwise. The reaction mixture was stirred at room temperature for 1 h and concentrated. The residue was purified by column chromatography (0% to 30% EtOAc/hexanes, gradient elution) to provide the title compound (909 mg, 72%) as a white crystalline solid. m/z (ESI, +ve ion)=244.1 [M+H]+. Step D. Tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-methoxy-2-methyl-6-morpholinopyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To an oven-dried flask was added 4-(6-Chloro-5-methoxy-2-methylpyrimidin-4-yl)morpholine (28.1 mg, 0.115 mmol), tert-butyl (1R,2S)-2-(3-amino-1-(tert-butoxycarbonyl)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (30.0 mg, 0.0576 mmol), Xantphos Pd G4 (5.6 mg, 0.0058 mmol), and 1,4-dioxane (0.6 mL). The mixture was degassed with bubbling argon for 10 min. At this time, Cs2CO3(37.6 mg, 0.115 mmol) was added and the reaction mixture was heated to 100° C. for 2 h. The reaction mixture was cooled was cooled to room temperature and diluted with EtOAc and washed with sat. aqueous NaHCO3. The aqueous layer was extracted an additional three times with EtOAc. Combined organic layers were washed with brine, dried with MgSO4, filtered, and concentrated. The residue was purified by column chromatography (0% to 50% acetone/hexanes, a gradient elution) to provide the title compound (25 mg, 59%) as a white foam. m/z (ESI, +ve ion)=728.3 [M+H]+. Step E To an oven-dried flask was added tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-methoxy-2-methyl-6-morpholinopyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1-carboxylate (25 mg, 0.034 mmol) followed DCM (1.7 mL) and trifluoroacetic acid (0.13 mL, 1.7 mmol). The reaction mixture was stirred at room temperature for 2 h. At this time, the mixture was concentrated and purified by prep HPLC (20% to 40% ACN/H2O, 0.1% TFA modifier, gradient elution) to afford Example 41 (8.1 mg, 45%) as a white amorphous solid after lyophilization. m/z (ESI, +ve ion)=528.2 [M+H]+.1H NMR (400 MHz, DMSO-d)=12.88 (br s, 1H), 10.45 (s, 1H), 9.90 (br s, 1H), 7.66 (d, J=8.3 Hz, 1H), 7.45 (s, 1H), 6.97 (d, J=8.6 Hz, 1H), 6.75 (d, J=8.3 Hz, 1H), 6.59 (dd, J=2.7, 8.5 Hz, 1H), 5.74 (d, J=2.5 Hz, 1H), 3.81-3.70 (m, 8H), 3.67 (s, 3H), 3.35 (s, 3H), 3.19 (t, J=8.5 Hz, 1H), 2.35 (dd, J=4.7, 8.0 Hz, 1H), 2.27 (s, 3H), 1.99 (dd, J=4.7, 9.0 Hz, 1H). Example 42. (1R,2S)-2-(3-{[6-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-(6-chloro-5-methoxy-pyrimidin-4-yl)-2-oxa-5-azabicyclo[2.2.2]octane This compound was prepared from 4,6-dichloro-5-methoxypyrimidine (200.0 mg, 1.120 mmol) and azetidine (63.4 mg, 0.08 mL, 1.12 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0˜50%) to provide the title compound (179.0 mg, 80%) as a white solid. Step B. tert-butyl (1R,2S)-2-[3-[[6-(azetidin-1-yl)-5-methoxy-pyrimidin-4-yl]amino]-1-tert-butoxycarbonyl-indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-(3-amino-1-tert-butoxycarbonyl-indazol-6-yl)-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (30.0 mg, 0.0600 mmol) and 5-(6-chloro-5-methoxy-pyrimidin-4-yl)-2-oxa-5-azabicyclo[2.2.2]octane (12.1 mg, 0.0605 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0˜80%) to provide the title compound (3.0 mg, 7.6%) as a yellow oil. Step C. (1R,2S)-2-[3-[[6-(azetidin-1-yl)-5-methoxy-pyrimidin-4-yl]amino]-1H-indazol-6-yl]-5′-methoxy-spiro[cyclopropane-1,3′-indoline]-2′-one This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-[3-[[6-(azetidin-1-yl)-5-methoxy-pyrimidin-4-yl]amino]-1-tert-butoxycarbonyl-indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (3.0 mg, 0.0044 mmol) and trifluoroacetic acid (0.03 mL, 0.4 mmol). The resulting brown solution was purified by Prep. HPLC (Gemini C18, 10 to 90% (0.1% TFA in water)/(0.1% TFA in Acetonitrile)) to provide the desired product Example 42 (1.7 mg, 81%) as a colorless film.1H NMR (400 MHz, METHANOL-d4) δ ppm 2.15-2.22 (m, 1H) 2.24 (dd, J=7.83, 4.80 Hz, 1H) 2.53 (quin, J=7.71 Hz, 2H) 3.33 (s, 3H) 3.33-3.38 (m, 1H) 3.83 (s, 3H) 4.42-4.59 (m, 4H) 5.55-5.63 (m, 1H) 6.58-6.66 (m, 1H) 6.78-6.89 (m, 1H) 6.96-7.08 (m, 1H) 7.42-7.50 (m, 1H) 7.80-7.91 (m, 1H) 8.11-8.19 (m, 1H); m/z (EST, +ve ion) 484.3 (M+H)+. Example 43. (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 1-(6-chloro-5-methoxy-pyrimidin-4-yl)azetidin-3-ol This compound was prepared using 4,6-dichloro-5-methoxypyrimidine (200.0 mg, 1.120 mmol) and azetidin-3-ol Hydrochloride (122.4 mg, 1.120 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0˜100%) to provide the title compound (216.0 mg, 90%) as a white solid. Step B. tert-butyl-[1-(6-chloro-5-methoxy-pyrimidin-4-yl)azetidin-3-yl]oxy-dimethyl-silane To a 50 ml round bottom flask containing 1-(6-chloro-5-methoxy-pyrimidin-4-yl)azetidin-3-ol (143.0 mg, 0.6600 mmol) in DCM (3.3 mL) were added tert-butylchlorodimethylsilane (119.9 mg, 0.8000 mmol) and Imidazole (112.9 mg, 1.660 mmol). The reaction mixture was stirred at rt and monitored by LCMS until the full conversion of the starting materials (approx. 80 min), cooled down to it, diluted with EtOAc and water, extracted with EtOAc for 3 times. The organic layer was then dried over Na2SO4. The residue was purified by column chromatography (ethyl acetate/hexane=0˜50%) to provide the title compound (180.0 mg, 82%) as a colorless oil. Step C. tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[6-[3-[tert-butyl(dimethyl)silyl]oxyazetidin-1-yl]-5-methoxy-pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (5c) This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-(3-amino-1-tert-butoxycarbonyl-indazol-6-yl)-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (30.0 mg, 0.0600 mmol) and tert-butyl-[1-(6-chloro-5-methoxy-pyrimidin-4-yl)azetidin-3-yl]oxy-dimethyl-silane (20.9 mg, 0.0605 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0˜50%) to provide the title compound (6.0 mg, 13%) as a yellow oil. Step D. (1R,2S)-2-[3-[[6-(3-hydroxyazetidin-1-yl)-5-methoxy-pyrimidin-4-yl]amino]-1H-indazol-6-yl]-5′-methoxy-spiro[cyclopropane-1,3′-indoline]-2′-one This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[6-[3-[tert-butyl(dimethyl)silyl]oxyazetidin-1-yl]-5-methoxy-pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (6.0 mg, 0.010 mmol) and trifluoroacetic acid (0.06 mL, 0.7 mmol). The resulting brown solution was purified by Prep. HPLC (Gemini C18, 20 to 40% (0.1% TFA in water)/(0.1% TFA in Acetonitrile)) to provide the desired product Example 43 (2.3 mg, 62%) as a colorless film.1H NMR (400 MHz, METHANOL-d4) δ ppm 2.16-2.21 (m, 1H) 2.25 (dd, J=7.96, 4.93 Hz, 1H) 3.33 (s, 3H) 3.33-3.38 (m, 1H) 3.83 (s, 3H) 4.16-4.27 (m, 2H) 4.61-4.86 (m, 3H) 5.60 (d, J=2.27 Hz, 1H) 6.62 (dd, J=8.59, 2.53 Hz, 1H) 6.83 (d, J=8.08 Hz, 1H) 7.03 (dd, J=8.59, 1.01 Hz, 1H) 7.48 (d, J=1.01 Hz, 1H) 7.87 (d, J=8.34 Hz, 1H) 8.17 (s, 1H); m/z (ESI, +ve ion) 500.1 (M+H)+. Example 44. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-(6-chloro-5-methoxy-pyrimidin-4-yl)-1,4-oxazepane This compound was prepared using 4,6-Dichloro-5-methoxypyrimidine (200.0 mg, 1.120 mmol) and 1,4-Oxazepane (113.0 mg, 1.120 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0˜50%) to provide the title compound (203.0 mg, 75%) as a colorless oil. Step B. tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[5-methoxy-6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-(3-amino-1-tert-butoxycarbonyl-indazol-6-yl)-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (30.0 mg, 0.0600 mmol) and 4-(6-chloro-5-methoxy-pyrimidin-4-yl)-1,4-oxazepane (14.7 mg, 0.0605 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0˜60%) to provide the title compound (13.0 mg, 31%) as a yellow oil. Step C. (1R,2S)-5′-methoxy-2-[3-[[5-methoxy-6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino]-1H-indazol-6-yl]spiro[cyclopropane-1,3′-indoline]-2′-one This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[5-methoxy-6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (13.0 mg, 0.0179 mmol) and trifluoroacetic acid (0.14 mL, 1.8 mmol). The resulting brown solution was purified by Prep. HPLC (Gemini C18, 10 to 90% (0.1% TFA in water)/(0.1% TFA in Acetonitrile)) to provide Example 44 (6.8 mg, 72%) as a colorless film.1H NMR (400 MHz, METHANOL-d4) δ ppm 1.98-2.05 (m, 2H) 2.16-2.21 (m, 1H) 2.25 (dd, J=7.96, 4.93 Hz, 1H) 3.33 (s, 3H) 3.34-3.38 (m, 1H) 3.77-3.81 (m, 2H) 3.82 (s, 3H) 3.87 (t, J=5.31 Hz, 2H) 4.04-4.17 (m, 4H) 5.60 (d, J=2.53 Hz, 1H) 6.62 (dd, J=8.59, 2.53 Hz, 1H) 6.83 (d, J=8.34 Hz, 1H) 7.04 (dd, J=8.59, 1.01 Hz, 1H) 7.50 (d, J=1.01 Hz, 1H) 7.91 (dd, J=8.59, 0.76 Hz, 1H) 8.24 (s, 1H); m/z (ESI, +ve ion) 528.2 (M+H)+. Example 45. (1R,2S)-2-(3-{[6-(azetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-(azetidin-1-yl)-6-chloro-pyrimidine This compound was prepared from 4,6-dichloropyrimidine (200.0 mg, 1.340 mmol) and azetidine (76.6 mg, 0.09 mL, 1.34 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0-50%) to provide the title compound (148.0 mg, 65%) as a white solid. Step B. tert-butyl (1R,2S)-2-[3-[[6-(azetidin-1-yl)pyrimidin-4-yl]amino]-1-tert-butoxycarbonyl-indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-(3-amino-1-tert-butoxycarbonyl-indazol-6-yl)-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (36.0 mg, 0.0700 mmol) and 4-(azetidin-1-yl)-6-chloro-pyrimidine (12.3 mg, 0.0700 mmol). The residue was purified by column chromatography (ethyl acetate/hexane=0˜90%) to provide the title compound (5.0 mg, 11%) as a yellow oil. Step C. (1R,2S)-2-[3-[[6-(azetidin-1-yl)pyrimidin-4-yl]amino]-1H-indazol-6-yl]-5′-methoxy-spiro[cyclopropane-1,3′-indoline]-2′-one This compound was prepared using the procedure described in Example 5 from tert-butyl (1R,2S)-2-[3-[[6-(azetidin-1-yl)pyrimidin-4-yl]amino]-1-tert-butoxycarbonyl-indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (5.0 mg, 0.0044 mmol) and trifluoroacetic acid (0.06 mL, 0.8 mmol). The resulting brown solution was purified by Prep. HPLC (Gemini C18, 10 to 90/o (0.1% TFA in water)/(0.1% TFA in Acetonitrile)) to provide the desired product Example 45 (1.4 mg, 40%) as a colorless film.1H NMR (400 MHz, METHANOL-d4) δ ppm 2.15-2.20 (m, 1H) 2.24 (dd, J=7.96, 4.93 Hz, 1H) 2.49-2.59 (m, 2H) 3.32 (s, 3H) 3.33-3.37 (m, 1H) 4.16-4.44 (m, 4H) 5.58 (d, J=2.53 Hz, 1H) 5.82-6.34 (m, 1H) 6.62 (dd, J=8.59, 2.53 Hz, 1H) 6.83 (d, J=8.34 Hz, 1H) 7.01 (dd, J=8.72, 0.88 Hz, 1H) 7.45 (d, J=0.76 Hz, 1H) 7.77 (d, J=8.59 Hz, 1H) 8.35 (s, 1H); m/z (ESI, +ve ion) 454.3 (M+H)+. Example 46. (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-[3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl]-5-chloropyrimidin 4-amine A mixture of 6-(3-((tert-butyldimethylsilyl)oxy)azetidin-1-yl)pyrimidin-4-amine (110.00 mg, 0.392 mmol, 1.00 equiv) and NCS (62.85 mg, 0.470 mmol, 1.20 equiv) in AcOH (0.60 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford the title compound (70 mg, 56.68%) as a yellow solid. m/z (ESI, +ve ion)=315.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.91 (d, J=1.0 Hz, 1H), 6.22 (s, 2H), 4.72-4.77 (m, 1H), 4.11-4.15 (m, 2H), 3.58-3.61 (m, 2H), 0.88 (s, 9H), 0.08 (s, 6H). Step B To a stirred mixture of 6-[3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl]-5-chloropyrimidin 4-amine (47.77 mg, 0.152 mmol, 1.20 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (80.00 mg, 0.127 mmol, 1.00 equiv) in toluene (5.00 mL) were added XantPhos (7.34 mg, 0.013 mmol, 0.10 equiv), Pd2(dba), (11.61 mg, 0.013 mmol, 0.1 equiv) and Cs2CO3(82.62 mg, 0.254 mmol, 2.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 90° C. under nitrogen atmosphere. The product was diluted by water (10 mL), extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (3×5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford the title compound (15 mg, 14.96%) as a white solid. m/z (ESI, +ve ion)=818.45 [M+H]+. Step C. (1R,2S)-2-(3-[[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl] amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A mixture of the compound from Step B (80.00 mg, 0.098 mmol, 1.00 equiv) and TFA (0.20 mL) in DCM (1 mL) was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (5 mM, NH4HCO3), gradient: 10% to 50% in 60 min to afford Example 46 (5.03 mg, 10%) as a white solid. m/z (ESI, +ve ion)=504.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.68 (s, 1H), 10.43 (s, 1H), 8.91 (s, 1H), 7.83 (d, J=4 Hz, 1H), 7.40 (s, 1H), 7.35 (d, J=8.4 Hz, 1H), 6.90 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59-6.57 (m, 1H), 5.72-5.68 (m, 2H), 4.52-4.43 (m, 3H), 3.99-3.96 (m, 2H), 3.18-3.16 (m, 1H), 3.34 (s, 3H), 2.53-2.50 (m, 1H), 1.99-1.95 (m, 1H). Example 47. (1R,2S)-2-(3-{[5-chloro-6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-chloro-6-(3-methoxyazetidin-1-yl) pyrimidin-4-amine A mixture of 6-(3-methoxyazetidin-1-yl)pyrimidin-4-amine (200.00 mg, 1.110 mmol, 1.00 equiv) and NCS (177.84 mg, 1.332 mmol, 1.2 equiv) in ACN (10.00 mL) was stirred for 16 h at room temperature under nitrogen atmosphere. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to the title compound (220 mg, 92.35%) as a white solid. m/z (ESI+ve ion)=215.10 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.02 (s, 1H), 5.08 (s, 2H), 4.52 (d, J=5.7 Hz, 1H), 4.35-4.17 (m, 4H), 3.35 (s, 3H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[[5-chloro-6-(3-methoxyazetidin-1-yl) pyrimidin-4-yl] amino] indazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (100.00 mg, 0.158 mmol, 1.00 equiv) and 5-chloro-6-(3-methoxyazetidin-1-yl) pyrimidin-4-amine (40.79 mg, 0.190 mmol, 1.2 equiv) in toluene (2.50 mL) were added Pd2(dba)3(14.50 mg, 0.016 mmol, 0.1 equiv) and XantPhos (9.16 mg, 0.016 mmol, 0.1 equiv) and Cs2CO3(103.19 mg, 0.317 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (10 mL). The resulting mixture was extracted with DCM (3×15 mL). The combined organic layers were washed with brine (2×20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford the title compound (62 mg, 54.51%) as a yellow solid. m/z=718.25 [M+H].1H NMR (400 MHz, Chloroform-d) δ 8.10 (d, J=11.5 Hz, 1H), 7.87-7.70 (m, 2H), 7.62 (s, 1H), 7.55 (m, 1H), 7.08 (d, J=8.5 Hz, 1H), 6.70 (m, 1H), 5.62 (d, J=2.7 Hz, 1H), 4.63-4.58 (m, 1H), 4.37-4.26 (m, 3H), 3.52 (t, J=8.7 Hz, 1H), 3.38 (d, J=10.9 Hz, 4H), 3.35 (s, 3H), 2.38 (m, 1H), 2.13 (m, 1H), 1.70 (d, J=2.6 Hz, 18H) Step C. (1R,2S)-2-(3-[[5-chloro-6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino]-1H-indazol-6-yl)-5-methoxy-1H-spiro[cyclopropane-1,3-indol]-2-one A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[[5-chloro-6-(3-methoxyazetidin-1-yl) pyrimidin-4-yl] amino] indazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (62.00 mg, 0.086 mmol, 1.00 equiv) and TFA (2.00 mL, 0.018 mmol, 0.20 equiv) in DCM (4.00 mL) was stirred for 1 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: YMC-Actus Triart C18 ExRS, 30 mm×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min. Gradient: 25% B to 48% B in 8 min, 254 nm; RT1: 7.28 min to afford Example 47 (25.0 mg, 55.91%) as a white solid. m/z (ESI, +ve ion)=518.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.68 (s, 1H), 10.42 (s, 1H), 8.93 (s, 1H), 7.83 (s, 1H), 7.53-7.22 (m, 2H), 6.90 (d, J=8.8 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58 (m, 1H), 5.71 (d, J=2.7 Hz, 1H), 4.44 (m, 2H), 4.25 (s, 1H), 4.05 (m, 2H), 3.17 (m, 7H), 2.32 (d, J=7.5 Hz, 1H), 1.98 (m, 1H). Example 48. (1R,2S)-2-(3-{[2-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 49. (1R,2S)-2-(3-{[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-(2,6-dichloro-5-methoxypyrimidin-4-yl) morpholine A mixture of 2,4,6-trichloro-5-methoxypyrimidine (200.00 mg, 0.937 mmol, 1.00 equiv) and morpholine (97.96 mg, 1.124 mmol, 1.2 equiv) in THF (5 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography with the following conditions: Column: Spherical C18, 20-40 μm, 80 g; Mobile Phase A: Water (plus 5 mM NH4HCO3); Mobile Phase B: ACN; Flow rate: 40 mL/min; Gradient: 40% B-60% B in 20 min; Detector: 254 nm. The fractions containing desired product were collected at 54% B and concentrated under reduced pressure to afford the title compound (130 mg, 52.53%) as an off-white solid. m/z (ESI+ve ion)=263.95 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 3.94-3.87 (m, 4H), 3.83-3.77 (m, 4H), 3.75 (s, 3H). Step B. The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[[2-chloro-5-methoxy-6-(morpholin-4-yl) pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (16b) and tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((4-chloro-5-methoxy-6-morpholinopyrimidin-2-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a stirred mixture of 4-(2,6-dichloro-5-methoxypyrimidin-4-yl) morpholine (30.34 mg, 1.20 equiv) and tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl) indazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (50.00 mg, 1.00 equiv) in toluene (1.25 mL) were added Pd2(dba)3(8.79 mg, 0.10 equiv) and XantPhos (5.56 mg, 0.10 equiv) and Cs2CO3(62.65 mg, 2.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure. The residue was purified by prep-TLC (PE/EA 1/1) to afford a mixture of the title compounds (50 mg, 70%) as a yellow oil. m/z (ESI +ve ion)=748.35 [M+H]+. Step C. (1R,2)-2-(3-((2-chloro-5-methoxy-6-morpholinopyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one and (1R,2S)-2-(3-[[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of compounds from Step B (50.00 mg) in TFA (2.00 mL) and DCM (4.00 mL) was stirred for 1 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 50% B in 8 min, 220 nm; RT1:7.30 min; RT2: 7.75 min) to afford Example 48 (8.1 mg) as an off-white solid. m/z (ESI+ve ion)=548.20 [M+H].1H NMR (400 MHz, DMSO-d6) δ 12.67 (s, 1H), 10.41 (s, 1H), 9.21 (s, 1H), 7.61-7.26 (m, 2H), 6.93 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58 (m, 1H), 5.70 (d, J=1.6 Hz, 1H), 3.71 (t, J=4.5 Hz, 4H), 3.65 (s, 3H), 3.62 (t, J=4.6 Hz, 4H), 3.19 (t, J=8.5 Hz, 3H), 3.18 (m, 1H), 2.32 (m, 1H), 1.99 (m, 1H). Compound Example 49 (8.2 mg) was also obtained as an off-white solid from the collection of fractions from the HPLC. m/z (ESI+ve ion)=548.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.46 (s, 1H), 10.41 (s, 1H), 9.43 (s, 1H), 7.49 (d, J=8.4 Hz, 1H), 7.38 (s, 1H), 6.83 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (m, 1H), 5.64 (d, J=2.0 Hz, 1H), 3.59 (s, 3H), 3.52 (m, 6H), 3.37 (s, 2H), 3.31 (m, 3H) 3.17 (t, J=7.2 Hz, 1H), 2.31 (m, 1H), 1.97 (m, 1H). Example 50. (1R,2S)-2-(3-{[1-(2-hydroxyethyl)-3-methoxy-1H-pyrazol-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-bromo-3-methoxy-1-[2-(oxan-2-yloxy)ethyl]pyrazole To the mixture of 4-bromo-3-methoxy-1H-pyrazole (500.00 mg, 2.825 mmol, 1.00 equiv) in DMF (5.00 mL) was added Cs2CO3(1104.47 mg, 3.390 mmol, 1.2 equiv) at 25° C. After stirred for 30 min, 2-(2-bromoethoxy)oxane (708.75 mg, 3.390 mmol, 1.2 equiv) was added. The mixture was stirred for 3 h. The reaction was diluted with water (50 mL) and extracted with EA (20 mL×3). The combined organic layer was washed with brine (50 mL×2), dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-50% EA in PE to give the title compound (750 mg, 82.65%) as a colorless oil. m/z (ESI+ve ion)=305.00 [M+H]+.1H NMR (300 MHz, Chloroform-d) S 7.35 (s, 1H), 4.55 (t, J=3.3 Hz, 1H), 4.16-4.10 (m, 2H), 4.05-3.97 (m, 1H), 3.96 (s, 3H), 3.75-3.65 (m, 2H), 3.52-3.45 (m, 1H), 1.85-1.52 (m, 6H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-([3-methoxy-1-[2-(oxan-2-yloxy)ethyl]pyrazol-4-yl]amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1-carboxylate (110.00 mg, 0.211 mmol, 1.00 equiv) and 4-bromo-3-methoxy-1-[2-(oxan-2-yloxy)ethyl]pyrazole (77.38 mg, 0.253 mmol, 1.20 equiv) in a dry dioxane (2.00 mL) were added Cs2CO3(137.69 mg, 0.422 mmol, 2.00 equiv), EPhos (22.60 mg, 0.042 mmol, 0.20 equiv) and EPhos Pd G4(38.82 mg, 0.042 mmol, 0.20 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by Prep-TLC (rinsed with PE/EA=2/1) to give the title compound (15 mg, 9.05%) as yellow oil. m/z (ESI+ve ion)=745.55 [M+H]+. Step C. (1R,2S)-2-(3-[[1-(2-hydroxyethyl)-3-methoxypyrazol-4-yl]amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a mixture solution of TFA (0.40 mL), H2O (0.20 mL) and THE (0.40 mL) was added tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-([3-methoxy-1-[2-(oxan-2-yloxy)ethyl]pyrazol-4-yl]amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (14.00 mg, 0.019 mmol, 1.00 equiv). The mixture was stirred at 50° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified with the following conditions: Column: XBridge Prep OBD C18 Column, 19×250 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 15% B to 35% B in 10 min; Detector: 254 & 220 nm; RT1: 8.62 min. The product-containing fractions was combined and concentrated to give Example 50 (2.7 mg) as a white solid. m/z (ESI+ve ion)=461.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.73 (s, 1H), 7.66 (d, J=8.0 Hz, 1H), 7.24 (s, 1H), 6.82 (t, J=8.4 Hz, 2H), 6.64-6.61 (m, 1H), 5.61 (d, J=2.4 Hz, 1H), 4.07-4.04 (m, 2H), 3.93 (s, 3H), 3.88-3.86 (m, 2H), 3.35 (s, 1H), 3.30 (s, 3H), 2.23-2.14 (m, 2H). Example 51. (1R,2S)-2-(3-{[2-cyclopropyl-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-cyclopropyl-6-hydroxy-5-methoxy-3H-pyrimidin-4-one Sodium 2-methylpropan-2-olate (1.48 g, 15.419 mmol, 2.50 equiv) was added to MeOH (10.00 mL) in portions over 20 min at 0° C. To this solution were added 1,3-dimethyl 2-methoxypropanedioate (0.10 g, 0.617 mmol, 1.00 equiv) 1,3-dimethyl 2-methoxypropanedioate (1.00 g, 6.167 mmol, 1.00 equiv) and cyclopropanecarboximidamide (0.52 g, 6.167 mmol, 1.00 equiv). The mixture was heated to reflux for 12 h and then cooled to 0° C. The PH was adjusted to ˜4 with Conc. HCl and filtered. The solid was collected and lyophilized to give crude the title compound (500 mg, crude) as a white solid. m/z (ESI+ve ion)=183.10 [M+H]+. Step B. 4,6-dichloro-2-cyclopropyl-5-methoxypyrimidine A mixture of crude 2-cyclopropyl-6-hydroxy-5-methoxy-3H-pyrimidin-4-one (500.00 mg) in POCl3(5.00 mL) was stirred for 3 h at 100° C. After cooled to room temperature, the mixture solution was added dropwise to cooled sat. aq. NaHCO3(150 mL). The mixture was extracted with EA (3×20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-20% EA in PE to give the title compound (370 mg) as a colorless oil. m/z (ESI+ve ion)=219.00 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 3.93 (s, 3H), 2.23-2.17 (m, 1H), 1.18-1.09 (m, 4H). Step C. 4-(6-chloro-2-cyclopropyl-5-methoxypyrimidin-4-yl)morpholine To the mixture of 4,6-dichloro-2-cyclopropyl-5-methoxypyrimidine (370.00 mg, 1.689 mmol, 1.00 equiv) and TEA (205.09 mg, 2.027 mmol, 1.20 equiv) in EtOH (5.00 mL) was added morpholine (176.57 mg, 2.027 mmol, 1.20 equiv). The mixture was stirred at 25° C. for 12 h. The solvent was moved and the residue was purified by silica gel column, eluted with 0-50% EA in PE to give the title compound (280 mg, 58.39%) as colorless oil. m/z (ESI+ve ion)=270.15 [M+H]+.1H NMR (400 MHz, Chloroform-d) 3.82-3.73 (m, 8H), 3.72 (s, 3H), 2.08-2.01 (m, 1H), 1.04-0.94 (m, 4H). Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-cyclopropyl-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.154 mmol, 1.00 equiv) and 4-(6-chloro-2-cyclopropyl-5-methoxypyrimidin-4-yl)morpholine (49.74 mg, 0.185 mmol, 1.2 equiv) in a dry dioxane (2 mL) were added Cs2CO3(100.14 mg, 0.308 mmol, 2 equiv), CPhos (13.42 mg, 0.031 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(31.81 mg, 0.031 mmol, 0.2 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-100% of EA in PE to give crude the title compound (60 mg, 51.79%) as a white solid. m/z (ESI +ve ion)=754.55 [M+H]+. Step E. (1R,2S)-2-(3-[[1-(2-hydroxyethyl)-3-methoxypyrazol-4-yl]amino]-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-cyclopropyl-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60.00 mg, 0.060 mmol, 1.00 equiv) in DCM (3.00 mL) and TFA (0.30 mL) was stirred for 4 h. The solvent was removed under reduced pressure and the residue was purified with the following conditions: Column: XBridge Prep OBD C18 Column, 19×250 mm 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 55% B in 10 min; Detector: 254&220 nm; RT1:8.62 min. The product-containing fractions was combined and concentrated to give Example 51 (27.1 mg, 77.28%) as a white solid. m/z (ESI+ve ion)=554.40 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.59 (d, J=8.4 Hz, 1H), 7.41 (s, 1H), 6.92-6.89 (m, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.63-6.61 (m, 1H), 5.65 (d, J=2.8 Hz, 1H), 3.84-3.79 (m, 4H), 3.74 (s, 3H), 3.67-3.64 (m, 4H), 3.38-3.36 (m, 1H), 3.32 (s, 3H), 2.25-2.23 (m, 1H), 2.20-2.17 (m, 1H), 1.78-1.73 (m, 1H), 0.79-0.77 (m, 2H), 0.74-0.68 (m, 2H). Example 52. (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. (2S,6R)-4-(6-Chloro-5-methoxypyrimidin-4-yl)-2,6-dimethylmorpholine To an oven-dried flask was added 4,6-dichloro-5-methoxypyrimidine (1.00 g, 5.59 mmol) followed by DMSO (19 mL), K2CO3(1.16 g, 8.34 mmol), and cis-2,6-dimethylmorpholine (0.64 mL, 5.9 mmol). The reaction mixture was stirred at room temperature for 1 h at which time it was quenched with sat. aqueous NH4Cl. The mixture was extracted three times with EtOAc. The combined organic layers were washed with water and brine, dried with MgSO4, filtered, and concentrated. The residue was purified by column chromatography (5% to 30% EtOAc/hexanes, gradient elution) to afford the title compound (1.30 g, 90%) as a white solid. m/z (ESI, +ve ion)=258.2 [M+H]+. Step B. Tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((6-((2S,6R)-2,6-dimethylmorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To an oven-dried flask was added (2S,6R)-4-(6-Chloro-5-methoxypyrimidin-4-yl)-2,6-dimethylmorpholine (36.4 mg, 0.141 mmol), tert-butyl (R,2S)-2-(3-amino-1-(tert-butoxycarbonyl)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (70.0 mg, 0.135 mmol), rac-binap Pd G4 (13.5 mg, 0.0134 mmol), BINAP (8.40 mg, 0.0134 mmol), Tripotassium phosphate (57.0 mg, 0.270 mmol) and 1,4-dioxane (3.4 mL). The mixture was degassed with bubbling argon for 10 min. At this time, the reaction mixture was heated to 70° C. for 2 h. The reaction mixture was cooled was cooled to room temperature and diluted with EtOAc and washed with sat. aqueous NaHCO3. The aqueous layer was extracted an additional three times with EtOAc. Combined organic layers were washed with brine, dried with MgSO4, filtered, and concentrated. The residue was purified by column chromatography (0% to 80% EtOAc/hexanes, a gradient elution) to provide the title compound (30 mg, 30%) as a white foam. m/z (ESI, +ve ion)=742.3 [M+H]+. Step C To an oven-dried flask was added tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((6-((2S,6R)-2,6-dimethylmorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (30 mg, 0.040 mmol) followed DCM (2.0 mL) and trifluoroacetic acid (0.15 mL, 2.0 mmol). The reaction mixture was stirred at room temperature for 2 h. At this time, the mixture was concentrated and purified by prep HPLC (20% to 40% ACN/H2O, 0.1% TFA modifier, gradient elution) to afford Example 52 (8.0 mg, 37%) as a white amorphous solid after lyophilization. m/z (ESI, +ve ion)=542.3 [M+H]+.1H NMR (400 MHz, DMSO-d) δ=12.99-12.60 (m, 1H), 10.45 (s, 1H), 9.81 (br s, 1H), 8.03 (s, 1H), 7.62 (d, J=8.3 Hz, 1H), 7.44 (s, 1H), 6.96 (d, J=8.3 Hz, 1H), 6.75 (d, J=8.6 Hz, 1H), 6.59 (dd, J=2.5, 8.6 Hz, 1H), 5.76-5.69 (m, 1H), 4.40 (br d, J=12.9 Hz, 2H), 3.69 (s, 3H), 3.68-3.59 (m, 2H), 3.34 (s, 3H), 3.19 (t, J=8.5 Hz, 1H), 2.72 (dd, J=10.9, 12.9 Hz, 2H), 2.34 (dd, J=4.7, 8.0 Hz, 1H), 1.99 (dd, J=4.7.9.0 Hz, 1H), 1.14 (d, J=6.3 Hz, 6H). Example 53. (1R,2S)-2-(3-((5-chloro-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 4-(6-Amino-5-chloropyrimidin-4-yl)thiomorpholine 1,1-dioxide To an oven-dried flask was added 5,6-dichloro-4-pyrimidinamine (1.0) g, 6.10 mmol) followed by toluene (9.0 mL) and thiomorpholine dioxide (0.850 mL, 6.31 mmol). The reaction mixture was heated to 80° C. for 16 hours and then cooled to room temperature. The reaction was then concentrated to a white solid to give the title compound (700 mg, 44%) as a white amorphous solid. Step B. Tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-chloro-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To an oven-dried flask was added tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (102 mg, 0.162 mmol), 4-(6-amino-5-chloropyrimidin-4-yl)thiomorpholine 1,1-dioxide (45 mg, 0.167 mmol), Xantphos Pd G4 (15.6 mg, 0.0162 mmol), and 1,4-dioxane (1.6 mL). The mixture was degassed with bubbling argon for 10 min. At this time, Cs2CO3(105 mg, 0.323 mmol) was added and the reaction mixture was heated to 100° C. for 1 h. The reaction mixture was cooled was cooled to room temperature and diluted with EtOAc and washed with sat. aqueous NaHCO3. The aqueous layer was extracted an additional three times with EtOAc. Combined organic layers were washed with brine, dried with MgSO4, filtered, and concentrated. The residue was purified by column chromatography (0% to 50% acetone/hexanes, a gradient elution) to provide the title compound (30 mg, 24%) as a white foam. m/z (ESI, +ve ion)=767.2 [M+H]+. Step C To an oven-dried flask was added tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-chloro-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (30 mg, 0.039 mmol) followed DCM (2.0 mL) and trifluoroacetic acid (0.15 mL, 2.0 mmol). The reaction mixture was stirred at room temperature for 2 h. At this time, the mixture was concentrated and purified by prep HPLC (20% to 40% ACN/H2O, 0.1% TFA modifier, gradient elution) to afford Example 53 (5.0 mg, 23%) as a white amorphous solid after lyophilization. m/z (ESI, +ve ion)=566.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ=12.75 (br s, 1H), 10.43 (s, 1H), 9.29 (s, 1H), 8.01 (s, 1H), 7.42 (s, 1H), 7.35 (d, J=8.3 Hz, 1H), 6.90 (dd, J=1.1, 8.5 Hz, 1H), 6.74 (d, J=8.3 Hz, 1H), 6.60-6.55 (m, 1H), 5.71 (d, J=2.8 Hz, 1H), 3.94 (br s, 4H), 3.33 (s, 3H), 3.32-3.26 (m, 4H), 3.21-3.15 (m, 1H), 2.33 (dd, J=4.7, 8.0 Hz, 1H), 2.01-1.95 (m, 1H). Example 54. (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 4-(6-Chloro-5-methoxypyrimidin-4-yl)thiomorpholine 1,1-dioxide To an oven-dried flask was added 4,6-dichloro-5-methoxypyrimidine (1.00 g, 5.59 mmol) followed by DMSO (19 mL), K2CO3(1.16 g, 8.34 mmol), and thiomorpholine dioxide (0.75 mL, 5.6 mmol). The reaction mixture was stirred at room temperature for overnight at which time it was quenched with sat. aqueous NH4Cl. The mixture was extracted three times with EtOAc. The combined organic layers were washed with water and brine, dried with MgSO4, filtered, and concentrated. The residue was purified by column chromatography (5% to 30% EtOAc/hexanes, gradient elution) to afford the title compound (973 mg, 63%) as a white solid. m/z (ESI, +ve ion)=278.0 [M+H]+. Step B. Tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((6-(1,1-dioxidothiomorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To an oven-dried flask was added 4-(6-Chloro-5-methoxypyrimidin-4-yl)thiomorpholine 1,1-dioxide (74.7 mg, 0.2689 mmol), tert-butyl (1R,2S)-2-(3-amino-1-(tert-butoxycarbonyl)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (70.0 mg, 0.135 mmol), Xantphos Pd G4 (13.0 mg, 0.134 mmol), and 1,4-dioxane (1.3 mL). The mixture was degassed with bubbling argon for 10 min. At this time, Cs2CO3(87.6 mg, 0.269 mmol) was added and the reaction mixture was heated to 100° C. for 2 h. The reaction mixture was cooled was cooled to room temperature and diluted with EtOAc and washed with sat. aqueous NaHCO3. The aqueous layer was extracted an additional three times with EtOAc. Combined organic layers were washed with brine, dried with MgSO4, filtered, and concentrated. The residue was purified by column chromatography (0% to 80% EtOAc/hexanes, a gradient elution) to provide the title compound (19 mg, 19%) as a white foam. m/z (ESI, +ve ion)=762.2 [M+H]+. Step C To an oven-dried flask was added tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((6-(1,1-dioxidothiomorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (19 mg, 0.025 mmol) followed DCM (1.25 mL) and trifluoroacetic acid (0.10 mL, 1.3 mmol). The reaction mixture was stirred at room temperature for 3 h. At this time, the mixture was concentrated and purified by prep HPLC (20% to 40% ACN/H2O, 0.1% TFA modifier, gradient elution) to afford Example 54 (10 mg, 71%) as a white amorphous solid after lyophilization. m/z (ESI, +ve ion)=562.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ=12.78 (br s, 1H), 10.45 (s, 1H), 9.57 (br s, 1H), 7.99 (s, 1H), 7.54 (d, J=8.3 Hz, 1H), 7.43 (s, 1H), 6.93 (d, J=8.3 Hz, 1H), 6.75 (d, J=8.3 Hz, 1H), 6.59 (dd, J=2.7, 8.5 Hz, 1H), 5.73 (d, J=2.5 Hz, 1H), 4.15 (br s, 4H), 3.71 (s, 3H), 3.34 (s, 3H), 3.30-3.24 (m, 4H), 3.22-3.16 (m, 1H), 2.34 (dd, J=4.7, 8.0 Hz, 1H), 1.99 (dd, J=4.7, 9.0 Hz, 1H). Example 55. (1R,2S)-2-(3-{[5-(2-hydroxyethyl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl N-(5-bromo-3-methoxypyrazin-2-yl)-N-(tert-butoxycarbonyl)carbamate To a stirred mixture of 5-bromo-3-methoxypyrazin-2-amine (2000.00 mg, 9.803 mmol, 1.00 equiv) and Boc2O (3209.09 mg, 14.705 mmol, 1.50 equiv) in THF (5.00 mL) were added DMAP (119.76 mg, 0.980 mmol, 0.10 equiv) and NaOH (784.15 mg, 19.606 mmol, 2.00 equiv). The resulting mixture was stirred for 2 hours at RT under N2atmosphere. The resulting mixture was extracted with EA (3×60 mL). The combined organic layers were washed with sat. brine (3×25 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE:EA (3:1) to afford the title compound (1.7 g, 42.5%) as white solid. m/z (ESI+ve ion-100)=403.85 [M+H−100]+. Step B. tert-butyl N-(tert-butoxycarbonyl)-N-{5-[(E)-2-ethoxyethenyl]-3-methoxypyrazin-2-yl}carbamate To a stirred mixture of tert-butyl N-(5-bromo-3-methoxypyrazin-2-yl)-N-(tert-butoxycarbonyl)carbamate (990 mg, 2.449 mmol, 1.00 equiv) and tributyl[(E)-2-ethoxyethenyl]stannane (1768.85 mg, 4.898 mmol, 2 equiv) in DMF (15 mL, 193.826 mmol, 79.15 equiv) were added LiCl (363.37 mg, 8.572 mmol, 3.5 equiv) and Pd(PPh3)2Cl2(171.89 mg, 0.245 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 1.5 h at 80° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (15 mL) at room temperature. The resulting mixture was extracted with DCM (3×15 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (730 mg, 75.38%) as a white solid. m/z (ESI+ve ion)=396.10 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.66 (s, 1H), 6.53 (d, J=7.1 Hz, 1H), 5.40 (d, J=7.1 Hz, 1H), 4.11 (m, 2H), 3.97 (s, 3H), 1.42 (s, 21H). Step C. tert-butyl N-(tert-butoxycarbonyl)-N-[5-(2-hydroxyethyl)-3-methoxypyrazin-2-yl] carbamate To a stirred mixture of tert-butyl N-(tert-butoxycarbonyl)-N-{5-[(E)-2-ethoxyethenyl]-3-methoxypyrazin-2-yl} carbamate (720.00 mg, 1.821 mmol, 1.00 equiv) in THF (5.50 mL) were added mercuric acetate (696.26 mg, 2.18 mmol, 1.20 equiv) in water (6.55 mL) dropwise at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 15 min at 0° C. under nitrogen atmosphere. To the above mixture was added NaBH4(275.53 mg, 7.284 mmol, 4.00 equiv) in saturated K2CO3(aq.) (5.44 mL) dropwise at room temperature. The resulting mixture was stirred for additional 20 min at room temperature. The reaction was quenched by the addition of Water (10 mL). The resulting mixture was extracted with DCM (3×15 mL). The combined organic layers were washed with brine (2×20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (1/1) to afford the title compound (600 mg, 89.21%) as a colorless solid. m/z (ESI+ve ion)=370.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.93 (s, 1H), 4.07-3.98 (m, 6H), 3.01 (t, J=5.8 Hz, 2H), 1.44 (s, 18H). Step D. tert-butyl N-(tert-butoxycarbonyl)-N-(6-[2-[(tert-butyldimethylsilyl) oxy]ethyl]-3-methoxypyrazin-2-yl)carbamate To a stirred mixture of tert-butyl N-(tert-butoxycarbonyl)-N-[5-(2-hydroxyethyl)-3-methoxypyrazin-2-yl] carbamate (166.00 mg, 0.449 mmol, 1.00 equiv) in DMF (4.00 mL) were added 1H-imidazole (73.42 mg, 0.000 mmol, 2.40 equiv) and TBDMS-Cl (81.27 mg, 0.539 mmol, 1.20 equiv) at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was quenched by the addition of water (15 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (6/1) to afford the title compound (105 mg, 48.31%) as a yellow oil. m/z (ESI+ve ion)=484.30 [M+H]+. Step E. 6-[2-[(tert-butyl dimethylsilyl)oxy]ethyl]-3-methoxypyrazin-2-amine Into a 50 mL round-bottom flask were added tert-butyl N-tert-butoxycarbonyl)-N-(6-[2-[(tert-butyldimethylsilyl) oxy]ethyl]-3-methoxypyrazin-2-yl)carbamate (560.00 mg, 1 equiv) and 1,1,1,3,3,3-hexafluoropropan-2-ol (20.00 mL) at room temperature. The resulting mixture was stirred for 16 h at 60° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (2/1) to afford the title compound (237 mg, 72.22%) as a white solid. m/z (ESI+ve ion)=284.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.33 (s, 1H), 5.98 (s, 2H), 3.85 (d, J=15.4 Hz, 5H), 2.63 (m, 2H), 0.81 (s, 9H), 0.06 (s, 6H). Step F. tert-butyl (1R,2S)-2-(1-tert-butoxycarbonyl)-3-((5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methoxypyrazin-2-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (100.00 mg, 0.158 mmol, 1.00 equiv) and 6-[2-[(tert-butyl dimethylsilyl)oxy]ethyl]-3-methoxypyrazin-2-amine (53.86 mg, 0.190 mmol, 1.20 equiv) in toluene (2.50 mL) were added Pd2(dba)3(14.50 mg, 0.016 mmol, 0.1 equiv) and XantPhos (9.16 mg, 0.016 mmol, 0.1 equiv) and Cs2CO3(103.19 mg, 0.317 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere and cooled down to room temperature. The reaction was quenched by the addition of water (5 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (3/1) to afford the title compound (84 mg, 67.40%) as a yellow solid. m/z (ESI+ve ion)=787.60 [M+H]+. Step G: (1R,2S)-2-(3-((5-(2-hydroxyethyl)-3-methoxypyrazin-2-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one A mixture of tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methoxypyrazin-2-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (84.00 mg, 0.107 mmol, 1.00 equiv) and TFA (2.0) mL, 26.926 mmol, 252.27 equiv) in DCM (4.00 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The resulting mixture was diluted with MeOH (5.00 mL). To the above mixture was added K2CO3(50.00 mg, 0.362 mmol, 3.39 equiv) in portions at room temperature. The resulting mixture was stirred for additional 2 h at room temperature then concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: YMC-Actus Triart C18 ExRS, 30 mm×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 45% B in 8 min, 254 nm; RT1: 6.58 min to afford Example 55 (21.5 mg, 42.63%) as a white solid. m/z (ESI+ve ion)=473.30 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.51 (s, 1H), 10.43 (s, 1H), 8.63 (s, 1H), 7.39 (m, 3H), 6.87 (d, J=9.2 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (m, 1H), 5.72 (d, J=2.4 Hz, 1H), 4.60 (m, 1H), 3.97 (s, 3H), 3.68 (q, J=6.5 Hz, 2H), 3.33 (s, 3H), 3.35 (d, J=15.2 Hz, 2H), 2.51 (d, J=1.6 Hz, 2H), 1.98 (m, 1H). Example 56. (1R,2S)-2-(3-{[6-(2-hydroxyethyl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl N-(tert-butoxycarbonyl)-N-(6-chloro-3-methoxypyrazin-2-yl)carbamate To a stirred mixture of 6-chloro-3-methoxypyrazin-2-amine (1500.00 mg, 9.400 mmol, 1.00 equiv) and Boc2O (3077.37 mg, 14.100 mmol, 1.5 equiv) in THF (10 mL) were added DMAP (114.84 mg, 0.940 mmol, 0.1 equiv) and Et3N (1902.43 mg, 18.801 mmol, 2 equiv). The resulting mixture was stirred for 2 hours at room temperature under N2atmosphere. The resulting mixture was extracted with EA (3×20 mL). The combined organic layers were washed with brine (3×15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE:EA (4:1) to afford the title compound (2.8 g, 82%) as a white solid. m/z (ESI, +ve ion)=204.1 [M+H−100−56]+. Step B. tert-butyl N-(tert-butoxycarbonyl)-N-[6-[(E)-2-ethoxyethenyl]-3-methoxypyrazin-2-yl]carbamate To a stirred mixture of tert-butyl N-(tert-butoxycarbonyl)-N-(6-chloro-3-methoxypyrazin-2-yl)carbamate (410.00 mg, 1.139 mmol, 1.00 equiv) and Pd(PPh3)2Cl2(79.98 mg, 0.114 mmol, 0.10 equiv) in DMF (10.00 mL) were added tributyl[(E)-2-ethoxyethenyl]stannane (823.05 mg, 2.278 mmol, 2.00 equiv) and LiCl (169.08 mg, 3.987 mmol, 3.50 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 1.5 h at 80° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of water (15 mL) at room temperature. The resulting mixture was extracted with DCM (3×15 mL). The combined organic layers were washed with brine (2×25 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography with the following conditions: Column: Spherical C18, 20-40 μm, 120 g; Mobile Phase A: Water (plus 5 mM NH4HCO3); Mobile Phase B: ACN; Flow rate: 50 mL/min; Gradient: 50% B-60% B in 15 min; Detector: 254 nm. The fractions containing desired product were collected at 61% B and concentrated under reduced pressure to afford the title compound (400 mg, 88.77%) as a yellow oil. m/z=396.30 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.76 (s, 1H), 6.41 (d, J=7.0 Hz, 1H), 5.38 (d, J=7.0 Hz, 1H), 4.12-3.92 (m, 5H), 1.39 (d, J=3.2 Hz, 21H). Step C. tert-butyl N-(tert-butoxycarbonyl)-N-[6-(2-hydroxyethyl)-3-methoxypyrazin-2-yl] carbamate To a stirred mixture of tert-butyl N-(tert-butoxycarbonyl)-N-[6-[(E)-2-ethoxyethenyl]-3-methoxypyrazin-2-yl] carbamate (333.00 mg, 0.842 mmol, 1.00 equiv) in THF (2.53 mg) was added mercuric acetate (322.02 mg, 1.010 mmol, 1.20 equiv) in water (3.03 m L) dropwise at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 15 min at 0° C. under nitrogen atmosphere. To the above mixture was added NaBH4(127.43 mg, 3.368 mmol, 4.00 equiv) in saturated K2CO3, aq. (3.00 mL) dropwise at room temperature. The resulting mixture was stirred for additional 1 h at room temperature. The reaction was quenched by the addition of water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography with the following conditions: Column: Spherical C18, 20-40 μm, 80 g; Mobile Phase A: Water (plus 5 mM NH4HCO); Mobile Phase B: ACN; Flow rate: 80 mL/min; Gradient: 40% B-60% B in 20 min; Detector: 254 nm. The fractions containing desired product were collected at 55% B and concentrated under reduced pressure to afford the title compound (217 mg, 69.76%) as an off-white solid. m/z=370.30 [M+H]+.1H NMR (400 MHz, DMSO-d) δ 8.11 (s, 1H), 4.68 (t, J=5.2 Hz, 1H), 3.93 (s, 3H), 3.68 (m, 2H), 2.83 (t, J=6.7 Hz, 2H), 1.35 (s, 18H). Step D. tert-butyl N-(tert-butoxycarbonyl)-N-(6-[2-[(tert-butyldimethylsilyl) oxy]ethyl]-3-methoxypyrazin-2-yl)carbamate To a stirred mixture of tert-butyl N-(tert-butoxycarbonyl)-N-[6-[(E)-2-ethoxyethenyl]-3-methoxypyrazin-2-yl] carbamate (182.00 mg, 0.493 mmol, 1.00 equiv) in DMF (4.55 mL) was added 1H-imidazole (80.49 mg, 0.000 mmol, 2.40 equiv) at room temperature under nitrogen atmosphere. To the above mixture was added TBS-Cl (89.11 mg, 0.592 mmol, 1.20 equiv) at 0° C. The resulting mixture was stirred for additional 2 h at room temperature. The reaction was quenched with water at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (4:1) to afford the title compound (238 mg, 99.88%) as a dark yellow oil. m/z (ESI+ve ion)=484.25 [M+H]+. Step E. 6-[2-[(tert-butyldimethylsilyl) oxy]ethyl]-3-methoxypyrazin-2-amine Into a 50 mL round-bottom flask were added tert-butyl N-(tert-butoxycarbonyl)-N-[6-[(E)-2-ethoxyethenyl]-3-methoxypyrazin-2-yl] carbamate (218.00 mg, 0.450 mmol) and 1,1,1,3,3,3-hexafluoropropan-2-ol (5.50 mL) at room temperature. The resulting mixture was stirred for 16 h at 60° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The reaction was quenched with water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (3/1) to afford the title compound (122 mg, 94%) as an off-white solid. m/z (ESI, +ve ion)=284.10 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.50-7.11 (m, 3H), 4.05-3.75 (m, 5H), 2.76 (t, J=6.6 Hz, 2H), 1.01-0.75 (m, 9H), 0.01 (d, J=0.7 Hz, 6H). Step F. tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-[2-[(tert-butyldimethylsilyl) oxy]ethyl]-3-methoxypyrazin-2-yl)amino]indazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate To a stirred mixture of 6-[2-[(tert-butyldimethylsilyl) oxy]ethyl]-3-methoxypyrazin-2-amine (53.80 mg, 1.20 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (100.00 mg, 1.00 equiv) in toluene (2.5 mL) were added Pd2(dba)3(14.5 mg, 0.1 equiv) and XantPhos (9.15 mg, 0.1 equiv) and Cs2CO3(103.25 mg, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched with Water at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (3/I) to afford the title compound (84.1 mg, 67.48%) as a dark yellow solid. m/z (ESI, +ve ion)=787.30 [M+H]+. Step G. (1R,2S)-2-(3-[[6-(2-hydroxyethyl)-3-methoxypyrazin-2-yl]amino]-1H-indazol-6-yl)-5-methoxy-1H-spiro[cyclopropane-1,3-indol]-2-one A mixture of tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-[2-[(tert-butyldimethylsilyl) oxy]ethyl]-3-methoxypyrazin-2-yl)amino]indazol-6-yl]-5-methoxy-2-oxospiro[cyclopropane-1,3-indole]-1-carboxylate (84.00 mg, 0.107 mmol, 1.00 equiv) and TFA (2.00 mL, 0.018 mmol, 0.16 equiv) in DCM (4.00 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. To the above mixture was added K2CO3(50.00 mg, 0.362 mmol, 3.39 equiv) in MeOH (5.00 mL) at room temperature. The resulting mixture was stirred for additional 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: YMC-Actus Triart C18 ExRS, 30 mm×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 43% B in 8 min, 254 nm; RT1: 7.22 min to afford Example 56 (24.5 mg, 48.09%) as a white solid. m/z (ESI, +ve ion)=473.20 [M+H]+,1H NMR (400 MHz, DMSO-d6) δ 12.50 (s, 1H), 10.40 (s, 1H), 8.75 (s, 1H), 7.50 (d, J=8.0 Hz, 1H), 7.35 (d, J=6.0 Hz, 2H), 6.86 (d, J=8.0 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58 (m, 1H), 5.67 (d, J=2.0 Hz, 1H), 4.48 (m, 1H), 3.96 (s, 3H), 3.52 (d, J=6.2 Hz, 2H), 3.49 (m, 4H), 3.19 (m, 2H), 2.29 (m, 1H), 1.98 (m, 1H). Example 57. 4-[5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-2-methylpyrimidin-4-yl]-1λ6-thiomorpholine-1,1-dione Step A. 4-(6-chloro-5-methoxy-2-methylpyrimidin-4-yl)-1lambda6-thiomor-pholine-1,1-dione To a stirred solution of 4,6-dichloro-5-methoxy-2-methylpyrimidine (400.0 mg, 1.0 equiv) and 1lambda6-thiomorpholine-1,1-dione (308.0 mg, 1.1 equiv) in THF (12.0 mL) was added TEA (420.0 mg, 2.0 equiv) at room temperature. The resulting mixture was stirred for 20 h at 70° C. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with 0-50% EtOAc in PE to afford the title compound (400.0 mg, 65.7% yield) as a white solid. m/z (ESI, +ve ion)=292.00 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 4.35-4.33 (m, 4H), 3.78 (s, 3H), 3.15-3.12 (m, 4H), 2.53 (s, 3H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxy-2-methylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100.0 mg, 1.0 equiv) and 4-(6-chloro-5-methoxy-2-methylpyrimidin-4-yl)-1lambda6-thiomor-pholine-1,1-dione (72.9 mg, 1.3 equiv) in toluene (2.5 mL) were added Cs2CO3(125.1 mg, 2.0 equiv), XantPhos (19.9 mg, 0.2 equiv) and Pd2(dba)3(35.1 mg, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×7 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% EtOAc in PE to afford the title compound (80.0 mg, 53.7%) as a yellow solid. m/z (ESI, +ve ion)=776.40 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.13 (s, 1H), 8.04 (s, 1H), 7.82 (d, J=8.0 Hz, 1H), 7.07 (d, J=8.0 Hz, 1H), 6.72-6.69 (m, 1H), 5.62 (d, J=0.0 Hz, 1H), 4.35-4.31 (m, 1H), 4.25 (s, 4H), 3.75 (s, 3H), 3.56-3.52 (m, 1H), 3.39 (s, 3H), 3.15 (s, 4H), 2.41-2.37 (m, 1H), 2.34 (s, 3H), 2.15-2.12 (m, 1H), 1.73-1.69 (m, 181-1). Step C. 4-[5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyc-lopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-2-methylpyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxy-2-methylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.0 mg) in DCM (4.0 mL) was added TFA (0.7 mL) dropwise at room temperature. The resulting mixture was stirred for 2 h at mom temperature. The resulting mixture was concentrated under vacuum. The crude product (50 mg) was purified by RP flash, eluted with ACN in water (5 mM NH4HCO3), 10% to 50% gradient in 30 min to afford Example 39 (22.0 mg, 37.0%) as a white solid. m/z (ESI, +ve ion)=576.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.59 (d, J=8.0 Hz, 1H), 7.43 (s, 1H), 6.92 (d, J=8.0 Hz, 1H), 6.84 (d, J=8.0 Hz, 1H), 6.64-6.61 (m, 1H), 5.66 (d, J=4.0 Hz, 1H), 4.23 (s, 4H), 3.76 (s, 3H), 3.37 (d, J=8.0 Hz, 1H), 3.35-3.30 (m, 3H), 3.21-3.15 (m, 4H), 3.01 (s, 1H), 2.88 (s, 1H), 2.26-2.20 (m, 4H), 2.19-2.17 (m, 1H). Example 58. 4-[5-chloro-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-2-methylpyrimidin-4-yl]-1λ6-thiomorpholine-1,1-dione Step A. 4-(6-amino-2-methylpyrimidin-4-yl)-lambda6-thiomorpholine-1,1-dione The mixture of 6-chloro-2-methylpyrimidin-4-amine (1 g, 6.965 mmol, 1.00 equiv) and 1lambda6-thiomorpholine-1,1-dione (2.82 g, 20.895 mmol, 3 equiv) was stirred for 80° C. for 12 h. The reaction mixture was turned out to be white solid. The solid was triturated with EA (20 mL) for 2 h and filtered. The filter cake was collected and dried in vacuo to give the title compound (500 mg, 29.33%) as a white solid. m/z (ESI+ve ion)=243.05 [M+H]+.1H NMR (400 MHz, DMSO-4) δ 6.74 (s, 2H), 5.70 (s, 1H), 4.00-3.98 (m, 4H), 3.16-3.13 (m, 4H), 2.27 (s, 3H). Step B. 4-(6-amino-5-chloro-2-methylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione To a stirred mixture of 4-(6-amino-2-methylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (500 mg, 2.064 mmol, 1.00 equiv) in THF (5 mL) was added NCS (220.44 mg, 1.651 mmol, 0.8 equiv) under nitrogen atmosphere. The mixture was stirred at 25° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to give the title compound (250 mg, 43.78%) as a colorless oil. m/z (ESI+ve ion)=277.15 [M+H].1H NMR (400 MHz, DMSO-d6) δ 6.89 (s, 2H), 3.85-3.83 (m, 4H), 3.24-3.21 (m, 4H), 2.25 (s, 3H). Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-chloro-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-2-methylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 4-(6-amino-5-chloro-2-methylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (52.59 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The reaction mixture was filtered and washed with EA (5 mL×3). The filtrate was concentrated in vacuo and the residue was purified by silica gel column eluted with 0-50% EA in PE to give the title compound (90 mg, 72.83%) as a yellow solid. m/z (ESI+ve ion)=780.35 [M+H].1H NMR (400 MHz, Chloroform-d) δ 8.15 (s, 1H), 7.84-7.80 (m, 2H), 7.07-7.02 (m, 1H), 6.71-6.68 (m, 1H), 5.59 (d, J=2.8 Hz, 1H), 4.15-4.12 (m, 4H), 3.53 (t, J=8.4 Hz, 1H), 3.40 (s, 3H), 3.23 (s, 4H), 2.41-2.37 (m, 4H), 2.14-2.11 (m, 1H), 1.71 (d, J=5.2 Hz, 18H). Step D. 4-[5-chloro-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-2-methylpyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-chloro-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-2-methylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.103 mmol, 1.00 eq) in DCM (1 mL) was added TFA (0.1 mL). The mixture was stirred for 6 h. The solvent was removed under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 27% B to 42% B in 8 min; wavelength: 254 nm; RT1(min): 7.5. The product-containing fractions were collected and concentrated in vacuo to give Example 58 (35 mg, 58.26%) as a white solid. m/z (ESI+ve ion)=580.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.68 (s, 1H), 10.42 (s, 1H), 9.16 (s, 1H), 7.40-7.37 (m, 2H), 6.91-6.88 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.69 (d, J=2.4 Hz, 1H), 3.92 (s, 4H), 3.32 (s, 3H), 3.29-3.26 (m, 4H), 3.20 (t, J=8.4 Hz, 1H), 2.34-2.30 (m, 1H), 2.13 (s, 3H), 2.00-1.97 (m, 1H). Example 59. (1R,2S)-2-(3-((2-cyclopropyl-&(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 6-chloro-2-cyclopropylpyrimidin-4-amine A mixture of 4,6-dichloro-2-cyclopropylpyrimidine (1.00 g, 5.290 mmol, 1.00 equiv) and NH3·H2O (13.00 mL, 0.371 mmol, 0.07 equiv) in THF (6.50 mL) was stirred for 6 h at 70° C. under nitrogen atmosphere and then cooled down to room temperature. The reaction was quenched with Water at room temperature. The resulting mixture was extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (860 mg, 95.85%) as an off-white solid. m/z=169.95 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 6.24 (s, 1H), 4.88 (s, 2H), 2.03 (m, 1H), 1.11 (m, 2H), 1.00 (m, 2H). Step B. 4-(6-amino-2-cyclopropylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione Into a 25 mL round-bottom flask were added 6-chloro-2-cyclopropylpyrimidin-4-amine (725.00 mg, 4.275 mmol, 1.00 equiv) and thiomorpholine 1,1-dioxide (2890.00 mg, 21.385 mmol, 5.00 equiv) at room temperature. The resulting mixture was stirred for 16 h at 100° C. under nitrogen atmosphere and cooled down to room temperature. The resulting mixture was diluted with MeOH (5 mL). The residue was purified by reverse phase flash chromatography with the following conditions: Column: Spherical C18, 20-40 um, 120 g; Mobile Phase A: Water (plus 5 mM NH4HCO); Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B-40% B in 20 min; Detector: 254 nm. The fractions containing desired product were collected at 22% B and concentrated under reduced pressure to afford the title compound (1.07 g, 93.28%) as a white solid. m/z (ESI+ve ion)=269.05 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 6.17 (s, 2H), 5.56 (s, 1H), 3.93 (t, J=5.1 Hz, 4H), 3.07 (t, J=5.1 Hz, 4H), 1.77 (m, 1H), 0.92-0.74 (m, 4H). Step C. tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-cyclopropyl-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 4-(6-amino-2-cyclopropylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (50.99 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Pd2(dba)3(14.50 mg, 0.016 mmol, 0.1 equiv) and XantPhos (9.16 mg, 0.016 mmol, 0.1 equiv) and Cs2C03(103.19 mg, 0.316 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The reaction was monitored by LCMS. The mixture was allowed to cool down to room temperature. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×8 mL). The combined organic layers were washed with brine (1×15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (96 mg, 78.54%) as a yellow solid. m/z (ESI+ve ion)=772.50 [M+H]+ Step D. 4-[2-cyclopropyl-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione A mixture of tert-butyl(1R,2S)-2-[I-(tert-butoxycarbonyl)-3-{[2-cyclopropyl-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (20.00 mg, 0.026 mmol, 1.00 equiv) and TFA (2.00 mL) in DCM (4.00 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 45% B in 8 min, 45% B: wavelength: 254 nm; RT1 (min): 6.88 to afford Example 59 (46.1 mg, 64.84%) as a white solid. m/z (ESI+ve ion)=572.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.36 (s, 1H), 10.42 (s, 1H), 9.74 (s, 1H), 7.96 (d, J=8.4 Hz, 1H), 7.33 (s, 1H), 7.08 (s, 1H), 6.92-6.85 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58 (m, 1H), 5.69 (d, J=2.8 Hz, 1H), 4.00 (d, J=6.5 Hz, 4H), 3.33 (s, 3H), 3.17 (m, 5H), 1.97 (m, 1H), 1.90 (m, 1H), 1.24 (s, 1H), 0.99-0.93 (m, 2H), 0.87 (m, 2H). Example 60. (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 4-(6-amino-2-methylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione The mixture of 6-chloro-2-methylpyrimidin-4-amine (1 g, 6.965 mmol, 1.00 equiv) and 1lambda6-thiomorpholine-1,1-dione (2.82 g, 20.895 mmol, 3 equiv) was stirred at 80° C. for 12 h. The reaction mixture was turned out to be white solid. The solid was triturated with EA (20 mL) for 2 h and filtered. The filter cake was collected and dried in vacuo to give title compound (500 mg, 29.33%) as a white solid. m/z (ESI+ve ion)=243.05 [M+H].1H NMR (400 MHz, DMSO-d6) δ 6.74 (s, 2H), 5.70 (s, 1H), 4.00-3.98 (m, 4H), 3.16-3.13 (m, 4H), 2.27 (s, 3H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-2-methylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 4-(6-amino-2-methylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (49.9 mg, 1.3 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbo-nyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100.0 mg, 1.0 equiv) in toluene (2.5 mL) were added Cs2CO3(103.3 mg, 2.0 equiv) and XantPhos (16.5 mg, 0.2 equiv) and Pd2(dba)3(29.0 mg, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×6 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EtOAc in PE to afford the title compound (100.0 mg, 84.7%) as a yellow solid. m/z (ESI, +ve ion)=746.40 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.16 (s, 1H), 7.82 (d, J=12.0 Hz, 1H), 7.75 (s, 1H), 7.61 (d, J=8.0 Hz, 1H), 7.13 (d, J=8.0 Hz, 1H), 6.71-6.68 (m, 1H), 5.57 (d, J=4.0 Hz, 1H), 4.35-4.31 (m, 4H), 4.17-4.12 (m, 1H), 3.55-3.50 (m, 1H), 3.42 (s, 3H), 3.17-3.11 (m, 4H), 2.49 (s, 3H), 2.40-2.37 (m, 1H), 2.14-2.11 (m, 1H), 1.70 (s, 18H). Step C. 4-[6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-2-methylpyrimidin-4-yl]-1lambda6-thiomorpholine-1,1 To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-2-methylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (90.0 mg) in DCM (4.0 mL) was added TFA (0.5 mL) dropwise at room temperature under air atmosphere. The resulting mixture was stirred for 5 h at room temperature. The resulting mixture was concentrated under vacuum. The crude product (50.0 mg) was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3). Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 40% B in 8 min, 40% B; wavelength: 254 nm; RT1(min): 6.6 to afford Example 60 (23.0 mg, 35.0% yield) as a white solid. m/z (ESI, +ve ion)=546.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.37 (s, 1H), 10.42 (s, 1H), 9.89 (s, 1H), 7.98 (d, J=4.0 Hz, 1H), 7.33 (s, 1H), 7.20 (s, 1H), 6.89 (d, J=4.0 Hz, 1H), 6.75 (d, J=8.0 Hz, 1H), 6.60-6.57 (m, 1H), 5.69 (d, J=0.0 Hz, 1H), 4.04 (d, J=5.6 Hz, 4H), 3.17 (s, 5H), 2.50 (s, 3H), 2.33 (d, J=8.0 Hz, 4H), 1.99-1.95 (m, 1H). Example 61. 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-6-(morpholin-4-yl)pyrimidine-2-carbonitrile Step A. 6-hydroxy-5-methoxy-2-sulfanyl-3H-pyrimidin-4-one To a stirred mixture of thiourea (3.00 g, 39.411 mmol, 1.00 equiv) and 1,3-dimethyl 2-methoxypropanedioate (6.39 g, 39.411 mmol, 1.00 equiv) in MeOH (80.00 mL) was added sodium 2-methylpropan-2-olate (7.58 g, 78.822 mmol, 2.00 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 7 h at 50° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The resulting mixture was diluted with water (50 mL). The resulting mixture was extracted with EtOAc (2×40 mL). The aqueous layer was acidified to pH 1 with conc. HCl. The precipitated solids were collected by filtration and washed with water (2×10 mL) and dried to afford the title compound (4 g, 58.27%) as a yellow solid. m/z (ESI, +ve ion)=175.05 [M+H]+. Step B. 2-(ethylsulfanyl)6-hydroxy-5-methoxy-3H-pyrimidin-4-one To a stirred mixture of 6-hydroxy-5-methoxy-2-sulfanyl-3H-pyrimidin-4-one (4.00 g, 22.966 mmol, 1.00 equiv) and iodoethane (7.16 g, 0.000 mmol, 2.00 equiv) in water (120 mL) was added NaOH (1.84 g, 0.046 mmol, 2.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at room temperature under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with water (2×20 mL). The filtrate was acidified to pH 1 with HCl (aq.). The precipitated solids were collected by filtration and washed with water (2×30 mL) and dried to afford the title compound (2.27 g, 48.88%) as a yellow solid. m/z (ESI, +ve ion)=203.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.23 (s, 1H), 11.25 (s, 1H), 3.59 (d, J=6.0 Hz, 3H), 3.07 (q, J=7.3 Hz, 2H), 1.28 (t, J=7.3 Hz, 3H). Step C. 4,6-dichloro-2-(ethylsulfanyl)-5-methoxypyrimidine A mixture of 2-(ethylsulfanyl)-6-hydroxy-5-methoxy-3H-pyrimidin-4-one (2.27 g, 11.225 mmol, 1.00 equiv) and PhNEt2(5.03 g, 0.034 mmol, 3.00 equiv) in POCl3(38.00 mL) was stirred for 3 h at 100° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched with sat. NaHCO3(aq. 30 mL) at 0° C. The resulting mixture was extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography with the following conditions: Column: Spherical C18, 20-40 um, 120 g; Mobile Phase A: Water (plus 0.05% TFA); Mobile Phase B: ACN; Flow rate: 80 mL/min; Gradient: 60% B-70% B in 15 min: Detector: 254 nm. The fractions containing desired product were collected at 64% B and concentrated under reduced pressure to afford the title compound (2.5 g, 93.15%) as a dark green oil. m/z=238.95 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 3.87 (s, 3H), 3.13-3.09 (m, 2H), 1.71-1.46 (m, 3H). Step D. 4-[6-chloro-2-(ethylsulfanyl)-5-methoxypyrimidin-4-yl] morpholine A mixture of 4,6-dichloro-2-(ethylsulfanyl)-5-methoxypyrimidine (400.00 mg, 1.673 mmol, 1.00 equiv) and morpholine (174.89 mg, 0.000 mmol, 1.20 equiv) in EtOH (10.00 mL) was stirred for 2 h at 85° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched with Water at room temperature. The resulting mixture was extracted with DCM (3×15 mL). The combined organic layers were washed with brine (2×20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (3/1) to afford the title compound (230 mg, 47.45%) as a white solid. m/z (ESI, +ve ion)=290.15 [M+H]+. Step E. 4-[6-chloro-2-(ethanesulfonyl)-5-methoxypyrimidin-4-yl] morpholine A mixture of 4-[6-chloro-2-(ethylsulfanyl)-5-methoxypyrimidin-4-yl] morpholine (430.00 mg, 1.484 mmol, 1.00 equiv) and m-CPBA (896.24 mg, 5.194 mmol, 3.5 equiv) in DCM (10.75 mL) were stirred for 3 h at room temperature under nitrogen atmosphere. The reaction was quenched with sat. Na2S2O3(aq., 15 mL) at room temperature. The resulting mixture was extracted with DCM (2×15 mL). The combined organic layers were washed with sat. NaHCO3(aq. 20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (2/1) to afford the title compound (450 mg, 94.24%) as an off-white solid. m/z (ESI, +ve ion)=322.00 [M+H]+. Step F. 4-chloro-5-methoxy-6-(morpholin-4-yl) pyrimidine-2-carbonitrile Into a 50 mL round-bottom flask were added 4-[6-chloro-2-(ethanesulfonyl)-5-methoxypyrimidin-4-yl] morpholine (465.00 mg, 1.445 mmol, 1.00 equiv) and DMSO (11.62 mL) at room temperature. To the above mixture was added NaCN (106.23 mg, 2.168 mmol, 1.5 equiv) in portions at room temperature. The resulting mixture was stirred for additional 2 h at room temperature. The reaction was quenched with Water (15 mL) at room temperature. The resulting mixture was extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (2/1) to afford the title compound (122 mg, 33.15%) as an off-white solid. m/z (ESI, +ve ion)=255.15 [M+H]+. Step G. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((2-cyano-5-methoxy-6-morpholinopyrimidin-4-yl)amino)-1H-indazol-6-yl 5′-methoxy-2′-oxospiro[cyclopropane-1′-indoline]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70 mg, 0.134 mmol, 1.00 equiv) and 4-chloro-5-methoxy-6-(morpholin-4-yl) pyrimidine-2-carbonitrile (34.24 mg, 0.134 mmol, 1 equiv) in dioxane (2 mL) were added Methanesulfonato(2-bis(3,5-di(trifluoromethyl)phenylphosphino)-3,6-dimethoxy-2′,6′-bis(dimethylamino)-1,1′-biphenyl)(2′-methylamino-1,1′-biphenyl-2-yl)palladium (II (15.35 mg, 0.013 mmol, 0.1 equiv) and 2′-(Bis(3,5-bis(trifluoromethyl)phenyl)phosphino)-3′,6′-dimethoxy-N2,N2,N6,N6-tetramethyl-[1,1′-biphenyl]-2,6-diamine (10.17 mg, 0.013 mmol, 0.1 equiv) and Cs2CO3(87.62 mg, 0.268 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (8 mL) at room temperature. The resulting mixture was extracted with DCM (3×5 mL). The combined organic layers were washed with brine (2×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (89 mg, 89.59%) as a brown yellow solid. m/z (ESI, +ve ion)=739.25 [M+H]+. Step H A mixture of tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((2-cyano-5-methoxy-6-morpholinopyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (89 mg, 0.120 mmol, 1.00 equiv) and TFA (2 mL, 26.926 mmol, 223.52 equiv) in DCM (4 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 19×250 mm, 5 un; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: MeOH-HPLC; Flow rate: 25 mL/min; Gradient: 50% B to 60% B in 10 min, 60% B to 60% B in 15 min, 60% B; wavelength: 254 nm; RT1(min): 9 to afford Example 61 (17.6 mg, 27.13%) as a pink solid. m/z (ESI, +ve ion)=539.25 [M+H]+.1H NMR (400 MHz, DMSO-d) S 12.72 (s, 1H), 10.42 (s, 1H), 9.40 (s, 1H), 7.47-7.39 (m, 2H), 6.96-6.91 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.69-6.55 (m, 1H), 5.68 (d, J=2.4 Hz, 1H), 3.72 (d, J=7.2 Hz, 7H), 3.68-3.62 (m, 4H), 3.31 (s, 3H), 3.24-3.15 (m, 2H), 2.34-2.32 (m, 1H), 2.01-1.97 (m, 1H). Example 62. 4-(1,1-dioxidothiomorpholino)-5-methoxy-6-((6-((1R,2S)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazol-3-yl)amino)pyrimidine-2-carbonitrile Step A. 4-[6-chloro-2-(ethylsulfanyl)-5-methoxypyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione To a stirred mixture of 4,6-dichloro-2-(ethylsulfanyl)-5-methoxypyrimidine (800.00 mg, 3.346 mmol, 1.00 equiv) and 1lambda6-thiomorpholine-1,1-dione (452.28 mg, 3.346 mmol, 1.00 equiv) in dioxane (20.00 mL) were added TEA (1354.22 mg, 13.383 mmol, 4 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 100° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (20 mL) at room temperature. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1) to afford the title compound (443 mg, 39.19%) as a white solid. m/z (ESI, +ve ion)=338.15 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 4.36-4.29 (m, 4H), 3.76 (s, 3H), 3.17-3.12 (m, 4H), 3.08 (q, J=7.3 Hz, 2H), 1.40 (t, J=7.3 Hz, 3H). Step B. 4-[6-chloro-2-(ethanesulfonyl)-5-methoxypyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione A mixture of 4-[6-chloro-2-(ethylsulfanyl)-5-methoxypyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione (417.00 mg, 1.234 mmol, 1.00 equiv) and m-CPBA (745.50 mg, 4.320 mmol, 3.50 equiv) in DCM (10.00 mL) was stirred for 3 h at room temperature under nitrogen atmosphere. The reaction was quenched with Na2S2O3(aq. 10 mL) at room temperature. The resulting mixture was extracted with DCM (2×15 mL). The combined organic layers were washed with NaHCO3(aq., 25 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (3/1) to afford the title compound (400 mg, 87.62%) as an off-white solid. m/z (ESI, +ve ion)=370.00 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 4.27 (t, J=5.1 Hz, 4H), 3.81 (s, 3H), 3.56-3.48 (m, 2H), 3.34 (t, J=5.1 Hz, 4H), 1.29-1.25 (m, 3H). Step C. 4-chloro-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxypyrimidine-2-carbonitrile To a stirred mixture of 4-[6-chloro-2-(ethanesulfonyl)-5-methoxypyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione (460 mg, 1.244 mmol, 1.00 equiv) and ACN (4.6 mL) in DMSO (11.5 mL) was added NaCN (91.43 mg, 1.866 mmol, 1.5 equiv) at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 0° C. under nitrogen atmosphere. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (158 mg, 41.96%) as an off-white solid. m/z (ESI, +ve ion)=302.90 [M+H]+. Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-cyano-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.192 mmol, 1.00 equiv) and 4-chloro-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxypyrimidine-2-carbonitrile (69.78 mg, 0.230 mmol, 1.2 equiv) in dioxane (5.00 mL, 58.992 mmol, 307.25 equiv) were added CPhos (8.39 mg, 0.019 mmol, 0.1 equiv) and Pd2(dba)3·CHCl3(19.88 mg, 0.019 mmol, 0.1 equiv) and Cs2CO3(125.17 mg, 0.384 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (10 mL) at room temperature. The residue was purified by reverse phase flash chromatography with the following conditions: Column: Spherical C18, 20-40 μm, 80 g; Mobile Phase A: Water (plus 5 mM NH4HCO3); Mobile Phase B: ACN; Flow rate: 40 mL/min; Gradient: 60% B-80% B in 20 min; Detector: 254 nm. The fractions containing desired product were collected at 70% B and concentrated under reduced pressure to afford the title compound (54 mg, 35.73%) as an off-white solid. m/z (ESI, +ve ion)=787.25 [M+H]+. Step E. 4-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxy-6-{(6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl} amino) pyrimidine-2-carbonitrile A mixture of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-{[2-cyano-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (54 mg, 0.069 mmol, 1.00 equiv) and TFA (2 mL, 26.926 mmol, 392.35 equiv) in DCM (4 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 19×250 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: MeOH-HPLC; Flow rate: 25 mL/min; Gradient: 50% B to 60% B in 10 min, 60% B to 60% B in 15 min, 60% B; Wavelength: 254 nm; RT1(min): 9 to afford Example 62 (10.7 mg, 26.31%) as a white solid. MS: m/z=587.25 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.59 (d, J=8.4 Hz, 1H), 7.43 (s, 1H), 6.98 (d, J=8.4 Hz, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.62 (m, 1H), 5.58 (d, J=2.4 Hz, 1H), 4.26 (d, J=5.9 Hz, 4H), 3.83 (s, 3H), 3.42 (m, 1H), 3.37 (s, 3H), 3.24 (t, J=4.8 Hz, 4H), 2.25 (m, 1H), 2.19 (m, 1H). Example 63. (1R,2S)-2-{3-[(1,3-dimethyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(1,3-dimethylpyrazol-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 1,3-dimethylpyrazol-4-amine (21.12 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h, then filtered and washed with EA (5 mL×3). The filtrate was concentrated in vacuo. The residue was purified by Prep-TLC (rinsed with EA/PE=1/1) to afford the title compound (60 mg, 58.55%) as a yellow solid. m/z (ESI+ve ion)=615.50 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.05 (s, 1H), 7.89 (s, 1H), 7.81 (d, J=8.8 Hz, 1H), 6.99 (d, J=8.4 Hz, 1H), 6.69 (d, J=9.2 Hz, 1H), 5.92 (s, 1H), 5.57 (d, J=1.6 Hz, 1H), 3.89 (s, 3H), 3.52-3.48 (m, 1H), 3.40 (d, J=1.2 Hz, 3H), 2.38-2.34 (m, 1H), 2.27 (s, 3H), 2.11-2.05 (m, 1H), 1.70 (s, 18H). Step B. (1R,2S)-2-{3-[(1,3-dimethylpyrazol-4-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(1,3-dimethylpyrazol-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (55 mg, 0.089 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (5 mL) was stirred at 25° C. for 4 h. The solvent was removed under reduced pressure and the residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 35% B in 8 min; Wavelength: 254 nm; RT1(min): 6.6. The product-containing fractions was collected and concentrated in vacuo to give Example 63 (8.8 mg, 22.54%) as an off-white solid. m/z (ESI+ve ion)=415.35 [M+H].1H NMR (400 MHz, Methanol-d4) δ 7.76 (s, 1H), 7.65 (d, J=8.4 Hz, 1H), 7.25 (s, 1H), 6.85-6.80 (m, 2H), 6.64-6.61 (m, 1H), 5.61 (d, J=2.4 Hz, 1H), 3.82 (s, 4H), 3.31 (s, 3H), 2.22 (s, 3H), 2.18-2.15 (m, 1H). Example 64. (1R,2S)-5′-methoxy-2-(3-{[1-methyl-3-(trifluoromethyl)-1H-pyrazol-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[1-methyl-3-(trifluoromethyl)pyrazol-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 1-methyl-3-(trifluoromethyl)pyrazol-4-amine (31.38 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), CPhos (13.83 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(32.78 mg, 0.032 mmol, 0.2 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h. The reaction mixture was filtered and washed with EA (5 mL×3). The filtrate was concentrated in vacuo. The residue was purified by Prep-TLC (rinsed with EA/PE=1/2) to afford crude the title compound (23a) (80 mg, 75%) as a yellow solid. m/z (ESI+ve ion)=669.50 [M+H]+. Step B. (1R,2S)-5′-methoxy-2-(3-{[1-methyl-3-(trifluoromethyl)pyrazol-4-yl]amino}-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of crude tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[1-methyl-3-(trifluoromethyl)pyrazol-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (75 mg) in TFA (0.5 mL) and DCM (3 mL) was stirred for 4 h at 25° C. The solvent was removed under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 50% B in 8 min, 50% B; wavelength: 254 nm; RT1: 7 min. The product-containing fractions was concentrated in vacuo to give Example 64 (29.8 mg, 40.01% over two steps) as a white solid. m/z (ESI +ve ion)=469.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 11.81 (s, 1H), 10.40 (s, 1H), 8.26 (s, 1H), 8.04 (s, 1H), 7.83 (d, J=8.4 Hz, 1H), 7.24 (s, 1H), 6.86-6.84 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.70 (d, J=2.4 Hz, 1H), 3.91 (s, 3H), 3.33 (s, 3H), 3.17 (t, J=8.4 Hz, 1H), 2.31-2.27 (m, 1H), 2.00-1.98 (m, 1H).19F NMR (376 MHz, DMSO-d6) δ −58.60 (s, 3F). Example 65. (1R,2S)-5′-methoxy-2-{3-[(1-methyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(1-methylpyrazol-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 1-methylpyrazol-4-amine (18.46 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), CPhos (13.83 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(32.78 mg, 0.032 mmol, 0.2 equiv) under argon atmosphere. After the mixture was stirred at 90° C. for 2 h, it was cooled down to room temperature, filtered and washed with EA (5 mL×3). The filtrate was concentrated in vacuo. The residue was purified by Prep-TLC (rinsed with EA/PE=1/2) to afford the title compound (30 mg, 31.54%) as a yellow solid. m/z (ESI+ve ion)=601.50 [M+H]+. Step B. (1R,2S)-5′-methoxy-2-{3-[(1-methylpyrazol-4-yl)amino]-1H-indazol-6-yl}-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(1-methylpyrazol-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (30 mg, 0.050 mmol, 1.00 equiv) in TFA (0.2 mL) and DCM (2 mL) was stirred for 5 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 45% B in 8 min; Wavelength: 254 nm; RT1(min): 6.5. The product-containing fractions were collected and concentrated in vacuo to give Example 65 (9.3 mg, 46.27%) as a white solid. m/z (ESI+ve ion)=401.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.87 (s, 1H), 7.67 (d, J=8.4 Hz, 1H), 7.52 (s, 1H), 7.26 (s, 1H), 6.83 (d, J=8.4 Hz, 2H), 6.64-6.61 (m, 1H), 5.61 (d, J=2.4 Hz, 1H), 3.88 (s, 3H), 3.36-3.33 (s, 1H), 3.29 (s, 3H), 2.22-2.15 (m, 2H). Example 66. 4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-1-methyl-1H-pyrazole-3-carbonitrile Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(1-methylpyrazol-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 4-amino-1-methylpyrazole-3-carbonitrile (23.21 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), CPhos (13.83 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(32.78 mg, 0.032 mmol, 0.2 equiv) under argon atmosphere. The mixture was stirred at 90° C. for 2 h and then cooled down to room temperature. The reaction mixture was filtered and washed with EA (5 mL×3). The filtrate was concentrated in vacuo. The residue was purified by Prep-TLC (rinsed with EA/PE=1/2) to afford crude title compound (25a) (28 mg, 22.61%) as a yellow solid. m/z (ESI+ve ion)=526.35 [M+H−100]+. Step B. 4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-1-methylpyrazole-3-carbonitrile The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(1-methylpyrazol-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (26 mg, 0.033 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (2.5 mL) was stirred at 25° C. for 5 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 18% B to 28% B in 8 min; wavelength: 254 nm; RT1(min): 7. The product containing fractions were collected and concentrated in vacuo to give Example 66 (4.0 mg, 28.00%) as a yellow solid. m/z (ESI+ve ion)=426.35 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.36 (s, 1H), 8.04 (d, J=8.4 Hz, 1H), 7.44 (s, 1H), 6.85-6.79 (m, 2H), 6.62-6.60 (m, 1H), 5.76 (d, J=2.0 Hz, 1H), 4.37 (s, 3H), 3.40-3.38 (m, 1H), 3.25 (s, 3H), 2.31-2.28 (m, 1H), 2.22-2.18 (m, 1H). Example 67. (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxy-2-methylpyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. (2R,6S)-4-(6-chloro-5-methoxy-2-methylpyrimidin-4-yl)-2,6-dimethylmorpholine A mixture of 4,6-dichloro-5-methoxy-2-methylpyrimidine (300 mg, 1.554 mmol, 1.00 equiv) and (2R,6S)-2,6-dimethylmorpholine (179.00 mg, 1.554 mmol, 1 equiv) in THF (7.5 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford the title compound (211 mg, 49.96%) as a colorless oil. m/z=272.00 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxy-2-methylpyrimidin-4-yl} amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.154 mmol, 1.00 equiv) and (2R,6S)-4-(6-chloro-5-methoxy-2-methylpyrimidin-4-yl)-2,6-dimethylmorpholine (41.76 mg, 0.154 mmol, 1 equiv) in dioxane (4 mL, 47.216 mmol, 307.25 equiv) were added methanesulfonato(2-bis(3,5-di(trifluoromethyl)phenylphosphino)-3,6-dimethoxy-2′,6′-bis(dimethylamino)-1,1′-biphenyl)(2′-methylamino-1,1′-biphenyl-2-yl)palladium (II) (17.54 mg, 0.015 mmol, 0.1 equiv) and 2′-(Bis(3,5-bis(trifluoromethyl)phenyl)phosphino)-3′,6′-dimethoxy-N2,N2,N6,N6-tetramethyl-[1,1′-biphenyl]-2,6-diamine (11.63 mg, 0.015 mmol, 0.1 equiv) and Cs2CO3(100.14 mg, 0.308 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The reaction was quenched by the addition of Water (8 mL) at room temperature. The resulting mixture was extracted with DCM (3×8 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford the title compound (74.5 mg, 64.14%) as a yellow solid. m/z (ESI, +ve ion)=756.30 [M+H]+. Step C. (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxy-2-methylpyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxy-2-methylpyrimidin-4-yl} amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (74.5 mg, 0.099 mmol, 1.00 equiv) and TFA (2 mL, 26.926 mmol) in DCM (4 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 35% B to 45% B in 8 min, 45% B; wavelength: 254 nm; RT1(min): 7.2 to afford Example 67 (28.1 mg, 50.80%) as a white solid. m/z (ESI, +ve ion)=556.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.53 (s, 1H), 10.43 (s, 1H), 8.75 (s, 1H), 7.36 (d, J=8.3 Hz, 1H), 7.37 (s, 1H), 6.88 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (m, 1H), 5.71 (d, J=2.4 Hz, 1H), 4.23 (d, J=12.8 Hz, 2H), 3.67 (t, J=8.6 Hz, 2H), 3.62 (s, 3H), 3.30 (s, 4H), 3.18-3.16 (m, 2H), 2.35-2.28 (m, 1H), 2.07 (s, 3H), 1.98 (m, 1H), 1.13 (d, J=6.4 Hz, 6H). Example 68. (1R,2S)-2-(3-{[2-(2-hydroxyethyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. methyl 2-(4-hydroxy-5-methoxy-6-oxo-1H-pyrimidin-2-yl)acetate To a mixture of t-BuONa (12.01 g, 124.992 mmol, 2.5 equiv) in MeOH (100 mL) were added ethyl 2-carbamimidoylacetate hydrochloride (8.33 g, 49.997 mmol, 1.00 equiv) and 1,3-dimethyl 2-methoxypropanedioate (8.11 g, 49.997 mmol, 1 equiv). The mixture was stirred for 3 h at 70° C. The mixture was quenched with water (50 mL) and then MeOH was evacuated under reduced pressure and then extracted with CHCl3(3×30 mL). The combined organic layers were dried over anhydrous Na2SO4and then concentrated under reduced pressure to afford the title compound (6 g, 56.03%) as a yellow oil. m/z (ESI, +ve ion)=215.05 [M+H]+ Step B. methyl 2-(4,6-dichloro-5-methoxypyrimidin-2-yl)acetate A mixture of methyl 2-(4-hydroxy-5-methoxy-6-oxo-1H-pyrimidin-2-yl)acetate (400.00 mg, 1 equiv) in POCl3(5.00 mL) was stirred for 3 h at 100° C. The mixture was concentrated under reduced pressure. The residue was purified with silica gel chromatography, eluted with 20% EA in PE to afford the title compound (70 mg, 14.93%) as a yellow solid. m/z (ESI, +ve ion)=250.95 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 3.97 (s, 3H), 3.94 (s, 2H), 3.75 (s, 3H). Step C. methyl 2-[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]acetate To a mixture of methyl 2-(4,6-dichloro-5-methoxypyrimidin-2-yl)acetate (500 mg, 1.992 mmol, 1.00 equiv) and TEA (403.05 mg, 3.984 mmol, 2 equiv) in THF (10 mL, 123.430 mmol, 61.98 equiv) was added morpholine (190.86 mg, 2.191 mmol, 1.1 equiv) at 0° C. The resulting mixture was stirred for 16 h at room temperature. The reaction was concentrated under reduced pressure to afford the title compound (600 mg, 99.85%) as a yellow oil. m/z (ESI, +ve ion)=302.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 3.79-3.87 (m, 4H), 3.77 (d, J=4.4 Hz, 7H), 3.73 (d, J=2.1 Hz, 6H). Step D. 2-[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]ethanol To a mixture of methyl 2-[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]acetate (1500 mg, 4.97 mmol, 1.00 equiv) in MeOH (50 mL) was added NaBH4(1128.5 mg, 29.82 mmol, 6 equiv) at 0° C. The resulting mixture was stirred for 3 h at room temperature. The reaction was quenched with water (50 mL), extracted with EA (3×100 mL). The combined organic layers were dried over anhydrous Na2SO4and then concentrated under reduced pressure. The residue was purified with silica gel chromatography, eluted with PE:EA=1:1 to afforded the title compound (800 mg, 58.79%) as a yellow oil. m/z (ESI, +ve ion)=274.15 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 4.01 (s, 2H), 3.80 (s, 8H), 3.74 (s, 3H), 2.96 (t, J=5.4 Hz, 2H). Step E. 4-(2-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-6-chloro-5-methoxypyrimidin-4-yl)morpholine To a mixture of 2-[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]ethanol (700 mg, 0.256 mmol, 1.00 equiv) and Imidazole (348.2 mg, 5.12 mmol, 2 equiv) in DMF (5 mL) was added TBSCl (578.2 mg, 3.84 mmol, 1.5 equiv). The resulting mixture was stirred for 16 h at room temperature. The mixture was concentrated under reduced pressure. The residue was purified with silica gel chromatography, eluted with PE/EA 4/1 to afford the title compound (860 mg, 86.68%) as a colorless oil. m/z (ESI, +ve ion)=388.15 [M+H]+. Step F. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a mixture of 4-(2-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-6-chloro-5-methoxypyrimidin-4-yl)morpholine (58 mg, 0.149 mmol, 1.00 equiv), tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (77.82 mg, 0.149 mmol, 1 equiv) and Cs2CO3(97.42 mg, 0.298 mmol, 2 equiv) in dioxane (5 mL) were added CPhos (6.53 mg, 0.015 mmol, 0.1 equiv) and Pd2(dba)3·CHCl3(15.47 mg, 0.015 mmol, 0.1 equiv) under N2atmosphere. The resulting mixture was stirred for 90° C. for 4 h. The mixture was concentrated under reduced pressure. The residue was purified with prep-TLC, eluted with PE/EA 1/1 to afford the title compound (20 mg, 15.34%) as a yellow oil. m/z (ESI, +ve ion)=872.40 [M+H]+ Step G. (1R,2S)-2-(3-{[2-(2-hydroxyethyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.115 mmol, 1.00 equiv) in DCM (2 mL) was added TFA (1 mL). The resulting mixture was stirred for 16 h at room temperature. The mixture was concentrated under reduced pressure. The residue was purified with prep-HPLC with following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 40% B in 8 min, 40% B; wavelength: 254 nm to afford Example 68 (26 mg, 40.66%) as a white solid. m/z (ESI, +ve ion)=558.50 [M+H]+,1H NMR (400 MHz, Methanol-d4) δ 7.64 (d, J=8.4 Hz, 1H), 7.41 (d, J=1.2 Hz, 1H), 6.93 (dd, J=8.4, 1.4 Hz, 1H), 6.84 (d, J=8.5 Hz, 1H), 6.63 (dd, J=8.5, 2.5 Hz, 1H), 5.65 (d, J=2.5 Hz, 1H), 3.75-3.85 (m, 9H), 3.73-3.71 (m, 4H), 3.38-3.37 (m, 1H), 3.30 (s, 3H), 2.71 (t, J=6.2 Hz, 2H), 2.24-2.18 (m, 2H). Example 69. 4-[2-cyclopropyl-5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidin-4-yl]-1 λ6-thiomorpholine-1,1-dione Step A. 4-(6-chloro-2-cyclopropyl-5-methoxypyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione To a stirred solution of 4,6-dichloro-2-cyclopropyl-5-methoxypyrimidine (400.0 mg, 1.0 equiv) and 1lambda6-thiomorpholine-1,1-dione (271.5 mg, 1.1 equiv) in THF (8.0 mL) was added TEA (369.5 mg, 2.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 70° C. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to the title compound (300.0 mg, 51.8% yield) as a white solid. m/z (ESI+ve ion)=318.05 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-cyclopropyl-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[3-amino-1-tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.192 mmol, 1.00 equiv) and 4-(6-chloro-2-cyclopropyl-5-methoxypyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (73.25 mg, 0.230 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(125.17 mg, 0.384 mmol, 2 equiv), CPhos (16.77 mg, 0.038 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(39.77 mg, 0.038 mmol, 0.2 equiv) under nitrogen atmosphere. The resulting mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (5 mL×3). The filtrate was concentrated and the residue was purified by prep-TLC (rinsed with EA/PE=1/1) to give crude the title compound (50 mg, 25.97%) as a light yellow solid. m/z (ESI+ve ion)=802.65 [M+H]+. Step C. 4-[2-cyclopropyl-5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-cyclopropyl-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (50 mg, 0.062 mmol, 1.00 equiv) in TFA (0.10 mL) and DCM (1.00 mL) was stirred at 25° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm. Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 45% B in 8 min; wavelength: 254/220 nm; RT1 (min): 6. The product-containing fractions was collected and concentrated in vacuo to give Example 69 (36 mg, 95.48%) as a white solid. m/z (ESI+ve ion)=602.40 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.54 (s, 1H), 10.43 (s, 1H), 8.88 (s, 1H), 7.43 (d, J=8.4 Hz, 1H), 7.37 (s, 1H), 6.88 (d, J=8.0 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59-6.57 (m, 1H), 5.69 (d, J=2.4 Hz, 1H), 4.07-4.05 (m, 4H), 3.63 (s, 3H), 3.32 (s, 3H), 3.23-3.17 (m, 5H), 2.34-2.30 (m, 1H), 2.00-1.97 (m, 1H), 1.64-1.60 (m, 1H), 0.66-0.64 (m, 4H). Example 70. 4-[5-chloro-2-cyclopropyl-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidin-4-yl]-1 λ6-thiomorpholine-1,1-dione Step A. 4-(6-amino-5-chloro-2-cyclopropylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione A mixture of 4-(6-amino-2-cyclopropylpyrimidin-4-yl)thiomorpholine 1,1-dioxide (400 mg, 1.491 mmol, 1.00 equiv) and NCS (238.86 mg, 1.789 mmol, 1.2 equiv) in THF (20 mL) was stirred for 5 h at 50° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (420 mg, 93.06%) as an off-white solid. m/z (ESI, +ve ion)=302.90 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 6.81 (s, 2H), 3.87-3.80 (m, 4H), 3.24-3.17 (m, 4H), 1.82 (m, 1H), 0.87 (d, J=6.4 Hz, 4H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-chloro-2-cyclopropyl-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-F-carboxylate (80 mg, 0.127 mmol, 1.00 equiv) and 4-(6-amino-5-chloro-2-cyclopropylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (46.03 mg, 0.152 mmol, 1.2 equiv) in toluene (4 mL) were added Xantphos Pd G4(12.19 mg, 0.013 mmol, 0.1 equiv) and XantPhos (7.33 mg, 0.013 mmol, 0.1 equiv) and Cs2CO3(82.56 mg, 0.254 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 9 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (46b) (50 mg, 48.95%) as a brown yellow solid. m/z=806.35 [M+H]+. Step C. 4-[5-chloro-2-cyclopropyl-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl} amino)pyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-chloro-2-cyclopropyl-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (50 mg, 0.062 mmol, 1.00 equiv) and TFA (2 mL, 26.926 mmol, 434.23 equiv) in DCM (4 mL) was stirred for 2 h at mom temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 m mol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 40% B in 8 min, 40% B: wavelength: 254 nm; RT1(min): 7.63 to afford Example 70 (16.1 mg, 42.24%) as a white solid. m/z (ESI, +ve ion)=606.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.67 (s, 1H), 10.42 (s, 1H), 9.13 (s, 1H), 7.40 (s, 1H), 7.35 (d, J=8.4 Hz, 1H), 6.90 (d, J=8.6 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58 (m, 1H), 5.68 (d, J=2.4 Hz, 1H), 3.91 (s, 4H), 3.27 (s, 4H), 3.20 (t, J=8.4 Hz, 1H), 2.50 (s, 3H), 2.35-2.28 (m, 1H), 1.99 (m, 1H), 1.63 (m, 1H), 0.66 (m, 4H). Example 71. (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyrazin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 3-methoxy-6-methylpyrazin-2-amine A solution of 6-chloro-3-methoxypyrazin-2-amine (200 mg, 1.253 mmol, 1.00 equiv) and Ni(dppp)Cl2(67.94 mg, 0.125 mmol, 0.1 equiv) in dioxane (3 mL) was stirred for 5 min at room temperature under nitrogen atmosphere. To the above mixture was added 1 M MeZnCl (5.01 mL, 5.012 mmol, 4 equiv) at room temperature. The resulting mixture was stirred for additional 3 min at room temperature. The resulting mixture was stirred for 4 h at 100° C. under nitrogen atmosphere then cooled down to room temperature. The reaction was quenched by the addition of Water (8 mL) at room temperature. The resulting mixture was extracted with EtOAc (3×8 mL). The combined organic layers were washed with brine (2×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM/MeOH 20/1) to afford the title compound as a white solid. m/z (ESI+ve ion)=140.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.11-7.10 (m, 1H), 6.15 (s, 2H), 3.83 (s, 3H), 2.14 (d, J=0.84 Hz, 3H). Step B. tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-methoxy-6-methylpyrazin-2-yl) amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 3-methoxy-6-methylpyrazin-2-amine (21.16 mg, 0.152 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.127 mmol, 1.00 equiv) in toluene (1.5 mL) were added XantPhos (7.33 mg, 0.013 mmol, 0.1 equiv) and Pd2(dba). (23.20 mg, 0.025 mmol, 0.2 equiv) and Cs2CO3(82.56 mg, 0.254 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 4 h at room temperature under nitrogen atmosphere then quenched by the addition of Water (5 mL) at room temperature. The resulting mixture was extracted with EtOAc (3×5 mL). The combined organic layers were washed with brine (2×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (2/1) to afford the title compound (70 mg, 85.97%) as a yellow oil. m/z (ESI+ve ion)=643.50 [M+H]+.1H NMR (400 MHz, Chloroform-d) 8.16 (s, 1H), 7.93 (d, J=8.4 Hz, 1H), 7.81 (d, J=8.9 Hz, 1H), 7.66 (s, 1H), 7.46 (s, 1H), 7.04 (d, J=8.3 Hz, 1H), 6.71-6.69 (m, 1H), 4.05 (s, 3H), 3.56-3.51 (m, 1H), 3.37 (s, 3H), 2.39-2.36 (m, 1H), 2.30 (s, 3H), 2.15-2.12 (m, 1H), 1.29 (s, 18H). Step C. (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyrazin-2-yl) amino]-1H-indazol-6-yl}-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-methoxy-6-methylpyrazin-2-yl) amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (65 mg, 0.101 mmol, 1.00 equiv) was added TFA (1 mL, 13.463 mmol, 133.12 equiv) and DCM (2 mL, 31.460 mmol, 311.07 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 4 h at room temperature under nitrogen atmosphere. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 50% B in 8 min, 50% B; wavelength: 254 nm; RT1(min): 6.5 to afford Example 71 (13.5 mg, 30.17%) as a white solid. m/z (ESI+ve ion)=443.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.52 (s, 1H), 10.41 (s, 1H), 8.71 (s, 1H), 7.44 (d, J=8.4 Hz, 1H), 7.37 (s, 1H), 7.31 (s, 1H), 6.87 (d, J=9.2 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.59-6.57 (m, 1H), 5.71 (d, J=2.4 Hz, 1H), 3.94 (s, 3H), 3.32 (s, 3H), 3.21-3.16 (m, 1H), 2.34-2.28 (m, 1H), 2.07 (s, 3H), 2.00-1.97 (m, 1H). Example 72. (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5,6-dichloro-2-methylpyrimidine To a stirred solution of 3-((tert-butyldimethylsilyl)oxy)azetidine (200.0 mg, 1.0 equiv) and 4,5,6-trichloro-2-methylpyrimidine (210.7 mg, 1.0 equiv) in THF (5.0 mL) were added TEA (324.0 mg, 3.0 equiv) dropwise at room temperature under air atmosphere. The resulting mixture was stirred for 2 h at room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with 0-50% EA in PE to afford the title compound (80.0 mg, 21.5%) as a white solid. m/z (ESI, +ve ion)=348.15 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimeth-ylsilyl)oxy]azetidin-1-yl}-5-chloro-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 4-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5,6-dichloro-2-methylpyrimidine (80.4 mg, 1.2 equiv) and tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100.0 mg, 1.0 equiv) in toluene (2.5 mL) were added Cs2CO3(126.0 mg, 2.0 equiv) and XantPhos (40.0 mg, 0.2 equiv) and Pd2(dba)3(70.0 mg, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×7 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EtOAc in PE to afford the title compound (100.0 mg, 62.5%) as a yellow solid. m/z (ESI, +ve ion)=832.40 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.12 (s, 1H), 7.82 (s, 1H), 7.80 (s, 1H), 7.04 (d, J=8.0 Hz, 1H), 6.70 (d, J=4.0 Hz, 1H), 6.68 (d, J=4.0 Hz, 1H), 5.59 (d, J=4.0 Hz, 1H), 4.71-4.68 (m, 1H), 4.61 (s, 2H), 4.20 (s, 2H), 3.55-3.50 (m, 1H), 3.38 (s, 3H), 2.39-2.36 (m, 1H), 2.31 (s, 3H), 2.14-2.11 (m, 1H), 1.70 (d, J=4.0 Hz, 18H), 0.93 (s, 9H), 0.11 (s, 6H). Step D. (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)-2-methylp-yrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloro-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (84.0 mg) in DCM (5.0 mL) was added TFA (1.5 mL) dropwise at room temperature. The resulting mixture was stirred for 28 h at room temperature. The resulting mixture was concentrated under vacuum. The crude product (50.0 mg) was purified by RP flash with the following conditions (column, silica gel; mobile phase. MeCN in water (5 mM NH4HCO3), 10/o to 50/o gradient in 10 min; detector, UV 254 nm) to afford Example 72 (33.0 mg, 63.4%) as a white solid. m/z (ESI, +ve ion)=518.25 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.53 (d, J=8.0 Hz, 1H), 7.43 (s, 1H), 6.93 (d, J=8.0 Hz, 1H), 6.84 (d, J=8.0 Hz, 1H), 6.64-6.61 (m, 1H), 5.65 (d, J=4.0 Hz 1H), 4.61-4.57 (m, 3H), 4.14-4.11 (m, 2H), 3.38 (m, 1H), 3.34 (s, 3H), 2.27-2.17 (m, 5H). Example 73. (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-5-methoxy-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-6-chloro-5-methoxy-2-methylpyrimidine To a stirred mixture of 4,6-dichloro-5-methoxy-2-methylpyrimidine (300 mg, 1.554 mmol, 1.00 equiv) and 3-((tert-butyldimethylsilyl)oxy)azetidine (436.78 mg, 2.331 mmol, 1.5 equiv) in THE (7.5 mL) was added TEA (471.80 mg, 4.662 mmol, 3 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 60° C. under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford the title compound (510 mg, 95.41%) as a white solid. m/z (ESI, +ve ion)=344.05 [M+H]+. Step B. tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl) oxy] azetidin-1-yl}-5-methoxy-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.154 mmol, 1.00 equiv) and 4-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-6-chloro-5-methoxy-2-methylpyrimidine (63.42 mg, 0.185 mmol, 1.2 equiv) in toluene (4 mL) were added CPhos (6.71 mg, 0.015 mmol, 0.1 equiv) and Pd2(dba)3·CHCl3(15.91 mg, 0.015 mmol, 0.1 equiv) and Cs2CO3(100.14 mg, 0.308 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford the title compound (85 mg, 59.45%) as a yellow solid. m/z (ESI, +ve ion)=828.70 [M+H]+. Step C. (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-5-methoxy-2-methylpyrimidin-4-yl] amino}-1H-indazol-6-yl 5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A mixture of tert-butyl(1R,2S)-2-[1-tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl) oxy]azetidin-1-yl}-5-methoxy-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (85 mg, 0.103 mmol, 1.00 equiv) and TFA (2 mL, 26.926 mmol, 262.31 equiv) in DCM (4 mL) was stirred for 16 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 30% B in 8 min, 30% B; wavelength: 254 nm; RT1(min): 7.8 to afford Example 73 (34.2 mg, 62.67%) as a white solid. m/z (ESI, +ve ion)=514.45 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.48 (s, 1H), 10.40 (s, 1H), 8.63 (s, 1H), 7.46 (d, J=8.4 Hz, 1H), 7.36 (s, 1H), 6.91-6.84 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58 (m, 1H), 5.71 (d, J=2.8 Hz, 1H), 5.60 (d, J=6.4 Hz, 1H), 4.53 (m, 1H), 4.31-4.23 (m, 2H), 4.09 (m, 1H), 3.82 (m, 2H), 3.58 (s, 3H), 3.18 (d, J=5.2 Hz, 3H), 2.31 (m, 1H), 2.04 (s, 3H), 1.98 (m, 1H). Example 74. (1R,2S)-2-{3-[(1,3-dimethyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2,5-dimethylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a mixture of 2,5-dimethylpyrazol-3-amine (120 mg, 1.080 mmol, 1.00 equiv), tBuXPhos Pd G3 (171.53 mg, 0.216 mmol, 0.2 equiv), tert-butyl(1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (68.18 mg, 0.1080 mmol, 1 equiv) and di-tert-butyl([2-[2,4,6-tris(propan-2-yl)phenyl]phenyl])phosphane (9.17 mg, 0.022 mmol, 0.2 equiv) in dioxane (6 mL) was added Cs2CO3(703.54 mg, 2.160 mmol, 2 equiv). The resulting mixture was stirred for 1 h at 90° C. then concentrated under reduced pressure. The residue was purified with silica gel chromatography, eluted with 8% MeOH in DCM to afford the title compound (50 mg, 7.53%) as a yellow solid. m/z (ESI+ve ion)=615.40 [M+H]+ Step B. (1R,2S)-2-{3-[(2,5-dimethylpyrazol-3-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one; trifluoroacetic acid To a mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2,5-dimethylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (30 mg, 0.049 mmol, 1.00 equiv) in DCM (0.8 mL, 12.584 mmol, 257.85 equiv) was added TFA (0.2 mL, 2.693 mmol, 55.17 equiv). The resulting mixture was stirred for 2 h at room temperature and then concentrated under reduced pressure. The crude product (30 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep Phenyl OBD Column, 19×250 mm, 5 μm; Mobile Phase A: Water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 28% B to 33% B in 10 min, 33% B; wavelength: 254 nm; RT1(min): 6 to afford Example 74 (9 mg, 34.89%) as an off-white solid. m/z (ESI, +ve ion)=415.05 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.68 (d, J=8.4 Hz, 1H), 7.40-7.39 (m, 1H), 6.93 (dd, J=8.5, 1.4 Hz, 1H), 6.85 (d, J=8.5 Hz, 1H), 6.63 (dd, J=8.5, 2.5 Hz, 1H), 5.60 (d, J=2.5 Hz, 1H), 4.85 (s, 2H), 3.86 (s, 3H), 3.34-3.4 (m, 1H), 3.32 (s, 3H), 2.32 (s, 3H), 2.25-2.17 (m, 2H). Example 75. (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(4-methoxy-2-methylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (90 mg, 0.173 mmol, 1.00 equiv) and 5-iodo-4-methoxy-1-methylpyrazole (45.27 mg, 0.190 mmol, 1.1 equiv) in toluene (2.0 mL) were added Cs2CO3(112.66 mg, 0.346 mmol, 2 equiv) and Pd-PEPPSI-IPentCl 2-methylpyridine (o-picoline (29.08 mg, 0.035 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (3×5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (rinsed with EA) to give the title compound (30 mg, 24.76%) as a light yellow oil. m/z (ESI+ve ion)=631.60 [M+H]+. Step B. (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-2-methylpyrazol-3-yl)amino]-1H-indazol-6-yl}-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(4-methoxy-2-methylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (30 mg, 0.048 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (5 mL) was stirred at 25° C. for 8 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 35% B in 8 min; wavelength: 254 nm; RT1 (min): 6.2. The product-containing fractions were concentrated in vacuo to give Example 75 (10.7 mg, 50.27%) as a white solid. m/z (ESI+ve ion)=431.15 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.35-7.28 (m, 3H), 6.85-6.79 (m, 2H), 6.64-6.62 (m, 1H), 5.59 (d, J=2.4 Hz, 1H), 3.71 (s, 3H), 3.67 (s, 3H), 3.31 (s, 4H), 2.12-2.14 (m, 2H). Example 76. (R,2S)-2-(3-{[5-chloro-2-cyclopropyl-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-2-cyclopropylpyrimidin-4-amine Into a 25 mL round-bottom flask were added 6-chloro-2-cyclopropylpyrimidin-4-amine (400 mg, 2.358 mmol, 1.00 equiv) and 3-[(tert-butyldimethylsilyl)oxy]azetidine (883.71 mg, 4.716 mmol, 2 equiv) at room temperature. The resulting mixture was stirred for 16 h at 100° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was diluted with DCM (3 mL). The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (260 mg, 34.40%) as an off-white solid. m/z (ESI, +ve ion)=321.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 5.32 (s, 2H), 5.07 (s, 1H), 4.73 (m, 1H), 4.27-4.19 (m, 2H), 3.85 (m, 2H), 1.99 (m, 1H), 1.13 (p, J=4.0 Hz, 2H), 1.00 (m, 2H), 0.92 (s, 9H), 0.10 (s, 6H). Step B. 6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloro-2-cyclopropylpyrimidin-4-amine A mixture of 6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-2-cyclopropylpyrimidin-4-amine (200 mg, 0.624 mmol, 1.00 equiv) and NCS (99.99 mg, 0.749 mmol, 1.2 equiv) in AcOH (10 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1) to afford the title compound (65 mg, 29.35%) as a white solid. m/z=355.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 6.45 (s, 2H), 4.64 (m, 1H), 4.38 (m, 2H), 3.84 (m, 2H), 1.73 (m, 1H), 0.87 (s, 9H), 0.81 (m, 4H), 0.06 (s, 6H). Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloro-2-cyclopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.127 mmol, 1.00 equiv) and 6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloro-2-cyclopropylpyrimidin-4-amine (53.96 mg, 0.152 mmol, 1.2 equiv) in toluene (4 mL) were added Xantphos Pd G4(12.19 mg, 0.013 mmol, 0.1 equiv) and XantPhos (7.33 mg, 0.013 mmol, 0.1 equiv) and Cs2CO3(82.56 mg, 0.254 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (8 mL) at room temperature. The resulting mixture was extracted with DCM (3×8 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (4/1) to afford the title compound (83 mg, 76.31%) as a white solid. m/z (ESI, +ve ion)=858.35 [M+H]+. Step D. (1R,2S)-2-(3-{[5-chloro-2-cyclopropyl-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloro-2-cyclopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (83 mg, 0.097 mmol, 1.00 equiv) and TFA (2 mL, 26.926 mmol, 278.51 equiv) in DCM (4 mL) was stirred for 16 h at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 45% B in 8 min, 45% B; wavelength: 254 nm; RT1(min): 7 to afford Example 76 (36.9 mg, 70.16%) as a white solid. m/z (ESI, +ve ion)=544.45 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.57 (s, 1H), 10.40 (s, 1H), 8.72 (s, 1H), 7.33-7.37 (m, 2H), 6.89 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.54-6.61 (m, 1H), 5.62 (m, 2H), 4.42-4.47 (m, 3H), 4.41 (m, 2H), 3.94 (m, 2H), 3.19 (m, 2H), 2.30 (s, 1H), 1.95-2.02 (m, 1H), 2.50-2.73 (m, 1H), 0.59 (d, J=16.0 Hz, 4H). Example 77. (1R,2S)-2-[3-({2-cyclopropyl-6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. (2R,6S)-4-(6-chloro-2-cyclopropyl-5-methoxypyrimidin-4-yl)-2,6-dimethylmorpholine The mixture of 4,6-dichloro-2-cyclopropyl-5-methoxypyrimidine (200 mg, 0.913 mmol, 1.00 equiv), (2R,6S)-2,6-dimethylmorpholine (105.15 mg, 0.913 mmol, 1 equiv) and TEA (184.76 mg, 1.826 mmol, 2 equiv) in THE (2 mL) was stirred at 60° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-50% of EA in PE to give the title compound (250 mg, 91.04%) as a colorless oil. m/z (ESI, +ve ion)=298.25 [M+H]+.1H NMR (400 MHz, Chloroform-d S 4.47 (d, J=13.2 Hz, 2H), 3.71-3.64 (m, 5H), 2.69-2.63 (m, 2H), 2.07-2.01 (m, 1H), 1.25 (s, 3H), 1.23 (s, 3H), 1.24-0.94 (m, 4H). Step B. 2-cyclopropyl-6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-amine To the mixture of (2R,6S)-4-(6-chloro-2-cyclopropyl-5-methoxypyrimidin-4-yl)-2,6-dimethylmorpholine (200 mg, 0.672 mmol, 1.00 equiv) and NH30.5 M in dioxane (4.03 mL, 2.016 mmol, 3 equiv) in dioxane (2.0 mL) were added sodium 2-methylpropan-2-olate (90.37 mg, 0.941 mmol, 1.4 equiv), Pd2(dba)3(12.30 mg, 0.013 mmol, 0.02 equiv) and t-BuBrettPhos (32.55 mg, 0.067 mmol, 0.1 equiv) under nitrogen atmosphere. The mixture was stirred at 80° C. for 12 h. The mixture was filtered and washed with EA (5 mL×3). The filtrate was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-100% of EA in PE to give the title compound (150 mg, 79.83%) as a light yellow solid. m/z (ESI, +ve ion)=279.10 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 4.89 (s, 2H), 4.31-4.27 (m, 2H), 3.72-3.66 (m, 2H), 3.62 (s, 3H), 2.62-2.56 (m, 2H), 1.93-1.87 (m, 1H), 1.24-1.22 (m, 6H), 1.01-0.87 (m, 4H). Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({2-cyclopropyl-6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 2-cyclopropyl-6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-amine (52.90 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The resulting mixture was filtered and washed with EA (5 mL×3). The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-50% EA in PE to give crude the title compound (70 mg, 50.88%) as a light yellow solid. m/z (ESI, +ve ion)=782.40 [M+H]+. Step D. (1R,2S)-2-[3-({2-cyclopropyl-6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({2-cyclopropyl-6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (55 mg, 0.070 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (3 mL) was stirred at 25° C. for 5 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 39% B to 55% B in 8 min; wavelength: 254 nm; RT1(min): 7.65. The product-containing fractions were concentrated in vacuo to give Example 77 (33.4 mg, 81.31%) as a white solid. m/z (ESI, +ve ion)=582.45 [M+H]+.1H NMR (400 MHz, DMSO-4) δ 12.50 (s, 1H), 10.42 (s, 1H), 8.72 (s, 1H), 7.43 (d, J=8.4 Hz, 1H), 7.36 (s, 1H), 6.88 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59-6.57 (m, 1H), 5.70 (d, J=2.4 Hz, 1H), 4.23 (d, J=12.8 Hz, 2H), 3.68-3.63 (m, 2H), 3.61 (s, 3H), 3.33 (s, 3H), 3.19 (t, J=8.0 Hz, 1H), 2.68 (s, 1H), 2.53 (s, 1H), 2.33-2.29 (m, 1H), 2.00-1.97 (m, 1H), 1.65-1.58 (m, 1H), 1.13 (d, J=6.4 Hz, 6H), 0.63-0.60 (m, 4H). Example 78. (1R,2S)-2-(3-((5-chloro-6-(1,1-dioxidothiomorpholino)-2-isopropylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 5-chloro-6-hydroxy-2-isopropyl-3H-pyrimidin-4-one To a stirred mixture of 2-methylpropanimidamide hydrochloride (3 g, 24.470 mmol, 1.00 equiv) and 1,3-dimethyl 2-chloropropanedioate (4.08 g, 24.470 mmol, 1 equiv) in MeOH (60 mL) was added sodium 2-methylpropan-2-olate (4.70 g, 48.940 mmol, 2 equiv) in portions at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 80° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The reaction was quenched by the addition of 2N HCl (aq., 20 mL) at room temperature. The resulting mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (45 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford the title compound (2 g, 43.33%) as an off-white solid. m/z (ESI, +ve ion)=189.00 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.40 (s, 2H), 2.72-2.89 (m, 1H), 1.20 (d, J=6.8 Hz, 6H). Step B. 4,5,6-trichloro-2-isopropylpyrimidine Into a 50 mL 3-necked round-bottom flask were added 5-chloro-6-hydroxy-2-isopropyl-3H-pyrimidin-4-one (200 mg, 1.060 mmol, 1.00 equiv) and POCl3(7 mL, 75.098 mmol, 70.82 equiv) at room temperature. The resulting mixture was stirred for 16 h at 100° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The reaction was quenched by the addition of sat. NaHCO3(aq., 50 mL) at 0° C. The resulting mixture was extracted with EtOAc (3×15 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound. m/z (ESI, +ve ion)=227.05 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 3.18 (m, 1H), 1.35 (d, J=6.8 Hz, 6H). Step C. 4-(5,6-dichloro-2-isopropylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione To a stirred mixture of 4,5,6-trichloro-2-isopropylpyrimidine (130 mg, 0.576 mmol, 1.00 equiv) and 1lambda6-thiomorpholine-1,1-dione (187.03 mg, 1.382 mmol, 2.4 equiv) in THF (3.25 mL, 40.115 mmol, 69.58 equiv) was added TEA (233.34 mg, 2.304 mmol, 4 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at room temperature under nitrogen atmosphere. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (150 mg, 80.25%) as a white solid. m/z (ESI, +ve ion)=323.95 [M+H]+. Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-chloro-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-2-isopropylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.154 mmol, 1.00 equiv) and 4-(5,6-dichloro-2-isopropylpyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (59.79 mg, 0.185 mmol, 1.2 equiv) in toluene (4 mL) were added CPhos (6.71 mg, 0.015 mmol, 0.1 equiv) and Pd2(dba)3·CHCl3(15.91 mg, 0.015 mmol, 0.1 equiv) and Cs2CO3(100.14 mg, 0.308 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The reaction was quenched by the addition of Water (8 mL) at room temperature. The resulting mixture was extracted with DCM (3×5 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (2/1) to afford the title compound (29 mg, 23.35%) as a brown solid. m/z (ESI, +ve ion)=808.20 [M+H]+. Step E. 4-[5-chloro-2-isopropyl-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′H-spiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidin-4-yl]-1lambda6-thiomorpholine-1,1-dione A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-chloro-6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-2-isopropylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (29 mg, 0.036 mmol, 1.00 equiv) and TFA (1.5 mL, 20.195 mmol, 562.91 equiv) in DCM (3 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: Column: Xselect CSH OBD Column 30×150 mm 5 um, n: Mobile Phase A: Water (0.1% FA), Mobile Phase B: MeOH-HPLC; Flow rate: 60 mL/min; Gradient: 55% B to 70% B in 8 min, 70% B: wavelength: 220 nm; RT1(min): 6.47 to afford Example 78 (14 mg, 63.79%) as a white solid. m/z (ESI, +ve ion)=608.15 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.50 (d, J=8.4 Hz, 1H), 7.44 (s, 1H), 6.81-6.92 (m, 2H), 6.62 (m, 1H), 5.62 (d, J=2.4 Hz, 1H), 4.09 (s, 4H), 3.29 (d, J=12.0 Hz, 8H), 2.72-2.67 (m, 1H), 2.25 (m, 1H), 2.18 (m, 1H), 1.06 (m, 6H). Example 79. (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methyl-1H-pyrazol-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(4-methoxy-1-methylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.192 mmol, 1.00 equiv) and 3-iodo-4-methoxy-1-methylpyrazole (45.72 mg, 0.192 mmol, 1.00 equiv) in toluene (2.5 mL) were added Cs2CO3(125.17 mg, 0.384 mmol, 2 equiv) and Pd-PEPPSI-IPentCl 2-methylpyridine (o-picoline (16.16 mg, 0.019 mmol, 0.1 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (5 mL×3). The filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (rinsed with EA) to give the title compound (40 mg, 26.41%) as a light-yellow oil. m/z (ESI, +ve ion)=631.55 [M+H]+. Step B. (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methylpyrazol-3-yl)amino]-1H-indazol-6-yl}-1′H-spiro[cyclopropane-1,3′-indol]-2′-one; trifluoroacetic acid The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(4-methoxy-1-methylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (35 mg, 0.055 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (3 mL) was stirred at 25° C. for 5 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: Xselect CSH OBD Column 30×150 mm 5 μm; Mobile Phase A: ACN, Mobile Phase B: Water (0.05% TFA); Flow rate: 60 mL/min; Gradient: 8% B to 25% B in 10 min; wavelength: 254 nm; RT1(min): 9.5. The product-containing fractions were concentrated in vacuo to give Example 79 (9.2 mg, 30.33%) as a white solid. m/z (ESI, +ve ion)=431.15 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.94-7.92 (m, 1H), 7.42 (d, J=23.2 Hz, 2H), 7.05 (d, J=8.4 Hz, 1H), 6.85 (d, J=8.4 Hz, 1H), 6.67-6.64 (m, 1H), 5.65 (d, J=2.0 Hz, 1H), 3.83 (d, J=7.2 Hz, 6H), 3.37 (s, 3H), 3.32 (s, 1H), 2.26-2.18 (m, 2H).19F NMR (376 MHz, MeOD) δ −77.05 (s, 3F). Example 80. (1R,2S)-2-{3-[(6-cyclopropyl-3-methoxypyrazin-2-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-cyclopropyl-3-methoxypyrazin-2-amine To a stirred solution of 6-chloro-3-methoxypyrazin-2-amine (200.0 mg, 1.253 mmol, 1.00 equiv) in toluene (4.0 mL) and water (0.4 mL) were added cyclopropyltrifluoro-lambda4-borane potassium (278.21 mg, 1.5 equiv) and bis(adamantan-1-yl) (butyl)phosphane (89.88 mg, 0.2 equiv) and Pd(OAc)2(28.14 mg, 0.1 equiv) and Cs2CO3(1225.12 mg, 3.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 5 h at 100° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of sat. NH4Cl (aq., 20 mL) at room temperature. The resulting mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (3×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% EA in PE to afford the title compound (150.0 mg, 72.45%) as a yellow solid. m/z (ESI, +ve ion)=166.05[M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.19 (s, 1H), 6.11 (s, 2H), 3.83 (s, 3H), 1.81-1.87 (m, 5.0 Hz, 1H), 0.82-0.68 (m, 4H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-cyclopropyl-3-methoxypyrazin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 6-cyclopropyl-3-methoxypyrazin-2-amine (26.0 mg, 0.157 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (99.39 mg, 1.0 equiv) in toluene (5.0 mL) were added Cs2C03 (102.56 mg, 0.314 mmol, 2.0 equiv) and XantPhos (18.21 mg, 0.031 mmol, 0.2 equiv) and Pd2(dba)3(28.82 mg, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2.0 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (4×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (80.0 mg, 76.01% yield) as a yellow solid. m/z (ESI, +ve ion)=669.45 [M+H]+.1H NMR (400 MHz, Chloroform-d) 8.13 (s, 1H), 7.90 (s, 1H), 7.82 (d, J=8.0 Hz, 1H), 7.54 (s, 1H), 7.06 (d, J=8.0 Hz, 1H), 6.72-6.66 (m, 1H), 5.58 (d, J=4.0 Hz, 1H), 4.07 (s, 3H), 3.50-3.45 (m, 1H), 3.38 (s, 3H), 2.38-2.35 (m, 1H), 2.15-2.13 (m, 1H), 1.51 (s, 18H), 0.89-0.79 (m, 4H). Step C. (1R,2S)-2-{3-[(6-cyclopropyl-3-methoxypyrazin-2-yl)amino]-1H-indazol-1-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one; trifluoroacetic acid To a stirred solution of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-[(6-cyclopropyl-3-methoxypyrazin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.0 mg) in DCM (5.0 mL) was added TFA (1.0 mL) dropwise at room temperature. The resulting mixture was stirred for 5.0 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (40 mg) was purified by Prep-HPLC with the following conditions: Column: XSelect CSH Prep C18 OBD Column, 19×250 mm, 5 μm; Mobile Phase A: Water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 43% B to 43% B in 10 min, 43% B; wavelength: 254 nm; RT1(min): 9 to afford Example 80 (27.8 mg, 48.9%) as a yellow solid. m/z (ESI, +ve ion)=469.40 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.76 (d, J=8.0 Hz, 1H), 7.52 (s, 1H), 7.46 (s, 1H), 6.98 (d, J=8.6 Hz, 1H), 6.85 (d, J=8.4 Hz, 1H), 6.63 (d, J=8.0 Hz, 1H), 5.63 (d, J=4.0 Hz, 1H), 4.12 (s, 3H), 3.32 (s, 4H), 2.26 (d, J=8.0 Hz, 1H), 2.20 (d, J=8.0 Hz, 1H), 1.99 (s, 1H), 0.91-0.82 (m, 2H), 0.80-0.77 (m, 2H).19F NMR (376 MHz, Methanol-d4) δ −77.42 Example 81. (1R,2S)-2-(3-{[2-cyclopropyl-6-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-6-chloro-2-cyclopropyl-5-methoxypyrimidine The mixture of 4,6-dichloro-2-cyclopropyl-5-methoxypyrimidine (200 mg, 0.913 mmol, 1.00 equiv), 3-((tert-butyldimethylsilyl)oxy)azetidine (205.26 mg, 1.096 mmol, 1.2 equiv) and TEA (184.76 mg, 1.826 mmol, 2 equiv) in THF (2 mL, 24.686 mmol, 27.04 equiv) was stirred at 60° C. for 8 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-50% EA in PE to give the title compound (300 mg, 84.38%) as a light yellow solid. m/z (ESI+ve ion)=370.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 4.75-4.70 (m, 1H), 4.44-4.40 (m, 2H), 4.06-4.02 (m, 2H), 3.72 (s, 3H), 2.03 (s, 1H), 1.05-1.02 (m, 2H), 0.92 (s, 11H), 0.10 (s, 6H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-2-cyclopropyl-5-methoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.192 mmol, 1.00 equiv) and 4-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-6-chloro-2-cyclopropyl-5-methoxypyrimidine (92.39 mg, 0.250 mmol, 1.3 equiv) in dry toluene (2.5 mL) were added Cs2CO3(125.17 mg, 0.384 mmol, 2 equiv), CPhos (16.77 mg, 0.038 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(39.77 mg, 0.038 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (5 mL×3). The filtrate was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-50% of EA in PE to give crude the title compound (70 mg, 36.27%) as a yellow solid. m/z (ESI+ve ion)=854.40 [M+H]+. Step C. (1R,2S)-2-(3-{[2-cyclopropyl-6-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one; trifluoroacetic acid The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-2-cyclopropyl-5-methoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70 mg, 0.082 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (3 mL) was stirred at 25° C. for 8 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: Xselect CSH OBD Column 30×150 mm 5 μm, Mobile Phase A: ACN, Mobile Phase B: Water (0.05% TFA); Flow rate: 60 mL/min; Gradient: 10% B to 35% B in 8 min; wavelength: 254 nm; RT1(min): 7. The product-containing fractions were collected and concentrated in vacuo to give Example 81 (46.1 mg, 86.06%) as a white solid. m/z (ESI +ve ion)=540.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.82 (s, 1H), 10.44 (s, 1H), 9.86 (s, 1H), 7.60 (s, 1H), 7.44 (s, 1H), 6.96 (d, J=8.4 Hz, 1H), 6.76 (d, J=8.4 Hz, 1H), 6.61-6.58 (m, 1H), 5.72 (m, 1H), 4.62-4.59 (m, 1H), 4.48 (s, 2H), 4.02 (s, 2H), 3.65 (s, 3H), 3.34 (s, 3H), 3.20 (t, J=8.4 Hz, 1H), 2.35-2.32 (m, 1H), 2.00-1.98 (m, 2H), 0.89-0.80 (m, 4H).19F NMR (376 MHz, DMSO-d6) δ −74.10 (s, 3F). Example 82. (1R,2S)-2-{3-[(3,6-dimethylpyrazin-2-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3,6-dimethylpyrazin-2-yl)amino] indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 3,6-dimethylpyrazin-2-amine (20.0 mg, 0.162 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (102.55 mg, 1.0 equiv) in toluene (5.0 mL) were added Cs2CO3(105.82 mg, 0.324 mmol, 2.0 equiv) and CPhos (14.18 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(33.62 mg, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2.0 h at 90.0° C. under nitrogen atmosphere. The mixture was cooled down to room temperature and then filtered, the filter cake was washed with EtOAc (4×5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford the title compound (40.0 mg, 39.3% yield) as a yellow solid. m/z (ESI, +ve ion)=627.30 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3,6-dimethylpyrazin-2-yl)amino] indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate; trifluoroacetic acid To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3,6-dimethylpyrazin-2-yl)amino] indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (50.0 mg) in DCM (5.0 mL) was added TFA (1.0 mL) dropwise at room temperature. The resulting mixture was stirred for 4 h at room temperature. The reaction was monitored by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product (30.0 mg) was purified by Prep-HPLC with the following conditions: (Column: Xselect CSH C18 OBD Column 30×150 mm 5 μm, Mobile Phase A: ACN, Mobile Phase B: Water (0.05% TFA); Flow rate: 60 mL/min; Gradient: 6% B to 30% B in 8 min. 30% B to 30% B in 11 min. 30% B; wavelength: 254 nm; RT1(min): 9.0-10.4 to afford Example 82 (14.5 mg, 41.4% yield) as a yellow solid. m/z (ESI, +ve ion)=427.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.85 (s, 1H), 7.61 (d, J=8.5 Hz, 1H), 7.48 (s, 1H), 6.95 (d, J=8.0 Hz, 1H), 6.85 (d, J=8.4 Hz, 1H), 6.63 (dd, J=8.5, 2.6 Hz, 1H), 5.62 (d, J=2.5 Hz, 1H), 3.42-3.33 (m, 4H), 2.64 (s, 3H), 2.35 (s, 3H), 2.27-2.15 (m, 2H). Example 83. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 3-methoxy-6-(prop-1-en-2-yl)pyrazin-2-amine To a stirred mixture of 6-chloro-3-methoxypyrazin-2-amine (400 mg, 2.507 mmol, 1.00 equiv) and 4,4,5,5-tetramethyl-2-(prop-1-en-2-yl)-1,3,2-dioxaborolane (842.47 mg, 5.014 mmol, 2 equiv) in dioxane (20 mL) and water (10 mL) were added Pd(dppf)Cl2(204.20 mg, 0.251 mmol, 0.1 equiv) and Na2CO3(797.06 mg, 7.521 mmol, 3 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 110° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (163 mg, 39.36%) as a white solid. m/z (ESI, +ve ion)=166.20 [M+H]+. Step B. 6-isopropyl-3-methoxypyrazin-2-amine A mixture of 3-methoxy-6-(prop-1-en-2-yl)pyrazin-2-amine (132 mg, 0.799 mmol, 1.00 equiv) and Pd(OH)2/C (112.21 mg, 0.799 mmol, 1 equiv) in EtOH (6.5 mL) was stirred for 16 h at room temperature under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOH (2×5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (2/1) to afford the title compound (74 mg, 55.38%) as a colorless oil. m/z (ESI+ve ion)=168.00 [M+H]+.1H NMR (400 MHz, chloroform-d) δ 7.30 (s, 1H), 4.80 (s, 2H), 3.98 (s, 3H), 2.85 (m, 1H), 1.26 (d, J=6.8 Hz, 6H). Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-isopropyl-3-methoxypyrazin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.127 mmol, 1.00 equiv) and 6-isopropyl-3-methoxypyrazin-2-amine (25.42 mg, 0.152 mmol, 1.2 equiv) in toluene (4 mL) were added Pd2(dba)3(11.60 mg, 0.013 mmol, 0.1 equiv), XantPhos (7.33 mg, 0.013 mmol, 0.1 equiv) and Cs2CO3(82.56 mg, 0.254 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with DCM (3×8 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (2/1) to afford the title compound (84.7 mg, 99.67%) as a brown yellow solid. m/z (ESI, +ve ion)=671.45 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.16 (s, 1H), 7.93 (d, J=8.4 Hz, 1H), 7.82 (d, J=8.9 Hz, 1H), 7.64 (s, 1H), 7.47 (s, 1H), 7.01 (d, J=8.2 Hz, 1H), 6.69 (m, 1H), 5.57 (d, J=2.8 Hz, 1H), 4.06 (s, 3H), 3.56-3.54 (m, 1H), 3.36 (s, 3H), 2.87-2.84 (m, 1H), 1.71 (d, J=6.6 Hz, 18H), 1.28 (t, J=7.2 Hz, 2H), 1.15 (t, J=6.4 Hz, 6H). Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-isopropyl-3-methoxypyrazin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-isopropyl-3-methoxypyrazin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (84.7 mg, 0.126 mmol, 1.00 equiv) and TFA (2 mL, 26.926 mmol, 213.24 equiv) in DCM (4 mL, 62.920 mmol, 498.29 equiv) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 60% Vo B in 8 min, 60% B; wavelength: 254 nm; RT1(min): 7 to afford Example 83 (33.6 mg, 56.49%) as a white solid. m/z=471.35 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.48 (s, 1H), 10.38 (s, 1H), 8.69 (s, 1H), 7.45 (d, J=8.4 Hz, 1H), 7.35 (d, J=4.4 Hz, 2H), 6.83 (d, J=8.5 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58 (m, 1H), 5.67 (d, J=2.4 Hz, 1H), 3.96 (s, 3H), 3.31 (s, 2H), 3.15-3.22 (m, 2H), 2.70-2.65 (m, 1H), 2.29 (m, 1H), 1.98 (m, 1H), 1.00 (m, 6H). Example 84. (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-2-isopropyl-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 6-hydroxy-2-isopropyl-5-methoxy-3H-pyrimidin-4-one To a stirred solution of Sodium t-butoxide (1.96 g, 2.5 equiv) in MeOH (10.0 mL) were added 2-methylpropanimidamide hydrochloride (1.0 g, 1.0 equiv) and 1,3-dimethyl 2-methoxypropanedioate (1.32 g, 1.0 equiv) at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 12 h at 80° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered. The filtrate was concentrated under reduced pressure to afford the title compound (1.0 g, 66.56%) as a yellow solid. m/z (ESI, +ve ion)=185.00 [M+H]+. Step B. 4,6-dichloro-2-isopropyl-5-methoxypyrimidine The mixture of crude 6-hydroxy-2-isopropyl-5-methoxy-3H-pyrimidin-4-one (1 g, 5.429 mmol, 1.00 equiv) in POCl3(10.00 mL) was stirred for 3 h at 100° C. After cooled to room temperature, the mixture solution was added dropwise to cooled sat·aq·NaHCO3(150 mL). The mixture was extracted with EA (50 mL×3). The combined organic layer was washed with brine (30 mL), dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-30% EA in PE to give the title compound (600 mg, 47.49%) as a colorless oil. m/z (ESI, +ve ion)=220.95 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 3.97 (s, 3H), 3.22-3.11 (m, 1H), 1.35 (s, 3H), 1.33 (s, 3H). Step C. 4-(6-chloro-2-isopropyl-5-methoxypyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione The mixture of 4,6-dichloro-2-isopropyl-5-methoxypyrimidine (200 mg, 0.905 mmol, 1.00 equiv) and 1lambda6-thiomorpholine-1,1-dione (122.29 mg, 0.905 mmol, 1 equiv) and TEA (183.08 mg, 1.810 mmol, 2 equiv) in THF (2 mL, 24.686 mmol, 27.29 equiv) was stirred at 60° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-50% of EA in PE to give the title compound (150 mg, 51.85%) as a white solid. m/z (ESI, +ve ion)=320.05 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 4.36-4.34 (m, 4H), 3.78 (s, 3H), 3.15-3.12 (m, 4H), 3.06-2.99 (m, 1H), 1.29 (s, 3H), 1.28 (s, 3H). Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-2-isopropyl-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (90 mg, 0.173 mmol, 1.00 equiv) and 4-(6-chloro-2-isopropyl-5-methoxypyrimidin-4-yl)-1lambda6-thiomorpholine-1,1dione (66.35 mg, 0.208 mmol, 1.2 equiv) in dry toluene (2.5 mL) were added Cs2CO3(112.66 mg, 0.346 mmol, 2 equiv), CPhos (15.10 mg, 0.035 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(35.79 mg, 0.035 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (3×5 mL). The filtrate was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-100% of EA in PE to give the title compound (70 mg, 45.33%) as a yellow solid. m/z (ESI, +ve ion)=804.30 [M+H]+. Step E The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[6-(1,1-dioxo-1lambda6-thiomorpholin-4-yl)-2-isopropyl-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70 mg, 0.087 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (5 mL) was stirred at 25° C. for 5 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 33% B to 43% B in 8 min; wavelength: 254 nm; RT1(min): 7.45. The product-containing fractions was collected and concentrated in vacuo to give Example 84 (31.9 mg, 60.26%) as a white solid. m/z (ESI, +ve ion)=604.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.52 (s, 1H), 10.41 (s, 1H), 8.93 (s, 1H), 7.46 (d, J=8.4 Hz, 1H), 7.36 (s, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.59-6.57 (m, 1H), 5.67 (s, 1H), 4.09 (s, 4H), 3.65 (s, 3H), 3.32 (s, 3H), 3.24-3.21 (m, 5H), 2.61-2.56 (m, 1H), 2.32-2.28 (m, 1H), 1.99-1.96 (m, 1H), 1.00 (d, J=6.8 Hz, 6H). Example 85. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(morpholin-4-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-(6-chloro-2-isopropyl-5-methoxypyrimidin-4-yl)morpholine The mixture of 4-(6-chloro-2-isopropyl-5-methoxypyrimidin-4-yl)-1lambda6-thiomorpholine-1,1-dione (100 mg, 0.452 mmol, 1.00 equiv), morpholine (39.41 mg, 0.452 mmol, 1 equiv) and TEA (91.54 mg, 0.904 mmol, 2 equiv) in THF (2 mL) was stirred at 60° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-50% of EA in PE to give the title compound (100 mg, 81.35%) as a white solid. m/z (ESI+ve ion)=272.00 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 4.36-4.34 (m, 4H), 3.78 (s, 3H), 3.15-3.12 (m, 4H), 3.06-3.00 (m, 1H), 1.30 (s, 3H), 1.28 (s, 3H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-isopropyl-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.154 mmol, 1.00 equiv) and 56a (50.11 mg, 0.185 mmol, 1.2 equiv) in dry toluene (2.0 mL) were added Cs2CO3(100.14 mg, 0.308 mmol, 2 equiv), CPhos (13.42 mg, 0.031 mmol, 0.2 equiv) and Pd2(dba)3·CHCl3(31.81 mg, 0.031 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (5 mL×3). The filtrate was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-100% of EA in PE to give crude the title compound (56b) (70 mg, 48.21%) as a light yellow solid. m/z (ESI+ve ion)=756.35 [M+H]+. Step C. (1R,2S)-2-(3-{[2-isopropyl-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of 4-(6-chloro-2-isopropyl-5-methoxypyrimidin-4-yl)morpholine (70 mg, 0.093 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (5 mL) was stirred at 25° C. for 5 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 38% B to 48% B in 8 min; wavelength: 254 nm; RT1(min): 7.33. The product-containing fractions were collected and concentrated in vacuo to give Example 85 (30.7 mg, 59.42%) as a white solid. m/z (ESI+ve ion)=556.30 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.48 (s, 1H), 10.41 (s, 1H), 8.78 (s, 1H), 7.46 (d, J=8.4 Hz, 1H), 7.35 (s, 1H), 6.84 (d, J=8.8 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.68 (d, J=2.4 Hz, 1H), 3.73-3.71 (m, 4H), 3.64 (s, 3H), 3.61-3.58 (m, 4H), 3.32 (s, 3H), 3.18 (t, J=8.0 Hz, 1H), 2.60-2.55 (m, 1H), 2.31-2.28 (m, 1H), 1.99-1.96 (m, 1H), 0.98 (d, J=6.8 Hz, 6H). Example 86. (1R,2S)-5′-methoxy-2-{3-[(5-methoxy-2-methylpyridin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-methoxy-2-methylpyridin-4-yl) amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 4-bromo-5-methoxy-2-methylpyridine (38.81 mg, 0.192 mmol, 1.0 equiv) and tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3-indole]-1′-carboxylate (100.0 mg, 1.0 equiv) in toluene (5.0 mL) were added Cs2C03 (125.17 mg, 0.384 mmol, 2.0 equiv) and XantPhos (22.23 mg, 0.038 mmol, 0.2 equiv) and Pd2(dba)3(35.18 mg, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×6 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (9/1) to afford the title compound (80.0 mg, 64.90%) as a yellow solid. m/z (ESI, +ve ion)=642.40 [M+H]+. Step B. (1R,2S)-5′-methoxy-2-{3-[(5-methoxy-2-methylpyridin-4-yl)amino]-1H-indazol-6-yl}-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-methoxy-2-methylpyridin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60.0 mg) in DCM (5.0 mL) was added TFA (1.0 mL) dropwise at room temperature. The resulting mixture was stirred for 5 h at room temperature then concentrated under reduced pressure. The crude product (30 mg) was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 45% B in 8 min, 45% B; wavelength: 254 nm; RT1(min): 6.12 to afford Example 86 (19.2 mg, 46.51%) as a white solid. m/z (ESI, +ve ion)=442.25 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.90 (s, 1H), 7.66 (d, J=8.0 Hz, 1H), 7.51 (s, 1H), 7.43 (s, 1H), 6.95 (d, J=8.0 Hz, 1H), 6.84 (d, J=8.0 Hz, 1H), 6.63 (d, J=8.0 Hz, 1H), 5.61 (d, J=4.0 Hz, 1H), 4.03 (s, 3H), 3.31 (s, 4H), 2.37 (s, 3H), 2.37-2.27 (m, 1H), 2.25-2.19 (m, 1H). Example 87. (1R,2S)-5′-methoxy-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-chloro-5-methoxy-2-methyl yrimidin-4-amine A mixture of 4,6-dichloro-5-methoxy-2-methylpyrimidine (500 mg, 2.590 mmol, 1.00 equiv) and 30% NH3·H2O (8 mL) in THF (4 mL) was stirred for 16 h at 70° C. under nitrogen atmosphere. The reaction was monitored by LCMS. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (288 mg, 64.05%) as a white solid. m/z (ESI, +ve ion)=174.00 [M+H]+. Step B. 5-methoxy-2-methylpyrimidin-4-amine A mixture of 6-chloro-5-methoxy-2-methylpyrimidin-4-amine (250 mg, 1.440 mmol, 1.00 equiv) and Pd/C (229.88 mg, 2.160 mmol, 1.5 equiv) in MeOH (8.5 mL, 209.941 mmol, 145.78 equiv) was stirred for 16 h at room temperature under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with MeOH (3×5 mL). The filtrate was concentrated under reduced pressure to give the title compound (158 mg, 78.84%) as a white solid. m/z (ESI, +ve ion)=140.05 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.93 (s, 1H), 8.43 (s, 1H), 7.92 (s, 1H), 3.89 (s, 3H), 2.47 (s, 3H). Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.127 mmol, 1.00 equiv) and 5-methoxy-2-methylpyrimidin-4-amine (21.16 mg, 0.152 mmol, 1.2 equiv) in toluene (4 mL) were added Pd2(dba)3(11.60 mg, 0.013 mmol, 0.1 equiv), XantPhos (7.33 mg, 0.013 mmol, 0.1 equiv) and Cs2CO2(82.56 mg, 0.254 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The reaction was quenched by the addition of Water (5 mL) at room temperature. The resulting mixture was extracted with DCM (3×5 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (80 mg, 98.25%) as a brown yellow solid. m/z (ESI, +ve ion)=643.20 [M+H]+. 1H NMR (400 MHz, Chloroform-d) δ 8.16 (s, 1H), 7.91-8.00 (m, 2H), 7.82 (d, J=8.8 Hz, 1H), 7.08 (d, J=8.4 Hz, 1H), 6.70 (m, 1H), 5.61 (d, J=2.4 Hz, 1H), 5.32 (s, 1H), 4.00 (s, 3H), 3.56-3.52 (m, 1H), 3.39 (s, 3H), 2.52 (s, 3H), 2.41-2.37 (m, 1H), 2.15-2.12 (m, 1H), 1.71 (d, J=5.6 Hz, 18H). Step D. (1R,2S)-5′-methoxy-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (87.5 mg, 0.136 mmol, 1.00 equiv) and TFA (2 mL, 26.926 mmol, 197.78 equiv) in DCM (4 mL, 62.920 mmol, 462.17 equiv) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 40% B in 8 min. 40% B: wavelength: 254 nm; RT1(min): 6.4 to afford Example 87 (23.3 mg, 38.60%) as a white solid. m/z (ESI, +ve ion)=443.30 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.62 (s, 1H), 10.42 (s, 1H), 8.98 (s, 1H), 7.92 (s, 1H), 7.45 (d, J=8.4 Hz, 1H), 7.39 (s, 1H), 6.90 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (m, 1H), 5.71 (d, J=2.4 Hz, 1H), 3.90 (s, 3H), 3.32 (s, 3H), 3.19 (t, J=8.4 Hz, 1H), 2.32 (m, 1H), 2.18 (s, 3H), 1.99 (m, 1H). Example 88. (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyridin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-methoxy-6-methylpyridin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 3-methoxy-6-methylpyridin-2-amine (24 mg, 1.3 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 1 equiv) in 1,4-dioxane (4 mL) and Cs2CO3(80 mg, 2 equiv) were added XantPhos (24 mg, 0.2 equiv) and Pd2(dba)3(16 mg, 0.2 equiv) at 25° C. under nitrogen atmosphere. The mixture was warmed up to 90° C. and stirred for 2 h. The mixture was concentrated under reduced pressure. The crude product 200 mg was purified by Prep-HPLC with PE:EA=1:1 to afford the title compound (59 mg, 70%) as an off-white solid. m/z (ESI+ve ion)=642.25 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.41 (s, 1H), 8.10 (s, 1H), 7.81 (d, J=8.9 Hz, 1H), 7.08 (dd, J=14.3, 8.2 Hz, 2H), 6.76-6.65 (m, 2H), 5.61 (d, J=2.6 Hz, 1H), 4.15 (q, J=7.1 Hz, 1H), 3.97 (s, 3H), 3.37 (s, 3H), 2.44-2.34 (m, 3H), 1.86-1.70 (m, 18H), 1.58-1.50 (m, 1H), 0.92-0.86 (m, 1H). Step B. (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyridin-2-yl)amino]-1H-indazol-6-yl}-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-methoxy-6-methylpyridin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (59 mg, 1 equiv) in DCM (5 mL) was added TFA (1 mL) at 25° C. The solution was stirred for 2 hours. The mixture was concentrated under reduced pressure. The residue was purified by C18 silica gel; mobile phase, MeCN in water (5 mM NH4HCO), 30% to 40/o gradient in 10 min; detector, UV 254 nm to afford Example 88 (20 mg, 49%) as an off-white solid. m/z (ESI+ve ion)=442.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.35 (s, 1H), 10.41 (s, 1H), 8.04 (s, 1H), 7.55 (d, J=8.4 Hz, 1H), 7.33 (s, 1H), 7.10 (d, J=7.9 Hz, 1H), 6.89-6.82 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.62-6.53 (m, 2H), 5.73 (d, J=2.5 Hz, 1H), 3.84 (s, 3H), 3.33 (s, 3H), 3.18 (t, J=8.4 Hz, 1H), 2.31 (dd, J=7.9, 4.7 Hz, 1H), 2.12 (s, 3H), 1.98 (dd, J=9.0, 4.7 Hz, 1H). Example 89. (1R,2S)-5′-methoxy-2-{3-[(2-methoxy-5-methylpyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-methoxy-5-methylpyridin-3-amine To a solution of 2-methoxy-5-methyl-3-nitropyridine (100.0 mg, 0.595 mmol, 1.00 equiv) in EtOH (4.0 mL) was added Pd/C (10%, 50.0 mg) under nitrogen atmosphere in a 50 mL round-bottom flask. The mixture was hydrogenated at room temperature for overnight under hydrogen atmosphere using a hydrogen balloon, filtered through a Celite pad, and concentrated under reduced pressure to afford the title compound (70.0 mg, 85.19%) as a white solid. m/z (ESI, +ve ion)=139.05 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.41 (d, J=4.0 Hz, 1H), 6.77 (d, J=4.0 Hz, 1H), 4.00 (s, 3H), 2.20 (s, 3H). Step B tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-methoxy-5-methylpyridin-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 2-methoxy-5-methylpyridin-3-amine (17.50 mg, 0.127 mmol, 1.0 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.0 mg, 1.0 equiv) in toluene (4.0 mL) were added Cs2CO3(82.56 mg, 0.254 mmol, 2.0 equiv) and XantPhos (14.66 mg, 0.025 mmol, 0.2 equiv) and Pd2(dba)3(23.2 mg, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×10 mL). The filtrate was concentrated under reduced pressure. The crude product was purified by prep-TLC (EA/PE=1/1) to afford the title compound (45.0 mg, 55.35%) as a yellow solid. m/z (ESI, +ve ion)=642.60 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.73 (s, 1H), 8.14 (s, 1H), 7.82 (d, J=8.0 Hz, 1H), 7.63 (s, 1H), 7.59 (d, J=8.0 Hz, 1H), 7.09 (s, 1H), 6.69 (d, J=8.0 Hz, 1H), 5.57 (s, 1H), 4.18 (s, 3H), 3.54 (d, J=8.0 Hz, 1H), 3.39 (s, 3H), 2.42-2.33 (m, 4H), 2.15-2.12 (m, 1H), 1.73 (s, 9H), 1.71 (s, 9H). Step C. (1R,2S)-5′-methoxy-2-{3-[(2-methoxy-5-methylpyridin-3-yl)amino]-1H-indazol-6-yl}-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-methoxy-5-methylpyridin-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60.0 mg) in DCM (5.0 mL) was added TFA (1.0 mL) dropwise at mom temperature. The resulting mixture was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (30.0 mg) was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmoL/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 35% B to 45% B in 8 min, 45% B; wavelength: 254 nm) to afford Example 89 (26.6 mg, 64.44%) as a white solid. m-z (ESI, +ve ion)=442.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.12 (s, 1H), 10.40 (s, 1H), 8.23 (d, J=4.0 Hz, 1H), 7.89 (d, J=8.0 Hz, 2H), 7.43 (d, J=1.9 Hz, 1H), 7.33 (s, 1H), 6.89 (d, J=8.0 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (d, J=8.0, 2.6 Hz, 1H), 5.70 (d, J=2.6 Hz, 1H), 3.96 (s, 3H), 3.33 (s, 3H), 3.18 (d, J=8.0 Hz, 1H), 2.34-2.28 (m, 1H), 2.21 (s, 3H), 1.98 (d, J=8.0 Hz, 1H). Example 90. (1R,2S)-5′-methoxy-2-{3-[(4-methoxypyridazin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 90 was prepared using similar procedures as other examples, but using a 4-methoxypyridazinyl starting material. Example 91. (1R,2S)-2-{3-[(3-cyclopropyl-1-methyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-cyclopropyl-2-methylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 5-cyclopropyl-2-methylpyrazol-3-amine (17.38 mg, 0.127 mmol, 1.0 equiv) and tert-butyl (R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.0 mg, 0.127 mmol, 1.00 equiv) in toluene (4.0 mL) were added Pd2(dba), (23.20 mg, 0.025 mmol, 0.2 equiv) and XantPhos (14.66 mg, 0.025 mmol, 0.2 equiv) and Cs2CO3(82.56 mg, 0.254 mmol, 2.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×9 mL). The filtrate was concentrated under reduced pressure. The crude product was purified by prep-TLC (EA/PE=1/1) to afford the title compound (50.0 mg, 61.60%) as a yellow solid. m/z (ESI, +ve ion)=641.25 [M+H]+. Step B. (1R,2S)-2-{3-[(5-cyclopropyl-2-methylpyrazol-3-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-[(5-cyclopropyl-2-methylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (50.0 mg, 0.078 mmol, 1.00 equiv) in DCM (5.0 mL) was added TFA (0.5 mL) dropwise at room temperature. The resulting mixture was stirred for 2 h at room temperature. The resulting mixture was concentrated under vacuum. The crude product (30 mg) was purified by Prep-HPLC with the following conditions (Column: Atlantis HILIC OBD Column, 19×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 26% B to 34% B in 8 min, 34% B; wavelength: 254 nm; RT1(min): 6.95 to afford Example 91 (18.7 mg, 54.40%) as a white solid. m/z (ESI, +ve ion)=441.20 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.55 (d, J=8.0 Hz, 1H), 7.32 (s, 1H), 6.81-6.89 (m, 2H), 6.63 (d, J=8.0, 4.0 Hz, 1H), 5.81 (s, 1H), 5.60 (d, J=4.0 Hz, 1H), 3.69 (s, 3H), 3.31 (s, 4H), 2.2-2.25 (m, 1H), 2.17 (d, J=8.0 Hz, 1H), 1.84 (d, J=8.0 Hz, 1H), 0.84-0.92 (m, 2H), 0.68 (s, 2H). Example 92. (1R,2S)-2-{3-[(3-cyclopropyl-1-ethyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-cyclopropyl-2-ethylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 5-cyclopropyl-2-ethylpyrazol-3-amine (16.76 mg, 0.111 mmol, 1.0 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70.0 mg, 0.111 mmol, 1.00 equiv) in toluene (3.0 mL) were added Pd(dba), (10.15 mg, 0.011 mmol, 0.1 equiv) and XantPhos (6.41 mg, 0.011 mmol, 0.1 equiv) and Cs2CO3(72.24 mg, 0.222 mmol, 2.0 equiv) at mom temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×8 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (40.0 mg, 55.11%) as a yellow solid. m/z (ESI, +ve ion)=655.55 [M+H]+. Step B. (1R,2S)-2-{3-[(5-cyclopropyl-2-ethylpyrazol-3-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-cyclopropyl-2-ethylpyrazol-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (35.0 mg, 0.053 mmol, 1.0) equiv) in DCM (5.0 mL) was added TFA (0.5 mL, 6.732 mmol, 125.93 equiv) dropwise at room temperature. The resulting mixture was stirred for 4 h at room temperature. The resulting mixture was concentrated under vacuum. The crude product (20.0 mg) was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 50% B in 8 min, 50% B; wavelength: 254 nm; RT1(min): 6 to afford Example 92 (9.1 mg, 37.45%) as a pink solid. m/z (ESI, +ve ion)=455.15 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.52 (d, J=8.0 Hz, 1H), 7.32 (s, 1H), 6.84 (d, J=8.0 Hz, 2H), 6.63 (d, J=8.0 Hz, 1H), 5.76 (s, 1H), 5.59 (d, J=4.0 Hz, 1H), 4.10-4.04 (m, 2H), 3.31 (s, 4H), 2.20 (d, J=8.0, Hz, 2H), 1.88-1.84 (t, J=8.0 Hz, 1H), 1.36 (t, J=4.0 Hz, 3H), 0.91-0.87 (m, 2H), 0.63-0.71 (m, 2H), 1.90-1.82 (m, 1H). Example 93. (1R,2S)-2-(3-{[2-(2-hydroxy-2-methylpropyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 1-[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]-2-methylpropan-2-ol To a stirred solution of methyl 2-[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]acetate (150.0 mg, 0.497 mmol, 1.00 equiv) in THF (5.0 mL) were added MeMgBr (2.4 mg, 2.485 mmol, 5.0 equiv, 1M in THF) and LaCl3·2LiCl (0.5 mL, 3 mmol, 0.6 M in THF) dropwise at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was quenched by the addition of sat. NH4Cl (aq., 20 mL) at room temperature. The aqueous layer was extracted with EtOAc (3×8 mL). The combined organic layer was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (100.0 mg, 66.66%) as a white oil. m/z (ESI, +ve ion)=315.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 3.65-3.76 (m, 11H), 2.72 (s, 2H), 1.16 (s, 6H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(2-hydroxy-2-methylpropyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 1-[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]-2-methylpropan-2-el (57.97 mg, 0.192 mmol, 1.0 equiv) and tert-butyl (1R,2S)-2-[3-amino-1-(tert-butoxycarbonyl)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100.0 mg, 1.0 equiv) in toluene (5.0 mL) were added Cs2CO3(125.17 mg, 0.384 mmol, 2.0 equiv) and XantPhos (44.46 mg, 0.077 mmol, 0.4 equiv) and Pd2(dba)3(70.36 mg, 0.4 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 100° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered; the filter cake was washed with EtOAc (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/2) to afford the title compound (30.0 mg, 20.00% yield) as a yellow solid. m/z (ESI, +ve ion)=786.40 [M+H]+. Step C. (1R,2S)-2-(3-{[2-(2-hydroxy-2-methylpropyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-{[2-(2-hydroxy-2-methylpropyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (30.0 mg) in DCM (5.0 mL) was added TFA (0.5 mL) dropwise at room temperature under air atmosphere. The resulting mixture was stirred for 2 h at room temperature under air atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product (25 mg) was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 28% B to 38% B in 8 min, 38% B: wavelength: 254 nm to afford Example 93 (13.8 mg, 61.54%) as a white solid. m/z (ESI, +ve ion)=586.50 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.59 (d, J=8.0 Hz, 1H), 7.44 (s, 1H), 6.91 (d, J=8.0 Hz, 1H), 6.84 (d, J=8.0 Hz, 1H), 6.63 (d, J=8.0 Hz, 1H), 5.63 (d, J=4.0 Hz, 1H), 3.77-3.86 (m, 7H), 3.68 (d, J=4.0 Hz, 4H), 3.50 (s, 1H), 3.36-3.39 (m, 3H), 2.64 (s, 2H), 2.27-2.23 (m, 1H), 2.19-2.17 (m, 1H), 1.13 (d, J=4.0 Hz, 6H). Example 94. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(morpholin-4-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl N-(5-bromo-3-methoxypyrazin-2-yl)-N-(tert-butoxycarbonyl)carbamate To a stirred mixture of 5-bromo-3-methoxypyrazin-2-amine (1 g, 4.901 mmol, 1.00 equiv) and (Boc)2O (2.35 g, 10.782 mmol, 2.2 equiv) in DCM (20 mL) was added TEA (1.09 g, 10.782 mmol, 2.2 equiv) and DMAP (0.06 g, 0.490 mmol, 0.1 equiv) at 25° C. under nitrogen atmosphere. The resulting mixture was warmed up to 50° C. and stirred for 16 hours. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA in PE, 1% to 21% gradient in 20 min to afford the title compound (1.4 g, 70.66%) as an off-white solid. m/z (ESI, +ve ion)=403.95 [M+H]+ Step B. tert-butyl N-(tert-butoxycarbonyl)-N-[3-methoxy-5-(morpholin-4-yl)pyrazin-2-yl]carbamate To a stirred mixture of tert-butyl N-(5-bromo-3-methoxypyrazin-2-yl)-N-(tert-butoxycarbonyl)carbamate (700 mg, 1.945 mmol, 1.00 equiv) and morpholine (254.24 mg, 2.917 mmol, 1.5 equiv) in 1,4-dioxane (2 mL) were added (DiMeIHeptCl)Pd(cinnamyl)Cl (CAS: 2138491-47-9, 454.22 mg, 0.389 mmol, 0.2 equiv) and Cs2CO3(296.26 mg, 3.890 mmol, 2 equiv) at 25° C. under nitrogen atmosphere. The mixture was warmed up to 100° C. and stirred for 2 h. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA in PE, 26% to 46% gradient in 20 min; Detector, UV: 254 nm to afford the title compound (270 mg, 33.81%) as an off-white solid. m/z (ESI, +ve ion)=411.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.48 (s, 1H), 3.93 (s, 3H), 3.89-3.82 (m, 4H), 3.58-3.53 (m, 4H), 1.45 (s, 18H). Step C. 3-methoxy-5-(morpholin-4-yl)pyrazin-2-amine Into a vial were added tert-butyl N-(tert-butoxycarbonyl)-N-[3-methoxy-5-(morpholin-4-yl)pyrazin-2-yl]carbamate (270 mg, 0.658 mmol, 1.00 equiv) and 1,1,1,3,3,3-hexafluoropropan-2-ol (2 mL). The mixture was warmed up to 60° C. and stirred for 16 h. The mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC (PF:EA=1:1) to afford the title compound (90 mg, 64%) as an off-white solid. m/z (ESI, +ve ion)=211.20 [M+H]+. Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-5-(morpholin-4-yl)pyrazin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 3-methoxy-5-(morpholin-4-yl)pyrazin-2-amine (10 mg, 0.048 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (30.04 mg, 0.048 mmol, 1 equiv) in 1,4-dioxane (1 mL, 11.350 mmol, 238.62 equiv) were added XantPhos (24 mg, 0.2 equiv), Cs2CO3(31.00 mg, 0.096 mmol, 2 equiv) and Pd2(dba)3(8.71 mg, 0.010 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was warmed up to 90° C. The mixture was concentrated under reduced pressure. The crude product was purified by silica gel chromatography, eluted with PE:EA=1:5 to afford the title compound (30 mg, 70%) as an off-white solid. m/z [ESI, +ve ion]=714.60, [M+H]+. Step E. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(morpholin-4-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-5-(morpholin-4-yl)pyrazin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (20 mg, 0.028 mmol) in DCM (2.5 mL) was added TFA (0.5 mL) at 25° C. The solution was stirred for 30 minutes. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC with following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO) Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 19% B to 29% B in 8 min, 29% B; wavelength: 254 nm; RT1(min): 7.28 to afford Example 94 (10 mg, 69%) as an off-white solid. m/z (ESI, +ve ion) 514.35 [M+H]+.1H NMR (400 MHz, Methanol-d4): δ 7.61 (d, J=8.4 Hz, 1H), 7.37 (s, 1H), 7.12 (s, 1H), 6.89 (d, J=8.5 Hz, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.63 (dd, J=8.4, 2.6 Hz, 1H), 5.65 (d, J=2.5 Hz, 1H), 4.06 (s, 3H), 3.87-3.75 (m, 4H), 3.33 (s, 5H), 3.30 (s, 3H), 2.25-2.16 (m, 2H). Example 95. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-2-(morpholin-4-yl)pyridin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl N-(tert-butoxycarbonyl)-N-(2-chloro-3-methoxypyridin-4-yl)carbamate To the mixture of 2-chloro-3-methoxypyridin-4-amine (300 mg, 1.892 mmol, 1.00 equiv) in DCM (10.00 mL) and TEA (574.25 mg, 5.676 mmol, 3 equiv) were added Boc2O (1238.55 mg, 5.676 mmol, 3 equiv) and DMAP (23.11 mg, 0.189 mmol, 0.1 equiv) at 0° C. The mixture was stirred at 25° C. for 12 h. The mixture was diluted with DCM (50 mL) and washed with sat. NaHCO3 (20 mL) and brine (20 mL). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel column eluted with 0-50% of EA in PE to give the title compound (420 mg, 61.88%) as a light yellow solid. m/z (ESI=ve ion)=359.25 [M+H]+ Step B. tert-butyl N-[3-methoxy-2-(morpholin-4-yl)pyridin-4-yl]carbamate To the mixture of 61a (300 mg, 0.836 mmol, 1.00 equiv) and morpholine (145.68 mg, 1.672 mmol, 2 equiv) in dry dioxane (5 mL, 59.020 mmol, 70.59 equiv) were added Cs2CO3(544.82 mg, 1.672 mmol, 2 equiv) and (DiMeIHeptCl)Pd(cinnamyl)C1 (195.20 mg, 0.167 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (5 mL×3). The filtrate was removed under reduced pressure. The residue was purified by silica gel column eluted with 0-100% of EA in PE to give crude the title compound (200 mg, 38.66%) as a yellow solid. m/z (ESI+ve ion)=310.20 [M+H]+ Step C. 3-methoxy-2-(morpholin-4-yl)pyridin-4-amine The mixture of tert-butyl N-[3-methoxy-2-(morpholin-4-yl)pyridin-4-yl]carbamate (200 mg, 0.323 mmol, 1.00 equiv) in TFA (0.5 mL, 6.732 mmol, 20.83 equiv) and DCM (3 mL) was stirred at 25° C. for 1 h. The solvent was removed under reduced pressure. The residue was purified by reverse phase flash with the following conditions: Column: AQ-C18 Column, 40 g, 40 g, 60 Å, 40-60 μm; Mobile Phase A: 10 mM aq. NH4HCO3, Mobile Phase B: MeCN; Flow rate: 60 mL/min; Gradient: 0% B to 0% B in 5 min, 0% B to 40% B in 30 min (41% hold 5 min); Detector: UV 254 & 280 nm. The product-containing fractions were concentrated to afford the title compound (60 mg, 88.71%) as a white solid. m/z (ESI+ve ion)=210.15 [M+H]+ Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-2-(morpholin-4-yl)pyridin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 3-methoxy-2-(morpholin-4-yl)pyridin-4-amine (13.25 mg, 0.063 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (40.0 mg, 0.063 mmol, 1.00 equiv) in toluene (2.0 mL) were added Pd2(dba)3(11.60 mg, 0.013 mmol, 0.2 equiv) and XantPhos (7.33 mg, 0.013 mmol, 0.2 equiv) and Cs2CO3(41.28 mg, 0.126 mmol, 2.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (25.0 mg, 55.37%) as a yellow solid. m/z (ESI, +ve ion)=713.55 [M+H]+. Step E. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-2-(morpholin-4-yl)pyridin-4-yl]amino}-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-24morpholin-4-yl)pyridin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (20.0 mg) in DCM (5.0 mL) was added TFA (0.5 mL) dropwise at room temperature under air atmosphere. The resulting mixture was stirred for 2.0 h at room temperature under air atmosphere. The resulting mixture was concentrated under vacuum. The crude product (22 mg) was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3). Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 28% B to 38% B in 8 min, 38% B; wavelength: 254 nm to afford Example 95 (10.8 mg, 75.09%) as a white solid. m/z (ESI, +ve ion)=513.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.66 (d, J=8.0 Hz, 2H), 7.42 (s, 1H), 7.13 (d, J=4.0 Hz, 1H), 6.95 (d, J=8.0 Hz, 1H), 6.84 (d, J=8.0 Hz, 1H), 6.68-6.66 (m, 1H), 5.61 (d, J=4.0 Hz, 1H), 4.85 (s, 4H), 3.85-3.93 (m, 7H), 3.31-3.28 (m, 4H), 2.21-2.31 (m, 2H). Example 96. (1R,2S)-2-{3-[(5-chloro-2-methylpyridin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-chloro-2-methylpyridin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (135 mg, 0.215 mmol, 1.00 equiv) and 5-chloro-2-methylpyridin-4-amine (30 mg, 0.215 mmol, 1 equiv) and Cs2CO3(139 mg, 0.430 mmol, 2 equiv) in 1,4-dioxane (5 mL) were added Pd2(dba)3(39 mg, 0.043 mmol, 0.2 equiv) and XantPhos (4.95 mg, 0.043 mmol, 0.2 equiv) at 25° C. under nitrogen atmosphere. The mixture was allowed to warmed up to 90° C. and stirred for 2 h. The mixture was allowed to cool down to 25° C. The residue was purified by silica gel column chromatography, eluted with EA in PE, 20% to 40% gradient in 20 min to afford the title compound (100 mg, 65%) as an off-white solid. m/z=646.30 [M+H]+. Step B. (1R,2S)-2-{3-[(5-chloro-2-methylpyridin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-chloro-2-methylpyridin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.152 mmol, 1.00 equiv) in DCM (5 mL) was added TFA (1 mL) at 25° C. The solution was stirred for 30 minutes. The residue was purified by RP flash, MeCN in water (5 mM NH4HCO3), 10% to 30% gradient in 20 min to afford Example 96 (37.4 mg, 55.30%) as an off-white solid. m/z (ESI, +ve ion)=446.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.71 (s, 1H), 10.44 (s, 1H), 8.55 (s, 1H), 8.23 (s, 1H), 7.64 (d, J=8.4 Hz, 1H), 7.44 (s, 1H), 7.22 (s, 1H), 6.95 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (dd, J=8.4, 2.6 Hz, 1H), 5.69 (d, J=2.5 Hz, 1H), 3.30-3.20 (m, 4H), 2.40-2.18 (m, 4H), 1.99-1.97 (m, 1H). Example 97. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-methoxy-2-(morpholin-4-yl)pyrimidin-4-amine To a stirred solution of 2-chloro-5-methoxypyrimidin-4-amine (200 mg, 1.253 mmol, 1.00 equiv) and morpholine (545.98 mg, 6.265 mmol, 5 equiv) in 1,4-dioxane (5 mL) was added TEA (253.66 mg, 2.506 mmol, 2 equiv) at 25° C. under nitrogen atmosphere. The mixture was allowed to warm up to 120° C. for 2 hours. The residue was purified by silica gel column chromatography, eluted with 50% EA in PE to afford the title compound (54 mg, 20.49%) as an off-white solid. m/z (ESI, +ve ion)=211.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.57 (s, 1H), 6.40 (s, 2H), 3.62-3.59 (m, 4H), 3.49-3.47 (m, 4H), 3.60 (s, 3H). Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-methoxy-2-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-methylidenespiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 5-methoxy-2-(morpholin-4-yl)pyrimidin-4-amine (44 mg, 0.209 mmol, 1.00 equiv), tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (132.16 mg, 0.209 mmol, 1 equiv) and Cs2CO3(136.38 mg, 0.418 mmol, 2 equiv) in 1,4-dioxane (2 mL) were added Pd2(dba); (38.33 mg, 0.042 mmol, 0.2 equiv) and XantPhos (24.22 mg, 0.042 mmol, 0.2 equiv) at 25° C. under nitrogen atmosphere. The mixture was allowed to warmed up to 90° C. The mixture was cooled down to 25° C. The residue was purified by silica gel column chromatography, eluted with EA in PE, 80% to 100% gradient in 20 min; Detector. UV 254 nm to afford the title compound (80 mg, 53.70%) as an off-white solid. m/z (ESI, +ve ion)=715.10[M+H]+. Step C. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-methoxy-2-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-methylidenespiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.112 mmol, 1.00 equiv) in DCM (5 mL) was added TFA (1 mL) at 25° C. The solution was stirred for 30 minutes. The residue was purified by prep-HPLC with following condition: Column: XBridge Prep OBD C18 Column, 19×250 mm, 5 μm; Mobile Phase A: Water (10 mmoL/L NH4HCO3), Mobile Phase B: MeOH-HPLC; Flow rate: 25 mL/min; Gradient: 55% B to 60% B in 10 min, 60% B: wavelength: 254 nm; RT1(min): 9 to afford Example 97 (30 mg, 52.12%) as an off-white solid. m/z (ESI, +ve ion)=514.25 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.78-7.72 (m, 3H), 7.37 (s, 1H), 6.91 (d, J=8.3 Hz, 1H), 6.83 (d, J=8.4 Hz, 1H), 6.63 (dd, J=8.5, 2.6 Hz, 1H), 5.52 (d, J=2.5 Hz, 1H), 3.92 (s, 3H), 3.57-3.47 (m, 9H), 3.38 (s, 3H), 2.31-2.28 (m, 1H), 2.09-2.06 (m, 1H). Example 98. (1R,2S)-2-{3-[(5-chloro-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl)-3-[(5-chloro-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 5-chloro-2-methylpyrimidin-4-amine (21.6 mg, 0.150 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (95.00 mg, 0.150 mmol, 1 equiv) in 1,4-dioxane (2 mL) and Cs2CO3(98.04 mg, 0.300 mmol, 2 equiv) were added Pd2(dba)3(27.55 mg, 0.030 mmol, 0.2 equiv) and XantPhos (17.41 mg, 0.030 mmol, 0.2 equiv) at 25° C. under nitrogen atmosphere. The mixture was warmed up to 90° C. and stirred for 2 h. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA in PE, 20% to 40% gradient in 20 min to afford the title compound (91 mg, 90%) as an off-white solid. m/z (ESI, +ve ion)=647.25 [M+H]+. Step B. (1R,2S)-2-{3-[(5-chloro-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-chloro-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (91 mg) in DCM (5 mL) was added TFA (1 mL) at 25° C. The solution was stirred for 30 minutes. Desired product could be detected by LCMS. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC with following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 27% B to 37% B in 8 min, 37% B; wavelength: 254 nm; RT1(min): 7 to afford Example 98 (29 mg) as an off-white solid. m/z (ESI, +ve ion)=447.15 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.27 (s, 1H), 7.60-7.36 (m, 2H), 6.94 (dd, J=8.4, 1.4 Hz, 1H), 6.84 (d, J=8.5 Hz, 1H), 6.63 (dd, J=8.5, 2.5 Hz, 1H), 5.61 (d, J=2.5 Hz, 1H), 3.38 (t, J=8.4 Hz, 1H), 3.30 (s, 3H), 2.32 (s, 3H), 2.47-2.08 (m, 2H). Example 99. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(morpholin-4-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl N-(tert-butoxycarbonyl)-N-[3-methoxy-6-(morpholin-4-yl) pyrazin-2-yl]carbamate To a stirred solution of tert-butyl (tert-butoxycarbonyl)(6-chloro-3-methoxypyrazin-2-yl)carbamate (100.0 mg, 0.278 mmol, 1.00 equiv) and morpholine (72.64 mg, 0.834 mmol, 3.0 equiv) in dioxane (5.0 mL) were added (DiMeIHeptCl)Pd(cinnamyl)Cl (CAS: 2138491-47-9, 64.89 mg, 0.056 mmol, 0.2 equiv) and Cs2CO3(181.11 mg, 0.556 mmol, 2.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 5 h at 100° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/3) to afford the title compound (60.0 mg, 27.61%) as a yellow solid. m/z (ESI, +ve ion)=411.22 [M+H]+. Step B. 3-methoxy-6-(morpholin-4-yl)pyrazin-2-amine To a stirred solution of tert-butyl N-(tert-butoxycarbonyl)-N-[3-methoxy-6-(morpholin-4-yl)pyrazin-2-yl]carbamate (100.0 mg, 0.244 mmol, 1.00 equiv) in DCM (20.0 mL) was added TFA (2.0 mL) dropwise at room temperature. The resulting mixture was stirred for 5 h at room temperature. The resulting mixture was concentrated under reduced pressure to afford the title compound (40.0 mg, 78.10%) as a light yellow solid. m/z (ESI, +ve ion)=211.10 [M+H]+. Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-6-(morpholin-4-yl)pyrazin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 3-methoxy-6-(morpholin-4-yl)pyrazin-2-amine (19.98 mg, 0.095 mmol, 1.0 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60.0 mg, 0.095 mmol, 1.00 equiv) in toluene (3.0 mL) were added Cs2CO3(61.92 mg, 0.190 mmol, 2.0 equiv), XantPhos (11.00 mg, 0.019 mmol, 0.2 equiv) and Pd2(dba)3(17.40 mg, 0.019 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (4/1) to afford the title compound (50.0 mg, 73.72%) as a yellow solid. m/z (ESI, +ve ion)=714.35 [M+H]+. Step D. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(morpholin-4-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-6-(morpholin-4-yl)pyrazin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (50.0 mg, 0.070 mmol, 1.00 equiv) in DCM (10.0 mL) was added TFA (1.0 mL) dropwise at room temperature. The resulting mixture was stirred for 5 h at room temperature. The reaction was monitored by LCMS. The resulting mixture was concentrated under vacuum. The crude product (30.0 mg) was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 35% B in 8 min, 35% B; wavelength: 254 nm to afford Example 99 (11.7 mg, 32.52%) as a pink solid. m/z (ESI, +ve ion)=514.40 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.60 (d, J=8.0 Hz, 1H), 7.45 (s, 1H), 6.96 (s, 1H), 6.86 (d, J=8.0 Hz, 2H), 6.64 (d, J=8.0 Hz, 1H), 5.61 (d, J=4.0 Hz, 1H), 4.01 (s, 3H), 3.5-3.57 (m, 4H), 3.30 (s, 4H), 3.08 (d, J=8.0 Hz, 4H), 2.28-2.24 (m, 1H), 2.19-2.15 (m, 1H). Example 100. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(oxetan-3-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl N-(tert-butoxycarbonyl)-N-[3-methoxy-6-(oxetan-3-yl)pyrazin-2-yl]carbamate The mixture of tert-butyl (tert-butoxycarbonyl)(6-chloro-3-methoxypyrazin-2-yl)carbamate (160 mg, 0.445 mmol, 1.00 equiv), trifluoro(oxetan-3-yl)-lambda4-borane potassium (145.84 mg, 0.890 mmol, 2 equiv), [Ir{dFCF3ppy}2(bpy)]PF6(44.90 mg, 0.045 mmol, 0.1 equiv), [4,4′-Bis(1,1-dimethylethyl)-2,2′-bipyridine] nickel (II) dichloride (26.55 mg, 0.067 mmol, 0.15 equiv) and Na2CO3(94.26 mg, 0.890 mmol, 2 equiv) in DMA (0.4 mL) and dioxane (1.6 mL) was stirred for 16 h at 25° C. under nitrogen atmosphere. The reaction mixture is irradiated under blue LEDs and away from light. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (110 mg, 64.85%) as a light yellow solid. m/z (ESI+ve ion)=382.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.97 (s, 1H), 5.05-5.02 (m, 2H), 4.95-4.92 (m, 2H), 4.45-4.37 (m, 1H), 4.01 (s, 3H), 1.44 (s, 18H). Step B. 3-methoxy-6-(oxetan-3-yl)pyrazin-2-amine To a stirred solution of tert-butyl N-(tert-butoxycarbonyl)-N-[3-methoxy-6-(oxetan-3-yl)pyrazin-2-yl]carbamate (100 mg, 0.262 mmol, 1.00 equiv) in DCM (2 mL) was added TFA (1 mL). The resulting mixture was stirred for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure to afford the title compound (45 mg, 94.73%) as an off-white solid. m/z (ESI+ve ion)=182.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.16 (s, 1H), 6.38 (s, 2H), 4.80-4.70 (m, 4H), 4.20-4.12 (m, 1H), 3.86 (s, 3H). Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-6-(oxetan-3-yl)pyrazin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (90 mg, 0.143 mmol, 1.00 equiv) and 3-methoxy-6-(oxetan-3-yl)pyrazin-2-amine (30.99 mg, 0.172 mmol, 1.2 equiv) in toluene (2.50 mL) were added Cs2CO3(92.88 mg, 0.286 mmol, 2 equiv), Pd2(dba); (26.10 mg, 0.029 mmol, 0.2 equiv) and XantPhos (16.49 mg, 0.029 mmol, 0.2 equiv) under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was filtered and washed with EA (3×5 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (80 mg, 81.97%) as a yellow solid. m/z (ESI+ve ion)=685.30 [M+H]+. Step D. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(oxetan-3-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-6-(oxetan-3-yl)pyrazin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (75 mg, 0.110 mmol, 1.00 equiv) in HFIP (5 mL) was stirred at 60° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 23% B to 33% B in 8 min; wavelength: 254 nm; RT1(min): 7. The product-containing fractions were collected and concentrated in vacuo to afford Example 100 (29.8 mg, 56.10%) as a white solid. m/z (ESI+ve ion)=485.25 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.69 (d, J=8.4 Hz, 1H), 7.41-7.40 (m, 2H), 6.92-6.89 (m, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.64-6.62 (m, 1H), 5.63 (d, J=2.4 Hz, 1H), 4.81-4.75 (m, 4H), 4.25-4.19 (m, 1H), 4.08 (s, 3H), 3.37 (d, J=8.4 Hz, 1H), 3.31 (s, 3H), 2.25-2.22 (m, 1H), 2.20-2.16 (m, 1H). Example 101. (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(propan-2-yl)pyridazin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-chloro-3-methoxypridazin-4-amine To a stirred solution of 3,6-dichloropyridazin-4-amine (500.0 mg, 3.049 mmol, 1.00 equiv) in MeOH (10.0 mL) was added NaOMe (658.87 mg, 12.196 mmol, 4.0 equiv) in portions at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 55° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (3×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford the title compound (300.0 mg, 61.66%) as a light yellow solid. m/z (ESI, +ve ion)=160.05 [M+H]+.1H NMR (300 MHz, DMSO-d6) δ 7.18 (s, 1H), 3.98 (d, J=0.8 Hz, 3H). Step B. 3-methoxy-6-(prop-1-en-2-yl)pyridazin-4-amine To a stirred solution of 6chloro-3-methoxypyridazin-4-amine (240.0 mg, 1.504 mmol, 1.00 equiv) and 4,4,5,5-tetramethyl-2-(prop-1-en-2-yl)-1,3,2-dioxaborolane (303.29 mg, 1.805 mmol, 1.2 equiv) in dioxane (10.0 mL) and water (1.0 mL) were added Pd(dppf)Cl2·DCM (122.52 mg, 0.150 mmol, 0.1 equiv) and Na2CO3(318.82 mg, 3.008 mmol, 2.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 110° C. under nitrogen atmosphere. The mixture was cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (110.0 mg, 44.27%) as a white solid. m/z. (ESI, +ve ion)=166.15 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.50 (d, J=8.0 Hz, 1H), 5.64 (s, 2H), 5.32 (d, J=4.0 Hz, 2H), 4.17 (s, 3H), 2.30 (d, J=4.0 Hz, 3H). Step C. 6-isopropyl-3-methoxypyridazin-4-amine To a solution of 3-methoxy-6-(prop-1-en-2-yl)pyridazin-4-amine (60.0 mg, 0.363 mmol, 1.00 equiv) in 3.0 mL EtOH was added Pd(OH)2/C (20%, 60.0 mg) under nitrogen atmosphere in a 50 mL round-bottom flask. The mixture was hydrogenated at room temperature for 2 h under hydrogen atmosphere using a hydrogen balloon, filtered through a Celite pad, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (30.0 mg, 49.40%) as a white solid. m/z (ESI, +ve ion)=168.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 6.56 (s, 1H), 4.13 (s, 3H), 3.17 (d, J=8.0 Hz, 1H), 1.30 (d, J=8.0 Hz, 6H). Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-isopropyl-3-methoxypyridazin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 6-isopropyl-3-methoxypyridazin-4-amine (21.18 mg, 0.127 mmol, 1.0 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.0 mg, 0.127 mmol, 1.00 equiv) in toluene (4.0 mL) were added Cs2CO3(82.56 mg, 0.254 mmol, 2.0 equiv), XantPhos (14.66 mg, 0.025 mmol, 0.2 equiv) and Pd2(dba)3(23.20 mg, 0.025 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EA (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/2) to afford the title compound (60.0 mg, 70.61%) as a yellow solid. m/z (ESI, +ve ion)=671.40 [M+H]+. Step E. (1R,2S)-2-{3-[(6-isopropyl-3-methoxypyridazin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-[(6-isopropyl-3-methoxypyridazin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60.0 mg, 0.089 mmol, 1.00 equiv) in DCM (10.0 mL) was added TFA (1.0 mL, 13.463 mmol) dropwise at room temperature. The resulting mixture was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (30.0 mg) was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 26% B to 36% B in 8 min, 36% B; wavelength: 254 nm to afford Example 101 (18.6 mg, 44.19%) as a white solid. m/z (ESI, +ve ion)=471.35 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.79 (s, 1H), 7.73 (d, J=8.0 Hz, 1H), 7.44 (s, 1H), 6.96 (d, J=8.0 Hz, 1H), 6.84 (d, J=8.0 Hz, 1H), 6.63 (d, J=8.0 Hz, 1H), 5.61 (d, J=4.0 Hz, 1H), 4.21 (s, 3H), 3.38 (d, J=8.0 Hz, 1H), 3.31 (s, 3H), 3.09-3.06 (m, 1H), 2.27-2.24 (m, 1H), 2.20-2.18 (m, 1H), 1.29 (d, J=8.0 Hz, 6H). Example 102. (1R,2S)-5′-methoxy-2-(3-{[6-(morpholin-4-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-hydroxy-2-isopropyl-3H-pyrimidin-4-one To MeOH (10 mL) was added sodium 2-methylpropan-2-olate (1.96 g, 20.392 mmol, 2.5 equiv) at 0° C. under nitrogen atmosphere. Then 2-methylpropanimidamide hydrochloride (I g, 8.157 mmol, 1.00 equiv) and dimethyl malonate (1.08 g, 8.157 mmol, 1 equiv) were added. The mixture was stirred at 80° C. for 16 h. After cooled to 0° C., the PH value was adjusted to 4 with conc. HCl and filtered. The filter cake was lyophilized overnight to give the title compound (1 g, 79.52%) as a white solid. m/z (ESI, +ve ion)=155.05[M+H]+.1H NMR (400 MHz, DMSO-4) δ 5.12 (s, 1H), 2.82-2.74 (m, 1H), 1.18 (s, 3H), 1.16 (s, 3H). Step B. 4,6-dichloro-2-isopropylpyrimidine The mixture of 6-hydroxy-2-isopropyl-3H-pyrimidin-4-one (1 g, 6.486 mmol, 1.00 equiv) in POCl3(10 mL) was stirred at 100° C. for 5 h. After cooled to room temperature, the PH of mixture was adjusted to 8 with sat. aq. NH4HCO3. The resulting mixture was extracted with EA (50 mL×3). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4and filtered. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-30% of EA in PE to afford the title compound (330 mg, 26.63%) as a colorless oil.1H NMR (400 MHz, Chloroform-d) δ 7.25 (s, 1H), 3.25-3.15 (m, 1H), 1.37 (s, 3H), 1.35 (s, 3H). Step C. 6-chloro-2-isopropylpyrimidin-4-amine The mixture of 4,6-dichloro-2-isopropylpyrimidine (330 mg, 1.727 mmol, 1.00 equiv) in NH4OH (3 mL) and THF (3 mL) was stirred at 60° C. for 8 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to give the title compound (250 mg, 84.33%) as a white solid. m/z (ESI, +ve ion)=172.05 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.09 (s, 2H), 6.25 (s, 1H), 2.81-2.72 (m, 1H), 1.15 (d, J=6.8 Hz, 6H). Step D. 2-isopropyl-6-(morpholin-4-yl)pyrimidin-4-amine The mixture of 6-chloro-2-isopropylpyrimidin-4-amine (250 mg, 1.457 mmol, 1.00 equiv), morpholine (253.81 mg, 2.914 mmol, 2 equiv) and TEA (442.19 mg, 4.371 mmol, 3 equiv) in dioxane (2 mL, 23.608 mmol, 16.21 equiv) was stirred at 120° C. for 8 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (200 mg, 61.77%) as a white solid. m/z (ESI, +ve ion)=223.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 6.11 (s, 2H), 5.44 (s, 1H), 3.66-3.64 (m, 4H), 3.40-3.37 (m, 4H), 2.71-2.64 (m, 1H), 1.15 (s, 3H), 1.13 (s, 3H). Step E. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-isopropyl-6-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of 2-isopropyl-6-(morpholin-4-yl)pyrimidin-4-amine (38.02 mg, 0.172 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (90 mg, 0.143 mmol, 1.00 equiv) in toluene (2.5 mL) were added Cs2CO3(92.88 mg, 0.286 mmol, 2 equiv), XantPhos (16.49 mg, 0.029 mmol, 0.2 equiv) and Pd2(dba)3(26.10 mg, 0.029 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The resulting mixture was filtered and washed with EA (3×5 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-50% EA in PE to give crude the title compound (70 mg, 67.66%) as a light yellow solid. m/z (ESI, +ve ion)=726.45 [M+H]+. Step F. (1R,2S)-2-(3-{[2-isopropyl-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-isopropyl-6-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70 mg, 0.096 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (5 mL) was stirred at 25° C. for 6 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 39% B to 49% B in 8 min; wavelength: 254 nm; RT1(min): 6.6. The product-containing fractions were collected and concentrated in vacuo to give Example 102 (30 mg, 59.18%) as a white solid. m/z (ESI, +ve ion)=526.40 [M+H]+.1H NMR (400 MHz, DMSO-d4) δ 12.29 (s, 1H), 10.41 (s, 1H), 9.66 (s, 1H), 7.98 (s, 1H), 7.32 (s, 1H), 7.06 (s, 1H), 6.90-6.87 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.69 (d, J=2.4 Hz, 1H), 3.71-3.68 (m, 4H), 3.52-3.50 (m, 4H), 3.32 (s, 3H), 3.20-3.15 (m, 1H), 2.85-2.78 (m, 1H), 2.34-2.31 (m, 1H), 1.99-1.96 (m, 1H), 1.22 (d, J=6.8 Hz, 6H). Example 103. (1R,2S)-2-(3-{[5-chloro-2-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-(morpholin-4-yl)pyrimidin-4-amine A solution of 2-chloropyrimidin-4-amine (258 mg, 1.992 mmol, 1.00 equiv), TEA (403.04 mg, 3.984 mmol, 2 equiv) and morpholine (208.21 mg, 2.390 mmol, 1.2 equiv) in dioxane (2 mL, 23.608 mmol, 11.85 equiv) was stirred for 16 h at 100° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography, eluted with 50% ACN in water (5 mM NH4HCO3) to afford the title compound (200 mg, 55.73%) as a white solid. m/z (ESI +ve ion)=181.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.75 (d, J=5.6 Hz, 1H), 6.40 (s, 2H), 5.76 (d, J=5.6 Hz, 1H), 3.63-3.56 (m, 8H) Step B. 5-chloro-2-(morpholin-4-yl)pyrimidin-4-amine To a stirred mixture of 2-(morpholin-4-yl)pyrimidin-4-amine (108 mg, 0.599 mmol, 1.00 equiv) in ACN (3 mL) and DMF (3 mL) was added NCS (88.03 mg, 0.659 mmol, 1.1 equiv). The resulting mixture was stirred for 16 h at room temperature. The resulting mixture was diluted with water (30 mL). The resulting mixture was extracted with EA (3×20 mL). The combined organic layers were washed with brine (5×5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography, eluted with 40% ACN in water (5 mM NH4HCO3) to afford the title compound (100 mg, 77.74%) as a white solid. m/z (ESI, +ve ion)=215.05 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.00-7.84 (m, 1H), 6.87-6.80 (m, 2H), 3.74-3.54 (m, 8H) Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-chloro-2-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 5-chloro-2-(morpholin-4-yl)pyrimidin-4-amine (25.29 mg, 0.118 mmol, 1.20 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (62 mg, 0.098 mmol, 1.00 equiv) in toluene (2 mL) were added Pd2(dba)3(8.99 mg, 0.010 mmol, 0.1 equiv) and XantPhos (5.68 mg, 0.010 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 100° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford crude the title compound (60 mg, 42.54%) as a yellow solid. m/z (ESI, +ve ion)=718.10 [M+H]+ Step D. (1R,2S)-2-(3-{[5-chloro-2-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-chloro-2-(morpholin-4-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60 mg, 0.084 mmol, 1.00 equiv) and TFA (0.5 mL) in DCM (1 mL) was stirred for 1 h at room temperature. The crude product was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 19×250 mm, 10 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 50% B in 8 min, 50% B; wavelength: 254 nm; RT1(min): 7.8 to afford Example 103 (26 mg, 60.03%) as a white solid. m/z (ESI+ve ion)=518.30 [M+H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.98 (s, 1H), 7.51 (d, J=8.6 Hz, 2H), 6.91-6.82 (m, 2H), 6.65 (dd, J=8.5, 2.5 Hz, 1H), 5.64 (d, J=2.5 Hz, 1H), 3.51-3.33 (m, 7H), 3.32-3.19 (m, 5H), 2.32-2.24 (m, 1H), 2.22-2.14 (m, 1H). Example 104. (1R,2S)-2-(3-{[5-(3-hydroxyazetidin-1-yl)-3-methoxypyridin-2-yl]amino}-1H-indazol-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 104 was prepared using procedures similar to those of other examples, but using a 5-(3-hydroxyazetidin-1-yl)-3-methoxypyridin-2-yl starting material. Example 105. (1R,2S)-5′-methoxy-2-(3-{[3-methyl-6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 105 was prepared using procedures similar to those of other examples, but using a 3-methyl-6-(propan-2-yl)pyrazin-2-yl starting material. Example 106. (1R,2S)-5′-methoxy-2-(3-{[6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Example 106 was prepared using procedures similar to those of other examples, but using a 6-(propan-2-yl)pyrazin-2-yl starting material. Example 107. (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-methoxy-1-methylindole-2,3-dione To the mixture of 5-methoxy-1H-indole-2,3-dione (1 g, 5.645 mmol, 1.00 equiv) in DMF (20 mL) was added NaH (0.14 g, 5.927 mmol, 1.05 equiv, 60% in mineral oil) under nitrogen atmosphere at 0° C. After stirred at 0° C. for 30 min, methyl iodide (0.88 g, 6.210 mmol, 1.1 equiv) was added drop-wise. The resulting mixture was stirred at 25° C. for 2 h. The reaction was quenched with sat. aq·NH4Cl (10 mL) and extracted with EA (3×30 mL). The combined organic layers were washed with brine (30 mL×2), dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-50% EA in PE to afford the title compound (900 mg, 83.40%) as a brown solid. m/z (ESI, +ve ion)=192.00 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.19-7.15 (m, 2H), 6.84-6.82 (m, 1H), 3.83 (s, 3H), 3.24 (s, 3H). Step B. 5-methoxy-1-methyl-3H-indol-2-one A solution of 5-methoxy-1-methylindole-2,3-dione (900 mg, 4.707 mmol, 1.00 equiv) in hydrazine hydrate (98%) (10 mL) was stirred for 5 h at 150° C. under argon atmosphere. The mixture was diluted with water (50 mL) and extracted with EA (3×50 mL). The combined organic layer was washed with brine (50 mL), dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-50% EA in PE to afford the title compound (700 mg, 83.92%) as a yellow solid. m/z (ESI, +ve ion)=178.05 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 6.90-6.89 (m, 1H), 6.84-6.82 (m, 1H), 6.73 (d, J=8.4 Hz, 1H), 3.81 (s, 3H), 3.52 (s, 2H), 3.21 (s, 3H). Step C. (1R,2S)-2-(1-benzylindazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′-one A solution of 5-methoxy-1-methyl-3H-indol-2-one (100 mg, 0.564 mmol, 1.00 equiv) in THF (3.00 mL) was cooled in an ice bath. NaH (28.44 mg, 1.184 mmol, 2.1 equiv, 60% in mineral oil) was added in portions, and the mixture was stirred at 0° C. for 30 min. A solution of (1S)-1-(1-benzylindazol-6-yl)-2-(methanesulfonyloxy)ethyl methanesulfonate (239.55 mg, 0.564 mmol, 1 equiv) in THF (7.14 mL) was added dropwise over 10 min and stirred for 3 h. The reaction was quenched with sat aq. NH4Cl (50 mL) and extracted with EA (50 mL×3). The organic layer was washed with brine (50 mL), dried with Na2SO4, and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-50% EA in PE to afford the title compound (130 mg, 56.26%) as a light yellow solid. m/z (ESI, +ve ion)=410.15 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.03 (d, J=0.8 Hz, 1H), 7.66 (d, J=8.4 Hz, 1H), 7.29-7.26 (m, 3H), 7.23 (s, 1H), 7.17-7.15 (m, 2H), 6.98-6.96 (m, 1H), 6.77 (d, J=8.4 Hz, 1H), 6.69-6.66 (m, 1H), 5.65-5.54 (m, 2H), 5.40 (d, J=2.4 Hz, 1H), 3.46 (t, J=8.4 Hz, 1H), 3.33 (s, 3H), 3.25 (s, 3H), 2.26-2.22 (m, 1H), 2.00-1.97 (m, 1H). Step D. (1R,2S)-2-(1H-indazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′-one A solution of (1R,2S)-2-(1-benzylindazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′-one (330 mg, 0.806 mmol, 1.00 equiv) in THF (2.5 mL) was cooled in ice before addition of Potassium tert-butoxide in THF (1.8 M, 8.95 mL, 16.120 mmol, 20 equiv). DMSO (1196.35 mg, 15.314 mmol, 19 equiv) was added and the mixture was purged gently with oxygen in an ice bath for 3 h. The reaction was quenched by sat. aq. NH4Cl (15 mL) and extracted with EA (20 mL×4). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to afford the title compound (220 mg, 85.48%) as a light yellow solid. m/z (ESI, +ve ion)=320.15 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.10 (s, 1H), 7.70 (d, J=8.0 Hz, 1H), 7.44 (s, 1H), 7.03 (d, J=8.4 Hz, 1H), 6.80-6.77 (m, 1H), 6.69-6.67 (m, 1H), 5.55 (t, J=2.4 Hz, 1H), 3.49 (t, J=8.4 Hz, 1H), 3.34 (d, J=3.2 Hz, 6H), 2.30-2.27 (m, 1H), 2.08-2.05 (m, 1H). Step E. (1R,2S)-2-(3-iodo-1H-indazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of (1R,2S)-2-(1H-indazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′-one (90 mg, 0.282 mmol, 1.00 equiv) and K2CO3(77.90 mg, 0.564 mmol, 2 equiv) in DMF (1.00 mL) was added a solution of 12 (121.59 mg, 0.479 mmol, 1.7 equiv) in DMF (1.00 mL) drop-wise. The resulting mixture was stirred for 3 h at room temperature. The mixture was poured in a mixture of water (5 mL) and sat. aq. Na2S2O3(5 mL) and the mixture was stirred at 25° C. for 1 h. The mixture was extracted with EA (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel column eluted with 0-90% of EA in PE to give the title compound (90 mg, 71.73%) as a white solid. m/z (ESI, +ve ion)=446.00 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 13.48 (s, 1H), 7.52 (s, 1H), 7.33 (d, J=8.4 Hz, 1H), 7.05-7.03 (m, 1H), 6.92 (d, J=8.4 Hz, 1H), 6.70-6.67 (m, 1H), 5.69 (d, J=2.8 Hz, 1H), 3.34 (s, 3H), 3.28-3.24 (m, 1H), 3.22 (s, 3H), 2.42-2.39 (m, 1H), 2.06-2.03 (m, 1H). Step F. tert-butyl 3-iodo-6-[(1R,2S)-5′-methoxy-1′-methyl-2′-oxospiro[cyclopropane-1,3′-indol]-2-yl]indazole-1-carboxylate To a stirred mixture of (1R,2S)-2-(3-iodo-1H-indazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′-one (90 mg, 0.202 mmol, 1.00 equiv) and TEA (40.91 mg, 0.404 mmol, 2 equiv) in DCM (2 mL, 31.460 mmol, 155.64 equiv) was added Boc2O (66.17 mg, 0.303 mmol, 1.5 equiv) dropwise at 25° C. The mixture was stirred at 25° C. for 4 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (97 mg, 87.99%) as a white solid. m/z (ESI, +ve ion)=546.05 [M+H]+.1H NMR (400 MHz, DMSO-d4) δ 8.11 (s, 1H), 7.27 (d, J=8.0 Hz, 1H), 7.18-7.16 (m, 1H), 6.79 (d, J=8.4 Hz, 1H), 6.71-6.68 (m, 1H), 5.64 (d, J=2.4 Hz, 1H), 3.39-3.43 (m, 4H), 3.38 (s, 3H), 3.32-2.29 (m, 1H), 2.11-2.08 (m, 1H), 1.71 (s, 9H). Step G. tert-butyl 3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloropyrimidin-4-yl)amino]-6-[(1R,2S)-5′-methoxy-1′-methyl-2′-oxospiro[cyclopropane-1,3′-indol]-2-yl]indazole-1-carboxylate To a stirred mixture of tert-butyl 3-iodo-6-[(1R,2S)-5′-methoxy-1′-methyl-2′-oxospiro[cyclopropane-1,3′-indol]-2-yl]indazole-1-carboxylate (90 mg, 0.165 mmol, 1.00 equiv) and 6-(3-((tert-butyldimethylsilyl)oxy)azetidin-1-yl)-5-chloropyrimidin-4-amine (62.36 mg, 0.198 mmol, 1.2 equiv) in toluene (2.50 mL) were added Cs2CO3(107.54 mg, 0.330 mmol, 2 equiv), Pd2(dba)3(15.11 mg, 0.017 mmol, 0.1 equiv) and XantPhos (9.55 mg, 0.017 mmol, 0.1 equiv) under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was filtered and washed with EA (3×5 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (70 mg, 43.44%) as a yellow solid. m/z (ESI, +ve ion)=732.30 [M+H]+. Step H. (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl 3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloropyrimidin-4-yl)amino]-6-[(1R,2S)-5′-methoxy-1′-methyl-2′-oxospiro[cyclopropane-1,3′-indol]-2-yl]indazole-1-carboxylate (65 mg, 0.089 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (2 mL) was stirred at 40° C. for 24 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 37% B in 8 min; wavelength: 254 nm; RT1(min): 6.8. The product-containing fractions were collected and concentrated in vacuo to Example 107 (22.3 mg, 48.41%) as a white solid. m/z (ESI, +ve ion)=518.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.83 (s, 1H), 7.51 (d, J=8.4 Hz, 1H), 7.43 (s, 1H), 6.95-6.92 (m, 2H), 6.73-6.70 (m, 1H), 5.70 (d, J=2.4 Hz, 1H), 4.65-4.59 (m, 3H), 4.19-4.113 (m, 2H), 3.41-3.38 (m, 1H), 3.36 (s, 3H), 3.32 (s, 3H), 2.29-2.19 (m, 2H). Example 108. (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-1′-ethyl-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 1-ethyl-5-methoxyindole-2,3-dione To the mixture of 5-methoxy-1H-indole-2,3-dione (1 g, 5.645 mmol, 1.00 equiv) in DMF (20 mL) was added NaH (0.14 g, 5.927 mmol, 1.05 equiv, 60% in mineral oil) under nitrogen atmosphere at 0° C. After stirred at 0° C. for 30 min, ethyl iodide (0.97 g, 6.210 mmol, 1.1 equiv) was added drop-wise. The resulting mixture was stirred at 25° C. for 2 h. The reaction was quenched with sat.aq. NH4Cl (10 mL) and extracted with EA (30 mL×3). The combined organic layers were washed with brine (30 mL×2), dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-50% EA in PE to afford the title compound (1 g, 86.33%) as a brown solid. m/z (ESI, +ve ion)=206.10 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.17-7.14 (m, 2H), 6.85-6.83 (m, 1H), 3.82 (s, 3H), 3.80-3.75 (m, 2H), 1.32 (t, J=7.2 Hz, 3H). Step B. 1-ethyl-5-methoxy-3H-indol-2-one A solution of 1-ethyl-5-methoxyindole-2,3-dione (1 g, 4.873 mmol, 1.00 equiv) in hydrazine hydrate (98%, 10 mL, 205.751 mmol, 43.71 equiv) was stirred for 5 h at 150° C. under argon atmosphere. The mixture was diluted with water (50 mL) and extracted with EA (50 mL×3). The combined organic layer was washed with brine (50 mL), dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-50% EA in PE to afford the title compound (800 mg, 85.85%) as a yellow solid. m/z (ESI, +ve ion)=192.05 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 6.91-6.90 (m, 1H), 6.84-6.81 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 3.81 (s, 3H), 3.79-3.74 (m, 2H), 3.51 (s, 2H), 1.28 (t, J=7.2 Hz, 3H). Step C. (1R,2S)-2-(1-benzylindazol-6-yl)-1′-ethyl-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′-one A solution of 1-ethyl-5-methoxy-3H-indol-2-one (180.20 mg, 0.942 mmol, 1 equiv) in THF (6 mL) was cooled in an ice bath. NaH (47.49 mg, 1.978 mmol, 2.1 equiv, 60% in mineral oil) was added in portions, and the mixture was stirred at 0° C. for 15 min. A solution of (1S)-1-(1-benzylindazol-6-yl)-2-(methanesulfonyloxy)ethyl methanesulfonate (400 mg, 0.942 mmol, 1.00 equiv) in THF (12 mL) was added drop-wise over 20 min and stirred for 3 h. The reaction was quenched with sat. aq. NH4Cl (50 mL) and extracted with EA (3×50 mL). The organic layer was washed with brine (50 mL), dried with Na2SO4, and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-50% EA in PE to afford the title compound (200 mg, 50.11%) as a light yellow solid. m/z (ESI, +ve ion)=424.30 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.03 (d, J=0.8 Hz, 1H), 7.65 (d, J=8.4 Hz, 1H), 7.27-7.25 (m, 3H), 7.22 (d, J=1.6 Hz, 1H), 7.17-7.15 (m, 2H), 6.97-6.94 (m, 1H), 6.79 (d, J=8.4 Hz, 1H), 6.68-6.65 (m, 1H), 5.70-5.49 (m, 2H), 5.40 (d, J=2.8 Hz, 1H), 3.97-3.79 (m, 2H), 3.45 (t, J=8.4 Hz, 1H), 3.24 (s, 3H), 2.25-2.22 (m, 1H), 1.98-1.95 (m, 1H), 1.34 (t, J=7.2 Hz, 3H). Step D. (1R,2S)-1′-ethyl-2-(1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′-one A solution of (1R,2S)-2-(1-benzylindazol-6-yl)-1′-ethyl-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′-one (280 mg, 0.661 mmol, 1.00 equiv) in THF (1.8 mL) was cooled in ice before addition of Potassium tert-butoxide in THF (1.8 M, 7.35 mL, 13.220 mmol, 20 equiv). DMSO (981.47 mg, 12.559 mmol, 19 equiv) was added and the mixture was purged gently with oxygen in an ice bath for 3 h. The reaction was quenched by sat. aq. NH4Cl (15 mL) and extracted with EA (20 mL×4). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to afford the title compound (220 mg, 99.81%) as a light yellow solid. m/z (ESI, +ve ion)=334.10 [M+H]+.1H NMR (400 MHz, DMSO-d4) δ 13.01 (s, 1H), 8.03 (s, 1H), 7.66 (d, J=8.4 Hz, 1H), 7.47 (s, 1H), 6.97-6.93 (m, 2H), 6.68-6.65 (m, 1H), 5.68 (d, J=2.4 Hz, 1H), 3.84-3.74 (m, 2H), 3.30 (s, 3H), 3.26 (t, J=8.4 Hz, 1H), 2.38-2.35 (m, 1H), 2.06-2.02 (m, 1H), 1.22-1.16 (m, 3H). Step E. (1R,2S)-1′-ethyl-2-(3-iodo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of (1R,2S)-1′-ethyl-2-(1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′-one (220 mg, 0.660 mmol, 1.00 equiv) and K2CO3(182.40 mg, 1.320 mmol, 2 equiv) in DMF (3 mL) was added a solution of I2(284.72 mg, 1.122 mmol, 1.7 equiv) in DMF (3 mL) dropwise. The mixture was stirred at 25° C. for 4 h. The reaction was quenched by the addition of sat. aq. Na2S2O3(10 mL). The aqueous layer was extracted with EA (3×20 mL). The combined organic layers were washed with brine (3×20 mL), dried over anhydrous Na2SO4and filtered. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to afford the title compound (245 mg, 80.84%) as a yellow solid. ma (ESI, +ve ion)=460.05 [M+H]+. Step F. tert-butyl 6-[(1R,2S)-1′-ethyl-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indol]-2-yl]-3-iodoindazole-1-carboxylate To a stirred mixture of (1R,2S)-1′-ethyl-2-(3-iodo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′-one (245 mg, 0.533 mmol, 1.00 equiv) and TEA (107.96 mg, 1.066 mmol, 2 equiv. in DCM (5 mL was added Boc2O (174.63 mg, 0.800 mmol, 1.5 equiv) at 25° C. The mixture was stirred at 25° C. for 4 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (268 mg, 89.81%) as a white solid. m/z (ESI, +ve ion)=560.05 [M+H]+.1H NMR (400 MHz, Chloroform-d) 8.08 (s, 1H), 7.42-7.39 (m, 1H), 7.19-7.17 (m, 1H), 6.81 (d, J=8.4 Hz, 1H), 6.70-6.67 (m, 1H), 5.64 (d, J=2.8 Hz, 1H), 3.94-3.82 (m, 2H), 3.47 (t, J=8.4 Hz, 1H), 3.42 (s, 3H), 2.32-2.28 (m, 1H), 2.09-2.06 (m, 1H), 1.70 (s, 9H), 1.35 (t, J=7.2 Hz, 3H). Step G. tert-butyl 3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloropyrimidin-4-yl)amino]-6-[(1R,2S)-1′-ethyl-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indol]-2-yl]indazole-1-carboxylate To a stirred mixture of tert-butyl 6-[(1R,2S)-1′-ethyl-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indol]-2-yl]-3-iodoindazole-1-carboxylate and 6-(3-((tert-butyldimethylsilyl)oxy)azetidin-1-yl)-5-chloropyrimidin-4-amine (74.30 mg, 0.236 mmol, 1.2 equiv) in toluene (3 mL) were added Cs2CO3(128.14 mg, 0.394 mmol, 2 equiv), Pd2(dba); (36.01 mg, 0.039 mmol, 0.2 equiv) and XantPhos (22.76 mg, 0.039 mmol, 0.2 equiv) under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was filtered and washed with EA (5 mL×3). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (90 mg, 49.06%) as a yellow solid. m/z (ESI, +ve ion)=746.35 [M+H]+. Step H. (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-1′-ethyl-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl 3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-chloropyrimidin-4-yl)amino]-6-[(1R,2S)-1′-ethyl-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indol]-2-yl]indazole-1-carboxylate (90 mg, 0.121 mmol, 1.00 equiv) in TFA (0.5 mL) and DCM (2.5 mL) was stirred at 40° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 24% B to 34% B in 8 min; wavelength: 254 nm; RT1(min): 7. The product-containing fractions were collected and concentrated in vacuo to give Example 71 (29 mg, 44.80%) as a white solid. m/z (ESI, +ve ion)=532.40 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.65 (s, 1H), 8.89 (s, 1H), 7.82 (s, 1H), 7.40 (s, 1H), 7.36 (d, J=8.4 Hz, 1H), 6.96 (d, J=8.8 Hz, 1H), 6.90-6.87 (m, 1H), 6.69-6.66 (m, 1H), 5.76 (d, J=2.4 Hz, 1H), 5.67 (d, J=6.0 Hz, 1H), 4.54-4.44 (m, 3H), 3.99-3.96 (m, 2H), 3.84-3.74 (m, 2H), 3.36 (s, 3H), 3.25 (t, J=8.4 Hz, 1H), 2.39-2.36 (m, 1H), 2.06-2.02 (m, 1H), 1.20 (t, J=6.8 Hz, 3H). Example 109. (1R,2S)-2-(3-{[5-(difluoromethoxy)-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. (1R,2S)-2-(3-iodo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one To a vial was added (1R,2S)-2-(1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (46.0 mg, 151 μmol) followed by DMF (100 μL) and MeOH (100 μL). To this suspension was added potassium carbonate (41.6 mg, 301 μmol). Finally, iodine (48.3 mg, 190 μmol) dissolved in DMF (100 μL) was added dropwise and allowed to stir at rt. After 2 h, another portion of iodine (9.3 mg, 36.58 mmol) in 100 μL of DMF was added to the mixture and stirred for 16 h at it. The mixture was quenched with Na2S2O3in water and stirred for 2 h. The mixture was diluted with excess water and a solid precipitated. The solid was collected by filtration and washed with water, affording the title compound (50.0 mg, 77%). m/z (ESI, +ve ion)=432.0 [M+H]+. Step B. tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-iodo-1H-indazole-1-carboxylate 4-Dimethylaminopyridine (1.5 mg, 11.6 μmol) was added to a solution of di-tert-butyl dicarbonate (73.4 μL, 313 μmol), (1R,2S)-2-(3-iodo-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one (50.0 mg, 116 μmol) and triethylamine (65.3 μL, 464 μmol) in DCM (400 μL). The solution was stirred at room temperature for 16 h and then concentrated to dryness. The product was purified by column chromatography (0 to 10/% EtOAc in hexane), affording the title compound (52.7 mg, 72%) as yellow foamy solid. m/z (ESI, +ve ion)=432.0 [M+H]+−2boc. 1H NMR (400 MHz, CDCl3) δ 8.09 (s, 1H), 7.79 (d, J=8.9 Hz, 1H), 7.38 (d, J=8.3 Hz, 1H), 7.13 (dd, J=8.3, 0.7 Hz, 1H), 6.67 (dd, J=8.9, 2.7 Hz, 1H), 5.51 (d, J=2.6 Hz, 1H), 3.50 (t, J=8.6 Hz, 1H), 3.37 (s, 3H), 2.36 (dd, J=9.2.4.8 Hz, 1H), 2.09 (dd, J=8.1, 4.8 Hz, 1H), 1.68 (d, J=5.0 Hz, 18H). Step C. 6-chloro-5-(difluoromethoxy)pyrimidin-4-amine Potassium hydroxide (1.5 g, 26.4 mmol) was added to a solution of 4-amino-6-chloropyrimidin-5-ol (200.0 mg, 1.32 mmol) dissolved in a mixture of MeCN and water (1:1) (13 mL) and the mixture was stirred in an ice-water bath. Diethyl(bromodifluoromethyl)phosphonate (483 μL, 2.64 mmol) was then added dropwise at 0° C. The reaction was then stirred at rt for 24 h. The mixture was directly loaded onto a C18 column chromatography and the product purified (isocratic 50% MeCN in aq. ammonium formate buffer) affording title compound (50.0 mg, 78%). m/z (ESI, +ve ion)=196.0 [M+H]+. Step D. 5-(difluoromethoxy)-6-morpholinopyrimidin-4-amine In a flask was dissolved 6-chloro-5-(difluoromethoxy)pyrimidin-4-amine (92.0 mg, 470 μmol) in DMF (2.75 mL) to which were added morpholine (100 μL, 1.13 mmol) and cesium carbonate (300 mg, 903 μmol). The reaction was heated to 90° C. and stirred for 22 h. The reaction was cooled down to room temperature and the mixture was passed through a pad of silica gel, washed successively with heptanes, DCM, and finally EtOAc:MeOH (8:2). The crude product eluted with EtOAc/MeOH and was collected and concentrated. The residue was further purified by C18 column chromatography (15-35% MeCN in aq. ammonium formate buffer) to afford title compound (58.0 mg, 47%) as a yellow solid. m/z (ESI, +ve ion)=246.9 [M+H]+. Step E. (1R,2S)-2-(3-((5-(difluoromethoxy)-6morpholinopyrimidin-4-yl)amino)-1H-indazol-1-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one To a microwave vial were added 5-(difluoromethoxy)-6-morpholinopyrimidin-4-amine (25.0 mg, 102 μmol), tert-butyl (1R,2S)-2-(14tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (64.1 mg, 102 μmol), Pd2(dba)3(5.83 mg, 10.1 μmol), XantPhos (6.00 mg, 10.2 μmol), and cesium carbonate (67.5 mg, 203 μmol). Degassed toluene (4.00 mL) was then added and the reaction was purged with nitrogen gas for 30 seconds. The vial was then sealed and the reaction was heated to 100° C. for 16 h in an oil bath. The cooled solution was passed through a pad of silica gel washing successively with hexanes, DCM, and then EtOAc:MeOH (8:2) as eluent. The EtOAc/MeOH portion was collected and concentrated. The crude residue was dissolved in DCM (5.00 mL) and TFA (5.00 mL) and stirred at rt for 1 h. The solvents were removed under vacuum and the residue was purified by silica gel chromatography (30% isopropanol in DCM) and then by C18 column chromatography (45% MeCN in aq. ammonium formate buffer) to afford Example 109 (8.0 mg, 14%) as a white solid. m/z (ESI, +ve ion)=550.2 [M+H]+. 1H NMR (500 MHz, DMSO) δ 12.62 (s, 1H), 10.41 (s, 1H), 9.11 (s, 1H), 7.89 (s, 1H), 7.45-7.36 (m, 2H), 6.95 (t, J=73.6 Hz, 1H), 6.90 (d, J=9.5 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (dd, J=8.5, 2.6 Hz, 1H), 5.72 (d, J=2.6 Hz, 1H), 3.76-3.63 (m, 4H), 3.62-3.48 (m, 4H), 3.33 (s, 3H), 3.18 (t, J=8.5 Hz, 1H), 2.32 (dd, J=7.9, 4.6 Hz, 1H), 2.02-1.95 (m, 1H), 19F NMR (470 MHz, DMSO) δ −82.03 (d, J=73.6 Hz). Example 110. (1R,2S)-2-(3-{[6-(azetidin-3-yl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-1-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl 3-{6-[bis(tert-butoxycarbonyl)amino]-5-methoxypyrazin-2-yl}azetidine-1-carboxylate To a stirred mixture of tert-butyl (tert-butoxycarbonyl)(6-chloro-3-methoxypyrazin-2-yl)carbamate (179.91 mg, 0.500 mmol, 1.00 equiv) and tert-butyl 3-(trifluoro-lambda4-boranyl)azetidine-1-carboxylate potassium (263.12 mg, 1.000 mmol, 2 equiv) in DMA (1 mL) and dioxane (4 mL) were added [Ir{dFCF3ppy}2(bpy)]PF6(50.49 mg, 0.050 mmol, 0.1 equiv), [4,4′-Bis(1,1-dimethylethyl)-2,2′-bipyridine]nickel (II) dichloride (29.85 mg, 0.075 mmol, 0.15 equiv) and Na2CO3(105.99 mg, 1.000 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at room temperature under blue LEDs. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA 1/1) to afford the title compound (175 mg, 72.83%) as a green solid. m/z (ESI+ve ion)=381.25 [M+H−100]+. Step B. tert-butyl 3-(6-amino-5-methoxypyrazin-2-yl)azetidine-1-carboxylate To a stirred mixture of tert-butyl 3-{6-[bis(tert-butoxycarbonyl)amino]-5-methoxypyrazin-2-yl}azetidine-1-carboxylate (120 mg, 0.250 mmol, 1.00 equiv) in MeOH (1 mL) was added NaOH (99.88 mg, 2.500 mmol, 10 equiv) in H2O (0.5 mL) dropwise at room temperature. The resulting mixture was stirred for 16 h at 60° C. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 50% gradient in 50 min; detector, UV 254 nm to afford the title compound (68.8 mg, 98.290) as a white solid. m/z (ESI, +ve ion)=281.25 [M+H]+.1H NMR (400 MHz DMSO-d6) δ 7.17 (d, J=1.0 Hz, 1H), 6.39 (s, 2H), 4.07 (s, 3H), 3.86 (d, J=1.0 Hz, 3H), 3.76-3.61 (m, 4H), 3.34 (s, 1H), 1.40 (d, J=1.0 Hz, 9H). Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({6-[1-(tert-butoxycarbonyl)azetidin-3-yl]-3-methoxypyrazin-2-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl 3-(6-amino-5-methoxypyrazin-2-yl)azetidine-1-carboxylate (31.96 mg, 0.114 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60 mg, 0.095 mmol, 1.00 equiv) in toluene (1 mL) were added Pd2(dba)3(8.70 mg, 0.010 mmol, 0.1 equiv), XantPhos (5.50 mg, 0.010 mmol, 0.1 equiv) and Cs2CO3(61.92 mg, 0.190 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA 1/1) to afford the title compound (45.3 mg, 60.82%) as a white solid. m/z (ESI+ve ion)=784.50 [M+H]+. Step E. (1R,2S)-2-(3-{[6-(azetidin-3-yl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({6-[1-(tert-butoxycarbonyl)azetidin-3-yl]-3-methoxypyrazin-2-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (40 mg, 0.051 mmol, 1.00 equiv) in DCM (2 mL) was added TFA (1 mL) dropwise at room temperature. The resulting mixture was stirred for 1 h at 60° C. The mixture was cooled down to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 40/o B in 8 min, 40% B; wavelength: 254 nm; RT1(min): 6.2 to afford Example 110 (20 mg, 80.90%) as an off-white solid. m/z (ESI, +ve ion)=484.20 [M+H]+. 1H NMR (400 MHz, Methanol-d4) δ 7.60 (d, J=8.4 Hz, 1H), 7.46 (s, 1H), 7.39 (s, 1H), 6.91 (d, J=8.4 Hz, 1H), 6.85 (d, J=8.0 Hz, 1H), 6.65-6.62 (m, 1H), 5.65 (d, J=2.4 Hz, 1H), 4.09-3.92 (m, 4H), 3.93 (m, 3H), 3.75 (m, 2H), 3.50-3.38 (m, 2H), 3.15 (t, J=1.6 Hz, 1H), 2.26 (m, 1H), 2.19 (m, 1H). Example 111. (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-2-isopropylpyrimidin-4-amine The mixture of 3-((tert-butyldimethylsilyl)oxy)azetidine (120 mg, 0.699 mmol, 1.00 equiv), 6-chloro-2-isopropylpyrimidin-4-amine (131.00 mg, 0.699 mmol, 1 equiv) and TEA (141.50 mg, 1.398 mmol, 2 equiv) in dioxane (2 mL) was stirred at 120° C. for 8 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (80 mg, 35.48%) as a white solid. m/z (ESI, +ve ion)=323.35[M+H]+.1H NMR (400 MHz, DMSO-d6) δ 6.09 (s, 2H), 5.09 (s, 1H), 4.75-4.70 (m, 1H), 4.14-4.11 (m, 2H), 3.60-3.57 (m, 2H), 2.68-2.59 (m, 1H), 1.13 (s, 3H), 1.11 (s, 3H), 0.88 (s, 9H), 0.07 (s, 6H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-2-isopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.127 mmol, 1.00 equiv) and 6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-2-isopropylpyrimidin-4-amine (49.03 mg, 0.152 mmol, 1.2 equiv) in toluene (2.0 mL) were added Cs2CO3(82.56 mg, 0.254 mmol, 2 equiv), XantPhos (14.66 mg, 0.025 mmol, 0.2 equiv) and Pd2(dba)3(23.20 mg, 0.025 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The resulting mixture was filtered and washed with EA (3×5 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-50% EA in PE to give crude title compound (70 mg, 66.89%) as a yellow solid. m/z (ESI, +ve ion)=826.50 [M+H]+. Step C. (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-2-isopropylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(6-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-2-isopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70 mg, 0.085 mmol, 1.00 equiv) in TFA (1 mL) and DCM (5 mL) was stirred at 25° C. for 48 h. The solvent was removed under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 45% B in 8 min; wavelength: 254 nm; RT1(min): 6.5. The product containing fractions were collected and concentrated in vacuo to give Example 111 (27.8 mg, 63.94%) as a white solid. m/z (ESI, +ve ion)=512.25 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.70-7.68 (m, 1H), 7.40-7.39 (m, 1H), 6.94-6.92 (m, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.64-6.62 (m, 1H), 6.31 (s, 1H), 5.61 (d, J=2.4 Hz, 1H), 4.71-4.65 (m, 1H), 4.29-4.25 (m, 2H), 3.85-3.80 (m, 2H), 3.38-3.36 (m, 1H), 3.30 (s, 3H), 2.90-2.83 (m, 1H), 2.26-2.17 (m, 2H), 1.27 (d, J=6.8 Hz, 6H). Example 114: (1R,2S)-2-(3-{[2-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-methoxypyrimidin-4-amine To a stirred solution of 2-chloro-5-methoxypyrimidin-4-amine (160.0 mg, 1.003 mmol, 1.00 equiv) and 3-[(tert-butyldimethylsilyl)oxy]azetidine (375.73 mg, 2.006 mmol, 2.0 equiv) in dioxane (2.0 mL) was added TEA (304.39 mg, 3.009 mmol, 3.0 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 100° C. under nitrogen atmosphere. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE:EA (1:1) to afford the title compound (80.0 mg, 25.70%) as a white solid. m/z (ESI, +ve ion)=311.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.55 (s, 1H), 4.72-4.67 (m, 1H), 4.32 (s, 2H), 3.94 (s, 2H), 3.81 (s, 3H), 0.91 (s, 9H), 0.09 (s, 6H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-methoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 2-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-methoxypyrimidin-4-amine (34.42 mg, 0.111 mmol, 1.0 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70.0 mg, 0.111 mmol, 1.00 equiv) in toluene (3.5 mL) were added XantPhos (12.83 mg, 0.022 mmol, 0.2 equiv), Pd2(dba)3(20.30 mg, 0.022 mmol, 0.2 equiv) and Cs2CO3(72.24 mg, 0.222 mmol, 2.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere then cooled down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE:EA (1:1) to afford the title compound (80.0 mg, 88.66%) as a yellow solid. m/z (ESI, +ve ion)=814.60 [M+H]+. Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-{3-[(tert-butyldimethylsilyl)oxy]azetidin-1-yl}-5-methoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (75.0 mg, 0.092 mmol, 1.00 equiv) in tetraethylene glycol (5.0 mL) was added KF (16.1 mg, 0.28 mmol, 3 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 8 h at room temperature under nitrogen atmosphere. The reaction was quenched with water at room temperature. The resulting mixture was extracted with EA (3×30 mL). The combined organic layers were washed with brine (3×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford the title compound (40.0 mg, 62.04%) as a yellow oil. m/z (ESI, +ve ion)=700.35[M+H]+. Step D. (1R,2S)-2-(3-{[2-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70.0 mg, 0.100 mmol, 1.00 equiv) in HFIP (5.0 mL) was stirred for 6 h at 60° C. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (45 mg) was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 17% B to 27% B in 8 min, 27% B; wavelength: 254 nm; RT1(min): 7.43 to afford Example 114 (18.7 mg, 37.42%) as a white solid. m/z (ESI, +ve ion)=500.35 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.58 (d, J=8.4 Hz, 1H), 7.51 (s, 1H), 7.30 (s, 1H), 6.77-6.71 (m, 2H), 6.53-6.50 (m, 1H), 5.49 (d, J=3.6 Hz, 1H), 4.35-4.30 (m, 1H), 3.88-3.79 (m, 6H), 3.53-3.48 (m, 2H), 3.25 (d, J=8.0 Hz, 2H), 3.19 (s, 3H), 2.14-2.11 (m, 1H), 2.08-2.05 (m, 1H). Example 116: (1R,2S)-2-(3-((5-chloro-2-cyclopropylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one Step A. 6-chloro-2-cyclopropylpyrimidin-4-amine To a stirred solution of 4,6-dichloro-2-cyclopropylpyrimidine (400.0 mg, 2.116 mmol, 1.00 equiv) in THF (1.6 mL) was added NH3·H2O (30%, 0.8 mL) dropwise at room temperature. The resulting mixture was stirred for 16 h at 70° C. then cooled down to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE:EA (1:1) to afford the title compound (250.0 mg, 69.66%) as a white solid. m/z (ESI, +ve ion)=170.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 6.25 (s, 1H), 4.92 (s, 2H), 2.06-2.00 (m, 1H), 1.13-1.04 (m, 2H), 1.04-0.98 (m, 2H). Step B. 2-cyclopropylpyrimidin-4-amine To a solution of 6chloro-2-cyclopropylpyrimidin-4-amine (200.0 mg, 1.179 mmol, 1.00 equiv) in 10.0 mL EtOH was added Pd/C (10%, 100 mg) in a 50 mL round bottom flask at room temperature under nitrogen atmosphere. The resulting mixture was hydrogenated at room temperature under 1 atm of hydrogen pressure for 1 h. then filtered through a Celite pad. The filtrate was concentrated under reduced pressure to afford the title compound (120.0 mg, 75.29%) as a white solid. m/z (ESI, +ve ion)=136.25 [M+H]+. Step C. 5-chloro-2-cyclopropylpyrimidin-4-amine To a stirred solution of 2-cyclopropylpyrimidin-4-amine (110.0 mg, 0.814 mmol, 1.00 equiv) in ACN (5.0 mL) was added NCS (130.40 mg, 0.977 mmol, 1.2 equiv) at room temperature. The resulting mixture was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE:EA (1:1) to afford the title compound (60.0 mg, 43.47%) as a white solid. m/z (ESI, +ve ion)=170.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.05 (s, 1H), 7.09 (s, 2H), 1.90-1.86 (m, 1H), 0.92-0.85 (m, 4H). Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-chloro-2-cyclopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 5-chloro-2-cyclopropylpyrimidin-4-amine (21.49 mg, 0.127 mmol, 1.0 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.00 mg, 0.127 mmol, 1.00 equiv) in toluene (4.00 mL) were added Pd2(dba)3(23.20 mg, 0.025 mmol, 0.2 equiv), XantPhos (14.66 mg, 0.025 mmol, 0.2 equiv) and Cs2CO3(82.56 mg, 0.254 mmol, 2.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere then cooled down to room temperature. The resulting mixture was filtered and the filter cake was washed with EA (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE:EA (1:1) to afford the title compound (60.00 mg, 70.35%) as a yellow solid. m/z (ESI, +ve ion)=673.50 [M+H]+. Step E. (1R,2S)-2-{3-[(5-chloro-2-cyclopropylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (0R,2S)-2-[I-(tert-butoxycarbonyl)-3-[(5-chloro-2-cyclopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (55.00 mg, 0.082 mmol, 1.00 equiv) in DCM (5.00 mL) was added TFA (1.00 mL) dropwise at room temperature. The resulting mixture was stirred for 4 h at room temperature then concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3). Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 29% B to 39% B in 8 min, 39% B: wavelength: 254 nm; RT1(min): 7.27 to afford Example 116 (20.00 mg, 51.76%) as a white solid. m/z (ESI, +ve ion)=473.20 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.20 (s, 1H), 7.49-7.41 (m, 2H), 6.94 (d, J=8.0 Hz, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.63-6.61 (m, 1H), 5.64 (d, J=2.4 Hz, 1H), 3.39 (d, J=8.4 Hz, 1H), 3.32 (s, 3H), 2.27-2.21 (m, 1H), 2.21-2.17 (m, 1H), 1.85 (d, J=4.4 Hz, 1H), 0.79-0.62 (m, 4H). Example 117: (1R,2S)-2-(3-{[5-chloro-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. N-(2-bromopyrimidin-4-yl)-N-(tert-butoxycarbonyl)carbamate To a stirred mixture of 2-bromopyrimidin-4-amine (500 mg, 2.874 mmol, 1.00 equiv) and Boc2O (1881.43 mg, 8.622 mmol, 3 equiv) in DCM (4 mL) were added TEA (1163.10 mg, 11.496 mmol, 4 equiv) and DMAP (35.11 mg, 0.287 mmol, 0.1 equiv) at 25° C. The resulting mixture was stirred for 16 h at 60° C. then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with (EA in PE, 2% to 22% gradient in 20 min) to afford the title compound (842 mg, 78.30%) as an off white solid. m/z (ESI, +ve ion)=374.23 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.62 (dd, J=19.3, 5.9 Hz, 1H), 7.73 (dd, J=12.4, 5.9 Hz, 1H), 1.60 (s, 18H). Step B. tert-butyl N-(tert-butoxycarbonyl)-N-[2-prop-1-en-2-yl)pyrimidin-4-yl]carbamate To a stirred solution of tert-butyl N-(2-bromopyrimidin-4-yl)-N-(tert-butoxycarbonyl)carbamate (269 mg, 0.719 mmol, 1.00 equiv) and 4,4,5,5-tetramethyl-2-(prop-1-en-2-yl)-1,3,2-dioxaborolane (120.79 mg, 0.719 mmol, 1 equiv) in H2O (0.2 mL) and 1,4-dioxane (0.4 mL) were added Pd(DtBPF)Cl2(93.70 mg, 0.144 mmol, 0.2 equiv) and K3PO4(305.15 mg, 1.438 mmol, 2 equiv) at 25° C. under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 100° C. then concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluted with EA in PE, 1% to 30% gradient in 20 min to afford the title compound (236 mg, 97.89%) as an off white solid. m/z (ESI, +ve ion)=336.40 [M+H]+. Step C. tert-butyl N-(tert-butoxycarbonyl)-N-(2-isopropylpyrimidin-4-yl)carbamate To a stirred solution of tert-butyl N-(tert-butoxycarbonyl)-N-[2-(prop-1-en-2-yl)pyrimidin-4-yl]carbamate (100 mg, 0.298 mmol, 1.00 equiv) and Pd(OH)2/C (25.12 mg) in EtOH (5 mL) at 25° C. under H2atmosphere. The resulting mixture was stirred for 2 h under 1 atm H2atmosphere then filtered. The filtrate was concentrated under reduced pressure to afford the title compound (80 mg, 79.52%) as an off white solid. m/z (ESI+ve ion)=138.31 [M+H−200]+. Step D. 2-isopropylpyrimidin-4-amine A solution of tert-butyl N-(tert-butoxycarbonyl)-N-(2-isopropylpyrimidin-4-yl)carbamate (236 mg, 0.699 mmol, 1.00 equiv) in 1,1,1,3,3,3-hexafluoropropan-2-ol (0.12 mL, 0.699 mmol, 1 equiv) was stirred for 4 hours at 60° C. then concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EA=1:4) to afford the title compound (74 mg, 77.12%) as an off white solid. m/z (ESI, +ve ion)=138.25 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.22 (d, J=5.8 Hz, 1H), 6.27 (d, J=5.8 Hz, 1H), 4.85 (s, 2H), 2.99 (hept, J=6.9 Hz, 1H), 1.31 (d, J=6.9 Hz, 6H). Step E. 5-chloro-2-isopropylpyrimidin-4-amine To a stirred solution of 2-isopropylpyrimidin-4-amine (100 mg, 0.729 mmol, 1.00 equiv) in THF (5 mL) was added NCS (107.07 mg, 0.802 mmol, 1.1 equiv) at 25° C. The resulting mixture was stirred for 16 h at 40° C. then concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EA=3:2) to afford the title compound (50 mg, 39.97%) as an off white solid. m/z (ESI, +ve ion)=172.20 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.24 (s, 1H), 5.32 (s, 2H), 3.10-3.02 (m, 1H), 1.30 (d, J=6.9 Hz, 6H). Step F. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-chloro-2-isopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (68.04 mg, 0.108 mmol, 1 equiv) and 5-chloro-2-isopropylpyrimidin-4-amine (20 mg, 0.108 mmol, 1.00 equiv) in toluene (0.2 mL) were added Pd2(dba)3(19.73 mg, 0.022 mmol, 0.2 equiv), XantPhos (12.47 mg, 0.022 mmol, 0.2 equiv) and Cs2CO3(70.38 mg, 0.216 mmol, 2 equiv) at 25° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. then concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EA=1:1) to afford the title compound (60 mg, 80.80%) as an off white solid. m/z (ESI, +ve ion)=675.50 [M+H]+. Step G. (1R,2S)-2-{3-[(5-chloro-2-isopropylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one Into a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-chloro-2-isopropylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60 mg, 0.089 mmol, 1.00 equiv) in DCM (2 mL) was added TFA (0.5 mL) at 25° C. The resulting mixture was stirred for 1 hours at 25° C. then concentrated under reduced pressure. The crude product (60 mg) was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 40% B in 8 min, 40% B; wavelength: 254 nm; RT1(min): 6.27 to afford Example 117 (20 mg, 47%) as an off-white solid. m/z (ESI, +ve ion)=475.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.74 (s, 1H), 10.40 (s, 1H), 9.46 (s, 1H), 8.37 (s, 1H), 7.42 (s, 1H), 7.37 (d, J=8.4 Hz, 1H), 6.88 (dd, J=8.5, 1.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (dd, J=8.4, 2.6 Hz, 1H), 5.67 (d, J=2.6 Hz, 1H), 3.30 (s, 3H), 3.20 (t, J=8.4 Hz, 1H), 2.72-2.68 (m, 1H), 2.31 (dd, J=8.0, 4.7 Hz, 1H), 1.99 (dd. J=9.0.4.6 Hz, 1H), 0.98 (d, J=6.9 Hz, 6H). Example 119: (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxetan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl N-(tert-butoxycarbonyl)-N-(2-chloro-5-methoxypyrimidin-4-yl)carbamate To a stirred mixture of 2-chloro-5-methoxypyrimidin-4-amine (300 mg, 1.880 mmol, 1.00 equiv) and triethylamine (570.74 mg, 5.640 mmol, 3 equiv) in THF (5 mL) were added di-tert-butyl dicarbonate (1025.80 mg, 4.700 mmol, 2.5 equiv) and DMAP (91.87 mg, 0.752 mmol, 0.4 equiv) at 0° C. under nitrogen atmosphere. After stirred for 12 h at 25° C., the solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% of EA in PE to afford the title compound (500 mg, 73.91%) as a white solid. m/z (ESI, +ve ion)=360.10 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.29 (s, 1H), 3.98 (s, 3H), 1.46 (s, 18H). Step B. tert-butyl N-(tert-butoxycarbonyl)-N-[5-methoxy-2-(oxetan-3-yl)pyrimidin-4-yl]carbamate Into a 8 mL sealed tube were added tert-butyl N-(tert-butoxycarbonyl)-N-(2-chloro-5-methoxypyrimidin-4-yl)carbamate (200 mg, 0.556 mmol, 1.00 equiv), trifluoro(oxetan-3-yl)potassio-lambda5-borane (182.30 mg, 1.112 mmol, 2 equiv), [Ir{dFCF3ppy}2(bpy)]PF6(56.12 mg, 0.056 mmol, 0.1 equiv), [4,4′-Bis(1,1-dimethylethyl)-2,2′-bipyridine] nickel (II) dichloride (33.18 mg, 0.083 mmol, 0.15 equiv) and Na2CO3(117.83 mg, 1.112 mmol, 2 equiv), DMA (4 mL) under nitrogen atmosphere. The reaction mixture is irradiated under blue LEDs. After stirred at room temperature for 12 h, the solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to afford crude the title compound (60 mg, 16.98%) as a yellow solid. m/z (ESI+ve ion)=382.20 [M+H]+. Step C. 5-methoxy-2-(oxetan-3-yl)pyrimidin-4-amine The mixture of crude tert-butyl N-(tert-butoxycarbonyl)-N-[5-methoxy-2-(oxetan-3-yl)pyrimidin-4-yl]carbamate (60 mg, 0.094 mmol, 1.00 equiv, 60%) in TFA (0.5 mL) and DCM (5 mL) was stirred at 25° C. for 8 h. The solvent was removed under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 5% B to 10% B in 8 min; wavelength: 254 nm; RT1(min): 5.78 to afford the title compound (10 mg, 58.47%) as a white solid. m/z (ESI, +ve ion)=182.10 [M+H]+. Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-methoxy-2-(oxetan-3-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (41.82 mg, 0.066 mmol, 1.00 equiv) and 5-methoxy-2-(oxetan-3-yl)pyrimidin-4-amine (12 mg, 0.066 mmol, 1.00 equiv) in toluene (1 mL) were added Cs2CO3(10.09 mg, 0.132 mmol, 2 equiv), XantPhos (7.66 mg, 0.013 mmol, 0.2 equiv) and Pd2(dba); (12.13 mg, 0.013 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. The mixture was filtered and washed with EA (10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (rinsed with EA) to afford the title compound (30 mg, 52.92%) as a yellow solid. m/z (ESI, +ve ion)=685.35 [M+H]+. Step E. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxetan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-{[5-methoxy-2-(oxetan-3-yl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (30 mg, 0.044 mmol, 1.00 equiv) in HFIP (3 mL) was stirred at 60° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XSelect CSH Prep C18 OBD Column, 19×250 mm, 5 μm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 25% B to 35% B in 10 min; wavelength: 254 nm; RT1 (min): 7 to give Example 119 (8 mg, 37.69%) as a white solid. m/z (ESI, +e ion)=485.25 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.96 (s, 1H), 7.61 (d, J=8.4 Hz, 1H), 7.44 (s, 1H), 6.91 (d, J=8.0 Hz, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.65-6.63 (m, 1H), 5.62 (d, J=2.4 Hz, 1H), 4.81-4.74 (m, 4H), 4.23-4.16 (m, 1H), 4.04 (s, 3H), 3.38-3.36 (m, 1H), 3.32 (s, 3H), 2.26-2.23 (m, 1H), 2.20-2.17 (m, 1H). Example 121: (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-amino-2-methylpyrimidin-5-ol hydrobromide To a stirred mixture of 5-methoxy-2-methylpyrimidin-4-amine (834 mg, 5.993 mmol, 1 equiv) in DCE (30 mL) was added BBr3(5.67 mL, 59.977 mmol, 10.01 equiv) dropwise at room temperature under nitrogen atmosphere. After the resulting mixture was stirred for 16 h at 50° C. under nitrogen atmosphere, 100 mL of DCE was added. The above clear solution was removed and the precipitate was collected and dissolved in MeOH (100 mL). The resulting mixture was concentrated under reduced pressure to afford the title compound (1.2 g, 97.18%) as an off-white solid. m/z (ESI+ve ion)=126.20 [M+H]+. Step B. 5-(difluoromethoxy)-2-methylpyrimidin-4-amine To a stirred mixture of 4-amino-2-methylpyrimidin-5-ol hydrobromide (80 mg, 0.388 mmol, 1 equiv) and 4 M aqueous of KOH (0.97 mL, 3.880 mmol, 10 equiv) in ACN (1 mL) was added diethyl bromodifluoromethylphosphonate (155.51 mg, 0.582 mmol, 1.5 equiv) dropwise at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 0° C. under nitrogen atmosphere then extracted with CHCl3(4×10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM:MeOH (20:1) to afford the title compound (30 mg, 44.12%) as an off-white solid. m/z (ESI, +ve ion)=176.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.91 (d, J=1.3 Hz, 1H), 7.44-6.77 (m, 3H). Step C. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-(difluoromethoxy)-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (65 mg, 0.103 mmol, 1.00 equiv) and 5-(difluoromethoxy)-2-methylpyrimidin-4-amine (21.63 mg, 0.124 mmol, 1.2 equiv) in toluene (2.5 mL) were added Pd2(dba)3(9.43 mg, 0.010 mmol, 0.1 equiv), Cs2CO3(67.08 mg, 0.206 mmol, 2 equiv) and XantPhos (5.96 mg, 0.010 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EA=1:1) to afford the title compound (50 mg, 71.57%) as a yellow solid. m/z (ESI, +ve ion)=679.30 [M+H]+. Step D. (1R,2S)-2-(3-((5-(difluoromethoxy)-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(difluoromethoxy)-2-methylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (10 mg, 0.015 mmol, 1 equiv) in DCM (1 mL) was added TFA (0.5 mL) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere then concentrated under vacuum. The crude product (10 mg) was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 23% B to 33% B in 8 min, 33% B; wavelength: 254 nm; RT1(min): 7.67 to afford Example 121 (5 mg, 70.92%) as a white solid. m/z (ESI, +ve ion)=479.15 [M+H]+.1H-NMR (Methanol-d4, ppm) δ 8.09 (s, 1H), 7.54 (d, J=8.4 Hz, 1H), 7.45 (s, 1H), 7.13-6.77 (m, 3H), 6.63 (d, J=8.4 Hz, 1H), 5.61 (s, 1H), 3.39-3.30 (m, 4H), 2.50 (s, 3H), 2.37-2.18 (m, 2H) Example 122: (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-1′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one tert-butyl (S)-6-(1,2-bis((methylsulfonyl)oxy)ethyl)-3-iodo-1H-indazole-1-carboxylate Step A. 3-iodo-1H-indazole-6-carbaldehyde To a flask was added molecular iodine (14.76 g, 58.16 mmol) and DMF (40 mL). This solution was stirred at room temperature and potassium carbonate (9.46 g, 68.42 mmol) was added. To this mixture was added 6-formyl-1H-indazole (5.0 g, 34.21 mmol) in DMF (40 mL) dropwise and the mixture was stirred for 16 h. Sodium thiosulfate (8.5 g) and potassium carbonate (0.5 g) dissolved in 60 mL water were then added and the mixture was stirred for 1 h. The solution was then poured into 300 mL of ice/water mixture and stirred until melted and the precipitate was collected by vacuum filtration, washed with water, and dried under vacuum to give the title compound as a beige solid (8.53 g, 92%). LCMS: m/z (ESI, +ve ion)=279.2 [M+H]+ Step B. 3-iodo-6-vinyl-1H-indazole To a solution of potassium tert-butoxide (7.74 g, 68.98 mmol) in THF (100 mL) cooled to 0° C. was added methyltriphenylphosphonium bromide (22.40 g, 62.71 mmol) in 4 portions over 15 minutes. The solution was allowed to warm to room temperature and stirred for 1 h. 3-iodo-1H-indazole-6-carbaldehyde (8.53 g, 31.36 mmol) was then added all at once and the mixture was stirred for 1 h. The mixture was diluted with DCM and washed with water (1×) and brine (1×). The organic layer was dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified by flash column chromatography (0-30% EtOAc in hexanes) to give the title compound as a white solid (6.51 g, 77%). LCMS: m/z (ESI, +ve ion)=271.0 [M+H]+1H NMR (400 MHz, CHLOROFORM-d) δ ppm 10.21 (m, J=3.67 Hz, 1H) 7.33-7.52 (m, 3H) 6.76-6.93 (m, 1H) 5.88 (br d, J=17.12 Hz, 1H) 5.39 (br d, J=11.74 Hz, 1H). Step C. tert-butyl 3-iodo-6-vinyl-1H-indazole-1-carboxylate To a solution of 3-iodo-6-vinyl-1H-indazole (8.45 g, 31.29 mmol). N-ethyl-N-isopropyl-propan-2-amine (10.9 mL, 62.58 mmol) and 4-dimethylaminopyridine (191 mg, 1.56 mmol) in acetonitrile (85 mL) at room temperature was added di-tert-butyl dicarbonate (10.24 g, 46.93 mmol) resulting in a light yellow homogeneous solution. The reaction mixture was stirred for 2 h then concentrated in vacuo and purified by flash column chromatography (0-20% EtOAc in hexanes) to give the title compound as a white solid (10.5 g, 91%). LCMS: m/z (ESI, +ve ion)=315.0 [M−tBu+H]+ Step D. tert-butyl (S)-6-(1,2-dihydroxyethyl)-3-iodo-1H-indazole-1-carboxylate To a solution of (DHQ)2PHAL (221 mg, 0.28 mmol) in tert-butanol (142 mL) was added potassium osmate (104.5 mg, 0.28 mmol), potassium carbonate (11.76 g, 85.09 mmol) and potassium hexacyanoferrate(III) (28.02 g, 85.09 mmol) as a solution in water (142 mL) and the mixture was stirred until dissolved. The reaction mixture was cooled to 0° C. and tert-butyl 3-iodo-6-vinyl-indazole-1-carboxylate (10.5 g, 28.36 mmol) was added in one portion and the reaction mixture was stirred at 0° C. for 1 h then slowly warmed to room temperature and stirred for 16 h. Sodium sulfite (30 g) was then added and the reaction mixture was stirred for 1 h. The mixture was diluted with DCM and filtered through celite. The filtrate was washed with water and the aqueous layer was extracted with DCM (2×). The combined organic extracts were dried with sodium sulfate, filtered, and the filtrate was concentrated in vacuo. The crude residue was purified by flash column chromatography (0-100% EtOAc in hexanes) to give the title compound as a white foam (8.47 g, 74%). LCMS: m/z (ESI, +ve ion)=349.0 [M−tBu+H]+. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 8.22 (s, 1H) 7.46-7.53 (m, 1H) 7.37-7.44 (m, 1H) 7.29 (s, 1H) 5.04 (m, J=4.16 Hz, 1H) 3.81-3.97 (m, 1H) 3.74 (m, J=3.42 Hz, 1H) 2.79 (s, 1H) 2.10-2.21 (m, 1H) 1.75 (s, 8H). Chiral HPLC (AD-H, 15 min isocratic 7% iPrOH in hexanes, 15 min) showed 98:1 enantiomeric ratio. (97% ee). Step E. tert-butyl (S)-6-(1,2-bis((methylsulfonyl)oxy)ethyl)-3-iodo-1H-indazole-1-carboxylate To a solution of tert-butyl 6-[(1S)-1,2-dihydroxyethyl]-3-iodo-indazole-1-carboxylate (2.0 g, 4.95 mmol) and triethylamine (4.14 mL, 29.69 mmol) and in DCM (50 mL) cooled to 0° C. was added ethanesulfonyl chloride (1.53 mL, 19.79 mmol) and the reaction mixture was stirred for 15 min at room temperature. The reaction mixture was concentrated in vacuo and the residue was adsorbed onto silica and purified by flash column chromatography (0-100% EtOAc in hexanes) to give the title compound as a white foam (2.59 g, 93%). LCMS: m/z (ESI, +ve ion)=461.0 [M−Boc+H]+. Step F. tert-butyl 3-iodo-6-((1R,2S)-1′-methyl-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate To a vial was added 1-methylindolin-2-one (50.7 mg, 0.34 mmol) followed by THF (6 mL). Sodium hydride (41.3 mg, 1.03 mmol) was added in one portion and the mixture was stirred for 5 min then tert-butyl 6-[(1S)-1,2-bis(methylsulfonyloxy)ethyl]-3-iodo-indazole-1-carboxylate (192.9 mg, 0.34 mmol) was added as a solution in THF (3 mL) dropwise by syringe and the reaction mixture was stirred at room temperature for 1 h. The reaction mixture was quenched with sat. ammonium chloride and extracted with EtOAc (3×). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by flash column chromatography (0-50% EtOAc in hexanes) to give the title compound as a brown solid (39.1 mg, 22%0) LCMS: m/z (ESI, +ve ion)=416.0 [M-Boc+H]+ Step G. tert-butyl 3-((5-methoxy-2-methylpyrimidin-4-yl)amino)-6-((1R,2S)-1′-methyl-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazole-1-carboxylate To a solution of tert-butyl 3-iodo-6-[(1S,2R)-1′-methyl-2′-oxo-spiro[cyclopropane-2,3′-indoline]-1-yl]indazole-1-carboxylate (39.1 mg, 0.08 mmol) in dry toluene (0.75 mL) was added Xantphos Pd G4 (7.3 mg, 0.01 mmol), Xantphos (4.4 mg, 0.01 mmol), cesium carbonate (49.4 mg, 0.15 mmol) and 5-methoxy-2-methyl-pyrimidin-4-amine (11.6 mg, 0.08 mmol). Argon was bubbled through the solution for 5 min then the reaction mixture was heated to 90° C. for 2 h. The reaction mixture was diluted with DCM, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by flash column chromatography (0-10% DCM in MeOH, 24 g) to afford the product as a brown solid (22.6 mg, 57%). LCMS: m/z (ESI, +ve ion)=527.2 [M+H]+ Step H. (1R,2S)-2-(3-((5-methoxy-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-1′-methylspiro[cyclopropane-1,3′-indolin]-2′-one To a solution of tert-butyl 3-[(5-methoxy-2-methyl-pyrimidin-4-yl)amino]-6-[(1S,2R)-1′-methyl-2′-oxo-spiro[cyclopropane-2,3′-indoline]-1-yl]indazole-1-carboxylate (22.6 mg, 0.04 mmol) in DCM (1.0 mL) was added trifluoroacetic acid (164 uL, 2.15 mmol) and the reaction mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated in vacuo and purified by RP-HPLC (20-40% ACN/water, 0.1% FA) to give the title compound as a lyophilized white solid (10.2 mg, 56%). LCMS: m/z (ESI, +ve ion)=427.3 [M+H]+.1H NMR1H NMR (400 MHz, MeOD) δ 7.84 (s, 1H), 7.47-7.38 (m, 1H), 7.34 (s, 1H), 7.09-6.98 (m, 1H), 6.97-6.88 (m, 1H), 6.85-6.76 (m, 1H), 6.58-6.48 (m, 1H), 5.99-5.89 (m, 1H), 3.95 (s, 3H), 3.32-3.25 (m, 1H), 3.24 (s, 3H), 2.28 (s, 3H), 2.21-2.12 (m, 1H), 2.12-2.04 (m, 1H). Example 125: (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-isopropyl-5-methoxypyrimidine-4,6-diol A mixture of 1,3-dimethyl 2-methoxypropanedioate (4 g, 24.670 mmol, 1 equiv) and 2-methylpropanimidamide hydrochloride (3.18 g, 25.904 mmol, 1.05 equiv) in NaOMe (15.55 g, 86.345 mmol, 3.5 equiv, 30% in methanol) was stirred for 1.5 h at 70° C. under nitrogen atmosphere. The mixture was cooled down to room temperature and stirred for 16 h at room temperature under nitrogen atmosphere. The mixture was cooled down to 0° C., 8 mL of conc. HCl was added. The resulting mixture was filtered, the filter cake was washed with Et2O (3×15 mL). The solid was collected and dried to afford the title compound (8 g, 88.03%) as an off-white solid. m/z (ESI, +ve ion)=185.10 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 11.58 (s, 2H), 3.59 (s, 3H), 2.83-2.73 (m, 1H), 1.15 (d, J=6.8 Hz, 6H). Step B. 4,6-dichloro-2-isopropyl-5-methoxypyrimidine To a stirred mixture of 2-isopropyl-5-methoxypyrimidine-4,6-diol (3.6 g, 19.545 mmol, 1 equiv) and TEA (2.18 g, 21.500 mmol, 1.1 equiv) in toluene (8 mL) was added POCl3(6.59 g, 42.999 mmol, 2.2 equiv) dropwise at 100° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 110° C. under nitrogen atmosphere. The mixture was cooled down to room temperature and then quenched by the addition of water (10 mL) at room temperature. The resulting mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with water (2×6 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford the title compound (3.2 g, 74.06%) as a colorless oil. m/z (ESI, +ve ion)=221.10 [M+H]+.1H NMR (400 MHz, Chloroform-d S 3.96 (s, 3H), 3.21-3.11 (m, 1H), 1.34 (d, J=6.8 Hz, 6H). Step C. 6-chloro-2-isopropyl-5-methoxypyrimidin-4-amine The mixture of 4,6-dichloro-2-isopropyl-5-methoxypyrimidine (1 g, 4.523 mmol, 1 equiv) in NH3H2O (30%, 10 mL) and THF (10 mL) was stirred at 60° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-50% EA in PE to afford the title compound (600 mg, 65.78%) as a white solid. m/z (ESI, +ve ion)=202.05 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.14 (s, 2H), 3.68 (s, 3H), 2.88-2.73 (m, 1H), 1.16 (d, J=6.8 Hz, 6H). Step D. 2-isopropyl-5-methoxypyrimidin-4-amine To the mixture of 6-chloro-2-isopropyl-5-methoxypyrimidin-4-amine (100 mg, 0.496 mmol, 1 equiv) in MeOH (5 mL) was added Pd/C (52.77 mg, 10%) under nitrogen atmosphere. The resulting mixture was degassed and purged with H2for three times then was stirred at 25° C. for 2 h. The mixture was filtered and washed with MeOH (10 mL). The filtrate was concentrated in vacuo to give the title compound (100 mg, 96.48%) as a grey solid. m/z (ESI, +ve ion)=168.05 [M+H]+. Step E. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-isopropyl-5-methoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1 equiv) and 2-isopropyl-5-methoxypyrimidin-4-amine (31.78 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) and XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOAc (10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% EA in PE to afford the title compound (20 mg, 18.83%) as a yellow solid. m/z (ESI, +ve ion)=671.35 [M+H]+ Step F. (1R,2S)-2-{3-[(2-isopropyl-5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-isopropyl-5-methoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (20 mg, 0.030 mmol, 1 equiv) in TFA (0.5 mL) and DCM (3 mL) was stirred for 4 h at 25° C. The solvent was removed under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 45% B in 8 min, 45% B; wavelength: 254 nm; RT1(min): 7 to give Example 125 (8.1 mg, 57.62%) as a white solid. m/z (ESI, +ve ion)=471.25 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.86 (s, 1H), 7.59 (d, J=8.4 Hz, 1H), 7.44-7.43 (m, 1H), 6.91-6.83 (m, 2H), 6.64-6.61 (m, 1H), 5.62 (d, J=2.4 Hz, 1H), 4.01 (s, 3H), 3.39-3.37 (m, 1H), 3.30 (s, 3H), 2.86-2.80 (m, 1H), 2.26-2.23 (m, 1H), 2.19-2.17 (m, 1H), 1.06 (t, J=6.8 Hz, 6H). Example 128: (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4,6-dichloro-2-isopropylpyrimidin-5-ol To the mixture of 4,6-dichloro-2-isopropyl-5-methoxypyrimidine (500 mg, 2.262 mmol, 1 equiv) in DCM (5 mL) was added BBr3(5.67 g, 22.620 mmol, 10 equiv) at 25° C. under nitrogen atmosphere. The mixture was stirred at 40° C. for 12 h then quenched with H2O (5 mL) at 0° C. The resulting mixture was extracted with EA (5×20 mL) The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to afford the title compound (450 mg, 96.10%) as a white solid. m/z (ESI, +ve ion)=207.00 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 5.69 (s, 1H), 3.20-3.10 (m, 1H), 1.33 (d, J=6.8 Hz, 6H). Step B. 4,6-dichloro-5-(difluoromethoxy)-2-isopropylpyrimidine To a stirred mixture of 4,6-dichloro-2-isopropylpyrimidin-5-ol (150 mg, 0.724 mmol, 1 equiv) in ACN (3.75 mL) was added a solution of KOH (812.93 mg, 14.480 mmol, 20 equiv) in H2O (3.75 mL). The resulting mixture was cooled to 0° C. and diethyl bromodifluoromethylphosphonate (386.87 mg, 1.448 mmol, 2 equiv) was added under nitrogen atmosphere. After the mixture was stirred at 0° C. for 0.5 h, it was diluted with brine (50 mL) and extracted with EA (3×20 mL). The combined organic layers were washed with brine (2×25 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-20% of EA in PE to afford the title compound (220 mg, 94.51%) as a colorless oil.1H NMR (400 MHz, DMSO-d4) δ 7.39-7.01 (m, 1H), 3.19-3.0) (m, 1H), 1.28-1.26 (m, 6H).19F NMR (377 MHz, DMSO-d6) δ −79.95 (s, 2F). Step C. 6-chloro-5-(difluoromethoxy)-2-isopropylpyrimidin-4-amine In a 40 mL sealed tube, the mixture of 4,6-dichloro-5-(difluoromethoxy)-2-isopropylpyrimidine (220 mg, 0.685 mmol, 1 equiv, 80%) in NH3·H2O (8 mL, 30%) and THF (2 mL) was stirred for 12 h at 70° C. After SM consumed, the solvent was removed under reduced pressure to give the title compound (200 mg, 98.34%) as a white solid. m/z (ESI, +ve ion)=238.00 [M+H]+. Step D. 5-(difluoromethoxy)-2-isopropylpyrimidin-4-amine To the mixture of 6-chloro-5-(difluoromethoxy)-2-isopropylpyrimidin-4-amine (200 mg, 0.842 mmol, 1 equiv) in MeOH (4 mL) was added Pd/C (89.57 mg, 10%) under nitrogen atmosphere. The resulting mixture was de-gassed and purged with H2for three times. After stirred for 3 h at 25° C. under H2atmosphere, SM was consumed. The mixture was filtered and washed with MeOH (10 mL). The filtrate was concentrated in vacuo. The residue was purified by RP-Flash with the following conditions: Column: AQ-C18 Column, 40 g, 60 Å, 40-60 μm; Mobile Phase A: 10 mM aq. NH4HCO3, Mobile Phase B: MeCN; Flow rate: 60 mL/min; Gradient: 0% B to 0% B in 5 min, 0% B to 30% B in 30 min; Detector: UV 254 & 220 nm to give the title compound (80 mg, 46.78%) as a white solid. m/z (ESI, +ve ion)=204.05 [M+H]+.1H NMR (400 MHz, DMSO-d4) δ 7.97-7.96 (m, 1H), 7.23-6.86 (m, 1H), 6.98-6.97 (m, 2H), 2.88-2.81 (m, 1H), 1.19-1.17 (m, 6H).19F NMR (377 MHz, DMSO) δ −81.50 (s, 2F). Step E. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(difluoromethoxy)-2-isopropylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1 equiv) and 5-(difluoromethoxy)-2-isopropylpyrimidin-4-amine (38.61 mg, 0.190 mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), Pd2(dba); (29.00 mg, 0.032 mmol, 0.2 equiv) and XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EA (10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to afford the title compound (50 mg, 40.21%) as a yellow solid. m/z (ESI, +ve ion)=707.35 [M+H]+ Step F. (1R,2S)-2-(3-{[5-4difluoromethoxy)-2-isopropylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred mixture of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-{[5-(difluoromethoxy)-2-isopropylpyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (50 mg, 0.071 mmol, 1 equiv) in DCM (5 mL) was added TFA (0.5 mL) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 4 h at room temperature under nitrogen atmosphere. The solvent was removed under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 40% B in 8 min; wavelength: 254 nm; RT11(min): 7 to give Example 128 (21.7 mg, 60.50%) as a white solid. m/z (ESI, +ve ion)=507.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.14-8.13 (m, 1H), 7.52 (d, J=8.4 Hz, 1H), 7.46 (s, 1H), 7.13-6.77 (m, 3H), 6.64-6.61 (m, 1H), 5.62 (d, J=2.4 Hz, 1H), 3.38 (d, J=8.4 Hz, 1H), 3.30 (s, 3H), 2.88-2.81 (m, 1H), 2.27-2.17 (m, 2H), 1.06 (t, J=6.8 Hz, 6H).19F NMR (376 MHz, Methanol-d4) δ −83.88 (s, 2F). Example 132: (1R,2S)-2-(3-{[5-chloro-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-chloro-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)pyrimidin-4-amine To a 50 ml round bottom flask containing 5,6-Dichloro-4-pyrimidinamine (1000.0 mg, 6.098 mmol) in toluene (9.0 mL) were added 2-Oxa-6-azaspiro[3.3]heptane (1:1) (625.4 mg, 6.308 mmol). The reaction mixture was heated to 100° C. and stirred, monitored by LCMS until the full conversion of the starting materials (approx. 2.5 hrs). Then the reaction mixture was cooled down to rt, diluted with acetone. The organic layer was then concentrated under reduced pressure and the residue was purified by column chromatography (DCM/MeOH=0˜4%) to provide the title compound 1a (868.0 mg, 63%) as a pale yellow oil. Step B. tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[5-chloro-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)pyrimidin-4-yl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a 50 ml round bottom flask were added cesium carbonate (60.8 mg, 0.187 mmol), tert-butyl (1R,2S)-2-(1-tert-butoxycarbonyl-3-iodo-indazol-6-yl)-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (59.0 mg, 0.934 mmol), Tris(dibenzylideneacetone)dipalladium(0) (8.6 mg, 0.0093 mmol), 1a (23.3 mg, 0.103 mmol), Xantphos (5.4 mg, 0.010 mmol) and dioxane (1.0 mL). The reaction mixture was stirred and purged with argon (in balloon) for 10 min to form a green suspension, and then heated to 100° C., resulting in a yellow suspension. The reaction was monitored by LCMS and TLC until the full conversion of the starting materials (approx. 2.5 hrs), cooled down to rt, diluted with EtOAc, washed with sat. aq. NaHCO3and dried over Na2SO4. The residue was purified by column chromatography (DCM/MeOH=0˜6%) to provide the title compound 1b (17.2 mg, 25%) as a yellow solid. Step C. (1R,2S)-2-[3-[[5-chloro-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)pyrimidin-4-yl]amino]-1H-indazol-6-yl]-5′-methoxy-spiro[cyclopropane-1,3′-indoline]-2′-one To a 50 mil round bottom flask containing 1b (17.2 mg, 0.0235 mmol) in DCM (0.50 mL) was added trifluoroacetic acid (0.13 mL, 1.6 mmol). The reaction mixture was stirred and monitored by LCMS until the full conversion of the starting materials (approx. 2 hrs), diluted with methanol, quenched with 1N NaOH. The resulting brown solution was purified by Prep. HPLC (Gemini C18, 10 to 40% (0.1% Formic Acid in water)/(0.1% Formic Acid in Acetonitrile)) to provide Example 132 product as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ ppm 1.97 (br dd, J=4.40, 2.93 Hz, 1H) 2.07 (br d, J=0.73 Hz, 1H) 2.28-2.37 (m, 1H) 3.17 (br d, J=2.45 Hz, 1H) 4.42 (br s, 4H) 4.71 (br s, 4H) 5.70 (br s, 1H) 6.58 (br d, J=7.34 Hz, 1H) 6.74 (br d, J=7.09 Hz, 1H) 6.84-6.93 (m, 1H) 7.27-7.36 (m, 1H) 7.40 (br d, J=0.73 Hz, 1H) 7.82 (br s, 1H) 8.93 (br s, 1H) 10.43 (br s, 1H) 12.68 (br s, 1H); m/z (ESI, +ve ion) 530.2 (M+H)+. Example 134: (1R,2S)-2-{3-[(5-ethoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-ethoxy-2-methylpyrimidin-4-amine To a stirred mixture of 4-amino-2-methylpyrimidin-5-ol hydrobromide (80 mg, 0.388 mmol, 1 equiv) in acetone (2 mL) were added ethyl iodide (72.67 mg, 0.466 mmol, 1.2 equiv) and Cs2CO3(404.82 mg, 1.242 mmol, 3.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 50° C. under nitrogen atmosphere then diluted with water (10 mL). The resulting mixture was extracted with CHCl3(4×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (DCM:MeOH, 10:1) to afford the title compound (40 mg, 67.25%) as a yellow solid. m/z (ESI, +ve ion)=154.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.68 (s, 1H), 6.48 (s, 2H), 4.02 (m, 2H), 2.25 (s, 3H), 1.33 (m, 3H). Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-ethoxy-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 5-ethoxy-2-methylpyrimidin-4-amine (26.20 mg, 0.172 mmol, 1.20 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.143 mmol, 1.00 equiv) in toluene (0.5 mL) was added Cs2CO3(92.88 mg, 0.286 mmol, 2 equiv) at room temperature under nitrogen atmosphere. To the above mixture were added XantPhos (16.49 mg, 0.029 mmol, 0.2 equiv) and Pd2(dba)3(26.10 mg, 0.029 mmol, 0.2 equiv) at room temperature. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere then filtered. The filter cake was washed with EA (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM:MeOH (10:1) to afford the title compound (64 mg, 68.37%) as a white solid. m/z (ESI, +ve ion)=657.50. Step C. (1R,2S)-2-{3-[(5-ethoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-ethoxy-2-methylpyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (70 mg, 0.107 mmol, 1 equiv) and DCM (2 mL, 31.460 mmol, 295.16 equiv) was added TFA (0.2 mL, 2.693 mmol, 25.26 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 22% B to 32% B in 8 min, 32% B: wavelength: 254 nm; RT1(min): 7 to afford Example 134 (32.1 mg, 65.91%) as a white solid. m/z (ESI, +ve ion)=457.20 [M+H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.84 (s, 1H), 7.62 (d, J=8.8 Hz, 1H), 7.44 (s, 1H), 6.93 (d, J=8.4 Hz, 1H), 6.85 (d, J=8.4 Hz, 1H), 6.64 (m, 1H), 5.64 (s, 1H), 4.27-4.21 (m, 2H), 3.36-3.39 (m, 4H), 2.31 (s, 3H), 2.27-2.17 (m, 2H), 1.54-1.50 (m, 3H). Example 154. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-(3,6-dihydro-2H-pyran-4-yl)-5-methoxypyrimidin-4-amine The mixture of 2-chloro-5-methoxypyrimidin-4-amine (159.6 mg, 1.000 mmol, 1 equiv) and 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (315.1 mg, 1.500 mmol, 1.50 equiv) was dissolved in 1,4-dioxane (8.5 mL) and water (1.75 mL) under the nitrogen atmosphere. To the solution were added K3PO4(636.8 mg, 3.000 mmol, 3.00 equiv) and Pd(dppf)Cl2·DCM (81.8 mg, 0.100 mmol, 0.10 equiv). The mixture was stirred at 90° C. overnight. After cooled to room temperature, the solvent was removed under reduced pressure. The residue was purified by Prep-TLC (PE/EA=1/10) to afford the title compound (165.7 mg, 79.94%) as a yellow solid. m/z (ESI, +ve ion)=208.30 [M+H]+. Step B. 5-methoxy-2-(tetrahydro-2H-pyran-4-yl)pyrimidin-4-amine To a solution of 2-(3,6-dihydro-2H-pyran-4-yl)-5-methoxypyrimidin-4-amine (165.7 mg, 0.800 mmol, 1 equiv) in MeOH (12 mL) was added 10% Pd/C (110 mg) under nitrogen atmosphere. The reaction system was degassed and purged with hydrogen three times. The mixture was hydrogenated at room temperature for 3 h under hydrogen atmosphere using a hydrogen balloon. The resulting mixture was filtered through a Celite pad and the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM/MeOH/TEA=100/10/1) to afford the title compound (143.2 mg, 85.59%) as a white solid. m/z (ESI, +ve ion)=210.10 [M+H]+. Step C. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-methoxy-2-(tetrahydro-2H-pyran-4-yl)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 0.83 equiv) and 5-methoxy-2-(tetrahydro-2H-pyran-4-yl)pyrimidin-4-amine (40 mg, 0.191 mmol, 1 equiv) were dissolved in toluene (3 mL) under nitrogen atmosphere. To the solution were added Cs2CO3(104.4 mg, 0.320 mmol, 1.68 equiv), XantPhos (19 mg, 0.033 mmol, 0.17 equiv) and Pd2(dba)3(29 mg, 0.032 mmol, 0.17 equiv). The reaction mixture was bubble with nitrogen for 5 minutes. The reaction mixture was then stirred at 90° C. for 2 h. The resulting mixture was filtered, the filter cake was washed with EA (5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EtOAc=1/20) to afford the title compound (75.9 mg, 55.70%) as an orange solid. m/z (ESI, +ve ion)=713.55 [M+H]+. Step D. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclo propane-1,3′-indol]-2′(1′H)-one To a solution of tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-methoxy-2-(tetrahydro-2H-pyran-4-yl)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate (75.9 mg, 0.106 mmol, 1 equiv) in DCM (2 mL) was added TFA (164 uL, 2.208 mmol, 20.74 equiv). The reaction mixture was stirred at room temperature overnight. The solvent was removed under reduced pressure. The residual was azeotropic roto-evaporated with toluene to remove TFA. The crude product was purified by Prep-HPLC: Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3). Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 35% B in 8 min, 35% B; wavelength: 254 nm; RT1(min): 7.67 to afford Example 154 (18.6 mg, 34.08%) as a white solid. m/z (ESI, +ve ion)=513.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.60 (s, 1H), 10.40 (s, 1H), 9.00 (s, 1H), 8.00 (s, 1H), 7.45 (d, J=4.2, 1H), 7.39 (s, 1H), 6.87 (d, J=8.8 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (dd, J=8.4, 2.8 Hz, 1H), 5.66 (d, J=2.8 Hz, 1H), 3.92 (s, 3H), 3.73 (t, J=13.2 Hz, 2H), 3.31-3.18 (m, 5H), 2.68-2.51 (m, 2H), 2.31-2.28 (m, 1H), 2.00-1.97 (m, 1H), 1.66-1.50 (m, 4H). Example 157. (1R,2S)-2-(3-{[2-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-(azetidin-1-yl)-5-methoxypyrimidin-4-amine The mixture of 2-chloro-5-methoxypyrimidin-4-amine (200 mg, 1.253 mmol, 1 equiv) and azetidine (214.69 mg, 3.759 mmol, 3 equiv) in THF (4 mL) was stirred at 60° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 6% B to 15% B in 8 min, 15% B; wavelength: 254 nm; RT1(min): 7) to afford the title compound (60 mg, 21.25%) as a white solid. m/z (ESI, +ve ion)=181.15 [M+H]+. Step B. tert-butyl (1R,2S)-2-(3-{[2-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1-(tert-butoxycarbonyl)indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To the mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (110 mg, 0.174 mmol, 1 equiv) and 2-(azetidin-1-yl)-5-methoxypyrimidin-4-amine (31.39 mg, 0.174 mmol, 1 equiv) in toluene (3 mL) were added Cs2CO3(113.38 mg, 0.348 mmol, 2 equiv). Pd2(dba)3(31.90 mg, 0.035 mmol, 0.2 equiv) and XantPhos (20.16 mg, 0.035 mmol, 0.2 equiv) under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. After cooled to room temperature, the mixture was filtered and washed with EA (20 mL). The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EtOAc in PE to afford the title compound (90 mg, 75.56%) as a yellow oil. m/z (ESI, +ve ion)=684.35 [M+H]+. Step C. (1R,2S)-2-(3-{[2-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one The mixture of tert-butyl (1R,2S)-2-(3-{[2-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1-tert-butoxycarbonyl)indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (90 mg, 0.132 mmol, 1 equiv) in HFIP (5 mL) was stirred at 65° C. for 12 h. The solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 23% B to 33% B in 8 min; wavelength: 254 nm; RT1(min): 7) to afford Example 157 (40 mg, 62.60%) as a white solid. m/z (ESI, +ve ion)=484.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.55 (s, 1H), 10.41 (s, 1H), 8.85 (s, 1H), 7.73 (s, 1H), 7.56 (d, J=8.4 Hz, 1H), 7.36 (s, 1H), 6.86-6.84 (m, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.59-6.56 (m, 1H), 5.65 (d, J=2.4 Hz, 1H), 3.79 (s, 3H), 3.67-3.50 (m, 4H), 3.32 (s, 3H), 3.18 (t, J=8.0 Hz, 1H), 2.32-2.29 (m, 1H), 2.08-1.96 (m, 3H). Example 160. (1R,2S)-2-{3-[(2-ethyl-5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-ethenyl-5-methoxypyrimidin-4-amine To a stirred solution of 2-chloro-5-methoxypyrimidin-4-amine (958 mg, 6.004 mmol, 1 equiv) and 2-ethenyl-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1109.62 mg, 7.205 mmol, 1.2 equiv) in dioxane (50 mL) and water (10 mL) were added Pd(dppf)Cl2·CH2Cl2(489.07 mg, 0.600 mmol, 0.1 equiv) and Na2CO3(1272.63 mg, 12.008 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 100° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was diluted with water (50 mL). The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (3×150 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (350 mg, 38.56%) as a white solid. m/z (ESI, +ve ion)=152.10 [M+H]+. Step B. 2-ethyl-5-methoxypyrimidin-4-amine To a solution of 2-ethenyl-5-methoxypyrimidin-4-amine (350 mg, 2.315 mmol, 1 equiv) in 5 mL MeOH was added Pd/C (10%, 35 mg) under nitrogen atmosphere. The mixture was hydrogenated at room temperature under hydrogen atmosphere for 2 h. The mixture was filtered through a Celite pad and the filtrate was concentrated under reduced pressure. The resulting mixture was filtered, the filter cake was washed with MeOH (3×50 mL). The filtrate was concentrated under reduced pressure to afford the title compound (350 mg, 98.68%) as a white solid. m/z (ESI, +ve ion)=154.10 [M+H]+. Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-ethyl-5-methoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (120 mg, 0.190 mmol, 1 equiv) and 2-ethyl-5-methoxypyrimidin-4-amine (34.93 mg, 0.228 mmol, 1.2 equiv) in toluene (6 mL) were added Pd2(dba)3(17.40 mg, 0.019 mmol, 0.1 equiv), XantPhos (11.00 mg, 0.019 mmol, 0.1 equiv) and Cs2CO3(123.83 mg, 0.380 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was diluted with water (20 mL). The resulting mixture was extracted with EA (3×20 mL). The combined organic layers were washed with brine (3×60 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA=1/1) to afford the title compound (90 mg, 72.11%) as a yellow solid. m/z (ESI, +ve ion)=657.25 [M+H]+. Step D. (1R,2S)-2-{3-[(2-ethyl-5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-ethyl-5-methoxypyrimidin-4-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (85 mg, 0.129 mmol, 1 equiv) in DCM (4 mL) was added TFA (2 mL) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 22% B to 32% B in 8 min, 32% B; wavelength: 254 nm; RT1(min): 7) to afford the title compound (32.8 mg, 54.57%) as a white solid. m/z (ESI+ve ion)=457.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.60 (s, 1H), 10.40 (s, 1H), 8.96 (s, 1H), 7.95 (s, 1H), 7.46 (d, J=8.4 Hz, 1H), 7.39 (s, 1H), 6.88 (d, J=10.4 Hz, 1H), 6.75 (d, J=8.0 Hz, 1H), 6.59 (dd, J=10.8, 2.4 Hz, 1H), 5.69 (d, J=2.4 Hz, 1H), 3.91 (s, 3H), 3.32 (s, 3H), 3.19 (t, J=8.4 Hz, 1H), 2.48-2.42 (m, 2H), 2.34-2.30 (m, 1H), 2.02-1.97 (m, 1H), 1.01 (t, J=7.6 Hz, 3H). Example 161. (1R,2S)-5′-methoxy-2-{3-[(7-methoxyquinolin-6-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(7-methoxyquinolin-6-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 7-methoxyquinolin-6-amine (33.10 mg, 0.190 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (120.00 mg, 0.190 mmol, 1.00 equiv) in toluene (5.00 mL) were added Pd2(dba)3(34.80 mg, 0.038 mmol, 0.20 equiv), XantPhos (21.99 mg, 0.038 mmol, 0.20 equiv) and Cs2CO3(123.83 mg, 0.380 mmol, 2.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered. The filter cake was washed with EA (3×30 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (82.00 mg, 63.67%) as a yellow solid. m/z (ESI, +ve ion)=678.30 [M+H]+. Step B. (1R,2S)-5′-methoxy-2-{3-[(7-methoxyquinolin-6-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(7-methoxyquinolin-6-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.00 mg, 0.118 mmol, 1.00 equiv) in HFIP (5.00 mL) was stirred for 6 h at 60° C. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (48 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 37% B to 45% B in 8 min, 45% B: wavelength: 254 nm; RT1(min): 7.82) to afford the title compound (27.20 mg, 48.02%) as a white solid. m/z (ESI+ve ion)=478.20 [M+H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.55-8.54 (m, 1H), 8.23 (s, 1H), 8.09 (d, J=7.6 Hz, 1H), 7.71 (d, J=8.0 Hz, 1H), 7.41-7.34 (m, 2H), 7.34-7.31 (m, 1H), 6.94 (d, J=8.0 Hz, 1H), 6.85 (d, J=8.4 Hz, 1H), 6.65-6.62 (m, 1H), 5.64 (d, J=2.4 Hz, 1H), 4.17 (s, 3H), 3.40-3.36 (m, 1H), 3.31 (s, 3H), 2.27-2.18 (m, 2H). Example 163. (1R,2S)-5′-methoxy-2-{3-[(3-methoxyquinolin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 3-methoxyquinolin-2-amine The mixture of 3-methoxyquinoline (500 mg, 3.141 mmol, 1 equiv) and m-CPBA (2092.12 mg, 12.124 mmol, 3.86 equiv) in CHCl3(9 mL) was stirred for 5 h at 25° C. under nitrogen atmosphere. The mixture was diluted with EtOAc (30 mL) and washed with water (30 mL), brine (30 mL). The organic layer was dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-100% of EtOAc in PE to give 840 mg of intermediate. The mixture of intermediate and benzoyl isocyanate (1386.41 mg, 9.423 mmol, 3 equiv) in DCM (9 mL) was stirred for 1 h at 55° C. After cooled to room temperature, the solvent was removed under reduced pressure. The residue was dissolved in MeOH (9 mL) and to this mixture was added sodium methoxide (848.43 mg, 15.705 mmol, 5 equiv) at 25° C. The resulting mixture was stirred at 75° C. for 2 h. After cooled to room temperature, the solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-10% of MeOH in DCM to afford the title compound (200 mg, 36.55%) as a white solid. m/z (ESI, +ve ion)=175.10 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-methoxyquinolin-2-yl)amino]indazol-4-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1 equiv) and 3-methoxyquinolin-2-amine (33.10 mg, 0.19) mmol, 1.2 equiv) in toluene (2.5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. After cooled to room temperature, the mixture was filtered and washed with EA (20 mL). The filtrate was concentrated in vacuo and the residue was purified by silica gel column, eluted with 0-100% of EtOAc in PE to give the title compound (85 mg, 79.19%) as a yellow solid. m/z (ESI, +ve ion)=678.30 [M+H]+. Step C. (1R,2S)-5′-methoxy-2-{3-[(3-methoxyquinolin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one The mixture of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-[(3-methoxyquinolin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (85 mg, 0.125 mmol, 1 equiv) in HFIP (5 mL) was stirred at 60° C. for 12 h. After cooled to 25° C., the solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 35% B to 45% B in 8 min-; wavelength: 254 nm; RT1(min): 7) to afford the title compound (33.6 mg, 56.05%) as a light yellow solid. m/z (ESI, +ve ion)=478.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.53 (s, 1H), 10.40 (s, 1H), 8.72 (s, 1H), 7.69 (d, J=7.6 Hz, 1H), 7.62 (d, J=8.4 Hz, 1H), 7.54 (s, 1H), 7.39 (s, 1H), 7.36-7.30 (m, 2H), 7.26-7.22 (m, 1H), 6.89 (d, J=8.8 Hz, 1H), 6.76 (d, J=8.4 Hz, 1H), 6.63-6.60 (m, 1H), 5.77 (d, J=2.4 Hz, 1H), 4.02 (s, 3H), 3.38 (s, 3H), 3.20 (t, J=8.0 Hz, 1H), 2.35-2.32 (m, 1H), 2.01-1.98 (m, 1H). Example 167. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(pyrrolidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-methoxy-2-(pyrrolidin-1-yl)pyrimidin-4-amine A solution of 2-chloro-5-methoxypyrimidin-4-amine (500 mg, 3.133 mmol, 1 equiv) in pyrrolidine (4.46 g, 62.660 mmol, 20 equiv) in sealed tube was stirred for 4 h at 80° C. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under vacuum. The residue was dissolved in DMF (0.5 mL). The residue was purified by reverse flash chromatography with the following conditions: Column: C18 Column, 40 g, 60 Å, 40-60 μm; Mobile Phase A: 10 mM aq. NH4HCO3, Mobile Phase B: MeCN; Flow rate: 60 mL/min; Gradient: 0% B to 0% B in 5 min, 0% B to 50% B in 30 min; Detector: UV 254 & 220 nm. The resulting mixture was concentrated under vacuum. This resulted in the title compound (350 mg, 50.03%) as a white solid. m/z (ESI, +ve ion)=195.10 [M+H]+. Step B. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((5-methoxy-2-(pyrrolidin-1-yl)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a stirred mixture of Pd2(dba)3(34.80 mg, 0.038 mmol, 0.2 equiv) and XantPhos (21.99 mg, 0.038 mmol, 0.2 equiv) in toluene (5 mL) were added Cs2CO3(123.83 mg, 0.380 mmol, 2 equiv), 5-methoxy-2-(pyrrolidin-1-yl)pyrimidin-4-amine (44.29 mg, 0.228 mmol, 1.20 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (120 mg, 0.190 mmol, 1.00 equiv) at 25° C. The resulting mixture was stirred for additional 2 h at 80° C. under nitrogen atmosphere. The mixture was allowed to cool down to 25° C. The resulting mixture was filtered, the filter cake was washed with DCM (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (EA/PE=1/1) to afford the title compound (90 mg, 66.51%) as a grey solid. m/z (ESI, +ve ion)=698.25 [M+H]+. Step C. (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(pyrrolidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one To a stirred solution of tert-butyl 6-{[1-(tert-butoxycarbonyl)-5-methoxy-2-oxo-3H-indol-3-yl]methyl}-3-{[5-methoxy-2-(pyrrolidin-1-yl)pyrimidin-4-yl]amino}indazole-1-carboxylate (90 mg, 0.043 mmol, 1 equiv) in DCM (1 mL) was added TFA (0.2 mL) dropwise at 25° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 25° C. under nitrogen atmosphere. The resulting mixture was diluted with toluene (2 mL). The resulting mixture was concentrated under vacuum. The crude product (80 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 um; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 43% B in 8 min, 43% B: wavelength: 254 nm; RT1(min): 7.3 to afford the title compound (25.5 mg, 39.77%) as a white solid. m/z (ESI, +ve ion)=498.15 [M+H]+.1H NMR (400 MHz, DMSO-d) δ 12.53 (s, 1H), 10.40 (s, 1H), 8.74 (s, 1H), 7.75 (s, 1H), 7.56 (d, J=8.4 Hz, 1H), 7.37 (s, 1H), 6.85 (dd. J=8.5, 1.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.4, 2.6 Hz, 1H), 5.63 (d, J=2.5 Hz, 1H), 3.78 (s, 3H), 3.29 (s, 3H), 3.19 (t, J=8.4 Hz, 1H), 3.11 (s, 4H), 2.29 (dd. J=7.9, 4.7 Hz, 1H), 1.97 (dd, J=9.0, 4.6 Hz, 1H), 1.68 (s, 4H). Example 169. (1R,2S)-2-[3-({2-[(3R)-3-fluoropyrrolidin-1-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-[(3R)-3-fluoropyrrolidin-1-yl]-5-methoxypyrimidin-4-amine To a stirred mixture of 2-chloro-5-methoxypyrimidin-4-amine (159 mg, 0.996 mmol, 1.00 equiv) and (3R)-3-fluoropyrrolidine (177.59 mg, 1.992 mmol, 2 equiv) in dioxane (0.5 mL) was added TEA (403.33 mg, 3.984 mmol, 4 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 100° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The residue was purified by reverse phase flash with the following conditions (column, C18; mobile phase, water (5 mM in NH4HCO3) in MeCN, 2% to 50% gradient in 30 min; detector, UV 254/210 nm.) to afford the title compound (130 mg, 61.47%) as a white solid. m/z (ESI, +ve ion)=213.10 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({2-[(3R)-3-fluoropyrrolidin-1-yl]-5-methoxypyrimidin-4-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodo-4,5-dihydroindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 2-[(3R)-3-fluoropyrrolidin-1-yl]-5-methoxypyrimidin-4-amine (40.20 mg, 0.190 mmol, 1.2 equiv) in toluene (5 mL) were added Cs2CO3(102.87 mg, 0.316 mmol, 2 equiv), XantPhos (18.27 mg, 0.032 mmol, 0.2 equiv) and Pd2(dba)3(28.91 mg, 0.032 mmol, 0.2 equiv) at room temperature. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×30 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (1/1) to afford the title compound (102 mg, 90.27%) as a yellow solid. m/z (ESI, +ve ion)=716.20 [M+H]+ Step C. (1R,2S)-2-[3-({2-[(3R)-3-fluoropyrrolidin-1-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({2-[(3R)-3-fluoropyrrolidin-1-yl]-5-methoxypyrimidin-4-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (92 mg, 0.129 mmol, 1 equiv) in TFA (1 mL) and DCM (5 mL) was stirred at room temperature for 1 h. The resulting mixture was concentrated under reduced pressure. The crude product (110 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmoL/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 35% B in 8 min, 35% B; wavelength: 254 nm; RT1(min): 7) to afford Example 169 (33.2 mg, 50.10%) as a white solid. m/z (ESI, +ve ion)=516.20 [M+H]+.1H NMR (400 MHz, DMSO-d) δ 12.56 (s, 1H), 10.40 (s, 1H), 8.85 (s, 1H), 7.77 (s, 1H), 7.54 (d, J=8.4 Hz, 1H), 7.38 (s, 1H), 6.85 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.58-6.60 (m, 1H), 5.63 (d, J=2.5 Hz, 1H), 5.16 (d, J=53.9 Hz, 1H), 3.80 (s, 3H), 3.30-3.48 (m, 6H), 3.23-3.05 (m, 2H), 2.29-2.31 (m, 1H), 1.87-1.92 (m, 3H). Example 170. (1R,2S)-2-(3-{[2-(3,3-difluoropyrrolidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-(3,3-difluoropyrrolidin-1-yl)-5-methoxypyrimidin-4-amine To a stirred mixture of 2-chloro-5-methoxypyrimidin-4-amine (159 mg, 0.996 mmol, 1.00 equiv) and 3,3-difluoropyrrolidine hydrochloride (286.09 mg, 1.992 mmol, 2 equiv) in dioxane (0.5 mL) was added TEA (403.33 mg, 3.984 mmol, 4 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 100° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The residue was purified by reverse phase flash with the following conditions (column, C18; mobile phase, water (5 mM NH4HCO3) in MeCN, 2% to 50% gradient in 30 min; detector, UV 254/210 nm.) to afford the title compound (184 mg, 80.21%) as a white solid. m/z (ESI, +ve ion)=231.05 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(3,3-difluoropyrrolidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and 2-(3,3-difluoropyrrolidin-1-yl)-5-methoxypyrimidin-4-amine (43.75 mg, 0.190 mmol, 1.2 equiv) in toluene (5 mL) were added Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) and Pd(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×30 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (130 mg, 78.31%) as a yellow solid. m/z (ESI, +ve ion)=734.25 [M+H]+. Step C. (1R,2S)-2-(3-{[2-(3,3-difluoropyrrolidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(3,3-difluoropyrrolidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (120 mg, 0.114 mmol, 1 equiv, 70%) in TFA (1 mL) and DCM (5 mL) was stirred for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (130 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 45% B in 8 min, 45% B; wavelength: 254 nm; RT1(min): 7.2) to afford the title compound (38.3 mg, 62.71%) as a white solid. m/z (ESI, +ve ion)=534.30 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.59 (s, 1H), 10.41 (s, 1H), 8.97 (s, 1H), 7.79 (s, 1H), 7.53 (d, J=8.4 Hz, 1H), 7.38 (s, 1H), 6.87 (d, J=8.8 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.57-6.60 (m, 1H), 5.62 (d, J=2.5 Hz, 1H), 3.81 (s, 3H), 3.57 (t, J=13.4 Hz, 2H), 3.32-3.30 (m, 5H), 3.20 (t, J=8.4 Hz, 1H), 2.33-2.17 (m, 3H), 1.96-2.00 (m, 1H). Example 173. (1R,2S)-2-(3-{[2-(3,3-difluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-(3,3-difluoroazetidin-1-yl)-5-methoxypyrimidin-4-amine To a stirred mixture of 3,3-difluoroazetidine hydrochloride (258.13 mg, 1.992 mmol, 2 equiv) and 2-chloro-5-methoxypyrimidin-4-amine (159 mg, 0.996 mmol, 1.00 equiv) in dioxane (1 mL) was added TEA (604.99 mg, 5.976 mmol, 6 equiv) under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 25° C. under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The resulting mixture was extracted with EA (3×30 mL). The combined organic layers were washed with brine (3×10 mL), dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (1/2) to afford the title compound (100 mg, 46.42%) as a grey solid. m/z (ESI, +ve ion)=217.10 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(3,3-difluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 2-(3,3-difluoroazetidin-1-yl)-5-methoxypyrimidin-4-amine (41.08 mg, 0.190 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) in toluene (5.0 mL) were added XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv), Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv) and Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (78 mg, 68.43%) as a yellow oil. m/z (ESI, +ve ion)=720.20 [M+H]+. Step C. (1R,2S)-2-(3-{[2-(3,3-difluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(3,3-difluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (78 mg, 0.106 mmol, 1 equiv) in HFIP (5 mL) was stirred at 60° C. for 12 h. The solvent was removed in vacuo. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 19×250 mm, 10 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 50% B in 8 min, 50% B; wavelength: 254 nm; RT1(min): 7.8 to afford Example 173 (27.6 mg, 50.13%) as a white solid. m/z (ESI, +ve ion)=520.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.69 (s, 1H), 7.59 (d, J=8.8 Hz, 1H), 7.47 (s, 1H), 6.88-6.83 (m, 2H), 6.63-6.61 (m, 1H), 5.60 (s, 1H), 4.12-3.94 (m, 4H), 3.75 (s, 3H), 3.51-3.49 (m, 1H), 3.39-3.13 (m, 3H), 2.29-2.25 (m, 11H), 2.21-2.17 (m, 1H). Example 174. (1R,2S)-2-(3-{[2-(3-fluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-(3-fluoroazetidin-1-yl)-5-methoxypyrimidin-4-amine To a stirred mixture of 3-fluoroazetidine hydrochloride (223.68 mg, 2.006 mmol, 2 equiv) and 2-chloro-5-methoxypyrimidin-4-amine (160 mg, 1.003 mmol, 1.00 equiv) in dioxane (1 mL) was added TEA (608.79 mg, 6.018 mmol, 6 equiv) under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 100° C. The resulting mixture was cool down to room temperature and then concentrated under vacuum. The resulting mixture was extracted with EA (3×30 mL). The combined organic layers were washed with brine (3×10 mL), dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (1/2) to afford the title compound (110 mg, 50.92%) as a grey solid. m/z (ESI, +ve ion)=199.10 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(3-fluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 2-(3-fluoroazetidin-1-yl)-5-methoxypyrimidin-4-amine (31.39 mg, 0.158 mmol, 1 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) in toluene (5.0 mL) were added XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv), Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv) and Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (72.0 mg, 64.79%) as a yellow oil. m/z (ESI, +ve ion)=702.20 [M+H]+. Step C. (1R,2S)-2-(3-{[2-(3-fluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(3-fluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (72 mg, 0.104 mmol, 1 equiv) in HFIP (5 mL) was stirred at 60° C. for 12 h. The solvent was removed in vacuo. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 19×250 mm, 10 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 50% B in 8 min, 50% B; wavelength: 254 nm; RT1(min): 7.8 to afford Example 174 (22 mg, 42.00%) as a white solid. m/z (ESI, +ve ion)=502.30 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 7.64 (d, J=8.8 Hz, 2H), 7.45 (s, 1H), 6.87-6.83 (m, 2H), 6.64-6.61 (m, 1H), 5.61 (d, J=2.4 Hz, 1H), 5.27-5.11 (m, 1H), 4.08-3.95 (m, 2H), 3.92 (s, 3H), 3.85-3.73 (m, 2H), 3.39-3.37 (m, 1H), 3.34 (s, 3H), 2.28-2.25 (m, 1H), 2.21-2.17 (m, 1H). Example 183: (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(morpholine-4-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A: (6-methoxy-5-nitro-2-pyridyl)-morpholino-methanone To a solution of 6-methoxy-5-nitro-2-pyridinecarboxylic acid (100 mg, 0.5 mmol) and HATU (230 mg, 0.61 mmol) in DCM (3 mL) was added morpholine (0.07 mL, 0.76 mmol) and N,N-diisopropylethylamine (0.22 mL, 1.26 mmol) and the reaction mixture was stirred at room temperature for 16 h. The reaction mixture was quenched by the addition of saturated, aq. sodium bicarbonate and extracted with DCM (3×). The combined organic layers were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EA in hexanes) to afford the product as a yellow solid (134 mg, 99%). m/z (ESI, +ve ion)=268.1 [M+H]+. Step B: (5-amino-6-methoxy-2-pyridyl)-morpholino-methanone To a solution of (6-methoxy-5-nitro-2-pyridyl)-morpholino-methanone (134 mg, 0.50 mmol) and ammonium formate (316 mg, 5.0 mmol) in ethanol (3.3 mL) under argon was added 10% palladium on carbon (107 mg, 0.1 mmol) and the reaction mixture was heated to 50° C. for 16 h. The reaction mixture was filtered through celite and concentrated in vacuo to give the desired product as a white foam (118 mg, 99%). m/z (ESI, +ve ion)=238.1 [M+H]+. Step C: tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[2-methoxy-6-(morpholine-4-carbonyl)-3-pyridyl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a solution of (5-amino-6-methoxy-2-pyridyl)-morpholino-methanone (41 mg, 0.17 mmol) in dry toluene (1.6 mL) was added tert-butyl (1R,2S)-2-(1-tert-butoxycarbonyl-3-iodo-indazol-6-yl)-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (100 mg, 0.16 mmol), Xantphos Pd G4 (15 mg, 0.02 mmol), Xantphos (9.2 mg, 0.02 mmol), and cesium carbonate (155 mg, 0.48 mmol). Argon was bubbled through the solution for 3 min then the reaction mixture was heated to 90° C. for 75 min. The reaction mixture was diluted with DCM, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% hexanes in acetone) to afford the product as a yellow foam (83.5 mg, 71%). m/z (ESI, +ve ion)=741.2 [M+H]+. Step D: (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(morpholine-4-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one A solution of tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[[2-methoxy-6-(morpholine-4-carbonyl)-3-pyridyl]amino]indazol-6-yl]-5′-methoxy-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (83.5 mg, 0.11 mmol) in hexafluoroisopropanol (3 mL) was heated to 70° C. for 16 h. The reaction mixture was concentrated in vacuo and the crude residue was purified by RP-HPLC using 10-70% ACN/water (10 mmol/L ammonium bicarbonate) to give the title compound as a white lyophilized solid (40.7 mg, 67%). m/z (ESI, +ve ion)=541.3 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.31 (s, 1H), 10.44 (s, 1H), 8.51-8.34 (m, 2H), 7.99-7.87 (m, 1H), 7.43-7.26 (m, 2H), 6.98-6.85 (m, 1H), 6.82-6.69 (m, 1H), 6.65-6.52 (m, 1H), 5.70 (s, 1H), 3.99 (s, 3H), 3.82-3.58 (m, 8H), 3.32 (s, 3H), 3.25-3.13 (m, 1H), 2.41-2.27 (m, 1H), 2.05-1.92 (m, 1H). Example 185: (1R,2S)-2-(3-{[6-(4,4-difluoropiperidine-1-carbonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one The title compound was prepared using the procedure for Example 183 starting from 6-methoxy-5-nitro-2-pyridinecarboxylic acid and 4,4-difluoropiperidine hydrochloride. m/z (ESI, +ve ion)=575.2 [M+H]+. 1H NMR (400 MHz, DMSO) δ 12.32 (s, 1H), 10.44 (d, J=3.4 Hz, 1H), 8.50-8.37 (m, 2H), 7.99-7.86 (m, 1H), 7.41-7.30 (m, 2H), 6.96-6.87 (m, 1H), 6.81-6.70 (m, 1H), 6.64-6.55 (m, 1H), 5.71 (s, 1H), 4.01 (s, 3H), 3.92-3.66 (m, 4H), 3.32 (s, 3H), 3.24-3.12 (m, 1H), 2.40-2.30 (m, 1H), 2.22-2.02 (m, 4H), 2.02-1.93 (m, 1H). Example 189. (1R,2S)-2-(3-{[2-(4,4-difluoropiperidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 2-(4,4-difluoropiperidin-1-yl)-5-methoxypyrimidin-4-amine To a stirred solution of 2-chloro-5-methoxypyrimidin-4-amine (159 mg, 0.996 mmol, 1 equiv) in dioxane (0.5 mL) was added TEA (0.83 mL, 5.976 mmol, 6 equiv) and 4,4-difluoropiperidine hydrochloride (314.05 mg, 1.992 mmol, 2 equiv) under nitrogen atmosphere. The resulting mixture was stirred for 16 hours at 100° C. The reaction was quenched by the addition of water (5 mL) at 25° C. The resulting mixture was extracted with DCM (3×5 mL). The combined organic layers were washed with brine (3×5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM/MeOH=10/1) to afford the title compound (116 mg, 47.66%) as white solid. m/z (ESI, +ve ion)=245.10 [M+H]+. Step B. tert-butyl (1R,2S)-2-(1-tert-butoxycarbonyl)-3-((2-(4,4-difluoropiperidin-1-yl)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a stirred solution of 2-(4,4-difluoropiperidin-1-yl)-5-methoxypyrimidin-4-amine (46.41 mg, 0.190 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) in toluene (1 mL) were added Pd2(dba)3(14.50 mg, 0.016 mmol, 0.1 equiv), XantPhos (9.16 mg, 0.016 mmol, 0.1 equiv) and Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv) under nitrogen atmosphere. The final reaction mixture was reacted for 2 hours at 90° C. The reaction was quenched by the addition of water (10 mL) at 25° C. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (3×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (EtOAc/PE=1/1) to afford the title compound (77 mg, 65.02%) as a yellow solid. m/z (ESI, +ve ion)=748.30 [M+H]+. Step C. (1R,2S)-2-(3-{[2-(4,4-difluoropiperidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[2-(4,4-difluoropiperidin-1-yl)-5-methoxypyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (77 mg, 0.103 mmol, 1 equiv) in HFIP (2 mL) was stirred for 16 hours at 60° C. The resulting mixture was concentrated under reduced pressure. The crude product (55.8 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 31% B to 41% B in 8 min, 41% B; wavelength: 254 nm; RT1(min): 7) to afford the title compound (11.2 mg, 19.86%) as a white solid. m/z (ESI, +ve ion)=548.10 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.60 (s, 1H), 10.40 (s, 1H), 9.01 (s, 1H), 7.77 (s, 1H), 7.44-7.43 (d, J=4 Hz, 2H), 6.81-6.74 (m, 2H), 6.59-6.56 (m, 1H), 5.63 (d, J=4 Hz, 1H), 3.81 (s, 3H), 3.44-3.37 (m, 4H), 3.34 (s, 3H), 3.20 (t, J=8 Hz, 1H), 2.33-2.30 (m, 1H), 1.99-1.96 (m, 1H), 1.72-1.68 (m, 2H), 1.53-1.50 (m, 2H). Example 193. (1R,2S)-2-(3-{[5-(methanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 3-methoxy-5-(methylsulfonyl)pyridin-2-amine To a stirred solution of 5-bromo-3-methoxypyridin-2-amine (203 mg, 1.000 mmol, 1 equiv) and (2S,4R)-4-hydroxy-N-(2-methylnaphthalen-1-yl)pyrrolidine-2-carboxamide (HMNPC, 27.03 mg, 0.100 mmol, 0.1 equiv) in DMSO (3 mL) were added CuI (19.04 mg, 0.100 mmol, 0.1 equiv), sodium methanesulfinate (132.69 mg, 1.300 mmol, 1.3 equiv) and K3PO4(212.22 mg, 1.000 mmol, 1 equiv) at 25° C. under nitrogen atmosphere. The resulting mixture was stirred for additional 24 h at 120° C. The mixture was allowed to cool down to 25° C. The residue was purified by reverse flash chromatography with the following conditions: column, C18 Column; mobile phase, water (5 mM NH4HCO3) in ACN, 20% to 60% gradient in 30 min; detector, UV 254 nm. The resulting mixture was concentrated under reduced pressure to afford the title compound (198.4 mg, 98.12%) as a yellow solid. m/z (ESI, +ve ion)=203.10 [M+H]+. Step B. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-methanesulfonyl-3-methoxypyridin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 5-methanesulfonyl-3-methoxypyridin-2-amine (38.43 mg, 0.190 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) in toluene (3 mL) were added Pd2(dba)3(14.50 mg, 0.016 mmol, 0.1 equiv), XantPhos (9.16 mg, 0.016 mmol, 0.1 equiv) and Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv) under nitrogen atmosphere. The final reaction mixture was stirred at 90° C. for 2 hours. The mixture was allowed to cool down to 25° C. The reaction was quenched by the addition of water (10 mL) at 25° C. The resulting mixture was extracted with DCM (3×10 mL). The combined organic layers were washed with brine (3×10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (EA/PE=1/1) to afford the title compound (108.6 mg, 97.17%) as a yellow solid. m/z (ESI, +ve ion)=706.10 [M+H]+. Step C. (1R,2S)-2-(3-{[5-(methanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-methanesulfonyl-3-methoxypyridin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (130 mg, 0.184 mmol, 1 equiv) in HFIP (2 mL, 18.996 mmol) was stirred for 16 hours at 60° C. The resulting mixture was concentrated under reduced pressure. The crude product (96.8 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep Phenyl OBD Column, 19×250 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 40% B in 10 min, 40% B; wavelength: 254 nm; RT1(min): 9) to afford the title compound (25.7 mg, 27.60%) as a white solid. m/z (ESI, +ve ion)=506.20 [M+H]+.1H-NMR (4 (0) MHz, DMSO-d6) δ 12.67 (s, 1H), 10.42 (s, 1H), 9.14 (s, 1H), 7.96 (s, 1H), 7.50 (s, 1H), 7.41-7.38 (m, 2H), 6.89 (d, J=16 Hz, 1H), 6.75 (d, J=8 Hz, 1H), 6.60-6.57 (m, 1H), 5.72 (d, J=4 Hz, 1H), 3.99 (s, 3H), 3.33 (s, 3H), 3.21 (s, 3H), 3.19-3.17 (t, J=8 Hz, 1H), 2.34-2.31 (m, 1H), 2.08-1.97 (m, 1H). Example 194. 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methyl-N-(propan-2-yl)pyridine-2-carboxamide Step A. N-isopropyl-6-methoxy-N-methyl-5-nitropyridine-2-carboxamide To a solution of 6-methoxy-5-nitropyridine-2-carboxylic acid (50 mg, 0.252 mmol, 1 equiv) in THF (4 mL) at 0° C. was added dropwise oxalyl chloride (39 mg, 0.307 mmol, 1.22 equiv) followed by one drop of DMF. The mixture was stirred for 2 h at 25° C. under nitrogen atmosphere. The resulting mixture was concentrated in vacuo under nitrogen atmosphere to afford the acid chloride which was used for next step without further purification. To a stirred solution of N-methylpropan-2-amine (25 mg, 0.342 mmol, 1.35 equiv) and DIEA (100 mg, 0.774 mmol, 3.07 equiv) in THF (1 mL) at 0° C. was added dropwise the solution of acid chloride in THF (1 mL) under nitrogen atmosphere. The mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was diluted with water (10 mL) and extracted with EA (2×30 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford the title compound (45 mg, 70.41%) as a light yellow oil. m/z (ESI, +ve ion)=254.05 [M+H]+. Step B. 5-amino-N-isopropyl-6-methoxy-N-methylpyridine-2-carboxamide Into a 25 mL round-bottom flask were added N-isopropyl-6-methoxy-N-methyl-5-nitropyridine-2-carboxamide (50 mg, 0.197 mmol, 1 equiv) and MeOH (2 mL) at 25° C. To the above mixture was added Pd/C (25 mg) at 25° C. under nitrogen atmosphere. The resulting mixture was degassed and purged with hydrogen for three times. The resulting mixture was stirred for 4 h at 25° C. under hydrogen atmosphere. The resulting mixture was filtered and the filter cake was washed with MeOH (3×10 mL). The filtrate was concentrated under reduced pressure to afford the title compound (37 mg, 83.94%) as a grey solid. m/z (ESI, +ve ion)=224.15 [M+H]+. Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({6-[isopropyl(methyl)carbamoyl]-2-methoxypyridin-3-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.127 mmol, 1 equiv) and 5-amino-N-isopropyl-6-methoxy-N-methylpyridine-2-carboxamide (31 mg, 0.139 mmol, 1.10 equiv) in toluene (2.5 mL) were added Cs2CO3(80 mg, 0.246 mmol, 1.94 equiv), Pd2(dba); (24 mg, 0.026 mmol, 0.21 equiv) and XantPhos (16 mg, 0.028 mmol, 0.22 equiv). After stirring for 2 h at 90° C. under a nitrogen atmosphere. The resulting mixture was filtered and the filter cake was washed with EA (3×50 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA=1/1) to afford the title compound (60 mg, 65.16%) as a light yellow solid. m/z (ESI, +ve ion)=727.40 [M+H]+ Step D. 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methyl-N-(propan-2-yl)pyridine-2-carboxamide Into a 8 mL vial were added tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({6-[isopropyl(methyl)carbamoyl]-2-methoxypyridin-3-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (60 mg, 0.083 mmol, 1 equiv) and HFIP (1 mL, 9.498 mmol) at 25° C. The mixture was stirred for 12 h at 60° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 34% B to 32% B in 8 min, 32% B; wavelength: 254 nm; RT1 (min): 7) to afford the title compound (25 mg, 57.51%) as a white solid. m/z (ESI, +ve ion)=527.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.27 (s, 1H), 10.43 (s, 1H), 8.40 (d, J=8.0 Hz, 1H), 8.31 (s, 1H), 7.94 (d, J=8.0 Hz, 1H), 7.35 (s, 1H), 7.20 (s, 1H), 6.92 (d, J=8.0 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.71 (d, J=2.4 Hz, 1H), 4.70-4.31 (m, 1H), 4.01 (s, 3H), 3.20 (s, 3H), 3.18 (t, J=8.4 Hz, 1H), 2.94-2.91 (m, 1H), 2.88-2.86 (m, 2H), 2.33-2.28 (m, 1H), 2.00-1.97 (m, 1H), 1.18 (s, 6H). Example 195. (1R,2S)-2-(3-{[5-ethoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-amino-2-chloropyrimidin-5-ol To a stirred mixture of 2-chloro-5-methoxypyrimidin-4-amine (318 mg, 1.993 mmol, 1 equiv) in DCE (10 mL) was added BBr3(1.88 mL, 19.930 mmol, 10 equiv) dropwise at room temperature under nitrogen atmosphere. After the resulting mixture was stirred for 16 h at room temperature under nitrogen atmosphere, 20 mL of DCE was added into the mixture and then stand for hours. Then the upper solution was removed. The deposited semi-solid was dissolved in MeOH (10 mL) and concentrated under reduced pressure to afford the title compound (480 mg, 63.82%) as a yellow solid. m/z (ESI, +ve ion)=145.95 [M+H]+ Step B. 2-chloro-5-ethoxypyrimidin-4-amine To a stirred mixture of 4-amino-2-chloropyrimidin-5-ol (500 mg, 1.374 mmol, 1 equiv, 40%) and Cs2CO3(1343.12 mg, 4.122 mmol, 3 equiv) in acetone (10 mL) was added iodoethane (171.45 mg, 1.099 mmol, 0.8 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for additional 16 h at 60° C. The resulting mixture was diluted with water (10 mL). The resulting mixture was extracted with EA (3×10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (146 mg, 61.20%) as a light yellow solid. m/z (ESI, +ve ion)=174.00 [M+H]+. Step C. 5-ethoxy-2-(methylsulfanyl)pyrimidin-4-amine To a stirred mixture of 2-chloro-5-ethoxypyrimidin-4-amine (120 mg, 0.691 mmol, 1 equiv) in DMF (2 mL) were added sodium thiomethoxide (290.65 mg, 4.146 mmol, 6 equiv) and TEA (629.54 mg, 6.219 mmol, 9 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for additional 4 h at 60° C. The resulting mixture was diluted with water (5 mL). The resulting mixture was extracted with EA (3×10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (52 mg, 39.39%) as a light yellow solid. m/z (ESI, +ve ion)=186.00 [M+H]+. Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-ethoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 5-ethoxy-2-(methylsulfanyl)pyrimidin-4-amine (35.20 mg, 0.190 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) in toluene (2.5 mL) were added Pd2(dba)3(14.50 mg, 0.016 mmol, 0.1 equiv), Cs2CO3(154.79 mg, 0.474 mmol, 3 equiv) and XantPhos (9.16 mg, 0.016 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for additional 2 h at 60° C. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (100 mg, 90.76%) as a yellow solid. m/z (ESI, +ve ion)=689.35 [M+H]+. Step E. (1R,2S)-2-(3-{[5-ethoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-ethoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.145 mmol, 1 equiv) in HFIP (5 mL) was stirred for 12 h at 60° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC-Actus Triart C18 ExRS, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 36% B to 36% B in 10 min, 36% B: wavelength: 254 nm; RT1(min): 10.2) to afford the title compound (18.5 mg, 26.03%) as a white solid. m/z (ESI, +ve ion)=489.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.69 (s, 1H), 10.40 (s, 1H), 9.17 (s, 1H), 7.93 (s, 1H), 7.42 (d, J=8.4 Hz, 2H), 6.91 (d, J=8.8 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.69 (d, J=2.0 Hz, 1H), 4.17-4.12 (m, 2H), 3.32 (s, 3H) 3.20 (t, J=8.8 Hz, 1H), 2.34-2.30 (m, 1H), 2.08 (s, 3H), 1.99 (t, J=4.8 Hz, 1H), 1.39 (t, J=8.0, 7.5 Hz, 3H). Example 203. 5-ethoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-carboxamide Step A. ethyl 5-ethoxy-6-nitropyridine-3-carboxylate To a stirred solution of 5-hydroxy-6-nitropyridine-3-carboxylic acid (200.00 mg, 1.086 mmol, 1.00 equiv) and K2CO3(375.34 mg, 2.715 mmol, 2.50 equiv) in DMF (2.00 mL) was added iodoethane (423.57 mg, 2.715 mmol, 2.50 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 24 h at 60° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was diluted with water (50 mL) and extracted with EA (3×60 mL). The combined organic layers were washed with brine (3×30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (140.00 mg, 53.65%) as a yellow solid. m/z (ESI, +ve ion)=241.10 [M+H]+. Step B. 5-ethoxy-6-nitropyridine-3-carboxylic acid To a stirred mixture of ethyl 5-ethoxy-6-nitropyridine-3-carboxylate (140.00 mg, 0.583 mmol, 1.00 equiv) in THF (2.00 mL), MeOH (2.00 mL) and water (1.00 mL) was added LiOH (27.92 mg, 1.166 mmol, 2.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 0.5 h at room temperature under nitrogen atmosphere. The mixture was acidified to pH 5 with 2 M aqueous of HCl. The resulting mixture was extracted with EA (3×50 mL). The combined organic layers were washed with brine (3×30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (5/1) to afford the title compound (85.00 mg, 68.74%) as a yellow solid. m/z (ESI, +ve ion)=213.10 [M+H]+. Step C. 5-ethoxy-N,N-dimethyl-6-nitropyridine-3-carboxamide To a stirred solution of 5-ethoxy-6-nitropyridine-3-carboxylic acid (75.00 mg, 0.354 mmol, 1.00 equiv) and dimethylamine hydrochloride (28.83 mg, 0.354 mmol, 1.00 equiv) in DMF (4.00 mL) were added HATU (268.83 mg, 0.708 mmol, 2.00 equiv) and DIEA (182.76 mg, 1.416 mmol, 4.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at room temperature under nitrogen atmosphere. The mixture was purified by reverse flash chromatography with the following conditions: C18 Column, 40 g, 60 Å, 40-60 μm; Mobile Phase A: 10 mM aq. NH4HCO3, Mobile Phase B: MeCN; Flow rate: 60 mL/min; Gradient: 0% B to 0% B in 5 min, 0% B to 40% B in 30 min; Detector: UV 254 & 220 nm to afford the title compound (60.00 mg, 70.95%) as a yellow solid. m/z (ESI, +ve ion)=240.15 [M+H]+. Step D. 6-amino-5-ethoxy-N,N-dimethylpyridine-3-carboxamide To a solution of 5-ethoxy-N,N-dimethyl-6-nitropyridine-3-carboxamide (55.00 mg, 0.230 mmol, 1.00 equiv) in MeOH (5.00 mL) was added Pd/C (10%, 24.47 mg) under nitrogen atmosphere. The mixture was degassed and purged with H2for three times. The mixture was hydrogenated at room temperature for 2 h under hydrogen atmosphere. The resulting mixture was filtered through a Celite pad and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (5/1) to afford the title compound (40.00 mg, 83.15%) as a yellow solid. m/z (ESI, +ve ion)=210.15 [M+H]+. Step E. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(dimethylcarbamoyl)-3-ethoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 6-amino-5-ethoxy-N,N-dimethylpyridine-3-carboxamide (33.14 mg, 0.158 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100.00 mg, 0.158 mmol, 1.00 equiv) in toluene (5.00 mL) were added Pd2(dba)3(29.00 mg, 0.032 mmol, 0.20 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.20 equiv) and Cs2CO3(103.19 mg, 0.316 mmol, 2.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered, the filter cake was washed with EA (3×30 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (80.00 mg, 70.87%) as a yellow solid. m/z (ESI, +ve ion)=713.40 [M+H]+. Step F. 5-ethoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-carboxamide A solution of tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-{[5-(dimethylcarbamoyl)-3-ethoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (76.00 mg, 0.107 mmol, 1.00 equiv) in HFIP (5.00 mL) was stirred for 16 h at 60° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 50% B in 8 min, 50% B; wavelength: 254 nm; RT1(min): 7.2) to afford the title compound (25.90 mg, 47.39%) as a white solid. m/z (ESI, +ve ion)=513.30 [M+H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.70 (s, 1H), 7.61 (d, J=8.4 Hz, 1H), 7.42 (s, 1H), 7.27 (d, J=1.2 Hz, 1H), 6.93 (d, J=8.4 Hz, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.67 (d, J=2.4 Hz, 1H), 4.27-4.24 (m, 2H), 3.37 (d, J=8.0 Hz, 1H), 3.34 (s, 3H), 3.12 (s, 6H), 2.25-2.17 (m, 2H), 1.56-1.53 (m, 3H). Example 204. 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-sulfonamide Step A. 5-(benzylsulfanyl)-3-methoxy-2-nitropyridine To the mixture of 5-bromo-3-methoxy-2-nitropyridine (500 mg, 2.146 mmol, 1 equiv) and K2CO3(355.86 mg, 2.575 mmol, 1.2 equiv) in DMF (5 mL) was added benzyl mercaptan (293.15 mg, 2.361 mmol, 1.1 equiv) at room temperature under nitrogen atmosphere. The mixture was stirred for 4 h at room temperature. The resulting mixture was diluted with water (50 mL). The resulting mixture was extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (20 mL×3), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to afford the title compound (450 mg, 75.90%) as a yellow solid. m/z (ESI, +ve ion)=277.05 [M+H]+. Step B. 5-methoxy-N,N-dimethyl-6-nitropyridine-3-sulfonamide To the mixture of 5-(benzylsulfanyl)-3-methoxy-2-nitropyridine (390 mg, 1.411 mmol, 1 equiv) in H2O (5 mL) and DCM (10 mL) was added conc. HCl (2.02 mL) and NaClO (5.85 mL, 8.645 mmol, 6.12 equiv, 8%-10% active chlorine aqueous solution) at 0° C. under nitrogen atmosphere. The mixture was stirred at 0° C. for 15 min. The organic phase was separated quickly and injected into the mixture of dimethylamine (0.64 mL, 1.270 mmol, 0.9 equiv, 2 M in THF) and TEA (428.49 mg, 4.233 mmol, 3 equiv) in THF (3 mL) at 0° C. under nitrogen atmosphere. The mixture was stirred at 0° C. for 10 min and 25° C. for 1 h. The resulting mixture diluted with brine (20 mL). The mixture was extracted with DCM (30 mL×3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with 0-100% of EA in PE to afford the title compound (240 mg, 65.09%) as a light yellow solid. m/z (ESI, +ve ion)=260.15 [M−H]+.1H NMR (400 MHz, Chloroform-d) δ 8.43 (d, J=1.7 Hz, 1H), 7.84 (d, J=1.7 Hz, 1H), 4.07 (s, 3H), 2.87 (s, 6H). Step C. 6-amino-5-methoxy-N,N-dimethylpyridine-3-sulfonamide To a stirred mixture of 5-methoxy-N,N-dimethyl-6-nitropyridine-3-sulfonamide (240 mg, 0.367 mmol, 1 equiv) in EtOH (10 mL) was added Pd/C (101.67 mg, 10%) at room temperature under nitrogen atmosphere. The reaction mixture was degassed and purged for H2for three times. Then the resulting mixture was stirred at 25° C. for 4 h under H2(about 2 atm) atmosphere. The mixture was filtered and washed with EA (20 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with 0-20% of MeOH in DCM to afford the title compound (150 mg, 70.40/6) as a grey solid. m/z (ESI, +ve ion)=232.10 [M+H]+. Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(dimethylsulfamoyl)-3-methoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (130 mg, 0.206 mmol, 1 equiv) and 6-amino-5-methoxy-N,N-dimethylpyridine-3-sulfonamide (57.13 mg, 0.247 mmol, 1.2 equiv) in toluene (6.50 mL) were added Cs2CO3(134.15 mg, 0.412 mmol, 2 equiv), XantPhos (23.82 mg, 0.041 mmol, 0.2 equiv) and Pd2(dba)3(37.70 mg, 0.041 mmol, 0.2 equiv) at room temperature under nitrogen atmosphere. The mixture was stirred at 90° C. for 2 h. After cooled to room temperature, the mixture was filtered and washed with EtOAc (20 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column eluted with 0-00% of EtOAc in PE to give the title compound (100 mg, 66.10%) as a yellow oil. m/z (ESI, +ve ion)=735.30 [M+H]+. Step E. 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-sulfonamide The mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(dimethylsulfamoyl)-3-methoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.136 mmol, 1 equiv) in HFIP (5 mL) was stirred at 60° C. for 16 h under nitrogen atmosphere. After cooled to room temperature, the solvent was removed under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 28% B to 36% B in 8 min, 36% B; wavelength: 254 nm; RT1(min): 7) to afford the title compound (52.3 mg, 71.82%) as a white solid. m/z (ESI, +ve ion)=535.15 [M+H]+.1H NMR (400 MHz, DMSO-4) δ 12.65 (s, 1H), 10.41 (s, 1H), 9.11 (s, 1H), 7.85 (s, 1H), 7.42 (d, J=8.0 Hz, 2H), 7.28 (s, 1H), 6.90 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.60-6.58 (m, 1H), 5.74 (d, J=2.0 Hz, 1H), 3.99 (s, 3H), 3.34 (s, 3H), 3.19 (t, J=8.4 Hz, 1H), 2.62 (s, 6H), 2.35-2.31 (m, 1H), 2.01-1.97 (m, 1H). Example 208. (1R,2S)-2-(3-{[3-ethoxy-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-bromo-3-ethoxy-2-nitropyridine To a stirred solution of 5-bromo-2-nitropyridin-3-ol (657 mg, 3.000 mmol, 1 equiv) and K2CO3(829.25 mg, 6.000 mmol, 2 equiv) in DMF (7 mL) were added iodoethane (561.49 mg, 3.600 mmol, 1.2 equiv) dropwise at 25° C. under nitrogen atmosphere. The reaction mixture was stirred for 2 h at 25° C. The reaction was quenched by the addition of Water (15 mL). The resulting mixture was extracted with DCM (3×15 mL). The combined organic layers were washed with brine (3×15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA=5/1) to afford the title compound (278 mg, 37.51%) as a yellow solid. m/z (ESI, +ve ion)=246.90, 248.90 [M+H]+. Step B. 5-bromo-3-ethoxypyridin-2-amine To a stirred solution of 5-bromo-3-ethoxy-2-nitropyridine (600 mg, 2.429 mmol, 1 equiv) and NH4C1 (649.55 mg, 12.145 mmol, 5 equiv) in EtOH (2 mL) and H2O (1 mL) was added Fe (678.15 mg, 12.145 mmol, 5 equiv) at 25° C. under nitrogen atmosphere. The mixture was stirred for 2 h at 25° C. The resulting mixture was filtered, the filter cake was washed with EA (3×20 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford the title compound (147 mg, 27.88%) as a yellow solid. m/z (ESI, +ve ion)=216.95, 218.95 [M+H]+. Step C. 3-ethoxy-5-(methylsulfonyl)pyridin-2-amine To a stirred solution of 5-bromo-3-ethoxypyridin-2-amine (137 mg, 0.631 mmol, 1 equiv) and (2S,4R)-4-hydroxy-N-(2-methylnaphthalen-1-yl)pyrrolidine-2-carboxamide (HMNPC, 17.06 mg, 0.063 mmol, 0.1 equiv) in DMSO (1.5 mL) were added sodium methanesulfinate (83.76 mg, 0.820 mmol, 1.3 equiv), CuI (12.02 mg, 0.063 mmol, 0.1 equiv) and K3PO4(133.97 mg, 0.631 mmol, 1 equiv) at 25° C. under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 80° C. under nitrogen atmosphere. The mixture was allowed to cool down to 25° C. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, water (5 mM NH4HCO3) in ACN, 20% to 60% gradient in 30 min; detector, UV 254 nm. The resulting mixture was concentrated under reduced pressure to afford the title compound (76 mg, 55.68%) as a yellow solid. m/z (ESI, +ve ion)=217.10 [M+H]+. Step D. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((3-ethoxy-5-(methylsulfonyl)pyridin-2-yl)amino)-1H-indazol-6-yl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a stirred solution of 3-ethoxy-5-methanesulfonylpyridin-2-amine (61.65 mg, 0.286 mmol, 1.2 equiv) and Cs2CO3(36.17 mg, 0.476 mmol, 2 equiv) in toluene (2 mL) were added tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (150 mg, 0.238 mmol, 1.00 equiv), Pd2(dba)3(43.50 mg, 0.048 mmol, 0.2 equiv) and XantPhos (27.49 mg, 0.048 mmol, 0.2 equiv) at 25° C. under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to 25° C. The reaction was quenched by the addition of water (15 mL). The resulting mixture was extracted with DCM (3×15 mL). The combined organic layers were washed with brine (3×15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford the title compound (99 mg, 57.90%) as a yellow solid. m/z (ESI, +ve ion)=720.25 [M+H]+. Step E. (1R,2S)-2-(3-{[3-ethoxy-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(3-ethoxy-5-methanesulfonylpyridin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (99 mg, 0.138 mmol, 1 equiv) in TFA (0.2 mL) and DCM (1 mL) was stirred for 2 h at 25° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product (69 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: Water (10 mmol/L NH4HCO3). Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 28% B to 32% B in 8 min, 32% B; wavelength: 254 nm; RT1(min): 7) to afford the title compound (31.6 mg, 44.18%) as a white solid. m/z (ESI, +ve ion)=520.20 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.67 (s, 1H), 10.41 (s, 1H), 9.05 (s, 1H), 7.96 (d, J=4 Hz, 1H), 7.48 (d, J=4 Hz, 1H), 7.40 (t, J=12 Hz, 2H), 6.89 (d, J=8 Hz, 1H), 6.75 (d, J=8 Hz, 1H), 6.60-6.57 (m, 1H), 5.71 (d, J=4 Hz, 1H), 4.28-4.23 (m, 2H), 3.31 (s, 3H), 3.21-3.17 (m, 4H), 2.34-2.31 (m, 1H), 2.00-1.97 (m, 1H), 1.46 (t, J=8 Hz, 3H). Example 212. 2,5-Dimethoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)benzene-1-sulfonamide Step A: Tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-((2,5-dimethoxy-4-sulfamoylphenyl)amino)-1H-indazole-1-carboxylate A microwave vial was charged with tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-iodo-1H-indazole-1-carboxylate (100 mg, 158 μmol), 2,5-dimethoxysulfanilamide (38.7 mg, 158 μmol), tris(dibenzylideneacetone)-dipalladium(0) (14.8 mg, 15.8 μmol), 4,4-bis(diphenylphosphino)-9,9-dimethylxanthene (9.65 mg, 15.8 μmol), cesium carbonate (105 mg, 317 μmol) and dioxane (2.00 mL). The vial was sealed, and the mixture was bubbled with nitrogen for 5 minutes at room temperature and stirred at 100° C. overnight. The reaction mixture was cooled to room temperature and diluted with 10 mL of water and was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated under reduced pressure to obtain the crude product (167 mg) as a brown color gum. m/z (ESI, +ve ion)=736.5 [M+H]+. Step B: 2,5-Dimethoxy-4-((6-((1R,2S)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazol-3-yl)amino)benzenesulfonamide The crude material prepared above (167 mg) was dissolved in anhydrous dichloromethane (1.0 mL), cooled to 0° C., and trifluoracetic acid (0.3 mL) was added dropwise. The reaction mixture was stirred at room temperature for 45 minutes, and concentrated under reduced pressure to obtain a crude product. The crude product was purified using reverse phase chromatography (2 to 50% MeCN in ammonium formate) to furnish the title compound (13.8 mg, 16%, 2 steps) as a white solid after lyophilization. m/z (ESI, +ve ion)=536.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 12.39 (s, 1H), 10.43 (s, 1H), 8.26 (s, 1H), 7.96 (s, 1H), 7.84 (d, J=8.4 Hz, 1H), 7.35 (s, 1H), 7.25 (s, 1H), 6.91 (d, J=8.2 Hz, 1H), 6.85 (s, 2H), 6.74 (d, J=8.4 Hz, 1H), 6.58 (dd, J=8.5, 2.6 Hz, 1H), 5.69 (d, J=2.5 Hz, 1H), 3.88 (s, 3H), 3.79 (s, 3H), 3.32 (s, 3H), 3.22-3.13 (m, 1H), 2.33 (dd, J=7.8, 4.8 Hz, 1H), 1.97 (dd, J=8.9, 4.6 Hz, 1H). Example 228. (1R,2S)-2-(3-{[5-(Ethanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-(Ethanesulfonyl)-3-methoxypyridin-2-amine To a solution of 5-bromo-3-methoxypyridin-2-amine (7.0 g, 34 mmol, 1 equiv) and sodium ethanesulfinate (5.20 g, 44.8 mmol, 1.30 equiv) in DMSO (40 mL) was added CuI (0.66 g, 3.5 mmol, 0.1 equiv), (4R)-4-hydroxy-N-(2-methylnaphthalen-1-yl)pyrrolidine-2-carboxamide (0.93 g, 3.5 mmol, 0.1 equiv) and K3PO4(7.32 g, 34.5 mmol, 1 equiv). After stirring for 16 h at 120° C. under a nitrogen atmosphere, the resulting mixture was diluted with water (200 mL). The resulting mixture was extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine (100 mL×3), dried over anhydrous Na2SO4and filtered. The filtrate was concentrated in vacuo. The residue was purified on silica gel column chromatography and eluted with 0-20% MeOH in DCM to afford the title compound (7.2 g, 97%) as a black solid. m/z (ESI +ve ion)=217.00 [M+H]+. Step B. Tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(ethanesulfonyl)-3-methoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a solution of 5-(ethanesulfonyl)-3-methoxypyridin-2-amine (2.71 g, 12.5 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[I-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (6.60 g, 10.5 mmol, 1.00 equiv) in toluene (130 mL) under a nitrogen atmosphere was added Pd2(dba)3(1.91 g, 2.09 mmol, 0.2 equiv), XantPhos (1.21 g, 2.09 mmol, 0.2 equiv) and Cs2CO3(6.81 g, 20.90 mmol, 2 equiv). After stirring for 1 h at 90° C. under a nitrogen atmosphere, the resulting mixture was cooled down to room temperature, filtered and washed with EtOAc (50 mL). The filtrate was concentrated under reduced pressure. The residue was purified on silica gel column chromatography and eluted with 0-100% EtOAc in PE to afford the title compound (6.2 g, 82%) as a yellow solid. m/z (ESI, +ve ion)=720.20 [M+H]+. Step C. (1R,2S)-2-(3-{[5-(Ethanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(ethanesulfonyl)-3-methoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (8.30 g, 11.5 mmol, 1 equiv) in 1,1,1,3,3,3-hexafluoropropan-2-ol (80 mL) was stirred for 16 h at 60° C. The resulting mixture was concentrated under reduced pressure. The residue was purified on silica gel column chromatography and eluted with 0-20% MeOH in DCM to afford the title compound (5.3 g, 88%) as a white solid. m/z (ESI, +ve ion)=520.20 [M+H]+.1H-NMR (400 MHz, DMSO-d) δ 12.67 (s, 1H), 10.41 (s, 1H), 9.16 (s, 1H), 7.92 (d, J=2.0 Hz, 1H), 7.44-7.39 (m, 3H), 6.91-6.89 (m, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.61-6.58 (m, 1H), 5.72 (d, J=2.0 Hz, 1H), 3.99 (s, 3H), 3.34 (s, 3H), 3.32-3.29 (m, 2H), 3.21-3.18 (m, 1H), 2.34-2.31 (m, 1H), 2.01-1.97 (m, 1H), 1.13 (t, J=7.2 Hz, 3H). Example 234. (1R,2S)-5′-Methoxy-2-(3-{[2-methoxy-5-(1,3-oxazol-2-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A: 5-Bromo-N-(2,2-dimethoxyethyl)-6-methoxynicotinamide To an ice-cold solution of 5-bromo-6-methoxynicotinic acid (1.00 g, 4.18 mmol) in anhydrous tetrahydrofuran (10.0 mL) was added 4-methylmorpholine (693 μL, 6.27 mmol), isopropyl chloroformate solution (1M in toluene, 6.27 mL, 6.27 mmol) and aminoacetaldehyde dimethyl acetal (680 mg, 6.27 mmol). The reaction mixture was stirred at room temperature overnight. The mixture was diluted with water and the aqueous phase was extracted with ethyl acetate twice. The combined organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated under reduced pressure to provide the title compound (1.36 g, 97%) as a white solid. The residue was used in the next step without further purification. m/z (ESI, +ve ion)=319.1 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 8.49 (d, J=2.2 Hz, 1H), 8.23 (d, J=2.2 Hz, 1H), 6.54-5.97 (m, 1H), 4.42 (dt, J=43.0, 5.2 Hz, 1H), 4.05 (s, 3H), 3.59 (t, J=5.4 Hz, 2H), 3.43 (s, 6H). Step B: 5-Bromo-6-methoxy-N-(2-oxoethyl)nicotinamide To an ice-cold solution of 5-bromo-N-(2,2-dimethoxyethyl)-6-methoxynicotinamide (500 mg, 1.57 mmol) in acetone (3.00 mL) was added water (0.75 mL) and hydrochloric acid (37%) (0.75 mL, 24.8 mmol). The reaction mixture was stirred at room temperature overnight. Then the reaction mixture was diluted with water and neutralized with saturated NaHCO3solution. The mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated under reduced pressure to obtain the title compound (383 mg, 48%) as a white solid. The residue was not purified further and used in the next step. m/z (ESI, +ve ion)=273.1 [M+H]+. Step C: 2-(5-Bromo-6-methoxypyridin-3-yl)oxazole A mixture of 5-bromo-6-methoxy-N-(2-oxoethyl)nicotinamide (200 mg, 732 μmol), Burgess reagent (358 mg, 1.46 mmol), and tetrahydrofuran (5.00 mL) was stirred at 70° C. for 30 minutes. The reaction mixture was diluted with water (10 mL). The mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated to obtain a crude product. The crude product was purified by column chromatography (0 to 50% ethyl acetate in hexane) to afford the title compound (32.0 mg, 17%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 8.76 (m, J=3.6 Hz, 1H), 8.46 (t, J=3.2 Hz, 1H), 7.72 (s, 1H), 7.25-7.20 (m, 1H), 4.08 (s, 3H). Step D: Tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-((2-methoxy-5-(oxazol-2-yl)pyridin-3-yl)amino)-1H-indazole-1-carboxylate A 10-mL vial was charged with 2-(5-bromo-6-methoxypyridin-3-yl)oxazole (27.0 mg, 106 μmol), aminoindazole (intermediate 5) (32.4 mg, 62.3 μmol), 4,4-bis(diphenylphosphino)-9,9-dimethylxanthene (7.4 mg, 13 μmol), tris(dibenzylideneacetone)-dipalladium(0) (11.4 mg, 13 μmol), cesium carbonate (61.5 mg, 187 μmol) and dioxane (3.00 mL). The vial was bubbled with nitrogen for 5 minutes, sealed and the reaction mixture was stirred at 100° C. for 30 minutes. Then the mixture was cooled to room temperature and diluted with water. The mixture was extracted with ethyl acetate and the combined organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated by rotary evaporation to obtain a crude product (60.0 mg). m/z (ESI, +ve ion)=695.5 [M+H]+. Step E: Tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-((2-methoxy-5-(oxazol-2-yl)pyridin-3-yl)amino)-1H-indazole-1-carboxylate Tert-butyl 6-((1R,2S)-1′-(tert-butoxycarbonyl)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-3-((2-methoxy-5-(oxazol-2-yl)pyridin-3-yl)amino)-1H-indazole-1-carboxylate (60.0 mg) was dissolved in anhydrous DCM (1.00 mL), cooled to 0° C., and trifluoroacetic acid (500 uL) was added dropwise. The reaction mixture was stirred at room temperature for 15 minutes. The reaction mixture was concentrated under reduced pressure. The product was purified by reverse phase chromatography (5 to 50% MeCN in ammonium formate buffer) to afford the title compound (16.8 mg, 54%, 2 steps) as a white solid. m/z (ESI, +ve ion)=495.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 12.34 (s, 1H), 10.43 (s, 1H), 9.07 (d, J 2.0, 1H), 8.37 (s, 1H), 8.25 (d, J 2.0, 1H), 8.21 (s, 1H), 7.97 (d, J 8.4, 1H), 7.36 (s, 2H), 6.91 (d, J 8.4, 1H), 6.74 (d, J 8.4, 1H), 6.58 (dd, J 8.4, 2.5, 1H), 5.71 (d, J 2.4, 1H), 4.07 (s, 3H), 3.31 (s, 3H), 3.23-3.09 (m, 1H), 2.35 (dd, J 8.0, 4.8, 1H), 2.07-1.93 (m, 1H). Example 248. (1R,2S)-5′-Methoxy-2-(3-{[3-methoxy-5-(propane-2-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 3-Methoxy-5-(propane-2-sulfonyl)pyridin-2-amine To a stirred mixture of 5-bromo-3-methoxypyridin-2-amine (200 mg, 0.985 mmol, 1 equiv) and (2S,4R)—N-(2,6-dimethylphenyl)-4-hydroxypyrrolidine-2-carboxamide (46.16 mg, 0.197 mmol, 0.2 equiv) in DMSO (4 mL) was added CuI (37.52 mg, 0.197 mmol, 0.2 equiv), sodium propane-2-sulfinate (166.65 mg, 1.280 mmol, 1.3 equiv) and K3PO4(209.09 mg, 0.985 mmol, 1 equiv) at room temperature under nitrogen atmosphere. The mixture was stirred at 120° C. for 16 h. After cooling to room temperature, the mixture was diluted with water (20 mL). The resulting mixture solution was extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with 0-10% of MeOH in DCM to afford the title compound (120 mg, 53%) as a light yellow solid. m/z (ESI, +ve ion)=231.05 [M+H]+. Step B. Tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-5-(propane-2-sulfonyl)pyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred solution of 3-methoxy-5-(propane-2-sulfonyl)pyridin-2-amine (43.76 mg, 0.190 mmol, 1.00 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (120.00 mg, 0.190 mmol, 1.00 equiv) in toluene (5.00 mL) was added Pd2(dba)3(17.40 mg, 0.019 mmol, 0.10 equiv) and XantPhos (11.00 mg, 0.019 mmol, 0.10 equiv) and Cs2CO3(123.83 mg, 0.380 mmol, 2.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered and the filter cake was washed with EtOAc (3×30 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE:EtOAc (1:1) to afford the title compound (82.00 mg, 59%) as a yellow solid. m/z (ESI+ve ion)=734.25 [M+H]+. Step C. (1R,2S)-5′-Methoxy-2-(3-{[3-methoxy-5-(propane-2-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[3-methoxy-5-(propane-2-sulfonyl)pyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80.00 mg, 0.109 mmol, 1.00 equiv) in HFIP (5.00 mL) was stirred for 16 h at 60° C. under nitrogen atmosphere. The mixture was allowed to cool to room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (40 mg) was purified by prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30×150 mm, 5 μm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 31% B to 41% B in 10 min, 41% B; Wavelength: 254 nm; RT1(min): 7.98 to afford the title compound (28.50 mg, 49%) as a white solid. m/z (ESI, +ve ion)=534.15 [M+H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.02 (d, J=2.0 Hz, 1H), 7.59 (d, J=8.4 Hz, 1H), 7.44 (d, J=2.0 Hz, 2H), 6.96 (d, J=8.8 Hz, 1H), 6.84 (d, J=8.4 Hz, 1H), 6.65-6.62 (m, 1H), 5.65 (d, J=2.4 Hz, 1H), 4.07 (s, 3H), 3.38 (d, J=7.6 Hz, 5H), 2.27-2.24 (m, 1H), 2.21-2.17 (m, 1H), 1.31 (d, J=6.8 Hz, 6H). Example 258. (1R,2S)-2-(3-{[5-(Cyclopropanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-(Cyclopropanesulfonyl)-3-methoxypyridin-2-amine To a stirred mixture of 5-bromo-3-methoxypyridin-2-amine (200 mg, 0.985 mmol, 1 equiv) and sodium cyclopropanesulfinate (151 mg, 1.18 mmol, 1.2 equiv) in DMSO (5 mL) was added 4-hydroxy-L-proline-derived 2,6-dimethyl-aniline amide DMPHPC (CAS: 2227488-62-0, 10.1 mg, 0.099 mmol, 0.1 equiv), CuI (18.8 mg, 0.10 mmol, 0.1 equiv) and K3PO4(209 mg, 0.99 mmol, 1 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 120° C. under nitrogen atmosphere. The mixture was allowed to cool to room temperature. Water (50 mL) was added and the resulting mixture was extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (20 mL×3), dried over anhydrous Na2SO4and concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EtOAc 1:1) to afford the title compound (180 mg, 80%) as a yellow solid. m/z (ESI, +ve ion)=229.00 [M+H]+. Step B. Tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(cyclopropanesulfonyl)-3-methoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a mixture of 5-(cyclopropanesulfonyl)-3-methoxypyridin-2-amine (43.4 mg, 0.190 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) in toluene (5 mL) was added Pd2(dba)3(29.0 mg, 0.032 mmol, 0.2 equiv), XantPhos (18.3 mg, 0.032 mmol, 0.2 equiv) and Cs2CO3(103 mg, 0.316 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA 1:1) to afford the title compound (95 mg, 82%) as a yellow solid. m/z (ESI, +ve ion)=732.30 [M+H]+. Step C. (1R,2S)-2-(3-{[5-cyclopropanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one Into an 8 mL vial was added tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(cyclopropanesulfonyl)-3-methoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (95 mg, 0.130 mmol, 1 equiv) and HFIP (5 mL) at room temperature. The resulting mixture was stirred for 16 h at 60° C. under nitrogen atmosphere. The mixture was allowed to cool to mom temperature. The solvent was removed under reduced pressure. The crude product was purified by prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 19×250 mm, 10 μm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 50% B in 8 min, 50% B; Wavelength: 254 nm; RT1(min): 7.8 to afford the title compound (30.2 mg, 43.76%) as a white solid. m/z (ESI, +ve ion)=532.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.67 (s, 1H), 10.42 (s, 1H), 9.14 (s, 1H), 7.94 (s, 1H), 7.45-7.40 (m, 3H), 6.90 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.61-6.58 (m, 1H), 5.73 (d, J=2.0 Hz, 1H), 4.00 (s, 3H), 3.34-3.30 (m, 3H), 3.21-3.17 (m, 1H), 2.89-2.85 (m, 1H), 2.35-2.32 (m, 1H), 2.0)−1.97 (m, 1H), 1.12-1.11 (m, 2H), 1.11-1.04 (m, 2H). Example 272. (1R,2S)-2-{3-[2-Ethoxy-4-(methanesulfonyl)anilino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 4-Bromo-2-ethoxyaniline Into a 40 mL vial was added 4-bromo-2-ethoxy-1-nitrobenzene (500 mg, 2.032 mmol, 1 equiv), iron (567 mg, 10.2 mmol, 5 equiv) and NH4Cl (544 mg, 10.2 mmol, 5 equiv) at 25° C. To the mixture was added water (2 mL) and ethyl alcohol (8 mL) at 25° C. under nitrogen atmosphere. The resulting mixture was stirred for additional 2 hours at 25° C. The mixture was filtered and the filter cake was washed with EtOAc (3×10 mL). The solvents were removed under reduced pressure. Water (10 mL) was added and the resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography that was eluted with PE:EtOAc (3:1) to afford the title compound (411 mg, 94%) as a brown oil. m/z (ESI+ve ion)=216.00, 218.00 [M+H]+. Step B. 2-Ethoxy-4-methanesulfonylaniline Into a 20 mL vial was added 4-bromo-2-ethoxyaniline (200 mg, 0.926 mmol, 1 equiv) and sodium methanesulfinate (122.84 mg, 1.204 mmol, 1.3 equiv), K3PO4(295 mg, 1.39 mmol, 1.5 equiv), CuI (35.26 mg, 0.185 mmol, 0.2 equiv), (2S,4R)—N-(2,6-dimethylphenyl)-4-hydroxypyrrolidine-2-carboxamide (21.66 mg, 0.093 mmol, 0.10 equiv), and DMSO (5 mL) at 25° C. The resulting mixture was stirred for 16 h at 120° C. under nitrogen atmosphere. The mixture was filtered and the filter cake was washed with EtOAc (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography that eluted with 0-100% EtOAc in PE to afford the title compound (120 mg, 60%) as a light yellow solid. m/z (ESI, +ve ion)=214.00 [M−H] Step C. Tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-ethoxy-4-methanesulfonylphenyl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate Into a 20 mL vial was added tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1 equiv), 2-ethoxy-4-methanesulfonylaniline (40.91 mg, 0.190 mmol, 1.2 equiv), Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv), Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv) and toluene (2.5 mL) at 25° C. The resulting mixture was stirred for an additional 2 h at 90° C. under nitrogen atmosphere. The mixture was filtered and washed with EtOAc (3×10 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography that eluted with PE:EtOAc (1:1) to afford the title compound (73 mg, 64%) as a light yellow solid. m/z (ESI, +ve ion)=717.25 [M−H]−. Step D. (1R,2S)-2-{3-[(2-Ethoxy-4-methanesulfonylphenyl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(2-ethoxy-4-methanesulfonylphenyl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.139 mmol, 1 equiv) in HFIP (5 mL) was stirred at 60° C. for 16 h. The mixture was allowed to cool to room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 36% B to 44% B in 8 min, 44% B; Wavelength: 254 nm; RT1(min): 7.70) to afford the title compound (30.5 mg, 42.23%) as a white solid. m/z (ESI, +ve ion)=519.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.42 (s, 1H), 10.43 (s, 1H), 8.18 (s, 1H), 7.99 (d, J=8.4 Hz, 1H), 7.73 (d, J=8.4 Hz, 1H), 7.43-7.39 (m, 3H), 6.93 (d, J=8.4 Hz, 1H), 6.76 (d, J=8.4 Hz, 1H), 6.60-6.58 (m, 1H), 5.69 (d, J=2.4 Hz, 1H), 4.29-4.24 (m, 2H), 3.325 (s, 3H), 3.20 (t, J=8.5 Hz, 1H), 3.15 (s, 3H), 2.35-2.32 (m, 1H), 2.01-1.99 (m, 1H), 1.48 (t, J=7.2 Hz, 3H). Example 274. (1R,2S)-2-(3-{[5-(Ethanesulfonyl)-3-ethoxypyridin-2-yl]amino}-1H-indazol-4-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. 5-(Ethanesulfonyl)-3-ethoxypyridin-2-amine To a stirred solution of 5-bromo-3-ethoxypyridin-2-amine (163 mg, 0.751 mmol, 1 equiv) and (2S,4R)-4-hydroxy-N-(2-methylnaphthalen-1-yl)pyrrolidine-2-carboxamide (20.30 mg, 0.075 mmol, 0.1 equiv) in DMSO (2 mL) was added sodium ethanesulfinate (113.35 mg, 0.976 mmol, 1.3 equiv), CuI (14.30 mg, 0.075 mmol, 0.1 equiv) and K3PO4(159.39 mg, 0.751 mmol, 1 equiv) at 20° C. under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 120° C. under nitrogen atmosphere. The mixture was allowed to cool to room temperature. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm. The mixture was concentrated under reduced pressure to afford the title compound (150 mg, 87%) as a yellow solid. m/z (ESI+ve ion)=231.10 [M+H]+ Step B. Tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(ethanesulfonyl)-3-ethoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 5-(ethanesulfonyl)-3-ethoxypyridin-2-amine (40.11 mg, 0.174 mmol, 1.1 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) and Cs2CO3(103.19 mg, 0.316 mmol, 2.0 equiv) was added Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv) and XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) in toluene (4 mL) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EtOAc 1:2) to afford the title compound (100 mg, 81%) as a light yellow oil. m/z (ESI+ve ion)=734.30 [M+H]+ Step C. (1R,2S)-2-(3-{[5-(Ethansulfonyl)-3-ethoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[5-(ethanesulfonyl)-3-ethoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.136 mmol, 1 equiv) in HFIP (3 mL) was stirred overnight at 60° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product (70 mg) was purified by prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 40% B in 10 min, 40% B; Wavelength: 254 nm. RT1(min): 6.78) to afford the title compound (23.2 mg, 32%) as a light yellow solid. m/z (ESI, +ve ion)=534.25 [M+H]1H NMR (400 MHz, DMSO-d6) δ 12.68 (s, 1H), 10.41 (s, 1H), 9.07 (s, 1H), 7.91 (d, J=1.9 Hz, 1H), 7.41 (dd, J=5.0, 2.9 Hz, 3H), 6.90 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.59 (dd, J=8.4, 2.6 Hz, 1H), 5.73 (d, J=2.6 Hz, 1H), 4.25 (q, J=6.9 Hz, 2H), 3.34 (s, 3H), 3.27 (d, J=7.2 Hz, 2H), 3.20 (t, J=8.4 Hz, 1H), 2.33 (dd. J=8.0, 4.6 Hz, 1H), 1.99 (dd, J=9.0, 4.7 Hz, 1H), 1.45 (t, J=6.9 Hz, 3H), 1.13 (t, J=7.3 Hz, 3H). Example 276. (1R,2S)-5′-chloro-2-(3-{[3-ethoxy-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one Step A. tert-butyl 5-chloro-2-oxoindoline-1-carboxylate To a 100 mL round-bottom flask was added 5-chloro-2-oxindole (500 mg, 2.98 mmol) in THF (15 mL). Next, sodium carbonate (2.85 g, 26.85 mmol) followed by di-tert-butyl dicarbonate (1.71 mL, 7.46 mmol) were added. The flask was equipped with a Findenser™, heated to 70° C. and stirred for 1 h. The reaction was allowed to cool to room temperature. The reaction mixture was filtered through a plug of Celite eluting with EtOAc (×3) and then the filtrate was concentrated in vacuo. The crude residue was purified by flash column chromatography (0-25% EtOAc in hexanes) to give the title compound as a light brown-orange solid (449.5 mg, 56.3%).1H NMR (400 MHz, CDCl3) δ 7.75 (d, J=8.3 Hz, 1H), 7.28 (s, 1H), 7.23 (s, 1H), 3.64 (s, 2H), 1.63 (s, 9H). Step B. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-iodo-1H-indazol-6-yl)-5′-chloro-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a 20 mL vial, was added tert-butyl 5-chloro-2-oxo-indoline-1-carboxylate (159.4 mg, 0.6 mmol) and tert-butyl (S)-6-(1,2-bis((methylsulfonyl)oxy)ethyl)-3-iodo-1H-indazole-1-carboxylate (333.7 mg, 0.6 mmol) in THF (6 mL). Next, cesium carbonate (582 mg, 1.79 mmol) was added. The reaction was stirred at room temperature for 3 h. The reaction mixture was quenched with sat. ammonium chloride and extracted with EtOAc (×3). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by flash column chromatography (0-40% EtOAc in hexanes) to give a diastereomeric mixture of the title compound as a clear film (42.5 mg, 11.2%). LCMS: m/z (ESI, +ve ion)=435.9 [M−2Boc+H]+ Step C. tert-butyl (1R,2S)-2-(1-(tert-butoxycarbonyl)-3-((3-ethoxy-5-(methylsulfonyl)pyridin-2-yl)amino)-1H-indazol-6-yl)-5′-chloro-2′-oxospiro[cyclopropane-1,3′-indoline]-1′-carboxylate To a 4 mL vial was added a diastereomeric mixture of tert-butyl (1R,2S)-2-(1-tert-butoxycarbonyl-3-iodo-indazol-6-yl)-5′-chloro-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (59.9 mg, 0.09 mmol), 3-ethoxy-5-methylsulfonyl-pyridin-2-amine (24.5 mg, 0.11 mmol), Xantphos (10.9 mg, 0.02 mmol), cesium carbonate (61.4 mg, 0.19 mmol) and tris(dibenzylideneacetone)dipalladium(0) (17.3 mg, 0.02 mmol) in dry toluene (1.9 mL). Argon was bubbled through the solution for 5 min then the reaction mixture was heated to 90° C. for 2 h. The reaction mixture was allowed to cool to room temperature. The reaction mixture was diluted with DCM, filtered through a plug of Celite eluting with DCM (3×), and then the filtrate was concentrated in vacuo. The crude residue was purified by flash column chromatography (0-100% acetone in hexanes) to give a diastereomeric mixture of the title compound as a brown solid (30.3 mg, 44.4%). LCMS: m/z (ESI, +ve ion)=624.2 [M−Boc+H]+ Step D. (1R,2S)-5′-chloro-2-(3-((3-ethoxy-5-(methylsulfonyl)pyridin-2-yl)amino)-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indolin]-2′-one To a 20 ml vial, was added a diastereomeric mixture of tert-butyl (1R,2S)-2-[1-tert-butoxycarbonyl-3-[(3-ethoxy-5-methylsulfonyl-2-pyridyl)amino]indazol-6-yl]-5′-chloro-2′-oxo-spiro[cyclopropane-1,3′-indoline]-1′-carboxylate (30.3 mg, 0.04 mmol) in hexafluoro-2-propanol (3 mL, 0.04 mmol). The reaction was heated to 60° C. and stirred overnight. The reaction was allowed to cool to room temperature. The solvent was removed under reduced pressure. The crude residue was dissolved in MeCN:H2O and then purified by RP-HPLC (Interchim) using water (00 mM ammonium bicarbonate):acetonitrile (20% to 65% MeCN over 30 min) to afford the title compound as a lyophilized white solid (1.9 mg, 8.5%). LCMS: m/z (ESI, +ve ion)=524.1 [M+H]+.1H NMR (400 MHz, DMSO) δ 12.74 (s, 1H), 10.77 (s, 1H), 9.08 (s, 1H), 7.96 (s, 1H), 7.46 (d, J=8.9 Hz, 2H), 7.40 (d, J=8.4 Hz, 1H), 7.07 (d, J=8.3 Hz, 1H), 6.87 (t, J=9.9 Hz, 2H), 6.13 (s, 1H), 4.24 (q, J=7.0 Hz, 2H), 3.23 (t, J=8.8 Hz, 1H), 3.19 (s, 3H), 2.05-2.00 (m, 1H), 1.66 (s, 1H), 1.44 (t, J=7.0 Hz, 3H). Example 278. (1R,2S)-2-(3-{[6-(2-hydroxypropan-2-yl)-3-methoxypyridin-2-yl]amino}-1H-indazol-4-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one Step A. 2-(6-bromo-5-methoxypyridin-2-yl)propan-2-ol To a stirred solution of 2-bromo-6-iodo-3-methoxypyridine (600 mg, 1.911 mmol, 1 equiv) in toluene (6 mL) was added 2.5 M n-BuLi in hexane (0.76 mL, 1.911 mmol, 1 equiv) dropwise at −78° C. under nitrogen atmosphere. The resulting mixture was stirred for 1 h at −78° C. under nitrogen atmosphere. To the above mixture was added acetone (0.28 mL, 3.822 mmol, 2 equiv) at −78° C. The resulting mixture was stirred for additional 16 h at room temperature. The reaction was quenched by the addition of sat. NH4Cl (aq., 20 mL) at room temperature. The resulting mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (3×20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography that eluted with PE:EtOAc (1:1) to afford the title compound (280 mg, 60%) as a colorless oil. m/z (ESI, +ve ion)=246.05, 248.05 [M+H]+. Step B. 2-(6-amino-5-methoxypyridin-2-yl)propan-2-ol To a stirred mixture of 2-(6-bromo-5-methoxypyridin-2-yl)propan-2-ol (246 mg, 1.000 mmol, 1 equiv), Cu2O (7.15 mg, 0.050 mmol, 0.05 equiv) and K2CO3(276.29 mg, 2.000 mmol, 2 equiv) in ethane-1,2-diol (6 mL) was added 1,2-bis(methylamino)ethane (8.81 mg, 0.100 mmol, 0.1 equiv) and 28% NH3·H2O (2.570 g, 20.000 mmol, 20 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 6 h at 60° C. under nitrogen atmosphere in a sealed tube. The mixture was allowed to cool to room temperature. The resulting mixture was diluted with water (15 mL). The mixture was extracted with EtOAc (3×15 mL). The combined organic layers were washed with brine (3×20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography that eluted with PE:EtOAc (1:2) to afford the title compound (60 mg, 33%) as a colorless oil. m/z (ESI, +ve ion)=183.15 [M+H]+. Step C. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[6-(2-hydroxypropan-2-yl)-3-methoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1 equiv) and 2-(6-amino-5-methoxypyridin-2-yl)propan-2-ol (37.51 mg, 0.205 mmol, 1.3 equiv) in toluene (6 mL, 56.392 mmol) was added Pd2(dba)3(29.00 mg, 0.032 mmol, 0.2 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) and Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was allowed to cool to room temperature. The resulting mixture was diluted with water (15 mL). The mixture was extracted with EtOAc (3×15 mL). The combined organic layers were washed with brine (3×20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (DCM:MeOH=10:1) to afford the title compound (76 mg, 70%) as a yellow solid. m/z (ESI, +ve ion)=686.35 [M+H]+. Step D. (1R,2S)-2-(3-{[6-(2-hydroxypropan-2-yl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one A solution of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-{[6-(2-hydroxypropan-2-yl)-3-methoxypyridin-2-yl]amino}indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (69 mg, 0.101 mmol, 1 equiv) in HFIP (6 mL) was stirred for 16 h at 60° C. The mixture was allowed to cool to room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (80 mg) was purified by prep-HPLC with the following conditions: (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: water 10 mmol/L NH4HCO), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 31% B to 39% B in 10 min. 39% B: wavelength: 254 nm; RT1(min): 7.22) to afford the title compound (28.7 mg, 58%) as a white solid. m/z (ESI, +ve ion)=486.20 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.34 (s, 1H), 10.40 (s, 1H), 8.09 (s, 1H), 7.50 (d, J=8.4 Hz, 1H), 7.32 (s, 1H), 7.19 (d, J=8.0 Hz, 1H), 6.95 (d, J=8.0 Hz, 1H), 6.81 (d, J=10.8 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.62-6.55 (m, 1H), 5.67 (d, J=2.4 Hz, 1H), 4.77 (s, 1H), 3.87 (s, 3H), 3.31 (s, 3H), 3.22-3.13 (m, 1H), 2.32-2.25 (m, 1H), 2.02-1.94 (m, 1H), 1.16 (d, J=16.0 Hz, 6H). Example 279. (1R,2S)-2-{3-[(4-ethoxy-6-methanesulfonylpyridin-3-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one Step A. 2-bromo-4-ethoxy-5-nitropyridine Into a 20 mL vial was added 2-bromo-4-chloro-5-nitropyridine (500 mg, 2.106 mmol, 1 equiv), EtONa (530 mg, 2.336 mmol, 1.11 equiv, 30/in EtOH) and THF (2 mL) at room temperature. The mixture was stirred for 12 h at 25° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EtOAc=2:1) to afford the title compound (320 mg, 62%) as a yellow solid. m/z (ESI, +ve ion)=246.90 [M+H]+. Step B. 6-bromo-4-ethoxypyridin-3-amine Into a 20 mL vial was added 2-bromo-4-ethoxy-5-nitropyridine (250 mg, 1.012 mmol, 1 equiv), Fe (250 mg, 4.477 mmol, 4.42 equiv), NH4Cl (250 mg, 4.674 mmol, 4.62 equiv), EtOH (5 mL) and water (1 mL) at room temperature. The resulting mixture was stirred for 4 h at 30° C. under nitrogen atmosphere. The resulting mixture was filtered and the filter cake was washed with MeOH (3×10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography that eluted with PE:EtOAc (1:1) to afford the title compound (180 mg, 82%) as a light yellow solid. m/z (ESI, +ve ion)=216.90 [M+H]+. Step C. 4-ethoxy-6-methanesulfonylpyridin-3-amine To a stirred mixture of 6-bromo-4-ethoxypyridin-3-amine (200 mg, 0.921 mmol, 1 equiv) and (2S,4R)—N-(2,6-dimethylphenyl)-4-hydroxypyrrolidine-2-carboxamide (30.22 mg, 0.129 mmol, 0.14 equiv), sodium methanesulfinate (129.81 mg, 1.271 mmol, 1.38 equiv) and K3PO4(199.49 mg, 0.939 mmol, 1.02 equiv) in DMSO (3 mL) was added CuI (19.30 mg, 0.101 mmol, 0.11 equiv) at room temperature under nitrogen atmosphere. The mixture was stirred for 4 h at 120° C. under nitrogen atmosphere. The mixture was allowed to cool to room temperature. The resulting mixture was diluted with water (10 mL). The mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography that eluted with PE:EtOAc (1:1) to afford the title compound (180 mg, 90%) as a light yellow solid. m/z (ESI, +ve ion)=217.05 [M+H]+ Step D. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(4-ethoxy-6-methanesulfonylpyridin-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a solution of 4-ethoxy-6-methanesulfonylpyridin-3-amine (50.00 mg, 0.231 mmol, 1.46 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) in toluene (3 mL) was added Cs2CO3(100 mg, 0.307 mmol, 1.94 equiv), Pd2(dba)3(30 mg, 0.033 mmol, 0.21 equiv) and XantPhos (25 mg, 0.043 mmol, 0.27 equiv) under nitrogen atmosphere. After stirring for 2 h at 90° C., the mixture was allowed to cool to room temperature. The resulting mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EtOAc=1:1) to afford the title compound (75 mg, 66%) as a light yellow solid. m/z (ESI, +ve ion)=720.25 [M+H]+. Step E. (1R,2S)-2-{3-[(4-ethoxy-6-methanesulfonylpyridin-3-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one Into a 8 mL vial was added tert-butyl (1R, 2S)-2-[1-(tert-butoxycarbonyl)-3-[(4-ethoxy-6-methanesulfonylpyridin-3-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (80 mg, 0.111 mmol, 1 equiv) and HFIP (0.5 mL) at room temperature. The resulting mixture was stirred for 12 h at 60° C. under nitrogen atmosphere. The mixture was allowed to cool to room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30×150 mm, 5 μm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 31% B to 39% B in 8 min, 39% B: wavelength: 254 nm; RT1(min): 7.7) to afford the title compound (30.3 mg, 53%) as a white solid. m/z (ESI, +ve ion)=520.25 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.46 (s, 1H), 10.42 (s, 1H), 9.18 (s, 1H), 8.37 (s, 1H), 7.83 (d, J=8.0 Hz, 1H), 7.54 (s, 1H), 7.39 (s, 1H), 6.94 (d, J=8.8 Hz, 1H), 6.76 (d, J=8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.70 (d, J=2.8 Hz, 1H), 4.39-4.35 (m, 2H), 3.33-3.28 (m, 3H), 3.21-3.17 (m, 4H), 2.35-2.32 (m, 1H), 2.01-1.97 (m, 1H), 1.48 (t, J=6.8 Hz, 3H). Example 280. (1R,2S)-2-{3-[(5-difluoromethanesulfonyl-3-methoxypyridin-2-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one Step A. 2-ethylhexyl 3-[(5-methoxy-6-nitropyridin-3-yl)sulfanyl]propanoate To a stirred mixture of 5-bromo-3-methoxy-2-nitropyridine (500 mg, 2.146 mmol, 1 equiv) and 2-ethylhexyl 3-sulfanylpropanoate (562.24 mg, 2.575 mmol, 1.2 equiv) in dioxane (10 mL) was added Pd(OAc)2(96.35 mg, 0.429 mmol, 0.2 equiv), XantPhos (248.32 mg, 0.429 mmol, 0.2 equiv) and DIEA (554.66 mg, 4.292 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 12 h at 100° C. under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography that eluted with PE:EtOAc (1:1) to afford the title compound (490 mg, 62%) as a yellow solid. m/z (ESI, +ve ion)=371.20 [M+H]+. Step B. 5-methoxy-6-nitropyridine-3-thiol To a stirred mixture of 2-ethylhexyl 3-[(5-methoxy-6-nitropyridin-3-yl)sulfanyl]propanoate (450 mg, 1.215 mmol, 1 equiv) and THF (10 mL) was added EtONa (99.19 mg, 1.458 mmol, 1.2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at room temperature under nitrogen atmosphere. 10 mL of water was added into the mixture and the mixture was extracted with MTBE (2×10 mL) The aqueous layer was used directly for the next step. m/z (ESI, +ve ion)=187.05 [M+H]+. Step C. difluoromethyl)thio)-3-methoxy-2-nitropyridine To a stirred mixture of 5-methoxy-6-nitropyridine-3-thiol Na salt (10 mL crude aqueous solution from previous step) and diethyl (bromodifluoromethyl)phosphonate (286.81 mg, 1.070 mmol, 10 equiv) in CH3CN (10 mL) was added KOH (1145 mg, 20.409 mmol, 20.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at room temperature under nitrogen atmosphere. The mixture was extracted with EtOAc (3×10 mL) and the combined organic layer was dried over Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography that eluted with PE:EtOAc (1:1) to afford 200 mg crude (contained phosphors impurity on H-NMR). The crude product was repurified by RP flash that eluted with 40% MeCN in water (10 mM NH4HCO3) to give the title compound (140 mg, 58%) as a yellow oil. m/z (ESI, +ve ion)=237.10 [M+H]+. Step D. 5-[(difluoromethyl)sulfanyl]-3-methoxypyridin-2-amine A solution of 5-[(difluoromethyl)sulfanyl]-3-methoxy-2-nitropyridine (60 mg, 0.254 mmol, 1 equiv) and 10% Pd/C (13.52 mg) in EtOH (5 mL) was stirred for 16 h at room temperature under hydrogen atmosphere. The resulting mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (DCM:MeOH=10:1) to afford the title compound (48 mg, 91.63%). m/z (ESI, +ve ion)=206.85 [M+H]+ Step E. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({5-[(difluoromethyl)sulfanyl]-3-methoxypyridin-2-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate To a stirred mixture of 5-[(difluoromethyl)sulfanyl]-3-methoxypyridin-2-amine (39.19 mg, 0.190 mmol, 1.2 equiv) and tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-iodoindazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (100 mg, 0.158 mmol, 1.00 equiv) in toluene (5 mL) was added Pd2(dba). (29.00 mg, 0.032 mmol, 0.2 equiv), XantPhos (18.33 mg, 0.032 mmol, 0.2 equiv) and Cs2CO3(103.19 mg, 0.316 mmol, 2 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90° C. under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EtOAc 1:1) to afford the title compound (85 mg, 76%) as a yellow solid. m/z (ESI, +ve ion)=710.30 [M+H]+ Step F. tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-difluoromethanesulfonyl-3-methoxypyridin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate A mixture of tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-({5-[(difluoromethyl)sulfanyl]-3-methoxypyridin-2-yl}amino)indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (50 mg, 0.070 mmol, 1 equiv) and mCPBA (24.31 mg, 0.140 mmol, 2 equiv) in DCM (3 mL) was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was quenched with aq. NaHCO3(10 mL), extracted with EtOAc (3×10 mL) and the combined organic layer was dried with Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (PE:EtOAc=1:1) to afford the title compound (45 mg, 86%) as a yellow solid. m/z (ESI, +ve ion)=742.15 [M+H]+ Step H. (1R,2S)-2-{3-[(5-difluoromethanesulfonyl-3-methoxypyridin-2-yl)amino]-1H-indazol-6-yl}-5′-methoxy-1′H-spiro[cyclopropane-1,3′-indol]-2′-one Into an 8 mL vial was added tert-butyl (1R,2S)-2-[1-(tert-butoxycarbonyl)-3-[(5-difluoromethanesulfonyl-3-methoxypyridin-2-yl)amino]indazol-6-yl]-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indole]-1′-carboxylate (45 mg, 0.061 mmol, 1 equiv) and HFIP (5 mL) at room temperature. The resulting mixture was stirred for 16 h at 60° C. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 19×250 mm, 10 μm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 50% B in 8 min, 50% B; wavelength: 254 nm; RT1(min): 7.8 to afford the title compound (8.6 mg, 26%) as a white solid. m/z (ESI, +ve ion)=542.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.78 (s, 1H), 10.42 (s, 1H), 9.60 (s, 1H), 8.01 (d, J=2.0 Hz, 1H), 7.44-7.34 (m, 3H), 7.33-7.08 (m, 1H), 6.92 (d, J=8.4 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 6.61 (d, J=2.4 Hz, 1H), 5.72 (d, J=2.4 Hz, 1H), 4.01 (s, 3H), 3.33-3.22 (m, 3H), 3.18 (t, J=8.0 Hz, 1H), 2.35-2.32 (m, 1H), 2.01-1.99 (m, 1H). The compounds in Table 1B were prepared using materials and methods analogous to those disclosed herein and methods known to those having ordinary skill in the art. TABLE 1BExample No.MS/1H NMR112m/z (ESI + ve ion) = 465.20 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 7.58 (d,J = 8.4 Hz, 1H), 7.34 (s, 1H), 6.88-6.83 (m, 2H), 6.32 (d, J = 4.4 Hz, 1H), 6.19-6.05(m, 1H), 6.01 (s, 1H), 5.59 (s, 1H), 4.52-4.47 (m, 2H), 3.34-3.32 (m, 4H),2.25-2.16 (m, 5H).19F NMR (376 MHz, Methanol-d4) δ-123.83.113m/z (ESI, +ve ion) = 528.30 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 7.85 (s,1H), 7.57 (d, J = 8.4 Hz, 1H), 7.43 (s, 1H), 6.94-6.61 (m, 2H), 6.73-6.70 (m, 1H),5.71 (d, J = 2.4 Hz, 1H), 3.84-3.80 (m, 7H), 3.71-3.68 (m, 4H), 3.38-3.36 (m,4H), 3.34 (s, 3H), 2.29-2.25 (m, 1H), 2.22-2.19 (m, 1H).115m/z (ESI, +ve ion) = 455.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.36 (s,1H), 10.41 (s, 1H), 10.03 (s, 1H), 9.08 (s, 1H), 8.02-7.94 (m, 2H), 7.36 (s, 1H),6.91 (dd, J = 8.5, 1.4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.59 (dd, J = 8.5, 2.6 Hz,1H), 5.71 (d, J = 2.6 Hz, 1H), 4.91-4.79 (m, 4H), 4.40-4.36 (m, 1H), 3.19 (t, J =8.4 Hz, 1H), 2.33 (dd, J = 8.0, 4.7 Hz, 1H), 1.99 (dd, J = 9.0, 4.7 Hz, 1H).118m/z (ESI + ve ion) = 489.15 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 8.38 (s,1H), 7.55-7.45 (m, 2H), 6.92 (dd, J = 8.5, 1.4 Hz, 1H), 6.85 (d, J = 8.4 Hz, 1H),6.65 (dd, J = 8.5, 2.5 Hz, 1H), 5.62 (d, J = 2.5 Hz, 1H), 4.81-4.69 (m, 4H), 4.20-4.16 (m, 1H), 3.38 (d, J = 8.4 Hz, 1H), 3.34-3.32(m, 3H), 2.25 (dd, J = 7.9, 4.8 Hz,1H), 2.19 (dd, J = 9.1, 4.8 Hz, 1H)120m/z (ESI, +ve ion) = 504.15 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 7.94 (s,1H), 7.59 (d, J = 8.4 Hz, 1H), 7.44 (s, 1H), 6.91-6.85 (m, 1H), 6.84 (d, J = 8.8 Hz,1H), 6.65-6.62 (m, 1H), 5.61 (d, J = 1.6 Hz, 1H), 4.46-4.41 (m, 1H), 3.97 (s, 2H),3.60 (s, 2H), 3.37 (d, J = 8.8 Hz, 1H), 3.33-3.16 (m, 3H), 2.27-2.24 (m, 1H), 2.19-2.17 (m, 1H)123m/z (ESI, +ve ion) = 461.15 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 7.89 (s,1H), 7.54 (d, J = 8.4 Hz, 1H), 7.18-7.15 (m, 1H), 6.85 (d, J = 8.4 Hz, 1H), 6.65-6.63 (m, 1H), 5.56 (d, J = 2.4 Hz, 1H), 4.02 (s, 3H), 3.31 (s, 3H), 3.25 (t, J = 8.8 Hz,1H), 2.35 (s, 3H), 2.25-2.20 (m, 2H).19F NMR (376 MHz, Methanol-d4) δ-136.60(s, 1F)124m/z (ESI, +ve ion) = 413.1 [M + H]+. 1H NMR (400 MHz, METHANOL-d4) δ ppm7.75 (s, 1H) 7.46 (d, J = 8.10 Hz, 1H) 7.31 (s, 1H) 6.89-7.02 (m, 1H) 6.77-6.87(m, 2H) 6.40-6.55 (m, 1H) 5.92 (d, J = 7.60 Hz, 1H) 3.90 (s, 3H) 3.23-3.28 (m, 1H) 2.21 (s, 3H) 1.99-2.15 (m, 2H)126m/z (ESI, +ve ion) = 427.1 [M + H]+. 1H NMR (400 MHz, MeOD) δ 7.85-7.68 (m,1H), 7.56-7.42 (m, 1H), 7.31 (d, J = 2.5 Hz, 1H), 6.87-6.67 (m, 3H), 5.78 (s, 1H),3.91 (s, 3H), 3.60-3.43 (m, 1H), 2.22 (s, 3H), 2.14-2.00 (m, 2H), 1.82 (s, 3H)127m/z (ESI, +ve ion) = 427.1 [M + H]+. 1H NMR (400 MHz, MeOD) δ 7.82-7.69 (m,1H), 7.49-7.41 (m, 1H), 7.33 (s, 1H), 6.97-6.87 (m, 2H), 6.84 (s, 1H), 6.77-6.69(m, 1H), 3.90 (s, 3H), 3.56-3.45 (m, 1H), 2.32-2.27 (m, 1H), 2.26 (s, 3H), 2.22(s, 3H), 2.15-2.06 (m, 1H)129m/z (ESI, +ve ion) = 431.2 [M + H]+. 1H NMR (400 MHz, MeOD) δ 7.74 (s, 1H),7.42 (d, J = 8.5 Hz, 1H), 7.26 (s, 1H), 7.04-6.94 (m, 1H), 6.80 (d, J = 8.5 Hz, 1H),6.70 (d, J = 7.4 Hz, 1H), 6.26 (t, J = 9.5 Hz, 1H), 3.89 (s, 3H), 3.18-3.08 (m, 1H),2.50-2.43 (m, 1H), 2.21 (s, 3H), 2.08-1.99 (m, 1H)130m/z (ESI, +ve ion) = 431.2 [M + H]+. 1H NMR (400 MHz, MeOD) δ 7.86 (s, 1H),7.58 (d, J = 8.7 Hz, 1H), 7.46 (s, 1H), 7.28-7.18 (m, 1H), 7.03 (d, J = 8.5 Hz, 1H),6.85-6.77 (m, 2H), 4.02 (s, 3H), 3.82-3.71 (m, 1H), 2.59-2.48 (m, 1H), 2.43-2.36 (m, 1H), 2.34 (s, 3H)131m/z (ESI, +ve ion) = 431.2 [M + H]+. 1H NMR (400 MHz, MeOD) δ 7.80-7.69 (m,1H), 7.51-7.43 (m, 1H), 7.34-7.29 (m, 1H), 6.84-6.76 (m, 1H), 6.64-6.56 (m,1H), 6.21 (t, J = 9.4 Hz, 1H), 5.93-5.80 (m, 1H), 3.90 (s, 3H), 3.60-3.44 (m, 1H),2.21 (s, 3H), 2.18-2.10 (m, 1H), 2.10-2.00 (m, 1H)133m/z (ESI, +ve ion) = 431.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.64 (s,1H), 10.63 (s, 1H), 8.98 (s, 1H), 7.92 (s, 1H), 7.49-7.39 (m, 2H), 6.93-6.79 (m,3H), 5.93-5.86 (m, 1H), 3.90 (s, 3H), 3.24 (t, J = 8.4 Hz, 1H), 2.56-2.38 (m, 1H),2.17 (s, 3H), 2.11-2.01 (m, 1H)135m/z (ESI, +ve ion) = 467.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.76 (s,1H), 10.65 (s, 1H), 9.60 (s, 1H), 8.13 (d, J = 1.2 Hz, 1H), 7.46-7.39 (m, 2H), 7.16(s, 1H), 6.93-6.90 (m, 1H), 6.90-6.79 (m, 2H), 5.92-5.86 (m, 1H), 3.25 (t, J = 8.4Hz, 1H), 2.46-2.41 (m, 1H), 2.22 (s, 3H), 2.10-2.00 (m, 1H)136m/z (ESI, +ve ion) = 471.35 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 7.87 (s,1H), 7.63 (d, J = 8.4 Hz, 1H), 7.44 (s, 1H), 6.95 (d, J = 8.4 Hz, 1H), 6.85 (d, J = 8.4Hz, 1H), 6.64-6.61 (m, 1H), 5.64 (s, 1H), 4.78-4.72 (m, 1H), 3.50-3.15 (m, 4H),2.31 (s, 3H), 2.27-2.17 (m, 2H), 1.46 (m, 6H).137m/z (ESI, +ve ion) = 481.0 [M + H]+. 1H NMR (400 MHz, DMSO) δ 12.74-12.58(m, 1H), 11.02 (d, J = 3.3 Hz, 1H), 9.01 (s, 1H), 7.92 (d, J = 2.8 Hz, 1H), 7.51-7.32 (m, 3H), 7.02 (q, J = 3.1 Hz, 1H), 6.99-6.87 (m, 1H), 6.40 (s, 1H), 3.89 (d, J =3.0 Hz, 3H), 3.29 (s, 1H), 2.16 (d, J = 3.2 Hz, 3H), 2.10-2.04 (m, 2H)138m/z (ESI, +ve ion) = 497.2 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.62 (s, 1H),10.82 (s, 1H), 9.01 (s, 1H), 7.91 (s, 1H), 7.45 (d, J = 8.4 Hz, 1H), 7.40 (s, 1H),7.08-6.99 (m, 1H), 6.96-6.84 (m, 2H), 6.03 (s, 1H), 3.89 (s, 3H), 3.31-3.20 (m, 1H),2.16 (s, 3H), 2.11-2.01 (m, 2H)139m/z (ESI, +ve ion) = 497.2 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.60 (s, 1H),10.55 (s, 1H), 8.97 (s, 1H), 7.91 (s, 1H), 7.41 (d, J = 8.6 Hz, 1H), 7.36 (s, 1H), 7.27(s, 1H), 7.23-7.11 (m, 1H), 6.92 (t, J = 8.3 Hz, 2H), 3.90 (s, 3H), 3.42 (s, 1H),2.36-2.26 (m, 2H), 2.19 (s, 3H)140m/z (ESI + ve ion) = 453.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 8.98 (s, 1H),7.92 (s, 1H), 7.41 (d, J = 7.6 Hz, 2H), 6.89 (d, J = 8.8 Hz, 1H), 6.77 (d, J = 8.4 Hz,1H), 6.59-6.56 (m, 1H), 6.02 (s, 1H), 5.70 (d, J = 2.8 Hz, 1H), 3.33(s, 3H), 3.21 (t,J = 8.4 Hz, 1H), 2.31 (dd, J = 4.8, 4.8 Hz, 1H), 2.172 (s, 3H), 1.991 (dd, J = 4.8, 4.4Hz, 1H), 1.85-1.81 (m, 1H), 0.96-0.91 (m, 2H), 0.67-0.63 (m, 2H)141m/z (ESI, +ve ion) = 521.20 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.23(s,1H), 7.54 (d, J = 8.6 Hz, 1H), 7.46(s, 1H), 7.17-6.80 (m, 3H), 6.66-6.63 (m, 1H),5.61 (d, J = 2.4 Hz, 1H), 4.48-4.47 (m, 4H), 4.22-4.18 (m, 1H), 3.33-3.32 (m,4H), 2.27-2.17 (m, 2H).19F NMR (376 MHz, Methanol-d4) δ-83.99142m/z (ESI, +ve ion) = 449.15 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.89 (s,1H), 7.54 (d, J = 8.4 Hz, 1H), 7.17-7.13 (m, 1H), 6.93-6.90 (m, 1H), 6.85-6.80(m, 1H), 5.78-5.75 (m, 1H), 4.03 (s, 3H), 3.28 (t, J = 8.8 Hz, 1H), 2.35 (s, 3H),2.28-2.26 (m, 2H). 19F NMR (376 MHz, Methanol-d4) δ −123.62 (s, 1F), −136.88(s, 1F)143m/z (ESI, +ve ion) = 485.10 [M + H]+. 1H NMR (400 MHz, Methanol-d4) δ 8.15 (s,1H), 7.48 (d, J = 8.4 Hz, 1H), 7.20-7.14 (m, 1H), 6.97-6.79 (m, 3H), 5.75-5.73(m, 1H), 3.32-3.27 (m, 1H), 2.39 (s, 3H), 2.30-2.22 (m, 2H)144m/z (ESI, +ve ion) = 461.20 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.85 (s,1H), 7.64 (d, J = 6.0 Hz, 1H), 7.23 (d, J = 10.0 Hz, 1H), 6.85 (d, J = 8.4 Hz, 1H),6.65-6.62 (m, 1H), 5.59 (d, J = 2.4 Hz, 1H), 4.00 (s, 3H), 3.29 (s, 3H), 3.22-3.18(m, 1H), 2, 29 (s, 3H), 2.23 (d, J = 8, 4 Hz, 2H)145m/z (ESI, +ve ion) = 443.4 [M + H]+.1HNMR (500 MHz, DMSO) δ 12.52 (s, 1H),10.41 (s, 1H), 8.86 (s, 1H), 7.91 (d, J = 8.4 Hz, 1H), 7.80 (s, 1H), 7.38 (s, 1H), 6.93(dd, J = 8.6, 1.1 Hz, 1H), 6.74 (d, J = 8.4 Hz, 1H), 6.58 (dd, J = 8.5, 2.6 Hz, 1H),5.69 (d, J = 2.6 Hz, 1H), 4.09 (s, 3H), 3.31 (s, 3H), 3.18 (t, J = 8.4 Hz, 1H), 2.40 (s,3H), 2.33 (dd, J = 7.9, 4.8 Hz, 1H), 1.98 (dd, J = 9.0, 4.7 Hz, 1H).146m/z (ESI, +ve ion) = 483.20 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.63 (s,1H), 10.40 (s, 1H), 8.82 (s, 1H), 7.92 (s, 1H), 7.49 (d, J = 8.4 Hz, 1H), 7.40 (s, 1H),6.92 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.57-6.54 (m, 1H), 5.72 (d, J =2.4 Hz, 1H), 3.97 (d, J = 6.8 Hz, 2H), 3.32-3.31 (m, 3H), 3.22 (t, J = 8.8 Hz, 1H),2.32-2.30 (m, 1H), 2.19 (s, 3H), 2.01-1.97 (m, 1H), 1.31-1.29 (m, 1H), 0.60-0.57 (m, 2H), 0.38-0.37 (m, 2H)147m/z (ESI, +ve ion) = 493.20 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.96 (s,1H), 7.59 (d, J = 8.4 Hz, 1H), 7.45 (s, 1H), 6.94-6.92 (m, 1H), 6.84 (d, J = 7.6 Hz,1H), 6.64-6.61 (m, 1H), 6.33-6.31 (m, 1H), 5.62 (d, J = 2.4 Hz, 1H), 4.49-4.41(m, 2H), 3.38 (d, J = 8.4 Hz, 1H), 3.31 (s, 3H), 2.32 (s, 3H), 2.27-2.24 (m, 1H),2.20-2.19 (m, 1H)148m/z (ESI + ve ion) = 441.20 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ7.61-7.57 (m, 2H), 7.35 (s, 1H), 6.90-6.83 (m, 3H), 6.67-6.61 (m, 2H), 5.62 (d,J = 2.4 Hz, 1H), 3.93 (s, 3H), 3.38-3.36 (m, 1H), 3.30 (s, 3H), 2.25-2.17 (m, 5H)149m/z (ESI, +ve ion) = 511.20 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.02 (s,1H), 7.58 (d, J = 8.4 Hz, 1H), 7.46 (s, 1H), 6.95-6.93 (d, J = 8 Hz, 1H), 6.85 (d, J =8.4 Hz, 1H), 6.64-6.62 (m, 1H), 5.63 (d, J = 2.4 Hz, 1H), 4.82-4.76 (m, 2H), 3.39-3.37 (m, 1H), 3.31 (s, 3H), 2.32 (s, 3H), 2.28-2.24 (m, 1H), 2.21-2.17 (m, 1H)150m/z (ESI, +ve ion) = 442.20 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.07 (d,J = 8.0 Hz, 1H), 7.68 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H), 6.91-6.89 (m, 1H), 6.84 (d,J = 8.4 Hz, 1H), 6.73 (d, J = 8, 0 Hz, 1H), 6.64-6.61 (m, 1H), 5.62 (d, J = 2.4 Hz,1H), 4.06 (s, 3H), 3.37 (d, J = 8.4 Hz, 1H), 3.30 (s, 3H), 2.39 (s, 3H), 2.25-2.22 (m,1H). 2.19-2.16 (m, 1H)151m/z (ESI, +ve ion) = 455.2 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.42 (s, 1H),10.43 (s, 1H), 9.98 (s, 1H), 7.91 (d, J = 8.5 Hz, 1H), 7.41 (s, 1H), 7.36 (s, 1H), 6.90(d, J = 8.5 Hz, 1H), 6.75 (d, J = 8.5 Hz, 1H), 6.58 (d, J = 8.4 Hz, 1H), 5.69 (s, 1H),3.21-3.14 (m, 1H), 2.98-2.85 (m, 1H), 2.37-2.27 (m, 4H), 2.02-1.94 (m, 1H),1.22 (d, J = 6.6 Hz, 6H)152m/z (ESI, +ve ion) = 441.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.60 (s,1H), 10.41 (s, 1H), 8.93 (s, 1H), 8.03 (s, 1H), 7.39-7.35 (m, 2H), 6.89 (d, J = 8.0Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.59 (dd, J = 8.4, 2.4 Hz, 1H), 5.70 (d, J = 2.4 Hz,1H), 3.33 (s, 3H), 3.20 (t, J = 8.4 Hz, 1H), 2.68-2.59 (m, 2H), 2.33-2.31 (m, 1H),2.17 (s, 3H), 1.99 (dd, J = 9.6, 4.4 Hz, 1H), 1.20 (t, J = 7.2 Hz, 3H)153m/z (ESI, +ve ion) = 455.1 [M + H]+.1H NMR (500 MHz, DMSO) δ 12.52 (s, 1H),10.40 (s, 1H), 9.15 (s, 1H), 7.42 (d, J = 8.4 Hz, 1H), 7.36 (s, 1H), 6.88 (d, J = 8.5Hz, 1H), 6.74 (d, J = 8.4 Hz, 1H), 6.58 (dd, J = 8.5, 2.6 Hz, 1H), 5.70 (d, J = 2.5 Hz,1H), 4.55 (t, J = 9.2 Hz, 2H), 3.31 (s, 3H), 3.22-3.10 (m, 3H), 2.31 (dd, J = 7.9, 4.7Hz, 1H), 2.22 (s, 3H), 1.97 (dd, J = 9.0, 4.7 Hz, 1H)155m/z (ESI, +ve ion) = 475.1 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.70 (s, 1H),10.43 (s, 1H), 9.30 (s, 1H), 7.94 (d, J = 2.8 Hz, 1H), 7.48-7.34 (m, 2H), 6.90 (d,J = 8.4 Hz, 1H), 6.74 (d, J = 8.1 Hz, 1H), 6.58 (d, J = 8.3 Hz, 1H), 5.69 (s, 1H), 3.89(s, 3H), 3.23-3.10 (m, 1H), 2.38-2.28 (m, 1H), 2.06 (s, 3H), 2.01-1.92 (m, 1H)156m/z (ESI, +ve ion) = 459.4 [M + H]+.1H NMR (500 MHz, DMSO) δ 12.67 (s, 1H),10.41 (s, 1H), 9.18 (s, 1H), 7.81 (s, 1H), 7.44 (d, J = 8.4 Hz, 1H), 7.39 (s, 1H), 6.90(d, J = 8.4 Hz, 1H), 6.74 (d, J = 8.4 Hz, 1H), 6.58 (dd, J = 8.5, 2.6 Hz, 1H), 5.68 (d,J = 2.5 Hz, 1H), 3.89-3.82 (m, 3H), 3.52 (s, 3H), 3.32 (s, 3H), 3.20-3.15 (m, 1H),2.31 (dd, J = 7.9, 4.8 Hz, 1H), 1.97 (dd, J = 9.1, 4.6 Hz, 1H)158m/z (ESI, +ve ion) = 496.15 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.86 (s,1H), 7.58 (d, J = 8.4 Hz, 1H), 7.43 (s, 1H), 7.36 (d, J = 1.6 Hz, 1H), 6.95 (d, J = 8.8Hz, 1H), 6.84 (d, J = 8.4 Hz, 1H), 6.65-6.62 (m, 1H), 5.66 (d, J = 2.4 Hz, 1H), 4.05(s, 3H), 3.38-3.32 (m, 4H), 2.27-2.23 (m, 1H), 2.20-2.18 (m, 1H)159m/z (ESI, +ve ion) = 453.4 [M + H]+.1H NMR (500 MHz, MeOD) δ = 7.52 (d,J = 8.5, 1H), 7.43 (s, 1H), 6.92 (dd, J = 8.5, 1.0, 1H), 6.82 (d, J = 8.5, 1H), 6.61 (dd,J = 8.5, 2.6, 1H), 5.60 (d, J = 2.5, 1H), 3.37-3.33 (m, 1H), 3.30 (s, 3H), 2.88 (t, J = 7.8,2H), 2.70 (t, J = 7.4, 2H), 2.23 (dd, J = 7.9, 4.8, 1H), 2.17 (dd, J = 9.1, 4.8, 1H), 2.14-2.07 (m, 2H)162m/z (ESI, +ve ion) = 459.15 [M + H]+.1H NMR (400 MHz, Methanol-d6) δ 8.34 (s,1H), 7.56 (d, J = 8.8 Hz, 1H), 7.47 (s, 1H), 6.94 (d, J = 8.4 Hz, 1H), 6.84 (d, J = 8.4Hz, 1H), 6.64-6.62 (m, 1H), 5.62 (d, J = 2.0 Hz, 1H), 3.80-3.60 (m, 1H), 3.31 (s,3H), 2.46 (s, 3H), 2.33 (s, 3H), 2.27-2.25 (m, 1H), 2.21-2.17 (m, 1H)164m/z (ESI, +ve ion) = 458.05 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.21 (s,1H), 10.42 (s, 1H), 8.15 (d, J = 2.4 Hz, 1H), 8.07 (s, 1H), 7.92 (d, J = 8.4 Hz, 1H),7.33 (s, 1H), 7.30 (d, J = 2.4 Hz, 1H), 6.90 (d, J = 7.6 Hz, 1H), 6.75 (d, J = 8.4 Hz,1H), 6.58 (dd, J = 10.8, 2.4 Hz, 1H), 5.70 (d, J = 2.4 Hz, 1H), 3.95 (s, 3H), 3.76 (s,3H), 3.32 (s, 3H), 3.18 (t, J = 8.4 Hz, 1H), 2.34-2.31 (m, 1H), 2.00-1.96 (m, 1H)165m/z (ESI, +ve ion) = 473.3 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.87 (s,1H), 10.43 (s, 1H), 8.41 (s, 1H), 8.29 (d, J = 2.0 Hz, 1H), 7.52 (d, J = 8.5 Hz, 1H),7.45 (s, 1H), 7.03 (d, J = 2.2 Hz, 1H), 6.95 (d, J = 7.9 Hz, 1H), 6.74 (d, J = 8.4 Hz,1H), 6.58 (dd, J = 8.4, 2.6 Hz, 1H), 5.70 (d, J = 2.5 Hz, 1H), 3.32 (s, 3H), 3.20 (t, J =8.3 Hz, 1H), 2.37-2.31 (m, 1H), 1.98 (dd, J = 9.1, 4.7 Hz, 1H)166m/z (ESI, +ve ion) = 499.3 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.30 (s, 1H),10.45 (s, 1H), 8.56-8.24 (m, 2H), 7.94 (s, 1H), 7.51-7.15 (m, 2H), 7.08-6.43(m, 3H), 5.71 (s, 1H), 4.01 (s, 3H), 3.21-3.11 (m, 4H), 2.99 (s, 3H), 2.51 (s, 3H),2.42-2.26 (m, 1H), 2.07-1.89 (m, 1H)168m/z (ESI, +ve ion) = 506.2 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.47 (s, 1H),10.44 (s, 1H), 8.79 (s, 1H), 8.52-8.36 (m, 1H), 7.99-7.86 (m, 1H), 7.71-7.54(m, 1H), 7.38 (s, 1H), 7.01-6.87 (m, 1H), 6.82-6.68 (m, 1H), 6.68-6.50 (m, 1H),5.70 (s, 1H), 4.09 (s, 3H), 3.19 (s, 4H), 2.39-2.28 (m, 1H), 2.08-1.93 (m, 1H)171m/z (ESI, +ve ion) = 486.3 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.74 (s,1H), 10.42 (d, J = 8.5 Hz, 1H), 9.31 (s, 1H), 8.39 (s, 1H), 7.64 (d, J = 11.1 Hz, 1H),7.43 (d, J = 6.1 Hz, 1H), 6.92 (d, J = 8.6 Hz, 1H), 6.74 (d, J = 8.4 Hz, 1H), 6.58 (dd,J = 8.5, 2.5 Hz, 1H), 6.38 (t, J = 6.6 Hz, 1H), 5.72 (d, J = 2.5 Hz, 1H), 4.07 (s, 3H),3.31-3.26 (m, 3H), 3.19 (t, J = 8.4 Hz, 1H), 2.37-2.29 (m, 1H), 1.96 (dt, J = 30.4,15.2 Hz, 1H)172m/z (ESI, +ve ion) = 454.3 [M + H]+.1H NMR (500 MHz, DMSO) δ = 12.83 (s, 1H),10.42 (s, 1H), 9.77 (s, 1H), 8.17 (s, 1H), 7.44 (d, J = 8.7, 2H), 6.97-6.94 (m, 1H),6.74 (d, J = 8.4, 1H), 6.58 (dd, J = 8.4, 2.6, 1H), 5.68 (d, J = 2.5, 1H), 4.03 (s, 3H), 3.30(s, 3H), 3.20 (t, J = 8.5, 1H), 2.33 (dd, J = 7.9, 4.7, 1H), 1.99 (dd, J = 9.0, 4.7, 1H)175m/z (ESI, +ve ion) = 519.10 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.28 (s,1H), 10.40 (s, 1H), 8.66 (s, 1H), 8.14 (s, 1H), 7.88 (d, J = 8 Hz, 1H), 7.34 (t,J = 8 Hz, 2H), 7.21 (d, J = 8 Hz, 1H), 6.89 (d, J = 8 Hz, 1H), 6.75 (d, J = 8 Hz, 1H),6.59-6.57 (m, 1H), 5.70 (s, 1H), 4.01 (s, 3H), 3.32-3.28 (m, 3H), 3.18-3.12 (m, 3H),2.34-2.31 (m, 1H), 2.00-1.96 (m, 1H), 1.12-1.11 (d, J = 4 Hz, 3H)176m/z (ESI, +ve ion) = 498.10 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.10 (s,1H), 10.42 (s, 1H), 8.19 (d, J = 2.0 Hz, 1H), 7.86-7.83 (m, 2H), 7.33 (s, 1H), 7.02(d, J = 8.4 Hz, 1H), 6, 88-6.86 (m, 2H), 6.76 (d, J = 8.4 Hz, 1H), 6.58-6.57 (m,1H), 5.70 (d, J = 2.4 Hz, 1H), 3.94 (s, 3H), 3.44-3.31 (m, 3H), 3.20-3.16 (m, 1H),2.96 (s, 6H), 2.34-2.31 (m, 1H), 1.98-1.96 (m, 1H)177m/z (ESI + ve ion) = 484.10 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.14 (s,1H), 10.42 (s, 1H), 8.52 (d, J = 2.0 Hz, 1H), 8.15 (d, J = 4.4 Hz, 1H), 7.81 (d, J =8.4 Hz, 1H), 7.74 (s, 1H), 7.33-7.29 (m, 2H), 7.03 (d, J = 8.4 Hz, 1H), 6.88 (d, J =8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.58-6.57(m, 1H), 5.71 (d, J = 2.4 Hz, 1H),3.94 (s, 3H), 3.33 (s, 3H), 3.20-3.16 (m, 1H), 2.74 (d, J = 4.4 Hz, 3H), 2.34-2.31(m, 1H), 2.00-1.96 (m, 1H)178m/z (ESI, +ve ion) = 533.30 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 8.43 (t,J = 1.9 Hz, 1H), 7.67 (d, J = 8.5 Hz, 1H), 7.41-7.34 (m, 2H), 7.20-7.17 (m, 1H),6.91 (d, J = 8.4 Hz, 1H), 6.84 (d, J = 8.5 Hz, 1H), 6.64-6.61 (m, 1H), 5.62 (d, J =2.5 Hz, 1H), 4.08 (d, J = 1.8 Hz, 3H), 3.39-3.37 (m, 1H), 3.32-3.29 (s, 4H), 2.26-2.22 (m, 1H), 2.20-2.17 (m, 1H), 1.26 (d, J = 6.8 Hz, 6H)179m/z (ESI, +ve ion) = 534.20 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 8.33 (d,J = 2.0 Hz, 1H), 7.67 (d, J = 8.8 Hz, 1H), 7.39 (s, 1H), 7.31-7, 28 (m, 1H), 7.16 (d,J = 8.4 Hz, 1H), 6.91 (d, J = 9.2, 1H), 6.84 (d, J = 8.4 Hz, 1H), 6.64-6.61 (m, 1H),5.63 (d, J = 2.8 Hz, 1H), 4.07 (s, 3H), 3.33-3.30 (m, 4H), 2.66 (s, 6H), 2.25-2.18(m, 2H)180m/z (ESI, +ve ion) = 540.10 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.13 (s,1H), 10.42 (s, 1H), 8.24 (d, J = 2.0 Hz, 1H), 7.87 (d, J = 8.0 Hz, 2H), 7.33 (s, 1H),7.04 (d, J = 8.4 Hz, 1H), 6.90-6.87 (m, 2H), 6, 75 (d, J = 8.4 Hz, 1H), 6.58-6.57(m, 1H), 5.70 (d, J = 2.8 Hz, 1H), 3.94 (s, 3H), 3.60-3.58 (m, 4 H), 3.52-3.49 (m,4H), 3.32 (s, 3H), 3.20-3.16 (m, 1H), 2.34-2.31 (m, 1H), 2.00-1.96 (m, 1H)181m/z (ESI, +ve ion) = 499.60 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 8.33 (d,J = 2.4 Hz, 1H), 7.73 (t, J = 5.2 Hz, 2H), 7.37 (s, 1H), 6.92 (d, J = 7.6 Hz, 1H), 6.84(d, J = 8.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.62 (d, J = 2.4 Hz, 1H), 4.13 (s, 3H),3.38-3.36 (m, 1H), 3.29 (s, 3H), 3.10 (s, 6H), 2.25-2.17 (m, 2H)182m/z (ESI, +ve ion) = 456.10 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.52 (s,1H), 10.41 (s, 1H), 8.73 (s, 1H), 7.76 (s, 1H), 7.43-7.34 (m, 2H), 6.84 (d, J = 8.4Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.57-6.60 (m, 1H), 5.64 (d, J = 2.6 Hz, 1H), 3.33(s, 3H), 3.18 (t, J = 8.4 Hz, 1H), 2.70 (s, 6H), 2.28-2.31 (m, 1H), 2.06 (s, 3H),1.96-1.99 (m, 1H)184m/z (ESI, +ve ion) = 547.1 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.39 (s, 1H),10.44 (s, 1H), 8.58 (s, 1H), 8.46-8.35 (m, 1H), 7.97-7.87 (m, 1H), 7.73-7.64(m, 1H), 7.36 (s, 1H), 6.97-6.87 (m, 1H), 6.79-6.71 (m, 1H), 6.64-6.53 (m, 1H),5.70 (s, 1H), 5.16 (t, J = 12.8 Hz, 2H), 4.46 (t, J = 12.7 Hz, 2H), 4.05 (s, 3H), 3.32(s, 3H), 3.23-3.11 (m, 1H), 2.40-2.27 (m, 1H), 2.04-1.92 (m, 1H)186m/z (ESI, +ve ion) = 493.05 [M + H]+.1H NMR (400 MHz, Methanol-d6) δ 7.82 (s,1H), 7.28 (s, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.76-6.60 (m, 2H), 5.76 (d, J = 2.4 Hz,1H), 3.98 (s, 3H), 3.40 (s, 3H), 3.35-3.34 (m, 1H), 2.27-2.24 (m, 1H), 2.17-2.10(m, 1H), 1.93 (s, 3H)187m/z (ESI, +ve ion) = 443.05 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.68 (s,1H), 10.42 (s, 1H), 9.07 (s, 1H), 7.92 (s, 1H), 7.39 (d, J = 7.7 Hz, 2H), 6.89 (d, J =8.4 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.57-6.60 (m, 1H), 5.67 (d, J = 2.5 Hz, 1H),3.48 (s, 3H), 3.33 (s, 3H), 3.19 (t, J = 8.4 Hz, 1H), 2.30-2.33 (m, 1H), 2.14 (s, 3H),1.97-2.00 (m, 1H)188m/z (ESI, +ve ion) = 553.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.34 (s,1H), 10.43 (s, 1H), 8.47 (s, 1H), 8.39 (d, J = 8.0 Hz, 1H), 7.92 (d, J = 8.4 Hz, 1H),7.60 (d, J = 8.2 Hz, 1H), 7.36 (s, 1H), 6.92 (d, J = 8.5 Hz, 1H), 6.75 (d, J = 8.4 Hz,1H), 6.60-6.57 (m, 1H), 5.70 (d, J = 2.0 Hz, 1H), 4.92 (s, 2H), 4.75-4.71 (m, 4H),4.20 (s, 2H), 4.08 (s, 3H), 3.30 (s, 3H), 3.19 (t, J = 8.4 Hz, 1H), 2.35-2.32 (m, 1H),2.00-1.97 (m, 1H190m/z (ESI, +ve ion) = 562.05 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.62 (s,1H), 10.42 (s, 1H), 9.11 (s, 1H), 7.79 (s, 1H), 7.47-7.44 (m, 2H), 6.76 (t, J = 16 Hz,2H), 6.59-6.57 (m, 1H), 5.62 (d, J = 4 Hz, 1H), 3.82 (s, 3H), 3, 75-3.72 (s, 2H),3.69-3.64 (m, 2H), 3.34 (s, 3H), 3.20 (t, J = 16 Hz, 1H), 2.93-2.89 (m, 2H), 2.51-2.50 (m, 2H), 2.36-2.32 (m, 1H), 2.00-1.98 (m, 1H)191m/z (ESI, +ve ion) = 485.10 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.32 (s,1H), 10.43 (s, 1H), 8.46-8.40 (m, 2H), 8.30 (d, J = 4.8 Hz, 1H), 7.93 (d, J = 8.4 Hz,1H), 7.61 (d, J = 8.0 Hz, 1H), 7.35 (s, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.4Hz, 1H), 6.59-6.57(m, 1H), 5.70 (d, J = 2.0 Hz, 1H), 4.12 (s, 3H), 3.33 (s, 3H),3.20-3.14 (m, 1H), 2.83 (d, J = 4.4 Hz, 3H), 2.35-2.32 (m, 1H), 2.00-1.97 (m,1H)192m/z (ESI, +ve ion) = 526.15 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.55 (s,1H), 10.40 (s, 1H), 8.87 (s, 1H), 7.73 (s, 1H), 7.54 (d, J = 8 Hz, 1H), 7.39 (s, 1H),6.84 (d, J = 12 Hz, 1H), 6.76 (d, J = 8 Hz, 1H), 6.63-6.60 (m, 1H), 5, 66 (s, 1H),4.60-4.53 (m, 4H), 3.81-3.74 (m, 6H), 3.31(s, 4H), 3.22-3.18 (t, J = 16 Hz, 1H),2.33-2.30 (m, 1H), 2.01-1.98 (m, 1H)196m/z (ESI, +ve ion) = 506.05 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.60 (s,1H), 10.41 (s, 1H), 8.98 (s, 1H), 7.49 (d, J = 8.5 Hz, 1H), 7.42-7.32 (m, 3H), 6.81(dd, J = 8.2, 1.3 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.60 (dd, J = 8.4, 2.6 Hz, 1H),5.65 (d, J = 2.5 Hz, 1H), 3.98 (s, 3H), 3.33 (s, 3H), 3.18 (t, J = 8.5 Hz, 1H), 2.74 (s,3H), 2.34-2.28 (m, 1H), 1.99-1.96 (m, 1H)197m/z (ESI, +ve ion) = 499.20 [M + H]+.1H-NMR. (400 MHz, Methanol-d4) δ 7.71 (d,J = 1.2 Hz, 1H), 7.60 (d, J = 8.4 Hz, 1H), 7.42 (s, 1H), 7.30 (d, J = 1.6 Hz, 1H), 6.93(d, J = 8.4 Hz, 1H), 6.84 (d, J = 8.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.67 (d, J = 2.4Hz, 1H), 4.03 (s, 3H), 3.37-3.33 (m, 1H), 3.32 (s, 3H), 3.12 (s, 6H), 2.24-2.17 (m,2H)198m/z (ESI, +ve ion) = 499.10 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.43 (s,1H), 10.42 (s, 1H), 8.56 (s, 1H), 8.12 (s, 1H), 8.07 (s, 1H), 7.86 (d, J = 8.4 Hz, 1H),7.39 (s, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.57-6.60 (m, 1H),5.70 (d, J = 2.5 Hz, 1H), 4.03 (s, 3H), 3.31 (s, 3H), 3.19 (t, J = 8.4 Hz, 1H), 2.97 (d,J = 5.4 Hz, 6H), 2.32-2.35 (m, 1H), 1.97-2.00 (m, 1H)199m/z (ESI, +ve ion) = 540.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.19 (s,1H), 10.43 (s, 1H), 8.07 (d, J = 8.0 Hz, 1H), 7.95 (s, 1H), 7.84 (d, J = 8.4 Hz, 1H),7.34 (s, 1H), 7.04 (s, 1H), 7.00 (d, J = 8.0 Hz, 1H), 6.90 (d, J = 8.8 Hz, 1H), 6.76 (d,J = 8.0 Hz, 1H), 6.60-6.57 (m, 1H), 5.70 (d, J = 2.4 Hz, 1H), 3.93 (s, 3H), 3.61 (s,4H), 3.54 (s, 4H), 3.30 (s, 3H), 3.20-3.16 (m, 1H), 2.34-2.31 (m, 1H), 2.00-1.97(m, 1H)200m/z (ESI, +ve ion) = 499.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.61 (s,1H), 10.42 (s, 1H), 9.09 (s, 1H), 7.99 (s, 1H), 7.44 (d, J = 8.4 Hz, 1H), 7.39 (s, 1H),6.88 (d, J = 8.4 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.60 (d, J = 8.4 Hz, 1H), 5.68 (s,1H), 3.92 (s, 3H), 3.83-3.79 (m, 1H), 3.63-3.57 (m, 3H), 3.34-3.24 (m, 3H),3.22-3.17 (m, 2H), 2.33-2.30 (m, 1H), 2.09-1.92 (m, 3H)201m/z (ESI, +ve ion) = 499.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.61 (s,1H), 10.42 (s, 1H), 9.10 (s, 1H), 7.99 (s, 1H), 7.42-7.38 (m, 2H), 6.86 (d, J = 8.4Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.67 (d, J = 2.4 Hz, 1H), 3.92(s, 3H), 3.87-3.83 (m, 1H), 3.65-3.61 (m, 1H), 3.53-3.49 (m, 2H), 3.34-3.24(m, 3H), 3.22-3.17 (m, 2H), 2.33-2.30 (m, 1H), 2.05-1.92 (m, 2H), 1.90-1.87(m, 1H)202m/z (ESI, +ve ion) = 520.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.47 (s,1H), 10.42 (s, 1H), 8.60 (s, 1H), 8.36 (d, J = 8.2 Hz, 1H), 7.87 (d, J = 8.4 Hz, 1H),7.59 (d, J = 8.1 Hz, 1H), 7.39 (s, 1H), 6.98-6.91 (m, 1H), 6.76 (d, J = 8.4 Hz, 1H),6.59 (dd, J = 8.4, 2.6 Hz, 1H), 5.69 (d, J = 2.6 Hz, 1H), 4.58-4.52 (m, 2H), 3.32 (s,3H), 3.22-3.20 (m, 1H), 3.18 (s, 3H), 2.35-2.32 (m, 1H), 2.01-1.98 (m, 1H), 1.47(t, J = 7.0 Hz, 3H)205m/z (ESI, +ve ion) = 504.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.32 (s,1H), 10.41 (s, 1H), 8.40-8.37 (m, 2H), 7.92 (d, J = 8.8 Hz, 1H), 7.49-7.46 (m,1H), 7.36 (s, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.0 Hz, 1H), 6.60-6.58 (m,1H), 5.71 (d, J = 2.0 Hz, 1H), 4.04 (s, 3H), 3.32 (s, 3H), 3.19 (t, J = 8.4 Hz, 1H),2.35-2.31 (m, 1H), 2.01-1.97 (m, 1H), 1.63 (d, J = 13.2 Hz, 6H)206m/z (ESI, +ve ion) = 516.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.26 (s,1H), 10.41 (s, 1H), 8.20 (s, 1H), 8.01 (d, J = 12.4 Hz, 1H), 7.89 (d, J = 8.4 Hz, 1H),7.35 (s, 1H), 6.91 (t, J = 6 Hz, 2H), 6.75 (d, J = 8.4 Hz, 1H), 6.60-6.57 (m, 1H),5.70 (d, J = 2.0 Hz, 1H), 4.03 (s, 3H), 3.35 (s, 3H), 3.19-3.12 (m, 1H), 3.14 (s, 3H),2.92 (s, 3H), 2.34-2.31 (m, 1H), 2.08-1.98 (m, 1H)207m/z (ESI, +ve ion) = 558.25 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.77 (d,J = 12.0 Hz, 1H), 7.69 (d, J = 8.4 Hz, 1H), 7.39 (s, 1H), 6.96-6.92 (m, 2H), 6.84 (d,J = 8.8 Hz, 1H), 6.64-6.61 (m, 1H), 5.61 (d, J = 2.8 Hz, 1H), 3.99 (s, 3H),3.77-3.68 (m, 6H), 3.48 (s, 2H), 3.38-3.32 (m, 1H), 3.30 (s, 3H), 2.26-2.23 (m, 2H)209m/z (ESI, +ve ion) = 520.1 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.42 (s, 1H),10.38 (s, 1H), 8.74 (s, 1H), 8.46-8.27 (m, 1H), 7.95-7.79 (m, 1H), 7.63-7.50(m, 1H), 7.32 (s, 1H), 6.94-6.82 (m, 1H), 6, 74-6.64 (m, 1H), 6.58-6.47 (m, 1H),5.76-5.57 (m, 2H), 4.00 (s, 3H), 3.26 (s, 3H), 3.18-3.04 (m, 2H), 2.35-2.23 (m,1H), 1.99-1.88 (m, 1H), 1.16-0.99 (m, 3H)210m/z (ESI, +ve ion) = 534.4 [M + H]+.1HNMR (400 MHz, DMSO-d6) δ 12.33 (s,1H), 10.43 (s, 1H), 8.17 (s, 1H), 7.92 (s, 1H), 7.81 (d, J = 8.4 Hz, 1H), 7.34 (d, J =17.1 Hz, 2H), 7.18 (q, J = 5.0 Hz, 1H), 6.90 (d, J = 8.5 Hz, 1H), 6.74 (d, J = 8.4 Hz,1H), 6.58 (dd, J = 8.5, 2.5 Hz, 1H), 5, 69 (d, J = 2.5 Hz, 1H), 3.92 (s, 3H), 3.31 (s,3H), 3.18 (t, J = 8.4 Hz, 1H), 2.43 (s, 3H), 2.39 (d, J = 5.0 Hz, 3H), 2.33 (dd, J = 7.9,4, 7 Hz, 1H), 1.98 (dd, J = 9.0, 4.6 Hz, 1H)211m/z (ESI, +ve ion) = 485.4 [M + H]+.1H NMR: (400 MHz, DMSO-d6) δ 12.67 (s,1H), 10.43 (s, 1H), 9.33 (s, 1H), 7.41 (d, J = 7.9 Hz, 2H), 6.90 (d, J = 9.1 Hz, 1H),6.74 (d, J = 8.4 Hz, 1H), 6.58 (dd, J = 8.5, 2.5 Hz, 1H), 5.71 (d, J = 2.5 Hz, 1H),4.74 (dd, J = 6.4, 2.5 Hz, 1H), 4.26 (dd, J = 15.7, 2.8 Hz, 1H), 3.97 (d, J = 15.7 Hz,1H), 3.32 (s, 3H), 3.24-3.10 (m, 1H), 2.33 (dd, J = 7.8, 4.6 Hz, 1H), 2.23 (s, 3H),1.98 (dd, J = 9.1, 4.7 Hz, 1H), 1.47 (d, J = 6, 5 Hz, 3H)213m/z (ESI, +ve ion) = 482.2 [M + H]+.1H NMR (500 MHz, DMSO) δ = 12.87 (s, 1H),10.43 (s, 1H), 10.18 (s, 1H), 7.47-7.43 (m, 2H), 6.88 (dd, J = 8.5, 0.9, 1H), 6.75 (d,J = 8.4, 1H), 6.58 (dd, J = 8.5, 2.6, 1H), 5.60 (d, J = 2.6, 1H), 3.31 (s, 3H), 3.19 (t,J = 8.4, 1H), 2.94-2.67 (m, 10H), 2.30 (dd, J = 7.9, 4.7, 1H), 2.13-2.04 (m, 2H),1.98 (dd, J = 9.0, 4.7, 1H)214m/z (ESI, +ve ion) = 513.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.29 (s,1H), 10.41 (s, 1H), 8.34 (d, J = 8.1 Hz, 1H), 8.15 (s, 1H), 7.86 (d, J = 8.3 Hz, 1H),7.36 (s, 1H), 7.25 (d, J = 8.0 Hz, 1H), 6.92 (d, J = 8.3 Hz, 1H), 6.76 (d, J = 8.4 Hz,1H), 6.59 (dd, J = 8.4, 2.6 Hz, 1H), 5.69 (d, J = 2.6 Hz, 1H), 4.49-4.44 (m, 2H),3.32 (s, 3H), 3.19 (t, J = 8.4 Hz, 1H), 3.15 (s, 3H), 2.99 (s, 3H), 2.32-2.30 (m, 1H),2.00-1.97 (m, 1H). 1.44 (t, J = 7.0 Hz, 3H)215m/z (ESI, +ve ion) = 511.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.05 (s,1H), 10.42 (s, 1H), 8.43 (d, J = 8.8 Hz, 1H), 7.94-7.90 (m, 2H), 7.78 (d, J = 8.4 Hz,1H), 7.30 (s, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.59 (d, J = 7.6Hz, 1H), 5.71 (s, 1H), 4.05-3.99 (m, 5H), 3.33 (s, 3H), 3.20-3.16 (m, 1H), 2.56-2.51 (m, 2H), 2.33 (d, J = 4.8 Hz, 1H), 2.07-2.03 (m, 2H), 2.00-1.98 (m, 1H)216m/z (ESI + ve ion) = 576.20 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.15 (s,1H), 7.64 (d, J = 8.4 Hz, 1H), 7.42 (s, 1H), 7.29-7.26 (m, 1H), 7.17 (d, J = 8.4 Hz,1H), 6.90 (d, J = 8, 4 Hz, 1H), 6.84 (d, J = 8.4 Hz, 1H), 6.64-6.62 (m, 1H), 5.64 (d,J = 2.4 Hz, 1H), 4.08 (s, 3H), 3.65-3.61 (m, 4H), 3.37 (d, J = 8.4 Hz, 1H), 3.31 (s,3H), 3.01-2.88 (m, 4H), 2.27-2.24 (m, 1H), 2.20-2.17 (m, 1H)217m/z (ESI, +ve ion) = 479.1 [M + H]+.1H NMR (400 MHz, DMSO) δ: 12.64 (s, 1H),10.44 (s, 1H), 9.08 (s, 1H), 8.55 (s, 1H), 7.64 (d, J = 7.3 Hz, 1H), 7.49-7.55 (m,2H), 7.41 (s, 1H), 7.35 (br. s., 1H), 6.89 (d, J = 7.1 Hz, 1H), 6.75 (d, J = 8.8 Hz,1H), 6.60 (d, J = 8.5 Hz, 1H), 5.75 (s, 1H), 4.07 (s, 3H), 3.36 (s, 3H), 3.21 (t,J = 9.7 Hz, 1H), 2.33 (s, 1H), 1.99 (s, 1H)218m/z (ESI, +ve ion) = 515.2 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.26 (s, 1H),10.37 (s, 1H), 8.44-8.25 (m, 2H), 7.94-7.77 (m, 1H), 7.41-7.19 (m, 2H), 6.97-6.79 (m, 1H), 6.78-6.62 (m, 1H), 6.62-6.45 (m, 1H), 5.64 (s, 1H), 3.97 (s, 3H),3.72 (s, 3H), 3.15-3.06 (m, 1H), 2.34-2.20 (m, 1H), 2.02-1.83 (m, 1H)219m/z (ESI, +ve ion) = 528.2 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.27 (s, 1H),10.37 (s, 1H), 9.60-9.49 (m, 1H), 8.48-8.28 (m, 2H), 7.94-7.80 (m, 1H), 7.59-7.46 (m, 1H), 7.36-7.23 (m, 1H), 6.93-6.79 (m, 1H), 6.76-6.63 (m, 1H), 6.58-6.45 (m, 1H), 5.63 (s, 1H), 4.80 (s, 1H), 4.05 (s, 3H), 3.15-3.07 (m, 1H), 3.07-2.99 (m, 1H), 2.33-2.20 (m, 1H), 1.97-1.86 (m, 1H), 0.96 (s, 6H)220m/z (ESI, +ve ion) = 511.4 [M + H]+. NMR (400 MHz, DMSO-d6) δ 12.22 (s, 1H),10.43 (s, 1H), 8.78 (d, J = 2.2 Hz, 1H), 8.10 (s, 1H), 7.92 (d, J = 8.4 Hz, 1H), 7.79(d, J = 2.2 Hz, 1H), 7.32 (s, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.74 (d, J = 8.4 Hz, 1H),6.58 (dd, J = 8.4, 2.4 Hz, 1H), 5.70 (d, J = 2.2 Hz, 1H), 3.98 (s, 3H), 3.79 (t,J = 7.0 Hz, 2H), 3.31 (s, 3H), 3.17 (t, J = 8.4 Hz, 1H), 2.46 (t, J = 8.0 Hz, 2H), 2.33(dd, J = 7.7, 4.7 Hz, 1H), 2.12-2.03 (m, 2H), 1.97 (dd, J = 8.9, 4.7 Hz, 1H)221m/z (ESI, +ve ion) = 534.4 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.33 (s,1H), 10.43 (s, 1H), 8.17 (s, 1H), 7.92 (s, 1H), 7.81 (d, J = 8.4 Hz, 1H), 7.34 (d, J =17.1 Hz, 2H), 7.18 (q, J = 5.0 Hz, 1H), 6.90 (d, J = 8.5 Hz, 1H), 6.74 (d, J = 8.4 Hz,1H), 6.58 (dd, J = 8.5, 2.5 Hz, 1H), 5.69 (d, J = 2.5 Hz, 1H), 3.92 (s, 3H), 3.31 (s,3H), 3.18 (t, J = 8.4 Hz, 1H), 2.43 (s, 3H), 2.39 (d, J = 5.0 Hz, 3H), 2.33 (dd, J = 7.9,4.7 Hz, 1H), 1.98 (dd, J = 9.0, 4.6 Hz, 1H)222m/z (ESI + ve ion) = 535.15 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.43 (s,1H), 10.42 (s, 1H), 8.69 (s, 1H), 8.43 (d, J = 8.0 Hz, 1H), 7.94 (d, J = 8.4 Hz, 1H),7.50 (d, J = 8.4 Hz, 1H), 7.38 (s, 1H), 6.95 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz,1H), 6.60-6.58 (m, 1H), 5.71 (d, J = 2.4 Hz, 1H), 4.06 (s, 3H), 3.32 (s, 3H), 3.21-3.17 (m, 1H), 2.81 (s, 6H), 2.35-2.32 (m, 1H), 2.01-1.96 (m, 1H).223m/z (ESI, +ve ion) = 549.10 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.45 (s,1H), 10.42 (s, 1H), 8.53 (s, 1H), 8.36 (d, J = 8.2 Hz, 1H), 7.87 (d, J = 8.4 Hz, 1H),7.47 (d, J = 8.1 Hz, 1H), 7.39 (s, 1H), 6.97-6.90 (m, 1H), 6.76 (d, J = 8.4 Hz, 1H),6.59 (dd, J = 8.4, 2.6 Hz, 1H), 5.69 (d, J = 2.6 Hz, 1H), 4.51 (q, J = 7.0 Hz, 2H),3.32 (s, 3H), 3.20 (t, J = 8.5 Hz, 1H), 2.81 (s, 6H), 2.34-2.32 (m, 1H), 2.00-1.98(m, 1H), 1.46 (t, J = 7.0 Hz, 3H)224m/z (ESI, +ve ion) = 549.10 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.45 (s,1H), 10.42 (s, 1H), 8.53 (s, 1H), 8.36 (d, J = 8.2 Hz, 1H), 7.87 (d, J = 8.4 Hz, 1H),7.47 (d, J = 8.1 Hz, 1H), 7.39 (s, 1H), 6.97-6.90 (m, 1H), 6.76 (d, J = 8.4 Hz, 1H),6.59 (dd, J = 8.4, 2.6 Hz, 1H), 5.69 (d, J = 2.6 Hz, 1H), 4.51 (q, J = 7.0 Hz, 2H),3.32 (s, 3H), 3.20 (t, J = 8.5 Hz, 1H), 2.81 (s, 6H), 2.34-2.32 (m, 1H), 2.00-1.98(m, 1H), 1.46 (t, J = 7.0 Hz, 3H225m/z (ESI, +ve ion) = 504.20 [M + H]+.1H NMR (400 MHz, Chloroform-d) δ7.93-7.90 (m, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.45 (d, J = 12 Hz, 2H), 6.93 (d, J = 8 Hz,1H), 6.84 (d, J = 4.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.66 (d, J = 2.4 Hz, 1H), 4.06 (s,3H), 3.42 (s, 1H), 3.36 (d, J = 8.8 3H), 2.26-2.20 (m, 1H), 2.19-2.16 (m, 1H), 1.81(s, 3H), 1.79 (s, 3H).226m/z (ESI, +ve ion) = 486.4 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.17 (s, 1H),10.41 (s, 1H), 8.53 (d, J = 2.1 Hz, 1H), 7.92-7.84 (m, 2H), 7.68 (d, J = 2.2 Hz,1H), 7.31 (s, 1H), 6.89-6.84 (m, 1H), 6.74 (d, J = 8.4 Hz, 1H), 6.58 (dd, J = 8.4,2.5 Hz, 1H), 5.70 (d, J = 2.5 Hz, 1H), 4.96 (s, 1H), 3.96 (s, 3H), 3.17 (t, J = 8.4 Hz,1H), 2.37-2.27 (m, 1H), 2.01-1.92 (m, 1H), 1.43 (s, 6H). 3 protons are underwater peak at 3.30 ppm227m/z (ESI, +ve ion) = 520.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.40 (s,1H), 10.41 (s, 1H), 8.64 (s, 1H), 8.27 (s, 1H), 7.92 (d, J = 8.4 Hz, 1H), 7.38 (s, 1H),6.93 (d, J = 8.4 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.60-6.58 (m, 1H), 5.71 (d, J =2.4 Hz, 1H), 4.04 (s, 3H), 3.33 (s, 3H), 3.31 (s, 3H), 3.19 (t, J = 8.4 Hz, 1H), 2.51 (s,3H), 2.34-2.33 (m, 1H), 2.00-1.97 (m, 1H).229m/z (ESI, +ve ion) = 505.10 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.02 (d,J = 2.8 Hz, 1H), 7.59 (d, J = 8.4 Hz, 1H), 7.44 (s, 1H), 6.95 (d, J = 8.8 Hz, 1H), 6.84(d, J = 8.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.62 (d, J = 2.4 Hz, 1H), 4.16 (s, 3H), 3.38(d, J = 8.4 Hz, 1H), 3.31 (s, 3H), 2.26-2.17 (m, 2H), 1.82 (s, 3H), 1.78 (s, 3H).230m/z (ESI, +ve ion) = 525.25 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.24 (d,J = 8.4 Hz, 1H), 7.73-7.67 (m, 2H), 7.40 (s, 1H), 6.94 (d, J = 8.4 Hz, 1H), 6.84 (d,J = 8.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.62 (d, J = 2.4 Hz, 1H), 4.20 (s, 3H), 3.38 (d,J = 8.4 Hz, 2H), 3.33-3.31 (s, 4H), 2.26-2.05 (m, 2H), 1.19-1.12 (m, 1H), 0.58-0.54 (m, 2H), 0.36-0.32 (m, 2H)231m/z (ESI + ve ion) = 513.25 [M + H]+. 1H-NMR (400 MHz, Methanol-d4) δ 8.23 (d,J = 8.4 Hz, 1H), 7.73-7.66 (m, 2H), 7.40 (s, 1H), 6.94 (d, J = 8.4 Hz, 1H), 6.84 (d,J = 8.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.62 (d, J = 2.4 Hz, 1H), 4.18 (s, 4H), 3.38 (d,J = 8.4 Hz, 1H), 3.30 (s, 3H), 2.26-2.17 (m, 2H), 1.31 (d, J = 6.4 Hz, 6H)232m/z (ESI + ve ion) = 567.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.31 (s,1H), 10.41 (s, 1H), 8.48-8.32 (m, 2H), 7.93 (d, J = 8.4 Hz, 1H), 7.51 (d, J = 8.2 Hz,1H), 7.36 (s, 1H), 6.92 (d, J = 8.8 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.59 (dd, J =8.4, 2.6 Hz, 1H), 5.71 (d, J = 2.6 Hz, 1H), 5.13 (s, 1H), 4.59 (d, J = 6.6 Hz, 1H),4.00 (s, 3H), 3.82 (s, 1H), 3.67 (d, J = 11.8 Hz, 3H), 3.32 (s, 3H), 3.21-3.17 (m,1H). 2.33 (m, 1H), 2.04-1.75 (m, 5H)233m/z (ESI, +ve ion) = 459.4 [M + H]+. NMR (400 MHz,) δ 12.45 (s, 1H), 10.42 (s,1H), 8.41 (s, 1H), 7.48 (d, J = 8.4 Hz, 1H), 7.35 (s, 1H), 6.87 (d, J = 8.5 Hz, 1H),6.74 (d, J = 8.4 Hz, 1H), 6.58 (dd, J = 8.5, 2.4 Hz, 1H), 6.29 (s, 1H), 5.67 (d,J = 2.4 Hz, 1H), 3.88 (s, 3H), 3.31 (s, 3H), 3.27 (s, 3H), 3.18 (t, J = 8.4 Hz, 1H),2.30 (dd, J = 7.8. 4.8 Hz, 1H), 1.97 (dd, J = 9.0, 4.6 Hz, 1H)235m/z (ESI, +ve ion) = 541.3 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.58 (s, 1H),10.44 (s, 1H), 8.75 (s, 1H), 7.83 (s, 1H), 7.47-7.37 (m, 2H), 7.34 (s, 1H), 6.87 (d,J = 8.4 Hz, 1H), 6.75 (d, J = 8.5 Hz, 1H), 6.59 (d, J = 8.3 Hz, 1H), 5.75 (s, 1H),4.59-4.37 (m, 1H), 4.31-4.08 (m, 5H), 3.93 (s, 3H), 3.88-3.74 (m, 2H), 3.26-3.11(m, 5H), 2.40-2.28 (m, 1H), 2.04-1.94 (m, 1H)236m/z (ESI, +ve ion) = 500.2 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.71 (s, 1H),10.47 (s, 1H), 9.31 (s, 1H), 7.78 (s, 1H), 7.54-7.34 (m, 2H), 6.90 (d, J = 8.5 Hz,1H), 6.77 (d, J = 8.5 Hz, 1H), 6.65-6.55 (m, 1H), 5.72 (s, 1H), 4.01 (s, 3H), 3.33(s, 3H), 3.25-3.10 (m, 4H), 2.98 (s, 3H), 2.38-2.29 (m, 1H), 2.05-1.96 (m, 1H)237m/z (ESI, +ve ion) = 514.3 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.69 (s, 1H),10.43 (s, 1H), 9.21 (s, 1H), 7.77 (s, 1H), 7.48-7.37 (m, 2H), 6.89 (d, J = 8.5 Hz,1H), 6.74 (d, J = 8.8 Hz, 1H), 6.67-6.52 (m, 1H), 5.71 (s, 1H), 4.52-4.38 (m, 2H),3.23-3.06 (m, 4H), 2.97 (s, 3H), 2.33 (s, 1H), 1.76 (s, 1H), 1.43 (s, 3H)238m/z (ESI, +ve ion) = 513.25 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 8.08 (s,1H), 7.70 (d, J = 8.4 Hz, 1H), 7.39 (s, 1H), 6.94-6.91 (m, 1H), 6.84 (d, J = 8.4 Hz,1H), 6.64-6.61 (m, 1H), 5.62 (d, J = 2.4 Hz, 1H), 4.06 (s, 3H), 3.38-3.36 (m, 1H),3.30 (s, 3H), 3.14 (s, 3H), 2.97 (s, 3H), 2.26-2.18 (m, 5H)239m/z (ESI, +ve ion) = 490.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.32 (s,1H), 10.42 (s, 1H), 8.53 (d, J = 8.4 Hz, 1H), 8.43 (s, 1H), 7.93 (d, J = 8.4 Hz, 1H),7.42 (d, J = 8.0 Hz, 1H), 7.36 (s, 1H), 6.93-6.91 (m, 1H), 6.75 (d, J = 8.4 Hz, 1H),6.60-6.57 (m, 1H), 5.71 (d, J = 2.4 Hz, 1H), 4.05 (s, 3H), 3.32 (s, 3H), 3.21-3.17(m, 1H), 2.78 (s, 3H), 2.34-2.37 (m, 1H), 2.00-1.97 (m, 1H)240m/z (ESI, +ve ion) = 490.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.32 (s,1H), 10.42 (s, 1H), 8.54 (d, J = 8.0 Hz, 1H), 8.43 (s, 1H), 7.94 (d, J = 8.4 Hz, 1H),7.42 (d, J = 8.4 Hz, 1H), 7.36 (s, 1H), 6.92 (d, J = 8.8 Hz, 1H), 6.75 (d, J = 8.4 Hz,1H), 6.60-6.57 (m, 1H), 5.71 (d, J = 2.4 Hz, 1H), 4.05 (s, 3H), 3.33 (s, 3H),3.21-3.17 (m, 1H), 2.78 (s, 3H), 2.35-2.32 (m, 1H), 2.08-1.97 (m, 1H)241m/z (ESI, +ve ion) = 506.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.43 (s,1H), 10.42 (s, 1H), 9.28 (s, 1H), 8.54 (s, 1H), 7.90 (d, J = 8.4 Hz, 1H), 7.58 (s, 1H),7.37 (s, 1H), 6.95-6.92 (m, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.60-6.58 (m, 1H), 5.71(d, J = 2.4 Hz, 1H), 4.09 (s, 3H), 3.33 (s, 3H), 3.32-3.21 (s, 4H), 2.35-2.32 (m,1H), 2.01-1.98 (m, 1H).242m/z (ESI, +ve ion) = 507.20 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.80 (s,1H), 10.42 (s, 1H), 9.76 (s, 1H), 8.03 (s, 1H), 7.44-7.41 (m, 2H), 6.92-6.90 (m,1H), 6.75 (d, J = 8.4 Hz, 1H), 6.60-6.57 (m, 1H), 5.71 (d, J = 2.4 Hz, 1H), 4.09 (s,3H). 3.33 (s, 3H), 3.21-3.15 (m, 4H), 2.35-2.31 (m, 1H), 2.00-1.97 (m, 1H)243m/z (ESI, +ve ion) = 551.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.27 (s,1H), 10.42 (s, 1H), 8.40 (d, J = 8.0 Hz, 1H), 8.32 (s, 1H), 7.93 (d, J = 8.8 Hz, 1H),7.35 (s, 1H), 7.27 (d, J = 8.4 Hz, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz,1H), 6.60-6.57 (m, 1H), 5.71 (d, J = 2.4 Hz, 1H), 3.98 (s, 3H), 3.32-3.21 (m, 3H),3.19 (t, J = 8.0 Hz, 1H), 2.87 (s, 2H), 2.34-2.31 (m, 1H), 2.05-1.97 (m, 1H), 0.69(s, 4H), 0.59 (s, 4H).244m/z (ESI, +ve ion) = 535.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.35 (s,1H), 10.42 (s, 1H), 8.60-8.57 (m, 1H), 8.52 (s, 1H), 8.42 (d, J = 8.0 Hz, 1H), 7.93(d, J = 8.4 Hz, 1H), 7.65 (d, J = 8.0 Hz, 1H), 7.36 (s, 1H), 6.92 (d, J = 8.4 Hz, 1H),6.75 (d, J = 8.4 Hz, 1H), 6.60-6.52 (m, 1H), 6.29-6.01 (m, 1H), 5.71 (d, J = 2.8Hz, 1H), 4.14 (s, 3H), 3.78-3.66 (m, 2H), 3.30 (s, 3H), 3.21-3.17 (m, 1H), 2.34-2.31 (m, 1H), 2.00-1.97 (m, 1H)245m/z (ESI, +ve ion) = 567.30 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.23 (s,1H), 10.41 (s, 1H), 8.40 (d, J = δ 0 Hz, 1H), 8.24 (s, 1H), 7.93 (d, J = 8.4 Hz, 1H),7.34 (s, 1H), 7.13 (d, J = 8.0 Hz, 1H), 6.92-6.90 (m, 1H), 6.76 (d, J = 8.4 Hz, 1H),6.60-6.57 (m, 1H), 5.71 (d, J = 2.4 Hz, 1H), 4.53 (s, 2H), 4.01 (s, 3H), 3.32 (d, J =5.6 Hz, 3H), 3.20 (t, J = 8.4 Hz, 1H), 2.34-2.31 (m, 1H), 2.01-1.97 (m, 1H), 1.86(d, J = 12 Hz, 1H), 1.61 (d, J = 5.2 Hz, 4H), 1.49 (d, J = 2.8 Hz, 1H), 1.29 (d, J = 6.8Hz, 6H)246m/z (ESI, +ve ion) = 567.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.29 (s,1H), 10.42 (s, 1H), 8.41-8.37 (m, 2H), 7.93 (d, J = 8.4 Hz, 1H), 7.35 (s, 1H), 7.30(d, J = 8.1 Hz, 1H), 6.92 (d, J = 8.0 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.59 (d, J =2.4 Hz, 1H), 5.71 (d, J = 2.6 Hz, 1H), 4.39-4.36 (m, 1H), 4.28-4.26 (m, 1H),4.13-4.09 (m, 1H), 4.01 (s, 4H), 3.32-3.29 (m, 5H), 3.19 (t, J = 8.4 Hz, 1H), 3.00-2.98(m, 1H). 2.34-2.31(m, 1H), 2.00-1.93(m, 1H), 1.83-1.80 (m, 3H).247m/z (ESI, +ve ion) = 510.10 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.82 (s,1H), 10.42 (s, 1H), 9.55 (s, 1H), 8.29 (d, J = 2.1 Hz, 1H), 8.19 (d, J = 2.2 Hz, 1H),7.45 (s, 1H), 7.36 (d, J = 8.4 Hz, 1H), 6.91 (d, J = 8.3 Hz, 1H), 6.75 (d, J = 8.4 Hz,1H), 6.60 (dd, J = 8.4, 2.6 Hz, 1H), 5.71 (d, J = 2.6 Hz, 1H), 3.34 (s, 3H), 3.25 (s,3H), 3.20 (t, J = 8.4 Hz, 1H), 2.34-2.32 (m, 1H), 2.08-1.94 (m, 1H)249m/z (ESI, +ve ion) = 504.15 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.29 (s,1H), 10.42 (s, 1H), 9.32 (s, 1H), 8.23 (s, 1H), 7.90 (d, J = 8.3 Hz, 1H), 7.51 (d, J =5.9 Hz, 1H), 7.34 (s, 1H), 6.91 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.59(dd, J = 8.5, 2.6 Hz, 1H), 5.71 (d, J = 2.6 Hz, 1H), 4.04 (s, 3H), 3.33 (s, 3H), 3.19 (t,J = 8.5 Hz, 1H), 2.35-2.32 (m, 1H), 2.01-1.97 (m, 1H), 1.63 (d, J = 13.5 Hz, 6H)250m/z (ESI, +ve ion) = 549.20 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 8.25 (d,J = 8.1 Hz, 1H), 7.73-7.70 (m, 2H), 7.40 (s, 1H), 6.95-6.93 (m, 1H), 6.84 (d, J =8.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.62 (d, J = 2.4 Hz, 1H), 4.78-4.76 (m, 1H),4.73-4.69 (m, 1H), 4.66-4.63 (m, 1H), 4.60-4.52 (m, 2H), 4.18 (s, 3H), 3.39-3.34(m, 1H), 3.31 (s, 3H), 2.27-2.23 (m, 1H), 2.21-2.17 (m, 1H)251m/z (ESI, +ve ion) = 503.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.56 (s,1H), 10.43 (s, 1H), 8.47 (s, 1H), 8.17 (d, J = 8.4 Hz, 1H), 7.64 (d, J = 8.4 Hz, 1H),7.56 (d, J = 8.4 Hz, 1H), 7.41 (s, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.77 (d, J = 8.4 Hz,1H), 6.61-6.53 (m, 1H), 5.71 (d, J = 2.4 Hz, 1H), 3.35 (s, 3H), 3.22-3.18 (m, 1H),3.07 (s, 3H), 2.99 (s, 3H), 2.35-2.32 (m, 1H), 2.08-1.91 (m, 1H)252m/z (ESI, +ve ion) = 486.20 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.33 (s,1H), 10.41 (s, 1H), 8.17 (s, 1H), 8.05 (s, 1H), 7.77-7.75 (m, 2H), 7.38 (s, 1H),6.93-6.91 (m, 1H), 6.76 (d, J = 8.0 Hz, 1H), 6.61-6.58 (m, 1H). 5.71 (d, J = 2.4 Hz,1H), 3.34 (s, 3H), 3.25 (t, J = 8.4 Hz, 1H), 2.57 (s, 3H), 2.34-2.31 (m, 1H),2.00-1.98 (m, 1H)253m/z (ESI, +ve ion) = 499.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.24 (s,1H), 10.42 (s, 1H), 9.18 (s, 1H), 8.14 (s, 1H), 7.91 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H),7.26 (s, 1H), 6.91 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.62-6.55 (m, 1H),5.71 (d, J = 2.4 Hz, 1H), 4.00 (s, 3H), 3.33 (s, 3H), 3.19 (t, J = 8.4 Hz, 1H), 3.07 (s,3H), 3.00 (s, 3H), 2.37-2.29 (m, 1H), 2.03-1.95 (m, 1H)254m/z (ESI + ve ion) = 533.20 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.57 (s,1H), 10.41 (s, 1H), 8.93 (s, 1H), 7.52 (s, 1H), 7.38 (d, J = 6.6 Hz, 2H), 6.87 (d, J =8.5 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.62-6.55 (m, 1H), 5.71 (d, J = 2.4 Hz, 1H),4.42 (t, J = 8.7 Hz, 4H), 4.06 (s, 3H), 3.88-3.79 (m, 1H), 3.33 (s, 3H), 3.18 (t, J =8.5 Hz, 1H), 2.33-2.30 (m, 1H), 2.00-1.97 (m, 1H)255m/z (ESI, +ve ion) = 577.15 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.96 (d,J = 2.0 Hz, 1H), 7.60 (d, J = 8.4 Hz, 1H), 7.44 (s, 1H), 7.35 (d, J = 1.6 Hz, 1H), 6.96(d, J = 8.4 Hz, 1H), 6.84 (d, J = 8.4 Hz, 1H), 6.64-6.62 (m, 1H), 5.65 (d,J = 2.4 Hz, 1H), 4.07 (s, 3H), 3.75-3.73 (m, 4H), 3.37 (d, J = 8.4 Hz, 1H), 3.32(s, 3H), 3.04-3.01 (m, 4H), 2.27-2.25 (m, 1H). 2.19-2.14 (m, 1H)256m/z (ESI, +ve ion) = 603.20 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 7.94 (d,J = 1.9 Hz, 1H), 7.59 (d, J = 8.4 Hz, 1H), 7.44 (s, 1H), 7.33 (d, J = 2.0 Hz, 1H), 6.95(d, J = 7.6 Hz, 1H), 6.84 (d, J = 8.4 Hz, 1H), 6.65-6.62 (m, 1H), 5.65 (d,J = 2.8 Hz, 1H), 4.39 (s, 2H), 4.07 (s, 3H), 3.51-3.50 (m, 3H), 3.38 (d,J = 11.2 Hz, 3H), 2.67 (d, J = 10.8 Hz, 2H), 2.27-2.24 (m, 1H), 2.21-2.17 (m, 1H),2.00-1.93 (m, 4H)257m/z (ESI, +ve ion) = 532.60 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.35 (s,1H), 10.43 (s, 1H), 8.42-8.40 (m, 2H), 7.92 (d, J = 8.4 Hz, 1H), 7.49-7.47 (m,1H), 7.36 (s, 1H), 6.93-6.91 (m, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.60-6.58 (m, 1H),5.71 (d, J = 2.4 Hz, 1H), 4.02 (s, 3H), 3.32 (d, J = 2.0 Hz, 3H), 3.19 (t, J = 8.4 Hz,1H), 2.35-2.32 (m, 1H), 2.01-1.87 (m, 5H), 1.03-0.95 (m, 6H)259m/z (ESI, +ve ion) = 576.15 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.02 (d,J = 1.9 Hz, 1H), 7.59 (d, J = 8.4 Hz, 1H), 7.44 (d, J = 2.0 Hz, 2H), 6.95 (d,J = 8.8 Hz, 1H), 6.84 (d, J = 8.8 Hz, 1H), 6.65-6.62 (m, 1H), 5.65 (d, J = 2.4 Hz, 1H),4.08 (s, 3H), 4.04-4.02 (m, 2H), 3.46-3.42 (m, 4H), 3, 40-3.37 (m, 3H), 2.27-2.24(m, 1H), 2.20-2.17 (m, 1H), 1.91-1.81 (m, 2H), 1.80-1.70 (m, 2H)260m/z (ESI, +ve ion) = 453.2 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.52 (s, 1H),10.44 (s, 1H), 8.89 (s, 1H), 8.41-8.29 (m, 1H), 7.96-7.88 (m, 1H), 7.63-7.54(m, 1H), 7.38 (s, 1H), 6.95-6.91 (m, 1H), 6.80-6.70 (m, 1H), 6.58 (s, 1H), 5, 69(s, 1H), 4.04 (s, 3H), 3.32 (s, 3H), 3.21-3.15 (m, 1H), 2.33 (s, 1H), 2.00 (s, 1H)261[0059] m/z (ESI, +ve ion) = 531.20 [M + H]+.1H NMR (400 MHz, Methanol-d4)6 8.17-8.13 (m, 1H), 7.61 (d, J = 8.4 Hz, 1H), 7.37 (s, 1H), 7.29-7.25 (m, 1H),7.16-7.13 (m, 1H), 6.89-6.82 (m, 2H), 6.63-6.60 (m, 1H), 5.62 (d, J = 2.4 Hz,1H), 4.03 (s, 3H), 3.37-3.29 (m, 4H), 2.24-2.19 (m, 1H), 2.18-2.15 (m, 1H),2.00-1.88 (m, 4H), 1.12-1.01 (m, 6H)262m/z (ESI, +ve ion) = 520.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12, 43 (s,1H), 10.41 (s, 1H), 9.29 (s, 1H), 8.55 (s, 1H), 7.90 (d, J = 8.4 Hz, 1H), 7.57 (s, 1H),7.37 (s, 1H), 6.93 (d(d, J = 8.4, 1, 4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.59 (dd, J =8.5, 2.6 Hz, 1H), 5.71 (d, J = 2.6 Hz, 1H), 4.09 (s, 3H), 3.37-3.34 (m, 2H), 3.33 (s,3H), 3.19 (t, J = 8.5 Hz, 1H), 2.34 (dd, J = 7.9, 4.7 Hz, 1H), 1.99 (dd, J = 9.0,4.7 Hz, 1H), 1.14 (t, J = 7.4 Hz, 3H)263m/z (ESI, +ve ion) = 511.3 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.57 (s, 1H),10.43 (s, 1H), 8.72 (s, 1H), 7.88-7.76 (m, 1H), 7.44-7.37 (m, 2H), 7.37-7.28(m, 1H), 6.96-6.82 (m, 1H), 6.81-6.70 (m, 1H), 6.66-6.53 (m, 1H), 5.75 (s, 1H),4.43-4.27 (m, 2H), 4.10-3.96 (m, 2H), 3.92 (s, 3H), 3.22-3.10 (m, 1H), 2.39-2.30 (m, 1H), 2.30-2.17 (m, 2H), 2.05-1.91 (m, 1H)264m/z (ESI, +ve ion) = 541.3 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.54 (s, 1H),10.43 (s, 1H), 8.61 (s, 1H), 7.61 (s, 1H), 7.46-7.40 (m, 1H), 7.39 (s, 1H), 7.21 (s,1H), 6.96-6.79 (m, 1H), 6.81-6.69 (m, 1H), 6.68-6.52 (m, 1H), 5.74 (s, 1H),3.91 (s, 3H), 3.65-3.56 (m, 4H), 3.56-3.49 (m, 4H), 3.22-3.13 (m, 1H), 2.39-2.28 (m, 1H), 2.03-1.93 (m, 1H)265m/z (ESI, +ve ion) = 496.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.24 (s,1H), 10.42 (s, 1H), 8.26 (s, 1H), 8.07 (s, 1H), 7.85 (d, J = 8.0 Hz, 1H), 7.34 (s, 1H),7.19 (s, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.60-6.58 (m, 1H),5.71 (d, J = 2.4 Hz, 1H), 4.34 (s, 2H), 3.98 (s, 3H), 3.35 (s, 3H), 3.19-3.14 (m, 1H),3.04 (s, 3H), 2.34-2.31 (m, 1H), 2.01-1.97 (m, 1H)266m/z (ESI, +ve ion) = 496.20 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ 12.27 (s,1H), 10.41 (s, 1H), 8.16 (s, 1H), 8.04 (d, J = 8.4 Hz, 1H), 7.80 (d, J = 8.4 Hz, 1H),7.36 (s, 1H), 7.28 (d, J = 8.4 Hz, 1H), 6.91-6.89 (m, 1H), 6.76 (d, J = 8.4 Hz, 1H),6.60-6.57 (m, 1H), 5.70 (d, J = 2.8 Hz, 1H), 4.64 (s, 2H), 3.99 (s, 3H), 3.33 (s, 3H),3.19 (t, J = 8.0 Hz, 1H), 3.06 (s, 3H), 2.34-2.30 (m, 1H), 2.00-1.97 (m, 1H)267m/z (ESI, +ve ion) = 563.25 [M + H]+.1H NMR (400 MHz, Methanol-d4) δ 8.08 (s,1H), 7.59 (d, J = 8.4 Hz, 1H), 7.47-7.44 (m, 2H), 6.97 (d, J = 8.4 Hz, 1H), 6.85 (d,J = 8.4 Hz, 1H), 6.64-6.62 (m, 1H), 5.65 (d, J = 2.4 Hz, 1H), 4.01 (s, 3H),3.94-3.88 (m, 2H), 3.69-3.66 (m, 2H), 3.38-3.33 (m, 4H), 2.27-2.14 (m, 4H)268m/z (ESI, +ve ion) = 547.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.66 (s,1H), 10.41 (s, 1H), 9.16 (s, 1H), 7.90 (d, J = 2.0 Hz, 1H), 7.46-7.42 (m, 2H), 7.33(d, J = 1.6 Hz, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 6.61-6.58(m, 1H), 5.74 (d, J = 2.8 Hz, 1H), 4.01 (s, 3H), 3, 70-3.67 (m, 4H), 3.34-3.285 (m,3H), 3.21-3.19 (m, 1H), 2.35-2.33 (m, 1H), 2.04-1.99 (m, 3H)269m/z (ESI, +ve ion) = 577.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.64 (s,1H), 10.41 (s, 1H), 9.07 (s, 1H), 7.88 (d, J = 2.0 Hz, 1H), 7.40-7.36 (m, 3H), 6.90(d, J = 8.4 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.59 (dd, J = 8.5, 2.6 Hz, 1H), 5.72 (d,J = 2.5 Hz, 1H), 4.56 (d, J = 6.0 Hz, 2H), 4.12 (d, J = 6.0 Hz, 2H), 3.97 (s, 3H), 3.34(s, 3H), 3.19 (t, J = 8.4 Hz, 1H), 2.34-2.31 (m, 1H), 2.00-1.97 (m, 1H), 1.46 (s,3H)270m/z (ESI, +ve ion) = 591.30 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.67 (s,1H), 10.42 (s, 1H), 9.15 (s, 1H), 7.95 (d, J = 1.9 Hz, 1H), 7.41 (d, J = 7.8 Hz, 2H),7.33 (d, J = 2.0 Hz, 1H), 6.90 (d, J = 8.5 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.59 (dd,J = 8.4, 2.6 Hz, 1H), 5.74 (d, J = 2.6 Hz, 1H), 4.77 (d, J = 6.0 Hz, 2H), 4.13 (d, J =6.0 Hz, 2H), 3.82 (s, 3H), 3.35 (s, 3H), 3.19 (t, J = 8.5 Hz, 1H), 2.50 (s, 3H),2.34-2.32 (m, 1H), 2.00-1.97 (m, 1H), 1.56 (s, 3H)271m/z (ESI, +ve ion) = 472.3 [M + H]+.1H NMR (400 MHz, DMSO) δ 12.19 (s, 1H),10.43 (s, 1H), 8.44 (s, 1H), 7.97 (s, 1H), 7.94-7.86 (m, 1H), 7.55 (s, 1H), 7.33 (s,1H), 6.94-6.83 (m, 1H), 6.79-6.71 (m, 1H), 6.64-6.52 (m, 1H), 5.70 (s, 1H),5.12 (s, 1H), 4.76-4.59 (m, 1H), 3.97 (s, 3H), 3.34-3.27 (m, 6H), 3.23-3.11 (m,1H), 2.38-2.29 (m, 1H), 2.02-1.92 (m, 1H)273m/z (ESI, I've ion) = 604.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.69 (s,1H), 10.42 (s, 1H), 9.06 (s, 1H), 7.83 (s, 1H), 7.42 (d, J = 4.8 Hz, 2H), 7.23 (s, 1H),6.90 (d, J = 8.4 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.63-6.56 (m, 1H), 5.74 (d, J =1.6 Hz, 1H), 4.29-4.19 (m, 2H), 3.34 (s, 3H), 3.19 (t, J = 8.4 Hz, 1H), 2.95-2.92(m, 4H), 2.50 (s, 1H), 2.38-2.31 (s, 4H), 2.17 (s, 3H), 2.03-1.95 (m, 1H), 1.44 (t,J = 6.8 Hz, 3H)275m/z (ESI, +ve ion) = 535.15 [M + H]+.1H-NMR (400 MHz, Methanol-d4) δ 8.01 (d,J = 2.0 Hz, 1H), 7.59 (d, J = 8.4 Hz, 1H), 7.43-7.41 (m, 2H), 6.96-6.94 (m, 1H),6.84 (d, J = 8.4 Hz, 1H), 6.64-6.61 (m, 1H), 5.66 (d, J = 2.8 Hz, 1H), 4.32-4.27(m, 2H), 3.39-3.36 (m, 1H), 3.32 (s, 3H), 2.56 (s, 3H), 2.26-2.23 (m, 1H), 2.20-2.17 (m, 1H), 1.56 (t, J = 6, 8 Hz, 3H)277m/z (ESI, +ve ion) = 473.25 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ 12.45 (s,1H), 10.41 (s, 1H), 8.33 (s, 1H), 7.49 (d, J = 8.8 Hz, 1H), 7.36 (s, 1H), 6.88 (d, J =8.4 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 6.59-6.57 (m, 1H), 6.25 (s, 1H), 5.68 (d, J =2.4 Hz, 1H), 4.14 (d, J = 7.2 Hz, 2H), 3.33 (s, 3H), 3.27 (s, 3H), 3.20-3.19 (m,1H), 2, 32-2.29 (m, 1H), 2.00-1.96 (m, 1H), 1.39 (t, J = 6.8 Hz, 3H) Biological Activity Examples Biological Activity Example No. 1: PLK4 Biochemical Assay Activity of human recombinant PLK4 (ThermoFisher, cat #PV6396) was measured by quantification of adenosine diphosphate (ADP) using the ADP-Glo Kinase Assay Kit (Promega, cat #V9102). Test compounds were solubilized in dimethyl sulfoxide (DMSO) and dispensed into 384-well white polystyrene nonbinding plates (Greiner, cat #781094) using the Echo acoustic dispenser (Labcyte Inc.) in a 11-point 3-fold titration in duplicates. 5 μL of 1.0 nM PLK4 protein in assay buffer (50 mM HEPES. pH 7.5, 0.01% Brij-35, 0.01% BSA, 10 mM MgCl2, 1 mM EGTA, 1 mM DTT) was added to the plates. Test compounds and PLK4 were incubated for 15 minutes at room temperature (RT). Then 5 μL of a 16 μM adenosine triphosphate (ATP) (Promega, cat #V915B) and 9.3 μM Myelin Basic Protein (MBP) (SignalChem, cat #M42-51N) substrate solution in assay buffer was added and the reaction mixture was incubated for 6 hours at RT. The final concentration of PLK4. ATP and MBP in the reactions were 0.5 nM, 8.0 μM and 4.7 μM, respectively. Reactions were stopped and the remaining ATP depleted by adding 10 μL of ADP-Glo reagent (Promega, cat #V912B) and incubating for 40 minutes at RT. The simultaneous conversion of the remaining ADP to ATP and measurement of the newly synthesized ATP was achieved by addition of 20 μL Kinase Detection reagent (Promega, eat #V914B), incubation for 30 min at RT, and luminescence detection using the EnVision plate reader (PerkinElmer). Reactions lacking PLK4 were used as 100% inhibition controls. Reactions containing DMSO alone were used as 0% inhibition controls. The IC50values reported in Table 2 were determined using four parameter non-linear regression curve fit. Biological Activity Example No. 2: CHP134 CellTiter-Glo (CTG) assay CHP-134 cells (DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig Germany) were cultured in RPMI 1640 supplemented with 10% fetal bovine serum, penicillin (100 U/ml), 1% L-Glutamine and streptomycin (100 mg/ml). Cells were seeded (200 cells/well) in 384-well plates for 16 hours. On day two, nine serial 1:3 compound dilutions were made in DMSO in a 96-well plate. The compounds were then further diluted into growth media using a BRAVO robot (Agilent, Santa Clara, CA). The diluted compounds were then added to quadruplicate wells in the 384-well cell plate and incubated at 37° C. and 5% CO2. After 5 days, relative numbers of viable cells were measured by luminescence using CellTiter-Glo® (Promega) according to the manufacturer's instructions and read on a SPARK Multimode Microplate Reader (Tecan, Mannedorf Switzerland). The IC50calculations for the values reported in Table 2 were carried out using Prism 6.0 software (GraphPad, San Diego). Biological Activity Example No. 3: Aurora a Kinase Biochemical Assay Activity of human recombinant Aurora A (ThermoFisher, cat #PR5935A) was measured by quantification of adenosine diphosphate (ADP) using the ADP-Glo Kinase Assay Kit (Promega, cat #V9102). Test compounds were solubilized in dimethyl sulfoxide (DMSO) and dispensed into 384-well white polystyrene nonbinding plates (Greiner, cat #781094) using the Echo acoustic dispenser (Labcyte Inc.) in a 1-point 3-fold titration in duplicates. 5 μL of 5.0 nM Aurora A in assay buffer (50 mM HEPES, pH 7.5, 0.01% Brij-35, 0.01% BSA, 10 mM MgCl2, 1 mM EGTA, 1 mM DTT) was added to the plates. Test compounds and Aurora A were incubated for 15 minutes at room temperature (RT). Then 5 μL of a 40 μM adenosine triphosphate (ATP) (Promega, cat #V915B) and 9.3 uM Myelin Basic Protein (MBP) (SignalChem, cat #M42-51N) substrate solution in assay buffer was added and the reaction mixture was incubated for 2 hours at RT. The final concentration of Aurora A, ATP and MBP in the reactions were 2.5 nM, 20 μM and 4.7 μM, respectively. Reactions were stopped and the remaining ATP depleted by adding 10 uL of ADP-Glo reagent (Promega, cat #V912B) and incubating for 40 minutes at RT. The simultaneous conversion of the remaining ADP to ATP and measurement of the newly synthesized ATP was achieved by addition of 20 μL Kinase Detection reagent (Promega, cat #V914B), incubation for 30 min at RT, and luminescence detection using the EnVision plate reader (PerkinElmer). Reactions lacking Aurora A were used as 100% inhibition controls. Reactions containing DMSO alone were used as 0% inhibition controls. The IC50values reported in Table 2 were determined using four parameter non-linear regression curve fit. Biological Activity Example No. 4: Aurora B Kinase Biochemical Assay Activity of human recombinant Aurora B (ThermoFisher, cat #PR9210B) was measured by quantification of adenosine diphosphate (ADP) using the ADP-Glo Kinase Assay Kit (Promega, cat #V9102). Test compounds were solubilized in dimethyl sulfoxide (DMSO) and dispensed into 384-well white polystyrene nonbinding plates (Greiner, cat #781094) using the Echo acoustic dispenser (Labcyte Inc.) in a 1-point 3-fold titration in duplicates. 5 μL of 20 nM Aurora B in assay buffer (50 mM HEPES, pH 7.5, 0.01% Brij-35, 0.01% BSA, 10 mM MgCl2. 1 mM EGTA, 1 mM DTT) was added to the plates. Test compounds and Aurora B were incubated for 15 minutes at room temperature (RT). Then 5 μL of a 228 μM adenosine triphosphate (ATP) (Promega, cat #V915B) and 9.3 M Myelin Basic Protein (MBP) (SignalChem, cat #M42-51N) substrate solution in assay buffer was added and the reaction mixture was incubated for 2 hours at RT. The final concentration of Aurora b, ATP and MBP in the reactions were 10 nM, 114 μM and 4.7 μM, respectively. Reactions were stopped and the remaining ATP depleted by adding 10 uL of ADP-Glo reagent (Promega, cat #V912B) and incubating for 40 minutes at RT. The simultaneous conversion of the remaining ADP to ATP and measurement of the newly synthesized ATP was achieved by addition of 20 μL Kinase Detection reagent (Promega, cat #V914B), incubation for 30 min at RT, and luminescence detection using the EnVision plate reader (PerkinElmer). Reactions lacking Aurora B were used as 100% inhibition controls. Reactions containing DMSO alone were used as 0% inhibition controls. The IC50values reported in Table 2 were determined using four parameter non-linear regression curve fit. As shown in Table 2, many of the compounds of Formula (I), (Ia), (Ib), (II), and (III) demonstrated potent inhibition of PLK4 and less potent inhibition of Aurora A kinase and Aurora B kinase. As such, the compounds of Formula (I), (Ia), (Ib), (II), and (III) demonstrated selective inhibition of PLK4. As also set forth in Table 2, many of the compounds of Formula (I), (Ia), (Ib), (II), and (III) demonstrated, surprisingly and unexpectedly, greater selectivity in the inhibition of PLK4 versus inhibition of Aurora A kinase and/or Aurora B kinase than the CFI-400495compound. In Table 2, ND means not determined. TABLE 2PLK4 IC50CTG IC50[Aurora A IC50(nM)]/[Aurora B IC50(nM)]/Ex. No.(nM)(nM)[PLK4 IC50(nM)][PLK4 IC50(nM)]13.495229.34318.0315.654439.94843.955465.486711.4513352.178618.1874.559693.573247.269148492.317.6091832.9880290.694499.734791.187240.6394101.587438.1954.6314103.0876114.8762952.33277.27677.87121124.9592025.5132.99667.558510.304351436.0374.160423.8662231545.66162.181605.685511.76066174.3041145.91134.897771822.91924.81202.254262.74489.796306.83232141.732239.8235.04845.9215.5309170.7409242.2552534.18249.648928.10123263.853272.083280.375295.9673031.2436.203596.18758313.02207.05322.16887323.938911.884217.5546335.911355.94658.474031343.215893.312619.95956357.52567.295682.162126366.173218.20835.7006323712.461799.35849.510433814.0943.690562.7608233926.4330.382141.0181614015.2167.061142.783037412.711844.12257.47471.41284236.241053.80842.301324314.491347.13621.904764420.661269.11945.111334515.49137.83094.60297462.5211428.992542.642135.8985471.302610.51879.416104.3011183.277741.51948.123292.34064919.1253.403153.193725061.69361.80927.4923511.232143.33031.6561241.883527.01122602009.69955.0706531.2514028.777251.79865433.24100001504.212117.2684551.2473251.80417.89896563.83726831047.694423.7686575.083959.7983527.559831.2992582.316157.41393.782140.6736598.442108.469639.23241607.35569.694097.854521619.7452385588.8148955.05396216.9110000540.9225946.77716372.2256.8402116.685136486.96210.211698.838556544.1982.484735.35641566234021.367527.606838671.3424394.3154976.155499.9255687.3824764761.5181347.967692.981686.11785.978480.7112701.10180.335051376.93512.7157713.062988.6843420.966738.66754720.99742.712006.018405.2156737.045100003290.277346.7708743.312293.055637.1376875157.2173.409723.27608760.43493.462143304.147535.4839771.757725.16738.7591577.689780.3561.3739656601778.8577991.86210.53786.98563802.365293.5346.511654.12262813.6339651478.393220.4239826.207187.852438.55325832.238277.2908.4004333.3333841.278394.839123.633151.017851.91107.25554.9743067.5398621.2459.31414.623669.3396871.58235.2973.4177133.9241882.6721813.3081997.75444.7979892.317100001736.729314.32899017.811505.33444.32903914.0193202.9436.42779.87061928.838132.835576.363439324.341733.3611055.053945.6124602533.85641.732951.3063192386.67754.264939611.3556.55534511.3656351.8943971.139221.649309.2186520.2809987.55465.76947787.0003305.5335995.802565.24155.05129.97241001.734566.93382.0069230.449810111.332288.91271.8451002.6481026.162114.735574.699771032.796416.6667977.11021075.704729.71455.295174.42151084.36610913889.143230.64591096.759187150000110701103.908679.6315232.19041111.535234.7231144.495111223.7197.5105121.898711310.6525171888.2629231.73701142.09110000590.1482811.57341158.0393462132.6035186.34151162.89133.5788.5813483.73701172.321108.71388.1947906.50581187.3212359457.0413902.33571191.5882664870.2770973.55161203.81510000444.0366684.66571211.7945324875.6967230.880712273.111.170911.865931234.7982110667.569884.0975412413.5738.0324235.895351250.8331159.61323.9707495.73881269.014459.366.219292.0568127219622.7686722.76861282.88714922750.25971337.028012934.5218.105417.1349130603.158.481160.189013153.78105.708440.85161322.1496185290.8329249.51141338.322192.843.391045.14531341.79782.48849.1620969.273713570.7728.331262.611213619.432573.3401979.413213797.2627213827.33703613931757.49.614016.56775881453714113.13327.4181891.089114221.4813.81826.30814384.7414.62140.6071441.413750.1852.795561.43671455.81929151001.5467280.80431461.491655.13512.4078745.80821474.22313032292.2093483.06891486.873569397.3799338.86461493.153844.23193.7837879.47991508.1283156720.7185366.75691519.74361.244096.31531527.156502.7949107.81161531.906875.61254.4596123.60971540.976260.11373.9754655.22541551.035192.2305.410668.0773156493.81571.947287.9510.6317303.95481588.591704.923851.72861591.9621298309.735058.05301601.932310.2545.5487149.58591612.21351.7291.4932258.41631628.394648.6776105.28951631.524226.32164.0420389.43571646.2771637397.0049161.22351654.6821063111.255976.93291660.80280.61151.371668.45391671.18212.3295.5932916.94921681.9661620946.5921265.00511690.995188.5666.9347431.45731701.358325.7694.1090587.92341715.87115171176.6309259.24031724.091263324.94202.961732.666331.7301.0878314.92871741.921392.3365.8511353.77411753.569220914009.52658627.06641762.84315521725.29021300.38691774.255367511750.881311750.88131783.294630.815179.113515179.11351793.81194813119.91605384.41351802.854740.85455.50111996.84651817.5762415.52273693.24181824.45453.6821198.87741831.245125534.2169322.57031846.9988827144.89857144.89851851.9561343240.7975180.16361861.779537641.9337232.54641876.116563109.5814112.27931886.277607974.4817191.70651891.045112383.7321514.54551902.6615115129.5378279.14321912.2664942416.593184.99561921.63911802835.2654678.46251931.1343901463.8448138.88891940.965126805.6995103.73061951.561603459.6154740.38461963.3181673732.3689100.81371971.07610821243.494473.81971988.692100005752.41605752.41601990.716108439.944157.70952003.256906.0197523.34152011.706455485.6389229.07392022.684221031.3433321.41792031.0423242922.2649333.78122040.81122342.9630222.83952051.6423445737.5152235.74912060.806618.858680.22332070.952497.3739111.65972080.8967402790424712091.537605.8273.2595254.65192101.34865.47462.3887316.54302118.658531.9935218.41072121.585161353.12302802132.131189.792.2102225.38712141.01132.23953.0168159.09992152.098191.6986.653953.574832162.688766.47712.0535718601.190482171.189450.2416.7367170.98402180.95573.81806.596870.85862192.504510.1763.578264.25712206.0131042.4081498.75272212.802848.9511.063521463.59742221.359456446.1368546.43112232.304163.11004.7743445.31252244.016202312450.199212450.19922256.1181884.6028381.49722264.74721462759.6376474.61552272.209532.3917.1570478.94972281.347180.4813.6599340.46022297.8471669.4278130.87802302.434394.612432.2103213.43462311.56397.34955.7692316.73072321.033141651.7909105.51792333.5724799332.3068107.27882342.294276.3608.9799831.73492352.1692053.4808260.35032362.262949.6021114.89832372.9433385.3211369.011212381.361133.4536.07641582.21892391.69948.61429.5857466.80472402.179421.9735.1996102.43232412.49311052066.9875253.46972426.5195509307.7159129.03812431.149187.9479.1122164.57782442.91545.31578.3505312.268042450.7381311730.3523319.64762460.84368.38426.8090176.86832473.73410131434.9223423.67432481.239119.6683.4543206.53752496.5632585.7077451.77512506.188819.6509110.60112511.3322871734.2342268.69362529.282518.4227365.86942535.982709.628888.71612545.192164297.687896.184972551.164380.3840.1202566.49482560.6498309.71143.42871152.508462573.9115921259.8465324.55242581.413167.61100.4954464.75582590.9708899.91294.8084835.29042609.3271330.5457294.84292617.915824.27302807.83812621.389507.43488.1209526.709862630.65599.41133.6923181.69232640.785859.9307.898066.67512651.9811067686.01716725.39122662.344791.6723.9761154.99142671.569325.1789.0376406.88332680.874123.51159.03890769.90842690.891346191.7977138.76402700.564268.9473.581561790.78012716.042825.7199492.88312720.83377.833273.7094448.85952731.45317033483.13832831.38332740.76372.262560.9436961.33682750.592162.71182302762.6144085377.96482173.29762772.1981120.56411049.13552780.8041269.9004423.63182791.33637425.14972792.66462801.932727.2256309.1097CFI-0.49629.08286.316012.0435400495 EMBODIMENTS Embodiment 1: A compound of Formula (I), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl, heteroaryl, C3-C10cycloalkyl, or heterocycloalkyl;each R1is independently deuterium, halogen, —CN, oxo, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;or two R1on adjacent atoms are taken together to form a C3-C10cycloalkyl or heterocycloalkyl; each optionally substituted with one or more R1b;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1aon the same atom are taken together to form an oxo;each R1bis independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1bon the same atom are taken together to form an oxo;n is 0, 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl; R3is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;each of R4, R4b, and R4cis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —NRcRd, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R5is hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each R6is independently hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R7is hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, or C1-C6aminoalkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2—NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C1alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)NH2, —S(═O)2NHCH3, —S(═O)2N(CH3h), —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3), —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 2: A compound of Formula (Ia), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl, heteroaryl, C3-C10cycloalkyl, or heterocycloalkyl;each R1is independently deuterium, halogen, —CN, oxo, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;or two R1on adjacent atoms are taken together to form a C3-C10cycloalkyl or heterocycloalkyl; each optionally substituted with one or more R1b;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1aon the same atom are taken together to form an oxo;each R1bis independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1bon the same atom are taken together to form an oxo;n is 0, 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;R3is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;each of R4a, R4b, and R4cis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —NRcRd, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R5is hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each R6is independently hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R7is hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, or C1-C6aminoalkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C1-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CHs, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3), —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)NH2, —S(═O)?NHCH3, —S(═O)N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3), —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)?N(CH3)2, —NH2, —NHCH3, —N(CH3), —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 3: A compound of Formula (Ib), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl, heteroaryl, C3-C10cycloalkyl, or heterocycloalkyl;each R1is independently deuterium, halogen, —CN, oxo, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C1heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;or two R1on adjacent atoms are taken together to form a C3-C10cycloalkyl or heterocycloalkyl; each optionally substituted with one or more R1b;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1aon the same atom are taken together to form an oxo;each R1bis independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1bon the same atom are taken together to form an oxo;n is 0, 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl; R3is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl; each of R4a, R4b, and R4cis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —NRcRd, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R5is hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each R6is independently hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R7is hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, or C1-C6aminoalkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C1-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C1-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CHs, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)NH2, —S(═O)?NHCH3, —S(═O)N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)?N(CH3)2, —NH2, —NHCH3, —N(CH3), —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 4: The compound according to any one of embodiments 1 to 3, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C6-C10aryl or heteroaryl. Embodiment 5: The compound according to embodiment 4, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C6-C10aryl. Embodiment 6: The compound according to embodiment 5, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is phenyl. Embodiment 7: The compound according to embodiment 4, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is heteroaryl. Embodiment 8: The compound according to embodiment 7, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is furanyl, pyrrolyl, thiophenyl, oxazolyl, imidazolyl, thiazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. Embodiment 9: The compound according to embodiment 8, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. Embodiment 10: The compound according to embodiment 9, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, or pyrimidinyl. Embodiment 11: The compound according to embodiment 10, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl. Embodiment 12: The compound according to embodiment 11, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl, or 5-pyrazolyl. Embodiment 13: The compound according to embodiment 12, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl. Embodiment 14: The compound according to embodiment 12, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazolyl. Embodiment 15: The compound according to embodiment 12, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrazolyl. Embodiment 16: The compound according to embodiment 12, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazolyl. Embodiment 17: The compound according to embodiment 10, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridinyl. Embodiment 18: The compound according to embodiment 17, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-pyridinyl, or 6-pyridinyl. Embodiment 19: The compound according to embodiment 18, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl. Embodiment 20: The compound according to embodiment 18, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridinyl. Embodiment 21: The compound according to embodiment 18, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridinyl. Embodiment 22: The compound according to embodiment 18, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridinyl. Embodiment 23: The compound according to embodiment 18, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridinyl. Embodiment 24: The compound according to embodiment 10, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazinyl. Embodiment 25: The compound according to embodiment 24, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, or 6-pyrazinyl. Embodiment 26: The compound according to embodiment 25, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl. Embodiment 27: The compound according to embodiment 25, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazinyl. Embodiment 28: The compound according to embodiment 25, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazinyl. Embodiment 29: The compound according to embodiment 25, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrazinyl. Embodiment 30: The compound according to embodiment 10, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrimidinyl. Embodiment 31: The compound according to embodiment 30, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. Embodiment 32: The compound according to embodiment 31, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl. Embodiment 33: The compound according to embodiment 31, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrimidinyl. Embodiment 34: The compound according to embodiment 31, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrimidinyl. Embodiment 35: The compound according to embodiment 31, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrimidinyl. Embodiment 36: The compound according to embodiment 9, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridazinyl. Embodiment 37: The compound according to embodiment 36, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, or 6-pyridazinyl. Embodiment 38: The compound according to embodiment 37, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl. Embodiment 39: The compound according to embodiment 37, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridazinyl. Embodiment 40: The compound according to embodiment 37, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridazinyl. Embodiment 41: The compound according to embodiment 37, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridazinyl. Embodiment 42: The compound according to any one of embodiments 1 to 3, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C3-C10cycloalkyl or heterocycloalkyl. Embodiment 43: The compound according to embodiment 42, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C3-C10cycloalkyl. Embodiment 44: The compound according to embodiment 42, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is heterocycloalkyl. Embodiment 45: Embodiment 40: The compound according to any one of embodiments 1 to 44, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a. Embodiment 46: The compound according to embodiment 45, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 47: The compound according to embodiment 46, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 48: The compound according to embodiment 47, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, —OC1-C6haloalkyl, —CF3, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 49: The compound according to embodiment 48, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, bromo, iodo, —CN, —OH, —ORa, C1-C6alkyl, —OC1-C6haloalkyl, —CF3, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 50: The compound according to embodiment 49, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, bromo, —CN, —OH, —OC1-C6alkyl, —OC1-C6haloalkyl, C1-C6alkyl, —CF3, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the —OC1-C6alkyl, C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 51: The compound according to embodiment 50, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, —CN, —OH, —OCH3, —OCH2CH3, —C1-C6alkyl(OR1a), —CH3, —CH2CH3, iso-propyl, n-propyl, n-butyl, i-butyl, t-butyl, —OCHF2, —OC1-C6hydroxyalkyl, —CF3, cyclopropyl, azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, 1,4-oxazepanyl, or thiazinyl; wherein each of the azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, thiazinyl, and 1,4-oxazepanyl is optionally and independently substituted with one or more R1a. Embodiment 52: The compound according to embodiment 51, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OH, —OCH3, —OCH2CH3, —CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, or 1,4-oxazepanyl; wherein each of the azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, and 1,4-oxazepanyl is optionally and independently substituted with one or more R1a. Embodiment 53: The compound according to embodiment 52, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, azetidinyl, piperidinyl, piperazinyl, or morpholinyl; wherein each of the azetidinyl, piperidinyl, piperazinyl, and morpholinyl, is optionally and independently substituted with one or more R1a. Embodiment 54: The compound according to embodiment 53, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, azetidinyl, piperidinyl, piperazinyl, or morpholinyl; wherein each of the azetidinyl, piperidinyl, piperazinyl, and morpholinyl, is optionally and independently substituted with one or more R1a. Embodiment 55: The compound according to embodiment 54, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more R1. Embodiment 56: The compound according to embodiment 55, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, iso-propyl, —OCHF2, —CF3, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. Embodiment 57: The compound according to embodiment 56, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. Embodiment 58: The compound according to embodiment 57, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3—CH3, —OCHF2, cyclopropyl, or morpholinyl; wherein morpholinyl is optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. Embodiment 59: The compound according to embodiment 58, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3—CH3, —OCHF2, cyclopropyl, or morpholinyl; wherein morpholinyl is optionally substituted with one or more —CH3. Embodiment 60: The compound according to any one of embodiments 1 to 59, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 1, 2, or 3. Embodiment 61: The compound according to embodiment 60, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 1. Embodiment 62: The compound according to embodiment 60, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 2. Embodiment 63: The compound according to embodiment 60, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 3. Embodiment 64: The compound according to any one of embodiments 1 to 63, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R2is hydrogen. Embodiment 65: The compound according to any one of embodiments 1 to 64, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R3is hydrogen. Embodiment 66: The compound according to any one of embodiments 1 to 65, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care independently hydrogen or halogen. Embodiment 67: The compound according to embodiment 66, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4ais halogen and R4b, and R4care hydrogen. Embodiment 68: The compound according to embodiment 66, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4care hydrogen and R4bis halogen. Embodiment 69: The compound according to embodiment 66, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare hydrogen and R4cis halogen. Embodiment 70: The compound according to embodiment 66, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare halogen and R4cis hydrogen. Embodiment 71: The compound according to embodiment 66, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare halogen and R4cis hydrogen. Embodiment 72: The compound according to embodiment 66, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4band R4care halogen. Embodiment 73: The compound according to embodiment 66, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care hydrogen. Embodiment 74: The compound according to any one of embodiments 1 to 73, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R5is hydrogen. Embodiment 75: The compound according to any one of embodiments 1 to 74, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R6is hydrogen. Embodiment 76: The compound according to any one of embodiments 1 to 75, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is hydrogen or C1-C6alkyl. Embodiment 77: The compound according to embodiment 76, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is hydrogen. Embodiment 78: The compound according to embodiment 76, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is C1-C6alkyl. Embodiment 79: The compound according to any one of embodiments 1 to 78, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each of R8a, R8b, R8c, and R8dis independently hydrogen, halogen, or —ORa. Embodiment 80: The compound according to embodiment 79, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each of R8a, R8b, and R8dare hydrogen and R8cis hydrogen, halogen, or —OR1a. Embodiment 81: The compound according to embodiment 80, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis halogen or —ORa. Embodiment 82: The compound according to embodiment 81, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis halogen. Embodiment 83: The compound according to embodiment 82, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis fluoro, chloro, bromo, or iodo. Embodiment 84: The compound according to embodiment 81, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis —ORa. Embodiment 85: The compound according to embodiment 84, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Rais C1-C6alkyl. Embodiment 86: The compound according to embodiment 85, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Rais —CH3. Embodiment 87: A compound of Formula (11), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl or heteroaryl, C3-C10cycloalkyl, and heterocycloalkyl;each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;n is 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen or C1-C6alkyl;R3is hydrogen or C1-C6alkyl;R4a, R4b, and R4care each independently hydrogen, deuterium, or halogen;R7is hydrogen or C1-C6alkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 88: The compound according to embodiment 87, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C6-C10aryl. Embodiment 89: The compound according to embodiment 88, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is phenyl. Embodiment 90: The compound according to embodiment 87, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is heteroaryl. Embodiment 91: The compound according to embodiment 90, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. Embodiment 92: The compound according to embodiment 91, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl. Embodiment 93: The compound according to embodiment 92, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl, or 5-pyrazolyl. Embodiment 94: The compound according to embodiment 93, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl. Embodiment 95: The compound according to embodiment 93, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazolyl. Embodiment 96: The compound according to embodiment 93, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrazolyl. Embodiment 97: The compound according to embodiment 93, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazolyl. Embodiment 98: The compound according to embodiment 91, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridinyl. Embodiment 99: The compound according to embodiment 98, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-pyridinyl, or 6-pyridinyl. Embodiment 100: The compound according to embodiment 99, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl. Embodiment 101: The compound according to embodiment 99, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridinyl. Embodiment 102: The compound according to embodiment 99, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridinyl. Embodiment 103: The compound according to embodiment 99, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridinyl. Embodiment 104: The compound according to embodiment 99, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridinyl. Embodiment 105: The compound according to embodiment 91, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazinyl. Embodiment 106: The compound according to embodiment 105, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, or 6-pyrazinyl. Embodiment 107: The compound according to embodiment 106, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl. Embodiment 108: The compound according to embodiment 106, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazinyl. Embodiment 109: The compound according to embodiment 106, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazinyl. Embodiment 110: The compound according to embodiment 106, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrazinyl. Embodiment 111: The compound according to embodiment 91, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrimidinyl. Embodiment 112: The compound according to embodiment 111, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. Embodiment 113: The compound according to embodiment 112, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl. Embodiment 114: The compound according to embodiment 112, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrimidinyl. Embodiment 115: The compound according to embodiment 112, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrimidinyl. Embodiment 116: The compound according to embodiment 112, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrimidinyl. Embodiment 117: The compound according to embodiment 91, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridazinyl. Embodiment 118: The compound according to embodiment 117, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, or 6-pyridazinyl. Embodiment 119: The compound according to embodiment 118, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl. Embodiment 120: The compound according to embodiment 118, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridazinyl. Embodiment 121: The compound according to embodiment 118, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridazinyl. Embodiment 122: The compound according to embodiment 118, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridazinyl. Embodiment 123: The compound according to embodiment 87, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C3-C10cycloalkyl or heterocycloalkyl. Embodiment 124: The compound according to embodiment 123, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is C3-C10cycloalkyl. Embodiment 125: The compound according to embodiment 123, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is heterocycloalkyl. Embodiment 126: The compound according to any one of embodiments 87 to 125, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, OC1-C6haloalkyl, C1-C6hydroxyalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 127: The compound according to embodiment 126, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 128: The compound according to embodiment 127, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, bromo, iodo, —CN, —OH, —ORa, C1-C6alkyl, —CF3, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 129: The compound according to embodiment 128, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, bromo, —CN, —OH, —ORa, C1-C6alkyl, —CF3, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 130: The compound according to embodiment 129, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, —CN, —OH, —OC1-C6alkyl, C1-C6alkyl, —CF3, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the —OC1-C6alkyl, C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a. Embodiment 131: The compound according to embodiment 130, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently fluoro, chloro, —CN, —OH, —OCH3, —OCH2CH3, —C1-C6alkyl(OR1a), —CH3, —CH2CH3, iso-propyl, n-propyl, n-butyl, i-butyl, t-butyl, —OC1-C6hydroxyalkyl, —CF3, —OCHF2, cyclopropyl, azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, 1,4-oxazepanyl, or thiazinyl; wherein each of the azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, thiazinyl, and 1,4-oxazepanyl is optionally and independently substituted with one or more R1a. Embodiment 132: The compound according to embodiment 131, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OH, —OCH3, —OCH2CH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, or 1,4-oxazepanyl; wherein each of the azetidinyl, oxetanyl, piperidinyl, piperazinyl, morpholinyl, and 1,4-oxazepanyl is optionally and independently substituted with one or more R1a. Embodiment 133: The compound according to embodiment 132, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, azetidinyl, piperidinyl, piperazinyl, or morpholinyl; wherein each of the azetidinyl, piperidinyl, piperazinyl, and morpholinyl, is optionally and independently substituted with one or more R1a. Embodiment 134: The compound according to embodiment 133, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —CN, —OCH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein each of the azetidinyl, piperidinyl, piperazinyl, and morpholinyl, is optionally and independently substituted with one or more R1a. Embodiment 135: The compound according to embodiment 134, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more R1a. Embodiment 136: The compound according to embodiment 135, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3, —CH3, iso-propyl, —CF3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. Embodiment 137: The compound according to embodiment 136, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3, —CH3, —OCHF2, cyclopropyl, piperidinyl, piperazinyl, or morpholinyl; wherein piperidinyl, piperazinyl, and morpholinyl are optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. Embodiment 138: The compound according to embodiment 137, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3—CH3, —OCHF2, cyclopropyl, or morpholinyl; wherein morpholinyl is optionally substituted with one or more —CH3, —CH2CH3, —CH2CH2CH3, —OH, and —OCH3. Embodiment 139: The compound according to embodiment 138, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each R1is independently chloro, —OCH3, —CH3, cyclopropyl, or morpholinyl; wherein morpholinyl is optionally substituted with one or more —CH3. Embodiment 140: The compound according to any one of embodiments 87 to 139, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 1, 2, or 3. Embodiment 141: The compound according to embodiment 140, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 1. Embodiment 142: The compound according to embodiment 140, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 2. Embodiment 143: The compound according to embodiment 140, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein n is 3. Embodiment 144: The compound according to any one of embodiments 87 to 143, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R2is hydrogen. Embodiment 145: The compound according to any one of embodiments 87 to 144, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R3is hydrogen. Embodiment 146: The compound according to any one of embodiments 87 to 145, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care independently hydrogen or halogen. Embodiment 147: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4ais halogen and R4b, and R4care hydrogen. Embodiment 148: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4care hydrogen and R4bis halogen. Embodiment 149: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare hydrogen and R4cis halogen. Embodiment 150: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4bare halogen and R4cis hydrogen. Embodiment 151: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4aand R4care halogen and R4bis hydrogen. Embodiment 152: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4ais halogen and R4band R4care hydrogen. Embodiment 153: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care hydrogen. Embodiment 154: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care halogen. Embodiment 155: The compound according to embodiment 146, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R4a, R4b, and R4care fluoro. Embodiment 156: The compound according to any one of embodiments 87 to 155, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is hydrogen or C1-C6alkyl. Embodiment 157: The compound according to embodiment 156, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is hydrogen. Embodiment 158: The compound according to embodiment 156, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R7is C1-C6alkyl. Embodiment 159: The compound according to any one of embodiments 87 to 158, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, or —ORa. Embodiment 160: The compound according to embodiment 159, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein each of R8a, R8band R8dare hydrogen and R8cis hydrogen, halogen, or —ORa. Embodiment 161: The compound according to embodiment 160, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis halogen or —ORa. Embodiment 162: The compound according to embodiment 161, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis halogen. Embodiment 163: The compound according to embodiment 162, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis fluoro, chloro, bromo, or iodo. Embodiment 164: The compound according to embodiment 161, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein R8cis —ORa. Embodiment 165: The compound according to embodiment 164, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Rais C1-C6alkyl. Embodiment 166: The compound according to embodiment 165, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Rais —CH3. Embodiment 167: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is heteroaryl; each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2—C(alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, 3, 4, 5, 6, 7, or 8; R7is hydrogen or C1-C6alkyl; R8cis halogen or —ORa; andeach R3is independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C1-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)?NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 168: The compound according to embodiment 167, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. Embodiment 169: The compound according to embodiment 168, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl. Embodiment 170: The compound according to embodiment 169, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl, or 5-pyrazolyl. Embodiment 171: The compound according to embodiment 170, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 1-pyrazolyl. Embodiment 172: The compound according to embodiment 170, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazolyl. Embodiment 173: The compound according to embodiment 170, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrazolyl. Embodiment 174: The compound according to embodiment 170, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazolyl. Embodiment 175: The compound according to embodiment 168, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridinyl. Embodiment 176: The compound according to embodiment 175, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-pyridinyl, or 6-pyridinyl. Embodiment 177: The compound according to embodiment 176, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl. Embodiment 178: The compound according to embodiment 176, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridinyl. Embodiment 179: The compound according to embodiment 176, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridinyl. Embodiment 180: The compound according to embodiment 176, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridinyl. Embodiment 181: The compound according to embodiment 176, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridinyl. Embodiment 182: The compound according to embodiment 168, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazinyl. Embodiment 183: The compound according to embodiment 182, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, or 6-pyrazinyl. Embodiment 184: The compound according to embodiment 183, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrazinyl. Embodiment 185: The compound according to embodiment 183, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyrazinyl. Embodiment 186: The compound according to embodiment 183, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrazinyl. Embodiment 187: The compound according to embodiment 183, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrazinyl. Embodiment 188: The compound according to embodiment 168, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrimidinyl. Embodiment 189: The compound according to embodiment 188, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. Embodiment 190: The compound according to embodiment 189, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl. Embodiment 191: The compound according to embodiment 189, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrimidinyl. Embodiment 192: The compound according to embodiment 189, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrimidinyl. Embodiment 193: The compound according to embodiment 189, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrimidinyl. Embodiment 194: The compound according to embodiment 168, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridazinyl. Embodiment 195: The compound according to embodiment 194, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, or 6-pyridazinyl. Embodiment 196: The compound according to embodiment 195, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridazinyl. Embodiment 197: The compound according to embodiment 195, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridazinyl. Embodiment 198: The compound according to embodiment 195, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridazinyl. Embodiment 199: The compound according to embodiment 195, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridazinyl. Embodiment 200: A compound according to embodiment 167, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein:Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl;each R1is independently halogen, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C1-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a;each R1is independently —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6hydroxyalkyl, or C1-C6heteroalkyl;n is 1, 2, or 3;R7is hydrogen;R8cis —ORa; andeach Rais C1-C6alkyl. Embodiment 201: The compound of embodiment 200, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrimidinyl. Embodiment 202: The compound according to embodiment 201, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. Embodiment 203: The compound according to embodiment 202, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl. Embodiment 204: The compound according to embodiment 202, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrimidinyl. Embodiment 205: The compound according to embodiment 202, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrimidinyl. Embodiment 206: The compound according to embodiment 202, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrimidinyl. Embodiment 207: A compound according to embodiment 167, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein:Ring A is pyridinyl or pyrimidinyl;each R1is independently fluoro, chloro, —CN, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a;each R1ais independently —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6hydroxyalkyl, or C1-C6heteroalkyl;n is 1, 2, or 3;R7is hydrogen;R8cis —OCH3; andeach Rais C1-C6alkyl. Embodiment 208: The compound according to embodiment 207, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyridinyl. Embodiment 209: The compound according to embodiment 208, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-pyridinyl, or 6-pyridinyl. Embodiment 210: The compound according to embodiment 209, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyridinyl. Embodiment 211: The compound according to embodiment 209, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 3-pyridinyl. Embodiment 212: The compound according to embodiment 209, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyridinyl. Embodiment 213: The compound according to embodiment 20), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyridinyl. Embodiment 214: The compound according to embodiment 209, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyridinyl. Embodiment 215: The compound according to embodiment 207, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrimidinyl. Embodiment 216: The compound according to embodiment 215, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, or 6-pyrimidinyl. Embodiment 217: The compound according to embodiment 216, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 2-pyrimidinyl. Embodiment 218: The compound according to embodiment 216, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 4-pyrimidinyl. Embodiment 219: The compound according to embodiment 216, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 5-pyrimidinyl. Embodiment 220: The compound according to embodiment 216, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is 6-pyrimidinyl. Embodiment 221: A compound according to embodiment 167, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein:Ring A is pyridinyl or pyrimidinyl;each R1is independently chloro, —CN, —OH, —OCH3, —OCH2CH3, —CHI, —CH2CH3, —CH(CH3)2, —CF3, —OCHF2, cyclopropyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, 1,1-dioxidothiomorpholinyl, or oxetanyl; wherein morpholinyl, piperidinyl, piperazinyl, azetidinyl, 1,1-dioxidothiomorpholinyl, or oxetanyl are each optionally and independently substituted with one or more R1a; each R1ais independently —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6hydroxyalkyl, or C1-C6heteroalkyl;n is 1, 2, or 3;R7is hydrogen;R8cis —OCH3; andeach Rais C1-C6alkyl. Embodiment 222: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is heteroaryl; each R1is independently halogen, —CN, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C1-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, or 3: R7is hydrogen or C1-C6alkyl; R8cis halogen or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; and each Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl. C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CHI)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)?, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C4heteroalkyl. Embodiment 223: The compound according to embodiment 222, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. Embodiment 224: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is heteroaryl; each R1is independently halogen, —ORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C1-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, or 3; R7is hydrogen or C1-C6alkyl; R8cis halogen or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; and each Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 225: The compound according to embodiment 222, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. Embodiment 226: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is heteroaryl; each R1is independently halogen, —ORa, —SRa, —S(═O)R3, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C3-C10cycloalkyl, or heterocycloalkyl; wherein each of the C1-C6alkyl, C3-C10cycloalkyl, and heterocycloalkyl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, or 3; R7is hydrogen or C1-C6alkyl; R8cis halogen or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3h, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; and each Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 227: The compound according to embodiment 226, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein Ring A is pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl. Embodiment 228: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —S(═O)2(C1-C6alkyl), —S(═O)2N(C1-C6alkyl)2, —OC1-C6alkyl, —C(═O)N(C1-C6alkyl)2, —C(═O)N(H)(C1-C6alkyl), —OC1-C6haloalkyl, C1-C6alkyl, —S(C1-C6alkyl), heterocycloalkyl, or —C(═O)(heterocycloalkyl); n is 1, 2, or 3; R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. Embodiment 229: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(CH3), —S(═O)2(CH2CH)—S(═O)2N(CH3)2, —OCH3, —CH2CHF2, —C(═O)N(CH3)2, —C(═O)N(H)(CH3), —OC1C6haloalkyl, —CH3, —CH2CH3, iso-propyl, n-propyl, —SCH3, azetidinyl, pyrrolidinyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl; n is 1, 2, or 3: R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. Embodiment 230: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is aryl; each R1is independently halogen, —CN, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, —OC1-C6haloalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C1-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a; each R1ais independently deuterium, —OH, —ORa, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; n is 1, 2, or 3: R7is hydrogen or C1-C6alkyl; R8cis hydrogen, C1-C6alkyl, halogen or —ORa; each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; and each Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(G-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)?, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 231: A compound selected from (1R,2S)-5′-methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(5-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloropyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-ethoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-2-{3-[(5-cyclopropylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloropyrimidin-4-yl)amino]-1H-indazol-6-yl)}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1S,2R)-5′-methoxy-2-{3-[(5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2-chloro-5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(piperidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyrazin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(6-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyridin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxypyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxypyridin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(4-methylpiperazin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(1,3,5-trimethyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-(trifluoromethyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloro-2-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methoxypyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-1-methyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(3-hydroxy-2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-2-(3-{[(3S)-3-hydroxy-2,3-dihydro-1-benzofuran-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[(3R)-3-hydroxy-2,3-dihydro-1-benzofuran-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,3-dihydropyrazolo[5,1-b][1,3]oxazol-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-oxo-2,3-dihydro-1-benzofuran-7-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,3-dihydrofuro[2,3-c]pyridin-7-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[(3S)-3-(hydroxymethyl)-2,3-dihydrofuro[2,3-c]pyridin-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[(3R)-3-(hydroxymethyl)-2,3-dihydrofuro[2,3-c]pyridin-7-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((6-(3-oxa-8-azabicyclo[3.2.1]octan-8-yl)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-{[6-(2-hydroxyethoxy)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one); (1R,2S)-5′-methoxy-2-(3-{[6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-methyl-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(1,4-oxazepan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(azetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-methoxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[4-chloro-5-methoxy-6-(morpholin-4-yl)pyrimidin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[1-(2-hydroxyethyl)-3-methoxy-1H-pyrazol-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-cyclopropyl-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((5-chloro-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-{[5-(2-hydroxyethyl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(2-hydroxyethyl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-5-methoxy-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((5-chloro-6-(1,1-dioxidothiomorpholino)-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(34 (2-cyclopropyl-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-2-methylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-6-(morpholin-4-yl)pyrimidine-2-carbonitrile; 4-(1,1-dioxidothiomorpholino)-5-methoxy-6-((6-((1R,2S)-5′-methoxy-2′-oxospiro[cyclopropane-1,3′-indolin]-2-yl)-1H-indazol-3-yl)amino)pyrimidine-2-carbonitrile; (1R,2S)-2-{3-[(1,3-dimethyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[1-methyl-3-(trifluoromethyl)-1H-pyrazol-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(1-methyl-1H-pyrazol-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl)}amino)-1-methyl-1H-pyrazole-3-carbonitrile; (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxy-2-methylpyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(2-hydroxyethyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((2-cyclopropyl-6-(1,1-dioxidothiomorpholino)-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-2-(3-((5-chloro-2-cyclopropyl-6-(1,1-dioxidothiomorpholino)pyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyrazin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-5-methoxy-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(1,3-dimethyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-cyclopropyl-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-[3-({2-cyclopropyl-6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((5-chloro-6-(1,1-dioxidothiomorpholino)-2-isopropylpyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methyl-1H-pyrazol-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(6-cyclopropyl-3-methoxypyrazin-2-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-cyclopropyl-6-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(3,6-dimethylpyrazin-2-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-((6-(1,1-dioxidothiomorpholino)-2-isopropyl-5-methoxypyrimidin-4-yl)amino)-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indolin]-2′-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(morpholin-4-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(5-methoxy-2-methylpyridin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyridin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methoxy-5-methylpyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxypyridazin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(3-cyclopropyl-1-methyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(3-cyclopropyl-1-ethyl-1H-pyrazol-5-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(2-hydroxy-2-methylpropyl)-5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(morpholin-4-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (0R,2S)-5′-methoxy-2-(3-{[3-methoxy-2-(morpholin-4-yl)pyridin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloro-2-methylpyridin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-chloro-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(morpholin-4-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (0R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(oxetan-3-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-6-(propan-2-yl)pyridazin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[6-(morpholin-4-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(3-hydroxyazetidin-1-yl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methyl-6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-(3-{[6-(propan-2-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxy-1′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-1′-ethyl-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(azetidin-3-yl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3-hydroxyazetidin-1-yl)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one, (1R,2S)-2-(3-{[1-(2,2-difluoroethyl)-3-methyl-1H-pyrazol-5-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-6-(morpholin-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-1′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(3-hydroxyazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[6-(oxetan-3-yl)pyrazin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-(oxetan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxetan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-2-(3-hydroxyazetidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-1′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2R)-2-{7-fluoro-3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1S,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methylspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-4′-fluoro-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1S,2S)-4′-fluoro-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-6′-fluoro-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-chloro-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-fluoro-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-ethoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-fluorospiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-[3-({2-methyl-5-[(propan-2-yl)oxy]pyrimidin-4-yl}amino)-1H-indazol-6-yl]spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-(trifluoromethyl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-(trifluoromethoxy)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1S,2S)-2-{3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-(trifluoromethoxy)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-cyclopropyl-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; and (1R,2S)-2-(3-{[5-(difluoromethoxy)-2-(oxetan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2R)-5′-fluoro-2-{7-fluoro-3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; and (1R,2R)-2-(3-{[5-(difluoromethoxy)-2-methylpyrimidin-4-yl]amino}-7-fluoro-1H-indazol-6-yl)-5′-fluorospiro[cyclopropane-1,3′-indol]-2′(1′H)-one or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Embodiment 232: A compound selected from (1R,2R)-2-{5-fluoro-3-[(5-methoxy-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-6-methylpyridazin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(cyclopropylmethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(2,2-difluoroethoxy)-2-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-[3-(2-methoxy-5-methylanilino)-1H-indazol-6-yl]spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methyl-5-(2,2,2-trifluoroethoxy)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methoxy-6-methylpyridin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methyl-6-(propan-2-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(5-ethyl-2-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methyl-6,7-dihydrofuro[3,2-d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxan-4-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,5-dimethoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(azetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(trifluoromethyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2-ethyl-5-methoxypyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(7-methoxyquinolin-6-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methyl-54methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxyquinolin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2,5-dimethoxypyridin-3-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2-chlorofuro[3,2-d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(pyrrolidin-1-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(methanesulfonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-2-[3-({2-[(3R)-3-fluoropyrrolidin-1-yl]-5-methoxypyrimidin-4-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(3,3-difluoropyrrolidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(2-chloro-5-methyl-5H-pyrrolo[3,2-d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidine-2-carbonitrile; (1R,2S)-2-(3-{[2-(3,3-difluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(3-fluoroazetidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[5-(ethanesulfonyl)-2-methoxyanilino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-methoxy-3-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylbenzamide; 4-methoxy-3-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methylbenzamide; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-5-(propane-2-sulfonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-methoxy-3-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylbenzene-1-sulfonamide; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-5-(morpholine-4-carbonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-carboxamide; (1R,2S)-2-(3-{[2-(dimethylamino)-5-methylpyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(morpholine-4-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(3,3-difluoroazetidine-1-carbonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(4,4-difluoropiperidine-1-carbonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(4-fluoro-3-{[5-methoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(2-methoxy-5-methylpyrimidin-4-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(2-oxa-6-azaspiro[3.3]heptane-6-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-(4,4-difluoropiperidin-1-yl)-5-methoxypyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-[5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrimidin-2-yl]-1λ6-thiomorpholine-1,1-dione; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methylpyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(2-oxa-6-azaspiro[3.3]heptan-6-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(methanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methyl-N-(propan-2-yl)pyridine-2-carboxamide; (1R,2S)-2-(3-{[5-ethoxy-2-(methylsulfanyl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (R,2S)-2-(3-{[6-(methanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-carboxamide; 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-4-(morpholine-4-carbonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; Diastereomer 1: (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxolan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; Diastereomer 2: (1R,2S)-5′-methoxy-2-(3-{[5-methoxy-2-(oxolan-3-yl)pyrimidin-4-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[2-ethoxy-6-(methanesulfonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-ethoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-carboxamide; 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-3-sulfonamide; (1R,2S)-2-(3-{[6-(dimethylphosphoryl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 2-fluoro-5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylbenzamide; (1R,2S)-2-(3-[5-fluoro-2-methoxy-4-(morpholine-4-carbonyl)anilino]-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; and (1R,2S)-2-(3-{[3-ethoxy-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one, or a pharmaceutically acceptable salt thereof. Embodiment 233: A compound selected from (1R,2S)-2-(3-{[6-(ethanesulfonyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-methoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,2-dimethylbenzene-1-sulfonamide; (1R,2S)-2-{3-[(2,5-dimethyl-5,7-dihydrothieno[3,4-d]pyrimidin-4-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 2,5-dimethoxy-4-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)benzene-1-sulfonamide; (1R,2S)-2-(3-{[2-(dimethylamino)-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; 6-ethoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(2-oxopyrrolidin-1-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-[2-methoxy-5-(morpholine-4-sulfonyl)anilino]-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(3-methoxy-1,5-naphthyridin-2-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; N,6-dimethoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methylpyridine-2-carboxamide; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N′-(propan-2-yl)pyridine-2-carbohydrazide; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-5-(2-oxopyrrolidin-1-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-5-(3-methyl-2-oxoimidazolidin-1-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-sulfonamide; 6-ethoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-sulfonamide; (1R,2S)-5′-methoxy-2-{3-[2-methoxy-5-(oxane-4-sulfonyl)anilino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(dimethylphosphoryl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(2-hydroxypropan-2-yl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(methanesulfonyl)-2-methoxy-5-methylpyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(ethanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(dimethylphosphoryl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; N-(cyclopropylmethyl)-6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carboxamide; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-(propan-2-yl)pyridine-2-carboxamide; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(3-oxa-8-azabicyclo[3.2.1]octane-8-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-1-methyl-6-oxo-1,6-dihydropyridazin-3-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-5-(1,3-oxazol-2-yl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(3-methoxyazetidine-1-carbonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyrazine-2-carboxamide; 6-ethoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyrazine-2-carboxamide; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N,3-trimethylpyridine-2-carboxamide; (1R,2S)-2-(3-{[6-(methanesulfinyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(methanesulfinyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-2-(3-{[6-(methanesulfonyl)-4-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(methanesulfonyl)-3-methoxypyrazin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; N,N-dicyclopropyl-6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carboxamide; N-(2,2-difluoroethyl)-6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carboxamide; (1R,2S)-2-[3-({6-[(2R,6S)-2,6-dimethylpiperidine-1-carbonyl]-2-methoxypyridin-3-yl}amino)-1H-indazol-6-yl]-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-(3-{[2-methoxy-6-(8-oxa-3-azabicyclo[3.2.1]octane-3-carbonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[3-chloro-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(propane-2-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(dimethylphosphoryl)-4-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; N-(1,3-difluoropropan-2-yl)-6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carboxamide; 6-chloro-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; (1R,2S)-2-{3-[(5-chloro-2-methyl-1,3-benzoxazol-6-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 4-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N,N-dimethylpyridine-2-carboxamide; 3-[6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyrazin-2-yl]-1λ6-thietane-1,1-dione; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(morpholine-4-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(8-oxa-3-azabicyclo[3.2.1]octane-3-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(diethylphosphoryl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-2-(3-{[5-(cyclopropanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(oxane-4-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 6-methoxy-5-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)pyridine-2-carbonitrile; (1R,2S)-2-{3-[5-(diethylphosphoryl)-2-methoxyanilino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(ethanesulfonyl)-4-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(azetidine-1-carbonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(morpholine-4-carbonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(6-methoxy-2-methyl-1-oxo-2,3-dihydro-1H-isoindol-5-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-{3-[(4-methoxy-2-methyl-1-oxo-2,3-dihydro-1H-isoindol-5-yl)amino]-1H-indazol-6-yl}spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-5′-methoxy-2-(3-{[3-methoxy-5-(1,2-oxazolidine-2-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(azetidine-1-sulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-(3-methyloxetan-3-yl)pyridine-3-sulfonamide; 5-methoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methyl-N-(3-methyloxetan-3-yl)pyridine-3-sulfonamide; (1R,2S)-2-(3-{[5-(1-hydroxyethyl)-2-methoxypyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[2-ethoxy-4-(methanesulfonyl)anilino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[3-ethoxy-5-(4-methylpiperazine-1-sulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[5-(ethanesulfonyl)-3-ethoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; 5-ethoxy-6-({6-[(1R,2S)-5′-methoxy-2′-oxo-1′,2′-dihydrospiro[cyclopropane-1,3′-indol]-2-yl]-1H-indazol-3-yl}amino)-N-methylpyridine-3-sulfonamide; (1R,2S)-5′-chloro-2-(3-{[3-ethoxy-5-(methanesulfonyl)pyridin-2-yl]amino}-1H-indazol-6-yl)spiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-{3-[(4-ethoxy-1-methyl-6-oxo-1,6-dihydropyridazin-3-yl)amino]-1H-indazol-6-yl}-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[6-(2-hydroxypropan-2-yl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; (1R,2S)-2-(3-{[4-ethoxy-6-(methanesulfonyl)pyridin-3-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one; and (1R,2S)-2-(3-{[5-(difluoromethanesulfonyl)-3-methoxypyridin-2-yl]amino}-1H-indazol-6-yl)-5′-methoxyspiro[cyclopropane-1,3′-indol]-2′(1′H)-one, or a pharmaceutically acceptable salt thereof. Embodiment 234: A compound of Formula (I), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein:Ring A is C6-C10aryl, heteroaryl, C3-C10cycloalkyl, or heterocycloalkyl;each R1is independently deuterium, halogen, —CN, oxo, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, —P(O)(R3)2, —P(O)2(Ra)2, C1-C6alkyl, C1-C6haloalkyl, —OC1-C10haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl; wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is optionally and independently substituted with one or more R1a;or two R1on adjacent atoms are taken together to form a C3-C10cycloalkyl or heterocycloalkyl; each optionally substituted with one or more R1b;each R1ais independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C6-C10deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1aon the same atom are taken together to form an oxo;each R1bis independently deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)R3, —S(═O)2R*, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;or two R1bon the same atom are taken together to form an oxo;n is 0, 1, 2, 3, 4, 5, 6, 7, or 8;R2is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;R3is hydrogen, C1-C6alkyl, C1-C6haloalkyl, or C1-C6deuteroalkyl;each of R4a, R4b, and R4cis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —NRcRd, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R5is hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each R6is independently hydrogen, deuterium, halogen, —CN, —OH, —ORa, —NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;R7is hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, or C1-C6aminoalkyl;each of R8a, R8b, R8c, and R8dis independently hydrogen, deuterium, halogen, —CN, —NO2, —OH, —ORa, —OC(═O)Ra, —OC(═O)ORb, —OC(═O)NRcRd, —SH, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRcRd, —NRcRd, —NRbC(═O)NRcRd, —NRbC(═O)Ra, —NRbC(═O)ORa, —NRbS(═O)2Ra, —C(═O)Ra, —C(═O)ORb, —C(═O)NRcRd, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, or heteroaryl;each Rais independently C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;each Rbis independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, C1-C6heteroalkyl, C6-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C6cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl; andeach Rcand Rdare independently hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6alkoxy, C1-C6aminoalkyl, C1-C6alkylamino, C1-C6heteroalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, heteroaryl, C1-C6alkyl(C3-C10cycloalkyl), C1-C6alkyl(heterocycloalkyl), C1-C6alkyl(C6-C10aryl), or C1-C6alkyl(heteroaryl); wherein each of the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C10cycloalkyl, heterocycloalkyl, C6-C10aryl, and heteroaryl is independently optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3)2, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl;or Rcand Rdare taken together with the atom to which they are attached to form a heterocycloalkyl optionally substituted with one or more oxo, deuterium, halogen, —CN, —OH, —OCH3, —S(═O)CH3, —S(═O)2CH3, —S(═O)2NH2, —S(═O)2NHCH3, —S(═O)2N(CH3)2, —NH2, —NHCH3, —N(CH3), —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, C1-C6alkyl, C1-C6haloalkyl, C1-C6deuteroalkyl, C1-C6hydroxyalkyl, C1-C6aminoalkyl, or C1-C6heteroalkyl. Embodiment 235: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(CH3), —S(═O)2(CH2CH3), —S(═O)2(i-Pr), —S(═O)2(cyclopropyl), —S(═O)2(C1C6haloalkyl), —S(═O)2N(CH3)2, —S(═O)2NH2, —S(═O)2N(CH3)(H), —OCH3, —OCH3, —OCH2CH3, —CH2CHF2, —C(═O)N(CH3), —C(═O)N(H)(CH3), —OC1C6haloalkyl, —CH3, —CH2CH3, iso-propyl, n-propyl, —SCH3, azetidinyl, pyrrolidinyl, oxazolyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl; n is 1, 2, or 3; R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. Embodiment 236: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —CF3, —CN, —S(═O) (C1-C6alkyl), —S(═O)2(C3-C10cycloalkyl), —S(═O)2N(C1-C6alkyl)2, —S(═O)2NH2, —S(═O)2N(C1-C6alkyl)(H), —OC1-C6alkyl, —CH2CHF2, —C(═O)N(C1-C6alkyl)2, —C(═O)N(H)(C1-C6alkyl), —OC1C6haloalkyl, —C1-C6alkyl, —SC1-C6alkyl, azetidinyl, pyrrolidinyl, oxazolyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl; n is 1, 2, or 3; R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. Embodiment 237: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof: wherein: Ring A is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(C1-C6alkyl), —S(═O)2(C3-C10cycloalkyl), —S(═O)2N(C1-C6alkyl)2, —S(═O)2NH2, —S(═O)2N(C1-C6alkyl)(H), —OC1-C6alkyl, —CH2CHF2, —C(═O)N(C1-C6alkyl)2, —C(═O)N(H)(C1-C6alkyl), —OC1C6haloalkyl, —C1-C6alkyl, —SC1-C6alkyl, azetidinyl, pyrrolidinyl, oxazolyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl, and wherein at least one of R1is —S(═O)2(C1-C6alkyl), —S(═O)2(C3-C10cycloalkyl), —S(═O)2N(C1-C6alkyl)2, —S(═O)2NH2, or —S(═O)2N(C1-C6alkyl)(H); n is 1, 2, or 3; R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. Embodiment 238: A compound of Formula (III), or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein: Ring A is heterocycloalkyl; each R1is independently halogen, —CF3, —CN, —S(═O)2(C1-C6alkyl), —S(═O)2(C2-C10cycloalkyl), —S(═O)2N(C1-C6alkyl)2, —S(═O)2NH2, —S(═O)2N(C1-C6alkyl)(H), —OC1-C6alkyl, —CH2CHF2, —C(═O)N(C1-C6alkyl)2, —C(═O)N(H)(C1-C6alkyl), —OC1C6haloalkyl, —C1-C6alkyl, —SC1-C6alkyl, azetidinyl, pyrrolidinyl, oxazolyl, fluoropyrrolidinyl, difluoropiperidinyl, difluoroazetidinyl, fluoroazetidinyl, morpholinyl, dioxidothiomorpholinyl, —C(═O)(morpholinyl), —C(═O)(azetidinyl), —C(═O)(difluoroazetidinyl), or —C(═O)difluoropiperidinyl; n is 1, 2, or 3; R7is hydrogen; and R8cis hydrogen, halogen, CH3, or —OCH3. Embodiment 239: A pharmaceutical composition comprising an amount of a compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, and one or more pharmaceutically acceptable excipients. Embodiment 240: A method of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Embodiment 241: A method of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition according to embodiment 239. Embodiment 242: The method according to embodiment 240 or 241, wherein the cancer in the subject is a solid tumor. Embodiment 243: The method according to embodiment 240 to 242, wherein the cancer is neuroblastoma, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (CNS), primary CNS lymphoma, spinal axis tumors, brain stem glioma, or pituitary adenoma. Embodiment 244: The method according to any one of embodiments 240 to 243, wherein the cancer in the subject expresses polo-like kinase 4 (PLK4). Embodiment 245: The method according to any one of embodiments 240 to 244, wherein the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering the compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or the pharmaceutical composition according to embodiment 233, to the subject. Embodiment 246: The method according to any one of embodiments 240 to 245, wherein the cancer in the subject exhibits an overexpression of the E3 ubiquitin-protein ligase (TRIM37) protein. Embodiment 247: The method according to embodiment 246, wherein the cancer in the subject exhibits an overexpression of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Embodiment 248: The method according to embodiment 246, wherein the cancer in the subject exhibits an amplification of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Embodiment 249: A method of treating cancer in a subject comprising administering to the subject a therapeutically effective amount of a compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein the cancer in the subject has been determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37) prior to administration of the compound to the subject. Embodiment 250: A method of treating cancer in a subject, wherein the cancer in the subject has been determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37), comprising administering to the subject a therapeutically effective amount of a compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Embodiment 251: A method of treating cancer in a subject, comprising:a. obtaining a biological sample of the cancer from the subject;b. determining whether the biological sample of the cancer overexpresses the gene that encodes the tripartite motif-containing protein 37 (TRIM37); andc. administering to the subject a therapeutically effective amount of a compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, if the biological sample of the cancer is determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Embodiment 252: The method according to any one of embodiments 240 to 251, wherein the cancer is neuroblastoma or breast cancer. Embodiment 253: The method of embodiment 252, wherein the cancer is neuroblastoma. Embodiment 254: The method of embodiment 252, wherein the cancer is breast cancer. Embodiment 255: The method according to any one of embodiments 240 to 254, wherein the compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, or the pharmaceutical composition according to embodiment 239 is administered to the subject with one or more additional therapeutic agents. Embodiment 256: The method according to embodiment 255, wherein the one or more additional therapeutic agents is selected from one or more mitotic inhibitors, alkylating agents, antimetabolites, antitumor antibiotics, anti-angiogenesis agents, topoisomerase I and II inhibitors, plant alkaloids, hormonal agents and antagonists, growth factor inhibitors, radiation, signal transduction inhibitors, such as inhibitors of protein tyrosine kinases and/or serine/threonine kinases, cell cycle inhibitors, biological response modifiers, enzyme inhibitors, antisense oligonucleotides or oligonucleotide derivatives, cytotoxics, and immuno-oncology agents. Embodiment 257: A method of inhibiting polo-like kinase 4 (PLK4) in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Embodiment 258: A method of inhibiting polo-like kinase 4 (PLK4) in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition according to embodiment 239. Embodiment 259: A method of inhibiting polo-like kinase 4 (PLK4) in a subject having cancer, comprising administering to the subject a therapeutically effective amount of a compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, wherein the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering the compound, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, to the subject. Embodiment 260: A method of inhibiting polo-like kinase 4 (PLK4) in a subject having cancer, comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition according to embodiment 239, wherein the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering the pharmaceutical composition to the subject. Embodiment 261: A compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, for use in a method of treating cancer in a subject in need thereof. Embodiment 262: A pharmaceutical composition according to embodiment 239 for use in a method of treating cancer in a subject in need thereof. Embodiment 263: A compound for use according to embodiment 261, wherein the cancer in the subject is a solid tumor. Embodiment 264: A compound for use according to embodiment 261 or 262, wherein the cancer is neuroblastoma, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (CNS), primary CNS lymphoma, spinal axis tumors, brain stem glioma, or pituitary adenoma. Embodiment 265: A compound for use according to any one of embodiments 261, 263, or 264 wherein the cancer in the subject expresses polo-like kinase 4 (PLK4). Embodiment 266: A compound for use according to any one of embodiments 261, 263, or 264, wherein the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering the compound to the subject. Embodiment 267: A compound for use according to any one of embodiments 261, 263, or 264, wherein the cancer in the subject exhibits an overexpression of the E3 ubiquitin-protein ligase (TRIM37) protein. Embodiment 268: A compound for use according to any one of embodiments 261, 263, or 264, wherein the cancer in the subject exhibits an overexpression of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Embodiment 269: A compound for use according to any one of embodiments 261, 263, or 264, wherein the cancer in the subject exhibits an amplification of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Embodiment 270: A compound for use according to any one of embodiments 261, 263, or 264, wherein the cancer in the subject has been determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37) prior to administration of the compound to the subject. Embodiment 271: A compound for use according to any one of embodiments 261, 263, or 264, wherein the cancer is neuroblastoma or breast cancer. Embodiment 272: A compound for use according to 271, wherein the cancer is neuroblastoma. Embodiment 273: A compound for use according to 271, wherein the cancer is breast cancer. Embodiment 274: A compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, for use in inhibiting polo-like kinase 4 (PLK4) in a subject having cancer. Embodiment 275: Use of a compound according to any one of embodiments 1 to 238, or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof, in the manufacture of a medicament for the treatment of cancer in a subject in need thereof. Embodiment 276: Use according to embodiment 275, wherein the cancer is neuroblastoma, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (CNS), primary CNS lymphoma, spinal axis tumors, brain stem glioma, or pituitary adenoma. Embodiment 277: Use according to embodiment 275 or 276, wherein the cancer in the subject expresses polo-like kinase 4 (PLK4). Embodiment 278: Use according to embodiment 275 or 276, wherein the cancer in the subject has been determined to express polo-like kinase 4 (PLK4) prior to administering the compound to the subject. Embodiment 279: Use according to any one of embodiments 274 to 278, wherein the cancer in the subject exhibits an overexpression of the E3 ubiquitin-protein ligase (TRIM37) protein. Embodiment 280: Use according to any one of embodiments 274 to 279, wherein the cancer in the subject exhibits an overexpression of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Embodiment 281: Use according to any one of embodiments 274 to 279, wherein the cancer in the subject exhibits an amplification of the gene that encodes the tripartite motif-containing protein 37 (TRIM37). Embodiment 282: Use according to any one of embodiments 274 to 279, wherein the cancer in the subject has been determined to overexpress the gene that encodes the tripartite motif-containing protein 37 (TRIM37) prior to administration of the compound to the subject. Embodiment 283: Use according to any one of embodiments 274 to 282, wherein the cancer is neuroblastoma or breast cancer. Embodiment 284: Use according to embodiment 283, wherein the cancer is neuroblastoma. Embodiment 285: Use according to embodiment 283, wherein the cancer is breast cancer.
1,015,559
11858916
DETAILED DESCRIPTION OF THE INVENTION The present invention concerns compounds of Formula I: or a pharmaceutically acceptable salt thereof, wherein A is A1 or A2, and whereineach R1is independently halogen, —CN, —C1-3alkyl, or —OC1-3alkyl, wherein the alkyl of C1-3alkyl and OC1-3alkyl is substituted with 0 to 3 F atoms;m is 0, 1, 2, or 3;X-L is N—CH2, CHCH2, or cyclopropyl;Y is CH or N;ZA1is CH, CR2, or N;ZA2is CH, CR2, or N;ZA3is CH, CR2, or N, provided that ZA2and ZA3are not simultaneously N; and further provided that one of ZA2and ZA3is N when X-L is N—CH2;each R2is independently F, Cl, or —CN;each R3is independently F, —OH, —CN, —C1-3alkyl, —OC1-3alkyl, or —C3-4cycloalkyl, or 2 R3s may together cyclize to form —C3-4spirocycloalkyl, wherein the alkyl of C1-3alkyl and OC1-3alkyl, cycloalkyl, or spirocycloalkyl may be substituted as valency allows with 0 to 3 F atoms and with 0 to 1 —OH;q is 0, 1, or 2;R4is —C1-3alkyl, —C0-3alkylene-C3-6cycloalkyl, —C0-3alkylene-R5, or —C1-3alkylene-R6, wherein said alkyl may be substituted as valency allows with 0 to 3 substituents independently selected from 0 to 3 F atoms and 0 to 1 substituent selected from —C0-1alkylene-CN, —C0-1alkylene-ORO, and —N(RN)2, andwherein said alkylene and cycloalkyl may be independently substituted as valency allows with 0 to 2 substituents independently selected from 0 to 2 F atoms and 0 to 1 substituent selected from —C0-1alkylene-CN, —C0-1alkylene-ORO, and —N(RN)2;R5is a 4- to 6-membered heterocycloalkyl, wherein said heterocycloalkyl may be substituted with 0 to 2 substituents as valency allows independently selected from:0 to 1 oxo (═O),0 to 1 —CN,0 2 F atoms, and0 to 2 substituents independently selected from —C1-3alkyl and —OC1-3alkyl, wherein the alkyl of C1-3alkyl and OC1-3alkyl may be substituted with 0 to 3 substituents as valency allows independently selected from:0 to 3 F atoms,0 to 1 —CN, and0 to 1 —ORO;R6is a 5- to 6-membered heteroaryl, wherein said heteroaryl may be substituted with 0 to 2 substituents as valency allows independently selected from:0 to 2 halogens,0 to 1 substituent selected from —OROand —N(RN)2, and0 to 2 —C1-3alkyl, wherein the alkyl may be substituted with 0 to 3 substituents as valency allows independently selected from:0 to 3 F atoms, and0 to 1 —ORO;each ROis independently H, or —C1-3alkyl, wherein C1-3alkyl may be substituted with 0 to 3 F atoms;each RNis independently H, or —C1-3alkyl;Z1, Z2, and Z3are each —CRZ, orone of Z1, Z2, and Z3is N and the other two are —CRZ; andeach RZis independently H, F, Cl, or —CH3. Another embodiment concerns a compound of Formula II or a pharmaceutically acceptable salt thereof, wherein A is A1 or A2, and whereineach R1is independently F, Cl, or —CN; andm is 0, 1, or 2. Another embodiment concerns a compound of Formula III or a pharmaceutically acceptable salt thereof, wherein A is A1 or A2, and whereineach R1is independently F, Cl, or —CN; andm is 0, 1, or 2. Another embodiment concerns a compound of Formulas I, II, or III, wherein A is A1, or a pharmaceutically acceptable salt thereof. Another embodiment concerns a compound of Formula IV or a pharmaceutically acceptable salt thereof, whereineach R1is independently F, Cl, or —CN;m is 0, or 1;q is 0 or 1; andR3is —CH3. Another embodiment concerns a compound of Formulas I, II, III, or IV, wherein ZA1is CH, or CR2; andR2is F; or a pharmaceutically acceptable salt thereof. Another embodiment concerns a compound of Formulas I, II, III, or IV, wherein ZA1is N, or a pharmaceutically acceptable salt thereof. Another embodiment concerns a compound of Formulas I, II, or OII, wherein A is A2,q is 0 or 1; andR3is —CH3;or a pharmaceutically acceptable salt thereof. Another embodiment concerns a compound of Formulas I, II, or III, wherein ZA2is CH, or CR2; and ZA3is N, or a pharmaceutically acceptable salt thereof. Another embodiment concerns a compound of Formulas I, II, or III, wherein ZA2is N and ZA3is CH, or CR2, or a pharmaceutically acceptable salt thereof. Another embodiment concerns a compound of Formula I, or a pharmaceutically acceptable salt thereof, wherein A is A1 or A2, and whereineach R1is independently F, Cl, or —CN; andm is 0, or 1. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, or III, or a pharmaceutically acceptable salt thereof, wherein X-L is N—CH2; and Y is CH or N. From the embodiments described herein, in such a case, X is N and L is CH2. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, or III, or a pharmaceutically acceptable salt thereof, wherein X-L is CHCH2; and Y is N. From the embodiments described herein, in such a case, X is CH and L is CH2. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, or III, or a pharmaceutically acceptable salt thereof, wherein X-L is CHCH2; and Y is CH. From the embodiments described herein, in such a case, X is CH and L is CH2. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, or II, or a pharmaceutically acceptable salt thereof, wherein X-L is cyclopropyl; and Y is N. In the embodiments where X-L is cyclopropyl, X-L being cyclopropyl would provide: Another embodiment concerns a compound of Formula V or a pharmaceutically acceptable salt thereof, whereineach R1is independently F, Cl, or —CN;m is 0, or 1;ZA2is CH, CR2, or N;ZA3is CH, CR2, or N, provided that ZA2and ZA3are not simultaneously N; andeach R2is F. Another embodiment concerns compounds a compound of Formula VI or a pharmaceutically acceptable salt thereof, whereineach R1is independently F, Cl, or —CN;m is 0, or 1;R3is —CH3;q is 0, 1, or 2;ZA2is CH, CR2, or N;ZA3is CH, CR2, or N, ZA2and ZA3are not simultaneously N;R2is F; andY is CH or N. Another embodiment concerns compounds of any of Formulas I, II, III, IV, V, or IV and any embodiments thereof, wherein R4is —CH2—R5, wherein R5is the 4- to 5-membered heterocycloalkyl, wherein said heterocycloalkyl may be substituted with 0 to 2 substituents as valency allows independently selected from:0 to 2 F atoms, and0 to 1 substituent selected from oxo (═O), —OCH3and —CH2OCH3; or a pharmaceutically acceptable salt thereof. In the embodiments concerning a compound of Formulas II, III, IV, V, or VI, where a variable is not defined, it takes on the definition provided in Formula I. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein R4is —CH2CH2OCH3, C1-3alkylene-R5, or C1-3alkylene-R6, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein R4is —C1-3alkyl, wherein said alkyl may be substituted as valency allows with 0 to 1 substituent selected from —C0-1alkylene-ORO, and —N(RN)2, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein R4is —(CH2)2OCH3, or —(CH2)2N(CH3)2, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein R4is —CH2—R5, wherein R5is the 4- to 5-membered heterocycloalkyl, wherein said heterocycloalkyl may be substituted with 0 to 1 substituent that is oxo (═O), or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein R4is —CH2—R5, wherein R5is the 4- to 5-membered heterocycloalkyl, wherein said heterocycloalkyl may be substituted with 0 to 2 substituents as valency allows independently selected from:0 to 2 F atoms, and0 to 1 substituent selected from —OCH3and —CH2OCH3;or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of any of Formulas I, II, III, IV, V, or IV and any embodiments thereof, wherein R4is —CH2—R6, wherein R6is the 5-membered heteroaryl, wherein said heteroaryl may be substituted with 0 to 2 substitutents as valency allows independently selected from:0 to 2 halogens, wherein the halogen is independently selected from F and Cl,0 to 1 —OCH3, and0 to 1 —CH3, —CH2CH3, —CF3, or —CH2CH2OCH3;or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of any of Formulas I, II, III, IV, V, or IV and any embodiments thereof, wherein R4is —C1-3alkyl, wherein said alkyl may be substituted as valency allows with 0 to 3 substituents independently selected from 0 to 3 F atoms and 0 to 1 substituent that is —C0-1alkylene-ORO. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heterocycloalkyl is wherein the heterocycloalkyl may be substituted with 0 to 2 substituents as valency allows, e.g., replacing hydrogen, independently selected from:0 to 1 oxo (O═),0 to 1 —CN,0 to 2 F atoms, and0 to 2 substituents independently selected from —C1-3alkyl and —OC1-3alkyl, wherein the alkyl of C1-3alkyl and OC1-3alkyl may be independently substituted with 0 to 3 substituents as valency allows independently selected from:0 to 3 F atoms,0 to 1 —CN, and0 to 1 —ORO,or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heterocycloalkyl is wherein the heterocycloalkyl may be substituted with 0 to 2 substituents as valency allows, e.g., replacing hydrogen, independently selected from:0 to 1 —CN,0 2 F atoms, and0 to 2 substituents independently selected from —C1-3alkyl and —OC1-3alkyl, wherein the alkyl of C1-3alkyl and OC1-3alkyl may be independently substituted with 0 to 3 substituents as valency allows independently selected from:0 to 3 F atoms,0 to 1 —CN, and0 to 1 —ORO, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heterocycloalkyl is wherein the heterocycloalkyl may be substituted with 0 to 1 substituent as valency allows, e.g., replacing hydrogen, selected from:—CN,F atom, and0 to 1 substituent independently selected from —C1-3alkyl and —OC1-3alkyl, wherein the alkyl of C1-3alkyl and OC1-3alkyl may be substituted with 0 to 3 substituents as valency allows independently selected from:0 to 3 F atoms,0 to 1 —CN, and0 to 1 —ORO,or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heterocycloalkyl is or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heterocycloalkyl is wherein the heterocycloalkyl may be substituted as valency allows with 0 to 1 methyl, wherein said methyl may be substituted with 0 to 3 F atoms, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heterocycloalkyl is wherein the heterocycloalkyl is unsubstituted, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein —CH2—R5and the nitrogen to which R4is attached provides: or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heteroaryl is wherein said heteroaryl may be substituted with 0 to 2 substituents as valency allows, e.g., replacing hydrogen, independently selected from:0 to 2 halogens, wherein the halogen is independently selected from F and Cl,0 to 1 substituent selected from —OROand —N(RN)2, or0 to 2 —C1-3alkyl, wherein the alkyl may be substituted with 0 to 3 substituents as valency allows independently selected from:0 to 3 F atoms, and0 to 1 —ORO;or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heteroaryl is wherein said heteroaryl may be substituted with 0 to 1 substituent as valency allows with —C1-2alkyl, wherein the alkyl may be substituted with 0 to 3 substituents as valency allows independently selected from:0 to 3 F atoms, and0 to 1 —ORO; andeach ROis independently H, or —C1-3alkyl;or a pharmaceutically acceptable salt thereof. One will recognize that any substituent would replace H on the carbon or nitrogen being substituted. A non-limiting example of substituted heteroaryls are: One will recognize that H is replaced with a substituent, e.g., R6s(substituent allowed on any heteroaryl of R6), to provide: wherein R6sis —C1-2alkyl, wherein the alkyl may be substituted with 0 to 3 substituents as valency allows independently selected from:0 to 3 F atoms, and0 to 1 —ORO; andeach ROis independently H, or —C1-3alkyl;or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of Formulas I, II, III, IV, V, or VI, wherein the heteroaryl is or a pharmaceutically acceptable salt thereof. Another embodiment concerns any one or more compound that is2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid;2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid;2-[(4-{2-[(4-cyano-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid;2-[(4-{2-[(4-cyano-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or2-[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or a pharmaceutically acceptable salt thereof. Another embodiment concerns any one or more compound that is2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid;2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid;2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid;2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or a pharmaceutically acceptable salt thereof. Another embodiment concerns any one or more compound that is2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid;2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, wherein chirality comes from C79;2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, wherein chirality comes from P7;2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid, wherein chirality comes from P7;2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, wherein chirality comes from C93;or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-[(4-{2-[(4-cyano-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-[(4-{2-[(4-cyano-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid;2-[(1S)-6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl]-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid; or2-[(1R)-6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl]-1-(1,3-oxazol-2-ylmethyl)-1H-benzimidazole-6-carboxylic acid; or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid;2-[(1S)6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or2-[(1R)6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid;2-[(1S)6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid; or2-[(1R)6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid;or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds, wherein the compound is 2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein Z1, Z2, and Z3are each CRZ, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein RZis H, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein Z1, Z2, and Z3are each CH, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein each R2is F, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein q is 0, or a pharmaceutically acceptable salt thereof. One will recognize that when q is 0, R3is absent. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein R3is —CH3, or —CF3; and q is 1, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein R3is —CH3; and q is 1, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein each R1is independently F, Cl, or —CN, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein R4is —CH2—R5, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein R4is —CH2—R6, or a pharmaceutically acceptable salt thereof. Another embodiment concerns compounds of other embodiments herein, e.g., compounds of Formulas I, II, III, IV, V, or VI, wherein the compound is the free acid. In another embodiment, the invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, in admixture with at least one pharmaceutically acceptable excipient. This would include a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, or VI or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, in admixture with at least one pharmaceutically acceptable excipient and one or more other therapeutic agent discussed herein. The invention also includes the following embodiments:a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, for use as a medicament;a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, for use in the prevention and/or treatment of cardiometabolic and associated diseases discussed herein, including T2DM, pre-diabetes, NASH, and cardiovascular disease;a method of treating a disease for which an agonist of GLP-1R is indicated, in a subject in need of such prevention and/or treatment, comprising administering to the subject a therapeutically effective amount of a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein;the use of a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, for the manufacture of a medicament for treating a disease or condition for which an agonist of the GLP-1R is indicated;a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, for use in the treatment of a disease or condition for which an agonist of GLP-1R is indicated; ora pharmaceutical composition for the treatment of a disease or condition for which an agonist of the GLP-1R is indicated, comprising a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein. Every Example or pharmaceutically acceptable salt thereof may be claimed individually or grouped together in any combination. The invention also relates to a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, for use in the treatment and/or prevention of cardiometabolic and associated diseases discussed herein, including T2DM, pre-diabetes, NASH, and cardiovascular disease. Another embodiment of the invention concerns a compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, for use in the treatment and/or prevention of cardiometabolic and associated diseases including diabetes (T1D and/or T2DM, including pre-diabetes), idiopathic T1D (Type 1b), latent autoimmune diabetes in adults (LADA), early-onset T2DM (EOD), youth-onset atypical diabetes (YOAD), maturity onset diabetes of the young (MODY), malnutrition-related diabetes, gestational diabetes, hyperglycemia, insulin resistance, hepatic insulin resistance, impaired glucose tolerance, diabetic neuropathy, diabetic nephropathy, kidney disease (e.g., acute kidney disorder, tubular dysfunction, proinflammatory changes to the proximal tubules), diabetic retinopathy, adipocyte dysfunction, visceral adipose deposition, sleep apnea, obesity (including hypothalamic obesity and monogenic obesity) and related comorbidities (e.g., osteoarthritis and urine incontinence), eating disorders (including binge eating syndrome, bulimia nervosa, and syndromic obesity such as Prader-Willi and Bardet-Biedl syndromes), weight gain from use of other agents (e.g., from use of steroids and antipsychotics), excessive sugar craving, dyslipidemia (including hyperlipidemia, hypertriglyceridemia, increased total cholesterol, high LDL cholesterol, and low HDL cholesterol), hyperinsulinemia, NAFLD (including related diseases such as steatosis, NASH, fibrosis, cirrhosis, and hepatocellular carcinoma), cardiovascular disease, atherosclerosis (including coronary artery disease), peripheral vascular disease, hypertension, endothelial dysfunction, impaired vascular compliance, congestive heart failure, myocardial infarction (e.g. necrosis and apoptosis), stroke, hemorrhagic stroke, ischemic stroke, traumatic brain injury, pulmonary hypertension, restenosis after angioplasty, intermittent claudication, post-prandial lipemia, metabolic acidosis, ketosis, arthritis, osteoporosis, Parkinson's Disease, left ventricular hypertrophy, peripheral arterial disease, macular degeneration, cataract, glomerulosclerosis, chronic renal failure, metabolic syndrome, syndrome X, premenstrual syndrome, angina pectoris, thrombosis, atherosclerosis, transient ischemic attacks, vascular restenosis, impaired glucose metabolism, conditions of impaired fasting plasma glucose, hyperuricemia, gout, erectile dysfunction, skin and connective tissue disorders, psoriasis, foot ulcerations, ulcerative colitis, hyper apo B lipoproteinemia, Alzheimer's Disease, schizophrenia, impaired cognition, inflammatory bowel disease, short bowel syndrome, Crohn's disease, colitis, irritable bowel syndrome, prevention or treatment of Polycystic Ovary Syndrome and treatment of addiction (e.g., alcohol and/or drug abuse). Abbreviations used herein are as follows: The term “alkyl”, as used herein, means a straight or branched chain monovalent hydrocarbon group of formula —CnH(2n+1). Non-limiting examples include methyl, ethyl, propyl, butyl, 2-methyl-propyl, 1,1-dimethylethyl, pentyl and hexyl. The term “alkylene”, as used herein, means a straight or branched chain divalent hydrocarbon group of formula —CnH2n—. Non-limiting examples include ethylene, and propylene. The term “cycloalkyl”, as used herein, means a cyclic, monovalent hydrocarbon group of formula —CnH(2n−1)containing at least three carbon atoms. Non-limiting examples include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The term “halogen”, as used herein, refers to fluoride, chloride, bromide, or iodide. The term “heterocycloalkyl”, as used herein, refers to a cycloalkyl group in which one or more of the ring methylene groups (—CH2—) has been replaced with a group selected from —O—, —S— or nitrogen, wherein the nitrogen may provide a point of attachment or may be substituted as provided within each embodiment. Where nitrogen provides a point of attachment, a structural drawing of a heterocycloalkyl would have an hydrogen on said nitrogen. Generally, the heterocycloalkyl may be substituted with 0 to 2 substituents as valency allows independently selected from oxo, —CN, halogen, alkyl and —Oalkyl and the alkyl may be further substituted. One will note that when there is 0 substitution, the heterocycloalkyl is unsubstituted. The term “heteroaryl”, as used herein, refers to a monocyclic aromatic hydrocarbon containing from 5 to 6 carbon atoms in which at least one of the ring carbon atoms has been replaced with a heteroatom selected from oxygen, nitrogen and sulfur. Such a heteroaryl group may be attached through a ring carbon atom or, where valency permits, through a ring nitrogen atom. Generally, the heteroaryl may be substituted with 0 to 2 substituents as valency allows independently selected from halogen, OH, alkyl, O-alkyl, and amino (e.g., NH2, NHalkyl, N(alkyl)2), and the alkyl may be further substituted. One will note that when there is 0 substitution, the heteroaryl is unsubstituted.Room temperature: RT.Methanol: MeOH.Ethanol: EtOH.Isopropanol: iPrOH.Ethyl acetate: EtOAc.Tetrahydrofuran: THF.Toluene: PhCH3.Cesium carbonate: Cs2CO3.Lithium bis(trimethylsilyl)amide: LiHMDS.Sodium t-butoxide: NaOtBu.Potassium t-butoxide: KOtBu.Lithium diisopropylamide: LDA.Triethylamine: Et3N.N,N-diisopropylethyl amine: DIPEA.Potassium carbonate: K2CO3.Dimethyl formamide: DMF.Dimethyl acetamide: DMAc.Dimethyl sulfoxide: DMSO.N-Methyl-2-pyrrolidinone: NMP.Sodium hydride: NaH.Trifluoroacetic acid: TFA.Trifluoroacetic anhydride: TFAA.Acetic anhydride: Ac2O.Dichloromethane: DCM.1,2-Dichloroethane: DCE.Hydrochloric acid: HCl.1,8-Diazabicyclo[5.4.0]undec-7-ene: DBU.Borane-dimethylsulfide complex: BH3-DMS.Borane-tetrahydrofuran complex: BH3-THF.Lithium aluminum hydride: LAH.Acetic acid: AcOH.Acetonitrile: MeCN.p-Toluenesulfonic acid: pTSA.Dibenzylidine acetone: DBA.2,2′-Bis(diphenylphosphino)-1,1′-binaphthalene: BINAP.1,1′-Ferrocenediyl-bis(diphenylphosphine): dppf.1,3-Bis(diphenylphosphino)propane: DPPP.3-Chloroperbenzoic acid: m-CPBA.Tert-Butyl methyl ether: MTBE.Methanesulfonyl: Ms.N-Methylpyrrolidinone: NMP.Thin layer chromatography: TLC.Supercritical fluid chromatography: SFC.4-(Dimethylamino)pyridine: DMAP.Tert-Butyloxycarbonyl: Boc.1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate: HATU.Petroleum ether: PE.2-(1H-Benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate: HBTU.2-Amino-2-(hydroxymethyl)propane-1,3-diol: tris.tris(dibenzylideneacetone)dipalladium: Pd2(dba)3 1H Nuclear magnetic resonance (NMR) spectra were in all cases consistent with the proposed structures. Characteristic chemical shifts (δ) are given in parts-per-million relative to the residual proton signal in the deuterated solvent (CHCl3at 7.27 ppm; CD2HOD at 3.31 ppm; MeCN at 1.94 ppm; DMSO at 2.50 ppm) and are reported using conventional abbreviations for designation of major peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad. As used herein, a wavy line, “” denotes a point of attachment of a substituent to another group. The compounds and intermediates described below were named using the naming convention provided with ACD/ChemSketch 2012, File Version C10H41, Build 69045 (Advanced Chemistry Development, Inc., Toronto, Ontario, Canada). The naming convention provided with ACD/ChemSketch 2012 is well-known by those skilled in the art and it is believed that the naming convention provided with ACD/ChemSketch 2012 generally comports with the IUPAC (International Union for Pure and Applied Chemistry) recommendations on Nomenclature of Organic Chemistry and the CAS Index rules. One will note that the chemical names may have only parentheses or may have parentheses and brackets. The stereochemical descriptors may also be placed different locations within the name itself, depending on the naming convention. One of ordinary skill in the art will recognize these formatting variations and understand they provide the same chemical structure. Pharmaceutically acceptable salts of the compounds of Formula I include acid addition and base salts. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulfate/sulfate, borate, camsylate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulfate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate, 1,5-naphathalenedisulfonic acid and xinafoate salts. Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, bis(2-hydroxyethyl)amine (diolamine), glycine, lysine, magnesium, meglumine, 2-aminoethanol (olamine), potassium, sodium, 2-Amino-2-(hydroxymethyl)propane-1,3-diol (tris or tromethamine) and zinc salts. Hemisalts of acids and bases may also be formed, for example, hemisulfate and hemicalcium salts. For a review on suitable salts, see Handbook of Pharmaceutical Salts: Properties, Selection, and Use by Stahl and Wermuth (Wiley-VCH, 2002). Pharmaceutically acceptable salts of compounds of Formula I may be prepared by one or more of three methods:(i) by reacting the compound of Formula I with the desired acid or base;(ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of Formula I or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base; or(iii) by converting one salt of the compound of Formula I to another by reaction with an appropriate acid or base or by means of a suitable ion exchange column. All three reactions are typically carried out in solution. The resulting salt may precipitate out and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionisation in the resulting salt may vary from completely ionised to almost non-ionised. The compounds of Formula I, and pharmaceutically acceptable salts thereof, may exist in unsolvated and solvated forms. The term ‘solvate’ is used herein to describe a molecular complex comprising the compound of Formula I, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable solvent molecules, for example, ethanol. The term ‘hydrate’ is employed when said solvent is water. A currently accepted classification system for organic hydrates is one that defines isolated site, channel, or metal-ion coordinated hydrates—seePolymorphism in Pharmaceutical Solidsby K. R. Morris (Ed. H. G. Brittain, Marcel Dekker, 1995). Isolated site hydrates are ones in which the water molecules are isolated from direct contact with each other by intervening organic molecules. In channel hydrates, the water molecules lie in lattice channels where they are next to other water molecules. In metal-ion coordinated hydrates, the water molecules are bonded to the metal ion. When the solvent or water is tightly bound, the complex may have a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content may be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm. Also included within the scope of the invention are multi-component complexes (other than salts and solvates) wherein the drug and at least one other component are present in stoichiometric or non-stoichiometric amounts. Complexes of this type include clathrates (drug-host inclusion complexes) and co-crystals. The latter are typically defined as crystalline complexes of neutral molecular constituents which are bound together through non-covalent interactions, but could also be a complex of a neutral molecule with a salt. Co-crystals may be prepared by melt crystallisation, by recrystallisation from solvents, or by physically grinding the components together—see Chem Commun, 17, 1889-1896, by O. Almarsson and M. J. Zaworotko (2004). For a general review of multi-component complexes, see J Pharm Sci, 64 (8), 1269-1288, by Haleblian (August 1975). The compounds of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline. The term ‘amorphous’ refers to a state in which the material lacks long range order at the molecular level and, depending upon temperature, may exhibit the physical properties of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid. Upon heating, a change from solid to liquid properties occurs which is characterised by a change of state, typically second order (‘glass transition’). The term ‘crystalline’ refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterised by a phase change, typically first order (‘melting point’). The compounds of Formula I may also exist in a mesomorphic state (mesophase or liquid crystal) when subjected to suitable conditions. The mesomorphic state is intermediate between the true crystalline state and the true liquid state (either melt or solution). Mesomorphism arising as the result of a change in temperature is described as ‘thermotropic’ and that resulting from the addition of a second component, such as water or another solvent, is described as ‘lyotropic’. Compounds that have the potential to form lyotropic mesophases are described as ‘amphiphilic’ and consist of molecules which possess an ionic (such as —COO−Na+, —COO−K+, or —SO3−Na+) or non-ionic (such as —N−N+(CH3)3) polar head group. For more information, seeCrystals and the Polarizing Microscopeby N. H. Hartshorne and A. Stuart, 4thEdition (Edward Arnold, 1970). The compounds of Formula I may exhibit polymorphism and/or one or more kinds of isomerism (e.g. optical, geometric or tautomeric isomerism). The compounds of Formula I may also be isotopically labelled. Such variation is implicit to the compounds of Formula I defined as they are by reference to their structural features and therefore within the scope of the invention. Compounds of Formula I containing one or more asymmetric carbon atoms can exist as two or more stereoisomers. Where a compound of Formula I contains an alkenyl or alkenylene group, geometric cis/trans (or Z/E) isomers are possible. Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism (‘tautomerism’) can occur. This can take the form of proton tautomerism in compounds of Formula I containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism. The pharmaceutically acceptable salts of compounds of Formula I may also contain a counterion which is optically active (e.g. d-lactate or l-lysine) or racemic (e.g. dl-tartrate or dl-arginine). Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). Alternatively, a racemic precursor containing a chiral ester may be separated by enzymatic resolution (see, for example, Int J Mol Sci 29682-29716 by A. C. L. M. Carvaho et. al. (2015)). In the case where the compound of Formula I contains an acidic or basic moiety, a salt may be formed with an optically pure base or acid such as 1-phenylethylamine or tartaric acid. The resulting diastereomeric mixture may be separated by fractional crystallization and one or both of the diastereomeric salts converted to the corresponding pure enantiomer(s) by means well known to a skilled person. Alternatively, the racemate (or a racemic precursor) may be covalently reacted with a suitable optically active compound, for example, an alcohol, amine or benzylic chloride. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization by means well known to a skilled person to give the separated diastereomers as single enantiomers with 2 or more chiral centers. Chiral compounds of Formula I (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1% diethylamine. Concentration of the eluate affords the enriched mixture. Chiral chromatography using sub- and supercritical fluids may be employed. Methods for chiral chromatography useful in some embodiments of the present invention are known in the art (see, for example, Smith, Roger M., Loughborough University, Loughborough, UK; Chromatographic Science Series (1998), 75 (SFC with Packed Columns), pp. 223-249 and references cited therein). In some relevant examples herein, columns were obtained from Chiral Technologies, Inc, West Chester, Pennsylvania, USA, a subsidiary of Daicel® Chemical Industries, Ltd., Tokyo, Japan. When any racemate crystallises, crystals of two different types are possible. The first type is the racemic compound (true racemate) referred to above wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts. The second type is the racemic mixture or conglomerate wherein two forms of crystal are produced in equimolar amounts each comprising a single enantiomer. While both of the crystal forms present in a racemic mixture have identical physical properties, they may have different physical properties compared to the true racemate. Racemic mixtures may be separated by conventional techniques known to those skilled in the art—see, for example,Stereochemistry of Organic Compoundsby E. L. Eliel and S. H. Wilen (Wiley, 1994). It must be emphasised that the compounds of Formula I have been drawn herein in a single tautomeric form, all possible tautomeric forms are included within the scope of the invention. The present invention includes all pharmaceutically acceptable isotopically-labeled compounds of Formula I wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as2H and3H, carbon, such as11C,13C and14C, chlorine, such as36Cl, fluorine, such as18F, iodine, such as123I and125I, nitrogen, such as13N and15N, oxygen, such as15O,17O and18O, phosphorus, such as32P, and sulfur, such as35S. Certain isotopically-labelled compounds of Formula I, for example those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e.3H, and carbon-14, i.e.14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, i.e.2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements. Substitution with positron emitting isotopes, such as11C,18F,15O and13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of Formula I can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed. Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D2O, d6-acetone, d6-DMSO. One way of carrying out the invention is to administer a compound of Formula I in the form of a prodrug. Thus, certain derivatives of a compound of Formula I which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into a compound of Formula I having the desired activity, for example by hydrolytic cleavage, particularly hydrolytic cleavage promoted by an esterase or peptidase enzyme. Such derivatives are referred to as ‘prodrugs’. Further information on the use of prodrugs may be found in ‘Prodrugs as Novel Delivery Systems’, Vol. 14, ACS Symposium Series(T. Higuchi and W. Stella) and ‘Bioreversible Carriers in Drug Design’, Pergamon Press, 1987 (Ed. E. B. Roche, American Pharmaceutical Association). Reference can also be made toNature Reviews/Drug Discovery,2008, 7, 355 andCurrent Opinion in Drug Discovery and Development,2007, 10, 550. Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities present in the compounds of Formula I with certain moieties known to those skilled in the art as ‘pro-moieties’ as described, for example, in ‘Design of Prodrugs’ by H. Bundgaard (Elsevier, 1985) and Y. M. Choi-Sledeski and C. G. Wermuth, ‘Designing Prodrugs and Bioprecursors’ in Practice of Medicinal Chemistry, (Fourth Edition), Chapter 28, 657-696 (Elsevier, 2015). Thus, a prodrug in accordance with the invention is (a) an ester or amide derivative of a carboxylic acid in a compound of Formula I; (b) an ester, carbonate, carbamate, phosphate or ether derivative of a hydroxyl group in a compound of Formula I; (c) an amide, imine, carbamate or amine derivative of an amino group in a compound form Formula I; (d) an oxime or imine derivative of a carbonyl group in a compound of Formula I; or (e) a methyl, primary alcohol or aldehyde group that can be metabolically oxidized to a carboxylic acid in a compound of Formula I. Some specific examples of prodrugs in accordance with the invention include:(i) where the compound of Formula I contains a carboxylic acid functionality (—COOH), an ester thereof, such as a compound wherein the hydrogen of the carboxylic acid functionality of the compound of Formula I is replaced by C1-C8alkyl (e.g. ethyl) or (C1-C8alkyl)C(═O)OCH2— (e.g.tBuC(═O)OCH2—);(ii) where the compound of Formula I contains an alcohol functionality (—OH), an ester thereof, such as a compound wherein the hydrogen of the alcohol functionality of the compound of Formula I is replaced by —CO(C1-C8alkyl) (e.g. methylcarbonyl) or the alcohol is esterified with an amino acid;(iii) where the compound of Formula I contains an alcohol functionality (—OH), an ether thereof, such as a compound wherein the hydrogen of the alcohol functionality of the compound of Formula I is replaced by (C1-C8alkyl)C(═O)OCH2— or —CH2OP(═O)(OH)2;(iv) where the compound of Formula I contains an alcohol functionality (—OH), a phosphate thereof, such as a compound wherein the hydrogen of the alcohol functionality of the compound of Formula I is replaced by —P(═O)(OH)2or —P(═O)(ONa)2or —P(═O)(O−)2Ca2+;(v) where the compound of Formula I contains a primary or secondary amino functionality (—NH2or —NHR where R≠H), an amide thereof, for example, a compound wherein, as the case may be, one or both hydrogens of the amino functionality of the compound of Formula I is/are replaced by (C1-C10)alkanoyl, —COCH2NH2or the amino group is derivatised with an amino acid;(vi) where the compound of Formula I contains a primary or secondary amino functionality (—NH2or —NHR where R≠H), an amine thereof, for example, a compound wherein, as the case may be, one or both hydrogens of the amino functionality of the compound of Formula I is/are replaced by —CH2OP(═O)(OH)2;(vii) where the carboxylic acid group within compound of Formula I is replaced by a methyl group, a —CH2OH group or an aldehyde group. Certain compounds of Formula I may themselves act as prodrugs of other compounds of Formula I. It is also possible for two compounds of Formula I to be joined together in the form of a prodrug. In certain circumstances, a prodrug of a compound of Formula I may be created by internally linking two functional groups in a compound of Formula I, for instance by forming a lactone. References to compounds of Formula I are taken to include the compounds themselves and prodrugs thereof. The invention includes such compounds of Formula I as well as pharmaceutically acceptable salts of such compounds and pharmaceutically acceptable solvates of said compounds and salts. Administration and Dosing Typically, a compound of the invention is administered in an amount effective to treat a condition as described herein. The compounds of the invention can be administered as compound per se, or alternatively, as a pharmaceutically acceptable salt. For administration and dosing purposes, the compound per se or pharmaceutically acceptable salt thereof will simply be referred to as the compounds of the invention. The compounds of the invention are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. The compounds of the invention may be administered orally, rectally, vaginally, parenterally, or topically. The compounds of the invention may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the bloodstream directly from the mouth. In another embodiment, the compounds of the invention may also be administered directly into the bloodstream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques. In another embodiment, the compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermally. In another embodiment, the compounds of the invention can also be administered intranasally or by inhalation. In another embodiment, the compounds of the invention may be administered rectally or vaginally. In another embodiment, the compounds of the invention may also be administered directly to the eye or ear. The dosage regimen for the compounds of the invention and/or compositions containing said compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely. In one embodiment, the total daily dose of a compound of the invention is typically from about 0.001 to about 100 mg/kg (i.e., mg compound of the invention per kg body weight) for the treatment of the indicated conditions discussed herein. In another embodiment, total daily dose of the compound of the invention is from about 0.01 to about 30 mg/kg, and in another embodiment, from about 0.03 to about 10 mg/kg, and in yet another embodiment, from about 0.1 to about 3. It is not uncommon that the administration of the compounds of the invention will be repeated a plurality of times in a day (typically no greater than 4 times). Multiple doses per day typically may be used to increase the total daily dose, if desired. For oral administration, the compositions may be provided in the form of tablets containing 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 30.0 50.0, 75.0, 100, 125, 150, 175, 200, 250 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, or in another embodiment, from about 1 mg to about 100 mg of active ingredient. Intravenously, doses may range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion. Suitable subjects according to the invention include mammalian subjects. In one embodiment, humans are suitable subjects. Human subjects may be of either gender and at any stage of development. Pharmaceutical Compositions In another embodiment, the invention comprises pharmaceutical compositions. Such pharmaceutical compositions comprise a compound of the invention presented with a pharmaceutically acceptable carrier. Other pharmacologically active substances can also be present. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof, and may include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol, or sorbitol in the composition. Pharmaceutically acceptable substances such as wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion. The compositions of this invention may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The form depends on the intended mode of administration and therapeutic application. Typical compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with antibodies in general. One mode of administration is parenteral (e.g. intravenous, subcutaneous, intraperitoneal, intramuscular). In another embodiment, the antibody is administered by intravenous infusion or injection. In yet another embodiment, the antibody is administered by intramuscular or subcutaneous injection. Oral administration of a solid dose form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the invention. In another embodiment, the oral administration may be in a powder or granule form. In another embodiment, the oral dose form is sub-lingual, such as, for example, a lozenge. In such solid dosage forms, the compounds of Formula I are ordinarily combined with one or more adjuvants. Such capsules or tablets may contain a controlled release formulation. In the case of capsules, tablets, and pills, the dosage forms also may comprise buffering agents or may be prepared with enteric coatings. In another embodiment, oral administration may be in a liquid dose form. Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents. In another embodiment, the invention comprises a parenteral dose form. “Parenteral administration” includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrasternal injections, and infusion. Injectable preparations (i.e., sterile injectable aqueous or oleaginous suspensions) may be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents. In another embodiment, the invention comprises a topical dose form. “Topical administration” includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration. Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams. A topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. When the compounds of this invention are administered by a transdermal device, administration will be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated—see, for example, B. C. Finnin and T. M. Morgan, J. Pharm. Sci., vol. 88, pp. 955-958, 1999. Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in a suitable carrier. A typical formulation suitable for ocular or aural administration may be in the form of drops of a micronized suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and aural administration include ointments, biodegradable (i.e., absorbable gel sponges, collagen) and non-biodegradable (i.e., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed linked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methylcellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis. For intranasal administration or administration by inhalation, the compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant. Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurized container, pump, spray, atomizer (preferably an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or without the use of a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin. In another embodiment, the invention comprises a rectal dose form. Such rectal dose form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate. Other carrier materials and modes of administration known in the pharmaceutical art may also be used. Pharmaceutical compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures. The above considerations in regard to effective formulations and administration procedures are well known in the art and are described in standard textbooks. Formulation of drugs is discussed in, for example, Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania, 1975; Liberman et al., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Kibbe et al., Eds., Handbook of Pharmaceutical Excipients (3rd Ed.), American Pharmaceutical Association, Washington, 1999. Co-Administration The compounds of the invention can be used alone, or in combination with other therapeutic agents. The invention provides any of the uses, methods or compositions as defined herein wherein the compound of any embodiment of Formula I herein, or pharmaceutically acceptable salt thereof, or pharmaceutically acceptable solvate of said compound or salt, is used in combination with one or more other therapeutic agent discussed herein. This would include a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, or VI or a pharmaceutically acceptable salt thereof, as defined in any of the embodiments described herein, in admixture with at least one pharmaceutically acceptable excipient and one or more other therapeutic agent discussed herein. The administration of two or more compounds “in combination” means that all of the compounds are administered closely enough in time that each may generate a biological effect in the same time frame. The presence of one agent may alter the biological effects of the other compound(s). The two or more compounds may be administered simultaneously, concurrently or sequentially. Additionally, simultaneous administration may be carried out by mixing the compounds prior to administration or by administering the compounds at the same point in time but as separate dosage forms at the same or different site of administration. The phrases “concurrent administration,” “co-administration,” “simultaneous administration,” and “administered simultaneously” mean that the compounds are administered in combination. In another embodiment, the invention provides methods of treatment that include administering compounds of the present invention in combination with one or more other pharmaceutical agents, wherein the one or more other pharmaceutical agents may be selected from the agents discussed herein. In one embodiment, the compounds of this invention are administered with an anti-diabetic agent including but not limited to a biguanide (e.g., metformin), a sulfonylurea (e.g., tolbutamide, glibenclamide, gliclazide, chlorpropamide, tolazamide, acetohexamide, glyclopyramide, glimepiride, or glipizide), a thiazolidinedione (e.g., pioglitazone, rosiglitazone, or lobeglitazone), a glitazar (e.g., saroglitazar, aleglitazar, muraglitazar or tesaglitazar), a meglitinide (e.g., nateglinide, repaglinide), a dipeptidyl peptidase 4 (DPP-4) inhibitor (e.g., sitagliptin, vildagliptin, saxagliptin, linagliptin, gemigliptin, anagliptin, teneligliptin, alogliptin, trelagliptin, dutogliptin, or omarigliptin), a glitazone (e.g., pioglitazone, rosiglitazone, balaglitazone, rivoglitazone, or lobeglitazone), a sodium-glucose linked transporter 2 (SGLT2) inhibitor (e.g., empagliflozin, canagliflozin, dapagliflozin, ipragliflozin, ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, or ertugliflozin), an SGLTL1 inhibitor, a GPR40 agonist (FFAR1/FFA1 agonist, e.g. fasiglifam), glucose-dependent insulinotropic peptide (GIP) and analogues thereof, an alpha glucosidase inhibitor (e.g. voglibose, acarbose, or miglitol), or an insulin or an insulin analogue, including the pharmaceutically acceptable salts of the specifically named agents and the pharmaceutically acceptable solvates of said agents and salts. In another embodiment, the compounds of this invention are administered with an anti-obesity agent including but not limited to peptide YY or an analogue thereof, a neuropeptide Y receptor type 2 (NPYR2) agonist, a NPYR1 or NPYR5 antagonist, a cannabinoid receptor type 1 (CB1R) antagonist, a lipase inhibitor (e.g., orlistat), a human proislet peptide (HIP), a melanocortin receptor 4 agonist (e.g., setmelanotide), a melanin concentrating hormone receptor 1 antagonist, a farnesoid X receptor (FXR) agonist (e.g. obeticholic acid), zonisamide, phentermine (alone or in combination with topiramate), a norepinephrine/dopamine reuptake inhibitor (e.g., buproprion), an opioid receptor antagonist (e.g., naltrexone), a combination of norepinephrine/dopamine reuptake inhibitor and opioid receptor antagonist (e.g., a combination of bupropion and naltrexone), a GDF-15 analog, sibutramine, a cholecystokinin agonist, amylin and analogues thereof (e.g., pramlintide), leptin and analogues thereof (e.g., metroleptin), a serotonergic agent (e.g., lorcaserin), a methionine aminopeptidase 2 (MetAP2) inhibitor (e.g., beloranib or ZGN-1061), phendimetrazine, diethylpropion, benzphetamine, an SGLT2 inhibitor (e.g., empagliflozin, canagliflozin, dapagliflozin, ipragliflozin, Ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, or ertugliflozin), an SGLTL1 inhibitor, a dual SGLT2/SGLT1 inhibitor, a fibroblast growth factor receptor (FGFR) modulator, an AMP-activated protein kinase (AMPK) activator, biotin, a MAS receptor modulator, or a glucagon receptor agonist (alone or in combination with another GLP-1R agonist, e.g., liraglutide, exenatide, dulaglutide, albiglutide, lixisenatide, or semaglutide), including the pharmaceutically acceptable salts of the specifically named agents and the pharmaceutically acceptable solvates of said agents and salts. In another embodiment, the compounds of this invention are administered with an agent to treat NASH including but not limited to PF-05221304, an FXR agonist (e.g., obeticholic acid), a PPAR α/δ agonist (e.g., elafibranor), a synthetic fatty acid-bile acid conjugate (e.g., aramchol), a caspase inhibitor (e.g., emricasan), an anti-lysyl oxidase homologue 2 (LOXL2) monoclonal antibody (e.g., simtuzumab), a galectin 3 inhibitor (e.g., GR-MD-02), a MAPK5 inhibitor (e.g., GS-4997), a dual antagonist of chemokine receptor 2 (CCR2) and CCR5 (e.g., cenicriviroc), a fibroblast growth factor 21 (FGF21) agonist (e.g., BMS-986036), a leukotriene D4 (LTD4) receptor antagonist (e.g., tipelukast), a niacin analogue (e.g., ARI 3037MO), an ASBT inhibitor (e.g., volixibat), an acetyl-CoA carboxylase (ACC) inhibitor (e.g., NDI 010976), a ketohexokinase (KHK) inhibitor, a diacylglyceryl acyltransferase 2 (DGAT2) inhibitor, a CB1 receptor antagonist, an anti-CB1R antibody, or an apoptosis signal-regulating kinase 1 (ASK1) inhibitor, including the pharmaceutically acceptable salts of the specifically named agents and the pharmaceutically acceptable solvates of said agents and salts. These agents and compounds of the invention can be combined with pharmaceutically acceptable vehicles such as saline, Ringer's solution, dextrose solution, and the like. The particular dosage regimen, i.e., dose, timing and repetition, will depend on the particular individual and that individual's medical history. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and may comprise buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or Igs; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). Liposomes containing these agents and/or compounds of the invention are prepared by methods known in the art, such as described in U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556. Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter. These agents and/or the compounds of the invention may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington, The Science and Practice of Pharmacy, 20th Ed., Mack Publishing (2000). Sustained-release preparations may be used. Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing the compound of Formulas I, II, III, IV, V, or VI, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or ‘poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as those used in LUPRON DEPOT™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(−)-3-hydroxybutyric acid. The formulations to be used for intravenous administration must be sterile. This is readily accomplished by, for example, filtration through sterile filtration membranes. Compounds of the invention are generally placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. Suitable emulsions may be prepared using commercially available fat emulsions, such as Intralipid™, Liposyn™, Infonutrol™, Lipofundin™ and Lipiphysan™. The active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g., egg phospholipids, soybean phospholipids or soybean lecithin) and water. It will be appreciated that other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emulsion. Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%. The fat emulsion can comprise fat droplets between 0.1 and 1.0 μm, particularly 0.1 and 0.5 μm, and have a pH in the range of 5.5 to 8.0. The emulsion compositions can be those prepared by mixing a compound of the invention with Intralipid™ or the components thereof (soybean oil, egg phospholipids, glycerol and water). Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulised by use of gases. Nebulised solutions may be breathed directly from the nebulising device or the nebulising device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner. Kits Another aspect of the invention provides kits comprising the compound of Formulas I, II, or III or pharmaceutical compositions comprising the compound of Formulas I, II, or III of the invention. A kit may include, in addition to the compound of Formulas I, II, or III, of the invention or pharmaceutical composition thereof, diagnostic or therapeutic agents. A kit may also include instructions for use in a diagnostic or therapeutic method. In some embodiments, the kit includes the compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutical composition thereof and a diagnostic agent. In other embodiments, the kit includes the compound of Formulas I, II, III, IV, V, or VI, or a pharmaceutical composition thereof. In yet another embodiment, the invention comprises kits that are suitable for use in performing the methods of treatment described herein. In one embodiment, the kit contains a first dosage form comprising one or more of the compounds of the invention in quantities sufficient to carry out the methods of the invention. In another embodiment, the kit comprises one or more compounds of the invention in quantities sufficient to carry out the methods of the invention and a container for the dosage and a container for the dosage. Preparation The compounds of Formulas I, II, III, IV, V, or VI, may be prepared by the general and specific methods described below, using the common general knowledge of one skilled in the art of synthetic organic chemistry. Such common general knowledge can be found in standard reference books such as Comprehensive Organic Chemistry, Ed. Barton and Ollis, Elsevier; Comprehensive Organic Transformations: A Guide to Functional Group Preparations, Larock, John Wiley and Sons; and Compendium of Organic Synthetic Methods, Vol. I-XII (published by Wiley-Interscience). The starting materials used herein are commercially available or may be prepared by routine methods known in the art. In the preparation of the compounds of Formulas I, II, III, IV, V, or VI, it is noted that some of the preparation methods described herein may require protection of remote functionality (e.g., primary amine, secondary amine, carboxyl in Formula I precursors). The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. The need for such protection is readily determined by one skilled in the art. The use of such protection/deprotection methods is also within the skill in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991. For example, certain compounds contain primary amines or carboxylic acid functionalities which may interfere with reactions at other sites of the molecule if left unprotected. Accordingly, such functionalities may be protected by an appropriate protecting group which may be removed in a subsequent step. Suitable protecting groups for amine and carboxylic acid protection include those protecting groups commonly used in peptide synthesis (such as N-t-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), and 9-fluorenylmethylenoxycarbonyl (Fmoc) for amines and lower alkyl or benzyl esters for carboxylic acids) which are generally not chemically reactive under the reaction conditions described and can typically be removed without chemically altering other functionality in the Formula I compounds. The Schemes described below are intended to provide a general description of the methodology employed in the preparation of the compounds of the present invention. Some of the compounds of the present invention may contain single or multiple chiral centers with the stereochemical designation (R) or (S). It will be apparent to one skilled in the art that all of the synthetic transformations can be conducted in a similar manner whether the materials are enantioenriched or racemic. Moreover the resolution to the desired optically active material may take place at any desired point in the sequence using well known methods such as described herein and in the chemistry literature. For example, intermediates (e.g., S29, S32, S37 and S48) and finals (e.g., S49) may be separated using chiral chromatographic methods. Alternatively, chiral salts may be utilized to isolate enantiomerically enriched intermediates and final compounds. In the Schemes that follow, the variables A, A1, A2, X, Y, L, Z1, Z2, Z3, ZA1, ZA2, ZA3, R1, R2, R3, R4, R5, R6, m, and q are as described herein for compounds of Formulas I, II, III, IV, V, or VI unless otherwise noted. For the Schemes provided below, each p is independently 0 or 1. For the Schemes provided below, each X1, X2, X3, and X4can independently be a leaving group such as any alkyl or aryl sulfonate (e.g., mesylate, tosylate, or triflate), or a halogen or any other group that can be displaced by an amine or utilized in a metal mediated coupling reaction. X4may also be a protected carboxylic acid (i.e., ester). When the protecting group is identified as Pg1, it can be an alkyl amine protecting group such as benzyl, benzhydryl, or the like; a carbamate protecting group such as Boc, Cbz, or the like; or an amide protecting group such trifluoroacetamide. When the protecting group is identified as Pg2, it can be acid protecting group such as methyl, ethyl, benzyl, t-butyl or the like. R4ais C1-2alkyl, C0-2alkylene-C3-6cycloalkyl, C0-2alkylene-R5, or C1-2alkylene-R6, wherein said alkyl, alkylene, or cycloalkyl may be independently substituted as valency allows with 0 to 3 F atoms and 0 to 1 substituent independently selected from C0-1alkylene-OROand —N(RN)2. The substituted piperidine of general structure S6 where Y=CH may be prepared as discussed in Scheme 1. The heterocycle of general structure S1 may be reacted with a substituted boronic acid or boronate ester (S2) in the presence of a palladium catalyst and ligand complex in the manner of a Suzuki reaction (Maluenda and Navarro, Molecules, 2015, 20, 7528-7557) to provide compounds of the general formula S3. When ZA1is CH or CR2, a preferred X1leaving group is F or SO2Me (from oxidation of SMe, as described in Scheme 3) and preferred X2substituents include Br and I. When ZA1is N, Cl is preferred for both X1and X2. Reduction of the olefin to provide compounds of general structure S4 could be performed under an atmosphere of hydrogen (15-100 psi H2) in an alcoholic solvent such as MeOH or EtOH or alternatively an aprotic organic solvent such as EtOAc or THF in the presence of an appropriate catalyst such as palladium on carbon, Pd(OH)2on carbon (Pearlman's catalyst), PtO2(Adams catalyst), or tris(triphenylphosphine)rhodium(I) chloride (Wilkinson's catalyst). Transfer hydrogenation reagents, for example ammonium formate or dihydrobenzene, or similar, may be employed using suitable catalyst. Alternatively, the reduction may be accomplished by alternative methods know to those skilled in the art using reagents such as triethyl silane or other silanes, under acid or metallic catalysis, or metallic reductants, such as magnesium or similar. Alternatively, the olefin can be functionalized by methods known to one skilled in the art to introduce R3groups. For example, the olefin could be hydroborated to produce an alcohol that could be alkylated or further converted to a nitrile, F or alkyl group. Conversion to compounds of general structure S6 can be accomplished by such manner as a Buchwald-Hartwig C—O coupling (Lundgren and Stradiotto, Aldrich Chimica Acta, 2012, 45, 59-65) between compounds of the general structure S4 and an appropriately substituted benzyl alcohol S5 in the presence of a palladium or copper catalyst and ligand complex. A preferred X1halogen is Cl. These reactions are generally performed between 0 and 110° C. in aprotic organic solvents such as but not limited to 1,4-dioxane and PhCH3with added base such as Cs2CO3, LiHMDS or NaOtBu. Alternatively, reaction of S4 with an appropriately substituted benzyl alcohol S5 in an aprotic solvent such as DMF or THF in the presence of a strong base such as NaH, KOtBu or LiHMDS can deliver compounds of the general structure S6. Preferred X1substituents for this reaction include F and Cl or sulfones (e.g. SO2Me). The substituted piperazine of general structure S6 where Y=N may be prepared as discussed in Scheme 2. Conversion of S1 to compounds of general structure S7 can be accomplished by reaction of S1 with an appropriately substituted benzyl alcohol S5 in an aprotic solvent such as DMF or THF in the presence of a strong base such as NaH, KOtBu or LiHMDS, to deliver compounds of the general structure S7. When ZA1is CH or CR2, a preferred X1leaving group is F, and preferred X2substituents include Br and I. When ZA1is N, Cl is preferred for both X1and X2. Conversion of S7 to compounds of general structure S6 where Y=N may be accomplished by such manner as a Buchwald-Hartwig C—N coupling between compounds of the general structure S7 and an appropriately substituted and protected piperazine S8 in the presence of a palladium or copper catalyst and ligand complex. These reactions are generally performed between 0 and 110° C. in aprotic organic solvents such as but not limited to 1,4-dioxane and PhCH3with added base such as Cs2CO3, LiHMDS or NaOtBu. As shown in Scheme 3, appropriately substituted piperidine esters of general structure S9 can be reacted with pyrimidines S11 in the presence of strong base such as LiHMDS or LDA or other suitable base in an aprotic organic solvent such as THF to deliver compounds of the general structure S12. In reactions of S9 with S11, X1is preferably Cl or more preferably SMe, X2is preferably Cl. Removal of Pg2through ester hydrolysis and decarboxylation of the resulting carboxylic acids may deliver piperidines S13 (X1=SMe) or S14 (X1=Cl). When X1is SMe, oxidation of the thioether with a peroxide such as meta-chloroperbenzoic acid may provide sulfoxides or sulfones S14 (X1=S(O)Me, SO2Me). Reaction of pyrimidine S14 (X1=Cl, S(O)Me, SO2Me) with an appropriately substituted benzyl alcohol S5 in an aprotic solvent such as DMF or THF in the presence of a strong base such as NaH, KOtBu, LiHMDS, or NaHMDS may deliver compounds of the general structure S15. Compounds of general structure S14 where Y=N may also be prepared as discussed in Scheme 4. Substituted pyrimidine S11 may react with piperidines of general structure S8 in the presence of a weak base, such as triethylamine or diisopropylethyl amine, in a solvent, such as methanol, ethanol, water, DMF, or THF, at a temperature around 30° C. to provide compounds of general structure S14. Cl is a preferred X2substituent. Compounds S14 may then be used to prepare S15 where Y=N as described in Scheme 1. for the preparation of S6 from S4, where preferred X1substituents include Cl, Br or I. As provided in Scheme 5, reaction of S17 with an appropriately substituted benzyl alcohol S5 in an aprotic solvent such as DMF or THF in the presence of a strong base such as NaH, KOtBu or LiHMDS can deliver compounds of the general structure S18. When electron withdrawing R2substituents are present bases such as K2CO3may be used. Preferred X1substituents for this reaction include F, Cl, and Br, while X2substituents may include Cl, Br or I. Alternatively, Buchwald-Hartwig C—O coupling conditions similar to the preparation of S6 may be used to prepare S18 with preferred X1substituents Cl, Br or I as described in Scheme 1. As provided in Scheme 6, reaction of pyrimidine S19 with an appropriately substituted benzyl alcohol S5 in an aprotic solvent such as DMF or THF in the presence of a strong base such as NaH, KOtBu or LiHMDS can deliver compounds of the general structure S20. A preferred X2substituent for this reaction is Cl. As provided in Scheme 7, reaction of pyrimidine S21 with an appropriately substituted benzyl alcohol S5 in an aprotic solvent such as DMF or THF in the presence of a strong base such as NaH, KOtBu or LiHMDS can deliver compounds of the general structure S22. Preferred X1and X2substituent for this reaction are Cl. The substituted piperidine of general structure S25 where Y=CH may be prepared as described in Scheme 8. Compounds of general structure S18, S20 and S22, collectively referred to as S23, may be reacted with a substituted boronic acid or boronate ester (S2) as described for S1 in Scheme 1. In the Suzuki reaction, the X2halogen is preferably Cl, Br or I. Reduction of the olefin as described in Scheme 1 may then provide compounds of general structure S25 where Y=CH. As provided in Scheme 9, conversion of S23 to compounds of general structure S25 where Y=N can be accomplished by such manner as a Buchwald-Hartwig C—N coupling between compounds of the general structure S23 and an appropriately substituted and protected piperazine in the presence of a palladium or copper catalyst and ligand complex as described in Scheme 2 for the conversion of S7 to S6. When X2is Cl and ZA3is N, S23 and S8 may react to produce S25 in the presence of a weak base, such as triethylamine or diisopropylethyl amine, in a solvent, such as methanol, ethanol, water, DMF, or THF, at a temperature around 30° C. Carboxylic acids of general structure S29 where X=N and L=CH2may be prepared as discussed in Scheme 10. Compounds of general structures S6, S15 and S25, prepared via methods described in Schemes 1-3, 8 and 9, and collectively referred to as S26, could be deprotected with many methods described in literature to provide amines of general structure S27. Amine compounds S27 can be alkylated with a protected 2-bromoacetate in the presence of a suitable base such as K2CO3, Et3N, NaH or LiHMDS in a polar aprotic solvent such as but not limited to DMF, DMAc, DMSO or NMP to deliver compounds of the general structure S28 where X=N and L=CH2. If Pg2is t-butyl, standard acidic deprotection methods such as TFA/DCM, HCl/1,4-dioxane, HCl/EtOAc or other suitable conditions may be used to deliver acids S29. If Pg2is methyl or ethyl, standard basic deprotection methods such as aqueous NaOH in methanol or ethanol, or other suitable conditions may be used to deliver acids S29. Compounds of general structure S29 where Y=N and X-L=cyclopropyl may be prepared as discussed in Scheme 11. Protected piperidinone S30 may be homologated to unsaturated ester S31 using methods well known to those skilled in the art. For example, Horner-Wadsworth-Emmons olefination of S30 with a phosphonate, such as ethyl (diethoxyphosphoryl)acetate, that has been deprotonated with a strong base such as sodium hydride or potassium tert-butoxide, may provide S31. The reaction is typically conducted in an aprotic solvent like THF or DME, at a temperature around 0 to −50° C. Conversion of S31 to the cyclopropane derivative S32 may be accomplished by treatment with sulfoxonium ylide derived from trimethylsulfoxonium iodide and a base, such as potassium tert-butoxide or sodium hydride. Removal of Pg1from S32 would then provide amines of general structure S33 where X-L is cyclopropyl. Aryl halides S7, S18, S20, and S22 are collectively represented by general structure S34. Coupling of S33 with compounds of general structure S34 in a manner similar to that described in Scheme 2 for the preparation of S6 from S7 and S8 provides S35 where Y=N and X-L is cyclopropyl. Removal of Pg2may then provide compounds of general structure S29 where Y=N and X-L=cyclopropyl. Alternatively, protected carboxylic acids of general structure S33 where X=N and L=CH2may be prepared as discussed in Scheme 12. Appropriately protected piperazines S8 can be alkylated with a protected 2-bromoacetate as described for the preparation of S28 in Scheme 10 to deliver compounds of the general structure S36. Removal of Pg1may then provide compounds of general structure S33 that may then be used to prepare S29 where X=N and L=CH2as described in Scheme 11. Compounds of general structure S29 where Y=N, A=A2, ZA2=N and ZA3=CH or CR2may also be prepared as discussed in Scheme 13. Substituted pyrimidine S16 may react with amines of general structure S33 in the presence of a weak base, such as triethylamine or diisopropylethyl amine, in a solvent, such as methanol, ethanol, water, DMF, or THF, at a temperature around 30° C. to provide compounds of general structure S37. Reaction of S37 with an appropriately substituted benzyl alcohol in an aprotic solvent such as DMF or THF in the presence of a strong base such as NaH, KOtBu or LiHMDS may deliver compounds of the general structure S38. Preferred X1and X2substituents for these reactions include Cl. Alternatively, Buchwald-Hartwig C—O coupling conditions similar to the preparation of S6 may be used to prepare S38 with preferred X1substituent Cl. Removal of Pg2may then give compounds of general structure S29 where Y=N, A=A2, ZA2=N and ZA3=CH or CR2. As shown in scheme 14, compounds of general structure S39 can react with amines R4NH2in the presence of bases such as sodium-, potassium-, or cesium carbonate, -bicarbonate, hydroxide, acetate, or an organic amine base such as Et3N, DIPEA, DBU, and the like in a polar aprotic solvent such as but not limited to THF, DMF, DMAc, DMSO or NMP or a protic solvent such as water, MeOH, EtOH or iPrOH or a mixture thereof to deliver compounds of the general structure S40. One will note that if an example provides an R4with a resolved enantiomeric center, the other enantiomer or a racemix mixture thereof could be obtained by selection of the appropriate starting material. Preferred X3substituents include F, Cl, and Br, preferred X4groups include Cl, Br, —CO2-Pg2. Reduction of the nitro group can be affected by hydrogenation at 1-6 atm H2with a metal catalyst such as palladium on carbon or Raney nickel in a protic solvent such as MeOH or EtOH or aprotic solvent such as DMF, THF or EtOAc. Alternatively, the nitro group may be reduced with iron, zinc, SnCl2or other suitable metal in an acidic media such as 1N HCl, AcOH or aqueous NH4Cl in THF to provide compounds of general structure S41 (Scheme 8a). Compounds such as S42 may be acylated by acyl halides by standard fashion or by carboxylates via standard amide coupling protocols to provide compounds S43. Reduction to compounds S44 may be performed under standard conditions with reducing agents such as LAH or BH3-THF or BH3-DMS (Scheme 14b). Diamine compounds S41 and S44 prepared via methods described in Schemes 14a and 14b, collectively designated as diamine S45 (Scheme 15), may be acylated with acids of general structure S29 under standard amide coupling protocols to deliver amines S46 which will exist as a mixture from 100% S46a to 100% S46b. This mixture of amines S46 may be cyclized to deliver compounds of general structure S47 by a variety of methods. Amines S46 may be heated with a dehydrating agent such as T3P® or an alkyl alcohol such as n-butanol under microwave conditions (10-60 min at 120-180° C.) to deliver compounds S47. Alternatively, the mixture of compounds S46 may be heated under acidic conditions such as AcOH from 60-100° C. or under basic conditions such as aqueous NaOH or KOH in 1,4-dioxane from 60-100° C. to provide S47. Compounds of general structure S47 (X4=Cl, Br or I) can be converted to esters of structure S48 by palladium-catalyzed carbonylation under a 15-100 psi carbon monoxide atmosphere at a temperature from 20-100 at a temperature from 20-100° C. with an appropriate alcohol such as MeOH or EtOH or other alkyl alcohol. Hydrolysis of ester S48 can be performed as described in Scheme 10 to provide acids S49. For compounds S46 where X4=CO2-Pg2conversion to ester S48 proceeds under similar conditions as described previously except for use of the basic cyclization method where compound S49 may be isolated directly from the reaction mixture. For compounds S48 where X4is CO2tBu, deprotection to acid 49 can be performed under acidic conditions described in Scheme 10. Alternatively, for compounds S48 where Pg2is a C1-C8alkyl, such as methyl, ethyl, hexyl or octyl, the ester deprotection may be performed with a variety of enzymes including esterases, proteases, peptidases, lipases, and glycosidases which are well known to those skilled in the art. The hydrolysis may also be performed by treating the ester with an aqueous solution of 1,5,7-triazabicyclo[4.4.0]dec-5-ene at RT. Additionally, diamine S45 may be converted to the 2-chloromethyl benzimidazole S50 (Scheme 16) by several methods. Treatment with 2-chloroacetyl chloride in an aprotic solvent such as 1,4-dioxane followed by heating at 40-100° C. for 2-18 h can deliver the desired benzimidazole S50 where Z1, Z2and Z3are CH. In the cases where Z1, Z2and Z3are not all CRz, after treatment with 2-chloroacetyl chloride in an aprotic solvent such as 1,4-dioxane for 30 min to 4 h, the solvent is exchanged for an acidic media such as AcOH or TFA followed by heating at 40-100° C. for 2-18 h to provide the desired compound S50. Diamine S45 can also be treated with chloroacetic anhydride at a temperature between 0 and 80° C. in an aprotic solvent such as, but not limited to 1,4-dioxane, THF or MeCN, followed by heating for 2 to 18 h at 60-100° C. to deliver the desired compound S50. In addition, diamine S45 can be treated with 2-chloro-1,1,1-trimethoxyethane in an aprotic solvent such as, but not limited to 1,4-dioxane, THF or MeCN, or a protic solvent, e.g., MeOH or EtOH, in the presence of an acid catalyst, e.g., pTSA, at 20-100° C. Alternatively, diamines S45 may be heated 100-180° C. with 2-hydroxyacetic acid in an aprotic solvent, such as but not limited to mesitylene, to provide a hydroxymethyl intermediate. Conversion of the hydroxymethyl group to the chloromethyl compound S50 may be accomplished by standard methods, including treatment with SOCl2in an aprotic solvent. Compounds of general structure S50 can be reacted with compounds S27 in the presence of bases such as sodium-, potassium-, or cesium carbonate, -bicarbonate, NaH or an organic amine base such as Et3N, DIPEA, DBU, and the like in a polar aprotic solvent, such as but not limited to THF, MeCN, DMF, DMAc, DMSO or NMP, to deliver compounds S47 (X4=Cl, Br, I) or compounds S48 (X4=CO2-Pg2) that are then used to obtain compounds S49 via methods described in Scheme 15. Alternatively, compounds of general structure S50 could be reacted with appropriately substituted and protected piperazines S8 to provide compounds S51 (Scheme 17). Removal of Pg1could be effected with many methods described in literature to provide amines S52. Conversion to compounds of general structure S47 (X4=Cl, Br or I) or S48 (X4=CO2-Pg2) may be accomplished by such manner as a Buchwald-Hartwig C—N coupling between compounds of the general structures S7 and as described previously in Scheme 2. Compounds of general structure S47 or S48 may then be used to obtain compounds of structure S49 via methods described in Scheme 15. In a manner similar to that described in Scheme 1 for the preparation of S4, compounds of the general structure S29 in which X=Y=CH, L=CH2, A=A2, ZA2=CH or CR2and ZA3=CH or CR2may also be prepared as described in Scheme 18a. Compounds of general structure S17 may be reacted with boronate esters S53 in the presence of a palladium catalyst and ligand complex in a manner similar to that described for the preparation of S3 from S1 and S2, to provide intermediate olefin S54. In this reaction, preferred X2substituents include Cl and Br. Reduction of the olefin will provide cyclohexyl derivatives S55, with the stereoselectivity of the reduction providing either the cis or the trans isomer, or a mixture thereof, depending on the conditions and the specific nature of the substituents. Reaction of S55 with alcohols S5 as described above will provide compounds of general structure S56. Removal of Pg2will provide compounds S29 (X=Y=CH, L=CH2, A=A2, ZA2=CH or CR2and ZA3=CH or CR2). Alternatively, compounds of the general structure S29 in which X=Y=CH, L=CH2may be prepared from substituted hetaryl halides of general structure S34 by reaction with S53 in the presence of a palladium catalyst and ligand complex (Scheme 18b). Reduction of the resulting olefin (S57) to give S58 followed by removal of Pg2will provide compounds S29 (X=Y=CH, L=CH2). Compounds S29 prepared in this manner may be converted into compounds of general structure S49 via the methods described above in Scheme 15. EXAMPLES The following illustrate the synthesis of various compounds of the present invention. Additional compounds within the scope of this invention may be prepared using the methods illustrated in these Examples, either alone or in combination with techniques generally known in the art. Experiments were generally carried out under inert atmosphere (nitrogen or argon), particularly in cases where oxygen- or moisture-sensitive reagents or intermediates were employed. Commercial solvents and reagents were generally used without further purification. Anhydrous solvents were employed where appropriate, generally AcroSeal® products from Acros Organics, Aldrich® Sure/Seal™ from Sigma-Aldrich, or DriSolv® products from EMD Chemicals. In other cases, commercial solvents were passed through columns packed with 4 Å molecular sieves, until the following QC standards for water were attained: a) <100 ppm for dichloromethane, toluene, N,N-dimethylformamide, and THF; b) <180 ppm for methanol, ethanol, 1,4-dioxane, and diisopropylamine. For very sensitive reactions, solvents were further treated with metallic sodium, calcium hydride, or molecular sieves, and distilled just prior to use. Products were generally dried under vacuum before being carried on to further reactions or submitted for biological testing. Mass spectrometry data is reported from either liquid chromatography-mass spectrometry (LCMS), atmospheric pressure chemical ionization (APCI) or gas chromatography-mass spectrometry (GCMS) instrumentation. The symbol ♦ denotes that the chlorine isotope pattern was observed in the mass spectrum. Chiral separations were used to separate enantiomers of some intermediates during the preparation of the compounds of the invention. When such separation was done, the separated enantiomers were designated as ENT-1 or ENT-2, according to their order of elution. For compounds with two chiral centers, the stereoisomers at each stereocenter were separated at different times. The designation of ENT-1 or ENT-2 of an intermediate or an example refers to the chiral center for the separation done at that step. It is recognized that when stereoisomers at a chiral center are separated in a compound with two or more centers, the separated enantiomers are diastereomers of each other. The ENT-1 or ENT-2 designation is used herein for consistency and refers to the separated chiral center. By way of example, but not limitation, Examples 35 and 36 have two chiral centers. The chiral center of the cyclopropyl moiety was separated when intermediate C77 was separated into ENT-1, giving intermediate C78, and ENT-2, giving intermediate C79. C78 was then used to prepare Example 35 that is identified by name as ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C78. In these preparations, after a mixture is subjected to separation procedures, the chiral center is identified with “abs” near that center, with the understanding that the separated enantiomers may not be enantiomerically pure. Typically, the enriched enantiomer at each chiral center is >90% of the isolated material. Preferrably, the enriched enantiomer at each center is >98% of the mixture. In some examples, the optical rotation of an enantiomer was measured using a polarimeter. According to its observed rotation data (or its specific rotation data), an enantiomer with a clockwise rotation was designated as the (+)-enantiomer and an enantiomer with a counter-clockwise rotation was designated as the (−)-enantiomer. Racemic compounds are indicated either by the absence of drawn or described stereochemistry, or by the presence of (+/−) adjacent to the structure; in this latter case, indicated stereochemistry represents the relative (rather than absolute) configuration of the compound's substituents. Reactions proceeding through detectable intermediates were generally followed by LCMS, and allowed to proceed to full conversion prior to addition of subsequent reagents. For syntheses referencing procedures in other Examples or Methods, reaction conditions (reaction time and temperature) may vary. In general, reactions were followed by thin-layer chromatography or mass spectrometry, and subjected to work-up when appropriate. Purifications may vary between experiments: in general, solvents and the solvent ratios used for eluents/gradients were chosen to provide appropriate Rfs or retention times. All starting materials in these Preparations and Examples are either commercially available or can be prepared by methods known in the art or as described herein. Preparation P1 (4-{2-[(4-Chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)acetic Acid (P1) Step 1. Synthesis of tert-butyl 2-fluoro-3′,6′-dihydro-3,4′-bipyridine-1′(2′H)-carboxylate (C1) A mixture of 3-bromo-2-fluoropyridine (14.5 g, 82.4 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-1(2H)-carboxylate (28.0 g, 90.6 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (3.80 g, 5.19 mmol), and potassium carbonate (25.0 g, 181 mmol) in 1,4-dioxane (200 mL) and water (50 mL) was stirred at 100° C. for 16 hours. The reaction mixture was then partitioned between saturated aqueous sodium chloride solution (200 mL) and EtOAc (300 mL); the organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 9% EtOAc in petroleum ether) afforded C1 as a yellow oil. Yield: 20.0 g, 71.9 mmol, 87%. Step 2. Synthesis of tert-butyl 4-(2-fluoropyridin-3-yl)piperidine-1-carboxylate (C2) A mixture of C1 (20.0 g, 71.9 mmol) and platinum(IV) oxide (1.97 g) in MeOH (500 mL) was hydrogenated at 15° C. for 16 hours, whereupon the reaction mixture was filtered. The filtrate was concentrated in vacuo, and the residue was subjected to silica gel chromatography (Gradient: 9% to 17% EtOAc in petroleum ether), affording C2 as a pale yellow oil. Yield: 20.0 g, 71.3 mmol, 99%. Step 3. Synthesis of tert-butyl 4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidine-1-carboxylate (C3) Sodium hydride (60% dispersion in mineral oil; 9.0 g, 220 mmol) was added portion-wise to a 0° C. mixture of C2 (20.0 g, 71.3 mmol) and (4-chloro-2-fluorophenyl)methanol (12.0 g, 74.7 mmol) in N,N-dimethylformamide (150 mL). The reaction mixture was then allowed to warm to 15° C. and stirred at 15° C. for 1 hour, whereupon it was cooled to 0° C. and slowly treated with saturated aqueous ammonium chloride solution (100 mL). The resulting mixture was extracted with EtOAc (300 mL), and the organic layer was washed with saturated aqueous sodium chloride solution (200 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Eluent: 20:1 petroleum ether/EtOAc) provided C3 as a white solid. Yield: 21.0 g, 49.9 mmol, 70%. Step 4. Synthesis of 2-[(4-chloro-2-fluorobenzyl)oxy]-3-(piperidin-4-yl)pyridine (C4) To a solution of C3 (14.0 g, 33.3 mmol) in dichloromethane (25 mL) was added trifluoroacetic acid (25 mL). After the reaction mixture had been stirred at 20° C. for 1.5 hours, it was concentrated in vacuo, affording C4 as a yellow oil. This material was taken directly into the following step. Step 5. Synthesis of ethyl (4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)acetate (C5) Potassium carbonate (30.0 g, 217 mmol) was added to a solution of C4 (from the previous step; ≤33.3 mmol) and ethyl bromoacetate (6.00 g, 35.9 mmol) in N,N-dimethylformamide (150 mL). The reaction mixture was stirred at 50° C. for 2 hours, whereupon it was filtered. The filtrate was diluted with EtOAc (250 mL), washed sequentially with water (250 mL) and with saturated aqueous sodium chloride solution (250 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 9% to 25% EtOAc in petroleum ether) provided C5 as a yellow oil. Yield: 9.80 g, 24.1 mmol, 72% over 2 steps. Step 6. Synthesis of (4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)acetic Acid (P1) To a solution of C5 (8.80 g, 21.6 mmol) in THF (45 mL) and MeOH (45 mL) was added aqueous sodium hydroxide solution (3 M; 90 mL, 270 mmol). After the reaction mixture had been stirred at 20° C. for 2 hours, its pH was adjusted to 4 by addition of 1 M hydrochloric acid. The resulting mixture was filtered, and the collected solid was washed three times with water, whereupon it was taken up in MeOH (20 mL) and water (60 mL). This mixture was concentrated in vacuo to remove organic solvents, and then lyophilized, affording P1 as a white solid. Yield: 5.90 g, 15.6 mmol, 72%. LCMS m/z 378.9♦ [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.03 (br d, 1H), 7.60 (br d, 1H), 7.55 (dd, 1H), 7.49 (br d, 1H), 7.32 (dd, 1H), 7.01 (dd, 1H), 5.41 (s, 2H), 3.26-3.17 (m, 4H), 2.90-2.79 (m, 1H), 2.66-2.55 (m, 2H), 1.85-1.73 (m, 4H). Preparation P2 Ammonium (4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)acetate (P2) Step 1. Synthesis of tert-butyl 4-(3-chloropyrazin-2-yl)-3,6-dihydropyridine-1(2H)-carboxylate (C6) A mixture of tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-1(2H)-carboxylate (20.0 g, 64.7 mmol), 2,3-dichloropyrazine (14.5 g, 97.3 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (2.60 g, 3.55 mmol), and sodium carbonate (15.1 g, 142 mmol) in 1,4-dioxane (120 mL) and water (50 mL) was stirred at 95° C. for 4 hours. The reaction mixture was then diluted with EtOAc (500 mL) and washed sequentially with water (100 mL) and with saturated aqueous sodium chloride solution (300 mL). The organic layer was dried over sodium sulfate, filtered, concentrated in vacuo, and subjected to silica gel chromatography (Gradient: 5% to 17% EtOAc in petroleum ether), affording C6 as a pale yellow oil. Yield: 15.7 g, 53.1 mmol, 82%. Step 2. Synthesis of tert-butyl 4-(3-chloropyrazin-2-yl)piperidine-1-carboxylate (C7) To a solution of C6 (12.6 g, 42.6 mmol) in MeOH (400 mL) was added tris(triphenylphosphine)rhodium(I) chloride (2.50 g, 2.70 mmol) and the reaction mixture was stirred for 2 hours at 30° C. under a balloon of hydrogen. After filtration, the filtrate was concentrated in vacuo, and the residue was combined with the product of a similar hydrogenation carried out using C6 (1.08 g, 3.65 mmol); silica gel chromatography (Gradient: 9% to 17% EtOAc in petroleum ether) provided C7 as a pale yellow solid. Combined yield: 12.3 g, 41.3 mmol, 89%. Step 3. Synthesis of tert-butyl 4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidine-1-carboxylate (C8) Palladium(II) acetate (271 mg, 1.21 mmol), 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphane) (BINAP; 1.51 g, 2.42 mmol), and cesium carbonate (13.1 mg, 40.3 mmol) were added to a solution of C7 (6.00 g, 20.1 mmol) and (4-chloro-2-fluorophenyl)methanol (3.56 g, 22.2 mmol) in 1,4-dioxane (60 mL). Nitrogen was bubbled through the suspension, after which the reaction mixture was stirred at 90° C. for 18 hours and filtered. The filtrate was concentrated in vacuo and purified using chromatography on silica gel (Gradient: 3% to 20% EtOAc in petroleum ether) to afford C8 as a yellow gum. Yield: 7.96 g, 18.9 mmol, 94%. Step 4. Synthesis of 2-[(4-chloro-2-fluorobenzyl)oxy]-3-(piperidin-4-yl)pyrazine (C9) To a solution of C8 (7.00 g, 16.6 mmol) in dichloromethane (80 mL) was added trifluoroacetic acid (20 mL). The reaction mixture was stirred at 15° C. for 1 hour, whereupon it was concentrated, and the residue was carefully treated with saturated aqueous sodium bicarbonate solution (100 mL). The resulting mixture was extracted with EtOAc (3×100 mL), and the combined organic layers were washed with saturated aqueous sodium chloride solution (300 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 10% to 25% MeOH in dichloromethane) provided C9 as a yellow solid. Yield: 5.2 g, 16 mmol, 96%. Step 5. Synthesis of ethyl (4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)acetate (C10) Ethyl bromoacetate (2.55 g, 15.3 mmol) and potassium carbonate (6.03 g, 43.6 mmol) were added to a solution of C9 (4.68 mg, 14.5 mmol) in N,N-dimethylformamide (50 mL). After the reaction mixture had been stirred at 15° C. for 18 hours, it was diluted with water (100 mL) and extracted with EtOAc (3×100 mL). The combined organic layers were washed sequentially with water (200 mL) and with saturated aqueous sodium chloride solution (200 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to provide C10 as a yellow gum. Yield: 4.75 g, 11.6 mmol, 80%. Step 6. Synthesis of Ammonium (4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)acetate (P2) To a solution of C10 (4.70 g, 11.5 mmol) in MeOH (40 mL) was added aqueous sodium hydroxide solution (3 M; 20 mL, 60 mmol). The reaction mixture was stirred at 15° C. for 2 hours, whereupon it was diluted with water (80 mL) and washed with EtOAc (3×80 mL). The aqueous layer was then adjusted to pH 6 via addition of 1 M hydrochloric acid, and concentrated in vacuo. The residue was treated with a mixture of dichloromethane and MeOH (10:1, 50 mL), stirred at 15° C. for 30 minutes, and filtered through diatomaceous earth. The filtrate was concentrated under reduced pressure and purified via reversed-phase HPLC (Column: Phenomenex Gemini C18, 10 μm; Mobile phase A: 10 mM ammonium bicarbonate in water; Mobile phase B: acetonitrile; Gradient: 10% to 33% B) to afford P2 as a white solid. Yield: 2.30 g, 5.80 mmol, 50%. LCMS m/z 380.1♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.13 (d, 1H), 8.06 (d, 1H), 7.55 (dd, 1H), 7.31-7.21 (m, 2H), 5.49 (s, 2H), 3.69 (br d, 2H), 3.61 (s, 2H), 3.43-3.33 (m, 1H), 3.25-3.11 (m, 2H), 2.25-2.06 (m, 4H). Preparation P3 (4-{2-[(4-Chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazin-1-yl)acetic Acid (P3) Step 1. Synthesis of 3-bromo-2-[(4-chloro-2-fluorobenzyl)oxy]pyridine (C11) (4-Chloro-2-fluorophenyl)methanol (2.21 g, 13.8 mmol) was added drop-wise via syringe to a 0° C. suspension of sodium hydride (60% dispersion in mineral oil; 0.80 g, 20.0 mmol) in THF (50 mL). The ice bath was then removed, and the reaction mixture was stirred at room temperature (14° C.) for 40 minutes, whereupon 3-bromo-2-fluoropyridine (2.00 g, 11.4 mmol) was added drop-wise via syringe. Stirring was continued at room temperature (14° C.) for 1 hour, whereupon the reaction mixture was poured into saturated aqueous ammonium chloride solution (20 mL) and extracted with EtOAc (3×20 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (50 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 5% dichloromethane in petroleum ether) afforded C11 as a white solid. Yield: 1.77 g, 49%. Step 2. Synthesis of tert-butyl 4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazine-1-carboxylate (C12) A suspension of C11 (1.00 g, 3.16 mmol), tert-butyl piperazine-1-carboxylate (650 mg, 3.49 mmol), 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphane) (236 mg, 0.379 mmol), tris(dibenzylideneacetone)dipalladium(0) (202 mg, 0.221 mmol), and cesium carbonate (2.06 g, 6.32 mmol) in 1,4-dioxane (10 mL) was degassed with nitrogen for 1 minute and then stirred at 85° C. for 16 hours. The reaction mixture was filtered, and the filtrate was concentrated in vacuo; purification of the residue using silica gel chromatography (Eluent: 20:1 petroleum ether/EtOAc) provided C12 as a pale yellow solid. Yield: 920 mg, 69%. LCMS m/z 422.1♦ [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.81 (dd, 1H), 7.46 (dd, 1H), 7.18-7.07 (m, 3H), 6.88 (dd, 1H), 5.47 (s, 2H), 3.61-3.52 (m, 4H), 3.07-2.97 (m, 4H), 1.48 (s, 9H). Step 3. Synthesis of 1-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazine (C13) Trifluoroacetic acid (1 mL) was added to a solution of C12 (160 mg, 0.379 mmol) in dichloromethane (2 mL). The reaction mixture was stirred at 20° C. for 30 minutes, whereupon it was concentrated in vacuo to afford C13 as a yellow oil, which was taken directly to the following step. Step 4. Synthesis of ethyl (4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazin-1-yl)acetate (C14) To a suspension of C13 (from the previous step; 50.379 mmol) and potassium carbonate (262 mg, 1.90 mmol) in N,N-dimethylformamide (1 mL) was added ethyl bromoacetate (82.3 mg, 0.493 mmol). After the reaction mixture had been stirred at 15° C. for 1.5 hours, it was diluted with EtOAc (50 mL) and washed with water (50 mL). The organic layer was then washed with saturated aqueous sodium chloride solution (50 mL), dried over magnesium sulfate, filtered, concentrated in vacuo, and purified via silica gel chromatography (Gradient: 17% to 50% EtOAc in petroleum ether), providing C14 as a yellow oil. Yield: 102 mg, 66% over 2 steps. LCMS m/z 408.0♦ [M+H]+. Step 5. Synthesis of (4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazin-1-yl)acetic Acid (P3) A solution of C14 (102 mg, 0.250 mmol) and aqueous sodium hydroxide solution (3 M; 0.3 mL, 0.9 mmol) in a mixture of MeOH (1 mL) and THF (1 mL) was stirred at 20° C. for 16 hours. The reaction mixture was then adjusted to pH 7 by addition of 1 M hydrochloric acid, and extracted with a mixture of dichloromethane and MeOH (10:1, 3×30 mL). The combined organic layers were dried over magnesium sulfate, filtered, and concentrated in vacuo to afford P3 as a yellow oil. Yield: 95.0 mg, 100%. Preparation P4 2-Chloro-3-[(4-chloro-2-fluorobenzyl)oxy]pyrazine (P4) To a 19° C. solution of (4-chloro-2-fluorophenyl)methanol (7.17 g, 44.65 mmol) and 2,3-dichloropyrazine (7.09 g, 47.59 mmol) in 1,4-dioxane (50 mL) was added sodium tert-butoxide (5.42 g, 56.40 mmol). The reaction mixture was stirred at 19° C. for 4 hours, whereupon it was poured into petroleum ether (150 mL) and then filtered through a pad of diatomaceous earth. Concentration of the filtrate in vacuo provided a residue, which was purified using silica gel chromatography (Gradient: 0% to 17% dichloromethane in petroleum ether); P4 was isolated as a white solid. Yield: 8.93 g, 73%. LCMS m/z 272.7 (dichloro isotope pattern observed) [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.03 (d, 1H), 7.98 (d, 1H), 7.48 (dd, 1H), 7.21-7.11 (m, 2H), 5.49 (s, 2H). Preparation P5 Methyl 6-azaspiro[2.5]octane-1-carboxylate (P5) Step 1. Synthesis of tert-butyl 4-(2-ethoxy-2-oxoethylidene)piperidine-1-carboxylate (C15) A solution of potassium tert-butoxide (65.9 g, 587 mmol) in THF (500 mL) was added to a 0° C. solution of ethyl (diethoxyphosphoryl)acetate (132 g, 589 mmol) in THF (500 mL), and the resulting suspension was stirred at 0° C. for 1 hour, whereupon it was cooled to −50° C. A solution of tert-butyl 4-oxopiperidine-1-carboxylate (90.0 g, 452 mmol) in THF (1.5 L) was added drop-wise at −50° C., and the reaction mixture was subsequently allowed to slowly warm to 20° C., and then to stir for 16 hours at 20° C. After addition of water (1 L), the mixture was concentrated in vacuo to remove THF. The aqueous residue was extracted with EtOAc (2×800 mL), and the combined organic layers were washed with saturated aqueous sodium chloride solution (500 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting material was washed several times with petroleum ether (200 mL) to provide C15 as a white solid. Yield: 95.0 g, 353 mmol, 78%.1H NMR (400 MHz, Chloroform-d) δ 5.71 (s, 1H), 4.16 (q, 2H), 3.55-3.43 (m, 4H), 2.94 (br t, 2H), 2.28 (br t, 2H), 1.47 (s, 9H), 1.28 (t, 3H). Step 2. Synthesis of 6-tert-butyl 1-ethyl 6-azaspiro[2.5]octane-1,6-dicarboxylate (C16) To a solution of trimethylsulfoxonium iodide (140 g, 636 mmol) in dimethyl sulfoxide (800 mL) was added potassium tert-butoxide (71.2 g, 634 mmol) in one portion at 20° C. After the reaction mixture had been stirred at 20° C. for 1.5 hours, a solution of C15 (95.0 g, 353 mmol) in dimethyl sulfoxide (800 mL) was added drop-wise, and stirring was continued at 20° C. for 16 hours. Saturated aqueous sodium chloride solution (2.0 L) was then added; the resulting mixture was neutralized by addition of ammonium chloride, and extracted with EtOAc (3.0 L). The combined organic layers were washed sequentially with water (2×1.0 L) and with saturated aqueous sodium chloride solution (2.0 L), dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via silica gel chromatography (Eluent: 10:1 petroleum ether/EtOAc) afforded C16 as a yellow oil.1H NMR analysis indicated that extraneous aliphatic material was present. Yield: 80 g, 280 mmol, 79%.1H NMR (400 MHz, Chloroform-d): δ 4.19-4.09 (m, 2H), 3.55-3.39 (m, 3H), 3.27 (ddd, 1H), 1.76-1.64 (m, 2H), 1.56 (dd, 1H, assumed; partially obscured by water peak), 1.47 (s, 9H), 1.47-1.37 (m, 2H), 1.27 (t, 3H), 1.17 (dd, 1H), 0.93 (dd, 1H). Step 3. Synthesis of 6-(tert-butoxycarbonyl)-6-azaspiro[2.5]octane-1-carboxylic Acid (C17) To a mixture of C16 (80 g, 280 mmol) in THF (500 mL) and water (500 mL) was added lithium hydroxide monohydrate (37.4 g, 891 mmol) in one portion. The reaction mixture was stirred at 25° C. for 16 hours, whereupon it was diluted with water (600 mL) and washed with EtOAc (3×300 mL). The organic layers were discarded, and the aqueous layer was acidified to pH 3 to 4 by addition of 6 M hydrochloric acid. The resulting mixture was extracted with EtOAc (3×600 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Trituration of the residue with petroleum ether (300 mL) provided C17 as a white solid. Yield: 42.0 g, 164 mmol, 59%. LCMS m/z 278.2 [M+Na+].1H NMR (400 MHz, DMSO-d6) δ 12.15-12.03 (br s, 1H), 3.43-3.25 (m, 3H, assumed; partially obscured by water peak), 3.23-3.12 (m, 1H), 1.64-1.50 (m, 2H), 1.52 (dd, 1H), 1.39 (s, 9H), 1.39-1.28 (m, 2H), 0.96-0.88 (m, 2H). Step 4. Synthesis of methyl 6-azaspiro[2.5]octane-1-carboxylate (P5) Thionyl chloride (5 mL) was added to a 15° C. solution of C17 (5.00 g, 19.6 mmol) in MeOH (50 mL), and the reaction mixture was stirred at 15° C. for 16 hours. It was then concentrated in vacuo, and the residue was poured into water (20 mL). The resulting mixture was adjusted to pH 9 by addition of aqueous sodium bicarbonate solution, whereupon it was extracted first with EtOAc (3×100 mL), and then with a mixture of dichloromethane and MeOH (10:1 ratio; 5×100 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford P5 as a pale brown solid. Yield: 3.0 g, 18 mmol, 92%.1H NMR (400 MHz, Chloroform-d) δ 3.68 (s, 3H), 2.92-2.88 (m, 2H), 2.88-2.82 (m, 1H), 2.74 (ddd, 1H), 1.76-1.62 (m, 2H), 1.51 (dd, 1H), 1.49-1.36 (m, 2H), 1.13 (dd, 1H), 0.90 (dd, 1H). Preparation P6 6-{6-[(4-Cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (P6) Step 1. Synthesis of 4-{[(6-chloropyridin-2-yl)oxy]methyl}-3-fluorobenzonitrile (C18) This reaction was carried out in two parallel batches. To a stirred suspension of potassium tert-butoxide (313 g, 2.79 mol) in THF (4.0 L) at 10° C. to 15° C. was added 3-fluoro-4-(hydroxymethyl)benzonitrile (281 g, 1.86 mol) in a portion-wise manner. The reaction mixture was stirred at 15° C. for 45 minutes, whereupon 2,6-dichloropyridine (230 g, 1.55 mol) was added in several portions, while maintaining the reaction temperature at 15° C. After a further 18 hours at 15° C., the reaction mixture was poured into saturated aqueous ammonium chloride solution (10 L). EtOAc (10 L) was added and the mixture was stirred for 15 minutes, then filtered through a pad of diatomaceous earth. The aqueous layer of the filtrate was extracted with EtOAc (2×6 L), and the combined organic layers were washed with saturated aqueous sodium chloride solution (5 L), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 10% to 15% EtOAc in petroleum ether) afforded C18 as a light yellow solid. Combined yield: 550 g, 2.09 mmol, 67%.1H NMR (400 MHz, CDCl3) δ 7.67 (dd, 1H), 7.58 (dd, 1H), 7.48 (dd, 1H), 7.40 (dd, 1H), 6.97 (d, 1H), 6.75 (d, 1H), 5.49 (s, 2H). Step 2. Synthesis of methyl 6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylate (C19) To a solution of C18 (3.00 g, 11.4 mmol) in toluene (80 mL) were added P5 (1.93 g, 11.4 mmol), tris(dibenzylideneacetone)dipalladium(0) (523 mg, 0.571 mmol), 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphane) (711 mg, 1.14 mmol), and cesium carbonate (11.2 g, 34.3 mmol). The reaction mixture was stirred at 100° C. for 16 hours, whereupon it was filtered through diatomaceous earth. The filtrate was concentrated in vacuo and purified using silica gel chromatography (Gradient: 0% to 15% EtOAc in petroleum ether) to provide C19 as a pale yellow oil. Yield: 2.30 g, 5.82 mmol, 51%. LCMS m/z 395.9 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.61 (dd, 1H), 7.44 (dd, 1H), 7.42 (dd, 1H), 7.36 (dd, 1H), 6.21 (d, 1H), 6.13 (d, 1H), 5.45 (s, 2H), 3.69 (s, 3H), 3.63-3.49 (m, 3H), 3.38 (ddd, 1H), 1.83-1.70 (m, 2H), 1.60 (dd, 1H), 1.51-1.45 (m, 2H), 1.21 (dd, 1H), 0.99 (dd, 1H). Step 3. Synthesis of 6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (P6) To a solution of C19 (770 mg, 1.95 mmol) in a mixture of THF (8.0 mL) and MeOH (8.0 mL) was added aqueous lithium hydroxide solution (2 M; 5.8 mL, 12 mmol), and the reaction mixture was stirred at 15° C. for 60 hours. It was then concentrated to remove organic solvents, and the aqueous residue was adjusted to pH 6 by addition of 1 M hydrochloric acid. The resulting mixture was extracted with dichloromethane (3×30 mL), and the combined organic layers were dried over magnesium sulfate, filtered, and concentrated in vacuo. Purification was carried out via chromatography on silica gel (Gradient: 0% to 1% MeOH in dichloromethane) followed by preparative thin-layer chromatography (Eluent: 20:1 dichloromethane/MeOH), affording P6 as a red gum. Yield: 530 mg, 1.39 mmol, 71%. LCMS m/z 382.1 [M+H]+. Preparation P7 Ammonium 6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]octane-1-carboxylate, ENT-2 (P7) Step 1. Synthesis of methyl 6-azaspiro[2.5]octane-1-carboxylate, Hydrochloride Salt (P5, HCl Salt) Thionyl chloride (8 mL) was added to a solution of C17 (12.4 g, 48.6 mmol) in MeOH (200 mL), and the reaction mixture was stirred at 30° C. for 16 hours. Concentration in vacuo afforded P5, HCl salt as a beige solid. Yield: 10.0 g, 48.6 mmol, quantitative.1H NMR (400 MHz, DMSO-d6) δ 9.29 (br s, 2H), 3.61 (s, 3H), 3.09-2.97 (m, 3H), 2.92-2.81 (m, 1H), 1.91-1.76 (m, 2H), 1.74 (dd, 1H), 1.73-1.57 (m, 2H), 1.06 (dd, 1H), 1.02 (dd, 1H). Step 2. Synthesis of methyl 6-(2-chloro-5-fluoropyrimidin-4-yl)-6-azaspiro[2.5]octane-1-carboxylate (C20) To a solution of P5, HCl salt (8.0 g, 39 mmol) and 2,4-dichloro-5-fluoropyrimidine (7.80 g, 46.7 mmol) in MeOH (150 mL) was added triethylamine (16.5 mL, 118 mmol). After the reaction mixture had been stirred at room temperature (30° C.) for 16 hours, it was concentrated in vacuo. The resulting gum was partitioned between EtOAc (80 mL) and water (80 mL); the aqueous layer was then further extracted with EtOAc (3×80 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (2×50 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography (Gradient: 0% to 21% EtOAc in petroleum ether) provided C20 as a yellow oil. Yield: 11.5 g, 38.4 mmol, 98%.1H NMR (400 MHz, CDCl3) δ 7.91 (d, 1H), 3.88-3.78 (m, 3H), 3.74-3.65 (m, 1H), 3.70 (s, 3H), 1.92-1.79 (m, 2H), 1.65 (dd, 1H, assumed; partially obscured by water peak), 1.59-1.54 (m, 2H), 1.25 (dd, 1H), 1.02 (dd, 1H). Step 3. Synthesis of methyl 6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]octane-1-carboxylate (C21) Palladium(II) acetate (888 mg, 3.96 mmol), di-tert-butyl[2′,4′,6′-tri(propan-2-yl)biphenyl-2-yl]phosphane (t-Bu XPhos; 3.27 g, 7.70 mmol), and cesium carbonate (31.6 g, 96.9 mmol) were added to a solution of C20 (11.5 g, 38.4 mmol) and (4-chloro-2-fluorophenyl)methanol (7.47 g, 46.5 mmol) in toluene (200 mL), whereupon the reaction vessel was evacuated and charged with nitrogen. This evacuation cycle was repeated twice, and the reaction mixture was then stirred at 110° C. for 16 hours. After filtration, the filtrate was concentrated in vacuo and purified via silica gel chromatography (Gradient: 0% to 22% EtOAc in petroleum ether) to afford C21 as a pale yellow oil. Yield: 13.4 g, 31.6 mmol, 82%. LCMS m/z 424.0♦ [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.87 (d, 1H), 7.45 (dd, 1H), 7.15-7.07 (m, 2H), 5.33 (s, 2H), 3.84-3.73 (m, 3H), 3.70 (s, 3H), 3.70-3.63 (m, 1H), 1.88-1.75 (m, 2H), 1.63 (dd, 1H), 1.56-1.50 (m, 2H), 1.23 (dd, 1H), 1.00 (dd, 1H). Step 4. Synthesis of 6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (C22) To a solution of C21 (21 g, 50 mmol) in a mixture of MeOH (80 mL) and THF (80 mL) was added aqueous sodium hydroxide solution (5 M; 30 mL, 150 mmol). The reaction mixture was stirred at 30° C. for 15 hours, whereupon it was concentrated in vacuo, adjusted to a pH of 6 to 7 by addition of 1 M hydrochloric acid, and extracted with a mixture of dichloromethane and MeOH (10:1, 4×100 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated under reduced pressure, and combined with the product of a similar reaction carried out using C21 (13.4 g, 31.6 mmol). Silica gel chromatography (Gradient: 0% to 10% MeOH in dichloromethane) provided C22 as a yellow gum. Combined yield: 27.0 g, 65.9 mmol, 81%. LCMS m/z 410.1♦ [M+H]+. Step 5. Isolation of ammonium 6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]octane-1-carboxylate, ENT-1 (C23) and ammonium 6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]octane-1-carboxylate, ENT-2 (P7) Separation of C22 (27.0 g, 65.9 mmol) into its component enantiomers was carried out via supercritical fluid chromatography [Column: Chiral Technologies Chiralpak AD, 10 μm; Mobile phase: 7:3 carbon dioxide/(MeOH containing 0.1% ammonium hydroxide)]. The first-eluting enantiomer, isolated as a white solid, was designated as ENT-1 (C23). Yield: 10.27 g, 25.06 mmol, 38%. LCMS m/z 410.0♦ [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.88 (d, 1H), 7.44 (dd, 1H), 7.12 (dd, 1H), 7.09 (dd, 1H), 5.33 (s, 2H), 3.89-3.69 (m, 4H), 1.86 (t, 2H), 1.63 (dd, 1H), 1.60-1.48 (m, 2H), 1.25 (dd, 1H), 1.05 (dd, 1H). The second-eluting enantiomer, designated as ENT-2 (P7), was further purified using reversed-phase HPLC (Column: Phenomenex Gemini C18, 10 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 15% to 38% B); this material was also obtained as a white solid. Yield: 9.86 g, 24.1 mmol, 37%. LCMS m/z 410.0♦ [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.88 (d, 1H), 7.45 (dd, 1H), 7.12 (dd, 1H), 7.09 (dd, 1H), 5.33 (s, 2H), 3.88-3.69 (m, 4H), 1.86 (t, 2H), 1.64 (dd, 1H), 1.61-1.49 (m, 2H), 1.26 (dd, 1H), 1.07 (dd, 1H). Preparation P8 4-Chloro-2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidine (P8) This reaction was carried out in two identical batches. A solution of (4-chloro-2-fluorophenyl)methanol (5.42 g, 33.8 mmol) in THF (60 mL) was added drop-wise, over a period of 25 minutes, to a −25° C. suspension of 4-chloro-2-(methylsulfonyl)pyrimidine (13.0 g, 67.5 mmol) and sodium hydride (60% dispersion in mineral oil; 2.43 g, 60.8 mmol) in THF (180 mL). The reaction mixture was allowed to stir at room temperature (25° C.) for 18 hours, whereupon the two batches were combined and concentrated in vacuo. The residue was diluted with saturated aqueous ammonium chloride solution (120 mL) and extracted with EtOAc (3×100 mL); the combined organic layers were dried over sodium sulfate, filtered, concentrated under reduced pressure, and purified via silica gel chromatography (Gradient: 0% to 6% EtOAc in petroleum ether), affording P8 as a white solid. Combined yield: 9.93 g, 36.4 mmol, 54%. LCMS m/z 272.7 (dichloro isotope pattern observed) [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.41 (d, 1H), 7.48 (dd, 1H), 7.17-7.10 (m, 2H), 7.02 (d, 1H), 5.46 (s, 2H). Preparation P9 6-{4-[(4-Chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (P9) Step 1. Synthesis of 2-chloro-4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidine (C24) Sodium hydride (60% dispersion in mineral oil; 264 mg, 6.59 mmol) was added in portions to a 0° C. solution of (4-chloro-2-fluorophenyl)methanol (1.06 g, 6.59 mmol) in THF (15 mL), and the resultant mixture was stirred at 10° C. for 30 minutes. A solution of 2,4-dichloro-5-fluoropyrimidine (1.00 mg, 5.99 mmol) in THF (5 mL) was then added portion-wise, and the reaction mixture was stirred at 10° C. for 3 hours, whereupon it was poured into aqueous ammonium chloride solution (100 mL) and extracted with EtOAc (2×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography (Gradient: 0% to 5% EtOAc in petroleum ether) provided C24 as a white solid. Yield: 1.21 g, 69%. Step 2. Synthesis of methyl 6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]octane-1-carboxylate (C25) A solution of C24 (1.10 g, 3.78 mmol), P5, HCl salt (855 mg, 4.16 mmol), and triethylamine (1.15 g, 11.3 mmol) in N,N-dimethylformamide (20 mL) was stirred at 100° C. for 6 hours, whereupon it was combined with a similar reaction carried out using C24 (100 mg, 0.344 mmol), poured into water (300 mL), and extracted with EtOAc (2×60 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo; purification via silica gel chromatography (Gradient: 0% to 10% EtOAc in petroleum ether) afforded C25 as an orange oil. Combined yield: 813 mg, 1.92 mmol, 47%. Step 3. Synthesis of 6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (P9) To a solution of C25 (1.79 g, 4.22 mmol) in MeOH (50 mL) was added aqueous sodium hydroxide solution (2 M; 21.1 mL, 42.2 mmol). The reaction mixture was stirred at 20° C. for 3 hours, then at 40° C. for 4 hours, whereupon it was acidified to pH 5 by addition of 12 M hydrochloric acid, diluted with water (300 mL), and extracted with dichloromethane (2×200 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Purification was carried out using silica gel chromatography (Gradient: 0% to 5% MeOH in dichloromethane) followed by reversed-phase HPLC (Column: Phenomenex Synergi C18, 30×150 mm, 4 μm; Mobile phase A: 0.225% formic acid in water; Mobile phase B: acetonitrile; Gradient: 50% to 80% B). The fractions from the HPLC separation were concentrated under reduced pressure to half of the original volume, and then extracted with dichloromethane (2×100 mL); the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo, providing P9 as a white foamy solid. Yield: 1.04 g, 60%. LCMS m/z 409.8♦ [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 7.97 (d, 1H), 7.42 (dd, 1H), 7.15 (br dd, 1H), 7.12 (dd, 1H), 5.43 (s, 2H), 3.86-3.74 (m, 3H), 3.73-3.64 (m, 1H), 1.79 (t, 2H), 1.63 (dd, 1H), 1.49 (t, 2H), 1.26 (dd, 1H), 1.07 (dd, 1H). Preparation P10 6-{6-[(4-Cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (P10) Step 1. Synthesis of 4-{[(3,6-difluoropyridin-2-yl)oxy]methyl}-3-fluorobenzonitrile (C26) To a solution of 2,3,6-trifluoropyridine (4.40 g, 33.1 mmol) and 3-fluoro-4-(hydroxymethyl)benzonitrile (5.00 g, 33.1 mmol) in 1-methylpyrrolidin-2-one (60 mL) was added potassium carbonate (13.7 g, 99.2 mmol). The reaction mixture was stirred at 100° C. for 16 hours, whereupon it was poured into water (100 mL) and extracted with EtOAc (3×300 mL). After the combined organic layers had been washed with saturated aqueous sodium chloride solution (4×200 mL), they were dried over sodium sulfate, filtered, concentrated in vacuo, and combined with the product of a similar reaction carried out using 2,3,6-trifluoropyridine (200 mg, 1.50 mmol). Silica gel chromatography (Gradient: 0% to 5% EtOAc in petroleum ether) provided C26 as a white solid. Combined yield: 6.93 g, 26.2 mmol, 76%. MS (ESI) m/z 265.1 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.69 (dd, 1H), 7.53-7.45 (m, 2H), 7.41 (dd, 1H), 6.50 (ddd, 1H), 5.52 (s, 2H). Step 2. Synthesis of methyl 6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylate (C27) This reaction was carried out in two identical batches. Triethylamine (766 mg, 7.57 mmol) was added to a solution of C26 (1.00 g, 3.78 mmol) and P5 (640 mg, 3.78 mmol) in dimethyl sulfoxide (9 mL), and the reaction mixture was stirred at 140° C. for 14 hours in a microwave reactor. The two reaction mixtures were then combined, poured into water (50 mL), and extracted with dichloromethane (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated in vacuo, and subjected to silica gel chromatography (Eluent: 20% EtOAc in petroleum ether) to afford C27 as a yellow oil. Combined yield: 696 mg, 1.68 mmol, 22%. LCMS m/z 413.9 [M+H]+.1H NMR (400 MHz, CDCl3): δ 7.65 (dd, 1H), 7.46 (dd, 1H), 7.37 (dd, 1H), 7.24 (dd, 1H), 6.12 (dd, 1H), 5.52 (s, 2H), 3.70 (s, 3H), 3.56-3.42 (m, 3H), 3.30 (ddd, 1H), 1.85-1.72 (m, 2H), 1.60 (dd, 1H), 1.53-1.46 (m, 2H), 1.21 (dd, 1H), 0.99 (dd, 1H). Step 3. Synthesis of 6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (P10) To a solution of C27 (646 mg, 1.56 mmol) in a mixture of THF (10 mL) and MeOH (1 mL) was added aqueous lithium hydroxide solution (2 M; 4.7 mL, 9.4 mmol). After the reaction mixture had been stirred at 25° C. for 16 hours, it was combined with a similar reaction carried out using C27 (50 mg, 0.12 mmol) and concentrated to dryness in vacuo. The residue was dissolved in water (5 mL) and adjusted to pH 6 via addition of 1 M hydrochloric acid; the precipitate was collected via filtration and washed with water (5 mL) to afford P10 as a yellow solid. Combined yield: 645 mg, 1.61 mmol, 96%. LCMS m/z 400.1 [M+H]+. Preparation P11 6-(6-Bromo-3-fluoropyridin-2-yl)-6-azaspiro[2.5]octane-1-carboxylic Acid (P11) Step 1. Synthesis of methyl 6-(6-bromo-3-fluoropyridin-2-yl)-6-azaspiro[2.5]octane-1-carboxylate (C28) Potassium carbonate (2.44 g, 17.7 mmol) was added to a solution of 2,6-dibromo-3-fluoropyridine (1.50 g, 5.88 mmol) and P5 (1.10 g, 6.50 mmol) in N,N-dimethylformamide (25 mL), and the resulting suspension was stirred at 100° C. for 16 hours. The reaction mixture was then combined with two similar reactions carried out using 2,6-dibromo-3-fluoropyridine (1.50 g, 5.88 mmol and 1.0 g, 3.9 mmol), and poured into water (300 mL). After extraction with tert-butyl methyl ether (2×200 mL), the combined organic layers were washed with saturated aqueous sodium chloride solution (200 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 10% EtOAc in petroleum ether), followed by reversed-phase HPLC (Column: Phenomenex Gemini C18, 10 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 60% to 88% B), afforded C28 as a white solid. Combined yield: 2.25 g, 6.56 mmol, 42%. LCMS m/z 344.7 (bromine isotope pattern observed) [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.06 (dd, 1H), 6.80 (dd, 1H), 3.69 (s, 3H), 3.63-3.50 (m, 3H), 3.40 (ddd, 1H), 1.91-1.78 (m, 2H), 1.64-1.50 (m, 3H, assumed; partially obscured by water peak), 1.22 (dd, 1H), 0.99 (dd, 1H). Step 2. Synthesis of 6-(6-bromo-3-fluoropyridin-2-yl)-6-azaspiro[2.5]octane-1-carboxylic Acid (P11) Aqueous sodium hydroxide solution (2 M; 6.56 mL, 13.1 mmol) was added to a solution of C28 (2.25 g, 6.56 mmol) in THF (35 mL) and MeOH (25 mL), and the reaction mixture was stirred at room temperature for 73 hours, whereupon it was concentrated under reduced pressure to two-thirds of the original volume, and acidified to pH 6 to 7 by careful addition of concentrated hydrochloric acid. The resulting mixture was diluted with saturated aqueous sodium chloride solution (100 mL) and extracted with dichloromethane (2×80 mL); the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo to provide P11 as an off-white solid. Yield: 2.15 g, 6.53 mmol, quantitative. LCMS m/z 330.7 (bromine isotope pattern observed) [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.06 (dd, 1H), 6.80 (dd, 1H), 3.62-3.44 (m, 4H), 1.90 (dd, 2H), 1.65-1.56 (m, 3H), 1.26 (dd, 1H), 1.07 (dd, 1H). Preparation P12 6-{6-[(4-Cyano-2-fluorobenzyl)oxy]-3,5-difluoropyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (P12) Step 1. Synthesis of 3-fluoro-4-{[(3,5,6-trifluoropyridin-2-yl)oxy]methyl}benzonitrile (C29) Reaction of 3-fluoro-4-(hydroxymethyl)benzonitrile with 2,3,5,6-tetrafluoropyridine was carried out using the method described for synthesis of C26 in Preparation P10. Compound C29 was obtained as a white solid. Yield: 1.15 g, 40%.1H NMR (400 MHz, Chloroform-d) δ 7.66 (dd, 1H), 7.49 (dd, 1H), 7.46-7.38 (m, 2H), 5.49 (s, 2H). Step 2. Synthesis of methyl 6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3,5-difluoropyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylate (C30) To a solution of C29 (522 mg, 1.85 mmol) in N,N-dimethylformamide (10 mL) were added P5 (348 mg, 2.06 mmol) and potassium carbonate (281 mg, 2.04 mmol). The reaction mixture was stirred at 110° C. for 10 hours, whereupon it was diluted with water (30 mL) and extracted with EtOAc (3×25 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (2×20 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 4% EtOAc in petroleum ether) afforded C30 as a light yellow gum. Yield: 400 mg, 50%. LCMS m/z 431.9 [M+H]+. Step 3. Synthesis of 6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3,5-difluoropyridin-2-yl}-6-azaspiro[2.5]octane-1-carboxylic Acid (P12) To a solution of C30 (540 mg, 1.25 mmol) in THF (16 mL) were added aqueous lithium hydroxide solution (2 M; 1.9 mL, 3.8 mmol) and MeOH (1.8 mL). After the reaction mixture had been stirred at 25° C. for 16 hours, aqueous lithium hydroxide solution (2 M; 1.9 mL, 3.8 mmol) was again added, and stirring was continued at 25° C. for 20 hours, whereupon the pH was adjusted to 5 to 6 by addition of 1 M hydrochloric acid, and the mixture was extracted with dichloromethane (2×25 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo to provide P12 (540 mg, assumed quantitative) as a white solid. This material was used in further chemistry without additional purification. LCMS m/z 417.9 [M+H]+. Preparation P13 2-[(4-Chloro-2-fluorobenzyl)oxy]-4-(piperidin-4-yl)pyrimidine, bis(p-toluenesulfonic Acid) Salt (P13) Step 1. Synthesis of 1-tert-butyl 4-ethyl 4-[2-(methylthio)pyrimidin-4-yl]piperidine-1,4-dicarboxylate (C31) A solution of lithium bis(trimethylsilyl)amide in THF (1 M; 2.3 L, 2.3 mol) was added to a −60° C. solution of 1-tert-butyl 4-ethyl piperidine-1,4-dicarboxylate (500 g, 1.93 mol) in THF (5 L). After the reaction mixture had stirred at −60° C. for 30 minutes, a solution of 4-chloro-2-(methylthio)pyrimidine (300 g, 1.87 mol) in THF (1.5 L) was added in a drop-wise manner. The reaction mixture was allowed to warm to room temperature over 1.5 hours and was then stirred at room temperature for 1 hour, before being cooled to 0° C. A solution of citric acid (386 g, 2.01 mol) in water (5 L) was then added, followed by saturated aqueous sodium chloride solution (5 L), and the resulting mixture was extracted with EtOAc (2×10 L). The combined organic layers were dried over sodium sulfate, filtered, concentrated in vacuo, and purified via silica gel chromatography (Gradient: 75% to 100% EtOAc in petroleum ether), affording C31 as a yellow oil. Yield: 690 g, 1.81 mol, 97%. LCMS m/z 382.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.61 (d, 1H), 7.25 (d, 1H), 4.13 (q, 2H), 3.69 (ddd, 2H), 3.16-2.96 (m, 2H), 2.48 (s, 3H), 2.27-2.18 (m, 2H), 1.96 (ddd, 2H), 1.39 (s, 9H), 1.13 (t, 3H). Step 2. Synthesis of tert-butyl 4-[2-(methylthio)pyrimidin-4-yl]piperidine-1-carboxylate (C32) A solution of C31 (690 g, 1.81 mol) in MeOH (4.6 L) and THF (2.3 L) was heated to 40° C., whereupon aqueous sodium hydroxide solution (2.0 M, 2 equivalents) was added. After the reaction mixture had been stirred at 40° C. for 8 hours, it was allowed to cool to room temperature and then adjusted to pH 4 by addition of 1 M aqueous citric acid solution. The resulting mixture was diluted with saturated aqueous sodium chloride solution (5 L) and extracted with EtOAc (3×5 L); the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo to provide C32, which was advanced directly into the following step. Yield: 550 g, 1.78 mol, 98%. LCMS m/z 310.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.52 (d, 1H), 7.12 (d, 1H), 4.10-3.98 (m, 2H), 2.90-2.74 (m, 2H), 2.80 (tt, 1H), 2.49 (s, 3H), 1.82 (br d, 2H), 1.60-1.47 (m, 2H), 1.41 (s, 9H). Step 3. Synthesis of tert-butyl 4-[2-(methylsulfonyl)pyrimidin-4-yl]piperidine-1-carboxylate (C33) 3-Chloroperoxybenzoic acid (80%; 765 g, 3.55 mol) was added to a 0° C. solution of C32 (550 g, 1.78 mol) in dichloromethane (14 L). The reaction mixture was allowed to warm to room temperature over 1.5 hours and was then stirred for an additional 15 hours before being filtered through a pad of diatomaceous earth. The filter pad was rinsed with dichloromethane (3×5 L), and the combined filtrates were washed sequentially with saturated aqueous sodium bicarbonate solution (2×1.1 L) and with saturated aqueous sodium chloride solution (1.5 L), dried over magnesium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Eluent: 50:1 dichloromethane/MeOH) afforded C33 as a yellow solid. Yield: 420 g, 1.23 mol, 69%. LCMS m/z 364.2 [M+Na+].1H NMR (400 MHz, CDCl3) δ 8.83 (d, 1H), 7.40 (d, 1H), 4.28 (br d, 2H), 3.37 (s, 3H), 2.99 (tt, 1H), 2.85 (ddd, 2H), 1.98 (br d, 2H), 1.81-1.67 (m, 2H), 1.48 (s, 9H). Step 4. Synthesis of tert-butyl 4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}piperidine-1-carboxylate (C34) To a solution of (4-chloro-2-fluorophenyl)methanol (201 g, 1.25 mol) in THF (12 L) was added a solution of sodium bis(trimethylsilyl)amide in THF (2 M; 1.2 equivalents). After the reaction mixture had been stirred for 15 minutes, a solution of C33 (410 g, 1.20 mol) in THF (1 L) was added, and stirring was continued at room temperature for 1 hour. Water (5 L) was then added, and the mixture was extracted with EtOAc (2×5 L); the combined organic layers were washed with saturated aqueous sodium chloride solution (5 L), dried over magnesium sulfate, filtered, and concentrated in vacuo. Purification via silica gel chromatography (Eluent: 5:1 petroleum ether/EtOAc) provided C34. Yield: 252 g, 597 mmol, 50%. LCMS m/z 422.1♦ [M+H]+.1H NMR (400 MHz, CDCl3) δ 8.43 (d, 1H), 7.48 (dd, 1H), 7.15-7.08 (m, 2H), 6.82 (d, 1H), 5.44 (s, 2H), 4.32-4.15 (m, 2H), 2.88-2.74 (m, 2H), 2.75 (tt, 1H), 1.88 (br d, 2H), 1.77-1.63 (m, 2H), 1.47 (s, 9H). Step 5. Synthesis of 2-[(4-chloro-2-fluorobenzyl)oxy]-4-(piperidin-4-yl)pyrimidine, bis(p-toluenesulfonic Acid) Salt (P13) p-Toluenesulfonic acid monohydrate (17.1 g, 89.9 mmol) was added in one portion to a solution of C34 (16.2 g, 38.4 mmol) in EtOAc (220 mL). The reaction mixture was heated to an internal temperature of 60° C. for 35 minutes, whereupon it was allowed to cool to room temperature overnight while stirring in the oil bath. LCMS analysis at this point indicated conversion to P13: LCMS m/z 322.2♦ [M+H]+. The solid was collected via filtration and washed with EtOAc (100 mL), affording P13 as a pink-white solid. Yield: 23.7 g, 35.6 mmol, 93%.1H NMR (400 MHz, DMSO-d6) δ 8.64-8.53 (br m, 1H), 8.56 (d, 1H), 8.37-8.23 (br m, 1H), 7.60 (dd, 1H), 7.53-7.48 (m, 1H), 7.49 (d, 4H), 7.34 (br d, 1H), 7.12 (d, 4H), 7.09 (d, 1H), 5.41 (s, 2H), 3.37 (br d, 2H), 3.08-2.93 (m, 3H), 2.29 (s, 6H), 2.01 (br d, 2H), 1.92-1.77 (m, 2H). Preparation P14 tert-Butyl (2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-2-methylpiperazine-1-carboxylate (P14) Step 1. Synthesis of tert-butyl (2S)-4-(2-chloro-5-fluoropyrimidin-4-yl)-2-methylpiperazine-1-carboxylate (C35) To a 0° C. solution of 2,4-dichloro-5-fluoropyrimidine (12.0 g, 71.9 mmol) in dichloromethane (130 mL) was added triethylamine (20 mL, 140 mmol), followed by a solution of tert-butyl (2S)-2-methylpiperazine-1-carboxylate (15.0 g, 74.9 mmol) in dichloromethane (70 mL). The reaction mixture was stirred at 30° C. for 15 hours, whereupon it was washed sequentially with water (150 mL) and with saturated aqueous sodium chloride solution (100 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo. Purification via chromatography on silica gel (Gradient: 20% to 50% EtOAc in petroleum ether) afforded C35 as a white solid. Yield: 21.4 g, 64.7 mmol, 90%. LCMS m/z 330.9♦ [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.94 (d, 1H), 4.54-4.45 (m, 1H), 4.39-4.29 (m, 1H), 4.28 (br d, 1H), 3.94 (br d, 1H), 3.35 (dd, 1H), 3.25-3.06 (m, 2H), 1.48 (s, 9H), 1.17 (d, 3H). Step 2. Synthesis of tert-butyl (2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-2-methylpiperazine-1-carboxylate (P14) Conversion of C35 to P14 was carried out via the method described for synthesis of C21 from C20 in Preparation P7. In this case, the crude product was purified using silica gel chromatography (Gradient: 0% to 20% EtOAc in petroleum ether) to afford P14 as a pale yellow oil. Yield: 10.7 g, 23.5 mmol, 98%.1H NMR (400 MHz, Chloroform-d) δ 7.88 (d, 1H), 7.44 (dd, 1H), 7.15-7.06 (m, 2H), 5.33 (s, 2H), 4.50-4.42 (m, 1H), 4.35-4.20 (m, 2H), 3.90 (br d, 1H), 3.28 (dd, 1H), 3.16 (ddd, 1H), 3.05 (ddd, 1H), 1.47 (s, 9H), 1.14 (d, 3H). Preparation P15 2-[(4-Chloro-2-fluorobenzyl)oxy]-4-[(3S)-3-methylpiperazin-1-yl]pyrimidine, bis(p-toluenesulfonate) Salt (P15) Step 1. Synthesis of tert-butyl (2S)-4-(2-chloropyrimidin-4-yl)-2-methylpiperazine-1-carboxylate (C36) A solution of tert-butyl (2S)-2-methylpiperazine-1-carboxylate (22.2 g, 111 mmol), 2,4-dichloropyrimidine (15.0 g, 101 mmol), and triethylamine (30 mL) in dichloromethane (200 mL) was stirred at 30° C. for 15 hours, whereupon it was washed sequentially with water (150 mL) and with saturated aqueous sodium chloride solution (100 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo. Chromatography on silica gel (Gradient: 20% to 50% EtOAc in petroleum ether) afforded C36 as a white solid. Yield: 25.0 g, 79.9 mmol, 79%. Step 2. Synthesis of tert-butyl (2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-2-methylpiperazine-1-carboxylate (C37) A solution of sodium bis(trimethylsilyl)amide in THF (1.0 M; 90.0 mL, 90.0 mmol) was added in a drop-wise manner to a solution of (4-chloro-2-fluorophenyl)methanol (13.3 g, 82.8 mmol) in THF (50 mL), and the resulting mixture was stirred at 60° C. for 15 minutes, whereupon it was added to a solution of C36 (20.0 g, 63.9 mmol) in THF (120 mL). Stirring was continued at 60° C. for 1 hour, and then the reaction mixture was partitioned between EtOAc and water, and combined with material from a similar reaction carried out using C36 (5.00 g, 16.0 mmol). The aqueous layer was extracted with EtOAc (2×120 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 30% EtOAc in petroleum ether) provided C37 as a yellow gum. Combined yield: 33.6 g, 76.9 mmol, 96%. Step 3. Synthesis of 2-[(4-chloro-2-fluorobenzyl)oxy]-4-[(3S)-3-methylpiperazin-1-yl]pyrimidine, bis(p-toluenesulfonate) Salt (P15) p-Toluenesulfonic acid monohydrate (17.8 g, 93.6 mmol) was added in one portion to a mixture of C37 (15.7 g, 35.9 mmol) and EtOAc (220 mL). The reaction mixture was heated to an internal temperature of 60° C. for 35 minutes, whereupon it was allowed to cool to room temperature in the oil bath while stirring overnight. The resulting solid was collected via filtration and washed with EtOAc (100 mL) to afford P15 as an off-white solid. Yield: 24.5 g, quantitative. LCMS m/z 337.2♦ [M+H]+.1H NMR (400 MHz, DMSO-d6), characteristic peaks: δ 9.21-9.06 (m, 1H), 8.89-8.73 (m, 1H), 8.22 (d, 1H), 7.62 (dd, 1H), 7.55 (dd, 1H), 7.48 (d, 4H), 7.37 (dd, 1H), 7.11 (d, 4H), 6.87 (d, 1H), 5.53 (s, 2H), 3.22 (dd, 1H), 3.18-3.05 (m, 1H), 2.29 (s, 6H), 1.27 (d, 3H). Preparation P16 Methyl 4-amino-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (P16) This entire sequence was carried out on large scale. In general, before reactions, as well as after addition of reagents, reactors were evacuated to −0.08 to −0.05 MPa and then filled with nitrogen to normal pressure. This process was generally repeated 3 times, and then oxygen content was assessed to ensure that it was ≤1.0%. For the processes of extraction and washing of organic layers, mixtures were generally stirred for 15 to 60 minutes and then allowed to settle for 15 to 60 minutes before separation of layers. Step 1. Synthesis of (2S)-2-[(benzyloxy)methyl]oxetane (C38) This reaction was carried out in three batches of approximately the same scale. A 2000 L glass-lined reactor was charged with 2-methylpropan-2-ol (774.7 kg). Potassium tert-butoxide (157.3 kg, 1402 mol) was added via a solid addition funnel, and the mixture was stirred for 30 minutes. Trimethylsulfoxonium iodide (308.2 kg, 1400 mol) was then added in the same manner, and the reaction mixture was heated at 55° C. to 65° C. for 2 to 3 hours, whereupon (2S)-2-[(benzyloxy)methyl]oxirane (92.1 kg, 561 mol) was added at a rate of 5 to 20 kg/hour. After the reaction mixture had been maintained at 55° C. to 65° C. for 25 hours, it was cooled to 25° C. to 35° C., and filtered through diatomaceous earth (18.4 kg). The filter cake was rinsed with tert-butyl methyl ether (3×340 kg), and the combined filtrates were transferred to a 5000 L reactor, treated with purified water (921 kg), and stirred for 15 to 30 minutes at 15° C. to 30° C. The organic layer was then washed twice using a solution of sodium chloride (230.4 kg) in purified water (920.5 kg), and concentrated under reduced pressure (≤−0.08 MPa) at ≤45° C. n-Heptane (187 kg) was added, and the resulting mixture was concentrated under reduced pressure (≤−0.08 MPa) at ≤45° C.; the organic phase was purified using silica gel chromatography (280 kg), with sodium chloride (18.5 kg) on top of the column. The crude material was loaded onto the column using n-heptane (513 kg), and then eluted with a mixture of n-heptane (688.7 kg) and EtOAc (64.4 kg). The three batches were combined, providing C38 as an 85% pure light yellow oil (189.7 kg, 906 mmol, 54%).1H NMR (400 MHz, Chloroform-d), C38 peaks only: δ 7.40-7.32 (m, 4H), 7.32-7.27 (m, 1H), 4.98 (dddd, 1H), 4.72-4.55 (m, 4H), 3.67 (dd, 1H), 3.62 (dd, 1H), 2.72-2.53 (m, 2H). Step 2. Synthesis of (2S)-oxetan-2-ylmethanol (C39) 10% Palladium on carbon (30.7 kg) was added through an addition funnel to a 10° C. to 30° C. solution of 85% pure C38 (from previous step; 185.3 kg, 884.8 mol) in THF (1270 kg) in a 3000 L stainless steel autoclave reactor. The addition funnel was rinsed with purified water and THF (143 kg), and the rinses were added to the reaction mixture. After the reactor contents had been purged with nitrogen, they were similarly purged with hydrogen, increasing the pressure to 0.3 to 0.5 MPa and then venting to 0.05 MPa. This hydrogen purge was repeated 5 times, whereupon the hydrogen pressure was increased to 0.3 to 0.4 MPa. The reaction mixture was then heated to 35° C. to 45° C. After 13 hours, during which the hydrogen pressure was maintained at 0.3 to 0.5 MPa, the mixture was vented to 0.05 MPa, and purged five times with nitrogen, via increasing the pressure to 0.15 to 0.2 MPa and then venting to 0.05 MPa. After the mixture had been cooled to 10° C. to 25° C., it was filtered, and the reactor was rinsed with THF (2×321 kg). The filter cake was soaked twice with this rinsing liquor and then filtered; concentration at reduced pressure (≤−0.06 MPa) was carried out at ≤40° C., affording C39 (62.2 kg, 706 mol, 80%) in THF (251 kg) Step 3. Synthesis of (2S)-oxetan-2-ylmethyl 4-methylbenzenesulfonate (C40) 4-(Dimethylamino)pyridine (17.5 kg, 143 mol) was added to a 10° C. to 25° C. solution of C39 (from the previous step; 62.2 kg, 706 mol) in THF (251 kg) and triethylamine (92.7 kg, 916 mol) in dichloromethane (1240 kg). After 30 minutes, p-toluenesulfonyl chloride (174.8 kg, 916.9 mol) was added in portions at intervals of 20 to 40 minutes, and the reaction mixture was stirred at 15° C. to 25° C. for 16 hours and 20 minutes. Purified water (190 kg) was added; after stirring, the organic layer was washed with aqueous sodium bicarbonate solution (prepared using 53.8 kg of sodium bicarbonate and 622 kg of purified water), and then washed with aqueous ammonium chloride solution (prepared using 230 kg of ammonium chloride and 624 kg of purified water). After a final wash with purified water (311 kg), the organic layer was filtered through a stainless steel Nutsche filter that had been preloaded with silica gel (60.2 kg). The filter cake was soaked with dichloromethane (311 kg) for 20 minutes, and then filtered; the combined filtrates were concentrated at reduced pressure (≤−0.05 MPa) and ≤40° C. until 330 to 400 L remained. THF (311 kg) was then added, at 15° C. to 30° C., and the mixture was concentrated in the same manner, to a final volume of 330 to 400 L. The THF addition and concentration was repeated, again to a volume of 330 to 400 L, affording a light yellow solution of C40 (167.6 kg, 692 mmol, 98%) in THF (251.8 kg)1H NMR (400 MHz, Chloroform-d), C40 peaks only: δ 7.81 (d, 2H), 7.34 (d, 2H), 4.91 (ddt, 1H), 4.62-4.55 (m, 1H), 4.53-4.45 (m, 1H), 4.14 (d, 2H), 2.75-2.63 (m, 1H), 2.60-2.49 (m, 1H), 2.44 (s, 3H). Step 4. Synthesis of (2S)-2-(azidomethyl)oxetane (C41) N,N-Dimethylformamide (473 kg), sodium azide (34.7 kg, 534 mol), and potassium iodide (5.2 kg, 31 mol) were combined in a 3000 L glass-lined reactor at 10° C. to 25° C. After addition of C40 (83.5 kg, 344.6 mol) in THF (125.4 kg), the reaction mixture was heated to 55° C. to 65° C. for 17 hours and 40 minutes, whereupon it was cooled to 25° C. to 35° C., and nitrogen was bubbled from the bottom valve for 15 minutes. tert-Butyl methyl ether (623 kg) and purified water (840 kg) were then added, and the resulting aqueous layer was extracted twice with tert-butyl methyl ether (312 kg and 294 kg). The combined organic layers were washed with purified water (2×419 kg) while maintaining the temperature at 10° C. to 25° C., affording C41 (31.2 kg, 276 mol, 80%) in a solution of the above organic layer (1236.8 kg). Step 5. Synthesis of 1-[(2S)-oxetan-2-yl]methanamine (C42) 10% Palladium on carbon (3.7 kg) was added through an addition funnel to a 10° C. to 30° C. solution of C41 (from the previous step; 1264 kg, 31.1 kg, 275 mol) in THF (328 kg) in a 3000 L stainless steel autoclave reactor. The addition funnel was rinsed with THF (32 kg), and the rinse was added to the reaction mixture. After the reactor contents had been purged with nitrogen, they were similarly purged with hydrogen, increasing the pressure to 0.05 to 0.15 MPa and then venting to 0.03 to 0.04 MPa. This hydrogen purge was repeated 5 times, whereupon the hydrogen pressure was increased to 0.05 to 0.07 MPa. The reaction temperature was increased to 25° C. to 33° C., and the hydrogen pressure was maintained at 0.05 to 0.15 MPa for 22 hours, while exchanging the hydrogen every 3 to 5 hours. The mixture was then purged five times with nitrogen, via increasing the pressure to 0.15 to 0.2 MPa and then venting to 0.05 MPa. After filtration, THF (92 kg and 93 kg) was used to wash the reactor and then soak the filter cake. The combined filtrates were concentrated at reduced pressure (≤−0.07 MPa) and ≤45° C., affording C42 (18.0 kg, 207 mol, 75%) in THF (57.8 kg)1H NMR (400 MHz, DMSO-d6), C42 peaks only: δ 4.62 (ddt, 1H), 4.49 (ddd, 1H), 4.37 (dt, 1H), 2.69 (d, 2H), 2.55-2.49 (m, 1H), 2.39 (m, 1H). Step 6. Synthesis of methyl 4-nitro-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (C43) Potassium carbonate (58.1 kg, 420 mol) was added to a solution of methyl 3-fluoro-4-nitrobenzoate (54.8 kg, 275 mol) in THF (148 kg) in a 100 L glass-lined reactor, and the mixture was stirred for 10 minutes. A solution of C42 (29.3 kg, 336 mol) in THF (212.9 kg) was added, and the reaction mixture was stirred at 20° C. to 30° C. for 12 hours, whereupon EtOAc (151 kg) was added, and the mixture was filtered through silica gel (29 kg). The filter cake was rinsed with EtOAc (150 kg and 151 kg), and the combined filtrates were concentrated at reduced pressure (≤−0.08 MPa) and ≤45° C. to a volume of 222 to 281 L. After the mixture had been cooled to 10° C. to 30° C., n-heptane (189 kg) was added, stirring was carried out for 20 minutes, and the mixture was concentrated at reduced pressure (≤−0.08 MPa) and ≤45° C. to a volume of 222 L. n-Heptane (181 kg) was again added into the mixture at a reference rate of 100 to 300 kg/hour, and stirring was continued for 20 minutes. The mixture was sampled until residual THF was ≤5% and residual EtOAc was 10% to 13%. The mixture was heated to 40° C. to 45° C. and stirred for 1 hour, whereupon it was cooled to 15° C. to 25° C. at a rate of 5° C. to 10° C. per hour, and then stirred at 15° C. to 25° C. for 1 hour. Filtration using a stainless steel centrifuge provided a filter cake, which was rinsed with a mixture of EtOAc (5.0 kg) and n-heptane (34 kg), and then stirred with THF (724 kg) at 10° C. to 30° C. for 15 minutes; filtration provided a yellow solid of C43 (57.3 kg, 210 mol, 76%).1H NMR (400 MHz, DMSO-d6) δ 8.34 (t, 1H), 8.14 (d, 1H), 7.63 (d, 1H), 7.13 (dd, 1H), 4.99 (dddd, 1H), 4.55 (ddd, 1H), 4.43 (dt, 1H), 3.87 (s, 3H), 3.67-3.61 (m, 2H), 2.67 (dddd, 1H), 2.57-2.47 (m, 1H). Step 7. Synthesis of methyl 4-amino-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (P16) To a solution of C43 (5.00 g, 18.8 mmol) in MeOH (150 mL) was added wet palladium on carbon (500 mg), and the mixture was stirred at 15° C. for 3 hours under a balloon of hydrogen. The reaction mixture was filtered; concentration of the filtrate in vacuo afforded P16 as a colorless oil. Yield: 4.40 g, 18.6 mmol, 99%.1H NMR (400 MHz, DMSO-d6) δ 7.16 (dd, 1H), 7.02 (d, 1H), 6.55 (d, 1H), 5.48 (s, 2H), 4.92-4.87 (m, 1H), 4.70 (t, 1H), 4.54 (ddd, 1H), 4.47 (ddd, 1H), 3.72 (s, 3H), 3.39-3.23 (m, 2H, assumed; partially obscured by water peak), 2.72-2.61 (m, 1H), 2.5-2.40 (m, 1H, assumed; partially obscured by solvent peak). Preparation P17 Methyl 2-(chloromethyl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (P17) A solution of C43 (from step 6 of Preparation P16; 51.8 kg, 190 mol) in THF (678 kg), in a 3000 L autoclave reactor, was treated with 10% palladium on carbon (5.2 kg) at 10° C. to 30° C. The addition pipe was rinsed with THF (46 kg) and the rinse was added to the reaction mixture. After the reactor contents had been purged with nitrogen, they were similarly purged with hydrogen, increasing the pressure to 0.1 to 0.2 MPa and then venting to 0.02 to 0.05 MPa. This hydrogen purge was repeated 5 times, whereupon the hydrogen pressure was increased to 0.1 to 0.25 MPa. The reaction mixture was stirred at 20° C. to 30° C., and every 2 to 3 hours, the mixture was purged with nitrogen three times, and then purged with hydrogen five times; after each final hydrogen exchange, the hydrogen pressure was increased to 0.1 to 0.25 MPa. After 11.25 hours total reaction time, the reaction mixture was vented to normal pressure, and purged five times with nitrogen, via increasing the pressure to 0.15 to 0.2 MPa and then venting to 0.05 MPa. It was then filtered, and the filter cake was rinsed twice with THF (64 kg and 63 kg); the combined rinse and filtrate were concentrated under reduced pressure (≤−0.08 MPa) and ≤40° C. to a volume of 128 to 160 L. THF (169 kg) was added, and the mixture was again concentrated to a volume of 128 to 160 L; this process was repeated a total of 4 times, affording a solution of the intermediate P16. THF (150 kg) was added to this solution, followed by 2-chloro-1,1,1-trimethoxyethane (35.1 kg, 227 mol) and p-toluenesulfonic acid monohydrate (1.8 kg, 9.5 mol). After the reaction mixture had been stirred for 25 minutes, it was heated at 40° C. to 45° C. for 5 hours, whereupon it was concentrated under reduced pressure to a volume of 135 to 181 L. 2-Propanol (142 kg) was added, and the mixture was again concentrated to a volume of 135 to 181 L, whereupon 2-propanol (36.5 kg) and purified water (90 kg) were added, and stirring was continued until a solution was obtained. This was filtered with an in-line liquid filter, and then treated with purified water (447 kg) at a reference rate of 150 to 400 kg/hour at 20° C. to 40° C. After the mixture had been cooled to 20° C. to 30° C., it was stirred for 2 hours, and the solid was collected via filtration with a centrifuge. The filter cake was rinsed with a solution of 2-propanol (20.5 kg) and purified water (154 kg); after drying, P17 was obtained as a white solid (32.1 kg, 109 mol, 57%).1H NMR (400 MHz, Chloroform-d) δ 8.14-8.11 (m, 1H), 8.01 (dd, 1H), 7.79 (br d, 1H), 5.26-5.18 (m, 1H), 5.04 (s, 2H), 4.66-4.58 (m, 2H), 4.53 (dd, 1H), 4.34 (dt, 1H), 3.96 (s, 3H), 2.82-2.71 (m, 1H), 2.48-2.37 (m, 1H). Preparation P18 Methyl 3-methyl-2-{[(2S)-2-methylpiperazin-1-yl]methyl}-3H-imidazo[4,5-b]pyridine-5-carboxylate (P18) Step 1. Synthesis of tert-butyl (3S)-4-(2-ethoxy-2-oxoethyl)-3-methylpiperazine-1-carboxylate (C44) To a solution of tert-butyl (3S)-3-methylpiperazine-1-carboxylate (1.20 g, 5.99 mmol) in N,N-dimethylformamide (15 mL) were added ethyl bromoacetate (1.20 g, 7.19 mmol) and potassium carbonate (2.48 g, 18.0 mmol). After the reaction mixture had been stirred at 50° C. for 2 hours, it was cooled to room temperature, diluted with EtOAc (25 mL), and washed with water (25 mL). The aqueous layer was extracted with EtOAc (2×30 mL), and the combined organic layers were washed with saturated aqueous sodium chloride solution (3×20 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via silica gel chromatography (Gradient: 0% to 10% MeOH in dichloromethane) afforded C44 as a yellow oil. Yield: 1.6 g, 5.6 mmol, 93%. Step 2. Synthesis of [(2S)-4-(tert-butoxycarbonyl)-2-methylpiperazin-1-yl]acetic Acid (C45) Aqueous sodium hydroxide solution (2 M; 5.34 mL, 10.7 mmol) and water (1 mL) were added to a solution of C44 (612 mg, 2.14 mmol) in MeOH (5 mL) and the reaction mixture was stirred at room temperature (10° C.) for 2 hours. After removal of organic solvent in vacuo, the residue was acidified to pH 7 by addition of 1 M hydrochloric acid, and the resulting mixture was concentrated under reduced pressure. The solid residue was then stirred with a mixture of dichloromethane and MeOH (10:1, 45 mL) at room temperature (10° C.) for 20 hours, whereupon it was filtered; the filtrate was concentrated in vacuo to provide C45 as a light yellow gum. Yield: 300 mg, 1.16 mmol, 54%. Step 3. Synthesis of tert-butyl (3S)-4-(2-{[6-chloro-2-(methylamino)pyridin-3-yl]amino}-2-oxoethyl)-3-methylpiperazine-1-carboxylate (C46) To a solution of C45 (270 mg, 1.04 mmol) and 2,4,6-tripropyl-1,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (50% solution in EtOAc; 1.27 g, 2.00 mmol) in N,N-dimethylformamide (3 mL) were added 6-chloro-N2-methylpyridine-2,3-diamine (182 mg, 1.15 mmol) and N,N-diisopropylethylamine (465 mg, 3.60 mmol). The reaction mixture was stirred at 25° C. for 16 hours, whereupon it was combined with a similar reaction carried out using C45 (25.5 mg, 98.8 μmol), diluted with water (20 mL), and extracted with EtOAc (3×20 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (2×20 mL), dried over sodium sulfate, filtered, concentrated in vacuo, and purified via silica gel chromatography (Gradient: 0% to 44% EtOAc in petroleum ether) to afford C46 as a black gum. Combined yield: 320 mg, 0.804 mmol, 70%. Step 4. Synthesis of tert-butyl (3S)-4-[(5-chloro-3-methyl-3H-imidazo[4,5-b]pyridin-2-yl)methyl]-3-methylpiperazine-1-carboxylate (C47) A mixture of C46 (320 mg, 0.804 mmol) in acetic acid (4 mL) was stirred at 100° C. for 16 hours, and then concentrated in vacuo. After the residue had been mixed with dichloromethane (10 mL), di-tert-butyl dicarbonate (287 mg, 1.32 mmol) and triethylamine (200 mg, 1.97 mmol) were added, and the reaction mixture was stirred at room temperature (15° C.) for 2 hours, whereupon it was washed with water (20 mL) and extracted with dichloromethane (3×20 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated in vacuo, and subjected to chromatography on silica gel (Gradient: 0% to 45% EtOAc in petroleum ether), providing C47 as a yellow gum. Yield: 180 mg, 0.474 mmol, 59%. Step 5. Synthesis of methyl 2-{[(2S)-4-(tert-butoxycarbonyl)-2-methylpiperazin-1-yl]methyl}-3-methyl-3H-imidazo[4,5-b]pyridine-5-carboxylate (C48) 1,3-Bis(diphenylphosphino)propane (43.0 mg, 0.104 mmol), palladium(II) acetate (12.7 mg, 56.6 μmol), and triethylamine (528 mg, 5.22 mmol) were added to a solution of C47 (180 mg, 0.474 mmol) in a mixture of MeOH (7 mL) and N,N-dimethylformamide, and the reaction mixture was stirred at 80° C., under carbon monoxide (50 psi), for 20 hours. It was then concentrated in vacuo and purified by chromatography on silica gel (Gradient: 0% to 78% EtOAc in petroleum ether), affording C48 as a yellow gum. Yield: 150 mg, 0.372 mmol, 78%. Step 6. Synthesis of methyl 3-methyl-2-{[(2S)-2-methylpiperazin-1-yl]methyl}-3H-imidazo[4,5-b]pyridine-5-carboxylate (P18) To a solution of C48 (100 mg, 0.248 mmol) in dichloromethane (4 mL) was added trifluoroacetic acid (1 mL), and the reaction mixture was stirred at room temperature (15° C.) for 2 hours. It was then concentrated in vacuo and subjected to strong cation exchange via solid-phase extraction (Agela Cleanert SCX column), affording P18 as a brown gum. Yield: 70 mg, 0.23 mmol, 93%. LCMS m/z 304.1 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.08 (AB quartet, 2H), 4.34 (d, 1H), 4.06 (s, 3H), 4.03 (s, 3H), 3.59 (d, 1H), 2.99-2.91 (m, 1H), 2.89-2.82 (m, 1H), 2.80-2.71 (m, 1H), 2.65-2.55 (m, 2H), 2.53-2.43 (m, 1H), 2.32-2.22 (m, 1H), 1.19 (d, 3H). Preparation P19 Methyl 2-(6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate (P19) Step 1. Synthesis of methyl 3-[(2-methoxyethyl)amino]-4-nitrobenzoate (C49) To a colorless solution of methyl 3-fluoro-4-nitrobenzoate (50 g, 250 mmol) in THF (400 mL) was added triethylamine (40.7 g, 402 mmol, 55.8 mL) followed by addition of 2-methoxyethanamine (30.2 g, 402 mmol) in THF (100 mL), drop-wise, at room temperature. The resultant yellow solution was stirred at 55° C. for 18 hours. The solution was cooled to room temperature and concentrated under reduced pressure to remove THF. The resultant yellow solid was dissolved in EtOAc (800 mL) and washed with saturated aqueous ammonium chloride solution (250 mL). The aqueous phase was separated and extracted with EtOAc (200 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (3×250 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to yield C49 (60.2 g, 94%) as a yellow solid.1H NMR (CDCl3) δ 8.23 (d, 1H), 8.17 (br s, 1H), 7.58 (d, 1H), 7.25 (dd, 1H), 3.95 (s, 3H), 3.69-3.73 (m, 2H), 3.56 (m, 2H), 3.45 (s, 3H); LCMS m/z 255.4 [M+H]+. Step 2. Synthesis of methyl 4-amino-3-[(2-methoxyethyl)amino]benzoate (C50) To solution of C49 (30 g, 118 mmol) in MeOH (500 mL) was added palladium on carbon (10 g, 94 mmol). This reaction was stirred at room temperature under 15 psi hydrogen for 18 hours. The black suspension was filtered through diatomaceous earth and the filter cake was washed with MeOH (500 mL). The combined filtrates were concentrated in vacuo to give C50 (26.5 g, quantitative) as a brown oil, which solidified on standing.1H NMR (400 MHz, CDCl3) δ 7.48 (dd, 1H), 7.36 (d, 1H), 6.69 (d, 1H), 3.87 (s, 3H), 3.77 (br s, 2H), 3.68 (t, 2H), 3.41 (s, 3H), 3.32 (t, 2H); LCMS m/z 224.7 [M+H]+. Step 3. Synthesis of tert-butyl 1-({4-(methoxycarbonyl)-2-[(2-methoxyethyl)amino]phenyl}carbamoyl)-6-azaspiro[2.5]octane-6-carboxylate (C51) To a room temperature (15° C.) solution of C17 (1.50 g, 5.88 mmol) and C50 (1.49 g, 6.64 mmol) in N,N-dimethylformamide (30 mL) was added O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HATU; 3.35 g, 8.81 mmol). The reaction mixture was stirred at 15° C. for 20 minutes, whereupon triethylamine (1.19 g, 11.8 mmol) was added, and the reaction mixture was stirred at 50° C. for 4 hours. It was then poured into water (160 mL) and extracted with EtOAc (3×100 mL); the combined organic layers were washed with saturated aqueous sodium chloride solution (3×100 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Purification using silica gel chromatography (Gradient: 0% to 5% MeOH in dichloromethane) afforded C51 as a yellow oil. Yield: 2.70 g, 5.85 mmol, 99%. Step 4. Synthesis of methyl 2-[6-(tert-butoxycarbonyl)-6-azaspiro[2.5]oct-1-yl]-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate (C52) A solution of C51 (2.70 g, 5.85 mmol) in acetic acid (25 mL) was stirred at 50° C. for 16 hours, whereupon it was carefully basified by addition of saturated aqueous potassium carbonate solution. The resulting mixture was extracted with EtOAc (2×100 mL), and the combined organic layers were dried over magnesium sulfate, filtered, and concentrated in vacuo. Chromatography on silica gel (Gradient: 50% to 100% EtOAc in petroleum ether) provided C52 as a yellow solid. Yield: 1.45 g, 3.27 mmol, 56%. Step 5. Synthesis of methyl 2-(6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate (P19) To a solution of C52 (550 mg, 1.24 mmol) in dichloromethane (10 mL) was added a solution of hydrogen chloride in EtOAc (4 M; 10 mL). The reaction mixture was stirred at 20° C. for 2 hours, whereupon it was concentrated under reduced pressure. The residue was treated with saturated aqueous potassium carbonate solution (10 mL) and extracted with dichloromethane (3×40 mL); the combined organic layers were dried over magnesium sulfate, filtered, and concentrated in vacuo to provide P19 as a yellow solid. Yield: 400 mg, 1.16 mmol, 94%.1H NMR (400 MHz, Chloroform-d) δ 8.06-8.02 (m, 1H), 7.93 (dd, 1H), 7.66 (d, 1H), 4.51 (ddd, component of ABXY pattern; 1H), 4.38 (ddd, component of ABXY pattern; 1H), 3.93 (s, 3H), 3.81-3.69 (m, 2H), 3.30 (s, 3H), 3.08-3.00 (m, 1H), 2.93 (ddd, 1H), 2.80-2.66 (m, 2H), 2.09 (dd, 1H), 1.88-1.77 (m, 1H), 1.67 (dd, 1H), 1.51-1.41 (m, 2H), 1.40-1.30 (m, 1H), 1.12 (dd, 1H). Preparation P20 Methyl 2-(chloromethyl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate (P20) Step 1. Synthesis of methyl 2-(chloromethyl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate (P20) To a solution of C50 (5.00 g, 22.3 mmol) in THF (100 mL) was added 2-chloro-1,1,1-trimethoxyethane (3.31 mL, 24.6 mmol), followed by p-toluenesulfonic acid monohydrate (84.8 mg, 0.446 mmol). The reaction mixture was heated at 45° C. for 5 hours, whereupon it was concentrated in vacuo; the residual oil was dissolved in EtOAc (10 mL) and heated until a solution formed. This was slowly stirred while cooling to room temperature overnight. The precipitate was collected via filtration and washed with heptane to afford P20 as a gray solid. Yield: 5.73 g, 20.3 mmol, 91%.1H NMR (600 MHz, Chloroform-d) δ 8.12 (br s, 1H), 8.01 (br d, 1H), 7.79 (d, 1H), 4.96 (s, 2H), 4.52 (t, 2H), 3.96 (s, 3H), 3.74 (t, 2H), 3.28 (s, 3H). Step 2. Synthesis of methyl 2-(chloromethyl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate, Hydrochloride Salt (P20, HCl Salt) A solution of C50 (5.0 g, 24 mmol) in 1,4-dioxane (100 mL) was heated to 100° C., a solution of chloroacetic anhydride (4.1 g, 24.5 mmol) in 1,4-dioxane (60 mL) was added via addition funnel over a period of 10 hours, and the reaction mixture was stirred for another 12 hours at 100° C. The following day, the reaction was cooled to room temperature and the 1,4-dioxane was removed under reduced pressure. The crude reaction mixture was dissolved in EtOAc and washed with saturated aqueous sodium bicarbonate solution. The EtOAc layer was separated, dried over sodium sulfate, and filtered. A solution of 4 M hydrogen chloride in 1,4-dioxane (1.1 equiv.) was added to the EtOAc solution of the product with constant stirring. The HCl salt of the desired product precipitated out as a pale yellow solid. The suspension was stirred for 1 hour and the product was then collected by filtration to obtain P20, HCl salt as a yellow solid (6.1 g, 86%).1H NMR (600 MHz, CD3OD) δ 8.64 (s, 1H), 8.30 (d, 1H), 7.92 (d, 1H), 5.32 (s, 2H), 4.84 (m, 2H), 3.99 (s, 3H), 3.83 (t, 2H), 3.31 (s, 3H). LCMS m/z 283.2 [M+H]+. Preparation P21 Methyl 1-(2-methoxyethyl)-2-(piperazin-1-ylmethyl)-1H-benzimidazole-6-carboxylate (P21) Step 1. Synthesis of methyl 2-{[4-(tert-butoxycarbonyl)piperazin-1-yl]methyl}-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate (C53) Compound P20 (1.59 g, 5.62 mmol) was added to a 15° C. mixture of tert-butyl piperazine-1-carboxylate (1.00 g, 5.37 mmol) and potassium carbonate (2.97 g, 21.5 mmol) in acetonitrile (15 mL), and the reaction mixture was stirred at 55° C. for 12 hours. It was then combined with a similar reaction carried out using P20 and tert-butyl piperazine-1-carboxylate (200 mg, 1.07 mmol), and the mixture was filtered. After the filtrate had been concentrated in vacuo, the residue was purified via chromatography on silica gel (Gradient: 0% to 60% EtOAc in petroleum ether) to provide C53 as a pale yellow solid. Combined yield: 2.30 g, 5.32 mmol, 83%. LCMS m/z 433.0 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.12 (d, 1H), 7.96 (dd, 1H), 7.73 (d, 1H), 4.58 (t, 2H), 3.95 (s, 3H), 3.89 (s, 2H), 3.73 (t, 2H), 3.46-3.37 (br m, 4H), 3.28 (s, 3H), 2.54-2.44 (br m, 4H), 1.45 (s, 9H). Step 2. Synthesis of methyl 1-(2-methoxyethyl)-2-(piperazin-1-ylmethyl)-1H-benzimidazole-6-carboxylate (P21) To a solution of C53 (2.30 g, 5.32 mmol) in dichloromethane (80 mL) was added a solution of hydrogen chloride in EtOAc (20 mL). The reaction mixture was stirred at 20° C. for 2 hours, whereupon it was concentrated in vacuo. The residue was diluted with water (20 mL), adjusted to a pH of 9 to 10 by addition of saturated aqueous sodium bicarbonate solution, and extracted with a mixture of EtOAc and MeOH (10:1, 15×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo to afford P21 as a pale yellow solid. Yield: 1.68 g, 5.05 mmol, 95%. LCMS m/z 332.8 [M+H]+.1H NMR (400 MHz, Chloroform-d) δ 8.13 (br s, 1H), 7.96 (br d, 1H), 7.72 (d, 1H), 4.59 (t, 2H), 3.95 (s, 3H), 3.86 (s, 2H), 3.75 (t, 2H), 3.29 (s, 3H), 2.87 (t, 4H), 2.50 (br m, 4H). Preparation P22 Methyl 2-(chloromethyl)-1-methyl-1H-benzimidazole-6-carboxylate (P22) Methyl 4-amino-3-(methylamino)benzoate (206 mg, 1.14 mmol) was dissolved in 1,4-dioxane (11.5 mL) and treated with chloroacetyl chloride (109 μL, 1.37 mmol). The mixture was stirred at 100° C. for 3 hours and cooled to room temperature. Triethylamine (0.8 mL, 7 mmol) and heptane (10 mL) were added and filtered. The filtrate was concentrated under reduced pressure and the crude product was purified by chromatography on silica gel (Eluent: 40% EtOAc in heptane) to afford 120 mg of P22 (44%).1H NMR (400 MHz, Chloroform-d) δ 8.14 (s, 1H), 8.01 (d, 1H), 7.78 (d, 1H), 4.87 (s, 2H), 3.97 (s, 3H), 3.94 (s, 3H); LCMS m/z 239.1 [M+H]+. Preparation P23 Methyl 4-amino-3-{[(1-ethyl-1H-imidazol-5-yl)methyl]amino}benzoate (P23) Step 1. Synthesis of methyl 3-{[(1-ethyl-1H-imidazol-5-yl)methyl]amino}-4-nitrobenzoate (C54) Triethylamine (3.65 mL, 26.2 mmol) was added to a solution of methyl 3-fluoro-4-nitrobenzoate (1.00 g, 5.02 mmol) and 1-(1-ethyl-1H-imidazol-5-yl)methanamine, dihydrochloride salt (1.00 g, 5.05 mmol) in a mixture of THF (12 mL) and MeOH (8 mL). The reaction mixture was stirred at 60° C. for 40 hours, whereupon it was concentrated in vacuo and purified using silica gel chromatography (Gradient: 0% to 2% MeOH in dichloromethane). Compound C54 was obtained as an orange solid. Yield: 1.27 g, 4.17 mmol, 83%.1H NMR (400 MHz, Chloroform-d) δ 8.24 (d, 1H), 7.98-7.91 (m, 1H), 7.68 (d, 1H), 7.57 (br s, 1H), 7.33 (dd, 1H), 7.11 (br s, 1H), 4.53 (d, 2H), 3.99 (q, 2H), 3.95 (s, 3H), 1.47 (t, 3H). Step 2. Synthesis of methyl 4-amino-3-{[(1-ethyl-1H-imidazol-5-yl)methyl]amino}benzoate (P23) A mixture of wet palladium on carbon (144 mg) and C54 (412 mg, 1.35 mmol) in MeOH (13 mL) was stirred under a balloon of hydrogen for 16 hours at 25° C. The reaction mixture was then filtered through a pad of diatomaceous earth and the filtrate was concentrated in vacuo to afford P23 as a gray solid. Yield: 340 mg, 1.24 mmol, 92%.1H NMR (400 MHz, Methanol-d4) δ 7.66 (br s, 1H), 7.38-7.29 (m, 2H), 6.97 (br s, 1H), 6.67 (d, 1H), 4.35 (s, 2H), 4.11 (q, 2H), 3.81 (s, 3H), 1.44 (t, 3H). Preparation P24 Methyl 2-(chloromethyl)-1-[(1-methyl-1H-imidazol-5-yl)methyl]-1H-benzimidazole-6-carboxylate, Hydrochloride Salt (P24) Step 1. Synthesis of methyl 3-{[(1-methyl-1H-imidazol-5-yl)methyl]amino}-4-nitrobenzoate (C55) To a colorless solution of methyl 3-fluoro-4-nitrobenzoate (1.0 g, 5.0 mmol) in N,N-dimethylformamide (10 mL) was added 1-(1-methyl-1H-imidazol-5-yl)methanamine (670 mg, 6.0 mmol) and triethylamine (762 mg, 7.53 mmol), slowly. The solution was stirred at 60° C. for 16 hours. The reaction mixture was poured into water (30 mL) and extracted with dichloromethane (3×30 mL). The combined organic extracts were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash chromatography (20% MeOH/dichloromethane). The obtained yellow solid was triturated with 30:1 petroleum ether/EtOAc to deliver C55 (1.2 g, 82%) as a yellow solid.1H NMR (CDCl3) δ 8.26 (d, 1H), 7.96 (br s, 1H), 7.71 (d, 1H), 7.50 (s, 1H), 7.35 (dd, 1H), 7.13 (s, 1H), 4.55 (d, 2H), 3.97 (s, 3H), 3.68 (s, 3H). Step 2. Synthesis of methyl 4-amino-3-{[(1-methyl-1H-imidazol-5-yl)methyl]amino}benzoate (C56) To a yellow suspension of C55 (5.46 g, 18.8 mmol) in MeOH (160 mL) was added wet 10% palladium on carbon (1 g). The mixture was stirred under 1 atmosphere of hydrogen for 36 hours at 20° C. The reaction mixture was filtered and the filter cake was rinsed with MeOH (200 mL). The filtrate was concentrated under reduced pressure to deliver C56 (4.8 g, 98%) as a brown solid.1H NMR (DMSO-d6) δ 7.56 (s, 1H), 7.18 (d, 1H), 7.13 (s, 1H), 6.87 (s, 1H), 6.55 (d, 1H), 5.50 (s, 2H), 4.84 (t, 1H), 4.23 (d, 2H), 3.73 (s, 3H), 3.63 (s, 3H). Step 3. Synthesis of methyl 2-(hydroxymethyl)-1-[(1-methyl-1H-imidazol-5-yl)methyl]-1H-benzimidazole-6-carboxylate (C57) A red mixture of C56 (780 mg, 3.00 mmol) and hydroxyacetic acid (342 mg, 4.49 mmol) in 1,3,5-trimethylbenzene (8 mL) was stirred at 140° C. under N2for 14 hours and at 25° C. for 48 hours. The clear yellow solution was decanted off to give a brown residue that was dissolved in MeOH (50 mL) and concentrated under reduced pressure. The crude product was purified by flash chromatography (20% MeOH/dichloromethane) to give C57 (318 mg, 35%) as a yellow foam.1H NMR (DMSO-d6) δ 8.13 (d, 1H), 7.83 (dd, 1H), 7.71 (d, 1H), 7.60 (s, 1H), 6.59 (s, 1H), 5.69 (s, 2H), 4.76 (s, 2H), 3.91 (s, 1H), 3.84 (s, 3H), 3.53 (s, 3H). Step 4. Synthesis of methyl 2-(chloromethyl)-1-[(1-methyl-1H-imidazol-5-yl)methyl]-1H-benzimidazole-6-carboxylate, Hydrochloride Salt (P24) To a yellow suspension of C57 (500 mg, 1.66 mmol) in dichloromethane (10 mL) and N,N-dimethylformamide (3 mL) was added SOCl2(990 mg, 0.60 mL, 8.32 mmol), drop-wise, at room temperature. The reaction mixture was stirred at room temperature for 1 hour, concentrated under reduced pressure, and the resultant brown residue was triturated with dichloromethane (10 mL). The solids were collected by filtration, rinsed with dichloromethane (5 mL), and dried under vacuum to give P24 (431 mg, 73%) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ 9.17 (s, 1H), 8.31 (s, 1H), 7.91-7.99 (m, 1H), 7.77-7.87 (m, 1H), 7.11 (s, 1H), 5.92 (s, 2H), 5.13 (s, 2H), 3.87 (s, 3H), 3.86 (s, 3H); MS (ES+): 319.0 (M+H). Preparation P25 tert-Butyl 4-nitro-3-[(1,3-oxazol-2-ylmethyl)amino]benzoate (P25) To a suspension of 1-(1,3-oxazol-2-yl)methanamine, hydrochloride salt (491 mg, 3.65 mmol) and tert-butyl 3-fluoro-4-nitrobenzoate (800 mg, 3.32 mmol) in N,N-dimethylformamide (5 mL) was added potassium carbonate (1.04 g, 6.63 mmol). The reaction was stirred at 60° C. for 2 hours. Additional 1-(1,3-oxazol-2-yl)methanamine, hydrochloride salt (100 mg, 1.0 mmol) was added and the reaction was stirred for an additional 30 minutes at 60° C. The reaction was cooled to room temperature then diluted with water (30 mL) and extracted with EtOAc (60 mL). The organic layer was washed with water, then saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered and concentrated under reduced pressure. The orange residue was purified by flash chromatography (12 g silica gel, 0-50% EtOAc/heptane gradient) to deliver P25 (764 mg, 75%) as an orange solid.1H NMR (CDCl3) δ 8.48 (br s, 1H), 8.23 (d, 1H), 7.68 (d, 1H), 7.61 (d, 1H), 7.28 (dd, 1H), 7.15 (s, 1H), 4.72 (d, 2H), 1.60 (s, 9H). Preparation P26 Methyl 5-amino-6-{[(2S)-oxetan-2-ylmethyl]amino}pyridine-2-carboxylate (P26) Step 1. Synthesis of methyl 5-nitro-6-{[(2S)-oxetan-2-ylmethyl]amino}pyridine-2-carboxylate (C58) Methyl 6-chloro-5-nitropyridine-2-carboxylate (270 g, 1.25 mol) and triethylamine (500 g, 5.1 mol) were added to a solution of C42 (152 g, 1.7 mol) in N,N-dimethylformamide (3 L) and THF (3 L) at 25° C. The mixture was stirred at 25° C. for 16 hours. The mixture was concentrated under reduced pressure to remove the THF, and water (5 L) was added. The mixture was extracted with EtOAc (2×5 L) and the combined organic solutions were washed with saturated aqueous sodium chloride solution (2×), dried and concentrated under reduced pressure. The crude material was combined with a second batch of crude product from a similar experiment (70 g) and the solids were triturated with petroleum ether:EtOAc (4:1, 500 mL) for 2 hours. The solids were collected by filtration and dried to provide C58 (304 g, 52%) as a yellow solid.1H NMR (400 MHz, CDCl3) δ 8.58 (br s, 1H), 8.56 (d, 1H), 7.39 (d, 1H), 5.08-5.18 (m, 1H), 4.73 (ddd, 1H), 4.61 (td, 1H), 4.06-4.16 (m, 1H), 3.98 (s, 3H), 3.88-3.97 (m, 1H), 2.68-2.80 (m, 1H), 2.55 (tdd, 1H). Step 2. Synthesis of methyl 5-amino-6-{[(2S)-oxetan-2-ylmethyl]amino}pyridine-2-carboxylate (P26) Compound C58 (10 g, 37 mmol) was suspended in MeOH (150 mL) and treated with 10% palladium on carbon (1.0 g) and the mixture was stirred at room temperature under 50 psi H2for 4 hours. The mixture was filtered through Celite® and the filtrate was concentrated under reduced pressure to yield P26 (8.4 g, 95%) as a yellow oil that solidified on standing.1H NMR (600 MHz, CDCl3) δ 7.49 (d, 1H), 6.86 (d, 1H), 5.06-5.15 (m, 1H), 4.68-4.77 (m, 1H), 4.53-4.63 (m, 2H), 3.91 (s, 3H), 3.80-3.86 (m, 2H), 3.72 (br s, 2H), 2.68-2.78 (m, 1H), 2.52-2.61 (m, 1H). Preparation P27 Methyl 2-(chloromethyl)-3-[(2S)-oxetan-2-ylmethyl]-3H-imidazo[4,5-b]pyridine-5-carboxylate (P27) In a 2 L, 3-neck flask equipped with a mechanical overhead stirrer P26 (43.0 g, 181 mmol) was taken up in THF (780 mL). The resultant pale pink suspension was treated with a solution of chloroacetic anhydride (33.5 g, 190 mmol in 100 mL THF) via addition funnel over 30 minutes. The resultant light amber solution was stirred at room temperature for 2 hours and then heated at 60° C. for 7 hours. The reaction mixture was cooled to room temperature. Approximately 400 mL of solvent from the reaction was removed under reduced pressure on a rotary evaporator. The resulting solution was diluted with EtOAc (500 mL) and treated with saturated aqueous sodium bicarbonate solution (200 mL). The biphasic mixture was stirred at room temperature for 30 minutes. The organic layer was separated and the aqueous layer was extracted with EtOAc (500 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (500 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to yield P27 (52.5 g, 98%) as a yellowish brown solid.1H NMR (600 MHz, CDCl3) δ 8.14 (d, 2H), 5.19-5.28 (m, 1H), 4.99-5.16 (m, 2H), 4.70-4.88 (m, 2H), 4.55-4.67 (m, 1H), 4.24-4.44 (m, 1H), 4.01 (s, 3H), 2.70-2.88 (m, 1H), 2.37-2.53 (m, 1H); LC-MS (ES+): 296.4 (M+H). Preparation P28 5-Chloro-2-(chloromethyl)-3-methyl-3H-imidazo[4,5-b]pyridine (P28) Step 1. Synthesis of 6-chloro-N-methyl-3-nitropyridin-2-amine (C59) To a suspension of 2,6-dichloro-3-nitropyridine (200 g, 1.04 mol) and sodium carbonate (132 g, 1.24 mol) in ethanol (1 L) was added 2.0 M methylamine in THF (622 mL, 1.24 mol), drop-wise, at 0° C. via syringe. After the addition, the reaction mixture was stirred at 18° C. for 6 hours. The yellow mixture was filtered and the filtrate was concentrated under reduced pressure to give a yellow solid. The crude product was purified by flash chromatography (petroleum ether/EtOAc 0-5%) to afford C59 (158 g, 81% yield) as a yellow solid.1H NMR (DMSO-d6) δ 8.72 (br s, 1H), 8.41 (d, 1H), 6.76 (d, 1H), 3.00 (d, 3H). Step 2. Synthesis of 6-chloro-N2-methylpyridine-2,3-diamine (C60) To a mixture of C59 (15.8 g, 84.2 mmol) in acetic acid (100 mL) was added iron powder (15.4 g, 276 mmol). The yellow mixture was stirred at 80° C. for 3 hours. The reaction was cooled to room temperature and filtered. The filter cake was washed with EtOAc (2×100). The combined organic layers were concentrated under reduced pressure and the crude product was purified by flash chromatography (120 g silica gel, 50% EtOAc/petroleum ether) to afford C60 (8.40 g, 63% yield) as a brown solid.1H NMR (CDCl3) δ 6.80 (d, 1H), 6.50 (d, 1H), 3.39 (br s, 2H), 3.01 (s, 3H). Step 3. Synthesis of 5-chloro-2-(chloromethyl)-3-methyl-3H-imidazo[4,5-b]pyridine (P28) To a solution of C60 (50.0 g, 317 mmol) in 1,4-dioxane (1.2 L) was added chloroacetyl chloride (55.5 mL, 698 mmol) and the mixture was stirred at 15° C. for 50 minutes. The brown mixture was concentrated under reduced pressure to give a brown solid which was taken up in trifluoroacetic acid (1.2 L) and stirred at 80° C. for 60 hours. The mixture was concentrated under reduced pressure to give a brown oil. The oil was diluted with EtOAc (1 L) and neutralized with saturated aqueous sodium bicarbonate solution. When CO2evolution subsided, the layers were separated and the aqueous layer was extracted with EtOAc (200 mL). The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography (10-25% EtOAc/petroleum ether gradient) to afford P28 (61.0 g, 79%) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ 8.13 (d, 1H), 7.37 (d, 1H), 5.11 (s, 2H), 3.84 (s, 3H). Preparation P29 5-Bromo-N3,6-dimethylpyridine-2,3-diamine (P29) Compound P29 was synthesized according to the literature procedure (Choi, J. Y. et al.,J. Med. Chem.2012, 55, 852-870). Preparation P30 Methyl 2-(6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-imidazo[4,5-b]pyridine-6-carboxylate, Hydrochloride Salt (P30) Step 1. Synthesis of tert-butyl 1-(6-bromo-1H-imidazo[4,5-b]pyridin-2-yl)-6-azaspiro[2.5]octane-6-carboxylate (C61) A mixture of C17 (1.00 g, 3.92 mmol), O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (2.98 g, 7.83 mmol), 5-bromopyridine-2,3-diamine (1.10 g, 5.85 mmol), and triethylamine (1.64 mL, 11.8 mmol) was dissolved in N,N-dimethylformamide, and the reaction mixture was stirred at room temperature overnight, whereupon it was diluted with water and extracted with EtOAc. The organic layer was concentrated in vacuo, and purified using silica gel chromatography. The resulting amide was dissolved in 1,4-dioxane (8 mL), treated with aqueous potassium hydroxide solution (4 M; 2 mL, 8 mmol), and stirred for 15 hours at 100° C. After a standard work-up, chromatography on silica gel (Gradient: EtOAc in hydrocarbon solvent), both C61 (255 mg) and uncyclized amide (255 mg) were obtained. The amide was resubjected to the potassium hydroxide reaction conditions, affording additional C61 (250 mg) as a solid. Combined yield: 505 mg, 1.24 mmol, 32%. Step 2. Synthesis of tert-butyl 1-[6-bromo-1-(2-methoxyethyl)-1H-imidazo[4,5-b]pyridin-2-yl]-6-azaspiro[2.5]octane-6-carboxylate (C62) A solution of C61 (225 mg, 0.552 mmol) and potassium tert-butoxide (136 mg, 1.21 mmol) in THF was stirred at room temperature for 20 minutes, whereupon 2-bromoethyl methyl ether (0.125 mL, 1.33 mmol) was added, and the reaction mixture was heated at 60° C. for 15 hours. LCMS analysis indicated formation of two isomers of the product. Silica gel chromatography provided purified C62 (120 mg) as a gum, and C63 (69 mg). The indicated regiochemistries for C62 and C63 were determined using nuclear Overhauser effect experiments. Yield of C62: 120 mg, 0.258 mmol, 47%. LCMS m/z 465.3 (bromine isotope pattern observed) [M+H]+.1H NMR (400 MHz, Chloroform-d), characteristic peaks: δ 8.40 (s, 1H), 7.70 (s, 1H), 4.40-4.20 (m, 2H), 3.75-3.58 (m, 3H), 3.36-3.25 (m, 2H), 3.21 (s, 3H), 3.14-3.08 (m, 1H), 1.98 (dd, 1H), 1.74 (dd, 1H), 1.70-1.63 (m, 1H), 1.48-1.42 (m, 2H), 1.36 (s, 9H), 1.11 (dd, 1H). Step 3. Synthesis of methyl 2-(6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-imidazo[4,5-b]pyridine-6-carboxylate, Hydrochloride Salt (P30) A mixture of C62 (120 mg, 0.258 mmol), 1,3-bis(diphenylphosphino)propane (31.9 mg, 77.4 μmol), and palladium(II) acetate (11.6 mg, 51.7 μmol) in N,N-dimethylformamide (0.5 mL) was treated with MeOH (4 mL) and triethylamine (0.36 mL, 2.6 mmol), and then heated at 80° C. under carbon monoxide (50 psi) for 20 hours. After a standard work-up, purification via silica gel chromatography (Eluent: 10:1 dichloromethane/MeOH) provided material that was then deprotected using a solution of hydrogen chloride in 1,4-dioxane to provide P30 as a solid. Yield: 40 mg, 0.105 mmol, 41%. LCMS m/z 345.3 [M+H]+. Preparation P31 5-Bromo-N3-(2-methoxyethyl)pyridine-2,3-diamine (P31) Step 1. Synthesis of N-(2-amino-5-bromopyridin-3-yl)-2-methoxyacetamide (C64) To a flask containing a solution of methoxyacetic acid (1.00 g, 11.1 mmol) in N,N-dimethylformamide (30 mL) was added O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (6.33 g, 16.7 mmol) and triethylamine (3.37 g, 33.3 mmol). After stirring for 20 minutes, 5-bromopyridine-2,3-diamine (2.3 g, 12 mmol) was added portion-wise, and the resulting reaction mixture was stirred overnight. After 15 hours, water was added, and the solution was extracted with EtOAc. The combined organic layers were dried, and the solvent was removed under reduced pressure. The crude compound was purified by flash chromatography (Gradient: 0% to 80% EtOAc in heptane) to yield C64 (2.3 g, 80%).1H NMR (400 MHz, CDCl3) δ 8.09 (s, 1H), 8.06 (d, 1H), 8.03 (s, 1H), 7.83 (d, 1H), 4.08 (s, 2H), 3.53 (s, 3H); LC-MS (ES+): 260.2 (M+H). Step 2. Synthesis of 5-bromo-N3-(2-methoxyethyl)pyridine-2,3-diamine (P31) To a solution of C64 (3.3 g, 13 mmol) in THF was added a solution of BH3in THF (1 M; 14 mL, 14 mmol) over the period of 10 minutes, and stirred at room temperature overnight, Water was added to the reaction slowly to quench the excess borane, and the mixture was then extracted with EtOAc. The EtOAc layer was dried and concentrated under reduced pressure. The crude product was dissolved in MeOH and HCl in 1,4-dioxane (1.0 equiv) was added and stirred for 2 hours. Excess MeOH was removed under reduced pressure to obtain the crude product. The compound was purified by flash chromatography with a gradient ranging from 0% to 70% EtOAc in heptane to obtain P31 as a brown oil (1.1 g, 35%).1H NMR (600 MHz, CDCl3) δ 7.83 (d, 1H), 6.95 (d, 1H), 5.56 (s, 2H), 3.77 (t, 1H), 3.66 (t, 2H), 3.42 (s, 3H), 3.22 (q, 2H); LC-MS (ES+): 246.1. Preparation P32 6-Bromo-2-(chloromethyl)-1-(2-methoxyethyl)-1H-imidazo[4,5-b]pyridine (P32) Compound P31 (400 mg, 1.63 mmol) was taken up in 1,4-dioxane (8 mL) and treated with chloroacetyl chloride (0.284 mL, 3.58 mmol). The mixture was stirred at room temperature. The solvent was removed under reduced pressure and the resultant residue was taken up in trifluoroacetic acid (8 mL) and stirred at 80° C. for 18 hours. The reaction was cooled to room temperature and concentrated under reduced pressure. The resultant brown oil was taken up in EtOAc (50 mL) and neutralized with saturated aqueous sodium bicarbonate solution. After the carbon dioxide evolution had subsided, the layers were separated and the aqueous layer was extracted with additional EtOAc (20 mL). The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure. The resultant crude product was purified by flash chromatography (Gradient: 0% to 80% EtOAc in heptane) to yield P32 (176 mg, 36%) as a tan solid.1H NMR (600 MHz, CDCl3) δ 8.59 (s, 1H), 7.90 (s, 1H), 4.93 (s, 2H), 4.45 (m, 2H), 3.72 (m, 2H), 3.29 (s, 3H); LC-MS (ES+): 306.1 (M+H). Preparation P33 6-Bromo-N4-(2-methoxyethyl)pyridine-3,4-diamine (P33) Step 1. Synthesis of 2-bromo-N-(2-methoxyethyl)-5-nitropyridin-4-amine (C65) A solution of 2,4-dibromo-5-nitropyridine (8.65 g, 30.7 mmol) in THF (100 mL) was treated with triethylamine (5.11 mL, 36.8 mmol) and 2-methoxyethanamine (2.77 g, 36.8 mmol), and the reaction mixture was stirred at 20° C. for 1 hour. It was then poured into water (200 mL) and extracted with EtOAc (2×200 mL); the combined organic layers were dried over sodium sulfate, filtered, concentrated in vacuo, and combined with a similar reaction carried out using 2,4-dibromo-5-nitropyridine (600 mg, 2.13 mmol). Purification using silica gel chromatography (Gradient: 0% to 35% EtOAc in petroleum ether) afforded C65 as a yellow solid. Combined yield: 9.02 g, 32.7 mmol, 99%. Step 2. Synthesis of 6-bromo-N4-(2-methoxyethyl)pyridine-3,4-diamine (P33) Reduced iron powder (607 mg, 10.9 mmol) and ammonium chloride (3.49 g, 65.2 mmol) were added to a solution of C65 (1.00 g, 3.62 mmol) in a mixture of MeOH (10 mL) and water (4 mL). The reaction mixture was stirred at 80° C. for 4 hours, whereupon it was poured into water (30 mL) and extracted with EtOAc (4×50 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (50 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to provide P33 as a pale brown solid. Yield: 900 mg, assumed quantitative.1H NMR (400 MHz, Chloroform-d) δ 7.63 (s, 1H), 6.59 (s, 1H), 4.58 (br s, 1H), 3.63 (t, 2H), 3.40 (s, 3H), 3.34-3.26 (m, 2H). Preparation P34 (4-{6-[(4-chloro-2-fluorobenzyl)oxy]pyridin-2-yl}cyclohexyl)acetic Acid (P34) Compound P34 was prepared as a mixture of cis and trans isomers by a route analogous to that employed in the preparation of P2 beginning from 2,6-dichloropyridine and methyl [4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-en-1-yl]acetate, via Suzuki coupling, chemoselective reduction, palladium-catalyzed etherification and ester hydrolysis. LCMS m/z 378.1 [M+H]+. Example 1 Ammonium 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (1) Step 1. Synthesis of methyl 4-{[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)acetyl]amino}-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (C66) To a solution of P1 (100 mg, 0.264 mmol) in N,N-dimethylformamide (5 mL) was added O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (151 mg, 0.396 mmol). After this mixture had been stirred at room temperature (10° C.) for 10 minutes, P16 (62.4 mg, 0.264 mmol) and N,N-diisopropylethylamine (102 mg, 0.792 mmol) were added, and the reaction mixture was stirred at room temperature (10° C.) for 16 hours. It was then treated with saturated aqueous ammonium chloride solution (10 mL) and extracted with a mixture of dichloromethane and MeOH (10:1, 3×30 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated in vacuo; purification via preparative thin-layer chromatography (Eluent: 20:1 dichloromethane/MeOH) afforded C66 as a colorless oil. Yield: 90 mg, 0.15 mmol, 57%. Step 2. Synthesis of methyl 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C67) A solution of C66 (90 mg, 0.15 mmol) in acetic acid (5 mL) was stirred at 50° C. for 16 hours. After the reaction mixture had been concentrated in vacuo, the residue was dissolved in a mixture of dichloromethane and MeOH (10:1, 30 mL) and washed with saturated aqueous sodium carbonate solution (20 mL). The aqueous layer was extracted with a mixture of dichloromethane and MeOH (10:1, 3×30 mL), and the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated in vacuo to provide C67 (90 mg) as a colorless oil, a portion of which was used directly in the following step. Step 3. Synthesis of Ammonium 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (1) To a solution of C67 (from the previous step; 80 mg, ≤0.13 mmol) in MeOH (2 mL) was added lithium hydroxide (4.96 mg, 0.209 mmol) and water (0.5 mL), and the reaction mixture was stirred at 50° C. for 16 hours. It was then concentrated in vacuo and purified via reversed-phase HPLC (Column: Agela Durashell C18, 5 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 13% to 33% B) to provide 1 as a white solid. Yield: 28 mg, 48 μmol, 37% over 2 steps. LCMS m/z 565.1♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.30 (d, 1H), 8.00-7.94 (m, 2H), 7.65 (d, 1H), 7.58 (dd, 1H), 7.51 (dd, 1H), 7.29-7.21 (m, 2H), 6.94 (dd, 1H), 5.42 (s, 2H), 5.29-5.21 (m, 1H), 4.9-4.83 (m, 1H, assumed; partially obscured by water peak), 4.71 (dd, 1H), 4.67-4.60 (m, 1H), 4.45 (dt, 1H), 3.99 (AB quartet, 2H), 3.11-3.04 (m, 1H), 3.02-2.94 (m, 1H), 2.92-2.74 (m, 2H), 2.57-2.47 (m, 1H), 2.40-2.25 (m, 2H), 1.90-1.69 (m, 4H). The compounds listed in Table 1 below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 1Structure and IUPAC name for Examples 2 to 18Ex.No.StructureIUPAC Name22-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-(1,3-oxazol-2-ylmethyl)- 1H-benzimidazole-6-carboxylic acid, trifluoroacetate salt32-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[(1-ethyl-1H-imidazol-5- yl)methyl]-1H-benzimidazole-6- carboxylic acid, trifluoroacetate salt42-[(4-{2-[(4-cyano-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-(1,3-oxazol-2-ylmethyl)- 1H-benzimidazole-6-carboxylic acid, trifluoroacetate salt52-[(4-{2-[(4-cyano-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[(2S)-oxetan-2- ylmethyl]-1H-benzimidazole-6- carboxylic acid62-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-(1,3-oxazol-5-ylmethyl)- 1H-benzimidazole-6-carboxylic acid, trifluoroacetate salt72-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-3-(1,3-oxazol-2-ylmethyl)- 3H-imidazo[4,5-b]pyridine-5- carboxylic acid, trifluoroacetate salt82-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[(3R)-tetrahydrofuran-3- ylmethyl]-1H-benzimidazole-6- carboxylic acid, trifluoroacetate salt92-[(4-{2-[(4-cyano-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-(2-methoxyethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt102-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[(1-methyl-1H-1,2,3- triazol-5-yl)methyl]-1H-benzimidazole- 6-carboxylic acid, trifluoroacetate salt112-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[(1-methyl-1H-imidazol- 5-yl)methyl]-1H-benzimidazole-6- carboxylic acid, trifluoroacetate salt12ammonium 2-[(4-{2-[(2,4- difluorobenzyl)oxy]pyridin-3- yl}piperidin-1-yl)methyl]-1-[(2S)- oxetan-2-ylmethyl]-1H-benzimidazole- 6-carboxylate132-[(4-{2-[(4-cyano-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[(2S)-tetrahydrofuran-2- ylmethyl]-1H-benzimidazole-6- carboxylic acid, trifluoroacetate salt142-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[2-(2-oxo-1,3- oxazolidin-3-yl)ethyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt152-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-3-[(1-ethyl-1H-imidazol-5- yl)methyl]-3H-imidazo[4,5-b]pyridine- 5-carboxylic acid, trifluoroacetate salt162-[(4-{2-[(4-cyano-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[(1-ethyl-1H-imidazol-5- yl)methyl]-1H-benzimidazole-6- carboxylic acid, trifluoroacetate salt172-[(4-{2-[(4-cyano-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-1-[(1-ethyl-1H-1,2,3- triazol-5-yl)methyl]-1H-benzimidazole- 6-carboxylic acid, trifluoroacetate salt181ammonium 2-[(4-{2-[(4-cyano-2- fluorobenzyl)oxy]pyridin-3-yl}piperidin- 1-yl)methyl]-3-[(2S)-oxetan-2- ylmethyl]-3H-imidazo[4,5-b]pyridine-5- carboxylate TABLE 1APhysicochemical data for Examples 2 to 18Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR2576.0♦;1H NMR (400 MHz, Methanol-d4): δ 8.36 (d, 1H), 8.09 (dd, 1H), 8.06 (dd,1H), 7.98 (d, 1H), 7.82 (d, 1H), 7.66 (dd, 1H), 7.56 (t, 1H), 7.31-7.22 (m, 2H),7.19 (d, 1H), 7.03 (dd, 1H), 5.86 (s, 2H), 5.48 (s, 2H), 4.89 (s, 2H), 3.96 (d, 2H),3.41 (t, 2H), 3.29-3.18 (m, 1H), 2.28-2.04 (m, 4H)3603.1♦; 2.43 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobilephase A: water containing 0.1% formic acid; Mobile phase B: acetonitrile;Gradient: 1.0% to 5% B over 0.6 minutes, then 5% to 100% B over 3.4 minutes;Flow rate: 0.8 mL/minute)4567.0;1H NMR (400 MHz, Methanol-d4): δ 8.36 (dd, 1H), 8.09-8.04 (m, 2H), 7.98(d, 1H), 7.82 (d, 1H), 7.73 (t, 1H), 7.67 (dd, 1H), 7.63 (dd, 1H), 7.59 (dd, 1H), 7.20(d, 1H), 7.04 (dd, 1H), 5.86 (s, 2H), 5.60 (s, 2H), 4.88 (s, 2H, assumed; partiallyobscured by water), 3.94 (d, 2H), 3.45-3.33 (m, 2H), 3.32-3.22 (m, 1H), 2.26-2.06 (m, 4H)5556.5;1H NMR (600 MHz, DMSO-d6): δ 12.76 (br s, 1H), 8.27 (s, 1H), 8.00 (d, 1H),7.92 (d, 1H), 7.80 (d, 1H), 7.77-7.72 (m, 1H), 7.71-7.58 (m, 3H), 7.05-6.97(m, 1H), 5.51 (s, 2H), 5.09 (d, 1H), 4.87-4.75 (m, 1H), 4.72-4.60 (m, 1H), 4.49(d, 1H), 4.43-4.32 (m, 1H), 3.95 (d, 1H), 3.80 (d, 1H), 3.01 (d, 1H), 2.88 (d, 1H),2.81 (t, 1H), 2.75-2.64 (m, 1H), 2.43 (d, 1H), 2.29-2.12 (m, 2H), 1.86-1.73 (m,2H), 1.72-1.54 (m, 2H)6576.1♦; 2.63 minutes (Analytical conditions identical to those used for Ex. 3)7577.0♦; 2.63 minutes (Column: Waters XBridge C18, 2.0 × 50 mm, 5 μm; Mobilephase A: water containing 0.1% formic acid; Mobile phase B: acetonitrilecontaining 0.1% formic acid; Gradient: 1.0% to 5% B over 0.6 minutes, then 5% to100% B over 3.4 minutes; Flow rate: 0.8 mL/minute)8579.1♦; 1.85 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 50 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine in 2-propanol; Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes,then 40% B for 1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.)9544.1; 2.52 minutes (Analytical conditions identical to those used for Ex. 3)10590.1♦; 2.70 minutes (Analytical conditions identical to those used for Ex. 3)11589.0♦; 2.47 minutes (Analytical conditions identical to those used for Ex. 3)12549.1; 1.45 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 50 mm, 3μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes, then40% B for 1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.)13570.1; 1.81 minutes (Analytical conditions identical to those used for Ex. 8)14608.0♦; 2.57 minutes (Analytical conditions identical to those used for Ex. 7)15604.0♦; 2.29 minutes (Analytical conditions identical to those used for Ex. 7)16594.1; 2.27 minutes (Analytical conditions identical to those used for Ex. 3)17595.0; 2.58 minutes (Column: Waters XBridge C18, 2.0 × 50 mm, 5 μm; Mobilephase A: water containing 10 mM ammonium carbonate; Mobile phase B:acetonitrile; Gradient: 1% to 5% B over 0.6 minutes, then 5% to 100% B over 3.4minutes; Flow rate: 0.8 mL/minute)181557.0; 4.34 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 100 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% to 40% B over 4.5 minutes, then 40% B for 2.5 minutes;Flow rate: 2.8 mL/minute; Column temperature: 40° C.)Table 1/1A:1Treatment of methyl 5-{[(4-{2-[(4-cyano-2-fluorobenzyl)oxy]pyridin-3-yl}piperidin-1-yl)acetyl]amino}-6-{[(2S)-oxetan-2-ylmethyl]amino}pyridine-2-carboxylate with aqueous sodium hydroxide solution in 1,4-dioxane at elevated temperature served to effect both ring closure and ester hydrolysis, delivering Example 18 after purification. Example 19 Ammonium 2-[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (19) Step 1. Synthesis of methyl 4-{[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)acetyl]amino}-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (C68) To a solution of P2 (800 mg, 2.02 mmol) and P16 (476 mg, 2.02 mmol) in N,N-dimethylformamide (12 mL) were added O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (1.15 g, 3.02 mmol) and triethylamine (408 mg, 4.03 mmol). The reaction mixture was stirred at 30° C. for 18 hours, whereupon it was diluted with saturated aqueous ammonium chloride solution (20 mL) and extracted with EtOAc (3×20 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (40 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to afford C68 as a yellow gum. Yield: 1.07 g, 1.79 mmol, 89%. Step 2. Synthesis of methyl 2-[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C69) A solution of C68 (1.08 g, 1.81 mmol) in acetic acid (8 mL) was stirred at 50° C. for 18 hours, whereupon it was concentrated in vacuo, carefully poured into saturated aqueous sodium bicarbonate solution (20 mL), and extracted with EtOAc (3×20 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (50 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Eluent: EtOAc) provided C69 as a yellow gum. Yield: 550 mg, 0.948 mmol, 52%. Step 3. Synthesis of Ammonium 2-[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (19) To a suspension of C69 (550 mg, 0.948 mmol) in a mixture of MeOH (5 mL) and THF (5 mL) was added aqueous sodium hydroxide solution (3 M; 6.32 mL, 19.0 mmol), and the reaction mixture was stirred at 15° C. for 3 hours. It was then concentrated in vacuo, diluted with water (8 mL), and washed with EtOAc (8 mL). The aqueous layer was adjusted to pH 6 to 7 by addition of 1 M hydrochloric acid, and extracted with EtOAc (3×10 mL). The combined organic extracts were washed with saturated aqueous sodium chloride solution (30 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. Purification via reversed-phase HPLC (Column: Agela Durashell C18, 5 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 26% to 56% B) afforded 19 as a white solid. Yield: 294 mg, 0.504 mmol, 53%. LCMS m/z 566.1♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.25 (br s, 1H), 8.06 (d, 1H), 7.99 (d, 1H), 7.95 (dd, 1H), 7.61 (d, 1H), 7.52 (dd, 1H), 7.28-7.21 (m, 2H), 5.46 (s, 2H), 5.30-5.22 (m, 1H), 4.93-4.83 (m, 1H, assumed; largely obscured by water peak), 4.70 (dd, 1H), 4.67-4.59 (m, 1H), 4.47 (dt, 1H), 3.96 (AB quartet, 2H), 3.13-3.02 (m, 2H), 2.95 (br d, 1H), 2.85-2.75 (m, 1H), 2.57-2.47 (m, 1H), 2.38-2.23 (m, 2H), 2.00-1.78 (m, 4H). The compounds listed in Table 2 below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 2Structure and IUPAC name for Examples 20 to 28Ex.No.StructureIUPAC Name202-[(4-{3-[(4-chloro-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-[(1-ethyl- 1H-imidazol-5-yl)methyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt212-[(4-{3-[(4-chloro-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-(1,3- oxazol-4-ylmethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt222-[(4-{3-[(4-chloro-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-(1,3- oxazol-2-ylmethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt232-[(4-{3-[(4-chloro-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-(2- methoxyethyl)-1H-benzimidazole-6- carboxylic acid, hydrochloride salt242-[(4-{3-[(4-chloro-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-(1,3- oxazol-5-ylmethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt252-[(4-{3-[(4-cyano-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-(1,3- oxazol-2-ylmethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt262-[(4-{3-[(4-chloro-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-[(1-methyl- 1H-imidazol-5-yl)methyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt272-[(4-{3-[(4-cyano-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-l-yl)methyl]-1-[(1-ethyl- 1H-imidazol-5-yl)methyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt28ammonium 2-[(4-{3-[(4-cyano-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-[(2S)- oxetan-2-ylmethyl]-1H- benzimidazole-6-carboxylate TABLE 2APhysicochemical data for Examples 20 to 28Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR20604.1♦; 2.38 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobilephase A: water containing 0.1% formic acid; Mobile phase B: acetonitrile;Gradient: 1.0% to 5% B over 0.6 minutes, then 5% to 100% B over 3.4 minutes;Flow rate: 0.8 mL/minute)21577.0♦; 2.64 minutes (Analytical conditions identical to those used for Ex. 20)22577.0♦; 2.66 minutes (Analytical conditions identical to those used for Ex. 20)23554.2♦;1H NMR (600 MHz, DMSO-d6), characteristic peaks: δ 8.31 (s, 1H), 8.22(s, 1H), 8.14 (s, 1H), 7.89 (d, 1H), 7.78 (d, 1H), 7.61 (dd, 1H), 7.52 (d, 1H), 7.34(d, 1H), 5.45 (s, 2H), 4.86-4.68 (br m, 2H), 4.67-4.58 (m, 2H), 3.87-3.71 (brm, 2H), 3.69-3.61 (m, 2H), 3.5-3.2 (m, 2H, assumed; partially obscured bywater peak), 3.20 (s, 3H), 2.17-1.99 (br m, 4H)24577.0♦; 2.60 minutes (Analytical conditions identical to those used for Ex. 20)25568.1; 2.45 minutes (Analytical conditions identical to those used for Ex. 20)26590.0♦; 2.31 minutes (Analytical conditions identical to those used for Ex. 20)27595.1; 2.16 minutes (Analytical conditions identical to those used for Ex. 20)28557.1; 1.52 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 50 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes, then40% B for 1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.) Example 29 2-[(4-{3-[(4-Chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-methyl-1H-imidazo[4,5-c]pyridine-6-carboxylic Acid, Trifluoroacetate Salt (29) Step 1. Synthesis of N-[6-bromo-4-(methylamino)pyridin-3-yl]-2-(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)acetamide (C70) To a solution of P2, HCl salt (180 mg, 0.47 mmol) in N,N-dimethylformamide (5 mL) were added 6-bromo-N4-methylpyridine-3,4-diamine (96 mg, 0.47 mmol), 0-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (270 mg, 0.71 mmol), and N,N-diisopropylethylamine (0.25 mL, 1.4 mmol). The reaction mixture was stirred at 25° C. for 16 hours, whereupon it was diluted with water (60 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by preparative thin-layer chromatography (EtOAc) to afford C70 as a red gum. Yield: 200 mg, 0.36 mmol, 75%. Step 2. Synthesis of 6-bromo-2-[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-methyl-1H-imidazo[4,5-c]pyridine (C71) To a solution of C70 (200 mg, 0.36 mmol) in 1,4-dioxane (9 mL) was added aqueous sodium hydroxide solution (2 M; 1.8 mL, 4 mmol). The solution was stirred at 100° C. for 2 hours. The mixture was then diluted with water (30 mL) and extracted with EtOAc (3×40 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. The material was combined with further crude material and purified by preparative thin-layer chromatography (EtOAc) to afford C71 as a yellow solid (140 mg, 0.26 mmol, 48% based on combined starting materials). Step 3. Synthesis of methyl 2-[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-methyl-1H-imidazo[4,5-c]pyridine-6-carboxylate (C72) 1,3-Bis(diphenylphosphino)propane (25 mg, 62 μmol), palladium(II) acetate (26 mg, 0.12 mmol), and triethylamine (0.35 mL, 2.5 mmol) were added to a solution of C71 (140 mg, 0.26 mmol) in a mixture of MeOH (3 mL) and N,N-dimethylformamide (2 mL), and the reaction mixture was stirred at 80° C., under carbon monoxide (50 psi), for 16 hours. It was then concentrated in vacuo and purified by preparative thin-layer chromatography (EtOAc) to afford C72 as a yellow solid (110 mg, 0.21 mmol, 82%). Step 4. Synthesis of 2-[(4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}piperidin-1-yl)methyl]-1-methyl-1H-imidazo[4,5-c]pyridine-6-carboxylic Acid, Trifluoroacetate Salt (29) Aqueous lithium hydroxide solution (2 M; 0.4 mL, 0.8 mmol) was added to a solution of C72 (80 mg, 0.15 mmol) in a mixture of THF (2 mL) and MeOH (2 mL), and the reaction mixture was stirred at 30° C. for 2 hours. After concentration in vacuo, the residue was adjusted to pH 4 with trifluoroacetic acid, and then purified using reversed-phase HPLC (Column: Waters XBridge C18 OBD, 5 μm; Mobile phase A: water containing 0.1% trifluoroacetic acid; Mobile phase B: acetonitrile; Gradient: 5% to 95% B); 29 was isolated as a white solid. Yield: 52.4 mg, 83.8 μmol, 56%. LCMS m/z 511.2♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 9.11 (s, 1H), 8.60 (s, 1H), 8.16 (d, 1H), 8.09 (d, 1H), 7.56 (dd, 1H), 7.30-7.22 (m, 2H), 5.50 (s, 2H), 4.91 (s, 2H), 4.02-3.93 (m, 2H), 4.01 (s, 3H), 3.54-3.41 (m, 3H), 2.38-2.18 (m, 4H). The compounds listed in Table 3 below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 3Structure and IUPAC name for Examples 30 to 32Ex. No.StructureIUPAC Name302-[(4-{3-[(4-chloro-2- fluorobenzyl)oxy]pyrazin-2- yl}piperidin-1-yl)methyl]-1-methyl-1H- imidazo[4,5-b]pyridine-6-carboxylic acid, hydrochloride salt312-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-1-(2- methoxyethyl)-1H-imidazo[4,5- c]pyridine-6-carboxylic acid, trifluoroacetate salt, single enantiomer; from P7322-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-1-(2- methoxyethyl)-1H-imidazo[4,5- c]pyridine-6-carboxylic acid, trifluoroacetate salt, single enantiomer; from C23 TABLE 3APhysicochemical data for Examples 30 to 32Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR30511.3;1H NMR (600 MHz, DMSO-d6), characteristic peaks: δ 9.02 (s, 1H), 8.65 (s,1H), 8.22 (s, 1H), 8.13 (s, 1H), 7.61 (dd, 8.0, 1H), 7.52 (d, 1H), 7.34 (d, 1H), 5.46(s, 2H), 4.95-4.71 (br m, 2H), 3.96 (s, 3H), 3.90-3.69 (br m, 2H), 3.5-3.2 (m,2H, assumed; partially obscured by water peak), 2.22-1.92 (br m, 4H)31585.1♦; 9.26 minutes (Column: Chiral Technologies Chiralcel AS-RH, 4.6 × 150mm, 5 μm: Mobile phase A: 0.069% trifluoroacetic acid in water; Mobile phase B:acetonitrile; Eluent: 20% B; Flow rate: 0.8 mL/minute)32585.1♦; 9.79 minutes (Column: Chiral Technologies Chiralcel OJ-RH, 4.6 × 150mm, 5 μm: Mobile phase A: 0.069% trifluoroacetic acid in water; Mobile phase B:acetonitrile; Gradient: 10% to 80% B over 25 minutes; Flow rate: 0.8 mL/minute) Example 33 Ammonium 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (33) Step 1. Synthesis of methyl 4-{[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazin-1-yl)acetyl]amino}-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (C73) A solution of P3 (95.0 mg, 0.250 mmol), P16 (70.5 mg, 0.298 mmol) and O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (143 mg, 0.376 mmol) in N,N-dimethylformamide (1 mL) was stirred at 20° C. for 30 minutes, whereupon triethylamine (50.6 mg, 0.500 mmol) was added. After the reaction mixture had been stirred at 15° C. for 2 hours, it was diluted with EtOAc (50 mL) and washed with water (50 mL). The organic layer was washed with saturated aqueous sodium chloride solution (50 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo; preparative thin-layer chromatography (Eluent: EtOAc) provided C73 as a yellow oil (150 mg), which was advanced directly to the following step. Step 2. Synthesis of methyl 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C74) A solution of C73 (from the previous step; 150 mg, 50.250 mmol) in acetic acid (2 mL) was stirred at 50° C. for 16 hours, whereupon it was concentrated in vacuo to dryness, mixed with EtOAc (60 mL), and washed with saturated aqueous sodium carbonate solution (50 mL). The organic layer was dried over magnesium sulfate, filtered, concentrated under reduced pressure, and purified using preparative thin-layer chromatography (Eluent: EtOAc) to afford C74 as a yellow oil. Yield: 80.5 mg, 0.139 mmol, 56% over 2 steps. Step 3. Synthesis of Ammonium 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyridin-3-yl}piperazin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (33) A solution of C74 (80.0 mg, 0.138 mmol) and aqueous sodium hydroxide solution (3 M; 0.3 mL, 0.9 mmol) in MeOH (1 mL) and THF (1 mL) was stirred at 50° C. for 1 hour. The reaction mixture was then adjusted to pH 7 by addition of 1 M hydrochloric acid, and extracted with a mixture of dichloromethane and MeOH (10:1, 3×30 mL). The combined organic layers were dried over magnesium sulfate, filtered, and concentrated in vacuo; purification using reversed-phase HPLC (Column: Agela Durashell C18, 5 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 20% to 50% B) provided 33 as a white solid. Yield: 39.7 mg, 68.0 μmol, 49%. LCMS m/z 566.0♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.32 (d, 1H), 7.97 (dd, 1H), 7.75 (dd, 1H), 7.67 (d, 1H), 7.55 (dd, 1H), 7.28-7.20 (m, 3H), 6.92 (dd, 1H), 5.42 (s, 2H), 5.29-5.21 (m, 1H), 4.9-4.81 (m, 1H, assumed; partially obscured by water peak), 4.70 (dd, 1H), 4.66-4.59 (m, 1H), 4.46 (dt, 1H), 3.98 (AB quartet, 2H), 3.16-3.04 (br m, 4H), 2.84-2.74 (m, 1H), 2.74-2.61 (m, 4H), 2.56-2.45 (m, 1H). Example 34 2-{[(2S)-4-{3-[(4-Chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}-2-methylpiperazin-1-yl]methyl}-3-methyl-3H-imidazo[4,5-b]pyridine-5-carboxylic Acid, Trifluoroacetate Salt (34) Step 1. Synthesis of methyl 2-{[(2S)-4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}-2-methylpiperazin-1-yl]methyl}-3-methyl-3H-imidazo[4,5-b]pyridine-5-carboxylate (C75) To a suspension of P18 (60.0 mg, 0.198 mmol), P4 (64.8 mg, 0.237 mmol), 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphane) (24.6 mg, 39.6 μmol), and tris(dibenzylideneacetone)dipalladium(0) (18.1 mg, 19.8 μmol) in toluene (2 mL) was added cesium carbonate (129 mg, 0.396 mmol). The reaction mixture was purged with nitrogen for 30 seconds and stirred at 100° C. for 16 hours, whereupon it was diluted with water (10 mL) and extracted with dichloromethane (3×10 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Preparative thin-layer chromatography (Eluent: 20:1 dichloromethane/MeOH) afforded C75 as a yellow solid. Yield: 60 mg, 0.11 μmol, 56%. Step 2. Synthesis of 2-{[(2S)-4-{3-[(4-chloro-2-fluorobenzyl)oxy]pyrazin-2-yl}-2-methylpiperazin-1-yl]methyl}-3-methyl-3H-imidazo[4,5-b]pyridine-5-carboxylic Acid, Trifluoroacetate Salt (34) To a solution of C75 (60 mg, 0.11 mmol) in MeOH (5 mL) was added THF (1 mL) and a solution of sodium hydroxide (22.2 mg, 0.556 mmol) in water (1 mL). After the reaction mixture had been stirred at 40° C. for 2 hours, it was concentrated in vacuo and acidified to pH 7 by addition of 1 M hydrochloric acid. The resulting mixture was extracted with dichloromethane (3×20 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. Purification via reversed-phase HPLC (Column: Waters XBridge C18 OBD, 5 μm; Mobile phase A: water containing 0.1% trifluoroacetic acid; Mobile phase B: acetonitrile; Gradient: 5% to 95% B) afforded 34 as a solid. Yield: 43.4 mg, 67.8 μmol, 62%. LCMS m/z 526.0♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.20 (AB quartet, 2H), 7.80 (d, 1H), 7.70 (d, 1H), 7.54 (dd, 1H), 7.27 (dd, 1H), 7.24 (br dd, 1H), 5.49 (AB quartet, 2H), 5.01 (d, 1H), 4.68 (d, 1H), 4.14-4.01 (m, 2H), 3.99 (s, 3H), 3.87-3.69 (m, 2H), 3.67-3.40 (m, 3H), 1.49 (d, 3H). Examples 35 and 36 Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C78 (35) and Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C79 (36) Step 1. Synthesis of methyl 4-{[(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)carbonyl]amino}-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (C76) To a solution of P6 (530 mg, 1.39 mmol), O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (794 mg, 2.09 mmol), and N,N-diisopropylethylamine (0.70 mL, 4.1 mmol) in N,N-dimethylformamide (4.0 mL) was added a solution of P16 (324 mg, 1.37 mmol) in N,N-dimethylformamide (4.0 mL). After the reaction mixture had been stirred at 25° C. for 15 hours, it was concentrated in vacuo to remove N,N-dimethylformamide. The resulting gum was washed with saturated aqueous sodium chloride solution (20 mL), and extracted with EtOAc (3×20 mL). The combined organic layers were dried over magnesium sulfate, filtered, concentrated under reduced pressure, and purified via silica gel chromatography (Gradient: 0% to 1.3% MeOH in dichloromethane) followed by preparative thin-layer chromatography (Eluent: 4:1 EtOAc/petroleum ether). Compound C76 was isolated as a red gum. Yield: 530 mg, 0.884 mmol, 64%. Step 2. Synthesis of methyl 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C77) A solution of C76 (530 mg, 0.884 mmol) in acetic acid (5.0 mL) was stirred at 60° C. for 16 hours, whereupon it was concentrated in vacuo, washed with saturated aqueous sodium bicarbonate solution (10 mL), and extracted with EtOAc (3×20 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. Purification was carried out twice using preparative thin-layer chromatography (Eluent for #1: 1:1 petroleum ether/EtOAc; Eluent for #2: 10:1 dichloromethane/MeOH), and then via reversed-phase HPLC (Column: Phenomenex Gemini C18, 10 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 60% to 90% B), affording C77 as a solid that consisted of a mixture of two stereoisomers at the cyclopropane. Yield: 140 mg, 0.241 mmol, 27%. Step 3. Isolation of methyl 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-1 (C78) and methyl 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-2 (C79) Separation of C77 (185 mg, 0.318 mmol) into the component stereoisomers at the cyclopropane was carried out using supercritical fluid chromatography [Column: Chiral Technologies Chiralcel OJ-H, 5 μm; Mobile phase: 3:2 carbon dioxide/(MeOH containing 0.1% ammonium hydroxide)]. The first-eluting stereoisomer, obtained as a yellow gum, was designated as ENT-1 (C78). Yield: 80 mg, 0.14 mmol, 44%. The second-eluting stereoisomer, isolated as a green gum, was designated as ENT-2 (C79). Yield: 100 mg, 0.17 mmol, assumed 50%. Step 4. Synthesis of Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C78 (35) A solution of C78 (75 mg, 0.13 mmol) in a mixture of THF (5.0 mL) and MeOH (2.0 mL) was treated with aqueous lithium hydroxide solution (2 M; 0.32 mL, 0.64 mmol), and the reaction mixture was stirred at 30° C. for 60 hours. It was then adjusted to pH 6 to 7 by addition of 1 M hydrochloric acid, and extracted with dichloromethane (4×15 mL); the combined organic layers were dried over magnesium sulfate, filtered, and concentrated in vacuo. Purification using reversed-phase HPLC (Column: Agela Durashell C18, 5 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 20% to 40% B) afforded 35 as a white solid. Yield: 11.3 mg, 18.8 μmol, 14%. LCMS m/z 568.1 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.22 (d, 1H), 7.94 (dd, 1H), 7.64-7.57 (m, 2H), 7.53-7.47 (m, 2H), 7.41 (dd, 1H), 6.27 (d, 1H), 6.09 (d, 1H), 5.40 (AB quartet, 2H), 5.23-5.15 (m, 1H), 4.69 (dd, 1H), 4.63-4.53 (m, 2H), 4.41 (dt, 1H), 3.98-3.90 (m, 1H), 3.61-3.52 (m, 1H), 3.42 (ddd, 1H), 3.23 (ddd, 1H), 2.84-2.73 (m, 1H), 2.58-2.48 (m, 1H), 2.38 (dd, 1H), 1.86 (ddd, 1H), 1.65 (dd, 1H), 1.54-1.44 (m, 2H), 1.36-1.27 (m, 1H), 1.24 (dd, 1H). Step 5. Synthesis of Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C79 (36) A solution of C79 (95 mg, 0.16 mmol) in a mixture of THF (5.0 mL) and MeOH (2.0 mL) was treated with aqueous lithium hydroxide solution (2 M; 0.40 mL, 0.80 mmol), and the reaction mixture was stirred at 30° C. for 50 hours. It was then adjusted to pH 5 to 6 by addition of 1 M hydrochloric acid, and extracted with dichloromethane (4×20 mL); the combined organic layers were dried over magnesium sulfate, filtered, and concentrated in vacuo. Purification using reversed-phase HPLC (Column: Agela Durashell C18, 5 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 21% to 41% B) afforded 36 as a white solid. Yield: 21.9 mg, 38.5 μmol, 24%. LCMS m/z 568.1 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.21 (d, 1H), 7.95 (dd, 1H), 7.62 (d, 1H), 7.60 (dd, 1H), 7.53-7.47 (m, 2H), 7.40 (dd, 1H), 6.25 (d, 1H), 6.08 (d, 1H), 5.40 (AB quartet, 2H), 5.28-5.20 (m, 1H), 4.69 (dd, 1H), 4.58-4.48 (m, 2H), 4.31 (dt, 1H), 3.95-3.86 (m, 1H), 3.59-3.50 (m, 1H), 3.41 (ddd, 1H), 3.22 (ddd, 1H), 2.81-2.71 (m, 1H), 2.52-2.41 (m, 2H), 1.86-1.77 (m, 1H), 1.66 (dd, 1H), 1.56-1.42 (m, 2H), 1.36-1.27 (m, 1H), 1.23 (dd, 1H). Example 37, in Table 4 below, was synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. Example 37 was purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 4Structure and IUPAC name for Example 37Ex.No.StructureIUPAC Name372-(6-{6-[(4-cyano-2- fluorobenzyl)oxy]pyridin-2-yl}-6- azaspiro[2.5]oct-1-yl)-1-[(1-methyl- 1H-imidazol-5-yl)methyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt TABLE 4APhysicochemical data for Example 37Ex No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR37592.2; 2.68 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobilephase A: water containing 0.1% formic acid; Mobile phase B: acetonitrile;Gradient: 1.0% to 5% B over 0.6 minutes, then 5% to 100% B over 3.4 minutes;Flow rate: 0.8 mL/minute) Example 38 Ammonium 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from P7 (38) Step 1. Synthesis of methyl 4-{[(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)carbonyl]amino}-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate, from P7 (C80) To a solution of P16 (540 mg, 2.29 mmol) and P7 (1.03 g, 2.51 mmol) in pyridine (10 mL) was added 1-[3-(dimethylamino)propyl]-3-ethylcarbodiimide hydrochloride (1.31 g, 6.86 mmol). The reaction mixture was stirred at 30° C. for 16 hours, whereupon it was poured into water (20 mL) and extracted with EtOAc (3×20 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo; silica gel chromatography (Eluent: 85% EtOAc in petroleum ether) afforded C80 as a white foam. Yield: 1.4 g, 2.23 mmol, 97%. Step 2. Synthesis of methyl 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from P7 (C81) A solution of C80 (1.4 g, 2.2 mmol) in acetic acid (20 mL) was stirred at 60° C. for 21 hours, whereupon the reaction mixture was concentrated to dryness in vacuo and neutralized by addition of aqueous sodium bicarbonate solution (20 mL). The resulting mixture was extracted with EtOAc (3×20 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Eluent: EtOAc) provided C81 as a foamy white solid. Yield: 1.1 g, 1.8 mmol, 82%. Step 3. Synthesis of Ammonium 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from P7 (38) Aqueous sodium hydroxide solution (2 M; 4.5 mL, 9.0 mmol) was added to a solution of C81 (1.1 g, 1.8 mmol) in a mixture of THF (11 mL) and MeOH (4.5 mL), and the reaction mixture was stirred at 25° C. for 16 hours. It was then combined with a similar reaction carried out using C81 (366 mg, 0.600 mmol), concentrated to dryness in vacuo, taken up in water (10 mL), and adjusted to pH 5 by addition of 1 M hydrochloric acid. The resulting mixture was extracted with dichloromethane (4×50 mL); the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography (Eluent: 20% MeOH in dichloromethane) provided a white solid, which was dissolved in acetonitrile (5 mL), and then treated with water (20 mL) and concentrated ammonium hydroxide (1 mL). This mixture was lyophilized to afford 38 as a white solid. Yield: 1.09 g, 1.78 mmol, 74%. LCMS m/z 596.0♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.20 (d, 1H), 7.95 (dd, 1H), 7.86 (d, 1H), 7.62 (d, 1H), 7.44 (dd, 1H), 7.18-7.11 (m, 2H), 5.30 (s, 2H), 5.28-5.20 (m, 1H), 4.73 (dd, 1H), 4.59-4.49 (m, 2H), 4.34 (dt, 1H), 4.28-4.20 (m, 1H), 3.99-3.90 (m, 1H), 3.70-3.60 (m, 1H), 3.53-3.44 (m, 1H), 2.84-2.73 (m, 1H), 2.56-2.44 (m, 2H), 2.00-1.89 (m, 1H), 1.69 (dd, 1H), 1.65-1.50 (m, 2H), 1.43-1.34 (m, 1H), 1.28 (dd, 1H). The compounds listed in Table 5 below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 5Structure and IUPAC name for Examples 39 to 45Ex.No.StructureIUPAC Name392-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-1-[(1- ethyl-1H-imidazol-5-yl)methyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt, from P74012-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-3-methyl- 3H-imidazo[4,5-b]pyridine-5- carboxylic acid, trifluoroacetate salt, from P74122-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-1-[2-(1H- 1,2,4-triazol-1-yl)ethyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt, from P74232-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-1-[2-(1H- 1,2,4-triazol-1-yl)ethyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt, from C23432-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-1-(1,3- oxazol-4-ylmethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt, from P7442-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-1-(1,3- oxazol-2-ylmethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt, from P7452-(6-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-6-azaspiro[2.5]oct-1-yl)-1-(1,3- oxazol-5-ylmethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt, from P7 TABLE 5APhysicochemical data for Examples 39 to 45Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR39634.1♦; 2.02 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 50 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes, then40% B for 1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.)401541.1♦; 3.54 minutes (Column: Chiral Technologies Chiralpak AS-3, 4.6 × 100 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% to 40% B for 4.5 minutes, then 40% B over 2.5 minutes;Flow rate: 2.8 mL/minute; Column temperature: 40° C.)412621.3♦; 5.81 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 100 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% to 40% B over 4.5 minutes, then 40% B over 2.5 minutes;Flow rate: 2.8 mL/minute; Column temperature: 40° C.)423621.3♦; 4.96 minutes (Analytical conditions identical to those used for Example 41)43607.1♦; 3.51 minutes (Column: Chiral Technologies Chiralcel OJ-3, 4.6 × 100 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inMeOH; Gradient: 5% to 40% B over 4.5 minutes, then 40% B for 2.5 minutes;Flow rate: 2.8 mL/minute; Column temperature: 40° C.)44607.1♦; 1.49 minutes (Column: Chiral Technologies Chiralcel OJ-3, 4.6 × 50 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes, then40% B for 1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.)45607.1♦; 3.84 minutes (Analytical conditions identical to those used for Example 43)Table 5/5A:1The amide product from coupling of P7 with C60 was cyclized by heating in trimethylsilyl polyphosphate at 120° C. for 30 minutes, affording a single enantiomer of 5-chloro-2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-3-methyl-3H-imidazo[4,5-b]pyridine.2The amide product from coupling of P7 with methyl 4-amino-3-{[2-(1H-1,2,4-triazol-1-yl)ethyl]amino}benzoate was cyclized by heating at elevated temperature with aqueous sodium hydroxide solution in 1,4-dioxane; ester hydrolysis was also effected, providing Example 41 after purification.3The amide product from coupling of C23 with methyl 4-amino-3-{[2-(1H-1,2,4-triazol-1-yl)ethyl]amino}benzoate was cyclized by heating to elevated temperature with aqueous sodium hydroxide solution in 1,4-dioxane; ester hydrolysis was also effected, providing Example 42 after purification. Examples 46 and 47 Ammonium 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate, ENT-1 (46) and Ammonium 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate, ENT-2 (47) Step 1. Synthesis of methyl 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate (C82) A vial containing a mixture of P8 (61 mg, 0.22 mmol), P19 (55 mg, 0.16 mmol), tris(dibenzylideneacetone)dipalladium(0) (14.7 mg, 16.0 μmol), 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphane) (20 mg, 32 μmol), and cesium carbonate (104 mg, 0.319 mmol) was evacuated and then purged with nitrogen for 10 minutes. After addition of 1,4-dioxane (0.8 mL), the reaction mixture was heated to 100° C. for 23 hours, whereupon it was diluted with dichloromethane (0.5 mL) and directly purified via silica gel chromatography (Gradient: 15% to 100% EtOAc in heptane). Compound C82 was isolated as a yellow solid (37.5 mg), which was taken directly into the following reaction. Step 2. Synthesis of Ammonium 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate, ENT-1 (46) and Ammonium 2-(6-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-6-azaspiro[2.5]oct-1-yl)-1-(2-methoxyethyl)-1H-benzimidazole-6-carboxylate, ENT-2 (47) Aqueous sodium hydroxide solution (2 M; 150 μL, 0.3 mmol) was added to a mixture of C82 (from the previous step; 37.5 mg, 564.6 μmol) in MeOH (0.4 mL) and THF (0.4 mL). The reaction mixture stirred at 35° C. for 2.25 hours, whereupon hydrochloric acid (1 M; 0.35 mL, 0.35 mmol) was added, and the reaction mixture was allowed to cool to room temperature. After removal of solvents in vacuo, the residue was separated into its component enantiomers using supercritical fluid chromatography [Column: Chiral Technologies Chiralpak AD-H, 5 μm; Mobile phase: 65:35 carbon dioxide/(MeOH containing 0.2% ammonium hydroxide)]. The first-eluting enantiomer was designated as ENT-1 (46). Yield: 4.3 mg, 7.3 μmol, 5% over 2 steps. LCMS m/z 566.4♦ [M+H]+. Retention time: 2.12 minutes [Analytical conditions, Column: Chiral Technologies Chiralpak AD-H, 4.6×100 mm, 5 μm; Mobile phase: 1:1 carbon dioxide/(MeOH containing 0.2% ammonium hydroxide); Flow rate: 1.5 mL/minute; Back pressure: 120 bar]. The second-eluting enantiomer was designated as ENT-2 (47). Yield: 4.1 mg, 7.0 μmol, 4% over 2 steps. LCMS m/z 566.4♦ [M+H]+. Retention time: 2.74 minutes (Analytical conditions identical to those used for 46). The compounds listed in Table 6 below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 6Structure and IUPAC name for Examples 48 and 49Ex.No.StructureIUPAC Name4812-(6-{6-[(4-chloro-2- fluorobenzyl)oxy]pyridin-2-yl}-6- azaspiro[2.5]oct-1-yl)-1-(2- methoxyethyl)-1H-imidazo[4,5- b]pyridine-6-carboxylic acid, ENT- 11,24912-(6-{6-[(4-chloro-2- fluorobenzyl)oxy]pyridin-2-yl}-6- azaspiro[2.5]oct-1-yl)-1-(2- methoxyethyl)-1H-imidazo[4,5- b]pyridine-6-carboxylic acid, ENT- 21,21In this case, the coupling between P30 and 2-chloro-6-[(4-chloro-2-fluorobenzyl)oxy]pryidine was catalyzed by chloro(2-dicyclohexylphosphino-2′,6′-diisopropoxy-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II) (RuPhos Pd G2) and 2-dicyclohexylphosphino-2′,6′-diisopropxybiphenyl.2A racemic mixture of Examples 48 and 49 was separated into its component enantiomers using supercritical fluid chromatography [Column: Phenomenex Lux Cellulose-2, 5 μm; Mobile phase: 7:3 carbon dioxide/(MeOH containing 0.2% ammonium hydroxide and 5% water)]. The first-eluting enantiomer was designated as ENT-1 (48), and the second-eluting enantiomer was designated as ENT-2 (49). TABLE 6APhysicochemical data for Examples 48 and 49Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR48566.3; 8.04 minutes [Column: Phenomenex Lux Cellulose-2, 4.6 × 100 mm, 5 μm;Mobile phase: 3:2 carbon dioxide/(MeOH containing 0.2% ammonium hydroxideand 5% water); Flow rate: 1.5 mL/minute; Column temperature: 40° C.; Backpressure: 200 bar]49566.3; 11.33 minutes (Analytical conditions identical to those used for Ex. 48) Example 50 and 51 Ammonium 2-(6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C85 (50) and Ammonium 2-(6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C86 (51) Step 1. Synthesis of methyl 4-{[(6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]oct-1-yl)carbonyl]amino}-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (C83) Reaction of P16 with P9 was carried out using the method described for synthesis of C80 from P16 and P7 in Example 38. In this case, purification was carried out using silica gel chromatography (Gradient: 0% to 2.1% MeOH in dichloromethane), affording C83, a mixture of stereoisomers at the cyclopropane, as a yellow foamy solid. Yield: 504 mg, 0.802 mmol, 94%. Step 2. Synthesis of methyl 2-(6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C84) A solution of C83 (270 mg, 0.430 mmol) in acetic acid (4.5 mL) was stirred at 60° C. for 7 hours, whereupon it was poured into water, basified to pH 8 by addition of sodium carbonate, and extracted with EtOAc (3×30 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated in vacuo, and purified via silica gel chromatography (Gradient: 0% to 80% EtOAc in petroleum ether) to provide C84, a mixture of stereoisomers at the cyclopropane, as a yellow oil. Yield: 250 mg, 0.410 mmol, 95%. Step 3. Isolation of methyl 2-(6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-1 (C85) and methyl 2-(6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-2 (C86) Separation of C84 (250 mg, 0.410 mmol) into its component stereoisiomers at the cyclopropane center was carried out via supercritical fluid chromatography [Column: Chiral Technologies Chiralcel OJ-H, 5 μm; Mobile phase: 3:2 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]. This first-eluting stereoisomer, a colorless oil, was designated as ENT-1 (C85). Yield: 104 mg, 0.170 mmol, 41%. The second-eluting stereoisomer, a white solid, was designated as ENT-2 (C86). Yield: 109 mg, 0.179 mmol, 44%. Step 4. Synthesis of Ammonium 2-(6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C85 (50) Aqueous sodium hydroxide solution (0.767 mL, 2 M, 1.53 mmol) was added to a solution of C85 (104 mg, 0.170 mmol) in MeOH (3 mL). After the reaction mixture had stirred at 30° C. for 6 hours, it was acidified by careful addition of 12 M hydrochloric acid, diluted with water (30 mL), and extracted with dichloromethane (3×30 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated in vacuo, and purified via reversed-phase HPLC (Column: Phenomenex Gemini C18, 10 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 25% to 55% B) to afford 50 as a white solid. Yield: 47.1 mg, 76.8 μmol, 45%. LCMS m/z 596.0♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.20 (d, 1H), 7.97-7.92 (m, 2H), 7.61 (d, 1H), 7.45 (dd, 1H), 7.20-7.13 (m, 2H), 5.42 (s, 2H), 5.28-5.20 (m, 1H), 4.70 (dd, 1H), 4.58-4.49 (m, 2H), 4.33 (dt, 1H), 4.24-4.16 (m, 1H), 3.92-3.83 (m, 1H), 3.56 (ddd, 1H), 3.39 (ddd, 1H), 2.82-2.71 (m, 1H), 2.54-2.42 (m, 2H), 1.90-1.80 (m, 1H), 1.68 (dd, 1H), 1.54-1.43 (m, 2H), 1.35-1.28 (m, 1H), 1.25 (dd, 1H). Step 5. Synthesis of Ammonium 2-(6-{4-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C86 (51) Conversion of C86 to 51 was carried out using the method described for synthesis of 50 from C85 in step 4 above. Compound 51 was isolated as a white solid. Yield: 46.0 mg, 75.0 μmol, 42%. LCMS m/z 596.0♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.23 (d, 1H), 7.97 (d, 1H), 7.95 (dd, 1H), 7.62 (d, 1H), 7.45 (dd, 1H), 7.20-7.13 (m, 2H), 5.43 (s, 2H), 5.23-5.15 (m, 1H), 4.70 (dd, 1H), 4.63-4.54 (m, 2H), 4.40 (dt, 1H), 4.29-4.21 (m, 1H), 3.96-3.87 (m, 1H), 3.57 (ddd, 1H), 3.41 (ddd, 1H), 2.85-2.74 (m, 1H), 2.59-2.48 (m, 1H), 2.42 (dd, 1H), 1.95-1.86 (m, 1H), 1.67 (dd, 1H), 1.54-1.42 (m, 2H), 1.36-1.24 (m, 2H). Examples 52 and 53 Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-1 (52) and Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-2 (53) Step 1. Synthesis of methyl 4-{[(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)carbonyl]amino}-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (C87) To a solution of P10 (695 mg, 1.74 mmol), P16 (411 mg, 1.74 mmol), and O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (860 mg, 2.26 mmol) in N,N-dimethylformamide (7 mL) was added triethylamine (880 mg, 8.70 mmol). The reaction mixture was stirred at 35° C. for 16 hours, whereupon it was combined with a similar reaction carried out using P10 (50 mg, 0.12 mmol), poured into water (20 mL), and extracted with EtOAc (3×20 mL). The combined organic layers were washed with aqueous ammonium chloride solution (2×30 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Purification was carried out via silica gel chromatography (Eluent: 80% EtOAc in petroleum ether) followed by preparative thin-layer chromatography (Eluent: EtOAc) to provide C87, a mixture of stereoisomers at the cyclopropane, as a yellow foam. Combined yield: 525 mg, 0.850 mmol, 46%. Step 2. Synthesis of methyl 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C88) A solution of C87 (351 mg, 0.568 mmol) in acetic acid (4 mL) was stirred at 60° C. for 16 hours, whereupon it was combined with a similar reaction carried out using C87 (174 mg, 0.282 mmol) and concentrated to dryness in vacuo. The residue was neutralized with aqueous sodium bicarbonate solution and extracted with dichloromethane (3×10 mL); the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford C88, a mixture of stereoisomers at the cyclopropane, as a yellow, foamy solid. Combined yield: 510 mg, 0.850 mmol, quantitative. Step 3. Synthesis of Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-1 (52) and Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-5-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-2 (53) To a solution of C88 (490 mg, 0.817 mmol) in a mixture of THF (8 mL) and MeOH (1 mL) was added aqueous lithium hydroxide solution (2 M; 1.63 mL, 3.26 mmol), and the reaction mixture was stirred at 25° C. for 16 hours. It was then combined with a similar reaction carried out using C88 (20 mg, 33 μmol), concentrated in vacuo, and diluted with water (5 mL). The resulting mixture was adjusted to pH 6 by addition of 1 M hydrochloric acid and extracted with a mixture of dichloromethane and MeOH (9:1, 3×10 mL). After the combined organic layers had been dried over sodium sulfate, they were filtered, concentrated in vacuo, and purified using reversed-phase HPLC (Column: Agela Durashell C18, 5 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 17% to 47% B) to provide a mixture of stereoisomers at the cyclopropane center 52 and 53 (161 mg, 0.275 mmol, 32%) as a white solid. The stereoisomers at the cyclopropane center were separated using supercritical fluid chromatography [Column: Chiral Technologies Chiralcel OJ-H, 5 μm; Mobile phase: 3:2 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]. The first-eluting stereoisomer, obtained as a white solid, was designated as ENT-1 (52). Yield: 49.2 mg, 81.6 μmol, 30% for the separation. LCMS m/z 586.1 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.23 (s, 1H), 7.94 (d, 1H), 7.67-7.59 (m, 2H), 7.55-7.49 (m, 2H), 7.28 (dd, 1H), 6.22 (dd, 1H), 5.48 (AB quartet, 2H), 5.23-5.14 (m, 1H), 4.70 (dd, 1H), 4.63-4.54 (m, 2H), 4.41 (dt, 1H), 3.90-3.82 (m, 1H), 3.54-3.45 (m, 1H), 3.38 (ddd, 1H), 3.19 (ddd, 1H), 2.84-2.73 (m, 1H), 2.59-2.48 (m, 1H), 2.39 (dd, 1H), 1.88 (ddd, 1H), 1.69-1.62 (m, 1H), 1.56-1.46 (m, 2H), 1.38-1.30 (m, 1H), 1.24 (dd, 1H). The second-eluting stereoisomer, which was also isolated as a white solid, was designated as ENT-2 (53). Yield: 37.9 mg, 62.9 μmol, 23% for the separation. LCMS m/z 586.1 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.21 (d, 1H), 7.94 (dd, 1H), 7.66-7.59 (m, 2H), 7.55-7.50 (m, 2H), 7.26 (dd, 1H), 6.20 (dd, 1H), 5.47 (AB quartet, 2H), 5.29-5.20 (m, 1H), 4.70 (dd, 1H), 4.58-4.48 (m, 2H), 4.32 (dt, 1H), 3.87-3.78 (m, 1H), 3.51-3.43 (m, 1H), 3.37 (ddd, 1H), 3.17 (ddd, 1H), 2.82-2.71 (m, 1H), 2.53-2.41 (m, 2H), 1.84 (ddd, 1H), 1.65 (dd, 1H), 1.59-1.44 (m, 2H), 1.38-1.27 (m, 1H), 1.23 (dd, 1H). Examples 54 and 55 Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C92 (54) and Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C93 (55) Step 1. Synthesis of methyl 4-({[6-(6-bromo-3-fluoropyridin-2-yl)-6-azaspiro[2.5]oct-1-yl]carbonyl}amino)-3-{[(2S)-oxetan-2-ylmethyl]amino}benzoate (C89) Using the method described for synthesis of C73 in Example 33, P11 was reacted with P16. Chromatography on silica gel afforded C89, a mixture of stereoisomers at the cyclopropane, as an off-white solid. Yield: 1.31 g, 2.39 mmol, 79%. Step 2. Synthesis of methyl 2-[6-(6-bromo-3-fluoropyridin-2-yl)-6-azaspiro[2.5]oct-1-yl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C90) A solution of C89 (900 mg, 1.64 mmol) in acetic acid (18 mL) was stirred at 60° C. for 16 hours, whereupon it was diluted with water (200 mL), carefully neutralized by addition of sodium carbonate, and extracted with EtOAc (3×100 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure; the residue was combined with the product of a similar reaction carried out using C89 (400 mg, 0.731 mmol) and purified using chromatography on silica gel (Gradient: 0% to 60% EtOAc in petroleum ether) to afford C90, a mixture of stereoisomers at the cyclopropane, as a white solid. Combined yield: 823 mg, 1.55 mmol, 65%. Step 3. Synthesis of methyl 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C91) A mixture of C90 (400 mg, 0.756 mmol), 3-fluoro-4-(hydroxymethyl)benzonitrile (228 mg, 1.51 mmol), 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (Xantphos; 43.7 mg, 75.6 μmol), palladium(II) acetate (8.48 mg, 37.8 μmol), and cesium carbonate (739 mg, 2.27 mmol) in toluene (8 mL) was stirred at 100° C. for 2 hours. The reaction mixture was combined with a similar reaction carried out using C90 (100 mg, 0.189 mmol), diluted with dichloromethane (50 mL), and filtered. The filtrate was then concentrated in vacuo and purified using silica gel chromatography (Gradient: 0% to 45% EtOAc in petroleum ether) to afford C91, a mixture of stereoisomers at the cyclopropane, as a light yellow foamy solid. Combined yield: 385 mg, 0.642 mmol, 68%. Step 4. Isolation of methyl 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-1 (C92) and methyl 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-2 (C93) The stereoisomers at the cyclopropane comprising C91 (385 mg, 0.642 mmol) were separated using supercritical fluid chromatography [Column: Chiral Technologies Chiralpak AD, 5 μm; Mobile phase: 3:2 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]. The first-eluting stereoisomer, isolated as a white foam, was designated as ENT-1 (C92). Yield: 192 mg, 0.320 mmol, 50%. The second-eluting stereoisomer, also isolated as a white foam, was designated as ENT-2 (C93). Yield: 193 mg, 0.322 mmol, 50%. Step 5. Synthesis of Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C92. (54) To a solution of C92 (100 mg, 0.167 mmol) in a mixture of THF (1 mL) and MeOH (1 mL) was added aqueous lithium hydroxide solution (2 M; 0.834 mL, 1.67 mmol). After the reaction mixture had been stirred at 30° C. for 3 hours, it was carefully neutralized by addition of 12 M hydrochloric acid, diluted with water (30 mL), and extracted with dichloromethane (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo; the residue was combined with the product of a similar reaction carried out using C92 (90 mg, 0.15 mmol) and purified via reversed-phase HPLC (Column: Agela Durashell C18, 5 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 27% to 47% B) to provide 54 as a white solid. Combined yield: 39 mg, 65 μmol, 20%. LCMS m/z 586.1 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.24 (d, 1H), 7.95 (dd, 1H), 7.62 (d, 1H), 7.60 (dd, 1H), 7.51 (dd, 1H), 7.47 (dd, 1H), 7.28 (dd, 1H), 6.22 (dd, 1H), 5.39 (s, 2H), 5.26-5.17 (m, 1H), 4.72 (dd, component of ABX pattern, 1H), 4.66-4.55 (m, 2H), 4.43 (dt, 1H), 3.84-3.74 (m, 1H), 3.50-3.34 (m, 2H), 3.19 (ddd, 1H), 2.87-2.75 (m, 1H), 2.61-2.49 (m, 1H), 2.39 (dd, 1H), 1.95 (ddd, 1H), 1.65 (dd, 1H), 1.61-1.49 (m, 2H), 1.42-1.32 (m, 1H), 1.23 (dd, 1H). Step 6. Synthesis of Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3-fluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, from C93. (55) Conversion of C93 to 55 was carried out using the method described for synthesis of 54 from C92 in step 5 above. Compound 55 was obtained as a white solid. Yield: 59.4 mg, 98.5 μmol, 31%. LCMS m/z 586.1 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.23 (d, 1H), 7.95 (dd, 1H), 7.62 (d, 1H), 7.59 (dd, 1H), 7.51 (dd, 1H), 7.46 (dd, 1H), 7.26 (dd, 1H), 6.20 (dd, 1H), 5.38 (s, 2H), 5.30-5.22 (m, 1H), 4.71 (dd, 1H), 4.61-4.50 (m, 2H), 4.35 (dt, 1H), 3.80-3.72 (m, 1H), 3.47-3.33 (m, 2H), 3.17 (ddd, 1H), 2.83-2.72 (m, 1H), 2.54-2.46 (m, 1H), 2.44 (dd, 1H), 1.95-1.84 (m, 1H), 1.65 (dd, 1H), 1.62-1.47 (m, 2H), 1.39-1.31 (m, 1H), 1.22 (dd, 1H). Examples 56, 57, and 58 Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3,5-difluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (56), Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3,5-difluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-1 (57), and Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3,5-difluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, ENT-2 (58) Step 1. Synthesis of methyl 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3,5-difluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C94) To a solution of P12 (488 mg, 1.17 mmol) and O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (522 mg, 1.37 mmol) in N,N-dimethylformamide (8 mL) were added P16 (276 mg, 1.17 mmol) and N,N-diisopropylethylamine (515 mg, 3.99 mmol). The reaction mixture was stirred at 25° C. for 16 hours, whereupon it was diluted with water (35 mL) and extracted with EtOAc (3×30 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (2×25 mL), dried over sodium sulfate, filtered, and concentrated in vacuo; silica gel chromatography (Gradient: 0% to 60% EtOAc in petroleum ether) provided the intermediate amide (500 mg, 0.787 mmol, 67%) as a colorless gum. This material was dissolved in acetic acid and heated at 60° C. for 16 hours, whereupon it was combined with a similar reaction mixture (derived from P12, 52.2 mg, 0.125 mmol) and concentrated in vacuo. The residue was dissolved in dichloromethane (25 mL) and washed with saturated aqueous sodium bicarbonate solution (30 mL). The aqueous layer was extracted with dichloromethane (2×20 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Preparative thin-layer chromatography (Eluent: 1:1 EtOAc/petroleum ether) afforded C94, a mixture of stereoisomers at the cyclopropane, as a yellow gum. Combined yield: 280 mg, 0.453 mmol, 35%. Step 2. Synthesis of Ammonium 2-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]-3,5-difluoropyridin-2-yl}-6-azaspiro[2.5]oct-1-yl)-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (56) Hydrolysis of C94 was carried out using the method described for synthesis of P12 from C30 in Preparation P12 providing 56, a mixture of stereoisomers at the cyclopropane, as a solid. The crude product was purified via reversed-phase HPLC (Column: Agela Durashell C18, 5 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 23% to 53% B). The first-eluting stereoisomer, isolated as a white solid, was designated as ENT-1 (57). Yield: 11.0 mg, 17.7 μmol, 21%. LCMS m/z 604.1 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.22 (br s, 1H), 7.95 (dd, 1H), 7.66-7.60 (m, 2H), 7.54 (dd, 1H), 7.49 (dd, 1H), 7.38 (dd, 1H), 5.48 (s, 2H), 5.31-5.22 (m, 1H), 4.72 (dd, 1H), 4.62-4.52 (m, 2H), 4.36 (dt, 1H), 3.69-3.60 (m, 1H), 3.36-3.25 (m, 2H, assumed; largely obscured by solvent peak), 3.16-3.07 (m, 1H), 2.84-2.73 (m, 1H), 2.55-2.47 (m, 1H), 2.45 (dd, 1H), 1.99-1.86 (m, 1H), 1.65 (dd, 1H), 1.62-1.49 (m, 2H), 1.42-1.33 (m, 1H), 1.22 (dd, 1H). The second-eluting stereoisomer, also isolated as a white solid, was designated as ENT-2 (58). Yield: 23.5 mg, 39.0 μmol, 32%. LCMS m/z 604.1 [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.25 (br s, 1H), 7.95 (dd, 1H), 7.67-7.60 (m, 2H), 7.54 (br d, 1H), 7.49 (br d, 1H), 7.40 (dd, 1H), 5.49 (s, 2H), 5.26-5.18 (m, 1H), 4.74 (dd, component of ABX system; 1H), 4.66-4.57 (m, 2H), 4.44 (dt, 1H), 3.71-3.63 (m, 1H), 3.37-3.26 (m, 2H, assumed; largely obscured by solvent peak), 3.17-3.08 (m, 1H), 2.88-2.76 (m, 1H), 2.61-2.50 (m, 1H), 2.40 (dd, 1H), 2.03-1.93 (m, 1H), 1.65 (dd, 1H), 1.63-1.52 (m, 2H), 1.43-1.34 (m, 1H), 1.24 (dd, 1H). The compounds listed in Table 7 below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 7Structure and IUPAC name for Examples 59 to 61ExNo.StructureIUPAC Name592-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]- 3,5-difluoropyridin-2-yl}-6- azaspiro[2.5]oct-1-yl)-1-methyl-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt602-(6-{6-[(4-chloro-2-fluorobenzyl)oxy]- 3,5-difluoropyridin-2-yl}-6- azaspiro[2.5]oct-1-yl)-1-[(4-methyl-4H- 1,2,4-triazol-3-yl)methyl]-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt612-(6-{6-[(4-cyano-2-fluorobenzyl)oxy]- 3,5-difluoropyridin-2-yl}-6- azaspiro[2.5]oct-1-yl)-1-(1,3-oxazol-2- ylmethyl)-1H-benzimidazole-6- carboxylic acid, trifluoroacetate salt TABLE 7APhysicochemical data for Examples 59 to 61Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR59548.1; 2.91 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobilephase A: water containing 0.1% formic acid; Mobile phase B: acetonitrile; Gradient:1.0% to 5% B over 0.6 minutes, then 5% to 100% B over 3.4 minutes; Flow rate: 0.8mL/minute)60638.3♦; 3.17 minutes (Analytical conditions identical to those used for Ex. 59)61615.1; 3.19 minutes (Analytical conditions identical to those used for Ex. 59) Example 62 2-[(4-{2-[(4-Chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic Acid (62) Step 1. Synthesis of methyl 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C95) A mixture of P13 (14.34 g, 21.5 mmol), P17 (6.34 g, 21.5 mmol), and potassium carbonate (19.3 g, 140 mmol) in 1,4-dioxane (144 mL) was stirred at 58° C. After 1.5 hours, additional potassium carbonate (5.0 g, 36 mmol) was introduced; 30 minutes later, acetonitrile (50 mL) was added, and after another 50 minutes, more acetonitrile (25 mL) was added. The reaction temperature was then increased to 63° C. overnight, whereupon it was removed from the heat and slowly diluted with water (300 mL). The resulting mixture was extracted with EtOAc (3×200 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 10% MeOH in dichloromethane) provided C95 as an off-white solid. Yield: 10.7 g, 18.4 mmol, 86%. Step 2. Synthesis of 2-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}piperidin-1-yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic Acid (62) A mixture of C95 (9.33 g, 16.1 mmol), MeOH (100 mL), and aqueous sodium hydroxide solution (1.0 M; 40.2 mL, 40.2 mmol) was treated with sufficient THF (12 mL) to form a solution; the reaction mixture was then stirred at room temperature overnight. LCMS analysis at this point indicated conversion to the product: LCMS m/z 566.3 [M+H]+. The reaction mixture was concentrated in vacuo to a final volume of approximately 50 mL, and then treated with 10% aqueous citric acid solution to a pH of 6. After extraction with a mixture of 2-propanol and dichloromethane (1:4; 3×75 mL), the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting white solid was diluted with cyclohexane and dichloromethane and then concentrated in vacuo to remove residual 2-propanol, affording 62. Yield: 9.25 g, 16 mmol, quantitative.1H NMR (400 MHz, DMSO-d6) δ 8.50 (d, 1H), 8.26 (br s, 1H), 7.80 (dd, 1H), 7.63 (d, 1H), 7.57 (dd, 1H), 7.48 (dd, 1H), 7.32 (dd, 1H), 7.09 (d, 1H), 5.39 (s, 2H), 5.13-5.04 (m, 1H), 4.80 (dd, 1H), 4.66 (dd, 1H), 4.52-4.44 (m, 1H), 4.37 (dt, 1H), 3.87 (AB quartet, 2H), 2.99 (br d, 1H), 2.86 (br d, 1H), 2.76-2.58 (m, 2H), 2.47-2.37 (m, 1H), 2.29-2.12 (m, 2H), 1.88-1.77 (m, 2H), 1.77-1.59 (m, 2H). The compounds listed in Table 8 below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 8Structure and IUPAC name for Examples 63 to 68Ex.No.StructureIUPAC Name632-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4- yl}piperidin-1-yl)methyl]-1-[(1-ethyl- 1H-imidazol-5-yl)methyl]-1H- benzimidazole-6-carboxylic acid642-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4- yl}piperidin-1-yl)methyl]-1-[(1-ethyl- 1H-1,2,3-triazol-5-yl)methyl]-1H- benzimidazole-6-carboxylic acid652-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4- yl}piperidin-1-yl)methyl]-1-(2- methoxyethyl)-1H-benzimidazole-6- carboxylic acid, hydrochloride salt662-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4- yl}piperidin-1-yl)methyl]-1-[(1- methyl-1H-imidazol-4-yl)methyl]- 1H-benzimidazole-6-carboxylic acid671ammonium 2-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4- yl}piperidin-l-yl)methyl]-3-[(2S)- oxetan-2-ylmethyl]-3H-imidazo[4,5- b]pyridine-5-carboxylate682-[(4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4- yl}piperidin-1-yl)methyl]-1-(1,3- oxazol-2-ylmethyl)-1H- benzimidazole-6-carboxylic acid, trifluoroacetate salt TABLE 8APhysicochemical data for Examples 63 to 68Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR63604; 2.64 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobile phaseA: 0.0375% trifluoroacetic acid in water; Mobile phase B: 0.01875% trifluoroaceticacid in acetonitrile; Gradient: 1% to 5% B over 0.6 minutes; 5% to 100% B over 3.4minutes; Flow rate: 0.8 mL/minute)64605; 2.22 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobile phaseA: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 5%B for 0.5 minutes; 5% to 100% B over 2.9 minutes; 100% B for 0.8 minutes; Flowrate: 0.8 mL/minute65554.2♦;1H NMR (600 MHz, DMSO-d6), characteristic peaks: δ 8.58 (s, 1H), 8.31 (s,1H), 7.89 (d, 1H), 7.78 (d, 1H), 7.65-7.57 (m, 1H), 7.50 (d, 1H), 7.34 (d, 1H), 7.15(s, 1H), 5.42 (s, 2H), 4.89-4.72 (br m, 2H), 4.70-4.59 (m, 2H), 3.88-3.71 (br m,2H), 3.70-3.57 (b m, 3H), 3.21 (s, 3H), 3.07-2.93 (br m, 2H), 2.24-2.01 (br m,4H)66590; 2.80 minutes (Analytical conditions identical to those used for Ex. 63671566.9♦; 1.81 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 50 mm, 3μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes, then40% B for 1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.)68577.0♦; 2.55 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobilephase A: water containing 0.1% formic acid; Mobile phase B: acetonitrile; Gradient:1.0% to 5% B over 0.6 minutes, then 5% to 100% B over 3.4 minutes; Flow rate: 0.8mL/minute)Table 8/8A:1Treatment of methyl 5-{[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}piperidin-1-yl)acetyl]amino}-6-{[(2S)-oxetan-2-ylmethyl]amino}pyridine-2-carboxylate with aqueous sodium hydroxide solution in 1,4-dioxane at elevated temperature served to effect both ring closure and ester hydrolysis, delivering Example 67 after purification. Example 69 Ammonium 2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (69) Step 1. Synthesis of 2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoro-4-[(3S)-3-methylpiperazin-1-yl]pyrimidine, Trifluoroacetate Salt (C96) Trifluoroacetic acid (36 mL) was added to a solution of P14 (7.62 g, 16.8 mmol) in dichloromethane (75 mL) and the reaction mixture was stirred at 18° C. for 1 hour, whereupon it was combined with a similar reaction carried out using P14 (3.09 g, 6.79 mmol). Concentration in vacuo afforded C96 as a brown oil. Combined yield: 11.0 g, 23.5 mmol, quantitative. Step 2. Synthesis of methyl 2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C97) A suspension of C96 (11.0 g, 23.5 mmol), P17 (6.94 g, 23.5 mmol), and potassium carbonate (16.3 g, 118 mmol) in acetonitrile (150 mL) was stirred at 50° C. for 19 hours, whereupon it was partitioned between water (300 mL) and EtOAc (300 mL). After extraction of the aqueous layer with EtOAc (2×200 mL), the combined organic layers were concentrated in vacuo and purified via silica gel chromatography (Gradient: 35% to 60% EtOAc in petroleum ether) to provide C97 as a white gum. Yield: 11.1 g, 18.1 mmol, 77%. Step 3. Synthesis of Ammonium 2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]-5-fluoropyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (69) Aqueous sodium hydroxide solution (2 M; 22 mL, 44 mmol) was added to a solution of C97 (5.00 g, 8.16 mmol) in a mixture of MeOH (73 mL) and THF (73 mL). After the reaction mixture had been stirred at 18° C. for 16 hours, it was heated to 45° C. for 3 hours, whereupon it was neutralized to pH 7 by addition of 1 M hydrochloric acid and concentrated in vacuo. The residue was diluted with water (100 mL) and extracted with a mixture of dichloromethane and MeOH (10:1, 4×100 mL); the combined organic layers were concentrated under reduced pressure and then purified using reversed-phase HPLC (Column: Phenomenex Gemini C18, 10 μm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 20% to 40% B) to afford 69 as a white solid. Yield: 2.75 g, 4.59 mmol, 56%. LCMS m/z 598.9♦ [M+H]+.1H NMR (400 MHz, Methanol-d4) δ 8.23 (dd, 1H), 7.96 (dd, 1H), 7.89 (d, 1H), 7.62 (dd, 1H), 7.46 (dd, 1H), 7.24-7.17 (m, 2H), 5.33 (s, 2H), 5.33-5.26 (m, 1H), 4.9-4.84 (m, 1H, assumed; partially obscured by water peak), 4.75 (dd, 1H), 4.60 (ddd, 1H), 4.49 (d, 1H), 4.33 (dt, 1H), 4.14-4.07 (m, 1H), 4.02 (br d, 1H), 3.70 (d, 1H), 3.52 (ddd, 1H), 3.36-3.29 (m, 1H, assumed; partially obscured by solvent peak), 2.81-2.70 (m, 2H), 2.72-2.63 (m, 1H), 2.54-2.44 (m, 1H), 2.39 (ddd, 1H), 1.18 (d, 3H). The compounds listed in Table 9 below below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 9Structure and IUPAC name for Examples 70 to 72Ex.No.StructureIUPAC Name70ammonium 2-{[(2S)-4-{2-[(4- cyanobenzyl)oxy]-5-fluoropyrimidin-4- yl}-2-methylpiperazin-1-yl]methyl}-1- (3,3,3-trifluoropropyl)-1H- benzimidazole-6-carboxylate712-{[(2S)-4-{2-[(4-chloro-2- fluorobenzyl)oxy]-5-fluoropyrimidin-4- yl}-2-methylpiperazin-1-yl]methyl}-1- [(2S)-tetrahydrofuran-2-ylmethyl]-1H- benzimidazole-6-carboxylic acid722-[(4-{2-[(4-chloro-2-fluorobenzyl)oxy]- 5-fluoropyrimidin-4-yl}piperazin-1- yl)methyl]-1-[(2S)-oxetan-2-ylmethyl]- 1H-benzimidazole-6-carboxylic acid TABLE 9APhysicochemical data for Examples 70 to 72Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR70598.3; 2.41 minutes (Column: Waters Atlantis dC18, 4.6 × 50 mm, 5 μm; Mobile phaseA: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acidin acetonitrile (v/v); Gradient: 5.0% to 95% B, linear over 4.0 minutes; Flow rate: 2mL/minute)71613; 2.36 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobile phase A:0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 5% B for0.5 minutes; 5% to 100% B over 2.9 minutes; 100% B for 0.8 minutes; Flow rate: 0.8mL/minute)72585.2♦; 1.87 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 50 mm, 3μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine in ethanol;Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes, then 40% B for1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.) Example 73 2-{[(2S)-4-{2-[(4-Chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic Acid (73) Step 1. Synthesis of methyl 2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate (C98) This reaction was carried out in two identical batches. A mixture of P15 (26.3 g 38.6 mmol) and potassium carbonate (31.0 g, 225 mmol) in acetone (190 mL) was stirred at room temperature for 5 minutes, placed in a 70° C. oil bath for 5 minutes, removed from the heat, and stirred for another 5 minutes. To this mixture was added P17 (11.0 g, 37.3 mmol) and the reaction mixture was heated at reflux for 24 hours. After cooling to room temperature, the reaction mixture was diluted with water (350 mL), stirred at room temperature for 1 hour, and extracted with EtOAc (2×250 mL). The EtOAc extracts from both reactions were combined, dried over sodium sulfate, filtered, and concentrated in vacuo; repeated chromatography on silica gel (Gradient: 0% to 10% MeOH in dichloromethane) provided C98 as a white solid. Combined yield: 29.2 g, 49.1 mmol, 66%. Step 2. Synthesis of 2-{[(2S)-4-{2-[(4-chloro-2-fluorobenzyl)oxy]pyrimidin-4-yl}-2-methylpiperazin-1-yl]methyl}-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylic Acid (73) Aqueous sodium hydroxide solution (1 M; 110 mL, 110 mmol) was added to a 0° C. solution of C98 (18.8 g, 31.6 mmol) in MeOH (220 mL); THF (40 mL) was then added until the mixture became clear. After the reaction mixture had been stirred at room temperature overnight, LCMS analysis indicated conversion to the product: LCMS m/z 581.2♦ [M+H]+. The reaction mixture was concentrated in vacuo to a volume of approximately 125 mL, diluted to 150 mL by addition of water, and slowly adjusted to pH 7 with 10% aqueous citric acid solution. The resulting slurry was filtered, and the filter cake was washed with copious water, affording 73 as a white solid. Yield: 18.2 g, 31.3 mmol, 99%.1H NMR (400 MHz, Methanol-d4) δ 8.30 (br s, 1H), 7.98 (dd, 1H), 7.94 (d, 1H), 7.67 (d, 1H), 7.49 (dd, 1H), 7.26-7.18 (m, 2H), 6.40 (d, 1H), 5.38 (s, 2H), 5.34-5.26 (m, 1H), 4.95-4.85 (m, 1H, assumed; partially obscured by water peak), 4.78 (dd, 1H), 4.62 (td, 1H), 4.50 (d, 1H), 4.35 (dt, 1H), 4.10-3.97 (br m, 1H), 3.97-3.83 (br m, 1H), 3.72 (d, 1H), 3.45-3.3 (m, 1H, assumed; partially obscured by solvent peak), 3.23 (dd, 1H), 2.82-2.70 (m, 2H), 2.70-2.59 (m, 1H), 2.53-2.42 (m, 1H), 2.36 (ddd, 1H), 1.19 (d, 3H). The compounds listed in Table 10 below were synthesized via procedures analogous to those described herein for syntheses of Examples and Preparations by using appropriate starting materials, which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described herein for other intermediates. The compounds were purified using methods well-known to those skilled in the art and may include silica gel chromatography, HPLC, or precipitation from the reaction mixture. TABLE 10Structure and IUPAC name for Examples 74 to 78Ex.No.StructureIUPAC Name7412-{[(3R)-4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4-yl}-3- methylpiperazin-1-yl]methyl}-1-(2- methoxyethyl)-1H-benzimidazole-6- carboxylic acid, trifluoroacetate salt75ammonium 2-{[(2R)-4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4-yl}-2- methylpiperazin-1-yl]methyl}-1-[(2S)- oxetan-2-ylmethyl]-1H-benzimidazole-6- carboxylate76ammonium 2-{[(2S,5R)-4-{2-[(4-chloro- 2-fluorobenzyl)oxy]pyrimidin-4-yl}-2,5- dimethylpiperazin-1-yl]methyl}-1-[(2S)- oxetan-2-ylmethyl]-1H-benzimidazole-6- carboxylate772-{[(2S)-4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4-yl}-2- methylpiperazin-1-yl]methyl}-3-[(2S)- oxetan-2-ylmethyl]-3H-imidazo[4,5- b]pyridine-5-carboxylic acid782-{[(2S)-4-{2-[(4-chloro-2- fluorobenzyl)oxy]pyrimidin-4-yl}-2- methylpiperazin-1-yl]methyl}-1-(1,3- oxazol-2-ylmethyl)-1H-benzimidazole-6- carboxylic acid, trifluoroacetate salt TABLE 10APhysicochemical data for Examples 74 to 78Ex. No.Mass spectrum, observed ion m/z [M + H]+; HPLC retention time or1H NMR741569; 2.16 minutes (Column: Waters XBridge C18, 2.1 × 50 mm, 5 μm; Mobilephase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile;Gradient: 5% B for 0.5 minutes; 5% to 100% B over 2.9 minutes; 100% B for 0.8minutes; Flow rate: 0.8 mL/minute)75581.0♦; 2.13 minutes (Column: Chiral Technologies Chiralpak AD-3, 4.6 × 50 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes, then40% B for 1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.)76595.0♦; 2.97 minutes (Column: Chiral Technologies Chiralcel OJ-3, 4.6 × 100 mm,3 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inethanol; Gradient: 5% to 40% B over 4.5 minutes, then 40% B for 2.5 minutes;Flow rate: 2.8 mL/minute; Column temperature: 40° C.)77582.1♦; 1.18 minutes (Column: Chiral Technologies Chiralcel OJ-3, 4.6 × 50 mm, 3μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.05% diethylamine inMeOH; Gradient: 5% B for 0.2 minutes, then 5% to 40% B over 1.4 minutes, then40% B for 1.05 minutes; Flow rate: 4 mL/minute; Column temperature: 40° C.)78592.2♦; 1.31 minutes (Analytical conditions identical to those used for Ex. 77)Table 10/10A,1Reaction of P8 with tert-butyl (3R)-3-methylpiperazine-1-carboxylate was carried out using cesium fluoride and N,N-diisopropylethylamine in acetonitrile at ≥100° C. Example 79 and 80 Ammonium 2-[(4-{6-[(4-chloro-2-fluorobenzyl)oxy]pyridin-2-yl}cyclohexyl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, Isomer 1 (79) and Ammonium 2-[(4-{6-[(4-chloro-2-fluorobenzyl)oxy]pyridin-2-yl}cyclohexyl)methyl]-1-[(2S)-oxetan-2-ylmethyl]-1H-benzimidazole-6-carboxylate, Isomer 2 (80) Compound C99, a mixture of cis and trans isomers at the cyclohexyl group, was prepared from P34 and P16 in a manner analogous to the preparation of Example 1 from intermediate P1. The isomeric mixture was subjected to separation by HPLC with the retention times shown below. Column: Chiral Technologies Chiralpak AD-H, 21×250 mm, 5 μm; Mobile phase A: carbon dioxide; Mobile phase B: 0.2% ammonium hydroxide in methanol; Eluent: 30% B; Flow rate: 75 mL/minute; Column temperature: 40° C.). The first eluting isomer was assigned as isomer 1 (79). Retention time 1.60 min, LCMS m/z 564.5 [M+H]+. The second eluting isomer was assigned as isomer 2 (80). Retention time 1.86 min, LCMS m/z 564.4 [M+H]+. CHO GLP-1R Clone H6—Assay 1 GLP-1R-mediated agonist activity was determined with a cell-based functional assay utilizing an HTRF (Homogeneous Time-Resolved Fluorescence) cAMP detection kit (cAMP HI Range Assay Kit; CisBio cat #62AM6PEJ) that measures cAMP levels in the cell. The method is a competitive immunoassay between native cAMP produced by the cells and exogenous cAMP labeled with the dye d2. The tracer binding is visualized by a mAb anti-cAMP labeled with Cryptate. The specific signal (i.e. energy transfer) is inversely proportional to the concentration of cAMP in either standard or experimental sample. The human GLP-1R coding sequence (NCBI Reference Sequence NP_002053.3, including naturally-occurring variant Gly168Ser) was subcloned into pcDNA3 (Invitrogen) and a cell line stably expressing the receptor was isolated (designated Clone H6). Saturation binding analyses (filtration assay procedure) using125I-GLP-17-36(Perkin Elmer) showed that plasma membranes derived from this cell line express a high GLP-1R density (Kd: 0.4 nM, Bmax: 1900 fmol/mg protein). Cells were removed from cryopreservation, re-suspended in 40 mL of Dulbecco's Phosphate Buffered Saline (DPBS—Lonza Cat #17-512Q) and centrifuged at 800×g for 5 min at 22° C. The cell pellet was then re-suspended in 10 mL of growth medium [DMEM/F12 1:1 Mixture with HEPES, L-Gln, 500 mL (DMEM/F12 Lonza Cat #12-719F), 10% heat inactivated fetal bovine serum (Gibco Cat #16140-071), 5 mL of 100× Pen-Strep (Gibco Cat #15140-122), 5 mL of 100× L-Glutamine (Gibco Cat #25030-081) and 500 μg/mL Geneticin (G418) (Invitrogen #10131035)]. A 1 mL sample of the cell suspension in growth media was counted on a Becton Dickinson ViCell to determine cell viability and cell count per mL. The remaining cell suspension was then adjusted with growth media to deliver 2000 viable cells per well using a Matrix Combi Multidrop reagent dispenser, and the cells were dispensed into a white 384 well tissue culture treated assay plate (Corning 3570). The assay plate was then incubated for 48 hours at 37° C. in a humidified environment in 5% carbon dioxide. Varying concentrations of each compound to be tested (in DMSO) were diluted in assay buffer (HBSS with Calcium/Magnesium (Lonza/BioWhittaker cat #10-527F)/0.1% BSA (Sigma Aldrich cat #A7409-1L)/20 mM HEPES (Lonza/BioWhittaker cat #17-737E) containing 100 μM 3-isobutyl-1-methylxanthin (IBMX; Sigma cat #15879). The final DMSO concentration is 1%. After 48 hours, the growth media was removed from the assay plate wells, and the cells were treated with 20 μL of the serially diluted compound in assay buffer for 30 minutes at 37° C. in a humidified environment in 5% carbon dioxide. Following the 30 minute incubation, 10 μL of labeled d2 cAMP and 10 μL of anti-cAMP antibody (both diluted 1:20 in cell lysis buffer; as described in the manufacturer's assay protocol) were added to each well of the assay plate. The plates were then incubated at room temperature and after 60 minutes, changes in the HTRF signal were read with an Envision 2104 multi-label plate reader using excitation of 330 nm and emissions of 615 and 665 nm. Raw data were converted to nM cAMP by interpolation from a cAMP standard curve (as described in the manufacturer's assay protocol) and the percent effect was determined relative to a saturating concentration of the full agonist GLP-17-36(1 μM) included on each plate. EC50determinations were made from agonist dose-response curves analyzed with a curve fitting program using a 4-parameter logistic dose response equation. CHO GLP-1R Clone C6—Assay 2 GLP-1R-mediated agonist activity was determined with a cell-based functional assay utilizing an HTRF (Homogeneous Time-Resolved Fluorescence) cAMP detection kit (cAMP HI Range Assay Kit; Cis Bio cat #62AM6PEJ) that measures cAMP levels in the cell. The method is a competitive immunoassay between native cAMP produced by the cells and exogenous cAMP labeled with the dye d2. The tracer binding is visualized by a mAb anti-cAMP labeled with Cryptate. The specific signal (i.e. energy transfer) is inversely proportional to the concentration of cAMP in either a standard or an experimental sample. The human GLP-1R coding sequence (NCBI Reference Sequence NP_002053.3, including naturally-occurring variant Leu260Phe) was subcloned into pcDNA5-FRT-TO and a clonal CHO cell line stably expressing a low receptor density was isolated using the Flp-In™ T-Rex™ System, as described by the manufacturer (ThermoFisher). Saturation binding analyses (filtration assay procedure) using125I-GLP-1 (Perkin Elmer) showed that plasma membranes derived from this cell line (designated clone C6) express a low GLP-1R density (Kd: 0.3 nM, Bmax: 240 fmol/mg protein), relative to the clone H6 cell line. Cells were removed from cryopreservation, re-suspended in 40 mL of Dulbecco's Phosphate Buffered Saline (DPBS—Lonza Cat #17-512Q) and centrifuged at 800×g for 5 min at 22° C. The DPBS was aspirated, and the cell pellet was re-suspended in 10 mL of complete growth medium (DMEM:F12 1:1 Mixture with HEPES, L-Gln, 500 mL (DMEM/F12 Lonza Cat #12-719F), 10% heat inactivated fetal bovine serum (Gibco Cat #16140-071), 5 mL of 100× Pen-Strep (Gibco Cat #15140-122), 5 mL of 100× L-Glutamine (Gibco Cat #25030-081), 700 μg/mL Hygromycin (Invitrogen Cat #10687010) and 15 μg/mL Blasticidin (Gibco Cat #R21001). A 1 mL sample of the cell suspension in growth media was counted on a Becton Dickinson ViCell to determine cell viability and cell count per mL. The remaining cell suspension was then adjusted with growth media to deliver 1600 viable cells per well using a Matrix Combi Multidrop reagent dispenser, and the cells were dispensed into a white 384 well tissue culture treated assay plate (Corning 3570). The assay plate was then incubated for 48 h at 37° C. in a humidified environment (95% O2, 5% CO2) Varying concentrations of each compound to be tested (in DMSO) were diluted in assay buffer [HBSS with Calcium/Magnesium (Lonza/BioWhittaker cat #10-527F)/0.1% BSA (Sigma Aldrich cat #A7409-1L)/20 mM HEPES (Lonza/BioWhittaker cat #17-737E)] containing 100 μM 3-isobutyl-1-methylxanthin (IBMX; Sigma cat #15879). The final DMSO concentration in the compound/assay buffer mixture is 1%. After 48 h, the growth media was removed from the assay plate wells, and the cells were treated with 20 μL of the serially diluted compound in assay buffer for 30 min at 37° C. in a humidified environment (95% O2, 5% CO2). Following the 30 min incubation, 10 μL of labeled d2 cAMP and 10 μL of anti-cAMP antibody (both diluted 1:20 in cell lysis buffer; as described in the manufacturer's assay protocol) were added to each well of the assay plate. The plates were then incubated at room temperature and after 60 minutes, changes in the HTRF signal were read with an Envision 2104 multi-label plate reader using excitation of 330 nm and emissions of 615 and 665 nm. Raw data were converted to nM cAMP by interpolation from a cAMP standard curve (as described in the manufacturer's assay protocol) and the percent effect was determined relative to a saturating concentration of the full agonist GLP-1 (1 μM) included on each plate. EC50determinations were made from agonist dose response curves analyzed with a curve fitting program using a 4-parameter logistic dose response equation. In Table 11, assay data are presented to two (2) significant figures as the geometric mean (EC50s) and arithmetic mean (Emax) based on the number of replicates listed (Number). A blank cell means there was no data for that Example or the Emax was not calculated. TABLE 11Biological activity for Examples 1-80.Assay 1Assay 1Assay 2Assay 2ExampleEC50EmaxAssay 1EC50EmaxAssay 2Number(nM)(%)Number(nM)(%)Number11.08452095723.27983881330.587949.088543.0776409835*2.28254295664.56838483478.981319081386.0723230763928773700813109.77632201003111.9834491003124.7784110933136.080427091314147735401203150.9681421913160.99873181304176.0863150913181.8953591003195.390842946200.348056.19142114733370913222.8855238232341894450944242.076333893256.380473913265.186438863270.8486416855286.894315098329180065430140793280011043130065332>200001333.193384110334190092335330853130001003360.4883315905379.3883190923381.175635917391.6903299534015077320001003417.68441309634234008934313753250703446.5813120773451272323083346>970010024786924170010014826005634918753290873500.717831980351470714>2000015210074364001003530.578331490354637633400933550.338335.8825560.388034.2903570.308834.19855843743596.8803120853605.09131101103611.16832674362**4.48681109812630.989211097364249631001004658885317001103661701104673.3903521203688.3773310923690.9189318110570700793718.982416090372*6.0967120130473***2.484789100147450086311000963752188337010037612883180100377*4.410031101103781779344081379>120004>20000280129841601003*Tested as ammonium salt and free acid**Tested as ammonium and tris salts***Tested as ammonium and tris salts, and free acid All patents, patent applications and references referred to herein are hereby incorporated by reference in their entirety.
294,867
11858917
DETAILED DESCRIPTION OF THE INVENTION The following terms are used to describe the present invention. In instances where a term is not specifically defined herein, that term is given an art-recognized meaning by those of ordinary skill applying that term in context to its use in describing the present invention. Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise (such as in the case of a group containing a number of carbon atoms in which case each carbon atom number falling within the range is provided), between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges is also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention. The term “compound”, as used herein, unless otherwise indicated, refers to any specific chemical compound disclosed herein and includes tautomers, regioisomers, geometric isomers, and where applicable, optical isomers (enantiomers) thereof, as well as pharmaceutically acceptable salts and derivatives (including prodrug forms) thereof. Within its use in context, the term compound generally refers to a single compound, but also may include other compounds such as stereoisomers, regioisomers and/or optical isomers (including racemic mixtures) as well as specific enantiomers or enantiomerically enriched mixtures of disclosed compounds. The term also refers, in context to prodrug forms of compounds which have been modified to facilitate the administration and delivery of compounds to a site of activity. It is noted that in describing the present compounds, numerous substituents, linkers and connector molecules and variables associated with same, among others, are described. It is understood by those of ordinary skill that molecules which are described herein are stable compounds as generally described hereunder. “Alkyl” refers to a fully saturated monovalent radical containing carbon and hydrogen, and which may be cyclic, branched or a straight chain. Examples of alkyl groups are methyl, ethyl, n-butyl, sec-butyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, isopropyl, 2-methylpropyl, cyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl, cyclopentylethyl, cyclohexylethyl and cyclohexyl, among others. Preferred alkyl groups are C1-C6or C1-C3alkyl groups. “Aryl” or “aromatic”, in context, refers to a substituted (as otherwise described herein) or unsubstituted monovalent aromatic radical having a single ring (e.g., benzene or phenyl) or condensed rings (e.g., naphthyl, anthracenyl, phenanthrenyl, etc.) and can be bound to the compound according to the present invention at any available stable position on the ring(s) or as otherwise indicated in the chemical structure presented. Other examples of aryl groups, in context, may include heterocyclic aromatic ring systems “heteroaryl” groups having one or more nitrogen, oxygen, or sulfur atoms in the ring (moncyclic) such as imidazole, furyl, pyrrole, furanyl, thiene, thiazole, pyridine, pyrimidine, pyrazine, triazole, oxazole or fused ring systems such as indole, quinoline, etc., among others, which may be optionally substituted as described above. Among the heteroaryl groups which may be mentioned include nitrogen-containing heteroaryl groups such as pyrrole, pyridine, pyridone, pyridazine, pyrimidine, pyrazine, pyrazole, imidazole, triazole, triazine, tetrazole, indole, isoindole, indolizine, purine, indazole, quinoline, isoquinoline, quinolizine, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, imidazopyridine, imidazotriazine, pyrazinopyridazine, acridine, phenanthridine, carbazole, carbazoline, perimidine, phenanthroline, phenacene, oxadiazole, benzimidazole, pyrrolopyridine, pyrrolopyrimidine and pyridopyrimidine; sulfur-containing aromatic heterocycles such as thiophene and benzothiophene; oxygen-containing aromatic heterocycles such as furan, pyran, cyclopentapyran, benzofuran and isobenzofuran; and aromatic heterocycles comprising 2 or more hetero atoms selected from among nitrogen, sulfur and oxygen, such as thiazole, thiadizole, isothiazole, benzoxazole, benzothiazole, benzothiadiazole, phenothiazine, isoxazole, furazan, phenoxazine, pyrazoloxazole, imidazothiazole, thienofuran, furopyrrole, pyridoxazine, furopyridine, furopyrimidine, thienopyrimidine and oxazole, among others. Alternative aryl and heteroaryl groups according to the present invention preferably include, for example, phenyl, naphthyl, pyridyl (2-, 3- or 4-pyridyl group), thiazolyl (2-, 4- or 5-thiazole), isothiazolyl, oxazolyl (2-, 4- or 5-oxazole), isoxazolyl, furanyl (2- or 3-furan) or thiophenyl (2- or 3-thiophene). Monocyclic and bicyclic aryl and heteroayl groups are as otherwise described herein. In alternative embodiments, preferred heteroaryl groups are 5- or 6-membered aryl or heteroaryl group according to the chemical structure: Where W is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(CH2O)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(CH2O)nO—(C1-C6alkyl), —(CH2O)nC(O)—(C1-C6alkyl), —(CH2O)nNHC(O)—R1, —(CH2O)nC(O)—NR1R2, NO2, CN, halogen (F, Cl, Br, I, preferably F or Cl) or a monocyclic aryl or heteroaryl group which itself is optionally substituted (especially an optionally substituted benzoyl or benzyl group); W′ is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl) or halogen (preferably F or Cl); and Y is O, S or N—R, where R is H or a C1-C3alkyl group. In still other embodiments, preferred aryl or heteroaryl groups include those which are substituted according to the chemical structures: Where W2is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(CH2O)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(CH2O)nO—(C1-C6alkyl), —(CH2O)nC(O)—(C1-C6alkyl), —(CH2O)nNHC(O)—R1, —(CH2O)nC(O)—NR1R2, NO2, CN or halogen (preferably F or Cl); X is a group —NH—, —NHC(O)—, —O—, —(CH2)m—, —S—, —S(O)—, SO2— or —NH—C(O)—NH—; and Y is O, S or N—R, where R is H or a C1-C3alkyl group. The term “substituted” shall mean substituted at a carbon (or nitrogen) position within context, hydroxyl, carboxyl, cyano (C═N), nitro (NO2), halogen (preferably, 1, 2 or 3 halogens, especially on an alkyl, especially a methyl group such as a trifluoromethyl), alkyl group (preferably, C1-C10, more preferably, C1-C6), aryl (especially phenyl and substituted phenyl for example benzyl or benzoyl), alkoxy group (preferably, C1-C6alkyl or aryl, including phenyl and substituted phenyl), ester (preferably, C1-C6alkyl or aryl) including alkylene ester (such that attachment is on the alkylene group, rather than at the ester function which is preferably substituted with a C1-C6alkyl or aryl group), preferably, C1-C6alkyl or aryl, halogen (preferably, F or Cl), nitro or amine (including a five- or six-membered cyclic alkylene amine, further including a C1-C6alkyl amine or C1-C6dialkyl amine which alkyl groups may be substituted with one or two hydroxyl groups), amido, which is preferably substituted with one or two C1-C6alkyl groups (including a carboxamide which is substituted with one or two C1-C6alkyl groups), alkanol (preferably, C1-C6alkyl or aryl), or alkanoic acid (preferably, C1-C6alkyl or aryl). The term “substituted” shall also mean within its context of use alkyl, alkoxy, halogen, amido, carboxamido, keto, carboxy, ester, keto, nitro, cyano and amine (especially including mono- or di-C1-C6alkyl substituted amines which may be optionally substituted with one or two hydroxyl groups). In certain embodiments preferred substituents will include for example, —NH—, —NHC(O)—, —O—, —(CH2)m— (m and n are at least 1 as otherwise described herein), —S—, —S(O)—, SO2— or —NH—C(O)—NH—, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(CH2O)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(OCH2)nO—(C1-C6alkyl), —(OCH2)nC(O)—(C1-C6alkyl), —(OCH2)nNHC(O)—R1, —(CH2O)nC(O)—NR1R2, NO2, CN or halogen (F, Cl, Br, I, preferably F or Cl), depending on the context of the use of the substituent. Any substitutable position in a compound according to the present invention may be substituted in the present invention, but no more than 3, more preferably no more than 2 substituents (in some instances only 1 or no substituents) is present on a ring. Preferably, the term “unsubstituted” shall mean substituted with one or more H atoms. The term “patient” or “subject” is used throughout the specification within context to describe an animal, generally a mammal and preferably a human, to whom treatment, including prophylactic treatment (prophylaxis), with the compositions according to the present invention is provided. For treatment of those infections, conditions or disease states which are specific for a specific animal such as a human patient or a patient of a particular gender, such as a human male patient, the term patient refers to that specific animal. Compounds according to the present invention are useful for treating and/or reducing the likelihood of HIV infections or the secondary effects of HIV infections, especially including AIDS and/or ARC. The term “effective” is used herein, unless otherwise indicated, to describe an amount of a compound or composition which, in context, is used to produce or effect an intended result, whether that result relates to the inhibition of the effects of a toxicant on a subject or the treatment of a subject for secondary conditions, disease states or manifestations of exposure to toxicants as otherwise described herein. This term subsumes all other effective amount or effective concentration terms (including the term “therapeutically effective”) which are otherwise described in the present application. The terms “treat”, “treating”, and “treatment”, etc., as used herein, refer to any action providing a benefit to a patient at risk for HIV infection or having an HIV infection, including improvement in the condition through lessening or suppression of titers of HIV or at least one symptom of HIV, prevention or delay in progression of the disease, prevention or delay in the onset of disease states or conditions which occur secondary to HIV, including AIDS or ARC, among others. Treatment, as used herein, encompasses both prophylactic and therapeutic treatment. The term “prophylactic” when used, means to reduce the likelihood of an occurrence or the severity of an occurrence within the context of the treatment of HIV, as otherwise described hereinabove. The term “human immunodeficieincy virus” or “HIV” shall be used to describe human immunodeficiency viruses 1 and 2 (HIV-1 and HIV-2), the growth or replication of which may be inhibited or disease states of which may be treated using one or more methods according to the present invention. Viruses which may be treated according to the present invention include, for example, human immunodeficiency viruses 1 and 2 (HIV-1 and HIV-2), among others. The term HIV includes mutant strains of HIV including “drug resistant” or “multiple drug resistant” strains of the HIV virus which have mutated to be resistant to one or more clinically approved anti-HIV agents, including, in particular, HIV strains which are resistant to one or more NRTI compounds and/or NNRTI compounds. Exemplary HIV drug resistant strains which may be effectively treated using compounds according to the present invention include the following, among others: (defined by their reverse transcriptase or RT mutation)-XXBRU, K65R, Y115F, F116Y, Q151M, M184V, L74V, V75T, 4XZT, T215Y, K103N, T215Y/M184V, 5705-72, 488-101, C910-6, LA1M184V, G910-6 L100I, K101E, K103N, V106A, D110E, V179D, Y181C, D185E, D186E, Y188H, G190E, E138K, M41L, D67N, K70R, T215Y/F, K219Q/E, Y181C, K103N, L100I, Y188C/H, among others, including HIV-1 isolates JR-FL, ADA, HXBc2, SF162 and BaL, among others. The terms “ARC” and “AIDS” refer to syndromes of the immune system caused by the human immunodeficiency virus, which are characterized by susceptibility to certain diseases and T cell counts which are depressed compared to normal counts. HIV progresses from Category 1 (Asymptomatic HIV Disease) to Category 2 (ARC), to Category 3 (AIDS), with the severity of the disease. A Category 1 HIV infection is characterized by the patient or subject being HIV positive, asymptomatic (no symptoms) and having never had fewer than 500 CD4 cells. If the patient has had any of the AIDS-defining diseases listed for categories 2 (ARC) or 3 (AIDS), then the patient is not in this category. If the patient's t-cell count has ever dropped below 500, that patient is considered either Category 2 (ARC) or Category 3 (AIDS). A Category 2 (ARC) infection is characterized by the following criteria: The patient's T-cells have dropped below 500 but never below 200, and that patient has never had any Category 3 diseases (as set forth below) but have had at least one of the following defining illnesses—Bacillary angiomatosisCandidiasis, oropharyngeal (thrush)Candidiasis, vulvovaginal; persistent, frequent, or poorly responsive to therapyCervical dysplasia (moderate or severe)/cervical carcinoma in situConstitutional symptoms, such as fever (38.5 C) or diarrhea lasting longer than 1 monthHairy leukoplakia, oralHerpes zoster (shingles), involving at least two distinct episodes or more than one dermatomeIdiopathic thrombocytopenic purpuraListeriosisPelvic inflammatory disease, particularly if complicated by tubo-ovarian abscessPeripheral neuropathy According to the U.S. government, in Category 2 ARC, the immune system shows some signs of damage but it isn't life-threatening. A Category 3 (AIDS) infection is characterized by the following criteria: T-cells have dropped below 200 or the patient has had at least one of the following defining illnesses— Brain ToxoplasmosisCandidiasis of bronchi, trachea, or lungsCandidiasis, esophagealCervical cancer, invasive**Coccidioidomycosis, disseminated or extrapulmonaryCryptococcosis, extrapulmonaryCryptosporidiosis, chronic intestinal (greater than 1 month's duration)Cytomegalovirus disease (other than liver, spleen, or nodes)Cytomegalovirus retinitis (with loss of vision)Encephalopathy, HIV-relatedHerpes simplex: chronic ulcer(s) (greater than 1 month's duration); or bronchitis, pneumonitis, or esophagitisHistoplasmosis, disseminated or extrapulmonaryIsosporiasis, chronic intestinal (greater than 1 month's duration)Kaposi's sarcomaLymphoma, Burkitt's (or equivalent term)Lymphoma, immunoblastic (or equivalent term)Lymphoma, primary, of brainMycobacterium aviumcomplex orM. kansasii, disseminated or extrapulmonaryMycobacterium tuberculosis, any site (pulmonary** or extrapulmonary)Mycobacterium, other species or unidentified species, disseminated or extrapulmonaryPneumocystis cariniipneumoniaPneumonia, recurrentProgressive multifocal leukoencephalopathySalmonellasepticemia, recurrentWasting syndrome due to HIV The term “coadministration” or “combination therapy” shall mean that at least two compounds or compositions are administered to the patient at the same time, such that effective amounts or concentrations of each of the two or more compounds may be found in the patient at a given point in time. Although compounds according to the present invention may be co-administered to a patient at the same time, the term embraces both administration of two or more agents at the same time or at different times, provided that effective concentrations of all coadministered compounds or compositions are found in the subject at a given time. In certain preferred aspects of the present invention, one or more of the bifunction ARM-HI compounds described above, are coadministered in combination with at least one additional anti-HIV agent as otherwise described herein in a cocktail for the treatment of HIV infections. In particularly preferred aspects of the invention, the coadministration of compounds results in synergistic anti-HIV activity of the therapy. The term “additional anti-HIV agent” shall mean a traditional anti-HIV agent (ie., a non-bifunctional ARM-HI compound as otherwise described herein) which may be co-administered to a patient along with ARM-HI compounds according to the present invention in treating a patient for HIV. Such compounds include, for example, agents such as nucleoside reverse transcriptase inhibitors (NRTI), non-nucleoside reverse transcriptase inhibitors, protease inhibitors and fusion inhibitors. Exemplary compounds include, for example, Amprenivir, Abacavir, Acemannan, Acyclovir, AD-439, AD-519, Adefovir dipivoxil, Alpha Interferon, Ansamycin, 097, AR 177, Beta-fluoro-ddA, BMS-232623 (CGP-73547), BMS-234475 (CGP-61755), CI-1012, Cidofovir, Curdlan sulfate, Cytomegalovirus Immune globin, Ganciclovir, Dideoxyinosine, DMP-450, Efavirenz (DMP-266), EL10, Famciclovir, FTC, GS 840, HBY097, Hypericin, Recombinant Human Interferon Beta, Interferon alfa-n3, Indinavir, ISIS-2922, KNI-272, Lamivudine (3TC), Lobucavir, Nelfinavir, Nevirapine, Novapren, Peptide T Octapeptide Sequence, Trisodium Phosphonoformate, PNU-140690, Probucol, RBC-CD4, Ritonavir, Saquinavir, Valaciclovir, Virazole Ribavirin, VX-478, Zalcitabine, Zidovudine (AZT), Tenofovir diisoproxil fumarate salt, Combivir, Abacavir succinate, T-20), AS-101, Bropirimine, CL246, EL10, FP-21399, Gamma Interferon, Granulocyte Macrophage Colony Stimulating Factor (GM-CSF), HIV Core Particle Immunostimulant, Interleukin-2 (IL-2), Immune Globulin Intravenous, IMREG-1, IMREG-2, Imuthiol Diethyl Dithio Carbamate, Alpha-2 Interferon, Methionine-Enkephalin, MTP-PE (Muramyl-Tripeptide), Granulocyte Colony Stimulating Factor (GCSF), Remune, rCD4 (Recombinant Soluble Human CD4-IgG), rCD4-IgG Hybrids, Recombinant Soluble Human CD4, Interferon Alfa 2a, SK&F1-6528, Soluble T4, Thymopentin, Tumor Necrosis Factor (TNF), AK602, Alovudine, Amdoxovir, AMD070, Atazanavir (Reyataz), AVX754 (apricitabine), Bevirimat, BI-201, BMS-378806, BMS-488043, BMS-707035, C31G, Carbopol 974P, Calanolide A, Carrageenan, Cellulose sulfate, Cyanovirin-N, Darunavir, Delavirdine, Didanosine (Videx), Efavirenz, Elvucitabine, Emtricitabine, Fosamprenavir (Lexiva), Fozivudine tidoxil, GS 9137, GSK-873,140 (aplaviroc), GSK-364735, GW640385 (brecanavir), HG0004, HGTV43, INCB9471, KP-1461, Lopinavir, Mifepristone (VGX410), MK-0518, PPL-100, PRO 140, PRO 542, PRO 2000, Racivir, SCH-D (vicriviroc), SP01A, SPL7013, TAK-652, Tipranavir (Aptivus), TNX-355, TMC125 (etravirine), UC-781, UK-427,857 (Maraviroc), Valproic acid, VRX496, Zalcitabine, Valganciclovir, Clindamycin with Primaquine, Fluconazole Pastille, Nystatin Pastille, Eflornithine, Pentamidine, Isethionate, Trimethoprim, Trimethoprim/sulfa, Piritrexim, Pentamidine isethionate, Spiramycin, Intraconazole-R51211, Trimetrexate, Daunorubicin, Recombinant Human Erythropoietin, Recombinant Human Growth Hormone, Megestrol Acetate, Testosterone, Aldesleukin (Proleukin), Amphotericin B, Azithromycin (Zithromax), Calcium hydroxyapatite, Doxorubicin, Dronabinol, Entecavir, Epoetin alfa, Etoposide, Fluconazole, Isoniazid, Itraconazole (Sporanox), Megestrol, Paclitaxel (Taxol), Peginterferon alfa-2, Poly-L-lactic acid (Sculptra), Rifabutin (Mycobutin), Rifampin, Somatropin and Sulfamethoxazole/Trimethoprim. Preferred anti-HIV compounds for use in the present invention include, for example, 3TC (Lamivudine), AZT (Zidovudine), (−)-FTC, ddI (Didanosine), ddC (zalcitabine), abacavir (ABC), tenofovir (PMPA), D-D4FC (Reverset), D4T (Stavudine), Racivir, L-FddC, L-FD4C, NVP (Nevirapine), DLV (Delavirdine), EFV (Efavirenz), SQVM (Saquinavir mesylate), RTV (Ritonavir), IDV (Indinavir), SQV (Saquinavir), NFV (Nelfinavir), APV (Amprenavir), LPV (Lopinavir), fusion inhibitors such as T20, among others, fuseon and mixtures thereof The term “pharmaceutically acceptable salt” is used throughout the specification to describe a salt form of one or more of the compounds herein which are presented to increase the solubility of the compound in saline for parenteral delivery or in the gastric juices of the patient's gastrointestinal tract in order to promote dissolution and the bioavailability of the compounds. Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable inorganic or organic bases and acids. Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium, magnesium and ammonium salts, among numerous other acids well known in the pharmaceutical art. Sodium and potassium salts may be particularly preferred as neutralization salts of carboxylic acid containing compositions according to the present invention. The term “salt” shall mean any salt consistent with the use of the compounds according to the present invention. In the case where the compounds are used in pharmaceutical indications, including the treatment of HIV infections, the term “salt” shall mean a pharmaceutically acceptable salt, consistent with the use of the compounds as pharmaceutical agents. The term “antibody binding terminal moiety”, “antibody binding terminus” or “antibody binding moiety” (ABT within the general formula of compounds according to the present invention) is used to describe that portion of a bifunctional ARM-HI compound according to the present invention which comprises at least one small molecule or hapten which can bind to antibodies within the patient. The term “hapten” is used to describe a small-molecular-weight inorganic or organic molecule that alone is not antigenic but which when linked to another molecule, such as a carrier protein (albumin, etc.) or in the case of the present invention, as an antibody terminus in the present compounds, is antigenic; and an antibody raised against the hapten (generally, the hapten bonded or complexed to the carrier) will react with the hapten alone. Because, in many instances, anti-hapten (anti-DNP) antibodies are already present in the human blood stream as endogenous antibodies because they naturally become raised to endogenous haptens (already present in patients), no pre-vaccination is necessary for ARM-HI activity. It is preferred that the antibody binding terminal comprise a hapten which is reactive with (binds to) an endogenous antibody that pre-exists in the patient prior to initiate therapy with the compounds of the present invention and does not have to be separately raised as part of a treatment regimen (for example, by vaccination or other approach for enhancing immunogenicity). Thus, haptens which comprise a di- or trinitro phenyl group as depicted below, or a digalactose hapten (Gal-Gal-Z, preferably Gal-Gal-sugar, preferably Gal-Gal-Glu), are preferred. Additionally, a compound according to the general structure: Where X″ is O, CH2, NR1, S; and R1is H, a C1-C3alkyl group or a —C(O)(C1-C3) group; May be used as haptens in the present invention. Further, a moiety according to the chemical structure: Where Xbis a bond, O, CH2, NR1or S may also be used as a hapten (ABT) in the present invention. Other ABT moieties include the following structures: Each of the above amino acid ABT moieties may be further substituted with a dinitrophenyl group through an X group, e.g., CH2—, sulfoxide, sulfone, etc. group as otherwise described herein to provide the following ABT moieties: In the above structures in each of the molecules (with the exception of the first, which is DNP amine), DNP may be linked to the structure where the NO2is linked. The di- or trinitro phenyl hapten (ABT) moiety for use in the present invention (Dinitropheny or DNP hapten is preferred) may be represented by the following formula: Where Y′ is H or NO2(preferably H); X is O, CH2, NR′, S(O), S(O)2, —S(O)2O, —OS(O)2, or OS(O)2O; and R1is H, a C1-C3alkyl group, or a —C(O)(C1-C3) group. The (Gal-Gal-Z) hapten is represented by the chemical formula: Where X′ is CH2, O, N—R1′, or S, preferably O; R1′is H or C1-C3alkyl; and Z is a bond, a monosaccharide, disaccharide, oligosaccharide, glycoprotein or glycolipid, preferably a sugar group, more preferably a sugar group selected from the monosaccharides, including aldoses and ketoses, and disaccharides, including those disaccharides described herein. Monosaccharide aldoses include monosaccharides such as aldotriose (D-glyceraldehdye, among others), aldotetroses (D-erythrose and D-Threose, among others), aldopentoses, (D-ribose, D-arabinose, D-xylose, D-lyxose, among others), aldohexoses (D-allose, D-altrose, D-Glucose, D-Mannose, D-gulose, D-idose, D-galactose and D-Talose, among others), and the monosaccharide ketoses include monosaccharides such as ketotriose (dihydroxyacetone, among others), ketotetrose (D-erythrulose, among others), ketopentose (D-ribulose and D-xylulose, among others), ketohexoses (D-Psicone, D-Fructose, D-Sorbose, D-Tagatose, among others), aminosugars, including galactoseamine, sialic acid, N-acetylglucosamine, among others and sulfosugars, including sulfoquinovose, among others. Exemplary disaccharides which find use in the present invention include sucrose (which may have the glucose optionally N-acetylated), lactose (which may have the galactose and/or the glucose optionally N-acetylated), maltose (which may have one or both of the glucose residues optionally N-acetylated), trehalose (which may have one or both of the glucose residues optionally N-acetylated), cellobiose (which may have one or both of the glucose residues optionally N-acetylated), kojibiose (which may have one or both of the glucose residues optionally N-acetylated), nigerose (which may have one or both of the glucose residues optionally N-acetylated), isomaltose (which may have one or both of the glucose residues optionally N-acetylated), β,β-trehalose (which may have one or both of the glucose residues optionally N-acetylated), sophorose (which may have one or both of the glucose residues optionally N-acetylated), laminaribiose (which may have one or both of the glucose residues optionally N-acetylated), gentiobiose (which may have one or both of the glucose residues optionally N-acetylated), turanose (which may have the glucose residue optionally N-acetylated), maltulose (which may have the glucose residue optionally N-acetylated), palatinose (which may have the glucose residue optionally N-acetylated), gentiobiluose (which may have the glucose residue optionally N-acetylated), mannobiose, melibiose (which may have the glucose residue and/or the galactose residue optionally N-acetylated), melibiulose (which may have the galactose residue optionally N-acetylated), rutinose, (which may have the glucose residue optionally N-acetylated), rutinulose and xylobiose, among others. Oligosaccharides for use in the present invention as Z can include any sugar of three or more (up to about 100) individual sugar (saccharide) units as described above (i.e., any one or more saccharide units described above, in any order, especially including glucose and/or galactose units as set forth above), or for example, fructo-oligosaccharides, galactooligosaccharides and mannan-oligosaccharides ranging from three to about ten-fifteen sugar units in size. Glycoproteins for use in the present invention include, for example, N-glycosylated and O-glycosylated glycoproteins, including the mucins, collagens, transferring, ceruloplasmin, major histocompatability complex proteins (MHC), enzymes, lectins and selectins, calnexin, calreticulin, and integrin glycoprotein IIb/IIa, among others. Glycolipids for use in the present invention include, for example, glyceroglycolipids (galactolipids, sulfolipids), glycosphingolipids, such as cerebrosides, galactocerebrosides, glucocerebrosides (including glucobicaranateoets), gangliosides, globosides, sulfatides, glycophosphphingolipids and glycocalyx, among others. Preferably, Z is a bond (linking a Gal-Gal disaccharide to a linker or connector molecule) or a glucose or glucosamine (especially N-acetylglucosamine). It is noted that Z is linked to a galactose residue through a hydroxyl group or an amine group on the galactose of Gal-Gal, preferably a hydroxyl group. A preferred hapten is Gal-Gal-Glu which is represented by the structure: Where Xs is OH or NHAc. Other ABT groups include, for example, the following groups: Where XRis O or S; and XMis O or S. It is noted in the carboxyethyl lysine ABT moiety either one, two or three of the nitrogen groups may be linked to the remaining portion of the molecule through the linker or one or both of the remaining nitrogen groups may be substituted with a dinitrophenyl through an X group as otherwise described herein. The term “pathogen binding terminus” or “pathogen binding terminal moiety” (“PBT”) is use to described that portion of a difunctional ARM-HI compound according to the present invention which comprises at least one small molecule or moiety which can bind specifically to is capable of binding to gp120 envelope protein on HIV virus or a cell surface of CD4 cells which are infected with HIV (HIV+) in said patient. PBT groups (i.e., the chemical moiety connected to linkers and ABT in the bifunctional chemical compound below) for use in the present invention include those which are found in the following bifunctional compounds having the following chemical structure: Where is an antibody binding terminus (moiety) comprising a hapten which is capable of binding to an antibody present in a patient (preferably a DNP group); is a linker molecule which chemical links ABT to RYor directly to the indole moiety at the carbon atom to which RYis attached and which optionally includes a connector CT which may be a bond or a connector molecule; is an aromatic or heteroaromatic group, preferably a monocyclic or bicyclic aromatic or heteroaromatic group; RYis absent or is an optionally substituted aryl or heteroaryl group or O, (CH2)j, —S—, —NHC(O)—, —NHC(O)NH—, S(O), S(O)2, —S(O)2O, —OS(O)2, or OS(O)2O; X2is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(CH2O)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(CH2O)nO—(C1-C6alkyl), —(CH2O)nC(O)—(C1-C6alkyl), —(CH2O)nNHC(O)—R1, —(CH2O)nC(O)—NR1R2, NO2, CN or halogen (F, Cl, Br, I, preferably F or Cl); X3is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(CH2O)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(CH2O)nO—(C1-C6alkyl), —(CH2O)nC(O)—(C1-C6alkyl), —(CH2O)nNHC(O)—R1, —(CH2O)nC(O)—NR1R2, NO2, CN, halogen (F, Cl, Br, I, preferably F or Cl) or a monocyclic aryl or heteroaryl group which itself is optionally substituted; R1is H or a C1-C3alkyl group; R1and R2are each independently H or a C1-C6alkyl group; i is 0 or 1, preferably 1; j is 1, 2 or 3; k is 0, 1, 2 or 3, preferably 0, 1 or 2; n is 0, 1, 2, 3, 4, 5, 6, preferably 0-3; Y3is H or a C1-C3alkyl group (preferably, disposed out of or into the plane, preferably out of the plane on the chiral carbon; and RNis H or a C1-C3alkyl group which is optionally substituted with one or two hydroxyl groups or up to three halogen groups (preferably F) or a pharmaceutically acceptable salt, enantiomer, solvate or polymorph thereof. Preferred PBT groups for use in the present invention include those (i.e., the chemical moiety connected to the linker and ABT below—connected to X) according to the chemical formula: Where is a monocyclic or bicyclic aryl or heteroaryl group according to the chemical structure: Where W is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(CH2O)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(CH2O)nO—(C1-C6alkyl), —(CH2O)nC(O)—(C1-C6alkyl), —(CH2O)nNHC(O)—R1, —(CH2O)nC(O)—NR1R2, NO2, CN, halogen (F, Cl, Br, I, preferably F or Cl) or a monocyclic aryl or heteroaryl group which itself is optionally substituted (especially an optionally substituted benzoyl or benzyl group); W′ is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl) or halogen (preferably F or Cl); is a group according to chemical structure: Where W2is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(CH2O)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(CH2O)nO—(C1-C6alkyl), —(CH2O)nC(O)—(C1-C6alkyl), —(CH2O)nNHC(O)—R1, —(CH2O)nC(O)—NR1R2, NO2, CN or halogen (preferably F or CO; X is a group —(CH2)nNH—, —(CH2)nNHC(O)—, —(CH2)nO—, —(CH2)m—, —(CH2)nS—, —(CH2)nS(O)—, —(CH2)nSO2— or —(CH2)nNH—C(O)—NH— which links to the linker; Y is O, S or N—R where R is H or a C1-C3alkyl group; X2is H, —(CH2)nOH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(CH2O)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(CH2O)nO—(C1-C6alkyl), —(CH2O)nC(O)—(C1-C6alkyl), —(CH2O)nNHC(O)—R1or —(CH2O)nC(O)—NR1R2, NO2; R1and R2are each independently H or a C1-C6alkyl group; Y3is H or a C1-C3alkyl group (preferably, disposed out of or into the plane, preferably out of the plane on the chiral carbon; and RNis H or a C1-C3alkyl group which is optionally substituted with one or two hydroxyl groups or up to three halogen groups (preferably F); i is 0 or 1, preferably 1; and m is 1, 2, 3, 4 or 5, preferably 1, 2 or 3, more preferably 1; and Each n is independently 0, 1, 2 or 3, or a pharmaceutically acceptable salt, enantiomer, solvate or polymorph thereof. The term “linker” refers to a chemical entity connecting an antibody binding terminus (ABT) moiety to a pathogen binding terminus (CBT) moiety, optionally through a connector moiety (CT) through covalent bonds. The linker between the two active portions of the molecule, that is the antibody binding terminus (ABT) and the pathogen binding terminus (PBT) ranges from about 5 Å to about 50 Å or more in length, about 6 Å to about 45 Å in length, about 7 Å to about 40 Å in length, about 8 Å to about 35 Å in length, about 9 Å to about 30 Å in length, about 10 Å to about 25 Å in length, about 7 Å to about 20 Å in length, about 5 Å to about 16 Å in length, about 5 Å to about 15 Å in length, about 6 Å to about 14 Å in length, about 10 Å to about 20 Å in length, about 11 Å to about 25 Å in length, etc. Linkers which are based upon ethylene glycol units and are between 4 and 14 glycol units in length may be preferred. By having a linker with a length as otherwise disclosed herein, the ABT moiety and the PBT moiety may be situated to advantageously take advantage of the biological activity of compounds according to the present invention which bind to HIV envelope protein gp120 (gp120) and attract endogenous antibodies to the virus and/or infected cells (e.g. HIV infected CD4 cells) to which the compounds are bound, resulting in the selective and targeted death of those viruses and/or cells. The selection of a linker component is based on its documented properties of biocompatibility, solubility in aqueous and organic media, and low immunogenicity/antigenicity. Although numerous linkers may be used as otherwise described herein, a linker based upon polyethyleneglycol (PEG) linkages, polypropylene glycol linkages, or polyethyleneglycol-co-polypropylene oligomers (up to about 100 units, about 1 to 100, about 1 to 75, about 1 to 60, about 1 to 50, about 1 to 35, about 1 to 25, about 1 to 20, about 1 to 15, 2 to 10, about 4 to 12, about 1 to 8, 1 to 3, 1 to 4, 2 to 6, 1 to 5, etc.) may be favored as a linker because of the chemical and biological characteristics of these molecules. The use of polyethylene (PEG) linkages is preferred. Alternative preferred linkers may include, for example, polyproline linkers and/or collagen linkers as depicted below (n is about 1 to 100, about 1 to 75, about 1 to 60, about 1 to 50, about 1 to 45, about 1 to 35, about 1 to 25, about 1 to 20, about 1 to 15, 2 to 10, about 4 to 12, about 5 to 10, about 4 to 6, about 1 to 8, about 1 to 6, about 1 to 5, about 1 to 4, about 1 to 3, etc.). Preferred linkers include those according to the chemical structures: Or a polypropylene glycol or polypropylene-co-polyethylene glycol linker having between 1 and 100 glycol units; Where Rais H, C1-C3alkyl or alkanol or forms a cyclic ring with R3(proline) and R3is a side chain derived from an amino acid preferably selected from the group consisting of alanine (methyl), arginine (propyleneguanidine), asparagine (methylenecarboxyamide), aspartic acid (ethanoic acid), cysteine (thiol, reduced or oxidized di-thiol), glutamine (ethylcarboxyamide), glutamic acid (propanoic acid), glycine (H), histidine (methyleneimidazole), isoleucine (1-methylpropane), leucine (2-methylpropane), lysine (butyleneamine), methionine (ethylmethylthioether), phenylalanine (benzyl), proline (R3forms a cyclic ring with Raand the adjacent nitrogen group to form a pyrrolidine group), hydroxyproline, serine (methanol), threonine (ethanol, 1-hydroxyethane), tryptophan (methyleneindole), tyrosine (methylene phenol) or valine (isopropyl); m (within this context) is an integer from 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5; n (within this context) is an integer from about 1 to 100, about 1 to 75, about 1 to 60, about 1 to 50, about 1 to 45, about 1 to 35, about 1 to 25, about 1 to 20, about 1 to 15, 2 to 10, about 4 to 12, about 5 to 10, about 4 to 6, about 1 to 8, about 1 to 6, about 1 to 5, about 1 to 4, about 1 to 3, etc.) or Another linker according to the present invention comprises a polyethylene glycol linker containing from 1 to 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5 ethylene glycol units, to which is bonded a lysine group (preferably at its carboxylic acid moiety) which binds one or two DNP groups to the lysine at the amino group(s) of lysine. Still other linkers comprise amino acid residues (D or L) to which are bonded to ABT moieties, in particular, DNP, among others at various places on amino acid residue as otherwise described herein. In another embodiment, as otherwise described herein, the amino acid has anywhere from 1-15 methylene groups separating the amino group from the acid group in providing a linker to the ABT moiety. Or another linker is according to the chemical formula: Where Z and Z′ are each independently a bond, —(CH2)i—O, —(CH2)i—S, —(CH2)i—N—R, wherein said —(CH2), group, if present in Z or Z′, is bonded to a connector, ABT or CBT; Each R is H, or a C1-C3alkyl or alkanol group; Each R2is independently H or a C1-C3alkyl group; Each Y is independently a bond, O, S or N—R; Each i is independently 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5; D is or a bond, with the proviso that Z, Z′ and D are not each simultaneously bonds; j is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5; m′ is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5; n is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5; X1is O, S or N—R; and R is as described above, or a pharmaceutical salt thereof. The term “connector”, symbolized in the generic formulas by [CT], is used to describe a chemical moiety which is optionally included in bifunctional compounds according to the present invention which forms from the reaction product of an activated ABT-linker with a PTB moiety (which also is preferably activated) or an ABT moiety with an activated linker-PTB as otherwise described herein. The connector group is often the resulting moiety which forms from the facile condensation of two or more separate chemical fragments which contain reactive groups which can provide connector groups as otherwise described to produce bifunctional or multifunctional compounds according to the present invention. It is noted that a connector may be distinguishable from a linker in that the connector is the result of a specific chemistry which is used to provide bifunctional compounds according to the present invention wherein the reaction product of these groups results in an identifiable connector group or part of a connector group which is distinguishable from the linker group, although in certain instances, incorporated into the linker group, as otherwise described herein. It is noted also that a connector group may be linked to a number of linkers to provide multifunctionality (i.e., more than one PBT moiety and/or more than one ABT moiety within the same molecule. It is noted that there may be some overlap between the description of the connector group and the linker group such that the connector group is actually incorporated or forms part of the linker, especially with respect to more common connector groups such as amide groups, oxygen (ether), sulfur (thioether) or amine linkages, urea or carbonate —OC(O)O— groups as otherwise described herein. It is further noted that a connector (or linker) may be connected to ABT, a linker or PBT at positions which are represented as being linked to another group using the using the symbol Where two or more such groups are present in a linker or connector, any of an ABT, a linker or a PBT may be bonded to such a group. Where that symbol is not used, the link may be at one or more positions of a moiety. Common connector groups which are used in the present invention include the following chemical groups: Where X2is O, S, NR4, S(O), S(O)2, —S(O)2O, —OS(O)2, or OS(O)2O; X3is O, S, NR4; and R4is H, a C1-C3alkyl or alkanol group, or a —C(O)(C1-C3) group. The triazole group, indicated above, is a preferred connector group. As discussed hereinabove, it is noted that each of the above groups may be further linked to a chemical moiety which bonds two or more of the above connector groups into a multifunctional connector, thus providing complex multifunctional compounds comprising more than one ABT and/or PBT group within the multifunctional compound. Initial work by the inventors involved in identifying bifunctional compounds ARM-HI according to the present invention, began with the small molecule BMS-378806 (Formula 1, below), (4-benzoyl-1-(2-(4-methoxy-1H-pyrrolo(2,3-b) pyridin-3-yl)-1,2-dioxoethyl)-2-methyl-, (2R)-Piperazine, CAS Number 357263-13-9, MW 406), a known inhibitor of the CD4-gp120 interaction. (Wang, et al. J. Med. Chem. 2003, 46, 4236-42396). It was shown in international application PCT/US2010/52344 (published as WO/2011/046946), which is incorporated by reference herein, that it was possible to derivatize Formula 1, at the carbon atom of the C4 methoxy group, in which the carbon atom of the C4 methoxy group could be replaced with various bulky substituents, (Wang, J, S.; Le, N.; Heredia, A.: Song, H. J.: Redfield, R.: Wang, L. X. Org. Biomol. Chem. 2005, 3, 1781-1786) so as to provide a linker which would attract DNP without sacrificing the compound's ability to inhibit viral entry. This hypothesis was supported by an analysis of a published computational docking model suggesting that the C4methoxy group in Formula 1 points toward the solvent in the complex. See Kong. R.; Tan, J.; Ma, X.; Chen, W.; Wang. C:. Biochim. Biophys. Acta 2006, 1764. 766-7728. Thus, in accordance with the invention of PCT/US2010/52344 (published as WO/2011/046946), Formula 1 was re-engineered to include the capability to recruit anti-DNP antibodies to gp120-expressing particles (infected cells or viruses), increasing the “visibility” of the combination to the human immune system. Consequently, an ARM-HI of Formula 4 was prepared in high yield (38% overall) via azide-alkyne cycloaddition (Rostovtsev, V. V.; Green, L. G.; Fokin, V. V.; Sharpless, K. B. Angew. Chem., Int, Ed. 2002, 41, 2596-2599. Tornoe, C.; Christensen, C.; Meldal, M. J. Org. Chem. 2002, 67, 3057-3064) of the compounds of Formula 2 and Formula 3, which were derived in turn from known intermediates. See, Wang, T.; et al. J. Med. Chem. 2003, 46, 4236-4239. As discussed above, ARM-HI compounds, including compounds according to the present invention, target HIV by inhibiting virus entry while targeting Env-expressing cells for immune recognition and clearance. (SeeFIG.1) Compounds set forth in the prior PCT application were shown to inhibit CD4 binding to HIV-1 gp120 and to out-compete the CD4-gp120 interaction. It was confirmed that ARM-HI has the ability to recruit antibodies to gp120 both in vitro and in tissue culture. Initial ELISA experiments demonstrated a concentration-dependent increase in anti-DNP antibody binding to the ARM-HI-gp120 complex but not to gp120 alone. Thus, ARM-HI is capable of templating a ternary complex that also includes gp120 and anti-DNP antibody. It was also confirmed that the ternary association could form in a complex cellular milieu, and that ARM-H bifunctional compounds have the ability to recruit anti-DNP antibodies to HIV-Env-expressing Chinese hamster ovary cells (CHO-gp120cells). Thus, the previous results presented in PCT/US2010/52344 (published as WO/2011/046946) provide strong evidence that ARM-HI bifunctional agents of the present invention are capable of recruiting anti-hapten (e.g. anti-DNP) antibodies to cells expressing the Env glycoprotein in a fashion that depends upon its simultaneous binding to both gp120 and anti-DNP antibodies and that the ternary complex formed from anti-DNP antibody, ARM-HI, and alive Env-expressing cell activates complement proteins and mediates cellular death. Notably, in the absence of anti-DNP antibody and complement-preserved serum (data in green), in cells lacking the Env glycoprotein (CHO-WT, data in black), or in the presence of compound which lacks the DNP group, no cell death is observed, suggesting that termolecular complex formation is necessary for complement-dependent cytotoxicity (CDC) and that ARM-HI itself is not toxic to cells. The present invention takes a novel approach and is directed to the development of further novel compositions which recruit anti-DNP antibodies and other anti-hapten antibodies, endogenous in most patients, to HIV via binding to the gp120 envelope protein, which additionally prevents HIV from binding to CD4 and T4 cells, providing novel compositions and therapy for treating HIV infection and the symptoms associated therewith. The present compounds exhibit substantially greater activity than the compounds which are disclosed in PCT/US2010/52344 (published as WO/2011/046946). The following detailed description outlines the design and synthesis of a number of bifunctional small-molecules capable of redirecting endogenous anti-hapten antibodies, especially including anti-dinitrophenyl (DNP) antibodies selectively to HIV, and inducing antibody-directed, cell-mediated cytotoxicity, which are based upon the results obtained for the compounds originally presented in PCT/US2010/52344 (WO/2011/046946). The following chemical synthesis which is presented in Scheme 1 (FIG.3) and Scheme 2 (FIG.4) may be used to synthesize the compound labeled as C-5 Furan inFIG.6which shows exceptional activity as an anti-HIV agent. The Scheme 1 and Scheme 2 chemical syntheses are genericized in Scheme 3,FIG.5, to provide generic methods (either directly or by analogy) for producing virtually all of the compounds which are described herein. By way of synthesis, the carboxylic acid azide compound 2 (the azide readily forming a triazole connector molecule with an acetylenic group which links the ABT group with the PBT group) is prepared as otherwise described herein. Pursuant to scheme 1,FIG.3, the oligo(ethylene oxide) azide compound 1 is modified in sodium hydride and solvent (THF) with bromoacetic acid to provide compound 2, which contains both an electrophilic moiety (the carboxy group can condense to form an amide with an amine group) and an azide which can react with an acetylenic group to form a triazole (connector group). The pathogen binding terminus (PBT) group in the present application is modified to contain an aryl group (ARYL2) on carbocyclic group of the indole bicyclic ring. Scheme 2 (FIG.4) provides a rather facile synthesis of C-5 furan from the bromo-substituted indole compound 4, which condenses a substituted furan compound (4) onto the indole ring as indicated in Scheme 2 to produce the furan-substituted PTB compound 5. Compound 5 is treated with trifluoroacetic acid in dichloromethane to produce intermediate 6 which is reacted with carboxyl azide compound 2 to produce compound 7. Compound 7 is then reacted with a compound containing an acetylenic moiety and an ABT group (in Scheme 2, a DNP group) under favorable conditions to produce the active bifunctional compound C5-furan (containing a PBT group containing a furanyl group and an ABT group linked together through a linking group, seeFIG.6). The chemical synthesis provided above may be presented in a more generalized fashion as set forth in Scheme 3,FIG.5. In the generic synthesis a bromo-substituted indole compound is first reacted with oxalyl chloride followed by protected piperazine in trifluoroacetic acid to produce compound 10. An aryl group substituting for the bromo group in the indole moiety may be introduced as Arene 1 (Scheme 3,FIG.5) by reacting an aryl hydroxy boron substituted compound containing an alkylene group substituted with a hydroxyl, an amine or a sulfhydryl group which provides compound 11, which may be further reacted with carboxylic acid azide compound 2 to form an azide containing compound which is then further reacted with an acetylene containing group (containing a linker group and a ABT group (DNP) to condense the acetylenic group onto the azide to form a triazole containing compound 13. Compound 13 may be further reacted with an appropriately substituted (carboxylic acid group which can be condensed onto free amine group of the piperazine moiety) aryl group (arene 2,FIG.5) to form the final bioactive biofunctional compounds according to the present invention. As noted, this generic synthesis may be used to provide a larger number of compounds which can accommodate numerous aryl groups and numerous ABT groups as indicated. Various analogs are also synthesized, specifically exemplary compounds of the invention which include alternative ABT substituents. Combing an ABT group (with or without a further linking group) containing an acetylenic group with an azide is rather facile and the formation of an azide group and/or an acetylenic group may be used to generally link the ABT group to the PBT group through a connector/linker as otherwise described herein. Thusly, in the present invention a PBT portion of a molecule is derivatized with a linker containing an azide group which can form a connector molecule in subsequent reactions. Once the derivative PBT molecule is formed, bifunctional compounds according to the present invention may be formed by condensation with appropriate ABT-containing molecules to produce the final bifunctional compounds according to the present invention. Using the above synthesis with appropriate modification, bifunctional compounds according to the present invention may be readily synthesized. These compounds contain a single PBT moiety to which is linked a compound comprising an ABT moiety. The above schemes provide exemplary synthesis of compounds according to the present invention with various iterations of same provided by analogy using well known methods as described herein and as understood by those of ordinary skill in the art. It is noted that the experimental section provides significant detail to allow the facile synthesis of a variety of bifunctional compounds as otherwise described herein. The schemes are not to be considered limiting in setting forth teachings which provide compounds according to the present invention. Turning to the biological data of bifunctional compounds according to the present invention, with reference toFIG.6, this figure shows a number of compounds which were tested in a viral inhibition assay. In this assay, IC50's of a number of prior art compounds were determined against different HIV-1 isolates as set forth in Table 1 below. In this assay, viral inhibition was determined by HIV Tat-induced luciferase (Luc) reporter gene expression after a single round of virus infection in TZM-bl cells according to the method of Platt, et al.,J. Virol.1998, 72, 2855-64. This biological data evidences that the present compounds are unexpectedly more active than are the compounds having a linker and ABT moiety at different positions of the indole ring, an unexpected result. Compound FIG. 6JR-FLADAHXBc2SF162BaLBMS-378806259534431.9359dncARM-HI12901003dncDncdncBMS-furan0.1170.1520.0970.0050.01m-phenyl52.837520820800150257C5-furan0.7206.3510.330934.8(PresentInvention)* Note-All values in nM;dnc = does not converge While specific analogs have been shown and described, the present invention is not limited to these specific analogs and other antibody recruiting compounds that can function as the antibody recruiting terminus connected by a linker to a binding terminus that will bind to the HIV glycoprotein gp120 (gp120 on the viral membrane as well as gp120 displayed on infected cells), would fall within the scope of the present invention. All of these compounds can be formulated into pharmaceutical compositions as otherwise described herein and used in the methods which are presented. Pharmaceutical compositions comprising combinations of an effective amount of at least one bifunctional compound according to the present invention, and one or more of the compounds otherwise described herein, all in effective amounts, in combination with a pharmaceutically effective amount of a carrier, additive or excipient, represents a further aspect of the present invention. The compositions of the present invention may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers and may also be administered in controlled-release formulations. Pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as prolamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. The compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term “parenteral” as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as Ph. Helv or similar alcohol. The pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. Alternatively, the pharmaceutical compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient which is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols. The pharmaceutical compositions of this invention may also be administered topically. Suitable topical formulations are readily prepared for each of these areas or organs. Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-acceptable transdermal patches may also be used. For topical applications, the pharmaceutical compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. In certain preferred aspects of the invention, the topical cream or lotion may be used prophylatically to prevent infection when applied topically in areas prone toward virus infection. In additional aspects, the compounds according to the present invention may be coated onto the inner surface of a condom and utilized to reduce the likelihood of infection during sexual activity. Alternatively, the pharmaceutical compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. For ophthalmic use, the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with our without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutical compositions may be formulated in an ointment such as petrolatum. The pharmaceutical compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. The amount of compound in a pharmaceutical composition of the instant invention that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host and disease treated, the particular mode of administration. Preferably, the compositions should be formulated to contain between about 0.05 milligram to about 750 milligrams or more, more preferably about 1 milligram to about 600 milligrams, and even more preferably about 10 milligrams to about 500 milligrams of active ingredient, alone or in combination with at least one other bifunctional compound according to the present invention or other anti-HIV agent which may be used to treat HIV infection or a secondary effect or condition thereof. It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease or condition being treated. A patient or subject (e.g. a male human) suffering from HIV infection can be treated by administering to the patient (subject) an effective amount of the ARM-HI compound according to the present invention including pharmaceutically acceptable salts, solvates or polymorphs, thereof optionally in a pharmaceutically acceptable carrier or diluent, either alone, or in combination with other known antiviral or pharmaceutical agents, preferably agents which can assist in treating HIV infection, including AIDS or ameliorate the secondary effects and conditions associated with HIV infection. This treatment can also be administered in conjunction with other conventional HIV therapies. These compounds can be administered by any appropriate route, for example, orally, parenterally, intravenously, intradermally, subcutaneously, or topically, in liquid, cream, gel, or solid form, or by aerosol form. The active compound is included in the pharmaceutically acceptable carrier or diluent in an amount sufficient to deliver to a patient a therapeutically effective amount for the desired indication, without causing serious toxic effects in the patient treated. A preferred dose of the active compound for all of the herein-mentioned conditions is in the range from about 10 ng/kg to 300 mg/kg, preferably 0.1 to 100 mg/kg per day, more generally 0.5 to about 25 mg per kilogram body weight of the recipient/patient per day. A typical topical dosage will range from 0.01-5% wt/wt in a suitable carrier. The compound is conveniently administered in any suitable unit dosage form, including but not limited to one containing less than 1 mg, 1 mg to 3000 mg, preferably 5 to 500 mg of active ingredient per unit dosage form. An oral dosage of about 25-250 mg is often convenient. The active ingredient is preferably administered to achieve peak plasma concentrations of the active compound of about 0.00001-30 mM, preferably about 0.1-30 μM. This may be achieved, for example, by the intravenous injection of a solution or formulation of the active ingredient, optionally in saline, or an aqueous medium or administered as a bolus of the active ingredient. Oral administration is also appropriate to generate effective plasma concentrations of active agent. The concentration of active compound in the drug composition will depend on absorption, distribution, inactivation, and excretion rates of the drug as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time. Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound or its prodrug derivative can be incorporated with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a dispersing agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or enteric agents. The active compound or pharmaceutically acceptable salt thereof can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors. The active compound or pharmaceutically acceptable salts thereof can also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as other anti-HIV agents, antibiotics, antifungals, anti-inflammatories, or antiviral compounds. In certain preferred aspects of the invention, one or more ARM-HI compounds according to the present invention are coadministered with another anti-HIV agent and/or another bioactive agent, as otherwise described herein. Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. If administered intravenously, preferred carriers are physiological saline or phosphate buffered saline (PBS). In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. Liposomal suspensions may also be pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811 (which is incorporated herein by reference in its entirety). For example, liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the active compound are then introduced into the container. The container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension. Detailed Synthetic Information Materials and General Information: Purchased starting materials were used as received unless otherwise noted. All moisture sensitive reactions were performed in an inert, dry atmosphere of nitrogen in flame dried glassware. Reagent grade solvents were used for extractions and flash chromatography. Reaction progress was checked by analytical thin-layer chromatography (TLC, Merck silica gel 60 F-254 plates). The plates were monitored either with UV illumination, or by charring with anisaldehyde (2.5% p-anisaldehyde, 1% AcOH, 3.5% H2SO4(conc.) in 95% EtOH) or ninhydrin (0.3% ninhydrin (w/v), 97:3 EtOH—AcOH) stains. Flash column chromatography was performed using silica gel (230-400 mesh). The solvent compositions reported for all chromatographic separations are on a volume/volume (v/v) basis. ELISA and CDC experiments were performed in triplicate and repeated at least three times unless otherwise noted. I Instrumentation:1H-NMR spectra were recorded at either 400 or 500 MHz and are reported in parts per million (ppm) on the δ scale relative to CDCl3(δ 7.26) as an internal standard unless otherwise noted. Data are reported as follows: chemical shift, multiplicity (s=singlet, d=doublet, t=triplet, q=quartet, br=broad, m=multiplet), coupling constants (Hz), and integration.13C-NMR spectra were recorded at either 100 or 125 MHz and are reported in parts per million (ppm) on the δ scale relative to CDCl3(δ 77.00). High resolution mass spectra (HRMS) were recorded on a 9.4T Bruker Qe FT-ICR MS (W. M. Keck Facility, Yale University). Analytical ultra high-performance liquid chromatography-mass spectrometry (UPLC/MS) was performed on a Waters UPLC/MS instrument equipped with a reverse-phase C18 column (1.7 μm particle size, 2.1×50 mm), dual atmospheric pressure chemical ionization (API)/electrospray (ESI) mass spectrometry detector, and photodiode array detector. Samples were eluted with a linear gradient of 20% acetonitrile-water→100% acetonitrile containing 0.1% formic acid over 3 min at a flow rate of 0.8 mL/min. Analytical UPLC/MS data are represented as follows: m/z; retention time (Rt) in minutes. High Pressure Liquid Chromatography (HPLC) using a Dynamax Rainin Solvent Delivery System equipped with a Varian Prostar Detector (Galaxie Chromatography Data System version 1.8.505.5), and absorbance measurements were made at 214 and 254 nm simultaneously. A Waters Xterra Prep MS C18 7.8×150 mm column was used for semi-preparative purifications using a water: acetonitrile (A:B) gradient containing 0.1% TFA at 5.0 mL/min, as specified below for individual compounds. Analytical HPLC analysis was performed using a Varian C8 4.6×250 mm Microsorb C8 column run at a flow rate of 1.0 mL/min water: acetonitrile (A: B) gradient containing 0.1% TFA. Unless otherwise noted, all micro-plate based assays were quantitated using a BioTek Synergy 3 Microplate reader and data was fitted and graphed using GraphPad Prism version 5.00 for Windows (GraphPad Software, San Diego Calif. USA, www.graphpad.com) or KaleidaGraph (Synergy Software). Azido polyethylene glycol 6 (0.60 g, 2 mmol, 1 equiv.) was dissolved in dry THF (10 mL) and cooled at 0° C., then sodium hydride (0.15 g, 6.3 mmol, 3.1 equiv.) was added in portions followed by bromoacetic acid (0.35 g, 2.5 mmol, 1.25 equiv.). The suspension was stirred at room temperature under nitrogen overnight. Water (1 mL) was added carefully and then stirred for 5 min. The reaction mixture was concentrated in vacuo. Dichloromethane was added and organic layer was washed with 2N HCl and brine. The organic layer was dried over Na2SO4and all solvents were evaporated. Pure 6 (0.72 g, 98%) was obtained as an oil.1H NMR (400 MHz, CDCl3) δ 4.16 (s, 2H), 3.76 (s, 2H), 3.72-3.58 (m, 20H), 3.39 (s, 2H). MS (ES+) 366 [M+H]+, 338 [M+H−N2] To a microwave vial (2.0-5.0 mL) containing 3 (52 mg, 0.11 mmol) in DMF/water (3.0/1.8 mL), added 5-((BOC-Amino)methyl)furan-2-boronic acid (Combi-Blocks LLC, San Diego Calif.; 37 mg, 0.154 mmol, 1.4 equiv) and NaHCO3(12.8 mg, 0.154 mmol, 1.4 equiv). Oxygen was removed from the solvent by bubbling nitrogen gas in solution for 10 min, to which Pd(PPh3)4(6.3 mg, 0.0055 mmol, 5 mol %) was added. The subsequent heterogenous solution was capped and heated in microwave reactor for 12 min at 150° C. when LC/MS analysis showed reaction completion. The volatile solvents were removed by rotary evaporation and crude material was purified by flash chromatography (flash chromatography (CombiFlash Automated Chromatographer, 12 g column; gradient elution ranging from 0% methanol:dichloromethane to 15% methanol:dichloromethane was performed over 30 column volumes) to yield pure 5 as a yellow solid (52 mg, 0.088 mmol, 80%).1H NMR (500 MHz, CDCl3) δ 11.26 (s, 1H), 8.11 (d, J=2.6, 1H), 7.40 (bs, 5H), 6.69 (d, J=8.4, 1H), 6.52 (d, J=3.2, 1H), 6.25 (d, J=3.2, 1H), 5.20 (t, J=6.2, 1H), 4.30 (d, J=6.4, 2H), 3.93 (s, 1H), 3.91-3.33 (m, 8H), 1.45 (s, 9H). UPLC/MS: (ES+) m/z (M+H)+; Rt= To 5 (51 mg, 0.86 mmol) in dichloromethane (800 μL), added trifluoroacetic acid (250 μL), resulting in a color change from yellow to dark brown. Solution was stirred at room temperature, open to air for 1 hr when TLC (20:1 dichloromethane/methanol) showed reaction completion. Volatiles were removed by rotary evaporation, co-evaporating several times with chloroform, resulting in 6 as a yellow solid which was used without further purification.1H NMR (400 MHz, CDCl3) δ 7.99 (s, 1H), 7.51-7.33 (m, 6H), 6.64 (d, J=8.3, 1H), 6.54 (d, J=3.1, 1H), 6.27 (s, 1H), 3.90 (s, 3H), 3.46 (s, 8H). To 2 (350 mg, 0.959 mmol 1.3 equiv)) dissolved in dichloromethane (20 mL), added diisopropylethylamine (250 μL), EDC HCl (300 mg, 1.56 mmol), HOBt (250 mg, 1.63 mmol) followed by 6 (350 mg, 0.719 mmol). After 5 hrs, TLC (20:1 dichloromethane:methanol) showed reaction completion. Reaction mixture was diluted with dichloromethane (10 mL) and washed with saturated NaHCO3(3×30 mL), 2 M HCl (1×30 mL) and brine (1×30 mL) and subsequently dried over anhydrous MgSO4. Solution was filtered and volatile solvents were removed by rotary evaporation. Crude yellow-orange product was purified by flash chromatography (flash chromatography (CombiFlash Automated Chromatographer, 24 g column; gradient elution ranging from 0% methanol: dichloromethane to 10% methanol:dichloromethane was performed over 30 column volumes) to yield pure 7 as a light yellow sticky solid (360 mg, 0.088 mmol, 60%).1H NMR (400 MHz, CDCl3) δ 11.32 (s, 1H), 8.12 (d, J=3.3, 1H), 8.07 (s, 1H), 7.38 (d, J=8.1, 6H), 6.67 (d, J=8.4, 1H), 6.51 (d, J=3.2, 1H), 6.29 (d, J=3.2, 1H), 4.52 (d, J=6.2, 2H), 4.01 (s, 2H), 3.92 (s, 3H), 3.80-3.42 (m, 28H), 3.32-3.25 (m, 2H). To a microwave vial containing 7 (200 mg, 0.240 mmol) in tBuOH/water (2 mL/2 mL), added alkyne 8 (100 mg, 0.261 mmol, 1.1 equiv), followed by 120 μL of 0.1M CuSO4and 240 μL of 0.1M sodium ascorbate. The reaction mixture was capped and heated to 130° C. for 30 minutes in microwave reactor when LC/MS showed reaction completion. Volatile solvents were removed by rotary evaporation and crude product was purified by HPLC (30-50% B, 36 min; Rt=27.03 min). Like fractions were combined and volatile solvents were removed by rotary evaporation, resulting in 9 as a yellow sticky solid (281 mg, 0.228 mmol, 95%).1H NMR (400 MHz, CDCl3) δ 11.21 (s, 1H), 9.09 (d, J=2.5, 1H), 8.77 (s, 1H), 8.18 (m, 3H), 7.74 (s, 1H), 7.52-7.34 (m, 6H), 6.92 (d, J=9.5, 1H), 6.69 (d, J=8.4, 1H), 6.54 (d, J=3.2, 1H), 6.32 (d, J=3.1, 1H), 4.65 (s, 2H), 4.58 (d, J=6.0, 2H), 4.45 (t, J=4.9, 2H), 4.08 (s, 2H), 3.94 (s, 3H), 3.83-3.28 (m, 46H). HRMS (ES+) calc'd for C58H74N10O20(M+H) m/z 1231.5154. Found 12315178; for (M+Na)+, calc'd 1253.4973, found 1253.5045. Anal. HPLC Retention Time=27.89 min at 0-60% B, 36 min To a microwave vial (2.0-5.0 mL) containing 10 (200 mg, 0.546 mmol, available from Patent No. WO 2011046946) in DMF/water (3.0/2 mL), added 5-((BOC-Amino)methyl)furan-2-boronic acid (Combi-Blocks LLC, San Diego Calif.; 241 mg, 0.622 mmol, 1.14 equiv) and NaHCO3(50 mg, 0.595 mmol, 1.1 equiv). Oxygen was removed from the solvent by bubbling nitrogen gas in solution for 10 min, to which Pd(PPh3)4(30 mg, 0.026 mmol, 5 mol %) was added. The subsequent heterogenous solution was capped and heated in microwave reactor for 15 min at 150° C. when LC/MS analysis showed reaction completion. The volatile solvents were removed by rotary evaporation and crude material was purified by flash chromatography (CombiFlash Automated Chromatographer, 24 g column; gradient elution ranging from 0% methanol:dichloromethane to 15% methanol:dichloromethane was performed over 30 column volumes) to yield pure 11 as a brown solid (225 mg, 0.467 mmol, 85%).1H NMR (400 MHz, CDCl3) δ 11.23 (s, 1H), 8.09 (s, 1H), 7.33 (d, J=8.2, 1H), 6.63 (d, J=8.2, 1H), 6.48 (d, J=3.2, 1H), 6.23 (d, J=3.2, 1H), 5.36 (s, 1H), 4.29 (s, 2H), 3.90 (brs, 5H), 3.64 (brs, 2H), 3.18 (brs, 2H), 3.09 (brs, 2H), 1.43 (s, 9H). To 11 (400 mg, 0.83 mmol) dissolved in dichloromethane (20 mL), added Fmoc-OSU (560 mg, 1.66 mmol, 2 equiv) followed by diisopropylethylamine (500 μL). Resulting yellow-orange mixture was stirred at room temperature under an atmosphere of nitrogen for 12 hr when TLC (10:1 dichloromethane-methanol) showed reaction completion. The organic mixture was washed with a saturated solution of ammonium chloride (3×20 mL) and brine (1×30 mL) and then dried over anhydrous magnesium sulfate. All volatiles were removed by rotary evaporation and crude material was purified by flash chromatography (CombiFlash Automated Chromatographer, 24 g column; flushing with dichloromethane for 5 column volumes then gradient elution ranging from 0% methanol: dichloromethane to 5% methanol: dichloromethane was performed over 20 column volumes) to yield pure 12 as a yellow solid (400 mg, 0.567 mmol, 69%).1H NMR (400 MHz, CDCl3) δ 11.25 (s, 1H), 8.12 (d, J=3.1, 1H), 7.76 (brs, 2H), 7.55 (brs, 2H), 7.42 (m, 3H), 7.31 (brs, 2H), 6.70 (d, J=8.3, 1H), 6.54 (d, J=3.1, 1H), 6.26 (d, J=3.1, 1H), 5.11 (m, 1H), 4.52 (d, J=6.2, 2H), 4.32 (d, J=6.4, 2H), 4.24 (brs, 1H), 3.94 (s, 3H), 3.42 (m, 8H), 1.47 (s, 9H). To 12 dissolved (600 mg, 0.85 mmol) in dichloromethane (15 mL), carefully added trifluoracetic acid (6 mL) and let stir at room temperature under an atmosphere of nitrogen when TLC (10:1 dichloromethane-methanol) indicated reaction completion (2 hr). All volatiles were removed by rotary evaporation and crude green residue was azeotroped with chloroform (3×10 mL) and used without further purification, yielding 13 as a TFA salt as brown solid (570 mg, 96%).1H NMR (400 MHz, CDCl3) δ 11.25 (s, 1H), 9.85 (s, 3H), 8.28 (s, 2H), 7.94 (s, 1H), 7.70 (s, 2H), 7.48 (s, 2H), 7.34 (s, 2H), 7.16 (d, J=7.5, 1H), 6.52 (d, J=8.1, 1H), 6.46 (s, 1H), 6.40 (s, 1H), 4.46 (d, J=4.4, 2H), 4.20 (s, 2H), 3.82 (s, 3H), 3.61-3.11 (m, 8H). To a solution of 13 (300 mg, 0.510 mmol) in dichloromethane (25 mL), added 2 (200 mg, 0.722 mmol, 1.4 equiv), added EDC-HCl (180 mg, 3 equiv), HOBT (150 mg, 3 equiv) and diisopropylethylamine (180 μL) and let stir under an atmosphere of nitrogen until TLC (20:1 dichloromethane-methanol) indicated reaction completion (5 hrs). The organic mixture was washed with saturated ammonium chloride (1×30 mL) and saturated sodium bicarbonate (1×30 mL) and volatiles were removed in vacuo. Resulting crude material was purified by flash chromatography (CombiFlash Automated Chromatographer, 24 g column; flushing with dichloromethane for 5 column volumes then gradient elution ranging from 0% methanol:dichloromethane to 5% methanol:dichloromethane was performed over 20 column volumes) to yield pure 14 as a yellow residue (380 mg, 0.44 mmol, 86%).1H NMR (400 MHz, CDCl3) δ 11.27 (s, 1H), 8.12 (d, J=3.2, 1H), 7.91 (t, J=6.1, 1H), 7.75 (brs, 2H), 7.55 (brs, 2H), 7.46-7.26 (m, 5H), 6.69 (d, J=8.3, 1H), 6.52 (d, J=3.2, 1H), 6.30 (d, J=3.2, 1H), 4.51 (m, 4H), 4.23 (brs, 1H), 4.04 (s, 2H), 3.94 (s, 3H), 3.75-3.26 (m, 24H). To 14 (288 mg, 0.33 mmol) in dichloromethane (2 mL), added piperadine (350 μL) and let stir under an atmosphere of nitrogen at room temperature until TLC (20:1 dichloromethane-methanol) showed reaction completion (2 hr). All volatiles were removed in vacuo and crude material was purified flash chromatography (CombiFlash Automated Chromatographer, 12 g column; gradient elution ranging from 0% methanol:dichloromethane to 20% methanol:dichloromethane was performed over 30 column volumes) to yield pure 15 as a yellow residue (170 mg, 0.265 mmol, 80%).1H NMR (400 MHz, CDCl3) δ 11.25 (s, 1H), 8.10 (s, 1H), 7.97 (s, 1H), 7.38 (d, J=8.3, 1H), 6.67 (d, J=8.4, 1H), 6.50 (d, J=3.2, 1H), 6.29 (d, J=3.2, 1H), 4.50 (d, J=6.2, 2H), 4.02 (s, 2H), 3.94 (s, 3H), 3.83-3.72 (m, 2H), 3.70-3.46 (m, 14H), 3.34-3.25 (m, 2H), 3.17-3.05 (m, 2H), 3.05-2.96 (m, 2H), 2.96-2.82 (m, 2H). To a microwave vial containing 15 (160 mg, 0.25 mmol) in water/tBuOH (2.3 mL/2.3 mL), added alkyne 8, 0.1M CuSO4(117 μL) in water and 0.1 M sodium ascorbate (234 μL). The reaction mixture was capped and heated to 130° C. for 30 minutes in microwave reactor when LC/MS showed reaction completion. Volatile solvents were removed by rotary evaporation and crude product was purified by flash chromatography (CombiFlash Automated Chromatographer, 12 g column; gradient elution ranging from 0% methanol: dichloromethane to 40% methanol: dichloromethane was performed over 140 column volumes) to yield 16 as a yellow residue (255 mg, 0.246 mmol, 98%).1H NMR (400 MHz, CDCl3) δ 11.25 (s, 1H), 9.06 (d, J=2.5, 1H), 8.75 (brs, 1H), 8.19 (d, J=9.5, 1H), 8.10 (s, 1H), 7.92 (brs, 1H), 7.66 (s, 1H), 7.37 (d, J=8.3, 1H), 6.89 (d, J=9.5, 1H), 6.67 (d, J=8.3, 1H), 6.51 (d, J=3.2, 1H), 6.28 (d, J=3.1, 1H), 4.62 (brs, 2H), 4.49 (d, J=6.1, 2H), 4.43 (t, J=5.0, 2H), 4.04 (s, 3H), 3.95 (brs, 4H), 3.91-3.41 (m, 34H), 3.13 (m, 3H). General Synthetic Procedure for Coupling of Aryl Carboxylic Acids to (16)—See Table 1. To a 16 (10 mg, 0.01 mmol) in dichloromethane (1 mL), added arene carboxylic acid (0.018 mmol, 1.8 equiv), EDC-HCl (3.5 mg, 0.018 mmol, 1.8 equiv), HOBT (3.0 mg, 0.019 mmol, 1.9 equiv) and diisopropylethylamine (104). Resulting solution was allowed to stir at room temperature under an atmosphere of nitrogen until LC/MS indicated reaction completion (5-14 hrs). Resulting solution was diluted with dichloromethane (5 mL) and then washed with a saturated solution of sodium bicarbonate (5 mL) and ammonium chloride (5 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and volatiles were removed in vacuo. Crude material was purified by HPLC. Coupling Product (18).1H NMR (400 MHz, CDCl3) δ 11.28 (s, 1H), 9.07 (s, 1H), 8.76 (s, 1H), 8.20 (d, J=9.4, 1H), 8.11 (s, 1H), 7.97 (s, 1H), 7.67 (brs, 1H), 7.43-7.30 (m, 2H), 7.22-7.09 (m, 3H), 6.90 (d, J=9.5, 1H), 6.76-6.63 (m, 1H), 6.52 (brs, 1H), 6.29 (brs, 1H), 4.63 (s, 2H), 4.47 (m, 4H), 4.05 (s, 3H), 3.96-3.33 (m, 40H), 2.32 (d, J=11.8, 3H) (Note: peak broadening effect of configurational isomerism about benzoyl-piperazine amide bond). Coupling Product (19).1H NMR (400 MHz, CDCl3) δ 11.20 (s, 1H), 9.06 (d, J=2.6, 1H), 8.74 (s, 1H), 8.20 (d, J=6.9, 1H), 8.12 (s, 1H), 8.00 (s, 1H), 7.67 (s, 1H), 7.39 (d, J=8.3, 1H), 7.33-7.27 (m, 2H), 7.21-7.13 (m, 1H), 6.89 (d, J=9.5, 1H), 6.68 (d, J=8.3, 1H), 6.52 (d, J=3.2, 1H), 6.30 (d, J=3.2, 1H), 4.63 (s, 2H), 4.50 (m, 2H), 4.43 (m, 2H), 4.07 (s, 2H), 3.95 (s, 3H), 3.78-3.42 (m, 38H), 2.38 (s, 3H). Coupling Product (20).1H NMR (400 MHz, CDCl3) δ 11.25 (s, 1H), 9.07 (d, J=2.5, 1H), 8.75 (s, 1H), 8.20 (dd, J=2.3, 9.4, 1H), 8.12 (s, 1H), 7.99 (s, 1H), 7.67 (s, 1H), 7.39 (d, J=8.3, 1H), 7.32 (m, 1H), 7.23 (m, 2H), 6.90 (d, J=9.5, 1H), 6.68 (d, J=8.4, 1H), 6.52 (d, J=3.1, 1H), 6.29 (d, J=3.0, 1H), 4.62 (s, 2H), 4.49 (d, J=5.9, 2H), 4.44 (m, 2H), 4.06 (s, 2H), 3.95 (s, 3H), 3.89-3.49 (m, 38H), 2.38 (s, 3H). Coupling Product (21).1H NMR (400 MHz, CDCl3) δ 11.21 (s, 1H), 9.05 (d, J=2.5, 1H), 8.74 (s, 1H), 8.19 (d, J=9.5, 1H), 8.11 (m, 2H), 7.63 (s, 1H), 7.36 (m, 4H), 7.24-7.18 (m, 1H), 6.88 (d, J=9.5, 1H), 6.68 (d, J=8.3, 1H), 6.52 (d, J=3.1, 1H), 6.30 (d, J=3.1, 1H), 4.59 (s, 2H), 4.52-4.24 (m, 8H), 4.08 (s, 2H), 3.94 (s, 3H), 3.85-3.33 (m, 34H), 2.68 (m, 2H), 1.24 (t, J=7.4, 3H). Coupling Product (22).1H NMR (400 MHz, CDCl3) δ 11.14 (s, 1H), 9.05 (d, J=2.5, 1H), 8.74 (s, 1H), 8.24-8.09 (m, 3H), 7.67 (s, 1H), 7.37 (m, 3H), 7.29 (brs, 1H), 6.88 (d, J=9.5, 1H), 6.69 (d, J=8.4, 1H), 6.53 (d, J=3.1, 1H), 6.31 (d, J=3.1, 1H), 4.60 (s, 2H), 4.50 (d, J=5.8, 2H), 4.41 (d, J=4.7, 2H), 4.10 (brs, 2H), 3.94 (s, 3H), 3.77-3.42 (m, 38H), 3.04-2.79 (m, 1H), 1.25 (d, J=6.5, 6H). Coupling Product (23).1H NMR (500 MHz, CDCl3) δ 11.08 (s, 1H), 9.05 (d, J=2.6, 1H), 8.73 (s, 1H), 8.19 (dd, J=2.6, 9.5, 1H), 8.13 (brs, 2H), 7.92 (m, 4H), 7.71 (s, 1H), 7.57 (s, 2H), 7.49 (d, J=8.1, 1H), 7.40 (d, J=8.3, 1H), 6.88 (d, J=9.5, 1H), 6.70 (d, J=8.1, 1H), 6.54 (d, J=3.1, 1H), 6.33 (d, J=3.2, 1H), 4.63 (s, 2H), 4.52 (s, 2H), 4.42 (s, 2H), 4.11 (s, 2H), 3.95 (s, 3H), 3.77 (d, J=5.1, 4H), 3.74-3.45 (m, 34H). Coupling Product (24).1H NMR (500 MHz, CDCl3) δ 11.27 (s, 1H), 9.07 (d, J=2.7, 1H), 8.75 (s, 1H), 8.20 (dd, J=2.6, 9.5, 1H), 8.12 (d, J=3.2, 1H), 7.91 (s, 1H), 7.66 (s, 1H), 7.39 (d, J=8.3, 1H), 7.37-7.32 (m, 1H), 7.24 (d, J=1.4, 1H), 7.02 (d, J=8.2, 1H), 6.88 (m, 2H), 6.69 (d, J=8.4, 1H), 6.52 (d, J=3.3, 1H), 6.29 (d, J=3.2, 1H), 4.61 (s, 2H), 4.49 (d, J=6.1, 2H), 4.43 (m, 2H), 4.06 (s, 3H), 3.95-3.50 (m, 40H), 2.01 (s, 1H). Coupling Product (25).1H NMR (500 MHz, CDCl3) δ 11.22 (s, 1H), 9.06 (d, J=2.6, 1H), 8.74 (s, 1H), 8.19 (d, J=9.5, 1H), 8.08 (d, J=3.3, 1H), 8.03-7.89 (brs, 1H), 7.70-7.49 (m, 1H), 7.39 (d, J=8.3, 1H), 7.25-7.2 (m, 1H), 6.91-6.87 (m, 3H), 6.68 (d, J=8.4, 1H), 6.53 (d, J=3.3, 1H), 6.31 (d, J=3.2, 1H), 4.57 (s, 2H), 4.49 (d, J=6.2, 2H), 4.45-4.25 (m, 2H), 4.07 (s, 2H), 3.94 (s, 3H), 3.83-3.39 (m, 38H), 2.01 (s, 1H). Coupling Product (26).1H NMR (400 MHz, CDCl3) δ 11.17 (s, 1H), 9.03 (d, J=2.4, 1H), 8.73 (s, 1H), 8.17 (d, J=9.5, 1H), 8.09 (brs, 2H), 7.63 (s, 1H), 7.38 (d, J=8.3, 1H), 7.31-7.27 (m, 1H), 6.88-6.82 (m, 3H), 6.68 (d, J=8.3, 1H), 6.52 (d, J=2.9, 1H), 6.31 (d, J=2.8, 1H), 4.57 (s, 2H), 4.49 (d, J=4.9, 2H), 4.44-4.22 (m, 2H), 4.08 (s, 2H), 3.93 (s, 3H), 3.83-3.43 (m, 38H). Coupling Product (27).1H NMR (400 MHz, CDCl3) δ 11.18 (s, 1H), 8.97 (d, J=2.3, 1H), 8.69 (s, 1H), 8.12 (d, J=9.5, 1H), 8.00 (m, 2H), 7.64 (brs, 1H), 7.33 (d, J=7.6, 1H), 6.83 (d, J=9.6, 1H), 6.62 (d, J=8.0, 1H), 6.50 (brs, 1H), 6.47-6.31 (m, 3H), 6.28 (brs, 1H), 4.57 (brs, 2H), 4.48 (brs, 2H), 4.43-4.31 (m, 2H), 4.04 (brs, 2H), 3.88 (s, 3H), 3.79-3.35 (m, 38H). Coupling Product (28).1H NMR (400 MHz, CDCl3) δ 11.23 (s, 1H), 9.06 (s, 1H), 8.75 (s, 1H), 8.19 (d, J=9.5, 1H), 8.11 (brs, 2H), 7.60 (s, 1H), 7.40 (m, 5H), 6.89 (d, J=9.5, 1H), 6.68 (d, J=8.4, 1H), 6.52 (s, 1H), 6.30 (s, 1H), 4.74 (s, 2H), 4.57 (s, 2H), 4.49 (s, 2H), 4.40 (s, 2H), 4.09 (s, 2H), 3.95 (s, 3H), 3.86-3.30 (m, 38H), 1.25 (s, 1H). Coupling Product (29).1H NMR (400 MHz, CDCl3) δ 11.23 (s, 1H), 9.06 (s, 1H), 8.75 (s, 1H), 8.70-8.49 (m, 1H), 8.20 (d, J=9.2, 1H), 8.13 (brs, 1H), 8.03 (brs, 1H), 7.88 (brs, 1H), 7.71 (brs, 1H), 7.63 (brs, 1H), 7.38 (m, 2H), 6.89 (d, J=9.5, 1H), 6.68 (brs, 1H), 6.52 (brs, 1H), 6.30 (brs, 1H), 4.59 (brs, 2H), 4.48 (brs, 2H), 4.41 (brs, 2H), 4.08 (brs, 2H), 3.95 (s, 3H), 3.92-3.12 (m, 38H), 2.00 (s, 3H). Coupling Product (30).1H NMR (400 MHz, CDCl3) δ 11.28 (s, 1H), 9.03 (d, J=2.6, 1H), 8.73 (m, 3H), 8.16 (m, 2H), 8.08 (s, 1H), 8.01 (m, 1H), 7.60 (brs, 1H), 7.50 (s, 1H), 7.36 (d, J=8.3, 1H), 6.86 (d, J=9.5, 1H), 6.67 (d, J=8.4, 1H), 6.50 (d, J=3.3, 1H), 6.28 (d, J=3.3, 1H), 4.53-4.42 (m, 4H), 4.35 (brs, 2H), 4.09 (brs, 2H), 3.94 (s, 3H), 3.79-3.39 (m, 38H). Coupling Product (31).1H NMR (400 MHz, CDCl3) δ 11.20 (s, 1H), 9.01 (s, 1H), 8.90 (s, 2H), 8.73 (s, 1H), 8.17 (s, 2H), 8.09 (s, 1H), 7.70 (s, 2H), 7.54 (s, 1H), 7.38 (s, 1H), 6.86 (brs, 1H), 6.69 (brs, 1H), 6.52 (brs, 1H), 6.30 (brs, 1H), 4.50 (m, 4H), 4.38 (brs, 2H), 4.11 (brs, 2H), 3.99-3.46 (m, 41H). TABLE 1Various synthesized compounds through general method as described fromcorresponding commercially available arene carboxylic acid. All activities are in μM.9181920212223CD40.295.141.360.4470.4690.9535.24MT-20.01~25~0.60.025~0.6N/A~5.02425262728293031CD40.490.200.2390.7450.4220.623.74.7MT-20.20.090.090.23~1.00.0481.39.3 SUMMARY The present invention meets the strategic need for a new treatment for HIV infection by providing bifunctional small molecules generally referred to as ARM-HI's which function through orthogonal pathways—both by inhibition the gp120-CD4 interaction, and by recruiting anti-DNP antibodies to gp120-expressing cells—in preventing the cell infection and spread of HIV. It is shown that: ARM-HI's according to the present invention exhibit substantially greater activity than ARM-H compounds previously published. The present antiviral approach has distinct advantages over other small-molecule, protein, and vaccine-based anti-HIV strategies. Although the human immune response has been demonstrated to generate neutralizing anti-gp120 antibodies around which the virus does not effectively mutate, vaccine-based approaches toward inducing such antibodies in human hosts have not yet proven successful. In theory, although the HIV virus mutates extremely rapidly in human hosts, since it must retain CD4-binding activity in order to remain infectious, antibody-recruiting small molecules that mimic the CD4 recognition motif such as the ARM-HI's of the invention have the hope of serving the same functional role as neutralizing anti-gp120 antibodies. Furthermore, as small molecules, these materials likely possess substantial advantages over protein-based therapeutics including low propensity for immunogenicity, high metabolic stability, ready large-scale production, and relatively low cost. The evidence suggests that a cellular immune response is necessary for viral inactivation in vivo, and the bifunctional small molecules of the invention have been shown to directly target gp120-expressing particles to macrophages and neutophils. This approach to antiviral therapy is also ideal as a prophylactic, as the bifunctional compound are not be expected to have any significant adverse side effects, being only active when virus is present. The complete disclosure of all patents, patent applications, and publications, and electronically available material (including, for instance, nucleotide sequence submissions in, e.g., GenBank and RefSeq, and amino acid sequence submissions in, e.g., SwissProt, PIR, PRF, PDB, and translations from annotated coding regions in GenBank and RefSeq) cited herein are incorporated by reference. Any inconsistency between the material incorporated by reference and the material set for in the specification as originally filed shall be resolved in favor of the specification as originally filed. The foregoing detailed description and examples have been given for clarity of understanding only. No unnecessary limitations are to be understood therefrom. The invention is not limited to the exact details shown and described, for variations obvious to one skilled in the art will be included within the invention defined by the following claims. All headings are for the convenience of the reader and should not be used to limit the meaning of the text that follows the heading, unless so specified.
92,943
11858918
DETAILED DESCRIPTION I. Definitions Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. A dash at the front or end of a chemical group is a matter of convenience to indicate the point of attachment to a parent moiety; chemical groups may be depicted with or without one or more dashes without losing their ordinary meaning. A prefix such as “Cu-v” or “Cu-Cv” indicates that the following group has from u to v carbon atoms, where u and v are integers. For example, “C1-6alkyl” or “C1-C6alkyl” indicates that the alkyl group has from 1 to 6 carbon atoms. “Alkyl” is a monovalent or divalent linear or branched saturated hydrocarbon radical. For example, an alkyl group can have 1 to 10 carbon atoms (i.e., C1-10alkyl) or 1 to 8 carbon atoms (i.e., C1-8alkyl) or 1 to 6 carbon atoms (i.e., C1-6alkyl) or 1 to 4 carbon atoms (i.e., C1-4alkyl). Examples of alkyl groups include, but are not limited to, methyl (Me, —CH3), ethyl (Et, —CH2CH3), 1-propyl (n-Pr, n-propyl, —CH2CH2CH3), 2-propyl (i-Pr, i-propyl, —CH(CH3)2), 1-butyl (n-Bu, n-butyl, —CH2CH2CH2CH3), 2-methyl-1-propyl (i-Bu, i-butyl, —CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, —CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, —C(CH3)3), 1-pentyl (n-pentyl, —CH2CH2CH2CH2CH3), 2-pentyl (—CH(CH3)CH2CH2CH3), 3-pentyl (—CH(CH2CH3)2), 2-methyl-2-butyl (—C(CH3)2CH2CH3), 3-methyl-2-butyl (—CH(CH3)CH(CH3)2), 3-methyl-1-butyl (—CH2CH2CH(CH3)2), 2-methyl-1-butyl (—CH2CH(CH3)CH2CH3), 1-hexyl (—CH2CH2CH2CH2CH2CH3), 2-hexyl (—CH(CH3)CH2CH2CH2CH3), 3-hexyl (—CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (—C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (—CH(CH3)CH(CH3)CH2CH3), 4-methyl-2-pentyl (—CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (—C(CH3)(CH2CH3)2), 2-methyl-3-pentyl (—CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (—C(CH3)2CH(CH3)2), 3,3-dimethyl-2-butyl (—CH(CH3)C(CH3)3, and octyl (—(CH2)7CH3). Alkyl groups can be unsubstituted or substituted. “Alkoxy” refers to the group —O-alkyl, where alkyl is as defined above. For example, C1-4alkoxy refers to an —O-alkyl group having 1 to 4 carbons. Alkoxy groups can be unsubstituted or substituted. “Alkoxyalkyl” is an alkoxy group attached to an alkyl as defined above, such that the alkyl is divalent. For example, C2-6alkoxyalkyl includes —CH2—OMe, —CH2—O-iPr, —CH2—CH2—OMe, —CH2—CH2—O—CH2—CH3, and —CH2—CH2—O-tBu. Alkoxyalkyl groups can be unsubstituted or substituted. “Hydroxyalkyl” is a hydroxy group attached to an alkyl as defined above, such that the alkyl is divalent. For example, C1-6hydroxyalkyl includes —CH2-0H, and —CH2—CH2—OH. Hydroxyalkyl groups can be unsubstituted or substituted. “Alkenyl” is a monovalent or divalent linear or branched hydrocarbon radical with at least one carbon-carbon double bond. For example, an alkenyl group can have 2 to 8 carbon atoms (i.e., C2-8alkenyl) or 2 to 6 carbon atoms (i.e., C2-6alkenyl) or 2 to 4 carbon atoms (i.e., C2-4alkenyl). Examples of alkenyl groups include, but are not limited to, ethenyl (—CH═CH2), allyl (—CH2CH═CH2), and —CH2—CH═CH—CH3. Alkenyl groups can be unsubstituted or substituted. “Alkynyl” is a monovalent or divalent linear or branched hydrocarbon radical with at least one carbon-carbon triple bond. For example, an alkynyl group can have 2 to 8 carbon atoms (i.e., C2-8alkynyl) or 2 to 6 carbon atoms (i.e., C2-6alkynyl) or 2 to 4 carbon atoms (i.e., C2-4alkynyl). Examples of alkynyl groups include, but are not limited to, acetylenyl (—C≡CH), propargyl (—CH2C≡CH), and —CH2—C≡C—CH3. Alkynyl groups can be unsubstituted or substituted. “Halogen” refers to fluoro (—F), chloro (—Cl), bromo (—Br) and iodo (—I). “Haloalkyl” is an alkyl as defined herein, wherein one or more hydrogen atoms of the alkyl are independently replaced by a halogen, which may be the same or different, such that the alkyl is divalent. The alkyl group and the halogen can be any of those described above. In some embodiments, the haloalkyl defines the number of carbon atoms in the alkyl portion, e.g., C1-4haloalkyl includes CF3, CH2F, CHF2, CH2CF3, CH2CH2CF3, CCl2CH2CH2CH3, and C(CH3)2(CF2H). Haloalkyl groups can be unsubstituted or substituted. “Haloalkoxy” is an alkoxy as defined herein, wherein one or more hydrogen atoms of the alkyl in the alkyoxy are independently replaced by a halogen, which may be the same or different, such that the alkyl is divalent. The alkoxy group and the halogen can be any of those described above. In some embodiments, the haloalkoxy defines the number of carbon atoms in the alkyl portion, e.g., C1-4haloalkoxy includes OCF3, OCH2F, OCH2CF3, OCH2CH2CF3, OCCl2CH2CH2CH3, and OC(CH3)2(CF2H). Haloalkoxy groups can be unsubstituted or substituted. “Cycloalkyl” is a monovalent or divalent single all carbon ring or a multiple condensed all carbon ring system wherein the ring in each instance is a non-aromatic saturated or unsaturated ring. For example, in some embodiments, a cycloalkyl group has 3 to 12 carbon atoms, 3 to 10 carbon atoms, 3 to 8 carbon atoms, 3 to 6 carbon atoms, 3 to 5 carbon atoms, or 3 to 4 carbon atoms. Exemplary single ring cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, and cyclooctyl. Cycloalkyl also includes multiple condensed ring systems (e.g., ring systems comprising 2 rings) having about 7 to 12 carbon atoms. The rings of the multiple condensed ring system can be connected to each other via fused, spiro, or bridged bonds when allowed by valency requirements. Exemplary multiple ring cycloalkyl groups include octahydropentalene, bicyclo[2. 2. 1]heptane, bicyclo[2. 2. 2]octane, bicyclo[2. 2. 2]oct-2-ene, and spiro[2. 5]octane. Cycloalkyl groups can be unsubstituted or substituted. “Alkylcycloalkyl” refers to an alkyl as defined herein, wherein one or more hydrogen atoms of the alkyl are independently replaced by a cycloalkyl group, which may be the same or different. The alkyl group and the cycloalkyl group can be any of those described above. In some embodiments, the number of carbon atoms in the alkyl and cycloalkyl portion can be designated separately, e.g., C1-6alkyl-C3-12cycloalkyl. Alkylcycloalkyl groups can be unsubstituted or substituted. “Aryl” as used herein refers to a monovalent or divalent single all carbon aromatic ring or a multiple condensed all carbon ring system wherein the ring is aromatic. For example, in some embodiments, an aryl group has 6 to 20 carbon atoms, 6 to 14 carbon atoms, 6 to 12 carbon atoms, or 6 to 10 carbon atoms. Aryl includes a phenyl radical. Aryl also includes multiple condensed ring systems (e.g., ring systems comprising 2, 3 or 4 rings) having about 9 to 20 carbon atoms in which multiple rings are aromatic. The rings of the multiple condensed ring system can be connected to each other via fused, spiro, or bridged bonds when allowed by valency requirements It is also understood that when reference is made to a certain atom-range membered aryl (e.g., 6-10 membered aryl), the atom range is for the total ring atoms of the aryl. For example, a 6-membered aryl would include phenyl and a 10-membered aryl would include naphthyl. Non-limiting examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, and the like. Aryl groups can be unsubstituted or substituted. “Alkylaryl” refers to an alkyl as defined herein, wherein one or more hydrogen atoms of the alkyl are independently replaced by an aryl group, which may be the same or different. The alkyl group and the aryl group can be any of those described above, such that the alkyl is divalent. In some embodiments, an alkylaryl group has 7 to 24 carbon atoms, 7 to 16 carbon atoms, 7 to 13 carbon atoms, or 7 to 11 carbon atoms. An alkylaryl group defined by the number of carbon atoms refers to the total number of carbon atoms present in the constitutive alkyl and aryl groups combined. For example, C7alkylaryl refers to benzyl, while C11alkylaryl includes 1-methylnaphthyl and n-pentylphenyl. In some embodiments the number of carbon atoms in the alkyl and aryl portion can be designated separately, e.g., C1-6alkyl-C6-10aryl. Non-limiting examples of alkylaryl groups include, but are not limited to, benzyl, 2,2-dimethylphenyl, n-pentylphenyl, 1-methylnaphthyl, 2-ethylnaphthyl, and the like. Alkylaryl groups can be unsubstituted or substituted. “Heterocyclyl” or “heterocycle” or “heterocycloalkyl” as used herein refers to a single saturated or partially unsaturated non-aromatic ring or a non-aromatic multiple ring system that has at least one heteroatom in the ring (i.e., at least one annular (i.e., ring-shaped) heteroatom selected from oxygen, nitrogen, and sulfur). Unless otherwise specified, a heterocyclyl group has from 3 to about 20 annular atoms, for example from 3 to 12 annular atoms, for example from 4 to 12 annular atoms, 4 to 10 annular atoms, or 3 to 8 annular atoms, or 3 to 6 annular atoms, or 3 to 5 annular atoms, or 4 to 6 annular atoms, or 4 to 5 annular atoms. Thus, the term includes single saturated or partially unsaturated rings (e.g., 3, 4, 5, 6 or 7-membered rings) having from about 1 to 6 annular carbon atoms and from about 1 to 3 annular heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur in the ring. The rings of the multiple condensed ring system can be connected to each other via fused, spiro, or bridged bonds when allowed by valency requirements. Heterocycles include, but are not limited to, azetidine, aziridine, imidazolidine, morpholine, oxirane (epoxide), oxetane, thietane, piperazine, piperidine, pyrazolidine, piperidine, pyrrolidine, pyrrolidinone, tetrahydrofuran, tetrahydrothiophene, dihydropyridine, tetrahydropyridine, quinuclidine, 2-oxa-6-azaspiro[3. 3]heptan-6-yl, 6-oxa-1-azaspiro[3. 3]heptan-1-yl, 2-thia-6-azaspiro[3. 3]heptan-6-yl, 2,6-diazaspiro[3. 3]heptan-2-yl, 2-azabicyclo[3. 1. 0]hexan-2-yl, 3-azabicyclo[3. 1. 0]hexanyl, 2-azabicyclo[2. 1. 1]hexanyl, 2-azabicyclo[2. 2. 1]heptan-2-yl, 4-azaspiro[2. 4]heptanyl, 5-azaspiro[2. 4]heptanyl, and the like. Heterocyclyl groups can be unsubstituted or substituted. “Alkylheterocyclyl” refers to an alkyl as defined herein, wherein one or more hydrogen atoms of the alkyl are independently replaced by a heterocyclyl group, which may be the same or different. The alkyl group and the heterocyclyl group can be any of those described above, such that the alkyl is divalent. In some embodiments, the number of atoms in the alkyl and heterocyclyl portion can be designated separately, e.g., C1-6alkyl-3 to 12 membered heterocyclyl having one to three heteroatoms each independently N, O, or S. Alkylheterocyclyl groups can be unsubstituted or substituted. “Heteroaryl” refers to a single aromatic ring that has at least one atom other than carbon in the ring, wherein the atom is selected from the group consisting of oxygen, nitrogen and sulfur; “heteroaryl” also includes multiple condensed ring systems that have at least one such aromatic ring, which multiple condensed ring systems are further described below. Thus, “heteroaryl” includes single aromatic rings of from about 1 to 6 carbon atoms and about 1-4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur. The sulfur and nitrogen atoms may also be present in an oxidized form provided the ring is aromatic. Exemplary heteroaryl ring systems include but are not limited to pyridyl, pyrimidinyl, oxazolyl or furyl. “Heteroaryl” also includes multiple condensed ring systems (e.g., ring systems comprising 2, 3 or 4 rings) wherein a heteroaryl group, as defined above, is condensed with one or more rings selected from heteroaryls (to form for example 1,8-naphthyridinyl) and aryls (to form, for example, benzimidazolyl or indazolyl) to form the multiple condensed ring system. Thus, a heteroaryl (a single aromatic ring or multiple condensed ring system) can have about 1-20 carbon atoms and about 1-6 heteroatoms within the heteroaryl ring. For example, tetrazolyl has 1 carbon atom and 4 nitrogen heteroatoms within the ring. The rings of the multiple condensed ring system can be connected to each other via fused, spiro, or bridged bonds when allowed by valency requirements. It is to be understood that the individual rings of the multiple condensed ring system may be connected in any order relative to one another. It is to be understood that the point of attachment for a heteroaryl or heteroaryl multiple condensed ring system can be at any suitable atom of the heteroaryl or heteroaryl multiple condensed ring system including a carbon atom and a heteroatom (e.g., a nitrogen). It also to be understood that when a reference is made to a certain atom-range membered heteroaryl (e.g., a 5 to 10 membered heteroaryl), the atom range is for the total ring atoms of the heteroaryl and includes carbon atoms and heteroatoms. It is also to be understood that the rings of the multiple condensed ring system may include an aryl ring fused to a heterocyclic ring with saturated or partially unsaturated bonds (e.g., 3, 4, 5, 6 or 7-membered rings) having from about 1 to 6 annular carbon atoms and from about 1 to 3 annular heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur in the ring. For example, a 5-membered heteroaryl includes thiazolyl and a 10-membered heteroaryl includes quinolinyl. Exemplary heteroaryls include but are not limited to pyridyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrazolyl, thienyl, indolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, furyl, oxadiazolyl, thiadiazolyl, quinolyl, isoquinolyl, benzothiazolyl, benzoxazolyl, indazolyl, quinoxalyl, quinazolyl, benzofuranyl, benzimidazolyl, thianaphthenyl, pyrrolo[2,3-b]pyridinyl, quinazolinyl-4(3H)-one, triazolyl, and tetrazolyl. Heteroaryl groups can be unsubstituted or substituted. “Alkylheteroaryl” refers to an alkyl as defined herein, wherein one or more hydrogen atoms of the alkyl are independently replaced by a heteroaryl group, which may be the same or different, such that the alkyl is divalent. The alkyl group and the heteroaryl group can be any of those described above. In some embodiments, the number of atoms in the alkyl and heteroaryl portion are designated separately, e.g., C1-6alkyl-5 to 10 membered heteroaryl having one to four heteroatoms each independently N, O, or S. Alkylheteroaryl groups can be unsubstituted or substituted. “Oxo” as used herein refers to ═O. “Substituted” as used herein refers to wherein one or more hydrogen atoms of the group are independently replaced by one or more substituents (e.g., 1, 2, 3, or 4 or more) as indicated. A “compound of the present disclosure” includes compounds disclosed herein, for example a compound of the present disclosure includes compounds of Formula (I), including the compounds of the Examples. In some embodiments, a “compound of the present disclosure” includes compounds of Formula (I). “Pharmaceutically acceptable excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals. “Therapeutically effective amount” or “effective amount” as used herein refers to an amount that is effective to elicit the desired biological or medical response, including the amount of a compound that, when administered to a subject for treating a disease, is sufficient to affect such treatment for the disease. The effective amount will vary depending on the compound, the disease, and its severity and the age, weight, etc., of the subject to be treated. The effective amount can include a range of amounts. As is understood in the art, an effective amount may be in one or more doses, i.e., a single dose or multiple doses may be required to achieve the desired treatment endpoint. An effective amount may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable or beneficial result may be or is achieved. Suitable doses of any co-administered compounds may optionally be lowered due to the combined action (e.g., additive or synergistic effects) of the compounds. “Co-administration” as used herein refers to administration of unit dosages of the compounds disclosed herein before or after administration of unit dosages of one or more additional therapeutic agents, for example, administration of the compound disclosed herein within seconds, minutes, or hours of the administration of one or more additional therapeutic agents. For example, in some embodiments, a unit dose of a compound of the present disclosure is administered first, followed within seconds or minutes by administration of a unit dose of one or more additional therapeutic agents. Alternatively, in other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound of the present disclosure within seconds or minutes. In some embodiments, a unit dose of a compound of the present disclosure is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of one or more additional therapeutic agents. In other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound of the present disclosure. Co-administration of a compound disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents, such that therapeutically effective amounts of each agent are present in the body of the subject. Provided are also pharmaceutically acceptable salts, hydrates, solvates, tautomeric forms, polymorphs, and prodrugs of the compounds described herein. “Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human pharmaceutical use. The compounds described herein may be prepared and/or formulated as pharmaceutically acceptable salts or when appropriate as a free base. Pharmaceutically acceptable salts are non-toxic salts of a free base form of a compound that possesses the desired pharmacological activity of the free base. These salts may be derived from inorganic or organic acids or bases. For example, a compound that contains a basic nitrogen may be prepared as a pharmaceutically acceptable salt by contacting the compound with an inorganic or organic acid. Non-limiting examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, methylsulfonates, propylsulfonates, besylates, xylenesulfonates, naphthalene-1-sulfonates, naphthalene-2-sulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ-hydroxybutyrates, glycolates, tartrates, and mandelates. Lists of other suitable pharmaceutically acceptable salts are found in Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams and Wilkins, Philadelphia, Pa., 2006. Examples of “pharmaceutically acceptable salts” of the compounds disclosed herein also include salts derived from an appropriate base, such as an alkali metal (for example, sodium, potassium), an alkaline earth metal (for example, magnesium), ammonium and N(C1≡C4alkyl)4+. Also included are base addition salts, such as sodium or potassium salts. Provided are also compounds described herein or pharmaceutically acceptable salts, isomers, or a mixture thereof, in which from 1 to n hydrogen atoms attached to a carbon atom may be replaced by a deuterium atom or D, in which n is the number of hydrogen atoms in the molecule. As known in the art, the deuterium atom is a non-radioactive isotope of the hydrogen atom. Such compounds may increase resistance to metabolism, and thus may be useful for increasing the half-life of the compounds described herein or pharmaceutically acceptable salts, isomer, or a mixture thereof when administered to a mammal. See, e.g., Foster, “Deuterium Isotope Effects in Studies of Drug Metabolism”, Trends Pharmacol. Sci., 5(12):524-527 (1984). Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogen atoms have been replaced by deuterium. Examples of isotopes that can be incorporated into the disclosed compounds also include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as2H,3H,11C,13C,14C,13N,15N,15O,17O,18O,31P,32P,35S,18F,36Cl,123I, and125I, respectively. Substitution with positron emitting isotopes, such as11C,18F,15O and13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of Formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed. The compounds of the embodiments disclosed herein, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (−), (R)- and (S)-, or (D)- and (L)-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high-pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. Where compounds are represented in their chiral form, it is understood that the embodiment encompasses, but is not limited to, the specific diastereomerically or enantiomerically enriched form. Where chirality is not specified but is present, it is understood that the embodiment is directed to either the specific diastereomerically or enantiomerically enriched form; or a racemic or scalemic mixture of such compound(s). As used herein, “scalemic mixture” is a mixture of stereoisomers at a ratio other than 1:1. “Stereoisomer” as used herein refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are non-superimposable mirror images of one another. “Tautomer” as used herein refers to a proton shift from one atom of a molecule to another atom of the same molecule. In some embodiments, the present disclosure includes tautomers of said compounds. “Solvate” as used herein refers to the result of the interaction of a solvent and a compound. Solvates of salts of the compounds described herein are also provided. Hydrates of the compounds described herein are also provided. “Hydrate” as used herein refers to a compound of the disclosure that is chemically associated with one or more molecules of water. “Prevention” or “preventing” means any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop. Compounds may, in some embodiments, be administered to a subject (including a human) who is at risk or has a family history of the disease or condition. “Prodrug” as used herein refers to a derivative of a drug that upon administration to the human body is converted to the parent drug according to some chemical or enzymatic pathway. In some embodiments, a prodrug is a biologically inactive derivative of a drug that upon administration to the human body is converted to the biologically active parent drug according to some chemical or enzymatic pathway. “Treatment” or “treat” or “treating” as used herein refers to an approach for obtaining beneficial or desired results. For purposes of the present disclosure, beneficial or desired results include, but are not limited to, alleviation of a symptom and/or diminishment of the extent of a symptom and/or preventing a worsening of a symptom associated with a disease or condition. In one embodiment, “treatment” or “treating” includes one or more of the following: a) inhibiting the disease or condition (e.g., decreasing one or more symptoms resulting from the disease or condition, and/or diminishing the extent of the disease or condition); b) slowing or arresting the development of one or more symptoms associated with the disease or condition (e.g., stabilizing the disease or condition, delaying the worsening or progression of the disease or condition); and c) relieving the disease or condition, e.g., causing the regression of clinical symptoms, ameliorating the disease state, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival. “At risk individual” as used herein refers to an individual who is at risk of developing a condition to be treated. An individual “at risk” may or may not have detectable disease or condition and may or may not have displayed detectable disease prior to the treatment of methods described herein. “At risk” denotes that an individual has one or more so-called risk factors, which are measurable parameters that correlate with development of a disease or condition and are known in the art. An individual having one or more of these risk factors has a higher probability of developing the disease or condition than an individual without these risk factor(s). II. Compounds In one embodiment, the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt thereof, whereinR1isi) C6-10aryl or 6-membered heteroaryl, each of which is optionally substituted with one to four R4; orii) 6-membered heteroaryl, wherein the heteroaryl is fused to a 5- or 6-membered ring having zero to three heteroatoms, each independently N, O, or S, to form a fused ring system, wherein the fused ring system is optionally substituted with one to four R5;ring A is which are each optionally substituted with one to three RA, each RAindependently C1-6alkyl, C1-6haloalkyl, C1-6alkoxy, C1-6haloalkoxy, halogen, —OH, —CN, or —N(R10a)(R10b);ring B is(i) C6-10aryl or heteroaryl, which is each optionally substituted with one to four RB, each RBindependently C1-9alkyl, C1-8haloalkyl, C1-6haloalkoxy, C2-6alkoxyalkyl, C2-6alkenyl, C2-6alkynyl, halogen, C3-10cycloalkyl, heterocyclyl, C6-10aryl, heteroaryl, oxo, —NO2, —CN, —N3, —O—R10a, —C(O)R10a, —C(O)O— R10a, —N(R10a)(R10b), N(R10a)2(R10b)+, —N(R10a)C(O)—R10b, —N(R10a)C(O)O— R10b, —N(R10a)C(O)N(R10b)(R10c), —N(R10a)S(O)2(R10b), —NR10aS(O)2N(R10b)(R10c), —NR10aS(O)2O(R10b), —OC(O)R10a, —OC(O)O R10a, —OC(O)—N(R10a)(R10b), —S—R10a, —S(O) R10a, —S(O)(NH)Rub, —S(O)2R10a, —S(O)2N(R10a)(R10b), —S(O)(NR10a) R10b, or —Si(R10a)3; or(ii) C6-10aryl or heteroaryl, which is each fused to a 5 or 6 membered ring having zero to four heteroatoms, each independently N, O, or S, to form a fused ring system, wherein the fused ring system is optionally substituted with one to five RB, each RBindependently C1-9alkyl, C1-8haloalkyl, C1-6haloalkoxy, C2-6alkoxyalkyl, C2-6alkenyl, C2-6alkynyl, halogen, C3-10cycloalkyl, heterocyclyl, C6-10aryl, heteroaryl, oxo, —NO2, —CN, —N3, —O—R10a, —C(O)R10a, —C(O)O— R10a, —N(R10a)(R10b), N(R10a)2(R10b)+, —N(R10a)C(O)—R10b, —N(R10a)C(O)O— R10b, —N(R10a)C(O)N(R10b)(R10c), —N(R10a)S(O)2(R10b), —NR10aS(O)2N(R10b)(R10c), —NR10aS(O)2O(R10b), —OC(O) R10a, —OC(O)O R10a, —OC(O)—N(R10a)(R10b), —S—R10a, —S(O) R10a, —S(O)(NH) R10a, —S(O)2R10a, —S(O)2N(R10a)(R10b), —S(O)(NR10a) R10b, or —Si(R10aV is —C(R11a)(R11b)—;X1, X2, and X3are each independently —C(H)═, or —C(R8)═;R2isi) C3-10cycloalkyl or heterocyclyl, wherein the cycloalkyl or heterocyclyl is each optionally substituted with one to four Z1, wherein each Z1is independently C1-6alkyl, C1-6alkoxy, C1-6hydroxyalkyl, C2-6alkoxyalkyl, halogen, C1-6haloalkyl, C1-6haloalkoxy, oxo, —OH, —CN, C1-6alkyl-CN, —C(O)R10a, —C(O)O— R10a, —C(O)NH2, —C(O)NH(C1-9alkyl), —N(R10a)(R10b), —N(R10a)C(O)O— R10b, —N(R10a)C(O)—R10b, —S(O)2R10a, —S(O)2(C1-9alkyl), —O—C3-6cycloalkyl or heteroaryl,wherein each —O—C3-6cycloalkyl or heteroaryl is optionally substituted with one to four Z1agroups, each Z1aindependently C1-6alkyl, C1-6alkoxy, halogen, C1-6haloalkyl, or C1-6haloalkoxy; or whereinR2ais C1-6alkyl, C1-6hydroxyalkyl, C2-6alkoxyalkyl, C1-6haloalkyl, C1-6alkyl-CN, —C3-6cycloalkyl or heteroaryl,wherein each alkyl, cycloalkyl, or heteroaryl is optionally substituted with one to four groups each independently C1-6alkyl, C1-6alkoxy, halogen, C1-6haloalkyl, or C1-6haloalkoxy;R2bis H, C1-3alkyl, C1-3haloalkyl; andR2cis H, C1-3alkyl, C1-3haloalkyl;R3is —C(O)OR3a;wherein R3ais H, C1-4alkyl-N(R9a)(R9b), —C1-4alkyl-N(R9a)C(O)—O—C1-4alkyl-OP(O)(OR9c)2, C1-4alkyl-C(O)N(R9a)(R9b), —C1-4alkyl-O—C(O)—C1-4alkyl, —C1-4alkyl-O—C(O)—O—C1-4alkyl, —C1-4alkyl-O—C(O)—C1-4alkyl-N(R9a)(R9b), —C1-4alkyl-O—C(O)—C1-4alkyl-OP(O)(OR9c)2, —CH2CH(N(R9a)2)C(O)OR9b, —P(O)(OR9c)2, —OP(O)(OR9c)2, —CH2P(O)(OR9c)2, —CH2OP(O)(OR9c)2, —OCH2P(O)(OR9c)2, C(O)OCH2P(O)(OR9c)2, —P(O)(R9c)(OR9d), —OP(O)(R9c)(OR9d), —CH2P(O)(R9c)(OR9d), —OCH2P(O)(R9c)(OR9d), —C(O)OCH2P(O)(R9c)(OR9d), —P(O)(N(R9c)2)2, —OP(O)(N(R9c)2)2, —CH2P(O)(N(R9c)2)2, —OCH2P(O)(N(R9c)2)2, —C(O)OCH2P(O)(N(R9c)2)2, —P(O)(N(R9c)2)(OR9d), —OP(O)(N(R9c)2)(OR9d), —CH2P(O)(N(R9c)2)(OR9d), —OCH2P(O)(N(R9c)2)(OR9d), —C(O)OCH2P(O)(N(R9c)2)(OR9d), —P(O)(R9c)(N(R9d)2), —OP(O)(R9c)(N(R9d)2), —CH2P(O)(R9c)(N(R9d)2), —OCH2P(O)(R9c)(N(R9d)2), —C(O)OCH2P(O)(R9)(N(R9d)2), or C1-6alkyl-heterocyclyl,wherein each alkyl or heterocyclyl is optionally substituted with one to four halogens;each R4is independently C1-6alkyl, C1-8haloalkyl, C1-6haloalkoxy, C2-6alkoxyalkyl, C2-6alkenyl, halogen, C3-10cycloalkyl, heterocyclyl, C6-10aryl, heteroaryl, oxo, —NO2, —CN, —N3, —O—R10a, —C(O)R10a, —C(O)O—R10a, —N(R10a)(R10b), —N(R10a)2(R10b), —N(R10a)—C(O)R10b, —N(R10a)C(O)O(R10b), —N(R10a)C(O)N(R10b)(R10c), —N(R10a)S(O)2(R10b), —N(R10a)S(O)2—N(R10b)(R10c), —N(R10a)S(O)2O(R10b), —OC(O)R10a, —OC(O)OR10a, —OC(O)—N(R10a)(R10b), —S—R10a, —S(O)R10a, —S(O)(NH)R10a, —S(O)2R10a, —S(O)2N(R10a)(R10b), —S(O)(NR10a)R10b, or —Si(R10a)3wherein each alkyl, haloalkyl, alkenyl or aryl is optionally substituted with one to four R5, andwherein each cycloalkyl, heterocyclyl or heteroaryl is optionally substituted with one to four R12;each R5is independently C1-9alkyl, C1-8haloalkyl, C1-6alkoxy, C1-6haloalkoxy, C2-6alkoxyalkyl, C2-6alkenyl, C2-6alkynyl, halogen, C3-15cycloalkyl, heterocyclyl, C6-10aryl, heteroaryl, oxo, —NO2, —N3, —CN, —O—R10a, —C(O)—R10a, —C(O)O—R10a, C(O)—N(R10a)(R10b), —N(R10a)(R10b), —N(R10a)2(R10b), —N(RR10a)C(O)—R10b, —N(R10a)C(O)O— R10b, —N(R10a)C(O)N(R10b)(R10c), —N(R10a)S(O)2(R10b), —NR10aS(O)2N(R10b)(R10c), —NR10aS(O)2O(R10b), —OC(O) R10a, —OC(O)O R10a, —OC(O)—N(R10a)(R10b), —S—R10a, —S(O) R10a, —S(O)(NH) R10a, —S(O)2R10a, —S(O)2N(R10a)(R10b), —S(O)(NR10a) R10b, or —Si(R10a)3wherein each alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl is optionally substituted with one to four R6;each R6is independently C1-9alkyl, C1-8haloalkyl, C1-8alkoxy, C1-6haloalkoxy, C2-6alkoxyalkyl, C2-6alkenyl, C2-6alkynyl, C3-15cycloalkyl, halogen, C3-15cycloalkyl, heterocyclyl, C6-10aryl, heteroaryl, oxo, —NO2, —N3, —CN, —O— R10a, —C(O)—R10a, —C(O)O—R10a, —C(O)—N(R10a)(R10b), —N(R10a)(R10b), —N(R10a)C(O)—R10b, —N(R10a)C(O)O— R10b, —N(R10a)C(O)N(R10b)(R10c), —N(R10a)S(O)2(R10b), —NR10aS(O)2N(R10b)(R10c), —NR10aS(O)2O(R10b), —OC(O) R10a, —OC(O)O R10a, —OC(O)—N(R10a)(R10b), —S—R10a, —S(O) R10a, —S(O)(NH) R10a, —S(O)2R10a, —S(O)2N(R10a)(R10b), —S(O)(NR10a) R10b, or —Si(R10a)3wherein each alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl is optionally substituted with one to four R7;each R7and R8is independently C1-9alkyl, C1-6alkoxy, C2-6alkoxyalkyl, C1-8haloalkyl, C2-6alkenyl, C2-6alkynyl, halogen, C3-15cycloalkyl, heterocyclyl, C6-10aryl, heteroaryl, oxo, —OH, —CN, —NO2, —NH2, —N3, —SH, —O(C1-9alkyl), —O(C1-8haloalkyl), —O(C2-6alkenyl), —O(C2-6alkynyl), —O(C3-15cycloalkyl), —O(heterocyclyl), —O(C6-10aryl), —O(heteroaryl), —NH(C1-9alkyl), —NH(C1-8haloalkyl), —NH(C2-6alkenyl), —NH(C2-6alkynyl), —NH(C3-15cycloalkyl), —NH(heterocyclyl), —NH(C6-10aryl), —NH(heteroaryl), —N(C1-9alkyl)2, —N(C1-8haloalkyl)2, —N(C2-6alkenyl)2, —N(C2-6alkynyl)2, —N(C3-15cycloalkyl)2, —N(heterocyclyl)2, —N(C6-10aryl)2, —N(heteroaryl)2, —N(C1-9alkyl)(C1-8haloalkyl), —N(C1-9alkyl)(C2-6alkenyl), —N(C1-9alkyl)(C2-6alkynyl), —N(C1-9alkyl)(C3-15cycloalkyl), —N(C1-9alkyl)(heterocyclyl), —N(C1-9alkyl)(C6-10aryl), —N(C1-9alkyl)(heteroaryl), —C(O)(C1-9alkyl), —C(O)(C1-8haloalkyl), —C(O)(C2-6alkenyl), —C(O)(C2-6alkynyl), —C(O)(C3-15cycloalkyl), —C(O)(heterocyclyl), —C(O)(C6-10aryl), —C(O)(heteroaryl), —C(O)O(C1-9alkyl), —C(O)O(C1-8haloalkyl), —C(O)O(C2-6alkenyl), —C(O)O(C2-6alkynyl), —C(O)O(C3-15cycloalkyl), —C(O)O(heterocyclyl), —C(O)O(C6-10aryl), —C(O)O(heteroaryl), —C(O)NH2, —C(O)NH(C1-9alkyl), —C(O)NH(C1-8haloalkyl), —C(O)NH(C2-6alkenyl), —C(O)NH(C2-6alkynyl), —C(O)NH(C3-15cycloalkyl), —C(O)NH(heterocyclyl), —C(O)NH(C6-10aryl), —C(O)NH(heteroaryl), —C(O)N(C1-9alkyl)2, —C(O)N(C1-8haloalkyl)2, —C(O)N(C2-6alkenyl)2, —C(O)N(C2-6alkynyl)2, —C(O)N(C3-15cycloalkyl)2, —C(O)N(heterocyclyl)2, —C(O)N(C6-10aryl)2, —C(O)N(heteroaryl)2, —NHC(O)(C1-9alkyl), —NHC(O)(C1-8haloalkyl), —NHC(O)(C2-6alkenyl), —NHC(O)(C2-6alkynyl), —NHC(O)(C3-15cycloalkyl), —NHC(O)(heterocyclyl), —NHC(O)(C6-10aryl), —NHC(O)(heteroaryl), —NHC(O)O(C1-9alkyl), —NHC(O)O(C1-8haloalkyl), —NHC(O)O(C2-6alkenyl), —NHC(O)O(C2-6alkynyl), —NHC(O)O(C3-15cycloalkyl), —NHC(O)O(heterocyclyl), —NHC(O)O(C6-10aryl), —NHC(O)O(heteroaryl), —NHC(O)NH(C1-9alkyl), —NHC(O)NH(C1-8haloalkyl), —NHC(O)NH(C2-6alkenyl), —NHC(O)NH(C2-6alkynyl), —NHC(O)NH(C3-15cycloalkyl), —NHC(O)NH(heterocyclyl), —NHC(O)NH(C6-10aryl), —NHC(O)NH(heteroaryl), —NHS(O)(C1-9alkyl), —N(C1-9alkyl)(S(O)(C1-9alkyl), —S(C1-9alkyl), —S(C1-8haloalkyl), —S(C2-6alkenyl), —S(C2-6alkynyl), —S(C3-15cycloalkyl), —S(heterocyclyl), —S(C6-10aryl), —S(heteroaryl), —S(O)N(C1-9alkyl)2, —S(O)(C1-9alkyl), —S(O)(C1-8haloalkyl), —S(O)(C2-6alkenyl), —S(O)(C2-6alkynyl), —S(O)(C3-15cycloalkyl), —S(O)(heterocyclyl), —S(O)(C6-10aryl), —S(O)(heteroaryl), —S(O)2(C1-9alkyl), —S(O)2(C1-8haloalkyl), —S(O)2(C2-6alkenyl), —S(O)2(C2-6alkynyl), —S(O)2(C3-15cycloalkyl), —S(O)2(heterocyclyl), —S(O)2(C6-10aryl), —S(O)2(heteroaryl), —S(O)(NH)(C1-9alkyl), —S(O)2NH(C1-9alkyl), or —S(O)2N(C1-9alkyl)2,wherein each alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl is optionally substituted with one to three C1-9alkyl, C1-8haloalkyl, halogen, —OH, —NH2, CO2H, —O(C1-9alkyl), —O(C1-8haloalkyl), —O(C3-15cycloalkyl), —O(heterocyclyl), —O(aryl), —O(heteroaryl), —NH(C1-9alkyl), —NH(C1-8haloalkyl), —NH(C3-15cycloalkyl), —NH(heterocyclyl), —NH(aryl), —NH(heteroaryl), —N(C1-9alkyl)2, —N(C3-15cycloalkyl)2, —NHC(O)(C1-8haloalkyl), —NHC(O)(C3-15cycloalkyl), —NHC(O)(heterocyclyl), —NHC(O)(aryl), —NHC(O)(heteroaryl), —NHC(O)O(C1-9alkyl), —NHC(O)O(C1-8haloalkyl), —NHC(O)O(C2-6alkynyl), —NHC(O)O(C3-15cycloalkyl), —NHC(O)O(heterocyclyl), —NHC(O)O(aryl), —NHC(O)O(heteroaryl), —NHC(O)NH(C1-9alkyl), S(O)2(C1-9alkyl), —S(O)2(C1-8haloalkyl), —S(O)2(C3-15cycloalkyl), —S(O)2(heterocyclyl), —S(O)2(aryl), —S(O)2(heteroaryl), —S(O)(NH)(C1-9alkyl), —S(O)2NH(C1-9alkyl), or —S(O)2N(C1-9alkyl)2,wherein each alkyl or heterocyclyl is optionally substituted with one to four halogens;each R9aand R9bis independently H, C1-6alkyl, or C1-6haloalkyl, or R9aand R9btogether form a 6-membered heterocyclyl;each R9c, R9d, R10a, R10b, and R10cis independently H, C1-9alkyl, C2-6alkenyl, C2-6alkynyl, C3-15cycloalkyl, heterocyclyl, C6-10aryl, or heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl is each optionally substituted with one to four R6;each R11aand R11bis independently —H, C1-6alkyl, oxo, or halogen;each R12is C1-9alkyl, C1-8haloalkyl, C2-6alkenyl, C2-6alkynyl, halogen, oxo, —OH, —CN, —NO2, —NH2, —N3, —SH, —O(C1-9alkyl), —O(C1-8haloalkyl), —O(C2-6alkenyl), —O(C2-6alkynyl), —O(C3-15cycloalkyl), —O(heterocyclyl), —O(C6-10aryl), —O(heteroaryl), —NH(C1-9alkyl), —NH(C1-8haloalkyl), —NH(C2-6alkenyl), —NH(C2-6alkynyl), —NH(C3-15cycloalkyl), —NH(heterocyclyl), —NH(C6-10aryl), —NH(heteroaryl), —N(C1-9alkyl)2, —N(C1-8haloalkyl)2, —N(C2-6alkenyl)2, —N(C2-6alkynyl)2, —N(C3-15cycloalkyl)2, —N(heterocyclyl)2, —N(C6-10aryl)2, —N(heteroaryl)2, —N(C1-9alkyl)(C1-8haloalkyl), —N(C1-9alkyl)(C2-6alkenyl), —N(C1-9alkyl)(C2-6alkynyl), —N(C1-9alkyl)(C3-15cycloalkyl), —N(C1-9alkyl)(heterocyclyl), —N(C1-9alkyl)(C6-10aryl), —N(C1-9alkyl)(heteroaryl), —C(O)(C1-9alkyl), —C(O)(C1-8haloalkyl), —C(O)(C2-6alkenyl), —C(O)(C2-6alkynyl), —C(O)(C3-15cycloalkyl), —C(O)(heterocyclyl), —C(O)(C6-10aryl), —C(O)(heteroaryl), —C(O)O(C1-9alkyl), —C(O)O(C1-8haloalkyl), —C(O)O(C2-6alkenyl), —C(O)O(C2-6alkynyl), —C(O)O(C3-15cycloalkyl), —C(O)O(heterocyclyl), —C(O)O(C6-10aryl), —C(O)O(heteroaryl), —C(O)NH2, —C(O)NH(C1-9alkyl), —C(O)NH(C1-8haloalkyl), —C(O)NH(C2-6alkenyl), —C(O)NH(C2-6alkynyl), —C(O)NH(C3-15cycloalkyl), —C(O)NH(heterocyclyl), —C(O)NH(C6-10aryl), —C(O)NH(heteroaryl), —C(O)N(C1-9alkyl)2, —C(O)N(C1-8haloalkyl)2, —C(O)N(C2-6alkenyl)2, —C(O)N(C2-6alkynyl)2, —C(O)N(C3-15cycloalkyl)2, —C(O)N(heterocyclyl)2, —C(O)N(C6-10aryl)2, —C(O)N(heteroaryl)2, —NHC(O)(C1-9alkyl), —NHC(O)(C1-8haloalkyl), —NHC(O)(C2-6alkenyl), —NHC(O)(C2-6alkynyl), —NHC(O)(C3-15cycloalkyl), —NHC(O)(heterocyclyl), —NHC(O)(C6-10aryl), —NHC(O)(heteroaryl), —NHC(O)O(C1-9alkyl), —NHC(O)O(C1-8haloalkyl), —NHC(O)O(C2-6alkenyl), —NHC(O)O(C2-6alkynyl), —NHC(O)O(C3-15cycloalkyl), —NHC(O)O(heterocyclyl), —NHC(O)O(C6-10aryl), —NHC(O)O(heteroaryl), —NHC(O)NH(C1-9alkyl), —NHC(O)NH(C1-8haloalkyl), —NHC(O)NH(C2-6alkenyl), —NHC(O)NH(C2-6alkynyl), —NHC(O)NH(C3-15cycloalkyl), —NHC(O)NH(heterocyclyl), —NHC(O)NH(C6-10aryl), —NHC(O)NH(heteroaryl), —NHS(O)(C1-9alkyl), —N(C1-9alkyl)(S(O)(C1-9alkyl), —S(C1-9alkyl), —S(C1-8haloalkyl), —S(C2-6alkenyl), —S(C2-6alkynyl), —S(C3-15cycloalkyl), —S(heterocyclyl), —S(C6-10aryl), —S(heteroaryl), —S(O)N(C1-9alkyl)2, —S(O)(C1-9alkyl), —S(O)(C1-8haloalkyl), —S(O)(C2-6alkenyl), —S(O)(C2-6alkynyl), —S(O)(C3-15cycloalkyl), —S(O)(heterocyclyl), —S(O)(C6-10aryl), —S(O)(heteroaryl), —S(O)2(C1-9alkyl), —S(O)2(C1-8haloalkyl), —S(O)2(C2-6alkenyl), —S(O)2(C2-6alkynyl), —S(O)2(C3-15), —S(O)2(heterocyclyl), —S(O)2(C6-10aryl), —S(O)2(heteroaryl), —S(O)(NH)(C1-9alkyl), —S(O)2NH(C1-9alkyl), or —S(O)2N(C1-9alkyl)2,wherein each alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl is optionally substituted with 1 to 3 C1-9alkyl, C1-8haloalkyl, halogen, —OH, —NH2, CO2H, —O(C1-9alkyl), —O(C1-8haloalkyl), —O(C3-15cycloalkyl), —O(heterocyclyl), —O(aryl), —O(heteroaryl), —NH(C1-9alkyl), —NH(C1-8haloalkyl), —NH(C3-15cycloalkyl), —NH(heterocyclyl), —NH(aryl), —NH(heteroaryl), —N(C1-9alkyl)2, —N(C3-15cycloalkyl)2, —NHC(O)(C1-8haloalkyl), —NHC(O)(C3-15cycloalkyl), —NHC(O)(heterocyclyl), —NHC(O)(aryl), —NHC(O)(heteroaryl), —NHC(O)O(C1-9alkyl), —NHC(O)O(C1-8haloalkyl), —NHC(O)O(C2-6alkynyl), —NHC(O)O(C3-15cycloalkyl), —NHC(O)O(heterocyclyl), —NHC(O)O(aryl), —NHC(O)O(heteroaryl), —NHC(O)NH(C1-9alkyl), S(O)2(C1-9alkyl), —S(O)2(C1-8haloalkyl), —S(O)2(C3-15cycloalkyl), —S(O)2(heterocyclyl), —S(O)2(aryl), —S(O)2(heteroaryl), —S(O)(NH)(C1-9alkyl), —S(O)2NH(C1-9alkyl), or —S(O)2N(C1-9alkyl)2,wherein each alkyl or heterocyclyl is optionally substituted with one to four halogens;wherein each heterocyclyl has three to twelve ring members and has one to four heteroatoms, each independently N, O, or S; and wherein each heteroaryl has five to twelve ring members and one to four heteroatoms, each independently N, O, or S. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R1C6-10aryl or 6-membered heteroaryl, each of which is optionally substituted with one to four R4 In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R1is phenyl or 6-membered heteroaryl, each of which is optionally substituted with one to four R4. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R1is optionally substituted with one to four R4. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R1is a 6-membered heteroaryl, each of which is optionally substituted with one to four R4. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R1is optionally substituted with one to four R4. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R1is substituted with three R4. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R1is substituted with two R4. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R1can be substituted with one R4. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof R1is 6-membered heteroaryl, wherein the heteroaryl is fused to a 5- or 6-membered ring having zero to three heteroatoms, each independently N, O, or S, to form a fused ring system, wherein the fused ring system is optionally substituted with one to four R5. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which are each optionally substituted with one to three RA. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which is optionally substituted with one to three RA. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which are each optionally substituted with one to three RA. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which is optionally substituted with one to three RA. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which is optionally substituted with one to two RA. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which is optionally substituted with one RA. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which is optionally substituted with one to three RA. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which is optionally substituted with one to two RA. In some embodiments, of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring A is which is optionally substituted with one RA. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each RAis independently C1-6alkyl, C1-6haloalkyl, C1-8alkoxy, C1-6haloalkoxy, halogen, —OH, —CN, or —N(R10a)(R10b). In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B is C6-10aryl or heteroaryl, which is each optionally substituted with one to four RB. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B is phenyl or 5 to 6 membered heteroaryl, wherein the phenyl or heteroaryl is optionally substituted with one to four RB. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B is which is optionally substituted with one to four RB. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B is which is optionally substituted with one to four RB. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B can be substituted with three RB. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B can be substituted with two RB. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B can be substituted with one RB. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B is a phenyl or 5 to 6 membered heteroaryl, which is each fused to a 5 or 6 membered ring having zero to four heteroatoms, each independently N, O, or S, to form a fused ring system, wherein the fused ring system is optionally substituted with one to five RB. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, ring B is which is optionally substituted with one to five RB In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each RBis independently C1-9alkyl, C1-8haloalkyl, halogen, heteroaryl, oxo, or —N(R10a)(R10b). In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, V is CH2—, —C(O)—, —C(F)2—, —CH(F)—, or —CH(CH3)—. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof X1, X2, and X3are each independently —CH═, —C(Br)═, —C(C≡CH2CH2CH3)═, or —C(C≡CC(CH3)(CH3)(CH2(OH)))═. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, X1, X2, and X3are each independently —CH═, —C(F)═, —C(Cl)═, —C(Br)═, or —C(CN)═. In some embodiments, X1, X2, and X3are each independently —CH═ or —C(F)═. In some embodiments, two of X1, X2, and X3are —CH═ and one is —C(F)═. In some embodiments X1is —C(F)═, and X2, and X3are each —CH═. In some embodiments X2is —C(F)═, and X1, and X3are each —CH═. In some embodiments X1, and X2are each —CH═, and X3is —C(F)═. In some embodiments, X1, X2, and X3is each —CH═. In some embodiments, X1, X2, and X3are each independently —CH═ or —C(Cl)═. In some embodiments, two of X1, X2, and X3are —CH═ and one is —C(Cl)═. In some embodiments X1is —C(Cl)═, and X2, and X3are each —CH═. In some embodiments X2is —C(Cl)═, and X1, and X3are each —CH═. In some embodiments X1, and X2are each —CH═, and X3is —C(Cl)═. In some embodiments, X1, X2, and X3is each —CH═. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R2is C3-10cycloalkyl or heterocyclyl, wherein the cycloalkyl or heterocyclyl is optionally substituted with one to four Z1. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R2is each optionally substituted with one to four Z1. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R2is each optionally substituted with one to four Z1. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R2is each optionally substituted with one to four Z1. In some embodiments of the compound of Formula (I) or a pharmaceutically acceptable salt thereof, R2is or In some embodiments of the compound of Formula (I) or a pharmaceutically acceptable salt thereof, R2is In some embodiments of the compound of Formula (I) or a pharmaceutically acceptable salt thereof, R2is In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R2is In some embodiments of the compound of Formula (I) or a pharmaceutically acceptable salt thereof, R2is whereinR2ais C1-6alkyl or C1-6haloalkyl;R2bis H, or C1-3alkyl;R2, is H, or C1-3alkyl; oralternatively, R2band R2ccan bind to each other to form a ring. In some embodiments of the compound of Formula (I) or a pharmaceutically acceptable salt thereof, R2is In some embodiments of the compound of Formula (I) or a pharmaceutically acceptable salt thereof, R2is In some embodiments of the compound of Formula (I) or a pharmaceutically acceptable salt thereof, each Z1is independently C1-6alkyl, C1-6alkoxy, C1-6hydroxyalkyl, C2-6alkoxyalkyl, halogen, C1-6haloalkyl, C1-6haloalkoxy, oxo, —OH, —CN, C1-6alkyl-CN, —C(O)R10a, —C(O)O—R10a, —C(O)NH2, —C(O)NH(C1-9alkyl), —N(R10a)(R10b), —N(R10a)C(O)O—R10b, —N(R10a)C(O)—R10b, —S(O)2R10a, or —S(O)2(C1-9alkyl). In some embodiments of the compound of Formula (I) or a pharmaceutically acceptable salt thereof, each Z1is independently C1-6alkyl, C1-6alkoxy, halogen, oxo, —C(O)R10a, —C(O)O—R10a, —C(O)NH2, —C(O)NH(C1-9alkyl), —N(R10a)(R10b), —N(R10a)C(O)O— R10b, —N(R10a)C(O)—R10b, —S(O)2R10a, or —S(O)2(C1-9alkyl). In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each Z1is independently C1-6alkyl, C1-6alkoxy, halogen, or C1-6haloalkyl. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each Z1is independently methyl, ethyl, or propyl. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each Z1is independently methyl, or ethyl. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each Z1is independently methyl. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, R3is —C(O)OH. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4is independently C1-9alkyl, C1-8haloalkyl, C1-6haloalkoxy, C2-6alkoxyalkyl, halogen, C3-10cycloalkyl, heterocyclyl, C6-10aryl, heteroaryl, oxo, —NO2, —CN, —N3, —O—R10a, —C(O)R10a, —C(O)O—R10R10a, —N(R10a)(R10b), —N(R10a)2(R10b), —N(R10a)—C(O)R10b, —N(R10a)S(O)2—N(R10b)(R10c), —S(O)R10a, or —S(O)(NR10a)R10b—S(O)2R10a. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4is independently C1-9alkyl, C1-8haloalkyl, C1-6haloalkoxy, C2-6alkoxyalkyl, halogen, C3-10cycloalkyl, heteroaryl, or —C(O)R10aIn some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4is independently —F, —Cl, —CN, C1-9alkyl, C3-10cycloalkyl, heteroaryl, or —C(O)R10a. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4is independently —F, —Cl, —CN, or heteroaryl. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4is independently —F, —Cl, of —CN. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4is independently —F, —Cl, or heteroaryl. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4heteroaryl is a pyrole, imidazole, triazole or a thiazole. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4heteroaryl is an imidazole or a triazole. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R4heteroaryl is a triazole. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R5is independently C1-9alkyl, C1-8haloalkyl, C1-6alkoxy, C1-6haloalkoxy, C2-6alkoxyalkyl, C2-6alkenyl, C2-6alkynyl, halogen, C3-15cycloalkyl, heterocyclyl, heteroaryl, C6-10aryl, oxo, —N3, —CN, —O—R10a, —C(O)—R10a, —C(O)O—R10a, C(O)—N(R10a)(R10b), —N(R10a)(R10b), —N(R10a)2(R10b)+, —N(R10aR10b)C(O)—R10b, —N(R10a)C(O)O—R10b, —N(R10a)C(O)N(R10b)(R10c), or —OC(O)—N(R10a)(R10b). In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R5is independently C1-9alkyl, C1-8haloalkyl, C1-6alkoxy, C2-6alkoxyalkyl, halogen, C3-15cycloalkyl, heteroaryl, or —CN. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R6is independently C1-9alkyl, C1-6alkoxy, C2-6alkoxyalkyl, C1-8haloalkyl, halogen, C3-15cycloalkyl, heterocyclyl, C6-10aryl, heteroaryl, —O(C1-9alkyl), —O(C1-8haloalkyl), —O(C2-6alkenyl), —O(C2-6alkynyl), —O(C3-15cycloalkyl), —O(heterocyclyl), —O(C6-10aryl), —O(heteroaryl), —NH(C1-9alkyl), —NH(C1-8haloalkyl), —NH(C2-6alkenyl), —NH(C2-6alkynyl), —NH(C3-15cycloalkyl), —NH(heterocyclyl), —NH(C6-10aryl), or —NH(heteroaryl). In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R6is independently C1-9alkyl, C1-6alkoxy, C2-6alkoxyalkyl, C1-8haloalkyl, C3-15cycloalkyl or halogen. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R7is independently C1-9alkyl, C1-6alkoxy, C2-6alkoxyalkyl, C1-8haloalkyl, oxo, —OH, —CN, —NH2, or halogen. In some embodiments of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, each R7is C2-6alkoxyalkyl. In some embodiments, a compound of Formula (II) is provided: or a pharmaceutically acceptable salt thereof, whereinR1is a phenyl or 6 membered heteroaryl optionally substituted with R4;X1is —C(H)═ or —C(R8)═;X4is —C(H)═, —C(R8)═ or N;each R4is independently halogen or —CN;each RBis independently C1-9alkyl, C1-8haloalkyl, or halogen;each R8is independently H or halogen; andn is 0, 1, 2, or 3. In some embodiments, a compound of Formula (I) is a compound according to Formula (II). In some embodiments, of a compound of Formula (II), or a pharmaceutically acceptable salt thereof, R1is phenyl, substituted with halogen or —CN. In some embodiments, of a compound of Formula (II), or a pharmaceutically acceptable salt thereof, R1is substituted with halogen or —CN. In some embodiments, of a compound of Formula (II), or a pharmaceutically acceptable salt thereof, R1is In some embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein RBis methyl, ethyl, F, or Cl. In some embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein RBis methyl or F. In some embodiments, embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein RBis F. In some embodiments of a compound of Formula (II), or a pharmaceutically acceptable salt thereof, R8is F or Cl. In some embodiments, a compound of Formula (III) is provided: or a pharmaceutically acceptable salt thereof, whereinX1is —C(H)═ or —C(R8)═;X4is —C(H)═, —C(R8)═ or N;X5is —C(H)═ or N;each RBis independently C1-9alkyl, C1-8haloalkyl, or halogen;each R4is independently pyrole, imidazole, triazole or a thiazole;each R8is independently halogen;m is 0, or 1; andn is 0, 1, 2, or 3. In some embodiments, a compound of Formula (I) is a compound according to Formula (III). In some embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein RBis methyl, ethyl, F, or Cl. In some embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein RBis methyl or F. In some embodiments, embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein RBis F. In some embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein R4is imidazole or triazole. In some embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein R4is triazole. In some embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein R4is In some embodiments, a compound of Formula (III), or a pharmaceutically acceptable salt thereof, wherein R4is In some embodiments of a compound of Formula (II), or a pharmaceutically acceptable salt thereof, R8is F or Cl. In some embodiments, the compound of Formula (I), Formula (II) or Formula (III), or pharmaceutically acceptable salt thereof, has the structure of a compound in Table 2. Also disclosed herein are the in vivo metabolic products of the compounds described herein, to the extent such products are novel and unobvious over the prior art. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, included are novel and unobvious compounds produced by a process comprising contacting a compound with a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radiolabeled (e.g.14C or3H) compound, administering it parenterally in a detectable dose (e.g. greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products can be easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g. by MS or NMR analysis. In general, analysis of metabolites can be done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, can be useful in diagnostic assays for therapeutic dosing of the compounds even if they possess no GLP-1R activity of their own. Recipes and methods for determining stability of compounds in surrogate gastrointestinal secretions are known. Compounds are defined herein as stable in the gastrointestinal tract where less than about 50 mole percent of the protected groups are deprotected in surrogate intestinal or gastric juice upon incubation for 1 hour at 37° C. Simply because the compounds are stable to the gastrointestinal tract does not mean that they cannot be hydrolyzed in vivo. The prodrugs typically will be stable in the digestive system but may be substantially hydrolyzed to the parental drug in the digestive lumen, liver, lung or other metabolic organ, or within cells in general. As used herein, a prodrug is understood to be a compound that is chemically designed to efficiently liberate the parent drug after overcoming biological barriers to oral delivery. II. Methods of Preparing Compounds The compounds of the present disclosure can be prepared by any method known in the art. The following exemplary general methods illustrate routes that may be used to obtain a compound of the present disclosure. Intermediate 1.3 may be assembled by reacting an amine with Intermediate 1.1, wherein X is a halogen, and R is an alkyl, alkylaryl, or aryl, in the presence of a suitable base (e.g. DIPEA, KOtBu, etc.) to give Intermediate 1.2. Intermediate 1.2 can be converted to Intermediate 1. 3 using suitable reducing conditions (e.g. H2and Pd/C, Fe and HCl, etc.). Compounds of Formula (I) having the structure of a compound of Formula 2.9 can be assembled by first coupling Intermediate 2.1, wherein X21and X22is each a leaving group, e.g., a halogen such as Cl or Br, with a heteroatom containing Intermediate 2.2 (where Y=O, NH, or S) using either a suitable base (e.g., DIPEA, KOtBu, etc.) or through metal mediated cross-coupling using a suitable palladium catalyst to give Intermediate 2.3 (Scheme 2). Intermediate 2. 4 (where M=Li, MgBr, MgCl, or MgI, purchased commercially or obtained through metalation of a corresponding halide) can be combined with Intermediate 2.3 using a suitable palladium catalyst to deliver Intermediate 2.5. Following conversion to the acid Intermediate 2.6 using standard conditions (e.g. LiOH, LiI and pyridine, etc.), the Intermediate 1.3 can be added using standard amide bond forming conditions (e.g. DIPEA with HATU, etc.) to give Intermediate 2.7, which can, in turn, be converted to the corresponding benzimidazole Intermediate 2.8 under the influence of an acid catalyst (e.g. HCl, AcOH, etc.) or dehydrating agents (e.g., POCl3, Tf2O/triphenylphosphine oxide, etc.). This intermediate can be converted to the compound of Formula (I) using standard ester hydrolysis conditions (e.g., LiOH, LiI and pyridine, etc.). While the above Scheme 2 is illustrated using Intermediate 2.1 as a dihalopyridine, any dihalogenated A-ring starting material can be used to obtain the analogous compound of Formula (I). In some embodiments, a compound of Formula (I) having the structure of a compound of Formula 2.9 can be assembled first by the combination of Intermediate 3.1 (wherein X31 is Cl, Br, or I) with Intermediate 1.3 (wherein R=alkyl, alkylaryl, or aryl) under standard amide bond forming conditions, e.g. DIPEA with HATU, etc. (Scheme 3). Treatment with a suitable acid catalyst (e.g. HCl, AcOH, etc.) or dehydrating agents (e.g., POCl3, Tf2O/triphenylphosphine oxide, etc.) can deliver Intermediate 3.3. Halogen metal exchange of —X31 to -M can be achieved using a suitable reagent (e.g. iPrMgBr, etc.) or transition metal coupling using a suitable palladium catalyst and metal source (e.g. B2Pin2, Bu6Sn2, etc.) to give Intermediate 2.8 which can be converted to the compound of Formula (I) using standard ester hydrolysis conditions (e.g. LiOH, LiI and pyridine, etc.). In some embodiments, a compound of Formula 2.9 can be formed by first conversion of Intermediate 2.3 to the metallated variant Intermediate 4.1 using a suitable palladium catalyst and metal source, e.g. B2Pin2, Bu6Sn2, etc. (Scheme 4). Intermediate 4.1 can be coupled to Intermediate 3.3 using a suitable palladium catalyst to deliver Intermediate 2.8 which can then be converted to the compound of Formula (I) using standard ester hydrolysis conditions, e.g. LiOH, LiI and pyridine, etc. A compound of Formula (I-A-1) and/or Formula (I) having the structure of a compound of Formula 5.3 can be assembled via first coupling to the halogen —X (wherein X is Cl, Br, or I) of Intermediate 5.1 using a suitable coupling partner and palladium catalyst to deliver Intermediate 5.2 which can be converted to a compound of Formula 5.3 using standard ester hydrolysis conditions, e.g. LiOH, LiI and pyridine, etc. (Scheme 5). A compound of Formula (I) having the structure of a compound of Formula 6.1 can be obtained through the reaction of Intermediate 2.9 with a sulfonamide under suitable coupling conditions (e.g. EDCI and DMAP, etc.) (Scheme 6). A compound of Formula (I) having the structure of a compound of Formula 7.3 can be assembled via first coupling to the halogen —X of Intermediate 7.1 using a suitable coupling partner and palladium catalyst to deliver Intermediate 7.2, which can be converted to a compound of Formula 7.3 using standard ester hydrolysis conditions (e.g. LiOH, LiI and pyridine, etc.) (Scheme 7). A compound of Formula (I) having the structure of a compound of Formula 2.9 can be assembled through first cross-coupling of an Intermediate 3.4 with Intermediate 2.1 using a suitable transition metal catalyst (e.g. palladium, etc.) (Scheme 8). This can then be coupled with a heteroatom containing Intermediate 2.2 (where Y=O, N or S) using either a suitable base (e.g. DIPEA, KOtBu, etc.) or through metal mediated cross-coupling using a suitable palladium catalyst to give Intermediate 2.8. Intermediate 2.8 can be converted to the compound of Formula (I) having the structure of a compound of Formula 2.9 using standard ester hydrolysis conditions (e.g. LiOH, LiI and pyridine, etc.). A compound of Formula (I) having the structure of a compound of Formula 2.9 can be assembled through first cross-coupling of an Intermediate 3.4 with Intermediate 9.1 using a suitable transition metal catalyst (e.g. palladium, etc.) (Scheme 9). The benzyl ether can then be removed through reduction using H2and a suitable catalyst (Pd/C, etc.) to yield intermediate 9.2. Intermediate 9.2 can then be alkylated using a suitable base (K2CO3, Cs2CO3, Ag2CO3, etc.) a suitable electrophile represented by intermediate 9.3 where X91can be —Cl, —Br, I, or —OTs. Intermediate 2.8 can be converted to the compound of Formula (I) having the structure of a compound of Formula 2.9 using standard ester hydrolysis conditions (e.g. LiOH, LiI and pyridine, etc.). A compound of Formula (I) having the structure of a compound of Formula 10.4 can be assembled through first alkylation if intermediate 9.2 with an intermediate of the type 10.1 using a suitable base (K2CO3, Cs2CO3, Ag2CO3, etc.) where X101and X102are each independently —Cl, —Br, —I, —OTs, or —OTf and Y1, Y2, Y3, and Y4are each independently —CH═ or —N═. Intermediate 10.2 is then converted to intermediate 10.3 using a suitable transition metal catalyst (e.g. palladium, etc.). Intermediate 10.3 can be converted to the compound of Formula (I) having the structure of a compound of Formula 10.4 using standard ester hydrolysis conditions (e.g. LiOH, LiI and pyridine, etc.). IV. Pharmaceutical Formulations In some embodiments, the present disclosure provides a pharmaceutical composition comprising a compound of the present disclosure (e.g. a compound of Formula (I)), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient. In some embodiments of the disclosure, the pharmaceutical composition comprises a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and one or more additional therapeutic agents, as more fully set forth below. Pharmaceutical compositions comprising the compounds disclosed herein, or pharmaceutically acceptable salts thereof, may be prepared with one or more pharmaceutically acceptable excipients which may be selected in accord with ordinary practice. Tablets may contain excipients including glidants, fillers, binders and the like. Aqueous compositions may be prepared in sterile form, and when intended for delivery by other than oral administration generally may be isotonic. In some embodiments, compositions may contain excipients such as those set forth in the Rowe et al, Handbook of Pharmaceutical Excipients, 6th edition, American Pharmacists Association, 2009. Excipients can include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkyl cellulose, hydroxyalkylmethyl cellulose, stearic acid and the like. In some embodiments, the composition is provided as a solid dosage form, including a solid oral dosage form. The compositions include those suitable for various administration routes, including oral administration. The compositions may be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient (e.g., a compound of the present disclosure or a pharmaceutical salt thereof) with one or more pharmaceutically acceptable excipients. The compositions may be prepared by uniformly and intimately bringing into association the active ingredient with liquid excipients or finely divided solid excipients or both, and then, if desired, shaping the product. Techniques and formulations generally are found in Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams and Wilkins, Philadelphia, Pa., 2006. Compositions described herein that are suitable for oral administration may be presented as discrete units (a unit dosage form) including but not limited to capsules, sachets or tablets each containing a predetermined amount of the active ingredient. In one embodiment, the pharmaceutical composition of the disclosure is a tablet. Pharmaceutical compositions disclosed herein comprise one or more compounds disclosed herein, or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable excipient and optionally other therapeutic agents. Pharmaceutical compositions containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more excipients including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed. The amount of active ingredient that may be combined with the inactive ingredients to produce a dosage form may vary depending upon the intended treatment subject and the mode of administration. For example, in some embodiments, a dosage form for oral administration to humans may contain approximately 1 to 1000 mg of active material formulated with an appropriate and convenient amount of a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutically acceptable excipient varies from about 5 to about 95% of the total compositions (weight:weight). In some embodiments, a composition comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof in one variation does not contain an agent that affects the rate at which the active ingredient is metabolized. Thus, it is understood that compositions comprising a compound of the present disclosure in one aspect do not comprise an agent that would affect (e.g., slow, hinder or retard) the metabolism of a compound of the present disclosure or any other active ingredient administered separately, sequentially or simultaneously with a compound of the present disclosure. It is also understood that any of the methods, kits, articles of manufacture and the like detailed herein in one aspect do not comprise an agent that would affect (e.g., slow, hinder or retard) the metabolism of a compound of the present disclosure or any other active ingredient administered separately, sequentially or simultaneously with a compound of the present disclosure. In some embodiments, the pharmaceutical compositions described above are for use in a human or an animal. The disclosure further includes a compound of the present disclosure for administration as a single active ingredient of a pharmaceutically acceptable composition which can be prepared by conventional methods known in the art, for example by binding the active ingredient to a pharmaceutically acceptable, therapeutically inert organic and/or inorganic carrier or excipient, or by mixing therewith. In one aspect, provided herein is the use of a compound of the present disclosure as a second or other active ingredient having a synergistic effect with other active ingredients in known drugs, or administration of the compound of the present disclosure together with such drugs. The compound of the present disclosure may also be used in the form of a prodrug or other suitably modified form which releases the active ingredient in vivo. V. Routes of Administration The compounds of the present disclosure (also referred to herein as the active ingredients), can be administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), transdermal, vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intratumoral, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient. An advantage of certain compounds disclosed herein is that they are orally bioavailable and can be dosed orally. A compound of the present disclosure may be administered to an individual in accordance with an effective dosing regimen for a desired period of time or duration, such as at least about one month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer. In one variation, the compound is administered on a daily or intermittent schedule for the duration of the individual's life. The dosage or dosing frequency of a compound of the present disclosure may be adjusted over the course of the treatment, based on the judgment of the administering physician. The compound may be administered to an individual (e.g., a human) in an effective amount. In some embodiments, the compound is administered once daily. The compound can be administered by any useful route and means, such as by oral or parenteral (e.g., intravenous) administration. Therapeutically effective amounts of the compound may include from about 0.00001 mg/kg body weight per day to about 10 mg/kg body weight per day, such as from about 0.0001 mg/kg body weight per day to about 10 mg/kg body weight per day, or such as from about 0.001 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.01 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.05 mg/kg body weight per day to about 0.5 mg/kg body weight per day, or such as from about 0.3 mg to about 30 mg per day, or such as from about 30 mg to about 300 mg per day. A compound of the present disclosure may be combined with one or more additional therapeutic agents in any dosage amount of the compound of the present disclosure (e.g., from 1 mg to 1000 mg of compound). Therapeutically effective amounts may include from about 1 mg per dose to about 1000 mg per dose, such as from about 50 mg per dose to about 500 mg per dose, or such as from about 100 mg per dose to about 400 mg per dose, or such as from about 150 mg per dose to about 350 mg per dose, or such as from about 200 mg per dose to about 300 mg per dose. Other therapeutically effective amounts of the compound of the present disclosure are about 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, or about 500 mg per dose. Other therapeutically effective amounts of the compound of the present disclosure are about 100 mg per dose, or about 125, 150, 175, 200, 225, 250, 275, 300, 350, 400, 450, or about 500 mg per dose. A single dose can be administered hourly, daily, or weekly. For example, a single dose can be administered once every 1 hour, 2, 3, 4, 6, 8, 12, 16 or once every 24 hours. A single dose can also be administered once every 1 day, 2, 3, 4, 5, 6, or once every 7 days. A single dose can also be administered once every 1 week, 2, 3, or once every 4 weeks. In some embodiments, a single dose can be administered once every week. A single dose can also be administered once every month. Kits that comprise a compound of the present disclosure, or an enantiomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing any of the above, are also included in the present disclosure. In one embodiment, a kit further includes instructions for use. In one aspect, a kit includes a compound of the disclosure, or a pharmaceutically acceptable salt, tautomer, stereoisomer, mixture of stereoisomers, prodrug, or deuterated analog thereof, and a label and/or instructions for use of the compounds in the treatment of the indications, such as the diseases or conditions, described herein. In one embodiment, kits comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, in combination with one or more (e.g., one, two, three, four, one or two, or one to three, or one to four) additional therapeutic agents are provided. Provided herein are also articles of manufacture that include a compound of the present disclosure or a pharmaceutically acceptable salt, tautomer, stereoisomer, mixture of stereoisomers, prodrug, or deuterated analog thereof in a suitable container. The container may be a vial, jar, ampoule, preloaded syringe, and intravenous bag. VI. Combination Therapy In some embodiments, a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, can be combined with a therapeutically effective amount of one or more (e.g., one, two, three, four, one or two, one to three, or one to four) additional therapeutic agents. In some embodiments, the additional therapeutic agent comprises an apoptotic signal-regulating kinase (ASK-1) inhibitor, a farnesoid X receptor (FXR) agonist, a peroxisome proliferator-activated receptor alpha (PPARα) agonist, fish oil, an acetyl-coA carboxylase (ACC) inhibitor, a TGFβ antagonist, a LPAR antagonist, a SGLT2 inhibitor, a Tpl2 inhibitor, or a GLP-1 agonist combination thereof. The benefit of combination may be increased efficacy and/or reduced side effects for a component as the dose of that component may be adjusted down to reduce its side effects while benefiting from its efficacy augmented by the efficacy of the compound of the present disclosure. In some embodiments, the therapeutic agent, or combination of therapeutic agents, are a(n) ACE inhibitor, 2-Acylglycerol O-acyltransferase 2 (DGAT2) inhibitor, Acetaldehyde dehydrogenase inhibitor, Acetyl CoA carboxylase inhibitor, Adrenergic receptor agonist, Alstrom syndrome protein 1 (ALMS1)/PKC alpha protein interaction inhibitor, Apelin receptor agonist, Diacylglycerol O acyltransferase 2 inhibitor, Adenosine A3 receptor agonist, Adenosine A3 receptor antagonist, Adiponectin receptor agonist, Aldehyde dehydrogenase 2 stimulator, AKT protein kinase inhibitor, AMP-activated protein kinases (AMPK), AMP kinase activator, ATP citrate lyase inhibitor, AMP activated protein kinase stimulator, Endothelial nitric oxide synthase stimulator, NAD-dependent deacetylase sirtuin-1 stimulator, Adrenergic receptor antagonist, Androgen receptor agonist, Amylin receptor agonist, Angiotensin II AT-1 receptor antagonist, Apical sodium-dependent bile acid transport inhibitor, Autophagy protein modulator, Autotaxin inhibitors, Axl tyrosine kinase receptor inhibitor, Bax protein stimulator, Beta-catenin inhibitor, Bioactive lipid, Calcitonin agonist, Cannabinoid receptor modulator, Caspase inhibitor, Caspase-3 stimulator, Cathepsin inhibitor, Caveolin 1 inhibitor, CCK receptor antagonist, CCL26 gene inhibitor, CCR2 chemokine antagonist, CCR2 chemokine antagonist, Angiotensin II AT-1 receptor antagonist, CCR3 chemokine antagonist, CCR5 chemokine antagonist, CD3 antagonist, CDGSH iron sulfur domain protein modulator, chitinase inhibitor, Chloride channel stimulator, Chitotriosidase 1 inhibitor, CNR1 inhibitor, Connective tissue growth factor ligand inhibitor, COT protein kinase inhibitor, Cyclin D1 inhibitor, Cytochrome P450 7A1 inhibitor, Cytochrome P450 reductase inhibitors, DGAT1/2 inhibitor, Diacylglycerol O acyltransferase 1 inhibitor (DGAT1), Cytochrome P450 2E1 inhibitor (CYP2E1), CXCR3 chemokine antagonist, CXCR4 chemokine antagonist, Dihydroceramide delta 4 desaturase inhibitor, Dihydroorotate dehydrogenase inhibitor, Dipeptidyl peptidase IV inhibitor, Endosialin modulator, Eotaxin ligand inhibitor, Extracellular matrix protein modulator, Farnesoid X receptor agonist, Fatty acid synthase inhibitors, FGF1 receptor agonist, Fibroblast growth factor (FGF-15, FGF-19, FGF-21) ligands, fibroblast activation protein inhibitor, Free fatty acid receptor 1 agonist, Galectin-3 inhibitor, GDNF family receptor alpha like agonist, Glucagon receptor agonist, Glucagon-like peptide 1 agonist, Glucocorticoid receptor antagonist, Glucose 6-phosphate 1-dehydrogenase inhibitor, G-protein coupled bile acid receptor 1 agonist, G-protein coupled receptor-119 agonist, G-protein coupled receptor 84 antagonist, Hedgehog (Hh) modulator, Hepatitis C virus NS3 protease inhibitor, Hepatocyte nuclear factor 4 alpha modulator (HNF4A), Hepatocyte growth factor modulator, Histone deacetylase inhibitor, STAT-3 modulator, HMG CoA reductase inhibitor, HSD17B13 gene inhibitor, 5-HT 2a receptor antagonist, Hydrolase inhibitor, Hypoxia inducible factor-2 alpha inhibitor, IL-10 agonist, IL-17 antagonist, IL-22 agonist, Ileal sodium bile acid cotransporter inhibitor, Insulin sensitizer, Insulin ligand agonist, Insulin receptor agonist, integrin modulator, Integrin Antagonist, Integrin alpha-V/beta-1 antagonist, Integrin alpha-V/beta-6 antagonist, intereukin-1 receptor-associated kinase 4 (IRAK4) inhibitor, IL-6 receptor agonist, interleukin 17 ligand inhibitor, Jak2 tyrosine kinase inhibitor, Jun N terminal kinase-1 inhibitor, Kelch like ECH associated protein 1 modulator, Ketohexokinase (KHK) inhibitor, Klotho beta stimulator, Leukotriene A4 hydrolase inhibitor, 5-Lipoxygenase inhibitor, Lipoprotein lipase inhibitor, Liver X receptor, LPL gene stimulator, Lysophosphatidate-1 receptor antagonist, Lysyl oxidase homolog 2 inhibitor, LXR inverse agonists, Macrophage mannose receptor 1 modulator, Matrix metalloproteinases (MMPs) inhibitor, MEKK-5 protein kinase inhibitor, MCH receptor-1 antagonist, Membrane copper amine oxidase (VAP-1) inhibitor, Methionine aminopeptidase-2 inhibitor, Methyl CpG binding protein 2 modulator, MicroRNA-132 (miR-132) antagonist, MicroRNA-21 (miR-21) inhibitor, Mitochondrial uncoupler, Mixed lineage kinase-3 inhibitor, Motile sperm domain protein 2 inhibitor, Myelin basic protein stimulator, NACHT LRR PYD domain protein 3 (NLRP3) inhibitor, NAD-dependent deacetylase sirtuin stimulator, NADPH oxidase inhibitor (NOX), NFE2L2 gene inhibitor, Nicotinic acid receptor 1 agonist, Opioid receptor mu antagonist, P2Y13 purinoceptor stimulator, Nuclear erythroid 2-related factor 2 stimulator, Nuclear receptor modulators, Nuclear transport of transcription factor modulator, P2X7 purinoceptor modulator, PACAP type I receptor agonist, PDE 3 inhibitor, PDE 4 inhibitor, PDE 5 inhibitor, PDGF receptor beta modulator, Phenylalanine hydroxylase stimulator, Phospholipase C inhibitor, Phosphoric diester hydrolase inhibitor, PPAR alpha agonist, PPAR delta agonist, PPAR gamma agonist, Peptidyl-prolyl cis-trans isomerase A inhibitor, PNPLA3 gene inhibitor, PPAR gamma modulator, Protease-activated receptor-2 antagonist, Protein kinase modulator, Protein NOV homolog modulator, PTGS2 gene inhibitor, renin inhibitor, Resistin/CAP1 (adenylyl cyclase associated protein 1) interaction inhibitor, Rho associated protein kinase inhibitor, RNA polymerase inhibitors, S-nitrosoglutathione reductase (GSNOR) enzyme inhibitor, Sodium glucose transporter-2 inhibitor, Sphingolipid delta 4 desaturase DES1 inhibitor, SREBP transcription factor inhibitor, STAT-1 inhibitor, Stearoyl CoA desaturase-1 inhibitor, STK25 inhibitor, Suppressor of cytokine signalling-1 stimulator, Suppressor of cytokine signalling-3 stimulator, Taste receptor type 2 agonist, Telomerase stimulator, TERT gene modulator, TGF beta (TGFB1) ligand inhibitor, TNF antagonist, Transforming growth factor β (TGF-β), Transforming growth factor β activated Kinase 1 (TAK1), Thyroid hormone receptor beta agonist, TLR-4 antagonist, Transglutaminase inhibitor, Tyrosine kinase receptor modulator, GPCR modulator, nuclear hormone receptor modulator, TLR-9 antagonist, VDR agonist, Vitamin D3 receptor modulators, WNT modulators, YAP/TAZ modulator or a Zonulin inhibitor, and combinations thereof. Non-limiting examples of the one or more additional therapeutic agents include:ACE inhibitors, such as enalapril;Acetaldehyde dehydrogenase inhibitors, such as ADX-629;Acetyl CoA carboxylase (ACC) inhibitors, such as NDI-010976 (firsocostat), DRM-01, gemcabene, GS-834356, PF-05175157, QLT-091382, PF-05221304;Acetyl CoA carboxylase/Diacylglycerol O acyltransferase 2 inhibitors, such as PF-07055341;Adenosine receptor agonists, such as CF-102 (namodenoson), CF-101 (piclidenoson), CF-502, CGS21680;Adenosine A3 receptor antagonist, such as FM-101;Adiponectin receptor agonists, such as ADP-355, ADP-399, ALY668-SR;Adrenergic receptor antagonist, such as bromocriptine, phentermine, VI-0521;Aldehyde dehydrogenase 2 stimulators, such as FP-045;Amylin/calcitonin receptor agonists, such as KBP-042, KBP-089;AMP activated protein kinase stimulators, such as C-455, PXL-770, O-304;AMP kinase activators/ATP citrate lyase inhibitors, such as bempedoic acid (ETC-1002, ESP-55016);AMP activated protein kinase/Endothelial nitric oxide synthase/NAD-dependent deacetylase sirtuin-1 stimulators, such as NS-0200 (leucine+metformin+sildenafil);Androgen receptor agonists, such as LPCN-1144, LPCN-1148, testosterone prodrug;Angiotensin II AT-1 receptor antagonists, such as irbesartan; Angiopoietin-related protein-3 inhibitors, such as vupanorsen (IONIS-ANGPTL3-LRx);Apelin receptor agonist, such as CB-5064, MBT-2;Apical sodium-dependent bile acid transport inhibitors, such as A-3907;Autophagy protein modulators, such as A-2906, GM-90194;Autotaxin (ectonucleotide pyrophosphatase/phosphodiesterase 2 (NPP2 or ENPP2)) inhibitors, such as FP10.47, PAT-505, PAT-048, GLPG-1690, X-165, PF-8380, TJC-0265, TJC-0316, AM-063, BBT-877;Axl tyrosine kinase receptor inhibitors, such as bemcentinib (BGB-324, R-428);Bax protein stimulators, such as CBL-514;Bioactive lipids, such as DS-102;Cannabinoid receptor modulators, such as namacizumab (nimacimab), GWP-42004, REV-200, CRB-4001, INV-101, SCN-002;Caspase inhibitors, such as emricasan;Pan cathepsin B inhibitors, such as VBY-376;Pan cathepsin inhibitors, such as VBY-825;CCK receptor antagonist, such as proglumide;CCL26 gene inhibitor, such as mosedipimod, KDDF-201410-10;CCR2/CCR5 chemokine antagonists, such as BMS-687681, cenicriviroc, maraviroc, CCX-872, leronlimab, WXSH-0213;CCR2/CCR5 chemokine antagonists and FXR agonists, such as LJC-242 (tropifexor+cenivriviroc);CCR2 chemokine antagonists, such as propagermanium;CCR2 chemokine/Angiotensin II AT-1 receptor antagonists, such as DMX-200, DMX-250;CCR3 chemokine antagonists, such as bertilimumab;CD3 antagonists, such as NI-0401 (foralumab);CDGSH iron sulfur domain protein modulators, such as EYP-002;Chitinase inhibitor, such as OATD-01;Chitotriosidase 1 inhibitors, such as OAT-2068;Chloride channel stimulators, such as cobiprostone, and lubiprostone;Casein kinase-1 (CK1) delta/epsilon inhibitors, such as PF-05006739;Connective tissue growth factor ligand inhibitor, such as PBI-4050;COT protein kinase inhibitors, such as GS-4875, GS-5290;CXCR4 chemokine antagonists, such as AD-214;Cytochrome P450 reductase inhibitors, such as SNP-630;Diglyceride acyltransferase 2 (DGAT2) inhibitors, such as IONIS-DGAT2Rx, PF-06865571;Diglyceride acyltransferase 1 (DGAT1) inhibitors, such as GSK-3008356;Diacylglycerol O acyltransferase 1 (DGAT1)/Cytochrome P450 2E1 inhibitors (CYP2E1), such as SNP-610;Dihydroorotate dehydrogenase inhibitor, such as vidofludimus;Dipeptidyl peptidase IV inhibitors, such as linagliptin, evogliptin;Eotaxin ligand inhibitors, such as bertilimumab, CM-101;Extracellular matrix protein modulators, such as CNX-024;Farnesoid X receptor (FXR) agonists, such as AGN-242266, AGN-242256, ASC-42, EDP-297 (EP-024297), RDX-023, BWL-200, AKN-083, EDP-305, GNF-5120, cilofexor tromethamine (GS-9674), HPG-1860, IOT-022, LMB-763, obeticholic acid, Px-102, Px-103, M790, M780, M450, M-480, MET-409, MET-642, PX20606, SYHA-1805, vonafexor (EYP-001), TERN-101, TC-100, INT-2228, TQA-3526, ZG-5266, HPD-001, alendronate;Farnesoid X receptor (FXR)/G-protein coupled bile acid receptor 1 (TGR5) agonists, such as INT-767;Fatty acid synthase inhibitors, such as TVB-2640, FT-8225;Fibroblast growth factor 19 (rhFGF19)/cytochrome P450 (CYP) 7A1 inhibitors, such as aldafermin (NGM-282);Fibroblast growth factor 21 (FGF-21) ligand modulators, such as AP-025, BMS-986171, B-1654, BIO89-100, BOS-580, Pegbelfermin (BMS-986036), B-1344, NN-9499;Fibroblast growth factor 21 (FGF-21)/glucagon like peptide 1 (GLP-1) agonists, such as YH-25723 (YH-25724; YH-22241), efruxifermin (AKR-001);FGF receptor agonists/Klotho beta stimulators, such as BFKB-8488A (RG-7992);Free fatty acid receptor 1 agonist, such as SCO-267;Galectin-3 inhibitors, such as belapectin (GR-MD-02), GB-1107 (Gal-300), GB-1211 (Gal-400), IMT-001;GDNF family receptor alpha like agonist, such as NGM-395;Glucagon-like peptide 1 (GLP1R) agonists, such as ALT-801, AC-3174, liraglutide, cotadutide (MEDI-0382), SAR-425899, LY-3305677, HM-15211, YH-25723, YH-GLP1, RPC-8844, PB-718, PF-06882961, semaglutide;Glucagon-like peptide 1 receptor agonist; Oxyntomodulin ligand; Glucagon receptor agonist, such as efinopegdutide;Gastric inhibitory polypeptide/Glucagon-like peptide-1 (GIP/GLP-1) receptor co-agonist, such as tirzepatide (LY-3298176);PEGylated long-acting glucagon-like peptide-1/glucagon (GLP-1R/GCGR) receptor dual agonist, such as DD-01;Glucagon/GLP1-receptor agonist, such as BI-456906, NN-6177;Glucocorticoid receptor antagonists, such as CORT-118335 (miricorilant);Glucose 6-phosphate 1-dehydrogenase inhibitors, such as ST001;Glucokinase stimulator, such as dorzagliatin, sinogliatin (RO-5305552);G-protein coupled bile acid receptor 1 (TGR5) agonists, such as RDX-009, INT-777, HY-209;G-protein coupled receptor 84 antagonist, such as PBI-4547;G-protein coupled receptor-119 agonist, such as DA-1241;Heat shock protein 47 (HSP47) inhibitors, such as ND-L02-s0201;Hedgehog protein TGF beta ligand inhibitors, such as Oxy-210;Histone deacetylase inhibitors/STAT-3 modulators, such as SFX-01;HMG CoA reductase inhibitors, such as atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin;HSD17B13 gene inhibitor, such as ALN-HSD, ARO-HSD;Hydrolase inhibitor, such as ABD-X;Hypoxia inducible factor-2 alpha inhibitors, such as PT-2567;IL-10 agonists, such as peg-ilodecakin;Ileal sodium bile acid cotransporter inhibitors, such as odevixibat (A-4250), volixibat potassium ethanolate hydrate (SHP-262), GSK2330672, CJ-14199, elobixibat (A-3309);Insulin sensitizers, such as, KBP-042, azemiglitazone potassium (MSDC-0602K), ION-224, MSDC-5514, Px-102, RG-125 (AZD4076), Tolimidone, VVP-100X, CB-4211, ETI-101;Insulin ligand/dsInsulin receptor agonists, such as ORMD-0801;Integrin antagonists, such as IDL-2965;IL-6 receptor agonists, such as KM-2702;Integrin alpha-V/beta-6 and alpha-V/beta-1 dual inhibitor; such as PLN-74809;Interleukin 17 ligand inhibitor, such as netakimab;Jak1/2 tyrosine kinase inhibitor, such as baricitinib;Jun N terminal kinase-1 inhibitor, such as CC-90001;Kelch like ECH associated protein 1 modulator, such as alpha-cyclodextrin-stabilized sulforaphane;Ketohexokinase (KHK) inhibitors, such as PF-06835919, LY-3478045, LY-3522348;beta Klotho (KLB)-FGF1c agonists, such as MK-3655 (NGM-313);Leukotriene A4 hydrolase inhibitor, such as LYS-006;5-Lipoxygenase inhibitors, such as tipelukast (MN-001), epeleuton (DS-102, AF-102);Lipoprotein lipase inhibitors, such as CAT-2003;LPL gene stimulators, such as alipogene tiparvovec;Liver X receptor (LXR) inhibitors, such as PX-665, PX-L603, PX-L493, BMS-852927, T-0901317, GW-3965, SR-9238;Lysophosphatidate-1 receptor antagonists, such as BMT-053011, UD-009 (CP-2090), AR-479, ITMN-10534, BMS-986020, KI-16198;Lysyl oxidase homolog 2 inhibitors, such as simtuzumab, PXS-5382A (PXS-5338);Macrophage mannose receptor 1 modulators, such as tilmanocept-Cy3 (technetium Tc 99m tilmanocept);Matrix metalloprotease inhibitors, such as ALS-L1023;Membrane copper amine oxidase (VAP-1) inhibitors, such as TERN-201, TT-01025;MEKK-5 protein kinase (ASK-1) inhibitors, such as CJ-16871, CS-17919, selonsertib (GS-4997), SRT-015, GS-444217, GST-HG-151, TERN-301;MCH receptor-1 antagonists, such as CSTI-100 (ALB-127158);Semicarbazide-Sensitive Amine Oxidase/Vascular Adhesion Protein-1 (SSAO/VAP-1) Inhibitors, such as PXS-4728A (BI-1467335);Methionine aminopeptidase-2 inhibitors, such as ZGN-1061, ZGN-839, ZN-1345;Methyl CpG binding protein 2 modulators, such as mercaptamine;Mineralocorticoid receptor antagonists (MCRA), such as MT-3995 (apararenone);Mitochondrial uncouplers, such as 2,4-dinitrophenol, HU6, Mito-99-0053;Mixed lineage kinase-3 inhibitors, such as URMC-099-C;Motile sperm domain protein 2 inhibitors, such as VB-601;Myelin basic protein stimulators, such as olesoxime;Myeloperoxidase inhibitors, such as PF-06667272, AZM-198;NADPH oxidase inhibitors, such as GKT-831, GenKyoTex, APX-311, setanaxib;Nicotinic acid receptor 1 agonists, such as ARI-3037MO;NACHT LRR PYD domain protein 3 (NLRP3) inhibitors, such as KDDF-201406-03, NBC-6, IFM-514, JT-194 (JT-349);NFE2L2 gene inhibitor, such as GeRP-amiR-144;Nuclear transport of transcription factor modulators, such as AMTX-100;Nuclear receptor modulators, such as DUR-928 (DV-928);Opioid receptor mu antagonists, such as methylnaltrexone;P2X7 purinoceptor modulators, such as SGM-1019;P2Y13 purinoceptor stimulators, such as CER-209;PDE 3/4 inhibitors, such as tipelukast (MN-001);PDE 5 inhibitors, such as sildenafil, MSTM-102;PDGF receptor beta modulators, such as BOT-191, BOT-509;Peptidyl-prolyl cis-trans isomerase inhibitors, such as CRV-431 (CPI-432-32), NVP-018, NV-556 (NVP-025);Phenylalanine hydroxylase stimulators, such as HepaStem;Phosphoric diester hydrolase inhibitor, such as ZSP-1601;PNPLA3 gene inhibitor, such as AZD-2693;PPAR agonists, such as Chiglitazar, elafibranor (GFT-505), seladelpar lysine (MBX-8025), deuterated pioglitazone R-enantiomer, pioglitazone, PXL-065 (DRX-065), saroglitazar, lanifibranor (IVA-337), CHS-131, pemafibrate (K-877), ZG-0588, ZSP-0678; ZSYM-008;Protease-activated receptor-2 antagonists, such as PZ-235;Protein kinase modulators, such as CNX-014;Protein NOV homolog modulators, such as BLR-200;PTGS2 gene inhibitors, such as STP-705, STP-707;Renin inhibitors, such as PRO-20;Resistin/CAP1 (adenylyl cyclase associated protein 1) interaction inhibitors, such as DWJ-211;Rev protein modulator, such as ABX-464;Rho associated protein kinase (ROCK) inhibitors, such as REDX-10178 (REDX-10325), KD-025, RXC-007, TDI-01;RNA polymerase inhibitors, such as rifaximin;Snitrosoglutathione reductase (GSNOR) enzyme inhibitors, such as SL-891;Sodium glucose transporter-2 (SGLT2) inhibitors, such as ipragliflozin, remogliflozin etabonate, ertugliflozin, dapagliflozin, tofogliflozin, sotagliflozin;Sodium glucose transporter-1/2 (SGLT 1/2) inhibitors, such as licogliflozin bis(prolinate) (LIK-066);SREBP transcription factor inhibitors, such as CAT-2003, HPN-01, MDV-4463;Stearoyl CoA desaturase-1 inhibitors, such as aramchol;Taste receptor type 2 agonists, such as ARD-101;Thyroid hormone receptor beta agonists, such as ALG-009, ASC-41, CNPT-101101; CNPT-101207, CS-27186, KY-41111, resmetirom (MGL-3196), MGL-3745, TERN-501, VK-2809, HP-515;TLR-2/TLR-4 antagonists, such as VB-201 (CI-201);TLR-4 antagonists, such as JKB-121, JKB-122, naltrexone;Tyrosine kinase receptor modulators, such as CNX-025, GFE-2137 (repurposed nitazoxanide);TLR-9 antagonist, such as GNKS-356, AVO-101;TNF antagonist, such as ALF-421;GPCR modulators, such as CNX-023;Nuclear hormone receptor modulators, such as Px-102;VDR agonist, such as CK-15;Xanthine oxidase inhibitors, such as ACQT-1127;Xanthine oxidase/Urate anion exchanger 1 (URAT1) inhibitors, such as RLBN-1001, RLBN-1127; orZonulin Inhibitors, such as larazotide acetate (INN-202). In certain specific embodiments, the one or more additional therapeutic agents are selected from A-4250, AC-3174, acetylsalicylic acid, AK-20, alipogene tiparvovec, AMX-342, AN-3015, anti-CXCR3 antibodies, anti-TAGE antibody, aramchol, ARI-3037MO, ASP-8232, AXA-1125, bertilimumab, Betaine anhydrous, BI-1467335, BMS-986036, BMS-986171, BMT-053011, BOT-191, BTT-1023, budesonide, BX-003, CAT-2003, cenicriviroc, CBW-511, CER-209, CF-102, CGS21680, CNX-014, CNX-023, CNX-024, CNX-025, cobiprostone, colesevelam, dabigatran etexilate mesylate, dapagliflozin, DCR-LIV1, deuterated pioglitazone R-enantiomer, 2,4-dinitrophenol, DRX-065, DS-102, DUR-928, edaravone (TTYP-01), EDP-305, elafibranor (GFT-505), emricasan, enalapril, ertugliflozin, evogliptin, F-351, fluasterone (ST-002), FT-4101, GDD-3898, GH-509, GKT-831, GNF-5120, GRI-0621, GR-MD-02, GS-300, GS-4997, GS-9674, GS-4875, GS-5290, HEC-96719, HTD-1801, HS-10356, HSG-4112, HST-202, HST-201, HU-6, hydrochlorothiazide, icosabutate (PRC-4016), icosapent ethyl ester, IMM-124-E, INT-767, INV-240, ION-455, IONIS-DGAT2Rx, ipragliflozin, Irbesarta, propagermanium, IVA-337, J2H-1702, JKB-121, KB-GE-001, KBLP-004, KBLP-009, KBP-042, KD-025, M790, M780, M450, metformin, sildenafil, LB-700, LC-280126, linagliptin, liraglutide, (LJN-452) tropifexor, LM-011, LM-002 (CVI-LM-002), LMB-763, LYN-100, MB-N-008, MBX-8025, MDV-4463, mercaptamine, MGL-3196, MGL-3745, MP-301, MSDC-0602K, namacizumab, NC-101, NDI-010976, ND-L02-s0201 (BMS-986263), NGM-282, NGM-313, NGM-386, NGM-395, NP-011, NP-135, NP-160, norursodeoxycholic acid, NV-422, NVP-022, 0-304, obeticholic acid (OCA), 25HC3S, olesoxime, PAT-505, PAT-048, peg-ilodecakin, pioglitazone, pirfenidone, PRI-724, PX20606, Px-102, PX-L603, PX-L493, PXS-4728A, PZ-235, PZH-2109, RCYM-001, RDX-009, remogliflozin etabonate, RG-125 (AZD4076), RP-005, RPI-500, S-723595, saroglitazar, SBP-301, semaglutide, SH-2442, SHC-028, SHC-023, simtuzumab, solithromycin, sotagliflozin, statins (atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin), TCM-606F, TEV-45478, TQA-3526, TQA-3563, tipelukast (MN-001), TLY-012, TRX-318, TVB-2640, TXR-611, TXR-612, TS-20004, UD-009, UN-03, ursodeoxycholic acid, VBY-376, VBY-825, VK-2809, vismodegib, volixibat potassium ethanolate hydrate (SHP-626), VVP-100X, WAV-301, WNT-974, WXSH-0038, WXSH-0078, XEN-103, XRx-117, XTYW-003, XW-003, XW-004, XZP-5610, ZGN-839, ZG-5216, ZSYM-008, or ZYSM-007. In some embodiments, the compound of the present disclosure is combined with one or more therapeutic agents selected from an anti-obesity agent including but not limited to peptide YY or an analogue thereof, a neuropeptide Y receptor type 2 (NPYR2) agonist, a NPYR1 agonist, an NPYR5 antagonist, a cannabinoid receptor type 1 (CB1 R) antagonist, a lipase inhibitor (e.g., orlistat), a human proislet peptide (HIP), a melanocortin receptor 4 agonist (e.g., setmelanotide), a melanin concentrating hormone receptor 1 antagonist, a famesoid X receptor (FXR) agonist (e.g. obeticholic acid), apoptotic signal-regulating kinase (ASK-1) inhibitor, zonisamide, phentermine (alone or in combination with topiramate), a norepinephrine/dopamine reuptake inhibitor (e.g., buproprion), an opioid receptor antagonist (e.g., naltrexone), a combination of norepinephrine/dopamine reuptake inhibitor and opioid receptor antagonist (e.g., a combination of bupropion and naltrexone), a GDF-15 analog, sibutramine, a cholecystokinin agonist, amylin and analogues thereof (e.g., pramlintide), leptin and analogues thereof (e.g., metroleptin), a serotonergic agent (e.g., lorcaserin), a methionine aminopeptidase 2 (MetAP2) inhibitor (e.g., beloranib or ZGN-1061), phendimetrazine, diethylpropion, benzphetamine, an SGLT2 inhibitor (e.g., empagliflozin, canagliflozin, dapagliflozin, ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, or ertugliflozin), an SGLTL1 inhibitor, a dual SGLT2/SGLT1 inhibitor, a fibroblast growth factor receptor (FGFR) modulator, an AMP-activated protein kinase (AMPK) activator, biotin, a MAS receptor modulator, or a glucagon receptor agonist (alone or in combination with another GLP-1 R agonist, e.g., liraglutide, exenatide, dulaglutide, albiglutide, lixisenatide, or semaglutide), an insulin sensitizer such as thiazolidinediones (TZDs), a peroxisome proliferator-activated receptor alpha (PPARα) agonist, fish oil, an acetyl-coA carboxylase (ACC) inhibitor, a transforming growth factor beta (TGFβ) antagonist, a GDNF family receptor alpha like (GFRAL) agonist, a melanocortin-4 receptor (MC4R) agonist, including the pharmaceutically acceptable salts of the specifically named agents and the pharmaceutically acceptable solvates of said agents and salts. VII. Methods of Treatment In some embodiments, compounds of Formula (I), or pharmaceutically acceptable salt thereof, are useful in a method of treating and/or preventing a GLP-1R mediated disease or condition. In some embodiments, a method for treating and/or preventing a GLP-1R mediated disease or condition includes administering to a subject in need thereof a pharmaceutically effective amount of a compound of the present disclosure or pharmaceutically acceptable salt thereof. In some embodiments, compounds of the present disclosure have desirable properties, including for example advantageous pharmacokinetic properties, physicochemical properties such as hepatic uptake properties, and/or bile salt export pump (BSEP) inhibition characteristics. In one embodiment, compounds of the present disclosure have desirable pharmacokinetic properties, such as prolonged exposures and/or higher oral bioavailability. In one embodiment, compounds of the present disclosure have desirable hepatic uptake properties, such as reduced transporter-mediated hepatic uptake. In one embodiment, compounds of the present disclosure demonstrate desirable BSEP inhibition. In some embodiments, the disease or condition comprises a liver disease or related diseases or conditions, e.g., liver fibrosis, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), liver cirrhosis, compensated liver fibrosis, decompensated liver fibrosis, hepatocellular carcinoma, Primary Biliary Cirrhosis (PBC), or Primary Sclerosing Cholangitis (PSC). In some embodiments, the disease or condition comprises a metabolic disease or related diseases or conditions, such as diabetes mellitus, obesity, or cardiometabolic diseases. GLP-1R agonists are currently being investigated in connection with certain disorders and conditions, including for example diabetes. GLP-1 analogs that are DPP4 resistant and have longer half-lives than endogenous GLP-1 have been reported to be associated with weight loss and improved insulin action. Liraglutide, a peptide GLP-1R agonist approved in connection with treatment of diabetes, has been reported to show favorable improvements in outcomes in NASH subjects. In some embodiments, the present disclosure relates to the use of compounds of Formula (I), or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the prevention and/or treatment of a disease or condition mediated by GLP-1R, such as a liver disease or metabolic disease. In some embodiments, the present disclosure relates to the use of compounds of Formula (I), or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the prevention and/or treatment of a disease or condition mediated by GLP-1R, such as a liver disease or metabolic disease. For example, some embodiments provide a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a use thereof, for treatment and/or prevention of chronic intrahepaatic or some forms of extrahepatic cholestatic conditions, of liver fibrosis, of acute intrahepatic cholestatic conditions, of obstructive or chronic inflammatory disorders that arise out of improper bile composition, of gastrointestinal conditions with a reduced uptake of dietary fat and fat-soluble dietary vitamins, of inflammatory bowel diseases, of lipid and lipoprotein disorders, of type II diabetes and clinical complications of type I and type II diabetes, of conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and specifically triglyceride accumulation and subsequent activation of profibrotic pathways, of obesity and metabolic syndrome (combined conditions of dyslipidemia, diabetes and abnormally high body-mass index), of acute myocardial infarction, of acute stroke, of thrombosis which occurs as an endpoint of chronic obstructive atherosclerosis, of persistent infections by intracellular bacteria or parasitic protozoae, of non-malignant hyperproliferative disorders, of malignant hyperproliferative disorders, of colon adenocarcinoma and hepatocellular carcinoma for instance, of liver steatosis and associated syndromes, of liver failure or liver malfunction as an outcome of chronic liver diseases or of surgical liver resection, of Hepatitis B infection, of Hepatitis C infection and/or of cholestatic and fibrotic effects that are associated with alcohol-induced cirrhosis or with viral-borne forms of hepatitis, of type I diabetes, pre-diabetes, idiopathic type 1 diabetes, latent autoimmune diabetes, maturity onset diabetes of the young, early onset diabetes, malnutrition-related diabetes, gestational diabetes, hyperglycemia, insulin resistance, hepatic insulin resistance, impaired glucose tolerance, diabetic neuropathy, diabetic nephropathy, kidney disease, diabetic retinopathy, adipocyte dysfunction, visceral adipose deposition, obesity, eating disorders, sleep apnea, weight gain, sugar craving, dyslipidemia, hyperinsulinemia, congestive heart failure, myocardial infarction, stroke, hemorrhagic stroke, ischemic stroke, traumatic brain injury, pulmonary hypertension, restenosis after angioplasty, intermittent claudication, post-prandial lipemia, metabolic acidosis, ketosis, arthritis, left ventricular hypertrophy, Parkinson's Disease, peripheral arterial disease, macular degeneration, cataract, glomerulosclerosis, chronic renal failure, metabolic syndrome, angina pectoris, premenstrual syndrome, thrombosis, atherosclerosis, impaired glucose metabolism, or vascular restenosis. In some embodiments, a method of treating and/or preventing a non-alcoholic fatty liver disease (NAFLD), comprises administering to a subject in need thereof a compound of the present disclosure or a pharmaceutically acceptable salt thereof. The disclosure also relates to a compound according to Formula (I) or a pharmaceutical composition comprising said compound for preventive and posttraumatic treatment of a cardiovascular disorder, such as acute myocardial infarction, acute stroke, or thrombosis which occur as an endpoint of chronic obstructive atherosclerosis. In some embodiments, a method for treating and/or preventing cardiovascular disorder comprises administering a compounds of Formula (I) to a subject in need thereof. The disclosure further relates to a compound or pharmaceutical composition for the treatment and/or prevention of obesity and associated disorders such as metabolic syndrome (combined conditions of dyslipidemias, diabetes and abnormally high body-mass index) which can be overcome by GLP-1R-mediated lowering of serum triglycerides, blood glucose and increased insulin sensitivity and GLP-1R-mediated weight loss. In some embodiments, a method for treating and/or preventing a metabolic disease comprises administering a compounds of Formula (I) to a subject in need thereof. In some embodiments, a method for treating and/or preventing a metabolic disease comprises administering a compounds of Formula (I), to a subject in need thereof. In a further embodiment, the compounds or pharmaceutical composition of the present disclosure are useful in preventing and/or treating clinical complications of Type I and Type II Diabetes. Examples of such complications include diabetic nephropathy, diabetic retinopathy, diabetic neuropathies, or Peripheral Arterial Occlusive Disease (PAOD). Other clinical complications of diabetes are also encompassed by the present disclosure. In some embodiments, a method for treating and/or preventing complications of Type I and Type II Diabetes comprises administering a compounds of Formula (I) to a subject in need thereof. In some embodiments, a method for treating and/or preventing complications of Type I and Type II Diabetes comprises administering a compounds of Formula (I) to a subject in need thereof. Furthermore, conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and/or triglyceride accumulation and subsequent activation of profibrotic pathways may also be prevented and/or treated by administering the compounds or pharmaceutical composition of the present disclosure. Such conditions and diseases can include NASH and chronic cholestatic conditions in the liver, Glomerulosclerosis and Diabetic Nephropathy in the kidney, Macular Degeneration and Diabetic Retinopathy in the eye and neurodegenerative diseases, such as Alzheimer's Disease in the brain, or Diabetic Neuropathies in the peripheral nervous system. In some embodiments, a method for treating and/or preventing conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and/or triglyceride accumulation and subsequent activation of profibrotic pathways comprises administering a compounds of Formula (I) to a subject in need thereof. In some embodiments, a method for treating and/or preventing conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and/or triglyceride accumulation and subsequent activation of profibrotic pathways comprises administering a compounds of Formula (I) to a subject in need thereof. In some embodiments, a method for treating and/or preventing NASH comprises administering a compounds of Formula (I) to a subject in need thereof. In some embodiments, a method for treating and/or preventing NASH comprises administering a compounds of Formula (I) to a subject in need thereof. Further provided herein is a pharmaceutical composition for use in treating a GLP-1R mediated disease or condition described herein, comprising a compound of the present disclosure or a pharmaceutically acceptable salt thereof. The present disclosure also describes a use for the manufacture of a medicament in treating a GLP-1R mediated disease or condition comprising a compound of the present disclosure or a pharmaceutically acceptable salt thereof. Medicaments as referred to herein may be prepared by conventional processes, including the combination of a compound according to the present disclosure and a pharmaceutically acceptable carrier. Also disclosed is a compound of the present disclosure or a pharmaceutically acceptable salt thereof for the treatment of a GLP-1R mediated disease or condition. Also disclosed is a compound of the present disclosure or a pharmaceutically acceptable salt thereof for the prevention of a GLP-1R mediated disease or condition. VIII. Examples Many general references providing commonly known chemical synthetic schemes and conditions useful for synthesizing the disclosed compounds are available (see, e.g., Smith, March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 7thedition, Wiley-Interscience, 2013). Compounds as described herein can be purified by any of the means known in the art, including chromatographic means, such as high-performance liquid chromatography (HPLC), preparative thin layer chromatography, flash column chromatography and ion exchange chromatography. Any suitable stationary phase can be used, including normal and reversed phases as well as ionic resins. For example, the disclosed compounds can be purified via silica gel and/or alumina chromatography. See, e.g., Introduction to Modern Liquid Chromatography, 2nd ed., ed. L. R. Snyder and J. J. Kirkland, John Wiley and Sons, 1979; and Thin Layer Chromatography, E. Stahl (ed.), Springer-Verlag, New York, 1969. During any of the processes for preparation of the subject compounds, it may be desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups as described in standard works, such as T. W. Greene and P. G. M. Wuts, “Protective Groups in Organic Synthesis,” 4th ed., Wiley, New York 2006. The protecting groups may be removed at a convenient subsequent stage using methods known from the art. Exemplary chemical entities useful in methods of the embodiments will now be described by reference to illustrative synthetic schemes for their general preparation herein and the specific examples that follow. Artisans will recognize that, to obtain the various compounds herein, starting materials may be suitably selected so that the ultimately desired substituents will be carried through the reaction scheme with or without protection as appropriate to yield the desired product. Alternatively, it may be desirable to employ, in the place of the ultimately desired substituent, a suitable group that may be carried through the reaction scheme and replaced as appropriate with the desired substituent. Furthermore, one of skill in the art will recognize that the transformations shown in the schemes below may be performed in any order that is compatible with the functionality of the pendant groups. Each of the reactions depicted in the general schemes can be run at a temperature from about 0° C. to the reflux temperature of the organic solvent used. The Examples provided herein describe the synthesis of compounds disclosed herein as well as intermediates used to prepare the compounds. It is to be understood that individual steps described herein may be combined. It is also to be understood that separate batches of a compound may be combined and then carried forth in the next synthetic step. In the following description of the Examples, specific embodiments are described. These embodiments are described in sufficient detail to enable those skilled in the art to practice certain embodiments of the present disclosure. Other embodiments may be utilized, and logical and other changes may be made without departing from the scope of the disclosure. The embodiments are also directed to processes and intermediates useful for preparing the subject compounds or pharmaceutically acceptable salts thereof. The following description is, therefore, not intended to limit the scope of the present disclosure. In some embodiments, the present disclosure generally provides a specific enantiomer or diastereomer as the desired product, although the stereochemistry of the enantiomer or diastereomer was not determined in all cases. When the stereochemistry of the specific stereocenter in the enantiomer or diastereomer is not determined, the compound is drawn without showing any stereochemistry at that specific stereocenter even though the compound can be substantially enantiomerically or disatereomerically pure. Representative syntheses of compounds of the present disclosure are described in schemes below, and the examples that follow. The compounds detailed in the Examples were synthesized according to the general synthetic methods described below. Compounds were named using ChemDraw version 18. 1. 0. 535 (PerkinElmer Informatics, Inc.) unless otherwise indicated. Abbreviations Certain abbreviations and acronyms are used in describing the experimental details. Although most of these would be understood by one skilled in the art, Table 1 contains a list of many of these abbreviations and acronyms. TABLE 1List of Abbreviations and AcronymsAbbreviationMeaningAcacetateAcOHAcetic acidACN or MeCNacetonitrileAmPhosdi-tert-butyl(4-dimethylaminophenyl)phosphineAq.aqueousAUCArea under the curveBnbenzylBpin(pinacolato)boronB2Pin2bis(pinacolato)diboronBubutylBzbenzoylBSAbovine serum albuminBzClbenzoyl chloridecAMPCyclic adenosine monophosphatecataCXium ® AMesylate[(di(1-adamantyl)-n-butylphosphine)-2-(2′-Pd G3amino-1,1′-biphenyl)]palladium(II)CANCerium Ammonium NitrateCDI1,1′-carbonyldiimidazoleCHOChinese hamster ovaryDBAdibenzalacetoneDBU1,8-Diazabicyclo[5. 4. 0]undec-7-eneDCMdichloromethaneDCEdichlorethaneDEAdiethylamineDeoxofluorBis(2-methoxyethyl)aminosulfur trifluorideDIPEAdiisopropylethylamineDMAP4-dimethylaminopyridineDMEdimethoxyethaneDMEMDulbecco's Modified Eagle MediumDMFdimethylformamideDMSOdimethylsulfoxideDPBSDulbecco's phosphate buffered salinedppf1,1′-Ferrocenediyl-bis(diphenylphosphine)EDCIN-(3-dimethylaminopropyl)-N′-ethylcarbodiimidehydrochlorideES/MSelectron spray mass spectrometryEtethylEtOAcEthyl acetateFBSfetal bovine serumHATU1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxidehexafluorophosphateHBSSHank's balanced salt solutionHEPES(4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid)Hex or hexhexaneHPLCHigh performance liquid chromatographyIPAisopropanolJohnPhos(2-Biphenyl)di-tert-butylphosphineKOtBupotassium tert-butoxideLCliquid chromatographyLCMSliquid chromatography/mass spectrometryMCPBAmeta-chloroperbenzoic acidMemethylm/zmass to charge ratioMS or msmass spectrumNMPN-methyl-2-pyrrolidoneNMRnuclear magnetic resonanceNOEnuclear Overhauser effectNOESYnuclear Overhauser effect spectroscopyOTsTosylate, p-toluenesulfonateOTfTritiate, trifluoromethanesulfonatePd Rockphos G3[(2-Di-tert-butylphosphino-3-methoxy-6-methyl-2′,4′,6′-triisopropyl-1,1′-biphenyl)-2-(2-aminobiphenyl)]palladium(II) methanesulfonatePEG300Polyethylene glycol 300PhphenylPh3PtriphenylphosphinepinpinacolPOBy mouth/orallyPOCl3Phosphorus oxychloridePyrpyridineRBFround bottom flaskRP-HPLC or RPreverse phase high performance liquidHPLCchromatographyRT/rtroom temperatureSFCsupercritical fluid chromatographytButert-butyltBuXPhos Pd G3[(2-Di-tert-butylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)-2-(2′-amino-1,1′-biphenyl)] palladium(II)methanesulfonateTCFHN,N,N′N′-tetramethylchloroformamidiniumhexafluorophosphateTEAtriethylamineTFAtrifluoroacetic acidTf2OTrifluoromethanesulfonic anhydrideTHFtetrahydrofuranTLCThin layer chromatographyTPPOTriphenylphosphine oxideTs4-toluenesulfonylXPhos Pd G2Chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II)XPhos Pd G3(2-Dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II) methanesulfonateδparts per million referenced to residual solvent peak A. Synthesis of Intermediates Preparation of Intermediate I-1 Methyl 4-amino-3-(2-methoxyethylamino)benzoate (I-1): To a solution of methyl 3-fluoro-4-nitro-benzoate (50.0 g, 251 mmol) in THF (400 mL) was added diisopropylethylamine (70.0 mL, 402 mmol) and 2-Methoxyethylamine (34.9 mL, 402 mmol). The resulting solution was heated to 55° C. for 6 hrs. Upon completion the solvent was removed, and the resulting residue taken up in EtOAc (150 mL), washed with brine (30 mL), concentrated and carried forward without further purification. Methyl 3-(2-methoxyethylamino)-4-nitro-benzoate (20.0 g, 78.7 mmol) was then dissolved in EtOAc:EtOH (1:1, 140 mL) after which 10% palladium on carbon (5.02 g, 4.72 mmol) was then added. The resulting suspension was stirred under a hydrogen balloon at room temperature for 16 hrs. The mixture was filtered through Celite washing with EtOAc (100 mL) and concentrated to give the desired compound without further purification: ES/MS: 225.2 (M+H+). Preparation of Intermediate I-2 Methyl 4-{[2-(4-bromo-2-fluoro-phenyl)acetyl]amino}-3-(2-methoxyethylamino)benzoate: To a solution of 2-(4-bromo-2-fluoro-phenyl)acetic acid (1.00 g, 4.29 mmol) in DMF (20.0 mL) was added methyl 4-amino-3-(2-methoxyethylamino)benzoate (1.18 g, 5.28 mmol) and O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (1.96 g, 5.15 mmol) followed by N,N-diisopropylethylamine (3.74 mL, 21.5 mmol) and the mixture was stirred for 2 hr. at room temperature. The mixture was concentrated in vacuo, the residue was taken up in EtOAc and washed with water (1×) and brine (1×). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was taken forward without further purification, assuming full conversion: ES/MS m/z: 441.2 (M+H+). Methyl 2-[(4-bromo-2-fluoro-phenyl)methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-2): The crude product from the previous step, methyl 4-{[2-(4-bromo-2-fluoro-phenyl)acetyl]amino}-3-(2-methoxyethylamino)benzoate (1.89 g, 4.29 mmol) was dissolved in AcOH (40.0 mL) and the mixture was heated to 60° C. for 2 hr. The mixture was concentrated in vacuo and the crude residue was taken up in DCM and washed with saturated aqueous sodium bicarbonate. The layers were separated, and the aqueous layer was extracted with DCM (2×). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound: ES/MS m/z: 421.9 (M+H+). Preparation of Intermediate I-3 4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (I-3): To a dried 100 mL RBF was added 3-fluoro-4-(hydroxymethyl)benzonitrile (2 g, 13.2 mmol). The material was dissolved in dry THE (20 mL) under a nitrogen atmosphere at 0° C. Sodium hydride (60% dispersion in mineral oil, 0.507 g, 13.2 mmol) was added in one portion, and the mixture was stirred for 30 mins at 0° C. under N2. Subsequently, 2,6-dibromopyridine (3.13 g, 13.2 mmol) was added, and the mixture was stirred room temperature overnight. The mixture was diluted with EtOAc (100 mL) and water (20 mL). The layers were separated, and the aqueous layer was extracted with ethyl acetate (2×30 mL). The combined organic layers were dried over MgSO4, filtered, and concentrated under reduced pressure. The crude material was purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford the Intermediate I-6: ES/MS: 307.058 (M+H+);1H NMR (400 MHz, Chloroform-d) δ 7.72-7.63 (m, 1H), 7.52-7.46 (m, 2H), 7.41 (dd, J=9.2, 1.5 Hz, 1H), 7.14 (dd, J=7.5, 0.7 Hz, 1H), 6.79 (dd, J=8.2, 0.7 Hz, 1H), 5.50 (t, J=0.9 Hz, 2H). Preparation of Intermediate I-4 Methyl (S)-4-amino-3-((oxetan-2-ylmethyl)amino)benzoate (I-4): Methyl (S)-4-amino-3-((oxetan-2-ylmethyl)amino)benzoate was prepared following procedure Intermediate I-1 substituting (S)-oxetan-2-ylmethanamine for 2-methoxyethylamine. ES/MS: 237.2 (M+H+). Intermediate I-5 Ethyl 3,5-difluoro-4-nitrobenzoate: Ethyl 4-amino-3,5-difluorobenzoate (5.00 g, 24.9 mmol) was taken up in acetic acid (50.0 mL) and sulfuric acid (12.1 M, 2.05 mL, 24.9 mmol) and hydrogen peroxide (30% aqueous solution, 46.7 mL, 74.6 mmol) were added sequentially. The mixture was heated to 100° C. for 1 hour. The mixture was then cooled to room temperature and then slowly poured into 300 mL of ice water while swirling. The mixture was then diluted with EtOAc (200 mL), transferred to a separatory funnel, and the organic phase collected. The aqueous phase was extracted with EtOAc (2×100 mL) and the combined organics were dried over MgSO4and concentrated in vacuo. The residue was purified by column chromatography (eluent: EtOAc/Hexanes gradient) to afford the product. Ethyl (S)-3-fluoro-4-nitro-5-((oxetan-2-ylmethyl)amino)benzoate: Ethyl 3,5-difluoro-4-nitro-benzoate (2.50 g, 10.8 mmol) and (S)-oxetan-2-ylmethanamine (989 mg, 11.4 mol) were taken up in tetrahydrofuran (12.0 mL) and N,N-dimethylformamide (6.0 mL), and N,N-diisopropylethylamine (9.42 mL, 54.1 mmol) was added. The mixture was heated to 50° C. for 16 hours. Following this time, the mixture was concentrated in vacuo and the residue purified by column chromatography (eluent: 0-25% EtOAc/Hexanes) to afford the product. ES/MS: 299.2 (M+H+). Ethyl (S)-4-amino-3-fluoro-5-((oxetan-2-ylmethyl)amino)benzoate (I-5): Ethyl (S)-3-fluoro-4-nitro-5-((oxetan-2-ylmethyl)amino)benzoate (2.20 g, 7.38 mmol) was taken up in ethanol (10 mL) and tetrahydrofuran (5 mL) and the mixture sparged with nitrogen for 5 minutes. Palladium on carbon (10 wt. % loading, 785 mg, 0.74 mmol) was then added and sparging continued for 5 minutes. Hydrogen was then bubbled through the solution for one minute and then the mixture was set up under balloon hydrogen atmosphere for 21 hours. Following this time, the reaction was stopped, and the mixture was filtered through Celite. The filter was washed with EtOAc (2×20 mL) and methanol (2×10 mL) and the filtrate concentrated in vacuo to afford ethyl (S)-4-amino-3-fluoro-5-((oxetan-2-ylmethyl)amino)benzoate (I-5). ES/MS: 269.2 (M+H+);1H NMR (400 MHz, chloroform) δ 7.44-7.30 (m, 2H), 5.13 (qd, J=7.1, 3.4 Hz, 1H), 4.72 (ddd, J=8.7, 7.4, 6.0 Hz, 1H), 4.62 (dt, J=9.1, 6.1 Hz, 1H), 4.33 (q, J=7.1 Hz, 2H), 3.58-3.30 (m, 2H), 2.76 (dtd, J=11.4, 8.0, 6.1 Hz, 1H), 2.56 (ddt, J=11.3, 9.0, 7.1 Hz, 1H), 1.37 (t, J=7.1 Hz, 3H). Preparation of Intermediate I-6 Tert-butyl 3-((2-methoxyethyl)amino)-4-nitrobenzoate: To a 500 mL RBF was added tert-butyl 3-fluoro-4-nitrobenzoate (10 g, 41.5 mmol). The material was dissolved in THE (150 mL), and 2-methoxyethanamine (7.2 mL, 82.9 mmol) and N,N-diisopropylethylamine (21.7 mL, 124 mmol) were added. The mixture was stirred at 50° C. overnight. Afterward, the mixture was concentrated to remove most of the THF, and the crude material was dissolved in EtOAc (400 mL). The organics were washed with 50% NH4Cl (2×100 mL) and with brine (1×50 mL). The organics were subsequently dried over MgSO4, filtered, and concentrated under reduced pressure. The crude material was carried forward without further purification: ES/MS: 297.1 (M+H+);1H NMR (400 MHz, Chloroform-d) δ 8.21 (d, J=8.9 Hz, 1H), 7.55 (d, J=1.7 Hz, 1H), 7.20 (dd, J=8.9, 1.7 Hz, 1H), 3.72 (dd, J=5.8, 4.8 Hz, 2H), 3.57 (q, J=5.2 Hz, 2H), 3.46 (s, 3H), 1.62 (s, 9H). Tert-butyl 4-amino-3-((2-methoxyethyl)amino)benzoate (I-6): To a 1 L RBF was added tert-butyl 3-((2-methoxyethyl)amino)-4-nitrobenzoate (13 g, 43.9 mmol), ethanol (100 mL), and EtOAc (50 mL). The mixture was stirred and sonicated until all material was dissolved. Nitrogen was bubbled through the mixture for 5 minutes, and then palladium on carbon (10% wt, 2.33 g, 2.19 mmol) was added. Hydrogen was bubbled through the mixture for 5 minutes, and the mixture was stirred overnight under a hydrogen balloon. Nitrogen was subsequently bubbled through the flask for 10 minutes, and then the mixture was filtered through Celite to remove the catalyst. The filtrate was concentrated under reduced pressure and was used without further purification: ES/MS: 267.2 (M+H+). Preparation of Intermediate I-7 Methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetate: A suspension of methyl 2-(4-bromo-2,5-difluorophenyl)acetate (10.5 g, 39.6 mmol), Bis(neopentyl glycolato)diboron (17.9 g, 79.2 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II); PdCl2(dppf) (2.94 g, 3.96 mmol), and potassium propionate (15.6 g, 139 mmol) in dioxane (50 mL) was degassed with Ar for 20 min. The mixture was sealed and heated at 100° C. for 2 hours. Sodium carbonate (2.0 M, 39.6 mL, 79.2 mmol) was added and the mixture was stirred at RT for 10 min. [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II); PdCl2(dppf) (1.47 g, 1.98 mmol) and I-3 (14 g, 45.6 mmol) were added, the mixture was degassed for 10 min with Ar, then sealed and heated at 100° C. for 1 hour. The mixture was diluted with EtOAc and washed with brine. The organic extract was dried over sodium sulfate and chromatographed (eluent: EtOAc/hexanes) to give the title product: ES/MS: 413.2 (M+H+). 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid (I-7). A solution of methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetate (12.5 g, 30.3 mmol) and lithium hydroxide (0.2 M, 19.7 mL, 39.4 mmol) in CH3CN (50 mL) was heated at 50° C. for 2 hours. The mixture was acidified with 1 N of hydrochloride to pH=6-7. The material crashed out and was filtered by filter funnel. The solid was washed with water and dried overnight to yield the product: ES/MS: 399.2 (M+H+);1H NMR (400 MHz, Methanol-d4) δ 7.83-7.77 (m, 1H), 7.78-7.65 (m, 2H), 7.64-7.59 (m, 2H), 7.58-7.51 (m, 1H), 7.26-7.14 (m, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.63 (s, 2H), 3.73 (d, J=1.2 Hz, 2H). Preparation of Intermediate I-8 Methyl (S)-2-(4-bromo-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-8): Methyl (S)-2-(4-bromo-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared following procedure Intermediate I-2 substituting I-4 for I-1 and 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 451.0, 453.0 (M+H+). Preparation of Intermediate I-9 Methyl (S)-2-(4-(6-(benzyloxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate: Methyl (S)-2-(4-bromo-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-8) (450 mg, 0.997 mmol), Pd(dppf)Cl2(74.0 mg, 0.100 mmol), potassium propionate (336 mg, 2.99 mmol), and bis(pinacolato)diboron (304 mg, 2.99 mmol) were taken up in 1,4-dioxane (4.00 mL) and the mixture sparged with argon for 5 minutes. The mixture was then heated to 110° C. for one hour. Following this time, complete conversion to the intermediate boronate ester was observed. The mixture was cooled to rt. and aqueous sodium carbonate (2.0 M, 0.997 mL, 1.99 mmol) was added. The mixture was stirred for 5 minutes, then 2-(benzyloxy)-6-bromopyridine (290 mg, 1.10 mmol) and Pd(dppf)Cl2(37.0 mg, 0.050 mmol) were added and the mixture heated to 90° C. for 1 hour. The mixture was then loaded directly onto SiO2for purification with normal phase column chromatography (eluent: EtOAc/CH2Cl2gradient) which afforded the desired product. ES/MS: 556.2 (M+H+) Methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-9): Methyl (S)-2-(4-(6-(benzyloxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (426.0 mg, 0.767 mmol) was taken up in ethanol (6.0 mL) and tetrahydrofuran (3.0 mL) and the solution sparged with nitrogen for 5 minutes. Pd/C (408 mg, 0.383 mmol) was added and nitrogen bubbled through the suspension for an additional 5 minutes. Hydrogen was then bubbled through the solution for 5 minutes before the mixture was set up under balloon hydrogen atmosphere. The mixture was stirred at RT for 30 minutes. Following this time, the suspension was filtered through celite, washed with EtOAc (3×10 mL). The filtrate was concentrated in vacuo to afford the methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-9). ES/MS: 466.2 (M+H+) Preparation of Intermediate I-10 Methyl 2-[[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-10): To a vial, methyl 2-[(4-bromo-2-fluoro-phenyl)methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-2) (200 mg, 0.475 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (145 mg, 0.570 mmol), (1,1′-bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (33.6 mg, 0.0475 mmol) and potassium acetate (0.140 g, 1.42 mmol) was added. Next, 1,4-dioxane (4.80 mL) was added and the mixture was heated to 100° C. for 24 hr. The mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give methyl 2-[[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-10). ES/MS m/z: 469.4 (M+H+) Preparation of Intermediate I-11 Tert-butyl 2-(2,5-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-11): tert-butyl 2-(2,5-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-10 substituting 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluorophenyl)acetic acid and tert-butyl 4-amino-3-((2-methoxyethyl)amino)benzoate (I-6) for methyl 4-amino-3-(2-methoxyethylamino)benzoate. ES/MS: 529.3 (M+H+);1H NMR (400 MHz, DMSO-d6) δ 8.13 (s, 1H), 7.74 (dd, J=8.4, 1.6 Hz, 1H), 7.56 (d, J=8.4 Hz, 1H), 7.33 (dd, J=9.3, 4.6 Hz, 1H), 7.17 (dd, J=9.1, 5.5 Hz, 1H), 4.54 (t, J=5.1 Hz, 2H), 4.39 (s, 2H), 3.65 (t, J=5.1 Hz, 2H), 3.20 (s, 3H), 1.57 (s, 9H), 1.31 (s, 12H). Preparation of Intermediate I-12 Tert-butyl 2-[[4-(6-chloropyridin-2-yl)-2,5-difluorophenyl]methyl]-3-(2-methoxyethyl)-1,3-benzodiazole-5-carboxylate (I-12): tert-butyl 2-[[2,5-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methyl]-3-(2-methoxyethyl)-1,3-benzodiazole-5-carboxylate (I-11) (30.0 g, 56.7 mmol, 1.00 equivalent) and 2-bromo-6-chloropyridine (14.2 g, 73.8 mmol, 1.30 equivalent) were dissolved in 1,4-dioxane (600 mL) and H2O (60 mL). To the solution Pd(dppf)Cl2(4.15 g, 5.68 mmol, 0.1 equivalent) and K2CO3(15.7 g, 114 mmol, 2.0 equivalent) were added. The resulting solution was heated to 90° C. overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with petroleum ether/EtOAc (2/1) to afford tert-butyl 2-[[4-(6-chloropyridin-2-yl)-2,5-difluorophenyl]methyl]-3-(2-methoxyethyl)-1,3-benzodiazole-5-carboxylate (1-12). ES/MS: 513.8 (M+H+);1H NMR (400 MHz, DMSO-d6) δ 8.14 (s, 1H), 8.02 (t, J=7.9 Hz, 1H), 7.87 (d, J=7.7 Hz, 1H), 7.78-7.69 (m, 2H), 7.59 (d, J=8.2 Hz, 2H), 7.41 (dd, J=11.4, 6.0 Hz, 1H), 4.58 (t, J=5.1 Hz, 2H), 4.44 (s, 2H), 3.68 (t, J=5.1 Hz, 2H), 3.22 (s, 3H), 1.58 (s, 9H). Preparation of Intermediate I-13 Ethyl (S)-4-(2-(4-bromo-2-fluorophenyl)acetamido)-3-fluoro-5-((oxetan-2-ylmethyl)amino)benzoate: A solution of 1-5 (500 mg, 1.86 mmol) and 2-(4-bromo-2-fluorophenyl)acetic acid (521 mg, 2.24 mmol) in MeCN (9.0 mL) was cooled to 0° C. and 1-methylimidazole (765 mg, 0.74 mL, 9.32 mmol) was added followed by N,N,N′,N′-Tetramethylchloroformamidinium Hexafluorophosphate (732 mg, 2.61 mmol). The mixture was warmed to RT and stirred for 30 minutes. The crude mixture was concentrated in vacuo, then partitioned between water and EtOAc. The organic layer was isolated and washed with an additional portion of water and then brine. The isolated organic layer was dried over sodium sulfate, isolated by vacuum filtration, concentrated in vacuo, and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide ethyl (S)-4-(2-(4-bromo-2-fluorophenyl)acetamido)-3-fluoro-5-((oxetan-2-ylmethyl)amino)benzoate. ES/MS: 483.0, 485.0 [M+H]+. Ethyl (S)-2-(4-bromo-2-fluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-13): To a solution of ethyl (S)-4-(2-(4-bromo-2-fluorophenyl)acetamido)-3-fluoro-5-((oxetan-2-ylmethyl)amino)benzoate (530 mg, 1.10 mmol) in DCE (12.0 mL) was added acetic acid (1.88 mL, 32.9 mmol). The mixture was heated to 60° C. for 12 hours. The mixture was concentrated and partitioned between EtOAc and saturated aqueous sodium bicarbonate. The organic layer was isolated and dried over sodium sulfate, isolated by vacuum filtration, concentrated in vacuo, and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide ethyl (S)-2-(4-bromo-2-fluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-13). ES/MS: 465.0, 467.0 [M+H]+. Preparation of Intermediate I-14 Ethyl (S)-2-(4-bromo-2,5-difluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-14): Ethyl (S)-2-(4-bromo-2,5-difluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-13 substituting 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluorophenyl)acetic acid. ES/MS: 483.0, 485.0 (M+H+). Preparation of Intermediate I-15 Ethyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-15): Ethyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-9 substituting 1-14 for 1-8. ES/MS: 498.2 (M+H+). Preparation of Intermediate I-16 Methyl (S)-2-(4-(6-chloropyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-16): Methyl (S)-2-(4-(6-chloropyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner a described for Intermediate I-12 substituting 1-4 for 1-6. ES/MS: 484.0 (M+H+). Preparation of Intermediate 17 Tert-butyl 2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-17): Tert-butyl 2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-9 substituting tert-butyl 2-(4-bromo-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (prepared in a manner as described for intermediate 1-2 substituting 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluorophenyl)acetic acid) and 1-6 for I-1. ES/MS: 496.9 (M+H+). Preparation of Intermediate I-18 Tert-butyl 2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-18): To a solution of tert-butyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-17) (500 mg, 1.0 mmol) in toluene (5 ml), 4-bromo-1-(bromomethyl)-2-fluoro-benzene (406 mg, 1.5 mmol) and Silver carbonate (835 mg, 3 mmol) were added. The solution was stirred at 70° C. for 8 hr., cooled and filtered. The solution was concentrated and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide tert-butyl 2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-18): ES/MS: 683.2, 684.1 (M+H+). Preparation of Intermediate I-19 Methyl (S)-2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-19): Methyl (S)-2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-18 substituting 1-9 for 1-17. ES/MS: 652.3 (M+H+). Preparation of Intermediate I-20 Ethyl (S)-2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-20): Ethyl (S)-2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-18 substituting 1-15 for 1-17. ES/MS: 684.2 (M+H+). Preparation of Intermediate I-21 Methyl (S)-2-(4-(6-((5-bromothiazol-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-21): To a solution of methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-9) (500 mg, 1.07 mmol) in acetonitrile (15 mL) was added cesium carbonate (560 mg, 1.72 mmol) and 5-bromo-2-(bromomethyl)thiazole (290 mg, 1.13 mmol) and the resulting mixture stirred for 1 hr. at 50° C. Upon completion the crude mixture was filtered through celite, rinsing with DCM. The filtrate was concentrated, and the crude residue purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide methyl (S)-2-(4-(6-((5-bromothiazol-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-21). ES/MS: 643.0 (M+H+). Preparation of Intermediate I-22 Ethyl (S)-2-(4-(6-((5-bromothiazol-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-22): Ethyl (S)-2-(4-(6-((5-bromothiazol-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-21 substituting I-15 for I-17. ES/MS: 673.0 (M+H+). Preparation of Intermediate I-23 Methyl (S)-2-(4-(6-((6-chloro-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-23): Methyl (S)-2-(4-(6-((6-chloro-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-19 substituting 5-(bromomethyl)-2-chloro-4-fluoropyridine for 4-bromo-1-(bromomethyl)-2-fluoro-benzene ES/MS: 609.2 (M+H+). Preparation of Intermediate I-24 Methyl (S)-2-(4-(6-((6-bromo-4-chloropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-24): Methyl (S)-2-(4-(6-((6-bromo-4-chloropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-19 substituting 2-bromo-5-(bromomethyl)-4-chloropyridine for 4-bromo-1-(bromomethyl)-2-fluoro-benzene ES/MS: 654.0, 656.0 (M+H+). Preparation of Intermediate I-25 Methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (I-25): Methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-1 substituting (±)-4,4-dimethyltetrahydrofuran-3-amine for 2-methoxyethylamine, as follows: to a solution of methyl 3-fluoro-4-nitro-benzoate (3.94 g, 19.8 mmol) and 4,4-dimethyltetrahydrofuran-3-amine hydrochloride (3.00 g, 19.8 mmol) in 2-methyltetrahydrofuran (40 mL) under argon was added DIPEA (17.2 mL, 98.9 mmol). The resulting solution was refluxed at 80° C. for 3 days. The mixture was concentrated in vacuo and partitioned between EtOAc and water. The aqueous phase was extracted with additional EtOAc. The combined organic phase was washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The resulting residue was redissolved in EtOH (52 ml) and tetrahydrofuran (26 mL) under argon, then 10% palladium on carbon (2.1 g, 1.98 mmol). The mixture was cycled between argon and vacuum, then placed under hydrogen atmosphere and stirred at rt for 18 hrs. The mixture was filtered through Celite and concentrated in vacuo. The crude was purified by silica gel flash column chromatography (EtOAc/hexane gradient) to yield methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (Intermediate I-25). ES/MS: 265.2 (M+H+). Preparation of Intermediate I-26 Tert-butyl (R)-4-amino-3-((2-methoxypropyl)amino)benzoate (I-26): Tert-butyl (R)-4-amino-3-((2-methoxypropyl)amino)benzoate was prepared in a manner as described for Intermediate I-6 substituting (R)-2-methoxypropan-1-amine for 2-methoxyethylamine. ES/MS: 281.1 (M+H+). Preparation of Intermediate I-27 Methyl 5-(((6-bromopyridin-2-yl)oxy)methyl)picolinate: To a solution of 6-bromopyridin-2-ol (3.00 g, 17 mmol) in acetonitrile (100 mL) was added silver carbonate (10.2 g, 37 mmol) and methyl 5-(bromomethyl)pyridine-2-carboxylate (5.00 g, 22 mmol) and the resultant mixture heated to 60° C. for 3 hours. Upon completion the mixture was filtered through celite, concentrated and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide the desired product. ES/MS: 325.2 (M+H+). 5-(((6-bromopyridin-2-yl)oxy)methyl)picolinic acid (I-27): To a solution of methyl 5-(((6-bromopyridin-2-yl)oxy)methyl)picolinate (5.57 g, 17 mmol) in acetonitrile (75 mL) was added lithium hydroxide (2.17 g, 51.7 mmol) as an solution in water (25 mL) and mixture stirred at RT temperature for 1 hr. pH was adjusted to ˜6 with 1N HCl, after which the mixture was diluted with EtOAc (200 mL) and the layers separated. The organic layer was washed with brine, dried over MgSO4, filtered and concentrated to give 5-(((6-bromopyridin-2-yl)oxy)methyl)picolinic acid (I-27) without further purification. ES/MS: 309.1 (M+H+). Preparation of Intermediate I-28 5-(((6-bromopyridin-2-yl)oxy)methyl)-N-(1-cyanocyclopropyl)picolinamide (I-28): To a solution of 5-(((6-bromopyridin-2-yl)oxy)methyl)picolinic acid (I-27) (5.32 g, 17 mmol) in DMF (80 mL), 1-aminocyclopropanecarbonitrile hydrochloride (3.14 g, 26 mmol), HATU (9.62 g, 25 mmol), and diisopropylethylamine (12 mL, 69 mmol) was added sequentially. The resultant solution was stirred at RT temperature for 30 minutes. Upon completion the mixture was diluted with EtOAc (250 mL, wash with water (2×50 mL), brine (1×40 mL), dried over MgSO4, filtered and concentrated to give 5-(((6-bromopyridin-2-yl)oxy)methyl)-N-(1-cyanocyclopropyl)picolinamide (I-28) without further purification. ES/MS: 375.1 (M+H+). The following intermediates were synthesized in an analogous manner as described for Intermediate I-28 Preparation of Intermediates I-29 and I-30 Tert-butyl 4-amino-3-(((3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)amino)benzoate (I-29 and I-30): Tert-butyl 3-(((3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)amino)-4-nitrobenzoate: A solution of tert-butyl 3-fluoro-4-nitro-benzoate (0.150 g, 0.622 mmol), (3aS,6aR)-2,3,3a,4,5,6a-hexahydrofuro[2,3-b]furan-4-amine (0.0984 g, 0.762 mmol), and N-ethyl-N-isopropyl-propan-2-amine (0.325 mL, 1.87 mmol) in NMP (4 mL) was heated at 90 C overnight. The mixture was diluted with EtOAc and washed with 5% LiCl and brine. The organic extract was dried over sodium sulfate and purified by flash chromatography (eluent: EtOAc/hexanes) to give tert-butyl 3-(((3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)amino)-4-nitrobenzoate as an unseparable mixture of two compounds. ES/MS: 351.0 (M+H+). Tert-butyl 4-amino-3-(((3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)amino)benzoate (I-29 and I-30): A solution of tert-butyl 3-[[(3aS,6aR)-2,3,3a,4,5,6a-hexahydrofuro[2,3-b]furan-4-yl]amino]-4-nitro-benzoate (156 mg, 0.445 mmol) in EtOH (15 mL) was degassed by cycling the mixture between argon and vacuum 3×. Pd/C (10.0%, 47.4 mg, 0.0445 mmol) was added to the solution and then the solution was degassed 1× by cycling the mixture between argon and vacuum and stirred at RT with a balloon of hydrogen overnight. The mixture was filtered over a Celite plug and rinsed with EtOAc. Concentrated and purified by flash chromatography (eluent: 30 to 40% EtOAc/hexanes) to give two distinct isomers of tert-butyl 4-amino-3-(((3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)amino)benzoate (I-29 and I- Isomer 1 (Less Polar, Eluted First), I-29 ES/MS: 321.0 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.45 (dd, J=8.1, 1.8 Hz, 1H), 7.37 (d, J=1.8 Hz, 1H), 6.74 (d, J=8.1 Hz, 1H), 5.87 (d, J=5.1 Hz, 1H), 4.32-4.06 (m, 2H), 4.05-3.81 (m, 2H), 3.66 (t, J=8.7 Hz, 1H), 3.24 (tt, J=8.7, 4.2 Hz, 1H), 2.03-1.90 (m, 1H), 1.90-1.79 (m, 1H), 1.60 (s, 9H). Isomer 2 (More Polar, Eluted Second), I-30 ES/MS: 321.0 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.45 (dd, J=8.1, 1.8 Hz, 1H), 7.38 (d, J=1.8 Hz, 1H), 6.75 (d, J=8.0 Hz, 1H), 5.87 (d, J=5.1 Hz, 1H), 4.34-4.05 (m, 2H), 4.02-3.82 (m, 2H), 3.67 (t, J=8.6 Hz, 1H), 3.24 (tt, J=8.6, 4.2 Hz, 1H), 2.02-1.81 (m, 2H), 1.60 (s, 9H). Preparation of Intermediate I-31 (5-bromo-3-fluoropyridin-2-yl)methyl 4-methylbenzenesulfonate (I-31): (5-bromo-3-fluoro-2-pyridyl)methanol (200 mg, 0.97 mmol), p-Toluenesulfonic anhydride (350 mg, 1.1 mmol), diisopropylethylamine (0.34 mL, 1.9 mmol), and DCM (10 mL were combined and stirred at ambient temperature for 16 hours. Upon completion the mixture was washed with saturated aqueous NaHCO3(5 mL) and brine (5 mL), dried over MgSO4, filtered and concentrated to give (5-bromo-3-fluoropyridin-2-yl)methyl 4-methylbenzenesulfonate (I-31), which was used without further purification. ES/MS The following intermediate was prepared in a manner as described for Intermediate I-31 Preparation of Intermediate I-32 (5-chloropyrazin-2-yl)methyl 4-methylbenzenesulfonate (I-32): (5-chloropyrazin-2-yl)methyl 4-methylbenzenesulfonate was prepared in a manner as described for Intermediate I-31 substituting (5-chloropyrazin-2-yl)methanol for (5-bromo-3-fluoro-2-pyridyl)methanol. Preparation of Intermediate I-33 Tert-butyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-33): To a solution of tert-butyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-17) (400 mg, 0.80 mmol) and (5-bromo-3-fluoro-2-pyridyl)methyl 4-methylbenzenesulfonate (I-31) (350 mg, 0.97 mmol) in 15 mL of acetonitrile was added Cs2CO3(400 mg, 1.20 mmol). The solution was then heated to 50° C. for 30 minutes. The solution was cooled to RT, filtered, and then concentrated. The crude material was purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide tert-butyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-33): product. ES/MS: 686.0 (M+H+). Preparation of Intermediate I-34 Tert-butyl 2-[[4-[6-[(5-chloropyrazin-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-34): tert-butyl 2-[[4-[6-[(5-chloropyrazin-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-33 substituting I-31 for I-32. ES/MS: 566.0 (M+H+). Preparation of Intermediate I-35 4-fluoro-5-(hydroxymethyl)thiophene-2-carbonitrile: (5-bromo-3-fluoro-2-thienyl)methanol (220 mg, 1.04 mmol), zinc cyanide (182 mg, 1.55 mmol), zinc powder (3 mg, 0.05 mmol), and Pd(PPh3)4(300 mg, 0.26 mmol) in DMF (10 mL) was degassed by bubbling argon for 1 minute, sealed and heated to 100° C. for 20 hours. Upon completion, the mixture was poured into H2O (10 mL) and extracted with EtOAc (2×20 mL). The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give 4-fluoro-5-(hydroxymethyl)thiophene-2-carbonitrile. ES/MS: 158.2 (M+1). 5-(bromomethyl)-4-fluoro-thiophene-2-carbonitrile (I-35): Carbon tetrabromide (111 mg, 0.34 mmol was added to a solution comprising 4-fluoro-5-(hydroxymethyl)thiophene-2-carbonitrile (48 mg, 0.31 mmol), triphenylphosphine (88 mg, 0.34 mmol) in DCM (3 mL) at RT. The mixture was stirred for 30 min at RT. Upon completion the mixture was concentrated and purified by flash chromatography (Eluent: EtOAc/hexane) to give 5-(bromomethyl)-4-fluoro-thiophene-2-carbonitrile (I-35). ES/MS: 221.2 (M+1). Preparation of Intermediate I-36 [5-(difluoromethyl)thiazol-2-yl]methanol: To a solution of [5-(difluoromethyl)thiazole-2-carbonyl]oxysodium (200 mg, 1.08 mmol) in DCM (5 mL), at RT, was added oxalyl chloride (2.0 M in DCM, 0.65 mL, 1.3 mmol). After stirring for 1 hour at RT, MeOH (1 mL) was added and the mixture was stirred for additional 30 minutes before pouring into H2O (10 mL) and extracted with EtOAc (2×20 mL). The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, and concentrated. The residue was re-dissolved in THE (5 mL) and cooled to 0° C. Diisobutylaluminium hydride (1.0 M in DCM, 3.3 mL, 3.3 mmol) was added, and the mixture was warmed to RT and stirred for 1 hour. Upon completion, the reaction was quenched with 2M NaOH (0.4 mL), H2O (0.4 mL), and diluted with EtOAc (10 mL). The mixture was then filtered through a plug of Celite. The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give the desired product. ES/MS: 166.2 (M+1). 2-(bromomethyl)-5-(difluoromethyl)thiazole (I-36): [5-(difluoromethyl)thiazol-2-yl]methanol (88 mg, 0.53 mmol), triphenylphosphine (150 mg, 0.56 mmol) in DCM (3 mL) was added carbon tetrabromide (190 mg, 0.56 mmol) at rt. The mixture was stirred at rt for 30 min. Upon completion, the mixture was concentrated and purified by flash chromatography (Eluent: EtOAc/hexane) to give I-36. ES/MS: 229.2 (M+1). Preparation of Intermediate I-37 [5-(2,2-difluoroethoxy)thiazol-2-yl]methanol: A suspension of methyl 5-hydroxythiazole-2-carboxylate (200 mg, 1.3 mmol), 2,2-difluoroethyl trifluoromethanesulfonate (300 mg, 1.4 mmol), and cesium carbonate (610 mg, 1.9 mmol) in MeCN (5 mL) was stirred at RT for 16 h. Upon completion, the mixture was filtered through a plug of Celite and concentrated. The residue was re-dissolved in THE (5 mL) and cooled to 0° C. Diisobutylaluminium hydride (1.0 M in DCM, 2.8 mL, 2.8 mmol) was added, and the mixture was warmed to RT and stirred for 1 hour. Upon completion, the reaction was quenched with 2M NaOH (0.4 mL), H2O (0.4 mL) and diluted with EtOAc (10 mL). The mixture was then filtered through a plug of Celite. The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give the desired product. 2-(bromomethyl)-5-(2,2-difluoroethoxy)thiazole (I-37): [5-(2,2-difluoroethoxy)thiazol-2-yl]methanol (91 mg, 0.47 mmol), triphenylphosphine (125 mg, 0.48 mmol) in DCM (5 mL) was added carbon tetrabromide (160 mg, 0.48 mmol) at RT. The mixture was stirred for 30 min at RT. Upon completion the mixture was concentrated and purified by flash chromatography (Eluent: EtOAc/hexane) to give I-37. ES/MS: 258.2 (M+1). Preparation of Intermediate I-38 2-(bromomethyl)-5-(2,2,2-trifluoroethoxy)thiazole (I-38): 2-(bromomethyl)-5-(2,2,2-trifluoroethoxy)thiazole was prepared in a manner as described for Intermediate I-37 substituting 2,2-difluoroethyl trifluoromethanesulfonate for 2,2,2-trifluoroethyl trifluoromethanesulfonate. ES/MS: 277.2 (M+1). Preparation of Intermediate I-39 Methyl (S)-2-(4-(6-((5-bromothiophen-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-39): Methyl (S)-2-(4-(6-((5-bromothiophen-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-19 substituting 5-(bromomethyl)-2-chloro-4-fluoropyridine for 2-bromo-5-(bromomethyl)thiophene ES/MS: 642.0 (M+H+). Preparation of Intermediate I-40 Methyl (S)-2-(4-(6-((5-bromo-3-fluorothiophen-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-40): Methyl (S)-2-(4-(6-((5-bromo-3-fluorothiophen-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-19 substituting 5-(bromomethyl)-2-chloro-4-fluoropyridine for 5-bromo-2-(bromomethyl)-3-fluorothiophene ES/MS: 660.0 (M+H+). Preparation of Intermediate I-41 Methyl 2-[[4-[6-[(5-bromo-1,3,4-thiadiazol-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-41): methyl 2-[[4-[6-[(5-bromo-1,3,4-thiadiazol-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-19 substituting 5-(bromomethyl)-2-chloro-4-fluoropyridine for 2-bromo-5-(bromomethyl)-1,3,4-thiadiazole ES/MS: 660.0 (M+H+). Preparation of Intermediate I-42 Thiazolo[5,4-b]pyridin-2-ylmethanol: methyl thiazolo[5,4-b]pyridine-2-carboxylate (100 mg, 0.52 mmol) in THE (5 mL) was cooled to 0° C. Diisobutylaluminium hydride (1.0 M in DCM, 1.5 mL, 1.5 mmol) was added, and the mixture was warmed to rt and stirred for 1 hour. Upon completion, the reaction was quenched with 2M NaOH (0.4 mL), H2O (0.4 mL), and diluted with EtOAc (10 mL). The mixture was then filtered through a plug of Celite. The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give the titled product. ES/MS: 167.2 (M+1). Thiazolo[5,4-b]pyridin-2-ylmethyl 4-methylbenzenesulfonate (I-42): To a solution of thiazolo[5,4-b]pyridin-2-ylmethanol (40 mg, 0.24 mmol), triethylamine (0.07 mL, 0.5 mmol) in DCM (5 mL) was added toluene-4-sulfonyl chloride (46 mg, 0.24 mmol) at rt. The mixture was stirred at rt for 20 hours. Upon completion the mixture was concentrated and purified by flash chromatography (Eluent: EtOAc/hexane) to give the title product. ES/MS: 321.2 (M+1). Preparation of Intermediate I-43 2-bromo-6-[(1-methylimidazol-4-yl)methoxy]pyridine (I-43): To a solution of 2-bromo-6-fluoro-pyridine (90.7 mg, 0.52 mmol) and (1-methylimidazol-4-yl)methanol (75.1 mg, 0.67 mmol) in 2.0 mL of acetonitrile was added Cs2CO3(338 mg, 1.04 mmol). The solution was then heated to 50° C. for 30 minutes. The solution was cooled to rt, filtered, then concentrated. The crude material was purified by normal phase chromatography 1-12% DCM/MeOH. The product containing fractions were combined and concentrated to give the title product. ES/MS m/z: 268.0, 270.2 (M+H+). The following intermediates were synthesized in a manner as described for intermediate I-43 Preparation of Intermediate I-44 2-bromo-6-[[1-(difluoromethyl)-2-methyl-imidazol-4-yl]methoxy]pyridine (I-44): This intermediate was prepared in a manner as described for Intermediate I-43 substituting [1-(difluoromethyl)-2-methyl-imidazol-4-yl]methanol for (1-methylimidazol-4-yl)methanol. ES/MS m/z: 318.2 (M+H+). Preparation of Intermediate I-45 Methyl 2-[(7-bromo-1,3-dihydroisobenzofuran-4-yl)methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-45): This intermediate was prepared in a manner as described for Intermediate I-13 substituting 2-(7-bromo-1,3-dihydroisobenzofuran-4-yl)acetic acid for (2-(4-bromo-2-fluorophenyl)acetic acid. ES/MS m/z: 447.1 (M+H+). Preparation of Intermediate I-46 Tert-butyl (3R,4R)-3-(2-amino-5-methoxycarbonyl-anilino)-4-fluoro-pyrrolidine-1-carboxylate (I-46): This intermediate was prepared in a manner as described for Intermediate I-6 substituting tert-butyl (3R,4R)-3-amino-4-fluoro-pyrrolidine-1-carboxylate for 2-methoxyethanamine. ES/MS m/z: 418.4 (M+Na+). Preparation of Intermediate I-47 Tert-butyl N-tert-butoxycarbonyl-N-(6-chloro-2-methylsulfanyl-pyrimidin-4-yl)carbamate: A solution of methylsulfanyl-pyrimidin-4-amine (1 g, 5.7 mmol), tert-butoxycarbonyl tert-butyl carbonate (2.61 g, 12 mmol), ethyldiisopropylamine (3 mL, 17.1 mmol) and 4-dimethylaminopyridine (140 mg, 1.14 mmol) in 20 mL of DCM, was stirred overnight. The mixture was washed with H2O and brine. The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-50% EtOAc in hexane) to give the title compound: ES/MS m/z: 375.2 (M+H+). Tert-butyl N-tert-butoxycarbonyl-N-[6-chloro-2-[(4-cyano-2-fluoro-phenyl)methoxy]pyrimidin-4-yl]carbamate (I-47): A solution of tert-butyl N-tert-butoxycarbonyl-N-(6-chloro-2-methylsulfanyl-pyrimidin-4-yl)carbamate (0.5 g, 1.33 mmol) and 3-chloroperoxybenzoic acid (0.6 g, 2.7 mmol, 77%) in 5 mL of DCM, was stirred for overnight. The mixture was washed with H2O and brine. The solvent was removed, and the resulting residue was dried in vacuo. The crude product was dissolved in 3 mL of DMF; to this solution 3-fluoro-4-(hydroxymethyl)benzonitrile (220 mg, 1.46 mmol) and potassium carbonate (366 mg, 2.65 mmol) was added and let sit at rt for 2 hr. After the 2 hours, the resulting solution was diluted with EtOAc (50 mL) and washed with H2O. The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-50% EtOAc in hexane) to give the title compound: ES/MS m/z: 479.1 (M+H+). Preparation of Intermediate I-48 2-[(2,6-dichloro-4-pyridyl)oxymethoxy]ethyl-trimethyl-silane: A solution of 2,6-dichloropyridin-4-ol (1 g, 6.1 mmol) and 2-(chloromethoxy)ethyl-trimethyl-silane (1.07 g, 6.4 mmol) in 10 mL of THF, lithium bis(trimethylsilyl)amide (6.4 mL, 6.4 mmol) was stirred overnight. The solution was washed with H2O and brine. The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-50% EtOAc in hexane) to give the title compound: ES/MS m/z: 294.1 (M+H+). 4-[(6-chloro-4-hydroxy-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (I-48): To a solution of 2-[(2,6-dichloro-4-pyridyl)oxymethoxy]ethyl-trimethyl-silane (1.0 g, 3.4 mmol) was dissolved in 5 mL of DMF, 3-fluoro-4-(hydroxymethyl)benzonitrile (0.77 g, 5.1 mmol) and potassium carbonate (0.94 g, 6.8 mmol) was added. The solution was heated to 120° C. for 6 hours. Upon completion, the solution was diluted with EtOAc (50 mL) and washed with H2O. The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude product was dissolved in 2 mL of THF. To the resulting solution tetrabutylammonium fluoride (4.0 mL, 4 mmol) was added. The mixture was stirred for 3 hr. The solvent was removed, and the resulting residue was purified by column chromatography (0-80% EtOAc in hexane) to give the title compound: ES/MS m/z: 279.2 (M+H+). Preparation of Intermediate I-49 4-[[6-chloro-4-(difluoromethyl)-2-pyridyl]oxymethyl]-3-fluoro-benzonitrile (I-49): To a solution of 2,6-dichloro-4-(difluoromethyl)pyridine (927 mg, 4.68 mmol) was dissolved in 10 mL of DMF, 3-fluoro-4-(hydroxymethyl)benzonitrile (708 mg, 4.68 mmol) and potassium carbonate (971 mg, 7 mmol) was added to the solution. It was heated to 60° C. overnight and diluted with EtOAc (50 mL) and washed with water. The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The solvent was removed, and the resulting residue was purified by column chromatography (0-60% EtOAc in hexane) to give the title compound: ES/MS m/z: 313.1 (M+H+). Preparation of Intermediate I-50 Methyl 6-(4-cyclopropyltriazol-1-yl)pyridine-3-carboxylate: A suspension of methyl 6-chloropyridine-3-carboxylate (300 mg, 1.75 mmol) and sodium azide (227 mg, 3.5 mmol) in THF, was heated to 60° C. for 5 hours. Upon completion, the mixture was diluted with EtOAc and washed with saturated solution of sodium bicarbonate (20 mL) and brine. The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. To the crude residue, ethynylcyclopropane (145 mg, 2.2 mmol) in tert-butanol (5 mL) was added. To the resulting mixtures, solutions of sodium ascorbate (0.19 mmol, 38 mg) in water (2.5 mL) and copper sulfate pentahydrate (0.19 mmol, 48 mg) in H2O (2.5 mL) were sequentially added. The mixture was stirred at rt conditions for 18 hr. Upon completion the mixture was diluted with 5 mL 1M aq. NH4OH and extracted with EtOAc (2×30 mL). The combined organic extracts were washed with brine, dried over Na2SO4and concentrated under reduced pressure. Crude product was purified by flash chromatography on silica gel (0-100% EtOAc in hexane) to give the title compound: ES/MS m/z: 245.2 (M+H+). [6-(4-cyclopropyltriazol-1-yl)-3-pyridyl]methyl 4-methylbenzenesulfonate (I-50): To a solution of methyl 6-(4-cyclopropyltriazol-1-yl)pyridine-3-carboxylate (300 mg, 1.23 mmol) in 2 mL of THF, 0.92 mL of 2 N of lithium borohydride in THF was added. The resulting solution was stirred for 8 hours and then diluted with 50 mL of EtOAc and washed with H2O and brine. The combined organic extracts were washed with brine, dried over Na2SO4and concentrated under reduced pressure. The resulting crude product was dissolved in 5 mL of DCM. Next, p-tolylsulfonyl 4-methylbenzenesulfonate (365 mg, 1.12 mmol) and ethyldiisopropylamine (0.37 mL, 2.13 mmol) was added to the solution. The mixture was stirred for overnight. Upon completion the resulting solution was diluted with 20 mL of DCM and washed with H2O and brine. The organic layer was dried over Na2SO4and concentrated under reduced pressure. The crude product was purified by flash chromatography on silica gel (0-100% EtOAc in hexane) to give the title compound: ES/MS m/z: 371.2 (M+H+). Preparation of Intermediate I-51 Methyl 6-(3-cyclopropyl-1,2,4-triazol-1-yl)pyridine-3-carboxylate: A suspension of methyl 6-chloropyridine-3-carboxylate (300 mg, 1.75 mmol), 3-cyclopropyl-1H-1,2,4-triazole (191 mg, 1.75 mmol) and potassium carbonate (483 mg, 3.5 mmol) in THF, was heated to reflux for 8 hours. Following this time, the solution was diluted with EtOAc and washed with saturated solution of sodium bicarbonate (20 mL) and brine. The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by flash chromatography on silica gel (0-100% EtOAc in hexane) to give the title compound: ES/MS m/z: 245.2 (M+H+). [6-(3-cyclopropyl-1,2,4-triazol-1-yl)-3-pyridyl]methyl 4-methylbenzenesulfonate (I-51): To a solution of methyl 6-(3-cyclopropyl-1,2,4-triazol-1-yl)pyridine-3-carboxylate (300 mg, 1.23 mmol) in 2 mL of THF, 0.92 mL of 2 N of lithium borohydride in THF was added. The solution was stirred overnight and diluted with 50 mL of EtOAc and washed with water and brine. The combined organic extracts were washed with brine, dried over Na2SO4and concentrated under reduced pressure. The crude product was dissolved in 5 mL of DCM, 4-methylbenzenesulfonyl chloride (241 mg, 1.26 mmol) and triethylamine (0.34 mL, 2.4 mmol) was added to the solution. The mixture was stirred for overnight and diluted with 20 mL of DCM and then washed with water and brine. The organic layer was dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by flash chromatography on silica gel (0-100% EtOAc in hexane) to give 1-51: ES/MS m/z: 371.1 (M+H+). Preparation of Intermediate I-52 [2-fluoro-4-(2-trimethylsilylethynyl)phenyl]methyl 4-methylbenzenesulfonate: (2-fluoro-4-iodo-phenyl)methanol (2 g, 7.94 mmol), ethynyl(trimethyl)silane (1.17 g, 11.9 mmol), copper iodide (75.6 mg, 0.4 mmol), bis(triphenylphosphine)palladium chloride (280 mg, 0.4 mmole) and triethylamine (3.3 mL, 23.8 mmol) was suspended in THE (15 mL). The mixture was degassed with nitrogen and stirred for 16 hours at rt. Following this, the mixture was filtered with celite and washed with 20 mL of DCM three times. The solvent was removed, and the resulting crude product was dried in vacuo. Next, the crude product was dissolved in 20 mL of DCM, followed by the addition of p-tolylsulfonyl 4-methylbenzenesulfonate (2.58 g, 7.92 mmol) and ethyldiisopropylamine (2.76 mL, 15.8 mmol) to the solution. The mixture was stirred for 5 hours and checked via LC/MS. Then the mixture was then diluted with 20 mL of DCM and washed with water and brine. The organic layer was dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by flash chromatography on silica gel (0-50% EtOAc in hexane) to give the title compound: ES/MS m/z: 377.1 (M+H+). Methyl 2-[[4-[6-[(4-ethynyl-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-52): Potassium carbonate (742 mg, 5.37 mmol) was added to a solution of methyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (500 mg, 1.07 mmol) and [2-fluoro-4-(2-trimethylsilylethynyl)phenyl]methyl 4-methylbenzenesulfonate (420 mg, 1.12 mmol) in 5 mL of DMF, and stirred for 3 hours. Following this time, 50 mL of water was added to the solution and the aqueous phase was extracted 2×EtOAc. The combined organic phase was dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude product was dissolved in MeOH (20 mL) and potassium carbonate (51.6 mg, 0.37 mmol) was added to the solution. After 2 hours, the solution was filtered, and the filtrate was concentrated in vacuo. The crude residue was purified by flash chromatography on silica gel (0-80% EtOAc in hexane) to give I-52. ES/MS m/z: 598.2 (M+H+). Preparation of Intermediate I-53 N-(1-cyanocyclopropyl)-4-methoxy-5-methyl-pyridine-2-carboxamide: N,N-Diisopropylethylamine (2.14 mL, 12.3 mmol) was added to a solution of 4-methoxy-5-methyl-pyridine-2-carboxylic acid; hydrochloride (500 mg, 2.46 mmol), 1-aminocyclopropanecarbonitrile; hydrochloride (349 mg, 2.95 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (1373 mg, 3.61 mmol) in DMF (10 mL). The mixture was stirred at rt overnight. Following this time, the mixture was diluted with EtOAc and washed with 5% LiCl, saturated NaHCO3, and brine. The organic extract was dried over sodium sulfate and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. 5-(bromomethyl)-N-(1-cyanocyclopropyl)-4-methoxy-pyridine-2-carboxamide: To a suspension of N-(1-cyanocyclopropyl)-4-methoxy-5-methyl-pyridine-2-carboxamide (400 mg, 1.73 mmol) in CCl4 (10 mL), was added N-Bromosuccinimide (403 mg, 2.26 mmol) followed by benzoyl peroxide (45.1 mg, 0.186 mmol). The resulting solution was heated at 90° C. for 1 hr. Upon completion the mixture was cooled to rt, diluted with 5 mL hexanes and the suspension was filtered. The filtrate was concentrated and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 310, 312 (M+H+). 5-[(6-bromo-2-pyridyl)oxymethyl]-N-(1-cyanocyclopropyl)-4-methoxy-pyridine-2-carboxamide (Intermediate I-53): A suspension of 5-(bromomethyl)-N-(1-cyanocyclopropyl)-4-methoxy-pyridine-2-carboxamide (112 mg, 0.36 mmol), 6-bromopyridin-2-ol (50 mg, 0.29 mmol), and silver carbonate (169 mg, 0.61 mmol) in CH3CN (5 mL) was heated at 50° C. overnight. Upon completion, the mixture was diluted with EtOAc and brine-filtered over Celite frit. The mixture was partitioned, and the organic phase was washed one more time with brine. The crude produced was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: EtOAc/hexanes) to give I-53. ES/MS: 403, 405 (M+H+). Preparation of Intermediate I-54 3-(bromomethyl)-1-cyclopropyl-pyrazole (I-54): Carbon tetrabromide (0.541 g, 0.00163 mol) was added to a solution of (1-cyclopropylpyrazol-3-yl)methanol (0.188 g, 1.36 mmol) and (4-diphenylphosphanylphenyl polymer bound) (78.7%, 0.542 g, 0.00163 mol) in DCM (10 mL) at 0° C. The mixture was gradually warmed to rt and stirred overnight. The resulting suspension was filtered, and the filtrate was diluted with DCM and washed with brine. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: EtOAc/hexanes) to give I-54. ES/MS: 201.2, 203.2 (M+H+). The following intermediates were prepared in a manner as described for intermediate I-54 Preparation of Intermediate I-55 [1-(oxetan-3-yl)pyrazol-3-yl]methanol: Diisobutylaluminium hydride (1000 mmol/L in DCM, 2.40 mL, 2.40 mmol) was added to a solution of methyl 1-(oxetan-3-yl)pyrazole-3-carboxylate (175 mg, 0.961 mmol) in THE (5 mL) at 0° C. and stirred for 1 hr. Following this time, the mixture was diluted with 5 mL Et2O and cooled to 0° C. Then, 0.100 mL water, 0.100 mL 15% NaOH, and 0.240 mL water was added. The solution was warmed to rt and stirred for 15 min. Following this time, MgSO4was added and the solution was stirred for an additional 15 min, then filtered to give titled product that was carried onto the next step without further purification. ES/MS: 155.2 (M+H+). 3-(bromomethyl)-1-(oxetan-3-yl)pyrazole (I-55): Carbon tetrabromide (0.281 g, 0.000848 mol) was added to a solution of [1-(oxetan-3-yl)pyrazol-3-yl]methanol (0.109 g, 0.707 mmol) and (4-diphenylphosphanylphenyl polymer bound) (78.7%, 0.282 g, 0.000848 mol) in DCM (10 mL) at 0° C. The mixture was gradually warmed to rt and stirred overnight. The resulting suspension was filtered, and the filtrate was diluted with DCM and washed with brine. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: Et2O/hexanes) to give I-55. ES/MS: 217.2, 219.2 (M+H+). Preparation of Intermediate I-56 Methyl 1-(4-pyridyl)pyrazole-3-carboxylate: In a 40 mL glass vial, a mixture of methyl 1H-pyrazole-3-carboxylate (472 mg, 3.74 mmol), 4-fluoropyridine; hydrochloride (500 mg, 3.74 mmol), and potassium carbonate (1358 mg, 9.83 mmol) in NMP (10 mL) was heated at 120° C. for 48 hr. Following this time, the mixture was diluted with EtOAc and washed with LiCl 5% 2× and brine. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 204.2 (M+H+). [1-(4-pyridyl)pyrazol-3-yl]methanol (I-56): To a solution of methyl 1-(4-pyridyl)pyrazole-3-carboxylate (106 mg, 0.520 mmol) in THE (5 mL) at 0° C., was added diisobutylaluminium hydride (1.0 M in DCM, 1.30 mL, 1.30 mmol). The solution was stirred for 1 hr. Following this time, the mixture was diluted with 5 mL Et2O and cooled to 0° C. Upon completion of the cooling, 0.05 mL water, 0.05 mL 15% NaOH, and 0.130 mL water was added to the solution. The solution was then warmed to rt and stirred for 15 min, followed by the addition of MgSO4. The solution was stirred and additional 15 min, and then filtered. The crude product was purified by flash chromatography (eluent: EtOAc/hexanes) to give I-56. ES/MS: 176.2 (M+H+). Preparation of Intermediate I-57 [1-(trifluoromethyl)pyrazol-3-yl]methanol: To a solution of 1-(trifluoromethyl)pyrazole-3-carboxylic acid (321 mg, 1.78 mmol) in THE (10 mL) at 0° C., was added lithium aluminum hydride (2.0M in THF) (2.00 mmol/L, 980 mL, 1.96 mmol). The solution was gradually warmed to rt and stirred for 1 hr. Following this time, the solution was diluted with Et2O, and cooled to 0° C. Upon completion of the cooling, 0.075 mL water, 0.075 mL 15% aqueous NaOH, and 0.225 mL water was added to the solution, which was then warmed to rt and stirred for an additional 15 min. Following the additional 15 min. MgSO4, was added and the solution was stirred another 15 min, then filtered to give title product that was carried onto the next step without further purification. ES/MS: 167.2 (M+H+). 3-(bromomethyl)-1-(trifluoromethyl)pyrazole (I-57): Carbon tetrabromide (0.465 g, 0.00140 mol) was added to a solution of [1-(trifluoromethyl)pyrazol-3-yl]methanol (0.194 g, 1.17 mmol) and (4-diphenylphosphanylphenyl polymer bound) (78.7%, 0.465 g, 0.00140 mol) in DCM (10 mL) at 0° C. The mixture was gradually warmed to rt and stirred overnight. The resulting suspension was filtered, and the filtrate was diluted with DCM and washed with brine. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: Et2O/hexanes) to give I-57. ES/MS: 230.2 (M+H+). Preparation of Intermediate I-58 Methyl 3-(trifluoromethyl)isothiazole-5-carboxylate: To a solution of 3-(trifluoromethyl)isothiazole-5-carboxylic acid (303 mg, 1.54 mmol) in MeOH (3 mL) at 0° C., thionyl chloride (0.125 mL, 1.69 mmol) was added. The resulting solution was gradually warmed to rt and stirred overnight. Following this time, more thionyl chloride (0.125 mL, 1.69 mmol) was added and stirred for 9 hr. Upon completion of this time, the mixture was concentrated and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. 1H NMR (400 MHz, Chloroform-d) δ 8.01 (s, 1H), 4.01 (s, 3H). [3-(trifluoromethyl)isothiazol-5-yl]methanol: To a solution of methyl 3-(trifluoromethyl)isothiazole-5-carboxylate (136 mg, 0.644 mmol) in THE (5 mL) at 0° C., was added diisobutylaluminium hydride (1.0 M in DCM, 1.61 mL, 1.61 mmol). The resulting solution was stirred for 3 hr. Upon completion of this time the mixture was diluted with 5 mL Et2O and cooled to 0° C. Once the mixture was cooled, 0.064 mL water, 0.064 mL 15% NaOH, and 0.161 mL water, was added and the solution was warmed to rt and stirred for 15 min. Following this time, MgSO4was added and the solution was stirred an additional 15 min, then filtered. The crude product was purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 184.2 (M+H+). 5-(bromomethyl)-3-(trifluoromethyl)isothiazole (I-58): To a solution of [3-(trifluoromethyl)isothiazol-5-yl]methanol (85 mg, 0.464 mmol) and (4-diphenylphosphanylphenyl polymer bound) (78.7%, 185 mg, 0.557 mol) in DCM (10 mL) at 0° C., was added carbon tetrabromide (185 mg, 0.557 mmol). The mixture was gradually warmed to rt and stirred overnight. The resulting suspension was filtered, and the filtrate was diluted with DCM and washed with brine. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: Et2O/hexanes) to give I-58. 1H NMR (400 MHz, Chloroform-d) δ 7.49 (d, J=0.8 Hz, 1H), 4.72 (d, J=0.8 Hz, 2H). Preparation of Intermediate I-59 Methyl 4-nitro-3-(spiro[2.2]pentan-2-ylamino)benzoate: A solution of methyl 3-fluoro-4-nitro-benzoate (0.205 g, 1.03 mmol), spiro[2.2]pentan-2-amine; hydrochloride (0.151 g, 1.26 mmol) and N,N-Diisopropylethylamine (0.538 mL, 3.09 mmol) in NMP (3 mL) was heated at 90° C. for 12 hr. Following this time, the mixture was diluted with EtOAc, washed with 5% LiCl, brine and water. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 263.2 (M+H+); 1H NMR (400 MHz, Chloroform-d) δ 8.22 (d, J=8.8 Hz, 1H), 8.06 (s, 1H), 7.79 (d, J=1.8 Hz, 1H), 7.29 (dd, J=8.9, 1.8 Hz, 1H), 3.98 (s, 3H), 3.01 (dd, J=6.3, 3.1 Hz, 1H), 1.54-1.42 (m, 1H), 1.14 (ddd, J=9.0, 5.5, 4.1 Hz, 1H), 1.06-0.98 (m, 2H), 0.95 (td, J=8.5, 4.7 Hz, 2H). Methyl 4-amino-3-(spiro[2.2]pentan-2-ylamino)benzoate (I-59): A solution of methyl 4-nitro-3-(spiro[2.2]pentan-2-ylamino)benzoate (101 mg, 0.4385 mmol) in EtOAc (8 mL) was degassed by cycling the mixture between argon and vacuum 3×. To the mixture was added platinum (1%), vanadium (2%) on carbon (50-70% wetted) and I-59 was carried onto the next step without further purification. ES/MS: 233.2 (M+H+); Preparation of Intermediates I-60 and I-61 Tert-butyl 3-[(6-bromo-2-pyridyl)oxymethyl]pyrazole-1-carboxylate: A suspension of tert-butyl 3-(bromomethyl)pyrazole-1-carboxylate (946 mg, 3.6 mmol), 6-bromopyridin-2-ol (500 mg, 2.9 mmol), and silver carbonate (1694 mg, 6.1 mmol) in CH3CN (15 mL) was heated at 50° C. for 15 hr. Following this time, 335 mg of 6-bromopyridin-2-ol and 5 mL CH3CN were added and the solution was heated at 50° C. for 5 hr. Upon completion of time, 500 mg of 6-bromopyridin-2-ol was added and heating was resumed for 2 hr. Following this time, the mixture was diluted with EtOAc and brine. The mixture was filtered over a plug of Celite. The mixture was partitioned, and the organic phase was washed with brine. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 298, 300 (M+H+). 2-bromo-6-(1H-pyrazol-3-ylmethoxy)pyridine: A solution of tert-butyl 3-[(6-bromo-2-pyridyl)oxymethyl]pyrazole-1-carboxylate (456 mg, 1.3 mmol) and TFA (0.49 mL, 6.4 mmol) in DCM (5 mL) was stirred at rt overnight. Following this time, the solution was diluted with DCM and washed with saturated sodium bicarbonate solution, dried over sodium sulfate, concentrated, and carried onto the next step without further purification. ES/MS: 254, 256 (M+H+). 2-[3-[(6-bromo-2-pyridyl)oxymethyl]pyrazol-1-yl]acetonitrile (I-60) and 2-[5-[(6-bromo-2-pyridyl)oxymethyl]pyrazol-1-yl]acetonitrile (I-61): To a suspension of 2-bromo-6-(1H-pyrazol-3-ylmethoxy)pyridine (0.115 g, 0.453 mmol) and cesium carbonate (0.177 g, 0.543 mmol) in DMF (3 mL), was added 2-chloroacetonitrile (0.0314 mL, 0.498 mmol). The solution was stirred at rt overnight. Following this time, the solution was then warmed to 40° C. for 2 hr., then diluted with EtOAc and washed with 5% LiCl 2× and brine. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: EtOAc/hexanes) to give the I-60 and I-61. 2-[3-[(6-bromo-2-pyridyl)oxymethyl]pyrazol-1-yl]acetonitrile (I-60): ES/MS: 293.2, 295.1 (M+H+); 1H NMR (400 MHz, Chloroform-d) δ 7.55 (d, J=2.4 Hz, 1H), 7.45 (dd, J=8.2, 7.5 Hz, 1H), 7.11 (dd, J=7.5, 0.7 Hz, 1H), 6.76 (dd, J=8.2, 0.7 Hz, 1H), 6.52 (d, J=2.4 Hz, 1H), 5.39 (s, 2H), 5.10 (s, 2H). 2-[5-[(6-bromo-2-pyridyl)oxymethyl]pyrazol-1-yl]acetonitrile (I-61): ES/MS: 293.2, 295.0 (M+H+); 1H NMR (400 MHz, Chloroform-d) δ 7.57 (d, J=1.9 Hz, 1H), 7.50 (dd, J=8.2, 7.5 Hz, 1H), 7.15 (d, J=7.4 Hz, 1H), 6.80 (d, J=8.1 Hz, 1H), 6.48 (d, J=1.9 Hz, 1H), 5.47 (s, 2H), 5.35 (s, 2H). Preparation of Intermediate I-62 Ethyl 1-(1-methylpyrazol-4-yl)pyrazole-3-carboxylate: In a 40 mL glass vial, a mixture of ethyl 1H-pyrazole-3-carboxylate (1000 mg, 7.14 mmol), 4-iodo-1-methyl-pyrazole (1484 mg, 7.14 mmol), cesium carbonate (5812 mg, 17.8 mmol), copper(I) oxide (60.0 mg, 0.419 mmol), and salicylaldoxime (120 mg, 0.875 mmol) in DMF (20 mL) was heated at 110° C. for 48 hr. Following this time, the mixture was diluted with EtOAc and washed with 5% LiCl, saturated sodium bicarbonate, and brine. The organic extract was dried over sodium sulfate and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 221.2 (M+H+). [1-(1-methylpyrazol-4-yl)pyrazol-3-yl]methanol: To a solution of ethyl 1-(1-methylpyrazol-4-yl)pyrazole-3-carboxylate (287 mg, 1.30 mmol) in THE (6 mL) at 0° C., diisobutylaluminium hydride (1.0 M in DCM, 3.26 mL, 3.26 mmol) was added. The resulting solution was stirred for 1 hr. while gradually warming to rt. Following this time, the solution was diluted with Et2O and cooled to 0° C. Upon completion of the cooling, 0.130 mL water, 0.130 mL 15% aqueous NaOH, and 0.326 mL water were added to the solution and then the resulting solution was warmed to rt. Following the warming, the solution was stirred for 15 min, then MgSO4was added and the solution was stirred for an additional 15 min, then filtered. The filtrate was concentrated and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 179.2 (M+H+). 3-(bromomethyl)-1-(1-methylpyrazol-4-yl)pyrazole (I-62): To a solution of [1-(1-methylpyrazol-4-yl)pyrazol-3-yl]methanol (136 mg, 0.762 mmol) and (4-diphenylphosphanylphenyl polymer bound) (78.7%, 303 mg, 0.914 mmol) in DCM (10 mL) at 0° C., was added carbon tetrabromide (303 mg, 0.914 mmol). The mixture was gradually warmed to rt and stirred overnight. The resulting suspension was filtered, and the filtrate was diluted with DCM and washed with brine. The organic extract was dried over sodium sulfate, concentrated, and purified by flash chromatography (eluent: Et2O/hexanes) to give I-62. ES/MS: 241.2, 243.2 (M+H+); 1H NMR (400 MHz, Chloroform-d) δ 7.71 (s, 1H), 7.69 (d, J=0.8 Hz, 1H), 7.61 (d, J=2.4 Hz, 1H), 6.47 (d, J=2.4 Hz, 1H), 4.56 (s, 2H), 3.96 (s, 3H). The following intermediate was prepared in a manner as described for intermediate I-62 Preparation of Intermediate I-63 2-(bromomethyl)thiazole-5-carbonitrile (I-63): To a solution of 2-methylthiazole-5-carbonitrile (200 mg, 1.61 mmol) in CCl4(8 mL), was added N-Bromosuccinimide (375 mg, 2.11 mmol), followed by benzoyl peroxide (42.0 mg, 0.173 mmol). The solution was heated at 90° C. for 9 hrs. Following this time, the mixture was cooled to rt and 5 mL hexanes was added. The suspension was filtered, and the filtrate was concentrated, and purified by flash chromatography (eluent: EtOAc/hexanes) to give I-63. ES/MS: 203.0, 205.2 (M+H+); 1H NMR (400 MHz, Chloroform-d) δ 8.23 (s, 1H), 4.74 (s, 2H). The following intermediates were prepared in a manner as described for intermediate I-63 Preparation of Intermediate I-64 3-(bromomethyl)-5-methoxy-1-methyl-1H-pyrazole (I-64): To a stirring solution of (5-methoxy-1-methyl-1H-pyrazol-3-yl)methanol (1.42 g, 10 mmol, 1.0 eq) in DCM (15 mL), was added at rt, CBr4 (7.2 g, 21 mmol, 2.0 eq) and triphenylphosphine (5.7 g, 21 mmol, 2.0 eq). The mixture was stirred for an additional 16 h. Upon completion, the mixture was diluted with water (150 mL), extracted with DCM (3×150 mL), washed with brine (50 mL), dried over Na2SO4and concentrated to get the crude product which was purified by column chromatography (0 to 2% MeOH-DCM) to afford 3-(bromomethyl)-5-methoxy-1-methyl-1H-pyrazole (I-64). The following intermediates were prepared in a manner as describe for intermediate I-64 Preparation of Intermediate I-65 (5,5-difluoro-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-2-yl)methanol (I-65): 5,5-difluoro-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole-2-carboxylic acid (105 mg, 0.56 mmol) was dissolved in THE (3 mL) and stirred at 0° C. for 5 min. Next, 1M Borane (3.4 mL) solution was added dropwise to the mixture at 0° C. over a period of 30 min. The ice bath was removed and stirring continued at rt for 7 hours. Following this time, the mixture was cooled in an ice bath and treated with 3M HCl (5 mL). The solution was heated for 1 h at 50° C. Upon completion of the time, the solution was washed with EtOAc (2×) and the aqueous layer was cooled in an ice bath and neutralized with 3M NaOH. The solution was extracted with EtOAc (3×), the combined organic layers were washed with brine, dried (Na2SO4) and concentrated in vacuo to obtain (5,5-difluoro-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-2-yl)methanol (I-65). Preparation of Intermediate I-66 Methyl 2-[[4-[6-[(5-bromopyrimidin-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-66): Methyl 2-[[4-[6-[(5-bromopyrimidin-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 to provide I-66. ES/MS: 637.4 (M+H+). Preparation of Intermediate I-67 Methyl 2-[[4-[6-[(5-chloropyrazin-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-67): Methyl 2-[[4-[6-[(5-chloropyrazin-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 to provide I-67. ES/MS: 593 (M+H+). Preparation of Intermediate I-68 Methyl 3-bromo-5-((4,4-dimethyltetrahydrofuran-3-yl) amino-4-nitrobenzoate: Methyl 3-bromo-5-fluoro-4-nitrobenzoate (0.5 g, 1.8 mmol) was dissolved in a 100 mL round bottom flask containing DMF (10 mL). Next, 4,4-dimethyltetrahydrofuran-3-amine hydrochloride (0.46 g, 3 mmol) and N,N-diisopropylethylamine (0.63 mL, 3.6 mmol) were added to the solution. The mixture was stirred at 50° C. overnight. Afterward, the mixture was concentrated to remove most of the THF, and the crude material was dissolved in EtOAc (40 mL). The organics were washed with 50% NH4Cl (2×10 mL) and with brine (1×50 mL). The organics were subsequently dried over MgSO4, filtered, and concentrated under reduced pressure. The crude material was carried forward without further purification: ES/MS: 374.2 (M+H+) Methyl 4-amino-3-bromo-5-[(4,4-dimethyltetrahydrofuran-3-yl)amino]benzoate (I-68): To a 100 mL round bottom flask, methyl 3-bromo-5-((4,4-dimethyltetrahydrofuran-3-yl) amino-4-nitrobenzoate (0.58 g, 1.6 mmol), Iron (0.43 g, 7.8 mmol), and Acetic acid (10 mL) were added. The mixture was stirred and heated at 100° C. for 1 h. Following this time, the mixture was filtered through Celite to remove the catalyst. The filtrate was concentrated under reduced pressure to give I-68 which was used without further purification: ES/MS: 344.2 (M+H+). Preparation of Intermediate I-69 Methyl 3-bromo-4-[[2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetyl]amino]-5-[(4,4-dimethyltetrahydrofuran-3-yl)amino]benzoate: To a solution of I-68 (200 mg, 0.58 mmol) and I-7 (180 mg, 0.45 mmol) in MeCN (5 mL) and cooled to 0° C. was added 1-methylimidazole (239 mg, 0.23 mL, 2.9 mmol) followed by N,N,N′,N′-Tetramethylchloroformamidinium Hexafluorophosphate (204 mg, 0.73 mmol). The mixture was warmed to rt and stirred for 30 min. Upon completion of the 30 min, the crude mixture was concentrated in vacuo, then partitioned between water and EtOAc. The organic layer was isolated and washed with an additional portion of water and then brine. The isolated organic layer was dried over sodium sulfate, isolated by vacuum filtration, concentrated in vacuo, and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide the desired product. ES/MS: 724.4 [M+H]+. Methyl 7-bromo-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (I-69): A solution of methyl 3-bromo-4-[[2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetyl]amino]-5-[(4,4-dimethyltetrahydrofuran-3-yl)amino]benzoate (100 mg, 0.14 mmol) in acetic acid (2 mL) was heated to 80° C. for 5 days. The mixture was concentrated and partitioned between EtOAc and saturated aqueous sodium bicarbonate. The organic layer was isolated and dried over sodium sulfate, isolated by vacuum filtration, concentrated in vacuo, and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide I-69. ES/MS: 706.5 [M+H]+. Preparation of Intermediate I-70 Methyl 2-(5-bromo-3-fluoropyridin-2-yl)acetate: Tert-butyl methyl malonate was added dropwise to a suspension of NaH (60% in mineral oil, 1.3 g, 34 mmol) in DMF (20 mL) at 5° C., and the suspension was stirred for 5 min. Next, 5-bromo-2,3-difluoropyridine was added dropwise, and the resulting suspension was warmed to 60° C. and stirred at that temperature overnight. Following this time, NH4Cl was added, and the mixture was extracted with ether. The organic phase was rinsed with brine, and concentrated. The residue was redissolved in DCM (10 mL). Next, TFA (10 mL) was added, and the resulting solution was warmed to 40° C. and stirred for 5 hours. Upon completion of time, the mixture was concentrated and purified by flash chromatography (EtOAc/hexanes) to give title product: ES/MS: 248.2 (M+H+). 5-bromo-3-fluoro-2-(2-methoxy-2-oxoethyl)pyridine 1-oxide: MCPBA (3.51 g, 16 mmol) was added to a solution of methyl 2-(5-bromo-3-fluoropyridin-2-yl)acetate (2.68 g, 11 mmol) in DCM (30 mL) at 0° C., and the resulting solution was allowed to warm to rt and stirred overnight. Next, the mixture was diluted with hexanes (20 mL) and filtered. The filtrate was concentrated and purified by flash chromatography (EtOAc/hexanes) to give title product: ES/MS: 265.2 (M+H+). Methyl 2-(6-amino-5-bromo-3-fluoropyridin-2-yl)acetate: P-toluenesulfonic anhydride (1.5 g) was added over the course of 1.5 hours to a solution of 5-bromo-3-fluoro-2-(2-methoxy-2-oxoethyl)pyridine 1-oxide (1.24 g, 4.7 mmol), saccharin (6.3 g, 34 mmol), and DIPEA (6.5 mL, 38 mmol) in chloroform (5 mL). The resulting solution was stirred at rt for 2 hours. Following this time, an additional amount of p-toluenesulfonic anhydride (1.3 g) was added and stirred for 1 hr. After the one hour, a further additional amount of p-toluenesulfonic anhydride (2.0 g) was added and stirred over the weekend. The reaction was quenched with Na2CO3and filtered. The phases were separated, and the aqueous phase was extracted with DCM. The combined organics were washed with 10% citric acid, dried, filtered, concentrated, and purified by flash chromatography (EtOAc/hexanes). The resulting product was suspended in 2M H2SO4(4 mL), and stirred at 90° C. for 18 hours, warmed to reflux, and stirred for 24 hours. The mixture was filtered, and filtrate washed with CHCl3. The aqueous phase was basified with NaOH to pH˜ 7 and filtered. Filtrate was acidified to pH˜5 and extracted with CHCl3(3×). Organics were dried and concentrated to give title product: ES/MS: 249.0 (M+H+).1H NMR (400 MHz, Chloroform-d) δ 7.59 (d, J=7.7 Hz, 1H), 5.05 (s, 2H), 3.80 (d, J=2.2 Hz, 2H).19F NMR (376 MHz, Chloroform-d) 6-137.76 (d, J=7.5 Hz). 2-(6-amino-5-bromo-3-fluoropyridin-2-yl)acetic acid: The title intermediate was prepared in a manner as described for intermediate I-7 (step 2) substituting methyl 2-(6-amino-5-bromo-3-fluoropyridin-2-yl)acetate for methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetate. Tert-butyl 2-((6-amino-5-bromo-3-fluoropyridin-2-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate: The title compound was prepared in a manner as described for Intermediate I-2, using 2-(6-amino-5-bromo-3-fluoropyridin-2-yl)acetic acid in place of 2-(4-bromo-2-fluoro-phenyl)acetic acid, and tert-butyl 4-amino-3-((2-methoxyethyl)amino)benzoate in place of methyl 4-amino-3-(2-methoxyethylamino)benzoate. ES/MS: 481.1 (M+H+).1H NMR (400 MHz, Chloroform-d) δ 8.07 (d, J=1.5 Hz, 1H), 7.94 (d, J=8.6 Hz, 1H), 7.78 (d, J=8.5 Hz, 1H), 7.51 (d, J=7.8 Hz, 1H), 4.79 (s, 2H), 4.57-4.39 (m, 4H), 3.70 (t, J=5.3 Hz, 2H), 3.30 (s, 3H), 1.65 (s, 9H). Tert-butyl 2-((2′-amino-6-((4-cyano-2-fluorobenzyl)oxy)-5′-fluoro-[2,3′-bipyridin]-6′-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-70): I-70 was prepared in a manner as described for Intermediate I-7, using dichlorobis(di-tert-butylphenylphosphine)palladium(II) in place of Pd(dppf)Cl2 in step 1, and tert-butyl 2-((6-amino-5-bromo-3-fluoropyridin-2-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate in place of methyl 2-(4-bromo-2,5-difluorophenyl)acetate. ES/MS: 627.5 (M+H+). Preparation of Intermediate I-71 Tert-butyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-71): The title compound was prepared in a manner as described for Intermediate I-2, using Intermediate I-6 and Intermediate I-7. ES/MS: 629.5 (M+H+). Preparation of Intermediate I-72 Methyl 4-amino-3-iodo-5-((2-methoxyethyl)amino)benzoate: The title compound was prepared in a manner as described for intermediate I-1. Reduction was executed by stirring Iron (603 mg, 10.8 mmol), acetic acid (12.0 mL, 1.8 mmol), and crude methyl 3-iodo-5-(2-methoxyethylamino)-4-nitro-benzoate (821 mg, 2.16 mmol) in methanol (5.0 mL) at reflux for 1 hour. The mixture was diluted with DCM, filtered, and organics were dried, filtered, concentrated, and carried on crude. Methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-iodo-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate: The title compound was prepared in a manner as described for Intermediate I-13, using methyl 4-amino-3-iodo-5-((2-methoxyethyl)amino)benzoate and Intermediate I-7. 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-iodo-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (I-72): In an 8 mL glass vial, a solution of methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-iodo-3-(2-methoxyethyl)benzimidazole-5-carboxylate (105 mg, 0.15 mmol) and lithium hydroxide, monohydrate (25 mg, 0.59 mmol) in THF/water (2:1, 3 mL) was heated at 70° C. until completion (15 min). Following completion of the mixture, trifluoroacetic anhydride (0.3 mL) was added and the solution purified directly by RP-HPLC (eluent: MeCN/H2O) to give I-72. ES/MS: 699.1 Preparation of Intermediate I-73 Tert-butyl (R)-2-(4-(6-((5-bromothiazol-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylate (I-73): tert-butyl (R)-2-(4-(6-((5-bromothiazol-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-21 substituting I-26 for I-4. ES/MS: 686.8 (M+H+). Preparation of Intermediate I-74 (1-methylthieno[2,3-c]pyrazol-5-yl)methanol: To a solution of 1-methylthieno[2,3-c]pyrazole-5-carboxylic acid (140 mg, 0.77 mmol) in THF (6 mL) was added CDI (249 mg, 1.54 mmol) and the resultant slurry stirred for 2 hours at ambient temperature. Following this time, NaBH4(145 mg, 3.84 mmol) was added portion wise and the mixture stirred overnight. Upon completion MeOH was added (2 mL) and the crude mixture concentrated directly. The crude residue purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide desired product. ES/MS: 169.1 (M+H+). 5-[(6-bromo-2-pyridyl)oxymethyl]-1-methyl-thieno[2,3-c]pyrazole (I-74): To a solution of 2-bromo-6-fluoro-pyridine (122 mg, 0.69 mmol) in acetonitrile (2 mL) was added (1-methylthieno[2,3-c]pyrazol-5-yl)methanol (106 mg, 0.63 mmol) and cesium carbonate (411 mg, 1.26 mmol) and the resultant mixture stirred for 2 hours at 80° C. Upon completion the mixture was diluted with EtOAc (25 mL), washed with water (5 mL) and brine (5 mL). The organic layer was dried over MgSO4, filtered, concentrated and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide I-74. ES/MS: 326.1 (M+H+). Preparation of Intermediate I-75 Methyl 2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-75): Methyl 2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-19 substituting I-2 for I-96. ES/MS: 623.3 (M+H+). Preparation of Intermediate I-76 Methyl 2-(4-(6-((6-chloro-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-76): Methyl 2-(4-(6-((6-chloro-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-23 substituting I-25 for I-4. ES/MS: 638.0 (M+H+). Preparation of Intermediate I-77 2-bromo-6-(1H-pyrazol-3-ylmethoxy)pyridine (I-77): To a solution of 6-bromopyridin-2-ol (180 mg, 1.0 mmol) in acetonitrile (4 mL) was added cesium carbonate (482 mg, 1.5 mmol) and tert-butyl 3-(bromomethyl)pyrazole-1-carboxylate (378 mg, 1.4 mmol) after which the mixture was heated to 65° C. for 30 minutes. Upon completion the mixture was filtered through celite, concentrated and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide desired product. The so obtained tert-butyl 3-[(6-bromo-2-pyridyl)oxymethyl]pyrazole-1-carboxylate (343 mg, 0.97 mmol) was dissolved in DCM (4.4 mL) and TFA (1.1 mL) and stirred at ambient temperature for 2 hours. Upon completion, the mixture was diluted with EtOAc (25 mL) washed with saturated aqueous NaHCO3until gas evolution ceased, dried over MgSO4, filtered and concentrated to give I-77 which was used without further purification. ES/MS: 254.2, 256.2 (M+H+). Preparation of Intermediate I-78 Methyl (S)-2-(4-(6-((5-bromo-1-methyl-1H-pyrazol-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-78): Methyl (S)-2-(4-(6-((5-bromo-1-methyl-1H-pyrazol-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 5-bromo-3-(bromomethyl)-1-methyl-1H-pyrazole for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 638.0, 640.0 (M+H+). Preparation of Intermediate I-79 2-[5-(hydroxymethyl)-2-methyl-pyrazol-3-yl]acetonitrile: To a solution of methyl 5-(bromomethyl)-1-methyl-pyrazole-3-carboxylate (750 mg, 3.22 mmol) in DMF (9.5 mL) and water (1.2 mL), was added sodium cyanide (241 mg, 4.83 mmol) and the resultant mixture stirred at rt for 3.5 hours. Upon completion the mixture was diluted with EtOAc (50 mL), washed with water (10 mL) and brine (10 mL). The organic layer was dried over MgSO4, filtered, concentrated and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide desired product. The so obtained methyl 5-(cyanomethyl)-1-methyl-pyrazole-3-carboxylate (328 mg, 1.83 mmol), was dissolved in THF (10 mL) and lithium borohydride (2.0M in THF, 1.83 mL, 3.66 mmol) was added at 0° C. The mixture was allowed to warm to rt and stir for 6 hr. at which point additional lithium borohydride (2.0M in THF, 1.83 mL, 3.66 mmol) was added and the mixture stirred for 2 hr. Upon completion the reaction was quenched by the addition of water (5 mL), diluted with EtOAc (50 mL) and the layers separated. The organic layer was dried over MgSO4, filtered, concentrated and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide desired product. 2-[5-(bromomethyl)-2-methyl-pyrazol-3-yl]acetonitrile (I-79): 2-[5-(Hydroxymethyl)-2-methyl-pyrazol-3-yl]acetonitrile (100 mg, 0.662 mmol) was taken up in dichloromethane (2.65 mL) and triphenylphosphine (0.208 g, 0.794 mmol) was added followed by the addition of carbontetrabromide (0.263 g, 0.794 mmol). The mixture was left to stir at rt for 5 minutes at which point the reaction was quenched by the addition of water (5 mL), diluted with EtOAc (25 mL) and the layers separated. The organic layer was dried over MgSO4, filtered, concentrated and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide I-79. ES/MS: 214.0, 216.0 (M+H+). Preparation of Intermediates I-80 and I-81 (Method 1) Methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (I-80, 1-81): Methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate as a mixture of 2 stereoisomers were separated by chiral SFC (SFC IB column with EtOH cosolvent) to give two distinct stereoisomers. Methyl (S)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate, isomer 1 (I-80): Isolated as the earlier eluting of two isomers by chiral SFC (4.6×100 mm 5 μm IB column, 10% EtOH in CO2). ES/MS: 265.2 (M+H+). Methyl (R)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate, isomer 2 (I-81): Isolated as the later-eluting of two isomers by chiral SFC (4.6×100 mm 5 μm IB column, 10% EtOH in CO2). ES/MS: 265.2 (M+H+). Preparation of Intermediate I-80 (Method 2) Methyl 4-amino-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate isomer 2 (I-80): A solution of methyl 3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-4-nitro-benzoate (Intermediate I-100, 17.8 g, 60.5 mmol) in EtOAc (380 mL) was degassed with argon then vacuum 3×. Palladium on carbon (10.0%, 6.07 g, 5.70 mmol) was added. The mixture was degassed with argon then vacuum 3 times and stirred at rt under an atmosphere of hydrogen until completion. The suspension was filtered over a Celite plug and rinsed with EtOAc. The mixture was concentrated to yield methyl 4-amino-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate, which was carried forward to subsequent steps without further purification. ES/MS: 265.0 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.49 (dd, J=8.0, 1.8 Hz, 1H), 7.35 (d, J=1.8 Hz, 1H), 6.73 (d, J=8.1 Hz, 1H), 4.36 (dd, J=9.1, 6.4 Hz, 1H), 3.89 (s, 3H), 3.76 (t, J=5.9 Hz, 1H), 3.72-3.63 (m, 2H), 3.63-3.59 (m, 1H), 1.22 (s, 3H), 1.15 (s, 3H). Preparation of Intermediates I-82 and I-83 (Method 1) Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-82, I-83): I-82 and I-83 were prepared separately in a manner as described for Intermediate I-8 substituting I-80 (for Intermediate I-82) and I-81 (for Intermediate I-83) for I-4. Methyl (S)-2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate isomer 1 (Intermediate I-82): ES/MS: 479.0, 481.0 (M+H+). Methyl (R)-2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate isomer 2 (Intermediate I-83): ES/MS: 479.0, 481.0 (M+H+). Preparation of Intermediate I-82 (Method 2) Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-82): To a solution of 2-(4-bromo-2,5-difluorophenyl)acetic acid (3301 mg, 13.2 mmol), methyl 4-amino-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate (Intermediate I-80, 3.16 g, 12.0 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (6360 mg, 16.7 mmol) in DMF (20 mL) and CH3CN (20 mL), was added N,N-Diisopropylethylamine (10.2 mL, 58.4 mmol). The solution was stirred at rt overnight. Then to the mixture was added 0.2 eq of 2-(4-bromo-2,5-difluoro-phenyl)acetic acid (600 mg, 2.39 mmol) and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (930 mg, 2.39 mmol) and continued stirring until complete conversion to product. The crude mixture was diluted with 200 mL EtOAc and washed with saturated NH4Cl (200 mL), 5% LiCl (100 mL), saturated NaHCO3(100 mL), and brine (100 mL). The organic extract was dried over sodium sulfate to give methyl 4-[[2-(4-bromo-2,5-difluoro-phenyl)acetyl]amino]-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate. A solution of methyl 4-[[2-(4-bromo-2,5-difluoro-phenyl)acetyl]amino]-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate (5.96 g, 12.0 mmol) in AcOH (60 mL) was heated to 180° C. for 90 min in a microwave reactor. The mixture was concentrated, then diluted with EtOAc and washed with saturated NaHCO3, and brine. The mixture was dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-82). ES/MS: 478.6, 480.6 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.55 (s, 1H), 8.02 (dd, J=8.5, 1.5 Hz, 1H), 7.78 (d, J=8.5 Hz, 1H), 7.37 (dd, J=8.7, 5.6 Hz, 1H), 7.12 (dd, J=8.4, 6.3 Hz, 1H), 4.59 (d, J=10.5 Hz, 2H), 4.40 (dd, J=11.1, 7.3 Hz, 1H), 4.31 (s, 2H), 3.97 (s, 4H), 3.80 (d, J=8.8 Hz, 1H), 1.35 (s, 3H), 0.67 (s, 3H). Preparation of Intermediate I-84 2-[(6-bromo-2-pyridyl)oxymethyl]thiazole-5-carbonitrile (I-84): To a solution of 6-bromopyridin-2-ol (500 mg, 2.9 mmol) in acetonitrile (10 mL) was added 2-(bromomethyl)thiazole-5-carbonitrile (I-63) (584 mg, 2.9 mmol) and cesium carbonate (1.34 g, 4.1 mmol) and the resultant mixture stirred at 65° C. for 1 hour. Upon completion the mixture was filtered through celite, concentrated and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide I-84. ES/MS: 296.0, 298.0 (M+H+). Preparation of Intermediate I-85 Methyl 2-[[4-[6-[(6-chloro-4-methoxy-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-85): Methyl 2-[[4-[6-[(6-chloro-4-methoxy-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-23 substituting 5-(bromomethyl)-2-chloro-4-methoxypyridine for 5-(bromomethyl)-2-chloro-4-fluoropyridine. ES/MS: 621.2 (M+H+). Preparation of Intermediate I-86 5-(bromomethyl)-4-methoxy-N-methylpicolinamide (I-86): 5-(bromomethyl)-4-methoxy-N-methylpicolinamide was prepared in a manner as described for Intermediate I-53 substituting methylamine HCl for 1-aminocyclopropane-1-carbonitrile. ES/MS: 259.2, 261.2 (M+H+). Preparation of Intermediate I-87 5-(bromomethyl)-4-chloro-N-methylpicolinamide (I-87): 5-(bromomethyl)-4-chloro-N-methylpicolinamide was prepared in a manner as described for Intermediate I-53 substituting methylamine HCl for 1-aminocyclopropane-1-carbonitrile and 4-chloro-5-methylpicolinic acid for 4-methoxy-5-methylpicolinic acid. ES/MS: 263.0, 265.0 (M+H+). Preparation of Intermediate I-88 Tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-88): To a solution of tert-butyl 2-[[4-[6-[(4-bromo-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (300 mg, 0.44 mmol) in 1,4-dioxane (20 mL) was added Bis(pinacolato)diboron (179 mg, 0.70 mmol), Pd(dppf)Cl2(33 mg, 0.044 mmol), and potassium propionate (148 mg, 1.3 mmol). Argon was bubbled through the mixture for 1 minute after which the reaction vessel was sealed and heated to 110° C. for 45 minutes in a microwave reactor. Upon completion the mixture was concentrated directly and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide I-88. ES/MS: 730.8 (M+H+). Preparation of Intermediate I-89 (3S,4R)-4-(5-methoxycarbonyl-2-nitro-anilino)tetrahydrofuran-3-carboxylic acid: To a solution of methyl 3-fluoro-4-nitro-benzoate (700 mg, 3.52 mmol) and (3S,4R)-4-aminotetrahydrofuran-3-carboxylic acid; hydrochloride (648 mg, 17.6 mmol) in DMF (2.5 mL) and THE (5 mL) was added diisopropylethylamine (3.1 mL, 17.6 mmol) and the resultant solution heated to 70° C. for 3 days. Upon completion, the mixture was diluted with EtOAc (50 mL), washed with water (10 mL), brine (10 mL), dried over MgSO4, filtered, concentrated to give the desired product which was used without further purification. ES/MS: 311.2 (M+H+). Methyl 3-[[(3R,4S)-4-(methylcarbamoyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate: (3S,4R)-4-(5-methoxycarbonyl-2-nitro-anilino)tetrahydrofuran-3-carboxylic acid (370 mg, 0.00119 mol), 1-hydroxybenzotriazole hydrate (0.192 g, 0.00125 mol), and methylamine (2000 mmol/L in THF, 1.19 mL, 0.00239 mol) were taken up in tetrahydrofuran (8.00 mL) and triethylamine (0.151 g, 0.00149 mol) was added followed by 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (0.251 g, 0.00131 mol). The mixture was stirred at rt overnight. Upon completion the mixture was diluted with EtOAc (50 mL), washed with water (10 mL), brine (10 mL), dried over MgSO4, filtered, and concentrated to give the desired product which was used without further purification. ES/MS: 324.2 (M+H+). Methyl 4-amino-3-[[(3R,4S)-4-(methylcarbamoyl)tetrahydrofuran-3-yl]amino]benzoate (I-89): To a solution of methyl 3-[[(3R,4S)-4-(methylcarbamoyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate (168 mg, 0.52 mmol) in EtOH (2 mL) and THE (1 mL) was added palladium on carbon (10%, 111 mg, 0.10 mmol). The mixture was then purged with H2gas and stirred under 1 atm of H2for 1 hr. Upon completion the mixture was filtered through celite, concentrated and the crude residue, I-89, was used without further purification. ES/MS: 294.2 (M+H+). Preparation of Intermediate I-90 Methyl 4-amino-3-(((3R,4S)-4-(dimethylcarbamoyl)tetrahydrofuran-3-yl)amino)benzoate (I-90): Methyl 4-amino-3-(((3R,4S)-4-(dimethylcarbamoyl)tetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-89 substituting dimethylamine for methylamine. ES/MS: 308.2 (M+H+). Preparation of Intermediate I-91 4-bromo-1-[2-[2-(2-methoxyethoxy)ethoxy]ethyl]pyrazole: To a solution of 4-bromo-1H-pyrazole (100 mg, 0.68 mmol) in 2-Me tetrahydrofuran (2 mL) was added Potassium Bis(trimethylsilyl)amide (204 mg, 1.0 mmol) and 1-[2-(2-bromoethoxy)ethoxy]-2-methoxy-ethane (309 mg, 1.4 mmol) and heated to 50° C. for 2 hours. Upon completion, the mixture was diluted with EtOAc (25 mL), washed with water (5 mL), brine (5 mL), dried over MgSO4, filtered and concentrated to give the desired product which was used without further purification. ES/MS: 293.3 (M+H+). 1-[2-[2-(2-methoxyethoxy)ethoxy]ethyl]-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (I-91): 4-bromo-1-[2-[2-(2-methoxyethoxy)ethoxy]ethyl]pyrazole was converted to 1-[2-[2-(2-methoxyethoxy)ethoxy]ethyl]-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (I-91) in a manner as described for intermediate I-88. ES/MS: 341.1 (M+H+). Preparation of Intermediate I-92 Tert-butyl 2-(4-(6-((6-chloropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-(I-92): Tert-butyl 2-(4-(6-((6-chloropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6- was prepared in manner as described for Intermediate I-18 substituting 5-(bromomethyl)-2-chloropyridine for 4-bromo-1-(bromomethyl)-2-fluoro-benzene. ES/MS: 621.2 (M+H+). Preparation of Intermediate I-93 Methyl (S)-2-(4-(6-((6-bromo-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-93): Methyl (S)-2-(4-(6-((6-bromo-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in manner as described for Intermediate I-19 substituting 2-bromo-5-(bromomethyl)-4-fluoropyridine for 4-bromo-1-(bromomethyl)-2-fluoro-benzene ES/MS: 654.0, 656.0 (M+H+). Preparation of Intermediate I-94 4-[(4-bromopyrimidin-2-yl)oxymethyl]-3-fluoro-benzonitrile (I-94): 4-bromo-2-chloro-pyrimidine (1.7 g, 8.8 mmol), 3-fluoro-4-(hydroxymethyl)benzonitrile (1.46 g, 9.7 mmol), potassium hydroxide (542 mg, 9.7 mmol), 18-crown-6 (116 mg, 0.44 mmol), and toluene (20 mL) were combined and heated to 110° C. for 2 hours. Upon completion the mixture was diluted with EtOAc (100 mL) washed with water (25 mL), washed with brine (25 mL), dried over MgSO4, filtered, concentrated and purified by silica gel chromatography (eluent: EtOAc/hexanes) to give I-94. ES/MS: 309.2 (M+H+). Preparation of Intermediate I-95 Methyl 2-(4-(6-chloropyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-95): Methyl (S)-2-(4-(6-chloropyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-12 substituting I-1 for I-6. ES/MS: 472.8 (M+H+). Preparation of Intermediate I-96 Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-96). Methyl 2-(2,5-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-2 substituting 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluorophenyl)acetic acid. ES/MS: 439.8 (M+H+). Preparation of Intermediate I-97 (5-chloro-1-methyl-pyrazol-3-yl)methanol: 5-chloro-1-methyl-pyrazole-3-carboxylic acid (700 mg, 4.36 mmol) was taken up in THE (20.0 mL) and 1,1′-Carbonyldiimidazole (1.41 g, 8.72 mmol) was added. The mixture was stirred at rt for 2 hours. Upon completion, the mixture was cooled to 0° C. and a solution of sodium borohydride (0.825 g, 21.8 mmol) in water (3.30 mL) was added slowly, after which the mixture was allowed to warm to rt over 40 minutes. Upon completion methanol (5 mL) was added, the mixture was concentrated directly and purified by flash chromatography (Eluent: EtOAc/hexane) to give the desired product. 3-(bromomethyl)-5-chloro-1-methyl-pyrazole (I-97): (5-chloro-1-methyl-pyrazol-3-yl)methanol (550 mg, 3.75 mmol) was taken up in DCM (25.0 mL) and (4-diphenylphosphanylphenyl) polymer bound (78.7%, 1.50 g, 0.00450 mol) was added. The mixture was cooled to 0° C. and carbon tetrabromide (1.49 g, 0.00450 mol) was added as a single portion. The mixture was stirred for 16 hr. at rt. Upon completion, the mixture was filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give I-97. ES/MS: 209.0, 211.0 (M+H+). The following intermediates were prepared in a manner as described for intermediate I-97 Preparation of Intermediate I-98 5-(hydroxymethyl)-N-methyl-pyridine-2-carboxamide: 5-(hydroxymethyl)pyridine-2-carboxylic acid (400 mg, 2.61 mmol), methanamine hydrochloride (194 mg, 2.87 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (1192 mg, 3.14 mmol) were taken up in DMF (5.00 mL). N,N-Diisopropylethylamine (2.27 mL, 13.1 mmol) was added after which the mixture was stirred for 30 min. Upon completion the mixture was diluted with EtOAc (50 mL) washed with water (10 mL), washed with brine (10 mL), dried over MgSO4, filtered, concentrated and purified by silica gel chromatography (eluent: MeOH/EtOAc/hexanes) to give desired product. ES/MS: 167.2 (M+H+). 5-(bromomethyl)-N-methyl-pyridine-2-carboxamide (I-98): 5-(hydroxymethyl)-N-methyl-pyridine-2-carboxamide (106 mg, 0.638 mmol) and triphenylphosphine (0.167 g, 0.638 mmol) were taken up in dichloromethane (2.60 mL) and carbon tetrabromide (0.212 g, 0.638 mmol), was added. The mixture was stirred for 15 min. Upon completion the mixture was filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give I-98. ES/MS: 229.0 (M+H+). The following intermediate was prepared in a manner as described for intermediate I-98 Preparation of Intermediate I-99 (R)-4,4-dimethyl-2-oxotetrahydrofuran-3-yl trifluoromethanesulfonate (I-99-1): A round-bottom flask was charged with (D)-(−)-pantolactone (4.20 g, 32.3 mmol, 1.0 equivalent). Anhydrous dichloromethane (20 mL) and pyridine (3.39 mL, 42.0 mmol, 1.30 equivalent) were added, and the resulting solution was cooled to −78° C. A solution of trifluoromethanesulfonic anhydride (5.96 mL, 35.4 mmol, 1.10 equivalent) in dichloromethane (100 mL) was added slowly to the mixture via an addition funnel while stirring at −78° C. Following the addition, the mixture was maintained with stirring at −78° C. for 30 min before the cooling bath was removed. The mixture was maintained with stirring at rt for an additional 3 hrs. The solvent was removed in vacuo, and the residue was dissolved in ethyl ether (200 mL) and washed with 10% aqueous sodium bicarbonate (100 mL), and brine (100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to afford (R)-4,4-dimethyl-2-oxotetrahydrofuran-3-yl trifluoromethanesulfonate. 1H NMR (500 MHz, CDCl3) δ=5.10 (s, 1H), 4.13 (d, J=9.5 Hz, 1H), 4.07 (d, J=9.5 Hz, 1H), 1.28 (s, 3H), 1.18 (s, 3H). (S)-3-azido-4,4-dimethyldihydrofuran-2(3H)-one (I-99-2): A round-bottom flask was charged with tetrabutylammonium azide (10.5 g, 36.8 mmol, 1.15 equivalent). Anhydrous toluene (50 mL) was added, and the resulting solution was cooled to 0° C. A solution of (R)-4,4-dimethyl-2-oxotetrahydrofuran-3-yl trifluoromethanesulfonate prepared as above (8.40 g, 32 mmol, 1.0 equivalent) in toluene (50 mL) was added slowly via an addition funnel at 0° C. The mixture was maintained at 0° C. for 30 min before the cooling bath was removed and the mixture was stirred at rt for 4 hrs. The mixture was diluted with ethyl ether (200 mL) and washed with 10% aqueous sodium bicarbonate (200 mL), and brine (100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to afford 3-azido-4,4-dimethyldihydrofuran-2(3H)-one. The crude material was carried forward immediately to the next step without purification. 3-azido-4,4-dimethyltetrahydrofuran-2-ol (I-99-3): 3-azido-4,4-dimethyldihydrofuran-2(3H)-one (4.16 g, 26.8 mmol) was taken in dichloromethane (40 mL), cooled to −78° C. then diisobutylaluminum hydride (1.0 M in toluene) (32.2 mL, 32.2 mmol, 1.2 equivalent) was added slowly followed at same temperature. The mixture was stirred at −78° C. for 2 hrs until no starting material remained. The reaction was quenched by adding saturated solution of potassium sodium tartarate (100 mL). The mixture was extracted with dichloromethane (3×50 mL). The organic extract was dried with anhydrous Na2SO4and concentrated under reduced pressure. The crude compound was purified by column chromatography (silica gel) to yield 3-azido-4,4-dimethyltetrahydrofuran-2-ol 1H NMR (400 MHz, CDCl3) 5==5.58-5.29 (m, 1H), 3.93-3.76 (m, 2H), 3.66-3.50 (m, 2H), 1.19-1.08 (m, 6H) 4-azido-3,3-dimethyltetrahydrofuran (I-99-4): 3-azido-4,4-dimethyltetrahydrofuran-2-ol was taken in dichloromethane (80 mL), cooled to −78° C. then BF3·Et2O (3.6 mL, 28.7 mmol, 1.5 equivalent) was added slowly followed by addition of triethylsilane (6.1 mL, 2.0 mmol) at same temperature. The mixture was stirred at 0° C. for 4 hrs until no starting material remained. Then water (100 mL) was added to the mixture. The resulting phases were separated, then the aqueous phase was extracted with dichloromethane (2×50 mL). The organic extract was dried with anhydrous Na2SO4and concentrated under reduced pressure. The crude compound was purified by column chromatography (silica gel) to yield (S)-4-azido-3,3-dimethyltetrahydrofuran. 1H NMR (400 MHz, CDCl3)=4.17 (dd, J=6.1, 9.8 Hz, 1H), 3.77 (dd, J=3.9, 9.8 Hz, 1H), 3.65 (dd, J=3.9, 6.1 Hz, 1H), 3.58-3.50 (m, 2H), 1.13 (d, J=6.1 Hz, 6H). 4,4-dimethyltetrahydrofuran-3-amine (I-99): 4-azido-3,3-dimethyltetrahydrofuran (1.89 g, 13.4 mmol) dissolved in ethyl acetate (50 mL) was added 10% palladium on carbon (2.14 g, 2.0 mmol, 0.15 equivalent). The mixture was stirred at rt for 16 hrs. under 1 atm of hydrogen before filtering through a plug of celite. The solution was acidified 4 M HCl in methanol (5.0 mL) before concentrating in vacuo to afford 4,4-dimethyltetrahydrofuran-3-amine as the hydrochloride salt. 1H NMR (400 MHz, CDCl3)=4.09-4.05 (m, 1H), 3.72-3.68 (m, 1H), 3.59-3.56 (m, 1H), 3.44-3.42 (m, 1H), 3.35 (m, 1H), 1.07 (s, 6H). Preparation of Intermediate I-100 Methyl 3-[[(S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-4-nitrobenzoate (I-100): To a suspension of methyl 3-fluoro-4-nitrobenzoate (15.0 g, 75.3 mmol) and 4,4-dimethyltetrahydrofuran-3-amine hydrochloride (Intermediate I-99, 12.6 g, 82.9 mmol) in THE (100 mL) and DMF (50 mL), was added N,N-diisopropylethylamine (65.6 mL, 377 mmol). The resulting solution was heated at 80° C. overnight. The crude mixture was diluted with EtOAc (300 mL), washed with 5% LiCl (250 mL) and brine (250 mL). The organic extract was dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford methyl 3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-4-nitrobenzoate, the structure of which was confirmed by X-ray crystallography. 1H NMR (400 MHz, Chloroform-d) δ 8.24 (d, J=8.8 Hz, 2H), 7.57 (d, J=1.6 Hz, 1H), 7.27 (dd, J=8.9, 1.7 Hz, 1H), 4.41 (dd, J=9.2, 6.8 Hz, 1H), 4.02 (s, 1H), 3.97 (s, 3H), 3.76-3.64 (m, 3H), 1.26 (s, 3H), 1.18 (s, 3H). ES/MS: 295.0 (M+H+). Methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-101): In a 200 mL flask, a suspension of methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-82, 2329 mg, 4.86 mmol), Bis(pinacolato)diboron (1506 mg, 5.93 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (541 mg, 0.729 mmol), and potassium propionate (1635 mg, 14.6 mmol) in dioxane (24 mL) was degassed with Argon for 5 min, then heated for 50 min at 110° C. The mixture was cooled to rt. To the mixture was added sodium carbonate (2000 mmol/L, 5.48 mL, 11.0 mmol). The mixture was stirred at rt for 5 min. To the mixture was added [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (358 mg, 0.483 mmol) and 4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluorobenzonitrile (Intermediate I-3, 2238 mg, 7.29 mmol). The mixture was degassed for 5 min with Argon, then heated thermally at 90° C. for 1 hr. 15 min. The mixture was diluted with EtOAc and filtered over Celite. The filtrate was washed with brine, dried over sodium sulfate, and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-101), the structure of which was confirmed by X-ray crystallography. ES/MS: 626.6 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.56 (s, 1H), 8.02 (dd, J=8.5, 1.5 Hz, 1H), 7.79 (dd, J=9.7, 5.6 Hz, 2H), 7.70 (dt, J=14.3, 7.6 Hz, 2H), 7.53 (dt, J=7.3, 1.1 Hz, 1H), 7.49 (dd, J=7.9, 1.5 Hz, 1H), 7.43 (dd, J=9.3, 1.5 Hz, 1H), 7.09 (dd, J=11.3, 6.0 Hz, 1H), 6.86 (d, J=8.1 Hz, 1H), 5.61 (s, 2H), 4.73-4.56 (m, 2H), 4.46-4.32 (m, 34H), 3.96 (d, J=6.8 Hz, 4H), 3.81 (d, J=8.7 Hz, 1H), 1.36 (s, 3H), 0.69 (s, 3H). Preparation of Intermediate I-101 (Method 2) Methyl (S)-4-(2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetamido)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (I-101-1): HATU (12.2 g, 32.3 mmol) followed by N,N-Diisopropylethylamine (19.6 mL, 112 mmol) was added to a solution of 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid (Intermediate I-7, 10.1 g, 25.3 mmol) and methyl (S)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (Intermediate I-80, 6.07 g, 23.0 mmol) in DMF (36 mL) and MeCN (36 mL), and the resulting solution was stirred at rt overnight. Next, the mixture was diluted with EtOAc and washed sequentially with sat. NH4Cl, 10% LiCl, sat. NaHCO3(×2), 1N NaOH, and brine. The organic phase was dried over MgSO4, filtered, and concentrated to give methyl (S)-4-(2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetamido)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (Intermediate I-101-1), which was taken forward assuming 100% yield. ES/MS: 645.2 (M+1). Methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-101): Trifluoromethanesulfonic anhydride (5.79 mL, 34.4 mmol) was added dropwise to a solution of methyl (S)-4-(2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetamido)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (I-101-1, 14.8 g, 23.0 mmol) and triphenylphosphine oxide (9.59 g, 34.4 mmol) in DCM under argon at 0° C. for 10 min. The mixture was stirred for 15 min, brought to rt, and stirred for 25 min. The reaction was quenched with sat. NaHCO3, and phases were separated. The aqueous layer was extracted with DCM. Combined organic layers were dried over MgSO4, filtered, concentrated, and purified by silica gel flash column chromatography (EtOAc/Hex gradient) to give methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-101). Preparation of Intermediate I-102 2-bromo-6-[(4-chloro-2-fluoro-phenyl)methoxy]pyridine (Intermediate I-102): A round-bottom flask was charged with (4-chloro-2-fluoro-phenyl)methanol (800 mg, 5.0 mmol, 1.1 equivalent), 2-bromo-6-fluoropyridine (800 mg, 4.6 mol, 1.0 equivalent), and cesium carbonate (2.3 g, 6.8 mmol, 1.5 equivalent). Anhydrous acetonitrile (15 mL) was added, and the resulting mixture was heated to reflux and stirred for 12 hrs. After cooling to rt, the mixture was filtered through a plug of Celite and then concentrated in vacuo. The residue was purified by column chromatography (silica gel, EtOAc/hexane gradient) to yield the title compound. ES/MS m/z: 317.8 (M+H)+. Preparation of Intermediate I-103 Methyl 2-[[4-[6-[(4-chloro-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-103). To a 25-mL microwave vial was charged with methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-82, 800 mg, 1.67 mmol, 1.0 equivalent), PdCl2(dppf)2 (186 mg, 0.25 mmol, 0.15 equivalent), potassium propionate (560 mg, 5.01 mmol, 3.0 equivalent), and B2Pin2(510 mg, 2.00 mol, 1.2 equivalent). Anhydrous 1,4-dioxane (10 mL) was added and the resulting mixture was purged with argon for 2 min. The mixture was sealed and heated to 120° C. by microwave and stirred for 1 h. After cooling down to rt, 2-bromo-6-[(4-chloro-2-fluoro-phenyl)methoxy]pyridine (Intermediate I-102, 580 mg, 1.84 mmol, 1.1 equivalent), PdCl2(dppf)2 (62 mg, 0.0834 mmol, 0.05 equivalent), 2 M aqueous Na2CO3(2.0 mL, 4.17 mmol, 2.5 equivalent) were added, respectively. The resulting mixture was heated to 100° C. under argon and stirred for 3 hrs. before cooling to rt and filtered through a plug of Celite and MgSO4. The filtrate was concentrated and purified by column chromatography (silica gel, EtOAc/hexane gradient) to yield the title compound. ES/MS m/z: 635.6 (M+H)+. Preparation of Intermediate I-104 Methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-104). 2-[4-[6-[(4-Cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetic acid (Intermediate I-7, 500 mg, 1.26 mmol), methyl 4-amino-3-[(4,4-dimethyltetrahydrofuran-3-yl)amino]benzoate (Intermediate I-25, 365 mg, 1.38 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (573 mg, 1.51 mmol) were combined in DMF (5.00 mL) and N,N-Diisopropylethylamine (1.09 mL, 6.28 mmol) was added. The mixture was stirred at r.t. for 3 hrs. The mixture was partitioned between EtOAc and sat. NH4C1. The organic phase was dried, filtered and concentrated in vacuo. The crude was taken up in acetic acid (2 mL) and heated to 100° C. for 72 hrs. The resulting mixture was then concentrated in vacuo and partitioned between EtOAc and sat. NaHCO3. The organic phase was dried, filtered and concentrated in vacuo. Silica gel flash chromatography (EtOAc/hexane) yielded the title compound. ES/MS m/z 627.2 (M+H)+. Preparation of Intermediate I-105 4-bromo-2-[(4-chloro-2-fluoro-phenyl)methoxy]pyrimidine (Intermediate I-105): To a 50 mL RBF was added (4-chloro-2-fluoro-phenyl)methanol (186 mg, 1.16 mmol) to THE (6 mL) and the flask was placed under nitrogen, and cooled to 0° C. Potassium tert-butoxide (1.05 mL, 0.362 mmol, 1 M THF) was added dropwise, and the solution was stirred 15 min at 0° C. To a separate 50 mL RBF was added 4-bromo-2-methylsulfonyl-pyrimidine (250 mg, 1.05 mmol) in THE (6 mL) and the mixture was cooled to −78° C. The first solution (deprotonated alcohol) was taken up in a syringe, and added dropwise to the second solution at −78° C. The solution was stirred for 1 h at −78° C., then 1 mL water was added dropwise, and the solution allowed to warm to rt. The solution was diluted with EtOAc and water, and 2 mL saturated aqueous NH4Cl was added to acidify the solution. Then the layers were separated, and the aqueous layer was extracted once with EtOAc. The combined organic layers were dried over MgSO4, filtered, concentrated, and purified by silica gel flash column chromatography (EtOAc/Hex gradient) to yield 4-bromo-2-[(4-chloro-2-fluoro-phenyl)methoxy]pyrimidine (Intermediate I-105). ES/MS m/z: 318.9 (M+H)+. Preparation of Intermediate I-106 Methyl (S)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluoro-4-nitrobenzoate (Intermediate I-106): To a solution of methyl 3,5-difluoro-4-nitro-benzoate (2.21 g, 10.2 mmol) and (S)-4,4-dimethyltetrahydrofuran-3-amine hydrochloride (Intermediate I-99, 1.70 g, 11.2 mmol) in tetrahydrofuran (12.5 mL) and N,N-dimethylformamide (6.0 mL) was added N,N-diisopropylethylamine (8.86 mL, 50.9 mmol). The mixture was stirred at 70° C. overnight before being cooled to rt. The mixture was diluted with water, extracted with EtOAc, washed with brine, dried over sodium sulfate, filtered, and concentrated to yield the title compound, which was carried forward to subsequent steps without further purification. Preparation of Intermediate I-107 Methyl (S)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate (Intermediate I-107): Methyl (S)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluoro-4-nitrobenzoate (Intermediate I-106, 2.88 g, 10.2 mmol) was dissolved in EtOAc, and put under argon. To this mixture was added 10% palladium on carbon (1.08 g, 1.02 mmol), and then the mixture was placed under hydrogen gas. The mixture was stirred overnight, then the mixture filtered through celite, and concentrated in vacuo. Purification by silica gel flash column chromatography (EtOAc/Hexane gradient) yielded methyl (S)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate (Intermediate I-107). ES/MS m/z: 283.2 (M+H)+. Preparation of Intermediate I-108 Methyl (S)-4-(2-(4-bromo-2,5-difluorophenyl)acetamido)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate (I-108-1): To a solution of methyl (S)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate (Intermediate I-107, 2000 mg, 7.08 mmol) and 2-(4-bromo-2,5-difluoro-phenyl)acetic acid (1867 mg, 7.44 mmol) in MeCN (10.0 mL) and cooled to 0° C. was added 1-methylimidazole (2.91 g, 2.82 mL, 35.4 mmol) followed by N,N,N′,N′-Tetramethylchloroformamidinium Hexafluorophosphate (2.39 g, 8.50 mmol). The mixture was warmed to rt and stirred for 30 min. The mixture was concentrated in vacuo to yield methyl (S)-4-(2-(4-bromo-2,5-difluorophenyl)acetamido)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate (I-108-1), which was carried forward to the next step without further purification. Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluorobenzimidazole-5-carboxylate (Intermediate I-108): To methyl (S)-4-(2-(4-bromo-2,5-difluorophenyl)acetamido)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate (I-108-1, 2.2 g, 4.3 mmol) in 21 mL of 1,2-dichloroethane was added phosphoryl trichloride (2.6 g, 17 mmol, 1.6 mL). The solution was heated to 80° C. for 24 hrs., then cooled to rt. An aliquot of 20 mL of water was added and stirring for 1 h, then aqueous sodium hydroxide (26 mL, 51 mmol, 2 M) was added. The mixture was diluted with DCM, layers separated, and the organic phase washed with brine, dried with MgSO4, filtered, and concentrated. Purification by silica chromatography (EtOAc/hexane gradient) yielded methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluorobenzimidazole-5-carboxylate (Intermediate I-108). ES/MS m/z: 497.0 (M+H)+. Preparation of Intermediate I-109 4-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (Intermediate I-109): To a solution of 6-bromo-2-chloro-3-fluoro-pyridine (4.90 g, 23.3 mmol) and 3-fluoro-4-(hydroxymethyl)benzonitrile (3.87 g, 25.6 mmol) in acetonitrile (40 mL) was added cesium carbonate (15.2 g, 25.6 mmol). The mixture was stirred at 60° C. overnight before being cooled to rt. The mixture was diluted with water, extracted three times with EtOAc, washed with brine, dried over sodium sulfate, filtered, and concentrated. Purification by silica gel flash column chromatography (EtOAc in Hexane gradient) yielded 4-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (Intermediate I-109). ES/MS m/z: 326.1 (M+H)+. Preparation of Intermediate I-110 Methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluoro-benzimidazole-5-carboxylate (Intermediate I-110): A 2 mL microwave vial was charged with methyl 2-[(4-bromo-2,5-difluorophenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluorobenzimidazole-5-carboxylate (Intermediate I-108, 400 mg, 0.69 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (184 mg, 0.73 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (77.7 mg, 0.1 mmol), potassium propionate (233 mg, 2.08 mmol), and 2.5 mL of 1,4-dioxane. This mixture had argon bubbled through for 5 min before being heated in a microwave for 1 h at 120° C. Then sodium carbonate (0.72 mL, 1.44 mmol, 2M) was added followed by 4-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]-3-fluorobenzonitrile (Intermediate I-109, 247 mg, 0.76 mmol). The mixture was heated to 110° C. for 1 h, then filtered, concentrated, and purified by silica gel chromatography (EtOAc/hexane gradient) to yield methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluorophenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluorobenzimidazole-5-carboxylate (Intermediate I-110). ES/MS m/z: 663.6 (M+H)+. Preparation of Intermediate I-111 Methyl (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-111): A 2 mL microwave vial was charged with methyl 2-[(4-bromo-2,5-difluorophenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluorobenzimidazole-5-carboxylate (Intermediate I-108, 45 mg, 0.09 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (24 mg, 0.09 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (10.2 mg, 0.01 mmol), potassium propionate (30.4 mg, 0.27 mmol), and 2.5 mL of 1,4-dioxane. This mixture had argon bubbled through for 5 min before being heated in a microwave for 1 h at 120° C. Then sodium carbonate (0.09 mL, 0.188 mmol, 2M) was added followed by 4-bromo-2-[(4-chloro-2-fluoro-phenyl)methoxy]pyrimidine (Intermediate I-105, 46.0 mg, 0.14 mmol). The mixture was heated to 110° C. for 1 h, then filtered, concentrated, and purified by silica gel chromatography (EtOAc/hexane gradient) to yield methyl (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-111). ES/MS m/z: 656.1 (M+H)+. Preparation of Intermediate I-112 (4-chloro-6-fluoropyridin-3-yl)methanol (I-112-1): NaBH4(117 mg, 3.09 mmol) was added to a solution of 4-chloro-6-fluoronicotinaldehyde (470 mg, 2.95 mmol) in MeOH (12 mL) at 0° C., and the solution was stirred for 5 min. The reaction was quenched with saturated NH4Cl and concentrated. It was dissolved in EtOAc, washed with water then brine, dried over MgSO4, filtered, and concentrated to give (4-chloro-6-fluoropyridin-3-yl)methanol (I-112-1). ES/MS: 161.9 (M+H)±. 4-chloro-5-(chloromethyl)-2-fluoropyridine (I-112-2): Thionyl chloride (0.53 mL, 7.4 mmol) was added to a solution of (4-chloro-6-fluoropyridin-3-yl)methanol (I-112-1, 476 mg, 2.95 mmol) in DCM (25 mL), and the resulting solution was stirred for 1 hr. More thionyl chloride (0.53 mL, 7.4 mmol) was added, and the resulting solution was stirred for 1 hr. More thionyl chloride (0.21 mL, 2.9 mmol) was added, and the resulting solution was stirred for 30 min. The mixture was concentrated, redissolved in DCM, and saturated NaHCO3was added dropwise slowly. Phases were separated, and the organic phase was dried over MgSO4, filtered, and concentrated to give 4-chloro-5-(chloromethyl)-2-fluoropyridine (I-112-2). 1H NMR (400 MHz, Chloroform-d) δ 8.30 (s, 1H), 7.05 (d, J=2.7 Hz, 1H), 4.67 (s, 2H). 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-fluoropyridine (I-112-3): A slurry of 6-bromopyridin-2-ol (557 mg, 3.2 mmol), 4-chloro-5-(chloromethyl)-2-fluoropyridine (I-112-2, 480 mg, 2.7 mmol), and Cs2CO3(2.17 g, 6.7 mmol) in CAN (9 mL) was heated at 70° C. for 30 min. The mixture was filtered through celite, concentrated, and purified by silica gel flash column chromatography (EtOAc in Hex gradient) to give 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-fluoropyridine (I-112-3). ES/MS: 317.0 (M+H)+. 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (Intermediate I-112): A slurry of 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-fluoropyridine (I-112-3, 110 mg, 0.31 mmol), 1H-1,2,3-triazole (0.018 mL, 0.31 mmol), and K2CO3(87 mg, 0.63 mmol) in DMSO (1.4 mL) was heated at 70° C. for 4 hrs. The mixture was diluted with brine and extracted 2× with EtOAc. The combined organic phase was dried over MgSO4, filtered, concentrated, and purified by silica gel flash column chromatography (EtOAc in Hex gradient) to give 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (Intermediate I-112) as the earlier eluting of two isomeric compounds. 1H NMR (400 MHz, Chloroform-d) δ 8.66 (s, 1H), 8.58 (s, 1H), 8.31 (s, 1H), 7.84 (d, J=1.2 Hz, 1H), 7.48 (t, J=7.8 Hz, 1H), 7.13 (d, J=7.5 Hz, 1H), 6.79 (d, J=8.2 Hz, 1H), 5.54 (s, 2H). Preparation of Intermediate I-113 Methyl (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-113): A slurry of methyl (S)-2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-82, 20 mg, 0.04 mmol), bis(pinacolato)diboron (13.8 mg, 0.05 mmol), potassium propionate (14 mg, 0.125 mmol), and bis(diphenylphosphino)ferrocene] dichloropalladium (II) (4.6 mg, 0.006 mmol) in dioxane (1 mL) was sparged with argon for 5 min, sealed, and heated at 110° C. for 1 hr. The mixture was cooled to rt, sodium carbonate (2M, 0.05 mL, 0.1 mmol) was added, and the mixture was stirred at rt for 5 min. Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (3.1 mg, 0.004 mmol) and a solution of 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (Intermediate I-112) in dioxane (1 mL) were added, and the mixture was degassed for 5 min with Ar, then heated at 90° C. for 2 hrs. The mixture was diluted with EtOAc, washed with brine (2×), dried over MgSO4, filtered, concentrated, and purified by silica gel flash chromatography (EtOAc in Hexane gradient) to give methyl (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-113). ES/MS m/z: 686.1 (M+H)+. Preparation of Intermediate I-114 4-[(6-bromo-2-pyridyl)oxymethyl]benzonitrile (Intermediate I-114): A mixture of 4-(bromomethyl)benzonitrile (200 mg, 1.0 mmol), 6-bromopyridin-2-ol (180 mg, 1.0 mmol), and cesium carbonate (400 mg, 1.2 mmol) was stirred in acetonitrile (5 mL) at rt for 3 hrs. The mixture was filtered through Celite and concentrated in vacuo to yield 4-[(6-bromo-2-pyridyl)oxymethyl]benzonitrile (Intermediate I-114). ES/MS m/z: 289.2 (M+H)+. Preparation of Intermediate I-115 Methyl 2-[[4-[6-[(4-cyanophenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-115): To methyl 2-[(4-bromo-3-fluoro-phenyl)methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (Intermediate I-82, 30 mg, 0.0626 mmol) was added bis(pinacolato)diboron (23 mg, 0.090 mmol), potassium propionate (23 mg, 0.208 mmol), Pd(dppf)C12(7.7 mg, 0.0104 mmol, and dioxane (1.5 mL). The mixture was degassed 30 sec under argon, and heated for 16 hrs. at 100° C. Then sodium carbonate (2M in water, 0.070 mL, 0.14 mmol) was added and the mixture stirred for 1 min at rt. Then more Pd(dppf)Cl2(3.9 mg, 0.0052 mmol) was added, followed by 4-[(6-bromo-2-pyridyl)oxymethyl]benzonitrile (Intermediate I-114, 20 mg, 0.0692 mmol). The mixture was sealed under argon and heated for 3 hrs. at 90° C. The organic layer was purified directly by silica gel flash chromatography (EtOAc in Hex gradient) to yield methyl 2-[[4-[6-[(4-cyanophenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-115). ES/MS m/z: 609.5 [M+H]+. Preparation of Intermediate I-116 Methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorophenyl)acetate (I-116-1): A mixture of methyl (4-bromo-2,3,6-trifluorophenyl)acetate (26.0 g, 91.9 mmol, 1.0 eq), Bis(pinacolato)diboron (30.3 g, 119.4 mmol, 1.3 eq), Pd(dppf)Cl2(3.36 g, 4.6 mmol, 0.05 eq), and potassium propionate (36.1 g, 321.7 mmol, 3.5 eq) in dioxane (400 mL) was degassed with Argon. The mixture was stirred at 100° C. overnight. The mixture was cooled to room temperature, followed by added aq. Na2CO3(2.0 M, 92 mL, 183.8 mmol, 2.0 eq), Pd(dppf)Cl2(3.36 g, 4.6 mmol, 0.05 eq) and 4-(((6-bromopyridin-2-yl)oxy)methyl)-3-fluorobenzonitrile (Intermediate I-3, 28.2 g, 91.9 mmol, 1.0 eq). The mixture was degassed with Argon gas, then the mixture was stirred at 100° C. for 2 hrs. The mixture was diluted with EtOAc (1000 mL) and washed with brine (500 mL×2). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash chromatography (petroleum ether/EtOAc gradient) to give methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorophenyl)acetate (I-116-1). 1H NMR (400 MHz, DMSO-d6): δ 7.93-7.90 (m, 2H), 7.78-7.72 (m, 2H), 7.65-7.60 (m, 1H), 7.56-7.54 (m, 1H), 7.04 (d, J=8.4 Hz, 1H), 5.59 (s, 2H), 3.88 (s, 2H), 3.68 (s, 3H). ES/MS m/z: 431.0 [M+H]+. 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorophenyl)acetic acid (Intermediate I-116): A solution of methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorophenyl)acetate (I-116-1, 12.0 g, 27.9 mmol, 1.0 eq) and aq. LiOH (2 M, 16.8 mL, 33.5 mmol, 1.3 eq) in CH3CN (130 mL) was stirred at room temperature for 5 hrs. The mixture was acidified with 1N HCl to pH=6. The precipitated solid was collected by filtration and washed with water (10 mL×2), then dried to afford 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorophenyl)acetic acid (Intermediate I-116). 1H NMR (400 MHz, DMSO-d6): δ 7.93-7.90 (m, 2H), 7.89-7.86 (m, 2H), 7.50-7.42 (m, 2H), 6.98 (d, J=8.4 Hz, 1H), 5.58 (s, 2H), 3.28 (s, 2H). ES/MS m/z: 417.0 [M+H]+. Preparation of Intermediate I-117 Methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate 117): To a solution of 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorophenyl)acetic acid (Intermediate I-116, 320 mg, 0.77 mmol) in DMF (20 mL) was added methyl 4-amino-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate (Intermediate I-80, 203 mg, 0.77 mmol), o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (292 mg, 0.77 mmol), and DIPEA (0.4 mL, 2.31 mmol). The resulting solution was stirred at room temperature for 2 hrs. after which the mixture was poured into 100 mL of saturated sodium bicarbonate solution and extracted with EtOAc (2×50 mL). The combined organic extracts were washed with 20 mL of brine, dried over MgSO4. The organic layer was filtered and concentrated. The resulting residue was dissolved in 5.0 mL of acetic acid and stirred at 100° C. for 16 hrs. The mixture was cooled down and the solvent removed in vacuo. The mixture was diluted with 50 mL of EtOAc and washed with 50 mL of saturated aqueous NaHCO3. The organic layer was dried over sodium sulfate. The organic layer was filtered and concentrated. It was purified by silica gel chromatography (eluent: EtOAc/hexanes) to give methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate 117). ES/MS: 645.7 (M+H)+. Preparation of Intermediate I-118 Methyl (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-118): To a vial was added methyl (S)-2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-82, 100 mg, 0.21 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (15.5 mg, 0.021 mmol), Bis(pinacolato)diboron (80 mg, 0.3 mmol) and potassium propionate (70 mg, 0.63 mmol) followed by 1,4-Dioxane (1.0 mL). Argon was bubbled through the solution for 3 min then the mixture was heated to 110° C. for 70 min. To the mixture was added aqueous sodium carbonate (2.00 M, 0.21 mL, 0.42 mmol), [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (6.6 mg, 0.009 mmol), and 4-bromo-2-[(4-chloro-2-fluoro-phenyl)methoxy]pyrimidine (Intermediate I-105, 99.3 mg, 0.3 mmol). Argon was bubbled through the solution for 3 min then the mixture was heated to 100° C. for 45 min. The mixture was filtered through celite, washing with DCM and concentrated in vacuo. The crude residue was purified by silica gel flash column chromatography (0-100% EtOAc in hexane) to give Methyl (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-118). ES/MS m/z: 637.8 (M+H+). Preparation of Intermediate I-119 Methyl 2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-119): To a solution of 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid (1.2 g, 4.46 mmol) in ACN (50 mL) was added methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate (Intermediate I-25, 1.26 g, 4.46 mmol), Chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (1.5 g, 5.35 mmol), and 1-methylimidazole (1.83 mL, 22.3 mmol). The resulting solution was stirred at room temperature for 16 hrs. after which the mixture was poured into 100 mL of saturated sodium bicarbonate solution and extracted with EtOAc (2×50 mL). The combined organic extracts were washed with 50 mL of brine, dried over MgSO4. The organic layer was filtered and concentrated. The mixture was purified by silica gel chromatography (eluent: EtOAc/hexanes) to yield Intermediate I-119-1, which was dissolved in 50.0 mL of acetic acid. It was stirred at 120° C. for 40 hrs. The mixture was cooled down and removed the solvent. The mixture was diluted with 100 mL of EtOAc and washed with 100 mL of saturated aqueous NaHCO3. The organic layer was dried over sodium sulfate. The organic layer was filtered and concentrated. It was purified by silica gel flash column chromatography (eluent: EtOAc/hexanes) to give methyl 2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-119). ES/MS: 515.6 (M+H)+. Preparation of Intermediate I-120 Methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-120): To methyl 2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-119, 300 mg, 0.58 mmol) was added [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (43.2 mg, 0.058 mmol), Bis(pinacolato)diboron (222 mg, 0.87 mmol) and potassium propionate (196 mg, 1.75 mmol) followed by 1,4-Dioxane (3.0 mL). Argon was bubbled through the solution for 3 min then the mixture was heated to 110° C. for 90 min. To the mixture was added aqueous sodium carbonate (2.00 M, 0.58 mL, 1.16 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (18.1 mg, 0.024 mmol) and 4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (Intermediate I-3, 268 mg, 0.87 mmol). Argon was bubbled through the solution for 3 min then the mixture was heated to 100° C. for 60 min. The mixture was filtered through celite, eluted with DCM, and the filtrate was concentrated in vacuo. The crude residue was purified by silica gel flash column chromatography (EtOAc/Hex gradient) to give methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-120). ES/MS m/z: 663.5 (M+H+). Preparation of Intermediate I-121 Tert-butyl 2,3-difluoro-4-nitrobenzoate: 2,3-difluoro-4-nitrobenzoic acid (1.00 g, 4.92 mmol) in THE (15 mL) at ambient temperature was treated with di-tert-butyl decarbonate (2.15 g, 9.9 mmol) followed by 4-dimethylaminopyridine (180 mg, 1.5 mmol) and the resulting mixture was heated to 40° C. for 3 hrs. Upon completion, the mixture was poured into water (20 mL) and extracted with EtOAc (3×30 mL). The combined organic extracts were washed with brine (15 mL), dried over MgSO4, and purified by silica gel chromatography (eluent: EtOAc/hexanes) to yield tert-butyl 2,3-difluoro-4-nitrobenzoate. Tert-butyl 4-amino-2-fluoro-3-((2-methoxyethyl)amino)benzoate (Intermediate I-121): tert-butyl 2,3-difluoro-4-nitrobenzoate (200 mg, 0.77 mmol) was dissolved in THE (2 mL) after which 2-methoxyethanamine (0.080 mL, 0.93 mmol) and diisopropylethylamine (0.40 mL, 2.3 mmol) were added and the resulting mixture heated to 60° C. for 16 hrs. Upon completion the mixture was concentrated directly, the crude residue then taken up in EtOAc (25 mL) and washed with saturated aq. NH4Cl (2×5 mL). The combined organic extracts were washed with brine (5 mL), dried over MgSO4, concentrated and carried forward without purification. Crude tert-butyl 2-fluoro-3-((2-methoxyethyl)amino)-4-nitrobenzoate was dissolved in EtOH (5 mL) after which iron (216 mg, 3.9 mmol) and saturated aq. NH4Cl (2 mL) were added. The resulting mixture was heated to 60° C. for 3 hrs. Upon completion the solids were removed by filtration washing with EtOAc (20 mL) and MeOH (20 mL). The filtrate was then concentrated, taken up in EtOAc (25 mL), and washed with water (5 mL) and brine (5 mL). The organic layer was then dried over MgSO4and purified by silica gel chromatography (eluent: EtOAc/hexanes) to give tert-butyl 4-amino-2-fluoro-3-((2-methoxyethyl)amino)benzoate (Intermediate I-121). ES/MS: 284.9 (M+H+). Preparation of Intermediate I-122 Tert-butyl 2-(4-bromo-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-122): To a solution of 2-(4-bromo-2,5-difluoro-phenyl)acetic acid (318 mg, 1.27 mmol) and tert-butyl 4-amino-2-fluoro-3-(2-methoxyethylamino)benzoate (Intermediate I-121, 250 mg, 0.9 mmol) in MeCN (8 mL) was added 1-methylimidazole (0.35 mL, 4.4 mmol) and N,N,N′,N′-tetramethylchloroformamidinium hexafluorophosphate (TCFH, 300 mg, 1.1 mmol). The solution was stirred at room temperature for 2 hrs., diluted with EtOAc and washed with HCl (1 M, aqueous). The organic layer was concentrated to provide tert-butyl 4-[[2-(4-bromo-2,5-difluoro-phenyl) acetyl] amino]-2-fluoro-3-(2-methoxyethylamino)benzoate (I-122-1), which was dissolved in 5 mL of DCE and 0.38 mL of acetic acid. The mixture was stirred at 60° C. for 4 hrs. The reaction was quenched with NaHCO3and extracted 2× with DCM. The organic phase was washed with brine, dried, filtered, concentrated, and purified by silica gel flash chromatography (EtOAc in Hexane gradient) to give tert-butyl 2-(4-bromo-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-122). 1H NMR (400 MHz, Chloroform-d) δ 7.86 (dd, J=8.6, 6.7 Hz, 1H), 7.65 (d, J=8.6 Hz, 1H), 7.35 (dd, J=8.7, 5.6 Hz, 1H), 7.13 (dd, J=8.3, 6.4 Hz, 1H), 4.62-4.40 (m, 4H), 3.78 (t, J=5.1 Hz, 2H), 3.29 (s, 3H), 1.64 (s, 9H). Preparation of Intermediate I-123 Tert-butyl 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-123). A suspension of tert-butyl 2-(4-bromo-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-122, 50 mg, 0.1 mmol), Bis(pinacolato)diboron (34 mg, 0.135 mmol), [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (11.5 mg, 0.016 mmol), and potassium propionate (35 mg, 0.3 mmol) in dioxane (2 mL) was degassed with Ar for 20 min. The mixture was sealed and heated at 100° C. for 2 hrs. Sodium carbonate (2.0 M, 0.1 mL, 0.2 mmol) was added and the mixture was stirred at rt for 10 min. [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (6 mg, 0.008 mmol) and 4-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (Intermediate I-109, 34 mg, 0.1 mmol) were added, the mixture was degassed for 10 min with Ar2, then sealed and heated at 90° C. for 3 hrs. The mixture was diluted with EtOAc and washed with brine. The organic extract was dried over sodium sulfate and chromatographed (eluent: EtOAc/hexanes) to give tert-butyl 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-123). ES/MS m/z: 665.2 (M+H+). Preparation of Intermediate I-1001 Tert-Butyl (1R,3R,5R)-3-[(2-amino-5-methoxycarbonyl-anilino)methyl]-2-azabicyclo[3.1.0]hexane-2-carboxylate (Intermediate I-1001): Tert-Butyl (1R,3R,5R)-3-[(2-amino-5-methoxycarbonyl-anilino)methyl]-2-azabicyclo[3.1.0]hexane-2-carboxylate was prepared in a manner as described for Intermediate I-1 substituting tert-butyl (1R,3R,5R)-3-(aminomethyl)-2-azabicyclo[3.1.0]hexane-2-carboxylate for 2-methoxyethylamine. ES/MS: 362.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.39 (d, J=8.1 Hz, 1H), 7.15 (s, 1H), 6.63 (d, J=8.1 Hz, 1H), 4.58 (t, J=11.0 Hz, 1H), 3.87 (s, 3H), 3.64 (d, J=7.3 Hz, 1H), 3.27-3.09 (m, 1H), 3.00 (t, J=11.0 Hz, 1H), 2.56 (q, J=11.7 Hz, 1H), 1.95-1.70 (m, 1H), 1.53 (s, 10H), 0.81 (q, J=7.1 Hz, 1H), 0.57 (d, J=5.7 Hz, 1H). Preparation of Intermediate I-1002 2-bromo-4-[[2-fluoro-4-(trifluoromethyl)phenyl]methoxy]pyrimidine (I-1002): To a solution of [2-fluoro-4-(trifluoromethyl)phenyl]methanol (1.1 g, 5.65 mmol) in tetrahydrofuran (3 mL) was added potassium tert-butoxide (0.333 g, 2.97 mmol). The solution was then stirred for 5 min at room temperature. Next, 2-bromo-4-fluoro-pyrimidine (0.500 g, 2.83 mmol) in N,N-dimethylformamide (5 mL) were added and the mixture was cooled to −78° C. Next, the mixture was removed from the dry-ice bath and stirred at rt over weekend. Following this, the mixture was diluted with EtOAc and washed with 5% LiCl (2×) and brine. The organic extract was dried over sodium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 351.0, 353.0 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.31 (d, J=5.7 Hz, 1H), 7.66 (t, J=7.5 Hz, 1H), 7.48 (dd, J=8.0, 1.5 Hz, 1H), 7.41 (dd, J=9.7, 1.7 Hz, 1H), 6.79 (d, J=5.7 Hz, 1H), 5.56 (s, 2H). 19F NMR (376 MHz, Chloroform-d) δ −63.43, −115.50 (d, J=7.5 Hz). Preparation of Intermediate I-1003 [2-fluoro-4-[1-(trifluoromethyl)pyrazol-4-yl]phenyl]methanol (I-1003): A suspension of (4-bromo-2-fluoro-phenyl)methanol (100 mg, 0.49 mmol), 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-(trifluoromethyl)pyrazole (153 mg, 0.59 mmol), 1,1′-Bis(di-isopropylphosphino)ferrocene palladium dichloride (29 mg, 0.049 mmol), and potassium carbonate (200 mg, 1.4 mmol) in 1,4-Dioxane anhydrous, 99.8% (2 mL) and Water (1 mL) was degassed with argon for 5 min, then heated thermally at 100° C. for 1 hr. Upon completion, the mixture was diluted with EtOAc and washed with brine. The organic extract was dried over sodium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 261.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.06 (s, 1H), 8.04 (t, J=0.9 Hz, 1H), 7.51 (t, J=7.8 Hz, 1H), 7.33 (dd, J=7.8, 1.7 Hz, 1H), 7.22 (dd, J=10.8, 1.7 Hz, 1H), 4.82 (s, 2H). 19F NMR (376 MHz, Chloroform-d) δ −61.00, −119.32. Preparation of Intermediate I-1004 Methyl 6-(1,2,4-triazol-1yl)pyridine-3-carboxylate: A suspension of methyl 6-chloropyridine-3-carboxylate (500 mg, 2.91 mmol), 1H-1,2,4-triazole (282 mg, 4.08 mmol), and Potassium carbonate (564 mg, 4.08 mmol) in NMP (10 mL) was heated at 80° C. overnight. Upon completion, the mixture was diluted with EtOAc and washed with 5% LiCl (2×) and brine. The organic extract was dried over sodium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 205.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 9.27 (s, 1H), 9.10 (dd, J=2.2, 0.8 Hz, 1H), 8.51 (dd, J=8.6, 2.2 Hz, 1H), 8.15 (s, 1H), 8.01 (dd, J=8.5, 0.8 Hz, 1H), 4.01 (s, 3H). 6-(1,2,4-triazol-1-yl)pyridine-3-carboxylic acid: A solution of methyl 6-(1,2,4-triazol-1-yl)pyridine-3-carboxylate (200 mg, 0.980 mmol) and Lithium hydroxide, monohydrate (215 mg, 5.12 mmol) in CH3CN (9 mL) and water (3 mL) was stirred at rt overnight. Next, the mixture was diluted with EtOAc. Adjusted to pH-6 with 1N HCl (2.8 mL) and then filtered and air-dried to give desired product, which was used in the next step without further purification. ES/MS: 191.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 9.48 (s, 1H), 9.01 (d, J=2.2 Hz, 1H), 8.52 (dd, J=8.5, 2.2 Hz, 1H), 8.38 (s, 1H), 7.99 (d, J=8.6 Hz, 1H). [6-(1,2,4-triazol-1-yl)-3-pyridyl]methanol (I-1004): 1,1′-carbonyldiimidazole (261 mg, 1.61 mmol) was added to a solution of 6-(1,2,4-triazol-1-yl)pyridine-3-carboxylic acid (153 mg, 0.805 mmol) in THE (6 mL) was added. The solution was stirred for 2 hr. Upon completion, the mixture was cooled to 0° C. and then a solution of NaBH4(152 mg, 4.02 mmol) in 1 mL water was added. Next, the mixture was stirred for 30 min at rt. Upon completion, 1 mL conc HCL slowly added, then the mixture concentrated to remove the THF. Upon removal of the THF, the mixture was neutralized with saturated NaHCO3and extracted with EtOAc 3×. The organic extract was washed with brine and dried over sodium sulfate. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 177.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 9.21 (s, 1H), 8.48 (s, 1H), 8.13 (s, 1H), 7.94 (t, J=1.6 Hz, 2H), 4.83 (s, 2H) Preparation of Intermediate I-1005 [6-(triazol-1-yl)-3-pyridyl]methanol (I-1005): [6-(triazol-1-yl)-3-pyridyl]methanol was prepared in a manner as described for Intermediate I-1004 substituting 1H-triazole for 1H-1,2,4-triazole. ES/MS: 177.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.62 (d, J=1.2 Hz, 1H), 8.56-8.45 (m, 1H), 8.23 (d, J=8.2 Hz, 1H), 7.98 (dd, J=8.4, 2.3 Hz, 1H), 7.86 (d, J=1.2 Hz, 1H), 4.85 (s, 2H). Preparation of Intermediate I-1006 Methyl 6-oxazol-5-ylpyridine-3-carboxylate: A suspension of methyl 6-chloropyridine-3-carboxylate (200 mg, 1.2 mmol), 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)oxazole (273 mg, 1.4 mmol), 1,1′-Bis(di-isopropylphosphino)ferrocene palladium dichloride (69 mg, 0.12 mmol), and potassium carbonate (477 mg, 3.5 mmol) in 1,4-Dioxane (4 mL) and water (2 mL) was degassed with argon for 5 min. The mixture was then heated thermally at 100° C. for 1 hr. Upon completion, the mixture was diluted with EtOAc and washed with brine. The organic extract was dried over sodium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 205.2 (M+H+) 0.1H NMR (400 MHz, Chloroform-d) δ 9.25 (dd, J=2.1, 0.9 Hz, 1H), 8.40 (dd, J=8.3, 2.1 Hz, 1H), 8.05 (s, 1H), 7.86 (s, 1H), 7.77 (dd, J=8.3, 0.9 Hz, 1H), 4.00 (s, 3H). 6-oxazol-5-ylpyridine-3-carboxylic acid: A solution of methyl 6-oxazol-5-ylpyridine-3-carboxylate (117 mg, 0.573 mmol) and lithium hydroxide, monohydrate (68.0 mg, 1.62 mmol), in CH3CN (3 mL) and water (1 mL) was stirred at rt overnight. The mixture was diluted with EtOAc. Next the mixture was adjusted to pH-6 with 1N HCl (1.6 mL) and then filtered and air-dried to give desired product, which was used in the next step without further purification. ES/MS: 191.2 (M+H+) 0.1H NMR (400 MHz, Chloroform-d) δ 8.31 (d, J=5.7 Hz, 1H), 7.66 (t, J=7.5 Hz, 1H), 7.48 (dd, J=8.0, 1.5 Hz, 1H), 7.41 (dd, J=9.7, 1.7 Hz, 1H), 6.79 (d, J=5.7 Hz, 1H), 5.56 (s, 2H). 19F NMR (376 MHz, Chloroform-d) δ −63.43, −115.50 (d, J=7.5 Hz). (6-oxazol-5-yl-3-pyridyl)methanol (I-1006): 1,1′-carbonyldiimidazole (141 mg, 0.872 mmol) was added to a suspension of 6-oxazol-5-ylpyridine-3-carboxylic acid (82.9 mg, 0.436 mmol) in THE (6 mL). The mixture was stirred for 2 hr. Upon completion, the mixture was cooled to 0° C.° C. then a solution of NaBH4(152 mg, 4.02 mmol) in 1 mL water was added. The mixture was stirred for 30 min at rt. Upon completion 1 mL of concentrated HCL was slowly added and then concentrated to remove THF. The mixture was then neutralized with saturated NaHCO3and extracted with EtOAc 3×. The organic extract was washed with brine and dried over sodium sulfate. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 177.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.65 (dd, J=2.0, 0.9 Hz, 1H), 8.00 (s, 1H), 7.85 (dd, J=8.1, 2.2 Hz, 1H), 7.75-7.67 (m, 2H), 4.81 (s, 2H). Preparation of Intermediate I-1007 2-imidazol-1-ylpyrimidine-5-carbaldehyde: A suspension of 2-chloropyrimidine-5-carbaldehyde (250 mg, 1.75 mmol), imidazole (150 mg, 2.21 mmol), and potassium carbonate (339 mg, 2.46 mmol) in DMF (10 mL) was heated at 50° C. overnight. The next day the mixture was diluted with EtOAc and washed with 5% LiCl (2×) and brine. The organic extract was dried over sodium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 175.2 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 2H), 8.70 (t, J=1.1 Hz, 1H), 8.03 (t, J=1.4 Hz, 1H), 7.16 (t, J=1.3 Hz, 1H). (2-imidazol-1-ylpyrimidin-5-yl)methanol (I-1007): sodium borohydride (23.3 mg, 0.616 mmol) was added to a solution of 2-imidazol-1-ylpyrimidine-5-carbaldehyde (107 mg, 0.616 mmol) in MeOH (5 mL) at 0° C. The mixture was gradually warmed to rt and stirred for 4 hr. Upon completion of time, the mixture was quenched with 1 mL water, diluted with EtOAc and concentrated to dryness to remove all MeOH. Then the mixture was diluted with EtOAc and washed with brine. Aqueous layer was extracted once more with EtOAc. The combined organic extracts were dried over sodium sulfate to give desired product. ES/MS: 177.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.77 (s, 1H), 8.75 (s, 2H), 7.97 (s, 1H), 7.26 (s, 1H), 4.84 (s, 2H). Preparation of Intermediate I-1008 [4-methoxy-6-(triazol-1-yl)-3-pyridyl]methanol (I-1008): [4-methoxy-6-(triazol-1-yl)-3-pyridyl]methanol was prepared in a manner as described for Intermediate I-1004 substituting 1H-triazole for 1H-1,2,4-triazole and substituting methyl 6-chloro-4-methoxy-pyridine-3-carboxylate for methyl 6-chloropyridine-3-carboxylate. ES/MS: 207.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.62 (d, J=1.2 Hz, 1H), 8.33 (s, 1H), 7.86 (d, J=1.1 Hz, 1H), 7.79 (s, 1H), 4.77 (s, 2H), 4.08 (s, 3H). Preparation of Intermediate I-1009 4-fluoro-5-methyl-pyridine-2-carbonitrile: 2-chloro-4-fluoro-5-methyl-pyridine (3.0 g, 21 mmol), zinc cyanide (1452 mg, 12 mmol), zinc (135 mg, 2.1 mmol), and 1,1′-Bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (852 mg, 1.0 mmol) were taken up in DMF (20 mL). The resulting mixture was sparged with argon for 5 minutes then heated to 100° C. in a sealed 20 mL MW vial for 3 hr. The reaction was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. 5-(bromomethyl)-4-fluoro-pyridine-2-carbonitrile: 4-fluoro-5-methyl-pyridine-2-carbonitrile (2.16 g, 15.9 mmol) was taken up in carbon tetrachloride (30.0 mL) and N-Bromosuccinimide (3389 mg, 19.0 mmol) was added followed by Benzoyl peroxide (231 mg, 0.952 mmol). The mixture was heated to 90° C. for 5 hr. Upon completion of time the mixture was filtered to remove succinimide (washed with DCM) and conc. in vacuo. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 215, 217 (M+H+). 5-[(6-bromo-2-pyridyl)oxymethyl]-4-fluoro-pyridine-2-carbonitrile: 6-bromopyridin-2-ol (1.3 g, 7.5 mmol) was taken up in acetonitrile (15 mL) and 5-(bromomethyl)-4-fluoro-pyridine-2-carbonitrile (1767 mg, 8.2 mmol) and cesium carbonate (3478 mg, 11 mmol) were added. The mixture was heated to 65° C. for 45 min. Next, the mixture was filtered through Celite and the filtrate was concentrated to dryness. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 308, 310 (M+H+). [5-[(6-bromo-2-pyridyl)oxymethyl]-4-fluoro-2-pyridyl]methanamine: To a solution of 5-[(6-bromo-2-pyridyl)oxymethyl]-4-fluoro-pyridine-2-carbonitrile (102 mg, 0.331 mmol) in THF (3 mL) at 0° C. was added diisobutylaluminium hydride (1000 mmol/L, 0.993 mL, 0.993 mmol). The resulting mixture was gradually warmed to rt. Additional aliquots of Dibal-H were added until the mixture no longer progressed (2×0.5 mL added). The mixture was then cooled to 0° C. and diluted with Et2O. Next, a solution of 100 uL of water, 100 uL 15% aqueous NaOH, and 250 uL water was slowly added to the mixture. The resulting mixture was warmed to rt and stirred for 15 min. Upon completion of time MgSO4was added the resulting mixture was stirred for 15 min, then filtered and concentrated to give desired product, which was carried onto the next step without purification. ES/MS: 312, 314 (M+H+). N-[[5-[(6-bromo-2-pyridyl)oxymethyl]-4-fluoro-2-pyridyl]methyl]acetamide (I-1009): To a suspension of [5-[(6-bromo-2-pyridyl)oxymethyl]-4-fluoro-2-pyridyl]methanamine (56.0 mg, 0.179 mmol) and pyridine (0.0217 mL, 0.269 mmol) in DCM (5 mL) at 0° C., acetic anhydride (0.0200 mL, 0.212 mmol) was added. The mixture was gradually warmed to rt and stirred overnight. Next, the organic extract was washed with brine and dried over sodium sulfate. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 354, 356 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.70 (d, J=9.6 Hz, 1H), 7.53-7.36 (m, 1H), 7.12 (d, J=7.5 Hz, 1H), 7.06 (d, J=9.8 Hz, 1H), 6.75 (d, J=8.1 Hz, 1H), 6.65 (s, 1H), 5.45 (s, 2H), 4.57 (d, J=5.1 Hz, 2H), 2.10 (s, 3H). Preparation of Intermediate I-1010 5-[(6-bromo-2-pyridyl)oxymethyl]-N-(2-cyanoethyl)pyridine-2-carboxamide (I-1010): 5-[(6-bromo-2-pyridyl)oxymethyl]-N-(2-cyanoethyl)pyridine-2-carboxamide was prepared in a manner as described for Intermediate I-28 substituting 3-aminopropanenitrile for 1-aminocyclopropanecarbonitrile hydrochloride. ES/MS: 361.0, 363.0 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.68 (d, J=2.0 Hz, 1H), 8.46 (d, J=6.7 Hz, 1H), 8.20 (d, J=8.0 Hz, 1H), 7.98 (dd, J=8.0, 2.1 Hz, 1H), 7.48 (t, J=7.8 Hz, 1H), 7.13 (d, J=7.5 Hz, 1H), 6.79 (d, J=8.1 Hz, 1H), 5.48 (s, 2H), 3.78 (q, J=6.5 Hz, 2H), 2.77 (t, J=6.5 Hz, 2H). Preparation of Intermediate I-1011 (4-bromo-2-fluoro-3-methyl-phenyl)methanol: To a solution of methyl 4-bromo-2-fluoro-3-methyl-benzoate (500 mg, 2.02 mmol) in THE (10 mL) at 0° C., diisobutylaluminum hydride (1000 mmol/L, 5.06 mL, 5.06 mmol) was added. The mixture was stirred for 1 hr. Next, the mixture was diluted with 25 mL Et2O and cooled to 0° C. Then 0.200 mL water, 0.200 mL 15% NaOH added and then an additional 0.500 mL water was added. The resulting mixture was warmed to rt and stirred for 15 min. Upon completion, MgSO4was added and the mixture was stirred for 15 min, then filtered and rinsed with Et2O to give crude residue which was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. 1H NMR (400 MHz, Chloroform-d) δ 7.37 (dd, J=8.2, 1.3 Hz, 1H), 7.16 (t, J=7.9 Hz, 1H), 4.73 (s, 2H), 2.36 (d, J=2.5 Hz, 3H). 1-bromo-4-(bromomethyl)-3-fluoro-2-methyl-benzene: To a solution of (4-bromo-2-fluoro-3-methyl-phenyl)methanol (358 mg, 1.63 mmol) and (4-diphenylphosphanylphenyl) polymer bound (78.7%, 651 mg, 1.966 mmol), in DCM (10 mL) at 0° C., carbon tetrabromide (650 mg, 1.96 mmol) was added. The mixture was then gradually warmed to rt overnight. The suspension was filtered, diluted with DCM and washed with brine. The organic extract was dried over sodium sulfate, concentrated and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. 1H NMR (400 MHz, Chloroform-d) δ 7.35 (dd, J=8.3, 1.3 Hz, 1H), 7.12 (t, J=8.0 Hz, 1H), 4.49 (d, J=1.2 Hz, 2H), 2.37 (d, J=2.5 Hz, 3H). 2-(4-bromo-2-fluoro-3-methyl-phenyl)acetonitrile: To a solution of 1-bromo-4-(bromomethyl)-3-fluoro-2-methyl-benzene (299 mg, 1.06 mmol) in DMSO (6 ml) at rt, potassium cyanide (138 mg, 2.12 mmol) was added. The mixture was then stirred at rt overnight. Next, the mixture was diluted with EtOAc and washed with 10% Na2CO3. The organic extract was washed once more with brine. Next, organic extract was dried over sodium sulfate to give desired product. 1H NMR (400 MHz, Chloroform-d) δ 7.41 (dd, J=8.3, 1.4 Hz, 1H), 7.17 (t, J=8.0 Hz, 1H), 3.73 (d, J=1.0 Hz, 2H), 2.37 (d, J=2.6 Hz, 3H). 2-(4-bromo-2-fluoro-3-methyl-phenyl)acetic acid (I-1011): A solution of 2-(4-bromo-2-fluoro-3-methyl-phenyl)acetonitrile (164 mg, 0.717 mmol) in concentrated HCl (2 mL) was heated at 100° C. overnight. Next, the mixture was cooled to rt, diluted with EtOAc and neutralized carefully with 6 NaOH (4 mL). The organic extract was dried organic extract over sodium sulfate, filtered and concentrated to give desired product. 1H NMR (400 MHz, Methanol-d4) δ 7.35 (dd, J=8.3, 1.3 Hz, 1H), 7.08 (t, J=8.0 Hz, 1H), 3.64 (d, J=1.8 Hz, 2H), 2.35 (d, J=2.5 Hz, 3H). Preparation of Intermediate I-1012 Methyl 2-[(4-bromo-2-fluoro-3-methyl-phenyl)methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1012): Methyl 2-[(4-bromo-2-fluoro-3-methyl-phenyl)methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-4 for I-1 and 2-(4-bromo-2-fluoro-3-methyl-phenyl)acetic acid (I-1011) for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 447.0, 449.0 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.11 (d, J=1.7 Hz, 1H), 7.98 (dd, J=8.5, 1.6 Hz, 1H), 7.75 (d, J=8.5 Hz, 1H), 7.29 (dd, J=8.3, 1.3 Hz, 1H), 6.99 (t, J=8.0 Hz, 1H), 5.14 (qd, J=6.8, 2.9 Hz, 1H), 4.72-4.51 (m, 1H), 4.51-4.27 (m, 5H), 3.96 (s, 3H), 2.73 (dtd, J=11.4, 8.1, 6.1 Hz, 1H), 2.51-2.38 (m, 1H), 2.35 (d, J=2.6 Hz, 3H). Preparation of Intermediate I-1013 [4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-phenyl]methanamine: To a solution of 4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (257 mg, 0.837 mmol) in THF (12 mL) at 0° C., diisobutylaluminium hydride (1000 mmol/L, 6.28 mL, 6.28 mmol) was added. The mixture was then gradually warmed to rt and stirred overnight. Next, the mixture was cooled to 0° C. and diluted with Et2O. Next, 250 uL of water was slowly added, followed by the addition of 250 uL 15% aq. NaOH, followed by an additional 625 uL water. The resulting mixture was warmed to rt and stirred for 15 min. Upon completion of time MgSO4was added and the mixture was stirred 15 min, then filtered and concentrated to give desired product, which was carried onto the next step without purification. ES/MS: 312.1, 314.1 (M+H+). N-[[4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-phenyl]methyl]acetamide (I-1013): To a suspension of [4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-phenyl]methanamine (194 mg, 0.623 mmol) and Pyridine (0.0753 mL, 0.935 mmol) in DCM (5 mL) at 0° C., acetic anhydride (0.0695 mL, 0.736 mmol) was added. The resulting mixture was gradually warmed to rt and stirred overnight. The organic extract was washed with brine and dried over sodium sulfate. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 353.1, 355.1 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.50 (t, J=7.6 Hz, 1H), 7.47-7.41 (m, 1H), 7.10 (dd, J=7.7, 3.9 Hz, 2H), 7.05 (d, J=10.6 Hz, 1H), 6.74 (d, J=8.1 Hz, 1H), 5.77 (s, 1H), 5.42 (s, 2H), 4.46 (d, J=5.8 Hz, 2H), 2.07 (d, J=1.0 Hz, 3H). Preparation of Intermediate I-1014 6-(bromomethyl)-1-methyl-benzotriazole (I-1014): 6-(bromomethyl)-1-methyl-benzotriazole was prepared in a manner as described for Intermediate I-36 substituting 3-methylbenzotriazole-5-carboxylic acid for [5-(difluoromethyl)thiazole-2-carbonyl]oxysodium. ES/MS: 226.2, 228.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.05 (dd, J=8.6, 0.8 Hz, 1H), 7.58 (dd, J=1.5, 0.8 Hz, 1H), 7.43 (dd, J=8.6, 1.5 Hz, 1H), 4.69 (s, 2H), 4.33 (s, 3H). Preparation of Intermediate I-1015 Tert-butyl 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline-2-carboxylate: A suspension of tert-butyl 5-(bromomethyl)isoindoline-2-carboxylate (510 mg, 1.6 mmol), 6-bromopyridin-2-ol (280 mg, 1.6 mmol), and silver carbonate (949 mg, 3.4 mmol) in CH3CN (10 mL) was heated at 50° C. overnight. The mixture was diluted with EtOAc and brine. Then the mixture was filtered over Celite frit, partitioned. The organic extract was next washed with brine once more. The organic extract was then dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 349, 351 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.53-7.42 (m, 1H), 7.39 (d, J=8.1 Hz, 1H), 7.28 (s, 1H), 7.10 (d, J=7.4 Hz, 1H), 6.75 (d, J=8.1 Hz, 1H), 5.38 (s, 2H), 4.70 (d, J=12.8 Hz, 4H), 1.54 (s, 9H). 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline; 2,2,2-trifluoroacetic acid: A solution of tert-butyl 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline-2-carboxylate (399 mg, 0.98 mmol) and TFA (0.38 mL, 4.9 mmol) in DCM (5 mL) was stirred at rt overnight. The mixture was then concentrated to dryness and carried onto the next step without purification. ES/MS: 305, 307 (M+H+). 1-[5-[(6-bromo-2-pyridyl)oxymethyl]isoindolin-2-yl]ethenone (I-1015): To a solution of 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline; 2,2,2-trifluoroacetic acid (206 mg, 0.491 mmol) and Pyridine (0.0792 mL, 0.983 mmol)) in DCM (5 mL) at 0° C., acetic anhydride (0.0548 mL, 0.580 mmol) was added. The mixture was gradually warmed to rt and stirred overnight. Next, the mixture was diluted with EtOAc, washed with brine, dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 347, 349 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.49-7.37 (m, 3H), 7.31 (dd, J=17.0, 7.8 Hz, 1H), 7.10 (d, J=7.5 Hz, 1H), 6.76 (dd, J=8.2, 3.4 Hz, 1H), 5.38 (s, 2H), 4.88-4.71 (m, 4H), 2.19 (s, 3H). Preparation of Intermediate I-1016 Methyl 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline-2-carboxylate (I-1016): Methyl 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline-2-carboxylate was prepared in a manner as described for Intermediate I-1015 substituting methyl chloroformate for acetic anhydride. ES/MS: 363, 365 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.45 (dd, J=8.2, 7.5 Hz, 1H), 7.42-7.34 (m, 2H), 7.34-7.23 (m, 1H), 7.10 (dd, J=7.5, 0.7 Hz, 1H), 6.75 (dd, J=8.2, 1.2 Hz, 1H), 5.38 (s, 2H), 4.75 (dt, J=23.3, 2.8 Hz, 4H), 3.81 (s, 3H). Preparation of Intermediate I-1018 Methyl 2-(chloromethyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-benzimidazole-5-carboxylate: To a solution of methyl 4-amino-3-[(4,4-dimethyltetrahydrofuran-3-yl)amino]-5-fluoro-benzoate 1-107 (1.13 g, 4.00 mmol) in CH3CN (20 mL), p-Toluenesulfonic Acid, monohydrate (0.0345 g, 0.200 mmol), and 2-Chloro-1,1,1-trimethoxyethane (0.547 mL, 5.20 mmol) were added. The mixture was heated at 60° C. overnight. Upon completion, more 2-Chloro-1,1,1-trimethoxyethane (0.547 mL, 5.20 mmol) and p-Toluenesulfonic Acid, monohydrate (0.0345 g, 0.200 mmol) were added and then the mixture was heated at 60° C. for 3 hr. Upon completion, the mixture was diluted with EtOAc washed with brine, dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 341.2 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.41 (s, 1H), 7.71 (dd, J=10.6, 1.4 Hz, 1H), 4.96 (d, J=12.6 Hz, 1H), 4.90-4.76 (m, 2H), 4.61 (dd, J=11.2, 1.9 Hz, 1H), 4.50 (dd, J=11.2, 7.0 Hz, 1H), 3.97 (d, J=1.2 Hz, 4H), 3.83 (d, J=9.3 Hz, 1H), 1.43 (s, 3H), 0.73 (s, 3H). Methyl 2-[(4-bromo-5-fluoro-2-oxo-1-pyridyl)methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-benzimidazole-5-carboxylate (I-1018): A suspension of 4-bromo-5-fluoro-1H-pyridin-2-one (115 mg, 0.599 mmol) methyl 2-(chloromethyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-benzimidazole-5-carboxylate (205 mg, 0.602 mmol), and potassium carbonate (414 mg, 3.00 mmol) in DMF (3 mL) was heated at 60° C. for 24 hr. Next, the mixture was cooled to rt, diluted with EtOAc, filtered, and concentrated. The mixture was diluted with EtOAc and washed with 5% LiCl and brine. the mixture was dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. Submitted for chiral-SFC separation (SFC IG column with EtOH cosolvent) gave 2 distinct stereoisomers. ES/MS: 497, 499 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.45 (s, 1H), 7.82 (d, J=3.9 Hz, 1H), 7.72 (dd, J=10.8, 1.2 Hz, 1H), 6.93 (d, J=6.4 Hz, 1H), 5.73 (d, J=14.8 Hz, 1H), 5.28 (d, J=6.6 Hz, 1H), 5.11 (d, J=14.8 Hz, 1H), 4.69-4.36 (m, 2H), 3.97 (s, 3H), 3.90-3.72 (m, 2H), 1.43 (s, 3H), 0.61 (s, 3H). Preparation of Intermediate I-1019 and I-1020 Methyl 2-[(4-bromo-5-fluoro-2-oxo-1-pyridyl)methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-benzimidazole-5-carboxylate: Methyl 2-[(4-bromo-5-fluoro-2-oxo-1-pyridyl)methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-benzimidazole-5-carboxylate was submitted for chiral-SFC separation (SFC IG column with EtOH cosolvent) gave 2 distinct stereoisomers. Methyl 2-[(4-bromo-5-fluoro-2-oxo-1-pyridyl)methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-benzimidazole-5-carboxylate (I-1019 isomer 1): RT 2.46 min. ES/MS: 496, 498 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.46 (s, 1H), 7.82 (d, J=3.9 Hz, 1H), 7.73 (dd, J=10.8, 1.2 Hz, 1H), 6.94 (d, J=6.4 Hz, 1H), 5.74 (d, J=14.8 Hz, 1H), 5.29 (d, J=6.5 Hz, 1H), 5.10 (d, J=14.8 Hz, 1H), 4.68-4.45 (m, 2H), 3.97 (s, 3H), 3.93-3.77 (m, 2H), 1.44 (s, 3H), 0.62 (s, 3H). Methyl 2-[(4-bromo-5-fluoro-2-oxo-1-pyridyl)methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-benzimidazole-5-carboxylate (I-1020 isomer 2): RT 5.88 min. ES/MS: 496, 498 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.46 (s, 1H), 7.82 (d, J=3.9 Hz, 1H), 7.73 (dd, J=10.8, 1.2 Hz, 1H), 6.94 (d, J=6.4 Hz, 1H), 5.74 (d, J=14.8 Hz, 1H), 5.29 (d, J=6.5 Hz, 1H), 5.10 (d, J=14.8 Hz, 1H), 4.68-4.45 (m, 2H), 3.97 (s, 3H), 3.93-3.77 (m, 2H), 1.44 (s, 3H), 0.62 (s, 3H). Preparation of Intermediate I-1021 [5-[(6-bromo-2-pyridyl)oxymethyl]isoindolin-2-yl]-cyclopropyl-methanone (I-1021): [5-[(6-bromo-2-pyridyl)oxymethyl]isoindolin-2-yl]-cyclopropyl-methanone was prepared in a manner as described for Intermediate I-1015 substituting cyclopropane carboxylic acid chloride for acetic anhydride. ES/MS: 373, 375 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.49-7.37 (m, 3H), 7.32 (t, J=8.7 Hz, 1H), 7.11 (d, J=7.5 Hz, 1H), 6.76 (d, J=8.1 Hz, 1H), 5.39 (s, 2H), 5.10-4.91 (m, 2H), 4.84 (d, J=5.8 Hz, 2H), 1.75 (dq, J=8.2, 4.6, 4.1 Hz, 1H), 1.09 (dt, J=4.8, 3.3 Hz, 2H), 0.87 (dt, J=7.9, 3.4 Hz, 2H). Preparation of Intermediate I-1022 5-[(6-bromo-2-pyridyl)oxymethyl]-2-methylsulfonyl-isoindoline (I-1022): A suspension of 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline; 2,2,2-trifluoroacetic acid (206 mg, 0.491 mmol) was washed with saturated NaHCO3. The mixture was then dried with sodium sulfate to give free amine, which was then dissolved in DCM (5 mL). The solution was cooled to 0° C. Next, pyridine (0.0292 mL, 0.363 mmol) was added, followed by a solution of methanesulfonyl chloride (1.00 mmol/L, 174 mL, 0.174 mmol). The resulting mixture was gradually warmed to rt and stirred overnight. Next, the mixture was diluted with EtOAc and washed with brine. The mixture was then dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 383, 385 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.49-7.37 (m, 3H), 7.29 (d, J=7.6 Hz, 1H), 7.11 (dd, J=7.5, 0.7 Hz, 1H), 6.76 (dd, J=8.1, 0.7 Hz, 1H), 5.39 (s, 2H), 4.74 (t, J=2.5 Hz, 4H), 2.90 (s, 3H). Preparation of Intermediate I-1023 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid (I-1023): To a solution of 4-bromo-2-chloro-5-methyl-benzoic acid (996 mg, 3.99 mmol) in DCM (10 mL) at 0° C., oxalyl dichloride (2000 mmol/L, 2.40 mL, 4.79 mmol) and 5 drops of DMF were added. The resulting mixture was stirred for 1 hr., then 10 mL MeOH was added and the resulting mixture was stirred overnight at rt. Next, the mixture was diluted with DCM, washed with saturated NaHCO3, dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. 1H NMR (400 MHz, Chloroform-d) δ 7.74 (s, 1H), 7.68 (d, J=1.1 Hz, 1H), 3.94 (d, J=1.2 Hz, 3H), 2.42 (s, 3H). (4-bromo-2-chloro-5-methyl-phenyl)methanol: To a solution of methyl 4-bromo-2-chloro-5-methyl-benzoate (931 mg, 3.53 mmol) in THE (15 mL) at 0° C., diisobutylaluminium hydride (1000 mmol/L, 8.83 mL, 8.83 mmol) was added. The mixture was then stirred for 1 hr. Next, the mixture was diluted with 25 mL Et2O and cooled to 0° C. Next, 0.350 mL water, and 0.350 mL 15% NaOH were added, then an additional 0.900 mL water was added. The mixture was then warmed to rt and stirred for 15 min. Next, MgSO4was added and the resulting mixture was stirred 15 min, filtered and rinsed with Et2O to give crude residue which was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. 1H MR (400 MHz, Chloroform-d) δ 7.56 (d, J=1.4 Hz, 1H), 7.38 (s, 1H), 4.73 (s, 2H), 2.41 (d, J=1.3 Hz, 4H). 1-bromo-4-(bromomethyl)-5-chloro-2-methyl-benzene: To a solution of (4-bromo-2-chloro-5-methyl-phenyl)methanol (639 mg, 2.71 mmol) and (4-diphenylphosphanylphenyl) polymer bound (78.7%, 1081 mg, 3.26 mmol), in DCM (10 mL) at 0° C., carbon tetrabromide (1080 mg, 3.26 mmol) was added. The mixture was then gradually warmed to rt overnight. Next, the mixture was filtered, diluted with DCM and washed with brine. The organic extract was dried over sodium sulfate, concentrated and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. 1H NMR (400 MHz, Chloroform-d) δ 7.60 (s, 1H), 7.32 (s, 1H), 4.53 (s, 2H), 2.39 (s, 3H). 2-(4-bromo-2-chloro-5-methyl-phenyl)acetonitrile: To a solution of 1-bromo-4-(bromomethyl)-5-chloro-2-methyl-benzene (721 mg, 2.42 mmol) in DMSO (12 ml) at rt, potassium cyanide (315 mg, 4.83 mmol) was added. The mixture was then stirred at rt overnight. The mixture was diluted with EtOAc and washed with 10% Na2CO3. The organic extract was washed once more with brine, then dried over sodium sulfate to give desired product. 1H NMR (400 MHz, Chloroform-d) δ 7.62 (s, 1H), 7.52-7.33 (m, 1H), 3.78 (s, 2H), 2.42 (s, 3H). 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid (I-1023): A solution of 2-(4-bromo-2-chloro-5-methyl-phenyl)acetonitrile (179 mg, 0.732 mmol) in concentrated HCl (5 mL) was heated at 100° C. overnight. The mixture was then cooled to rt, and diluted with EtOAc, and neutralized carefully with 6 N NaOH (10 mL). Next the organic extract was dried over sodium sulfate to give desired product. 1H NMR (400 MHz, Methanol-d4) δ 7.59 (s, 1H), 7.29 (s, 1H), 3.71 (s, 2H), 2.37 (s, 3H). Preparation of Intermediate I-1024 Methyl 2-[(4-bromo-2-chloro-5-methyl-phenyl)methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1024): methyl 2-[(4-bromo-2-chloro-5-methyl-phenyl)methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-4 for I-1 and 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid (I-1023) for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 463, 465 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.12 (d, J=1.6 Hz, 1H), 8.00 (dd, J=8.5, 1.4 Hz, 1H), 7.78 (d, J=8.5 Hz, 1H), 7.62 (s, 1H), 7.13 (s, 1H), 5.13 (dd, J=7.1, 2.9 Hz, 1H), 4.77-4.59 (m, 1H), 4.59-4.23 (m, 5H), 3.97 (d, J=1.0 Hz, 3H), 2.74 (ddt, J=14.1, 11.7, 6.6 Hz, 1H), 2.52-2.35 (m, 1H), 2.29 (s, 3H). Preparation of Intermediate I-1025 2-[2,5-difluoro-4-[6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]acetic acid (I-1025): 2-[2,5-difluoro-4-[6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]acetic acid was prepared in a manner as described for Intermediate I-7 substituting methyl 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline-2-carboxylate (I-1016) for I-3. ES/MS: 455.1 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 7.82-7.65 (m, 2H), 7.53-7.45 (m, 2H), 7.44 (d, J=7.3 Hz, 2H), 7.32 (dd, J=10.9, 8.0 Hz, 1H), 7.23-7.09 (m, 1H), 6.85 (d, J=8.2 Hz, 1H), 5.48 (s, 2H), 4.71 (d, J=4.2 Hz, 4H), 3.78 (s, 3H), 3.73 (d, J=1.3 Hz, 2H). Preparation of Intermediate I-1026 Ethyl 4-[[2-(4-bromo-2-chloro-5-methyl-phenyl)acetyl]amino]-3-fluoro-5-[[(2S)-oxetan-2-yl]methylamino]benzoate: To a solution of 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid (501 mg, 1.90 mmol) and ethyl 4-amino-3-fluoro-5-[[(2S)-oxetan-2-yl]methylamino]benzoate (510 mg, 1.90 mmol) in CH3CN (19 mL) at 0° C., 1-Methylimidazole (0.758 mL, 9.50 mmol) was added, followed by the addition of TCFH (640 mg, 2.28 mmol). The mixture was stirred at rt overnight. Next, the mixture was diluted with EtOAc, washed with saturated NH4C1, 1N NaOH, and brine, and then subsequently dried over sodium sulfate, concentrated, and carried onto next step below. ES/MS: 513, 515 (M+H+). Ethyl 2-[(4-bromo-2-chloro-5-methyl-phenyl)methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1026): A solution of ethyl 4-[[2-(4-bromo-2-chloro-5-methyl-phenyl)acetyl]amino]-3-fluoro-5-[[(2S)-oxetan-2-yl]methylamino]benzoate (977 mg, 1.90 mmol) and acetic acid (3426 mg, 57.0 mmol) in DCE (15 mL) was heated at 60° C. overnight. The mixture was cooled to rt, diluted with EtOAc and carefully neutralized with NaHCO3(4.78 g). The organic layer was washed with brine and dried over sodium sulfate and concentrated. Purification by chromatography (eluent: EtOAc/hexanes) gave desired product. ES/MS: 495, 497 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.94 (d, J=1.3 Hz, 1H), 7.69 (dd, J=11.0, 1.3 Hz, 1H), 7.62 (s, 1H), 7.12 (s, 1H), 5.08 (qd, J=6.9, 2.8 Hz, 1H), 4.74-4.59 (m, 1H), 4.59-4.24 (m, 8H), 2.73 (dtd, J=11.4, 8.1, 6.1 Hz, 1H), 2.39 (ddt, J=11.5, 9.0, 7.2 Hz, 1H), 2.29 (s, 3H), 1.44 (t, J=7.2 Hz, 3H). Preparation of Intermediate I-1027 2-[2,5-difluoro-4-[6-[(2-methylsulfonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]acetic acid (I-1027): 2-[2,5-difluoro-4-[6-[(2-methylsulfonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]acetic acid was prepared in a manner as described for Intermediate I-7 substituting 5-[(6-bromo-2-pyridyl)oxymethyl]-2-methylsulfonyl-isoindoline (I-1022) for 1-3. ES/MS: 475.0 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 7.75 (t, J=7.8 Hz, 1H), 7.67 (dd, J=10.7, 6.5 Hz, 1H), 7.53-7.43 (m, 3H), 7.33 (d, J=7.8 Hz, 1H), 7.19 (dd, J=11.9, 6.2 Hz, 1H), 6.83 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 4.71 (d, J=4.7 Hz, 5H), 3.56 (s, 2H), 2.92 (s, 3H). Preparation of Intermediate I-1028 Tert-butyl 5-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]isoindoline-2-carboxylate: A suspension of 6-bromo-2-chloro-3-fluoro-pyridine (1000 mg, 4.75 mmol), tert-butyl 5-(hydroxymethyl)isoindoline-2-carboxylate (1.18 g, 4.75 mmol), and cesium carbonate (3.10 g, 9.50 mmol) in CH3CN (20 mL) was heated at 60° C. for 30 hr. Upon completion the solid was filtered off, the filtrate was diluted with EtOAc and washed with brine. Next, the filtrate was dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 366.8, 368.8 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.41 (q, J=7.9, 6.4 Hz, 2H), 7.33-7.18 (m, 2H), 7.04 (dd, J=8.1, 2.7 Hz, 1H), 5.45 (s, 2H), 4.78-4.63 (m, 4H), 1.54 (s, 9H). 5-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]isoindoline; 2,2,2-trifluoroacetic acid: A solution of tert-butyl 5-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]isoindoline-2-carboxylate (449 mg, 1.1 mmol) and TFA (0.81 mL, 11 mmol) in DCM (5 mL) was stirred at rt overnight. The mixture was concentrated to dryness and carried onto the next step without purification. ES/MS: 323.2, 325.2 (M+H+). Methyl 5-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]isoindoline-2-carboxylate (I-1028): To a solution of 5-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]isoindoline; 2,2,2-trifluoroacetic acid (464 mg, 1.06 mmol) and N,N-diisopropylethylamine (0.370 mL, 2.12 mmol) in DCM (10 mL) at 0° C., methyl chloroformate (0.107 mL, 1.38 mmol) was added. The resulting mixture was gradually warmed to rt and stirred overnight. Next, the mixture was diluted with EtOAc and washed with brine. Dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 381, 383 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 7.48-7.37 (m, 2H), 7.31 (d, J=7.8 Hz, 1H), 7.25 (dd, J=9.3, 8.1 Hz, 1H), 7.05 (dd, J=8.1, 2.7 Hz, 1H), 5.46 (s, 2H), 4.83-4.66 (m, 4H), 3.81 (s, 3H). Preparation of Intermediate I-1029 2-[2,5-difluoro-4-[5-fluoro-6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]acetic acid (I-1029): 2-[2,5-difluoro-4-[5-fluoro-6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]acetic acid was prepared in a manner as described for Intermediate I-7 substituting methyl 5-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]isoindoline-2-carboxylate (I-1028) for I-3. ES/MS: 473.0 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 7.68 (dd, J=10.7, 6.4 Hz, 1H), 7.57 (dd, J=9.9, 8.2 Hz, 1H), 7.53-7.41 (m, 3H), 7.40-7.29 (m, 1H), 7.22 (dd, J=11.6, 6.1 Hz, 1H), 5.56 (s, 2H), 4.71 (d, J=5.2 Hz, 4H), 3.78 (s, 3H), 3.73 (s, 2H). Preparation of Intermediate I-1030 Methyl 2-[(4-bromo-2-chloro-5-methyl-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1030): Methyl 2-[(4-bromo-2-chloro-5-methyl-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting 1-80 for I-1 and 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid (I-1023) for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 490.6, 492.6 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.56 (s, 1H), 8.02 (dd, J=8.5, 1.4 Hz, 1H), 7.80 (d, J=8.5 Hz, 1H), 7.64 (s, 1H), 7.06 (s, 1H), 4.60-4.47 (m, 2H), 4.45-4.26 (m, 3H), 3.95 (d, J=10.4 Hz, 5H), 3.78 (d, J=8.8 Hz, 1H), 2.28 (s, 3H), 2.07 (s, 1H), 1.31 (s, 3H), 0.67 (s, 3H). Preparation of Intermediate I-1031 Methyl 4-[[2-(4-bromo-2-chloro-5-methyl-phenyl)acetyl]amino]-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-5-fluoro-benzoate: To a solution of 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid (399 mg, 1.51 mmol) and methyl 4-amino-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-5-fluoro-benzoate (427 mg, 1.51 mmol) in CH3CN (7 mL) at 0° C., 1-Methylimidazole (0.603 mL, 7.56 mmol) was added, followed by the addition of TCFH (509 mg, 1.82 mmol). The resulting mixture was stirred at rt overnight. Next, the mixture was diluted with EtOAc and washed with saturated NH4C1, 1N NaOH, and brine. Next, the mixture was dried over sodium sulfate, concentrated and carried onto next step below without purification. ES/MS: 527, 529 (M+H+). Methyl 2-[(4-bromo-2-chloro-5-methyl-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluoro-benzimidazole-5-carboxylate (I-1031): To a solution of methyl 4-[[2-(4-bromo-2-chloro-5-methyl-phenyl)acetyl]amino]-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-5-fluoro-benzoate (798 mg, 1.51 mmol) and Triphenylphosphine oxide (1262 mg, 4.54 mmol) in DCM (15 mL) at 0° C., Triflic anhydride (1000 mmol/L, 2.27 mL, 2.27 mmol) was added. The resulting mixture was gradually warmed to rt and stirred for 1 hr. Next, the mixture was diluted with DCM and washed with saturated NaHCO3solution and brine. The mixture was then dried over sodium sulfate, concentrated and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 508.6, 510.6 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.38 (s, 1H), 7.71 (dd, J=10.8, 1.2 Hz, 1H), 7.64 (s, 1H), 7.06 (s, 1H), 4.55-4.42 (m, 2H), 4.41 (d, J=2.8 Hz, 2H), 4.32 (dd, J=11.0, 7.0 Hz, 1H), 3.97 (s, 3H), 3.92 (d, J=8.8 Hz, 1H), 3.77 (d, J=8.8 Hz, 1H), 2.27 (s, 3H), 1.29 (s, 3H), 0.64 (s, 3H). Preparation of Intermediate I-1032 Ethyl (S)-4-amino-3-fluoro-5-((oxetan-2-ylmethyl)amino)benzoate (I-1032): Ethyl (S)-4-amino-3-fluoro-5-((oxetan-2-ylmethyl)amino)benzoate was prepared in a manner as described for Intermediate I-1 substituting ethyl 3,5-difluoro-4-nitrobenzoate for methyl 3-fluoro-4-nitro-benzoate. ES/MS: 257.2 (M+H+); ′H NMR (400 MHz, CDCl3) δ 7.32 (dd, J=10.8, 1.7 Hz, 1H), 7.20 (t, J=1.4 Hz, 1H), 4.35 (q, J=7.1 Hz, 2H), 3.78 (s, 3H), 3.68 (dd, J=5.6, 4.5 Hz, 2H), 3.43 (s, 3H), 3.35 (dd, J=5.6, 4.5 Hz, 2H), 1.39 (t, J=7.1 Hz, 3H). Preparation of Intermediate I-1033 Ethyl 2-(4-bromo-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1033): Ethyl 2-(4-bromo-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-2 substituting ethyl 4-amino-3-fluoro-5-((2-methoxyethyl)amino)benzoate (I-1032) for I-1 and 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 471.0, 473.0 (M+H+). 1H NMR (400 MHz, CDCl3) δ 7.91 (d, J=1.3 Hz, 1H), 7.70 (dd, J=10.8, 1.3 Hz, 1H), 7.34 (dd, J=8.7, 5.5 Hz, 1H), 7.12 (dd, J=8.6, 6.3 Hz, 1H), 4.44 (q, J=7.1 Hz, 2H), 4.39 (s, 2H), 4.37-4.29 (m, 2H), 3.66 (t, J=5.1 Hz, 2H), 3.25 (s, 3H), 1.45 (t, J=7.1 Hz, 3H). Preparation of Intermediate I-1034 q6-[(6-bromo-2-pyridyl)oxymethyl]-1-methyl-benzotriazole (I-1034): A suspension of 6-(bromomethyl)-1-methyl-benzotriazole (62 mg, 0.28 mmol), 6-bromopyridin-2-ol (60 mg, 0.34 mmol), and silver carbonate (203 mg, 0.74 mmol) in CH3CN (5 mL) was heated at 50° C. overnight. Next, the mixture was diluted with EtOAc and brine. The mixture was then filtered over Celite frit and partitioned. The partitioned layers were washed and separated. The organic layer was washed once more with brine, then dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 318.8, 320.8 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.08 (d, J=8.6 Hz, 1H), 7.71 (s, 1H), 7.48 (dd, J=8.4, 7.1 Hz, 2H), 7.13 (d, J=7.4 Hz, 1H), 6.80 (d, J=8.1 Hz, 1H), 5.57 (s, 2H), 4.35 (s, 3H). Preparation of Intermediate I-1035 5-[(6-bromo-2-pyridyl)oxymethyl]-1-methyl-benzotriazole (I-1035): 5-[(6-bromo-2-pyridyl)oxymethyl]-1-methyl-benzotriazole was prepared in a manner as described for Intermediate I-1034 substituting 5-(bromomethyl)-1-methyl-benzotriazole for 6-(bromomethyl)-1-methyl-benzotriazole. ES/MS: 318.8, 320.8 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.18 (t, J=1.1 Hz, 1H), 7.65 (dd, J=8.5, 1.4 Hz, 1H), 7.55 (dd, J=8.5, 0.9 Hz, 1H), 7.47 (dd, J=8.1, 7.5 Hz, 1H), 7.11 (d, J=7.5 Hz, 1H), 6.78 (d, J=8.2 Hz, 1H), 5.55 (s, 2H), 4.34 (s, 3H). Preparation of Intermediate I-1036 6-bromo-2-[(4-chloro-2-fluoro-phenyl)methoxy]-3-fluoro-pyridine (I-1036): 6-bromo-2-[(4-chloro-2-fluoro-phenyl)methoxy]-3-fluoro-pyridine was prepared in a manner as described for Intermediate I-1034 substituting 1-(bromomethyl)-4-chloro-2-fluoro-benzene for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromo-3-fluoro-pyridin-2-ol for 6-bromopyridin-2-ol. 1H NMR (400 MHz, Chloroform-d) δ 7.51 (t, J=8.0 Hz, 1H), 7.30-7.22 (m, 1H), 7.21-7.11 (m, 2H), 7.07 (dd, J=8.1, 2.7 Hz, 1H), 5.48 (d, J=1.2 Hz, 2H). Preparation of Intermediate I-1037 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-7-chloro-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1037): Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-7-chloro-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-1031 substituting 2-(4-bromo-2,5-difluoro-phenyl)acetic acid for 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid and methyl 4-amino-3-chloro-5-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate I-1038 for methyl 4-amino-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-5-fluoro-benzoate. ES/MS: 513.0, 514.9 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.50 (s, 1H), 8.07 (d, J=1.3 Hz, 1H), 7.38 (dd, J=8.7, 5.6 Hz, 1H), 7.23-7.07 (m, 1H), 4.61-4.40 (m, 3H), 4.40-4.29 (m, 2H), 3.97 (s, 3H), 3.92 (d, J=8.9 Hz, 1H), 3.78 (d, J=8.9 Hz, 1H), 1.32 (s, 3H), 0.63 (s, 3H). Preparation of Intermediate I-1038 Methyl 4-amino-3-chloro-5-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate (I-1038): Methyl 4-amino-3-chloro-5-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate was prepared in a manner as described for Intermediate I-68 substituting methyl 3-chloro-5-fluoro-4-nitro-benzoate for methyl 3-bromo-5-fluoro-4-nitro-benzoate. ES/MS: 299.2 (M+). Preparation of Intermediate I-1041 Tert-butyl 4-[[2-(4-bromo-2,5-difluoro-phenyl)acetyl]amino]-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate: To a solution of 2-(4-bromo-2,5-difluoro-phenyl)acetic acid (575 mg, 2.29 mmol), tert-butyl 4-amino-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate (696 mg, 2.27 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (1292 mg, 3.40 mmol) in DMF (10 mL), N,N-diisopropylethylamine (1.22 mL, 6.98 mmol was added). The mixture was stirred at rt overnight. Next the mixture was diluted with EtOAc and washed with 5% LiCl, saturated NaHCO3, and brine. Dried over sodium sulfate, concentrated and carried onto next step below without purification. ES/MS: 539, 541 (M+H+). Tert-butyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1041): To a solution of tert-butyl 4-[[2-(4-bromo-2,5-difluoro-phenyl)acetyl]amino]-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-5-fluoro-benzoate (1236 mg, 2.22 mmol) and triphenylphosphine oxide, 99% (1851 mg, 6.65 mmol) in DCM (20 mL) at 0° C., triflic anhydride (0.559 mL, 3.33 mmol) was added. The mixture was gradually warmed to rt and stirred for 1 hr. Next, the mixture was diluted with DCM and washed with saturated NaHCO3solution and brine. Next, the mixture was dried over sodium sulfate, concentrated and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 521, 523 (M+H+). 1H NMR (400 MHz, Chloroform-d) δ 8.55 (s, 1H), 7.97 (dd, J=8.5, 1.5 Hz, 1H), 7.77 (d, J=8.5 Hz, 1H), 7.37 (dd, J=8.7, 5.5 Hz, 1H), 7.15 (t, J=7.4 Hz, 1H), 4.59 (t, J=8.9 Hz, 2H), 4.47-4.25 (m, 3H), 3.96 (d, J=8.8 Hz, 1H), 3.81 (d, J=8.8 Hz, 1H), 1.34 (s, 3H), 0.66 (s, 3H). Preparation of Intermediate I-1042 Tert-butyl 3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-4-nitro-benzoate: To a suspension of tert-butyl 3-fluoro-4-nitro-benzoate (3.50 g, 14.5 mmol), (3S)-4,4-dimethyltetrahydrofuran-3-amine; hydrochloride (2.50 g, 16.5 mmol) and (3S)-4,4-dimethyltetrahydrofuran-3-amine; hydrochloride (2.50 g, 16.5 mmol) in THE (30 mL) and DMF (15 mL), N,N-diisopropylethylamine (12.6 mL, 72.5 mmol) was added. The resulting mixture was heated at 80° C. for 18 hr. Upon completion, the mixture was diluted with EtOAc (300 mL), washed with 5% LiCl (250 mL) and brine (250 mL). The organic extract was dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford desired product. ES/MS: 337.2 (M+H+); 1H NMR (400 MHz, Chloroform-d) δ 8.21 (t, J=7.8 Hz, 2H), 7.54 (d, J=1.6 Hz, 1H), 7.22 (dd, J=8.9, 1.7 Hz, 1H), 4.41 (dd, J=9.2, 6.8 Hz, 1H), 4.00 (q, J=7.0 Hz, 1H), 3.81-3.59 (m, 3H), 1.63 (s, 9H), 1.26 (s, 3H), 1.18 (s, 3H). Tert-butyl 4-amino-3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate (I-1042): A solution of tert-butyl 3-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-4-nitro-benzoate (4.35 g, 12.9 mmol) in EtOAc (86 mL) was degassed by cycling the mixture between argon and vacuum 3×. Next, palladium on carbon (10.0%, 1.38 g, 1.29 mmol) was added followed by degassing by cycling the mixture between argon and vacuum. Next, the mixture was stirred at rt with a balloon of hydrogen for 24 hr. Upon completion, the mixture was filtered through Celite and concentrated in vacuo to give desired product. ES/MS m/z: 307.2 (M+H)+. 1H NMR (400 MHz, Chloroform-d) δ 7.40 (d, J=16.5 Hz, 2H), 6.81 (s, 1H), 4.33 (d, J=8.6 Hz, 1H), 3.82-3.54 (m, 4H), 1.60 (s, 9H), 1.23 (s, 3H), 1.17 (s, 2H). Preparation of Intermediate I-1043 Methyl (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-iodobenzyl)oxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1043): A slurry of methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (250 mg, 0.54 mmol), 1-(bromomethyl)-2-fluoro-4-iodobenzene (203 mg, 0.65 mmol), and silver carbonate (370 mg, 1.3 mmol) in toluene (5 mL) was heated at 70° C. for 1 hr. The mixture was filtered through celite, washing with EtOAc, concentrated, and purified (24 g GOLD, 0-100% EtOAc in Hex). ES/MS: 700.1 (M+H+). Preparation of Intermediate I-1044 Methyl (S)-2-(4-(6-((6-bromo-2-chloropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1044): Methyl (S)-2-(4-(6-((6-bromo-2-chloropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-18 substituting 6-bromo-3-(bromomethyl)-2-chloropyridine for 4-bromo-1-(bromomethyl)-2-fluorobenzene and methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-9) for tert-butyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-17). ES/MS: 671.1 (M+H+). Preparation of Intermediate I-1045 Methyl (S)-2-(4-(6-((6-bromo-2-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1045): Methyl (S)-2-(4-(6-((6-bromo-2-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-18 substituting 6-bromo-3-(bromomethyl)-2-fluoropyridine for 4-bromo-1-(bromomethyl)-2-fluorobenzene and methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-9) for tert-butyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-17). ES/MS: 653.2 (M+H+). Preparation of Intermediate I-1046 Methyl (S)-2-(4-(6-((6-bromo-2-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1046): Methyl (S)-2-(4-(6-((6-bromo-2-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-18 substituting 6-bromo-3-(chloromethyl)-2-methoxypyridine for 4-bromo-1-(bromomethyl)-2-fluorobenzene and methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-9) for tert-butyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-17). ES/MS: 665.2 (M+H+). Preparation of Intermediate I-1047 Methyl (S)-2-(4-(6-((5-bromo-6-chloropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1047): Methyl (S)-2-(4-(6-((6-bromo-2-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 3-bromo-2-chloro-6-(chloromethyl)pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 671.1 (M+H+). Preparation of Intermediate I-1048 4-formyl-1-methyl-1H-pyrazole-5-carbonitrile (I-1048): n-BuLi (2.5M, 0.645 mL, 1.61 mmol) was added dropwise to a solution of 4-bromo-2-methyl-pyrazole-3-carbonitrile (250 mg, 1.34 mmol) in THE (12 mL) at −78° C., and the solution was stirred for 15 min. DMF (0.21 mL, 2.69 mmol) was added dropwise and the solution was stirred for 20 min. Reaction was quenched with saturated NH4Cl and water, and warmed to rt. It was extracted with EtOAc (2×) and organic was washed with brine, dried over MgSO4, filtered, concentrated, and purified (ISCO 12 g, 0-80% EtOAc in Hex). Preparation of Intermediate I-1049 (4-chloro-6-fluoropyridin-3-yl)methanol: NaBH4(117 mg, 3.09 mmol) was added to a solution of 4-chloro-6-fluoronicotinaldehyde (470 mg, 2.95 mmol) in MeOH (12 mL) at 0° C., and the solution was stirred for 5 min. The mixture was quenched with saturated NH4Cl and concentrated. The concentrate was dissolved in EtOAc, washed with water then brine, dried over MgSO4, filtered, and concentrated to give (4-chloro-6-fluoropyridin-3-yl)methanol (1), which was taken forward crude. ES/MS: 161.9 (M+1). 4-chloro-5-(chloromethyl)-2-fluoropyridine: Thionyl chloride (0.53 mL, 7.4 mmol) was added to a solution of (4-chloro-6-fluoropyridin-3-yl)methanol (476 mg, 2.95 mmol) in DCM (25 mL), and the resulting solution was stirred for 1 hour. Thionyl chloride (0.53 mL, 7.4 mmol) was added, and the resulting solution was stirred for 1 hour. Thionyl chloride (0.21 mL, 2.9 mmol) was added, and the resulting solution was stirred for 30 min. The mixture was concentrated, redissolved in DCM, and saturated NaHCO3was added dropwise carefully. Next, the phases were separated, and organic was dried over MgSO4, filtered, and concentrated to give 4-chloro-5-(chloromethyl)-2-fluoropyridine (2), which was taken forward crude.1H NMR (400 MHz, Chloroform-d) δ 8.30 (s, 1H), 7.05 (d, J=2.7 Hz, 1H), 4.67 (s, 2H). 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-fluoropyridine: A slurry of 6-bromopyridin-2-ol (557 mg, 3.2 mmol), 4-chloro-5-(chloromethyl)-2-fluoropyridine (480 mg, 2.7 mmol), and Cs2CO3(2.17 g, 6.7 mmol) in CAN (9 mL) was heated at 70° C. for 30 min. The mixture was filtered through celite, concentrated, and purified (ISCO 12 g GOLD, 0-100% EtOAc in Hex) to give 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-fluoropyridine (3). ES/MS: 317.0 (M+1). 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (I-1049): A slurry of 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-fluoropyridine (110 mg, 0.31 mmol), 1H-1,2,3-triazole (0.018 mL, 0.31 mmol), and K2CO3(87 mg, 0.63 mmol) in DMSO (1.4 mL) was heated at 70° C. under an atmosphere of air for 4 hours. The mixture was diluted with brine and extracted 2× with EtOAc. Organic was dried over MgSO4, filtered, concentrated, and purified (ISCO 12 g GOLD, 0-100% EtOAc in Hex, product eluted at ˜80% EtOAc) to give 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (4) as the first eluting of two peaks. 1H NMR (400 MHz, Chloroform-d) δ 8.66 (s, 1H), 8.58 (s, 1H), 8.31 (s, 1H), 7.84 (d, J=1.2 Hz, 1H), 7.48 (t, J=7.8 Hz, 1H), 7.13 (d, J=7.5 Hz, 1H), 6.79 (d, J=8.2 Hz, 1H), 5.54 (s, 2H). Preparation of Intermediate I-1050 6-bromo-2-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-3-fluoropyridine: 6-bromo-2-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-3-fluoropyridine (I-1050) was prepared in a manner as described for Intermediate I-1049 substituting 6-bromo-3-fluoropyridin-2-ol for 6-bromopyridin-2-ol 1H NMR (400 MHz, Chloroform-d) δ 8.68 (s, 1H), 8.58 (d, J=1.2 Hz, 1H), 8.32 (s, 1H), 7.84 (d, J=1.2 Hz, 1H), 7.27 (dd, J=9.3, 8.1 Hz, 1H), 7.09 (dd, J=8.1, 2.7 Hz, 1H), 5.60 (s, 2H). Preparation of Intermediate I-1051 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-imidazol-1-yl)pyridine: 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-imidazol-1-yl)pyridine (I-1051) was prepared in a manner as described for Intermediate I-1049 substituting 1H-imidazole for 1H-1,2,3-triazole.1H NMR (400 MHz, Chloroform-d) δ 8.65 (s, 1H), 8.58 (d, J=1.2 Hz, 1H), 8.31 (s, 1H), 7.84 (d, J=1.2 Hz, 1H), 7.47 (t, J=7.8 Hz, 1H), 7.13 (d, J=7.5 Hz, 1H), 6.78 (d, J=8.2 Hz, 1H), 5.54 (s, 2H). Preparation of Intermediate I-1052 4-(((6-bromo-3,5-difluoropyridin-2-yl)oxy)methyl)benzonitrile (I-1052): A suspension of 4-(hydroxymethyl)benzonitrile (377 mg, 2.8 mmol), 2-bromo-3,5,6-trifluoropyridine (600 mg, 2.8 mmol), and cesium carbonate (1.8 g, 5.7 mmol) in ACN (9.5 mL) was stirred at rt overnight. The mixture was diluted with EtOAc, filtered through celite, concentrated, and purified (0-100% EtOAc in Hex). 1H NMR (400 MHz, Chloroform-d) δ 7.69 (d, J=8.2 Hz, 2H), 7.58 (d, J=8.0 Hz, 2H), 7.30 (dd, J=8.5, 6.5 Hz, 1H), 5.48 (s, 2H). Preparation of Intermediate I-1053 Methyl 3-chloro-5-(1H-1,2,3-triazol-1-yl)picolinate: Glyoxal dimethyl acetal (60% in H2O, 0.17 mL, 1.13 mmol) was added to a solution of tosyl hydrazide (200 mg, 1.07 mmol) in MeOH (2.7 mL) in a microwave vial. The mixture was stirred for 2 hours at rt, and methyl 5-amino-3-chloro-pyridine-2-carboxylate (220 mg, 1.18 mmol) followed by the addition of AcOH (0.06 mL, 1.07 mmol). The reaction was sealed and heated to 75° C. overnight. Next, the mixture was neutralized with DIPEA, concentrated, and purified (0-100% EtOAc in Hex). 1H NMR (400 MHz, Chloroform-d) δ 9.00 (d, J=2.2 Hz, 1H), 8.36 (d, J=2.2 Hz, 1H), 8.12 (d, J=1.3 Hz, 1H), 7.95 (d, J=1.3 Hz, 1H), 4.06 (s, 3H). (3-chloro-5-(1H-1,2,3-triazol-1-yl)pyridin-2-yl)methanol: NaBH4(57 mg, 1.5 mmol) was added to a solution of methyl 3-chloro-5-(1H-1,2,3-triazol-1-yl)picolinate (180 mg, 0.75 mmol) and sodium methoxide (0.5M in MeOH, 0.075 mL, 0.038 mmol) in MeOH (0.7 mL), and the resulting suspension was stirred at rt for 2 hr. The mixture was quenched with saturated NH4Cl and partitioned between EtOAc and water. The organic was washed with brine, dried over MgSO4, filtered, concentrated, and taken forward crude. ES/MS: 210.9 (M+H+). 2-(((6-bromopyridin-2-yl)oxy)methyl)-3-chloro-5-(1H-1,2,3-triazol-1-yl)pyridine (I-1053): A suspension of (3-chloro-5-(1H-1,2,3-triazol-1-yl)pyridin-2-yl)methanol (50 mg, 0.24 mmol), 2-bromo-6-fluoropyridine (50 mg, 0.29 mmol), and cesium carbonate (155 mg, 0.48 mmol) in ACN (1 mL) was stirred at 70° C. overnight. The mixture was diluted with EtOAc, filtered through celite, concentrated, and purified (0-100% EtOAc in Hex). ES/MS: 367.8 (M+H+). Preparation of Intermediate I-1054 Methyl 3-((cis-4-(fluoromethyl)tetrahydrofuran-3-yl)amino)-4-nitrobenzoate: To a solution of methyl 3-((cis-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)-4-nitrobenzoate (100 mg, 0.338 mmol) in 1,2-dimethoxyethane (4 mL) at −50° C. was added a solution of DAST (0.054 mL, 0.41 mmol) in 1,2-dimethoxyethane (2 mL). The reaction was stirred for 10 min at −50° C., warmed up to rt and stirred for 4 hr. To the mixture was added cold saturated NaHCO3(20 mL). The product was extracted with EtOAc (3×). The combined organic layers were washed with brine, dried over MgSO4, filtered, concentrated, redissolved in DMSO, and purified (RP HPLC, 0-100% ACN in H2O). ES/MS: 299.0 (M+H+). Preparation of Intermediate I-1055 Methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-2-fluorobenzoate: Methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-2-fluorobenzoate (I-1055) was prepared in a manner as described for Intermediate I-1 substituting methyl 2,3-difluoro-4-nitrobenzoate for methyl 3-fluoro-4-nitro-benzoate and 4,4-dimethyltetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 283.0 (M+H+). Preparation of Intermediate I-1056 (Peak 1) and 1-1057 (Peak 2) Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-1H-benzo[d]imidazole-6-carboxylate: Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-13 substituting methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-2-fluorobenzoate for ethyl (S)-4-amino-3-fluoro-5-((oxetan-2-ylmethyl)amino)benzoate and 2-(4-bromo-2,5-difluoro-phenyl)acetic acid for 2-(4-bromo-2-fluorophenyl)acetic acid, separating the two enantiomers (Peak 1, I-1056) and (Peak 2, I-1057) by chiral SFC. ES/MS: 497.0 (M+H+). Preparation of Intermediate I-1058 (2-fluoro-4-(4-((trimethylsilyl)methyl)-1H-1,2,3-triazol-1-yl)phenyl)methanol: A suspension of trimethyl(prop-2-yn-1-yl)silane (0.21 mL, 1.44 mmol), (4-azido-2-fluoro-phenyl) methanol (200 mg, 1.2 mmol), sodium ascorbate (84 mg, 0.48 mmol), copper(II) sulfate pentahydrate (60 mg, 0.24 mmol) was stirred at rt overnight. The mixture was then diluted with 2M NH4OH and extracted with EtOAc (2×). The organic layer was washed with water then brine, dried over MgSO4, filtered, concentrated, and purified (12 g, 0-100% EtOAc in Hex). ES/MS: 280.1 (M+H+). 1-(4-(chloromethyl)-3-fluorophenyl)-4-((trimethylsilyl)methyl)-1H-1,2,3-triazole: Thionyl chloride (0.18 mL, 2.4 mmol) was added to a solution of (2-fluoro-4-(4-((trimethylsilyl)methyl)-1H-1,2,3-triazol-1-yl)phenyl)methanol (270 mg, 0.97 mmol) in DCM (9 mL), and the resulting solution was stirred for 1 hour. The mixture was concentrated, redissolved in DCM, and saturated NaHCO3was added dropwise. The phases were separated, and the organic extract was dried over MgSO4, filtered, concentrated, and taken forward crude. ES/MS: 298.0 (M+H+). 2-bromo-6-((2-fluoro-4-(4-((trimethylsilyl)methyl)-1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridine: A suspension of 1-(4-(chloromethyl)-3-fluorophenyl)-4-((trimethylsilyl)methyl)-1H-1,2,3-triazole (80 mg, 0.27 mmol), 6-bromopyridin-2-ol (51 mg, 0.30 mmol), and cesium carbonate (219 mg, 0.67 mmol) in were heated at 70° C. for 30 min. The mixture was diluted with EtOAc, washed with water then brine, dried over MgSO4, filtered, concentrated, and taken forward crude. ES/MS: 435.1 (M+H+). 2-bromo-6-((2-fluoro-4-(4-methyl-1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridine (I-1058): TBAF (1M in THF, 0.54 mL, 0.54 mmol) was added dropwise to a solution of 2-bromo-6-((2-fluoro-4-(4-((trimethylsilyl)methyl)-1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridine (117 mg, 0.27 mmol) in THE at 0° C. The resulting solution was stirred at 0° C. for 1 hr. Next, the mixture was partitioned between EtOAc and sat. NH4Cl. The organic layer was washed with sat. NH4Cl, and brine, dried over MgSO4, filtered, concentrated, and purified (0-100% EtOAc in Hex). ES/MS: 363.1 (M+H+). Preparation of Intermediate I-1059 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-N-((1-methyl-1H-pyrazol-3-yl)methyl)picolinamide: 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-N-((1-methyl-1H-pyrazol-3-yl)methyl)picolinamide was prepared in a manner as described for Intermediate I-1273 substituting (1-methylpyrazol-3-yl)methanamine for methylamine hydrochloride. ES/MS: 437.9 (M+H+). Intermediate I-1059 2-bromo-6-((4-chloro-3-fluorobenzyl)oxy)pyridine (I-1059a): 2-bromo-6-((4-chloro-3-fluorobenzyl)oxy)pyridine was prepared in a manner as described for Intermediate I-84 substituting 4-(bromomethyl)-1-chloro-2-fluorobenzene for 2-(bromomethyl)thiazole-5-carbonitrile. Preparation of Intermediate I-1060 2-bromo-6-((3,4-dichlorobenzyl)oxy)pyridine (I-1060): 2-bromo-6-((3,4-dichlorobenzyl)oxy)pyridine was prepared in a manner as described for Intermediate I-84 substituting 4-(bromomethyl)-1,2-dichlorobenzene for 2-(bromomethyl)thiazole-5-carbonitrile. Preparation of Intermediate I-1061 5-((5-bromo-2-fluorophenoxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (I-1061): 5-((5-bromo-2-fluorophenoxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine was prepared in a manner as described for Intermediate I-1049 substituting 5-bromo-2-fluorophenol for 6-bromopyridin-2-ol.1H NMR (400 MHz, Chloroform-d) δ 8.67 (s, 1H), 8.59 (d, J=1.1 Hz, 1H), 8.34 (s, 1H), 7.86 (d, J=1.2 Hz, 1H), 7.23 (dd, J=7.4, 2.3 Hz, 1H), 7.13 (ddd, J=8.6, 4.0, 2.2 Hz, 1H), 7.02 (dd, J=10.7, 8.7 Hz, 1H), 5.27 (s, 2H). Preparation of Intermediate I-1062 4-chloro-6-(1H-1,2,3-triazol-1-yl)nicotinaldehyde: Potassium carbonate (1.7 g, 13 mmol) was added to a solution of 4-chloro-6-fluoro-pyridine-3-carbaldehyde (1.0 g, 6.3 mmol) and 1H-1,2,3-triazole (0.36 mL, 6.3 mmol) in DMSO (28 mL). The solution was stirred at rt for 2 hours. Next, the solution was partitioned between brine and EtOAc. The phases were separated and the aqueous phase. was extracted with EtOAc. The combined organic phases were dried over MgSO4, filtered, concentrated, and purified (0-100% EtOAc in Hex) to give 4-chloro-6-(1H-1,2,3-triazol-1-yl)nicotinaldehyde. (4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methanol: NaBH4(103 mg, 2.72 mmol) was added to a solution of 4-chloro-6-(triazol-1-yl)pyridine-3-carbaldehyde (540 mg, 2.59 mmol) in MeOH (10 mL) at 0° C. The mixture was stirred for 5 min. Next, the mixture was quenched with saturated NH4Cl and concentrated. The concentrate was dissolved in EtOAc, washed with water then brine, dried over MgSO4, filtered, concentrated, and taken forward crude. ES/MS: 211.0 (M+1). 4-bromo-2-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyrimidine (I-1062): KHMDS (1M in THF, 0.70 mL, 0.70 mmol) was added dropwise to a solution of [4-chloro-6-(triazol-1-yl)-3-pyridyl]methanol (140 mg, 0.67 mmol) and 4-bromo-2-(methylsulfonyl)pyrimidine (158 mg, 0.67 mmol) in THE at −30° C. The mixture was stirred at −30° C. for 30 min. Next, the mixture was quenched with water dropwise then brine, and extracted 2× with EtOAc. The organic extract was dried over MgSO4, filtered, concentrated, and purified (30% EtOAc in Hex for 5 min then 30-70% for 5 min) to give dp.1H NMR (400 MHz, Chloroform-d) δ 8.68 (s, 1H), 8.58 (d, J=1.2 Hz, 1H), 8.34-8.30 (m, 2H), 7.84 (d, J=1.2 Hz, 1H), 7.23 (d, J=5.1 Hz, 1H), 5.61 (s, 2H). ES/MS: 367.0 (M+1). Preparation of Intermediate I-1063 (2-chloro-6-methoxypyridin-3-yl)methanol: NaBH4(116 mg, 3.06 mmol) was added to a solution of 2-chloro-6-methoxynicotinaldehyde (500 mg, 2.91 mmol) in MeOH (12 mL) at 0° C., and the solution was stirred for 5 min. The mixture was quenched with saturated NH4Cl and concentrated. The concentrate was dissolved in EtOAc, washed with water then brine. Next, the concentrates were dried over MgSO4, filtered, and concentrated to give (2-chloro-6-methoxypyridin-3-yl)methanol, which was taken forward crude. ES/MS: 173.9 (M+1). 2-chloro-3-(chloromethyl)-6-methoxypyridine: Thionyl chloride (0.30 mL, 4.09 mmol) was added to a solution of (2-chloro-6-methoxypyridin-3-yl)methanol (284 mg, 1.64 mmol) in DCM (13 mL), and the resulting solution was stirred for 1 hour. The mixture was concentrated, then redissolved in DCM. Next, saturated NaHCO3was added dropwise carefully. The phases were separated, and organic phase was dried over MgSO4, filtered, and concentrated to give 2-chloro-3-(chloromethyl)-6-methoxypyridine which was taken forward crude. 3-(((6-bromopyridin-2-yl)oxy)methyl)-2-chloro-6-methoxypyridine (I-1063): A slurry of 6-bromopyridin-2-ol (67 mg, 0.38 mmol), 2-chloro-3-(chloromethyl)-6-methoxypyridine (82 mg, 0.43 mmol), and Cs2CO3(348 mg, 1.1 mmol) in ACN (2 mL) was heated at 70° C. for 30 min. The mixture was filtered through celite, concentrated, and purified (ISCO 12 g GOLD, 0-100% EtOAc in Hex) to give 3-(((6-bromopyridin-2-yl)oxy)methyl)-2-chloro-6-methoxypyridine. ES/MS: 330.8 (M+1).1H NMR (400 MHz, Chloroform-d) δ 7.70 (d, J=7.6 Hz, 1H), 7.44 (dd, J=8.2, 7.5 Hz, 1H), 7.08 (dd, J=7.5, 0.6 Hz, 1H), 6.92 (d, J=7.6 Hz, 1H), 6.74 (dd, J=8.1, 0.7 Hz, 1H), 5.32 (s, 2H), 4.00 (s, 3H). Preparation of Intermediate I-1064 Methyl 4-amino-3-((cis-4-(fluoromethyl)tetrahydrofuran-3-yl)amino)benzoate (I-1340): A mixture of methyl 3-[[(3R,4S)-4-(fluoromethyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate (I-1054, 31 mg, 0.1 mmol) and Pd/C (10%, 11.1 mg, 0.01 mmol) in EtOH (4 mL) was degassed and backfilled with argon 3× then with H23×. The mixture was stirred at rt under H2for 20 min. The mixture was diluted with EtOAc, filtered through celite, concentrated to give the title compound. ES/MS: 269.1 (M+H+). Preparation of Intermediate I-1065 Methyl 4-amino-3-(((1-(fluoromethyl)cyclopropyl)methyl)amino)benzoate (I-1065-1-1): N,N-diisopropylethylamine (0.525 mL, 3.01 mmol was added to a suspension of methyl 3-fluoro-4-nitro-benzoate (120 mg, 0.603 mmol), racemic cis 4-aminotetrahydrofuran-3-yl]methanol (77.7 mg, 0.66 mmol) in THF (4 mL) and DMF (2 mL). The mixture was heated at 80 deg. for 18 hr. The crude mixture was diluted with EtOAc, washed with 5% LiCi and brine. The organic extract was dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford the title compound. ES/MS: 297.2 (M+H+). Methyl 3-[[-4-(methoxymethyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate (I-1065-2): NaH (24.2 mg, 0.633 mmol, 60% dispersion) was added to a solution of methyl 3-[[4-(hydroxymethyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate (75 mg, 0.253 mmol) in DMF (4 mL) at 0 deg. To this solution was added methyl iodide (35.9 mg, 0.253 mmol) and the solution stirred for 1 hour at rt. The solution was then split between saturated aqueous ammonium chloride and EtOAc, the aqueous layer was extracted with EtOAc five times, washed with brine. The organic extract was dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford desired product. ES/MS: 311.2 (M+H+). Methyl 4-amino-3-[[-4-(methoxymethyl)tetrahydrofuran-3-yl]amino]benzoate (I-1065, racemic, relative stereochemistry known): A solution of methyl 3-[[-4-(methoxymethyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate (80 mg, 0.258 mmol) in EtOAc (5 mL) was degassed by cycling between argon and vacuum 3×. Then palladium on carbon (10.0%, 27.4 mg, 0.258 mmol) was added and then the mixture was degassed by cycling between argon and vacuum and stirred at rt with a balloon of hydrogen for 24 hr. The mixture was filtered through Celite and concentrated in vacuo to give the title compound. ES/MS: 281.2 (M+H+) Preparation of Intermediate I-1066 Methyl 4-amino-3-(((1-(fluoromethyl)cyclopropyl)methyl)amino)benzoate (cis, relative stereochemistry known): Methyl 4-amino-3-(((1-(fluoromethyl)cyclopropyl)methyl)amino)benzoate was added N,N-diisopropylethylamine (0.525 mL, 3.01 mmol) was added to a suspension of methyl 3-fluoro-4-nitro-benzoate (120 mg, 0.603 mmol), 4-aminotetrahydrofuran-3-yl]methanol (77.7 mg, 0.66 mmol) in THE (4 mL) and DMF (2 mL). The mixture was heated at 80° C. for 18 hr. The crude mixture was diluted with EtOAc, washed with 5% LiCl and brine. The organic extract was dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford desired product. ES/MS: 297.2 (M+H+) Methyl 3-[[-4-(difluoromethoxymethyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate: Trimethyl(bromodifluoromethyl)silane (308 mg, 1.52 mmol, 0.237 mL) and potassium hydrogen fluoride (356, 4.56 mmol) were added to a solution of methyl 3-[[-4-(hydroxymethyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate (75 mg, 0.253 mmol) in DCM (2 mL) and water (2 mL). The solution was stirred vigorously overnight at rt. The solution was then split between water and DCM, washed with brine, and dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford desired product. ES/MS: 347.6 (M+H+) Methyl 4-amino-3-[[-4-(difluoromethoxymethyl)tetrahydrofuran-3-yl]amino]benzoate (I-1066): A solution of methyl 3-[[-4-(difluoromethoxymethyl)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate (88 mg, 0.254 mmol) in EtOAc (5 mL) was degassed by cycling the mixture between argon and vacuum 3×. Next, palladium on carbon (10.0%, 27.0 mg, 0.254 mmol) was added and then the mixture was degassed by cycling the mixture between argon and vacuum and stirred at rt with a balloon of hydrogen for 24 hr. The mixture was filtered through Celite and concentrated in vacuo to give desired product. ES/MS: 317.2 (M+H+) Preparation of Intermediate I-1067 Methyl 4-amino-3-[[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]amino]benzoate (I-1067): Methyl 4-amino-3-[[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]amino]benzoate (I-1067) was prepared in a manner as described for I-1 substituting (1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-amine for 2-methoxyethylamine. ES/MS: 263.5 (M+H+). Preparation of Intermediate I-1068 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4-cyclopropyltetrahydrofuran-3-yl)benzimidazole-5-carboxylic acid (I-1068): Methyl 4-amino-3-[(4-cyclopropyltetrahydrofuran-3-yl)amino]benzoate (I-1068) as a mixture of four different stereoisomers was prepared in a manner as described for I-72 substituting 4-cyclopropyltetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine and methyl 3-fluoro-4-nitro-benzoate for methyl 3-fluoro-5-iodo-4-nitrobenzoate. ES/MS: 624.6 (M+H+). Preparation of Intermediate I-1069 Methyl 4-amino-3-[[(2S,3R)-2-ethyltetrahydrofuran-3-yl]amino]benzoate (I-1069): Methyl 4-amino-3-[[(2S,3R)-2-ethyltetrahydrofuran-3-yl]amino]benzoate (I-1069) was prepared in a manner as described for I-1 substituting (2S,3R)-2-ethyltetrahydrofuran-3-amine for 2-methoxyethylamine. ES/MS: 265.2 (M+H+). Preparation of Intermediate I-1070 Methyl 4-amino-3-[(2,2,5,5-tetramethyltetrahydrofuran-3-yl)amino]benzoate (I-1070): Methyl 4-amino-3-[(2,2,5,5-tetramethyltetrahydrofuran-3-yl)amino]benzoate (I-1070) was prepared in a manner as described for I-1 substituting 2,2,5,5-tetramethyltetrahydrofuran-3-amine for 2-methoxyethylamine. ES/MS: 293.2 (M+H+). Preparation of Intermediate I-1071 Methyl 4-amino-3-(2,5-dioxaspiro[3.4]octan-7-ylamino)benzoate (I-1071): Methyl 4-amino-3-(2,5-dioxaspiro[3.4]octan-7-ylamino)benzoate (I-1071) was prepared in a manner as described for I-1 substituting 2,5-dioxaspiro[3.4]octan-7-amine for 2-methoxyethylamine. ES/MS: 279.2 (M+H+). Preparation of Intermediate I-1072 Methyl 4-amino-3-(4-oxaspiro[2.4]heptan-6-ylamino)benzoate (I-1072): methyl 4-amino-3-(4-oxaspiro[2.4]heptan-6-ylamino)benzoate (I-1072) was prepared in a manner as described for Intermediate I-1 substituting 4-oxaspiro[2.4]heptan-6-amine for 2-methoxyethylamine. ES/MS: 263.2 (M+H+). Preparation of Intermediate I-1073 4-[[6-(4-amino-3-fluoro-5-methyl-phenyl)-2-pyridyl]oxymethyl]-3-fluoro-benzonitrile: A suspension of 4-bromo-2-fluoro-6-methyl-aniline (160 mg, 0.78 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II); PdCl2(dppf) (88 mg, 0.12 mmol), potassium propionate (264 mg, 2.35 mmol), and Bis(pinacolato)diboron (209 mg, 0.823 mmol) was degassed, then heated at 120° C. for 1 hr. Next, sodium carbonate (2000 mmol/L, 0.81 mL, 1.63 mmol), 4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (289 mg, 0.94 mmol) was added, and then the mixture was heated at 110° C. for 1 hr. The mixture was diluted with EtOAc and water. The organic extract was dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 352.2 (M+H+) 4-[[6-(4-bromo-3-fluoro-5-methyl-phenyl)-2-pyridyl]oxymethyl]-3-fluoro-benzonitrile: To CuBr2(110 mg, 0.617 mmol) and tert-butyl nitrite (0.07 mL, 0.617 mmol) in MeCN, preheated to 60 deg. Next, 4-[[6-(4-amino-3-fluoro-5-methyl-phenyl)-2-pyridyl]oxymethyl]-3-fluoro-benzonitrile (145 mg, 0.411 mmol) as a solution in MeCN (2 mL) was added dropwise. The mixture was then warmed to 80° C. and stirred 1 h. Next, the mixture was left to cool to rt, then diluted with DCM, poured over 0.5 N HCl, extracted with DCM, dried (MgSO4), concentrated, and purified by column chromatography to yield the product. ES/MS: 415.0 (M+) Tert-butyl 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2-fluoro-6-methyl-phenyl]acetate: A suspension of 4-[[6-(4-bromo-3-fluoro-5-methyl-phenyl)-2-pyridyl]oxymethyl]-3-fluoro-benzonitrile (103 mg, 0.248 mmol), XPhos Pd G3 (18.7 mg, 0.025 mmol), and bromo-(2-tert-butoxy-2-oxo-ethyl)zinc (1.49 mL, 0.744 mmol, 0.5 M) was added to a dry vial under argon, then heated at 65° C. for 12 h. The mixture was diluted with EtOAc and water. The organic extract was dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 451.2 (M+H+) 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2-fluoro-6-methyl-phenyl]acetic acid (I-1073): A solution of tert-butyl 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2-fluoro-6-methyl-phenyl]acetate (32 mg, 0.071 mmol) in DCM had 2,2,2-trifluoroacetic acid (0.54 mL, 7.1 mmol) added and the solution was stirred at rt for 3 h. The solution was concentrated to give 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2-fluoro-6-methyl-phenyl]acetic acid (I-1073) which was carried forward without further purification. ES/MS: 395.2 (M+H+) Preparation of Intermediate I-1074 2-bromo-6-[[5-(1,1-difluoroethyl)-2-pyridyl]methoxy]pyridine (I-1074): 2-bromo-6-[[5-(1,1-difluoroethyl)-2-pyridyl]methoxy]pyridine (I-1074) was prepared in a manner as described for Intermediate I-1034 substituting 2-(bromomethyl)-5-(1,1-difluoroethyl)pyridine for 6-(bromomethyl)-1-methyl-benzotriazole. ES/MS: 330.2 (M+H+). Preparation of Intermediate I-1075 Methyl 2-[(4-bromo-2-fluoro-5-methyl-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluoro-benzimidazole-5-carboxylate (I-1075): This compound was made analogous to 1-1031 replacing 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid for 2-(4-bromo-2-fluoro-5-methyl-phenyl)acetic acid. ES/MS: 493.1, 495.0 (M+H+). Preparation of Intermediate I-1076 Methyl 2-[(4-bromo-2-fluoro-5-methyl-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1076): This compound was made analogous to 1-2 replacing 2-(4-bromo-2-fluoro-phenyl)acetic acid with 2-(4-bromo-2-fluoro-5-methyl-phenyl)acetic acid and I-1 with 1-80. ES/MS: 492.6, 494.6 (M+H+). Preparation of Intermediate I-1077 6-bromo-3-fluoro-2-[[4-(triazol-1-yl)phenyl]methoxy]pyridine (I-1077): 6-bromo-3-fluoro-2-[[4-(triazol-1-yl)phenyl]methoxy]pyridine (I-1077) was prepared in a manner as described for intermediate 1-1034 substituting 1-(4-(bromomethyl)phenyl)-1H-1,2,3-triazole for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromo-3-fluoro-pyridin-2-ol for 6-bromopyridin-2-ol ES/MS: 348.2, 350.2 (M+H+). Preparation of Intermediate I-1086 4-[(4-bromopyrimidin-2-yl)oxymethyl]benzonitrile (I-1086): 4-[(4-bromopyrimidin-2-yl)oxymethyl]benzonitrile (I-1086) was made analogous to I-94 replacing 3-fluoro-4-(hydroxymethyl)benzonitrile with 4-(hydroxymethyl)benzonitrile. ES/MS: 475.0, 477.0 (M+H+). Preparation of Intermediate I-1087 4-bromo-2-[[4-(triazol-1-yl)phenyl]methoxy]pyrimidine (I-1087):4-bromo-2-[[4-(triazol-1-yl)phenyl]methoxy]pyrimidine (I-1087) was made analogous to I-94 replacing 3-fluoro-4-(hydroxymethyl)benzonitrile with [4-(triazol-1-yl)phenyl]methanol. ES/MS: 332.0, 334.0 (M+H+). Preparation for Intermediate I-1088 2-bromo-6-[[4-(triazol-1-yl)phenyl]methoxy]pyridine (I-1088): 2-bromo-6-[[4-(triazol-1-yl)phenyl]methoxy]pyridine (I-1088) was prepared in a manner as described for intermediate 1-1034 substituting 1-(4-(bromomethyl)phenyl)-1H-1,2,3-triazole for 6-(bromomethyl)-1-methyl-benzotriazole. ES/MS: 331.2, 333.2 (M+H+). Preparation of Intermediate I-1091 4-bromo-2-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidine (I-1091): 4-bromo-2-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidine (I-1091) was made analogous to I-94 replacing 3-fluoro-4-(hydroxymethyl)benzonitrile with (2-fluoro-4-(trifluoromethyl)phenyl)methanol. ES/MS: 351.0, 353.0 (M+H+). Preparation of Intermediate I-1098 6-bromo-2-((2,4-dichlorobenzyl)oxy)-3-fluoropyridine (I-1098): 6-bromo-2-((2,4-dichlorobenzyl)oxy)-3-fluoropyridine (I-1098) was prepared in a manner as described for intermediate I-1034 substituting 1-(bromomethyl)-2,4-dichlorobenzene for 6-(bromomethyl)-1-methyl-benzotriazol and 6-bromo-3-fluoro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 351.2 (M+H+). Preparation of Intermediate I-1099 2-bromo-6-[(2,4-dichlorophenyl)methoxy]pyridine (I-1099): 2-bromo-6-[(2,4-dichlorophenyl)methoxy]pyridine (I-1099) was prepared in a manner as described for intermediate I-1040 substituting (2,4-dichlorophenyl)methanol for 6-(bromomethyl)-1-methyl-benzotriazole and 2-bromo-6-fluoro-pyridine for 4-bromo-2-fluoro-pyrimidine. ES/MS: 334.0, 336.0 (M+H+). Preparation of Intermediate I-1100 4-bromo-2-[(2,4-dichlorophenyl)methoxy]pyrimidine (I-1100): 4-bromo-2-[(2,4-dichlorophenyl)methoxy]pyrimidine (I-1100) was made analogous to I-94 replacing 3-fluoro-4-(hydroxymethyl)benzonitrile with (2,4-dichlorophenyl)methanol. ES/MS: 334.0, 336.0 (M+H+). Preparation of Intermediate I-1101 (2-chloro-4-(4-(trimethylsilyl)-11H-1,2,3-triazol-1-yl)phenyl)methanol (I-1101): (2-chloro-4-(4-(trimethylsilyl)-1H-1,2,3-triazol-1-yl)phenyl)methanol (I-1101) was made analogous to 1-50 replacing methyl 6-chloropyridine-3-carboxylate with methyl 2-chloro-4-fluoro-benzoate and ethynylcyclopropane with ethynyl(trimethyl)silane. ES/MS: 282.0 (M+H+). Preparation of Intermediate I-1102 2-bromo-6-[[2-chloro-4-(triazol-1-yl)phenyl]methoxy]pyridine (I-1102): 2-bromo-6-[[2-chloro-4-(triazol-1-yl)phenyl]methoxy]pyridine (I-1102) was made analogous to 1-1040 substituting [2-chloro-4-(4-trimethylsilyltriazol-1-yl)phenyl]methanol for 6-(bromomethyl)-1-methyl-benzotriazole and 2-bromo-6-fluoro-pyridine for 4-bromo-2-fluoro-pyrimidine. ES/MS: 366.0, 368.0 (M+H+). Preparation of Intermediate I-1103 Tert-butyl 2-[(4-bromo-2,3,6-trifluoro-phenyl)methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-1103): tert-butyl 2-[(4-bromo-2,3,6-trifluoro-phenyl)methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-1103) was made analogous to I-2 replacing 2-(4-bromo-2-fluoro-phenyl)acetic acid with 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid and I-1 with 1-6. ES/MS: 500.0, 502.0 (M+H+). Preparation of Intermediate I-1104 Ethyl 4-amino-5-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-2-methoxy-benzoate (I-1104): ethyl 4-amino-5-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]-2-methoxy-benzoate (I-1104) was made analogous to I-1 replacing methyl 3-fluoro-4-nitro-benzoate with methyl 5-fluoro-2-methoxy-4-nitro-benzoate and 2-Methoxyethylamine with (3S)-4,4-dimethyltetrahydrofuran-3-amine; hydrochloride. ES/MS: 309.0 (M+H+). Preparation of Intermediate I-1105 Ethyl 4-amino-2-chloro-5-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate (I-1105): ethyl 4-amino-2-chloro-5-[[(3S)-4,4-dimethyltetrahydrofuran-3-yl]amino]benzoate (I-1105) was made analogous to I-1 replacing methyl 3-fluoro-4-nitro-benzoate with ethyl 2-chloro-5-fluoro-4-nitro-benzoate and 2-Methoxyethylamine with (3S)-4,4-dimethyltetrahydrofuran-3-amine; hydrochloride. ES/MS: 313.0 (M+H+). Preparation of Intermediate I-1106 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-6-methoxy-benzimidazole-5-carboxylate (I-1106): methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-6-methoxy-benzimidazole-5-carboxylate (I-1106) was made analogous to I-2 replacing 2-(4-bromo-2-fluoro-phenyl)acetic acid with 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and I-1 with I-1104. ES/MS: 510.0 (M+H+). Preparation of Intermediate I-1107 Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-6-chloro-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1107): ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-6-chloro-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1107) was made prepared in a manner as described for I-2 replacing 2-(4-bromo-2-fluoro-phenyl)acetic acid with 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and I-1 with I-1105. ES/MS: 528.0 (M+H+). Preparation of Intermediate I-1110 Methyl (R)-4-amino-3-((tetrahydrofuran-3-yl)amino)benzoate (I-1110): methyl (R)-4-amino-3-((tetrahydrofuran-3-yl)amino)benzoate (I-1110) was prepared in a manner as described for Intermediate I-1 substituting 4-methylbenzenesulfonic acid; (3R)-tetrahydrofuran-3-amine for 2-methoxyethylamine. ES/MS: 237.2 (M+H+). Preparation of Intermediate I-1111 Methyl (S)-4-amino-3-((tetrahydrofuran-3-yl)amino)benzoate (I-1111): methyl (S)-4-amino-3-((tetrahydrofuran-3-yl)amino)benzoate (I-1111) was prepared in a manner as described for Intermediate I-1 substituting 4-methylbenzenesulfonic acid; (3S)-tetrahydrofuran-3-amine for 2-methoxyethylamine. ES/MS: 237.1 (M+H+). Preparation of Intermediate I-1112 Methyl 4-amino-3-((cis-4-methoxytetrahydrofuran-3-yl)amino)benzoate (I-1112): methyl 4-amino-3-((cis-4-methoxytetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-1 substituting cis-4-methoxytetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 367.2 (M+H+) Preparation of Intermediate I-1113 Tert-butyl 3-(((5-oxaspiro[2.4]heptan-6-yl)methyl)amino)-4-aminobenzoate (I-1113): tert-butyl 3-(((5-oxaspiro[2.4]heptan-6-yl)methyl)amino)-4-aminobenzoate was prepared in a manner as described for Intermediate I-6 substituting (5-oxaspiro[2.4]heptan-6-yl)methanamine hydrochloride for 2-methoxyethylamine. ES/MS: 319.0 (M+H+) Preparation of Intermediate I-1114 Tert-butyl 4-amino-3-(((2-cyclopropyltetrahydrofuran-2-yl)methyl)amino)benzoate (I-1114): tert-butyl 3-(((5-oxaspiro[2.4]heptan-6-yl)methyl)amino)-4-aminobenzoate was prepared in a manner as described for Intermediate I-6 substituting (2-cyclopropyltetrahydrofuran-2-yl)methanamine for 2-methoxyethylamine. ES/MS: 333.3 (M+H+) Preparation of Intermediate I-1115 Tert-butyl 3-(((2,6-dioxabicyclo[3.2.1]octan-1-yl)methyl)amino)-4-aminobenzoate (I-1115): tert-butyl 3-(((2,6-dioxabicyclo[3.2.1]octan-1-yl)methyl)amino)-4-aminobenzoate was prepared in a manner as described for Intermediate I-6 substituting 2,6-dioxabicyclo[3.2.1]octan-1-ylmethanamine hydrochloride for 2-methoxyethylamine. ES/MS: 335.2 (M+H+) Preparation of Intermediate I-1116 Methyl 4-amino-3-(((2-((tert-butoxycarbonyl)amino)tetrahydrofuran-2-yl)methyl)amino)benzoate (I-1116): methyl 4-amino-3-(((2-((tert-butoxycarbonyl)amino)tetrahydrofuran-2-yl)methyl)amino)benzoate was prepared in a manner as described for Intermediate I-1 substituting tert-butyl (2-(aminomethyl)tetrahydrofuran-2-yl)carbamate hydrochloride for 2-methoxyethylamine. ES/MS: 333.3 (M+H+) Preparation of Intermediate I-1117 Methyl 4-amino-3-(2-oxabicyclo[2.1.1]hexan-4-ylamino)benzoate (I-1117): methyl 4-amino-3-(2-oxabicyclo[2.1.1]hexan-4-ylamino)benzoate was prepared in a manner as described for Intermediate I-1 substituting 2-oxabicyclo[2.1.1]hexan-4-amine hydrochloride for 2-methoxyethylamine. ES/MS: 249.2 (M+H+) Preparation of Intermediate I-1118 Methyl 4-amino-3-[(1-methyl-2-oxabicyclo[2.1.1]hexan-4-yl)amino]benzoate (I-1118): methyl 4-amino-3-[(1-methyl-2-oxabicyclo[2.1.1]hexan-4-yl)amino]benzoate was prepared in a manner as described for Intermediate I-1 substituting 1-methyl-2-oxabicyclo[2.1.1]hexan-4-amine hydrochloride for 2-methoxyethylamine. ES/MS: 249.2 (M+H+) Preparation of Intermediate I-1119 Methyl 4-amino-3-(2-oxabicyclo[3.1.1]heptan-4-ylamino)benzoate (I-1119): methyl 4-amino-3-(2-oxabicyclo[3.1.1]heptan-4-ylamino)benzoate was prepared in a manner as described for Intermediate I-1 substituting 2-oxabicyclo[3.1.1]heptan-4-amine for 2-methoxyethylamine. ES/MS: 263.2 (M+H+) Preparation of Intermediate I-1120 Methyl 4-amino-3-(3-oxabicyclo[3.1.0]hexan-1-ylamino)benzoate (I-1120): methyl 4-amino-3-(3-oxabicyclo[3.1.0]hexan-1-ylamino)benzoate was prepared in a manner as described for Intermediate I-1 substituting 3-oxabicyclo[3.1.0]hexan-1-amine hydrochloride for 2-methoxyethylamine. ES/MS: 249.2 (M+H+) Preparation of Intermediate I-1121 Methyl 4-amino-3-[[cis-4-methoxytetrahydropyran-3-yl]amino]benzoate (I-1121): methyl 4-amino-3-[[cis-4-methoxytetrahydropyran-3-yl]amino]benzoate was prepared in a manner as described for Intermediate I-1 substituting cis-4-methoxytetrahydropyran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 281.1 (M+H+) Preparation of Intermediate I-1122 Methyl 4-amino-3-[[(1r,4r,6s)-2-oxabicyclo[2.2.1]heptan-6-yl]amino]benzoate (I-1122): methyl 4-amino-3-[[(1r,4r,6s)-2-oxabicyclo[2.2.1]heptan-6-yl]amino]benzoate was prepared in a manner as described for Intermediate I-1 substituting (1r,4r,6s)-2-oxabicyclo[2.2.1]heptan-6-amine hydrochloride for 2-methoxyethylamine. ES/MS: 263.2 (M+H+) Preparation of Intermediate I-1123 Methyl 4-amino-3-[[trans-4-(difluoromethyl)tetrahydrofuran-3-yl]amino]benzoate (I-1123): methyl 4-amino-3-[[trans-4-(difluoromethyl)tetrahydrofuran-3-yl]amino]benzoate was prepared in a manner as described for Intermediate I-1 substituting trans-4-(difluoromethyl)tetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 287.2 (M+H+) Preparation of Intermediate I-1124 Methyl 4-amino-3-(5-oxaspiro[2.4]heptan-7-ylamino)benzoate (I-1124): methyl 4-amino-3-(5-oxaspiro[2.4]heptan-7-ylamino)benzoate was prepared in a manner as described for Intermediate I-69 substituting 5-oxaspiro[2.4]heptan-7-amine hydrochloride for 4,4-dimethyltetrahydrofuran-3-amine hydrochloride. ES/MS: 263.2 (M+H+) Preparation of Intermediate I-1125 Methyl 4-amino-3-[[cis-4-(difluoromethyl)tetrahydrofuran-3-yl]amino]benzoate (I-1125): methyl 4-amino-3-[[trans-4-(difluoromethyl)tetrahydrofuran-3-yl]amino]benzoate was prepared in a manner as described for Intermediate I-1 substituting cis-4-(difluoromethyl)tetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 287.2 (M+H+) Preparation of Intermediate I-1126 Methyl 4-amino-3-[[trans-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1126): methyl 4-amino-3-[[trans-4-methyltetrahydrofuran-3-yl]amino]benzoate was prepared in a manner as described for Intermediate I-1 substituting trans-4-methyltetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 251.2 (M+H+) Preparation of Intermediate I-1129 2-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-5-(trifluoromethyl)pyridine (I-1129): 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine (107 mg, 0.41 mmol), 6-bromopyridin-2-ol (60 mg, 0.35 mmol), and silver carbonate (190 mg, 0.69 mmol) in toluene (5 mL) was stirred at 90° C. for 1 h. The mixture was cooled to rt and filtered through a Celite plug. The resulting solution was concentrated and purified by flash chromatography (eluent: EtOAc/hexanes) to yield the desired product. ES/MS: 351.0 (M+H+) Preparation of Intermediate I-1131 Ethyl 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline-2-carboxylate (I-1131): DIPEA (0.075 mL, 0.43 mmol) and ethyl chloroformate (0.016 mL, 0.17 mmol) were added to a solution of 5-[(6-bromo-2-pyridyl)oxymethyl]isoindoline; 2,2,2-trifluoroacetic acid (60 mg, 0.14 mmol) in DCM at rt. The mixture was stirred at rt for 2 hr. before concentrating in vacuo. Purification by flash chromatography (eluent: EtOAc/hexanes) yielded the desired product. ES/MS: 379.0 (M+H+) Preparation of Intermediate I-1132 Methyl 2-[(4-bromo-2-chloro-5-fluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1132): Methyl 2-[(4-bromo-2-chloro-5-fluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting methyl 4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate 1-80 for methyl 4-amino-3-(2-methoxyethylamino)benzoate I-1 and 2-(4-bromo-2-chloro-5-fluoro-phenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 497.3 (M+H+). Preparation of Intermediate I-1133 Methyl 4-amino-3-fluoro-5-((cis-4-methoxytetrahydrofuran-3-yl)amino)benzoate (I-1133): methyl 4-amino-3-fluoro-5-((cis-4-methoxytetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-1 substituting trans-4-methoxytetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine and methyl 3,5-difluoro-4-nitro-benzoate for methyl 3-fluoro-4-nitro-benzoate. ES/MS: 285.2 (M+H+) Preparation of Intermediate I-1134 Ethyl 2-[[2,5-difluoro-4-(5-fluoro-6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1134): ethyl 2-[[2,5-difluoro-4-(5-fluoro-6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate 1-9 substituting 1-14 for 1-8 and 2-benzyloxy-6-bromo-3-fluoro-pyridine for 2-benzyloxy-6-bromopyridine. ES/MS: 516.1 (M+H+). Preparation of Intermediate I-1135 Ethyl 2-(4-bromo-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1135): ethyl 2-(4-bromo-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for I-2 substituting ethyl 4-amino-3-fluoro-5-((2-methoxyethyl)amino)benzoate I-1032 for methyl 4-amino-3-(2 methoxyethylamino)benzoate I-1 and 2-(4-bromo-2,5,6-trifluorophenyl)acetic acid for 2-(4-bromo-2,5-difluorophenyl)acetic acid. ES/MS: 490.2 (M+H+). Preparation of Intermediate I-1136 Ethyl 7-fluoro-3-(2-methoxyethyl)-2-[[2,3,6-trifluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]benzimidazole-5-carboxylate (I-1136): ethyl 7-fluoro-3-(2-methoxyethyl)-2-[[2,3,6-trifluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-9 substituting ethyl 2-(4-bromo-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate I-1135 for I-8. ES/MS: 504.1 (M+H+). Preparation of Intermediate I-1140 Tert-butyl 4-amino-3-((cis-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)benzoate (I-1140): tert-butyl 4-amino-3-((cis-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-6 substituting (cis-4-aminotetrahydrofuran-3-yl)methanol for 2-methoxyethylamine. ES/MS: 339.2 (M+H+) Preparation of Intermediate I-1141 Tert-butyl 4-amino-3-((trans-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)benzoate (I-1141): tert-butyl 4-amino-3-((trans-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-6 substituting (trans-4-aminotetrahydrofuran-3-yl)methanol for 2-methoxyethylamine. ES/MS: 339.2 (M+H+) Preparation of Intermediate I-1142 Methyl 3-((5-oxaspiro[2.4]heptan-7-yl)amino)-4-amino-5-fluorobenzoate (I-1142): methyl 3-((5-oxaspiro[2.4]heptan-7-yl)amino)-4-amino-5-fluorobenzoate was prepared in a manner as described for Intermediate I-1 substituting 5-oxaspiro[2.4]heptan-7-amine hydrochloride for 2-methoxyethylamine and methyl 3,5-difluoro-4-nitro-benzoate for methyl 3-fluoro-4-nitro-benzoate. ES/MS: 281.2 (M+H+) Preparation of Intermediate I-1143 2-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-5-methyl-pyridine (I-1143): 2-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-5-methyl-pyridine was prepared in a manner as described for Intermediate I-43 substituting (3-fluoro-5-methyl-2-pyridyl)methanol for (1-methylimidazol-4-yl)methanol. ES/MS: 298.8 (M+H+) Preparation of Intermediate I-1144 Methyl 4-amino-3-fluoro-5-((cis-4-methoxytetrahydrofuran-3-yl)amino)benzoate (I-1144): methyl 4-amino-3-fluoro-5-((cis-4-methoxytetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-1 substituting 5-oxaspiro[2.4]heptan-7-amine hydrochloride for 2-methoxyethylamine and methyl 3,5-difluoro-4-nitro-benzoate for methyl 3-fluoro-4-nitro-benzoate. ES/MS: 281.2 (M+H+) Preparation of Intermediate I-1149 Methyl (S)-4-amino-3-chloro-5-((oxetan-2-ylmethyl)amino)benzoate (I-1149): methyl (S)-4-amino-3-chloro-5-((oxetan-2-ylmethyl)amino)benzoate was prepared in a manner as described for Intermediate I-68 substituting (S)-oxetan-2-ylmethanamine for 2-methoxyethylamine and methyl 3-chloro-5-fluoro-4-nitrobenzoate for methyl 3-fluoro-4-nitro-benzoate. ES/MS: 271.0 (M+H+). Preparation of Intermediate I-1150 Methyl (S)-2-(4-bromo-2,5-difluorobenzyl)-4-chloro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1150): methyl (S)-2-(4-bromo-2,5-difluorobenzyl)-4-chloro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-2 substituting methyl (S)-4-amino-3-chloro-5-((oxetan-2-ylmethyl)amino)benzoate 1-1149 for methyl 4-amino-3-(2-methoxyethylamino)benzoate I-1 and 2-(4-bromo-2,5-difluoro-phenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 504.9 (M+H+). Preparation of Intermediate I-1152 Methyl 4-nitro-3-(3-oxabicyclo[3.1.1]heptan-1-ylamino)benzoate (I-1152): methyl 4-nitro-3-(3-oxabicyclo[3.1.1]heptan-1-ylamino)benzoate was prepared in a manner as described for Intermediate I-1 substituting 3-oxabicyclo[3.1.1]heptan-1-amine hydrochloride for 2-methoxyethylamine. ES/MS: 293.0 (M+H+). Preparation of Intermediate I-1153 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-(3-oxabicyclo[3.1.1]heptan-1-yl)benzimidazole-5-carboxylate (I-1153): Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-(3-oxabicyclo[3.1.1]heptan-1-yl)benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting methyl 4-nitro-3-(3-oxabicyclo[3.1.1]heptan-1-ylamino)benzoate 1-1152 for methyl 4-amino-3-(2-methoxyethylamino)benzoate I-1 and 2-(4-bromo-2,5-difluoro-phenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 477.0 (M+H+). Preparation of Intermediate I-1154 2-chloro-4-[(4-chloro-2-fluoro-phenyl)methoxy]-5-fluoro-pyrimidine (I-1154): 2-chloro-4-[(4-chloro-2-fluoro-phenyl)methoxy]-5-fluoro-pyrimidine was prepared in a manner as described for Intermediate I-1129 substituting 1-(bromomethyl)-4-chloro-2-fluoro-benzene for 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine and 2-chloro-5-fluoro-pyrimidin-4-ol for 6-bromopyridin-2-ol. ES/MS: 291.0 (M+H+) Preparation of Intermediate I-1155 Methyl 4-amino-3-[[cis-4-(cyclopropoxy)tetrahydrofuran-3-yl]amino]benzoate (I-1155): Methyl 4-amino-3-[[cis-4-(cyclopropoxy)tetrahydrofuran-3-yl]amino]benzoate was prepared in a manner as described for Intermediate I-68 substituting cis-4-(cyclopropoxy)tetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 293.0 (M+H+) Preparation of Intermediate I-1156 Methyl 3-[[cis-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate (I-1156): Methyl 3-[[cis-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl]amino]-4-nitro-benzoate was prepared in a manner as described for Intermediate I-1 substituting cis-4-(2,2-difluoroethoxy)tetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 317.2 (M+H+). Preparation of Intermediate I-1157 Methyl 6-(triazol-1-yl)-4-(trifluoromethyl)pyridine-3-carboxylate: 2H-triazole (170 mg, 2.5 mmol), methyl 6-chloro-4-(trifluoromethyl)pyridine-3-carboxylate (500 mg, 2.0 mmol), and K2CO3(580 mg, 4.2 mmol) in DMF (15 mL) was stirred at 80° C. for 10 h. The mixture was diluted with EtOAc and water, and the layers were separated. The combined organic extracts were washed with brine, dried over MgSO4, filtered, and concentrated. Purification by flash chromatography (eluent: EtOAc/hexanes) yielded the desired product. ES/MS: 273.1 (M+H+) 5-(bromomethyl)-2-(triazol-1-yl)-4-(trifluoromethyl)pyridine (I-1157): Methyl 6-(triazol-1-yl)-4-(trifluoromethyl)pyridine-3-carboxylate (140 mg, 0.51 mmol) in THE (10 mL) was cooled to 0° C. DIABL (1M in THF, 2.0 mL, 2.0 mmol) was added dropwise and the mixture was slowly warmed to rt for 5 h. The mixture was next quenched with saturated aqueous NH4Cl and diluted with EtOAc, and the layers were separated. The combined organic extracts were washed with brine, dried over MgSO4, filtered, and concentrated. The residue was dissolved in DCM and cooled to 0° C. Triphenylphosphine (100 mg, 0.37 mmol) and CBr4(120 mg, 0.37 mmol) were added, respectively. The mixture was warmed to rt and stirred for 2 h before concentrating in vacuo. Purification by flash chromatography (eluent: EtOAc/hexanes) yielded the desired product. ES/MS: 308.0 (M+H+) Preparation of Intermediate I-1158 4-[(6-bromo-3-chloro-2-pyridyl)oxymethyl]benzonitrile (I-1158): 4-[(6-bromo-3-chloro-2-pyridyl)oxymethyl]benzonitrile was prepared in a manner as described for Intermediate I-1129 substituting 4-(bromomethyl)benzonitrile for 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine and 6-bromo-3-chloro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 324.0 (M+H+) Preparation of Intermediate I-1159 4-[(6-bromo-3-chloro-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (I-1159): 4-[(6-bromo-3-chloro-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile was prepared in a manner as described for Intermediate I-1129 substituting 4-(bromomethyl)-3-fluorobenzonitrile for 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine and 6-bromo-3-chloro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 342.0 (M+H+) Preparation of Intermediate I-1160 Methyl (S)-4-amino-5-((4,4-dimethyltetrahydrofuran-3-yl)amino)-2-fluorobenzoate (I-1160): Methyl (S)-4-amino-5-((4,4-dimethyltetrahydrofuran-3-yl)amino)-2-fluorobenzoate was prepared in a manner as described for Intermediate I-1 substituting (S)-4,4-dimethyltetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine and methyl 2,5-difluoro-4-nitrobenzoate for methyl 3-fluoro-4-nitrobenzoate. ES/MS: 283.2 (M+H+) Preparation of Intermediate I-1161 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-6-fluoro-benzimidazole-5-carboxylate (I-1161): Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-6-fluoro-benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting methyl (S)-4-amino-5-((4,4-dimethyltetrahydrofuran-3-yl)amino)-2-fluorobenzoate 1-1160 for methyl 4-amino-3-(2-methoxyethylamino)benzoate I-1 and 2-(4-bromo-2,5-difluoro-phenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 498.0 (M+H+). Preparation of Intermediate I-1162 Ethyl (S)-5-amino-4-((4,4-dimethyltetrahydrofuran-3-yl)amino)picolinate (I-1162): ethyl (S)-5-amino-4-((4,4-dimethyltetrahydrofuran-3-yl)amino)picolinate was prepared in a manner as described for Intermediate I-1 substituting (S)-4,4-dimethyltetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine and ethyl 4-chloro-5-nitropicolinate for methyl 3-fluoro-4-nitrobenzoate. ES/MS: 280.0 (M+H+) Preparation of Intermediate I-1163 Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-1-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]imidazo[4,5-c]pyridine-6-carboxylate (I-1163): Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-1-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]imidazo[4,5-c]pyridine-6-carboxylate was prepared in a manner as described for Intermediate I-2 substituting ethyl (S)-5-amino-4-((4,4-dimethyltetrahydrofuran-3-yl)amino)picolinate 1-1162 for methyl 4-amino-3-(2-methoxyethylamino)benzoate I-1 and 2-(4-bromo-2,5-difluoro-phenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 495.0 (M+H+). Preparation of Intermediate I-1164 3-[(6-bromo-2-pyridyl)oxymethyl]-1-methyl-pyridin-2-one (I-1164): 3-[(6-bromo-2-pyridyl)oxymethyl]-1-methyl-pyridin-2-one was prepared in a manner as described for Intermediate I-43 substituting [3-(hydroxymethyl)-1-methyl-pyridin-2-one for (1-methylimidazol-4-yl)methanol. ES/MS: 295.0 (M+H+) Preparation of Intermediate I-1165 4-[(6-bromo-2-pyridyl)oxymethyl]-1-methyl-pyridin-2-one (I-1165): 3-[(6-bromo-2-pyridyl)oxymethyl]-1-methyl-pyridin-2-one was prepared in a manner as described for Intermediate I-43 substituting [4-(hydroxymethyl)-1-methyl-pyridin-2-one for (1-methylimidazol-4-yl)methanol. ES/MS: 295.0 (M+H+) Preparation of Intermediate I-1166 Ethyl (S)-5-amino-6-((4,4-dimethyltetrahydrofuran-3-yl)amino)picolinate (I-1166): ethyl (S)-5-amino-6-((4,4-dimethyltetrahydrofuran-3-yl)amino)picolinate was prepared in a manner as described for Intermediate I-1 substituting (S)-4,4-dimethyltetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine and ethyl 6-chloro-5-nitropicolinate for methyl 3-fluoro-4-nitrobenzoate. ES/MS: 280.0 (M+H+) Preparation of Intermediate I-1167 Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]imidazo[4,5-b]pyridine-5-carboxylate (I-1167): Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]imidazo[4,5-b]pyridine-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting ethyl (S)-5-amino-6-((4,4-dimethyltetrahydrofuran-3-yl)amino)picolinate I-1166 for methyl 4-amino-3-(2-methoxyethylamino)benzoate I-1 and 2-(4-bromo-2,5-difluoro-phenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 495.0 (M+H+). Preparation of Intermediate I-1168 2-bromo-6-[(3-fluoro-4-pyridyl)methoxy]pyridine (I-1168): 2-bromo-6-[(3-fluoro-4-pyridyl)methoxy]pyridine was prepared in a manner as described for Intermediate I-43 substituting (3-fluoro-4-pyridyl)methanol for (1-methylimidazol-4-yl)methanol. ES/MS: 283.0 (M+H+) Preparation of Intermediate I-1169 2-bromo-6-[(3-methoxy-4-pyridyl)methoxy]pyridine (I-1169): 2-bromo-6-[(3-methoxy-4-pyridyl)methoxy]pyridine was prepared in a manner as described for Intermediate I-43 substituting (3-methoxy-4-pyridyl)methanol for (1-methylimidazol-4-yl)methanol. ES/MS: 295.0 (M+H+) Preparation of Intermediate I-1170 2-bromo-6-[(4-cyclopropylphenyl)methoxy]pyridine (I-1170): 2-bromo-6-[(4-cyclopropylphenyl)methoxy]pyridine was prepared in a manner as described for Intermediate I-43 substituting (4-cyclopropylphenyl)methanol for (1-methylimidazol-4-yl)methanol. ES/MS: 305.2 (M+H+) Preparation of Intermediate I-1172 6-bromo-3-chloro-2-[(4-chlorophenyl)methoxy]pyridine (I-1172): 6-bromo-3-chloro-2-[(4-chlorophenyl)methoxy]pyridine was prepared in a manner as described for Intermediate I-1129 substituting 1-(bromomethyl)-4-chloro-benzene for 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine and 6-bromo-3-chloro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 352.2 (M+H+) Preparation of Intermediate I-1173 2-(((6-bromopyridin-2-yl)oxy)methyl)-5-(1,1-difluoroethyl)thiazole (I-1173): 2-(((6-bromopyridin-2-yl)oxy)methyl)-5-(1,1-difluoroethyl)thiazole was prepared in a manner as described for Intermediate I-1129 substituting 2-(bromomethyl)-5-(1,1-difluoroethyl)thiazole for 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine. ES/MS: 335.0 (M+H+) Preparation of Intermediate I-1174 4-bromo-2-((4-chlorobenzyl)oxy)pyrimidine (I-1174): 4-bromo-2-((4-chlorobenzyl)oxy)pyrimidine was prepared in a manner as described for Intermediate I-43 substituting (4-chlorophenyl)methanol for (1-methylimidazol-4-yl)methanol and 4-bromo-2-fluoropyrimidine for 2-bromo-6-fluoropyridine. ES/MS: 299.2 (M+H+) Preparation of Intermediate I-1175 4-(((4-bromopyrimidin-2-yl)oxy)methyl)-3-fluorobenzonitrile (I-1175): 4-(((4-bromopyrimidin-2-yl)oxy)methyl)-3-fluorobenzonitrile was prepared in a manner as described for Intermediate I-43 substituting 3-fluoro-4-(hydroxymethyl)benzonitrile for (1-methylimidazol-4-yl)methanol and 4-bromo-2-fluoropyrimidine for 2-bromo-6-fluoropyridine. ES/MS: 308.0 (M+H+) Preparation of Intermediate I-1177 Methyl (S)-2-(4-(6-((5-bromo-1,3,4-thiadiazol-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1177): Methyl (S)-2-(4-(6-((5-bromo-1,3,4-thiadiazol-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-1096 substituting 2-bromo-5-(chloromethyl)-1,3,4-thiadiazole for 2-bromo-5-(bromomethyl)thiazole. ES/MS: 642.1 (M+H+) Preparation of Intermediate I-1178 Methyl 4-amino-3-(((1-(fluoromethyl)cyclopropyl)methyl)amino)benzoate (I-1178): Methyl 4-amino-3-(((1-(fluoromethyl)cyclopropyl)methyl)amino)benzoate was prepared in a manner as described for Intermediate I-1 substituting (1-(fluoromethyl)cyclopropyl)methanamine for 2-methoxyethylamine. ES/MS: 253.2 (M+H+). Preparation of Intermediate I-1179 Tert-butyl 4-amino-3-(((1-(fluoromethyl)cyclopropyl)methyl)amino)benzoate (I-1179): Tert-butyl 4-amino-3-(((1-(fluoromethyl)cyclopropyl)methyl)amino)benzoate was prepared in a manner as described for Intermediate I-6 substituting (1-(fluoromethyl)cyclopropyl)methanamine for 2-methoxyethylamine. ES/MS: 295.2 (M+H+). Preparation of Intermediate I-1180 Tert-butyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate (I-1180): Tert-butyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-1179 for I-1 and 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 509, 511 (M+H+). Preparation of Intermediate I-1076 Methyl 2-[(4-bromo-2-fluoro-5-methyl-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1076): Methyl 2-[(4-bromo-2-fluoro-5-methyl-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-80 for I-1 and 2-(4-bromo-2-fluoro-5-methyl-phenyl)acetic acid I-1277 for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 475, 477 (M+H+). Preparation of Intermediate I-1182 Methyl 2-[(4-bromophenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1182): Methyl 2-[(4-bromophenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-80 for I-1 and 2-(4-bromophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 443, 445 (M+H+). Preparation of Intermediate I-1184 Methyl 3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-2-[[2-fluoro-4-(3-hydroxyphenyl)-5-methyl-phenyl]methyl]benzimidazole-5-carboxylate (I-1184): A suspension of methyl 2-[(4-bromo-2-fluoro-5-methyl-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (130 mg, 0.273 mmol) (I-1076), (3-hydroxyphenyl)boronic acid (75.4 mg, 0.547 mmol), [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II); PdCl2(dppf) (20.3 mg, 0.0273 mmol, sodium carbonate (2000 mmol/L, 0.273 mL, 0.547 mmol) was degassed. The mixture was heated at 100° C. for 1 hr. Next, the mixture was diluted with EtOAc and water. The organic extract was dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 489.1 (M+H+) Preparation of Intermediate I-1185 Methyl 2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1185): Methyl 2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-2 substituting 1-25 for I-1 and 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 497, 499 (M+H+). Preparation of Intermediate I-1186 Tert-butyl 2-(4-bromo-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1186): Tert-butyl 2-(4-bromo-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-6 for I-1 and 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 499, 501 (M+H+). Preparation of Intermediate I-1187 Tert-butyl 2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-((1-(fluoromethyl)cyclopropyl)methyl)-1H-benzo[d]imidazole-6-carboxylate (I-1187): A suspension of tert-butyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate (500 mg, 0.98 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II), PdCl2(dppf) (72.8 mg, 0.098 mmol), potassium propionate (330 mg, 2.94 mmol), and Bis(pinacolato)diboron (324 mg, 1.28 mmol) was degassed, then heated at 110° C. for 2 hr. Next, sodium carbonate (2000 mmol/L, 0.98 mL, 1.96 mmol), 6-bromopyridin-2-ol 256 mg, 1.47 mmol), and [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II); PdCl2(dppf) (30.4 mg, 0.04 mmol) was added and then the resulting mixture was degassed. The mixture was then heated at 100° C. for 1 hr. Upon completion the mixture was diluted with EtOAc and water. The organic extract was dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 523.7 (M+H+) Preparation of Intermediate I-1188 Tert-butyl 2-[(4-bromo-2,3,6-trifluoro-phenyl)methyl]-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate (I-1188): Tert-butyl 2-[(4-bromo-2,3,6-trifluoro-phenyl)methyl]-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-1179 for I-1 and 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 527, 529 (M+H+). Preparation of Intermediate I-1189 Methyl 4-amino-3-fluoro-5-[[1-(fluoromethyl)cyclopropyl]methylamino]benzoate (I-1189): Methyl 4-amino-3-fluoro-5-[[1-(fluoromethyl)cyclopropyl]methylamino]benzoate was prepared in a manner as described for Intermediate I-1 substituting (1-(fluoromethyl)cyclopropyl)methanamine for 2-methoxyethylamine and methyl 3,5-difluoro-4-nitrobenzoate for methyl 3-fluoro-4-nitrobenzoate. ES/MS: 270.2 (M+H+). Preparation of Intermediate I-1190 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-7-fluoro-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate (I-1190): Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-7-fluoro-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-13 substituting 1-1189 for 1-5 and 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 485, 487 (M+H+). Preparation of Intermediate I-1191 Methyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate (I-1191): Methyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-1187 substituting 1-1190 for 1-1180. ES/MS: 499, 501 (M+H+). Preparation of Intermediate I-1192 Methyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate (I-1192): Methyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[1-(fluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 1-1191 for 1-9 and 5-bromo-2-(chloromethyl)-3-fluoro-pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 687, 689 (M+H+). Preparation of Intermediate I-1193 Ethyl 3-[[1-(difluoromethyl)cyclopropyl]methylamino]-5-fluoro-4-nitro-benzoate (I-1193): Ethyl 3-[[1-(difluoromethyl)cyclopropyl]methylamino]-5-fluoro-4-nitro-benzoate was prepared in a manner as described for Intermediate I-1 substituting [1-(difluoromethyl)cyclopropyl]methanamine for 2-methoxyethylamine and ethyl 3,5-difluoro-4-nitrobenzoate for methyl 3-fluoro-4-nitrobenzoate. ES/MS: 302.2 (M+H+). Preparation of Intermediate I-1194 Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[[1-(difluoromethyl)cyclopropyl]methyl]-7-fluoro-benzimidazole-5-carboxylate (I-1194): Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[[1-(difluoromethyl)cyclopropyl]methyl]-7-fluoro-benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-13 substituting I-1193 for I-5 and 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 517.1, 519 (M+H+). Preparation of Intermediate I-1195 Tert-butyl 4-amino-3-(((1-(difluoromethyl)cyclopropyl)methyl)amino)benzoate (I-1195): Tert-butyl 4-amino-3-(((1-(difluoromethyl)cyclopropyl)methyl)amino)benzoate was prepared in a manner as described for Intermediate I-6 substituting (1-(difluoromethyl)cyclopropyl)methanamine for 2-methoxyethylamine. ES/MS: 295.2 (M+H+). Preparation of Intermediate I-1196 Tert-butyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[[1-(difluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate (I-1196): Tert-butyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[[1-(difluoromethyl)cyclopropyl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-1195 for I-1 and 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 527, 529 (M+H+). Preparation of Intermediate I-1197 Methyl 2-[[4-[6-[(6-bromo-2-fluoro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1197): Methyl 2-[[4-[6-[(6-bromo-2-fluoro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 6-bromo-3-(bromomethyl)-2-fluoropyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 653, 655 (M+H+). Preparation of Intermediate I-1198 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (I-1198): Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting I-25 for I-1 and 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 479, 481 (M+H+). Preparation of Intermediate I-1199 Methyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (I-1199): Methyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-1187 substituting I-1198 for I-1180. ES/MS: 493, 495 (M+H+). Preparation of Intermediate I-1200 Methyl 2-[[4-[6-[(4-bromo-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (I-1200): Methyl 2-[[4-[6-[(4-bromo-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting I-1199 for I-9 and 4-bromo-1-(bromomethyl)-2-fluoro-benzene for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 680, 682 (M+H+). Preparation of Intermediate I-1201 Methyl 2-(4-bromo-2,3,6-trifluorobenzyl)-4-fluoro-1-((1-(fluoromethyl)cyclopropyl)methyl)-1H-benzo[d]imidazole-6-carboxylate (I-1201): Methyl 2-(4-bromo-2,3,6-trifluorobenzyl)-4-fluoro-1-((1-(fluoromethyl)cyclopropyl)methyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-13 substituting 1-1189 for 1-5 and 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 485, 487 (M+H+). Preparation of Intermediate I-1202 Methyl 7-fluoro-3-[[1-(fluoromethyl)cyclopropyl]methyl]-2-[[2,3,6-trifluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]benzimidazole-5-carboxylate (I-1202): Methyl 7-fluoro-3-[[1-(fluoromethyl)cyclopropyl]methyl]-2-[[2,3,6-trifluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-1187 substituting 1-1201 for 1-1180. ES/MS: 517, 519 (M+H+). Preparation of Intermediate I-1203 Methyl 2-[[4-[6-[(6-bromo-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1203): Methyl 2-[[4-[6-[(6-bromo-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 2-bromo-5-(bromomethyl)pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 635, 637 (M+H+). Preparation of Intermediate I-1204 Methyl 2-[[4-[6-[(6-bromo-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1204): Methyl 2-[[4-[6-[(6-bromo-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting I-31 for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 653, 655 (M+H+). Preparation of Intermediate I-1205 Methyl 2-[[4-[6-[(5-bromo-3-methyl-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1205): Methyl 2-[[4-[6-[(5-bromo-3-methyl-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 5-bromo-2-(chloromethyl)-3-methyl-pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 649, 651 (M+H+). Preparation of Intermediate I-1206 Ethyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-1206): Ethyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-(2-methoxyethyl)benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-1187 substituting I-1033 for I-1180. ES/MS: 485, 487 (M+H+). Preparation of Intermediate I-1208 Ethyl 2-[[4-[6-[(6-bromo-3-pyridyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1208): Ethyl 2-[[4-[6-[(6-bromo-3-pyridyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting ethyl 2-[[2,5-difluoro-4-(5-fluoro-6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate for I-9 and 2-bromo-5-(bromomethyl)pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 686, 687 (M+H+). Preparation of Intermediate I-1209 Ethyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1209): Ethyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting ethyl 2-[[2,5-difluoro-4-(5-fluoro-6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate for I-9 and 5-bromo-2-(chloromethyl)-3-fluoro-pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 703, 705 (M+H+). Preparation of Intermediate I-1210 Methyl 2-[[4-[6-[(5-bromo-3-chloro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1210): Methyl 2-[[4-[6-[(5-bromo-3-chloro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 5-bromo-3-chloro-2-(chloromethyl)pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 670, 672 (M+H+). Preparation of Intermediate I-1211 Methyl 2-[[4-[6-[(4-bromo-2-methoxy-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1211): Methyl 2-[[4-[6-[(4-bromo-2-methoxy-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 4-bromo-1-(bromomethyl)-2-methoxy-benzene for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 664, 666 (M+H+). Preparation of Intermediate I-1212 Methyl 2-[[4-[6-[(4-bromophenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1212): Methyl 2-[[4-[6-[(4-bromophenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 1-bromo-4-(bromomethyl)benzene for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 634, 636 (M+H+). Preparation of Intermediate I-1217 2-(((6-bromopyridin-2-yl)oxy)methyl)-5-chloro-3-fluoropyridine (I-1217): 2-(((6-bromopyridin-2-yl)oxy)methyl)-5-chloro-3-fluoropyridine was prepared in a manner as described for Intermediate I-84 substituting 2-(chloromethyl)-3-fluoro-pyridine for 2-(bromomethyl)thiazole-5-carbonitrile. ES/MS: 317, 319 (M+H+). Preparation of Intermediate I-1218 5-[(6-bromo-2-pyridyl)oxymethyl]-2-chloro-4-methoxy-pyridine (I-1218): 5-[(6-bromo-2-pyridyl)oxymethyl]-2-chloro-4-methoxy-pyridine was prepared in a manner as described for Intermediate I-84 substituting 5-(bromomethyl)-2-chloro-4-methoxy-pyridine for 2-(bromomethyl)thiazole-5-carbonitrile. ES/MS: 329, 331 (M+H+). Preparation of Intermediate I-1219 Methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1219): Methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-1187 substituting I-82 for I-1180. ES/MS: 493, 495 (M+H+). Preparation of Intermediate I-1220 Methyl (S)-2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1220): Methyl (S)-2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for Intermediate I-21 substituting I-1219 for I-9 and 4-bromo-1-(bromomethyl)-2-fluoro-benzene for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 680, 682 (M+H+). Preparation of Intermediate I-1222 2-bromo-6-[(4-chloro-2-fluoro-phenyl)methoxy]-4-(difluoromethyl)pyridine (I-1222): 2-bromo-6-[(4-chloro-2-fluoro-phenyl)methoxy]-4-(difluoromethyl)pyridine was prepared in a manner as described for Intermediate I-49 substituting (4-chloro-2-fluoro-phenyl)methanol for 3-fluoro-4-(hydroxymethyl)benzonitrile and 2,6-dibromo-4-(difluoromethyl)pyridine for 2,6-dichloro-4-(difluoromethyl)pyridine. ES/MS: 366, 368 (M+H+). Preparation of Intermediate I-1226 Methyl (S)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate: Methyl (S)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)-5-fluorobenzoate was prepared the following the procedure for 1-5 substituting (3S)-4,4′-dimethyltetrahydrofuran-3-amine hydrochloride for (S)-oxetan-2-ylmethanamine. ES/MS: 338.5 (M+Na+): Preparation of Intermediate I-1229 Methyl (S)-2-(4-bromo-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1229): Methyl (S)-2-(4-bromo-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for I-1041 substituting INTERMEDIATE I-80 for I-1042 and 2-(4-bromo-2-fluoro-phenyl)acetic acid for 2-(4-bromo-2,5-difluoro-phenyl)acetic acid. ES/MS: 469.8 (M+H+). Preparation of Intermediate I-1230 Methyl (S)-2-(4-bromo-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (I-1230): Methyl (S)-2-(4-bromo-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for 1-2 substituting Intermediate I-1226 for I-1042 and 2-(4-bromo-2-fluoro-phenyl)acetic acid for 2-(4-bromo-2,5-di-fluoro-phenyl)acetic acid. ES/MS: 479.6 (M+H+); Preparation of Intermediate I-1231 Methyl (S)-2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate: Methyl (S)-2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for 1-1041 substituting Intermediate I-80 for 1-1042 and 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid for 2-(4-bromo-2,5-difluorophenyl)acetic acid. ES/MS: 498.0 (M+H+); Preparation of Intermediate I-1232 Methyl (S)-2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate: Methyl (S)-2-(4-bromo-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for 1-2 substituting Intermediate I-1226 for I-1042 and 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid for 2-(4-bromo-2,5-difluorophenyl)acetic acid. ES/MS: 515.7 (M+H+); Preparation of Intermediate I-1233 Methyl (S)-2-(4-bromo-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate: Methyl (S)-2-(4-bromo-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for 1-1041 substituting Intermediate I-80 for 1-1042 and 2-(4-bromo-2,6-difluorophenyl)acetic acid for 2-(4-bromo-2,5-difluorophenyl)acetic acid. ES/MS: 479.9 (M+H+); Preparation of Intermediate I-1234 Methyl (S)-2-(4-bromo-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate: Methyl (S)-2-(4-bromo-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for 1-2 substituting Intermediate I-1226 for 1-1042 and 2-(4-bromo-2,6-trifluorophenyl)acetic acid for 2-(4-bromo-2,6-difluorophenyl)acetic acid. ES/MS: 497.6 (M+H+); Preparation of Intermediate I-1235 Ethyl (S)-2-(4-bromo-2,3,6-trifluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate: Ethyl (S)-2-(4-bromo-2,3,6-trifluorobenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for I-2 substituting I-5 for I-1 and 2-(4-bromo-2,3,6-trifluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 501.3 (M+H+); Preparation of Intermediate I-1237 Methyl (S)-2-((4-bromo-5-fluoro-2-oxopyridin-1(2H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate: Methyl (S)-2-((4-bromo-5-fluoro-2-oxopyridin-1(2H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for 1-1018 substituting Intermediate I-80 for 1-107. ES/MS: 322.5 (M+H+). Preparation of Intermediate I-1238 Methyl (S)-2-((4-bromo-5-chloro-2-oxopyridin-1(2H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate: Methyl (S)-2-((4-bromo-5-chloro-2-oxopyridin-1(2H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate was prepared following the procedure for I-1018 substituting Intermediate I-1226 for 1-107 and 4-chloro-5-fluoro-1H-pyridin-2-one for 4-bromo-5-fluoro-1H-pyridin-2-one. ES/MS: 514.4 (M+H+). Preparation of Intermediate I-1239 Methyl 2-(4-bromo-2-chloro-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate: Methyl 2-(4-bromo-2-chloro-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for 1-1031 substituting methyl 4-amino-3-[(4,4-dimethyltetrahydrofuran-3-yl)amino]-5-fluoro-benzoate 1-107 for ethyl 4-amino-3-fluoro-5-[[(2S)-oxetan-2-yl]methylamino]benzoate and 2-(4-bromo-2-chloro-5-fluoro-phenyl)acetic acid. For 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid. ES/MS: 513.8 (M+H+). Preparation of Intermediate I-1240 Methyl 2-(4-bromo-2-chloro-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate: Methyl 2-(4-bromo-2-chloro-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for 1-1031 substituting methyl 4-amino-3-[(4,4-dimethyltetrahydrofuran-3-yl)amino]-benzoate 1-107 for ethyl 4-amino-3-fluoro-5-[[(2S)-oxetan-2-yl]methylamino]benzoate and 2-(4-bromo-2-chloro-5-fluoro-phenyl)acetic acid. For 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid. ES/MS: 494.6 (M+H+). Preparation of Intermediate I-1242 4-bromo-2-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyrimidine: 4-bromo-2-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyrimidine was prepared in a manner as described for Intermediate I-1034 substituting 1-(bromomethyl)-4-difluoromethyl-2-fluoro-benzene for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromo-pyrimidin-3-ol for 6-bromopyridin-2-ol. ES/MS: 289.7 (M+H+). Preparation of Intermediate I-1243 6-bromo-2-((6-chloro-4-methoxypyridin-3-yl)methoxy)-3-fluoropyridine: 6-bromo-2-((6-chloro-4-methoxypyridin-3-yl)methoxy)-3-fluoropyridine was prepared in a manner as described for Intermediate I-1034 substituting 5-(bromomethyl)-2-chloro-4-methoxypyridine for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromo-3-fluoro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 348.8 (M+H+). Preparation of Intermediate I-1246 6-bromo-2-[(4-chloro-phenyl)methoxy]-3-fluoro-pyridine: 6-bromo-2-[(4-chloro-phenyl)methoxy]-3-fluoro-pyridine was prepared in a manner as described for Intermediate I-1034 substituting 1-(bromomethyl)-4-chloro-benzene for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromo-3-fluoro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 317.6 (M+H+). Preparation of Intermediate I-1247 4-(((6-bromo-3-fluoropyridin-2-yl)oxy)methyl)benzonitrile: 4-(((6-bromo-3-fluoropyridin-2-yl)oxy)methyl)benzonitrile was prepared in a manner as described for Intermediate I-1034 substituting 1-(bromomethyl)-4-benzonitrile for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromo-3-fluoro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 308.3 (M+H+). Preparation of Intermediate I-1248 2-bromo-6-((4-chlorobenzyl)oxy)pyridine: 2-bromo-6-((4-chlorobenzyl)oxy)pyridine was prepared in a manner as described for Intermediate I-1034 substituting 1-(bromomethyl)-4-chloro-benzene for 6-(bromomethyl)-1-methyl-benzotriazole. ES/MS: 298.6 (M+H+). Preparation of Intermediate I-1249 4-(((4-bromopyrimidin-2-yl)oxy)methyl)benzonitrile: 4-(((4-bromopyrimidin-2-yl)oxy)methyl)benzonitrile was prepared in a manner as described for Intermediate I-1034 substituting 1-(bromomethyl)-4-benzonitrile for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromo-3-fluoro-pyrimidin-2-ol for 6-bromopyridin-2-ol. ES/MS: 290.1 (M+H+). Preparation of Intermediate I-1253 cis-isomer 1 and I-1253 trans-isomer 2 Tert-butyl (4-hydroxy-4-methyltetrahydrofuran-3-yl)carbamate: A suspension of tert-butyl (4-oxotetrahydrofuran-3-yl)carbamate (500 mg, 2.5 mmol) in diethyl ether (212.5 mL) was cooled to 0° C. Next, methylmagnesium bromide (2.48 mL, 3.0 Min diethyl ether, 7.45 mmol) was added slowly. The mixture was allowed to warm to 25° C. and then stirred for 16 h. Then the mixture was diluted with diethyl ether and washed with brine. Then the mixture was dried over sodium sulfate, concentrated, and used without further purification. 1H NMR (400 MHz, CDCl3) δ 5.02 (s, 1H), 4.68 (d, J=8.1 Hz, 1H), 4.32-4.24 (m, 1H), 4.21-4.13 (m, 1H), 4.06-3.91 (m, 1H), 3.85-3.72 (m, 2H), 3.62-3.46 (m, 1H), 1.47 (d, J=3.5 Hz, 12H), 1.38 (s, 2H). 4-amino-3-methyltetrahydrofuran-3-ol: A suspension of tert-butyl (4-hydroxy-4-methyltetrahydrofuran-3-yl)carbamate (491 mg, 2.26 mmol) and hydrochloric acid (1.25 mL, 4.0M in dioxane, 5.00 mmol) in DCM (5.00 mL) was stirred overnight. The mixture was diluted in EtOAc and washed with brine, dried over sodium sulfate, concentrated and used without further purification. 1H NMR (400 MHz, CDCl3) δ 3.95 (ddt, J=7.8, 3.8, 2.0 Hz, 1H), 3.77 (tq, J=7.2, 5.3, 3.7 Hz, 2H), 3.72-3.60 (m, 2H), 3.55 (dd, J=9.4, 2.0 Hz, 2H), 3.52-3.41 (m, 2H), 3.38 (t, J=1.8 Hz, 0H), 3.33-3.23 (m, 2H), 3.09 (t, J=6.2 Hz, 2H), 1.20 (t, J=1.8 Hz, 5H), 1.17 (d, J=2.0 Hz, 4H), 1.02-0.92 (m, 6H). Methyl 3-((4-hydroxy-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate: A suspension of 4-amino-3-methyltetrahydrofuran-3-ol (424 mg, 2.76 mmol), methyl 3-fluoro-4-nitro-benzoate (500 mg, 2.51 mmol) and N,N-Diisopropylethylamine (2.19 mL, 1.26 mmol) in THE (4 mL) and DMF (2 mL) was stirred at 82° C. for 16 h. The mixture was diluted in EtOAc and washed with brine, dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MZ: 297.1 (M+H+). Isomer 1: 1H NMR (400 MHz, CDCl3) δ 3.95 (ddt, J=7.8, 3.8, 2.0 Hz, 1H), 3.77 (tq, J=7.2, 5.3, 3.7 Hz, 2H), 3.72-3.60 (m, 2H), 3.55 (dd, J=9.4, 2.0 Hz, 2H), 3.52-3.41 (m, 2H), 3.38 (t, J=1.8 Hz, 0H), 3.33-3.23 (m, 2H), 3.09 (t, J=6.2 Hz, 2H), 1.20 (t, J=1.8 Hz, 5H), 1.17 (d, J=2.0 Hz, 4H), 1.02-0.92 (m, 6H). Isomer 2 Methyl 3-((4-methoxy-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate: A suspension of methyl 3-((4-hydroxy-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (144 mg, 0.49 mmol), sodium hydride (20.5 mg, 0.54 mmol) and iodomethane (33 uL, 0.54 mmol) in DMF (1.00 mL) was stirred at 0° C. for 15 min. Next, the mixture was warmed to 25° C. and stirred for 16 h. The mixture was diluted in EtOAc and washed with brine, dried over sodium sulfate, concentrated, and purified by chromatography (eluent: EtOAc/hexanes) to give desired product EZ/MS: 311.3 (M+H+). Methyl 4-amino-3-((4-methoxy-4-methyltetrahydrofuran-3-yl)amino)benzoate: A suspension of methyl 3-((4-methoxy-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (133 mg, 0.43 mmol), and Palladium on carbon (10% loading, 45.5 mg, 0.43 mmol) in ethanol (5.0 mL) and placed under hydrogen at 25° C. for 5 h. The mixture was diluted in EtOAc and washed with brine, dried over sodium sulfate, concentrated and used without further purification. EZ/MS: 281.1 (M+H+) Preparation of Intermediate I-1255 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid: was prepared in a manner as described for the Intermediate I-7 substituting I-102 for I-3. ES/MZ: 316.6 (M+H+). Preparation of Intermediate I-1256 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorophenyl)acetic acid: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorophenyl)acetic acid: was prepared in a manner as described for the Intermediate I-7 substituting I-1036 for I-3. ES/MZ: 425.1 (M+H+). Preparation of Intermediate I-1257 cis-isomer 1 tert-butyl (4-hydroxy-4-methyltetrahydrofuran-3-yl)carbamate) (Isomer 1 I-1257, Isomer 2 I-1258-1): was obtained via preparative chiral HPLC (Chiralpak IH column, Hexanes/iPrOH eluent) on racemic I-1257-0, giving two distinct stereoisomers (I-1257-1 as the earlier eluting isomer, I-1258-1 as the later eluting isomer). tert-butyl (4-hydroxy-4-methyltetrahydrofuran-3-yl)carbamate (I-1257-2): A mixture of tert-butyl (4-oxotetrahydrofuran-3-yl)carbamate (500 mg, 2.5 mmol) (Peak 1 I-1257-1) in diethyl ether (212.5 mL) was cooled to 0° C., then methylmagnesium bromide (2.48 mL, 3.0 M in diethyl ether, 7.45 mmol) was added slowly. The mixture was allowed to warm to 25° C. and allowed to stirred for 16 h. Then the mixture was diluted with diethyl ether and washed with brine. Then the mixture was dried over sodium sulfate and concentrated to yield the title compound. 1H NMR (400 MHz, CDCl3) δ 5.02 (s, 1H), 4.68 (d, J=8.1 Hz, 1H), 4.32-4.24 (m, 1H), 4.21-4.13 (m, 1H), 4.06-3.91 (m, 1H), 3.85-3.72 (m, 2H), 3.62-3.46 (m, 1H), 1.47 (d, J=3.5 Hz, 12H), 1.38 (s, 2H). 4-amino-3-methyltetrahydrofuran-3-ol (I-1257-3): A suspension of tert-butyl (4-hydroxy-4-methyltetrahydrofuran-3-yl)carbamate (491 mg, 2.26 mmol) (I-1257-2) and hydrochloric acid (1.25 mL, 4.0M in dioxane, 5.00 mmol) in DCM (5.00 mL) was stirred overnight. The mixture was diluted in EtOAc and washed with brine, dried over sodium sulfate, and concentrated to yield the title compound. 1H NMR (400 MHz, CDCl3) δ 3.95 (ddt, J=7.8, 3.8, 2.0 Hz, 1H), 3.77 (tq, J=7.2, 5.3, 3.7 Hz, 2H), 3.72-3.60 (m, 2H), 3.55 (dd, J=9.4, 2.0 Hz, 2H), 3.52-3.41 (m, 2H), 3.38 (t, J=1.8 Hz, 0H), 3.33-3.23 (m, 2H), 3.09 (t, J=6.2 Hz, 2H), 1.20 (t, J=1.8 Hz, 5H), 1.17 (d, J=2.0 Hz, 4H), 1.02-0.92 (m, 6H). methyl 3-((4-hydroxy-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (cis-isomer Peak 1, I-1257-4A, relative stereochemistry established): A suspension of 4-amino-3-methyltetrahydrofuran-3-ol (424 mg, 2.76 mmol) (I-1257-3), methyl 3-fluoro-4-nitro-benzoate (500 mg, 2.51 mmol) and N,N-Diisopropylethylamine (2.19 mL, 1.26 mmol) in THF (4 mL) and DMF (2 mL) was stirred at 82° C. for 16 h. Purification by silica gel flash column chromatography (EtOAc/hexane) provided both cis (I-1257-4A, Isomer 1, the earlier eluting of two diastereomers) and trans (I-1257-4B, Isomer 2, the later eluting of two diastereomers) isomers. ES/MZ: 297.1 (M+H+). Isomer 1, I-1257-4A: 1H NMR (400 MHz, CDCl3) δ 3.95 (ddt, J=7.8, 3.8, 2.0 Hz, 1H), 3.77 (tq, J=7.2, 5.3, 3.7 Hz, 2H), 3.72-3.60 (m, 2H), 3.55 (dd, J=9.4, 2.0 Hz, 2H), 3.52-3.41 (m, 2H), 3.38 (t, J=1.8 Hz, 0H), 3.33-3.23 (m, 2H), 3.09 (t, J=6.2 Hz, 2H), 1.20 (t, J=1.8 Hz, 5H), 1.17 (d, J=2.0 Hz, 4H), 1.02-0.92 (m, 6H). Two dimensional NOESY NMR identified a O17-OH to N11-NH NOE correlation and a C12-CH to C21-CH3NOE correlation to confirm relative stereochemistry of I-1257-4A as the cis diastereomer. Isomer 2, I-1257-4B: 1H NMR (400 MHz, CDCl3) δ 8.27 (d, J=8.9 Hz, 1H), 8.11 (d, J=7.7 Hz, 1H), 7.75 (d, J=1.7 Hz, 1H), 7.33 (dd, J=8.9, 1.7 Hz, 1H), 4.53 (dd, J=9.6, 6.3 Hz, 1H), 4.24 (q, J=7.0, 6.4 Hz, 1H), 3.98 (s, 3H), 3.91 (d, J=9.8 Hz, 1H), 3.80 (d, J=9.8 Hz, 1H), 3.73 (dd, J=9.6, 4.6 Hz, 1H), 1.42 (s, 3H). Methyl 3-((cis-4-methoxy-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (cis-isomer peak 1, I-1257-5, relative stereochemistry established): A mixture of methyl 3-((4-hydroxy-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (144 mg, 0.49 mmol) (cis-isomer peak 1, I-1257-4A), sodium hydride (20.5 mg, 0.54 mmol) and iodomethane (33 uL, 0.54 mmol) in DMF (1.00 mL) was stirred at 0° C. for 15 min then warmed to 25° C. and stirred for 16 h. The mixture was diluted in EtOAc and washed with brine, then dried over sodium sulfate, concentrated, and purified by silica gel flash column chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 311.3 (M+H+). Methyl 4-amino-3-((4-methoxy-4-methyltetrahydrofuran-3-yl)amino)benzoate (cis-isomer peak 1, I-1257, relative stereochemistry established): A mixture of methyl 3-((4-methoxy-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (133 mg, 0.43 mmol) (cis-isomer peak 1, I-1257-5), and 10% palladium on carbon (45.5 mg, 0.043 mmol) in ethanol (5.0 mL) and placed under hydrogen at 25° C. for 5 h. The mixture was diluted in EtOAc and washed with brine, dried over sodium sulfate, and concentrated to yield the title compound. ES/MS: 281.1 (M+H+) Preparation of Intermediate I-1258 cis-isomer 2 Methyl 4-amino-3-((4-methoxy-4-methyltetrahydrofuran-3-yl)amino)benzoate (cis-isomer 2, I-1258, relative stereochemistry established): methyl 4-amino-3-((4-methoxy-4-methyltetrahydrofuran-3-yl)amino)benzoate (cis-isomer 2, I-1258, relative stereochemistry established) was prepared in a manner as described for cis-isomer peak 1 I-1257, using Intermediate isomer 2 I-1258-1 in place of isomer 1 I-1257-1. Preparation of Intermediate I-1260 2-bromo-6-((4-chloro-2-(trifluoromethyl)benzyl)oxy)pyridine: 2-bromo-6-((4-chloro-2-(trifluoromethyl)benzyl)oxy)pyridine benzonitrile was prepared in a manner as described for the Intermediate I-1034 substituting 1-(bromomethyl)-4-chloro-2-(trifluoromethyl)benzene for 6-(bromomethyl)-1-methyl-benzotriazole. ES/MZ: 366.6 (M+H+). Preparation of Intermediate I-1261 6-bromo-2-((2,6-difluorobenzyl)oxy)-3-fluoropyridine: 6-bromo-2-((2,6-difluorobenzyl)oxy)-3-fluoropyridine was prepared in a manner as described for the Intermediate I-1034 substituting 3-(((6-bromopyridin-2-yl)oxy)methyl)-6-(trifluoromethyl)pyridazine for 6-(bromomethyl)-1-methyl-benzotriazole ES/MZ: 334.1 (M+H+). Preparation of Intermediate I-1262 6-bromo-2-((2,6-difluorobenzyl)oxy)-3-fluoropyridine: 6-bromo-2-((2,6-difluorobenzyl)oxy)-3-fluoropyridine was prepared in a manner as described for the Intermediate I-1034 substituting 2-(bromomethyl)-1,3-difluorobenzene for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromopyridin-2-ol for 6-bromo-3-fluoropyridin-2-ol. ES/MZ: 318.1 (M+H+). Preparation of Intermediate I-1263 2-bromo-6-((2,6-difluorobenzyl)oxy)pyridine: 2-bromo-6-((2,6-difluorobenzyl)oxy)pyridine was prepared in a manner as described for the intermediate 1-1034 substituting 2-(bromomethyl)-1,3-difluorobenzene for 6-(bromomethyl)-1-methyl-benzotriazole. ES/MZ: 300.1 (M+H+). Preparation of Intermediate I-1264 6-bromo-3-fluoro-2-((2-fluorobenzyl)oxy)pyridine: 6-bromo-3-fluoro-2-((2-fluorobenzyl)oxy)pyridine was prepared in a manner as described for the Intermediate I-1034 substituting 1-(bromomethyl)-2-fluorobenzene for 6-(bromomethyl)-1-methyl-benzotriazole and 6-bromopyridin-2-ol for 6-bromo-3-fluoropyridin-2-ol. ES/MZ: 300.1 (M+H+). Preparation of Intermediate I-1265 2-bromo-6-((2-fluorobenzyl)oxy)pyridine: 2-bromo-6-((2-fluorobenzyl)oxy)pyridine was prepared in a manner as described for the Intermediate I-1034 substituting 1-(bromomethyl)-2-fluorobenzene for 6-(bromomethyl)-1-methyl-benzotriazole. ES/MZ: 282.1 (M+H+). Preparation of Intermediate I-1266 Methyl rac-cis-4-amino-3-(((1-(fluoromethyl)cyclopropyl)methyl)amino)benzoate (I-1065-1): To a mixture of methyl 3-fluoro-4-nitro-benzoate (120 mg, 0.603 mmol), racemic cis-4-aminotetrahydrofuran-3-yl]methanol (77.7 mg, 0.66 mmol) in THE (4 mL) and DMF (2 mL), was added N,N-diisopropylethylamine (0.525 mL, 3.01 mmol). The mixture was heated at 80° C. for 18 hr. The crude mixture was diluted with EtOAc, washed with 5% LiCl and brine. The organic extract was dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford the title compound. ES/MS: 297.2 (M+H+). Methyl 3-((cis-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)-4-nitrobenzoate (Peak 1-I-1266-1A, Peak 2-I-1266-1B, relative stereochemistry established): was obtained via preparative chiral SFC (Daicel Chiralpak AD-H column with EtOH/CO2eluent) on I-1065-1, which gave 2 distinct stereoisomers. Methyl 3-(((3R,4R)-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)-4-nitrobenzoate (Peak 1, I-1266-1A): the earlier eluting of the two stereoisomers. Methyl 3-(((3S,4S)-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)-4-nitrobenzoate (Peak 2, I-1266-1B): the later eluting of the two stereoisomers. Methyl 3-(((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1266-2): To a mixture of methyl 3-(((3S,4S)-4-(hydroxymethyl)tetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1266-1A) (75 mg, 0.253 mmol) in DMF (4 mL) was added NaH (24.2 mg, 0.633 mmol, 60% dispersion) at 0° C. To this mixture was added methyl iodide (35.9 mg, 0.253 mmol) and the mixture stirred for 1 hour at rt. The mixture was then split between saturated aqueous ammonium chloride and EtOAc, the aqueous layer was extracted with EtOAc five times, washed with brine. The organic extract was dried over sodium sulfate and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to afford the title compound. ES/MS: 311.2 (M+H+) Methyl 4-amino-3-(((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)amino)benzoate (I-1266, relative stereochemistry established): A mixture of methyl 3-(((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1266-2) (80 mg, 0.258 mmol) in EtOAc (5 mL) was degassed with cycles of argon then vacuum 3×. Added Palladium on carbon (10.0%, 27.4 mg, 0.258 mmol). The mixture was degassed with cycles of argon then vacuum and Preparation of Intermediate I-1267 Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate: Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described for the Intermediate I-82, substituting methyl 4-amino-3-(((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)amino)benzoate for I-80. Preparation of Intermediate I-1268 2-bromo-6-[[6-(triazol-1-yl)-3-pyridyl]methoxy]pyridine (I-1268): 2-bromo-6-[[6-(triazol-1-yl)-3-pyridyl]methoxy]pyridine was prepared in a manner as described for Intermediate I-84 substituting 5-(bromomethyl)-2-(triazol-1-yl)pyridine for 2-(bromomethyl)thiazole-5-carbonitrile. ES/MS: 332, 334 (M+H+). Preparation of Intermediate I-1269 Ethyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1269): A suspension of ethyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (300 mg, 0.603 mmol), 5-bromo-2-(chloromethyl)-3-fluoro-pyridine (162 mg, 0.724 mmol), and cesium carbonate (491 mg, 1.51 mmol) in CH3CN was heated at 70° C. for 1 hr. The mixture was filtered, concentrated, and purified by silica gel column chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 686.0 (M+H+). Preparation of Intermediate I-1270 Methyl 4-amino-3-fluoro-5-[[(2S)-2-methoxypropyl]amino]benzoate (I-1270): Methyl 4-amino-3-fluoro-5-[[(2S)-2-methoxypropyl]amino]benzoate was prepared in a manner as described for Intermediate I-5 substituting (S)-2-methoxypropan-1-amine for (S)-oxetan-2-ylmethanamine and methyl 3,5-difluoro-4-nitrobenzoate for Ethyl 3,5-difluoro-4-nitrobenzoate. ES/MS: 257.1 (M+H+). Preparation of Intermediate I-1271 Ethyl 4-amino-3-fluoro-5-[[(2R)-2-methoxypropyl]amino]benzoate (I-1271) Ethyl 4-amino-3-fluoro-5-[[(2R)-2-methoxypropyl]amino]benzoate was prepared in a manner as described for Intermediate I-5 substituting (R)-2-methoxypropan-1-amine for (S)-oxetan-2-ylmethanamine. ES/MS: 271.2 (M+H+). Preparation of Intermediate I-1272 Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-7-fluoro-3-[(2R)-2-methoxypropyl]benzimidazole-5-carboxylate (I-1272): Ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-7-fluoro-3-[(2R)-2-methoxypropyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting Ethyl 4-amino-3-fluoro-5-[[(2R)-2-methoxypropyl]amino] (I-1271) for I-1 and 2-(4-bromo-2,5-difluorophenyl)acetic acid for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 485, 487 (M+H+). Preparation of Intermediate I-1273 Methyl 5-(bromomethyl)-4-chloro-pyridine-2-carboxylate (I-1273-1): Methyl 4-chloro-5-methyl-pyridine-2-carboxylate (1.00 g, 5.39 mmol) was taken up in carbon tetrachloride (10.0 mL) and N-Bromosuccinimide (1255 mg, 7.05 mmol) was added followed by Benzoyl peroxide (140 mg, 0.580 mmol). The mixture was heated to 90° C. for 45 min. The mixture was concentrated. in vacuo. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 264, 266 (M+H+). Methyl 5-[(6-bromo-2-pyridyl)oxymethyl]-4-chloro-pyridine-2-carboxylate (I-1273-2): 6-Bromopyridin-2-ol (600 mg, 3.4 mmol), methyl 5-(bromomethyl)-4-chloro-pyridine-2-carboxylate (1003 mg, 3.8 mmol), and cesium carbonate (1685 mg, 5.2 mmol) were taken up in Acetonitrile (12 mL) and the mixture was heated to 65° C. for 1 hr. The mixture was filtered through Celite and concentrated in vacuo. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 357, 359 (M+H+). 5-[(6-bromo-2-pyridyl)oxymethyl]-4-chloro-pyridine-2-carboxylic acid (I-1273-3): Methyl 5-[(6-bromo-2-pyridyl)oxymethyl]-4-chloro-pyridine-2-carboxylate (1.17 g, 3.27 mmol) was taken up in CH3CN (15.0 mL) and water (5.00 mL) and then lithium hydroxide, monohydrate (408 mg, 9.72 mmol) was added. The mixture was stirred at rt for 1 hr. The mixture was diluted with EtOAc then acidified to pH-6 with 1N HCl. A precipitate formed. The precipitate was collected and washed with H2O and Et2O. Remaining material extracted into EtOAc (dried over MgSO4and conc. in vacuo). The combined solids were used in subsequent reactions without purification. ES/MS: 343, 345 (M+H+). 5-[(6-bromo-2-pyridyl)oxymethyl]-4-chloro-N-methyl-pyridine-2-carboxamide (I-1273): N,N-Diisopropylethylamine (1.22 mL, 6.99 mmol) was added to a solution of 5-[(6-bromo-2-pyridyl)oxymethyl]-4-chloro-pyridine-2-carboxylic acid (600 mg, 1.75 mmol), methanamine; hydrochloride (236 mg, 3.49 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (976 mg, 2.57 mmol) in DMF (10 mL). The mixture was stirred at rt for 10 min. The mixture was partitioned with EtOAc and water. The organic extract was dried over MgSO4, filtered, and concentrated to give desired product. ES/MS: 356, 358 (M+H+). Preparation of Intermediate I-1274 Methyl 5-[(5-bromo-2-fluoro-phenoxy)methyl]-4-chloro-pyridine-2-carboxylate (I-1274): Methyl 5-[(5-bromo-2-fluoro-phenoxy)methyl]-4-chloro-pyridine-2-carboxylate was prepared in a similar manner as described for Intermediate I-1273 substituting 5-bromo-2-fluoro-phenol for 6-bromopyridin-2-ol in step 2. ES/MS: 374, 376 (M+H+). Preparation of Intermediate I-1275 Ethyl 2-[(4-bromo-2-fluoro-5-methyl-phenyl)methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1275): ethyl 2-[(4-bromo-2-fluoro-5-methyl-phenyl)methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-2 substituting 1-5 for I-1 and 2-(4-bromo-2-fluoro-5-methyl-phenyl)acetic acid 1-1277 for 2-(4-bromo-2-fluoro-phenyl)acetic acid. ES/MS: 479, 481 (M+H+). Preparation of Intermediate I-1276 (4-bromo-2-fluoro-5-methyl-phenyl)methanol (I-1276): Sodium borohydride (436 mg, 11.5 mmol) was added in one portion to a solution of 4-bromo-2-fluoro-5-methyl-benzaldehyde (5.0 g, 23.0 mmol) in EtOH (115 mL) at 0° C. The mixture was placed under an inert atmosphere of argon and allowed to warm to room temperature and stirred for 2 h. The mixture was then quenched with saturated sodium NaHCO3(2×30 ml), extracted with EtOAc (2×30 ml) and dried over magnesium sulfate. The mixture was then filtered and concentrated in vacuo. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product.1HNMR (400 MHz, CDCl3) δ 7.29 (d, J=7.8 Hz, 1H), 7.25 (d, J=9.3 Hz, 1H), 4.68 (s, 2H), 2.36 (s, 3H).19F NMR (376 MHz, Chloroform-d) δ −122.82. Preparation of Intermediate I-1277 2-(4-bromo-2-fluoro-5-methyl-phenyl)acetic acid (I-1277): 2-(4-bromo-2-fluoro-5-methyl-phenyl)acetic acid was prepared in a manner as described for Intermediate I-1023 substituting (4-bromo-2-fluoro-5-methyl-phenyl)methanol 1-1276 for (4-bromo-2-chloro-5-methyl-phenyl)methanol in step 3.1H NMR (400 MHz, CDCl3) δ 7.28 (d, J=9.0 Hz, 1H), 7.12 (d, J=7.8 Hz, 1H), 3.64 (s, 2H), 2.34 (s, 3H).19F NMR (376 MHz, CDCl3) δ −119.92. Preparation of Intermediate I-1278 Ethyl 2-[(4-bromo-2-chloro-5-fluoro-phenyl)methyl]-7-fluoro-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-1278): Ethyl 2-[(4-bromo-2-chloro-5-fluoro-phenyl)methyl]-7-fluoro-3-(2-methoxyethyl)benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-26 substituting ethyl 4-amino-3-fluoro-5-((2-methoxyethyl)amino)benzoate 1-1032 for ethyl 4-amino-3-fluoro-5-[[(2S)-oxetan-2-yl]methylamino]benzoate and 2-(4-bromo-2-chloro-5-fluoro-phenyl)acetic acid for 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid. ES/MS: 488, 489 (M+H+). Preparation of Intermediate I-1279 2-benzyloxy-4-bromo-1-fluoro-benzene: A suspension of 5-bromo-2-fluoro-phenol (5.00 g, 0.0262 mol), Benzyl bromide, reagent grade, 98% (3.74 mL, 0.0314 mol), and potassium carbonate (7.96 g, 0.0576 mol) in acetone (30 mL) was heated at 70° C. for 1 hr. The mixture was partitioned with EtOAc and water. The organic extract was dried over MgSO4, concentrated and purified by chromatography (eluent: EtOAc/hexanes) to give desired product. Ethyl 2-[[4-(3-benzyloxy-4-fluoro-phenyl)-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate: A suspension of ethyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (500 mg, 1.03 mmol) (I-14), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II); PdCl2(dppf) (76.7 mg, 0.103 mmol), potassium propionate (348 mg, 3.10 mmol), and Bis(pinacolato)diboron (394 mg, 1.55 mmol) was degassed, then heated at 110° C. for 2 hr. Next, sodium carbonate (2000 mmol/L, 1.03 mL, 2.07 mmol), 2-benzyloxy-4-bromo-1-fluoro-benzene (320 mg, 1.14 mmol), and [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II); PdCl2(dppf) (38.4 mg, 0.0517 mmol) were added and followed by degassing. The reaction was heated at 100° C. for 1 hr. The mixture was diluted with EtOAc and water. The organic extract was dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 606.1 (M+H+). Ethyl 2-[[2,5-difluoro-4-(4-fluoro-3-hydroxy-phenyl)phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1279): A suspension of ethyl 2-[[4-(3-benzyloxy-4-fluoro-phenyl)-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (626 mg, 1.04 mmol) and Palladium on carbon 10 wt. % (5.00%, 2204 mg, 1.04 mmol) was degassed by cycling the mixture between argon and vacuum 3×. Next, the mixture was stirred at rt with a balloon of hydrogen for 2 hr. The mixture was then filtered through Celite and concentrated in vacuo to give desired product. ES/MS: 515.6 (M+H+) Preparation of Intermediate I-1281 Ethyl 2-[[4-[6-[(6-bromo-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (I-1281): Ethyl 2-[[4-[6-[(6-bromo-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-1269 substituting 2-bromo-5-(bromomethyl)pyridine for 5-bromo-2-(chloromethyl)-3-fluoro-pyridine. ES/MS: 667, 669 (M+H+). Preparation of Intermediate I-1282 [2-fluoro-4-(triazol-1-yl)phenyl]methanol: A suspension of ethynyl(trimethyl)silane (0.372 mL, 0.00269 mol), (4-azido-2-fluoro-phenyl)methanol (300 mg, 0.00179 mol), sodium ascorbate (0.126 g, 0.000718 mol), copper sulfate monohydrate (0.0638 g, 0.000359 mol), and potassium carbonate (0.298 g, 0.00215 mol) was stirred at rt overnight. The mixture was partitioned with EtOAc and water. The organic extract was dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 194.1 (M+H+). 1-[4-(bromomethyl)-3-fluoro-phenyl]triazole (I-1282): A solution of [2-fluoro-4-(triazol-1-yl)phenyl]methanol (245 mg, 0.00127 mol) and phosphorus tribromide (0.149 mL, 0.00159 mol) in DCM (2 mL) was stirred at rt for 3 hr. The mixture was carefully quenched with saturated NaHCO3solution and partitioned with EtOAc and water. The organic extract was dried over magnesium sulfate and concentrated to give desired product, which was used without further purification. ES/MS: 256, 258 (M+H+). Preparation of Intermediate I-1283 2-bromo-6-[[2-fluoro-4-(triazol-1-yl)phenyl]methoxy]pyridine (I-1283): 2-bromo-6-[[2-fluoro-4-(triazol-1-yl)phenyl]methoxy]pyridine was prepared in a manner as described for Intermediate I-84 substituting 1-[4-(bromomethyl)-3-fluoro-phenyl]triazole 1-1282 for 2-(bromomethyl)thiazole-5-carbonitrile. ES/MS: 349, 351 (M+H+). Preparation of Intermediate I-1284 (4-chloro-2,5-difluorophenyl)methanol: To a solution of 4-chloro-2,5-difluorobenzoic acid (10.0 g, 51.9 mmol) and B(OMe)3(15.6 g, 150 mmol, 17.0 mL) in THE (50.0 mL) at 0° C. was added BH3-Me2S (10 M, 7.79 mL). The mixture was stirred at 20° C. for 16 h. The reaction was poured into 1N NaOH (100 mL) and extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (50 mL), dried over sodium sulfate, and concentrated to dryness. The crude residue was recrystallized from petroleum ether to give desired product. 1H NMR (CDCl3400 MHz): δ 7.19-7.23 (m, 1H), 7.05 (dd, J=8.99, 5.93 Hz, 1H), 4.66 (d, J=5.62 Hz, 2H), 1.76 (t, J=5.93 Hz, 1H). 2-bromo-6-[(4-chloro-2,5-difluoro-phenyl)methoxy]pyridine (I-1284): To a solution of (4-chloro-2,5-difluorophenyl)methanol (6.0 g, 33.6 mmol) in THE (60 mL) at 0° C. was slowly added KOtBu (1.0 M, 50.4 mL, 50.4 mmol). The mixture was stirred at 20° C. for 1 hr, then cooled to 0° C. and added a solution of 2,6-dibromopyridine (7.16 g, 30.2 mmol) in THE (20 mL). The reaction was stirred at 20° C. for 1 hr, then poured into saturated NH4Cl solution and extracted 3× with EtOAc. The organic extracts were washed with brine, dried over magnesium sulfate and concentrated. The crude residue was triturated with petroleum ether, and the resulting suspension was filtered to give desired product. ES/MS: 332.9 (M+H+).1H NMR (CDCl3400 MHz): δ 7.48 (t, J=7.82 Hz, 1H), 7.35 (dd, J=8.93, 6.24 Hz, 1H), 7.19 (dd, J=8.80, 5.87 Hz, 1H), 7.13 (d, J=7.46 Hz, 1H), 6.78 (d, J=8.19 Hz, 1H), 5.40 (s, 2H).19F NMR (CDCl3374 MHz): δ −120.89-−120.98 (m, 1F), −121.30-−121.40 (m, 1F). Preparation of Intermediate I-1285 Methyl 4-amino-3-((4-cyclopropyltetrahydrofuran-3-yl)amino)benzoate (I-1285): methyl 4-amino-3-((4-cyclopropyltetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-68 substituting 4-cyclopropyltetrahydrofuran-3-amine hydrochloride for 2-methoxyethylamine. ES/MS: 277.2 (M+H+). Preparation of Intermediate I-1286 4-(((2-chloropyrimidin-4-yl)oxy)methyl)-3-fluorobenzonitrile (I-1286): 4-(((2-chloropyrimidin-4-yl)oxy)methyl)-3-fluorobenzonitrile was prepared in a manner as described for Intermediate I-1129 substituting 4-(bromomethyl)-3-fluorobenzonitrile for 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine and 2-chloropyrimidin-4-ol for 6-bromopyridin-2-ol. ES/MS: 264.0 (M+H+). Preparation of Intermediate I-1287 4-[(6-chloro-3-fluoro-2-pyridyl)oxymethyl]benzonitrile (I-1287): A mixture of 6-chloro-3-fluoro-pyridin-2-ol (265 mg, 1.0 equivalent), 4-(bromomethyl)benzonitrile (370 mg, 1.05 equivalent), cesium carbonate (0.878 g, 1.5 equivalent) in acetonitrile (9 mL) was stirred at in metal heating block at 90° C. until all starting material was consumed as determined by LCMS. The mixture was filtered, concentrated in vacuo, and purified by silica gel flash column chromatography to yield the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.68 (dd, J=8.2, 1.2 Hz, 2H), 7.59 (d, J=7.9 Hz, 2H), 7.39-7.30 (m, 1H), 6.90 (ddd, J=8.1, 2.7, 1.1 Hz, 1H), 5.49 (s, 2H). Preparation of Intermediate I-1288 Racemic (3S,4R)-4-methyltetrahydrofuran-3-amine (I-1288): Diisopropyl azodicarboxylate (0.74 mL, 1.2 equivalent) was added to a mixture of racemic (3R,4S)-4-methyltetrahydrofuran-3-ol (320 mg, 1.0 equivalent), triphenylphosphine (985 mg, 1.2 equivalent), diisopropylethylamine (0.56 mL, 1.02 equivalent) in THE (15 mL) precooled to 0° C. equivalent. The mixture was stirred at 0° C. for 15 min, then diphenylphosphoryl azide (0.81 mL, 1.2 equivalent) was added. The mixture was allowed to warm to 20° C. with stirring for 3 hr., then cooled back to 0° C. Additional triphenylphosphine (1.06 g, 1.3 equivalent) as a solution in THE (6 mL) was added to the mixture, and the mixture was allowed to stir at 20° C. for 2.5 hr. Upon completion of time, water (1.5 mL) was added, and the mixture was stirred at 65° C. for 18 hr. (OPERATIONAL NOTE: significant gas evolution was observed). The mixture was cooled and partitioned between ethyl acetate and a mixture of 1 N aqueous NaOH, saturated aqueous NaHCO3and brine; extracted aqueous phase twice more with EtOAc. The combined organic phases were dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound, which was carried forward crude without further purification. ES/MS m/z: 101.9 (M+H+). Preparation of Intermediate I-1289 Racemic methyl 3-[[(3R,4S)-4-methyltetrahydrofuran-3-yl]amino]-4-nitrobenzoate (I-1289-1): Methyl 3-fluoro-4-nitrobenzoate (687 mg, 1.1 equivalent), THE (8.8 mL), DMF (4.4 mL), and diisopropylethylamine (2.73 mL, 5.0 equivalent) were added to a crude racemic (3R,4S)-4-methyltetrahydrofuran-3-amine (I-1288, 317 mg, 1.0 equivalent)equivalent. The mixture was stirred at 80° C. for 16 hr., or until completion as determined by LCMS. The mixture was then diluted with ethyl acetate and washed with saturated aqueous NH4Cl, then brine, then dried over magnesium sulfate, filtered and concentrated in vacuo to yield the title compound. Racemic methyl 4-amino-3-[[(3R,4S)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1289): A mixture of crude racemic methyl 3-[[(3R,4S)-4-methyltetrahydrofuran-3-yl]amino]-4-nitrobenzoate (I-1289-1, 878 mg, 1.0 equivalent), 10% palladium on carbon (333 mg, 0.10 equivalent) and ethanol (60 mL) was stirred under an atmosphere of hydrogen for 6 hr., or until completion as determined by LCMS. The mixture was filtered, concentrated in vacuo and purified by silica gel flash column chromatography (hexane/EtOAc) to yield the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.45 (dd, J=8.1, 1.8 Hz, 1H), 7.34 (d, J=1.9 Hz, 1H), 6.69 (d, J=8.1 Hz, 1H), 4.09-4.02 (m, 3H), 3.85 (s, 3H), 3.68 (h, J=3.8 Hz, 1H), 3.57 (dd, J=8.5, 7.2 Hz, 1H), 2.66-2.54 (m, 1H), 1.01 (d, J=7.0 Hz, 3H). ES/MS m/z: 251.0 (M+H+). Preparation of Intermediate I-1290, I-1291 Methyl 4-amino-3-[[(3S,4R)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1290), Methyl 4-amino-3-[[(3R,4S)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1291): Racemic I-1289 was purified by preparative chiral SFC (IG, 20% MeOH) to yield methyl 4-amino-3-[[(3S,4R)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1290) as the earlier eluting isomer (Peak 1) and methyl 4-amino-3-[[(3R,4S)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1291) as the later-eluting isomer (Peak 2). Methyl 4-amino-3-[[(3S,4R)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1290): ES/MS m/z: 251.0 (M+H+). Methyl 4-amino-3-[[(3R,4S)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1291): ES/MS m/z: 251.0 (M+H+). Preparation of Intermediate I-1292 Methyl 4-[[2-(4-bromo-2,5-difluoro-phenyl)acetyl]amino]-3-[[(3S,4R)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1292-1): A mixture of methyl 4-amino-3-[[(3S,4R)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1290, 57 mg, 1.0 equivalent), 2-(4-bromo-2,5-difluorophenyl)acetic acid (63 mg, 1.1 equivalent), chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (77 mg, 1.2 equivalent), 1-methylimidazole (94 mg, 5.0 equivalent) and acetonitrile (2.3 mL) was stirred at 20° C. for 1 hr. The mixture was then diluted with EtOAc, washed with saturated aqueous NH4Cl. Next, the mixture was saturated aqueous NaHCO3, then brine. The organic phase was dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound, which was carried forward without further purification. ES/MS m/z: 483.2 (M+H+). Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S,4R)-4-methyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1292): A mixture of methyl 4-[[2-(4-bromo-2,5-difluoro-phenyl)acetyl]amino]-3-[[(3S,4R)-4-methyltetrahydrofuran-3-yl]amino]benzoate (I-1292-1, 110 mg, 1.0 equivalent), glacial acetic acid (0.91 mL) and 1,2-dichloroethane (4.6 mL) was stirred at 80-95° C. for 48 hr., or until completion as determined by LCMS. The mixture was diluted with EtOAc, quenched with saturated aqueous NaHCO3and batchwise addition of solid NaHCO3. The organic phase was washed with brine, dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound. ES/MS m/z: 465.2 (M+H+). Preparation of Intermediate I-1293 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3R,4S)-4-methyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1293): The title compound was prepared in a similar manner as described for Intermediate I-1292, substituting Intermediate I-1291 in place of Intermediate I-1290. ES/MS m/z: 465.2 (M+H+). Preparation of Intermediate I-1294 2,5-difluoro-4-(hydroxymethyl)benzonitrile (I-1294-1): Sodium borohydride (1.35 g, 1.05 equivalent) was added to a mixture of 2,5-difluoro-4-formylbenzonitrile (5.66 g, 1.0 equivalent) in methanol (100 mL). equivalent. The mixture was stirred at 20° C. for 2 hr., then concentrated in vacuo. The crude material was quenched with slow addition of saturated aqueous ammonium chloride, then extracted with ethyl acetate. The organic phases were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to yield the title compound, which was carried forward without further purification. 1H NMR (400 MHz, CDCl3) δ 7.43 (dd, J=8.9, 5.6 Hz, 1H), 7.28 (dd, J=8.5, 4.9 Hz, 1H), 4.84 (d, J=4.8 Hz, 2H), 1.96 (t, J=5.7 Hz, 1H). ES/MS m/z: 170.2 (M+H+). 4-(bromomethyl)-2,5-difluorobenzonitrile (I-1294-2): Carbon tetrabromide (412 mg, 1.05 equivalent) was added to a mixture of 2,5-difluoro-4-(hydroxymethyl)benzonitrile (I-1294-1, 200 mg, 1.0 equivalent), triphenylphosphine (326 mg, 1.05 equivalent) and DCM (11 mL) precooled to 0° C. equivalent. The mixture was allowed to warm to 20° C. with stirring for 15 min, then the mixture was purified directly by silica gel flash column chromatography (EtOAc/hexane) to yield the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.40-7.28 (m, 2H), 4.45 (s, 2H). 4-[(6-chloro-3-fluoro-2-pyridyl)oxymethyl]-2,5-difluorobenzonitrile (I-1294): A mixture of 4-(bromomethyl)-2,5-difluorobenzonitrile (I-1294-2, 182 mg, 1.05 equivalent), 6-chloro-3-fluoropyridin-2-ol (110 mg, 1.0 equivalent), cesium carbonate (364 mg, 1.5 equivalent) and acetonitrile (3.8 mL) was stirred at 90° C. for 1 hr. The mixture was then diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.47 (dd, J=8.7, 5.5 Hz, 1H), 7.43-7.35 (m, 2H), 6.97 (dd, J=8.2, 2.6 Hz, 1H), 5.55 (s, 2H). Preparation of Intermediate I-1295 2,5-difluoro-4-((methylsulfinyl)(methylthio)methyl)benzonitrile (I-1295-1): n-BuLi (2.50 M in hexane, 134 mL, 2.1 equivalent) was added to a mixture of methyl methylthiomethyl sulfoxide (39.5 g, 1.0 equivalent) in THE (380 mL) at −80° C. equivalent The mixture was stirred at −80° C. for 30 min, then a solution of 2,4,5-trifluorobenzonitrile (25.0 g, 1.00 equivalent) in THF (380 mL) was added. The mixture was stirred at −80° C. for 30 min, with monitoring by TLC. The mixture was then quenched with water and extracted with ethyl acetate. The organic phase was washed with brine, then concentrated in vacuo. Silica gel flash column chromatography (EtOAc/petroleum ether) yielded the title compound. 1H NMR (CDCl3400 MHz) δ=7.47-7.32 (m, 2H), 4.98-4.84 (m, 1H), 2.56-2.50 (m, 3H), 2.42-2.34 (m, 3H). 2,5-difluoro-4-formylbenzonitrile (I-1295-2): A mixture of 2,5-difluoro-4-((methylsulfinyl)(methylthio)methyl)benzonitrile (I-1295-1, 34.0 g, 1.00 equivalent) and concentrated sulfuric acid (42.9 mL, 6.18 equivalent) was stirred at 25° C. for 60 hr. The mixture was then partitioned between water and ethyl acetate. The organic phase was washed with brine, then concentrated in vacuo to yield the title compound. 1H NMR (CDCl3400 MHz) δ=10.34 (d, J=2.6 Hz, 1H), 7.70 (dd, J=5.3, 7.9 Hz, 1H), 7.53 (dd, J=4.6, 8.6 Hz, 1H). 2,5-difluoro-4-(hydroxymethyl)benzonitrile (I-1295-3): Sodium borohydride (4.98 g, 1.1 equivalent) was added To a mixture of 2,5-difluoro-4-formylbenzonitrile (I-1295-2, 20.0 g, 1.0 equivalent) in methanol (300 mL) pre-cooled to 0° C. The mixture was stirred for 1 hr., with monitoring by TLC. The mixture was quenched with 1N HCl, then the mixture was concentrated in vacuo. The resulting mixture was partitioned between water and ethyl acetate. The organic phase was washed with brine, then concentrated in vacuo. Silica gel flash column chromatography (EtOAc/petroleum ether) yielded the title compound.1H NMR (CDCl3400 MHz) δ=7.89 (dd, J=5.2, 9.2 Hz, 1H), 7.51 (dd, J=5.6, 9.6 Hz, 1H), 5.65 (t, J=5.7 Hz, 1H), 4.59 (td, J=1.2, 5.7 Hz, 2H). 4-(((6-bromopyridin-2-yl)oxy)methyl)-2,5-difluorobenzonitrile (I-1295): Triphenylphosphine (9.31 g, 1.2 equivalent), then diisopropyl azodicarboxylate (7.17 g, 1.2 equivalent) were added to a mixture of 2,5-difluoro-4-(hydroxymethyl)benzonitrile (I-1295-3, 5.00 g, 1.00 equivalent) and 6-bromo-2-pyridinol (5.14 g, 1.00 equivalent) in THF (125 mL)equivalent. The mixture was stirred at 25° C. for 1 hr., with monitoring by TLC. The mixture was partitioned between water and ethyl acetate. The organic phase was washed with brine, then concentrated in vacuo. Silica gel flash column chromatography (EtOAc/petroleum ether) yielded the title compound.1H NMR (CDCl3400 MHz) δ=7.49 (t, J=7.8 Hz, 1H), 7.42 (dd, J=5.5, 8.6 Hz, 1H), 7.34 (dd, J=5.0, 8.3 Hz, 1H), 7.14 (d, J=7.5 Hz, 1H), 6.80 (d, J=8.2 Hz, 1H), 5.46 (s, 2H). ES/MS m/z: 324.0 (M+H+). Preparation of Intermediate I-1296 2-bromo-6-((4-chloro-2,3-difluorobenzyl)oxy)pyridine (I-1296): A mixture of 1-(bromomethyl)-4-chloro-2,3-difluorobenzene (729 mg, 1.05 equivalent), 6-bromo-2-pyridinol (500 mg, 1.00 equivalent), cesium carbonate (1.40 g, 1.5 equivalent) and acetonitrile (14 mL) was stirred at 80° C. for 3 hr. The mixture was then diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.45 (dd, J=8.2, 7.5 Hz, 1H), 7.25-7.21 (m, 1H), 7.18 (ddd, J=8.4, 6.1, 1.7 Hz, 1H), 7.10 (dd, J=7.5, 0.7 Hz, 1H), 6.73 (dd, J=8.2, 0.7 Hz, 1H), 5.41 (d, J=1.4 Hz, 2H). ES/MS m/z: 333.9 (M+H+). Preparation of Intermediate I-1297 2-bromo-6-((4-chloro-2,6-difluorobenzyl)oxy)pyridine (I-1297): A mixture of 1-(bromomethyl)-4-chloro-2,6-difluorobenzene (305 mg, 1.1 equivalent), 6-bromo-2-pyridinol (200 mg, 1.00 equivalent), cesium carbonate (600 mg, 1.5 equivalent) and acetonitrile (5.5 mL) was stirred at 80° C. for 6 hr. The mixture was then diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.43 (dd, J=8.2, 7.5 Hz, 1H), 7.10 (dd, J=7.5, 0.6 Hz, 1H), 7.03-6.93 (m, 2H), 6.69 (dd, J=8.2, 0.6 Hz, 1H), 5.38 (s, 2H). ES/MS m/z: 335.8 (M+H+). Preparation of Intermediate I-1298 (4-chloro-2,5-difluorophenyl)methanol (I-1298-1): BH3-Me2S (7.79 mL, 1.5 equivalent) was added to a mixture of 4-chloro-2,5-difluorobenzoic acid (10.0 g, 1.00 equivalent) and B(OMe)3(15.6 g, 2.9 equivalent) in THF (50 mL) pre-cooled to 0° C. The mixture was allowed to warm to 20° C. and stirred for 16 hr., with monitoring by TLC. The mixture was then poured into 1 N aqueous sodium hydroxide and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. Next, recrystallization from petroleum ether yielded the title compound.1H NMR (CDCl3400 MHz): δ 7.19-7.23 (m, 1H), 7.05 (dd, J=8.99, 5.93 Hz, 1H), 4.66 (d, J=5.62 Hz, 2H), 1.76 (t, J=5.93 Hz, 1H). 4-(((6-bromopyridin-2-yl)oxy)methyl)-2,5-difluorobenzonitrile (I-1298): Potassium tert butoxide (1 M in THF, 50.4 mL, 1.5 equivalent) was slowly added to a mixture of (4-chloro-2,5-difluorophenyl)methanol (I-1298-1, 6.0 g, 1.00 equivalent) in THF (60 mL) pre-cooled to 0° C. equivalent. The mixture was stirred at 20° C. for 1 hr., then it was cooled back to 0° C. 2,6-dibromopyridine (7.16 g, 0.90 equivalent) as a solution in THE (20 mL) was then added and the mixture was again stirred at 20° C. for 1 hr., with monitoring by TLC. The mixture was poured into saturated aqueous ammonium chloride and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. Titration with petroleum ether yielded the title compound.1H NMR (CDCl3400 MHz): δ 7.48 (t, J=7.82 Hz, 1H), 7.35 (dd, J=8.93, 6.24 Hz, 1H), 7.19 (dd, J=8.80, 5.87 Hz, 1H), 7.13 (d, J=7.46 Hz, 1H), 6.78 (d, J=8.19 Hz, 1H), 5.40 (s, 2H). ES/MS m/z: 324.0 (M+H+). Preparation of Intermediate I-1300 4-(bromomethyl)-2,3-difluorobenzonitrile (I-1300-1): Carbon tetrabromide (165 mg, 1.05 equivalent) was added to a mixture of 2,3-difluoro-4-(hydroxymethyl)benzonitrile (80 mg, 1.0 equivalent), triphenylphosphine (130 mg, 1.05 equivalent) and DCM (4.7 mL) precooled to 0° C. equivalent. The mixture was allowed to warm to 20° C. with stirring for 15 min, then the mixture was purified directly by silica gel flash column chromatography (EtOAc/hexane) to yield the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.40 (ddd, J=7.5, 5.4, 1.9 Hz, 1H), 7.30 (td, J=6.3, 3.1 Hz, 1H), 4.49 (d, J=1.4 Hz, 2H). 4-[(6-bromo-2-pyridyl)oxymethyl]-2,3-difluorobenzonitrile (I-1300): A mixture of 4-(bromomethyl)-2,3-difluorobenzonitrile (I-1300-1, 18 mg, 1.0 equivalent), 6-bromo-2-pyridinol (27 mg, 2.00 equivalent), cesium carbonate (51 mg, 2.0 equivalent) and acetonitrile (0.8 mL) was stirred at 90° C. for 3 hr. The mixture was then diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.48 (t, J=7.9 Hz, 1H), 7.46-7.36 (m, 2H), 7.13 (d, J=7.5 Hz, 1H), 6.77 (d, J=8.1 Hz, 1H), 5.50 (d, J=1.3 Hz, 2H). ES/MS m/z: 325.0 (M+H+). Preparation of Intermediate I-1301 4-[(6-bromo-2-pyridyl)oxymethyl]-3,5-difluorobenzonitrile (I-1301): A mixture of 4-(bromomethyl)-3,5-difluorobenzonitrile (90 mg, 1.0 equivalent), 6-bromo-2-pyridinol (74 mg, 1.1 equivalent), cesium carbonate (253 mg, 2.0 equivalent) and acetonitrile (3.9 mL) was stirred at 90° C. for 30 min. The mixture was then diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. ES/MS m/z: 325.0 (M+H+). Preparation of Intermediate I-1302 6-bromo-3-fluoro-2-[[2-fluoro-4-(trifluoromethyl)phenyl]methoxy]pyridine (I-1301): A mixture of [2-fluoro-4-(trifluoromethyl)phenyl]methanol (923 mg, 1.0 equivalent), 6-bromo-2-chloro-3-fluoropyridine (1000 mg, 1.0 equivalent), cesium carbonate (3100 mg, 2.0 equivalent) and acetonitrile (9.5 mL) was stirred at 60° C. for 16 hr. The mixture was then diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound as the earlier eluting of two isomers. 1H NMR (400 MHz, Chloroform-d) δ 7.72 (t, J=7.5 Hz, 1H), 7.52-7.42 (m, 1H), 7.39 (dd, J=9.7, 1.7 Hz, 1H), 7.32-7.21 (m, 2H), 7.09 (dd, J=8.1, 2.7 Hz, 1H), 5.57 (s, 2H). Preparation of Intermediate I-1303 Methyl 4-(difluoromethyl)-2-fluorobenzoate (I-1303-1): Diethylaminosulfur trifluoride (72.8 g, 1.20 equivalent) was added to a mixture of methyl 2-fluoro-4-formylbenzoate (50.0 g, 1.00 equivalent) in dichloromethane (500 mL) pre-cooled to 0° C. equivalent. The mixture was allowed to warm to 25° C. with stirring for 3 hr., with monitoring by silica gel TLC. The mixture was slowly poured into a mixture of ice and water, and the pH adjusted to pH 7-8 with aqueous sodium bicarbonate. The mixture was extracted with EtOAc. The organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. Purification by silica gel flash column chromatography (EtOAc/petroleum ether) yielded the title compound.1H NMR (DMSO-d6400 MHz): δ 7.97-8.10 (m, 1H) 7.51-7.65 (m, 2H) 6.95-7.32 (m, 1H) 3.82-3.95 (m, 3H). (4-(difluoromethyl)-2-fluorophenyl)methanol (I-1303-2): LiBH4(14.4 g, 3.0 equivalent) was added to a mixture of methyl 4-(difluoromethyl)-2-fluorobenzoate (I-1303-1, 45.0 g, 1.00 equivalent) in THF (450 mL) pre-cooled to 0° C. The mixture was stirred at 50° C. for 3 hr., with monitoring by silica gel TLC. The mixture was diluted with water, acidified to pH 7 with aqueous HCl, and stirred at 20° C. for 1 hr. The mixture was filtered and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to yield the title compound.1H NMR (CDCl3400 MHz): δ 7.29-7.52 (m, 1H) 7.07-7.24 (m, 2H) 6.36-6.72 (m, 1H) 4.64-5.03 (m, 2H). 4-chloro-2-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyrimidine (I-1303): NaH (60% in mineral oil, 4.54 g, 2.00 equivalent) was added to a mixture of (4-(difluoromethyl)-2-fluorophenyl)methanol (I-1303-2, 10.0 g, 1.00 equivalent) in THE (80 mL)equivalent. The mixture was cooled to −25° C., then 4-chloro-2-(methylsulfonyl)pyrimidine (14.2 g, 1.30 equivalent) was added as a mixture in THE (100 mL). The mixture was stirred at 25° C. for 6 hr, with monitoring by silica gel TLC. The mixture was diluted with water and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. Purification by silica gel flash column chromatography (EtOAc/petroleum ether) yielded the title compound.1H NMR (CDCl3400 MHz): δ 8.34-8.49 (m, 1H) 7.68 (t, J=7.60 Hz, 1H) 7.23-7.37 (m, 3H) 7.00-7.10 (m, 1H) 6.46-6.84 (m, 1H) 5.51-5.63 (m, 2H). ES/MS m/z: 288.0 (M+H+). Preparation of Intermediate I-1304 2-bromo-6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridine (I-1304): triphenylphosphine (35.7 g, 1.2 equivalent), then diisopropyl azodicarboxylate (27.6 g, 1.2 equivalent) were added to a mixture of (4-(difluoromethyl)-2-fluorophenyl)methanol (I-1303-2, 20.0 g, 1.00 equivalent) in THF (200 mL)equivalent. The mixture was stirred at 25° C. for 1 hr., or with monitoring by TLC. The mixture was partitioned between water and ethyl acetate. The organic phase was washed with brine, then concentrated in vacuo. Silica gel flash column chromatography (EtOAc/petroleum ether) yielded the title compound.1H NMR (CDCl3400 MHz): δ=7.60 (t, J=7.6 Hz, 1H), 7.43 (dd, J=7.5, 8.1 Hz, 1H), 7.31-7.25 (m, 2H), 7.23 (s, 1H), 7.08 (dd, J=0.7, 7.5 Hz, 1H), 6.77-6.46 (m, 2H), 5.44 (s, 2H). ES/MS m/z: 331.0 (M+H+). Preparation of Intermediate I-1305 Methyl 2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetate (I-1305-1): A mixture of methyl 2-(4-bromo-2,5-difluoro-phenyl)acetate (500 mg, 1.0 equivalent), PdCl2(dppf) (140 mg, 0.10 equivalent), potassium propionate (635 mg, 3.0 equivalent), and bis(pinacolato)diboron (623 mg, 1.3 equivalent) in 1,4-dioxane (9.4 mL) was purged with argon for 1 min. The mixture was sealed and heated to 90° C. for 2 h. After cooling down to room temperature, 2-bromo-6-[[4-(difluoromethyl)-2-fluoro-phenyl]methoxy]pyridine (Intermediate I-1304, 580 mg, 1.84 mmol, 1.1 equivalent), PdCl2(dppf) (66 mg, 0.05 equivalent), 2 M aqueous Na2CO3 (1.89 mL, 2.0 equivalent) were added, respectively. The resulting mixture was heated to 100° C. under argon and stirred for 2 hr., with monitoring by LCMS. The mixture was cooled to rt and partitioned between water and ethyl acetate. The organic phase was washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. Next, purification by silica gel flash column chromatography (EtOAc/hexane gradient) yielded the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.77 (dd, J=10.5, 6.4 Hz, 1H), 7.65 (dt, J=22.8, 7.7 Hz, 2H), 7.51 (dd, J=7.7, 1.6 Hz, 1H), 7.30 (d, J=8.1 Hz, 2H), 7.09 (dd, J=11.2, 6.0 Hz, 1H), 6.81 (d, J=8.2 Hz, 1H), 6.63 (s, 1H), 5.56 (s, 2H), 3.75 (s, 3H), 3.70 (s, 2H). ES/MS m/z: 438.0 (M+H)+. 2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid (I-1305): A mixture of methyl 2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetate (I-1305-1, 535 mg, 1.0 equivalent), lithium hydroxide (2.0 M in H2O, 1.22 mL, 2.0 equivalent) and acetonitrile (4.3 mL) was stirred at 50-60° C. for 4 hr., with monitoring by LCMS. The mixture was quenched with aqueous HCl until pH<2, then extracted with ethyl acetate. The organic phase was washed with brine, dried over MgSO4, filtered and concentrated in vacuo to yield the title compound. 1H NMR (400 MHz, Methanol-d4) δ 7.82-7.68 (m, 2H), 7.66 (t, J=7.5 Hz, 1H), 7.57-7.41 (m, 1H), 7.41-7.26 (m, 2H), 7.20 (dd, J=11.5, 6.0 Hz, 1H), 6.96-6.56 (m, 2H), 5.58 (s, 2H), 3.84-3.61 (m, 2H). ES/MS m/z: 424.0 (M+H)+. Preparation of Intermediate I-1306 Racemic methyl 3-[(2-methoxy-1,2-dimethyl-propyl)amino]-4-nitrobenzoate (I-1306-1): Methyl 3-fluoro-4-nitrobenzoate (310 mg, 1.0 equivalent), THF (2.0 mL), DMF (1.0 mL), and diisopropylethylamine (1.36 mL, 5.0 equivalent) were added to racemic 3-methoxy-3-methylbutan-2-amine hydrochloride (251 mg, 1.05 equivalent)equivalent. The mixture was stirred at 80° C. for 20 hr., with monitoring by LCMS. The mixture was diluted with ethyl acetate and washed with saturated aqueous NH4Cl, then brine, then dried over magnesium sulfate, filtered and concentrated in vacuo to yield the title compound. ES/MS m/z: 296.9 (M+H)+. Racemic methyl 4-amino-3-((3-methoxy-3-methylbutan-2-yl)amino)benzoate (I-1306): A mixture of crude racemic methyl 3-[(2-methoxy-1,2-dimethyl-propyl)amino]-4-nitrobenzoate (I-1306-1, 461 mg, 1.0 equivalent), 10% palladium on carbon (83 mg, 0.10 equivalent) and ethanol (16 mL) was stirred under an atmosphere of hydrogen for 16 hr., with monitoring by LCMS. The mixture was filtered, concentrated in vacuo and purified by silica gel flash column chromatography (hexane/EtOAc) to yield the title compound. 1H NMR (400 MHz, Methanol-d4) δ 7.34-7.26 (m, 2H), 6.68 (d, J=8.0 Hz, 1H), 3.82 (s, 3H), 3.47 (q, J=6.5 Hz, 1H), 3.25 (s, 3H), 1.27 (s, 6H), 1.16 (d, J=6.5 Hz, 3H). ES/MS m/z: 266.9 (M+H)+. Preparation of Intermediate I-1307 Racemic methyl 4-[[2-(4-bromo-2,5-difluoro-phenyl)acetyl]amino]-3-[(2-methoxy-1,2-dimethyl-propyl)amino]benzoate (I-1307-1): A mixture of racemic methyl 4-amino-3-((3-methoxy-3-methylbutan-2-yl)amino)benzoate (I-1306, 440 mg, 1.00 equivalent), 2-(4-bromo-2,5-difluorophenyl)acetic acid (435 mg, 1.05 equivalent), chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (487 mg, 1.05 equivalent), 1-methylimidazole (678 mg, 5.0 equivalent) and acetonitrile (8.1 mL) was stirred at 20° C. for 1 hr., with monitoring by LCMS. The mixture was then diluted with EtOAc, washed with saturated aqueous NH4Cl, then saturated aqueous NaHCO3, then brine. The organic phase was dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound, which was carried forward without further purification. ES/MS m/z: 499.0 (M+H+). Racemic methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(3-methoxy-3-methylbutan-2-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1307): A mixture of racemic methyl 4-[[2-(4-bromo-2,5-difluoro-phenyl)acetyl]amino]-3-[(2-methoxy-1,2-dimethyl-propyl)amino]benzoate (I-1307-1, 825 mg, 1.0 equivalent), glacial acetic acid (3.3 mL) and 1,2-dichloroethane (8.3 mL) was stirred at 80-95° C. for 96 hr., or until completion as determined by LCMS. The mixture was diluted with EtOAc, quenched with saturated aqueous NaHCO3and batchwise addition of solid NaHCO3. The organic phase was washed with brine, dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound. 1H NMR (400 MHz, Chloroform-d) δ 8.56-8.50 (m, 1H), 7.91 (dd, J=8.5, 1.5 Hz, 1H), 7.69 (d, J=8.5 Hz, 1H), 7.29 (dd, J=8.6, 5.6 Hz, 1H), 7.03 (dd, J=8.6, 6.3 Hz, 1H), 4.39-4.22 (m, 3H), 3.90 (s, 3H), 3.15 (s, 3H), 1.56 (d, J=7.1 Hz, 3H), 1.26 (s, 3H), 0.98 (s, 3H). ES/MS m/z: 481.2 (M+H+). Preparation of Intermediate I-1308 1-((3-bromophenoxy)methyl)-4-chloro-2-fluorobenzene (I-1308): A mixture of 1-(bromomethyl)-4-chloro-2-fluorobenzene (581 mg, 1.0 equivalent), 3-bromophenol (450 mg, 1.0 equivalent), cesium carbonate (2.54 g, 3.0 equivalent) and acetone (13 mL) was stirred at 70° C. for 2 hr. The mixture was then diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.42 (t, J=8.0 Hz, 1H), 7.21-7.07 (m, 5H), 6.89 (ddd, J=7.8, 2.5, 1.4 Hz, 1H), 5.07 (s, 2H). Preparation of Intermediate I-1309 1-[(5-bromo-2-fluoro-phenoxy)methyl]-4-chloro-2-fluorobenzene (I-1309): A mixture of 1-(bromomethyl)-4-chloro-2-fluorobenzene (585 mg, 1.0 equivalent), 5-bromo-2-fluorophenol (500 mg, 1.0 equivalent), cesium carbonate (2.56 g, 3.0 equivalent) and acetone (13 mL) was stirred at 70° C. for 2 hr. The mixture was then diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.46 (t, J=8.0 Hz, 1H), 7.23-7.12 (m, 3H), 7.06 (dd, J=4.0, 2.3 Hz, 1H), 6.98 (dd, J=10.7, 8.6 Hz, 1H), 5.12 (s, 2H). Preparation of Intermediate I-1310 Methyl 3-[(2-methoxy-2-methyl-propyl)amino]-4-nitrobenzoate (I-1310-1): Methyl 3-fluoro-4-nitrobenzoate (500 mg, 1.0 equivalent), 2-methyltetrahydrofuran (3.4 mL), DMF (1.7 mL), and diisopropylethylamine (2.2 mL, 5.0 equivalent) were added To 2-methoxy-2-methyl-propan-1-amine (285 mg, 1.1 equivalent)equivalent. The mixture was stirred at 80° C. for 16 hr., with monitoring by LCMS. The mixture was diluted with ethyl acetate and washed with saturated aqueous NH4Cl, then brine, then dried over magnesium sulfate, filtered and concentrated in vacuo to yield the title compound. ES/MS m/z: 282.8 (M+H)+. Methyl 4-amino-3-[(2-methoxy-2-methylpropyl)amino]benzoate (I-1310): A mixture of crude methyl 3-[(2-methoxy-2-methyl-propyl)amino]-4-nitrobenzoate (I-13010-1, 555 mg, 1.0 equivalent), 10% palladium on carbon (105 mg, 0.10 equivalent) and ethanol (20 mL) was stirred under an atmosphere of hydrogen for 16 hr., with monitoring by LCMS. The mixture was filtered, concentrated in vacuo and purified by silica gel flash column chromatography (hexane/EtOAc) to yield the title compound. 1H NMR (400 MHz, Methanol-d4) δ 7.32 (d, J=8.0 Hz, 1H), 7.24 (d, J=1.8 Hz, 1H), 6.68 (d, J=8.1 Hz, 1H), 3.82 (s, 3H), 3.25 (s, 3H), 3.11 (s, 2H), 1.31 (s, 6H). ES/MS m/z: 252.9 (M+H)+. Preparation of Intermediate I-1311 Methyl 4-[[2-(4-bromo-2,5-difluorophenyl)acetyl]amino]-3-[(2-methoxy-2-methylpropyl)amino]benzoate (I-1311-1): A mixture of methyl 4-amino-3-[(2-methoxy-2-methylpropyl)amino]benzoate (I-1310, 655 mg, 1.00 equivalent), 2-(4-bromo-2,5-difluorophenyl)acetic acid (684 mg, 1.05 equivalent), chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (765 mg, 1.05 equivalent), 1-methylimidazole (1066 mg, 5.0 equivalent) and acetonitrile (13 mL) was stirred at 20° C. for 2.5 hr., with monitoring by LCMS. The mixture was then diluted with EtOAc, washed with saturated aqueous NH4Cl, then saturated aqueous NaHCO3, then brine. The organic phase was dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound, which was carried forward without further purification. ES/MS m/z: 485.0 (M+H+). Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(2-methoxy-2-methylpropyl)-1H-benzo[d]imidazole-6-carboxylate (I-1311): A mixture of methyl 4-[[2-(4-bromo-2,5-difluorophenyl)acetyl]amino]-3-[(2-methoxy-2-methylpropyl)amino]benzoate (I-1311-1, 1260 mg, 1.0 equivalent), glacial acetic acid (5.3 mL) and 1,2-dichloroethane (13 mL) was stirred at 80° C. for 16 hr., with monitoring by LCMS. The mixture was diluted with EtOAc, quenched with saturated aqueous NaHCO3and batchwise addition of solid NaHCO3. The organic phase was washed with brine, dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound. 1H NMR (400 MHz, Chloroform-d) δ 8.16 (s, 1H), 8.05 (d, J=8.6 Hz, 1H), 7.86 (d, J=8.4 Hz, 1H), 7.35 (s, 1H), 7.30 (dd, J=8.7, 5.5 Hz, 1H), 4.62 (s, 2H), 4.20 (s, 2H), 3.96 (s, 3H), 3.14 (s, 3H), 1.23 (s, 6H). ES/MS m/z: 467.2 (M+H+). Preparation of Intermediate I-1312 2-bromo-6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridine (I-1312): A mixture of [2-fluoro-4-(trifluoromethyl)phenyl]methanol (1213 mg, 1.1 equivalent), 2-bromo-6-fluoropyridine (1000 mg, 1.0 equivalent), cesium carbonate (3.71 g, 2.0 equivalent) and acetonitrile (8.0 mL) was stirred at 25° C. for 16 hr. with monitoring by LCMS. The mixture was then filtered and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. ES/MS m/z: 351.8 (M+H+). Preparation of Intermediate I-1313 4-chloro-5-fluoro-2-[[2-fluoro-4-(trifluoromethyl)phenyl]methoxy]pyrimidine (I-1313): Sodium hydride (46 mg, 60% in mineral oil, 1.20 equivalent), then [2-fluoro-4-(trifluoromethyl)phenyl]methanol (326 mg, 1.00 equivalent) were added to mixture of 2,4-dichloro-5-fluoro-pyrimidine (280 mg, 1.0 equivalent) and THE (3.2 mL)equivalent. The mixture was stirred at 20° C. for 16 hr. The mixture was partitioned between ethyl acetate and brine. The organic phase was dried over MgSO4, filtered and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. 1H NMR (400 MHz, Chloroform-d) δ 8.25 (d, J=2.2 Hz, 1H), 7.67 (t, J=7.5 Hz, 1H), 7.48 (dd, J=7.8, 1.6 Hz, 1H), 7.40 (dd, J=9.6, 1.7 Hz, 1H), 5.61 (s, 2H). ES/MS m/z: 324.9 (M+H+). Preparation of Intermediate I-1319 3-(bromomethyl)-6-chloro-2-fluoro-pyridine (I-1319): Carbon tetrabromide (431 mg, 1.05 equivalent) was added to a mixture of (6-chloro-2-fluoro-3-pyridyl)methanol (200 mg, 1.0 equivalent), triphenylphosphine (357 mg, 1.10 equivalent) and DCM (5.0 mL) precooled to 0° C. equivalent The mixture stirred at 0° C. for 1 hr., then the mixture was concentrated in vacuo. Purification by silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. Preparation of Intermediate I-1322 4-(((6-bromo-5-fluoropyridin-2-yl)oxy)methyl)-3-fluorobenzonitrile (I-1322): A mixture of 3-Fluoro-4-(hydroxymethyl)benzonitrile (160 mg, 1.06 mmol), cesium carbonate (332 mg, 1.02 mmol), and 2-bromo-3,6-difluoro-pyridine (98.0 mg, 0.505 mmol) in MeCN (2 mL) was stirred for 4 days at 20 C. The mixture was diluted with ethyl acetate, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. 1H NMR (400 MHz, CDCl3) δ 7.67 (t, J=7.5 Hz, 1H), 7.50 (dd, J=8.0, 1.5 Hz, 1H), 7.42 (ddd, J=8.7, 4.0, 2.4 Hz, 2H), 6.78 (dd, J=8.8, 2.8 Hz, 1H), 5.47 (s, 2H). ES/MS m/z: 327.1 (M+H+). Preparation of Intermediate I-1324 Tert-butyl 2-(4-(6-(benzyloxy)pyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1324-1): A mixture of tert-butyl 2-(4-bromo-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (49 mg, 0.965 equivalent), PdCl2(dppf) (11.4 mg, 0.15 equivalent), potassium propionate (34 mg, 3.0 equivalent), and bis(pinacolato)diboron (34 mg, 1.3 equivalent) in 1,4-dioxane (2.0 mL) was purged with argon for 1 min. The mixture was sealed and heated to 100° C. for 2 h. After cooling down to room temperature, 2-(benzyloxy)-6-bromopyridine (27 mg, 1.1 equivalent), PdCl2(dppf) (5.7 mg, 0.075 equivalent), 2 M aqueous Na2CO3(0.10 mL, 2.0 equivalent) were added, respectively. The resulting mixture was heated to 90° C. under argon and stirred for 2 hr., with monitoring LCMS. The mixture was filtered and concentrated in vacuo. Purification by silica gel flash column chromatography (EtOAc/hexane gradient) yielded the title compound. Tert-butyl 2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1324): A mixture of tert-butyl 2-(4-(6-(benzyloxy)pyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1324-1, 40 mg, 1.0 equivalent), 10% palladium on carbon (10 mg, 0.14 equivalent), and ethanol (3.0 mL) was stirred under an atmosphere of hydrogen for 3 hr. The mixture was filtered and concentrated in vacuo to yield the title compound. ES/MS m/z: 514.7 (M+H+). Preparation of Intermediate I-1325 Tert-butyl 2-fluoro-3-((2-methoxyethyl)amino)-4-nitrobenzoate (I-1325-1): Tert-butyl 2,3-difluoro-4-nitrobenzoate (600 mg, 1.0 equivalent), THE (10 mL), and diisopropylethylamine (2.0 mL, 5.0 equivalent) were added to 2-methoxyethanamine (191 mg, 1.1 equivalent)equivalent. The mixture was stirred at 60° C. for 16 hr., with monitoring by LCMS. The mixture was diluted with ethyl acetate and washed with saturated aqueous NH4Cl, then brine, then dried over magnesium sulfate, filtered and concentrated in vacuo to yield the title compound, which was carried forward without further purification. Tert-butyl 4-amino-2-fluoro-3-((2-methoxyethyl)amino)benzoate (I-1325-2): A mixture of crude tert-butyl 2-fluoro-3-((2-methoxyethyl)amino)-4-nitrobenzoate (I-1325-1, 560 mg, 1.0 equivalent), 10% palladium on carbon (190 mg, 0.10 equivalent) and ethanol (15 mL) was stirred under an atmosphere of hydrogen for 4 hr., with monitoring by LCMS. The mixture was filtered, concentrated in vacuo and purified by silica gel flash column chromatography (hexane/EtOAc) to yield the title compound. Tert-butyl 4-(2-(4-bromo-2,5-difluorophenyl)acetamido)-2-fluoro-3-((2-methoxyethyl)amino)benzoate (I-1325-3): A mixture of tert-butyl 4-amino-2-fluoro-3-((2-methoxyethyl)amino)benzoate (I-1325-2, 250 mg, 1.00 equivalent), 2-(4-bromo-2,5-difluorophenyl)acetic acid (318 mg, 1.44 equivalent), chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (300 mg, 1.22 equivalent), 1-methylimidazole (360 mg, 5.0 equivalent) and acetonitrile (8 mL) was stirred at 20° C. for 2 hr., with monitoring by LCMS. The mixture was then diluted with EtOAc, then washed with 1 N HCl. The organic phase was dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound, which was carried forward without further purification. Tert-butyl 2-(4-bromo-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1325): A mixture of tert-butyl 4-(2-(4-bromo-2,5-difluorophenyl)acetamido)-2-fluoro-3-((2-methoxyethyl)amino)benzoate (I-1325-3, 331 mg, 1.0 equivalent), glacial acetic acid (0.38 mL) and 1,2-dichloroethane (5 mL) was stirred at 60° C. for 4 hr., with monitoring by LCMS. The mixture was diluted with EtOAc, quenched with saturated aqueous NaHCO3. The organic phase was washed with brine, dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound. 1H NMR (400 MHz, Chloroform-d) δ 7.86 (dd, J=8.6, 6.7 Hz, 1H), 7.65 (d, J=8.6 Hz, 1H), 7.35 (dd, J=8.7, 5.6 Hz, 1H), 7.13 (dd, J=8.3, 6.4 Hz, 1H), 4.62-4.40 (m, 4H), 3.78 (t, J=5.1 Hz, 2H), 3.29 (s, 3H), 1.64 (s, 9H). Preparation of Intermediate I-1326 Methyl (S)-4-(2-(4-bromo-3-fluorophenyl)acetamido)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (I-1326-1): A mixture of methyl (S)-4-amino-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (I-80, 200 mg, 1.0 equivalent), 2-(4-bromo-3-fluorophenyl)acetic acid (83 mg, 1.1 equivalent), chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (265 mg, 1.25 equivalent), 1-methylimidazole (311 mg, 5.0 equivalent) and acetonitrile (3.0 mL) was stirred at 20° C. for 2 hr., with monitoring by LCMS. The mixture was then diluted with EtOAc, then washed with 1 N HCl. The organic phase was dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound, which was carried forward without further purification. Methyl (S)-2-(4-bromo-3-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1326): A mixture of methyl (S)-4-(2-(4-bromo-3-fluorophenyl)acetamido)-3-((4,4-dimethyltetrahydrofuran-3-yl)amino)benzoate (I-1326-1, 250 mg, 1.0 equivalent) and glacial acetic acid (0.2 mL) was stirred at 180° C. for 2 hr., with monitoring by LCMS. The mixture was diluted with EtOAc, quenched with saturated aqueous NaHCO3and batchwise addition of solid NaHCO3. The organic phase was washed with brine, dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound. ES/MS m/z: 462.9 (M+H+). Preparation of Intermediate I-1327 Methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (330-1): A mixture of methyl (S)-2-(4-bromo-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1229, 800 mg, 1.67 mmol, 1.0 equivalent), PdCl2(dppf) (186 mg, 0.25 mmol, 0.15 equivalent), potassium propionate (560 mg, 5.01 mmol, 3.0 equivalent), and bis(pinacolato)diboron (510 mg, 2.00 mol, 1.2 equivalent) was mixed with 1,4-dioxane (10 mL) and the resulting mixture was purged with argon for 2 min. The mixture was sealed and heated to 120° C. by microwave. The mixture was then stirred for 1 h. After cooling down to room temperature, 4-[(6-bromo-3-fluoro-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (Intermediate I-109a, 580 mg, 1.84 mmol, 1.1 equivalent), PdCl2(dppf) (62 mg, 0.0834 mmol, 0.05 equivalent), 2 M aqueous Na2CO3(2.0 mL, 4.17 mmol, 2.5 equivalent) were added, respectively. The resulting mixture was heated to 100° C. under argon and stirred for 3 hrs. before cooling to rt. Once cooled the mixture was filtered through a plug of Celite and MgSO4. The filtrate was concentrated and purified by column chromatography (silica gel, EtOAc/hexane gradient) to yield the title compound. ES/MS m/z: 627.7 (M+H+). Methyl (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2-fluoro-4-(5-fluoro-6-hydroxypyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylate (I-1327): A mixture of methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate 330-1, 200 mg, 1.0 equivalent) in 4M HCl solution in dioxane (8.0 mL, 10 equivalent) was heated at 80° C. overnight. The mixture was diluted with EtOAc and washed with brine, saturated aqueous NaHCO3and water. The organic phase was dried, concentrated in vacuo, and purified by preparative reverse-phase HPLC (CH3CN/water, 0.1% TFA) to yield the title compound. ES/MS m/z: 494.7 (M+H+). Preparation of Intermediate I-1328 Methyl (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2-fluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1328): Methyl (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2-fluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner similar to Intermediate I-1327. ES/MS m/z: 476.6 (M+H+). Preparation of Intermediate I-1329 2-(2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)acetic acid (I-1329-1). Pd(dppf)Cl2·DCM (7.75 g, 9.5 mmol, 0.05 eq) was added to a solution of 2-(4-bromo-2-fluorophenyl)acetic acid (45.0 g, 0.19 mol, 1.0 eq), 4,4,4′,4′, 5,5,5′, 5′-octamethyl-2, 2′-bi(1, 3,2-dioxaborolane) (58.8 g, 0.23 mol, 1.2 eq) and potassium acetate (74.6 g, 0.76 mol, 4.0 eq) in DMF (450 mL). The mixture was flushed thoroughly with nitrogen for 5-10 mins. The mixture was then purged with nitrogen for three times. The mixture was heated at 90° C. for overnight. HPLC and LCMS showed completion. After cooling to room temperature, the mixture was then filtered through a pad of Celite and washed with EtOAc (500 mL). The filtrate was concentrated. Next, water (1200 mL) was added and the pH was adjusted to ˜ 3 with 3 N HCl. The resulting mixture was extracted with EtOAc (600 mL×3). The combined organic layers were washed with brine, dried over MgSO4, concentrated under reduced pressure. The residue was purified by silica gel flash column chromatography, eluting with 0-20% EtOAc in hexanes to give the title compound.1HNMR (400 MHz, DMSO-d6) δ 12.50 (br s, 1H), 7.45-7.35 (m, 1H), 7.35-7.31 (m, 2H), 3.65 (s, 2H), 1.23 (s, 12H). 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorophenyl)acetic acid (I-1329): 2-(2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)acetic acid (I-1329-1, 13.7 g, 48.9 mmol, 1.0 eq) and 4-(((6-bromopyridin-2-yl)oxy)methyl)-3-fluorobenzonitrile (I-3, 15.0 g, 48.9 mmol, 1.0 eq) were taken up in 1,4-dioxane (240 mL) and aqueous sodium carbonate (2.0 M, 67.5 mL, 135.0 mmol, 2.8 eq) and the mixture sparged with nitrogen for 5 minutes. Pd(dppf)Cl2·DCM (566 mg, 0.49 mmol) was then added and the system was purged with nitrogen for three times. The mixture was heated to 90° C. and stirred for 3 h. HPLC and LCMS showed completion. Upon completion the mixture was cooled to room temperature. After cooling the mixture was filtered and washed with EtOAc (100 mL×3). The filtrate was concentrated. Next, water (500 mL) was added, filtered and the filter cake was triturated with EtOAc (100 mL×3) for three times. The solid obtained by filtration and then water (800 mL) was added, the pH was adjusted to ˜ 6 with 1 N HCl. The resulting mixture was extracted with EtOAc (600 mL×3). The combined organic layers were washed with brine, dried over MgSO4, concentrated under reduced pressure to give the title compound.1HNMR (400 MHz, DMSO-d6) δ 12.53 (br s, 1H), 7.94-7.91 (m, 1H), 7.87-7.81 (m, 3H), 7.78-7.71 (m, 2H), 7.64 (d, J=7.6 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 6.92 (d, J=8.0 Hz, 1H), 5.62 (s, 2H), 3.69 (s, 2H). ES/MS m/z: 381.1 (M+H)+. Preparation of Intermediate I-1330 Tert-butyl 2-(4-bromo-2-fluorobenzyl)-1-((1-(fluoromethyl)cyclopropyl)methyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1330): Tert-butyl 2-(4-bromo-2-fluorobenzyl)-1-((1-(fluoromethyl)cyclopropyl)methyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner described for Intermediate I-2 using Intermediate I-1196 and 2-(4-bromo-2-fluorophenyl)acetic acid. ES/MS m/z: 509.7 (M+H)+. Preparation of Intermediate I-1332 Methyl (S)-2-(2,5-difluoro-4-(6-((4-iodobenzyl)oxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1332): A mixture of methyl (S)-2-(2,5-difluoro-4-(6-hydroxypyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-9,157 mg, 1.0 equivalent), 1-(bromomethyl)-4-iodobenzene (100 mg, 1.0 equivalent), cesium carbonate (150 mg, 1.4 equivalent) and acetonitrile (3.0 mL) was stirred at 50° C. for 1.5 hr, with monitoring by LCMS. The mixture was then filtered and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. ES/MS m/z: 683.2 (M+H+). Preparation of Intermediate I-1333 Methyl (S)-2-(4-(6-((5-bromopyrimidin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1333): Methyl (S)-2-(4-(6-((5-bromopyrimidin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner described for preparation of Intermediate I-1332, using Intermediate I-9. ES/MS m/z: 638 (M+H+). Preparation of Intermediate I-1335 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-N-((1-methyl-1H-pyrazol-3-yl)methyl)picolinamide (I-1335): A mixture of Intermediate I-1273-3 (from Intermediate I-1273, 50 mg, 1.0 equivalent), (1-methylpyrazol-3-yl)methanamine (24 mg, 1.5 equivalent), HATU (83 mg, 1.5 equivalent), diisopropylethylamine (60 μL, 2.5 equivalent) in DMF (2 mL) was stirred at room temperature for 2 hr. The mixture was diluted with EtOAc, washed with 10% LiCl. Next, a mixture of saturated aqueous NaHCO3and brine, then brine, dried over MgSO4, filtered, and concentrated in vacuo. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. ES/MS m/z: 435.9 (M+H+). Preparation of Intermediate I-1336 Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (I-1336): Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner described for Intermediate I-108 substituting with 4,4-dimethyltetrahydrofuran-3-amine hydrochloride for (S)-4,4-dimethyltetrahydrofuran-3-amine hydrochloride. ES/MS: 338.5 (M+Na+). Preparation of Intermediate I-1337 Racemic methyl 2-(4-bromo-2,5-difluorobenzyl)-4-fluoro-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1337): Racemic methyl 2-(4-bromo-2,5-difluorobenzyl)-4-fluoro-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner described for preparation of Intermediate I-1326, using Intermediate I-1133. ES/MS: 499.0 (M+H+). Preparation of Intermediate I-1338 4-[(2-chloropyrimidin-4-yl)oxymethyl]-3-fluoro-benzonitrile (I-1338): Potassium tert-butoxide (0.237 g, 2.11 mmol) was added to a mixture of 3-fluoro-4-(hydroxymethyl)benzonitrile (0.609 g, 4.03 mmol) in tetrahydrofuran (1.00 mL) and the resulting mixture was stirred for 5 min at room temperature. This solution was then added to a mixture of 2,4-dichloropyrimidine (300 mg, 2.01 mmol) in N,N-dimethylformamide (1.50 mL) pre-cooled to −78° C. and the mixture was warmed slowly to room temperature and stirred for 1 h, or until LCMS showed complete conversion to desired product. The mixture was poured into 50 mL of water and stirred for 5 min, then the precipitate was filtered off to give the title compound. 1H NMR (400 MHz, Chloroform-d) δ 8.39 (d, J=5.7 Hz, 1H), 7.66 (t, J=7.4 Hz, 1H), 7.53 (dd, J=8.0, 1.5 Hz, 1H), 7.44 (dd, J=9.2, 1.5 Hz, 1H), 6.78 (d, J=5.7 Hz, 1H), 5.57 (s, 2H). ES/MS m/z: 264.1 (M+H+). Preparation of Intermediate I-1339 Ethyl 2-(4-bromo-2-chloro-5-methylbenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1339-1): A mixture of ethyl 4-amino-3-fluoro-5-(2-methoxyethylamino)benzoate (I-1032, 73 mg, 1.0 equivalent), 2-(4-bromo-2-chloro-5-methyl-phenyl)acetic acid (I-1023, 90 mg, 1.2 equivalent), chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (100 mg, 1.25 equivalent), 1-methylimidazole (70 mg, 3.0 equivalent) and DMF (5.0 mL) was stirred at 20° C. for 5 hr., with monitoring by LCMS. The mixture was then diluted with EtOAc, then washed with water, then brine. The organic phase was dried over MgSO4, filtered, and concentrated in vacuo to yield the title compound, which was carried forward without further purification. Ethyl 2-[(4-bromo-2-chloro-5-methyl-phenyl)methyl]-7-fluoro-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-1339): A mixture of crude ethyl 2-(4-bromo-2-chloro-5-methylbenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1339-1, 143 mg, 1.0 equivalent) and glacial acetic acid (5.0 mL) was stirred at reflux for 20 hr., with monitoring by LCMS. The mixture was concentrated in vacuo and purified by silica gel flash column chromatography (EtOAc/hexane) to yield the title compound. ES/MS m/z: 483.3 (M+H+). Preparation of Intermediate I-1340 6-bromo-3-fluoro-2-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridine (I-1340): 6-bromo-3-fluoro-2-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridine was prepared in a similar manner to 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-fluoropyridine (Intermediate I-1049) using (2-fluoro-4-(1H-1,2,3-triazol-1-yl)phenyl)methanol (from preparation of Intermediate I-1282) and 6-bromo-3-fluoropyridin-2-ol. ES/MS: 367.1 (M+1). Preparation of Intermediate I-1341 (6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methanol (I-1341): To a mixture of 1-1005 (120 mg, 1.0 equivalent), triphenylphosphine 197 mg, 1.10 equivalent) in DCM (5 mL) pre-cooled to 0° C. was added carbon tetrabromide (260 mg, 1.15 equivalent). The mixture was stirred at 0° C., then allowed to warm to 20° C., concentrated in vacuo, and purified by silica gel flash column chromatography (EtOAc/hexane) to yield the title compound. ES/MS m/z: 177.2 (M+H+). Preparation of Intermediate I-1347 Methyl 2-[[4-[6-[(6-chloro-4-fluoro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1347): Methyl 2-[[4-[6-[(6-chloro-4-fluoro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 1-1219 for 1-9 and 5-(bromomethyl)-2-chloro-4-fluoro-pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 637.3 (M+H+). Preparation of Intermediate I-1348 Methyl 2-[[4-[6-[(4,6-dichloro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1348): Methyl 2-[[4-[6-[(4,6-dichloro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 1-1219 for 1-9 and 2,4-dichloro-5-(chloromethyl)pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 653.3 (M+H+). Preparation of Intermediate I-1349 Methyl 2-[[4-[6-[(4-bromophenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1349): Methyl 2-[[4-[6-[(4-bromophenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 1-1219 for 1-9 and 1-bromo-4-(bromomethyl)benzene for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 662, 664 (M+H+). Preparation of Intermediate I-1350 Methyl 2-[[4-[6-[(6-chloro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (I-1350): Methyl 2-[[4-[6-[(6-chloro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate was prepared in a manner as described for Intermediate I-21 substituting 1-1219 for 1-9 and 5-(bromomethyl)-2-chloro-pyridine for 5-bromo-2-(bromomethyl)thiazole. ES/MS: 619.2 (M+H+). Preparation of Intermediate I-1351 Methyl 3-(((3R,4R)-4-hydroxytetrahydrofuran-3-yl)amino)-4-nitrobenzoate: Methyl 3-(((3R,4R)-4-hydroxytetrahydrofuran-3-yl)amino)-4-nitrobenzoate was prepared in a manner described for Intermediate I-103 substituting (3R,4R)-4-aminotetrahydrofuran-3-ol for (S)-4,4-dimethyltetrahydrofuran-3-amine and methyl 3-fluoro-4-nitrobenzoate for methyl 3,5-difluoro-4-nitrobenzoate. ES/MS m/z: 283.0 (M+H+). Methyl 3-(((3R,4R)-4-methoxytetrahydrofuran-3-yl)amino)-4-nitrobenzoate: A mixture of methyl 3-(((3R,4R)-4-hydroxytetrahydrofuran-3-yl)amino)-4-nitrobenzoate (200 mg, 0.71 mmol), sodium hydride (19.5 mg, 0.85 mmol) and iodomethane (71 uL, 0.85 mmol) in DMF (4.00 mL) was stirred at 0° C. for 15 min. Upon completion the mixture was then warmed to 25° C. and stirred for 16 h. The mixture was neutralized with 1M hydrochloride acid diluted in EtOAc and washed with brine. Next the mixture was dried over sodium sulfate, concentrated, and purified by chromatography (EtOAc/hexane) to give desired product ES/MS m/z: 297.2 (M+H+). Methyl 4-amino-3-(((3R,4R)-4-methoxytetrahydrofuran-3-yl)amino)benzoate: A mixture of methyl 3-(((3R,4R)-4-methoxytetrahydrofuran-3-yl)amino)-4-nitrobenzoate (210 mg, 0.68 mmol), and Palladium on carbon (10% loading, 72.2 mg, 0.71 mmol) in ethanol (5.0 mL) and placed under hydrogen at 25° C. for 1 h. The mixture was diluted in EtOAc and washed with brine. Next, the mixture was dried over sodium sulfate, concentrated and used without further purification. ES/MS m/z: 267.1 (M+H+). Preparation of Intermediate I-1352 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid (I-1352): 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid was prepared in a manner as described for 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid (Intermediate I-7) using 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (I-1049) in place of 4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (I-3). ES/MS: 457.9 (M+H+). Preparation of Intermediate I-1353 and I-1354 1-(5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloropyridin-2-yl)-4,5,6,7-tetrahydro-1H-benzo[d][1,2,3]triazole (I-1353) and 2-(5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloropyridin-2-yl)-4,5,6,7-tetrahydro-2H-benzo[d][1,2,3]triazole (I-1354): 1-(5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloropyridin-2-yl)-4,5,6,7-tetrahydro-1H-benzo[d][1,2,3]triazole (I-1353) and 2-(5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloropyridin-2-yl)-4,5,6,7-tetrahydro-2H-benzo[d][1,2,3]triazole (I-1354) were prepared in a manner as described for 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (Intermediate I-1049) using 4,5,6,7-tetrahydro-1H-benzo[d][1,2,3]triazole in place of 1H-1,2,3-triazole and running the final step at room temperature overnight. 1-(5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloropyridin-2-yl)-4,5,6,7-tetrahydro-1H-benzo[d][1,2,3]triazole (I-1353):1H NMR (400 MHz, Chloroform-d) δ 8.62 (s, 1H), 8.24 (s, 1H), 7.47 (dd, J=8.1, 7.5 Hz, 1H), 7.13 (dd, J=7.5, 0.7 Hz, 1H), 6.78 (dd, J=8.2, 0.7 Hz, 1H), 5.53 (s, 2H), 3.13 (s, 2H), 2.83 (s, 2H), 1.87 (d, J=3.7 Hz, 4H). 2-(5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloropyridin-2-yl)-4,5,6,7-tetrahydro-2H-benzo[d][1,2,3]triazole (I-1354):1H NMR (400 MHz, Chloroform-d) δ 8.67 (s, 1H), 8.06 (s, 1H), 7.45 (t, J=7.8 Hz, 1H), 7.11 (d, J=7.5 Hz, 1H), 6.77 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 2.84 (t, J=3.4 Hz, 4H), 1.91 (p, J=3.3 Hz, 4H). Preparation of Intermediate I-1355 Methyl 4-amino-3-((2-(difluoromethoxy)ethyl)amino)benzoate (I-1355): Methyl 4-amino-3-((2-(difluoromethoxy)ethyl)amino)benzoate (I-1355) was prepared in a similar manner to tert-butyl 4-amino-3-((2-methoxyethyl)amino)benzoate (Intermediate I-6) using 2-(difluoromethoxy)ethan-1-amine in place of 2-methoxyethanamine, and methyl 3-fluoro-4-nitrobenzoate in place of tert-butyl 3-fluoro-4-nitrobenzoate. ES/MS: 261.0 (M+1). Preparation of Intermediate I-1356 Methyl 4-amino-3-((2-isopropoxyethyl)amino)benzoate (I-1356): Methyl 4-amino-3-((2-isopropoxyethyl)amino)benzoate (Intermediate I-1356) was prepared in a similar manner to tert-butyl 4-amino-3-((2-methoxyethyl)amino)benzoate (Intermediate I-6) using 2-isopropoxyethan-1-amine in place of 2-methoxyethanamine, and methyl 3-fluoro-4-nitrobenzoate in place of tert-butyl 3-fluoro-4-nitrobenzoate. ES/MS: 253.0 (M+1). Preparation of Intermediate I-1357 Methyl 4-amino-3-((2-cyclopropoxyethyl)amino)benzoate (I-1357): Methyl 4-amino-3-((2-cyclopropoxyethyl)amino)benzoate was prepared in a similar manner to tert-butyl 4-amino-3-((2-methoxyethyl)amino)benzoate (Intermediate I-6) using 2-cyclopropoxyethan-1-amine in place of 2-methoxyethanamine, and methyl 3-fluoro-4-nitrobenzoate in place of tert-butyl 3-fluoro-4-nitrobenzoate. ES/MS: 251.0 (M+1). Preparation of Intermediate I-1358 Methyl 4-amino-3-((2-isobutoxyethyl)amino)benzoate (I-1358): Methyl 4-amino-3-((2-isobutoxyethyl)amino)benzoate was prepared in a similar manner to tert-butyl 4-amino-3-((2-methoxyethyl)amino)benzoate (Intermediate I-6) using 2-isobutoxyethan-1-amine in place of 2-methoxyethanamine, and methyl 3-fluoro-4-nitrobenzoate in place of tert-butyl 3-fluoro-4-nitrobenzoate. ES/MS: 267.0 (M+1). Preparation of Intermediate I-1359 Tert-butyl 2-(4-bromo-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-1359): tert-butyl 2-(4-bromo-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a similar manner to Intermediate I-11. ES/MS: 481.05 (M+1).1H NMR (400 MHz, DMSO-d6) δ 8.12 (s, 1H), 7.77-7.71 (m, 2H), 7.59-7.55 (m, 1H), 7.44 (dd, J=9.1, 6.3 Hz, 1H), 4.55 (t, J=5.2 Hz, 2H), 4.36 (s, 2H), 3.66 (t, J=5.0 Hz, 2H), 3.20 (s, 3H), 1.57 (s, 9H). Preparation of Intermediate I-1360 1-(5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloropyridin-2-yl)-1H-benzo[d][1,2,3]triazole (I-1360): 1-(5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloropyridin-2-yl)-1H-benzo[d][1,2,3]triazole was prepared in a manner as described for 5-(((6-bromopyridin-2-yl)oxy)methyl)-4-chloro-2-(1H-1,2,3-triazol-1-yl)pyridine (Intermediate I-1049) using 1H-benzo[d][1,2,3]triazole in place of 1H-1,2,3-triazole.1H NMR (400 MHz, Chloroform-d) δ 8.76 (s, 1H), 8.64 (dt, J=8.5, 1.0 Hz, 1H), 8.43 (s, 1H), 8.14 (dt, J=8.3, 1.0 Hz, 1H), 7.63 (ddd, J=8.3, 7.0, 1.1 Hz, 1H), 7.48 (ddd, J=8.5, 7.2, 1.3 Hz, 2H), 7.14 (dd, J=7.6, 0.6 Hz, 1H), 6.80 (dd, J=8.2, 0.6 Hz, 1H), 5.57 (s, 2H). Preparation of Intermediate I-1361 Methyl 4-amino-3-[[(1R,2R)-2-methoxy-1-methyl-propyl]amino]benzoate (I-1361) was prepared in a manner as described for Intermediate I-1, using (2R,3R)-3-methoxybutan-2-amine hydrochloride. ES/MS m/z: 253.0 (M+H+). Preparation of Intermediate I-1362 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(1R,2R)-2-methoxy-1-methyl-propyl]benzimidazole-5-carboxylate (I-1362) was prepared in a manner as described for Intermediate I-2, using 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and Intermediate I-1361. ES/MS m/z: 467.1 (M+H+). Preparation of Intermediate I-1363 Methyl 4-amino-3-[[(1S,2S)-2-methoxy-1-methyl-propyl]amino]benzoate (I-1363) was prepared in a manner as described for Intermediate I-1, using (2S,3S)-3-methoxybutan-2-amine hydrochloride. ES/MS m/z: 253.0 (M+H+). Preparation of Intermediate I-1364 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(1S,2S)-2-methoxy-1-methyl-propyl]benzimidazole-5-carboxylate (I-1364) was prepared in a manner as described for Intermediate I-2 using 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and Intermediate I-1363. ES/MS m/z: 467.1 (M+H+). Preparation of Intermediate I-1365 Methyl 4-amino-3-[(2-methoxy-2-methyl-propyl)amino]benzoate (I-1361): methyl 4-amino-3-[(2-methoxy-2-methyl-propyl)amino]benzoate was prepared in a manner as described for Intermediate I-1, using 2-methoxy-2-methyl-propan-1-amine. ES/MS m/z: 252.9 (M+H+). Preparation of Intermediate I-1366 Methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-(2-methoxy-2-methyl-propyl)benzimidazole-5-carboxylate (I-1366) was prepared in a manner as described for Intermediate I-2 using 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and Intermediate I-1365. ES/MS m/z: 467.2 (M+H+). Preparation of Intermediate I-1367 Methyl 4-amino-3-(((3R,4R)-4-ethoxytetrahydrofuran-3-yl)amino)benzoate (I-1367): methyl 4-amino-3-(((3R,4R)-4-ethoxytetrahydrofuran-3-yl)amino)benzoate was prepared in a manner described for Intermediate I-1351 substituting iodoethane for iodomethane. ES/MS m/z: 281.3 (M+H+). Preparation of Intermediate I-1368 Methyl 4-amino-3-(((3R,4R)-4-isopropoxytetrahydrofuran-3-yl)amino)benzoate (I-1368): methyl 4-amino-3-(((3R,4R)-4-isopropoxytetrahydrofuran-3-yl)amino)benzoate was prepared in a manner described for Intermediate I-1351 substituting 2-bromopropane for iodomethane. ES/MS m/z: 295.5 (M+H+). Preparation of Intermediates 1-1369, 1-1370, 1-1371, 1-1372 (4-amino-3-methyl-tetrahydrofuran-3-yl)methanol: Lithium aluminum hydride (2.0M in THF, 5.77 mL, 5.77 mmol) at 0° C. was added to a solution of ethyl 4-amino-3-methyl-tetrahydrofuran-3-carboxylate (500 mg, 2.89 mmol) in THE (1.0 M, 5.0 mL). The mixture was stirred at 0° C. The mixture was then warmed to 25° C. and stirred for 2 h. the mixture was then quenched with water (2 mL) and NaOH (2 m, 2.0 mL) diluted in EtOAc, dried over magnesium sulfate, filtered and concentrated in vaccuo.1H NMR (400 MHz, CDCl3) δ 4.14-4.04 (m, 1H), 3.81 (d, J=8.8 Hz, 1H), 3.72 (dd, J=18.4, 9.9 Hz, 1H), 3.60-3.50 (m, 2H), 3.48 (d, J=8.7 Hz, 1H), 3.39 (d, J=5.0 Hz, 1H), 3.28 (t, J=5.7 Hz, 1H), 1.10 (s, 2H). Methyl 3-((4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate: methyl 3-((4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate was prepared in a manner described for Intermediate I-103 substituting (4-amino-3-methyl-tetrahydrofuran-3-yl)methanol for (S)-4,4-dimethyltetrahydrofuran-3-amine and methyl 3-fluoro-4-nitrobenzoate for methyl 3,5-difluoro-4-nitrobenzoate. The mixture was purified by silica gel flash column chromatography (EtOAc/hexane) provided both cis and trans diastereomers. ES/MS m/z: 311.0 (M+H+). Methyl-rac-cis-3-((4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (rac-I-1369-1): The title compound was the earlier of two stereoisomers to elute during silica gel flash column chromatography. 1H NMR (400 MHz, CDCl3) δ 8.25 (d, J=8.9 Hz, 1H), 7.60 (d, J=1.6 Hz, 1H), 7.27 (s, 1H), 4.39 (dd, J=9.0, 6.9 Hz, 1H), 4.15 (d, J=7.1 Hz, 1H), 4.04 (d, J=9.1 Hz, 1H), 3.98 (s, 3H), 3.92-3.84 (m, 2H), 3.82-3.67 (m, 4H), 1.27 (s, 3H). Two-dimensional NOESY NMR identified no H17 correlation of C17-CH3to N11-NH and a strong C12 C—H correlation with C17-CH3to confirm relative stereochemistry as cis diastereomer. Methyl-rac-trans-3-((4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (rac-I-1371-1): The title compound was the later of two stereoisomers to elute during silica gel flash column chromatography. 1H NMR (400 MHz, CDCl3) δ 8.25 (d, J=8.9 Hz, 1H), 7.83 (d, J=1.7 Hz, 1H), 7.30 (d, J=1.6 Hz, 1H), 4.49-4.36 (m, 2H), 3.95 (d, J=15.9 Hz, 5H), 3.74-3.59 (m, 4H), 1.16 (s, 3H). Two-dimensional NOESY NMR identified a C17-CH3to N11-NH correlation and a very weak C12 C—H correlation with C17-CH3correlation to confirm relative stereochemistry as the trans diastereomer. Methyl 3-(((3S,4S)-4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1369, I-1370, relative stereochemistry established): was obtained via preparative chiral SFC (Daicel Chiralpak AD-H column with EtOH/CO2eluent) which gave 2 distinct stereoisomers. Methyl 3-(((3S,4S)-4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1369, Isomer 1, relative stereochemistry established): The earlier-eluting of two stereoisomers. Methyl 3-(((3S,4S)-4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1370, Isomer 2, relative stereochemistry established): The later-eluting of two stereoisomers. Methyl 3-(((3S,4R)-4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1371, I-1372, relative stereochemistry established): was obtained via preparative chiral SFC (Daicel Chiralpak IC column with MeOH/CO2eluent) which gave 2 distinct stereoisomers. Methyl 3-(((3S,4R)-4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1371, Isomer 1, relative stereochemistry established): The earlier-eluting of two stereoisomers. Methyl 3-(((3S,4R)-4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1372, Isomer 2, relative stereochemistry established): The later-eluting of two stereoisomers. Preparation of Intermediate I-1373 (Isomer 1): Methyl 3-(((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1373-1, relative stereochemistry established): N1,N1,N8,N8-tetramethylnaphthalene-1,8-diamine (66.3 mg, 0.31 mmol) followed by Trimethyloxonium Tetrafluoroborate (43.9 mg, 0.34 mmol) were added to a solution of methyl 3-((4-(hydroxymethyl)-4-methyltetrahydrofuran-3-yl)amino)-4-nitrobenzoate (I-1369) (96 mg, 0.31 mmol) in DCM (3.0 mL, 2.0 M) at 0° C. The mixture was stirred at 25° C. for 16 h, then diluted with DCM, filtered and purified by column chromatography. ES/MS m/z: 325.1 (M+H+). Methyl 4-amino-3-(((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)amino)benzoate (I-1373, Isomer 1, relative stereochemistry established): methyl 4-amino-3-(((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)amino)benzoate (relative stereochemistry established) was prepared in a manner as described for Intermediate I-107 using I-1373-1. ES/MS m/z: 295.2 (M+H+). Preparation of Intermediate I-1374 (Isomer 2) Methyl 4-amino-3-(((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)amino)benzoate (I-1374, Isomer 2, relative stereochemistry established): The title compound was prepared in a manner as described for Intermediate I-1373, using Intermediate I-1370. Preparation of Intermediate I-1375 (Isomer 1) Methyl 4-amino-3-(((3S,4R)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)amino)benzoate (I-1375, Isomer 1, relative stereochemistry established): The title compound was prepared in a manner as described for Intermediate I-1373, using Intermediate I-1371. Preparation of Intermediate I-1376 (Isomer 2) Methyl 4-amino-3-(((3S,4R)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)amino)benzoate (I-1376, Isomer 2, relative stereochemistry established): The title compound was prepared in a manner as described for Intermediate I-1373, using Intermediate I-1372. Preparation of Intermediate I-1377 (Isomer 1) Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1377, Isomer 1, relative stereochemistry established): Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1377) was prepared in a manner as described for Intermediate I-2 using 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and Intermediate I-1373. ES/MS m/z: 509.2 (M+H+). Preparation of Intermediate I-1378 (Isomer 2) Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1378, Isomer 2, relative stereochemistry established): Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1378) Intermediate I-2 using 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and Intermediate I-1374. ES/MS: 509.0 (M+H+). Preparation of Intermediate I-1379 (prophetic, Isomer 1) Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-((3S,4R)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1379, Isomer 1, relative stereochemistry established): methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1379) can be prepared in a manner as described for Intermediate I-2 using 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and Intermediate I-1375. Preparation of Intermediate I-1380 (Isomer 2) Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-((3S,4R)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-1380, Isomer 2, relative stereochemistry established): Methyl 2-(4-bromo-2,5-difluorobenzyl)-1-(4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1380) was prepared in a manner as described for Intermediate I-2 using 2-(4-bromo-2,5-difluoro-phenyl)acetic acid and Intermediate I-1376. ES/MS: 509.1 (M+H+). Preparation of Intermediate I-1381 6-bromo-3-fluoro-2-[(4-fluorophenyl)methoxy]pyridine (I-1381): 6-bromo-3-fluoro-2-[(4-fluorophenyl)methoxy]pyridine was prepared in a manner as described for Intermediate I-1129 substituting 1-(bromomethyl)-4-fluoro-benzene for 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine and 6-bromo-3-fluoro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 301.2 (M+H+). Preparation of Intermediate I-1382 2-bromo-6-[[4-(trifluoromethyl)phenyl]methoxy]pyridine (I-1382): 2-bromo-6-[[4-(trifluoromethyl)phenyl]methoxy]pyridine was prepared in a manner as described for Intermediate I-43 substituting (4-(trifluoromethyl)phenyl)methanol for (1-methylimidazol-4-yl)methanol. ES/MS: 334.0 (M+H+). Preparation of Intermediate I-1383 6-bromo-3-fluoro-2-[[4-(trifluoromethyl)phenyl]methoxy]pyridine (I-1383): 6-bromo-3-fluoro-2-[[4-(trifluoromethyl)phenyl]methoxy]pyridine was prepared in a manner as described for Intermediate I-1129 substituting 1-(bromomethyl)-4-(trifluoromethyl)benzene for 2-(bromomethyl)-3-fluoro-5-(trifluoromethyl)pyridine and 6-bromo-3-fluoro-pyridin-2-ol for 6-bromopyridin-2-ol. ES/MS: 350.0 (M+H+). Preparation of Intermediate I-1384 4-bromo-2-[[4-(trifluoromethyl)phenyl]methoxy]pyrimidine (I-1384): 4-bromo-2-[[4-(trifluoromethyl)phenyl]methoxy]pyrimidine was prepared in a manner as described for Intermediate I-43 substituting (4-(trifluoromethyl)phenyl)methanol for (1-methylimidazol-4-yl)methanol and 4-bromo-2-fluoropyrimidine for 4-bromo-2-fluoropyridine. ES/MS: 334.0 (M+H+). Preparation of Intermediate I-1385 Racemic (3S,4S)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-ol (I-1385-1): To a mixture of 3,6-dioxabicyclo[3.1.0]hexane (20.0 g, 1.0 equivalent) in DCM (200 mL) at 0° C. under argon atmosphere was added BF3-etherate (6.58 g, 0.2 equivalent). The resulting mixture was stirred at 0° C. for 20 min. Then 2,2-difluoroethan-1-ol (27.9 g, 1.2 equivalent) was added to the mixture at 0° C. The mixture was stirred for 16 h at RT, with monitoring by silica gel TLC (EtOAc/petroleum ether). The mixture was quenched with aqueous NaHCO3solution and extracted with DCM twice. The combined organic layer was washed with brine solution, dried over Na2SO4and evaporated under vacuum. Purification by silica gel flash column chromatography (ethyl acetate: petroleum ether) yielded the title compound. 1H NMR (400 MHz, CDCl3) δ 5.99-5.71 (m, 1H), 4.33-4.31 (m, 1H), 4.11-4.06 (m, 1H), 4.00-3.95 (m, 2H), 3.80-3.53 (m, 4H). Racemic (3S,4S)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl 4-methylbenzenesulfonate (I-1385-2): To a mixture of racemic (3S,4S)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-ol (I-1385-1, 1.3 g, 1.0 equivalent) and pyridine (7.8 mL) was added p-TsCl (1.59 g, 1.2 equivalent) at 0° C. and then the resulting solution was stirred at 60° C. for 16 h, with monitoring by TLC (EtOAc/petroleum ether). The mixture was cooled to RT, evaporated under reduced pressure. Then the mixture was purified by silica gel flash column chromatography (ethyl acetate: petroleum ether) yielded the title compound. 1H NMR (400 MHz, CDCl3) δ 7.86 (d, 2H), 7.51 (d, 2H), 6.21-6.05 (m, 1H), 4.95-4.93 (m, 1H), 4.18-4.16 (m, 1H), 3.93-3.89 (m, 1H), 3.84-3.80 (m, 1H), 3.67-3.55 (m, 4H), 2.43 (s, 3H). Racemic (3R,4R)-3-azido-4-(2,2-difluoroethoxy)tetrahydrofuran (I-1385-3): To a mixture of racemic (3S,4S)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl 4-methylbenzenesulfonate (I-1385-2, 1.3 g, 1.0 equivalent) in DMF (9.1 mL) was added sodium azide (1.24 g, 5.0 equivalent) at RT. The resulting mixture was stirred at 130° C. for 16 hr., with monitoring by TLC (EtOAc/petroleum ether). The mixture was cooled to RT, quenched with ice cold water. The organic layer was extracted with diethyl ether twice. The combined organic layer was washed with brine solution, dried over Na2SO4. To the organic layer was added methanol (4V) and the resulting mixture was concentrated under vacuum (30° C., 350 mmHg) to remove most of diethyl ether to yield the title compound, which was used directly in the next step. 1H NMR (400 MHz, CDCl3) δ 6.31-6.02 (m, 1H), 4.40-4.36 (m, 1H), 4.16-4.12 (m, 1H), 3.89-3.80 (m, 4H), 3.71-3.61 (m, 2H). Racemic (3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-amine (I-1385): To a mixture of racemic (3R,4R)-3-azido-4-(2,2-difluoroethoxy)tetrahydrofuran (I-1385-3, 1.22 g, 1.0 equivalent) in MeOH (20.4 mL) was added 10% Pd/C (0.2 g) The mixture was stirred under an atmosphere of hydrogen in a Parr apparatus (60) psi at RT for 16 hr, with monitoring by TLC (5% MeOH in DCM). The mixture was passed through a plug of celite and concentrated under reduced pressure. The mixture was diluted with DCM, then 4M HCl in 1,4-dioxane was added at 0° C. and stirred at RT for 1 h. The mixture was concentrated under reduced pressure and triturated with diethyl ether to yield the title compound. 1H NMR (400 MHz, DMSO-d6) δ 6.34-6.04 (m, 1H), 4.32-4.29 (m, 1H), 3.91-3.78 (m, 6H), 3.72-3.68 (m, 1H). ES/MS m/z 168.1 (M+H+). Preparation of Intermediate I-1386 Racemic methyl 4-amino-3-(((3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)amino)benzoate (I-1386): racemic methyl 4-amino-3-(((3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)amino)benzoate (relative stereochemistry known) was prepared in a manner as described for Intermediate I-1, using Intermediate I-1385. ES/MS m/z: 317.2 (M+H+). Preparation of Intermediate I-1387 4-bromo-2-((4-fluorobenzyl)oxy)pyrimidine (I-1387): 4-bromo-2-((4-fluorobenzyl)oxy)pyrimidine was prepared in a manner as described for Intermediate I-43 substituting (4-fluorophenyl)methanol for (1-methylimidazol-4-yl)methanol and 4-bromo-2-fluoropyrimidine for 4-bromo-2-fluoropyridine. ES/MS: 283.0 (M+H+). Preparation of Intermediate I-1395 Methyl 4-amino-3-((2,2-dimethyltetrahydrofuran-3-yl)amino)benzoate (I-1395): methyl 4-amino-3-((2,2-dimethyltetrahydrofuran-3-yl)amino)benzoate was prepared in a manner as described for Intermediate I-1, using 2,2-dimethyltetrahydrofuran-3-amine. B. Compound Examples Procedure 1: Example 1 Tert-butyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-indole-6-carboxylate: Tert-butyl (S)-4-amino-3-((tetrahydrofuran-3-yl)amino)benzoate (25 mg, 0.091 mmol), o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (47 mg, 0.12 mmol), and DIPEA (0.04 mL, 0.25 mmol were added to a solution of 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)acetic acid (33 mg, 0.083 mmol) in DMF (1 mL). The resulting solution was stirred at rt for 2 hrs. The mixture was then poured into H2O (5 mL) and extracted with EtOAc (2×15 mL). The combined organic extracts were washed with brine (5 mL), dried over MgSO4, and purified by silica gel chromatography (eluent: EtOAc/hexanes) to give desired product. The crude intermediate was then dissolved in acetic acid (1 mL) and heated to 60° C. for 3 hrs. The mixture was concentrated directly and purified by silica gel chromatography (eluent: EtOAc/hexanes): ES/MS: 641.6 (M+H+). (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 1): 0.25 mL TFA was added to a solution of tert-butyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-indole-6-carboxylate (76 mg, 0.057 mmol) in DCM (2 mL). The solution was stirred at 40° C. for 1 hour. The mixture was further diluted with EtOAc (30 mL), washed with water (3×5 mL), concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide Example 1. ES/MS: 585.3 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J=1.4 Hz, 1H), 7.97-7.85 (m, 3H), 7.83-7.71 (m, 3H), 7.68 (d, J=8.5 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.58-5.49 (m, 1H), 4.58 (s, 2H), 4.31 (td, J=8.7, 2.8 Hz, 1H), 4.22 (dd, J=10.4, 2.8 Hz, 1H), 3.97 (dd, J=10.5, 7.7 Hz, 1H), 3.72 (td, J=9.3, 7.0 Hz, 1H), 2.23-2.06 (m, 1H). Example 2: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxopyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxopyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 1. 1H NMR (400 MHz, DMSO) δ 8.22 (s, 1H), 8.09 (d, J=1.4 Hz, 1H), 7.97-7.82 (m, 3H), 7.81-7.71 (m, 3H), 7.68 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.38 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.68 (tt, J=10.0, 5.2 Hz, 1H), 5.60 (s, 2H), 4.51 (s, 2H), 3.89 (t, J=10.1 Hz, 1H), 3.62 (dd, J=10.9, 4.6 Hz, 1H), 2.86 (dd, J=17.9, 10.4 Hz, 1H), 2.62 (dd, J=17.8, 5.7 Hz, 1H). Example 3: (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxopyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxopyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 1. 1H NMR (400 MHz, DMSO) δ 8.22 (s, 1H), 8.09 (d, J=1.5 Hz, 1H), 7.96-7.82 (m, 3H), 7.81-7.71 (m, 3H), 7.68 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.6, 1.7 Hz, 1H), 7.38 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.68 (tt, J=10.1, 5.2 Hz, 1H), 5.60 (s, 2H), 4.51 (s, 2H), 3.89 (t, J=10.1 Hz, 1H), 3.61 (dd, J=11.0, 4.6 Hz, 1H), 2.86 (dd, J=17.9, 10.4 Hz, 1H), 2.62 (dd, J=17.9, 5.7 Hz, 1H). Example 4: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((1R,2S)-2-(difluoromethyl)cyclopropyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((1R,2S)-2-(difluoromethyl)cyclopropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 1. 1H NMR (400 MHz, Methanol-d4) δ 8.64-8.45 (m, 1H), 8.15 (td, J=9.0, 1.5 Hz, 1H), 7.91-7.78 (m, 2H), 7.78-7.66 (m, 2H), 7.66-7.48 (m, 3H), 7.33 (ddd, J=17.3, 11.2, 6.1 Hz, 1H), 7.09-6.83 (m, 1H), 6.16 (td, J=56.2, 4.3 Hz, 1H), 5.67-5.59 (m, 2H), 4.72 (d, J=3.1 Hz, 2H), 3.91 (q, J=5.4, 4.0 Hz, 1H), 2.36 (d, J=57.4 Hz, 1H), 1.94 (d, J=9.4 Hz, 1H), 1.86-1.75 (m, 1H). Example 5: (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 1. 1H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J=1.4 Hz, 1H), 7.97-7.85 (m, 3H), 7.83-7.71 (m, 3H), 7.68 (d, J=8.5 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.58-5.49 (m, 1H), 4.58 (s, 2H), 4.31 (td, J=8.7, 2.8 Hz, 1H), 4.22 (dd, J=10.4, 2.8 Hz, 1H), 3.97 (dd, J=10.5, 7.7 Hz, 1H), 3.72 (td, J=9.3, 7.0 Hz, 1H), 2.23-2.06 (m, 1H). Example 6: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-cyclopentyl-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-cyclopentyl-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 1. 1H NMR (400 MHz, DMSO-d6) δ 8.18 (d, J=1.4 Hz, 1H), 7.97-7.85 (m, 3H), 7.83-7.68 (m, 4H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.41 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.16 (p, J=8.8 Hz, 1H), 4.58 (s, 2H), 2.22-2.08 (m, 4H), 2.06-1.94 (m, 2H), 1.89-1.69 (m, 2H). Procedure 2: Example 7 Tert-butyl 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-(trifluoromethyl)-1H-pyrazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate: A suspension of tert-butyl 2-[[4-[6-[(4-bromo-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (25 mg, 0.037 mmol), 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-(trifluoromethyl)pyrazole (15 mg, 0.058 mmol), 1,1′-Bis(di-phenylphosphino)ferrocene palladium dichloride (4.0 mg, 0.0054 mmol), and potassium carbonate (15 mg, 0.11 mmol) in 1,4-Dioxane anhydrous, 99.8% (1.0 mL) and H2O (0.50 mL) was degassed with argon for 5 min. The mixture was then heated at 100° C. for 30 min. Following this time, the mixture was diluted with EtOAc, and washed with brine. The organic extract was dried over sodium sulfate and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. 1H NMR (400 MHz, Chloroform-d) δ 8.09-7.99 (m, 3H), 7.96 (dd, J=8.5, 1.5 Hz, 1H), 7.87 (dd, J=10.7, 6.3 Hz, 1H), 7.77 (d, J=8.5 Hz, 1H), 7.68 (t, J=7.8 Hz, 1H), 7.59 (t, J=7.7 Hz, 1H), 7.50 (dd, J=7.4, 1.5 Hz, 1H), 7.37-7.20 (m, 2H), 7.08 (dd, J=11.3, 6.0 Hz, 1H), 6.82 (d, J=8.2 Hz, 1H), 5.57 (s, 2H), 4.46 (s, 2H), 4.38 (t, J=5.2 Hz, 2H), 3.69 (t, J=5.2 Hz, 2H), 3.28 (s, 3H), 1.65 (s, 9H). 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-(trifluoromethyl)-1H-pyrazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid: To a solution of tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-[1-(trifluoromethyl)pyrazol-4-yl]phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (22.7 mg, 0.0308 mmol) in DCM (3 mL), was added 2,2,2-trifluoroacetic acid (0.103 mL, 1.35 mmol). The mixture was stirred at 40° C. for 1 hr. After 1 hr. more TFA (100 uL) was added and the mixture was heated until completion (3 hr.). The crude residue was concentrated to dryness and purified by RP-HPLC (eluent: MeCN/H2O). The resulting product fractions were diluted with EtOAc and neutralized with sodium bicarbonate solution. The organic extract was dried over sodium sulfate, filtered and concentrated to yield Example 7. ES/MS: 682.2 (M+H+); 1H NMR (400 MHz, DMSO-d6) δ 9.08 (s, 1H), 8.54 (s, 1H), 8.20 (s, 1H), 7.91-7.82 (m, 2H), 7.79 (dd, J=8.4, 1.6 Hz, 1H), 7.73 (d, J=11.5 Hz, 1H), 7.67-7.54 (m, 3H), 7.52 (d, J=7.3 Hz, 1H), 7.38 (dd, J=11.7, 6.1 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 6.53 (s, 1H), 5.53 (s, 2H), 4.59 (t, J=5.1 Hz, 2H), 4.44 (s, 2H), 3.68 (t, J=5.1 Hz, 2H), 3.21 (s, 3H). Procedure 3: Example 8 Methyl (R)-1-(5-(tert-butoxycarbonyl)-5-azaspiro[2.4]heptan-7-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)pyridin-2-yl)-2,5-difluorobenzyl)-1H-indole-6-carboxylate: N,N-Diisopropylethylamine (0.173 mL, 0.995 mmol) was added to a solution of 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetic acid (130 mg, 0.326 mmol), tert-butyl (7R)-7-(2-amino-5-methoxycarbonyl-anilino)-5-azaspiro[2.4]heptane-5-carboxylate (122 mg, 0.338 mmol), and o-(7-Azabenzotriazol-1-yl)-N,′,N′,N′-tetramethyluronium hexafluorophosphate (184 mg, 0.484 mmol) in DMF (3 mL). The solution was stirred overnight at rt. Following this time, the solution was diluted with EtOAc and washed with 5% LiCl, saturated NaHCO3, and brine. The organic extract was dried over sodium sulfate. The crude product was diluted with DCE (5 mL) and added acetic acid (1.26 mL, 22.0 mmol). The mixture was heated at 60° C. overnight. The next day the mixture was stirred at 80° C. until completion (8 hr.). The mixture was diluted with DCM and neutralized carefully with NaHCO3. The organic extract was dried over sodium sulfate. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. 19F NMR (376 MHz, Chloroform-d) δ −115.41 (t, J=8.2 Hz), −119.67, −124.02. Methyl (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-azaspiro[2.4]heptan-7-yl)-1H-indole-6-carboxylate: A solution of methyl 3-[(7R)-5-tert-butoxycarbonyl-5-azaspiro[2.4]heptan-7-yl]-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]benzimidazole-5-carboxylate (60.5 mg, 0.0836 mmol) and 2,2,2-trifluoroacetic acid (0.318 mL, 4.18 mmol) in DCM (5 mL) was stirred at rt for 4 hr. Following this time, the mixture was diluted with EtOAc and neutralized carefully with NaHCO3. The organic extract was dried over sodium sulfate. The extract was then concentrated and carried onto the next step without purification. ES/MS: 624.2 (M+H+); (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-azaspiro[2.4]heptan-7-yl)-1H-indole-6-carboxylic acid: A solution of methyl 3-[(7R)-5-azaspiro[2.4]heptan-7-yl]-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]benzimidazole-5-carboxylate (16.0 mg, 0.0257 mmol) and Lithium hydroxide, monohydrate (300 mmol/L, 0.257 mL, 0.0770 mmol) in CH3CN (3 mL) in a 40 mL glass vial was heated at 100° C. until completion (2 h). The mixture was diluted with EtOAc and brine. Next, 0.110 mL 1M citric acid was added to the mixture. The organic extract was dried over sodium sulfate, filtered and concentrated. Purified by RP-HPLC (eluent: MeCN/H2O). The resulting product fractions were diluted with EtOAc and neutralized with sodium bicarbonate solution. The organic extract was dried over sodium sulfate, filtered and concentrated to give Example 8. ES/MS: 610.2 (M+H+); 1H NMR (400 MHz, DMSO-d6) δ 8.42 (s, 1H), 7.97-7.85 (m, 2H), 7.82-7.67 (m, 4H), 7.63 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.7, 1.6 Hz, 1H), 7.31 (dd, J=11.5, 6.2 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.07 (s, 1H), 4.41 (s, 2H), 3.68 (t, J=10.4 Hz, 2H), 3.14 (d, J=11.0 Hz, 2H), 0.83 (d, J=23.4 Hz, 2H), 0.58 (m, 2H). Example 9: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-azaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-azaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 3. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.98-7.81 (m, 2H), 7.77 (ddd, J=16.8, 8.1, 1.5 Hz, 5H), 7.62 (d, J=8.5 Hz, 1H), 7.57-7.45 (m, 1H), 7.30 (d, J=10.5 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 6.54 (s, OH), 5.60 (s, 2H), 5.40 (s, OH), 5.01 (s, 1H), 4.40 (s, 2H), 3.60 (s, 1H), 3.11-2.99 (m, 1H), 0.76 (d, J=8.8 Hz, 1H), 0.58 (s, 3H), −0.06 (s, 1H). Example 10: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-methoxypyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3 S,4S)-4-methoxypyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 3. 1H NMR (400 MHz, DMSO-d6) δ 9.34 (s, 2H), 8.27 (d, J=1.4 Hz, 1H), 7.99-7.84 (m, 4H), 7.83-7.67 (m, 5H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.28 (td, J=10.2, 7.9 Hz, 1H), 4.68 (q, J=7.0 Hz, 1H), 4.56-4.34 (m, 2H), 3.34 (d, J=11.4 Hz, 1H), 3.15 (s, 3H). Example 11: (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 3. 1H NMR (400 MHz, DMSO) δ 13.09 (s, 1H), 8.35 (s, 1H), 7.96-7.86 (m, 2H), 7.83-7.70 (m, 3H), 7.59-7.42 (m, 3H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H). Example 12: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 3. 1H NMR (400 MHz, DMSO) δ 13.10 (s, 1H), 8.35 (s, 1H), 7.96-7.86 (m, 2H), 7.83-7.70 (m, 3H), 7.59-7.48 (m, 2H), 7.46 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.03 (d, J=6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.61 (s, 3H). Example 13: 4-bromo-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 4-bromo-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 3. 1H NMR (400 MHz, Methanol-d4) δ 8.71 (s, 1H), 8.17 (d, J=1.2 Hz, 1H), 7.90-7.67 (m, 3H), 7.66-7.47 (m, 3H), 7.19 (dd, J=11.5, 6.0 Hz, 1H), 6.92 (d, J=8.2 Hz, 1H), 5.63 (s, 2H), 4.55 (dd, J=11.4, 1.9 Hz, 3H), 4.44 (dd, J=11.3, 6.8 Hz, 1H), 3.93 (d, J=8.9 Hz, 1H), 3.78 (d, J=8.8 Hz, 1H), 3.37-3.34 (m, 2H), 1.30 (s, 3H), 0.61 (s, 3H). Procedure 4: Example 14 Methyl (R)-1-(5-acetyl-5-azaspiro[2.4]heptan-7-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-indole-6-carboxylate: Acetic anhydride (1000 mmol/L in DCM, 0.0696 mL, 0.0696 mmol) was added to a solution of methyl 3-[(7R)-5-azaspiro[2.4]heptan-7-yl]-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]benzimidazole-5-carboxylate, see Procedure 3, (34.7 mg, 0.0556 mmol) and N,N-Diisopropylethylamine (0.0291 mL, 0.167 mmol) in DCM (3 mL). The mixture was stirred overnight at rt. Following this time, the mixture was diluted with brine and EtOAc. The organic extract was dried over sodium sulfate. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 666.2 (M+H+). (R)-1-(5-acetyl-5-azaspiro[2.4]heptan-7-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-indole-6-carboxylic acid (Example 14): In a 40 mL glass vial a solution of methyl 3-[(7R)-5-acetyl-5-azaspiro[2.4]heptan-7-yl]-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]benzimidazole-5-carboxylate (23.7 mg, 0.0356 mmol) and lithium hydroxide, monohydrate (300 mmol/L, 0.356 mL, 0.107 mmol) in CH3CN (3 mL) was heated at 100° C. until completion (20 min). Upon completion, the solution was diluted with EtOAc and brine, followed by the addition of 0.110 mL 1M citric acid. The organic extract was dried over sodium sulfate, filtered and concentrated. Purified by RP-HPLC (eluent: MeCN/H2O). The resulting product fractions were diluted with EtOAc and neutralized with sodium bicarbonate solution. The organic extract was dried over sodium sulfate, filtered and concentrated to yield Example 14. ES/MS: 652.2 (M+H+); 1H NMR (400 MHz, DMSO-d6) δ 8.06 (s, 1H), 7.96-7.86 (m, 2H), 7.83-7.67 (m, 4H), 7.64-7.55 (m, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.34 (s, 1H), 5.19 (d, J=8.0 Hz, 1H), 4.35 (dt, J=28.0, 16.9 Hz, 3H), 4.23-4.07 (m, 1H), 4.07-3.82 (m, 2H), 3.82-3.53 (m, 1H), 2.10 (s, 2H), 2.03 (s, 1H), 1.07-0.78 (m, 2H), 0.78-0.53 (m, 1H), 0.29-0.04 (m, 1H). Example 15: (S)-1-(5-acetyl-5-azaspiro[2.4]heptan-7-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-1-(5-acetyl-5-azaspiro[2.4]heptan-7-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner described in Procedure 4. 1H NMR (400 MHz, DMSO-d6) δ 12.79 (s, 1H), 8.12 (s, OH), 8.06 (d, J=1.4 Hz, 1H), 7.96-7.84 (m, 2H), 7.83-7.66 (m, 4H), 7.63 (dd, J=8.5, 3.3 Hz, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.34 (s, OH), 5.19 (d, J=8.0 Hz, 1H), 4.63-4.22 (m, 3H), 4.19-4.05 (m, 1H), 3.99 (dd, J=13.5, 3.3 Hz, 1H), 3.92 (d, J=10.7 Hz, 1H), 3.82-3.46 (m, 2H), 2.10 (s, 2H), 2.03 (s, 1H), 1.08-0.75 (m, 2H), 0.71 (dd, J=10.2, 5.3 Hz, 1H), 0.17 (ddt, J=16.4, 11.2, 5.8 Hz, 1H). Example 16: 1-((3S,4S)-1-acetyl-4-methoxypyrrolidin-3-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-((3S,4S)-1-acetyl-4-methoxypyrrolidin-3-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner described in Procedure 4. 1H NMR (400 MHz, DMSO-d6) δ 8.12 (dd, J=37.9, 1.4 Hz, 1H), 7.96-7.81 (m, 3H), 7.79-7.71 (m, 3H), 7.68 (d, J=8.5 Hz, 1H), 7.56-7.51 (m, 1H), 7.43-7.33 (m, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.33 (dq, J=27.6, 8.0 Hz, 1H), 4.55 (dd, J=13.1, 6.1 Hz, 2H), 4.43 (dd, J=16.8, 5.7 Hz, 1H), 4.24-4.07 (m, 1H), 4.07-3.93 (m, 1H), 3.41 (ddd, J=84.8, 11.4, 6.6 Hz, 1H), 3.17 (d, J=5.9 Hz, 3H), 2.05 (d, J=24.1 Hz, 3H). Example 17: 1-((3R,4S)-4-acetamidotetrahydrofuran-3-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-((3R,4S)-4-acetamidotetrahydrofuran-3-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 4. Obtained as a mixture of enantiomers. 1H NMR (400 MHz, DMSO-d6) δ 8.25 (s, 1H), 8.00 (s, 1H), 7.97-7.86 (m, 4H), 7.76 (m, 3H), 7.54 (d, J=8.4 Hz, 2H), 6.99 (d, J=8.2 Hz, 2H), 5.64 (s, 2H), 4.64 (d, J=17.2 Hz, 5H), 4.53 (d, J=16.0 Hz, 3H). Example 18: 1-((3S,3aR,6aS)-5-acetylhexahydro-2H-furo[2,3-c]pyrrol-3-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-((3S,3aR,6aS)-5-acetylhexahydro-2H-furo[2,3-c]pyrrol-3-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner described in Procedure 4. 1H NMR (400 MHz, DMSO-d6) δ 8.57 (dd, J=15.4, 1.4 Hz, 1H), 7.95-7.85 (m, 2H), 7.84-7.68 (m, 4H), 7.63 (dd, J=8.5, 7.0 Hz, 1H), 7.54 (dt, J=7.5, 1.9 Hz, 1H), 7.44 (dt, J=11.3, 5.4 Hz, 1H), 7.00 (dd, J=8.3, 1.9 Hz, 1H), 5.66-5.52 (m, 3H), 4.76-4.57 (m, 2H), 4.57-4.45 (m, 3H), 4.13-4.05 (m, 2H), 4.00 (d, J=13.2 Hz, 0H), 3.89 (d, J=12.4 Hz, 1H), 3.55 (dd, J=12.5, 4.6 Hz, 1H), 3.40 (d, J=9.1 Hz, 0H), 3.28 (t, J=9.9 Hz, 0H), 3.21-3.06 (m, 1H), 2.76 (dd, J=12.9, 9.8 Hz, 1H), 2.70-2.57 (m, 1H), 2.42-2.31 (m, 1H), 1.74 (s, 3H), 0.89 (s, 1H). Procedure 5: Example 19 Tert-butyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)-1H-indole-6-carboxylate: To a solution of 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetic acid (120 mg, 0.301 mmol), tert-butyl 3-[[(3aS,4S,6aR)-2,3,3a,4,5,6a-hexahydrofuro[2,3-b]furan-4-yl]amino]-4-amino-benzoate (102 mg, 0.318 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (170 mg, 0.447 mmol) in DMF (3 mL), was added N,N-Diisopropylethylamine purified by redistillation, 99.5% (0.160 mL, 0.919 mmol). Next, the solution was stirred overnight at rt. Following this time, the solution was diluted with EtOAc and washed with 5% LiCl, saturated NaHCO3, and brine. The organic extract was dried over sodium sulfate. The crude product was diluted with DCE (5 mL) and acetic acid was added (1.26 mL, 22.0 mmol). The mixture was heated at 60° C. overnight. Following this time, the mixture was increased to 80° C. until completion (8 hr.). The mixture was diluted with DCM and neutralized carefully with NaHCO3. The organic extract was dried over sodium sulfate. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 683.2 (M+H+). 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)-1H-indole-6-carboxylic acid: To a solution of tert-butyl 3-[(3aS,4S,6aR)-2,3,3a,4,5,6a-hexahydrofuro[2,3-b]furan-4-yl]-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]benzimidazole-5-carboxylate (77.0 mg, 0.113 mmol) in DCM (3 mL), was added 2,2,2-trifluoroacetic acid (0.376 mL, 4.95 mmol). The solution was stirred at 40° C. for 75 min. Following this time, the crude residue was concentrated to dryness and purified by RP-HPLC (eluent: MeCN/H2O). The resulting product fractions were diluted with EtOAc and neutralized with sodium bicarbonate solution. The organic extract was dried over sodium sulfate, filtered and concentrated to give Example 19. ES/MS: 627.2 (M+H+); 1H NMR (400 MHz, DMSO-d6) δ 12.78 (s, 1H), 8.51 (d, J=1.4 Hz, 1H), 7.97-7.85 (m, 2H), 7.84-7.68 (m, 5H), 7.64 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.3, 1.6 Hz, 1H), 7.41 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.80 (d, J=5.3 Hz, 1H), 5.60 (s, 2H), 5.48-5.30 (m, 1H), 4.71 (dd, J=11.1, 2.5 Hz, 1H), 4.65-4.30 (m, 2H), 4.16 (dd, J=11.1, 7.0 Hz, 1H), 3.82-3.62 (m, 1H), 3.57-3.45 (m, 1H), 3.30 (s, 1H), 1.73-1.56 (m, 1H). Example 20: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,3aS,6aR)-hexahydrofuro[2,3-b]furan-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner described in Procedure 5. 1H NMR (400 MHz, DMSO-d6) δ 12.78 (s, 1H), 8.51 (d, J=1.4 Hz, 1H), 7.98-7.84 (m, 2H), 7.84-7.68 (m, 4H), 7.64 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.41 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.80 (d, J=5.3 Hz, 1H), 5.60 (s, 2H), 5.40 (ddd, J=9.5, 7.0, 2.6 Hz, 1H), 4.72 (dd, J=11.0, 2.6 Hz, 1H), 4.58-4.27 (m, 2H), 4.16 (dd, J=11.1, 6.9 Hz, 1H), 3.72 (td, J=8.6, 2.2 Hz, 1H), 3.62-3.42 (m, 1H), 1.61 (dtd, J=13.2, 10.7, 8.7 Hz, 1H), 0.77 (dd, J=13.3, 6.3 Hz, 1H). Example: 21: 2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 5. 1H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.13 (dd, J=8.6, 1.4 Hz, 1H), 8.00-7.81 (m, 2H), 7.81-7.73 (m, 1H), 7.68 (t, J=7.5 Hz, 1H), 7.58 (dd, J=7.4, 1.6 Hz, 1H), 7.46-7.30 (m, 3H), 7.01-6.85 (m, 1H), 6.79 (s, 1H), 5.61 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.70-4.61 (m, 3H), 4.52 (dd, J=11.5, 6.7 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.40 (s, 3H), 0.74 (s, 3H). Example 22: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-(difluoromethoxy)propyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-(difluoromethoxy)propyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 5. 1H NMR (400 MHz, Methanol-d4) δ 8.53 (t, J=0.9 Hz, 1H), 8.19 (dd, J=8.6, 1.4 Hz, 1H), 7.90-7.79 (m, 2H), 7.79-7.67 (m, 2H), 7.60 (ddt, J=9.3, 8.0, 1.5 Hz, 3H), 7.37 (dd, J=11.2, 6.0 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 6.26 (t, J=74.2 Hz, 1H), 5.64 (s, 2H), 4.82-4.62 (m, 5H), 1.54 (d, J=5.1 Hz, 3H). Procedure 6: Example 23 Tert-butyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: Cs2CO3(400 mg, 1.20 mmol) was added to solution of tert-butyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (400 mg, 0.80 mmol) and (5-bromo-3-fluoro-2-pyridyl)methyl 4-methylbenzenesulfonate (350 mg, 0.97 mmol) in 15 mL of acetonitrile. The resulting mixture was heated to 50° C. for 30 minutes. Following this time, the solution was cooled to rt, filtered, then concentrated. The crude material was purified by silica gel column chromatography (eluent: EtOAc/hexanes) to provide desired product. ES/MS: 686.0 (M+H+). 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid: 0.5 mL TFA was added to a solution of tert-butyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (64 mg, 0.094 mmol) in DCM (2.5 mL). The resulting solution stirred at rt for 1 hour. Upon completion the mixture was diluted with EtOAc (30 mL), washed with water (3×5 mL), concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide the final compound Example 23. ES/MS: 628.0 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.59 (t, J=1.3 Hz, 1H), 8.30 (d, J=1.5 Hz, 1H), 8.25 (dd, J=9.5, 1.9 Hz, 1H), 7.94-7.84 (m, 2H), 7.74 (dd, J=10.6, 6.4 Hz, 1H), 7.66 (d, J=8.5 Hz, 1H), 7.52 (dd, J=7.5, 1.6 Hz, 1H), 7.41 (dd, J=11.5, 6.1 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 5.58 (d, J=1.8 Hz, 2H), 4.66 (t, J=5.1 Hz, 2H), 4.52 (s, 2H), 3.72-3.68 (m, 2H), 3.21 (s, 3H). Example 24: 2-(4-(6-((5-chloropyrazin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((5-chloropyrazin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner described in Procedure 6. 1H NMR (400 MHz, DMSO-d6) δ 8.82 (d, J=1.3 Hz, 1H), 8.68 (d, J=1.3 Hz, 1H), 8.27 (d, J=1.5 Hz, 1H), 7.91 (t, J=7.9 Hz, 1H), 7.85 (dd, J=8.5, 1.5 Hz, 1H), 7.71 (dd, J=10.5, 6.4 Hz, 1H), 7.64 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.39 (dd, J=11.5, 6.1 Hz, 1H), 7.03 (d, J=8.3 Hz, 1H), 5.63 (s, 2H), 4.63 (t, J=5.1 Hz, 2H), 4.49 (s, 2H), 3.69 (t, J=5.0 Hz, 2H), 3.21 (s, 3H). Example 25: 2-(4-(6-((2,6-dimethylpyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((2,6-dimethylpyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.56-8.44 (m, 2H), 8.17 (dt, J=8.6, 1.4 Hz, 1H), 7.91-7.80 (m, 2H), 7.73 (dd, J=14.4, 8.3 Hz, 2H), 7.56 (dd, J=7.5, 1.6 Hz, 1H), 7.33 (dd, J=11.2, 6.1 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 5.65 (s, 2H), 4.76 (t, J=5.0 Hz, 2H), 4.71 (s, 2H), 3.81 (t, J=4.9 Hz, 2H), 3.30 (s, 3H), 2.83 (s, 3H), 2.74 (s, 3H). Example 26: 2-(4-(6-((2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.32 (dd, J=1.5, 0.7 Hz, 1H), 8.12 (dd, J=8.6, 1.5 Hz, 1H), 7.83 (dd, J=10.7, 6.3 Hz, 1H), 7.78-7.69 (m, 2H), 7.56-7.47 (m, 1H), 7.22 (ddd, J=8.7, 5.6, 3.2 Hz, 1H), 7.18-7.13 (m, 1H), 7.13-7.04 (m, 1H), 7.04-6.94 (m, 1H), 6.86 (dd, J=8.3, 0.7 Hz, 1H), 5.50 (d, J=1.2 Hz, 2H), 4.63-4.55 (m, 4H), 3.76 (t, J=5.0 Hz, 2H), 3.29 (s, 3H). Example 27: 2-(2,5-difluoro-4-(6-((2,4,5-trifluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2,4,5-trifluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.46 (d, J=1.3 Hz, 1H), 8.15 (dd, J=8.6, 1.5 Hz, 1H), 7.89 (dd, J=10.8, 6.3 Hz, 1H), 7.81 (t, J=7.9 Hz, 1H), 7.74 (d, J=8.5 Hz, 1H), 7.56 (dd, J=7.4, 1.6 Hz, 1H), 7.48 (ddd, J=10.8, 8.9, 6.6 Hz, 1H), 7.30 (dd, J=11.3, 6.0 Hz, 1H), 7.22 (td, J=10.0, 6.5 Hz, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 4.74 (t, J=5.0 Hz, 2H), 4.68 (s, 2H), 3.81 (t, J=4.9 Hz, 2H), 3.30 (s, 3H). Example 28: 2-(4-(6-((2,5-difluoro-4-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((2,5-difluoro-4-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.37 (d, J=1.4 Hz, 1H), 8.14 (dd, J=8.6, 1.5 Hz, 1H), 7.87 (dd, J=10.8, 6.3 Hz, 1H), 7.78-7.71 (m, 2H), 7.52 (dd, J=7.3, 1.6 Hz, 1H), 7.27-7.10 (m, 2H), 6.96 (dd, J=10.0, 6.1 Hz, 1H), 6.86 (d, J=8.1 Hz, 1H), 5.46 (s, 2H), 4.65 (t, J=5.0 Hz, 2H), 4.62 (s, 2H), 3.84-3.70 (m, 2H), 3.30 (s, 3H), 2.25 (d, J=2.0 Hz, 3H). Example 29: 2-(2,5-difluoro-4-(6-((5-methylpyrazin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((5-methylpyrazin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.65 (d, J=1.4 Hz, 1H), 8.58 (t, J=1.0 Hz, 1H), 8.53 (d, J=1.4 Hz, 1H), 8.25 (dd, J=8.6, 1.4 Hz, 1H), 7.85 (d, J=7.7 Hz, 1H), 7.82 (t, J=3.2 Hz, 1H), 7.79 (d, J=8.6 Hz, 1H), 7.58 (dd, J=7.5, 1.6 Hz, 1H), 7.37 (dd, J=11.2, 6.1 Hz, 1H), 6.98 (d, J=8.1 Hz, 1H), 5.61 (s, 2H), 4.84 (d, J=5.0 Hz, 2H), 4.78 (s, 2H), 3.91-3.79 (m, 2H), 3.32 (s, 3H), 2.56 (s, 3H). Example 30: 2-(2,5-difluoro-4-(6-(imidazo[1,2-a]pyridin-6-ylmethoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-(imidazo[1,2-a]pyridin-6-ylmethoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.99 (t, J=1.3 Hz, 1H), 8.49 (d, J=1.5 Hz, 1H), 8.29-8.22 (m, 1H), 8.15 (td, J=8.6, 1.5 Hz, 2H), 8.06 (d, J=2.2 Hz, 1H), 8.00-7.83 (m, 3H), 7.75 (d, J=8.5 Hz, 1H), 7.60 (dd, J=7.4, 1.6 Hz, 1H), 7.34 (dd, J=11.3, 6.0 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.69 (d, J=1.1 Hz, 2H), 4.76 (t, J=5.0 Hz, 2H), 4.70 (s, 2H), 3.88-3.73 (m, 2H), 3.31 (s, 3H). Example 31: 2-(4-(6-((3,5-difluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((3,5-difluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.56 (t, J=1.0 Hz, 1H), 8.39 (d, J=2.4 Hz, 1H), 8.23 (dd, J=8.6, 1.4 Hz, 1H), 7.94 (dd, J=10.9, 6.3 Hz, 1H), 7.88-7.74 (m, 2H), 7.66 (ddd, J=9.7, 8.5, 2.4 Hz, 1H), 7.58 (dd, J=7.3, 1.6 Hz, 1H), 7.36 (dd, J=11.2, 6.1 Hz, 1H), 6.91 (dd, J=8.3, 0.6 Hz, 1H), 5.62 (d, J=2.0 Hz, 2H), 4.83 (t, J=5.0 Hz, 2H), 4.77 (s, 2H), 3.85 (dd, J=5.4, 4.4 Hz, 2H), 3.33 (s, 3H). Example 32: 2-(2,5-difluoro-4-(6-((4-(trifluoromethoxy)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((4-(trifluoromethoxy)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.50 (d, J=1.4 Hz, 1H), 8.18 (dd, J=8.5, 1.5 Hz, 1H), 7.92-7.68 (m, 3H), 7.66-7.46 (m, 3H), 7.40-7.20 (m, 3H), 6.92 (d, J=8.3 Hz, 1H), 5.52 (s, 2H), 4.77 (t, J=5.0 Hz, 2H), 4.71 (s, 2H), 3.82 (t, J=4.9 Hz, 2H), 3.31 (s, 3H). Example 33: 2-(2,5-difluoro-4-(6-((2-methyl-6-(trifluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-methyl-6-(trifluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Methanol-d4) δ 8.53 (d, J=1.3 Hz, 1H), 8.20 (dd, J=8.6, 1.4 Hz, 1H), 8.05 (d, J=7.9 Hz, 1H), 7.91-7.70 (m, 3H), 7.64 (d, J=7.9 Hz, 1H), 7.57 (dd, J=7.4, 1.6 Hz, 1H), 7.35 (dd, J=11.2, 6.1 Hz, 1H), 6.98 (d, J=8.2 Hz, 1H), 5.62 (s, 2H), 4.83-4.64 (m, 4H), 3.88-3.78 (m, 2H), 3.32 (s, 3H), 2.69 (s, 3H). Example 34: 2-(4-(6-((2-chloro-6-(trifluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((2-chloro-6-(trifluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Chloroform-d) δ 8.24 (t, J=0.9 Hz, 1H), 8.13 (dd, J=8.6, 1.4 Hz, 1H), 8.10-8.03 (m, 1H), 7.99 (d, J=8.6 Hz, 1H), 7.82-7.69 (m, 2H), 7.65 (d, J=7.8 Hz, 1H), 7.56 (dd, J=7.5, 1.3 Hz, 1H), 7.35-7.32 (m, 1H), 6.92 (dd, J=8.2, 0.7 Hz, 1H), 5.63 (s, 2H), 4.74 (s, 2H), 4.57 (t, J=4.9 Hz, 2H), 3.78 (t, J=4.8 Hz, 2H), 3.32 (s, 3H). Example 35: 2-(2,5-difluoro-4-(6-((6-(2,2,2-trifluoroethoxy)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((6-(2,2,2-trifluoroethoxy)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6. 1H NMR (400 MHz, Chloroform-d) δ 8.24 (d, J=19.5 Hz, 2H), 8.09 (d, J=8.5 Hz, 1H), 7.96 (d, J=8.4 Hz, 1H), 7.91-7.73 (m, 2H), 7.68 (t, J=8.0 Hz, 1H), 7.50 (d, J=7.5 Hz, 1H), 6.90 (d, J=8.4 Hz, 1H), 6.79 (d, J=8.2 Hz, 1H), 5.42 (s, 2H), 4.78 (dd, J=16.8, 8.5 Hz, 4H), 4.56 (s, 2H), 3.78 (d, J=5.2 Hz, 2H), 3.33 (s, 3H). Procedure 7: Example 36 Tert-butyl 2-[[4-[6-[[5-[1-(difluoromethyl)pyrazol-4-yl]pyrazin-2-yl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: Aqueous sodium carbonate solution (0.13 mL, 0.2 mmol) was added to a solution of tert-butyl 2-[[4-[6-[(5-chloropyrazin-2-yl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-34) (60 mg, 0.097 mmol), 1-(difluoromethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (30 mg, 0.12 mmol), and bis(diphenylphosphino)ferrocene] dichloropalladium(II) (14 mg, 0.0020 mmol) in 1,4-dioxane (1.5 mL)ed. The resulting solution was degassed by bubbling argon for 1 minute, sealed and heated to 100° C. for 2 hr. Upon completion the mixture was poured into water (5 mL) and extracted with EtOAc (2×5 mL). The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give the desired product. ES/MS: 704.2 (M+1). 2-[[4-[6-[[5-[1-(difluoromethyl)pyrazol-4-yl]pyrazin-2-yl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid; 2,2,2-trifluoroacetic acid (Example 36): 0.5 mL TFA was added to a solution of tert-butyl 2-[[4-[6-[[5-[1-(difluoromethyl)pyrazol-4-yl]pyrazin-2-yl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (37 mg, 0.053 mmol) in DCM (2.5 mL). The resulting solution stirred at rt for 3 hours. Following this time, the mixture was diluted with EtOAc (30 mL), washed with water (3×5 mL), concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide the final compound Example 36. ES/MS: 648.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 9.13 (d, J=1.5 Hz, 1H), 8.98 (s, 1H), 8.78 (d, J=1.4 Hz, 1H), 8.45 (s, 1H), 8.26 (d, J=1.5 Hz, 1H), 8.09-7.71 (m, 4H), 7.63 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.6, 1.7 Hz, 1H), 7.39 (dd, J=11.5, 6.1 Hz, 1H), 7.03 (d, J=8.3 Hz, 1H), 5.62 (s, 2H), 4.63 (t, J=5.1 Hz, 2H), 4.48 (s, 2H), 3.69 (t, J=5.1 Hz, 2H), 3.20 (s, 3H). Example 37: 2-(2,5-difluoro-4-(6-((5-(1-methyl-11H-1,2,3-triazol-4-yl)pyrazin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((5-(1-methyl-1H-1,2,3-triazol-4-yl)pyrazin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 7. 1H NMR (400 MHz, DMSO-d6) δ 9.19 (d, J=1.5 Hz, 1H), 8.94 (d, J=1.4 Hz, 1H), 8.45 (s, 1H), 8.28 (d, J=1.5 Hz, 1H), 7.92 (t, J=7.9 Hz, 1H), 7.86 (dd, J=8.4, 1.5 Hz, 1H), 7.72 (dd, J=10.5, 6.4 Hz, 1H), 7.64 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.05 (d, J=8.3 Hz, 1H), 5.69 (s, 2H), 4.64 (t, J=5.1 Hz, 2H), 4.49 (s, 2H), 4.31 (s, 3H), 3.69 (t, J=5.0 Hz, 2H), 3.20 (s, 3H). Example 38: 2-(2,5-difluoro-4-(6-((5-(1-methyl-1H-pyrazol-4-yl)pyrazin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((5-(1-methyl-1H-pyrazol-4-yl)pyrazin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 7. 1H NMR (400 MHz, DMSO-d6) δ 8.97 (d, J=1.5 Hz, 1H), 8.70 (d, J=1.4 Hz, 1H), 8.41 (s, 1H), 8.28 (d, J=1.5 Hz, 1H), 8.10 (s, 1H), 7.90 (t, J=7.9 Hz, 1H), 7.86 (dd, J=8.4, 1.5 Hz, 1H), 7.78 (dd, J=10.5, 6.4 Hz, 1H), 7.65 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.01 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 4.65 (t, J=5.2 Hz, 2H), 4.51 (s, 2H), 3.90 (s, 3H), 3.69 (t, J=5.0 Hz, 2H), 3.21 (s, 3H). Procedure 8: Example 39 tert-butyl 2-[[4-[6-[(5-cyano-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: A solution of tert-butyl 2-[[4-[6-[(5-bromo-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-33) (70 mg, 0.10 mmol), zinc cyanide (14 mg, 0.12 mmol), zinc powder (0.3 mg, 0.005 mmol), and Pd(PPh3)4(60 mg, 0.050 mmol) in DMF (1.5 mL) was degassed by bubbling argon for 1 minute. The solution was then sealed and heated to 100° C. for 20 hrs. Upon completion the mixture was poured into water (5 mL) and extracted with EtOAc (2×5 mL). The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give the desired product. ES/MS: 630.2 (M+1). 2-[[4-[6-[(5-cyano-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid; 2,2,2-trifluoroacetic acid: 0.5 mL TFA was added to a solution of tert-butyl 2-[[4-[6-[(5-cyano-3-fluoro-2-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (51 mg, 0.081 mmol) in DCM (2.5 mL). The resulting solution stirred at rt for 3 hours. Upon completion the mixture was diluted with EtOAc (30 mL), washed with water (3×5 mL), concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide the final compound Example 39. ES/MS: 574.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.89 (t, J=1.2 Hz, 1H), 8.48 (dd, J=9.9, 1.7 Hz, 1H), 8.29 (d, J=1.5 Hz, 1H), 7.93-7.82 (m, 2H), 7.66 (d, J=8.5 Hz, 1H), 7.61 (dd, J=10.6, 6.4 Hz, 1H), 7.51 (dd, J=7.5, 1.6 Hz, 1H), 7.39 (dd, J=11.6, 6.1 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.69 (d, J=1.7 Hz, 2H), 4.65 (t, J=5.1 Hz, 2H), 4.51 (s, 2H), 3.69 (t, J=5.0 Hz, 2H), 3.21 (s, 3H). Procedure 9: Example 40 3-[(3R,4R)-1-acetyl-4-fluoro-pyrrolidin-3-yl]-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]benzimidazole-5-carboxylic acid (Example 40): 1-methylimidazole (82.4 mg, 0.08 mL, 1.00 mmol) followed by N,N,N′,N′-Tetramethylchloroformamidinium Hexafluorophosphate (60.0 mg, 0.214 mmol) were added to a solution of I-46 (82.0 mg, 0.21 mmol) and I-7 (70.0 mg, 0.176 mmol) in MeCN (2.50 mL) and cooled to 0° C. Next, the mixture was warmed to rt and stirred for 30 min. The crude mixture was concentrated in vacuo, then partitioned between 1M HCl and EtOAc. The organic layer was isolated and back extracted with an additional portion of EtOAc. The isolated organic layer was dried over magnesium sulfate, isolated by vacuum filtration, and concentrated in vacuo. The resulting crude material was dissolved in 1.5 mL of AcOH and the solution stirred for 3 days at 80° C. Upon completion the solution was concentrated, and the crude taken up into 1 mL of DMF and purified by reverse phase chromatography 10-70% MeCN/H2O with 0.1% TFA. Product containing fractions were combined and concentrated to give 3-[(3R,4R)-1-acetyl-4-fluoro-pyrrolidin-3-yl]-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]benzimidazole-5-carboxylic acid. Example 40. ES/MS m/z: 644.4 (M+H+);1H NMR (400 MHz, DMSO) δ 8.06 (dd, J=43.2, 1.5 Hz, 1H), 7.95-7.87 (m, 2H), 7.84 (dt, J=8.4, 1.9 Hz, 1H), 7.81-7.71 (m, 4H), 7.68 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.45-7.36 (m, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.83-5.62 (m, 1H), 5.62 (s, 2H), 4.63-4.39 (m, 2H), 4.32-4.06 (m, 2H), 4.02-3.86 (m, 1H), 2.09 (d, J=13.4 Hz, 3H). Procedure 10: Example 41 Tert-butyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-4-(difluoromethyl)-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: Tert-butyl 2-[[2,5-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (50 mg, 0.09 mmol), 4-[[6-chloro-4-(difluoromethyl)-2-pyridyl]oxymethyl]-3-fluoro-benzonitrile (32.5 mg, 0.1 mmol), 2 N of sodium carbonate (0.1 mL, 0.2 mmol) and (1,1′-bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (10.5 mg, 0.014 mmole) were suspended in dioxane (0.5 mL). The mixture was degassed by nitrogen. After which, the mixture was heated to 120° C. in the microwave reactor for 30 minutes. Upon completion the solvent was removed the solvent and crude product was dissolved in 1 mL of DMF. The mixture was filtered and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give the desired product. ES/MS: 679.3 (M+H+). 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-4-(difluoromethyl)-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (Example 41): Tert-butyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-4-(difluoromethyl)-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (35 mg, 0.052 mmol) was added to 1 mL of DCM and 0.4 mL of TFA stirred for 2 hrs. Upon completion the solvent was removed, and the crude residue was dissolved in 1 mL of DMF. The mixture was filtered and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give Example 41. ES/MS: 623.2 (M+H+). Example 42: 2-(4-(4-chloro-6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(4-chloro-6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 10. 1H NMR (400 MHz, Methanol-d4) δ 8.33 (d, J=1.4 Hz, 1H), 8.12 (dd, J=8.6, 1.5 Hz, 1H), 7.80-7.65 (m, 3H), 7.58-7.48 (m, 3H), 7.19 (dd, J=11.3, 6.0 Hz, 1H), 6.94 (d, J=1.4 Hz, 1H), 5.60 (s, 2H), 4.64-4.56 (m, 4H), 3.77 (t, J=5.0 Hz, 2H), 3.29 (s, 3H). Example 43: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-4-methoxypyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-4-methoxypyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 10. 1H NMR (400 MHz, DMSO-d6) δ 8.29 (d, J=1.5 Hz, 1H), 7.97-7.89 (m, 1H), 7.86 (dd, J=8.4, 1.5 Hz, 1H), 7.81-7.70 (m, 3H), 7.65 (d, J=8.4 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.08 (t, J=1.5 Hz, 1H), 6.57 (d, J=1.9 Hz, 1H), 5.58 (s, 2H), 4.65 (t, J=5.1 Hz, 2H), 4.51 (s, 2H), 3.87 (s, 3H), 3.70 (t, J=5.0 Hz, 2H), 3.21 (s, 3H). Example 44: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-4-hydroxypyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-4-hydroxypyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 10. 1H NMR (400 MHz, Methanol-d4) δ 8.52 (d, J=1.2 Hz, 1H), 8.19 (dd, J=8.6, 1.5 Hz, 1H), 7.83-7.74 (m, 2H), 7.68 (dt, J=12.8, 6.7 Hz, 1H), 7.63-7.52 (m, 2H), 7.30 (dd, J=11.3, 6.1 Hz, 1H), 7.07 (t, J=1.6 Hz, 1H), 6.29 (d, J=1.8 Hz, 1H), 5.58 (s, 2H), 4.78 (t, J=5.0 Hz, 2H), 4.71 (s, 2H), 3.82 (t, J=4.9 Hz, 2H). Example 45: 2-(4-(6-amino-2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-amino-2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 10. 1H NMR (400 MHz, DMSO-d6) δ 8.25 (s, 1H), 7.92 (d, J=10.0 Hz, 1H), 7.87-7.78 (m, 2H), 7.74 (s, 2H), 7.62 (d, J=8.5 Hz, 1H), 7.38 (dd, J=11.7, 6.0 Hz, 1H), 7.26 (s, 2H), 6.71 (d, J=1.4 Hz, 1H), 5.50 (s, 2H), 4.61 (t, J=5.1 Hz, 2H), 4.47 (s, 2H), 3.68 (t, J=5.1 Hz, 2H), 3.20 (s, 3H). Procedure 11: Example 46 Methyl 2-[6-(3-hydroxyphenyl)-3-pyridyl]acetate: Methyl 2-(6-chloro-3-pyridyl)acetate (300 mg, 1.62 mmol), (3-hydroxyphenyl)boronic acid (223 mg, 1.62 mmol), 2 N of sodium carbonate (1.62 mL, 3.23 mmol) and (1,1′-bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (57 mg, 0.08 mmole) was suspended in dioxane (5 mL). The mixture was degassed by nitrogen. After which, the mixture was heated to 100° C. in the microwave reactor for 45 minutes. Following this time, the solvent was removed, and the crude product was dissolved in 1 mL of DCM. The crude residue was purified by column chromatography (0-50% EtOAc in hexane) to give the title compound: ES/MS m/z: 244.1 (M+H+). 2-[6-[3-[(4-cyano-2-fluoro-phenyl)methoxy]phenyl]-3-pyridyl]acetic acid: A solution of methyl 2-[6-(3-hydroxyphenyl)-3-pyridyl]acetate (200.0 mg, 0.82 mmol), 4-(bromomethyl)-3-fluoro-benzonitrile (194 mg, 0.9 mmol) and potassium carbonate (227 mg, 1.64 mmol) in 5 mL of DMF, was stirred for 2 hrs. Next, 30 mL of water was added to the mixture and the compound was crashed out, filtered and washed with water. To the solid was added 3 mL of ACN and 1 mL of 1 N lithium hydroxide. The mixture was heated to 80° C. for 30 minutes. Upon completion the mixture was neutralized with 1 N hydrochloride in water and the organics were extracted with EtOAc (30 mL twice). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound: ES/MS m/z: 363.2 (M+H+). 2-[[6-[3-[(4-cyano-2-fluoro-phenyl)methoxy]phenyl]-3-pyridyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid: Tert-butyl 4-amino-3-(2-methoxyethylamino)benzoate (73.5 mg, 0.27 mmol) and O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (126 mg, 0.33 mmol) followed by N,N-diisopropylethylamine (0.24 mL, 1.38 mmol) were added to a solution of 2-[6-[3-[(4-cyano-2-fluoro-phenyl)methoxy]phenyl]-3-pyridyl]acetic acid (100 mg, 0.27 mmol) in DMF (1.0 mL). the resulting mixture was stirred for 2 hrs. at rt. Upon completion the mixture was diluted in 20 mL of EtOAc and washed with water (1×) and brine (1×). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was taken forward without further purification, assuming full conversion. The mixture was dissolved in AcOH (2.0 mL) and the mixture was heated to 60° C. for 2 hrs. The mixture was concentrated in vacuo and the crude residue was taken up in DCM and washed with saturated aqueous sodium bicarbonate. The layers were separated, and the aqueous layer was extracted with DCM (2×). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was added 1 mL of DCM and 0.4 mL of TFA stirred for 4 hours. Removed the solvent. The crude residue was dissolved in 2 mL of DMF. The mixture was filtered and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give Example 46. ES/MS: 537.4 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.74 (t, J=1.5 Hz, 1H), 8.52 (t, J=1.0 Hz, 1H), 8.19 (dd, J=8.6, 1.5 Hz, 1H), 8.12-8.00 (m, 2H), 7.80 (t, J=7.7 Hz, 1H), 7.75 (dd, J=8.6, 0.7 Hz, 1H), 7.70 (t, J=2.1 Hz, 1H), 7.67-7.60 (m, 3H), 7.50 (t, J=8.0 Hz, 1H), 7.21 (ddd, J=8.3, 2.6, 0.9 Hz, 1H), 5.35 (s, 2H), 4.81 (t, J=4.9 Hz, 2H), 4.77 (s, 2H), 3.84 (t, J=4.9 Hz, 2H), 3.32 (s, 3H). Procedure 12: Example 47 Methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-(3-hydroxy-3-methyl-but-1-ynyl)benzimidazole-5-carboxylate: Methyl 7-bromo-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (I-69) (20 mg, 0.028 mmol), 2-methylbut-3-yn-2-ol (11 mg, 0.13 mmol), copper iodide (3 mg, 0.16 mmol), bis(triphenylphosphine)palladium Chloride (4 mg, 0.006 mmole) and diisopropylamine (0.04 mL) was suspended in DMF (1 mL). The mixture was degassed by nitrogen. After which, the mixture was heated to 90° C. for 2 hrs. Next, the solvent was removed, and the crude product was dissolved in 1 mL of DMF. The mixture was filtered and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give the desired product. ES/MS: 709.6 (M+H+). (S) 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-(3-hydroxy-3-methyl-but-1-ynyl)benzimidazole-5-carboxylic acid: 1 mL of ACN and 0.4 mL of 1 N lithium hydroxide was added to a vial of Methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)-7-(3-hydroxy-3-methyl-but-1-ynyl)benzimidazole-5-carboxylate (10 mg, 0.014 mmol). The mixture was heated to 80° C. for 30 min. Upon completion the filtrate was purified by RP-HPLC (eluent: water/MeCN*0.1% TFA) to give Example 47. ES/MS: 695.3 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.69 (s, 1H), 8.04 (d, J=1.4 Hz, 1H), 7.93-7.67 (m, 3H), 7.67-7.48 (m, 3H), 7.15 (dd, J=11.0, 6.7 Hz, 1H), 7.00-6.85 (m, 1H), 5.63 (s, 2H), 5.00-4.92 (m, 1H), 4.64-4.39 (m, 4H), 3.93 (d, J=8.9 Hz, 1H), 3.78 (d, J=8.9 Hz, 1H), 2.00 (d, J=39.2 Hz, 1H), 1.64 (s, 6H), 1.30 (s, 3H), 0.61 (s, 3H). Example 48: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-(cyclopropylethynyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-(cyclopropylethynyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 12. 1H NMR (400 MHz, Methanol-d4) δ 8.67 (s, 1H), 8.01 (d, J=1.4 Hz, 1H), 7.90-7.69 (m, 3H), 7.69-7.47 (m, 3H), 7.19 (dd, J=11.3, 6.0 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.63 (s, 2H), 5.00-4.90 (m, 2H), 4.56 (dd, J=19.4, 8.0 Hz, 3H), 4.43 (dd, J=11.4, 6.8 Hz, 1H), 3.94 (d, J=8.9 Hz, 1H), 3.78 (d, J=8.9 Hz, 1H), 1.61 (ddd, J=12.8, 8.3, 5.1 Hz, 1H), 1.28 (s, 3H), 1.02-0.84 (m, 4H), 0.61 (s, 3H). Procedure 13: Example 49 2-((2′-amino-6-((4-cyano-2-fluorobenzyl)oxy)-5′-fluoro-[2,3′-bipyridin]-6′-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 49): 0.7 mL TFA was added to a solution of tert-butyl 2-((2′-amino-6-((4-cyano-2-fluorobenzyl)oxy)-5′-fluoro-[2,3′-bipyridin]-6′-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-70, 30 mg, 0.048 mmol) in DCM (2 mL). The resulting mixture was stirred at rt for 40 min. Upon completion the solution was heated at 35° C. for 1 hour. The mixture was concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give Example 49. ES/MS: 571.3 (M+H+). H NMR (400 MHz, Methanol-d4) δ 8.60 (t, J=1.0 Hz, 1H), 8.28 (dd, J=8.6, 1.4 Hz, 1H), 7.91-7.81 (m, 3H), 7.70 (t, J=7.5 Hz, 1H), 7.63-7.55 (m, 2H), 7.48 (d, J=7.5 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.56 (s, 2H), 4.87-4.82 (m, 4H), 3.96-3.71 (m, 2H), 3.30 (s, 3H). Procedure 14: Example 50 2-((8-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-6-fluoroimidazo[1,2-a]pyridin-5-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 50): A solution of tert-butyl 2-((2′-amino-6-((4-cyano-2-fluorobenzyl)oxy)-5′-fluoro-[2,3′-bipyridin]-6′-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-70, 41 mg, 0.065 mmol) and chloroacetaldehyde (50% in H2O, 0.09 mL, 0.65 mmol) in isopropanol (2 mL) was heated at 90° C. for 40 min. Upon completion the mixture was cooled to rt, diluted with EtOAc (25 mL), washed with water (5 mL), brine (5 mL), dried over MgSO4, filtered, concentrated and the crude residue purified by flash chromatography (EtOAc/hexanes). The residue was dissolved in TFA (0.5 mL) and stirred for 25 min. Following this time, the mixture was concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give Example 50: ES/MS: 595.3 (M+H+).1H NMR (400 MHz, Methanol-d4) δ 8.57 (d, J=9.5 Hz, 1H), 8.35 (d, J=1.4 Hz, 1H), 8.18 (s, 2H), 8.07 (dd, J=8.4, 7.5 Hz, 1H), 7.96 (dd, J=8.5, 1.5 Hz, 1H), 7.85 (d, J=7.4 Hz, 1H), 7.79 (t, J=7.6 Hz, 1H), 7.66-7.55 (m, 2H), 7.50 (d, J=8.5 Hz, 1H), 7.19 (d, J=8.3 Hz, 1H), 5.73 (s, 2H), 5.22 (dd, J=6.9, 2.1 Hz, 1H), 4.78 (t, J=4.8 Hz, 2H), 3.96-3.83 (m, 2H), 3.41 (s, 3H). Procedure 15: Example 51 and 52 2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)fluoromethyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid and 2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)difluoromethyl)-1-(2-methoxyethyl)-11H-benzo[d]imidazole-6-carboxylic acid: A suspension of tert-butyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-71, 50 mg, 0.08 mmol), n-fluorobenzenesulfonimide (75 mg, 0.24 mmol), potassium tert-butoxide (18 mg, 0.16 mmol) in THE (1.0 mL) was stirred for 30 min at rt. Upon completion additional n-fluorobenzenesulfonimide (75 mg, 0.24 mmol) was added followed by the addition of LiHMDS (1M in THF, 0.16 mL, 0.16 mmol) and the mixture was stirred for 15 min. Following this the mixture was diluted with EtOAc (25 mL), washed with water (5 mL), brine (5 mL), dried over MgSO4, filtered, concentrated and the crude residue, redissolved in TFA, and stirred for 10 min. Upon completion the mixture was concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give the title compounds. 2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)fluoromethyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 51): ES/MS: 591.2 (M+H+);1H NMR (400 MHz, Methanol-d4) δ 8.38 (d, J=1.4 Hz, 1H), 8.02 (dd, J=8.6, 1.5 Hz, 1H), 7.88-7.63 (m, 4H), 7.63-7.48 (m, 4H), 7.41 (d, J=45.5 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.79-4.62 (m, 2H), 3.89-3.67 (m, 2H), 3.25 (s, 3H). 19F NMR (376 MHz, Methanol-d4) δ −78.14, −117.82 (dd, J=9.6, 7.2 Hz), −121.92 (ddd, J=17.8, 11.2, 6.2 Hz), −124.47 (ddd, J=17.3, 11.0, 5.8 Hz), −179.57 (d, J=45.4 Hz). 2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)difluoromethyl)-1-(2-methoxyethyl)-11H-benzo[d]imidazole-6-carboxylic acid (Example 52): ES/MS: 609.3 (M+H+);1H NMR (400 MHz, Methanol-d4) δ 8.47-8.20 (m, 1H), 8.16-7.83 (m, 1H), 7.83-7.71 (m, 2H), 7.71-7.33 (m, 6H), 6.99-6.80 (m, 1H), 5.60-5.35 (m, 2H), 4.71-4.56 (m, 2H), 3.86-3.66 (m, 2H), 3.22 (s, 3H). 19F NMR (376 MHz, Methanol-d4) δ −78.33 (d, J=11.0 Hz), −91.17 (t, J=11.0 Hz), −117.83 (d, J=9.4 Hz), −120.07, −121.85. Procedure 16: Example 53 2-((8-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-6-fluoro-[1,2,4]triazolo[1,5-a]pyridin-5-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 53): A suspension of tert-butyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-70, 30 mg, 0.048 mmol) and DMF-DMA (200 mg, 1.7 mmol) in isopropanol (0.7 mL) was heated at 60° C. for 20 min, followed by an additional heating at 90° C. for 3 hours. Upon completion the mixture was concentrated, redissolved in isopropanol (0.7 mL) and pyridine. Upon completion hydroxylamine-o-sulfonic acid was added to the solution and heated at 40° C. for 25 min. Following this time, the solution was concentrated, dissolved in TFA (0.5 mL) and stirred for 10 min. Upon completion the mixture was concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give Example 53. ES/MS: 596.7 (M+H+). H NMR (400 MHz, Methanol-d4) δ 8.78 (d, J=7.5 Hz, 1H), 8.59 (d, J=10.5 Hz, 1H), 8.53 (d, J=6.9 Hz, 1H), 8.48 (s, 1H), 8.22-8.08 (m, 1H), 7.94 (t, J=7.9 Hz, 1H), 7.78 (t, J=7.6 Hz, 1H), 7.74-7.55 (m, 3H), 7.05 (d, J=8.3 Hz, 1H), 5.72 (s, 2H), 5.37-5.25 (m, 2H), 4.92 (t, J=4.9 Hz, 2H), 3.89 (t, J=4.9 Hz, 2H), 3.28 (s, 3H). Procedure 17: Example 54 2-(1-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorophenyl)-2-hydroxyethyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 54): LiHMDS (1M in THF, 0.12 mL, 0.12 mmol) was added to a solution of tert-butyl 2-((3′-((4-cyano-2-fluorobenzyl)oxy)-2,5-difluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-71, 46 mg, 0.073 mmol) and paraformaldehyde (8.8 mg, 0.29 mmol) in THE (1.0 mL) at 0° C., and stirred for 5 min. Upon completion the reaction was quenched with aqueous TFA and concentrated. The residue was redissolved in DCM (0.3 mL) and TFA (0.5 mL) was added and stirred for 2 hours. The mixture was concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give Example 54. ES/MS: 603.3 (M+H+);1H NMR (400 MHz, Methanol-d4) δ 8.49 (t, J=1.0 Hz, 1H), 8.20 (dd, J=8.6, 1.4 Hz, 1H), 7.86 (d, J=8.6 Hz, 1H), 7.84-7.75 (m, 2H), 7.72 (t, J=7.5 Hz, 1H), 7.66-7.50 (m, 4H), 7.42 (dd, J=11.8, 6.0 Hz, 1H), 6.95 (d, J=8.2 Hz, 1H), 5.62 (s, 2H), 5.25 (t, J=6.1 Hz, 1H), 4.82-4.73 (m, 1H), 4.67 (ddd, J=15.3, 7.2, 3.5 Hz, 1H), 4.40 (dd, J=11.0, 6.5 Hz, 1H), 4.32 (dd, J=11.0, 5.7 Hz, 1H), 3.85-3.66 (m, 2H), 3.21 (s, 3H). Procedure 18: Example 55 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-4-(pyrimidin-5-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 55): A solution of methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-iodo-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-72, 7 mg, 0.01 mmol), pyrimidin-5-ylboronic acid (3.7 mg, 0.03 mmol), Pd(dppf)Cl2(0.7 mg, 0.001 mmol), and K2CO3(5.5 mg, 0.04 mmol) in toluene (0.4 mL) was degassed with argon, and stirred for 30 min at 90° C. Upon completion the mixture was diluted with EtOAc (25 mL), washed with water (5 mL), brine (5 mL), dried over MgSO4, filtered, concentrated and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give Example 55. ES/MS: 651.3 (M+H+);1H NMR (400 MHz, Acetonitrile-d3) δ 8.32 (d, J=1.4 Hz, 1H), 8.21 (d, J=1.4 Hz, 1H), 7.99 (s, 2H), 7.88-7.77 (m, 2H), 7.73 (t, J=7.5 Hz, 1H), 7.64-7.51 (m, 3H), 7.21 (dd, J=11.7, 6.1 Hz, 1H), 6.92 (d, J=8.3 Hz, 1H), 5.63 (s, 2H), 4.56 (t, J=5.0 Hz, 2H), 4.49 (s, 2H), 3.74 (t, J=5.0 Hz, 2H), 3.25 (s, 3H). Example 56: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-4-(1-(2-morpholinoethyl)-1H-pyrazol-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-4-(1-(2-morpholinoethyl)-1H-pyrazol-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 18. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.53 (s, 1H), 8.33 (d, J=0.7 Hz, 1H), 8.16 (d, J=1.5 Hz, 1H), 8.12 (d, J=1.5 Hz, 1H), 7.88-7.76 (m, 2H), 7.73 (t, J=7.6 Hz, 1H), 7.64-7.50 (m, 3H), 7.24 (dd, J=11.7, 6.1 Hz, 1H), 6.92 (dd, J=8.3, 0.6 Hz, 1H), 5.63 (s, 2H), 4.61 (t, J=5.8 Hz, 2H), 4.55-4.43 (m, 4H), 3.88 (s, 8H), 3.72 (t, J=5.0 Hz, 2H), 3.63 (t, J=5.8 Hz, 2H), 3.23 (s, 3H). Procedure 19: Example 57 & 58 2-(2,5-difluoro-4-(6-((4-fluoro-6-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid as a mixture of 2 stereoisomers were separated by chiral SFC (SFC IA column with MeOH cosolvent) to give two distinct stereoisomers. 2-(2,5-difluoro-4-(6-((4-fluoro-6-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 1, Example 57): ES/MS: 669.6 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.69 (d, J=10.4 Hz, 1H), 8.51 (s, 1H), 8.35 (s, 1H), 8.06 (s, 1H), 8.00-7.86 (m, 2H), 7.83 (dd, J=8.4, 1.5 Hz, 1H), 7.73-7.61 (m, 2H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.48 (dd, J=11.2, 6.3 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.55 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.65-4.51 (m, 2H), 4.49-4.36 (m, 2H), 3.89 (s, 3H), 3.79 (d, J=8.7 Hz, 1H), 3.74 (d, J=8.6 Hz, 1H), 1.34 (s, 3H), 0.62 (s, 3H). 2-(2,5-difluoro-4-(6-((4-fluoro-6-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 2, Example 58): ES/MS: 669.6 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.69 (d, J=10.4 Hz, 1H), 8.51 (s, 1H), 8.35 (s, 1H), 8.06 (s, 1H), 8.00-7.86 (m, 2H), 7.83 (dd, J=8.4, 1.5 Hz, 1H), 7.73-7.61 (m, 2H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.48 (dd, J=11.2, 6.3 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.55 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.65-4.51 (m, 2H), 4.49-4.36 (m, 2H), 3.89 (s, 3H), 3.79 (d, J=8.7 Hz, 1H), 3.74 (d, J=8.6 Hz, 1H), 1.34 (s, 3H), 0.62 (s, 3H). Procedure 20: Example 59, 60 (Method 1) 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylic acid (Example 78) as a mixture of 2 stereoisomers was separated by chiral SFC (SFC AD-H column with EtOH cosolvent) to give two distinct stereoisomers. (R)-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylic acid (Isomer 1, Example 59). Earlier eluting of two isomers ES/MS: 613.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 12.77 (s, 1H), 8.48 (s, 1H), 7.96-7.86 (m, 2H), 7.83-7.70 (m, 4H), 7.61 (d, J=8.5 Hz, 1H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.45 (dd, J=11.3, 6.2 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.58-4.47 (m, 2H), 4.44 (dd, J=11.1, 6.8 Hz, 1H), 4.36 (d, J=16.9 Hz, 1H), 3.82-3.70 (m, 2H), 1.33 (s, 3H), 1.24 (s, 1H), 0.60 (s, 3H). (S)-2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylic acid (Isomer 2, Example 60): Later eluting of two isomers. ES/MS: 613.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 7.96-7.85 (m, 2H), 7.85-7.68 (m, 4H), 7.63 (d, J=8.5 Hz, 1H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.46 (dd, J=11.3, 6.2 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.02 (d, J=6.6 Hz, 1H), 4.58-4.49 (m, 2H), 4.49-4.34 (m, 2H), 3.82-3.70 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluorophenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylic acid (Example 60, Method 2): Example 60 was also prepared in a manner similar to Procedure 22, with the following modifications: A suspension of methyl 2-[[4-[6-[(4-cyano-2-fluorophenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-101, 2020 mg, 3.2 mmol) and lithium hydroxide, monohydrate (300 mmol/L, 24 mL, 7.2 mmol) in in CH3CN (24 mL) was heated at 90° C. for until complete conversion to product (˜20 min). The mixture was cooled to rt and diluted with EtOAc and brine. Then 7.3 mL 1M citric acid was added. The organic extract was dried over sodium sulfate, concentrated and purified by reverse-phase preparative HPLC (water/MeCN gradient, with 0.1% TFA) to provide Example 60. 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluorophenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylic acid (Example 60, Method 3). Example 60 was also prepared in a manner similar to Procedure 22 with the following modifications: LiOH (2N in H2O, 20.5 mL, 41.1 mmol) was added to a solution of methyl 2-[[4-[6-[(4-cyano-2-fluorophenyl)methoxy]-2-pyridyl]-2,5-difluorophenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-101, 11.7 g, 18.7 mmol) in ACN (90 mL) and water (70 mL), and the resulting solution was heated to 95° C. for 40 min. The mixture was partitioned between EtOAc and brine acidified with 20.5 mL 1M citric acid. Phases were separated, and aqueous phase again extracted with EtOAc. Combined organic layers were dried over MgSO4, filtered, concentrated, and purified by silica gel flash chromatography (EtOAc/Hexane gradient) to give Example 60. Procedure 21: Example 61 Tert-butyl 2-[[2,5-difluoro-4-[6-[(2-fluoro-4-imidazol-1-yl-phenyl)methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: Copper(I) chloride (0.6 mg, 0.006 mmol) and imidazole (6.5 mg, 0.096 mmol) were added to a solution of tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-88) (70 mg, 0.096 mmol) in MeOH (3 mL). The mixture was stirred at 60° C. for 3 hrs. Following this time, the mixture was concentrated directly, and the crude residue purified by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give the desired product. 2-[[2,5-difluoro-4-[6-[(2-fluoro-4-imidazol-1-yl-phenyl)methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (Example 61): 0.25 mL TFA was added to a solution of tert-butyl 2-[[2,5-difluoro-4-[6-[(2-fluoro-4-imidazol-1-yl-phenyl)methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (15 mg, 0.022 mmol) in DCM (2 mL). The resulting solution stirred at 40° C. for 1 hour. Following this time mixture was concentrated directly and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide Example 61. ES/MS: 614.1 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 9.32 (s, 1H), 8.25-8.16 (m, 2H), 7.94-7.77 (m, 5H), 7.73-7.64 (m, 2H), 7.60 (d, 1H), 7.54 (dd, 1H), 7.40 (dd, 1H), 6.97 (d, 1H), 5.60 (s, 2H), 4.61 (t, 2H), 4.46 (s, 2H), 3.69 (t, 2H), 3.22 (s, 3H). Procedure 22: Example 62 Methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-spiro[2.2]pentan-2-yl-benzimidazole-5-carboxylate: N,N-Diisopropylethylamine (0.147 mL, 0.842 mmol) was added to a solution of 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetic acid (110 mg, 0.276 mmol), methyl 4-amino-3-(spiro[2.2]pentan-2-ylamino)benzoate (60.0%, 89.5 mg, 0.231 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (156 mg, 0.410 mmol) in DMF (5 mL)ed. The solution was stirred at rt overnight. Following this time, the solution was diluted with EtOAc and washed with 5% LiCl, saturated NaHCO3, and brine. The organic extract was dried over sodium sulfate. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes). The resulting product was diluted with DCE (4 mL) and acetic acid (0.12 mL, 2.1 mmol) was added. The mixture was heated at 80° C. for 4 hr. Upon completion the mixture was diluted with DCM and neutralized carefully with NaHCO3. The organic extract was dried over sodium sulfate. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 595.2 (M+H+). 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-spiro[2.2]pentan-2-yl-benzimidazole-5-carboxylic acid (Example 62): Lithium hydroxide, monohydrate (0.3 M, 0.148 mL, 1.86 mmol) in CH3CN (3 mL) was added to a solution of methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-spiro[2.2]pentan-2-yl-benzimidazole-5-carboxylate (8.80 mg, 0.0148 mmol) in CH3CN (3 mL)d. The mixture was heated at 80° C. until completion (˜2 hr.). Upon completion the mixture was diluted with EtOAc and brine and 0.120 mL 1M citric acid was added. The organic extract was dried over sodium sulfate, filtered and concentrated. Purified by RP-HPLC (eluent: MeCN/H2O). The resulting product fractions were diluted with EtOAc and neutralized with sodium bicarbonate solution. The organic extract was dried over sodium sulfate, filtered and concentrated to give Example 62. ES/MS: 581.2 (M+H+); 1H NMR (400 MHz, DMSO-d6) δ 7.94 (s, 1H), 7.92-7.87 (m, 2H), 7.80-7.65 (m, 4H), 7.61 (d, J=8.5 Hz, 1H), 7.53 (d, J=7.3 Hz, 1H), 7.44 (dd, J=11.5, 6.0 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.34 (s, 2H), 3.91 (dd, J=7.2, 4.0 Hz, 1H), 1.94-1.77 (m, 2H), 1.36 (s, 1H), 1.08 (td, J=9.2, 4.6 Hz, 2H), 0.90 (m, 1H). Example 63: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(spiro[2.4]heptan-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(spiro[2.4]heptan-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.24 (d, J=1.4 Hz, 1H), 8.00-7.82 (m, 2H), 7.82-7.67 (m, 4H), 7.62 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.31 (dd, J=11.4, 6.1 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.97 (t, J=8.4 Hz, 1H), 4.38 (s, 2H), 2.26 (ddt, J=30.5, 18.5, 6.2 Hz, 3H), 2.04 (d, J=7.1 Hz, 1H), 1.87 (dq, J=11.9, 6.6, 5.8 Hz, 1H), 1.77-1.57 (m, 1H), 0.83-0.61 (m, 1H), 0.55 (dt, J=9.3, 5.1 Hz, 1H), 0.48 (dd, J=9.2, 3.2 Hz, 1H), −0.09 (dd, J=10.0, 4.9 Hz, 1H). Example 64: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(spiro[2.3]hexan-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(spiro[2.3]hexan-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 12.84 (s, 1H), 8.50 (d, J=1.5 Hz, 1H), 7.99-7.84 (m, 2H), 7.82 (dd, J=8.4, 1.5 Hz, 1H), 7.78-7.70 (m, 3H), 7.64 (d, J=8.2 Hz, 1H), 7.51 (dd, J=7.5, 1.7 Hz, 1H), 7.22 (dd, J=11.5, 6.0 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.37 (dd, J=9.1, 7.0 Hz, 1H), 4.40 (s, 2H), 2.99-2.78 (m, 1H), 2.76-2.52 (m, 1H), 2.45-2.15 (m, 2H), 0.59 (td, J=7.4, 6.7, 4.3 Hz, 2H), 0.44-0.22 (m, 1H), 0.22-0.05 (m, 1H). Example 65: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((1S,2R)-2-methoxycyclobutyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((1S,2R)-2-methoxycyclobutyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.45 (d, J=1.4 Hz, 1H), 7.95-7.86 (m, 2H), 7.82 (dd, J=8.5, 1.5 Hz, 1H), 7.79-7.69 (m, 3H), 7.63 (d, J=8.5 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.34 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.48-5.32 (m, 1H), 4.59-4.42 (m, 2H), 4.30 (dd, J=5.9, 3.2 Hz, 1H), 3.22-3.07 (m, 1H), 2.95 (s, 3H), 2.63 (pd, J=9.7, 8.8, 3.3 Hz, 1H), 2.24 (ddd, J=14.2, 9.1, 5.5 Hz, 1H), 2.12 (t, J=11.7 Hz, 1H). Example 66: 1-(2-oxabicyclo[2.1.1]hexan-4-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-(2-oxabicyclo[2.1.1]hexan-4-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J=1.4 Hz, 1H), 7.94-7.88 (m, 2H), 7.84-7.71 (m, 4H), 7.64 (d, J=8.4 Hz, 1H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.41 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 4.77 (s, 1H), 4.40 (s, 2H), 3.94 (s, 2H), 2.61 (t, J=1.7 Hz, 4H). Example 67: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(pyrrolidin-1-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(pyrrolidin-1-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J=1.4 Hz, 1H), 7.94-7.86 (m, 2H), 7.84 (dd, J=8.5, 1.5 Hz, 1H), 7.78-7.71 (m, 3H), 7.69 (d, J=8.5 Hz, 1H), 7.52 (dd, J=7.6, 1.6 Hz, 1H), 7.38 (dd, J=11.6, 6.0 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.59 (s, 2H), 4.39 (s, 2H), 3.38-3.25 (m, 4H), 2.13-1.94 (m, 4H). Example 68: 1-(2-oxabicyclo[3.1.1]heptan-4-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-(2-oxabicyclo[3.1.1]heptan-4-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.68 (d, J=1.4 Hz, 1H), 7.96-7.86 (m, 2H), 7.82 (dd, J=8.5, 1.5 Hz, 1H), 7.80-7.70 (m, 3H), 7.63 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.44 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.50-5.36 (m, 1H), 4.73-4.61 (m, 2H), 4.58-4.37 (m, 3H), 3.07 (p, J=5.6 Hz, 1H), 2.36 (ddd, J=9.6, 5.7, 3.5 Hz, 1H), 2.27 (t, J=10.0 Hz, 1H), 2.09 (q, J=5.3, 4.3 Hz, 1H), 2.04-1.96 (m, 1H). Example 69: 1-(3-oxabicyclo[3.1.0]hexan-1-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-(3-oxabicyclo[3.1.0]hexan-1-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 7.95-7.88 (m, 2H), 7.83 (dd, J=8.4, 1.5 Hz, 1H), 7.82-7.71 (m, 4H), 7.65 (d, J=8.6 Hz, 1H), 7.58-7.52 (m, 1H), 7.46 (s, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 4.49 (d, J=7.5 Hz, 2H), 4.21 (dd, J=8.9, 2.8 Hz, 1H), 4.03-3.91 (m, 1H), 3.79-3.70 (m, 3H), 1.83-1.54 (m, 1H), 1.42 (s, 1H). Example 70: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methoxytetrahydro-2H-pyran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methoxytetrahydro-2H-pyran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.91 (t, J=9.9 Hz, 2H), 7.85-7.68 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.40 (dd, J=11.4, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 4.87 (dd, J=9.0, 4.7 Hz, 1H), 4.63 (d, J=16.9 Hz, 1H), 4.59-4.46 (m, 2H), 3.90 (dd, J=10.7, 4.6 Hz, 1H), 3.84-3.77 (m, 2H), 3.14 (s, 3H), 1.99 (q, J=4.6, 4.0 Hz, 3H). Example 71: 1-((1R,4R,6S)-2-oxabicyclo[2.2.1]heptan-6-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-((1R,4R,6S)-2-oxabicyclo[2.2.1]heptan-6-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 7.96-7.87 (m, 2H), 7.83 (d, J=8.5 Hz, 1H), 7.81-7.70 (m, 3H), 7.63 (d, J=8.4 Hz, 1H), 7.59-7.48 (m, 1H), 7.41 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.07 (dd, J=11.5, 5.6 Hz, 1H), 4.54 (d, J=12.8 Hz, 3H), 4.06 (d, J=7.1 Hz, 1H), 3.95-3.83 (m, 1H), 2.81 (t, J=3.1 Hz, 1H), 2.40 (t, J=13.9 Hz, 1H), 2.18-1.95 (m, 2H), 1.83 (d, J=10.4 Hz, 1H). Example 72: 1-((3R,4S)-4-((tert-butoxycarbonyl)amino)tetrahydrofuran-3-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-((3R,4S)-4-((tert-butoxycarbonyl)amino)tetrahydrofuran-3-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid (as a racemic mixture of enantiomers) was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.44 (s, 1H), 7.94-7.86 (m, 2H), 7.81-7.70 (m, 4H), 7.54 (dd, J=11.0, 7.7 Hz, 2H), 7.30 (dd, J=11.2, 6.4 Hz, 1H), 7.09 (s, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.62 (s, 2H), 5.40 (s, 1H), 4.63-4.43 (m, 4H), 4.30 (d, J=17.0 Hz, 1H), 4.20 (dd, J=10.9, 7.0 Hz, 1H), 4.08-3.89 (m, 3H), 1.12 (s, 9H). Example 73: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2R,3R)-2-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2R,3R)-2-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (as a racemic mixture of enantiomers) was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.32 (d, J=1.4 Hz, 1H), 7.94-7.87 (m, 2H), 7.83 (dd, J=8.5, 1.5 Hz, 1H), 7.80-7.71 (m, 3H), 7.65 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.5 Hz, 1H), 7.43 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 4.54 (d, J=5.8 Hz, 4H), 4.38-4.32 (m, 2H), 4.00 (p, J=6.2 Hz, 1H), 3.73 (q, J=9.0 Hz, 1H), 0.80 (d, J=6.3 Hz, 3H). Example 74: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2,2-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2,2-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.25 (d, J=1.5 Hz, 1H), 7.96-7.87 (m, 2H), 7.82-7.69 (m, 4H), 7.63 (d, J=8.4 Hz, 1H), 7.54 (d, J=6.7 Hz, 1H), 7.43 (dd, J=11.4, 6.4 Hz, 2H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.01 (s, 2H), 4.48 (t, J=14.0 Hz, 3H), 4.29 (d, J=3.5 Hz, 2H), 3.99 (d, J=8.4 Hz, 2H), 1.29 (s, 3H), 0.77 (s, 3H). Example 75: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(methylcarbamoyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(methylcarbamoyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (as a racemic mixture of enantiomers) was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.37 (d, J=1.4 Hz, 1H), 7.99 (q, J=4.6 Hz, 1H), 7.96-7.82 (m, 3H), 7.82-7.71 (m, 3H), 7.68 (d, J=8.4 Hz, 1H), 7.52 (dd, J=7.6, 1.7 Hz, 1H), 7.23 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.71 (td, J=7.1, 3.3 Hz, 1H), 5.60 (s, 2H), 4.59-4.48 (m, 2H), 4.48-4.42 (m, 1H), 4.27 (dd, J=10.3, 3.4 Hz, 1H), 4.13 (dd, J=10.5, 8.3 Hz, 1H), 3.65 (t, J=9.4 Hz, 1H), 3.45 (ddd, J=9.9, 8.0, 6.2 Hz, 1H), 2.50 (s, 3H). Example 76: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(dimethylcarbamoyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(dimethylcarbamoyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (as a racemic mixture of enantiomers) was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.53 (d, J=1.5 Hz, 0H), 8.46 (d, J=1.5 Hz, 1H), 7.97-7.83 (m, 5H), 7.83-7.63 (m, 7H), 7.61-7.46 (m, 2H), 7.37 (dd, J=11.5, 6.1 Hz, 0H), 7.21 (dd, J=11.6, 6.1 Hz, 1H), 7.00 (dd, J=8.3, 2.0 Hz, 2H), 5.87 (d, J=8.6 Hz, 0H), 5.82 (s, 1H), 5.61 (d, J=2.3 Hz, 3H), 4.69 (t, J=8.6 Hz, 1H), 4.61-4.42 (m, 3H), 4.40-4.31 (m, 2H), 4.31-4.20 (m, 1H), 4.20-4.05 (m, 2H), 3.77 (td, J=8.8, 5.6 Hz, 1H), 3.57 (t, J=9.1 Hz, 1H), 3.02 (s, 1H), 2.78 (s, 3H), 2.72 (s, 3H). Example 77: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-fluorotetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-fluorotetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (as a racemic mixture of enantiomers) was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.29 (d, J=1.4 Hz, 1H), 7.96-7.89 (m, 2H), 7.89-7.82 (m, 1H), 7.82-7.71 (m, 3H), 7.68 (d, J=8.5 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.41 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.66 (dq, J=5.0, 2.3 Hz, 1H), 5.60 (s, 2H), 5.54 (q, J=5.3, 3.7 Hz, 1H), 4.55 (q, J=16.8 Hz, 2H), 4.46-4.24 (m, 3H), 4.08 (ddd, J=24.6, 11.4, 3.1 Hz, 1H). Example 78: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 78 was prepared in a manner similar to Procedure 22, with the following modifications: Methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (Intermediate I-104, 468 mg, 0.000746 mol) was taken up in Acetonitrile (3.80 mL) and lithium hydroxide (300 mmol/L, 3.73 mL, 0.00112 mol) was added. The mixture was heated to 100° C. After 10 min the mixture was removed from heat, then diluted with water and acidified to pH˜5 with 5% aqueous citric acid. The aqueous phase was extracted 3× with EtOAc. Combined organics were dried over MgSO4, filtered and concentrated in vacuo. Purification by preparative HPLC, MeCN/H2O gradient with 0.1% TFA yielded 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 78). 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.96-7.87 (m, 2H), 7.86-7.71 (m, 5H), 7.63 (d, J=8.5 Hz, 1H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.46 (dd, J=11.3, 6.2 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.60-4.49 (m, 2H), 4.49-4.33 (m, 2H), 3.83-3.68 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). Example 79: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,3aR,6aR)-hexahydrofuro[3,4-b]furan-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,3aR,6aR)-hexahydrofuro[3,4-b]furan-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.47 (d, J=1.4 Hz, 1H), 7.96-7.85 (m, 3H), 7.85-7.65 (m, 5H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.25 (td, J=5.2, 2.6 Hz, 1H), 5.09 (dd, J=7.0, 4.1 Hz, 1H), 4.60 (s, 2H), 4.23 (d, J=5.1 Hz, 2H), 3.96 (dd, J=12.2, 9.2 Hz, 2H), 3.58-3.48 (m, 2H), 3.18 (ddt, J=6.9, 4.5, 2.3 Hz, 1H). Example 80: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,3aR,6aS)-hexahydro-2H-furo[2,3-c]pyrrol-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,3aR,6aS)-hexahydro-2H-furo[2,3-c]pyrrol-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (d, J=1.4 Hz, 1H), 7.98-7.87 (m, 2H), 7.84 (dd, J=8.4, 1.5 Hz, 1H), 7.81-7.70 (m, 3H), 7.66 (d, J=8.5 Hz, 1H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.43 (dd, J=11.5, 6.1 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.61 (s, 3H), 4.86 (dd, J=11.1, 2.3 Hz, 1H), 4.76 (td, J=6.0, 2.5 Hz, 1H), 4.55-4.36 (m, 2H), 4.23 (dd, J=11.1, 7.3 Hz, 1H), 3.71-3.48 (m, 2H), 3.46-3.29 (m, 1H), 3.04 (s, 1H), 2.46-2.29 (m, 2H). Example 81: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(1,1-dioxidotetrahydrothiophen-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(1,1-dioxidotetrahydrothiophen-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.41 (d, J=1.3 Hz, 1H), 7.94-7.88 (m, 2H), 7.86 (dd, J=8.5, 1.4 Hz, 1H), 7.81-7.71 (m, 3H), 7.68 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.4, 1.6 Hz, 1H), 7.38 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.73 (ddd, J=18.8, 10.9, 7.8 Hz, 1H), 5.61 (s, 2H), 4.53 (s, 2H), 3.78 (dd, J=14.3, 10.6 Hz, 1H), 3.70-3.53 (m, 2H), 3.32 (td, J=13.1, 6.8 Hz, 1H), 2.86-2.60 (m, 2H). Example 82: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxaspiro[2.5]octan-8-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxaspiro[2.5]octan-8-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.28 (d, J=1.4 Hz, 1H), 7.96-7.86 (m, 2H), 7.81-7.70 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.6, 1.7 Hz, 1H), 7.48 (dd, J=11.4, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.27-5.19 (m, 1H), 4.55 (d, J=16.8 Hz, 1H), 4.37 (d, J=16.8 Hz, 1H), 4.22 (d, J=11.5 Hz, 1H), 4.00 (dd, J=11.8, 2.1 Hz, 1H), 3.79-3.68 (m, 1H), 3.19 (d, J=11.7 Hz, 1H), 2.89 (td, J=12.5, 11.5, 4.5 Hz, 1H), 2.05 (d, J=11.6 Hz, 1H), 0.61 (dt, J=10.1, 5.2 Hz, 1H), 0.40 (s, 1H), 0.22 (dt, J=10.2, 5.3 Hz, 1H). Example 83: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(3,3-dimethyltetrahydro-2H-pyran-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(3,3-dimethyltetrahydro-2H-pyran-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22. 1H NMR (400 MHz, DMSO-d6) δ 8.30 (s, 1H), 7.96-7.86 (m, 2H), 7.86-7.70 (m, 5H), 7.61 (d, J=8.4 Hz, 1H), 7.55 (dd, J=7.5, 1.7 Hz, 1H), 7.46 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 4.75 (dd, J=12.6, 3.9 Hz, 1H), 4.63 (d, J=16.9 Hz, 1H), 4.42 (d, J=16.9 Hz, 1H), 4.19-4.11 (m, 1H), 3.63-3.53 (m, 2H), 3.47 (d, J=11.4 Hz, 1H), 2.91 (dt, J=12.7, 5.8 Hz, 1H), 1.75 (d, J=10.8 Hz, 1H), 1.20 (s, 3H), 0.91 (s, 3H). Procedure 23: Example 84 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)-7-(2-pyridyl)benzimidazole-5-carboxylic acid (Example 84): A solution of methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-iodo-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (I-72, 7 mg, 0.01 mmol), bromo(2-pyridyl)zinc (11.2 mg, 0.05 mmol), and Pd(dppf)Cl2(0.7 mg, 0.001 mmol) in THE (0.4 mL) was degassed with argon, and stirred for 30 min at 90° C. Following this time, the mixture was purified directly by RP-HPLC (eluent: water/MeCN 0.1% TFA) to give Example 84. ES/MS: 650.3 (M+H+); 1H NMR (400 MHz, Acetonitrile-d3) δ 8.83 (s, 1H), 8.75 (d, J=6.4 Hz, 2H), 8.54 (s, 1H), 8.43 (s, 1H), 7.90-7.75 (m, 3H), 7.72 (t, J=7.4 Hz, 1H), 7.57 (t, J=8.1 Hz, 3H), 7.26 (dd, J=11.6, 6.0 Hz, 1H), 6.94 (d, J=8.3 Hz, 1H), 5.63 (s, 2H), 4.77-4.41 (m, 4H), 3.75 (t, J=5.0 Hz, 2H), 3.23 (s, 3H). Procedure 24: Example 85 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3R,4S)-4-methoxy-1-methoxycarbonyl-pyrrolidin-3-yl]benzimidazole-5-carboxylic acid (Example 85): The title compound was prepared according to procedure 4 substituting methyl chloroformate for acetic anhydride. ES/MS: 672.2 (M+H+);1H NMR (400 MHz, DMSO-d6) δ 8.26 (d, J=5.2 Hz, 1H), 7.99-7.87 (m, 2H), 7.83-7.71 (m, 4H), 7.60 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.34 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.49 (d, J=5.7 Hz, 1H), 4.62-4.43 (m, 2H), 4.15 (d, J=8.8 Hz, 2H), 3.88 (t, J=9.9 Hz, 1H), 3.69 (d, J=4.8 Hz, 3H), 3.10 (s, 3H) (note: 3 protons hidden by solvent). Example 86: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxy-1-(methoxycarbonyl)pyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxy-1-(methoxycarbonyl)pyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 24. 1H NMR (400 MHz, DMSO-d6) δ 8.12 (s, 1H), 7.99-7.87 (m, 2H), 7.84 (dd, J=8.4, 1.4 Hz, 1H), 7.79-7.72 (m, 3H), 7.67 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.38 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.33 (s, 1H), 4.63-4.37 (m, 3H), 3.68 (s, 4H), 3.36 (d, J=9.4 Hz, 1H), 3.17 (s, 3H). Example 87: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methoxy-1-(methoxycarbonyl)pyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methoxy-1-(methoxycarbonyl)pyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 24. 1H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J=5.9 Hz, 1H), 7.97-7.87 (m, 2H), 7.84-7.58 (m, 4H), 7.54 (dd, J=7.6, 1.7 Hz, 1H), 7.35 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.55-5.42 (m, 1H), 4.62-4.43 (m, 2H), 4.16 (d, J=9.1 Hz, 2H), 3.88 (t, J=9.9 Hz, 1H), 3.10 (s, 3H). Procedure 25: Example 88 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3R,4R)-4-methoxy-1-methylsulfonyl-pyrrolidin-3-yl]benzimidazole-5-carboxylic acid (Example 88): The title compound was prepared in a manner as described in Procedure 24 substituting methane sulfonyl chloride for methyl chloroformate. ES/MS: 692.2 (M+H+);1H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J=1.5 Hz, 1H), 7.96-7.83 (m, 3H), 7.80-7.73 (m, 3H), 7.68 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.4, 1.7 Hz, 1H), 7.38 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.34 (dt, J=9.6, 6.6 Hz, 1H), 4.61-4.38 (m, 3H), 4.06-3.92 (m, 2H), 3.30 (dd, J=10.1, 6.9 Hz, 1H), 3.18 (s, 3H), 3.13 (s, 3H). Example 89: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methoxy-1-(methylsulfonyl)pyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methoxy-1-(methylsulfonyl)pyrrolidin-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in procedure 25. 1H NMR (400 MHz, DMSO-d6) δ 8.41 (s, 1H), 7.97-7.87 (m, 2H), 7.82-7.71 (m, 4H), 7.60 (d, J=8.4 Hz, 1H), 7.56-7.52 (m, 1H), 7.34 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.53 (td, J=8.6, 5.1 Hz, 1H), 4.62-4.46 (m, 2H), 4.19 (q, J=4.6, 3.7 Hz, 1H), 4.05 (dd, J=10.3, 8.2 Hz, 1H), 3.84 (t, J=9.7 Hz, 1H), 3.10 (s, 3H), 3.08 (s, 3H). Procedure 26: Example 90 Methyl 2-[[2,5-difluoro-4-[6-[[4-fluoro-6-(1-methylpyrazol-4-yl)-3-pyridyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate: Methyl 2-[[4-[6-[(6-chloro-4-fluoro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (I-76) (120 mg, 0.19 mmol), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (78 mg, 0.38 mmol), 2 N of sodium carbonate (0.19 mL, 0.38 mmol) and (1,1′-bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (14 mg, 0.019 mmol) was suspended in dioxane (1 mL). The mixture was degassed by bubbling argon after which, the mixture was heated to 120° C. in the microwave reactor for 30 minutes. Upon completion the mixture was poured into water (5 mL) and extracted with EtOAc (2×15 mL). The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and purified by flash chromatography (Eluent: EtOAc/hexane) to give the desired product. ES/MS: 683.7 (M+H+). 2-[[2,5-difluoro-4-[6-[[4-fluoro-6-(1-methylpyrazol-4-yl)-3-pyridyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylic acid (Example 90): 1 mL of ACN and 0.3 mL of 1 N lithium hydroxide were added to a vial of methyl 2-[[2,5-difluoro-4-[6-[[4-fluoro-6-(1-methylpyrazol-4-yl)-3-pyridyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (81 mg, 0.12 mmol). The mixture was heated to 100° C. for 10 minutes. Upon completion a 5% citric acid solution in water was added (2 mL), the resulting mixture diluted with EtOAc (25 mL), washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and purified by RP-HPLC (eluent: water/MeCN*0.1% TFA) to give Example 90. ES/MS: 669.6 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.69 (d, J=10.4 Hz, 1H), 8.51 (s, 1H), 8.35 (s, 1H), 8.06 (s, 1H), 8.00-7.86 (m, 2H), 7.83 (dd, J=8.4, 1.5 Hz, 1H), 7.73-7.61 (m, 2H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.48 (dd, J=11.2, 6.3 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.55 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.65-4.51 (m, 2H), 4.49-4.36 (m, 2H), 3.89 (s, 3H), 3.79 (d, J=8.7 Hz, 1H), 3.74 (d, J=8.6 Hz, 1H), 1.34 (s, 3H), 0.62 (s, 3H). Example 91: (R)-2-(2,5-difluoro-4-(6-((4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(2,5-difluoro-4-(6-((4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 26. 1H NMR (400 MHz, DMSO-d6) δ 8.65 (s, 1H), 8.60 (s, 1H), 8.32 (s, 1H), 8.27 (d, J=1.5 Hz, 1H), 7.96-7.79 (m, 3H), 7.75 (s, 1H), 7.62 (d, J=8.4 Hz, 1H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.42 (dd, J=11.5, 6.1 Hz, 1H), 6.97 (d, J=8.3 Hz, 1H), 5.50 (s, 2H), 4.54 (dd, J=15.2, 3.2 Hz, 1H), 4.49 (s, 2H), 4.38 (dd, J=15.2, 8.8 Hz, 1H), 4.16 (s, 3H), 3.97 (s, 3H), 3.71 (ddd, J=9.3, 6.2, 3.2 Hz, 1H), 3.09 (s, 3H), 1.24 (d, J=6.1 Hz, 3H). Procedure 27: Example 92 Methyl 2-[[4-[6-[(6-chloro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate: To a solution of methyl 2-[[2,5-difluoro-4-(6-hydroxy-2-pyridyl)phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (30.0 mg, 0.061 mmol) and 5-(bromomethyl)-2-chloro-pyridine (13 mg, 0.061 mmol) in 11 mL of acetonitrile was added Cs2CO3(40 mg, 0.12 mmol). The solution was then heated to 50° C. for 30 minutes. Upon completion the solution was cooled to rt, filtered, then concentrated. The crude material was purified by normal phase chromatography (eluent: EtOAc/hexanes). 2-[[4-[6-[(6-chloro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylic acid (Example 92): To a methyl 2-[[4-[6-[(6-chloro-3-pyridyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(4,4-dimethyltetrahydrofuran-3-yl)benzimidazole-5-carboxylate (20.0 mg, 0.032 mmol) in 3 mL of acetonitrile and 1 mL of water was added aqueous LiOH (0.02 mL, 0.04 mmol, 2 M). The solution was then stirred at 80° C. for 1 hour. Upon completion the solution was cooled to rt, filtered through celite and concentrated. The crude material was purified by reverse phase chromatography (eluent ACN/water with 0.1% TFA added) to give Example 92. ES/MS m/z: 605.9 (M+H+); 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.51 (d, J=2.4 Hz, 1H), 8.17 (dd, J=8.6, 1.3 Hz, 1H), 8.05-7.88 (m, 2H), 7.88-7.72 (m, 2H), 7.58 (dd, J=7.5, 1.6 Hz, 1H), 7.48 (d, J=8.2 Hz, 1H), 7.40 (dd, J=11.1, 6.1 Hz, 1H), 6.94 (d, J=8.3 Hz, 1H), 5.55 (s, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.78-4.61 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). Example 93: 2-(4-(6-((5-chloropyrazin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((5-chloropyrazin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27. 1H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.66 (d, J=1.4 Hz, 1H), 8.59 (d, J=1.3 Hz, 1H), 8.20 (d, J=8.6 Hz, 1H), 7.94-7.72 (m, 3H), 7.60 (dd, J=7.5, 1.6 Hz, 1H), 7.41 (dd, J=11.2, 6.0 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.64 (s, 2H), 5.15 (d, J=6.6 Hz, 1H), 4.79-4.59 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). Procedure 28: Example 94 Methyl 2-[[4-[6-[[5-(difluoromethyl)pyrazin-2-yl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: A suspension of methyl 2-[[4-(6-chloro-2-pyridyl)-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (40.0 mg, 0.085 mmol), [5-(difluoromethyl)pyrazin-2-yl]methanol (17 mg, 0.11 mmol), Pd RockPhos G3 (11 mg, 0.013 mmol), and cesium carbonate (83 mg, 0.25 mmol) in toluene (1.5 mL) was degassed with Ar for 5 min, then heated at 100° C. overnight. Upon completion the mixture was diluted with EtOAc and washed with brine. The organic extract was dried over sodium sulfate and purified by flash chromatography (eluent: EtOAc/hexanes) to give the title compound. ES/MS: 596.2 (M+H+). 2-[[4-[6-[[5-(difluoromethyl)pyrazin-2-yl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (Example 94): A suspension of methyl 2-[[4-[6-[[5-(difluoromethyl)pyrazin-2-yl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (30.0 mg, 0.0209 mmol) and lithium hydroxide, monohydrate (300 mmol/L, 0.50 mL, 0.15 mmol) in CH3CN (0.5 mL) was heated at 105° C. for 6 min. Upon completion the reaction was quenched with 5% aqueous citric acid (1 mL) diluted with EtOAc (25 mL) and washed with brine (5 mL). The organic extract was dried over sodium sulfate, filtered and concentrated. The crude residue was purified by RP-HPLC (eluent: MeCN/H2O) to give Example 94. ES/MS: 582.2 (M+H+); 1H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.93 (d, J=1.4 Hz, 1H), 8.25 (d, J=1.5 Hz, 1H), 7.92 (t, J=7.9 Hz, 1H), 7.84 (dd, J=8.4, 1.5 Hz, 1H), 7.68-7.59 (m, 2H), 7.53 (dd, J=7.6, 1.6 Hz, 1H), 7.38 (dd, J=11.5, 6.1 Hz, 1H), 7.29-6.93 (m, 2H), 5.71 (s, 2H), 4.62 (t, J=5.1 Hz, 2H), 4.47 (s, 2H), 3.69 (t, J=5.1 Hz, 2H), 3.21 (s, 3H). Example 95: 2-(4-(6-((6-(1H-imidazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-(1H-imidazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 28. 1H NMR (400 MHz, DMSO-d6) δ 8.65 (d, J=2.2 Hz, 1H), 8.55 (s, 1H), 8.20 (d, J=1.4 Hz, 1H), 8.14 (dd, J=8.4, 2.3 Hz, 1H), 7.97 (s, 1H), 7.92-7.83 (m, 3H), 7.79 (dd, J=8.4, 1.5 Hz, 1H), 7.60 (d, J=8.5 Hz, 1H), 7.53 (d, J=7.1 Hz, 1H), 7.39 (dd, J=11.5, 6.1 Hz, 1H), 7.13 (s, 1H), 6.97 (d, J=8.3 Hz, 1H), 5.56 (s, 2H), 5.40 (s, 1H), 4.59 (d, J=5.6 Hz, 2H), 4.44 (s, 2H), 3.69 (t, J=5.0 Hz, 2H), 3.22 (s, 3H). Example 96: 2-(4-(6-((4-(1,1-difluoroethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-(1,1-difluoroethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared a manner as described in Procedure 28. 1H NMR (400 MHz, DMSO-d6) δ 12.84 (s, 1H), 8.32 (d, 1H), 7.93-7.84 (m, 2H), 7.79 (dd, 1H), 7.70-7.59 (m, 1H), 7.59 (d, 4H), 7.51 (dd, 1H), 7.43 (dd, 1H), 6.97 (dd, 1H), 5.53 (s, 2H), 4.67 (t, 2H), 3.70 (t, 2H), 3.21 (s, 3H), 2.49 (s, OH), 1.96 (t, 3H). Procedure 29: Example 97 Methyl 2-((7-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-1,3-dihydroisobenzofuran-4-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate: In a 8 mL glass vial, a suspension of methyl 2-[(7-bromo-1,3-dihydroisobenzofuran-4-yl)methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-45) (80.0 mg, 0.177 mmol), bis(pinacolato)diboron (48 mg, 0.19 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II); PdCl2(dppf) (10 mg, 0.014 mmol), and potassium acetate (38 mg, 0.39 mmol) in dioxane (1.5 mL) was degassed with Ar for 5 min. Upon completion the mixture was heated at 120° C. for 30 min in a microwave reactor. Following this time, the mixture was cooled to rt. Sodium carbonate (2.00 M, 0.18 mL, 0.36 mmol) was added and the mixture was stirred at rt for 5 min. Upon completion 4-[(6-bromo-2-pyridyl)oxymethyl]-3-fluoro-benzonitrile (I-3) (64 mg, 0.21 mmol) was added to the mixture followed by degassing the mixture for 5 min with argon and then heated at 120° C. for 20 min in a microwave reactor. Following this time, the mixture was diluted with EtOAc filtered through celite, concentrated and purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 593.3. 2-((7-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-1,3-dihydroisobenzofuran-4-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 97): A suspension of methyl 2-((7-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-1,3-dihydroisobenzofuran-4-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (41 mg, 0.069 mmol) and lithium hydroxide, monohydrate (1M, 0.2 mL, 0.2 mmol) in CH3CN (1.25 mL) in a 4 mL glass vial was heated at 100° C. for 5 min. Upon completion the mixture was diluted with acetonitrile/DMF (1:1, 1 mL). 2 drops of TFA was added and the mixture was then purified directly by RP-HPLC (eluent: MeCN/H2O) to obtain Example 97. ES/MS: 579.2; 1H NMR (400 MHz, DMSO) δ 8.26 (s, 1H), 7.98-7.92 (m, 1H), 7.91-7.81 (m, 3H), 7.78-7.68 (m, 2H), 7.65 (d, J=8.4 Hz, 1H), 7.59 (d, J=7.6 Hz, 1H), 7.27 (d, J=8.0 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 5.36 (s, 2H), 4.99 (s, 2H), 4.58 (t, J=5.3 Hz, 2H), 4.42 (s, 2H), 3.63 (t, J=5.1 Hz, 2H), 3.20 (s, 3H). Procedure 30: Example 98 Tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-[1-[2-[2-(2-methoxyethoxy)ethoxy]ethyl]pyrazol-4-yl]phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: 1-[2-[2-(2-methoxyethoxy)ethoxy]ethyl]-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (36 mg, 0.18 mmol), Pd(dppf)Cl2(12 mg, 0.037 mmol) and aqueous potassium carbonate (2.0M, 36 uL, 0.073 mmol) were added to a solution of tert-butyl 2-[[4-[6-[(4-bromo-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-18) (25 mg, 0.037 mmol) in 1,4-dioxane (2 mL). The mixture was bubbled with argon for 1 minute, then the reaction vessel sealed and heated to 100° C. for 10 minutes. Upon completion the mixture was concentrated directly, diluted with acetonitrile/water (9:1, 10 mL), passed through a C18 SPE column, concentrated and purified by RP-HPLC (eluent: MeCN/H2O) to obtain the desired product. ES/MS: 816.4 (M+H+). 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-[1-[2-[2-(2-methoxyethoxy)ethoxy]ethyl]pyrazol-4-yl]phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (Example 98): 1 mL TFA 1 mL TFA was added to a solution of tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-[1-[2-[2-(2-methoxyethoxy)ethoxy]ethyl]pyrazol-4-yl]phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (15 mg, 0.018 mmol) in DCM (2 mL). The resulting solution was stirred at rt for 1 hr. Upon completion the mixture was concentrated directly and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to give Example 98. ES/MS: 760.4 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.25 (d, 1H), 7.96 (s, 1H), 7.93-7.79 (m, 2H), 7.63 (d, 1H), 7.59-7.35 (m, 4H), 6.94 (d, 1H), 5.50 (s, 1H), 4.63 (d, OH), 4.62 (s, 1H), 4.48 (s, 1H), 4.26 (d, 1H), 3.79 (s, 1H), 3.48 (dt, 5H), 3.36 (q, 2H), 3.19 (dd, 4H) (note: multiple protons hidden under water peak). Example 99: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-11H-1,2,3-triazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-1,2,3-triazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, DMSO-d6) δ 8.29 (d, 1H), 7.99 (s, 1H), 7.92-7.82 (m, 3H), 7.73 (t, 1H), 7.65 (d, 1H), 7.62 (dd, 1H), 7.53 (dd, 1H), 7.50 (dd, 1H), 7.42 (dd, 1H), 6.98 (dd, 1H), 5.59 (s, 2H), 4.65 (t, 2H), 4.52 (s, 2H), 4.11 (s, 3H), 3.70 (t, 2H), 3.22 (s, 3H). Example 100: 2-(4-(6-((4-(1-(difluoromethyl)-1H-pyrazol-4-yl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-(1-(difluoromethyl)-1H-pyrazol-4-yl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, DMSO-d6) δ 12.87 (s, 1H), 8.82 (s, 1H), 8.35 (s, 1H), 8.29 (d, 1H), 7.95-7.82 (m, 4H), 7.72-7.54 (m, 5H), 7.52 (dd, 1H), 7.42 (dd, 1H), 6.96 (d, 1H), 5.52 (s, 2H), 4.65 (t, 2H), 4.52 (s, 2H), 3.70 (t, 2H), 3.22 (s, 3H). Example 101: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-pyrazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-pyrazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, DMSO-d6) δ 12.86 (s, 1H), 8.29 (d, 1H), 7.94-7.81 (m, 3H), 7.73-7.62 (m, 2H), 7.57-7.45 (m, 3H), 7.47-7.38 (m, 2H), 6.98 (d, 1H), 6.49 (d, 1H), 5.58 (s, 2H), 4.66 (t, 2H), 4.52 (s, 2H), 3.88 (s, 3H), 3.70 (t, 2H), 3.22 (s, 3H). Example 102: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-3-(trifluoromethyl)-1H-pyrazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-3-(trifluoromethyl)-1H-pyrazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, DMSO-d6) δ 8.27 (d, 1H), 7.94-7.80 (m, 3H), 7.73 (t, 1H), 7.67-7.37 (m, 5H), 7.03-6.95 (m, 2H), 5.59 (s, 2H), 4.64 (t, 2H), 4.50 (s, 2H), 3.70 (t, 2H), 3.21 (s, 3H). Example 103: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(pyridin-3-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(pyridin-3-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, DMSO-d6) δ 9.05 (d, 1H), 8.69 (dd, 1H), 8.35 (dt, 1H), 8.27 (d, 1H), 7.94-7.80 (m, 3H), 7.80-7.60 (m, 5H), 7.53 (dd, 1H), 7.42 (dd, 1H), 6.98 (d, 1H), 5.59 (s, 2H), 4.64 (t, 2H), 4.50 (s, 2H), 3.70 (t, 2H), 3.21 (s, 3H). Example 104: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(2-methylpyridin-3-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(2-methylpyridin-3-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described on Procedure 30.1H NMR (400 MHz, DMSO-d6) δ 8.74 (dd, 1H), 8.27 (d, 1H), 8.20 (d, 1H), 7.95-7.80 (m, 3H), 7.75 (dt, 2H), 7.63 (d, 1H), 7.57-7.32 (m, 4H), 6.99 (d, 1H), 5.60 (s, 2H), 4.64 (t, 2H), 4.50 (s, 2H), 3.70 (t, 2H), 3.22 (s, 3H), 2.59 (s, 3H). Example 105: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(6-methylpyridin-3-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(6-methylpyridin-3-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, DMSO-d6) δ 12.88 (s, 1H), 9.03 (d, 1H), 8.51 (dd, 1H), 8.27 (d, 1H), 7.94-7.65 (m, 7H), 7.63 (d, 1H), 7.53 (dd, 1H), 7.42 (dd, 1H), 6.97 (d, 1H), 5.59 (s, 2H), 4.64 (t, 2H), 3.70 (t, 2H), 3.21 (s, 3H), 2.66 (s, 3H). Example 106: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-(2-methoxyethyl)-1H-pyrazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-(2-methoxyethyl)-1H-pyrazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, DMSO-d6) δ 8.28-8.21 (m, 4H), 7.96 (s, 2H), 7.92-7.79 (m, 6H), 7.63 (d, 2H), 7.57-7.47 (m, 7H), 7.45 (d, 1H), 7.43 (s, 1H), 6.94 (d, 2H), 5.49 (s, 4H), 4.63 (t, 4H), 4.49 (s, 4H), 4.27 (t, 4H), 3.70 (dt, 9H), 3.24 (s, 6H), 3.22 (s, 6H). Example 107: 2-(2,5-difluoro-4-(6-((6-(1-methyl-11H-1,2,3-triazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((6-(1-methyl-1H-1,2,3-triazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, Acetonitrile-d3) δ 8.85 (d, 1H), 8.34 (s, 1H), 8.09-7.98 (m, 3H), 7.93-7.70 (m, 4H), 7.56 (dd, 1H), 7.27 (dd, 1H), 6.96-6.89 (m, 1H), 5.60 (s, 2H), 4.57 (dd, 4H), 4.35 (s, 3H), 3.75 (t, 2H), 3.26 (s, 3H). Example 108: 2-(2,5-difluoro-4-(6-((6-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((6-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, Acetonitrile-d3) δ 8.80 (d, 1H), 8.43-8.30 (m, 3H), 8.16 (d, 1H), 8.08 (dd, 1H), 7.98-7.77 (m, 3H), 7.76 (d, 1H), 7.54 (dd, 1H), 7.29 (dd, 1H), 6.92 (d, 1H), 5.60 (s, 2H), 4.65 (d, 1H), 4.63 (s, 3H), 3.95 (s, 3H), 3.78 (t, 2H), 3.28 (s, 3H). Example 109: 2-(4-(6-((6-(1-(difluoromethyl)-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-(1-(difluoromethyl)-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, Acetonitrile-d3) δ 8.79 (d, 1H), 8.63 (s, 1H), 8.45-8.40 (m, 1H), 8.31 (s, 1H), 8.11 (ddd, 2H), 7.97-7.87 (m, 1H), 7.87-7.76 (m, 3H), 7.62-7.52 (m, 1H), 7.45 (s, OH), 7.35-7.26 (m, 1H), 6.92 (d, 1H), 5.59 (s, 2H), 4.65 (t, 2H), 3.78 (t, 2H), 3.27 (s, 3H), 1.97-1.86 (m, 1H). Example 110: 2-(2,5-difluoro-4-(6-((6-(1-methyl-1H-pyrazol-5-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((6-(1-methyl-1H-pyrazol-5-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 30.1H NMR (400 MHz, Acetonitrile-d3) δ 8.81 (d, 1H), 8.39 (s, 1H), 8.08 (dd, 1H), 7.97 (dd, 1H), 7.91 (dd, 1H), 7.85-7.75 (m, 2H), 7.72 (d, 1H), 7.56 (dd, 1H), 7.47 (d, 1H), 7.30 (dd, 1H), 6.93 (d, 1H), 6.69 (d, 1H), 5.58 (s, 2H), 4.62 (s, 3H), 4.62 (d, 1H), 4.16 (s, 3H), 3.76 (t, 2H), 3.26 (s, 3H). Procedure 31: Example 111 Tert-butyl 2-[[4-[6-[(4-ethynyl-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: Cuprous iodide (4 mg, 0.021 mmol), trans-Dichlorobis(triphenylphosphine)palladium (7.7 mg, 0.011 mmol), trimethylsilyl acetylene (11 uL, 0.22 mmol) and diisopropylethylamine (0.15 mL, 0.88 mmol) were added to a solution of tert-butyl 2-[[4-[6-[(4-bromo-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-18) (30 mg, 0.044 mmol) in DMF (2 mL). The resulting solution was degassed by bubbling argon for 1 min, sealed and heated to 100° C. for 3 hours. Upon completion the mixture was poured into water (5 mL) and extracted with EtOAc (2×15 mL). The organic layers were combined, washed with brine (5 mL), dried over MgSO4, filtered, concentrated, and the crude residue was dissolved in THF (2 mL) and TBAF (1.0M in THF, 0.35 mL, 0.35 mmol) was added. After stirring at rt for 10 min the mixture was diluted with EtOAc (25 mL), washed with water (5 mL) and brine (5 mL). The organic layer was dried over MgSO4, filtered, concentrated and purified by flash chromatography (Eluent: EtOAc/hexane) to give the desired product. ES/MS: 628.3 (M+H+). 2-[[4-[6-[(4-ethynyl-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (Example 111): 1 mL TFA was added to a solution of tert-butyl 2-[[4-[6-[(4-ethynyl-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (27 mg, 0.043 mmol) in DCM (2 mL). The resulting solution stirred at rt for 1 hr. Upon completion the mixture was concentrated directly and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide Example 111. ES/MS: 572.4 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.28 (d, J=1.4 Hz, 1H), 7.93-7.84 (m, 2H), 7.81 (dd, J=10.5, 6.5 Hz, 1H), 7.65 (d, J=8.4 Hz, 1H), 7.57 (t, J=7.8 Hz, 1H), 7.52 (dd, J=7.5, 1.7 Hz, 1H), 7.41 (ddd, J=11.4, 4.4, 2.8 Hz, 2H), 7.35 (dd, J=7.8, 1.6 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.53 (s, 2H), 4.65 (t, J=5.1 Hz, 2H), 4.51 (s, 2H), 4.34 (s, 1H), 3.70 (t, J=5.0 Hz, 2H), 3.22 (s, 3H). Procedure 32: Example 112 Tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(2-methyl-1,2,4-triazol-3-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: In a 8 mL glass vial, a suspension of tert-butyl 2-[[4-[6-[(4-bromo-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-18) (140 mg, 0.21 mmol), Bis(pinacolato)diboron (68 mg, 0.27 mmol), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II); PdCl2(dppf) (23 mg, 0.031 mmol), and potassium propionate (69 mg, 0.62 mmol) in dioxane (1.5 mL) was degassed with Ar for 1 min. Upon completion the mixture was heated at 110° C. for 45 min. Following this time, the mixture was cooled to rt, followed by the addition of sodium carbonate (2.00 M, 0.21 mL, 0.41 mmol) and stirred at rt for 5 min. Upon completion 5-bromo-1-methyl-1,2,4-triazole (50 mg, 0.31 mmol) was added to the mixture and then the mixture was degassed for 5 min with argon, followed by heating at 90° C. for 45 min. Following this time, the mixture was diluted with EtOAc filtered through celite, concentrated and purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(2-methyl-1,2,4-triazol-3-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (Example 112): 1.6 mL TFA was added to a solution of tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(2-methyl-1,2,4-triazol-3-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (112 mg, 0.16 mmol) in DCE (4 mL). The resulting solution stirred at 60° C. for 1 hr. Upon completion the mixture was concentrated directly and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide Example 112. ES/MS: 629.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.31 (d, J=1.4 Hz, 1H), 8.03 (s, 1H), 7.92-7.82 (m, 3H), 7.79-7.61 (m, 4H), 7.54 (dd, J=7.6, 1.7 Hz, 1H), 7.43 (dd, J=11.5, 6.1 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 4.67 (t, J=5.1 Hz, 2H), 4.54 (s, 2H), 4.00 (s, 3H), 3.70 (d, J=5.0 Hz, 2H), 3.21 (s, 3H). Procedure 33: Example 113 Tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(3-methylimidazol-4-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: In a 8 mL glass vial, a suspension of tert-butyl 2-[[4-[6-[(4-bromo-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-18) (90 mg, 0.13 mmol), tributyl-(3-methylimidazol-4-yl)stannane (73 mg, 0.20), tetrakis(triphenylphosphine) palladium (23 mg, 0.020 mmol), and lithium chloride (17 mg, 0.40 mmol) in dioxane (2 mL) was degassed with Ar for 1 min. Following the degassing the mixture was heated at 100° C. for 16 hrs. Upon completion the mixture was cooled to rt, concentrated directly and the crude residue purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 684.2 (M+H+). 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(3-methylimidazol-4-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (Example 113): 1.6 mL TFA was added to a solution of tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(3-methylimidazol-4-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (112 mg, 0.16 mmol) in DCE (4 mL). The resulting solution stirred at 60° C. for 1 hr. Upon completion the mixture was concentrated directly and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide Example 113. ES/MS: 628.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 9.18 (d, J=1.4 Hz, 1H), 8.25 (d, J=1.4 Hz, 1H), 7.97-7.80 (m, 4H), 7.76 (t, J=7.8 Hz, 1H), 7.65-7.57 (m, 2H), 7.52 (ddd, J=15.7, 7.7, 1.7 Hz, 2H), 7.41 (dd, J=11.5, 6.0 Hz, 1H), 6.98 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 4.63 (t, J=5.1 Hz, 2H), 4.48 (s, 2H), 3.88 (s, 3H), 3.70 (s, 2H), 3.22 (s, 3H). Procedure 34: Example 114 Tert-butyl 2-[[4-[6-[[4-[1-(2,2-difluoroethyl)pyrazol-4-yl]-2-fluoro-phenyl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate: Tert-butyl 2-[[2,5-difluoro-4-[6-[[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methoxy]-2-pyridyl]phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (I-88) (23 mg, 0.032 mmol), 4-bromo-1-(2,2-difluoroethyl)pyrazole (6.7 mg, 0.032 mmol), 1,1′-Bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (2.6 mg, 0.0032 mmol), sodium carbonate (6.7 mg, 0.063 mmol) 1,4-dioxane (1 mL), and water (0.5 mL) were combined in a glass vial, degassed via bubbling argon for 1 min, and then the mixture heated to 100° C. for 10 min in a microwave reactor. Upon completion the mixture was concentrated directly, diluted with 9:1 acetonitrile:water, passed through a C18 SPE column, concentrated and carried forward without further purification. 2-[[4-[6-[[4-[1-(2,2-difluoroethyl)pyrazol-4-yl]-2-fluoro-phenyl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylic acid (Example 114): 0.25 mL TFA was added to a solution of tert-butyl 2-[[4-[6-[[4-[1-(2,2-difluoroethyl)pyrazol-4-yl]-2-fluoro-phenyl]methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (23 mg, 0.031 mmol) in DCM (2 mL). The resulting solution stirred at 40° C. for 1 hr. The mixture was concentrated directly and purified by RP-HPLC (eluent: water/MeCN 0.1% TFA). The combined fractions were then frozen and placed on a lyophilizer to provide Example 114. ES/MS: 678.2 (M+H+). 1H NMR (400 MHz, Acetonitrile-d3) δ 8.28 (s, 1H), 8.00 (s, 1H), 7.96 (s, 1H), 7.93-7.88 (m, 2H), 7.80 (t, 1H), 7.69 (d, 1H), 7.56 (t, 3H), 7.42-7.35 (m, 2H), 7.23 (dd, 1H), 6.87 (d, 1H), 6.24 (tt, 1H), 5.56 (s, 2H), 4.55-4.51 (m, 2H), 4.49 (s, 2H), 3.73 (t, 2H), 3.25 (s, 3H). Procedure 35: Example 115 Methyl 2-[[4-[6-[(4-chloro-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-103). A mixture of methyl 2-[(4-bromo-2,5-difluoro-phenyl)methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-82, 800 mg, 1.67 mmol, 1.0 equivalent), PdCl2(dppf) (186 mg, 0.25 mmol, 0.15 equivalent), potassium propionate (560 mg, 5.01 mmol, 3.0 equivalent), and bis(pinacolato)diboron (510 mg, 2.00 mol, 1.2 equivalent) was mixed with 1,4-dioxane (10 mL) and the resulting mixture was purged with argon for 2 min. The mixture was sealed and heated to 120° C. by microwave followed by stirring for 1 hr. After cooling down to room temperature, 2-bromo-6-[(4-chloro-2-fluoro-phenyl)methoxy]pyridine (Intermediate I-102, 580 mg, 1.84 mmol, 1.1 equivalent), PdCl2(dppf) (62 mg, 0.0834 mmol, 0.05 equivalent), 2 M aqueous Na2CO3(2.0 mL, 4.17 mmol, 2.5 equivalent) were added, respectively. The resulting mixture was heated to 100° C. under argon and stirred for 3 hrs. before cooling to rt and filtered through a plug of Celite and MgSO4. The filtrate was concentrated and purified by column chromatography (silica gel, EtOAc/hexane gradient) to yield the title compound. ES/MS m/z: 635.6 (M+H)+. 2-[[4-[6-[(4-chloro-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylic acid (Example 115): 2-[[4-[6-[(4-chloro-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylic acid was prepared in a manner similar to Procedure 22, with the following modifications: LiOH (1.0 M in H2O, 3.5 mL, 3.5 mmol) was added to a solution of methyl 2-[[4-[6-[(4-chloro-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-103, 550 mg, 0.865 mmol) in acetonitrile (10 mL), and the resulting solution was heated to reflux for 8 min. The reaction was quenched with 1 mL acetic acid and diluted with EtOAc (100 mL). The organic extracts were washed with water (4×50 mL), dried over MgSO4, filtered, concentrated in vacuo. The crude material was purified by reverse phase chromatography (MeCN/water gradient with 0.1% TFA added). The product containing fractions were combined and concentrated to give Example 115. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.94-7.76 (m, 3H), 7.62 (dd, J=12.6, 8.3 Hz, 2H), 7.57-7.42 (m, 3H), 7.33 (dd, J=8.2, 2.0 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.54 (dd, J=14.1, 2.9 Hz, 2H), 4.49-4.33 (m, 2H), 3.82-3.72 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 622.2 (M+H)+. Example 116: 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluorobenzimidazole-5-carboxylic acid 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluorobenzimidazole-5-carboxylic acid was prepared in a manner similar to Procedure 22, with the following modifications: To a solution of methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-5-fluoro-2-pyridyl]-2,5-difluorophenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]-7-fluorobenzimidazole-5-carboxylate (Intermediate I-110, 437 mg, 0.66 mmol) in 15 mL of acetonitrile and 5 mL of water was added aqueous LiOH (2.0 mL, 2.0 mmol, 2 M). The solution was then stirred at rt overnight. The following day, trifluoroacetic acid (0.15 mL, 2.0 mmol) was added, then concentrated into 1 mL of DMF. The crude material was purified by reverse phase preparative HPLC (ACN/water gradient with 0.1% TFA added) to give Example 116. ES/MS m/z: 648.6 (M+H+); 1H NMR (400 MHz, DMSO) δ 13.12 (s, 1H), 8.79 (d, J=5.2 Hz, 1H), 8.36 (s, 1H), 7.96 (dd, J=10.2, 6.2 Hz, 1H), 7.68-7.60 (m, 2H), 7.60-7.51 (m, 2H), 7.51 (d, J=2.1 Hz, 1H), 7.35 (dd, J=8.2, 2.0 Hz, 1H), 5.54 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.58 (d, J=17.0 Hz, 1H), 4.53 (d, J=11.8 Hz, 1H), 4.48-4.38 (m, 2H), 3.76 (d, J=16.5 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H). Example 117: (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner similar to Procedure 22, with the following modifications: To a solution of methyl (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-111, 37 mg, 0.06 mmol) in 3 mL of acetonitrile and 1 mL of water was added aqueous LiOH (0.04 mL, 0.08 mmol, 2 M). The solution was then stirred at rt overnight. The following day, a drop of trifluoroacetic acid was added, then concentrated into 1 mL of DMF. The crude material was purified by reverse-phase preparative HPLC (ACN/water gradient with 0.1% TFA added) to yield Example 117. ES/MS m/z: 642.0 (M+H+);1H NMR (400 MHz, DMSO) δ 8.79 (d, J=5.2 Hz, 1H), 8.36 (s, 1H), 7.96 (dd, J=10.2, 6.2 Hz, 1H), 7.68-7.48 (m, 5H), 7.35 (dd, J=8.3, 2.1 Hz, 1H), 5.54 (s, 2H), 5.04 (d, J=6.5 Hz, 1H), 4.59 (d, J=17.0 Hz, 1H), 4.53 (d, J=11.8 Hz, 1H), 4.48-4.38 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H). Example 118: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner similar to Procedure 22 with the following modifications: A solution of methyl (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-113) and LiOH (2N, 0.1 mL, 0.2 mmol) in ACN (0.5 mL) and H2O (0.4 mL) was heated at 100° C. for 6 min. The mixture was neutralized with acetic acid (0.01 mL, 0.2 mmol), diluted with DMSO, filtered, and purified (RP HPLC, 0-100% ACN in H2O, 0.1% TFA) to give Example 118. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.3 Hz, 1H), 8.82 (s, 1H), 8.47 (s, 1H), 8.30 (s, 1H), 8.02 (d, J=1.3 Hz, 1H), 7.97-7.84 (m, 2H), 7.79 (dd, J=8.5, 1.5 Hz, 1H), 7.61 (d, J=8.5 Hz, 1H), 7.56 (d, J=7.2 Hz, 1H), 7.46 (dd, J=11.1, 6.4 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.67 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.57-4.48 (m, 2H), 4.46-4.33 (m, 2H), 3.79-3.70 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 672.1 (M+H)+. Example 119: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-benzimidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-benzimidazole-6-carboxylic acid was prepared in a manner similar to Procedure 22, with the following modifications: To methyl 2-[[4-[6-[(4-cyanophenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[(3S)-4,4-dimethyltetrahydrofuran-3-yl]benzimidazole-5-carboxylate (Intermediate I-115, 52 mg, 0.0854 mmol) dissolved in acetonitrile (0.8 mL) was added lithium hydroxide monohydrate (4.3 mg, 0.103 mmol), and water (0.4 mL). The resulting mixture was stirred for 1 h at 90° C. The mixture was diluted with EtOAc and washed with brine, dried, filtered and concentrated in vacuo. Purification by reverse-phase preparative HPLC (CH3CN/water gradient, with 0.1% TFA) yielded Example 119. 1H NMR (400 MHz, Methanol-d4) δ 8.87 (s, 1H), 8.16 (dd, J=8.6, 1.4 Hz, 1H), 7.91-7.72 (m, 5H), 7.66 (d, J=8.4 Hz, 2H), 7.58 (dd, J=7.5, 1.6 Hz, 1H), 7.37 (dd, J=11.2, 6.1 Hz, 1H), 6.97 (d, J=8.2 Hz, 1H), 5.59 (s, 2H), 5.12 (d, J=6.6 Hz, 1H), 4.76-4.60 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 595.6 (M+H)+. Example 120: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-benzimidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-benzimidazole-6-carboxylic acid was prepared in a manner similar to Procedure 22, with the following modifications: Methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-117, 400 mg, 0.62 mmol) was dissolved in acetonitrile (3 mL), after which 1.0 mL of 2 N LiOH was added and the resulting mixture stirred at 80° C. for 40 min. To the mixture was added 0.5 mL acetic acid. The solvent was removed in vacuo, and the crude product dissolved in 4 mL of DMF and purified by reverse-phase preparative HPLC (eluent: water/MeCN 0.1% TFA) to yield Example 120. ES/MS m/z: 631.2 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.11 (dd, J=8.6, 1.4 Hz, 1H), 7.89 (t, J=7.9 Hz, 1H), 7.81-7.66 (m, 3H), 7.66-7.55 (m, 3H), 7.01 (d, J=8.3 Hz, 1H), 5.65 (s, 2H), 5.17 (d, J=6.6 Hz, 1H), 4.84-4.68 (m, 2H), 4.68-4.49 (m, 2H), 4.00 (d, J=8.8 Hz, 1H), 3.87 (d, J=8.8 Hz, 1H), 1.48 (s, 3H), 0.82 (s, 3H). Example 121: (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 121) was prepared in a manner similar to Procedure 22, with the following modifications: To a mixture of methyl (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-118, 50.0 mg, 0.079 mmol) in acetonitrile (1.00 mL) and 0.3 mL of 2 N lithium hydroxide was added and the resulting mixture stirred at 80° C. for 35 min. To the mixture was added 0.3 mL of acetic acid. The solvent was removed in vacuo and the residue was dissolved in 1 mL of DMF and purified by reverse-phase preparative HPLC (eluent: water/MeCN 0.1% TFA) to yield Example 121. ES/MS m/z: 623.5 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 1H), 8.72 (d, J=5.2 Hz, 1H), 8.14 (dd, J=8.6, 1.4 Hz, 1H), 8.06 (dd, J=10.4, 6.1 Hz, 1H), 7.75 (d, J=8.6 Hz, 1H), 7.68 (dd, J=5.2, 1.6 Hz, 1H), 7.60 (t, J=8.1 Hz, 1H), 7.46 (dd, J=11.2, 5.9 Hz, 1H), 7.32-7.22 (m, 2H), 5.61 (s, 2H), 5.12 (d, J=6.6 Hz, 1H), 4.81-4.58 (m, 3H), 4.53 (dd, J=11.5, 6.8 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H). Example 122: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner similar to Procedure 22, with the following modifications: To a mixture of methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-120, 167.0 mg, 0.25 mmol) in acetonitrile (3.00 mL) and 1 mL of 2 N lithium hydroxide was added and the resulting mixture stirred at 80° C. for 35 min. To the mixture was added 0.5 mL of acetic acid. Solvent was removed in vacuo and the resulting residue was dissolved in 1 mL of DMF and purified by reverse-phase preparative HPLC (eluent: water/MeCN 0.1% TFA) to yield Example 122. ES/MS m/z: 649.7 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.57 (s, 1H), 7.86 (t, J=7.9 Hz, 1H), 7.74 (t, J=7.5 Hz, 1H), 7.69-7.50 (m, 5H), 6.98 (d, J=8.2 Hz, 1H), 5.64 (s, 2H), 5.10 (d, J=6.6 Hz, 1H), 4.75-4.61 (m, 2H), 4.55 (dd, J=11.4, 6.8 Hz, 1H), 4.45 (d, J=17.2 Hz, 1H), 3.95 (d, J=8.8 Hz, 1H), 3.84 (d, J=8.8 Hz, 1H), 1.46 (s, 3H), 0.78 (s, 3H). Examples 123 and 124: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner similar to Procedure 20, with the following modifications: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (Example 122) as a mixture of 2 stereoisomers was separated by preparative chiral SFC (AD-H column with MeOH cosolvent) to give two distinct stereoisomers. (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (Example 123): Earlier eluting isomer was validated with X-ray Crystallography). ES/MS m/z: 649.6 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.56 (s, 1H), 7.87 (t, J=7.9 Hz, 1H), 7.75 (t, J=7.5 Hz, 1H), 7.69-7.53 (m, 5H), 6.98 (d, J=8.3 Hz, 1H), 5.64 (s, 2H), 5.10 (d, J=6.6 Hz, 1H), 4.78-4.60 (m, 2H), 4.55 (dd, J=11.3, 6.8 Hz, 1H), 4.44 (d, J=17.1 Hz, 1H), 3.95 (d, J=8.8 Hz, 1H), 3.84 (d, J=8.8 Hz, 1H), 1.46 (s, 3H), 0.78 (s, 3H). (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (Example 124): Later eluting isomer ES/MS m/z: 649.5 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.56 (s, 1H), 7.87 (t, J=7.9 Hz, 1H), 7.75 (t, J=7.5 Hz, 1H), 7.69-7.53 (m, 5H), 6.98 (d, J=8.3 Hz, 1H), 5.64 (s, 2H), 5.10 (d, J=6.6 Hz, 1H), 4.78-4.60 (m, 2H), 4.55 (dd, J=11.3, 6.8 Hz, 1H), 4.44 (d, J=17.1 Hz, 1H), 3.95 (d, J=8.8 Hz, 1H), 3.84 (d, J=8.8 Hz, 1H), 1.46 (s, 3H), 0.78 (s, 3H). Example 125: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner similar to Procedure 1 (step 2), with the following modifications: To a solution of tert-butyl 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (Intermediate I-123, 37 mg, 0.0557 mmol) in dichloromethane (1 mL) was added trifluoroacetic acid (0.11 mL, 0.667 mmol). The mixture was stirred at rt for 3 hrs., then concentrated in vacuo. Purification by reverse-phase preparative HPLC (eluent: water/MeCN 0.1% TFA) gave Example 125. 1H NMR (400 MHz, Chloroform-d) δ 8.03 (dd, J=8.7, 6.6 Hz, 1H), 7.82-7.63 (m, 3H), 7.60-7.37 (m, 4H), 7.18 (dd, J=11.1, 6.0 Hz, 1H), 5.66 (s, 2H), 5.32 (s, 1H), 4.64 (d, J=6.9 Hz, 4H), 3.83 (t, J=4.8 Hz, 2H), 3.34 (s, 3H). ES/MS m/z: 609.5 (M+H+). Example 126: (S)-4-chloro-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-4-chloro-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1037. 1H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 7.95-7.83 (m, 2H), 7.81 (d, J=1.3 Hz, 1H), 7.61 (t, J=8.2 Hz, 1H), 7.54 (dd, J=7.6, 1.6 Hz, 1H), 7.47 (ddd, J=17.2, 10.7, 4.1 Hz, 2H), 7.33 (dd, J=8.2, 2.0 Hz, 1H), 6.95 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.69-4.25 (m, 4H), 3.88-3.67 (m, 2H), 1.32 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 656.0 (M+H+). Example 127: (S)-4-chloro-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-4-chloro-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1037. 1H NMR (400 MHz, DMSO-d6) δ 8.46 (s, 1H), 7.94 (dd, J=10.0, 1.4 Hz, 1H), 7.88 (dd, J=10.2, 8.2 Hz, 1H), 7.84-7.67 (m, 4H), 7.63-7.49 (m, 1H), 7.45 (dd, J=11.4, 6.2 Hz, 1H), 5.70 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.61-4.34 (m, 4H), 3.79-3.72 (m, 2H), 3.17 (s, 1H), 1.32 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 665.0 (M+H+). Example 129: (S)-4-chloro-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-4-chloro-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-94 and I-1037. 1H NMR (400 MHz, DMSO-d6) δ 13.12 (s, 1H), 8.79 (d, J=5.2 Hz, 1H), 8.47 (s, 1H), 7.95 (dt, J=10.5, 3.2 Hz, 2H), 7.83-7.72 (m, 3H), 7.66 (dd, J=5.2, 1.7 Hz, 1H), 7.55 (dd, J=11.5, 5.9 Hz, 1H), 5.64 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.66-4.35 (m, 5H), 3.75 (q, J=8.7 Hz, 2H), 1.32 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 648.0 (M+H+). Example 130: (S)-4-chloro-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid S)-4-chloro-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1037. 1H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 8.02-7.84 (m, 2H), 7.86-7.79 (m, 2H), 7.79-7.70 (m, 2H), 7.55 (dd, J=7.6, 1.6 Hz, 1H), 7.45 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.70-4.28 (m, 4H), 3.88-3.56 (m, 3H), 1.32 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 647.0 (M+H+). Example 131: (S)-2-((6-((4-chloro-2-fluorobenzyl)oxy)-5,5′-difluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((6-((4-chloro-2-fluorobenzyl)oxy)-5,5′-difluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1036 and I-1020. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 8.27 (d, J=7.0 Hz, 1H), 7.90 (dd, J=10.2, 8.2 Hz, 1H), 7.67-7.48 (m, 4H), 7.35 (dd, J=8.3, 2.0 Hz, 1H), 6.98 (d, J=7.6 Hz, 1H), 5.64-5.35 (m, 4H), 5.14 (d, J=6.5 Hz, 1H), 4.54 (d, J=11.3 Hz, 1H), 4.43 (dd, J=11.3, 6.7 Hz, 1H), 3.93-3.69 (m, 2H), 1.38 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 657.0 (M+H+). Example 132: (S)-2-((6-((4-cyano-2-fluorobenzyl)oxy)-5,5′-difluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((6-((4-cyano-2-fluorobenzyl)oxy)-5,5′-difluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1020. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 8.27 (d, J=7.0 Hz, 1H), 8.03-7.85 (m, 2H), 7.85-7.69 (m, 2H), 7.62-7.57 (m, 1H), 7.55 (d, J=11.2 Hz, 1H), 6.93 (d, J=7.6 Hz, 1H), 5.68 (s, 2H), 5.61-5.33 (m, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.61-4.45 (m, 1H), 4.45-4.28 (m, 1H), 3.89-3.68 (m, 2H), 1.38 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 648.0 (M+H+). Example 133: (S)-2-(2-chloro-4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1031. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.92-7.82 (m, 3H), 7.64 (d, J=8.0 Hz, 2H), 7.51 (d, J=11.2 Hz, 1H), 7.43 (s, 1H), 7.33 (s, 1H), 7.25 (d, J=7.3 Hz, 1H), 6.98 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.62-4.44 (m, 2H), 4.44-4.27 (m, 2H), 3.91-3.64 (m, 2H), 2.23 (s, 3H), 1.33 (s, 3H). ES/MS m/z: 625.3 (M+H+). Example 134: (S)-2-(2-chloro-4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1030. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.92-7.83 (m, 4H), 7.81 (dd, J=8.5, 1.5 Hz, 1H), 7.63 (t, J=8.2 Hz, 3H), 7.42 (s, 1H), 7.33 (s, 1H), 7.25 (d, J=7.3 Hz, 1H), 6.98 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.66-4.46 (m, 2H), 4.46-4.27 (m, 2H), 2.23 (s, 3H), 1.33 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 607.5 (M+H+). Example 135: (S)-2-(2-chloro-4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1247 and I-1030. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.01-7.83 (m, 2H), 7.83-7.73 (m, 1H), 7.66 (d, J=8.1 Hz, 2H), 7.61 (d, J=8.5 Hz, 1H), 7.43 (s, 1H), 7.34 (s, 1H), 7.29 (dd, J=8.1, 2.8 Hz, 1H), 5.59 (s, 2H), 5.00 (d, J=6.7 Hz, 1H), 4.61-4.48 (m, 2H), 4.48-4.27 (m, 2H), 3.80 (d, J=8.6 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 2.23 (s, 2H), 1.33 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 624.6 (M+H+). Example 136: (S)-2-(2-chloro-4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1247 and I-1031. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.95-7.78 (m, 3H), 7.66 (d, J=8.1 Hz, 2H), 7.51 (d, J=11.2 Hz, 1H), 7.43 (s, 1H), 7.34 (s, 1H), 7.29 (dd, J=8.1, 2.8 Hz, 1H), 5.59 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.64-4.45 (m, 2H), 4.45-4.26 (m, 2H), 3.88-3.69 (m, 2H), 2.23 (s, 3H), 1.33 (s, 4H), 0.65 (s, 3H). ES/MS m/z: 642.6 (M+H+). Example 137: (S)-2-((6-((4-chloro-2-fluorobenzyl)oxy)-5′-fluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((6-((4-chloro-2-fluorobenzyl)oxy)-5′-fluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1020. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 8.26 (d, J=6.9 Hz, 1H), 7.99-7.88 (m, 1H), 7.61 (t, J=8.2 Hz, 1H), 7.57-7.51 (m, 2H), 7.49 (dd, J=10.0, 2.1 Hz, 1H), 7.33 (dd, J=8.2, 2.0 Hz, 1H), 7.05 (d, J=8.3 Hz, 1H), 7.00 (d, J=7.5 Hz, 1H), 5.64-5.40 (m, 4H), 5.14 (d, J=6.6 Hz, 1H), 4.54 (d, J=11.3 Hz, 1H), 4.43 (dd, J=11.3, 6.8 Hz, 1H), 3.79-3.67 (m, 2H), 1.38 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 638.6 (M+H+). Example 138: (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1031. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.97-7.89 (m, 1H), 7.85 (dd, J=10.5, 8.1 Hz, 1H), 7.74 (dd, J=3.6, 1.9 Hz, 2H), 7.51 (dd, J=11.3, 1.2 Hz, 1H), 7.45 (s, 1H), 7.38-7.29 (m, 2H), 5.62 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.63-4.45 (m, 2H), 4.45-4.34 (m, 2H), 3.79 (d, J=8.7 Hz, 1H), 3.72 (d, J=8.6 Hz, 1H), 2.27 (s, 3H), 1.33 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 660.6 (M+H+). Example 139: (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1031. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.96-7.82 (m, 2H), 7.72 (d, J=5.8 Hz, 2H), 7.53-7.48 (m, 1H), 7.44 (s, 1H), 7.34 (s, 1H), 7.28 (d, J=7.3 Hz, 1H), 6.97 (d, J=8.3 Hz, 1H), 5.53 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.62-4.48 (m, 2H), 4.44-4.33 (m, 2H), 3.79 (d, J=8.7 Hz, 1H), 3.72 (d, J=8.7 Hz, 1H), 2.27 (s, 3H), 1.32 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 642.6 (M+H+). Example 140: (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1030. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.96-7.90 (m, 1H), 7.90-7.79 (m, 2H), 7.74 (d, J=5.6 Hz, 2H), 7.63 (d, J=8.5 Hz, 1H), 7.45 (s, 1H), 7.35 (s, 1H), 7.31 (dd, J=8.1, 2.8 Hz, 1H), 5.61 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.64-4.46 (m, 2H), 4.46-4.26 (m, 2H), 3.74 (m, 2H), 2.26 (s, 3H), 1.33 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 642.6 (M+H+). Example 141: (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1030. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.96-7.85 (m, 2H), 7.81 (d, J=8.6 Hz, 1H), 7.78-7.66 (m, 2H), 7.63 (d, J=8.5 Hz, 1H), 7.44 (s, 1H), 7.35 (s, 1H), 7.27 (d, J=7.3 Hz, 1H), 6.98 (d, J=8.3 Hz, 1H), 5.53 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.63-4.47 (m, 2H), 4.47-4.31 (m, 2H), 3.74 (m, 2H), 2.26 (s, 3H), 1.32 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 624.6 (M+H+). Example 142: (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35 starting with Intermediates I-109 and I-1026. 1H NMR (400 MHz, Methanol-d4) δ 8.17 (d, J=1.2 Hz, 1H), 7.73 (t, J=7.6 Hz, 1H), 7.68-7.54 (m, 4H), 7.43 (s, 1H), 7.16 (q, J=3.8, 3.3 Hz, 2H), 5.63 (s, 2H), 5.22-5.02 (m, 1H), 4.75-4.59 (m, 3H), 4.59-4.38 (m, 3H), 2.77 (q, J=8.5, 7.4 Hz, 1H), 2.49 (t, J=9.8 Hz, 1H), 2.25 (s, 3H). ES/MS m/z: 633.0 (M+H+). Example 143: (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35 starting with Intermediates I-3 and I-1026. 1H NMR (400 MHz, Methanol-d4) δ 8.16 (d, J=1.3 Hz, 1H), 7.81 (dd, J=8.3, 7.3 Hz, 1H), 7.72-7.60 (m, 2H), 7.60-7.49 (m, 2H), 7.41 (s, 1H), 7.19-7.10 (m, 2H), 6.91 (d, J=8.3 Hz, 1H), 5.54 (s, 2H), 5.12 (dd, J=7.4, 2.4 Hz, 1H), 4.76-4.59 (m, 3H), 4.59-4.35 (m, 3H), 2.77 (dtd, J=11.4, 8.2, 6.1 Hz, 1H), 2.48 (ddt, J=11.5, 9.1, 7.2 Hz, 1H), 2.24 (s, 3H). ES/MS m/z: 615.0 (M+H+). Example 144: (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35 starting with Intermediate I-1024 and I 8. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J=1.4 Hz, 1H), 7.98 (dd, J=8.5, 1.5 Hz, 1H), 7.72 (t, J=7.6 Hz, 1H), 7.68-7.64 (m, 1H), 7.64-7.55 (m, 3H), 7.42 (s, 1H), 7.19 (s, 1H), 7.15 (dd, J=8.1, 2.8 Hz, 1H), 5.63 (s, 2H), 5.17 (qd, J=7.1, 2.6 Hz, 1H), 4.76-4.39 (m, 6H), 2.95-2.66 (m, 1H), 2.50 (ddt, J=11.5, 9.1, 7.2 Hz, 1H), 2.25 (s, 3H). ES/MS m/z: 615.0 (M+H+). Example 145: (S)-2-((6-((4-cyano-2-fluorobenzyl)oxy)-5′-fluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((6-((4-cyano-2-fluorobenzyl)oxy)-5′-fluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1020. 1H NMR (400 MHz, DMSO-d6) δ 13.14 (s, 1H), 8.36 (s, 1H), 8.26 (d, J=6.9 Hz, 1H), 7.92 (t, J=9.0 Hz, 2H), 7.80-7.67 (m, 2H), 7.62-7.48 (m, 2H), 7.10 (d, J=8.3 Hz, 1H), 6.95 (d, J=7.4 Hz, 1H), 5.69-5.30 (m, 4H), 5.14 (d, J=6.7 Hz, 1H), 4.54 (d, J=11.2 Hz, 1H), 4.43 (dd, J=11.3, 6.7 Hz, 1H), 3.92-3.61 (m, 2H), 1.38 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 630.2 (M+H+). Example 146: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluoro-3-methylbenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluoro-3-methylbenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediate I-1012 and I-8. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J=1.4 Hz, 1H), 7.98 (dd, J=8.5, 1.5 Hz, 1H), 7.81 (dd, J=8.3, 7.3 Hz, 1H), 7.73-7.63 (m, 2H), 7.63-7.51 (m, 2H), 7.21-7.09 (m, 3H), 6.91 (d, J=8.2 Hz, 1H), 5.56 (s, 2H), 5.15 (qd, J=7.1, 2.5 Hz, 1H), 4.70 (dd, J=15.7, 7.0 Hz, 1H), 4.66-4.39 (m, 6H), 2.77 (dtd, J=11.4, 8.2, 6.1 Hz, 1H), 2.56-2.38 (m, 1H), 2.22 (d, J=2.6 Hz, 3H). ES/MS m/z: 581.2 (M+H+). Example 147: (S)-2-(4-(6-((6-(acetamidomethyl)-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-(acetamidomethyl)-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediate I-1009 and I-8. 1H NMR (400 MHz, Methanol-d4) δ 8.67 (d, J=9.6 Hz, 1H), 8.56 (s, OH), 8.32 (d, J=1.4 Hz, 1H), 7.99 (dd, J=8.5, 1.5 Hz, 1H), 7.88 (dd, J=10.7, 6.3 Hz, 1H), 7.79 (t, J=7.9 Hz, 1H), 7.68 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.6, 1.6 Hz, 1H), 7.20 (dd, J=11.2, 6.0 Hz, 2H), 6.87 (d, J=8.1 Hz, 1H), 5.61 (s, 2H), 5.21 (tt, J=7.3, 3.6 Hz, 1H), 4.80-4.35 (m, 9H), 2.89-2.65 (m, 1H), 2.61-2.35 (m, 1H), 2.04 (s, 3H). ES/MS m/z: 632.2 (M+H+). Example 148: (S)-2-(2,5-difluoro-4-(5-fluoro-4-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(5-fluoro-4-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediate I-1313 and I-14. 1H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J=2.8 Hz, 1H), 8.14 (s, 1H), 7.86 (dd, J=9.6, 6.3 Hz, 2H), 7.81-7.73 (m, 1H), 7.72-7.63 (m, 1H), 7.50 (dd, J=11.4, 1.2 Hz, 1H), 7.42 (dd, J=11.1, 6.0 Hz, 1H), 5.77 (s, 2H), 5.18-4.94 (m, 1H), 4.78 (dd, J=15.5, 7.1 Hz, 1H), 4.69-4.40 (m, 4H), 4.35 (dt, J=8.9, 5.9 Hz, 1H), 2.70 (ddd, J=16.1, 6.3, 2.8 Hz, 1H), 2.43-2.26 (m, 1H). ES/MS m/z: 665.0 (M+H+). Example 149: (S)-2-(2,5-difluoro-4-(4-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(4-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediate I-1002 and I-14. 1H NMR (400 MHz, DMSO-d6) δ 8.73 (d, J=5.8 Hz, 1H), 8.12 (s, 1H), 7.88 (dd, J=10.2, 6.2 Hz, 1H), 7.83 (t, J=7.6 Hz, 1H), 7.77 (dd, J=9.9, 1.6 Hz, 1H), 7.65 (d, J=8.0 Hz, 1H), 7.50 (dd, J=11.5, 1.2 Hz, 1H), 7.41 (dd, J=11.1, 6.0 Hz, 1H), 7.07 (d, J=5.8 Hz, 1H), 5.68 (s, 2H), 5.07 (qd, J=6.9, 2.8 Hz, 1H), 4.78 (dd, J=15.6, 7.1 Hz, 1H), 4.70-4.42 (m, 4H), 4.35 (dt, J=9.1, 6.0 Hz, 1H), 2.86-2.64 (m, 1H), 2.44-2.25 (m, 1H). ES/MS m/z: 647.0 (M+H+). Example 150: 1-(((1R,3R,5R)-2-acetyl-2-azabicyclo[3.1.0]hexan-3-yl)methyl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-(((1R,3R,5R)-2-acetyl-2-azabicyclo[3.1.0]hexan-3-yl)methyl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 4, starting with Intermediates I-1001 and I-7. 1H NMR (400 MHz, DMSO-d6) δ 8.37 (s, 1H), 7.98-7.85 (m, 2H), 7.83 (dd, J=8.4, 1.5 Hz, 1H), 7.75 (q, J=5.4 Hz, 3H), 7.62 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.46 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.69 (s, 1H), 4.62-4.44 (m, 4H), 4.31 (dd, J=14.4, 8.5 Hz, 1H), 2.26 (d, J=14.1 Hz, 1H), 2.11 (s, 2H), 1.80 (dd, J=15.7, 13.0 Hz, 1H), 1.65 (d, J=7.0 Hz, 1H), 0.96-0.80 (m, 2H). ES/MS m/z: 652.2 (M+H+). Example 151: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-3,5-difluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-3,5-difluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1052 and I-1233. 1H NMR (400 MHz, DMSO-d6) δ 8.46 (s, 1H), 8.20 (t, J=9.9 Hz, 1H), 7.89 (d, J=8.2 Hz, 2H), 7.78-7.73 (m, 1H), 7.71 (d, J=8.1 Hz, 2H), 7.61 (d, J=8.4 Hz, 2H), 7.56 (d, J=8.5 Hz, 1H), 5.67 (s, 2H), 5.08 (d, J=6.6 Hz, 1H), 4.57 (d, J=17.7 Hz, 2H), 4.46 (dd, J=11.1, 6.6 Hz, 1H), 4.29 (d, J=16.8 Hz, 1H), 3.80-3.71 (m, 2H), 1.37 (s, 3H), 0.64 (s, 3H). ES/MS m/z: 631.2 (M+H+). Example 152: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-3,5-difluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-3,5-difluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1052 and I-1229. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.15 (t, J=9.9 Hz, 1H), 7.88 (d, J=8.1 Hz, 2H), 7.81 (dd, J=8.5, 1.4 Hz, 1H), 7.75-7.65 (m, 4H), 7.62 (d, J=8.5 Hz, 1H), 7.53 (t, J=8.0 Hz, 1H), 5.65 (s, 2H), 5.00 (d, J=6.6 Hz, 1H), 4.57-4.48 (m, 2H), 4.48-4.36 (m, 2H), 3.78 (d, J=8.6 Hz, 1H), 3.72 (d, J=8.6 Hz, 1H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 613.3 (M+H+). Example 153: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1050 and I-108. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.84 (s, 1H), 8.35 (s, 1H), 8.32 (s, 1H), 8.03 (d, J=1.3 Hz, 1H), 7.88 (ddd, J=11.3, 7.4, 4.3 Hz, 2H), 7.64-7.55 (m, 1H), 7.55-7.44 (m, 2H), 5.76 (s, 2H), 5.03 (d, J=6.5 Hz, 1H), 4.59-4.47 (m, 2H), 4.47-4.35 (m, 2H), 3.74 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 708.0 (M+H+). Example 154: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1050 and I-1230. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.3 Hz, 1H), 8.86 (s, 1H), 8.34 (s, 1H), 8.32 (s, 1H), 8.02 (d, J=1.3 Hz, 1H), 7.99-7.91 (m, 2H), 7.85 (dd, J=10.2, 8.2 Hz, 1H), 7.73 (dd, J=8.3, 2.8 Hz, 1H), 7.50 (dd, J=9.7, 6.9 Hz, 2H), 5.78 (s, 2H), 5.00 (d, J=6.7 Hz, 1H), 4.50 (dd, J=14.1, 8.4 Hz, 2H), 4.46-4.34 (m, 2H), 3.79-3.66 (m, 2H), 1.29 (s, 3H), 0.57 (s, 3H). ES/MS m/z: 690.0 (M+H+). Example 155: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1050 and I-10. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.86 (s, 1H), 8.32 (s, 1H), 8.23 (s, 1H), 8.02 (d, J=1.3 Hz, 1H), 7.97-7.88 (m, 2H), 7.88-7.79 (m, 2H), 7.72 (dd, J=8.2, 2.7 Hz, 1H), 7.60 (d, J=8.4 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 5.77 (s, 2H), 4.59 (s, 2H), 4.47 (s, 2H), 3.66 (t, J=5.0 Hz, 2H), 3.20 (s, 3H). ES/MS m/z: 632.2 (M+H+). Example 156: (S)-2-(2-chloro-4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1049 and I-1030. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.3 Hz, 1H), 8.76 (s, 1H), 8.49 (s, 1H), 8.29 (s, 1H), 8.03 (d, J=1.2 Hz, 1H), 7.89 (t, J=7.8 Hz, 1H), 7.80 (dd, J=8.5, 1.5 Hz, 1H), 7.62 (d, J=8.5 Hz, 1H), 7.52 (s, 1H), 7.36 (s, 1H), 7.29 (d, J=7.3 Hz, 1H), 6.98 (d, J=8.3 Hz, 1H), 5.58 (s, 2H), 5.00 (d, J=6.8 Hz, 1H), 4.61-4.48 (m, 2H), 4.46-4.33 (m, 2H), 3.84-3.69 (m, 2H), 2.33 (s, 3H), 1.32 (s, 3H), 0.64 (s, 3H). ES/MS m/z: 684.2 (M+H+). Example 157: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1049 and I-1231. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (s, 1H), 8.83 (s, 1H), 8.47 (s, 1H), 8.31 (s, 1H), 8.03 (d, J=1.3 Hz, 1H), 7.96 (t, J=7.9 Hz, 1H), 7.77 (d, J=8.2 Hz, 2H), 7.60 (dd, J=10.7, 7.7 Hz, 2H), 7.06 (d, J=8.3 Hz, 1H), 5.68 (s, 2H), 5.08 (d, J=6.5 Hz, 1H), 4.68-4.53 (m, 2H), 4.46 (dd, J=11.3, 6.9 Hz, 1H), 4.35 (d, J=17.2 Hz, 1H), 3.76 (d, J=2.8 Hz, 2H), 1.38 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 690.1 (M+H+). Example 158: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1049 and I-1229. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.3 Hz, 1H), 8.83 (s, 1H), 8.47 (s, 1H), 8.30 (s, 1H), 8.02 (d, J=1.3 Hz, 1H), 8.00-7.93 (m, 2H), 7.88 (t, J=7.9 Hz, 1H), 7.80 (dd, J=8.4, 1.5 Hz, 1H), 7.70 (d, J=7.5 Hz, 1H), 7.61 (d, J=8.5 Hz, 1H), 7.50 (t, J=8.1 Hz, 1H), 6.95 (d, J=8.2 Hz, 1H), 5.69 (s, 2H), 4.98 (d, J=6.8 Hz, 1H), 4.54-4.47 (m, 2H), 4.47-4.35 (m, 2H), 3.81-3.68 (m, 2H), 1.29 (s, 3H), 0.57 (s, 3H). ES/MS m/z: 654.2 (M+H+). Example 159: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1050 and I-1231. 1H NMR (400 MHz, DMSO-d6) δ 8.91 (d, J=1.3 Hz, 1H), 8.85 (s, 1H), 8.47 (s, 1H), 8.33 (s, 1H), 8.03 (d, J=1.2 Hz, 1H), 7.99-7.87 (m, 1H), 7.77 (d, J=8.6 Hz, 2H), 7.64 (d, J=8.4 Hz, 1H), 7.58 (d, J=8.4 Hz, 1H), 5.77 (s, 2H), 5.08 (d, J=6.7 Hz, 1H), 4.64 (d, J=17.5 Hz, 1H), 4.57 (d, J=11.3 Hz, 1H), 4.51-4.42 (m, 1H), 4.35 (d, J=17.6 Hz, 1H), 3.76 (d, J=2.5 Hz, 2H), 1.38 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 708.1 (M+H+). Example 160: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1050 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.84 (s, 1H), 8.47 (s, 1H), 8.32 (s, 1H), 8.03 (d, J=1.2 Hz, 1H), 7.87 (td, J=9.7, 9.1, 7.4 Hz, 2H), 7.82-7.75 (m, 1H), 7.60 (t, J=9.5 Hz, 2H), 7.47 (dd, J=11.6, 6.1 Hz, 1H), 5.76 (s, 2H), 5.01 (d, J=6.8 Hz, 1H), 4.58-4.47 (m, 2H), 4.48-4.32 (m, 2H), 3.80-3.68 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 690.1 (M+H+). Example 161: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1050 and I-1229. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.86 (s, 1H), 8.48 (s, 1H), 8.32 (s, 1H), 8.02 (d, J=1.3 Hz, 1H), 7.94 (t, J=8.5 Hz, 2H), 7.85 (dd, J=10.2, 8.3 Hz, 1H), 7.82-7.77 (m, 1H), 7.73 (dd, J=8.3, 2.7 Hz, 1H), 7.61 (d, J=8.4 Hz, 1H), 7.50 (t, J=8.1 Hz, 1H), 5.78 (s, 2H), 4.99 (d, J=6.6 Hz, 1H), 4.54-4.47 (m, 2H), 4.46-4.35 (m, 2H), 3.81-3.69 (m, 2H), 1.29 (s, 3H), 0.57 (s, 3H). ES/MS m/z: 672.2 (M+H+). Example 162: (S)-2-(4-(6-((3-chloro-5-(1H-1,2,3-triazol-1-yl)pyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((3-chloro-5-(1H-1,2,3-triazol-1-yl)pyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1053 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 9.12 (d, J=2.2 Hz, 1H), 8.95 (d, J=1.2 Hz, 1H), 8.62 (d, J=2.3 Hz, 1H), 8.47 (s, 1H), 8.04 (d, J=1.2 Hz, 1H), 7.89 (t, J=7.9 Hz, 1H), 7.79 (dd, J=8.5, 1.4 Hz, 1H), 7.73 (dd, J=10.6, 6.4 Hz, 1H), 7.60 (d, J=8.4 Hz, 1H), 7.53 (d, J=7.3 Hz, 1H), 7.42 (dd, J=11.5, 6.0 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.73 (s, 2H), 5.02-4.95 (m, 1H), 4.56-4.45 (m, 2H), 4.46-4.30 (m, 2H), 3.82-3.67 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 672.2 (M+H+). Example 163: (S)-2-(2,5-difluoro-4-(5-fluoro-6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(5-fluoro-6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1340 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.2 Hz, 1H), 8.48 (s, 1H), 8.00 (d, J=1.1 Hz, 1H), 7.95 (dd, J=10.8, 2.0 Hz, 1H), 7.88-7.83 (m, 3H), 7.91-7.76 (m, 2H), 7.61 (d, J=8.5 Hz, 1H), 7.59-7.53 (m, 1H), 7.46 (dd, J=11.4, 6.1 Hz, 1H), 5.68 (s, 2H), 5.01 (d, J=6.8 Hz, 1H), 4.57-4.48 (m, 2H), 4.48-4.30 (m, 2H), 3.82-3.69 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 673.2 (M+H+). Example 164: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(4-methyl-1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(4-methyl-1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1058 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.60 (s, 1H), 8.48 (s, 1H), 7.92-7.81 (m, 3H), 7.81-7.75 (m, 3H), 7.61 (d, J=8.4 Hz, 1H), 7.57-7.51 (m, 1H), 7.45 (dd, J=11.3, 6.2 Hz, 1H), 6.98 (d, J=8.3 Hz, 1H), 5.58 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.59-4.48 (m, 2H), 4.50-4.34 (m, 2H), 3.83-3.70 (m, 2H), 2.33 (s, 3H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 669.2 (M+H+). Example 165: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1057. 1H NMR (400 MHz, DMSO-d6) δ 7.94-7.86 (m, 2H), 7.80-7.71 (m, 3H), 7.66 (d, J=7.7 Hz, 1H), 7.54 (d, J=7.4 Hz, 1H), 7.41 (p, J=11.0, 9.8 Hz, 2H), 6.99 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.22 (d, J=112.0 Hz, 1H), 4.54 (d, J=24.0 Hz, 2H), 4.47-4.14 (m, 2H), 3.94-3.58 (m, 2H), 1.29 (d, J=21.9 Hz, 3H), 0.72 (d, J=14.3 Hz, 3H). ES/MS m/z: 631.1 (M+H+). Example 166: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-7-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1056. 1H NMR (400 MHz, DMSO-d6) δ 7.95-7.86 (m, 2H), 7.79-7.71 (m, 3H), 7.66 (t, J=7.6 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.43 (q, J=10.6, 7.8 Hz, 2H), 6.99 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.22 (d, J=111.9 Hz, 1H), 4.54 (d, J=24.1 Hz, 2H), 4.37 (d, J=18.0 Hz, 1H), 3.92-3.67 (m, 1H), 1.29 (d, J=21.9 Hz, 3H), 0.72 (d, J=14.2 Hz, 3H). ES/MS m/z: 631.2 (M+H+). Example 167: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1283 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (s, 1H), 8.48 (s, 1H), 8.00 (s, 1H), 7.95-7.87 (m, 3H), 7.87-7.81 (m, 2H), 7.79 (dd, J=8.3, 1.7 Hz, 2H), 7.61 (d, J=8.5 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.46 (dd, J=11.3, 6.1 Hz, 1H), 6.98 (d, J=8.3 Hz, 1H), 5.59 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.58-4.47 (m, 2H), 4.47-4.28 (m, 2H), 3.81-3.63 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 655.2 (M+H+). Example 168: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1312 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 7.90 (t, J=7.9 Hz, 1H), 7.83-7.70 (m, 4H), 7.62 (t, J=7.8 Hz, 2H), 7.54 (d, J=7.3 Hz, 1H), 7.44 (dd, J=11.3, 6.1 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.00 (d, J=6.6 Hz, 1H), 4.57-4.47 (m, 2H), 4.43 (dd, J=11.1, 6.8 Hz, 1H), 4.36 (d, J=17.0 Hz, 1H), 3.83-3.69 (m, 2H), 1.32 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 656.2 (M+H+). Example 169: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1312 and I-108. 1H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.90 (t, J=7.8 Hz, 1H), 7.81-7.71 (m, 3H), 7.62 (d, J=8.0 Hz, 1H), 7.53 (dd, J=12.7, 9.2 Hz, 2H), 7.45 (dd, J=11.4, 6.1 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.59-4.47 (m, 2H), 4.47-4.34 (m, 2H), 3.74 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 674.1 (M+H+). Example 170: 2-(4-(6-((6-chloro-2-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-chloro-2-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1063 and I-1336. 1H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.92-7.81 (m, 2H), 7.79 (dd, J=10.3, 6.5 Hz, 1H), 7.56-7.49 (m, 2H), 7.46 (dd, J=11.3, 6.2 Hz, 1H), 7.12 (d, J=7.7 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 5.41 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.60-4.47 (m, 2H), 4.47-4.33 (m, 2H), 3.93 (s, 3H), 3.74 (q, J=8.7 Hz, 2H), 3.10 (qd, J=7.3, 4.8 Hz, 1H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 653.2 (M+H+). Example 171: (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1049 and I-108. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.4 Hz, 1H), 8.82 (s, 1H), 8.35 (s, 1H), 8.30 (s, 1H), 8.02 (d, J=1.4 Hz, 1H), 7.92 (d, J=7.7 Hz, 1H), 7.90-7.86 (m, 1H), 7.57 (dd, J=7.6, 1.5 Hz, 1H), 7.51 (d, J=11.4 Hz, 1H), 7.47 (dd, J=11.4, 6.4 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.67 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.58-4.47 (m, 2H), 4.47-4.31 (m, 2H), 3.74 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 690.1 (M+H+). Procedure 36: Example 172 Methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(fluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate. Chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (35 mg, 1.2 equivalent) followed by 1-methylimidazole (42 μL, 5.0 equivalent) were added to a mixture of 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-3-fluoro-phenyl]acetic acid (I-7, 42 mg, 1.0 equivalent) and methyl 4-amino-3-[[(3R,4S)-4-(fluoromethyl)tetrahydrofuran-3-yl]amino]benzoate (I-1064, 28 mg, 1 equivalent) in MeCN (1 mL), and the mixture was stirred for 2 hours. An additional I-7 (10 mg, 0.25 equivalent) and chloro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate (7 mg, 0.25 equivalent) were added, and the mixture was stirred overnight. The mixture was diluted with EtOAc and washed sequentially with saturated aqueous NH4Cl, saturated aqueous NaHCO3, and brine, dried over MgSO4, filtered, and concentrated in vacuo. The crude was mixed with acetic acid (0.42 mL) and 1,2-dichloroethane (1.6 mL), and the mixture was heated in a sealed vial overnight at 95° C. The mixture was then heated at 180° C. in the microwave for 1 hour. The mixture was quenched by dropwise addition of saturated NaHCO3and diluted with EtOAc. Phases were separated, and organic layer was washed with saturated NaHCO3then brine, dried over MgSO4, filtered, concentrated, and purified (0-100% EtOAc in Hex) to give methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(fluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate. 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(fluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 172): Methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(fluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (35 mg, 1.0 equivalent) was mixed with lithium hydroxide monohydrate (2 M in water, 83 μL, 3 equivalent) and acetonitrile (0.8 mL). All components were heated at 100° C. for 6 min. The mixture was neutralized with AcOH, diluted with DMSO, and purified (0-100% ACN in H2O, 0.1% TFA) to give the title compound. 1H NMR (400 MHz, DMSO-d6) δ 8.53 (s, 1H), 7.94-7.90 (m, 1H), 7.89 (d, J=8.0 Hz, 1H), 7.81 (dd, J=8.4, 1.5 Hz, 1H), 7.79-7.70 (m, 3H), 7.62 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.39 (dd, J=11.4, 6.1 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.61 (d, J=9.4 Hz, 3H), 4.56 (d, J=6.1 Hz, 1H), 4.52 (s, 1H), 4.42 (d, J=17.0 Hz, 1H), 4.34-4.10 (m, 2H), 4.06 (td, J=9.0, 1.7 Hz, 1H), 3.97-3.77 (m, 2H), 3.31 (dq, J=15.6, 7.6 Hz, 1H). ES/MS m/z: 617.3 (M+H+). Example 173: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1049 and I-1336. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.3 Hz, 1H), 8.82 (s, 1H), 8.35 (s, 1H), 8.30 (s, 1H), 8.02 (d, J=1.2 Hz, 1H), 7.94-7.91 (m, 1H), 7.90-7.87 (m, 1H), 7.57 (dd, J=7.6, 1.6 Hz, 1H), 7.52 (dd, J=11.2, 1.2 Hz, 1H), 7.47 (dd, J=11.4, 6.5 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.67 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.53 (dd, J=14.2, 11.9 Hz, 2H), 4.47-4.35 (m, 2H), 3.74 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 690.0 (M+H+). Example 174: 2-(4-(6-((4-chloro-6-(1H-imidazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-imidazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1051 and I-1336. 1H NMR (400 MHz, DMSO-d6) δ 9.33 (s, 1H), 8.78 (s, 1H), 8.35 (s, 1H), 8.30 (s, 1H), 8.27 (t, J=1.6 Hz, 1H), 7.92 (d, J=7.9 Hz, 1H), 7.89 (t, J=3.3 Hz, 1H), 7.57 (dt, J=4.4, 1.7 Hz, 2H), 7.52 (dd, J=11.2, 1.2 Hz, 1H), 7.49-7.43 (m, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.66 (s, 2H), 5.03 (d, J=6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.47-4.35 (m, 2H), 3.74 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 689.4 (M+H+). Example 175: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1312 and I-1336. 1H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.90 (t, J=7.9 Hz, 1H), 7.83-7.77 (m, 2H), 7.74 (dd, J=10.4, 1.9 Hz, 1H), 7.64-7.60 (m, 1H), 7.57-7.49 (m, 2H), 7.46 (dd, J=11.4, 6.1 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.61-4.47 (m, 2H), 4.47-4.34 (m, 2H), 3.74 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 674.1 (M+H+). Example 176A: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared from racemic Example 184 by preparative chiral SFC (Daicel Chiralcel AD-H column, 45% MeOH in CO2) as the later eluting of two isomers. 1H NMR (400 MHz, DMSO) δ 8.45 (s, 1H), 7.96-7.86 (m, 2H), 7.80-7.71 (m, 4H), 7.53 (d, J=8.2 Hz, 2H), 7.35 (dd, J=11.0, 5.7 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.48 (s, 1H), 4.49 (d, J=11.9 Hz, 2H), 4.42 (d, J=17.2 Hz, 1H), 4.21 (dd, J=10.8, 6.8 Hz, 1H), 4.16-4.03 (m, 1H), 3.78 (s, 1H), 3.09 (s, 2H), 3.07-2.99 (m, 1H), 2.92 (s, 3H), 2.59 (s, 1H). Example 176B: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared from racemic Example 184 by preparative chiral SFC (Daicel Chiralcel AD-H column, 45% MeOH in CO2) as the earlier eluting of two isomers. 1H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 7.96-7.86 (m, 2H), 7.82-7.71 (m, 4H), 7.62-7.51 (m, 2H), 7.36 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.50 (s, 1H), 4.56-4.47 (m, 2H), 4.43 (d, J=16.8 Hz, 1H), 4.21 (dd, J=10.9, 6.6 Hz, 1H), 4.08 (t, J=8.7 Hz, 1H), 3.77 (t, J=8.2 Hz, 1H), 3.14-3.00 (m, 2H), 2.92 (s, 3H), 2.63-2.52 (m, 1H), 1.24 (s, 1H). ES/MS m/z: 629.0 (M+H+). Example 177: (S)-2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1077 and I-108. 1H NMR (400 MHz, DMSO) δ 13.05 (s, 1H), 8.84 (d, J=1.1 Hz, 1H), 8.35 (s, 1H), 8.01-7.93 (m, 3H), 7.85 (ddd, J=13.3, 10.3, 7.4 Hz, 2H), 7.75 (d, J=8.3 Hz, 2H), 7.60-7.43 (m, 3H), 5.66 (s, 2H), 5.03 (d, J=6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.47-4.34 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: x (M+H+). Example 178: (S)-2-(4-(2-((4-cyanobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-cyanobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1076 and I-1086. 1H NMR (400 MHz, DMSO) δ 8.74 (d, 1H), 8.49 (s, 1H) 7.80-7.90 (m, 3H), 7.70-7.60 (m, 3H), 7.46-7.36 (m, 3H), 5.56 (s, 2H), 5.02 (d, 1H), 4.53-4.43 (m, 4H), 3.82-3.72 (m, 2H), 2.33 (s, 3H), 1.32 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 592.0 (M+H+). Example 179: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1076 and I-7. 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.92-7.78 (m, 4H), 7.63 (dd, J=8.3, 4.5 Hz, 3H), 7.33-7.20 (m, 3H), 5.51 (s, 2H), 4.56-4.38 (m, 3H), 3.82-3.70 (m, 2H), 3.74 (s, 21H), 2.22 (s, 3H), 1.31 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 591.2 (M+H+). Example 180: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1247 and I-7. 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.90-7.78 (m, 4H), 7.64 (dd, J=12.6, 8.3 Hz, 3H), 7.33-7.21 (m, 3H), 5.59 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.56-4.38 (m, 3H), 4.33 (d, J=16.9 Hz, 1H), 3.79 (d, J=8.6 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 2.22 (s, 3H), 1.31 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 610.6 (M+H+). Example 181: (S)-2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1077 and I-82. 1H NMR (400 MHz, DMSO) δ 8.84 (d, J=1.2 Hz, 1H), 8.51 (s, 1H), 8.01-7.93 (m, 3H), 7.91-7.83 (m, 1H), 7.87-7.79 (m, 2H), 7.78-7.71 (m, 2H), 7.64 (d, J=8.4 Hz, 1H), 7.60-7.52 (m, 1H), 7.48 (dd, J=11.4, 6.1 Hz, 1H), 5.66 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.60-4.51 (m, 2H), 4.48-4.37 (m, 2H), 3.79 (d, J=8.6 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 655.2 (M+H+). Example 182: (S)-2-(4-(2-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1087 and I-82. 1H NMR (400 MHz, DMSO) δ 8.87-8.77 (m, 2H), 8.50 (s, 1H), 8.01-7.92 (m, 4H), 7.82 (dd, J=8.5, 1.5 Hz, 1H), 7.74 (d, J=8.4 Hz, 2H), 7.67-7.60 (m, 2H), 7.56 (dd, J=11.4, 5.9 Hz, 1H), 5.60 (s, 2H), 5.21 (s, 15H), 5.03 (d, J=6.6 Hz, 1H), 4.59 (d, J=17.1 Hz, 1H), 4.55 (d, J=11.7 Hz, 1H), 4.48-4.38 (m, 2H), 3.78 (d, J=8.6 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 638.3 (M+H+). Example 183: (S)-2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1088 and I-82. 1H NMR (400 MHz, DMSO) δ 8.83 (d, J=1.2 Hz, 1H), 8.50 (s, 1H), 7.98 (d, J=1.2 Hz, 1H), 7.98-7.91 (m, 2H), 7.90 (t, J=7.8 Hz, 1H), 7.86 (dd, J=10.3, 6.6 Hz, 1H), 7.82 (dd, J=8.4, 1.5 Hz, 1H), 7.76-7.69 (m, 2H), 7.63 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.6, 1.6 Hz, 1H), 7.47 (dd, J=11.2, 6.3 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.57 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.59-4.54 (m, 1H), 4.53 (s, 1H), 4.48-4.35 (m, 2H), 4.03 (s, 21H), 3.78 (d, J=8.7 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 636.6 (M+H+). Example 184: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1065. 1H NMR (400 MHz, DMSO) δ 12.76 (s, 1H), 8.55 (s, 1H), 7.96-7.86 (m, 2H), 7.81 (dd, J=8.3, 1.5 Hz, 1H), 7.81-7.68 (m, 3H), 7.62 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.6, 1.7 Hz, 1H), 7.38 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.52 (s, 1H), 4.58-4.41 (m, 3H), 4.21 (dd, J=10.8, 6.5 Hz, 1H), 4.08 (t, J=8.7 Hz, 1H), 3.77 (t, J=8.1 Hz, 1H), 3.17-3.01 (m, 2H), 2.91 (s, 3H), 2.60 (t, J=8.7 Hz, 1H). ES/MS m/z: 628.2 (M+H+). Example 185: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-((difluoromethoxy)methyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-((difluoromethoxy)methyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates 1-7 and 1-1066. 1H NMR (400 MHz, DMSO) δ 8.53 (s, 1H), 7.96-7.86 (m, 2H), 7.81 (dd, J=8.4, 1.5 Hz, 1H), 7.81-7.70 (m, 3H), 7.66-7.50 (m, 2H), 7.38 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 6.6-6.25 (m, 1H), 5.61 (s, 2H), 4.56 (d, J=16.9 Hz, 1H), 4.50 (d, J=10.9 Hz, 1H), 4.42 (d, J=17.0 Hz, 1H), 4.19 (dd, J=10.9, 6.7 Hz, 1H), 4.09 (t, J=8.6 Hz, 1H), 3.93-3.86 (m, 1H), 3.61-3.51 (m, 1H). ES/MS m/z: 664.6 (M+H+). Example 186: (S)-2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1088 and I-108. 1H NMR (400 MHz, DMSO) δ 13.14 (s, 1H), 8.83 (d, J=1.2 Hz, 1H), 8.36 (s, 1H), 8.00-7.82 (m, 5H), 7.73 (d, J=8.2 Hz, 2H), 7.58-7.49 (m, 2H), 7.47 (dd, J=11.2, 6.2 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 5.04 (d, J=6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 4.26 (s, 13H), 3.75 (q, J=8.7 Hz, 2H), 2.55 (s, 1H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 654.6 (M+H+). Example 187: 1-((1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-((1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 36, starting with Intermediates I-7 and I-1067. 1H NMR (400 MHz, DMSO) δ 12.98 (s, 1H), 8.31 (d, J=1.5 Hz, 1H), 7.96-7.86 (m, 2H), 7.82 (dd, J=8.5, 1.3 Hz, 1H), 7.82-7.70 (m, 3H), 7.66 (d, J=8.4 Hz, 1H), 7.55 (dd, J=7.5, 1.7 Hz, 1H), 7.41 (dd, J=11.3, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.02 (dt, J=11.1, 5.0 Hz, 1H), 4.85 (dt, J=12.8, 4.2 Hz, 2H), 4.58 (d, J=17.0 Hz, 1H), 4.47 (d, J=17.0 Hz, 1H), 2.37 (d, J=10.9 Hz, 1H), 2.10-1.96 (m, 1H), 1.90 (d, J=10.3 Hz, 1H), 1.68 (q, J=5.3 Hz, 2H). ES/MS m/z: 610.6 (M+H+). Example 188: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1247 and I-82. 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.92-7.77 (m, 4H), 7.72 (t, J=8.8 Hz, 3H), 7.62 (d, J=8.5 Hz, 1H), 7.59-7.52 (m, 1H), 7.46 (dd, J=11.4, 6.1 Hz, 1H), 5.67 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.58-4.49 (m, 2H), 4.44 (dd, J=11.2, 6.8 Hz, 1H), 4.38 (d, J=16.9 Hz, 1H), 3.82-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 612.6 (M+H+). Example 189: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-cyclopropyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 190: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-cyclopropyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 191: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-cyclopropyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 192: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-cyclopropyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 189, 190, 191 and 192 were prepared in a manner as described in Procedure 36, starting with Intermediates I-7 and I-1068. This yielded a mixture of four different stereoisomers which were purified by preparative chiral SFC (Daicel Chiralpak IG column, 35% EtOH in CO2) to yield the title compounds. Example 189: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-cyclopropyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was the fourth of four stereoisomers to elute under the conditions described above. 1H NMR (400 MHz, DMSO) δ 8.56 (s, 1H), 8.22 (s, 1H), 7.96-7.87 (m, 2H), 7.82-7.70 (m, 4H), 7.60 (d, J=8.4 Hz, 1H), 7.55 (dd, J=7.5, 1.7 Hz, 1H), 7.41 (dd, J=11.4, 6.1 Hz, 1H), 7.04-6.95 (m, 1H), 5.61 (s, 2H), 5.48 (t, J=7.2 Hz, 1H), 4.63-4.53 (m, 2H), 4.47 (d, J=17.0 Hz, 1H), 4.23 (dd, J=10.9, 6.6 Hz, 1H), 4.12 (t, J=8.5 Hz, 1H), 3.82 (t, J=8.4 Hz, 1H), 3.02-2.87 (m, 1H), 2.18 (p, J=8.4 Hz, 1H), 1.24 (s, 1H), 1.16 (t, J=7.3 Hz, 2H), 0.24 (s, 2H), 0.11 (d, J=7.1 Hz, 2H), −0.12 (s, 1H). ES/MS m/z: 624.6 (M+H+). Example 190: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-cyclopropyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was the third of four stereoisomers to elute under the conditions described above. 1H NMR (400 MHz, DMSO) δ 12.74 (s, 1H), 8.55 (s, 1H), 7.96-7.87 (m, 2H), 7.81-7.70 (m, 4H), 7.63-7.51 (m, 2H), 7.41 (dd, J=11.3, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.47 (t, J=7.2 Hz, 1H), 4.62-4.52 (m, 2H), 4.46 (d, J=16.9 Hz, 1H), 4.23 (dd, J=10.8, 6.7 Hz, 1H), 4.12 (t, J=8.5 Hz, 1H), 3.81 (t, J=8.4 Hz, 1H), 2.17 (p, J=8.5 Hz, 1H), 1.24 (s, 1H), 0.23 (s, 2H), 0.11 (s, 1H), −0.13 (s, 1H). ES/MS m/z: 624.6 (M+H+). Example 191: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-cyclopropyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was the second of four stereoisomers to elute under the conditions described above. 1H NMR (400 MHz, DMSO) δ 8.28 (s, 1H), 7.96-7.86 (m, 2H), 7.84-7.72 (m, 4H), 7.62 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.4 Hz, 1H), 7.41 (dd, J=11.4, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.31 (s, 1H), 4.76-4.42 (m, 2H), 4.35 (t, J=8.2 Hz, 1H), 4.31-4.12 (m, 2H), 3.58 (t, J=9.6 Hz, 1H), 2.89 (d, J=7.4 Hz, 1H), 2.68 (s, 1H), 2.34 (s, 1H), 1.92 (s, OH), 1.24 (s, 2H), 1.14 (t, J=7.2 Hz, 2H), 1.04-0.78 (m, 2H), 0.38 (d, J=9.7 Hz, 2H), 0.11 (s, 1H), −0.38 (d, J=5.2 Hz, 1H). ES/MS m/z: 624.6 (M+H+). Example 192: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-cyclopropyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was the first of four stereoisomers to elute under the conditions described above. 1H NMR (400 MHz, DMSO) δ 12.79 (s, 1H), 8.28 (s, 1H), 8.00-7.84 (m, 2H), 7.77 (dd, J=15.6, 7.3 Hz, 3H), 7.63 (d, J=8.5 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.41 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.32 (s, 1H), 4.62-4.42 (m, 2H), 4.40-4.09 (m, 2H), 3.58 (t, J=9.6 Hz, 1H), 1.92 (d, J=8.1 Hz, 0H), 1.02-0.82 (m, 1H), 0.37 (d, J=13.0 Hz, 1H), 0.10 (d, J=5.4 Hz, 1H), −0.37 (d, J=4.9 Hz, 1H). ES/MS m/z: 624.6 (M+H+). Example 193: (S)-2-(4-(2-((4-cyanobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-cyanobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1086 and I-82. 1H NMR (400 MHz, DMSO) δ 8.79 (d, J=5.2 Hz, 1H), 8.36 (s, 1H), 7.91 (dd, J=17.7, 7.2 Hz, 3H), 7.70 (d, J=8.2 Hz, 2H), 7.65 (dd, J=5.2, 1.8 Hz, 1H), 7.60-7.48 (m, 2H), 5.62 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.58 (d, J=17.1 Hz, 1H), 4.53 (d, J=11.8 Hz, 1H), 4.48-4.37 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 595.6 (M+H+). Example 194: (S)-2-(4-(2-((4-cyanobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-cyanobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1086 and I-108. 1H NMR (400 MHz, DMSO) δ 8.79 (d, J=5.2 Hz, 1H), 8.36 (s, 1H), 7.91 (dd, J=17.7, 7.2 Hz, 3H), 7.70 (d, J=8.2 Hz, 2H), 7.65 (dd, J=5.2, 1.8 Hz, 1H), 7.60-7.48 (m, 2H), 5.62 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.58 (d, J=17.1 Hz, 1H), 4.53 (d, J=11.8 Hz, 1H), 4.48-4.37 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 613.6 (M+H+). Example 195: (S)-2-(4-(6-((5-(1,1-difluoroethyl)pyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((5-(1,1-difluoroethyl)pyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1074 and I-82. 1H NMR (400 MHz, DMSO) δ 13.08 (s, 1H), 8.49 (d, J=1.3 Hz, 1H), 8.35 (s, 1H), 7.93 (dd, J=10.2, 8.3 Hz, 1H), 7.82 (dd, J=10.2, 6.7 Hz, 1H), 7.66-7.59 (m, 1H), 7.56-7.43 (m, 2H), 5.98 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.59-4.48 (m, 2H), 4.47-4.35 (m, 2H), 3.84 (s, 14H), 3.74 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 634.6 (M+H+). Example 196: (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1232 and I-105. 1H NMR (400 MHz, DMSO) δ 13.08 (s, 1H), 8.49 (d, J=1.3 Hz, 1H), 8.35 (s, 1H), 7.93 (dd, J=10.2, 8.3 Hz, 1H), 7.82 (dd, J=10.2, 6.7 Hz, 1H), 7.66-7.59 (m, 1H), 7.56-7.43 (m, 2H), 5.98 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.59-4.48 (m, 2H), 4.47-4.35 (m, 2H), 3.84 (s, 14H), 3.74 (q, J=8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 658.6 (M+H+). Example 197: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2S,3R)-2-ethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2S,3R)-2-ethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 36, starting with Intermediates I-7 and I-1069. 1H NMR (400 MHz, DMSO) δ 8.32 (d, J=1.5 Hz, 1H), 7.96-7.64 (m, 7H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.42 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.09-4.99 (m, 1H), 4.60-4.44 (m, 2H), 4.23 (ddt, J=13.1, 8.7, 3.6 Hz, 2H), 4.00 (q, J=8.0 Hz, 1H), 2.39-2.26 (m, 1H), 1.66-1.47 (m, 2H), 0.86 (t, J=7.4 Hz, 3H). ES/MS m/z: 612.6 (M+H+). Example 198: (S)-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1175 and I-82. 1H NMR (400 MHz, DMSO) δ 8.79 (d, J=5.2 Hz, 1H), 8.49 (s, 1H), 7.98-7.88 (m, 2H), 7.84-7.72 (m, 3H), 7.69-7.59 (m, 2H), 7.55 (dd, J=11.4, 5.9 Hz, 1H), 5.64 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.62-4.50 (m, 2H), 4.48-4.37 (m, 2H), 3.82-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 614.5 (M+H+). Example 199: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2,2,5,5-tetramethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2,2,5,5-tetramethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 36, starting with Intermediates I-7 and I-1070. 1H NMR (400 MHz, DMSO) δ 8.37 (d, J=1.5 Hz, 1H), 7.95-7.63 (m, 7H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.44 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.23 (dd, J=10.7, 8.3 Hz, 1H), 4.67 (d, J=17.0 Hz, 1H), 4.44 (d, J=16.9 Hz, 1H), 3.00-2.90 (m, 1H), 2.39 (dd, J=12.7, 8.2 Hz, 1H), 1.50 (s, 3H), 1.39 (d, J=17.9 Hz, 6H), 1.07 (s, 3H). ES/MS m/z: 641.2 (M+H+). Example 200: 2-(2,5-difluoro-4-(2-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidin-4-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(2-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidin-4-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1091 and I-108. 1H NMR (400 MHz, DMSO) δ 13.09 (s, 1H), 8.80 (d, J=5.1 Hz, 1H), 8.36 (s, 1H), 7.93 (dd, J=10.2, 6.2 Hz, 1H), 7.87-7.73 (m, 2H), 7.66 (dt, J=5.2, 2.6 Hz, 2H), 7.60-7.48 (m, 2H), 5.65 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.58 (d, J=17.1 Hz, 1H), 4.53 (d, J=11.9 Hz, 1H), 4.48-4.38 (m, 2H), 4.11 (s, 7H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 675.0 (M+H+). Example 201: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-108. 1H NMR (400 MHz, DMSO) δ 13.07 (s, 1H), 8.36 (s, 1H), 7.93-7.81 (m, 2H), 7.65-7.42 (m, 5H), 7.33 (dd, J=8.2, 2.1 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.04 (d, J=6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J=8.6 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 640.2 (M+H+). Example 202: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2,5-dioxaspiro[3.4]octan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2,5-dioxaspiro[3.4]octan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1071. 1H NMR (400 MHz, DMSO) δ 8.20 (s, 1H), 7.96-7.86 (m, 2H), 7.83-7.70 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.53 (d, J=7.0 Hz, 1H), 7.34 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.53 (s, 1H), 4.89 (d, J=7.0 Hz, 1H), 4.75 (d, J=7.1 Hz, 1H), 4.63 (q, J=6.8 Hz, 2H), 4.47 (s, 2H), 4.25 (dd, J=10.4, 3.4 Hz, 1H), 4.16 (dd, J=10.4, 7.6 Hz, 1H), 3.31 (s, 1H), 2.93 (dd, J=14.0, 9.1 Hz, 1H), 2.51-2.39 (m, 1H), 1.24 (s, 1H). ES/MS m/z: 627.2 (M+H+). Example 203: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4-oxaspiro[2.4]heptan-6-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4-oxaspiro[2.4]heptan-6-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1072. 1H NMR (400 MHz, DMSO) δ 12.95 (s, 1H), 8.60 (d, J=1.5 Hz, 1H), 7.96-7.86 (m, 3H), 7.82-7.66 (m, 4H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.42 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.73 (qd, J=7.6, 3.6 Hz, 1H), 5.60 (s, 2H), 4.61 (s, 2H), 4.17 (qd, J=10.2, 5.5 Hz, 2H), 2.51-2.40 (m, 2H), 1.20-1.09 (m, 1H), 0.82 (td, J=10.4, 6.0 Hz, 2H), 0.65-0.54 (m, 1H). ES/MS m/z: 611.2 (M+H+). Example 204: (S)-2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1295 and I-108. 1H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 8.05 (dd, J=9.2, 5.2 Hz, 1H), 7.92 (t, J=7.9 Hz, 1H), 7.77 (ddd, J=12.5, 9.8, 6.1 Hz, 2H), 7.60-7.42 (m, 3H), 7.02 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J=8.7 Hz, 3H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 649.2 (M+H+). Example 205: (S)-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1175 and I-108. 1H NMR (400 MHz, DMSO) δ 8.79 (d, J=5.1 Hz, 1H), 8.36 (s, 1H), 7.98-7.89 (m, 2H), 7.83-7.72 (m, 2H), 7.65-7.67 (m, 1H), 7.60-7.48 (m, 2H), 5.64 (s, 2H), 5.04-5.02 (m, 1H), 4.63-4.49 (m, 2H), 4.48-4.37 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 632.2 (M+H+). Example 206: (S)-2-(4-(4-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(4-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1338 and I-108. 1H NMR (400 MHz, DMSO) δ 8.82-8.71 (m, 1H), 8.36 (s, 1H), 7.98-7.85 (m, 2H), 7.83-7.72 (m, 2H), 7.60-7.43 (m, 2H), 7.08 (d, J=5.8 Hz, 1H), 5.66 (d, J=13.7 Hz, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.63-4.49 (m, 2H), 4.48-4.37 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (d, J=2.8 Hz, 3H), 0.62 (d, J=2.7 Hz, 3H). ES/MS m/z: 632.2 (M+H+). Example 207: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1075. 1H NMR (400 MHz, DMSO) δ 13.06 (s, 1H), 8.35 (s, 1H), 7.96-7.89 (m, 1H), 7.85 (dd, J=10.5, 8.1 Hz, 1H), 7.74 (dd, J=3.5, 1.6 Hz, 2H), 7.52 (dd, J=11.2, 1.2 Hz, 1H), 7.34-7.24 (m, 3H), 5.61 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.55-4.38 (m, 3H), 4.34 (d, J=16.8 Hz, 1H), 3.80-3.69 (m, 2H), 2.26 (s, 3H), 1.31 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 645.2 (M+H+). Example 208: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1075. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.94-7.86 (m, 2H), 7.75-7.70 (m, 2H), 7.52 (d, 1H), 7.34-7.22 (m, 3H), 6.96 (d, 1H), 5.53 (s, 2H), 5.02 (d, 1H), 4.53-4.41 (m, 3H), 4.34 (d, 1H), 3.77-3.69 (dd, 2H), 2.26 (s, 3H), 1.31 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 627.2 (M+H+). Example 209: (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-1110 and I-1329. 1H NMR (400 MHz, MeOD) δ 8.81 (d, J=1.4 Hz, 1H), 8.22 (dd, J=8.6, 1.4 Hz, 1H), 7.97-7.86 (m, 2H), 7.86-7.79 (m, 2H), 7.73 (t, J=7.6 Hz, 1H), 7.61 (dd, J=9.7, 1.5 Hz, 1H), 7.57 (dd, J=7.7, 1.7 Hz, 2H), 7.50 (t, J=7.9 Hz, 1H), 6.92 (d, J=8.3 Hz, 1H), 5.65 (s, 2H), 4.78 (d, J=2.6 Hz, 2H), 4.48 (t, J=8.8 Hz, 1H), 4.40-4.32 (m, 1H), 4.04 (dd, J=11.0, 7.5 Hz, 1H), 3.80 (td, J=9.7, 6.9 Hz, 1H), 2.54 (dt, J=14.3, 7.3 Hz, 1H), 2.35-2.20 (m, 1H). ES/MS m/z: 567.4 (M+H+). Example 210: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-1111 and I-1329. 1H NMR (400 MHz, MeOD) δ 8.84 (d, J=4.2 Hz, 1H), 8.25 (dd, J=8.5, 4.2 Hz, 1H), 8.01-7.87 (m, 2H), 7.82 (t, J=7.7 Hz, 2H), 7.73 (t, J=7.6 Hz, 1H), 7.64-7.49 (m, 4H), 6.93 (d, J=8.2 Hz, 1H), 5.65 (s, 2H), 4.81 (s, 2H), 4.53-4.45 (m, 1H), 4.38 (d, J=11.1 Hz, 1H), 4.06 (dd, J=11.1, 7.5 Hz, 1H), 3.81 (td, J=9.7, 6.7 Hz, 1H), 2.56 (q, J=12.0, 10.2 Hz, 1H), 2.29 (d, J=13.8 Hz, 1H). ES/MS m/z: 567.3 (M+H+). Example 211: (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1174 and I-1075. 1H NMR (400 MHz, DMSO-d6) δ 8.73 (d, J=5.0 Hz, 1H), 8.35 (s, 1H), 7.56-7.39 (m, 7H), 7.38 (d, J=7.5 Hz, 1H), 5.46 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.57-4.31 (m, 4H), 3.75 (q, J=8.7 Hz, 2H), 2.36 (s, 3H), 1.32 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 619.3 (M+H+). Example 212: (S)-2-(2-chloro-4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1174 and I-1031. 1H NMR (400 MHz, DMSO-d6) δ 8.74 (d, J=5.1 Hz, 1H), 8.36 (s, 1H), 7.63 (s, 1H), 7.55-7.43 (m, 6H), 7.41 (s, 1H), 5.46 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.59 (d, J=17.1 Hz, 1H), 4.52 (d, J=11.1 Hz, 1H), 4.47-4.36 (m, 2H), 3.85-3.68 (m, 2H), 2.37 (s, 3H), 1.34 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 635.0 (M+H+). Example 213: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1050 and I-1135. 1H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J=1.3 Hz, 1H), 8.86 (s, 1H), 8.33 (s, 1H), 8.11 (d, J=1.3 Hz, 1H), 8.04 (d, J=1.2 Hz, 1H), 7.94 (dd, J=10.2, 8.2 Hz, 1H), 7.77 (dd, J=10.4, 5.7 Hz, 1H), 7.69-7.60 (m, 1H), 7.54-7.46 (m, 1H), 5.77 (s, 2H), 4.68 (t, J=5.1 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J=4.9 Hz, 2H), 3.25 (s, 3H). ES/MS m/z: 686.0 (M+H+). Example 214: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 38, starting with Intermediates I-1049 and I-11. 1H NMR (400 MHz, DMSO-d6) δ 8.91 (s, 1H), 8.83 (s, 1H), 8.31 (s, 1H), 8.22 (s, 1H), 8.03 (s, 1H), 7.95-7.84 (m, 2H), 7.80 (d, J=8.7 Hz, 1H), 7.62 (s, 1H), 7.55 (d, J=7.7 Hz, 1H), 7.40 (dd, J=11.6, 6.2 Hz, 1H), 7.01 (d, J=8.4 Hz, 1H), 5.67 (s, 2H), 4.60 (s, 2H), 4.46 (s, 2H), 3.69 (t, J=5.0 Hz, 2H), 3.21 (s, 3H). ES/MS m/z: 632.0 (M+H+). Example 215: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1050 and I-11. 1H NMR (400 MHz, DMSO-d6) δ 8.91 (d, J=1.3 Hz, 1H), 8.85 (s, 1H), 8.33 (s, 1H), 8.22 (s, 1H), 8.04 (d, J=1.3 Hz, 1H), 7.93-7.83 (m, 2H), 7.83-7.78 (m, 1H), 7.61 (d, J=8.4 Hz, 1H), 7.57 (d, J=8.9 Hz, 1H), 7.41 (dd, J=10.9, 6.5 Hz, 1H), 5.76 (s, 2H), 4.60 (d, J=5.7 Hz, 2H), 4.46 (s, 2H), 3.69 (t, J=5.0 Hz, 2H), 3.21 (s, 3H). ES/MS m/z: 650.0 (M+H+). Example 216: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(hydroxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(hydroxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 5, starting with Intermediates I-7 and I-1140. 1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.98-7.85 (m, 2H), 7.85-7.70 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.37 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.53 (d, J=7.6 Hz, 1H), 4.55 (s, 2H), 4.50 (d, J=10.8 Hz, 1H), 4.21 (dd, J=10.9, 6.5 Hz, 1H), 4.06 (t, J=8.7 Hz, 1H), 3.80 (t, J=8.3 Hz, 1H), 3.18-3.09 (m, 1H), 3.00 (q, J=7.8 Hz, 1H), 2.75 (t, J=9.5 Hz, 1H). ES/MS m/z: 615.0 (M+H+). Example 217: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-(hydroxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-(hydroxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 5, starting with Intermediates I-7 and I-1141. 1H NMR (400 MHz, DMSO-d6) δ 8.42 (s, 1H), 7.96-7.86 (m, 2H), 7.84 (dd, J=8.5, 1.5 Hz, 1H), 7.77 (tt, J=8.8, 4.8 Hz, 3H), 7.65 (d, J=8.5 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.39 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.26 (s, 1H), 4.65-4.46 (m, 2H), 4.37 (t, J=8.5 Hz, 1H), 4.21 (dd, J=10.5, 3.2 Hz, 1H), 4.08 (dd, J=10.4, 7.9 Hz, 1H), 3.62-3.46 (m, 3H), 2.76 (d, J=7.3 Hz, 1H). ES/MS m/z: 615.0 (M+H+). Example 218: (S)-2-(2-chloro-4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1174 and I-1030. 1H NMR (400 MHz, DMSO-d6) δ 8.74 (d, J=5.0 Hz, 1H), 8.50 (s, 1H), 7.81 (dd, J=8.4, 1.5 Hz, 1H), 7.62 (t, J=4.2 Hz, 2H), 7.55-7.42 (m, 5H), 7.41 (s, 1H), 5.46 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.56 (dd, J=22.6, 14.0 Hz, 2H), 4.49-4.35 (m, 2H), 3.81 (d, J=8.7 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 2.36 (s, 3H), 1.34 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 618.0 (M+H+). Example 219: (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1174 and I-1076. 1H NMR (400 MHz, DMSO-d6) δ 8.73 (d, J=5.1 Hz, 1H), 8.49 (s, 1H), 7.81 (dd, J=8.5, 1.5 Hz, 1H), 7.62 (d, J=8.5 Hz, 1H), 7.54-7.45 (m, 4H), 7.44-7.33 (m, 3H), 5.45 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.58-4.29 (m, 4H), 3.82-3.67 (m, 2H), 2.36 (s, 3H), 1.32 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 601.0 (M+H+). Example 220: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1142. 1H NMR (400 MHz, DMSO-d6) δ 8.39 (s, 1H), 7.92-7.79 (m, 2H), 7.61 (t, J=8.2 Hz, 1H), 7.57-7.45 (m, 3H), 7.43-7.26 (m, 2H), 6.95 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.24 (s, 1H), 4.42 (s, 2H), 4.35-4.23 (m, 2H), 4.03 (d, J=8.9 Hz, 1H), 3.90 (d, J=8.9 Hz, 1H), 1.00-0.44 (m, 3H), −0.11 (s, 1H). ES/MS m/z: 638.0 (M+H+). Example 221: 2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1268 and I-1033. 1H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J=1.2 Hz, 1H), 8.76 (d, J=2.1 Hz, 1H), 8.25 (dd, J=8.3, 2.2 Hz, 1H), 8.17 (d, J=8.4 Hz, 1H), 8.10 (d, J=1.3 Hz, 1H), 8.00 (d, J=1.2 Hz, 1H), 7.95-7.80 (m, 2H), 7.57-7.47 (m, 2H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.63 (s, 2H), 4.62 (t, J=5.1 Hz, 2H), 4.47 (s, 2H), 3.68 (t, J=5.0 Hz, 2H), 3.21 (s, 3H). ES/MS m/z: 616.0 (M+H+). Example 222: 2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1088 and I-1135. 1H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J=1.2 Hz, 1H), 8.11 (d, J=1.3 Hz, 1H), 8.04-7.89 (m, 4H), 7.73 (d, J=8.5 Hz, 3H), 7.63-7.55 (m, 1H), 7.53-7.44 (m, 1H), 7.05 (d, J=8.3 Hz, 1H), 5.58 (s, 2H), 4.68 (t, J=5.2 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J=4.9 Hz, 2H), 3.24 (s, 3H). ES/MS m/z: 633.0 (M+H+). Example 223: 2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1077 and I-1135. 1H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J=1.2 Hz, 1H), 8.11 (d, J=1.3 Hz, 1H), 8.02-7.86 (m, 4H), 7.73 (dd, J=18.0, 8.6 Hz, 3H), 7.65-7.56 (m, 1H), 7.50 (dd, J=11.4, 1.3 Hz, 1H), 5.67 (s, 2H), 4.68 (t, J=5.0 Hz, 2H), 4.54 (s, 2H), 3.73 (t, J=5.0 Hz, 2H), 3.24 (s, 3H). ES/MS m/z: 651.0 (M+H+). Example 224: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1142. 1H NMR (400 MHz, DMSO-d6) δ 8.39 (s, 1H), 8.01-7.93 (m, 1H), 7.87 (dd, J=10.2, 8.2 Hz, 1H), 7.84-7.70 (m, 3H), 7.54 (dd, J=14.2, 10.3 Hz, 2H), 7.37 (dd, J=11.2, 6.3 Hz, 1H), 5.70 (s, 2H), 5.24 (s, 1H), 4.41 (s, 2H), 4.35-4.24 (m, 2H), 4.02 (d, J=8.9 Hz, 1H), 3.90 (d, J=8.9 Hz, 1H), 0.93-0.50 (m, 3H), −0.11 (s, 1H). ES/MS m/z: 647.0 (M+H+). Example 225: 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1339. 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J=1.2 Hz, 1H), 7.96-7.91 (m, 1H), 7.85 (dd, J=10.5, 8.1 Hz, 1H), 7.74 (d, J=4.7 Hz, 2H), 7.55-7.47 (m, 1H), 7.43 (s, 1H), 7.31 (d, J=9.4 Hz, 2H), 5.61 (s, 2H), 4.61 (t, J=5.1 Hz, 2H), 4.47 (s, 2H), 3.69 (t, J=5.0 Hz, 2H), 3.22 (s, 3H), 2.25 (s, 3H). ES/MS m/z: 621.0 (M+H+). Example 226: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1049 and I-1135. 1H NMR (400 MHz, DMSO-d6) δ 8.91 (d, J=1.3 Hz, 1H), 8.84 (s, 1H), 8.31 (s, 1H), 8.11 (d, J=1.3 Hz, 1H), 8.03 (d, J=1.3 Hz, 1H), 7.96 (t, J=7.9 Hz, 1H), 7.84-7.74 (m, 1H), 7.61 (d, J=7.3 Hz, 1H), 7.50 (d, J=11.4 Hz, 1H), 7.06 (d, J=8.3 Hz, 1H), 5.68 (s, 2H), 4.68 (t, J=5.1 Hz, 2H), 4.56 (s, 2H), 3.73 (t, J=5.0 Hz, 2H), 3.25 (s, 3H). ES/MS m/z: 668.0 (M+H+). Example 227: 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-methylbenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1339. 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J=1.3 Hz, 1H), 7.95-7.84 (m, 2H), 7.72 (d, J=5.8 Hz, 2H), 7.50 (dd, J=11.4, 1.3 Hz, 1H), 7.43 (s, 1H), 7.29 (s, 1H), 7.27 (d, J=7.3 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.52 (s, 2H), 4.60 (t, J=5.1 Hz, 2H), 4.47 (s, 2H), 3.68 (t, J=5.0 Hz, 2H), 3.22 (s, 3H), 2.25 (s, 3H). ES/MS m/z: 603.0 (M+H+). Example 228: (S)-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1246 and I-108. 1H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.83 (ddd, J=20.6, 10.3, 7.3 Hz, 2H), 7.59-7.50 (m, 4H), 7.50-7.43 (m, 3H), 5.57 (s, 2H), 5.04 (d, J=6.8 Hz, 1H), 4.64-4.49 (m, 2H), 4.49-4.33 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 640.1 (M+H+). Example 229: (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1174 and I-1232. 1H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J=5.1 Hz, 1H), 8.36 (s, 1H), 7.90-7.79 (m, 1H), 7.69 (dd, J=5.1, 1.7 Hz, 1H), 7.60-7.41 (m, 5H), 5.52 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.70 (d, J=17.4 Hz, 1H), 4.56 (d, J=11.5 Hz, 1H), 4.52-4.31 (m, 2H), 3.76 (s, 2H), 1.40 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 641.6 (M+H+). Example 230: (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1174 and I-108. 1H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J=5.1 Hz, 1H), 8.36 (s, 1H), 7.95 (dd, J=10.2, 6.2 Hz, 1H), 7.63 (dd, J=5.3, 1.8 Hz, 1H), 7.59-7.49 (m, 4H), 7.47 (d, J=8.4 Hz, 2H), 5.51 (s, 2H), 5.04 (d, J=6.5 Hz, 1H), 4.56 (dd, J=22.4, 14.1 Hz, 2H), 4.49-4.35 (m, 2H), 3.75 (q, J=8.6 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 623.6 (M+H+). Example 231: (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chlorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1174 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J=5.2 Hz, 1H), 8.48 (s, 1H), 7.93 (dd, J=10.2, 6.2 Hz, 1H), 7.79 (dd, J=8.5, 1.4 Hz, 1H), 7.65-7.59 (m, 2H), 7.59-7.51 (m, 3H), 7.47 (d, J=8.5 Hz, 2H), 5.51 (s, 2H), 5.01 (d, J=6.5 Hz, 1H), 4.61-4.49 (m, 2H), 4.49-4.34 (m, 2H), 3.76 (q, J=8.6 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 606.6 (M+H+). Example 232: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1135. 1H NMR (400 MHz, DMSO-d6) δ 8.11 (d, J=1.3 Hz, 1H), 7.97-7.86 (m, 2H), 7.83-7.74 (m, 2H), 7.64 (ddt, J=16.9, 8.4, 1.7 Hz, 2H), 7.50 (dd, J=11.4, 1.3 Hz, 1H), 5.70 (s, 2H), 4.68 (t, J=5.1 Hz, 2H), 4.54 (s, 2H), 3.73 (t, J=5.0 Hz, 2H), 3.24 (s, 3H). ES/MS m/z: 626.6 (M+H+). Example 233: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared via preparative chiral SFC (Daicel Chiralpak AD-H column, EtOH/CO2) of Example 287, as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.97-7.87 (m, 2H), 7.84-7.71 (m, 4H), 7.61 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.39 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.82-5.40 (m, 3H), 4.62-4.49 (m, 2H), 4.40 (d, J=17.0 Hz, 1H), 4.29-4.13 (m, 2H), 4.09 (t, J=9.1 Hz, 1H), 3.58-3.49 (m, 2H). ES/MS m/z: 634.6 (M+H+). Example 234: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared via preparative chiral SFC (Daicel Chiralpak AD-H column, EtOH/CO2) of Example 287, as the later-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.97-7.87 (m, 2H), 7.84-7.71 (m, 4H), 7.61 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.39 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.82-5.40 (m, 3H), 4.62-4.49 (m, 2H), 4.40 (d, J=17.0 Hz, 1H), 4.29-4.13 (m, 2H), 4.09 (t, J=9.1 Hz, 1H), 3.58-3.49 (m, 2H). ES/MS m/z: 634.6 (M+H+). Example 235: (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-2-(2-fluoro-4-(5-fluoro-6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-2-(2-fluoro-4-(5-fluoro-6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1340 and I-1230. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.2 Hz, 1H), 8.35 (s, 1H), 8.01 (d, J=1.2 Hz, 1H), 7.99-7.90 (m, 3H), 7.90-7.79 (m, 3H), 7.72 (dd, J=8.3, 2.8 Hz, 1H), 7.56-7.46 (m, 2H), 5.71 (s, 2H), 5.01 (d, J=6.5 Hz, 1H), 4.59-4.46 (m, 2H), 4.46-4.34 (m, 2H), 3.82-3.69 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 673.3 (M+H+). Example 236: (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2-fluoro-4-(5-fluoro-6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2-fluoro-4-(5-fluoro-6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1340 and I-1229. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.50 (s, 1H), 8.01 (d, J=1.2 Hz, 1H), 7.99-7.90 (m, 3H), 7.90-7.81 (m, 4H), 7.72 (dd, J=8.2, 2.7 Hz, 1H), 7.63 (d, J=8.5 Hz, 1H), 7.51 (t, J=8.1 Hz, 1H), 5.71 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.63-4.49 (m, 2H), 4.49-4.37 (m, 2H), 3.83-3.67 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 655.4 (M+H+). Example 237: (S)-2-(2,5-difluoro-4-(6-((3-fluoro-5-methylpyridin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((3-fluoro-5-methylpyridin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1143 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.51 (s, 1H), 8.29 (s, 1H), 7.93-7.79 (m, 3H), 7.63 (dd, J=11.7, 9.4 Hz, 2H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.46 (dd, J=11.5, 6.0 Hz, 1H), 6.94 (d, J=8.3 Hz, 1H), 5.56 (d, J=1.8 Hz, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.61-4.50 (m, 2H), 4.49-4.36 (m, 2H), 3.86-3.57 (m, 2H), 2.34 (s, 3H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 603.4 (M+H+). Example 238: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared was prepared via preparative chiral SFC (Daicel AD-H column, MeOH/CO2) of Example 256, as the later-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J=1.3 Hz, 1H), 7.99-7.83 (m, 2H), 7.83-7.68 (m, 3H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.49 (dd, J=11.2, 1.3 Hz, 1H), 7.31 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.73-5.47 (m, 3H), 4.53 (s, 2H), 4.40 (dd, J=10.7, 3.4 Hz, 1H), 4.17-4.09 (m, 2H), 4.01 (dd, J=10.6, 8.2 Hz, 1H), 3.78 (dd, J=10.5, 4.6 Hz, 1H), 2.87 (s, 3H). ES/MS m/z: 633.0 (M+H+). Example 239: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-((3S,4S)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-((3S,4S)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared was prepared was prepared via preparative chiral SFC (Daicel AD-H column, MeOH/CO2) of Example 256, as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J=1.3 Hz, 1H), 7.99-7.83 (m, 2H), 7.83-7.68 (m, 3H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.49 (dd, J=11.2, 1.3 Hz, 1H), 7.31 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.73-5.47 (m, 3H), 4.53 (s, 2H), 4.40 (dd, J=10.7, 3.4 Hz, 1H), 4.17-4.09 (m, 2H), 4.01 (dd, J=10.6, 8.2 Hz, 1H), 3.78 (dd, J=10.5, 4.6 Hz, 1H), 2.87 (s, 3H). ES/MS m/z: 633.0 (M+H+). Example 240: (S)-2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1022 and I-108. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.94-7.78 (m, 2H), 7.55-7.42 (m, 5H), 7.35 (d, J=7.8 Hz, 1H), 6.94 (d, J=8.3 Hz, 1H), 5.49 (s, 2H), 5.04 (d, J=6.5 Hz, 1H), 4.64 (d, J=3.4 Hz, 4H), 4.60-4.34 (m, 4H), 3.75 (q, J=8.7 Hz, 2H), 2.96 (s, 3H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 706.6 (M+H+). Example 241: (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-2-(2,3,6-trifluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-2-(2,3,6-trifluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1283 and I-1232. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.2 Hz, 1H), 8.36 (s, 1H), 8.01 (d, J=1.2 Hz, 1H), 7.97-7.89 (m, 2H), 7.89-7.74 (m, 3H), 7.64-7.58 (m, 1H), 7.51 (dd, J=11.2, 1.2 Hz, 1H), 7.04 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.11 (d, J=6.4 Hz, 1H), 4.67 (d, J=17.4 Hz, 1H), 4.56 (d, J=11.3 Hz, 1H), 4.46 (dd, J=11.3, 6.6 Hz, 1H), 4.38 (d, J=17.5 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 690.6 (M+H+). Example 242: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1232. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.93 (t, J=7.9 Hz, 1H), 7.76 (ddd, J=10.4, 5.7, 2.0 Hz, 1H), 7.65-7.57 (m, 2H), 7.51 (ddd, J=10.0, 5.3, 1.6 Hz, 2H), 7.33 (dd, J=8.3, 2.0 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.52 (s, 2H), 5.11 (d, J=6.4 Hz, 1H), 4.67 (d, J=17.4 Hz, 1H), 4.56 (d, J=11.3 Hz, 1H), 4.46 (dd, J=11.3, 6.7 Hz, 1H), 4.38 (d, J=17.4 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 659.5 (M+H+). Example 243: (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1248 and I-1232. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.93 (t, J=7.9 Hz, 1H), 7.73 (ddd, J=10.7, 5.6, 1.9 Hz, 1H), 7.57 (dd, J=7.4, 1.7 Hz, 1H), 7.55-7.49 (m, 3H), 7.46 (d, J=8.5 Hz, 2H), 7.01 (d, J=8.3 Hz, 1H), 5.49 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.67 (d, J=17.4 Hz, 1H), 4.56 (d, J=11.2 Hz, 1H), 4.46 (dd, J=11.3, 6.6 Hz, 1H), 4.37 (d, J=17.4 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 641.6 (M+H+). Example 244: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1232. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.96 (d, J=7.7 Hz, 1H), 7.92-7.85 (m, 2H), 7.74-7.62 (m, 3H), 7.61-7.56 (m, 1H), 7.54-7.46 (m, 1H), 7.06 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.66 (d, J=17.4 Hz, 1H), 4.55 (d, J=11.2 Hz, 1H), 4.46 (dd, J=11.3, 6.6 Hz, 1H), 4.37 (d, J=17.4 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 630.6 (M+H+). Example 245: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1231. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.95 (t, J=7.9 Hz, 1H), 7.88 (d, J=8.2 Hz, 2H), 7.78 (dd, J=8.4, 1.4 Hz, 1H), 7.69 (d, J=8.0 Hz, 2H), 7.68-7.61 (m, 1H), 7.58 (dd, J=8.2, 2.9 Hz, 2H), 7.07 (d, J=8.3 Hz, 1H), 5.59 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.64 (d, J=17.4 Hz, 1H), 4.57 (d, J=11.1 Hz, 1H), 4.47 (dd, J=11.2, 6.8 Hz, 1H), 4.35 (d, J=17.3 Hz, 1H), 3.82-3.72 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 612.6 (M+H+). Example 246: (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1248 and I-1231. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.93 (t, J=7.9 Hz, 1H), 7.78 (dd, J=8.4, 1.5 Hz, 1H), 7.71 (ddd, J=10.4, 5.6, 2.0 Hz, 1H), 7.63-7.50 (m, 4H), 7.46 (d, J=8.4 Hz, 2H), 7.02 (d, J=8.3 Hz, 1H), 5.49 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.64 (d, J=17.3 Hz, 1H), 4.57 (d, J=11.1 Hz, 1H), 4.47 (dd, J=11.2, 6.7 Hz, 1H), 4.35 (d, J=17.3 Hz, 1H), 3.83-3.73 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 623.6 (M+H+). Example 247: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1231. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 7.93 (t, J=7.9 Hz, 1H), 7.79 (dd, J=8.5, 1.4 Hz, 1H), 7.77-7.71 (m, 1H), 7.65-7.56 (m, 3H), 7.50 (dd, J=10.0, 2.0 Hz, 1H), 7.34 (dd, J=8.3, 2.0 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.52 (s, 2H), 5.10 (d, J=6.6 Hz, 1H), 4.66 (d, J=17.3 Hz, 1H), 4.58 (d, J=11.1 Hz, 1H), 4.47 (dd, J=11.2, 6.7 Hz, 1H), 4.37 (d, J=17.3 Hz, 1H), 3.84-3.69 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 641.6 (M+H+). Example 248: (S)-2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1022 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.92-7.78 (m, 3H), 7.63 (d, J=8.5 Hz, 1H), 7.56-7.41 (m, 4H), 7.35 (d, J=7.8 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.48 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.64 (q, J=2.5 Hz, 4H), 4.58-4.51 (m, 2H), 4.49-4.36 (m, 2H), 3.84-3.68 (m, 2H), 2.96 (s, 3H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 688.6 (M+H+). Example 249 (S)-2-(4-(6-((6-chloro-2-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-chloro-2-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 249) was made in a manner similar to that described in Procedure 27, substituting intermediates I-1219 and I-1319. Methyl (S)-2-(4-(6-((6-chloro-2-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (249-1): ES/MS m/z: 637.9 (M+H+). A mixture of methyl (S)-2-(4-(6-((6-chloro-2-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (249-1, 38 mg, 1.0 equivalent), lithium hydroxide (0.3 M in water, 0.60 mL, 3.0 equivalent) and acetonitrile (0.8 mL) was heated at 105° C. for 4 min. The mixture was quenched with acetic acid and purified by preparative reverse-phase HPLC (acetonitrile/water, 0.1% TFA) to yield the title compound. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 8.18 (dd, J=9.6, 7.8 Hz, 1H), 7.90 (t, J=7.9 Hz, 1H), 7.87-7.78 (m, 2H), 7.63 (d, J=8.5 Hz, 1H), 7.55 (dd, J=7.8, 2.5 Hz, 2H), 7.47 (dd, J=11.2, 6.3 Hz, 1H), 6.98 (d, J=8.3 Hz, 1H), 5.53 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.59-4.50 (m, 2H), 4.49-4.35 (m, 2H), 3.83-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 624.6 (M+H+). (S)-2-(4-(6-((6-chloro-2-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 249): 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 8.18 (dd, J=9.6, 7.8 Hz, 1H), 7.90 (t, J=7.9 Hz, 1H), 7.87-7.78 (m, 2H), 7.63 (d, J=8.5 Hz, 1H), 7.55 (dd, J=7.8, 2.5 Hz, 2H), 7.47 (dd, J=11.2, 6.3 Hz, 1H), 6.98 (d, J=8.3 Hz, 1H), 5.53 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.59-4.50 (m, 2H), 4.49-4.35 (m, 2H), 3.83-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 624.6 (M+H+). Example 250: 4-fluoro-1-(2-methoxyethyl)-2-(2,3,6-trifluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid 4-fluoro-1-(2-methoxyethyl)-2-(2,3,6-trifluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1282 and I-1136. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.11 (d, J=1.3 Hz, 1H), 8.03-7.90 (m, 3H), 7.89-7.73 (m, 3H), 7.59 (d, J=7.3 Hz, 1H), 7.50 (d, J=11.3 Hz, 1H), 7.03 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 4.68 (t, J=5.1 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J=4.9 Hz, 2H), 3.24 (s, 3H). ES/MS m/z: 650.6 (M+H+). Example 251: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with 4-chloro-2-fluorobenzyl bromide and Intermediate I-1136. 1H NMR (400 MHz, DMSO-d6) δ 8.11 (d, J=1.3 Hz, 1H), 7.93 (t, J=7.9 Hz, 1H), 7.77-7.69 (m, 1H), 7.64-7.56 (m, 2H), 7.54-7.46 (m, 2H), 7.33 (dd, J=8.2, 2.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.52 (s, 2H), 4.68 (t, J=5.1 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J=5.0 Hz, 2H), 3.24 (s, 3H). ES/MS m/z: 617.6 (M+H+). Procedure 37: Example 252 Methyl (S)-2-(2,5-difluoro-4-(6-((2-fluoro-6-methylpyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate. A mixture of methyl (S)-2-(4-(6-((6-chloro-2-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (249-1, from Example 249, 35 mg, 1.0 equivalent), methylboronic acid (32.9 mg, 10 equivalent), Pd(dppf)Cl2(6.1 mg, 0.15 equivalent), potassium carbonate (76 mg, 10 equivalent), and 1,4-dioxane (1.5 mL) was degassed by bubbling through argon for 2 min, then heated to 120° C. in a microwave reactor for 60 min. The mixture was filtered, concentrated in vacuo, and purified by silica gel flash column chromatography to yield methyl (S)-2-(2,5-difluoro-4-(6-((2-fluoro-6-methylpyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate. (S)-2-(2,5-difluoro-4-(6-((2-fluoro-6-methylpyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 252): Methyl (S)-2-(2,5-difluoro-4-(6-((2-fluoro-6-methylpyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (31 mg, 1.0 equivalent) was mixed with lithium hydroxide monohydrate (0.3 M in water, 500 μL, 3 equivalent) and acetonitrile (0.8 mL). All components were heated at 105° C. for 4 min. The mixture was neutralized with AcOH, diluted with DMSO, and purified by preparative HPLC (0-100% ACN in H2O, 0.1% TFA) to give the title compound. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.99 (dd, J=10.1, 7.5 Hz, 1H), 7.94-7.78 (m, 3H), 7.64 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.47 (dd, J=11.3, 6.2 Hz, 1H), 7.24 (dd, J=7.6, 1.9 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 5.48 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.62-4.51 (m, 2H), 4.51-4.35 (m, 2H), 3.81-3.69 (m, 2H), 2.43 (s, 3H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 602.6 (M+H+). Example 253: 2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1341 and I-1136. 1H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J=1.3 Hz, 1H), 8.76 (d, J=2.2 Hz, 1H), 8.26 (dd, J=8.4, 2.2 Hz, 1H), 8.18 (d, J=8.4 Hz, 1H), 8.11 (d, J=1.3 Hz, 1H), 8.03-7.91 (m, 2H), 7.81-7.72 (m, 1H), 7.59 (dd, J=7.5, 1.8 Hz, 1H), 7.54-7.46 (m, 1H), 7.05 (d, J=8.3 Hz, 1H), 5.63 (s, 2H), 4.68 (t, J=5.1 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J=5.0 Hz, 2H), 3.25 (s, 3H). ES/MS m/z: 633.6 (M+H+). Example 254: (S)-2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1268 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J=1.2 Hz, 1H), 8.76 (d, J=2.1 Hz, 1H), 8.49 (s, 1H), 8.26 (dd, J=8.4, 2.2 Hz, 1H), 8.17 (d, J=8.4 Hz, 1H), 8.00 (d, J=1.2 Hz, 1H), 7.96-7.85 (m, 2H), 7.81 (dd, J=8.4, 1.4 Hz, 1H), 7.62 (d, J=8.5 Hz, 1H), 7.55 (d, J=7.4 Hz, 1H), 7.47 (dd, J=10.4, 7.2 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.63 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.59-4.49 (m, 2H), 4.49-4.30 (m, 2H), 3.78 (d, J=8.7 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 638.2 (M+H+). Example 255: 2-(2-chloro-4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-5-fluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-5-fluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1248 and I-1278. 1H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J=1.3 Hz, 1H), 7.98 (d, J=7.2 Hz, 1H), 7.88 (t, J=7.9 Hz, 1H), 7.55-7.48 (m, 4H), 7.48-7.42 (m, 3H), 6.97 (d, J=8.2 Hz, 1H), 5.46 (s, 2H), 4.63 (t, J=5.1 Hz, 2H), 4.52 (s, 2H), 3.70 (t, J=5.0 Hz, 2H), 3.22 (s, 3H). ES/MS m/z: 598.0 (M+H+). Example 256: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-((4S)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-((4S)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1337. 1H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J=1.3 Hz, 1H), 7.95-7.86 (m, 2H), 7.75 (tdd, J=9.4, 7.2, 3.0 Hz, 3H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.48 (dd, J=11.2, 1.3 Hz, 1H), 7.31 (dd, J=11.5, 6.1 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.64-5.51 (m, 3H), 4.53 (s, 2H), 4.40 (dd, J=10.6, 3.4 Hz, 1H), 4.13 (td, J=8.8, 7.6, 3.7 Hz, 2H), 4.00 (dd, J=10.6, 8.2 Hz, 1H), 3.78 (dd, J=10.4, 4.6 Hz, 1H), 2.87 (s, 3H). ES/MS m/z: 633.0 (M+H+). Example 257: (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1132. 1H NMR (400 MHz, DMSO-d6) δ 8.51 (s, 1H), 7.97-7.87 (m, 3H), 7.82 (dd, J=8.5, 1.5 Hz, 1H), 7.78-7.70 (m, 2H), 7.63 (d, J=8.5 Hz, 1H), 7.58-7.45 (m, 2H), 7.02 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.63 (d, J=17.1 Hz, 1H), 4.55 (d, J=11.7 Hz, 1H), 4.49-4.36 (m, 2H), 3.81 (d, J=8.6 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 629.0 (M+H+). Example 258: 2-(2-chloro-5-fluoro-4-(6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-5-fluoro-4-(6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1016 and I-1278. 1H NMR (400 MHz, DMSO-d6) δ 8.10 (s, 1H), 8.01 (d, J=7.1 Hz, 1H), 7.87 (t, J=7.9 Hz, 1H), 7.56-7.39 (m, 5H), 7.38-7.30 (m, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.47 (s, 2H), 4.64 (t, J=7.8 Hz, 6H), 4.53 (s, 2H), 3.70 (t, J=5.1 Hz, 2H), 3.66 (d, J=2.4 Hz, 3H), 3.22 (s, 3H). ES/MS m/z: 663.0 (M+H+). Example 259: (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared via preparative chiral SFC (Daicel AD-H column, MeOH/CO2) of Example 268, as the later-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.57 (s, 1H), 7.98-7.83 (m, 3H), 7.81-7.71 (m, 3H), 7.67 (d, J=8.5 Hz, 1H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.40 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.27 (s, 1H), 4.47 (s, 2H), 4.37-4.23 (m, 2H), 4.05 (d, J=8.9 Hz, 1H), 3.91 (d, J=8.9 Hz, 1H), 0.75 (dd, J=24.6, 17.1 Hz, 3H), −0.07 (s, 1H). ES/MS m/z: 610.6 (M+H+). Example 260: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared via preparative chiral SFC (Daicel AD-H column, MeOH/CO2) of Example 268, as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.57 (s, 1H), 7.98-7.83 (m, 3H), 7.81-7.71 (m, 3H), 7.67 (d, J=8.5 Hz, 1H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.40 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.27 (s, 1H), 4.47 (s, 2H), 4.37-4.23 (m, 2H), 4.05 (d, J=8.9 Hz, 1H), 3.91 (d, J=8.9 Hz, 1H), 0.75 (dd, J=24.6, 17.1 Hz, 3H), −0.07 (s, 1H). ES/MS m/z: 610.6 (M+H+). Example 261: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3 S,4S)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared via preparative chiral SFC (Daicel AD-H column, EtOH/CO2) of Example 269, as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J=1.4 Hz, 1H), 7.96-7.82 (m, 3H), 7.82-7.70 (m, 3H), 7.67 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.41 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 6.41 (td, J=55.8, 4.9 Hz, 1H), 5.65-5.50 (m, 3H), 4.59 (d, J=17.0 Hz, 1H), 4.49 (t, J=8.9 Hz, 1H), 4.41 (d, J=16.9 Hz, 1H), 4.28 (dd, J=10.5, 3.3 Hz, 1H), 4.15 (dd, J=10.5, 7.9 Hz, 1H), 3.81 (t, J=9.2 Hz, 1H), 3.25 (d, J=14.7 Hz, 1H). ES/MS m/z: 635.2 (M+H+). Example 262: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared via preparative chiral SFC (Daicel AD-H column, EtOH/CO2) of Example 269, as the later-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J=1.4 Hz, 1H), 7.96-7.82 (m, 3H), 7.82-7.70 (m, 3H), 7.67 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.41 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 6.41 (td, J=55.8, 4.9 Hz, 1H), 5.65-5.50 (m, 3H), 4.59 (d, J=17.0 Hz, 1H), 4.49 (t, J=8.9 Hz, 1H), 4.41 (d, J=16.9 Hz, 1H), 4.28 (dd, J=10.5, 3.3 Hz, 1H), 4.15 (dd, J=10.5, 7.9 Hz, 1H), 3.81 (t, J=9.2 Hz, 1H), 3.25 (d, J=14.7 Hz, 1H). ES/MS m/z: 635.2 (M+H+). Example 263: (S)-2-(2,5-difluoro-4-(6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1016 and I-108. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.92-7.76 (m, 2H), 7.57-7.40 (m, 5H), 7.34 (dd, J=10.8, 7.9 Hz, 1H), 6.94 (d, J=8.3 Hz, 1H), 5.48 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.69-4.60 (m, 4H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 3.66 (d, J=3.3 Hz, 3H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 687.2 (M+H+). Example 264: 2-(2,5-difluoro-4-(6-((3-fluoro-5-(trifluoromethyl)pyridin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((3-fluoro-5-(trifluoromethyl)pyridin-2-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1129 and I-1336. 1H NMR (400 MHz, DMSO-d6) δ 8.85 (s, 1H), 8.37 (d, J=11.1 Hz, 2H), 7.89 (t, J=7.9 Hz, 1H), 7.60 (dd, J=10.6, 6.4 Hz, 1H), 7.56-7.48 (m, 2H), 7.44 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.71 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.58-4.48 (m, 2H), 4.46-4.33 (m, 2H), 3.74 (q, J=8.7 Hz, 2H), 1.32 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 675.2 (M+H+). Example 265: (S)-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1175 and I-1075. 1H NMR (400 MHz, DMSO-d6) δ 8.75 (d, J=5.1 Hz, 1H), 8.35 (s, 1H), 7.94 (d, J=10.0 Hz, 1H), 7.80-7.70 (m, 2H), 7.52 (dd, J=11.3, 1.2 Hz, 1H), 7.48 (d, J=5.1 Hz, 1H), 7.42 (d, J=10.5 Hz, 1H), 7.38 (d, J=7.5 Hz, 1H), 5.59 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.56-4.46 (m, 2H), 4.42 (dd, J=11.3, 6.7 Hz, 1H), 4.36 (d, J=16.9 Hz, 1H), 3.77 (d, J=8.7 Hz, 1H), 3.72 (d, J=8.6 Hz, 1H), 2.36 (s, 3H), 1.32 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 628.2 (M+H+). Example 266: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1126. 1H NMR (400 MHz, DMSO-d6) δ 8.39 (d, J=1.5 Hz, 1H), 7.96-7.81 (m, 3H), 7.81-7.71 (m, 3H), 7.68 (d, J=8.5 Hz, 1H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.44 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.06 (td, J=7.6, 3.6 Hz, 1H), 4.56 (d, J=16.9 Hz, 1H), 4.48 (d, J=16.8 Hz, 1H), 4.37 (t, J=8.2 Hz, 1H), 4.22 (dd, J=10.5, 3.6 Hz, 1H), 4.13 (dd, J=10.5, 8.1 Hz, 1H), 3.38 (t, J=9.4 Hz, 1H), 2.68 (dq, J=9.7, 6.7 Hz, 1H), 1.06 (d, J=6.7 Hz, 3H). ES/MS m/z: 599.2 (M+H+). Example 267: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((4R)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((4R)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1125. 1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.97-7.87 (m, 2H), 7.84-7.67 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.4, 1.7 Hz, 1H), 7.39 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.80-5.56 (m, 3H), 4.55 (d, J=12.6 Hz, 2H), 4.39 (d, J=17.0 Hz, 1H), 4.27-4.12 (m, 2H), 4.09 (t, J=9.2 Hz, 1H), 3.62-3.39 (m, 2H). ES/MS m/z: 635.2 (M+H+). Example 268: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(5-oxaspiro[2.4]heptan-7-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1124. 1H NMR (400 MHz, DMSO-d6) δ 8.57 (s, 1H), 7.98-7.83 (m, 3H), 7.81-7.71 (m, 3H), 7.67 (d, J=8.5 Hz, 1H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.40 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.27 (s, 1H), 4.47 (s, 2H), 4.37-4.23 (m, 2H), 4.05 (d, J=8.9 Hz, 1H), 3.91 (d, J=8.9 Hz, 1H), 0.75 (dd, J=24.6, 17.1 Hz, 3H), −0.07 (s, 1H). ES/MS m/z: 611.2 (M+H+). Example 269: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(difluoromethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1123. 1H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J=1.4 Hz, 1H), 7.96-7.82 (m, 3H), 7.82-7.70 (m, 3H), 7.67 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.41 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 6.41 (td, J=55.8, 4.9 Hz, 1H), 5.65-5.50 (m, 3H), 4.59 (d, J=17.0 Hz, 1H), 4.49 (t, J=8.9 Hz, 1H), 4.41 (d, J=16.9 Hz, 1H), 4.28 (dd, J=10.5, 3.3 Hz, 1H), 4.15 (dd, J=10.5, 7.9 Hz, 1H), 3.81 (t, J=9.2 Hz, 1H), 3.25 (d, J=14.7 Hz, 1H). ES/MS m/z. 635.2 (M+H+). Example 270: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((1s,4s)-1-methyl-2-oxabicyclo[2.1.1]hexan-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((1S,4S)-1-methyl-2-oxabicyclo[2.1.1]hexan-4-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1118. 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J=1.4 Hz, 1H), 7.95-7.87 (m, 2H), 7.84-7.74 (m, 4H), 7.64 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.38 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 4.39 (s, 2H), 3.99 (s, 2H), 2.63-2.55 (m, 2H), 1.51 (s, 3H). ES/MS m/z: 611.2 (M+H+). Example 271: 1-((3-aminotetrahydrofuran-3-yl)methyl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-((3-aminotetrahydrofuran-3-yl)methyl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 3, starting with Intermediates I-7 and I-1116. 1H NMR (400 MHz, DMSO-d6) δ 8.40 (s, 3H), 8.37 (s, 1H), 7.96-7.88 (m, 2H), 7.84 (dd, J=8.5, 1.4 Hz, 1H), 7.79-7.71 (m, 3H), 7.66 (d, J=8.4 Hz, 1H), 7.56-7.50 (m, 1H), 7.46 (dd, J=11.6, 6.1 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 4.80 (s, 2H), 4.41 (s, 2H), 4.00 (q, J=7.9 Hz, 1H), 3.97-3.88 (m, 1H), 3.88-3.79 (m, 2H), 2.37-2.12 (m, 2H). Example 272: racemic 1-(((5S)-2,6-dioxabicyclo[3.2.1]octan-1-yl)methyl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 1-(((5S)-2,6-dioxabicyclo[3.2.1]octan-1-yl)methyl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 5, starting with Intermediates I-7 and I-1115. 1H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J=1.5 Hz, 1H), 7.94-7.88 (m, 2H), 7.84 (dd, J=8.5, 1.5 Hz, 1H), 7.80-7.70 (m, 3H), 7.63 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.6, 1.7 Hz, 1H), 7.39 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.86-4.65 (m, 2H), 4.52 (d, J=4.7 Hz, 3H), 3.98 (d, J=9.3 Hz, 1H), 3.91 (d, J=9.3 Hz, 1H), 2.13 (dd, J=11.0, 6.4 Hz, 1H), 1.64-1.38 (m, 4H). ES/MS m/z: 641.2 (M+H+). Example 273: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2-cyclopropyltetrahydrofuran-2-yl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2-cyclopropyltetrahydrofuran-2-yl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 5, starting with Intermediates I-7 and I-1114. 1H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J=1.5 Hz, 1H), 7.95-7.83 (m, 3H), 7.80-7.71 (m, 3H), 7.64 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.43 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.62 (s, 2H), 4.57 (s, 2H), 3.75-3.58 (m, 2H), 1.99-1.86 (m, 2H), 1.86-1.72 (m, 1H), 1.66-1.51 (m, 1H), 1.12 (qd, J=8.1, 5.2 Hz, 1H), 0.31 (dq, J=9.1, 4.4, 3.9 Hz, 1H), 0.26-0.15 (m, 1H), 0.10-−0.12 (m, 2H). ES/MS m/z: 639.2 (M+H+). Example 274: 1-(5-oxaspiro[2.4]heptan-6-ylmethyl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-(5-oxaspiro[2.4]heptan-6-ylmethyl)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 5, starting with Intermediates I-7 and I-1113. 1H NMR (400 MHz, DMSO-d6) δ 8.32 (d, J=1.5 Hz, 1H), 7.95-7.83 (m, 3H), 7.80-7.71 (m, 3H), 7.65 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.43 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.67 (dd, J=15.2, 2.9 Hz, 1H), 4.62-4.46 (m, 3H), 4.40 (qd, J=7.1, 2.9 Hz, 1H), 3.67 (d, J=8.0 Hz, 1H), 3.51 (d, J=8.0 Hz, 1H), 2.03 (dd, J=12.4, 6.6 Hz, 1H), 1.78 (dd, J=12.3, 7.0 Hz, 1H), 0.70-0.45 (m, 4H). ES/MS m/z: 625.2 (M+H+). Example 275: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 276: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(cis-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate: N,N-Diisopropylethylamine (1.2 mL, 6.90 mmol) was added to a solution of 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetic acid (I-7, 550 mg, 1.38 mmol), methyl 4-amino-3-((cis-4-methoxytetrahydrofuran-3-yl)amino)benzoate (I-1112, 368 mg, 1.38 mmol), and o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (630 mg, 1.66 mmol) in DMF (10 mL). The solution was stirred at rt overnight. Following this time, the solution was diluted with EtOAc and washed with 5% LiCl, saturated NaHCO3, and brine. The organic extract was dried over sodium sulfate. The crude residue was purified by flash chromatography (eluent: EtOAc/hexanes). The resulting product was diluted with acetic acid (10 mL). The mixture was heated at 100° C. for 20 hr. Upon completion the mixture was concentrated and purified by flash chromatography (eluent: EtOAc/hexanes) to yield desired product. ES/MS: 629.2 (M+H+) 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(cis-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 1, GS-1157494) and 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(cis-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (later-eluting of two stereoisomers, Example 275): Lithium hydroxide, monohydrate (2.0 M, 1.30 mL, 1.30 mmol) was added to a solution of methyl 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(cis-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (400 mg, 0.636 mmol) in CH3CN (4.0 mL). The mixture was heated at 100° C. until completion (˜4 min). Next, the mixture was acidified with acetic acid and diluted with DMSO (2 mL). The mixture was purified by RP-HPLC (eluent: MeCN/H2O 0.1% TFA). The resulting product fractions were combined and lyophilized, and subsequent separation using preparative chiral SFC (Chiralpak AD-H column with MeOH/CO2) to give two distinct stereoisomers. 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(cis-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 1, Earlier eluting of two stereoisomers, Example 276): 1H NMR (400 MHz, DMSO-d6) δ 8.39 (d, J=1.5 Hz, 1H), 7.98-7.84 (m, 2H), 7.80-7.68 (m, 4H), 7.58 (d, J=8.4 Hz, 1H), 7.53 (d, J=6.8 Hz, 1H), 7.29 (dd, J=11.6, 6.0 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 5.54 (t, J=9.6 Hz, 1H), 4.50 (s, 2H), 4.42 (dd, J=10.4, 3.7 Hz, 1H), 4.19-4.08 (m, 2H), 4.02 (t, J=9.4 Hz, 1H), 3.82 (dd, J=10.3, 4.5 Hz, 1H), 2.87 (s, 3H). ES/MS: 615.2 (M+H+). 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(cis-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 2, later eluting of two stereoisomers, Example 275) 1H NMR (400 MHz, DMSO-d6) δ 8.38 (s, 1H), 7.97-7.86 (m, 2H), 7.75 (dt, J=6.5, 5.2 Hz, 4H), 7.60-7.43 (m, 2H), 7.29 (dd, J=11.4, 6.1 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.53 (d, J=10.2 Hz, 1H), 4.49 (s, 2H), 4.42 (dd, J=10.3, 3.8 Hz, 1H), 4.17-4.06 (m, 2H), 4.02 (dd, J=10.3, 8.3 Hz, 1H), 3.82 (dd, J=10.1, 4.5 Hz, 1H), 2.87 (s, 3H). ES/MS: 615.2 (M+H+). Example 277: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1182. 1H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.21 (dd, J=8.6, 1.4 Hz, 1H), 8.06 (d, J=8.2 Hz, 2H), 7.83 (d, J=8.6 Hz, 1H), 7.77 (t, J=7.5 Hz, 1H), 7.68-7.52 (m, 4H), 7.49 (d, J=8.1 Hz, 2H), 5.73 (s, 2H), 5.10 (d, J=6.2 Hz, 1H), 4.80-4.62 (m, 2H), 4.62-4.53 (m, 1H), 4.45 (dd, J=11.6, 6.7 Hz, 1H), 3.99 (d, J=9.0 Hz, 1H), 3.80 (d, J=8.9 Hz, 1H), 1.28 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 595.2 (M+H+). Example 278: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1076. 1H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.21 (dd, J=8.6, 1.4 Hz, 1H), 7.93-7.78 (m, 2H), 7.70 (t, J=7.6 Hz, 1H), 7.61-7.53 (m, 2H), 7.38 (d, J=7.6 Hz, 1H), 7.25 (d, J=10.8 Hz, 1H), 7.17 (d, J=7.4 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.57 (s, 2H), 5.15 (d, J=6.5 Hz, 1H), 4.79-4.62 (m, 3H), 4.52 (dd, J=11.7, 6.6 Hz, 1H), 4.01 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 2.31 (s, 3H), 1.37 (s, 3H), 0.73 (s, 3H). ES/MS m/z: 609.3 (M+H+). Example 279: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1076. 1H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J=8.6, 1.4 Hz, 1H), 7.80 (d, J=8.6 Hz, 1H), 7.73 (t, J=7.6 Hz, 1H), 7.66 (dd, J=10.1, 8.1 Hz, 1H), 7.64-7.58 (m, 2H), 7.37 (d, J=7.6 Hz, 1H), 7.26 (d, J=10.8 Hz, 1H), 7.19 (dd, J=8.1, 2.8 Hz, 1H), 5.64 (s, 2H), 5.13 (d, J=6.4 Hz, 1H), 4.77-4.59 (m, 3H), 4.51 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=9.0 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 2.31 (s, 3H), 1.37 (s, 3H), 0.72 (s, 3H). ES/MS m/z: 627.3 (M+H+). Example 280: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1076. 1H NMR (400 MHz, Methanol-d4) δ 8.94 (s, 1H), 8.23 (dd, J=8.6, 1.4 Hz, 1H), 7.83 (t, J=8.0 Hz, 2H), 7.51 (t, J=8.3 Hz, 1H), 7.41 (d, J=7.6 Hz, 1H), 7.30 (d, J=10.8 Hz, 1H), 7.28-7.19 (m, 2H), 7.16 (d, J=7.3 Hz, 1H), 6.91 (d, J=8.3 Hz, 1H), 5.46 (s, 2H), 5.17 (d, J=7.0 Hz, 1H), 4.81-4.58 (m, 3H), 4.52 (dd, J=11.7, 6.7 Hz, 1H), 4.01 (d, J=8.9 Hz, 1H), 3.85 (d, J=9.0 Hz, 1H), 2.36 (s, 3H), 1.38 (s, 3H), 0.74 (s, 3H). ES/MS m/z: 618.25 (M+H+). Example 281: 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid Procedure 38 Tert-butyl 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (281-1): A mixture of tert-butyl 2-[(4-bromo-2,3,6-trifluoro-phenyl)methyl]-3-(2-methoxyethyl)benzimidazole-5-carboxylate (160 mg, 1.0 equivalent), [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (I-1186, 24 mg, 0.10 equivalent), potassium propionate (108 mg, 3.0 equivalent), and Bis(pinacolato)diboron (98 mg, 1.2 equivalent) in dioxane (3 mL) was degassed with Ar for 5 min. Upon completion the mixture was then heated at 110° C. for 1.5 h with a reflux condenser under Argon. Next, sodium carbonate (2 M in water, 0.32 mL, 2 equivalent) was added and the mixture stirred for 5 min. Upon completion 4-[(6-bromo-2-pyridyl)oxymethyl]benzonitrile (I-114, 139 mg, 1.5 equivalent) and [1,1′-Bis(diphenylphosphino)ferrocene] dichloropalladium(II) (12 mg, 0.05 equivalent) were added to the mixture. The resulting mixture was again degassed for 5 min with Argon, then heated at 80° C. for 2 hr. Upon completion, the mixture was filtered through Celite, concentrated in vacuo, and purified by silica gel flash column chromatography (ethyl acetate/hexane) to yield tert-butyl 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (281-1). 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 281). Tert-butyl 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylate (281-1, 100 mg) in 1 mL of DCM and 0.4 mL of TFA was stirred for 4 hours at room temperature. The mixture was concentrated in vacuo and purified by preparative HPLC (MeCN in water, 0.1% TFA) to give the title compound. 1H NMR (400 MHz, Methanol-d4) δ 8.67 (s, 1H), 7.95 (dd, J=8.6, 1.5 Hz, 1H), 7.82-7.64 (m, 3H), 7.59 (d, J=8.5 Hz, 1H), 5.82 (s, 2H), 5.08 (d, J=6.8 Hz, 1H), 4.72 (d, J=11.0 Hz, 1H), 4.68-4.51 (m, 4H), 4.42 (d, J=17.1 Hz, 1H), 3.99 (d, J=8.7 Hz, 1H), 3.84 (d, J=8.7 Hz, 1H), 1.50-1.40 (m, 6H), 0.77 (s, 3H). ES/MS (m/z): 573.6 (M+H+). Example 282: (S)-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1246 and I-1231. 1H NMR (400 MHz, Methanol-d4) δ 8.83 (s, 1H), 8.09 (dd, J=8.5, 1.4 Hz, 1H), 7.73-7.64 (m, 3H), 7.64-7.55 (m, 1H), 7.51 (d, J=8.4 Hz, 2H), 7.47-7.20 (m, 2H), 5.57 (s, 2H), 5.16 (d, J=6.6 Hz, 1H), 4.81-4.63 (m, 2H), 4.57 (dd, J=11.5, 6.7 Hz, 2H), 3.99 (d, J=8.8 Hz, 1H), 3.86 (d, J=8.8 Hz, 1H), 1.48 (s, 3H), 0.81 (s, 3H). ES/MS m/z: 640.5 (M+H+). Example 283: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1247 and I-1231. 1H NMR (400 MHz, Methanol-d4) δ 8.83 (s, 1H), 8.09 (dd, J=8.6, 1.4 Hz, 1H), 7.84-7.74 (m, 2H), 7.74-7.66 (m, 4H), 7.66-7.51 (m, 2H), 5.68 (s, 2H), 5.16 (d, J=6.6 Hz, 1H), 4.80-4.66 (m, 2H), 4.57 (dd, J=11.6, 6.7 Hz, 2H), 3.99 (d, J=8.9 Hz, 1H), 3.86 (d, J=8.9 Hz, 1H), 1.48 (s, 3H), 0.81 (s, 3H). ES/MS m/z: 631.5 (M+H+). Example 284: (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-105 and I-1076. 1H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.71 (d, J=5.1 Hz, 1H), 8.19 (dd, J=8.6, 1.4 Hz, 1H), 7.79 (d, J=8.6 Hz, 1H), 7.58 (t, J=8.2 Hz, 1H), 7.44 (dd, J=14.0, 9.0 Hz, 2H), 7.34 (d, J=5.1 Hz, 1H), 7.31-7.20 (m, 2H), 5.57 (s, 2H), 5.15 (d, J=6.2 Hz, 1H), 4.79-4.58 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 2.43 (s, 3H), 1.39 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 619.2 (M+H+). Example 285: (S)-2-(2,5-difluoro-4-(2-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidin-4-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(2-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyrimidin-4-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1091 and I-82. 1H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.73 (d, J=5.2 Hz, 1H), 8.13 (dd, J=8.5, 1.4 Hz, 1H), 8.03 (dd, J=10.4, 6.1 Hz, 1H), 7.82 (t, J=7.5 Hz, 1H), 7.74 (d, J=8.6 Hz, 1H), 7.69 (dd, J=5.2, 1.7 Hz, 1H), 7.56 (d, J=7.9 Hz, 2H), 7.45 (dd, J=11.2, 5.9 Hz, 1H), 5.71 (s, 2H), 5.11 (d, J=6.6 Hz, 1H), 4.79-4.58 (m, 3H), 4.52 (dd, J=11.5, 6.8 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 657.3 (M+H+). Example 286: 2-(4-(6-((2-chloro-6-methylpyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((2-chloro-6-methylpyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1199. 1H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J=8.6, 1.4 Hz, 1H), 7.93-7.80 (m, 3H), 7.78 (d, J=8.6 Hz, 1H), 7.59 (dd, J=7.4, 1.6 Hz, 1H), 7.40 (dd, J=11.2, 6.0 Hz, 1H), 7.27 (d, J=7.8 Hz, 1H), 6.98 (d, J=8.2 Hz, 1H), 5.57 (s, 2H), 5.15 (d, J=6.4 Hz, 1H), 4.75 (d, J=17.3 Hz, 1H), 4.71-4.62 (m, 2H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 2.51 (s, 3H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 619.4 (M+H+). Example 287: (S)-2-(4-(6-((5-chloro-3-fluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((5-chloro-3-fluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1217 and I-108. 1H NMR (400 MHz, Methanol-d4) δ 8.56 (s, 1H), 8.43 (d, J=1.9 Hz, 1H), 7.93-7.74 (m, 3H), 7.68 (dd, J=11.1, 1.2 Hz, 1H), 7.60-7.50 (m, 1H), 7.21 (dd, J=11.5, 6.0 Hz, 1H), 6.89 (d, J=8.2 Hz, 1H), 5.63 (d, J=1.9 Hz, 2H), 4.95 (d, J=7.3 Hz, 1H), 4.62-4.44 (m, 4H), 3.94 (d, J=8.8 Hz, 1H), 3.80 (d, J=8.8 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 641.3 (M+H+). Example 288: (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1218 and I-108. 1H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 8.29 (s, 1H), 7.88 (dd, J=10.8, 6.3 Hz, 1H), 7.84-7.75 (m, 1H), 7.70 (dd, J=11.0, 1.2 Hz, 1H), 7.58-7.50 (m, 1H), 7.29-7.23 (m, 1H), 7.22 (d, J=11.4 Hz, 1H), 6.89 (dd, J=8.3, 0.7 Hz, 1H), 5.58-5.44 (m, 2H), 4.98 (d, J=6.7 Hz, 1H), 4.66-4.35 (m, 4H), 4.02 (s, 3H), 3.94 (d, J=8.8 Hz, 1H), 3.80 (d, J=8.8 Hz, 1H), 1.36 (s, 3H), 0.68 (s, 3H). ES/MS m/z: 653.5 (M+H+). Example 289A: (S)-2-(4-(6-((5-chloro-3-fluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 289B: (R)-2-(4-(6-((5-chloro-3-fluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 289A and 289B were prepared by preparative chiral SFC (Daicel Chiralpak AD-H column, EtOH/CO2) of Example 296. (S)-2-(4-(6-((5-chloro-3-fluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 289A) was isolated as the later-eluting of two stereoisomers. 1H NMR (400 MHz, Methanol-d4) δ 8.81 (s, 1H), 8.43 (d, J=1.9 Hz, 1H), 8.09 (dd, J=8.6, 1.4 Hz, 1H), 7.95-7.78 (m, 3H), 7.74 (d, J=8.6 Hz, 1H), 7.58 (dd, J=7.6, 1.5 Hz, 1H), 7.31 (dd, J=11.4, 6.0 Hz, 1H), 6.91 (d, J=8.3 Hz, 1H), 5.63 (d, J=1.9 Hz, 2H), 5.06 (d, J=6.6 Hz, 1H), 4.74-4.43 (m, 4H), 3.98 (d, J=8.8 Hz, 1H), 3.83 (d, J=8.8 Hz, 1H), 1.40 (s, 3H), 0.72 (s, 3H). ES/MS m/z: 623.7 (M+H+). (R)-2-(4-(6-((5-chloro-3-fluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 289B) was isolated as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.43 (d, J=1.9 Hz, 1H), 8.18 (dd, J=8.6, 1.4 Hz, 1H), 7.91 (dd, J=10.9, 6.3 Hz, 1H), 7.88-7.74 (m, 3H), 7.59 (dd, J=7.4, 1.5 Hz, 1H), 7.38 (dd, J=11.2, 6.1 Hz, 1H), 6.92 (d, J=8.2 Hz, 1H), 5.63 (d, J=1.9 Hz, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.80-4.61 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 623.7 (M+H+). Example 290A: (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 290B: (R)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 289A and 289B were prepared by preparative chiral SFC (Daicel Chiralpak AD-H column, EtOH/CO2) of Example 296. (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 290A) was isolated as the later-eluting of two stereoisomers. 1H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.28 (s, 1H), 8.16 (dd, J=8.6, 1.4 Hz, 1H), 7.93 (dd, J=10.8, 6.3 Hz, 1H), 7.85-7.72 (m, 2H), 7.58 (dd, J=7.4, 1.5 Hz, 1H), 7.39 (dd, J=11.2, 6.0 Hz, 1H), 7.18 (s, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 5.13 (d, J=6.6 Hz, 1H), 4.76-4.58 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (s, 4H), 3.84 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 636.1 (M+H+). (R)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 290B) was isolated as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.28 (s, 1H), 8.16 (dd, J=8.6, 1.4 Hz, 1H), 7.93 (dd, J=10.8, 6.3 Hz, 1H), 7.85-7.72 (m, 2H), 7.58 (dd, J=7.4, 1.5 Hz, 1H), 7.39 (dd, J=11.2, 6.0 Hz, 1H), 7.18 (s, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 5.13 (d, J=6.6 Hz, 1H), 4.76-4.58 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (s, 4H), 3.84 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 636.1 (M+H+). Example 291: 2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1136. 1H NMR (400 MHz, Methanol-d4) δ 8.28 (s, 1H), 8.19 (d, J=1.2 Hz, 1H), 7.86 (dd, J=10.8, 6.3 Hz, 1H), 7.79 (t, J=7.9 Hz, 1H), 7.72 (dd, J=11.1, 1.2 Hz, 1H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.20 (q, J=6.2 Hz, 2H), 6.88 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 4.63 (t, J=5.0 Hz, 2H), 4.57 (s, 2H), 4.01 (s, 3H), 3.75 (t, J=4.9 Hz, 2H), 3.28 (s, 3H). ES/MS m/z: 613.4 (M+H+). Example 292: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-116 and I-1032. 1H NMR (400 MHz, Methanol-d4) δ 8.15 (d, J=1.3 Hz, 1H), 7.86 (t, J=7.9 Hz, 1H), 7.75 (t, J=7.5 Hz, 1H), 7.70-7.56 (m, 5H), 6.97 (d, J=8.3 Hz, 1H), 5.64 (s, 2H), 4.67 (t, J=5.0 Hz, 2H), 4.60 (s, 2H), 3.81 (t, J=4.9 Hz, 2H). ES/MS m/z: 609.3 (M+H+). Example 293: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-116 and I-25. 1H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.11 (dd, J=8.6, 1.4 Hz, 1H), 7.89 (t, J=7.9 Hz, 1H), 7.82-7.66 (m, 3H), 7.66-7.50 (m, 3H), 7.01 (d, J=8.3 Hz, 1H), 5.64 (s, 2H), 5.17 (d, J=6.5 Hz, 1H), 4.85-4.50 (m, 4H), 4.00 (d, J=8.9 Hz, 1H), 3.87 (d, J=8.9 Hz, 1H), 1.48 (s, 3H), 0.82 (s, 3H). ES/MS m/z: 631.3 (M+H+). Example 294: 2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1199. 1H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.28 (s, 1H), 8.16 (dd, J=8.6, 1.4 Hz, 1H), 7.93 (dd, J=10.8, 6.3 Hz, 1H), 7.85-7.72 (m, 2H), 7.58 (dd, J=7.4, 1.5 Hz, 1H), 7.39 (dd, J=11.2, 6.0 Hz, 1H), 7.18 (s, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 5.13 (d, J=6.6 Hz, 1H), 4.76-4.58 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (s, 4H), 3.84 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 636.1 (M+H+). Example 295: 2-(4-(6-((6-chloro-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-chloro-4-fluoropyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1199. 1H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.57 (d, J=9.6 Hz, 1H), 8.19 (dd, J=8.6, 1.4 Hz, 1H), 7.95 (dd, J=10.8, 6.3 Hz, 1H), 7.89-7.75 (m, 2H), 7.59 (dd, J=7.5, 1.6 Hz, 1H), 7.41 (dd, J=10.1, 4.8 Hz, 2H), 6.93 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.15 (d, J=6.6 Hz, 1H), 4.81-4.61 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 623.7 (M+H+). Example 296: 2-(4-(6-((5-chloro-3-fluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((5-chloro-3-fluoropyridin-2-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1199. 1H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.43 (d, J=1.9 Hz, 1H), 8.18 (dd, J=8.6, 1.4 Hz, 1H), 7.91 (dd, J=10.9, 6.3 Hz, 1H), 7.88-7.74 (m, 3H), 7.59 (dd, J=7.4, 1.5 Hz, 1H), 7.38 (dd, J=11.2, 6.1 Hz, 1H), 6.92 (d, J=8.2 Hz, 1H), 5.63 (d, J=1.9 Hz, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.80-4.61 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 623.7 (M+H+). Example 297: (S)-2-((6-((4-chlorobenzyl)oxy)-5′-fluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((6-((4-chlorobenzyl)oxy)-5′-fluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1237 and I-1248. 1H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.24 (d, J=6.9 Hz, 1H), 7.92 (t, J=7.9 Hz, 1H), 7.81 (d, J=8.4 Hz, 1H), 7.66 (d, J=8.5 Hz, 1H), 7.57-7.49 (m, 3H), 7.45 (d, J=8.4 Hz, 2H), 7.06 (d, J=8.3 Hz, 1H), 6.97 (d, J=7.4 Hz, 1H), 5.55 (d, J=16.0 Hz, 1H), 5.46 (d, J=8.7 Hz, 3H), 5.10 (d, J=6.6 Hz, 1H), 4.61-4.39 (m, 2H), 3.83-3.69 (m, 2H), 1.38 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 603.09 (M+H+). Example 298: (S)-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1230 and I-94. 1H NMR (400 MHz, DMSO) δ 8.75 (d, J=5.2 Hz, 1H), 8.35 (s, 1H), 8.06 (d, J=9.4 Hz, 2H), 7.95 (dd, J=10.0, 1.5 Hz, 1H), 7.90-7.69 (m, 3H), 7.65-7.46 (m, 2H), 5.64 (s, 2H), 5.01 (d, J=6.5 Hz, 1H), 4.73-4.37 (m, 4H), 3.74 (q, J=8.7 Hz, 2H), 1.31 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 614.38 (M+H+). Example 299: (S)-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1246 and I-82. 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 8.10-7.72 (m, 3H), 7.72-7.40 (m, 8H), 5.57 (s, 2H), 5.02 (d, J=6.5 Hz, 1H), 4.82-4.27 (m, 4H), 3.85-3.64 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 622.17 (M+H+). Example 300: (S)-2-((6-((4-chlorobenzyl)oxy)-5,5′-difluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((6-((4-chlorobenzyl)oxy)-5,5′-difluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1237 and I-1246. 1H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.25 (d, J=7.1 Hz, 1H), 8.04-7.76 (m, 2H), 7.74-7.40 (m, 6H), 6.95 (d, J=7.5 Hz, 1H), 5.50 (d, J=28.8 Hz, 4H), 5.10 (d, J=6.7 Hz, 1H), 4.70-4.34 (m, 2H), 3.73 (d, J=8.6 Hz, 2H), 1.38 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 621.09 (M+H+). Example 301: (S)-2-(2-chloro-4-(2-((4-cyanobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(2-((4-cyanobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1030 and I-1248. 1H NMR (400 MHz, DMSO) δ 8.75 (d, J=5.1 Hz, 1H), 8.50 (s, 1H), 8.00-7.74 (m, 3H), 7.74-7.53 (m, 4H), 7.53-7.28 (m, 2H), 5.57 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.66-4.29 (m, 4H), 3.93-3.59 (m, 2H), 2.33 (s, 3H), 1.34 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 608.29 (M+H+). Example 302: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1233 and I-102. 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.88 (dd, J=8.2, 5.1 Hz, 3H), 7.82-7.71 (m, 2H), 7.71-7.44 (m, 4H), 7.34 (dd, J=8.2, 2.0 Hz, 1H), 6.95 (d, J=8.2 Hz, 1H), 5.55 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.58 (d, J=18.1 Hz, 2H), 4.47 (dd, J=11.2, 6.7 Hz, 1H), 4.32 (d, J=17.3 Hz, 1H), 3.86-3.71 (m, 2H), 1.38 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 622.2 (M+H+). Example 303: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1233 and I-1036. 1H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.93-7.75 (m, 5H), 7.65 (t, J=8.2 Hz, 1H), 7.61-7.50 (m, 2H), 7.36 (dd, J=8.2, 2.1 Hz, 1H), 5.64 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.64-4.42 (m, 3H), 4.30 (d, J=17.3 Hz, 1H), 3.76 (t, J=6.4 Hz, 2H), 1.38 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 640.13 (M+H+). Example 305: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1234 and I-102. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.89 (dd, J=9.8, 8.3 Hz, 3H), 7.76 (d, J=7.5 Hz, 1H), 7.63 (t, J=8.2 Hz, 1H), 7.55-7.43 (m, 2H), 7.33 (dd, J=8.2, 2.1 Hz, 1H), 6.94 (d, J=8.1 Hz, 1H), 5.55 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.71-4.49 (m, 2H), 4.46 (dd, J=11.3, 6.6 Hz, 1H), 4.32 (d, J=17.3 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 642.72 (M+H+). Example 306: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1234 and I-1036. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.93-7.74 (m, 4H), 7.65 (t, J=8.2 Hz, 1H), 7.51 (ddd, J=11.1, 8.9, 1.6 Hz, 2H), 7.36 (dd, J=8.2, 2.1 Hz, 1H), 5.64 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.71-4.39 (m, 3H), 4.31 (d, J=17.4 Hz, 1H), 3.76 (s, 2H), 1.38 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 658.85 (M+H+). Example 307: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1234 and I-109. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 8.02-7.69 (m, 7H), 7.53-7.47 (m, 1H), 5.74 (s, 2H), 5.11 (d, J=6.4 Hz, 1H), 4.74-4.50 (m, 2H), 4.46 (dd, J=11.1, 6.6 Hz, 1H), 4.31 (d, J=17.3 Hz, 1H), 3.76 (s, 2H), 1.38 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 649.36 (M+H+). Example 308: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1230 and I-1092. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.98-7.79 (m, 6H), 7.72 (d, J=3.2 Hz, 1H), 7.60-7.42 (m, 2H), 7.19 (dd, J=8.2, 2.6 Hz, 3H), 5.69 (s, 2H), 5.01 (s, 1H), 4.67-4.27 (m, 4H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 613.21 (M+H+). Example 309: (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-2-(2-fluoro-4-(6-((5-methoxy-1,3,4-thiadiazol-2-yl)methoxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-2-(2-fluoro-4-(6-((5-methoxy-1,3,4-thiadiazol-2-yl)methoxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1230 and I-1244. 1H NMR (400 MHz, DMSO) δ 8.18 (s, 1H), 8.00 (dd, J=15.7, 9.9 Hz, 2H), 7.89 (t, J=7.8 Hz, 1H), 7.74 (d, J=7.7 Hz, 1H), 7.48 (t, J=8.7 Hz, 2H), 6.92 (d, J=8.2 Hz, 1H), 5.75 (s, 2H), 4.93 (s, 1H), 4.63-4.22 (m, 2H), 4.09 (s, 3H), 3.84-3.65 (m, 2H), 1.26 (s, 3H), 0.56 (s, 3H). ES/MS m/z: 608.25 (M+H+). Example 310: (S)-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1232 and I-1246. 1H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.90 (dd, J=10.2, 8.2 Hz, 1H), 7.70 (td, J=6.3, 3.3 Hz, 1H), 7.64-7.45 (m, 6H), 5.58 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.67 (d, J=17.4 Hz, 1H), 4.61-4.30 (m, 3H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 658.14 (M+H+). Example 311A: 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[4-methoxy-4-methyl-tetrahydrofuran-3-yl]benzimidazole-5-carboxylic acid Example 311B: 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[4-methoxy-4-methyl-tetrahydrofuran-3-yl]benzimidazole-5-carboxylic acid Procedure 39 Methyl 4-[[2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetyl]amino]-3-[[4-methoxy-4-methyl-tetrahydrofuran-3-yl]amino]benzoate (311-1): A mixture of 2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetic acid (I-7, 145 mg, 0.365 mmol), racemic cis methyl 4-amino-3-[3-4-methoxy-4-methyl-tetrahydrofuran-3-yl]amino]benzoate (Intermediate I-1253 cis-isomer 1, 93 mg, 0.332 mmol), o-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (176 mg, 0.342 mmol) and N,N-diisopropylethylamine (0.28 mL, 1.62 mmol) in DMF (1.5 mL) and CAN (1.5 mL) was stirred at 25° C. for 5 h. The mixture was diluted in EtOAc and washed with brine, dried over sodium sulfate, concentrated, and purified by silica gel flash column chromatography (EtOAc/hexanes) to give the title compound. ES/MS: 661.26 (M+H+). Methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[4-methoxy-4-methyl-tetrahydrofuran-3-yl]benzimidazole-5-carboxylate (311-2): A mixture of methyl 4-[[2-[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]acetyl]amino]-3-[[3-4-methoxy-4-methyl-tetrahydrofuran-3-yl]amino]benzoate (311-1, 226 mg, 0.34 mmol), triphenylphosphine oxide (285 mg, 1.0 mmol) and triflic anhydride (0.086 mL, 0.51 mmol) in DCM (5.0 mL) was stirred at 0° C. for 10 min then warmed to 25° C. and stirred for 1 h. The mixture was diluted in EtOAc and washed with brine, dried over sodium sulfate, concentrated, and purified by silica gel flash column chromatography (EtOAc/hexanes) to give the title compound. ES/MS: 643.7 (M+H+). 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[4-methoxy-4-methyl-tetrahydrofuran-3-yl]benzimidazole-5-carboxylic acid (311-3): A mixture in 3 mL of methyl 2-[[4-[6-[(4-cyano-2-fluoro-phenyl)methoxy]-2-pyridyl]-2,5-difluoro-phenyl]methyl]-3-[4-methoxy-4-methyl-tetrahydrofuran-3-yl]benzimidazole-5-carboxylate (311-2, 114 mg, 0.177 mmol) in acetonitrile and 1 mL of water was added aqueous LiOH (1.8 mL, 0.53 mmol, 0.3 M). The mixture was then stirred at 100° C. for 5 min. Upon completion the mixture was cooled to rt, filtered through celite and concentrated. The crude material was purified by reverse phase chromatography (ACN/water with 0.1% TFA added) to yield the title compound as a racemic mixture. ES/MZ: 629.3 (M+H+). Relative stereochemistry was confirmed by . . . . Racemic 311-3 was purified by preparative chiral SFC (Daicel Chiralpak AD-H column, EtOH—NH3—CO2) to yield Example 311A and Example 311B as separate stereoisomers. Example 311A: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was isolated as the later eluting of two stereoisomers. 1H NMR (400 MHz, DMSO) δ 8.44 (s, 1H), 7.98-7.84 (m, 2H), 7.77 (ddd, J=15.0, 8.4, 1.6 Hz, 5H), 7.68-7.48 (m, 3H), 7.37 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.29 (s, 1H), 4.61-4.33 (m, 3H), 4.28-4.14 (m, 2H), 2.96 (s, 3H), 1.43 (s, 3H). ES/MS m/z: 629.32 (M+H+). Example 311B: was isolated as the earlier eluting of two stereoisomers. 1H NMR (400 MHz, DMSO) δ 8.44 (s, 1H), 7.98-7.84 (m, 2H), 7.77 (ddd, J=15.0, 8.4, 1.6 Hz, 5H), 7.68-7.48 (m, 3H), 7.37 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.29 (s, 1H), 4.61-4.33 (m, 3H), 4.28-4.14 (m, 2H), 2.96 (s, 3H), 1.43 (s, 3H). ES/MS m/z: 629.32 (M+H+). Example 312: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 313: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 312 and 313 were prepared in a manner as described in Procedure 39, starting with Intermediates I-7 and I-1253 trans-isomer 2, and using preparative chiral SFC (Daicel Chiralpak IG column, EtOH—CO2) to yield Example 312 and Example 313 as separate stereoisomers. Example 312: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was isolated as the later eluting of two stereoisomers. 1H NMR (400 MHz, DMSO) δ 8.46 (s, 1H), 7.99-7.85 (m, 2H), 7.85-7.69 (m, 4H), 7.64 (d, J=8.5 Hz, 1H), 7.55 (d, J=7.3 Hz, 1H), 7.46 (dd, J=10.4, 7.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.23 (d, J=6.8 Hz, 1H), 4.82-4.25 (m, 4H), 4.13 (d, J=10.1 Hz, 1H), 3.80 (d, J=10.1 Hz, 1H), 3.31 (s, 3H), 0.84 (s, 3H). ES/MS m/z: 629.32 (M+H+). Example 313: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was isolated as the earlier eluting of two stereoisomers. 1H NMR (400 MHz, DMSO) δ 8.46 (s, 1H), 7.99-7.85 (m, 2H), 7.85-7.69 (m, 4H), 7.64 (d, J=8.5 Hz, 1H), 7.55 (d, J=7.3 Hz, 1H), 7.46 (dd, J=10.4, 7.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.23 (d, J=6.8 Hz, 1H), 4.82-4.25 (m, 4H), 4.13 (d, J=10.1 Hz, 1H), 3.80 (d, J=10.1 Hz, 1H), 3.31 (s, 3H), 0.84 (s, 3H). ES/MS m/z: 629.32 (M+H+). Example 316: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1232 and I-1092. 1H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.92 (dd, J=19.4, 8.4 Hz, 3H), 7.72 (d, J=8.0 Hz, 2H), 7.68-7.56 (m, 2H), 7.51 (d, J=11.2 Hz, 1H), 5.68 (s, 2H), 5.11 (d, J=6.4 Hz, 1H), 4.82-4.31 (m, 3H), 3.76 (s, 2H), 0.66 (s, 3H). ES/MS m/z: 649.31 (M+H+). Example 317: (S)-2-(2-chloro-4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1031 and I-1241. 1H NMR (400 MHz, DMSO) δ 8.74 (d, J=5.1 Hz, 1H), 8.36 (s, 1H), 7.69-7.57 (m, 2H), 7.57-7.45 (m, 3H), 7.41 (s, 1H), 7.35 (dd, J=8.3, 2.1 Hz, 1H), 5.02 (d, J=6.7 Hz, 1H), 4.64-4.31 (m, 4H), 3.81-3.67 (m, 2H), 1.33 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 653.73 (M+H+). Example 318: (S)-2-(2-chloro-4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1030 and I-1241. 1H NMR (400 MHz, DMSO) δ 8.75 (d, J=5.1 Hz, 1H), 8.36 (s, 1H), 7.94 (d, J=10.0 Hz, 1H), 7.83-7.70 (m, 2H), 7.61 (s, 1H), 7.57-7.46 (m, 2H), 7.41 (s, 1H), 5.59 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.73-4.33 (m, 4H), 3.85-3.65 (m, 2H), 1.33 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 636.25 (M+H+). Example 319: (S)-2-(2-chloro-4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1031 and I-94. 1H NMR (400 MHz, DMSO) δ 8.75 (d, J=5.1 Hz, 1H), 8.36 (s, 1H), 7.94 (d, J=10.0 Hz, 1H), 7.83-7.70 (m, 2H), 7.61 (s, 1H), 7.57-7.46 (m, 2H), 7.41 (s, 1H), 5.59 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.73-4.33 (m, 4H), 3.85-3.65 (m, 2H), 1.33 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 644.8 (M+H+). Example 320: (S)-2-(2-chloro-4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(2-((4-cyano-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1030 and I-94. 1H NMR (400 MHz, DMSO) δ 8.75 (d, J=5.1 Hz, 1H), 8.51 (s, 1H), 7.94 (d, J=10.0 Hz, 1H), 7.88-7.69 (m, 3H), 7.68-7.57 (m, 2H), 7.49 (d, J=5.1 Hz, 1H), 7.42 (s, 1H), 5.59 (s, 2H), 5.02 (d, J=6.8 Hz, 1H), 4.73-4.23 (m, 4H), 3.88-3.68 (m, 2H), 1.33 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 626.81 (M+H+). Example 321: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1233 and I-3. 1H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.98-7.70 (m, 9H), 7.57 (d, J=8.5 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.65 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.57 (d, J=16.7 Hz, 2H), 4.47 (dd, J=11.1, 6.7 Hz, 1H), 4.31 (d, J=17.1 Hz, 1H), 3.86-3.69 (m, 2H), 1.38 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 613.44 (M+H+). Example 322: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1233 and I-109. 1H NMR (400 MHz, DMSO) δ 8.47 (s, 1H), 8.01-7.69 (m, 8H), 7.56 (d, J=8.5 Hz, 1H), 5.74 (s, 2H), 5.08 (d, J=6.7 Hz, 1H), 4.76-4.44 (m, 3H), 4.29 (d, J=17.1 Hz, 1H), 3.77 (d, J=3.8 Hz, 2H), 1.38 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 631.32 (M+H+). Example 323: (S)-2-(2-chloro-4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1030 and I-1243. 1H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.30 (s, 1H), 7.83 (dd, J=10.2, 8.2 Hz, 2H), 7.64 (d, J=8.4 Hz, 1H), 7.53 (s, 1H), 7.37 (s, 1H), 7.32-7.23 (m, 3H), 5.43 (s, 2H), 5.02 (d, J=6.8 Hz, 1H), 4.72-4.34 (m, 4H), 3.93 (s, 4H), 2.32 (s, 3H), 1.33 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 666.25 (M+H+). Example 324: (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1230 and I-1243. 1H NMR (400 MHz, DMSO) δ 8.36 (d, J=8.3 Hz, 2H), 7.96-7.87 (m, 2H), 7.81 (dd, J=10.2, 8.2 Hz, 1H), 7.70 (dd, J=8.2, 2.8 Hz, 1H), 7.57-7.46 (m, 2H), 7.27 (s, 1H), 5.54 (s, 2H), 5.01 (d, J=6.6 Hz, 1H), 4.64-4.32 (m, 5H), 3.96 (s, 3H), 1.31 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 653.53 (M+H+). Example 325: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-108 and I-1092. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 8.10-7.80 (m, 3H), 7.80-7.65 (m, 3H), 7.65-7.35 (m, 3H), 5.68 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.69-4.30 (m, 4H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 631.17 (M+H+). Example 326: (S)-2-((5′-chloro-6-((4-cyano-2-fluorobenzyl)oxy)-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((5′-chloro-6-((4-cyano-2-fluorobenzyl)oxy)-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1238 and I-3. 1H NMR (400 MHz, DMSO) δ 8.34 (d, J=20.1 Hz, 2H), 8.04-7.85 (m, 2H), 7.74 (d, J=4.7 Hz, 2H), 7.55 (dd, J=11.2, 1.2 Hz, 1H), 7.40 (d, J=7.3 Hz, 1H), 7.07 (d, J=8.3 Hz, 1H), 6.67 (s, 1H), 5.65-5.38 (m, 4H), 5.13 (d, J=6.5 Hz, 1H), 4.64-4.39 (m, 2H), 3.83-3.53 (m, 2H), 1.38 (s, 3H), 0.68 (s, 3H). ES/MS m/z: 646.34 (M+H+). Example 327: (S)-2-((5′-chloro-6-((4-chloro-2-fluorobenzyl)oxy)-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((5′-chloro-6-((4-chloro-2-fluorobenzyl)oxy)-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1238 and I-102. 1H NMR (400 MHz, DMSO) δ 8.34 (d, J=16.2 Hz, 2H), 7.91 (dd, J=8.3, 7.3 Hz, 1H), 7.68-7.44 (m, 3H), 7.44-7.27 (m, 2H), 7.02 (d, J=8.3 Hz, 1H), 6.70 (s, 1H), 5.45 (s, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.63-4.29 (m, 2H), 3.93-3.63 (m, 2H), 1.39 (s, 3H), 0.68 (s, 3H). ES/MS m/z: 655.53 (M+H+). Example 328: (S)-2-((6-((4-cyano-2-fluorobenzyl)oxy)-5,5′-difluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((6-((4-cyano-2-fluorobenzyl)oxy)-5,5′-difluoro-2′-oxo-[2,4′-bipyridin]-1′(2′H)-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1237 and I-109. 1H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.24 (d, J=7.0 Hz, 1H), 8.04-7.84 (m, 2H), 7.87-7.70 (m, 3H), 7.66 (d, J=8.5 Hz, 1H), 7.59 (dt, J=8.6, 2.0 Hz, 1H), 6.92 (d, J=7.6 Hz, 1H), 5.67 (s, 2H), 5.63-5.32 (m, 2H), 5.10 (d, J=6.6 Hz, 1H), 4.55 (d, J=11.1 Hz, 1H), 4.44 (dd, J=11.1, 6.8 Hz, 1H), 3.83-3.59 (m, 2H), 1.37 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 630.22 (M+H+). Example 329: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1230 and I-102. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 8.10-7.90 (m, 2H), 7.85 (t, J=7.8 Hz, 1H), 7.74-7.57 (m, 2H), 7.57-7.44 (m, 3H), 7.32 (dd, J=8.2, 2.1 Hz, 1H), 6.89 (d, J=8.2 Hz, 1H), 5.53 (s, 2H), 5.01 (d, J=6.6 Hz, 1H), 4.66-4.23 (m, 4H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 622.45 (M+H+). Example 330: Methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate The title compound was prepared in a manner similar to that described in Procedure 35, starting with Intermediates I-109 and I-1229. Methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (330-1): ES/MS m/z: 627.7 (M+H+). Methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Example 330): 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 8.05-7.68 (m, 9H), 7.62 (d, J=8.5 Hz, 1H), 7.49 (t, J=7.9 Hz, 1H), 5.72 (s, 2H), 4.99 (d, J=6.7 Hz, 1H), 4.60-4.29 (m, 3H), 3.88-3.67 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 613.3 (M+H+). Example 331: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1229 and I-1021H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.00-7.90 (m, 2H), 7.90-7.78 (m, 2H), 7.74-7.57 (m, 3H), 7.57-7.43 (m, 2H), 7.33 (dd, J=8.2, 2.1 Hz, 1H), 6.90 (d, J=8.2 Hz, 1H), 5.53 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.62-4.27 (m, 5H), 3.88-3.67 (m, 2H), 1.30 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 604.3 (M+H+). Example 332: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1229 and I-1036. 1H NMR (400 MHz, DMSO) δ 8.50 (s, 1H), 7.98-7.86 (m, 2H), 7.86-7.80 (m, 2H), 7.76-7.58 (m, 3H), 7.57-7.44 (m, 2H), 7.35 (dd, J=8.2, 2.1 Hz, 1H), 5.62 (s, 2H), 5.00 (d, J=6.6 Hz, 1H), 4.61-4.33 (m, 5H), 3.83-3.67 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 622.16 (M+H+). Example 334: (S)-2-(2-chloro-4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2-chloro-4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1031 and I-1036. 1H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.83 (dd, J=10.5, 8.1 Hz, 1H), 7.59 (t, J=8.2 Hz, 1H), 7.55-7.45 (m, 3H), 7.41-7.22 (m, 3H), 5.51 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.59-4.47 (m, 1H), 4.47-4.32 (m, 2H), 3.88-3.60 (m, 2H), 2.31 (s, 3H), 1.33 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 670.33 (M+H+). Example 335: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1231 and I-1036. 1H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.91 (dd, J=10.2, 8.2 Hz, 1H), 7.78 (dd, J=8.4, 1.5 Hz, 1H), 7.75-7.69 (m, 1H), 7.69-7.48 (m, 4H), 7.36 (dd, J=8.4, 2.1 Hz, 1H), 5.61 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.76-4.27 (m, 4H), 3.92-3.69 (m, 2H), 1.39 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 658.12 (M+H+). Example 336: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1232 and I-1036. 1H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.91 (dd, J=10.2, 8.2 Hz, 1H), 7.74 (ddd, J=10.5, 5.7, 2.0 Hz, 1H), 7.69-7.56 (m, 2H), 7.52 (ddd, J=11.1, 3.4, 1.6 Hz, 2H), 7.35 (dd, J=8.2, 2.1 Hz, 1H), 5.61 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.67 (d, J=17.4 Hz, 1H), 4.62-4.32 (m, 3H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 676.38 (M+H+). Example 337: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1230 and I-109. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 8.09-7.64 (m, 8H), 7.57-7.39 (m, 2H), 5.72 (s, 2H), 5.00 (d, J=6.6 Hz, 1H), 4.59-4.28 (m, 2H), 3.75-3.80 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H). ES/MS m/z: 630.35 (M+H+). Example 339: (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1232 and I-1243. 1H NMR (400 MHz, DMSO) δ 8.36 (d, J=2.6 Hz, 2H), 7.90 (dd, J=10.2, 8.2 Hz, 1H), 7.75 (ddd, J=10.4, 5.8, 2.0 Hz, 1H), 7.61 (ddd, J=8.1, 2.9, 1.5 Hz, 1H), 7.51 (dd, J=11.2, 1.2 Hz, 1H), 7.27 (s, 1H), 5.53 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.67 (d, J=17.5 Hz, 1H), 4.61-4.31 (m, 3H), 3.95 (s, 3H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 689.58 (M+H+). Example 340: (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-108 and I-1243. 1H NMR (400 MHz, DMSO) δ 8.35 (s, 2H), 7.84 (ddd, J=10.2, 7.4, 4.4 Hz, 2H), 7.61-7.43 (m, 3H), 7.27 (s, 1H), 5.52 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.63-4.49 (m, 2H), 4.49-4.28 (m, 2H), 3.95 (s, 3H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 671.6 (M+H+). Example 341: (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1231 and I-1243. 1H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.36 (s, 1H), 7.90 (dd, J=10.2, 8.2 Hz, 1H), 7.78 (dd, J=8.4, 1.5 Hz, 1H), 7.60 (dd, J=8.7, 3.3 Hz, 2H), 7.28 (s, 1H), 5.53 (s, 2H), 5.09 (d, J=6.5 Hz, 1H), 4.75-4.52 (m, 2H), 4.47 (dd, J=11.1, 6.7 Hz, 1H), 4.36 (d, J=17.4 Hz, 1H), 3.96 (s, 3H), 3.86-3.71 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 671.51 (M+H+). Example 342: (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-chloro-4-methoxypyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1243. 1H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.35 (s, 1H), 7.89-7.76 (m, 3H), 7.64 (d, J=8.5 Hz, 1H), 7.59-7.41 (m, 2H), 7.27 (s, 1H), 5.52 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.63-4.32 (m, 4H), 3.95 (s, 3H), 3.88-3.66 (m, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 653.76 (M+H+). Example 343: 2-(2-chloro-4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1239 and I-1241. 1H NMR (400 MHz, DMSO) δ 8.79 (d, J=5.2 Hz, 1H), 8.36 (s, 1H), 8.17 (d, J=7.0 Hz, 1H), 7.70-7.45 (m, 5H), 7.35 (dd, J=8.2, 2.1 Hz, 1H), 5.54 (s, 2H), 5.04 (d, J=6.7 Hz, 1H), 4.67 (d, J=17.1 Hz, 1H), 4.59-4.37 (m, 3H), 3.84-3.66 (m, 2H), 1.36 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 657.36 (M+H+). Example 344: 2-(2-chloro-4-(2-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-4-(2-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyrimidin-4-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1239 and I-1242. 1H NMR (400 MHz, DMSO) δ 8.80 (d, J=5.2 Hz, 1H), 8.36 (s, 1H), 8.16 (d, J=7.0 Hz, 1H), 7.75 (t, J=7.6 Hz, 1H), 7.65 (dd, J=5.2, 1.8 Hz, 1H), 7.59 (d, J=11.7 Hz, 1H), 7.50 (dd, J=16.7, 9.4 Hz, 3H), 7.07 (t, J=55.6 Hz, 1H), 5.61 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.66 (d, J=17.1 Hz, 1H), 4.58-4.37 (m, 3H), 3.85-3.70 (m, 2H), 1.36 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 673.175 (M+H+). Example 345: 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1240 and I-109. 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 8.00-7.83 (m, 3H), 7.83-7.71 (m, 3H), 7.65-7.43 (m, 3H), 5.69 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.69-4.49 (m, 2H), 4.49-4.35 (m, 2H), 3.80 (d, J=8.6 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 647.46 (M+H+). Example 346: 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-5-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1239 and I-109. 1H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 8.00-7.83 (m, 3H), 7.83-7.70 (m, 2H), 7.60-7.43 (m, 3H), 5.70 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.62 (d, J=17.0 Hz, 1H), 4.53 (d, J=11.9 Hz, 1H), 4.47-4.36 (m, 2H), 3.83-3.69 (m, 2H), 1.35 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 665.3 (M+H+). Example 347: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1041 and I-1036. 1H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.84 (ddd, J=11.9, 10.3, 7.4 Hz, 2H), 7.63 (t, J=8.1 Hz, 1H), 7.59-7.43 (m, 4H), 7.35 (dd, J=8.2, 2.0 Hz, 1H), 5.61 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.61-4.47 (m, 2H), 4.48-4.32 (m, 2H), 3.75 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 658.8 (M+H+). Example 348: 2-(4-(4-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(4-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1041 and I-1336. 1H NMR (400 MHz, MeOD) δ 8.60 (d, J=5.8 Hz, 1H), 8.55 (s, 1H), 7.90 (dd, J=10.3, 6.1 Hz, 1H), 7.66 (d, J=11.0 Hz, 1H), 7.56 (t, J=8.0 Hz, 1H), 7.35-7.13 (m, 3H), 6.91 (d, J=5.8 Hz, 1H), 5.59 (s, 2H), 4.91 (s, 8H), 4.65-4.55 (m, 1H), 4.55-4.42 (m, 3H), 3.93 (d, J=8.8 Hz, 1H), 3.79 (d, J=8.8 Hz, 1H), 3.33 (p, J=1.7 Hz, 5H), 1.36 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 641.2 (M+H+). Example 349: 2-[[2,5-difluoro-4-[6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylic acid Procedure 40 Methyl 5-[[6-[4-[2-[4-ethoxycarbonyl-2-fluoro-6-[[(2S)-oxetan-2-yl]methylamino]anilino]-2-oxo-ethyl]-2,5-difluoro-phenyl]-2-pyridyl]oxymethyl]isoindoline-2-carboxylate (349-1). 1-Methylimidazole (0.0186 mL, 0.233 mmol) followed by TCFH (15.7 mg, 0.0559 mmol) was added to a solution of 2-[2,5-difluoro-4-[6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]acetic acid (20.9 mg, 0.0460 mmol) and ethyl 4-amino-3-fluoro-5-[[(2S)-oxetan-2-yl]methylamino]benzoate (12.5 mg, 0.0466 mmol) in CH3CN (2 mL) at 0° C. The mixture was stirred for 1 hr., then diluted with EtOAc and washed with saturated NH4Cl, 1N NaOH, and brine. Dried over sodium sulfate, concentrated and carried onto next step below without purification. ES/MS: 706 (M+H+). Ethyl 2-[[2,5-difluoro-4-[6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (349-2): A solution of methyl 5-[[6-[4-[2-[4-ethoxycarbonyl-2-fluoro-6-[[(2S)-oxetan-2-yl]methylamino]anilino]-2-oxo-ethyl]-2,5-difluoro-phenyl]-2-pyridyl]oxymethyl]isoindoline-2-carboxylate (32.0 mg, 0.0454 mmol) and acetic acid (81.8 mg, 1.36 mmol) in DCE (4 mL) was heated at 60° C. overnight, then at 75° C. for 9 hr. The mixture was cooled to rt. Diluted with EtOAc and carefully neutralized with NaHCO3(114 mg). Washed organic layer with brine and dried over sodium sulfate. The crude residue was purified by chromatography (eluent: EtOAc/hexanes) to give desired product. ES/MS: 687.2 (M+H+); NMR (400 MHz, Chloroform-d) δ 7.94 (s, 1H), 7.84 (dd, J=10.8, 6.3 Hz, 1H), 7.73-7.61 (m, 2H), 7.49 (dd, J=7.7, 1.5 Hz, 1H), 7.46-7.35 (m, 2H), 7.33 (s, 1H), 7.11 (dd, J=11.3, 6.1 Hz, 1H), 6.82 (d, J=8.3 Hz, 1H), 5.48 (s, 2H), 5.24-5.08 (m, 1H), 4.84-4.69 (m, 5H), 4.68-4.21 (m, 6H), 3.81 (s, 3H), 2.76 (dq, J=14.0, 7.5 Hz, 1H), 2.41 (dt, J=17.7, 7.9 Hz, 1H), 1.44 (t, J=7.1 Hz, 3H). 2-[[2,5-difluoro-4-[6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylic acid (Example 349): A suspension of ethyl 2-[[2,5-difluoro-4-[6-[(2-methoxycarbonylisoindolin-5-yl)methoxy]-2-pyridyl]phenyl]methyl]-7-fluoro-3-[[(2S)-oxetan-2-yl]methyl]benzimidazole-5-carboxylate (10.0 mg, 0.0146 mmol) and lithium hydroxide, monohydrate (300 mmol/L, 0.146 mL, 0.0437 mmol) in CH3CN (3 mL) in a 40 ml glass vial was heated at 100° C. until completion. The mixture was then diluted with EtOAc and brine, followed by the addition of 0.090 mL 1M citric acid. The organic extract was dried over sodium sulfate, filtered and concentrated. The organic extract was then purified by RP-HPLC (eluent: MeCN/H2O). The resulting product fractions were diluted with EtOAc and neutralized with sodium bicarbonate solution. The organic extract was dried over sodium sulfate, filtered and concentrated to give title product. ES/MS: 659.2 (M+H+); 1H NMR (400 MHz, Methanol-d4) δ 8.16 (d, J=1.3 Hz, 1H), 7.82-7.77 (m, 1H), 7.75 (d, J=7.9 Hz, 1H), 7.65 (dd, J=11.2, 1.2 Hz, 1H), 7.50 (dd, J=7.4, 1.6 Hz, 1H), 7.44 (t, J=9.7 Hz, 2H), 7.31 (dd, J=11.5, 7.9 Hz, 1H), 7.17 (dd, J=11.5, 6.0 Hz, 1H), 6.86 (d, J=8.2 Hz, 1H), 5.49 (s, 2H), 5.16 (qd, J=7.1, 2.5 Hz, 1H), 4.77-4.50 (m, 10H), 4.50-4.35 (m, 1H), 3.78 (d, J=1.8 Hz, 3H), 2.88-2.69 (m, 1H), 2.53-2.40 (m, 1H). Example 350: 2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2-chloro-5-fluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2-chloro-5-fluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 18, starting with Intermediates I-1268 and I-1278. 1H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J=1.2 Hz, 1H), 8.75 (d, J=2.1 Hz, 1H), 8.25 (dd, J=8.4, 2.2 Hz, 1H), 8.17 (d, J=8.4 Hz, 1H), 8.10 (d, J=1.3 Hz, 1H), 8.05-7.97 (m, 2H), 7.91 (t, J=7.9 Hz, 1H), 7.58-7.48 (m, 2H), 7.45 (d, J=11.7 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 4.63 (t, J=5.1 Hz, 2H), 4.53 (s, 2H), 3.70 (t, J=5.0 Hz, 2H), 3.22 (s, 3H). ES/MS m/z: 632.4 (M+H+). Example 351: 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-fluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2-chloro-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-5-fluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 18, starting with Intermediates I-3 and I-1278. 1H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J=1.2 Hz, 1H), 7.98-7.85 (m, 3H), 7.74 (d, J=5.4 Hz, 2H), 7.55-7.47 (m, 2H), 7.43 (d, J=11.8 Hz, 1H), 7.01 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.62 (t, J=5.1 Hz, 2H), 4.52 (s, 2H), 3.70 (t, J=5.0 Hz, 2H), 3.22 (s, 3H). ES/MS m/z: 607.6 (M+H+). Example 352: (R)-2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 18, starting with Intermediates I-1268 and I-1272. 1H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J=1.2 Hz, 1H), 8.75 (d, J=2.1 Hz, 1H), 8.25 (dd, J=8.4, 2.2 Hz, 1H), 8.17 (d, J=8.5 Hz, 1H), 8.12 (d, J=1.3 Hz, 1H), 8.00 (d, J=1.2 Hz, 1H), 7.96-7.82 (m, 2H), 7.60-7.46 (m, 2H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.62 (s, 2H), 4.54 (dd, J=15.2, 3.1 Hz, 1H), 4.47 (s, 2H), 4.37 (dd, J=15.2, 8.8 Hz, 1H), 3.79-3.60 (m, 1H), 3.08 (s, 3H), 1.23 (d, J=6.1 Hz, 3H). ES/MS m/z: 630.2 (M+H+). Example 353: (R)-2-(4-(6-((4-chloro-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-chloro-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 18, starting with Intermediates I-1284 and I-1272. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.2 Hz, 1H), 8.12 (d, J=1.3 Hz, 1H), 8.01 (d, J=1.2 Hz, 1H), 7.97-7.78 (m, 5H), 7.58-7.45 (m, 2H), 7.40 (dd, J=11.5, 6.0 Hz, 1H), 6.98 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.54 (dd, J=15.2, 3.1 Hz, 1H), 4.47 (s, 2H), 4.37 (dd, J=15.2, 8.8 Hz, 1H), 3.78-3.59 (m, 1H), 3.08 (s, 3H), 1.23 (d, J=6.1 Hz, 3H). ES/MS m/z: 647.2 (M+H+). Example 354: (R)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 18, starting with Intermediates I-1283 and I-1272. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.2 Hz, 1H), 8.12 (d, J=1.3 Hz, 1H), 8.01 (d, J=1.2 Hz, 1H), 7.97-7.78 (m, 5H), 7.58-7.45 (m, 2H), 7.40 (dd, J=11.5, 6.0 Hz, 1H), 6.98 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.54 (dd, J=15.2, 3.1 Hz, 1H), 4.47 (s, 2H), 4.37 (dd, J=15.2, 8.8 Hz, 1H), 3.78-3.59 (m, 1H), 3.08 (s, 3H), 1.23 (d, J=6.1 Hz, 3H). ES/MS m/z: 647.2 (M+H+). Example 355: (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 18, starting with Intermediates I-3 and I-1272. 1H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J=1.2 Hz, 1H), 7.97-7.85 (m, 2H), 7.82-7.70 (m, 3H), 7.58-7.46 (m, 2H), 7.39 (dd, J=11.5, 6.0 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 4.53 (dd, J=15.2, 3.1 Hz, 1H), 4.46 (s, 2H), 4.36 (dd, J=15.2, 8.8 Hz, 1H), 3.68 (ddd, J=9.1, 6.1, 3.1 Hz, 1H), 3.08 (s, 3H), 1.23 (d, J=6.1 Hz, 3H). ES/MS m/z: 605.2 (M+H+). Example 357: (S)-2-(4-(6-((4-(difluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-(difluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1219 and 4-(difluoromethyl)benzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.87 (s, 1H), 8.15 (dd, J=8.7, 1.3 Hz, 1H), 7.99-7.71 (m, 3H), 7.71-7.45 (m, 5H), 7.37 (dd, J=11.2, 6.0 Hz, 1H), 7.03-6.86 (m, 1H),&nbsp; 5.56 (s, 2H), 5.12 (d, J=6.6 Hz, 1H), 4.78-4.61 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H),&nbsp; 1.40 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 620.3 (M+H+). Example 358: (S)-2-(2,5-difluoro-4-(6-((4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1219 and 4-(trifluoromethyl)benzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.96 (s, 1H), 8.24 (dd, J=8.6, 1.3 Hz, 1H), 7.83 (d, J=8.6 Hz, 1H), 7.78-7.67 (m, 2H), 7.58 (d, J=8.6 Hz, 2H), 7.46 (d, J=7.5 Hz, 1H), 7.19 (d, J=10.5 Hz, 1H), 7.02 (dd, J=9.0, 2.9 Hz, 1H), 5.51 (s, 2H), 5.19 (d, J=6.5 Hz, 1H), 4.83-4.62 (m, 3H), 4.52 (dd, J=11.8, 6.6 Hz, 1H), 4.01 (d, J=9.0 Hz, 1H), 3.84 (d, J=9.0 Hz, 1H), 2.21 (s, 3H), 1.36 (s, 3H), 0.73 (s, 3H). ES/MS m/z: 638.0 (M+H+). Example 359: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-3-fluoropyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid ((S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-3-fluoropyridin-2-yl)-2-fluoro-5-methylbenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1322 and I-1076. 1H NMR (400 MHz, Methanol-d4) δ 8.96 (s, 1H), 8.24 (dd, J=8.6, 1.3 Hz, 1H), 7.83 (d, J=8.6 Hz, 1H), 7.78-7.67 (m, 2H), 7.58 (d, J=8.6 Hz, 2H), 7.46 (d, J=7.5 Hz, 1H), 7.19 (d, J=10.5 Hz, 1H), 7.02 (dd, J=9.0, 2.9 Hz, 1H), 5.51 (s, 2H), 5.19 (d, J=6.5 Hz, 1H), 4.83-4.62 (m, 3H), 4.52 (dd, J=11.8, 6.6 Hz, 1H), 4.01 (d, J=9.0 Hz, 1H), 3.84 (d, J=9.0 Hz, 1H), 2.21 (s, 3H), 1.36 (s, 3H), 0.73 (s, 3H). ES/MS m/z: 627.0 (M+H+). Example 360: (S)-2-(2,5-difluoro-4-(6-((4-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((4-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1219 and 4-fluorobenzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.18 (dd, J=8.6, 1.3 Hz, 1H), 7.93 (dd, J=10.8, 6.2 Hz, 1H), 7.87-7.73 (m, 2H), 7.66-7.48 (m, 3H), 7.40 (dd, J=11.2, 6.0 Hz, 1H), 7.21-7.02 (m, 2H), 6.90 (d, J=8.2 Hz, 1H), 5.46 (s, 2H), 5.15 (d, J=6.5 Hz, 1H), 4.79-4.63 (m, 2H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 3.33 (p, J=1.7 Hz, 3H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 588.5 (M+H+). Example 361: (S)-2-(2,5-difluoro-4-(6-((4-methylbenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((4-methylbenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1219 and 4-methylbenzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J=8.6, 1.3 Hz, 1H), 7.93 (dd, J=10.9, 6.3 Hz, 1H), 7.88-7.72 (m, 2H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.38 (dd, J=14.9, 7.0 Hz, 3H), 7.19 (d, J=7.8 Hz, 2H), 6.88 (d, J=8.2 Hz, 1H), 5.44 (s, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.77-4.61 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 2.34 (s, 3H), 1.42 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 584.6 (M+H+). Example 362: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,6-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1233. 1H NMR (400 MHz, Methanol-d4) δ 8.99-8.75 (m, 1H), 8.15 (dd, J=8.6, 1.4 Hz, 1H), 7.95-7.66 (m, 8H), 7.61 (d, J=7.5 Hz, 1H), 6.97 (d, J=8.2 Hz, 1H), 5.62 (s, 2H), 5.17 (d, J=6.5 Hz, 1H), 4.81-4.46 (m, 4H), 4.00 (d, J=8.9 Hz, 1H), 3.86 (d, J=8.9 Hz, 1H), 1.45 (s, 3H), 0.79 (s, 3H). ES/MS m/z: 595.3 (M+H+). Example 363: (S)-2-(4-(6-(benzyloxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-(benzyloxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1324 and I-82. 1H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J=8.6, 1.4 Hz, 1H), 7.92 (dd, J=10.9, 6.3 Hz, 1H), 7.87-7.70 (m, 2H), 7.56 (dd, J=7.4, 1.7 Hz, 1H), 7.52-7.44 (m, 2H), 7.42-7.33 (m, 4H), 6.91 (d, J=8.2 Hz, 1H), 5.49 (s, 2H), 5.15 (d, J=6.5 Hz, 1H), 4.77-4.62 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 570.6 (M+H+). Example 364: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-3-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-3-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1322 and I-1229. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J=8.6, 1.3 Hz, 1H), 7.91 (d, J=8.1 Hz, 1H), 7.87-7.66 (m, 5H), 7.63-7.54 (m, 3H), 6.97 (dd, J=8.9, 2.7 Hz, 1H), 5.61 (s, 2H), 5.12 (d, J=6.6 Hz, 1H), 4.76-4.61 (m, 3H), 4.60-4.49 (m, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.83 (d, J=8.9 Hz, 1H), 1.37 (s, 3H), 0.73 (s, 3H). ES/MS m/z: 613.2 (M+H+). Example 365: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-3-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-3-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1322 and I-108. 1H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 7.82-7.65 (m, 3H), 7.65-7.55 (m, 2H), 7.35 (dd, J=9.9, 5.7 Hz, 1H), 7.27 (dd, J=9.9, 6.0 Hz, 1H), 7.01 (dd, J=9.0, 2.9 Hz, 1H), 5.54 (s, 2H), 4.99 (d, J=6.7 Hz, 1H), 4.69-4.42 (m, 4H), 3.95 (d, J=8.8 Hz, 1H), 3.80 (d, J=8.8 Hz, 1H), 1.36 (s, 3H), 0.68 (s, 3H). ES/MS m/z: 649.2 (M+H+). Example 366: 2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1247 and I-1325. 1H NMR (400 MHz, Chloroform-d) δ 8.00 (t, J=7.5 Hz, 1H), 7.80-7.68 (m, 3H), 7.68-7.57 (m, 3H), 7.56-7.36 (m, 2H), 7.15 (dd, J=11.1, 6.0 Hz, 1H), 5.60 (s, 2H), 4.67-4.55 (m, 4H), 3.82 (t, J=4.8 Hz, 2H), 3.33 (s, 3H). ES/MS m/z: 591.5 (M+H+). Example 367: (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1248 and I-108. 1H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 7.90-7.82 (m, 1H), 7.77 (t, J=7.9 Hz, 1H), 7.70 (dd, J=11.0, 1.2 Hz, 1H), 7.58-7.44 (m, 3H), 7.42-7.34 (m, 2H), 7.24 (dd, J=11.4, 6.0 Hz, 1H), 6.88 (d, J=8.2 Hz, 1H), 5.46 (s, 2H), 4.97 (d, J=6.7 Hz, 1H), 4.65-4.34 (m, 4H), 3.93 (d, J=8.8 Hz, 1H), 3.79 (d, J=8.8 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 622.8 (M+H+). Example 368: (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1272. 1H NMR (400 MHz, Methanol-d4) δ 7.99 (d, J=1.2 Hz, 1H), 7.86-7.68 (m, 2H), 7.68-7.47 (m, 5H), 7.12 (dd, J=11.6, 6.1 Hz, 1H), 5.71 (s, 2H), 4.52 (d, J=16.6 Hz, 1H), 4.45-4.34 (m, 1H), 4.27 (dd, J=15.1, 8.9 Hz, 1H), 3.75 (ddd, J=9.2, 6.1, 3.1 Hz, 1H), 3.13 (s, 3H), 2.08-1.89 (m, 1H), 1.29 (d, J=6.2 Hz, 3H). ES/MS m/z: 623.3 (M+H+). Example 369: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-3-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-3-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1326. 1H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.20 (dd, J=8.6, 1.4 Hz, 1H), 8.02 (t, J=8.3 Hz, 1H), 7.92-7.71 (m, 2H), 7.69-7.46 (m, 4H), 7.40-7.21 (m, 2H), 5.70 (s, 2H), 5.10 (d, J=6.5 Hz, 1H), 4.83-4.58 (m, 3H), 4.47 (dd, J=11.6, 6.7 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.82 (d, J=8.9 Hz, 1H), 1.33 (s, 3H), 0.69 (s, 3H). ES/MS m/z: 613.6 (M+H+). Example 370: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1327 and 4-cyanobenzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 1H), 8.15 (dd, J=8.6, 1.4 Hz, 1H), 7.96-7.82 (m, 2H), 7.78 (d, J=2.9 Hz, 1H), 7.77-7.70 (m, 3H), 7.70-7.56 (m, 3H), 7.56-7.42 (m, 1H), 5.69 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.74-4.58 (m, 3H), 4.49 (dd, J=11.5, 6.7 Hz, 1H), 4.18 (ddd, J=12.3, 9.0, 3.7 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.82 (d, J=8.9 Hz, 1H), 3.76-3.72 (m, 1H), 3.56 (dt, J=11.8, 2.8 Hz, 1H), 1.36 (s, 3H), 0.71 (s, 3H). ES/MS m/z: 595.7 (M+H+). Example 371: (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1328 and 4-chlorobenzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.96 (s, 1H), 8.25 (dd, J=8.6, 1.4 Hz, 1H), 8.06-7.92 (m, 2H), 7.88-7.73 (m, 2H), 7.68-7.55 (m, 2H), 7.55-7.44 (m, 2H), 7.44-7.28 (m, 2H), 6.88 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 5.19 (d, J=6.4 Hz, 1H), 4.78 (d, J=7.2 Hz, 2H), 4.65 (dd, J=11.7, 1.3 Hz, 1H), 4.51 (dd, J=11.8, 6.6 Hz, 1H), 4.01 (d, J=9.0 Hz, 1H), 3.84 (d, J=9.0 Hz, 1H), 1.37 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 587.0 (M+H+). Example 372: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1230. 1H NMR (400 MHz, Methanol-d4) δ 8.57 (s, 1H), 7.91-7.62 (m, 8H), 7.52 (d, J=7.5 Hz, 1H), 7.38 (t, J=7.8 Hz, 1H), 6.89 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.94 (s, 1H), 4.60-4.48 (m, 3H), 4.43 (dd, J=11.4, 6.8 Hz, 1H), 3.93 (d, J=8.9 Hz, 1H), 3.78 (d, J=8.8 Hz, 1H), 1.29 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 595.7 (M+H+). Example 373: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1328 and 4-cyanobenzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.21 (dd, J=8.6, 1.4 Hz, 1H), 8.00-7.87 (m, 2H), 7.87-7.79 (m, 2H), 7.74 (d, J=8.3 Hz, 2H), 7.67 (d, J=8.1 Hz, 2H), 7.61-7.46 (m, 2H), 6.93 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.15 (d, J=6.5 Hz, 1H), 4.73 (d, J=5.6 Hz, 2H), 4.64 (dd, J=11.7, 1.4 Hz, 1H), 4.50 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.83 (d, J=8.9 Hz, 1H), 1.36 (s, 3H), 0.73 (s, 3H). ES/MS m/z: 577.7 (M+H+). Example 374: 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-fluoro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)benzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1282 and I-1324. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.2 Hz, 1H), 8.01 (d, J=1.2 Hz, 1H), 7.98-7.76 (m, 5H), 7.65 (dd, J=8.5, 6.8 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.48-7.32 (m, 2H), 6.98 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 4.61 (t, J=5.2 Hz, 2H), 4.43 (s, 2H), 3.73 (t, J=5.2 Hz, 2H), 3.23 (s, 3H). ES/MS m/z: 633.3 (M+H+). Example 375: 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-7-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1325. 1H NMR (400 MHz, Methanol-d4) δ 7.94 (dd, J=8.6, 6.6 Hz, 1H), 7.82 (t, J=7.8 Hz, 1H), 7.79-7.72 (m, 3H), 7.67 (d, J=8.1 Hz, 2H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.50 (d, J=8.6 Hz, 1H), 7.25 (dd, J=11.4, 6.0 Hz, 1H), 6.95 (d, J=8.2 Hz, 1H), 5.59 (s, 2H), 4.74 (t, J=5.0 Hz, 2H), 4.59 (s, 2H), 3.84 (t, J=4.9 Hz, 2H), 3.32 (s, 3H). ES/MS m/z: 573.3 (M+H+). Example 376: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-108. 1H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 7.86-7.72 (m, 4H), 7.71-7.63 (m, 3H), 7.54 (dd, J=7.5, 1.6 Hz, 1H), 7.23 (dd, J=11.4, 6.0 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 4.97 (d, J=6.7 Hz, 1H), 4.65-4.41 (m, 4H), 3.94 (d, J=8.8 Hz, 1H), 3.80 (d, J=8.8 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 613.63 (M+H+). Example 377: 2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1136 and 4-chlorobenzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.15 (d, J=1.2 Hz, 1H), 7.83 (dd, J=8.3, 7.5 Hz, 1H), 7.66 (ddd, J=11.3, 5.4, 1.8 Hz, 2H), 7.56 (dd, J=7.4, 1.7 Hz, 1H), 7.53-7.45 (m, 2H), 7.45-7.33 (m, 2H), 6.92 (d, J=8.3 Hz, 1H), 5.48 (s, 2H), 4.67 (t, J=5.0 Hz, 2H), 4.60 (s, 2H), 3.86-3.78 (m, 2H), 3.33 (s, 3H). ES/MS m/z: 600.4 (M+H+). Example 378: 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 36, starting with Intermediates I-116 and I-1032. 1H NMR (400 MHz, Methanol-d4) δ 8.15 (d, J=1.3 Hz, 1H), 7.85 (t, J=7.9 Hz, 1H), 7.75 (d, J=8.3 Hz, 2H), 7.72-7.64 (m, 3H), 7.63-7.56 (m, 2H), 6.97 (d, J=8.3 Hz, 1H), 5.59 (s, 2H), 4.68 (d, J=5.0 Hz, 2H), 4.60 (s, 2H), 3.75 (dd, J=2.9, 1.7 Hz, 1H), 3.62-3.51 (m, 2H), 3.32 (s, 3H). ES/MS m/z: 591.6 (M+H+). Example 379: (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 380: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 379 and 380 were prepared via preparative chiral SFC (Lux Cellulose-2, EtOH/CO2eluent) of Example 384. (R)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 379) was isolated as the later-eluting of two stereoisomers. NMR ES/MS m/z: 595.6 (M+H+). (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 379) was isolated as the earlier-eluting of two stereoisomers. NMR ES/MS m/z: 595.6 (M+H+). Example 381: (R)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid (R)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1033. 1H NMR (400 MHz, Methanol-d4) δ 8.17 (d, J=1.2 Hz, 1H), 7.83-7.59 (m, 8H), 7.50 (dd, J=7.5, 1.5 Hz, 1H), 7.19 (dd, J=11.5, 6.0 Hz, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.55 (s, 2H), 4.61-4.45 (m, 3H), 4.37 (dd, J=15.2, 9.1 Hz, 1H), 3.72 (dqt, J=10.1, 6.9, 3.4 Hz, 1H), 3.15 (s, 3H), 1.30 (d, J=6.1 Hz, 3H). ES/MS m/z: 587.73 (M+H+). Example 382: 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1206 and 4-cyanobenzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.19 (s, 1H), 7.80 (d, J=7.8 Hz, 1H), 7.77-7.69 (m, 4H), 7.66 (d, J=8.3 Hz, 2H), 7.51 (dd, J=7.5, 1.6 Hz, 1H), 7.19 (dd, J=11.4, 6.0 Hz, 1H), 6.92 (d, J=8.2 Hz, 1H), 5.57 (s, 2H), 4.63 (t, J=5.0 Hz, 2H), 4.57 (s, 2H), 3.75 (t, J=4.9 Hz, 2H), 3.27 (s, 3H). ES/MS m/z: 573.6 (M+H+). Example 383A: (S)-2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 383B: (R)-2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 383A and 383B were prepared via preparative chiral SFC (Daicel Chiralpak AD-H column, EtOH/CO2eluent) of Example 21. (S)-2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 383A) was isolated as the later-eluting of two stereoisomers. 1H NMR (400 MHz, Methanol-d4) δ 8.79 (s, 1H), 8.07 (dd, J=8.5, 1.4 Hz, 1H), 7.97-7.77 (m, 2H), 7.69 (dd, J=24.1, 8.1 Hz, 2H), 7.55 (dd, J=7.5, 1.5 Hz, 1H), 7.43-7.20 (m, 3H), 6.97-6.86 (m, 1H), 5.59 (s, 2H), 5.04 (d, J=6.7 Hz, 1H), 4.67-4.44 (m, 4H), 3.97 (d, J=8.8 Hz, 1H), 3.82 (d, J=8.8 Hz, 1H), 3.75-3.61 (m, 2H), 1.39 (s, 3H), 0.71 (s, 3H). ES/MS m/z: 638.36 (M+H+). (R)-2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 383B): 1H NMR (400 MHz, Methanol-d4) δ 8.79 (s, 1H), 8.08 (dd, J=8.5, 1.5 Hz, 1H), 7.94-7.76 (m, 2H), 7.69 (dd, J=24.9, 8.1 Hz, 2H), 7.56 (dd, J=7.5, 1.6 Hz, 1H), 7.45-7.20 (m, 3H), 6.92 (d, J=8.4 Hz, 1H), 5.60 (s, 2H), 5.05 (d, J=6.7 Hz, 1H), 4.76-4.43 (m, 4H), 3.97 (d, J=8.8 Hz, 1H), 3.82 (d, J=8.8 Hz, 1H), 3.74-3.66 (m, 1H), 1.39 (s, 3H), 0.71 (s, 3H). ES/MS m/z: 638.3 (M+H+). Example 384: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2-fluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 36, starting with Intermediates I-1329 and I-25. 1H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.14 (dd, J=8.6, 1.4 Hz, 1H), 8.03-7.87 (m, 2H), 7.87-7.67 (m, 4H), 7.67-7.44 (m, 3H), 6.92 (d, J=8.2 Hz, 1H), 5.66 (s, 2H), 5.08 (d, J=6.6 Hz, 1H), 4.72-4.56 (m, 3H), 4.49 (dd, J=11.5, 6.8 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.82 (d, J=8.8 Hz, 1H), 3.72-3.63 (m, 1H), 1.36 (s, 3H), 0.71 (s, 3H). ES/MS m/z: 595.6 (M+H+). Example 385A: (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 385B: (R)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 385A and 385B were prepared via preparative chiral SFC (Daicel Chiralpak AD-H column, EtOH/CO2eluent) of Example 387. (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 385A) was isolated as the later-eluting of two stereoisomers. 1H NMR (400 MHz, Methanol-d4) δ 8.84 (s, 1H), 8.13 (dd, J=8.5, 1.4 Hz, 1H), 8.00-7.70 (m, 3H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.48 (d, J=8.2 Hz, 2H), 7.44-7.23 (m, 3H), 6.91 (d, J=8.2 Hz, 1H), 5.48 (s, 2H), 5.18-5.02 (m, 1H), 4.75-4.48 (m, 4H), 3.99 (d, J=8.9 Hz, 1H), 3.85 (s, 1H), 3.74-3.61 (m, 1H), 1.40 (s, 3H), 0.74 (s, 3H). ES/MS m/z: 604.85 (M+H+). (R)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 385B) was isolated as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, Methanol-d4) δ 8.82 (s, 1H), 8.15-8.01 (m, 1H), 7.98-7.66 (m, 3H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.48 (d, J=8.2 Hz, 2H), 7.42-7.34 (m, 3H), 6.90 (d, J=8.2 Hz, 1H), 5.48 (s, 2H), 5.12-4.99 (m, 1H), 4.74-4.43 (m, 4H), 3.98 (d, J=8.9 Hz, 1H), 3.92-3.73 (m, 2H), 1.40 (s, 3H), 0.73 (s, 3H). ES/MS m/z: 604.8 (M+H+). Example 386: 2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1206 and 4-chlorobenzyl bromide. 1H NMR (400 MHz, DMSO-d6) δ 8.10 (s, 1H), 7.97-7.73 (m, 2H), 7.60-7.31 (m, 7H), 6.95 (d, J=8.2 Hz, 1H), 5.47 (s, 2H), 4.62 (t, J=5.1 Hz, 2H), 4.46 (s, 2H), 3.68 (t, J=5.0 Hz, 2H), 3.66-3.51 (m, 1H), 3.21 (s, 3H). ES/MS m/z: 582.65 (M+H+). Example 387: 2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-11H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1199 and 4-chlorobenzyl bromide. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J=8.6, 1.4 Hz, 1H), 7.90 (dd, J=10.9, 6.3 Hz, 1H), 7.87-7.70 (m, 2H), 7.56 (dd, J=7.3, 1.6 Hz, 1H), 7.53-7.44 (m, 2H), 7.43-7.31 (m, 3H), 6.92 (d, J=8.2 Hz, 1H), 5.48 (s, 2H), 5.14 (d, J=6.5 Hz, 1H), 4.78-4.60 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 605.25 (M+H+). Example 388: (S)-2-((3′-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-2,4′,5-trifluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((3′-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-2,4′,5-trifluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1061. 1H NMR (400 MHz, DMSO-d6) δ 8.93 (d, J=1.2 Hz, 1H), 8.83 (s, 1H), 8.48 (s, 1H), 8.33 (s, 1H), 8.04 (d, J=1.3 Hz, 1H), 7.80 (dd, J=8.4, 1.4 Hz, 1H), 7.62 (t, J=8.2 Hz, 2H), 7.56 (dd, J=10.1, 6.5 Hz, 1H), 7.47-7.43 (m, 1H), 7.40 (dd, J=11.3, 8.6 Hz, 1H), 7.30-7.23 (m, 1H), 5.46 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.58-4.48 (m, 2H), 4.44 (dd, J=11.1, 6.8 Hz, 1H), 4.36 (d, J=16.8 Hz, 1H), 3.82-3.68 (m, 2H), 1.34 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 689.1 (M+H+). Example 389: 2-(4-(2-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(2-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-96 and I-1062. 1H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J=1.3 Hz, 1H), 8.84 (s, 1H), 8.80 (d, J=5.2 Hz, 1H), 8.32 (s, 1H), 8.21 (s, 1H), 8.03 (d, J=1.2 Hz, 1H), 7.97 (dd, J=10.1, 6.3 Hz, 1H), 7.79 (dd, J=8.6, 1.5 Hz, 1H), 7.65 (dd, J=5.3, 1.8 Hz, 1H), 7.59 (d, J=8.4 Hz, 1H), 7.48 (dd, J=11.5, 5.8 Hz, 1H), 5.69 (s, 2H), 4.59 (d, J=5.5 Hz, 2H), 4.48 (s, 2H), 3.68 (t, J=5.0 Hz, 2H), 3.20 (s, 3H). ES/MS m/z: 633.1 (M+H+). Example 390: 2-((3′-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-2,4′,5-trifluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-((3′-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-2,4′,5-trifluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-96 and I-1061. 1H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J=1.3 Hz, 1H), 8.83 (s, 1H), 8.33 (s, 1H), 8.25 (d, J=1.5 Hz, 1H), 8.04 (d, J=1.3 Hz, 1H), 7.83 (dd, J=8.4, 1.5 Hz, 1H), 7.62 (dd, J=8.5, 3.5 Hz, 2H), 7.56 (dd, J=10.2, 6.5 Hz, 1H), 7.39 (dd, J=11.0, 7.5 Hz, 2H), 7.31-7.19 (m, 1H), 5.45 (s, 2H), 4.63 (t, J=5.2 Hz, 2H), 4.47 (s, 2H), 3.68 (d, J=5.3 Hz, 2H), 3.21 (s, 3H). ES/MS m/z: 649.1 (M+H+). Example 391: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 38, starting with Intermediates I-1186 and I-1049. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.83 (s, 1H), 8.31 (s, 1H), 8.21 (d, J=1.5 Hz, 1H), 8.03 (d, J=1.3 Hz, 1H), 7.96 (t, J=7.8 Hz, 1H), 7.82-7.73 (m, 2H), 7.58 (dd, J=13.1, 7.9 Hz, 2H), 7.06 (d, J=8.4 Hz, 1H), 5.67 (s, 2H), 4.65 (s, 2H), 4.53 (s, 2H), 3.72 (t, J=5.0 Hz, 2H), 3.24 (s, 3H). ES/MS m/z: 650.1 (M+H+). Example 392: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 38, starting with Intermediates I-1186 and I-1050. 1H NMR (400 MHz, DMSO-d6) δ 8.91 (d, J=1.3 Hz, 1H), 8.85 (s, 1H), 8.32 (s, 1H), 8.21 (s, 1H), 8.03 (d, J=1.3 Hz, 1H), 7.99-7.86 (m, 1H), 7.80-7.70 (m, 1H), 7.63 (d, J=8.5 Hz, 1H), 7.56 (d, J=8.4 Hz, 1H), 5.76 (s, 2H), 4.65 (s, 2H), 4.52 (s, 2H), 3.72 (t, J=4.9 Hz, 2H), 3.24 (s, 3H). ES/MS m/z: 668.1 (M+H+). Example 393: (S)-2-(4-(6-((3,4-dichlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((3,4-dichlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1060. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.89 (t, J=7.9 Hz, 1H), 7.80 (dt, J=10.4, 3.3 Hz, 3H), 7.63 (dd, J=16.7, 8.4 Hz, 2H), 7.57-7.41 (m, 3H), 6.98 (d, J=8.3 Hz, 1H), 5.48 (s, 2H), 5.01 (d, J=6.7 Hz, 1H), 4.58-4.48 (m, 2H), 4.48-4.31 (m, 2H), 3.81-3.71 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 638.2 (M+H+). Example 394: (S)-2-(4-(6-((4-chloro-3-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-3-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1059. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.89 (t, J=7.9 Hz, 1H), 7.83-7.75 (m, 2H), 7.61 (dt, J=8.0, 3.8 Hz, 2H), 7.58-7.49 (m, 2H), 7.45 (dd, J=11.2, 6.2 Hz, 1H), 7.39-7.35 (m, 1H), 6.98 (d, J=8.2 Hz, 1H), 5.48 (s, 2H), 5.01 (d, J=6.5 Hz, 1H), 4.59-4.48 (m, 2H), 4.48-4.31 (m, 2H), 3.81-3.68 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 622.2 (M+H+). Example 396: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1247 and I-1167. 1H NMR (400 MHz, DMSO) δ 8.08 (d, J=8.6 Hz, 1H), 7.96 (d, J=8.3 Hz, 1H), 7.87 (dd, J=9.9, 8.3 Hz, 3H), 7.77-7.67 (m, 3H), 7.55 (dd, J=8.5, 2.4 Hz, 1H), 7.46 (dd, J=11.4, 6.0 Hz, 1H), 5.67 (s, 2H), 4.96 (dd, J=8.4, 5.3 Hz, 1H), 4.74 (dd, J=9.6, 5.1 Hz, 1H), 4.58 (d, J=17.1 Hz, 1H), 4.48 (d, J=17.1 Hz, 1H), 4.37 (dt, J=9.0, 5.0 Hz, 2H), 3.63 (d, J=7.9 Hz, 1H), 1.24 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 614.0 (M+H+). Example 397: (S)-5-chloro-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-5-chloro-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1107. 1H NMR (400 MHz, DMSO) δ 8.31 (s, 1H), 7.89 (dd, J=17.8, 8.0 Hz, 3H), 7.78-7.65 (m, 4H), 7.53 (d, J=7.4 Hz, 1H), 7.45 (t, J=8.7 Hz, 1H), 7.01 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 5.00 (d, J=6.5 Hz, 1H), 4.56-4.47 (m, 2H), 4.46-4.31 (m, 3H), 4.36 (s, 15H), 4.16 (s, 1H), 3.78-3.68 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 629.0 (M+H+). Example 398: (S)-2-(4-(6-((2,4-dichlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2,4-dichlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1098 and I-82. 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.92-7.69 (m, 4H), 7.64 (dd, J=8.4, 6.1 Hz, 2H), 7.60-7.53 (m, 1H), 7.53-7.42 (m, 2H), 5.63 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.54 (dd, J=15.0, 2.5 Hz, 2H), 4.48-4.34 (m, 2H), 3.78 (d, J=8.7 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 656.0 (M+H+). Example 399: (S)-2-(4-(6-((2,4-dichlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2,4-dichlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1099 and I-82. 1H NMR (400 MHz, DMSO) δ 8.50 (s, 1H), 7.90 (t, J=7.9 Hz, 1H), 7.86-7.75 (m, 2H), 7.70 (d, J=2.1 Hz, 1H), 7.66-7.59 (m, 2H), 7.58-7.51 (m, 1H), 7.51-7.42 (m, 2H), 7.00 (d, J=8.2 Hz, 1H), 5.54 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.73 (s, 8H), 4.55 (d, J=17.4 Hz, 2H), 4.48-4.37 (m, 2H), 4.35 (s, 4H), 3.82-3.70 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 638.0 (M+H+). Example 400: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-hydroxy-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-hydroxy-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1106. 1H NMR (400 MHz, DMSO) δ 8.40 (s, 1H), 7.93-7.80 (m, 2H), 7.61 (t, J=8.1 Hz, 1H), 7.57-7.41 (m, 4H), 7.33 (dd, J=8.3, 1.9 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 4.98 (d, J=6.6 Hz, 1H), 4.57-4.47 (m, 2H), 4.47-4.34 (m, 2H), 3.77 (d, J=8.7 Hz, 1H), 3.72 (d, J=8.6 Hz, 1H), 2.58 (s, 3H), 1.31 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 636.0 (M+H+). Example 401: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-hydroxy-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-hydroxy-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1106. 1H NMR (400 MHz, DMSO) δ 8.40 (s, 1H), 7.95-7.83 (m, 3H), 7.74 (dd, J=10.4, 6.5 Hz, 1H), 7.68 (d, J=8.0 Hz, 2H), 7.53 (d, J=7.4 Hz, 1H), 7.45 (s, 1H), 7.50-7.40 (m, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.58 (s, 2H), 4.97 (d, J=6.6 Hz, 1H), 4.52 (d, J=7.5 Hz, 1H), 4.49 (s, 1H), 4.47-4.33 (m, 2H), 3.80-3.68 (m, 2H), 2.58 (s, 3H), 1.31 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 609 (M+H+). Example 402: (S)-5-chloro-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-5-chloro-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1107. 1H NMR (400 MHz, DMSO) δ 8.32 (s, 1H), 7.93-7.79 (m, 2H), 7.73 (s, 1H), 7.61 (t, J=8.2 Hz, 1H), 7.59-7.41 (m, 3H), 7.33 (dd, J=8.3, 2.0 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.00 (d, J=6.5 Hz, 1H), 4.57-4.47 (m, 2H), 4.46-4.32 (m, 2H), 3.78-3.68 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 656.0 (M+H+). Example 403: 2-(4-(6-((2-chloro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((2-chloro-4-(1H-1,2,3-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 38, starting with Intermediates I-1102 and I-1103. 1H NMR (400 MHz, DMSO) δ 8.92 (s, 1H), 8.22 (s, 1H), 8.16 (d, J=2.1 Hz, 1H), 8.03-7.92 (m, 3H), 7.87-7.75 (m, 2H), 7.75-7.67 (m, 1H), 7.58 (t, J=7.7 Hz, 2H), 7.08 (d, J=8.3 Hz, 1H), 5.63 (s, 2H), 4.65 (t, J=5.1 Hz, 2H), 4.53 (s, 2H), 3.73 (t, J=5.0 Hz, 2H), 3.24 (s, 3H), 2.56 (d, J=7.5 Hz, 1H). ES/MS m/z: 649.0 (M+H+). Example 404: (S)-2-(4-(2-((2,4-dichlorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(2-((2,4-dichlorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1099 and I-108. 1H NMR (400 MHz, Chloroform-d) δ 8.31 (d, J=5.1 Hz, 1H), 7.51 (t, J=8.1 Hz, 1H), 7.21 (d, J=5.2 Hz, 1H), 7.19-7.13 (m, 2H), 5.51-5.46 (m, 2H); 1H NMR (400 MHz, DMSO) δ 8.78 (d, J=5.1 Hz, 1H), 8.57 (d, J=1.5 Hz, 1H), 7.92 (ddt, J=10.3, 3.5, 1.7 Hz, 3H), 7.75 (dq, J=11.3, 8.2 Hz, 3H), 7.62 (dd, J=5.2, 1.8 Hz, 1H), 7.51 (dd, J=11.5, 6.0 Hz, 1H), 5.81-5.70 (m, 1H), 5.62 (s, 2H), 4.65 (s, 2H), 4.27 (dd, J=10.8, 7.9 Hz, 1H), 4.20 (dd, J=10.8, 3.3 Hz, 1H), 2.40 (dd, J=13.2, 9.4 Hz, 1H), 2.02 (dd, J=13.3, 6.9 Hz, 1H), 1.50 (s, 3H), 1.27 (s, 3H). ES/MS m/z: 658.2 (M+H+). Example 405: (S)-2-(4-(4-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(4-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1338 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.72 (d, J=5.8 Hz, 1H), 8.50 (s, 1H), 7.92 (dd, J=10.1, 6.2 Hz, 1H), 7.82 (dd, J=8.5, 1.5 Hz, 1H), 7.64 (dt, J=8.2, 4.0 Hz, 2H), 7.50 (ddd, J=16.8, 10.5, 4.0 Hz, 2H), 7.35 (dd, J=8.4, 2.1 Hz, 1H), 7.04 (d, J=5.8 Hz, 1H), 5.58 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.60-4.52 (m, 2H), 4.49-4.38 (m, 2H), 3.85-3.69 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 623.0 (M+H+). Example 406: 1-(3-oxabicyclo[3.1.1]heptan-1-yl)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid 1-(3-oxabicyclo[3.1.1]heptan-1-yl)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1153. 1H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J=1.4 Hz, 1H), 7.95-7.76 (m, 3H), 7.65-7.58 (m, 2H), 7.52 (ddd, J=12.1, 8.8, 1.9 Hz, 2H), 7.43 (dd, J=11.5, 6.1 Hz, 1H), 7.33 (dd, J=8.3, 2.1 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 4.38 (s, 2H), 4.11 (s, 2H), 3.94 (d, J=2.1 Hz, 2H), 2.90-2.77 (m, 2H), 2.69 (ddd, J=12.3, 6.5, 3.7 Hz, 3H). ES/MS m/z: 620.0 (M+H+). Example 407: racemic 1-((4R)-2-oxabicyclo[3.1.1]heptan-4-yl)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 1-((4R)-2-oxabicyclo[3.1.1]heptan-4-yl)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1342. 1H NMR (400 MHz, DMSO-d6) δ 8.71 (d, J=1.4 Hz, 1H), 7.92-7.81 (m, 3H), 7.67-7.58 (m, 2H), 7.55-7.43 (m, 3H), 7.34 (dd, J=8.2, 2.1 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.46 (q, J=4.5, 3.8 Hz, 1H), 4.74-4.63 (m, 2H), 4.60-4.40 (m, 3H), 3.08 (p, J=5.5 Hz, 1H), 2.36 (ddd, J=11.0, 5.8, 3.6 Hz, 1H), 2.28 (dd, J=10.9, 9.2 Hz, 1H), 2.13-2.04 (m, 1H), 2.00 (dd, J=10.7, 9.2 Hz, 1H). ES/MS m/z: 620.0 (M+H+). Example 408: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methoxytetrahydro-2H-pyran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 409: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methoxytetrahydro-2H-pyran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 408 and Example 409 were prepared via preparative chiral SFC (Daicel Chiralpak AD-H column, EtOH/CO2eluent) of Example 70. 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methoxytetrahydro-2H-pyran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 408) was isolated as the later-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.91 (t, J=9.9 Hz, 2H), 7.85-7.68 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.40 (dd, J=11.4, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 4.87 (dd, J=9.0, 4.7 Hz, 1H), 4.63 (d, J=16.9 Hz, 1H), 4.59-4.46 (m, 2H), 3.90 (dd, J=10.7, 4.6 Hz, 1H), 3.84-3.77 (m, 2H), 3.14 (s, 3H), 1.99 (q, J=4.6, 4.0 Hz, 3H). ES/MS m/z: 629.3 (M+H+). 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methoxytetrahydro-2H-pyran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 409) was isolated as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.91 (t, J=9.9 Hz, 2H), 7.85-7.68 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.40 (dd, J=11.4, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 4.87 (dd, J=9.0, 4.7 Hz, 1H), 4.63 (d, J=16.9 Hz, 1H), 4.59-4.46 (m, 2H), 3.90 (dd, J=10.7, 4.6 Hz, 1H), 3.84-3.77 (m, 2H), 3.14 (s, 3H), 1.99 (q, J=4.6, 4.0 Hz, 3H). ES/MS m/z: 629.3 (M+H+). Example 410: (S)-2-(4-(4-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyrimidin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(4-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyrimidin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1154 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.83 (d, J=2.8 Hz, 1H), 8.50 (s, 1H), 7.90 (dd, J=10.1, 6.2 Hz, 1H), 7.81 (dd, J=8.4, 1.5 Hz, 1H), 7.65 (q, J=8.3 Hz, 2H), 7.55 (dd, J=10.0, 2.0 Hz, 1H), 7.49 (dd, J=11.0, 6.0 Hz, 1H), 7.37 (dd, J=8.3, 2.0 Hz, 1H), 5.67 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.61-4.50 (m, 2H), 4.48-4.37 (m, 2H), 3.85-3.69 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 641.0 (M+H+). Example 411: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-cyclopropoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-cyclopropoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1155. 1H NMR (400 MHz, DMSO-d6) δ 8.39 (d, J=1.5 Hz, 1H), 8.00-7.89 (m, 2H), 7.84-7.71 (m, 4H), 7.60 (d, J=8.4 Hz, 1H), 7.53 (d, J=7.2 Hz, 1H), 7.37 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.59 (d, J=14.5 Hz, 3H), 4.50 (s, 2H), 4.40 (ddd, J=13.3, 8.4, 4.9 Hz, 2H), 4.19-4.09 (m, 1H), 4.04 (dd, J=10.4, 8.2 Hz, 1H), 3.87 (dd, J=10.2, 4.6 Hz, 1H), 2.91 (tt, J=5.9, 2.9 Hz, 1H), 0.22 (dd, J=10.9, 5.2 Hz, 1H), 0.11 (ddd, J=12.6, 9.1, 5.0 Hz, 2H), −0.04-−0.40 (m, 1H). ES/MS m/z: 641.0 (M+H+). Example 412: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-7 and I-1156. 1H NMR (400 MHz, DMSO-d6) δ 8.43 (s, 1H), 8.03-7.86 (m, 2H), 7.85-7.67 (m, 4H), 7.60 (d, J=8.5 Hz, 1H), 7.53 (d, J=7.3 Hz, 1H), 7.34 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.87-5.45 (m, 3H), 4.54 (s, 2H), 4.51-4.41 (m, 2H), 4.14 (s, OH), 4.05 (dd, J=10.5, 8.3 Hz, 1H), 3.93-3.83 (m, 2H), 3.62-3.43 (m, 1H), 3.24-2.99 (m, 1H). ES/MS m/z: 665.0 (M+H+). Example 413: (S)-2-(4-(6-((2-cyano-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Methyl (S)-2-(4-(6-((2-bromo-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (413-1): A mixture of Intermediate I-1219 (44 mg, 1.0 equivalent), 2-bromo-1-(bromomethyl)-4-(trifluoromethyl)benzene (30 mg, 1.05 equivalent), cesium carbonate (47 mg, 1.6 equivalent) and acetonitrile (3 mL) was stirred at 50 C for 1 hr. The mixture was filtered, concentrated and purified by silica gel flash column chromatography (EtOAc/hexane) to yield the title compound. ES/MS m/z: 730.0 (M+H+). Methyl (S)-2-(4-(6-((2-cyano-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (413-2): A mixture of methyl (S)-2-(4-(6-((2-bromo-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (413-1, 42 mg, 1.0 equivalent), zinc cyanide (20 mg, 3.0 equivalent), zinc(0) (38 mg, 1.0 equivalent), Pd(PPh3)4(20 mg, 0.30 equivalent), and DMF (3.8 mL) was purged with argon for 1 min, then stirred at 100° C. for 5 hr. Silica gel flash column chromatography (EtOAc/hexane) yielded the title compound. ES/MS m/z: 677.0 (M+H+). (S)-2-(4-(6-((2-cyano-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 413): A mixture of methyl (S)-2-(4-(6-((2-cyano-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (413-2, 38 mg, 1.0 equivalent), lithium hydroxide (0.3 M in water, 0.56 mL, 3.0 equivalent) and acetonitrile (0.50 mL) was stirred at 105° C. for 6 min. The mixture was quenched with acetic acid and purified by reverse-phase preparative HPLC (MeCN/water, 0.1% TFA) to yield the title compound. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.43 (d, J=1.9 Hz, 1H), 8.12 (dd, J=8.3, 1.9 Hz, 1H), 7.99-7.88 (m, 2H), 7.81 (dd, J=8.5, 1.5 Hz, 1H), 7.72 (dd, J=10.4, 6.4 Hz, 1H), 7.63 (d, J=8.5 Hz, 1H), 7.57 (dd, J=7.5, 1.6 Hz, 1H), 7.46 (dd, J=11.4, 6.0 Hz, 1H), 7.04 (d, J=8.3 Hz, 1H), 5.75 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.59-4.50 (m, 2H), 4.48-4.35 (m, 2H), 3.83-3.67 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 663.0 (M+H+). Example 414: (S)-2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)-4-(trifluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-(1H-1,2,3-triazol-1-yl)-4-(trifluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1157 and I-1219. 1H NMR (400 MHz, DMSO-d6) δ 9.04 (s, 1H), 8.97 (d, J=1.3 Hz, 1H), 8.50 (s, 1H), 8.41 (s, 1H), 8.06 (d, J=1.2 Hz, 1H), 7.93 (t, J=7.9 Hz, 1H), 7.87-7.76 (m, 2H), 7.63 (d, J=8.4 Hz, 1H), 7.57 (dd, J=7.5, 1.6 Hz, 1H), 7.47 (dd, J=11.4, 6.1 Hz, 1H), 7.02 (d, J=8.3 Hz, 1H), 5.77 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.55 (d, J=17.2 Hz, 2H), 4.49-4.35 (m, 2H), 3.88-3.58 (m, 2H), 1.32 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 706 (M+H+). Example 415: (S)-2-(4-(5-chloro-6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(5-chloro-6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1158 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 8.14 (d, J=8.1 Hz, 1H), 7.92-7.85 (m, 2H), 7.78 (dd, J=8.4, 1.5 Hz, 1H), 7.75-7.64 (m, 3H), 7.64-7.54 (m, 2H), 5.69 (s, 2H), 5.08 (d, J=6.6 Hz, 1H), 4.70-4.54 (m, 2H), 4.47 (dd, J=11.1, 6.7 Hz, 1H), 4.35 (d, J=17.3 Hz, 1H), 3.83-3.71 (m, 2H), 1.39 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 647.0 (M+H+). Example 416: (S)-2-(4-(5-chloro-6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(5-chloro-6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1159 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 8.15 (d, J=8.0 Hz, 1H), 7.95 (d, J=10.0 Hz, 1H), 7.77 (dd, J=5.8, 2.7 Hz, 3H), 7.71 (dd, J=10.6, 5.4 Hz, 1H), 7.63 (d, J=8.0 Hz, 1H), 7.59 (d, J=8.5 Hz, 1H), 5.72 (s, 2H), 5.09 (d, J=6.5 Hz, 1H), 4.65 (d, J=17.3 Hz, 1H), 4.57 (d, J=11.1 Hz, 1H), 4.47 (dd, J=11.1, 6.8 Hz, 1H), 4.35 (d, J=17.2 Hz, 1H), 3.85-3.75 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 665.0 (M+H+). Example 417: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1161 and I-102. 1H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J=6.5 Hz, 1H), 7.89 (t, J=7.8 Hz, 1H), 7.83 (dd, J=9.8, 7.0 Hz, 1H), 7.62 (d, J=8.2 Hz, 1H), 7.56-7.47 (m, 3H), 7.47-7.42 (m, 1H), 7.33 (dd, J=8.2, 2.0 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.00 (d, J=6.5 Hz, 1H), 4.52 (d, J=17.4 Hz, 2H), 4.47-4.30 (m, 2H), 3.82-3.67 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 640.0 (M+H+). Example 418: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1161. 1H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J=6.5 Hz, 1H), 7.96-7.83 (m, 3H), 7.77-7.65 (m, 3H), 7.53 (d, J=7.4 Hz, 1H), 7.45 (dd, J=10.2, 7.0 Hz, 2H), 7.01 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 5.00 (d, J=6.5 Hz, 1H), 4.51 (d, J=15.7 Hz, 2H), 4.47-4.30 (m, 2H), 3.81-3.69 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 613.0 (M+H+). Example 419: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-5-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1161. 1H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J=6.4 Hz, 1H), 7.98-7.85 (m, 2H), 7.83-7.69 (m, 3H), 7.54 (d, J=7.3 Hz, 1H), 7.46 (dd, J=10.8, 7.4 Hz, 2H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.00 (d, J=6.5 Hz, 1H), 4.52 (d, J=17.0 Hz, 2H), 4.47-4.31 (m, 2H), 3.82-3.69 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 631.0 (M+H+). Example 420: (S)-2-(4-(6-((6-(difluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((6-(difluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1219. 1H NMR (400 MHz, DMSO-d6) δ 8.83 (d, J=2.0 Hz, 1H), 8.50 (s, 1H), 8.12 (dd, J=8.1, 2.0 Hz, 1H), 7.91 (t, J=7.9 Hz, 1H), 7.88-7.78 (m, 2H), 7.74 (d, J=8.0 Hz, 1H), 7.63 (d, J=8.5 Hz, 1H), 7.55 (d, J=7.3 Hz, 1H), 7.47 (dd, J=10.9, 6.6 Hz, 1H), 7.16-6.78 (m, 2H), 5.61 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.55 (d, J=16.8 Hz, 2H), 4.50-4.32 (m, 2H), 3.95-3.65 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 621.0 (M+H+). Example 421: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-imidazo[4,5-c]pyridine-6-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-imidazo[4,5-c]pyridine-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1162. 1H NMR (400 MHz, DMSO-d6) δ 9.03 (s, 1H), 8.62 (s, 1H), 7.97-7.83 (m, 3H), 7.75 (dd, J=10.2, 6.5 Hz, 1H), 7.69 (d, J=8.1 Hz, 2H), 7.54 (d, J=7.3 Hz, 1H), 7.50 (dd, J=11.2, 6.3 Hz, 1H), 7.02 (d, J=8.3 Hz, 1H), 5.59 (s, 2H), 5.09 (d, J=6.3 Hz, 1H), 4.62 (d, J=17.3 Hz, 1H), 4.54 (d, J=11.2 Hz, 1H), 4.49-4.37 (m, 2H), 3.80-3.71 (m, 2H), 1.37 (s, 3H), 0.63 (s, 3H). ES/MS m/z: 596.0 (M+H+). Example 422: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-imidazo[4,5-c]pyridine-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-imidazo[4,5-c]pyridine-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1162. 1H NMR (400 MHz, DMSO-d6) δ 9.02 (s, 1H), 8.61 (s, 1H), 7.98-7.88 (m, 2H), 7.84-7.71 (m, 3H), 7.55 (d, J=7.4 Hz, 1H), 7.50 (dd, J=11.0, 6.4 Hz, 1H), 7.01 (d, J=8.2 Hz, 1H), 5.61 (s, 2H), 5.09 (d, J=6.1 Hz, 1H), 4.62 (d, J=17.3 Hz, 1H), 4.54 (d, J=11.2 Hz, 1H), 4.50-4.36 (m, 2H), 3.80-3.74 (m, 2H), 1.37 (s, 3H), 0.63 (s, 3H). ES/MS m/z: 614.0 (M+H+). Example 423: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-imidazo[4,5-c]pyridine-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-imidazo[4,5-c]pyridine-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1162. 1H NMR (400 MHz, DMSO-d6) δ 9.01 (s, 1H), 8.61 (s, 1H), 7.96-7.80 (m, 2H), 7.61 (t, J=8.2 Hz, 1H), 7.58-7.45 (m, 3H), 7.34 (dd, J=8.2, 1.9 Hz, 1H), 6.97 (d, J=8.2 Hz, 1H), 5.52 (s, 2H), 5.09 (d, J=6.2 Hz, 1H), 4.62 (d, J=17.1 Hz, 1H), 4.54 (d, J=11.2 Hz, 1H), 4.50-4.38 (m, 2H), 3.79-3.73 (m, 2H), 1.37 (s, 3H), 0.63 (s, 3H). ES/MS m/z: 623.0 (M+H+). Example 424: (S)-2-(2,5-difluoro-4-(6-((6-(2,2,2-trifluoroethoxy)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((6-(2,2,2-trifluoroethoxy)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1219. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.36 (d, J=2.2 Hz, 1H), 7.95 (dd, J=8.5, 2.4 Hz, 1H), 7.88 (td, J=8.9, 7.9, 4.5 Hz, 2H), 7.84-7.77 (m, 1H), 7.62 (d, J=8.5 Hz, 1H), 7.53 (d, J=7.3 Hz, 1H), 7.47 (dd, J=10.8, 6.8 Hz, 1H), 7.02 (d, J=8.5 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.47 (s, 2H), 5.00 (q, J=8.8 Hz, 3H), 4.60-4.48 (m, 2H), 4.48-4.32 (m, 2H), 3.83-3.71 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 669.0 (M+H+). Example 425: (S)-2-(2,5-difluoro-4-(6-((6-(trifluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((6-(trifluoromethyl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1219. 1H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J=2.0 Hz, 1H), 8.49 (s, 1H), 8.20 (d, J=8.1 Hz, 1H), 7.93 (dd, J=17.1, 8.2 Hz, 2H), 7.79 (dd, J=9.3, 7.5 Hz, 2H), 7.62 (d, J=8.5 Hz, 1H), 7.55 (d, J=7.4 Hz, 1H), 7.46 (dd, J=10.7, 6.8 Hz, 1H), 7.02 (d, J=8.3 Hz, 1H), 5.65 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.58-4.49 (m, 2H), 4.49-4.30 (m, 2H), 3.85-3.71 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 639.0 (M+H+). Example 426: (S)-2-(2,5-difluoro-4-(6-((1-methyl-2-oxo-1,2-dihydropyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((1-methyl-2-oxo-1,2-dihydropyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1164 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.92-7.79 (m, 3H), 7.71 (dd, J=6.8, 2.0 Hz, 1H), 7.65 (d, J=8.4 Hz, 1H), 7.57-7.50 (m, 2H), 7.46 (dd, J=11.3, 6.1 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 6.25 (t, J=6.8 Hz, 1H), 5.29 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.61-4.52 (m, 2H), 4.48-4.36 (m, 2H), 3.86-3.68 (m, 2H), 3.48 (s, 3H), 1.34 (s, 3H), 0.61 (d, J=3.4 Hz, 3H). ES/MS m/z: 601.0 (M+H+). Example 427: (S)-2-(2,5-difluoro-4-(6-((1-methyl-2-oxo-1,2-dihydropyridin-4-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((1-methyl-2-oxo-1,2-dihydropyridin-4-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1165 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.91 (t, J=7.9 Hz, 1H), 7.80 (dd, J=9.3, 7.8 Hz, 2H), 7.68 (d, J=7.0 Hz, 1H), 7.63 (d, J=8.5 Hz, 1H), 7.54 (d, J=7.4 Hz, 1H), 7.46 (dd, J=10.1, 7.4 Hz, 1H), 7.02 (d, J=8.3 Hz, 1H), 6.42 (s, 1H), 6.29 (dd, J=7.0, 1.8 Hz, 1H), 5.34 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.58-4.50 (m, 2H), 4.48-4.35 (m, 2H), 3.85-3.66 (m, 2H), 3.40 (s, 3H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 601.0 (M+H+). Example 428: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1167. 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J=8.3 Hz, 1H), 7.95 (dd, J=9.3, 7.6 Hz, 2H), 7.88 (dd, J=10.2, 8.2 Hz, 1H), 7.81-7.70 (m, 3H), 7.59-7.53 (m, 1H), 7.47 (dd, J=11.4, 6.1 Hz, 1H), 5.70 (s, 2H), 4.96 (dd, J=8.5, 5.3 Hz, 1H), 4.74 (dd, J=9.6, 5.2 Hz, 1H), 4.65-4.44 (m, 2H), 4.37 (dt, J=8.9, 5.1 Hz, 2H), 3.63 (d, J=7.9 Hz, 1H), 1.24 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 632.0 (M+H+). Example 429: (S)-2-(2,5-difluoro-4-(6-((3-fluoropyridin-4-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((3-fluoropyridin-4-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1168 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.61 (d, J=1.7 Hz, 1H), 8.50 (s, 1H), 8.45 (d, J=4.9 Hz, 1H), 7.92 (t, J=7.9 Hz, 1H), 7.82 (dd, J=8.4, 1.4 Hz, 1H), 7.74 (dd, J=10.5, 6.4 Hz, 1H), 7.64 (d, J=8.5 Hz, 1H), 7.61-7.53 (m, 2H), 7.46 (dd, J=11.4, 6.1 Hz, 1H), 7.03 (d, J=8.2 Hz, 1H), 5.62 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.55 (d, J=16.4 Hz, 2H), 4.51-4.35 (m, 2H), 3.83-3.71 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 589.0 (M+H+). Example 430: (S)-2-(2,5-difluoro-4-(6-((3-methoxypyridin-4-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((3-methoxypyridin-4-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1169 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.61 (s, 1H), 8.49 (s, 1H), 8.44 (d, J=5.3 Hz, 1H), 7.94 (t, J=7.9 Hz, 1H), 7.81 (dd, J=8.5, 1.4 Hz, 1H), 7.75 (d, J=5.4 Hz, 1H), 7.67 (t, J=8.4 Hz, 1H), 7.62 (d, J=8.4 Hz, 1H), 7.56 (d, J=7.4 Hz, 1H), 7.46 (t, J=8.7 Hz, 1H), 7.09 (d, J=8.3 Hz, 1H), 5.63 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.60-4.48 (m, 2H), 4.48-4.31 (m, 2H), 4.05 (s, 3H), 3.87-3.65 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 601.1 (M+H+). Example 431: (S)-2-(4-(6-((4-cyclopropylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyclopropylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1170 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.94-7.75 (m, 3H), 7.64 (d, J=8.4 Hz, 1H), 7.58-7.41 (m, 2H), 7.36 (d, J=7.8 Hz, 2H), 7.09 (d, J=7.8 Hz, 2H), 6.92 (d, J=8.2 Hz, 1H), 5.41 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.55 (d, J=16.6 Hz, 2H), 4.50-4.33 (m, 2H), 3.86-3.67 (m, 2H), 1.91 (tt, J=8.7, 5.0 Hz, 1H), 1.34 (s, 3H), 0.99-0.85 (m, 2H), 0.66 (dt, J=6.5, 4.6 Hz, 2H), 0.61 (s, 3H). ES/MS m/z: 610.0 (M+H+). Example 432: (S)-2-(4-(5-chloro-6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(5-chloro-6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1158 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.09 (d, J=8.0 Hz, 1H), 7.90 (d, J=8.2 Hz, 2H), 7.82-7.74 (m, 2H), 7.70 (d, J=8.0 Hz, 2H), 7.62 (d, J=8.4 Hz, 1H), 7.60-7.52 (m, 1H), 7.48 (dd, J=11.4, 6.1 Hz, 1H), 5.69 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.59-4.51 (m, 2H), 4.51-4.34 (m, 2H), 3.82-3.67 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 629.0 (M+H+). Example 433: (S)-2-(4-(5-chloro-6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(5-chloro-6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1172 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.07 (d, J=8.0 Hz, 1H), 7.88-7.78 (m, 2H), 7.62 (d, J=8.5 Hz, 1H), 7.58-7.51 (m, 3H), 7.51-7.41 (m, 3H), 5.58 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.57-4.50 (m, 2H), 4.49-4.33 (m, 2H), 3.83-3.68 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 638.0 (M+H+). Example 434: (S)-2-(4-(5-chloro-6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(5-chloro-6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1158 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.10 (d, J=8.0 Hz, 1H), 7.95 (d, J=10.0 Hz, 1H), 7.83-7.74 (m, 4H), 7.62 (d, J=8.5 Hz, 1H), 7.58 (dd, J=8.1, 1.4 Hz, 1H), 7.48 (dd, J=11.4, 6.1 Hz, 1H), 5.71 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.34 (m, 2H), 3.83-3.68 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 647.0 (M+H+). Example 435: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(hydroxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 436: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(hydroxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 435 and Example 436 were prepared via preparative chiral SFC (Daicel Chiralpak AD-H column, MeOH—CO2eluent) of Example 216. 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(hydroxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 435) was isolated as the later-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.98-7.85 (m, 2H), 7.85-7.70 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.37 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.53 (d, J=7.6 Hz, 1H), 4.55 (s, 2H), 4.50 (d, J=10.8 Hz, 1H), 4.21 (dd, J=10.9, 6.5 Hz, 1H), 4.06 (t, J=8.7 Hz, 1H), 3.80 (t, J=8.3 Hz, 1H), 3.18-3.09 (m, 1H), 3.00 (q, J=7.8 Hz, 1H), 2.75 (t, J=9.5 Hz, 1H). ES/MS m/z: 615.0 (M+H+). 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(hydroxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 436) was isolated as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.98-7.85 (m, 2H), 7.85-7.70 (m, 4H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H), 7.37 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 5.53 (d, J=7.6 Hz, 1H), 4.55 (s, 2H), 4.50 (d, J=10.8 Hz, 1H), 4.21 (dd, J=10.9, 6.5 Hz, 1H), 4.06 (t, J=8.7 Hz, 1H), 3.80 (t, J=8.3 Hz, 1H), 3.18-3.09 (m, 1H), 3.00 (q, J=7.8 Hz, 1H), 2.75 (t, J=9.5 Hz, 1H). ES/MS m/z: 615.0 (M+H+). Example 437: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates 102 and I-1167. 1H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J=8.3 Hz, 1H), 7.96 (d, J=8.3 Hz, 1H), 7.92-7.80 (m, 2H), 7.60 (t, J=8.2 Hz, 1H), 7.57-7.44 (m, 3H), 7.33 (dd, J=8.2, 2.0 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 4.97 (dd, J=8.4, 5.3 Hz, 1H), 4.74 (dd, J=9.6, 5.2 Hz, 1H), 4.64-4.47 (m, 2H), 4.37 (dt, J=8.9, 5.1 Hz, 2H), 3.63 (d, J=7.9 Hz, 1H), 1.24 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 623.0 (M+H+). Example 438: (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid (S)-2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1248 and I-1167. 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J=8.3 Hz, 1H), 7.96 (d, J=8.3 Hz, 1H), 7.88 (t, J=7.9 Hz, 1H), 7.82 (dd, J=10.3, 6.6 Hz, 1H), 7.58-7.40 (m, 6H), 6.96 (d, J=8.3 Hz, 1H), 5.47 (s, 2H), 4.97 (dd, J=8.4, 5.3 Hz, 1H), 4.75 (dd, J=9.6, 5.2 Hz, 1H), 4.65-4.45 (m, 2H), 4.37 (dt, J=8.9, 5.1 Hz, 2H), 3.63 (d, J=7.9 Hz, 1H), 1.24 (s, 3H), 0.63 (s, 3H). ES/MS m/z: 605.0 (M+H+). Example 439: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1167. 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J=8.3 Hz, 1H), 8.00-7.85 (m, 3H), 7.84-7.68 (m, 3H), 7.58-7.52 (m, 1H), 7.46 (dd, J=11.3, 6.2 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.61 (s, 2H), 4.96 (dd, J=8.4, 5.3 Hz, 1H), 4.74 (dd, J=9.6, 5.1 Hz, 1H), 4.64-4.46 (m, 2H), 4.37 (dt, J=8.9, 5.1 Hz, 2H), 3.63 (d, J=7.9 Hz, 1H), 1.23 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 614.0 (M+H+). Example 440: (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid (S)-2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-3-(4,4-dimethyltetrahydrofuran-3-yl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-114 and I-1167. 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J=8.3 Hz, 1H), 7.96 (d, J=8.3 Hz, 1H), 7.93-7.83 (m, 3H), 7.74 (dd, J=10.2, 6.6 Hz, 1H), 7.68 (d, J=8.0 Hz, 2H), 7.58-7.51 (m, 1H), 7.46 (dd, J=11.2, 6.3 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.58 (s, 2H), 4.96 (dd, J=8.5, 5.2 Hz, 1H), 4.74 (dd, J=9.6, 5.1 Hz, 1H), 4.64-4.44 (m, 2H), 4.37 (dt, J=9.0, 5.1 Hz, 2H), 3.63 (d, J=7.9 Hz, 1H), 1.24 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 596.0 (M+H+). Example 441: (S)-4-chloro-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-4-chloro-2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1246 and I-1037. 1H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 7.96-7.74 (m, 3H), 7.55 (d, J=8.3 Hz, 3H), 7.51-7.39 (m, 3H), 5.57 (s, 2H), 5.03 (d, J=6.5 Hz, 1H), 4.54 (t, J=13.8 Hz, 2H), 4.47-4.36 (m, 2H), 3.84-3.68 (m, 2H), 1.32 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 656.0 (M+H+). Example 442: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-1,2,3-triazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Procedure 41 Methyl (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-1,2,3-triazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (442-1): A mixture of methyl (S)-2-(4-(6-((4-bromo-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (I-1220, 50 mg, 1.0 equivalent), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)triazole (31 mg, 2.0 equivalent), sodium carbonate (2 M in water, 74 μL, 2.0 equivalent), Pd(dppf)Cl2(5.2 mg, 0.10 equivalent), and 1,4-dioxane (1 mL) was degassed with argon, then stirred at 120 C for 1 hr. The mixture was filtered and then concentrated in vacuo. For this particular example the crude was carried forward without further purification. However, other examples could be purified by preparative HPLC (MeCN in water with 0.1% TFA), or by silica gel flash column chromatography (EtOAc/hexane) to yield the title compound. (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-1,2,3-triazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 442): Crude methyl (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-1,2,3-triazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (442-1) (30 mg, 1.0 equivalent) was mixed with 1 mL of MeCN and 0.3 mL of 2 N aqueous lithium hydroxide. The mixture was heated to 80° C. for 30 minutes. Then 0.3 mL of acetic acid was added to the mixture. The mixture was filtered and purified by reverse phase preparative HPLC (eluent: water/MeCN 0.1% TFA) to give title compound. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J=8.6, 1.4 Hz, 1H), 7.96-7.81 (m, 3H), 7.81-7.67 (m, 2H), 7.59 (dd, J=7.3, 1.6 Hz, 1H), 7.49-7.35 (m, 3H), 7.06-6.75 (m, 1H), 5.64 (s, 2H), 5.14 (d, J=6.5 Hz, 1H), 4.80-4.61 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.13 (s, 3H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). Example 443: (S)-2-(4-(6-((4-(1-(difluoromethyl)-1H-pyrazol-4-yl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-(1-(difluoromethyl)-1H-pyrazol-4-yl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediate I-1220 and 1-(difluoromethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole. 1H NMR (400 MHz, Methanol-d4) δ 8.93 (s, 1H), 8.49 (d, J=0.7 Hz, 1H), 8.21 (dd, J=8.7, 1.4 Hz, 1H), 8.11 (s, 1H), 7.95 (dd, J=10.9, 6.3 Hz, 1H), 7.89-7.72 (m, 2H), 7.71-7.31 (m, 6H), 6.93 (dd, J=8.3, 0.7 Hz, 1H), 5.57 (s, 2H), 5.17 (d, J=6.4 Hz, 1H), 4.81-4.57 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 704.1 (M+H+). Example 444: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(2-methylthiazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(2-methylthiazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediates I-1220 and 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)thiazole. 1H NMR (400 MHz, Methanol-d4) δ 8.93 (s, 1H), 8.21 (dd, J=8.6, 1.4 Hz, 1H), 8.01-7.90 (m, 2H), 7.90-7.75 (m, 2H), 7.59 (ddd, J=8.2, 5.0, 3.4 Hz, 2H), 7.51-7.32 (m, 3H), 7.02-6.87 (m, 1H), 5.58 (s, 2H), 5.17 (d, J=6.5 Hz, 1H), 4.83-4.61 (m, 3H), 4.52 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=9.0 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 2.74 (s, 3H), 1.41 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 685.1 (M+H+). Example 445: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(2-methyloxazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(2-methyloxazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediates I-1220 and 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)oxazole. 1H NMR (400 MHz, Methanol-d4) δ 8.93 (s, 1H), 8.21 (dd, J=8.6, 1.4 Hz, 1H), 7.94 (dd, J=10.9, 6.3 Hz, 1H), 7.90-7.76 (m, 2H), 7.67-7.53 (m, 2H), 7.54-7.37 (m, 4H), 6.93 (d, J=8.2 Hz, 1H), 5.57 (s, 2H), 5.17 (d, J=6.4 Hz, 1H), 4.83-4.61 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 2.53 (s, 3H), 1.41 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 669.2 (M+H+). Example 446: (S)-2-(4-(6-((2-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-1219 and 2-(chloromethyl)benzonitrile. 1H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J=8.6, 1.4 Hz, 1H), 7.94-7.80 (m, 2H), 7.79 (d, J=8.1 Hz, 2H), 7.75-7.66 (m, 2H), 7.58 (dd, J=7.5, 1.6 Hz, 1H), 7.51 (qd, J=7.6, 1.6 Hz, 1H), 7.40 (dd, J=11.2, 6.0 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.68 (s, 2H), 5.15 (d, J=6.5 Hz, 1H), 4.79-4.60 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 595.3 (M+H+). Example 447: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-4-(difluoromethyl)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-4-(difluoromethyl)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1222 and I-82. 1H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (dd, J=8.6, 1.4 Hz, 1H), 7.96 (dd, J=10.7, 6.3 Hz, 1H), 7.77 (d, J=8.6 Hz, 1H), 7.70 (s, 1H), 7.56 (t, J=8.0 Hz, 1H), 7.43 (dd, J=11.3, 6.0 Hz, 1H), 7.31-7.19 (m, 2H), 7.07 (s, 1H), 6.88 (t, J=55.4 Hz, 1H), 5.58 (s, 2H), 5.13 (d, J=6.6 Hz, 1H), 4.79-4.59 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 672.2 (M+H+). Example 448: (S)-2-(4-(6-((4-(1H-1,2,4-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-(1H-1,2,4-triazol-1-yl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-[4-(bromomethyl)phenyl]-1,2,4-triazole. 1H NMR (400 MHz, Methanol-d4) δ 9.10 (s, 1H), 8.90 (s, 1H), 8.23-8.08 (m, 2H), 7.91 (dd, J=10.9, 6.3 Hz, 1H), 7.88-7.81 (m, 3H), 7.78 (d, J=8.6 Hz, 1H), 7.68 (d, J=8.5 Hz, 2H), 7.58 (dd, J=7.5, 1.6 Hz, 1H), 7.40 (dd, J=11.2, 6.0 Hz, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.58 (s, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.79-4.57 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 637.0 (M+H+). Example 449: (S)-2-(4-(6-((4-chloro-2-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 2-(bromomethyl)-5-chloro-benzonitrile. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J=8.6, 1.5 Hz, 1H), 7.93-7.81 (m, 3H), 7.78 (d, J=8.6 Hz, 1H), 7.71 (d, J=1.6 Hz, 2H), 7.59 (dd, J=7.4, 1.6 Hz, 1H), 7.38 (dd, J=11.1, 6.0 Hz, 1H), 6.97 (d, J=8.2 Hz, 1H), 5.66 (s, 2H), 5.13 (d, J=6.6 Hz, 1H), 4.81-4.60 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.8 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 629.2 (M+H+). Example 450: (S)-2-(4-(6-((4-chloro-2-fluoro-5-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluoro-5-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-4-chloro-2-fluoro-5-methyl-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J=8.6, 1.3 Hz, 1H), 7.94 (dd, J=10.9, 6.3 Hz, 1H), 7.90-7.73 (m, 2H), 7.58 (d, J=7.5 Hz, 1H), 7.48 (d, J=7.8 Hz, 1H), 7.39 (dd, J=11.2, 6.0 Hz, 1H), 7.22 (d, J=9.7 Hz, 1H), 6.91 (d, J=8.3 Hz, 1H), 5.50 (s, 2H), 5.14 (d, J=6.3 Hz, 1H), 4.81-4.61 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 2.34 (s, 3H), 1.42 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 636.3 (M+H+). Example 451: (S)-2-(2,5-difluoro-4-(6-((2,3,4-trifluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2,3,4-trifluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-2,3,4-trifluoro-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.20 (dd, J=8.6, 1.4 Hz, 1H), 7.94 (dd, J=10.8, 6.3 Hz, 1H), 7.89-7.74 (m, 2H), 7.59 (dd, J=7.5, 1.6 Hz, 1H), 7.48-7.31 (m, 2H), 7.13 (tdd, J=9.2, 7.1, 2.1 Hz, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.55 (s, 2H), 5.16 (d, J=6.6 Hz, 1H), 4.83-4.60 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.01 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 624.3 (M+H+). Example 452: (S)-2-(4-(6-((3-chloro-4-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((3-chloro-4-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 2-chloro-4-(chloromethyl)-1-methyl-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.93 (s, 1H), 8.21 (d, J=8.5 Hz, 1H), 7.90 (ddd, J=11.1, 6.5, 4.8 Hz, 1H), 7.87-7.77 (m, 2H), 7.64-7.53 (m, 1H), 7.47-7.39 (m, 2H), 7.38-7.12 (m, 2H), 7.02-6.78 (m, 1H), 5.66-5.39 (m, 2H), 5.17 (d, J=6.5 Hz, 1H), 4.81-4.60 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.01 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 2.52-2.31 (m, 3H), 1.42 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 618.1 (M+H+). Example 453: (S)-2-(4-(6-((4-chloro-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 2-(bromomethyl)-5-chloro-1,3-difluoro-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.18 (dd, J=8.6, 1.3 Hz, 1H), 8.04 (dd, J=10.9, 6.3 Hz, 1H), 7.91-7.65 (m, 2H), 7.67-7.44 (m, 1H), 7.40 (dd, J=11.2, 6.0 Hz, 1H), 7.17 (d, J=7.3 Hz, 2H), 6.85 (d, J=8.3 Hz, 1H), 5.55 (s, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.81-4.60 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 640.2 (M+H+). Example 454: (S)-2-(4-(6-((2-chloro-4,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2-chloro-4,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-2-chloro-4,5-difluoro-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 1H), 8.15 (dd, J=8.6, 1.4 Hz, 1H), 7.91-7.79 (m, 2H), 7.76 (d, J=8.5 Hz, 1H), 7.60 (dd, J=7.5, 1.6 Hz, 1H), 7.56-7.44 (m, 2H), 7.37 (dd, J=11.2, 6.0 Hz, 1H), 6.98 (d, J=8.2 Hz, 1H), 5.55 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.76-4.60 (m, 3H), 4.52 (dd, J=11.6, 6.8 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.8 Hz, 1H), 1.41 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 640.3 (M+H+). Example 455: (S)-2-(2,5-difluoro-4-(6-((2,4,5-trifluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2,4,5-trifluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(chloromethyl)-2,4,5-trifluoro-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (dd, J=8.6, 1.4 Hz, 1H), 7.93 (dd, J=10.8, 6.3 Hz, 1H), 7.83 (t, J=7.9 Hz, 1H), 7.77 (d, J=8.6 Hz, 1H), 7.59 (dd, J=7.4, 1.6 Hz, 1H), 7.49 (ddd, J=10.8, 8.9, 6.6 Hz, 1H), 7.38 (dd, J=11.2, 6.0 Hz, 1H), 7.22 (td, J=10.1, 6.6 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.52 (s, 2H), 5.21-5.06 (m, 1H), 4.78-4.58 (m, 3H), 4.53 (dd, J=11.5, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 624.1 (M+H+). Example 456: (S)-2-(4-(6-((2,5-difluoro-4-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2,5-difluoro-4-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-2,5-difluoro-4-methyl-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.18 (dd, J=8.6, 1.4 Hz, 1H), 7.93 (dd, J=10.9, 6.3 Hz, 1H), 7.89-7.73 (m, 2H), 7.58 (dd, J=7.5, 1.5 Hz, 1H), 7.39 (dd, J=11.2, 6.0 Hz, 1H), 7.20 (dd, J=9.7, 6.0 Hz, 1H), 7.04 (dd, J=10.1, 6.1 Hz, 1H), 6.92 (d, J=8.3 Hz, 1H), 5.50 (s, 2H), 5.14 (d, J=6.6 Hz, 1H), 4.81-4.60 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 2.26 (d, J=1.9 Hz, 3H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 620.3 (M+H+). Example 457: (S)-2-(4-(6-((3,4-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((3,4-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 4-(chloromethyl)-1,2-difluoro-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (dd, J=8.6, 1.4 Hz, 1H), 7.91 (dd, J=10.8, 6.3 Hz, 1H), 7.87-7.73 (m, 2H), 7.57 (dd, J=7.4, 1.6 Hz, 1H), 7.46-7.35 (m, 2H), 7.35-7.17 (m, 2H), 6.93 (d, J=8.3 Hz, 1H), 5.47 (s, 2H), 5.13 (d, J=6.5 Hz, 1H), 4.82-4.60 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 606.2 (M+H+). Example 458: (S)-2-(2,5-difluoro-4-(6-((2,4,6-trifluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2,4,6-trifluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 2-(bromomethyl)-1,3,5-trifluoro-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J=8.7, 1.4 Hz, 1H), 8.05 (dd, J=10.9, 6.3 Hz, 1H), 7.88-7.75 (m, 2H), 7.59 (dd, J=7.5, 1.6 Hz, 1H), 7.40 (dd, J=11.3, 6.0 Hz, 1H), 7.01-6.89 (m, 2H), 6.84 (d, J=8.3 Hz, 1H), 5.54 (s, 2H), 5.14 (d, J=7.1 Hz, 1H), 4.81-4.60 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 624.3 (M+H+). Example 459: (S)-2-(4-(6-((5-chloro-2-fluoro-4-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((5-chloro-2-fluoro-4-methylbenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-chloro-5-(chloromethyl)-4-fluoro-2-methyl-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 1H), 8.14 (dd, J=8.6, 1.4 Hz, 1H), 7.92 (dd, J=10.8, 6.3 Hz, 1H), 7.86-7.73 (m, 2H), 7.57 (dd, J=7.5, 1.6 Hz, 1H), 7.53 (d, J=6.7 Hz, 1H), 7.36 (dd, J=11.2, 6.0 Hz, 1H), 7.12 (d, J=10.4 Hz, 1H), 6.92 (d, J=8.3 Hz, 1H), 5.50 (s, 2H), 5.11 (d, J=6.5 Hz, 1H), 4.79-4.58 (m, 3H), 4.52 (dd, J=11.5, 6.7 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.8 Hz, 1H), 2.37 (s, 3H), 1.41 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 636.2 (M+H+). Example 460: (S)-2-(4-(6-((2-chloro-4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2-chloro-4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 4-(bromomethyl)-3-chloro-benzonitrile. 1H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (d, J=8.6 Hz, 1H), 7.98-7.83 (m, 2H), 7.83-7.66 (m, 4H), 7.60 (d, J=7.4 Hz, 1H), 7.37 (dd, J=11.2, 6.0 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.67 (s, 2H), 5.12 (d, J=6.4 Hz, 1H), 4.74-4.58 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 629.15 (M+H+). Example 461: (S)-2-(4-(6-((4-cyano-2-methoxybenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-methoxybenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 4-(bromomethyl)-3-methoxy-benzonitrile. 1H NMR (400 MHz, Methanol-d4) δ 8.84 (s, 1H), 8.12 (dd, J=8.6, 1.4 Hz, 1H), 7.89-7.77 (m, 2H), 7.75 (d, J=8.6 Hz, 1H), 7.58 (d, J=7.7 Hz, 2H), 7.38 (d, J=1.4 Hz, 1H), 7.37-7.26 (m, 2H), 6.96 (d, J=8.3 Hz, 1H), 5.58 (s, 2H), 5.08 (d, J=6.6 Hz, 1H), 4.74-4.47 (m, 4H), 3.97 (d, J=2.4 Hz, 4H), 3.84 (d, J=8.9 Hz, 1H), 1.40 (s, 3H), 0.74 (s, 3H). ES/MS m/z: 625.2 (M+H+). Example 462: (S)-2-(4-(6-((4-chloro-2-methoxybenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-methoxybenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-4-chloro-2-methoxy-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.77 (s, 1H), 8.10-8.01 (m, 1H), 7.88 (dd, J=10.9, 6.3 Hz, 1H), 7.79 (t, J=7.9 Hz, 1H), 7.72 (d, J=8.5 Hz, 1H), 7.55 (dd, J=7.5, 1.5 Hz, 1H), 7.39 (d, J=8.1 Hz, 1H), 7.27 (dd, J=11.3, 6.1 Hz, 1H), 7.05 (d, J=1.9 Hz, 1H), 6.95 (dd, J=8.1, 2.0 Hz, 1H), 6.88 (d, J=8.2 Hz, 1H), 5.48 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.69-4.46 (m, 4H), 3.97 (d, J=8.8 Hz, 1H), 3.91 (s, 3H), 3.82 (d, J=8.8 Hz, 1H), 1.39 (s, 3H), 0.71 (s, 3H). ES/MS m/z: 634.3 (M+H+). Example 463: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-methylbenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-methylbenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-2-fluoro-4-methyl-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.96 (s, 1H), 8.23 (dd, J=8.6, 1.4 Hz, 1H), 7.96 (dd, J=10.9, 6.3 Hz, 1H), 7.89-7.74 (m, 2H), 7.56 (dd, J=7.5, 1.6 Hz, 1H), 7.51-7.33 (m, 2H), 6.97 (dd, J=14.8, 9.5 Hz, 2H), 6.88 (d, J=8.2 Hz, 1H), 5.49 (s, 2H), 5.20 (d, J=6.4 Hz, 1H), 4.89-4.60 (m, 3H), 4.53 (dd, J=11.8, 6.6 Hz, 1H), 4.01 (d, J=9.0 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 2.34 (s, 3H), 1.42 (s, 3H), 0.78 (s, 3H). ES/MS m/z: 602.3 (M+H+). Example 464: (S)-2-(4-(6-((2-chloro-4-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2-chloro-4-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-2-chloro-4-fluoro-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.94 (s, 1H), 8.21 (dd, J=8.7, 1.4 Hz, 1H), 7.91 (dd, J=10.9, 6.3 Hz, 1H), 7.88-7.73 (m, 2H), 7.68-7.54 (m, 2H), 7.43 (dd, J=11.1, 6.1 Hz, 1H), 7.28 (dd, J=8.6, 2.6 Hz, 1H), 7.10 (td, J=8.5, 2.6 Hz, 1H), 6.94 (d, J=8.3 Hz, 1H), 5.56 (s, 2H), 5.18 (d, J=6.5 Hz, 1H), 4.85-4.60 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 622.2 (M+H+). Example 465: (S)-2-(2,5-difluoro-4-(6-((4-fluoro-2-methylbenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((4-fluoro-2-methylbenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-4-fluoro-2-methyl-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.20 (dd, J=8.6, 1.4 Hz, 1H), 7.95 (dd, J=10.8, 6.4 Hz, 1H), 7.85-7.70 (m, 2H), 7.56 (dd, J=7.4, 1.7 Hz, 1H), 7.43 (td, J=10.9, 9.7, 5.9 Hz, 2H), 6.99 (dd, J=9.9, 2.8 Hz, 1H), 6.90 (dd, J=11.5, 8.4 Hz, 2H), 5.47 (s, 2H), 5.16 (d, J=6.4 Hz, 1H), 4.83-4.61 (m, 3H), 4.53 (dd, J=11.7, 6.7 Hz, 1H), 4.01 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 2.43 (s, 3H), 1.42 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 602.3 (M+H+). Example 466: (S)-2-(4-(6-((2,4-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2,4-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-1219 and 1-(bromomethyl)-2,4-difluoro-benzene. 1H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J=8.6, 1.4 Hz, 1H), 7.96 (dd, J=10.8, 6.3 Hz, 1H), 7.89-7.70 (m, 2H), 7.65-7.52 (m, 2H), 7.41 (dd, J=11.2, 6.0 Hz, 1H), 7.10-6.94 (m, 2H), 6.90 (d, J=8.2 Hz, 1H), 5.52 (s, 2H), 5.16 (d, J=6.4 Hz, 1H), 4.82-4.60 (m, 3H), 4.53 (dd, J=11.6, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.85 (d, J=8.9 Hz, 1H), 1.42 (s, 3H), 0.77 (s, 3H). ES/MS m/z: 606.3 (M+H+). Example 467: 2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1267 and I-1092. 1H NMR (400 MHz, DMSO) δ 7.89 (dd, J=16.6, 8.2 Hz, 3H), 7.70 (dd, J=12.0, 7.3 Hz, 2H), 7.52 (d, J=7.4 Hz, 1H), 7.39-7.30 (m, 1H), 7.01 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 5.49 (s, 1H), 4.48 (t, J=13.9 Hz, 2H), 4.35 (t, J=5.1 Hz, 1H), 4.26-4.14 (m, 1H), 2.92 (s, 3H), 2.08 (s, 2H). ES/MS m/z: 629.1 (M+H+). Example 468: 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3 S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1267 and I-114. 1H NMR (400 MHz, DMSO) δ 7.89 (dd, J=16.6, 8.2 Hz, 3H), 7.70 (dd, J=12.0, 7.3 Hz, 2H), 7.52 (d, J=7.4 Hz, 1H), 7.40-7.29 (m, 1H), 7.01 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 5.49 (s, 1H), 4.48 (t, J=13.9 Hz, 2H), 4.35 (t, J=5.1 Hz, 1H), 4.29-4.16 (m, 1H), 2.92 (s, 3H), 2.08 (s, 2H). ES/MS m/z: 611.2 (M+H+). Example 469: (S)-2-(4-(6-((4-chloro-2-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1260. 1H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.97-7.85 (m, 2H), 7.85-7.73 (m, 3H), 7.70 (dd, J=10.5, 6.4 Hz, 1H), 7.62 (d, J=8.5 Hz, 1H), 7.55 (d, J=7.3 Hz, 1H), 7.44 (dd, J=11.4, 6.1 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.64 (s, 2H), 5.00 (d, J=6.7 Hz, 1H), 4.63-4.47 (m, 2H), 4.47-4.31 (m, 2H), 3.80-3.66 (m, 2H), 2.55 (s, 3H), 1.32 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 672.1 (M+H+). Example 470: (S)-2-(2,5-difluoro-4-(6-((6-(trifluoromethyl)pyridazin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((6-(trifluoromethyl)pyridazin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1261. 1H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.29 (d, J=8.8 Hz, 1H), 8.10 (d, J=8.8 Hz, 1H), 7.94 (t, J=7.9 Hz, 1H), 7.83 (dd, J=8.5, 1.5 Hz, 1H), 7.73-7.51 (m, 3H), 7.45 (dd, J=11.4, 6.1 Hz, 1H), 7.09 (d, J=8.3 Hz, 1H), 5.90 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.63-4.31 (m, 4H), 3.85-3.65 (m, 2H), 1.32 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 640.0 (M+H+). Example 471: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 39, starting with Intermediates I-1256 and I-1292. 1H NMR (400 MHz, DMSO) δ 8.50 (s, 1H), 7.97-7.73 (m, 3H), 7.71-7.50 (m, 4H), 7.45 (dd, J=11.2, 6.0 Hz, 1H), 7.35 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.43 (s, 1H), 4.66-4.40 (m, 3H), 4.31-4.18 (m, 1H), 4.18-4.09 (m, 1H), 3.59 (d, J=8.8 Hz, 4H), 0.53 (d, J=7.0 Hz, 3H). ES/MS m/z: 626.0 (M+H+). Example 472: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3 S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 39, starting with Intermediates I-1255 and I-1292. 1H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 7.97-7.75 (m, 3H), 7.62 (d, J=8.7 Hz, 2H), 7.56-7.42 (m, 3H), 7.35-7.31 (m, 1H), 6.95 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.44 (t, J=7.2 Hz, 1H), 4.57-4.39 (m, 3H), 4.24 (dd, J=10.9, 6.7 Hz, 1H), 4.13 (t, J=8.5 Hz, 1H), 3.60 (t, J=8.5 Hz, 1H), 0.53 (d, J=7.0 Hz, 3H). ES/MS m/z: 608.0 (M+H+). Example 473: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid, isomer 1 (absolute stereochemistry not known) 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid, isomer 1 (absolute stereochemistry not known) was prepared in a manner as described in Procedure 39, starting with Intermediates I-1255 and I-1258. 1H NMR (400 MHz, DMSO) δ 8.47 (s, 1H), 7.98-7.70 (m, 3H), 7.61 (t, J=8.3 Hz, 2H), 7.57-7.46 (m, 2H), 7.39 (dd, J=11.5, 6.0 Hz, 1H), 7.33 (dd, J=8.3, 2.1 Hz, 1H), 6.96 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.33 (d, J=8.2 Hz, 1H), 4.73-4.38 (m, 4H), 4.30-4.11 (m, 2H), 3.63 (d, J=10.0 Hz, 1H), 2.97 (s, 3H), 1.43 (s, 3H). ES/MS m/z: 638.0 (M+H+). Example 474: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid, isomer 2 (absolute stereochemistry not known) 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxy-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid, isomer 2 (absolute stereochemistry not known) was prepared in a manner as described in Procedure 39 starting with Intermediates I-1255 and I-1257. 1H NMR (400 MHz, DMSO) δ 8.45 (s, 1H), 7.95-7.76 (m, 3H), 7.67-7.56 (m, 2H), 7.56-7.47 (m, 2H), 7.43-7.30 (m, 2H), 6.95 (d, J=8.3 Hz, 1H), 5.51 (s, 2H), 5.31 (s, 1H), 4.64-4.35 (m, 3H), 4.29-4.14 (m, 2H), 3.63 (d, J=10.1 Hz, 1H), 2.96 (s, 3H), 1.43 (s, 3H). ES/MS m/z: 638.0 (M+H+). Example 475: (S)-2-(4-(6-((2,6-difluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2,6-difluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1262. 1H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 7.96-7.77 (m, 3H), 7.65 (d, J=8.4 Hz, 1H), 7.60-7.38 (m, 3H), 7.19 (t, J=8.0 Hz, 2H), 5.63 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.67-4.29 (m, 4H), 3.85-3.69 (m, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 624.2 (M+H+). Example 476: (S)-2-(4-(6-((2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1263. 1H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.00-7.79 (m, 3H), 7.65 (d, J=8.4 Hz, 1H), 7.60-7.38 (m, 3H), 7.18 (t, J=7.9 Hz, 2H), 6.91 (d, J=8.3 Hz, 1H), 5.53 (s, 2H), 5.04 (d, J=6.6 Hz, 1H), 4.67-4.32 (m, 4H), 3.87-3.70 (m, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 606.2 (M+H+). Example 477: (S)-2-(2,5-difluoro-4-(5-fluoro-6-((2-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(5-fluoro-6-((2-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1264. 1H NMR (400 MHz, DMSO) δ 8.50 (s, 1H), 8.03-7.79 (m, 3H), 7.75-7.51 (m, 3H), 7.46 (dq, J=13.9, 7.0, 6.5 Hz, 2H), 7.26 (q, J=9.3, 7.7 Hz, 2H), 5.62 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.66-4.28 (m, 4H), 3.83-3.66 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 606.2 (M+H+). Example 478: (S)-2-(2,5-difluoro-4-(6-((2-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-1265. 1H NMR (400 MHz, DMSO) δ 8.52 (s, 1H), 7.98-7.77 (m, 3H), 7.78-7.36 (m, 5H), 7.36-7.13 (m, 2H), 6.96 (d, J=8.3 Hz, 1H), 5.53 (s, 2H), 5.05 (d, J=6.6 Hz, 1H), 4.72-4.37 (m, 4H), 3.90-3.66 (m, 2H), 1.34 (s, 3H), 0.62 (s, 3H). ES/MS m/z: 588.3 (M+H+). Example 479: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-82 and I-109. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.42 (s, 1H), 7.73 (t, J=7.7 Hz, 1H), 7.68-7.51 (m, 6H), 5.68 (s, 2H), 4.93 (d, J=6.6 Hz, 1H), 4.59 (dd, J=11.3, 1.5 Hz, 1H), 4.53-4.40 (m, 2H), 4.32 (d, J=17.2 Hz, 1H), 3.85 (d, J=8.7 Hz, 1H), 3.78 (d, J=8.7 Hz, 1H), 1.41 (s, 3H), 0.72 (s, 3H). ES/MS m/z: 667.2 (M+H+). Example 480: 2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1287 and I-1292. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.57 (s, 1H), 7.86 (dd, J=8.4, 1.5 Hz, 1H), 7.78-7.68 (m, 3H), 7.65 (d, J=8.1 Hz, 2H), 7.62-7.54 (m, 2H), 7.52 (ddd, J=8.3, 3.1, 1.4 Hz, 1H), 7.20 (dd, J=11.6, 6.1 Hz, 1H), 5.63 (s, 2H), 5.24 (t, J=7.2 Hz, 1H), 4.55 (dd, J=11.0, 1.4 Hz, 1H), 4.37 (s, 2H), 4.23 (dd, J=11.0, 6.7 Hz, 1H), 4.13 (t, J=8.5 Hz, 1H), 3.67 (t, J=8.4 Hz, 1H), 2.88 (hept, J=7.6 Hz, 1H), 0.57 (d, J=7.0 Hz, 3H). ES/MS m/z: 599.3 (M+H+). Example 481: 2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1294 and I-1292. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.56 (s, 1H), 7.87 (dd, J=8.5, 1.5 Hz, 1H), 7.70 (dd, J=10.7, 6.5 Hz, 1H), 7.65-7.48 (m, 5H), 7.20 (dd, J=11.6, 6.1 Hz, 1H), 5.65 (s, 2H), 5.23 (t, J=7.3 Hz, 1H), 4.56 (dd, J=10.9, 1.4 Hz, 1H), 4.37 (s, 2H), 4.23 (dd, J=10.9, 6.7 Hz, 1H), 4.13 (t, J=8.5 Hz, 1H), 3.68 (t, J=8.3 Hz, 1H), 2.87 (hept, J=7.7 Hz, 1H), 0.57 (d, J=7.0 Hz, 3H). ES/MS m/z: 635.2 (M+H+). Example 482: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1231. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.54 (s, 1H), 7.83 (dd, J=8.5, 1.5 Hz, 1H), 7.72 (t, J=7.7 Hz, 1H), 7.66-7.45 (m, 6H), 5.66 (s, 2H), 4.92 (d, J=6.7 Hz, 1H), 4.60 (dd, J=11.2, 1.5 Hz, 1H), 4.53-4.40 (m, 2H), 4.31 (d, J=17.1 Hz, 1H), 3.86 (d, J=8.7 Hz, 1H), 3.77 (d, J=8.7 Hz, 1H), 1.41 (s, 3H), 0.70 (s, 3H). ES/MS m/z: 649.2 (M+H+). Example 483: (S)-2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1294 and I-108. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.41 (s, 1H), 7.72 (dd, J=10.7, 6.4 Hz, 1H), 7.67-7.49 (m, 5H), 7.23 (dd, J=11.5, 6.1 Hz, 1H), 5.66 (s, 2H), 4.85 (d, J=6.7 Hz, 1H), 4.54 (dd, J=11.3, 1.5 Hz, 1H), 4.43 (dd, J=11.3, 6.8 Hz, 1H), 4.39-4.27 (m, 2H), 3.84 (d, J=8.8 Hz, 1H), 3.75 (d, J=8.8 Hz, 1H), 1.35 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 667.2 (M+H+). Example 484: 2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(2-((4-chloro-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-105 and I-1241. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.63 (d, J=5.1 Hz, 1H), 8.57 (s, 1H), 7.94 (dd, J=10.5, 6.3 Hz, 1H), 7.86 (dd, J=8.5, 1.5 Hz, 1H), 7.62-7.52 (m, 3H), 7.31-7.19 (m, 3H), 5.53 (s, 2H), 5.24 (t, J=7.2 Hz, 1H), 4.55 (dd, J=11.0, 1.4 Hz, 1H), 4.40 (s, 2H), 4.23 (dd, J=11.0, 6.7 Hz, 1H), 4.13 (t, J=8.5 Hz, 1H), 3.67 (t, J=8.4 Hz, 1H), 2.89 (hept, J=7.5 Hz, 1H), 0.57 (d, J=7.1 Hz, 3H). ES/MS m/z: 609.2 (M+H+). Example 485: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,3,6-trifluorobenzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1135 and I-1036. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.03 (d, J=1.3 Hz, 1H), 7.69-7.64 (m, 1H), 7.64-7.50 (m, 4H), 7.31-7.22 (m, 2H), 5.59 (s, 2H), 4.53 (t, J=5.0 Hz, 2H), 4.46 (s, 2H), 3.75 (t, J=5.0 Hz, 2H), 3.27 (s, 3H). ES/MS m/z: 636.2 (M+H+). Example 486: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-1292. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.57 (d, J=1.5 Hz, 1H), 7.86 (dd, J=8.5, 1.6 Hz, 1H), 7.75-7.68 (m, 2H), 7.62-7.53 (m, 4H), 7.51 (ddd, J=8.2, 3.1, 1.4 Hz, 1H), 7.20 (dd, J=11.6, 6.1 Hz, 1H), 5.66 (s, 2H), 5.24 (t, J=7.2 Hz, 1H), 4.54 (dd, J=11.0, 1.4 Hz, 1H), 4.37 (s, 2H), 4.23 (dd, J=11.0, 6.7 Hz, 1H), 4.13 (t, J=8.5 Hz, 1H), 3.67 (t, J=8.4 Hz, 1H), 2.87 (dq, J=15.2, 7.6 Hz, 1H), 0.57 (d, J=7.1 Hz, 3H). “ES/MS m/z: 617.2 (M+H+). Example 487: (S)-2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1295 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.55 (s, 1H), 7.86 (dd, J=8.5, 1.5 Hz, 1H), 7.83-7.78 (m, 1H), 7.75 (dd, J=10.7, 6.4 Hz, 1H), 7.62-7.57 (m, 1H), 7.53 (ddt, J=10.5, 9.2, 4.8 Hz, 3H), 7.22 (dd, J=11.5, 6.1 Hz, 1H), 6.92 (dd, J=8.3, 0.7 Hz, 1H), 5.58 (s, 2H), 4.84 (d, J=6.6 Hz, 1H), 4.56 (dd, J=11.2, 1.6 Hz, 1H), 4.42 (dd, J=11.2, 6.9 Hz, 1H), 4.36 (d, J=6.3 Hz, 2H), 3.85 (d, J=8.7 Hz, 1H), 3.75 (d, J=8.7 Hz, 1H), 1.34 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 631.2 (M+H+). Example 488: (S)-2-(4-(6-((4-chloro-2,3-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2,3-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1296 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.69 (s, 1H), 8.01 (dd, J=8.6, 1.5 Hz, 1H), 7.85 (dd, J=10.8, 6.4 Hz, 1H), 7.77 (dd, J=8.2, 7.5 Hz, 1H), 7.72 (d, J=8.5 Hz, 1H), 7.50 (dd, J=7.5, 1.7 Hz, 1H), 7.36-7.22 (m, 3H), 6.86 (d, J=8.2 Hz, 1H), 5.53 (d, J=1.3 Hz, 2H), 4.93 (d, J=6.6 Hz, 1H), 4.62-4.50 (m, 3H), 4.43 (dd, J=11.6, 6.8 Hz, 1H), 3.89 (d, J=8.9 Hz, 1H), 3.76 (d, J=8.9 Hz, 1H), 1.34 (s, 3H), 0.68 (s, 3H). ES/MS m/z: 640.2 (M+H+). Example 489: (S)-2-(4-(6-((4-chloro-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1297 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.62 (s, 1H), 7.98-7.89 (m, 2H), 7.75 (t, J=7.9 Hz, 1H), 7.65 (d, J=8.5 Hz, 1H), 7.50 (dd, J=7.5, 1.7 Hz, 1H), 7.26 (dd, J=11.5, 6.1 Hz, 1H), 7.14 (d, J=7.4 Hz, 2H), 6.78 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 4.89 (d, J=6.7 Hz, 1H), 4.55 (dd, J=11.4, 1.5 Hz, 1H), 4.49-4.37 (m, 3H), 3.87 (d, J=8.8 Hz, 1H), 3.75 (d, J=8.8 Hz, 1H), 1.34 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 640.2 (M+H+). Example 490: (S)-2-(4-(6-((4-chloro-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1298 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.56 (s, 1H), 7.88 (dd, J=8.5, 1.5 Hz, 1H), 7.85-7.73 (m, 2H), 7.60 (d, J=8.5 Hz, 1H), 7.54-7.48 (m, 1H), 7.44 (dd, J=9.5, 6.3 Hz, 1H), 7.35 (dd, J=9.2, 6.1 Hz, 1H), 7.22 (dd, J=11.5, 6.1 Hz, 1H), 6.87 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 4.85 (d, J=6.7 Hz, 1H), 4.55 (dd, J=11.2, 1.5 Hz, 1H), 4.42 (dd, J=11.1, 6.9 Hz, 1H), 4.38 (d, J=6.0 Hz, 2H), 3.86 (d, J=8.7 Hz, 1H), 3.75 (d, J=8.7 Hz, 1H), 1.34 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 640.2 (M+H+). Example 491: (S)-2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2,5-difluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1294 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.59 (s, 1H), 7.90 (dd, J=8.5, 1.5 Hz, 1H), 7.74 (dd, J=10.7, 6.5 Hz, 1H), 7.66-7.49 (m, 5H), 7.25 (dd, J=11.6, 6.1 Hz, 1H), 5.68 (s, 2H), 4.87 (d, J=6.8 Hz, 1H), 4.59 (dd, J=11.2, 1.5 Hz, 1H), 4.46 (dd, J=11.1, 6.8 Hz, 1H), 4.39 (d, J=6.7 Hz, 2H), 3.89 (d, J=8.7 Hz, 1H), 3.78 (d, J=8.7 Hz, 1H), 1.37 (s, 3H), 0.68 (s, 3H). ES/MS m/z: 649.2 (M+H+). Example 492: (S)-2-(4-(6-((4-cyano-2,3-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2,3-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1300 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.70 (s, 1H), 8.02 (dd, J=8.6, 1.5 Hz, 1H), 7.83 (d, J=7.5 Hz, 1H), 7.81-7.75 (m, 1H), 7.72 (d, J=8.5 Hz, 1H), 7.57-7.51 (m, 2H), 7.51-7.44 (m, 1H), 7.29 (dd, J=11.4, 6.1 Hz, 1H), 6.92 (d, J=8.3 Hz, 1H), 5.62 (s, 2H), 4.93 (d, J=6.6 Hz, 1H), 4.62-4.54 (m, 1H), 4.51 (d, J=8.4 Hz, 2H), 4.44 (dd, J=11.5, 6.7 Hz, 1H), 3.90 (d, J=8.9 Hz, 1H), 3.77 (d, J=8.9 Hz, 1H), 1.35 (s, 3H), 0.69 (s, 3H). ES/MS m/z: 631.25 (M+H+). Example 493: (S)-2-(4-(6-((4-cyano-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1301 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.60 (s, 1H), 7.95-7.88 (m, 2H), 7.81 (t, J=7.8 Hz, 1H), 7.64 (d, J=8.5 Hz, 1H), 7.58 (dd, J=7.3, 1.6 Hz, 1H), 7.48 (d, J=6.7 Hz, 2H), 7.26 (dd, J=11.6, 6.0 Hz, 1H), 6.84 (d, J=8.2 Hz, 1H), 5.63 (s, 2H), 4.88 (d, J=6.9 Hz, 1H), 4.60 (dd, J=11.3, 1.4 Hz, 1H), 4.46 (dd, J=11.3, 6.8 Hz, 1H), 4.41 (d, J=5.9 Hz, 2H), 3.89 (d, J=8.7 Hz, 1H), 3.78 (d, J=8.6 Hz, 1H), 1.38 (s, 3H), 0.69 (s, 3H). ES/MS m/z: 631.3 (M+H+). Example 494: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4R)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-3 and I-1292. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.60 (d, J=1.5 Hz, 1H), 7.89 (dd, J=8.5, 1.6 Hz, 1H), 7.84-7.75 (m, 2H), 7.72 (t, J=7.5 Hz, 1H), 7.62 (d, J=8.5 Hz, 1H), 7.58 (d, J=8.7 Hz, 2H), 7.54 (dd, J=7.5, 1.8 Hz, 1H), 7.23 (dd, J=11.5, 6.1 Hz, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.62 (s, 2H), 5.27 (t, J=7.2 Hz, 1H), 4.57 (dd, J=10.9, 1.4 Hz, 1H), 4.41 (s, 2H), 4.26 (dd, J=11.0, 6.7 Hz, 1H), 4.16 (t, J=8.5 Hz, 1H), 3.70 (t, J=8.4 Hz, 1H), 2.91 (hept, J=7.1 Hz, 1H), 0.59 (d, J=7.1 Hz, 3H). ES/MS m/z: 599.3 (M+H+). Example 495: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1036 and I-108. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.41 (s, 1H), 7.81 (dd, J=10.7, 6.5 Hz, 1H), 7.63-7.50 (m, 4H), 7.30-7.19 (m, 3H), 5.59 (d, J=1.1 Hz, 2H), 4.85 (d, J=6.6 Hz, 1H), 4.54 (dd, J=11.3, 1.6 Hz, 1H), 4.43 (dd, J=11.3, 6.8 Hz, 1H), 4.37 (d, J=7.1 Hz, 2H), 3.84 (d, J=8.8 Hz, 1H), 3.75 (d, J=8.7 Hz, 1H), 1.35 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 658.2 (M+H+). Example 496: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-109 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.58 (s, 1H), 7.89 (dd, J=8.5, 1.5 Hz, 1H), 7.79-7.71 (m, 2H), 7.65-7.52 (m, 5H), 7.24 (dd, J=11.6, 6.1 Hz, 1H), 5.70 (s, 2H), 4.86 (d, J=6.6 Hz, 1H), 4.59 (dd, J=11.2, 1.6 Hz, 1H), 4.45 (dd, J=11.3, 7.0 Hz, 1H), 4.38 (d, J=6.4 Hz, 2H), 3.88 (d, J=8.7 Hz, 1H), 3.78 (d, J=8.7 Hz, 1H), 1.37 (s, 3H), 0.68 (s, 3H). ES/MS m/z: 631.3 (M+H+). Example 497: (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1036 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.56 (s, 1H), 7.87 (dd, J=8.5, 1.5 Hz, 1H), 7.80 (dd, J=10.8, 6.5 Hz, 1H), 7.64-7.47 (m, 4H), 7.32-7.16 (m, 3H), 5.59 (s, 2H), 4.85 (d, J=6.8 Hz, 1H), 4.56 (dd, J=11.1, 1.6 Hz, 1H), 4.48-4.42 (m, 1H), 4.42-4.30 (m, 2H), 3.86 (d, J=8.7 Hz, 1H), 3.75 (d, J=8.7 Hz, 1H), 1.34 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 640.232 (M+H+). Example 498: (S)-2-(4-(6-((4-cyano-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1301 and I-108. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.46 (s, 1H), 7.90 (dd, J=10.8, 6.4 Hz, 1H), 7.78 (t, J=7.9 Hz, 1H), 7.63 (dd, J=11.2, 1.2 Hz, 1H), 7.55 (dd, J=7.5, 1.8 Hz, 1H), 7.45 (d, J=6.7 Hz, 2H), 7.26 (dd, J=11.4, 6.1 Hz, 1H), 6.81 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 4.89 (d, J=6.7 Hz, 1H), 4.55 (dd, J=11.4, 1.5 Hz, 1H), 4.50-4.37 (m, 3H), 3.86 (d, J=8.8 Hz, 1H), 3.76 (d, J=8.8 Hz, 1H), 1.36 (s, 3H), 0.69 (s, 3H). ES/MS m/z: 649.2 (M+H+). Example 499: 2-(4-(6-((4-chloro-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2,6-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1297 and I-1336. 1H NMR (400 MHz, DMSO-d6) δ 13.08 (s, 1H), 8.35 (s, 1H), 7.92 (dd, J=10.4, 6.4 Hz, 1H), 7.87 (t, J=7.9 Hz, 1H), 7.58-7.40 (m, 5H), 6.91 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.59-4.47 (m, 2H), 4.47-4.36 (m, 2H), 3.74 (q, J=8.7 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 658.1 (M+H+). Example 500: (S)-2-(4-(6-((4-cyano-2,3-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2,3-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1300 and I-108. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.48 (s, 1H), 7.83-7.72 (m, 2H), 7.65 (dd, J=11.1, 1.2 Hz, 1H), 7.56-7.44 (m, 3H), 7.25 (dd, J=11.5, 6.1 Hz, 1H), 6.90 (d, J=8.2 Hz, 1H), 5.61 (d, J=1.3 Hz, 2H), 4.91 (d, J=6.6 Hz, 1H), 4.54 (dd, J=11.5, 1.4 Hz, 1H), 4.48-4.37 (m, 3H), 3.86 (d, J=8.8 Hz, 1H), 3.76 (d, J=8.9 Hz, 1H), 1.35 (s, 3H), 0.69 (s, 3H). ES/MS m/z: 649.2 (M+H+). Example 501: 2-(4-(6-((4-chloro-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2,5-difluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1284 and I-1336. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.49 (s, 1H), 7.82 (dd, J=10.8, 6.4 Hz, 1H), 7.76 (dd, J=8.3, 7.4 Hz, 1H), 7.67 (dd, J=11.0, 1.2 Hz, 1H), 7.50-7.39 (m, 2H), 7.33 (dd, J=9.2, 6.1 Hz, 1H), 7.25 (dd, J=11.5, 6.1 Hz, 1H), 6.87 (dd, J=8.3, 0.6 Hz, 1H), 5.47 (s, 2H), 4.93 (d, J=6.6 Hz, 1H), 4.57-4.46 (m, 3H), 4.47-4.37 (m, 1H), 3.85 (d, J=8.9 Hz, 1H), 3.76 (d, J=8.9 Hz, 1H), 1.34 (s, 3H), 0.69 (s, 3H). ES/MS m/z: 658.1 (M+H+). Example 502A: (S)-2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid Example 502B: (R)-2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid Example 502A and Example 502B were obtained from preparative chiral SFC (Daicel Chiraltek AD-H column, EtOH/CO2eluent) of Example 506. (S)-2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (Example 502A) was isolated as the later-eluting of two stereoisomers. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.49 (s, 1H), 7.89-7.81 (m, 1H), 7.79 (d, J=7.9 Hz, 1H), 7.70-7.62 (m, 2H), 7.54 (dd, J=7.4, 1.7 Hz, 1H), 7.40-7.33 (m, 2H), 7.25 (dd, J=11.4, 6.1 Hz, 1H), 6.89 (d, J=8.2 Hz, 1H), 6.78 (t, J=55.9 Hz, 1H), 5.59 (s, 2H), 4.90 (d, J=6.6 Hz, 1H), 4.55 (dd, J=11.5, 1.4 Hz, 1H), 4.48-4.38 (m, 3H), 3.86 (d, J=8.8 Hz, 1H), 3.76 (d, J=8.8 Hz, 1H), 1.35 (s, 3H), 0.69 (s, 3H). ES/MS m/z: 656.1 (M+H+). (R)-2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid (Example 502B) was isolated as the earlier-eluting of two stereoisomers. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.44 (s, 1H), 7.91-7.77 (m, 2H), 7.70 (t, J=7.7 Hz, 1H), 7.60 (dd, J=11.3, 1.2 Hz, 1H), 7.57 (dd, J=7.5, 1.8 Hz, 1H), 7.44-7.34 (m, 2H), 7.26 (dd, J=11.5, 6.1 Hz, 1H), 6.98-6.88 (m, 1H), 6.74 (d, J=55.9 Hz, 1H), 5.62 (s, 2H), 4.88 (d, J=6.5 Hz, 1H), 4.57 (dd, J=11.2, 1.5 Hz, 1H), 4.50-4.32 (m, 3H), 3.87 (d, J=8.7 Hz, 1H), 3.77 (d, J=8.8 Hz, 1H), 1.38 (s, 3H), 0.69 (s, 3H). ES/MS m/z: 656.2 (M+H+). Example 503: 2-(2,5-difluoro-4-(5-fluoro-6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(5-fluoro-6-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1302 and I-1336. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.41 (s, 1H), 7.80-7.72 (m, 2H), 7.63-7.48 (m, 5H), 7.23 (dd, J=11.5, 6.1 Hz, 1H), 5.69 (s, 2H), 4.85 (d, J=6.7 Hz, 1H), 4.55 (d, J=11.2 Hz, 1H), 4.45 (d, J=8.1 Hz, 1H), 4.42-4.29 (m, 2H), 3.84 (d, J=8.8 Hz, 1H), 3.75 (d, J=8.9 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 692.1 (M+H+). Example 504: 2-(4-(2-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(2-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyrimidin-4-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1303 and I-1336. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.65 (d, J=5.2 Hz, 1H), 8.45 (s, 1H), 7.95 (dd, J=10.4, 6.2 Hz, 1H), 7.69 (t, J=7.6 Hz, 1H), 7.62 (dd, J=11.1, 1.2 Hz, 1H), 7.58 (dd, J=5.2, 1.8 Hz, 1H), 7.42-7.35 (m, 2H), 7.30 (dd, J=11.5, 5.9 Hz, 1H), 6.79 (t, J=55.9 Hz, 1H), 5.61 (s, 2H), 4.88 (d, J=6.7 Hz, 1H), 4.55 (d, J=11.4 Hz, 1H), 4.50-4.36 (m, 3H), 3.85 (d, J=8.8 Hz, 1H), 3.76 (d, J=8.8 Hz, 1H), 1.35 (s, 3H), 0.68 (s, 3H). ES/MS m/z: 657.1 (M+H+). Example 505: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 36, starting with Intermediates I-7 and I-1289. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.94-7.87 (m, 2H), 7.81-7.71 (m, 3H), 7.61 (d, J=8.4 Hz, 1H), 7.54 (d, J=7.2 Hz, 1H), 7.44 (dd, J=11.4, 6.3 Hz, 2H), 6.99 (d, J=8.2 Hz, 1H), 5.60 (s, 2H), 5.42 (t, J=7.1 Hz, 1H), 4.56-4.39 (m, 3H), 4.26-4.17 (m, 1H), 4.12 (t, J=8.5 Hz, 1H), 3.59 (t, J=8.5 Hz, 1H), 2.92-2.82 (m, 1H), 0.52 (d, J=7.0 Hz, 3H). ES/MS m/z: 599.2 (M+H+). Example 506: 2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-(difluoromethyl)-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-4-fluoro-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 36, starting with Intermediates I-1305 and I-1336. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.41 (s, 1H), 7.88-7.77 (m, 2H), 7.67 (t, J=7.7 Hz, 1H), 7.62 (d, J=11.0 Hz, 1H), 7.58-7.51 (m, 1H), 7.41-7.32 (m, 2H), 7.23 (dd, J=11.5, 6.1 Hz, 1H), 6.94-6.61 (m, 3H), 5.60 (s, 2H), 4.87 (d, J=6.7 Hz, 1H), 4.57 (d, J=11.4 Hz, 1H), 4.52-4.45 (m, 1H), 4.45-4.32 (m, 2H), 3.85 (d, J=8.8 Hz, 1H), 3.83-3.74 (m, 1H), 1.35 (s, 3H), 0.67 (s, 3H). ES/MS m/z: 656.1 (M+H+). Example 507: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(3-methoxy-3-methylbutan-2-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(3-methoxy-3-methylbutan-2-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1307. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.58 (dd, J=1.6, 0.6 Hz, 1H), 7.87-7.77 (m, 2H), 7.77-7.72 (m, 1H), 7.59-7.47 (m, 3H), 7.28-7.20 (m, 2H), 7.16 (dd, J=11.5, 6.1 Hz, 1H), 6.84 (dd, J=8.3, 0.7 Hz, 1H), 5.51 (d, J=1.0 Hz, 2H), 4.55 (q, J=7.1 Hz, 1H), 4.38 (d, J=2.4 Hz, 2H), 3.18 (s, 2H), 1.64 (d, J=7.1 Hz, 3H), 1.34 (s, 3H), 1.02 (s, 3H). ES/MS m/z: 624.1 (M+H+). Example 508: (S)-2-((3′-((4-chloro-2-fluorobenzyl)oxy)-2,5-difluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((3′-((4-chloro-2-fluorobenzyl)oxy)-2,5-difluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1308 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.55 (s, 1H), 7.87 (dd, J=8.5, 1.5 Hz, 1H), 7.60 (d, J=8.5 Hz, 1H), 7.53 (t, J=8.2 Hz, 1H), 7.39 (t, J=7.9 Hz, 1H), 7.32 (dd, J=10.2, 6.4 Hz, 1H), 7.31-7.13 (m, 5H), 7.04 (ddd, J=8.4, 2.6, 1.0 Hz, 1H), 5.16 (s, 2H), 4.84 (dd, J=6.9, 1.6 Hz, 1H), 4.55 (dd, J=11.2, 1.6 Hz, 1H), 4.43 (dd, J=11.2, 6.9 Hz, 1H), 4.35 (d, J=6.5 Hz, 2H), 3.85 (d, J=8.7 Hz, 1H), 3.75 (d, J=8.7 Hz, 1H), 1.34 (s, 3H), 0.65 (s, 3H). ES/MS m/z: 621.2 (M+H+). Example 509: (S)-2-((3′-((4-chloro-2-fluorobenzyl)oxy)-2,4′,5-trifluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-((3′-((4-chloro-2-fluorobenzyl)oxy)-2,4′,5-trifluoro-[1,1′-biphenyl]-4-yl)methyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1309 and I-82. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.55 (s, 1H), 7.87 (dd, J=8.5, 1.5 Hz, 1H), 7.60 (d, J=8.5 Hz, 1H), 7.53 (t, J=8.2 Hz, 1H), 7.39-7.10 (m, 7H), 5.19 (s, 2H), 4.84 (dd, J=6.9, 1.6 Hz, 1H), 4.55 (dd, J=11.2, 1.6 Hz, 1H), 4.42 (dd, J=11.1, 6.8 Hz, 1H), 4.35 (d, J=6.6 Hz, 2H), 3.85 (d, J=8.7 Hz, 1H), 3.74 (d, J=8.7 Hz, 1H), 1.34 (s, 3H), 0.64 (s, 3H). ES/MS m/z: 639.2 (M+H+). Example 510: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxy-2-methylpropyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxy-2-methylpropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1311. 1H NMR (400 MHz, Acetonitrile-d3) δ 8.24 (s, 1H), 7.91-7.73 (m, 3H), 7.61-7.48 (m, 3H), 7.29-7.13 (m, 3H), 6.84 (d, J=8.2 Hz, 1H), 5.52 (s, 2H), 4.44 (s, 2H), 4.30 (s, 2H), 3.14 (s, 3H), 1.22 (s, 6H). ES/MS m/z: 610.2 (M+H+). Example 511: 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-cyanobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 6, starting with Intermediates I-17 and 4-(bromomethyl)benzonitrile. 1H NMR (400 MHz, Methanol-d4) δ 8.25 (s, 1H), 7.97 (dd, J=8.5, 1.5 Hz, 1H), 7.78 (t, J=7.9 Hz, 1H), 7.75-7.60 (m, 6H), 7.51 (d, J=7.4 Hz, 1H), 7.12 (dd, J=11.6, 6.0 Hz, 1H), 6.90 (d, J=8.3 Hz, 1H), 5.57 (s, 2H), 4.55 (t, J=5.1 Hz, 2H), 4.47 (s, 2H), 3.72 (t, J=5.0 Hz, 2H), 3.25 (s, 3H). ES/MS m/z: 555.3 (M+H+). Example 512: (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-benzo[d][1,2,3]triazol-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-benzo[d][1,2,3]triazol-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1035 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 8.15 (s, 1H), 7.96-7.83 (m, 3H), 7.82 (dd, J=8.5, 1.5 Hz, 1H), 7.71 (dd, J=8.6, 1.4 Hz, 1H), 7.63 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.5 Hz, 1H), 7.47 (dd, J=11.1, 6.4 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.65 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.63-4.54 (m, 1H), 4.53 (s, 1H), 4.49-4.35 (m, 2H), 4.31 (s, 3H), 3.81-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 624.6 (M+H+). Example 513: (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-benzo[d][1,2,3]triazol-6-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-benzo[d][1,2,3]triazol-6-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1034 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.04 (d, J=8.6 Hz, 1H), 8.01 (s, 1H), 7.90 (t, J=7.9 Hz, 1H), 7.87-7.78 (m, 2H), 7.62 (d, J=8.5 Hz, 1H), 7.54 (ddd, J=7.6, 5.1, 1.4 Hz, 2H), 7.46 (dd, J=11.4, 6.2 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.67 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.64-4.48 (m, 2H), 4.48-4.32 (m, 2H), 4.30 (s, 3H), 3.82-3.72 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 624.6 (M+H+). Example 514: 2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-4-fluoro-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1022 and I-1033. 1H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J=1.3 Hz, 1H), 7.91-7.78 (m, 2H), 7.54-7.42 (m, 3H), 7.39 (dd, J=11.6, 6.1 Hz, 1H), 7.35 (d, J=7.8 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.48 (s, 2H), 4.71-4.53 (m, 6H), 4.46 (s, 2H), 3.68 (t, J=5.0 Hz, 2H), 3.21 (s, 3H), 2.96 (s, 3H). ES/MS m/z: 666.6 (M+H+). Example 515: (S)-2-(2,5-difluoro-4-(5-fluoro-6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(5-fluoro-6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 40, starting with Intermediates I-1029 and I-5. 1H NMR (400 MHz, Methanol-d4) δ 8.15 (d, J=1.3 Hz, 1H), 7.75 (dd, J=10.8, 6.4 Hz, 1H), 7.66 (d, J=11.3 Hz, 1H), 7.59 (dd, J=9.9, 8.2 Hz, 1H), 7.55-7.42 (m, 3H), 7.39-7.30 (m, 1H), 7.18 (dd, J=11.5, 6.0 Hz, 1H), 5.58 (s, 2H), 5.18 (d, J=7.5 Hz, 1H), 4.78-4.65 (m, 6H), 4.65-4.31 (m, 4H), 3.78 (d, J=1.7 Hz, 3H), 2.90-2.60 (m, 1H), 2.56-2.36 (m, 1H). ES/MS m/z: 676.6 (M+H+). Example 516: (S)-2-(2,5-difluoro-4-(5-fluoro-6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(5-fluoro-6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-1029 and I-4. 1H NMR (400 MHz, Methanol-d4) δ 8.27 (s, 1H), 8.02-7.92 (m, 1H), 7.75 (dd, J=10.7, 6.2 Hz, 1H), 7.67-7.54 (m, 2H), 7.54-7.43 (m, 3H), 7.33 (d, J=8.1 Hz, 0H), 7.17 (dd, J=11.5, 6.0 Hz, 1H), 5.59 (s, 2H), 5.20 (d, J=10.9 Hz, 1H), 4.73 (s, 5H), 4.68-4.50 (m, 3H), 4.50-4.36 (m, 1H), 3.78 (d, J=1.9 Hz, 3H), 2.78 (s, 1H), 2.51 (s, 1H). ES/MS m/z: 658.6 (M+H+). Example 517: (S)-2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-4-fluoro-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 40, starting with Intermediates I-1027 and I-5. 1H NMR (400 MHz, Methanol-d4) δ 8.15 (s, 1H), 7.84-7.71 (m, 2H), 7.67 (d, J=11.3 Hz, 1H), 7.54-7.40 (m, 3H), 7.33 (d, J=7.9 Hz, 1H), 7.18 (dd, J=11.5, 6.1 Hz, 1H), 6.87 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 5.17 (d, J=8.1 Hz, 1H), 4.70 (d, J=4.3 Hz, 5H), 4.66-4.48 (m, 4H), 4.45 (dt, J=9.0, 5.9 Hz, 1H), 2.91 (s, 3H), 2.79 (s, 1H), 2.55-2.41 (m, 1H). ES/MS m/z: 678.6 (M+H+). Example 518: (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-4-fluoro-1-(2-methoxypropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 40, starting with Intermediates I-7 and I-1270. 1H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J=1.3 Hz, 1H), 7.97-7.85 (m, 2H), 7.75 (ddd, J=9.7, 7.8, 6.5 Hz, 3H), 7.58-7.44 (m, 2H), 7.39 (dd, J=11.5, 6.1 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 5.60 (s, 2H), 4.53 (dd, J=15.2, 3.2 Hz, 1H), 4.46 (s, 2H), 4.36 (dd, J=15.2, 8.8 Hz, 1H), 3.76-3.61 (m, 1H), 3.08 (s, 3H), 1.23 (d, J=6.2 Hz, 3H). ES/MS m/z: 605.5 (M+H+). Example 519: (S)-2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-(methylsulfonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1022 and I-8. 1H NMR (400 MHz, Methanol-d4) δ 8.30 (s, 1H), 7.99 (dd, J=8.5, 1.5 Hz, 1H), 7.86-7.73 (m, 2H), 7.66 (d, J=8.5 Hz, 1H), 7.56-7.49 (m, 1H), 7.46 (d, J=10.9 Hz, 2H), 7.33 (d, J=7.8 Hz, 1H), 7.18 (dd, J=11.6, 6.0 Hz, 1H), 6.87 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 5.21 (d, J=8.8 Hz, 1H), 4.79-4.65 (m, 5H), 4.65-4.35 (m, 4H), 2.91 (s, 3H), 2.87-2.69 (m, 1H), 2.51 (d, J=11.2 Hz, 0H). ES/MS m/z: 661.2 (M+H+). Example 520: (S)-2-(4-(6-((2-(cyclopropanecarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2-(cyclopropanecarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1021 and I-8. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J=1.4 Hz, 1H), 7.99 (dd, J=8.5, 1.5 Hz, 1H), 7.85-7.72 (m, 2H), 7.67 (d, J=8.5 Hz, 1H), 7.56-7.43 (m, 3H), 7.36 (dd, J=7.9, 3.8 Hz, 1H), 7.24-7.09 (m, 1H), 6.87 (d, J=8.2 Hz, 1H), 5.52 (d, J=2.5 Hz, 2H), 5.19 (td, J=7.2, 2.5 Hz, 1H), 5.08 (d, J=4.3 Hz, 2H), 4.77 (d, J=6.9 Hz, 2H), 4.73-4.37 (m, 6H), 2.88-2.67 (m, 1H), 2.50 (dq, J=11.4, 7.7 Hz, 1H), 1.97-1.84 (m, 1H), 0.97 (dq, J=6.0, 3.1 Hz, 2H), 0.91 (ddt, J=8.8, 6.0, 2.8 Hz, 2H). ES/MS m/z: 651.2 (M+H+). Example 521: (S)-2-(2,5-difluoro-4-(6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1016 and I-8. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (s, 1H), 7.99 (dd, J=8.5, 1.5 Hz, 1H), 7.86-7.70 (m, 2H), 7.67 (d, J=8.5 Hz, 1H), 7.53-7.41 (m, 3H), 7.36-7.25 (m, 1H), 7.17 (dd, J=11.4, 6.0 Hz, 1H), 6.87 (d, J=8.3 Hz, 1H), 5.50 (s, 2H), 5.20 (d, J=7.2 Hz, 1H), 4.72 (q, J=3.8, 3.2 Hz, 6H), 4.66-4.34 (m, 4H), 3.78 (d, J=1.9 Hz, 3H), 2.91-2.74 (m, 1H), 2.50 (dt, J=17.5, 7.8 Hz, 1H). ES/MS m/z: 641.2 (M+H+). Example 522: (S)-2-(4-(6-((2-acetylisoindolin-5-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((2-acetylisoindolin-5-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1015 and I-8. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (s, 1H), 7.99 (dd, J=8.5, 1.5 Hz, 1H), 7.79 (q, J=8.7, 7.8 Hz, 2H), 7.67 (d, J=8.5 Hz, 1H), 7.58-7.42 (m, 3H), 7.42-7.27 (m, 1H), 7.26-7.13 (m, 1H), 6.87 (d, J=8.3 Hz, 1H), 5.52 (s, 2H), 5.30-5.12 (m, 1H), 4.73 (dd, J=16.6, 7.0 Hz, 3H), 4.66-4.37 (m, 5H), 2.91-2.65 (m, 1H), 2.60-2.39 (m, 1H), 2.18 (d, J=5.9 Hz, 3H). ES/MS m/z: 625.2 (M+H+). Example 523: (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-benzo[d][1,2,3]triazol-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-benzo[d][1,2,3]triazol-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-9 and 5-(bromomethyl)-1-methyl-benzotriazole. 1H NMR (400 MHz, Methanol-d4) δ 8.32 (s, 1H), 8.10 (s, 1H), 8.00 (dd, J=8.5, 1.5 Hz, 1H), 7.89-7.71 (m, 4H), 7.66 (s, 1H), 7.53 (d, J=7.4 Hz, 1H), 7.18 (dd, J=11.4, 6.0 Hz, 1H), 6.92 (d, J=8.3 Hz, 1H), 5.68 (s, 2H), 5.20 (q, J=6.8 Hz, 1H), 4.74 (dd, J=15.7, 6.9 Hz, 1H), 4.68-4.47 (m, 4H), 4.45 (dt, J=9.3, 5.9 Hz, 1H), 4.35 (d, J=1.0 Hz, 3H), 2.80 (dt, J=15.9, 7.7 Hz, 1H), 2.58-2.38 (m, 1H). ES/MS m/z: 597.2 (M+H+). Example 524: (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-indazol-6-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-indazol-6-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-9 and 6-(bromomethyl)-1-methyl-indazole. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J=1.5 Hz, 1H), 8.02-7.93 (m, 2H), 7.87-7.72 (m, 4H), 7.71-7.61 (m, 2H), 7.52 (dd, J=7.5, 1.6 Hz, 1H), 7.32 (dd, J=8.4, 1.2 Hz, 1H), 7.17 (dd, J=11.5, 6.0 Hz, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.64 (s, 2H), 5.19 (qd, J=7.1, 2.5 Hz, 1H), 4.72 (dd, J=15.7, 6.9 Hz, 1H), 4.67-4.48 (m, 5H), 4.48-4.34 (m, 1H), 4.06 (s, 3H), 2.91-2.65 (m, 1H), 2.58-2.40 (m, 1H). ES/MS m/z: 596.2 (M+H+). Example 525: (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-benzo[d][1,2,3]triazol-6-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-11H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-benzo[d][1,2,3]triazol-6-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediate I-9 and I-1014. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J=1.4 Hz, 1H), 8.00 (d, J=2.1 Hz, 1H), 7.98 (d, J=2.2 Hz, 1H), 7.92 (s, 1H), 7.86-7.73 (m, 2H), 7.66 (d, J=8.5 Hz, 1H), 7.60 (dd, J=8.7, 1.3 Hz, 1H), 7.53 (dd, J=7.5, 1.5 Hz, 1H), 7.18 (dd, J=11.4, 6.0 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.70 (s, 2H), 5.19 (dd, J=7.2, 2.5 Hz, 1H), 4.72 (dd, J=15.7, 6.9 Hz, 1H), 4.67-4.49 (m, 4H), 4.48-4.36 (m, 1H), 4.33 (s, 3H), 2.79 (ddt, J=14.2, 11.6, 6.8 Hz, 1H), 2.56-2.41 (m, 1H). ES/MS m/z: 597.2 (M+H+). Example 526: (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-indazol-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((1-methyl-1H-indazol-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-9 and 5-(bromomethyl)-1-methyl-indazole. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J=1.4 Hz, 1H), 7.99 (d, J=9.1 Hz, 2H), 7.91-7.81 (m, 2H), 7.76 (q, J=6.7, 5.5 Hz, 1H), 7.67 (d, J=8.5 Hz, 1H), 7.63-7.53 (m, 2H), 7.51 (dd, J=7.5, 1.6 Hz, 1H), 7.18 (dd, J=11.4, 6.0 Hz, 1H), 6.86 (d, J=8.2 Hz, 1H), 5.59 (s, 2H), 5.20 (qd, J=7.2, 2.5 Hz, 1H), 4.81-4.67 (m, 1H), 4.67-4.49 (m, 4H), 4.49-4.35 (m, 1H), 4.07 (d, J=0.9 Hz, 3H), 2.89-2.71 (m, 1H), 2.64-2.38 (m, 1H). ES/MS m/z: 596.2 (M+H+). Example 527: (S)-2-(4-(6-(benzo[c][1,2,5]thiadiazol-5-ylmethoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-(benzo[c][1,2,5]thiadiazol-5-ylmethoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-9 and 5-(bromomethyl)-2,1,3-benzothiadiazole. 1H NMR (400 MHz, Methanol-d4) δ 8.29 (s, 1H), 8.11 (s, 1H), 8.04 (d, J=9.0 Hz, 1H), 7.98 (dd, J=8.5, 1.5 Hz, 1H), 7.86-7.72 (m, 3H), 7.65 (d, J=8.5 Hz, 1H), 7.55 (d, J=7.5 Hz, 1H), 7.16 (dd, J=11.5, 6.1 Hz, 1H), 6.97 (d, J=8.2 Hz, 1H), 5.71 (s, 2H), 5.19 (d, J=7.3 Hz, 1H), 4.72 (dd, J=15.7, 6.9 Hz, 1H), 4.67-4.34 (m, 5H), 2.79 (t, J=9.1 Hz, 1H), 2.59-2.41 (m, 1H). ES/MS m/z: 600.2 (M+H+). Example 528: (S)-4-chloro-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-4-chloro-2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-112 and I-1037. 1H NMR (400 MHz, DMSO) δ 8.89 (s, 1H), 8.82 (s, 1H), 8.47 (s, 1H), 8.30 (s, 1H), 8.03 (s, 1H), 7.98-7.85 (m, 2H), 7.81 (s, 1H), 7.57 (d, J=7.3 Hz, 1H), 7.46 (dd, J=11.4, 6.1 Hz, 1H), 7.01 (d, J=8.2 Hz, 1H), 5.68 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.67-4.29 (m, 4H), 3.84-3.68 (m, 2H), 1.31 (s, 3H), 0.59 (s, 3H). ES/MS m/z: 706.0 (M+H+). Example 529: (S)-2-(2,5-difluoro-4-(6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-(methoxycarbonyl)isoindolin-5-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1016 and I-82. 1H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.91-7.77 (m, 3H), 7.55-7.39 (m, 5H), 7.34 (dd, J=10.6, 7.8 Hz, 1H), 6.94 (d, J=8.3 Hz, 1H), 5.47 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.64 (t, J=6.9 Hz, 4H), 4.58-4.50 (m, 2H), 4.44 (dd, J=11.2, 6.8 Hz, 1H), 4.39 (d, J=16.9 Hz, 1H), 3.81-3.71 (m, 2H), 3.67 (s, 3H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 669.6 (M+H+). Example 530: (S)-2-(4-(6-(benzo[d]thiazol-6-ylmethoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-(benzo[d]thiazol-6-ylmethoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-9 and I-1346. 1H NMR (400 MHz, Methanol-d4) δ 9.21 (s, 1H), 8.52 (d, J=1.4 Hz, 1H), 8.20 (dd, J=8.6, 1.4 Hz, 2H), 8.08 (d, J=8.4 Hz, 1H), 7.87 (dd, J=10.8, 6.3 Hz, 1H), 7.83-7.73 (m, 2H), 7.69 (dd, J=8.5, 1.6 Hz, 1H), 7.55 (dd, J=7.5, 1.6 Hz, 1H), 7.31 (dd, J=11.1, 6.0 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.66 (s, 2H), 5.24 (tt, J=7.3, 3.8 Hz, 1H), 4.93 (dd, J=15.5, 7.5 Hz, 1H), 4.83-4.63 (m, 4H), 4.54 (dt, J=9.1, 6.0 Hz, 1H), 2.98-2.75 (m, 1H), 2.67-2.50 (m, 1H). ES/MS m/z: 599.1 (M+H+). Example 531: (S)-2-(2,5-difluoro-4-(6-((3-methyl-2-oxo-2,3-dihydrobenzo[d]thiazol-6-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((3-methyl-2-oxo-2,3-dihydrobenzo[d]thiazol-6-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 27, starting with Intermediates I-9 and 5-(bromomethyl)-3-methyl-1,3-benzothiazol-2-one. 1H NMR (400 MHz, Methanol-d4) δ 8.57 (d, J=1.3 Hz, 1H), 8.21 (dd, J=8.6, 1.4 Hz, 1H), 7.90 (dd, J=10.8, 6.3 Hz, 1H), 7.85-7.74 (m, 2H), 7.69 (d, J=1.6 Hz, 1H), 7.54 (ddd, J=8.3, 6.4, 1.7 Hz, 2H), 7.37 (dd, J=11.2, 6.1 Hz, 1H), 7.26 (d, J=8.3 Hz, 1H), 6.91 (d, J=8.2 Hz, 1H), 5.52 (s, 2H), 5.26 (qd, J=7.4, 2.4 Hz, 1H), 4.98 (dd, J=15.5, 7.5 Hz, 1H), 4.86-4.64 (m, 4H), 4.54 (dt, J=9.1, 6.0 Hz, 1H), 3.47 (s, 3H), 2.88 (dtd, J=11.5, 8.2, 6.1 Hz, 1H), 2.58 (ddt, J=11.5, 9.1, 7.2 Hz, 1H). ES/MS m/z: 629.2 (M+H+). Example 532: (S)-methyl 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (S)-methyl 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared and described previously as Intermediate I-103. 1H NMR (400 MHz, Chloroform-d) δ 8.55 (s, 1H), 8.01 (dd, J=8.5, 1.5 Hz, 1H), 7.88 (dd, J=10.8, 6.3 Hz, 1H), 7.79 (d, J=8.5 Hz, 1H), 7.66 (t, J=7.9 Hz, 1H), 7.54-7.41 (m, 3H), 7.14 (dt, J=9.8, 2.3 Hz, 2H), 7.08 (dd, J=11.3, 6.0 Hz, 1H), 6.79 (d, J=8.2 Hz, 1H), 5.49 (s, 2H), 4.65 (d, J=7.0 Hz, 1H), 4.57 (dd, J=11.0, 1.8 Hz, 1H), 4.47-4.30 (m, 3H), 3.96-3.92 (m, 4H), 3.79 (d, J=8.8 Hz, 1H), 1.34 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 636.3 (M+H+). Example 533: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 39, starting with Intermediates I-1255 and I-1351. 1H NMR (400 MHz, DMSO) δ 8.43 (d, J=1.5 Hz, 1H), 7.91-7.75 (m, 3H), 7.67-7.57 (m, 2H), 7.53 (ddd, J=10.1, 7.3, 2.5 Hz, 2H), 7.43-7.28 (m, 2H), 5.60 (s, 2H), 4.54 (s, 2H), 4.42 (dd, J=10.4, 3.6 Hz, 1H), 4.19-4.07 (m, 2H), 4.02 (dd, J=10.5, 8.2 Hz, 1H), 3.80 (dd, J=10.5, 4.7 Hz, 1H), 2.88 (s, 3H). ES/MS m/z: 624.1 (M+H+). Example 534: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-methoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 39, starting with Intermediates I-1256 and I-1351. 1H NMR (400 MHz, DMSO) δ 1H NMR (400 MHz, DMSO) δ 8.45 (d, J=1.6 Hz, 1H), 7.94-7.77 (m, 3H), 7.68-7.55 (m, 2H), 7.55-7.46 (m, 2H), 7.39-7.27 (m, 2H), 6.96 (d, J=8.2 Hz, 1H), 5.65-5.54 (m, 1H), 5.50 (s, 2H), 4.57 (s, 2H), 4.43 (dd, J=10.4, 3.6 Hz, 1H), 4.13 (dt, J=8.5, 2.1 Hz, 2H), 4.02 (dd, J=10.6, 8.2 Hz, 1H), 3.79 (dd, J=10.8, 4.9 Hz, 1H), 2.88 (s, 3H). ES/MS m/z: 642.2 (M+H+). Example 535: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-pyrazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-pyrazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediate I-1220 and (1-methylpyrazol-4-yl)boronic acid. 1H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.20 (dd, J=8.6, 1.4 Hz, 1H), 8.10-7.92 (m, 2H), 7.91-7.73 (m, 3H), 7.58 (dd, J=7.4, 1.6 Hz, 1H), 7.51 (t, J=7.8 Hz, 1H), 7.46-7.28 (m, 3H), 6.91 (d, J=8.2 Hz, 1H), 5.54 (s, 2H), 5.15 (d, J=6.4 Hz, 1H), 4.80-4.61 (m, 3H), 4.52 (dd, J=11.7, 6.7 Hz, 1H), 4.00 (d, J=8.9 Hz, 1H), 3.93 (s, 3H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H). ES/MS m/z: 668.2 (M+H+). Example 536: (S)-2-(2,5-difluoro-4-(5-fluoro-6-((4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(5-fluoro-6-((4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1383 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 7.87 (dd, J=10.3, 8.2 Hz, 1H), 7.84-7.69 (m, 6H), 7.62 (d, J=8.4 Hz, 1H), 7.55 (ddd, J=8.2, 2.9, 1.4 Hz, 1H), 7.46 (dd, J=11.5, 6.1 Hz, 1H), 5.68 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.55-4.48 (m, 2H), 4.47-4.32 (m, 2H), 3.82-3.69 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 656.0 (M+H+). Example 537: (S)-2-(2,5-difluoro-4-(2-((4-(trifluoromethyl)benzyl)oxy)pyrimidin-4-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(2-((4-(trifluoromethyl)benzyl)oxy)pyrimidin-4-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1384 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.79 (d, J=5.2 Hz, 1H), 8.48 (s, 1H), 7.91 (dd, J=10.2, 6.3 Hz, 1H), 7.83-7.75 (m, 3H), 7.73 (d, J=8.2 Hz, 2H), 7.64 (dd, J=5.2, 1.9 Hz, 1H), 7.61 (d, J=8.5 Hz, 1H), 7.55 (dd, J=11.5, 5.9 Hz, 1H), 5.63 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.61-4.49 (m, 2H), 4.49-4.36 (m, 2H), 3.82-3.67 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 639.0 (M+H+). Example 538: 2-(2,5-difluoro-4-(5-fluoro-6-((4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(5-fluoro-6-((4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35 starting with Intermediates I-1383 and I-1267. 1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.87 (dd, J=10.3, 8.2 Hz, 1H), 7.80 (dd, J=9.7, 8.3 Hz, 3H), 7.76-7.68 (m, 3H), 7.61 (d, J=8.5 Hz, 1H), 7.55 (ddd, J=8.2, 2.9, 1.5 Hz, 1H), 7.39 (dd, J=11.3, 6.2 Hz, 1H), 5.68 (s, 2H), 5.52 (t, J=7.3 Hz, 1H), 4.57-4.41 (m, 3H), 4.20 (dd, J=10.9, 6.5 Hz, 1H), 4.08 (t, J=8.7 Hz, 1H), 3.77 (t, J=8.2 Hz, 1H), 3.19-3.01 (m, 2H), 2.91 (s, 3H), 2.60 (t, J=8.8 Hz, 1H). ES/MS m/z: 672.0 (M+H+). Example 539: racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Racemic 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 3, starting with Intermediates I-7 and I-1386. 1H NMR (400 MHz, DMSO-d6) δ 8.43 (s, 1H), 8.03-7.86 (m, 2H), 7.85-7.67 (m, 4H), 7.60 (d, J=8.5 Hz, 1H), 7.53 (d, J=7.3 Hz, 1H), 7.34 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.3 Hz, 1H), 5.87-5.45 (m, 3H), 4.54 (s, 2H), 4.51-4.41 (m, 2H), 4.14 (s, OH), 4.05 (dd, J=10.5, 8.3 Hz, 1H), 3.93-3.83 (m, 2H), 3.62-3.43 (m, 1H), 3.24-2.99 (m, 1H). ES/MS m/z: 665.0 (M+H+). Example 540: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Example 541: 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 540 and 541: -(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 540) and 2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(2,2-difluoroethoxy)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 541) were prepared via preparative chiral SFC (Chiralpak OJ-H column, MeOH/CO2eluent) of Example 539. Example 542: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1267. 1H NMR (400 MHz, MeOD) δ 8.71 (s, 1H), 7.95 (dd, J=8.5, 1.5 Hz, 1H), 7.82 (dd, J=10.8, 6.4 Hz, 1H), 7.73 (t, J=7.9 Hz, 1H), 7.62 (d, J=8.5 Hz, 1H), 7.49 (t, J=8.0 Hz, 2H), 7.28-7.04 (m, 3H), 6.82 (d, J=8.2 Hz, 1H), 5.46 (d, J=14.0 Hz, 3H), 4.60-4.41 (m, 3H), 4.23 (dd, J=10.9, 6.6 Hz, 1H), 4.15-4.06 (m, 1H), 3.89 (dd, J=9.0, 7.0 Hz, 1H), 3.35 (s, OH), 3.21-3.04 (m, 2H), 2.92 (s, 3H), 2.57 (t, J=8.9 Hz, 1H). ES/MS m/z: 638.1 (M+H+). Example 543: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1036 and I-1267. 1H NMR (400 MHz, MeOD) δ 8.72 (s, 1H), 7.94 (dd, J=8.5, 1.5 Hz, 1H), 7.76 (dd, J=10.7, 6.4 Hz, 1H), 7.70-7.44 (m, 4H), 7.26 (dd, J=8.6, 2.6 Hz, 1H), 7.23-7.03 (m, 2H), 5.59 (s, 2H), 5.45 (t, J=7.2 Hz, 1H), 4.65-4.46 (m, 3H), 4.24 (dd, J=10.9, 6.6 Hz, 1H), 4.11 (t, J=8.9 Hz, 1H), 3.89 (dd, J=9.0, 7.0 Hz, 1H), 3.22-3.06 (m, 2H), 2.92 (s, 3H). ES/MS m/z: 656.1 (M+H+). Example 544: 2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chlorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1248 and I-1267. 1H NMR (400 MHz, MeOD) δ 8.72 (s, 1H), 7.95 (dd, J=8.4, 1.5 Hz, 1H), 7.80 (dd, J=10.8, 6.3 Hz, 1H), 7.74 (t, J=7.8 Hz, 1H), 7.64 (d, J=8.5 Hz, 1H), 7.49 (dd, J=7.5, 1.7 Hz, 1H), 7.45 (d, J=8.5 Hz, 2H), 7.36-7.33 (m, 2H), 7.14 (dd, J=11.5, 6.0 Hz, 1H), 6.84 (d, J=8.2 Hz, 1H), 5.44 (s, 3H), 4.61-4.38 (m, 3H), 4.24 (dd, J=10.9, 6.6 Hz, 1H), 4.19-4.04 (m, 1H), 3.90 (dd, J=9.0, 7.0 Hz, 1H), 2.93 (s, 3H). ES/MS m/z: 620.2 (M+H+). Example 545: 2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chlorobenzyl)oxy)-5-fluoropyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1246 and I-1267. 1H NMR (400 MHz, MeOD) δ 8.73 (s, 1H), 7.95 (dd, J=8.5, 1.5 Hz, 1H), 7.81-7.75 (m, 1H), 7.64 (d, J=8.5 Hz, 1H), 7.58 (dd, J=9.8, 8.2 Hz, 1H), 7.54-7.46 (m, 3H), 7.42-7.33 (m, 2H), 7.15 (dd, J=11.5, 6.1 Hz, 1H), 5.59-5.36 (m, 3H), 4.63-4.41 (m, 3H), 4.26 (dd, J=10.9, 6.6 Hz, 1H), 4.13 (t, J=8.9 Hz, 1H), 3.91 (dd, J=9.0, 7.0 Hz, 1H), 2.93 (s, 3H). ES/MS m/z: 638.1 (M+H+). Example 546: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid, isomer 1 (relative stereochemistry shown) 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid, isomer 1 (relative stereochemistry shown) was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1377. 1H NMR (400 MHz, MeOD) δ 8.73 (s, 1H), 7.95 (dd, J=8.5, 1.5 Hz, 1H), 7.83 (dd, J=10.7, 6.3 Hz, 1H), 7.74 (t, J=7.9 Hz, 1H), 7.64 (d, J=8.5 Hz, 1H), 7.55-7.47 (m, 2H), 7.24-7.16 (m, 2H), 7.12 (dd, J=11.5, 6.0 Hz, 1H), 6.83 (d, J=8.2 Hz, 1H), 5.49 (s, 2H), 4.96 (d, J=6.5 Hz, 1H), 4.61-4.46 (m, 2H), 4.46-4.34 (m, 2H), 3.98 (d, J=9.1 Hz, 1H), 3.73 (d, J=9.0 Hz, 1H), 3.03 (d, J=9.4 Hz, 1H), 2.88 (s, 3H), 2.69 (d, J=9.4 Hz, 1H), 1.45 (s, 3H). ES/MS m/z: 652.2 (M+H+). Example 547: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid, isomer 2 (relative stereochemistry shown) 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3S,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid, isomer 2 (relative stereochemistry shown) was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1378. 1H NMR (400 MHz, MeOD) δ 8.74 (s, 1H), 7.96 (dd, J=8.5, 1.5 Hz, 1H), 7.84 (dd, J=10.8, 6.4 Hz, 1H), 7.77 (t, J=7.9 Hz, 1H), 7.65 (d, J=8.5 Hz, 1H), 7.53 (t, J=8.0 Hz, 2H), 7.30-7.17 (m, 2H), 7.13 (dd, J=11.5, 6.0 Hz, 1H), 6.85 (d, J=8.2 Hz, 1H), 5.51 (s, 2H), 4.97 (d, J=6.5 Hz, 1H), 4.58 (d, J=17.2 Hz, 2H), 4.51-4.34 (m, 2H), 3.99 (d, J=9.0 Hz, 1H), 3.74 (d, J=9.1 Hz, 1H), 3.05 (d, J=9.4 Hz, 1H), 2.89 (s, 3H), 2.70 (d, J=9.2 Hz, 1H), 1.46 (s, 3H). ES/MS m/z: 652.2 (M+H+). Example 548 (prophetic example): 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 1, relative stereochemistry shown) 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 1, relative stereochemistry shown) was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1379. Example 549: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 2, relative stereochemistry shown) 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4S)-4-(methoxymethyl)-4-methyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Isomer 2, relative stereochemistry shown) was prepared in a manner as described in Procedure 35, starting with Intermediates I-102 and I-1380. 1H NMR (400 MHz, MeOD) δ 8.75 (s, 1H), 7.99 (dd, J=8.5, 1.5 Hz, 1H), 7.86 (dd, J=10.7, 6.4 Hz, 1H), 7.79 (t, J=7.9 Hz, 1H), 7.68 (d, J=8.5 Hz, 1H), 7.61-7.49 (m, 2H), 7.25 (ddd, J=12.2, 8.9, 2.1 Hz, 2H), 7.10 (dd, J=11.5, 6.0 Hz, 1H), 6.88 (d, J=8.2 Hz, 1H), 5.54 (s, 2H), 5.23 (d, J=7.0 Hz, 1H), 4.69-4.38 (m, 3H), 4.33 (dd, J=11.0, 7.1 Hz, 1H), 3.97 (d, J=9.2 Hz, 1H), 3.85 (d, J=9.2 Hz, 1H), 3.50 (s, 3H), 0.72 (s, 3H). ES/MS m/z: 652.2 (M+H+). Example 550: (S)-2-(2,5-difluoro-4-(6-((6-(1-methyl-11H-1,2,3-triazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((6-(1-methyl-1H-1,2,3-triazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediate I-1350 and 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)triazole. NMR ES/MS m/z: 652.2 (M+H+). Example 551: (S)-2-(4-(6-((4-(1-(difluoromethyl)-1H-pyrazol-4-yl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-(1-(difluoromethyl)-1H-pyrazol-4-yl)benzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediate I-1349 and 1-(difluoromethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole. 1H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.52-8.01 (m, 2H), 7.91 (ddd, J=10.9, 6.3, 2.0 Hz, 1H), 7.87-7.71 (m, 2H), 7.71-7.23 (m, 7H), 6.91 (dd, J=8.3, 5.7 Hz, 1H), 5.50 (d, J=8.5 Hz, 2H), 5.14 (s, 1H), 4.79-4.60 (m, 3H), 4.52 (ddd, J=11.6, 6.7, 3.2 Hz, 1H), 3.99 (dd, J=8.9, 3.2 Hz, 1H), 3.84 (dd, J=8.9, 4.1 Hz, 1H), 1.41 (d, J=5.4 Hz, 3H), 0.76 (d, J=4.4 Hz, 3H). ES/MS m/z: 686.2 (M+H+). Example 552: (S)-2-(4-(6-((4-chloro-6-(1-(difluoromethyl)-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(4-(6-((4-chloro-6-(1-(difluoromethyl)-1H-pyrazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediate I-1348 and 1-(difluoromethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.80-8.63 (m, 2H), 8.29 (s, 1H), 8.17 (dd, J=8.6, 1.4 Hz, 1H), 8.05-7.89 (m, 2H), 7.90-7.74 (m, 2H), 7.74-7.35 (m, 3H), 6.96 (dd, J=8.3, 0.7 Hz, 1H), 5.67 (s, 2H), 5.13 (d, J=6.5 Hz, 1H), 4.79-4.59 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 3.99 (d, J=8.9 Hz, 1H), 3.83 (d, J=8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 721.2 (M+H+). Example 553: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(2-methyl-2H-1,2,3-triazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(2-methyl-2H-1,2,3-triazol-4-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediate I-1220 and 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)triazole. 1H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.18 (dd, J=8.6, 1.4 Hz, 1H), 8.02 (s, 1H), 7.94 (dd, J=10.9, 6.3 Hz, 1H), 7.89-7.72 (m, 2H), 7.71-7.50 (m, 4H), 7.39 (dd, J=11.2, 6.0 Hz, 1H), 6.93 (d, J=8.2 Hz, 1H), 5.58 (s, 2H), 5.14 (d, J=6.5 Hz, 1H), 4.80-4.59 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.22 (s, 3H), 3.99 (d, J=8.9 Hz, 1H), 3.83 (d, J=8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 669.1 (M+H+). Example 554: (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-1,2,3-triazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((2-fluoro-4-(1-methyl-1H-1,2,3-triazol-5-yl)benzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediate I-1220 and 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)triazole. 1H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (dd, J=8.6, 1.4 Hz, 1H), 7.97-7.86 (m, 2H), 7.84 (dd, J=8.3, 7.5 Hz, 1H), 7.80-7.68 (m, 2H), 7.59 (dd, J=7.2, 1.6 Hz, 1H), 7.49-7.33 (m, 3H), 6.95 (dd, J=8.3, 0.6 Hz, 1H), 5.64 (s, 2H), 5.13 (d, J=6.6 Hz, 1H), 4.81-4.57 (m, 3H), 4.52 (dd, J=11.6, 6.7 Hz, 1H), 4.13 (s, 3H), 3.99 (d, J=8.9 Hz, 1H), 3.84 (d, J=8.9 Hz, 1H), 1.41 (s, 3H), 0.75 (s, 3H). ES/MS m/z: 669.1 (M+H+). Example 555: (S)-2-(2,5-difluoro-4-(6-((4-fluoro-6-(1-methyl-11H-1,2,3-triazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(6-((4-fluoro-6-(1-methyl-1H-1,2,3-triazol-4-yl)pyridin-3-yl)methoxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 41, starting with Intermediate I-1347 and 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)triazole. 1H NMR (400 MHz, Methanol-d4) δ 8.89 (d, J=9.8 Hz, 1H), 8.83 (s, 1H), 8.23 (s, 1H), 8.12 (dd, J=8.6, 1.4 Hz, 1H), 8.03-7.90 (m, 1H), 7.88-7.69 (m, 3H), 7.65-7.51 (m, 1H), 7.36 (dd, J=11.2, 6.1 Hz, 1H), 6.94 (dd, J=8.2, 0.7 Hz, 1H), 5.68 (s, 2H), 5.09 (d, J=6.4 Hz, 1H), 4.76-4.56 (m, 3H), 4.52 (dd, J=11.5, 6.7 Hz, 1H), 4.40 (s, 3H), 3.99 (d, J=8.9 Hz, 1H), 3.83 (d, J=8.9 Hz, 1H), 1.40 (s, 3H), 0.74 (s, 3H). ES/MS m/z: 670.1 (M+H+). Example 556: 2-(2,5-difluoro-4-(6-((4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(2,5-difluoro-4-(6-((4-(trifluoromethyl)benzyl)oxy)pyridin-2-yl)benzyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1382 and I-1267. 1H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.94-7.88 (m, 1H), 7.81 (dd, J=8.5, 1.5 Hz, 1H), 7.78-7.69 (m, 5H), 7.61 (d, J=8.4 Hz, 1H), 7.53 (dd, J=7.5, 1.7 Hz, 1H), 7.38 (dd, J=11.4, 6.1 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.59 (s, 2H), 5.52 (s, 1H), 4.58-4.40 (m, 3H), 4.21 (dd, J=10.8, 6.5 Hz, 1H), 4.08 (t, J=8.8 Hz, 1H), 3.82-3.72 (m, 2H), 3.18-2.99 (m, 2H), 2.91 (s, 3H). ES/MS m/z: 654.0 (M+H+). Example 557: (S)-2-(2,5-difluoro-4-(5-fluoro-6-((4-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(5-fluoro-6-((4-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1381 and I-82. 1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.89-7.77 (m, 3H), 7.63 (d, J=8.5 Hz, 1H), 7.61-7.52 (m, 3H), 7.48 (dd, J=11.5, 6.1 Hz, 1H), 7.30-7.17 (m, 2H), 5.55 (s, 2H), 5.03 (d, J=6.6 Hz, 1H), 4.62-4.55 (m, 1H), 4.53 (d, J=1.5 Hz, 1H), 4.48-4.35 (m, 2H), 3.84-3.68 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 606.0 (M+H+). Example 558: (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2,3,6-trifluoro-4-(5-fluoro-6-((4-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2,3,6-trifluoro-4-(5-fluoro-6-((4-fluorobenzyl)oxy)pyridin-2-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1381 and I-1231. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.90 (dd, J=10.3, 8.2 Hz, 1H), 7.78 (dd, J=8.5, 1.5 Hz, 1H), 7.76-7.67 (m, 1H), 7.63-7.53 (m, 4H), 7.31-7.20 (m, 2H), 5.56 (s, 2H), 5.09 (d, J=6.6 Hz, 1H), 4.73-4.52 (m, 2H), 4.47 (dd, J=11.2, 6.7 Hz, 1H), 4.35 (d, J=17.4 Hz, 1H), 3.84-3.70 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 624.0 (M+H+). Example 559: 2-((3′-((4-chloro-2-fluorobenzyl)oxy)-2,4′,5-trifluoro-[1,1′-biphenyl]-4-yl)methyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-((3′-((4-chloro-2-fluorobenzyl)oxy)-2,4′,5-trifluoro-[1,1′-biphenyl]-4-yl)methyl)-1-((3S,4S)-4-(methoxymethyl)tetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1309 and I-1267. 1H NMR (400 MHz, MeOD) δ 8.74 (s, 1H), 7.97 (dd, J=8.5, 1.5 Hz, 1H), 7.69-7.62 (m, 1H), 7.62-7.52 (m, 1H), 7.41-7.29 (m, 2H), 7.29-7.22 (m, 2H), 7.22-7.10 (m, 2H), 5.49 (t, J=7.3 Hz, 1H), 5.27-5.17 (m, 2H), 4.65-4.47 (m, 3H), 4.33-4.22 (m, 1H), 4.00-3.87 (m, 1H), 2.96 (s, 3H). ES/MS m/z: 655.2 (M+H+). Example 560: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-(difluoromethoxy)ethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-(difluoromethoxy)ethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-1355 and I-1352. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.82 (s, 1H), 8.30 (s, 1H), 8.25 (d, J=1.5 Hz, 1H), 8.02 (d, J=1.3 Hz, 1H), 7.94-7.85 (m, 2H), 7.80 (dd, J=8.4, 1.6 Hz, 1H), 7.61 (d, J=8.5 Hz, 1H), 7.57-7.52 (m, 1H), 7.40 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 6.64 (t, J=75.4 Hz, 1H), 5.67 (s, 2H), 4.72 (t, J=5.1 Hz, 2H), 4.44 (s, 2H), 4.20 (t, J=5.0 Hz, 2H). ES/MS m/z: 668.0 (M+H+). Example 561: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-isopropoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-isopropoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-1356 and I-1352. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.82 (s, 1H), 8.30 (s, 1H), 8.27 (d, J=1.5 Hz, 1H), 8.02 (d, J=1.2 Hz, 1H), 7.94-7.86 (m, 2H), 7.83 (dd, J=8.4, 1.5 Hz, 1H), 7.62 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.4, 1.7 Hz, 1H), 7.40 (dd, J=11.4, 6.1 Hz, 1H), 5.66 (s, 2H), 4.58 (t, J=5.1 Hz, 2H), 4.52 (s, 2H), 3.71 (t, J=5.0 Hz, 2H), 3.45 (p, J=6.0 Hz, 1H), 0.95 (s, 3H), 0.93 (s, 3H). ES/MS m/z: 660.0 (M+H+). Example 562: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-cyclopropoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-cyclopropoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-1357 and I-1352. 1H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J=1.3 Hz, 1H), 8.83 (s, 1H), 8.31 (s, 1H), 8.26 (d, J=1.5 Hz, 1H), 8.03 (d, J=1.3 Hz, 1H), 7.96-7.86 (m, 2H), 7.84 (dd, J=8.5, 1.6 Hz, 1H), 7.63 (d, J=8.4 Hz, 1H), 7.55 (dd, J=7.5, 1.7 Hz, 1H), 7.40 (dd, J=11.5, 6.1 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 5.67 (s, 2H), 4.60 (t, J=5.1 Hz, 2H), 4.46 (s, 2H), 3.79 (t, J=5.0 Hz, 2H), 3.25 (tt, J=6.1, 3.0 Hz, 1H), 0.31 (ddd, J=13.8, 6.6, 2.7 Hz, 2H), 0.24 (tt, J=6.3, 3.2 Hz, 2H). ES/MS m/z: 658.1 (M+H+). Example 563: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxy-2-methylpropyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxy-2-methylpropyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1366 and I-1049. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.2 Hz, 1H), 8.81 (s, 1H), 8.39 (d, J=1.5 Hz, 1H), 8.30 (s, 1H), 8.03 (d, J=1.3 Hz, 1H), 7.93-7.86 (m, 2H), 7.84 (dd, J=8.5, 1.3 Hz, 1H), 7.62 (d, J=8.4 Hz, 1H), 7.55 (dd, J=7.4, 1.7 Hz, 1H), 7.41 (dd, J=11.5, 6.1 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.66 (s, 2H), 4.55 (s, 2H), 4.50 (s, 2H), 3.10 (s, 3H), 1.21 (s, 6H). ES/MS m/z: 660.2 (M+H+). Example 564: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2S,3S)-3-methoxybutan-2-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2S,3S)-3-methoxybutan-2-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1364 and I-102. 1H NMR (400 MHz, DMSO-d6) δ 8.32 (d, J=1.5 Hz, 1H), 7.90-7.85 (m, 1H), 7.85-7.78 (m, 2H), 7.64-7.57 (m, 2H), 7.53-7.47 (m, 2H), 7.38 (dd, J=11.5, 6.1 Hz, 1H), 7.32 (dd, J=8.2, 2.1 Hz, 1H), 6.94 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 4.70 (s, 1H), 4.45 (q, J=16.8 Hz, 2H), 3.93-3.81 (m, 1H), 3.00 (s, 3H), 1.62 (d, J=7.1 Hz, 3H), 1.21 (d, J=6.1 Hz, 3H). ES/MS m/z: 610.2 (M+H+). Example 565: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2R,3R)-3-methoxybutan-2-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((2R,3R)-3-methoxybutan-2-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1362 and I-102. 1H NMR (400 MHz, DMSO-d6) δ 8.33 (d, J=1.5 Hz, 1H), 7.91-7.85 (m, 1H), 7.85-7.79 (m, 2H), 7.63 (d, J=8.3 Hz, 1H), 7.59 (d, J=8.2 Hz, 1H), 7.50 (td, J=10.0, 9.3, 1.9 Hz, 2H), 7.39 (dd, J=11.5, 6.1 Hz, 1H), 7.32 (dd, J=8.3, 2.1 Hz, 1H), 6.94 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 4.71 (s, 1H), 4.46 (q, J=16.8 Hz, 2H), 3.88 (dd, J=8.1, 6.1 Hz, 1H), 3.00 (s, 3H), 1.62 (d, J=7.1 Hz, 3H), 1.21 (d, J=6.1 Hz, 3H). ES/MS m/z: 610.2 (M+H+). Example 566: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-isobutoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid -(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-isobutoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-1358 and I-1255. 1H NMR (400 MHz, DMSO-d6) δ 8.26 (d, J=1.5 Hz, 1H), 7.91-7.85 (m, 1H), 7.84-7.82 (m, 1H), 7.81 (d, J=1.5 Hz, 1H), 7.66-7.56 (m, 1H), 7.61 (d, J=3.2 Hz, 1H), 7.54-7.46 (m, 2H), 7.37 (dd, J=11.5, 6.1 Hz, 1H), 7.33 (dd, J=8.4, 2.1 Hz, 1H), 6.95 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 4.61 (t, J=5.1 Hz, 2H), 4.50 (s, 2H), 3.71 (t, J=5.0 Hz, 2H), 3.10 (d, J=6.5 Hz, 2H), 1.66 (hept, J=6.7 Hz, 1H), 0.72 (s, 3H), 0.70 (s, 3H). ES/MS m/z: 624.3 (M+H+). Example 567: 2-(4-(6-((4-chloro-6-(4,5,6,7-tetrahydro-2H-benzo[d][1,2,3]triazol-2-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(4,5,6,7-tetrahydro-2H-benzo[d][1,2,3]triazol-2-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 38, starting with Intermediates I-1359 and I-1354. 1H NMR (400 MHz, DMSO-d6) δ 8.73 (s, 1H), 8.26 (d, J=1.5 Hz, 1H), 8.04 (s, 1H), 7.93-7.86 (m, 2H), 7.84 (dd, J=8.4, 1.5 Hz, 1H), 7.64 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.41 (dd, J=11.5, 6.1 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 5.62 (s, 2H), 4.63 (t, J=5.1 Hz, 2H), 4.50 (s, 2H), 3.69 (t, J=5.0 Hz, 2H), 3.21 (s, 3H), 2.77 (d, J=5.5 Hz, 4H), 1.83 (q, J=3.3 Hz, 4H). ES/MS m/z: 686.3 (M+H+). Example 568: 2-(4-(6-((4-chloro-6-(4,5,6,7-tetrahydro-1H-benzo[d][1,2,3]triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(4,5,6,7-tetrahydro-1H-benzo[d][1,2,3]triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 38, starting with Intermediates I-1359 and I-1353. 1H NMR (400 MHz, DMSO-d6) δ 8.79 (s, 1H), 8.24 (d, J=1.5 Hz, 1H), 8.20 (s, 1H), 7.91 (t, J=7.9 Hz, 1H), 7.88-7.79 (m, 2H), 7.62 (d, J=8.4 Hz, 1H), 7.54 (dd, J=7.5, 1.7 Hz, 1H), 7.40 (dd, J=11.5, 6.0 Hz, 1H), 7.00 (d, J=8.2 Hz, 1H), 5.64 (s, 2H), 4.62 (t, J=5.2 Hz, 2H), 4.48 (s, 2H), 3.68 (t, J=5.1 Hz, 2H), 3.21 (s, 3H), 3.03 (s, 2H), 2.71 (s, 2H), 1.82-1.71 (m, 4H). ES/MS m/z: 686.1 (M+H+). Example 569: 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-isobutoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-6-(1H-1,2,3-triazol-1-yl)pyridin-3-yl)methoxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(2-isobutoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 22, starting with Intermediates I-1358 and I-1352. 1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J=1.3 Hz, 1H), 8.82 (s, 1H), 8.30 (s, 1H), 8.28 (d, J=1.5 Hz, 1H), 8.02 (d, J=1.3 Hz, 1H), 7.94-7.90 (m, 1H), 7.90-7.86 (m, 1H), 7.84 (dd, J=8.4, 1.5 Hz, 1H), 7.63 (d, J=8.5 Hz, 1H), 7.54 (dd, J=7.3, 1.7 Hz, 1H), 7.39 (dd, J=11.4, 6.1 Hz, 1H), 7.00 (dd, J=8.3, 0.6 Hz, 1H), 5.66 (s, 2H), 4.63 (t, J=5.0 Hz, 2H), 4.53 (s, 2H), 3.71 (t, J=4.9 Hz, 2H), 3.10 (d, J=6.5 Hz, 2H), 1.65 (hept, J=6.7 Hz, 1H), 0.72 (s, 3H), 0.70 (s, 3H). ES/MS m/z: 674.1 (M+H+). Example 570: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-isopropoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-isopropoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 37, starting with Intermediates I-1346 and I-1368. 1H NMR (400 MHz, DMSO) δ 8.36 (d, J=1.5 Hz, 1H), 7.93-7.79 (m, 3H), 7.71 (d, J=8.5 Hz, 1H), 7.60 (t, J=8.2 Hz, 1H), 7.56-7.46 (m, 2H), 7.46-7.26 (m, 2H), 6.95 (d, J=8.3 Hz, 1H), 5.50 (s, 2H), 5.29 (dt, J=7.4, 3.5 Hz, 1H), 4.67-4.13 (m, 6H), 3.78-3.59 (m, 1H), 3.37 (m, J=7.0 Hz, 1H), 1.00 (d, J=7.0 Hz, 6H). ES/MS m/z: 652.2 (M+H+). Example 571: 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-ethoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid 2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-((3R,4R)-4-ethoxytetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 37, starting with Intermediates I-1346 and I-1367. 1H NMR (400 MHz, DMSO) δ 8.34 (d, J=1.5 Hz, 1H), 7.93-7.79 (m, 3H), 7.69 (d, J=8.5 Hz, 1H), 7.60 (t, J=8.2 Hz, 1H), 7.50 (ddd, J=8.6, 5.6, 2.0 Hz, 2H), 7.45-7.27 (m, 2H), 6.95 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 5.39-5.18 (m, 1H), 4.64-4.46 (m, 2H), 4.46-4.34 (m, 2H), 4.34-4.12 (m, 2H), 3.71 (h, J=5.2 Hz, 1H), 3.45-3.30 (m, 2H), 1.00 (t, J=7.0 Hz, 3H). ES/MS m/z: 638.1 (M+H+). Example 572: (R)-1-(2-oxabicyclo[3.1.1]heptan-4-yl)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid Example 573: (S)-1-(2-oxabicyclo[3.1.1]heptan-4-yl)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid Examples 572 and 572: (R)-1-(2-oxabicyclo[3.1.1]heptan-4-yl)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 572) and (S)-1-(2-oxabicyclo[3.1.1]heptan-4-yl)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 573) were prepared via preparative chiral SFC (Chiralpak OJ-H column, MeOH/CO2eluent) of Example 407. Example 574: (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2,3,6-trifluoro-4-(2-((4-fluorobenzyl)oxy)pyrimidin-4-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-1-(4,4-dimethyltetrahydrofuran-3-yl)-2-(2,3,6-trifluoro-4-(2-((4-fluorobenzyl)oxy)pyrimidin-4-yl)benzyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1387 and I-1231. 1H NMR (400 MHz, DMSO) δ 8.84 (d, J=5.1 Hz, 1H), 8.48 (s, 1H), 7.84 (ddd, J=9.9, 5.6, 2.0 Hz, 1H), 7.78 (dd, J=8.4, 1.5 Hz, 1H), 7.68 (dd, J=5.1, 1.8 Hz, 1H), 7.63-7.53 (m, 3H), 7.30-7.20 (m, 2H), 5.50 (s, 2H), 5.13-5.04 (m, 1H), 4.68 (d, J=17.3 Hz, 1H), 4.57 (d, J=11.5 Hz, 1H), 4.51-4.34 (m, 2H), 3.79-3.74 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H). ES/MS m/z: 607.0 (M+H+). Example 575: (S)-2-(2,5-difluoro-4-(2-((4-fluorobenzyl)oxy)pyrimidin-4-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (S)-2-(2,5-difluoro-4-(2-((4-fluorobenzyl)oxy)pyrimidin-4-yl)benzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Procedure 35, starting with Intermediates I-1387 and I-82. 1H NMR (400 MHz, DMSO) δ 8.78 (d, J=5.2 Hz, 1H), 8.48 (s, 1H), 7.94 (dd, J=10.2, 6.3 Hz, 1H), 7.80 (dd, J=8.4, 1.5 Hz, 1H), 7.70-7.50 (m, 5H), 7.29-7.19 (m, 2H), 5.49 (s, 2H), 5.02 (d, J=6.7 Hz, 1H), 4.61-4.50 (m, 2H), 4.49-4.36 (m, 2H), 3.81-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H). ES/MS m/z: 589.1 (M+H+). Example 576: (5-methyl-2-oxo-1,3-dioxol-4-yl)methyl (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate Procedure 42 (5-methyl-2-oxo-1,3-dioxol-4-yl)methyl (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate: Na2CO3(17 mg, 0.16 mmol) followed by 4-(chloromethyl)-5-methyl-1,3-dioxol-2-one (0.018 mL, 0.16 mmol) were added to a mixture of (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 115, 50 mg, 0.08 mmol) in DMF (1.0 mL). The resulting mixture was stirred at rt overnight. The mixture was diluted with EtOAc and washed with water then 10% LiCl. Organic phase was dried over MgSO4, filtered, concentrated, and purified by silica gel flash column chromatography (EtOAc in Hex) to yield the title compound. 1H NMR (400 MHz, DMSO-d6) δ 8.52 (s, 1H), 7.88 (dd, J=8.2, 7.5 Hz, 1H), 7.85-7.79 (m, 2H), 7.65 (d, J=8.6 Hz, 1H), 7.60 (t, J=8.2 Hz, 1H), 7.55-7.51 (m, 1H), 7.49 (dd, J=10.1, 2.1 Hz, 1H), 7.47-7.42 (m, 1H), 7.33 (dd, J=8.3, 2.1 Hz, 1H), 6.95 (d, J=8.2 Hz, 1H), 5.50 (s, 2H), 5.23 (s, 2H), 5.02 (d, J=6.6 Hz, 1H), 4.53 (d, J=13.9 Hz, 2H), 4.48-4.30 (m, 2H), 3.80-3.68 (m, 2H), 2.23 (s, 3H), 1.34 (s, 3H), 1.21 (d, J=16.9 Hz, 1H), 0.59 (s, 3H). ES/MS m/z: 734.0 (M+H+). Example 577: (phosphonooxy)methyl (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate Procedure 43 ((di-tert-butoxyphosphoryl)oxy)methyl (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (577-1): Na2CO3(17 mg, 0.16 mmol) followed by di-tert-butyl (chloromethyl) phosphate (41 mg, 0.161 mmol) were added to a mixture of (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylic acid (Example 115, 50 mg, 0.08 mmol) in DMF (1.0 mL)> The resulting mixture was stirred at rt overnight. The mixture was diluted with EtOAc and washed with water then 10% LiCl. Nest, the organic phase was dried over MgSO4, filtered, concentrated, and purified by silica gel flash column chromatography (EtOAc in Hex) to yield the title compound. ES/MS: 844.1 (M+H+). (phosphonooxy)methyl (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (Example 577): A mixture of ((di-tert-butoxyphosphoryl)oxy)methyl (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (577-1, 68 mg, 0.0805 mmol) in DCM (2 mL) and TFA (0.5 mL) was stirred at rt for 30 min. Solvent was evaporated to give the title compound. 1H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.92-7.80 (m, 3H), 7.68 (d, J=8.5 Hz, 1H), 7.60 (t, J=8.2 Hz, 1H), 7.53 (dd, J=7.5, 1.6 Hz, 1H), 7.49 (dd, J=10.0, 2.1 Hz, 1H), 7.47-7.43 (m, 1H), 7.33 (dd, J=8.4, 2.1 Hz, 1H), 6.95 (d, J=8.4 Hz, 1H), 5.82-5.68 (m, 2H), 5.51 (s, 2H), 5.03 (d, J=6.7 Hz, 1H), 4.60-4.55 (m, 1H), 4.53 (s, 1H), 4.48-4.35 (m, 2H), 3.79 (d, J=8.7 Hz, 1H), 3.73 (d, J=8.6 Hz, 1H), 1.34 (s, 3H), 0.60 (s, 3H). ES/MS m/z: 731.9 (M+H+). Example 578: (isobutyryloxy)methyl (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate (isobutyryloxy)methyl (S)-2-(4-(6-((4-chloro-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(4,4-dimethyltetrahydrofuran-3-yl)-1H-benzo[d]imidazole-6-carboxylate was prepared in a manner as described in Procedure 42, using Example 115 and chloromethyl isobutyrate. ES/MS m/z: 722.1 (M+H+). Comparative Example 1 (CE-1) (S)-2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)piperidin-1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid: (S)-2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)piperidin-1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Patent Application WO2018/109607 (see Example 4A-01) and references therein. Comparative Example 2 (CE-2) (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid (Comparative Example 2): (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)-2,5-difluorobenzyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid was prepared in a manner as described in Patent Application WO2021/081207 (see Example 444) and references therein. TABLE 2CompoundsES/MSEx.StructureProcedurem/z1H NMR11585.31H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J = 1.4 Hz, 1H), 7.97-7.85 (m, 3H), 7.83-7.71 (m, 3H), 7.68 (d, J = 8.5 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.58- 5.49 (m, 1H), 4.58 ( s, 2H), 4.31 (td, J = 8.7, 2.8 Hz, 1H), 4.22 (dd, J = 10.4, 2.8 Hz, 1H), 3.97 (dd, J = 10.5,7.7 Hz, 1H), 3.72 (td, J =9.3, 7.0 Hz, 1H), 2.23-2.06(m, 1H).21598.21H NMR (400 MHz, DMSO) δ 8.22 (s, 1H), 8.09 (d, J = 1.4 Hz, 1H), 7.97- 7.82 (m, 3H), 7.81-7.71 (m, 3H), 7.68 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.38 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.68 (tt, J = 10.0, 5.2 Hz, 1H), 5.60 (s, 2H), 4.51 (s, 2H), 3.89 (t, J = 10.1 Hz, 1H), 3.62 (dd, J = 10.9, 4.6 Hz, 1H), 2.86 (dd, J = 17.9, 10.4 Hz, 1H), 2.62 (dd, J = 17.8, 5.7 Hz, 1H).31598.31H NMR (400 MHz, DMSO) δ 8.22 (s, 1H), 8.09 (d, J = 1.5 Hz, 1H), 7.96- 7.82 (m, 3H), 7.81-7.71 (m, 3H), 7.68 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.6, 1.7 Hz, 1H), 7.38 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.68 (tt, J = 10.1, 5.2 Hz, 1H), 5.60 (s, 2H), 4.51 (s, 2H), 3.89 (t, J = 10.1 Hz, 1H), 3.61 (dd, J = 11.0, 4.6 Hz, 1H), 2.86 (dd, J = 17.9, 10.4 Hz, 1H), 2.62 (dd, J = 17.9, 5.7 Hz, 1H).41605.31H NMR (400 MHz, Methanol-d4) δ 8.64-8.45 (m, 1H), 8.15 (td, J = 9.0, 1.5 Hz, 1H), 7.91-7.78 (m, 2H), 7.78-7.66 (m, 2H), 7.66-7.48 (m, 3H), 7.33 (ddd, J = 17.3, 11.2, 6.1 Hz, 1H), 7.09-6.83 (m, 1H), 6.16 (td, J = 56.2, 4.3 Hz, 1H), 5.67-5.59 (m, 2H), 4.72 (d, J = 3.1 Hz, 2H), 3.91 (q, J = 5.4, 4.0 Hz, 1H), 2.36 (d, J = 57.4 Hz, 1H), 1.94 (d, J = 9.4 Hz, 1H), 1.86-1.75 (m, 1H).51585.51H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J = 1.4 Hz, 1H), 7.97-7.85 (m, 3H), 7.83-7.71 (m, 3H), 7.68 (d, J = 8.5 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.58- 5.49 (m, 1H), 4.58 (s, 2H), 4.31 (td, J = 8.7, 2.8 Hz, 1H), 4.22 (dd, J = 10.4, 2.8 Hz, 1H), 3.97 (dd, J = 10.5,7.7 Hz, 1H), 3.72 (td, J =9.3, 7.0 Hz, 1H), 2.23-2.06(m, 1H).61583.61H NMR (400 MHz, DMSO-d6) δ 8.18 (d, J = 1.4 Hz, 1H), 7.97-7.85 (m, 3H), 7.83-7.68 (m, 4H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.41 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.16 (p, J = 8.8 Hz, 1H), 4.58 (s, 2H), 2.22-2.08 (m, 4H), 2.06- 1.94 (m, 2H), 1.89-1.69 (m, 2H).72MH+ 682.21H NMR (400 MHz, DMSO-d6) δ 9.08 (s, 1H), 8.54 (s, 1H), 8.20 (s, 1H), 7.91-7.82 (m, 2H), 7.79 (dd, J = 8.4, 1.6 Hz, 1H), 7.73 (d, J = 11.5 Hz, 1H), 7.67-7.54 (m, 3H), 7.52 (d, J = 7.3 Hz, 1H), 7.38 (dd, J = 11.7, 6.1 Hz, 1H), 6.96 (d, J = 8.2 Hz, 1H), 6.53 (s, 1H), 5.53 (s, 2H), 4.59 (t, J = 5.1 Hz, 2H), 4.44 (s, 2H), 3.68 (t, J = 5.1 Hz, 2H), 3.21 (s, 3H).83(MH+) 610.21H NMR (400 MHz, DMSO-d6) δ 8.42 (s, 1H), 7.97-7.85 (m, 2H), 7.82- 7.67 (m, 4H), 7.63 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.7, 1.6 Hz, 1H), 7.31 (dd, J = 11.5, 6.2 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.07 (s, 1H), 4.41 (s, 2H), 3.68 (t, J = 10.4 Hz, 2H), 3.14 (d, J = 11.0 Hz, 2H), 0.83 (d, J = 23.4 Hz, 2H), 0.58 (m, 2H).93(MH+) 610.21H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.98-7.81 (m, 2H), 7.77 (ddd, J = 16.8, 8.1, 1.5 Hz, 5H), 7.62 (d, J = 8.5 Hz, 1H), 7.57-7.45 (m, 1H), 7.30 (d, J = 10.5 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 6.54 (s, 0H), 5.60 (s, 2H), 5.40 (s, 0H), 5.01 (s, 1H), 4.40 (s, 2H), 3.60 (s, 1H), 3.11-2.99 (m, 1H), 0.76 (d, J = 8.8 Hz, 1H), 0.58 (s, 3H), −0.06 (s, 1H).103614.21H NMR (400 MHz, DMSO-d6) δ 9.34 (s, 2H), 8.27 (d, J = 1.4 Hz, 1H), 7.99-7.84 (m, 4H), 7.83- 7.67 (m, 5H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.28 (td, J = 10.2, 7.9 Hz, 1H), 4.68 (q, J = 7.0 Hz, 1H), 4.56-4.34 (m, 2H), 3.34 (d, J = 11.4 Hz, 1H), 3.15 (s, 3H).113631.21H NMR (400 MHz, DMSO) δ 13.09 (s, 1H), 8.35 (s, 1H), 7.96-7.86 (m, 2H), 7.83-7.70 (m, 3H), 7.59-7.42 (m, 3H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H).123631.21H NMR (400 MHz, DMSO) δ 13.10 (s, 1H), 8.35 (s, 1H), 7.96-7.86 (m, 2H), 7.83-7.70 (m, 3H), 7.59-7.48 (m, 2H), 7.46 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.03 (d, J = 6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.33 (s, 3H), 0.61 (s, 3H).133693.191H NMR (400 MHz, Methanol-d4) δ 8.71 (s, 1H), 8.17 (d, J = 1.2 Hz, 1H), 7.90-7.67 (m, 3H), 7.66- 7.47 (m, 3H), 7.19 (dd, J = 11.5, 6.0 Hz, 1H), 6.92 (d, J = 8.2 Hz, 1H), 5.63 (s, 2H), 4.55 (dd, J = 11.4, 1.9 Hz, 3H), 4.44 (dd, J = 11.3, 6.8 Hz, 1H), 3.93 (d, J = 8.9 Hz, 1H), 3.78 (d, J = 8.8 Hz, 1H), 3.37-3.34 (m, 2H), 1.30 (s, 3H), 0.61 (s, 3H).144(MH+) 652.21H NMR (400 MHz, DMSO-d6) δ 8.06 (s, 1H), 7.96-7.86 (m, 2H), 7.83- 7.67 (m, 4H), 7.64-7.55 (m, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.34 (s, 1H), 5.19 (d, J = 8.0 Hz, 1H), 4.35 (dt, J = 28.0, 16.9 Hz, 3H), 4.23-4.07 (m, 1H), 4.07-3.82 (m, 2H), 3.82- 3.53 (m, 1H), 2.10 (s, 2H), 2.03 (s, 1H), 1.07-0.78 (m, 2H), 0.78-0.53 (m, 1H), 0.29-0.04 (m, 1H).154(MH+) 652.21H NMR (400 MHz, DMSO-d6) δ 12.79 (s, 1H), 8.12 (s, 0H), 8.06 (d, J = 1.4 Hz, 1H), 7.96-7.84 (m, 2H), 7.83-7.66 (m, 4H), 7.63 (dd, J = 8.5, 3.3 Hz, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.34 (s, 0H), 5.19 (d, J = 8.0 Hz, 1H), 4.63-4.22 (m, 3H), 4.19-4.05 (m, 1H), 3.99 (dd, J = 13.5, 3.3 Hz, 1H), 3.92 (d, J = 10.7 Hz, 1H), 3.82-3.46 (m, 2H), 2.10 (s,2H), 2.03 (s, 1H), 1.08-0.75 (m, 2H), 0.71 (dd, J =10.2, 5.3 Hz, 1H), 0.17 (ddt,J = 16.4, 11.2, 5.8 Hz, 1H).164656.21H NMR (400 MHz, DMSO-d6) δ 8.12 (dd, J = 37.9, 1.4 Hz, 1H), 7.96- 7.81 (m, 3H), 7.79-7.71 (m, 3H), 7.68 (d, J = 8.5 Hz, 1H), 7.56-7.51 (m, 1H), 7.43-7.33 (m, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.33 (dq, J = 27.6, 8.0 Hz, 1H), 4.55 (dd, J = 13.1, 6.1 Hz, 2H), 4.43 (dd, J = 16.8, 5.7 Hz, 1H), 4.24- 4.07 (m, 1H), 4.07-3.93 (m, 1H), 3.41 (ddd, J = 84.8, 11.4, 6.6 Hz, 1H), 3.17 (d, J = 5.9 Hz, 3H), 2.05 (d, J = 24.1 Hz, 3H).174642.21H NMR (400 MHz, DMSO-d6) δ 8.25 (s, 1H), 8.00 (s, 1H), 7.97-7.86 (m, 4H), 7.76 (m, 3H), 7.54 (d, J = 8.4 Hz, 2H), 6.99 (d, J = 8.2 Hz, 2H), 5 .64 (s, 2H), 4.64 (d, J = 17.2 Hz, 5H), 4.53 (d, J = 16.0 Hz, 3H).184668.21H NMR (400 MHz, DMSO-d6) δ 8.57 (dd, J = 15.4, 1.4 Hz, 1H), 7.95- 7.85 (m, 2H), 7.84-7.68 (m, 4H), 7.63 (dd, J = 8.5, 7.0 Hz, 1H), 7.54 (dt, J = 7.5, 1.9 Hz, 1H), 7.44 (dt, J = 11.3, 5.4 Hz, 1H), 7.00 (dd, J = 8.3, 1.9 Hz, 1H), 5.66-5.52 (m, 3H), 4.76- 4.57 (m, 2H), 4.57-4.45 (m, 3H), 4.13-4.05 (m, 2H), 4.00 (d, J = 13.2 Hz, 0H), 3.89 (d, J = 12.4 Hz, 1H), 3.55 (dd, J = 12.5, 4.6 Hz, 1H), 3.40 (d, J = 9.1 Hz, 0H), 3.28 (t, J = 9.9 Hz, 0H),3.21-3.06 (m, 1H), 2.76(dd, J = 12.9, 9.8 Hz, 1H),2.70-2.57 (m, 1H), 2.42-2.31 (m, 1H), 1.74 (s, 3H),0.89 (s, 1H).195MH+ 627.21H NMR (400 MHz, DMSO-d6) δ 12.78 (s, 1H), 8.51 (d, J = 1.4 Hz, 1H), 7.97-7.85 (m, 2H), 7.84- 7.68 (m, 5H), 7.64 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.3, 1.6 Hz, 1H), 7.41 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.80 (d, J = 5.3 Hz, 1H), 5.60 (s, 2H), 5.48-5.30 (m, 1H), 4.71 (dd, J = 11.1, 2.5 Hz, 1H), 4.65-4.30 (m, 2H), 4.16 (dd, J = 11.1, 7.0 Hz, 1H), 3.82-3.62 (m, 1H), 3.57- 3.45 (m, 1H), 3.30 (s, 1H), 1.73-1.56 (m, 1H).205MH+ 627.21H NMR (400 MHz, DMSO-d6) δ 12.78 (s, 1H), 8.51 (d, J = 1.4 Hz, 1H), 7.98-7.84 (m, 2H), 7.84- 7.68 (m, 4H), 7.64 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.41 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.80 (d, J = 5.3 Hz, 1H), 5.60 (s, 2H), 5.40 (ddd, J = 9.5, 7.0, 2.6 Hz, 1H), 4.72 (dd, J = 11.0, 2.6 Hz, 1H), 4.58-4.27 (m, 2H), 4.16 (dd, J = 11.1, 6.9 Hz, 1H), 3.72 (td, J = 8.6,2.2 Hz, 1H), 3.62-3.42 (m,1H), 1.61 (dtd, J = 13.2,10.7, 8.7 Hz, 1H), 0.77 (dd,J = 13.3, 6.3 Hz, 1H).215638.41H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.13 (dd, J = 8.6, 1.4 Hz, 1H), 8.00-7.81 (m, 2H), 7.81-7.73 (m, 1H), 7.68 (t, J = 7.5 Hz, 1H), 7.58 (dd, J = 7.4, 1.6 Hz, 1H), 7.46- 7.30 (m, 3H), 7.01-6.85 (m, 1H), 6.79 (s, 1H), 5.61 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.70-4.61 (m, 3H), 4.52 (dd, J = 11.5, 6.7 Hz, 1H), 3.99 (d, J = 8.9 Hz,1H), 3.84 (d, J = 8.9 Hz,1H), 1.40 (s, 3H), 0.74 (s,3H).225623.31H NMR (400 MHz, Methanol-d4) δ 8.53 (t, J = 0.9 Hz, 1H), 8.19 (dd, J = 8.6, 1.4 Hz, 1H), 7.90-7.79 (m, 2H), 7.79-7.67 (m, 2H), 7.60 (ddt, J = 9.3, 8.0, 1.5 Hz, 3H), 7.37 (dd, J = 11.2, 6.0 Hz, 1H), 6.96 (d, J = 8.2 Hz, 1H), 6.26 (t, J = 74.2 Hz, 1H), 5.64 (s, 2H), 4.82-4.62 (m, 5H), 1.54 (d, J = 5.1 Hz, 3H).2366281H NMR (400 MHz, DMSO-d6) δ 8.59 (t, J = 1.3 Hz, 1H), 8.30 (d, J = 1.5 Hz, 1H), 8.25 (dd, J = 9.5, 1.9 Hz, 1H), 7.94-7.84 (m, 2H), 7.74 (dd, J = 10.6, 6.4 Hz, 1H), 7.66 (d, J = 8.5 Hz, 1H), 7.52 (dd, J = 7.5, 1.6 Hz, 1H), 7.41 (dd, J = 11.5, 6.1 Hz, 1H), 6.96 (d, J = 8.2 Hz, 1H), 5.58 (d, J = 1.8 Hz, 2H), 4.66 (t, J = 5.1 Hz, 2H), 4.52 (s, 2H), 3.72-3.68 (m, 2H), 3.21 (s, 3H).2465661H NMR (400 MHz, DMSO-d6) δ 8.82 (d, J = 1.3 Hz, 1H), 8.68 (d, J = 1.3 Hz, 1H), 8.27 (d, J = 1.5 Hz, 1H), 7.91 (t, J = 7.9 Hz, 1H), 7.85 (dd, J = 8.5, 1.5 Hz, 1H), 7.71 (dd, J = 10.5, 6.4 Hz, 1H), 7.64 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.39 (dd, J = 11.5, 6.1 Hz, 1H), 7.03 (d, J = 8.3 Hz, 1H), 5.63 (s, 2H), 4.63 (t, J = 5.1 Hz, 2H), 4.49 (s, 2H), 3.69 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H).256559.41H NMR (400 MHz, Methanol-d4) δ 8.56-8.44 (m, 2H), 8.17 (dt, J = 8.6, 1.4 Hz, 1H), 7.91-7.80 (m, 2H), 7.73 (dd, J = 14.4, 8.3 Hz, 2H), 7.56 (dd, J = 7.5, 1.6 Hz, 1H), 7.33 (dd, J = 11.2, 6.1 Hz, 1H), 6.96 (d, J = 8.2 Hz, 1H), 5.65 (s, 2H), 4.76 (t, J = 5.0 Hz, 2H), 4.71 (s, 2H), 3.81 (t, J = 4.9 Hz, 2H), 3.30 (s, 3H), 2.83 (s, 3H), 2.74 (s, 3H).266566.41H NMR (400 MHz, Methanol-d4) δ 8.32 (dd, J = 1.5, 0.7 Hz, 1H), 8.12 (dd, J = 8.6, 1.5 Hz, 1H), 7.83 (dd, J = 10.7, 6.3 Hz, 1H), 7.78- 7.69 (m, 2H), 7.56-7.47 (m, 1H), 7.22 (ddd, J = 8.7, 5.6, 3.2 Hz, 1H), 7.18-7.13 (m, 1H), 7.13-7.04 (m, 1H), 7.04-6.94 (m, 1H), 6.86 (dd, J = 8.3, 0.7 Hz, 1H), 5.50 (d, J = 1.2 Hz, 2H), 4.63-4.55 (m, 4H), 3.76 (t, J = 5.0 Hz, 2H), 3.29 (s, 3H).276584.41H NMR (400 MHz, Methanol-d4) δ 8.46 (d, J = 1.3 Hz, 1H), 8.15 (dd, J = 8.6, 1.5 Hz, 1H), 7.89 (dd, J = 10.8, 6.3 Hz, 1H), 7.81 (t, J = 7.9 Hz, 1H), 7.74 (d, J = 8.5 Hz, 1H), 7.56 (dd, J = 7.4, 1.6 Hz, 1H), 7.48 (ddd, J = 10.8, 8.9, 6.6 Hz, 1H), 7.30 (dd, J = 11.3, 6.0 Hz, 1H), 7.22 (td, J = 10.0, 6.5 Hz, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 4.74 (t, J = 5.0 Hz, 2H), 4.68 (s, 2H), 3.81 (t, J = 4.9 Hz, 2H), 3.30 (s, 3H).286580.41H NMR (400 MHz, Methanol-d4) δ 8.37 (d, J = 1.4 Hz, 1H), 8.14 (dd, J = 8.6, 1.5 Hz, 1H), 7.87 (dd, J = 10.8, 6.3 Hz, 1H), 7.78- 7.71 (m, 2H),7.52 (dd, J = 7.3, 1.6 Hz, 1H), 7.27-7.10 (m, 2H), 6.96 (dd, J = 10.0, 6.1 Hz, 1H), 6.86 (d, J = 8.1 Hz, 1H), 5.46 (s, 2H), 4.65 (t, J = 5.0 Hz, 2H), 4.62 (s, 2H), 3.84-3.70 (m, 2H), 3.30 (s, 3H), 2.25 (d, J = 2.0 Hz, 3H).296546.41H NMR (400 MHz, Methanol-d4) δ 8.65 (d, J = 1.4 Hz, 1H), 8.58 (t, J = 1.0 Hz, 1H), 8.53 (d, J = 1.4 Hz, 1H), 8.25 (dd, J = 8.6, 1.4 Hz, 1H), 7.85 (d, J = 7.7 Hz, 1H), 7.82 (t, J = 3.2 Hz, 1H), 7.79 (d, J = 8.6 Hz, 1H), 7.58 (dd, J = 7.5, 1.6 Hz, 1H), 7.37 (dd, J = 11.2, 6.1 Hz, 1H), 6.98 (d, J = 8.1 Hz, 1H), 5.61 (s, 2H), 4.84 (d, J = 5.0 Hz, 2H), 4.78 (s, 2H), 3.91-3.79 (m, 2H), 3.32 (s, 3H), 2.56 (s, 3H).306570.21H NMR (400 MHz, Methanol-d4) δ 8.99 (t, J = 1.3 Hz, 1H), 8.49 (d, J = 1.5 Hz, 1H), 8.29-8.22 (m, 1H), 8.15 (td, J = 8.6, 1.5 Hz, 2H), 8.06 (d, J = 2.2 Hz, 1H), 8.00-7.83 (m, 3H), 7.75 (d, J = 8.5 Hz, 1H), 7.60 (dd, J = 7.4, 1.6 Hz, 1H), 7.34 (dd, J = 11.3, 6.0 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.69 (d, J = 1.1 Hz, 2H), 4.76 (t, J = 5.0 Hz, 2H), 4.70 (s, 2H), 3.88-3.73 (m, 2H), 3.31 (s, 3H).316567.31H NMR (400 MHz, Methanol-d4) δ 8.56 (t, J = 1.0 Hz, 1H), 8.39 (d, J = 2.4 Hz, 1H), 8.23 (dd, J = 8.6, 1.4 Hz, 1H), 7.94 (dd, J = 10.9, 6.3 Hz, 1H), 7.88- 7.74 (m, 2H), 7.66 (ddd, J = 9.7, 8.5, 2.4 Hz, 1H), 7.58 (dd, J = 7.3, 1.6 Hz, 1H), 7.36 (dd, J = 11.2, 6.1 Hz, 1H), 6.91 (dd, J = 8.3, 0.6 Hz, 1H), 5.62 (d, J = 2.0 Hz, 2H), 4.83 (t, J = 5.0 Hz, 2H), 4.77 (s, 2H), 3.85 (dd, J = 5.4, 4.4 Hz, 2H), 3.33 (s, 3H).326614.521H NMR (400 MHz, Methanol-d4) δ 8.50 (d, J = 1.4 Hz, 1H), 8.18 (dd, J = 8.5, 1.5 Hz, 1H), 7.92-7.68 (m, 3H), 7.66-7.46 (m, 3H), 7.40-7.20 (m, 3H), 6.92 (d, J = 8.3 Hz, 1H), 5.52 (s, 2H), 4.77 (t, J = 5.0 Hz, 2H), 4.71 (s, 2H), 3.82 (t, J = 4.9 Hz, 2H), 3.31 (s, 3H).336613.451H NMR (400 MHz, Methanol-d4) δ 8.53 (d, J = 1.3 Hz, 1H), 8.20 (dd, J = 8.6, 1.4 Hz, 1H), 8.05 (d, J = 7.9 Hz, 1H), 7.91-7.70 (m, 3H), 7.64 (d, J = 7.9 Hz, 1H), 7.57 (dd, J = 7.4, 1.6 Hz, 1H), 7.35 (dd, J = 11.2, 6.1 Hz, 1H), 6.98 (d, J = 8.2 Hz, 1H), 5.62 (s, 2H), 4.83- 4.64 (m, 4H), 3.88-3.78 (m, 2H), 3.32 (s, 3H), 2.69 (s, 3H).346633.581H NMR (400 MHz, Chloroform-d) δ 8.24 (t, J = 0.9 Hz, 1H), 8.13 (dd, J = 8.6, 1.4 Hz, 1H), 8.10-8.03 (m, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.82-7.69 (m, 2H), 7.65 (d, J = 7.8 Hz, 1H), 7.56 (dd, J = 7.5, 1.3 Hz, 1H), 7.35-7.32 (m, 1H), 6.92 (dd, J = 8.2, 0.7 Hz, 1H), 5.63 (s, 2H), 4.74 (s, 2H), 4.57 (t, J = 4.9 Hz, 2H), 3.78 (t, J = 4.8 Hz, 2H), 3.32 (s, 3H).356629.291H NMR (400 MHz, Chloroform-d) δ 8.24 (d, J = 19.5 Hz, 2H), 8.09 (d, J = 8.5 Hz, 1H), 7.96 (d, J = 8.4 Hz, 1H), 7.91-7.73 (m, 2H), 7.68 (t, J = 8.0 Hz, 1H), 7.50 (d, J = 7.5 Hz, 1H), 6.90 (d, J = 8.4 Hz, 1H), 6.79 (d, J = 8.2 Hz, 1H), 5.42 (s, 2H), 4.78 (dd, J = 16.8, 8.5 Hz, 4H), 4.56 (s, 2H), 3.78 (d, J = 5.2 Hz, 2H), 3.33 (s, 3H).367648.21H NMR (400 MHz, DMSO-d6) δ 9.13 (d, J = 1.5 Hz, 1H), 8.98 (s, 1H), 8.78 (d, J = 1.4 Hz, 1H), 8.45 (s, 1H), 8.26 (d, J = 1.5 Hz, 1H), 8.09-7.71 (m, 4H), 7.63 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.6, 1.7 Hz, 1H), 7.39 (dd, J = 11.5, 6.1 Hz, 1H), 7.03 (d, J = 8.3 Hz, 1H), 5.62 (s, 2H), 4.63 (t, J = 5.1 Hz, 2H), 4.48 (s, 2H), 3.69 (t, J = 5.1 Hz, 2H), 3.20 (s, 3H).377613.21H NMR (400 MHz, DMSO-d6) δ 9.19 (d, J = 1.5 Hz, 1H), 8.94 (d, J = 1.4 Hz, 1H), 8.45 (s, 1H), 8.28 (d, J = 1.5 Hz, 1H), 7.92 (t, J = 7.9 Hz, 1H), 7.86 (dd, J = 8.4, 1.5 Hz, 1H), 7.72 (dd, J = 10.5, 6.4 Hz, 1H), 7.64 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.05 (d, J = 8.3 Hz, 1H), 5.69 (s, 2H), 4.64 (t, J = 5.1 Hz, 2H), 4.49 (s, 2H), 4.31 (s, 3H), 3.69 (t, J = 5.0 Hz, 2H), 3.20 (s, 3H).387612.21H NMR (400 MHz, DMSO-d6) δ 8.97 (d, J = 1.5 Hz, 1H), 8.70 (d, J = 1.4 Hz, 1H), 8.41 (s, 1H), 8.28 (d, J = 1.5 Hz, 1H), 8.10 (s, 1H), 7.90 (t, J = 7.9 Hz, 1H), 7.86 (dd, J = 8.4, 1.5 Hz, 1H), 7.78 (dd, J = 10.5, 6.4 Hz, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.01 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 4.65 (t, J = 5.2 Hz, 2H), 4.51 (s, 2H), 3.90 (s, 3H), 3.69 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H).398574.21H NMR (400 MHz, DMSO-d6) δ 8.89 (t, J = 1.2 Hz, 1H), 8.48 (dd, J = 9.9, 1.7 Hz, 1H), 8.29 (d, J = 1.5 Hz, 1H), 7.93-7.82 (m, 2H), 7.66 (d, J = 8.5 Hz, 1H), 7.61 (dd, J = 10.6, 6.4 Hz, 1H), 7.51 (dd, J = 7.5, 1.6 Hz, 1H), 7.39 (dd, J = 11.6, 6.1 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.69 (d, J = 1.7 Hz, 2H), 4.65 (t, J = 5.1 Hz, 2H), 4.51 (s, 2H), 3.69 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H).409644.41H NMR (400 MHz, DMSO) δ 8.06 (dd, J = 43.2, 1.5 Hz, 1H), 7.95-7.87 (m, 2H), 7.84 (dt, J = 8.4, 1.9 Hz, 1H), 7.81-7.71 (m, 4H), 7.68 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.45-7.36 (m, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.83-5.62 (m, 1H), 5.62 (s, 2H), 4.63-4.39 (m, 2H), 4.32-4.06 (m, 2H), 4.02-3.86 (m, 1H), 2.09 (d, J = 13.4 Hz, 3H).4110623.41H NMR (400 MHz, Methanol-d4) δ 8.30 (d, J = 1.1 Hz, 1H), 8.10 (dd, J = 8.5, 1.5 Hz, 1H), 7.77 (dd, J = 10.6, 6.3 Hz, 1H), 7.72 (d, J = 8.8 Hz, 1H), 7.68 (d, J = 7.5 Hz, 1H), 7.63 (s, 1H), 7.55-7.44 (m, 2H), 7.17 (dd, J = 11.2, 6.0 Hz, 1H), 7.02 (s, 1H), 6.73 (t, J = 55.5 Hz, 1H), 5.62 (s, 2H), 4.57 (d, J = 6.4 Hz, 4H), 3.76 (t, J = 4.9 Hz, 2H), 3.29 (s, 3H).4210607.61H NMR (400 MHz, Methanol-d4) δ 8.33 (d, J = 1.4 Hz, 1H), 8.12 (dd, J = 8.6, 1.5 Hz, 1H), 7.80-7.65 (m, 3H), 7.58-7.48 (m, 3H), 7.19 (dd, J = 11.3,6.0 Hz, 1H), 6.94 (d, J = 1.4 Hz, 1H), 5.60 (s, 2H), 4.64- 4.56 (m, 4H), 3.77 (t, J = 5.0 Hz, 2H), 3.29 (s, 3H).4310603.51H NMR (400 MHz, DMSO-d6) δ 8.29 (d, J = 1.5 Hz, 1H), 7.97-7.89 (m, 1H),7.86 (dd, J = 8.4, 1.5 Hz, 1H), 7.81-7.70 (m, 3H), 7.65 (d, J = 8.4 Hz, 1H), 7.40 (dd, J= 11.5, 6.1 Hz, 1H), 7.08 (t, J = 1.5 Hz, 1H), 6.57 (d, J = 1.9 Hz, 1H), 5.58 (s, 2H), 4.65 (t, J = 5.1 Hz, 2H), 4.51 (s, 2H), 3.87 (s, 3H), 3.70 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H).4410589.31H NMR (400 MHz, Methanol-d4) δ 8.52 (d, J = 1.2 Hz, 1H), 8.19 (dd, J = 8.6, 1.5 Hz, 1H), 7.83-7.74 (m, 2H), 7.68 (dt, J = 12.8, 6.7 Hz, 1H), 7.63-7.52 (m, 2H), 7.30 (dd, J = 11.3, 6.1 Hz, 1H), 7.07 (t, J = 1.6 Hz, 1H), 6.29 (d, J = 1.8 Hz, 1H), 5.58 (s, 2H), 4.78 (t, J = 5.0 Hz, 2H), 4.71 (s, 2H), 3.82 (t, J = 4.9 Hz, 2H).4510589.21H NMR (400 MHz, DMSO-d6) δ 8.25 (s, 1H), 7.92 (d, J = 10.0 Hz, 1H), 7.87-7.78 (m, 2H), 7.74 (s, 2H), 7.62 (d, J = 8.5 Hz, 1H), 7.38 (dd, J = 11.7,6.0 Hz, 1H), 7.26 (s, 2H), 6.71 (d, J = 1.4 Hz, 1H), 5.50 (s, 2H), 4.61 (t, J = 5.1 Hz, 2H), 4.47 (s, 2H), 3.68 (t, J = 5.1 Hz, 2H), 3.20 (s, 3H).4611537.41H NMR (400 MHz, Methanol-d4) δ 8.74 (t, J = 1.5 Hz, 1H), 8.52 (t, J =1.0 Hz, 1H), 8.19 (dd, J = 8.6, 1.5 Hz, 1H), 8.12-8.00 (m, 2H), 7.80 (t, J = 7.7 Hz, 1H), 7.75 (dd, J = 8.6, 0.7 Hz, 1H), 7.70 (t, J = 2.1 Hz, 1H), 7.67-7.60 (m, 3H), 7.50 (t, J = 8.0 Hz, 1H), 7.21 (ddd, J = 8.3, 2.6, 0.9 Hz, 1H), 5.35 (s, 2H), 4.81 (t, J = 4.9 Hz, 2H), 4.77 (s, 2H), 3.84 (t, J = 4.9 Hz, 2H), 3.32 (s, 3H).4712695.291H NMR (400 MHz, Methanol-d4) δ 8.69 (s, 1H), 8.04 (d, J = 1.4 Hz, 1H), 7.93-7.67 (m, 3H), 7.67- 7.48 (m, 3H), 7.15 (dd, J = 11.0, 6.7 Hz, 1H), 7.00- 6.85 (m, 1H), 5.63 (s, 2H), 5.00-4.92 (m, 1H),4.64- 4.39 (m, 4H), 3.93 (d, J = 8.9 Hz, 1H), 3.78 (d, J = 8.9 Hz, 1H), 2.00 (d, J = 39.2 Hz, 1H), 1.64 (s, 6H), 1.30 (s, 3H), 0.61 (s, 3H).4812677.321H NMR (400 MHz, Methanol-d4) δ 8.67 (s, 1H), 8.01 (d, J = 1.4 Hz, 1H), 7.90-7.69 (m, 3H), 7.69- 7.47 (m, 3H), 7.19 (dd, J = 11.3, 6.0 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.63 (s, 2H), 5.00-4.90 (m, 2H), 4.56 (dd, J = 19.4, 8.0 Hz, 3H), 4.43 (dd, J = 11.4, 6.8 Hz, 1H), 3.94 (d, J = 8.9 Hz, 1H), 3.78 (d, J = 8.9 Hz, 1H), 1.61 (ddd, J = 12.8, 8.3, 5.1 Hz, 1H), 1.28 (s, 3H), 1.02-0.84 (m, 4H), 0.61 (s, 3H).4913571.31H NMR (400 MHz, Methanol-d4) δ 8.60 (t, J = 1.0 Hz, 1H), 8.28 (dd, J = 8.6, 1.4 Hz, 1H), 7.91-7.81 (m, 3H), 7.70 (t, J = 7.5 Hz, 1H), 7.63-7.55 (m, 2H), 7.48 (d, J = 7.5 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.56 (s, 2H), 4.87-4.82 (m, 4H), 3.96-3.71 (m, 2H), 3.30 (s, 3H).5014595.31H NMR (400 MHz, Methanol-d4) δ 8.57 (d, J = 9.5 Hz, 1H), 8.35 (d, J = 1.4 Hz, 1H), 8.18 (s, 2H), 8.07 (dd, J = 8.4, 7.5 Hz, 1H), 7.96 (dd, J = 8.5, 1.5 Hz, 1H), 7.85 (d, J = 7.4 Hz, 1H), 7.79 (t, J = 7.6 Hz, 1H), 7.66-7.55 (m, 2H), 7.50 (d, J = 8.5 Hz, 1H), 7.19 (d, J = 8.3 Hz, 1H), 5.73 (s, 2H), 5.22 (dd, J = 6.9, 2.1 Hz,1H), 4.78 (t, J = 4.8 Hz, 2H),3.96-3.83 (m, 2H), 3.41 (s,3H).5115591.21H NMR (400 MHz, Methanol-d4) δ 8.38 (d, J = 1.4 Hz, 1H), 8.02 (dd, J = 8.6, 1.5 Hz, 1H), 7.88-7.63 (m, 4H), 7.63-7.48 (m, 4H), 7.41 (d, J = 45.5 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.79- 4.62 (m, 2H), 3.89-3.67 (m, 2H), 3.25 (s, 3H). 19F NMR (376 MHz, Methanol- d4) δ −78.14, −117.82 (dd, J =9.6, 7.2 Hz), −121.92 (ddd,J = 17.8, 11.2, 6.2 Hz),−124.47 (ddd, J = 17.3, 11.0,5.8 Hz), −179.57 (d, J = 45.4Hz).5215609.31H NMR (400 MHz, Methanol-d4) δ 8.47-8.20 (m, 1H), 8.16-7.83 (m, 1H), 7.83-7.71 (m, 2H), 7.71-7.33 (m, 6H), 6.99- 6.80 (m, 1H), 5.60-5.35 (m, 2H), 4.71-4.56 (m, 2H), 3.86-3.66 (m, 2H), 3.22 (s, 3H). 19F NMR (376 MHz, Methanol-d4) δ −78.33 (d, J = 11.0 Hz), −91.17 (t, J = 11.0 Hz), −117.83 (d, J = 9.4 Hz), −120.07, −121.85.5316596.71H NMR (400 MHz, Methanol-d4) δ 8.78 (d, J = 7.5 Hz, 1H), 8.59 (d, J = 10.5 Hz, 1H), 8.53 (d, J = 6.9 Hz, 1H), 8.48 (s, 1H), 8.22-8.08 (m, 1H), 7.94 (t, J = 7.9 Hz, 1H), 7.78 (t, J = 7.6 Hz, 1H), 7.74-7.55 (m, 3H), 7.05 (d, J = 8.3 Hz, 1H), 5.72 (s, 2H), 5.37- 5.25 (m, 2H), 4.92 (t, J = 4.9 Hz, 2H), 3.89 (t, J = 4.9 Hz, 2H), 3.28 (s, 3H).5417603.31H NMR (400 MHz, Methanol-d4) δ 8.49 (t, J = 1.0 Hz, 1H), 8.20 (dd, J = 8.6, 1.4 Hz, 1H), 7.86 (d, J = 8.6 Hz, 1H), 7.84-7.75 (m, 2H), 7.72 (t, J = 7.5 Hz, 1H), 7.66-7.50 (m, 4H), 7.42 (dd, J = 11.8, 6.0 Hz, 1H), 6.95 (d, J = 8.2 Hz, 1H), 5.62 (s, 2H), 5.25 (t, J = 6.1 Hz, 1H), 4.82-4.73 (m, 1H), 4.67 (ddd, J = 15.3, 7.2, 3.5 Hz, 1H), 4.40 (dd, J =11.0, 6.5 Hz, 1H), 4.32 (dd,J = 11.0, 5.7 Hz, 1H), 3.85-3.66 (m, 2H), 3.21 (s, 3H).5518651.31H NMR (400 MHz, Acetonitrile-d3) δ 8.32 (d, J = 1.4 Hz, 1H), 8.21 (d, J = 1.4 Hz, 1H), 7.99 (s, 2H), 7.88-7.77 (m, 2H), 7.73 (t, J = 7.5 Hz, 1H), 7.64-7.51 (m, 3H), 7.21 (dd, J = 11.7, 6.1 Hz, 1H), 6.92 (d, J = 8.3 Hz, 1H), 5.63 (s, 2H), 4.56 (t, J = 5.0 Hz, 2H), 4.49 (s, 2H), 3.74 (t, J = 5.0 Hz, 2H), 3.25 (s, 3H).5618752.21H NMR (400 MHz, Acetonitrile-d3) δ 8.53 (s, 1H), 8.33 (d, J = 0.7 Hz, 1H), 8.16 (d, J = 1.5 Hz, 1H), 8.12 (d, J = 1.5 Hz, 1H), 7.88-7.76 (m, 2H), 7.73 (t, J = 7.6 Hz, 1H), 7.64- 7.50 (m, 3H), 7.24 (dd, J = 11.7, 6.1 Hz, 1H), 6.92 (dd, J = 8.3, 0.6 Hz, 1H), 5.63 (s, 2H), 4.61 (t, J = 5.8 Hz, 2H), 4.55-4.43 (m, 4H), 3.88 (s, 8H), 3.72 (t, J = 5.0 Hz, 2H), 3.63 (t, J = 5.8 Hz, 2H), 3.23 (s, 3H).5719669.61H NMR (400 MHz, DMSO-d6) δ 8.69 (d, J = 10.4 Hz, 1H), 8.51 (s, 1H), 8.35 (s, 1H), 8.06 (s, 1H), 8.00-7.86 (m, 2H), 7.83 (dd, J = 8.4, 1.5 Hz, 1H), 7.73-7.61 (m, 2H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.48 (dd, J = 11.2, 6.3 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.55 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.65-4.51 (m, 2H), 4.49-4.36 (m, 2H), 3.89 (s, 3H), 3.79 (d, J = 8.7 Hz, 1H), 3.74 (d, J = 8.6 Hz, 1H), 1.34 (s, 3H), 0.62 (s, 3H).5819669.61H NMR (400 MHz, DMSO-d6) δ 8.69 (d, J = 10.4 Hz, 1H), 8.51 (s, 1H), 8.35 (s, 1H), 8.06 (s, 1H), 8.00-7.86 (m, 2H), 7.83 (dd, J = 8.4, 1.5 Hz, 1H), 7.73-7.61 (m, 2H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.48 (dd, J = 11.2, 6.3 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.55 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.65-4.51 (m, 2H), 4.49-4.36 (m, 2H), 3.89 (s, 3H), 3.79 (d, J = 8.7 Hz, 1H), 3.74 (d, J = 8.6 Hz, 1H), 1.34 (s, 3H), 0.62 (s, 3H).5920613.21H NMR (400 MHz, DMSO-d6) δ 12.77 (s, 1H), 8.48 (s, 1H), 7.96-7.86 (m, 2H), 7.83-7.70 (m, 4H), 7.61 (d, J = 8.5 Hz, 1H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.45 (dd, J = 11.3, 6.2 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.58-4.47 (m, 2H), 4.44 (dd, J = 11.1, 6.8 Hz, 1H), 4.36 (d, J = 16.9 Hz, 1H), 3.82-3.70 (m, 2H), 1.33 (s, 3H), 1.24 (s, 1H), 0.60 (s, 3H).6020613.21H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 7.96-7.85 (m, 2H), 7.85- 7.68 (m, 4H), 7.63 (d, J = 8.5 Hz, 1H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.46 (dd, J = 11.3, 6.2 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.02 (d, J = 6.6 Hz, 1H), 4.58-4.49 (m, 2H), 4.49- 4.34 (m, 2H), 3.82-3.70 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H).6121614.11H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 9.32 (s, 1H), 8.25-8.16 (m, 2H), 7.94-7.77 (m, 5H), 7.73-7.64 (m, 2H), 7.60 (d, 1H), 7.54 (dd, 1H), 7.40 (dd, 1H), 6.97 (d, 1H), 5.60 (s, 2H), 4.61 (t, 2H), 4.46 (s, 2H), 3.69 (t, 2H), 3.22 (s, 3H).6222(M + H+) 581.21H NMR (400 MHz, DMSO-d6) δ 7.94 (s, 1H), 7.92-7.87 (m, 2H), 7.80- 7.65 (m, 4H), 7.61 (d, J = 8.5 Hz, 1H), 7.53 (d, J = 7.3 Hz, 1H), 7.44 (dd, J = 11.5, 6.0 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.34 (s, 2H), 3.91 (dd, J = 7.2, 4.0 Hz, 1H), 1.94-1.77 (m, 2H), 1.36 (s, 1H), 1.08 (td, J = 9.2, 4.6 Hz, 2H), 0.90 (m, 1H).6322(M + H+) 609.21H NMR (400 MHz, DMSO-d6) δ 8.24 (d, J = 1.4 Hz, 1H), 8.00-7.82 (m, 2H), 7.82-7.67 (m, 4H), 7.62 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.31 (dd, J = 11.4, 6.1 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.97 (t, J = 8.4 Hz, 1H), 4.38 (s, 2H), 2.26 (ddt, J = 30.5, 18.5, 6.2 Hz, 3H), 2.04 (d, J = 7.1 Hz, 1H), 1.87 (dq, J = 11.9, 6.6,5.8 Hz, 1H), 1.77-1.57 (m,1H), 0.83-0.61 (m, 1H),0.55 (dt, J = 9.3, 5.1 Hz,1H), 0.48 (dd, J = 9.2, 3.2Hz, 1H), −0.09 (dd, J = 10.0,4.9 Hz, 1H).6422(M + H+) 595.11H NMR (400 MHz, DMSO-d6) δ 12.84 (s, 1H), 8.50 (d, J = 1.5 Hz, 1H), 7.99-7.84 (m, 2H), 7.82 (dd, J = 8.4, 1.5 Hz, 1H), 7.78-7.70 (m, 3H), 7.64 (d, J = 8.2 Hz, 1H), 7.51 (dd, J = 7.5, 1.7 Hz, 1H), 7.22 (dd, J = 11.5, 6.0 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.37 (dd, J = 9.1, 7.0 Hz, 1H), 4.40 (s, 2H), 2.99- 2.78 (m, 1H), 2.76-2.52(m, 1H), 2.45-2.15 (m,2H), 0.59 (td, J = 7.4, 6.7,4.3 Hz, 2H), 0.44-0.22 (m,1H), 0.22-0.05 (m, 1H).6522599.21H NMR (400 MHz, DMSO-d6) δ 8.45 (d, J = 1.4 Hz, 1H), 7.95-7.86 (m, 2H), 7.82 (dd, J = 8.5, 1.5 Hz, 1H), 7.79-7.69 (m, 3H), 7.63 (d, J = 8.5 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.34 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.48- 5.32 (m, 1H), 4.59-4.42 (m, 2H), 4.30 (dd, J = 5.9,3.2 Hz, 1H), 3.22-3.07 (m,1H), 2.95 (s, 3H), 2.63 (pd,J = 9.7, 8.8, 3.3 Hz, 1H), 2.24(ddd, J = 14.2, 9.1, 5.5 Hz,1H), 2.12 (t, J = 11.7 Hz,1H).6622597.21H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 1.4 Hz, 1H), 7.94-7.88 (m, 2H), 7.84-7.71 (m, 4H), 7.64 (d, J = 8.4 Hz, 1H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.41 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 4.77 (s, 1H), 4.40 (s, 2H), 3.94 (s, 2H), 2.61 (t, J = 1.7 Hz, 4H).6722584.21H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J = 1.4 Hz, 1H), 7.94-7.86 (m, 2H), 7.84 (dd, J = 8.5, 1.5 Hz, 1H), 7.78-7.71 (m, 3H), 7.69 (d, J = 8.5 Hz, 1H), 7.52 (dd, J = 7.6, 1.6 Hz, 1H), 7.38 (dd, J = 11.6, 6.0 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.59 (s, 2H), 4.39 (s, 2H), 3.38-3.25 (m, 4H), 2.13-1.94 (m, 4H).6822611.21H NMR (400 MHz, DMSO-d6) δ 8.68 (d, J = 1.4 Hz, 1H), 7.96-7.86 (m, 2H), 7.82 (dd, J = 8.5, 1.5 Hz, 1H), 7.80-7.70 (m, 3H), 7.63 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.44 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.50- 5.36 (m, 1H), 4.73-4.61 (m, 2H), 4.58-4.37 (m, 3H), 3.07 (p, J = 5.6 Hz,1H), 2.36 (ddd, J = 9.6, 5.7,3.5 Hz, 1H), 2.27 (t, J = 10.0Hz, 1H), 2.09 (q, J = 5.3, 4.3Hz, 1H), 2.04-1.96 (m,1H).6922597.21H NMR (400 MHz, DMSO-d6) δ 7.95-7.88 (m, 2H), 7.83 (dd, J = 8.4, 1.5 Hz, 1H), 7.82-7.71 (m, 4H), 7.65 (d, J = 8.6 Hz, 1H), 7.58-7.52 (m, 1H), 7.46 (s, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 4.49 (d, J = 7.5 Hz, 2H),4.21 (dd, J = 8.9, 2.8 Hz, 1H), 4.03- 3.91 (m, 1H), 3.79-3.70 (m, 3H), 1.83-1.54 (m, 1H), 1.42 (s, 1H).7022629.31H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.91 (t, J = 9.9 Hz, 2H), 7.85- 7.68 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.40 (dd, J = 11.4, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 4.87 (dd, J = 9.0, 4.7 Hz, 1H), 4.63 (d, J = 16.9 Hz, 1H), 4.59-4.46 (m, 2H), 3.90 (dd, J = 10.7, 4.6 Hz, 1H), 3.84-3.77 (m, 2H), 3.14 (s, 3H), 1.99 (q, J = 4.6, 4.0 Hz, 3H).7122611.21H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 7.96-7.87 (m, 2H), 7.83 (d, J = 8.5 Hz, 1H), 7.81-7.70 (m, 3H), 7.63 (d, J = 8.4 Hz, 1H), 7.59-7.48 (m, 1H), 7.41 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.07 (dd, J = 11.5, 5.6 Hz, 1H), 4.54 (d, J = 12.8 Hz, 3H), 4.06 (d, J = 7.1 Hz, 1H), 3.95-3.83 (m, 1H), 2.81 (t, J = 3.1 Hz,1H), 2.40 (t, J = 13.9 Hz,1H), 2.18-1.95 (m, 2H),1.83 (d, J = 10.4 Hz, 1H).7222700.21H NMR (400 MHz, DMSO-d6) δ 8.44 (s, 1H), 7.94-7.86 (m, 2H), 7.81- 7.70 (m, 4H), 7.54 (dd, J = 11.0, 7.7 Hz, 2H), 7.30 (dd, J = 11.2, 6.4 Hz, 1H), 7.09 (s, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.62 (s, 2H), 5.40 (s, 1H), 4.63-4.43 (m, 4H), 4.30 (d, J = 17.0 Hz, 1H), 4.20 (dd, J = 10.9, 7.0 Hz, 1H), 4.08-3.89 (m, 3H), 1.12 (s, 9H).7322599.21H NMR (400 MHz, DMSO-d6) δ 8.32 (d, J = 1.4 Hz, 1H), 7.94-7.87 (m, 2H), 7.83 (dd, J = 8.5, 1.5 Hz, 1H), 7.80-7.71 (m, 3H), 7.65 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.5 Hz, 1H), 7.43 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 4.54 (d, J = 5.8 Hz, 4H), 4.38-4.32 (m, 2H), 4.00 (p, J = 6.2 Hz, 1H), 3.73 (q, J = 9.0 Hz, 1H), 0.80 (d, J = 6.3 Hz, 3H).7422613.21H NMR (400 MHz, DMSO-d6) δ 8.25 (d, J = 1.5 Hz, 1H), 7.96-7.87 (m, 2H), 7.82-7.69 (m, 4H), 7.63 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 6.7 Hz, 1H), 7.43 (dd, J = 11.4, 6.4 Hz, 2H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.01 (s, 2H), 4.48 (t, J = 14.0 Hz, 3H), 4.29 (d, J = 3.5 Hz, 2H), 3.99 (d, J = 8.4 Hz, 2H), 1.29 (s, 3H), 0.77 (s, 3H).7522642.21H NMR (400 MHz, DMSO-d6) δ 8.37 (d, J = 1.4 Hz, 1H), 7.99 (q, J = 4.6 Hz, 1H), 7.96-7.82 (m, 3H), 7.82-7.71 (m, 3H), 7.68 (d, J = 8.4 Hz, 1H), 7.52 (dd, J = 7.6, 1.7 Hz, 1H), 7.23 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.71 (td, J = 7.1, 3.3 Hz, 1H), 5.60 (s, 2H), 4.59-4.48 (m, 2H), 4.48-4.42 (m, 1H), 4.27 (dd, J = 10.3, 3.4 Hz, 1H), 4.13 (dd, J =10.5, 8.3 Hz, 1H), 3.65 (t, J = 9.4 Hz, 1H), 3.45 (ddd, J = 9.9, 8.0, 6.2 Hz, 1H), 2.50 (s, 3H).7622656.21H NMR (400 MHz, DMSO-d6) δ 8.53 (d, J = 1.5 Hz, 0H), 8.46 (d, J = 1.5 Hz, 1H), 7.97-7.83 (m, 5H), 7.83-7.63 (m, 7H), 7.61- 7.46 (m, 2H), 7.37 (dd, J = 11.5, 6.1 Hz, 0H), 7.21 (dd, J = 11.6, 6.1Hz, 1H), 7.00 (dd, J = 8.3, 2.0 Hz, 2H), 5.87 (d, J = 8.6 Hz, 0H), 5.82 (s, 1H), 5.61 (d, J = 2.3 Hz, 3H), 4.69 (t, J = 8.6 Hz, 1H), 4.61-4.42 (m, 3H), 4.40-4.31 (m, 2H), 4.31- 4.20 (m, 1H), 4.20-4.05(m, 2H), 3.77 (td, J = 8.8,5.6 Hz, 1H), 3.57 (t, J = 9.1Hz, 1H), 3.02 (s, 1H), 2.78(s, 3H), 2.72 (s, 3H).7722603.21H NMR (400 MHz, DMSO-d6) δ 8.29 (d, J = 1.4 Hz, 1H), 7.96-7.89 (m, 2H), 7.89-7.82 (m, 1H), 7.82-7.71 (m, 3H), 7.68 (d, J = 8.5 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.41 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.66 (dq, J = 5.0, 2.3 Hz, 1H), 5.60 (s, 2H), 5.54 (q, J = 5.3, 3.7 Hz, 1H), 4.55 (q, J = 16.8 Hz, 2H), 4.46-4.24 (m, 3H), 4.08 (ddd, J = 24.6, 11.4,3.1 Hz, 1H).7822613.21H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.96-7.87 (m, 2H), 7.86- 7.71 (m, 5H),7.63 (d, J = 8.5 Hz, 1H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.46 (dd, J = 11.3, 6.2 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.60-4.49 (m, 2H), 4.49-4.33 (m, 2H), 3.83- 3.68 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H).″7922627.21H NMR (400 MHz, DMSO-d6) δ 8.47 (d, J = 1.4 Hz, 1H), 7.96-7.85 (m, 3H), 7.85-7.65 (m, 5H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.25 (td, J = 5.2, 2.6 Hz, 1H), 5.09 (dd, J = 7.0, 4.1 Hz, 1H), 4.60 (s, 2H), 4.23 (d, J = 5.1 Hz, 2H), 3.96 (dd, J = 12.2, 9.2 Hz, 2H), 3.58-3.48 (m, 2H), 3.18 (ddt, J = 6.9, 4.5, 2.3 Hz, 1H).8022626.21H NMR (400 MHz, DMSO-d6) δ 8.50 (d, J = 1.4 Hz, 1H), 7.98-7.87 (m, 2H), 7.84 (dd, J = 8.4, 1.5 Hz, 1H), 7.81-7.70 (m, 3H), 7.66 (d, J = 8.5 Hz, 1H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.43 (dd, J = 11.5, 6.1 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.61 (s, 3H), 4.86 (dd, J = 11.1, 2.3 Hz, 1H), 4.76 (td, J = 6.0, 2.5 Hz, 1H), 4.55-4.36 (m, 2H), 4.23 (dd, J = 11.1, 7.3 Hz, 1H), 3.71-3.48 (m, 2H), 3.46-3.29 (m, 1H), 3.04 (s, 1H), 2.46-2.29 (m, 2H).8122633.21H NMR (400 MHz, DMSO-d6) δ 8.41 (d, J = 1.3 Hz, 1H), 7.94-7.88 (m, 2H), 7.86 (dd, J = 8.5, 1.4 Hz, 1H), 7.81-7.71 (m, 3H), 7.68 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.4, 1.6 Hz, 1H), 7.38 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.73 (ddd, J = 18.8, 10.9, 7.8 Hz, 1H), 5.61 (s, 2H), 4.53 (s, 2H), 3.78 (dd, J = 14.3, 10.6 Hz, 1H), 3.70- 3.53 (m, 2H), 3.32 (td, J = 13.1, 6.8 Hz, 1H), 2.86- 2.60 (m, 2H).8222625.21H NMR (400 MHz, DMSO-d6) δ 8.28 (d, J = 1.4 Hz, 1H), 7.96-7.86 (m, 2H), 7.81-7.70 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.6, 1.7 Hz, 1H), 7.48 (dd, J = 11.4, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.27- 5.19 (m, 1H), 4.55 (d, J = 16.8 Hz, 1H), 4.37 (d, J = 16.8 Hz, 1H), 4.22 (d, J = 11.5 Hz, 1H), 4.00 (dd, J = 11.8, 2.1 Hz, 1H), 3.79-3.68 (m, 1H), 3.19 (d, J =11.7 Hz, 1H), 2.89 (td, J =12.5, 11.5, 4.5 Hz, 1H), 2.05(d, J = 11.6 Hz, 1H), 0.61(dt, J = 10.1, 5.2 Hz, 1H),0.40 (s, 1H), 0.22 (dt, J =10.2, 5.3 Hz, 1H).8322627.21H NMR (400 MHz, DMSO-d6) δ 8.30 (s, 1H), 7.96-7.86 (m, 2H), 7.86- 7.70 (m, 5H), 7.61 (d, J = 8.4 Hz, 1H), 7.55 (dd, J = 7.5, 1.7 Hz, 1H), 7.46 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 4.75 (dd, J = 12.6, 3.9 Hz, 1H), 4.63 (d, J = 16.9 Hz, 1H), 4.42 (d, J = 16.9 Hz, 1H), 4.19-4.11 (m, 1H), 3.63-3.53 (m, 2H), 3.47 (d, J = 11.4 Hz, 1H),2.91 (dt, J = 12.7, 5.8 Hz,1H), 1.75 (d, J = 10.8 Hz,1H), 1.20 (s, 3H), 0.91 (s,3H).8423650.31H NMR (400 MHz, Acetonitrile-d3) δ 8.83 (s, 1H), 8.75 (d, J = 6.4 Hz, 2H), 8.54 (s, 1H), 8.43 (s, 1H), 7.90-7.75 (m, 3H), 7.72 (t, J = 7.4 Hz, 1H), 7.57 (t, J = 8.1 Hz, 3H), 7.26 (dd, J = 11.6, 6.0 Hz, 1H), 6.94 (d, J = 8.3 Hz, 1H), 5.63 (s, 2H), 4.77-4.41 (m, 4H), 3.75 (t, J = 5.0 Hz, 2H), 3.23 (s, 3H).8524672.21H NMR (400 MHz, DMSO-d6) δ 8.26 (d, J = 5.2 Hz, 1H), 7.99-7.87 (m, 2H), 7.83-7.71 (m, 4H), 7.60 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.34 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.49 (d, J = 5.7 Hz, 1H), 4.62-4.43 (m, 2H), 4.15 (d, J = 8.8 Hz, 2H), 3.88 (t, J = 9.9 Hz, 1H), 3.69 (d, J = 4.8 Hz, 3H), 3.10 (s, 3H) (note: 3 protons hidden by solvent).8624672.21H NMR (400 MHz, DMSO-d6) δ 8.12 (s, 1H), 7.99-7.87 (m, 2H), 7.84 (dd, J = 8.4, 1.4 Hz, 1H), 7.79-7.72 (m, 3H), 7.67 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.38 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.33 (s, 1H), 4.63- 4.37 (m, 3H), 3.68 (s, 4H), 3.36 (d, J = 9.4 Hz, 1H), 3.17 (s, 3H).8724672.21H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J = 5.9 Hz, 1H), 7.97-7.87 (m, 2H), 7.84-7.58 (m, 4H), 7.54 (dd, J = 7.6, 1.7 Hz, 1H), 7.35 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.55- 5.42 (m, 1H), 4.62-4.43 (m, 2H), 4.16 (d, J = 9.1 Hz, 2H), 3.88 (t, J = 9.9 Hz, 1H), 3.10 (s, 3H).8825692.21H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J = 1.5 Hz, 1H), 7.96-7.83 (m, 3H), 7.80-7.73 (m, 3H), 7.68 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.4, 1.7 Hz, 1H), 7.38 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.34 (dt, J = 9.6, 6.6 Hz, 1H), 4.61- 4.38 (m, 3H), 4.06-3.92 (m, 2H), 3.30 (dd, J = 10.1, 6.9 Hz, 1H), 3.18 (s, 3H), 3.13 (s, 3H).8925692.21H NMR (400 MHz, DMSO-d6) δ 8.41 (s, 1H), 7.97-7.87 (m, 2H), 7.82- 7.71 (m, 4H),7.60 (d, J = 8.4 Hz, 1H), 7.56-7.52 (m, 1H), 7.34 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.53 (td, J = 8.6, 5.1 Hz, 1H), 4.62- 4.46 (m, 2H), 4.19 (q, J = 4.6, 3.7 Hz, 1H), 4.05 (dd, J = 10.3, 8.2 Hz, 1H), 3.84 (t, J = 9.7 Hz, 1H), 3.10 (s, 3H), 3.08 (s, 3H).9026669.61H NMR (400 MHz, DMSO-d6) δ 8.69 (d, J = 10.4 Hz, 1H), 8.51 (s, 1H), 8.35 (s, 1H), 8.06 (s, 1H), 8.00-7.86 (m, 2H), 7.83 (dd, J = 8.4, 1.5 Hz, 1H), 7.73-7.61 (m, 2H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.48 (dd, J = 11.2, 6.3 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.55 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.65-4.51 (m, 2H), 4.49-4.36 (m, 2H), 3.89 (s, 3H), 3.79 (d, J = 8.7 Hz, 1H), 3.74 (d, J = 8.6 Hz, 1H), 1.34 (s, 3H), 0.62 (s, 3H).9126655.31H NMR (400 MHz, DMSO-d6) δ 8.65 (s, 1H), 8.60 (s, 1H), 8.32 (s, 1H), 8.27 (d, J = 1.5 Hz, 1H), 7.96-7.79 (m, 3H), 7.75 (s, 1H), 7.62 (d, J = 8.4 Hz, 1H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.42 (dd, J = 11.5, 6.1 Hz, 1H), 6.97 (d, J = 8.3 Hz, 1H), 5.50 (s, 2H), 4.54 (dd, J = 15.2, 3.2 Hz, 1H), 4.49 (s, 2H), 4.38 (dd, J = 15.2, 8.8 Hz, 1H), 4.16 (s, 3H), 3.97 (s, 3H), 3.71 (ddd, J = 9.3, 6.2, 3.2 Hz, 1H), 3.09 (s, 3H), 1.24 (d, J = 6.1 Hz, 3H).9227605.861H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.51 (d, J = 2.4 Hz, 1H), 8.17 (dd, J = 8.6, 1.3 Hz, 1H), 8.05-7.88 (m, 2H), 7.88-7.72 (m, 2H), 7.58 (dd, J = 7.5, 1.6 Hz, 1H), 7.48 (d, J = 8.2 Hz, 1H), 7.40 (dd, J = 11.1, 6.1 Hz, 1H), 6.94 (d, J = 8.3 Hz, 1H), 5.55 (s, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.78-4.61 (m, 3H), 4.52 (dd, J = 11.6,6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.84 (d, J = 8.9 Hz,1H), 1.41 (s, 3H), 0.76 (s,3H).9327606.771H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.66 (d, J = 1.4 Hz, 1H), 8.59 (d, J = 1.3 Hz, 1H), 8.20 (d, J = 8.6 Hz, 1H), 7.94-7.72 (m, 3H), 7.60 (dd, J = 7.5, 1.6 Hz, 1H), 7.41 (dd, J = 11.2, 6.0 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.64 (s, 2H), 5.15 (d, J = 6.6 Hz, 1H), 4.79-4.59 (m, 3H), 4.53 (dd, J = 11.7, 6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.85 (d, J = 8.9 Hz,1H), 1.41 (s, 3H), 0.76 (s,3H).9428582.21H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.93 (d, J = 1.4 Hz, 1H), 8.25 (d, J = 1.5 Hz, 1H), 7.92 (t, J = 7.9 Hz, 1H), 7.84 (dd, J = 8.4, 1.5 Hz, 1H), 7.68-7.59 (m, 2H), 7.53 (dd, J = 7.6, 1.6 Hz, 1H), 7.38 (dd, J = 11.5, 6.1Hz, 1H), 7.29-6.93 (m, 2H), 5.71 (s, 2H), 4.62 (t, J = 5.1 Hz, 2H), 4.47 (s, 2H), 3.69 (t, J = 5.1 Hz, 2H), 3.21 (s, 3H).9528(M + H+) 597.21H NMR (400 MHz, DMSO-d6) δ 8.65 (d, J = 2.2 Hz, 1H), 8.55 (s, 1H), 8.20 (d, J = 1.4 Hz, 1H), 8.14 (dd, J = 8.4, 2.3 Hz, 1H), 7.97 (s, 1H), 7.92-7.83 (m, 3H), 7.79 (dd, J = 8.4, 1.5 Hz, 1H), 7.60 (d, J = 8.5 Hz, 1H), 7.53 (d, J = 7.1 Hz, 1H), 7.39 (dd, J = 11.5, 6.1 Hz, 1H), 7.13 (s, 1H), 6.97 (d, J = 8.3 Hz, 1H), 5.56 (s, 2H), 5.40 (s, 1H), 4.59 (d, J = 5.6 Hz, 2H), 4.44 (s, 2H), 3.69 (t, J = 5.0 Hz, 2H), 3.22 (s, 3H).9628594.41H NMR (400 MHz, DMSO-d6) δ 12.84 (s, 1H), 8.32 (d, 1H), 7.93-7.84 (m, 2H), 7.79 (dd, 1H), 7.70- 7.59 (m, 1H), 7.59 (d, 4H), 7.51 (dd, 1H), 7.43 (dd, 1H), 6.97 (dd, 1H), 5.53 (s, 2H), 4.67 (t, 2H), 3.70 (t, 2H), 3.21 (s, 3H), 2.49 (s, 0H), 1.96 (t, 3H).9729579.21H NMR (400 MHz, DMSO) δ 8.26 (s, 1H), 7.98- 7.92 (m, 1H), 7.91-7.81 (m, 3H), 7.78-7.68 (m, 2H), 7.65 (d, J = 8.4 Hz, 1H), 7.59 (d, J = 7.6 Hz, 1H), 7.27 (d, J = 8.0 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 5.36 (s, 2H), 4.99 (s, 2H), 4.58 (t, J = 5.3 Hz, 2H), 4.42 (s, 2H), 3.63 (t, J = 5.1 Hz, 2H), 3.20 (s, 3H).9830760.41H NMR (400 MHz, DMSO-d6) δ 8.25 (d, 1H), 7.96 (s, 1H), 7.93-7.79 (m, 2H), 7.63 (d, 1H), 7.59- 7.35 (m, 4H), 6.94 (d, 1H), 5.50 (s, 1H), 4.63 (d, 0H), 4.62 (s, 1H), 4.48 (s, 1H), 4.26 (d, 1H), 3.79 (s, 1H), 3.48 (dt, 5H), 3.36 (q, 2H), 3.19 (dd, 4H) (note: multiple protons hidden under water peak).9930629.11H NMR (400 MHz, DMSO-d6) δ 8.29 (d, 1H), 7.99 (s, 1H), 7.92-7.82 (m, 3H), 7.73 (t, 1H), 7.65 (d, 1H), 7.62 (dd, 1H), 7.53 (dd, 1H), 7.50 (dd, 1H), 7.42 (dd, 1H), 6.98 (dd, 1H), 5.59 (s, 2H), 4.65 (t, 2H), 4.52 (s, 2H), 4.11 (s, 3H), 3.70 (t, 2H), 3.22 (s, 3H).10030664.21H NMR (400 MHz, DMSO-d6) δ 12.87 (s, 1H), 8.82 (s, 1H), 8.35 (s, 1H), 8.29 (d, 1H), 7.95-7.82 (m, 4H), 7.72-7.54 (m, 5H), 7.52 (dd, 1H), 7.42 (dd, 1H), 6.96 (d, 1H), 5.52 (s, 2H), 4.65 (t, 2H), 4.52 (s, 2H), 3.70 (t, 2H), 3.22 (s, 3H).10130628.31H NMR (400 MHz, DMSO-d6) δ 12.86 (s, 1H), 8.29 (d, 1H), 7.94-7.81 (m, 3H), 7.73-7.62 (m, 2H), 7.57-7.45 (m, 3H), 7.47- 7.38 (m, 2H), 6.98 (d, 1H), 6.49 (d, 1H), 5.58 (s, 2H), 4.66 (t, 2H), 4.52 (s, 2H), 3.88 (s, 3H), 3.70 (t, 2H), 3.22 (s, 3H).10230696.21H NMR (400 MHz, DMSO-d6) δ 8.27 (d, 1H), 7.94-7.80 (m, 3H), 7.73 (t, 1H), 7.67-7.37 (m, 5H), 7.03-6.95 (m, 2H), 5.59 (s, 2H), 4.64 (t, 2H), 4.50 (s, 2H), 3.70 (t, 2H), 3.21 (s, 3H).10330625.21H NMR (400 MHz, DMSO-d6) δ 9.05 (d, 1H), 8.69 (dd, 1H), 8.35 (dt, 1H), 8.27 (d, 1H), 7.94-7.80 (m, 3H), 7.80-7.60 (m, 5H), 7.53 (dd, 1H), 7.42 (dd, 1H), 6.98 (d, 1H), 5.59 (s, 2H), 4.64 (t, 2H), 4.50 (s, 2H), 3.70 (t, 2H), 3.21 (s, 3H).10430639.31H NMR (400 MHz, DMSO-d6) δ 8.74 (dd, 1H), 8.27 (d, 1H), 8.20 (d, 1H), 7.95-7.80 (m, 3H), 7.75 (dt, 2H), 7.63 (d, 1H), 7.57- 7.32 (m, 4H), 6.99 (d, 1H), 5.60 (s, 2H), 4.64 (t, 2H), 4.50 (s, 2H), 3.70 (t, 2H), 3.22 (s, 3H), 2.59 (s, 3H).10530639.31H NMR (400 MHz, DMSO-d6) δ 12.88 (s, 1H), 9.03 (d, 1H), 8.51 (dd, 1H), 8.27 (d, 1H), 7.94-7.65 (m, 7H), 7.63 (d, 1H), 7.53 (dd, 1H), 7.42 (dd, 1H), 6.97 (d, 1H), 5.59 (s, 2H), 4.64 (t, 2H), 3.70 (t, 2H), 3.21 (s, 3H), 2.66 (s, 3H).10630672.21H NMR (400 MHz, DMSO-d6) δ 8.28-8.21 (m, 4H), 7.96 (s, 2H), 7.92- 7.79 (m, 6H), 7.63 (d, 2H), 7.57-7.47 (m, 7H), 7.45 (d, 1H), 7.43 (s, 1H), 6.94 (d, 2H), 5.49 (s, 4H), 4.63 (t, 4H), 4.49 (s, 4H), 4.27 (t, 4H), 3.70 (dt, 9H), 3.24 (s, 6H), 3.22 (s, 6H).10730612.11H NMR (400 MHz, Acetonitrile-d3) δ 8.85 (d, 1H), 8.34 (s, 1H), 8.09- 7.98 (m, 3H), 7.93-7.70 (m, 4H), 7.56 (dd, 1H), 7.27 (dd, 1H), 6.96-6.89 (m, 1H), 5.60 (s, 2H), 4.57 (dd, 4H), 4.35 (s, 3H), 3.75 (t, 2H), 3.26 (s, 3H).10830611.21H NMR (400 MHz, Acetonitrile-d3) δ 8.80 (d, 1H), 8.43-8.30 (m, 3H), 8.16 (d, 1H), 8.08 (dd, 1H), 7.98-7.77 (m, 3H), 7.76 (d, 1H), 7.54 (dd, 1H), 7.29 (dd, 1H), 6.92 (d, 1H), 5.60 (s, 2H), 4.65 (d, 1H), 4.63 (s, 3H), 3.95 (s, 3H), 3.78 (t, 2H), 3.28 (s, 3H).10930647.21H NMR (400 MHz, Acetonitrile-d3) δ 8.79 (d, 1H), 8.63 (s, 1H), 8.45- 8.40 (m, 1H), 8.31 (s, 1H), 8.11 (ddd, 2H), 7.97-7.87 (m, 1H), 7.87-7.76 (m, 3H), 7.62-7.52 (m, 1H), 7.45 (s, 0H), 7.35-7.26 (m, 1H), 6.92 (d, 1H), 5.59 (s, 2H), 4.65 (t, 2H), 3.78 (t, 2H), 3.27 (s, 3H), 1.97- 1.86 (m, 1H).11030611.21H NMR (400 MHz, Acetonitrile-d3) δ 8.81 (d, 1H), 8.39 (s, 1H), 8.08 (dd, 1H), 7.97 (dd, 1H), 7.91 (dd, 1H), 7.85-7.75 (m, 2H), 7.72 (d, 1H), 7.56 (dd, 1H), 7.47 (d, 1H), 7.30 (dd, 1H), 6.93 (d, 1H), 6.69 (d, 1H), 5.58 (s, 2H), 4.62 (s, 3H), 4.62 (d, 1H), 4.16 (s, 3H), 3.76 (t, 2H), 3.26 (s, 3H).11131572.41H NMR (400 MHz, DMSO-d6) δ 8.28 (d, J = 1.4 Hz, 1H), 7.93-7.84 (m, 2H), 7.81 (dd, J = 10.5, 6.5 Hz, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.57 (t, J = 7.8 Hz, 1H), 7.52 (dd, J = 7.5, 1.7 Hz, 1H), 7.41 (ddd, J = 11.4, 4.4, 2.8 Hz, 2H), 7.35 (dd, J = 7.8, 1.6 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.53 (s, 2H), 4.65 (t, J = 5.1 Hz, 2H), 4.51 (s, 2H), 4.34 (s, 1H), 3.70 (t, J = 5.0 Hz, 2H), 3.22 (s, 3H).11232629.21H NMR (400 MHz, DMSO-d6) δ 8.31 (d, J = 1.4 Hz, 1H), 8.03 (s, 1H), 7.92- 7.82 (m, 3H), 7.79-7.61 (m, 4H), 7.54 (dd, J = 7.6, 1.7 Hz, 1H), 7.43 (dd, J = 11.5, 6.1 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 4.67 (t, J = 5.1 Hz, 2H), 4.54 (s, 2H), 4.00 (s, 3H), 3.70 (d, J = 5.0 Hz, 2H), 3.21 (s, 3H).11333628.21H NMR (400 MHz, DMSO-d6) δ 9.18 (d, J = 1.4 Hz, 1H), 8.25 (d, J = 1.4 Hz, 1H), 7.97-7.80 (m, 4H), 7.76 (t, J = 7.8 Hz, 1H), 7.65- 7.57 (m, 2H), 7.52 (ddd, J = 15.7, 7.7, 1.7 Hz, 2H), 7.41 (dd, J = 11.5, 6.0 Hz, 1H), 6.98 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 4.63 (t, J = 5.1 Hz, 2H), 4.48 (s, 2H), 3.88 (s, 3H), 3.70 (s, 2H), 3.22 (s, 3H).11434678.21H NMR (400 MHz, Acetonitrile-d3) δ 8.28 (s, 1H), 8.00 (s, 1H), 7.96 (s, 0H), 7.93-7.88 (m, 2H), 7.80 (t, 1H), 7.69 (d, 1H), 7.56 (t, 3H), 7.42-7.35 (m, 2H), 7.23 (dd, 1H), 6.87 (d, 1H), 6.24 (tt, 1H), 5.56 (s, 2H), 4.55-4.51 (m, 2H), 4.49 (s, 2H), 3.73 (t, 2H), 3.25 (s, 3H).11522622.21H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.94-7.76 (m, 3H), 7.62 (dd, J = 12.6, 8.3 Hz, 2H), 7.57-7.42 (m, 3H), 7.33 (dd, J = 8.2, 2.0 Hz, 1H), 6.96 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.54 (dd, J = 14.1, 2.9 Hz, 2H), 4.49-4.33 (m, 2H), 3.82-3.72 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).11622648.61H NMR (400 MHz, DMSO) δ 13.12 (s, 1H), 8.79 (d, J = 5.2 Hz, 1H), 8.36 (s, 1H), 7.96 (dd, J = 10.2, 6.2 Hz, 1H), 7.68- 7.60 (m, 2H), 7.60-7.51 (m, 2H), 7.51 (d, J = 2.1 Hz, 1H), 7.35 (dd, J = 8.2, 2.0 Hz, 1H), 5.54 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.58 (d, J = 17.0 Hz, 1H), 4.53 (d, J = 11.8 Hz, 1H), 4.48-4.38 (m, 2H), 3.76 (s, 10H), 3.76 (d, J = 16.5 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H).11722642.01H NMR (400 MHz, DMSO) δ 8.79 (d, J = 5.2 Hz, 1H), 8.36 (s, 1H), 7.96 (dd, J = 10.2, 6.2 Hz, 1H), 7.68-7.48 (m, 5H), 7.35 (dd, J = 8.3, 2.1 Hz, 1H), 5.54 (s, 2H), 5.04 (d, J = 6.5 Hz, 1H), 4.59 (d, J = 17.0 Hz, 1H), 4.53 (d, J = 11.8 Hz, 1H), 4.48-4.38 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H).11822672.11H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.3 Hz, 1H), 8.82 (s, 1H), 8.47 (s, 1H), 8.30 (s, 1H), 8.02 (d, J = 1.3 Hz, 1H), 7.97- 7.84 (m, 2H), 7.79 (dd, J = 8.5, 1.5 Hz, 1H), 7.61 (d, J = 8.5 Hz, 1H), 7.56 (d, J = 7.2 Hz, 1H), 7.46 (dd, J = 11.1, 6.4 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.67 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.57- 4.48 (m, 2H), 4.46-4.33(m, 2H), 3.79-3.70 (m,2H), 1.33 (s, 3H), 0.60 (s,3H).11922595.61H NMR (400 MHz, Methanol-d4) δ 8.87 (s, 1H), 8.16 (dd, J = 8.6, 1.4 Hz, 1H), 7.91-7.72 (m, 5H), 7.66 (d, J = 8.4 Hz, 2H), 7.58 (dd, J = 7.5, 1.6 Hz, 1H), 7.37 (dd, J = 11.2, 6.1 Hz, 1H), 6.97 (d, J = 8.2 Hz, 1H), 5.59 (s, 2H), 5.12 (d, J = 6.6 Hz, 1H), 4.76-4.60 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 3.99 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H).12022631.21H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.11 (dd, J = 8.6, 1.4 Hz, 1H), 7.89 (t, J = 7.9 Hz, 1H), 7.81-7.66 (m, 3H), 7.66- 7.55 (m, 3H), 7.01 (d, J = 8.3 Hz, 1H), 5.65 (s, 2H), 5.17 (d, J = 6.6 Hz, 1H), 4.84-4.68 (m, 2H), 4.68 - 4.49 (m, 2H), 4.00 (d, J = 8.8 Hz, 1H), 3.87 (d, J = 8.8 Hz, 1H), 1.48 (s, 3H), 0.82 (s, 3H).12122623.51H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 1H), 8.72 (d, J = 5.2 Hz, 1H), 8.14 (dd, J = 8.6, 1.4 Hz, 1H), 8.06 (dd, J = 10.4, 6.1 Hz, 1H), 7.75 (d, J = 8.6 Hz, 1H), 7.68 (dd, J = 5.2, 1.6 Hz, 1H), 7.60 (t, J = 8.1 Hz, 1H), 7.46 (dd, J = 11.2, 5.9 Hz, 1H), 7.32-7.22 (m, 2H), 5.61 (s, 2H), 5.12 (d, J = 6.6 Hz, 1H), 4.81-4.58 (m, 3H), 4.53 (dd, J = 11.5,6.8 Hz, 1H), 3.99 (d, J = 8.9Hz, 1H), 3.84 (d, J = 8.9 Hz,1H), 1.42 (s, 3H), 0.76 (s,3H).12222649.71H NMR (400 MHz, Methanol-d4) δ 8.57 (s, 1H), 7.86 (t, J = 7.9 Hz, 1H), 7.74 (t, J = 7.5 Hz, 1H), 7.69- 7.50 (m, 5H), 6.98 (d, J = 8.2 Hz, 1H), 5.64 (s, 2H), 5.10 (d, J = 6.6 Hz, 1H), 4.75-4.61 (m, 2H), 4.55 (dd, J = 11.4, 6.8 Hz, 1H), 4.45 (d, J = 17.2 Hz, 1H), 3.95 (d, J = 8.8 Hz, 1H), 3.84 (d, J = 8.8 Hz, 1H), 1.46 (s, 3H), 0.78 (s, 3H).12330649.61H NMR (400 MHz, Methanol-d4) δ 8.56 (s, 1H), 7.87 (t, J = 7.9 Hz, 1H), 7.75 (t, J = 7.5 Hz, 1H), 7.69- 7.53 (m, 5H), 6.98 (d, J = 8.3 Hz, 1H), 5.64 (s, 2H), 5.10 (d, J = 6.6 Hz, 1H), 4.78-4.60 (m, 2H), 4.55 (dd, J = 11.3, 6.8 Hz, 1H), 4.44 (d, J = 17.1 Hz, 1H), 3.95 (d, J = 8.8 Hz, 1H), 3.84 (d, J = 8.8 Hz, 1H), 1.46 (s, 3H), 0.78 (s, 3H)12430649.51H NMR (400 MHz, Methanol-d4) δ 8.56 (s, 1H), 7.87 (t, J = 7.9 Hz, 1H), 7.75 (t, J = 7.5 Hz, 1H), 7.69- 7.53 (m, 5H), 6.98 (d, J = 8.3 Hz, 1H), 5.64 (s, 2H), 5.10 (d, J = 6.6 Hz, 1H), 4.78-4.60 (m, 2H), 4.55 (dd, J = 11.3, 6.8 Hz, 1H), 4.44 (d, J = 17.1 Hz, 1H), 3.95 (d, J = 8.8 Hz, 1H), 3.84 (d, J = 8.8 Hz, 1H), 1.46 (s, 3H), 0.78 (s, 3H).1251 (step 2)609.51H NMR (400 MHz, Chloroform-d) δ 8.03 (dd, J = 8.7, 6.6 Hz, 1H), 7.82- 7.63 (m, 3H), 7.60-7.37 (m, 4H), 7.18 (dd, J = 11.1, 6.0 Hz, 1H), 5.66 (s, 2H), 5.32 (s, 1H), 4.64 (d, J = 6.9 Hz, 4H), 3.83 (t, J = 4.8 Hz, 2H), 3.34 (s, 3H).12635(M+) 656.01H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 7.95-7.83 (m, 2H), 7.81 (d, J = 1.3 Hz, 1H), 7.61 (t, J = 8.2 Hz, 1H), 7.54 (dd, J = 7.6, 1.6 Hz, 1H), 7.47 (ddd, J = 17.2, 10.7, 4.1 Hz, 2H), 7.33 (dd, J = 8.2, 2.0 Hz, 1H), 6.95 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.69-4.25 (m, 4H), 3.88-3.67 (m, 2H), 1.32 (s, 3H), 0.60 (s, 3H).12735(M+) 665.01H NMR (400 MHz, DMSO-d6) δ 8.46 (s, 1H), 7.94 (dd, J = 10.0, 1.4 Hz, 1H), 7.88 (dd, J = 10.2, 8.2 Hz, 1H), 7.84-7.67 (m, 4H), 7.63-7.49 (m, 1H), 7.45 (dd, J = 11.4, 6.2 Hz, 1H), 5.70 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.61-4.34 (m, 4H), 3.79-3.72 (m, 2H), 3.17 (s, 1H), 1.32 (s, 3H), 0.59 (s, 3H).12935(M+) 648.01H NMR (400 MHz, DMSO-d6) δ 13.12 (s, 1H), 8.79 (d, J = 5.2 Hz, 1H), 8.47 (s, 1H), 7.95 (dt, J = 10.5, 3.2 Hz, 2H), 7.83- 7.72 (m, 3H), 7.66 (dd, J = 5.2, 1.7 Hz, 1H), 7.55 (dd, J = 11.5, 5.9 Hz, 1H), 5.64 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.66-4.35 (m, 5H), 3.75 (q, J = 8.7 Hz, 2H), 1.32 (s, 3H), 0.60 (s, 3H).13035(M+) 647.01H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 8.02-7.84 (m, 2H), 7.86- 7.79 (m, 2H), 7.79-7.70 (m, 2H), 7.55 (dd, J = 7.6, 1.6 Hz, 1H), 7.45 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.70-4.28 (m, 4H), 3.88- 3.56 (m, 3H), 1.32 (s, 3H), 0.59 (s, 3H).13135(M+) 657.01H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 8.27 (d, J = 7.0 Hz, 1H), 7.90 (dd, J = 10.2, 8.2 Hz, 1H), 7.67-7.48 (m, 4H), 7.35 (dd, J = 8.3, 2.0 Hz, 1H), 6.98 (d, J = 7.6 Hz, 1H), 5.64-5.35 (m, 4H), 5.14 (d, J = 6.5 Hz, 1H), 4.54 (d, J = 11.3 Hz, 1H), 4.43 (dd, J = 11.3, 6.7 Hz, 1H), 3.93-3.69 (m, 2H), 1.38 (s, 3H), 0.67 (s, 3H).13235(M+) 648.01H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 8.27 (d, J = 7.0 Hz, 1H), 8.03-7.85 (m, 2H), 7.85- 7.69 (m, 2H), 7.62-7.57 (m, 1H), 7.55 (d, J = 11.2 Hz, 1H), 6.93 (d, J = 7.6 Hz, 1H), 5.68 (s, 2H), 5.61- 5.33 (m, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.61-4.45 (m, 1H), 4.45-4.28 (m, 1H), 3.89-3.68 (m, 2H), 1.38 (s, 3H), 0.67 (s, 3H).13335(M+) 625.31H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.92-7.82 (m, 3H), 7.64 (d, J = 8.0 Hz, 2H), 7.51 (d, J = 11.2 Hz, 1H), 7.43 (s, 1H), 7.33 (s, 1H), 7.25 (d, J = 7.3 Hz, 1H), 6.98 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.62-4.44 (m, 2H), 4.44-4.27 (m, 2H), 3.91-3.64 (m, 2H), 2.23 (s, 3H), 1.33 (s, 3H).13435(M+) 607.51H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.92-7.83 (m, 4H), 7.81 (dd, J = 8.5, 1.5 Hz, 1H), 7.63 (t, J = 8.2 Hz, 3H), 7.42 (s, 1H), 7.33 (s, 1H), 7.25 (d, J = 7.3 Hz, 1H), 6.98 (d, J = 8.2 Hz, 1H),5.51 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.66-4.46 (m, 2H), 4.46-4.27 (m, 2H), 2.23 (s, 3H), 1.33 (s, 3H), 0.65 (s, 3H).13535(M+) 624.61H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.01-7.83 (m, 2H), 7.83- 7.73 (m, 1H), 7.66 (d, J = 8.1 Hz, 2H), 7.61 (d, J = 8.5 Hz, 1H), 7.43 (s, 1H), 7.34 (s, 1H), 7.29 (dd, J = 8.1, 2.8 Hz, 1H), 5.59 (s, 2H), 5.00 (d, J = 6.7 Hz, 1H), 4.61- 4.48 (m, 2H), 4.48-4.27 (m, 2H), 3.80 (d, J = 8.6 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 2.23 (s, 2H), 1.33 (s, 3H), 0.65 (s, 3H).13635(M+) 642.61H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.95-7.78 (m, 3H), 7.66 (d, J = 8.1 Hz, 2H), 7.51 (d, J = 11.2 Hz, 1H), 7.43 (s, 1H), 7.34 (s, 1H), 7.29 (dd, J = 8.1, 2.8 Hz, 1H), 5.59 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.64-4.45 (m, 2H), 4.45-4.26 (m, 2H), 3.88- 3.69 (m, 2H), 2.23 (s, 3H), 1.33 (s, 4H), 0.65 (s, 3H).13735(M+) 638.61H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 8.26 (d, J = 6.9 Hz, 1H), 7.99-7.88 (m, 1H), 7.61 (t, J = 8.2 Hz, 1H), 7.57-7.51 (m, 2H), 7.49 (dd, J = 10.0, 2.1 Hz, 1H), 7.33 (dd, J = 8.2, 2.0 Hz, 1H), 7.05 (d, J = 8.3 Hz, 1H), 7.00 (d, J = 7.5 Hz, 1H), 5.64-5.40 (m, 4H), 5.14 (d, J = 6.6 Hz, 1H), 4.54 (d, J = 11.3 Hz, 1H), 4.43 (dd, J = 11.3, 6.8Hz, 1H), 3.79-3.67 (m,2H), 1.38 (s, 3H), 0.67 (s,3H).13835(M+) 660.61H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.97-7.89 (m, 1H), 7.85 (dd, J = 10.5, 8.1Hz, 1H), 7.74 (dd, J = 3.6, 1.9 Hz, 2H), 7.51 (dd, J = 11.3, 1.2 Hz, 1H), 7.45 (s, 1H), 7.38- 7.29 (m, 2H), 5.62 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.63-4.45 (m, 2H), 4.45- 4.34 (m, 2H), 3.79 (d, J = 8.7 Hz, 1H), 3.72 (d, J = 8.6 Hz, 1H), 2.27 (s, 3H), 1.33 (s, 3H), 0.65 (s, 3H).13935(M+) 642.61H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.96-7.82 (m, 2H), 7.72 (d, J = 5.8 Hz, 2H), 7.53-7.48 (m, 1H), 7.44 (s, 1H), 7.34 (s, 1H), 7.28 (d, J = 7.3 Hz, 1H), 6.97 (d, J = 8.3 Hz, 1H), 5.53 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.62-4.48 (m, 2H), 4.44-4.33 (m, 2H), 3.79 (d, J = 8.7 Hz, 1H), 3.72 (d, J = 8.7 Hz, 1H), 2.27 (s, 3H), 1.32 (s, 3H), 0.65 (s, 3H).14035(M+) 642.61H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.96-7.90 (m, 1H), 7.90- 7.79 (m, 2H), 7.74 (d, J = 5.6 Hz, 2H), 7.63 (d, J = 8.5 Hz, 1H), 7.45 (s, 1H), 7.35 (s, 1H), 7.31 (dd, J = 8.1, 2.8 Hz, 1H), 5.61 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.64- 4.46 (m, 2H), 4.46-4.26 (m, 2H), 3.74 (m, 2H), 2.26 (s, 3H), 1.33 (s, 3H), 0.65 (s, 3H).14135(M+) 624.61H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.96-7.85 (m, 2H), 7.81 (d, J = 8.6 Hz, 1H), 7.78-7.66 (m, 2H), 7.63 (d, J = 8.5 Hz, 1H), 7.44 (s, 1H), 7.35 (s, 1H), 7.27 (d, J = 7.3 Hz, 1H), 6.98 (d, J = 8.3 Hz, 1H), 5.53 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.63-4.47 (m, 2H), 4.47-4.31 (m, 2H), 3.74 (m, 2H), 2.26 (s, 3H), 1.32 (s, 3H), 0.65 (s, 3H).14235(M+) 633.01H NMR (400 MHz, DMSO-d6) δ 13.14 (s, 1H), 8.36 (s, 1H), 8.26 (d, J = 6.9 Hz, 1H), 7.92 (t, J = 9.0 Hz, 2H), 7.80-7.67 (m, 2H), 7.62-7.48 (m, 2H), 7.10 (d, J = 8.3 Hz, 1H), 6.95 (d, J = 7.4 Hz, 1H), 5.69-5.30 (m, 4H), 5.14 (d, J = 6.7 Hz, 1H), 4.54 (d, J = 11.2 Hz, 1H), 4.43 (dd, J = 11.3, 6.7 Hz, 1H), 3.92-3.61 (m, 2H), 1.38 (s, 3H), 0.67 (s, 3H).14335(M+) 615.01H NMR (400 MHz, Methanol-d4) δ 8.16 (d, J = 1.3 Hz, 1H), 7.81 (dd, J = 8.3, 7.3 Hz, 1H), 7.72-7.60 (m, 2H), 7.60-7.49 (m, 2H), 7.41 (s, 1H), 7.19- 7.10 (m, 2H), 6.91 (d, J = 8.3 Hz, 1H), 5.54 (s, 2H), 5.12 (dd, J = 7.4, 2.4 Hz, 1H), 4.76-4.59 (m, 3H), 4.59-4.35 (m, 3H), 2.77 (dtd, J = 11.4, 8.2, 6.1 Hz, 1H), 2.48 (ddt, J = 11.5, 9.1, 7.2 Hz, 1H), 2.24 (s, 3H).14435(M+) 615.01H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J = 1.4 Hz, 1H), 7.98 (dd, J = 8.5, 1.5 Hz, 1H), 7.72 (t, J = 7.6 Hz, 1H), 7.68-7.64 (m, 1H), 7.64-7.55 (m, 3H), 7.42 (s, 1H), 7.19 (s, 1H), 7.15 (dd, J = 8.1, 2.8 Hz, 1H), 5.63 (s, 2H), 5.17 (qd, J = 7.1, 2.6 Hz, 1H), 4.76- 4.39 (m, 6H), 2.95-2.66 (m, 1H), 2.50 (ddt, J = 11.5, 9.1, 7.2 Hz, 1H), 2.25 (s, 3H).14535(MH+) 630.21H NMR (400 MHz, DMSO-d6) δ 13.14 (s, 1H), 8.36 (s, 1H), 8.26 (d, J = 6.9 Hz, 1H), 7.92 (t, J = 9.0 Hz, 2H), 7.80-7.67 (m, 2H), 7.62-7.48 (m, 2H), 7.10 (d, J = 8.3 Hz, 1H), 6.95 (d, J = 7.4 Hz, 1H), 5.69-5.30 (m, 4H), 5.14 (d, J = 6.7 Hz, 1H), 4.54 (d, J = 11.2 Hz, 1H), 4.43 (dd, J = 11.3, 6.7 Hz, 1H), 3.92-3.61 (m, 2H), 1.38 (s, 3H), 0.67 (s, 3H).14635(M+) 581.21H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J = 1.4 Hz, 1H), 7.98 (dd, J = 8.5, 1.5 Hz, 1H), 7.81 (dd, J = 8.3, 7.3 Hz, 1H), 7.73- 7.63 (m, 2H), 7.63-7.51 (m, 2H), 7.21-7.09 (m, 3H), 6.91 (d, J = 8.2 Hz, 1H), 5.56 (s, 2H), 5.15 (qd, J = 7.1, 2.5 Hz, 1H), 4.70 (dd, J = 15.7, 7.0 Hz, 1H), 4.66- 4.39 (m, 6H), 2.77 (dtd, J = 11.4, 8.2, 6.1 Hz, 1H), 2.56- 2.38 (m, 1H), 2.22 (d, J = 2.6 Hz, 3H).14735(M + H+) 632.21H NMR (400 MHz, Methanol-d4) δ 8.67 (d, J = 9.6 Hz, 1H), 8.56 (s, 0H), 8.32 (d, J = 1.4 Hz, 1H), 7.99 (dd, J = 8.5, 1.5 Hz, 1H), 7.88 (dd, J = 10.7, 6.3 Hz, 1H), 7.79 (t, J = 7.9 Hz, 1H), 7.68 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.6, 1.6 Hz, 1H), 7.20 (dd, J = 11.2, 6.0 Hz, 2H), 6.87 (d, J = 8.1 Hz, 1H), 5.61 (s, 2H), 5.21 (tt, J = 7.3, 3.6 Hz, 1H), 4.80-4.35 (m, 9H), 2.89-2.65(m, 1H), 2.61-2.35 (m,1H), 2.04 (s, 3H).14835(M + H+) 665.01H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J = 2.8 Hz, 1H), 8.14 (s, 1H), 7.86 (dd, J = 9.6, 6.3 Hz, 2H), 7.81-7.73 (m, 1H), 7.72- 7.63 (m, 1H), 7.50 (dd, J = 11.4, 1.2 Hz, 1H), 7.42 (dd, J = 11.1, 6.0 Hz, 1H), 5.77 (s, 2H), 5.18-4.94 (m, 1H), 4.78 (dd, J = 15.5, 7.1 Hz, 1H), 4.69-4.40 (m, 4H), 4.35 (dt, J = 8.9, 5.9 Hz, 1H), 2.70 (ddd, J = 16.1, 6.3, 2.8 Hz, 1H), 2.43-2.26 (m, 1H).14935(M + H+) 647.01H NMR (400 MHz, DMSO-d6) δ 8.73 (d, J = 5.8 Hz, 1H), 8.12 (s, 1H), 7.88 (dd, J = 10.2, 6.2 Hz, 1H), 7.83 (t, J = 7.6 Hz, 1H), 7.77 (dd, J = 9.9, 1.6 Hz, 1H), 7.65 (d, J = 8.0 Hz, 1H), 7.50 (dd, J = 11.5, 1.2 Hz, 1H), 7.41 (dd, J = 11.1, 6.0 Hz, 1H), 7.07 (d, J = 5.8 Hz, 1H), 5.68 (s, 2H), 5.07 (qd, J = 6.9, 2.8 Hz, 1H), 4.78 (dd, J = 15.6, 7.1 Hz, 1H), 4.70- 4.42 (m, 4H), 4.35 (dt, J =9.1, 6.0 Hz, 1H), 2.86-2.64(m, 1H), 2.44-2.25 (m,1H).1504(MH+) 652.21H NMR (400 MHz, DMSO-d6) δ 8.37 (s, 1H), 7.98-7.85 (m, 2H), 7.83 (dd, J = 8.4, 1.5 Hz, 1H), 7.75 (q, J = 5.4 Hz, 3H), 7.62 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.46 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.69 (s, 1H), 4.62-4.44 (m, 4H), 4.31 (dd, J = 14.4, 8.5 Hz, 1H), 2.26 (d, J = 14.1 Hz, 1H), 2.11 (s, 2H), 1.80 (dd, J = 15.7, 13.0 Hz, 1H), 1.65 (d, J = 7.0 Hz, 1H), 0.96- 0.80 (m, 2H).15135(M + H+) 631.21H NMR (400 MHz, DMSO-d6) δ 8.46 (s, 1H), 8.20 (t, J = 9.9 Hz, 1H), 7.89 (d, J = 8.2 Hz, 2H), 7.78- 7.73 (m, 1H), 7.71 (d, J = 8.1 Hz, 2H), 7.61 (d, J = 8.4 Hz, 2H), 7.56 (d, J = 8.5 Hz, 1H), 5.67 (s, 2H), 5.08 (d, J = 6.6 Hz, 1H), 4.57 (d, J = 17.7 Hz, 2H), 4.46 (dd, J = 11.1, 6.6 Hz, 1H), 4.29 (d, J = 16.8 Hz, 1H), 3.80-3.71 (m, 2H), 1.37 (s, 3H), 0.64 (s, 3H).15235(M + H+) 613.31H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.15 (t, J = 9.9 Hz, 1H), 7.88 (d, J = 8.1 Hz, 2H), 7.81 (dd, J = 8.5, 1.4 Hz, 1H), 7.75- 7.65 (m, 4H), 7.62 (d, J = 8.5 Hz, 1H), 7.53 (t, J = 8.0 Hz, 1H), 5.65 (s, 2H), 5.00 (d, J = 6.6 Hz, 1H), 4.57- 4.48 (m, 2H), 4.48-4.36 (m, 2H), 3.78 (d, J = 8.6 Hz, 1H), 3.72 (d, J = 8.6 Hz, 1H), 1.30 (s, 3H), 0.58 (s, 3H).15335(M + H+) 708.01H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.84 (s, 1H), 8.35 (s, 1H), 8.32 (s, 1H), 8.03 (d, J = 1.3 Hz, 1H), 7.88 (ddd, J = 11.3, 7.4, 4.3 Hz, 2H), 7.64-7.55 (m, 1H), 7.55-7.44 (m, 2H), 5.76 (s, 2H), 5.03 (d, J = 6.5 Hz, 1H), 4.59-4.47 (m, 2H), 4.47-4.35 (m, 2H), 3.74 (q, J = 8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H).15435(M + H+) 690.01H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.3 Hz, 1H), 8.86 (s, 1H), 8.34 (s, 1H), 8.32 (s, 1H), 8.02 (d, J = 1.3 Hz, 1H), 7.99- 7.91 (m, 2H),7.85 (dd, J = 10.2, 8.2 Hz, 1H), 7.73 (dd, J = 8.3, 2.8 Hz, 1H), 7.50 (dd, J = 9.7, 6.9 Hz, 2H), 5.78 (s, 2H), 5.00 (d, J = 6.7 Hz, 1H), 4.50 (dd, J = 14.1, 8.4 Hz, 2H), 4.46-4.34 (m, 2H), 3.79-3.66 (m, 2H), 1.29 (s, 3H), 0.57 (s, 3H).15535(M + H+) 632.21H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.86 (s, 1H), 8.32 (s, 1H), 8.23 (s, 1H), 8.02 (d, J = 1.3 Hz, 1H), 7.97- 7.88 (m, 2H), 7.88-7.79 (m, 2H), 7.72 (dd, J = 8.2, 2.7 Hz, 1H), 7.60 (d, J = 8.4 Hz, 1H), 7.45 (t, J = 8.0 Hz, 1H), 5.77 (s, 2H), 4.59 (s, 2H), 4.47 (s, 2H), 3.66 (t, J = 5.0 Hz, 2H), 3.20 (s, 3H).15635(M + H+) 684.21H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.3 Hz, 1H), 8.76 (s, 1H), 8.49 (s, 1H), 8.29 (s, 1H), 8.03 (d, J = 1.2 Hz, 1H), 7.89 (t, J = 7.8 Hz, 1H), 7.80 (dd, J = 8.5, 1.5 Hz, 1H), 7.62 (d, J = 8.5 Hz, 1H), 7.52 (s, 1H), 7.36 (s, 1H), 7.29 (d, J = 7.3 Hz, 1H), 6.98 (d, J = 8.3 Hz, 1H), 5.58 (s, 2H), 5.00 (d, J = 6.8 Hz, 1H), 4.61-4.48 (m, 2H), 4.46-4.33 (m,2H), 3.84-3.69 (m, 2H),2.33 (s, 3H), 1.32 (s, 3H),0.64 (s, 3H).15735(M + H+) 690.11H NMR (400 MHz, DMSO-d6) δ 8.90 (s, 1H), 8.83 (s, 1H), 8.47 (s, 1H), 8.31 (s, 1H), 8.03 (d, J = 1.3 Hz, 1H), 7.96 (t, J = 7.9 Hz, 1H), 7.77 (d, J = 8.2 Hz, 2H), 7.60 (dd, J = 10.7, 7.7 Hz, 2H), 7.06 (d, J = 8.3 Hz, 1H), 5.68 (s, 2H), 5.08 (d, J = 6.5 Hz, 1H), 4.68-4.53 (m, 2H), 4.46 (dd, J = 11.3, 6.9 Hz, 1H), 4.35 (d, J = 17.2 Hz, 1H), 3.76 (d, J = 2.8 Hz, 2H), 1.38 (s, 3H), 0.65 (s, 3H).15835(M + H+) 654.21H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.3 Hz, 1H), 8.83 (s, 1H), 8.47 (s, 1H), 8.30 (s, 1H), 8.02 (d, J = 1.3 Hz, 1H), 8.00- 7.93 (m, 2H), 7.88 (t, J = 7.9 Hz, 1H), 7.80 (dd, J = 8.4, 1.5 Hz, 1H), 7.70 (d, J = 7.5 Hz, 1H), 7.61 (d, J = 8.5 Hz, 1H), 7.50 (t, J = 8.1 Hz, 1H), 6.95 (d, J = 8.2 Hz, 1H), 5.69 (s, 2H), 4.98 (d, J = 6.8 Hz, 1H), 4.54-4.47 (m,2H), 4.47-4.35 (m, 2H),3.81-3.68 (m, 2H), 1.29 (s,3H), 0.57 (s, 3H).15935(M + H+) 708.11H NMR (400 MHz, DMSO-d6) δ 8.91 (d, J = 1.3 Hz, 1H), 8.85 (s, 1H), 8.47 (s, 1H), 8.33 (s, 1H), 8.03 (d, J = 1.2 Hz, 1H), 7.99- 7.87 (m, 1H), 7.77 (d, J = 8.6 Hz, 2H), 7.64 (d, J = 8.4 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 5.77 (s, 2H), 5.08 (d, J = 6.7 Hz, 1H), 4.64 (d, J = 17.5 Hz, 1H), 4.57 (d, J = 11.3 Hz, 1H), 4.51 -4.42 (m, 1H), 4.35 (d, J = 17.6Hz, 1H), 3.76 (d, J = 2.5 Hz,2H), 1.38 (s, 3H), 0.65 (s,3H).16035(M + H+) 690.11H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.84 (s, 1H), 8.47 (s, 1H), 8.32 (s, 1H), 8.03 (d, J = 1.2 Hz, 1H), 7.87 (td, J = 9.7, 9.1, 7.4 Hz, 2H), 7.82-7.75 (m, 1H), 7.60 (t, J = 9.5 Hz, 2H), 7.47 (dd, J = 11.6, 6.1 Hz, 1H), 5.76 (s, 2H), 5.01 (d, J = 6.8 Hz, 1H), 4.58-4.47 (m, 2H), 4.48-4.32 (m, 2H), 3.80- 3.68 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H).16135(M + H+) 672.21H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.86 (s, 1H), 8.48 (s, 1H), 8.32 (s, 1H), 8.02 (d, J = 1.3 Hz, 1H), 7.94 (t, J = 8.5 Hz, 2H), 7.85 (dd, J = 10.2, 8.3 Hz, 1H), 7.82- 7.77 (m, 1H), 7.73 (dd, J = 8.3, 2.7 Hz, 1H), 7.61 (d, J = 8.4 Hz, 1H), 7.50 (t, J = 8.1 Hz, 1H), 5.78 (s, 2H), 4.99 (d, J = 6.6 Hz, 1H), 4.54- 4.47 (m, 2H), 4.46-4.35(m, 2H), 3.81-3.69 (m,2H), 1.29 (s, 3H), 0.57 (s,3H).16235(M + H+) 672.21H NMR (400 MHz, DMSO-d6) δ 9.12 (d, J = 2.2 Hz, 1H), 8.95 (d, J = 1.2 Hz, 1H), 8.62 (d, J = 2.3 Hz, 1H), 8.47 (s, 1H), 8.04 (d, J = 1.2 Hz, 1H), 7.89 (t, J = 7.9 Hz, 1H), 7.79 (dd, J = 8.5, 1.4 Hz, 1H), 7.73 (dd, J = 10.6, 6.4 Hz, 1H), 7.60 (d, J = 8.4 Hz, 1H), 7.53 (d, J = 7.3 Hz, 1H), 7.42 (dd, J = 11.5, 6.0 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.73 (s, 2H),5.02-4.95 (m, 1H), 4.56-4.45 (m, 2H), 4.46-4.30(m, 2H), 3.82-3.67 (m,2H), 1.30 (s, 3H), 0.58 (s,3H).16335(M + H+) 673.21H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.2 Hz, 1H), 8.48 (s, 1H), 8.00 (d, J = 1.1 Hz, 1H), 7.95 (dd, J = 10.8, 2.0 Hz, 1H), 7.88- 7.83 (m, 3H), 7.91-7.76 (m, 2H), 7.61 (d, J = 8.5 Hz, 1H), 7.59-7.53 (m, 1H), 7.46 (dd, J = 11.4, 6.1Hz, 1H), 5.68 (s, 2H), 5.01 (d, J = 6.8 Hz, 1H), 4.57-4.48 (m, 2H), 4.48-4.30 (m, 2H), 3.82-3.69 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H).16435(M + H+) 669.21H NMR (400 MHz, DMSO-d6) δ 8.60 (s, 1H), 8.48 (s, 1H), 7.92-7.81 (m, 3H), 7.81-7.75 (m, 3H), 7.61 (d, J = 8.4 Hz, 1H), 7.57-7.51 (m, 1H), 7.45 (dd, J = 11.3, 6.2 Hz, 1H), 6.98 (d, J = 8.3 Hz, 1H), 5.58 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.59-4.48 (m, 2H), 4.50-4.34 (m, 2H), 3.83-3.70 (m, 2H), 2.33 (s, 3H), 1.33 (s, 3H), 0.60 (s, 3H).16535(M + H+) 631.11H NMR (400 MHz, DMSO-d6) δ 7.94-7.86 (m, 2H), 7.80-7.71 (m, 3H), 7.66 (d, J = 7.7 Hz, 1H), 7.54 (d, J = 7.4 Hz, 1H), 7.41 (p, J = 11.0,9.8 Hz, 2H), 6.99 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.22 (d, J = 112.0 Hz, 1H), 4.54 (d, J = 24.0 Hz, 2H), 4.47-4.14 (m, 2H), 3.94-3.58 (m, 2H), 1.29 (d, J = 21.9 Hz, 3H), 0.72 (d, J = 14.3 Hz, 3H).16635(M + H+) 631.21H NMR (400 MHz, DMSO-d6) δ 7.95-7.86 (m, 2H), 7.79-7.71 (m, 3H), 7.66 (t, J = 7.6 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.43 (q, J = 10.6, 7.8 Hz, 2H), 6.99 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.22 (d, J = 111.9 Hz, 1H), 4.54 (d, J = 24.1 Hz, 2H), 4.37 (d, J = 18.0 Hz, 1H), 3.92-3.67 (m, 1H), 1.29 (d, J = 21.9 Hz, 3H), 0.72 (d, J = 14.2 Hz, 3H).16735(M + H+) 655.21H NMR (400 MHz, DMSO-d6) δ 8.89 (s, 1H), 8.48 (s, 1H), 8.00 (s, 1H), 7.95-7.87 (m, 3H), 7.87- 7.81 (m, 2H), 7.79 (dd, J = 8.3, 1.7 Hz, 2H), 7.61 (d, J = 8.5 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.46 (dd, J = 11.3, 6.1 Hz, 1H), 6.98 (d, J = 8.3 Hz, 1H), 5.59 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.58- 4.47 (m, 2H), 4.47-4.28 (m, 2H), 3.81-3.63 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H).16835(M + H+) 656.21H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 7.90 (t, J = 7.9 Hz, 1H), 7.83- 7.70 (m, 4H), 7.62 (t, J = 7.8 Hz, 2H), 7.54 (d, J = 7.3 Hz, 1H), 7.44 (dd, J = 11.3, 6.1 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.00 (d, J = 6.6 Hz, 1H), 4.57- 4.47 (m, 2H), 4.43 (dd, J = 11.1, 6.8 Hz, 1H), 4.36 (d, J = 17.0 Hz, 1H), 3.83-3.69 (m, 2H), 1.32 (s, 3H), 0.59 (s, 3H).16935(M + H+) 674.11H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.90 (t, J = 7.8 Hz, 1H), 7.81- 7.71 (m, 3H), 7.62 (d, J = 8.0 Hz, 1H), 7.53 (dd, J = 12.7, 9.2 Hz, 2H), 7.45 (dd, J = 11.4, 6.1 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.59-4.47 (m, 2H), 4.47-4.34 (m, 2H), 3.74 (q, J = 8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H).17035(M + H+) 653.21H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.92-7.81 (m, 2H), 7.79 (dd, J = 10.3, 6.5 Hz, 1H), 7.56-7.49 (m, 2H), 7.46 (dd, J = 11.3, 6.2 Hz, 1H), 7.12 (d, J = 7.7 Hz, 1H), 6.96 (d, J = 8.2 Hz, 1H), 5.41 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.60-4.47 (m, 2H), 4.47-4.33 (m, 2H), 3.93 (s, 3H), 3.74 (q, J = 8.7 Hz, 2H), 3.10 (qd, J = 7.3, 4.8 Hz, 1H), 1.33 (s, 3H), 0.60 (s, 3H).17135(M + H+) 690.11H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.4 Hz, 1H), 8.82 (s, 1H), 8.35 (s, 1H), 8.30 (s, 1H), 8.02 (d, J = 1.4 Hz, 1H), 7.92 (d, J = 7.7 Hz, 1H), 7.90-7.86 (m, 1H), 7.57 (dd, J = 7.6, 1.5 Hz, 1H), 7.51 (d, J = 11.4 Hz, 1H), 7.47 (dd, J = 11.4, 6.4 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.67 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.58-4.47 (m, 2H), 4.47-4.31 (m, 2H), 3.74 (q, J =8.7 Hz, 2H), 1.33 (s, 3H),0.60 (s, 3H).17236(M + H+) 617.31H NMR (400 MHz, DMSO-d6) δ 8.53 (s, 1H), 7.94-7.90 (m, 1H), 7.89 (d, J = 8.0 Hz, 1H), 7.81 (dd, J = 8.4, 1.5 Hz, 1H), 7.79- 7.70 (m, 3H), 7.62 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.39 (dd, J = 11.4, 6.1 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.61 (d, J = 9.4 Hz, 3H), 4.56 (d, J = 6.1 Hz, 1H), 4.52 (s, 1H), 4.42 (d, J = 17.0 Hz, 1H),4.34-4.10 (m, 2H), 4.06 (td, J =9.0, 1.7 Hz, 1H), 3.97-3.77(m, 2H), 3.31 (dq, J = 15.6,7.6 Hz, 1H).17335(M + H+) 690.01H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.3 Hz, 1H), 8.82 (s, 1H), 8.35 (s, 1H), 8.30 (s, 1H), 8.02 (d, J = 1.2 Hz, 1H), 7.94- 7.91 (m, 1H), 7.90-7.87 (m, 1H), 7.57 (dd, J = 7.6, 1.6 Hz, 1H), 7.52 (dd, J = 11.2, 1.2 Hz, 1H), 7.47 (dd, J = 11.4, 6.5 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.67 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.53 (dd, J = 14.2, 11.9Hz, 2H), 4.47-4.35 (m,2H), 3.74 (q, J = 8.7 Hz,2H), 1.33 (s, 3H), 0.60 (s,3H).17435(M + H+) 689.41H NMR (400 MHz, DMSO-d6) δ 9.33 (s, 1H), 8.78 (s, 1H), 8.35 (s, 1H), 8.30 (s, 1H), 8.27 (t, J = 1.6 Hz, 1H), 7.92 (d, J = 7.9 Hz, 1H), 7.89 (t, J = 3.3 Hz, 1H), 7.57 (dt, J = 4.4, 1.7 Hz, 2H), 7.52 (dd, J = 11.2, 1.2 Hz, 1H), 7.49-7.43 (m, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.66 (s, 2H), 5.03 (d, J = 6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.47-4.35 (m,2H), 3.74 (q, J = 8.7 Hz,2H), 1.33 (s, 3H), 0.61 (s,3H).17535(M + H+) 674.11H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.90 (t, J = 7.9 Hz, 1H), 7.83- 7.77 (m, 2H), 7.74 (dd, J = 10.4, 1.9 Hz, 1H), 7.64- 7.60 (m, 1H), 7.57-7.49 (m, 2H), 7.46 (dd, J = 11.4, 6.1 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.61- 4.47 (m, 2H), 4.47-4.34 (m, 2H), 3.74 (q, J = 8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H).176 A22629.01H NMR (400 MHz, DMSO) δ 8.45 (s, 1H), 7.96- 7.86 (m, 2H), 7.80-7.71 (m, 4H), 7.53 (d, J = 8.2 Hz, 2H), 7.35 (dd, J = 11.0, 5.7 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.48 (s, 1H), 4.49 (d, J = 11.9 Hz, 2H),4.42 (d, J = 17.2 Hz, 1H), 4.21 (dd, J = 10.8, 6.8 Hz, 1H), 4.16-4.03 (m, 1H), 3.78 (s, 1H), 3.09 (s, 2H), 3.07-2.99 (m, 1H), 2.92 (s, 3H), 2.59 (s, 1H). (s, 1H).176 B22629.01H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 7.96- 7.86 (m, 2H), 7.82-7.71 (m, 4H), 7.62-7.51 (m, 2H), 7.36 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.50 (s, 1H), 4.56-4.47 (m, 2H), 4.43 (d, J = 16.8 Hz, 1H), 4.21 (dd, J = 10.9, 6.6 Hz, 1H), 4.08 (t, J = 8.7 Hz, 1H), 3.77 (t, J = 8.2 Hz, 1H), 3.14- 3.00 (m, 2H), 2.92 (s, 3H), 2.63-2.52 (m, 1H), 1.24 (s, 1H).17735673.01H NMR (400 MHz, DMSO) δ 13.05 (s, 1H), 8.84 (d, J = 1.1 Hz, 1H), 8.35 (s, 1H), 8.01-7.93 (m, 3H), 7.85 (ddd, J = 13.3, 10.3, 7.4 Hz, 2H), 7.75 (d, J = 8.3 Hz, 2H), 7.60-7.43 (m, 3H), 5.66 (s, 2H), 5.03 (d, J = 6.5 Hz, 1H), 4.59- 4.49 (m, 2H), 4.47-4.34 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H).178355921H NMR (400 MHz, DMSO) δ 8.74 (d, 1H), 8.49 (s, 1H) 7.80-7.90 (m, 3H), 7.70-7.60 (m, 3H), 7.46-7.36 (m, 3H), 5.56 (s, 2H), 5.02 (d, 1H), 4.53-4.43 (m, 4H), 3.82-3.72 (m, 2H), 2.33 (s, 3H), 1.32 (s, 3H), 0.60 (s, 3H).17935591.21H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.92- 7.78 (m, 4H), 7.63 (dd, J = 8.3, 4.5 Hz, 3H), 7.33-7.20 (m, 3H), 5.51 (s, 2H), 4.56- 4.38 (m, 3H), 3.82-3.70 (m, 2H), 3.74 (s, 21H), 2.22 (s, 3H), 1.31 (s, 3H), 0.60 (s, 3H).18035609.01H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.90- 7.78 (m, 4H), 7.64 (dd, J = 12.6, 8.3 Hz, 3H), 7.33- 7.21 (m, 3H), 5.59 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.56-4.38 (m, 3H), 4.33 (d, J = 16.9 Hz, 1H), 3.79 (d, J = 8.6 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 2.22 (s, 3H), 1.31 (s, 3H), 0.60 (s, 3H).18135655.21H NMR (400 MHz, DMSO) δ 8.84 (d, J = 1.2 Hz, 1H), 8.51 (s, 1H), 8.01- 7.93 (m, 3H), 7.91-7.83 (m, 1H), 7.87-7.79 (m, 2H), 7.78-7.71 (m, 2H), 7.64 (d, J = 8.4 Hz, 1H), 7.60-7.52 (m, 1H), 7.48 (dd, J = 11.4, 6.1 Hz, 1H), 5.66 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.60-4.51 (m, 2H), 4.48-4.37 (m, 2H), 3.79 (d, J = 8.6 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 1.33 (s, 3H), 0.61 (s, 3H).18235638.31H NMR (400 MHz, DMSO) δ 8.87-8.77 (m, 2H), 8.50 (s, 1H), 8.01- 7.92 (m, 4H), 7.82 (dd, J = 8.5, 1.5 Hz, 1H), 7.74 (d, J = 8.4 Hz, 2H), 7.67-7.60 (m, 2H), 7.56 (dd, J = 11.4, 5.9 Hz, 1H), 5.60 (s, 2H), 5.21 (s, 15H), 5.03 (d, J = 6.6 Hz, 1H), 4.59 (d, J = 17.1Hz, 1H), 4.55 (d, J = 11.7 Hz, 1H), 4.48-4.38 (m, 2H), 3.78 (d, J = 8.6 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 1.34 (s, 3H), 0.62 (s, 3H).18335636.61H NMR (400 MHz, DMSO) δ 8.83 (d, J = 1.2 Hz, 1H), 8.50 (s, 1H), 7.98 (d, J = 1.2 Hz, 1H), 7.98- 7.91 (m, 2H), 7.90 (t, J = 7.8 Hz, 1H), 7.86 (dd, J = 10.3, 6.6 Hz, 1H), 7.82 (dd, J = 8.4, 1.5 Hz, 1H), 7.76-7.69 (m, 2H), 7.63 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.6, 1.6 Hz, 1H), 7.47 (dd, J = 11.2, 6.3 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.57 (s, 2H), 5.03(d, J = 6.6 Hz, 1H), 4.59-4.54 (m, 1H), 4.53 (s, 1H),4.48-4.35 (m, 2H), 4.03 (s,21H), 3.78 (d, J = 8.7 Hz,1H), 3.73 (d, J = 8.6 Hz,1H), 1.33 (s, 3H), 0.61 (s,3H).18422628.21H NMR (400 MHz, DMSO) δ 12.76 (s, 1H), 8.55 (s, 1H), 7.96-7.86 (m, 2H), 7.81 (dd, J = 8.3, 1.5 Hz, 1H), 7.81-7.68 (m, 3H), 7.62 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.6, 1.7 Hz, 1H), 7.38 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.52 (s, 1H), 4.58-4.41 (m, 3H), 4.21 (dd, J = 10.8, 6.5 Hz, 1H), 4.08 (t, J = 8.7 Hz, 1H),3.77 (t, J = 8.1 Hz, 1H), 3.17-3.01 (m, 2H), 2.91 (s, 3H),2.60 (t, J = 8.7 Hz, 1H).18522664.61H NMR (400 MHz, DMSO) δ 8.53 (s, 1H), 7.96- 7.86 (m, 2H), 7.81 (dd, J = 8.4, 1.5 Hz, 1H), 7.81-7.70 (m, 3H), 7.66-7.50 (m, 2H), 7.38 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 6.6-6.25 (m, 1H), 5.61 (s, 2H), 4.56 (d, J = 16.9 Hz, 1H), 4.50 (d, J = 10.9 Hz, 1H), 4.42 (d, J = 17.0 Hz, 1H), 4.19 (dd, J = 10.9, 6.7 Hz, 1H), 4.09 (t, J = 8.6 Hz, 1H), 3.93-3.86 (m, 1H), 3.61-3.51 (m, 1H).18635654.61H NMR (400 MHz, DMSO) δ 13.14 (s, 1H), 8.83 (d, J = 1.2 Hz, 1H), 8.36 (s, 1H), 8.00-7.82 (m, 5H), 7.73 (d, J = 8.2 Hz, 2H), 7.58-7.49 (m, 2H), 7.47 (dd, J = 11.2, 6.2 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 5.04 (d, J = 6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 4.26 (s, 13H), 3.75 (q, J = 8.7 Hz, 2H), 2.55 (s, 1H), 1.34 (s, 3H), 0.61 (s, 3H).18736610.61H NMR (400 MHz, DMSO) δ 12.98 (s, 1H), 8.31 (d, J = 1.5 Hz, 1H), 7.96-7.86 (m, 2H), 7.82 (dd, J = 8.5, 1.3 Hz, 1H), 7.82-7.70 (m, 3H), 7.66 (d, J = 8.4 Hz, 1H), 7.55 (dd, J = 7.5, 1.7 Hz, 1H), 7.41 (dd, J = 11.3, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.02 (dt, J = 11.1, 5.0 Hz, 1H), 4.85 (dt, J = 12.8, 4.2 Hz, 2H), 4.58 (d, J = 17.0 Hz, 1H), 4.47 (d, J =17.0 Hz, 1H), 2.37 (d, J =10.9 Hz, 1H), 2.10-1.96(m, 1H), 1.90 (d, J = 10.3Hz, 1H), 1.68 (q, J = 5.3 Hz,2H).18835612.61H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.92- 7.77 (m, 4H), 7.72 (t, J = 8.8 Hz, 3H), 7.62 (d, J = 8.5 Hz, 1H), 7.59-7.52 (m, 1H), 7.46 (dd, J = 11.4, 6.1 Hz, 1H), 5.67 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H),4.58- 4.49 (m, 2H), 4.44 (dd, J = 11.2, 6.8 Hz, 1H), 4.38 (d, J = 16.9 Hz, 1H), 3.82-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).18936624.61H NMR (400 MHz, DMSO) δ 8.56 (s, 1H), 8.22 (s, 1H), 7.96-7.87 (m, 2H), 7.82-7.70 (m, 4H), 7.60 (d, J = 8.4 Hz, 1H), 7.55 (dd, J = 7.5, 1.7 Hz, 1H), 7.41 (dd, J = 11.4, 6.1 Hz, 1H), 7.04- 6.95 (m, 1H), 5.61 (s, 2H), 5.48 (t, J = 7.2 Hz, 1H), 4.63- 4.53 (m, 2H), 4.47 (d, J = 17.0 Hz, 1H), 4.23 (dd, J = 10.9, 6.6 Hz, 1H), 4.12 (t, J = 8.5 Hz, 1H), 3.82 (t, J =8.4 Hz, 1H), 3.02-2.87 (m,1H), 2.18 (p, J = 8.4 Hz,1H), 1.24 (s, 1H), 1.16 (t,J = 7.3 Hz, 2H), 0.24 (s, 2H),0.11 (d, J = 7.1 Hz, 2H),−0.12 (s, 1H).19036624.61H NMR (400 MHz, DMSO) δ 12.74 (s, 1H), 8.55 (s, 1H), 7.96-7.87 (m, 2H), 7.81-7.70 (m, 4H), 7.63-7.51 (m, 2H), 7.41 (dd, J = 11.3, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.47 (t, J = 7.2 Hz, 1H), 4.62-4.52 (m, 2H), 4.46 (d, J = 16.9 Hz, 1H), 4.23 (dd, J = 10.8, 6.7 Hz, 1H), 4.12 (t, J = 8.5 Hz, 1H), 3.81 (t, J = 8.4 Hz, 1H),2.17 (p, J = 8.5 Hz, 1H),1.24 (s, 1H), 0.23 (s, 2H),0.11 (s, 1H), −0.13 (s, 1H).19136624.61H NMR (400 MHz, DMSO) δ 8.28 (s, 1H), 7.96- 7.86 (m, 2H), 7.84-7.72 (m, 4H), 7.62 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.4 Hz, 1H), 7.41 (dd, J = 11.4, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.31 (s, 1H), 4.76-4.42 (m, 2H), 4.35 (t, J = 8.2 Hz, 1H), 4.31- 4.12 (m, 2H), 3.58 (t, J = 9.6 Hz, 1H), 2.89 (d, J = 7.4Hz, 1H), 2.68 (s, 1H), 2.34(s, 1H), 1.92 (s, 0H), 1.24 (s,2H), 1.14 (t, J = 7.2 Hz, 2H),1.04-0.78 (m, 2H), 0.38 (d,J = 9.7 Hz, 2H), 0.11 (s,1H), −0.38 (d, J = 5.2 Hz,1H).19236624.61H NMR (400 MHz, DMSO) δ 12.79 (s, 1H), 8.28 (s, 1H), 8.00-7.84 (m, 2H), 7.77 (dd,J= 15.6, 7.3 Hz, 3H), 7.63 (d, J = 8.5 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.41 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.32 (s, 1H), 4.62-4.42 (m, 2H), 4.40-4.09 (m, 2H), 3.58 (t, J = 9.6 Hz, 1H), 1.92 (d, J = 8.1 Hz, 0H), 1.02-0.82 (m,1H), 0.37 (d, J = 13.0 Hz,1H), 0.10 (d, J = 5.4 Hz,1H), −0.37 (d, J = 4.9 Hz,1H).19335595.61H NMR (400 MHz, DMSO) δ 8.79 (d, J = 5.2 Hz, 1H), 8.36 (s, 1H), 7.91 (dd, J = 17.7, 7.2 Hz, 3H), 7.70 (d, J = 8.2 Hz, 2H), 7.65 (dd, J = 5.2, 1.8 Hz, 1H), 7.60-7.48 (m, 2H), 5.62 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.58 (d, J = 17.1 Hz, 1H), 4.53 (d, J = 11.8 Hz, 1H), 4.48-4.37 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H).19435613.61H NMR (400 MHz, DMSO) δ 8.79 (d, J = 5.2 Hz, 1H), 8.36 (s, 1H), 7.91 (dd, J = 17.7, 7.2 Hz, 3H), 7.70 (d, J = 8.2 Hz, 2H), 7.65 (dd, J = 5.2, 1.8 Hz, 1H), 7.60-7.48 (m, 2H), 5.62 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.58 (d, J = 17.1 Hz, 1H), 4.53 (d, J = 11.8 Hz, 1H), 4.48-4.37 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H).19535634.61H NMR (400 MHz, DMSO) δ 13.08 (s, 1H), 8.49 (d, J = 1.3 Hz, 1H), 8.35 (s, 1H), 7.93 (dd, J = 10.2, 8.3 Hz, 1H), 7.82 (dd, J = 10.2, 6.7 Hz, 1H), 7.66- 7.59 (m, 1H), 7.56-7.43 (m, 2H), 5.98 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.59 - 4.48 (m, 2H), 4.47-4.35 (m, 2H), 3.84 (s, 14H), 3.74 (q, J = 8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H).19635658.61H NMR (400 MHz, DMSO) δ 13.08 (s, 1H), 8.49 (d, J = 1.3 Hz, 1H), 8.35 (s, 1H), 7.93 (dd, J = 10.2, 8.3 Hz, 1H), 7.82 (dd, J = 10.2, 6.7 Hz, 1H), 7.66- 7.59 (m, 1H), 7.56-7.43 (m, 2H), 5.98 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.59 - 4.48 (m, 2H), 4.47-4.35 (m, 2H), 3.84 (s, 14H), 3.74 (q, J = 8.7 Hz, 2H), 1.33 (s, 3H), 0.60 (s, 3H).19736612.61H NMR (400 MHz, DMSO) δ 8.32 (d, J = 1.5 Hz, 1H), 7.96-7.64 (m, 7H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.42 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.09- 4.99 (m, 1H), 4.60-4.44 (m, 2H), 4.23 (ddt, J = 13.1, 8.7, 3.6 Hz, 2H), 4.00 (q, J = 8.0 Hz, 1H), 2.39-2.26 (m, 1H), 1.66-1.47 (m, 2H), 0.86 (t, J = 7.4 Hz, 3H).19835614.51H NMR (400 MHz, DMSO) δ 8.79 (d, J = 5.2 Hz, 1H), 8.49 (s, 1H), 7.98- 7.88 (m, 2H), 7.84-7.72 (m, 3H), 7.69-7.59 (m, 2H), 7.55 (dd, J = 11.4, 5.9 Hz, 1H), 5.64 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.62- 4.50 (m, 2H), 4.48-4.37 (m, 2H), 3.82-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).19936641.21H NMR (400 MHz, DMSO) δ 8.37 (d, J = 1.5 Hz, 1H), 7.95-7.63 (m, 7H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.44 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.23 (dd, J = 10.7, 8.3 Hz, 1H), 4.67 (d, J = 17.0 Hz, 1H), 4.44 (d, J = 16.9 Hz, 1H), 3.00-2.90 (m, 1H), 2.39 (dd, J = 12.7, 8.2 Hz, 1H), 1.50 (s, 3H), 1.39 (d, J = 17.9 Hz, 6H), 1.07 (s, 3H).200356751H NMR (400 MHz, DMSO) δ 13.09 (s, 1H), 8.80 (d, J = 5.1 Hz, 1H), 8.36 (s, 1H), 7.93 (dd, J = 10.2, 6.2 Hz, 1H), 7.87- 7.73 (m, 2H), 7.66 (dt, J = 5.2, 2.6 Hz, 2H), 7.60-7.48 (m, 2H), 5.65 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.58 (d, J = 17.1 Hz, 1H), 4.53 (d, J = 11.9 Hz, 1H), 4.48-4.38 (m, 2H), 4.11 (s, 7H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H).20135640.21H NMR (400 MHz, DMSO) δ 13.07 (s, 1H), 8.36 (s, 1H), 7.93-7.81 (m, 2H), 7.65-7.42 (m, 5H), 7.33 (dd, J = 8.2, 2.1 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.04 (d, J = 6.5 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J = 8.6 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H).20222627.21H NMR (400 MHz, DMSO) δ8.20 (s, 1H), 7.96- 7.86 (m, 2H), 7.83-7.70 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.53 (d, J = 7.0 Hz, 1H), 7.34 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.53 (s, 1H), 4.89 (d, J = 7.0 Hz, 1H), 4.75 (d, J = 7.1 Hz, 1H), 4.63 (q, J = 6.8 Hz, 2H), 4.47 (s, 2H), 4.25 (dd, J = 10.4, 3.4 Hz, 1H),4.16 (dd, J = 10.4, 7.6 Hz, 1H), 3.31 (s, 1H), 2.93 (dd, J = 14.0, 9.1 Hz, 1H), 2.51- 2.39 (m, 1H), 1.24 (s, 1H).20322611.21H NMR (400 MHz, DMSO) δ 12.95 (s, 1H), 8.60 (d, J = 1.5 Hz, 1H), 7.96-7.86 (m, 3H), 7.82- 7.66 (m, 4H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.42 (dd, J = 11.5, 6.1Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.73 (qd, J = 7.6, 3.6 Hz, 1H), 5.60 (s, 2H), 4.61 (s, 2H), 4.17 (qd, J = 10.2, 5.5 Hz, 2H), 2.51- 2.40 (m, 2H), 1.20-1.09 (m, 1H), 0.82 (td, J = 10.4, 6.0 Hz, 2H), 0.65-0.54 (m, 1H).20435649.21H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 8.05 (dd, J = 9.2, 5.2 Hz, 1H), 7.92 (t, J = 7.9 Hz, 1H), 7.77 (ddd, J = 12.5, 9.8, 6.1Hz, 2H), 7.60-7.42 (m, 3H), 7.02 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J = 8.7 Hz, 3H), 1.34 (s, 3H), 0.61 (s, 3H).20535632.21H NMR (400 MHz, DMSO) δ 8.79 (d, J = 5.1 Hz, 1H), 8.36 (s, 1H), 7.98- 7.89 (m, 2H), 7.83-7.72 (m, 2H), 7.65-7.67 (m, 1H), 7.60-7.48 (m, 2H), 5.64 (s, 2H), 5.04-5.02 (m, 1H), 4.63-4.49 (m, 2H), 4.48-4.37 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H).20635632.21H NMR (400 MHz, DMSO) δ 8.82-8.71 (m, 1H), 8.36 (s, 1H), 7.98- 7.85 (m, 2H), 7.83-7.72 (m, 2H), 7.60-7.43 (m, 2H), 7.08 (d, J = 5.8 Hz, 1H), 5.66 (d, J = 13.7 Hz, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.63-4.49 (m, 2H), 4.48-4.37 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (d, J = 2.8 Hz, 3H), 0.62 (d, J = 2.7 Hz, 3H).20735645.21H NMR (400 MHz, DMSO) δ 13.06 (s, 1H), 8.35 (s, 1H), 7.96-7.89 (m, 1H), 7.85 (dd, J = 10.5, 8.1 Hz, 1H), 7.74 (dd, J = 3.5, 1.6 Hz, 2H), 7.52 (dd, J = 11.2, 1.2 Hz, 1H), 7.34- 7.24 (m, 3H), 5.61 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.55-4.38 (m, 3H), 4.34 (d, J = 16.8 Hz, 1H), 3.80- 3.69 (m, 2H), 2.26 (s, 3H), 1.31 (s, 3H), 0.60 (s, 3H).20835627.21H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.94- 7.86 (m, 2H), 7.75-7.70 (m, 2H), 7.52 (d, 1H), 7.34-7.22 (m, 3H), 6.96 (d, 1H), 5.53 (s, 2H), 5.02 (d, 1H),4.53- 4.41 (m, 3H), 4.34 (d, 1H), 3.77-3.69 (dd, 2H), 2.26 (s, 3H), 1.31 (s, 3H), 0.59 (s, 3H).20922567.3781H NMR (400 MHz, MeOD) δ 8.81 (d, J = 1.4 Hz, 1H), 8.22 (dd, J = 8.6, 1.4 Hz, 1H), 7.97-7.86 (m, 2H), 7.86-7.79 (m, 2H), 7.73 (t, J = 7.6 Hz, 1H), 7.61 (dd, J = 9.7, 1.5 Hz, 1H), 7.57 (dd, J = 7.7, 1.7 Hz, 2H), 7.50 (t, J = 7.9 Hz, 1H), 6.92 (d, J = 8.3 Hz, 1H), 5.65 (s, 2H), 4.78 (d, J = 2.6 Hz, 2H), 4.48 (t, J = 8.8 Hz, 1H), 4.40-4.32 (m, 1H), 4.04 (dd, J = 11.0, 7.5 Hz,1H), 3.80 (td, J = 9.7, 6.9Hz, 1H), 2.54 (dt, J = 14.3,7.3 Hz, 1H), 2.35-2.20 (m,1H).21022567.2871H NMR (400 MHz, MeOD) δ 8.84 (d, J = 4.2 Hz, 1H), 8.25 (dd, J = 8.5, 4.2 Hz, 1H), 8.01-7.87 (m, 2H), 7.82 (t, J = 7.7 Hz, 2H), 7.73 (t, J = 7.6 Hz, 1H), 7.64- 7.49 (m, 4H), 6.93 (d, J = 8.2 Hz, 1H), 5.65 (s, 2H), 4.81 (s, 2H), 4.53-4.45 (m, 1H), 4.38 (d, J = 11.1Hz, 1H), 4.06 (dd, J = 11.1,7.5 Hz, 1H), 3.81 (td, J = 9.7, 6.7 Hz, 1H), 2.56 (q, J = 12.0, 10.2 Hz, 1H), 2.29 (d, J = 13.8 Hz, 1H).21135619.31H NMR (400 MHz, DMSO-d6) δ 8.73 (d, J = 5.0 Hz, 1H), 8.35 (s, 1H), 7.56- 7.39 (m, 7H), 7.38 (d, J = 7.5 Hz, 1H), 5.46 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.57-4.31 (m, 4H), 3.75 (q, J = 8.7 Hz, 2H), 2.36 (s, 3H), 1.32 (s, 3H), 0.61 (s, 3H).212356351H NMR (400 MHz, DMSO-d6) δ 8.74 (d, J = 5.1 Hz, 1H), 8.36 (s, 1H), 7.63 (s, 1H), 7.55-7.43 (m, 6H), 7.41 (s, 1H), 5.46 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.59 (d, J = 17.1 Hz, 1H), 4.52 (d, J = 11.1 Hz, 1H), 4.47-4.36 (m, 2H), 3.85- 3.68 (m, 2H), 2.37 (s, 3H), 1.34 (s, 3H), 0.66 (s, 3H).213296861H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J = 1.3 Hz, 1H), 8.86 (s, 1H), 8.33 (s, 1H), 8.11 (d, J = 1.3 Hz, 1H), 8.04 (d, J = 1.2 Hz, 1H), 7.94 (dd, J = 10.2, 8.2 Hz, 1H), 7.77 (dd, J = 10.4, 5.7 Hz, 1H), 7.69-7.60 (m, 1H), 7.54-7.46 (m, 1H), 5.77 (s, 2H), 4.68 (t, J = 5.1 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J = 4.9 Hz, 2H), 3.25 (s, 3H).214356321H NMR (400 MHz, DMSO-d6) δ 8.91 (s, 1H), 8.83 (s, 1H), 8.31 (s, 1H), 8.22 (s, 1H), 8.03 (s, 1H), 7.95-7.84 (m, 2H), 7.80 (d, J = 8.7 Hz, 1H), 7.62 (s, 1H), 7.55 (d, J = 7.7 Hz, 1H), 7.40 (dd, J = 11.6, 6.2 Hz, 1H), 7.01 (d, J = 8.4 Hz, 1H), 5.67 (s, 2H), 4.60 (s, 2H), 4.46 (s, 2H), 3.69 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H).215356501H NMR (400 MHz, DMSO-d6) δ 8.91 (d, J = 1.3 Hz, 1H), 8.85 (s, 1H), 8.33 (s, 1H), 8.22 (s, 1H), 8.04 (d, J = 1.3 Hz, 1H), 7.93- 7.83 (m, 2H), 7.83-7.78 (m, 1H), 7.61 (d, J = 8.4 Hz, 1H), 7.57 (d, J = 8.9 Hz, 1H), 7.41 (dd, J = 10.9, 6.5 Hz, 1H), 5.76 (s, 2H), 4.60 (d, J = 5.7 Hz, 2H), 4.46 (s, 2H), 3.69 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H).21656151H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.98-7.85 (m, 2H), 7.85- 7.70 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.37 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.53 (d, J = 7.6 Hz, 1H), 4.55 (s, 2H), 4.50 (d, J = 10.8 Hz, 1H), 4.21 (dd, J = 10.9, 6.5 Hz, 1H), 4.06 (t, J = 8.7 Hz, 1H), 3.80 (t, J = 8.3 Hz, 1H),3.18-3.09 (m, 1H), 3.00 (q,J = 7.8 Hz, 1H), 2.75 (t, J =9.5 Hz, 1H).21756151H NMR (400 MHz, DMSO-d6) δ 8.42 (s, 1H), 7.96-7.86 (m, 2H), 7.84 (dd, J = 8.5, 1.5 Hz, 1H), 7.77 (tt, J = 8.8, 4.8 Hz, 3H), 7.65 (d, J = 8.5 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.39 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.26 (s, 1H), 4.65-4.46 (m, 2H), 4.37 (t, J = 8.5 Hz, 1H), 4.21 (dd, J = 10.5, 3.2 Hz, 1H),4.08 (dd, J = 10.4, 7.9 Hz,1H), 3.62-3.46 (m, 3H),2.76 (d, J = 7.3 Hz, 1H).218356181H NMR (400 MHz, DMSO-d6) δ 8.74 (d, J = 5.0 Hz, 1H), 8.50 (s, 1H), 7.81 (dd, J = 8.4, 1.5 Hz, 1H), 7.62 (t, J = 4.2 Hz, 2H), 7.55- 7.42 (m, 5H), 7.41 (s, 1H), 5.46 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.56 (dd, J = 22.6, 14.0 Hz, 2H), 4.49-4.35 (m, 2H), 3.81 (d, J = 8.7 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 2.36 (s, 3H), 1.34 (s, 3H), 0.66 (s, 3H).219356011H NMR (400 MHz, DMSO-d6) δ 8.73 (d, J = 5.1 Hz, 1H), 8.49 (s, 1H), 7.81 (dd, J = 8.5, 1.5 Hz, 1H), 7.62 (d, J = 8.5 Hz, 1H), 7.54-7.45 (m, 4H), 7.44- 7.33 (m, 3H), 5.45 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.58-4.29 (m, 4H), 3.82- 3.67 (m, 2H), 2.36 (s, 3H), 1.32 (s, 3H), 0.60 (s, 3H).220356381H NMR (400 MHz, DMSO-d6) δ 8.39 (s, 1H), 7.92-7.79 (m, 2H), 7.61 (t, J = 8.2 Hz, 1H), 7.57-7.45 (m, 3H), 7.43-7.26 (m, 2H), 6.95 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.24 (s, 1H), 4.42 (s, 2H), 4.35- 4.23 (m, 2H), 4.03 (d, J = 8.9 Hz, 1H), 3.90 (d, J = 8.9 Hz, 1H), 1.00-0.44 (m, 3H), −0.11 (s, 1H).221356161H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J = 1.2 Hz, 1H), 8.76 (d, J = 2.1 Hz, 1H), 8.25 (dd, J = 8.3, 2.2 Hz, 1H), 8.17 (d, J = 8.4 Hz, 1H), 8.10 (d, J = 1.3 Hz, 1H), 8.00 (d, J = 1.2 Hz, 1H), 7.95-7.80 (m, 2H), 7.57-7.47 (m, 2H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.63 (s, 2H), 4.62 (t, J = 5.1 Hz, 2H), 4.47 (s, 2H), 3.68 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H).222356331H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J = 1.2 Hz, 1H), 8.11 (d, J = 1.3 Hz, 1H), 8.04-7.89 (m, 4H), 7.73 (d, J = 8.5 Hz, 3H), 7.63-7.55 (m, 1H), 7.53- 7.44 (m, 1H), 7.05 (d, J = 8.3 Hz, 1H), 5.58 (s, 2H), 4.68 (t, J = 5.2 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J = 4.9 Hz, 2H), 3.24 (s, 3H).223356511H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J = 1.2 Hz, 1H), 8.11 (d, J = 1.3 Hz, 1H), 8.02-7.86 (m, 4H), 7.73 (dd, J = 18.0, 8.6 Hz, 3H), 7.65-7.56 (m, 1H), 7.50 (dd, J = 11.4, 1.3 Hz, 1H), 5.67 (s, 2H), 4.68 (t, J = 5.0 Hz, 2H), 4.54 (s, 2H), 3.73 (t, J = 5.0 Hz, 2H), 3.24 (s, 3H).224356471H NMR (400 MHz, DMSO-d6) δ 8.39 (s, 1H), 8.01-7.93 (m, 1H), 7.87 (dd, J = 10.2, 8.2 Hz, 1H), 7.84-7.70 (m, 3H), 7.54 (dd, J = 14.2, 10.3 Hz, 2H), 7.37(dd, J = 11.2, 6.3 Hz, 1H), 5.70 (s, 2H), 5.24 (s, 1H),4.41 (s, 2H), 4.35- 4.24 (m, 2H), 4.02 (d, J = 8.9 Hz, 1H), 3.90 (d, J = 8.9 Hz, 1H), 0.93-0.50 (m, 3H), −0.11 (s, 1H).225356211H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 1.2 Hz, 1H), 7.96-7.91 (m, 1H), 7.85 (dd, J = 10.5, 8.1 Hz, 1H), 7.74 (d, J = 4.7 Hz, 2H), 7.55-7.47 (m, 1H), 7.43 (s, 1H), 7.31 (d, J = 9.4 Hz, 2H), 5.61 (s, 2H), 4.61 (t, J = 5.1 Hz, 2H), 4.47 (s, 2H), 3.69 (t, J = 5.0 Hz, 2H), 3.22 (s, 3H), 2.25 (s, 3H).226356681H NMR (400 MHz, DMSO-d6) δ 8.91 (d, J = 1.3 Hz, 1H), 8.84 (s, 1H), 8.31 (s, 1H), 8.11 (d, J = 1.3 Hz, 1H), 8.03 (d, J = 1.3 Hz, 1H), 7.96 (t, J = 7.9 Hz, 1H), 7.84-7.74 (m, 1H), 7.61 (d, J = 7.3 Hz, 1H), 7.50 (d, J = 11.4 Hz, 1H), 7.06 (d, J = 8.3 Hz, 1H), 5.68 (s, 2H), 4.68 (t, J = 5.1 Hz, 2H), 4.56 (s, 2H), 3.73 (t, J = 5.0 Hz, 2H), 3.25 (s, 3H).227356031H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 1.3 Hz, 1H), 7.95-7.84 (m, 2H), 7.72 (d, J = 5.8 Hz, 2H), 7.50 (dd, J = 11.4, 1.3 Hz, 1H), 7.43 (s, 1H), 7.29 (s, 1H), 7.27 (d, J = 7.3 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.52 (s, 2H), 4.60 (t, J = 5.1 Hz, 2H), 4.47 (s, 2H), 3.68 (t, J = 5.0 Hz, 2H), 3.22 (s, 3H), 2.25 (s, 3H).22835640.11H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.83 (ddd, J = 20.6, 10.3, 7.3 Hz, 2H), 7.59-7.50 (m, 4H), 7.50-7.43 (m, 3H), 5.57 (s, 2H), 5.04 (d, J = 6.8 Hz, 1H), 4.64-4.49 (m, 2H), 4.49-4.33 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H).22935641.61H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J = 5.1 Hz, 1H), 8.36 (s, 1H), 7.90- 7.79 (m, 1H), 7.69 (dd, J = 5.1, 1.7 Hz, 1H), 7.60-7.41 (m, 5H), 5.52 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.70 (d, J = 17.4 Hz, 1H), 4.56 (d, J = 11.5 Hz, 1H), 4.52-4.31 (m, 2H), 3.76 (s, 2H), 1.40 (s, 3H), 0.67 (s, 3H).23035623.61H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J = 5.1 Hz, 1H), 8.36 (s, 1H), 7.95 (dd, J = 10.2, 6.2 Hz, 1H), 7.63 (dd, J = 5.3, 1.8 Hz, 1H), 7.59-7.49 (m, 4H), 7.47 (d, J = 8.4 Hz, 2H), 5.51 (s, 2H), 5.04 (d, J = 6.5 Hz, 1H), 4.56 (dd, J = 22.4, 14.1 Hz, 2H), 4.49-4.35 (m, 2H), 3.75 (q, J = 8.6 Hz, 2H), 1.34 (s, 3H), 0.62 (s, 3H).23135606.61H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J = 5.2 Hz, 1H), 8.48 (s, 1H), 7.93 (dd, J = 10.2, 6.2 Hz, 1H), 7.79 (dd, J = 8.5, 1.4 Hz, 1H), 7.65-7.59 (m, 2H), 7.59-7.51 (m, 3H), 7.47 (d, J = 8.5 Hz, 2H), 5.51 (s, 2H), 5.01 (d, J = 6.5 Hz, 1H), 4.61-4.49 (m, 2H), 4.49-4.34 (m, 2H), 3.76 (q, J = 8.6 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H).23235626.61H NMR (400 MHz, DMSO-d6) δ 8.11 (d, J = 1.3 Hz, 1H), 7.97-7.86 (m, 2H), 7.83-7.74 (m, 2H), 7.64 (ddt, J = 16.9, 8.4, 1.7 Hz, 2H), 7.50 (dd, J = 11.4, 1.3 Hz, 1H), 5.70 (s, 2H), 4.68 (t, J = 5.1 Hz, 2H), 4.54 (s, 2H), 3.73 (t, J = 5.0 Hz, 2H), 3.24 (s, 3H).23335 with Chiral separation peak 2634.61H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.97-7.87 (m, 2H), 7.84- 7.71 (m, 4H),7.61 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.39 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.82-5.40 (m, 3H), 4.62-4.49 (m, 2H), 4.40 (d, J = 17.0 Hz, 1H), 4.29-4.13 (m, 2H), 4.09 (t, J = 9.1 Hz, 1H), 3.58- 3.49 (m, 2H).23435 with Chiral separation peak 1634.61H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.97-7.87 (m, 2H), 7.84- 7.71 (m, 4H),7.61 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.39 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.82-5.40 (m, 3H), 4.62-4.49 (m, 2H), 4.40 (d, J = 17.0 Hz, 1H), 4.29-4.13 (m, 2H), 4.09 (t, J = 9.1 Hz, 1H), 3.58- 3.49 (m, 2H).23535673.31H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.2 Hz, 1H), 8.35 (s, 1H), 8.01 (d, J = 1.2 Hz, 1H), 7.99- 7.90 (m, 3H), 7.90-7.79 (m, 3H), 7.72 (dd, J = 8.3, 2.8 Hz, 1H), 7.56-7.46 (m, 2H), 5.71 (s, 2H), 5.01 (d, J = 6.5 Hz, 1H), 4.59-4.46 (m, 2H), 4.46-4.34 (m, 2H), 3.82-3.69 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H).23635655.41H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.50 (s, 1H), 8.01 (d, J = 1.2 Hz, 1H), 7.99- 7.90 (m, 3H), 7.90-7.81 (m, 4H), 7.72 (dd, J = 8.2, 2.7 Hz, 1H), 7.63 (d, J = 8.5 Hz, 1H), 7.51 (t, J = 8.1 Hz, 1H), 5.71 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.63-4.49 (m, 2H), 4.49-4.37 (m, 2H), 3.83-3.67 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H).23735603.41H NMR (400 MHz, DMSO-d6) δ 8.51 (s, 1H), 8.29 (s, 1H), 7.93-7.79 (m, 3H), 7.63 (dd, J = 11.7, 9.4 Hz, 2H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.46 (dd, J = 11.5, 6.0 Hz, 1H), 6.94 (d, J = 8.3 Hz, 1H), 5.56 (d, J = 1.8 Hz, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.61-4.50 (m, 2H), 4.49-4.36 (m, 2H), 3.86-3.57 (m, 2H), 2.34 (s, 3H), 1.33 (s, 3H), 0.61 (s, 3H).2383633.01H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J = 1.3 Hz, 1H), 7.99-7.83 (m, 2H), 7.83-7.68 (m, 3H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.49 (dd, J = 11.2, 1.3 Hz, 1H), 7.31 (dd, J = 11.5, 6.1Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.73-5.47 (m, 3H), 4.53 (s, 2H), 4.40 (dd, J = 10.7, 3.4 Hz, 1H), 4.17- 4.09 (m, 2H), 4.01 (dd, J = 10.6, 8.2 Hz, 1H), 3.78 (dd, J = 10.5, 4.6 Hz, 1H), 2.87 (s, 3H).2393633.01H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J = 1.3 Hz, 1H), 7.99-7.83 (m, 2H), 7.83-7.68 (m, 3H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.49 (dd, J = 11.2, 1.3 Hz, 1H), 7.31 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.73 -5.47 (m, 3H), 4.53 (s, 2H), 4.40 (dd, J = 10.7, 3.4 Hz, 1H), 4.17- 4.09 (m, 2H), 4.01 (dd, J = 10.6, 8.2 Hz, 1H), 3.78 (dd, J = 10.5, 4.6 Hz, 1H), 2.87 (s, 3H).24035706.61H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.94-7.78 (m, 2H), 7.55- 7.42 (m, 5H), 7.35 (d, J = 7.8 Hz, 1H), 6.94 (d, J = 8.3 Hz, 1H), 5.49 (s, 2H), 5.04 (d, J = 6.5 Hz, 1H), 4.64 (d, J = 3.4 Hz, 4H), 4.60-4.34 (m, 4H), 3.75 (q, J = 8.7 Hz, 2H), 2.96 (s, 3H), 1.34 (s, 3H), 0.62 (s, 3H).24135690.61H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.2 Hz, 1H), 8.36 (s, 1H), 8.01 (d, J = 1.2 Hz, 1H), 7.97- 7.89 (m, 2H), 7.89-7.74 (m, 3H), 7.64-7.58 (m, 1H), 7.51 (dd, J = 11.2, 1.2 Hz, 1H), 7.04 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.11 (d, J = 6.4 Hz, 1H), 4.67 (d, J = 17.4 Hz, 1H), 4.56 (d, J = 11.3 Hz, 1H), 4.46 (dd, J = 11.3, 6.6 Hz, 1H), 4.38 (d, J = 17.5 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H).24235659.51H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.93 (t, J = 7.9 Hz, 1H), 7.76 (ddd, J = 10.4, 5.7, 2.0 Hz, 1H), 7.65-7.57 (m, 2H), 7.51 (ddd, J = 10.0, 5.3, 1.6 Hz, 2H), 7.33 (dd, J = 8.3, 2.0 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.52 (s, 2H), 5.11 (d, J = 6.4 Hz, 1H), 4.67 (d, J = 17.4 Hz, 1H), 4.56 (d, J = 11.3 Hz, 1H), 4.46 (dd, J = 11.3, 6.7 Hz, 1H), 4.38 (d, J = 17.4 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H).24335641.61H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.93 (t, J = 7.9 Hz, 1H), 7.73 (ddd, J = 10.7, 5.6, 1.9 Hz, 1H), 7.57 (dd, J = 7.4, 1.7 Hz, 1H), 7.55-7.49 (m, 3H), 7.46 (d, J = 8.5 Hz, 2H), 7.01 (d, J = 8.3 Hz, 1H), 5.49 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.67 (d, J = 17.4 Hz, 1H), 4.56 (d, J = 11.2 Hz, 1H), 4.46 (dd, J = 11.3, 6.6 Hz, 1H), 4.37 (d, J = 17.4 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H).24435630.61H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.96 (d, J = 7.7 Hz, 1H), 7.92-7.85 (m, 2H), 7.74- 7.62 (m, 3H), 7.61-7.56 (m, 1H), 7.54-7.46 (m, 1H), 7.06 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.66 (d, J = 17.4 Hz, 1H), 4.55 (d, J = 11.2 Hz, 1H), 4.46 (dd, J = 11.3, 6.6 Hz, 1H), 4.37 (d, J = 17.4 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.66 (s, 3H).24535612.61H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.95 (t, J = 7.9 Hz, 1H), 7.88 (d, J = 8.2 Hz, 2H), 7.78 (dd, J = 8.4, 1.4 Hz, 1H), 7.69 (d, J = 8.0 Hz, 2H), 7.68-7.61 (m, 1H), 7.58 (dd, J = 8.2, 2.9 Hz, 2H), 7.07 (d, J = 8.3 Hz, 1H), 5.59 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.64 (d, J = 17.4 Hz, 1H), 4.57 (d, J = 11.1 Hz, 1H), 4.47 (dd, J = 11.2, 6.8 Hz, 1H), 4.35 (d,J = 17.3 Hz, 1H), 3.82-3.72(m, 2H), 1.39 (s, 3H), 0.66(s, 3H).24635623.61H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.93 (t, J = 7.9 Hz, 1H), 7.78 (dd, J = 8.4, 1.5 Hz, 1H), 7.71 (ddd, J = 10.4, 5.6,2.0 Hz, 1H), 7.63-7.50 (m, 4H), 7.46 (d, J = 8.4 Hz, 2H), 7.02 (d, J = 8.3 Hz, 1H), 5.49 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.64 (d, J = 17.3 Hz, 1H), 4.57 (d, J = 11.1 Hz, 1H), 4.47 (dd, J = 11.2, 6.7 Hz, 1H), 4.35 (d,J = 17.3 Hz, 1H), 3.83-3.73(m, 2H), 1.39 (s, 3H), 0.66(s, 3H).24735641.61H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 7.93 (t, J = 7.9 Hz, 1H), 7.79 (dd, J = 8.5, 1.4 Hz, 1H), 7.77-7.71 (m, 1H), 7.65- 7.56 (m, 3H), 7.50 (dd, J = 10.0, 2.0 Hz, 1H), 7.34 (dd, J = 8.3, 2.0 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.52 (s, 2H), 5.10 (d, J = 6.6 Hz, 1H), 4.66 (d, J = 17.3 Hz, 1H), 4.58 (d, J = 11.1 Hz, 1H), 4.47 (dd, J = 11.2, 6.7Hz, 1H), 4.37 (d, J = 17.3Hz, 1H), 3.84-3.69 (m,2H), 1.39 (s, 3H), 0.66 (s,3H).24835688.61H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.92-7.78 (m, 3H), 7.63 (d, J = 8.5 Hz, 1H), 7.56-7.41 (m, 4H), 7.35 (d, J = 7.8 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.48 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.64 (q, J = 2.5 Hz, 4H), 4.58-4.51 (m, 2H), 4.49-4.36 (m, 2H), 3.84-3.68 (m, 2H), 2.96 (s, 3H), 1.34 (s, 3H), 0.62 (s, 3H).24927624.61H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 8.18 (dd, J = 9.6, 7.8 Hz, 1H), 7.90 (t, J = 7.9 Hz, 1H), 7.87-7.78 (m, 2H), 7.63 (d, J = 8.5 Hz, 1H), 7.55 (dd, J = 7.8, 2.5 Hz, 2H), 7.47 (dd, J = 11.2, 6.3 Hz, 1H), 6.98 (d, J = 8.3 Hz, 1H), 5.53 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.59-4.50 (m, 2H), 4.49-4.35 (m, 2H), 3.83- 3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).25027650.61H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.11 (d, J = 1.3 Hz, 1H), 8.03-7.90 (m, 3H), 7.89-7.73 (m, 3H), 7.59 (d, J = 7.3 Hz, 1H), 7.50 (d, J = 11.3 Hz, 1H), 7.03 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 4.68 (t, J = 5.1 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J = 4.9 Hz, 2H), 3.24 (s, 3H).25127617.61H NMR (400 MHz, DMSO-d6) δ 8.11 (d, J = 1.3 Hz, 1H), 7.93 (t, J = 7.9 Hz, 1H), 7.77-7.69 (m, 1H), 7.64-7.56 (m, 2H), 7.54- 7.46 (m, 2H), 7.33 (dd, J = 8.2, 2.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.52 (s, 2H), 4.68 (t, J = 5.1 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J = 5.0 Hz, 2H), 3.24 (s, 3H).25237602.61H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.99 (dd, J = 10.1, 7.5 Hz, 1H), 7.94-7.78 (m, 3H), 7.64 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.47 (dd, J = 11.3, 6.2 Hz, 1H), 7.24 (dd, J = 7.6, 1.9 Hz, 1H), 6.96 (d, J = 8.2 Hz, 1H), 5.48 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.62- 4.51 (m, 2H), 4.51-4.35 (m, 2H), 3.81-3.69 (m, 2H),2.43 (s, 3H), 1.34 (s, 3H), 0.61 (s, 3H).25327633.61H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J = 1.3 Hz, 1H), 8.76 (d, J = 2.2 Hz, 1H), 8.26 (dd, J = 8.4, 2.2 Hz, 1H), 8.18 (d, J = 8.4 Hz, 1H), 8.11 (d, J = 1.3 Hz, 1H), 8.03-7.91 (m, 2H), 7.81-7.72 (m, 1H), 7.59 (dd, J = 7.5, 1.8 Hz, 1H), 7.54-7.46 (m, 1H), 7.05 (d, J = 8.3 Hz, 1H),5.63 (s, 2H), 4.68 (t, J = 5.1 Hz, 2H), 4.55 (s, 2H), 3.73 (t, J = 5.0 Hz, 2H), 3.25 (s, 3H).25435638.21H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J = 1.2 Hz, 1H), 8.76 (d, J = 2.1 Hz, 1H), 8.49 (s, 1H), 8.26 (dd, J = 8.4, 2.2 Hz, 1H), 8.17 (d, J = 8.4 Hz, 1H), 8.00 (d, J = 1.2 Hz, 1H), 7.96-7.85 (m, 2H), 7.81 (dd, J = 8.4, 1.4 Hz, 1H), 7.62 (d, J = 8.5 Hz, 1H), 7.55 (d, J = 7.4 Hz, 1H), 7.47 (dd, J = 10.4, 7.2 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.63 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.59-4.49(m, 2H), 4.49-4.30 (m,2H), 3.78 (d, J = 8.7 Hz,1H), 3.73 (d, J = 8.6 Hz,1H), 1.34 (s, 3H), 0.61 (s,3H).255355981H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J = 1.3 Hz, 1H), 7.98 (d, J = 7.2 Hz, 1H), 7.88 (t, J = 7.9 Hz, 1H), 7.55-7.48 (m, 4H), 7.48- 7.42 (m, 3H), 6.97 (d, J = 8.2 Hz, 1H), 5.46 (s, 2H), 4.63 (t, J = 5.1 Hz, 2H), 4.52 (s, 2H), 3.70 (t, J = 5.0 Hz, 2H), 3.22 (s, 3H).256356331H NMR (400 MHz, DMSO-d6) δ 8.27 (d, J = 1.3 Hz, 1H), 7.95-7.86 (m, 2H), 7.75 (tdd, J = 9.4, 7.2, 3.0 Hz, 3H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.48 (dd, J = 11.2, 1.3 Hz, 1H), 7.31 (dd, J = 11.5, 6.1 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.64-5.51 (m, 3H), 4.53 (s, 2H), 4.40 (dd, J = 10.6, 3.4 Hz, 1H), 4.13 (td, J = 8.8, 7.6, 3.7 Hz, 2H),4.00 (dd, J = 10.6, 8.2 Hz, 1H), 3.78 (dd, J = 10.4, 4.6 Hz, 1H), 2.87 (s, 3H).257356291H NMR (400 MHz, DMSO-d6) δ 8.51 (s, 1H), 7.97-7.87 (m, 3H), 7.82 (dd, J = 8.5, 1.5 Hz, 1H), 7.78-7.70 (m, 2H), 7.63 (d, J = 8.5 Hz, 1H), 7.58-7.45 (m, 2H), 7.02 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.63 (d, J = 17.1 Hz, 1H), 4.55 (d, J = 11.7 Hz, 1H), 4.49-4.36 (m, 2H), 3.81 (d, J = 8.6 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H).258356631H NMR (400 MHz, DMSO-d6) δ 8.10 (s, 1H), 8.01 (d, J = 7.1 Hz, 1H), 7.87 (t, J = 7.9 Hz, 1H), 7.56- 7.39 (m, 5H), 7.38-7.30 (m, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.47 (s, 2H), 4.64(t, J = 7.8 Hz, 6H), 4.53 (s, 2H), 3.70 (t, J = 5.1 Hz, 2H), 3.66 (d, J = 2.4 Hz, 3H), 3.22 (s, 3H).259chiral separation Peak 2610.61H NMR (400 MHz, DMSO-d6) δ 8.57 (s, 1H), 7.98-7.83 (m, 3H), 7.81- 7.71 (m, 3H),7.67 (d, J = 8.5 Hz, 1H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.40 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.27 (s, 1H), 4.47 (s, 2H), 4.37-4.23 (m, 2H), 4.05 (d, J = 8.9 Hz, 1H), 3.91 (d, J = 8.9 Hz, 1H), 0.75 (dd, J = 24.6, 17.1 Hz, 3H), −0.07 (s, 1H).260chiral separation Peak 1610.61H NMR (400 MHz, DMSO-d6) δ 8.57 (s, 1H), 7.98-7.83 (m, 3H), 7.81- 7.71 (m, 3H), 7.67 (d, J = 8.5 Hz, 1H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.40 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.27 (s, 1H), 4.47 (s, 2H), 4.37-4.23 (m, 2H), 4.05 (d, J = 8.9 Hz, 1H), 3.91 (d, J = 8.9 Hz, 1H), 0.75 (dd, J = 24.6, 17.1 Hz, 3H), −0.07 (s, 1H).261chiral separation Peak 1635.21H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J = 1.4 Hz, 1H), 7.96-7.82 (m, 3H), 7.82-7.70 (m, 3H), 7.67 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.41 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 6.41 (td, J = 55.8, 4.9 Hz, 1H), 5.65-5.50 (m, 3H), 4.59 (d, J = 17.0 Hz, 1H), 4.49 (t, J = 8.9 Hz, 1H), 4.41 (d, J = 16.9 Hz, 1H), 4.28 (dd, J = 10.5, 3.3 Hz,1H), 4.15 (dd, J = 10.5, 7.9Hz, 1H), 3.81 (t, J = 9.2 Hz,1H), 3.25 (d, J = 14.7 Hz,1H).262chiral separation Peak 2635.21H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J = 1.4 Hz, 1H), 7.96-7.82 (m, 3H), 7.82-7.70 (m, 3H), 7.67 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.41 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 6.41 (td, J = 55.8, 4.9 Hz, 1H), 5.65-5.50 (m, 3H),4.59(d, J = 17.0 Hz, 1H), 4.49 (t, J = 8.9 Hz, 1H), 4.41 (d, J = 16.9 Hz, 1H), 4.28 (dd, J = 10.5, 3.3 Hz,1H), 4.15 (dd, J = 10.5, 7.9Hz, 1H), 3.81 (t, J = 9.2 Hz,1H), 3.25 (d, J = 14.7 Hz,1H).26335687.21H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 7.92-7.76 (m, 2H), 7.57- 7.40 (m, 5H), 7.34 (dd, J = 10.8, 7.9 Hz, 1H), 6.94 (d, J = 8.3 Hz, 1H), 5.48 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.69-4.60 (m, 4H),4.59- 4.49 (m, 2H), 4.48-4.35 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 3.66 (d, J = 3.3 Hz, 3H), 1.34 (s, 3H), 0.62 (s, 3H).26435675.21H NMR (400 MHz, DMSO-d6) δ 8.85 (s, 1H), 8.37 (d, J = 11.1 Hz, 2H), 7.89 (t, J = 7.9 Hz, 1H), 7.60 (dd, J = 10.6, 6.4 Hz, 1H), 7.56-7.48 (m, 2H), 7.44 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.71 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.58-4.48 (m, 2H), 4.46-4.33 (m, 2H), 3.74 (q, J = 8.7 Hz, 2H), 1.32 (s, 3H), 0.60 (s, 3H).26535628.21H NMR (400 MHz, DMSO-d6) δ 8.75 (d, J = 5.1 Hz, 1H), 8.35 (s, 1H), 7.94 (d, J = 10.0 Hz, 1H), 7.80- 7.70 (m, 2H), 7.52 (dd, J = 11.3, 1.2 Hz, 1H), 7.48 (d, J = 5.1 Hz, 1H), 7.42 (d, J = 10.5 Hz, 1H), 7.38 (d, J = 7.5 Hz, 1H), 5.59 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.56-4.46 (m, 2H), 4.42 (dd, J = 11.3, 6.7 Hz, 1H), 4.36 (d, J = 16.9 Hz, 1H),3.77 (d, J = 8.7 Hz, 1H),3.72 (d, J = 8.6 Hz, 1H),2.36 (s, 3H), 1.32 (s, 3H),0.61 (s, 3H).26622599.21H NMR (400 MHz, DMSO-d6) δ 8.39 (d, J = 1.5 Hz, 1H), 7.96-7.81 (m, 3H), 7.81-7.71 (m, 3H), 7.68 (d, J = 8.5 Hz, 1H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.44 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.06 (td, J = 7.6, 3.6 Hz, 1H), 4.56 (d, J = 16.9 Hz, 1H), 4.48 (d, J = 16.8 Hz, 1H), 4.37 (t, J = 8.2 Hz, 1H), 4.22 (dd, J = 10.5,3.6 Hz, 1H), 4.13 (dd, J =10.5, 8.1 Hz, 1H), 3.38 (t, J =9.4 Hz, 1H), 2.68 (dq, J =9.7, 6.7 Hz, 1H), 1.06 (d, J =6.7 Hz, 3H).26722635.21H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.97-7.87 (m, 2H), 7.84- 7.67 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.4, 1.7 Hz, 1H), 7.39 (dd, J = 11.4, 6.1Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.80-5.56 (m, 3H), 4.55 (d, J = 12.6 Hz, 2H), 4.39 (d, J = 17.0 Hz, 1H), 4.27-4.12 (m, 2H), 4.09 (t, J = 9.2 Hz, 1H), 3.62-3.39 (m, 2H).26822611.21H NMR (400 MHz, DMSO-d6) δ 8.57 (s, 1H), 7.98-7.83 (m, 3H), 7.81- 7.71 (m, 3H), 7.67 (d, J = 8.5 Hz, 1H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.40 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.27 (s, 1H), 4.47 (s, 2H), 4.37-4.23 (m, 2H), 4.05 (d, J = 8.9 Hz, 1H), 3.91 (d, J = 8.9 Hz, 1H), 0.75 (dd, J = 24.6, 17.1 Hz, 3H), −0.07 (s, 1H).26922635.21H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J = 1.4 Hz, 1H), 7.96-7.82 (m, 3H), 7.82-7.70 (m, 3H), 7.67 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.41 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 6.41 (td, J = 55.8, 4.9 Hz, 1H), 5.65-5.50 (m, 3H), 4.59 (d, J = 17.0 Hz, 1H), 4.49 (t, J = 8.9 Hz, 1H), 4.41 (d, J = 16.9 Hz, 1H),4.28 (dd, J = 10.5, 3.3 Hz,1H), 4.15 (dd, J = 10.5, 7.9Hz, 1H), 3.81 (t, J = 9.2 Hz,1H), 3.25 (d, J = 14.7 Hz,1H).27022611.21H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 1.4 Hz, 1H), 7.95-7.87 (m, 2H), 7.84-7.74 (m, 4H), 7.64 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.38 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 4.39 (s, 2H), 3.99 (s, 2H), 2.63- 2.55 (m, 2H), 1.51 (s, 3H).2713614.21H NMR (400 MHz, DMSO-d6) δ 8.40 (s, 3H), 8.37 (s, 1H), 7.96-7.88 (m, 2H), 7.84 (dd, J = 8.5, 1.4 Hz, 1H), 7.79-7.71 (m, 3H), 7.66 (d, J = 8.4 Hz, 1H), 7.56-7.50 (m, 1H), 7.46 (dd, J = 11.6, 6.1 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 4.80 (s, 2H), 4.41 (s, 2H), 4.00 (q, J = 7.9 Hz, 1H), 3.97-3.88 (m, 1H), 3.88- 3.79 (m, 2H), 2.37- 2.12 (m, 2H).2725641.21H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J = 1.5 Hz, 1H), 7.94-7.88 (m, 2H), 7.84 (dd, J = 8.5, 1.5 Hz, 1H), 7.80-7.70 (m, 3H), 7.63 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.6, 1.7 Hz, 1H), 7.39 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.86- 4.65 (m, 2H), 4.52 (d, J = 4.7 Hz, 3H), 3.98 (d, J = 9.3 Hz, 1H), 3.91 (d, J = 9.3 Hz, 1H), 2.13 (dd, J = 11.0, 6.4 Hz, 1H), 1.64-1.38 (m, 4H).2735639.21H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J = 1.5 Hz, 1H), 7.95-7.83 (m, 3H), 7.80-7.71 (m, 3H), 7.64 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.43 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.62 (s, 2H), 4.57 (s, 2H), 3.75- 3.58 (m, 2H), 1.99-1.86 (m, 2H), 1.86-1.72 (m, 1H), 1.66-1.51 (m, 1H), 1.12 (qd, J = 8.1, 5.2 Hz, 1H), 0.31 (dq, J = 9.1, 4.4, 3.9 Hz, 1H), 0.26-0.15 (m, 1H), 0.10-−0.12 (m, 2H).2745625.21H NMR (400 MHz, DMSO-d6) δ 8.32 (d, J = 1.5 Hz, 1H), 7.95-7.83 (m, 3H), 7.80-7.71 (m, 3H), 7.65 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.43 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H),4.67 (dd, J = 15.2, 2.9 Hz, 1H), 4.62- 4.46 (m, 3H), 4.40 (qd, J = 7.1, 2.9 Hz, 1H), 3.67 (d, J = 8.0 Hz, 1H), 3.51 (d, J = 8.0 Hz, 1H), 2.03 (dd, J = 12.4, 6.6 Hz, 1H), 1.78 (dd, J = 12.3, 7.0 Hz, 1H), 0.70- 0.45 (m, 4H).275chiral separation Peak 2615.21H NMR (400 MHz, DMSO-d6) δ 8.38 (s, 1H), 7.97-7.86 (m, 2H), 7.75 (dt, J = 6.5, 5.2 Hz, 4H), 7.60-7.43 (m, 2H), 7.29 (dd, J = 11.4, 6.1 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.53 (d, J = 10.2 Hz, 1H), 4.49 (s, 2H), 4.42 (dd, J = 10.3, 3.8 Hz, 1H), 4.17-4.06 (m, 2H), 4.02 (dd, J = 10.3, 8.3 Hz, 1H), 3.82 (dd, J = 10.1,4.5 Hz, 1H), 2.87 (s, 3H).276chiral separation Peak 1615.21H NMR (400 MHz, DMSO-d6) δ 8.39 (d, J = 1.5 Hz, 1H), 7.98-7.84 (m, 2H), 7.80-7.68 (m, 4H), 7.58 (d, J = 8.4 Hz, 1H), 7.53 (d, J = 6.8 Hz, 1H), 7.29 (dd, J = 11.6, 6.0 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 5.54 (t, J = 9.6 Hz, 1H), 4.50 (s, 2H), 4.42 (dd, J = 10.4, 3.7 Hz, 1H), 4.19-4.08 (m, 2H), 4.02 (t, J = 9.4 Hz, 1H), 3.82(dd, J = 10.3, 4.5 Hz, 1H),2.87 (s, 3H).; 1H NMR (400MHz, DMSO-d6) δ 8.38 (s,1H), 7.97-7.86 (m, 2H),7.75 (dt, J = 6.5, 5.2 Hz,4H), 7.60-7.43 (m, 2H),7.29 (dd, J = 11.4, 6.1 Hz,1H), 6.99 (d, J = 8.2 Hz,1H), 5.60 (s, 2H), 5.53 (d, J =10.2 Hz, 1H), 4.49 (s, 2H),4.42 (dd, J = 10.3, 3.8 Hz,1H), 4.17-4.06 (m, 2H),4.02 (dd, J = 10.3, 8.3 Hz,1H), 3.82 (dd, J = 10.1,4.5Hz, 1H), 2.87 (s, 3H).27735595.21H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.21 (dd, J = 8.6, 1.4 Hz, 1H), 8.06 (d, J = 8.2 Hz, 2H), 7.83 (d, J = 8.6 Hz, 1H), 7.77 (t, J = 7.5 Hz, 1H), 7.68-7.52 (m, 4H), 7.49 (d, J = 8.1 Hz, 2H), 5.73 (s, 2H), 5.10 (d, J = 6.2 Hz, 1H), 4.80-4.62 (m, 2H), 4.62-4.53 (m, 1H), 4.45 (dd, J = 11.6, 6.7 Hz, 1H), 3.99 (d, J = 9.0 Hz, 1H), 3.80 (d, J = 8.9 Hz, 1H), 1.28 (s, 3H), 0.65 (s, 3H).27835609.31H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.21 (dd, J = 8.6, 1.4 Hz, 1H), 7.93-7.78 (m, 2H), 7.70 (t, J = 7.6 Hz, 1H), 7.61- 7.53 (m, 2H), 7.38 (d, J = 7.6 Hz, 1H), 7.25 (d, J = 10.8 Hz, 1H), 7.17 (d, J = 7.4 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.57 (s, 2H), 5.15 (d, J = 6.5 Hz, 1H), 4.79- 4.62 (m, 3H), 4.52 (dd, J = 11.7, 6.6 Hz, 1H), 4.01 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 2.31 (s, 3H), 1.37 (s, 3H), 0.73 (s, 3H).27935627.261H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J = 8.6, 1.4 Hz, 1H), 7.80 (d, J = 8.6 Hz, 1H), 7.73 (t, J = 7.6 Hz, 1H), 7.66 (dd, J = 10.1, 8.1 Hz, 1H), 7.64-7.58 (m, 2H), 131 (d, J = 7.6 Hz, 1H), 7.26 (d, J = 10.8 Hz, 1H), 7.19 (dd, J = 8.1, 2.8 Hz, 1H), 5.64 (s, 2H), 5.13 (d, J = 6.4 Hz, 1H), 4.77-4.59 (m, 3H), 4.51 (dd, J = 11.6,6.7 Hz, 1H), 4.00 (d, J = 9.0Hz, 1H), 3.84 (d, J = 8.9 Hz,1H), 2.31 (s, 3H), 1.37 (s,3H), 0.72 (s, 3H).28035618.251H NMR (400 MHz, Methanol-d4) δ 8.94 (s, 1H), 8.23 (dd, J = 8.6, 1.4 Hz, 1H), 7.83 (t, J = 8.0 Hz, 2H), 7.51 (t, J = 8.3 Hz, 1H), 7.41 (d, J = 7.6 Hz, 1H), 7.30 (d, J = 10.8 Hz, 1H), 7.28- 7.19 (m, 2H), 7.16 (d, J = 7.3 Hz, 1H), 6.91 (d, J = 8.3 Hz, 1H), 5.46 (s, 2H), 5.17 (d, J = 7.0 Hz, 1H), 4.81- 4.58 (m, 3H), 4.52 (dd, J = 11.7, 6.7 Hz, 1H), 4.01 (d,J = 8.9 Hz, 1H), 3.85 (d, J =9.0 Hz, 1H), 2.36 (s, 3H),1.38 (s, 3H), 0.74 (s, 3H).28138573.61H NMR (400 MHz, Methanol-d4) δ 8.67 (s, 1H), 7.95 (dd, J = 8.6, 1.5 Hz, 1H), 7.82-7.64 (m, 3H), 7.59 (d, J = 8.5 Hz, 1H), 5.82 (s, 2H), 5.08 (d, J = 6.8 Hz, 1H), 4.72 (d, J= 11.0 Hz, 1H), 4.68-4.51 (m, 4H), 4.42 (d, J = 17.1 Hz, 1H), 3.99 (d, J = 8.7 Hz, 1H), 3.84 (d, J = 8.7 Hz, 1H), 1.50-1.40 (m, 6H), 0.77 (s, 3H).28235640.51H NMR (400 MHz, Methanol-d4) δ 8.83 (s, 1H), 8.09 (dd, J = 8.5, 1.4 Hz, 1H), 7.73-7.64 (m, 3H), 7.64-7.55 (m, 1H), 7.51 (d, J = 8.4 Hz, 2H), 7.47-7.20 (m, 2H), 5.57 (s, 2H), 5.16 (d, J = 6.6 Hz, 1H), 4.81- 4.63 (m, 2H), 4.57 (dd, J = 11.5, 6.7 Hz, 2H), 3.99 (d, J = 8.8 Hz, 1H), 3.86 (d, J = 8.8 Hz, 1H), 1.48 (s, 3H), 0.81 (s, 3H).28335631.51H NMR (400 MHz, Methanol-d4) δ 8.83 (s, 1H), 8.09 (dd, J = 8.6, 1.4 Hz, 1H), 7.84-7.74 (m, 2H), 7.74-7.66 (m, 4H), 7.66- 7.51 (m, 2H), 5.68 (s, 2H), 5.16 (d, J = 6.6 Hz, 1H), 4.80-4.66 (m, 2H), 4.57 (dd, J = 11.6, 6.7 Hz, 2H), 3.99 (d, J = 8.9 Hz, 1H), 3.86 (d, J = 8.9 Hz, 1H), 1.48 (s, 3H), 0.81 (s, 3H).28435619.21H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.71 (d, J = 5.1 Hz, 1H), 8.19 (dd, J = 8.6, 1.4 Hz, 1H), 7.79 (d, J = 8.6 Hz, 1H), 7.58 (t, J = 8.2 Hz, 1H), 7.44 (dd, J = 14.0, 9.0 Hz, 2H), 7.34 (d, J = 5.1 Hz, 1H), 7.31-7.20 (m, 2H), 5.57 (s, 2H), 5.15 (d, J = 6.2 Hz, 1H), 4.79-4.58 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz,1H), 3.84 (d, J = 8.9 Hz,1H), 2.43 (s, 3H), 1.39 (s,3H), 0.75 (s, 3H).28535657.31H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.73 (d, J = 5.2 Hz, 1H), 8.13 (dd, J = 8.5, 1.4 Hz, 1H), 8.03 (dd, J = 10.4, 6.1 Hz, 1H), 7.82 (t, J = 7.5 Hz, 1H), 7.74 (d, J = 8.6 Hz, 1H), 7.69 (dd, J = 5.2, 1.7 Hz, 1H), 7.56 (d, J = 7.9 Hz, 2H), 7.45 (dd, J = 11.2, 5.9 Hz, 1H), 5.71 (s, 2H), 5.11 (d, J = 6.6 Hz, 1H), 4.79- 4.58 (m, 3H), 4.52 (dd, J =11.5, 6.8 Hz, 1H), 3.99 (d,J = 8.9 Hz, 1H), 3.84 (d, J =8.9 Hz, 1H), 1.42 (s, 3H),0.76 (s, 3H).28627619.41H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J = 8.6, 1.4 Hz, 1H), 7.93-7.80 (m, 3H), 7.78 (d, J = 8.6 Hz, 1H), 7.59 (dd, J = 7.4, 1.6 Hz, 1H), 7.40 (dd, J = 11.2, 6.0 Hz, 1H), 7.27 (d, J = 7.8 Hz, 1H), 6.98 (d, J = 8.2 Hz, 1H), 5.57 (s, 2H), 5.15 (d, J = 6.4 Hz, 1H), 4.75 (d, J = 17.3 Hz, 1H), 4.71-4.62 (m, 2H), 4.53 (dd, J = 11.7,6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.85 (d, J = 8.9 Hz,1H), 2.51 (s, 3H), 1.41 (s,3H), 0.76 (s, 3H).28735641.31H NMR (400 MHz, Methanol-d4) δ 8.56 (s, 1H), 8.43 (d, J = 1.9 Hz, 1H), 7.93-7.74 (m, 3H), 7.68 (dd, J = 11.1, 1.2 Hz, 1H), 7.60-7.50 (m, 1H), 7.21 (dd, J = 11.5, 6.0 Hz, 1H), 6.89 (d, J = 8.2 Hz, 1H), 5.63 (d, J = 1.9 Hz, 2H), 4.95 (d, J = 7.3 Hz, 1H), 4.62-4.44 (m, 4H), 3.94 (d, J = 8.8 Hz, 1H), 3.80 (d, J = 8.8 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H).28835653.51H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 8.29 (s, 1H), 7.88 (dd, J = 10.8, 6.3 Hz, 1H), 7.84- 7.75 (m, 1H), 7.70 (dd, J = 11.0, 1.2 Hz, 1H), 7.58- 7.50 (m, 1H), 7.29-7.23 (m, 1H), 7.22 (d, J = 11.4 Hz, 1H), 6.89 (dd, J = 8.3, 0.7 Hz, 1H), 5.58-5.44 (m, 2H), 4.98 (d, J = 6.7 Hz, 1H), 4.66-4.35 (m, 4H), 4.02 (s, 3H), 3.94 (d, J = 8.8 Hz, 1H), 3.80 (d, J = 8.8 Hz, 1H), 1.36 (s, 3H), 0.68 (s, 3H).).289 Achiral separation Peak 2623.71H NMR (400 MHz, Methanol-d4) δ 8.81 (s, 1H), 8.43 (d, J = 1.9 Hz, 1H), 8.09 (dd, J = 8.6, 1.4 Hz, 1H), 7.95-7.78 (m, 3H), 7.74 (d, J = 8.6 Hz, 1H), 7.58 (dd, J = 7.6, 1.5 Hz, 1H), 7.31 (dd, J = 11.4, 6.0 Hz, 1H), 6.91 (d, J = 8.3 Hz, 1H), 5.63 (d, J = 1.9 Hz, 2H), 5.06 (d, J = 6.6 Hz, 1H), 4.74-4.43 (m, 4H), 3.98 (d, J = 8.8 Hz, 1H), 3.83 (d, J = 8.8 Hz, 1H), 1.40 (s, 3H), 0.72 (s, 3H).289 Bchiral separation Peak 1623.71H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.43 (d, J = 1.9 Hz, 1H), 8.18 (dd, J = 8.6, 1.4 Hz, 1H), 7.91 (dd, J = 10.9, 6.3 Hz, 1H), 7.88-7.74 (m, 3H), 7.59 (dd, J = 7.4, 1.5 Hz, 1H), 7.38 (dd, J = 11.2, 6.1 Hz, 1H), 6.92 (d, J = 8.2 Hz, 1H), 5.63 (d, J = 1.9 Hz, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.80-4.61 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz,1H), 4.00 (d, J = 8.9 Hz,1H), 3.85 (d, J = 8.9 Hz,1H), 1.41 (s, 3H), 0.76 (s,3H).290 Achiral separation Peak 2636.11H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.28 (s, 1H), 8.16 (dd, J = 8.6, 1.4 Hz, 1H), 7.93 (dd, J = 10.8, 6.3 Hz, 1H), 7.85- 7.72 (m, 2H), 7.58 (dd, J = 7.4, 1.5 Hz, 1H), 7.39 (dd, J = 11.2, 6.0 Hz, 1H), 7.18 (s, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 5.13 (d, J = 6.6 Hz, 1H), 4.76-4.58 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (s, 4H), 3.84 (d, J = 8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H).290 Bchiral separation Peak 1636.11H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.28 (s, 1H), 8.16 (dd, J = 8.6, 1.4 Hz, 1H), 7.93 (dd, J = 10.8, 6.3 Hz, 1H), 7.85- 7.72 (m, 2H), 7.58 (dd, J = 7.4, 1.5 Hz, 1H), 7.39 (dd, J = 11.2, 6.0 Hz, 1H), 7.18 (s, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 5.13 (d, J = 6.6 Hz, 1H), 4.76-4.58 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (s, 4H), 3.84 (d, J = 8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H).29137613.41H NMR (400 MHz, Methanol-d4) δ 8.28 (s, 1H), 8.19 (d, J = 1.2 Hz, 1H), 7.86 (dd, J = 10.8, 6.3 Hz, 1H), 7.79 (t, J = 7.9 Hz, 1H), 7.72 (dd, J = 11.1, 1.2 Hz, 1H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.20 (q, J = 6.2 Hz, 2H), 6.88 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 4.63 (t, J = 5.0 Hz, 2H), 4.57 (s, 2H), 4.01 (s, 3H), 3.75 (t, J = 4.9 Hz, 2H), 3.28 (s, 3H).29222609.31H NMR (400 MHz, Methanol-d4) δ 8.15 (d, J = 1.3 Hz, 1H), 7.86 (t, J = 7.9 Hz, 1H), 7.75 (t, J = 7.5 Hz, 1H), 7.70-7.56 (m, 5H), 6.97 (d, J = 8.3 Hz, 1H), 5.64 (s, 2H), 4.67 (t, J = 5.0 Hz, 2H), 4.60 (s, 2H), 3.81 (t, J = 4.9 Hz, 2H).29322631.31H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.11 (dd, J = 8.6, 1.4 Hz, 1H), 7.89 (t, J = 7.9 Hz, 1H), 7.82-7.66 (m, 3H), 7.66- 7.50 (m, 3H), 7.01 (d, J = 8.3 Hz, 1H), 5.64 (s, 2H), 5.17 (d, J = 6.5 Hz, 1H), 4.85-4.50 (m, 4H), 4.00 (d, J = 8.9 Hz, 1H), 3.87 (d, J = 8.9 Hz, 1H), 1.48 (s, 3H), 0.82 (s, 3H).29427636.11H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.28 (s, 1H), 8.16 (dd, J = 8.6, 1.4 Hz, 1H), 7.93 (dd, J = 10.8, 6.3 Hz, 1H), 7.85- 7.72 (m, 2H), 7.58 (dd, J = 7.4, 1.5 Hz, 1H), 7.39 (dd, J = 11.2, 6.0 Hz, 1H), 7.18 (s, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 5.13 (d, J = 6.6 Hz, 1H), 4.76-4.58 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (s, 4H), 3.84 (d, J = 8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H).29527623.71H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.57 (d, J = 9.6 Hz, 1H), 8.19 (dd, J = 8.6, 1.4 Hz, 1H), 7.95 (dd, J = 10.8, 6.3 Hz, 1H), 7.89-7.75 (m, 2H), 7.59 (dd, J = 7.5, 1.6 Hz, 1H), 7.41 (dd, J = 10.1, 4.8 Hz, 2H), 6.93 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.15 (d, J = 6.6 Hz, 1H), 4.81- 4.61 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz, 1H), 3.85 (d, J = 8.9 Hz, 1H), 1.42 (s, 3H), 0.77 (s, 3H).29627623.71H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.43 (d, J = 1.9 Hz, 1H), 8.18 (dd, J = 8.6, 1.4 Hz, 1H), 7.91 (dd, J = 10.9, 6.3 Hz, 1H), 7.88-7.74 (m, 3H), 7.59 (dd, J = 7.4, 1.5 Hz, 1H), 7.38 (dd, J = 11.2, 6.1 Hz, 1H), 6.92 (d, J = 8.2 Hz, 1H), 5.63 (d, J = 1.9 Hz, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.80-4.61 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz,1H), 4.00 (d, J = 8.9 Hz,1H), 3.85 (d, J = 8.9 Hz,1H), 1.41 (s, 3H), 0.76 (s,3H).29735603.091H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.24 (d, J = 6.9 Hz, 1H), 7.92 (t, J = 7.9 Hz, 1H), 7.81 (d, J = 8.4 Hz, 1H), 7.66 (d, J = 8.5 Hz, 1H), 7.57-7.49 (m, 3H), 7.45 (d, J = 8.4 Hz, 2H), 7.06 (d, J = 8.3 Hz, 1H), 6.97 (d, J = 7.4 Hz, 1H), 5.55 (d, J = 16.0 Hz, 1H), 5.46 (d, J = 8.7 Hz, 3H), 5.10 (d, J = 6.6 Hz, 1H), 4.61-4.39 (m, 2H), 3.83-3.69 (m, 2H), 1.38 (s, 3H), 0.66 (s, 3H).29835614.381H NMR (400 MHz, DMSO) δ 8.75 (d, J = 5.2 Hz, 1H), 8.35 (s, 1H), 8.06 (d, J = 9.4 Hz, 2H), 7.95 (dd, J = 10.0, 1.5 Hz, 1H), 7.90- 7.69 (m, 3H), 7.65-7.46 (m, 2H), 5.64 (s, 2H), 5.01 (d, J = 6.5 Hz, 1H), 4.73- 4.37 (m, 4H), 3.74 (q, J = 8.7 Hz, 2H), 1.31 (s, 3H), 0.59 (s, 3H).29935622.171H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 8.10- 7.72 (m, 3H), 7.72-7.40 (m, 8H), 5.57 (s, 2H), 5.02 (d, J = 6.5 Hz, 1H), 4.82- 4.27 (m, 4H), 3.85-3.64 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).30035621.091H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.25 (d, J = 7.1 Hz, 1H), 8.04- 7.76 (m, 2H), 7.74-7.40 (m, 6H), 6.95 (d, J = 7.5 Hz, 1H), 5.50 (d, J = 28.8 Hz, 4H), 5.10 (d, J = 6.7 Hz, 1H), 4.70-4.34 (m, 2H), 3.73 (d, J = 8.6 Hz, 2H), 1.38 (s, 3H), 0.66 (s, 3H).30135608.291H NMR (400 MHz, DMSO) δ 8.75 (d, J = 5.1 Hz, 1H), 8.50 (s, 1H), 8.00- 7.74 (m, 3H), 7.74-7.53 (m, 4H), 7.53-7.28 (m, 2H), 5.57 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.66-4.29 (m, 4H), 3.93-3.59 (m, 2H), 2.33 (s, 3H), 1.34 (s, 3H), 0.66 (s, 3H).30235622.211H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.88 (dd, J = 8.2, 5.1 Hz, 3H), 7.82-7.71 (m, 2H), 7.71- 7.44 (m, 4H), 7.34 (dd, J = 8.2, 2.0 Hz, 1H), 6.95 (d, J = 8.2 Hz, 1H), 5.55 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.58 (d, J = 18.1 Hz, 2H), 4.47 (dd, J = 11.2, 6.7 Hz, 1H), 4.32 (d, J = 17.3 Hz, 1H), 3.86-3.71 (m, 2H), 1.38 (s, 3H), 0.66 (s, 3H).30335640.131H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.93- 7.75 (m, 5H), 7.65 (t, J = 8.2 Hz, 1H), 7.61-7.50 (m, 2H), 7.36 (dd, J = 8.2, 2.1 Hz, 1H), 5.64 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.64- 4.42 (m, 3H), 4.30 (d, J = 17.3 Hz, 1H), 3.76 (t, J = 6.4 Hz, 2H), 1.38 (s, 3H), 0.65 (s, 3H).30535642.721H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.89 (dd, J = 9.8, 8.3 Hz, 3H), 7.76 (d, J = 7.5 Hz, 1H), 7.63 (t, J = 8.2 Hz, 1H), 7.55- 7.43 (m, 2H), 7.33 (dd, J = 8.2, 2.1 Hz, 1H), 6.94 (d, J = 8.1 Hz, 1H), 5.55 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.71-4.49 (m, 2H), 4.46 (dd, J = 11.3, 6.6 Hz, 1H), 4.32 (d, J = 17.3 Hz, 1H), 3.76 (s, 2H), 1.39 (s, 3H), 0.66 (s, 3H).30635658.851H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.93- 7.74 (m, 4H), 7.65 (t, J = 8.2 Hz, 1H), 7.51 (ddd, J = 11.1, 8.9, 1.6 Hz, 2H), 7.36 (dd, J = 8.2, 2.1 Hz, 1H), 5.64 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.71-4.39 (m, 3H), 4.31 (d, J = 17.4 Hz, 1H), 3.76 (s, 2H), 1.38 (s, 3H), 0.66 (s, 3H).30735649.361H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 8.02- 7.69 (m, 7H), 7.53-7.47 (m, 1H), 5.74 (s, 2H), 5.11 (d, J = 6.4 Hz, 1H), 4.74- 4.50 (m, 2H), 4.46 (dd, J = 11.1, 6.6 Hz, 1H), 4.31 (d, J = 17.3 Hz, 1H), 3.76 (s, 2H), 1.38 (s, 3H), 0.66 (s, 3H).30835613.211H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.98- 7.79 (m, 6H), 7.72 (d, J = 3.2 Hz, 1H), 7.60-7.42 (m, 2H), 7.19 (dd, J = 8.2, 2.6 Hz, 3H), 5.69 (s, 2H), 5.01 (s, 1H), 4.67-4.27 (m, 4H), 1.30 (s, 3H), 0.58 (s, 3H).30935608.251H NMR (400 MHz, DMSO) δ 8.18 (s, 1H), 8.00 (dd, J = 15.7, 9.9 Hz, 2H), 7.89 (t, J = 7.8 Hz, 1H), 7.74 (d, J = 7.7 Hz, 1H), 7.48 (t, J = 8.7 Hz, 2H), 6.92 (d, J = 8.2 Hz, 1H), 5.75 (s, 2H), 4.93 (s, 1H), 4.63-4.22 (m, 2H), 4.09 (s, 3H), 3.84- 3.65 (m, 2H), 1.26 (s, 3H), 0.56 (s, 3H).31035658.141H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.90 (dd, J = 10.2, 8.2 Hz, 1H), 7.70 (td, J = 6.3, 3.3 Hz, 1H), 7.64-7.45 (m, 6H), 5.58 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.67 (d, J = 17.4 Hz, 1H), 4.61-4.30 (m, 3H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H).311 A39629.321H NMR (400 MHz, DMSO) δ 8.44 (s, 1H), 7.98- 7.84 (m, 2H), 7.77 (ddd, J = 15.0, 8.4, 1.6 Hz, 5H), 7.68-7.48 (m, 3H), 7.37 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.29 (s, 1H), 4.61-4.33 (m, 3H), 4.28- 4.14 (m, 2H), 2.96 (s, 3H), 1.43 (s, 3H).311 B39629.321H NMR (400 MHz, DMSO) δ 8.44 (s, 1H), 7.98- 7.84 (m, 2H), 7.77 (ddd, J = 15.0, 8.4, 1.6 Hz, 5H), 7.68-7.48 (m, 3H), 7.37 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.29 (s, 1H), 4.61-4.33 (m, 3H), 4.28- 4.14 (m, 2H), 2.96 (s, 3H), 1.43 (s, 3H).31239629.321H NMR (400 MHz, DMSO) δ 8.46 (s, 1H), 7.99- 7.85 (m, 2H), 7.85-7.69 (m, 4H), 7.64 (d, J = 8.5 Hz, 1H), 7.55 (d, J = 7.3 Hz, 1H), 7.46 (dd, J = 10.4, 7.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.23 (d, J = 6.8 Hz, 1H), 4.82-4.25 (m, 4H), 4.13 (d, J = 10.1 Hz, 1H), 3.80 (d, J = 10.1 Hz, 1H), 3.31 (s, 3H), 0.84 (s, 3H).31339629.321H NMR (400 MHz, DMSO) δ 8.46 (s, 1H), 7.99- 7.85 (m, 2H), 7.85-7.69 (m, 4H), 7.64 (d, J = 8.5 Hz, 1H), 7.55 (d, J = 7.3 Hz, 1H), 7.46 (dd, J = 10.4, 7.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.23 (d, J = 6.8 Hz, 1H), 4.82-4.25 (m, 4H), 4.13 (d, J = 10.1 Hz, 1H), 3.80 (d, J = 10.1 Hz, 1H), 3.31 (s, 3H), 0.84 (s, 3H).31635649.311H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.92 (dd, J = 19.4, 8.4 Hz, 3H), 7.72 (d, J = 8.0 Hz, 2H), 7.68-7.56 (m, 2H), 7.51 (d, J = 11.2 Hz, 1H), 5.68 (s, 2H), 5.11 (d, J = 6.4 Hz, 1H), 4.82-4.31 (m, 3H), 3.76 (s, 2H), 0.66 (s, 3H).31735649.311H NMR (400 MHz, DMSO) δ 8.74 (d, J = 5.1 Hz, 1H), 8.36(s, 1H), 7.69- 7.57 (m, 2H), 7.57-7.45 (m, 3H), 7.41 (s, 1H), 7.35 (dd, J = 8.3, 2.1 Hz, 1H), 5.02 (d, J = 6.7 Hz, 1H), 4.64-4.31 (m, 4H), 3.81- 3.67 (m, 2H), 1.33 (s, 3H), 0.66 (s, 3H).31835636.251H NMR (400 MHz, DMSO) δ 8.75 (d, J = 5.1 Hz, 1H), 8.36 (s, 1H), 7.94 (d, J = 10.0 Hz, 1H), 7.83- 7.70 (m, 2H), 7.61 (s, 1H), 7.57-7.46 (m, 2H), 7.41 (s, 1H), 5.59 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.73-4.33 (m, 4H), 3.85-3.65 (m, 2H), 1.33 (s, 3H), 0.65 (s, 3H).31935644.81H NMR (400 MHz, DMSO) δ 8.75 (d, J = 5.1 Hz, 1H), 8.36 (s, 1H), 7.94 (d, J = 10.0 Hz, 1H), 7.83- 7.70 (m, 2H), 7.61 (s, 1H), 7.57-7.46 (m, 2H), 7.41 (s, 1H), 5.59 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.73-4.33 (m, 4H), 3.85-3.65 (m, 2H), 1.33 (s, 3H), 0.65 (s, 3H).32035626.811H NMR (400 MHz, DMSO) δ 8.75 (d, J = 5.1 Hz, 1H), 8.51 (s, 1H), 7.94 (d, J = 10.0 Hz, 1H), 7.88- 7.69 (m, 3H), 7.68-7.57 (m, 2H), 7.49 (d, J = 5.1 Hz, 1H), 7.42 (s, 1H), 5.59 (s, 2H), 5.02 (d, J = 6.8 Hz, 1H), 4.73-4.23 (m, 4H), 3.88-3.68 (m, 2H), 1.33 (s, 3H), 0.66 (s, 3H).32135613.441H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.98- 7.70 (m, 9H), 7.57 (d, J = 8.5 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.65 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.57 (d, J = 16.7 Hz, 2H), 4.47 (dd, J = 11.1, 6.7 Hz, 1H), 4.31 (d, J = 17.1 Hz, 1H), 3.86- 3.69 (m, 2H), 1.38 (s, 3H), 0.65 (s, 3H).32235631.321H NMR (400 MHz, DMSO) δ 8.47 (s, 1H), 8.01- 7.69 (m, 8H), 7.56 (d, J = 8.5 Hz, 1H), 5.74 (s, 2H), 5.08 (d, J = 6.7 Hz, 1H), 4.76-4.44 (m, 3H), 4.29 (d, J = 17.1 Hz, 1H), 3.77 (d, J = 3.8 Hz, 2H), 1.38 (s, 3H), 0.65 (s, 3H).32335666.251H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.30 (s, 1H), 7.83 (dd, J = 10.2, 8.2 Hz, 2H), 7.64 (d, J = 8.4 Hz, 1H), 7.53 (s, 1H), 7.37 (s, 1H), 7.32-7.23 (m, 3H), 5.43 (s, 2H), 5.02 (d, J = 6.8 Hz, 1H), 4.72-4.34 (m, 4H), 3.93 (s, 4H), 2.32 (s, 3H), 1.33 (s, 3H), 0.66 (s, 3H).32435653.531H NMR (400 MHz, DMSO) δ 8.36 (d, J = 8.3 Hz, 2H), 7.96-7.87 (m, 2H), 7.81 (dd, J = 10.2, 8.2 Hz, 1H), 7.70 (dd, J = 8.2, 2.8 Hz, 1H), 7.57-7.46 (m, 2H), 7.27 (s, 1H), 5.54 (s, 2H), 5.01 (d, J = 6.6 Hz, 1H), 4.64-4.32 (m, 5H), 3.96 (s, 3H), 1.31 (s, 3H), 0.59 (s, 3H).32535631.171H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 8.10- 7.80 (m, 3H), 7.80-7.65 (m, 3H), 7.65-7.35 (m, 3H), 5.68 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.69-4.30 (m, 4H), 1.34 (s, 3H), 0.61 (s, 3H).32635646.341H NMR (400 MHz, DMSO) δ 8.34 (d, J = 20.1 Hz, 2H), 8.04-7.85 (m, 2H), 7.74 (d, J = 4.7 Hz, 2H), 7.55 (dd, J = 11.2, 1.2 Hz, 1H), 7.40 (d, J = 7.3 Hz, 1H), 7.07 (d, J = 8.3 Hz, 1H), 6.67 (s, 1H), 5.65- 5.38 (m, 4H), 5.13 (d, J = 6.5 Hz, 1H), 4.64-4.39 (m, 2H), 3.83-3.53 (m, 2H), 1.38 (s, 3H), 0.68 (s, 3H).32735655.531H NMR (400 MHz, DMSO) δ 8.34 (d, J = 16.2 Hz, 2H), 7.91 (dd, J = 8.3, 7.3 Hz, 1H), 7.68-7.44 (m, 3H), 7.44-7.27 (m, 2H), 7.02 (d, J = 8.3 Hz, 1H), 6.70 (s, 1H), 5.45 (s, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.63-4.29 (m, 2H), 3.93- 3.63 (m, 2H), 1.39 (s, 3H), 0.68 (s, 3H).32835630.221H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.24 (d, J = 7.0 Hz, 1H), 8.04- 7.84 (m, 2H), 7.87-7.70 (m, 3H), 7.66 (d, J = 8.5 Hz, 1H), 7.59 (dt, J = 8.6, 2.0 Hz, 1H), 6.92 (d, J = 7.6 Hz, 1H), 5.67 (s, 2H), 5.63- 5.32 (m, 2H), 5.10 (d, J = 6.6 Hz, 1H), 4.55 (d, J = 11.1 Hz, 1H), 4.44 (dd, J = 11.1, 6.8 Hz, 1H), 3.83- 3.59 (m, 2H), 1.37 (s, 3H), 0.65 (s, 3H).32935622.451H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 8.10- 7.90 (m, 2H), 7.85 (t, J = 7.8 Hz, 1H), 7.74-7.57 (m, 2H), 7.57-7.44 (m, 3H), 7.32 (dd, J = 8.2, 2.1 Hz, 1H), 6.89 (d, J = 8.2 Hz, 1H), 5.53 (s, 2H), 5.01 (d, J = 6.6 Hz, 1H), 4.66-4.23 (m, 4H), 1.30 (s, 3H), 0.58 (s, 3H).33035613.261H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 8.05- 7.68 (m, 9H), 7.62 (d, J = 8.5 Hz, 1H), 7.49 (t, J = 7.9 Hz, 1H), 5.72 (s, 2H), 4.99 (d, J = 6.7 Hz, 1H), 4.60- 4.29 (m, 3H), 3.88-3.67 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H).33135604.281H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.00- 7.90 (m, 2H), 7.90-7.78 (m, 2H), 7.74-7.57 (m, 3H), 7.57-7.43 (m, 2H), 7.33 (dd, J = 8.2, 2.1 Hz, 1H), 6.90 (d, J = 8.2 Hz, 1H), 5.53 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.62-4.27 (m, 5H), 3.88-3.67 (m, 2H), 1.30 (s, 3H), 0.59 (s, 3H).33235622.161H NMR (400 MHz, DMSO) δ 8.50 (s, 1H), 7.98- 7.86 (m, 2H), 7.86-7.80 (m, 2H), 7.76-7.58 (m, 3H), 7.57-7.44 (m, 2H), 7.35 (dd, J = 8.2, 2.1 Hz, 1H), 5.62 (s, 2H), 5.00 (d, J = 6.6 Hz, 1H), 4.61-4.33 (m, 5H), 3.83-3.67 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H).33435670.331H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.91 (dd, J = 10.2, 8.2 Hz, 1H), 7.78 (dd, J = 8.4, 1.5 Hz, 1H), 7.75-7.69 (m, 1H), 7.69-7.48 (m, 4H), 7.36 (dd, J = 8.4, 2.1 Hz, 1H), 5.61 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.76-4.27 (m, 4H), 3.92-3.69 (m, 2H), 1.39 (s, 3H), 0.65 (s, 3H).33535658.121H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.91 (dd, J = 10.2, 8.2 Hz, 1H), 7.78 (dd, J = 8.4, 1.5 Hz, 1H), 7.75-7.69 (m, 1H), 7.69-7.48 (m, 4H), 7.36 (dd, J = 8.4, 2.1 Hz, 1H), 5.61 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.76-4.27 (m, 4H), 3.92-3.69 (m, 2H), 1.39 (s, 3H), 0.65 (s, 3H).33635676.381H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.91 (dd, J = 10.2, 8.2 Hz, 1H), 7.74 (ddd, J = 10.5, 5.7,2.0 Hz, 1H), 7.69-7.56 (m, 2H), 7.52 (ddd, J = 11.1, 3.4, 1.6 Hz, 2H), 7.35 (dd, J = 8.2, 2.1 Hz, 1H), 5.61 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.67 (d, J = 17.4 Hz, 1H), 4.62-4.32 (m, 3H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H).33735630.351H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 8.09- 7.64 (m, 8H), 7.57-7.39 (m, 2H), 5.72 (s, 2H), 5.00 (d, J = 6.6 Hz, 1H), 4.59- 4.28 (m, 2H), 3.75-3.80 (m, 2H), 1.30 (s, 3H), 0.58 (s, 3H).33935689.581H NMR (400 MHz, DMSO) δ 8.36 (d, J = 2.6 Hz, 2H), 7.90 (dd, J = 10.2, 8.2 Hz, 1H), 7.75 (ddd, J = 10.4, 5.8, 2.0 Hz, 1H), 7.61 (ddd, J = 8.1, 2.9, 1.5 Hz, 1H), 7.51 (dd, J = 11.2, 1.2 Hz, 1H), 7.27 (s, 1H),5.53 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.67 (d, J = 17.5 Hz, 1H), 4.61-4.31 (m, 3H), 3.95 (s, 3H), 3.76 (s, 2H), 1.39 (s, 3H), 0.67 (s, 3H).34035671.61H NMR (400 MHz, DMSO) δ 8.35 (s, 2H), 7.84 (ddd, J = 10.2, 7.4, 4.4 Hz, 2H), 7.61-7.43 (m, 3H), 7.27 (s, 1H), 5.52 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.63-4.49 (m, 2H), 4.49- 4.28 (m, 2H), 3.95 (s, 3H), 1.34 (s, 3H), 0.62 (s, 3H).34135671.511H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.36 (s, 1H), 7.90 (dd, J = 10.2, 8.2 Hz, 1H), 7.78 (dd, J = 8.4, 1.5 Hz, 1H), 7.60 (dd, J = 8.7, 3.3 Hz, 2H), 7.28 (s, 1H), 5.53 (s, 2H), 5.09 (d, J = 6.5 Hz, 1H), 4.75-4.52 (m, 2H), 4.47 (dd, J = 11.1, 6.7 Hz, 1H), 4.36 (d, J = 17.4 Hz, 1H), 3.96 (s, 3H), 3.86-3.71 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H).34235653.761H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.35 (s, 1H), 7.89-7.76 (m, 3H), 7.64 (d, J = 8.5 Hz, 1H), 7.59-7.41 (m, 2H), 7.27 (s, 1H), 5.52 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.63-4.32 (m, 4H), 3.95 (s, 3H), 3.88- 3.66 (m, 2H), 1.34 (s, 3H), 0.62 (s, 3H).34335657.361H NMR (400 MHz, DMSO) δ 8.79 (d, J = 5.2 Hz, 1H), 8.36 (s, 1H), 8.17 (d, J = 7.0 Hz, 1H), 7.70- 7.45 (m, 5H), 7.35 (dd, J = 8.2, 2.1 Hz, 1H), 5.54 (s, 2H), 5.04 (d, J = 6.7 Hz, 1H), 4.67 (d, J = 17.1 Hz, 1H), 4.59-4.37 (m, 3H), 3.84-3.66 (m, 2H), 1.36 (s, 3H), 0.67 (s, 3H).34435673.1751H NMR (400 MHz, DMSO) δ 8.80 (d, J = 5.2 Hz, 1H), 8.36 (s, 1H), 8.16 (d, J = 7.0 Hz, 1H), 7.75 (t, J = 7.6 Hz, 1H), 7.65 (dd, J = 5.2, 1.8 Hz, 1H), 7.59 (d, J = 11.7 Hz, 1H), 7.50 (dd, J = 16.7, 9.4 Hz, 3H), 7.07 (t, J = 55.6 Hz, 1H), 5.61 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.66 (d, J = 17.1 Hz, 1H), 4.58-4.37 (m, 3H), 3.85- 3.70 (m, 2H), 1.36 (s, 3H), 0.67 (s, 3H).34535647.461H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 8.00- 7.83 (m, 3H), 7.83-7.71 (m, 3H), 7.65-7.43 (m, 3H), 5.69 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.69-4.49 (m, 2H), 4.49-4.35 (m, 2H), 3.80 (d, J = 8.6 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H).34635665.2841H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 8.00- 7.83 (m, 3H), 7.83-7.70 (m, 2H), 7.60-7.43 (m, 3H), 5.70 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.62 (d, J = 17.0 Hz, 1H), 4.53 (d, J = 11.9 Hz, 1H), 4.47-4.36 (m, 2H), 3.83-3.69 (m, 2H), 1.35 (s, 3H), 0.66 (s, 3H).34735658.81H NMR (400 MHz, DMSO) δ 8.36 (s, 1H), 7.84 (ddd, J = 11.9, 10.3, 7.4 Hz, 2H), 7.63 (t, J = 8.1 Hz, 1H), 7.59-7.43 (m, 4H), 7.35 (dd, J = 8.2, 2.0 Hz, 1H), 5.61 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.61-4.47 (m, 2H), 4.48-4.32 (m, 2H), 3.75 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H).34835641.21H NMR (400 MHz, MeOD) δ 8.60 (d, J = 5.8 Hz, 1H), 8.55 (s, 1H), 7.90 (dd, J = 10.3, 6.1 Hz, 1H), 7.66 (d, J = 11.0 Hz, 1H), 7.56 (t, J = 8.0 Hz, 1H), 7.35- 7.13 (m, 3H), 6.91 (d, J = 5.8 Hz, 1H), 5.59 (s, 2H), 4.91 (s, 8H), 4.65-4.55 (m, 1H), 4.55-4.42 (m, 3H), 3.93 (d, J = 8.8 Hz, 1H), 3.79 (d, J = 8.8 Hz, 1H), 3.33 (p, J = 1.7 Hz, 5H), 1.36 (s, 3H), 0.67 (s, 3H).34940(M+) 659.21H NMR (400 MHz, Methanol-d4) δ 8.16 (d, J = 1.3 Hz, 1H), 7.82-7.77 (m, 1H), 7.75 (d, J = 7.9 Hz, 1H), 7.65 (dd, J = 11.2, 1.2 Hz, 1H), 7.50 (dd, J = 7.4, 1.6 Hz, 1H), 7.44 (t, J = 9.7 Hz, 2H), 7.31 (dd, J = 11.5, 7.9 Hz, 1H), 7.17 (dd, J = 11.5, 6.0 Hz, 1H), 6.86 (d, J = 8.2 Hz, 1H), 5.49 (s, 2H), 5.16 (qd, J = 7.1, 2.5 Hz, 1H), 4.77-4.50 (m, 10H),4.50-4.35 (m, 1H), 3.78 (d,J = 1.8 Hz, 3H), 2.88-2.69(m, 1H), 2.53-2.40 (m,1H).35018632.41H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J = 1.2 Hz, 1H), 8.75 (d, J = 2.1 Hz, 1H), 8.25 (dd, J = 8.4, 2.2 Hz, 1H), 8.17 (d, J = 8.4 Hz, 1H), 8.10 (d, J = 1.3 Hz, 1H), 8.05-7.97 (m, 2H), 7.91 (t, J = 7.9 Hz, 1H), 7.58- 7.48 (m, 2H), 7.45 (d, J = 11.7 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 4.63 (t, J = 5.1 Hz, 2H), 4.53 (s, 2H), 3.70 (t, J = 5.0 Hz, 2H), 3.22 (s, 3H).35118607.61H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J = 1.2 Hz, 1H), 7.98-7.85 (m, 3H), 7.74 (d, J = 5.4 Hz, 2H), 7.55-7.47 (m, 2H), 7.43 (d, J = 11.8 Hz, 1H), 7.01 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.62 (t, J = 5.1 Hz, 2H), 4.52 (s, 2H), 3.70 (t, J = 5.0 Hz, 2H), 3.22 (s, 3H).35218630.21H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J = 1.2 Hz, 1H), 8.75 (d, J = 2.1 Hz, 1H), 8.25 (dd, J = 8.4, 2.2 Hz, 1H), 8.17 (d, J = 8.5 Hz, 1H), 8.12 (d, J = 1.3 Hz, 1H), 8.00 (d, J = 1.2 Hz, 1H), 7.96-7.82 (m, 2H), 7.60-7.46 (m, 2H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.62 (s, 2H), 4.54 (dd, J = 15.2, 3.1 Hz, 1H),4.47 (s, 2H), 4.37 (dd, J = 15.2, 8.8 Hz, 1H), 3.79-3.60 (m, 1H), 3.08 (s, 3H), 1.23 (d, J = 6.1 Hz, 3H).35318632.51H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.2 Hz, 1H), 8.12 (d, J = 1.3 Hz, 1H), 8.01 (d, J = 1.2 Hz, 1H), 7.97-7.78 (m, 5H), 7.58-7.45 (m, 2H), 7.40 (dd, J = 11.5, 6.0 Hz, 1H), 6.98 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.54 (dd, J = 15.2, 3.1 Hz, 1H), 4.47 (s, 2H), 4.37 (dd, J = 15.2, 8.8 Hz, 1H), 3.78-3.59 (m, 1H), 3.08 (s, 3H), 1.23 (d, J = 6.1 Hz, 3H).35418647.21H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.2 Hz, 1H), 8.12 (d, J = 1.3 Hz, 1H), 8.01 (d, J = 1.2 Hz, 1H), 7.97-7.78 (m, 5H), 7.58-7.45 (m, 2H), 7.40 (dd, J = 11.5, 6.0 Hz, 1H), 6.98 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.54 (dd, J = 15.2, 3.1 Hz, 1H), 4.47 (s, 2H), 4.37 (dd, J = 15.2, 8.8 Hz, 1H), 3.78-3.59 (m, 1H), 3.08 (s, 3H), 1.23 (d, J = 6.1 Hz, 3H).35518605.21H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J = 1.2 Hz, 1H), 7.97-7.85 (m, 2H), 7.82-7.70 (m, 3H), 7.58-7.46 (m, 2H), 7.39 (dd, J = 11.5, 6.0 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 4.53 (dd, J = 15.2, 3.1 Hz, 1H), 4.46 (s, 2H), 4.36 (dd, J = 15.2, 8.8 Hz, 1H), 3.68 (ddd, J = 9.1, 6.1, 3.1 Hz, 1H), 3.08 (s, 3H), 1.23 (d, J = 6.1 Hz, 3H).35727620.31H NMR (400 MHz, Methanol-d4) δ 8.87 (s, 1H), 8.15 (dd, J = 8.7, 1.3 Hz, 1H), 7.99-7.71 (m, 3H), 7.71-7.45 (m, 5H), 7.37 (dd, J = 11.2, 6.0 Hz, 1H), 7.03-6.86 (m, 1H), &nbsp; 5.56 (s, 2H), 5.12 (d, J = 6.6 Hz, 1H), 4.78-4.61 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 3.99 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), &nbsp; 1.40 (s, 3H), 0.75 (s, 3H).35827638.01H NMR (400 MHz, Methanol-d4) δ 8.96 (s, 1H), 8.24 (dd, J = 8.6, 1.3 Hz, 1H), 7.83 (d, J = 8.6 Hz, 1H), 7.78-7.67 (m, 2H), 7.58 (d, J = 8.6 Hz, 2H), 7.46 (d, J = 7.5 Hz, 1H), 7.19 (d, J = 10.5 Hz, 1H), 7.02 (dd, J = 9.0, 2.9 Hz, 1H), 5.51 (s, 2H), 5.19 (d, J = 6.5 Hz, 1H), 4.83-4.62 (m, 3H),4.52 (dd, J = 11.8, 6.6 Hz, 1H), 4.01 (d, J = 9.0Hz, 1H), 3.84 (d, J = 9.0 Hz,1H), 2.21 (s, 3H), 1.36 (s,3H), 0.73 (s, 3H).35935627.01H NMR (400 MHz, Methanol-d4) δ 8.96 (s, 1H), 8.24 (dd, J = 8.6, 1.3 Hz, 1H),7.83 (d, J = 8.6 Hz, 1H), 7.78-7.67 (m, 2H), 7.58 (d, J = 8.6 Hz, 2H), 7.46 (d, J = 7.5 Hz, 1H), 7.19 (d, J = 10.5 Hz, 1H), 7.02 (dd, J = 9.0, 2.9 Hz, 1H), 5.51 (s, 2H), 5.19 (d, J = 6.5 Hz, 1H), 4.83-4.62 (m, 3H), 4.52 (dd, J = 11.8, 6.6 Hz, 1H), 4.01 (d, J = 9.0Hz, 1H), 3.84 (d, J = 9.0 Hz,1H), 2.21 (s, 3H), 1.36 (s,3H), 0.73 (s, 3H).36027588.51H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.18 (dd, J = 8.6, 1.3 Hz, 1H), 7.93 (dd, J = 10.8, 6.2 Hz, 1H), 7.87-7.73 (m, 2H), 7.66-7.48 (m, 3H), 7.40 (dd, J = 11.2, 6.0 Hz, 1H), 7.21-7.02 (m, 2H), 6.90 (d, J = 8.2 Hz, 1H), 5.46 (s, 2H), 5.15 (d, J = 6.5 Hz, 1H), 4.79-4.63 (m, 2H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz,1H), 3.84 (d, J = 8.9 Hz,1H), 3.33 (p, J = 1.7 Hz,3H), 1.41 (s, 3H), 0.76 (s,3H).36127584.61H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J = 8.6, 1.3 Hz, 1H), 7.93 (dd, J = 10.9, 6.3 Hz, 1H), 7.88-7.72 (m, 2H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.38 (dd, J = 14.9, 7.0 Hz, 3H), 7.19 (d, J = 7.8 Hz, 2H), 6.88 (d, J = 8.2 Hz, 1H), 5.44 (s, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.77-4.61 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.84 (d, J = 8.9 Hz,1H), 2.34 (s, 3H), 1.42 (s,3H), 0.76 (s, 3H).36235595.31H NMR (400 MHz, Methanol-d4) δ 8.99-8.75 (m, 1H), 8.15 (dd, J = 8.6, 1.4 Hz, 1H), 7.95-7.66 (m, 8H), 7.61 (d, J = 7.5 Hz, 1H), 6.97 (d, J = 8.2 Hz, 1H), 5.62 (s, 2H), 5.17 (d, J = 6.5 Hz, 1H), 4.81-4.46 (m, 4H), 4.00 (d, J = 8.9 Hz, 1H), 3.86 (d, J = 8.9 Hz, 1H), 1.45 (s, 3H), 0.79 (s, 3H).36335570.61H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J = 8.6, 1.4 Hz, 1H), 7.92 (dd, J = 10.9, 6.3 Hz, 1H), 7.87-7.70 (m, 2H), 7.56 (dd, J = 7.4, 1.7 Hz, 1H), 7.52-7.44 (m, 2H), 7.42-7.33 (m, 4H), 6.91 (d, J = 8.2 Hz, 1H), 5.49 (s, 2H), 5.15 (d, J = 6.5 Hz, 1H), 4.77-4.62 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz,1H), 3.85 (d, J = 8.9 Hz,1H), 1.42 (s, 3H), 0.77 (s,3H).36435613.21H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J = 8.6, 1.3 Hz, 1H), 7.91 (d, J = 8.1 Hz, 1H), 7.87-7.66 (m, 5H), 7.63-7.54 (m, 3H), 6.97 (dd, J = 8.9, 2.7 Hz, 1H), 5.61 (s, 2H), 5.12 (d, J = 6.6 Hz, 1H), 4.76-4.61 (m, 3H), 4.60-4.49 (m, 1H), 4.00 (d, J = 8.9 Hz, 1H), 3.83 (d, J = 8.9 Hz, 1H), 1.37 (s, 3H), 0.73 (s, 3H).36535649.21H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 7.82-7.65 (m, 3H), 7.65- 7.55 (m, 2H), 7.35 (dd, J = 9.9, 5.7 Hz, 1H), 7.27 (dd, J = 9.9, 6.0 Hz, 1H), 7.01 (dd, J = 9.0, 2.9 Hz, 1H), 5.54 (s, 2H), 4.99 (d, J = 6.7 Hz, 1H), 4.69-4.42 (m, 4H), 3.95 (d, J = 8.8 Hz, 1H), 3.80 (d, J = 8.8 Hz, 1H), 1.36 (s, 3H), 0.68 (s, 3H).36635591.51H NMR (400 MHz, Chloroform-d) δ 8.00 (t, J = 7.5 Hz, 1H), 7.80-7.68 (m, 3H), 7.68-7.57 (m, 3H), 7.56-7.36 (m, 2H), 7.15 (dd, J = 11.1, 6.0 Hz, 1H), 5.60 (s, 2H), 4.67-4.55 (m, 4H), 3.82 (t, J = 4.8 Hz, 2H), 3.33 (s, 3H).36735622.81H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 7.90-7.82 (m, 1H), 7.77 (t, J = 7.9 Hz, 1H), 7.70 (dd, J = 11.0, 1.2 Hz, 1H), 7.58- 7.44 (m, 3H), 7.42-7.34 (m, 2H), 7.24 (dd, J = 11.4, 6.0 Hz, 1H), 6.88 (d, J = 8.2 Hz, 1H), 5.46 (s, 2H), 4.97 (d, J = 6.7 Hz, 1H), 4.65- 4.34 (m, 4H), 3.93 (d, J = 8.8 Hz, 1H), 3.79 (d, J = 8.8 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H).36835623.31H NMR (400 MHz, Methanol-d4) δ 7.99 (d, J = 1.2 Hz, 1H), 7.86-7.68 (m, 2H), 7.68-7.47 (m, 5H), 7.12 (dd, J = 11.6, 6.1 Hz, 1H), 5.71 (s, 2H),4.52 (d, J = 16.6 Hz, 1H), 4.45-4.34 (m, 1H), 4.27 (dd, J = 15.1, 8.9 Hz, 1H), 3.75 (ddd, J = 9.2, 6.1, 3.1 Hz, 1H), 3.13 (s, 3H), 2.08-1.89 (m, 1H), 1.29 (d, J = 6.2 Hz, 3H).36935613.61H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.20 (dd, J = 8.6, 1.4 Hz, 1H), 8.02 (t, J = 8.3 Hz, 1H), 7.92-7.71 (m, 2H), 7.69- 7.46 (m, 4H), 7.40-7.21 (m, 2H), 5.70 (s, 2H), 5.10 (d, J = 6.5 Hz, 1H), 4.83- 4.58 (m, 3H), 4.47 (dd, J = 11.6, 6.7 Hz, 1H), 3.99 (d, J = 8.9 Hz, 1H), 3.82 (d, J = 8.9 Hz, 1H), 1.33 (s, 3H), 0.69 (s, 3H).37027595.71H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 1H), 8.15 (dd, J = 8.6, 1.4 Hz, 1H), 7.96-7.82 (m, 2H), 7.78 (d, J = 2.9 Hz, 1H), 7.77-7.70 (m, 3H), 7.70- 7.56 (m, 3H), 7.56-7.42 (m, 1H), 5.69 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.74- 4.58 (m, 3H), 4.49 (dd, J = 11.5, 6.7 Hz, 1H), 4.18 (ddd, J = 12.3, 9.0, 3.7 Hz, 1H), 3.99 (d, J = 8.9 Hz, 1H),3.82 (d, J = 8.9 Hz, 1H),3.76-3.72 (m, 1H), 3.56(dt, J = 11.8, 2.8 Hz, 1H),1.36 (s, 3H), 0.71 (s, 3H).37124587.01H NMR (400 MHz, Methanol-d4) δ 8.96 (s, 1H), 8.25 (dd, J = 8.6, 1.4 Hz, 1H), 8.06-7.92 (m, 2H), 7.88-7.73 (m, 2H), 7.68- 7.55 (m, 2H), 7.55-7.44 (m, 2H), 7.44-7.28 (m, 2H), 6.88 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 5.19 (d, J = 6.4 Hz, 1H), 4.78 (d, J = 7.2 Hz, 2H), 4.65 (dd, J = 11.7, 1.3 Hz, 1H), 4.51 (dd, J = 11.8, 6.6 Hz, 1H), 4.01(d, J = 9.0 Hz, 1H), 3.84 (d,J = 9.0 Hz, 1H), 1.37 (s,3H), 0.75 (s, 3H).37235595.71H NMR (400 MHz, Methanol-d4) δ 8.57 (s, 1H), 7.91-7.62 (m, 8H), 7.52 (d, J = 7.5 Hz, 1H), 7.38 (t, J = 7.8 Hz, 1H), 6.89 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.94 (s, 1H), 4.60-4.48 (m, 3H), 4.43 (dd, J = 11.4, 6.8 Hz, 1H), 3.93 (d, J = 8.9 Hz, 1H), 3.78 (d, J = 8.8 Hz, 1H), 1.29 (s, 3H), 0.62 (s, 3H).37327577.71H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.21 (dd, J = 8.6, 1.4 Hz, 1H), 8.00-7.87 (m, 2H), 7.87-7.79 (m, 2H), 7.74 (d, J = 8.3 Hz, 2H), 7.67 (d, J = 8.1 Hz, 2H), 7.61-7.46 (m, 2H), 6.93 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.15 (d, J = 6.5 Hz, 1H), 4.73 (d, J = 5.6 Hz, 2H), 4.64 (dd, J = 11.7, 1.4 Hz, 1H), 4.50 (dd, J = 11.7, 6.7 Hz, 1H), 4.00(d, J = 8.9 Hz, 1H), 3.83 (d,J = 8.9 Hz, 1H), 1.36 (s,3H), 0.73 (s, 3H).37427633.31H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.2 Hz, 1H), 8.01 (d, J = 1.2 Hz, 1H), 7.98-7.76 (m, 5H), 7.65 (dd, J = 8.5, 6.8 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.48-7.32 (m, 2H), 6.98 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 4.61 (t, J = 5.2 Hz, 2H), 4.43 (s, 2H), 3.73 (t, J = 5.2 Hz, 2H), 3.23 (s, 3H).37535573.31H NMR (400 MHz, Methanol-d4) δ 7.94 (dd, J = 8.6, 6.6 Hz, 1H), 7.82 (t, J = 7.8 Hz, 1H), 7.79-7.72 (m, 3H), 7.67 (d, J = 8.1 Hz, 2H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.50 (d, J = 8.6 Hz, 1H), 7.25 (dd, J = 11.4, 6.0 Hz, 1H), 6.95 (d, J = 8.2 Hz, 1H), 5.59 (s, 2H), 4.74 (t, J = 5.0 Hz, 2H), 4.59 (s, 2H), 3.84 (t, J = 4.9 Hz, 2H), 3.32 (s, 3H).37635613.631H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 7.86-7.72 (m, 4H), 7.71- 7.63 (m, 3H), 7.54 (dd, J = 7.5, 1.6 Hz, 1H), 7.23 (dd, J = 11.4, 6.0 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 4.97 (d, J = 6.7 Hz, 1H), 4.65-4.41 (m, 4H), 3.94 (d, J = 8.8 Hz, 1H), 3.80 (d, J = 8.8 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H).37727600.41H NMR (400 MHz, Methanol-d4) 5 8.15 (d, J = 1.2 Hz, 1H), 7.83 (dd, J = 8.3, 7.5 Hz, 1H), 7.66 (ddd, J = 11.3, 5.4, 1.8 Hz, 2H), 7.56 (dd, J = 7.4, 1.7 Hz, 1H), 7.53-7.45 (m, 2H), 7.45 - 7.33 (m, 2H), 6.92 (d, J = 8.3 Hz, 1H), 5.48 (s, 2H), 4.67 (t, J = 5.0 Hz, 2H), 4.60 (s, 2H), 3.86- 3.78 (m, 2H), 3.33 (s, 3H).37836591.61H NMR (400 MHz, Methanol-d4) δ 8.15 (d, J = 1.3 Hz, 1H), 7.85 (t, J = 7.9 Hz, 1H), 7.75 (d, J = 8.3 Hz, 2H), 7.72-7.64 (m, 3H), 7.63-7.56 (m, 2H), 6.97 (d, J = 8.3 Hz, 1H), 5.59 (s, 2H), , 4.68 (d, J = 5.0 Hz, 2H), 4.60 (s, 2H), 3.75 (dd, J = 2.9, 1.7 Hz, 1H), 3.62- 3.51 (m, 2H), 3.32 (s, , 3H).379chiral separation Peak 2595.61H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.29-8.11 (m, 1H), 8.04- 7.89 (m, 2H), 7.89-7.69 (m, 3H), 7.67-7.42 (m, 4H), 6.92 (d, J = 8.1 Hz, 1H), 5.66 (s, 2H), 5.17- 5.06 (m, 1H), 4.79-4.60 (m, 3H), 4.50 (dd, J = 11.5, 6.7 Hz, 1H), 4.32-4.14 (m, 1H), 3.99 (d, J = 8.9 Hz, 1H), 3.56 (dq, J = 9.8, 2.9 Hz, 1H), 1.36 (s, 3H), 0.72 (s, 3H).380chiral separation Peak 1595.61H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.28-8.12 (m, 1H), 8.06- 7.89 (m, 2H), 7.89-7.70 (m, 3H), 7.68-7.43 (m, 4H), 6.93 (d, J = 8.2 Hz, 1H), 5.66 (s, 2H), 5.20- 5.11 (m, 1H), 4.81-4.61 (m, 3H), 4.61-4.49 (m, 1H), 4.28-4.11 (m, 1H), 4.00 (d, J = 8.9 Hz, 1H), 3.59-3.46 (m, 1H), 1.37 (s, 3H), 0.73 (s, 3H).38135587.731H NMR (400 MHz, Methanol-d4) δ 8.17 (d, J = 1.2 Hz, 1H), 7.83-7.59 (m, 8H), 7.50 (dd, J = 7.5, 1.5 Hz, 1H), 7.19 (dd, J = 11.5, 6.0 Hz, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.55 (s, 2H), 4.61- 4.45 (m, 3H), 4.37 (dd, J = 15.2, 9.1 Hz, 1H), 3.72 (dqt, J = 10.1,6.9, 3.4 Hz, 1H), 3.15 (s, 3H), 1.30 (d, J = 6.1 Hz, 3H).38227573.61H NMR (400 MHz, Methanol-d4) δ 8.19 (s, 1H), 7.80 (d, J = 7.8 Hz, 1H), 7.77-7.69 (m, 4H), 7.66 (d, J = 8.3 Hz, 2H), 7.51 (dd, J = 7.5, 1.6 Hz, 1H), 7.19 (dd, J = 11.4, 6.0 Hz, 1H), 6.92 (d, J = 8.2 Hz, 1H), 5.57 (s, 2H), 4.63 (t, J = 5.0 Hz, 2H), 4.57 (s, 2H), 3.75 (t, J = 4.9 Hz, 2H), 3.27 (s, 3H).383 Achiral separation Peak 2638.361H NMR (400 MHz, Methanol-d4) δ 8.79 (s, 1H), 8.07 (dd, J = 8.5, 1.4 Hz, 1H), 7.97-7.77 (m, 2H), 7.69 (dd, J = 24.1, 8.1 Hz, 2H), 7.55 (dd, J = 7.5, 1.5 Hz, 1H), 7.43-7.20 (m, 3H), 6.97-6.86 (m, 1H), 5.59 (s, 2H), 5.04 (d, J = 6.7 Hz, 1H), 4.67-4.44 (m, 4H), 3.97 (d, J = 8.8 Hz, 1H), 3.82 (d, J = 8.8 Hz, 1H), 3.75-3.61 (m, 2H), 1.39 (s, 3H), 0.71 (s, 3H).383 Bchiral separation Peak 1638.31H NMR (400 MHz, Methanol-d4) δ 8.79 (s, 1H), 8.08 (dd, J = 8.5, 1.5 Hz, 1H), 7.94-7.76 (m, 2H), 7.69 (dd, J = 24.9, 8.1 Hz, 2H), 7.56 (dd, J = 7.5, 1.6 Hz, 1H), 7.45-7.20 (m, 3H), 6.92 (d, J = 8.4 Hz, 1H), 5.60 (s, 2H), 5.05 (d, J = 6.7 Hz, 1H), 4.76-4.43 (m, 4H), 3.97 (d, J = 8.8 Hz, 1H), 3.82 (d, J = 8.8 Hz, 1H), 3.74-3.66 (m, 1H), 1.39 (s, 3H), 0.71 (s, 3H).38436595.61H NMR (400 MHz, Methanol-d4) δ 8.85 (s, 1H), 8.14 (dd, J = 8.6, 1.4 Hz, 1H), 8.03-7.87 (m, 2H), 7.87-7.67 (m, 4H), 7.67- 7.44 (m, 3H), 6.92 (d, J = 8.2 Hz, 1H), 5.66 (s, 2H), 5.08 (d, J = 6.6 Hz, 1H), 4.72-4.56 (m, 3H), 4.49 (dd, J = 11.5, 6.8 Hz, 1H), 3.99 (d, J = 8.9 Hz, 1H), 3.82 (d, J = 8.8 Hz, 1H), 3.72-3.63 (m, 1H), 1.36 (s, 3H), 0.71 (s, 3H).385 Achiral separation Peak 2604.851H NMR (400 MHz, Methanol-d4) δ 8.84 (s, 1H), 8.13 (dd, J = 8.5, 1.4 Hz, 1H), 8.00-7.70 (m, 3H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.48 (d, J = 8.2 Hz, 2H), 7.44-7.23 (m, 3H), 6.91 (d, J = 8.2 Hz, 1H), 5.48 (s, 2H), 5.18-5.02 (m, 1H), 4.75-4.48 (m, 4H), 3.99 (d, J = 8.9 Hz, 1H), 3.85 (s, 1H), 3.74-3.61 (m, 1H), 1.40 (s, 3H), 0.74 (s, 3H).385 Bchiral separation Peak 1604.81H NMR (400 MHz, Methanol-d4) δ 8.82 (s, 1H), 8.15-8.01 (m, 1H), 7.98- 7.66 (m, 3H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.48 (d, J = 8.2 Hz, 2H), 7.42-7.34 (m, 3H), 6.90 (d, J = 8.2 Hz, 1H), 5.48 (s, 2H), 5.12- 4.99 (m, 1H), 4.74-4.43 (m, 4H), 3.98 (d, J = 8.9 Hz, 1H), 3.92-3.73 (m, 2H), 1.40 (s, 3H), 0.73 (s, 3H).38627582.651H NMR (400 MHz, DMSO-d6) δ 8.10 (s, 1H), 7.97-7.73 (m, 2H), 7.60- 7.31 (m, 7H), 6.95 (d, J = 8.2 Hz, 1H), 5.47 (s, 2H), 4.62 (t, J = 5.1 Hz, 2H), 4.46 (s, 2H), 3.68 (t, J = 5.0 Hz, 2H), 3.66-3.51 (m, 1H), 3.21 (s, 3H).38727605.251H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J = 8.6, 1.4 Hz, 1H), 7.90 (dd, J = 10.9, 6.3 Hz, 1H), 7.87-7.70 (m, 2H), 7.56 (dd, J = 7.3, 1.6 Hz, 1H), 7.53-7.44 (m, 2H), 7.43-7.31 (m, 3H), 6.92 (d, J = 8.2 Hz, 1H), 5.48 (s, 2H), 5.14 (d, J = 6.5 Hz, 1H), 4.78-4.60 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz,1H), 3.84 (d, J = 8.9 Hz,1H), 1.41 (s, 3H), 0.76 (s,3H).38835689.11H NMR (400 MHz, DMSO-d6) δ 8.93 (d, J = 1.2 Hz, 1H), 8.83 (s, 1H), 8.48 (s, 1H), 8.33 (s, 1H), 8.04 (d, J = 1.3 Hz, 1H), 7.80 (dd, J = 8.4, 1.4 Hz, 1H), 7.62 (t, J = 8.2 Hz, 2H), 7.56 (dd, J = 10.1, 6.5 Hz, 1H), 7.47- 7.43 (m, 1H), 7.40 (dd, J = 11.3, 8.6 Hz, 1H), 7.30- 7.23 (m, 1H), 5.46 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.58-4.48 (m, 2H), 4.44(dd, J = 11.1, 6.8 Hz, 1H),4.36 (d, J = 16.8 Hz, 1H),3.82-3.68 (m, 2H), 1.34 (s,3H), 0.60 (s, 3H).38935633.11H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J = 1.3 Hz, 1H), 8.84 (s, 1H), 8.80 (d, J = 5.2 Hz, 1H), 8.32 (s, 1H), 8.21 (s, 1H), 8.03 (d, J = 1.2 Hz, 1H), 7.97 (dd, J = 10.1, 6.3 Hz, 1H), 7.79 (dd, J = 8.6, 1.5 Hz, 1H), 7.65 (dd, J = 5.3, 1.8 Hz, 1H), 7.59 (d, J = 8.4 Hz, 1H), 7.48 (dd, J = 11.5, 5.8 Hz, 1H), 5.69 (s, 2H), 4.59 (d, J = 5.5 Hz, 2H), 4.48 (s, 2H), 3.68 (t, J = 5.0 Hz, 2H), 3.20 (s, 3H).39035649.11H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J = 1.3 Hz, 1H), 8.83 (s, 1H), 8.33 (s, 1H), 8.25 (d, J = 1.5 Hz, 1H), 8.04 (d, J = 1.3 Hz, 1H), 7.83 (dd, J = 8.4, 1.5 Hz, 1H), 7.62 (dd, J = 8.5, 3.5 Hz, 2H), 7.56 (dd, J = 10.2, 6.5 Hz, 1H), 7.39 (dd, J = 11.0, 7.5 Hz, 2H), 7.31- 7.19 (m, 1H), 5.45 (s, 2H), 4.63 (t, J = 5.2 Hz, 2H), 4.47 (s, 2H), 3.68 (d, J = 5.3 Hz, 2H), 3.21 (s, 3H).39138650.11H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.83 (s, 1H), 8.31 (s, 1H), 8.21 (d, J = 1.5 Hz, 1H), 8.03 (d, J = 1.3 Hz, 1H), 7.96 (t, J = 7.8 Hz, 1H), 7.82-7.73 (m, 2H), 7.58 (dd, J = 13.1, 7.9 Hz, 2H), 7.06 (d, J = 8.4 Hz, 1H), 5.67 (s, 2H), 4.65 (s, 2H), 4.53 (s, 2H), 3.72 (t, J = 5.0 Hz, 2H), 3.24 (s, 3H).39238668.11H NMR (400 MHz, DMSO-d6) δ 8.91 (d, J = 1.3 Hz, 1H), 8.85 (s, 1H), 8.32 (s, 1H), 8.21 (s, 1H), 8.03 (d, J = 1.3 Hz, 1H), 7.99- 7.86 (m, 1H), 7.80-7.70 (m, 1H), 7.63 (d, J = 8.5 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 5.76 (s, 2H), 4.65 (s, 2H), 4.52 (s, 2H), 3.72 (t, J = 4.9 Hz, 2H), 3.24 (s, 3H).39335638.21H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.89 (t, J = 7.9 Hz, 1H), 7.80 (dt, J = 10.4, 3.3 Hz, 3H), 7.63 (dd, J = 16.7, 8.4 Hz, 2H), 7.57-7.41 (m, 3H), 6.98 (d, J = 8.3 Hz, 1H), 5.48 (s, 2H), 5.01 (d, J = 6.7 Hz, 1H), 4.58-4.48 (m, 2H), 4.48-4.31 (m, 2H), 3.81-3.71 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H).39435622.21H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.89 (t, J = 7.9 Hz, 1H), 7.83- 7.75 (m, 2H), 7.61 (dt, J = 8.0, 3.8 Hz, 2H), 7.58-7.49 (m, 2H), 7.45 (dd, J = 11.2, 6.2 Hz, 1H), 7.39-7.35 (m, 1H), 6.98 (d, J = 8.2 Hz, 1H), 5.48 (s, 2H), 5.01 (d, J = 6.5 Hz, 1H), 4.59-4.48 (m, 2H), 4.48-4.31 (m, 2H), 3.81-3.68 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H).39635614.01H NMR (400 MHz, DMSO) δ 8.08 (d, J = 8.6 Hz, 1H), 7.96 (d, J = 8.3 Hz, 1H), 7.87 (dd, J = 9.9, 8.3 Hz, 3H), 7.77-7.67 (m, 3H), 7.55 (dd, J = 8.5, 2.4 Hz, 1H), 7.46 (dd, J = 11.4, 6.0 Hz, 1H), 5.67 (s, 2H), 4.96 (dd, J = 8.4, 5.3 Hz, 1H), 4.74 (dd, J = 9.6, 5.1 Hz, 1H), 4.58 (d, J = 17.1 Hz, 1H), 4.48 (d, J = 17.1 Hz, 1H), 4.37 (dt, J = 9.0, 5.0 Hz, 2H), 3.63 (d, J = 7.9 Hz, 1H), 1.24 (s, 3H), 0.62 (s, 3H).39735629.01H NMR (400 MHz, DMSO) δ 8.31 (s, 1H), 7.89 (dd, J = 17.8, 8.0 Hz, 3H), 7.78-7.65 (m, 4H), 7.53 (d, J = 7.4 Hz, 1H), 7.45 (t, J = 8.7 Hz, 1H), 7.01 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 5.00 (d, J = 6.5 Hz, 1H), 4.56- 4.47 (m, 2H), 4.46-4.31 (m, 3H), 4.36 (s, 15H), 4.16 (s, 1H), 3.78-3.68 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H).39835656.01H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.92- 7.69 (m, 4H), 7.64 (dd, J = 8.4, 6.1 Hz, 2H), 7.60-7.53 (m, 1H), 7.53-7.42 (m, 2H), 5.63 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.54 (dd, J = 15.0, 2.5 Hz, 2H), 4.48- 4.34 (m, 2H), 3.78 (d, J = 8.7 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 1.33 (s, 3H), 0.61 (s, 3H).39935638.01H NMR (400 MHz, DMSO) δ 8.50 (s, 1H), 7.90 (t, J = 7.9 Hz, 1H), 7.86- 7.75 (m, 2H), 7.70 (d, J = 2.1 Hz, 1H), 7.66-7.59 (m, 2H), 7.58-7.51 (m, 1H), 7.51-7.42 (m, 2H), 7.00 (d, J = 8.2 Hz, 1H), 5.54 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.73 (s, 8H), 4.55 (d, J = 17.4 Hz, 2H), 4.48-4.37 (m, 2H), 4.35 (s, 4H), 3.82- 3.70 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H).40035636.01H NMR (400 MHz, DMSO) δ 8.40 (s, 1H), 7.93- 7.80 (m, 2H), 7.61 (t, J = 8.1 Hz, 1H), 7.57-7.41 (m, 4H), 7.33 (dd, J = 8.3, 1.9 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 4.98 (d, J = 6.6 Hz, 1H), 4.57-4.47 (m, 2H), 4.47-4.34 (m, 2H), 3.77 (d, J = 8.7 Hz, 1H), 3.72 (d, J = 8.6 Hz, 1H), 2.58 (s, 3H), 1.31 (s, 3H), 0.60 (s, 3H).401609.1H NMR (400 MHz, DMSO) δ 8.40 (s, 1H), 7.95- 7.83 (m, 3H), 7.74 (dd, J = 10.4, 6.5 Hz, 1H), 7.68 (d, J = 8.0 Hz, 2H), 7.53 (d, J = 7.4 Hz, 1H), 7.45 (s, 1H), 7.50-7.40 (m, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.58 (s, 2H), 4.97 (d, J = 6.6 Hz, 1H), 4.52 (d, J = 7.5 Hz, 1H), 4.49 (s, 1H), 4.47- 4.33 (m, 2H), 3.80-3.68 (m, 2H), 2.58 (s, 3H), 1.31 (s, 3H), 0.59 (s, 3H).402356561H NMR (400 MHz, DMSO) δ 8.32 (s, 1H), 7.93- 7.79 (m, 2H), 7.73 (s, 1H), 7.61 (t, J = 8.2 Hz, 1H), 7.59- 7.41 (m, 3H), 7.33 (dd, J = 8.3, 2.0 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.00 (d, J = 6.5 Hz, 1H), 4.57-4.47 (m, 2H), 4.46- 4.32 (m, 2H), 3.78-3.68 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H).40338649.01H NMR (400 MHz, DMSO) δ 8.92 (s, 1H), 8.22 (s, 1H), 8.16 (d, J = 2.1 Hz, 1H), 8.03-7.92 (m, 3H), 7.87-7.75 (m, 2H), 7.75- 7.67 (m, 1H), 7.58 (t, J = 7.7 Hz, 2H), 7.08 (d, J = 8.3 Hz, 1H), 5.63 (s, 2H), 4.65 (t, J = 5.1 Hz, 2H), 4.53 (s, 2H), 3.73 (t, J = 5.0 Hz, 2H), 3.24 (s, 3H), 2.56 (d, J = 7.5 Hz, 1H).40435658.21H NMR (400 MHz, Chloroform-d) δ 8.31 (d, J = 5.1 Hz, 1H), 7.51 (t, J = 8.1 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 7.19-7.13 (m, 2H), 5.51-5.46 (m, 2H).; 1H NMR (400 MHz, DMSO) δ 8.78 (d, J = 5.1 Hz, 1H), 8.57 (d, J = 1.5 Hz, 1H), 7.92 (ddt, J = 10.3, 3.5, 1.7 Hz, 3H), 7.75 (dq, J = 11.3, 8.2 Hz, 3H), 7.62 (dd, J = 5.2, 1.8 Hz, 1H), 7.51 (dd,J = 11.5, 6.0 Hz, 1H), 5.81-5.70 (m, 1H), 5.62 (s, 2H),4.65 (s, 2H), 4.27 (dd, J =10.8, 7.9 Hz, 1H), 4.20 (dd,J = 10.8, 3.3 Hz, 1H), 2.40(dd, J = 13.2, 9.4 Hz, 1H),2.02 (dd, J = 13.3, 6.9 Hz,1H), 1.50 (s, 3H), 1.27 (s,3H).40535623.01H NMR (400 MHz, DMSO-d6) δ 8.72 (d, J = 5.8 Hz, 1H), 8.50 (s, 1H), 7.92 (dd, J = 10.1, 6.2 Hz, 1H), 7.82 (dd, J = 8.5, 1.5 Hz, 1H), 7.64 (dt, J = 8.2, 4.0 Hz, 2H), 7.50 (ddd, J = 16.8, 10.5, 4.0 Hz, 2H), 7.35 (dd, J = 8.4, 2.1 Hz, 1H), 7.04 (d, J = 5.8 Hz, 1H), 5.58 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.60-4.52 (m, 2H), 4.49-4.38 (m, 2H), 3.85- 3.69 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).40635620.01H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J = 1.4 Hz, 1H), 7.95-7.76 (m, 3H), 7.65-7.58 (m, 2H), 7.52 (ddd, J = 12.1, 8.8, 1.9 Hz, 2H), 7.43 (dd, J = 11.5, 6.1 Hz, 1H), 7.33 (dd, J = 8.3, 2.1 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 4.38 (s, 2H), 4.11 (s, 2H), 3.94 (d, J = 2.1 Hz, 2H), 2.90-2.77 (m, 2H), 2.69 (ddd, J = 12.3, 6.5, 3.7 Hz, 3H).40735620.01H NMR (400 MHz, DMSO-d6) δ 8.71 (d, J = 1.4 Hz, 1H), 7.92-7.81 (m, 3H), 7.67-7.58 (m, 2H), 7.55-7.43 (m, 3H), 7.34 (dd, J = 8.2, 2.1 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.46 (q, J = 4.5, 3.8 Hz, 1H), 4.74-4.63 (m, 2H), 4.60-4.40 (m, 3H), 3.08 (p, J = 5.5 Hz, 1H), 2.36 (ddd, J = 11.0, 5.8, 3.6 Hz, 1H), 2.28 (dd, J = 10.9, 9.2 Hz, 1H), 2.13-2.04 (m, 1H), 2.00 (dd, J = 10.7, 9.2 Hz, 1H).408chiral separation Peak 2629.31H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.91 (t, J = 9.9 Hz, 2H), 7.85- 7.68 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.40 (dd, J = 11.4, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 4.87 (dd, J = 9.0, 4.7 Hz, 1H), 4.63 (d, J = 16.9 Hz, 1H), 4.59-4.46 (m, 2H), 3.90 (dd, J = 10.7, 4.6 Hz, 1H), 3.84-3.77 (m, 2H), 3.14 (s, 3H), 1.99 (q, J = 4.6, 4.0 Hz, 3H).409chiral separation Peak 1629.31H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.91 (t, J = 9.9 Hz, 2H), 7.85- 7.68 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.40 (dd, J = 11.4, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 4.87 (dd, J = 9.0, 4.7 Hz, 1H), 4.63 (d, J = 16.9 Hz, 1H), 4.59-4.46 (m, 2H), 3.90 (dd, J = 10.7, 4.6 Hz, 1H), 3.84-3.77 (m, 2H), 3.14 (s, 3H), 1.99 (q, J = 4.6, 4.0 Hz, 3H).410356411H NMR (400 MHz, DMSO-d6) δ 8.83 (d, J = 2.8 Hz, 1H), 8.50 (s, 1H), 7.90 (dd, J = 10.1, 6.2 Hz, 1H), 7.81 (dd, J = 8.4, 1.5 Hz, 1H), 7.65 (q, J = 8.3 Hz, 2H), 7.55 (dd, J = 10.0, 2.0 Hz, 1H), 7.49 (dd, J = 11.0, 6.0 Hz, 1H), 131 (dd, J = 8.3, 2.0 Hz, 1H), 5.67 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.61-4.50 (m, 2H), 4.48-4.37 (m, 2H), 3.85- 3.69 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).411226411H NMR (400 MHz, DMSO-d6) δ 8.39 (d, J = 1.5 Hz, 1H), 8.00-7.89 (m, 2H), 7.84-7.71 (m, 4H), 7.60 (d, J = 8.4 Hz, 1H), 7.53 (d, J = 7.2 Hz, 1H), 7.37 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.59 (d, J = 14.5 Hz, 3H), 4.50 (s, 2H), 4.40 (ddd, J = 13.3, 8.4, 4.9 Hz, 2H), 4.19-4.09 (m, 1H), 4.04 (dd, J = 10.4, 8.2 Hz, 1H), 3.87 (dd, J = 10.2, 4.6 Hz, 1H), 2.91 (tt, J = 5.9, 2.9 Hz,1H), 0.22 (dd, J = 10.9, 5.2Hz, 1H), 0.11 (ddd, J = 12.6,9.1, 5.0 Hz, 2H), −0.04-−0.40 (m, 1H).412226651H NMR (400 MHz, DMSO-d6) δ 8.43 (s, 1H), 8.03-7.86 (m, 2H), 7.85- 7.67 (m, 4H), 7.60 (d, J = 8.5 Hz, 1H), 7.53 (d, J = 7.3 Hz, 1H), 7.34 (dd, J = 11.5, 6.1Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.87-5.45 (m, 3H), 4.54 (s, 2H), 4.51- 4.41 (m, 2H), 4.14 (s, 0H), 4.05 (dd, J = 10.5, 8.3 Hz, 1H), 3.93-3.83 (m, 2H), 3.62-3.43 (m, 1H), 3.24- 2.99 (m, 1H).413276631H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.43 (d, J = 1.9 Hz, 1H), 8.12 (dd, J = 8.3, 1.9 Hz, 1H), 7.99-7.88 (m, 2H), 7.81 (dd, J = 8.5, 1.5 Hz, 1H), 7.72 (dd, J = 10.4, 6.4 Hz, 1H), 7.63 (d, J = 8.5 Hz, 1H), 7.57 (dd, J = 7.5, 1.6 Hz, 1H), 7.46 (dd, J = 11.4, 6.0 Hz, 1H), 7.04 (d, J = 8.3 Hz, 1H), 5.75 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.59-4.50 (m, 2H), 4.48-4.35(m, 2H), 3.83-3.67 (m,2H), 1.33 (s, 3H), 0.60 (s,3H).414277061H NMR (400 MHz, DMSO-d6) δ 9.04 (s, 1H), 8.97 (d, J = 1.3 Hz, 1H), 8.50 (s, 1H), 8.41 (s, 1H), 8.06 (d, J = 1.2 Hz, 1H), 7.93 (t, J = 7.9 Hz, 1H), 7.87- 7.76 (m, 2H), 7.63 (d, J = 8.4 Hz, 1H), 7.57 (dd, J = 7.5, 1.6 Hz, 1H), 7.47 (dd, J = 11.4, 6.1 Hz, 1H), 7.02 (d, J = 8.3 Hz, 1H), 5.77 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.55 (d, J = 17.2 Hz, 2H), 4.49-4.35 (m, 2H), 3.88-3.58 (m, 2H), 1.32 (s, 3H), 0.60 (s, 3H).415356471H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 8.14 (d, J = 8.1 Hz, 1H), 7.92-7.85 (m, 2H), 7.78 (dd, J = 8.4, 1.5 Hz, 1H), 7.75-7.64 (m, 3H), 7.64- 7.54 (m, 2H), 5.69 (s, 2H), 5.08 (d, J = 6.6 Hz, 1H), 4.70-4.54 (m, 2H), 4.47 (dd, J = 11.1, 6.7 Hz, 1H), 4.35 (d, J = 17.3 Hz, 1H), 3.83-3.71 (m, 2H), 1.39 (s, 3H), 0.65 (s, 3H).416356651H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 8.15 (d, J = 8.0 Hz, 1H), 7.95 (d, J = 10.0 Hz, 1H), 7.77 (dd, J = 5.8, 2.7 Hz, 3H), 7.71 (dd, J = 10.6, 5.4 Hz, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.59 (d, J = 8.5 Hz, 1H), 5.72 (s, 2H), 5.09 (d, J = 6.5 Hz, 1H), 4.65 (d, J = 17.3 Hz, 1H), 4.57 (d, J = 11.1 Hz, 1H), 4.47 (dd, J = 11.1, 6.8 Hz, 1H), 4.35 (d, J = 17.2 Hz, 1H), 3.85-3.75 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H).417356401H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J = 6.5 Hz, 1H), 7.89 (t, J = 7.8 Hz, 1H), 7.83 (dd, J = 9.8, 7.0 Hz, 1H), 7.62 (d, J = 8.2 Hz, 1H), 7.56-7.47 (m, 3H), 7.47-7.42 (m, 1H), 7.33 (dd, J = 8.2, 2.0 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.00 (d, J = 6.5 Hz, 1H), 4.52 (d, J = 17.4 Hz, 2H), 4.47-4.30 (m, 2H), 3.82-3.67 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).418356131H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J = 6.5 Hz, 1H), 7.96-7.83 (m, 3H), 7.77-7.65 (m, 3H), 7.53 (d, J = 7.4 Hz, 1H), 7.45 (dd, J = 10.2, 7.0 Hz, 2H), 7.01 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 5.00 (d, J = 6.5 Hz, 1H), 4.51 (d, J = 15.7 Hz, 2H), 4.47-4.30 (m, 2H), 3.81-3.69 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H).419356311H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J = 6.4 Hz, 1H), 7.98-7.85 (m, 2H), 7.83-7.69 (m, 3H), 7.54 (d, J = 7.3 Hz, 1H), 7.46 (dd, J = 10.8, 7.4 Hz, 2H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.00 (d, J = 6.5 Hz, 1H), 4.52 (d, J = 17.0 Hz, 2H), 4.47-4.31 (m, 2H), 3.82-3.69 (m, 2H), 1.33 (s, 3H), 0.61 (s, 3H).420276211H NMR (400 MHz, DMSO-d6) δ 8.83 (d, J = 2.0 Hz, 1H), 8.50 (s, 1H), 8.12 (dd, J = 8.1, 2.0 Hz, 1H), 7.91 (t, J = 7.9 Hz, 1H), 7.88- 7.78 (m, 2H), 7.74 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.5 Hz, 1H), 7.55 (d, J = 7.3 Hz, 1H), 7.47 (dd, J = 10.9, 6.6 Hz, 1H), 7.16-6.78 (m, 2H), 5.61 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.55 (d, J = 16.8 Hz, 2H), 4.50-4.32 (m, 2H), 3.95-3.65 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).421355961H NMR (400 MHz, DMSO-d6) δ 9.03 (s, 1H), 8.62 (s, 1H), 7.97-7.83 (m, 3H), 7.75 (dd, J = 10.2, 6.5 Hz, 1H), 7.69 (d, J = 8.1 Hz, 2H), 7.54 (d, J = 7.3 Hz, 1H), 7.50 (dd, J = 11.2, 6.3 Hz, 1H), 7.02 (d, J = 8.3 Hz, 1H), 5.59 (s, 2H), 5.09 (d, J = 6.3 Hz, 1H), 4.62 (d, J = 17.3 Hz, 1H), 4.54 (d, J = 11.2 Hz, 1H), 4.49-4.37 (m, 2H), 3.80-3.71 (m, 2H), 1.37 (s, 3H), 0.63 (s, 3H).422356141H NMR (400 MHz, DMSO-d6) δ 9.02 (s, 1H), 8.61 (s, 1H), 7.98-7.88 (m, 2H), 7.84-7.71 (m, 3H), 7.55 (d, J = 7.4 Hz, 1H), 7.50 (dd, J = 11.0, 6.4 Hz, 1H), 7.01 (d, J = 8.2 Hz, 1H), 5.61 (s, 2H), 5.09 (d, J = 6.1 Hz, 1H), 4.62 (d, J = 17.3 Hz, 1H), 4.54 (d, J = 11.2 Hz, 1H), 4.50-4.36 (m, 2H), 3.80-3.74 (m, 2H), 1.37 (s, 3H), 0.63 (s, 3H).423356231H NMR (400 MHz, DMSO-d6) δ 9.01 (s, 1H), 8.61 (s, 1H), 7.96-7.80 (m, 2H), 7.61 (t, J = 8.2 Hz, 1H), 7.58-7.45 (m, 3H), 7.34 (dd, J = 8.2, 1.9 Hz, 1H), 6.97 (d, J = 8.2 Hz, 1H), 5.52 (s, 2H), 5.09 (d, J = 6.2 Hz, 1H), 4.62 (d, J = 17.1 Hz, 1H), 4.54 (d, J = 11.2 Hz, 1H), 4.50-4.38 (m, 2H), 3.79-3.73 (m, 2H), 1.37 (s, 3H), 0.63 (s, 3H).424276691H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.36 (d, J = 2.2 Hz, 1H), 7.95 (dd, J = 8.5, 2.4 Hz, 1H), 7.88 (td, J = 8.9, 7.9, 4.5 Hz, 2H), 7.84-7.77 (m, 1H), 7.62 (d, J = 8.5 Hz, 1H), 7.53 (d, J = 7.3 Hz, 1H), 7.47 (dd, J = 10.8, 6.8 Hz, 1H), 7.02 (d, J = 8.5 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.47 (s, 2H), 5.00 (q, J = 8.8 Hz, 3H), 4.60-4.48 (m, 2H), 4.48-4.32 (m, 2H), 3.83-3.71 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).425276391H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J = 2.0 Hz, 1H), 8.49 (s, 1H), 8.20 (d, J = 8.1 Hz, 1H), 7.93 (dd, J = 17.1, 8.2 Hz, 2H), 7.79 (dd, J = 9.3, 7.5 Hz, 2H), 7.62 (d, J = 8.5 Hz, 1H), 7.55 (d, J = 7.4 Hz, 1H), 7.46 (dd, J = 10.7, 6.8 Hz, 1H), 7.02 (d, J = 8.3 Hz, 1H), 5.65 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.58-4.49 (m, 2H), 4.49-4.30 (m, 2H), 3.85-3.71 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H).42635601.01H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.92-7.79 (m, 3H), 7.71 (dd, J = 6.8, 2.0 Hz, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.57-7.50 (m, 2H), 7.46 (dd, J = 11.3, 6.1 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 6.25 (t, J = 6.8 Hz, 1H), 5.29 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.61-4.52 (m, 2H), 4.48-4.36 (m, 2H), 3.86- 3.68 (m, 2H), 3.48 (s, 3H), 1.34 (s, 3H), 0.61 (d, J = 3.4 Hz, 3H).42735601.01H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.91 (t, J = 7.9 Hz, 1H), 7.80 (dd, J = 9.3, 7.8 Hz, 2H), 7.68 (d, J = 7.0 Hz, 1H), 7.63 (d, J = 8.5 Hz, 1H), 7.54 (d, J = 7.4 Hz, 1H), 7.46 (dd, J = 10.1, 7.4 Hz, 1H), 7.02 (d, J = 8.3 Hz, 1H), 6.42 (s, 1H), 6.29 (dd, J = 7.0, 1.8 Hz, 1H), 5.34 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.58-4.50 (m, 2H), 4.48-4.35 (m, 2H), 3.85- 3.66 (m, 2H), 3.40 (s, 3H), 1.34 (s, 3H), 0.61 (s, 3H42835632.01H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 8.3 Hz, 1H), 7.95 (dd, J = 9.3, 7.6 Hz, 2H), 7.88 (dd, J = 10.2, 8.2 Hz, 1H), 7.81- 7.70 (m, 3H), 7.59-7.53 (m, 1H), 7.47 (dd, J = 11.4, 6.1 Hz, 1H), 5.70 (s, 2H), 4.96 (dd, J = 8.5, 5.3 Hz, 1H), 4.74 (dd, J = 9.6, 5.2 Hz, 1H), 4.65-4.44 (m, 2H), 4.37 (dt, J = 8.9, 5.1 Hz, 2H), 3.63 (d, J = 7.9 Hz, 1H), 1.24 (s, 3H), 0.62 (s, 3H).42935589.0.1H NMR (400 MHz, DMSO-d6) δ 8.61 (d, J = 1.7 Hz, 1H), 8.50 (s, 1H), 8.45 (d, J = 4.9 Hz, 1H), 7.92 (t, J = 7.9 Hz, 1H), 7.82 (dd, J = 8.4, 1.4 Hz, 1H), 7.74 (dd, J = 10.5, 6.4 Hz, 1H), 7.64 (d, J = 8.5 Hz, 1H), 7.61-7.53 (m, 2H), 7.46 (dd, J = 11.4, 6.1 Hz, 1H), 7.03 (d, J = 8.2 Hz, 1H), 5.62 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.55 (d, J = 16.4 Hz, 2H), 4.51-4.35 (m, 2H), 3.83-3.71(m, 2H), 1.33 (s, 3H), 0.61(s, 3H).43035601.11H NMR (400 MHz, DMSO-d6) δ 8.61 (s, 1H), 8.49 (s, 1H), 8.44 (d, J = 5.3 Hz, 1H), 7.94 (t, J = 7.9 Hz, 1H), 7.81 (dd, J = 8.5, 1.4 Hz, 1H), 7.75 (d, J = 5.4 Hz, 1H), 7.67 (t, J = 8.4 Hz, 1H), 7.62 (d, J = 8.4 Hz, 1H), 7.56 (d, J = 7.4 Hz, 1H), 7.46 (t, J = 8.7 Hz, 1H), 7.09 (d, J = 8.3 Hz, 1H), 5.63 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.60-4.48 (m, 2H),4.48-4.31 (m, 2H), 4.05 (s,3H), 3.87-3.65 (m, 2H),1.33 (s, 3H), 0.61 (s, 3H).43135610.01H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.94-7.75 (m, 3H), 7.64 (d, J = 8.4 Hz, 1H), 7.58-7.41 (m, 2H), 7.36 (d, J = 7.8 Hz, 2H), 7.09 (d, J = 7.8 Hz, 2H), 6.92 (d, J = 8.2 Hz, 1H), 5.41 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.55 (d, J = 16.6 Hz, 2H), 4.50-4.33 (m, 2H), 3.86-3.67 (m, 2H), 1.91 (tt, J = 8.7, 5.0 Hz,1H), 1.34 (s, 3H), 0.99-0.85 (m, 2H), 0.66 (dt, J =6.5, 4.6 Hz, 2H), 0.61 (s,3H).43235629.01H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.09 (d, J = 8.0 Hz, 1H), 7.90 (d, J = 8.2 Hz, 2H), 7.82-7.74 (m, 2H), 7.70 (d, J = 8.0 Hz, 2H), 7.62 (d, J = 8.4 Hz, 1H), 7.60-7.52 (m, 1H), 7.48 (dd, J = 11.4, 6.1 Hz, 1H), 5.69 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.59- 4.51 (m, 2H), 4.51-4.34 (m, 2H), 3.82-3.67 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).43335638.01H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.07 (d, J = 8.0 Hz, 1H), 7.88-7.78 (m, 2H), 7.62 (d, J = 8.5 Hz, 1H), 7.58-7.51 (m, 3H), 7.51-7.41 (m, 3H), 5.58 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.57-4.50 (m, 2H), 4.49-4.33 (m, 2H), 3.83-3.68 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).43435647.01H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.10 (d, J = 8.0 Hz, 1H), 7.95 (d, J = 10.0 Hz, 1H), 7.83-7.74 (m, 4H), 7.62 (d, J = 8.5 Hz, 1H), 7.58 (dd, J = 8.1, 1.4 Hz, 1H), 7.48 (dd, J = 11.4, 6.1 Hz, 1H), 5.71 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.59-4.49 (m, 2H), 4.48-4.34 (m, 2H), 3.83- 3.68 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).435chiral separation Peak 2615.01H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.98-7.85 (m, 2H), 7.85- 7.70 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.37 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.53 (d, J = 7.6 Hz, 1H), 4.55 (s, 2H), 4.50 (d, J = 10.8 Hz, 1H), 4.21 (dd, J = 10.9, 6.5 Hz, 1H), 4.06 (t, J = 8.7 Hz, 1H), 3.80 (t, J = 8.3 Hz, 1H),3.18-3.09 (m, 1H), 3.00 (q,J = 7.8 Hz, 1H), 2.75 (t, J =9.5 Hz, 1H).436chiral separation Peak 1615.01H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.98-7.85 (m, 2H), 7.85- 7.70 (m, 4H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.3 Hz, 1H), 7.37 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 5.53 (d, J = 7.6 Hz, 1H), 4.55 (s, 2H), 4.50 (d, J = 10.8 Hz, 1H), 4.21 (dd, J = 10.9, 6.5 Hz, 1H), 4.06 (t, J = 8.7 Hz, 1H), 3.80 (t, J = 8.3 Hz, 1H), 3.18-3.09 (m, 1H), 3.00 (q, J = 7.8 Hz, 1H), 2.75 (t, J = 9.5 Hz, 1H).43735623.01H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J = 8.3 Hz, 1H), 7.96 (d, J = 8.3 Hz, 1H), 7.92-7.80 (m, 2H), 7.60 (t, J = 8.2 Hz, 1H), 7.57- 7.44 (m, 3H), 7.33 (dd, J = 8.2, 2.0 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 4.97 (dd, J = 8.4, 5.3 Hz, 1H), 4.74 (dd, J = 9.6, 5.2 Hz, 1H), 4.64-4.47 (m, 2H), 4.37 (dt, J = 8.9, 5.1 Hz, 2H), 3.63 (d, J = 7.9 Hz, 1H), 1.24 (s, 3H), 0.62 (s, 3H).43835605.01H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 8.3 Hz, 1H), 7.96 (d, J = 8.3 Hz, 1H), 7.88 (t, J = 7.9 Hz, 1H), 7.82 (dd, J = 10.3, 6.6 Hz, 1H), 7.58-7.40 (m, 6H), 6.96 (d, J = 8.3 Hz, 1H), 5.47 (s, 2H), 4.97 (dd, J = 8.4, 5.3 Hz, 1H), 4.75 (dd, J = 9.6, 5.2 Hz, 1H), 4.65- 4.45 (m, 2H), 4.37 (dt, J = 8.9, 5.1 Hz, 2H), 3.63 (d, J = 7.9 Hz, 1H), 1.24 (s, 3H), 0.63 (s, 3H).43935614.01H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 8.3 Hz, 1H), 8.00-7.85 (m, 3H), 7.84-7.68 (m, 3H), 7.58-7.52 (m, 1H), 7.46 (dd, J = 11.3, 6.2 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.61 (s, 2H), 4.96 (dd, J = 8.4, 5.3 Hz, 1H), 4.74 (dd, J = 9.6, 5.1 Hz, 1H), 4.64- 4.46 (m, 2H), 4.37 (dt, J = 8.9, 5.1 Hz, 2H), 3.63 (d, J = 7.9 Hz, 1H), 1.23 (s, 3H), 0.62 (s, 3H).44035596.01H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 8.3 Hz, 1H), 7.96 (d, J = 8.3 Hz, 1H), 7.93-7.83 (m, 3H), 7.74 (dd, J = 10.2, 6.6 Hz, 1H), 7.68 (d, J = 8.0 Hz, 2H), 7.58-7.51 (m, 1H), 7.46 (dd, J = 11.2, 6.3 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.58 (s, 2H), 4.96 (dd, J = 8.5, 5.2 Hz, 1H), 4.74 (dd, J = 9.6, 5.1 Hz, 1H), 4.64- 4.44 (m, 2H), 4.37 (dt, J =9.0, 5.1 Hz, 2H), 3.63 (d, J =7.9 Hz, 1H), 1.24 (s, 3H),0.62 (s, 3H).44135656.01H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 7.96-7.74 (m, 3H), 7.55 (d, J = 8.3 Hz, 3H), 7.51-7.39 (m, 3H), 5.57 (s, 2H), 5.03 (d, J = 6.5 Hz, 1H), 4.54 (t, J = 13.8 Hz, 2H), 4.47-4.36 (m, 2H), 3.84-3.68 (m, 2H), 1.32 (s, 3H), 0.60 (s, 3H).44241669.21H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J = 8.6, 1.4 Hz, 1H), 7.96-7.81 (m, 3H), 7.81-7.67 (m, 2H), 7.59 (dd, J = 7.3, 1.6 Hz, 1H), 7.49-7.35 (m, 3H), 7.06- 6.75 (m, 1H), 5.64 (s, 2H), 5.14 (d, J = 6.5 Hz, 1H), 4.80-4.61 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 4.13 (s, 3H), 4.00 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 1.41 (s, 3H), 0.76 (s, 3H).44341704.11H NMR (400 MHz, Methanol-d4) δ 8.93 (s, 1H), 8.49 (d, J = 0.7 Hz, 1H), 8.21 (dd, J = 8.7, 1.4 Hz, 1H), 8.11 (s, 1H), 7.95 (dd, J = 10.9, 6.3 Hz, 1H), 7.89- 7.72 (m, 2H), 7.71-7.31 (m, 6H), 6.93 (dd, J = 8.3, 0.7 Hz, 1H), 5.57 (s, 2H), 5.17 (d, J = 6.4 Hz, 1H), 4.81-4.57 (m, 3H), 4.53 (dd, J = 11.7, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 1.41 (s, 3H), 0.77 (s, 3H).44441685.11H NMR (400 MHz, Methanol-d4) δ 8.93 (s, 1H), 8.21 (dd, J = 8.6, 1.4 Hz, 1H), 8.01-7.90 (m, 2H), 7.90-7.75 (m, 2H), 7.59 (ddd, J = 8.2, 5.0, 3.4 Hz, 2H), 7.51-7.32 (m, 3H), 7.02-6.87 (m, 1H), 5.58 (s, 2H), 5.17 (d, J = 6.5 Hz, 1H), 4.83-4.61 (m, 3H), 4.52 (dd, J = 11.7, 6.7 Hz, 1H), 4.00 (d, J = 9.0 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 2.74 (s, 3H), 1.41 (s, 3H), 0.77 (s, 3H).44541669.21H NMR (400 MHz, Methanol-d4) δ 8.93 (s, 1H), 8.21 (dd, J = 8.6, 1.4 Hz, 1H), 7.94 (dd, J = 10.9, 6.3 Hz, 1H), 7.90-7.76 (m, 2H), 7.67-7.53 (m, 2H), 7.54-7.37 (m, 4H), 6.93 (d, J = 8.2 Hz, 1H), 5.57 (s, 2H), 5.17 (d, J = 6.4 Hz, 1H), 4.83-4.61 (m, 3H), 4.53 (dd, J = 11.7, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 2.53 (s, 3H), 1.41 (s, 3H), 0.77 (s, 3H).44627595.31H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J = 8.6, 1.4 Hz, 1H), 7.94-7.80 (m, 2H), 7.79 (d, J = 8.1 Hz, 2H), 7.75-7.66 (m, 2H), 7.58 (dd, J = 7.5, 1.6 Hz, 1H), 7.51 (qd, J = 7.6, 1.6 Hz, 1H), 7.40 (dd, J = 11.2, 6.0 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.68 (s, 2H), 5.15 (d, J = 6.5 Hz, 1H), 4.79-4.60 (m, 3H), 4.53 (dd, J = 11.7,6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.84 (d, J = 8.9 Hz,1H), 1.41 (s, 3H), 0.76 (s,3H).44735672.21H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (dd, J = 8.6, 1.4 Hz, 1H), 7.96 (dd, J = 10.7, 6.3 Hz, 1H), 7.77 (d, J = 8.6 Hz, 1H), 7.70 (s, 1H), 7.56 (t, J = 8.0 Hz, 1H), 7.43 (dd, J = 11.3, 6.0 Hz, 1H), 7.31- 7.19 (m, 2H), 7.07 (s, 1H), 6.88 (t, J = 55.4 Hz, 1H), 5.58 (s, 2H), 5.13 (d, J = 6.6 Hz, 1H), 4.79-4.59 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 1.42 (s, 3H), 0.76 (s, 3H).448276371H NMR (400 MHz, Methanol-d4) δ 9.10 (s, 1H), 8.90 (s, 1H), 8.23-8.08 (m, 2H), 7.91 (dd, J = 10.9, 6.3 Hz, 1H), 7.88-7.81 (m, 3H), 7.78 (d, J = 8.6 Hz, 1H), 7.68 (d, J = 8.5 Hz, 2H), 7.58 (dd, J = 7.5, 1.6 Hz, 1H), 7.40 (dd, J = 11.2, 6.0 Hz, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.58 (s, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.79- 4.57 (m, 3H), 4.52 (dd, J =11.6, 6.7 Hz, 1H), 4.00 (d,J = 8.9 Hz, 1H), 3.84 (d, J =8.9 Hz, 1H), 1.40 (s, 3H),0.75 (s, 3H).44927629.21H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J = 8.6, 1.5 Hz, 1H), 7.93-7.81 (m, 3H), 7.78 (d, J = 8.6 Hz, 1H), 7.71 (d, J = 1.6 Hz, 2H), 7.59 (dd, J = 7.4, 1.6 Hz, 1H), 7.38 (dd, J = 11.1, 6.0 Hz, 1H), 6.97 (d, J = 8.2 Hz, 1H), 5.66 (s, 2H), 5.13 (d, J = 6.6 Hz, 1H), 4.81-4.60 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.84 (d, J = 8.8 Hz,1H), 1.41 (s, 3H), 0.76 (s,3H).45027636.31H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J = 8.6, 1.3 Hz, 1H), 7.94 (dd, J = 10.9, 6.3 Hz, 1H), 7.90-7.73 (m, 2H), 7.58 (d, J = 7.5 Hz, 1H), 7.48 (d, J = 7.8 Hz, 1H), 7.39 (dd, J = 11.2, 6.0 Hz, 1H), 7.22 (d, J = 9.7 Hz, 1H), 6.91 (d, J = 8.3 Hz, 1H), 5.50 (s, 2H), 5.14 (d, J = 6.3 Hz, 1H), 4.81-4.61 (m, 3H), 4.53 (dd, J = 11.6,6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.85 (d, J = 8.9 Hz,1H), 2.34 (s, 3H), 1.42 (s,3H), 0.76 (s, 3H).45127624.31H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.20 (dd, J = 8.6, 1.4 Hz, 1H), 7.94 (dd, J = 10.8, 6.3 Hz, 1H), 7.89-7.74 (m, 2H), 7.59 (dd, J = 7.5, 1.6 Hz, 1H), 7.48-7.31 (m, 2H), 7.13 (tdd, J = 9.2, 7.1, 2.1 Hz, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.55 (s, 2H), 5.16 (d, J = 6.6 Hz, 1H), 4.83- 4.60 (m, 3H), 4.53 (dd, J = 11.7, 6.7 Hz, 1H), 4.01 (d,J = 8.9 Hz, 1H), 3.85 (d, J =8.9 Hz, 1H), 1.42 (s, 3H),0.77 (s, 3H).45227618.11H NMR (400 MHz, Methanol-d4) δ 8.93 (s, 1H), 8.21 (d, J = 8.5 Hz, 1H), 7.90 (ddd, J = 11.1, 6.5, 4.8 Hz, 1H), 7.87-7.77 (m, 2H), 7.64-7.53 (m, 1H), 7.47-7.39 (m, 2H), 7.38- 7.12 (m, 2H), 7.02-6.78 (m, 1H), 5.66-5.39 (m, 2H), 5.17 (d, J = 6.5 Hz, 1H), 4.81-4.60 (m, 3H), 4.53 (dd, J =11.7, 6.7 Hz, 1H), 4.01 (d, J = 8.9 Hz,1H), 3.85 (d, J = 8.9 Hz,1H), 2.52-2.31 (m, 3H),1.42 (s, 3H), 0.77 (s, 3H).45327640.21H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.18 (dd, J = 8.6, 1.3 Hz, 1H), 8.04 (dd, J = 10.9, 6.3 Hz, 1H), 7.91-7.65 (m, 2H), 7.67-7.44 (m, 1H), 7.40 (dd, J = 11.2, 6.0 Hz, 1H), 7.17 (d, J = 7.3 Hz, 2H), 6.85 (d, J = 8.3 Hz, 1H), 5.55 (s, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.81-4.60 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.85 (d, J = 8.9 Hz,1H), 1.42 (s, 3H), 0.77 (s,3H).45427640.31H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 1H), 8.15 (dd, J = 8.6, 1.4 Hz, 1H), 7.91-7.79 (m, 2H), 7.76 (d, J = 8.5 Hz, 1H), 7.60 (dd, J = 7.5, 1.6 Hz, 1H), 7.56-7.44 (m, 2H), 7.37 (dd, J = 11.2, 6.0 Hz, 1H), 6.98 (d, J = 8.2 Hz, 1H), 5.55 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.76-4.60 (m, 3H), 4.52 (dd, J = 11.6, 6.8 Hz, 1H), 3.99 (d, J = 8.9Hz, 1H), 3.84 (d, J = 8.8 Hz,1H), 1.41 (s, 3H), 0.75 (s,3H).45527624.11H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (dd, J = 8.6, 1.4 Hz, 1H), 7.93 (dd, J = 10.8, 6.3 Hz, 1H), 7.83 (t, J = 7.9 Hz, 1H), 7.77 (d, J = 8.6 Hz, 1H), 7.59 (dd, J = 7.4, 1.6 Hz, 1H), 7.49 (ddd, J = 10.8, 8.9, 6.6 Hz, 1H), 7.38 (dd, J = 11.2, 6.0 Hz, 1H), 7.22 (td, J = 10.1, 6.6 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.52 (s, 2H), 5.21-5.06 (m,1H), 4.78-4.58 (m, 3H),4.53 (dd, J = 11.5, 6.7 Hz,1H), 4.00 (d, J = 8.9 Hz,1H), 3.84 (d, J = 8.9 Hz,1H), 1.41 (s, 3H), 0.75 (s,3H).456620.31H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.18 (dd, J = 8.6, 1.4 Hz, 1H), 7.93 (dd, J = 10.9, 6.3 Hz, 1H), 7.89-7.73 (m, 2H), 7.58 (dd, J = 7.5, 1.5 Hz, 1H), 7.39 (dd, J = 11.2, 6.0 Hz, 1H), 7.20 (dd, J = 9.7, 6.0 Hz, 1H), 7.04 (dd, J = 10.1, 6.1 Hz, 1H), 6.92 (d, J = 8.3 Hz, 1H), 5.50 (s, 2H), 5.14 (d, J = 6.6 Hz, 1H), 4.81-4.60 (m, 3H),4.53 (dd, J = 11.7, 6.7 Hz,1H), 4.00 (d, J = 8.9 Hz,1H), 3.84 (d, J = 8.9 Hz,1H), 2.26 (d, J = 1.9 Hz,3H), 1.41 (s, 3H), 0.76 (s,3H).45727606.21H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (dd, J = 8.6, 1.4 Hz, 1H), 7.91 (dd, J = 10.8, 6.3 Hz, 1H), 7.87-7.73 (m, 2H), 7.57 (dd, J = 7.4, 1.6 Hz, 1H), 7.46-7.35 (m, 2H), 7.35-7.17 (m, 2H), 6.93 (d, J = 8.3 Hz, 1H), 5.47 (s, 2H), 5.13 (d, J = 6.5 Hz, 1H), 4.82-4.60 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz,1H), 3.84 (d, J = 8.9 Hz,1H), 1.41 (s, 3H), 0.75 (s,3H).45827624.31H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.17 (dd, J = 8.7, 1.4 Hz, 1H), 8.05 (dd, J = 10.9, 6.3 Hz, 1H), 7.88-7.75 (m, 2H), 7.59 (dd, J = 7.5, 1.6 Hz, 1H), 7.40 (dd, J = 11.3, 6.0 Hz, 1H), 7.01-6.89 (m, 2H), 6.84 (d, J = 8.3 Hz, 1H), 5.54 (s, 2H), 5.14 (d, J = 7.1 Hz, 1H), 4.81-4.60 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9Hz, 1H), 3.85 (d, J = 8.9 Hz,1H), 1.42 (s, 3H), 0.76 (s,3H).45927636.21H NMR (400 MHz, Methanol-d4) δ 8.86 (s, 1H), 8.14 (dd, J = 8.6, 1.4 Hz, 1H), 7.92 (dd, J = 10.8, 6.3 Hz, 1H), 7.86-7.73 (m, 2H), 7.57 (dd, J = 7.5, 1.6 Hz, 1H), 7.53 (d, J = 6.7 Hz, 1H), 7.36 (dd, J = 11.2, 6.0 Hz, 1H), 7.12 (d, J = 10.4 Hz, 1H), 6.92 (d, J = 8.3 Hz, 1H), 5.50 (s, 2H), 5.11 (d, J = 6.5 Hz, 1H), 4.79-4.58 (m, 3H), 4.52 (dd, J = 11.5,6.7 Hz, 1H), 3.99 (d, J = 8.9Hz, 1H), 3.84 (d, J = 8.8 Hz,1H), 2.37 (s, 3H), 1.41 (s,3H), 0.75 (s, 3H).46027629.151H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (d, J = 8.6 Hz, 1H), 7.98-7.83 (m, 2H), 7.83- 7.66 (m, 4H), 7.60 (d, J = 7.4 Hz, 1H), 7.37 (dd, J = 11.2, 6.0 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.67 (s, 2H), 5.12 (d, J = 6.4 Hz, 1H), 4.74-4.58 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 3.99 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H).46127625.21H NMR (400 MHz, Methanol-d4) δ 8.84 (s, 1H), 8.12 (dd, J = 8.6, 1.4 Hz, 1H), 7.89-7.77 (m, 2H), 7.75 (d, J = 8.6 Hz, 1H), 7.58 (d, J = 7.7 Hz, 2H), 7.38 (d, J = 1.4 Hz, 1H), 7.37-7.26 (m, 2H), 6.96 (d, J = 8.3 Hz, 1H), 5.58 (s, 2H), 5.08 (d, J = 6.6 Hz, 1H), 4.74-4.47 (m, 4H), 3.97 (d, J = 2.4 Hz, 4H), 3.84 (d, J = 8.9 Hz, 1H), 1.40 (s, 3H), 0.74 (s, 3H).46227634.31H NMR (400 MHz, Methanol-d4) δ 8.77 (s, 1H), 8.10-8.01 (m, 1H), 7.88 (dd, J = 10.9, 6.3 Hz, 1H), 7.79 (t, J = 7.9 Hz, 1H), 7.72 (d, J = 8.5 Hz, 1H), 7.55 (dd, J = 7.5, 1.5 Hz, 1H), 7.39 (d, J = 8.1 Hz, 1H), 7.27 (dd, J = 11.3, 6.1 Hz, 1H), 7.05 (d, J = 1.9 Hz, 1H), 6.95 (dd, J = 8.1, 2.0 Hz, 1H), 6.88 (d, J = 8.2 Hz, 1H), 5.48 (s, 2H),5.02 (d, J = 6.7 Hz, 1H),4.69-4.46 (m, 4H), 3.97 (d,J = 8.8 Hz, 1H), 3.91 (s,3H), 3.82 (d, J = 8.8 Hz,1H), 1.39 (s, 3H), 0.71 (s,3H).46327602.31H NMR (400 MHz, Methanol-d4) δ 8.96 (s, 1H), 8.23 (dd, J = 8.6, 1.4 Hz, 1H), 7.96 (dd, J = 10.9, 6.3 Hz, 1H), 7.89-7.74 (m, 2H), 7.56 (dd, J = 7.5, 1.6 Hz, 1H), 7.51-7.33 (m, 2H), 6.97 (dd, J = 14.8, 9.5 Hz, 2H), 6.88 (d, J = 8.2 Hz, 1H), 5.49 (s, 2H), 5.20 (d, J = 6.4 Hz, 1H), 4.89-4.60 (m, 3H), 4.53 (dd, J = 11.8, 6.6 Hz, 1H), 4.01 (d, J = 9.0Hz, 1H), 3.85 (d, J = 8.9 Hz,1H), 2.34 (s, 3H), 1.42 (s,3H), 0.78 (s, 3H).46427622.21H NMR (400 MHz, Methanol-d4) δ 8.94 (s, 1H), 8.21 (dd, J = 8.7, 1.4 Hz, 1H), 7.91 (dd, J = 10.9, 6.3 Hz, 1H), 7.88-7.73 (m, 2H), 7.68-7.54 (m, 2H), 7.43 (dd, J = 11.1, 6.1 Hz, 1H), 7.28 (dd, J = 8.6, 2.6 Hz, 1H), 7.10 (td, J = 8.5, 2.6 Hz, 1H), 6.94 (d, J = 8.3 Hz, 1H), 5.56 (s, 2H), 5.18 (d, J = 6.5 Hz, 1H), 4.85- 4.60 (m, 3H), 4.53 (dd, J =11.7, 6.7 Hz, 1H), 4.00 (d,J = 8.9 Hz, 1H), 3.85 (d, J =8.9 Hz, 1H), 1.42 (s, 3H),0.77 (s, 3H).46527602.31H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.20 (dd, J = 8.6, 1.4 Hz, 1H), 7.95 (dd, J = 10.8, 6.4 Hz, 1H), 7.85-7.70 (m, 2H), 7.56 (dd, J = 7.4, 1.7 Hz, 1H), 7.43 (td, J = 10.9, 9.7, 5.9 Hz, 2H), 6.99 (dd, J = 9.9, 2.8 Hz, 1H), 6.90 (dd, J = 11.5, 8.4 Hz, 2H), 5.47 (s, 2H), 5.16 (d, J = 6.4 Hz, 1H), 4.83-4.61 (m, 3H), 4.53 (dd, J = 11.7, 6.7 Hz,1H), 4.01 (d, J = 8.9 Hz,1H), 3.85 (d, J = 8.9 Hz,1H), 2.43 (s, 3H), 1.42 (s,3H), 0.77 (s, 3H).46627606.31H NMR (400 MHz, Methanol-d4) δ 8.91 (s, 1H), 8.19 (dd, J = 8.6, 1.4 Hz, 1H), 7.96 (dd, J = 10.8, 6.3 Hz, 1H), 7.89-7.70 (m, 2H), 7.65-7.52 (m, 2H), 7.41 (dd, J = 11.2, 6.0 Hz, 1H), 7.10-6.94 (m, 2H), 6.90 (d, J = 8.2 Hz, 1H), 5.52 (s, 2H), 5.16 (d, J = 6.4 Hz, 1H), 4.82-4.60 (m, 3H), 4.53 (dd, J = 11.6, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz,1H), 3.85 (d, J = 8.9 Hz,1H), 1.42 (s, 3H), 0.77 (s,3H).46735629.11H NMR (400 MHz, DMSO) δ 7.89 (dd, J = 16.6, 8.2 Hz, 3H), 7.70 (dd, J = 12.0, 7.3 Hz, 2H), 7.52 (d, J = 7.4 Hz, 1H), 7.39-7.30 (m, 1H), 7.01 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 5.49 (s, 1H), 4.48 (t, J = 13.9 Hz, 2H), 4.35 (t, J = 5.1 Hz, 1H), 4.26-4.14 (m, 1H), 2.92 (s, 3H), 2.08 (s, 2H).46835611.21H NMR (400 MHz, DMSO) δ 7.89 (dd, J = 16.6, 8.2 Hz, 3H), 7.70 (dd, J = 12.0, 7.3 Hz, 2H), 7.52 (d, J = 7.4 Hz, 1H), 7.40-7.29 (m, 1H), 7.01 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 5.49 (s, IH), 4.48 (t, J = 13.9 Hz, 2H), 4.35 (t, J = 5.1 Hz, 1H), 4.29-4.16 (m, 1H), 2.92 (s, 3H), 2.08 (s, 2H).46935672.11H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 7.97- 7.85 (m, 2H), 7.85-7.73 (m, 3H), 7.70 (dd, J = 10.5, 6.4 Hz, 1H), 7.62 (d, J = 8.5 Hz, 1H), 7.55 (d, J = 7.3 Hz, 1H), 7.44 (dd, J = 11.4, 6.1 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.64 (s, 2H), 5.00 (d, J = 6.7 Hz, 1H), 4.63-4.47 (m, 2H), 4.47-4.31 (m, 2H), 3.80-3.66 (m, 2H), 2.55 (s, 3H), 1.32 (s, 3H), 0.59 (s, 3H).47035640.01H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 8.29 (d, J = 8.8 Hz, 1H), 8.10 (d, J = 8.8 Hz, 1H), 7.94 (t, J = 7.9 Hz, 1H), 7.83 (dd, J = 8.5, 1.5 Hz, 1H), 7.73-7.51 (m, 3H), 7.45 (dd, J = 11.4, 6.1 Hz, 1H), 7.09 (d, J = 8.3 Hz, 1H), 5.90 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.63- 4.31 (m, 4H), 3.85-3.65 (m, 2H), 1.32 (s, 3H), 0.60 (s, 3H).47139626.01H NMR (400 MHz, DMSO) δ 8.50 (s, 1H), 7.97- 7.73 (m, 3H), 7.71-7.50 (m, 4H), 7.45 (dd, J = 11.2, 6.0 Hz, 1H), 7.35 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.43 (s, 1H), 4.66-4.40 (m, 3H), 4.31-4.18 (m, 1H), 4.18- 4.09 (m, 1H), 3.59 (d, J = 8.8 Hz, 4H), 0.53 (d, J = 7.0 Hz, 3H).47239608.01H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 7.97- 7.75 (m, 3H), 7.62 (d, J = 8.7 Hz, 2H), 7.56-7.42 (m, 3H), 7.35-7.31 (m, 1H), 6.95 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.44 (t, J = 7.2 Hz, 1H), 4.57-4.39 (m, 3H), 4.24 (dd, J = 10.9, 6.7 Hz, 1H), 4.13 (t, J = 8.5 Hz, 1H), 3.60 (t, J = 8.5 Hz, 1H), 0.53 (d, J = 7.0 Hz, 3H).47339638.01H NMR (400 MHz, DMSO) δ 8.47 (s, 1H), 7.98- 7.70 (m, 3H), 7.61 (t, J = 8.3 Hz, 2H), 7.57-7.46 (m, 2H), 7.39 (dd, J = 11.5, 6.0 Hz, 1H), 7.33 (dd, J = 8.3, 2.1 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.33 (d, J = 8.2 Hz, 1H), 4.73- 4.38 (m, 4H), 4.30-4.11 (m, 2H), 3.63 (d, J = 10.0 Hz, 1H), 2.97 (s, 3H), 1.43 (s, 3H).47439638.01H NMR (400 MHz, DMSO) δ 8.45 (s, 1H), 7.95- 7.76 (m, 3H), 7.67-7.56 (m, 2H), 7.56-7.47 (m, 2H), 7.43-7.30 (m, 2H), 6.95 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.31 (s, 1H), 4.64-4.35 (m, 3H), 4.29- 4.14 (m, 2H), 3.63 (d, J = 10.1 Hz, 1H), 2.96 (s, 3H), 1.43 (s, 3H).47535624.21H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 7.96- 7.77 (m, 3H), 7.65 (d, J = 8.4 Hz, 1H), 7.60-7.38 (m, 3H), 7.19 (t, J = 8.0 Hz, 2H), 5.63 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.67-4.29 (m, 4H), 3.85-3.69 (m, 2H), 1.34 (s, 3H), 0.62 (s, 3H).47635606.21H NMR (400 MHz, DMSO) δ 8.51 (s, 1H), 7.96- 7.77 (m, 3H), 7.65 (d, J = 8.4 Hz, 1H), 7.60-7.38 (m, 3H), 7.19 (t, J = 8.0 Hz, 2H), 5.63 (s, 2H), 5.04 (d, J = 6.6 Hz, 1H), 4.67-4.29 (m, 4H), 3.85-3.69 (m, 2H), 1.34 (s, 3H), 0.62 (s, 3H).47735606.21H NMR (400 MHz, DMSO) δ 8.50 (s, 1H), 8.03- 7.79 (m, 3H), 7.75-7.51 (m, 3H), 7.46 (dq, J = 13.9, 7.0, 6.5 Hz, 2H), 7.26 (q, J = 9.3, 7.7 Hz, 2H), 5.62 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.66-4.28 (m, 4H), 3.83-3.66 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).47835588.31H NMR (400 MHz, DMSO) δ 8.52 (s, 1H), 7.98- 7.77 (m, 3H), 7.78-7.36 (m, 5H), 7.36-7.13 (m, 2H), 6.96 (d, J = 8.3 Hz, 1H), 5.53 (s, 2H), 5.05 (d, J = 6.6 Hz, 1H), 4.72-4.37 (m, 4H), 3.90-3.66 (m, 2H), 1.34 (s, 3H), 0.62 (s, 3H).47935667.21H NMR (400 MHz, Acetonitrile-d3) δ 8.42 (s, 1H), 7.73 (t, J = 7.7 Hz, 1H), 7.68-7.51 (m, 6H), 5.68 (s, 2H), 4.93 (d, J = 6.6 Hz, 1H), 4.59 (dd, J = 11.3, 1.5 Hz, 1H), 4.53-4.40 (m, 2H), 4.32 (d, J = 17.2 Hz, 1H), 3.85 (d, J = 8.7 Hz, 1H), 3.78 (d, J = 8.7 Hz, 1H), 1.41 (s, 3H), 0.72 (s, 3H).48035599.31H NMR (400 MHz, Acetonitrile-d3) δ 8.57 (s, 1H), 7.86 (dd, J = 8.4, 1.5 Hz, 1H), 7.78-7.68 (m, 3H), 7.65 (d, J = 8.1 Hz, 2H), 7.62-7.54 (m, 2H), 7.52 (ddd, J = 8.3, 3.1, 1.4 Hz, 1H), 7.20 (dd, J = 11.6, 6.1 Hz, 1H), 5.63 (s, 2H), 5.24 (t, J = 7.2 Hz, 1H), 4.55 (dd, J = 11.0, 1.4 Hz, 1H), 4.37 (s, 2H), 4.23 (dd, J = 11.0, 6.7 Hz, 1H), 4.13 (t,J = 8.5 Hz, 1H), 3.67 (t, J =8.4 Hz, 1H), 2.88 (hept, J =7.6 Hz, 1H), 0.57 (d, J = 7.0Hz, 3H).48135635.21H NMR (400 MHz, Acetonitrile-d3) δ 8.56 (s, 1H), 7.87 (dd, J = 8.5, 1.5 Hz, 1H), 7.70 (dd, J = 10.7, 6.5 Hz, 1H), 7.65-7.48 (m, 5H), 7.20 (dd, J = 11.6, 6.1 Hz, 1H), 5.65 (s, 2H), 5.23 (t, J = 7.3 Hz, 1H), 4.56 (dd, J = 10.9, 1.4 Hz, 1H),4.37 (s, 2H), 4.23 (dd, J = 10.9, 6.7 Hz, 1H), 4.13 (t, J = 8.5 Hz, 1H), 3.68 (t, J = 8.3 Hz, 1H), 2.87 (hept, J = 7.7 Hz, 1H), 0.57 (d, J = 7.0 Hz, 3H).48235649.21H NMR (400 MHz, Acetonitrile-d3) δ 8.54 (s, 1H), 7.83 (dd, J = 8.5, 1.5 Hz, 1H), 7.72 (t, J = 7.7 Hz, 1H), 7.66-7.45 (m, 6H), 5.66 (s, 2H), 4.92 (d, J = 6.7 Hz, 1H), 4.60 (dd, J = 11.2, 1.5 Hz, 1H), 4.53-4.40 (m, 2H), 4.31 (d, J = 17.1Hz, 1H), 3.86 (d, J = 8.7 Hz, 1H), 3.77 (d, J = 8.7 Hz, 1H), 1.41 (s, 3H), 0.70 (s, 3H).48335667.21H NMR (400 MHz, Acetonitrile-d3) δ 8.41 (s, 1H), 7.72 (dd, J = 10.7, 6.4 Hz, 1H), 7.67-7.49 (m, 5H), 7.23 (dd, J = 11.5, 6.1 Hz, 1H), 5.66 (s, 2H), 4.85 (d, J = 6.7 Hz, 1H), 4.54 (dd, J = 11.3, 1.5 Hz, 1H),4.43 (dd, J = 11.3, 6.8 Hz, 1H), 4.39-4.27 (m, 2H), 3.84 (d, J = 8.8 Hz, 1H), 3.75 (d, J = 8.8 Hz, 1H), 1.35 (s, 3H), 0.67 (s, 3H).48435609.21H NMR (400 MHz, Acetonitrile-d3) δ 8.63 (d, J = 5.1 Hz, 1H), 8.57 (s, 1H), 7.94 (dd, J = 10.5, 6.3 Hz, 1H), 7.86 (dd, J = 8.5, 1.5 Hz, 1H), 7.62-7.52 (m, 3H), 7.31-7.19 (m, 3H), 5.53 (s, 2H), 5.24 (t, J = 7.2 Hz, 1H), 4.55 (dd, J = 11.0, 1.4 Hz, 1H), 4.40 (s, 2H), 4.23 (dd, J = 11.0, 6.7 Hz, 1H), 4.13 (t, J = 8.5 Hz, 1H), 3.67 (t, J = 8.4 Hz, 1H), 2.89 (hept, J = 7.5 Hz, 1H), 0.57 (d, J = 7.1 Hz, 3H).48535636.21H NMR (400 MHz, Acetonitrile-d3) δ 8.03 (d, J = 1.3 Hz, 1H), 7.69-7.64 (m, 1H), 7.64-7.50 (m, 4H), 7.31-7.22 (m, 2H), 5.59 (s, 2H), 4.53 (t, J = 5.0 Hz, 2H), 4.46 (s, 2H), 3.75 (t, J = 5.0 Hz, 2H), 3.27 (s, 3H).48635617.21H NMR (400 MHz, Acetonitrile-d3) δ 8.57 (d, J = 1.5 Hz, 1H), 7.86 (dd, J = 8.5, 1.6 Hz, 1H), 7.75-7.68 (m, 2H), 7.62-7.53 (m, 4H), 7.51 (ddd, J = 8.2, 3.1, 1.4 Hz, 1H), 7.20 (dd, J = 11.6, 6.1 Hz, 1H), 5.66 (s, 2H), 5.24 (t, J = 7.2 Hz, 1H), 4.54 (dd, J = 11.0, 1.4 Hz, 1H), 4.37 (s, 2H), 4.23 (dd, J = 11.0, 6.7 Hz, 1H), 4.13 (t, J = 8.5 Hz, 1H), 3.67 (t, J =8.4 Hz, 1H), 2.87 (dq, J =15.2, 7.6 Hz, 1H), 0.57 (d,J = 7.1 Hz, 3H).48735631.21H NMR (400 MHz, Acetonitrile-d3) δ 8.55 (s, 1H), 7.86 (dd, J = 8.5, 1.5 Hz, 1H), 7.83-7.78 (m, 1H), 7.75 (dd, J = 10.7, 6.4 Hz, 1H), 7.62-7.57 (m, 1H), 7.53 (ddt, J = 10.5, 9.2, 4.8 Hz, 3H), 7.22 (dd, J = 11.5, 6.1 Hz, 1H), 6.92 (dd, J = 8.3, 0.7 Hz, 1H), 5.58 (s, 2H), 4.84 (d, J = 6.6 Hz, 1H), 4.56 (dd, J = 11.2, 1.6 Hz, 1H), 4.42 (dd, J = 11.2,6.9 Hz, 1H), 4.36 (d, J = 6.3Hz, 2H), 3.85 (d, J = 8.7 Hz,1H), 3.75 (d, J = 8.7 Hz,1H), 1.34 (s, 3H), 0.65 (s,3H).48835640.21H NMR (400 MHz, Acetonitrile-d3) δ 8.69 (s, 1H), 8.01 (dd, J = 8.6, 1.5 Hz, 1H), 7.85 (dd, J = 10.8, 6.4 Hz, 1H), 7.77 (dd, J = 8.2, 7.5 Hz, 1H), 7.72 (d, J = 8.5 Hz, 1H), 7.50 (dd, J = 7.5, 1.7 Hz, 1H), 7.36-7.22 (m, 3H), 6.86 (d, J = 8.2 Hz, 1H), 5.53 (d, J = 1.3 Hz, 2H), 4.93 (d, J = 6.6 Hz, 1H), 4.62-4.50 (m, 3H), 4.43 (dd, J = 11.6, 6.8 Hz,1H), 3.89 (d, J = 8.9 Hz,1H), 3.76 (d, J = 8.9 Hz,1H), 1.34 (s, 3H), 0.68 (s,3H).48935640.21H NMR (400 MHz, Acetonitrile-d3) δ 8.62 (s, 1H), 7.98-7.89 (m, 2H), 7.75 (t, J = 7.9 Hz, 1H), 7.65 (d, J = 8.5 Hz, 1H), 7.50 (dd, J = 7.5, 1.7 Hz, 1H), 7.26 (dd, J = 11.5, 6.1 Hz, 1H), 7.14 (d, J = 7.4 Hz, 2H), 6.78 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 4.89 (d, J = 6.7 Hz, 1H), 4.55 (dd, J = 11.4, 1.5 Hz, 1H), 4.49-4.37 (m, 3H), 3.87 (d, J = 8.8 Hz,1H), 3.75 (d, J = 8.8 Hz,1H), 1.34 (s, 3H), 0.67 (s,3H).490640.21H NMR (400 MHz, Acetonitrile-d3) δ 8.56 (s, 1H), 7.88 (dd, J = 8.5, 1.5 Hz, 1H), 7.85-7.73 (m, 2H), 7.60 (d, J = 8.5 Hz, 1H), 7.54-7.48 (m, 1H), 7.44 (dd, J = 9.5, 6.3 Hz, 1H), 7.35 (dd, J = 9.2, 6.1 Hz, 1H), 7.22 (dd, J = 11.5, 6.1 Hz, 1H), 6.87 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 4.85 (d, J = 6.7 Hz, 1H), 4.55 (dd, J = 11.2, 1.5 Hz, 1H), 4.42(dd, J = 11.1, 6.9 Hz, 1H),4.38 (d, J = 6.0 Hz, 2H),3.86 (d, J = 8.7 Hz, 1H),3.75 (d, J = 8.7 Hz, 1H),1.34 (s, 3H), 0.65 (s, 3H).49135649.21H NMR (400 MHz, Acetonitrile-d3) δ 8.59 (s, 1H), 7.90 (dd, J = 8.5, 1.5 Hz, 1H), 7.74 (dd, J = 10.7, 6.5 Hz, 1H), 7.66-7.49 (m, 5H), 7.25 (dd, J = 11.6, 6.1 Hz, 1H), 5.68 (s, 2H), 4.87 (d, J = 6.8 Hz, 1H), 4.59 (dd, J = 11.2, 1.5 Hz, 1H), 4.46 (dd, J = 11.1, 6.8 Hz, 1H), 4.39 (d, J = 6.7 Hz, 2H), 3.89 (d, J = 8.7 Hz, 1H), 3.78 (d, J = 8.7 Hz, 1H), 1.37 (s, 3H), 0.68 (s, 3H).49235631.31H NMR (400 MHz, Acetonitrile-d3) δ 8.70 (s, 1H), 8.02 (dd, J = 8.6, 1.5 Hz, 1H),7.83 (d, J = 7.5 Hz, 1H), 7.81-7.75 (m, 1H), 7.72 (d, J = 8.5 Hz, 1H), 7.57-7.51 (m, 2H), 7.51- 7.44 (m, 1H), 7.29 (dd, J = 11.4, 6.1 Hz, 1H), 6.92 (d, J = 8.3 Hz, 1H), 5.62 (s, 2H), 4.93 (d, J = 6.6 Hz, 1H), 4.62-4.54 (m, 1H), 4.51 (d, J = 8.4 Hz, 2H), 4.44 (dd, J =11.5, 6.7 Hz, 1H), 3.90 (d,J = 8.9 Hz, 1H), 3.77 (d, J =8.9 Hz, 1H), 1.35 (s, 3H),0.69 (s, 3H).49335631.31H NMR (400 MHz, Acetonitrile-d3) δ 8.60 (s, 1H), 7.95-7.88 (m, 2H), 7.81 (t, J = 7.8 Hz, 1H), 7.64 (d, J = 8.5 Hz, 1H), 7.58 (dd, J = 7.3, 1.6 Hz, 1H), 7.48 (d, J = 6.7 Hz, 2H), 7.26 (dd, J = 11.6, 6.0 Hz, 1H), 6.84 (d, J = 8.2 Hz, 1H), 5.63 (s, 2H), 4.88 (d, J = 6.9 Hz, 1H), 4.60 (dd, J = 11.3, 1.4 Hz, 1H), 4.46 (dd, J = 11.3, 6.8 Hz, 1H), 4.41 (d, J = 5.9Hz, 2H), 3.89 (d, J = 8.7 Hz,1H), 3.78 (d, J = 8.6 Hz,1H), 1.38 (s, 3H), 0.69 (s,3H).49435599.31H NMR (400 MHz, Acetonitrile-d3) δ 8.60 (d, J = 1.5 Hz, 1H), 7.89 (dd, J = 8.5, 1.6 Hz, 1H), 7.84-7.75 (m, 2H), 7.72 (t, J = 7.5 Hz, 1H), 7.62 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.7 Hz, 2H), 7.54 (dd, J = 7.5, 1.8 Hz, 1H), 7.23 (dd, J = 11.5, 6.1 Hz, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.62 (s, 2H), 5.27 (t, J = 7.2 Hz, 1H), 4.57 (dd, J = 10.9, 1.4 Hz, 1H),4.41(s, 2H), 4.26 (dd, J = 11.0,6.7 Hz, 1H), 4.16 (t, J = 8.5Hz, 1H), 3.70 (t, J = 8.4 Hz,1H), 2.91 (hept, J = 7.1 Hz,1H), 0.59 (d, J = 7.1 Hz,3H).49535658.21H NMR (400 MHz, Acetonitrile-d3) δ 8.41 (s, 1H), 7.81 (dd, J = 10.7, 6.5 Hz, 1H), 7.63-7.50 (m, 4H), 7.30-7.19 (m, 3H), 5.59 (d, J = 1.1 Hz, 2H), 4.85 (d, J = 6.6 Hz, 1H), 4.54 (dd, J = 11.3, 1.6 Hz, 1H), 4.43 (dd, J = 11.3, 6.8 Hz, 1H), 4.37 (d, J = 7.1 Hz, 2H), 3.84 (d, J = 8.8 Hz, 1H), 3.75 (d, J = 8.7 Hz, 1H), 1.35 (s, 3H), 0.67 (s, 3H).49635631.31H NMR (400 MHz, Acetonitrile-d3) δ 8.58 (s, 1H), 7.89 (dd, J = 8.5, 1.5 Hz, 1H), 7.79-7.71 (m, 2H), 7.65-7.52 (m, 5H), 7.24 (dd, J = 11.6, 6.1 Hz, 1H), 5.70 (s, 2H), 4.86 (d, J = 6.6 Hz, 1H), 4.59 (dd, J = 11.2, 1.6 Hz, 1H), 4.45 (dd, J = 11.3, 7.0 Hz, 1H),4.38 (d, J = 6.4 Hz, 2H), 3.88 (d, J = 8.7 Hz, 1H), 3.78 (d, J = 8.7 Hz, 1H), 1.37 (s, 3H), 0.68 (s, 3H).49735640.21H NMR (400 MHz, Acetonitrile-d3) δ 8.56 (s, 1H), 7.87 (dd, J = 8.5, 1.5 Hz, 1H), 7.80 (dd, J = 10.8, 6.5 Hz, 1H), 7.64-7.47 (m, 4H), 7.32-7.16 (m, 3H), 5.59 (s, 2H), 4.85 (d, J = 6.8 Hz, 1H), 4.56 (dd, J = 11.1, 1.6 Hz, 1H), 4.48-4.42 (m, 1H), 4.42-4.30 (m, 2H), 3.86 (d, J = 8.7 Hz, 1H), 3.75 (d, J = 8.7 Hz, 1H), 1.34 (s, 3H), 0.65 (s, 3H).49835649.21H NMR (400 MHz, Acetonitrile-d3) δ 8.46 (s, 1H), 7.90 (dd, J = 10.8, 6.4 Hz, 1H), 7.78 (t, J = 7.9 Hz, 1H), 7.63 (dd, J = 11.2, 1.2 Hz, 1H), 7.55 (dd, J = 7.5, 1.8 Hz, 1H), 7.45 (d, J = 6.7 Hz, 2H), 7.26 (dd, J = 11.4, 6.1 Hz, 1H), 6.81 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 4.89 (d, J = 6.7 Hz, 1H), 4.55 (dd, J = 11.4, 1.5 Hz, 1H), 4.50- 4.37 (m, 3H), 3.86 (d, J =8.8 Hz, 1H), 3.76 (d, J = 8.8Hz, 1H), 1.36 (s, 3H), 0.69(s, 3H).49935658.11H NMR (400 MHz, DMSO-d6) δ 13.08 (s, 1H), 8.35 (s, 1H), 7.92 (dd, J = 10.4, 6.4 Hz, 1H), 7.87 (t, J = 7.9 Hz, 1H), 7.58-7.40 (m, 5H), 6.91 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.59-4.47 (m, 2H), 4.47-4.36 (m, 2H), 3.74 (q, J = 8.7 Hz, 2H), 1.34 (s, 3H), 0.61 (s, 3H).50035649.21H NMR (400 MHz, Acetonitrile-d3) δ 8.48 (s, 1H), 7.83-7.72 (m, 2H), 7.65 (dd, J = 11.1, 1.2 Hz, 1H), 7.56-7.44 (m, 3H), 7.25 (dd, J =11.5, 6.1Hz, 1H), 6.90 (d, J = 8.2 Hz, 1H), 5.61 (d, J = 1.3 Hz, 2H), 4.91 (d, J = 6.6 Hz, 1H), 4.54 (dd, J = 11.5, 1.4 Hz, 1H), 4.48-4.37 (m, 3H), 3.86 (d, J = 8.8 Hz, 1H), 3.76 (d, J = 8.9 Hz, 1H), 1.35 (s, 3H), 0.69 (s, 3H).50135658.11H NMR (400 MHz, Acetonitrile-d3) δ 8.49 (s, 1H), 7.82 (dd, J = 10.8, 6.4 Hz, 1H), 7.76 (dd, J = 8.3, 7.4 Hz, 1H), 7.67 (dd, J = 11.0, 1.2 Hz, 1H), 7.50- 7.39 (m, 2H), 7.33 (dd, J = 9.2, 6.1 Hz, 1H), 7.25 (dd, J = 11.5, 6.1Hz, 1H), 6.87 (dd, J = 8.3, 0.6 Hz, 1H), 5.47 (s, 2H), 4.93 (d, J = 6.6 Hz, 1H), 4.57-4.46 (m, 3H), 4.47-4.37 (m, 1H), 3.85 (d, J = 8.9 Hz, 1H), 3.76 (d, J = 8.9 Hz, 1H), 1.34 (s, 3H), 0.69 (s, 3H).502 Achiral separation Peak 2656.11H NMR (400 MHz, Acetonitrile-d3) δ 8.49 (s, 1H), 7.89-7.81 (m, 1H), 7.79 (d, J = 7.9 Hz, 1H), 7.70-7.62 (m, 2H), 7.54 (dd, J = 7.4, 1.7 Hz, 1H), 7.40-7.33 (m, 2H), 7.25 (dd, J = 11.4, 6.1 Hz, 1H), 6.89 (d, J = 8.2 Hz, 1H), 6.78 (t, J = 55.9 Hz, 1H), 5.59 (s, 2H), 4.90 (d, J = 6.6 Hz, 1H), 4.55 (dd, J = 11.5, 1.4 Hz, 1H), 4.48-4.38 (m, 3H), 3.86 (d, J = 8.8 Hz,1H), 3.76 (d, J = 8.8 Hz,1H), 1.35 (s, 3H), 0.69 (s,3H).502 Bchiral separation Peak 1656.21H NMR (400 MHz, Acetonitrile-d3) δ 8.44 (s, 1H), 7.91-7.77 (m, 2H), 7.70 (t, J = 7.7 Hz, 1H), 7.60 (dd, J = 11.3, 1.2 Hz, 1H), 7.57 (dd, J = 7.5, 1.8 Hz, 1H), 7.44-7.34 (m, 2H), 7.26 (dd, J =11.5, 6.1Hz, 1H), 6.98-6.88 (m, 1H), 6.74 (d, J = 55.9 Hz, 1H), 5.62 (s, 2H), 4.88 (d, J = 6.5 Hz, 1H), 4.57 (dd, J = 11.2, 1.5 Hz, 1H), 4.50-4.32 (m,3H), 3.87 (d, J = 8.7 Hz,1H), 3.77 (d, J = 8.8 Hz,1H), 1.38 (s, 3H), 0.69 (s,3H).50335692.11H NMR (400 MHz, Acetonitrile-d3) δ 8.41 (s, 1H), 7.80-7.72 (m, 2H), 7.63-7.48 (m, 5H), 7.23 (dd, J = 11.5, 6.1 Hz, 1H), 5.69 (s, 2H), 4.85 (d, J = 6.7 Hz, 1H), 4.55 (d, J = 11.2 Hz, 1H), 4.45 (d, J = 8.1 Hz, 1H), 4.42-4.29 (m, 2H), 3.84 (d, J = 8.8 Hz, 1H), 3.75 (d, J = 8.9 Hz, 1H), 1.35 (s, 3H), 0.66 (s, 3H).50435657.11H NMR (400 MHz, Acetonitrile-d3) δ 8.65 (d, J = 5.2 Hz, 1H), 8.45 (s, 1H), 7.95 (dd, J = 10.4, 6.2 Hz, 1H), 7.69 (t, J = 7.6 Hz, 1H), 7.62 (dd, J = 11.1, 1.2 Hz, 1H), 7.58 (dd, J = 5.2, 1.8 Hz, 1H), 7.42-7.35 (m, 2H), 7.30 (dd, J = 11.5, 5.9 Hz, 1H), 6.79 (t, J = 55.9 Hz, 1H), 5.61 (s, 2H), 4.88 (d, J = 6.7 Hz, 1H), 4.55 (d, J = 11.4 Hz, 1H), 4.50- 4.36 (m, 3H), 3.85 (d, J =8.8 Hz, 1H), 3.76 (d, J = 8.8Hz, 1H), 1.35 (s, 3H), 0.68(s, 3H).50535599.21H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.94-7.87 (m, 2H), 7.81- 7.71 (m, 3H),7.61 (d, J = 8.4 Hz, 1H), 7.54 (d, J = 7.2 Hz, 1H), 7.44 (dd, J = 11.4, 6.3 Hz, 2H), 6.99 (d, J = 8.2 Hz, 1H), 5.60 (s, 2H), 5.42 (t, J = 7.1 Hz, 1H), 4.56- 4.39 (m, 3H), 4.26-4.17 (m, 1H), 4.12 (t, J = 8.5 Hz, 1H), 3.59 (t, J = 8.5 Hz, 1H), 2.92-2.82 (m, 1H), 0.52 (d, J = 7.0 Hz, 3H).50635656.11H NMR (400 MHz, Acetonitrile-d3) δ 8.41 (s, 1H), 7.88-7.77 (m, 2H), 7.67 (t, J = 7.7 Hz, 1H), 7.62 (d, J = 11.0 Hz, 1H), 7.58- 7.51 (m, 1H), 7.41-7.32 (m, 2H), 7.23 (dd, J = 11.5, 6.1 Hz, 1H), 6.94-6.61 (m, 3H), 5.60 (s, 2H),4.87 (d, J = 6.7 Hz, 1H), 4.57 (d, J = 11.4 Hz, 1H), 4.52-4.45 (m, 1H), 4.45-4.32 (m, 2H), 3.85 (d, J = 8.8 Hz, 1H), 3.83-3.74 (m, 1H), 1.35 (s, 3H), 0.67 (s, 3H).50735624.11H NMR (400 MHz, Acetonitrile-d3) δ 8.58 (dd, J = 1.6, 0.6 Hz, 1H), 7.87- 7.77 (m, 2H), 7.77-7.72 (m, 1H), 7.59-7.47 (m, 3H), 7.28-7.20 (m, 2H), 7.16 (dd, J = 11.5, 6.1Hz, 1H), 6.84 (dd, J = 8.3, 0.7 Hz, 1H), 5.51 (d, J = 1.0 Hz, 2H), 4.55 (q, J = 7.1 Hz, 1H), 4.38 (d, J = 2.4 Hz, 2H), 3.18 (s, 2H), 1.64 (d, J = 7.1 Hz, 3H), 1.34 (s, 3H), 1.02 (s, 3H).50835621.21H NMR (400 MHz, Acetonitrile-d3) δ 8.55 (s, 1H), 7.87 (dd, J = 8.5, 1.5 Hz, 1H), 7.60 (d, J = 8.5 Hz, 1H), 7.53 (t, J = 8.2 Hz, 1H), 7.39 (t, J = 7.9 Hz, 1H), 7.32 (dd, J = 10.2, 6.4 Hz, 1H), 7.31-7.13 (m, 5H), 7.04 (ddd, J = 8.4, 2.6, 1.0 Hz, 1H), 5.16 (s, 2H), 4.84 (dd, J = 6.9, 1.6 Hz, 1H), 4.55 (dd, J = 11.2, 1.6 Hz, 1H), 4.43 (dd, J = 11.2, 6.9 Hz, 1H),4.35 (d, J = 6.5 Hz, 2H),3.85 (d, J = 8.7 Hz, 1H),3.75 (d, J = 8.7 Hz, 1H),1.34 (s, 3H), 0.65 (s, 3H).50935639.21H NMR (400 MHz, Acetonitrile-d3) δ 8.55 (s, 1H), 7.87 (dd, J = 8.5, 1.5 Hz, 1H), 7.60 (d, J = 8.5 Hz, 1H), 7.53 (t, J = 8.2 Hz, 1H), 7.39-7.10 (m, 7H), 5.19 (s, 2H), 4.84 (dd, J = 6.9, 1.6 Hz, 1H), 4.55 (dd, J = 11.2, 1.6 Hz, 1H), 4.42 (dd, J = 11.1, 6.8 Hz, 1H), 4.35 (d, J = 6.6 Hz, 2H), 3.85 (d, J = 8.7 Hz, 1H), 3.74 (d, J = 8.7 Hz, 1H), 1.34 (s, 3H), 0.64 (s, 3H).51035610.21H NMR (400 MHz, Acetonitrile-d3) δ 8.24 (s, 1H), 7.91-7.73 (m, 3H), 7.61-7.48 (m, 3H), 7.29- 7.13 (m, 3H),6.84 (d, J = 8.2 Hz, 1H), 5.52 (s, 2H), 4.44 (s, 2H), 4.30 (s, 2H), 3.14 (s, 3H), 1.22 (s, 6H).5116555.31H NMR (400 MHz, Methanol-d4) δ 8.25 (s, 1H), 7.97 (dd, J = 8.5, 1.5 Hz, 1H), 7.78 (t, J = 7.9 Hz, 1H), 7.75-7.60 (m, 6H), 7.51 (d, J = 7.4 Hz, 1H), 7.12 (dd, J = 11.6, 6.0 Hz, 1H), 6.90 (d, J = 8.3 Hz, 1H), 5.57 (s, 2H), 4.55 (t, J = 5.1 Hz, 2H), 4.47 (s, 2H), 3.72 (t, J = 5.0 Hz, 2H), 3.25 (s, 3H).51235(M+) 624.611H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 8.15 (s, 1H), 7.96-7.83 (m, 3H), 7.82 (dd, J = 8.5, 1.5 Hz, 1H), 7.71 (dd, J = 8.6, 1.4 Hz, 1H), 7.63 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.5 Hz, 1H), 7.47 (dd, J = 11.1, 6.4 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.65 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.63-4.54 (m, 1H), 4.53 (s, 1H), 4.49-4.35 (m, 2H), 4.31 (s, 3H), 3.81-3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).51335(M+) 624.61H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 8.04 (d, J = 8.6 Hz, 1H), 8.01 (s, 1H), 7.90 (t, J = 7.9 Hz, 1H), 7.87-7.78 (m, 2H), 7.62 (d, J = 8.5 Hz, 1H), 7.54 (ddd, J = 7.6, 5.1, 1.4 Hz, 2H), 7.46 (dd, J = 11.4, 6.2 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.67 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.64-4.48 (m, 2H), 4.48- 4.32 (m, 2H), 4.30 (s, 3H), 3.82-3.72 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).51435(M+) 666.61H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J = 1.3 Hz, 1H), 7.91-7.78 (m, 2H), 7.54-7.42 (m, 3H), 7.39 (dd, J = 11.6, 6.1 Hz, 1H), 7.35 (d, J = 7.8 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.48 (s, 2H), 4.71- 4.53 (m, 6H), 4.46 (s, 2H), 3.68 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H), 2.96 (s, 3H).51540(M+) 676.61H NMR (400 MHz, Methanol-d4) δ 8.15 (d, J = 1.3 Hz, 1H), 7.75 (dd, J = 10.8, 6.4 Hz, 1H), 7.66 (d, J = 11.3 Hz, 1H), 7.59 (dd, J = 9.9, 8.2 Hz, 1H), 7.55-7.42 (m, 3H), 7.39-7.30 (m, 1H), 7.18 (dd, J = 11.5, 6.0 Hz, 1H), 5.58 (s, 2H), 5.18 (d, J = 7.5 Hz, 1H), 4.78- 4.65 (m, 6H), 4.65-4.31 (m, 4H), 3.78 (d, J = 1.7 Hz, 3H), 2.90-2.60 (m, 1H), 2.56-2.36 (m, 1H).51622(M+) 658.61H NMR (400 MHz, Methanol-d4) δ 8.27 (s, 1H), 8.02-7.92 (m, 1H), 7.75 (dd, J = 10.7, 6.2 Hz, 1H), 7.67-7.54 (m, 2H), 7.54- 7.43 (m, 3H), 7.33 (d, J = 8.1 Hz, 0H), 7.17 (dd, J = 11.5, 6.0 Hz, 1H), 5.59 (s, 2H), 5.20 (d, J = 10.9 Hz, 1H), 4.73 (s, 5H),4.68- 4.50 (m, 3H), 4.50-4.36 (m, 1H), 3.78 (d, J = 1.9 Hz, 3H), 2.78 (s, 1H), 2.51 (s, 1H).51740(M+) 678.61H NMR (400 MHz, Methanol-d4) δ 8.15 (s, 1H), 7.84-7.71 (m, 2H), 7.67 (d, J = 11.3 Hz, 1H), 7.54- 7.40 (m, 3H), 7.33 (d, J = 7.9 Hz, 1H), 7.18 (dd, J = 11.5, 6.1 Hz, 1H), 6.87 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 5.17 (d, J = 8.1 Hz, 1H), 4.70 (d, J = 4.3 Hz, 5H), 4.66-4.48 (m, 4H), 4.45 (dt, J = 9.0, 5.9 Hz, 1H), 2.91 (s, 3H), 2.79 (s, 1H), 2.55-2.41 (m, 1H).51840605.51H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J = 1.3 Hz, 1H), 7.97-7.85 (m, 2H), 7.75 (ddd, J = 9.7, 7.8, 6.5 Hz, 3H), 7.58-7.44 (m, 2H), 7.39 (dd, J = 11.5, 6.1 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 5.60 (s, 2H), 4.53 (dd, J = 15.2, 3.2 Hz, 1H), 4.46 (s, 2H), 4.36 (dd, J = 15.2, 8.8 Hz, 1H), 3.76-3.61 (m, 1H), 3.08 (s, 3H), 1.23 (d, J = 6.2 Hz, 3H).51935(M+) 661.21H NMR (400 MHz, Methanol-d4) δ 8.30 (s, 1H), 7.99 (dd, J = 8.5, 1.5 Hz, 1H), 7.86-7.73 (m, 2H), 7.66 (d, J = 8.5 Hz, 1H), 7.56-7.49 (m, 1H), 7.46 (d, J = 10.9 Hz, 2H), 7.33 (d, J = 7.8 Hz, 1H), 7.18 (dd, J = 11.6, 6.0 Hz, 1H), 6.87 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 5.21 (d, J = 8.8 Hz, 1H), 4.79-4.65 (m, 5H), 4.65- 4.35 (m, 4H), 2.91 (s, 3H), 2.87-2.69 (m, 1H), 2.51 (d, J = 11.2 Hz, 0H).52035(M+) 651.21H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J = 1.4 Hz, 1H), 7.99 (dd, J = 8.5, 1.5 Hz, 1H), 7.85-7.72 (m, 2H), 7.67 (d, J = 8.5 Hz, 1H), 7.56-7.43 (m, 3H), 7.36 (dd, J = 7.9, 3.8 Hz, 1H), 7.24-7.09 (m, 1H), 6.87 (d, J = 8.2 Hz, 1H), 5.52 (d, J = 2.5 Hz, 2H), 5.19 (td, J = 7.2, 2.5 Hz, 1H), 5.08 (d, J = 4.3 Hz, 2H), 4.77 (d, J = 6.9 Hz,2H), 4.73-4.37 (m, 6H),2.88-2.67 (m, 1H), 2.50(dq, J = 11.4, 7.7 Hz, 1H),1.97-1.84 (m, 1H), 0.97(dq, J = 6.0, 3.1 Hz, 2H),0.91 (ddt, J = 8.8, 6.0, 2.8Hz, 2H).52135(M+) 641.21H NMR (400 MHz, Methanol-d4) δ 8.31 (s, 1H), 7.99 (dd, J = 8.5, 1.5 Hz, 1H), 7.86-7.70 (m, 2H), 7.67 (d, J = 8.5 Hz, 1H), 7.53-7.41 (m, 3H), 7.36- 7.25 (m, 1H), 7.17 (dd, J = 11.4, 6.0 Hz, 1H), 6.87 (d, J = 8.3 Hz, 1H), 5.50 (s, 2H), 5.20 (d, J = 7.2 Hz, 1H), 4.72 (q, J = 3.8, 3.2 Hz, 6H), 4.66-4.34 (m, 4H), 3.78 (d, J = 1.9 Hz, 3H), 2.91-2.74 (m, 1H), 2.50 (dt, J = 17.5, 7.8 Hz, 1H).52235(M+) 625.21H NMR (400 MHz, Methanol-d4) δ 8.31 (s, 1H), 7.99 (dd, J = 8.5, 1.5 Hz, 1H), 7.79 (q, J = 8.7, 7.8 Hz, 2H), 7.67 (d, J = 8.5 Hz, 1H), 7.58-7.42 (m, 3H), 7.42-7.27 (m, 1H), 7.26- 7.13 (m, 1H), 6.87 (d, J = 8.3 Hz, 1H), 5.52 (s, 2H), 5.30-5.12 (m, 1H), 4.73 (dd, J = 16.6, 7.0 Hz, 3H), 4.66-4.37 (m, 5H), 2.91- 2.65 (m, 1H), 2.60-2.39 (m, 1H), 2.18 (d, J = 5.9 Hz, 3H).52327(M+) 597.21H NMR (400 MHz, Methanol-d4) δ 8.32 (s, 1H), 8.10 (s, 1H), 8.00 (dd, J = 8.5, 1.5 Hz, 1H), 7.89-7.71 (m, 4H), 7.66 (s, 1H), 7.53 (d, J = 7.4 Hz, 1H), 7.18 (dd, J = 11.4, 6.0 Hz, 1H), 6.92 (d, J = 8.3 Hz, 1H), 5.68 (s, 2H), 5.20 (q, J = 6.8 Hz, 1H), 4.74 (dd, J = 15.7, 6.9 Hz, 1H), 4.68-4.47 (m, 4H), 4.45 (dt, J = 9.3, 5.9 Hz, 1H), 4.35 (d, J = 1.0 Hz, 3H), 2.80 (dt, J = 15.9, 7.7 Hz, 1H), 2.58-2.38 (m, 1H).52427(M+) 596.21H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J = 1.5 Hz, 1H), 8.02-7.93 (m, 2H), 7.87-7.72 (m, 4H), 7.71-7.61 (m, 2H), 7.52 (dd, J = 7.5, 1.6 Hz, 1H), 7.32 (dd, J = 8.4, 1.2 Hz, 1H), 7.17 (dd, J = 11.5, 6.0 Hz, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.64 (s, 2H), 5.19 (qd, J = 7.1, 2.5 Hz, 1H), 4.72 (dd, J = 15.7, 6.9 Hz, 1H), 4.67- 4.48 (m, 5H), 4.48-4.34 (m, 1H), 4.06 (s, 3H), 2.91- 2.65 (m, 1H), 2.58-2.40 (m, 1H).525(M+) 597.21H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J = 1.4 Hz, 1H), 8.00 (d, J = 2.1 Hz, 1H), 7.98 (d, J = 2.2 Hz, 1H), 7.92 (s, 1H), 7.86- 7.73 (m, 2H), 7.66 (d, J = 8.5 Hz, 1H), 7.60 (dd, J = 8.7, 1.3 Hz, 1H), 7.53 (dd, J = 7.5, 1.5 Hz, 1H), 7.18 (dd, J = 11.4, 6.0 Hz, 1H),6.93 (d, J = 8.2 Hz, 1H), 5.70 (s, 2H), 5.19 (dd, J = 7.2, 2.5 Hz, 1H), 4.72 (dd, J = 15.7, 6.9 Hz, 1H), 4.67-4.49 (m,4H), 4.48-4.36 (m, 1H),4.33 (s, 3H), 2.79 (ddt, J =14.2, 11.6, 6.8 Hz, 1H),2.56-2.41 (m, 1H).52627(M+) 596.21H NMR (400 MHz, Methanol-d4) δ 8.31 (d, J = 1.4 Hz, 1H), 7.99 (d, J = 9.1 Hz, 2H), 7.91-7.81 (m, 2H), 7.76 (q, J = 6.7, 5.5 Hz, 1H), 7.67 (d, J = 8.5 Hz, 1H), 7.63-7.53 (m, 2H), 7.51 (dd, J = 7.5, 1.6 Hz, 1H), 7.18 (dd, J = 11.4, 6.0 Hz, 1H), 6.86 (d, J = 8.2 Hz, 1H), 5.59 (s, 2H), 5.20 (qd, J = 7.2, 2.5 Hz, 1H), 4.81- 4.67 (m, 1H), 4.67-4.49(m, 4H), 4.49-4.35 (m,1H), 4.07 (d, J = 0.9 Hz,3H), 2.89-2.71 (m, 1H),2.64-2.38 (m, 1H).52727(M+) 600.21H NMR (400 MHz, Methanol-d4) δ 8.29 (s, 1H), 8.11 (s, 1H), 8.04 (d, J = 9.0 Hz, 1H), 7.98 (dd, J = 8.5, 1.5 Hz, 1H), 7.86-7.72 (m, 3H), 7.65 (d, J = 8.5 Hz, 1H), 7.55 (d, J = 7.5 Hz, 1H), 7.16 (dd, J = 11.5, 6.1 Hz, 1H), 6.97 (d, J = 8.2 Hz, 1H), 5.71 (s, 2H), 5.19 (d, J = 7.3 Hz, 1H), 4.72 (dd, J = 15.7, 6.9 Hz, 1H), 4.67- 4.34 (m, 5H), 2.79 (t, J = 9.1 Hz, 1H), 2.59-2.41 (m, 1H).52835(M+) 706.01H NMR (400 MHz, DMSO) δ 8.89 (s, 1H), 8.82 (s, 1H), 8.47 (s, 1H), 8.30 (s, 1H), 8.03 (s, 1H), 7.98- 7.85 (m, 2H), 7.81 (s, 1H), 7.57 (d, J = 7.3 Hz, 1H), 7.46 (dd, J = 11.4, 6.1Hz, 1H), 7.01 (d, J = 8.2 Hz, 1H), 5.68 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.67-4.29 (m, 4H), 3.84-3.68 (m, 2H), 1.31 (s, 3H), 0.59 (s, 3H).52935669.61H NMR (400 MHz, DMSO) δ 8.49 (s, 1H), 7.91- 7.77 (m, 3H), 7.55-7.39 (m, 5H), 7.34 (dd, J = 10.6, 7.8 Hz, 1H), 6.94 (d, J = 8.3 Hz, 1H), 5.47 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.64 (t, J = 6.9 Hz, 4H), 4.58-4.50 (m, 2H), 4.44 (dd, J = 11.2, 6.8 Hz, 1H), 4.39 (d, J = 16.9 Hz, 1H), 3.81-3.71 (m, 2H), 3.67 (s, 3H), 1.34 (s, 3H), 0.61 (s, 3H).53027599.11H NMR (400 MHz, Methanol-d4) δ 9.21 (s, 1H), 8.52 (d, J = 1.4 Hz, 1H), 8.20 (dd, J = 8.6, 1.4 Hz, 2H), 8.08 (d, J = 8.4 Hz, 1H), 7.87 (dd, J = 10.8, 6.3 Hz, 1H), 7.83-7.73 (m, 2H), 7.69 (dd, J = 8.5, 1.6 Hz, 1H), 7.55 (dd, J = 7.5, 1.6 Hz, 1H), 7.31 (dd, J = 11.1, 6.0 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.66 (s, 2H), 5.24 (tt, J = 7.3, 3.8 Hz, 1H),4.93 (dd, J = 15.5, 7.5 Hz,1H), 4.83-4.63 (m, 4H),4.54 (dt, J = 9.1, 6.0 Hz,1H), 2.98-2.75 (m, 1H),2.67-2.50 (m, 1H).53127629.21H NMR (400 MHz, Methanol-d4) δ 8.57 (d, J = 1.3 Hz, 1H), 8.21 (dd, J = 8.6, 1.4 Hz, 1H), 7.90 (dd, J = 10.8, 6.3 Hz, 1H), 7.85- 7.74 (m, 2H), 7.69 (d, J = 1.6 Hz, 1H), 7.54 (ddd, J = 8.3, 6.4, 1.7 Hz, 2H), 7.37 (dd, J = 11.2, 6.1 Hz, 1H), 7.26 (d, J = 8.3 Hz, 1H), 6.91 (d, J = 8.2 Hz, 1H), 5.52 (s, 2H), 5.26 (qd, J = 7.4, 2.4 Hz, 1H), 4.98 (dd,J = 15.5, 7.5 Hz, 1H), 4.86-4.64 (m, 4H), 4.54 (dt, J =9.1, 6.0 Hz, 1H), 3.47 (s,3H), 2.88 (dtd, J = 11.5, 8.2,6.1 Hz, 1H), 2.58 (ddt, J =11.5, 9.1, 7.2 Hz, 1H).532636.31H NMR (400 MHz, Chloroform-d) δ 8.55 (s, 1H), 8.01 (dd, J = 8.5, 1.5 Hz, 1H), 7.88 (dd, J = 10.8, 6.3 Hz, 1H), 7.79 (d, J = 8.5 Hz, 1H), 7.66 (t, J = 7.9 Hz, 1H), 7.54-7.41 (m, 3H), 7.14 (dt, J = 9.8, 2.3 Hz, 2H), 7.08 (dd, J = 11.3, 6.0 Hz, 1H), 6.79 (d, J = 8.2 Hz, 1H), 5.49 (s, 2H), 4.65 (d, J = 7.0 Hz, 1H), 4.57 (dd, J = 11.0, 1.8 Hz, 1H), 4.47-4.30 (m, 3H), 3.96-3.92(m, 4H), 3.79 (d, J = 8.8 Hz,1H), 1.34 (s, 3H), 0.66 (s,3H).53339624.11H NMR (400 MHz, DMSO) δ 8.43 (d, J = 1.5 Hz, 1H), 7.91-7.75 (m, 3H), 7.67-7.57 (m, 2H), 7.53 (ddd, J = 10.1, 7.3, 2.5 Hz, 2H), 7.43-7.28 (m, 2H), 5.60 (s, 2H), 4.54 (s, 2H), 4.42 (dd, J = 10.4, 3.6 Hz, 1H), 4.19-4.07 (m, 2H), 4.02 (dd, J = 10.5, 8.2 Hz, 1H), 3.80 (dd, J = 10.5, 4.7 Hz, 1H), 2.88 (s, 3H).53439642.21H NMR (400 MHz, DMSO) δ 1H NMR (400 MHz, DMSO) δ 8.45 (d, J = 1.6 Hz, 1H), 7.94-7.77 (m, 3H), 7.68-7.55 (m, 2H), 7.55-7.46 (m, 2H), 7.39- 7.27 (m, 2H), 6.96 (d, J = 8.2 Hz, 1H), 5.65-5.54 (m, 1H), 5.50 (s, 2H), 4.57 (s, 2H), 4.43 (dd, J = 10.4, 3.6 Hz, 1H), 4.13 (dt, J = 8.5, 2.1 Hz, 2H), 4.02 (dd, J = 10.6, 8.2 Hz, 1H), 3.79 (dd, J = 10.8, 4.9 Hz, 1H), 2.88 (s, 3H).53541668.21H NMR (400 MHz, Methanol-d4) δ 8.92 (s, 1H), 8.20 (dd, J = 8.6, 1.4 Hz, 1H), 8.10-7.92 (m, 2H), 7.91-7.73 (m, 3H), 7.58 (dd, J = 7.4, 1.6 Hz, 1H), 7.51 (t, J = 7.8 Hz, 1H), 7.46- 7.28 (m, 3H), 6.91 (d, J = 8.2 Hz, 1H), 5.54 (s, 2H), 5.15 (d, J = 6.4 Hz, 1H), 4.80-4.61 (m, 3H), 4.52 (dd, J = 11.7, 6.7 Hz, 1H), 4.00 (d, J = 8.9 Hz, 1H),3.93 (s, 3H), 3.84 (d, J = 8.9Hz, 1H), 1.41 (s, 3H), 0.76(s, 3H).53635656.01H NMR (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 7.87 (dd, J = 10.3, 8.2 Hz, 1H), 7.84-7.69 (m, 6H), 7.62 (d, J = 8.4 Hz, 1H), 7.55 (ddd, J = 8.2, 2.9, 1.4 Hz, 1H), 7.46 (dd, J = 11.5, 6.1Hz, 1H), 5.68 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.55-4.48 (m, 2H), 4.47- 4.32 (m, 2H), 3.82-3.69 (m, 2H), 1.33 (s, 3H), 0.60 (s, 3H).53735639.01H NMR (400 MHz, DMSO-d6) δ 8.79 (d, J = 5.2 Hz, 1H), 8.48 (s, 1H), 7.91 (dd, J = 10.2, 6.3 Hz, 1H), 7.83-7.75 (m, 3H), 7.73 (d, J = 8.2 Hz, 2H), 7.64 (dd, J = 5.2, 1.9 Hz, 1H), 7.61 (d, J = 8.5 Hz, 1H), 7.55 (dd, J = 11.5, 5.9 Hz, 1H), 5.63 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.61-4.49 (m, 2H), 4.49-4.36 (m, 2H), 3.82- 3.67 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).53835672.01H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 7.87 (dd, J = 10.3, 8.2 Hz, 1H), 7.80 (dd, J = 9.7, 8.3 Hz, 3H), 7.76-7.68 (m, 3H), 7.61 (d, J = 8.5 Hz, 1H), 7.55 (ddd, J = 8.2, 2.9, 1.5 Hz, 1H), 7.39 (dd, J = 11.3, 6.2 Hz, 1H), 5.68 (s, 2H), 5.52 (t, J = 7.3 Hz, 1H), 4.57-4.41 (m, 3H), 4.20 (dd, J = 10.9, 6.5 Hz, 1H), 4.08 (t, J = 8.7 Hz, 1H), 3.77 (t, J = 8.2 Hz, 1H), 3.19- 3.01 (m, 2H), 2.91 (s, 3H), 2.60 (t, J = 8.8 Hz, 1H).5393665.01H NMR (400 MHz, DMSO-d6) δ 8.43 (s, 1H), 8.03-7.86 (m, 2H), 7.85- 7.67 (m, 4H), 7.60 (d, J = 8.5 Hz, 1H), 7.53 (d, J = 7.3 Hz, 1H), 7.34 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.87-5.45 (m, 3H), 4.54 (s, 2H), 4.51- 4.41 (m, 2H), 4.14 (s, 0H), 4.05 (dd, J = 10.5, 8.3 Hz, 1H), 3.93-3.83 (m, 2H), 3.62-3.43 (m, 1H), 3.24- 2.99 (m, 1H).54035665.01H NMR (400 MHz, DMS0-d6) δ 8.43 (s, 1H), 8.03-7.86 (m, 2H), 7.85- 7.67 (m, 4H), 7.60 (d, J = 8.5 Hz, 1H), 7.53 (d, J = 7.3 Hz, 1H), 7.34 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.87-5.45 (m, 3H), 4.54 (s, 2H), 4.51- 4.41 (m, 2H), 4.14 (s, 0H), 4.05 (dd, J = 10.5, 8.3 Hz, 1H), 3.93-3.83 (m, 2H), 3.62- 3.43 (m, 1H), 3.24- 2.99 (m, 1H).54135665.01H NMR (400 MHz, DMSO-d6) δ 8.43 (s, 1H), 8.03-7.86 (m, 2H), 7.85- 7.67 (m, 4H), 7.60 (d, J = 8.5 Hz, 1H), 7.53 (d, J = 7.3 Hz, 1H), 7.34 (dd, J = 11.5, 6.1Hz, 1H), 7.00 (d, J = 8.3 Hz, 1H), 5.87-5.45 (m, 3H), 4.54 (s, 2H), 4.51- 4.41 (m, 2H), 4.14 (s, 0H), 4.05 (dd, J = 10.5, 8.3 Hz, 1H), 3.93-3.83 (m, 2H), 3.62-3.43 (m, 1H), 3.24- 2.99 (m, 1H).54235638.11H NMR (400 MHz, MeOD) δ 8.71 (s, 1H), 7.95 (dd, J = 8.5, 1.5 Hz, 1H), 7.82 (dd, J = 10.8, 6.4 Hz, 1H), 7.73 (t, J = 7.9 Hz, 1H), 7.62 (d, J = 8.5 Hz, 1H), 7.49 (t, J = 8.0 Hz, 2H), 7.28- 7.04 (m, 3H), 6.82 (d, J = 8.2 Hz, 1H), 5.46 (d, J = 14.0 Hz, 3H), 4.60-4.41 (m, 3H), 4.23 (dd, J = 10.9, 6.6 Hz, 1H), 4.15-4.06 (m, 1H), 3.89 (dd, J = 9.0, 7.0 Hz, 1H), 3.35 (s, 0H), 3.21- 3.04 (m, 2H), 2.92 (s, 3H), 2.57 (t, J = 8.9 Hz, 1H).54335656.11H NMR (400 MHz, MeOD) δ 8.72 (s, 1H), 7.94 (dd, J = 8.5, 1.5 Hz, 1H), 7.76 (dd, J = 10.7, 6.4 Hz, 1H), 7.70-7.44 (m, 4H), 7.26 (dd, J = 8.6, 2.6 Hz, 1H), 7.23-7.03 (m, 2H), 5.59 (s, 2H), 5.45 (t, J = 7.2 Hz, 1H), 4.65-4.46 (m, 3H), 4.24 (dd, J = 10.9, 6.6 Hz, 1H), 4.11 (t, J = 8.9 Hz, 1H), 3.89 (dd, J = 9.0, 7.0 Hz, 1H), 3.22-3.06 (m, 2H), 2.92 (s, 3H).54435620.21H NMR (400 MHz, MeOD) δ 8.72 (s, 1H), 7.95 (dd, J = 8.4, 1.5 Hz, 1H), 7.80 (dd, J = 10.8, 6.3 Hz, 1H), 7.74 (t, J = 7.8 Hz, 1H), 7.64 (d, J = 8.5 Hz, 1H), 7.49 (dd, J = 7.5, 1.7 Hz, 1H), 7.45 (d, J = 8.5 Hz, 2H), 7.36-7.33 (m, 2H), 7.14 (dd, J = 11.5, 6.0 Hz, 1H), 6.84 (d, J = 8.2 Hz, 1H), 5.44 (s, 3H), 4.61- 4.38 (m, 3H), 4.24 (dd, J = 10.9, 6.6 Hz, 1H), 4.19- 4.04 (m, 1H), 3.90 (dd, J = 9.0, 7.0 Hz, 1H), 2.93 (s, 3H).54535638.11H NMR (400 MHz, MeOD) δ 8.73 (s, 1H), 7.95 (dd, J = 8.5, 1.5 Hz, 1H), 7.81-7.75 (m, 1H), 7.64 (d, J = 8.5 Hz, 1H), 7.58 (dd, J = 9.8, 8.2 Hz, 1H), 7.54- 7.46 (m, 3H), 7.42-7.33 (m, 2H), 7.15 (dd, J = 11.5, 6.1 Hz, 1H), 5.59-5.36 (m, 3H), 4.63-4.41 (m, 3H), 4.26 (dd, J = 10.9, 6.6 Hz, 1H), 4.13 (t, J = 8.9 Hz, 1H), 3.91 (dd, J = 9.0, 7.0 Hz, 1H), 2.93 (s, 3H).54635652.21H NMR (400 MHz, MeOD) δ 8.73 (s, 1H), 7.95 (dd, J = 8.5, 1.5 Hz, 1H), 7.83 (dd, J = 10.7, 6.3 Hz, 1H), 7.74 (t, J = 7.9 Hz, 1H), 7.64 (d, J = 8.5 Hz, 1H), 7.55-7.47 (m, 2H), 7.24- 7.16 (m, 2H), 7.12 (dd, J = 11.5, 6.0 Hz, 1H), 6.83 (d, J = 8.2 Hz, 1H), 5.49 (s, 2H), 4.96 (d, J = 6.5 Hz, 1H), 4.61-4.46 (m, 2H), 4.46- 4.34 (m, 2H), 3.98 (d, J = 9.1 Hz, 1H), 3.73 (d, J = 9.0 Hz, 1H), 3.03 (d, J = 9.4 Hz, 1H), 2.88 (s, 3H), 2.69 (d, J = 9.4 Hz, 1H), 1.45 (s, 3H).54735652.21H NMR (400 MHz, MeOD) δ 8.74 (s, 1H), 7.96 (dd, J = 8.5, 1.5 Hz, 1H), 7.84 (dd, J = 10.8, 6.4 Hz, 1H), 7.77 (t, J = 7.9 Hz, 1H), 7.65 (d, J = 8.5 Hz, 1H), 7.53 (t, J = 8.0 Hz, 2H), 7.30- 7.17 (m, 2H), 7.13 (dd, J = 11.5, 6.0 Hz, 1H), 6.85 (d, J = 8.2 Hz, 1H), 5.51 (s, 2H), 4.97 (d, J = 6.5 Hz, 1H), 4.58 (d, J = 17.2 Hz, 2H), 4.51-4.34 (m, 2H), 3.99 (d, J = 9.0 Hz, 1H), 3.74 (d, J = 9.1 Hz, 1H), 3.05 (d, J = 9.4 Hz, 1H), 2.89 (s, 3H), 2.70 (d, J = 9.2 Hz, 1H), 1.46 (s, 3H).54935652.21H NMR (400 MHz, MeOD) δ 8.75 (s, 1H), 7.99 (dd, J = 8.5, 1.5 Hz, 1H), 7.86 (dd, J = 10.7, 6.4 Hz, 1H), 7.79 (t, J = 7.9 Hz, 1H), 7.68 (d, J = 8.5 Hz, 1H), 7.61-7.49 (m, 2H), 7.25 (ddd, J = 12.2, 8.9, 2.1 Hz, 2H), 7.10 (dd, J = 11.5, 6.0 Hz, 1H), 6.88 (d, J = 8.2 Hz, 1H), 5.54 (s, 2H), 5.23 (d, J = 7.0 Hz, 1H), 4.69-4.38 (m, 3H), 4.33 (dd, J = 11.0, 7.1 Hz, 1H), 3.97 (d, J = 9.2 Hz, 1H), 3.85 (d, J = 9.2 Hz, 1H), 3.50 (s, 3H), 0.72 (s, 3H).55041652.21H NMR (400 MHz, Methanol-d4) δ 8.95-8.82 (m, 2H), 8.21-8.12 (m, 2H), 8.06 (dd, J = 8.2, 2.3 Hz, 1H), 7.95-7.81 (m, 3H), 7.76 (dd, J = 8.6, 0.6 Hz, 1H), 7.63-7.49 (m, 1H), 7.38 (dd, J = 11.1, 6.1 Hz, 1H), 6.96 (dd, J = 8.3, 0.7 Hz, 1H), 5.63 (s, 2H), 5.12 (d, J = 6.5 Hz, 1H), 4.75-4.59 (m, 3H),4.59- 4.45 (m, 1H), 4.39 (s, 3H), 3.99 (d, J = 8.9 Hz, 1H), 3.83 (d, J = 8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H).55141686.21H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.52-8.01 (m, 2H), 7.91 (ddd, J = 10.9, 6.3, 2.0 Hz, 1H), 7.87-7.71 (m, 2H), 7.71-7.23 (m, 7H), 6.91 (dd, J = 8.3, 5.7 Hz, 1H), 5.50 (d, J = 8.5 Hz, 2H), 5.14 (s, 1H), 4.79-4.60 (m, 3H), 4.52 (ddd, J = 11.6, 6.7, 3.2 Hz, 1H), 3.99 (dd, J = 8.9, 3.2 Hz, 1H), 3.84 (dd, J = 8.9, 4.1 Hz, 1H), 1.41 (d, J = 5.4 Hz, 3H), 0.76 (d, J = 4.4 Hz, 3H).55241721.21H NMR (400 MHz, Methanol-d4) δ 8.89 (s, 1H), 8.80- 8.63 (m, 2H), 8.29 (s, 1H), 8.17 (dd, J = 8.6, 1.4 Hz, 1H), 8.05-7.89 (m, 2H), 7.90-7.74 (m, 2H), 7.74-7.35 (m, 3H), 6.96 (dd, J = 8.3, 0.7 Hz, 1H), 5.67 (s, 2H), 5.13 (d, J = 6.5 Hz, 1H), 4.79-4.59 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 3.99 (d, J = 8.9 Hz, 1H), 3.83 (d, J = 8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H).55341669.11H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.18 (dd, J = 8.6, 1.4 Hz, 1H), 8.02 (s, 1H), 7.94 (dd, J = 10.9, 6.3 Hz, 1H), 7.89- 7.72 (m, 2H), 7.71-7.50 (m, 4H), 7.39 (dd, J = 11.2, 6.0 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 5.58 (s, 2H), 5.14 (d, J = 6.5 Hz, 1H), 4.80- 4.59 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 4.22 (s, 3H), 3.99 (d, J = 8.9 Hz, 1H), 3.83 (d, J = 8.9 Hz, 1H), 1.40 (s, 3H), 0.75 (s, 3H).55441669.11H NMR (400 MHz, Methanol-d4) δ 8.88 (s, 1H), 8.16 (dd, J = 8.6, 1.4 Hz, 1H), 7.97-7.86 (m, 2H), 7.84 (dd, J = 8.3, 7.5 Hz, 1H), 7.80-7.68 (m, 2H), 7.59 (dd, J = 7.2, 1.6 Hz, 1H), 7.49-7.33 (m, 3H), 6.95 (dd, J = 8.3, 0.6 Hz, 1H), 5.64 (s, 2H), 5.13 (d, J = 6.6 Hz, 1H), 4.81-4.57 (m, 3H), 4.52 (dd, J = 11.6, 6.7 Hz, 1H), 4.13 (s, 3H), 3.99 (d, J = 8.9 Hz, 1H), 3.84 (d, J = 8.9 Hz, 1H), 1.41 (s, 3H), 0.75 (s, 3H).55541670.11H NMR (400 MHz, Methanol-d4) δ 8.89 (d, J = 9.8 Hz, 1H), 8.83 (s, 1H), 8.23 (s, 1H), 8.12 (dd, J = 8.6, 1.4 Hz, 1H), 8.03-7.90 (m, 1H), 7.88-7.69 (m, 3H), 7.65-7.51 (m, 1H), 7.36 (dd, J =11.2, 6.1 Hz, 1H), 6.94 (dd, J = 8.2, 0.7 Hz, 1H), 5.68 (s, 2H), 5.09 (d, J = 6.4 Hz, 1H), 4.76- 4.56 (m, 3H), 4.52 (dd, J = 11.5, 6.7 Hz, 1H), 4.40 (s,3H), 3.99 (d, J = 8.9 Hz,1H), 3.83 (d, J = 8.9 Hz,1H), 1.40 (s, 3H), 0.74 (s,3H).55635654.01H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.94-7.88 (m, 1H), 7.81 (dd, J = 8.5, 1.5 Hz, 1H), 7.78-7.69 (m, 5H), 7.61 (d, J = 8.4 Hz, 1H), 7.53 (dd, J = 7.5, 1.7 Hz, 1H), 7.38 (dd, J = 11.4, 6.1 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.59 (s, 2H), 5.52 (s, 1H), 4.58- 4.40 (m, 3H), 4.21 (dd, J = 10.8, 6.5 Hz, 1H), 4.08 (t, J = 8.8 Hz, 1H), 3.82-3.72 (m, 2H), 3.18-2.99 (m, 2H), 2.91 (s, 3H).55735606.01H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.89-7.77 (m, 3H), 7.63 (d, J = 8.5 Hz, 1H), 7.61-7.52 (m, 3H), 7.48 (dd, J = 11.5, 6.1 Hz, 1H), 7.30-7.17 (m, 2H), 5.55 (s, 2H), 5.03 (d, J = 6.6 Hz, 1H), 4.62-4.55 (m, 1H), 4.53 (d, J = 1.5 Hz, 1H), 4.48-4.35 (m, 2H), 3.84-3.68 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H).55835624.01H NMR (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 7.90 (dd, J = 10.3, 8.2 Hz, 1H), 7.78 (dd, J = 8.5, 1.5 Hz, 1H), 7.76-7.67 (m, 1H), 7.63-7.53 (m, 4H), 7.31-7.20 (m, 2H), 5.56 (s, 2H), 5.09 (d, J = 6.6 Hz, 1H), 4.73-4.52 (m, 2H), 4.47 (dd, J = 11.2, 6.7 Hz, 1H), 4.35 (d, J = 17.4 Hz, 1H), 3.84-3.70 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H).55935655.21H NMR (400 MHz, MeOD) δ 8.74 (s, 1H), 7.97 (dd, J = 8.5, 1.5 Hz, 1H), 7.69-7.62 (m, 1H), 7.62- 7.52 (m, 1H), 7.41-7.29 (m, 2H), 7.29-7.22 (m, 2H), 7.22-7.10 (m, 2H), 5.49 (t, J = 7.3 Hz, 1H), 5.27- 5.17 (m, 2H), 4.65-4.47 (m, 3H), 4.33-4.22 (m, 1H), 4.00-3.87 (m, 1H), 2.96 (s, 3H).56022668.01H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.82 (s, 1H), 8.30 (s, 1H), 8.25 (d, J = 1.5 Hz, 1H), 8.02 (d, J = 1.3 Hz, 1H), 7.94-7.85 (m, 2H), 7.80 (dd, J = 8.4, 1.6 Hz, 1H), 7.61 (d, J = 8.5 Hz, 1H), 7.57-7.52 (m, 1H), 7.40 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 6.64 (t, J = 75.4 Hz, 1H), 5.67 (s, 2H), 4.72 (t, J = 5.1 Hz, 2H), 4.44 (s, 2H), 4.20 (t, J = 5.0 Hz, 2H).56122660.01H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.82 (s, 1H), 8.30 (s, 1H), 8.27 (d, J = 1.5 Hz, 1H), 8.02 (d, J = 1.2 Hz, 1H), 7.94-7.86 (m, 2H), 7.83 (dd, J = 8.4, 1.5 Hz, 1H), 7.62 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.4, 1.7 Hz, 1H), 7.40 (dd, J = 11.4, 6.1 Hz, 1H), 5.66 (s, 2H), 4.58 (t, J = 5.1 Hz, 2H), 4.52 (s, 2H), 3.71 (t, J = 5.0 Hz, 2H), 3.45 (p, J = 6.0 Hz, 1H), 0.95 (s, 3H), 0.93 (s, 3H56222658.11H NMR (400 MHz, DMSO-d6) δ 8.90 (d, J = 1.3 Hz, 1H), 8.83 (s, 1H), 8.31 (s, 1H), 8.26 (d, J = 1.5 Hz, 1H), 8.03 (d, J = 1.3 Hz, 1H), 7.96-7.86 (m, 2H), 7.84 (dd, J = 8.5, 1.6 Hz, 1H), 7.63 (d, J = 8.4 Hz, 1H), 7.55 (dd, J = 7.5, 1.7 Hz, 1H), 7.40 (dd, J = 11.5, 6.1 Hz, 1H), 7.01 (d, J = 8.3 Hz, 1H), 5.67 (s, 2H), 4.60 (t, J = 5.1 Hz, 2H), 4.46 (s, 2H), 3.79 (t, J = 5.0 Hz, 2H), 3.25 (tt, J = 6.1, 3.0 Hz, 1H), 0.31 (ddd, J = 13.8, 6.6,2.7 Hz, 2H), 0.24 (tt, J = 6.3, 3.2 Hz, 2H).56335660.21H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.2 Hz, 1H), 8.81 (s, 1H), 8.39 (d, J = 1.5 Hz, 1H), 8.30 (s, 1H), 8.03 (d, J = 1.3 Hz, 1H), 7.93-7.86 (m, 2H), 7.84 (dd, J = 8.5, 1.3 Hz, 1H), 7.62 (d, J = 8.4 Hz, 1H), 7.55 (dd, J = 7.4, 1.7 Hz, 1H), 7.41 (dd, J = 11.5, 6.1 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.66 (s, 2H), 4.55 (s, 2H), 4.50 (s, 2H), 3.10 (s, 3H), 1.21 (s, 6H).56435601.21H NMR (400 MHz, DMSO-d6) δ 8.32 (d, J = 1.5 Hz, 1H), 7.90-7.85 (m, 1H), 7.85-7.78 (m, 2H), 7.64-7.57 (m, 2H), 7.53- 7.47 (m, 2H), 7.38 (dd, J = 11.5, 6.1 Hz, 1H), 7.32 (dd, J = 8.2, 2.1 Hz, 1H), 6.94 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 4.70 (s, 1H), 4.45 (q, J = 16.8 Hz, 2H), 3.93-3.81 (m, 1H), 3.00 (s, 3H), 1.62 (d, J = 7.1 Hz, 3H), 1.21 (d, J = 6.1 Hz, 3H).56535610.21H NMR (400 MHz, DMSO-d6) δ 8.33 (d, J = 1.5 Hz, 1H), 7.91-7.85 (m, 1H), 7.85-7.79 (m, 2H), 7.63 (d, J = 8.3 Hz, 1H), 7.59 (d, J = 8.2 Hz, 1H), 7.50 (td, J = 10.0, 9.3, 1.9 Hz, 2H), 7.39 (dd, J = 11.5, 6.1 Hz, 1H), 7.32 (dd, J = 8.3, 2.1 Hz, 1H), 6.94 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 4.71 (s, 1H), 4.46 (q, J = 16.8 Hz, 2H), 3.88 (dd, J = 8.1, 6.1 Hz, 1H), 3.00 (s, 3H), 1.62 (d, J = 7.1 Hz, 3H), 1.21 (d, J = 6.1 Hz, 3H).56622624.31H NMR (400 MHz, DMSO-d6) δ 8.26 (d, J = 1.5 Hz, 1H), 7.91-7.85 (m, 1H), 7.84-7.82 (m, 1H), 7.81 (d, J = 1.5 Hz, 1H), 7.66-7.56 (m, 1H), 7.61 (d, J = 3.2 Hz, 1H), 7.54-7.46 (m, 2H), 7.37 (dd, J = 11.5, 6.1 Hz, 1H), 7.33 (dd, J = 8.4, 2.1 Hz, 1H), 6.95 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 4.61 (t, J = 5.1 Hz, 2H), 4.50 (s, 2H), 3.71 (t, J = 5.0 Hz, 2H), 3.10 (d, J = 6.5 Hz, 2H), 1.66 (hept, J = 6.7 Hz, 1H), 0.72 (s, 3H), 0.70 (s, 3H).56738686.31H NMR (400 MHz, DMSO-d6) δ 8.73 (s, 1H), 8.26 (d, J = 1.5 Hz, 1H), 8.04 (s, 1H), 7.93-7.86 (m, 2H), 7.84 (dd, J = 8.4, 1.5 Hz, 1H), 7.64 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.41 (dd, J = 11.5, 6.1 Hz, 1H), 6.99 (d, J = 8.2 Hz, 1H), 5.62 (s, 2H), 4.63 (t, J = 5.1 Hz, 2H), 4.50 (s, 2H), 3.69 (t, J = 5.0 Hz, 2H), 3.21 (s, 3H), 2.77 (d, J = 5.5 Hz, 4H), 1.83 (q, J = 3.3 Hz, 4H).56838686.11H NMR (400 MHz, DMSO-d6) δ 8.79 (s, 1H), 8.24 (d, J = 1.5 Hz, 1H), 8.20 (s, 1H), 7.91 (t, J = 7.9 Hz, 1H), 7.88-7.79 (m, 2H), 7.62 (d, J = 8.4 Hz, 1H), 7.54 (dd, J = 7.5, 1.7 Hz, 1H), 7.40 (dd, J = 11.5, 6.0 Hz, 1H), 7.00 (d, J = 8.2 Hz, 1H), 5.64 (s, 2H), 4.62 (t, J = 5.2 Hz, 2H), 4.48 (s, 2H), 3.68 (t, J = 5.1 Hz, 2H), 3.21 (s, 3H), 3.03 (s, 2H), 2.71 (s, 2H), 1.82-1.71 (m, 4H).56922674.11H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 1.3 Hz, 1H), 8.82 (s, 1H), 8.30 (s, 1H), 8.28 (d, J = 1.5 Hz, 1H), 8.02 (d, J = 1.3 Hz, 1H), 7.94-7.90 (m, 1H), 7.90-7.86 (m, 1H), 7.84 (dd, J = 8.4, 1.5 Hz, 1H), 7.63 (d, J = 8.5 Hz, 1H), 7.54 (dd, J = 7.3, 1.7 Hz, 1H), 7.39 (dd, J = 11.4, 6.1 Hz, 1H), 7.00 (dd, J = 8.3, 0.6 Hz, 1H), 5.66 (s, 2H), 4.63 (t, J = 5.0 Hz, 2H), 4.53 (s, 2H), 3.71 (t, J = 4.9 Hz, 2H), 3.10 (d, J = 6.5 Hz, 2H), 1.65 (hept, J = 6.7 Hz, 1H), 0.72 (s, 3H), 0.70 (s, 3H).57037652.21H NMR (400 MHz, DMSO) δ 8.36 (d, J = 1.5 Hz, 1H), 7.93-7.79 (m, 3H), 7.71 (d, J = 8.5 Hz, 1H), 7.60 (t, J = 8.2 Hz, 1H), 7.56-7.46 (m, 2H), 7.46- 7.26 (m, 2H), 6.95 (d, J = 8.3 Hz, 1H), 5.50 (s, 2H), 5.29 (dt, J = 7.4, 3.5 Hz, 1H), 4.67-4.13 (m, 6H), 3.78-3.59 (m, 1H), 3.37 (m, J = 7.0 Hz, 1H), 1.00 (d, J = 7.0 Hz, 6H).57137638.11H NMR (400 MHz, DMSO) δ 8.34 (d, J = 1.5 Hz, 1H), 7.93-7.79 (m, 3H), 7.69 (d, J = 8.5 Hz, 1H), 7.60 (t, J = 8.2 Hz, 1H), 7.50 (ddd, J = 8.6, 5.6, 2.0 Hz, 2H), 7.45-7.27 (m, 2H), 6.95 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 5.39- 5.18 (m, 1H), 4.64-4.46 (m, 2H), 4.46-4.34 (m, 2H), 4.34-4.12 (m, 2H), 3.71 (h, J = 5.2 Hz, 1H), 3.45-3.30 (m, 2H), 1.00 (t, J = 7.0 Hz, 3H).57235620.01H NMR (400 MHz, DMSO-d6) δ 8.71 (d, J = 1.4 Hz, 1H), 7.92-7.81 (m, 3H), 7.67-7.58 (m, 2H), 7.55-7.43 (m, 3H), 7.34 (dd, J = 8.2, 2.1 Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.46 (q, J = 4.5, 3.8 Hz, 1H), 4.74-4.63 (m, 2H), 4.60-4.40 (m, 3H), 3.08 (p, J = 5.5 Hz, 1H), 2.36 (ddd, J = 11.0, 5.8, 3.6 Hz, 1H), 2.28 (dd, J = 10.9, 9.2 Hz, 1H), 2.13-2.04 (m, 1H), 2.00 (dd, J = 10.7, 9.2 Hz, 1H).57335620.01H NMR (400 MHz, DMSO-d6) δ 8.71 (d, J = 1.4 Hz, 1H), 7.92-7.81 (m, 3H), 7.67-7.58 (m, 2H), 7.55-7.43 (m, 3H), 7.34 (dd, J = 8.2, 2.1Hz, 1H), 6.96 (d, J = 8.3 Hz, 1H), 5.51 (s, 2H), 5.46 (q, J = 4.5, 3.8 Hz, 1H), 4.74-4.63 (m, 2H), 4.60-4.40 (m, 3H), 3.08 (p, J = 5.5 Hz, 1H), 2.36 (ddd, J = 11.0, 5.8, 3.6 Hz, 1H), 2.28 (dd, J = 10.9, 9.2 Hz, 1H), 2.13-2.04 (m, 1H), 2.00 (dd, J = 10.7, 9.2 Hz, 1H).57435607.01H NMR (400 MHz, DMSO) δ 8.84 (d, J = 5.1 Hz, 1H), 8.48 (s, 1H), 7.84 (ddd, J = 9.9, 5.6, 2.0 Hz, 1H), 7.78 (dd, J = 8.4, 1.5 Hz, 1H), 7.68 (dd, J = 5.1, 1.8 Hz, 1H), 7.63-7.53 (m, 3H), 7.30-7.20 (m, 2H), 5.50 (s, 2H), 5.13-5.04 (m, 1H), 4.68 (d, J = 17.3 Hz, 1H), 4.57 (d, J = 11.5 Hz, 1H), 4.51-4.34 (m, 2H), 3.79-3.74 (m, 2H), 1.39 (s, 3H), 0.66 (s, 3H).57535589.11H NMR (400 MHz, DMSO) δ 8.78 (d, J = 5.2 Hz, 1H), 8.48 (s, 1H), 7.94 (dd, J = 10.2, 6.3 Hz, 1H), 7.80 (dd, J = 8.4, 1.5 Hz, 1H), 7.70-7.50 (m, 5H), 7.29-7.19 (m, 2H), 5.49 (s, 2H), 5.02 (d, J = 6.7 Hz, 1H), 4.61-4.50 (m, 2H), 4.49-4.36 (m, 2H), 3.81- 3.70 (m, 2H), 1.34 (s, 3H), 0.61 (s, 3H)57642734.01H NMR (400 MHz, DMSO-d6) δ 8.52 (s, 1H), 7.88 (dd, J = 8.2, 7.5 Hz, 1H), 7.85-7.79 (m, 2H), 7.65 (d, J = 8.6 Hz, 1H), 7.60 (t, J = 8.2 Hz, 1H), 7.55- 7.51 (m, 1H), 7.49 (dd, J = 10.1, 2.1 Hz, 1H), 7.47- 7.42 (m, 1H), 7.33 (dd, J = 8.3, 2.1 Hz, 1H), 6.95 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 5.23 (s, 2H), 5.02 (d, J = 6.6 Hz, 1H), 4.53 (d, J = 13.9 Hz, 2H), 4.48-4.30 (m, 2H), 3.80-3.68 (m, 2H), 2.23 (s, 3H), 1.34 (s, 3H), 1.21 (d, J = 16.9 Hz, 1H), 0.59 (s, 3H57743731.91H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.92-7.80 (m, 3H), 7.68 (d, J = 8.5 Hz, 1H), 7.60 (t, J = 8.2 Hz, 1H), 7.53 (dd, J = 7.5, 1.6 Hz, 1H), 7.49 (dd, J = 10.0, 2.1 Hz, 1H), 7.47- 7.43 (m, 1H), 7.33 (dd, J = 8.4, 2.1 Hz, 1H), 6.95 (d, J = 8.4 Hz, 1H), 5.82-5.68 (m, 2H), 5.51 (s, 2H), 5.03 (d, J = 6.7 Hz, 1H), 4.60-4.55 (m, 1H), 4.53 (s, 1H), 4.48- 4.35 (m, 2H), 3.79 (d, J = 8.7 Hz, 1H), 3.73 (d, J = 8.6 Hz, 1H), 1.34 (s, 3H), 0.60 (s, 3H).57842722.1 C. Biological Data GLP-1R Activation—cAMP Assay 1 GLP-1R activation by compounds of the present disclosure were quantified by measuring cAMP increase in CHO cells stably expressing GLP-1R (MultiSpan product #C1267-1a). The cells were harvested and plated in growth medium (DMEM/F-12 (Corning product #10-090-CV) supplemented with 10% FBS (HyClone product #SH30071-03), penicillin/streptomycin (Corning product #30-002CI) and 10 g/ml puromycin (Gibco product #A11138-03)) at 1,000 cells/well in a 384-well plate (Greiner product #781080). The cells were then incubated overnight at 37° C., 5% CO2. The next day, the medium was removed and the cells were washed with DPBS (Corning product #21-031-CM) before adding the assay medium (HBSS, Corning product #21-023-CV) with 20 mM Hepes (Gibco product #15630-080) and 0.1% BSA (Rockland Immunochemicals product #BSA-1000)). Following the medium change, the cells were incubated for 1 hour at 37° C., 5% CO2. The tested GLP-1 compound was added to the cells in a 10-point dose response followed by a 30 minutes incubation at 37° C., 5% CO2. cAMP concentration increase was then detected using Cisbio's cAMP Gs Dynamic Kit (product #62AM4PEC) according to the manufacturer's protocol. The response was plotted against the log of the agonist concentration and fitted to a sigmoidal equation to determine the EC50. GLP-1R Activation—cAMP Assay 2 GLP-1R activation by small molecule agonists was quantified by measuring cAMP increase in CHO cells stably expressing GLP-1R (MultiSpan product #C1267-1a). 50 nL of agonists were pre-spotted in a 10-point dose response onto 384-well plate (Corning product #CL3826) using the Labcyte Echo System. The cells were harvested and plated in assay buffer (HIBSS (Corning product #21-023-CV) with 20 mM Hepes (Gibco product #15630-080) and 0.100 BSA (Rockland Immunochemicals product #BSA-1000) at 1,000 cells/well, 10 μL/well, onto the pre-spotted plates. The cells are then incubated for 30 minutes at 37° C., 500 CO2. cAMP concentration increase was then detected using Cisbio's cAMP Gs Dynamic Kit (product #62AM4PEC) according to the manufacturer's protocol. The response was plotted against the log of the agonist concentration and fitted to a sigmoidal equation to determine the EC50. All compounds were run on one of the two GLP-1R Activation cAMP assays and the result are listed in Table 3, below. TABLE 3ActivityExampleGLP-1R Activation -No.Assay EC50 (nM)11.122155.78311.5342.6550.7768.4970.60821.2497.8810952.92110.26120.03130.191478.661570.99164800.27175.611849.19190.60200.35210.11220.72234.57249.96257.552618.682713.08281.462935.983049.903130.43323.43332.58343.25352.67361.04371.97384.02398.9840154.83414.88424.254320.80440.814516.734620.95470.78480.214945.59505.24510.835221.27535.115431.605525.8456902.70572.06580.58590.32600.04610.83627.32632.55641.28655.056612.86677.25680.336913.84700.26711.197237.00732.40744.387515.6376100.00771.91780.36795.62804.918140.86821.60830.62841.34851443.1186140.288722.5388136.088915.33900.70912.06920.13930.359423.95950.97960.989721.22981.43990.601000.451011.4610222.791031.2310441.301051.551062.221071.401083.281092.511104.931111.4611223.0311311.001145.991150.1221160.0051170.0441180.0531190.0311200.1011210.0831220.0311230.5281240.0271250.1121260.3321270.3531290.2711300.0381310.1451320.0431330.0211340.0771350.3611360.3121370.0391380.1361390.0231400.1581410.0791420.8941430.6141441.0841450.0221460.3791470.2331480.1741490.0821500.0631510.2341520.1151530.0111540.0341550.4161560.0641570.0811580.0321590.0431600.0351610.0151620.1871630.0391640.0851650.5531660.2221670.0271680.3881690.1711700.2271710.051720.1291730.0181740.0511750.106176A0.085176B0.7481770.0211780.4991790.0641800.0891810.0251820.1781830.1291840.1571850.5941860.0181870.3911880.0321890.1491900.0611910.3531920.4441930.1321940.2651950.5391960.5081976.621980.50819990.912000.5082010.0282024.00620338.2992040.092050.0972060.1912070.1682080.24920912.32921014.112110.2582120.3792130.0422140.2992150.212160.0562171.1062180.2792190.1892200.2162210.5082220.1362230.1262240.5152257.1412260.0222274.7992280.0472290.1352300.0342310.0492320.1182333.1082340.4482350.1912360.2032370.522380.12390.042400.1272410.0132420.022430.0222440.0152450.0512460.0652470.1512480.1862490.1372500.0482510.0792520.0592530.0712540.0882550.1082560.1172570.0642580.242590.5782600.4112614.0792621.0732630.7092640.1552650.1712660.3632670.0552680.1752690.1642706094.512710.7172722.112730.9422740.6432750.1682760.1082770.342780.0852790.1952800.1652810.6552820.1922830.4772840.1352850.0962860.0992870.0512880.041289A0.122289B0.931290A0.063290B0.7642910.0762920.1332930.042940.0212950.0422960.0662970.1512980.6632990.3423000.2683010.7143020.1813030.173050.1253060.1233070.5083080.183090.3683100.032311A0.064311B0.1593120.3973131.1423160.143170.4513180.7453190.3463200.8273210.0833220.3463230.7343240.0493250.1553260.2363270.2663280.5553290.0623300.0113310.2183320.1593340.0673350.3063360.0163370.0253390.0133400.0053410.0243420.0123430.1133440.1473450.1973460.0753470.1323480.553490.0423500.1713510.0743520.5353531.1793540.5283551.2123570.1713580.1243590.0683600.0793610.1313620.2663630.3693640.1583650.1243661.1713670.2743680.5623690.3643700.0423710.1273720.1633730.2943740.5713750.2723760.0563770.1883780.3213790.5743800.0963810.2673820.604383A0.747383B0.9713840.205385A0.089385B1.1173860.0933870.0433880.0533890.553903.1173910.1073920.0363930.6523940.2433960.1573970.5463980.2173990.1244000.444010.2474020.5614030.0814040.0684050.33740610.8294070.4214080.6334090.2914100.3494111.3314120.4624130.2814140.2614150.4914160.2354170.194180.1994190.1124200.4134213.0214223.1574232.5444240.594250.4944261.484270.5014280.1054290.1394300.024310.3774320.1144330.2094340.0584350.064360.9934370.6444380.5194390.1714400.9564410.7224420.0424430.214440.3654450.2794460.24471.3864480.3524490.0214501.0394510.6214520.5644530.5274540.4774550.1264560.3324570.1574580.3154590.84600.0964610.0674620.1414630.2094640.1724650.184660.1244670.0774680.1184691.5994701.6094710.0414720.0694730.0924740.1814750.6014760.6144770.1714780.254790.2494800.2234810.8834820.0114830.0224840.1554850.2034860.0544870.1594880.2824890.3424900.1824910.0794920.1914930.2784940.0514950.1444960.0264970.2064980.5084990.5085000.2035010.508502A0.281502B0.9395030.0815040.1485050.0435060.2425070.3515080.3025090.1445100.5095110.5885120.5155130.2085140.1525150.0745160.0885170.0245180.8875190.1025200.275210.1115220.1655230.3625240.6055250.4295260.5095270.1845280.0475290.5985300.3925310.3965323678.85330.55340.3535350.0995360.0835370.1215380.1095390.4625400.0525410.4345420.0335430.0385440.0485450.0445460.7065470.0245490.7275500.0665510.115520.995530.1325540.0265550.0475560.2625570.0315580.1325590.1955600.1925610.5585620.2075630.0415642.2935650.1945664.0875671005681.5645690.9875700.9555711.2665720.3225730.1885740.4575750.186 Pharmacokinetic (PK) Studies Test Articles: Examples 60, 115, 117, and 119 were tested along with comparative examples CE-1 and CE-2. Dose Formulation Preparation Compounds CE-2, Example 60, Example 115, Example 117, and Example 119 were combined with a mixture of 1000 NMP (v:v) and 9000 PEG300 (v:v), and the mixture was stirred until a homogeneous solution was obtained. Compound CE-1 was combined with a mixture of 15% 2-hydroxypropyl-β-cyclodextrin (w:v) and 85% Mili-Q water (v:v), and the mixture was stirred until a homogeneous solution was obtained. Study Design Young adult non-naive Cynomolgus monkeys with weight about 2.5-5 kg (n=1-3/test article/route) were fasted overnight and then dosed with either CE-1, CE2, Example 60, Example 115, Example 117 or Example 119. Food was supplied approximately 4 hours post dose and animals were provided with free access to water all the time. Animals were restrained at designated time points for blood sampling. Approximately 500 μL of blood samples was taken via cephalic or saphenous vein into EDTA-K2 tubes. For oral administration (PO): blood samples were generally collected pre-dose, and at 15 min, 30 min, 1 hr., 2 hr., 4 hr., 8 hr., 12 hr., and 24 hr. time points after dosing. Blood was be maintained on wet ice, in chilled cryoracks, or at approximately 5° C. prior to centrifugation to obtain plasma. Centrifugation was conducted within 1 hour of collection. Plasma (approximately 200 μL) was placed into a micronic tubes containing 4 μL of formic acid (the final concentration of formic acid in plasma was approximately 2%) and samples were vortex mixed. Samples were maintained on dry ice prior to storage at approximately −70° C. Bioanalytical Analysis LC-MS/MS assays were utilized to quantify the concentration of the test articles in plasma as follows. Sample Preparation 50 μL plasma sample was mixed with 350 μL of ACN combined with an internal standard. The mixture was then vortexed for about 1-5 min, and centrifuged at about 4,000-5,800 rpm for about 10-15 mm. An aliquot of 1 μL supernatant was used for LC-MS/MS analysis. LC-MS/MS Method MS conditions: A Sciex API 6500 equipped with electrospray ionization in the positive ion selected reaction monitoring mode was used for detection. HPLC conditions Mobile Phase A: H2O-0.11% Formic Acid Mobile Phase B: ACN-0.1% Formic Acid MobileTimePhase(min)B (%)0.10350.55501.10951.22951.26351.50stopColumn: Phenomenex Kinetex XBC18 (2.1×50 mm, 2.6 μm) with a flow rate of 1.20 mL/minColumn temperature: 60° C.Retention time: approximately between 0.85 to 1.05 min Pharmacokinetics (PK) Parameters Pharmacokinetics parameters, including area under the curve (AUC0-24), maximum plasma concentration (Cmax), time to reach maximum plasma concentration (Tmax), oral bioavailability (F %), etc., were calculated using Dotmatics software in non-compartmental model. The results of the pharmacokinetics studies are presented inFIG.1and Table 4. As is apparent inFIG.1CE-1 demonstrates low exposures (<10 nM plasma concentrations) over a period of 12 h after a single oral dose of 1 mg/kg that subsequently drop to below the detection limit. In comparison, CE-2 achieves disproportionately higher exposures 2 mg/kg at a 2-fold higher dose than CE-1 and has sustained exposures over 24 h after a single oral dose. However, the oral bioavailability for CE-1 and CE-2 is low, 2.5% and 27.4%, respectively. In contrast, as illustrated inFIGS.1&2the plasma concentrations of orally-administered Examples 60, 115, 117, and 119 at the same dose level as CE-2 (i.e., 2 mg/kg were higher than CE-1 (117-407-fold higher AUC0-24despite only 2-fold higher dose), and CE-2 (2-7-fold higher AUC0-24for the same dose) over 24 hours after oral dosing. Further, the oral bioavailability of Examples 60, 115, 117 and were higher than that of CE-1 and CE-2. These data show that compounds disclosed herein can provide prolonged exposures and/or higher oral bioavailability at least within 24 h following a single oral dose administration of compound. TABLE 4In vivo Monkey Oral PK ProfilesOral doseCmaxAUC0-24Compound(Arbitrary(ArbitraryTmax(ArbitraryNo.unit)unit)(hr)units)F %Example 602 mg/kg385 ± 821.5 ± 0.95570 ± 219047.6 ± 12.3Example 1152 mg/kg1540 ± 1662.7 ± 1.217900 ± 204042.0 ± 16.5Example 1172 mg/kg1600 ± 8324.7 ± 2.312800 ± 401052.2 ± 16.3Example 1192 mg/kg1190 ± 1371.7± 0.619400 ± 331047.6 ± 8.1Compound CE-11 mg/kg4.5 ± 0.82.2 ± 1.847.7 ± 13.32.5 ± 0.2Compound CE-22 mg/kg224 ± 1304.0 ± 2.02810 ± 133027.4 ± 12.9 Although the foregoing has been described in some detail by way of illustration and Example for purposes of clarity of understanding, one of skill in the art will appreciate that certain changes and modifications may be practiced within the scope of the appended claims. In addition, each reference provided herein is incorporated by reference in its entirety to the same extent as if each reference was individually incorporated by reference. Where a conflict exists between the instant application and a reference provided herein, the instant application shall dominate.
1,242,560
11858919
DETAILED DESCRIPTION Provided herein are compounds, e.g., the compounds of Formulae I, II, III, and IV, or pharmaceutically acceptable salts thereof, that are useful in the treatment of cancer, myelodysplastic syndrome or hemoglobinopathy in a subject. In a non-limiting aspect, these compounds can inhibit histone deacetylases. In a particular embodiment, the compounds provided herein are considered HDAC1 and/or HDAC2 inhibitors. As such, in one aspect, the compounds provided herein are useful in the treatment of cancer, myelodysplastic syndrome or hemoglobinopathy in a subject by acting as a HDAC1 and/or HDAC2 inhibitor. Definitions Listed below are definitions of various terms used herein. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art. Generally, the nomenclature used herein and the laboratory procedures in cell culture, molecular genetics, organic chemistry, and peptide chemistry are those well-known and commonly employed in the art. As used herein, the articles “a” and “an” refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element. Furthermore, use of the term “including” as well as other forms, such as “include,” “includes,” and “included,” is not limiting. As used herein, the term “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. As used herein when referring to a measurable value such as an amount, a temporal duration, and the like, the term “about” is meant to encompass variations of ±20% or +10%, including ±5%, 1%, and ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods. The term “treat,” “treated,” “treating,” or “treatment” includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated. In certain embodiments, the treatment comprises bringing into contact with HDAC1 and/or HDAC2 an effective amount of a compound provided herein for conditions related to cancers, hemoglobinopathies, or myelodysplastic syndrome. As used herein, the term “prevent” or “prevention” means no disorder or disease development if none had occurred, or no further disorder or disease development if there had already been development of the disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the disorder or disease. As used herein, the term “patient,” “individual,” or “subject” refers to a human or a non-human mammal. Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline and marine mammals. Preferably, the patient, subject, or individual is human. As used herein, the terms “effective amount,” “pharmaceutically effective amount,” and “therapeutically effective amount” refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation. As used herein, the term “pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained. As used herein, the term “pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts described herein include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts discussed herein can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. The phrase “pharmaceutically acceptable salt” is not limited to a mono, or 1:1, salt. For example, “pharmaceutically acceptable salt” also includes bis-salts, such as a bis-hydrochloride salt. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety. As used herein, the term “composition” or “pharmaceutical composition” refers to a mixture of at least one compound with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary, and topical administration. As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful to the patient such that it may perform its intended function. Typically, such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound disclosed herein, and not injurious to the patient. Some examples of materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations. As used herein, “pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound disclosed herein, and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions. The “pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound disclosed herein. Other additional ingredients that may be included in the pharmaceutical compositions are known in the art and described, for example, in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, Pa.), which is incorporated herein by reference. The term “HDAC” refers to histone deacetylases, which are enzymes that remove the acetyl groups from the lysine residues in core histones, thus leading to the formation of a condensed and transcriptionally silenced chromatin. There are currently 18 known histone deacetylases, which are classified into four groups. Class I HDACs, which include HDAC1, HDAC2, HDAC3, and HDAC8, are related to the yeast RPD3 gene. Class II HDACs, which include HDAC4, HDAC5, HDAC6, HDAC7, HDAC9, and HDAC10, are related to the yeast Hda1 gene. Class III HDACs, which are also known as the sirtuins are related to the Sir2 gene and include SIRT1-7. Class IV HDACs, which contains only HDAC11, has features of both Class I and II HDACs. The term “HDAC” refers to any one or more of the 18 known histone deacetylases, unless otherwise specified. As used herein, the term “alkyl,” by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e., C1-C6-alkyl means an alkyl having one to six carbon atoms) and includes straight and branched chains. In an embodiment, C1-C6alkyl groups are provided herein. Examples include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, and hexyl. Other examples of C1-C6-alkyl include ethyl, methyl, isopropyl, isobutyl, n-pentyl, and n-hexyl. As used herein, the term “alkoxy,” refers to the group —O-alkyl, wherein alkyl is as defined herein. Alkoxy includes, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, t-butoxy and the like. In an embodiment, C1-C6alkoxy groups are provided herein. As used herein, the term “halo” or “halogen” alone or as part of another substituent means, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom, preferably, fluorine, chlorine, or bromine, more preferably, fluorine or chlorine. As used herein, the term “cycloalkyl” means a non-aromatic carbocyclic system that is partially or fully saturated having 1, 2 or 3 rings wherein such rings may be fused. The term “fused” means that a second ring is present (i.e., attached or formed) by having two adjacent atoms in common (i.e., shared) with the first ring. Cycloalkyl also includes bicyclic structures that may be bridged or spirocyclic in nature with each individual ring within the bicycle varying from 3-8 atoms. The term “cycloalkyl” includes, but is not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[3.1.0]hexyl, spiro[3.3]heptanyl, and bicyclo[1.1.1]pentyl. In an embodiment, C4-C7cycloalkyl groups are provided herein. As used herein, the term “heterocycloalkyl” means a non-aromatic carbocyclic system containing 1, 2, 3 or 4 heteroatoms selected independently from N, O, and S and having 1, 2 or 3 rings wherein such rings may be fused, wherein fused is defined above. Heterocycloalkyl also includes bicyclic structures that may be bridged or spirocyclic in nature with each individual ring within the bicycle varying from 3-8 atoms, and containing 0, 1, or 2 N, O, or S atoms. The term “heterocycloalkyl” includes cyclic esters (i.e., lactones) and cyclic amides (i.e., lactams) and also specifically includes, but is not limited to, epoxidyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl (i.e., oxanyl), pyranyl, dioxanyl, aziridinyl, azetidinyl, pyrrolidinyl, 2,5-dihydro-1H-pyrrolyl, oxazolidinyl, thiazolidinyl, piperidinyl, morpholinyl, piperazinyl, thiomorpholinyl, 1,3-oxazinanyl, 1,3-thiazinanyl, 2-azabicyclo[2.1.1]hexanyl, 5-azabicyclo[2.1.1]hexanyl, 6-azabicyclo[3.1.1] heptanyl, 2-azabicyclo[2.2.1]heptanyl, 3-azabicyclo[3.1.1]heptanyl, 2-azabicyclo[3.1.1]heptanyl, 3-azabicyclo[3.1.0]hexanyl, 2-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[3.2.1]octanyl, 8-azabicyclo[3.2.1]octanyl, 3-oxa-7-azabicyclo[3.3.1]nonanyl, 3-oxa-9-azabicyclo[3.3.1]nonanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 6-oxa-3-azabicyclo[3.1.1]heptanyl, 2-azaspiro[3.3]heptanyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2-oxaspiro[3.3]heptanyl, 2-oxaspiro[3.5]nonanyl, 3-oxaspiro[5.3]nonanyl, and 8-oxabicyclo[3.2.1]octanyl. In an embodiment, C2-C7heterocycloalkyl groups are provided herein. As used herein, the term “aromatic” refers to a carbocycle or heterocycle with one or more polyunsaturated rings and having aromatic character, i.e., having (4n+2) delocalized π (pi) electrons, where n is an integer. As used herein, the term “aryl” means an aromatic carbocyclic system containing 1, 2 or 3 rings, wherein such rings may be fused, wherein fused is defined above. If the rings are fused, one of the rings must be fully unsaturated and the fused ring(s) may be fully saturated, partially unsaturated or fully unsaturated. The term “aryl” includes, but is not limited to, phenyl, naphthyl, indanyl, and 1,2,3,4-tetrahydronaphthalenyl. In some embodiments, aryl groups have 6 carbon atoms. In some embodiments, aryl groups have from six to ten carbon atoms. In some embodiments, aryl groups have from six to sixteen carbon atoms. In an embodiment, C5-C7aryl groups are provided herein. As used herein, the term “heteroaryl” means an aromatic carbocyclic system containing 1, 2, 3, or 4 heteroatoms selected independently from N, O, and S and having 1, 2, or 3 rings wherein such rings may be fused, wherein fused is defined above. The term “heteroaryl” includes, but is not limited to, furanyl, thiophenyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, imidazo[1,2-a]pyridinyl, pyrazolo[1,5-a]pyridinyl, 5,6,7,8-tetrahydroisoquinolinyl, 5,6,7,8-tetrahydroquinolinyl, 6,7-dihydro-5H-cyclopenta[b]pyridinyl, 6,7-dihydro-5H-cyclopenta[c]pyridinyl, 1,4,5,6-tetrahydrocyclopenta[c]pyrazolyl, 2,4,5,6-tetrahydrocyclopenta[c]pyrazolyl, 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazolyl, 6,7-dihydro-5H-pyrrolo[1,2-b][1,2,4]triazolyl, 5,6,7,8-tetrahydro-[1,2,4]triazolo[1,5-a]pyridinyl, 4,5,6,7-tetrahydropyrazolo[1,5-a]pyridinyl, 4,5,6,7-tetrahydro-1H-indazolyl and 4,5,6,7-tetrahydro-2H-indazolyl. In an embodiment, C2-C7heteroaryl groups are provided herein. It is to be understood that if an aryl, heteroaryl, cycloalkyl, or heterocycloalkyl moiety may be bonded or otherwise attached to a designated moiety through differing ring atoms (i.e., shown or described without denotation of a specific point of attachment), then all possible points are intended, whether through a carbon atom or, for example, a trivalent nitrogen atom. For example, the term “pyridinyl” means 2-, 3- or 4-pyridinyl, the term “thienyl” means 2- or 3-thioenyl, and so forth. As used herein, the term “substituted” means that an atom or group of atoms has replaced hydrogen as the substituent attached to another group. As used herein, the term “optionally substituted” means that the referenced group may be substituted or unsubstituted. In one embodiment, the referenced group is optionally substituted with zero substituents, i.e., the referenced group is unsubstituted. In another embodiment, the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from groups described herein. Compounds In an aspect, provided herein are compounds of Formula I: or a pharmaceutically acceptable salt thereof; wherein: R1is selected from the group consisting of aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with —OH or —OSO2NHCH3; R2is selected from the group consisting of —H, C1-C6alkyl, alkyl-cycloalkyl, alkyl-heterocycloalkyl, alkyl-aryl, alkyl-heteroaryl, aryl, heteroaryl, and halo; and R3and R4are independently, at each occurrence, selected from the group consisting of —H, halo, alkyl, aryl, heteroaryl, cycloalkyl, and heterocycloalkyl. In an embodiment, R1is selected from the group consisting of aryl, aryl-OH, and heteroaryl; R2is selected from the group consisting of —H, methyl, alkyl-cycloalkyl, and alkyl-heterocycloalkyl; and R3and R4are independently, at each occurrence, selected from the group consisting of —H, halo, and methyl. In another embodiment, R1is selected from the group consisting of phenyl, phenyl-OH, thienyl, and furanyl. In yet another embodiment, R1is phenyl. In still another embodiment, R1is phenyl-OH. In an embodiment, R1is thienyl. In another embodiment, R1is furanyl. In yet another embodiment, R2is selected from the group consisting of —H, methyl, CH2-cycloalkyl, and C1-C3alkyl-5-6 membered heterocycloalkyl. In another embodiment, R2is —H. In yet another embodiment, R2is methyl. In still another embodiment, R2is CH2-cycloalkyl. In an embodiment, R2is C1-C3alkyl-5-6 membered heterocycloalkyl. In another embodiment, R3and R4are independently, at each occurrence, selected from the group consisting of —H, halo, and methyl. In yet another embodiment, R3and R4are independently, at each occurrence, —H. In still another embodiment, R3and R4are independently, at each occurrence, halo. In an embodiment, R3and R4are independently, at each occurrence, methyl. In another embodiment, aryl is a 6-10 membered aromatic ring and heteroaryl is a 5-10 membered heteroaromatic ring. In another aspect, provided herein are compounds of Formula II: or a pharmaceutically acceptable salt thereof; wherein: R1is selected from the group consisting of aryl and heteroaryl, wherein aryl or heteroaryl are optionally substituted with —OH or —OSO2NHCH3; R2is selected from the group consisting of absent, —H, C1-C6alkyl, alkyl-cycloalkyl, alkyl-heterocycloalkyl, aryl, and heteroaryl; R3is selected from the group consisting of absent, —H, C1-C6alkyl, alkyl-cycloalkyl, alkyl-heterocycloalkyl, aryl, and heteroaryl; R4is selected from the group consisting of —H, —OH, alkyl, aryl, cycloalkyl, and heteroaryl; and indicates an optional double bond. In an embodiment of Formula II, R1is aryl or heteroaryl; R2is selected from the group consisting of absent, —H, methyl, and alkyl-heterocycloalkyl; R3is selected from the group consisting of absent, —H, and alkyl-heterocycloalkyl; and R4is selected from the group consisting of —H, —OH, cycloalkyl, and methyl. In another embodiment, R1is phenyl or thienyl. In yet another embodiment, R1is phenyl. In still another embodiment, R1is thienyl. In an embodiment, R2is selected from the group consisting of absent, —H, methyl, and C2-alkyl-5-6 membered heterocycloalkyl. In still another embodiment, R2is absent. In an embodiment, R2is —H. In another embodiment, R2is methyl. In yet another embodiment, R2is C2-alkyl-5-6 membered heterocycloalkyl. In still another embodiment, R3is selected from the group consisting of absent, —H, and —C2-alkyl-5-6 membered heterocycloalkyl. In an embodiment, R3is absent. In another embodiment, R3is —H. In yet another embodiment, R3is —C2-alkyl-5-6 membered heterocycloalkyl. In still another embodiment, R4is selected from the group consisting of —H, cyclopropyl, and methyl. In an embodiment, R4is —H. In another embodiment R4is cyclopropyl. In yet another embodiment, R4is methyl. In another embodiment, aryl is a 6-10 membered aromatic ring and heteroaryl is a 5-10 membered heteroaromatic ring. In still another embodiment, the compound of Formula I or Formula II is selected from the group consisting of: TABLE 1CompoundNo.Structure001002003004005006007008009010011012013014015016017018019020021022023 or a pharmaceutically acceptable salt thereof. In yet another aspect, provided herein is a compound of Formula III: or a pharmaceutically acceptable salt thereof; wherein: X and Y are, independently at each occurrence, selected from the group consisting of CH and N, provided at least one of X and Y are N; R1is —N(R4)2or —OR4; R2is selected from the group consisting of halo, aryl, and heteroaryl; wherein aryl or heteroaryl are optionally substituted by one or more R5groups; R3is independently, at each occurrence, selected from the group consisting of halo, cycloalkyl, heterocycloalkyl, —N(R6)—C1-C3alkyl-N(R6)2, —O—C1-C3alkyl-N(R6)2, and —N(R6)—C1-C3alkyl-OR6; wherein cycloalkyl and heterocycloalkyl are optionally substituted by one or more R5groups; R4is independently, at each occurrence, selected from the group consisting of —H, C1-C6alkyl, and C1-C6alkoxy; R5is independently, at each occurrence, —H or C1-C6alkyl; R6is independently, at each occurrence, selected from the group consisting of —H, C1-C6alkyl, and C1-C6alkoxy; andn is 0, 1, 2, or 3. In an embodiment of Formula III, R1is —NH2or —OH; R2is halo or heteroaryl, wherein heteroaryl is optionally substituted by one or more R5; R3is independently, at each occurrence, selected from the group consisting of cycloalkyl, heterocycloalkyl, —N(R6)—C1-C3alkyl-N(R6)2, —O—C1-C3alkyl-N(R6)2, and —N(R6)—C1-C3alkyl-OR6, wherein cycloalkyl and heterocycloalkyl are optionally substituted by one or more R5; R4is independently, at each occurrence, —H or C1-C6alkyl; R5is independently, at each occurrence, —H or C1-C6alkyl; R6is independently, at each occurrence, —H or C1-C6alkyl; and n is 1, 2, or 3. In another embodiment, R1is —NH2or —OH. In yet another embodiment, R1is —NH2. In still another embodiment, R1is —OH. In another embodiment, R2is halo or heteroaryl, wherein heteroaryl is optionally substituted by one or more R5. In an embodiment, R2is halo. In another embodiment, R2is heteroaryl, wherein heteroaryl is optionally substituted by one or more R5. In yet another embodiment, R3is independently, at each occurrence, selected from the group consisting of halo, cycloalkyl, heterocycloalkyl, —N(R6)—C1-C3alkyl-N(R6)2, —O—C1-C3alkyl-N(R6)2, and —N(R6)—C1-C3alkyl-OR6, wherein cycloalkyl and heterocycloalkyl are optionally substituted by one or more R5groups. In still another embodiment, n is 1 and R3is cycloalkyl, wherein cycloalkyl is optionally substituted by one or more R5. In an embodiment, n is 1 and R3is heterocycloalkyl, wherein heterocycloalkyl is optionally substituted by one or more R5. In another embodiment, n is 1, and R3is —N(R6)—C1-C3alkyl-N(R6)2. In yet another embodiment, n is 1, and R3is —O—C1-C3alkyl-N(R6)2. In still another embodiment, n is 1, and R3is —N(R6)—C1-C3alkyl-OR6. In an embodiment, R4is independently, at each occurrence, —H or C1-C6alkyl. In another embodiment, R4is —H. In yet another embodiment, R4is C1-C6alkyl. In still another embodiment, R5is independently, at each occurrence, —H or C1-C6alkyl. In an embodiment, R5is —H. In another embodiment, R5is C1-C6alkyl. In yet another embodiment, R6is independently, at each occurrence, —H or C1-C6alkyl. In still another embodiment, R6is —H. In an embodiment, R6is C1-C6alkyl. In another embodiment, n is 1 or 2. In yet another embodiment, n is 1. In still another embodiment, n is 2. In an embodiment, n is 3. In another embodiment, the compound of Formula III is a compound of Formula IV: or a pharmaceutically acceptable salt thereof. In an embodiment of Formula IV, R3is independently, at each occurrence, selected from the group consisting of cycloalkyl, heterocycloalkyl, —N(R6)—C1-C3alkyl-N(R6)2, —O—C1-C3alkyl-N(R6)2, and —N(R6)—C1-C3alkyl-OR6, wherein cycloalkyl and heterocycloalkyl are optionally substituted by one or more R5. In yet another embodiment, n is 1, and R3is cycloalkyl optionally substituted by one or more R5. In still another embodiment, n is 1, and R3is heterocycloalkyl optionally substituted by one or more R5. In an embodiment, n is 1, and R3is —N(R6)—C1-C3alkyl-N(R6)2. In another embodiment, n is 1, and R3is —O—C1-C3alkyl-N(R6)2. In yet another embodiment, n is 1, and R3is —N(R6)—C1-C3alkyl-OR6. In another embodiment of Formula IV, R6is, independently at each occurrence, —H or C1-C6alkyl. In yet another embodiment, R6is —H. In still another embodiment, R6is C1-C6alkyl. In an embodiment, n is 1, 2, or 3. In another embodiment, n is 1. In yet another embodiment, n is 2. In still another embodiment, n is 3. In an embodiment, the compound of Formula III or Formula IV is selected from the group consisting of: TABLE 2Com -poundNo.Structure024025026027028029030031032 or a pharmaceutically acceptable salt thereof. In an aspect, provided herein are pharmaceutical compositions comprising any of the compounds described herein, or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier. In one embodiment, the disclosed compounds may exist as tautomers. All tautomers are included within the scope of the compounds presented herein. Compounds described herein also include isotopically-labeled compounds wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds described herein include and are not limited to2H,3H,11C,13C,14C,36Cl,18F,123I,125I,13N,15N,15O,17O,18O,32P, and35S. In another embodiment, isotopically-labeled compounds are useful in drug or substrate tissue distribution studies. In another embodiment, substitution with heavier isotopes such as deuterium affords greater metabolic stability (for example, increased in vivo half-life or reduced dosage requirements). In yet another embodiment, the compounds described herein include a2H (i.e., deuterium) isotope. In still another embodiment, substitution with positron emitting isotopes, such as11C,18F,15O and13N, is useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds are prepared by any suitable method or by processes using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed. The specific compounds described herein, and other compounds encompassed by one or more of the formulas described herein having different substituents are synthesized using techniques and materials described herein and as described, for example, in Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989), March, Advanced Organic Chemistry 4thEd., (Wiley 1992); Carey and Sundberg, Advanced Organic Chemistry 4th Ed., Vols. A and B (Plenum 2000, 2001), and Green and Wuts, Protective Groups in Organic Synthesis 3rd Ed., (Wiley 1999) (all of which are incorporated by reference for such disclosure). General methods for the preparation of compounds as described herein are modified by the use of appropriate reagents and conditions, for the introduction of the various moieties found in the Formulas as provided herein. Compounds described herein are synthesized using any suitable procedures starting from compounds that are available from commercial sources, or are prepared using procedures described herein. Methods of Treatment The compounds provided herein can be used in a method of treating a disease or condition in a subject, said method comprising administering to the subject in need thereof a compound provided herein, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound provided herein, or pharmaceutically acceptable salt thereof. In an aspect, provided herein is a method of selectively inhibiting HDAC1 and/or HDAC2 over other HDACs (e.g., HDAC3 and HDAC6) in a subject in need thereof, comprising administering to the subject a compound of Formula I, II, III, IV, or any of the compounds of Table 1 or Table 2, or pharmaceutically acceptable salts thereof. In an embodiment, a compound provided herein has a selectivity for HDAC1 and/or HDAC2 of 5 to 1000 fold over other HDACs. In another embodiment, a compound provided herein has a selectivity for HDAC1 and/or HDAC2 when tested in a HDAC enzyme assay of about 5 to 1000 fold over other HDACs. In certain embodiments, the compound has a selectivity for HDAC1 and/or HDAC2 of 15 to 40 fold over other HDACs. In another aspect, provided herein is a method of treating a disease mediated by HDAC1 and/or HDAC2 in a subject in need thereof, comprising administering to the subject a compound of Formula I, II, III, IV, or any of the compounds of Table 1 or Table 2. In an embodiment, the disease is selected from the group consisting of alopecia, spinal muscular atrophy, inflammatory bowel disease, and Crohn's ulcerative colitis. In an embodiment, the compounds provided herein are able to treat a subject suffering from or susceptible to a hemoglobinopathy. In another embodiment, the compounds are able to treat sickle-cell disease or beta-thalessemia. In another embodiment, the compounds provided herein are useful in the treatment of myelodysplastic syndromes. In an aspect, provided herein is a method of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or pharmaceutically acceptable salt thereof. In still another embodiment, the cancer is lung cancer, colon and rectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, brain cancer, kidney cancer, ovarian cancer, stomach cancer, skin cancer, bone cancer, gastric cancer, breast cancer, glioma, glioblastoma, neuroblastoma, hepatocellular carcinoma, papillary renal carcinoma, head and neck squamous cell carcinoma, leukemia, lymphomas, myelomas, retinoblastoma, cervical cancer, melanoma and/or skin cancer, bladder cancer, uterine cancer, testicular cancer, esophageal cancer, and solid tumors. In some embodiments, the cancer is lung cancer, colon cancer, breast cancer, neuroblastoma, leukemia, or lymphomas. In other embodiments, the cancer is lung cancer, colon cancer, breast cancer, neuroblastoma, leukemia, or lymphoma. In a further embodiment, the cancer is non-small cell lung cancer (NSCLC) or small cell lung cancer. In an embodiment, the cancer is neuroblastoma. In another embodiment, the cancer is a hematologic cancer. In yet another embodiment, the hematologic cancer is leukemia or lymphoma. In still another embodiment, the lymphoma is Hodgkin's lymphoma or Non-Hodgkin's lymphoma. In an embodiment, the leukemia is myeloid, lymphocytic, myelocytic, lymphoblastic, or megakaryotic leukemia. In another embodiment, the leukemia is acute myelogenous leukemia or megakaryocytic leukemia. In another aspect, provided herein is a method of treating or preventing hearing loss in a subject in need thereof comprising administering to a subject a therapeutically effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof. In another aspect, provided herein is a method of treating sickle cell disease, beta thalassemia, myelodysplastic syndrome, acute myelogenous leukemia, neuroblastoma, or megakaryocytic leukemia in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a provided herein, or a pharmaceutically acceptable salt thereof. In yet another aspect, provided herein is a method for the treatment of a disease mediated by HDAC1 and/or HDAC2 in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound as described herein. In an embodiment, the subject is identified as in need of such treatment. In another embodiment, the method of treatment comprises administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutical composition comprising a compound provided herein, in such amounts and for such time as is necessary to achieve the desired result. In yet another embodiment, the method involves the administration of a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, to a subject (including, but not limited to a human or animal) in need of treatment (including a subject identified as in need). Administration/Dosage/Formulations In another aspect, provided herein is a pharmaceutical composition comprising at least one compound disclosed herein, together with a pharmaceutically acceptable carrier. Actual dosage levels of the active ingredients in the pharmaceutical compositions may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. In particular, the selected dosage level will depend upon a variety of factors including the activity of the particular compound employed, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds or materials used in combination with the compound, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well, known in the medical arts. A medical doctor, e.g., physician or veterinarian, having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could begin administration of the pharmaceutical composition to dose the disclosed compound at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In particular embodiments, it is especially advantageous to formulate the compound in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the patients to be treated; each unit containing a predetermined quantity of the disclosed compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle. The dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the disclosed compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding/formulating such a disclosed compound for the treatment of pain, a depressive disorder, or drug addiction in a patient. In one embodiment, the compounds provided herein are formulated using one or more pharmaceutically acceptable excipients or carriers. In one embodiment, the pharmaceutical compositions provided herein comprise a therapeutically effective amount of a disclosed compound and a pharmaceutically acceptable carrier. Routes of administration of any of the compositions discussed herein include oral, nasal, rectal, intravaginal, parenteral, buccal, sublingual or topical. The compounds may be formulated for administration by any suitable route, such as for oral or parenteral, for example, transdermal, transmucosal (e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and perivaginally), (intra)nasal and (trans)rectal), intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, subcutaneous, intramuscular, intradermal, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration. In one embodiment, the preferred route of administration is oral. Suitable compositions and dosage forms include, for example, tablets, capsules, caplets, pills, gel caps, troches, dispersions, suspensions, solutions, syrups, granules, beads, transdermal patches, gels, powders, pellets, magmas, lozenges, creams, pastes, plasters, lotions, discs, suppositories, liquid sprays for nasal or oral administration, dry powder or aerosolized formulations for inhalation, compositions and formulations for intravesical administration and the like. It should be understood that the formulations and compositions are not limited to the particular formulations and compositions that are described herein. For oral application, particularly suitable are tablets, dragees, liquids, drops, suppositories, or capsules, caplets and gel caps. The compositions intended for oral use may be prepared according to any method known in the art and such compositions may contain one or more agents selected from the group consisting of inert, non-toxic pharmaceutically excipients that are suitable for the manufacture of tablets. Such excipients include, for example an inert diluent such as lactose; granulating and disintegrating agents such as cornstarch; binding agents such as starch; and lubricating agents such as magnesium stearate. The tablets may be uncoated or they may be coated by known techniques for elegance or to delay the release of the active ingredients. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert diluent. For parenteral administration, the disclosed compounds may be formulated for injection or infusion, for example, intravenous, intramuscular or subcutaneous injection or infusion, or for administration in a bolus dose or continuous infusion. Suspensions, solutions or emulsions in an oily or aqueous vehicle, optionally containing other formulatory agents such as suspending, stabilizing or dispersing agents may be used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures, embodiments, claims, and examples described herein. Such equivalents were considered to be within the scope of this disclosure and covered by the claims appended hereto. For example, it should be understood, that modifications in reaction conditions, including but not limited to reaction times, reaction size/volume, and experimental reagents, such as solvents, catalysts, pressures, atmospheric conditions, e.g., nitrogen atmosphere, and reducing/oxidizing agents, with art-recognized alternatives and using no more than routine experimentation, are within the scope of the present application. It is to be understood that wherever values and ranges are provided herein, all values and ranges encompassed by these values and ranges, are meant to be encompassed within the scope of the present disclosure. Moreover, all values that fall within these ranges, as well as the upper or lower limits of a range of values, are also contemplated by the present application. The following examples further illustrate aspects of the present disclosure. However, they are in no way a limitation of the teachings of the present disclosure as set forth. EXAMPLES The compounds and methods disclosed herein are further illustrated by the following examples, which should not be construed as further limiting. The practice of the present disclosure will employ, unless otherwise indicated, conventional techniques of organic synthesis, cell biology, cell culture, and molecular biology, which are within the skill of the art. Abbreviations Ac acetyl° C. degree Celsiusdba dibenzylideneacetoneDCM dichloromethaneDIPEA N,N-DiisopropylethylamineDMAP 4-dimethylaminopyridineDMF dimethylformamideDMSO dimethylsulfoxidedppf 1,1′-bis(diphenylphosphino)ferroceneEDCI 1-ethyl-3-[3-(dimethylamino)propyl]carbodiimidhydrochlorideEA ethyl acetateHATU O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium HexafluorophosphateHOAt 1-hydroxy-7-azabenzotriazoleHOBt 1-hydroxybenzotriazoleHPLC high performance liquid chromatographym-CPBA m-chloroperoxybenzoicacidPE petroleum etherPh phenylPy pyridineRuphos 2-Dicyclohexylphosphino-2′,6′-di-i-propoxy-1,1′-biphenylS-Phos 2-Dicyclohexylphosphino-2′,6′-dimethoxybiphenylTFA trifluoroacetic acidTHE tetrahydrofuranTLC thin layer chromatographyTol toluene or tolylXantphos 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene Synthesis Procedures Example 1—Synthesis of Compound 001 Step 1: Methyl-4-(methylamino)benzoate (1.0 g, 6.6 mmol) and K2CO3(2.28 g, 16.53 mmol) were combined in EA and water (v:v=1, 30 mL) and stirred at rt for 20 mins. 2-Chloroacetyl chloride (0.63 mL, 7.94 mmol) was added dropwise. The mixture was stirred at rt for 24 h. The organic layer was collected and dried over Na2SO4. The crude solution was concentrated to afford compound 2 as a white solid (0.9 g, 57%). Step 2: Compound 2 (0.9 g, 3.734 mmol) was dissolved in toluene (10 mL). KOAc (736 mg, 0.75 mmol), xantphos (433 mg, 0.2 eq), and Pd(OAc)2(168 mg, 0.2 eq) were added. The mixture was stirred at 60° C. overnight. After the reaction was complete, the mixture was filtered. The filtrate was concentrated and washed with 10 mL EA. The solid was collected to afford compound 3 (1.0 g, crude). Step 3: Compound 3 (1.0 g, 5 mmol) was stirred in 2M HCl (15 mL) at 100° C. overnight. The mixture was concentrated to get compound 4 (823 mg, 86%) as yellow solid. Step 4: To a solution of compound 4 (95 mg, 0.5 mmol) in Py (5 mL) was added EDCI (191 mg, 1 mmol) followed by amine tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (123 mg, 0.9 eq). The mixture was stirred at rt overnight. The mixture was concentrated to get a residue, which was extracted with EA/water to get compound 5 (250 mg, crude). Step 5: Compound 5 (220 mg, crude) was dissolved in DCM (2 mL). TFA (1 mL) was added, and the reaction mixture was stirred at rt for 1 h. The mixture was concentrated to get compound 001 (38 mg, 20%) as yellow solid. LCMS: m/z=364 (M+H)+ 1H NMR (400 MHz, DMSO) δ 9.65 (s, 1H), 7.97 (d, J=8.2 Hz, 1H), 7.93 (s, 1H), 7.61 (d, J=1.1 Hz, 1H), 7.52 (d, J=1.8 Hz, 1H), 7.33 (dd, J=8.4, 1.9 Hz, 1H), 7.11 (d, J=8.2 Hz, 1H), 6.82 (d, J=8.4 Hz, 1H), 6.61 (d, J=3.2 Hz, 1H), 6.51 (dd, J=3.2, 1.8 Hz, 1H), 5.14 (s, 2H), 3.64 (d, J=8.1 Hz, 2H), 3.17 (d, J=10.2 Hz, 3H). Example 2—Synthesis of Compound 002 Step 1: Methyl 2-oxoindoline-5-carboxylate (382 g, 2 mmol), (bromomethyl)cyclopropane (270 mg, 2 mmol), and K2CO3(276 mg, 2 mmol) was stirred in DMF (5 mL) at rt overnight. The mixture was diluted with EA (300 mL), filtered (flash silica gel), and concentrated to get a residue, which was purified by silica gel to get Compound 2 (294 mg, 60%) as yellow solid. Step 2: Compound 2 (490 mg, 2 mmol) was added to 6M HCl (5 mL) and stirred at 80° C. overnight. The mixture was concentrated to get Compound 3 (462 mg, 100%) as yellow solid. Step 3: Compound 3 (231 mg, 1 mmol), amine tert-butyl 3-aminobiphenyl-4-ylcarbamate (284 mg, 1 mmol), and EDCI (382 mg, 2 mmol) were combined in Py (5 mL) and was stirred at rt overnight. The mixture was concentrated to get a residue, which was purified by silica gel to get Compound 4 (298 mg, 60%) as yellow solid. Step 4: Compound 4 (249 mg, 0.5 mmol) was dissolved in DCM (2 mL). TFA (1 mL) was added and the reaction mixture was stirred at rt for 1 h. The mixture was concentrated to get Compound 002 (119, 80%) as yellow solid. LCMS: m/z=498 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.66 (s, 1H), 8.00 (m, 1H), 7.93 (s, 1H), 7.46 (s, 1H), 7.40 (m, 1H), 7.37 (m, 1H), 7.36 (m, 1H), 7.31 (m, 1H), 7.30 (m 2H), 7.06 (m, 1H), 6.81 (m, 1H), 5.14 (s, 2H). 3.68 (m, 2H), 3.62 (m, 2H), 1.17 (m, 1H), 0.48 (m, 2H), 0.36 (m, 2H) Example 3—Synthesis of Compound 003 Step 1: To a solution of methyl 2-oxoindoline-5-carboxylate (0.95 g, 5 mmol) and 4-(2-chloroethyl)morpholine (1.16 g, 6 mmol) in CH3CN (20 mL) was added K2CO3(1.38 g, 10 mmol) at rt. The reaction mixture was stirred at 75° C. for 5 h. The reaction mixture was cooled and ice water was added. The crude mixture was extracted with EA/water. The EA layer was collected and purified on combiflash with DCM/MeOH (0-10%) to afford Compound 2 as purple solid (0.9 g, 60%). Step 2: Compound 2 (608 mg, 2 mmol) was added into 2M HCl (10 mL), the mixture was heated to 100° C. for 6 h. When the reaction was complete, the mixture was concentrated for next step (570 mg, 98%). Step 3: To a solution of compound 3 (290 mg, 1 mol) and tert-butyl 3-aminobiphenyl-4-ylcarbamate (255 mg, 0.9 mmol) in Py (10 mL) was added EDCI (382 mg, 2 mmol). The mixture was stirred at rt overnight. The reaction mixture was concentrated to get a residue, and extracted with EA/water. The organic layer was separated, dried and purified on combiflash to afford compound 4 (392 mg, 70%) as a purple oil. Step 4: Compound 4 (392 mg, 0.7 mmol) and TFA (1.5 mL) were combined in DCM (5 mL). The reaction was stirred at rt for 0.5 h. The mixture was purified by Prep-HPLC (acid condition) to obtain Compound 003 (80 mg, 25%) as a white solid. LCMS: m/z=463 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.67 (s, 1H), 7.96 (d, J=18.5 Hz, 2H), 7.46 (d, J=2.1 Hz, 1H), 7.36 (dd, J=5.1, 1.1 Hz, 1H), 7.30 (dd, J=8.3, 2.1 Hz, 1H), 7.25 (dd, J=3.6, 1.1 Hz, 1H), 7.19 (s, 1H), 7.06 (dd, J=5.1, 3.6 Hz, 1H), 6.82 (d, J=8.3 Hz, 1H), 5.14 (s, 2H), 3.86 (s, 2H), 3.67 (s, 3H), 3.54 (s, 4H), 3.34 (s, 8H), 5.72-0.52 (m, 41H), 2.48-2.38 (m, 2H). Example 4—Synthesis of Compound 004 Step 1: To a solution of compound 1 (177 mg, 05 mmol) in Py (5 mL) was added EDCI (382 mg, 1 mmol) followed by amine tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (260 mg, 0.9 eq). The mixture was stirred at rt overnight. The reaction was concentrated to get a residue, which was extracted with EA/water to get compound 2 (218 mg, 62%). Step 2: Compound 2 (218 mg, 0.62) and TFA (1 mL) were combined in DCM (2 mL), and the reaction was stirred at rt for 1 h. The mixture was concentrated to get a compound 004 (138 mg, 82%) as yellow solid. LCMS: m/z=350 (M+H).1H NMR (400 MHz, DMSO) δ 9.65 (s, 1H), 8.21 (s, 1H), 7.97 (d, J=8.2 Hz, 1H), 7.93 (s, 1H), 7.61 (d, J=1.1 Hz, 1H), 7.52 (d, J=1.8 Hz, 1H), 7.33 (dd, J=8.4, 1.9 Hz, 1H), 7.11 (d, J=8.2 Hz, 1H), 6.82 (d, J=8.4 Hz, 1H), 6.61 (d, J=3.2 Hz, 1H), 6.51 (dd, J=3.2, 1.8 Hz, 1H), 5.14 (s, 2H), 3.64 (d, J=8.1 Hz, 2H). Example 5—Synthesis of Compound 006 Step 1: Compound 1 (200 mg, 1 mmol), 1-(2-chloroethyl) piperidine (183 mg, 1 mmol), and KOH (112 mg, 2 mmol) were combined in CH3CN (5 mL). The reaction was stirred at 50° C. for 1 h. After completed, the mixture was concentrated and purified by column chromatography (DCM/MeOH=10/1). Compound 2 was afforded as a purple oil (100 mg, 32%). Step 2: Compound 2 (100 mg, 0.33 mmol) was dissolved in HCl (2N in water). The reaction mixture was refluxed for 4 h. The mixture was concentrated for next step (100 mg, crude). Step 3: Compound 3 (100 mg, 0.35 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (81 mg, 0.28 mmol), EDCI (202 mg, 1.05 mmol) were combined in Py (5 mL). The reaction was stirred at rt for 3 h. After completed, the mixture was concentrated and purified by column chromatography (DCM/MeOH=10/1). Colorless oil was afforded as compound 4 (40 mg, 20%). Step 4: Compound 4 (40 mg, 0.07 mmol) was dissolved in DCM (4 ml). Then TFA (2 ml) was added. The mixture was stirred at rt for 2 h. The crude reaction was concentrated and washed by Et2O. Pink solid was afforded to obtain compound 006 (45 mg, TFA salt). LCMS: m/z=461.1 (M+H)+.1H NMR (400 MHz, D2O) δ 7.87 (d, J=8.0 Hz, 1H), 7.80 (s, 1H), 7.56 (d, J=6.7 Hz, 1H), 7.42 (s, 1H), 7.37 (d, J=4.9 Hz, 1H), 7.32 (d, J=3.5 Hz, 1H), 7.22 (d, J=8.6 Hz, 1H), 7.07 (dd, J=12.9, 6.6 Hz, 2H), 4.08 (t, J=6.1 Hz, 2H), 3.58 (t, J=12.4 Hz, 4H), 3.34 (t, J=6.3 Hz, 2H), 2.92 (t, J=11.2 Hz, 2H), 1.88 (d, J=15.0 Hz, 2H), 1.78-1.58 (m, 3H), 1.41 (d, J=12.5 Hz, 1H). Example 6—Synthesis of Compound 007 Step 1: To a solution of tert-butyl 4-bromo-2-nitrophenylcarbamate (634 mg, 2 mmol) and 4-hydroxyphenylboronic acid (276 mg, 2 mmol) in dioxane and water (10 mL:5 ml, v:v=1) was added K2CO3(552 mg, 4 mmol) and Pd(PPh3)4(231 mg, 0.1 eq). The mixture was stirred at 100° C. overnight. The mixture was then extracted with EA/water. The organic layer was separated, dried, and purified on combiflash (PE/EA=3) to afford compound 2 (462 mg, 70%) as a brown solid. Step 2: NaH (85 mg, 2.125 mmol, 60%) was slowly added to a solution of compound 2 (280 mg, 0.85 mol) in dry DMF (10 mL). After the reaction stirred for 5 min, methylsulfamoyl chloride (130 mg, 0.1 eq) was added into the reaction. The mixture was stirred at rt for overnight. The mixture was extracted with EA/water. The organic layer was separated, dried and concentrated to afford compound 3 (350 mg, crude) as a yellow oil. Step 3: To a solution of compound 3 (350 mg, crude) in EtOH (10 mL) was added carbol (175 mg, 50% amount) and FeCl3(14 mg, 0.1 eq) at 60° C. NH2—NH2(1 mL) was added dropwise, and the mixture was stirred at 60° C. for 30 min. After the reaction had completed, the mixture was filtered. The filtrate was collected and concentrated, washed with water, and the solid was dried to afford the amine 4 (280 mg, 80%). Step 4: 1-Methyl-2-oxoindoline-5-carboxylic acid (136 mg, 0.7 mmol) was dissolved in Py (10 mL). EDCI (286 mg, 1.5 mmol) was added, followed by amine 4 (280 mg, crude). The mixture was stirred at rt overnight. The reaction mixture was concentrated, diluted with water, and the precipitate was collected to afford the crude solid 5 (300 mg, crude). Step 5: The crude material of compound 5 (100 mg, crude) and TFA (1 mL) were combined in DCM (2 mL). The reaction was stirred at rt for 1 h. The mixture was concentrated and purified on prep-HPLC (acid condition) to get a compound 007 (17 mg, 5.4%, 4 steps) as yellow solid. LCMS: m/z=374 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.65 (s, 1H), 8.00 (d, J=8.1 Hz, 1H), 7.92 (s, 1H), 7.40 (d, J=2.1 Hz, 1H), 7.36 (d, J=8.6 Hz, 2H), 7.21 (dd, J=8.3, 2.2 Hz, 1H), 7.11 (d, J=8.2 Hz, 1H), 6.83 (d, J=8.3 Hz, 1H), 6.78 (d, J=8.6 Hz, 2H), 4.94 (s, 2H), 3.65 (s, 2H), 3.18 (s, 3H). Example 7—Synthesis of Compound 009 Step 1: Compound 1 (830 mg, 5 mmol) and cyclopropanecarboxylic acid (430 mg, 5 mmol) were combined in 4M HCl (20 mL). The reaction was refluxed overnight. After completed, the mixture was concentrated and purified by Prep-HPLC. White solid was afforded as Compound 2 (312 mg, 31%). Step 2: Compound 2 (100 mg, 0.50 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (144 mg, 0.50 mmol), and EDCI (288 mg, 1.50 mmol) were combined in Py (5 mL). The reaction was stirred at rt overnight. After completed, the mixture was concentrated and purified by column chromatography (DCM/MeOH=10/1). Compound 3 was isolated as a colorless oil (54 mg, 23%). Step 3: Compound 3 (54 mg, 0.11 mmol) was dissolved in DCM (2 ml). Then TFA (1 ml) was added. The mixture was stirred at rt for 2 h. The crude material was concentrated and washed with Et2O. Compound 009 was isolated as a white solid (30 mg, TFA salt). LCMS: m/z=375.0 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.96 (s, 1H), 8.26 (s, 1H), 8.07 (d, J=8.0 Hz, 1H), 7.76 (d, J=8.5 Hz, 1H), 7.50 (s, 1H), 7.35 (dd, J=8.5, 2.5 Hz, 2H), 7.28 (d, J=3.0 Hz, 1H), 7.07 (t, J=3.5 Hz, 1H), 6.88 (s, 1H), 2.42 (t, J=3.0 Hz, 1H), 1.37 (dd, J=2.5 Hz, 4H). Example 8—Synthesis of Compound 010 Step 1: Compound 1 (50 mg, 0.28 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (66 mg, 0.23 mmol), and EDCI (164 mg, 0.85 mmol) were combined in Py (3 mL). The reaction was stirred at rt overnight. After completed, the mixture was concentrated and washed with water and ether. The white solid was afforded as Compound 2 (100 mg, 72%). Step 2: Compound 2 (100 mg, 0.22 mmol) and TFA (2 mL) were combined in DCM (4 mL) and stirred at rt for 2 h. The mixture was concentrated and purified by Prep-HPLC (base method). White solid was afforded to obtain compound 010 (56 mg, 72%). LCMS: m/z=349.1 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.80 (s, 1H), 8.36 (s, 1H), 8.31 (d, J=1.1 Hz, 1H), 7.90 (dd, J=8.5, 1.6 Hz, 1H), 7.74 (d, J=8.5 Hz, 1H), 7.51 (d, J=2.1 Hz, 1H), 7.37 (dd, J=5.1, 1.0 Hz, 1H), 7.32 (dd, J=8.3, 2.2 Hz, 1H), 7.26 (dd, J=3.6, 1.1 Hz, 1H), 7.06 (dd, J=5.1, 3.6 Hz, 1H), 6.84 (d, J=8.3 Hz, 1H), 5.18 (s, 2H), 3.93 (s, 3H). Example 9—Synthesis of Compound 011 Step 1: Compound 3 (231 mg, 1 mmol), amine tert-butyl 2-amino-4-(thiophen-2 yl)phenylcarbamate (290 mg, 1 mmol), and EDCI (382 mg, 2 mmol) were combined in Py (5 mL). The reaction was stirred at rt overnight. The mixture was concentrated to get a residue, which was purified by silica gel to get compound 4 (302 mg, 60%) as yellow solid. Step 2: Compound 4 (252 mg, 0.5 mmol) and TFA (1 mL) were combined in DCM (2 mL). The reaction was stirred at rt for 1 h. The mixture was concentrated to get a compound 011 (161, 80%) as yellow solid. LCMS: m/z=404 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.66 (s, 1H), 8.00 (m, 1H), 7.93 (s, 1H), 7.46 (s, 1H), 7.36 (m, 1H), 7.31 (m, 1H), 7.30 (m 2H), 7.06 (m, 1H), 6.81 (m, 1H), 5.14 (s, 2H). 3.68 (m, 2H), 3.62 (m, 2H), 1.17 (m, 1H), 0.48 (m, 2H), 0.36 (m, 2H) Example 10—Synthesis of Compound 012 Step 1: To a solution of compound 4 (95 mg, 05 mmol) in Py (5 mL) was added EDCI (191 mg, 1 mmol) followed by amine tert-butyl 3-aminobiphenyl-4-ylcarbamate (120 mg, 0.9 eq). The mixture was stirred at rt for overnight. The mixture was concentrated to get a residue, which was extracted with EA/water to get compound 5 (220 mg, crude). Step 2: Compound 5 (220 mg, crude) and TFA (1 mL) were combined in DCM (2 mL). The reaction was stirred at rt for 1 h. The mixture was concentrated to get a Compound 012 (25, 16%) as yellow solid. LCMS: m/z=368 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.65 (s, 1H), 7.61 (d, J=1.1 Hz, 1H), 7.52-7.41 (m, 5H), 7.33 (dd, J=8.4, 1.9 Hz, 1H), 7.11 (d, J=8.2 Hz, 1H), 6.82 (d, J=8.4 Hz, 1H), 6.61 (d, J=3.2 Hz, 1H), 6.51 (dd, J=3.2, 1.8 Hz, 1H), 5.14 (s, 2H), 3.64 (d, J=8.1 Hz, 2H), 3.17 (d, J=10.2 Hz, 3H). Example 11—Synthesis of Compound 013 Step 1: To a solution of compound 3 (290 mg, 1 mmol) and tert-butyl 3-aminobiphenyl-4-ylcarbamate (255 mg, 0.9 mmol) in Py (10 mL) was added EDCI (382 mg, 2 mmol). The mixture was stirred at rt overnight. The mixture was concentrated to get a residue and extracted with EA/water. The organic layer was separated, dried, and purified on combiflash to afford compound 4 (376 mg, 68%) as a purple oil. Step 2: Compound 4 (376 mg, 0.68 mmol) and TFA (1.5 mL) were combined in DCM (5 mL). The reaction was stirred at rt for 0.5 h. The mixture was purified by Prep-HPLC (acid condition) to obtain compound 013 (100 mg, 32%) as a white solid. LCMS: m/z=457 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.67 (s, 1H), 7.56-7.49 (m, 4H), 7.47 (d, J=8, 1H), 7.36 (dd, J=5.1, 1.1 Hz, 1H), 7.30 (dd, J=8.3, 2.1 Hz, 1H), 7.26 (dd, J=3.6, 1.1 Hz, 1H), 7.20 (s, 1H), 7.06 (dd, J=5.1, 3.6 Hz, 1H), 6.82 (d, J=8.3 Hz, 1H), 5.14 (s, 2H), 3.86 (s, 2H), 3.67 (s, 3H), 3.54 (s, 4H), 3.34 (s, 8H), 5.72-0.52 (m, 41H), 2.48-2.38 (m, 2H). Example 12—Synthesis of Compound 014 Step 1: Compound 1 (177 g, 1 mmol), iodomethane (426 mg, 3 mmol), and NaH (60%, 160 mg, 4 mmol) were combined in THE (5 mL). The reaction mixture was stirred at 60° C. for 3 h. The mixture was concentrated to get a residue The crude residue was dissolved in Py (5 mL). tert-Butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (290 mg, 1 mmol) and EDCI (382 mg, 2 mmol) were added and the reaction was stirred at rt overnight. The mixture was concentrated to get a residue, which was purified by silica gel to get compound 2 (246 mg, 50%, 2 steps) as yellow solid. Step 2: Compound 2 (246 mg, 0.5 mmol) and TFA (1 mL) were combined in DCM (2 mL), and the reaction was stirred at rt overnight. The mixture was concentrated to get compound 014 (156 mg, 80%) as yellow solid. LCMS: m/z=492 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.88 (s, 1H), 8.02 (d, J=8.0 Hz, 2H), 7.53 (s, 1H), 7.42 (t, J=7.5 Hz, 2H), 7.34 (d, J=3.4 Hz, 1H), 7.17 (d, J=8.2 Hz, 1H), 7.09 (d, J=4.3 Hz, 1H), 6.99 (d, J=8.3 Hz, 1H), 3.21 (s, 3H), 1.34 (s, 6H). Example 13—Synthesis of Compound 015 Step 1: To a solution of compound 1 (95 mg, 05 mmol) in Py (5 mL) was added EDCI (191 mg, 1 mmol) followed by amine tert-butyl 2-amino-4-(furan-2-yl)phenylcarbamate (123 mg, 0.9 eq). The mixture was stirred at rt overnight. The mixture was concentrated to get a residue, which was extracted with EA/water to get compound 2 (226 mg, crude). Step 2: Compound 2 (225 mg, crude) and TFA (1 mL) were combined in DCM (2 mL) and the reaction was stirred at rt for 1 h. The mixture was concentrated to get a compound 015 (20 mg, 13%, 2 steps) as yellow solid. LCMS: m/z=348 (M+H).1H NMR (400 MHz, DMSO) δ 9.65 (s, 1H), 8.01 (d, J=8.2 Hz, 1H), 7.93 (s, 1H), 7.61 (d, J=1.1 Hz, 1H), 7.52 (d, J=1.8 Hz, 1H), 7.33 (dd, J=8.4, 1.9 Hz, 1H), 7.11 (d, J=8.2 Hz, 1H), 6.82 (d, J=8.4 Hz, 1H), 6.61 (d, J=3.2 Hz, 1H), 6.51 (dd, J=3.2, 1.8 Hz, 1H), 5.14 (s, 2H), 3.64 (d, J=8.1 Hz, 2H), 3.17 (d, J=10.2 Hz, 3H). Example 14—Synthesis of Compound 016 Step 1: Compound 1 (191 mg, 1 mmol), 4-(3-chloropropyl) morpholine (163 mg, 1 mmol), and K2CO3(276 mg, 2 mmol) were combined in CH3CN (5 mL), and the reaction was stirred at 80° C. for 2 h. After completed, the mixture was concentrated and purified by column chromatography (DCM/MeOH=10/1). Purple oil was afforded as Compound 2 (260 mg, crude). Step 2: Compound 2 (200 mg, crude) was dissolved in HCl (2N in water). The reaction was stirred at 100° C. for 3 h. The mixture was concentrated for next step (200 mg, crude). Step 3: Compound 3 (200 mg, 0.66 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (154 mg, 0.53 mmol), and EDCI (380 mg, 1.98 mmol) were dissolved in Py (5 mL). The reaction was stirred at rt overnight. After completed, the mixture was concentrated and purified by column chromatography (DCM/MeOH=10/1). Colorless oil was afforded as Compound 4 (290 mg, 77%). Step 4: Compound 4 (290 mg, 0.50 mmol) was dissolved in DCM (4 ml). Then TFA (2 ml) was added. The mixture was stirred at rt for 2 h. The reaction was concentrated and washed with Et2O. Pink solid was afforded to obtain compound 016 (200 mg, TFA salt). LCMS: m/z=477.1 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.77 (s, 2H), 8.03 (d, J=8.9 Hz, 1H), 7.97 (s, 1H), 7.49 (d, J=1.9 Hz, 1H), 7.39 (d, J=4.3 Hz, 1H), 7.34 (dd, J=8.3, 2.1 Hz, 1H), 7.28 (d, J=2.6 Hz, 1H), 7.22 (d, J=8.3 Hz, 1H), 7.07 (dd, J=5.0, 3.6 Hz, 1H), 6.89 (d, J=8.3 Hz, 1H), 3.97 (d, J=12.2 Hz, 2H), 3.81 (t, J=6.5 Hz, 2H), 3.68 (s, 2H), 3.61 (d, J=11.5 Hz, 2H), 3.40 (t, J=12.1 Hz, 2H), 3.19 (s, 2H), 3.05 (s, 2H), 2.00 (d, J=9.5 Hz, 2H). Example 15—Synthesis of Compound 018 Step 1: Compound 1 (100 mg, 0.62 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (151 mg, 0.52 mmol), HOAt (141 mg, 1.04 mmol), EDCI (200 mg, 1.04 mmol), and Et3N (0.2 mL) were combined in THE (5 mL), and the reaction was stirred at rt overnight. After completed, the mixture was concentrated and the crude was carried over for next step (300 mg, crude). Step 2: The crude Compound 2 (300 mg, crude) and TFA (3 mL) were combined in DCM (6 mL). The reaction was stirred at rt for 2 h. The mixture was purified by Prep-HPLC (base method). White solid was afforded to obtain compound 018 (130 mg, 43%, 2 steps). LCMS: m/z=334.8 (M+H)+.1H NMR (400 MHz, DMSO) δ 9.77 (s, 1H), 8.39 (d, J=33.4 Hz, 2H), 7.90 (d, J=7.6 Hz, 1H), 7.71 (s, 1H), 7.51 (s, 1H), 7.39-7.21 (m, 3H), 7.07 (dd, J=13.2, 9.0 Hz, 1H), 6.83 (d, J=8.2 Hz, 1H). Example 16—Synthesis of Compound 019 Step 1: Compound 1 (100 mg, 0.57 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (139 mg, 0.48 mmol), and EDCI (276 mg, 1.44 mmol) were combined in Py (5 mL), and the reaction was stirred at rt overnight. After completed, the mixture was concentrated and washed with ether (200 mg, 78%). Step 2: Compound 2 (200 mg, 0.446 mmol) and TFA (2 mL) were combined in DCM (4 mL). The reaction was stirred at rt for 2 h. The mixture was concentrated and washed with Et2O. White solid was afforded to obtain compound 019 (270 mg, TFA salt). LCMS: m/z=349.1 (M+H).1H NMR (400 MHz, DMSO) δ 10.03 (s, 1H), 9.06 (s, 1H), 8.46 (s, 1H), 8.14 (d, J=8.2 Hz, 1H), 7.93 (d, J=8.6 Hz, 1H), 7.55 (s, 1H), 7.39 (dd, J=14.1, 3.5 Hz, 2H), 7.31 (d, J=3.2 Hz, 1H), 7.13-7.04 (m, 1H), 6.93 (d, J=8.3 Hz, 1H), 4.02 (s, 3H). Example 17—Synthesis of Compound 020 Step 1: 2-Methyl-3H-benzo[d]imidazole-5-carboxylic acid (176 mg, 1 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (290 mg, 1 mmol) and EDCI (382 mg, 2 mmol) were combined in Py (5 mL), and the reaction was stirred at rt overnight. The mixture was concentrated to get a residue, which was purified by silica gel to get compound 2 (314 mg, 70%) as yellow solid. Step 2: Compound 2 (224 mg, 0.5 mmol) and TFA (1 mL) were combined in DCM (2 mL), and the reaction was stirred at rt for 1 h. The mixture was concentrated to get a compound 020 (170 mg, 100%) as yellow solid. LCMS: m/z=349 (M+H)+.1H NMR (400 MHz, DMSO) δ 10.03 (s, 1H), 8.39 (s, 1H), 8.14 (d, J=8.5 Hz, 1H), 7.88 (d, J=8.6 Hz, 1H), 7.51 (s, 1H), 7.43-7.34 (m, 2H), 7.29 (d, J=3.5 Hz, 1H), 7.10-7.04 (m, 1H), 6.89 (d, J=8.4 Hz, 1H), 2.82 (s, 3H). Examples 18 and 19—Synthesis of Compounds 021 and 022 Step 1: Compound 1 (352 g, 2 mmol), 4-(2-chloroethyl)morpholine (300 mg, 2 mmol), and K2CO3(276 mg, 2 mmol) were combined in DMF (5 mL). The reaction was stirred at rt overnight. The mixture was diluted with EA (300 mL), filtered (flash silica gel), and concentrated to get a residue, which was purified by silica gel to get the mixture of 2a and 2b (231 mg, 40%) as yellow solid. Step 2: A mixture of compound 2a and 2b (289 mg, 1 mmol) and LiOH—H2O (42 mg, 1 mmol) were combined in MeOH (2 mL), and was stirred at 60° C. overnight. The mixture was concentrated. The crude material was combined with tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (290 mg, 1 mmol) and EDCI (382 mg, 2 mmol) in Py (5 mL), and was stirred at rt overnight. The mixture was concentrated to get a residue, which was purified by silica gel to get the mixture of 4a and 4b (383 mg, 70%) as yellow solid. Step 3: A mixture of compound 4a and 4b (274 mg, 0.5 mmol) and TFA (1 mL) were combined in DCM (2 mL) stirred at rt for 1 h. The mixture was concentrated to get a residue, which was purified by chiral HPLC to compound 021 (88 mg, 40%) as yellow solid. LCMS: m/z=442 (M+H)+.1H NMR (500 MHz, DMSO) δ 9.79 (s, 1H), 8.39 (s, 1H), 8.35 (s, 1H), 7.93 (m, 1H), 7.95-7.94 (m, 1H), 7.58-7.56 (m, 3H), 7.41-7.38 (m, 2H), 7.35-7.32 (m, 1H), 7.25 (m, 1H), 6.89-6.88 (m, 1H), 5.120 (s, 2H), 4.43-4.41 (m, 2H), 3.53-3.51 (m, 4H), 2.72-2.71 (m, 2H), 2.46 (m, 4H). Compound 022 (88 mg, 40%). LCMS: m/z=442 (M+H)+.1H NMR (500 MHz, DMSO) δ 1H NMR (500 MHz, DMSO) δ 9.76 (s, 1H), 8.40 (s, 1H), 8.367 (s, 1H), 7.93 (m, 1H), 7.95-7.94 (m, 1H), 7.58-7.56 (m, 3H), 7.41-7.38 (m, 2H), 7.35-7.32 (m, 1H), 7.25 (m, 1H), 6.89-6.88 (m, 1H), 5.120 (s, 2H), 4.43-4.41 (m, 2H), 3.53-3.51 (m, 4H), 2.72-2.71 (m, 2H), 2.46 (m, 4H). Example 20—Synthesis of Compound 024 Step 1: Under N2, ethyl 7-bromoisoquinoline-3-carboxylate (589 mg, 2.10 mmol) and N1,N1-dimethylethane-1,2-diamine (742 mg, 8.42 mmol), tris(dibenzylideneacetone) dipalladium (48 mg, 0.052 mmol), xantphos (60 mg, 0.10 mmol) and Cs2CO3(1.37 g, 4.19 mmol) were combined in toluene (20 mL). The reaction mixture was heated at 100° C. for 18 h. The reaction was cooled to rt. and filtered. The filtrate was concentrated in vacuo, and the residue was purified by silica gel chromatography to give compound 2 as a yellow solid (486 mg, 74%). Step 2: Ethyl 7-(2-(dimethylamino)ethylamino)isoquinoline-3-carboxylate (486 mg, 1.69 mmol) and LiOH (213 mg, 5.07 mmol) were combined in THE (12.8 mL), water (4.2 mL) and MeOH (2.1 mL). The reaction was stirred at rt until the reaction was complete. MeOH and THE were then removed in vacuo. The aqueous phase was adjusted to pH 4. Then it was concentrated in vacuo to give crude 3 (assay: 39.5%). Step 3: 7-(2-(Dimethylamino)ethylamino)isoquinoline-3-carboxylic acid (993 mg, crude, 392 mg, 1.51 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (439 mg, 1.51 mmol), and EDCI (580 mg, 3.02 mmol) were dissolved in pyridine (6 mL). The reaction was stirred at rt for 18 h. The reaction was concentrated in vacuo, and the residue was purified by reverse phase chromatography to give compound 4 (711 mg, 79%). Step 4: tert-Butyl-2-(7-(2-(dimethylamino)ethylamino)isoquinoline-3-carboxamido)-4-(thiophen-2-yl)phenylcarbamate (711 mg) was dissolved in DCM (4 mL) TFA (4 mL) was added at 0° C. The reaction was stirred at 0° C. to rt for 2 h. The reaction mixture was concentrated in vacuo, and the residue was purified by prep-HPLC to give product 024 as an off-white solid (386.4 mg, 67%). LCMS: m/z=432.1 (M+H)+.1H-NMR (400 MHz, DMSO) δ 9.84 (s, 1H), 9.21 (s, 1H), 8.74 (s, 1H), 7.88 (d, J=8.5, 1H), 7.53 (s, 1H), 7.38 (d, J=8.0, 2H), 7.32 (d, J=8.5, 1H), 7.26 (s, 1H), 7.06 (s, 2H), 6.84 (d, J=8.5, 1H), 5.13 (s, 2H), 3.27 (t, 2H), 2.52 (t, 2H), 2.17 (s, 6H). Example 21—Synthesis of Compound 025 Step 1: Under N2, ethyl 7-bromoquinoline-3-carboxylate (1.00 g, 3.57 mmol), N,N-dimethylethane-1,2-diamine (346 mg, 3.93 mmol), tris(dibenzylideneacetone) dipalladium (82 mg, 0.089 mmol), Ruphos (83 mg, 0.18 mmol) and Cs2CO3(2.33 g, 7.14 mmol) were combined in toluene (40 mL). The reaction was heated at 100° C. and stirred for 18 h. The reaction was then was cooled to rt. and filtered. The filtrate was concentrated in vacuo, and the residue was purified by reverse phase chromatography to give compound 2 as an oil (665 mg, 65%). Step 2: Ethyl 7-(2-(dimethylamino)ethylamino)quinoline-3-carboxylate (665 mg, 2.31 mmol) and LiOH (292 mg, 6.94 mmol) were combined in THE (17.6 mL), water (5.8 mL) and MeOH (2.9 mL). The reaction was stirred at rt until completed. MeOH and THE were then removed in vacuo. The aqueous phase was adjusted to pH 7 and concentrated in vacuo to give crude 3. Step 3: 7-(2-(Dimethylamino)ethylamino)quinoline-3-carboxylic acid (20 mg, 0.077 mmol), EDCI (18 mg, 0.093 mmol), and HOAT (12 mg, 0.085 mmol) were combined in DIPEA (25.5 mL, 0.154 mmol). The reaction was stirred at rt for 10 min, then tert-butyl 2-amino-4-(thiophen-2-yl)phenyl carbamate (22 mg, 0.077 mmol) was added. The reaction was heated to 50° C. and stirred for 16 h. The crude material was purified by reverse phase column chromatography to give compound 4. Step 4: tert-Butyl 2-(7-(2-(dimethylamino)ethylamino)quinoline-3-carboxamido)-4-(thiophen-2-yl)phenylcarbamate (377 mg, 0.71 mmol) was dissolved in DCM (4 mL). TFA (2 mL) was added at 0° C. The reaction mixture was stirred at 0° C. to rt. for 2 h. The crude reaction was concentrated in vacuo, and the residue was purified by prep-HPLC to give product 025 as a yellow solid (111 mg, 36%). LCMS: m/z=432.1 (M+H)+.1H-NMR (400 MHz, DMSO) δ 9.84 (s, 1H), 9.21 (s, 1H), 8.74 (s, 1H), 7.88 (d, J=8.5, 1H), 7.53 (s, 1H), 7.38 (d, J=8.0, 2H), 7.32 (d, J=8.5, 1H), 7.26 (s, 1H), 7.06 (s, 2H), 6.84 (d, J=8.5, 1H), 5.22 (s, 2H), 3.28 (t, 2H), 2.54 (t, 2H), 2.23 (s, 6H). Example 22—Synthesis of Compound 026 Step 1: 3-(Methylamino)propan-1-ol (8.9 g, 100 mmol), Boc2O (21.8 g, 100 mmol), and Et3N (10.1 g, 100 mmol) were combined in DCM (200 mL). The reaction was stirred at rt overnight. The reaction mixture was diluted with EA (300 mL) and H2O (300 mL), and stirred for 1 h. The organic layer was separated, dried, and concentrated in vacuo to get compound 2 (18 g, 94%) as yellow oil. Step 2: Compound 2 (1.89 g, 10 mmol), ethyl 7-bromoquinoline-3-carboxylate (1.40 g, 5 mmol), Pd2(dba)3(458 mg, 0.5 mmol), Brettphos (268 mg, 0.5 mmol) and Cs2CO3(4.87 g, 15 mmol) were combined in toluene (35 mL). The reaction was stirred at 85° C. under N2atmosphere overnight. The reaction mixture was diluted with EA (50 mL) and H2O (50 mL), and stirred for 1 h. The organic layer was separated, dried, and concentrated in vacuo to get a residue, which was purified by silica gel column chromatography to get compound 3 (1.16 g, 60%) as yellow solid. Step 3: Compound 3 (3.88 g, 10 mmol) and LiOH (840 mg, 20 mmol) were combined in EtOH (10 mL). The reaction was stirred at rt overnight. HCl (2M, 40 mL) was added to the crude reaction mixture. The resulting mixture was filtered to get compound 4 (3.50 g, 97%) as yellow solid. Step 4: Compound 4 (360 mg, 1 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenylcarbamate (290 mg, 1 mmol) and EDCI (573 mg, 3 mmol) were combined in Py (10 mL). The reaction was stirred at rt overnight. The reaction was diluted EA (20 mL) and H2O (20 mL), and stirred for 1 h. The organic layer was separated, dried, and concentrated in vacuo to get a residue, which was purified by silica gel column chromatography to get compound 5 (506 mg, 80%) as yellow solid. Step 5: Compound 5 (316 mg, 0.5 mmol) and TFA (2 mL) were combined in DCM (5 mL). The reaction was stirred at rt for 1 h. The mixture was concentrated to get 026 (200 mg, 92%) as yellow solid. LCMS: m/z=433 (M+H)+.1H NMR (400 MHz, DMSO) δ 11.212 (s, 1H), 9.688 (s, 1H), 9.525 (s, 1H), 9.129 (s, 2H), 8.312-8.289 (d, J=9.2, 1H), 7.844 (S, 1H), 7.712 (s, 1H), 7.635-7.500 (m, 3H), 7.493-7.408 (m, 1H), 7.388-7.356 (m, 1H), 7.156-7.140 (m, 1H), 4.395-4.365 (m, 2H), 3.129-3.100 (m, 2H), 2.596-2.569 (m, 2H), 2.253-2.219 (m, 2H). Example 23—Synthesis of Compound 027 Step 1: 7-(4-Methylpiperazin-1-yl)quinoline-3-carboxylic acid (200 mg, 0.74 mmol) and EDCI (248 mg, 1.30 mmol) were dissolved in pyridine (10 mL). The reaction was stirred at rt for 20 min. tert-Butyl 2-amino-4-(1-methyl-1H-imidazol-2-yl)phenylcarbamate (230 mg, 0.80 mmol) was then added. The reaction was stirred at rt for 18 h, then concentrated in vacuo. The residue was purified by prep-TLC to give compound 3. Step 2: tert-Butyl 4-(1-methyl-1H-imidazol-2-yl)-2-(7-(4-methylpiperazin-1-yl)quinoline-3-carboxamido)phenylcarbamate (0.74 mmol) was dissolved in water and MeOH. HCl/dioxane (1 mL) was added at 0° C. The reaction was stirred at 0° C. to rt for 18 h, then concentrated in vacuo, and the residue was purified by prep-HPLC to give product 027 as a grey solid (30 mg). LCMS: m/z=442.2 (M+H)+.1H-NMR (400 MHz, DMSO) δ 9.88 (s, 1H), 9.24 (s, 1H), 8.77 (s, 1H), 7.91 (d, J=8.5, 1H), 7.57 (d, J=8.0, 1H), 7.55 (s, 1H), 7.28-7.32 (m, 2H), 7.15 (s, 1H), 6.88 (s, 1H), 6.85 (d, J=8.4, 1H), 5.31 (s, 2H), 3.71 (s, 3H), 3.42 (t, 4H), 2.25 (s, 3H). Example 24—Synthesis of Compound 028 Step 1: To a solution of 4-bromo-2-nitrophenol (500 mg, 2.3 mmol) in DCM (20 mL) was added Et3N (464 mg, 4.6 mmol) and DMAP (28 mg, 0.1 mmol, cat.) followed by acetic anhydride (258 mg, 2.5 mmol). The mixture was stirred at rt overnight. After the reaction was complete, the mixture was purified by silica gel chromatography (petroleum ether/EA=3:1) to afford the compound 2 as yellow solid (576 mg, 96%) Step 2: Under N2, 4-bromo-2-nitrophenyl acetate (1.8 g, 6.9 mmol) was dissolved in t-BuOH (30 mL). Thiophen-2-ylboronic acid (876 mg, 6.9 mmol), Pd2(dba)3(631 mg, 0.69 mmol), Xphos (328 mg, 0.69 mmol) and K3PO4(2.9 g, 14 mmol) were added. The reaction was stirred at 130° C. under microwave for 30 min. The residue was purified by column chromatography on silica gel (200-300 mesh, eluting with petroleum ether/EtOAc=3:1) to give compound 3 (485 mg, 26%). Step 3: To a solution of 2-nitro-4-(thiophen-2-yl)phenyl acetate (500 mg, 1.9 mmol) in EtOH (15 mL) was added Pd/C (50 mg, 10% w/w), AcOH (11 mg, 0.19 mmol, cat.). The mixture was stirred under H2at 60° C. for 2 h. After completed, the mixture was filtered to afford the desired amine 4 (430 mg, 97%) Step 4: A mixture of 2-(4-(tert-butoxycarbonyl)piperazin-1-yl)quinoline-6-carboxylic acid (357 mg, 1 mmol), compound 4 (321 mg, 0.9 mmol) and EDCI (382 mg, 2 mmol) were dissolved in pyridine (8 mL). The reaction solution was stirred at rt for 18 h, then concentrated in vacuo. The crude residue was washed with water and dried in vacuo. The residue was purified by prep-TLC to give product 5 (460 mg, crude) as a yellow oil. Step 5: To a solution of crude 5 (460 mg, crude) in MeOH (10 mL) was added ammonia (2 mL). The mixture was stirred at 40° C. for 2 h. After completed, the mixture was purified by prep-TLC to afford the desired product 6 (127 mg, 22%, 2 steps). Step 6: At 0° C., to a mixture of tert-butyl 4-(6-((2-hydroxy-5-(thiophen-2-yl)phenyl)carbamoyl)-quinolin-2-yl)piperazine-1-carboxylate (127 mg) in DCM (8 mL) was added TFA (2 mL). The reaction was stirred at 0° C. to rt for 2 h, then concentrated in vacuo. The crude residue was purified by prep-HPLC to give 028 as a white solid (12.5 mg, FA salt). LCMS: m/z=431.2 (M+H)+. 1H NMR (400 MHz, DMSO) δ 10.62 (s, 1H), 9.21 (s, 1H), 8.82 (d, J=2.0 Hz, 1H), 8.56 (s, 1H), 8.11 (d, J=9.2 Hz, 1H), 7.79 (dd, J=9.4, 2.6 Hz, 1H), 7.53 (brs, 1H), 7.47 (dd, J=5.0, 1.0 Hz, 1H), 7.32 (dd, J=3.6, 1.2 Hz, 1H), 7.29 (dd, J=8.4, 2.4 Hz, 1H), 7.11 (dd, J=5.2, 3.6 Hz, 1H), 7.00 (d, J=8.4 Hz, 1H), 3.49 (s, 4H), 3.12 (s, 4H). Example 25—Synthesis of Compound 029 Step 1: Under N2, ethyl 7-bromoquinoline-3-carboxylate (500 mg, 1.78 mmol), 2-methoxy-ethanamine (134 mg, 1.78 mmol), tris(dibenzylideneacetone) dipalladium (41 mg, 0.045 mmol), Ruphos (42 mg, 0.089 mmol) and Cs2CO3(1.16 g, 3.57 mmol) were combined in toluene (15 mL). The reaction was heated to 100° C. and stirred for 18 h. The reaction was then cooled to rt. and filtered. The filtrate was concentrated in vacuo, and the residue was purified by prep-TLC to give compound 2 (288 mg, 47%). Step 2: Ethyl 7-(2-methoxyethylamino)quinoline-3-carboxylate (288 mg, 1.05 mmol) and LiOH (133 mg, 3.15 mmol) were combined in THE (8 mL), water (2.6 mL) and MeOH (1.3 mL). The reaction was stirred at rt until the reaction was complete. MeOH and THE were then removed in vacuo, and the aqueous phase was adjusted to pH 7. The aqueous phase was then concentrated in vacuo to give crude 3. Step 3: 7-(2-Methoxyethylamino)quinoline-3-carboxylic acid (240 mg, 0.97 mmol), tert-butyl 2-amino-4-(thiophen-2-yl)phenyl carbamate (160 mg, 0.55 mmol) and EDCI (380 mg, 1.98 mmol) were combined in pyridine (12 mL). The residue was washed with water, purified by column chromatography on silica gel (200-300 mesh, eluting with petroleum ether/EtOAc=1:2) to give compound 4 as a yellow solid (300 mg, yield: 67%). Step 4: tert-Butyl 2-(7-(2-methoxyethylamino)quinoline-3-carboxamido)-4-(thiophen-2-yl)phenylcarbamate (300 mg, 0.58 mmol) was dissolved in DCM (4 mL). TFA (3 mL) was added at 0° C. and the reaction stirred at 0° C. to rt for 2 h. The reaction was concentrated in vacuo and the residue was purified by prep-HPLC to give product 029 as a yellow solid (120.4 mg, 50%). LCMS: m/z=419.1 (M+H)+.1H-NMR (400 MHz, DMSO) δ 9.84 (s, 1H), 9.21 (s, 1H), 8.74 (s, 1H), 7.88 (d, J=8.5, 1H), 7.53 (s, 1H), 7.38 (d, J=8.0, 2H), 7.32 (d, J=8.5, 1H), 7.26 (s, 1H), 7.06 (s, 2H), 6.84 (d, J=8.5, 1H), 5.21 (s, 2H), 3.59 (t, 2H), 3.37 (t, 2H), 3.31 (s, 3H). Example 26—Synthesis of Compound 030 Step 1: Under N2, ethyl 7-bromoquinoline-3-carboxylate (280 mg, 1 mmol), 2-methoxy-N-methylethanamine (89 mg, 1 mmol), Pd2(dba)3(92 mg, 0.1 mmol), Ruphos (93 mg, 0.2 mmol) and Cs2CO3(652 mg, 2 mmol) were combined in toluene (10 mL). The reaction was stirred at 100° C. overnight. The residue was purified by column chromatography on silica gel (200-300 mesh, eluting with petroleum ether/EtOAc=1:2) to give compound 2 (250 mg, 89%). Step 2: Ethyl 7-((2-methoxyethyl)(methyl) amino) quinoline-3-carboxylate (250 mg, 0.87 mmol) and LiOH (55 mg, 1.3 mmol) were combined in EtOH (4 mL) and water (0.5 mL). The reaction was stirred at 60° C. for 4 h, then concentrated in vacuo to remove EtOH. The aqueous phase was adjusted to pH 7, then it was concentrated in vacuo to give the compound 3 (200 mg, 89%). Step 3: 7-((2-Methoxyethyl)(methyl) amino) quinoline-3-carboxylic acid (200 mg, 0.77 mmol), tert-butyl (2-amino-4-(thiophen-2-yl)phenyl)carbamate (223 mg, 0.77 mmol) and EDCI (444 mg, 2.31 mmol) were dissolved in pyridine (5 mL). The reaction was stirred at rt overnight. The reaction was then concentrated in vacuo, and the crude material was purified by column chromatography on silica gel (200-300 mesh, eluting DCM/CH3OH=1:1) to give compound 4 (300 mg, 76%). Step 4: tert-Butyl 2-(7-((2-methoxyethyl)(methyl) amino) quinoline-3-carboxamido)-4-(thiophen-2-yl) phenylcarbamate (300 mg, 0.56 mmol) and TFA (2 mL) were combined in DCM (4 mL). The reaction was stirred at rt for 2 h, then concentrated in vacuo. The crude material was washed with diethyl ether, and purified by prep-TLC (DCM/CH3OH=1:1) to give the product 030 (70 mg, 29%). LCMS: m/z=433.1 (M+H)+.1H-NMR (500 MHz, DMSO) δ 9.84 (s, 1H), 9.21 (s, 1H), 8.74 (s, 1H), 7.88 (d, J=8.5, 1H), 7.53 (s, 1H), 7.38 (d, J=8.0, 2H), 7.32 (d, J=8.5, 1H), 7.26 (s, 1H), 7.06 (s, 2H), 6.84 (d, J=8.5, 1H), 5.24 (s, 2H), 3.72 (s, 2H), 3.59 (s, 2H), 3.31 (s, 3H), 3.12 (s, 3H). Example 27—Synthesis of Compound 031 Step 1: Under N2, ethyl 7-bromoquinoline-3-carboxylate (500 mg, 1.79 mmol), N1,N1,N2-trimethylethane-1,2-diamine (182 mg, 1.79 mmol), Pd(OAc)2(40 mg, 0.18 mmol), Xphos (172 mg, 0.36 mmol) and Cs2CO3(1174 mg, 3.6 mmol) were combined in toluene (10 mL). The reaction was stirred at 100° C. overnight. The residue was purified by column chromatography on silica gel (200-300 mesh, eluting with petroleum ether/EtOAc=1:2) to give the compound 2 (280 mg, crude). Step 2: Ethyl 7-((2-(dimethylamino) ethyl)(methyl) amino) quinoline-3-carboxylate (280 mg, 0.93 mmol) and LiOH (59 mg, 1.4 mmol) were combined in EtOH (4 mL) and water (0.5 mL), and the reaction was stirred at 60° C. for 4 h. The reaction was concentrated in vacuo. The aqueous phase was adjusted to pH 7, then concentrated in vacuo to give the compound 3 (280 mg, crude). Step 3: 7-((2-(Dimethylamino) ethyl)(methyl) amino) quinoline-3-carboxylic acid (280 mg, 1 mmol), tert-butyl (2-amino-4-(thiophen-2-yl)phenyl)carbamate (232 mg, 0.8 mmol), and EDCI (576 mg, 3 mmol) were dissolved in pyridine (5 mL) and stirred at rt overnight. The reaction was then concentrated in vacuo and the crude material was carried over for next step (400 mg, crude). Step 4: tert-Butyl 2-(7-((2-(dimethylamino) ethyl)(methyl) amino) quinoline-3-carboxamido)-4-(thiophen-2-yl) phenylcarbamate (400 mg, 0.73 mmol) and TFA (2 mL) were combined in DCM (4 mL), and the reaction was stirred at rt for 2 h. The reaction was then concentrated in vacuo and purified by prep-HPLC (base method) to give the product 031 (57 mg, 18%). LCMS: m/z=446.1 (M+H)+.1H-NMR (500 MHz, DMSO) δ 9.88 (s, 1H), 9.22 (s, 1H), 8.75 (s, 1H), 8.26 (s, 1H), 7.90 (d, J=9.0, 1H), 7.53 (d, J=2.0, 1H), 7.31-7.36 (m, 3H), 7.26 (d, J=3.0, 1H), 7.06 (t, 2H), 6.84 (d, J=8.5, 1H), 3.69 (t, J=6.5, 2H), 3.11 (s, 3H), 2.66 (t, J=6.5, 2H), 2.37 (s, 6H). Example 28—Synthesis of Compound 032 Step 1: Under N2, Ethyl 7-bromoquinoline-3-carboxylate (1.1 g, 3.93 mmol), 1-methylpiperazine (432 mg, 4.31 mmol), Pd2(dba)3(89 mg, 0.097 mmol), Ruphos (91 mg, 0.195 mmol) and Cs2CO3(2.563 g, 7.87 mmol) were combined in toluene (53 mL) and stirred at 100° C. for 18 h. The crude material was purified by column chromatography on silica gel (200-300 mesh, eluting with petroleum ether/EtOAc=1:2) to give the compound 2 (1.1 g, 93.6%). Step 2: Ethyl 7-(4-methylpiperazin-1-yl)quinoline-3-carboxylate (1.1 g, 3.67 mmol) and LiOH (463 mg, 11.02 mmol) were combined in THE (23 mL), MeOH (3.8 mL) and water (7.6 mL). The reaction was stirred at rt for 4 h, then concentrated in vacuo to remove THE and MeOH. The aqueous phase was adjusted to pH 7, then concentrated in vacuo to give the compound 3 (2 g, crude). Step 3: 7-(4-Methylpiperazin-1-yl)quinoline-3-carboxylic acid (1.065 g, crude), tert-butyl (2-amino-4-(thiophen-2-yl)phenyl)carbamate (484 mg, 1.67 mmol) and EDCI (750 mg, 3.91 mmol) were dissolved in pyridine (19 mL) and stirred at rt overnight. The reaction was concentrated in vacuo, and the crude residue was purified by column chromatography on silica gel (200-300 mesh, eluting DCM/CH3OH=1:1) to give compound 5 (1.9 g, crude). Step 4: tert-Butyl (2-(7-(4-methylpiperazin-1-yl)quinoline-3-carboxamido)-4-(thiophen-2-yl)phenyl)carbamate (1.9 g, crude) and TFA (5 mL) were combined in DCM (5 mL). The reaction was stirred at rt for 1 h then concentrated in vacuo. The crude material was washed with diethyl ether, and the residue was purified by prep-HPLC (0.1% NaHCO3/water-acetonitrile) to give the product 032 (365 mg, 45.6%). LCMS: m/z=444 (M+H)+.1H-NMR (400 MHz, DMSO) δ 9.84 (s, 1H), 9.21 (s, 1H), 8.74 (s, 1H), 7.88 (d, J=8.5, 1H), 7.53 (s, 1H), 7.38 (d, J=8.0, 2H), 7.32 (d, J=8.5, 1H), 7.26 (s, 1H), 7.06 (s, 2H), 6.84 (d, J=8.5, 1H), 5.25 (s, 2H), 3.41 (t, 4H), 2.50 (t, 4H), 2.25 (s, 3H). Example 29—Pharmacokinetics Male SD rats were fasted overnight. Compounds of the invention were dissolved in dimethyl acetamide at 10 times the final concentration, then Solutol HS 15 (BASF) was added to a final concentration of 10%. Finally, 80% saline was added and vortexed to achieve a clear solution. For the IV dosing three animals were injected via the foot dorsal vein with 1 mg/kg compound. For the PO dosing 5 mg/kg of compound was delivered by oral gavage. Blood was collected via the tail vein into K2EDTA tubes at 5 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours and 24 hours after dosing. The blood was centrifuged at 2000 g for 5 minutes at 4° C. to obtain plasma. The plasma was extracted with acetonitrile and the level of compound was analyzed by LC/MS/MS. The level of compound in plasma was calculated from a standard curve in rat plasma. The IV clearance (L/h/kg) and area under the curve (h*ng/mL) were calculated using WinNonLin software. The dose adjusted area under the curve for the IV and oral dosing were used to calculate the oral bioavailability. Pharmacokinetic properties were assessed in a rat cassette dosing experiment. The IV clearance (IV Clr.) is in units of L/hr/kg. The oral maximum plasma concentration (PO Cmax) is in units of ng/ml. The oral plasma half-life (PO T½) is in units of hours. The oral area under the curve (PO AUC) is in units of hours*ng/ml. The fraction absorbed by the oral route (F %) is a percentage of the oral area under the curve to the IV area under the curve, dose adjusted. A summary of results is presented in Table 3 and Table 4, below. TABLE 3PK Parameters (Rat IV)CompoundIV Clr. (L/h/kg)IV t1/2(h)IV AUC (h*ng/mL)00140050-401500020.01-0.51-56500-66000030.1-15-101800-19000040.01-0.515-2010650-107500090.01-0.515-2010750-108500110.1-15-102000-21000120.01-0.55-1029850-299500130.1-11-52700-28000180.01-0.510-1519650-197500210.1-11-53700-38000240-0.515-2020-250250-0.51-515-200290-0.11-320-250300-0.10-0.51-50310-0.20.5-11-50320-0.50.5-20-15 TABLE 4PK Parameters (Rat PO)CompoundCmaxPO t1/2(h)PO AUC (h*ng/mL)F %0016950-7000139950-14005065-750022450-25005-1022350-2245065-75003950-10005-1011450-11550130-1400043500-355010-1561250-61350110-1150094150-420010-1565250-65350115-125011700-7505-108000-810080-900128200-825015-209450-955060-700131900-19501-511750-1185080-900185550-56005-1081850-8195075-850215250-53001-521350-21450110-120 Example 21—HDAC Enzyme Assays Compounds for testing were diluted in DM30 to 50 fold the final concentration and a ten-point three-fold dilution series was made. The compounds were diluted in assay buffer (50 mM HEPES, pH 7.4, 100 mM KCl, 0.001% Tween-20, 0.05% BSA, 20 μM TCEP) to 6-fold their final concentration. The HDAC enzymes (purchased from BPS Biosciences) were diluted to 1.5-fold their final concentration in assay buffer. The tripeptide substrate and trypsin at 0.05 μM final concentration were diluted in assay buffer at 6-fold their final concentration. The final enzyme concentrations used in these assays were 3.3 ng/ml (HDAC1), 0.2 ng/ml (HDAC2) and 0.08 ng/ml (HDAC3). The final substrate concentrations used were 16 μM (HDAC1), 10 μM (HDAC2) and 17 μM (HDAC3). Five μl of compounds and 20 μl of enzyme were added to wells of a black, opaque 384 well plate in duplicate. Enzyme and compound were incubated together at room temperature for 10 minutes. Five μl of substrate was added to each well, the plate was shaken for 60 seconds and placed into a Victor 2 microtiter plate reader. The development of fluorescence was monitored for 60 min and the linear rate of the reaction was calculated. The IC50was determined using Graph Pad Prism by a four parameter curve fit. TABLE 5IC50(nM)CompoundHDAC1HDAC2HDAC30015-1010-15340-35000220-2545-50925-9350031-105-25145-5900045-1025-30530-54000610-1570-751130-114000725-3060-652740-2750008625-650>2000>200000910-1540-45470-480010375-380210-220985-99501110-1535-40490-50001225-3520-30875-88501325-3570-801040-10500144185-41952140-21503370-338001515-2530-40640-6500161-105-15710-7200171-1010-20450-4600181-1020-35150-23501915-2035-40690-7000205-1025-301300-13100215-1015-20430-440022885-8951240-12501210-12200240-55-1015-900255-1515-20110-20002625-3020-2585-95027>2000>2000>200002850-6050-60290-30002915-2550-105750-76003050-55155-1601200-12300310-515-25240-2500325-1025-35280-300 The disclosed subject matter is not to be limited in scope by the specific embodiments and examples described herein. Indeed, various modifications of the disclosure in addition to those described will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications are intended to fall within the scope of the appended claims. All references (e.g., publications or patents or patent applications) cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual reference (e.g., publication or patent or patent application) was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. Other embodiments are within the following claims.
86,340
11858920
BEST MODE FOR CARRYING OUT THE INVENTION Hereinafter, the present invention will be described in detail by way of embodiments, examples, etc., but the present invention is not limited to embodiments, examples, etc. described below and can be arbitrarily changed and then practiced within a range not departing from the gist of the present invention. Note that all the documents and publications cited herein are incorporated herein by reference in their entireties regardless of purposes thereof. One embodiment of the present invention relates to an episulfide compound represented by formula (1) below (hereinafter also referred to as just “the episulfide compound (1)”). Further, another embodiment of the present invention relates to a composition for optical materials containing the episulfide compound represented by formula (1) below and an episulfide compound represented by formula (2) below (hereinafter also referred to as just “the episulfide compound (2)”), and a composition for optical materials further containing a compound which can be polymerized with the compound represented by formula (2) below, etc. (e.g., a compound (c), a compound (d) and sulfur, which will be described later) according to need. In formula (1), X1and X2represent O (oxygen atom) or S (sulfur atom). In a specific embodiment, regarding the episulfide compound (1), X1and X2are O (i.e., X1═X2═O), or X1is O and X2is S (i.e., X1═O and X2═S) in formula (1). In one embodiment, X1and X2are O (i.e., X1═X2═O) in formula (1). When an epoxy ring is contained, rapid polymerization reaction progress is suppressed, and a polymer having excellent transparency, wherein striae are reduced, tends to be obtained thereby. In one embodiment, the episulfide compound (1) is a mixture of a compound in which X1═X2═O, a compound in which X1═O and X2═S and a compound in which X1═X2═S, and these compounds may be present at any ratio. In one embodiment, the episulfide compound (1) includes at least one of a compound in which X1═X2═O in formula (1) (hereinafter also referred to as “the episulfide compound (B1)”) and a compound in which X1═O and X2═S in formula (1) (hereinafter also referred to as “the episulfide compound (B2)”), and according to need, a compound in which X1═X2═S in formula (1) (hereinafter also referred to as “the episulfide compound (B3)”). In one embodiment, the episulfide compound (1) includes the episulfide compound (B1) and the episulfide compound (B2). In one embodiment, the episulfide compound (1) includes the episulfide compound (B1), the episulfide compound (B2) and the episulfide compound (B3). When the episulfide compound (1) is a mixture of the episulfide compound (B1), the episulfide compound (B2) and/or the episulfide compound (B3), the ratio between them is not particularly limited. In one embodiment, the episulfide compound (1) includes 1 to 99% by mass (preferably 5 to 95% by mass, and more preferably 10 to 99% by mass) of the episulfide compound (B1), 1 to 99% by mass (preferably 5 to 95% by mass, and more preferably 10 to 90% by mass) of the episulfide compound (B2) and 0 to 95% by mass (preferably 5 to 95% by mass, and more preferably 10 to 90% by mass) of the episulfide compound (B3) relative to the total mass (100% by mass) of the episulfide compound. Further, the ratio of the epoxy ring contained in the episulfide compound (1) (the ratio of the number of epoxy rings to the total number of epoxy rings and epithio rings) is preferably 50% or more, more preferably 51% or more, and even more preferably 52% or more. One embodiment of the present invention is a composition for optical materials containing the episulfide compound represented by formula (1) and the episulfide compound represented by formula (2), and the ratio (content) of the compound represented by formula (1) (the episulfide compound (1)) in the composition for optical materials is preferably 0.001 to 5.0% by mass, more preferably 0.005 to 3.0% by mass, and particularly preferably 0.01 to 1.0% by mass relative to the total amount (100% by mass) of the composition for optical materials. When the content of the compound represented by formula (1) is less than 0.001% by mass, sufficient effects (e.g., high transparency and reduction of striae) may not be obtained, and when the content is more than 5.0% by mass, mold releasability may be deteriorated. Further, in one embodiment of the present invention, the ratio (content) of the compound represented by formula (2) (episulfide compound (2)) in the composition for optical materials is preferably 40 to 99.999% by mass, more preferably 50 to 99.995% by mass, and particularly preferably 60 to 99.99% by mass relative to the total amount (100% by mass) of the composition for optical materials. When the content is 40% by mass or more, excellent optical characteristics of the episulfide compound tend to be obtained. It is not known exactly why excellent effects as described above (e.g., transparency, low-level striae, peeling prevention property and mold releasability) are obtained by mixing the episulfide compound (1) with the episulfide compound (2), but it is speculated that it is because polymerization reaction progress is moderated. In one preferred embodiment, in the episulfide compound (1) represented by formula (1) contained in the composition for optical materials, the ratio of the total of the episulfide compound (B1) in which both X1and X2are O in formula (1) and the episulfide compound (B2) in which X1is O and X2is S in formula (1) is 50% by mass or more (more preferably 60% by mass or more, and even more preferably 70% by mass or more). In one embodiment, in the episulfide compound represented by formula (1) contained in the composition for optical materials, the ratio of the episulfide compound (B1) in which both X1and X2are O in formula (1) is 10% by mass or more (more preferably 25% by mass or more, and even more preferably 40% by mass or more). Hereinafter, the method for producing the compound represented by formula (1) and the compound represented by formula (2) will be described, but the production method is not particularly limited. The compound represented by formula (1) and the compound represented by formula (2) can be obtained by reacting an epoxy compound represented by formula (3) below with thiourea. Note that when obtaining the compound represented by formula (1) by reacting the epoxy compound represented by formula (3) with thiourea, interrupting the reaction before completed is a technique for efficiently obtaining a mixture of the compound represented by formula (1) and the compound represented by formula (2). Specifically, in a mixed solvent of a polar organic solvent which can dissolve thiourea and a non-polar organic solvent which can dissolve the epoxy compound represented by formula (3), the reaction is performed in the presence of an acid or acid anhydride or an ammonium salt, and the reaction is terminated before completed. That is, in one embodiment, the method for producing the composition for optical materials includes a step of reacting an epoxy compound represented by the above formula (3) with thiourea and optionally adding the episulfide compound represented by formula (2) thereto to obtain a mixture of the episulfide compound represented by formula (1) and the episulfide compound represented by formula (2). In the case where the composition for optical materials is constituted by using the episulfide compound (1) whose content ratio of the epoxy ring is high (for example, in the case where the episulfide compound (B1) and the episulfide compound (B2) are contained at a high content ratio (for example, the total of B1 and B2 is 50% by mass or more)), usually, after the episulfide compound represented by formula (1) is obtained by reacting the epoxy compound represented by formula (3) with thiourea, the episulfide compound (2) represented by formula (2) is required to be added. That is, in one embodiment, the method for producing the composition for optical materials includes a step of reacting an epoxy compound represented by the above formula (3) with thiourea and adding the episulfide compound represented by formula (2) thereto to obtain a mixture of the episulfide compound represented by formula (1) and the episulfide compound represented by formula (2). As the episulfide compound represented by formula (2), a synthesized product obtained by completely reacting the compound of formula (3) with thiourea may be used. In the method for obtaining the compound represented by formula (1) and the compound represented by formula (2) by means of the aforementioned reaction, thiourea is used in a mole number corresponding to epoxy of the epoxy compound represented by formula (3), i.e., a theoretical amount, but when importance is placed on the reaction rate and the purity, thiourea is used in the theoretical amount to 2.5 times the theoretical amount (mol). The amount is preferably from 1.3 times the theoretical amount (mol) to 2.4 times the theoretical amount (mol), and more preferably from 1.5 times the theoretical amount (mol) to 2.3 times the theoretical amount (mol). Examples of the polar organic solvent which can dissolve thiourea include: alcohols such as methanol and ethanol; ethers such as diethyl ether, tetrahydrofuran and dioxane; and hydroxy ethers such as methyl cellosolve, ethyl cellosolve and butyl cellosolve. Among them, alcohols are preferred, and methanol is most preferred. Examples of the non-polar organic solvent which can dissolve the epoxy compound represented by formula (3) include: aliphatic hydrocarbons such as pentane, hexane and heptane; aromatic hydrocarbons such as benzene and toluene; and halogenated hydrocarbons such as dichloromethane, chloroform and chlorobenzene. Among them, aromatic hydrocarbons are preferred, and toluene is most preferred. The ratio of the polar organic solvent/the non-polar organic solvent is 0.1 to 10.0 (volume ratio), and preferably 0.2 to 5.0 (volume ratio). When the volume ratio is less than 0.1, thiourea is not sufficiently dissolved and the reaction does not proceed sufficiently, and when the volume ratio is more than 10.0, polymer formation may become pronounced. The reaction temperature is 10° C. to 30° C. When the reaction temperature is lower than 10° C., not only the reaction rate is reduced, but also thiourea is not sufficiently dissolved and the reaction does not proceed sufficiently, and when the temperature is higher than 30° C., polymer formation becomes pronounced. Specific examples of the acid or acid anhydride to be used include: inorganic acidic compounds such as nitric acid, hydrochloric acid, perchloric acid, hypochlorous acid, chlorine dioxide, hydrofluoric acid, sulfuric acid, fuming sulfuric acid, sulfuryl chloride, boric acid, arsenic acid, arsenious acid, pyroarsenic acid, phosphoric acid, phosphorous acid, hypophosphoric acid, phosphorus oxychloride, phosphorous oxybromide, phosphorus sulfide, phosphorus trichloride, phosphorus tribromide, phosphorus pentachloride, hydrocyanic acid, chromic acid, nitric anhydride, sulphuric anhydride, boron oxide, arsenic pentoxide, phosphorus pentoxide, chromic anhydride, silica gel, silica alumina, aluminium chloride and zinc chloride; organic carboxylic acids such as formic acid, acetic acid, peracetic acid, thioacetic acid, oxalic acid, tartaric acid, propionic acid, butyric acid, succinic acid, valeric acid, caproic acid, caprylic acid, naphthenic acid, methyl mercaptopropionate, malonic acid, glutaric acid, adipic acid, cyclohexanecarboxylic acid, thiodipropionic acid, dithiodipropionic acid acetic acid, maleic acid, benzoic acid, phenylacetic acid, o-toluic acid, m-toluic acid, p-toluic acid, salicylic acid, 2-methoxybenzoic acid, 3-methoxybenzoic acid, benzoylbenzoic acid, phthalic acid, isophthalic acid, terephthalic acid, salicylic acid, benzilic acid, α-naphthalenecarboxylic acid, β-naphthalenecarboxylic acid, acetic anhydride, propionic anhydride, butyric anhydride, succinic anhydride, maleic anhydride, benzoic anhydride, phthalic anhydride, pyromellitic dianhydride, trimellitic anhydride and trifluoroacetic anhydride; phosphoric acids such as mono-, di- or trimethyl phosphate, mono-, di- or triethyl phosphate, mono-, di- or triisobutyl phosphate, mono-, di- or tributyl phosphate and mono-, di- or trilauryl phosphate, and phosphorous acids in which the phosphate moiety of any of the phosphoric acids is changed to a phosphite; organic phosphorous compounds such as dialkyl phosphorodithioates typified by dimethyl phosphorodithioate; phenols such as phenol, catechol, t-butyl catechol, 2,6-di-t-butyl cresol, 2,6-di-t-butyl ethylphenol, resorcin, hydroquinone, phloroglucin, pyrogallol, cresol, ethyl phenol, butyl phenol, nonyl phenol, hydroxyphenylacetic acid, hydroxyphenylpropionic acid, hydroxyphenylacetamide, methyl hydroxyphenylacetate, ethyl hydroxyphenylacetate, hydroxyphenethyl alcohol, hydroxyphenethyl amine, hydroxybenzaldehyde, phenylphenol, bisphenol A, 2,2′-methylene-bis(4-methyl-6-t-butyl phenol), bisphenol F, bisphenol S, α-naphthol, β-naphthol, aminophenol, chlorophenol and 2,4,6-trichlorophenol; and sulfonic acids such as methanesulfonic acid, ethanesulfonic acid, butanesulfonic acid, dodecanesulfonic acid, benzenesulfonic acid, o-toluenesulfonic acid, m-toluenesulfonic acid, p-toluenesulfonic acid, ethylbenzenesulfonic acid, butylbenzenesulfonic acid, dodecylbenzenesulfonic acid, p-phenolsulfonic acid, o-cresolsulfonic acid, metanilic acid, sulfanilic acid, 4B-acid, diaminostilbenesulfonic acid, biphenylsulfonic acid, α-naphthalenesulfonic acid, β-naphthalenesulfonic acid, peri acid, Laurent's acid and phenyl-J-acid (7-anilino-4-hydroxy-2-naphthalenesulfonic acid). Several of them may be used in combination. Preferred are organic carboxylic acids such as formic acid, acetic acid, peracetic acid, thioacetic acid, oxalic acid, tartaric acid, propionic acid, butyric acid, succinic acid, valeric acid, caproic acid, caprylic acid, naphthenic acid, methyl mercaptopropionate, malonic acid, glutaric acid, adipic acid, cyclohexanecarboxylic acid, thiodipropionic acid, dithiodipropionic acid acetic acid, maleic acid, benzoic acid, phenylacetic acid, o-toluic acid, m-toluic acid, p-toluic acid, salicylic acid, 2-methoxybenzoic acid, 3-methoxybenzoic acid, benzoylbenzoic acid, phthalic acid, isophthalic acid, terephthalic acid, salicylic acid, benzilic acid, α-naphthalenecarboxylic acid, β-naphthalenecarboxylic acid, acetic anhydride, propionic anhydride, butyric anhydride, succinic anhydride, maleic anhydride, benzoic anhydride, phthalic anhydride, pyromellitic dianhydride, trimellitic anhydride and trifluoroacetic anhydride. More preferred are acid anhydrides such as acetic anhydride, propionic anhydride, butyric anhydride, succinic anhydride, maleic anhydride, benzoic anhydride, phthalic anhydride, pyromellitic dianhydride, trimellitic anhydride and trifluoroacetic anhydride. Acetic anhydride is most preferred. The amount of the acid or acid anhydride to be added is usually 0.001 to 10% by mass, and preferably 0.01 to 5% by mass relative to the total amount of the reaction solution. When the amount to be added is less than 0.001% by mass, polymer formation becomes pronounced, resulting in reduction in the yield of the reaction, and when the amount is more than 10% by mass, the yield may be significantly reduced. Further, specific examples of the ammonium salt include ammonium chloride, ammonium bromide, ammonium iodide, ammonium formate, ammonium acetate, ammonium propionate, ammonium benzoate, ammonium sulfate, ammonium nitrate, ammonium carbonate, ammonium phosphate and ammonium hydroxide. Ammonium nitrate, ammonium sulfate and ammonium chloride are more preferred, and ammonium nitrate is most preferred. The reaction is monitored by NMR, IR, liquid chromatograph or gas chromatograph and terminated in a state where the compound represented by formula (1) remains. In one embodiment, the reaction is terminated in a state where the compounds (B1), (B2) and (B3) exist at a desired ratio. In another embodiment, the reaction is terminated in a state where the amount of the compound represented by formula (1) is 0.05 to 20% by mass, more preferably 0.1 to 15% by mass, particularly preferably 0.5 to 10% by mass, and most preferably 0.5 to 4% by mass. The compound represented by formula (1) thus obtained is subjected to column purification, thereby isolating the compound (B1), the compound (B2) and the compound (B3) respectively. In the case of obtaining the compound represented by formula (2), the reaction is monitored by NMR, IR, liquid chromatograph or gas chromatograph and terminated in a state where epoxy rings have been completely converted into epithio rings. The composition for optical materials can be obtained by mixing the compound represented by formula (1) obtained by the above-described reaction or the compound (B1), the compound (B2) and/or the compound (B3) obtained by isolation therefrom with the compound represented by formula (2). [1,2,3,5,6-pentathiepane (c)] The composition for optical materials of the present invention may contain 1,2,3,5,6-pentathiepane (c) according to need. 1,2,3,5,6-pentathiepane (c) is a compound represented by formula (c) below and has the effect of improving the refractive index of an optical material (resin) obtained from the composition for optical materials of the present invention. The method for obtaining 1,2,3,5,6-pentathiepane (c) is not particularly limited. A commercially-available product may be used as 1,2,3,5,6-pentathiepane (c). Alternatively, 1,2,3,5,6-pentathiepane (c) may be collected and extracted from natural material such as crude oil, animals and plants or may be synthesized according to a publicly-known method. Examples of synthesis methods include those described in: N. Takeda et al., Bull. Chem. Soc. Jpn., 68, 2757 (1995); F. Feher et al., Angew. Chem. Int. Ed., 7, 301 (1968); G. W. Kutney et al., Can. J. Chem, 58, 1233 (1980); etc. When using 1,2,3,5,6-pentathiepane (c), the ratio thereof in the composition for optical materials is preferably 5 to 70% by mass, and more preferably 5 to 50% by mass relative to the total amount of the composition for optical materials. When the ratio is within the above-described range, a balance between the improvement of the refractive index and transparency of the optical material can be achieved. [Polythiol (d)] The composition for optical materials may contain a polythiol (d) according to need. The polythiol (d) is a thiol compound having at least two mercapto groups per one molecule. The polythiol (d) has the effect of improving the color tone of resin obtained from the composition for optical materials of the present invention at the time of heating. The polythiol to be used in the present invention is not particularly limited, but in terms of being highly effective for the improvement of the color tone, preferred specific examples thereof include 1,2,6,7-tetramercapto-4-thiaheptane, methanedithiol, (sulfanylmethyldisulfanyl)methanethiol, bis(2-mercaptoethyl)sulfide, 2,5-bis(mercaptomethyl)-1,4-dithiane, 1,2-bis(2-mercaptoethylthio)-3-mercaptopropane, 4,8-dimercaptomethyl-1,11-dimercapto-3,6,9-trithiaundecane, 4,7-dimercaptomethyl-1,11-dimercapto-3,6,9-trithiaundecane, 5,7-dimercaptomethyl-1,11-dimercapto-3,6,9-trithiaundecane, 1,1,3,3-tetrakis(mercaptomethylthio)propane, tetramercaptopentaerythritol, 1,3-bis(mercaptomethyl)benzene, 1,4-bis(mercaptomethyl)benzene and thiiranemethanethiol. Particularly preferred are bis(2-mercaptoethyl)sulfide, 1,2,6,7-tetramercapto-4-thiaheptane, 1,2-bis(2-mercaptoethylthio)-3-mercaptopropane and 1,3-bis(mercaptomethyl)benzene, and most preferred is 1,2,6,7-tetramercapto-4-thiaheptane. As these materials, a commercially-available product or a product obtained by synthesis according to a publicly-known method can be used. Further, two or more of these materials can be used in combination. The ratio of the polythiol (d) in the composition for optical materials is preferably 0 to 25% by mass (e.g., 0.1 to 25% by mass), more preferably 0 to 20% by mass (e.g., 0.5 to 20% by mass), even more preferably 0 to 15% by mass (e.g., 0.5 to 15% by mass), and particularly preferably 0 to 12% by mass (e.g., 0.5 to 12% by mass) relative to the total amount of the composition for optical materials. [Sulfur] The composition for optical materials may contain sulfur according to need. Sulfur has the effect of improving the refractive index of the optical material (resin) obtained from the composition for optical materials of the present invention. The sulfur to be used in the present invention may be in any form. Specific examples of the sulfur include finely-powdered sulfur, colloidal sulfur, precipitated sulfur, crystalline sulfur and sublimed sulfur, and from the viewpoint of the dissolution rate, finely-powdered sulfur having fine particles is preferred. It is preferred that the particle size (diameter) of the sulfur to be used in the present invention is less than 10 mesh. When the particle size of the sulfur is more than 10 mesh, it is difficult to dissolve the sulfur completely. The particle size of the sulfur is more preferably less than 30 mesh, and most preferably less than 60 mesh. The purity of the sulfur to be used in the present invention is preferably at least 98%, more preferably at least 99.0%, even more preferably at least 99.5%, and most preferably at least 99.9%. When the purity of the sulfur is at least 98%, the color tone of the obtained optical material is improved compared to the case of lower than 98%. As the sulfur satisfying the above-described conditions, a commercially-available product can be easily obtained and suitably used. In the composition for optical materials, the ratio of the sulfur is usually 0 to 40% by mass (e.g., 1 to 40% by mass), preferably 0 to 30% by mass (e.g., 5 to 30% by mass or 10 to 30% by mass), more preferably 0 to 25% by mass (e.g., 5 to 25% by mass), and particularly preferably 0 to 20% by mass (e.g., 5 to 20% by mass) relative to the total amount of the composition for optical materials. When obtaining an optical material by polymerizing and curing the composition for optical materials of the present invention, it is preferred to add a polymerization catalyst. As the polymerization catalyst, amines, phosphines and onium salts may be used, but onium salts are particularly preferred. Among them, quaternary ammonium salts, quaternary phosphonium salts, tertiary sulfonium salts and secondary iodonium salts are preferred. Among them, quaternary ammonium salts and quaternary phosphonium salts, which have good compatibility with the composition for optical materials, are more preferred, and quaternary phosphonium salts are even more preferred. More preferred examples of the polymerization catalyst include quaternary ammonium salts such as tetra-n-butylammonium bromide, triethylbenzyl ammonium chloride, cetyldimethylbenzyl ammonium chloride and 1-n-dodecyl pyridinium chloride and quaternary phosphonium salts such as tetra-n-butylphosphonium bromide and tetraphenyl phosphonium bromide. Among them, tetra-n-butylammonium bromide, triethylbenzyl ammonium chloride and tetra-n-butylphosphonium bromide are even more preferred polymerization catalysts. The amount of the polymerization catalyst to be added cannot be determined categorically because it varies depending on the components of the composition, the mixing ratio and the method for polymerization and curing, but the amount is usually 0.0001 to 10% by mass, preferably 0.001 to 5% by mass, more preferably 0.01 to 1% by mass, and most preferably 0.01 to 0.5% by mass relative to the total amount (100% by mass) of the composition for optical materials. When the amount of the polymerization catalyst to be added is more than 10% by mass, the composition may be rapidly polymerized. When the amount of the polymerization catalyst to be added is less than 0.0001% by mass, the composition for optical materials may be insufficiently cured, resulting in poor heat resistance. Moreover, in the production of the optical material according to the production method of the present invention, it is surely possible to add additives such as an ultraviolet absorber, a blueing agent and a pigment to the composition for optical materials to further improve practicability of the optical material obtained. Preferred examples of the ultraviolet absorber include benzotriazole-based compounds, and 2-(2-hydroxy-5-methylphenyl)-2H-benzotriazol, 5-chloro-2-(3,5-di-tert-butyl-2-hydroxyphenyl)-2H-benzotriazol, 2-(2-hydroxy-4-octylphenyl)-2H-benzotriazol, 2-(2-hydroxy-4-methoxyphenyl)-2H-benzotriazol, 2-(2-hydroxy-4-ethoxyphenyl)-2H-benzotriazol, 2-(2-hydroxy-4-butoxyphenyl)-2H-benzotriazol, 2-(2-hydroxy-4-octyloxyphenyl)-2H-benzotriazol and 2-(2-hydroxy-5-t-octylphenyl)-2H-benzotriazol are particularly preferred compounds. The amount of each of the antioxidant and the ultraviolet absorber to be added is usually 0.01 to 5% by mass relative to the total amount (100% by mass) of the composition for optical materials. When polymerizing and curing the composition for optical materials, for the purpose of extension of the pot life, dispersion of heat generated by polymerization, etc., a polymerization modifier may be added according to need. Examples of the polymerization modifier include halides of groups 13 to 16 of the long form of the periodic table. Among them, halides of silicon, germanium, tin and antimony are preferred, and chlorides of germanium, tin and antimony, which have an alkyl group, are more preferred. Further, dibutyltin dichloride, butyltin trichloride, dioctyltin dichloride, octyltin trichloride, dibutyldichlorogermanium, butyltrichlorogermanium, diphenyldichlorogermanium, phenyltrichlorogermanium and triphenylantimony dichloride are even more preferred, and dibutyltin dichloride is the most preferred compound. These polymerization modifiers may be used solely, or two or more of them may be used in combination. The amount of the polymerization modifier to be added is 0.0001 to 5.0% by mass, preferably 0.0005 to 3.0% by mass, and more preferably 0.001 to 2.0% by mass relative to the total amount (100% by mass) of the composition for optical materials. When the amount of the polymerization modifier to be added is less than 0.0001% by mass, sufficient pot life cannot be ensured in the obtained optical material, and when the amount of the polymerization modifier to be added is more than 2.0% by mass, the composition for optical materials may not be sufficiently cured, and the heat resistance of the obtained optical material may be reduced. The composition for optical materials thus obtained is injected into a mold or the like and polymerized to obtain an optical material. At the time of cast-molding the composition for optical materials of the present invention, it is preferred to filter and remove impurities using, for example, a filter having a pore diameter of about 0.1 to 5 μm in terms of improving the quality of the optical material of the present invention. The composition for optical materials of the present invention is usually polymerized as described below. Specifically, the curing time is usually 1 to 100 hours, and the curing temperature is usually −10° C. to 140° C. The polymerization is conducted by carrying out a step of retaining the composition at a predetermined polymerization temperature for a predetermined amount of time, a step of increasing the temperature at a rate of 0.1° C. to 100° C./h and a step of decreasing the temperature at a rate of 0.1° C. to 100° C./h, or a combination of these steps. Further, it is preferred to anneal the obtained optical material at a temperature of 50 to 150° C. for about 10 minutes to 5 hours after curing is completed in terms of eliminating distortion of the optical material of the present invention. Moreover, the obtained optical material may be subjected to a surface treatment such as dyeing, hard coating, impact-resistant coating, antireflection treatment and imparting antifog properties according to need. The optical material of the present invention can be suitably used as an optical lens. EXAMPLES Hereinafter, the present invention will be specifically described by way of working examples and comparative examples. However, the present invention is not limited to the below-described working examples. Note that optical materials obtained according to the below-described methods of working examples and comparative examples were evaluated according to the below-described methods. 1. Method for Evaluating Mold Releasability 10 lenses having a lens power of +12D were prepared according to the method described in the Examples below. When the lenses were released from molds, the case where no lens was broken was rated as “A”, the case where 1 lens was broken was rated as “B”, and the case where 2 or more lenses were broken was rated as “C”. A and B are regarded as acceptable. A and B are preferred, and A is particularly preferred. 2. Method for Evaluating Peeling Traces 10 lenses having a lens power of −15D were prepared according to the method described in the Examples below. After released from molds, the lenses were annealed at 120° C. for 30 minutes, and then surface conditions thereof were visually observed. Regarding the 10 lenses prepared, the case where no peeling trace was generated in the lenses was rated as “A”, the case where 1 lens had peeling traces was rated as “B”, and the case where 2 or more lenses had peeling traces was rated as “C”. A and B are regarded as acceptable. A and B are preferred, and A is particularly preferred. 3. Method for Evaluating Transparency According to the methods described in the Examples and Comparative Examples below, 10 lenses were prepared, and the lenses were observed under a fluorescent light in a dark room. The case where no white turbidity was observed in the 10 lenses was rated as “A”. The case where white turbidity was not observed in 7 to 9 lenses was rated as “B”. The case where white turbidity was not observed in 6 lenses or less was rated as “C”. A and B are regarded as acceptable. 4. Method for Evaluating Striae According to the methods described in the Examples and Comparative Examples below, 10 lenses were prepared, and the lenses were visually observed according to the schlieren method. The case where no stria was observed in the 10 lenses was rated as “A”. The case where striae were not observed in 7 to 9 lenses was rated as “B”. The case where striae were not observed in 6 lenses or less was rated as “C”. A and B are regarded as acceptable. Example 1 To 20.1 g (0.047 mol) of tetrakis(β-epoxypropylthiomethyl)methane, 100 mL of toluene, 100 mL of methanol, 1.24 g (0.012 mol) of acetic anhydride and 30.5 g (0.40 mol) of thiourea were added, and the mixture was stirred at 20° C. for 6 hours. After that, 400 mL of toluene and 400 mL of 5% sulfuric acid were added thereto, the toluene layer was washed with water three times, and the solvent was distilled away, thereby obtaining 16.8 g of a crude product of tetrakis(β-epithiopropylthiomethyl)methane. The crude product was further subjected to silica gel column purification, thereby obtaining 11.2 g of a compound (1) (hereinafter referred to as “the compound b”). It was subjected to the NMR measurement, and it was confirmed that a compound in which X1═X2═O, a compound in which X1═O and X2═S and a compound in which X1═X2═S were contained therein at a mass ratio of 40:30:30. The compound (1) used in the below-described experiments was synthesized according to this method. X1═X2═O (Episulfide compound (B1)) 1H-NMR (CDCl3): 2.54 ppm (1H), 2.34 ppm, 2.09 ppm (2H), 2.81 ppm (3H), 2.61 ppm, 2.36 ppm (6H), 2.88 ppm, 2.63 ppm (2H), 2.67 ppm, 2.43 ppm (6H), 2.30 ppm (8H) 13C-NMR (CDCl3): 32.6 ppm (1C), 26.4 ppm (1C), 53.8 ppm (3C), 46.8 ppm (3C), 44.8 ppm (1C), 41.5 ppm (3C), 37.5 ppm (1C), 37.6 ppm (3C), 38.9 ppm (1C) X1═O and X2═S (Episulfide compound (B2)) 1H-NMR (CDCl3): 2.54 ppm (2H), 2.34 ppm, 2.09 ppm (4H), 2.81 ppm (2H), 2.61 ppm, 2.36 ppm (4H), 2.88 ppm, 2.63 ppm (4H), 2.67 ppm, 2.43 ppm (4H), 2.30 ppm (8H) 13C-NMR (CDCl3): 32.6 ppm (2C), 26.4 ppm (2C), 53.8 ppm (2C), 46.8 ppm (2C), 44.8 ppm (2C), 41.5 ppm (2C), 37.5 ppm (2C), 37.6 ppm (2C), 38.9 ppm (1C) X1═X2═S (Episulfide compound (B3)) 1H-NMR (CDCl3): 2.54 ppm (4H), 2.34 ppm, 2.09 ppm (8H), 2.88 ppm, 2.63 ppm (8H), 2.30 ppm (8H) 13C-NMR (CDCl3): 32.6 ppm (3C), 26.4 ppm (3C), 53.8 ppm (1C), 46.8 ppm (1C), 44.8 ppm (3C), 41.5 ppm (1C), 37.5 ppm (3C), 37.6 ppm (1C), 38.9 ppm (1C) Examples 2-7 and Reference Example 1 Tetrakis(β-epithiopropylthiomethyl)methane that is the episulfide compound represented by formula (2) (hereinafter referred to as “the compound a”) was mixed with the compound represented by formula (1) obtained in Example 1 (hereinafter referred to as “the compound b”) to prepare a composition in which the ratio between the compound a and the compound b was as shown in Table 1. To 55 parts by mass of the obtained composition, 35 parts by mass of 1,2,3,5,6-pentathiepane (c), 10 parts by mass of 1,2,6,7-tetramercapto-4-thiaheptane and 0.05 part by mass of tetra-n-butylphosphonium bromide as a polymerization catalyst were added, and then the mixture was sufficiently mixed to be homogeneous. Subsequently, the obtained mixture was subjected to the deaeration treatment at a vacuum degree of 1.3 kPa and injected into a mold composed of two glass plates and a tape. It was heated at 30° C. for 10 hours, then the temperature was elevated to 100° C. at a constant rate over 10 hours, and finally, it was heated at 100° C. for 1 hour to be polymerized and cured. After cooling, the obtained product was released from the mold and annealed at 120° C. for 30 minutes to obtain a molded plate (12D or −15D). Mold releasability, peeling traces, transparency and striae of the obtained optical material were evaluated. The evaluation results are shown in Table 1. Comparative Example 1 A molded plate was obtained in a manner similar to that in Example 2, except that the compound a was used instead of the composition obtained by mixing the compound a and the compound b. The evaluation results are shown in Table 1. TABLE 1Compound ratio inAdditionCompound bamount of(% by mass)Compound aCompound bX1=X1= O,X1=MoldPeelingExamples(% by mass)(% by mass)X2= OX2= SX2= SreleasabilitytracesTransparencyStriaeExample 299.9990.001403030AABBExample 399.9950.005403030AABBExample 499.990.01403030AAAAExample 599.01.0403030AAAAExample 697.03.0403030BAAAExample 795.05.0403030BBAAComparative100Not added———AACCExample 1Reference93.07.0403030CCAAExample 1 Examples 8-13 and Comparative Example 2 Tetrakis(β-epithiopropylthiomethyl)methane that is the episulfide compound represented by formula (2) (hereinafter referred to as “the compound a”) was mixed with the compound represented by formula (1) obtained in Example 1 (hereinafter referred to as “the compound b”) to prepare a composition in which the ratio between the compound a and the compound b was as shown in Table 2. To 60 parts by mass of the obtained composition, 25 parts by mass of 1,2,3,5,6-pentathiepane (c), 5 parts by mass of 1,2,6,7-tetramercapto-4-thiaheptane, 10 parts by mass of sulfur and 0.05 part by mass of tetra-n-butylphosphonium bromide as a polymerization catalyst were added, and then the mixture was sufficiently mixed to be homogeneous. Subsequently, the obtained mixture was subjected to the deaeration treatment at a vacuum degree of 1.3 kPa and injected into a mold composed of two glass plates and a tape. It was heated at 30° C. for 10 hours, then the temperature was elevated to 100° C. at a constant rate over 10 hours, and finally, it was heated at 100° C. for 1 hour to be polymerized and cured. After cooling, the obtained product was released from the mold and annealed at 120° C. for 30 minutes to obtain a molded plate (12D or −15D). Mold releasability, peeling traces, transparency and striae of the obtained optical material were evaluated. The evaluation results are shown in Table 2. TABLE 2Compound ratio inAdditionCompound bamount of(% by mass)Compound aCompound bX1=X1= O,X1=MoldPeelingExamples(% by mass)(% by mass)X2= OX2= SX2= SreleasabilitytracesTransparencyStriaeExample 899.9990.001403030AABBExample 999.9950.005403030AABBExample 1099.990.01403030AAAAExample 1199.01.0403030AAAAExample 1297.03.0403030BAAAExample 1395.05.0403030BBAAComparative100Not added———AACCExample 2 As shown in Table 1 and Table 2 above, it is confirmed that in the case of the compositions of the Examples containing the episulfide compound represented by formula (1) (compound b) and the episulfide compound represented by formula (2) (compound a), mold release failure and peeling defects at the time of polymerization and curing are suppressed and high-quality optical materials having high transparency and low-level striae are obtained.
37,199
11858921
DETAILED DESCRIPTION OF THE INVENTION The present invention discloses novel uracil derivatives that are inhibitors of transient receptor potential ankyrin 1 (TRPA1), possessing appropriate pharmacological and pharmacokinetic properties enabling their use as medicaments for the treatment of conditions and/or diseases treatable by inhibition of TRPA1. The compounds of the present invention may provide several advantages, such as enhanced potency, high metabolic and/or chemical stability, high selectivity, safety and tolerability, enhanced solubility, enhanced permeability, desirable plasma protein binding, enhanced bioavailability, suitable pharmacokinetic profiles, and the possibility to form stable salts. The Compounds of the Invention The present invention provides uracil derivatives that are surprisingly potent inhibitors of TRPA1 (Assay A), further characterised byimproved stability in human liver microsomes (Assay B)improved stability in human hepatocytes (Assay C). Compounds of the present invention differ structurally from examples 53, 72, 73, 86 and 90 in WO2017/060488 and from example 31 in L. Schenkel, et al., J. Med. Chem. 2016, 59, 2794-2809, in that they contain a substituted uracil core as well as substituents adjacent to a secondary aliphatic alcohol. These structural differences unexpectedly lead to a favourable combination of (i) inhibition of TRPA1, (ii) stability in human liver microsomes, and (iii) stability in human hepatocytes. Stability in human liver microsomes refers to the susceptibility of compounds to biotransformation in the context of selecting and/or designing drugs with favorable pharmacokinetic properties as a first screening step. The primary site of metabolism for many drugs is the liver. Human liver microsomes contain the cytochrome P450s (CYPs), and thus represent a model system for studying phase I drug metabolism in vitro. Enhanced stability in human liver microsomes is associated with several advantages, including increased bioavailability and adequate half-life, which can enable lower and less frequent dosing of patients. Thus, enhanced stability in human liver microsomes is a favorable characteristic for compounds that are to be used for drugs. Therefore, compounds of the present invention in addition to being able to inhibit TRPA1 are expected to have a favorable in vivo clearance and thus the desired duration of action in humans. Stability in human hepatocytes refers to the susceptibility of compounds to biotransformation in the context of selecting and/or designing drugs with favorable pharmacokinetic properties. The primary site of metabolism for many drugs is the liver. Human hepatocytes contain the cytochrome P450s (CYPs) and other drug metabolizing enzymes, and thus represent a model system for studying drug metabolism in vitro. (Importantly, in contrast to liver microsomes assay, the hepatocytes assay covers also phase II biotransformations as well as liver-specific transporter-mediated processes, and therefore represents a more complete system for drug metabolism studies). Enhanced stability in human hepatocytes is associated with several advantages, including increased bioavailability and adequate half-life, which can enable lower and less frequent dosing of patients. Thus, enhanced stability in human hepatocytes is a favorable characteristic for compounds that are to be used for drugs. The present invention provides novel compounds according to formula (I) wherein A is selected from the group consisting of phenyl, thiophenyl, benzofuranyl and benzothiophenyl, and wherein A is unsubstituted or substituted with one or two members of the group R1consisting of halogen and C1-4-alkyl. Another embodiment of the present invention relates to a compound of formula (I) wherein A is selected from the group consisting of phenyl, thiophenyl, benzofuranyl and benzothiophenyl, and wherein A is unsubstituted or substituted with one or two members of the group R1consisting of F, Cl, I and CH3. Another embodiment of the present invention relates to a compound of formula (I) wherein A is selected from the group consisting of phenyl, benzofuranyl and benzothiophenyl, and wherein A is unsubstituted or substituted with one or two members of the group R1consisting of halogen and C1-4-alkyl. Another embodiment of the present invention relates to a compound of formula (I) wherein A is selected from the group consisting of phenyl, benzofuranyl and benzothiophenyl, and wherein A is unsubstituted or substituted with one or two members of the group R1consisting of F, Cl, I and CH3. Another embodiment of the present invention relates to a compound of formula (I) wherein A is selected from the group consisting of and wherein A is unsubstituted or substituted with one or two members of the group R1, and R1is defined as in any of the preceding embodiments. Preferred is the compound according to formula (I) selected from the group consisting of USED TERMS AND DEFINITIONS Terms not specifically defined herein should be given the meanings that would be given to them by one of skill in the art in light of the disclosure and the context. As used in the specification, however, unless specified to the contrary, the following terms have the meaning indicated and the following conventions are adhered to. In the groups, radicals, or moieties defined below, the number of carbon atoms is often specified preceding the group, for example, C1-6-alkyl means an alkyl group or radical having 1 to 6 carbon atoms. In general in groups like HO, H2N, (O)S, (O)2S, NC (cyano), HOOC, F3C or the like, the skilled artisan can see the radical attachment point(s) to the molecule from the free valences of the group itself. For combined groups comprising two or more subgroups, the last named subgroup is the radical attachment point, for example, the substituent “aryl-C1-3-alkyl” means an aryl group which is bound to a C1-3-alkyl-group, the latter of which is bound to the core or to the group to which the substituent is attached. In case a compound of the present invention is depicted in form of a chemical name and as a formula in case of any discrepancy the formula shall prevail. An asterisk may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined. The numeration of the atoms of a substituent starts with the atom that is closest to the core or to the group to which the substituent is attached. For example, the term “3-carboxypropyl-group” represents the following substituent: wherein the carboxy group is attached to the third carbon atom of the propyl group. The terms “1-methylpropyl-”, “2,2-dimethylpropyl-” or “cyclopropylmethyl-” group represent the following groups: The asterisk may be used in sub-formulas to indicate the bond that is connected to the core molecule as defined. The term “C1-n-alkyl”, wherein n is an integer selected from 2, 3, 4 or 5, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydro-carbon radical with 1 to n C atoms. For example the term C1-5-alkyl embraces the radicals H3C—, H3C—CH2—, H3C—CH2—CH2—, H3C—CH(CH3)—, H3C—CH2—CH2—CH2—, H3C—CH2—CH(CH3)—, H3C—CH(CH3)—CH2—, H3C—C(CH3)2—, H3C—CH2—CH2—CH2—CH2—, H3C—CH2—CH2—CH(CH3)—, H3C—CH2—CH(CH3)—CH2—, H3C—CH(CH3)—CH2—CH2—, H3C—CH2—C(CH3)2—, H3C—C(CH3)2—CH2—, H3C—CH(CH3)—CH(CH3)— and H3C—CH2—CH(CH2CH3)—. The term “fluoro” added to an “alkyl”, “alkylene” or “cycloalkyl” group (saturated or un-saturated) means such a alkyl or cycloalkyl group wherein one or more hydrogen atoms are replaced by a fluorine atom. Examples include, but are not limited to: H2FC—, HF2C— and F3C—. The term phenyl refers to the radical of the following ring The term thiophenyl refers to the radical of the following ring The term benzofuranyl refers to the radical of the following ring The term benzothiophenyl refers to the radical of the following ring The term uracil refers to the radical of the following core The term “substituted” as used herein, means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound. Unless specifically indicated, throughout the specification and the appended claims, a given chemical formula or name shall encompass tautomers and all stereo, optical and geo-metrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc.) and racemates thereof as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound. In general, substantially pure stereoisomers can be obtained according to synthetic principles known to a person skilled in the field, e.g. by separation of corresponding mixtures, by using stereochemically pure starting materials and/or by stereoselective synthesis. It is known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis, e.g. starting from optically active starting materials and/or by using chiral reagents. Enantiomerically pure compounds of this invention or intermediates may be prepared via asymmetric synthesis, for example by preparation and subsequent separation of appropriate diastereomeric compounds or intermediates which can be separated by known methods (e.g. by chromatographic separation or crystallization) and/or by using chiral reagents, such as chiral starting materials, chiral catalysts or chiral auxiliaries. Further, it is known to the person skilled in the art how to prepare enantiomerically pure compounds from the corresponding racemic mixtures, such as by chromatographic separation of the corresponding racemic mixtures on chiral stationary phases; or by resolution of a racemic mixture using an appropriate resolving agent, e.g. by means of diastereomeric salt formation of the racemic compound with optically active acids or bases, subsequent resolution of the salts and release of the desired compound from the salt; or by derivatization of the corresponding racemic compounds with optically active chiral auxiliary reagents, subsequent diastereomer separation and removal of the chiral auxiliary group; or by kinetic resolution of a racemate (e.g. by enzymatic resolution); by enantioselective crystallization from a conglomerate of enantiomorphous crystals under suitable conditions; or by (fractional) crystallization from a suitable solvent in the presence of an optically active chiral auxiliary. The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use without excessive toxicity, irritation, allergic response, or other problem or complication, and commensurate with a reasonable benefit/risk ratio. As used herein, “pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound forms a salt or a complex with an acid or a base. Examples of acids forming a pharmaceutically acceptable salt with a parent compound containing a basic moiety include mineral or organic acids such as benzenesulfonic acid, benzoic acid, citric acid, ethanesulfonic acid, fumaric acid, gentisic acid, hydrobromic acid, hydrochloric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, 4-methyl-benzenesulfonic acid, phosphoric acid, salicylic acid, succinic acid, sulfuric acid and tartaric acid. Examples for cations and bases forming a pharmaceutically acceptable salt with a parent compound containing an acidic moiety include Na+, K+, Ca2+, Mg2+, NH4+, L-arginine, 2,2′-iminobisethanol, L-lysine, N-methyl-D-glucamine or tris(hydroxymethyl)-amino-methane. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficient amount of the appropriate base or acid in water or in an organic diluent like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile, or a mixture thereof. Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention (e.g. trifluoroacetate salts,) also comprise a part of the present invention. BIOLOGICAL ASSAYS Evaluation of TRPA1 Activity Assay A: TRPA1 Assay The activity of the compounds of the invention may be demonstrated using the following in vitro TRPA1 cell assay: Method: A human HEK293 cell line over-expressing the human TRPA1 ion channel (Perkin Elmer, Product No. AX-004-PCL) is used as a test system for compound efficacy and potency. Compound activity is determined by measuring the effect of compounds on intracellular calcium concentration induced by AITC (Allylisothiocyanat) agonism in a FLIPRtetra system (Molecular Devices). Cell Culture: The cells are obtained as frozen cells in cryo-vials and stored until use at −150° C. Cells are grown in culture medium (MEM/EBSS medium with 10% FCS and 0.4 mg/ML Geneticin). It is important that density does not exceed 90% confluence. For sub-culturing cells are detached from flasks by Versene. At the day before the assay, cells are detached, washed twice with medium (MEM/EBSS medium with 10% FCS) and 20000 cells in 20 μl/well are seeded to Poly D-Lysin biocoated 384-well plates (black, clear bottom, Cat. 356697) from Corning. Plates are incubated for 24 hours at 37° C./5% CO2 before use in the assay. Compound Preparation The test compounds are dissolved in 100% DMSO at a concentration of 10 mM and in a first step diluted in DMSO to a concentration of 5 mM, followed by serial dilution steps in 100% DMSO. Dilution factor and number of dilution steps may vary according to needs. Typically 8 different concentrations by 1:5 dilutions are prepared, further intermediate dilutions (1:20) of the substances are carried out with HBSS/HEPES buffer (1×HEPES, Cat. 14065 from Gibco, 20 mM HEPES, Cat. 83264 from SIGMA, 0.1% BSA Cat. 11926 from Invitrogen, pH 7.4 FLIPR Assay: At the assay day cells are washed 3× with assay puffer, 20 μL buffer remaining in the wells after washing. 10 μL Ca6 kit (Cat. R8191 MolecularDevices) loading buffer in HBSS/HEPES is added to the cells and the plates are incubated with lid for 120 minutes at 37°/5% CO2. 10 μL of compound or controls in HBSS/HEPES buffer/5% DMSO from the intermediate dilution plate are carefully added to the wells. Luminescence (indicating the calcium influx or release) is read on the FLIPRtetra device for 10 minutes to monitor the compound induced effects (e.g. agonism). Finally 10 μL of the agonist AITC 50 μM dissolved in HBSS/HEPES buffer/0.05% DMSO (final concentration 10 μM) is added to the wells followed by an additional read on the FLIPRtetra device for 10 minutes. The area under the signal curve (AUC) after AITC addition is used for IC50/% inhibition calculations Data evaluation and calculation: Each assay microtiter plate contains wells with vehicle (1% DMSO) controls instead of compound as controls for AITC induced luminescence (100% CTL; high controls) and wells with vehicle controls without AITC as controls for non-specific changes in luminescence (0% CTL; low controls). The analysis of the data is performed by the calculation of the area under signal curve of the individual wells. Based on this values the % value for the measurement of each substance concentration is calculated (AUC(sample)−AUC(low))*100/(AUC(high)−AUC(low)) using MegaLab software (in house development). The IC50 values are calculated from the % control values using MegaLab software. Calculation: [y=(a−d)/(1+(x/c){circumflex over ( )}b)+d], a=low value, d=high value; x=conc M; c=IC50 M; b=hill; y=% ctrl TABLE 1Biological data for compounds of the inventionas obtained in Assay AhTRPA1 IC50Example[nM]1382638415533657784 TABLE 2Biological data for prior art compounds (examples 53,72, 73, 86, 90 in WO2017/060488) as obtained in Assay A.Example inhTRPA1 IC50WO2017/060488[nM]53367214732886679041 TABLE 3Biological data for prior art compounds (example31 in L. Schenkel, et al., J. Med. Chem. 2016, 59, 2794-2809)as obtained in Assay A.Example in Med. Chem.hTRPA1 IC502016, 59, 2794-2809[nM]3152 Evaluation of Microsomal Clearance Assay B: Microsomal Clearance: The metabolic degradation of the test compound is assayed at 37° C. with pooled liver microsomes. The final incubation volume of 100 μl per time point contains TRIS buffer pH 7.6 at RT (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/ml) and the test compound at a final concentration of 1 μM. Following a short preincubation period at 37° C., the reactions are initiated by addition of beta-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH, 1 mM) and terminated by transferring an aliquot into solvent after different time points (0, 5, 15, 30, 60 min). Additionally, the NADPH-independent degradation is monitored in incubations without NADPH, terminated at the last time point. The [%] remaining test compound after NADPH independent incubation is reflected by the parameter c(control) (metabolic stability). The quenched incubations are pelleted by centrifugation (10000 g, 5 min). An aliquot of the supernatant is assayed by LC-MS/MS for the amount of parent compound. The half-life (t½ INVITRO) is determined by the slope of the semilogarithmic plot of the concentration-time profile. The intrinsic clearance (CL_INTRINSIC) is calculated by considering the amount of protein in the incubation: CL_INTRINSIC [μl/min/mg protein]=(Ln 2/(half-life [min]*protein content [mg/ml]))*1000 CL_INTRINSIC_INVIVO [ml/min/kg]=(CL_INTRINSIC [μL/min/mg protein]×MPPGL [mg protein/g liver]×liver factor [g/kg bodyweight])/1000 Qh[%]=CL[ml/min/kg]/hepatic blood flow [ml/min/kg]) Hepatocellularity, human: 120×10e6 cells/g liver Liver factor, human: 25.7 g/kg bodyweight Blood flow, human: 21 ml/(min×kg) TABLE 4Biological data for compounds of the invention as obtained in Assay BExamplehuman LM [% Qh]1<232<233434<235<236247<23 TABLE 5Biological data for prior art compounds (examples 53, 72, 73,86, 90 in WO2017/060488) as obtained in Assay B.Example inWO2017/060488human LM [% Qh]53<237230733886<239039 TABLE 6Biological data for prior art compounds (example31 in L. Schenkel, et al., J. Med. Chem. 2016, 59, 2794-2809)as obtained in Assay B.Example in Med. Chem.2016, 59, 2794-2809human LM [% Qh]31<23 Evaluation of Hepatocyte Clearance Assay C: Hepatocyte Clearance The metabolic degradation of the test compound is assayed in a hepatocyte suspension. Hepatocytes (cryopreserved) are incubated in Dulbecco's modified eagle medium (supplemented with 3.5 μg glucagon/500 mL, 2.5 mg insulin/500 mL and 3.75 mg/500 mL hydrocortison) containing 5% species serum. Following a 30 min preincubation in an incubator (37° C., 10% CO2) 5 μl of test compound solution (80 μM; from 2 mM in DMSO stock solution diluted 1:25 with medium) are added into 395 μl hepatocyte suspension (cell density in the range 0.25-5 Mio cells/mL depending on the species, typically 1 Mio cells/mL; final concentration of test compound 1 μM, final DMSO concentration 0.05%). The cells are incubated for six hours (incubator, orbital shaker) and samples (25 μl) are taken at 0, 0.5, 1, 2, 4 and 6 hours. Samples are transferred into acetonitrile and pelleted by centrifugation (5 min). The supernatant is transferred to a new 96-deepwell plate, evaporated under nitrogen and resuspended. Decline of parent compound is analyzed by HPLC-MS/MS CLint is calculated as follows CL_INTRINSIC=Dose/AUC=(CO/CD)/(AUD+clast/k)×1000/60. C0: initial concentration in the incubation [μM], CD: cell density of vital cells [10e6 cells/mL], AUD: area under the data [μM×h], clast: concentration of last data point [μM], k: slope of the regression line for parent decline [h−1]. The calculated in vitro hepatic intrinsic clearance can be scaled up to the intrinsic in vivo hepatic Clearance and used to predict hepatic in vivo blood clearance (CL) by the use of a liver model (well stirred model). CL_INTRINSIC_INVIVO [ml/min/kg]=(CL_INTRINSIC [μL/min/10e6 cells]×hepato-cellularity [10e6 cells/g liver]×liver factor [g/kg bodyweight])/1000 CL[ml/min/kg]=CL_INTRINSIC_INVIVO [ml/min/kg]×hepatic blood flow [ml/min/kg]/(CL_INTRINSIC_INVIVO [ml/min/kg]+hepatic blood flow [ml/min/kg]) Qh[%]=CL[ml/min/kg]/hepatic blood flow [ml/min/kg]) Hepatocellularity, human: 120×10e6 cells/g liver Liver factor, human: 25.7 g/kg bodyweight Blood flow, human: 21 ml/(min×kg) TABLE 7Biological data for compounds of the invention as obtained in Assay Chuman HepatocytesExample[% Qh]11522431742151561778 TABLE 8Biological data for prior artcompounds (examples 53, 72,73, 86, 90 in WO2017/060488)as obtained in Assay C.Example inhumanWO2017/Hepatocytes060488[% Qh]53257250733686129061 TABLE 9Biological data for prior artcompounds (example 31 inL. Schenkel, et al., J. Med.Chem. 2016, 59, 2794-2809)as obtained in Assay C.Example inMed. Chem.human2016, 59,Hepatocytes2794-2809[% Qh]3173 Evaluation of Permeability Caco-2 cells (1-2×105cells/1 cm2area) are seeded on filter inserts (Costar transwell polycarbonate or PET filters, 0.4 μm pore size) and cultured (DMEM) for 10 to 25 days. Compounds are dissolved in appropriate solvent (like DMSO, 1-20 mM stock solutions). Stock solutions are diluted with HTP-4 buffer (128.13 mM NaCl, 5.36 mM KCl, 1 mM MgSO4, 1.8 mM CaCl2, 4.17 mM NaHCO3, 1.19 mM Na2HPO4×7H2O, 0.41 mM NaH2PO4×H2O, 15 mM HEPES, 20 mM glucose, 0.25% BSA, pH 7.2) to prepare the transport solutions (0.1-300 μM compound, final DMSO<=0.5%). The transport solution (TL) is applied to the apical or basolateral donor side for measuring A-B or B-A permeability (3 filter replicates), respectively. Samples are collected at the start and end of experiment from the donor and at various time intervals for up to 2 hours also from the receiver side for concentration measurement by HPLC-MS/MS or scintillation counting. Sampled receiver volumes are replaced with fresh receiver solution. Evaluation of Plasma Protein Binding This equilibrium dialysis (ED) technique is used to determine the approximate in vitro fractional binding of test compounds to plasma proteins. Dianorm Teflon dialysis cells (micro 0.2) are used. Each cell consists of a donor and an acceptor chamber, separated by an ultrathin semipermeable membrane with a 5 kDa molecular weight cutoff. Stock solutions for each test compound are prepared in DMSO at 1 mM and diluted to a final concentration of 1.0 μM. The subsequent dialysis solutions are prepared in pooled human or rat plasma (with NaEDTA) from male and female donors. Aliquots of 200 μL dialysis buffer (100 mM potassium phosphate, pH 7.4) are dispensed into the buffer chamber. Aliquots of 200 μL test compound dialysis solution are dispensed into the plasma chambers. Incubation is carried out for 2 hours under rotation at 37° C. At the end of the dialysis period, the dialysate is transferred into reaction tubes. The tubes for the buffer fraction contain 0.2 mL ACN/water (80/20). Aliquots of 25 μL of the plasma dialysate are transferred into deep well plates and mixed with 25 μL ACN/water (80/20), 25 μL buffer, 25 μL calibration solution and 25 μL Internal Standard solution. Protein precipitation is done by adding 200 μL ACN. Aliquots of 50 μL of the buffer dialysate are transferred into deep well plates and mixed with 25 μL blank plasma, 25 μL Internal Standard solution and 200 μL ACN. Samples are measured on HPLC-MS/MS-Systems and evaluated with Analyst-Software. Percent bound is calculated with the formula: % bound=(plasma concentration−buffer concentration/plasma 30 concentration)×100. Evaluation of Solubility Saturated solutions are prepared in well plates (format depends on robot) by adding an appropriate volume of selected aqueous media (typically in the range of 0.25-1.5 ml) into each well which contains a known quantity of solid drug substance (typically in the range 0.5-5.0 mg). The wells are shaken or stirred for a predefined time period (typically in a range of 2-24 h) and than filtered using appropriate filter membranes (typically PTFE-filters with 0.45 μm pore size). Filter absorption is avoided by discarding the first few drops of filtrate. The amount of dissolved drug substance is determined by UV spectroscopy. In addition the pH of the aqueous saturated solution is measured using a glass-electrode pH meter. Evaluation of Pharmacokinetic Characteristics The test compound is administered either intravenously or orally to the respective test species. Blood samples are taken at several time points post application of the test compound, anticoagulated and centrifuged. The concentration of analytes—the administered compound and/or metabolites—are quantified in the plasma samples. PK parameters are calculated using non compartment methods. AUC and Cmax are normalized to a dose of 1 μmol/kg. Evaluation of Metabolism in Human Hepatocytes In Vitro The metabolic pathway of a test compound is investigated using primary human hepatocytes in suspension. After recovery from cryopreservation, human hepatocytes are incubated in Dulbecco's modified eagle medium containing 5% human serum and supplemented with 3.5 μg glucagon/500 ml, 2.5 mg insulin/500 ml and 3.75 mg/500 ml hydrocortisone. Following a 30 min preincubation in a cell culture incubator (37° C., 10% CO2), test compound solution is spiked into the hepatocyte suspension to obtain a final cell density of 1.0*106to 4.0*106cells/ml (depending on the metabolic turnover rate of the compound observed with primary human hepatocytes), a final test compound concentration of 10 μM, and a final DMSO concentration of 0.05%. The cells are incubated for six hours in a cell culture incubator on a horizontal shaker, and samples are removed from the incubation after 0, 0.5, 1, 2, 4 or 6 hours, depending on the metabolic turnover rate. Samples are quenched with acetonitrile and pelleted by centrifugation. The supernatant is transferred to a 96-deepwell plate, evaporated under nitrogen and resuspended prior to bioanalysis by liquid chromatography-high resolution mass spectrometry for identification of putative metabolites. The structures are assigned tentatively based on Fourier-Transform-MSndata. Metabolites are reported as percentage of the parent in human hepatocyte incubation with a threshold of ≥4%. METHOD OF TREATMENT The present invention is directed to compounds of general formula 1 which are useful in the prevention and/or treatment of a disease and/or condition associated with or modulated by TRPA1 activity, including but not limited to the treatment and/or prevention of fibrotic disease, inflammatory and immunoregulatory disorders, respiratory or gastrointestinal diseases or complaints, ophthalmic diseases, inflammatory diseases of the joints and inflammatory diseases of the nasopharynx, eyes, and skin and pain and neurological disorders. Said disorders, diseases and complaints include cough, idiopathic pulmonary fibrosis, other pulmonary interstitial diseases and other fibrotic, asthma or allergic diseases, eosinophilic diseases, chronic obstructive pulmonary disease, as well as inflammatory and immunoregulatory disorders, such as rheumatoid arthritis and atherosclerosis, as well as pain and neurological disorders, such as acute pain, surgical pain, chronic pain and depression and bladder disorders. The compounds of general formula 1 are useful for the prevention and/or treatment of:(1) Cough such as chronic idiopathic cough or chronic refractory cough, cough associated with asthma, COPD, lung cancer, post-viral infection and idiopathic pulmonary fibrosis and other pulmonary interstitial diseases.(2) Pulmonary fibrotic diseases such as pneumonitis or interstitial pneumonitis associated with collagenosis, e.g. lupus erythematodes, systemic scleroderma, rheumatoid arthritis, polymyositis and dermatomysitis, idiopathic interstitial pneumonias, such as pulmonary lung fibrosis (IPF), non-specific interstitial pneumonia, respiratory bronchiolitis associated interstitial lung disease, desquamative interstitial pneumonia, cryptogenic orgainizing pneumonia, acute interstitial pneumonia and lymphocytic interstitial pneumonia, lymangioleiomyomatosis, pulmonary alveolar proteinosis, Langerhan's cell histiocytosis, pleural parenchymal fibroelastosis, interstitial lung diseases of known cause, such as interstitial pneumonitis as a result of occupational exposures such as asbestosis, silicosis, miners lung (coal dust), farmers lung (hay and mould), Pidgeon fanciers lung (birds) or other occupational airbourne triggers such as metal dust or mycobacteria, or as a result of treatment such as radiation, methotrexate, amiodarone, nitrofurantoin or chemotherapeutics, or for granulomatous disease, such as granulomatosis with polyangitis, Churg-Strauss syndrome, sarcoidosis, hypersensitivity pneumonitis, or interstitial pneumonitis caused by different origins, e.g. aspiration, inhalation of toxic gases, vapors, bronchitis or pneumonitis or interstitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy, M. boeck or sarcoidosis, granulomatosis, cystic fibrosis or mucoviscidosis, alpha-1-antitrypsin deficiency, acute lung injury as a result of Covid-19/SARS-Cov-2 infection or pulmonary fibrosis secondary to Covid-19/SARS-Cov-2 infection.(3) Other fibrotic diseases such as hepatic bridging fibrosis, liver cirrhosis, non-alcoholic steatohepatitis (NASH), atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, glial scar, arterial stiffness, arthrofibrosis, Dupuytren's contracture, keloid, scleroderma/systemic sclerosis, mediastinal fibrosis, myelofibrosis, Peyronie's disease, nephrogenic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis.(4) Inflammatory, auto-immune or allergic diseases and conditions such as allergic or non-allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal polyposis, chronic rhinosinusitis, acute rhinosinusitis, asthma, pediatric asthma, allergic bronchitis, alveolitis, hyperreactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory distress syndrome, bronchial and pulmonary edema, bronchitis or pneumonitis, eosinophilic cellulites (e.g., Well's syndrome), eosinophilic pneumonias (e.g., Loeffler's syndrome, chronic eosinophilic pneumonia), eosinophilic fasciitis (e.g., Shulman's syndrome), delayed-type hypersensitivity, non-allergic asthma; exercise induced bronchoconstriction; chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bronchitis, cough, pulmonary emphysema; systemic anaphylaxis or hypersensitivity responses, drug allergies (e.g., to penicillin, cephalosporin), eosinophiliamyalgia syndrome due to the ingestion of contaminated tryptophane, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, Graves' disease, Sjogren's syndrome psoriatic arthritis, multiple sclerosis, systemic lupus erythematosus, myasthenia gravis, immune thrombocytopenia (adult ITP, neonatal thrombocytopenia, pediatric ITP), immune hemolytic anemia (auto-immune and drug induced), Evans syndrome (platelet and red cell immune cytopaenias), Rh disease of the newborn, Goodpasture's syndrome (anti-GBM disease), Celiac, autoimmune cardio-myopathy juvenile onset diabetes; glomerulonephritis, autoimmune thyroiditis, Behcet's disease; graft rejection (e.g., in transplantation), including allograft rejection or graftversus-host disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloarthropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and inflammatory dermatoses such as an dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria; vasculitis (e.g., necrotizing, cutaneous, and hypersensitivity vasculitis); erythema nodosum; eosinophilic myositis, eosinophilic fasciitis, cancers with leukocyte infiltration of the skin or organs; ophthalmic diseases such as age related macular degeneration, diabetic retinopathy and diabetic macular edema, keratitis, eosinophilic keratitis, keratoconjunctivitis, vernal keratoconjunctivitis, scarring, anterior segment scarring, blepharitis, blepharoconjunctivitis, bullous disorders, cicatricial pemphigoid, conjunctival melanoma, papillary conjunctivitis, dry eye, episcleritis, glaucoma, gliosis, Granuloma annulare, Graves' ophthalmopathy, intraocular melanoma, Pinguecula, proliferative vitreoretinopathy, pterygia, scleritis, uveitis, acute gout flares, gout or osteoarthritis.(5) Pain such as chronic idiopathic pain syndrome, neuropathic pain, dysesthesia, allodynia, migraine, dental pain and post-surgical pain.(6) Depression, anxiousness, diabetic neuropathy and bladder disorders such as bladder outlet obstruction, overactive bladder, cystitis; myocardial reperfusion injury or brain ischaemia injury. Accordingly, the present invention relates to a compound of general formula 1 for use as a medicament. Furthermore, the present invention relates to the use of a compound of general formula 1 for the treatment and/or prevention of a disease and/or condition associated with or modulated by TRPA1 activity. Furthermore, the present invention relates to the use of a compound of general formula 1 for the treatment and/or prevention of fibrotic disease, inflammatory and immunoregulatory disorders, respiratory or gastrointestinal diseases or complaints, ophthalmic diseases, inflammatory diseases of the joints and inflammatory diseases of the nasopharynx, eyes, and skin, pain and neurological disorders. Said disorders, diseases and complaints include cough, idiopathic pulmonary fibrosis, other pulmonary interstitial diseases and other fibrotic, asthma or allergic diseases, eosinophilic diseases, chronic obstructive pulmonary disease, as well as inflammatory and immunoregulatory disorders, such as rheumatoid arthritis and atherosclerosis, as well as pain and neurological disorders, such as acute pain, surgical pain, chronic pain and depression and bladder disorders. Furthermore, the present invention relates to the use of a compound of general formula 1 for the treatment and/or prevention of:(1) Cough such as chronic idiopathic cough or chronic refractory cough, cough associated with asthma, COPD, lung cancer, post-viral infection and idiopathic pulmonary fibrosis and other pulmonary interstitial diseases.(2) Pulmonary fibrotic diseases such as pneumonitis or interstitial pneumonitis associated with collagenosis, e.g. lupus erythematodes, systemic scleroderma, rheumatoid arthritis, polymyositis and dermatomysitis, idiopathic interstitial pneumonias, such as pulmonary lung fibrosis (IPF), non-specific interstitial pneumonia, respiratory bronchiolitis associated interstitial lung disease, desquamative interstitial pneumonia, cryptogenic orgainizing pneumonia, acute interstitial pneumonia and lymphocytic interstitial pneumonia, lymangioleiomyomatosis, pulmonary alveolar proteinosis, Langerhan's cell histiocytosis, pleural parenchymal fibroelastosis, interstitial lung diseases of known cause, such as interstitial pneumonitis as a result of occupational exposures such as asbestosis, silicosis, miners lung (coal dust), farmers lung (hay and mould), Pidgeon fanciers lung (birds) or other occupational airbourne triggers such as metal dust or mycobacteria, or as a result of treatment such as radiation, methotrexate, amiodarone, nitrofurantoin or chemotherapeutics, or for granulomatous disease, such as granulomatosis with polyangitis, Churg-Strauss syndrome, sarcoidosis, hypersensitivity pneumonitis, or interstitial pneumonitis caused by different origins, e.g. aspiration, inhalation of toxic gases, vapors, bronchitis or pneumonitis or interstitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy, M. boeck or sarcoidosis, granulomatosis, cystic fibrosis or mucoviscidosis, alpha-1-antitrypsin deficiency, acute lung injury as a result of Covid-19/SARS-Cov-2 infection or pulmonary fibrosis secondary to Covid-19/SARS-Cov-2 infection.(3) Other fibrotic diseases such as hepatic bridging fibrosis, liver cirrhosis, non-alcoholic steatohepatitis (NASH), atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, glial scar, arterial stiffness, arthrofibrosis, Dupuytren's contracture, keloid, scleroderma/systemic sclerosis, mediastinal fibrosis, myelofibrosis, Peyronie's disease, nephrogenic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis.(4) Inflammatory, auto-immune or allergic diseases and conditions such as allergic or non-allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal polyposis, chronic rhinosinusitis, acute rhinosinusitis, asthma, pediatric asthma, allergic bronchitis, alveolitis, hyperreactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory distress syndrome, bronchial and pulmonary edema, bronchitis or pneumonitis, eosinophilic cellulites (e.g., Well's syndrome), eosinophilic pneumonias (e.g., Loeffler's syndrome, chronic eosinophilic pneumonia), eosinophilic fasciitis (e.g., Shulman's syndrome), delayed-type hypersensitivity, non-allergic asthma; exercise induced bronchoconstriction; chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bronchitis, cough, pulmonary emphysema; systemic anaphylaxis or hypersensitivity responses, drug allergies (e.g., to penicillin, cephalosporin), eosinophiliamyalgia syndrome due to the ingestion of contaminated tryptophane, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, Graves' disease, Sjogren's syndrome psoriatic arthritis, multiple sclerosis, systemic lupus erythematosus, myasthenia gravis, immune thrombocytopenia (adult ITP, neonatal thrombocytopenia, pediatric ITP), immune hemolytic anemia (auto-immune and drug induced), Evans syndrome (platelet and red cell immune cytopaenias), Rh disease of the newborn, Goodpasture's syndrome (anti-GBM disease), Celiac, autoimmune cardio-myopathy juvenile onset diabetes; glomerulonephritis, autoimmune thyroiditis, Behcet's disease; graft rejection (e.g., in transplantation), including allograft rejection or graftversus-host disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloarthropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and inflammatory dermatoses such as an dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria; vasculitis (e.g., necrotizing, cutaneous, and hypersensitivity vasculitis); erythema nodosum; eosinophilic myositis, eosinophilic fasciitis, cancers with leukocyte infiltration of the skin or organs; ophthalmic diseases such as age related macular degeneration, diabetic retinopathy and diabetic macular edema, keratitis, eosinophilic keratitis, keratoconjunctivitis, vernal keratoconjunctivitis, scarring, anterior segment scarring, blepharitis, blepharoconjunctivitis, bullous disorders, cicatricial pemphigoid, conjunctival melanoma, papillary conjunctivitis, dry eye, episcleritis, glaucoma, gliosis, Granuloma annulare, Graves' ophthalmopathy, intraocular melanoma, Pinguecula, proliferative vitreoretinopathy, pterygia, scleritis, uveitis, acute gout flares, gout or osteoarthritis.(5) Pain such as chronic idiopathic pain syndrome, neuropathic pain, dysesthesia, allodynia, migraine, dental pain and post-surgical pain.(6) Depression, anxiousness, diabetic neuropathy and bladder disorders such as bladder outlet obstruction, overactive bladder, cystitis; myocardial reperfusion injury or brain ischaemia injury. In a further aspect the present invention relates to a compound of general formula 1 for use in the treatment and/or prevention of above mentioned diseases and conditions. In a further aspect the present invention relates to the use of a compound of general formula 1 for the preparation of a medicament for the treatment and/or prevention of above mentioned diseases and conditions. In a further aspect of the present invention the present invention relates to methods for the treatment or prevention of above mentioned diseases and conditions, which method comprises the administration of an effective amount of a compound of general formula 1 to a human being. Combination Therapy The compounds of the invention may further be combined with one or more, preferably one additional therapeutic agent. According to one embodiment the additional therapeutic agent is selected from the group of therapeutic agents useful in the treatment of diseases or conditions described hereinbefore, in particular associated with fibrotic diseases, inflammatory and immunoregulatory disorders, respiratory or gastrointestinal diseases or complaints, inflammatory diseases of the joints or of the nasopharynx, eyes, and skin or conditions such as for example cough, idiopathic pulmonary fibrosis, other pulmonary interstitial diseases, asthma or allergic diseases, eosinophilic diseases, chronic obstructive pulmonary disease, atopic dermatitis as well as autoimmune pathologies, such as rheumatoid arthritis and atherosclerosis, or therapeutic agents useful for the treatment of ophthalmic diseases, pain and depression. Additional therapeutic agents that are suitable for such combinations include in particular those, which, for example, potentiate the therapeutic effect of one or more active substances with respect to one of the indications mentioned and/or allow the dosage of one or more active substances to be reduced. Therefore, a compound of the invention may be combined with one or more additional therapeutic agents selected from the group consisting of antifibrotic agents, anti-tussive agents, anti-inflammatory agents, anti-atopic dermatitis agents, analgesics, anti-convulsants, anxiolytics, sedatives, skeletal muscle relaxants or anti-depressants. Antifibrotic agents are for example nintedanib, pirfenidone, phosphodiesterase-IV (PDE4) inhibitors such as roflumilast, autotaxin inhibitors such as GLPG-1690 or BBT-877; connective tissue growth factor (CTGF) blocking antibodies such as Pamrevlumab; B-cell activating factor receptor (BAFF-R) blocking antibodies such as Lanalumab; alpha-V/beta-6 blocking inhibitors such as BG-00011/STX-100, recombinant pentraxin-2 (PTX-2) such as PRM-151; c-Jun N-terminal kinase (JNK) inhibitors such as CC-90001; galectin-3 inhibitors such as TD-139; G-protein coupled receptor 84 (GPR84) inhibitors such as GLPG-1205; G-protein coupled receptor 84/G-protein coupled receptor 40 dual inhibitors such as PBI-4050; Rho Associated Coiled-Coil Containing Protein Kinase 2 (ROCK2) inhibitors such as KD-025; heat shock protein 47 (HSP47) small interfering RNA such as BMS-986263/ND-L02-s0201; Wnt pathway inhibitor such as SM-04646; LD4/PDE3/4 inhibitors such as Tipelukast; recombinant immuno-modulatory domains of histidyl tRNA synthetase (HARS) such as ATYR-1923; prostaglandin synthase inhibitors such as ZL-2102/SAR-191801; 15-hydroxy-eicosapentaenoic acid (15-HEPE e.g. DS-102); Lysyl Oxidase Like 2 (LOXL2) inhibitors such as PAT-1251, PXS-5382/PXS-5338; phosphoinositide 3-kinases (PI3K)/mammalian target of rapamycin (mTOR) dual inhibitors such as HEC-68498; calpain inhibitors such as BLD-2660; mitogen-activated protein kinase kinase kinase (MAP3K19) inhibitors such as MG-S-2525; chitinase inhibitors such as OATD-01; mitogen-activated protein kinase-activated protein kinase 2 (MAPKAPK2) inhibitors such as MMI-0100; transforming growth factor beta 1 (TGF-beta1) small interfering RNA such as TRK250/BNC-1021; or lysophosphatidic acid receptor antagonists such as BMS-986278. Anti-tussive agents are, for example, purinoceptor 3 (P2X3) receptor antagonists such as gefapixant, S-600918, BAY-1817080, or BLU-5937; neurokinin 1 (NK-1) receptor antagonist such as Orvepitant, Aprepitant; nicotinic acetylcholine receptor alpha 7 subunit stimulator such as ATA-101/bradanicline; codeine, gabapentin, pregablin, or azithromycin. Anti-inflammatory agents are, for example, corticosteroids such as prednisolone or dexamethasone; cyclo-oxygenase-2 (COX2) inhibitors such as celecoxib, rofecoxib, parecoxib, valdecoxib, deracoxib, etoricoxib or lumiracoxib; prostaglandin E2 antagonists; leukotriene B4 antagonists; leukotriene D4 antagonists such as monteleukast; 5-lipoxygenase inhibitors; or other nonsteroidal anti-inflammatory agents (NSAIDs) such as aspirin, diclofenac, diflunisal, etodolac, ibuprofen or indomethacin. Anti-atopic dermatitis agents are, for example, cyclosporin, methotrexate, mycophenolate mofetil, azathioprine, phosphodiesterase inhibitors (e.g. apremilast, crisaborole), Janus Associated Kinase (JAK) inhibitors (e.g. tofacitinib), neutralizing antibodies against IL-4/IL-13 (e.g. dupilamab), IL-13 (e.g. lebrikizumab, tralokinumab) and IL-31 (nemolizumab). Analgesics are, for example, of the opioid type, such as morphine, oxymorphine, levopanol, oxycodon, propoxyphene, nalmefene, fentanyl, hydrocondon, hydromorphone, meripidine, methadone, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine, pentazocine; or of the non-opioid type, such as acetophenamine. Anti-depressants are, for example, tricyclic anti-depressants such as amitriptyline, clomipramine, despramine, doxepin, desipramine, imipramine, nortriptyline; selective serotonin reuptake inhibitor anti-depressants (SSRIs) such as fluoxetine, paroxetine, sertraline, citalopram, escitalopram; norepinephrine reuptake inhibitor anti-depressants (SNRIs) such as maprotiline, lofepramine, mirtazapine, oxaprotiline, fezolamine, tomoxetine, mianserin, buproprion, hydroxybuproprion, nomifensine, viloxazine; dual serotonin-norepinephrine reuptake inhibitor anti-depressants (SNRIs) such as duloxetine, venlafaxine, desvenlafaxine, levomilnacipran; atypical antidepressants such as trazodone, mirtazapine, vortioxetine, vilazodone, bupropion; or monoamine oxidase inhibitor anti-depressants (MAOIs) such as tranylcypromine, phenelzine, or isocarboxazid. Anxiolytics are, for example, benzodiazepines such as alprazolam, bromazepam, chlordiazepoxide, clonazepam, clorazepate, diazepam, flurazepam, lorazepam, oxazepam, temazepam, triazolam, or tofisopam; or they are nonbenzodiazepine hypnoticssuch as eszopiclone, zaleplon, zolpidem, or zopiclone; or they are carbamates e.g. meprobamate, carisoprodol, tybamate, or lorbamate; or they are antihistamines such as hydroxyzine, chlor-pheniramine or diphenhydramine. Sedatives are, for example, barbiturate sedatives, such as amobarbital, aprobarbital, buta-barbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital, secobarbital, talbutal, theamylal, or thiopental; or they are non-barbiturate sedatives such as glutethimide, meprobamate, methaqualone or dichloalphenazone. Skeletal muscle relaxants are, for example, baclofen, meprobamate, carisoprodol, cyclobenzaprine, metaxalone, methocarbamol, tizanidine, chlorzoxazone or orphenadrine. Other suitable combination partners are inhibitors of Acetylcholinesterase inhibitors such as donepezil; 5-HT-3 anatgonists such as ondansetron; metabotropic glutamate receptor antagonists; antiarrhythmics such as mexiletine or phenytoin; or NMDA receptor antagonists. Further suitable combination partners are incontinence medications, for example, anticholinergics such as oxybutynin, tolterodine, darifenacin, fesoterodine, solifenacin or trospium; or they are bladder muscle relaxants such as mirabegron; or they are alpha blockers such as tamsulosin, alfuzosin, silodosin, doxazosin or terazosin. The dosage for the combination partners mentioned above is usually ⅕ of the lowest dose normally recommended up to 1/1 of the normally recommended dose. Therefore, in another aspect, this invention relates to the use of a compound according to the invention in combination with one or more additional therapeutic agents described hereinbefore and hereinafter for the treatment of diseases or conditions which may be affected or which are mediated by TRPA1, in particular diseases or conditions as described hereinbefore and hereinafter. In a further aspect this invention relates to a method for treating a disease or condition which can be influenced by the inhibition of TRPA1 in a patient that includes the step of administering to the patient in need of such treatment a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with a therapeutically effective amount of one or more additional therapeutic agents. In a further aspect this invention relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with one or more additional therapeutic agents for the treatment of diseases or conditions which can be influenced by the inhibition of TRPA1 in a patient in need thereof. In yet another aspect the present invention relates to a method for the treatment of a disease or condition mediated by TRPA1 activity in a patient that includes the step of administering to the patient, preferably a human, in need of such treatment a therapeutically effective amount of a compound of the present invention in combination with a therapeutically effective amount of one or more additional therapeutic agents described in hereinbefore and hereinafter. The use of the compound according to the invention in combination with the additional therapeutic agent may take place simultaneously or at staggered times. The compound according to the invention and the one or more additional therapeutic agents may both be present together in one formulation, for example a tablet or capsule, or separately in two identical or different formulations, for example as a so-called kit-of-parts. Consequently, in another aspect, this invention relates to a pharmaceutical composition that comprises a compound according to the invention and one or more additional therapeutic agents described hereinbefore and hereinafter, optionally together with one or more inert carriers and/or diluents. In yet another aspect the present invention relates to the use of a compound according to the invention in a cough-measuring device. Other features and advantages of the present invention will become apparent from the following more detailed examples which illustrate, by way of example, the principles of the invention. Preparation The compounds according to the present invention and their intermediates may be obtained using methods of synthesis which are known to the one skilled in the art and described in the literature of organic synthesis. Preferably, the compounds are obtained in analogous fashion to the methods of preparation explained more fully hereinafter, in particular as described in the experimental section. In some cases, the order in carrying out the reaction steps may be varied. Variants of the reaction methods that are known to the one skilled in the art but not described in detail here may also be used. The general processes for preparing the compounds according to the invention will become apparent to the one skilled in the art studying the following schemes. Any functional groups in the starting materials or intermediates may be protected using conventional protecting groups. These protecting groups may be cleaved again at a suitable stage within the reaction sequence using methods familiar to the one skilled in the art. The compounds according to the invention are prepared by the methods of synthesis described hereinafter in which the substituents of the general formulae have the meanings given herein before. These methods are intended as an illustration of the invention without restricting its subject matter and the scope of the compounds claimed to these examples. Where the preparation of starting compounds is not described, they are commercially obtainable or may be prepared analogously to known compounds or methods described herein. Substances described in the literature are prepared according to the published methods of synthesis. Abbreviations are as defined in the Examples section. In scheme 1, compounds of formula I can be synthesized via N-alkylation of the intermediate (A) with chloromethylen-oxadiazoles (B) in presence of a base such as potassium carbonate. In scheme 2, uracil derivative (C), CAS: 154942-22-0, can be synthesized from methylurea and 1,3-diethyl 2-(ethoxymethylidene)propanedioate under neat conditions at elevated temperature. Primary amide (A) can be synthesized from ester (C) by stirring with ammonia in a solvent such as water or an alcohol at elevated temperature in a sealed vessel. In scheme 3, alpha-cyano ketones (E), synthesized from carboxylic esters (D), are reduced enantioselectively by using an appropriate catalytic systems using a transition metal complex (of e.g. Ru or Ir) in combination with a chiral ligand (e.g. [I(1S,2S)-2-amino-1,2-diphenylethyl](4-toluenesulfonyl)amido) and a hydrogen source such as formic acid triethylamine complex to provide alcohols (F). Hydroxylamine is added to these alcohols (F) to provide the dihydroxypropanimidamides (G). Ring-closure to chloromethylen-oxadiazoles (B) can be achieved by stirring the reaction mixture together with chloro acetyl chloride in presence of a base such as DIPEA. EXAMPLES Preparation The compounds according to the invention and their intermediates may be obtained using methods of synthesis which are known to the one skilled in the art and described in the literature of organic synthesis for example using methods described in “Comprehensive Organic Transformations”, 2nd Edition, Richard C. Larock, John Wiley & Sons, 2010, and “March's Advanced Organic Chemistry”, 7th Edition, Michael B. Smith, John Wiley & Sons, 2013. Preferably the compounds are obtained analogously to the methods of preparation explained more fully hereinafter, in particular as described in the experimental section. In some cases the sequence adopted in carrying out the reaction schemes may be varied. Variants of these reactions that are known to the skilled artisan but are not described in detail herein may also be used. The general processes for preparing the compounds according to the invention will become apparent to the skilled man on studying the schemes that follow. Starting compounds are commercially available or may be prepared by methods that are described in the literature or herein, or may be prepared in an analogous or similar manner. Before the reaction is carried out, any corresponding functional groups in the starting compounds may be protected using conventional protecting groups. These protecting groups may be cleaved again at a suitable stage within the reaction sequence using methods familiar to the skilled man and described in the literature for example in “Protecting Groups”, 3rd Edition, Philip J. Kocienski, Thieme, 2005, and “Protective Groups in Organic Synthesis”, 4th Edition, Peter G. M. Wuts, Theodora W. Greene, John Wiley & Sons, 2006. The terms “ambient temperature” and “room temperature” are used inter-changeably and designate a temperature of about 20° C., e.g. between 19 and 24° C. Abbreviations ACNacetonitrileAq.aqueous° C.Degree celsiusCyH/CHcyclohexaneconc.concentratedDCMdichloro methaneDCE1,2-DichloroethaneDIPEAN,N-diisopropylethylamineDMAN,N-dimethylacetamideDMFN,N-dimethylformamideDMSOdimethyl sulfoxideESI-MSElectrospray ionisation mass spectrometryEtOAcethyl acetateEtOHethanolexexampleeqequivalentFAformic acidhhourHATU1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium3-oxid hexafluorophosphateHClHydrochloric acidHPLCHigh performance liquid chromatographyK2CO3potassium carbonateLliterMmolarMeOHmethanolMgSO4magnesium sulphateminminutemLmilliliterMTBEtert-butylmethyletherNH3ammoniaNMPN-Methyl-2-pyrrolidonPEpetrol etherRTroom temperature (about 20° C.)sat.saturatedTBTUBenzotriazolyl tetramethyluroniumtetrafluoroborateTEAtriethylamineTFAtrifluoroacetic acidTHFtetrahydrofuran PREPARATION OF INTERMEDIATES Intermediate I Intermediate I.1 (General Route) (3S)-3-(4-chlorophenyl)-3-hydroxypropanenitrile 10.0 g (55.7 mmol) 4-Chlorobenzoylacetonitrile are added to 100 mL ACN under inert atmosphere. 142 mg (0.23 mmol) Chloro([(1S,2S)-2-amino-1,2-diphenylethyl](4-toluenesulfonyl)amido)(mesitylene)ruthenium (II) (CAS 174813-81-1) are added, followed by drop-wise addition of 8.30 mL (19.8 mmol) formic acid triethylamine complex (5:2). After stirring at RT for 3 h, the solvent is removed in vacuo. To the remaining crude mixture is added water and this mixture is extracted two times with EtOAc. The organic layers are combined, dried over MgSO4, filtered, and the solvent is removed in vacuo to provide intermediate I.1. C9H8ClNO(M = 181.6 g/mol)ESI-MS:226 [M + HCOO]−Rt(HPLC):0.81 min (method B) The following compounds are prepared using procedures analogous to those described for intermediate I.1 using appropriate starting materials. As is appreciated by those skilled in the art, these analogous examples may involve variations in general reaction conditions. HPLC retention time[min](method), or1H NMR (300 MHz,Int.Starting materialsStructureESI-MSDMSO-d6) δ ppmI.2184 [M + Na]+0.76 (B)1.3256 [M + H − H2O]+0.84 (B)1.4IV.1266 [M + HCOO]−3.03 (D)1.5—δ 7.98-7.91 (m, 1 H), 7.82-7.77 (m, 1 H), 7.37-7.31 (m, 3 H), 6.56 (d, J = 5.0 Hz, 1 H), 5.28-5.20 (m, 1 H), 3.14-2.94 (m, 2 H)1.6IV.2266 [M + HCOO]−3.12 (D)1.7IV.3—δ 7.63-7.56 (m, 1 H), 7.46 (dd, J = 8.9, 2.7 Hz, 1 H), 7.14 (td, J = 9.2, 2.7 Hz, 1 H), 6.88 (s, 1 H), 6.41 (d, J = 5.5 Hz, 1H), 5.10- 5.01 (m, 1 H), 3.16-2.98 (m, 2 H) Intermediate II Intermediate II.1 (General Route) (3S)-3-(4-chlorophenyl)-N,3-dihydroxypropanimidamidl To 9.82 g (54.1 mmol) (3S)-3-(4-chlorophenyl)-3-hydroxypropanenitrile (intermediate I.1) in 100 mL MeOH are added 8.00 mL (136 mmol) hydroxylamine (50% in water) and the mixture is stirred at 75° C. for 1.5 h. After cooling to RT, all volatiles are removed in vacuo to yield the crude product, which is used without further purification. C9H11ClN2O2(M = 214.6 g/mol)ESI-MS:215 [M + H]+Rt(HPLC):0.60 min (method B) The following compounds are prepared using procedures analogous to those described for intermediate II.1 using appropriate starting materials. As is appreciated by those skilled in the art, these analogous examples may involve variations in general reaction conditions. HPLCretention timeStarting[min]Int.materialsStructureESI-MS(method)II.2I.2195 [M + H]+0.57 (B)II.31.3307 [M + H]+0.71 (B)II.41.4255 [M + H]+2.07 (D)II.51.5237 [M + H]+1.93 (D)II.61.6255 [M + H]+2.18 (D)II.71.7239 [M + H]+1.90 (D) Intermediate III Intermediate III.1 (General Route) (1S)-2-[5-(chloromethyl)-1,2,4-oxadiazol-3-yl]-1-(4-chlorophenyl)ethan-1-ol To 11.2 g (52.4 mmol) of intermediate II.1 in 55 mL NMP are added 10.0 mL (57.8 mmol) DIPEA. The mixture is cooled to 0° C. before 4.60 mL (57.7 mmol) chloroacetyl chloride dissolved in 5 mL NMP are slowly added and the mixture is stirred at 0° C. for 45 min. The mixture is then heated up to 95° C. and stirring is continued for 4 h. After cooling down to RT, 200 mL water are added and the resulting mixture is extracted three times with EtOAc. The organic layers are combined, dried over MgSO4, filtered and the solvent is removed in vacuo. The residue is purified by column chromatography (silica gel; PE/EtOAc, 7/3). C11H10C12N2O2(M=273.1 g/mol) ESI-MS: 271 [M−H]− Rt(HPLC): 0.93 min (method B) The following compounds are prepared using procedures analogous to those described for intermediate III.1 using appropriate starting materials. As is appreciated by those skilled in the art, these analogous examples may involve variations in general reaction conditions. HPLCretention timeStarting[min]Int.materialsStructureESI-MS(method)III.2II.2251 [M − H]−0.92 (C)III.3II.3387 [M + Na]+1.01 (B)III.4II.4311 [M − H]−6.02 (E)III.5II.5295 [M + H]+5.88 (E)III.6II.6311 [M − H]−6.12 (E)III.7II.7295 [M − H]−5.67 (E) Intermediate IV Intermediate IV.1 (General Route) 3-(6-fluoro-1-benzothiophen-2-yl)-3-oxopropanenitrile To 0.63 g (3.00 mmol) methyl 6-fluoro-1-benzothiophene-2-carboxylate in 9.0 mL dry tolu-ene and 0.78 mL dry ACN are added 0.36 g (9.00 mmol) of NaH (60% in oil) under inert atmosphere at RT. The mixture is heated to reflux and stirred for 16 h, cooled to room temperature, poured on ice/water (30 mL), and treated with 2M HCl to reach pH=1. EtOAc (20 mL) is added and the phases are separated. The aqueous phase is extracted once more with EtOAc (20 mL), the combined organic phases are washed with brine (20 mL), and the solvent is removed under reduced pressure. The crude product is purified by silica gel column chromatography using a gradient of EtOAc/hexane (30% to 40%). C11H6FNOS(M = 219.23 g/mol)ESI-MS:218 [M − H]−Rt(HPLC):3.31 min (D) The following compounds are prepared using procedures analogous to those described for intermediate IV.1 using appropriate starting materials. As is appreciated by those skilled in the art, these analogous examples may involve variations in general reaction conditions. HPLCretention time[min]Int.Starting materialsStructureESI-MS(method)IV.2218 [M − H]−3.33 (D)IV.3202 [M − H]−3.08 (D) Intermediate V ethyl 3-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate 500 mg (6.75 mmol) methylurea and 1.36 g (6.75 mmol) 1,3-diethyl 2-(methoxymethylidene) propanedioate are stirred under neat conditions at 120° C. for 2 h, at RT for 17 h, at 100° C. for 66 h, at 150° C. for 17 h, and at 120° C. for 17 h. Subsequently, the mixture is diluted with EtOAc and refluxed. The mixture is slowly cooled to RT and the precipitated intermediate is filtered off. C8H10N2O4(M = 198.2 g/mol)ESI-MS:199 [M + H]+Rt(HPLC):0.24 min (method A) Intermediate VI 3-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide 10.0 g (50.46 mmol) ethyl 3-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate (CAS: 154942-22-0, intermediate V) in 33% aq. ammonia (120 mL) are stirred in a sealed vessel at 100° C. for 10 h. The reaction mixture is cooled to RT and concentrated under reduced pressure. The residue is triturated with ACN, filtered off, and dried at 50° C. to provide intermediate VI. C6H7N3O3(M = 169.1 g/mol)ESI-MS:170 [M + H]+Rt(HPLC):0.48 min (method B) Preparation of Final Compounds Example 1 (General Procedure) 1-({3-[(2S)-2-(4-chlorophenyl)-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl}methyl)-3-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide A mixture of 19 mg (0.11 mmol) intermediate VI, 30 mg (0.11 mmol) intermediate III.1, and 30 mg (0.22 mmol) K2CO3in 1.0 mL DMF is stirred at RT for 1 h. The reaction mixture is filtered and the filtrate is purified by reversed phase HPLC (ACN/H2O gradient, 0.1% TFA) to yield the desired product. C17H16ClN5O5(M = 405.79 g/mol)ESI-MS:406 [M + H]+Rt(HPLC):0.44 min (method A) 1H NMR (400 MHz, DMSO-d6) δ ppm: 2.92-3.07 (m, 2H), 3.23 (s, 3H), 4.96 (dd, J=7.9, 5.8 Hz, 1H), 5.48 (d, J=1.9 Hz, 2H), 7.31-7.40 (m, 4H), 7.65 (d, J=3.3 Hz, 1H), 8.19 (d, J=3.3 Hz, 1H), 8.80 (s, 1H). The following compounds are prepared using procedures analogous to those described for example 1 general procedure, using appropriate starting materials. As is appreciated by those skilled in the art, these analogous examples may involve variations in general reaction conditions. StartingReactionEx.materialsStructureconditions2VI + III.51.05 eq III.5, 2 eq K2CO3, DMF, RT, 2 h3VI + III.41.05 eq III.4, 2 eq K2CO3, DMF, RT, 2 h4VI + III.61.05 eq III.6, 2 eq K2CO3, DMF, RT, 2 h5VI + III.71.05 eq III.7, 2 eq K2CO3, DMF, RT, 3 h6VI + III.31.0 eq III.3, 2 eq K2CO3, DMF, RT, 18 h7VI + III.21.0 eq III.2, 2 eq K2CO3, DMF, RT, 18 h Analytical data for the compounds described in the table above: HPLCretentiontime[min]Ex.ESI-MS(method)1H NMR (400 MHz, DMSO-d6) δ ppm24280.473.14-3.20 (m, 2 H), 3.23 (s, 3 H), 5.31 (t, J = 6.7 Hz,[M + H]+(A)1 H), 5.50 (d, J = 1.7 Hz, 2 H), 7.25 (s, 1 H), 7.32(quind, J = 7.4, 1.4 Hz, 2 H), 7.66 (br d, J = 3.4 Hz,1 H), 7.74 (dd, J = 7.0, 1.6 Hz, 1 H), 7.85-7.94(m, 1 H), 8.19 (br d, J = 3.4 Hz, 1 H), 8.81 (s, 1 H)34460.483.15-3.19 (m, 2 H), 3.23 (s, 3 H), 5.25-5.33 (m, 1 H),[M + H]+(A)5.50 (d, J = 1.9 Hz, 2 H), 6.16 (d, J = 5.2 Hz, 1 H),7.20 (td, J = 9.1, 2.4 Hz, 1 H), 7.25 (s, 1 H), 7.66(d, J = 3.3 Hz, 1 H), 7.76 (dd, J = 8.7, 5.3 Hz, 1 H),7.82 (dd, J = 9.4, 2.4 Hz, 1 H),8.18 (d, J = 3.3 Hz, 1 H), 8.80 (s, 1 H)44460.493.15-3.29 (m, 5 H), 5.11 (dt, J = 7.8, 5.7 Hz, 1 H),[M + H]+(A)5.48 (s, 2 H), 6.00 (d, J = 5.7 Hz, 1 H), 6.77 (s, 1 H),7.29 (dd, J = 8.7, 2.3 Hz, 1 H), 7.57(d, J = 8.7 Hz, 1 H), 7.64-7.67 (m, 2 H),8.18 (d, J = 3.4 Hz, 1 H), 8.78 (s, 1 H)54300.443.13-3.31 (m, 5 H), 5.10 (dd, J = 7.9, 5.6 Hz, 1 H),[M + H]+(A)5.48 (s, 2 H), 6.77 (s, 1 H), 7.09 (td, J = 9.2, 2.7 Hz,1 H), 7.38 (dd, J = 8.9, 2.7 Hz, 1 H), 7.55(dd, J = 9.0, 4.2 Hz, 1 H), 7.65 (br d, J = 3.2 Hz, 1 H),8.18 (br d, J = 3.3 Hz, 1 H), 8.78 (s, 1 H)64980.492.95-3.01 (m, 2 H), 3.23 (s, 3 H), 4.86-4.95[M + H]+(A)(m, 1 H), 5.48 (d, J = 1.9 Hz, 2 H), 7.13-7.20(m, 2 H), , 7.59-7.70 (m, 3 H), 8.19(d, J = 3.4 Hz, 1 H), 8.80 (s, 1H)73860.442.26 (s, 3 H), 2.89-3.05 (m, 2 H), 3.23 (s, 3 H), 4.91[M + H]+(A)(dd, J = 8.2, 5.5 Hz, 1 H), 5.48 (d, J = 1.3 Hz, 2 H),7.07-7.12 (m, 2 H), 7.18-7.24 (m, 2H),7.65 (br d, J = 3.2 Hz, 1 H), 8.19(br d, J = 3.3 Hz, 1 H), 8.80 (s, 1 H) Analytical HPLC methods Method A timeVol % waterVol %Flow(min)(incl. 0.1% TFA)ACN[mL/min]0.009911.60.029911.61.0001001.61.1001001.6 Analytical column: XBridge BEH C18_2.1×30 mm, 1.7 μm; column temperature: 60° C. timeVol % waterVol %Flow(min)(incl. 0.1% TFA)ACN[mL/min]0.009732.20.209732.21.2001002.21.2501003.01.4001003.0 Analytical column: Stable Bond (Agilent) 1.8 μm; 3.0×30 mm; column temp: 60° C. Method C timeVol % waterVol %Flow(min)(incl. 0.1% TFA)ACN[mL/min]0.009732.20.209732.21.2001002.21.2501003.01.4001003.0 Analytical column: Sunfire (Waters) 2.5 μm; 3.0×30 mm; column temperature: 60° C. Method D Gradient/SolventVol % waterVol % CANFlowTime [min](incl. 0.1% FA)(incl. 0.1% FA)[ml/min]0.019550.54.005950.55.005950.55.209550.56.009550.5 Analytical column: ACQUITY UPLC C18_2.1×50 mm_1.8 μm. 100 Å; column temperature: 25° C. Method E Gradient/SolventVol % waterVol % CANFlowTime [min](incl. 0.1% FA)(incl. 0.1% FA)[ml/min]0.009550.510.005950.510.505950.511.009550.512.009550.5 Analytical column: ACQUITY UPLC C18_2.1×50 mm_1.8 μm. 100 Å; column temperature: 25° C.
67,150
11858922
DETAILED DESCRIPTION The present invention is predicated, at least in part, on the finding that certain sulfonyl ureas and related compounds have advantageous properties and show useful activity in the inhibition of activation of the NLRP3 inflammasome and/or inhibition of IL-1β and/or IL-17 and/or IL-18, and/or IL-1α, and/or IL-37, and/or IL-33 as well as interfere with or modulate the activity of T helper cells such as Th17. Particularly, the compounds of the invention are useful in the treatment of a wide range of disorders in which the inflammation process, or the NLRP3 inflammasome and/or IL-1β and/or IL-17 and/or IL-18, and/or IL-1α, and/or IL-37, and/or IL-33 and/or Th17 cells play a part. Evidence from human CAPS patients and mouse models of CAPS has lead the present inventors to believe that NLRP3 inhibition will be a superior treatment over IL-1 biologics, as inhibition of all NLRP3-dependent processes will be more effective than inhibition of a single NLRP3-dependent process, such as IL-1 signalling. Individuals with CAPS display dysregulated secretion of both IL-1β and IL-18, and CAPS patients treated with anti-IL-1 biologics have residual disease. Symptoms such as bony overgrowth and joint deformity are not prevented by IL-1 biologics. In addition, symptoms involving the central nervous system such as hearing loss are difficult to control using IL-1 biologics, which appear to poorly penetrate the central nervous system. Studies in mouse models of CAPS indicate that deficiency in either IL-1 signalling or IL-18 alone is insufficient to block systemic inflammation, particularly in older animals. In a severe model of CAPS, only a complete loss of caspase-1 signalling fully rescued the disease. Specific inhibition of NLRP3 by sulfonyurea-containing compounds, such as those of the first aspect, may block all processes downstream of NLRP3, including ASC speck formation and caspase-8 and caspase-1 activation. Consequently, NLRP3 inhibition will block all caspase-1 dependent processes such as IL-1β, IL-18 and IL-37 processing and secretion, gasdermin D cleavage, pyroptosis, and release of IL-1α, IL-33 and HMGB. Furthermore, NLRP3-dependent extracellular release of the ASC speck will be blocked, and caspase-8-dependent pro-IL-1β and pro-IL-18 cleavage and apoptotic cell death will be prevented. Thus, specific inhibition of NLRP3 by compounds of the first aspect will prevent multiple downstream inflammatory signals and should therefore prove more effective as an anti-inflammatory therapy than IL-1 blockade alone. Anti-IL-1 biologics block IL-1 derived from NLRP3-independent sources, such as IL-1 produced by other inflammasomes (e.g. NLRC4, NLRP1, NLRP6, AIM2), and IL-1 generated by the latter pathways may be important for host defence against pathogens. For example, patients receiving IL-1/IL-1R antagonists exhibit increased incidence of upper airway infections. Specific inhibition of NLRP3 by the present compounds may thus exert less generalised immunosuppression compared to anti-IL-1 biologics. IL-1β and IL-18, generated by the NLRP3/caspase-1 axis, play critical roles in driving IL-17 production by CD4 Th17 cells and γδ T cells. IL-1β and IL-18 synergise with IL-23 to induce IL-17 production by memory CD4 Th17 cells and by γδ T cells in the absence of TCR engagement. IL-1-driven IL-17 has also been implicated in psoriasis, type I diabetes, rheumatoid arthritis, type 2 diabetes mellitus, atherosclerosis, obesity, gout, and recently, asthma. In essence, each of these diseases has been shown to involve the activation of tissue macrophages, dendritic cells, or brain microglia, driven by the frustrated phagocytosis of metabolites that accumulate extracellularly. NLRP3 senses this phagocytic event, leading to IL-1 release, triggering inflammation to clear the offensive material. Disease will result if this process becomes chronic or over-activated, which explains why so many diseases have been shown to involve NLRP3. Inhibitors that act to prevent NLRP3 activation hence can have utility in IL-17 driven, as well as IL-1 driven diseases. In this patent specification, the terms ‘comprises’, ‘comprising’, ‘includes’, ‘including’, or similar terms are intended to mean a non-exclusive inclusion, such that a method or composition that comprises a list of elements does not include those elements solely, but may well include other elements not listed. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as would be commonly understood by those of ordinary skill in the art to which this invention belongs. The term “pharmaceutically acceptable salt”, as used herein, refers to salts which are toxicologically safe for systemic or localised administration such as salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. The pharmaceutically acceptable salts may be selected from the group including alkali and alkali earth, ammonium, aluminium, iron, glucosamine, chloride, sulphate, sulphonate, bisulphate, nitrate, citrate, tartrate, bitartrate, phosphate, carbonate, bicarbonate, malate, maleate, napsylate, fumarate, succinate, acetate, benzoate, terephthalate, palmoate, piperazine, pectinate and S-methyl methionine salts and the like. The term “alkyl” refers to a straight-chain or branched alkyl substituent containing from, for example, 1 to about 12 carbon atoms, preferably 1 to about 9 carbon atoms, more preferably 1 to about 6 carbon atoms, even more preferably from 1 to about 4 carbon atoms, still yet more preferably from 1 to 2 carbon atoms. Examples of alkyl groups may be selected from the group consisting of methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, 2-methylbutyl, 3-methylbutyl, hexyl, heptyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-ethylbutyl, 3-ethylbutyl, octyl, nonyl, decyl, undecyl, dodecyl and the like. The number of carbons referred to relates to the carbon backbone and carbon branching, but does not include carbon atoms belonging to any substituents, for example the carbon atoms of an alkoxy substituent branching off the main carbon chain. Substituted alkyl includes alkyl substituted with one or more moieties selected from the group consisting of halo (e.g., Cl, F, Br, and I); halogenated alkyl (e.g., CF3, 2-Br-ethyl, CH2F, CH2Cl, CH2CF3, or CF2CF3); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of “optionally substituted”. An “alkylene” group is similarly defined as a divalent alkyl group. The term “alkenyl” refers to unsaturated linear or branched hydrocarbon groups, having 2 to 12 carbon atoms, preferably 2 to 9 carbon atoms, more preferably 2 to 6 carbon atoms, and having at least one carbon-carbon double bond. Where appropriate, the alkenyl group may have a specified number of carbon atoms, for example, C2-C6alkenyl which includes alkenyl groups having 2, 3, 4, 5 or 6 carbon atoms in linear or branched arrangements. The number of carbons referred to relates to the carbon backbone and carbon branching, but does not include carbon atoms belonging to any substituents. Examples of alkenyl groups may be selected from the group consisting of ethenyl, propenyl, isopropenyl, butenyl, s- and t-butenyl, pentenyl, hexenyl, hepta-1,3-dienyl, hexa-1,3-dienyl, nona-1,3,5-trienyl and the like. Substituted alkenyl includes alkenyl substituted with one or more moieties selected from the group consisting of halo (e.g., Cl, F, Br, and I); halogenated alkyl (e.g., CF3, 2-Br-ethyl, CH2F, CH2Cl, CH2CF3, or CF2CF3); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of “optionally substituted”. An “alkenylene” group is similarly defined as a divalent alkenyl group. The term “alkynyl” refers to unsaturated linear or branched hydrocarbon groups, having 2 to 12 carbon atoms, preferably 2 to 9 carbon atoms, more preferably 2 to 6 carbon atoms, and having at least one carbon-carbon triple bond. Where appropriate, the alkynyl group may have a specified number of carbon atoms, for example, C2-C6alkynyl which includes alkynyl groups having 2, 3, 4, 5 or 6 carbon atoms in linear or branched arrangements. The number of carbons referred to relates to the carbon backbone and carbon branching, but does not include carbon atoms belonging to any substituents. Examples of alkynyl groups may be selected from the group consisting of ethynyl, propargyl, but-1-ynyl, but-2-ynyl and the like. Substituted alkynyl includes alkynyl substituted with one or more moieties selected from the group consisting of halo (e.g., Cl, F, Br, and I); halogenated alkyl (e.g., CF3, 2-Br-ethyl, CH2F, CH2Cl, CH2CF3, or CF2CF3); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of “optionally substituted”. An “alkynylene” group is similarly defined as a divalent alkynyl group. The term “alkoxy” as used herein means straight or branched chain alkyl groups linked by an oxygen atom (i.e., —O-alkyl), wherein alkyl is as described above. In particular embodiments, alkoxy refers to oxygen-linked groups comprising 1 to 10 carbon atoms (“C1-10alkoxy”). In further embodiments, alkoxy refers to oxygen-linked groups comprising 1 to 8 carbon atoms (“C1-8alkoxy”), 1 to 6 carbon atoms (“C1-6alkoxy”), 1 to 4 carbon atoms (“C1-4alkoxy”) or 1 to 3 carbon atoms (“C1-3alkoxy”). The terms “cycloalkyl” and “cycloalkenyl” refer to saturated and unsaturated mono-cyclic, bicyclic or tricyclic carbon groups. Where appropriate, the cycloalkyl or cycloalkenyl group may have a specified number of carbon atoms, for example, C3-C6cycloalkyl or cycloalkenyl includes within its scope a carbocyclic group having 3, 4, 5 or 6 carbon atoms. Examples of cycloalkyl and cycloalkenyl groups may be selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl and the like. Substituted cycloalkyl or cycloalkenyl includes substitutions with one or more moieties selected from the group consisting of halo (e.g., Cl, F, Br, and I); halogenated alkyl (e.g., CF3, 2-Br-ethyl, CH2F, CH2Cl, CH2CF3, or CF2CF3); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of “optionally substituted”. The term “alkylthio” as used herein means a thio group with one alkyl substituent (i.e., —S-alkyl), where alkyl is defined as above. The term “amino” as used herein means a moiety represented by the structure NR23, and includes primary amines, and secondary and tertiary amines substituted by alkyl (i.e., alkylamino). Thus, R23may represent, for example, two hydrogen atoms, two alkyl moieties, or one hydrogen atom and one alkyl moiety. The term “aryl” refers to a monocyclic, bicyclic, tricyclic or other polycyclic carbon ring of up to 8 members in each ring, wherein at least one ring is aromatic as defined by the Hückel 4n+2 rule. The term includes polycyclic systems comprising saturated carbon rings or heteroaryl or heterocyclic groups so long as at least one ring is aryl, as described. An “arylene” group is similarly defined as a divalent aryl group. The terms “aralkyl” and “arylalkyl” as used herein mean an aryl group as defined above linked to the molecule through an alkylene group as defined above. For the purposes of the present invention, where a combination of groups is referred to as one moiety, for example, arylalkyl, arylalkenyl, arylalkynyl, alkylaryl, alkenylaryl or alkynylaryl, the last mentioned group contains the atom by which the moiety is attached to the rest of the molecule. A typical example of an arylalkyl group is benzyl. The term “heteroaryl” refers to an aryl group containing from one or more (particularly one to four) non-carbon ring atom(s) (particularly N, O or S) or a combination thereof. A heteroaryl group may be optionally substituted at one or more carbon or nitrogen atom(s). Heteroaryl rings may also be fused with one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings. Heteroaryl includes, but is not limited to, 5-membered heteroaryls having one hetero atom (e.g., thiophenes, pyrroles, furans); 5-membered heteroaryls having two heteroatoms in 1,2 or 1,3 positions (e.g., oxazoles, pyrazoles, imidazoles, thiazoles, purines); 5-membered heteroaryls having three heteroatoms (e.g., triazoles, thiadiazoles); 5-membered heteroaryls having four heteroatoms (e.g., tetrazoles); 6-membered heteroaryls with one heteroatom (e.g., pyridine, quinoline, isoquinoline, phenanthrine, 5,6-cycloheptenopyridine); 6-membered heteroaryls with two heteroatoms (e.g., pyridazines, cinnolines, phthalazines, pyrazines, pyrimidines, quinazolines); 6-membered heteroaryls with three heteroatoms (e.g., 1,3,5-triazine); and 6-membered heteroaryls with four heteroatoms. “Substituted heteroaryl” means a heteroaryl having one or more groups as substituents and including those defined under “optionally substituted”. A “heteroarylene” group is similarly defined as a divalent heteroaryl group. “Heterocyclyl” as used herein refers to a non-aromatic ring having 3 to 8 atoms in the ring, preferably 5 to 8 atoms in the ring, and of those atoms 1 to 4 are heteroatoms (particularly N, O or S). Heterocyclic rings may also be fused with one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings. Heterocyclic includes partially and fully saturated heterocyclic groups. Heterocyclic systems may be attached to another moiety via any number of carbon atoms or heteroatoms of the radical and may be both saturated and unsaturated. Non-limiting examples of heterocyclic groups include C4-C6selenocycles, pyrrolidinyl, pyrrolinyl, pyranyl, piperidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolinyl, dithiolyl, oxathiolyl, dioxanyl, dioxinyl, oxazinyl, azepinyl, diazepinyl, thiazepinyl, oxepinyl and thiapinyl, imidazolinyl, thiomorpholinyl, and the like. The term “acyl” as used herein means C(O)R19wherein R19is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, aralkyl, heteroaryl or heterocyclyl. The term “halo” as used herein refers to fluoro, chloro, bromo and iodo groups. Similarly the term “halogen” as used herein refers to fluorine, chlorine, bromine and iodine. “Optionally substituted” in reference to a substituent group refers to substituent groups optionally substituted with one or more moieties, for example, those selected from the group consisting of optionally substituted C1-10alkyl (e.g., optionally substituted C1-6alkyl); optionally substituted C3-6cycloalkyl (e.g., optionally substituted cyclopropyl), optionally substituted hydroxylalkyl; optionally substituted C1-10alkoxy (e.g., optionally substituted C1-6alkoxy); optionally substituted C2-10alkenyl; optionally substituted C2-10alkynyl; optionally substituted C6-12aryl; aryloxy; optionally substituted heteroaryl; optionally substituted heterocyclyl; halo (e.g., Cl, F, Br, and I); hydroxyl; halogenated alkyl (e.g., CF3, 2-Br-ethyl, CH2F, CH2CF3, and CF2CF3); amino (e.g., NH2, NR24H, and NR24R25); alkylamino; arylamino; acyl; amido; CN; NO2; N3; CH2OH; CONH2; CONR24R25; CO2R24; CH2OR24; NHCOR24; NHCO2R24; C1-3alkylthio; sulfate; sulfonic acid; sulfonate esters such as alkyl or aralkyl sulfonyl, including methanesulfonyl; phosphonic acid; phosphate; phosphonate; mono-, di-, or triphosphate esters; trityl or monomethoxytrityl; R24SO; R24SO2; CF3S; and CF3SO2; trialkylsilyl such as dimethyl-t-butylsilyl or diphenylmethylsilyl; and R24and R25are each independently selected from H or optionally substituted C1-10alkyl, C1-6alkyl or C1-4alkyl. Optional substituents also include cyclic structures such as cyclic hydrocarbon (e.g. cycloalkyl, cycloalkenyl), heterocyclic, aryl and heteroaryl rings, fused to the parent moiety. Whenever a range of the number of atoms in a structure is indicated (e.g., a C1-C12, C1-C10, C1-C9, C1-C6, C1-C4, or C2-C20, C2-C12, C2-C10, C2- C9, C2-C8, C2-C6, C2-C4alkyl, alkenyl, etc.), it is specifically contemplated that any sub-range or individual number of carbon atoms falling within the indicated range also can be used. Thus, for instance, the recitation of a range of 1-12 carbon atoms (e.g., C1-C12), 1-9 carbon atoms (e.g., C1-C9), 1-6 carbon atoms (e.g., C1-C6), 1-4 carbon atoms (e.g., C1-C4), 1-3 carbon atoms (e.g., C1-C3), or 2-8 carbon atoms (e.g., C2-C8) as used with respect to any chemical group (e.g., alkyl, etc.) referenced herein encompasses and specifically describes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and/or 12 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 1-2 carbon atoms, 1-3 carbon atoms, 1-4 carbon atoms, 1-5 carbon atoms, 1-6 carbon atoms, 1-7 carbon atoms, 1-8 carbon atoms, 1-9 carbon atoms, 1-10 carbon atoms, 1-11 carbon atoms, 1-12 carbon atoms, 2-3 carbon atoms, 2-4 carbon atoms, 2-5 carbon atoms, 2-6 carbon atoms, 2-7 carbon atoms, 2-8 carbon atoms, 2-9 carbon atoms, 2-10 carbon atoms, 2-11 carbon atoms, 2-12 carbon atoms, 3-4 carbon atoms, 3-5 carbon atoms, 3-6 carbon atoms, 3-7 carbon atoms, 3-8 carbon atoms, 3-9 carbon atoms, 3-10 carbon atoms, 3-11 carbon atoms, 3-12 carbon atoms, 4-5 carbon atoms, 4-6 carbon atoms, 4-7 carbon atoms, 4-8 carbon atoms, 4-9 carbon atoms, 4-10 carbon atoms, 4-11 carbon atoms, and/or 4-12 carbon atoms, etc., as appropriate). According to a first aspect of the invention, there is provided a compound of formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof: wherein Q is selected from O, S and Se;J is S or Se;W1and W2, when present, are independently selected from N and C;R1and R2are independently selected from the group consisting of hydrogen, C1-C12alkyl, C2-C12alkenyl, C2-C12alkynyl, aryl, heterocyclyl, heteroaryl, cycloalkyl, cycloalkenyl, amino, amido, alkylthio, acyl, arylalkyl and acylamido, all of which may be optionally substituted; andwherein at least one of W1and W2is present and is a nitrogen atom and when R1or R2are cyclic then the respective W1or W2may form part of the ring structure. It will be apparent to the skilled addressee that the structure of formula (I) covers compounds wherein either W1or W2is nitrogen as well as compounds wherein both W1and W2are nitrogen. Additionally, when W1and/or W2is nitrogen then that nitrogen may be part of a chain linking to R1or R2or may be an atom which forms part of a ring structure. In preferred embodiments, R1W1— is (R1)2N— or (R1)HN— or (R1)—, and —W2R2is —N(R2)2or —NH(R2) or —(R2), provided that a nitrogen atom of R1W1— and/or a nitrogen atom of —W2R2is linked to (i.e. bonded to) the remainder of the molecule. In one embodiment, a sp3hybridised nitrogen atom of R1W1— is linked to J. In another embodiment, a sp3hybridised nitrogen atom of —W2R2is linked to the remainder of the molecule of formula (I). In preferred embodiments, Q is O and J is S. In one embodiment, R1and R2are independently C1-C10alkyl which may be optionally substituted. In an embodiment, R1and R2are independently C1-C8alkyl which may be optionally substituted. In a further embodiment, R1and R2are independently C1-C6alkyl which may be optionally substituted. In one embodiment, R1and R2are independently C1-C10alkenyl which may be optionally substituted. In a further embodiment, R1and R2are independently C1-C8alkenyl which may be optionally substituted. In an embodiment, R1and R2are independently C1-C6alkenyl which may be optionally substituted. In one embodiment, R1and R2are independently C1-C10alkynyl which may be optionally substituted. In a further embodiment, R1and R2are independently C1-C8alkynyl which may be optionally substituted. In one embodiment, R1and R2are independently C1-C6alkynyl which may be optionally substituted. In an embodiment, one or more hydrogens of the alkyl, alkenyl or alkynyl groups is deuterated. In certain embodiments, R1and R2are independently C3-C8cycloalkyl or cycloalkenyl which may each be optionally substituted. In one embodiment, R1and R2are independently C4-C7cycloalkyl or cycloalkenyl which may each be optionally substituted. In one embodiment, R1and R2are independently selected from C5or C6cycloalkyl or cycloalkenyl, each of which may be optionally substituted. In one embodiment, R1and R2are independently C6-C8aryl which may be optionally substituted. In one embodiment, R1and R2are independently C6-C7aryl which may be optionally substituted. In one embodiment, W2and R2may form an indacene group, including substituted, for example halogenated, and hydrogenated variants thereof. In one embodiment, W2and R2may form a hexahydro-indacene group, preferably a hexahydro-s-indacene group. In one embodiment, R1and R2are independently C5-C8heteroaryl which may be optionally substituted. In one embodiment, R1and R2are independently C5-C7heteroaryl which may be optionally substituted. In one embodiment, R1and R2are independently selected from C5or C6heteroaryl, each of which may be optionally substituted. In one embodiment, R1and R2are independently C3-C8heterocyclyl which may be optionally substituted. In one embodiment, R1and R2are independently C4-C7heterocyclyl which may be optionally substituted. In one embodiment, R1and R2are independently selected from C5or C6heterocyclyl, each of which may be optionally substituted. It will be understood that W1and W2may independently represent —N— or —NH depending on the degree of substitution with R1. That is, when R1is, for example, C3alkyl then W1may either be mono- or disubstituted with C3alkyl. Thus in all definitions where R1or R2are described as e.g. alkyl, alkenyl etc. then this may also be read as dialkyl, dialkenyl etc. In one embodiment W1/R1or W2/R2may form a selenocycle. In one embodiment —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions, wherein —W2R2may optionally be further substituted. For example, —W2R2may be a phenyl group substituted at the 2- and 6-positions. Typical substituents at the α and α′ positions include alkyl, cycloalkyl, alkoxy, cycloalkoxy, alkenyl, cycloalkenyl, alkynyl, acyl, aryl, alkylaryl, alkoxyaryl, heteroaryl, heterocyclyl, arylalkyl and heteroarylalkyl groups. More typically, the substituents at the α and α′ positions are independently selected from alkyl and cycloalkyl groups, such as C3-C6branched or C3-C6cyclic alkyl groups, e.g. isopropyl, cyclopropyl, cyclohexyl or t-butyl groups. Other typical substituents at the α and α′ positions include cyclic hydrocarbon, heterocyclic, aryl or heteroaryl rings which are fused to the parent aryl or heteroaryl group across the α,β and/or α′,β′ positions respectively. Such fused aryl and fused heteroaryl groups are described in greater detail below. As used herein, the nomenclature α, β, α′, β′ refers to the position of the atoms of the aryl or heteroaryl group relative to the point of attachment of the —W2R2moiety to the remainder of the molecule. For example, where —W2R2is a 1,2,3,5,6,7-hexahydro-s-indacen-4-yl moiety, the α, β, α′ and β′ positions are as follows: In one embodiment —W2R2is a fused aryl or a fused heteroaryl group, wherein the aryl or heteroaryl group is fused to one or more cyclic hydrocarbon, heterocyclic, aryl or heteroaryl rings, wherein —W2R2may be optionally substituted. In another embodiment, —W2R2is a fused aryl or a fused heteroaryl group, wherein the aryl or heteroaryl group is fused to two or more cyclic hydrocarbon, heterocyclic, aryl or heteroaryl rings, wherein —W2R2may be optionally substituted. Typically, the two or more cyclic hydrocarbon, heterocyclic, aryl or heteroaryl rings are each ortho-fused to the aryl or heteroaryl group, i.e. each fused cyclic hydrocarbon, heterocyclic, aryl or heteroaryl ring has only two atoms and one bond in common with the aryl or heteroaryl group. In yet another embodiment, —W2R2is a fused aryl or a fused heteroaryl group, wherein a first cyclic hydrocarbon, heterocyclic, aryl or heteroaryl ring is fused to the aryl or heteroaryl group across the α,β positions and a second cyclic hydrocarbon, heterocyclic, aryl or heteroaryl ring is fused to the aryl or heteroaryl group across the α′,β′ positions, and wherein —W2R2may be optionally substituted. Typically in any embodiment where —W2R2is a fused aryl or a fused heteroaryl group, R1W1— is (R1)2N— or R1NH—, J is S and Q is O, wherein R1is as previously defined. In one embodiment, —W2R2has a formula selected from: wherein A1and A2are each independently selected from an optionally substituted alkylene or alkenylene group, which may optionally include one or more heteroatoms N, O or S in its carbon skeleton, and wherein B1is hydrogen or any optional substituent. B1and any optional substituent attached to A1or A2may together with the atoms to which they are attached form a further fused cyclic hydrocarbon, heterocyclic, aryl or heteroaryl ring which may itself be optionally substituted. Similarly, any optional substituent attached to A1and any optional substituent attached to A2may also together with the atoms to which they are attached form a further fused cyclic hydrocarbon, heterocyclic, aryl or heteroaryl ring which may itself be optionally substituted. Typically, B1is hydrogen or a halo, hydroxyl, —CN, —NO2, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy or C1-C4haloalkoxy group. Typically, any ring containing A1or A2is a five or a six membered ring. In a further embodiment, —W2R2has a formula selected from: Examples of compounds where —W2R2is a fused aryl or a fused heteroaryl group include the compounds of Examples 1-43 below and the compounds: In one embodiment, R1W1— comprises a heteroaryl group, wherein R1W1— may be optionally substituted. Typically in any embodiment where R1W1-comprises a heteroaryl group, a nitrogen atom of R1W1— is linked to J. Typically, in any embodiment where R1W1— comprises a heteroaryl group and a nitrogen atom of R1W1— is linked to J, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically in any embodiment where R1W1— comprises a heteroaryl group, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. More typically, in any embodiment where R1W1— comprises a heteroaryl group, a nitrogen atom of R1W1— is linked to J and —W2R2is an aryl group, wherein the aryl group is substituted at the α and α′ positions and optionally at other positions. In one embodiment, R1W1— is R1NH— or (R1)2N— wherein at least one R1comprises a heteroaryl group, or two R1together with the nitrogen atom to which they are attached form a heteroaryl group or a cyclic group which is substituted with a heteroaryl group, wherein R1W1— may be optionally substituted. In one embodiment, R1W1— is Het-L-NH— or Het-L-NR1—, wherein Het is an optionally substituted heteroaryl group, -L- is a bond or an optionally substituted alkylene, alkenylene, alkynylene or arylene group, which may optionally include one or more heteroatoms N, O or S in its carbon skeleton, and R1is as previously defined. Typically, -L- is a bond or a C1-C2alkylene group. In one embodiment, Het is an optionally substituted monocyclic or bicyclic heteroaryl group. Typically, such a group is unsubstituted or substituted with one or more halo, alkyl, alkoxy, aryl, alkylaryl, alkoxyaryl, heteroaryl or halogenated alkyl groups. In a further embodiment, Het is selected from an optionally substituted furanyl, thiophenyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, indolizinyl, indolyl, isoindolyl, benzofuranyl, benzothiophenyl, indazolyl, benzimidazolyl, benzthiazolyl, purinyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl or pteridinyl group. Typically, such a group is unsubstituted or substituted with one or more halo, alkyl, alkoxy, acyl, aryl, alkylaryl, alkoxyaryl, heteroaryl, arylalkyl, heteroarylalkyl, or halogenated alkyl groups. Examples of compounds where R1W1— comprises a heteroaryl group include the compounds of Examples 4, 8, 13, 14, 15, 17-23, 27, 29, 30, 35, 39, 42 and 43 below and the compounds: In one embodiment, R1W1— comprises a heterocyclic group containing a nitrogen atom and at least one further heteroatom in the heterocyclic ring, wherein R1W1— may be optionally substituted. For example, R1W1— may comprise a heterocyclic group containing at least two nitrogen atoms in the heterocyclic ring, wherein R1W1— may be optionally substituted. Typically in any embodiment where R1W1— comprises a heterocyclic group containing a nitrogen atom and at least one further heteroatom in the heterocyclic ring, a nitrogen atom of R1W1— is linked to J, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically in any embodiment where R1W1— comprises a heterocyclic group containing a nitrogen atom and at least one further heteroatom, such as a nitrogen atom, in the heterocyclic ring, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. In one embodiment, R1W1— is (R1)2N— wherein both R1and the nitrogen atom to which they are attached together form an optionally substituted heterocyclic group containing at least one further heteroatom N, O or S in the heterocyclic ring. Typically, R1W1— is (R1)2N— wherein both R1and the nitrogen atom to which they are attached together form an optionally substituted heterocyclic group containing at least one further nitrogen atom in the heterocyclic ring. Typically, the heterocyclic group is monocyclic or bicyclic. In one embodiment, R1W1— is (R1)2N—, wherein (R1)2N— is an optionally substituted piperazinyl, morpholinyl or thiomorpholinyl group. Typically such a group is unsubstituted or substituted with one or more halo, alkyl, alkoxy, acyl, aryl, alkylaryl, alkoxyaryl, heteroaryl, arylalkyl, heteroarylalkyl, or halogenated alkyl groups. Examples of compounds where R1W1— comprises a heterocyclic group containing a nitrogen atom and at least one further heteroatom in the heterocyclic ring include the compounds of Examples 1, 2, 13, 16, 17 and 41 below and the compounds: In one embodiment, R1W1— is R1NH— or (R1)2N— wherein at least one R1comprises a fused bicyclic group, or two R1together with the nitrogen atom to which they are attached form a fused bicyclic group, wherein R1W1— may be optionally substituted. The fused bicyclic group may be optionally substituted and may be carbocyclic or heterocyclic. Both rings of the bicyclic group may be aromatic, or one ring may be aromatic and the other non-aromatic, or both rings may be non-aromatic. Typically in any embodiment where R1W1— is R1NH— or (R1)2N— wherein at least one R1comprises a fused bicyclic group, or two R1together with the nitrogen atom to which they are attached form a fused bicyclic group, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically in any embodiment where R1W1— is R1NH— or (R1)2N— wherein at least one R1comprises a fused bicyclic group, or two R1together with the nitrogen atom to which they are attached form a fused bicyclic group, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. In one embodiment, R1W1— is Bic-L-NH— or Bic-L-NR1—, wherein Bic is an optionally substituted fused bicyclic group, -L- is a bond or an optionally substituted alkylene, alkenylene, alkynylene or arylene group, which may optionally include one or more heteroatoms N, O or S in its carbon skeleton, and R1is as previously defined. Typically, -L- is a bond or a C1-C2alkylene group. In one embodiment, Bic comprises a 5-membered ring fused to a six-membered ring. Typically in such an embodiment, the six-membered ring is aromatic. Examples of such groups include optionally substituted indolyl, isoindolyl, indolinyl, indazolyl, indenyl, indanyl and 1,3-benzodioxolyl groups. Examples of compounds where R1W1— is R1NH— or (R1)2N— wherein at least one R1comprises a fused bicyclic group, or two R1together with the nitrogen atom to which they are attached form a fused bicyclic group, include the compounds of Examples 8, 28, 34, 36, 37, 38, 40, 42 and 43 below and the compounds: In one embodiment, R1W1— is halo-substituted. Typically in such an embodiment, R1W1— is substituted with one or more fluoro and/or chloro groups. Typically in any embodiment where R1W1— is halo-substituted, a nitrogen atom of R1W1— is linked to J, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically in any embodiment where R1W1— is halo-substituted, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. In one embodiment, R1W1— is Har-L-NH— or Har-L-NR1—, wherein Har is an aryl or a heteroaryl group substituted with one or more halo, halogenated alkyl or halogenated alkoxy groups, wherein Har may optionally be further substituted, -L- is a bond or an optionally substituted alkylene, alkenylene, alkynylene or arylene group, which may optionally include one or more heteroatoms N, O or S in its carbon skeleton, and R1is as previously defined. Typically, -L- is a bond, a —O—(C1-C2alkylene)- group or a C1-C2alkylene group. In one embodiment, Har is a phenyl or toluyl group substituted with one or more chloro, fluoro, trifluoromethyl and/or trifluoromethoxy groups. Examples of compounds where R1W1— is halo-substituted include the compounds of Examples 5, 6, 9-11 and 32 below and the compounds: In one embodiment, R1W1— is Het-X—(CH2)m—NH—, Ar—X—(CH2)m—NH—, Het-X—(CH2)m—NR1— or Ar—X—(CH2)m—NR1—; wherein Het is as defined above; Ar is an optionally substituted aryl group; —X— is a bond, —NH—, —S— or —O—; and m is 2-6. Typically m is 2-4. More typically m is 2. Typically in such an embodiment, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically in such an embodiment, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. Examples of compounds where R1W1— is Het-X—(CH2)m—NH—, Ar—X—(CH2)m—NH—, Het-X—(CH2)m—NR1— or Ar—X—(CH2)m—NR1— include the compounds of Examples 5, 15, 21 and 24-26 below. In one embodiment, R1W1— is substituted with an alkylsulphonyl or a cyano group. For example, R1W1— may be R1NH— or (R1)2N— wherein at least one R1is a cyano- or alkylsulphonyl-substituted aryl or arylalkyl group, which may optionally be substituted with further substituents. Typically in such an embodiment, a nitrogen atom of R1W1— is linked to J, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically in such an embodiment, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. Examples of compounds where R1W1— is substituted with an alkylsulphonyl or a cyano group include the compounds of Examples 7 and 33 below. In one embodiment, R1W1— is R1N(Me)-, wherein R1is an optionally substituted aryl or heteroaryl group. Typically in such an embodiment, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically in such an embodiment, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. An example of such a compound is the compound of Example 31 below. In one embodiment, R1W1— is Am-M-NH— or Am-M-NR1—, wherein Am is a primary, secondary or tertiary amino group, -M- is a branched alkylene, a cycloalkylene or a cycloalkyl-substituted alkylene group, and R1is as previously defined. Typically, Am is a dialkylamino group and -M- is a cycloalkyl-substituted alkylene group. Typically in such an embodiment, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically in such an embodiment, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. An example of such a compound is the compound of Example 12 below. In one embodiment, R1W1— comprises a heterocyclic group, wherein the heterocyclic group is substituted with one or more hydroxyl, halo, alkyl, alkoxy, halogenated alkyl or halogenated alkoxy groups. Optionally the substituted heterocyclic group contains a single heteroatom in the heterocyclic ring, such as nitrogen. Alternatively the substituted heterocyclic group may contain two or more heteroatoms in the heterocyclic ring, such as nitrogen and one or more further heteroatoms selected from O, N or S. Typically, the substituted heterocyclic group is substituted with one or more hydroxyl or C1-C4alkyl groups. Typically in any embodiment where R1W1— comprises a substituted heterocyclic group, a nitrogen atom of R1W1— is linked to J. Typically, in any embodiment where R1W1— comprises a substituted heterocyclic group and a nitrogen atom of R1W1— is linked to J, J is S, Q is O and —W2R2is —R2wherein R2is as previously defined. Typically, in any embodiment where R1W1— comprises a substituted heterocyclic group, —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions. Specific examples of such compounds include the compounds of Examples 2 and 41 below and: In one embodiment, when —W2R2is an aryl or a heteroaryl group, wherein the aryl or the heteroaryl group is substituted at the α and α′ positions and optionally at other positions, —W2R2is halo substituted at a position other than the α and α′ position of the aryl or the heteroaryl group. For example, —W2R2may be a phenyl group, wherein the phenyl group is fluoro, chloro and/or bromo substituted at one or more of the 3-, 4- and 5-positions, and substituted at the 2- and 6-positions with groups independently selected from alkyl, cycloalkyl, alkoxy, cycloalkoxy, alkenyl, cycloalkenyl, alkynyl, acyl, aryl, alkylaryl, alkoxyaryl, heteroaryl, heterocyclyl, arylalkyl and heteroarylalkyl groups. Examples of such compounds include: In one embodiment, —W2R2is an optionally substituted —NHR2or —N(R2)2group, wherein optionally two R2together with the nitrogen atom to which they are attached may form a cyclic group, and R1W1— is Het, wherein Het is as defined above. Typically in such an embodiment, J is S and Q is O. Typically in such an embodiment, Het is an optionally substituted monocyclic or bicyclic heteroaryl group. More typically, Het is an optionally substituted five membered monocyclic heteroaryl group or an optionally substituted fused bicyclic heteroaryl group containing a five membered and a six membered ring. In another embodiment, —W2R2is an optionally substituted —N(R2)2group, wherein the two R2together with the nitrogen atom to which they are attached form an optionally substituted cyclic aromatic group, such as an optionally substituted pyrrolyl, imidazolyl, pyrazolyl or triazolyl group. Typically in such an embodiment J is S, Q is O, and R1W1— is R1— wherein R1is as previously defined. Typically, the pyrrolyl, imidazolyl, pyrazolyl or triazolyl group is substituted at least at the 2- and 5-positions, wherein the 2,5-disubstituents are each independently selected from alkyl, cycloalkyl, alkoxy, cycloalkoxy, alkenyl, cycloalkenyl, alkynyl, acyl, aryl, alkylaryl, alkoxyaryl, heteroaryl, heterocyclyl, arylalkyl and heteroarylalkyl groups. More typically, the 2,5-disubstituents are each independently selected from alkyl or cycloalkyl groups, such as C3-C6branched or C3-C6cyclic alkyl groups, e.g. isopropyl, cyclopropyl, cyclohexyl or t-butyl groups. Alternatively, —W2R2may be wherein A1and A2are each independently selected from an optionally substituted alkylene or alkenylene group, which may optionally include one or more heteroatoms N, O or S in its carbon skeleton. Typically, each ring containing A1or A2is a five or a six membered ring. In a further embodiment, —W2R2has a formula selected from Example 4 below is an example of a compound where —W2R2is an optionally substituted —N(R2)2group, wherein the two R2together with the nitrogen atom to which they are attached form a cyclic aromatic group. In one embodiment, R1W1— comprises a heterocyclic group, wherein the heterocyclic group contains a single heteroatom in the heterocyclic ring, such as a nitrogen atom, wherein R1W1— may be optionally substituted, and —W2R2is a fused aryl or a fused heteroaryl group, wherein the aryl or heteroaryl group is fused to one or more cyclic hydrocarbon, heterocyclic, aryl or heteroaryl rings, wherein —W2R2may be optionally substituted. In another embodiment, R1W1— comprises a heterocyclic group containing a nitrogen atom and at least one further heteroatom in the heterocyclic ring, wherein R1W1— may be optionally substituted, wherein a nitrogen atom of R1W1— is linked to J, and wherein —W2R2is a monocyclic aryl or a monocyclic heteroaryl group, wherein the monocyclic aryl or the monocyclic heteroaryl group is substituted at the α and α′ positions, wherein the substituents at the α and α′ positions are independently selected from alkyl, cycloalkyl, alkoxy, cycloalkoxy, alkenyl, cycloalkenyl, alkynyl, acyl, aryl, alkylaryl, alkoxyaryl, heteroaryl, heterocyclyl, arylalkyl and heteroarylalkyl groups, wherein —W2R2may optionally be further substituted. In one embodiment of the compound of formula (I), at least one of W1and R1or W2and R2combine to form a moiety selected from the group consisting of: wherein, each dashed line may independently be a bond;T is O or S;A, B, D, E, W, X, Y and Z, when present, may each be independently selected from O, C(R3), C(R3)2, N, N(R3) and S;each incidence of R3is independently selected from the group consisting of hydrogen, halide, cyano, C1-C6alkyl, C1-C6trifluoroalkyl, C1-C6alkoxy, C═O, SO2, acyl, amino, hydroxyl, C5-C6heteroaryl, C5-C6heterocyclyl and C3-C6cycloalkyl, each of which may be optionally substituted as appropriate; andn is 0, 1, 2 or 3. It will be appreciated that each ring shown in the above structures with an R3group extending therefrom indicates that an R3group may extend from one or more or all of the available positions on said ring for substitution. Each of these ‘nitrogen-linked’ moieties may be combined with any of the ‘carbon-linked’ moieties described for the compound of the first aspect to form the respective R1and R2combination to give the final structure. In one embodiment, the compound of formula (I) is selected from a compound of formula (II), (III), (IV), (V) or (VI) or a pharmaceutically acceptable salt, solvate or prodrug thereof: wherein W1and W2, if present, and R1and R2are as described in any one or more of the embodiments described for the first aspect;each incidence of R15is independently selected from C1to C4alkyl, C1to C4hydroxylalkyl and C3to C5cycloalkyl; andA is optionally substituted heteroaryl or heterocycle, as previously defined, linked to the sulfonyl sulphur through a ring nitrogen. In one embodiment of any one of formula II to VI, R1or A may be selected from the group consisting of pyrazole, imidazole, triazole, tetrazole, pyrrole, morpholine, piperazine, 4-methyl piperazine, and fused bicyclics or tricyclics comprising a benzene ring fused with at least one 5-membered heterocycle, in one embodiment an indole, each of which may be substituted or unsubstituted. In certain embodiments of any one of formula II to VI, R1or A may be pyrazole or triazole optionally substituted at a ring atom with a group selected from halo, isopropyl, morpholinyl, piperidinyl, and piperazinyl, each of which groups may themselves be optionally substituted with C1-C6alkyl. In one embodiment, R15is selected from isopropyl, cyclopropyl and C3to C5hydroxylalkyl. In one embodiment, A is C3-C8heteroaryl or heterocyclyl, each of which may be optionally substituted. In one embodiment, A is C4-C7heteroaryl or heterocyclyl, each of which may be optionally substituted. In one embodiment, A is selected from C5or C6heteroaryl or heterocyclyl, each of which may be optionally substituted. A may be selected from pyrazole, imidazole, triazole, tetrazole, pyrrole, morpholine, piperazine, 4-methyl piperazine, and fused bicyclics or tricyclics comprising a benzene ring fused with at least one 5-membered heterocycle, such as indole, all of which groups may be optionally substituted at a ring atom with a group selected from halo, isopropyl, morpholinyl, piperidinyl, and piperazinyl, each of which groups may themselves be optionally substituted with C1-C6alkyl. In one embodiment, the compound of formula (I), (II), (III), (IV) (V) or (VI) is selected from the group consisting of: In one embodiment of the first aspect, when J is sulphur, Q is oxo, W2is carbon and R2is cycloalkane, heterocycle or aryl, then R2is not a monocyclic cycloalkane, heterocycle or aryl group. In one embodiment of the first aspect, when J is sulphur, Q is oxo, and W2is carbon, then R2is not an alkyl group. In one embodiment of the first aspect, when J is sulphur, Q is oxo, and W2is a carbon which is part of a ring system, then R2is not a substituted phenyl group. In one embodiment of the first aspect, when J is sulphur, Q is oxo, W1and R1together form an alkylamine or dialkylamine and W2is a carbon which is part of a ring system, then R2is not a substituted or unsubstituted phenyl, a tetrahydrobenzothiophene, or other bicyclic thiophene, a pyridine or a pyrimidine group. In one embodiment of the first aspect, when J is sulphur, Q is oxo, W1is a nitrogen as part of a piperidine or morpholine R1group and W2is a carbon which is part of a ring system, then R2is not a pyridine or pyrimidine group. In one embodiment of the first aspect, when J is sulphur, Q is oxo, W1is a nitrogen as part of a piperidine, piperazine, morpholine, pyrazole, imidazole, pyrrolidine, isoquinoline or thienopyridine R1group and W2is a carbon which is part of a ring system, then R2is not a tetrahydrobenzothiophene, or other bicyclic thiophene, or a methyl-substituted pyridine group. In one embodiment of the first aspect, when J is sulphur, Q is oxo, W1is a nitrogen and W2is a carbon which is part of a ring system, then R2may be an indacene, or substituted or hydrogenated variant thereof, or a phenyl substituted with at least one group selected from halo, C1-C4alkyl and C3-C5cycloalkyl. In certain embodiments, the indacene may be a hexahydroindacene and the substituted phenyl group may be selected from 2,6-diisopropyl-4-chlorophenyl, 2,6-dicyclopropylphenyl and 2,6-dicyclopropyl-4-chloro-phenyl. In one embodiment of the first aspect, when J is sulphur, Q is oxo, W1and R1together are selected from alkylamine, dialkylamine, arylamine, diarylamine, piperidine, morpholine, thiomorpholine, pyridine, pyrazole, azepine, hydroazepine, imidazole, pyrrolidine, isoquinoline or thienopyridine, then W2and R2together are not any group selected, independently, from substituted or unsubstituted phenyl, alkyl, cycloalkyl, pyrimidine, and a triazine group. In one specific embodiment, the compound of formula (I), (II), (III), (IV), (V) or (VI) may not be a compound selected from the group consisting of: In one embodiment, the compound of formula (I) has a molecular weight of from 200 to 2000 Da. Preferably the compound of formula (I) has a molecular weight of from 300 to 1000 Da. More preferably, the compound of formula (I) has a molecular weight of from 350 to 500 Da. The compounds of the present invention may provide one or more benefits over prior art sulfonyl ureas selected from: improved microsomal stability; improved permeability; reduced Pgp liability; reduced plasma protein binding; increased half-life; improved oral bioavailability; improved AUC; improved Cmax; reduced Cyp inhibition; and improved solubility. In one embodiment, the compounds of formula (I) offer improved pharmacokinetic characteristics. CRID3, a known sulfonylurea, has a half-life of 3.2 hours (mouse) which may lead to substantial trough levels from QD or BD dosing when the t½ is extrapolated to man. The compounds of formula (I) may differ in, for example, their protein binding, metabolism and oral availability. In one embodiment, the compounds of formula (I) have a tPSA of less than 90 Å2. In one further embodiment, the compounds of formula (I) have a tPSA of less than 90 Å2and a molecular weight of less than 405. In some embodiments of the present invention, therapeutically inactive prodrugs are provided. Prodrugs are compounds which, when administered to a mammal, are converted in whole or in part to a compound of the invention. In most embodiments, the prodrugs are pharmacologically inert chemical derivatives that can be converted in vivo to the active drug molecules to exert a therapeutic effect. Any of the compounds described herein can be administered as a prodrug to increase the activity, bioavailability, or stability of the compound or to otherwise alter the properties of the compound. Typical examples of prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound. Prodrugs include, but are not limited to, compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, and/or dephosphorylated to produce the active compound. In certain embodiments, the compounds of formula (I) may exhibit improved properties compared to known anti-diabetes drugs. Such compounds of formula (I) may be viewed as very potent versions of current sulfonylurea anti-diabetes drugs. Known diabetes drugs do not target NLRP3 to any therapeutically significant extent and so it would be necessary to use very high doses to have any significant effect on the NLRP3 inflammasome. The compounds of formula (I), show advantageously improved properties in a significant decrease in IC50 versus the NLRP3 inflammasome and additionally have the benefits, not realised by existing diabetes drugs, associated with NLRP3 inhibition such as improved wound healing and other advantages described herein. In one embodiment, the compound of formula (I) displaying these improved properties is selected from the group consisting of: In a further embodiment, one or more of the compounds of formula (I) may be useful as photoswitchable compounds which may applied in a range of uses including but not limited to insulin release. In certain embodiments of the invention one or more compounds of formula (I) may be appropriate for use as probes, such as photoaffinity probes, or as reactive intermediates which can be modified either directly or by means of a linking moiety to give biotinylated, fluorescent or photoaffinity probes. It will be appreciated that the compounds of formula (I) may be modified or derivatised by means well understood in the art to allow linkage to a molecule such as biotin, or a fluorescent group or photoaffinity label. A number of prodrug ligands are known. In general, alkylation, acylation, or other lipophilic modification of one or more heteroatoms of the compound, such as a free amine or carboxylic acid residue, may reduce polarity and allow for the compound's passage into cells. Examples of substituent groups that can replace one or more hydrogen atoms on a free amine and/or carboxylic acid moiety include, but are not limited to, the following: aryl; steroids; carbohydrates (including sugars); 1,2-diacylglycerol; alcohols; acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester (including alkyl or arylalkyl sulfonyl, such as methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as provided in the definition of an aryl given herein); optionally substituted arylsulfonyl; lipids (including phospholipids); phosphatidylcholine; phosphocholine; amino acid residues or derivatives; amino acid acyl residues or derivatives; peptides; cholesterols; or other pharmaceutically acceptable leaving groups which, when administered in vivo, provide the free amine. Any of these moieties can be used in combination with the disclosed active agents to achieve a desired effect. In some embodiments, compounds with one or more chiral centers are provided. While racemic mixtures of compounds of the invention may be active, selective, and bioavailable, isolated isomers may be of interest as well. The compounds disclosed herein as active agents may contain chiral centers, which may be either of the (R) or (S) configuration, or which may comprise a mixture thereof. Accordingly, the present invention also includes stereoisomers of the compounds described herein, where applicable, either individually or admixed in any proportions. Stereoisomers may include, but are not limited to, enantiomers, diastereomers, racemic mixtures, and combinations thereof. Such stereoisomers can be prepared and separated using conventional techniques, either by reacting enantiomeric starting materials, or by separating isomers of compounds and prodrugs of the present invention. Isomers may include geometric isomers. Examples of geometric isomers include, but are not limited to, cis isomers or trans isomers across a double bond. Other isomers are contemplated among the compounds of the present invention. The isomers may be used either in pure form or in admixture with other isomers of the compounds described herein. Various methods are known in the art for preparing optically active forms and determining activity. Such methods include standard tests described herein and other similar tests which are well known in the art. Examples of methods that can be used to obtain optical isomers of the compounds according to the present invention include the following:i) physical separation of crystals whereby macroscopic crystals of the individual enantiomers are manually separated. This technique may particularly be used when crystals of the separate enantiomers exist (i.e., the material is a conglomerate), and the crystals are visually distinct;ii) simultaneous crystallization whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state;iii) enzymatic resolutions whereby partial or complete separation of a racemate is achieved by virtue of differing rates of reaction for the enantiomers with an enzyme;iv) enzymatic asymmetric synthesis, a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer;v) chemical asymmetric synthesis whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries;vi) diastereomer separations whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers. The resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer;vii) first- and second-order asymmetric transformations whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer. The desired enantiomer is then released from the diastereomers;viii) kinetic resolutions comprising partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions;ix) enantiospecific synthesis from non-racemic precursors whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis;x) chiral liquid chromatography whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase. The stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions;xi) chiral gas chromatography whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase;xii) extraction with chiral solvents whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent; andxiii) transport across chiral membranes whereby a racemate is placed in contact with a thin membrane barrier. The barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through. The compound optionally may be provided in a composition that is enantiomerically enriched, such as a mixture of enantiomers in which one enantiomer is present in excess, in particular, to the extent of 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more, including 100%. The terms (R), (S), (R,R), (S,S), (R,S) and (S,R) as used herein mean that the composition contains a greater proportion of the named isomer of the compound in relation to other isomers. In a preferred embodiment, these terms indicate that the composition contains at least 90% by weight of the named isomer and 10% by weight or less of the one or more other isomers; or more preferably about 95% by weight of the named isomer and 5% or less of the one or more other isomers. In some embodiments, the composition may contain at least 99% by weight of the named isomer and 1% or less by weight of the one or more other isomers, or may contain 100% by weight of the named isomer and 0% by weight of the one of more other isomers. These percentages are based on the total amount of the compound of the present invention present in the composition. The compounds of the present invention may be utilized per se or in the form of a pharmaceutically acceptable ester, amide, salt, solvate, prodrug, or isomer. For example, the compound may be provided as a pharmaceutically acceptable salt. If used, a salt of the drug compound should be both pharmacologically and pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare the free active compound or pharmaceutically acceptable salts thereof and are not excluded from the scope of this invention. Such pharmacologically and pharmaceutically acceptable salts can be prepared by reaction of the drug with an organic or inorganic acid, using standard methods detailed in the literature. Examples of pharmaceutically acceptable salts of the compounds useful according to the invention include acid addition salts. Salts of non-pharmaceutically acceptable acids, however, may be useful, for example, in the preparation and purification of the compounds. Suitable acid addition salts according to the present invention include organic and inorganic acids. Preferred salts include those formed from hydrochloric, hydrobromic, sulfuric, phosphoric, citric, tartaric, lactic, pyruvic, acetic, succinic, fumaric, maleic, oxaloacetic, methanesulfonic, ethanesulfonic, p-toluenesulfonic, benzenesulfonic, and isethionic acids. Other useful acid addition salts include those formed with propionic acid, glycolic acid, oxalic acid, malic acid, malonic acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, and the like. Particular examples of pharmaceutically acceptable salts include, but are not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ-hydroxybutyrates, glycolates, tartrates, methanesulfonates, propanesulfonates, naphthalene-1-sulfonates, naphthalene-2-sulfonates, and mandelates. An acid addition salt may be reconverted to the free base by treatment with a suitable base. Preparation of basic salts of acid moieties which may be present on a compound or prodrug useful according to the present invention may be prepared in a similar manner using a pharmaceutically acceptable base, such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, triethylamine, or the like. Esters of the active agent compounds according to the present invention may be prepared through functionalization of hydroxyl and/or carboxyl groups that may be present within the molecular structure of the compound. Amides and prodrugs may also be prepared using techniques known to those skilled in the art. For example, amides may be prepared from esters, using suitable amine reactants, or they may be prepared from an anhydride or an acid chloride by reaction with ammonia or a lower alkyl amine. Moreover, esters and amides of compounds of the invention can be made by reaction with a carbonylating agent (e.g., ethyl formate, acetic anhydride, methoxyacetyl chloride, benzoyl chloride, methyl isocyanate, ethyl chloroformate, methanesulfonyl chloride) and a suitable base (e.g., 4-dimethylaminopyridine, pyridine, triethylamine, potassium carbonate) in a suitable organic solvent (e.g., tetrahydrofuran, acetone, methanol, pyridine, N,N-dimethylformamide) at a temperature of 0° C. to 60° C. Prodrugs are typically prepared by covalent attachment of a moiety, which results in a compound that is therapeutically inactive until modified by an individual's metabolic system. Examples of pharmaceutically acceptable solvates include, but are not limited to, compounds according to the invention in combination with water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine. In the case of solid compositions, it is understood that the compounds used in the methods of the invention may exist in different forms. For example, the compounds may exist in stable and metastable crystalline forms and isotropic and amorphous forms, all of which are intended to be within the scope of the present invention. If a compound useful as an active agent according to the invention is a base, the desired salt may be prepared by any suitable method known to the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, pyranosidyl acids such as glucuronic acid and galacturonic acid, alpha-hydroxy acids such as citric acid and tartaric acid, amino acids such as aspartic acid and glutamic acid, aromatic acids such as benzoic acid and cinnamic acid, sulfonic acids such a p-toluenesulfonic acid or ethanesulfonic acid, or the like. If a compound described herein as an active agent is an acid, the desired salt may be prepared by any suitable method known to the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal or alkaline earth metal hydroxide or the like. Illustrative examples of suitable salts include organic salts derived from amino acids such as glycine and arginine, ammonia, primary, secondary and tertiary amines, and cyclic amines such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminium and lithium. According to a second aspect of the invention there is provided a pharmaceutical composition comprising a compound of the first aspect disclosed herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, diluent and/or excipient. Suitably, the pharmaceutically acceptable carrier, diluent and/or excipient may be or include one or more of diluents, solvents, pH buffers, binders, fillers, emulsifiers, disintegrants, polymers, lubricants, oils, fats, waxes, coatings, viscosity-modifying agents, glidants and the like. The salt forms of the compounds of the invention are especially useful due to their improved solubility. In one embodiment, the pharmaceutical composition includes a cyclodextrin. The cyclodextrin may be selected from alpha, beta or gamma cyclodextrins. In one embodiment, the cyclodextrin is selected from a methyl cyclodextrin, a hydroxypropyl cyclodextrin and a sulfobutylether cyclodextrin. It has been found that cyclodextrins provide significant advantages in formulation and delivery of the compounds of the invention. Cyclodextrin formulations such as for example, one or more compounds of the invention with hydroxypropyl beta cyclodextrin or methyl beta cyclodextrin, may have uses in cholesterol sequestration/cholesterol lowering or via NLRP3 inhibition for Non-alcoholic steatohepatitis (NASH) and also in Alzheimer's Disease (AD). Diluents may include one or more of microcrystalline cellulose, lactose, mannitol, calcium phosphate, calcium sulfate, kaolin, dry starch, powdered sugar, and the like. Binders may include one or more of povidone, starch, stearic acid, gums, hydroxypropylmethyl cellulose and the like. Disintegrants may include one or more of starch, croscarmellose sodium, crospovidone, sodium starch glycolate and the like. Solvents may include one or more of ethanol, methanol, isopropanol, chloroform, acetone, methylethyl ketone, methylene chloride, water and the like. Lubricants may include one or more of magnesium stearate, zinc stearate, calcium stearate, stearic acid, sodium stearyl fumarate, hydrogenated vegetable oil, glyceryl behenate and the like. A glidant may be one or more of colloidal silicon dioxide, talc or corn starch and the like. Buffers may include phosphate buffers, borate buffers and carbonate buffers, although without limitation thereto. Fillers may include one or more gels inclusive of gelatin, starch and synthetic polymer gels, although without limitation thereto. Coatings may comprise one or more of film formers, solvents, plasticizers and the like. Suitable film formers may be one or more of hydroxypropyl methyl cellulose, methyl hydroxyethyl cellulose, ethyl cellulose, hydroxypropyl cellulose, povidone, sodium carboxymethyl cellulose, polyethylene glycol, acrylates and the like. Suitable solvents may be one or more of water, ethanol, methanol, isopropanol, chloroform, acetone, methylethyl ketone, methylene chloride and the like. Plasticizers may be one or more of propylene glycol, castor oil, glycerin, polyethylene glycol, polysorbates, and the like. Reference is made to the Handbook of Excipients 6thEdition, Eds. Rowe, Sheskey & Quinn (Pharmaceutical Press), which provides non-limiting examples of excipients which may be useful according to the invention. It will be appreciated that the choice of pharmaceutically acceptable carriers, diluents and/or excipients will, at least in part, be dependent upon the mode of administration of the formulation. By way of example only, the composition may be in the form of a tablet, capsule, caplet, powder, an inhalable liquid (e.g. solution, suspension), an injectable liquid, a suppository, a slow release formulation, an osmotic pump formulation or any other form that is effective and safe for administration. Suitably, the pharmaceutical composition is for the treatment or prevention of a disease, disorder or condition in a mammal. A third aspect of the invention resides in a method of treatment or prevention of a disease, disorder or condition including the step of administering an effective amount of a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect, to thereby treat or prevent the disease, disorder or condition. A fourth aspect of the invention provides for a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect, for use in the treatment or prevention of a disease, disorder or condition. A fifth aspect of the invention provides for use of a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, in the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition. As generally used herein, the terms “administering” or “administration”, and the like, describe the introduction of the compound or composition to a mammal such as by a particular route or vehicle. Routes of administration may include topical, parenteral and enteral which include oral, buccal, sub-lingual, nasal, anal, gastrointestinal, subcutaneous, intramuscular and intradermal routes of administration, although without limitation thereto. By “treat”, “treatment” or “treating” is meant administration of the compound or composition to a subject to at least ameliorate, reduce or suppress existing signs or symptoms of the disease, disorder or condition experienced by the subject. By “prevent”, “preventing” or “preventative” is meant prophylactically administering the formulation to a subject such as a mammal who does not exhibit signs or symptoms of a disease, disorder or condition, but who is expected or anticipated to likely exhibit such signs or symptoms in the absence of prevention. Preventative treatment may at least lessen or partly ameliorate expected symptoms or signs. As used herein, “effective amount” refers to the administration of an amount of the relevant active agent sufficient to prevent the occurrence of symptoms of the condition being treated, or to bring about a halt in the worsening of symptoms or to treat and alleviate or at least reduce the severity of the symptoms. The effective amount will vary in a manner which would be understood by a person of skill in the art with patient age, sex, weight, etc. An appropriate dosage or dosage regime can be ascertained through routine trial. As used herein, the terms “subject” or “individual” or “patient” may refer to any mammalian subject. Mammals may include, but are not restricted to, primates, livestock animals (e.g. sheep, cows, horses, donkeys, pigs), laboratory test animals (e.g. rabbits, mice, rats, guinea pigs, hamsters), companion animals (e.g. cats, dogs) and captive wild animals (e.g. foxes, deer, dingoes). A preferred subject is a human in need of treatment for a disease, disorder or condition as described herein. However, it will be understood that the aforementioned terms do not imply that symptoms are necessarily present. In one particular embodiment, the disease, disorder or condition is one which is responsive to inhibition of activation of the NLRP3 inflammasome. According to this embodiment, the compound of the first aspect, or pharmaceutically effective salt, solvate or prodrug thereof is a specific inhibitor of NLRP3. In a further embodiment, the disease, disorder or condition is responsive to modulation of one or more of IL-1β, IL-17, IL-18, IL-1α, IL-37, IL-33 and Th17 cells. In one embodiment, the modulation is inhibition of one or more of IL-1β, IL-17, IL-18, IL-1α, IL-37, and IL-33. In one embodiment, the modulation of Th17 cells, is by inhibition of production and/or secretion of IL-17. In general embodiments, the disease, disorder or condition is a disease, disorder or condition of the immune system, the cardiovascular system, the endocrine system, the gastrointestinal tract, the renal system, the respiratory system, the central nervous system, is a cancer or other malignancy and/or is caused by or associated with a pathogen. It will be appreciated that these general embodiments defined according to broad categories of diseases, disorders and conditions are not mutually exclusive. In this regard any particular disease, disorder or condition may be categorized according to more than one of the above general embodiments. A non-limiting example is Type I diabetes which is an autoimmune disease and a disease of the endocrine system. In one embodiment, the disease, disorder or condition is of the immune system. In particular embodiments, the disease, disorder or condition is an inflammatory disease, disorder or condition or an autoimmune disease, disorder or condition. In one embodiment, the disease, disorder or condition is of the skin. In one embodiment, the disease, disorder or condition is of the cardiovascular system. In one embodiment, the disease, disorder or condition is a cancer, tumour or other malignancy. As used herein, cancers tumours and malignancies, refer to diseases disorders or conditions, or to cells or tissues associated with the diseases, disorders or conditions, characterized by aberrant or abnormal cell proliferation, differentiation and/or migration often accompanied by an aberrant or abnormal molecular phenotype that includes one or more genetic mutations or other genetic changes associated with oncogenesis, expression of tumour markers, loss of tumour suppressor expression or activity and/or aberrant or abnormal cell surface marker expression. In general embodiments, cancers, tumours and malignancies may include sarcomas, lymphomas, leukemias, solid tumours, blastomas, gliomas, carcinomas, melanomas and metastatic cancers, although without limitation thereto. A more comprehensive listing of cancers tumours and malignancies may be found at the National Cancer Institute's website http://www.cancer.gov/cancertopics/types/alphalist. In one embodiment, the disease, disorder or condition is of the renal system. In one embodiment, the disease, disorder or condition is of the gastro-intestinal tract. In one embodiment, the disease, disorder or condition is of the respiratory system. In a further embodiment, the disease, disorder or condition is of the endocrine system. In one embodiment, the disease, disorder or condition is of the central nervous system (CNS). In one embodiment, the disease, disorder or condition is caused by, or is associated with, a pathogen. The pathogen may be a virus, a bacterium, a protist, a worm or a fungus or any other organism capable of infecting a mammal, although without limitation thereto. Non-limiting examples of viruses include influenza virus, cytomegalovirus, Epstein Barr Virus, human immunodeficiency virus (HIV), alphavirus such as Chikungunya and Ross River virus, flaviviruses such as Dengue virus, Zika virus and papillomavirus, although without limitation thereto. Non-limiting examples of pathogenic bacteria includeStaphylococcus aureus, Helicobacter pylori, Bacillus anthracis, Bordatella pertussis, Corynebacterium diptheriae, Clostridium tetani, Clostridium botulinum, Streptococcus pneumoniae, Streptococcus pyogenes, Listeria monocytogenes, Hemophilus influenzae, Pasteurelia multicida, Shigella dysenteriae, Mycobacterium tuberculosis, Mycobacterium leprae, Mycoplasma pneumoniae, Mycoplasma homrinis, Neisseria meningitidis, Neisseria gonorrhoeae, Rickettsia rickettsii, Legionella pneumophila, Klebsiella pneumoniae, Pseudomonas aeruginosa, Propionibacterium acnes, Treponema pallidum, Chlamydia trachomatis, Vibrio cholerae, Salmonella typhimurium, Salmonella typhi, Borrelia burgdorferiandYersinia pestis, although without limitation thereto. Non-limiting examples of protists includePlasmodium, Babesia, Giardia, Entamoeba, Leishmaniaand Trypanosomes, although without limitation thereto. Non-limiting examples of worms include helminths inclusive of schistisimes, roundworms, tapeworms and flukes, although without limitation thereto. Non-limiting examples of fungi includeCandidaandAspergillusspecies, although without limitation thereto. Further relevant diseases, disorders or conditions may be selected from the group consisting of those recited in the journal article Menu et al., Clinical and Experimental Immunology, 166, 1-15, 2011, found at: http://onlinelibrary.wiley.com/store/10.1111/j.1365-2249.2011.04440.x/asset/j.1365-2249.2011.04440.x.pdf?v=1&t=i60c1phf&s=d26f50a2622926cc6b4bc855bd911ae9dc9750cf. In particular embodiments, the disease, disorder or condition is selected from the group consisting of constitutive inflammation including the cryopyrin-associated periodic syndromes (CAPS): Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome (FCAS) and neonatal-onset multisystem inflammatory disease (NOMID); including autoinflammatory diseases: familial Mediterranean fever (FMF), TNF receptor associated periodic syndrome (TRAPS), mevalonate kinase deficiency (MKD), hyperimmunoglobulinemia D and periodic fever syndrome (HIDS), deficiency of interleukin 1 receptor (DIRA) antagonist, Majeed syndrome, pyogenic arthritis, pyoderma gangrenosum and acne syndrome (PAPA), haploinsufficiency of A20 (HA20), pediatric granulomatous arthritis (PGA), PLCG2-associated antibody deficiency and immune dysregulation (PLAID), PLCG2-associated autoinflammation, antibody deficiency and immune dysregulation (APLAID) and sideroblastic anemia with B-cell immunodeficiency, periodic fevers, and developmental delay (SIFD); autoimmune diseases including multiple sclerosis (MS), type-1 diabetes, psoriasis, rheumatoid arthritis, Behcet's disease, Sjogren's syndrome and Schnitzler syndrome; macrophage activation syndrome; Blau syndrome; respiratory diseases including chronic obstructive pulmonary disorder (COPD), asthma such as allergic asthma and steroid-resistant asthma, asbestosis, silicosis and cystic fibrosis; dermatitis including contact dermatitis; central nervous system diseases including Parkinson's disease, Alzheimer's disease, motor neuron disease, Huntington's disease, cerebral malaria and brain injury from pneumococcal meningitis; metabolic diseases including Type 2 diabetes, atherosclerosis, obesity, gout, pseudo-gout; ocular diseases including those of the ocular epithelium, age-related macular degeneration (AMD), uveitis, corneal infection and dry eye; kidney disease including chronic kidney disease, oxalate nephropathy, nephrocalcinosis and diabetic nephropathy; liver disease including non-alcoholic steatohepatitis (NASH) and alcoholic liver disease; inflammatory reactions in skin including contact hypersensitivity and sunburn; inflammatory reactions in the joints including osteoarthritis, systemic juvenile idiopathic arthritis, adult-onset Still's disease, relapsing polychondritis; viral infections including alpha virus (Chikungunya, Ross River) and flavivirus (Dengue, Zika), flu, HIV; hidradenitis suppurativa (HS) and other cyst-causing skin diseases; cancers including lung cancer metastasis, pancreatic cancers, gastric cancers, myelodisplastic syndrome, leukemia; polymyositis; stroke including ischemic stroke; myocardial infarction including recurrent myocardial infarction; congestive heart failure; embolism; cardiovascular disease; Graft versus Host Disease; hypertension; colitis; helminth infection; bacterial infection; abdominal aortic aneurism; wound healing; depression, psychological stress; ischaemia reperfusion injury and any disease where an individual has been determined to carry a germline or somatic non-silent mutation in NLRP3. In one embodiment, the disease, disorder or condition is an autoinflammatory disease such as cryopyrin-associated periodic syndromes (CAPS), Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome (FCAS), familial Mediterranean fever (FMF), Neonatal onset multisystem inflammatory disease (NOMID), Tumor Necrosis Factor (TNF) Receptor-Associated Periodic Syndrome (TRAPS), hyperimmunoglobulinemia D and periodic fever syndrome (HIDS), deficiency of interleukin 1 receptor antagonist (DIRA), Majeed syndrome, or pyogenic arthritis, pyoderma gangrenosum and acne syndrome (PAPA). In another embodiment, the disease, disorder or condition is Parkinson's disease or Huntington's disease. In another embodiment, the disease, disorder or condition is gout or juvenile idiopathic arthritis. In another embodiment, the disease, disorder or condition is non-alcoholic steatohepatitis (NASH). In another embodiment, the disease, disorder or condition is oxalate nephropathy or nephrocalcinosis. In another embodiment, the disease, disorder or condition is uveitis. In another embodiment, the disease, disorder or condition is hidradenitis suppurativa (HS). In another embodiment, the disease, disorder or condition is myelodisplastic syndrome, macrophage activation syndrome, Schnitzler syndrome, adult-onset Still's disease, or Behget's Disease. In one non-limiting example of those described, the disease, disorder or condition being treated is NASH. NLRP3 inflammasome activation is central to inflammatory recruitment in NASH, and inhibition of NLRP3 may both prevent and reverse liver fibrosis. Compounds of the present invention, by interrupting the function of NLRP3 inflammasomes in liver tissue, can cause histological reductions in liver inflammation, decreased recruitment of macrophages and neutrophils, and suppression of NF-κB activation. Inhibition of the NLRP3 can reduce hepatic expression of pro-IL-1β and normalized hepatic and circulating IL-1β, IL-6 and MCP-1 levels thereby assisting in treatment of the disease. In a further non-limiting example of those described, the disease, disorder or condition being treated is severe steroid resistant (SSR) asthma. Respiratory infections induce an NLRP3 inflammasome/caspase-1/IL-1β signaling axis in the lungs that promotes SSR asthma. The NLRP3 inflammasome recruits, and activates, pro-caspase-1 to induce IL-1β responses. NLRP3 inflammasome-induced IL-1β responses are therefore important in the control of infections, however, excessive activation results in aberrant inflammation and has been associated with the pathogenesis of SSR asthma and COPD. The administration of compounds of the first aspect that target specific disease processes, are more therapeutically attractive than non-specifically inhibiting inflammatory responses with steroids or IL-1β. Targeting the NLRP3 inflammasome/caspase-1/IL-1β signaling axis with the compounds of the first aspect may therefore be useful in the treatment of SSR asthma and other steroid-resistant inflammatory conditions. In one further non-limiting example of those described, the disease, disorder or condition being treated is Parkinson's disease. Parkinson's is the most common neurodegenerative movement disorder and is characterized by a selective loss of dopaminergic neurons, accompanied by the accumulation of mis-folded α-synuclein (Syn) into Lewy bodies that are pathological hallmarks of the disease. Chronic microglial neuroinflammation is evident early in the disease, and has been proposed to drive pathology. A central role for microglial NLRP3 is postulated in Parkinson's progression. The NLRP3 inflammasome is activated by fibrillar Syn via a Syk kinase dependent mechanism, and also occurs in the absence of Syn pathology at the early stages of dopaminergic degeneration, and drives neuronal loss. The compounds of the first aspect may block NLRP3 inflammasome activation by fibrillar Syn or mitochondrial dysfunction and thereby confer effective neuroprotection of the nigrostriatal dopaminergic system and assist with treatment of Parkinson's. In a sixth aspect of the invention there is provided a method of diagnosing a disease, disorder or condition in a mammal including the step of administering a labelled compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, to the mammal or to a biological sample obtained from the mammal to facilitate diagnosis of the disease, disorder or condition in the mammal. Inflammasome activation, in particular that of the NLRP3 inflammasome, is known to drive initiation, progression and chronic development of a vast number of inflammatory diseases. The sulfonylureas and related compounds of the first aspect are potent and specific direct inhibitors of NLRP3. Accordingly, a chemical probe specific for NLRP3, which is present in immune cells during inflammation has potential utility in diagnosing inflammatory and other related diseases. An NRLP3 activation probe comprising a compound of the first aspect could act as an effective surrogate biomarker of inflammatory disease for ex vivo (blood) or in vivo (MRI, PET etc.) diagnostics. A compound of the first aspect (or a pharmaceutically effective salt, solvate or prodrug thereof) could also be used in other ex-vivo and/or in in-vitro diagnostic methods. The use of the compounds of formula (I) in diagnosing inflammatory and other related diseases may be achieved by near infrared fluorescent imaging and ex vivo characterisation of immune cells by degree of inhibition of IL-1beta, pro-caspase 1 cleavage and IL-18 levels. In particular, peripheral blood monocytes (PMBCs), macrophages, dendritic cells, CD4+T cells, Th17 cells, Th1 cells and Th2 cells are relevant. In vivo diagnostics may use magnetic resonance imaging (MRI), employing2H (deuterium),13C,19F and/or15N labelled variants of compounds of the present invention given to a patient IV, IM, SC, PO, topical, IT, etc. In vivo diagnostics using positron emission tomography (PET) are also appropriate. PET is a molecular imaging technique that requires specific probes radiolabelled with short-lived positron emitting radionuclides. Typical isotopes include11C,13N,15O,18F,64Cu,62Cu,124I,76Br,82Rb and68Ga, with18F being the most clinically utilized. In particular it is possible to produce in a simple manner a stable64Cu or62Cu salt of one or more of the compounds of formula (I) by simple ion exchange with a sodium (or other monovalent cation) salt of said compounds. This enables rapid preparation of a diagnostic probe for radioimaging, PET and the like whereby the intensity, location and temporal accretion of the diagnostic probe is able to identify the degree and/or the location of immune cells with activated NLRP3 as a surrogate biomarker of the patients inflammatory state, and site of inflammation within the body. They will also be useful for application to biological samples removed from the body i.e. in vitro diagnosis. FIG.1evidences complex formation between the sodium form of MCC950 (CRID3), a sulfonylurea, and copper chloride by using isothermal titration calorimetry (ITC). The results show that copper (II) ions form a strong complex with MCC950, comparable to the complex with EDTAx2Na and much stronger than with EDTA free acid. Formation of MCC950:Cu(II) complex was endothermic with enthalpy being positive which suggests that the process was entropy driven with the presence of strong hydrophobic interactions. This is a strong indication that compounds of formula (I), bearing the same core functional sulfonyl and urea moieties, will achieve the same degree of complexation thereby proving for their use in diagnostics, as described above. A seventh aspect of the invention resides in a method of modulating the activity of a biological target comprising the step of exposing the biological target to a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof. The method may be an ex-vivo or an in-vitro method. The biological target may be selected from the group consisting of NLRP3 inflammasome, IL-1β, IL-17, IL-18, IL-1α, IL-37, IL-33 and Th17 cells. Preferably the target is NLRP3 inflammasome. The modulation may be as described previously for the third to fifth aspects. As generally used herein, a biological sample may include cells, tissues, fluids, molecules or other biological materials obtained, or obtainable, from a mammal. Non-limiting examples include urine, blood and fractions thereof such as serum, plasma, lymphocytes and erythrocytes, cerebrospinal fluid, PAP smears, nasal and ocular secretions, amniotic fluid, faeces, semen, tissue and/or organ biopsies and nucleic acid (e.g. DNA, RNA) or protein samples, although without limitation thereto. The following experimental section describes in more detail the characterisation of certain of the compounds of the invention and their efficacy. The intention is to illustrate certain specific embodiments of the compounds of the invention and their efficacy without limiting the invention in any way. EXPERIMENTAL General Methods Method A: A1: To a solution of R2amine intermediate (1 eq.) with or without base such as, but not exclusively, triethylamine (1.2 eq.) in an anhydrous aprotic solvent such as, but not exclusively, tetrahydrofuran or dichloromethane was added triphosgene (0.4 to 1.1 eq.). The reaction was stirred at ambient temperature or, where necessary, heated at reflux until completion, typically from 2 to 18 h. A2: To di-t-butyldicarbonate (1.2-1.4 eq.) in anhydrous acetonitrile or THF was added DMAP (15-100 mol %), after 5 minutes, a solution of R2amine intermediate (1.0 eq.) in acetonitrile was added. The reaction mixture was stirred for 30-60 min at room temperature. Method B: B1: The R2carboxylic acid intermediate (1 eq.) was dissolved in an aprotic solvent such as toluene with or without 2 drops of DMF and a chlorinating agent such as thionyl chloride (2 eq.) added. The reaction mixture was heated at reflux until completion, then concentrated in vacuo to give the corresponding R2acid chloride intermediate. Alternative methods or forming the acid chloride are also equally useful here for example the above procedure can be carried out without toluene and DMF thereby using thionyl chloride as both solvent and chlorinating agent. The R2acid chloride intermediate was dissolved in acetone and added drop-wise to a solution of sodium azide (1.5 eq) in a water:acetone (50:50) solution at 0° C. Iced water was added to precipitate the resulting R2acylazide intermediate which was dissolved in toluene and dried (MgSO4) prior to adding the solution in a drop-wise fashion to anhydrous toluene at reflux while maintaining a constant flow of inert gas. The reaction was heated until completion, typically 2 h, to give the R2isocyanate. B2: The R2acid chloride (formed as indicated in method B1) in dry CH2Cl2was added NaN3(2.0 eq.) at 0° C. The reaction mixture was stirred at room temperature for 1 h and extracted into EtOAc. The organic layer was washed with H2O (15 mL), dried (MgSO4), and carefully evaporated to give acyl azide. The acyl azide was dissolved in dry toluene and heated to 100° C. for 2 h. The solvent was removed to give crude R2isocyanate. Method C: C1: R1sulfonamide intermediate (1 eq.) was dissolved in anhydrous THF and treated with NaH (1 eq.) under reduced pressure. The mixture was heated to reflux for 2 h then cooled to room temperature and R2isocyanate intermediate in THF added under nitrogen atmosphere. The reaction mixture was stirred at reflux until completion. C2: R1sulfonamide intermediate (1 eq.) was dissolved in anhydrous THF or anhydrous methanol and treated with NaH (1 eq.) under reduced pressure. Once effervescence ceased the R2isocyanate intermediate was added and the reaction mixture was stirred at ambient temperature overnight. C3: To R1sulfonamide intermediate (1 eq) in anhydrous THF (5 mL/mmol) was added NaH (1 eq) at 0° C. and stirred for 30 min to 2 h, or until completion, at ambient temperature under nitrogen atmosphere. Again cooled to 0° C., R2isocyanate (1.0 eq) in THF was added and stirred at ambient temperature until completion, typically 2 to 16 h. C4: To crude R2isocyanate (1.0 eq) in anhydrous THF or DCM (5-11 mL/mmol) was added R1sulfonamide (1.0 eq) followed by base such as triethylamine, DIPEA, or DBU (1-2 eq) and the reaction mixture stirred at ambient temperature overnight. C5: To R1sulfonamide intermediate (1 eq) in anhydrous MeOH (5 mL/mmol) was added NaOMe (1 eq) [alternatively: a 1.0 mM solution of freshly prepared sodium methoxide (1 eq) was added to a 1.0 mM solution of R1sulfonamide (1 eq) in anhydrous methanol]. The solvent was then removed in vacuo. The salt was suspended in anhydrous aprotic solvent such as acetonitrile or THF, the R2isocyanate (1.0 eq) in anhydrous aprotic solvent such as acetonitrile or THF was added and the mixture stirred at ambient temperature overnight. The solution was then heated at reflux until completion, typically 90 min. C6: R1sulfonamide (1.0 eq.) was dissolved in anhydrous THF under a nitrogen atmosphere. Solid sodium methoxide (1.0 eq mmol) was added in one portion. This mixture was stirred at ambient temperature for 3 h. A solution of the R2isocyanate (1.17 eq) in THF was added drop wise. The reaction mixture was stirred at room temperature overnight. Method D: A solution of amine (1.0 eq) in acetonitrile (7-12 mL/mmol) at 0° C. was treated with c.HCl (1.25-2.25 mL/mmol) in H2O (0.5-1.2 mL/mmol) followed by aqueous solution of NaNO2(1.2 eq) dissolved in H2O (0.3-0.5 mL/mmol of NaNO2). The resulting solution was stirred at 0° C. for 45 min. AcOH (0.5-1.2 mL/mmol), CuCl2·2H2O (0.5 eq) and CuCl (0.05 eq) were sequentially added to the above mixture and purged with SO2gas for 20 min at 0° C. The resulting reaction mixture was stirred at 0° C.-10° C. until completion. Method E: E1: A solution of sulfonyl chloride (1 eq) in THF (10-20 mL/mmol) was cooled to −78° C. and ammonia gas was bubbled through the solution for 15 min, stirring was continued for a further 30 min then allowed to warm to ambient temperature and stirred for 2 h or until completion. E2: A solution of sulfonyl chloride (1 eq) in acetone (20 mL/mmol) was treated with a solution of NH4HCO3(4 eq) dissolved in water (1.5 mL/mmol of NH4HCO3) at ambient temperature and stirred for 4 h or until completion. Method F General Procedure for the Synthesis of Triazoles Alkyne (1 eq) and azide (1.2 eq), 5 mol % CuSO4, 10 mol % NaAsc solution in DMSO (500 μL) were stirred at room temperature until completion, typically 12 h. Synthesis of R1Sulfonamide Intermediates: Morpholine (1.98 mL, 22.9 mmol) was added slowly to a mixture of sulfuryl chloride (5.5 mL, 68.8 mmol) in acetonitrile (15 mL) at ambient temperature. The resulting reaction mixture was heated to reflux for 24 h. The solvent was removed in vacuo and the residue azeotroped twice with toluene to give morpholine-4-sulfonyl chloride as a light yellow oil (2.8 g, 67%). The crude product was used directly in the next step without further purification.1H NMR (400 MHz, DMSO-d6): δ 3.79 (t, J=4.0 Hz, 4H), 3.28 (t, J=4.0 Hz, 4H). Morpholine-4-sulfonyl chloride (0.5 g, 4.3 mmol) in acetone (0.5 mL) was added to aq NH3(1.5 mL, NH4OH in H2O, 28% NH3basis) at 0° C. and stirred at same temperature for 2 h. The solvent was removed in vacuo and the residue azeotroped twice with toluene. The residue was purified by column chromatography on silica gel using 2% MeOH-DCM eluent to give morpholine-4-sulfonamide as white solid (270 mg, 60%).1H NMR (400 MHz, DMSO-d6): δ 6.82 (bs, 2H), 3.65 (t, J=4.0 Hz, 4H), 2.92 (t, J=4.0 Hz, 4H). 4-methylpiperazine-1-sulfonamide 1-Methylpiperazine (2.0 g, 19.9 mmol) was added slowly to a mixture of sulfuryl chloride (4.83 mL, 59.9 mmol) in acetonitrile (15 mL) at room temperature, the resulting reaction mixture was heated to reflux for 24 h. The solvent was removed in vacuo and the residue azeotroped twice with toluene to give 4-methylpiperazine-1-sulfonyl chloride hydrochloride salt as a brown solid (2.1 g, crude). The crude product was used directly in the next step without purification.1H NMR (400 MHz, DMSO-d6): δ=3.95 (bs, 2H), 3.60 (bs, 4H), 3.39-3.34 (m, 2H), 2.81 (3H, s). To a solution of 4-methylpiperazine-1-sulfonyl chloride hydrochloride in acetone (5.0 mL) was added aq NH3(5.0 mL, NH4OH in H2O, 28% NH3basis) at 0° C., the resulting reaction mixture was stirred at room temperature for about 2 h. The solvent was removed in vacuo and the residue azeotroped twice with toluene. The residue was purified by reverse phase column chromatography using acetonitrile/water as mobile phase to afford 4-methylpiperazine-1-sulfonamide as an off white solid (125 mg, 21%).1H NMR (400 MHz, DMSO-d6): δ=6.71 (bs, 2H), 2.91 (t, J=4.0 Hz, 4H), 2.34 (t, J=4.0 Hz, 4H), 2.15 (s, 3H). 1-isopropyl-1H-pyrazole-3-sulfonamide 1-Isopropyl-1H-pyrazol-3-amine was reacted to 1-isopropyl-1H-pyrazole-3-sulfonyl chloride, a brown liquid, using general method D (0.5 g, 43%).1H NMR (400 MHz, CDCl3): δ=7.55 (s, 1H), 6.88 (s, 1H), 4.66-4.63 (m, 1H), 3.6 (br.s., 2H), 1.59 (d, J=6.8 Hz, 6H). LCMS (m/z): 209.0 (M+1)+. The sulfonyl chloride was converted using general method E1 to give the titled compound as yellow solid (0.45 g, 82%).1H NMR (300 MHz, DMSO-d6): δ=7.9 (d, J=2.4 Hz, 1H), 7.36 (s, 2H), 6.55 (d, J=2.1 Hz, 1H), 4.57-4.53 (m, 1H), 1.42 (d, J=6.9 Hz, 6H). LCMS (m/z): 190.0 (M+1)+. Other R1sulphonamide intermediates are commercially available and/or may be prepared by routine synthetic methods. WO 2016/131098 for example (see pages 90-130) discloses the synthesis of the following R1sulphonamide intermediates which may be used in the synthesis of compounds of the present invention:CyclohexanesulfonamideCyclopentanesulfonamide5-((dimethylamino)methyl)furan-2-sulfonamideFuran-2-sulfonamide5-methylfuran-2-sulfonamide5-ethyl-N-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)furan-2-sulfonamide4-(prop-1-en-2-yl)furan-2-sulfonamided6-4-(prop-1-en-2-yl)furan-2-sulfonamide4-(prop-1-en-2-yl)furan-2-sulfonamide4-(2-hydroxypropan-2-yl)-5-methylfuran-2-sulfonamided6-4-(2-hydroxypropan-2-yl)-5-methylfuran-2-sulfonamide1-benzyl-1H-1,2,4-triazole-3-sulfonamide1-methyl-1H-pyrazole-3-sulfonamide1-(trifluoromethyl)-1H-pyrazole-3-sulfonamide1-isopropyl-1H-pyrazole-4-sulfonamide1-cyclopropyl-1H-pyrazole-3-sulfonamide1-(tert-butyl)-1H-pyrazole-3-sulfonamide1-cyclohexyl-1H-pyrazole-3-sulfonamide1-phenyl-1H-pyrazole-3-sulfonamide1-benzyl-1H-pyrazole-3-sulfonyl chloride1-(1-phenylethyl)-1H-pyrazole-3-sulfonamide1-(2-(piperidin-1-yl)ethyl)-1H-pyrazole-3-sulfonamide1,5-dimethyl-1H-pyrazole-3-sulfonamide1-methyl-5-(trifluoromethyl)-1H-pyrazole-3-sulfonamide1-isopropyl-5-(trifluoromethyl)-1H-pyrazole-3-sulfonamide5-isopropyl-1-methyl-1H-pyrazole-3-sulfonamide5-(2-hydroxypropan-2-yl)-1-methyl-1H-pyrazole-3-sulfonamide1-benzyl-5-(2-hydroxypropan-2-yl)-1H-pyrazole-3-sulfonamide5-(2-hydroxypropan-2-yl)-1-phenyl-1H-pyrazole-3-sulfonamide5-(dimethylamino)naphthalene-1-sulfonamideN-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-2,3-dihydrobenzo[b]thiophene-6-sulfonamide 1,1-dioxide3-azidobenzenesulfonamideN-(3-Sulfamoylphenyl)pent-4-ynamideBenzene-1,3-disulfonamideN1,N1-dimethylbenzene-1,3-disulfonamideMethyl 3-sulfamoylbenzoate3-(4-phenyl-1H-1,2,3-triazol-1-yl)benzenesulfonamideN-(prop-2-yn-1-yl)-3-(4-sulfamoylphenyl)propanamidebenzo[d][1,3]dioxole-5-sulfonamidePyridine-4-sulfonamidePyridine-3-sulfonamidePyridine-2-sulfonamide4-(trifluoromethyl)pyridine-2-sulfonamide3-(3-(trifluoromethyl)-3H-diazirin-3-yl)benzenesulfonamide2-(methyl(7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)amino)-N-(4-sulfamoylphenethyl)acetamide4-(2-(7-Nitrobenzo[c][1,2,5]oxadiazol-4-ylamino)ethyl)benzenesulfonamide2-(7-(Dimethylamino)-2-oxo-2H-chromen-4-yl)-N-(4-sulfamoylphenethyl)acetamide Synthesis of R1and R2Amine Intermediates 9H-carbazol-9-amine 9H-carbazole (2.0 g, 12 mmol) was dissolved in acetonitrile (80 mL) and acetic acid (20 mL) then cooled to 0° C. and c.HCl:water (4:2, 6 mL) added. The solution was treated with a solution of sodium nitrite (1 g, 14.4 mmol) in water (4 mL) drop-wise over 10 mins. The reaction was stirred at 0-10° C. for 3 hours or until completion then diluted with water and extracted using ethyl acetate. The organics were washed with water, brine then dried (Na2SO4) and concentrated in vacuo to give 9-nitro-9H-carbazole as a yellow solid used directly in the next reaction step. Zinc (9.7 g, 150 mmol) and ammonium chloride (8 g, 150 mmol) in THF (50 mL) and water (15 mL) was cooled to 0° C. and 9-nitro-9H-carbazole in THF (5 mL) was added dropwise and stirring continued for 2 h or until completion. The reaction was diluted using ethyl acetate and filtered through celite then the organic phase was washed using water, brine, dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica using 5% ethyl acetate:hexanes eluent to give the titled compound as a semi-solid (3.3 g, 42%).1H NMR (400 MHz, DMSO-d6) δ=8.10 (d, J=7.7 Hz, 1H), 7.59 (d, J=7.7 Hz, 1H), 7.45 (t, J=7.7 Hz, 1H), 7.17 (t, J=7.7 Hz, 1H), 5.83 (s, 1H). Other R1and R2amine intermediates are commercially available and/or may be prepared by routine synthetic methods. WO 2016/131098 for example (see pages 130-157) discloses the synthesis of the following R1and R2amine intermediates which may be used in the synthesis of compounds of the present invention:1-methyl-1H-pyrazol-3-amine HCl1-(trifluoromethyl)-1H-pyrazol-3-amine1-isopropyl-1H-pyrazol-3-amine1-cyclopropyl-1H-pyrazol-3-amine1-(tert-butyl)-1H-pyrazol-3-amine1-cyclohexyl-1H-pyrazol-3-amine1-phenyl-1H-pyrazol-3-amine1-benzyl-1H-pyrazol-3-amine1-(1-phenylethyl)-1H-pyrazol-3-amine1-(2-(piperidin-1-yl)ethyl)-1H-pyrazol-3-amine1,5-dimethyl-1H-pyrazol-3-amine1-methyl-5-(trifluoromethyl)-1H-pyrazol-3-amine1-methyl-5-(prop-1-en-2-yl)-1H-pyrazol-3-amineEthyl 1-benzyl-3-nitro-1H-pyrazole-5-carboxylateEthyl 1-benzyl-3-nitro-1H-pyrazole-5-carboxylate3-(2,5-dimethyl-1H-pyrrol-1-yl)-1-phenyl-1H-pyrazole8-bromo-1,2,3,5,6,7-hexahydro-s-indacen-4-amine8-chloro-1,2,3,5,6,7-hexahydro-s-indacen-4-amine8-methyl-1,2,3,5,6,7-hexahydro-s-indacen-4-amine3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-amine4-bromo-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-amine3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-4-aminebenzo[1,2-b:4,5-b′]difuran-4-amine3-(3-(trifluoromethyl)-3H-diazirin-3-yl)aniline Synthesis of R2Acid Intermediates R2acid intermediates are commercially available and/or may be prepared by routine synthetic methods. WO 2016/131098 for example (see pages 166-169) discloses the synthesis of the following R2acid intermediates which may be used in the synthesis of compounds of the present invention:2,3,6,7-tetrahydrobenzo[1,2-b:4,5-b′]difuran-4-carboxylic acidBenzo[d][1,3]dioxole-4-carboxylic acid Compounds Example 1: N-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl) morpholine-4-sulfonamide 4-Isocyanato-1,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and morpholine-4-sulfonamide were used in general method C2 to give the titled compound as a white solid (25 mg, 24%).1H NMR (400 MHz, DMSO-d6): δ=7.98 (bs, 1H), 6.94 (s, 1H), 3.63 (t, J=4.0 Hz, 4H), 3.18 (t, J=4.0 Hz, 4H), 2.81 (t, J=8.0 Hz, 4H), 2.68 (t, J=8.0 Hz, 4H), 2.02-1.95 (m, 4H); LCMS Purity: >95%; LCMS (m/z): 366 [M+H]+. Example 2: N-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-methylpiperazine-1-sulfonamide 4-Isocyanato-1,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4-Methylpiperazine-1-sulfonamide were used in general method C3 to give the titled compound as a white solid (60 mg, 55%).1H NMR (600 MHz, DMSO-d6): δ=7.96 (bs, 1H), 6.94 (s, 1H), 3.20 (t, J=6.0 Hz, 4H), 2.80 (t, J=6.0 Hz, 4H), 2.69 (t, J=6.0 Hz, 4H), 2.37 (t, J=6.0 Hz, 4H), 2.19 (s, 3H), 2.00-1.95 (m, 4H).13C NMR (150 MHz, DMSO-d6): δ=150.6, 143.5, 137.4, 129.6, 118.1, 54.3, 46.5, 45.9, 32.9, 30.7, 25.5. LCMS Purity: >95%; LCMS (m/z): 379 [M+H]+. HRMS calculated for C18H27N4O3S1(M+H)+379.1798, found 379.1795. Example 3: N-[1,2,3,5,6,7-hexahydro-s-indacen-4-yl]-N′-[(dimethylamino) sulfonyl]urea 4-Isocyanato-1,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and N,N-dimethylsulfamide were used in general method C2 to give the titled compound as a white solid (29 mg, 31%).1H NMR (400 MHz, DMSO-d6): δ=7.96 (s, 1H), 6.94 (s, 1H), 2.81 (t, J=8 Hz, 4H), 2.79 (s, 6H), 2.70 (t, J=8 Hz, 4H), 2.02-1.96 (m, 4H).13C NMR (150 MHz, DMSO-d6): δ=143.4, 142.9, 137.4, 125.1 117.9, 38.6, 32.9, 30.7, 25.5; LCMS (m/z): 324 [M+H]+; HRMS calculated for C15H21N3O3S1(M+H)+, 324.13764, found 324.13891. Example 4: N-((9H-carbazol-9-yl)carbamoyl)-1-isopropyl-1H-pyrazole-3-sulfonamide 9H-carbazol-9-amine (1.0 g, 5.5 mmol) in THF (20 mL) was cooled to 0° C. and sodium hydride (0.45 g, 11 mmol) was added portion-wise. The reaction mixture was stirred for 30 mins then phenylchloroformate (1.72 g, 11 mmol) added drop-wise. The solution was allowed to warm to ambient temperature and stirred for a further 5 h. The reaction was quenched using NaHCO3(aq) and the solution extracted using ethyl acetate. The organic phase was washed using water, brine then dried (Na2SO4) and concentrated in vacuo. The crude phenyl (9H-carbazol-9-yl)carbamate was purified by column chromatography on silica using 10% EtOAc:hexanes eluent and the resulting white solid used directly in the next synthetic step. 1-Isopropyl-1H-pyrazole-3-sulfonamide (0.1 g, 0.53 mmol) in THF (10 mL) was treated with NaH (60 mg, 1.06 mmol) and the reaction heated to 80° C. for 2 h. The mixture was cooled to ambient temperature, phenyl (9H-carbazol-9-yl)carbamate (2 equivalents) added and the reaction heated once more to 80° C. for 2 h. On completion the reaction was diluted using sat. aq. NH4Cl and extracted using ethyl acetate (2×25 mL). The combined organics were washed with water, brine, dried (Na2SO4) and concentrated in vacuo. The product was purified using preparative thin layer chromatography on silica with 50% EtOAc:hexane to give the titled product as a white solid (15 mg, 7%).1H NMR (400 MHz, CD3OD) δ 8.02 (d, J=7.7 Hz, 2H), 7.72 (s, 1H), 7.41-7.27 (m, 4H), 7.19 (t, J=7.4 Hz, 2H), 6.70 (s, 1H), 4.59 (m, 1H), 1.48 (d, J=6.6 Hz, 6H). Copper Complexation of MCC950 Complex formation between the sodium form of MCC950 and copper chloride was tested and detected by using isothermal titration calorimetry (ITC). MCC950 (also known as CRID3) is a sulphonylurea having the formula: An autoITC 200 (GE life sciences) was used to measure the change in heat induced by Cu2+-MCC950 interactions and data analysed using the MicroCal Origin version 7.0 software package adapted for auto-ITC data analysis. Copper chloride (5 or 2.5 mM of CuCl2) dissolved in water (miliQ water; Elga) was titrated into a cell containing 0.4 mM MCC950Na also dissolved in water. The titration consisted of 19×2 μL injections at 25° C. As a control EDTA was used, in free acid form or as a sodium di-salt, at a concentration of 0.4 mM. Experiments were replicated three times and results were averaged. Thermograms and binding isotherms were used to determine the enthalpy (ΔH), binding constants (K), stoichiometry (N), and entropy (TΔS) using a single-site binding model. The change in standard Gibbs free energy (ΔG) was calculated using the Gibbs-Helmholz thermodynamic equation: ΔG=−RTInK, where R is the ideal gas constant (1.985 cal mol−1K−1) and T is the temperature (298 K). Copper (II) ions were seen to form a strong complex with MCC950, comparable to the complex with EDTAx2Na and much stronger than with EDTA free acid. Formation of MCC950:Cu(II) complex was endothermic with enthalpy being positive thereby suggesting that the process was entropy driven with the presence of strong hydrophobic interactions. The thermodynamics are displayed in the table below and results are also shown inFIG.1: KΔHTΔSΔGComplexN(104M−1)(kcal mol−1)(kcal mol−1)(kcal mol−1)MCC950 × Na:CuCl20.31 ± 0.005.69 ± 0.36.56 ± 0.1513.04 ± 0.12−6.48 ± 0.03 Biological Testing Methodology NLRP3 inhibition assays The following assays can be used to determine inhibitory activity of test compounds on the NLRP3 inflammasome using common stimuli such as adenosine triphosphate, nigericin, LeuLeu-OMe or monosodium urate crystals (MSU). Cell Culture To generate HMDM (Human Monocyte Derived Macrophages), human monocytes are isolated from buffy coat blood using Ficoll-Plaque Plus (GE Healthcare) and density centrifugation. CD14+cell selection is performed using MACS magnetic beads (Miltenyl Biotec). Isolated CD14+monocytes are differentiated in culture for 7 days with 10 ng/ml human CSF-1 (Miltenyl Biotec) in Iscove's modified Dulbecco's medium (IMDM) containing L-glutamine supplemented with 10% FBS and 1% penicillin/streptomycin (Life Technologies) as described by Croker et al 2013Immunol Cell Biol91:625. Stably transfected ASC-cerulean macrophages as described by Hett et al. (Nat. Chem. Biol., 9, 398-405, 2013) are cultured in DMEM supplemented with 10% FCS and 1% P/S. NLRP3 Inflammasome Activation Assays HMDM are seeded at 1×105/ml. The following day the overnight medium is replaced and cells are stimulated withEscherichia coliserotype 0111:B4 (Sigma Aldrich) for 3 h. Medium is removed and replaced with serum free medium (SFM) containing test compound 30 min prior to NLRP3 stimulation. Cells are then stimulated with: adenosine 5′-triphosphate disodium salt hydrate (5 mM 1 h), nigericin (10 μM 1 h), LeuLeu-OMe (1 mM 2 h) or MSU (200 μg/ml 15 h). ATP can be sourced from Sigma Aldrich, nigericin and MSU from Invivogen and LeuLeu-Ome from Chem-Impex International. Measurement of IL-1β, IL-18, TNFα and Cell Death For ELISA and cell death assays cells are seeded in 96 well plates. Supernatants are removed and analysed using ELISA kits according to the manufacturer's instructions (DuoSet® R&D Systems, ReadySetGo!® eBioscience, BD OptEIA™, or Perkin Elmer AlphaLISA®). Cell death is assessed by measurement of LDH release relative to a 100% cell lysis control using the CytoTox96® non-radioactive cytotoxicity assay (Promega). Murine Studies on Compound Levels in Blood Plasma and Brain General Experimental: Carbutamide was purchased from Sigma Aldrich (Catalogue No. 381578). Acetonitrile was Chromasolv® HPLC grade (Sigma Aldrich, Sydney, Australia), the formic acid was AR grade 99%-100% Normapur (VWR International Pty Ltd, Brisbane, Australia), DMSO was ReagentPlus® grade (D5879, Sigma Aldrich, Sydney, Australia) and the H2O Milli-Q was filtered. The HPLC vial and polypropylene inserts from Agilent Technologies (Melbourne, Australia), while the 1.5 mL Eppendorf tubes Protein LoBind Tubes were from VWR International Pty Ltd (Brisbane, Australia). Preparation of Precipitation Solution: 100 mL ACN and 5 μL of 10 mM carbutamide in DMSO (ACN with 135 ng/mL carbutamide MS internal standard). Preparation of Standard Curve in Plasma: A 1 mg/mL of test compound in 10 mM NH4HCO3was prepared and diluted 10-fold to give a 100,000 ng/mL stock solution. A series of 10-fold dilutions of the 100,000 ng/mL stock solution with 10 mM NH4HCO3gave concentrations of 10,000, 1,000, 100 and 10 ng/mL. The 100,000 ng/mL stock solution was diluted to 3:7 with 10 mM NH4HCO3to give a concentration of 30,000 ng/mL and a series of 10-fold dilutions gave concentrations of 3,000, 300, 30 and 3 ng/mL. 20 μL of test compound-containing solution and 160 μL precipitation solution were added to 20 μL of mouse plasma in a low binding Eppendorf tube. The samples were vortexed, allowed to stand at 4° C. for 10 mins and centrifuged at 14,000×g for 8 min. 150 μL of the supernatant was transferred to an HPLC vial insert. The samples were stored at 4° C. until analysis. Preparation of Standard Curve in Brain Homogenate: The sample solutions prepared for the plasma standard curve were used for the brain homogenate standard curve. The mouse brain homogenate from the saline control was thawed and vortexed for 3 min or until homogenous, sonicated for 1 min. When the foam settled, 50 μL of mouse brain homogenate was transferred into an Eppendorf tube, followed by 50 μL of test compound in 10 mM NH4HCO3, 150 μL of H2O and 500 μL of ice cold precipitation solution with vortexing after every addition. The standards were allowed to stand at 4° C. for 10 mins and then centrifuged at 14,000×g for 8 min. 200 μL of the supernatant was transferred to HPLC vial insert ensuring that no air bubbles were present and the samples stored at 4° C. until analysis. Dosing of Mice and Transcardial Perfusion Dosing: Oral gavage at 20 mg/kg Time Point: 2 hour Prepare stock compounds for dosing at 4 mg/ml in sterile PBS. Mice were weighed and dosed by oral gavage at 20 mg/kg for each compound. After 2 hours mice were anesthetized using a combination of Zoletil (50 mg/kg) and Xylazine (10 mg/kg) and blood was collected by cardiac puncture into tubes containing 20 μL of 100 mM EDTA. The blood was centrifuged at 2000×g for 15 minutes at 4° C. to collect plasma. Preparation of Plasma Samples for Analysis: 20 μL of NH4HCO3and 160 μL precipitation solution were added to 20 μL of mouse plasma in a low binding Eppendorf tube. The samples were vortexed, allowed to stand at 4° C. for 10 mins and centrifuged at 14,000×g for 8 min. 150 μL of the supernatant was transferred to an HPLC vial insert ensuring that no air bubbles were present. The samples were stored at 4° C. until analysis. Brain Homogenate Preparation: The brains of mice were perfused with PBS for 5 minutes then dissected and weighed. Brain homogenate was prepared by homogenizing total brain (0.5 g) with 4 volumes (2 ml) of deionized water and stored at −20° C. before analysis. The homogenate was thawed, vortexed for 3 min or until homogenous, and sonicated for 1 min. When the foam settled, 50 μL of mouse brain homogenate was transferred into an Eppendorf tube, followed by 50 μL of 10 mM NH4HCO3, 150 μL of H2O and 500 μL of ice cold precipitation solution with vortexing after every addition. 200 μL of the supernatant was transferred to HPLC vial insert ensuring that no air bubbles were present and the samples stored at 4° C. until analysis. Preparation of Brain Samples for Analysis: 50 μL of mouse brain was transferred into an Eppendorf tube, followed by 50 μL of 10 mM NH4HCO3, 150 μL of H2O and 500 μL of ice cold precipitation solution with vortexing after every addition. The solutions were allowed to stand at 4° C. for 10 mins and then centrifuged at 14,000×g for 8 min. 200 μL of the supernatant was transferred to HPLC vial insert ensuring that no air bubbles were present and the samples stored at 4° C. until analysis. LC-MS/MS: The samples were analysed on an AB Sciex 4000QTrap MS with 2 Shimadzu Nexera LC-30AD Solvent Delivery Units, Shimadzu Nexera SIL-30AC Auto-Sampler, Shimadzu Prominence DGU-20A5Degasser, Shimadzu Prominence CBM-20A System Controller and Shimadzu Prominence CTO-20A Column Oven. The column oven was set to 40° C., while the Autosampler was set to 15° C. 2 μL injections were made and MS analyses were undertaken in Selected Reaction Monitoring (SRM) mode using Turbo Spray (−)-ESI with Low Resolution Q1 and Low Resolution Q3. MS parameters: CUR: 30.00, IS: −4300.00, TEM: 500.00, GS1: 50.00, GS2: 50.00, ihe: ON, CAD: High, DP −60.00, EP −10.00, CXP −15.00. MCC950 SRM: Q1 403.2 to Q3 204.3 Da, dwell 150 msec, CE −27 and carbutamide (IS) SRM: Q1 270.0 to Q3 171.0 Da, dwell 100 msec, CE −25. HPLC Column: Waters Atlantis® T3 5 μm 2.1×50 mm with Atlantis® T3 5 μm 2.1×10 mm guard column. Flow rates and solvent: 0.35 ml/min, solvent A: 0.1% formic acid in H2O, solvent B: 0.1% formic acid in ACN; isocratic 2% B from 0→2 mins, gradient 2%→100% B from 2→5 mins, isocratic 100% from 5→9 mins, gradient 100%→2% B from 9→9.1 mins and isocratic 2% B from 9.1→13 mins. The peak areas from the SRM data for carbutamide and test compound were analysed using the AB Sciex's Analyst software using the Quantitation Wizard. The peak area was plotted against the ng/mL concentration in 20 μL 3 to 30,000 ng/mL test compound solutions and the lower and upper range of linear response was determined. These data were then plotted in Microsoft Excel and the linear response equation used to determine the test compound concentration in the 20 μL plasma solutions. Similarly, for the brain homogenate samples, the peak areas of the 50 μL 3 to 3,000 ng/mL test compound solutions were used to determine the test compound concentration in the 50 μL brain homogenate solutions. Results TABLE 1Topological Polar Surface Area (tPSA) and molecular weight of selectcompounds.ExCompound structureCompound nametPSAM.W.1N-((1,2,3,5,6,7-hexahydro-s- indacen-4-yl)carbamoyl) morpholine-4-sulfonamide883652N-((1,2,3,5,6,7-hexahydro-s- indacen-4-yl)carbamoyl)-4- methylpiperazine-1- sulfonamide903783N-[1,2,3,5,6,7-hexahydro-s- indacen-4-yl]-N′- [(dimethylamino)sulfonyl] urea793234N-((9H-carbazol-9- yl)carbamoyl)-1-isopropyl- 1H-pyrazole-3-sulfonamide106397 TABLE 2Inhibition of IL-1β release IC50 in nM cell based assay usingAvg.IL-1βIC50HRMSHRMSHMDMExNameChem FormulaHRMS formulaCalcfound(nM)2N-((1,2,3,5,6,7-C18H26N4O3SC18H27N4O3S1379.1798379.1795+++hexahydro-s-indacen-4-yl)carbamoyl)-4-methylpiperazine-1-sulfonamide4N-((9H-carbazol-9-yl)C19H19N5O3SC19H20N5O3S398.1281398.1282++carbamoyl)-1-isopropyl-1H-pyrazole-3-sulfonamideHMDM (<1 μM = ‘+++’/<10 μM = ‘++’/<50 μM = ‘+’). (ESI+ for all compounds) Examples 5-43 Nuclear magnetic resonance (NMR) spectra were recorded at 400 MHZ; the chemical shifts are reported in parts per million. Spectra were recorded using a Bruker Avance III spectrometer at 400 MHz fitted with a BBO 5 mm liquid probe. Mass spectra were recorded with a Waters Acquity UPLC system equipped with Acquity UPLC BEH. Mobile phases typically consisted of acetonitrile mixed with water containing 10 mM ammonium bicarbonate. Preparative HPLC was carried out using a Waters Xbridge BEH C18, 5 μm, 19×50 mm column using a gradient MeCN in aqueous 10 mM ammonium bicarbonate. Fractions were collected following detection by mass with positive and negative ion electrospray detector on a Waters FractionLynx LCMS. ((1,2,3,5,6,7-Hexahydro-s-indacen-4-yl)carbamoyl)sulfamoyl chloride A stirred solution of chlorosulfonyl isocyanate (2.63 ml, 30.3 mmol) in diethyl ether (20 mL) was cooled to −20° C., then a solution of 1,2,3,5,6,7-hexahydro-s-indacen-4-amine (5 g, 28.9 mmol) in diethyl ether (100 mL) was added slowly over 10 minutes. The reaction was stirred for 1 hour, then most of the ether removed in vacuo. Iso-hexane (200 mL) was added and the mixture was sonicated for 5 minutes. The solid was filtered and dried overnight to afford the title compound (7.5 g). 1H NMR (400 MHz, CDCl3) δ 7.95 (s, 1H), 7.10 (s, 1H), 2.93 (t, J=7.5 Hz, 4H), 2.86 (t, J=7.4 Hz, 4H), 2.11 (p, J=7.4 Hz, 4H) (exchangeable proton not visible). General Procedure Amines (0.1 mmol) were pre-dissolved in DMA (0.5 mL), then 4-methylmorpholine (0.040 g, 0.400 mmol) was added. A solution of ((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)sulfamoyl chloride (0.025 g, 0.08 mmol) in THF (1 mL) was added to each well, the reactions were capped and shaken overnight at room temperature. The samples were purified by RPHPLC; Waters X-Bridge BEH C18 prep column, 5 μm, 10×50 mm, Basic (0.1% ammonium bicarbonate) 6.5 min method, 10-40% acetonitrile. Examples 5-40 shown in Table 3 below were prepared this way. TABLE 3LCMSExactMassRetentionExStructuremassiontime5449.1450.11.36437.1438.11.37463.1464.11.038424.2425.21.249433.1434.11.3610469.1470.11.3511439.1440.11.2112448.3449.31.0913474.2475.21.2914375.1376.11.0915406.1407.11.0116406.2407.21.0517488.2489.21.2418462.2463.21.2719482.1483.21.2920482.1483.21.4721468.2469.20.9622513.2514.21.1323391.1392.11.0924429.2430.21.525413.2414.21.3826429.2430.21.527451.2452.21.0928397.1398.21.2129400.2401.20.9130386.1387.10.9331385.1386.21.2132417.2418.21.2233410.1411.11.1334497.2498.21.135416.2417.21.136457.1458.11.1137411.2412.21.2738467.2468.21.0939400.2401.21.0240440.2441.21.38 Example 41: N-((1,2,3,5,6,7-Hexahydro-s-indacen-4-yl)carbamoyl)-4-methyl-piperazine-1-sulfonamide 1-Methylpiperazine (70 μl, 0.631 mmol) was added to a suspension of ((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)sulfamoyl chloride (50 mg, 0.159 mmol) in THF (1 mL). The reaction mixture was stirred at room temperature overnight. DMF (1 mL) was added to aid solubility, the mixture was stirred for a further 1 hour, then filtered through a plug of cotton wool and purified by preparative HPLC to afford the title compound (2.8 mg) as a colourless solid. 1H NMR (400 MHz, D2O/NaOD) δ 6.82 (s, 1H), 2.89 (br s, 4H), 2.59 (t, J=7.5 Hz, 4H), 2.48 (t, J=7.3 Hz, 4H), 2.26 (br s, 4H), 1.97 (s, 3H), 1.76 (p, J=7.5 Hz, 4H). LCMS m/z 379 (M+H)+(ES+); 377 (M−H)−(ES−) Example 42: 1-(1,2,3,5,6,7-Hexahydro-s-indacen-4-yl)-3-[(1-methyl-1H-indazol-6-yl)sulfamoyl]urea ((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)sulfamoyl chloride (75 mg, 0.238 mmol) was added to a solution of 1-methyl-1H-indazol-6-amine (105 mg, 0.715 mmol) in THF (2 mL). The mixture was stirred for 6 hours, filtered, then purified by preparative HPLC to afford the title compound (2.3 mg). 1H NMR (400 MHz, D2O/NaOD) δ 7.53 (s, 1H), 7.19 (d, J=8.8 Hz, 1H), 6.77 (s, 1H), 6.60 (s, 1H), 6.46 (dd, J=8.7, 1.8 Hz, 1H), 2.37 (t, J=7.5 Hz, 4H), 2.36 (s, 3H), 1.97 (t, J=7.5 Hz, 4H), 1.41 (p, J=7.5 Hz, 4H). LCMS m/z 426 (M+H)+(ES+); 424 (M−H)−(ES−) Example 43: 3-(1,2,3,5,6,7-Hexahydro-s-indacen-4-yl)-1-[(1H-indol-5-yl)sulfamoyl]urea 1H-Indol-5-amine (84 mg, 0.635 mmol) and triethylamine (150 μl, 1.076 mmol) were dissolved in THF (2 mL). ((1,2,3,5,6,7-Hexahydro-s-indacen-4-yl)carbamoyl) sulfamoyl chloride (100 mg, 0.318 mmol) was then added as a solid, the mixture was stirred for 30 minutes, filtered and purified by preparative HPLC to afford the title compound (7 mg) as a pale brown solid. 1H NMR (400 MHz, DMSO-d6) δ 11.11 (s, 1H), 9.85 (s, 1H), 9.70 (s, 1H), 7.64 (s, 1H), 7.41 (d, J=2.0 Hz, 1H), 7.37 (t, J=2.8 Hz, 1H), 7.34 (d, J=8.6 Hz, 1H), 7.00 (dd, J=8.6, 2.0 Hz, 1H), 6.94 (s, 1H), 6.39-6.36 (m, 1H), 2.81 (t, J=7.4 Hz, 4H), 2.58 (t, J=7.4 Hz, 4H), 1.95 (p, J=7.5 Hz, 4H). LCMS m/z 411 (M+H)+(ES+); 409 (M−H)−(ES−) Biological Assay NLRP3 and Pyroptosis It is well established that the activation of NLRP3 leads to cell pyroptosis and this feature plays an important part in the manifestation of the clinical disease (Yan-gang Liu et al., Cell Death & Disease, 2017, 8(2), e2579; Alexander Wree et al., Hepatology, 2014, 59(3), 898-910; Alex Baldwin et al., Journal of Medicinal Chemistry, 2016, 59(5), 1691-1710; Ema Ozaki et al., Journal of Inflammation Research, 2015, 8, 15-27; Zhen Xie & Gang Zhao, Neuroimmunology Neuroinflammation, 2014, 1(2), 60-65; Mattia Cocco et al., Journal of Medicinal Chemistry, 2014, 57(24), 10366-10382; T. Satoh et al., Cell Death & Disease, 2013, 4, e644). Therefore, it is anticipated that inhibitors of NLRP3 will block pyroptosis, as well as the release of pro-inflammatory cytokines (e.g. IL-1β) from the cell. THP-1 Cells: Culture and Preparation THP-1 cells (ATCC #TIB-202) were grown in RPMI containing L-glutamine (Gibco #11835) supplemented with 1 mM sodium pyruvate (Sigma #S8636) and penicillin (100 units/ml)/streptomycin (0.1 mg/ml) (Sigma #P4333) in 10% Fetal Bovine Serum (FBS) (Sigma #F0804). The cells were routinely passaged and grown to confluency (˜106cells/ml). On the day of the experiment, THP-1 cells were harvested and resuspended into RPMI medium (without FBS). The cells were then counted and viability (>90%) checked by Trypan blue (Sigma #T8154). Appropriate dilutions were made to give a concentration of 625,000 cells/ml. To this diluted cell solution was added LPS (Sigma #L4524) to give a 1 μg/ml Final Assay Concentration (FAC). 40 μl of the final preparation was aliquoted into each well of a 96-well plate. The plate thus prepared was used for compound screening. THP-1 Cells Pyroptosis Assay The following method step-by-step assay was followed for compound screening.1. Seed THP-1 cells (25,000 cells/well) containing 1.0 μg/ml LPS in 40 μl of RPMI medium (without FBS) in 96-well, black walled, clear bottom cell culture plates coated with poly-D-lysine (VWR #734-0317)2. Add 5 μl compound (8 points half-log dilution, with 10 μM top dose) or vehicle (DMSO 0.1% FAC) to the appropriate wells3. Incubate for 3 hrs at 37° C. in 5% CO24. Add 5 μl nigericin (Sigma #N7143) (FAC 5 μM) to all wells5. Incubate for 1 hr at 37° C. and 5% CO26. At the end of the incubation period, spin plates at 300×g for 3 mins and remove supernatant7. Then add 50 μl of resazurin (Sigma #R7017) (FAC 100 μM resazurin in RPMI medium without FBS) and incubate plates for a further 1-1.5 h at 37° C. and 5% CO28. Plates were read in an Envision reader at Ex 560 nm and Em 590 nm9. IC50data is fitted to a non-linear regression equation (log inhibitor vs response-variable slope 4-parameters) 96-Well Plate Map 123456789101112AHighComp 1Comp 2Comp 3Comp 4Comp 5Comp 6Comp 7Comp 8Comp 9Comp 10LowBHighComp 1Comp 2Comp 3Comp 4Comp 5Comp 6Comp 7Camp SComp 9Comp 10LowCHighComp 1Comp 2Comp 3Comp 4Comp 5Comp 6Comp 7Comp 8Comp 9Comp 10LowDHighComp 1Comp 2Comp 3Comp 4Comp 5Comp 6Comp 7Comp 8Comp 9Comp 10LowEHighComp 1Comp 2Comp 3Comp 4Comp 5Comp 6Comp 7Comp 8Comp 9Comp 10LowFHighComp 1Comp 2Comp 3Comp 4Comp 5Comp 6Comp 7Comp 8Comp 9Comp 10LowGHighComp 1Comp 2Comp 3Comp 4Comp 5Comp 6Comp 7Comp 8Comp 9Comp 10LowHHighComp 1Comp 2Comp 3Comp 4Comp 5Comp 6Comp 7Comp 8Comp 9Comp 10LowHighMCC950 (10 uM)Compound 8-point half-log dilutionLowDrug free contro1 The results of the pyroptosis assay performed are summarised in Table 4 below. TABLE 4Results of pyroptosis assayExampleIC50ExampleIC50No.(nM)No.(nM)5++6++7++8not measured9not measured10not measured11++12++13++14++15++16not measured17+18++19+++20++21++22not measured23++24++25++26++27++28++29++30+31++32++33++34++35+++36++37++38not measured39++40++41not measured42+++43+++(<1 μM = ‘+++’/<10 μM = ‘++’/<50 μM = ‘+’)
127,887
11858923
DETAILED DESCRIPTION This disclosure provides secondary amine-substituted coumarin compounds particularly suitable for methods of fluorescence detection and sequencing by synthesis. Embodiments described herein relate to dyes and their derivatives of the structure of Formula (I). According to a first aspect the disclosure provides compounds of Formula (I) or salts thereof. Particular limitations for the various substituents are shown below. Each single group can be combined with any other individual limitation unless otherwise specified. To improve fluorescent properties of the biomarkers and especially their bioconjugates in water-based solutions, the compound of Formula (I) is a compound in which:i) R2is —SO3H; and/orii) R4is —SO3H; and/oriii) R5is —SO3H or —SO2NH2. In some aspects, X is O or S. In some aspects, X is O. In some aspects, X is S. In some aspects, X is NRn, where Rnis H or C1-6alkyl, and in some aspects, Rnis H. In some aspects, R3is H. In some aspects, R3is methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, or hexyl. In other aspects, R3is ethyl. In other aspects, R3is substituted C2-6alkyl. In other aspects, R3is C2-6alkyl substituted with —CO2H. In other aspects, R3is optionally substituted C2-6alkenyl or optionally substituted C2-6alkynyl. In some aspects, R3is linked to R2or R4to form an optionally substituted ring. Where coupling to a linker or nucleotide is via R3, R3should be of sufficient length to allow coupling to a functional group attached thereto. In some aspects, R3is not —CH2COOH or —CH2COO−. Optionally, R3is —(CH2)nCOOH where n is 2-6. In some aspects, n is 2, 3, 4, 5 or 6. In other aspects, n is 2 or 5. In some aspects, n is 2. In some aspects, n is 5. Optionally, R3is —(CH2)nSO3H where n is 2-6. In some aspects, n is 2, 3, 4, 5 or 6. In other aspects, n is 2 or 5. In some aspects, n is 2. In some aspects, n is 5. The benzene ring of the indole moiety is optionally substituted in any one, two, three, or four positions by a substituent shown as R5. Where m is zero, the benzene ring is unsubstituted. Where m is greater than 1, each R5may be the same or different. In some aspects, m is 0. In other aspects, m is 1. In other aspects, m is 2. In some aspects, m is 1, 2, or 3, and each R5is independently halo, —CN, —CO2H, amino, —OH, —SO3H, or —SO2NH2. In some aspects, R5is —(CH2)xCOOH where x is 2-6. In some aspects, x is 2, 3, 4, 5 or 6. In other aspects, x is 2 or 5. In some aspects, x is 2. In some aspects, x is 5. In some aspects, R5is halo, —CN, —CO2H, —SO3H, —SO2NH2, or optionally substituted C1-6alkyl. In some aspects, R5is halo, —CO2H, —SO3H, or —SO2NH2. In some aspects, R5is C2-6alkyl substituted with —CO2H, —SO3H, or —SO2NH2. In some aspects, each R5is independently optionally substituted C1-6alkyl, halo, —CN, —CO2H, amino, —OH, —SO3H, or —SO2NH2. In some aspects, R1is H. In some aspects, R1is halo. In some aspects, R1is Cl. In some aspects, R1is C1-6alkyl. In some aspects, R1is methyl. In some aspects, R is H. In some aspects, R is halo. In some aspects, R is Cl. In some aspects, R is C1-6alkyl. In some aspects, R is methyl. In some aspects, R is not —CN. In some aspects, R is H, halo, —CO2H, amino, —OH, C-amido, N-amido, —NO2, —SO3H, —SO2NH2, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted aminoalkyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In some aspects, R2is H. In some aspects, R2is optionally substituted alkyl. In some aspects, R2is C1-4alkyl optionally substituted with —CO2H or —SO3H. In some aspects, R2is —SO3H. In some aspects, R2is linked to R3to form an optionally substituted heterocyclic ring, such as a pyrrolidine or piperidine, optionally substituted with one or more alkyl groups. In some aspects, R2is H, optionally substituted alkyl, C1-4alkyl optionally substituted with —CO2H or —SO3H, or —SO3H. In some aspects, R2is H or —SO3H. In some aspects, R4is H. In some aspects, R4is optionally substituted alkyl. In some aspects, R4is C1-4alkyl optionally substituted with —CO2H or —SO3H. In some aspects, R4is —SO3H. In some aspects, R4is linked to R3to form an optionally substituted heterocyclic ring, such as a pyrrolidine or piperidine, optionally substituted with one or more alkyl groups. Particular examples of a compound of Formula (I) include where X is O or S; R is H; R1is H; R3is —(CH2)nCOOH where n is 2-6; R5is H, —SO3H, or —SO2NH2; R2is H or —SO3H; and R4is H or —SO3H. Particular examples of a compound of Formula (I) include where X is O or S; R is H; R1is H; R3is —(CH2)2COOH; R5is H, —SO3H, or —SO2NH2; R2is H or —SO3H; and R4is H or —SO3H. Particular examples of a compound of Formula (I) include where X is O or S; R is H; R1is H; R3is —(CH2)5COOH; R5is H, —SO3H, or —SO2NH2; R2is H or —SO3H; and R4is H or —SO3H. In some aspects of Formula (II), X′ is O. In some aspects, X′ is S. In some aspects, X′ is NRp, where Rpis H or C1-6alkyl. In some aspects, X′ is NRp, where Rpis H. In some aspects, R6is H. In some aspects, R6is C1-4alkyl. In some aspects, R7is H. In some aspects, R7is optionally substituted C1-4alkyl, —CO2H, —SO3H, —SO2NH2, —SO2NH(C1-4alkyl), or —SO2N(C1-4alkyl)2. In some aspects, R7is C1-4alkyl optionally substituted with —CO2H. In some aspects, R8is H. In some aspects, R8is —CO2H, —SO3H, or —SO2NH2. In some aspects, R8is —SO3H. In some aspects, R10is H. In some aspects, R10is —CO2H, —SO3H, or —SO2NH2. In some aspects, R10is —SO3H. In some aspects, R8is H and R10is —SO3H. In some aspects, R8is —SO3H and R10is H. In some aspects, one of R8and R10is H, halo, —CN, —CO2H, amino, —OH, —SO3H, —SO2NH2, —SO2NH(C1-4alkyl), —SO2N(C1-4alkyl)2, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, optionally substituted C2-6alkynyl, or optionally substituted C1-6alkoxy, and the other of R8and R10is taken with R9to form an optionally substituted 4- to 7-membered heterocyclic ring. In some aspects, R9is C2-6alkyl. In some aspects, R9is C1-6alkyl substituted with —CO2H, —CO2C1-4alkyl, —CONH2, —CONH(C1-4alkyl), —CON(C1-4alkyl)2, —CN, —SO3H, —SO2NH2, —SO2NH(C1-4alkyl), or —SO2N(C1-4alkyl)2. In some aspects, R9is C1-6alkyl substituted with —CO2H. In some aspects, R9is —(CH2)y—CO2H, where y is 2, 3, 4, or 5. In some aspects, each R11is independently halo, —CO2H, —SO3H, —SO2NH2, —SO2NH(C1-4alkyl), —SO2N(C1-4alkyl)2, or optionally substituted alkyl. In other aspects, each R11is independently halo, —CO2H, —SO3H, or —SO2NH2. In some aspects, q is 0. In other aspects, q is 1. In still other aspects, q is 2. Specific examples of secondary amine-substituted coumarin dyes include: and salts thereof. A particularly useful compound is a nucleotide or oligonucleotide labeled with a dye as described herein. The labeled nucleotide or oligonucleotide may have the label attached to the nitrogen atom of coumarin molecule via an alkyl-carboxy group to form an alkyl-amide. The labeled nucleotide or oligonucleotide may have the label attached to the C5 position of a pyrimidine base or the C7 position of a 7-deaza purine base through a linker moiety. The labeled nucleotide or oligonucleotide may also have a blocking group covalently attached to the ribose or deoxyribose sugar of the nucleotide. The blocking group may be attached at any position on the ribose or deoxyribose sugar. In particular embodiments, the blocking group is at the 3′ OH position of the ribose or deoxyribose sugar of the nucleotide. Provided herein are kits including two or more nucleotides wherein at least one nucleotide is a nucleotide labeled with a compound of the present disclosure. The kit may include two or more labeled nucleotides. The nucleotides may be labeled with two or more fluorescent labels. Two or more of the labels may be excited using a single excitation source, which may be a laser. For example, the excitation bands for the two or more labels may be at least partially overlapping such that excitation in the overlap region of the spectrum causes both labels to emit fluorescence. In particular embodiments, the emission from the two or more labels will occur in different regions of the spectrum such that presence of at least one of the labels can be determined by optically distinguishing the emission. The kit may contain four labeled nucleotides, where the first of four nucleotides is labeled with a compound as disclosed herein. In such a kit, each of the four nucleotides can be labeled with a compound that is the same or different from the label on the other three nucleotides. Thus, one or more of the compounds can have a distinct absorbance maximum and/or emission maximum such that the compound(s) is(are) distinguishable from other compounds. For example, each compound can have a distinct absorbance maximum and/or emission maximum such that each of the compounds is distinguishable from the other three compounds. It will be understood that parts of the absorbance spectrum and/or emission spectrum other than the maxima can differ and these differences can be exploited to distinguish the compounds. The kit may be such that two or more of the compounds have a distinct absorbance maximum. The compounds of the invention typically absorb light in the region below 500 nm. The compounds, nucleotides, or kits that are set forth herein may be used to detect, measure, or identify a biological system (including, for example, processes or components thereof). Exemplary techniques that can employ the compounds, nucleotides or kits include sequencing, expression analysis, hybridization analysis, genetic analysis, RNA analysis, cellular assay (e.g., cell binding or cell function analysis), or protein assay (e.g., protein binding assay or protein activity assay). The use may be on an automated instrument for carrying out a particular technique, such as an automated sequencing instrument. The sequencing instrument may contain two lasers operating at different wavelengths. Disclosed herein are methods of synthesizing compounds of the disclosure. Dyes according to the present disclosure may be synthesized from a variety of different suitable starting materials. Methods for preparing coumarin dyes are well known in the art. Definitions The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. It is noted that, as used in this specification and the appended claims, the singular forms “a”, “an” and “the” include plural referents unless expressly and unequivocally limited to one referent. It will be apparent to those skilled in the art that various modifications and variations can be made to various embodiments described herein without departing from the spirit or scope of the present teachings. Thus, it is intended that the various embodiments described herein cover other modifications and variations within the scope of the appended claims and their equivalents. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. The use of the term “including” as well as other forms, such as “include”, “includes,” and “included,” is not limiting. The use of the term “having” as well as other forms, such as “have”, “has,” and “had,” is not limiting. As used in this specification, whether in a transitional phrase or in the body of the claim, the terms “comprise(s)” and “comprising” are to be interpreted as having an open-ended meaning. That is, the above terms are to be interpreted synonymously with the phrases “having at least” or “including at least.” For example, when used in the context of a process, the term “comprising” means that the process includes at least the recited steps but may include additional steps. When used in the context of a compound, composition, or device, the term “comprising” means that the compound, composition, or device includes at least the recited features or components, but may also include additional features or components. As used herein, the term “covalently attached” or “covalently bonded” refers to the forming of a chemical bonding that is characterized by the sharing of pairs of electrons between atoms. For example, a covalently attached polymer coating refers to a polymer coating that forms chemical bonds with a functionalized surface of a substrate, as compared to attachment to the surface via other means, for example, adhesion or electrostatic interaction. It will be appreciated that polymers that are attached covalently to a surface can also be bonded via means in addition to covalent attachment. The term “halogen” or “halo,” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, e.g., fluorine, chlorine, bromine, or iodine, with fluorine and chlorine being preferred. As used herein, “alkyl” refers to a straight or branched hydrocarbon chain that is fully saturated (i.e., contains no double or triple bonds). The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 9 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 6 carbon atoms. The alkyl group may be designated as “C1-4alkyl” or similar designations. By way of example only, “C1-6alkyl” indicates that there are one to six carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, and the like. As used herein, “alkoxy” refers to the formula —OR wherein R is an alkyl as is defined above, such as “C1-9alkoxy”, including but not limited to methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, and tert-butoxy, and the like. As used herein, “alkenyl” refers to a straight or branched hydrocarbon chain containing one or more double bonds. The alkenyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term “alkenyl” where no numerical range is designated. The alkenyl group may also be a medium size alkenyl having 2 to 9 carbon atoms. The alkenyl group could also be a lower alkenyl having 2 to 6 carbon atoms. The alkenyl group may be designated as “C2-6alkenyl” or similar designations. By way of example only, “C2-6alkenyl” indicates that there are two to six carbon atoms in the alkenyl chain, i.e., the alkenyl chain is selected from the group consisting of ethenyl, propen-1-yl, propen-2-yl, propen-3-yl, buten-1-yl, buten-2-yl, buten-3-yl, buten-4-yl, 1-methyl-propen-1-yl, 2-methyl-propen-1-yl, 1-ethyl-ethen-1-yl, 2-methyl-propen-3-yl, buta-1,3-dienyl, buta-1,2-dienyl, and buta-1,2-dien-4-yl. Typical alkenyl groups include, but are in no way limited to, ethenyl, propenyl, butenyl, pentenyl, and hexenyl, and the like. As used herein, “alkynyl” refers to a straight or branched hydrocarbon chain containing one or more triple bonds. The alkynyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term “alkynyl” where no numerical range is designated. The alkynyl group may also be a medium size alkynyl having 2 to 9 carbon atoms. The alkynyl group could also be a lower alkynyl having 2 to 6 carbon atoms. The alkynyl group may be designated as “C2-6alkynyl” or similar designations. By way of example only, “C2-6alkynyl” indicates that there are two to six carbon atoms in the alkynyl chain, i.e., the alkynyl chain is selected from the group consisting of ethynyl, propyn-1-yl, propyn-2-yl, butyn-1-yl, butyn-3-yl, butyn-4-yl, and 2-butynyl. Typical alkynyl groups include, but are in no way limited to, ethynyl, propynyl, butynyl, pentynyl, and hexynyl, and the like. As used herein, “heteroalkyl” refers to a straight or branched hydrocarbon chain containing one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur, in the chain backbone. The heteroalkyl group may have 1 to 20 carbon atom, although the present definition also covers the occurrence of the term “heteroalkyl” where no numerical range is designated. The heteroalkyl group may also be a medium size heteroalkyl having 1 to 9 carbon atoms. The heteroalkyl group could also be a lower heteroalkyl having 1 to 6 carbon atoms. The heteroalkyl group may be designated as “C1-6heteroalkyl” or similar designations. The heteroalkyl group may contain one or more heteroatoms. By way of example only, “C4-6heteroalkyl” indicates that there are four to six carbon atoms in the heteroalkyl chain and additionally one or more heteroatoms in the backbone of the chain. The term “aromatic” refers to a ring or ring system having a conjugated pi electron system and includes both carbocyclic aromatic (e.g., phenyl) and heterocyclic aromatic groups (e.g., pyridine). The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of atoms) groups provided that the entire ring system is aromatic. As used herein, “aryl” refers to an aromatic ring or ring system (i.e., two or more fused rings that share two adjacent carbon atoms) containing only carbon in the ring backbone. When the aryl is a ring system, every ring in the system is aromatic. The aryl group may have 6 to 18 carbon atoms, although the present definition also covers the occurrence of the term “aryl” where no numerical range is designated. In some embodiments, the aryl group has 6 to 10 carbon atoms. The aryl group may be designated as “C6-10aryl,” “C6or C10aryl,” or similar designations. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, azulenyl, and anthracenyl. An “aralkyl” or “arylalkyl” is an aryl group connected, as a substituent, via an alkylene group, such as “C7-14aralkyl” and the like, including but not limited to benzyl, 2-phenylethyl, 3-phenylpropyl, and naphthylalkyl. In some cases, the alkylene group is a lower alkylene group (i.e., a C1-6alkylene group). As used herein, “heteroaryl” refers to an aromatic ring or ring system (i.e., two or more fused rings that share two adjacent atoms) that contain(s) one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur, in the ring backbone. When the heteroaryl is a ring system, every ring in the system is aromatic. The heteroaryl group may have 5-18 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term “heteroaryl” where no numerical range is designated. In some embodiments, the heteroaryl group has 5 to 10 ring members or 5 to 7 ring members. The heteroaryl group may be designated as “5-7 membered heteroaryl,” “5-10 membered heteroaryl,” or similar designations. Examples of heteroaryl rings include, but are not limited to, furyl, thienyl, phthalazinyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, indolyl, isoindolyl, and benzothienyl. A “heteroaralkyl” or “heteroarylalkyl” is heteroaryl group connected, as a substituent, via an alkylene group. Examples include but are not limited to 2-thienylmethyl, 3-thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazollylalkyl, and imidazolylalkyl. In some cases, the alkylene group is a lower alkylene group (i.e., a C1-6alkylene group). As used herein, “carbocyclyl” means a non-aromatic cyclic ring or ring system containing only carbon atoms in the ring system backbone. When the carbocyclyl is a ring system, two or more rings may be joined together in a fused, bridged or spiro-connected fashion. Carbocyclyls may have any degree of saturation provided that at least one ring in a ring system is not aromatic. Thus, carbocyclyls include cycloalkyls, cycloalkenyls, and cycloalkynyls. The carbocyclyl group may have 3 to 20 carbon atoms, although the present definition also covers the occurrence of the term “carbocyclyl” where no numerical range is designated. The carbocyclyl group may also be a medium size carbocyclyl having 3 to 10 carbon atoms. The carbocyclyl group could also be a carbocyclyl having 3 to 6 carbon atoms. The carbocyclyl group may be designated as “C3-6carbocyclyl” or similar designations. Examples of carbocyclyl rings include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,3-dihydro-indene, bicycle[2.2.2]octanyl, adamantyl, and spiro[4.4]nonanyl. As used herein, “cycloalkyl” means a fully saturated carbocyclyl ring or ring system. Examples include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. As used herein, “heterocyclyl” means a non-aromatic cyclic ring or ring system containing at least one heteroatom in the ring backbone. Heterocyclyls may be joined together in a fused, bridged or spiro-connected fashion. Heterocyclyls may have any degree of saturation provided that at least one ring in the ring system is not aromatic. The heteroatom(s) may be present in either a non-aromatic or aromatic ring in the ring system. The heterocyclyl group may have 3 to 20 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term “heterocyclyl” where no numerical range is designated. The heterocyclyl group may also be a medium size heterocyclyl having 3 to 10 ring members. The heterocyclyl group could also be a heterocyclyl having 3 to 6 ring members. The heterocyclyl group may be designated as “3-6 membered heterocyclyl” or similar designations. In preferred six membered monocyclic heterocyclyls, the heteroatom(s) are selected from one up to three of 0, N or S, and in preferred five membered monocyclic heterocyclyls, the heteroatom(s) are selected from one or two heteroatoms selected from O, N, or S. Examples of heterocyclyl rings include, but are not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, imidazolidinyl, morpholinyl, oxiranyl, oxepanyl, thiepanyl, piperidinyl, piperazinyl, dioxopiperazinyl, pyrrolidinyl, pyrrolidonyl, pyrrolidinonyl, 4-piperidonyl, pyrazolinyl, pyrazolidinyl, 1,3-dioxinyl, 1,3-dioxanyl, 1,4-dioxinyl, 1,4-dioxanyl, 1,3-oxathianyl, 1,4-oxathiinyl, 1,4-oxathianyl, 2H-1,2-oxazinyl, trioxanyl, hexahydro-1,3,5-triazinyl, 1,3-dioxolyl, 1,3-dioxolanyl, 1,3-dithiolyl, 1,3-dithiolanyl, isoxazolinyl, isoxazolidinyl, oxazolinyl, oxazolidinyl, oxazolidinonyl, thiazolinyl, thiazolidinyl, 1,3-oxathiolanyl, indolinyl, isoindolinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydro-1,4-thiazinyl, thiamorpholinyl, dihydrobenzofuranyl, benzimidazolidinyl, and tetrahydroquinoline. An “O-carboxy” group refers to a “—OC(═O)R” group in which R is selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl, as defined herein. A “C-carboxy” group refers to a “—C(═O)OR” group in which R is selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl, as defined herein. A non-limiting example includes carboxyl (i.e., —C(═O)OH). A “sulfonyl” group refers to an “—SO2R” group in which R is selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl, as defined herein. A “sulfino” group refers to a “—S(═O)OH” group. An “S-sulfonamido” group refers to a “—SO2NRARB” group in which RAand RBare each independently selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl, as defined herein. An “N-sulfonamido” group refers to a “—N(RA)SO2RB” group in which RAand Rbare each independently selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl, as defined herein. A “C-amido” group refers to a “—C(═O)NRARB” group in which RAand RBare each independently selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl, as defined herein. An “N-amido” group refers to a “—N(RA)C(═O)RB” group in which RAand RBare each independently selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl, as defined herein. An “amino” group refers to a “—NRARB” group in which RAand RBare each independently selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl, as defined herein. A non-limiting example includes free amino (i.e., —NH2). An “aminoalkyl” group refers to an amino group connected via an alkylene group. An “alkoxyalkyl” group refers to an alkoxy group connected via an alkylene group, such as a “C2-8alkoxyalkyl” and the like. As used herein, a substituted group is derived from the unsubstituted parent group in which there has been an exchange of one or more hydrogen atoms for another atom or group. Unless otherwise indicated, when a group is deemed to be “substituted,” it is meant that the group is substituted with one or more substituents independently selected from C1-C6alkyl, C1-C6alkenyl, C1-C6alkynyl, C1-C6heteroalkyl, C3-C7carbocyclyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), C3-C7-carbocyclyl-C1-C6-alkyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), 3-10 membered heterocyclyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), 3-10 membered heterocyclyl-C1-C6-alkyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), aryl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), aryl(C1-C6)alkyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), 5-10 membered heteroaryl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), 5-10 membered heteroaryl(C1-C6)alkyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), halo, —CN, hydroxy, C1-C6alkoxy, C1-C6alkoxy(C1-C6)alkyl (i.e., ether), aryloxy, sulfhydryl (mercapto), halo(C1-C6)alkyl (e.g., —CF3), halo(C1-C6)alkoxy (e.g., —OCF3), C1-C6alkylthio, arylthio, amino, amino(C1-C6)alkyl, nitro, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, acyl, cyanato, isocyanato, thiocyanato, isothiocyanato, sulfinyl, sulfonyl, —SO3H, sulfino, —OSO2C1-4alkyl, and oxo (═O). Wherever a group is described as “optionally substituted” that group can be substituted with the above substituents. In some embodiments, substituted alkyl, alkenyl, or alkynyl groups are substituted with one or more substituents selected from the group consisting of halo, —CN, SO3−, SRa, ORa, NRbRc, oxo, CONRbRc, COOH, and COORb, where Ra, Rband Rcare each independently selected from H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, and substituted aryl. Compounds described herein can be represented as several mesomeric forms. Where a single structure is drawn, any of the relevant mesomeric forms are intended. The coumarin compounds described herein are represented by a single structure but can equally be shown as any of the related mesomeric forms. Exemplary mesomeric structures are shown below for Formula (I): In each instance where a single mesomeric form of a compound described herein is shown, the alternative mesomeric forms are equally contemplated. As understood by one of ordinary skill in the art, a compound described herein may exist in ionized form, e.g., —CO2−or —SO3−. If a compound contains a positively or negatively charged substituent group, for example, SO3−, it may also contain a negatively or positively charged counterion such that the compound as a whole is neutral. In other aspects, the compound may exist in a salt form, where the counterion is provided by a conjugate acid or base. It is to be understood that certain radical naming conventions can include either a mono-radical or a di-radical, depending on the context. For example, where a substituent requires two points of attachment to the rest of the molecule, it is understood that the substituent is a di-radical. For example, a substituent identified as alkyl that requires two points of attachment includes di-radicals such as —CH2—, —CH2CH2—, —CH2CH(CH3)CH2—, and the like. Other radical naming conventions clearly indicate that the radical is a di-radical such as “alkylene” or “alkenylene.” When two “adjacent” R groups are said to form a ring “together with the atom to which they are attached,” it is meant that the collective unit of the atoms, intervening bonds, and the two R groups are the recited ring. For example, when the following substructure is present: and R1and R2are defined as selected from the group consisting of hydrogen and alkyl, or R1and R2together with the atoms to which they are attached form an aryl or carbocyclyl, it is meant that R1and R2can be selected from hydrogen or alkyl, or alternatively, the substructure has structure: where A is an aryl ring or a carbocyclyl containing the depicted double bond. Labeled Nucleotides According to an aspect of the disclosure, there are provided dye compounds suitable for attachment to substrate moieties, particularly comprising linker groups to enable attachment to substrate moieties. Substrate moieties can be virtually any molecule or substance to which the dyes of the disclosure can be conjugated, and, by way of non-limiting example, may include nucleosides, nucleotides, polynucleotides, carbohydrates, ligands, particles, solid surfaces, organic and inorganic polymers, chromosomes, nuclei, living cells, and combinations or assemblages thereof. The dyes can be conjugated by an optional linker by a variety of means including hydrophobic attraction, ionic attraction, and covalent attachment. In some aspects, the dyes are conjugated to the substrate by covalent attachment. More particularly, the covalent attachment is by means of a linker group. In some instances, such labeled nucleotides are also referred to as “modified nucleotides.” The present disclosure further provides conjugates of nucleosides and nucleotides labeled with one or more of the dyes set forth herein (modified nucleotides). Labeled nucleosides and nucleotides are useful for labeling polynucleotides formed by enzymatic synthesis, such as, by way of non-limiting example, in PCR amplification, isothermal amplification, solid phase amplification, polynucleotide sequencing (e.g., solid phase sequencing), nick translation reactions and the like. The attachment to the biomolecules may be via the R, R1, R2, R3, R4, R5, or X position of the compound of Formula (I). In some aspects, the connection is via the R3or R5group of Formula (I). For Formula (II), attachment may be at any position R6-11or X′. In some embodiments, the substituent group is a substituted alkyl, for example, alkyl substituted with —CO2H or an activated form of carboxyl group, for example, an amide or ester, which may be used for attachment to the amino or hydroxyl group of the biomolecules. In one embodiment, the R, R1, R2, R3, R4, R5, or X group of Formula (I) or the R6-11or X′ groups of Formula (II) may contain an activated ester or amide residue most suitable for further amide/peptide bond formation. The term “activated ester” as used herein, refers to a carboxyl group derivative which is capable of reacting in mild conditions, for example, with a compound containing an amino group. Non-limiting examples of activated esters include but not limited to p-nitrophenyl, pentafluorophenyl and succinimido esters. In some embodiments, the dye compounds may be covalently attached to oligonucleotides or nucleotides via the nucleotide base. For example, the labeled nucleotide or oligonucleotide may have the label attached to the C5 position of a pyrimidine base or the C7 position of a 7-deaza purine base through a linker moiety. The labeled nucleotide or oligonucleotide may also have a 3′-OH blocking group covalently attached to the ribose or deoxyribose sugar of the nucleotide. A particular useful application of the new fluorescent dyes as described herein is for labeling biomolecules, for example, nucleotides or oligonucleotides. Some embodiments of the present application are directed to a nucleotide or oligonucleotide labeled with the new fluorescent compounds as described herein. Linkers The dye compounds as disclosed herein may include a reactive linker group at one of the substituent positions for covalent attachment of the compound to a substrate or another molecule. Reactive linking groups are moieties capable of forming a bond (e.g., a covalent or non-covalent bond), in particular a covalent bond. In a particular embodiment the linker may be a cleavable linker. Use of the term “cleavable linker” is not meant to imply that the whole linker is required to be removed. The cleavage site can be located at a position on the linker that ensures that part of the linker remains attached to the dye and/or substrate moiety after cleavage. Cleavable linkers may be, by way of non-limiting example, electrophilically cleavable linkers, nucleophilically cleavable linkers, photocleavable linkers, cleavable under reductive conditions (for example disulfide or azide containing linkers), oxidative conditions, cleavable via use of safety-catch linkers and cleavable by elimination mechanisms. The use of a cleavable linker to attach the dye compound to a substrate moiety ensures that the label can, if required, be removed after detection, avoiding any interfering signal in downstream steps. Useful linker groups may be found in PCT Publication No. WO2004/018493 (herein incorporated by reference), examples of which include linkers that may be cleaved using water-soluble phosphines or water-soluble transition metal catalysts formed from a transition metal and at least partially water-soluble ligands. In aqueous solution the latter form at least partially water-soluble transition metal complexes. Such cleavable linkers can be used to connect bases of nucleotides to labels such as the dyes set forth herein. Particular linkers include those disclosed in PCT Publication No. WO2004/018493 (herein incorporated by reference) such as those that include moieties of the formulae: (wherein X is selected from the group comprising O, S, NH and NQ wherein Q is a C1-10 substituted or unsubstituted alkyl group, Y is selected from the group comprising O, S, NH and N(allyl), T is hydrogen or a C1-C10substituted or unsubstituted alkyl group and * indicates where the moiety is connected to the remainder of the nucleotide or nucleoside). In some aspects, the linkers connect the bases of nucleotides to labels such as, for example, the dye compounds described herein. In particular embodiments, the length of the linker between a fluorescent dye (fluorophore) and a guanine base can be altered, for example, by introducing a polyethylene glycol spacer group, thereby increasing the fluorescence intensity compared to the same fluorophore attached to the guanine base through other linkages known in the art. Exemplary linkers and their properties are set forth in PCT Publication No. WO2007020457 (herein incorporated by reference). The design of linkers, and especially their increased length, can allow improvements in the brightness of fluorophores attached to the guanine bases of guanosine nucleotides when incorporated into polynucleotides such as DNA. Thus, when the dye is for use in any method of analysis which requires detection of a fluorescent dye label attached to a guanine-containing nucleotide, it is advantageous if the linker comprises a spacer group of formula —((CH2)2O)n—, wherein n is an integer between 2 and 50, as described in WO 2007/020457. Nucleosides and nucleotides may be labeled at sites on the sugar or nucleobase. As known in the art, a “nucleotide” consists of a nitrogenous base, a sugar, and one or more phosphate groups. In RNA, the sugar is ribose and in DNA is a deoxyribose, i.e., a sugar lacking a hydroxyl group that is present in ribose. The nitrogenous base is a derivative of purine or pyrimidine. The purines are adenine (A) and guanine (G), and the pyrimidines are cytosine (C) and thymine (T) or in the context of RNA, uracil (U). The C-1 atom of deoxyribose is bonded to N-1 of a pyrimidine or N-9 of a purine. A nucleotide is also a phosphate ester of a nucleoside, with esterification occurring on the hydroxyl group attached to the C-3 or C-5 of the sugar. Nucleotides are usually mono, di- or triphosphates. A “nucleoside” is structurally similar to a nucleotide but is missing the phosphate moieties. An example of a nucleoside analog would be one in which the label is linked to the base and there is no phosphate group attached to the sugar molecule. Although the base is usually referred to as a purine or pyrimidine, the skilled person will appreciate that derivatives and analogues are available which do not alter the capability of the nucleotide or nucleoside to undergo Watson-Crick base pairing. “Derivative” or “analogue” means a compound or molecule whose core structure is the same as, or closely resembles that of a parent compound but which has a chemical or physical modification, such as, for example, a different or additional side group, which allows the derivative nucleotide or nucleoside to be linked to another molecule. For example, the base may be a deazapurine. In particular embodiments, the derivatives should be capable of undergoing Watson-Crick pairing. “Derivative” and “analogue” also include, for example, a synthetic nucleotide or nucleoside derivative having modified base moieties and/or modified sugar moieties. Such derivatives and analogues are discussed in, for example, Scheit,Nucleotide analogs(John Wiley & Son, 1980) and Uhlman et al.,Chemical Reviews90:543-584, 1990. Nucleotide analogues can also comprise modified phosphodiester linkages including phosphorothioate, phosphorodithioate, alkyl-phosphonate, phosphoranilidate, phosphoramidate linkages and the like. A dye may be attached to any position on the nucleotide base, for example, through a linker. In particular embodiments, Watson-Crick base pairing can still be carried out for the resulting analog. Particular nucleobase labeling sites include the C5 position of a pyrimidine base or the C7 position of a 7-deaza purine base. As described above a linker group may be used to covalently attach a dye to the nucleoside or nucleotide. In particular embodiments the labeled nucleoside or nucleotide may be enzymatically incorporable and enzymatically extendable. Accordingly, a linker moiety may be of sufficient length to connect the nucleotide to the compound such that the compound does not significantly interfere with the overall binding and recognition of the nucleotide by a nucleic acid replication enzyme. Thus, the linker can also comprise a spacer unit. The spacer distances, for example, the nucleotide base from a cleavage site or label. Nucleosides or nucleotides labeled with the dyes described herein may have the formula: whereDye is a dye compound,B is a nucleobase, such as, for example uracil, thymine, cytosine, adenine, guanine and the like,L is an optional linker group which may or may not be present,R′ can be H, monophosphate, diphosphate, triphosphate, thiophosphate, a phosphate ester analog, —O— attached to a reactive phosphorous containing group, or —O— protected by a blocking group,R″ can be H, OH, a phosphoramidite, or a 3′-OH blocking group, andR′″ is H or OH. Where R″ is phosphoramidite, R′ is an acid-cleavable hydroxyl protecting group which allows subsequent monomer coupling under automated synthesis conditions. In a particular embodiment, the blocking group is separate and independent of the dye compound, i.e., not attached to it. Alternatively, the dye may comprise all or part of the 3′-OH blocking group. Thus R″ can be a 3′-OH blocking group which may or may not comprise the dye compound. In yet another alternative embodiment, there is no blocking group on the 3′ carbon of the pentose sugar and the dye (or dye and linker construct) attached to the base, for example, can be of a size or structure sufficient to act as a block to the incorporation of a further nucleotide. Thus, the block can be due to steric hindrance or can be due to a combination of size, charge and structure, whether or not the dye is attached to the 3′ position of the sugar. In still yet another alternative embodiment, the blocking group is present on the 2′ or 4′ carbon of the pentose sugar and can be of a size or structure sufficient to act as a block to the incorporation of a further nucleotide. The use of a blocking group allows polymerization to be controlled, such as by stopping extension when a modified nucleotide is incorporated. If the blocking effect is reversible, for example, by way of non-limiting example by changing chemical conditions or by removal of a chemical block, extension can be stopped at certain points and then allowed to continue. In another particular embodiment, a 3′-OH blocking group will comprise a moiety disclosed in WO2004/018497 and WO2014/139596, which are hereby incorporated by references. For example the blocking group may be azidomethyl (—CH2N3) or substituted azidomethyl (e.g., —CH(CHF2)N3or CH(CH2F)N3), or allyl. In a particular embodiment, the linker (between dye and nucleotide) and blocking group are both present and are separate moieties. In particular embodiments, the linker and blocking group are both cleavable under substantially similar conditions. Thus, deprotection and deblocking processes may be more efficient because only a single treatment will be required to remove both the dye compound and the blocking group. However, in some embodiments a linker and blocking group need not be cleavable under similar conditions, instead being individually cleavable under distinct conditions. The disclosure also encompasses polynucleotides incorporating dye compounds. Such polynucleotides may be DNA or RNA comprised respectively of deoxyribonucleotides or ribonucleotides joined in phosphodiester linkage. Polynucleotides may comprise naturally occurring nucleotides, non-naturally occurring (or modified) nucleotides other than the labeled nucleotides described herein or any combination thereof, in combination with at least one modified nucleotide (e.g., labeled with a dye compound) as set forth herein. Polynucleotides according to the disclosure may also include non-natural backbone linkages and/or non-nucleotide chemical modifications. Chimeric structures comprised of mixtures of ribonucleotides and deoxyribonucleotides comprising at least one labeled nucleotide are also contemplated. Non-limiting exemplary labeled nucleotides as described herein include: wherein L represents a linker and R represents a sugar residue as described above. In some embodiments, non-limiting exemplary fluorescent dye conjugates are shown below: Kits The present disclosure also provides kits including modified nucleosides and/or nucleotides labeled with dyes. Such kits will generally include at least one modified nucleotide or nucleoside labeled with a dye set forth herein together with at least one further component. The further component(s) may be one or more of the components identified in a method set forth herein or in the Examples section below. Some non-limiting examples of components that can be combined into a kit of the present disclosure are set forth below. In a particular embodiment, a kit can include at least one modified nucleotide or nucleoside labeled with a dye set forth herein together with modified or unmodified nucleotides or nucleosides. For example, modified nucleotides labeled with dyes according to the disclosure may be supplied in combination with unlabeled or native nucleotides, and/or with fluorescently labeled nucleotides or any combination thereof. Accordingly, the kits may comprise modified nucleotides labeled with dyes according to the disclosure and modified nucleotides labeled with other, for example, prior art dye compounds. Combinations of nucleotides may be provided as separate individual components (e.g., one nucleotide type per vessel or tube) or as nucleotide mixtures (e.g., two or more nucleotides mixed in the same vessel or tube). Where kits comprise a plurality, particularly two, or three, or more particularly four, modified nucleotides labeled with a dye compound, the different nucleotides may be labeled with different dye compounds, or one may be dark, with no dye compounds. Where the different nucleotides are labeled with different dye compounds, it is a feature of the kits that the dye compounds are spectrally distinguishable fluorescent dyes. As used herein, the term “spectrally distinguishable fluorescent dyes” refers to fluorescent dyes that emit fluorescent energy at wavelengths that can be distinguished by fluorescent detection equipment (for example, a commercial capillary-based DNA sequencing platform) when two or more such dyes are present in one sample. When two modified nucleotides labeled with fluorescent dye compounds are supplied in kit form, it is a feature of some embodiments that the spectrally distinguishable fluorescent dyes can be excited at the same wavelength, such as, for example by the same laser. When four modified nucleotides labeled with fluorescent dye compounds are supplied in kit form, it is a feature of some embodiments that two of the spectrally distinguishable fluorescent dyes can both be excited at one wavelength and the other two spectrally distinguishable dyes can both be excited at another wavelength. Particular excitation wavelengths are 488 nm and 532 nm. In one embodiment, a kit includes a modified nucleotide labeled with a compound of the present disclosure and a second modified nucleotide labeled with a second dye wherein the dyes have a difference in absorbance maximum of at least 10 nm, particularly 20 nm to 50 nm. More particularly, the two dye compounds have Stokes shifts of between 15-40 nm where “Stokes shift” is the distance between the peak absorption and peak emission wavelengths. In a further embodiment, a kit can further include two other modified nucleotides labeled with fluorescent dyes wherein the dyes are excited by the same laser at 532 nm. The dyes can have a difference in absorbance maximum of at least 10 nm, particularly 20 nm to 50 nm. More particularly the two dye compounds can have Stokes shifts of between 20-40 nm. Particular dyes which are spectrally distinguishable from dyes of the present disclosure and which meet the above criteria are polymethine analogues as described in U.S. Pat. No. 5,268,486 (for example Cy3) or WO 0226891 (Alexa 532; Molecular Probes A20106) or unsymmetrical polymethines as disclosed in U.S. Pat. No. 6,924,372, each of which is incorporated herein by reference. Alternative dyes include rhodamine analogues, for example tetramethyl rhodamine and analogues thereof. In an alternative embodiment, the kits of the disclosure may contain nucleotides where the same base is labeled with two different compounds. A first nucleotide may be labeled with a compound of the disclosure. A second nucleotide may be labeled with a spectrally distinct compound, for example a ‘green’ dye absorbing at less than 600 nm. A third nucleotide may be labeled as a mixture of the compound of the disclosure and the spectrally distinct compound, and the fourth nucleotide may be ‘dark’ and contain no label. In simple terms, therefore, the nucleotides 1-4 may be labeled ‘blue’, ‘green’, ‘blue/green’, and dark. To simplify the instrumentation further, four nucleotides can be labeled with two dyes excited with a single laser, and thus the labeling of nucleotides 1-4 may be ‘blue 1’, ‘blue 2’, ‘blue 1/blue 2’, and dark. Nucleotides may contain two dyes of the present disclosure. A kit may contain two or more nucleotides labeled with dyes of the disclosure. Kits may contain a further nucleotide where the nucleotide is labeled with a dye that absorbs in the region of 520 nm to 560 nm. Kits may further contain an unlabeled nucleotide. Although kits are exemplified herein in regard to configurations having different nucleotides that are labeled with different dye compounds, it will be understood that kits can include 2, 3, 4 or more different nucleotides that have the same dye compound. In particular embodiments, a kit may include a polymerase enzyme capable of catalyzing incorporation of the modified nucleotides into a polynucleotide. Other components to be included in such kits may include buffers and the like. The modified nucleotides labeled with dyes according to the disclosure, and other any nucleotide components including mixtures of different nucleotides, may be provided in the kit in a concentrated form to be diluted prior to use. In such embodiments a suitable dilution buffer may also be included. Again, one or more of the components identified in a method set forth herein can be included in a kit of the present disclosure. Methods of Sequencing Modified nucleotides (or nucleosides) comprising a dye compound according to the present disclosure may be used in any method of analysis such as method that include detection of a fluorescent label attached to a nucleotide or nucleoside, whether on its own or incorporated into or associated with a larger molecular structure or conjugate. In this context the term “incorporated into a polynucleotide” can mean that the 5′ phosphate is joined in phosphodiester linkage to the 3′ hydroxyl group of a second (modified or unmodified) nucleotide, which may itself form part of a longer polynucleotide chain. The 3′ end of a modified nucleotide set forth herein may or may not be joined in phosphodiester linkage to the 5′ phosphate of a further (modified or unmodified) nucleotide. Thus, in one non-limiting embodiment, the disclosure provides a method of detecting a modified nucleotide incorporated into a polynucleotide which comprises: (a) incorporating at least one modified nucleotide of the disclosure into a polynucleotide and (b) detecting the modified nucleotide(s) incorporated into the polynucleotide by detecting the fluorescent signal from the dye compound attached to said modified nucleotide(s). This method can include: a synthetic step (a) in which one or more modified nucleotides according to the disclosure are incorporated into a polynucleotide and a detection step (b) in which one or more modified nucleotide(s) incorporated into the polynucleotide are detected by detecting or quantitatively measuring their fluorescence. Some embodiments of the present application are directed to methods of sequencing including: (a) incorporating at least one labeled nucleotide as described herein into a polynucleotide; and (b) detecting the labeled nucleotide(s) incorporated into the polynucleotide by detecting the fluorescent signal from the new fluorescent dye attached to said modified nucleotide(s). In one embodiment, at least one modified nucleotide is incorporated into a polynucleotide in the synthetic step by the action of a polymerase enzyme. However, other methods of joining modified nucleotides to polynucleotides, such as, for example, chemical oligonucleotide synthesis or ligation of labeled oligonucleotides to unlabeled oligonucleotides, can be used. Therefore, the term “incorporating,” when used in reference to a nucleotide and polynucleotide, can encompass polynucleotide synthesis by chemical methods as well as enzymatic methods. In a specific embodiment, a synthetic step is carried out and may optionally comprise incubating a template polynucleotide strand with a reaction mixture comprising fluorescently labeled modified nucleotides of the disclosure. A polymerase can also be provided under conditions which permit formation of a phosphodiester linkage between a free 3′ hydroxyl group on a polynucleotide strand annealed to the template polynucleotide strand and a 5′ phosphate group on the modified nucleotide. Thus, a synthetic step can include formation of a polynucleotide strand as directed by complementary base-pairing of nucleotides to a template strand. In all embodiments of the methods, the detection step may be carried out while the polynucleotide strand into which the labeled nucleotides are incorporated is annealed to a template strand, or after a denaturation step in which the two strands are separated. Further steps, for example chemical or enzymatic reaction steps or purification steps, may be included between the synthetic step and the detection step. In particular, the target strand incorporating the labeled nucleotide(s) may be isolated or purified and then processed further or used in a subsequent analysis. By way of example, target polynucleotides labeled with modified nucleotide(s) as described herein in a synthetic step may be subsequently used as labeled probes or primers. In other embodiments, the product of the synthetic step set forth herein may be subject to further reaction steps and, if desired, the product of these subsequent steps purified or isolated. Suitable conditions for the synthetic step will be well known to those familiar with standard molecular biology techniques. In one embodiment, a synthetic step may be analogous to a standard primer extension reaction using nucleotide precursors, including modified nucleotides as described herein, to form an extended target strand complementary to the template strand in the presence of a suitable polymerase enzyme. In other embodiments, the synthetic step may itself form part of an amplification reaction producing a labeled double stranded amplification product comprised of annealed complementary strands derived from copying of the target and template polynucleotide strands. Other exemplary synthetic steps include nick translation, strand displacement polymerization, random primed DNA labeling, etc. A particularly useful polymerase enzyme for a synthetic step is one that is capable of catalyzing the incorporation of modified nucleotides as set forth herein. A variety of naturally occurring or modified polymerases can be used. By way of example, a thermostable polymerase can be used for a synthetic reaction that is carried out using thermocycling conditions, whereas a thermostable polymerase may not be desired for isothermal primer extension reactions. Suitable thermostable polymerases which are capable of incorporating the modified nucleotides according to the disclosure include those described in WO 2005/024010 or WO06120433, each of which is incorporated herein by reference. In synthetic reactions which are carried out at lower temperatures such as 37° C., polymerase enzymes need not necessarily be thermostable polymerases, therefore the choice of polymerase will depend on a number of factors such as reaction temperature, pH, strand-displacing activity and the like. In specific non-limiting embodiments, the disclosure encompasses methods of nucleic acid sequencing, re-sequencing, whole genome sequencing, single nucleotide polymorphism scoring, any other application involving the detection of the modified nucleotide or nucleoside labeled with dyes set forth herein when incorporated into a polynucleotide. Any of a variety of other applications benefitting the use of polynucleotides labeled with the modified nucleotides comprising fluorescent dyes can use modified nucleotides or nucleosides with dyes set forth herein. In a particular embodiment the disclosure provides use of modified nucleotides comprising dye compounds according to the disclosure in a polynucleotide sequencing-by-synthesis reaction. Sequencing-by-synthesis generally involves sequential addition of one or more nucleotides or oligonucleotides to a growing polynucleotide chain in the 5′ to 3′ direction using a polymerase or ligase in order to form an extended polynucleotide chain complementary to the template nucleic acid to be sequenced. The identity of the base present in one or more of the added nucleotide(s) can be determined in a detection or “imaging” step. The identity of the added base may be determined after each nucleotide incorporation step. The sequence of the template may then be inferred using conventional Watson-Crick base-pairing rules. The use of the modified nucleotides labeled with dyes set forth herein for determination of the identity of a single base may be useful, for example, in the scoring of single nucleotide polymorphisms, and such single base extension reactions are within the scope of this disclosure. In an embodiment of the present disclosure, the sequence of a template polynucleotide is determined by detecting the incorporation of one or more nucleotides into a nascent strand complementary to the template polynucleotide to be sequenced through the detection of fluorescent label(s) attached to the incorporated nucleotide(s). Sequencing of the template polynucleotide can be primed with a suitable primer (or prepared as a hairpin construct which will contain the primer as part of the hairpin), and the nascent chain is extended in a stepwise manner by addition of nucleotides to the 3′ end of the primer in a polymerase-catalyzed reaction. In particular embodiments, each of the different nucleotide triphosphates (A, T, G and C) may be labeled with a unique fluorophore and also comprises a blocking group at the 3′ position to prevent uncontrolled polymerization. Alternatively, one of the four nucleotides may be unlabeled (dark). The polymerase enzyme incorporates a nucleotide into the nascent chain complementary to the template polynucleotide, and the blocking group prevents further incorporation of nucleotides. Any unincorporated nucleotides can be washed away and the fluorescent signal from each incorporated nucleotide can be “read” optically by suitable means, such as a charge-coupled device using laser excitation and suitable emission filters. The 3′-blocking group and fluorescent dye compounds can then be removed (deprotected) (simultaneously or sequentially) to expose the nascent chain for further nucleotide incorporation. Typically, the identity of the incorporated nucleotide will be determined after each incorporation step, but this is not strictly essential. Similarly, U.S. Pat. No. 5,302,509 (which is incorporated herein by reference) discloses a method to sequence polynucleotides immobilized on a solid support. The method, as exemplified above, utilizes the incorporation of fluorescently labeled, 3′-blocked nucleotides A, G, C, and T into a growing strand complementary to the immobilized polynucleotide, in the presence of DNA polymerase. The polymerase incorporates a base complementary to the target polynucleotide but is prevented from further addition by the 3′-blocking group. The label of the incorporated nucleotide can then be determined, and the blocking group removed by chemical cleavage to allow further polymerization to occur. The nucleic acid template to be sequenced in a sequencing-by-synthesis reaction may be any polynucleotide that it is desired to sequence. The nucleic acid template for a sequencing reaction will typically comprise a double stranded region having a free 3′ hydroxyl group that serves as a primer or initiation point for the addition of further nucleotides in the sequencing reaction. The region of the template to be sequenced will overhang this free 3′ hydroxyl group on the complementary strand. The overhanging region of the template to be sequenced may be single stranded but can be double-stranded, provided that a “nick is present” on the strand complementary to the template strand to be sequenced to provide a free 3′ OH group for initiation of the sequencing reaction. In such embodiments, sequencing may proceed by strand displacement. In certain embodiments, a primer bearing the free 3′ hydroxyl group may be added as a separate component (e.g., a short oligonucleotide) that hybridizes to a single-stranded region of the template to be sequenced. Alternatively, the primer and the template strand to be sequenced may each form part of a partially self-complementary nucleic acid strand capable of forming an intra-molecular duplex, such as for example a hairpin loop structure. Hairpin polynucleotides and methods by which they may be attached to solid supports are disclosed in PCT Publication Nos. WO0157248 and WO2005/047301, each of which is incorporated herein by reference. Nucleotides can be added successively to a growing primer, resulting in synthesis of a polynucleotide chain in the 5′ to 3′ direction. The nature of the base which has been added may be determined, particularly but not necessarily after each nucleotide addition, thus providing sequence information for the nucleic acid template. Thus, a nucleotide is incorporated into a nucleic acid strand (or polynucleotide) by joining of the nucleotide to the free 3′ hydroxyl group of the nucleic acid strand via formation of a phosphodiester linkage with the 5′ phosphate group of the nucleotide. The nucleic acid template to be sequenced may be DNA or RNA, or even a hybrid molecule comprised of deoxynucleotides and ribonucleotides. The nucleic acid template may comprise naturally occurring and/or non-naturally occurring nucleotides and natural or non-natural backbone linkages, provided that these do not prevent copying of the template in the sequencing reaction. In certain embodiments, the nucleic acid template to be sequenced may be attached to a solid support via any suitable linkage method known in the art, for example via covalent attachment. In certain embodiments template polynucleotides may be attached directly to a solid support (e.g., a silica-based support). However, in other embodiments of the disclosure the surface of the solid support may be modified in some way so as to allow either direct covalent attachment of template polynucleotides, or to immobilize the template polynucleotides through a hydrogel or polyelectrolyte multilayer, which may itself be non-covalently attached to the solid support. Arrays in which polynucleotides have been directly attached to silica-based supports are those for example disclosed in WO00006770 (incorporated herein by reference), wherein polynucleotides are immobilized on a glass support by reaction between a pendant epoxide group on the glass with an internal amino group on the polynucleotide. In addition, polynucleotides can be attached to a solid support by reaction of a sulfur-based nucleophile with the solid support, for example, as described in WO2005/047301 (incorporated herein by reference). A still further example of solid-supported template polynucleotides is where the template polynucleotides are attached to hydrogel supported upon silica-based or other solid supports, for example, as described in WO00/31148, WO01/01143, WO02/12566, WO03/014392, U.S. Pat. No. 6,465,178 and WO00/53812, each of which is incorporated herein by reference. A particular surface to which template polynucleotides may be immobilized is a polyacrylamide hydrogel. Polyacrylamide hydrogels are described in the references cited above and in WO2005/065814, which is incorporated herein by reference. Specific hydrogels that may be used include those described in WO 2005/065814 and U.S. Pub. No. 2014/0079923. In one embodiment, the hydrogel is PAZAM (poly(N-(5-azidoacetamidylpentyl) acrylamide-co-acrylamide)). DNA template molecules can be attached to beads or microparticles, for example, as described in U.S. Pat. No. 6,172,218 (which is incorporated herein by reference). Attachment to beads or microparticles can be useful for sequencing applications. Bead libraries can be prepared where each bead contains different DNA sequences. Exemplary libraries and methods for their creation are described in Nature, 437, 376-380 (2005); Science, 309, 5741, 1728-1732 (2005), each of which is incorporated herein by reference. Sequencing of arrays of such beads using nucleotides set forth herein is within the scope of the disclosure. Template(s) that are to be sequenced may form part of an “array” on a solid support, in which case the array may take any convenient form. Thus, the method of the disclosure is applicable to all types of high-density arrays, including single-molecule arrays, clustered arrays, and bead arrays. Modified nucleotides labeled with dye compounds of the present disclosure may be used for sequencing templates on essentially any type of array, including but not limited to those formed by immobilization of nucleic acid molecules on a solid support. However, the modified nucleotides labeled with dye compounds of the disclosure are particularly advantageous in the context of sequencing of clustered arrays. In clustered arrays, distinct regions on the array (often referred to as sites, or features) comprise multiple polynucleotide template molecules. Generally, the multiple polynucleotide molecules are not individually resolvable by optical means and are instead detected as an ensemble. Depending on how the array is formed, each site on the array may comprise multiple copies of one individual polynucleotide molecule (e.g., the site is homogenous for a particular single- or double-stranded nucleic acid species) or even multiple copies of a small number of different polynucleotide molecules (e.g., multiple copies of two different nucleic acid species). Clustered arrays of nucleic acid molecules may be produced using techniques generally known in the art. By way of example, WO 98/44151 and WO00/18957, each of which is incorporated herein, describe methods of amplification of nucleic acids wherein both the template and amplification products remain immobilized on a solid support in order to form arrays comprised of clusters or “colonies” of immobilized nucleic acid molecules. The nucleic acid molecules present on the clustered arrays prepared according to these methods are suitable templates for sequencing using the modified nucleotides labeled with dye compounds of the disclosure. The modified nucleotides labeled with dye compounds of the present disclosure are also useful in sequencing of templates on single molecule arrays. The term “single molecule array” or “SMA” as used herein refers to a population of polynucleotide molecules, distributed (or arrayed) over a solid support, wherein the spacing of any individual polynucleotide from all others of the population is such that it is possible to individually resolve the individual polynucleotide molecules. The target nucleic acid molecules immobilized onto the surface of the solid support can thus be capable of being resolved by optical means in some embodiments. This means that one or more distinct signals, each representing one polynucleotide, will occur within the resolvable area of the particular imaging device used. Single molecule detection may be achieved wherein the spacing between adjacent polynucleotide molecules on an array is at least 100 nm, more particularly at least 250 nm, still more particularly at least 300 nm, even more particularly at least 350 nm. Thus, each molecule is individually resolvable and detectable as a single molecule fluorescent point, and fluorescence from said single molecule fluorescent point also exhibits single step photobleaching. The terms “individually resolved” and “individual resolution” are used herein to specify that, when visualized, it is possible to distinguish one molecule on the array from its neighboring molecules. Separation between individual molecules on the array will be determined, in part, by the particular technique used to resolve the individual molecules. The general features of single molecule arrays will be understood by reference to published applications WO00/06770 and WO 01/57248, each of which is incorporated herein by reference. Although one use of the modified nucleotides of the disclosure is in sequencing-by-synthesis reactions, the utility of the modified nucleotides is not limited to such methods. In fact, the nucleotides may be used advantageously in any sequencing methodology which requires detection of fluorescent labels attached to nucleotides incorporated into a polynucleotide. In particular, the modified nucleotides labeled with dye compounds of the disclosure may be used in automated fluorescent sequencing protocols, particularly fluorescent dye-terminator cycle sequencing based on the chain termination sequencing method of Sanger and co-workers. Such methods generally use enzymes and cycle sequencing to incorporate fluorescently labeled dideoxynucleotides in a primer extension sequencing reaction. So-called Sanger sequencing methods, and related protocols (Sanger-type), utilize randomized chain termination with labeled dideoxynucleotides. Thus, the present disclosure also encompasses modified nucleotides labeled with dye compounds which are dideoxynucleotides lacking hydroxyl groups at both of the 3′ and 2′ positions, such modified dideoxynucleotides being suitable for use in Sanger type sequencing methods and the like. Modified nucleotides labeled with dye compounds of the present disclosure incorporating 3′ blocking groups, it will be recognized, may also be of utility in Sanger methods and related protocols since the same effect achieved by using modified dideoxy nucleotides may be achieved by using modified nucleotides having 3′-OH blocking groups: both prevent incorporation of subsequent nucleotides. Where nucleotides according to the present disclosure, and having a 3′ blocking group are to be used in Sanger-type sequencing methods it will be appreciated that the dye compounds or detectable labels attached to the nucleotides need not be connected via cleavable linkers, since in each instance where a labeled nucleotide of the disclosure is incorporated; no nucleotides need to be subsequently incorporated and thus the label need not be removed from the nucleotide. EXAMPLES Additional embodiments are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the claims. Additional embodiments are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the claims. Table 2 summarizes spectral properties of the new coumarin fluorescent dyes disclosed in the examples. Table 3 summarizes the structure and spectral properties of various nucleotides labeled with new dyes disclosed herein. Example 1-1-1: 7-(5-Carboxypentyl)amino-3-(benzothiazol-2-yl)coumarin 3-(Benzothiazol-2-yl)-7-fluoro-coumarin derivative (FC-1, 0.4 g, 1.345 mmol, 1 eqv) and 6-aminohexanoic acid (AC-05, 0.25 g, 1.906 mmol, 1.417 eqv) was added to anhydrous dimethyl sulfoxide (DMSO, 3 mL). After the addition was complete, the mixture was stirred for a few minutes at room temperature and then N,N-diisopropyl-N-ethylamine (DIPEA, 0.25 g, 2 mmol, 2 eqv) was added to this mixture. The reaction mixture was stirred for 3 hours at 120° C. After standing at room temperature for 1 hour, the yellow, semi-solid reaction mixture was diluted with water (5 mL) and stirred overnight. The resulting precipitate was collected by suction filtration. Yield 0.36 g (65.5%). MS (DUIS): MW Calculated 408.47. Found m/z: (+) 409 (M+1)+; (−), 407 (M−1)−.1H NMR (400 MHz, DMSO-d6) δ: 12.03 (m, 2H), 9.00 (s, 1H), 8.12 (d, J=7.9 Hz, 1H), 7.99 (d, J=8.1 Hz, 1H), 6.73 (dd, J=8.8, 2.1 Hz, 1H), 6.54 (d, J=2.0 Hz, 1H), 3.18 (q, J=6.5 Hz, 2H), 2.23 (t, J=7.3 Hz, 2H), 1.57 (dp, J=14.7, 7.2 Hz, 4H), 1.39 (dq, J=9.2, 4.5, 3.5 Hz, 2H). Example 1-1-2: 7-(5-Carboxypentyl)amino-3-(benzimidazol-2-yl)coumarin 3-(Benzimidazol-2-yl)-7-fluoro-coumarin (FC-2, 0.28 g, 1 mmol, 1 eqv) and 6-aminohexanoic acid (AC-05, 0.13 g, 1 mmol, 1 eqv) was added to anhydrous dimethyl sulfoxide (DMSO, 2 mL). The resulting mixture was stirred for a few minutes at room temperature and then DIPEA (0.25 g, 2 mmol, 2 eqv) was added. The reaction mixture was stirred for 4 hours at temperature 130° C. Additional portions of 6-aminohexanoic acid (AC-1, 0.13 g, 1 mmol, 1 eqv) and DIPEA (0.26 g, 2 mmol, 2 eqv) was added to the reaction mixture and heating was continued at 130° C. was continued for 5 hours. After standing at room temperature for 1 hour, the pale-yellow reaction mixture was diluted with water (5 mL) and stirred overnight. The resulting precipitate was collected by suction filtration. Yield 0.26 g (68.5%). Purity, structure and composition of the product were confirmed by HPLC, NMR and LCMS. MS (DUIS): MW Calculated 391.15. Found m/z: (+) 392 (M+1)+; (−) 390 (M−1)−, 781 (2M−1)−. Example 1-1-3: 7-(2-Carboxyethyl)amino-3-(benzothiazol-2-yl)coumarin Step A: 7-[2-(t-Butyloxycarbonyl)ethyl]amino-3-(benzothiazol-2-yl)coumarin 3-(Benzothiazol-2-yl)-7-fluoro-coumarin (FC-1, 0.3 g, 1.01 mmol, 1 eqv) and t-butyl 3-aminopropionate hydrochloride (AC-C2, 0.2 g, 1.1 mmol, 1.09 eqv) was added to anhydrous dimethyl sulfoxide (DMSO, 2 mL) and the resulting mixture was stirred for a few minutes at room temperature and then DIPEA (0.26 g, 2 mmol, 2 eqv) was added. The resulting mixture was stirred for 2 hours at 100° C. After standing at room temperature for 1 hour, the yellow reaction mixture was diluted with water (7 mL) and was stirred overnight. The resulting precipitate was collected by suction filtration. Yield 0.38 g (69%). MS (DUIS): MW Calculated 422.13. Found m/z: (+) 423 (M+1)+; (−), 421 (M−1)−.1H NMR (400 MHz, DMSO-d6) δ: 9.28 (s, 1H), 9.01 (s, 1H), 8.27-8.16 (m, 1H), 8.10 (tt, J=8.3, 0.9 Hz, 2H), 8.05-7.92 (m, 1H), 7.72 (d, J=8.8 Hz, 1H), 7.66-7.55 (m, 1H), 7.51 (dddd, J=11.4, 8.2, 7.1, 1.3 Hz, 2H), 7.46-7.32 (m, 2H), 6.74 (dd, J=8.7, 2.1 Hz, 1H), 6.58 (d, J=2.1 Hz, 1H), 3.41 (q, J=6.3 Hz, 2H), 2.55 (t, J=6.4 Hz, 2H), 1.41 (s, 9H). Step B A solution of 7-[2-(t-butyloxycarbonyl)ethyl]amino-3-(benzothiazol-2-yl)coumarin (I-1-3tBu, 0.2 g, 0.473 mmol) in anhydrous dichloromethane (20 mL) was treated with trifluoroacetic acid (0.5 mL) and the resulting mixture was stirred for 24 hours at room temperature. The solvents were distilled off and the residue was triturated with water (10 mL). The resulting precipitate was collected by suction filtration. Yield 0.15 g (86%). Purity, structure and composition were confirmed by HPLC, NMR and LCMS. MS (DUIS): MW Calculated 366.39. Found m/z: (+) 367 (M+1)+; (−), 365 (M−1)−. Example 1-1-4: 7-(3-Carboxypropyl)amino-3-(benzothiazol-2-yl)coumarin Step A: 7-[3-(t-Butyloxycarbonyl)propyl]amino-3-(benzothiazol-2-yl)coumarin 3-(Benzothiazol-2-yl)-7-fluoro-coumarin (FC-1, 0.6 g, 2.02 mmol, 1 eqv) and t-butyl 4-aminobutanoate hydrochloride (AC-C3, 0.5 g, 2.56 mmol, 1.27 eqv) were added to anhydrous dimethyl sulfoxide (DMSO, 5 mL). After the addition was complete, the mixture was stirred for a few minutes at room temperature and then DIPEA (0.65 g, 5 mmol, 4 eqv) was added. The reaction mixture was stirred for 3 hours at temperature 100° C. After standing at room temperature for 1 hour, the yellow semi-solid reaction mixture was diluted with water (10 mL) and was left stirring overnight. The resulting precipitate was collected by suction filtration. Yield 0.7 g (79%). Purity, structure and composition were confirmed by HPLC, NMR and LCMS. MS (DUIS): MW Calculated 436.53. Found m/z: (+) 437 (M+1)+; (−), 435 (M−1)−. Step B A solution of 7-[3-(t-Butyloxycarbonyl)propyl]amino-3-(benzothiazol-2-yl)coumarin (I-1-4tBu, 0.7 g, 1.604 mmol) in anhydrous dichloromethane (25 mL) was treated with trifluoroacetic acid (1 mL) and the reaction mixture was stirred for 24 hours at room temperature. The solvents were distilled off and the residue was triturated with water (10 mL). The resulting precipitate was collected by suction filtration. Yield 0.59 g (97%). Purity, structure and composition were confirmed by HPLC, NMR and LCMS. MS (DUIS): MW Calculated 366.39. Found m/z: (+) 381 (M+1)+; (−), 379 (M−1)−.1H NMR (400 MHz, DMSO-d6) δ: 12.17 (s, 1H), 9.01 (s, 1H), 8.12 (d, J=8.0 Hz, 1H), 7.99 (d, J=8.1 Hz, 1H), 7.71 (d, J=8.8 Hz, 1H), 7.48-7.30 (m, 2H), 6.73 (dd, J=8.8, 2.1 Hz, 1H), 6.57 (d, J=2.1 Hz, 1H), 3.21 (q, J=6.6 Hz, 2H), 2.36 (d, J=7.3 Hz, 2H), 1.80 (p, J=7.3 Hz, 2H). Example 1-1-5: 7-(5-Carboxypentyl)amino-3-(5-chloro-benzoxazol-2-yl)coumarin 3-(5-Chloro-benzoxazol-2-yl)-7-fluoro-coumarin (FC-3, 0.32 g, 1 mmol, 1 eqv) and 6-aminohexanoic acid (AC-05, 0.26 g, 2 mmol, 2 eqv) were added to anhydrous dimethyl sulfoxide (DMSO, 5 mL) in round bottomed flask. After the addition was complete, the mixture was stirred for a few minutes at room temperature and then DIPEA (0.52 g, 4 mmol, 2 eqv) was added. The reaction mixture was stirred for 7 hours at temperature 135° C. Additional portions of 6-aminohexanoic acid (AC-1, 0.13 g, 1 mmol, 1 eqv) and DIPEA (0.26 g, 2 mmol, 2 eqv) were added and heating was continued at 135° C. for 5 hours. After standing at room temperature for 1 hour, the pale-yellow reaction mixture was diluted with water (15 mL) and was stirred overnight. The resulting precipitate was collected by suction filtration. Yield 0.09 g (21%). Purity, structure and composition of the product were confirmed by HPLC, NMR and LCMS. MS (DUIS): MW Calculated 426.10. Found m/z: (+) 427 (M+1)+; (−) 425 (M−1)−, 851 (2M−1)−. Example 2: 7-(5-Carboxypentyl)amino-3-(benzothiazol-2-yl)coumarin-6-sulfonic acid (2A) and 7-(5-Carboxypentyl)amino-3-(benzothiazol-2-yl)coumarin-8-sulfonic acid (2B) Compound I-1-1 (0.1 g, 0.245 mmol) was added in small portions with stirring to 20% fuming sulfuric acid (1 mL) that was cooled in a dry-ice/acetone bath. After the addition was complete, the mixture was stirred for 1 hour at 0° C., warmed to room temperature, and then stirred for 2 hours at room temperature. The solution was poured into anhydrous ether (25 mL). After standing at room temperature for 1 hour, the resulting precipitate was collected by suction filtration. Yield 78 mg (65%).1H NMR (d6-DMSO) showed compound 2A plus a small amount (˜4%) of compound 2B. Example 2A, Sodium Salt: The precipitate from above was resuspended in water (2 mL) and the pH of the suspension was adjusted to ˜5 by addition of 5 M NaOH solution. The resulting mixture was poured into 10 mL of methanol and the suspension was filtered. The filtrate was evaporated to dryness to give the dye as sodium salt (I-2A-Na). Purity, structure and composition were confirmed by HPLC, NMR and LCMS. MS (DUIS): MW Calculated 488.07. Found m/z: (+) 489 (M+1)+; (−) 243 (M−1)2−, 487 (M−1)−. Preparation of Triethylammonium Salts of 2A and 2B: Compound I-1-1 (0.41 g, 1 mmol) was added in small portions with stirring to 20% fuming sulfuric acid (5 mL) that was cooled in a dry-ice/acetone bath. After the addition was complete, the mixture was stirred for 1 hour at 0° C., warmed to room temperature, and then stirred for 2 hours at room temperature. The solution was poured into anhydrous ether (50 mL). After standing at room temperature for 1 hour, the organic solvent layer is decanted and the semi-solid bottom layer was dissolved in acetonitrile-water (1:1, 10 mL). The pH of the solution was adjusted to ˜7.0 by addition of 2 M TEAB solution in water. The resulting solution was filtered through a 20 μm Nylon filter and the isomers were separated by preparative HPLC. The solution of the isomers were concentrated in vacuo then re-dissolved in water (20 μL) and solvent removed in vacuo to dryness to give the dyes as triethylammonium salts. Purity and composition were confirmed by HPLC and LCMS. Example 3: 7-(5-Carboxypentyl)amino-3-[5-sulfonato(benzothiazol-2-yl)-coumarin-6-sulfonate triethylammonium salt Compound I-1-1 (0.08 g, 0.2 mmol) was added in small portions with stirring to 20% fuming sulfuric acid (2 mL) that was cooled in a dry-ice/acetone bath. After the addition was complete, the mixture was stirred for 1 hour at 0° C., warmed to room temperature, and then stirred for 2 hours at 70° C. The mixture was then stirred overnight at room temperature. The solution was poured into anhydrous ether (30 mL). After stirring at room temperature for 1 hour, the resulting precipitate was collected by suction filtration. Yield 43 mg (38%). The precipitate was resuspended in water (2 mL) and the pH of the suspension was adjusted to ˜7.5 by addition of 2 M TEAB solution in water. The resulting mixture was filtered through a 20 μm Nylon filter and purified by preparative HPLC. The dye fraction was concentrated in vacuo then re-dissolved in water (20 μL) and solvent removed in vacuo to dryness to give the dye as the bis-triethylammonium salt. Purity and composition were confirmed by HPLC and LCMS. MS (DUIS): MW Calculated 568.03. Found m/z: (+) 569 (M+1)+. Fluorescence intensities of exemplary dye solutions were compared with a commercial dye for the same spectral region. The results are shown in Table 2 and demonstrate significant advantages of the exemplary dyes for fluorescence based analytical applications. TABLE 2Spectral properties of the new fluorescent dyes disclosed in the examplesSpectral propertiesin EtOH-Water 1:1Relative Fluor.*FluorescenceIntensity,NumberStructureAbs. max nmmax nm%I-1-1460499275I-1-2437488175I-1-3453499230I-1-4455500220I-1-5430490200I-2A465503395I-2B466505280I-3472515330StandardAtto465 from AttoTec455508100*Excitation of fluorescence @ 460 nm Example 4: General Procedure for the Synthesis of Fully Functional Nucleotide Conjugates with New Fluorescent Dyes Coumarin fluorescent dyes disclosed herein were coupled with appropriate amino-substituted adenine (A) and cytosine (C) nucleotide derivatives A-LN3-NH2or C-LN3-NH2: after activation of carboxylic group of a dye with appropriate reagents according to the following adenine exemplary scheme: The general product for the adenine coupling is as shown below: ffA-LN3-Dye refers to a fully functionalized A nucleotide with an LN3 linker and labeled with a coumarin dye disclosed herein. The R group in each of the structures refers to the coumarin dye moiety after conjugation. The dye (10 μmol) is dried by placing into a 5 mL round-bottomed flask and is dissolved in anhydrous dimethylformamide (DMF, 1 mL) then the solvent is distilled off in vacuo. This procedure is repeated twice. The dried dye is dissolved in anhydrous N,N-dimethylacetamide (DMA, 0.2 mL) at room temperature. N,N,N′,N′-Tetramethyl-O—(N-succinimidyl)uronium tetrafluoroborate (TSTU, 1.5 eq., 15 μmol, 4.5 mg) is added to the dye solution, then DIPEA (3 eq., 30 μmol, 3.8 mg, 5.2 μL) is added via micropipette to this solution. The reaction flask is sealed under nitrogen gas. The reaction progress is monitored by TLC (eluent Acetonitrile-Water 1:9) and HPLC. Meanwhile, a solution of the appropriate amino-substituted nucleotide derivative (A-LN3-NH2, 20 mM, 1.5 eq, 15 μmol, 0.75 mL) is concentrated in vacuo then re-dissolved in water (20 μL). A solution of the activated dye in DMA is transferred to the flask containing the solution of N-LN3-NH2. More DIPEA (3 eq, 30 μmol, 3.8 mg, 5.2 μL) is added along with triethylamine (1 μL). Progress of coupling is monitored hourly by TLC, HPLC, and LCMS. When the reaction is complete, triethylamine bicarbonate buffer (TEAB, 0.05 M˜3 mL) is added to the reaction mixture via pipette. Initial purification of the fully functionalized nucleotide is carried out by running the quenched reaction mixture through a DEAE-Sephadex® column to remove most of remaining unreacted dye. For example, Sephadex is poured into an empty 25 g Biotage cartridge, solvent system TEAB/MeCN. The solution from the Sephadex column is concentrated in vacuo. The remaining material is re-dissolved in the minimum volume of water and acetonitrile, before filtering through a 20 μm Nylon filter. The filtered solution is purified by preparative-HPLC. The composition of prepared compounds was confirmed by LCMS. TABLE 3Structure and spectral properties of various nucleotideslabeled with new coumarin based dyes disclosed herein.Spectral properties in SREAbsorption,Fluorescence,Relative Fluor.Compd.nmnmIntensity, %ffA-I-1-1448505480ffA-I-1-3454499500ffA-I-2A475510575ffA-Standard465504100 A comparison of fluorescence intensities in solution of nucleotides labeled with dyes disclosed herein with appropriate data for nucleotides labeled with the a commercial dye for the same spectral region (Atto465 from AttoTec GmbH) demonstrate the advantage of the dyes described herein for labeling of biomolecules to use in fluorescence based analytical applications. Example 5: Sequencing Analysis FfA's based on dyes disclosed herein were used in sequencing on an Illumina sequencer using sequencing-by-synthesis chemistry and the images inFIGS.1A,1B, and1Cdemonstrate their utility. Scan temp 22° C., Expose times: blue 500 ms, green 500 ms; SFA flow cell. The numbers in bracket in the charts below indicate the ratio of new blue ffA to greenA. The A, C, G, and T nucleotides were labeled as follows: A: Ex 2A (FIG.1A), Ex I-1-3 (FIG.1B), and Ex I-1-1 (FIG.1C); C: NR440; G: Dark; and T: AF550POPOS0. NR440 (See WO2018/060482) AF550POPOS0 (See WO2017/051201)
86,778
11858924
DETAILED DESCRIPTION Many embodiments are detailed throughout the specification and will be apparent to a reader skilled in the art. The specification is not to be interpreted as being limited to any particular embodiment(s) described herein. I. Definitions With respect to the embodiments disclosed in this specification, the following terms have the meanings set forth below: Reference to “a” or “an” means “one or more.” Throughout, the plural and singular should be treated as interchangeable, other than the indication of number. Unless the context requires otherwise, the words “comprise” or “comprises” or “comprising” are used on the basis and clear understanding that they are to be interpreted inclusively, rather than exclusively, and that Applicants intend each of those words to be so interpreted in construing this patent, including the claims below. The term “halogen” (alone or in combination with another term(s)) means a fluorine radical (which may be depicted as —F), chlorine radical (which may be depicted as —Cl), bromine radical (which may be depicted as —Br), or iodine radical (which may be depicted as —I). The term “hydroxy” (alone or in combination with another term(s)) means —OH. The term “cyano” (alone or in combination with another term(s)) means —CN. The term “oxo” (alone or in combination with another term(s)) means an oxo radical, and may be depicted as ═O. The term “alkyl” (alone or in combination with another term(s)) means a straight- or branched-chain saturated hydrocarbyl substituent (i.e., a substituent containing only carbon and hydrogen). Alkyl typically contains from 1 to about 20 carbon atoms, more typically from 1 to about 12 carbon atoms, even more typically from 1 to about 8 carbon atoms, and still even more typically from 1 to about 6 carbon atoms. Examples of such substituents include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso-amyl, 2,2,-dimethylpropyl, hexyl, heptyl, and octyl. The term “cycloalkyl” (alone or in combination with another term(s)) means a saturated carbocyclyl substituent containing from 3 to about 14 carbon ring atoms, more typically from 3 to about 12 carbon ring atoms, and even more typically from 3 to about 8 carbon ring atoms. A cycloalkyl includes a single carbon ring, which typically contains from 3 to 6 carbon ring atoms. Examples of single-ring cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. The term “cycloalkylalkyl” (alone or in combination with another term(s)) means an alkyl substituted with cycloalkyl. Examples of such substituents include cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, and cyclohexylmethyl. The term “alkoxy” (alone or in combination with another term(s)) means an alkylether substituent, i.e., alkyl-O—. Examples of alkoxy include methoxy (CH3—O—), ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, sec-butoxy, and tert-butoxy. Thus, for example:(i) the term “alkoxyalkyl” (alone or in combination with another term(s)) means alkyl substituted with alkoxy such as “methoxymethyl” which may be depicted as: (ii) the term “cycloalkylalkoxy” (alone or in combination with another term(s)) means alkoxy substituted with cycloalkyl such as “cyclopropylmethoxy” which may be depicted as: (iii) the term “alkoxyalkoxy” (alone or in combination with another term(s)) means alkoxy substituted with another alkoxy such as “methoxyethoxy” which may be depicted as: (iv) the term “alkoxyalkoxyalkyl” (alone or in combination with another term(s)) means alkyl substituted with alkoxyalkoxy such as “methoxyethoxymethyl” which may be depicted as: (v) the term “alkoxyphenyl” (alone or in combination with another term(s)) means phenyl substituted with alkoxy such as “4-methoxyphenyl” which may be depicted as: and(vi) the term “alkoxyphenylalkyl” (alone or in combination with another term(s)) means alkyl substituted with alkoxyphenyl such as “4-methoxyphenylmethyl” which may be depicted as: In some instances, the number of carbon atoms in a substituent (e.g., alkyl, cycloalkyl, etc.) is indicated by the prefix “Cx-y—”, wherein x is the minimum and y is the maximum number of carbon atoms in the substituent. Thus, for example, “C1-6-alkyl” refers to an alkyl substituent containing from 1 to 6 carbon atoms. Illustrating further, C3-6-cycloalkyl refers to a cycloalkyl substituent containing from 3 to 6 carbon ring atoms. The prefix “halo” indicates that the substituent to which the prefix is attached is substituted with one or more independently selected halogen radicals. For example, haloalkyl means an alkyl substituent wherein at least one hydrogen radical is replaced with a halogen radical. Where there are more than one hydrogens replaced with halogens, the halogens may be the identical or different. Examples of haloalkyls include fluoromethyl, difluoromethyl, trifluoromethyl, difluoroethyl, 1,1,1-trifluoroethyl, pentafluoroethyl, difluoropropyl, heptafluoropropyl chloromethyl, dichloromethyl, trichloromethyl, difluorochloromethyl, dichlorofluoromethyl, and dichloropropyl. Similarly, “haloalkoxy” means an alkoxy substituent wherein at least one hydrogen radical is replaced by a halogen radical. Where there are more than one hydrogens replaced with halogens, the halogens may be the identical or different. Examples of haloalkoxy substituents include fluoromethoxy, difluoromethoxy, trifluoromethoxy (also known as “perfluoromethyloxy”), 1,1,1,-trifluoroethoxy, and chloromethoxy. The term “carbonyl” (alone or in combination with another term(s)) means —C(O)—, which also may be depicted as: Thus, for example:(i) the term “alkylcarbonyl” (alone or in combination with another term(s)) means alkyl-C(O)— such as “methylcarbonyl” (i.e., acetyl) which may be depicted as: other alkylcarbonyl substituents such as ethylcarbonyl, propylcarbonyl, butylcarbonyl, pentylcarbonyl, and hexylcarbonyl;(ii) the term “alkylcarbonylalkyl” (alone or in combination with another term(s)) means alkyl substituted with alkylcarbonyl such as “methylcarbonylmethyl” which may be depicted as: (iii) the term “cycloalkylcarbonyl” (alone or in combination with another term(s)) means cycloalkyl-C(O)— such as “cyclopropylcarbonyl” which may be depicted as: and(iv) the term “cycloalkylcarbonylalkyl” (alone or in combination with another term(s)) means alkyl substituted with cycloalkylcarbonyl such as “cyclopropylcarbonylmethyl” which may be depicted as: The term “thio” or “thia” (alone or in combination with another term(s)) means a divalent sulfur atom, which also may be depicted as —S—. The term “sulfinyl” or “sulfoxido” (alone or in combination with another term(s)) means —S(O)—, which also may be depicted as: The term “sulfonyl” (alone or in combination with another term(s)) means —S(O)2—, which also may be depicted as: Thus, for example, “alkylsulfonyl” (alone or in combination with another term(s)) means alkyl-S(O)2—. Examples of alkylsulfonyl substituents include methylsulfonyl, ethylsulfonyl, and propylsulfonyl. Similarly, the term “alkylsulfonylalkyl” (alone or in combination with another term(s)) means alkyl substituted with alkylsulfonyl such as “methylsulfonylmethyl” may be depicted as: The term “alkylcarbonylaminoalkyl” (alone or in combination with another term(s)) means alkyl-C(O)—N(H)-alkyl- such as “methylcarbonylaminomethyl” which also may be depicted as: The term “heterocyclyl” (alone or in combination with another term(s)) means a saturated, partially saturated, or completely unsaturated (i.e., heteroaryl”) ring structure containing a total of 3 to 14 ring atoms. At least one of the ring atoms is a heteroatom (i.e., oxygen, nitrogen, or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, oxygen, nitrogen, and sulfur. Heterocyclyl includes monocyclic saturated, partially unsaturated, and completely unsaturated ring structures having, for example, 3 to 7 members, such as 3 to 6 members, 5 to 7 members such as 5 or 6 members, where at least one member and up to 4 members, particularly 1, 2 or 3 members of the ring are heteroatoms selected from N, O and S, and the remaining ring atoms are carbon atoms, in stable combinations known to those of skill in the art. Examples of monocyclic heterocyclyls include furanyl, dihydrofuranyl, tetrahydrofuranyl, thiophenyl, dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, isopyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, isoimidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, dithiolyl, oxathiolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thiodiazolyl, oxathiazolyl, dioxazolyl, oxathiazolyl, oxathiolyl, oxathiolanyl, pyranyl, dihydropyranyl, pyridinyl, piperidinyl, pyridazinyl, pyrimidinyl, pyrazinyl, piperazinyl, triazinyl, oxazinyl, isoxazinyl, oxazolidinyl, isoxazolidinyl, oxathiazinyl, morpholinyl, azepinyl, oxepinyl, thiepinyl, and diazepinyl. Heterocyclyl further includes bicyclic ring structures fused together (i.e., fused bicyclic) or two rings with only one common atom (i.e., spiro), wherein at least one such ring contains a heteroatom as a ring atom (i.e., nitrogen, oxygen, or sulfur). Examples of heterocyclyls having two ring structures fused together include indolizinyl, pyrindinyl, pyranopyrrolyl, 4H-quinolizinyl, purinyl, naphthyridinyl, pyridopyridinyl, pteridinyl, indolyl, isoindolyl, indoleninyl, isoindazolyl, benzazinyl, phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl, benzopyranyl, benzothiopyranyl, benzoxazolyl, indoxazinyl, anthranilyl, benzodioxolyl, benzodioxanyl, benzoxadiazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, isobenzothienyl, benzothiazolyl, benzothiadiazolyl, benzimidazolyl, benzotriazolyl, benzoxazinyl, benzisoxazinyl, and tetrahydroisoquinolinyl. A substituent is “substitutable” if it comprises at least one carbon or nitrogen atom that is bonded to one or more hydrogen atoms. Thus, for example, hydrogen, halogen, and cyano do not fall within this definition. If a substituent is described as being “substituted”, a non-hydrogen radical is in the place of a hydrogen radical on a carbon or nitrogen of the substituent. Thus, for example, a substituted alkyl substituent is an alkyl substituent wherein at least one non-hydrogen radical is in the place of a hydrogen radical on the alkyl substituent. To illustrate, monofluoroalkyl is alkyl substituted with a fluoro radical, and difluoroalkyl is alkyl substituted with two fluoro radicals. It should be recognized that if there are more than one substitutions on a substituent, each non-hydrogen radical may be identical or different (unless otherwise stated). If a substituent is described as being “optionally substituted”, the substituent may be either (1) not substituted, or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogens on the carbon (to the extent there are any) may separately and/or together be replaced with an independently selected optional substituent. If a nitrogen of a substituent is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogens on the nitrogen (to the extent there are any) may each be replaced with an independently selected optional substituent. If substituents are described as being “independently selected” from a group, each substituent is selected independent of the other. Each substituent therefore may be identical to or different from the other substituent(s). The term “pharmaceutically acceptable” is used adjectivally in this specification to mean that the modified noun is appropriate for use as a pharmaceutical product or as a part of a pharmaceutical product. For example, “pharmaceutically acceptable salts” are salts that are suitable for use in mammals, particularly humans, and include salts with an inorganic base, organic base, inorganic acid, organic acid, or basic or acidic amino acid that are suitable for use in mammals, particularly humans. A “therapeutically effective amount” of a pharmacological agent is an amount that is sufficient to effect beneficial or desired results, including clinical results, and, as such, will depend upon the situation in which it is being administered. Where the pharmacological agent is being administered to treat liver disease, for example, a therapeutically effective amount of the agent is an amount of the agent that is sufficient, either alone or in combination with additional therapies, to provide an anti-liver disease effect in a subject as compared to the response obtained without administration of the agent. The term “preventing” is readily understood by an ordinarily skilled physician and, with respect to treatment of a particular condition, can include is intended to have its normal meaning and includes primary prophylaxis to prevent the development of the condition and secondary prophylaxis whereby the condition has already developed and the patient is temporarily or permanently protected against exacerbation or worsening of the disease or the development of new symptoms associated with the condition. The terms “treating” is readily understood by an ordinarily skilled physician and, with respect to treatment of a particular condition, can include (1) diminishing the extent or cause of the condition being treated, and/or (2) alleviating or ameliorating one or more symptoms associated with that condition. Treatment of liver disease, for example, can include stabilizing (i.e., not worsening), delaying, or slowing the spread or progression of the liver disease; prolonging survival as compared to expected survival if not receiving treatment; and/or otherwise ameliorating or palliating the cancer or the severity of the liver disease, in whole or in part. II. Compounds In one embodiment, the present disclosure provides compounds having the structure of Formula (I): and pharmaceutically acceptable salts thereof, wherein:X1is selected from the group consisting of —S—, —S(O)—, and —S(O)2—;R1is selected from the group consisting of hydrogen, halogen, hydroxy, C1-3-alkyl, and C1-6-alkoxy;R2is selected from the group consisting of:(a) heterocyclyl containing a total of 4 to 10 ring atoms, wherein the heterocyclyl ring: (i) is a saturated, partially saturated, or completely unsaturated monocyclic or fused bicyclic ring, (ii) has one, two, or three nitrogen ring atoms with the remaining ring atoms being carbon, and (iii) is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, cyano, C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, and wherein: (a) the C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen, and (b) the C1-6-alkyl may be further substituted with one or more hydroxy;(b) heterocyclyl containing a total of 5 to 10 ring atoms, wherein the heterocyclyl ring: (i) is a saturated, partially saturated, or completely unsaturated monocyclic or fused bicyclic ring, (ii) has (a) one nitrogen ring atom and one oxygen ring atom with the remaining ring atoms being carbon, or (b) one nitrogen ring atom and one sulfur ring atom with the remaining ring atoms being carbon, and (iii) is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen; and(c) spiro heterocyclyl containing a total of 6 to 11 ring atoms, wherein the spiro heterocyclyl: (i) comprises two saturated rings, (ii) has: (a) one or two nitrogen ring atoms with the remaining ring atoms being carbon, (b) one or two nitrogen ring atoms and one or two oxygen ring atoms with the remaining ring atoms being carbon, or (c) one nitrogen ring atom and one sulfur ring atom with the remaining ring atoms being carbon, and (iii) is optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl;R3is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl;R4is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl;R5is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl; andR6is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl. In some embodiments, the present disclosure provides compounds having the structure of Formula (II): and pharmaceutically acceptable salts thereof, wherein X1, R1, R2, R3, R4, R5, and R6are as defined above for the compounds of Formula (I). In one aspect, X1is —S—. In another aspect, X1is —S(O)—. In another aspect, X1is —S(O)2—. In some embodiments, R1is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl. In one aspect, R1is selected from the group consisting of hydrogen, chloro, fluoro, and methyl. In another aspect, R1is hydrogen. In another aspect, R1is chloro. In another aspect, R1is fluoro. In another aspect, R1is methyl. In some embodiments, R3is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl. In one aspect, R3is selected from the group consisting of hydrogen, chloro, fluoro, and methyl. In another aspect, R3is hydrogen. In another aspect, R3is chloro. In another aspect, R3is fluoro. In another aspect, R3is methyl. In some embodiments, R4is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl. In one aspect, R4is hydrogen. In another aspect, R4is chloro. In another aspect, R4is fluoro. In another aspect, R4is methyl. In some embodiments, R5is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl. In one aspect, R5is selected from the group consisting of hydrogen, chloro, fluoro, and methyl. In another aspect, R5is hydrogen. In another aspect, R5is fluoro. In another aspect, R5is chloro. In another aspect, R5is methyl. In some embodiments, R6is selected from the group consisting of hydrogen, halogen, and C1-3-alkyl. In one aspect, R6is selected from the group consisting of hydrogen, chloro, fluoro, and methyl. In another aspect, R6is hydrogen. In another aspect, R6is chloro. In another aspect, R6is fluoro. In another aspect, R6is methyl. In some embodiments, one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of halogen and C1-3-alkyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro and fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is chloro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In some embodiments, at least two of the R1, R3, R4, R5, and R6substituents are independently selected from the group consisting of halogen and C1-3-alkyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, two of the R1, R3, R4, R5, and R6substituents are independently selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In some embodiments, at least one of the R1, R3, R4, R5, and R6substituents is chloro. In some embodiments, at least one of the R1, R3, R4, R5, and R6substituents is fluoro. In some embodiments, at least one of the R1, R3, R4, R5, and R6substituents is methyl. In some embodiments, the present disclosure provides compounds having the structure of Formula (III-A): and pharmaceutically acceptable salts thereof, wherein R1and R2are as defined in the various embodiments above. In some embodiments, the present disclosure provides compounds having the structure of Formula (III-B): and pharmaceutically acceptable salts thereof, wherein R2and R3are as defined in the various embodiments above. In some embodiments, the present disclosure provides compounds having the structure of Formula (III-C): and pharmaceutically acceptable salts thereof, wherein R2and R4are as defined in the various embodiments above. In some embodiments, the present disclosure provides compounds having the structure of Formula (III-D): and pharmaceutically acceptable salts thereof, wherein R2and R5are as defined in the various embodiments above. In some embodiments, the present disclosure provides compounds having the structure of Formula (III-E): and pharmaceutically acceptable salts thereof, wherein R2and R6are as defined in the various embodiments above. In some embodiments, the present disclosure provides compounds having the structure of Formula (IV): and pharmaceutically acceptable salts thereof, wherein R2is as defined above for the compounds of Formula (I). In some embodiments, the present disclosure provides compounds having the structure of Formula (IV-A): or pharmaceutically acceptable salts thereof, wherein R2is as defined above for the compounds of Formula (I). A. R2is Monocyclic or Fused Bicyclic Heterocyclyl (Nitrogen and Carbon Ring Atoms) In some embodiments, the present disclosure provides compounds having the structure of Formulae (I), (II), (III-A), (III-B), (III-C), (III-D), (III-E), (IV), or (IV-A), and pharmaceutically acceptable salts thereof, wherein R2is heterocyclyl containing a total of 4 to 10 ring atoms, wherein the heterocyclyl ring: (i) is a saturated, partially saturated, or completely unsaturated monocyclic or fused bicyclic ring, (ii) has one, two, or three nitrogen ring atoms with the remaining ring atoms being carbon, and (iii) is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, cyano, C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, and wherein: (a) the C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen, and (b) the C1-6-alkyl may be further substituted with one or more hydroxy. In one aspect, the R2heterocyclyl ring is a saturated monocyclic ring. In another aspect, the R2heterocyclyl ring is a partially saturated monocyclic ring. In another aspect, the R2heterocyclyl ring is a completely unsaturated monocyclic ring. In another aspect, the R2heterocyclyl ring is a saturated fused bicyclic ring. In another aspect, the R2heterocyclyl ring is a partially saturated fused bicyclic ring. In another aspect, the R2heterocyclyl ring is a completely unsaturated fused bicyclic ring. In another aspect, the R2heterocyclyl ring has one nitrogen ring atom with the remaining ring atoms being carbon. In another aspect, the R2heterocyclyl ring has two nitrogen ring atoms with the remaining ring atoms being carbon. In another aspect, the R2heterocyclyl ring has three nitrogen ring atoms with the remaining ring atoms being carbon. In some embodiments, the R2heterocyclyl ring is selected from the group consisting of: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, cyano, C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, and wherein: (a) the C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen, and (b) the C1-6-alkyl may be further substituted with one or more hydroxy. In some embodiments, the R2heterocyclyl ring is selected from the group consisting of: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, cyano, C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, and wherein: (a) the C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen, and (b) the C1-6-alkyl may be further substituted with one or more hydroxy. In some embodiments, the R2heterocyclyl ring is selected from the group consisting wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, cyano, C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, and wherein: (a) the C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, C1-3-alkoxyphenyl-C1-3-alkyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen, and (b) the C1-6-alkyl may be further substituted with one or more hydroxy. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of azetidinyl, pyrrolidinyl, piperidinyl, and morpholinyl, wherein the azetidinyl, pyrrolidinyl, piperidinyl, and morpholinyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, wherein the tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more chloro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more fluoro. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more hydroxy. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more oxo. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more cyano. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkyl, wherein the C1-3-alkyl may be further substituted with one or more substituents independently selected from halogen and hydroxy. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C3-6-cycloalkyl, wherein the C3-6-cycloalkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C3-6-cycloalkyl-C1-3-alkyl, wherein the C3-6-cycloalkyl-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkoxy, wherein the C1-3-alkoxy may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C3-6-cycloalkoxy, wherein the C3-6-cycloalkoxy may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkoxy-C2-3-alkoxy, wherein the C1-3-alkoxy-C2-3-alkoxy may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkylcarbonyl, wherein the C1-3-alkylcarbonyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C3-6-cycloalkylcarbonyl, wherein the C3-6-cycloalkylcarbonyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkyl-carbonylamino-C1-3-alkyl, wherein the C1-3-alkyl-carbonylamino-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, and C1-3-alkoxyphenyl-C1-3-alkyl, wherein the phenyl, tolyl, phenyl-C1-3-alkyl, and C1-3-alkoxyphenyl-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of azetidinyl, pyrrolidinyl, piperidinyl, and morpholinyl, wherein the azetidinyl, pyrrolidinyl, piperidinyl, and morpholinyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, wherein the tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, oxo, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, difluoropropyl, trifluoropropyl, methoxy, ethoxy, isopropoxy, difluoromethoxy, trifluoromethoxy, methoxymethyl, trifluoromethoxymethyl, methylcarbonylaminomethyl, methylsulfonylmethyl, morpholinyl, and tetrahydropyranyl. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, difluoroethyl, difluoropropyl, cyclopropyl, methoxy, trifluoromethoxy, ethoxy, and methoxymethyl. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, and morpholinyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tolyl, C1-3-alkoxyphenyl, phenyl-C1-3-alkyl, and morpholinyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, difluoropropyl, trifluoropropyl, cyclopropyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, methylamidomethyl, methylsulfonylmethyl, and morpholinyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, difluoroethyl, trifluoroethyl, difluoropropyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoromethoxymethyl, methoxymethyl, methylamidomethyl, and morpholinyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylazetidin-1-yl)-quinoline-4-carboxamide (Example 20);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoroazetidin-1-yl)quinoline-4-carboxamide (Example 21);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylazetidin-1-yl)-quinoline-4-carboxamide (Example 22);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoroazetidin-1-yl)-quinoline-4-carboxamide (Example 23);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 24);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 25);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(trifluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 26);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 27);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 28);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(methoxymethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 29);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3R)-3-methoxy-2-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 30);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxamide (Example 31);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxyazetidin-1-yl)quinoline-4-carboxamide (Example 66);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 113);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-morpholinoazetidin-1-yl)-quinoline-4-carboxamide (Example 122);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-((trifluoromethoxy)methyl)-azetidin-1-yl)quinoline-4-carboxamide (Example 130);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methyl-3-(2,2,2-trifluoroethyl)-azetidin-1-yl)quinoline-4-carboxamide (Example 131);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(trifluoromethoxy)azetidin-1-yl)quinoline-4-carboxamide (Example 132);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoroethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 133);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropyl-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 134);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 135);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethoxy)azetidin-1-yl)quinoline-4-carboxamide (Example 136);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 137);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-fluoroazetidin-1-yl)-quinoline-4-carboxamide (Example 138);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoropropyl)azetidin-1-yl)quinoline-4-carboxamide (Example 140);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(3,3,3-trifluoropropyl)azetidin-1-yl)quinoline-4-carboxamide (Example 142);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(trifluoromethyl)-azetidin-1-yl)quinoline-4-carboxamide (Example 143);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoroethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 144);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropylazetidin-1-yl)-quinoline-4-carboxamide (Example 145);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2-fluoroethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 146);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(1,1-difluoroethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 147);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-isopropylazetidin-1-yl)quinoline-4-carboxamide (Example 148);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 151);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethoxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 152);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-hydroxyazetidin-1-yl)-quinoline-4-carboxamide (Example 154);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 157);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 158);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoro-2-methylazetidin-1-yl)quinoline-4-carboxamide (Example 159);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-hydroxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 165);(R)-6-(3-(Acetamidomethyl)-3-methylazetidin-1-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 181);(R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-phenylazetidin-1-yl)-quinoline-4-carboxamide (Example 182);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(p-tolyl)azetidin-1-yl)quinoline-4-carboxamide (Example 183);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(4-fluorophenyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 185);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(m-tolyl)azetidin-1-yl)quinoline-4-carboxamide (Example 186);(R)-6-(3-(4-Chlorobenzyl)azetidin-1-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-quinoline-4-carboxamide (Example 187);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methyl-3-((methylsulfonyl)-methyl)azetidin-1-yl)quinoline-4-carboxamide (Example 188); and pharmaceutically acceptable salts thereof. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylazetidin-1-yl)-quinoline-4-carboxamide (Example 22);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 24);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 27);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 28);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(methoxymethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 29);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3R)-3-methoxy-2-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 30);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxamide (Example 31);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxyazetidin-1-yl)quinoline-4-carboxamide (Example 66);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 113);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 135);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-fluoroazetidin-1-yl)-quinoline-4-carboxamide (Example 138);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoropropyl)azetidin-1-yl)quinoline-4-carboxamide (Example 140);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(trifluoromethyl)-azetidin-1-yl)quinoline-4-carboxamide (Example 143);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(1,1-difluoroethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 147);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 151);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethoxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 152);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 157);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-hydroxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 165); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, C1-6-alkyl, hydroxy-C1-3-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-6-alkyl, hydroxy-C1-3-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, C1-3-alkyl, hydroxy-C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, hydroxy-C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, difluoropropyl, hydroxymethyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, and trifluoromethoxymethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, fluoromethyl, hydroxymethyl, methoxy, and trifluoromethoxy. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoropyrrolidin-1-yl)-quinoline-4-carboxamide (Example 47);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 48);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4S)-3,4-difluoropyrrolidin-1-yl)-quinoline-4-carboxamide (Example 50);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-fluoropyrrolidin-1-yl)-quinoline-4-carboxamide (Example 51);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-fluoropyrrolidin-1-yl)-quinoline-4-carboxamide (Example 52);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 54);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 55);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(trifluoromethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 56);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 57);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 65);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methoxypyrrolidin-1-yl)-quinoline-4-carboxamide (Example 161);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide (Example 167);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide (Example 168);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3-fluoro-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide (Example 198);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-2-cyclopropylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 199);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-2-cyclopropylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 200);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methoxypyrrolidin-1-yl)-quinoline-4-carboxamide (Example 207);N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 211);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(methoxymethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 212);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,4S)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxamide (Example 213);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4R)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxamide (Example 214);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 220);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 221);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3,3-difluoro-4-hydroxy-pyrrolidin-1-yl)quinoline-4-carboxamide (Example 224);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R*,4R*)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 225);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R*,4R*)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 226); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, and C1-3-alkoxy-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, difluoropropyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, and trifluoromethoxymethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, fluoromethyl, methoxy, and trifluoromethoxy. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxopyrrolidin-1-yl)quinoline-4-carboxamide (Example 11);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide (Example 12);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 201);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 202); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-6-alkyl, hydroxy-C1-3-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, and wherein the C1-6-alkyl, hydroxy-C1-3-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkyl-carbonylamino-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-3-alkyl, C3-6-cycloalkyl, cyclopropyl-C1-3-alkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, cyclopropyl-C1-3-alkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, and tetrahydropyranyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, difluoropropyl, cyclopropyl, cyclopropylmethyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and tetrahydropyranyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, difluoropropyl, cyclopropyl, and tetrahydropyranyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-methyl-1H-pyrazol-4-yl)-quinoline-4-carboxamide (Example 129);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-cyclopropyl-1H-pyrazol-4-yl)-quinoline-4-carboxamide (Example 170);(R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,3-dimethyl-1H-pyrazol-4-yl)-quinoline-4-carboxamide (Example 171);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,5-dimethyl-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxamide (Example 172);(R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxamide (Example 173); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl C1-3-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl C1-3-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylsulfonyl-C1-3-alkyl, phenyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-3-alkyl, C3-6-cycloalkyl, cyclopropyl-C1-3-alkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, tetrahydrofuranyl, tetrahydropyranyl, and tetrahydrooxepanyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, cyclopropyl-C1-3-alkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, and tetrahydropyranyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, difluoropropyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and tetrahydropyranyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, difluoropropyl, cyclopropyl, and tetrahydropyranyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-methyl-1H-pyrazol-5-yl)-quinoline-4-carboxamide (Example 174);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)quinoline-4-carboxamide (Example 176);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1H-pyrazol-5-yl)quinoline-4-carboxamide (Example 196);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-isopropyl-1H-pyrazol-5-yl)-quinoline-4-carboxamide (Example 197); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, phenyl, tolyl, and C1-3-alkoxyphenyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, phenyl, tolyl, and C1-3-alkoxyphenyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, C1-3-alkyl, cyclopropyl, cyclopropyl-C1-3-alkyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, cyclopropyl-C1-3-alkyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, cyclopropylmethyl, methoxy, ethoxy, propoxy, isopropoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, and trifluoromethoxymethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, propoxy, isopropoxy, trifluoromethoxy, and trifluoroethoxy. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,4S,5R)-4-hydroxy-3,5-dimethylpiperidin-1-yl)quinoline-4-carboxamide (Example 7);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 8);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(piperidin-1-yl)quinoline-4-carboxamide (Example 32);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-dimethylpiperidin-1-yl)-quinoline-4-carboxamide (Example 33);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 34);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-difluoropiperidin-1-yl)-quinoline-4-carboxamide (Example 35);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoropiperidin-1-yl)-quinoline-4-carboxamide (Example 36);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-(fluoromethyl)-4-methyl-piperidin-1-yl)quinoline-4-carboxamide (Example 37);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-difluoro-3,3-dimethyl-piperidin-1-yl)quinoline-4-carboxamide (Example 38);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-(trifluoromethyl)piperidin-1-yl)-quinoline-4-carboxamide (Example 39);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoropiperidin-1-yl)quinoline-4-carboxamide (Example 40);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 41);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxy-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 42);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-isopropoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 43);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-difluoro-2-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 44);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)piperidin-1-yl)quinoline-4-carboxamide (Example 45);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-ethyl-4-hydroxypiperidin-1-yl)quinoline-4-carboxamide (Example 189);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-hydroxy-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 190);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-ethyl-4-methoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 191);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-hydroxy-4-isopropylpiperidin-1-yl)quinoline-4-carboxamide (Example 192);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4S,5S)-4-hydroxy-3,4,5-trimethylpiperidin-1-yl)quinoline-4-carboxamide (Example 193);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoropiperidin-1-yl)quinoline-4-carboxamide (Example 205);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-4-phenylpiperidin-1-yl)-quinoline-4-carboxamide (Example 223); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-oxopiperidin-1-yl)quinoline-4-carboxamide (Example 230), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, oxo, C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, and phenyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl, C3-6-cycloalkylcarbonyl, and phenyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, oxo, C1-3-alkyl, cyclopropyl, cyclopropyl-C1-3-alkyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, cyclopropyl-C1-3-alkyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, oxo, methyl, ethyl, isopropyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, cyclopropylmethyl, methoxy, ethoxy, propoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, and trifluoromethoxymethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, oxo, methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, and methoxymethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-1-methylpiperidin-4-yl)quinoline-4-carboxamide (Example 195), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, and C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, and C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, propoxy, isopropoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, and trifluoromethoxymethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, isopropoxy, trifluoromethoxy, trifluoroethoxy, and methoxymethyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxopiperidin-1-yl)quinoline-4-carboxamide (Example 14);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-2-oxopiperidin-1-yl)-quinoline-4-carboxamide (Example 15);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 203);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 204); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-6-alkyl, hydroxy-C1-3-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3- alkyloxy, and C1-3-alkoxy-C2-3-alkyloxy-C1-3alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkyloxy, and C1-3-alkoxy-C2-3-alkyloxy-C1-3alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, C1-3-alkyl, hydroxy-C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting fluoro, hydroxy, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, propoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, and trifluoromethoxymethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting fluoro, hydroxy, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, and methoxymethyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(pyridin-3-yl)quinoline-4-carboxamide (Example 126);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-methylpyridin-3-yl)quinoline-4-carboxamide (Example 128);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(difluoromethyl)pyridin-3-yl)-quinoline-4-carboxamide (Example 169);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methylpyridin-3-yl)quinoline-4-carboxamide (Example 177);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methoxypyridin-3-yl)quinoline-4-carboxamide (Example 178); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-6-alkyl, hydroxy-C1-3-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, and C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, and C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, C1-3-alkyl, hydroxy-C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, propoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, and trifluoromethoxymethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, and methoxymethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,5-dimethyl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-4-carboxamide (Example 123), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, C1-6-alkyl, hydroxy-C1-3-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, and C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkoxy-C2-3-alkoxy, and C1-3-alkoxy-C2-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, C1-3-alkyl, hydroxy-C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkyl, cyclopropyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more fluoro. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, propoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, and trifluoromethoxymethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, hydroxy, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, and methoxymethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,6,7,8-tetrahydroimidazo[1,2-a]pyridin-3-yl)quinoline-4-carboxamide (Example 175) or a pharmaceutically acceptable salt thereof. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,4S,5R)-4-hydroxy-3,5-dimethylpiperidin-1-yl)quinoline-4-carboxamide (Example 7);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 8);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxopyrrolidin-1-yl)quinoline-4-carboxamide (Example 11);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide (Example 12);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxopiperidin-1-yl)quinoline-4-carboxamide (Example 14);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-2-oxopiperidin-1-yl)-quinoline-4-carboxamide (Example 15);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylazetidin-1-yl)-quinoline-4-carboxamide (Example 20);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoroazetidin-1-yl)quinoline-4-carboxamide (Example 21);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylazetidin-1-yl)-quinoline-4-carboxamide (Example 22);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoroazetidin-1-yl)-quinoline-4-carboxamide (Example 23);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 24);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 25);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(trifluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 26);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 27);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 28);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(methoxymethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 29);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3R)-3-methoxy-2-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 30);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxamide (Example 31);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(piperidin-1-yl)quinoline-4-carboxamide (Example 32);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-dimethylpiperidin-1-yl)-quinoline-4-carboxamide (Example 33);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 34);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-difluoropiperidin-1-yl)-quinoline-4-carboxamide (Example 35);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoropiperidin-1-yl)-quinoline-4-carboxamide (Example 36);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-(fluoromethyl)-4-methyl-piperidin-1-yl)quinoline-4-carboxamide (Example 37);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-difluoro-3,3-dimethyl-piperidin-1-yl)quinoline-4-carboxamide (Example 38);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-(trifluoromethyl)piperidin-1-yl)-quinoline-4-carboxamide (Example 39);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoropiperidin-1-yl)quinoline-4-carboxamide (Example 40);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 41);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxy-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 42);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-isopropoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 43);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-difluoro-2-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 44);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)piperidin-1-yl)quinoline-4-carboxamide (Example 45);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoropyrrolidin-1-yl)-quinoline-4-carboxamide (Example 47);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 48);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4S)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxamide (Example 50);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-fluoropyrrolidin-1-yl)-quinoline-4-carboxamide (Example 51);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-fluoropyrrolidin-1-yl)-quinoline-4-carboxamide (Example 52);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(hexahydrocyclopenta[c]pyrrol-2(1H)-yl)quinoline-4-carboxamide (Example 53);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 54);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 55);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(trifluoromethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 56);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 57);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-fluoroazepan-1-yl)quinoline-4-carboxamide (Example 59);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-fluoroazepan-1-yl)quinoline-4-carboxamide (Example 60);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 65);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxyazetidin-1-yl)quinoline-4-carboxamide (Example 66);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 113);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,5,6,7-tetrahydro-1H-indazol-1-yl)quinoline-4-carboxamide (Example 114);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,5,6,7-tetrahydro-2H-indazol-2-yl)quinoline-4-carboxamide (Example 115);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-methyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxamide (Example 116);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6,6-dimethyl-5,6-dihydrocyclopenta[c]pyrazol-2(4H)-yl)quinoline-4-carboxamide (Example 117);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxamide (Example 118);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,6-difluoro-1H-indol-1-yl)-quinoline-4-carboxamide (Example 119);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-fluoro-1H-indol-1-yl)quinoline-4-carboxamide (Example 120);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methyl-1H-pyrrol-1-yl)-quinoline-4-carboxamide (Example 121);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-morpholinoazetidin-1-yl)-quinoline-4-carboxamide (Example 122);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,5-dimethyl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-4-carboxamide (Example 123);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-fluoropyridin-4-yl)quinoline-4-carboxamide (Example 124);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-fluoropyridin-2-yl)quinoline-4-carboxamide (Example 125);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(pyridin-3-yl)quinoline-4-carboxamide (Example 126);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(pyrimidin-5-yl)quinoline-4-carboxamide (Example 127);R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-methylpyridin-3-yl)quinoline-4-carboxamide (Example 128);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-methyl-1H-pyrazol-4-yl)-quinoline-4-carboxamide (Example 129);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-((trifluoromethoxy)methyl)-azetidin-1-yl)quinoline-4-carboxamide (Example 130);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methyl-3-(2,2,2-trifluoroethyl)-azetidin-1-yl)quinoline-4-carboxamide (Example 131);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(trifluoromethoxy)azetidin-1-yl)quinoline-4-carboxamide (Example 132);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoroethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 133);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropyl-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 134);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 135);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethoxy)azetidin-1-yl)-quinoline-4-carboxamide (Example 136);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 137);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-fluoroazetidin-1-yl)-quinoline-4-carboxamide (Example 138);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoropropyl)azetidin-1-yl)quinoline-4-carboxamide (Example 140);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(3,3,3-trifluoropropyl)azetidin-1-yl)quinoline-4-carboxamide (Example 142);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(trifluoromethyl)-azetidin-1-yl)quinoline-4-carboxamide (Example 143);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoroethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 144);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropylazetidin-1-yl)-quinoline-4-carboxamide (Example 145);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2-fluoroethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 146);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(1,1-difluoroethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 147);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-isopropylazetidin-1-yl)quinoline-4-carboxamide (Example 148);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 151);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethoxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 152);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-hydroxyazetidin-1-yl)-quinoline-4-carboxamide (Example 154);6-(6-Azabicyclo[3.2.0]heptan-6-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-quinoline-4-carboxamide (Example 156);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 157);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 158);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoro-2-methylazetidin-1-yl)quinoline-4-carboxamide (Example 159);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6,6-difluoro-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxamide (Example 160);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methoxypyrrolidin-1-yl)-quinoline-4-carboxamide (Example 161);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-hydroxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 165);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide (Example 167);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide (Example 168);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(difluoromethyl)pyridin-3-yl)-quinoline-4-carboxamide (Example 169);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-cyclopropyl-1H-pyrazol-4-yl)-quinoline-4-carboxamide (Example 170);(R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,3-dimethyl-1H-pyrazol-4-yl)-quinoline-4-carboxamide (Example 171);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,5-dimethyl-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxamide (Example 172);(R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxamide (Example 173);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-methyl-1H-pyrazol-5-yl)-quinoline-4-carboxamide (Example 174);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,6,7,8-tetrahydroimidazo[1,2-a]-pyridin-3-yl)quinoline-4-carboxamide (Example 175);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)quinoline-4-carboxamide (Example 176);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methylpyridin-3-yl)quinoline-4-carboxamide (Example 177);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methoxypyridin-3-yl)quinoline-4-carboxamide (Example 178);(R)-6-(3-(Acetamidomethyl)-3-methylazetidin-1-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 181);(R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-phenylazetidin-1-yl)-quinoline-4-carboxamide (Example 182);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(p-tolyl)azetidin-1-yl)quinoline-4-carboxamide (Example 183);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(4-fluorophenyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 185);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(m-tolyl)azetidin-1-yl)quinoline-4-carboxamide (Example 186);(R)-6-(3-(4-Chlorobenzyl)azetidin-1-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-quinoline-4-carboxamide (Example 187);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methyl-3-((methylsulfonyl)-methyl)azetidin-1-yl)quinoline-4-carboxamide (Example 188);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-ethyl-4-hydroxypiperidin-1-yl)-quinoline-4-carboxamide (Example 189);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-hydroxy-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 190);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-ethyl-4-methoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 191);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-hydroxy-4-isopropylpiperidin-1-yl)quinoline-4-carboxamide (Example 192);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4S,5S)-4-hydroxy-3,4,5-trimethylpiperidin-1-yl)quinoline-4-carboxamide (Example 193);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-1-methylpiperidin-4-yl)-quinoline-4-carboxamide (Example 195);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1H-pyrazol-5-yl)quinoline-4-carboxamide (Example 196);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-isopropyl-1H-pyrazol-5-yl)-quinoline-4-carboxamide (Example 197);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3-fluoro-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide (Example 198);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-2-cyclopropylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 199);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-2-cyclopropylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 200);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 201);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 202);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 203);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 204);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoropiperidin-1-yl)quinoline-4-carboxamide (Example 205);6-(3-Azabicyclo[3.1.0]hexan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-quinoline-4-carboxamide (Example 206);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methoxypyrrolidin-1-yl)-quinoline-4-carboxamide (Example 207);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((1R,5S,6R)-6-(trifluoromethyl)-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxamide (Example 208);(R)-6-(7-Azabicyclo[2.2.1]heptan-7-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-quinoline-4-carboxamide (Example 209);6-(2-Azabicyclo[2.2.1]heptan-2-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-quinoline-4-carboxamide (Example 210);N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methylpyrrolidin-1-yl)-quinoline-4-carboxamide (Example 211);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(methoxymethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 212);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,4S)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxamide (Example 213);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4R)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxamide (Example 214);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-phenyl-1H-imidazol-1-yl)-quinoline-4-carboxamide (Example 217);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-phenyl-1H-pyrrol-1-yl)-quinoline-4-carboxamide (Example 218);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,5,6,7-tetrahydro-1H-indol-1-yl)quinoline-4-carboxamide (Example 219);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 220);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 221);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-4-phenylpiperidin-1-yl)-quinoline-4-carboxamide (Example 223);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3,3-difluoro-4-hydroxy-pyrrolidin-1-yl)quinoline-4-carboxamide (Example 224);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R*,4R*)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 225);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R*,4R*)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 226);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-oxopiperidin-1-yl)quinoline-4-carboxamide (Example 230); and pharmaceutically acceptable salts thereof. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,4S,5R)-4-hydroxy-3,5-dimethylpiperidin-1-yl)quinoline-4-carboxamide (Example 7);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 8);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide (Example 12);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide (Example 15);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylazetidin-1-yl)-quinoline-4-carboxamide (Example 22);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 24);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 27);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 28);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(methoxymethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 29);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3R)-3-methoxy-2-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 30);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxamide (Example 31);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(piperidin-1-yl)quinoline-4-carboxamide (Example 32);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 34);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 41);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxy-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 42);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-isopropoxypiperidin-1-yl)-quinoline-4-carboxamide (Example 43);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4S)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxamide (Example 50);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-fluoropyrrolidin-1-yl)-quinoline-4-carboxamide (Example 52);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxyazetidin-1-yl)quinoline-4-carboxamide (Example 66);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)azetidin-1-yl)-quinoline-4-carboxamide (Example 113);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)-3-methyl-azetidin-1-yl)quinoline-4-carboxamide (Example 135);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-fluoroazetidin-1-yl)-quinoline-4-carboxamide (Example 138);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoropropyl)azetidin-1-yl)quinoline-4-carboxamide (Example 140);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(trifluoromethyl)-azetidin-1-yl)quinoline-4-carboxamide (Example 143);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(1,1-difluoroethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 147);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxy-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 151);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethoxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 152);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 157);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methoxypyrrolidin-1-yl)-quinoline-4-carboxamide (Example 161);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-hydroxy-3-methylazetidin-1-yl)-quinoline-4-carboxamide (Example 165);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(difluoromethyl)pyridin-3-yl)-quinoline-4-carboxamide (Example 169);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,5-dimethyl-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxamide (Example 172);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,6,7,8-tetrahydroimidazo[1,2-a]pyridin-3-yl)quinoline-4-carboxamide (Example 175);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-ethyl-4-hydroxypiperidin-1-yl)quinoline-4-carboxamide (Example 189);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-hydroxy-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 190);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-1-methylpiperidin-4-yl)-quinoline-4-carboxamide (Example 195);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3-fluoro-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide (Example 198);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 201);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 202);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide Isomer 1 (Example 203);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide Isomer 2 (Example 204);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoropiperidin-1-yl)quinoline-4-carboxamide (Example 205);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methoxypyrrolidin-1-yl)-quinoline-4-carboxamide (Example 207);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,4S)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxamide (Example 213);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxamide (Example 221);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3,3-difluoro-4-hydroxy-pyrrolidin-1-yl)quinoline-4-carboxamide (Example 224); and pharmaceutically acceptable salts thereof. B. R2is Monocyclic or Fused Bicyclic Heterocyclyl (Nitrogen, Oxygen (or Sulfur), and Carbon Ring Atoms) In some embodiments, the present disclosure provides compounds having the structure of Formulae (I), (II), (III-A), (III-B), (III-C), (III-D), (III-E), (IV), or (IV-A), and pharmaceutically acceptable salts thereof, wherein R2is heterocyclyl containing a total of 5 to 10 ring atoms, wherein the heterocyclyl ring: (i) is a saturated, partially saturated, or completely unsaturated monocyclic or fused bicyclic ring, (ii) has (a) one nitrogen ring atom and one oxygen ring atom with the remaining ring atoms being carbon, or (b) one nitrogen ring atom and one sulfur ring atom with the remaining ring atoms being carbon, and (iii) is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is a saturated monocyclic ring. In another aspect, the R2heterocyclyl ring is a partially saturated monocyclic ring. In another aspect, the R2heterocyclyl ring is a completely unsaturated monocyclic ring. In another aspect, the R2heterocyclyl ring is a saturated fused bicyclic ring. In another aspect, the R2heterocyclyl ring is a partially saturated fused bicyclic ring. In another aspect, the R2heterocyclyl ring is a completely unsaturated fused bicyclic ring. In another aspect, the R2heterocyclyl ring has one nitrogen ring atom and one oxygen ring atom with the remaining ring atoms being carbon. In another aspect, the R2heterocyclyl ring has one nitrogen ring atom and one sulfur ring atom with the remaining ring atoms being carbon. In some embodiments, the R2heterocyclyl ring contains a total of 6 to 10 ring atoms. In some embodiments, the R2heterocyclyl ring is selected from the group consisting of: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is selected from the group consisting of: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is selected from the group consisting of: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-5-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, C3-5-cycloalkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-2-alkyl, C3-4-cycloalkyl, C1-2-alkoxy, C1-2-alkoxy-C1-3-alkyl, C1-2-alkylcarbonyl-C1-2-alkyl, and C1-2-alkylsulfonyl-C1-2-alkyl, wherein the C1-2-alkyl, C3-4-cycloalkyl, C1-2-alkoxy, C1-2-alkoxy-C1-2-alkyl, C1-2-alkylcarbonyl-C1-2-alkyl, and C1-2-alkylsulfonyl-C1-2-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more fluoro. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkyl, wherein the C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C3-6-cycloalkyl, wherein the C3-6-cycloalkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkoxy-C1-3-alkyl, wherein the C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, isopropoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, methoxyethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In some embodiments, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, isopropyl, cyclopropyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the R2heterocyclyl ring is: wherein the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-5-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-5-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, cyano, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, methoxyethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, trifluoromethyl, difluoropropyl, methoxy, trifluoromethoxy, and methoxymethyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-3-fluoro-6-morpholinoquinoline-4-carboxamide (Example 2);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-difluoromorpholino)quinoline-4-carboxamide (Example 4);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2,6,6-tetrafluoromorpholino)-quinoline-4-carboxamide (Example 5);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-8-methyl-6-morpholinoquinoline-4-carboxamide (Example 9);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-7-methyl-6-morpholinoquinoline-4-carboxamide (Example 10);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethyl-3-oxomorpholino)-quinoline-4-carboxamide (Example 16);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 67);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 68);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(fluoromethyl)morpholino)-quinoline-4-carboxamide (Example 69);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6R)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 70);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)morpholino)-quinoline-4-carboxamide (Example 71);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methylmorpholino)-quinoline-4-carboxamide (Example 72);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide (Example 73);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide (Example 74);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 78);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methylmorpholino)-quinoline-4-carboxamide (Example 80);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3S)-2,3-dimethyl-morpholino)quinoline-4-carboxamide (Example 81);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3S)-2,3-dimethy-lmorpholino)quinoline-4-carboxamide (Example 82);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3R)-2,3-dimethyl-morpholino)quinoline-4-carboxamide (Example 83);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-(trifluoromethyl)-morpholino)quinoline-4-carboxamide Isomer 1 (Example 84);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-(trifluoromethyl)-morpholino)quinoline-4-carboxamide Isomer 2 (Example 85);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,5R)-2,5-dimethy-lmorpholino)quinoline-4-carboxamide (Example 87);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylmorpholino)-quinoline-4-carboxamide (Example 88);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(methoxymethyl)-morpholino)quinoline-4-carboxamide (Example 89);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5R)-3,5-dimethyl-morpholino)quinoline-4-carboxamide (Example 90);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-((methylsulfonyl)-methyl)morpholino)quinoline-4-carboxamide (Example 92);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(methoxymethyl)-morpholino)quinoline-4-carboxamide (Example 93);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-((methylsulfonyl)-methyl)morpholino)quinoline-4-carboxamide (Example 94);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-(2-methoxyethyl)-morpholino)quinoline-4-carboxamide (Example 95);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3S)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxamide (Example 96);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3R)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxamide (Example 97);7-Bromo-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxamide (Example 100);(R)-5-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 101);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methylmorpholino)-quinoline-4-carboxamide (Example 102);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5S)-3,5-dimethylmorpholino)-quinoline-4-carboxamide (Example 103);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,5R)-3,5-dimethylmorpholino)-quinoline-4-carboxamide (Example 105);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-ethylmorpholino)quinoline-4-carboxamide (Example 106);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 107);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methylmorpholino)-quinoline-4-carboxamide (Example 108);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-2-methyl-6-morpholinoquinoline-4-carboxamide (Example 109);(R)-7-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 179);(R)-8-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 180); and pharmaceutically acceptable salts thereof. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-8-methyl-6-morpholinoquinoline-4-carboxamide (Example 9);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 67);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 68);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(fluoromethyl)morpholino)-quinoline-4-carboxamide (Example 69);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6R)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 70);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)morpholino)-quinoline-4-carboxamide (Example 71);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methylmorpholino)-quinoline-4-carboxamide (Example 72);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide (Example 73);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide (Example 74);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 78);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methylmorpholino)-quinoline-4-carboxamide (Example 80);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3S)-2,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 81);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3S)-2,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 82);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylmorpholino)-quinoline-4-carboxamide (Example 88);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(methoxymethyl)-morpholino)quinoline-4-carboxamide (Example 89);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(methoxymethyl)-morpholino)quinoline-4-carboxamide (Example 93);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3R)-3-(methoxymethyl)-2-methylmorpholino)-quinoline-4-carboxamide (Example 97);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methylmorpholino)-quinoline-4-carboxamide (Example 102);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-ethylmorpholino)quinoline-4-carboxamide (Example 106);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 107); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, isopropyl, cyclopropyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-thiomorpholinoquinoline-4-carboxamide (Example 3), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, isopropyl, cyclopropyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethyl-3-oxomorpholino)quinoline-4-carboxamide (Example 16), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, isopropyl, cyclopropyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6,6-dimethyl-2-oxo-1,3-oxazinan-3-yl)quinoline-4-carboxamide (Example 112), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, isopropyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compound or pharmaceutically acceptable salt is 6-(6-oxa-3-azabicyclo[3.1.1]heptan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 77), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:6-((1S,4S)-2-oxa-5-azabicyclo[2.2.1]heptan-5-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 75);6-((1R,4R)-2-oxa-5-azabicyclo[2.2.1]heptan-5-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 76); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compound or pharmaceutically acceptable salt is 6-(3-oxa-8-azabicyclo[3.2.1]octan-8-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 98), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, isopropyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compound or pharmaceutically acceptable salt is 6-((1R,5S)-9-oxa-3-azabicyclo[3.3.1]nonan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 194), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,2-oxazinan-2-yl)quinoline-4-carboxamide (Example 1), or a pharmaceutically acceptable salt thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxamide (Example 61);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxamide (Example 62);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxamide (Example 63);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxamide (Example 64);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,4-oxazepan-4-yl)quinoline-4-carboxamide (Example 91); and pharmaceutically acceptable salts thereof. In some embodiments, the R2heterocyclyl ring is: wherein the heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, and wherein the C1-6-alkyl, C3-6-cycloalkyl, C1-6-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In one aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl, wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, C1-3-alkylcarbonyl-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl may be further substituted with one or more halogen. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of cyano, fluoro, methyl, ethyl, propyl, isopropyl, cyclopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methoxymethyl, trifluoromethoxymethyl, and methylsulfonylmethyl. In another aspect, the R2heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoropropyl, methoxy, and trifluoromethoxy. In some embodiments, the compound or pharmaceutically acceptable salt is 6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 79), or a pharmaceutically acceptable salt thereof. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,2-oxazinan-2-yl)quinoline-4-carboxamide (Example 1);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-3-fluoro-6-morpholinoquinoline-4-carboxamide (Example 2);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-thiomorpholinoquinoline-4-carboxamide (Example 3);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-difluoromorpholino)quinoline-4-carboxamide (Example 4);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2,6,6-tetrafluoromorpholino)-quinoline-4-carboxamide (Example 5);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,3-dihydro-4H-benzo[b][1,4]oxazin-4-yl)quinoline-4-carboxamide (Example 6);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-8-methyl-6-morpholinoquinoline-4-carboxamide (Example 9);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-7-methyl-6-morpholinoquinoline-4-carboxamide (Example 10);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,5-dimethyl-2-oxooxazolidin-3-yl)quinoline-4-carboxamide (Example 13);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethyl-3-oxomorpholino)-quinoline-4-carboxamide (Example 16);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-7-methyl-1,4-oxazepan-4-yl)-quinoline-4-carboxamide (Example 61);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-7-methyl-1,4-oxazepan-4-yl)-quinoline-4-carboxamide (Example 62);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methyl-1,4-oxazepan-4-yl)-quinoline-4-carboxamide (Example 63);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methyl-1,4-oxazepan-4-yl)-quinoline-4-carboxamide (Example 64);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 67);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 68);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(fluoromethyl)-morpholino)quinoline-4-carboxamide (Example 69);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6R)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 70);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)-morpholino)quinoline-4-carboxamide (Example 71);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methylmorpholino)-quinoline-4-carboxamide (Example 72);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide (Example 73);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide (Example 74);6-((1S,4S)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 75);6-((1R,4R)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 76);6-(6-Oxa-3-azabicyclo[3.1.1]heptan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 77);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 78);6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 79);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methylmorpholino)-quinoline-4-carboxamide (Example 80);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3S)-2,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 81);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3S)-2,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 82);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3R)-2,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 83);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-(trifluoromethyl)-morpholino)quinoline-4-carboxamide Isomer 1 (Example 84);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-(trifluoromethyl)-morpholino)quinoline-4-carboxamide Isomer 2 (Example 85);6-(3-Oxa-9-azabicyclo[3.3.1]nonan-9-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 86);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,5R)-2,5-dimethylmorpholino)-quinoline-4-carboxamide (Example 87);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylmorpholino)-quinoline-4-carboxamide (Example 88);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(methoxymethyl)-morpholino)quinoline-4-carboxamide (Example 89);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5R)-3,5-dimethyl-morpholino)quinoline-4-carboxamide (Example 90);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,4-oxazepan-4-yl)quinoline-4-carboxamide (Example 91);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-((methylsulfonyl)-methyl)morpholino)quinoline-4-carboxamide (Example 92);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(methoxymethyl)-morpholino)quinoline-4-carboxamide (Example 93);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-((methylsulfonyl)-methyl)morpholino)quinoline-4-carboxamide (Example 94);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-(2-methoxyethyl)-morpholino)quinoline-4-carboxamide (Example 95);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3S)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxamide (Example 96);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3R)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxamide (Example 97);6-(3-Oxa-8-azabicyclo[3.2.1]octan-8-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 98);7-Bromo-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5R)-3,5-dimethyl-morpholino)quinoline-4-carboxamide (Example 100);(R)-5-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 101);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methylmorpholino)-quinoline-4-carboxamide (Example 102);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5S)-3,5-dimethylmorpholino)-quinoline-4-carboxamide (Example 103);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,5R)-3,5-dimethylmorpholino)-quinoline-4-carboxamide (Example 105);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-ethylmorpholino)quinoline-4-carboxamide (Example 106);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 107);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methylmorpholino)-quinoline-4-carboxamide (Example 108);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-2-methyl-6-morpholinoquinoline-4-carboxamide (Example 109);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6,6-dimethyl-2-oxo-1,3-oxazinan-3-yl)quinoline-4-carboxamide (Example 112);(R)-7-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 179);(R)-8-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 180);6-((1R,5S)-9-Oxa-3-azabicyclo[3.3.1]nonan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 194);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,4-dimethyloxazol-5-yl)quinoline-4-carboxamide (Example 215);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,5-dimethylisoxazol-4-yl)-quinoline-4-carboxamide (Example 216); and pharmaceutically acceptable salts thereof. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,2-oxazinan-2-yl)quinoline-4-carboxamide (Example 1);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-3-fluoro-6-morpholinoquinoline-4-carboxamide (Example 2);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-difluoromorpholino)quinoline-4-carboxamide (Example 4);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-8-methyl-6-morpholinoquinoline-4-carboxamide (Example 9);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 67);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 68);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(fluoromethyl)morpholino)-quinoline-4-carboxamide (Example 69);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6R)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 70);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)morpholino)-quinoline-4-carboxamide (Example 71);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methylmorpholino)-quinoline-4-carboxamide (Example 72);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide (Example 73);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide (Example 74);6-((1S,4S)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 75);6-((1R,4R)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 76);6-(6-Oxa-3-azabicyclo[3.1.1]heptan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 77);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 78);6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 79);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methylmorpholino)-quinoline-4-carboxamide (Example 80);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3S)-2,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 81);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3S)-2,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 82);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylmorpholino)-quinoline-4-carboxamide (Example 88);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(methoxymethyl)-morpholino)quinoline-4-carboxamide (Example 89);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(methoxymethyl)-morpholino)quinoline-4-carboxamide (Example 93);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3R)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxamide (Example 97);6-(3-Oxa-8-azabicyclo[3.2.1]octan-8-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 98);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methylmorpholino)-quinoline-4-carboxamide (Example 102);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-ethylmorpholino)quinoline-4-carboxamide (Example 106);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylmorpholino)-quinoline-4-carboxamide (Example 107);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-2-methyl-6-morpholinoquinoline-4-carboxamide (Example 109);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6,6-dimethyl-2-oxo-1,3-oxazinan-3-yl)quinoline-4-carboxamide (Example 112); and pharmaceutically acceptable salts thereof. C. R2is Spiro Heterocyclyl In some embodiments, the present disclosure provides compounds having the structure of Formulae (I), (II), (III-A), (III-B), (III-C), (III-D), (III-E), (IV), or (IV-A), or pharmaceutically acceptable salts thereof, wherein R2is spiro heterocyclyl containing a total of 6 to 11 ring atoms, wherein the spiro heterocyclyl: (i) comprises two saturated rings, (ii) has: (a) one or two nitrogen ring atoms with the remaining ring atoms being carbon, (b) one or two nitrogen ring atoms and one or two oxygen ring atoms with the remaining ring atoms being carbon, or (c) one nitrogen ring atom and one sulfur ring atom with the remaining ring atoms being carbon, and (iii) is optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl has: (a) one or two nitrogen ring atoms with the remaining ring atoms being carbon, or (b) one or two nitrogen ring atoms and one or two oxygen ring atoms with the remaining ring atoms being carbon. In another aspect, the R2spiro heterocyclyl has one or two nitrogen ring atoms and, optionally, one or two oxygen ring atoms with the remaining ring atoms being carbon. In another aspect, the R2spiro heterocyclyl has one nitrogen ring atom with the remaining ring atoms being carbon. In another aspect, the R2spiro heterocyclyl has two nitrogen ring atoms with the remaining ring atoms being carbon. In another aspect, the R2spiro heterocyclyl has one nitrogen ring atom and one oxygen ring atom with the remaining ring atoms being carbon. In another aspect, the R2spiro heterocyclyl has one nitrogen ring atom and two oxygen ring atoms with the remaining ring atoms being carbon. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are selected from the group consisting of: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are selected from the group consisting of: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are selected from the group consisting of: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are selected from the group consisting of: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more halogen. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more fluoro. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more C1-3-alkyl. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more C1-3-haloalkyl. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more C1-3-alkoxy. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more C1-3-haloalkoxy. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more C1-3-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more methylcarbonyl. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, ethylcarbonyl, and isopropylcarbonyl. In some embodiments, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, and trifluoroethoxy. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-azaspiro[3.3]heptan-2-yl)-quinoline-4-carboxamide (Example 17);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-fluoro-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide (Example 155);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(trifluoromethyl)-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide (Example 150);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methyl-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide (Example 149); and pharmaceutically acceptable salts thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, and methylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, trifluoromethyl, and methylcarbonyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-6-(6-acetyl-2,6-diazaspiro[3.3]heptan-2-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 184), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-thia-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 222), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 18);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 153); and pharmaceutically acceptable salts thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 166), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide (Example 139);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,5-difluoro-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide (Example 141); and pharmaceutically acceptable salts thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,8-dioxa-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide (Example 163), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, and trifluoroethoxy. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(8-oxa-5-azaspiro[3.5]nonan-5-yl)quinoline-4-carboxamide (Example 104), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-oxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxamide (Example 162), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,9-dioxa-4-azaspiro[5.5]undecan-4-yl)quinoline-4-carboxamide (Example 99), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, and trifluoroethoxy. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(7-oxo-6-oxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxamide (Example 111), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, and trifluoroethoxy. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxo-1-oxa-3-azaspiro[5.5]undecan-3-yl)quinoline-4-carboxamide (Example 110), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxa-7-azaspiro[3.5]nonan-7-yl)quinoline-4-carboxamide (Example 229), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,4-dioxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxamide (Example 228), or a pharmaceutically acceptable salt thereof. In some embodiments, the two saturated rings of the R2spiro heterocyclyl are: wherein one or both of the rings are optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-6-alkyl, C1-6-haloalkyl, C1-6-alkoxy, C1-6-haloalkoxy, and C1-6-alkylcarbonyl. In one aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of halogen, C1-3-alkyl, C1-3-haloalkyl, C1-3-alkoxy, C1-3-haloalkoxy, and C1-3-alkylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, trifluoropropyl, methoxy, ethoxy, trifluoromethoxy, trifluoroethoxy, methylcarbonyl, and ethylcarbonyl. In another aspect, the R2spiro heterocyclyl ring is optionally substituted with one or more substituents independently selected from the group consisting of fluoro, methyl, ethyl, fluoromethyl, difluoromethyl, and trifluoromethyl. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,5-dioxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxamide (Example 227), or a pharmaceutically acceptable salt thereof. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide (Example 17);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 18);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-azaspiro[3.3]heptan-1-yl)quinoline-4-carboxamide (Example 19);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-azaspiro[2.5]octan-5-yl)quinoline-4-carboxamide (Example 46);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxamide (Example 49);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-6-(fluoromethyl)-5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxamide (Example 58);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,9-dioxa-4-azaspiro[5.5]undecan-4-yl)quinoline-4-carboxamide (Example 99);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(8-oxa-5-azaspiro[3.5]nonan-5-yl)quinoline-4-carboxamide (Example 104);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxo-1-oxa-3-azaspiro[5.5]undecan-3-yl)quinoline-4-carboxamide (Example 110);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(7-oxo-6-oxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxamide (Example 111);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide (Example 139);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,5-difluoro-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide (Example 141);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methyl-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide (Example 149);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(trifluoromethyl)-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide (Example 150);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 153);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-fluoro-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide (Example 155);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-oxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxamide (Example 162);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,8-dioxa-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide (Example 163);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-azaspiro[2.3]hexan-5-yl)quinoline-4-carboxamide (Example 164);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 166);(R)-6-(6-Acetyl-2,6-diazaspiro[3.3]heptan-2-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (Example 184);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-thia-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 222);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,5-dioxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxamide (Example 227);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,4-dioxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxamide (Example 228);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxa-7-azaspiro[3.5]nonan-7-yl)quinoline-4-carboxamide (Example 229); and pharmaceutically acceptable salts thereof. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 18);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,9-dioxa-4-azaspiro[5.5]undecan-4-yl)quinoline-4-carboxamide (Example 99);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxo-1-oxa-3-azaspiro[5.5]undecan-3-yl)quinoline-4-carboxamide (Example 110);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(trifluoromethyl)-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide (Example 150);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 153);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,8-dioxa-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide (Example 163);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 166);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-thia-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide (Example 222); and pharmaceutically acceptable salts thereof. D. R2is Azetidinyl In some embodiments, the present disclosure provides compounds having the structure of Formula (V): and pharmaceutically acceptable salts thereof, wherein:R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, chloro, fluoro, and methyl;R20aand R20bare independently selected from the group consisting of hydrogen and C1-3-alkyl;R20cand R20dare independently selected from the group consisting of hydrogen, fluoro, hydroxy, C1-3-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-3-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, phenyl, tolyl, phenyl-C1-3-alkyl, morpholinyl, C1-3-alkylsulfonyl-C1-3-alkyl, and C1-3-alkyl-carbonylamino-C1-3-alkyl; and wherein the C1-3-alkyl, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl, C1-3-alkoxy, C3-6-cycloalkoxy, C1-3-alkoxy-C1-3-alkyl, phenyl, and phenyl-C1-3-alkyl may be further substituted with one or more halogen; andR20eand R20fare independently selected from the group consisting of hydrogen and C1-3-alkyl. In some embodiments of the compounds having the structure of Formula (V), one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, one of the R1, R3, R4, R5, and R6substituents is chloro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen.In some embodiments of the compounds having the structure of Formula (V), R1, R3, R4, R5, and R6are all hydrogen. In some embodiments of the compounds having the structure of Formula (V):R20aand R20bare independently selected from the group consisting of hydrogen and methyl;R20cand R20dare independently selected from the group consisting of hydrogen, fluoro, hydroxy, C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, C1-3-alkoxy-C1-3-alkyl, morpholinyl, C1-3-alkylsulfonyl-C1-3-alkyl, and C1-3-alkyl-carbonylamino-C1-3-alkyl; and wherein the C1-3-alkyl, C3-6-cycloalkyl, C1-3-alkoxy, and C1-3-alkoxy-C1-3-alkyl may be further substituted with one or more halogen; andR20eand R20fare independently selected from the group consisting of hydrogen and methyl. In some embodiments of the compounds having the structure of Formula (V), R20cand R20dare independently selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, C1-3-alkoxy, morpholinyl, and C1-3-alkyl-carbonylamino-C1-3-alkyl; and wherein the C1-3-alkyl and C1-3-alkoxy may be further substituted with one or more halogen. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 27);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxamide (Example 113);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxy-3-methylazetidin-1-yl)quinoline-4-carboxamide (Example 151); and pharmaceutically acceptable salts thereof. E. R2is Morpholinyl In some embodiments, the present disclosure provides compounds having the structure of Formula (VI): and pharmaceutically acceptable salts thereof, wherein:R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, chloro, fluoro, and methyl;R30aand R30bare independently selected from the group consisting of hydrogen, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy-C1-3-alkyl;R30cand R30aare independently selected from the group consisting of hydrogen, halogen, C1-3-alkyl, halo-C1-3-alkyl, C1-3-alkoxy-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl;R30eand R30fare independently selected from the group consisting of hydrogen, halogen, C1-3-alkyl, halo-C1-3-alkyl, C1-3-alkoxy-C1-3-alkyl, and C1-3-alkylsulfonyl-C1-3-alkyl; andR30gand R30hare independently selected from the group consisting of hydrogen, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy-C1-3-alkyl. In some embodiments of the compounds having the structure of Formula (VI), one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, one of the R1, R3, R4, R5, and R6substituents is chloro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In some embodiments of the compounds having the structure of Formula (VI), R1, R3, R4, R5, and R6are all hydrogen. In some embodiments of the compounds having the structure of Formula (VI):R30aand R30bare independently selected from the group consisting of hydrogen, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy-C1-2-alkyl;R30cand R30dare independently selected from the group consisting of hydrogen, halogen, C1-2-alkyl, halo-C1-2-alkyl, C1-2-alkoxy-C1-2-alkyl, and C1-2-alkylsulfonyl-C1-2-alkyl;R30eand R30fare independently selected from the group consisting of hydrogen, halogen, C1-2-alkyl, halo-C1-2-alkyl, C1-2-alkoxy-C1-2-alkyl, and C1-2-alkylsulfonyl-C1-2-alkyl; andR30gand R30hare independently selected from the group consisting of hydrogen, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy-C1-2-alkyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-8-methyl-6-morpholinoquinoline-4-carboxamide (Example 9);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide (Example 67);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6S)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 68);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(fluoromethyl)morpholino)-quinoline-4-carboxamide (Example 69);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6R)-2,6-dimethylmorpholino)-quinoline-4-carboxamide (Example 70);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)morpholino)-quinoline-4-carboxamide (Example 71);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methylmorpholino)-quinoline-4-carboxamide (Example 72);N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methylmorpholino)-quinoline-4-carboxamide (Example 80); and pharmaceutically acceptable salts thereof. F. R2is Piperidin-1-yl In some embodiments, the present disclosure provides compounds having the structure of Formula (VII): and pharmaceutically acceptable salts thereof, wherein:R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, chloro, fluoro, and methyl;R40aand R40bare independently selected from the group consisting of hydrogen, C1-3-alkyl, and halo-C1-3-alkyl;R40cand R40dare independently selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy;R40eand R40fare independently selected from the group consisting of hydrogen, fluoro, hydroxy, oxo, C1-3-alkyl, halo-C1-3-alkyl, cyclopropyl, and C1-3-alkoxy;R40gand R40hare independently selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy; andR40iand R40jare independently selected from the group consisting of hydrogen, C1-3-alkyl, and halo-C1-3-alkyl. In some embodiments of the compounds having the structure of Formula (VII), one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, one of the R1, R3, R4, R5, and R6substituents is chloro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In some embodiments of the compounds having the structure of Formula (VII), R1, R3, R4, R5, and R6are all hydrogen. In some embodiments of the compounds having the structure of Formula (VII):R40aand R40bare independently selected from the group consisting of hydrogen, C1-3-alkyl, and halo-C1-3-alkyl;R40cand R40dare independently selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy;R40eand R40fare independently selected from the group consisting of hydrogen, fluoro, hydroxy, C1-3-alkyl, halo-C1-3-alkyl, cyclopropyl, and C1-3-alkoxy;R40gand R40hare independently selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy; andR40iand R40jare independently selected from the group consisting of hydrogen, C1-3-alkyl, and halo-C1-3-alkyl. In some embodiments of the compounds having the structure of Formula (VII):R40aand R40bare independently selected from the group consisting of hydrogen, C1-2-alkyl, and halo-C1-2-alkyl;R40cand R40dare independently selected from the group consisting of hydrogen, fluoro, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy;R40eand R40fare independently selected from the group consisting of hydrogen, fluoro, hydroxy, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy;R40gand R40hare independently selected from the group consisting of hydrogen, fluoro, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy; andR40iand R40jare independently selected from the group consisting of hydrogen, C1-2-alkyl, and halo-C1-2-alkyl. In some embodiments, the compounds and pharmaceutically acceptable salts are selected from the group consisting of:(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxypiperidin-1-yl)quinoline-4-carboxamide (Example 8);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxy-4-methylpiperidin-1-yl)quinoline-4-carboxamide (Example 42);(R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoropiperidin-1-yl)quinoline-4-carboxamide (Example 205);(R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-oxopiperidin-1-yl)quinoline-4-carboxamide (Example 230) and pharmaceutically acceptable salts thereof. G. R2is Piperidin-4-yl In some embodiments, the present disclosure provides compounds having the structure of Formula (VIII): and pharmaceutically acceptable salts thereof, wherein:R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, chloro, fluoro, and methyl;R50aand R50bare independently selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy;R50cand R50dare independently selected from the group consisting of hydrogen, C1-3-alkyl, and halo-C1-3-alkyl, or together are oxo;R50eis selected from the group consisting of hydrogen, C1-3-alkyl, halo-C1-3-alkyl, C1-3-alkoxy-C2-3-alkyl, C1-3-alkyl-carbonyl, and C3-6-cycloalkyl-carbonyl;R50fand R50gare independently selected from the group consisting of hydrogen, C1-3-alkyl, and halo-C1-3-alkyl, or together are oxo;R50hand R50iare independently selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy; andR50jis selected from the group consisting of hydrogen and fluoro. In some embodiments of the compounds having the structure of Formula (VIII), one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, one of the R1, R3, R4, R5, and R6substituents is chloro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In some embodiments of the compounds having the structure of Formula (VIII), R1, R3, R4, R5, and R6are all hydrogen. In some embodiments of the compounds having the structure of Formula (VIII):R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, fluoro, and methyl;R50aand R50bare independently selected from the group consisting of hydrogen, fluoro, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy;R50cand R50dare independently selected from the group consisting of hydrogen, C1-2-alkyl, and halo-C1-2-alkyl;R50eis selected from the group consisting of hydrogen, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy-C2-3-alkyl;R50fand R50gare independently selected from the group consisting of hydrogen, C1-2-alkyl, and halo-C1-2-alkyl;R50hand R50iare independently selected from the group consisting of hydrogen, fluoro, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy; andR50jis selected from the group consisting of hydrogen and fluoro. In some embodiments of the compounds having the structure of Formula (VIII), at least one of R50a, R50b, R50h, R50i, and R50jis fluoro. H. R2is 3-Oxomorpholinyl In some embodiments, the present disclosure provides compounds having the structure of Formula (IX): and pharmaceutically acceptable salts thereof, wherein:R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, chloro, fluoro, and methyl;R60aand R60bare independently selected from the group consisting of hydrogen and C1-3-alkyl;R60cand R60dare independently selected from the group consisting of hydrogen and C1-3-alkyl; andR60eand R60fare independently selected from the group consisting of hydrogen and C1-3-alkyl. In some embodiments of the compounds having the structure of Formula (IX), one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, one of the R1, R3, R4, R5, and R6substituents is chloro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In some embodiments of the compounds having the structure of Formula (IX), R1, R3, R4, R5, and R6are all hydrogen. In some embodiments of the compounds having the structure of Formula (IX):R4is methyl; andR1, R3, R5, and R6are all hydrogen. In some embodiments of the compounds having the structure of Formula (IX):R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, fluoro, and methyl;R60aand R60bare independently selected from the group consisting of hydrogen and C1-2-alkyl;R60cand R60dare independently selected from the group consisting of hydrogen and C1-2-alkyl; andR60eand R60fare independently selected from the group consisting of hydrogen and C1-2-alkyl. I. R2is 5,8-Dioxa-2-azaspiro[3.4]octan-2-yl In some embodiments, the present disclosure provides compounds having the structure of Formula (X): and pharmaceutically acceptable salts thereof, wherein:R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, chloro, fluoro, and methyl;R70aand R70bare independently selected from the group consisting of hydrogen and C1-3-alkyl;R70cand R70dare independently selected from the group consisting of hydrogen and C1-3-alkyl;R70eand R70fare independently selected from the group consisting of hydrogen and C1-3-alkyl; andR70gand R70hare independently selected from the group consisting of hydrogen and C1-3-alkyl. In some embodiments of the compounds having the structure of Formula (X), one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, one of the R1, R3, R4, R5, and R6substituents is chloro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In some embodiments of the compounds having the structure of Formula (X), R1, R3, R4, R5, and R6are all hydrogen. In some embodiments of the compounds having the structure of Formula (X):R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, fluoro, and methyl;R70aand R70bare independently selected from the group consisting of hydrogen and C1-2-alkyl;R70cand R70dare independently selected from the group consisting of hydrogen and C1-2-alkyl;R70eand R70fare independently selected from the group consisting of hydrogen and C1-2-alkyl; andR70gand R70hare independently selected from the group consisting of hydrogen and C1-2-alkyl. J. R2is Pyridin-3-yl In some embodiments, the present disclosure provides compounds having the structure of Formula (XI): and pharmaceutically acceptable salts thereof, wherein:R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, chloro, fluoro, and methyl;R80ais selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy;R80bis selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy;R80cis selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy; andR80dis selected from the group consisting of hydrogen, fluoro, C1-3-alkyl, halo-C1-3-alkyl, and C1-3-alkoxy. In some embodiments of the compounds having the structure of Formula (XI), one of the R1, R3, R4, R5, and R6substituents is selected from the group consisting of chloro, fluoro, and methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In one aspect, one of the R1, R3, R4, R5, and R6substituents is chloro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is fluoro, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In another aspect, one of the R1, R3, R4, R5, and R6substituents is methyl, and the remaining R1, R3, R4, R5, and R6substituents are all hydrogen. In some embodiments of the compounds having the structure of Formula (XI), R1, R3, R4, R5, and R6are all hydrogen. In some embodiments of the compounds having the structure of Formula (XI):R1, R3, R4, R5, and R6are independently selected from the group consisting of hydrogen, fluoro, and methyl;R80ais selected from the group consisting of hydrogen, fluoro, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy;R80bis selected from the group consisting of hydrogen, fluoro, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy;R80cis selected from the group consisting of hydrogen, fluoro, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy; andR80dis selected from the group consisting of hydrogen, fluoro, C1-2-alkyl, halo-C1-2-alkyl, and C1-2-alkoxy. In some embodiments, the compound or pharmaceutically acceptable salt is (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(difluoromethyl)pyridin-3-yl)quinoline-4-carboxamide (Example 169), or a pharmaceutically acceptable salt thereof. K. Additional Embodiments Any embodiment of the compounds described in the present disclosure can be combined with any other suitable embodiment described herein to provide additional embodiments. For example, where one embodiment individually or collectively describes possible groups for R1, R3, R4, R5, and/or R6and a separate embodiment describes possible groups for R2, it is understood that these embodiments can be combined to provide an additional embodiment describing the possible groups described for R1, R3, R4, R5, and/or R6together with the possible groups described for R2. In other words, for any of the embodiments of the compounds described in the present disclosure, the R2substituent can be as defined in any of the embodiments of R2described below. The compounds of the present disclosure have a pharmaceutically acceptable FAP inhibitory activity measured as described for the hFAP inhibition assay (tight binders) reported in the Examples below. In one aspect, the compounds have an FAP inhibitory activity at IC50concentrations below about 100 nM. In another aspect, the compounds have an FAP inhibitory activity at IC50concentrations below about 50 nM. In another aspect, the compounds have an FAP inhibitory activity at IC50concentrations below about 10 nM. In another aspect, the compounds have an FAP inhibitory activity at IC50concentrations below about 1 nM. In some embodiments, the compounds of the present disclosure possess a pharmaceutically acceptable surface plasmon resonance (SPR) pKdvalue measured as described for the SPR assay reported in the Examples below. In one aspect, the compounds have a surface plasmon resonance (SPR) pKdvalue greater than about 7. In another aspect, the compounds have a surface plasmon resonance (SPR) pKdvalue greater than about 8. In another aspect, the compounds have an SPR pKdvalue greater than about 9. In another aspect, the compounds have an SPR pKdvalue greater than about 10. In some embodiments, the compounds of the present disclosure have a pharmaceutically acceptable selectivity for FAP relative to PREP measured as described for the hFAP inhibition assay (tight binders) and the hPREP inhibition assay reported in the Examples below. In one aspect, the compounds are at least about 50 times more selective for FAP relative to PREP. In another aspect, the compounds are at least about 100 times more selective for FAP relative to PREP. In another aspect, the compounds are at least about 1,000 times more selective for FAP relative to PREP. In another aspect, the compounds are at least about 10,000 times more selective for FAP relative to PREP. In another aspect, the compounds have a PREP IC50value greater than about 0.1 μM. In another aspect, the compounds have a PREP IC50value greater than about 1.0 μM. In another aspect, the compounds have a PREP IC50value greater than about 10.0 μM. In some embodiments, the compounds of the present disclosure have a pharmaceutically acceptable selectivity for FAP relative to DPP7 measured as described for the hFAP inhibition assay (tight binders) and the DPP7 selectivity assay reported in the Examples below. In one aspect, the compounds are at least about 50 times more selective for FAP relative to DPP7. In another aspect, the compounds are at least about 100 times more selective for FAP relative to DPP7. In another aspect, the compounds are at least about 1,000 times more selective for FAP relative to DPP7. In another aspect, the compounds are at least about 10,000 times more selective for FAP relative to DPP7. In another aspect, the compounds have an IC50value for DPP7 that is greater than about 0.1 μM. In another aspect, the compounds have an IC50value for DPP7 that is greater than about 1 μM. In another aspect, the compounds have an IC50value for DPP7 that is greater than about 10 μM. In some embodiments, the compounds of the present disclosure have a pharmaceutically acceptable selectivity for FAP relative to DPP8 and/or DPP9 measured as described for the hFAP inhibition assay (tight binders), DPP8 selectivity assay, and DPP9 selectivity assay reported in the Examples below. In one aspect, the compounds are selective for FAP relative to DPP8. In another aspect, the compounds are selective for FAP relative to DPP9. In another aspect, the compounds are selective for FAP relative to both DPP8 and DPP9. In one aspect, the compounds are at least about 50 times more selective for FAP relative to DPP8 and/or DPP9. In another aspect, the compounds are at least about 100 times more selective for FAP relative to DPP8 and/or DPP9. In another aspect, the compounds are at least about 500 times more selective for FAP relative to DPP8 and/or DPP9. In another aspect, the compounds are at least about 1,000 times more selective for FAP relative to DPP8 and/or DPP9. In another aspect, the compounds have an IC50value for DPP8 and/or DPP9 that is greater than about 0.01 μM. In another aspect, the compounds have an IC50value for DPP8 and/or DPP9 that is greater than about 0.1 μM. In another aspect, the compounds have an IC50value for DPP8 and/or DPP9 that is greater than about 0.4 μM. In some embodiments, the compounds of the present disclosure have a pharmaceutically acceptable metabolic stability measured as described for the human liver microsomes (HLM) assay reported in the Examples below. In one aspect, the compounds have an HLM CLintvalue less than about 300 μL/min/mg. In another aspect, the compounds have an HLM CLintvalue less than about 100 μL/min/mg. In another aspect, the compounds have an HLM CLintvalue less than about 50 μL/min/mg. In some embodiments, the compounds of the present disclosure have a pharmaceutically acceptable metabolic stability measured as described for the rat hepatocytes (rHep) assay reported in the Examples below. In one aspect, the compounds have an rHep CLintvalue less than about 300 μL/min/106cells. In another aspect, the compounds have an rHep CLintvalue less than about 100 μL/min/106cells. In another aspect, the compounds have an rHep CLintvalue less than about 50 μL/min/106cells. In some embodiments, the compounds of the present disclosure have a pharmaceutically acceptable Caco-2 AB intrinsic permeability measured as described for the Caco-2 AB intrinsic permeability assay reported in the Examples below. In one aspect, the compounds have a Caco-2 intrinsic apparent permeability of at least about 0.1×106cm/s. In another aspect, the compounds have a Caco-2 intrinsic apparent permeability of at least about 0.5×106cm/s. In another aspect, the compounds have a Caco-2 intrinsic apparent permeability of at least about 1×106cm/s. In some embodiments, the compounds of the present disclosure have a pharmaceutically acceptable Caco-2 bidirectional (ABBA) A to B apparent permeability measured as described for the Caco-2 bidirectional (ABBA) A to B apparent permeability assay reported in the Examples below. In one aspect, the compounds have a Caco-2 bidirectional (ABBA) A to B apparent permeability of at least about 0.1×106cm/s. In another aspect, the compounds have a Caco-2 bidirectional (ABBA) A to B apparent permeability of at least about 0.25×106cm/s. In another aspect, the compounds have a Caco-2 bidirectional (ABBA) A to B apparent permeability of at least about 0.5×106cm/s. In some embodiments, the compounds of the present disclosure have a pharmaceutically acceptable kinetic solubility measured as described for the kinetic solubility assay reported in the Examples below. In one aspect, the compounds have a kinetic solubility of at least about 1 μM. In another aspect, the compounds have a kinetic solubility of at least about 10 μM. In another aspect, the compounds have a kinetic solubility of at least about 25 μM. In another aspect, the compounds have a kinetic solubility of at least about 50 μM. L. Salts The compounds of the present disclosure may exist in salt form or in non-salt form (i.e., as a free base), and the present disclosure covers both salt forms and non-salt forms. The compounds may form acid addition salts or base addition salts. In general, an acid addition salt can be prepared using various inorganic or organic acids. Such salts can typically be formed by, for example, mixing the compound with an acid (e.g. a stoichiometric amount of an acid) using various methods known in the art. This mixing may occur in water, an organic solvent (e.g. ether, ethyl acetate, ethanol, methanol, isopropanol, or acetonitrile), or an aqueous/organic mixture. In another aspect, the acid addition salts are, for example, trifluoroacetate, formate, acetate or hydrochloric. In general, a base addition salt can be prepared using various inorganic or organic bases, for example an alkali or alkaline earth metal salt such as a sodium, calcium or magnesium salt, or other metal salts, such as potassium or zinc, or an ammonium salt, or a salt with an organic base such as methylamine, dimethylamine, trimethylamine, piperidine or morpholine. The skilled person will be aware of the general principles and techniques of preparing pharmaceutical salts, such as those described in, for exampleJ Pharm Sci.1977 66, 1. Examples of pharmaceutically acceptable salts are also described in “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002). M. Isomers The compounds and salts of the present disclosure may exist in one or more geometrical, optical, enantiomeric, and diastereomeric forms, including, but not limited to, cis- and trans-forms, E- and Z-forms, and R-, S- and meso-forms. Unless otherwise stated a reference to a particular compound includes all such isomeric forms, including racemic and other mixtures thereof. Where appropriate such isomers can be separated from their mixtures by the application or adaptation of known methods (e.g. chromatographic techniques and recrystallisation techniques). Where appropriate such isomers can be prepared by the application or adaptation of known methods. In some embodiments, a single stereoisomer is obtained by isolating it from a mixture of isomers (e.g., a racemate) using, for example, chiral chromatographic separation. In other embodiments, a single stereoisomer is obtained through direct synthesis from, for example, a chiral starting material. A particular enantiomer of a compound described herein may be more active than other enantiomers of the same compound. In one embodiment, the compound, or a pharmaceutically acceptable salt thereof, is a single enantiomer being in an enantiomeric excess (% ee) of ≥90, ≥95%, ≥96%, ≥97, ≥98% or ≥99%. In one aspect, the single enantiomer is present in an enantiomeric excess (% ee) of ≥99%. In another embodiment, the present disclosure relates to a pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt thereof, which is a single enantiomer being in an enantiomeric excess (% ee) of ≥90, ≥95%, ≥96%, ≥97, ≥98% or ≥99%, or a pharmaceutically acceptable salt thereof, in association with one or more pharmaceutically acceptable excipients. In one aspect, the single enantiomer is present in an enantiomeric excess (% ee) of ≥99%. N. Additional Forms The compounds and salts of the present disclosure may exist in various tautomeric forms and the specification encompasses all such tautomeric forms. “Tautomers” are structural isomers that exist in equilibrium resulting from the migration of a hydrogen atom. The compounds of the present disclosure, and pharmaceutically acceptable salts thereof, may exist as solvates (such as a hydrates) as well as unsolvated forms, and the present specification covers all such solvates. The compounds of the present disclosure, and pharmaceutically acceptable salts thereof, may exist in crystalline or amorphous form, and the present specification covers all such forms. Compounds and salts of the present disclosure may be isotopically-labeled (or “radio-labeled”). In that instance, one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature. The specification encompasses isotopically-labelled forms of compounds disclosed herein. Examples of isotopes that may be incorporated include2H (also written as “D” for deuterium),3H (also written as “T” for tritium),11C,13C,14C,13N,15N,15O,17,18and36Cl. The isotope that is used will depend on the specific application of that radio-labeled derivative. For example, for in vitro receptor labeling and competition assays,3H or14C are often useful. For radio-imaging applications,11C is often useful. In some embodiments, the radionuclide is3H. In some embodiments, the radionuclide is14C. In some embodiments, the radionuclide is11C. O. Intermediates In some embodiments, the present disclosure provides additional compounds that are useful as intermediates for preparing the compounds of the present disclosure, and pharmaceutically acceptable salts thereof. III. Methods of Use The disclosed compounds of the present disclosure, and pharmaceutically acceptable salts thereof, are inhibitors of Prolyl endopeptidase fibroblast activation protein (FAP) activity. FAP is an endopeptidase that enzymatically cleaves substrates involved in glucose and lipid metabolism, fibrinolysis, and collagen production. FAP is believed to cleave and inactivate human Fibroblast Growth Factor 21 (FGF-21) (Biochem J2016, 473, 605), a protein involved in the regulation of glucose and lipid metabolism. It is hypothesized that inhibition of FAP increases endogenous FGF-21 levels and signaling, and results, for example, in decreased steatosis, improved insulin sensitivity, improved glucose tolerance, reduced body weight, and/or reduced cardiovascular disease mortality. FAP is also believed to cleave human α2-Antiplasmin (α2AP) (Blood2004 103, 3783), a protein involved in the regulation of fibrosis and fibrinolysis. Tissue repair involves coagulation which results in fibrin deposition. The fibrin of a clot is usually lysed, primarily by plasmin when converted from its inactive form (plasminogen) by plasminogen activators. Fibrinolysis is inhibited by Plasminogen Activator Inhibitor-1 (PAI-1), Plasminogen Activator Inhibitor-2 (PAI-2), and α2AP, (Experimental&Molecular Medicine2020, 52, 367) all of which are induced by tissue trauma. FAP converts α2AP into a more active form that decreases plasmin activity and increases fibrin deposition at the site of an injury. It is hypothesized that inhibition of FAP increases fibrinolysis and improves tissue regeneration at the site of injury (J Thromb Haemost2013, 11, 2029; Proteomics Clin. Appl.2014, 8, 454). FAP is further believed to promote collagen production and deposition and to play a role in increased fibrosis through altered extracellular matrix (ECM) turnover (J Biol Chem2016, 8, 291). It is hypothesized that inhibition of FAP results in a decrease in collagen deposition and a reduction in inflammation (Inflamm Bowel Dis.2018, 18, 332). In view of the above, it is hypothesized that inhibition of FAP collectively reduces fibrosis and inflammation by decreasing hepatic stellate cell activity and increasing fibrinolysis, and further provides positive metabolic effects through increased FGF21 signaling and improved glucose tolerance. In some embodiments, therefore, the present disclosure provides a method for treating or preventing an FAP-mediated condition in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides a method for treating or preventing a condition characterized by overexpression of FAP in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides a method for treating or preventing liver disease in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the liver disease is a fatty liver disease. In another aspect, the liver disease is Nonalcoholic Fatty Liver Disease (NAFLD). In another aspect, the NAFLD is selected from the group consisting of isolated steatosis, Nonalcoholic Steatohepatitis (NASH), liver fibrosis, and cirrhosis. In another aspect, the liver disease is end stage liver disease. In another aspect, the subject is also suffering from or susceptible to one or more conditions selected from the group consisting of obesity, dyslipidemia, insulin resistance, Type 2 diabetes, and renal insufficiency. In some embodiments, the present disclosure provides a method for treating liver disease in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, wherein the subject has a body mass index (BMI of 27 kg/m2to 40 kg/m2. In one aspect, the subject has a BMI of 30 kg/m2to 39.9 kg/m2. In another aspect, the subject has a BMI of at least 40 kg/m2. In another aspect, the subject is overweight. In another aspect, the subject is obese. In another aspect, the liver disease is NAFLD. In another aspect, the liver disease is NASH. In another aspect, the liver disease is liver fibrosis. In another aspect, the liver disease is cirrhosis. In some embodiments, the present disclosure provides a method for treating liver disease in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, wherein the subject is also suffering from or susceptible to dyslipidemia. In another aspect, the liver disease is NAFLD. In another aspect, the liver disease is NASH. In another aspect, the liver disease is liver fibrosis. In another aspect, the liver disease is cirrhosis. In some embodiments, the present disclosure provides a method for treating liver disease in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, wherein the subject is also suffering from or susceptible to insulin resistance. In another aspect, the liver disease is NAFLD. In another aspect, the liver disease is NASH. In another aspect, the liver disease is liver fibrosis. In another aspect, the liver disease is cirrhosis. In some embodiments, the present disclosure provides a method for treating liver disease in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, wherein the subject is also suffering from or susceptible to at least one of Type 2 diabetes and renal insufficiency. In another aspect, the liver disease is NAFLD. In another aspect, the liver disease is NASH. In another aspect, the liver disease is liver fibrosis. In another aspect, the liver disease is cirrhosis. In some embodiments, the present disclosure provides a method for treating liver disease in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, wherein the subject is also suffering from or susceptible to Type 2 diabetes. In another aspect, the liver disease is NAFLD. In another aspect, the liver disease is NASH. In another aspect, the liver disease is liver fibrosis. In another aspect, the liver disease is cirrhosis. In some embodiments, the present disclosure provides a method for treating liver disease in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, wherein the subject is also suffering from or susceptible to renal insufficiency. In another aspect the liver disease is NAFLD. In another aspect the liver disease is NASH. In another aspect, the liver disease is liver fibrosis. In another aspect, the liver disease is cirrhosis. In some embodiments, the present disclosure provides a method for reducing liver fat in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the subject is suffering from or susceptible to NAFLD. In another aspect, the subject is suffering from or susceptible to NASH. In another aspect, the subject is suffering from or susceptible to liver fibrosis. In another aspect, the subject is suffering from or susceptible to cirrhosis. In another aspect, the subject is also suffering from or susceptible to one or more conditions selected from the group consisting of obesity, dyslipidemia, insulin resistance, Type 2 diabetes, and renal insufficiency. In some embodiments, the present disclosure provides a method for treating or preventing Nonalcoholic Fatty Liver Disease (NAFLD) in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the NAFLD is Stage 1 NAFLD. In another aspect, the NAFLD is Stage 2 NAFLD. In another aspect, the NAFLD is Stage 3 NAFLD. In another aspect, the NAFLD is Stage 4 NAFLD. See, e.g., “The Diagnosis and Management of Nonalcoholic Fatty Liver Disease: Practice Guidance From the American Association for the Study of Liver Diseases,”Hepatology,2018, Vol. 67, No. 1. In another aspect, the subject is also suffering from or susceptible to one or more conditions selected from the group consisting of obesity, dyslipidemia, insulin resistance, Type 2 diabetes, and renal insufficiency. In some embodiments, the present disclosure provides a method for treating or preventing Nonalcoholic Steatohepatitis (NASH) in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the NASH is Stage 1 NASH. In another aspect, the NASH is Stage 2 NASH. In another aspect, the NASH is Stage 3 NASH, In another aspect, the NASH is Stage 4 NASH. In another aspect, the subject is also suffering from or susceptible to one or more conditions selected from the group consisting of obesity, dyslipidemia, insulin resistance, Type 2 diabetes, and renal insufficiency. In some embodiments, the present disclosure provides a method for treating or preventing liver fibrosis in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the subject is suffering from Stage 3 liver fibrosis. In another aspect, the subject is also suffering from or susceptible to one or more conditions selected from the group consisting of obesity, dyslipidemia, insulin resistance, Type 2 diabetes, and renal insufficiency. In some embodiments, the present disclosure provides a method for treating or preventing cirrhosis in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the subject is suffering from stage F4 cirrhosis. In another aspect, the subject is also suffering from or susceptible to one or more conditions selected from the group consisting of obesity, dyslipidemia, insulin resistance, Type 2 diabetes, and renal insufficiency. In some embodiments, the present disclosure provides a method for treating or preventing type 2 diabetes mellitus in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the subject is a subject is suffering from diabetic kidney disease. In another aspect, the subject is suffering from renal insufficiency. In another aspect, the administration of the compound is an adjunct to diet and exercise. In another aspect, the administration of the compound also reduces body weight and/or treats obesity. In another aspect, the subject has a BMI of 27 kg/m2to 40 kg/m2. In another aspect, the subject has a BMI of 30 kg/m2to 39.9 kg/m2. In another aspect, the subject has a BMI of at least 40 kg/m2. In another aspect, the subject is overweight. In another aspect, the subject is obese. In some embodiments, the present disclosure provides a method of improving glycemic control in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the subject is a subject is suffering from type 2 diabetes. In another aspect, the subject is a subject is suffering from diabetic kidney disease. In another aspect, the subject is suffering from renal insufficiency. In another aspect, the administration of the compound is an adjunct to diet and exercise. In another aspect, the administration of the compound also reduces body weight and/or treats obesity. In another aspect, the subject has a BMI of 27 kg/m2to 40 kg/m2. In another aspect, the subject has a BMI of 30 kg/m2to 39.9 kg/m2. In another aspect, the subject has a BMI of at least 40 kg/m2. In another aspect, the subject is overweight. In another aspect, the subject is obese. In some embodiments, the present disclosure provides a method of improving glycemic control in a subject with type 2 diabetes and diabetic kidney disease by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable sail thereof. In one aspect, the administration of the compound is an adjunct to diet and exercise. In another aspect, the administration of the compound also reduces body weight and/or treats obesity. In another aspect, the subject has a BMI of 27 kg/m2to 4.0 kg/m2. In another aspect, the subject has aa BMI of 30 kg/m2to 39.9 kg/m2. In another aspect, the subject has a BMI of at least 40 kg/m2. In another aspect, the subject is overweight. In another aspect, the subject is obese. In some embodiments, the present disclosure provides a method of improving glycemic control in a subject with type 2 diabetes and renal insufficiency by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the administration of the compound is an adjunct to diet and exercise. In another aspect, the administration of the compound also reduces body weight and/or treats obesity. In another aspect, the subject has a BMI of 27 kg/m2to 40 kg/m2. In another aspect, the subject has a BMI of 30 kg/m2to 39.9 kg/m2. In another aspect, the subject has a BMI of at least 40 kg/m2. In another aspect, the subject is overweight. In another aspect, the subject is obese. In some embodiments, the present disclosure provides a method of treating or preventing insulin resistance in a subject thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In another aspect, the subject is a subject is suffering from type 2 diabetes. In another aspect, the subject is a subject is suffering from diabetic kidney disease. In another aspect, the subject is suffering from renal insufficiency. Insulin resistance can be measured, for example, using the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) and/or the MATSUDA index. The HOMA-IR is explained, for example, inDiabetologia1985, 28, 412, which is herein incorporated by reference in its entirety. The MATSUDA index is explained, for example, inDiabetes Care1999, 22, 1462, which is herein incorporated by reference in its entirety. In some embodiments, the present disclosure provides a method of treating or preventing glucose intolerance in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the subject is a subject is suffering from type 2 diabetes. In another aspect, the subject is a subject is suffering from diabetic kidney disease. In another aspect, the subject is suffering from renal insufficiency. In some embodiments, the present disclosure provides a method of treating a cardiovascular condition in a subject in need of treatment by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the cardiovascular condition is selected from the group consisting of heart failure, cardiomyopathy, atherosclerosis, venous thromboembolism, and atrial fibrillation. In one aspect, the cardiovascular condition is heart failure. In another aspect, the cardiovascular condition is heart failure with preserved ejection fraction (HFpEF). In another aspect, the cardiovascular condition is cardiomyopathy. In another aspect, the cardiomyopathy is selected from the group consisting of hypertrophic cardiomyopathy, dilated cardiomyopathy, restrictive cardiomyopathy, hypertrophic cardiomyopathy, ischemic cardiomyopathy, ischemic cardiomyopathy, dilated cardiomyopathy, and idiopathic cardiomyopathy. In another aspect, the cardiovascular condition is atherosclerosis. In another aspect, the cardiovascular condition is venous thromboembolism. In another aspect, the cardiovascular condition is atrial fibrillation. In some embodiments, the present disclosure provides a method of treating obesity or an obesity-related condition in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the obesity-related condition is an obesity-related metabolic condition. In another aspect, the obesity-related condition is selected from the group consisting of insulin resistance, pre-diabetes, type 2 diabetes, glucose intolerance, increased fasting glucose, and glucagonomas. In another aspect, the obesity-related condition is dyslipidemia. In another aspect, the obesity-related condition is a cardiovascular condition is selected from the group consisting of heart failure, cardiomyopathy, atherosclerosis, venous thromboembolism, and atrial fibrillation. In another aspect, the obesity-related condition is renal disease. In some embodiments, the present disclosure provides a method of reducing body weight in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the subject is a subject is suffering from type 2 diabetes. In another aspect, the subject is a subject is suffering from diabetic kidney disease. In another aspect, the subject is suffering from renal insufficiency. In another aspect, the administration of the compound is an adjunct to diet and exercise. In another aspect, the administration of the compound also reduces body weight and/or treats obesity. In another aspect, the subject has a BMI of 27 kg/m2to 40 kg/m2. In another aspect, the subject has a BMI of 30 kg/m2to 39.9 kg/m2. In another aspect, the subject has a BMI of at least 40 kg/m2. In another aspect, the subject is overweight. In another aspect, the subject is obese. In another aspect, the subject's weight is reduced, for example, by at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, or 40%. In some embodiments, the present disclosure provides a method of reducing body fat in a subject in need of treatment by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In another aspect, the subject is a subject is suffering from type 2 diabetes. In another aspect, the subject is a subject is suffering from diabetic kidney disease. In another aspect, the subject is suffering from renal insufficiency. In another aspect, the administration of the compound is an adjunct to diet and exercise. In another aspect, the administration of the compound also reduces body weight and/or treats obesity. In another aspect, the subject has a BMI of 27 kg/m2to 40 kg/m2. In another aspect, the subject has a BMI of 30 kg/m2to 39.9 kg/m2. In another aspect, the subject has a BMI of at least 40 kg/m2. In another aspect, the subject is overweight. In another aspect, the subject is obese. In another aspect, the fat is liver fat. In some embodiments, the present disclosure provides a method for treating or preventing fibrosis in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the fibrosis is interstitial lung disease. In another aspect, the fibrosis is interstitial lung disease with progressive fibrosis. In another aspect, the interstitial lung disease is pulmonary fibrosis. In another aspect, the interstitial lung disease is idiopathic pulmonary fibrosis (IPF). In some embodiments, the present disclosure provides a method for promoting tissue remodeling in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the subject has suffered cardiac tissue damage due to a myocardial infarction. In some embodiments, the present disclosure provides a method of promoting wound healing and/or reducing adhesions in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the administration of the compound promotes wound healing and/or reduces adhesions through increased fibrinolysis. In some embodiments, the present disclosure provides a method for treating or preventing a keloid disorder in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the keloid disorder is selected from the group consisting of scar formation, keloid tumors, and keloid scar. In some embodiments, the present disclosure provides a method for treating or preventing inflammation in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the inflammation is chronic inflammation. In one aspect, the chronic inflammation is selected from the group consisting of rheumatoid arthritis, osteoarthritis, and Crohn's disease. In another aspect, the chronic inflammation is rheumatoid arthritis. In some embodiments, the present disclosure provides a method of treating cancer in a subject in need of treatment by administering to the subject a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, the cancer is selected from the group consisting of breast cancer, pancreatic cancer, small intestine cancer, colon cancer, rectal cancer, lung cancer, head and neck cancer, ovarian cancer, hepatocellular carcinoma, esophageal cancer, hypopharynx cancer, nasopharynx cancer, larynx cancer, myeloma cells, bladder cancer, cholangiocellular carcinoma, clear cell renal carcinoma, neuroendocrine tumor, oncogenic osteomalacia, sarcoma, CUP (carcinoma of unknown primary), thymus carcinoma, desmoid tumors, glioma, astrocytoma, cervix carcinoma, and prostate cancer. In another aspect, the cancer is hepatocellular carcinoma. The subject treated typically will be a human or non-human mammal, particularly a human. Suitable subjects can also include domestic or wild animals; companion animals (including dogs, cats, and the like); livestock (including horses, cows and other ruminants, pigs, poultry, rabbits, and the like); primates (including monkeys such as rhesus monkeys, cynomolgus (also known as crab-eating or long-tailed) monkeys, marmosets, tamarins, chimpanzees, macaques, and the like); and rodents (including rats, mice, gerbils, guinea pigs, and the like). In some embodiments, the present disclosure provides the compounds of the present disclosure, or pharmaceutically acceptable salts thereof, for use as medicaments. In some embodiments, the present disclosure provides for the use of the compounds of the Formula I, or pharmaceutically acceptable salts thereof, for treating or preventing an FAP-mediated condition as discussed above. In some embodiments, the present disclosure provides for the use of the compounds of the Formula I, or pharmaceutically acceptable salts thereof, for the manufacture of medicaments for treating or preventing an FAP-mediated condition as discussed above. IV. Combination Therapies and Fixed-Dose Combinations The compounds of the present disclosure may be used in the methods described above as either as single pharmacological agents or in combination with other pharmacological agents or techniques. Such combination therapies may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment. These combination therapies (and corresponding combination products) employ the compounds of the present disclosure within the dosage ranges described in this application and the other pharmacological agent(s), typically within its approved dosage range(s). In some embodiments, the present disclosure provides a combination suitable for use in the treatment of a condition selected from the previously discussed conditions, wherein the combination comprises a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and a sodium-glucose transport protein 2 (SGLT2) inhibitor. In one aspect, the SGLT2 inhibitor is selected from the group consisting of canagliflozin, dapagliflozin, empagliflozin, ertugliflozin, ipragliflozin, luseogliflozin, and remogliflozin. In another aspect, the SGLT2 inhibitor is dapagliflozin. In some embodiments, the present disclosure provides a combination suitable for use in the treatment of a condition selected from the previously discussed conditions, wherein the combination comprises a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and metformin. In some embodiments, the present disclosure provides a combination suitable for use in the treatment of a condition selected from the previously discussed conditions, wherein the combination comprises a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and a glucagon-like peptide-1 receptor (GLP1) agonist. In one aspect, the SGLT2 inhibitor is selected from the group consisting of exenatide, liraglutide, lixisenatide, albiglutide, dulaglutide, and semaglutide. In some embodiments, the present disclosure provides a combination suitable for use in the treatment of a condition selected from the previously discussed conditions, wherein the combination comprises a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and a dipeptidyl peptidase 4 (DPP4) inhibitor. In one aspect, the DPP4 inhibitor is selected from the group consisting of sitagliptin, vildagliptin, saxagliptin, linagliptin, gemigliptin, anagliptin, teneligliptin, alogliptin, trelagliptin, omarigliptin, evogliptin, gosogliptin, and dutogliptin. In some embodiments, the present disclosure provides a combination suitable for use in the treatment of a condition selected from the previously discussed conditions, wherein the combination comprises a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and a peroxisome proliferator-activated receptor (PPAR) agonist. In one aspect, the PPAR agonist is a PPARα agonist. In another aspect, the PPAR agonist is a PPARγ agonist. In another aspect, the PPAR agonist is a PPARα/γ agonist. In another aspect, the PPAR agonist is selected from the group consisting of clofibrate, gemfibrozil, ciprofibrate, bezafibrate, and fenofibrate. In another aspect, the PPAR agonist is a thiazolidinedione. In another aspect, the thiazolidinedione is selected from the group consisting of pioglitazone, rosiglitazone, lobeglitazone, and rivoglitazone. In another aspect, the PPAR agonist stimulates liver expression of FGF21. In some embodiments, the present disclosure provides a pharmaceutical composition comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof; one or more pharmacological agents selected from SGLT2 inhibitors, metformin, GLP1 agonists, DPP4 inhibitors, and PPAR agonists; and a pharmaceutically acceptable diluent or carrier. Such a combination can be used for the manufacture of a medicament for use in the treatment of a condition selected from the previously discussed conditions. In one aspect, the pharmaceutical composition comprises an SGLT2 inhibitor. In another aspect, the pharmaceutical composition comprises metformin. In another aspect, the pharmaceutical composition comprises a GLP1 agonist. In another aspect, the pharmaceutical composition comprises a DPP4 inhibitor. In another aspect, the pharmaceutical composition comprises a PPAR agonist. In some embodiments, the present disclosure provides a combination suitable for use in the treatment of cancer, wherein the combination comprises a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and an immune checkpoint inhibitor. In one aspect, the immune checkpoint inhibitor is selected from the group consisting of anti-PD-1 antibodies, anti-PD-L1 antibodies, anti-CTLA4 antibodies, TLR7 agonists, CD40 agonists, Lag-3 antagonists, and OX40 agonists. In another aspect, the immune checkpoint inhibitor is an anti-PD-1 antibody (e.g., pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo), etc.). In another aspect, the immune checkpoint inhibitor is an anti-PD-L1 antibody (e.g., atezolizumab (Tecentriq), avelumab (Bavencio), durvalumab (Imfinzi), etc.). In another aspect, the immune checkpoint inhibitor is an anti-CTLA4 antibody (e.g., ipilimumab (Yervoy), tremelimumab, etc.). In another aspect, the cancer is selected from the group consisting of pancreatic cancer, colon cancer, and rectal cancer. V. Pharmaceutical Compositions The compounds of the present disclosure, and pharmaceutically acceptable salts thereof, may be administered as pharmaceutical compositions, comprising one or more pharmaceutically acceptable excipients. Therefore, in some embodiments the present disclosure provides pharmaceutical compositions comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient. The excipient(s) selected for inclusion in a particular composition will depend on factors such as the mode of administration and the form of the composition provided. Suitable pharmaceutically acceptable excipients are well known to persons skilled in the art and are described, for example, in the Handbook of Pharmaceutical Excipients, Sixth Edition, Pharmaceutical Press, edited by Rowe, Ray C; Sheskey, Paul J; Quinn, Marian. Pharmaceutically acceptable excipients may function as, for example, adjuvants, diluents, carriers, stabilisers, flavourings, colorants, fillers, binders, disintegrants, lubricants, glidants, thickening agents and coating agents. As persons skilled in the art will appreciate, certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the composition and what other excipients are present in the composition. The compositions may be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder) or for parenteral administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous or intramuscular dosing), or as a suppository for rectal dosing. The compositions may be obtained by conventional procedures using conventional pharmaceutical excipients, well known in the art. Thus, compositions intended for oral use may contain, for example, one or more coloring, sweetening, flavoring and/or preservative agents. The total daily dose will necessarily be varied depending upon the subject treated, the particular route of administration, any therapies being co-administered, and the severity of the illness being treated, and may include single or multiple doses. Specific dosages can be adjusted, for example, depending upon the condition being treated; the age, body weight, general health condition, sex, and diet of the subject; administration routes; dose intervals; excretion rate; and other drugs being co-administered to the subject. An ordinarily skilled physician provided with the disclosure of the present application will be able to determine appropriate dosages and regimens for administration of the therapeutic agent to the subject, and to adjust such dosages and regimens as necessary during the course of treatment, in accordance with methods well-known in the therapeutic arts. The compound of the present disclosure, or a pharmaceutically acceptable salt thereof, typically will be administered to a warm-blooded animal at a unit dose within the range 2.5 to 5000 mg/m2body area of the animal, or approximately 0.05 to 100 mg/kg, and this normally provides a therapeutically-effective dose. A unit dose form such as a tablet or capsule can contain, for example, 0.1 to 500 mg, 0.1 to 250 mg, 0.1 to 100 mg, of active ingredient. In some embodiments, the present disclosure provides pharmaceutical compositions for use in therapy, comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient. In some embodiments, the present disclosure provides pharmaceutical compositions for use in the treatment of an FAP-mediated condition, comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient. In one aspect, the FAP-mediated condition is selected from the group consisting of liver disease, type 2 diabetes mellitus, cardiovascular conditions, obesity, obesity-related conditions, fibrosis, keloid disorder, inflammation, and cancer. VI. Kits The present disclosure further provides kits comprising a unit dosage form comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, contained within a packaging material and a label or package insert which indicates that the unit dosage form can be used for treating one or more of the previously described conditions. In some embodiments, the kit comprises a unit dosage form comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, contained within a packaging material and a label or package insert which indicates that the pharmaceutical composition can be used for treating an FAP-mediated condition. In another aspect, the FAP-mediated condition is liver disease. In another aspect, the liver disease is selected from the group consisting of fatty liver disease, end stage liver disease, and cirrhosis. In another aspect, the liver disease is selected from the group consisting of Nonalcoholic Steatohepatitis (NASH) and Nonalcoholic Fatty Liver Disease (NAFLD). In some embodiments, kit comprises: (a) a first unit dosage form comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof; (b) a second unit dosage form comprising a pharmacological agent selected from the group consisting of SGLT2 inhibitors, metformin, GLP1 agonists, DPP4 inhibitors, and PPAR agonists; (c) a container means for containing said first and second dosage forms; and (d) a label or package insert which indicates that the first unit dosage form and second unit dosage form can be used for treating an FAP-mediated condition. VII. Methods of Preparation The present disclosure further provides processes for the preparation of the compounds of Formulae (I), (II), (III-A), (III-B), (III-C), (III-D), (III-E), (IV), (IV-A), (V), (VI), (VII), (VIII), (IX), (X), and (XI), and pharmaceutically acceptable salts thereof. Schemes 1 to 14 below illustrate synthetic routes to compounds of Formula (II) wherein R1, R2, R3, R4, R5, R6and X1are as defined in formula (I), R7is an alkyl group (e.g., methyl, ethyl, or tert-butyl), and X2, X3and X4are leaving groups (e.g., Cl, Br, I, or OTf). One of skill in the art will appreciate that these methods are representative and are not inclusive of all possible methods for preparing the compounds of the present disclosure. The RX substituents in each Scheme are as defined for the compounds of the present disclosure unless otherwise stated. It is understood that the processes for preparation described in Schemes 1 to 14 can be performed starting from any enantiomer, or a racemic mixture, of compounds of formula (2), (4), (6), (8), (9), (10), (11), (12), (13) or (14), to give compounds of Formula (II) or any stereoisomer of Formula (II). Scheme 1 illustrates synthetic routes to certain compounds of formula (II). A compound of formula (2) may be reacted with a compound of formula (3) to give a compound of formula (II). The reaction may be performed using suitable coupling reagents (e.g., HATU, HOBt/EDC, or T3P) in the presence of a base (typically an organic base such as DIPEA or TEA) using a solvent such as DCM, DMF, EtOAc or MeCN, or mixtures thereof, and at temperatures ranging from typically 0° C. to 60° C. Scheme 2 illustrates additional synthetic routes to certain compounds of formula (II). A compound of formula (4) may be reacted with a compound of formula (5) to give a compound of formula (II). The reaction may be performed using suitable coupling reagents (e.g., HATU, HOBt/EDC, or T3P) in the presence of a base (typically an organic base, such as DIPEA or TEA) using a solvent such as DCM, DMF, EtOAc or MeCN, or mixtures thereof, and at temperatures ranging from typically 0° C. to 60° C. Scheme 3 illustrates additional synthetic routes to certain compounds of formula (II). A compound of formula (6) may be transformed into a compound of formula (II) by dehydration using a suitable reagent (typically TFAA or T3P) in a solvent such as DCM, DMF, EtOAc or MeCN, or mixtures thereof, and at a temperature ranging from typically 0° C. to 120° C. Scheme 4 illustrates synthetic routes to certain compounds of formula (2). A compound of formula (4) may be reacted with (tert-butoxycarbonyl)-glycine (7) to give a compound of formula (8). The reaction may be performed using suitable coupling reagents (e.g., HATU, HOBt/EDC or T3P) in the presence of a base (typically an organic base, such as DIPEA or TEA) using a solvent such as DCM, DMF, EtOAc or MeCN, or mixtures thereof, and at temperatures ranging from typically 0° C. to 120° C. A compound of formula (2) may be formed by reacting a compound of formula (8) with a suitable acid (e.g., HCl) in a solvent such as 1,4-dioxane, EtOAc, MeOH or water, or mixtures thereof. Alternatively, the reaction may be performed using acids such as TFA, neat or in a solvent such as DCM, at temperatures ranging from typically 0° C. to 60° C. Scheme 5 illustrates synthetic routes to certain compounds of formula (4). A compound of formula (10) may be formed by reacting a compound of formula (9) with NH3, either neat or as a solution, e.g. in water or MeOH, or with an ammonia synthetic equivalent (e.g., NH4Cl). The reaction may be performed using suitable coupling reagents (e.g., HATU, HOBt/EDC, T3P or Boc2O) in the presence of a base (typically an organic base, such as DIPEA or TEA) using a solvent such as THF, DMF, EtOAc or MeCN, or mixtures thereof, and at temperatures ranging from typically 0° C. to 120° C. A compound of formula (10) may be transformed into a compound of formula (11) by dehydration using a suitable reagent (typically TFAA or T3P) in a solvent such as DCM, DMF, EtOAc or MeCN, or mixtures thereof, and at a temperature ranging from typically 0° C. to 120° C. A compound of formula (4) may be formed by reacting a compound of formula (11) with a suitable acid (e.g., HCl or TsOH) in a solvent such as MeCN, 1,4-dioxane, EtOAc, MeOH or water, or mixtures thereof. Alternatively, the reaction may be performed using acids such as TFA, neat or in a solvent such as DCM, at temperatures ranging from typically 0° C. to 60° C. Scheme 6 illustrates synthetic routes to certain compounds of formula (6). A compound of formula (12) may be reacted with a compound of formula (3) to give a compound of formula (6). The reaction may be performed under conditions described for the analogous reaction described in Scheme 1. Scheme 7 illustrates synthetic routes to certain compounds of formula (12). A compound of formula (12) may be formed from compounds of formula (13) and (7), via a compound of formula (14). The reactions may be performed under conditions described for the analogous reactions described in Scheme 4. Scheme 8 illustrates synthetic routes to certain compounds of formula (5). A compound of formula (3) may be reacted with a compound of formula (15) to give a compound of formula (16). The reaction may be performed using suitable coupling reagents (e.g., HATU, HOBt/EDC or T3P) in the presence of a base (typically an organic base such as DIPEA or TEA) using a solvent such as DCM, DMF, EtOAc or MeCN, or mixtures thereof, and at temperatures ranging from typically 0° C. to 120° C. A compound of formula (5) may be formed by reacting a compound of formula (16) with a base (e.g., NaOH or LiOH) in an organic solvent (e.g., dioxane, THF, or MeOH, or mixtures thereof), and optionally in the presence of water. The reaction may be performed in a temperature interval from 0° C. to reflux. Alternatively, for compounds of formula (16) where R7=tert-butyl, the reaction may be performed with a suitable acid (e.g., HCl) in a solvent such as 1,4-dioxane, EtOAc, MeOH or water, or mixtures thereof. Alternatively, the reaction may be performed using acids such as TFA, neat or in a solvent such as DCM, at temperatures ranging from typically 0° C. to 60° C. Scheme 9 illustrates synthetic routes to certain compounds of formula (3). A compound of formula (18) may be formed by reacting a compound of formula (17) with an alcohol (e.g., MeOH or EtOH) in the presence of an acid (e.g., HCl or H2SO4) in a suitable solvent, or using the alcohol as solvent. Alternatively, the reaction may be promoted by reagents such as SOCl2in a suitable solvent, or using the alcohol (e.g., MeOH or EtOH) as solvent. Alternatively, a compound of formula (18) may be reacted with an alcohol (e.g., MeOH or EtOH) promoted by coupling reagents (e.g., EDC or TBTU) in the presence of base (such as DIPEA, TEA, or DMAP) using a solvent such as DCM, DMF, EtOAc or MeCN, or mixtures thereof, and at temperatures ranging from typically 0° C. to 120° C. A compound of formula (19) wherein R2is as defined in Formula (I), and where the attachment point to the quinoline is through a nitrogen atom, may be formed by reacting a compound of formula (18) with an amine H-R2(20), wherein R2is as defined in Formula (I). The reaction may be catalyzed with a suitable Pd-reagent, e.g. Pd2(dba)3with a suitable phosphine ligand (e.g., XPhos, CPhos, SPhos, RuPhos, DavePhos or XantPhos) in the presence of a base (such as Cs2CO3) in a suitable solvent (such as 1,4-dioxane), optionally in the presence of water, at temperatures ranging from room temperature to reflux. A compound of formula (19) wherein R2is as defined in formula (I), and where the attachment point to the quinoline is through a nitrogen atom, may be formed by reacting a compound of formula (18) with an amine H-R2(20), wherein R2is as defined in formula (I). The reaction may be catalyzed with a suitable Cu-reagent (e.g., CuI or Cu2O) in the presence of a base (such as K2CO3or Cs2CO3) in a suitable solvent (such as DMF) at temperatures ranging from room temperature to 160° C. A compound of formula (3) may be formed by reacting a compound of formula (19) with a base (e.g., NaOH or LiOH) in an organic solvent (e.g., 1,4-dioxane, THF, or MeOH, or mixtures thereof), and optionally in the presence of water. The reaction may be performed in a temperature interval from 0° C. to reflux. Alternatively, for compounds of formula (19) where R7=tert-butyl, the reaction may be performed with a suitable acid (e.g., HCl) in a solvent such as 1,4-dioxane, EtOAc, MeOH or water, or mixtures thereof. Alternatively, for compounds of formula (19) where R7=tert-butyl, the reaction may be performed using acids such as TFA, neat or in a solvent such as DCM, at temperatures ranging from typically 0° C. to 60° C. Alternatively, a compound of formula (3) wherein R2is as defined in Formula (I), and where the attachment point to the quinoline is through a nitrogen atom, may be formed directly from a compound of formula (17) by reaction with an amine H-R2(20), wherein R2is as defined in formula (I). The reaction may be performed under conditions described for the analogous reactions described above in Scheme 9. Scheme 10 illustrates synthetic routes to certain compounds of formula (3). A compound of formula (19) wherein R2is as defined in formula (I), and where the attachment point to the quinoline is through a carbon atom, may be formed by reacting a compound of formula (18) with a compound B-R2(21), wherein B is a boronic acid, boronate ester or trifluoroborate salt, and wherein R2is as defined in formula (I). The reaction may be catalyzed with a suitable Pd-reagent (e.g., Pd(dppf)Cl2) in the presence of a base (such as Na2CO3or K2CO3) in a suitable solvent (such as 1,4-dioxane), optionally in the presence of water, at temperatures ranging from room temperature to reflux. A compound of formula (3) may be formed by reacting a compound of formula (19) under conditions described for the analogous reactions described in Scheme 9. Alternatively, a compound of formula (3) wherein R2is as defined in Formula (I), and where the attachment point to the quinoline is through a carbon atom, may be formed directly from a compound of formula (17) by reaction with a compound B-R2(21), wherein B is a boronic acid, boronate ester or trifluoroborate salt, and wherein R2is as defined in formula (I). The reaction may be performed under conditions described for the analogous reaction described above in Scheme 10. Scheme 11 illustrates synthetic routes to certain compounds of formula (19). A compound of formula (22), wherein B is a boronic acid, boronate ester or trifluoroborate salt, may be formed by reacting a compound of formula (18) with a bis-boronic species (e.g., B2(OH)4(hypodiboric acid) or B2pin2(4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane)). The reaction may be catalyzed with a suitable Pd-reagent (e.g., Pd(dppf)Cl2) in the presence of a base (such as Na2CO3or K2CO3) in a suitable solvent (such as ethanol or 1,4-dioxane), optionally in the presence of water, at temperatures ranging from room temperature to reflux. A compound of formula (19) wherein R2is as defined in formula (I), and where the attachment point to the quinoline is through a carbon atom, may be formed by reacting a compound of formula (22) with an arylhalide or aryl pseudohalide of formula (23), wherein R2is as defined in formula (I) and X3is attached to R2via a carbon atom. The reaction may be catalyzed with a suitable Pd-reagent (e.g., Pd(dppf)Cl2) in the presence of a base (such as Na2CO3or K2CO3) in a suitable solvent (such as 1,4-dioxane), optionally in the presence of water, at temperatures ranging from room temperature to reflux. Scheme 12 illustrates synthetic routes to certain compounds of formula (17). A compound of formula (26) may be formed by reacting a compound of formula (24) with a 2-ketocarboxylic acid of formula (25), or a salt thereof (e.g., a sodium salt), in the presence of a base (e.g., NaOH) in water at reflux temperature, or at elevated temperatures ranging from typically 100° C. to 160° C. in a sealed vessel, or in a sealed tube in a microwave reactor. A compound of formula (17) may be formed by heating a compound of formula (26), either neat, or in a suitable solvent (e.g., water) at elevated temperatures ranging from typically 150° C. to 250° C. in a sealed vessel, or in sealed tube in a microwave reactor. Scheme 13 illustrates synthetic routes to certain compounds of formula (19). A compound of formula (28) wherein R2is as defined in formula (I), and where the attachment point to the quinoline is through a nitrogen atom or a carbon atom, may be formed from a compound of formula (27) using synthetic methodology performed under conditions described for the analogous reactions described in Scheme 9, 10 and 11. A compound of formula (19) may be formed by reacting a compound of formula (28) with carbon monoxide (1-10 atm), typically at a pressure of 10 atm, at a temperature ranging from typically 80° C. to 120° C. in a sealed vessel. The reaction may be catalyzed with a suitable Pd-reagent (e.g., Pd(dppf)Cl2) in the presence of a base (e.g., TEA) in the presence of a suitable alcohol (such as MeOH or EtOH) in a suitable solvent, or using the alcohol as solvent. Scheme 14 illustrates synthetic routes to certain compounds of formula (31). A compound of formula (29) in which R50is as defined in Formula (VIII) may be formed from a compound of formula (27) by metal halogen exchange using an organometallic reagent (e.g., BuLi) followed by reaction with an electrophile such as a ketone of formula (30). The reaction may be performed in a solvent such as THF at a temperature ranging from typically −78° C. to room temperature. A compound of formula (31) in which R50jis a fluorine, may be formed by reacting a compound of formula (29) with a fluorinating agent (e.g., DAST) in a solvent such as DCM at a temperature ranging from typically −20° C. to reflux. A compound of formula (31) may be converted into a compound of formula (VIII) using synthetic methodology analogous to those described in Scheme 13, 10 and 1. It should be understood that: (i) the organic reactions described in this disclosure are performed according to laboratory practice known to person skilled in the art; (ii) some of the reactions described in this disclosure may optionally be performed in different orders than laid out herein; (iii) chiral isomers of compounds in this disclosure can be resolved at any stage in the synthetic process using chiral resolving agents described in the literature and known to person skilled in the art, or using chiral chromatography methods described in the literature and known to person skilled in the art, or as described further in the Examples; (iv) additional and/or other protective groups may optionally be needed in some of the steps described above, and (v) a deprotection step therefore optionally may be performed, using method described in the literature and known to person skilled in the art. The protection and deprotection of functional groups is described in “Protective Groups in Organic Synthesis” 3rdEd, T. W. Greene and P. G. M. Wutz, Wiley-Interscience (1999), which publication is incorporated herein by reference. VIII. Examples The following descriptions of experiments, procedures, examples, and intermediates are intended to exemplify embodiments of the disclosure and are in no way intended to be limiting. Other compounds of this disclosure may be prepared using the methods illustrated in these examples, either alone or in combination with techniques generally known in the art. A. General Conditions Unless stated otherwise:(i) operations were carried out at room temperature (rt), i.e., in the range 17 to 25° C. and under an atmosphere of an inert gas such as N2unless otherwise stated;(ii) where reactions refer to the use of a microwave reactor, one of the following microwave reactors were used: Biotage Initiator, Personal Chemistry Emrys Optimizer, Personal Chemistry Smith Creator or CEM Explorer;(iii) in general, the course of reactions was followed by thin layer chromatography (TLC) and/or analytical high performance liquid chromatography (HPLC or UPLC) which was usually coupled to a mass spectrometer (LCMS).(iv) when necessary, organic solutions were dried over anhydrous MgSO4or Na2SO4, or by using ISOLUTE® Phase Separator, and work-up procedures were carried out using traditional phase separating techniques.(v), evaporations were carried out either by rotary evaporation in vacuo or in a Genevac HT-4/EZ-2 or Biotage V10;(vi) unless otherwise stated, flash column chromatography was performed on straight phase silica, using either Merck Silica Gel (Art. 9385) or pre-packed cartridges such as Biotage® SNAP cartridges (40-63 μm silica, 4-330 g), Biotage® Sfar Silica HC D cartridges (20 μm, 10-100 g), Interchim PuriFlash™ cartridges (25 μm, 4-120 g), Interchim PuriFlash™ cartridges (50 μm, 25-330 g), Grace™ GraceResolv™ Silica Flash Cartridges (4-120 g) or Agela Flash Colum Silica-CS cartridges (80-330 g), or on reversed phase silica using Agela Technologies C-18, spherical cartridges (20-35 μm, 100 A, 80-330 g), manually or automated using a Grace Reveleris® X2 Flash system or similar system;(vii) preparative reverse phase HPLC and preparative reverse phase SFC were performed using standard HPLC and SFC instruments, respectively, equipped with either a MS and/or UV triggered fraction collecting instrument, using either isocratic or a gradient of the mobile phase as described in the experimental section, and one of the following methods as described below; HPLC Prep Methods: PrepMethod A: The compound was purified by preparative HPLC on a YMC-Actus Triart C18 ExRS column (5 μm, 150×30 mm ID) using a gradient of MeCN in H2O/NH4HCO3(10 mM) as mobile phase; PrepMethod B: The compound was purified by preparative HPLC on a XBridge™ C18 OBD column (5 μm, 150×30 mm ID) using a gradient of MeCN in a H2O/NH4HCO3(10 mM)/NH3(0.1%, aq) buffer system as mobile phase; PrepMethod C: The compound was purified by preparative HPLC on a XSelect CSH OBD column (5 μm, 150×30 mm ID) using a gradient of MeCN in H2O/FA (0.1%) as mobile phase; PrepMethod D: The compound was purified by preparative HPLC on a XSelect CSH C18 OBD column (5 μm, 250×19 mm ID) using a gradient of MeCN in H2O/FA (0.1%) as mobile phase; PrepMethod E: The compound was purified by preparative HPLC on a Kromasil C8 column (10 μm, 250×20 mm ID) using a gradient of MeCN in H2O/MeCN/FA (95/5/0.2) as mobile phase; PrepMethod F: The compound was purified by preparative HPLC on a Waters™ Sunfire™ C18 OBD column (5 μm, 150×30 mm ID) using a gradient of MeCN in H2O/FA (0.1%) as mobile phase; PrepMethod G: The compound was purified by preparative HPLC on a Kromasil C8 column (10 μm, 250×50 mm ID) using a gradient of MeCN in H2O/MeCN/FA (95/5/0.2) as mobile phase; PrepMethod H: The compound was purified by preparative HPLC on a XBridge™ C18 column (10 μm, 250×50 mm ID) using a gradient of MeCN in H2O/MeCN/NH3(95/5/0.2) as mobile phase; PrepMethod I: The compound was purified by preparative HPLC on a XBridge™ C18 OBD column (5 μm, 250×19 mm ID) using a gradient of MeCN in H2O/NH4HCO3(10 mM) as mobile phase; PrepMethod N: The compound was purified by preparative HPLC on a XBridge™ C18 column (10 μm, 250×19 mm ID) using a gradient of MeCN in H2O/MeCN/NH3(95/5/0.2) as mobile phase; PrepMethod O: The compound was purified by preparative HPLC on a XBridge™ C18 column (5 μm, 250×19 mm ID) using a gradient of MeOH in H2O/NH4HCO3(10 mM) as mobile phase; PrepMethod P: The compound was purified by preparative HPLC on a XBridge™ Shield C18 column (5 μm, 150×30 mm ID) using a gradient of MeCN in H2O/FA (0.1%) as mobile phase; PrepMethod Q: The compound was purified by preparative HPLC on a Xbridge™ C18 ODB column (5 μm, 150×19 mm ID) using a gradient of MeCN in a H2O/NH3(0.2%, pH 10) buffer system as mobile phase; PrepMethod R: The compound was purified by preparative HPLC on a XBridge™ C18 OBD column (5 μm, 150×30 mm ID) using a gradient of MeCN in H2O/NH4HCO3(10 mM) as mobile phase; PrepMethod T: The compound was purified by preparative HPLC on a XBridge™ Shield C18 column (5 μm, 150×30 mm ID) using a gradient of MeCN in a H2O/NH4CO3(10 mM)/NH3(0.1%, aq) buffer system as mobile phase; PrepMethod U: The compound was purified by preparative HPLC on a Xselect CSH F-Phenyl OBD column, (5 μm, 250×19 mm ID) using a gradient of MeCN in H2O/FA (0.1%) as mobile phase; PrepMethod V: The compound was purified by preparative HPLC on a Waters™ Sunfire™ C18 OBD column (5 μm, 150×30 mm ID) using a gradient of MeCN in H2O/FA (0.1 M) as mobile phase; PrepMethod X: The compound was purified by preparative HPLC on a Xselect CSH OBD column, (5 μm, 150×30 mm ID) using a gradient of MeCN in H2O/FA (0.1%) as mobile phase; SFC Prep Methods: PrepMethod SFC-A: The compound was purified by preparative SFC on a Phenomenex Luna® HILIC column (5 μm, 250×30 mm ID) using EtOH/FA (20 mM) in CO2as mobile phase; PrepMethod SFC-B: The compound was purified by preparative SFC on a DAICEL DCpak® P4VP, (5 μm, 250×20 mm ID) using MeOH/2M NH3in MeOH (99.5/0.5) in CO2as mobile phase; PrepMethod SFC-C: The compound was purified by preparative SFC on a Waters™ BEH, (5 μm, 250×30 mm ID) using MeOH/H2O (NH350 mM) (97/3) in CO2as mobile phase; PrepMethod SFC-D: The compound was purified by preparative SFC on a Waters™ BEH (5 μm, 30×250 mm ID) using EtOH/FA (20 mM) in CO2as mobile phase; PrepMethod SFC-E: The compound was purified by preparative SFC on a Waters™ BEH, (5 μm, 250×30 mm ID) using MeOH/NH3(20 mM) in CO2as mobile phase; PrepMethod SFC-G: The compound was purified by preparative SFC on a Waters™ BEH, (3.5 μm, 100×3 mm ID) using MeOH/NH3(20 mM) in CO2as mobile phase; PrepMethod SFC-H: The compound was purified by preparative SFC on a Phenomenex Luna® HILIC column (5 μm, 250×30 mm ID) using MeOH/NH3(20 mM) in CO2as mobile phase. Relevant fractions were collected, combined and freeze-dried to give the purified compound or relevant fractions were collected, combined and concentrated at reduced pressure, extracted with DCM or EtOAc, and the organic phase was dried either over Na2SO4or by using a phase-separator, and then concentrated at reduced pressure to give the purified compound;(viii) chiral preparative chromatography was carried out using HPLC or SFC on a standard HPLC or SFC instruments, respectively, and using either isocratic or gradient run with mobile phase as described in the experimental section;(x) yields, where present, are not necessarily the maximum attainable, and when necessary, reactions were repeated if a larger amount of the reaction product was required;(xi) where certain compounds were obtained as an acid-addition salt, for example a mono-hydrochloride salt or a di-hydrochloride salt, the stoichiometry of the salt was based on the number and nature of the basic groups in the compound, the exact stoichiometry of the salt was generally not determined, for example by means of elemental analysis data;(xii) in general, the structures of the end-products of the Formula (I) were confirmed by nuclear magnetic resonance (NMR) and/or mass spectral techniques; proton NMR chemical shift values were measured on the delta scale using Bruker Avance III 300, 400, 500 and 600 spectrometers, operating at1H frequencies of 300, 400, 500 and 600 MHz, respectively. The experiments were typically recorded at 25° C. Chemical shifts are given in ppm with the solvent as internal standard. Protons on heteroatoms such as NH and OH protons are only reported when detected in NMR and can therefore be missing. In certain instances, protons can be masked or partially masked by solvent peaks and will therefore either be missing and not reported or reported as multiplets overlapping with solvent. The following abbreviations have been used (and derivatives thereof, e.g., dd, doublet of doublets, etc.): s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad; qn, quintet; p, pentet. In some cases, the structures of the end-products of the Formula (I) might appear as rotamers in the NMR-spectrum, in which instances only peaks of the major rotamer are reported. Electrospray mass spectral data were obtained using a Waters Acquity UPLC coupled to a Waters single quadrupole mass spectrometer or similar equipment, acquiring both positive and negative ion data, and generally, only ions relating to the parent structure are reported; high resolution electrospray mass spectral data were obtained using a Waters XEVO qToF mass spectrometer or similar equipment, coupled to a Waters Acquity UPLC, acquiring either positive and negative ion data, and generally, only ions relating to the parent structure are reported(xiii) intermediates were not necessarily fully purified but their structures and purity were assessed by TLC, analytical HPLC/UPLC, and/or NMR analysis and/or mass spectrometry;(xiv) unless stated otherwise compounds containing an asymmetric carbon and/or sulfur atom were not resolved;(xv) in general Examples and Intermediate compounds are named using ChemDraw Professional version 19.0.0.22 from PerkinElmer. ChemDraw Professional version 19.0.0.22 generates the names of chemical structures using the Cahn-Ingold-Prelog (CIP) rules for stereochemistry and follows IUPAC rules as closely as possible when generating chemical names. Stereoisomers are differentiated from each other by stereodescriptors cited in names and assigned in accordance with the CIP rules. ChemDraw is optionally using labels in the graphical representation of stereocenters such as ‘&’ and ‘or’ to describe the configuration of the stereochemical centers present in the structure. In general chemical structures of Examples and Intermediates containing the label ‘&’ at a stereocenter, means the configuration of such Example or Intermediate at that stereocenter is a mixture of both (R) and (S); and a label ‘or’ means the configuration of such Example or Intermediate at that stereocenter is either (S) or (R). Absolute, unspecified, ‘&’, and ‘or’ stereocenters can all be present in a single structure. In general for structures of Examples and Intermediates where all of the stereocenters are designated as ‘&’, the structure is named with a “rac-” prefix. For structures of Examples and Intermediates where all of the stereocenters are designated as ‘or’, the structure is named with a “rel-” prefix. In general Examples and Intermediate compounds are named using the descriptors (RS) and (SR) to denote general ‘&’ centers for chemical structures with multiple chiral centers where only some are designated as ‘&’. The descriptors (R*) and (S*) are used to denote the general ‘or’ centers for chemical structures with multiple chiral centers where only some are designated as ‘or’. In general, the descriptors (r) and (s) are used to describe the absolute configuration of any pseudoasymmetric centers in the structures of Examples and Intermediates. In general, the label “Isomer 1” corresponds to the first eluted isomer, and “Isomer 2” corresponds to the second eluted isomer, on a given chiral HPLC column and eluent, and are used to distinguish two isomers containing one or more stereocenters with absolute unknown configuration;(xvi) where reactions refer to being degassed or purged, this can be performed for example by purging the reaction solvent with a constant flow of nitrogen for a suitable period of time (for example 5 to 10 min)(xvii) in addition to the ones mentioned above, the following abbreviations have been used: α2APα2-antiplasminAlaalanineAMC7-amino-4-methylcoumarinaqaqueousatmatmosphereBMIbody mass indexBoc2Odi-tert-butyl dicarbonateBSAbovine serum albuminn-BuLin-butyllithiumtert-2-methylpropan-2-olBuOHC.CelciusChaps(3-((3-cholamidopropyl)dimethylammonio)-1-propanesulfonate)CLintintrinsic clearanceconcconcentrationm-CPBA3-chlorobenzoperoxoic acidCPhos2′-(dicyclohexylphosphaneyl)-N2,N2,N6,N6-tetramethyl-[1,1′-biphenyl]-2,6-diamineCPMEcyclopentyl methyl etherCRconcentration responseDAdaltonDASTdiethylaminosulfur trifluorideDavePhos2′-(dicyclohexylphosphaneyl)-N,N-dimethyl-[1,1′-biphenyl]-2-amineDCMdichloromethaneDEAdiethylamineDIPEAN-ethyl-N-isopropyl-propan-2-amineDMAP4-dimethylaminopyridineDMFN,N-dimethylformamideDMPUN,N′-dimethylpropyleneureaDMSOdimethyl sulfoxideDPP4dipeptidyl peptidase 4dppf1,1′-bis(diphenylphosphino)ferroceneEDC3-(ethyliminomethyleneamino)-N,N-dimethyl-propan-1-amine; hydrochlorideEDTAethylenediaminetetraacetic acid% eeenantiomeric excessEPhos PdCAS No 2132978-44-8G4ESIelectrospray ionizationEtOAcethyl acetateEtOHethanolEt2Zndiethyl zinkFAformic acidFACfinal assay concentrationFAPprolyl endopeptidase fibroblastactivation proteinFGF21fibroblast growth factor 21ggramGLP1glucagon-like peptide-1 receptorglyglycinehhour(s)HATU(1-[bis(dimethylamino)-methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphatehDPP7human dipeptidylpeptidase VIIhDPP8human dipeptidylpeptidase VIIIhDPP9human dipeptidylpeptidase IXHEPES(4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid)hFAPhuman prolyl endopeptidasefibroblast activation proteinHFIP1,1,1,3,3,3-hexafluoropropan-2-olHIShistidineHLMhuman liver microsomesHOBt1-hydroxybenzotriazole; hydrateHOMA-homeostatic model assessmentIRof insulin resistanceHPLChigh performance liquidchromatographyHRMShigh resolution massspectrometryIC50half-maximum inhibitoryconcentrationIDinner diameterIPApropan-2-olKOtBupotassium: tert-butoxideLCliquid chromatographyMMolarMeCNacetonitrileMeIiodomethaneMeOHmethanolminminute(s)mLmilliliterMSmass spectrometryMTBEtert-butyl methyl etherMWmolecular weightm/zmass spectrometry peak(s)NAFLDnonalcoholic fatty liver diseaseNASHnon-alcoholic steatohepatitisNBSN-bromosuccinimideNCSN-chlorosuccinimideNHSN-hydroxysuccinimideNinickelNiBr2nickel(II) bromide 2-methoxyethylO(CH2CH2OCH3)2ether complexNMPN-methyl-2-pyrrolidoneNMRnuclear magnetic resonanceNVnot validOTftrifluoromethanesulfonatePappapparent permeability coefficientPBSphosphate buffered salinePCRpolymeras chain reactionPd Catalyst [CAS:methanesulfonato(2-bis(3,5-1810068-35-9]di(trifluoromethyl)phenylphosphino)-3,6-dimethoxy-2′,6′-bis(dimethylamino)-1,1′-biphenyl)(2′-methylamino-1,1′-biphenyl-2-yl)palladium(II)Pd(dba)2bis(dibenzylideneacetone)palladiumPd2(dba)3(tris(dibenzylideneacetone)-dipalladium(0)Pd(dppf)Cl2•DCM[1,1′-bis(diphenylphosphino)-ferrocene]-dichloropalladium(II)complex with dichloromethane (1:1)Pd(dtbpf)Cl21,1′-bis(di-tert-butylphosphino)-ferrocene palladium dichloridePd(OAc)2palladium(II)acetatePKpharmacokineticsPPARperoxisome proliferator-activatedreceptorPREPprolyl endopeptidaseProprolinerHeprat hepatocytesrpmrevolution per minutertroom temperatureRUresponse unitRuPhosdicyclohexyl(2′,6′-diisopropoxy-[1,1′-biphenyl]-2-yl)phosphaneRuPhos Pd G2chloro(2-dicyclohexylphosphino-2′,6′-diisopropoxy-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II) [CASNumber 1375325-68-0]RuPhos Pd G3dicyclohexyl(2′,6′-diisopropoxy-[1,1′-biphenyl]-2-yl)phosphane (2′-amino-[1,1′-biphenyl]-2-yl)((methylsulfonyl)oxy)palladium[CAS No1445085-77-7]RuPhos Pd G4dicyclohexyl(2′,6′-diisopropoxy-[1,1′-biphenyl]-2-yl)phosphane (2′-(methylamino)-[1,1′-biphenyl]-2-yl)((methylsulfonyl)oxy)palladium[CAS Number 1599466-85-9]satsaturatedSDstandard deviationSFCsupercritical fluid chromatographySGLT2sodium-glucose transport protein 2SPhosdicyclohexyl(2′,6′-dimethoxy-[1,1′-biphenyl]-2-yl)phosphaneSPhos Pd G3(2-dicyclohexylphosphino-2′,6′-dimethoxybiphenyl) [2-(2′-amino-1,1′-biphenyl)]palladium(II)methanesulfonate [CAS No1445085-82-4]TBTU2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethylaminium tetrafluoro-borateTCEPtris(2-carboxyethyl)phosphinehydrochlorideTEAtriethylamineTFAtrifluoro acetic acidTFAAtrifluoro acetic acid anhydrideTHFtetrahydrofuranTLCthin layer chromatographyT3Ppropanephosphonic acid anhydrideTriphosgenebis(trichloromethyl) carbonateTris HCltris(hydroxymethyl)aminomethanehydrochlorideTriton X-100t-octylphenoxypolyethoxyethanolTsOHp-toluenesulfonic acidμLmicroliterUPLCultra performance liquidchromatographyUVultravioletv/vvolume by volumeXantPhos(9,9-dimethyl-9H-xanthene-4,5-diyl)bis(diphenylphosphane)XantPhos Pd G4[CAS No 1621274-19-8]XPhosdicyclohexyl(2′,4′,6′-triisopropyl-[1,1′-biphenyl]-2-yl)phosphaneXPhos-Pd-G2chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)-]palladium(II), X-Phosaminobiphenyl palladium chlorideprecatalyst [CAS No 1310584-14-5]XPhos Pd G3(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II)methanesulfonate [CAS Number1445085-55-1] B. Intermediate Compounds Intermediate 1: tert-Butyl (R)-4-cyanothiazolidine-3-carboxylate Step a) tert-Butyl (R)-4-carbamoylthiazolidine-3-carboxylate Boc2O (18.6 mL, 80.2 mmol) was added to a stirred solution of (R)-3-(tert-butoxycarbonyl)thiazolidine-4-carboxylic acid (17.0 g, 72.9 mmol) and pyridine (7.07 mL, 87.5 mmol) in EtOAc (170 mL) and the reaction mixture was stirred at rt for 3 h. Then, a solution of NH3(aq, 25%, 6 mL) was added dropwise and the mixture was stirred at rt overnight. The reaction mixture was diluted with EtOAc, the phases were separated and the organic phase was washed with sat NaCl, dried, filtered through a pad of silica gel, washed with EtOAc and evaporated to give the crude title compound (16.9 g, 100%) as a colorless oil, which was used directly in the next step. Step b) tert-Butyl (R)-4-cyanothiazolidine-3-carboxylate TFAA (12.4 mL, 87.5 mmol) as a solution in EtOAc (20 mL) was added to a solution of crude tert-butyl (R)-4-carbamoylthiazolidine-3-carboxylate (16.9 g, 72.9 mmol) and pyridine (14.7 mL, 182 mmol) in EtOAc (150 mL) at rt. The mixture was stirred at rt for 4 h and then diluted with EtOAc, washed with aq, HCl (1 M), and sat NaHCO3. The organic phase was dried, filtered through a pad of silica gel, washed with EtOAc, and evaporated to give a light yellow oil which solidified on standing. The crude solid material was suspended in heptane:EtOAc (4:1, 50 mL) and stirred at rt overnight. The solids were filtered off, washed with heptane:EtOAc (4:1), and dried to give the title compound (12.0 g, 83%) as a colorless solid;1H NMR (400 MHz, CDCl3) δ 5.20-4.79 (m, 1H), 4.60-4.53 (m, 1H), 4.53-4.36 (m, 1H), 3.40-3.18 (m, 2H), 1.51 (s, 9H). Intermediate 2: (R)-Thiazolidine-4-carbonitrile hydrochloride A solution of aq HCl (12 M, 11 mL) in MeOH (140 mL) was added slowly to a solution of tert-butyl (R)-4-cyanothiazolidine-3-carboxylate Intermediate 1 (6.0 g, 28 mmol) in MeOH (140 mL) at rt. The clear colorless solution was stirred at rt for 2 h. Solvents were evaporated to give the title compound (4.22 g, 100%) as a colorless solid;1H NMR (400 MHz, CD3OD) δ 4.90 (dd, 1H), 4.35-4.24 (m, 2H), 3.37-3.24 (m, 2H). Intermediate 3: tert-Butyl (R)-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)carbamate DIPEA (19.6 mL, 112 mmol) was added to a suspension of (R)-thiazolidine-4-carbonitrile hydrochloride Intermediate 2 (4.22 g, 28 mmol), (tert-butoxycarbonyl)glycine (6.13 g, 35.0 mmol) and T3P (41.6 mL, 70.0 mmol, 50% solution in EtOAc) in EtOAc (120 mL). The mixture was heated at 60° C. for 4 h. The mixture was diluted with EtOAc, and sequentially washed with water, aq HCl (1 M) and sat NaHCO3. The organic phase was dried, filtered and evaporated. The residue was filtered through a pad of silica gel, washed with heptane:EtOAc (1:1) and evaporated to give an oil which was triturated with heptane:DCM to give the title compound (7.60 g, 100%) as an almost colorless solid;1H NMR (400 MHz, CDCl3) δ 5.36-5.25 (m, 2H), 4.59-4.52 (m, 2H), 4.14-3.90 (m, 2H), 3.29 (d, 2H), 1.45 (s, 9H). Intermediate 4: (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride A solution of aq HCl (12 M, 5.6 mL) in MeOH (140 mL) was added slowly to a solution of tert-butyl (R)-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)carbamate Intermediate 3 (7.60 g, 28.0 mmol) in MeOH (140 mL), then the solution was stirred at rt overnight. The solvents were evaporated to give the title compound (5.80 g, 100%) as a colorless solid;1H NMR (400 MHz, CD3OD) δ 5.34 (t, 1H), 4.72 (d, 1H), 4.62 (d, 1H), 4.11-3.94 (m, 2H), 3.41-3.36 (m, 2H). Intermediate 5: Ethyl 6-(1,2-oxazinan-2-yl)quinoline-4-carboxylate 1,2-Oxazinane hydrochloride (37 mg, 0.30 mmol) was added to a mixture of ethyl 6-bromoquinoline-4-carboxylate (56 mg, 0.20 mmol), Cs2CO3(195 mg, 0.60 mmol), Pd2dba3(9.0 mg, 10 μmol) and XPhos (9.5 mg, 0.02 mmol) in dioxane (1 mL). The flask was sealed, purged with N2(g) and the mixture was heated at 90° C. overnight. The mixture was diluted with EtOAc and washed with water. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 20-50% EtOAc in heptane) to give the title compound (50 mg, 87%) as a yellow oil which solidified on standing; MS m/z (ESI) [M+H]+287.2. Intermediate 6: 6-(1,2-Oxazinan-2-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 0.52 mL) was added to a solution of ethyl 6-(1,2-oxazinan-2-yl)quinoline-4-carboxylate Intermediate 5 (50 mg, 0.17 mmol) in MeOH (2 mL) and water (1 mL). The mixture was stirred at rt for 3 h, then neutralized with aq HCl (1 M), and evaporated to give the title compound (45 mg, 100%) as a yellow-red semisolid; MS m/z (ESI), [M+H]+259.1. Intermediate 7: Ethyl 3-fluoro-6-morpholinoquinoline-4-carboxylate Morpholine (70 mg, 0.80 mmol) was added to a mixture of ethyl 6-chloro-3-fluoroquinoline-4-carboxylate (101 mg, 0.4 mmol), Cs2CO3(0.391 g, 1.20 mmol), Pd2dba3(18 mg, 0.02 mmol) and XPhos (19 mg, 0.04 mmol) in dioxane (2 mL). The flask was sealed, purged with N2(g) and the mixture was heated at 90° C. for 1 h. The mixture was diluted with EtOAc and washed with water. The organic phase was dried, filtered and evaporated to give the title compound (122 mg, 100%) as a yellow semisolid; MS m/z (ESI) [M+H]+305.2. Intermediate 8: 3-Fluoro-6-morpholinoquinoline-4-carboxylic acid Aq NaOH (1 M, 1.25 mL) was added to a solution of ethyl 3-fluoro-6-morpholinoquinoline-4-carboxylate Intermediate 7 (190 mg, 0.62 mmol) in MeOH (6 mL) and water (3 mL). The mixture was stirred at rt for 3 h. The mixture was neutralized with aq HCl (1 M), and evaporated to give the title compound (172 mg, 100%) as a red-yellow semisolid; MS m/z (ESI) [M+H]+277.3. Intermediate 9: Ethyl 6-thiomorpholinoquinoline-4-carboxylate Thiomorpholine (41 mg, 0.40 mmol) was added to a mixture ethyl 6-bromoquinoline-4-carboxylate (56 mg, 0.2 mmol), Cs2CO3(0.130 g, 0.40 mmol), Pd2dba3(9.2 mg, 0.01 mmol) and XPhos (9.5 mg, 0.02 mmol) in dioxane (1 mL). The flask was sealed, purged with N2(g) and the mixture was heated at 90° C. overnight. The mixture was diluted with EtOAc, washed with water (3×). The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 20-50% EtOAc in heptane) to give the title compound (55 mg, 91%) as a yellow oil; MS m/z (ESI) [M+H]+303.2. Intermediate 10: 6-Thiomorpholinoquinoline-4-carboxylic acid LiOH (22 mg, 0.91 mmol) was added to a solution of ethyl 6-thiomorpholinoquinoline-4-carboxylate Intermediate 9 (55 mg, 0.18 mmol) in MeOH (1 mL) and water (1 mL). The mixture was stirred at rt overnight, and then neutralized with aq HCl (1 M). The solvents were evaporated under reduced pressure to give the title compound (50 mg, 100%) as a yellow-red semisolid; MS m/z (ESI) [M+H]+275.1. Intermediate 11: Ethyl 6-(2,2-difluoromorpholino)quinoline-4-carboxylate 2,2-Difluoromorpholine hydrochloride (72 mg, 0.45 mmol) was added to a mixture of ethyl 6-bromoquinoline-4-carboxylate (84 mg, 0.30 mmol), Cs2CO3(0.293 g, 0.90 mmol), Pd2dba3(14 mg, 0.02 mmol) and XPhos (14 mg, 0.03 mmol) in dioxane (1.5 mL). The flask was sealed, purged with N2(g) and the mixture was heated at 90° C. overnight. The mixture was diluted with EtOAc and washed with water. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (heptane:EtOAc 1:1) to give the title compound (85 mg, 88%) as a yellow solid; MS m/z (ESI) [M+H]+323.4. Intermediate 12: 6-(2,2-Difluoromorpholino)quinoline-4-carboxylic acid Aq NaOH (1 M, 0.5 mL) was added to a solution of ethyl 6-(2,2-difluoromorpholino)quinoline-4-carboxylate Intermediate 11 (80 mg, 0.25 mmol) in MeOH (2.5 mL) and water (1 mL). The mixture was stirred at rt overnight, and then neutralized with aq HCl (1 M). The solvents were evaporated under reduced pressure to give the title compound (73 mg, 100%) as a yellow solid; MS m/z (ESI) [M+H]+295.3. Intermediate 13: Ethyl 6-(2,2,6,6-tetrafluoromorpholino)quinoline-4-carboxylate 2,2,6,6-Tetrafluoromorpholine (64 mg, 0.40 mmol) was added to a mixture of ethyl 6-bromoquinoline-4-carboxylate (56 mg, 0.2 mmol), Cs2CO3(0.195 g, 0.60 mmol), Pd2dba3(9.2 mg, 0.01 mmol) and XPhos (9.5 mg, 0.02 mmol) in dioxane (1 mL). The flask was sealed, purged with N2(g) and the mixture was heated at 90° C. overnight. The mixture was diluted with EtOAc and washed with water. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 20-50% EtOAc in heptane) to give the title compound (60 mg, 84%) as an almost colorless solid; MS m/z (ESI) [M+H]+359.2. Intermediate 14: 6-(2,2,6,6-Tetrafluoromorpholino)quinoline-4-carboxylic acid Aq NaOH (1 M, 0.5 mL, 0.50 mmol) was added a solution of ethyl 6-(2,2,6,6-tetrafluoromorpholino)quinoline-4-carboxylate Intermediate 13 (60 mg, 0.17 mmol) in MeOH (2 mL) and water (1 mL). The mixture was stirred at rt for 3 h, and then neutralized with aq HCl (1 M). The solvents were evaporated under reduced pressure to give the title compound (55 mg, 99%) as a light yellow solid; MS m/z (ESI) [M+H]+331.1. Intermediate 15: Ethyl 6-(2,3-dihydro-4H-benzo[b][1,4]oxazin-4-yl)quinoline-4-carboxylate 3,4-Dihydro-2H-benzo[b][1,4]oxazine (54 mg, 0.40 mmol) was added to a mixture of ethyl 6-bromoquinoline-4-carboxylate (56 mg, 0.2 mmol), Cs2CO3(0.130 g, 0.40 mmol) and RuPhos Pd G2 (16 mg, 0.02 mmol) in dioxane (1 mL). The flask was sealed, purged with N2(g) and the mixture was heated at 90° C. overnight. The mixture was diluted with EtOAc and washed with water. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 20-50% EtOAc in heptane) to give the title compound (53 mg, 79%) as a yellow semisolid; MS m/z (ESI) [M+H]+335.2. Intermediate 16: 6-(2,3-Dihydro-4H-benzo[b][1,4]oxazin-4-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 0.5 mL) was added a solution of ethyl 6-(2,3-dihydro-4H-benzo[b][1,4]oxazin-4-yl)quinoline-4-carboxylate Intermediate 15 (53 mg, 0.16 mmol) in MeOH (1 mL) and water (0.5 mL). The mixture was stirred at rt overnight, and then neutralized with aq HCl (1 M). The solvents were evaporated under reduced pressure to give the title compound (49 mg, 100%) as a yellow-red semisolid; MS m/z (ESI) [M+H]+307.2. Intermediate 17: Ethyl 6-((3S,4s,5R)-4-hydroxy-3,5-dimethylpiperidin-1-yl)quinoline-4-carboxylate (3R,4s,5S)-3,5-Dimethylpiperidin-4-ol hydrochloride (50 mg, 0.30 mmol) was added to a mixture of ethyl 6-bromoquinoline-4-carboxylate (56 mg, 0.2 mmol), Cs2CO3(0.195 g, 0.60 mmol), Pd2(dba)3(9 mg, 0.01 mmol) and XPhos (9.5 mg, 0.02 mmol) in 1,4-dioxane (1 mL). The flask was sealed, purged with N2(g), and heated at 90° C. overnight. The mixture was diluted with EtOAc and washed with water. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 20-50% EtOAc in heptane) to give the title compound (20 mg, 30%) as a yellow semisolid; MS m/z (ESI) [M+H]+329.2. Intermediate 18: 6-((3S,4s,5R)-4-Hydroxy-3,5-dimethylpiperidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 0.18 mL) was added to a solution of ethyl 6-((3S,4s,5R)-4-hydroxy-3,5-dimethylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 17 (20 mg, 0.06 mmol) in MeOH (1 mL) and water (0.5 mL). The mixture was stirred at rt overnight. The mixture was neutralized with aq HCl (1 M), and evaporated to give the title compound (18 mg, 98%) as a yellow-red semisolid; MS m/z (ESI) [M+H]+301.2. Intermediate 19: tert-Butyl 6-(4-methoxypiperidin-1-yl)quinoline-4-carboxylate 4-Methoxypiperidine (179 mg, 1.56 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (WO2019/154886) (400 mg, 1.30 mmol), Cs2CO3(846 mg, 2.60 mmol), XPhos (124 mg, 0.26 mmol) and Pd2(dba)3(119 mg, 0.13 mmol) in 1,4-dioxane (15 mL) at 15° C. under N2(g). The resulting mixture was stirred at 100° C. for 3 h. The reaction mixture was concentrated and the residue was dissolved in EtOAc (125 mL), and washed sequentially with water (75 mL) and sat brine (75 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 3:1) to afford the title compound (300 mg, 68%) as a yellow solid; MS m/z (ESI) [M+H]+343.1. Intermediate 20: 6-(4-Methoxypiperidin-1-yl)quinoline-4-carboxylic acid HCl (0.025 mL, 4 M in 1,4-dioxane) was added to tert-butyl 6-(4-methoxypiperidin-1-yl)quinoline-4-carboxylate Intermediate 19 (280 mg, 0.82 mmol) in 1,4-dioxane (15 mL) at 20° C. under air. The resulting mixture was stirred at 20° C. for 20 h. The precipitate was collected by filtration, washed with 1,4-dioxane (100 mL) and dried under vacuum to afford the title compound (200 mg, 85%) as a yellow solid; MS m/z (ESI) [M+H]+287.1. Intermediate 21: 6-Bromo-8-methylquinoline-2,4-dicarboxylic acid 5-Bromo-7-methylindoline-2,3-dione (500 mg, 2.08 mmol) was added to a solution of sodium 2-oxopropanoate (275 mg, 2.50 mmol) in aq NaOH (20%, 10 mL) in a microwave tube. The tube was sealed and heated at 110° C. for 1 h in a microwave reactor and then cooled to rt. The reaction mixture was acidified to pH 2 with aq HCl (2 M). The precipitate was collected by filtration, washed with water (100 mL) and dried under vacuum to give the title compound (550 mg, 85%) as a yellow solid; MS m/z (ESI) [M+H]+309.9. Intermediate 22: 6-Bromo-8-methylquinoline-4-carboxylic acid 6-Bromo-8-methylquinoline-2,4-dicarboxylic acid Intermediate 21 (430 mg, 1.39 mmol) was dissolved in water (15 mL) in a steel reactor. The reactor was sealed, and heated at 190° C. for 6 h in an oil bath, and then cooled to rt. The precipitate was collected by filtration, washed with water (100 mL), and dried under vacuum to afford the title compound (233 mg, 63%) as a yellow solid; MS m/z (ESI) [M+H]+265.9. Intermediate 23: Methyl 6-bromo-8-methylquinoline-4-carboxylate SOCl2(0.082 mL, 1.13 mmol) was added slowly to 6-bromo-8-methylquinoline-4-carboxylic acid Intermediate 22 (300 mg, 1.13 mmol) in MeOH (10 mL) at 15° C. under air. The resulting mixture was stirred at 60° C. for 16 h. The reaction mixture was concentrated, the residue was dissolved in DCM (100 mL), and washed sequentially with water (50 mL) and sat brine (50 mL). The organic layer was dried over Na2SO4, filtered, and evaporated. The crude product was purified by preparative TLC (DCM:MeOH, 40:1) to afford the title compound (250 mg, 79%) as a yellow solid; MS m/z (ESI) [M+H]+279.9. Intermediate 24: Methyl 8-methyl-6-morpholinoquinoline-4-carboxylate Morpholine (311 mg, 3.57 mmol) was added to a mixture of methyl 6-bromo-8-methylquinoline-4-carboxylate Intermediate 23 (200 mg, 0.71 mmol), Cs2CO3(465 mg, 1.43 mmol), Pd Catalyst [CAS: 1810068-35-9] (82 mg, 0.07 mmol) in 1,4-dioxane (15 mL) at 20° C. under N2(g). The resulting mixture was heated at 100° C. for 5 h. The reaction mixture was concentrated and diluted with EtOAc (100 mL), and washed sequentially with water (50 mL) and sat brine (50 mL). The organic layer was dried over Na2SO4, filtered, and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 5:1) to afford the title compound (190 mg, 93%) as a yellow solid; MS m/z (ESI) [M+H]+287.0. Intermediate 25: 8-Methyl-6-morpholinoquinoline-4-carboxylic acid A solution of NaOH (126 mg, 3.14 mmol) in water (3 mL) was added to a stirred solution of methyl 8-methyl-6-morpholinoquinoline-4-carboxylate Intermediate 24 (180 mg, 0.63 mmol) in MeOH (9 mL) at 20° C. The resulting mixture was stirred at 20° C. for 2 h. The reaction mixture was acidified to pH 5 with aq HCl (2 M). The reaction mixture was concentrated and diluted with EtOAc (125 mL), and washed sequentially with water (50 mL) and sat brine (50 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (150 mg, 88%) as a red solid; MS m/z (ESI) [M+H]+273.0. Intermediate 26: 6-Bromo-7-methylquinoline-2,4-dicarboxylic acid 5-Bromo-6-methylindoline-2,3-dione (800 mg, 3.33 mmol) was added to sodium 2-oxopropanoate (440 mg, 4.00 mmol) in aq NaOH (20%, 15 mL) in a microwave reactor tube. The tube was sealed and heated at 110° C. for 1 h in a microwave reactor and then cooled to rt. The reaction mixture was acidified to pH 2 with aq HCl (2 M). The precipitate was collected by filtration, washed with water (50 mL), and dried under vacuum to afford the title compound (540 mg, 52%) as a brown solid; MS m/z (ESI) [M+H]+309.9. Intermediate 27: 6-Bromo-7-methylquinoline-4-carboxylic acid 6-Bromo-7-methylquinoline-2,4-dicarboxylic acid Intermediate 26 (400 mg, 1.29 mmol) was dissolved in water (10 mL) in a steel reactor. The reactor was sealed, and heated at 190° C. for 6 h in an oil bath, and then cooled to rt. The precipitate was collected by filtration, washed with water (125 mL), and dried under vacuum to afford the title compound (200 mg, 58%) as a dark solid; MS m/z (ESI) [M+H]+265.9. Intermediate 28: Methyl 6-bromo-7-methylquinoline-4-carboxylate SOCl2(0.34 mL, 4.7 mmol) was added slowly to 6-bromo-7-methylquinoline-4-carboxylic acid Intermediate 27 (250 mg, 0.94 mmol) in MeOH (10 mL) at 15° C. under air. The resulting mixture was heated at 60° C. for 2 h. The reaction mixture was concentrated, the residue was diluted with DCM (75 mL), and washed sequentially with water (20 mL) and sat brine (20 mL). The organic layer was dried over Na2SO4, filtered, and evaporated. The residue was purified by preparative TLC (DCM:MeOH, 40:1) to afford the title compound (215 mg, 82%) as a yellow solid; MS m/z (ESI) [M+H]+279.9. Intermediate 29: Methyl 7-methyl-6-morpholinoquinoline-4-carboxylate Morpholine (112 mg, 1.29 mmol) was added to methyl 6-bromo-7-methylquinoline-4-carboxylate Intermediate 28 (180 mg, 0.64 mmol), Pd Catalyst [CAS: 1810068-35-9](73 mg, 0.06 mmol) and Cs2CO3(419 mg, 1.29 mmol) in 1,4-dioxane (10 mL) at 15° C. under N2(g). The resulting mixture was heated at 100° C. for 16 h. The reaction mixture was filtered through filter paper and the filtrate was concentrated. The residue was diluted with EtOAc (100 mL), and washed sequentially with sat brine (25 mL) and water (25 mL). The organic layer was dried over Na2SO4, filtered, and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 5:1) to afford the title compound (138 mg, 75%) as a yellow solid; MS m/z (ESI) [M+H]+287.0. Intermediate 30 7-Methyl-6-morpholinoquinoline-4-carboxylic acid NaOH (112 mg, 2.79 mmol) in water (3 mL) was added to a stirred solution of methyl 7-methyl-6-morpholinoquinoline-4-carboxylate Intermediate 29 (160 mg, 0.56 mmol) in MeOH (9 mL) at 16° C. under N2(g). The resulting mixture was stirred at 20° C. for 2 h. The reaction mixture was adjusted to pH 5 with aq HCl (2 M). The reaction mixture was concentrated, and the residue was dissolved in EtOAc (50 mL), and washed sequentially with sat brine (25 mL) and water (25 mL). The organic layer was dried over Na2SO4, filtered, and evaporated to afford the title compound (120 mg, 79%) as a yellow solid; MS m/z (ESI) [M+H]+273.0. Intermediate 31: tert-Butyl 6-(2-oxopyrrolidin-1-yl)quinoline-4-carboxylate Pyrrolidin-2-one (57 mg, 0.67 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (160 mg, 0.52 mmol), Cs2CO3(254 mg, 0.78 mmol) and XPhos Pd G3 (44 mg, 0.05 mmol) in 1,4-dioxane (5 mL) at 15° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was filtered through Celite©. The filtrate was concentrated under reduced pressure and the residue was purified by preparative TLC (EtOAc:petroleum ether, 2:1), to afford the title compound (160 mg, 99%) as a beige oil which solidified on standing; MS m/z (ESI) [M+H]+313.15. Intermediate 32: 6-(2-Oxopyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (5 mL, 65 mmol) was added to a stirred solution of tert-butyl 6-(2-oxopyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 31 (140 mg, 0.45 mmol) in DCM (5 mL) at 27° C. The resulting solution was stirred at 27° C. for 7 h. The solvent was removed under reduced pressure to afford the title compound (115 mg, 100%) as a yellow solid; MS m/z (ESI) [M+H]+257.0. Intermediate 33: tert-Butyl 6-(3,3-dimethyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxylate 3,3-Dimethylpyrrolidin-2-one (132 mg, 1.17 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), Cs2CO3(634 mg, 1.95 mmol), Pd2(dba)3(89 mg, 0.10 mmol) and XPhos (93 mg, 0.19 mmol) in 1,4-dioxane (15 mL) at 20° C. under N2(g). The resulting mixture was heated at 100° C. for 3 h. The reaction mixture was concentrated, diluted with EtOAc (100 mL), and washed sequentially with water (25 mL) and sat brine (25 mL). The organic layer was dried over Na2SO4, filtered, and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 5:1) to afford the title compound (220 mg, 66%) as a yellow solid; MS m/z (ESI) [M+H]+341.0. Intermediate 34: 6-(3,3-Dimethyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (2 mL, 26 mmol) was added to tert-butyl 6-(3,3-dimethyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 33 (200 mg, 0.59 mmol) in DCM (10 mL) at 20° C. under air. The resulting mixture was stirred at 20° C. for 3 h. The reaction mixture was concentrated, redissolved in DCM (50 mL), and washed with water (15 mL). The organic layer was dried over Na2SO4, filtered, and evaporated to afford crude product. The residue was purified by preparative TLC (DCM:MeOH, 10:1) to afford the title compound (120 mg, 72%) as a yellow solid; MS m/z (ESI) [M+H]+285.0. Intermediate 35: tert-Butyl 6-(5,5-dimethyl-2-oxooxazolidin-3-yl)quinoline-4-carboxylate 5,5-Dimethyloxazolidin-2-one (134 mg, 1.17 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), Cs2CO3(634 mg, 1.95 mmol), Pd2(dba)3(89 mg, 0.10 mmol) and XPhos (93 mg, 0.19 mmol) in 1,4-dioxane (10 mL) at 20° C. under N2(g). The resulting mixture was stirred at 100° C. for 3 h. The reaction mixture was concentrated, and redissolved in EtOAc (100 mL), and washed sequentially with water (20 mL) and sat brine (20 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford crude product. The residue was purified by preparative TLC (EtOAc: petroleum ether, 1:5) to afford the title compound (240 mg, 72%) as a yellow solid; MS m/z (ESI) [M+H]+343.1. Intermediate 36: 6-(5,5-Dimethyl-2-oxooxazolidin-3-yl)quinoline-4-carboxylic acid HCl (4 M in 1,4-dioxane) (10 mL) was added to tert-butyl 6-(5,5-dimethyl-2-oxooxazolidin-3-yl)quinoline-4-carboxylate Intermediate 35 (220 mg, 0.64 mmol) at 20° C. under air. The resulting mixture was stirred at 40° C. for 6 h. The reaction mixture was concentrated, the residue was dissolved in EtOAc (50 mL), and washed with water (20 mL). The organic layer was dried over Na2SO4, filtered, and evaporated to afford the title compound (150 mg, 82%) as a yellow solid; MS m/z (ESI) [M+H]+287.0. Intermediate 37: tert-Butyl 6-(2-oxopiperidin-1-yl)quinoline-4-carboxylate Piperidin-2-one (575 mg, 5.80 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (500 mg, 1.45 mmol), Cs2CO3(945 mg, 2.90 mmol), Pd2(dba)3(13 mg, 0.01 mmol) and XPhos (14 mg, 0.03 mmol) in 1,4-dioxane (20 mL) at 15° C. The resulting suspension was stirred at 100° C. for 15 h under N2(g). The reaction mixture was filtered through Celite©. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 4:1) to afford the title compound (132 mg, 28%) as a brown solid; MS m/z (ESI) [M+H]+327.2. Intermediate 38: 6-(2-Oxopiperidin-1-yl)quinoline-4-carboxylic acid TFA (0.11 mL, 1.5 mmol) was added to a solution of tert-butyl 6-(2-oxopiperidin-1-yl)quinoline-4-carboxylate Intermediate 37 (120 mg, 0.37 mmol) in DCM (2 mL). The resulting solution was stirred at 25° C. for 6 h. The solvent was removed under reduced pressure afford the title compound (130 mg) as a brown solid; MS m/z (ESI) [M+H]+271.15. Intermediate 39: tert-Butyl 6-(3,3-dimethyl-2-oxopiperidin-1-yl)quinoline-4-carboxylate 3,3-Dimethylpiperidin-2-one (825 mg, 6.49 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (500 mg, 1.62 mmol), Cs2CO3(793 mg, 2.43 mmol), Pd2(dba)3(15 mg, 0.02 mmol), and XPhos (15 mg, 0.03 mmol) in 1,4-dioxane (15 mL) under N2(g). The resulting suspension was stirred at 100° C. for 24 h. The reaction mixture was filtered through silica. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (80 mg, 14%) as a brown solid. MS m/z (ESI) [M+H]+355.2. Intermediate 40: 6-(3,3-Dimethyl-2-oxopiperidin-1-yl)quinoline-4-carboxylic acid TFA (0.061 mL, 0.79 mmol) was added to a solution of tert-butyl 6-(3,3-dimethyl-2-oxopiperidin-1-yl)quinoline-4-carboxylate Intermediate 39 (70 mg, 0.20 mmol) in DCM (2 mL). The resulting solution was stirred at 25° C. for 6 h. The solvent was removed under reduced pressure to afford the title compound (60 mg, 100%) as a brown solid; MS m/z (ESI) [M+H]+299.15. Intermediate 41: tert-Butyl 6-(2,2-dimethyl-3-oxomorpholino)quinoline-4-carboxylate 2,2-Dimethylmorpholin-3-one (122 mg, 0.94 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (291 mg, 0.94 mmol), Cs2CO3(616 mg, 1.89 mmol), Pd2(dba)3(86 mg, 0.09 mmol) and CPhos (82 mg, 0.19 mmol) in 1,4-dioxane (1 mL) at 25° C. The resulting suspension was stirred at 100° C. for 2 h. The reaction mixture was extracted with DCM (3×100 mL). The organic phases were combined and washed with sat brine (2×60 mL). The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:2) to afford the title compound (299 mg, 89%) as a yellow solid; MS m/z (ESI) [M+H]+357.2. Intermediate 42: 6-(2,2-Dimethyl-3-oxomorpholino)quinoline-4-carboxylic acid HCl (4 M in 1,4-dioxane, 10 mL) was added to tert-butyl 6-(5,5-dimethyl-2-oxooxazolidin-3-yl)quinoline-4-carboxylate Intermediate 41 (289 mg, 0.84 mmol) at 20° C. under air. The resulting mixture was stirred at 40° C. for 12 h. The solvent was removed under reduced pressure to afford the title compound (315 mg) as a tan solid; MS m/z (ESI) [M+H]+301.1. Intermediate 43: tert-Butyl 6-(2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate 2-Azaspiro[3.3]heptane hydrochloride (347 mg, 2.60 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (400 mg, 1.30 mmol), Cs2CO3(1.69 g, 5.19 mmol), Pd2(dba)3(119 mg, 0.13 mmol), and XPhos (124 mg, 0.26 mmol) in 1,4-dioxane (10 mL) at 10° C. The resulting suspension was stirred at 100° C. for 3 h under N2(g). The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (150 mg, 36%) as a yellow solid. MS m/z (ESI) [M+H]+325.3. Intermediate 44: 6-(2-Azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid TFA (53 mg, 0.46 mmol) was added to a solution of tert-butyl 6-(2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Intermediate 43 (150 mg, 0.46 mmol) in 1,4-dioxane (3 mL), and stirred at 25° C. overnight. The solvent was removed under reduced pressure to give the title compound; MS m/z (ESI) [M+H]+269.1. Intermediate 45: tert-Butyl 6-(3,3-dimethyl-1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylate 3,3-Dimethyl-1-oxa-6-azaspiro[3.3]heptane hydrochloride (223 mg, 1.36 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (280 mg, 0.91 mmol), Cs2CO3(888 mg, 2.73 mmol) and Pd Catalyst [CAS: 1810068-35-9] (52 mg, 0.05 mmol) in 1,4-dioxane (15 mL) at 25° C. The resulting suspension was stirred at 100° C. for 4 h under N2(g). The solid was filtered off. The filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (200 mg, 62%) as a yellow solid; MS m/z (ESI) [M+H]+355.2. Intermediate 46: 6-(3,3-Dimethyl-1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylic acid TFA (3 mL) was added to a solution of tert-butyl 6-(3,3-dimethyl-1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylate Intermediate 45 (200 mg, 0.56 mmol) in DCM (6 mL). The reaction was stirred at rt for 15 h. The solvent was removed under reduced pressure, and the residue was dried under vacuum to give the title compound (168 mg, 100%) as a yellow solid; MS m/z (ESI) [M+H]+299.2. Intermediate 47: Methyl 6-(1-azaspiro[3.3]heptan-1-yl)quinoline-4-carboxylate 1-Azaspiro[3.3]heptane hydrochloride (301 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Pd2(dba)3(103 mg, 0.11 mmol), XPhos (161 mg, 0.34 mmol) and Cs2CO3(1.10 g, 3.38 mmol) in 1,4-dioxane (15 mL) under N2(g). The reaction was stirred at 100° C. for 15 h. The solid was filtered off. The filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (250 mg, 79%) as a yellow solid; MS m/z (ESI) [M+H]+283.2. Intermediate 48: 6-(1-Azaspiro[3.3]heptan-1-yl)quinoline-4-carboxylic acid LiOH (106 mg, 4.43 mmol) was added to a solution of methyl 6-(1-azaspiro[3.3]heptan-1-yl)quinoline-4-carboxylate Intermediate 47 (250 mg, 0.89 mmol) in MeOH (10 mL) and water (2 mL). The reaction was stirred at rt for 5 h. The solvent was removed under reduced pressure. The residue was diluted with water and adjusted to pH 6 with citric acid. The reaction mixture was extracted with EtOAc, and the organic phases were washed with water. The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (215 mg, 90%) as an orange solid; MS m/z (ESI) [M+H]+269.2. Intermediate 49: Methyl 6-(2,2-dimethylazetidin-1-yl)quinoline-4-carboxylate 2,2-Dimethylazetidine hydrochloride (147 mg, 1.21 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (215 mg, 0.81 mmol), Cs2CO3(1.05 g, 3.23 mmol) and RuPhos Pd G3 (68 mg, 0.08 mmol) in 1,4-dioxane (5 mL) at 5° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was diluted with EtOAc (10 mL) and filtered through Celite©. The filter pad was washed with EtOAc, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (196 mg, 90%) as a brown gum; MS m/z (ESI) [M+H]+271.2. Intermediate 50: 6-(2,2-Dimethylazetidin-1-yl)quinoline-4-carboxylic acid NaOH (96 mg, 2.4 mmol) in water (2 mL) was added slowly to a stirred solution of methyl 6-(2,2-dimethylazetidin-1-yl)quinoline-4-carboxylate Intermediate 49 (130 mg, 0.48 mmol) in MeOH (6 mL) cooled to 0° C. The resulting solution was stirred at 10° C. for 2 h. The reaction mixture was diluted with water (20 mL) and acidified with aq HCl (1 M). The mixture was extracted with EtOAc (5×75 mL). The organic layers were combined and washed with water (4×25 mL). The aqueous layers were combined and extracted with EtOAc (4×25 mL). All organic layers were combined, dried over Na2SO4, filtered and evaporated to afford the title compound (120 mg, 97%) as a yellow solid; MS m/z (ESI) [M+H]+257.2. Intermediate 51: Methyl 6-(3-fluoroazetidin-1-yl)quinoline-4-carboxylate 3-Fluoroazetidine hydrochloride (252 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(1.10 mg, 3.38 mmol), Pd2(dba)3(103 mg, 0.11 mmol) and XPhos (161 mg, 0.34 mmol) in 1,4-dioxane (20 mL) was stirred at 100° C. for 2 h. The precipitate was collected by filtration, washed with MeOH, and dried under vacuum. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (180 mg, 61%) as a yellow solid; MS m/z (ESI) [M+H]+261.0. Intermediate 52: 6-(3-Fluoroazetidin-1-yl)quinoline-4-carboxylic acid LiOH (83 mg, 3.5 mmol) was added to a solution of methyl 6-(3-fluoroazetidin-1-yl)quinoline-4-carboxylate Intermediate 51 (180 mg, 0.69 mmol) in MeOH (10 mL) and water (2 mL). The reaction was stirred at rt for 2 h. The reaction mixture was adjusted to pH 5 with aq HCl (1 M). The reaction mixture was diluted with water, and extracted with EtOAc. The organic layer was dried over Na2SO4, filtered and evaporated to give the title compound (150 mg, 88%) as a yellow solid; MS m/z (ESI) [M+H]+247.1. Intermediate 53: Methyl 6-(3,3-dimethylazetidin-1-yl)quinoline-4-carboxylate 3,3-Dimethylazetidine hydrochloride (274 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(1.10 mg, 3.38 mmol), Pd2(dba)3(103 mg, 0.11 mmol) and XPhos (161 mg, 0.34 mmol) in 1,4-dioxane (20 mL). The mixture was stirred at 100° C. for 4 h under N2(g). The precipitate was collected by filtration, washed with MeOH and dried under vacuum to afford the crude product, which was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (200 mg, 66%) as a yellow solid; MS m/z (ESI) [M+H]+271.10. Intermediate 54: 6-(3,3-Dimethylazetidin-1-yl)quinoline-4-carboxylic acid LiOH (89 mg, 3.7 mmol) was added to a solution of methyl 6-(3,3-dimethylazetidin-1-yl)quinoline-4-carboxylate Intermediate 53 (200 mg, 0.74 mmol) MeOH (10 mL) and water (2 mL). The reaction was stirred at rt for 2 h, and then adjusted to pH 5 with aq HCl (1 M). The reaction mixture was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (180 mg, 95%) as a yellow solid; MS m/z (ESI) [M+H]+257.2. Intermediate 55: Methyl 6-(3,3-difluoroazetidin-1-yl)quinoline-4-carboxylate 3,3-Difluoroazetidine hydrochloride (294 mg, 2.27 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (302 mg, 1.13 mmol), Cs2CO3(1.11 g, 3.40 mmol), Pd2(dba)3(104 mg, 0.11 mmol) and XPhos (108 mg, 0.23 mmol) in 1,4-dioxane (3 mL) at 10° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was diluted with water (50 mL), and extracted with EtOAc (3×50 mL). The organic phases were combined and washed with sat brine (150 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (313 mg, 99%) as a brown solid; MS m/z (ESI) [M+H]+279.2. Intermediate 56: 6-(3,3-Difluoroazetidin-1-yl)quinoline-4-carboxylic acid NaOH (224 mg, 5.61 mmol) was added to methyl 6-(3,3-difluoroazetidin-1-yl)quinoline-4-carboxylate Intermediate 55 (312 mg, 1.12 mmol) in MeOH (3 mL) and water (1 mL) at 10° C. The resulting solution was stirred at 10° C. for 1 h under N2(g). The solvent was removed under reduced pressure. The reaction mixture was diluted with water (50 mL), adjusted to pH 5 with aq HCl (1 M), and extracted with EtOAc (6×50 mL). The combined organic phases were dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC PrepMethod F (gradient: 0-50%) to afford the title compound (127 mg, 43%) as a yellow solid; MS m/z (ESI) [M+H]+265.2. Intermediate 57: Methyl 6-(3-fluoro-3-methylazetidin-1-yl)quinoline-4-carboxylate 3-Fluoro-3-methylazetidine hydrochloride (176 mg, 1.40 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (212 mg, 0.70 mmol), Cs2CO3(685 mg, 2.10 mmol), Pd2(dba)3(64 mg, 0.07 mmol) and XPhos (67 mg, 0.14 mmol) in 1,4-dioxane (3 mL) at 10° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM. The solvents were removed under reduced pressure. The residue was suspended in water (5 mL), and extracted with EtOAc (3×20 mL). The organic layers were combined and was washed with brine (20 mL). the organic phase was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (153 mg, 80%) as a brown gum; MS m/z (ESI) [M+H]+275.0. Intermediate 58: 6-(3-Fluoro-3-methylazetidin-1-yl)quinoline-4-carboxylic acid NaOH (112 mg, 2.79 mmol) was added to methyl 6-(3-fluoro-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 57 (153 mg, 0.56 mmol) in MeOH (3 mL) and water (1 mL) at 5° C. The resulting solution was stirred at 5° C. for 1 h under N2(g). The solvent was removed under reduced pressure. The reaction mixture was diluted with water and adjusted to pH 5 with aq HCl (1 M). The mixture was diluted with water (10 mL) and extracted with EtOAc (25 mL). The combined organic layers were dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC PrepMethod F (gradient: 0-50%) to afford the title compound (88 mg, 61%) as an orange solid; MS m/z (ESI) [M+H]+261.2. Intermediate 59: tert-Butyl 6-(3-methylazetidin-1-yl)quinoline-4-carboxylate 3-Methylazetidine (102 mg, 1.43 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (400 mg, 1.30 mmol), Cs2CO3(1.06 g, 3.24 mmol), Pd2(dba)3(119 mg, 0.13 mmol) and DavePhos (102 mg, 0.26 mmol) in 1,4-dioxane (2 mL). The resulting mixture was heated at 100° C. for 3 h under N2(g). The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 3:1) to afford the title compound (350 mg, 90%) as a yellow solid; MS m/z (ESI) [M+H]+299.3. Intermediate 60: 6-(3-Methylazetidin-1-yl)quinoline-4-carboxylic acid TFA (4.5 mL) was added to tert-butyl 6-(3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 59 (300 mg, 1.01 mmol) in DCM (9 mL). The resulting mixture was stirred at 40° C. for 3 h. The solvents were removed under reduced pressure to give the title compound (200 mg, 82%) as a violet solid; MS m/z (ESI) [M+H]+243.2. Intermediate 61: tert-Butyl 6-(3-(trifluoromethyl)azetidin-1-yl)quinoline-4-carboxylate 3-(Trifluoromethyl)azetidine (112 mg, 0.89 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (250 mg, 0.81 mmol), Cs2CO3(661 mg, 2.03 mmol), XPhos (77 mg, 0.16 mmol) and Pd2(dba)3(74 mg, 0.08 mmol) in 1,4-dioxane (2 mL). The resulting mixture was heated at 100° C. for 3 h under N2(g). The reaction mixture was filtered through Celite©, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (180 mg, 63%) as a yellow oil; MS m/z (ESI) [M+H]+253.2. Intermediate 62: 6-(3-(Trifluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid TFA (42 mg, 0.37 mmol) was added to tert-butyl 6-(3-(trifluoromethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 61 (130 mg, 0.37 mmol) in DCM (1.5 mL). The resulting mixture was heated at 60° C. for 5 h. The solvent was removed under reduced pressure to give the title compound (100 mg, 91%) as a violet solid; MS m/z (ESI) [M+H]+297.1. Intermediate 63: tert-Butyl 6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate KOtBu (336 mg, 3.00 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), 3-(fluoromethyl)-3-methylazetidine (206 mg, 2.00 mmol), Pd(OAc)2(150 mg, 0.67 mmol), and XantPhos (300 mg, 0.52 mmol) in 1,4-dioxane (3 mL) at 10° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (147 mg, 44%) as a brown gum; MS m/z (ESI) [M+H]+331.3. Intermediate 64: 6-(3-(Fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid TFA (3 mL) was added to tert-butyl 6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 63 (147 mg, 0.44 mmol) in DCM (3 mL) at 10° C. The resulting solution was stirred at 10° C. overnight under N2(g). The solvent was removed under reduced pressure to afford the title compound (298 mg) as a red gum; MS m/z (ESI) [M+H]+275.05. Intermediate 65: tert-Butyl 6-(3-(difluoromethyl)azetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(149 mg, 0.16 mmol) was added to 3-(difluoromethyl)azetidine (191 mg, 1.78 mmol), tert-butyl 6-bromoquinoline-4-carboxylate (500 mg, 1.62 mmol), Cs2CO3(1.06 g 3.24 mmol) and XPhos (155 mg, 0.32 mmol) in 1,4-dioxane (10 mL) at 25° C. under N2(g). The resulting mixture was stirred at 100° C. for 5 h. The reaction mixture was concentrated, and the residue was dissolved in EtOAc (125 mL), and washed sequentially with sat brine (75 mL) and water (75 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 3:1) to afford the title compound (380 mg, 70%) as a yellow solid; MS m/z (ESI) [M+H]+335.05. Intermediate 66: 6-(3-(Difluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid TFA (0.17 mL, 2.2 mmol) was added to tert-butyl 6-(3-(difluoromethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 65 (250 mg, 0.75 mmol) in DCM (5 mL) at 20° C. under air. The resulting mixture was stirred at 25° C. for 16 h. The reaction mixture was concentrated and diluted with DCM (100 mL), and washed sequentially with sat NaHCO3(25 mL), sat brine (25 mL), and water (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (DCM:MeOH, 5:1) to afford the title compound (150 mg, 72%) as a yellow solid. MS m/z (ESI) [M+H]+279.2. Intermediate 67: Ethyl 6-(3-(methoxymethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate 3-(Methoxymethyl)-3-methylazetidine hydrochloride (108 mg, 0.71 mmol) was added to a mixture of ethyl 6-bromoquinoline-4-carboxylate (100 mg, 0.36 mmol), Cs2CO3(465 mg, 1.43 mmol), XPhos (34 mg, 0.07 mmol) and Pd2(dba)3(33 mg, 0.04 mmol) in 1,4-dioxane (2 mL). The vial was sealed, purged with N2(g), and the reaction was heated at 100° C. for 2 h. After cooling to rt, water (10 mL) and DCM (10 mL) were added, the mixture was stirred, filtered through a phase separator. The phase separator was rinsed with more DCM and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 5-50% EtOAc in heptane) to give the title compound (87 mg, 78%); MS m/z (ESI) [M+H]+315.3. Intermediate 68: 6-(3-(Methoxymethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 0.51 mL) was added to ethyl 6-(3-(methoxymethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 67 (80 mg, 0.25 mmol) in MeOH (2 mL). The reaction was stirred at 50° C. for 20 min, then cooled to rt. Aq HCl (3.8 M, 0.17 mL) was added, the mixture stirred, then evaporated to give the title compound (73 mg, 100%); MS m/z (ESI) [M+H]+287.3. Intermediate 69: tert-Butyl 6-((2S,3R)-3-methoxy-2-methylazetidin-1-yl)quinoline-4-carboxylate (2S,3R)-3-Methoxy-2-methylazetidine oxalate (285 mg, 1.95 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), Cs2CO3(1.27 mg, 3.89 mmol) and Pd Catalyst [CAS: 1810068-35-9] (55 mg, 0.05 mmol) in 1,4-dioxane (20 mL) at 25° C. The resulting suspension was heated at 100° C. for 48 h under N2(g). The solid was filtered off, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (200 mg, 63%) as a yellow solid; MS m/z (ESI) [M+H]+329.2. Intermediate 70: 6-((2S,3R)-3-Methoxy-2-methylazetidin-1-yl)quinoline-4-carboxylic acid TFA (3 mL) was added to a solution of tert-butyl 6-((2S,3R)-3-methoxy-2-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 69 (200 mg, 0.61 mmol) in DCM (6 mL). The solution was stirred at rt for 4 h. The solvent was removed under reduced pressure to give the crude title compound (166 mg, 100%); MS m/z (ESI) [M+H]+273.2. Intermediate 71: Methyl 6-(3-cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxylate 3-Cyclopropyl-3-fluoroazetidine (143 mg, 1.24 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(21 mg, 0.06 mmol), Pd2(dba)3(103 mg, 0.11 mmol) and XPhos (107 mg, 0.23 mmol) in 1,4-dioxane (2 mL) at 20° C. under N2(g). The mixture was heated at 100° C. for 5 h. The reaction mixture was concentrated and the residue was diluted with EtOAc (75 mL), and washed sequentially with sat NH4Cl (25 mL), brine (25 mL), and water (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 3:1) to afford the title compound (180 mg, 53%) as a yellow solid; MS m/z (ESI) [M+H]+301.1. Intermediate 72: 6-(3-Cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (113 mg, 2.83 mmol) in water (4 mL) was added to a stirred solution of methyl 6-(3-cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxylate Intermediate 71 (170 mg, 0.57 mmol) in MeOH (12 mL) at 20° C. The resulting mixture was stirred at 25° C. for 2 h. The reaction mixture was adjusted to pH 5 with aq HCl (2 M). The reaction mixture was concentrated, and the residue was redissolved in DCM (50 mL), and washed sequentially with brine (25 mL) and water (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (130 mg, 80%) as a red solid; MS m/z (ESI) [M+H]+287.0. Intermediate 73: Methyl 6-(piperidin-1-yl)quinoline-4-carboxylate Piperidine (0.061 mL, 0.62 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (150 mg, 0.56 mmol), Cs2CO3(367 mg, 1.13 mmol), DavePhos (44 mg, 0.11 mmol) and Pd2(dba)3(52 mg, 0.06 mmol) in 1,4-dioxane (2 mL) at 16° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (5 mL). The organic layer was filtered and the solvents were evaporated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH, 30:1) to afford the title compound (109 mg, 71%) as a yellow oil; MS m/z (ESI) [M+H]+271.2. Intermediate 74: 6-(Piperidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (81 mg, 2.0 mmol) in water (1 mL) was added slowly to a stirred solution of methyl 6-(piperidin-1-yl)quinoline-4-carboxylate Intermediate 73 (109 mg, 0.40 mmol) in MeOH (4 mL) cooled to 0° C. The resulting solution was stirred at 16° C. for 1 h. The reaction mixture was diluted with water (20 mL), adjusted to pH 5 with aq HCl (2 M), and extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×25 mL), dried over Na2SO4, filtered and evaporated to afford the title compound (90 mg, 87%) as a yellow oil; MS m/z (ESI) [M+H]+257.1. Intermediate 75: Methyl 6-(4,4-dimethylpiperidin-1-yl)quinoline-4-carboxylate 4,4-Dimethylpiperidine hydrochloride (93 mg, 0.62 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (150 mg, 0.56 mmol), Cs2CO3(551 mg, 1.69 mmol), Pd2(dba)3(52 mg, 0.06 mmol) and XPhos (54 mg, 0.11 mmol) in 1,4-dioxane (3 mL) at 15° C. The resulting suspension was heated at 100° C. for 4 h under N2(g). The reaction mixture was diluted with DCM (5 mL) and filtered. The filtrate was dried over Na2SO4, filtered and evaporated to afford crude product. The residue was purified by preparative TLC (DCM:MeOH, 20:1) to afford the title compound (140 mg, 83%) as a yellow oil; MS m/z (ESI) [M+H]+299.1. Intermediate 76: 6-(4,4-Dimethylpiperidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (94 mg, 2.4 mmol) in water (1 mL) was added slowly to a stirred solution of methyl 6-(4,4-dimethylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 75 (140 mg, 0.47 mmol) in MeOH (4 mL) cooled to 0° C. The resulting solution was stirred at 15° C. for 1 h. The reaction mixture was diluted with water (20 mL), adjusted to pH 5 with aq HCl (2 M), and extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×30 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (101 mg, 76%) as a yellow solid; MS m/z (ESI) [M+H]+285.3. Intermediate 77: Methyl 6-(4-fluoro-4-methylpiperidin-1-yl)quinoline-4-carboxylate 4-Fluoro-4-methylpiperidine hydrochloride (76 mg, 0.50 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (120 mg, 0.45 mmol), Cs2CO3(441 mg, 1.35 mmol), Pd2(dba)3(41 mg, 0.05 mmol) and DavePhos (36 mg, 0.09 mmol) in 1,4-dioxane (2 mL) at 15° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (2 mL), filtered, and evaporated under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH, 20:1) to afford the title compound (102 mg, 75%) as a yellow oil; MS m/z (ESI) [M+H]+303.1. Intermediate 78: 6-(4-Fluoro-4-methylpiperidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (67 mg, 1.7 mmol) in water (1 mL) was added to a stirred solution of methyl 6-(4-fluoro-4-methylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 77 (101 mg, 0.33 mmol) in MeOH (4 mL) at 18° C. The resulting solution was stirred at 18° C. for 1 h. The reaction mixture was diluted with water (20 mL), and adjusted to pH 4 with aq HCl (2 M). The mixture was extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (65 mg, 68%) as a yellow solid; MS m/z (ESI) [M+H]+289.2. Intermediate 79: Methyl 6-(4,4-difluoropiperidin-1-yl)quinoline-4-carboxylate 4,4-Difluoropiperidine (89 mg, 0.73 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (150 mg, 0.56 mmol), Cs2CO3(551 mg, 1.69 mmol), Pd2(dba)3(52 mg, 0.06 mmol) and XPhos (54 mg, 0.11 mmol) in 1,4-dioxane (15 mL) at 20° C. The resulting mixture was stirred at 100° C. for 15 h under N2(g). The solvents were evaporated under reduced pressure and the residue was purified by preparative TLC (petroleum ether:EtOAc, 5:1) to afford the title compound (160 mg, 93%) as a pale yellow oil; MS m/z (ESI) [M+H]+307.2. Intermediate 80: 6-(4,4-Difluoropiperidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 3 mL) was added to methyl 6-(4,4-difluoropiperidin-1-yl)quinoline-4-carboxylate Intermediate 79 (150 mg, 0.49 mmol) in MeOH (10 mL) at 20° C. The resulting mixture was stirred at 20° C. for 3 h. The reaction mixture was adjusted to pH 5 with aq HCl (1 M). Solvents were evaporated to afford the title compound (140 mg, 98%) as a yellow solid; MS m/z (ESI) [M+H]+293.2. Intermediate 81: Methyl 6-(3,3-difluoropiperidin-1-yl)quinoline-4-carboxylate 3,3-Difluoropiperidine hydrochloride (98 mg, 0.62 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (150 mg, 0.56 mmol), Cs2CO3(551 mg, 1.69 mmol), Pd2(dba)3(52 mg, 0.06 mmol) and XPhos (54 mg, 0.11 mmol) in 1,4-dioxane (3 mL) at 15° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (5 mL) and filtered. The filtrate was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative TLC (DCM:MeOH, 20:1) to afford the title compound (162 mg, 94%) as a yellow gum; MS m/z (ESI) [M+H]+307.1. Intermediate 82: 6-(3,3-Difluoropiperidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (104 mg, 2.61 mmol) in water (1 mL) was added dropwise to a stirred solution of methyl 6-(3,3-difluoropiperidin-1-yl)quinoline-4-carboxylate Intermediate 81 (160 mg, 0.52 mmol) in MeOH (4 mL) cooled to 0° C. The resulting solution was stirred at 15° C. for 1 h. The reaction mixture was diluted with water (20 mL), and adjusted to pH 4 with aq HCl (2 M). The mixture was extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (143 mg, 94%) as a yellow solid; MS m/z (ESI) [M+H]+293.1. Intermediate 83: Methyl 6-(4-(fluoromethyl)-4-methylpiperidin-1-yl)quinoline-4-carboxylate 4-(Fluoromethyl)-4-methylpiperidine hydrochloride (162 mg, 0.97 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (266 mg, 0.88 mmol), Cs2CO3(859 mg, 2.64 mmol), Pd2(dba)3(81 mg, 0.09 mmol) and DavePhos (69 mg, 0.18 mmol) in 1,4-dioxane (5 mL) at 11° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (3 mL) and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (170 mg, 61%) as a yellow oil; MS m/z (ESI) [M+H]+317.3. Intermediate 84: 6-(4-(Fluoromethyl)-4-methylpiperidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (107 mg, 2.69 mmol) in water (1 mL) was added slowly to a stirred solution of methyl 6-(4-(fluoromethyl)-4-methylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 83 (170 mg, 0.54 mmol) in MeOH (3 mL) cooled to 0° C. The resulting solution was stirred at 10° C. for 1 h. The reaction mixture was diluted with water (20 mL), adjusted to pH 5 with aq HCl (2 M), and extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (129 mg, 79%) as a yellow solid; MS m/z (ESI) [M+H]+303.2. Intermediate 85: Methyl 6-(4,4-difluoro-3,3-dimethylpiperidin-1-yl)quinoline-4-carboxylate 4,4-Difluoro-3,3-dimethylpiperidine hydrochloride (230 mg, 1.24 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(1.10 g, 3.38 mmol), XPhos (107 mg, 0.23 mmol) and Pd2(dba)3(103 mg, 0.11 mmol) in 1,4-dioxane (10 mL) at 11° C. The resulting suspension was heated at 100° C. for 5 h under N2(g). The reaction mixture was filtered. The filtrate was concentrated and redissolved in EtOAc (75 mL), and washed sequentially with brine (25 mL) and water (20 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (EtOAc:petroleum ether, 1:1) to afford the title compound (310 mg, 82%) as a yellow oil; MS m/z (ESI) [M+H]+335.1. Intermediate 86: 6-(4,4-Difluoro-3,3-dimethylpiperidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (179 mg, 4.49 mmol) in water (4 mL) was added to a stirred solution of methyl-(4,4-difluoro-3,3-dimethylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 85 (300 mg, 0.90 mmol) in MeOH (12 mL) at 20° C. The resulting mixture was stirred at 20° C. for 3 h. The reaction mixture adjusted to pH 5 with aq HCl (2 M). Solvents were evaporated under reduced pressure, the residue was dissolved in EtOAc (100 mL) and washed with brine (25 mL). The organic layer was dried over Na2SO4, filtered, and evaporated to afford the title compound (250 mg, 87%) as a yellow solid; MS m/z (ESI) [M+H]+321.1. Intermediate 87: Methyl 6-(4-(trifluoromethyl)piperidin-1-yl)quinoline-4-carboxylate 4-(Trifluoromethyl)piperidine (169 mg, 1.10 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (303 mg, 1.00 mmol), Cs2CO3(0.98 g, 3.00 mmol) Pd2(dba)3(92 mg, 0.10 mmol) and DavePhos (79 mg, 0.20 mmol) in 1,4-dioxane (1 mL) at 10° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (3 mL) and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 1:1) to afford the title compound (243 mg, 72%) as a yellow oil which solidified on standing; MS m/z (ESI) [M+H]+339.1. Intermediate 88: 6-(4-(Trifluoromethyl)piperidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (142 mg, 3.55 mmol) in water (1 mL) was added slowly to a stirred solution of methyl 6-(4-(trifluoromethyl)piperidin-1-yl)quinoline-4-carboxylate Intermediate 87 (240 mg, 0.71 mmol) in MeOH (4 mL) cooled to 0° C. The resulting solution was stirred at 12° C. for 1 h. The reaction mixture was diluted with water (20 mL), and adjusted to pH 5 with aq HCl (2 M). The aqueous layer was extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (190 mg, 83%) as a brown solid; MS m/z (ESI) [M+H]+325.0. Intermediate 89: rac-Methyl (R)-6-(3-fluoropiperidin-1-yl)quinoline-4-carboxylate 3-Fluoropiperidine (64 mg, 0.62 mmol) was added to a solution of methyl 6-bromoquinoline-4-carboxylate (150 mg, 0.56 mmol), Cs2CO3(367 mg, 1.13 mmol), DavePhos (44 mg, 0.11 mmol) and Pd2(dba)3(52 mg, 0.06 mmol) in 1,4-dioxane (20 mL) at 16° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1), to afford the title compound (140 mg, 86%) as a yellow gum; MS m/z (ESI) [M+H]+289.0. Intermediate 90: rac-(R)-6-(3-Fluoropiperidin-1-yl)quinoline-4-carboxylic acid NaOH (78 mg, 1.9 mmol) in water (3.5 mL) was added to a stirred solution of methyl 6-(3-fluoropiperidin-1-yl)quinoline-4-carboxylate Intermediate 89 (140 mg, 0.49 mmol) in MeOH (14 mL) at 0° C., and then stirred for 1 h at rt. The reaction mixture was diluted with water (15 mL), adjusted to pH 5 with aq HCl (2 M), and extracted with EtOAc (3×25 mL). The combined organic phases were washed with brine (15 mL), the organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (120 mg, 90%) as an orange solid; MS m/z (ESI) [M+H]+275.2. Intermediate 91: rac-Methyl (R)-6-(3-methoxypiperidin-1-yl)quinoline-4-carboxylate 3-Methoxypiperidine (71 mg, 0.62 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (150 mg, 0.56 mmol), Cs2CO3(367 mg, 1.13 mmol), DavePhos (44 mg, 0.11 mmol) and Pd2(dba)3(52 mg, 0.06 mmol) in 1,4-dioxane (20 mL) at 16° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:2) to afford the title compound (150 mg, 89%) as a yellow gum; MS m/z (ESI) [M+H]+301.1. Intermediate 92: rac-(R)-6-(3-Methoxypiperidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (104 mg, 2.59 mmol) in water (3 mL) was added slowly to a stirred solution of methyl 6-(piperidin-1-yl)quinoline-4-carboxylate Intermediate 91 (140 mg, 0.52 mmol) in MeOH (12 mL) cooled to 0° C. The resulting solution was stirred at 16° C. for 1 h. The reaction mixture was diluted with water (20 mL), and adjusted to pH 5 with aq HCl (2 M), and extracted with EtOAc (4×50 mL). The combined organic layers were dried over Na2SO4, filtered and evaporated. The crude product was purified by reversed phase flash chromatography on a C18 column (gradient: 0-50% MeCN in water) to give the title compound (133 mg, 100%) as a yellow gum. Intermediate 93: tert-Butyl 6-(4-methoxy-4-methylpiperidin-1-yl)quinoline-4-carboxylate 4-Methoxy-4-methylpiperidine hydrochloride (105 mg, 0.63 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (150 mg, 0.49 mmol), Cs2CO3(206 mg, 0.63 mmol), XPhos (46 mg, 0.10 mmol) and Pd2(dba)3(45 mg, 0.05 mmol) in 1,4-dioxane (5 mL) at 25° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was filtered, the filtrated was concentrated under reduced pressure, and purified by preparative TLC (EtOAc:petroleum ether, 3:2) to afford the title compound (140 mg, 81%) as a yellow oil which solidified on standing; MS m/z (ESI) [M+H]+357.2. Intermediate 94: 6-(4-Methoxy-4-methylpiperidin-1-yl)quinoline-4-carboxylic acid HCl in 1,4-dioxane (4 M, 5 mL) was added slowly to a stirred solution of tert-butyl 6-(4-methoxy-4-methylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 93 (110 mg, 0.31 mmol) in 1,4-dioxane (5 mL) at 25° C. The resulting solution was stirred at 50° C. for 15 h. The solvent was removed under reduced pressure to afford the title compound (90 mg, 97%) as a red solid; MS m/z (ESI) [M+H]+301.2. Intermediate 95: tert-Butyl 6-(4-isopropoxypiperidin-1-yl)quinoline-4-carboxylate 4-Isopropoxypiperidine hydrochloride (117 mg, 0.65 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (200 mg, 0.65 mmol), Cs2CO3(634 mg, 1.95 mmol), Pd2(dba)3(59 mg, 0.06 mmol) and XPhos (91 mg, 0.19 mmol) in 1,4-dioxane (10 mL) under N2(g). The reaction was heated at 80° C. for 20 h. The solvent was removed under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 5:1) to afford the title compound (180 mg, 75%) as a yellow solid; MS m/z (ESI) [M+H]+371.1. Intermediate 96: 6-(4-Isopropoxypiperidin-1-yl)quinoline-4-carboxylic acid TFA (3 mL) was added to a solution of tert-butyl 6-(4-isopropoxypiperidin-1-yl)quinoline-4-carboxylate Intermediate 95 (120 mg, 0.32 mmol) in DCM (6 mL). The reaction was stirred at rt for 2 h. The solvent was removed under reduced pressure to give the title compound (100 mg, 98%) as a yellow solid; MS m/z (ESI) [M+H]+315.05. Intermediate 97: rac-Methyl (R)-6-(4,4-difluoro-2-methylpiperidin-1-yl)quinoline-4-carboxylate 4,4-Difluoro-2-methylpiperidine hydrochloride (387 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(1.10 g, 3.38 mmol) and Pd Catalyst [CAS: 1810068-35-9] (64 mg, 0.06 mmol) in 1,4-dioxane (20 mL) at 25° C. The resulting suspension was heated at 85° C. for 18 h under N2(g). The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (170 mg, 47%) as a yellow solid; MS m/z (ESI) [M+H]+321.2. Intermediate 98: rac-(R)-6-(4,4-Difluoro-2-methylpiperidin-1-yl)quinoline-4-carboxylic acid LiOH (64 mg, 2.65 mmol) was added to a solution of methyl 6-(4,4-difluoro-2-methylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 97 (170 mg, 0.53 mmol) in MeOH (10 mL) and water (2 mL). The reaction was stirred at rt for 2 h. The solvent was removed under reduced pressure. The reaction mixture was diluted with water, the pH was adjusted to 6 with aq HCl (2 M), and evaporated under reduced pressure. The residue was dissolved in EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (150 mg, 92%) as a yellow solid; MS m/z (ESI) [M+H]+307.1. Intermediate 99: Methyl (S)-6-(2-(fluoromethyl)piperidin-1-yl)quinoline-4-carboxylate (S)-2-(Fluoromethyl)piperidine hydrobromide (447 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(1.10 g, 3.38 mmol) and Pd Catalyst [CAS: 1810068-35-9] (64 mg, 0.06 mmol) in 1,4-dioxane (20 mL) at 25° C. The resulting suspension was heated at 100° C. for 18 h under N2(g). The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (110 mg, 32%) as a yellow solid; MS m/z (ESI) [M+H]+303.2. Intermediate 100: (S)-6-(2-(Fluoromethyl)piperidin-1-yl)quinoline-4-carboxylic acid LiOH (44 mg, 1.82 mmol) was added to a solution of methyl (S)-6-(2-(fluoromethyl)piperidin-1-yl)quinoline-4-carboxylate Intermediate 99 (110 mg, 0.36 mmol) in MeOH (5 mL) and water (1 mL). The reaction was stirred at rt for 2 h. The solvent was removed under reduced pressure. The residue was diluted with water and adjusted to pH 6 with aq HCl (2 M), and evaporated under reduced pressure. The residue was dissolved in EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (90 mg, 86%); MS m/z (ESI) [M+H]+289.2. Intermediate 101: Methyl 6-(5-azaspiro[2.5]octan-5-yl)quinoline-4-carboxylate 5-Azaspirol[2.5]octane hydrochloride (325 mg, 2.20 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (605 mg, 2.00 mmol), Cs2CO3(2.60 g, 8.00 mmol), Pd2(dba)3(183 mg, 0.20 mmol) and DavePhos (157 mg, 0.40 mmol) in 1,4-dioxane (5 mL) at 10° C. The resulting suspension was heated at 100° C. overnight under N2(g). The reaction mixture was diluted with DCM (3 mL) and filtered. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (273 mg, 46%) as a yellow oil which solidified on standing; MS m/z (ESI) [M+H]+297.05. Intermediate 102: 6-(5-Azaspiro[2.5]octan-5-yl)quinoline-4-carboxylic acid A solution of NaOH (151 mg, 3.78 mmol) in water (2 mL) was added slowly to a stirred solution of methyl 6-(5-azaspiro[2.5]octan-5-yl)quinoline-4-carboxylate Intermediate 101 (270 mg, 0.76 mmol) in MeOH (6 mL) cooled to 0° C. The resulting solution was stirred at 10° C. for 1 h. The solvent was removed under reduced pressure. The residue was dissolved with water (5 mL) and adjusted to pH 4 with aq HCl (2 M). The solvent was removed under reduced pressure. The crude product was purified by preparative HPLC PrepMethod P (gradient: 0-50%) to afford the title compound (115 mg, 54%) as an orange solid; MS m/z (ESI) [M+H]+283.1. Intermediate 103: Methyl 6-(3,3-difluoropyrrolidin-1-yl)quinoline-4-carboxylate 3,3-Difluoropyrrolidine hydrochloride (324 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(1.10 g, 3.38 mmol), Pd2(dba)3(52 mg, 0.06 mmol) and XPhos (161 mg, 0.34 mmol) in 1,4-dioxane (20 mL). The reaction was heated at 100° C. for 4 h. The precipitate was collected by filtration, washed with MeOH and dried under vacuum to afford crude product, which was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (180 mg, 55%) as a yellow solid; MS m/z (ESI) [M+H]+293.0. Intermediate 104: 6-(3,3-Difluoropyrrolidin-1-yl)quinoline-4-carboxylic acid LiOH (74 mg, 3.1 mmol) was added to a solution of methyl 6-(3,3-difluoropyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 103 (180 mg, 0.62 mmol) in MeOH (10 mL) and water (2 mL). The mixture was stirred at rt for 2 h. The reaction mixture was adjusted to pH 5 with aq HCl (2 M). The reaction mixture was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (150 mg, 88%) as a yellow solid; MS m/z (ESI) [M+H]+279.1. Intermediate 105: tert-Butyl 6-(3,3-dimethylpyrrolidin-1-yl)quinoline-4-carboxylate 3,3-Dimethylpyrrolidine hydrochloride (149 mg, 1.10 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), Cs2CO3(977 mg, 3.00 mmol), Pd2(dba)3(92 mg, 0.10 mmol), and XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (5 mL) at 8° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (3 mL). The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified preparative TLC (EtOAc:petroleum ether, 1:1) to afford the title compound (285 mg, 87%) as a brown solid; MS m/z (ESI) [M+H]+327.3. Intermediate 106: 6-(3,3-Dimethylpyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (2.5 mL, 32 mmol) was added to a stirred solution of tert-butyl 6-(3,3-dimethylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 105 (265 mg, 0.81 mmol) in DCM (5 mL) at 8° C. The resulting solution was stirred at 8° C. overnight. The solvent was removed under reduced pressure to afford the title compound (343 mg) as a dark red gum; MS m/z (ESI) [M+H]+271.1. Intermediate 107: tert-Butyl 6-(5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxylate 5-Azaspiro[2.4]heptane hydrochloride (147 mg, 1.10 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), Cs2CO3(977 mg, 3.00 mmol), Pd2(dba)3(92 mg, 0.10 mmol), and XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (5 mL) at 8° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (3 mL) and filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 1:1) to afford the title compound (243 mg, 75%) as a brown oil which solidified on standing; MS m/z (ESI) [M+H]+325.2. Intermediate 108: 6-(5-Azaspiro[2.4]heptan-5-yl)quinoline-4-carboxylic acid TFA (5.0 mL, 65 mmol) was added to a stirred solution of tert-butyl 6-(5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxylate Intermediate 107 (240 mg, 0.74 mmol) in DCM (5 mL) at 8° C. The resulting solution was stirred at 8° C. overnight. The solvent was removed under reduced pressure to afford the title compound (423 mg) as a dark red solid; MS m/z (ESI) [M+H]+269.1. Intermediate 109: tert-Butyl 6-((3R,4S)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxylate (3R,4S)-3,4-Difluoropyrrolidine hydrochloride (158 mg, 1.10 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), Cs2CO3(977 mg, 3.00 mmol), Pd2(dba)3(92 mg, 0.10 mmol), and XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (1 mL) at 5° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (3 mL). The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 2:1) to afford the title compound (263 mg, 79%) as a brown solid; MS m/z (ESI) [M+H]+335.2. Intermediate 110: 6-((3R,4S)-3,4-Difluoropyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (2.5 mL, 32 mmol) was added to a stirred solution of tert-butyl 6-((3S,4R)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 109 (233 mg, 0.70 mmol) in DCM (5 mL) at 9° C. The resulting solution was stirred at 9° C. overnight. The solvent was removed under reduced pressure to afford the title compound (350 mg) as a red dark solid; MS m/z (ESI) [M+H]+279.0. Intermediate 111: tert-Butyl (S)-6-(3-fluoropyrrolidin-1-yl)quinoline-4-carboxylate (S)-3-Fluoropyrrolidine hydrochloride (138 mg, 1.10 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), Cs2CO3(977 mg, 3.00 mmol), Pd2(dba)3(92 mg, 0.10 mmol), and XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (5 mL) at 9° C. The resulting suspension was heated at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (3 mL) and filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 1:1) to afford the title compound (246 mg, 78%) as a brown oil which solidified on standing; MS m/z (ESI) [M+H]+317.3. Intermediate 112: (S)-6-(3-Fluoropyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (4 mL, 52 mmol) was added to a stirred solution of tert-butyl (S)-6-(3-fluoropyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 111 (217 mg, 0.69 mmol) in DCM (5 mL) at 9° C. The resulting solution was stirred at 9° C. overnight. The solvent was removed under reduced pressure to afford the title compound (305 mg) as a dark red oil which solidified on standing; MS m/z (ESI) [M+H]+261.2. Intermediate 113: tert-Butyl (R)-6-(3-fluoropyrrolidin-1-yl)quinoline-4-carboxylate (R)-3-Fluoropyrrolidine hydrochloride (138 mg, 1.10 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), Cs2CO3(977 mg, 3.00 mmol), Pd2(dba)3(92 mg, 0.10 mmol), and XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (5 mL) at 8° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (3 mL) and filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 1:1) to afford the title compound (245 mg, 77%) as a brown oil which solidified on standing; MS m/z (ESI) [M+H]+317.3. Intermediate 114: (R)-6-(3-Fluoropyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (4 mL, 52 mmol) was added to a stirred solution of tert-butyl (R)-6-(3-fluoropyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 113 (230 mg, 0.73 mmol) in DCM (5 mL) at 9° C. The resulting solution was stirred at 9° C. overnight. The solvent was removed under reduced pressure to afford the title compound (447 mg) as a dark red oil which solidified on standing; MS m/z (ESI) [M+H]+261.2. Intermediate 115: Methyl 6-(hexahydrocyclopenta[c]pyrrol-2(1H)-yl)quinoline-4-carboxylate Octahydrocyclopenta[c]pyrrole hydrochloride (333 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(1.10 g, 3.38 mmol), Pd2(dba)3(103 mg, 0.11 mmol) and XPhos (161 mg, 0.34 mmol) in 1,4-dioxane (20 mL). The mixture was heated 100° C. for 4 h under N2(g). The precipitate was collected by filtration, washed with MeOH, and dried under vacuum to afford crude product, which was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (200 mg, 60%) as a yellow solid; MS m/z (ESI) [M+H]+297.1. Intermediate 116: 6-(Hexahydrocyclopenta[c]pyrrol-2(1H)-yl)quinoline-4-carboxylic acid LiOH (40 mg, 1.69 mmol) was added to a solution of methyl 6-(hexahydrocyclopenta[c]pyrrol-2(1H)-yl)quinoline-4-carboxylate Intermediate 115 (100 mg, 0.34 mmol) in MeOH (10 mL) and water (2 mL). The reaction was stirred at rt for 2 h. The reaction mixture was adjusted to pH 5 with aq HCl (2 M). The reaction mixture was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (85 mg, 89%) as a yellow solid; MS m/z (ESI) [M+H]+283.2. Intermediate 117: tert-Butyl (S)-6-(3-methylpyrrolidin-1-yl)quinoline-4-carboxylate (S)-3-Methyl-pyrrolidine hydrochloride (87 mg, 0.71 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (200 mg, 0.65 mmol), Cs2CO3(634 mg, 1.95 mmol), Pd2(dba)3(59 mg, 0.06 mmol), and XantPhos (75 mg, 0.13 mmol) in 1,4-dioxane (5 mL) at 20° C. The resulting suspension was then heated at 100° C. for 2 h under N2(g). The reaction mixture was filtered through Celite©. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (125 mg, 61%) as a brown solid; MS m/z (ESI) [M+H]+313.3. Intermediate 118: (S)-6-(3-Methylpyrrolidin-1-yl)quinoline-4-carboxylic acid HCl (4 M in 1,4-dioxane, 0.3 mL) was added to tert-butyl (S)-6-(3-methylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 117 (240 mg, 0.77 mmol) in 1,4-dioxane (5 mL) at 0° C. The reaction was stirred at 25° C. for 19 h. The solvent was removed under reduced pressure to give the title compound (197 mg); MS m/z (ESI) [M+H]+257.3. Intermediate 119: tert-Butyl (R)-6-(3-methylpyrrolidin-1-yl)quinoline-4-carboxylate (R)-3-Methyl-pyrrolidine hydrochloride (118 mg, 0.97 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (200 mg, 0.65 mmol), Cs2CO3(423 mg, 1.30 mmol), Pd2(dba)3(30 mg, 0.03 mmol), and XantPhos (38 mg, 0.06 mmol) in 1,4-dioxane (5 mL) at 20° C. The resulting suspension was then heated at 100° C. for 2 h under N2(g). The reaction mixture was filtered through Celite©. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to afford the title compound (135 mg, 66%) as a brown solid; MS m/z (ESI) [M+H]+313.4. Intermediate 120: (R)-6-(3-Methylpyrrolidin-1-yl)quinoline-4-carboxylic acid HCl (4 M in 1,4-dioxane, 0.65 mL) was added to tert-butyl (R)-6-(3-methylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 119 (270 mg, 0.86 mmol) in 1,4-dioxane (5 mL) at 0° C. The reaction was stirred at 25° C. for 19 h. The solvent was removed under reduced pressure to give the title compound (222 mg); MS m/z (ESI) [M+H]+257.2. Intermediate 121: Methyl (S)-6-(2-(trifluoromethyl)pyrrolidin-1-yl)quinoline-4-carboxylate (S)-2-(Trifluoromethyl)pyrrolidine (314 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), Cs2CO3(1.10 g, 3.38 mmol) and Pd Catalyst [CAS: 1810068-35-9] (129 mg, 0.11 mmol) in 1,4-dioxane (15 mL) at 25° C. The resulting suspension was heated at 100° C. for 18 h under N2(g). The solid was filtered off and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (150 mg, 41%) as a yellow solid; MS m/z (ESI) [M+H]+325.1. Intermediate 122: (S)-6-(2-(Trifluoromethyl)pyrrolidin-1-yl)quinoline-4-carboxylic acid LiOH (55 mg, 2.31 mmol) was added to a solution of methyl (S)-6-(2-(trifluoromethyl)pyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 121 (150 mg, 0.46 mmol) in MeOH (5 mL) and water (1 mL). The reaction was stirred at rt for 2 h. The solvent was removed under reduced pressure. The residue was diluted with water, adjusted to pH 6 with aq HCl (1 M), and extracted with EtOAc. The combined organic phases were washed with water. The organic layer was dried over Na2SO4, filtered and evaporated to afford the title compound (130 mg, 91%) as a yellow solid; MS m/z (ESI) [M+H]+311.05. Intermediate 123: tert-Butyl 6-(2,2-dimethylpyrrolidin-1-yl)quinoline-4-carboxylate 2,2-Dimethylpyrrolidine (193 mg, 1.95 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), Cs2CO3(952 mg, 2.92 mmol) and Pd Catalyst [CAS: 1810068-35-9] (55 mg, 0.05 mmol) in 1,4-dioxane (15 mL) at 25° C. The resulting suspension was stirred at 100° C. for 18 h under N2(g). The solid was filtered off and the filtrate was concentrated under vacuum. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (160 mg, 50%) as a yellow solid; MS m/z (ESI) [M+H]+327.3. Intermediate 124: 6-(2,2-Dimethylpyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (3 mL) was added to a solution of tert-butyl 6-(2,2-dimethylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 123 (150 mg, 0.46 mmol) in DCM (6 mL). The reaction was stirred at rt for 4 h. The solvent was removed under reduced pressure to give the title compound (300 mg); MS m/z (ESI) [M+H]+271.2. Intermediate 125: tert-Butyl (R)-6-(6-(fluoromethyl)-5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxylate (R)-6-(Fluoromethyl)-5-azaspiro[2.4]heptane hydrochloride (202 mg, 1.22 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (250 mg, 0.81 mmol), Cs2CO3(793 mg, 2.43 mmol) and Pd Catalyst [CAS: 1810068-35-9] (46 mg, 0.04 mmol) in 1,4-dioxane (15 mL) at 25° C. The resulting suspension was stirred at 100° C. for 4 h under N2(g). The reaction mixture was filtered, and the filtrate was concentrated under vacuum. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (180 mg, 62%) as a yellow solid; MS m/z (ESI) [M+H]+357.3. Intermediate 126: (R)-6-(6-(Fluoromethyl)-5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxylic acid TFA (3 mL) was added to a solution of tert-butyl (R)-6-(6-(fluoromethyl)-5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxylate Intermediate 125 (180 mg, 0.50 mmol) in DCM (6 mL). The reaction was stirred at rt for 5 h. The solvent was removed under reduced pressure to give the title compound (152 mg, 100%); MS m/z (ESI) [M+H]+301.2. Intermediate 127: tert-Butyl (S)-6-(2-methylpyrrolidin-1-yl)quinoline-4-carboxylate (S)-2-Methylpyrrolidine (111 mg, 1.30 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (200 mg, 0.65 mmol), Cs2CO3(634 mg, 1.95 mmol) and Pd Catalyst [CAS: 1810068-35-9] (37 mg, 0.03 mmol) in 1,4-dioxane (5 mL). The mixture was stirred under an atmosphere of N2(g) at 100° C. overnight. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 3:1) to afford the title compound (120 mg, 59%) as a yellow solid; MS m/z (ESI) [M+H]+313.3. Intermediate 128: (S)-6-(2-Methylpyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (44 mg, 0.38 mmol) was added to a solution of tert-butyl (S)-6-(2-methylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 127 (120 mg, 0.38 mmol) in DCM (3 mL). The reaction was stirred at 25° C. overnight. The solvent was removed under reduced pressure to give the title compound; MS m/z (ESI) [M+H]+257.15. Intermediate 129: Methyl (R)-6-(3-fluoroazepan-1-yl)quinoline-4-carboxylate (R)-3-Fluoroazepane hydrobromide (395 mg, 2.00 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate hydrochloride Intermediate 422 (302 mg, 1.00 mmol), Cs2CO3(976 mg, 2.99 mmol), Pd2(dba)3(150 mg, 0.16 mmol) and XPhos (150 mg, 0.31 mmol) in 1,4-dioxane (20 mL) at 10° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (2 mL) and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (195 mg, 65%) as a yellow gum; MS m/z (ESI) [M+H]+303.1. Intermediate 130: (R)-6-(3-Fluoroazepan-1-yl)quinoline-4-carboxylic acid NaOH (97 mg, 2.42 mmol) was added to methyl (R)-6-(3-fluoroazepan-1-yl)quinoline-4-carboxylate Intermediate 129 (195 mg, 0.48 mmol) in MeOH (3 mL) and water (1 mL) at 13° C. The resulting suspension was stirred at 13° C. for 1 h. The reaction mixture was acidified to pH 4 with aq HCl (1 M). The reaction mixture was diluted with water (20 mL), and extracted with EtOAc (3×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC PrepMethod P to afford the title compound (20 mg, 14%); MS m/z (ESI) [M+H]+289.2. Intermediate 131: Methyl (S)-6-(3-fluoroazepan-1-yl)quinoline-4-carboxylate (S)-3-Fluoroazepane hydrobromide (395 mg, 2.00 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate hydrochloride Intermediate 422 (302 mg, 1.00 mmol), Cs2CO3(976 mg, 2.99 mmol), Pd2(dba)3(91 mg, 0.10 mmol) and XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (5 mL) at 13° C. The resulting suspension was stirred at 100° C. overnight under N2(g). The reaction mixture was diluted with DCM (20 mL) and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (173 mg, 57%) as a yellow gum; MS m/z (ESI) [M+H]+303.1. Intermediate 132: (S)-6-(3-Fluoroazepan-1-yl)quinoline-4-carboxylic acid NaOH (80 mg, 2.00 mmol) was added to methyl (S)-6-(3-fluoroazepan-1-yl)quinoline-4-carboxylate Intermediate 131 (173 mg, 0.40 mmol) in MeOH (3 mL) and water (1 mL) 13° C. The resulting solution was stirred at 13° C. for 1 h. The reaction mixture was acidified to pH 4 with aq HCl (1 M). The reaction mixture was diluted with water (20 mL), and extracted with EtOAc (3×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC PrepMethod P to afford the title compound (84 mg, 73%) as an orange solid; MS m/z (ESI) [M+H]+289.2. Intermediate 133: tert-Butyl (R)-6-(7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylate (R)-7-Methyl-1,4-oxazepane hydrochloride (100 mg, 0.66 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (185 mg, 0.60 mmol), Cs2CO3(587 mg, 1.80 mmol), and RuPhos Pd G3 (50 mg, 0.06 mmol) in 1,4-dioxane (3 mL) at 10° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was filtered, and the filtrate was washed with water (3 mL). The aqueous layer was extracted with EtOAc (3×15 mL). The organic layers were combined and washed with water (3×5 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (155 mg, 75%) as a brown gum; MS m/z (ESI) [M+H]+343.15. Intermediate 134: (R)-6-(7-Methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylic acid TFA (5 mL, 65 mmol) was added to a stirred solution of tert-butyl (R)-6-(7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylate Intermediate 133 (136 mg, 0.40 mmol) in DCM (5 mL) at 3° C. The resulting solution was stirred at 3° C. overnight. The solvent was removed under reduced pressure to afford the title compound (265 mg) as a dark red gum. MS m/z (ESI) [M+H]+287.1. Intermediate 135: tert-Butyl (S)-6-(7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylate (S)-7-Methyl-1,4-oxazepane hydrochloride (100 mg, 0.66 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (185 mg, 0.60 mmol), Cs2CO3(587 mg, 1.80 mmol) and RuPhos Pd G3 (50 mg, 0.06 mmol) in 1,4-dioxane (3 mL) at 10° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was filtered, the filtrate was washed with water (3 mL). The aqueous layer was extracted with EtOAc (3×15 mL). The organic layers were combined and washed with water (3×5 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (166 mg, 81%) as a brown gum; MS m/z (ESI) [M+H]+343.15. Intermediate 136: (S)-6-(7-Methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylic acid TFA (5 mL, 65 mmol) added slowly to a stirred solution of tert-butyl (S)-6-(7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylate Intermediate 135 (148 mg, 0.43 mmol) in DCM (5 mL) at 3° C. The resulting solution was stirred at 3° C. overnight. The solvent was removed under reduced pressure to afford the title compound (405 mg) as a dark red gum; MS m/z (ESI) [M+H]+287.1. Intermediate 137: Methyl (S)-6-(3-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylate (S)-3-Methyl-1,4-oxazepane hydrochloride (228 mg, 1.50 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), Cs2CO3(1.47 g, 4.51 mmol), and Pd Catalyst [CAS: 1810068-35-9] (43 mg, 0.04 mmol) in 1,4-dioxane (3 mL) at 5° C. The resulting suspension was stirred at 100° C. for 2 days under N2(g). The reaction mixture was diluted with EtOAc. The solvent was removed under reduced pressure. The residue was diluted with water (50 mL), and extracted with EtOAc (3×50 mL). The organic layers were combined and washed with brine (200 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to afford the title compound (147 mg, 65%) as an orange solid; MS m/z (ESI) [M+H]+301.0. Intermediate 138: (S)-6-(3-Methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylic acid NaOH (91 mg, 2.3 mmol) was added to methyl (R)-6-(3-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylate Intermediate 137 (136 mg, 0.45 mmol) in MeOH (3 mL) and water (1 mL) at 10° C. The resulting solution was stirred at 10° C. for 1 h. The solvent was removed under reduced pressure. The residue was diluted with water (20 mL) and adjusted to pH 5 with aq aq HCl (1 M). The mixture was diluted with water (10 mL), and extracted with EtOAc (6×30 mL). The organic layers were combined, dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC PrepMethod F to afford the title compound (80 mg, 62%) as an orange solid; MS m/z (ESI) [M+H]+287.0. Intermediate 139: tert-Butyl (R)-6-(2-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylate (R)-2-Methyl-1,4-oxazepane (168 mg, 1.46 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), Cs2CO3(952 mg, 2.92 mmol), Pd2(dba)3(89 mg, 0.10 mmol) and XantPhos (113 mg, 0.19 mmol) in 1,4-dioxane (5 mL). The reaction was stirred under an atmosphere of N2(g) at 100° C. for 3 h. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc, 2:1) to afford the title compound (150 mg, 45%) as a yellow solid; MS m/z (ESI) [M+H]+343.3. Intermediate 140: (R)-6-(2-Methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylic acid TFA (50 mg, 0.44 mmol) was added to tert-butyl (R)-6-(2-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylate Intermediate 139 (150 mg, 0.44 mmol) in DCM (3 mL). The reaction was stirred at 25° C. overnight. The solvent was removed by under reduced pressure to give the title compound; MS m/z (ESI) [M+H]+287.3. Intermediate 141: Ethyl 6-(3-methoxyazetidin-1-yl)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (140 mg, 0.50 mmol), Cs2CO3(651 mg, 2.00 mmol), RuPhos Pd G4 (43 mg, 0.05 mmol), 3-methoxyazetidine hydrochloride (80 mg, 0.65 mmol) and dioxane (1.2 mL) under N2(g) was stirred at 90° C. for 5.5 h. The reaction mixture was diluted with EtOAc (5 mL), SiliaMetS® Thiol (150 mg; loading 1.4 mmol/g) was added and the mixture was stirred for 1 h. The mixture was filtered through a pad of Celite® 521, the filter pad was washed with EtOAc (12 mL) and the filtrate was concentrated. The residue was purified using preparative HPLC, PrepMethod H, (gradient: 30-70%) to give the title compound (104 mg, 0.36 mmol); MS (ESI) m/z [M+H]+287.3. Intermediate 142: 6-(3-Methoxyazetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 184 μL, 0.70 mmol) was added to a solution of ethyl 6-(3-methoxyazetidin-1-yl)quinoline-4-carboxylate Intermediate 141 (100 mg, 0.35 mmol) in MeOH (2 mL) and the reaction mixture was stirred at rt overnight. Aq HCl (3.8 M, 230 μL, 0.87 mmol) was added dropwise and the resulting mixture was concentrated and freeze-dried from a mixture of MeCN/H2O to give the crude title compound (0.102 g); MS m/z (ESI) [M+H]+259.1. Intermediate 143: Ethyl 6-morpholinoquinoline-4-carboxylate Morpholine (0.22 mL, 2.5 mmol) was added to a mixture of ethyl 6-bromoquinoline-4-carboxylate (0.355 g, 1.27 mmol), Pd(dba)2(36 mg, 0.06 mmol), RuPhos (59 mg, 0.13 mmol) and K3PO4(0.538 g, 2.53 mmol) in tert-BuOH (2.3 mL). The flask was sealed, purged with N2(g), and heated at 90° C. overnight. The reaction mixture was diluted with EtOAc, washed sequentially with water and brine. The organic layer was dried by passing through a phase separator and concentrated under reduced pressure to give the title compound (110 mg, 30%); MS m/z (ESI), [M+H]+287.2. Intermediate 144: 6-Morpholinoquinoline-4-carboxylic acid NaOH (31 mg, 0.77 mmol) was added to a solution of ethyl 6-morpholinoquinoline-4-carboxylate Intermediate 143 (110 mg, 0.38 mmol) in MeOH (4 mL), and heated at 60° C. for 2 h. The reaction mixture was cooled to rt, and aq HCl (0.023 mL) was added. The reaction mixture was concentrated under reduced pressure to give the title compound (95 mg, 96%); MS m/z (ESI), [M+H]+259.1. Intermediate 145: tert-Butyl (R)-6-(2-(fluoromethyl)morpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (0.616 g, 2 mmol), (R)-2-(fluoromethyl)morpholine hydrochloride (0.405 g, 2.60 mmol), RuPhos Pd G4 (0.170 g, 0.20 mmol), Cs2CO3(1.955 g, 6.00 mmol) and dioxane (5 mL) under N2(g) was stirred vigorously at 85-90° C. for 19 h. After cooling to rt the reaction mixture was diluted with EtOAc (8 mL) and stirred with SiliaMetS® Thiol scavenger (0.7 g; 1.4 mmol/g) at rt overnight. The reaction mixture was filtered through Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated under reduced pressure. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%) to give the title compound (0.62 g, 89%) as a yellow syrup; MS (ESI) m/z [M+H]+347.3. Intermediate 146: (R)-6-(2-(Fluoromethyl)morpholino)quinoline-4-carboxylic acid A solution of tert-butyl (R)-6-(2-(fluoromethyl)morpholino)quinoline-4-carboxylate Intermediate 145 (0.554 g, 1.60 mmol) in 90% TFA (aq, 3 mL) was stirred at 50° C. for 70 min. The volatiles were removed under reduced pressure and the residue was concentrated from heptane twice to give the crude title compound (1.09 g); MS (ESI) m/z [M+H]+291.1. Intermediate 147: Ethyl 6-((2R,6R)-2,6-dimethylmorpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), Cs2CO3(0.456 g, 1.40 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol), (2R,6R)-2,6-dimethylmorpholine hydrochloride (70 mg, 0.46 mmol) and dioxane (0.9 mL) under N2(g) was stirred at 90° C. for 4.5 h. After cooling to rt, SilaMetS® Thiol scavenger (150 mg; loading 1.4 mmol/g) was added and the mixture was stirred overnight, diluted with EtOAc (3 mL) and filtered through a pad of Celite® 521. The filter pad was washed with EtOAc (10 mL) and the filtrate was concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%) to give the title compound (83 mg, 76%) as a yellow film; MS (ESI) m/z [M+H]+315.2. Intermediate 148: 6-((2R,6R)-2,6-Dimethylmorpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M. 158 μL, 0.60 mmol) was added to a solution of ethyl 6-((2R,6R)-2,6-dimethylmorpholino)quinoline-4-carboxylate Intermediate 147 (81 mg, 0.26 mmol) in MeOH (2 mL) and the reaction was stirred at rt for 2.5 h and then at 50° C. for 30 min after which the reaction was allowed to reach rt. Aq HCl (3.8 M, 0.12 mL, 0.45 mmol) was added dropwise and the resulting mixture was concentrated under reduced pressure at rt to give the crude title compound (99 mg) as a red solid; MS (ESI) m/z [M+H]+287.3. Intermediate 149: tert-Butyl 6-((2R,6S)-2,6-dimethylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol), Cs2CO3(473 mg, 1.45 mmol), RuPhos Pd G3 (40 mg, 0.05 mmol), (2R,6S)-2,6-dimethylmorpholine (69 mg, 0.63 mmol) and dioxane (1.2 mL) under N2(g) was stirred at 85° C. for 22 h. After cooling to rt the reaction mixture was diluted with EtOAc (2 mL). SiliaMetS® Thiol (ca 150 mg; loading 1.4 mmol/g) was added and the mixture was stirred for 2 h and filtered through a pad of Celite® 521. The filter pad was washed with EtOAc (9 mL) and the combined filtrate was concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%) to give the title compound (141 mg, 85%); MS (ESI) m/z [M+H]+343.4. Intermediate 150: tert-Butyl (S)-6-(2-(fluoromethyl)morpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol), Cs2CO3(473 mg, 1.45 mmol), RuPhos Pd G3 (40 mg, 0.05 mmol) (S)-2-(fluoromethyl)morpholine hydrochloride (98 mg, 0.63 mmol) and dioxane (1.2 mL) under N2(g) was stirred at 85° C. for 19 h. After cooling to rt, the reaction mixture was diluted with EtOAc (2 mL). SilaMetS® Thiol scavenger (ca 150 mg; loading 1.4 mmol/g) was added and the mixture was stirred for 2 h and filtered through a pad of Celite® 521. The filter pad was washed with EtOAc (9 mL) and the combined filtrates were concentrated. The residue was purified by preparative HPLC PrepMethod H, (gradient: 35-75%) to give the title compound (137 mg, 82%); MS m/z (ESI) [M+H]+347.3. Intermediate 151: Ethyl (R)-6-(2-methylmorpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.280 g, 1.0 mmol), (R)-2-methylmorpholine hydrochloride (0.179 g, 1.30 mmol), RuPhos Pd G4 (0.085 g, 0.10 mmol), Cs2CO3(0.977 g, 3.00 mmol) and dioxane (2.5 mL) under N2(g) was stirred vigorously at 85-90° C. for 1 h 45 min. After cooling to rt the reaction mixture was diluted with EtOAc (5 mL) and stirred with SilaMetS® Thiol scavenger (0.5 g; loading: 1.4 mmol/g) at rt for 1.5 h. The reaction mixture was filtered through Celite® 521. The filter was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 30-70%) to give the title compound (0.248 g, 83%) as a yellow solid; MS (ESI) m/z [M+H]+301.3. Intermediate 152: (R)-6-(2-Methylmorpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 0.42 mL, 1.6 mmol) was added to a solution of ethyl (R)-6-(2-methylmorpholino)quinoline-4-carboxylate Intermediate 151 (241 mg, 0.80 mmol) in MeOH (5 mL) and the reaction was stirred at rt overnight and at 50° C. for 1 h. Additional aq NaOH (3.8 M, 106 μL, 0.40 mmol) was added and the heating was continued for 20 min. Aq HCl (3.8 M, 528 μL, 2.01 mmol) was added dropwise and the resulting mixture was concentrated under reduced pressure. The residue was concentrated from MeCN to give the crude (R)-6-(2-methylmorpholino)quinoline-4-carboxylic acid (0.309 g) as a red solid; MS (ESI) m/z [M+H]+273.1. Intermediate 153: Ethyl (R)-6-(2-(trifluoromethyl)morpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), Cs2CO3(0.456 g, 1.40 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol) and (R)-2-(trifluoromethyl)-morpholine hydrochloride (0.088 g, 0.46 mmol) in dioxane (0.9 mL) under N2(g) was stirred at 90° C. for 4.5 h. After cooling to rt SilaMetS® Thiol scavenger (0.150 g; loading 1.4 mmol/g) was added and the mixture was stirred overnight at rt. The mixture was diluted with EtOAc (3 mL) and filtered through a pad of Celite® 521. The filter pad was washed with EtOAc (10 mL) and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%) to give the title compound (0.113 g, 91%) as a yellow film; MS (ESI) m/z [M+H]+355.2. Intermediate 154: (R)-6-(2-(Trifluoromethyl)morpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 0.16 mL, 0.60 mmol) was added to a solution of ethyl (R)-6-(2-(trifluoromethyl)morpholino)quinoline-4-carboxylate Intermediate 153 (0.106 g, 0.30 mmol) in MeOH (2 mL) and the reaction was stirred at rt for 2.5 h and then at 50° C. for 30 min. The reaction was allowed to reach rt, aq HCl (3.8 M, 0.118 mL, 0.45 mmol) was added dropwise and the resulting mixture was concentrated to give the crude title compound (0.121 g) as a red solid; MS (ESI) m/z [M+H]+327.1. Intermediate 155: Ethyl (S)-6-(2-(trifluoromethyl)morpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), Cs2CO3(0.456 g, 1.40 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol) and (S)-2-(trifluoromethyl)-morpholine hydrochloride (0.088 g, 0.46 mmol) in dioxane (0.9 mL) under N2(g) was stirred at 90° C. for 4.5 h. After cooling to rt SilaMetS® Thiol scavenger (0.150 g; loading 1.4 mmol/g) was added and the mixture was stirred overnight at rt. The mixture was diluted with EtOAc (3 mL) and filtered through a pad of Celite® 521. The filter pad was washed with EtOAc (10 mL) and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%) to give the title compound (0.114 g, 92%) as a yellow film; MS (ESI) m/z [M+H]+355.2. Intermediate 156: (S)-6-(2-(Trifluoromethyl)morpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 159 μL, 0.60 mmol) was added to a solution of ethyl (S)-6-(2-(trifluoromethyl)morpholino)quinoline-4-carboxylate Intermediate 155 (0.107 g, 0.30 mmol) in MeOH (2 mL) and the reaction was stirred at rt for 2.5 h and then at 50° C. for 30 min. The reaction was allowed to reach rt, aq HCl (3.8 M, 119 μL, 0.45 mmol) was added and the resulting mixture was concentrated to give the crude title compound (0.120 g) as a red solid; MS (ESI) m/z [M+H]+327.17. Intermediate 157: Ethyl 6-((1S,4S)-2-oxa-5-azabicyclo[2.2.1]heptan-5-yl)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), (1S,4S)-2-oxa-5-azabicyclo[2.2.1]heptane (0.045 g, 0.46 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol), Cs2CO3(0.342 g, 1.05 mmol) and dioxane (0.9 mL) under N2(g) was stirred vigorously at 85-90° C. for 1 h 50 min. After cooling to rt the reaction mixture was diluted with EtOAc (3 mL) and stirred with SilaMetS® Thiol scavenger (0.15 g; 1.4 mmol/g) at rt overnight. The reaction mixture was filtered through Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 25-65%) to give the title compound (0.097 g, 93%) as a yellow solid; MS (ESI) m/z [M+H]+299.2. Intermediate 158: 6-((1S,4S)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 0.16 mL, 0.62 mmol) was added to a solution of ethyl 6-((1S,4S)-2-oxa-5-azabicyclo[2.2.1]heptan-5-yl)quinoline-4-carboxylate Intermediate 157 (0.092 g, 0.31 mmol) in MeOH (2 mL) and the reaction mixture was stirred at 50° C. for 90 min and then at rt overnight. Aq HCl (3.8 M, 0.122 mL, 0.46 mmol) was added dropwise and the resulting mixture was concentrated at rt to give the crude title compound (0.110 g) as a red solid; MS (ESI) m/z [M+H]+271.1. Intermediate 159: Ethyl 6-((1R,4R)-2-oxa-5-azabicyclo[2.2.1]heptan-5-yl)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), (1R,4R)-2-oxa-5-azabicyclo[2.2.1]heptane hydrochloride (0.062 g, 0.46 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol), Cs2CO3(0.342 g, 1.05 mmol) and dioxane (0.9 mL) under N2(g) was stirred vigorously at 85-90° C. for 85 min. After cooling to rt the reaction mixture was diluted with EtOAc (3 mL) and stirred with SilaMetS® Thiol scavenger (0.15 g; 1.4 mmol/g) at rt overnight. The mixture was filtered through Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 15-55%) to give the title compound (0.087 g, 83%) as a solid; MS (ESI) m/z [M+H]+299.3. Intermediate 160: 6-((1R,4R)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 0.15 mL, 0.56 mmol) was added to a solution of ethyl 6-((1R,4R)-2-oxa-5-azabicyclo[2.2.1]heptan-5-yl)quinoline-4-carboxylate Intermediate 159 (0.084 g, 0.28 mmol) in MeOH (2 mL) and the reaction was stirred at 50° C. for 1 h 30 min and then at rt overnight. Aq HCl (3.8 M, 0.11 mL, 0.42 mmol) was added dropwise and the resulting mixture was concentrated at rt to give the crude title compound (0.099 g) as a red solid; MS (ESI) m/z [M+H]+271.2. Intermediate 161: Ethyl 6-(6-oxa-3-azabicyclo[3.1.1]heptan-3-yl)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), Cs2CO3(0.456 g, 1.40 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol), 6-oxa-3-azabicyclo[3.1.1]heptane 4-methylbenzenesulfonate (0.125 g, 0.46 mmol) and dioxane (0.9 mL) was stirred at 90° C. for 4.5 h under N2(g). After cooling to rt SilaMetS® Thiol scavenger (0.15 g; loading 1.4 mmol/g) was added and the mixture was stirred overnight at rt, diluted with EtOAc (3 mL) and filtered through a pad of Celite® 521. The filter pad was washed with EtOAc (10 mL) and the combined filtrate was concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 20-60%) to give the compound as a yellow film; MS (ESI) m/z [M+H]+299.3. Intermediate 162: 6-(6-Oxa-3-azabicyclo[3.1.1]heptan-3-yl)quinoline-4-carboxylic acid Aq NaOH 3.8 M, 0.16 mL, 0.60 mmol) was added to a solution of ethyl 6-(6-oxa-3-azabicyclo[3.1.1]heptan-3-yl)quinoline-4-carboxylate Intermediate 161 (89 mg, 0.30 mmol) in MeOH (2 mL) and the reaction was stirred at rt overnight. 1 M TFA in MeOH (299 μL, 0.30 mmol) was added dropwise and the resulting mixture was concentrated at rt. The residue was slurried in MeCN and concentrated to give the crude title compound (0.123 g) as a red solid; MS (ESI) m/z [M+H]+271.2. Intermediate 163: Ethyl 6-((2S,6S)-2,6-dimethylmorpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), (2S,6S)-2,6-dimethylmorpholine (0.052 g, 0.46 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol), Cs2CO3(0.342 g, 1.05 mmol) and dioxane (0.9 mL) under N2(g) was stirred vigorously at 85-90° C. for 1.5 h. After cooling to rt the reaction mixture was diluted with EtOAc (3 mL), stirred with SilaMetS® Thiol scavenger (0.15 g; 1.4 mmol/g) at rt overnight and filtered through Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H (gradient: 35-75%) to give the title compound (0.109 g, 99%) as a yellow syrup; MS (ESI) m/z [M+H]+315.3. Intermediate 164: 6-((2S,6S)-2,6-Dimethylmorpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 149 μL, 0.57 mmol) was added to a solution of ethyl 6-((2S,6S)-2,6-dimethylmorpholino)quinoline-4-carboxylate Intermediate 163 (89 mg, 0.28 mmol) in MeOH (2 mL) and the reaction was stirred at rt overnight. Additional aq NaOH (3.8 M, 37 μL, 0.14 mmol) was added and the reaction mixture was stirred at 50° C. for 45 min. The reaction was allowed to reach rt, Aq HCl (3.8 M, 149 μL, 0.57 mmol) was added dropwise and the resulting mixture was concentrated and freeze-dried from MeCN/H2O to give the crude title compound (0.116 g) as a red solid; m/z (ESI) [M+H]+287.2. Intermediate 165: Ethyl 6-(8-oxa-3-azabicyclo[3.2.1]octan-3-quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), 8-oxa-3-azabicyclo[3.2.1]octane (0.051 g, 0.46 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol), Cs2CO3(0.342 g, 1.05 mmol) and dioxane (0.9 mL) under N2(g) was stirred vigorously at 85-90° C. for 1 h 50 min. After cooling to rt, the reaction mixture was diluted with EtOAc (3 mL) and stirred with SilaMetS® Thiol scavenger (0.15 g, 1.4 mmol/g) at rt overnight. The mixture was filtered through Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 25-65%) to give the title compound (0.099 g, 91%) as a yellow solid; MS (ESI) m/z [M+H]+313.2. Intermediate 166: 6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 160 μL, 0.61 mmol) was added to a solution of ethyl 6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)quinoline-4-carboxylate Intermediate 165 (95 mg, 0.30 mmol) in MeOH (2 mL) and the reaction was stirred at 50° C. for 1 h 30 min, and then at rt overnight. Aq HCl (3.8 M, 0.12 mL, 0.46 mmol) was added dropwise and the resulting mixture was concentrated at rt to give the crude title compound (0.108 g) as a red solid; MS (ESI) m/z [M+H]+285.1. Intermediate 167: Ethyl (S)-6-(2-methylmorpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), (S)-2-methylmorpholine (0.046 g, 0.46 mmol), RuPhos Pd G4 (0.030 g, 0.04 mmol), Cs2CO3(0.342 g, 1.05 mmol) and dioxane (0.9 mL) under N2(g) was stirred vigorously at 85-90° C. for 2 h. After cooling to rt, the reaction mixture was diluted with EtOAc (3 mL) and stirred with SilaMetS® Thiol scavenger (0.15 g; 1.4 mmol/g) at rt overnight. The mixture was filtered through Celite® 521, the filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H (gradient: 20-60%) to give the title compound (0.096 g, 91%) as a yellow solid; MS (ESI) m/z [M+H]+301.3. Intermediate 168: (S)-6-(2-Methylmorpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 0.16 mL, 0.61 mmol) was added to a solution of ethyl (S)-6-(2-methylmorpholino)quinoline-4-carboxylate Intermediate 167 (91 mg, 0.30 mmol) in MeOH (2 mL) and stirred at 50° C. for 1 h 30 min and then at rt overnight. Aq HCl (3.8 M, 120 μL, 0.45 mmol) was added dropwise and the resulting mixture was concentrated to give the crude title compound (0.108 g) as a red solid; MS (ESI) m/z [M+H]+273.1. Intermediate 169: Ethyl 6-((2R,3S)-2,3-dimethylmorpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.140 g, 0.5 mmol), (2R,3S)-2,3-dimethylmorpholine hydrochloride (0.099 g, 0.65 mmol), Pd Catalyst [CAS: 1810068-35-9](0.057 g, 0.05 mmol), Cs2CO3(0.489 g, 1.50 mmol) and anhydrous dioxane (1.3 mL) under N2(g) was stirred vigorously at 80-85° C. overnight. The reaction mixture was allowed to reach rt and was stirred with SilaMetS® Thiol scavenger (0.2 g; 1.4 mmol/g) overnight. The mixture was filtered (Celite® 521) and the filter pad was washed with EtOAc (10 mL). The combined filtrates were concentrated and the residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%). The compound was dissolved in DCM, washed with H2O and the organic layer was concentrated to give the title compound (0.111 g, 71%) as a yellow solid; MS (ESI) m/z [M+H]+315.3. Intermediate 170: 6-((2R,3S)-2,3-Dimethylmorpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 228 μL, 0.87 mmol) was added to a solution of ethyl 6-((2R,3S)-2,3-dimethylmorpholino)quinoline-4-carboxylate Intermediate 169 (109 mg, 0.35 mmol) in MeOH (3 mL) and the reaction mixture was stirred at 50° C. for 70 min. The reaction mixture was allowed to cool to rt, aq HCl (3.8 M, 0.18 mL, 0.69 mmol) was added dropwise and the resulting mixture was concentrated and freeze-dried from MeCN/H2O to give the crude title compound (0.137 g) as a red solid; MS (ESI) m/z [M+H]+287.2. Intermediate 171: Ethyl 6-((2S,3S)-2,3-dimethylmorpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (98 mg, 0.35 mmol), (2S,3S)-2,3-dimethylmorpholine hydrobromide (WO2014/016849) (95 mg, 0.48 mmol), Pd Catalyst [CAS: 1810068-35-9] (44 mg, 0.04 mmol), Cs2CO3(342 mg, 1.05 mmol) and anhydrous dioxane (0.9 mL) under N2(g) was stirred vigorously at 80-85° C. overnight. The reaction mixture was allowed to reach rt, diluted with EtOAc (2 mL) and stirred with SilaMetS® Thiol scavenger (0.15 g; 1.4 mmol/g) at rt overnight. The mixture was filtered, and the filter cake was washed with EtOAc. The combined filtrates were concentrated and the residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-65%) to give the title compound (0.088 g, 80%) as a yellow film; m/z (ESI) [M+H]+315.3. Intermediate 172: 6-((2S,3S)-2,3-Dimethylmorpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 182 μL, 0.69 mmol) was added to a solution of ethyl 6-((2S,3S)-2,3-dimethylmorpholino)quinoline-4-carboxylate Intermediate 171 (87 mg, 0.28 mmol) in MeOH (2.3 mL) and stirred at 50° C. for 50 min. The reaction was allowed to cool to rt, aq HCl (3.8 M, 0.146 mL, 0.55 mmol) was added dropwise and the resulting mixture was concentrated and freeze-dried from MeCN/H2O give the crude title compound (0.107 g) as a dark orange solid; MS (ESI) m/z [M+H]+287.2. Intermediate 173: Ethyl 6-((2R,3R)-2,3-dimethylmorpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (98 mg, 0.35 mmol), (2R,3R)-2,3-dimethylmorpholine hydrobromide (89 mg, 0.45 mmol), Pd Catalyst [CAS: 1810068-35-9](45 mg, 0.04 mmol), Cs2CO3(342 mg, 1.05 mmol) and dioxane (0.9 mL) under N2(g) was stirred vigorously at 80-85° C. for 17.5 h. The reaction was allowed to reach rt, diluted with EtOAc (2 mL), and was stirred with SilaMetS® Thiol scavenger (0.15 g, 1.4 mmol/g) at rt overnight. The mixture was filtered, and the filter cake was washed with EtOAc (10 mL). The combined filtrates were concentrated and the residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-70%) to give the title compound (0.088 g, 80%) as a yellow film; MS (ESI) m/z [M+H]+315.27. Intermediate 174: 6-((2R,3R)-2,3-Dimethylmorpholino)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 178 μL, 0.68 mmol) was added to a solution of ethyl 6-((2R,3R)-2,3-dimethylmorpholino)quinoline-4-carboxylate Intermediate 173 (85 mg, 0.27 mmol) in MeOH (2.3 mL) and the reaction mixture was stirred at 50° C. for 45 min. The reaction was allowed to reach rt, aq HCl (3.8 M, 142 μL, 0.54 mmol) was added dropwise and the resulting mixture was concentrated and freeze-dried from MeCN/H2O to give the crude title compound (0.111 g) as a dark orange solid; MS (ESI) m/z [M+H]+287.3. Intermediate 175: rac-tert-Butyl (R)-6-(3-(trifluoromethyl)morpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (298 mg, 0.97 mmol), Pd Catalyst [CAS: 1810068-35-9] (110 mg, 0.10 mmol), rac-(R)-3-(trifluoromethyl)morpholine hydrochloride (241 mg, 1.26 mmol), Cs2CO3(945 mg, 2.90 mmol) and dioxane (2.4 mL) under N2(g) was stirred vigorously at 80-85° C. overnight. The reaction mixture was cooled to rt, SilaMetS® Thiol scavenger (360 mg, loading 1.4 mmol/g) was added, and the mixture was stirred overnight, diluted with EtOAc and filtered (Celite® 521). The filter cake was washed with EtOAc, and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%) to give the title compound (0.26 g, 70%); MS (ESI) m/z [M+H]+383.29. Intermediate 176: rel-tert-Butyl (R)-6-(3-(trifluoromethyl)morpholino)quinoline-4-carboxylate Isomer 1 Intermediate 177: rel-tert-butyl (R)-6-(3-(trifluoromethyl)morpholino)quinoline-4-carboxylate Isomer 2 The isomers of rac-tert-butyl (R)-6-(3-(trifluoromethyl)morpholino)quinoline-4-carboxylate Intermediate 175 (259 mg, 0.68 mmol) were separated by preparative chiral SFC on a Chiralpak IC (5 μm, 250×30 mm ID) using 20% IPA/DEA 100/20 mM in CO2, 120 bar as mobile phase, to give the first eluting compound rel-tert-butyl (R)-6-(3-(trifluoromethyl)-morpholino)quinoline-4-carboxylate Isomer 1 Intermediate 176 (0.114 g, 44%) and the second eluting compound rel-tert-butyl (R)-6-(3-(trifluoromethyl)morpholino)quinoline-4-carboxylate Isomer 2 Intermediate 177 (0.122 g, 47%). Intermediate 178: rel-(R)-6-(3-(Trifluoromethyl)morpholino)quinoline-4-carboxylic acid Isomer 1 A solution of rel-tert-butyl (R)-6-(3-(trifluoromethyl)morpholino)quinoline-4-carboxylate Isomer 1 Intermediate 176 (101 mg, 0.26 mmol) in 90% TFA (aq, 0.5 mL) was heated at 50° C. for 1 h. The reaction solution was concentrated and the residue was freeze-dried from MeCN/H2O to give the crude title compound (0.150 g) as a red viscous oil; MS (ESI) m/z [M+H]+327.1. Intermediate 179: rel-(R)-6-(3-(Trifluoromethyl)morpholino)quinoline-4-carboxylic acid Isomer 2 A solution of Intermediate 177 rel-tert-butyl (R)-6-(3-(trifluoromethyl)morpholino)-quinoline-4-carboxylate Isomer 2 (108 mg, 0.28 mmol) in 90% TFA (aq, 0.5 mL) was heated at 50° C. for 1 h. The reaction solution was concentrated and the residue was freeze-dried from MeCN/H2O to give the crude title compound (0.152 g) as a red oil; MS (ESI) m/z [M+H]+327.1. Intermediate 180: tert-Butyl 6-(3-oxa-9-azabicyclo[3.3.1]nonan-9-yl)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (82 mg, 0.27 mmol), Pd Catalyst [CAS: 1810068-35-9] (30 mg, 0.03 mmol), 3-oxa-9-azabicyclo[3.3.1]nonane hydrochloride (56 mg, 0.34 mmol), Cs2CO3(260 mg, 0.80 mmol) and dioxane (0.65 mL) under N2(g) was stirred vigorously at 80-85° C. overnight. The reaction mixture was allowed to reach rt, diluted with EtOAc (3 mL) and the mixture was stirred with SilaMetS® Thiol scavenger (140 mg; 1.4 mmol/g) at rt for 2 h. The mixture was filtered through Celite® 521 and the filtrate was concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-80%) to give the title compound (0.065 g, 69%); MS (ESI) m/z [M+H]+355.4. Intermediate 181: 6-(3-Oxa-9-azabicyclo[3.3.1]nonan-9-yl)quinoline-4-carboxylic acid A solution of tert-butyl 6-(3-oxa-9-azabicyclo[3.3.1]nonan-9-yl)quinoline-4-carboxylate Intermediate 180 (56 mg, 0.15 mmol) in 90% TFA (aq, 0.5 mL) was stirred at 50° C. for 1.5 h. The reaction solution was concentrated and freeze-dried from MeCN/H2O to give the crude title compound (0.070 g); MS (ESI) m/z [M+H]+299.3. Intermediate 182: tert-Butyl 6-((2R,5R)-2,5-dimethylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol), Cs2CO3(473 mg, 1.45 mmol), Pd Catalyst [CAS: 1810068-35-9] (55 mg, 0.05 mmol), (2R,5R)-2,5-dimethylmorpholine hydrochloride (96 mg, 0.63 mmol) and dioxane (1.2 mL) under N2(g) was stirred at 85° C. for 19 h. After cooling to rt the reaction mixture was diluted with EtOAc (2 mL) and stirred with SilaMetS® Thiol scavenger (150 mg; loading 1.4 mmol/g) for 2 h. The mixture was filtered through a pad of Celite® 521, the filter pad was washed with EtOAc (9 mL) and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%) to give the title compound (0.147 g, 89%); MS (ESI) m/z [M+H]+343.3. Intermediate 183: 6-((2R,5R)-2,5-Dimethylmorpholino)quinoline-4-carboxylic acid A vial was charged with tert-butyl 6-((2R,5R)-2,5-dimethylmorpholino)quinoline-4-carboxylate Intermediate 182 (0.12 g, 0.35 mmol) and 90% TFA (aq, 0.5 mL) and heated at 50° C. for 1 h 40 min. The reaction mixture was concentrated. A mixture of H2O and MeCN was added to the residue and the resulting mixture was concentrated to give the crude title compound (0.158 g); MS (ESI) m/z [M+H]+287.2. Intermediate 184: tert-Butyl 6-(2,2-dimethylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol), Cs2CO3(473 mg, 1.45 mmol), RuPhos Pd G3 (40 mg, 0.05 mmol), 2,2-dimethylmorpholine (0.069 g, 0.63 mmol) and dioxane (1.2 mL) under N2(g) was stirred at 85° C. for 22 h. After cooling to rt the reaction mixture was diluted with EtOAc (2 mL), SilaMetS® Thiol scavenger (0.150 g; loading 1.4 mmol/g) was added and the mixture was stirred for 2 h. The mixture was filtered through a pad of Celite® 521, the filter pad was washed with EtOAc (9 mL) and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%) to give the title compound (0.136 mg, 82%); MS (ESI) m/z [M+H]+343.4. Intermediate 185: 6-(2,2-Dimethylmorpholino)quinoline-4-carboxylic acid A vial was charged with crude tert-butyl 6-(2,2-dimethylmorpholino)quinoline-4-carboxylate Intermediate 184 (0.120 g, 0.35 mmol) and 90% TFA (aq, 0.5 mL). The vial was heated at 50° C. for 1 h 40 min. The reaction mixture was concentrated. A mixture of H2O and MeCN was added to the residue and the mixture was concentrated to give the crude title compound (0.145 g); MS (ESI) m/z [M+H]+287.2. Intermediate 186: tert-Butyl (S)-6-(3-(methoxymethyl)morpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (150 mg, 0.49 mmol), (S)-3-(methoxymethyl)morpholine hydrochloride (106 mg, 0.63 mmol), Pd Catalyst [CAS: 1810068-35-9] (56 mg, 0.05 mmol), Cs2CO3(476 mg, 1.46 mmol) and dioxane (1.2 mL) was stirred vigorously at 80-85° C. for 15 h. The reaction mixture was diluted with EtOAc and filtered through Celite®. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-75% EtOAc in heptane) to give the title compound (0.146 g, 84%) as a yellow solid; MS (ESI) m/z [M+H]+359.3. Intermediate 187: tert-Butyl 6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol), Cs2CO3(473 mg, 1.45 mmol), Pd Catalyst [CAS: 1810068-35-9] (55 mg, 0.05 mmol), (3S,5R)-3,5-dimethylmorpholine hydrochloride (96 mg, 0.63 mmol) and dioxane (1.2 mL) under N2(g) was stirred at 85° C. for 60 h. After cooling to rt the reaction mixture was diluted to 3.5 mL with EtOAc and was filtered through a pad of Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-75% EtOAc in heptane) to afford the title compound (73 mg, 41%) as a yellow oil; MS (ESI) m/z [M+H]+343.3. Intermediate 188: 6-((3S,5R)-3,5-Dimethylmorpholino)quinoline-4-carboxylic acid A solution of tert-butyl 6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylate Intermediate 187 (67 mg, 0.20 mmol) in 90% TFA (aq, 1 mL) was stirred at 50° C. for 3 h. The reaction was concentrated and the residue was concentrated from a mixture of DCM and heptane and dried under vacuum to give the crude title compound (0.127 g); MS (ESI) m/z [M+H]+287.2. Intermediate 189: tert-Butyl 6-(1,4-oxazepan-4-yl)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol), Cs2CO3(473 mg, 1.45 mmol), RuPhos Pd G3 (40 mg, 0.05 mmol), 1,4-oxazepane hydrochloride (83 mg, 0.6 mmol) and dioxane (1.2 mL) under N2(g) was stirred at 85° C. for 22 h. After cooling to rt the reaction mixture was diluted with EtOAc (2 mL). SilaMetS® Thiol scavenger (ca 150 mg; loading 1.4 mmol/g) was added and the mixture were stirred for 2 h and filtered through a pad of Celite® 521. The filter pad was washed with EtOAc (9 mL) and the filtrate was concentrated. The residue was purified by preparative HPLC, PrepMethod G (gradient: 25-100%). The relevant fractions were partially concentrated to remove the MeCN and the aqueous mixture was basified with 8% NaHCO3(aq) and extracted with DCM. The organic layer was concentrated to give the title compound (0.117 g, 74%); MS (ESI) m/z [M+H]+329.3. Intermediate 190: (R)-4-Benzyl-2-((methylthio)methyl)morpholine KI (7.3 g, 44.5 mmol) was added to a solution of (R)-4-benzyl-2-(chloromethyl)morpholine (200 g, 0.89 mol) in DMF (2.0 L) followed by 18% NaSCH3(aq, 690 g, 1.78 mol) at 0° C. The mixture was stirred at rt for 12 h. The reaction mixture was poured into ice-water and extracted with EtOAc (3×500 mL). The combined organic layers were washed with water (500 mL) and brine (500 mL), dried over Na2SO4, filtered and evaporated to dryness to give the title compound (192 g, 91%) as a colorless oil; MS m/z (ESI) [M+H]+238.0. Intermediate 191: (R)-4-Benzyl-2-((methylsulfonyl)methyl)morpholine m-CPBA (494 g, 2.435 mol) was added portion wise at 10° C. to a solution of (R)-4-benzyl-2-((methylthio)methyl)morpholine Intermediate 190 (192 g, 0.812 mol) in anhydrous DCM (2 L). After addition, the mixture was stirred at rt for 16 h. The mixture was filtered and the filtrate was washed with sat NaHSO3(2×1 L), sat NaHCO3(3×1 L), and brine (800 mL), dried over Na2SO4, filtered and concentrated under vacuum. The residue was triturated by MTBE/petroleum ether (100 mL/500 mL) and the solids were collected by filtration to afford of the title compound (130 g, 60%) as a white solid; MS m/z (ESI) [M+H]+269.9. Intermediate 192: (R)-2-((Methylsulfonyl)methyl)morpholine A mixture of (R)-4-benzyl-2-((methylsulfonyl)methyl)morpholine Intermediate 191 (120 g, 0.44 mol) and Pd(OH)2/C (12 g) in MeOH (1000 mL) was hydrogenated (50 Psi H2) at 40° C. for 24 h. The resulting mixture was filtered to remove Pd(OH)2/C and the filtrate was concentrated to give the title compound (68 g, 86%) as a white solid; MS m/z (ESI) [M+H]+180.2. Intermediate 193: tert-Butyl (R)-6-(2-((methylsulfonyl)methyl)morpholino)quinoline-4-carboxylate The title compound was synthesized analogous to the procedure of Intermediate 189 starting from tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol) and (R)-2-((methylsulfonyl)methyl)morpholine Intermediate 192 (0.108 g, 0.6 mmol) to give the title compound (147 mg, 75%); MS (ESI) m/z [M+H]+407.2. Intermediate 194: tert-Butyl (S)-6-(2-(methoxymethyl)morpholino)quinoline-4-carboxylate The title compound was synthesized and purified analogous to the procedure of Intermediate 187 starting from tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol) and (S)-2-(methoxymethyl)morpholine hydrochloride (0.106 g, 0.63 mmol) with a reaction time of 20 h to give the title compound (0.16 g, 88%); MS (ESI) m/z [M+H]+359.3. Intermediate 195: (S)-4-Benzyl-2-((methylthio)methyl)morpholine KI (7.3 g, 0.045 mol) was added to a solution of (S)-4-benzyl-2-(chloromethyl)morpholine (200 g, 0.89 mol) in DMF (2.0 L) followed by 18% NaSCH3(aq, 690 g, 1.78 mol) at 0° C. and the mixture was stirred at rt for 12 h. The reaction mixture was poured into ice-water and extracted with EtOAc (3×0.5 L). The combined organic layers were washed with water (0.5 L) and brine (0.5 L), dried over Na2SO4and evaporated to dryness to give the title compound (200 g, 95%) as a colorless oil; MS m/z (ESI) [M+H]+238.0. Intermediate 196: (S)-4-Benzyl-2-((methylsulfonyl)methyl)morpholine m-CPBA (494 g, 2.44 mol) was added portion wise below 10° C. to a solution of (S)-4-benzyl-2-((methylthio)methyl)morpholine Intermediate 195 (192 g, 0.812 mol) in anhydrous DCM (2 L), and the mixture was stirred at rt for 16 h. The mixture was filtered and the filtrate was washed with sat NaHSO3(2×1 L), sat NaHCO3(3×1 L) and brine (0.8 L), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was triturated by a mixture of MTBE (100 mL) and petroleum ether (500 mL). The solids were collected by filtration to give the title compound (150 g. 68%) as a white solid; MS m/z (ESI) [M+H]+270.0. Intermediate 197: (S)-2-((Methylsulfonyl)methyl)morpholine A mixture of (S)-4-benzyl-2-((methylsulfonyl)methyl)morpholine Intermediate 196 (120 g, 0.44 mol) and Pd(OH)2/C (12 g) in MeOH (1 L) was hydrogenated (50 Psi H2) at 40° C. for 24 h. The resulting mixture was filtered to remove Pd(OH)2/C and the filtrate was concentrated to give the title compound (49 g, 62%) as white solid; MS m/z (ESI) [M+H]+180.2. Intermediate 198: tert-Butyl (S)-6-(2-((methylsulfonyl)methyl)morpholino)quinoline-4-carboxylate The title compound was synthesized and purified analogous to the procedure of Intermediate 187 starting from tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol) and (S)-2-((methylsulfonyl)methyl)morpholine Intermediate 197 (0.113 g, 0.63 mmol) with a reaction time of 20 h to give the title compound (0.111 g, 50%); MS (ESI) m/z [M+H]+407.2. Intermediate 199: (R)-4-Benzyl-3-(2-methoxyethyl)morpholine NaH (24.4 g, 0.61 mol) was added at to a solution of (R)-2-(4-benzylmorpholin-3-yl)ethan-1-ol WO 2011111875 (90 g, 0.41 mol) in THF (1.30 L) at 0° C. and the reaction was stirred for 1 h. Mel (63.6 g, 448 mmol) was added at 0° C. and stirred for 6 h. The reaction was quenched with H2O (0.80 L), extracted with EtOAc (2×1.50 L). The organic phase was dried over Na2SO4, filtered and concentrated to give the title compound (100 g, 100%); MS m/z (ESI) [M+H]+236.1. Intermediate 200: (R)-3-(2-Methoxyethyl)morpholine hydrochloride Pd(OH)2/C (25 g) was added to a solution of (R)-4-benzyl-3-(2-methoxyethyl)morpholine Intermediate 199 (100 g, 0.425 mol) in MeOH (1.0 L). The mixture was hydrogenated at 50° C. for 16 h. The reaction mixture was filtered and HCl/EtOAc was added to the filtrate. The filtrate was concentrated and the residue was triturated with EtOAc to give the title compound (52 g, 68%); MS m/z (ESI) [M+H]+145.9. Intermediate 201: tert-Butyl (R)-6-(3-(2-methoxyethyl)morpholino)quinoline-4-carboxylate The title compound was synthesized and purified analogous to the procedure of Intermediate 187 starting from tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol) and (R)-3-(2-methoxyethyl)morpholine hydrochloride Intermediate 200 (0.114 g, 0.63 mmol) with a reaction time of 20 h to give the title compound (0.151 g, 79%); MS (ESI) m/z [M+H]+373.3. Intermediate 202: (2S,3S)-4-Benzyl-3-(methoxymethyl)-2-methylmorpholine NaH (19.5 g, 814 mmol) was added to a solution of ((2S,3S)-4-benzyl-2-methylmorpholin-3-yl)methanol (WO2015/144093) (90 g, 407 mmol) in THF (1.2 L) at 0° C., and the suspension was stirred at 5° C. for 1 h. Mel (63.6 g, 448 mmol) was added at 0° C. and the reaction was stirred for 2 h. The temperature was increased to rt and the reaction was stirred overnight. The reaction was quenched by addition of H2O at 0° C. and extracted with DCM (2×2 L). The combined organic layers were concentrated to give the title compound (100 g) as an oil; MS m/z (ESI) [M+H]+235.9. Intermediate 203: (2S,3S)-3-(Methoxymethyl)-2-methylmorpholine hydrochloride Pd(OH)2/C (20 g) was added to a solution of (2S,3S)-4-benzyl-3-(methoxymethyl)-2-methylmorpholine Intermediate 202 (100 g, 407 mmol) in MeOH (1 L) under N2(g). The system was evacuated and backfilled with N2(g) (3×) and hydrogenated (50 psi) at 50° C. for 18 h. The reaction was filtered and the filtrate was concentrated. HCl in EtOAc was added to the residue and the mixture was stirred for 30 min. The solid was collected by filtration, washed with EtOAc and dried to give the title compound (57 g, 79%) as a solid; MS m/z (ESI) [M+H]+146.1. Intermediate 204: tert-Butyl 6-((2S,3S)-3-(methoxymethyl)-2-methylmorpholino)-quinoline-4-carboxylate The title compound was synthesized and purified analogous to the procedure of Intermediate 187 starting from tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol) and (2S,3S)-3-(methoxymethyl)-2-methylmorpholine hydrochloride Intermediate 203 (0.114 g, 0.63 mmol) with a reaction time of 20 h to give the title compound (0.155 g, 71%); MS (ESI) m/z [M+H]+373.4. Intermediate 205: (2R,3R)-4-Benzyl-3-(methoxymethyl)-2-methylmorpholine NaH (23.4 g, 584 mmol, 60%) was added to a solution of ((2R,3R)-4-benzyl-2-methylmorpholin-3-yl)methanol (86 g, 0.39 mol) in THF (1.5 L) at −5-0° C. and the mixture was stirred at 0° C. for 2 h. Mel (60.7 g, 428 mmol) was added dropwise and the mixture was stirred at rt overnight. The mixture was quenched with H2O and extracted with DCM (3×1 L). The combined organic layers were dried over anhydrous MgSO4, filtered and concentrated under reduced pressure to give the title compound (88 g, 86%); MS m/z (ESI) [M+H]+236.1. Intermediate 206: (2R,3R)-3-(Methoxymethyl)-2-methylmorpholine hydrochloride Pd(OH)2(20 g) was added to a solution of (2R,3R)-4-benzyl-3-(methoxymethyl)-2-methylmorpholine Intermediate 205 (86 g, 370 mmol) in MeOH (1 L). The mixture was stirred at 50° C. under H2(g) (50 Psi) overnight. The mixture was filtered, 4 M HCl in EtOAc (200 mL) was added to the filtrate and the solvent was evaporated to give the title compound (55 g, 82%) as a light yellow solid; MS m/z (ESI) [M+H]+146.0. Intermediate 207: tert-Butyl 6-((2R,3R)-3-(methoxymethyl)-2-methylmorpholino)-quinoline-4-carboxylate The title compound was synthesized analogous to the procedure of Intermediate 187 starting from tert-butyl 6-bromoquinoline-4-carboxylate (0.149 g, 0.48 mmol) and (2R,3R)-3-(methoxymethyl)-2-methylmorpholine hydrochloride Intermediate 206 (0.110 g, 0.61 mmol) with a reaction time of 14.5 h. The compound was purified by straight phase flash chromatography on silica (0-40% EtOAc in heptane) to give the title compound (0.128 g, 71%); MS (ESI) m/z [M+H]+373.4. Intermediate 208: tert-Butyl 6-(3-oxa-8-azabicyclo[3.2.1]octan-8-yl)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (200 mg, 0.65 mmol), 3-oxa-8-azabicyclo[3.2.1]octane hydrochloride (126 mg, 0.84 mmol), Pd Catalyst [CAS: 1810068-35-9] (74 mg, 0.06 mmol), Cs2CO3(634 mg, 1.95 mmol) and dioxane (1.6 mL) under N2(g) was stirred vigorously at 80-85° C. for 27 h. After cooling to rt the reaction mixture was diluted with EtOAc and filtered through Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The compound was purified by straight phase flash chromatography on silica (0-75% EtOAc in heptane) to give the title compound (0.20 g, 90%) as a yellow solid; MS (ESI) m/z [M+H]+341.3. Intermediate 209: tert-Butyl 6-(1,9-dioxa-4-azaspiro[5.5]undecan-4-yl)quinoline-4-carboxylate The title compound was synthesized analogous to the procedure of Intermediate 189 starting from tert-butyl 6-bromoquinoline-4-carboxylate (149 mg, 0.48 mmol) and 1,9-dioxa-4-azaspiro[5.5]undecane hydrochloride (0.116 g, 0.6 mmol) to give the title compound (147 mg, 75%); MS (ESI) m/z [M+H]+385.3. Intermediate 210: Ethyl 6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (160 mg, 0.57 mmol) and (3R,5S)-3,5-dimethylmorpholine hydrochloride (106 mg, 0.70 mmol), Pd Catalyst [CAS: 1810068-35-9] (49 mg, 0.04 mmol), Cs2CO3(558 mg, 1.71 mmol) and dioxane (1 mL) under N2(g) was stirred vigorously at 90° C. for 13 h. Another batch of Pd Catalyst [CAS: 1810068-35-9](20 mg, 0.02 mmol) was added and the reaction was stirred vigorously at 90° C. overnight. After cooling to rt the reaction mixture was diluted with EtOAc and filtered through Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-65% EtOAc in heptane) to give the title compound (0.070 g, 39%) as a yellow film; MS (ESI) m/z [M+H]+315.2. Intermediate 211: Ethyl 7-bromo-6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylate NBS (39 mg, 0.21 mmol) was added to a solution of ethyl 6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylate Intermediate 210 (65 mg, 0.21 mmol) in HFIP (2 mL) at 0° C. and the reaction was stirred for 1 h at 0° C. NBS (8 mg, 0.04 mmol) was added and the reaction was stirred for 10 min at 0° C. The reaction mixture was concentrated and the compound was purified by straight phase flash chromatography on silica (gradient: 0-65% EtOAc in heptane) to give the title compound (0.012 g, 15%); MS (ESI) m/z [M+H]+393.1, 395.1. Intermediate 212: 7-Bromo-6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylic acid 2 M NaOH (aq, 75 μL, 0.15 mmol) was added to a suspension of ethyl 7-bromo-6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylate Intermediate 211 (12 mg, 0.03 mmol in MeOH (200 μL). The reaction was heated at 50° C. for 25 min. The solution was cooled to rt, aq HCl (3.8 M, 39.5 μL, 0.15 mmol) was added and the resulting mixture was concentrated under a stream of N2(g). The residue was slurried in EtOAc and concentrated (3×) to give the crude title compound (0.020 g) as an orange solid; MS (ESI) m/z [M+H]+365.1, 367.1. Intermediate 213: Ethyl 5-chloro-6-morpholinoquinoline-4-carboxylate hydrochloride NCS (56 mg, 0.42 mmol) was added to a solution of ethyl 6-morpholinoquinoline-4-carboxylate Intermediate 143 (100 mg, 0.35 mmol) in MeCN (1.5 mL) at rt and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated and the residue was dissolved in EtOAc (2 mL). 4 M HCl in dioxane (87 μL, 0.35 mmol) was added and the mixture was stirred until precipitation appeared complete. The solids were collected by filtration, washed with EtOAc and dried under vacuum to give the title compound (0.103 g, 83%) as a yellow solid; MS (ESI) m/z [M+H]+321.2. Intermediate 214: tert-Butyl (S)-6-(3-methylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (104 mg, 0.34 mmol), 2′-(bis(3,5-bis(trifluoromethyl)phenyl)phosphaneyl)-3′,6′-dimethoxy-N2,N2,N6,N6-tetramethyl-[1,1′-biphenyl]-2,6-diamine (26 mg, 0.03 mmol), Pd2(dba)3(9 mg, 10 μmol), sodium 2-methylbutan-2-olate (48 mg, 0.44 mmol) and (S)-3-methylmorpholine (41 mg, 0.40 mmol) in CPME (0.7 mL) under argon was heated at 80-85° C. for 3 h. The mixture was diluted with EtOAc and washed with H2O. The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-75% EtOAc in heptane) to give the title compound (0.052 g, 47%) as a yellow film; MS (ESI) m/z [M+H]+329.4. Intermediate 215: tert-Butyl 6-((3S,5S)-3,5-dimethylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (100 mg, 0.32 mmol), 2′-(bis(3,5-bis(trifluoromethyl)phenyl)phosphaneyl)-3′,6′-dimethoxy-N2,N2,N6,N6-tetramethyl-[1,1′-biphenyl]-2,6-diamine (25 mg, 0.03 mmol), Pd2(dba)3(9 mg, 9.7 μmol), sodium 2-methylbutan-2-olate (39 mg, 0.36 mmol), (3S,5S)-3,5-dimethylmorpholine (59 mg, 0.51 mmol) and CPME (0.7 mL) under argon was stirred at 80° C. for 4 h. After cooling to rt the reaction mixture was diluted with EtOAc and washed with H2O. The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-75% EtOAc in heptane) to give the title compound (0.054 g, 48%) as a yellow film; MS (ESI) m/z [M+H]+343.4. Intermediate 216: tert-Butyl 6-(8-oxa-5-azaspiro[3.5]nonan-5-yl)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (150 mg, 0.49 mmol), 8-oxa-5-azaspiro[3.5]nonane hydrochloride (104 mg, 0.63 mmol), Pd Catalyst [CAS: 1810068-35-9](56 mg, 0.05 mmol), Cs2CO3(476 mg, 1.46 mmol) and dioxane (1.2 mL) under N2(g) was stirred vigorously at 80-85° C. for 18 h. After cooling to rt the reaction mixture was diluted with EtOAc and filtered through Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified on straight phase flash chromatography on silica (gradient: 0-65% EtOAc in heptane) to give the title compound (0.103 g, 60%) as a yellow film; MS (ESI) m/z [M+H]+355.4. Intermediate 217: tert-Butyl 6-((3R,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylate Under N2(g) a vial was charged with a stirring bar, tert-butyl 6-bromoquinoline-4-carboxylate (40 mg, 0.13 mmol), (3R,5R)-3,5-dimethylmorpholine hydrochloride (30 mg, 0.19 mmol), Pd Catalyst [CAS: 1810068-35-9] (18 mg, 15 μmol, Cs2CO3(127 mg, 0.39 mmol) and dioxane (0.6 mL). The vial was stirred vigorously at 80-85° C. for 18 h and was then allowed to cool to rt. The reaction was repeated but using (26 mg, 23 μmol) of the Pd Catalyst [CAS: 1810068-35-9]. The reaction mixtures were diluted with EtOAc and the combined mixtures were filtered through a pad of Celite® 521. The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-65% EtOAc in heptane) to give the title compound (48 mg, 54%) as a yellow oil; MS (ESI) m/z [M+H]+343.4. Intermediate 218: tert-Butyl (S)-6-(3-ethylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (87 mg, 0.28 mmol), Pd Catalyst [CAS: 1810068-35-9] (9 mg, 7.72 μmol), (S)-3-ethylmorpholine hydrochloride (86 mg, 0.56 mmol), Cs2CO3(276 mg, 0.85 mmol) and CPME (1.5 mL) under argon was stirred at 80° C. overnight. Pd Catalyst [CAS: 1810068-35-9] (17 mg, 0.01 mmol) was added and the stirring was continued at 80° C. to a reaction time of 2 days. The reaction mixture was diluted with EtOAc and washed with H2O. The aqueous layer was extracted with EtOAc and the combined organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-75% EtOAc in heptane) to give the title compound (0.050 g, 51%) as a yellow film; MS (ESI) m/z [M+H]+343.4. Intermediate 219: (S)-6-(3-Ethylmorpholino)quinoline-4-carboxylic acid A solution of tert-butyl (S)-6-(3-ethylmorpholino)quinoline-4-carboxylate Intermediate 218 (32 mg, 0.09 mmol) in 90% TFA (aq, 0.5 mL) was stirred at 25-30° C. for 4 h. The solution was concentrated under reduced pressure and the residue was concentrated from heptane (2×) to give the crude title compound (0.048 g, 99%); MS (ESI) m/z [M+H]+287.3. Intermediate 220: tert-Butyl 6-(3,3-dimethylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (100 mg, 0.32 mmol), Pd Catalyst [CAS: 1810068-35-9] (50 mg, 0.04 mmol), 3,3-dimethylmorpholine (75 mg, 0.65 mmol), Cs2CO3(211 mg, 0.65 mmol) and CPME (1.5 mL) was stirred at 80° C. under argon for 2 days. The reaction mixture was diluted with EtOAc and washed with H2O. The aqueous layer was extracted with EtOAc and the combined organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-75% EtOAc in heptane) to give the title compound (0.053 g, 48%) as a yellow film; MS (ESI) m/z [M+H]+343.3. Intermediate 221: tert-Butyl (R)-6-(3-methylmorpholino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (83 mg, 0.27 mmol), 2′-(bis(3,5-bis(trifluoromethyl)phenyl)phosphaneyl)-3′,6′-dimethoxy-N2,N2,N6,N6-tetramethyl-[1,1′-biphenyl]-2,6-diamine (31 mg, 0.04 mmol), Pd2(dba)3(12 mg, 0.01 mmol), sodium 2-methylbutan-2-olate (39 mg, 0.35 mmol), (R)-3-methylmorpholine (33 mg, 0.32 mmol) and CPME (0.6 mL) under argon was stirred at 80-85° C. for 3 h. After cooling to rt, the reaction mixture was partitioned between EtOAc and H2O. The mixture was filtered and the layers were allowed to separate. The aqueous layer was extracted with EtOAc (2×) and the combined organic layer was passed through a phase separator and concentrated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-75% EtOAc in heptane) to give the title compound (0.067 g, 76%) as a yellow film; MS (ESI) m/z [M+H]+329.3. Intermediate 222: Ethyl 2-methyl-6-morpholinoquinoline-4-carboxylate Pd(dba)2(16 mg, 0.03 mmol), RuPhos (25 mg, 0.05 mmol) and K3PO4(231 mg, 1.09 mmol) was added to ethyl 6-bromo-2-methylquinoline-4-carboxylate (160 mg, 0.54 mmol), in tert-BuOH (3 mL). Morpholine (47 mg, 0.54 mmol) was added, and the reaction flask was fitted with a rubber septum, purged with N2(g) and heated at 90° C. overnight. The reaction mixture was diluted with EtOAc and washed with H2O and brine. The organic layer was evaporated and the residue was purified by preparative HPLC, PrepMethod E, (gradient: 15-55%) to give the title compound (89 mg, 55%); MS (ESI) m/z (M+H)+301.3. Intermediate 223: tert-Butyl 6-(2-oxo-1-oxa-3-azaspiro[5.5]undecan-3-yl)quinoline-4-carboxylate Step a) tert-Butyl 6-((2-(1-hydroxycyclohexyl)ethyl)amino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (0.200 g, 0.65 mmol), XantPhos Pd G4 (0.062 g, 0.06 mmol), Cs2CO3(0.634 g, 1.95 mmol), 1-(2-aminoethyl)-cyclohexan-1-ol hydrochloride (0.152 g, 0.84 mmol) and anhydrous dioxane (3.25 mL) under N2(g) was stirred vigorously at 85° C. for 3 h. The vial was allowed to reach rt, SilaMetS® Thiol scavenger (300 mg; loading: 1.4 mmol/g) was added and the mixture was stirred overnight. The mixture was diluted with EtOAc and H2O and the resulting mixture was filtered. The layers of the filtrate were separated and the organic layer was dried over Na2SO4, filtered and concentrated to give crude tert-butyl 6-((2-(1-hydroxycyclohexyl)ethyl)amino)-quinoline-4-carboxylate. Step b) tert-Butyl 6-(2-oxo-1-oxa-3-azaspiro[5.5]undecan-3-yl)quinoline-4-carboxylate Triphosgene (0.148 g, 0.5 mmol) was added to a stirred mixture of the crude tert-butyl 6-((2-(1-hydroxycyclohexyl)ethyl)amino)quinoline-4-carboxylate from Step a) and DIPEA (0.453 mL, 2.60 mmol) in EtOAc (12 mL) at rt. The reaction mixture was stirred at rt for 10 min and was then heated at 120° C. for 20 min. DIPEA (0.113 mL, 0.65 mmol) was added and the reaction was heated at 120° C. for 5 min. The reaction mixture was diluted with EtOAc and washed with H2O and 8% NaHCO3(aq), passed through a phase separator and concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%). The purified product was dissolved in DCM, washed with H2O and concentrated to give the title compound (0.12 g, 47%) as a pale yellow solid; MS (ESI) m/z [M+H]+397.5. Intermediate 224: tert-Butyl 6-((2-(1-hydroxycyclopentyl)ethyl)amino)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (0.200 g, 0.65 mmol), XantPhos Pd G4 (0.062 g, 0.06 mmol), Cs2CO3(0.634 g, 1.95 mmol), 1-(2-aminoethyl)-cyclopentan-1-ol (170 mg, 1.32 mmol) and anhydrous dioxane (3.25 mL) under N2(g) was stirred vigorously at 80-85° C. for 75 min and was then allowed to reach rt. SilaMetS® Thiol scavenger (300 mg; loading 1.4 mmol/g) was added and the mixture was stirred for 1.5 h, diluted with EtOAc and filtered (Celite® 521). The filter pad was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod H, (gradient: 35-75%). Pure fractions were partially concentrated to remove most of the MeCN and the resulting aqueous mixture was extracted with DCM and the phases were separated using a phase separator. The organic layer was concentrated to give the title compound (0.163 g, 71%) as a yellow viscous oil; MS (ESI) m/z [M+H]+357.4. Intermediate 225: tert-Butyl 6-(7-oxo-6-oxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxylate Triphosgene (74 mg, 0.25 mmol) was added to a stirred mixture of tert-butyl 6-((2-(1-hydroxycyclopentyl)ethyl)amino)quinoline-4-carboxylate Intermediate 224 (159 mg, 0.41 mmol) and DIPEA (289 μL, 1.66 mmol) in anhydrous DCM (4.5 mL) at rt. The reaction mixture was stirred at rt for 1.5 h and was then heated at 120° C. for 15 min. The solution was concentrated and the residue was purified by preparative HPLC, PrepMethod H, (gradient: 30-70%). The purified product was dissolved in DCM and washed with H2O. The aqueous layer was extracted with DCM (×4) and the combined organic layers were concentrated to give the title compound (0.106 g, 67%) as a pale yellow foam; MS (ESI) m/z [M+H]+383.4. Intermediate 226: tert-Butyl 6-(6,6-dimethyl-2-oxo-1,3-oxazinan-3-yl)quinoline-4-carboxylate A mixture of tert-butyl 6-bromoquinoline-4-carboxylate (280 mg, 0.91 mmol), 6,6-dimethyl-1,3-oxazinan-2-one (WO2013/050454) (176 mg, 1.36 mmol), Pd2(dba)3(25 mg, 0.03 mmol), XPhos (26 mg, 0.05 mmol) and Cs2CO3(592 mg, 1.82 mmol) in 1,4-dioxane (5 mL) under N2(g) was stirred at 100° C. for 24 h. The solvent was removed under reduced pressure and the residue was purified by preparative TLC (eluent: petroleum ether, EtOAc 1:1), to afford the title compound (0.040 g, 12%) as a white solid; MS m/z (ESI) [M+H]+357.25. Intermediate 227: tert-Butyl 6-(3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxylate Cs2CO3(977 mg, 3.00 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), 3-(fluoromethyl)azetidine hydrochloride (251 mg, 2.00 mmol), Pd2(dba)3(92 mg, 0.10 mmol) and DavePhos (79 mg, 0.20 mmol) in 1,4-dioxane (3 mL) at 13° C. The resulting suspension was stirred at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM. The solvent was removed under reduced pressure and the residue was purified by preparative TLC (petroleum ether:EtOAc, 1:1) to give the title compound (0.282 g, 89%) as a brown gum; MS m/z (ESI) [M+H]+317. Intermediate 228: 6-(3-(Fluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid TFA (5 mL) was added to tert-butyl 6-(3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 227 (274 mg, 0.87 mmol) in DCM (5 mL) at 10° C. The resulting solution was stirred at 10° C. overnight. The solvent was removed under reduced pressure to afford the crude title compound (0.466 g); MS m/z (ESI) [M+H]+261. Intermediate 229: tert-Butyl 6-(4,5,6,7-tetrahydro-1H-indazol-1-yl)quinoline-4-carboxylate Intermediate 230: tert-Butyl 6-(4,5,6,7-tetrahydro-2H-indazol-2-yl)quinoline-4-carboxylate Cs2CO3(1057 mg, 3.24 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (500 mg, 1.62 mmol), 4,5,6,7-tetrahydro-1H-indazole (297 mg, 2.43 mmol) and EPhos Pd G4 (149 mg, 0.16 mmol) in 1,4-dioxane (10 mL) at 15° C. The resulting suspension was stirred at 100° C. for 16 h under N2(g). The mixture was filtered through a pad of Celite® and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 2:1) followed by preparative HPLC, PrepMethod F, (gradient: 70-90%), to give the first eluting compound tert-butyl 6-(4,5,6,7-tetrahydro-1H-indazol-1-yl)quinoline-4-carboxylate Intermediate 229 (0.040 g, 7%) as a pale yellow solid; MS m/z (ESI) [M+H]+350;1H NMR (300 MHz, CDCl3) δ 9.01 (d, 1H), 8.92 (d, 1H), 8.25 (d, 1H), 8.12 (dd, 1H), 7.91 (d, 1H), 7.57 (s, 1H), 2.92 (t, 2H), 2.65 (t, 2H), 1.89 (t, 4H), 1.70 (s, 9H). and the second eluting compound tert-butyl 6-(4,5,6,7-tetrahydro-2H-indazol-2-yl)quinoline-4-carboxylate Intermediate 230 (0.245 g, 43%) as a white solid; MS m/z (ESI) [M+H]+350;1H NMR (300 MHz, CDCl3) δ 9.01 (d, 1H), 8.92 (d, 1H), 8.25 (d, 1H), 8.12 (dd, 1H), 7.91 (d, 1H), 7.57 (s, 1H), 2.92 (t, 2H), 2.65 (t, 2H), 1.89 (t, 4H), 1.70 (s, 9H). The configuration of the regioisomers were assigned by NOESY NMR Intermediate 231: tert-Butyl 6-(5-methyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxylate Cs2CO3(529 mg, 1.62 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (250 mg, 0.81 mmol), 5-methyl-3-(trifluoromethyl)-1H-pyrazole (183 mg, 1.22 mmol) and EPhos Pd G4 (75 mg, 0.08 mmol) in 1,4-dioxane (5 mL) at 10° C. under N2(g). The resulting suspension was stirred at 100° C. for 15 h. The reaction mixture was filtered through Celite® and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 2:1) followed by preparative HPLC, PrepMethod F, (gradient: 60-85%), to give the title compound (0.133 g, 43%) as a white solid; MS m/z (ESI) [M+H]+378. Intermediate 232: 6,6-Dimethyl-2,4,5,6-tetrahydrocyclopenta[c]pyrazole hydrochloride Hydrazine hydrate (5.3 g, 106 mmol) was added dropwise with stirring at 25° C. over 10 min to stirred (5E)-5-[(dimethylamino)methylidene]-2,2-dimethylcyclopentan-1-one (18 g, 108 mmol) in a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of N2(g). EtOH (180 mL, 3.10 mol) was added and the resulting solution was heated at reflux overnight in an oil bath. The reaction mixture was cooled to 25° C. The resulting mixture was concentrated under vacuum. The residue was dissolved in 200 mL of ether and the HCl salt of the title compound was precipitated (12.6 g, 68%) as an off-white solid; MS m/z (ESI) [M+H]+137.1. Intermediate 233: tert-Butyl 6-(6,6-dimethyl-5,6-dihydrocyclopenta[c]pyrazol-2(4H)-yl)quinoline-4-carboxylate Cs2CO3(793 mg, 2.43 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (250 mg, 0.81 mmol), 6,6-dimethyl-2,4,5,6-tetrahydrocyclopenta[c]pyrazole hydrochloride Intermediate 232 (210 mg, 1.22 mmol) and EPhos Pd G4 (75 mg, 0.08 mmol) in 1,4-dioxane (1 mL) at 15° C. The resulting suspension was stirred at 100° C. for 16 h under N2(g). The reaction mixture was filtered through Celite® and the filter cake was washed with DCM (10 mL). The filtrate was concentrated under reduced pressure and the residue was purified by preparative TLC (EtOAc:petroleum ether, 2:1), to give the title compound (0.268 g, 91%) as a yellow solid; MS m/z (ESI) [M+H]+364. Intermediate 234: 6-(3-(Trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxylic acid Cs2CO3(776 mg, 2.38 mmol) was added to 6-bromoquinoline-4-carboxylic acid (200 mg, 0.79 mmol), 3-(trifluoromethyl)-1H-pyrazole (162 mg, 1.19 mmol) and Cu2O (11 mg, 0.08 mmol) in DMF (5 mL) at 10° C. The resulting suspension was stirred at 120° C. for 20 h under N2(g). The reaction mixture was filtered through Celite®. The reaction mixture was adjusted to pH≈6 with aq HCl (2 M) and the mixture was filtered through Celite® again. The filtrate was purified by preparative HPLC, PrepMethod C, (gradient: 40-60%) to give the title compound (0.088 g, 36%) as a white solid; MS m/z (ESI) [M+H]+308. Intermediate 235: 6-(4,6-Difluoro-1H-indol-1-yl)quinoline-4-carboxylic acid CuI (15 mg, 0.08 mmol) was added to a solution of 6-bromoquinoline-4-carboxylic acid (200 mg, 0.79 mmol), 4,6-difluoro-1H-indole (243 mg, 1.59 mmol) and K2CO3(219 mg, 1.59 mmol) in DMF (10 mL) under N2(g). The reaction was stirred at 150° C. for 15 h. The solvent was removed under reduced pressure. The reaction mixture was diluted with H2O (15 mL) and the reaction mixture was adjusted to pH 6 with aq HCl (1 M). The precipitate was collected by filtration and washed with H2O (30 mL) to provide a brown solid. The solid was dissolved in DMF (15 mL) and filtered. The filtrate was concentrated under vacuum to give the crude title compound (0.20 g, 78%) as a brown solid; MS m/z (ESI) [M+H]+325. Intermediate 236: 6-(5-Fluoro-1H-indol-1-yl)quinoline-4-carboxylic acid Cs2CO3(238 mg, 0.73 mmol) was added to tert-butyl 6-bromoquinoline-4-carboxylate (150 mg, 0.49 mmol), 5-fluoro-1H-indole (99 mg, 0.73 mmol) and Cu2O (7 mg, 0.05 mmol) in dry DMF (5 mL) at 10° C. The resulting suspension was stirred at 120° C. for 19 h under N2(g). The reaction mixture was adjusted to pH≈6 with aq HCl (1 M), and filtered through a Celite® pad. The filtrate was purified by preparative HPLC, PrepMethod F, (gradient: 45-70%) to give the title compound (0.035 g, 23%) as a pale yellow solid; MS m/z (ESI) [M+H]+307. Intermediate 237: 6-(3-Methyl-1H-pyrrol-1-yl)quinoline-4-carboxylic acid K2CO3(123 mg, 0.89 mmol) was added to 6-bromoquinoline-4-carboxylic acid (150 mg, 0.60 mmol), 3-methyl-1H-pyrrole (78 μL, 0.89 mmol) and CuI (11 mg, 0.06 mmol) in dry DMF (3 mL) at 10° C. The resulting suspension was stirred at 150° C. for 15 h under N2(g). The reaction mixture was adjusted to pH≈6 with aq HCl (1 M). The mixture was filtered through a Celite® pad. The filtrate was purified by preparative HPLC, PrepMethod D, (gradient: 50-65%), to give the title compound (0.050 g, 33%) as a grey solid; MS m/z (ESI) [M+H]+253. Intermediate 238: Ethyl 6-(3-morpholinoazetidin-1-yl)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (140 mg, 0.50 mmol), Cs2CO3(651 mg, 2.00 mmol), RuPhos Pd G4 (43 mg, 0.05 mmol) and 4-(azetidin-3-yl)morpholine hydrochloride (116 mg, 0.65 mmol) and dioxane (1.2 mL) under N2(g) was stirred at 90° C. for 5.5 h. The reaction mixture was diluted with EtOAc (5 mL). SilaMetS® Thiol scavenger (150 mg; loading 1.4 mmol/g) was added and the mixture was stirred for 1 h and then filtered through a pad of Celite® 521. The filter pad was washed with EtOAc (12 mL) and the combined filtrates were concentrated. The residue was purified by preparative reversed phase HPLC, PrepMethod H, (gradient: 15-65%) to give the title compound (0.141 g, 83%) as a yellow solid; MS (ESI) m/z [M+H]+342.3. Intermediate 239: 6-(3-Morpholinoazetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (3.8 M, 0.220 mL, 0.84 mmol) was added to a solution of ethyl 6-(3-morpholinoazetidin-1-yl)quinoline-4-carboxylate Intermediate 238 (143 mg, 0.42 mmol) in MeOH (2 mL) and the reaction was stirred at rt overnight. Aq HCl (3.8 M, 0.220 mL, 0.84 mmol) was added dropwise and the resulting mixture was concentrated under reduced pressure at rt. The residue was concentrated under reduced pressure from MeCN to give the crude title compound (0.176 g); MS (ESI) m/z [M+H]+314.3. Intermediate 240: 6-(4,5,6,7-Tetrahydro-1H-indazol-1-yl)quinoline-4-carboxylic acid TFA (4 mL) was added slowly to a stirred solution of tert-butyl 6-(4,5,6,7-tetrahydro-1H-indazol-1-yl)quinoline-4-carboxylate Intermediate 229 (36 mg, 0.10 mmol) in DCM (4 mL) at 15° C. The resulting solution was stirred at 15° C. for 15 h. The solvent was removed under reduced pressure to afford the crude title compound (0.063 g) as a beige solid; MS m/z (ESI) [M+H]+294. Intermediate 241: 6-(4,5,6,7-Tetrahydro-2H-indazol-2-yl)quinoline-4-carboxylic acid TFA (5 mL) was added slowly to a stirred solution of tert-butyl 6-(4,5,6,7-tetrahydro-2H-indazol-2-yl)quinoline-4-carboxylate Intermediate 230 (239 mg, 0.68 mmol) in DCM (5 mL) at 15° C. The resulting solution was stirred at 15° C. for 15 h. The solvent was removed under reduced pressure to afford the crude title compound (0.527 g) as a yellow oil which solidified on standing; MS m/z (ESI) [M+H]+294. Intermediate 242: 6-(5-Methyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxylic acid TFA (5 mL) was added to a stirred solution of tert-butyl 6-(5-methyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxylate Intermediate 231 (130 mg, 0.34 mmol) in DCM (5 mL) at 15° C. The resulting solution was stirred at 15° C. for 18 h. The solvent was removed under reduced pressure to afford the crude title compound (0.230 g) as a brown gum; MS m/z (ESI) [M+H]+322. Intermediate 243: 6-(6,6-Dimethyl-5,6-dihydrocyclopenta[c]pyrazol-2(4H)-yl)quinoline-4-carboxylic acid TFA (5 mL) was added to a stirred solution of tert-butyl 6-(6,6-dimethyl-5,6-dihydrocyclopenta[c]pyrazol-2(4H)-yl)quinoline-4-carboxylate Intermediate 233 (260 mg, 0.72 mmol) in DCM (5 mL) at 15° C. The resulting solution was stirred at 15° C. for 15 h. The solvent was removed under reduced pressure to give the crude title compound (0.552 g) as a yellow oil; MS m/z (ESI) [M+H]+308. Intermediate 244: 6-(5,5-Dimethyl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-4-carboxylic acid 1,4-Dioxane (3 mL) and water (0.75 mL) were added to a mixture of 6-bromoquinoline-4-carboxylic acid (100 mg, 0.40 mmol), 5,5-dimethyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole (114 mg, 0.44 mmol), Cs2CO3(323 mg, 0.99 mmol) and Pd(dtbpf)Cl2(26 mg, 0.04 mmol). The reaction mixture was purged with N2(g) and then stirred at rt overnight. DMSO (3 mL) was added to the reaction mixture and it was concentrated under reduced pressure. The residue was purified by preparative HPLC, PrepMethod E, (gradient: 5-45%) to give the title compound (74 mg, 61%) as a light yellow solid; MS (ESI) m/z [M+H]+308.3. Intermediate 245: 6-(2-Fluoropyridin-4-yl)quinoline-4-carboxylic acid The title compound was prepared as described for Intermediate 244 using 6-bromoquinoline-4-carboxylic acid (100 mg, 0.40 mmol) and (2-fluoropyridin-4-yl)boronic acid (61 mg, 0.44 mmol). The crude product was purified by preparative HPLC, PrepMethod E, (gradient: 0-40%) to give the title compound (5 mg, 5%) as white solid; MS (ESI) m/z [M+H]+269.2. Intermediate 246: 6-(5-Fluoropyridin-2-yl)quinoline-4-carboxylic acid 1,4-Dioxane (3 mL) and water (0.75 mL) were added to a mixture of 6-bromoquinoline-4-carboxylic acid (100 mg, 0.40 mmol), (5-fluoropyridin-2-yl)boronic acid (67 mg, 0.48 mmol), Cs2CO3(323 mg, 0.99 mmol) and Pd(dtbpf)Cl2(26 mg, 0.04 mmol). The reaction mixture was purged with N2(g) and then stirred at rt overnight. (5-Fluoropyridin-2-yl)boronic acid (67 mg, 0.48 mmol), Pd(dtbpf)Cl2(26 mg, 0.04 mmol) and Cs2CO3(323 mg, 0.99 mmol) were added to the reaction mixture and it was purged with N2(g), and heated at 100° C. overnight in a microwave reactor. DMSO was added to the reaction mixture (2 mL) and it was concentrated under reduced pressure. The residue was purified by preparative HPLC, PrepMethod E, (gradient: 0-40%) to give the title compound (6 mg, 6%) as white solid; MS (ESI) m/z [M+H]+269.2. Intermediate 247: Methyl 6-(3-((trifluoromethoxy)methyl)azetidin-1-yl)quinoline-4-carboxylate Cs2CO3(255 mg, 0.78 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (160 mg, 0.60 mmol), 3-((trifluoromethoxy)methyl)azetidine hydrochloride (150 mg, 0.78 mmol), Pd2(dba)3(55 mg, 0.06 mmol) and XPhos (57 mg, 0.12 mmol) in 1,4-dioxane (1 mL) at 30° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 3:2), to give the title compound (0.20 g, 98%) as a brown oil which solidified on standing; MS (ESI) m/z [M+H]+431. Intermediate 248: 6-(3-((Trifluoromethoxy)methyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (109 mg, 2.72 mmol) in water (2 mL) was added slowly to a stirred solution of methyl 6-(3-((trifluoromethoxy)methyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 247 (185 mg, 0.54 mmol) in MeOH (6 mL), cooled to 0° C., and the resulting mixture was stirred at 30° C. for 1 h. The reaction mixture was diluted with water (20 mL), pH was adjusted to 6 with aq HCl (2 M), and extracted with EtOAc (8×75 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure, to give the title compound (0.175 g, 99%) as an orange solid; MS (ESI) m/z [M+H]+327. Intermediate 249: Methyl 6-(3-methyl-3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Cs2CO3(255 mg, 0.78 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (160 mg, 0.60 mmol), 3-methyl-3-(2,2,2-trifluoroethyl)azetidine hydrochloride (148 mg, 0.78 mmol), Pd2(dba)3(55 mg, 0.06 mmol) and XPhos (57 mg, 0.12 mmol) in 1,4-dioxane (1 mL) at 30° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 3:2), to give the title compound (0.199 g, 98%) as a brown oil which solidified on standing; MS (ESI) m/z [M+H]+339. Intermediate 250: 6-(3-Methyl-3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (109 mg, 2.73 mmol) in water (2 mL) was added slowly to a stirred solution of methyl 6-(3-methyl-3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 249 (185 mg, 0.55 mmol) in MeOH (6 mL) cooled to 0° C. and the reaction mixture was stirred at 30° C. for 1 h. The reaction mixture was diluted with water (20 mL), pH was adjusted to 6 with aq HCl (2 M), and extracted with EtOAc (8×75 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.173 g, 98%) as a yellow solid; MS (ESI) m/z [M+H]+325. Intermediate 251: Methyl 6-(3-(trifluoromethoxy)azetidin-1-yl)quinoline-4-carboxylate Cs2CO3(478 mg, 1.47 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), 3-(trifluoromethoxy)azetidine hydrochloride (260 mg, 1.47 mmol), Pd2(dba)3(103 mg, 0.11 mmol) and XPhos (107 mg, 0.23 mmol) in 1,4-dioxane (10 mL) at 35° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 2:1) to give the title compound (0.354 g, 96%) as a brown oil which solidified on standing; MS (ESI) m/z [M+H]+327. Intermediate 252: 6-(3-(Trifluoromethoxy)azetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (201 mg, 5.03 mmol) in water (2 mL) was added to a stirred solution of methyl 6-(3-(trifluoromethoxy)azetidin-1-yl)quinoline-4-carboxylate Intermediate 251 (328 mg, 1.01 mmol) in MeOH (6 mL) cooled to 0° C., and the reaction mixture was stirred at 37° C. for 1 h. The reaction mixture was diluted with water (20 mL), the pH was adjusted to 6 with aq HCl (2 M), and extracted with EtOAc (6×75 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.30 g, 96%) as a yellow solid; MS (ESI) m/z [M+H]+313. Intermediate 253: Methyl 6-(3-(2,2-difluoroethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(302 mg, 0.93 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (190 mg, 0.71 mmol), 3-(2,2-difluoroethyl)-3-methylazetidine hydrochloride (159 mg, 0.93 mmol), Pd2(dba)3(65 mg, 0.07 mmol) and XPhos (68 mg, 0.14 mmol) in 1,4-dioxane (10 mL) at 28° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 3:2) followed by purification on reversed phase flash chromatography on a C18 column, (gradient: 0-8% MeCN in water) to give the title compound (0.203 g) as a tan gum; MS (ESI) m/z [M+H]+321. Intermediate 254: 6-(3-(2,2-Difluoroethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (97 mg, 2.4 mmol) in water (2 mL) was added slowly to a stirred solution of methyl 6-(3-(2,2-difluoroethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 253 (173 mg) in MeOH (6 mL), cooled to 0° C., and the reaction mixture was stirred at 28° C. for 1 h. The reaction mixture was diluted with water (20 mL), pH was adjusted to 6 with aq HCl (2 M), and extracted with EtOAc (4×100 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure give the title compound (0.14 g, 94%) as an orange solid; MS (ESI) m/z [M+H]+307. Intermediate 255: Methyl 6-(3-cyclopropyl-3-methylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(207 mg, 0.64 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (130 mg, 0.49 mmol), 3-cyclopropyl-3-methylazetidine (71 mg, 0.64 mmol), Pd2(dba)3(45 mg, 0.05 mmol) and XPhos (47 mg, 0.10 mmol) in 1,4-dioxane (10 mL) at 30° C., and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 3:2), to give the title compound (0.14 g, 97%) as a brown gum; MS (ESI) m/z [M+H]+297. Intermediate 256: 6-(3-Cyclopropyl-3-methylazetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (74 mg, 1.9 mmol) in water (2 mL) was added to a stirred suspension of methyl 6-(3-cyclopropyl-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 255 (110 mg, 0.37 mmol) in MeOH (6 mL) cooled to 0° C., and the reaction mixture was stirred at 23° C. for 1 h. The reaction mixture was diluted with water (15 mL), pH was adjusted to 6 with aq HCl (2 M), and extracted with EtOAc (4×75 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.10 g, 95%) as a yellow solid; MS (ESI) m/z [M+H]+283. Intermediate 257: Methyl 6-(3-(difluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(207 mg, 0.64 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (130 mg, 0.49 mmol), 3-(difluoromethyl)-3-methylazetidine hydrochloride (100 mg, 0.64 mmol), Pd2(dba)3(45 mg, 0.05 mmol) and XPhos (47 mg, 0.10 mmol) in 1,4-dioxane (10 mL) at 30° C., and the reaction mixture was stirred at 100° C. for 4 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 6:5), to give the title compound (0.145 g, 97%) as a brown gum; MS (ESI) m/z [M+H]+307. Intermediate 258: 6-(3-(Difluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid NaOH (78 mg, 2.0 mmol) was added to a stirred solution of methyl 6-(3-(difluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 257 (120 mg, 0.39 mmol) in MeOH (8 mL) and water (2 mL) at 0° C., and the reaction mixture was stirred at 23° C. for 1 h. The reaction mixture was diluted with water (15 mL), and pH was adjusted to 6 with aq HCl (2 M), and extracted with EtOAc (6×75 mL). The combined organic layer was, dried over Na2SO4, filtered and evaporated at reduced pressure, to give the title compound (0.11 g, 96%) as a yellow solid; MS (ESI) m/z [M+H]+293. Intermediate 259: Methyl 6-(3-(difluoromethoxy)azetidin-1-yl)quinoline-4-carboxylate Cs2CO3(398 mg, 1.22 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (250 mg, 0.94 mmol), 3-(difluoromethoxy)azetidine hydrochloride (195 mg, 1.22 mmol), Pd2(dba)3(86 mg, 0.09 mmol) and XPhos (90 mg, 0.19 mmol) in 1,4-dioxane (10 mL) at 28° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 3:2) followed by reversed phase flash chromatography on a C18 column (gradient: 0-4% MeCN in water) to give the title compound (0.281 g) as a brown gum; MS (ESI) m/z [M+H]+309. Intermediate 260: 6-(3-(Difluoromethoxy)azetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (154 mg, 3.85 mmol) in water (3 mL) was added slowly to a stirred solution of crude methyl 6-(3-(difluoromethoxy)azetidin-1-yl)quinoline-4-carboxylate Intermediate 259 (260 mg) in MeOH (9 mL) cooled to 0° C., and the reaction mixture was stirred at 28° C. for 1 h. The reaction mixture was diluted with water (20 mL), pH was adjusted to 6 with aq HCl (2 M), and extracted with EtOAc (8×100 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.207 g, 91%) as an orange solid; MS (ESI) m/z [M+H]+295. Intermediate 261: Methyl 6-(3-ethyl-3-methylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(276 mg, 0.85 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (150 mg, 0.56 mmol), 3-ethyl-3-methylazetidine hydrochloride (115 mg, 0.85 mmol), Pd2(dba)3(52 mg, 0.06 mmol) and XPhos (54 mg, 0.11 mmol) in 1,4-dioxane (10 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 6 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:1), to give the title compound (0.157 g, 98%) as a brown oil which solidified on standing; MS (ESI) m/z [M+H]+285. Intermediate 262: 6-(3-Ethyl-3-methylazetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (98 mg, 2.5 mmol) in water (2 mL) was added slowly to a stirred suspension of methyl 6-(3-ethyl-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 261 (140 mg, 0.49 mmol) in MeOH (6 mL) cooled to 0° C., and the reaction mixture was stirred at 30° C. for 1 h. The reaction mixture was diluted with water (15 mL), pH was adjusted to 6 with aq HCl (2 M), and extracted with EtOAc (6×75 mL). The combined organic layer was, dried over Na2SO4, filtered and evaporated at reduced pressure, to give the title compound (0.13 g, 98%) as an orange solid; MS (ESI) m/z [M+H]+271. Intermediate 263: Methyl 6-(3-ethyl-3-fluoroazetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(138 mg, 0.15 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (400 mg, 1.50 mmol), 3-ethyl-3-fluoroazetidine (186 mg, 1.80 mmol), Cs2CO3(980 mg, 3.01 mmol) and XPhos (143 mg, 0.30 mmol) in 1,4-dioxane (15 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was concentrated and diluted with EtOAc (125 mL), and washed sequentially with sat brine (75 mL) and water (75 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (MeOH:DCM, 1:5), to give the title compound (0.20 g, 46%) as a yellow solid; MS (ESI) m/z [M+H]+289. Intermediate 264: 6-(3-Ethyl-3-fluoroazetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (104 mg, 2.60 mmol) in water (4 mL) was added to a stirred solution of methyl 6-(3-ethyl-3-fluoroazetidin-1-yl)quinoline-4-carboxylate Intermediate 263 (150 mg, 0.52 mmol) in MeOH (12 mL) at 20° C., and the reaction mixture was stirred at 25° C. for 2 h. The pH of the reaction mixture was adjusted to 4 using aq HCl (2 M, 7 mL). The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (100 mL), and washed sequentially with sat brine (50 mL) and water (50 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.12 g, 84%) as a red solid; MS (ESI) m/z [M+H]+275.0. Intermediate 265: Methyl 6-(2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylate Pd2(dba)3(103 mg, 0.11 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), 2-azaspirol[3.4]octane (150 mg, 1.35 mmol), Cs2CO3(735 mg, 2.25 mmol) and XPhos (107 mg, 0.23 mmol) in 1,4-dioxane (15 mL) at 20° C., and the reaction mixture was stirred at 100° C. for 5 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (125 mL), and washed sequentially with water (75 mL) and sat brine (75 mL). The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:2), to give the title compound (0.20 g, 60%) as a yellow solid; MS (ESI) m/z [M+H]+397.0. Intermediate 266: 6-(2-Azaspiro[3.4]octan-2-yl)quinoline-4-carboxylic acid A solution of NaOH (135 mg, 3.37 mmol) in water (3 mL) was added to a stirred solution of methyl 6-(2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylate Intermediate 265 (200 mg, 0.67 mmol) in MeOH (9 mL) at 20° C., and the reaction mixture was stirred at 25° C. for 2 h. The pH of the reaction mixture was adjusted to 4 using aq HCl (2 M, 25 mL). The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (100 mL), and washed sequentially with water (25 mL) and sat brine (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.15 g, 79%) as a red solid; MS (ESI) m/z [M+H]+283.3. Intermediate 267: Methyl 6-(3-(2,2-difluoropropyl)azetidin-1-yl)quinoline-4-carboxylate XPhos (107 mg, 0.23 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), 3-(2,2-difluoropropyl)azetidine (183 mg, 1.35 mmol), Pd2(dba)3(103 mg, 0.11 mmol) and Cs2CO3(735 mg, 2.25 mmol) in 1,4-dioxane (15 mL) at 20° C., and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (100 mL), and washed sequentially with water (25 mL) and sat brine (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:3), to give the title compound (0.25 g, 69%) as a yellow oil; MS (ESI) m/z [M+H]+321.0. Intermediate 268: 6-(3-(2,2-Difluoropropyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (181 mg, 4.53 mmol) in water (3 mL) was added to a stirred solution of methyl 6-(3-(2,2-difluoropropyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 267 (290 mg, 0.91 mmol) in MeOH (9 mL) at 20° C., and the reaction mixture was stirred at 25° C. for 2 h. The pH was adjusted to 5 with aq HCl (2 M). The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (125 mL), and washed sequentially with sat brine (75 mL) and water (75 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.20 g, 72%) as a yellow solid; MS (ESI) m/z [M+H]+307.0. Intermediate 269: Methyl 6-(5,5-difluoro-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylate Pd2(dba)3(55 mg, 0.06 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (160 mg, 0.60 mmol), 5,5-difluoro-2-azaspiro[3.4]octane (106 mg, 0.72 mmol), Cs2CO3(24 mg, 0.08 mmol) and XPhos (57 mg, 0.12 mmol) in 1,4-dioxane (15 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (125 mL), and washed sequentially with sat brine (75 mL) and water (75 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:3), to give the title compound (0.15 g, 75%) as a yellow solid; MS (ESI) m/z [M+H]+333.0. Intermediate 270: 6-(5,5-Difluoro-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylic acid A solution of NaOH (120 mg, 3.01 mmol) in water (3 mL) was added to a stirred solution of methyl 6-(5,5-difluoro-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylate Intermediate 269 (200 mg, 0.60 mmol) in MeOH (9 mL) at 20° C. and the reaction mixture was stirred at 25° C. for 3 h. The pH of the reaction mixture was adjusted to 5 with aq HCl (2 M). The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (100 mL), and washed sequentially with sat brine (20 mL) and water (20 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.15 g, 78%) as a red solid; MS (ESI) m/z [M+H]+319.0. Intermediate 271: Methyl 6-(3-(3,3,3-trifluoropropyl)azetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(43 mg, 0.05 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (250 mg, 0.94 mmol), 3-(3,3,3-trifluoropropyl)azetidine hydrochloride (267 mg, 1.41 mmol), XPhos (67 mg, 0.14 mmol) and Cs2CO3(918 mg, 2.82 mmol) in 1,4-dioxane (10 mL) and the reaction mixture was stirred at 100° C. for 2 h. The solid was filtered off, and the filtrate was concentrated under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.25 g, 79%) as an orange gum; MS (ESI) m/z [M+H]+339. Intermediate 272: 6-(3-(3,3,3-Trifluoropropyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of methyl 6-(3-(3,3,3-trifluoropropyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 271 (230 mg, 0.68 mmol) and LiOH (49 mg, 2.0 mmol) in MeOH (10 mL) and water (2 mL) was stirred at rt for 2 h. The solvent was removed under reduced pressure, and the reaction mixture was diluted with water. The pH of the reaction mixture was adjusted to 6 with aq HCl (0.1M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.20 g, 91%) as an orange solid; MS (ESI) m/z [M+H]+325. Intermediate 273: Methyl 6-(3-fluoro-3-(trifluoromethyl)azetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(34 mg, 0.04 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 3-fluoro-3-(trifluoromethyl)azetidine hydrochloride (202 mg, 1.13 mmol), XPhos (54 mg, 0.11 mmol) and Cs2CO3(735 mg, 2.25 mmol) in 1,4-dioxane (10 mL) and the reaction mixture was stirred at 100° C. for 2 h. The solid was filtered off and the filtrate was concentrated under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.20 g, 81%) as a yellow solid; MS (ESI) m/z [M+H]+329. Intermediate 274: 6-(3-Fluoro-3-(trifluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of methyl 6-(3-fluoro-3-(trifluoromethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 273 (200 mg, 0.61 mmol) and LiOH (44 mg, 1.83 mmol) in MeOH (8 mL) and water (2 mL) was stirred at rt for 2 h. The solvent was removed under reduced pressure and the reaction mixture was diluted with water. The pH of the reaction mixture was set to 6 with aq HCl (1 M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.17 g, 89%) as a yellow solid; MS (ESI) m/z [M+H]+315. Intermediate 275: Methyl 6-(3-(2,2-difluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(43 mg, 0.05 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (250 mg, 0.94 mmol), 3-(2,2-difluoroethyl)azetidine 2,2,2,-trifluoroacetate (247 mg, 1.13 mmol), XPhos (67 mg, 0.14 mmol) and Cs2CO3(918 mg, 2.82 mmol) in 1,4-dioxane (10 mL) and the reaction mixture was stirred at 100° C. for 2 h. The solid was filtered off, and the filtrate was concentrated under reduced pressure. The residue was diluted with EtOAc, and washed sequentially with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.23 g, 80%) as an orange gum; MS (ESI) m/z [M+H]+307. Intermediate 276: 6-(3-(2,2-Difluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of methyl 6-(3-(2,2-difluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 275 (230 mg, 0.75 mmol) and LiOH (54 mg, 2.3 mmol) in MeOH (10 mL) and water (2 mL) was stirred at rt for 2 h. The solvent was removed under reduced pressure and the reaction mixture was diluted with water. The pH of the reaction mixture was adjusted to 6 with aq HCl (1 M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.19 g, 87%) as an orange solid; MS (ESI) m/z [M+H]+293. Intermediate 277: Methyl 6-(3-cyclopropylazetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(43 mg, 0.05 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (250 mg, 0.94 mmol), 3-cyclopropylazetidine hydrochloride (151 mg, 1.13 mmol), XPhos (67 mg, 0.14 mmol) and Cs2CO3(918 mg, 2.82 mmol) in 1,4-dioxane (10 mL) and the reaction mixture was stirred at 100° C. for 2 h. The solid was filtered off, and the filtrate was concentrated under reduced pressure. The residue was diluted with EtOAc, and washed sequentially with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.20 g, 75%) as an orange solid; MS (ESI) m/z [M+H]+283. Intermediate 278: 6-(3-Cyclopropylazetidin-1-yl)quinoline-4-carboxylic acid A solution of methyl 6-(3-cyclopropylazetidin-1-yl)quinoline-4-carboxylate Intermediate 277 (200 mg, 0.71 mmol) and LiOH (51 mg, 2.1 mmol) in MeOH (10 mL) and water (2 mL) was stirred at rt for 2 h. The solvent was removed under reduced pressure and the reaction mixture was diluted with water. The pH of the reaction mixture was adjusted to 6 with aq HCl (1 M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.17 g, 89%) as an orange solid; MS (ESI) m/z [M+H]+269. Intermediate 279: Methyl 6-(3-(2-fluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(34 mg, 0.04 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 3-(2-fluoroethyl)azetidine 2,2,2-trifluoroacetate (197 mg, 0.98 mmol), XPhos (54 mg, 0.11 mmol) and Cs2CO3(735 mg, 2.25 mmol) in 1,4-dioxane (12 mL) and the reaction mixture was stirred at 100° C. for 2 h. The solid was filtered off and the filtrate was concentrated under reduced pressure. The residue was diluted with EtOAc, and washed sequentially with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.17 g, 78%) as a yellow solid; MS (ESI) m/z [M+H]+289. Intermediate 280: 6-(3-(2-Fluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of methyl 6-(3-(2-fluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 279 (170 mg, 0.59 mmol) and LiOH (42 mg, 1.77 mmol) in MeOH (8 mL) and water (2 mL) was stirred at rt for 2 h. The solvent was removed under reduced pressure and the reaction mixture was diluted with water. The pH of the reaction mixture was adjusted to 6 with aq HCl (1 M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.145 g, 90%) as a yellow solid; MS (ESI) m/z [M+H]+275. Intermediate 281: Methyl 6-(3-(1,1-difluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(34 mg, 0.04 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 3-(1,1-difluoroethyl)azetidine hydrochloride (142 mg, 0.90 mmol), XPhos (54 mg, 0.11 mmol) and Cs2CO3(735 mg, 2.3 mmol) in 1,4-dioxane (10 mL) and the reaction mixture was stirred at 100° C. for 2 h. The solid was filtered off and the filtrate was concentrated under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.18 g, 78%) as a yellow solid; MS (ESI) m/z [M+H]+307. Intermediate 282: 6-(3-(1,1-Difluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of methyl 6-(3-(1,1-difluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 281 (180 mg, 0.59 mmol) and LiOH (42 mg, 1.8 mmol) in MeOH (8 mL) and water (2 mL) was stirred at rt for 3 h. The solvent was removed under reduced pressure, and the reaction mixture was diluted with water. The pH of the reaction mixture was adjusted to 6 with aq HCl (1 M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.155 g, 90%) as a yellow solid; MS (ESI) m/z [M+H]+293. Intermediate 283: Methyl 6-(3-isopropylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(765 mg, 2.35 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (250 mg, 0.94 mmol), 3-isopropylazetidine hydrochloride (255 mg, 1.88 mmol), Pd2(dba)3(86 mg, 0.09 mmol) and CPhos (410 mg, 0.94 mmol) in 1,4-dioxane (5.0 mL) at 25° C. and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was diluted with DCM (10 mL) and the solvent was removed under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:1), to give the title compound (0.262 g, 98%) as an orange gum; MS (ESI) m/z [M+H]+285. Intermediate 284: 6-(3-Isopropylazetidin-1-yl)quinoline-4-carboxylic acid NaOH (181 mg, 4.52 mmol) was added to methyl 6-(3-isopropylazetidin-1-yl)quinoline-4-carboxylate Intermediate 283 (257 mg, 0.90 mmol) in MeOH (9 mL) and water (3 mL) at 25° C. and the reaction mixture was stirred at 25° C. for 1 h. The solvent was removed under reduced pressure and the residue was diluted with water (50 mL). The pH of the reaction mixture was adjusted to 3 with aq HCl (1 M) and extracted with EtOAc (3×50 mL), The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.242 g, 99%) as an orange solid; MS (ESI) m/z [M+H]+271. Intermediate 285: Methyl 6-(6-methyl-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Cs2CO3(612 mg, 1.88 mmol) was added to methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 6-methyl-2-azaspiro[3.3]heptane hydrochloride (222 mg, 1.50 mmol), Pd2(dba)3(69 mg, 0.08 mmol) and SPhos (62 mg, 0.15 mmol) in 1,4-dioxane (5 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was diluted with DCM (10 mL) and the solvent was removed under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:1), to give the title compound (0.22 g, 99%) as an orange solid; MS (ESI) m/z [M+H]+297. Intermediate 286: 6-(6-Methyl-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid NaOH (145 mg, 3.63 mmol) was added to a solution of methyl 6-(6-methyl-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Intermediate 285 (215 mg, 0.73 mmol) in MeOH (6 mL) and water (2 mL) at 25° C. and the reaction solution was stirred at 25° C. for 1 h. The solvent was removed under reduced pressure, the residue was diluted with water (50 mL) and pH was adjusted to 3 with aq HCl (1 M). The aqueous phase was extracted with EtOAc (3×50 mL), the combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.20 g, 98%) as an orange solid; MS (ESI) m/z [M+H]+283. Intermediate 287: Methyl 6-(6-(trifluoromethyl)-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Cs2CO3(612 mg, 1.88 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 6-(trifluoromethyl)-2-azaspiro[3.3]heptane hydrochloride (303 mg, 1.50 mmol), Pd2(dba)3(69 mg, 0.08 mmol) and SPhos (62 mg, 0.15 mmol) in 1,4-dioxane (5 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was diluted with DCM (10 mL), and the solvent was removed under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:1), to give the title compound (0.26 g, 99%) as a yellow solid; MS (ESI) m/z [M+H]+351. Intermediate 288: 6-(6-(Trifluoromethyl)-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid NaOH (148 mg, 3.70 mmol) was added to a solution of methyl 6-(6-(trifluoromethyl)-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Intermediate 287 (259 mg, 0.74 mmol) in MeOH (9 mL) and water (3 mL) at 25° C. and the reaction mixture was stirred at 25° C. for 1 h. The solvent was removed under reduced pressure and the reaction mixture was diluted with water (50 mL) and pH was adjusted to 3 with aq HCl (1 M). The aqueous phase was extracted with EtOAc (3×50 mL), the combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.24 g, 97%) as an orange solid; MS (ESI) m/z [M+H]+337. Intermediate 289: Methyl 6-(3-methoxy-3-methylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(918 mg, 2.82 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), 3-methoxy-3-methylazetidine hydrochloride (310 mg, 2.25 mmol), Pd2(dba)3(103 mg, 0.11 mmol) and RuPhos (105 mg, 0.23 mmol) in 1,4-dioxane (5 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 2 h under N2(g). The reaction mixture was diluted with DCM (10 mL), and the solvent was removed under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:1), to give the title compound (0.321 g, 99%) as a brown gum; MS (ESI) m/z [M+H]+287. Intermediate 290: 6-(3-Methoxy-3-methylazetidin-1-yl)quinoline-4-carboxylic acid NaOH (223 mg, 5.59 mmol) was added to a solution of methyl 6-(3-methoxy-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 289 (320 mg, 1.12 mmol) in MeOH (6 mL) and water (2 mL) at 25° C., and the reaction mixture was stirred at 25° C. for 1 h. The solvent was removed under reduced pressure, the reaction mixture was diluted with water (50 mL) and pH was adjusted to 3 with aq HCl (1 M). The aqueous phase was extracted with EtOAc (3×50 mL), the combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.296 g, 97%) as an orange solid; MS (ESI) m/z [M+H]+273. Intermediate 291: Methyl 6-(3-ethoxy-3-methylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(918 mg, 2.82 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), 3-ethoxy-3-methylazetidine hydrochloride (342 mg, 2.25 mmol), Pd2(dba)3(103 mg, 0.11 mmol) and RuPhos (105 mg, 0.23 mmol) in 1,4-dioxane (5 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was diluted with DCM (10 mL), and the solvent was removed under reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 1:1), to give the title compound (0.33 g, 97%) as a red gum; MS (ESI) m/z [M+H]+301. Intermediate 292: 6-(3-Ethoxy-3-methylazetidin-1-yl)quinoline-4-carboxylic acid NaOH (216 mg, 5.41 mmol) was added to a solution of methyl 6-(3-ethoxy-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 291 (325 mg, 1.08 mmol) in MeOH (9 mL) and water (3 mL) at 25° C., and the reaction was stirred at 25° C. for 1 h. The solvent was removed under reduced pressure, the reaction mixture was diluted with water (50 mL) and pH was adjusted to 3 with aq HCl (1 M). The aqueous phase was extracted with EtOAc (3×50 mL), and the combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.305 g, 98%) as an orange solid; MS (ESI) m/z [M+H]+287. Intermediate 293: Methyl 6-(1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylate Cs2CO3(430 mg, 1.32 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (270 mg, 1.01 mmol), 1-oxa-6-azaspiro[3.3]heptane oxalate (250 mg, 1.32 mmol) and RuPhos Pd G3 (85 mg, 0.10 mmol) in 1,4-dioxane (5 mL) at 35° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (MeOH:DCM, 1:10), to give the title compound (0.283 g, 98%) as a yellow solid; MS (ESI) m/z [M+H]+285. Intermediate 294: 6-(1-Oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylic acid A solution of NaOH (177 mg, 4.43 mmol) in water (2 mL) was added to a stirred solution of methyl 6-(1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylate Intermediate 293 (252 mg, 0.89 mmol) in MeOH (6 mL) at 0° C., and the reaction mixture was stirred at 37° C. for 1 h. The reaction mixture was diluted with water (20 mL), and the pH was adjusted to 6 with aq HCl (2 M). The reaction mixture was extracted with EtOAc (8×75 mL), the combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure, to give the title compound (0.214 g, 89%) as an orange solid; MS (ESI) m/z [M+H]+271. Intermediate 295: Methyl 6-(3-ethyl-3-hydroxyazetidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(103 mg, 0.11 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), 3-ethylazetidin-3-ol (137 mg, 1.35 mmol), Cs2CO3(735 mg, 2.25 mmol) and DavePhos (89 mg, 0.23 mmol) in 1,4-dioxane (15 mL) at 20° C. and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was concentrated under reduced pressure and the residue was diluted with EtOAc (75 mL). The mixture was washed sequentially with water (20 mL) and sat brine (20 mL), and the organic layer was dried over Na2SO4, filtered, and evaporated at reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:5) to give the title compound (0.20 g, 62%) as a yellow solid; MS (ESI) m/z [M+H]+287.0. Intermediate 296: 6-(3-Ethyl-3-hydroxyazetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (140 mg, 3.49 mmol) in water (3 mL) was added to a stirred solution of methyl 6-(3-ethyl-3-hydroxyazetidin-1-yl)quinoline-4-carboxylate Intermediate 295 (200 mg, 0.70 mmol) in MeOH (9 mL) at 25° C., and the reaction mixture was stirred at 25° C. for 3 h. The pH of the reaction mixture was adjusted to 4 with aq HCl (2 M), and the reaction mixture was concentrated under reduced pressure. The residue was diluted with EtOAc (75 mL), and the organic layer was washed sequentially with water (25 mL) and sat brine (25 mL), dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.15 g, 79%) as a red solid; MS (ESI) m/z [M+H]+273.0. Intermediate 297: Methyl 6-(6-fluoro-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Cs2CO3(612 mg, 1.88 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 6-fluoro-2-azaspiro[3.3]heptane trifluoroacetate (344 mg, 1.50 mmol), Pd2(dba)3(69 mg, 0.08 mmol) and DavePhos (59 mg, 0.15 mmol) in 1,4-dioxane (5 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was diluted with DCM (10 mL) and the solvent was removed under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:1), to give the title compound (0.22 g, 97%) as an orange gum; MS (ESI) m/z [M+H]+301. Intermediate 298: 6-(6-Fluoro-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid NaOH (146 mg, 3.65 mmol) was added to a solution of methyl 6-(6-fluoro-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Intermediate 297 (219 mg, 0.73 mmol) in MeOH (6 mL) and water (2 mL) at 25° C., and the reaction was stirred at 25° C. for 1 h. The solvent was removed under reduced pressure and the residue was diluted with water (50 mL), and pH was adjusted to 3 with aq HCl (1 M). The aqueous phase was extracted with EtOAc (3×50 mL), the combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure to give the title compound (0.205 g, 98%) as an orange solid; MS (ESI) m/z [M+H]+287. Intermediate 299: rac-Methyl 6-((1R,5R)-6-azabicyclo[3.2.0]heptan-6-yl)quinoline-4-carboxylate A solution of methyl 6-bromoquinoline-4-carboxylate (400 mg, 1.50 mmol) in 1,4-dioxane (10 mL) was added to a mixture of 6-azabicyclo[3.2.0]heptane (146 mg, 1.50 mmol), Pd2(dba)3(14 mg, 0.02 mmol) XPhos (14 mg, 0.03 mmol) and Cs2CO3(980 mg, 3.01 mmol), and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was filtered through silica, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.17 g, 40%) as a yellow solid; MS (ESI) m/z [M+H]+382.2. Intermediate 300: rac-6-((1R,5R)-6-Azabicyclo[3.2.0]heptan-6-yl)quinoline-4-carboxylic acid LiOH (25 mg, 1.1 mmol) was added to a stirred solution of methyl 6-(6-azabicyclo[3.2.0]heptan-6-yl)quinoline-4-carboxylate Intermediate 299 (150 mg, 0.53 mmol) in THF (3 mL) and water (3 mL), and the reaction mixture was stirred at 25° C. for 4 h. The pH of the reaction mixture was adjusted to 5 with aq HCl (2 M), and the reaction mixture was concentrated under reduced pressure to give the title compound (0.13 g, 91%) as a red solid; MS (ESI) m/z [M+H]+269.2. Intermediate 301: Methyl 6-(3-hydroxy-3-(hydroxymethyl)azetidin-1-yl)quinoline-4-carboxylate Cs2CO3(814 mg, 2.50 mmol) was added to methyl 6-bromoquinoline-4-carboxylate (266 mg, 1.00 mmol), 3-(hydroxymethyl)azetidin-3-ol oxalate (193 mg, 1.30 mmol), Pd2(dba)3(92 mg, 0.10 mmol) and XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (5 mL) at 12° C., and the reaction mixture was stirred at 100° C. for 15 h. The reaction mixture was filtered through Celite®, and the filtrated was concentrated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH, 10:1), to give the title compound (0.134 g, 46%) as a brown solid; MS (ESI) m/z [M+H]+289. Intermediate 302: Methyl 6-(3-fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxylate A solution of DAST (972 μL, 7.35 mmol) in anhydrous DCM (5 mL) was added dropwise to a stirred solution of methyl 6-(3-hydroxy-3-(hydroxymethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 301 (212 mg, 0.74 mmol) in anhydrous DCM (25 mL), cooled to −60° C., over a period of 10 min. The reaction mixture was warmed to 25° C. and stirred for 8 h. The reaction mixture was poured into sat NaHCO3(25 mL, aq), and extracted with DCM (5×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH, 10:1), to give the title compound (0.12 g, 56%) as a brown oil; MS (ESI) m/z [M+H]+293. Intermediate 303: 6-(3-Fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (45 mg, 1.13 mmol) in water (1 mL) was added slowly to a stirred solution of methyl 6-(3-fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 302 (110 mg, 0.38 mmol) in MeOH (3 mL), cooled to 0° C., and the reaction mixture was stirred at 28° C. for 1.5 h. The reaction mixture was acidified with aq HCl (2 M), and extracted with EtOAc (5×100 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure to give the title compound (0.10 g, 95%) as a beige solid; MS (ESI) m/z [M+H]+279. Intermediate 304: Methyl 6-(3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Cs2CO3(563 mg, 1.73 mmol) was added to methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 3-(2,2,2-trifluoroethyl)azetidine hydrochloride (172 mg, 0.98 mmol), Pd2(dba)3(69 mg, 0.08 mmol) and XPhos (72 mg, 0.15 mmol) in 1,4-dioxane (20 mL) at 28° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC(DCM:MeOH, 10:1), to give the title compound (0.415 g) as a crude; MS (ESI) m/z [M+H]+325. Intermediate 305: 6-(3-(2,2,2-Trifluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (134 mg, 3.36 mmol) in water (2 mL) was added to a stirred solution of methyl 6-(3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 304 (363 mg, 1.12 mmol) in MeOH (6 mL), cooled to 0° C., and the reaction mixture was stirred at 20° C. for 2 h. The reaction mixture was acidified to pH 6 with aq HCl (2 M, aq), and extracted with EtOAc (10×100 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure, to give the title compound (0.333 g, 96%) as an orange solid; MS (ESI) m/z [M+H]+311. Intermediate 306: rac-Methyl (R)-6-(3,3-difluoro-2-methylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(735 mg, 2.25 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 3,3-difluoro-2-methylazetidine hydrochloride (216 mg, 1.50 mmol) and RuPhos Pd G3 (63 mg, 0.08 mmol) in 1,4-dioxane (4 mL) at 20° C., and the reaction mixture was stirred at 100° C. for 4 h. The reaction mixture was diluted with DCM, and filtered through silica, and the filter cake was washed with DCM. The filtrate was concentrated under reduced pressure, and the crude product was purified by preparative TLC (petroleum ether:EtOAc, 1:1), to give the title compound (0.215 g, 98%) as a brown gum; MS (ESI) m/z [M+H]+293. Intermediate 307: rac-(R)-6-(3,3-Difluoro-2-methylazetidin-1-yl)quinoline-4-carboxylic acid NaOH (146 mg, 3.66 mmol) was added to a solution of methyl 6-(3,3-difluoro-2-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 306 (214 mg, 0.73 mmol) in MeOH (1.5 mL) and water (0.5 mL) at 20° C., and the reaction mixture was stirred at 20° C. for 1 h. The reaction mixture was concentrated under reduced pressure, diluted with water, and the pH was adjusted to 3 with aq HCl (1 M). The reaction mixture was diluted with water (50 mL), and washed with EtOAc (5×50 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure to give the title compound (0.198 g, 97%) as a yellow solid; MS (ESI) m/z [M+H]+279. Intermediate 308: tert-Butyl 6-(6,6-difluoro-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylate Cs2CO3(132 mg, 0.41 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (50 mg, 0.16 mmol), 6,6-difluoro-3-azabicyclo[3.1.0]hexane hydrochloride (28 mg, 0.18 mmol), Pd2(dba)3(15 mg, 0.02 mmol) and XPhos (15 mg, 0.03 mmol) in 1,4-dioxane (5 mL) at 20° C., and the reaction mixture was stirred at 100° C. for 5 h The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 2:1), followed by reversed phase flash chromatography on a C18 column (gradient: 0-75% MeCN in water) to give the title compound (0.137 g) as a crude yellow solid; MS (ESI) m/z [M+H]+347. Intermediate 309: 6-(6,6-Difluoro-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylic acid TFA (5 mL) was added to a stirred solution of tert-butyl 6-(6,6-difluoro-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylate Intermediate 308 (123 mg, 0.36 mmol) in DCM (5 mL) at 15° C., and the reaction mixture was stirred at 15° C. for 15 h. The solvent was removed under reduced pressure, to give the title compound (0.198 g, 97%) as a crude, dark red solid; MS (ESI) m/z [M+H]+291. Intermediate 310: Methyl (R)-6-(3-methoxypyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(735 mg, 2.25 mmol), Pd2(dba)3(10 mg, 0.01 mmol) and XPhos (11 mg, 0.02 mmol) was added to a solution of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol) and (R)-3-methoxypyrrolidine (228 mg, 2.25 mmol) in 1,4-dioxane (10 mL), and the reaction mixture was stirred at 100° C. for 2 h. The solvent was removed under reduced pressure and the crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.30 g, 93%) as a red oil which solidified on standing; MS (ESI) m/z [M+H]+287.1. Intermediate 311: (R)-6-(3-Methoxypyrrolidin-1-yl)quinoline-4-carboxylic acid LiOH (90 mg, 3.8 mmol) was add to a solution of methyl (R)-6-(3-methoxypyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 310 (270 mg, 0.94 mmol) in THF (0.5 mL) and water (0.5 mL), and the reaction mixture was stirred at 25° C. for 3 h. The pH of the reaction mixture was adjusted to 5 with aq HCl (2 M), and the reaction mixture was concentrated under reduced pressure to give the title compound (0.20 g, 78%) as a red solid; MS (ESI) m/z [M+H]+273. Intermediate 312: Methyl 6-(3-oxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylate Pd2(dba)3(52 mg, 0.06 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), 3-oxa-9-azaspiro[5.5]undecane (263 mg, 1.69 mmol), Cs2CO3(735 mg, 2.25 mmol) and XantPhos (98 mg, 0.17 mmol) in 1,4-dioxane (15 mL) and the reaction mixture was stirred at 80° C. for 2 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.24 g, 62%) as a yellow gum; MS (ESI) m/z [M+H]+341. Intermediate 313: 6-(3-Oxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylic acid A solution of methyl 6-(3-oxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylate Intermediate 312 (230 mg, 0.68 mmol) and LiOH (81 mg, 3.4 mmol) in MeOH (10 mL) and water (2 mL) was stirred at rt for 3 h. The solvent was removed under reduced pressure and the reaction mixture was diluted with water, and the pH was adjusted to 6 with aq HCl (1 M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.20 g, 91%) as a yellow solid; MS (ESI) m/z [M+H]+327. Intermediate 314: Methyl 6-(5,8-dioxa-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylate A solution of methyl 6-bromoquinoline-4-carboxylate (400 mg, 1.50 mmol) in 1,4-dioxane (15 mL) was added to a mixture of 5,8-dioxa-2-azaspiro[3.4]octane (260 mg, 2.25 mmol), Pd2(dba)3(14 mg, 0.02 mmol) XPhos (14 mg, 0.03 mmol) and Cs2CO3(980 mg, 3.01 mmol), and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was filtered through silica, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 2:1), to give the title compound (0.35 g, 78%) as a yellow solid; MS (ESI) m/z [M+H]+301.1. Intermediate 315: 6-(5,8-Dioxa-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylic acid LiOH (115 mg, 4.79 mmol) was added to a stirred solution of methyl 6-(5,8-dioxa-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylate Intermediate 314 (360 mg, 1.20 mmol) in THF (5 mL) and water (5 mL), and the reaction mixture was stirred at 25° C. for 4 h. The pH of the reaction mixture was adjusted to 5 with aq HCl (2 M) and the reaction mixture was concentrated under reduced pressure to give the title compound (0.30 g, 87%) as a red solid; MS (ESI) m/z [M+H]+287.1. Intermediate 316: Methyl 6-(5-azaspiro[2.3]hexan-5-yl)quinoline-4-carboxylate Cs2CO3(612 mg, 1.88 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 5-azaspiro[2.3]hexane hydrochloride (135 mg, 1.13 mmol), Pd2(dba)3(69 mg, 0.08 mmol) and XPhos (72 mg, 0.15 mmol) in 1,4-dioxane (4 mL) at 15° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was diluted with DCM, and the solvent was removed under reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 1:1), to give the title compound (0.189 g, 94%) as a yellow solid; MS (ESI) m/z [M+H]+269. Intermediate 317: 6-(5-Azaspiro[2.3]hexan-5-yl)quinoline-4-carboxylic acid NaOH (140 mg, 3.50 mmol) was added to a solution of methyl 6-(5-azaspiro[2.3]hexan-5-yl)quinoline-4-carboxylate Intermediate 316 (188 mg, 0.70 mmol) in MeOH (1.5 mL) and water (0.5 mL) at 15° C., and the reaction mixture was stirred at 15° C. for 1 h. The reaction mixture was concentrated under reduced pressure, diluted with water, and the pH was adjusted to 3 with aq HCl (1 M). The reaction mixture was diluted with EtOAc (50 mL), and washed with water (10×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure to give the title compound (0.327 g) as an orange solid; MS (ESI) m/z [M+H]+255. Intermediate 318: Methyl 6-(3-hydroxy-3-methylazetidin-1-yl)quinoline-4-carboxylate Cs2CO3(1.30 g, 4.01 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate hydrochloride Intermediate 422 (303 mg, 1.00 mmol), 3-methylazetidin-3-ol hydrochloride (155 mg, 1.25 mmol), Pd2(dba)3(92 mg, 0.10 mmol) and XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (7 mL) at 20° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 10:1), to give the title compound (0.193 g, 71%) as a yellow solid; MS (ESI) m/z [M+H]+273. Intermediate 319: 6-(3-Hydroxy-3-methylazetidin-1-yl)quinoline-4-carboxylic acid A solution of NaOH (110 mg, 2.75 mmol) in water (4 mL) was added slowly to a stirred solution of methyl 6-(3-hydroxy-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 318 (150 mg, 0.55 mmol) in MeOH (12 mL), cooled to 0° C., and the reaction mixture was stirred at 20° C. for 1 h. The pH of the reaction mixture was adjusted to 6 with aq HCl (2 M), and the mixture was extracted with EtOAc (5×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated under reduced pressure to give the title compound (0.095 g, 67%) as a yellow oil which solidified on standing; MS (ESI) m/z [M+H]+259. Intermediate 320: Methyl 6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylate Cs2CO3(1.53 g, 4.70 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (500 mg, 1.88 mmol), 2-oxa-6-azaspiro[3.3]heptane oxalate (462 mg, 2.44 mmol) and RuPhos Pd G3 (157 mg, 0.19 mmol) in 1,4-dioxane (15 mL) at 28° C., and the reaction mixture was stirred at 100° C. for 4 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH, 10:1), to give the title compound (0.254 g, 48%) as a yellow solid; MS (ESI) m/z [M+H]+285. Intermediate 321: 6-(2-Oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylic acid A solution of NaOH (196 mg, 4.90 mmol) in water (3 mL) was added slowly to a stirred suspension of methyl 6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylate Intermediate 320 (232 mg, 0.82 mmol) in MeOH (12 mL) cooled to 0° C., and the reaction mixture was stirred at 25° C. for 1 h. The reaction mixture was diluted with water (20 mL), the pH was adjusted to 6 with aq HCl (2 M), and the reaction mixture was extracted with EtOAc (6×100 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure to give the title compound (0.19 g, 86%) as a red solid; MS (ESI) m/z [M+H]+271. Intermediate 322: Methyl (R)-6-(3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(52 mg, 0.06 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), (R)-3-methylpyrrolidin-3-ol (228 mg, 2.25 mmol), XPhos (81 mg, 0.17 mmol) and Cs2CO3(735 mg, 2.25 mmol) in 1,4-dioxane (15 mL) and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was filtered, and the filtrate was concentrated under vacuum. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 1:3), to give the title compound (0.28 g, 87%) as a yellow solid; MS (ESI) m/z [M+H]+287. Intermediate 323: (R)-6-(3-Hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid A solution of methyl (R)-6-(3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 322 (280 mg, 0.98 mmol) and LiOH (94 mg, 3.91 mmol) in MeOH (10 mL) and water (2 mL) was stirred at rt for 1 h. The solvent was removed under reduced pressure. The residue was diluted with water, and the pH was adjusted to 6 with aq HCl (1 M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.22 g, 83%) as an orange solid; MS (ESI) m/z [M+H]+273. Intermediate 324: Methyl (S)-6-(3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxylate Pd2(dba)3(52 mg, 0.06 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), (S)-3-methylpyrrolidin-3-ol hydrochloride (233 mg, 1.69 mmol), XPhos (81 mg, 0.17 mmol) and Cs2CO3(1.10 g, 3.38 mmol) in 1,4-dioxane (10 mL), and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The reaction mixture was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was purified by preparative TLC (petroleum ether:EtOAc, 1:3), to give the title compound (0.26 g, 81%) as a yellow solid; MS (ESI) m/z [M+H]+287. Intermediate 325: (S)-6-(3-Hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid A solution of methyl (S)-6-(3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 324 (250 mg, 0.87 mmol) and LiOH (63 mg, 2.6 mmol) in MeOH (10 mL) and water (2 mL) was stirred at rt for 2 h. The solvent was removed under reduced pressure. The reaction mixture was diluted with water, and the pH was adjusted to 6 with aq HCl (1 M). The precipitate was collected by filtration, washed with water and dried under vacuum to give the title compound (0.21 g, 88%) as an orange solid; MS (ESI) m/z [M+H]+273. Intermediate 326: 6-(6-(Difluoromethyl)pyridin-3-yl)quinoline-4-carboxylic acid A mixture of (6-(difluoromethyl)pyridin-3-yl)boronic acid (103 mg, 0.60 mmol), 6-bromoquinoline-4-carboxylic acid (150 mg, 0.60 mmol), Cs2CO3(388 mg, 1.19 mmol) and Pd(dtbpf)Cl2(58 mg, 0.09 mmol) in 1,4-dioxane (4.8 mL) and water (1.2 mL) was stirred at rt for 2 h. The reaction mixture was concentrated under reduced pressure, diluted with water and the pH was adjusted to 2 with aq HCl (1 M). The precipitate was filtered off, washed with diethyl ether and dried under vacuum to give the title compound (143 mg, 80%); MS (ESI) m/z [M+H]+301.1. Intermediate 327: 6-(1-Cyclopropyl-1H-pyrazol-4-yl)quinoline-4-carboxylic acid A mixture of 6-bromoquinoline-4-carboxylic acid (150 mg, 0.60 mmol), (1-cyclopropyl-1H-pyrazol-4-yl)boronic acid (136 mg, 0.89 mmol), Cs2CO3(388 mg, 1.19 mmol) and Pd(dtbpf)Cl2(58 mg, 0.09 mmol) in 1,4-dioxane (4.8 mL) and water (1.2 mL) was stirred at rt for 2 h. The reaction mixture was concentrated under reduced pressure, diluted with water and the pH was adjusted to 2 with aq HCl (1 M). The precipitate was filtered off, washed with diethyl ether and dried under vacuum to give the title compound (0.124 g, 75%); MS (ESI) m/z [M+H]+280.1. Intermediate 328: 6-(1,3-Dimethyl-1H-pyrazol-4-yl)quinoline-4-carboxylic acid A mixture of 1,3-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (0.448 g, 2.02 mmol), 6-bromoquinoline-4-carboxylic acid (0.508 g, 2.02 mmol), Cs2CO3(1.31 g, 4.03 mmol) and Pd(dtbpf)Cl2(0.131 g, 0.20 mmol) in 1,4-dioxane (10 mL) and water (2.5 mL) was stirred overnight. Pd(dtbpf)Cl2(25 mg, 0.04 mmol) was added and the reaction was stirred for 6 h. Pd(dtbpf)Cl2(25 mg, 0.04 mmol) was added and the reaction was stirred for 4 days. The reaction mixture was diluted with a few mL of DMSO and evaporated under reduced pressure, and the crude product was purified by preparative HPLC, PrepMethod E, (gradient: 0-30%) to give the title compound (0.314 g, 58%); MS (ESI) m/z [M+H]+268. Intermediate 329: 6-(3,5-Dimethyl-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxylic acid A mixture of 3,5-dimethyl-1-(tetrahydro-2H-pyran-4-yl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (68 mg, 0.22 mmol), 6-bromoquinoline-4-carboxylic acid (56 mg, 0.22 mmol), Cs2CO3(145 mg, 0.44 mmol) and Pd(dtbpf)Cl2(14 mg, 0.02 mmol) in 1,4-dioxane (1 mL) and water (0.25 mL) was stirred overnight. The reaction mixture was diluted with a few mL of DMSO and evaporated under reduced pressure, and the crude product was purified by preparative HPLC, PrepMethod E, (gradient: 5-45%) to give the title compound (40 mg, 51%); MS (ESI) m/z [M+H]+352. Intermediate 330: 6-(1-(Tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxylic acid A mixture of (1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)boronic acid (40 mg, 0.21 mmol), 6-bromoquinoline-4-carboxylic acid (52 mg, 0.21 mmol), Cs2CO3(134 mg, 0.41 mmol) and Pd(dtbpf)Cl2(13 mg, 0.02 mmol) in 1,4-dioxane (1 mL) and water (0.25 mL) was stirred overnight. The reaction mixture was diluted with a few mL of DMSO and evaporated under reduced pressure, and the crude product was purified by preparative HPLC, PrepMethod E, (gradient: 5-45%) to give the title compound (26 mg, 39%); MS (ESI) m/z [M+H]+324. Intermediate 331: 6-(1-Methyl-1H-pyrazol-5-yl)quinoline-4-carboxylic acid A mixture of 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (41 mg, 0.20 mmol), 6-bromoquinoline-4-carboxylic acid (50 mg, 0.20 mmol), Cs2CO3(194 mg, 0.60 mmol) and Pd(dtbpf)Cl2(22 mg, 0.03 mmol) in 1,4-dioxane (1 mL) and water (0.25 mL) was stirred overnight. The reaction mixture was diluted with a few mL of DMSO and evaporated under reduced pressure, and the crude product was purified by preparative HPLC, PrepMethod E, (gradient: 0-30%) to give the title compound (29 mg, 58%); MS (ESI) m/z [M+H]+254. Intermediate 332: (5,6,7,8-Tetrahydroimidazo[1,2-a]pyridin-3-yl)boronic acid A solution of n-BuLi (280 mL, 0.69 mol, 2.5 M in hexane) was cannulated into a solution of 5,6,7,8-tetrahydroimidazo[1,2-a]pyridine in THF at −60° C. and over 20 min. The reaction mixture was stirred at rt for 30 min, and then cooled to −60° C., and added dropwise to a solution of B(C3H7O)3(150 g, 0.7 mol). The reaction mixture was stirred at rt for 6 h, and then quenched by the addition of aq HCl (1 M). The reaction mixture was concentrated under reduced pressure and the pH was adjusted to 2 with aq HCl (1 M). The reaction mixture was filtered, and the filter cake was washed with EtOAc, and dried in vacuo to give the title compound (54 g, 57%); MS (ESI) m/z [M+H]+167.1. Intermediate 333: 6-(5,6,7,8-Tetrahydroimidazo[1,2-a]pyridin-3-yl)quinoline-4-carboxylic acid A mixture of (5,6,7,8-tetrahydroimidazo[1,2-a]pyridin-3-yl)boronic acid Intermediate 332 (37 mg, 0.22 mmol), 6-bromoquinoline-4-carboxylic acid (56 mg, 0.22 mmol), Cs2CO3(145 mg, 0.44 mmol) and Pd(dtbpf)C2(14 mg, 0.02 mmol) in 1,4-dioxane (1 mL) and water (0.25 mL) was stirred overnight. The reaction mixture was diluted with a few mL of DMSO and evaporated under reduced pressure, and the crude compound was purified by preparative HPLC, PrepMethod E, (gradient: 0-45%) to give the title compound (25 mg, 38%); MS (ESI) m/z [M+H]+294. Intermediate 334: Methyl 6-bromo-7-chloroquinoline-4-carboxylate SOCl2(410 μL, 5.62 mmol) was added slowly to a solution of 6-bromo-7-chloroquinoline-4-carboxylic acid (322 mg, 1.12 mmol) in MeOH (10 mL) at 20° C., and the reaction mixture was stirred at 60° C. for 4 h. The reaction mixture was diluted with DCM (75 mL), and washed sequentially with water (20 mL) and sat brine (20 mL, aq). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH, 40:1), to give the title compound (0.188 g, 56%) as a yellow solid; MS (ESI) m/z [M+H]+301.9. Intermediate 335: Methyl 7-chloro-6-morpholinoquinoline-4-carboxylate Pd2(dba)3(46 mg, 0.05 mmol) was added to a suspension of methyl 6-bromo-7-chloroquinoline-4-carboxylate Intermediate 334 (150 mg, 0.50 mmol), morpholine (217 mg, 2.50 mmol), Cs2CO3(22 mg, 0.07 mmol) and XantPhos (58 mg, 0.10 mmol) in 1,4-dioxane (15 mL) at 20° C., and the reaction mixture was stirred at 100° C. for 5 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (75 mL), and washed sequentially with water (15 mL) and sat brine (15 mL, aq). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH, 40:1), to give the title compound (0.074 g, 48%) as a yellow solid; MS (ESI) m/z [M+H]+307.0. Intermediate 336: 7-Chloro-6-morpholinoquinoline-4-carboxylic acid NaOH (46 mg, 1.1 mmol) was added to a solution of methyl 7-chloro-6-morpholinoquinoline-4-carboxylate Intermediate 335 (70 mg, 0.23 mmol) in MeOH (9 mL) and water (1 mL) at 20° C., and the reaction mixture was stirred at 25° C. for 3 h. The pH of the reaction mixture was adjusted to 3 using aq HCl (2 M, 15 mL). The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (20 mL), and washed with water (10 mL). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure to give the title compound (0.060 g, 90%) as a yellow solid; MS (ESI) m/z [M+H]+292.9. Intermediate 337: Methyl 6-bromo-8-chloroquinoline-4-carboxylate SOCl2(159 μL, 2.18 mmol) was added slowly to a solution of 6-bromo-8-chloroquinoline-4-carboxylic acid (125 mg, 0.44 mmol) in MeOH (10 mL) at 20° C., and the reaction mixture was stirred at 60° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (50 mL), and washed sequentially with water (15 mL) and sat brine (15 mL). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:5), to give the title compound (0.10 g, 76%) as a yellow solid; MS (ESI) m/z [M+H]+301.9. Intermediate 338: Methyl 8-chloro-6-morpholinoquinoline-4-carboxylate Pd2(dba)3(30 mg, 0.03 mmol) was added to a suspension of methyl 6-bromo-8-chloroquinoline-4-carboxylate Intermediate 337 (100 mg, 0.33 mmol), morpholine (145 mg, 1.66 mmol), XantPhos (38 mg, 0.07 mmol) and Cs2CO3(217 mg, 0.67 mmol) in 1,4-dioxane (15 mL) at 20° C., and the reaction mixture was stirred at 100° C. for 2 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (50 mL), and washed sequentially with sat brine (15 mL) and water (15 mL). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:5), to give the title compound (0.082 g, 80%) as a yellow solid; MS (ESI) m/z [M+H]+307. Intermediate 339: 8-Chloro-6-morpholinoquinoline-4-carboxylic acid A solution of NaOH (52 mg, 1.3 mmol) in water (3 mL) was added to a stirred solution of methyl 8-chloro-6-morpholinoquinoline-4-carboxylate Intermediate 338 (80 mg, 0.26 mmol) in MeOH (9 mL) at 20° C., and the reaction mixture was stirred at 25° C. for 2 h. The pH of the reaction mixture was adjusted to 4 with aq HCl (2 M). The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (50 mL), and washed sequentially with sat brine (20 mL) and water (15 mL). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure to give the title compound (0.070 g, 92%) as a red solid; MS (ESI) m/z [M+H]+293.0. Intermediate 340: Ethyl 6-(3-(acetamidomethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate Ethyl 6-bromoquinoline-4-carboxylate (100 mg, 0.36 mmol), N-((3-methylazetidin-3-yl)methyl)acetamide hydrochloride, (128 mg, 0.71 mmol), Cs2CO3(465 mg, 1.43 mmol), XPhos (34 mg, 0.07 mmol) and Pd2(dba)3(33 mg, 0.04 mmol) were weighed into a 5 mL vial. Dioxane (2 mL) was added and the reaction mixture was purged with N2(g) for 10 min. The vial was sealed and the reaction mixture was stirred at 100° C. for 2 h. After cooling to rt, water (10 mL) and DCM (10 mL) were added and the reaction mixture was stirred and filtered through a phase separator and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 20-100% of EtOAc (containing 5% 2 M NH3in MeOH) in heptane) to give the title compound (47 mg, 39%); MS (ESI) m/z [M+H]+342.3. Intermediate 341: 6-(3-(Acetamidomethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 264 μL, 0.26 mmol, 1 M) was added to a solution of ethyl 6-(3-(acetamidomethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylate Intermediate 340 (45 mg, 0.13 mmol) in MeOH (1.5 mL). The reaction mixture was stirred at 50° C. for 20 min, then cooled to rt. Aq HCl (3.8 M, 87 μL, 0.33 mmol, 3.8 M) was added, and the reaction mixture was evaporated under reduced pressure and co-evaporated under reduced pressure with EtOH (2×) and MeCN (1×), to give the crude title compound; MS (ESI) m/z [M+H]+314.2. Intermediate 342: Ethyl 6-(3-fluoro-3-phenylazetidin-1-yl)quinoline-4-carboxylate Ethyl 6-bromoquinoline-4-carboxylate (120 mg, 0.43 mmol), 3-fluoro-3-phenylazetidine hydrochloride (161 mg, 0.86 mmol), Cs2CO3(558 mg, 1.71 mmol), XPhos (41 mg, 0.09 mmol) and Pd2(dba)3(39 mg, 0.04 mmol) were weighed into a 5 mL vial. Dioxane (2 mL) was added and the reaction mixture was purged with N2(g) for 10 min. The vial was sealed and the reaction mixture was stirred at 100° C. for 2 h. After cooling to rt, water (10 mL) and DCM (10 mL) were added and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The residue was purified by straight phase flash chromatography on silica (gradient: 5-50% of EtOAc in heptane) to give the title compound (26 mg, 17%); MS (ESI) m/z [M+H]+351.3. Intermediate 343: 6-(3-Fluoro-3-phenylazetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 143 μL, 0.14 mmol) was added to a solution of ethyl 6-(3-fluoro-3-phenylazetidin-1-yl)quinoline-4-carboxylate Intermediate 342 (25 mg, 0.07 mmol) in MeOH (1 mL), and the reaction mixture was stirred at 50° C. for 20 min, and then cooled to rt. Aq HCl (3.8 M, 47 μL, 0.18 mmol) was added, and the reaction mixture was evaporated under reduced pressure and co-evaporated under reduced pressure with EtOH (2×), and MeCN (1×), to give the crude title compound; MS (ESI) m/z [M+H]+: 323.2. Intermediate 344: Ethyl 6-(3-(p-tolyl)azetidin-1-yl)quinoline-4-carboxylate Ethyl 6-bromoquinoline-4-carboxylate (150 mg, 0.54 mmol), 3-(p-tolyl)azetidine hydrochloride, (148 mg, 0.80 mmol), Cs2CO3(698 mg, 2.14 mmol), XPhos (51 mg, 0.11 mmol) and Pd2(dba)3(49 mg, 0.05 mmol) were weighed into a 5 mL vial. Dioxane (2 mL) was added and the reaction mixture was purged with N2(g) for 10 min. The vial was sealed and the reaction mixture was stirred at 100° C. for 3 h. After cooling to rt, water (10 mL) and DCM (15 mL) were added and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The residue was purified by straight phase flash chromatography on silica (gradient: 5-50% of EtOAc in heptane) to give the title compound (22 mg, 12%); MS (ESI) m/z [M+H]+347.3. Intermediate 345: 6-(3-(p-Tolyl)azetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1M, 115 μL, 0.12 mmol) was added to a solution of ethyl 6-(3-(p-tolyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 344 (20 mg, 0.06 mmol) in MeOH (1 mL). The reaction mixture was stirred at 50° C. for 20 min, then cooled to rt. Aq HCl (1 M, 144 μL, 0.14 mmol) was added and the reaction mixture was stirred, evaporated under reduced pressure, and co-evaporated under reduced pressure with EtOH (2×), and MeCN (1×), to give the crude title compound; MS (ESI) m/z [M+H]+319.2. Intermediate 346: Ethyl 6-(6-acetyl-2,6-diazaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Ethyl 6-bromoquinoline-4-carboxylate (120 mg, 0.43 mmol), 1-(2,6-diazaspiro[3.3]-heptan-2-yl)ethan-1-one oxalate, (148 mg, 0.64 mmol), Cs2CO3(558 mg, 1.71 mmol), XPhos (41 mg, 0.09 mmol) and Pd2(dba)3(39 mg, 0.04 mmol) were weighed into a 5 mL vial. Dioxane (2.5 mL) was added and the reaction mixture was purged with N2(g) for 10 min. The vial was sealed and the reaction mixture was stirred at 100° C. for 3 h. After cooling to rt, water (10 mL) and DCM (15 mL) were added, and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The residue was purified by straight phase flash chromatography on silica (gradient: 25-100% of EtOAc (containing 5% 2 M NH3in MeOH) in heptane) to give the title compound (120 mg, 83%); MS (ESI) m/z [M+H]+340.29. Intermediate 347: 6-(6-Acetyl-2,6-diazaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 707 μL, 0.71 mmol) was added to a solution of ethyl 6-(6-acetyl-2,6-diazaspiro[3.3]heptan-2-yl)quinoline-4-carboxylate Intermediate 346 (120 mg, 0.35 mmol) in MeOH (4 mL). The reaction mixture was stirred at 50° C. for 20 min, and then cooled to rt. Aq HCl (1 M, 884 μL, 0.88 mmol) was added, and the reaction mixture was stirred, evaporated under reduced pressure, and co-evaporated under reduced pressure with EtOH (2×), and MeCN (1×), to give the crude title compound; MS (ESI) m/z [M+H]+312.2. Intermediate 348: Ethyl 6-(3-(4-fluorophenyl)azetidin-1-yl)quinoline-4-carboxylate Ethyl 6-bromoquinoline-4-carboxylate (120 mg, 0.43 mmol), 3-(4-fluorophenyl)-azetidine hydrochloride (121 mg, 0.64 mmol), Cs2CO3(558 mg, 1.71 mmol), XPhos (41 mg, 0.09 mmol) and Pd2(dba)3(39 mg, 0.04 mmol) were weighed into a 5 mL vial. Toluene (2.5 mL) was added and the reaction mixture was purged with N2(g) for 10 min. The vial was sealed and the reaction mixture was stirred at 110° C. for 2 h. After cooling to rt, water (10 mL) and DCM (15 mL) were added, and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The residue was purified by straight phase flash chromatography on silica (gradient: 5-50% of EtOAc in heptane) to give the title compound (130 mg, 87%); MS (ESI) m/z [M+H]+351.2. Intermediate 349: 6-(3-(4-Fluorophenyl)azetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 685 μL, 0.68 mmol) was added to a solution of ethyl 6-(3-(4-fluorophenyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 348 (120 mg, 0.34 mmol) in MeOH (4 mL). The reaction mixture was stirred at 50° C. for 20 min, then cooled to rt. Aq HCl (1 M, 856 μL, 0.86 mmol) was added, and the reaction mixture was stirred, evaporated under reduced pressure and co-evaporated under reduced pressure with EtOH (2×), and MeCN (1×), to give the crude title compound; MS (ESI) m/z [M+H]+323.19. Intermediate 350: Ethyl 6-(3-(m-tolyl)azetidin-1-yl)quinoline-4-carboxylate Ethyl 6-bromoquinoline-4-carboxylate (120 mg, 0.43 mmol), 3-(m-tolyl)azetidine hydrochloride (118 mg, 0.64 mmol), Cs2CO3(558 mg, 1.71 mmol), XPhos (41 mg, 0.09 mmol) and Pd2(dba)3(39 mg, 0.04 mmol) were weighed into a 5 mL vial. Toluene (2.5 mL) was added and the reaction mixture was purged with N2(g) for 10 min. The vial was sealed and the reaction mixture was stirred at 100° C. for 3 h. After cooling to rt, water (10 mL) and DCM (15 mL) were added and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The residue was purified by straight phase flash chromatography on silica (gradient: 5-50% of EtOAc in heptane) to give the title compound (128 mg, 86%); MS (ESI) m/z [M+H]+347.36. Intermediate 351: 6-(3-(m-Tolyl)azetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 687 μL, 0.69 mmol) was added to a solution of ethyl 6-(3-(m-tolyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 350 (119 mg, 0.34 mmol) in MeOH (4 mL). The reaction mixture was stirred at 50° C. for 20 min, then cooled to rt. Aq HCl (1 M, 859 μL, 0.86 mmol) was added, and the reaction mixture was stirred, evaporated under reduced pressure and co-evaporated under reduced pressure with EtOH (2×), MeCN (1×), to give the crude title compound; MS (ESI) m/z [M+H]+319.2. Intermediate 352: Ethyl 6-(3-(4-chlorobenzyl)azetidin-1-yl)quinoline-4-carboxylate Ethyl 6-bromoquinoline-4-carboxylate (100 mg, 0.36 mmol), 3-(4-chlorobenzyl)azetidine hydrochloride (117 mg, 0.54 mmol), Cs2CO3(465 mg, 1.43 mmol), XPhos (34 mg, 0.07 mmol) and Pd2(dba)3(33 mg, 0.04 mmol) were weighed into a 5 mL vial. Toluene (2 mL) was added and the reaction mixture was purged with N2(g) for 10 min. The vial was sealed and the reaction mixture was stirred at 110° C. for 1 h. After cooling to rt, water (10 mL) and DCM (15 mL) were added and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The residue was purified by straight phase flash chromatography on silica (gradient: 5-50% of EtOAc in heptane) to give the title compound (110 mg, 81%); MS (ESI) m/z [M+H]+381.22. Intermediate 353: 6-(3-(4-Chlorobenzyl)azetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 525 μL, 0.53 mmol) was added to a solution of ethyl 6-(3-(4-chlorobenzyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 352 (100 mg, 0.26 mmol) in MeOH (3 mL). The reaction mixture was stirred at 50° C. for 20 min, then cooled to rt. Aq HCl (1 M, 656 μL, 0.66 mmol) was added, and the reaction mixture was stirred, evaporated under reduced pressure, and co-evaporated under reduced pressure with EtOH (2×), and MeCN (1×), to give the crude title compound; MS (ESI) m/z [M+H]+353.2. Intermediate 354: Ethyl 6-(3-methyl-3-((methylsulfonyl)methyl)azetidin-1-yl)quinoline-4-carboxylate Ethyl 6-bromoquinoline-4-carboxylate (100 mg, 0.36 mmol), 3-methyl-3-((methylsulfonyl)methyl)azetidine hydrochloride (143 mg, 0.71 mmol), Cs2CO3(465 mg, 1.43 mmol), XPhos (34 mg, 0.07 mmol) and Pd2(dba)3(33 mg, 0.04 mmol) were weighed into a 5 mL vial. Dioxane (2 mL) was added, and the reaction mixture was purged with N2(g) for 10 min. The vial was sealed and the reaction mixture was stirred at 100° C. for 2 h. After cooling to rt, water (10 mL) and DCM (10 mL) were added, and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The residue was purified by straight phase flash chromatography on silica (gradient: 10-100% of EtOAc in heptane) to give the title compound (77 mg, 60%); MS (ESI) m/z [M+H]+363.21. Intermediate 355: 6-(3-Methyl-3-((methylsulfonyl)methyl)azetidin-1-yl)quinoline-4-carboxylic acid Aq NaOH (1 M, 386 μL, 0.39 mmol) was added to a solution of ethyl 6-(3-methyl-3-((methylsulfonyl)methyl)azetidin-1-yl)quinoline-4-carboxylate Intermediate 354 (70 mg, 0.19 mmol) in MeOH (2 mL). The reaction mixture was stirred at 50° C. for 20 min, then cooled to rt. Aq HCl (3.8 M, 127 μL, 0.48 mmol) was added, and the reaction mixture was stirred, evaporated under reduced pressure, and co-evaporated under reduced pressure with EtOH (2×), and MeCN (1×), to give the crude title compoundMS (ESI) m/z [M+H]+335.1. Intermediate 356: tert-Butyl 6-(4-ethyl-4-hydroxypiperidin-1-yl)quinoline-4-carboxylate Cs2CO3(206 mg, 0.63 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (150 mg, 0.49 mmol), 4-ethylpiperidin-4-ol hydrochloride (105 mg, 0.63 mmol), XPhos (46 mg, 0.10 mmol) and Pd2(dba)3(45 mg, 0.05 mmol) in 1,4-dioxane (10 mL) and the reaction mixture was stirred at 100° C. for 15 h. 4-Ethylpiperidin-4-ol hydrochloride (105 mg, 0.63 mmol), Cs2CO3(206 mg, 0.63 mmol), XPhos (46 mg, 0.10 mmol) and Pd2(dba)3(45 mg, 0.05 mmol) were added and the reaction mixture was stirred at 100° C. for an additional 24 h. The reaction mixture was filtered and the filtrate concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 3:2), to give the title compound (0.147 g, 85%) as a brown gum; MS (ESI) m/z [M+H]+357. Intermediate 357: 6-(4-Ethyl-4-hydroxypiperidin-1-yl)quinoline-4-carboxylic acid HCl in 1,4-dioxane (4 M, 5 mL) was added to a stirred solution of tert-butyl 6-(4-ethyl-4-hydroxypiperidin-1-yl)quinoline-4-carboxylate Intermediate 356 (130 mg, 0.36 mmol) in 1,4-dioxane (5 mL) and the reaction mixture was stirred at 50° C. for 15 h. Volatiles were removed under reduced pressure, to give the title compound (0.105 g, 96%) as a red solid; MS (ESI) m/z [M+H]+301. Intermediate 358: tert-Butyl 6-(4-hydroxy-4-methylpiperidin-1-yl)quinoline-4-carboxylate Cs2CO3(793 mg, 2.43 mmol) was added to a stirred solution of tert-butyl 6-bromoquinoline-4-carboxylate (250 mg, 0.81 mmol), 4-methylpiperidin-4-ol (140 mg, 1.22 mmol), XPhos (58 mg, 0.12 mmol) and Pd2(dba)3(74 mg, 0.08 mmol) in 1,4-dioxane (10 mL) and stirred at 80° C. for 2 h. Volatiles were removed under reduced pressure, diluted with EtOAc, and washed with H2O. The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.20 g, 72%) as a yellow solid; MS (ESI) m/z [M+H]+343. Intermediate 359: 6-(4-Hydroxy-4-methylpiperidin-1-yl)quinoline-4-carboxylic acid A solution of DCM (6 mL) and TFA (3 mL) was added to tert-butyl 6-(4-hydroxy-4-methylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 358 (100 mg, 0.29 mmol) and stirred at rt for 2 h. Volatiles were removed under reduced pressure to give the title compound (0.084 g, 100%) as a yellow solid; MS (ESI) m/z [M+H]+287. Intermediate 360: tert-Butyl 6-(4-ethyl-4-methoxypiperidin-1-yl)quinoline-4-carboxylate Cs2CO3(634 mg, 1.95 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (200 mg, 0.65 mmol), 4-ethyl-4-methoxypiperidine hydrochloride (128 mg, 0.71 mmol), XPhos (93 mg, 0.19 mmol) and Pd2(dba)3(59 mg, 0.06 mmol) in 1,4-dioxane (10 mL) and stirred at 80° C. for 20 h. Volatiles were removed under reduced pressure, diluted with EtOAc (20 mL), and washed with H2O (20 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.20 g, 83%) as a yellow solid; MS (ESI) m/z [M+H]+371. Intermediate 361: 6-(4-Ethyl-4-methoxypiperidin-1-yl)quinoline-4-carboxylic acid A solution of DCM (10 mL) and TFA (3 mL) was added to tert-butyl 6-(4-ethyl-4-methoxypiperidin-1-yl)quinoline-4-carboxylate Intermediate 360 (100 mg, 0.29 mmol) and stirred at rt for 5 h. The solvent was removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.085 g, 100%) as a solid; MS (ESI) m/z [M+H]+315. Intermediate 362: tert-Butyl 6-(4-hydroxy-4-isopropylpiperidin-1-yl)quinoline-4-carboxylate Cs2CO3(275 mg, 0.84 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (200 mg, 0.65 mmol), 4-isopropylpiperidin-4-ol (121 mg, 0.84 mmol), XPhos (62 mg, 0.13 mmol) and Pd2(dba)3(59 mg, 0.06 mmol) in 1,4-dioxane (10 mL) and stirred at 100° C. for 15 h. The reaction mixture was filtered, the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 3:2), to give the title compound (0.209 g, 87%) as a brown gum; MS (ESI) m/z [M+H]+371. Intermediate 363: 6-(4-Hydroxy-4-isopropylpiperidin-1-yl)quinoline-4-carboxylic acid HCl in 1,4-dioxane (4 M, 5 mL) was added slowly to a stirred solution of tert-butyl 6-(4-hydroxy-4-isopropylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 362 (185 mg, 0.50 mmol) in 1,4-dioxane (5 mL) at 28° C. and the reaction mixture was stirred at 50° C. for 15 h. Volatiles were removed under reduced pressure, to give the title compound (0.155 g, 91%) as a red solid; MS (ESI) m/z [M+H]+315. Intermediate 364: tert-Butyl (3R,4s,5S)-4-hydroxy-3,4,5-trimethylpiperidine-1-carboxylate MeMgBr (120 mL, 360 mmol) was slowly added to a stirred solution of tert-butyl (3R,5S)-3,5-dimethyl-4-oxopiperidine-1-carboxylate (30.8 g, 136 mmol) in THF (500 mL) at 0° C. The reaction mixture was allowed to return to rt and was stirred overnight. The reaction was quenched with a sat NH4Cl (aq) and extracted with EtOAc (3×400 mL). The combined organic layers were concentrated to give the title compound (33 g, 100%) as a white solid;1H NMR (300 MHz, CDCl3) δ 3.95-3.55 (m, 2H), 2.80-2.55 (m, 2H), 1.58-1.50 (m, 2H), 1.50 (s, 9H), 1.18 (s, 3H), 0.92-0.90 (m, 6H). Intermediate 365: (3R,4s,5S)-3,4,5-Trimethylpiperidin-4-ol Methanolic HCl (400 mL) was added to a solution of tert-butyl (3R,4s,5S)-4-hydroxy-3,4,5-trimethylpiperidine-1-carboxylate Intermediate 364 (33 g, 0.0.23 mol) in MeOH (100 mL). The reaction mixture was stirred at rt for 5 h and then concentrated to give the title compound (24.5 g, 100%) as a white solid; MS (ESI) m/z [M+H]+144.1. Intermediate 366: tert-Butyl 6-((3R,4s,5S)-4-hydroxy-3,4,5-trimethylpiperidin-1-yl)quinoline-4-carboxylate Cs2CO3(137 mg, 0.42 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (100 mg, 0.32 mmol), (3R,4s,5S)-3,4,5-trimethylpiperidin-4-ol Intermediate 365 (76 mg, 0.42 mmol), XPhos (30 mg, 0.06 mmol) and Pd2(dba)3(30 mg, 0.03 mmol) in 1,4-dioxane (5 mL) at 25° C. The resulting suspension was stirred at 100° C. for 23 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.118 g, 98%) as an orange solid; MS (ESI) m/z [M+H]+371. Intermediate 367: 6-((3R,4s,5S)-4-Hydroxy-3,4,5-trimethylpiperidin-1-yl)quinoline-4-carboxylic acid HCl in dioxane (4 M, 5 mL) was added slowly to a stirred solution of tert-butyl 6-((3R,4s,5S)-4-hydroxy-3,4,5-trimethylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 366 (105 mg, 0.28 mmol) in 1,4-dioxane (5 mL) at 20° C. and the reaction mixture was stirred at 50° C. for 3 h. Volatiles were removed under reduced pressure, to give the title compound (0.85 g, 95%) as a dark red solid; MS (ESI) m/z [M+H]+315. Intermediate 368: Ethyl 6-((1R,5S)-9-oxa-3-azabicyclo[3.3.1]nonan-3-yl)quinoline-4-carboxylate A mixture of ethyl 6-bromoquinoline-4-carboxylate (0.098 g, 0.35 mmol), 9-oxa-3-azabicyclo[3.3.1]nonane hydrochloride (0.074 g, 0.46 mmol), RuPhos Pd G4 (30 mg, 0.04 mmol), Cs2CO3(0.342 g, 1.05 mmol) and dioxane (0.9 mL) was degassed by 5×vacuum/N2(g) cycles and stirred vigorously at 90° C. for 17 h. The reaction mixture was allowed to return to rt and was diluted with EtOAc (3 mL) and stirred with SilaMet S-thiol scavenger (0.15 g; 1.4 mmol/g) at rt for 7 h. The mixture was filtered through Celite® 521. The filter was washed with EtOAc and the combined filtrates were concentrated. The residue was purified by preparative HPLC, PrepMethod G (gradient: 35-75%) to give the title compound (0.085 g, 75%) as a yellow syrup; MS (ESI) m/z [M+H]+327.3. Intermediate 369: 6-((1R,5S)-9-Oxa-3-azabicyclo[3.3.1]nonan-3-yl)quinoline-4-carboxylic acid NaOH (3.8 M, 156 μL, aq) was added to a solution of ethyl 6-((1R,5S)-9-oxa-3-azabicyclo[3.3.1]nonan-3-yl)quinoline-4-carboxylate Intermediate 368 (77 mg, 0.24 mmol) in MeOH (2 mL) and the reaction mixture was stirred at 50° C. for 1 h. Aq HCl (3 M, 125 μL) was added dropwise and the resulting mixture was concentrated and lyophilized from MeCN/H2O give the title compound (0.097 g) as a red solid; MS (ESI) m/z [M+H]+299.22. Intermediate 370: 4-(4-Chloroquinolin-6-yl)-1-methylpiperidin-4-ol 6-Bromo-4-chloroquinoline (1 g, 4 mmol) was dissolved in THF (25 mL) in a dried two necked flask under argon, cooled to −70° C., and n-BuLi (2.47 mL, 6.19 mmol) was slowly added dropwise so that the internal temperature did not exceed −65° C. The reaction mixture was stirred for 1 h at −70° C. 1-Methylpiperidin-4-one (0.70 g, 6.2 mmol) in THF (3 mL) was slowly added dropwise so that the internal temperature did not exceed −65° C. The reaction mixture was stirred for 1 h at −70° C. and then allowed to return to rt and stirred for 3 h. The reaction mixture was diluted with EtOAc, and washed with H2O. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by preparative TLC (DCM:MeOH; 10:1), to give the title compound (0.30 g, 26%) as a pale yellow solid; MS (ESI) m/z [M+H]+277. Intermediate 371: Methyl 6-(4-hydroxy-1-methylpiperidin-4-yl)quinoline-4-carboxylate TEA (0.45 mL, 3.3 mmol) was added to a stirred solution of 4-(4-chloroquinolin-6-yl)-1-methylpiperidin-4-ol Intermediate 370 (300 mg, 1.08 mmol), Pd(OAc)2(24 mg, 0.11 mmol), PdCl2(dppf) (40 mg, 0.05 mmol) and dppf (90 mg, 0.16 mmol) in MeOH (15 mL). The resulting mixture was stirred under an atmosphere of CO (g) at 10 atm of pressure and 100° C. for 12 h. The reaction mixture was filtered and the filtrate was concentrated under vacuum. The residue was purified by preparative TLC (DCM:MeOH; 10:1), to give the title compound (0.25 g, 77%) as a pale yellow solid; MS (ESI) m/z [M+H]+301. Intermediate 372: Methyl 6-(4-fluoro-1-methylpiperidin-4-yl)quinoline-4-carboxylate DAST (0.22 mL, 1.7 mmol) in DCM (2 mL) was added dropwise to a solution of methyl 6-(4-hydroxy-1-methylpiperidin-4-yl)quinoline-4-carboxylate Intermediate 371 (250 mg, 0.83 mmol) in DCM (10 mL) at 0° C. The reaction mixture was allowed to return to rt and stirred for 2 h. The solvent was removed under reduced pressure and the reaction mixture was diluted with EtOAc, and washed with H2O. The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (DCM:MeOH; 10:1), to give the title compound (0.25 g, 53%) as a white solid; MS (ESI) m/z [M+H]+303. Intermediate 373: 6-(4-Fluoro-1-methylpiperidin-4-yl)quinoline-4-carboxylic acid Methyl 6-(4-fluoro-1-methylpiperidin-4-yl)quinoline-4-carboxylate Intermediate 372 (230 mg, 0.55 mmol) and LiOH (132 mg, 5.52 mmol) was dissolved in a mixture of MeOH (8 mL) and H2O (2 mL). The reaction mixture was stirred at rt for 2 h and volatiles were removed under reduced pressure. The reaction mixture was diluted with H2O and pH 6 was set with aq HCl (0.1 M). EtOAc was added, and the mixture was washed with H2O. The organic layer was dried over Na2SO4, filtered and evaporated to give the title compound (0.145 g, 91%) as a white solid; MS (ESI) m/z [M+H]+289. Intermediate 374: 6-(1H-Pyrazol-5-yl)quinoline-4-carboxylic acid A mixture of 1-(tetrahydro-2H-pyran-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (59 mg, 0.21 mmol), 6-bromoquinoline-4-carboxylic acid (53 mg, 0.21 mmol), Cs2CO3(137 mg, 0.42 mmol) and Pd(dtbpf)Cl2(14 mg, 0.02 mmol) in 1,4-dioxane (1 mL) and H2O (0.250 mL) was stirred at rt overnight. The reaction mixture was diluted with DMSO and concentrated under reduced pressure. The compound was purified by preparative HPLC, PrepMethod E (gradient: 5-45%) to give the title compound (32 mg, 64%); MS (ESI) m/z [M+H]+240.2. Intermediate 375: 6-(1-Isopropyl-1H-pyrazol-5-yl)quinoline-4-carboxylic acid A mixture of 1-isopropyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (48 mg, 0.20 mmol), 6-bromoquinoline-4-carboxylic acid (51 mg, 0.20 mmol), Cs2CO3(198 mg, 0.61 mmol) and Pd(dtbpf)Cl2(13 mg, 0.02 mmol in 1,4-dioxane (1 mL) and H2O (0.25 mL) was stirred at rt overnight. The reaction mixture concentrated under reduced pressure to give the title compound (56 mg); MS (ESI) m/z [M+H]+282.2. Intermediate 376: rac-tert-Butyl (R)-6-(3-fluoro-3-methylpyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(587 mg, 1.80 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (185 mg, 0.60 mmol), 3-fluoro-3-methylpyrrolidine hydrochloride (168 mg, 1.20 mmol) and Pd2(dba)3(55 mg, 0.06 mmol), XPhos (57 mg, 0.12 mmol) in 1,4-dioxane (4 mL). The resulting suspension was stirred at 100° C. for 2 h. The reaction mixture was diluted with EtOAc and H2O (50 mL) and extracted with EtOAc (3×50 mL). The organic layers were combined and washed with brine (150 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.188 g, 95%) as a brown gum; MS (ESI) m/z [M+H]+331. Intermediate 377: rac-(R)-6-(3-Fluoro-3-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (2 mL) was added to rac-tert-butyl (R)-6-(3-fluoro-3-methylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 376 (187 mg, 0.57 mmol) in DCM (2 mL) at 10° C. and stirred overnight. Volatiles were removed under reduced pressure to give the title compound (0.494 g) as a red gum; MS (ESI) m/z [M+H]+275. Intermediate 378: rac-(R)-6-(3-Methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(244 mg, 0.75 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (133 mg, 0.50 mmol), 3-methylpyrrolidin-2-one (64 mg, 0.65 mmol) and XPhos Pd G3 (42 mg, 0.05 mmol) in 1,4-dioxane (7 mL) at 20° C. The resulting suspension was stirred at 100° C. for 2 h and filtered through a Celite® pad. The filtrate was concentrated under reduced pressure and the resulting residue was purified by preparative TLC (EtOAc:petroleum ether; 3:1) to give the title compound (0.55 g) as a beige solid; MS (ESI) m/z [M+H]+285. Intermediate 379: rac-(R)-6-(3-Methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxylic acid NaOH (366 mg, 9.14 mmol) was added to a stirred solution of rac-(R)-6-(3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 378 (520 mg, 1.83 mmol) in MeOH (9 mL) and H2O (3 mL) at 20° C. and stirred for 1 h. The reaction mixture was acidified with aq HCl (2 M). The aq layer was extracted with EtOAc (6×50 mL), the combined organic layers were washed with H2O (3×25 mL) and the organic layer was dried over Na2SO4, filtered and evaporated to give the title compound (0.31 g, 63%) as a beige oil; MS (ESI) m/z [M+H]+271. Intermediate 380: rac-tert-Butyl (R)-6-(3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxylate Cs2CO3(634 mg, 1.95 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (400 mg, 1.30 mmol), 3-methylpiperidin-2-one (588 mg, 5.19 mmol) and Pd2(dba)3(12 mg, 0.01 mmol), XPhos (12 mg, 0.03 mmol) in 1,4-dioxane (20 mL). The resulting suspension was stirred at 100° C. for 24 h. The reaction mixture was filtered through silica and volatiles were removed under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 4:1), to give the title compound (0.33 g, 75%) as a brown solid; MS (ESI) m/z [M+H]+341.20. Intermediate 381: rac-(R)-6-(3-Methyl-2-oxopiperidin-1-yl)quinoline-4-carboxylic acid TFA (0.27 mL, 3.5 mmol) was added to a solution of rac-tert-butyl (R)-6-(3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxylate Intermediate 380 (300 mg, 0.88 mmol) in DCM (5 mL) and stirred at 25° C. for 6 h. Volatiles were removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.33 g) as a brown solid; MS (ESI) m/z [M+H]+285.1. Intermediate 382: tert-Butyl 6-(4-fluoropiperidin-1-yl)quinoline-4-carboxylate Cs2CO3(977 mg, 3.00 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), 4-fluoropiperidine hydrochloride (153 mg, 1.10 mmol), and Pd2(dba)3(92 mg, 0.10 mmol), XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (1 mL). The resulting suspension was stirred at 100° C. for 2 h and filtered through Celite®. The solvent was removed under reduced pressure and the residue was purified by preparative TLC (EtOAc:petroleum ether; 2:1), to give the title compound (0.33 g, 100%) as a yellow gum; MS (ESI) m/z [M+H]+331. Intermediate 383: 6-(4-Fluoropiperidin-1-yl)quinoline-4-carboxylic acid tert-Butyl 6-(4-fluoropiperidin-1-yl)quinoline-4-carboxylate Intermediate 382 (320 mg, 0.97 mmol) was added to a mixture of TFA (4.5 mL) in DCM (4.5 mL) and stirred at 20° C. for 15 h. Volatiles were removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.674 g) as a dark red gum; MS (ESI) m/z [M+H]+275. Intermediate 384: tert-Butyl 6-(3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylate Cs2CO3(977 mg, 3.00 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), 3-azabicyclo[3.1.0]hexane hydrochloride (239 mg, 2.00 mmol) and Pd2(dba)3(92 mg, 0.10 mmol), XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (3 mL). The resulting suspension was stirred at 100° C. for 2 h. The reaction mixture was diluted with DCM and concentrated under reduced pressure. The reaction mixture was partitioned between H2O and EtOAc, extracted with EtOAc (3×20 mL) and the organic phases washed with brine (20 mL). The organic phases were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.281 g, 91%) as a yellow solid; MS (ESI) m/z [M+H]+311. Intermediate 385: 6-(3-Azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylic acid tert-Butyl 6-(3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylate Intermediate 384 (320 mg, 0.97 mmol) was added to a mixture of TFA (3 mL) in DCM (3 mL) and stirred at 10° C. overnight. The solvent was removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.435 g) as a red gum; MS (ESI) m/z [M+H]+255. Intermediate 386: Methyl (S)-6-(3-methoxypyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(735 mg, 2.25 mmol) was added a mixture of methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), (S)-3-methoxypyrrolidine (171 mg, 1.69 mmol) and Pd2(dba)3(10 mg, 0.01 mmol), XPhos (11 mg, 0.02 mmol) in 1,4-dioxane (10 mL). The resulting suspension was stirred at 100° C. for 2 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.298 g, 92%) as a yellow solid; MS (ESI) m/z [M+H]+287. Intermediate 387: (S)-6-(3-Methoxypyrrolidin-1-yl)quinoline-4-carboxylic acid Methyl (S)-6-(3-methoxypyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 386 (270 mg, 0.94 mmol) and LiOH (132 mg, 5.52 mmol) was dissolved in a mixture of THF (5 mL) and H2O (5 mL) under air. The reaction mixture was stirred at 25° C. for 3 h. The solvent was removed under reduced pressure and the reaction mixture was diluted with H2O and pH 5 was set with aq HCl (2 M). The reaction mixture was concentrated under vacuum to give the title compound (0.20 g, 78%) as a red solid; MS (ESI) m/z [M+H]+273.1. Intermediate 388: tert-Butyl 6-((1R,5S,6r)-6-(trifluoromethyl)-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylate Cs2CO3(977 mg, 3.00 mmol) was added a stirred suspension of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), (1R,5S,6r)-6-(trifluoromethyl)-3-azabicyclo[3.1.0]hexane hydrochloride (375 mg, 2.00 mmol) and Pd2(dba)3(92 mg, 0.10 mmol), XPhos (95 mg, 0.20 mmol) in 1,4-dioxane (3 mL). The resulting suspension was stirred at 100° C. for 2 h. The reaction mixture was diluted with DCM and concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (50 mL) and washed with H2O (3×50 mL), the organic layer dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.455 g, 92%) as a brown solid; MS (ESI) m/z [M+H]+379. Intermediate 389: 6-((1R,5S,6r)-6-(Trifluoromethyl)-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylic acid tert-Butyl 6-((1R,5S,6r)-6-(trifluoromethyl)-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylate Intermediate 388 (454 mg, 1.20 mmol) was added to a mixture of TFA (5 mL) in DCM (5 mL) and stirred at 13° C. overnight under air. The solvent was removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.728 g) as a red gum; MS (ESI) m/z [M+H]+323. Intermediate 390: Methyl 6-(7-azabicyclo[2.2.1]heptan-7-yl)quinoline-4-carboxylate Cs2CO3(1.47 g, 4.51 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 7-azabicyclo[2.2.1]heptane hydrochloride (201 mg, 1.50 mmol) and Pd Catalyst [CAS: 1810068-35-9] (43 mg, 0.04 mmol) in 1,4-dioxane (3 mL). The resulting suspension was stirred at 100° C. for 2 days. The reaction mixture was diluted with H2O (50 mL), extracted with EtOAc (3×50 mL), the organic layers were washed with brine (200 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.039 g, 18%) as a yellow solid; MS (ESI) m/z [M+H]+283. Intermediate 391: 6-(7-Azabicyclo[2.2.1]heptan-7-yl)quinoline-4-carboxylic acid Methyl 6-(7-azabicyclo[2.2.1]heptan-7-yl)quinoline-4-carboxylate Intermediate 390 (36 mg, 0.13 mmol) and NaOH (25 mg, 0.63 mmol) was dissolved in a mixture of MeOH (3 mL) and H2O (1 mL) under air. The reaction mixture was stirred at 10° C. for 1 h. The solvent was removed under reduced pressure. The reaction mixture was diluted with H2O (20 mL) and pH 3 was set with aq HCl (1 M). The reaction mixture was diluted with H2O (10 mL) and extracted with EtOAc (6×30 mL), dried over Na2SO4, filtered and concentrated under vacuum. The material was purified by preparative HPLC, PrepMethod P (gradient: 5-17%) to give the title compound (0.030 g, 89%) as a yellow solid; MS (ESI) m/z [M+H]+269. Intermediate 392: rac-tert-Butyl 6-((1R,4S)-2-azabicyclo[2.2.1]heptan-2-yl)quinoline-4-carboxylate Cs2CO3(651 mg, 2.00 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (154 mg, 0.50 mmol), 2-azabicyclo[2.2.1]heptane (97 mg, 1.0 mmol) and Pd Catalyst [CAS: 1810068-35-9] (29 mg, 0.02 mmol) in 1,4-dioxane (5 mL). The resulting suspension was stirred at 100° C. for 24 h. 2-Azabicyclo[2.2.1]heptane (97 mg, 1.00 mmol) and Cs2CO3(651 mg, 2.00 mmol) was added and the suspension was stirred at 100° C. for an additional 18 h. The reaction mixture was diluted with EtOAc (10 mL) and filtered through Celite®, the filter pad was washed with EtOAc (3×2 mL) and the combined filtrate was concentrated under vacuum. The residue was purified by preparative TLC (EtOAc:petroleum ether; 2:1), to give the title compound (0.059 g, 36%) as a yellow gum; MS (ESI) m/z [M+H]+325. Intermediate 393: rac-6-((1R,4S)-2-Azabicyclo[2.2.1]heptan-2-yl)quinoline-4-carboxylic acid tert-Butyl 6-(2-azabicyclo[2.2.1]heptan-2-yl)quinoline-4-carboxylate Intermediate 392 (50 mg, 0.15 mmol) was added to a mixture of TFA (3 mL) in DCM (5 mL) and stirred at 10° C. overnight under air. Volatiles were removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.11 g) as a dark purple gum; MS (ESI) m/z [M+H]+269. Intermediate 394: tert-Butyl (R)-6-(2-methylpyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(634 mg, 1.95 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (200 mg, 0.65 mmol), (R)-2-methylpyrrolidine (111 mg, 1.30 mmol) and Pd Catalyst [CAS: 1810068-35-9] (37 mg, 0.03 mmol) in 1,4-dioxane (5 mL). The resulting suspension was stirred at 100° C. overnight. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 3:1), to give the title compound (0.19 g, 94%) as a yellow gum; MS (ESI) m/z [M+H]+313.3. Intermediate 395: (R)-6-(2-Methylpyrrolidin-1-yl)quinoline-4-carboxylic acid tert-Butyl (R)-6-(2-methylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 394 (190 mg, 0.61 mmol) was added to a mixture of TFA (69 mg, 0.61 mmol) in DCM (5 mL) and stirred at 25° C. overnight under air. The solvent was removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.18 g) as a brown solid; MS (ESI) m/z [M+H]+257. Intermediate 396: tert-Butyl (S)-6-(2-(methoxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(952 mg, 2.92 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), (S)-2-(methoxymethyl)pyrrolidine (224 mg, 1.95 mmol) and Pd Catalyst [CAS: 1810068-35-9] (56 mg, 0.05 mmol) in 1,4-dioxane (15 mL). The resulting suspension was stirred at 100° C. for 8 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 2:1), to give the title compound (0.18 g, 54%) as a yellow solid; MS (ESI) m/z [M+H]+343. Intermediate 397: (S)-6-(2-(Methoxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylic acid tert-Butyl (S)-6-(2-(methoxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 396 (180 mg, 0.53 mmol) was added to a mixture of TFA (5 mL) in DCM (10 mL) and stirred at rt for 6 h under air. The solvent was removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.15 g, 100%) as a dark yellow; MS (ESI) m/z [M+H]+287. Intermediate 398: tert-Butyl 6-((3S,4S)-3,4-Difluoropyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(952 mg, 2.92 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), (3S,4S)-3,4-difluoropyrrolidine (115 mg, 1.07 mmol) and Pd2(dba)3(89 mg, 0.10 mmol), XantPhos (113 mg, 0.19 mmol) in 1,4-dioxane (6 mL). The resulting suspension was stirred at 100° C. for 2 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.30 g, 92%) as a red solid; MS (ESI) m/z [M+H]+335.3. Intermediate 399: 6-((3S,4S)-3,4-Difluoropyrrolidin-1-yl)quinoline-4-carboxylic acid tert-Butyl 6-((3S,4S)-3,4-Difluoropyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 398 (300 mg, 0.90 mmol) was added to a mixture of TFA (512 mg, 4.49 mmol) in DCM (5 mL) and stirred at 25° C. overnight under air. The solvent was removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.3 g) as a solid; MS (ESI) m/z [M+H]+279.2. Intermediate 400: rac-tert-Butyl 6-((3R,4R)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(952 mg, 2.92 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), rac-(3R,4R)-3,4-difluoropyrrolidine (115 mg, 1.07 mmol) and Pd2(dba)3(89 mg, 0.10 mmol), XantPhos (113 mg, 0.19 mmol) in 1,4-dioxane (8 mL). The resulting suspension was stirred at 100° C. for 2 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 2:1), to give the title compound (0.27 g, 83%) as a yellow solid; MS (ESI) m/z [M+H]+335.3. Intermediate 401: rac-6-((3R,4R)-3,4-Difluoropyrrolidin-1-yl)quinoline-4-carboxylic acid rac-tert-Butyl 6-((3R,4R)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 400 (260 mg, 0.78 mmol) was added to a mixture of TFA (443 mg, 3.89 mmol) in DCM (5 mL) and stirred at 25° C. for 5 h under air. The solvent was removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.23 g, 100%) as a solid; MS (ESI) m/z [M+H]+279.2. Intermediate 402: 6-(2,4-Dimethyloxazol-5-yl)quinoline-4-carboxylic acid A mixture of 2,4-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)oxazole (54 mg, 0.24 mmol), 6-bromoquinoline-4-carboxylic acid (61 mg, 0.24 mmol), Cs2CO3(237 mg, 0.73 mmol) and Pd(dtbpf)Cl2(13 mg, 0.02 mmol) in 1,4-dioxane (2 mL) and H2O (0.5 mL) was stirred at rt overnight. The reaction mixture concentrated under reduced pressure and purified by preparative HPLC, PrepMethod E (gradient: 5-45%) to give the title compound (18 mg, 28%); MS (ESI) m/z [M+H]+269. Intermediate 403: 6-(3,5-Dimethylisoxazol-4-yl)quinoline-4-carboxylic acid A mixture of (3,5-dimethylisoxazol-4-yl)boronic acid (37 mg, 0.26 mmol), 6-bromoquinoline-4-carboxylic acid (60 mg, 0.24 mmol), Cs2CO3(194 mg, 0.60 mmol) and Pd(dtbpf)Cl2(16 mg, 0.02 mmol) in 1,4-dioxane (2 mL) and H2O (0.5 mL) was stirred under argon at rt overnight. H2O was added to the reaction mixture and the aq layer was extracted with EtOAc. The aq layer was acidified with aq HCl (2 M) to pH 3 and concentrated under reduced pressure to give the title compound that was used in the next step without further purification; MS (ESI) m/z [M+H]+269.2. Intermediate 404: 6-(2-Phenyl-1H-imidazol-1-yl)quinoline-4-carboxylic acid A mixture of 2-phenyl-1H-imidazole (86 mg, 0.60 mmol), 6-bromoquinoline-4-carboxylic acid (100 mg, 0.40 mmol), Cs2CO3(194 mg, 0.60 mmol) and Cu2O (6 mg, 0.04 mmol) in DMF (3 mL) was stirred at 150° C. for 14 h. The pH of the reaction mixture was adjusted to pH 6 with aq HCl (1 M) and filtered through a Celite® pad. The reaction mixture was concentrated under reduced pressure and purified by preparative HPLC, PrepMethod B (gradient: 2-30%) to give the title compound (0.033 g, 26%) as a yellow solid; MS (ESI) m/z [M+H]+316. Intermediate 405: 6-(3-Phenyl-1H-pyrrol-1-yl)quinoline-4-carboxylic acid K2CO3(208 mg, 1.50 mmol) was added a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 3-phenyl-1H-pyrrole (161 mg, 1.13 mmol) and CuI (14 mg, 0.08 mmol) in DMF (10 mL). The resulting suspension was stirred at 150° C. for 15 h. The reaction mixture was concentrated under reduced pressure and diluted with H2O, and pH 6 was set with aq HCl (1 M). The formed precipitate was collected by filtration, washed with H2O (30 mL) and the collected brown solid was dissolved in DMF (15 mL), solids were filtered off and the filtrate was concentrated under reduced pressure to give the title compound (0.15 g, 64%) as a brown solid; MS (ESI) m/z [M+H]+315. Intermediate 406: 6-(4,5,6,7-Tetrahydro-1H-indol-1-yl)quinoline-4-carboxylic acid A mixture of 4,5,6,7-tetrahydro-1H-indole (216 mg, 1.79 mmol), 6-bromoquinoline-4-carboxylic acid (300 mg, 1.19 mmol), Cs2CO3(1.16 g, 3.57 mmol) and EPhos Pd G4 (109 mg, 0.12 mmol) in 1,4-dioxane (4 mL) was stirred at 100° C. overnight. A second reaction batch was set up. A mixture of 4,5,6,7-tetrahydro-1H-indole (144 mg, 1.19 mmol), 6-bromoquinoline-4-carboxylic acid (200 mg, 0.79 mmol), Cs2CO3(776 mg, 2.38 mmol) and EPhos Pd G4 (73 mg, 0.08 mmol) in 1,4-dioxane (3 mL) was stirred at 100° C. overnight. The reaction mixture of the two batches were combined and filtered through Celite®, pH 6 was set with aq HCl (2 M) and the filtrate was filtered through Celite® and concentrated. The residue was purified by preparative TLC (DCM:MeOH; 5:1), to give the title compound (0.055 g, 14%) as a yellow solid; MS (ESI) m/z [M+H]+293. Intermediate 407: tert-Butyl (R)-6-(3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(634 mg, 1.95 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), (R)-pyrrolidin-3-ylmethanol (118 mg, 1.17 mmol) and Pd2(dba)3(89 mg, 0.10 mmol), XantPhos (77 mg, 0.19 mmol) in 1,4-dioxane (15 mL). The resulting suspension was stirred at 100° C. for 5 h. The reaction mixture was concentrated under reduced pressure and diluted with EtOAc (125 mL). The organic layer was washed with H2O (2×75 mL), sat NH4Cl (75 mL, aq), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH; 10:1), to give the title compound (0.28 g, 88%) as a yellow oil; MS (ESI) m/z [M+H]+329.1. Intermediate 408: (R)-6-(3-(Hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylic acid HCl in 1,4-dioxane (4 M, 8 mL) was added slowly to tert-butyl (R)-6-(3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 407 (200 mg, 0.61 mmol) and the reaction mixture was stirred at 20° C. for 12 h. Volatiles were removed under reduced pressure, and the reaction mixture was diluted with EtOAc (75 mL). The organic layer was washed with H2O (15 mL), brine (20 mL), dried over Na2SO4, filtered and concentrated to give the title compound (0.10 g, 60%) as a red solid; MS (ESI) m/z [M+H]+273. Intermediate 409: tert-Butyl (S)-6-(3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(846 mg, 2.60 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (400 mg, 1.30 mmol), (S)-pyrrolidin-3-ylmethanol (158 mg, 1.56 mmol) and Pd2(dba)3(119 mg, 0.13 mmol), XantPhos (102 mg, 0.26 mmol) in 1,4-dioxane (3 mL). The resulting suspension was stirred at 100° C. for 5 h. The reaction mixture was concentrated under reduced pressure and diluted with EtOAc (125 mL). The organic layer was washed with H2O (50 mL), brine (75 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH; 10:1), to give the title compound (0.357 g, 84%) as a yellow solid; MS (ESI) m/z [M+H]+329.1. Intermediate 410: (S)-6-(3-(Hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylic acid HCl in 1,4-dioxane (4 M, 5 mL) was added slowly to tert-butyl (S)-6-(3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 409 (200 mg, 0.61 mmol) and the reaction mixture was stirred at 20° C. for 12 h. Volatiles were removed under reduced pressure, and the reaction mixture was diluted with EtOAc (75 mL). The organic layer was washed with H2O (25 mL), brine (25 mL), dried over Na2SO4, filtered and concentrated to give the title compound (0.12 g, 72%) as a red solid; MS (ESI) m/z [M+H]+273.05. Intermediate 411: Methyl 6-(1-thia-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylate Cs2CO3(367 mg, 1.13 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (150 mg, 0.56 mmol), 1-thia-6-azaspiro[3.3]heptane hydrochloride (171 mg, 1.13 mmol) and RuPhos Pd G3 (94 mg, 0.11 mmol) in 1,4-dioxane (10 mL) at 35° C. The resulting suspension was stirred at 100° C. for 15 h. Cs2CO3(367 mg, 1.13 mmol), 1-thia-6-azaspiro[3.3]heptane hydrochloride (171 mg, 1.13 mmol) and RuPhos Pd G3 (94 mg, 0.11 mmol) was added and the reaction mixture stirred at 100° C. for 20 h. The reaction mixture was filtered and the filtrate concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 6:5) to give the title compound (0.098 g, 58%) as a brown gum; MS (ESI) m/z [M+H]+301. Intermediate 412: 6-(1-Thia-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylic acid To a solution of methyl 6-(1-thia-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylate Intermediate 411 (84 mg, 0.28 mmol) dissolved in MeOH (6 mL) was added a solution of NaOH (56 mg, 1.4 mmol) in H2O (2 mL) under air at 0° C. The reaction mixture was stirred at 30° C. for 1 h. The reaction mixture was diluted with H2O (10 mL) and pH 6 was set with aq HCl (2 M). The reaction mixture was diluted with H2O (10 mL) and extracted with EtOAc (6×75 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give the title compound (0.075 g, 94%) as an orange solid; MS (ESI) m/z [M+H]+287. Intermediate 413: Methyl 6-(4-hydroxy-4-phenylpiperidin-1-yl)quinoline-4-carboxylate Cs2CO3(490 mg, 1.50 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 4-phenylpiperidin-4-ol (266 mg, 1.50 mmol), XPhos (72 mg, 0.15 mmol) and Pd2(dba)3(34 mg, 0.04 mmol) in 1,4-dioxane (8 mL). The resulting suspension was stirred at 90° C. for 2 h and filtered. The filtrate was concentrated under reduced pressure. EtOAc (25 mL) was added and the mixture was washed with water (2×15 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 2:1), to give the title compound (0.20 g, 73%) as a yellow solid; MS (ESI) m/z [M+H]+285. Intermediate 414: Methyl 6-(4-fluoro-4-phenylpiperidin-1-yl)quinoline-4-carboxylate DAST (146 μL, 1.10 mmol) was added dropwise to a solution of methyl 6-(4-hydroxy-4-phenylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 413 (200 mg, 0.55 mmol) in DCM (10 mL) at −50° C. and stirred for 3 h. Volatiles were removed under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:2), to give the title compound (0.15 g, 75%) as a yellow solid; MS (ESI) m/z [M+H]+365. Intermediate 415: 6-(4-Fluoro-4-phenylpiperidin-1-yl)quinoline-4-carboxylic acid Methyl 6-(4-fluoro-4-phenylpiperidin-1-yl)quinoline-4-carboxylate Intermediate 414 (140 mg, 0.38 mmol) and LiOH (18 mg, 0.77 mmol) was dissolved in a mixture of MeOH (10 mL) and H2O (1 mL). The reaction mixture was stirred at 30° C. for 2 h and volatiles were removed under reduced pressure. The reaction mixture was diluted with H2O (15 mL) and pH 6 was set with aq HCl (1 M). The precipitate was collected by filtration, washed with H2O and dried under vacuum to give the title compound (0.115 g, 85%) as a yellow solid; MS (ESI) m/z [M+H]+351. Intermediate 416: rac-tert-Butyl (R)-6-(3,3-difluoro-4-hydroxypyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(634 mg, 1.95 mmol) was added to a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (300 mg, 0.97 mmol), 4,4-difluoropyrrolidin-3-ol (144 mg, 1.17 mmol), DavePhos (77 mg, 0.19 mmol) and Pd2(dba)3(89 mg, 0.10 mmol) in 1,4-dioxane (15 mL). The resulting suspension was stirred at 100° C. for 2 h and filtered. The reaction mixture was diluted with EtOAc (125 mL) and washed with water (75 mL), brine (75 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH; 5:1), to give the title compound (0.28 g, 82%) as a yellow solid; MS (ESI) m/z [M+H]+351.15. Intermediate 417: rac-(R)-6-(3,3-Difluoro-4-hydroxypyrrolidin-1-yl)quinoline-4-carboxylic acid A solution of DCM (5 mL) and TFA (1.5 mL) was added to rac-tert-butyl (R)-6-(3,3-difluoro-4-hydroxypyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 416 (160 mg, 0.46 mmol) and stirred at 20° C. for 16 h. Volatiles were removed under reduced pressure and the reaction mixture was diluted with DCM (75 mL), washed with H2O (25 mL), brine (25 mL), dried over Na2SO4, filtered and evaporated to give the title compound (0.12 g, 89%) as a red solid; MS (ESI) m/z [M+H]+295.0. Intermediate 418: rac-tert-Butyl 6-((3R,4R)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(977 mg, 3.00 mmol) was added to a stirred solution of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), rac-(3R,4R)-3,4-dimethylpyrrolidine hydrochloride (149 mg, 1.10 mmol), XPhos (95 mg, 0.20 mmol)) and Pd2(dba)3(92 mg, 0.10 mmol) in 1,4-dioxane (5 mL) and stirred at 100° C. for 2 h. The reaction mixture was filtered, and the filtrate washed with H2O (3 mL). The aq layer was extracted with EtOAc (4×20 mL), the combined organic layers were washed with H2O (3×10 mL). The organic layers was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.307 g, 94%) as a yellow solid; MS (ESI) m/z [M+H]+327. Intermediate 419: rac-6-((3R,4R)-3,4-Dimethylpyrrolidin-1-yl)quinoline-4-carboxylic acid TFA (5 mL) was added to a solution of rac-tert-butyl 6-((3R,4R)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 418 (270 mg, 0.83 mmol) in DCM (5 mL) and stirred at 6° C. for 15 h. Volatiles were removed under reduced pressure to give the title compound (0.493 g, 97%) as a dark red gum; MS (ESI) m/z [M+H]+271. Intermediate 420: rac-tert-Butyl (R)-6-(2-cyclopropylpyrrolidin-1-yl)quinoline-4-carboxylate Cs2CO3(977 mg, 3.00 mmol) was added a mixture of tert-butyl 6-bromoquinoline-4-carboxylate (308 mg, 1.00 mmol), rac-(R)-2-cyclopropylpyrrolidine hydrochloride (162 mg, 1.10 mmol) and Pd Catalyst [CAS: 1810068-35-9] (57 mg, 0.05 mmol) in 1,4-dioxane (5 mL). The resulting suspension was stirred at 100° C. for 48 h. The reaction mixture was diluted with EtOAc (10 mL) and filtered through Celite®, the filter pad was washed with EtOAc (3×2 mL) and the combined filtrates were concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether; 1:1), to give the title compound (0.192 g, 57%) as a yellow solid; MS (ESI) m/z [M+H]+339. Intermediate 421: rac-(R)-6-(2-Cyclopropylpyrrolidin-1-yl)quinoline-4-carboxylic acid rac-tert-Butyl (R)-6-(2-cyclopropylpyrrolidin-1-yl)quinoline-4-carboxylate Intermediate 420 (180 mg, 0.53 mmol) was added to a mixture of TFA (5 mL) in DCM (5 mL) and stirred at 10° C. for 15 h. Volatiles were removed under reduced pressure and the solid was further dried under vacuum to give the title compound (0.37 g, 100%) as a red gum; MS (ESI) m/z [M+H]+283. Intermediate 422: Methyl 6-bromoquinoline-4-carboxylate hydrochloride A solution of 6-bromoquinoline-4-carboxylic acid (3.0 g, 12 mmol) in MeOH (70 mL) was added dropwise to stirred SOCl2(7.08 g, 59.5 mmol) over a period of 25 min. The reaction mixture was heated at 60° C. for 13 h. The reaction mixture was concentrated at reduced pressure to give the title compound (3.50 g, 97%) as a yellow solid; MS (ESI) m/z [M+H]+266. Intermediate 423: Methyl 6-(1,5-dioxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylate Cs2CO3(490 mg, 1.50 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 1,5-dioxa-9-azaspiro[5.5]undecane (236 mg, 1.50 mmol), Pd2(dba)3(69 mg, 0.08 mmol) and SPhos (62 mg, 0.15 mmol) in 1,4-dioxane (4.0 mL) at 15° C. and the reaction mixture was stirred at 100° C. for 2 h under an atmosphere of N2(g). The solvent was removed under reduced pressure. The residue was diluted with water (30 mL), and the water phase was extracted with EtOAc (3×50 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative TLC (EtOAc:petroleum ether, 1:1), to give the title compound (0.25 g, 97%) as an orange gum; MS (ESI) m/z [M+H]+343.1. Intermediate 424: 6-(1,5-Dioxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylic acid NaOH (146 mg, 3.65 mmol) was added to a solution of methyl 6-(1,5-dioxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylate Intermediate 423 (250 mg, 0.73 mmol) in MeOH (9.0 mL) and water (3.0 mL) at 15° C. and the reaction mixture was stirred at 15° C. for 1 h. The solvent was removed under reduced pressure. The residue was diluted with water (10 mL), and pH was adjusted to 3 with aq HCl (1 M). The aqueous phase was extracted with EtOAc (3×50 mL), and the combined organic layer was dried over Na2SO4, filtered and evaporated to give the title compound (0.221 g, 92%) as an orange solid; MS (ESI) m/z [M+H]+329.0. Intermediate 425: Methyl 6-(1,4-dioxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxylate Cs2CO3(490 mg, 1.50 mmol) was added to a mixture of methyl 6-bromoquinoline-4-carboxylate (200 mg, 0.75 mmol), 1,4-dioxa-8-azaspiro[4.5]decane (215 mg, 1.50 mmol), Pd2(dba)3(69 mg, 0.08 mmol) and SPhos (62 mg, 0.15 mmol) in 1,4-dioxane (5.0 mL) at 25° C., and the reaction mixture was stirred at 100° C. for 2 h under an atmosphere of N2(g). The solvent was removed under reduced pressure. The residue was diluted with water (50 mL), and the water phase was extracted with EtOAc (3×50 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The residue was purified by preparative TLC (EtOAc:petroleum ether, 1:1), to give the title compound (0.104 g, 42%) as a brown gum; MS (ESI) m/z [M+H]+329.0. Intermediate 426: 6-(1,4-Dioxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxylic acid NaOH (60 mg, 1.50 mmol) was added to a solution of methyl 6-(1,4-dioxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxylate Intermediate 425 (99 mg, 0.30 mmol) in MeOH (9.0 mL) and water (3.0 mL) at 15° C., and the reaction mixture was stirred at 15° C. for 1 h. The solvent was removed under reduced pressure. The residue was diluted with water (20 mL) and the pH was adjusted to 3 with aq HCl (1 M). The aqueous phase was extracted with EtOAc (3×50 mL), and the combined organic layer was dried over Na2SO4, filtered and evaporated to give the crude title compound (0.137 g) as an orange solid; MS (ESI) m/z [M+H]+315. Intermediate 427: Methyl 6-(2-oxa-7-azaspiro[3.5]nonan-7-yl)quinoline-4-carboxylate Cs2CO3(918 mg, 2.82 mmol) was added to methyl 6-bromoquinoline-4-carboxylate (300 mg, 1.13 mmol), 2-oxa-7-azaspiro[3.5]nonane oxalate (490 mg, 2.25 mmol) and SPhos Pd G3 (98 mg, 0.11 mmol) in 1,4-dioxane (5 mL) at 15° C., and the reaction mixture was stirred at 100° C. for 16 h under an atmosphere of N2(g). Cs2CO3(367 mg, 1.13 mmol) and SPhos Pd G3 (98 mg, 0.11 mmol) were added and the reaction mixture was stirred at 100° C. for a further 5 h. The solvent was removed under reduced pressure. The residue was diluted with water (50 mL) and extracted with EtOAc (3×50 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (EtOAc:petroleum ether, 2:1), to give the title compound (0.142 g, 40%) as a brown gum; MS (ESI) m/z [M+H]+313.1. C. Final Compounds Example 1: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,2-oxazinan-2-yl)-quinoline-4-carboxamide DIPEA (0.15 mL, 0.87 mmol) was added to a mixture of 6-(1,2-oxazinan-2-yl)quinoline-4-carboxylic acid Intermediate 6 (45 mg, 0.17 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (54 mg, 0.26 mmol) and HATU (79 mg, 0.21 mmol) in EtOAc (1.5 mL) and MeCN (1.5 mL). The mixture was stirred at rt for 4 h. The mixture was diluted with EtOAc, washed sequentially with water and sat NaHCO3. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-10% MeOH in EtOAc). Appropriate fractions were pooled and evaporated to give a yellow oil. The oil was dissolved in MeCN:water and freeze dried overnight to give the title compound (65 mg, 91%) as a fluffy yellow powder; HRMS (ESI) m/z [M+H]+calcd for C20H22N5O3S: 412.1438 found: 412.1438;1H NMR (500 MHz, DMSO-d6) δ 9.02 (brs, 1H), 8.78 (d, 1H), 7.95 (d, 1H), 7.85 (d, 1H), 7.62 (dd, 1H), 7.48 (d, 1H), 5.34-5.31 (m, 1H), 4.88 (d, 1H), 4.71 (d, 1H), 4.31 (d, 2H), 4.09 (t, 2H), 3.55 (t, 2H), 3.46-3.35 (m, 2H), 1.91-1.83 (m, 2H), 1.72-1.65 (m, 2H). Example 2: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-3-fluoro-6-morpholino-quinoline-4-carboxamide DIPEA (0.33 mL, 1.9 mmol) was added to a mixture of 3-fluoro-6-morpholinoquinoline-4-carboxylic acid Intermediate 8 (172 mg, 0.62 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (129 mg, 0.62 mmol) and HATU (308 mg, 0.81 mmol) in EtOAc (3 mL) and MeCN (3 mL). The mixture which was stirred at rt for 3 h. The mixture was diluted with EtOAc, washed sequentially with water and sat NaHCO3. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 75-100% EtOAc in heptane), and then further purified using preparative HPLC, PrepMethod SFC-D, to give the title compound (60 mg, 22%); HRMS (ESI) m/z [M+H]+calcd for C20H21FN5O3S: 430.1344 found: 430.1318;1H NMR (600 MHz, DMSO-d6) δ 9.28 (t, 1H), 8.70 (s, 1H), 7.93 (d, 1H), 7.65 (dd, 2H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.41 (dd, 1H), 4.32 (dd, 1H), 3.80 (t, 4H), 3.44-3.36 (m, 6H). Example 3: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-thiomorpholinoquinoline-4-carboxamide DIPEA (0.16 mL, 0.91 mmol) was added to a mixture of 6-thiomorpholinoquinoline-4-carboxylic acid Intermediate 10 (50 mg, 0.18 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (38 mg, 0.18 mmol) and HATU (83 mg, 0.22 mmol) in EtOAc (1.5 mL) and MeCN (1.5 mL). The mixture was stirred at rt overnight. The mixture was diluted with EtOAc, washed sequentially with water and sat NaHCO3. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-10% MeOH in EtOAc). Appropriate fractions were pooled and evaporated to give a yellow oil. The oil was dissolved in MeCN/water and freeze dried overnight to give the title compound (55 mg, 71%) as a fluffy yellow powder; HRMS (ESI) m/z [M+H]+calcd for C20H22N5O2S2: 428.1210 found: 428.1210;1H NMR (500 MHz, DMSO-d6) δ 9.01 (t, 1H), 8.67 (d, 1H), 7.89 (d, 1H), 7.73 (d, 1H), 7.64 (dd, 1H), 7.39 (d, 1H), 5.33 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.37-4.25 (m, 2H), 3.79 (t, 4H), 3.44-3.35 (m, 2H), 2.71 (ddt, 4H). Example 4: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-difluoromorpholino)-quinoline-4-carboxamide DIPEA (0.13 mL, 0.75 mmol) was added to a mixture of 6-(2,2-difluoromorpholino)quinoline-4-carboxylic acid Intermediate 12 (74 mg, 0.25 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (52 mg, 0.25 mmol) and HATU (124 mg, 0.33 mmol) in EtOAc (2 mL) and MeCN (2 mL). The mixture was stirred at rt for 3 h. The mixture was diluted with EtOAc, washed sequentially with water and sat NaHCO3. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-10% MeOH in EtOAc), and then further purified using preparative HPLC, PrepMethod SFC-D, to give the title compound (40 mg, 36%); HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O3S: 448.1250 found: 448.1252;1H NMR (600 MHz, DMSO-d6) δ 9.08 (t, 1H), 8.76 (d, 1H), 7.97 (d, 1H), 7.84 (d, 1H), 7.75 (dd, 1H), 7.47 (d, 1H), 5.32 (dd, 1H), 4.91 (d, 1H), 4.73 (d, 1H), 4.38-4.29 (m, 2H), 4.29-4.23 (m, 2H), 3.89-3.80 (m, 2H), 3.61-3.54 (m, 2H), 3.45-3.35 (m, 2H). Example 5: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2,6,6-tetrafluoromorpholino)quinoline-4-carboxamide DIPEA (0.14 mL, 0.83 mmol) was added to a mixture of 6-(2,2,6,6-tetrafluoromorpholino)quinoline-4-carboxylic acid Intermediate 14 (55 mg, 0.17 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (52 mg, 0.25 mmol) and HATU (76 mg, 0.20 mmol) in EtOAc (1.5 mL) and MeCN (1.5 mL). The mixture was stirred at rt overnight. The mixture was diluted with EtOAc, and washed sequentially with water and sat NaHCO3. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-10% MeOH in EtOAc), and then further purified using preparative HPLC, PrepMethod SFC-D, to give the title compound (20 mg, 25%); HRMS (ESI) m/z [M+H]+calcd for C20H18F4N5O3S: 484.1060 found: 484.1052;1H NMR (600 MHz, DMSO-d6) δ 9.11 (t, 1H), 8.80 (d, 1H), 8.01 (d, 1H), 7.98 (d, 1H), 7.81 (dd, 1H), 7.48 (d, 1H), 5.28 (dd, 1H), 4.92 (d, 1H), 4.73 (d, 1H), 4.40-4.30 (m, 2H), 4.29-4.21 (m, 4H), 3.47-3.36 (m, 2H, overlapping with solvent). Example 6: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,3-dihydro-4H-benzo[b][1,4]oxazin-4-yl)quinoline-4-carboxamide DIPEA (0.14 mL, 0.80 mmol) was added to a mixture of 6-(2,3-dihydro-4H-benzo[b][1,4]oxazin-4-yl)quinoline-4-carboxylic acid Intermediate 16 (49 mg, 0.16 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (50 mg, 0.24 mmol) and HATU (73 mg, 0.19 mmol) in EtOAc (1.5 mL) and MeCN (1.5 mL). The mixture was stirred at rt overnight. The mixture was diluted with EtOAc and washed sequentially with water and sat NaHCO3. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-10% MeOH in EtOAc). Appropriate fractions were pooled and evaporated to give a yellow oil. The oil was dissolved in MeCN/water and freeze dried overnight to give the title compound (59 mg, 80%) as a fluffy yellow powder; HRMS (ESI) m/z [M+H]+calcd for C24H22N5O3S: 460.1438 found: 460.1428;1H NMR (500 MHz, DMSO-d6) δ 9.06 (t, 1H), 8.84 (d, 1H), 8.11 (d, 1H), 8.02 (d, 1H), 7.81 (dd, 1H), 7.53 (d, 1H), 7.05 (dd, 1H), 6.88 (dd, 1H), 6.85-6.72 (m, 2H), 5.32 (dd, 1H), 4.87 (d, 1H), 4.70 (d, 1H), 4.30 (t, 4H), 3.86 (q, 2H), 3.39 (dd, 2H). Example 7: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,4S,5R)-4-hydroxy-3,5-dimethylpiperidin-1-yl)quinoline-4-carboxamide DIPEA (0.052 mL, 0.30 mmol) was added to a mixture of 6-((3S,4s,5R)-4-hydroxy-3,5-dimethylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 18 (18 mg, 0.06 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (19 mg, 0.09 mmol) and HATU (27 mg, 0.07 mmol) in EtOAc (1.5 mL) and MeCN (1.5 mL). The mixture was stirred at rt overnight, and then diluted with EtOAc, washed sequentially with water and sat NaHCO3. The organic phase was dried, filtered and evaporated. The residue was purified by straight phase flash chromatography on silica (gradient: 0-10% MeOH in EtOAc), and then further purified by preparative HPLC, PrepMethod SFC-A, to give the title compound (5 mg, 18%); HRMS (ESI) m/z [M+H]+calcd for C23H28N5O3S: 454.1908 found: 454.1886;1H NMR (600 MHz, DMSO-d6) δ 8.98 (t, 1H), 8.63 (d, 1H), 7.86 (d, 1H), 7.70-7.60 (m, 2H), 7.37 (d, 1H), 5.30 (dd, 1H), 4.89 (d, 1H), 4.73 (d, 1H), 4.48 (brs, 1H), 4.35-4.25 (m, 2H), 3.60-3.50 (m, 3H), 2.74 (t, 4H), 1.85-1.75 (m, 2H), 0.98 (dd, 6H). Example 8: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxypiperidin-1-yl)quinoline-4-carboxamide DIPEA (0.24 mL, 1.4 mmol) was added to 6-(4-methoxypiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 20 (200 mg, 0.70 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (179 mg, 1.05 mmol) and HATU (266 mg, 0.70 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 20° C. under air. The resulting mixture was stirred at 20° C. for 4 h. The reaction mixture was concentrated, and the residue was dissolved in EtOAc (100 mL), and washed sequentially with sat NaHCO3(25 mL), sat brine (50 mL), and water (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 12-37%) to give the title compound (70 mg, 23%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750 found: 440.1744;1H NMR (500 MHz, DMSO-d6) δ 9.14 (t, 1H), 8.78 (d, 1H), 7.95 (d, 1H), 7.84-7.71 (m, 2H), 7.57 (d, 1H), 5.33 (dd, 1H), 4.91 (d, 1H), 4.72 (d, 1H), 4.40-4.27 (m, 2H), 3.79-3.71 (m, overlapping with solvent), 3.47-3.33 (m, 3H), 3.29 (s, 3H), 3.22-3.07 (m, 2H), 2.03-1.94 (m, 2H), 1.60-1.50 (m, 2H). Example 9: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-8-methyl-6-(4-methoxy-piperidin-1-yl)morpholinoquinoline-4-carboxamide DIPEA (0.205 mL, 1.18 mmol) was added to 8-methyl-6-morpholinoquinoline-4-carboxylic acid Intermediate 25 (160 mg, 0.59 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (151 mg, 0.88 mmol) and HATU (223 mg, 0.59 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 20° C. The resulting mixture was stirred at 20° C. for 3 h. The reaction mixture was concentrated and diluted with EtOAc (75 mL), and washed sequentially with sat NaHCO3(20 mL), water (15 mL), and sat brine (20 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford crude product. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 12-35%) to give the title compound (50 mg, 20%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594 found: 426.1600;1H NMR (400 MHz, DMSO-d6) δ 9.17 (t, 1H), 8.81 (d, 1H), 7.77 (s, 1H), 7.68-7.62 (m, 2H), 5.32 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.42-4.25 (m, 2H), 3.79 (t, 4H), 3.46-3.32 (m, overlapping with solvent), 2.73 (s, 3H). Example 10: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-7-methyl-6-morpholino-quinoline-4-carboxamide DIPEA (0.31 mL, 1.8 mmol) was added to 7-methyl-6-morpholinoquinoline-4-carboxylic acid Intermediate 30 (160 mg, 0.59 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (151 mg, 0.88 mmol) and T3P (50% solution in EtOAc, 712 mg, 2.24 mmol) in EtOAc (1 mL) and MeCN (1 mL) at 20° C. under N2(g). The resulting mixture was stirred at 20° C. for 4 h. The reaction mixture was concentrated, and the residue was dissolved in EtOAc (75 mL), and washed sequentially with sat brine (25 mL) and water (25 mL). The organic layer was dried over Na2SO4, filtered, and evaporated. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 12-35%) to give the title compound (50 mg, 20%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594 found: 426.1582;1H NMR (400 MHz, DMSO-d6) δ 9.05 (t, 1H), 8.80 (d, 1H), 7.91 (s, 1H), 7.88 (s, 1H), 7.45 (d, 1H), 5.35 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.32 (t, 2H), 3.79 (t, 4H), 3.45-3.35 (m, overlapping with solvent), 3.10-2.90 (m, 4H), 2.54-2.46 (m, overlapping with solvent). Example 11: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxopyrrolidin-1-yl)-quinoline-4-carboxamide DIPEA (1.8 mL, 10 mmol) was added to a stirred suspension of 6-(2-oxopyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 32 (224 mg, 0.52 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (214 mg, 1.03 mmol), HOBt (395 mg, 2.58 mmol) and EDC (494 mg, 2.58 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 29° C. The resulting solution was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The residue was dissolved in a mixture of NaHCO3(aq, 40 mL) and EtOAc (80 mL). The phases were separated and the aqueous layer was extracted with EtOAc (5×75 mL). The organic layers were combined and washed with water (3×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford crude product. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 15-30%) to give the title compound (90 mg, 43%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C20H20N5O3S: 410.1282 found: 410.1288;1H NMR (300 MHz, DMSO-d6) δ 9.10 (t, 1H), 8.90 (d, 1H), 8.55 (dd, 1H), 8.31-8.16 (m, 1H), 8.07 (d, 1H), 7.56 (d, 1H), 5.43-5.24 (m, 1H), 4.90 (m, 1H), 4.77 (d, 1H), 4.50-4.16 (m, 2H), 4.07-3.89 (m, 2H), 3.70-3.20 (m, overlapping with solvent) 2.57 (t, 2H), 2.22-2.02 (m, 2H). Example 12: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (0.22 mL, 1.3 mmol) was added to a mixture of 6-(3,3-dimethyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 34 (120 mg, 0.42 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (108 mg, 0.63 mmol) and T3P (50% solution in EtOAc, 1.07 g, 1.69 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 20° C. under N2(g). The resulting mixture was stirred at 20° C. for 3 h. The reaction mixture was concentrated and diluted with EtOAc (100 mL), and washed sequentially with water (20 mL) and sat brine (20 mL). The organic layer was dried over Na2SO4, filtered, and evaporated to afford crude product. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 25-40%) to afford the title compound (100 mg, 54%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O3S: 438.1594 found: 438.1616;1H NMR (400 MHz, CD3OD) δ 8.93-8.85 (m, 1H), 8.66-8.59 (m, 1H), 8.45 (d, 1H), 8.10 (d, 1H), 7.68 (d, 1H), 5.35 (dd, 1H), 4.95-4.83 (m, overlapping with solvent), 4.79 (d, 1H), 4.43 (s, 2H), 4.17-4.00 (m, 2H), 3.60-3.35 (m, 2H), 2.12 (t, 2H), 1.27 (s, 3H), 1.26 (s, 3H). Example 13: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,5-dimethyl-2-oxooxazolidin-3-yl)quinoline-4-carboxamide DIPEA (0.37 mL, 2.10 mmol) was added to a mixture of 6-(5,5-dimethyl-2-oxooxazolidin-3-yl)quinoline-4-carboxylic acid Intermediate 36 (200 mg, 0.70 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (179 mg, 1.05 mmol) and T3P (50% solution in EtOAc, 1.78 g, 2.79 mmol) in EtOAc (8 mL) and MeCN (8 mL) at 20° C. under air. The resulting mixture was stirred at 25° C. for 3 h. The reaction mixture was concentrated and diluted with EtOAc (75 mL), and washed sequentially with water (20 mL) and sat brine (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford crude product. The crude product was purified by preparative HPLC, PrepMethod B, (gradient: 20-40%) to afford the title compound (70 mg, 23%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O4S: 440.1388 found: 440.1380;1H NMR (400 MHz, DMSO-d) δ 9.12 (t, 1H), 8.90 (d, 1H), 8.57 (dd, 1H), 8.21 (d, 1H), 8.10 (d, 1H), 7.55 (d, 1H), 5.33 (dd, 1H), 4.91 (d, 1H), 4.72 (d, 1H), 4.37-4.31 (m, 2H), 4.10-3.99 (m, 2H), 3.50-3.33 (m, overlapping with solvent), 1.55 (s, 3H), 1.52 (s, 3H). Example 14: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxopiperidin-1-yl)-quinoline-4-carboxamide DIPEA (230 mg, 1.78 mmol) was added to a stirred mixture of 6-(2-oxopiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 38 (120 mg, 0.44 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (152 mg, 0.89 mmol) and T3P (1.13 mL, 50% in EtOAc) in DMF (5 mL). The resulting solution was stirred at 25° C. for 6 h. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH, 19:1), followed by further purification by preparative HPLC, PrepMethod B, (gradient 13-33%) to give the title compound (48 mg, 26%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438 found: 424.1444;1H NMR (300 MHz, DMSO-d6) δ 9.12 (t, 1H), 8.95 (d, 1H), 8.28 (d, 1H), 8.02 (d, 1H), 7.78 (dd, 1H), 7.56 (d, 1H), 5.33 (dd, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.31 (d, 2H), 3.85-3.65 (m, 2H), 3.43-3.34 (m, overlapping with solvent), 2.46-2.35 (m, overlapping with solvent), 2.00-1.78 (m, 4H). Example 15: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide DIPEA (104 mg, 0.80 mmol) was added to a mixture of 6-(3,3-dimethyl-2-oxopiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 40 (60 mg, 0.20 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (69 mg, 0.40 mmol) and T3P (0.51 mL, 50% in EtOAc) in DMF (2 mL). The resulting solution was stirred at 25° C. for 6 h. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH, 18:1), followed by further purification by preparative HPLC, PrepMethod B, (gradient 22-42%) to give the title compound (25 mg, 28%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O3S: 452.1750 found: 452.1752;1H NMR (300 MHz, DMSO-d6) δ 9.12 (t, 1H), 8.94 (d, 1H), 8.29 (d, 1H), 8.01 (d, 1H), 7.69 (dd, 1H), 7.55 (d, 1H), 5.32 (dd, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.31 (d, 2H), 3.77 (t, 2H), 3.43-3.34 (m, overlapping with solvent), 2.04-1.91 (m, 2H), 1.88-1.75 (m, 2H), 1.24 (s, 3H), 1.24 (s, 3H). Example 16: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethyl-3-oxomorpholino)quinoline-4-carboxamide DIPEA (0.88 mL, 5.1 mmol) was added to a mixture of 6-(2,2-dimethyl-3-oxomorpholino)quinoline-4-carboxylic acid Intermediate 42 (304 mg, 1.01 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (260 mg, 1.52 mmol) and HATU (1.15 g, 3.04 mmol) in EtOAc (10 mL) and MeCN (10 mL) at 20° C. The resulting mixture was stirred at 25° C. for 6 h. The reaction mixture was diluted with sat NaHCO3. The phases were separated and the aqueous layer was extracted with DCM (3×100 mL). The combined organic phases were dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (DCM:MeOH, 17:1), followed by further purification by preparative HPLC, PrepMethod C, (gradient 17-27%) to give the title compound (200 mg, 44%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O4S: 454.1544 found: 454.1542;1H NMR (300 MHz, DMSO-d6) δ 9.16 (t, 1H), 8.99 (d, 1H), 8.42 (d, 1H), 8.08 (d 1H), 7.86 (dd, 1H), 7.60 (d, 1H), 5.45-5.25 (m, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.34 (d, 2H), 4.11-3.96 (m, 2H), 3.94-3.80 (m, 2H), 3.59-3.35 (m, overlapping with solvent), 1.47 (s, 6H). Example 17: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide DIPEA (289 mg, 2.24 mmol) was added to a mixture of 6-(2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid Intermediate 44 (100 mg, 0.37 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (96 mg, 0.56 mmol) and HATU (283 mg, 0.75 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction was stirred under an atmosphere of air at 25° C. for 3 h. The solvent was removed under reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient 20-40%) to give the title compound (44 mg, 28%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O2S: 422.1646 found: 422.1650;1H NMR (300 MHz, DMSO-d6) δ 9.20-9.05 (m, 1H), 8.75 (d, 1H), 7.96 (d, 1H), 7.61 (d, 1H), 7.30-7.15 (m, 2H), 5.40-5.30 (m, 1H), 4.91 (d, 1H), 4.73 (d, 1H), 4.34 (d, 2H), 3.99 (s, 4H), 3.48-3.32 (m, 2H), 2.21 (t, 4H), 1.95-1.75 (m, 2H). Example 18: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide DIPEA (0.50 mL, 2.9 mmol) was added to a mixture of 6-(3,3-dimethyl-1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylic acid Intermediate 46 (170 mg, 0.57 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (178 mg, 0.85 mmol), EDC (218 mg, 1.14 mmol) and HOBt (175 mg, 1.14 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction was stirred at 50° C. for 3 h. The solvent was removed under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 15-25%) to give the title compound (160 mg, 62%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O3S: 452.1750 found: 452.1748;1H NMR (300 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.62 (d, 1H), 7.87 (d, 1H), 7.38 (d, 1H), 7.28 (d, 1H), 7.13 (dd, 1H), 5.35-5.25 (m, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.35-4.20 (m, 4H), 4.16 (s, 2H), 3.94-3.83 (m, 2H), 3.42-3.34 (m, 2H), 1.22 (s, 6H). Example 19: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-azaspiro[3.3]heptan-1-yl)quinoline-4-carboxamide DIPEA (0.65 mL, 3.7 mmol) was added to a mixture of 6-(1-azaspiro[3.3]heptan-1-yl)quinoline-4-carboxylic acid Intermediate 48 (200 mg, 0.75 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (232 mg, 1.12 mmol), EDC (286 mg, 1.49 mmol) and HOBt (228 mg, 1.49 mmol) in EtOAc (6 mL) and MeCN (6 mL). The mixture was stirred at 45° C. for 5 h. The solvent was removed under reduced pressure. The residue was dissolved in EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod B, (gradient: 28-48%) to afford the title compound (220 mg, 70%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O2S: 422.1646 found: 422.1626;1H NMR (300 MHz, CD3OD) δ 8.55 (d, 1H), 7.89 (d, 1H), 7.49 (d, 1H), 7.37 (dd, 1H), 7.28 (d, 1H), 5.40-5.25 (m, 1H), 4.85-4.63 (m, 2H), 4.37 (d, 2H), 3.87 (t, 2H), 3.51-3.34 (m, 2H), 3.04-2.88 (m, 2H), 2.51 (t, 2H), 2.17-2.03 (m, 2H), 2.02-1.70 (m, 2H). Example 20: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylazetidin-1-yl)quinoline-4-carboxamide TEA (0.62 mL, 4.5 mmol) was added to a stirred suspension of 6-(2,2-dimethylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 50 (114 mg, 0.44 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (185 mg, 0.89 mmol), HOBt (341 mg, 2.22 mmol) and EDC (426 mg, 2.22 mmol) in EtOAc (13 mL) and MeCN (13 mL) at 10° C. The resulting suspension was stirred at 10° C. overnight. The solvent was removed under reduced pressure. The residue was dissolved with a mixture of a solution of NaHCO3(50 mL) and EtOAc (100 mL). The aqueous layer was extracted with EtOAc (4×100 mL). The organic layers were combined and washed with water (2×50 mL). The aqueous layers were combined and extracted with EtOAc (2×25 mL). The organic layers were combined, dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod B, (gradient: 34-48%) to afford the title compound (82 mg, 45%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O2S: 410.1646 found: 410.1640;1H NMR (300 MHz, DMSO-d6) δ 9.00-8.89 (m, 1H), 8.58 (d, 1H), 7.83 (d, 1H), 7.37 (d, 1H), 7.14 (dd, 1H), 7.09 (d, 1H), 5.30 (dd, 1H), 4.88 (d, 1H), 4.71 (d, 1H), 4.28 (d, 2H), 3.75 (t, 2H), 3.35 (m, overlapping with solvent), 2.18-2.03 (m, 1H), 1.49 (s, 3H), 1.48 (s, 3H). Example 21: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoroazetidin-1-yl)quinoline-4-carboxamide DIPEA (0.53 mL, 3.1 mmol) was added to a mixture of 6-(3-fluoroazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 52 (150 mg, 0.61 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (253 mg, 1.22 mmol), EDC (234 mg, 1.22 mmol) and HOBt (187 mg, 1.22 mmol) in EtOAc (5 mL) and MeCN (5 mL). The mixture was stirred at rt for 10 h. The solvent was removed under reduced pressure. The residue was dissolved in EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 17-29%) to afford the title compound (110 mg, 45%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C19H19FN5O2S: 400.1238 found: 400.1246;1H NMR (300 MHz, CD3OD) δ 8.60 (d, 1H), 7.91 (d, 1H), 7.50 (d, 1H), 7.32 (d, 1H), 7.18 (dd, 1H), 5.64-5.32 (m, 2H), 4.83-4.69 (m, overlapping with solvent), 4.50-4.25 (m, 4H), 4.20-4.00 (m, 2H), 3.52-3.34 (m, overlapping with solvent). Example 22: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylazetidin-1-yl)quinoline-4-carboxamide DIPEA (0.58 mL, 3.3 mmol) was added to a suspension of 6-(3,3-dimethylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 54 (170 mg, 0.66 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (344 mg, 1.66 mmol), EDC (254 mg, 1.33 mmol) and HOBt (203 mg, 1.33 mmol) in MeCN (10 mL) and EtOAc (10 mL). The mixture was stirred at rt for 20 h. The solvent was removed under reduced pressure. The residue was dissolved in EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 19-30%) to afford the title compound (90 mg, 33%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O2S: 410.1646 found: 410.1642;1H NMR (300 MHz, CD3OD) δ 8.54 (d, 1H), 7.87 (d, 1H), 7.48 (d, 1H), 7.21-7.07 (m, 2H), 5.40-5.28 (m, 1H), 4.82-4.59 (m, 2H), 4.38 (d, 2H), 3.74 (s, 4H), 3.53-3.35 (m, 2H), 1.35 (s, 6H). Example 23: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoroazetidin-1-yl)quinoline-4-carboxamide DIPEA (1.38 mL, 7.87 mmol) was added to 6-(3,3-difluoroazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 56 (104 mg, 0.39 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (123 mg, 0.59 mmol), HOBt (603 mg, 3.94 mmol) and EDC (755 mg, 3.94 mmol) in EtOAc (5 mL) and MeCN (5 mL) at 10° C. The resulting solution was stirred at 10° C. overnight under N2(g). The solvent was removed under reduced pressure. The residue was diluted with sat NaHCO3(50 mL), and extracted with EtOAc (6×50 mL). The organic layers were combined and washed with sat brine (5×200 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient: 18-45%) to afford the title compound (116 mg, 70%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C19H18F2N5O2S: 418.1144 found: 418.1158;1H NMR (300 MHz, DMSO-d6) δ 9.05-8.95 (m, 1H), 8.72 (d, 1H), 7.96 (d, 1H), 7.55-7.38 (m, 2H), 7.25 (dd, 1H), 5.45-5.30 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.44 (t, 4H), 4.32 (d, 2H), 3.42-3.35 (m, overlapping with solvent). Example 24: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-methyl-azetidin-1-yl)quinoline-4-carboxamide DIPEA (0.43 mL, 2.5 mmol) was added to 6-(3-fluoro-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 58 (65 mg, 0.25 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (103 mg, 0.50 mmol) and TBTU (282 mg, 0.74 mmol) in MeCN (6 mL) and EtOAc (6 mL) at 10° C. The resulting solution was stirred at 40° C. for 4 h under N2(g). The solvent was removed under reduced pressure. The reaction mixture was diluted with sat NaHCO3(50 mL), and extracted with EtOAc (6×50 mL). The organic layers were combined and washed with sat brine (5×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod D, (gradient: 20-45%) to afford the title compound (57 mg, 56%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C20H21FN5O2S: 414.1394 found: 414.1394;1H NMR (300 MHz, DMSO-d6) δ 9.05-8.96 (m, 1H), 8.66 (d, 1H), 7.91 (d, 1H), 7.42 (d, 1H), 7.31 (d, 1H), 7.17 (dd, 1H), 5.40-5.25 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.31 (d, 2H), 4.20-4.00 (m, 4H), 3.45-3.34 (m, overlapping with solvent), 1.66 (d, 3H). Example 25: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methylazetidin-1-yl)-quinoline-4-carboxamide TEA (0.83 g, 8.3 mmol) was added to a mixture of 6-(3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 60 (200 mg, 0.83 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (141 mg, 0.83 mmol), EDC (317 mg, 1.65 mmol) and HOBt (253 mg, 1.65 mmol) in EtOAc (2.5 mL) and MeCN (2.5 mL). The resulting mixture was stirred at 25° C. for 3 h. The reaction mixture was diluted with EtOAc (20 mL), and washed with sat brine (3×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 15-30%) to afford the title compound (190 mg, 58%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C20H22N5O2S: 396.1488 found: 396.1490;1H NMR (300 MHz, CD3OD) δ 8.69 (d, 1H), 7.99 (d, 1H), 7.80 (d, 1H), 7.37 (dd, 1H), 7.34-7.27 (m, 1H), 5.40-5.25 (m, 1H), 4.83-4.65 (m, overlapping with solvent), 4.44 (s, 2H), 4.35-4.18 (m, 3H), 3.77-3.67 (m, 2H), 3.36 (m, overlapping with solvent), 3.00-2.85 (m, 1H), 1.35 (d, 3H). Example 26: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(trifluoromethyl)-azetidin-1-yl)quinoline-4-carboxamide DIPEA (0.59 mL, 3.4 mmol) was added to a mixture of 6-(3-(trifluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 62 (100 mg, 0.34 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (116 mg, 0.68 mmol) and HATU (261 mg, 0.68 mmol) in EtOAc (2 mL) and MeCN (2 mL). The resulting mixture was stirred at 25° C. for 4 h. The solvent was removed under reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 19-30%) to afford the title compound (60 mg, 40%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C20H19F3N5O2S: 450.1206 found: 450.1194;1H NMR (400 MHz, CD3OD) δ 8.79 (brs, 1H), 8.07 (d, 1H), 7.85 (d, 1H), 7.60-7.40 (m, 2H), 5.45-5.25 (m, 1H), 4.79 (d, overlapping with solvent), 4.52-4.35 (m, 4H), 4.28-4.15 (m, 2H), 3.80-3.60 (m, 1H), 3.45-3.34 (m, 2H). Example 27: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide DIPEA (0.25 mL, 1.4 mmol) was added to a mixture of 6-(3-(fluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 64 (268 mg, 0.28 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (118 mg, 0.57 mmol), HOBt (192 mg, 1.42 mmol) and EDC (333 mg, 1.74 mmol) in MeCN (3 mL) and EtOAc (3 mL) at 13° C. The resulting solution was stirred at 13° C. overnight under N2(g). The solvent was removed under reduced pressure. The residue was diluted with sat NaHCO3(100 mL) and extracted with EtOAc (5×100 mL). The organic layers were combined and washed with brine (3×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 17-30%) to afford the title compound (66 mg, 54%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H23FN5O2S: 428.1550 found: 428.1538;1H NMR (400 MHz, DMSO-d6) δ 8.94 (t, 1H), 8.63 (d, 1H), 7.89 (d, 1H), 7.41 (d, 1H), 7.23-7.16 (m, 1H), 7.12 (dd, 1H), 5.40-5.25 (m, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.50 (d, 2H), 4.30 (d, 2H), 3.88 (d, 2H), 3.68 (dd, 2H), 3.44-3.34 (m, overlapping with solvent), 1.35 (s, 3H). Example 28: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)-azetidin-1-yl)quinoline-4-carboxamide DIPEA (0.19 mL, 1.1 mmol) was added to a mixture of 6-(3-(difluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 66 (120 mg, 0.43 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (90 mg, 0.43 mmol), HOBt (99 mg, 0.65 mmol) and EDC (124 mg, 0.65 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 20° C. under N2(g). The resulting mixture was stirred at 25° C. for 3 h. The reaction mixture was concentrated and diluted with DCM (100 mL), and washed sequentially with sat NH4Cl (50 mL), brine (50 mL), and water (50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 12-23%) to afford the title compound (71 mg, 38%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O2S: 432.1300 found: 432.1292;1H NMR (400 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.64 (d, 1H), 7.90 (d, 1H), 7.41 (d, 1H), 7.22 (d, 1H), 7.15 (dd, 1H), 6.39 (td, 1H), 5.33 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.29 (d, 2H), 4.15-4.05 (m, 2H), 3.95-3.85 (m, 2H), 3.44-3.30 (m, overlapping with solvent). Example 29: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(methoxymethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide DIPEA (0.16 mL, 0.91 mmol) was added to a mixture of 6-(3-(methoxymethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 68 (65 mg, 0.23 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (57 mg, 0.27 mmol) and HATU (129 mg, 0.34 mmol) in MeCN (2 mL) and EtOAc (2 mL). The mixture was stirred at rt overnight. DCM (10 mL) and sat NaHCO3(aq, 7 mL) were added to the reaction mixture, and the mixture was stirred and filtered through a phase separator. The phase separator was washed with DCM, and the combined organic layer was evaporated. The crude product was purified by preparative HPLC, PrepMethod SFC-D, (gradient 2-94%) to give the title compound (38 mg, 38%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750 found: 440.1748;1H NMR (600 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.60 (d, 1H), 7.86 (d, 1H), 7.39 (d, 1H), 7.12 (d, 1H), 7.09 (dd, 1H), 5.32 (dd, 1H), 4.87 (d, 1H), 4.71 (d, 1H), 4.28 (d, 2H), 3.80 (dd, 2H), 3.60 (dd, 2H), 3.32-3.41 (m, overlapping with solvent), 3.31 (s, 3H), 1.30 (s, 3H). Example 30: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3R)-3-methoxy-2-methylazetidin-1-yl)quinoline-4-carboxamide DIPEA (0.51 mL, 2.9 mmol) was added to a mixture of 6-((2S,3R)-3-methoxy-2-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 70 (160 mg, 0.59 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (183 mg, 0.88 mmol), EDC (225 mg, 1.18 mmol) and HOBt (180 mg, 1.18 mmol) in EtOAc (5 mL) and MeCN (5 mL). The mixture was stirred at 50° C. for 3 h. The solvent was removed under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 13-23%) to give the title compound (220 mg, 88%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594 found: 426.1576;1H NMR (300 MHz, DMSO-d6) δ 9.01-8.91 (m, 1H), 8.62 (d, 1H), 7.87 (d, 1H), 7.38 (d, 1H), 7.29 (d, 1H), 7.16 (dd, 1H), 5.31 (dd, 1H), 4.88 (d, 1H), 4.71 (d, 1H), 4.35-4.20 (m, 3H), 4.10-3.85 (m, 2H), 3.50-3.29 (m, overlapping with solvent), 3.27 (s, 3H), 1.50 (d, 3H). Example 31: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxamide DIPEA (0.15 mL, 0.87 mmol) was added to a mixture of 6-(3-cyclopropyl-3-fluoroazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 72 (100 mg, 0.35 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (73 mg, 0.35 mmol), EDC (100 mg, 0.52 mmol) and HOBt (71 mg, 0.52 mmol) in EtOAc (8 mL) and MeCN (8 mL) at 20° C. under N2(g). The resulting mixture was stirred at 25° C. for 12 h. The reaction mixture was concentrated and diluted with EtOAc (100 mL), and washed sequentially with brine (50 mL) and water (50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 12-23%) to give the title compound (68 mg, 34%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H23FN5O2S: 440.1550 found: 440.1546;1H NMR (400 MHz, DMSO-d6) δ 9.11 (t, 1H), 8.78 (d, 1H), 7.99 (d, 1H), 7.60 (d, 1H), 7.40 (d, 1H), 7.29 (dd, 1H), 5.34 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.41-4.25 (m, 2H), 4.10-3.95 (m, 2H), 3.46-3.32 (m, 2H), 1.50-1.35 (m, 1H), 0.67-0.61 (2H, m), 0.49 (d, 2H), Example 32: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(piperidin-1-yl)quinoline-4-carboxamide TEA (0.81 mL, 5.9 mmol) was added slowly to a mixture of 6-(piperidin-1-yl)quinoline-4-carboxylic acid Intermediate 74 (60 mg, 0.23 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (194 mg, 0.94 mmol) and T3P (0.57 mL, 50% in EtOAc) in EtOAc (4 mL) at 15° C. under N2(g). The resulting mixture was stirred at 15° C. overnight under N2(g). The reaction mixture was filtered, the filtrate was washed sequentially with brine (3×10 mL) and water (2×10 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 15-30%) to give the title compound (12 mg, 12%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O2S: 410.1646 found: 410.1648;1H NMR (400 MHz, DMSO-d6) δ 9.10-8.90 (m, 1H), 8.66 (d, 1H), 7.86 (d, 1H), 7.72-7.53 (m, 2H), 7.39 (d, 1H), 5.50-5.20 (m, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.36-4.22 (m, 2H), 3.40-3.34 (m, overlapping with solvent), 1.75-1.50 (m, 6H). Example 33: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-dimethylpiperidin-1-yl)quinoline-4-carboxamide TEA (0.34 mL, 2.5 mmol) was added slowly to a mixture of 6-(4,4-dimethylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 76 (70 mg, 0.25 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (102 mg, 0.49 mmol), EDC (142 mg, 0.74 mmol) and HOBt (113 mg, 0.74 mmol) in DMF (5 mL) at 10° C. under N2(g). The resulting suspension was stirred at 10° C. overnight. The reaction mixture was diluted with sat NaHCO3(25 mL), and extracted with EtOAc (4×20 mL). The organic layers were combined and washed with brine (3×10 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 10-25%) to give the title compound (12 mg, 11%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H28N5O2S: 438.1958 found: 438.1946;1H NMR (300 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.63 (d, 1H), 7.84 (d, 1H), 7.74-7.50 (m, 2H), 7.37 (d, 1H), 5.40-5.20 (m, 1H), 4.87 (d, 1H), 4.69 (d, 1H), 4.27 (d, 2H), 3.37-3.32 (m, overlapping with solvent), 1.65-1.35 (m, 4H), 0.96 (s, 6H). Example 34: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-4-methyl-piperidin-1-yl)quinoline-4-carboxamide DIPEA (0.30 mL, 1.7 mmol) was added slowly to a mixture of 6-(4-fluoro-4-methylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 78 (50 mg, 0.17 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (72 mg, 0.35 mmol) and HATU (198 mg, 0.52 mmol) in DMF (5 mL) at 10° C. under N2(g). The resulting mixture was stirred at 10° C. overnight. The reaction mixture was diluted with water (10 mL), and extracted with EtOAc (3×25 mL). The organic layers were combined and washed with brine (3×20 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 10-25%) to give the title compound (13 mg, 17%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H25FN5O2S: 442.1708 found: 442.1726;1H NMR (300 MHz, DMSO-d6) δ 9.05-8.91 (m, 1H), 8.65 (d, 1H), 7.87 (d, 1H), 7.74-7.62 (m, 2H), 7.37 (d, 1H), 5.40-5.20 (m, 1H), 4.87 (d, 1H), 4.69 (d, 1H), 4.32-4.24 (m, 2H), 3.82-3.60 (m, 2H), 3.55-3.07 (m, overlapping with solvent), 1.94-1.62 (m, 4H), 1.35 (d, 3H). Example 35: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-difluoropiperidin-1-yl)quinoline-4-carboxamide DIPEA (0.84 mL, 4.8 mmol) was added to 6-(4,4-difluoropiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 80 (140 mg, 0.48 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (98 mg, 0.57 mmol) and T3P (0.91 mL, 50% in EtOAc) in DMF (8 mL) at 20° C. The resulting mixture was stirred at 50° C. for 35 h. The reaction mixture was diluted with EtOAc (50 mL), and washed with water (3×50 mL), filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 25-55%) to give the title compound (28 mg, 13%) as a pale yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22F2N5O2S: 446.1456 found: 446.1452;1H NMR (300 MHz, DMSO-d6) δ 9.01 (t, 1H), 8.68 (d, 1H), 7.90 (d, 1H), 7.83 (d, 1H), 7.70 (dd, 1H), 7.39 (d, 1H), 5.37-5.20 (m, 1H), 4.88 (d, 1H), 4.69 (d, 1H), 4.40-4.20 (m, 2H), 3.65-3.45 (m, 4H), 3.42-3.33 (m, overlapping with solvent), 2.16-1.94 (m, 4H). Example 36: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoropiperidin-1-yl)quinoline-4-carboxamide TEA (1.05 mL, 7.53 mmol) was added to 6-(3,3-difluoropiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 82 (110 mg, 0.38 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (782 mg, 3.78 mmol) and T3P (2.4 mL, 50% in EtOAc) in DMF (5 mL) at 10° C. under N2(g). The resulting mixture was stirred at 10° C. overnight under N2(g). The reaction mixture was diluted with sat NaHCO3(25 mL), and extracted with EtOAc (4×25 mL). The organic layers were combined and washed with brine (5×20 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 10-25%) to give the title compound (28 mg, 17%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22F2N5O2S: 446.1456 found: 446.1460;1H NMR (300 MHz, DMSO-d6) δ 9.10-8.90 (m, 1H), 8.70 (d, 1H), 8.19-7.86 (m, 1H), 7.84-7.60 (m, 2H), 7.42 (d, 1H), 5.36-5.26 (m, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.30 (d, 2H), 3.70 (t, 2H), 3.55-3.30 (m, overlapping with solvent), 2.18-1.99 (m, 2H), 1.98-1.80 (m, 2H). Example 37: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-(fluoromethyl)-4-methylpiperidin-1-yl)quinoline-4-carboxamide TEA (1.10 mL, 7.87 mmol) was added to 6-(4-(fluoromethyl)-4-methylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 84 (119 mg, 0.39 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (327 mg, 1.57 mmol), HOBt (532 mg, 3.94 mmol) and EDC (755 mg, 3.94 mmol) in DMF (5 mL) at 10° C. under N2(g). The resulting suspension was stirred at 10° C. overnight under N2(g). The reaction mixture was diluted with water (50 mL), and extracted with EtOAc (4×50 mL). The organic layers were combined and washed with brine (5×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 10-25%) to give the title compound (16 mg, 9%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H27FN5O2S: 456.1864 found: 456.1850;1H NMR (300 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.64 (d, 1H), 7.93-7.80 (m, 1H), 7.65 (m, 2H), 7.38 (d, 1H), 5.82-5.23 (m, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.35-4.07 (m, 4H), 3.70-3.00 (m, overlapping with solvent), 1.75-1.55 (m, 2H), 1.53-1.34 (m, 2H), 1.01 (d, 3H). Example 38: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,4-difluoro-3,3-dimethylpiperidin-1-yl)quinoline-4-carboxamide DIPEA (0.22 mL, 1.3 mmol) was added to 6-(4,4-difluoro-3,3-dimethylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 86 (200 mg, 0.62 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (194 mg, 0.94 mmol) and HATU (237 mg, 0.62 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 15° C. under N2(g). The resulting mixture was stirred at 20° C. for 3 h. The reaction mixture was filtered through silica. The filtrate was concentrated and redissolved in DCM (100 mL), and washed sequentially with brine (50 mL) and water (50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod F, (gradient 30-40%) to give the title compound (75 mg, 25%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26F2N5O2S: 474.1770 found: 474.1760;1H NMR (300 MHz, DMSO-d6) δ 9.03 (t, 1H), 8.68 (d, 1H), 7.90 (d, 1H), 7.80 (d, 1H), 7.71 (dd, 1H), 7.39 (d, 1H), 5.37-5.29 (m, 1H), 4.91 (d, 1H), 4.72 (d, 1H), 4.31 (d, 2H), 3.62-3.35 (m, overlapping with solvent), 2.28-2.12 (m, 2H), 1.12 (s, 6H). Example 39: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-(trifluoromethyl)-piperidin-1-yl)quinoline-4-carboxamide DIPEA (0.83 mL, 4.8 mmol) was added to 6-(4-(trifluoromethyl)piperidin-1-yl)quinoline-4-carboxylic acid Intermediate 88 (154 mg, 0.47 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (197 mg, 0.95 mmol) and TBTU (540 mg, 1.42 mmol) in DMF (5 mL) at 10° C. under N2(g). The resulting solution was stirred at 10° C. overnight under N2(g). The reaction mixture was diluted with water (30 mL). The aqueous layer was extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (DCM:MeOH, 10:1), and further purified by preparative HPLC, PrepMethod D, (gradient 10-40%) to give the title compound (27 mg, 12%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H23F3N5O2S: 478.1518 found: 478.1514;1H NMR (300 MHz, DMSO-d6) δ 9.05 (t, 1H), 8.72 (d, 1H), 7.92 (d, 1H), 7.81-7.63 (m, 2H), 7.46 (d, 1H), 5.40-5.20 (m, 1H), 4.90 (d, 1H), 4.71 (m, 1H), 4.40-4.20 (m, 2H), 4.09 (d, 2H), 3.70-3.34 (m, overlapping with solvent), 2.91 (t, 2H), 1.94 (d, 2H), 1.70-1.45 (m, 2H). Example 40: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3-fluoropiperidin-1-yl)quinoline-4-carboxamide TEA (0.51 mL, 3.7 mmol) was added to a mixture of 6-(3-fluoropiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 90 (100 mg, 0.36 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (125 mg, 0.73 mmol), HOBt (279 mg, 1.82 mmol), and EDC (349 mg, 1.82 mmol) in DMF (15 mL) at rt. The mixture was stirred for 15 h at rt under N2(g). The reaction mixture was diluted with water (25 mL), and extracted with EtOAc (3×25 mL). The combined organic layers were dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (DCM:MeOH, 12:1), and further purified by preparative HPLC, PrepMethod C, (gradient 14-25%) to give the title compound (28 mg, 18%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H23FN5O2S: 428.1550 found: 428.1514;1H NMR (300 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.66 (d, 1H), 7.87 (d, 1H), 7.75-7.55 (m, 2H), 7.39 (d, 1H), 4.87 (d, 1H), 4.72 (d, 1H), 5.40-5.20 (m, 1H), 5.00-4.50 (m, 3H), 4.30 (d, 1H), 3.66-3.20 (m, overlapping with solvent), 2.06-1.47 (4H, m). Example 41: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3-methoxypiperidin-1-yl)quinoline-4-carboxamide DIPEA (0.73 mL, 4.2 mmol) was added to a mixture of 6-(3-methoxypiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 92 (120 mg, 0.42 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (144 mg, 0.84 mmol) and HATU (478 mg, 1.26 mmol) in DMF (15 mL) at rt. The mixture was stirred for 15 h at rt under N2(g). The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (3×25 mL). The combined organic phases were washed with brine (20 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford yellow oil, which was purified by preparative TLC (DCM:MeOH, 10:1), and then further purified by preparative HPLC, PrepMethod P, (gradient 57-67%) to give the title compound (11 mg, 6%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1722;1H NMR (300 MHz, DMSO-d6) δ 9.10-8.90 (m, 1H), 8.64 (d, 1H), 7.86 (d, 1H), 7.77-7.56 (m, 2H), 7.38 (d, 1H), 5.40-5.20 (m, 1H), 4.87 (d, 1H), 4.70 (d, 1H), 4.28 (d, 2H), 3.85-3.70 (m, 1H), 3.66-3.33 (m, overlapping with solvent), 3.10-2.80 (m, 2H), 2.10-1.72 (2H, d), 1.65-1.30 (m, 2H). Example 42: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-methoxy-4-methyl-piperidin-1-yl)quinoline-4-carboxamide DIPEA (0.22 mL, 1.3 mmol) was added to a stirred suspension of 6-(4-methoxy-4-methylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 94 (75 mg, 0.25 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (104 mg, 0.50 mmol), HOBt (101 mg, 0.75 mmol) and EDC (144 mg, 0.75 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 25° C. The resulting solution was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The residue was redissolved in a mixture of sat NaHCO3(aq, 25 mL) and EtOAc (100 mL). The phases were separated, the aqueous layer was extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (3×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 16-26%) to give the title compound (70 mg, 61%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H28N5O3S: 454.1908 found: 454.1918;1H NMR (300 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.64 (d, 1H), 7.85 (d, 1H), 7.75-7.55 (m, 2H), 7.38 (d, 1H), 5.40-5.20 (m, 1H), 4.88 (d, 1H), 4.69 (d, 1H), 4.32-4.22 (m, 2H), 3.55-3.33 (m, overlapping with solvent), 3.25-3.13 (m, overlapping with solvent), 1.90-1.68 (m, 2H), 1.66-1.49 (m, 2H), 1.12 (s, 3H). Example 43: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-isopropoxypiperidin-1-yl)quinoline-4-carboxamide DIPEA (0.17 mL, 0.95 mmol) was added to a mixture of 6-(4-isopropoxypiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 96 (100 mg, 0.32 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (99 mg, 0.48 mmol), EDC (91 mg, 0.48 mmol) and HOBt (73 mg, 0.48 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 20-30%) to give the title compound (105 mg, 71%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C24H30N5O3S: 468.2064 found: 468.2062;1H NMR (300 MHz, CD3OD) δ 8.62 (d, 1H), 7.90 (d, 1H), 7.75 (s, 1H), 7.67 (dd, 1H), 7.49 (d, 1H), 5.43-5.23 (m, 1H), 4.90-4.65 (m, overlapping with solvent), 4.38 (s, 2H), 3.93-3.75 (m, 3H), 3.74-3.55 (m, 1H), 3.52-3.35 (m, 2H), 3.20-3.05 (m, 2H), 2.13-1.93 (m, 2H), 1.79-1.55 (m, 2H), 1.16 (d, 6H). Example 44: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-4,4-difluoro-2-methylpiperidin-1-yl)quinoline-4-carboxamide DIPEA (0.43 mL, 2.5 mmol) was added to mixture of 6-(4,4-difluoro-2-methylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 98 (150 mg, 0.49 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (153 mg, 0.73 mmol), EDC (141 mg, 0.73 mmol) and HOBt (112 mg, 0.73 mmol) in EtOAc (3 mL) and MeCN (3 mL). The reaction was stirred at 40° C. for 4 h. The solvent was removed under reduced pressure. The residue was redissolved in EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 22-33%) to give the title compound (120 mg, 53%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24F2N5O2S: 460.1614 found: 460.1608;1H NMR (400 MHz, CD3OD) δ 8.68 (d, 1H), 7.96 (d, 1H), 7.86 (dd, 1H), 7.71 (dd, 1H), 7.52 (dd, 1H), 5.38-5.32 (m, 1H), 4.83-4.73 (m, overlapping with solvent), 4.59-4.44 (m, 1H), 4.42-4.31 (m, 2H), 3.93-3.69 (m, 1H), 3.50-3.33 (m, overlapping with solvent), 2.38-1.96 (m, 4H), 1.23 (td, 3H). Example 45: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)-piperidin-1-yl)quinoline-4-carboxamide DIPEA (0.27 mL, 1.6 mmol) was added to a solution of (S)-6-(2-(fluoromethyl)piperidin-1-yl)quinoline-4-carboxylic acid Intermediate 100 (90 mg, 0.31 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (97 mg, 0.47 mmol), EDC (90 mg, 0.47 mmol) and HOBt (72 mg, 0.47 mmol) in EtOAc (4 mL) and MeCN (4 mL). The reaction was stirred at 40° C. for 3 h. The solvent was removed under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 17-27%) to give the title compound (45 mg, 33%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H25FN5O2S: 442.1708 found: 442.1708;1H NMR (300 MHz, CD3OD) δ 8.59 (d, 1H), 7.89 (d, 1H), 7.75-7.55 (m, 2H), 7.47 (d, 1H), 5.40-5.27 (m, 1H), 4.87-4.65 (m, overlapping with solvent), 4.58-4.43 (m, 2H), 4.37 (s, 2H), 3.86-3.66 (m, 1H), 3.54-3.34 (m, overlapping with solvent), 3.25-3.13 (m, 1H), 2.03-1.60 (m, 6H). Example 46: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-azaspiro[2.5]octan-5-yl)quinoline-4-carboxamide DIPEA (0.54 mL, 3.1 mmol) was added to a stirred suspension of 6-(5-azaspiro[2.5]octan-5-yl)quinoline-4-carboxylic acid Intermediate 102 (107 mg, 0.31 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (129 mg, 0.62 mmol) and TBTU (354 mg, 0.93 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 9° C. The resulting solution was stirred at 9° C. overnight. The solvent was removed under reduced pressure. The residue was dissolved in a mixture of sat NaHCO3(aq, 60 mL) and EtOAc (80 mL). The aqueous layer was extracted with EtOAc (4×75 mL). The organic layers were combined and washed with water (4×50 mL). The aqueous layers were combined and extracted with EtOAc (3×25 mL). The organic layers were combined and dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 16-26%) to afford the title compound (45 mg, 32%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O2S: 436.1802, found: 436.1784;1H NMR (300 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.64 (d, 1H), 7.85 (d, 1H), 7.70-7.50 (m, 2H), 7.38 (d, 1H), 5.31 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.37-4.20 (m, 2H), 3.50-3.20 (m, overlapping with solvent), 3.16 (s, 2H), 1.89-1.69 (m, 2H), 1.53-1.35 (m, 2H), 0.63-0.39 (m, 2H), 0.38-0.20 (m, 2H). Example 47: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-difluoropyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (0.63 mL, 3.6 mmol) was added to a mixture of 6-(3,3-difluoropyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 104 (200 mg, 0.72 mmol), (R)-3-glycyl-thiazolidine-4-carbonitrile hydrochloride Intermediate 4 (299 mg, 1.44 mmol), EDC (276 mg, 1.44 mmol), HOBt (194 mg, 1.44 mmol) in EtOAc (6 mL) and MeCN (6 mL). The reaction at 25° C. for 10 h. The solvent was removed under reduced pressure. The reaction mixture was diluted with EtOAc (25 mL) and washed with water (3×10 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 17-29%) to give the title compound (120 mg, 38%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O2S: 432.1300, found: 432.1310;1H NMR (300 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.63 (d, 1H), 7.93 (d, 1H), 7.43-7.30 (m, 3H), 5.40-5.20 (m, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.29 (d2H), 3.85 (t, 2H), 3.63 (t, 2H), 3.44-3.32 (m, overlapping with solvent), 2.70-2.50 (m, overlapping with solvent). Example 48: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethylpyrrolidin-1-yl)quinoline-4-carboxamide TEA (2.07 mL, 14.8 mmol) was added to a stirred suspension of 6-(3,3-dimethylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 106 (347 mg, 0.74 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (385 mg, 1.86 mmol), HOBt (568 mg, 3.71 mmol) and EDC (711 mg, 3.71 mmol) in MeCN (7 mL) and EtOAc (7 mL) at 7° C. The resulting suspension was stirred at 7° C. overnight. The solvent was removed under reduced pressure. The residue was suspended in EtOAc and washed with a solution of sat NaHCO3(50 mL). The aqueous layer was extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×20 mL) and concentrated under reduced pressure. The crude product was purified by preparative HPLC, PrepMethod P, to give the title compound (174 mg, 55%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O2S: 424.1802 found: 424.1806;1H NMR (300 MHz, DMSO-d6) δ 8.91 (t, 1H), 8.53 (d, 1H), 7.84 (d, 1H), 7.34 (d, 1H), 7.29-7.15 (m, 2H), 5.30 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.27 (d, 2H), 3.51-3.33 (m, overlapping with solvent), 3.15 (s, 2H), 1.80 (t, 2H), 1.14 (s, 3H), 1.13 (s, H). Example 49: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxamide DIPEA (2.12 mL, 12.2 mmol) was added to a mixture of 6-(5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxylic acid Intermediate 108 (346 mg, 0.61 mmol), (R)-3-glycyl-thiazolidine-4-carbonitrile hydrochloride Intermediate 4 (253 mg, 1.22 mmol), HOBt (465 mg, 3.04 mmol) and EDC (583 mg, 3.04 mmol) in EtOAc (8 mL) and MeCN (8 mL) at 5° C. The resulting solution was stirred at 5° C. overnight. The solvent was removed under reduced pressure. The residue was dissolved in a mixture of a sat NaHCO3(aq, 80 mL) and EtOAc (100 mL). The phases were separated, and the aqueous layer was extracted with EtOAc (4×100 mL). The organic layers were combined, and washed with water (4×25 mL). The aqueous layers were combined and extracted with EtOAc (3×20 mL). The organic layers were combined, dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, to give the title compound (120 mg, 47%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O2S: 422.1646, found: 422.1654;1H NMR (300 MHz, DMSO-d6) δ 8.91 (t, 1H), 8.54 (d, 1H), 7.85 (d, 1H), 7.34 (d, 1H), 7.30-7.14 (m, 2H), 5.29 (dd, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.27 (d, 2H), 3.54 (t, 2H), 3.40-3.25 (m, overlapping with solvent), 1.94 (t, 2H), 0.75-0.57 (m, 4H). Example 50: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4S)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxamide TEA (1.86 mL, 13.4 mmol) was added to a stirred suspension of 6-((3S,4R)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 110 (335 mg, 0.67 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (347 mg, 1.67 mmol), HOBt (511 mg, 3.34 mmol) and EDC (640 mg, 3.34 mmol) in EtOAc (9 mL) and MeCN (9 mL) at 5° C. The resulting suspension was stirred at 5° C. overnight. The solvent was removed under reduced pressure. The residue was suspended in sat NaHCO3(50 mL), and extracted with EtOAc (4×50 mL). The organic layers were combined and washed with water (4×25 mL). The aqueous layers were combined and extracted with EtOAc (3×25 mL). The organic layers were combined and dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, to give the title compound (128 mg, 44%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H20N5O2S: 432.1300, found: 432.1294;1H NMR (300 MHz, DMSO-d6) δ 9.06-8.89 (m, 1H), 8.62 (d, 1H), 7.93 (d, 1H), 7.40 (d, 1H), 7.38-7.30 (m, 2H), 5.65-5.25 (m, 3H), 4.89 (d, 1H), 4.72 (d, 1H), 4.34-4.15 (m, 2H), 3.92-3.75 (m, 2H), 3.71-3.53 (m, 2H), 3.41-3.34 (m, overlapping with solvent). Example 51: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-fluoropyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (2.64 mL, 15.1 mmol) was added to a stirred solution of (S)-6-(3-fluoropyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 112 (282 mg, 0.76 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (392 mg, 1.89 mmol) and TBTU (860 mg, 2.27 mmol) in EtOAc (7 mL) and MeCN (7 mL) at 6° C. The resulting solution was stirred at 5° C. overnight. The solvent was removed under reduced pressure. The residue was dissolved in a mixture of sat NaHCO3(aq, 70 mL) and EtOAc (100 mL). The phases were separated, and the aqueous layer was extracted with EtOAc (4×100 mL). The organic layers were combined and washed with water (4×25 mL). The aqueous layers were combined and extracted with EtOAc (4×20 mL). The organic layers were combined, dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, to give the title compound (120 mg, 38%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C20H21FN5O2S: 414.1394, found: 414.1384;1H NMR (300 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.60 (d, 1H), 7.90 (d, 1H), 7.45-7.30 (m, 3H), 5.49 (d, 1H), 5.36-5.25 (m, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.30 (d, 2H), 3.80-3.34 (m, overlapping with solvent), 2.43-2.01 (m, 2H). Example 52: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-fluoropyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (1.39 mL, 7.96 mmol) was added to a stirred solution of (R)-6-(3-fluoropyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 114 (297 mg, 0.80 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (248 mg, 1.19 mmol) and TBTU (905 mg, 2.39 mmol) in MeCN (7 mL) and EtOAc (7 mL) at 6° C. The resulting solution was stirred at 6° C. overnight. The solvent was removed under reduced pressure. The residue was dissolved with a mixture of sat NaHCO3(70 mL) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (4×100 mL). The organic layers were combined and washed with water (4×50 mL). The aqueous layers were combined and extracted with EtOAc (4×20 mL). The organic layers were combined and dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, to give the title compound (113 mg, 34%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C20H21FN5O2S: 414.1394, found: 414.1406;1H NMR (300 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.59 (d, 1H), 7.90 (d, 1H), 7.48-7.22 (m, 3H), 5.49 (d, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.35-4.24 (m, 2H), 3.74-3.68 (m, 1H), 3.64-3.37 (m, overlapping with solvent), 2.38-2.17 (m, 2H). Example 53: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(hexahydrocyclopenta[c]pyrrol-2(1H)-yl)quinoline-4-carboxamide DIPEA (0.26 mL, 1.5 mmol) was added to a mixture of 6-(hexahydrocyclopenta[c]pyrrol-2(1H)-yl)quinoline-4-carboxylic acid Intermediate 116 (85 mg, 0.30 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (94 mg, 0.45 mmol), EDC (87 mg, 0.45 mmol) and HOBt (69 mg, 0.45 mmol) in EtOAc (5 mL) and MeCN (5 mL). The reaction was stirred at 40° C. for 3 h. The solvent was removed under reduced pressure. The residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 23-33%) to give the title compound (40 mg, 30%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O2S: 436.1802, found: 436.1808;1H NMR (300 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.58 (d, 1H), 7.86 (d, 1H), 7.40-7.26 (m, 3H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.29 (d, 2H), 3.62-3.50 (m, 2H), 3.45-3.33 (m, overlapping with solvent), 3.19-3.10 (m, 2H), 2.83-2.77 (m, 3H), 1.92-1.43 (m, 5H). Example 54: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (0.20 mL, 1.2 mmol) was added to a mixture of (S)-6-(3-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 118 (100 mg, 0.39 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (100 mg, 0.59 mmol) and HATU (297 mg, 0.78 mmol) in MeCN (10 mL) and EtOAc (10 mL). The reaction was stirred at 25° C. for 2 h. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH, 8:1), and then further purified by preparative HPLC, PrepMethod F, (gradient 20-30%) to give the title compound (56 mg, 35%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O2S: 410.1646, found: 410.1634;1H NMR (400 MHz, DMSO-d6) δ 9.12 (t, 1H), 8.71 (d, 1H), 7.97 (d, 1H), 7.60 (d, 1H), 7.47 (dd, 1H), 7.30 (d, 1H), 5.31 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.49-4.17 (m, 2H), 3.66-3.30 (m, overlapping with solvent), 3.06-2.92 (m, 1H), 2.47-2.30 (m, 1H), 2.22-2.10 (m, 1H), 1.75-1.57 (m, 1H), 1.13 (d, 3H). Example 55: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (0.36 mL, 2.1 mmol) was added to a mixture of (R)-6-(3-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 120 (175 mg, 0.68 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (175 mg, 1.02 mmol), and HATU (519 mg, 1.37 mmol) in MeCN (10 mL) and EtOAc (10 mL). The reaction was stirred at 25° C. for 2 h. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH, 8:1), and then further purified by preparative HPLC, PrepMethod F, (gradient 17-36%) to give the title compound (80 mg, 29%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O2S: 410.1646, found: 410.1650;1H NMR (400 MHz, DMSO-d6) δ 9.11 (t, 1H), 8.71 (d, 1H), 7.97 (d, 1H), 7.60 (d, 1H), 7.47 (dd, 1H), 7.30 (d, 1H), 5.32 (dd, 1H), 4.90 (d, 1H), 4.73 (d, 1H), 4.45-4.20 (m, 2H), 3.66-3.57 (m, 1H), 3.56-3.33 (m, overlapping with solvent), 3.04-2.93 (m, 1H), 2.47-2.30 (m, 1H), 2.25-2.05 (m, 1H), 1.76-1.56 (m, 1H), 1.13 (d, 3H). Example 56: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(trifluoromethyl)-pyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (0.37 mL, 2.1 mmol) was added to a mixture of (S)-6-(2-(trifluoromethyl)pyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 122 (130 mg, 0.42 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (131 mg, 0.63 mmol), EDC (161 mg, 0.84 mmol) and HOBt (128 mg, 0.84 mmol) in EtOAc (5 mL) and MeCN (5 mL). The reaction was stirred at 50° C. for 4 h. The solvent was removed under reduced pressure. The residue was redissolved in EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 25-35%) to give the title compound (110 mg, 57%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H21F3N5O2S: 464.1362, found: 464.1366;1H NMR (400 MHz, CD3OD) δ 8.61 (d, 1H), 7.94 (d, 1H), 7.75-7.61 (m, 1H), 7.60-7.40 (m, 2H), 5.32 (dd, 1H), 4.84-4.66 (m, overlapping with solvent), 4.47 (d, 1H), 4.31 (d, H), 3.91-3.74 (m, 1H), 3.50-3.34 (m, overlapping with solvent), 2.38-2.06 (m, 4H). Example 57: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethylpyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (0.40 mL, 2.3 mmol) was added to a mixture of 6-(2,2-dimethylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 124 (125 mg, 0.46 mmol), (R)-3-glycyl-thiazolidine-4-carbonitrile hydrochloride Intermediate 4 (144 mg, 0.69 mmol), EDC (133 mg, 0.69 mmol) and HOBt (106 mg, 0.69 mmol) in EtOAc (5 mL) and MeCN (5 mL). The reaction was stirred at 40° C. for 4 h. The solvent was removed under reduced pressure. The residue was dissolved in EtOAc and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 15-25%) to give the title compound (110 mg, 56%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O2S: 424.1802, found: 424.1810;1H NMR (300 MHz, CD3OD) δ 8.49 (d, 1H), 7.83 (d, 1H), 7.54 (dd, 1H), 7.46 (d, 1H), 7.36 (d, 1H), 5.40-5.20 (m, 1H), 4.84-4.65 (m, overlapping with solvent), 4.37 (s, 2H), 3.58-3.48 (m, 2H), 3.47-3.33 (m overlapping with solvent), 2.10-1.90 (m, 4H), 1.53 (s, 6H). Example 58: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-6-(fluoromethyl)-5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxamide DIPEA (0.47 mL, 2.7 mmol) was added to a mixture of (R)-6-(6-(fluoromethyl)-5-azaspiro[2.4]heptan-5-yl)quinoline-4-carboxylic acid Intermediate 126 (160 mg, 0.53 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (166 mg, 0.80 mmol), EDC (204 mg, 1.07 mmol) and HOBt (163 mg, 1.07 mmol) in EtOAc (6 mL) and MeCN (6 mL). The reaction was stirred at 50° C. for 5 h. The solvent was removed under reduced pressure. The residue was redissolved in EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient 18-28%) to give the title compound (145 mg, 60%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H25FN5O2S: 454.1708, found: 454.1712;1H NMR (300 MHz, DMSO-d6) δ 8.94 (t, 1H), 8.57 (d, 1H), 7.87 (d, 1H), 7.44-7.31 (m, 3H), 5.27 (dd, 1H), 4.87 (d, 1H), 4.73-4.12 (m, 6H), 3.51-3.15 (m, overlapping with solvent), 2.35 (dd, 1H), 1.64 (d, 1H), 0.75-0.52 (m, 4H). Example 59: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-fluoroazepan-1-yl)quinoline-4-carboxamide TEA (0.50 mL, 3.6 mmol) was added to (R)-6-(3-fluoroazepan-1-yl)quinoline-4-carboxylic acid Intermediate 130 (52 mg, 0.18 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (150 mg, 0.72 mmol), HOBt (276 mg, 1.80 mmol) and EDC (346 mg, 1.80 mmol) in DMF (5 mL) at 15° C. The resulting solution was stirred at 40° C. overnight under N2(g). The reaction mixture was diluted with sat NaHCO3(30 mL), and extracted with EtOAc (9×50 mL). The organic layers were combined and washed with brine (5×200 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford crude product. The crude product was purified by preparative HPLC, PrepMethod F, to give the title compound (39 mg, 49%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H25FN5O2S: 442.1708, found: 442.1702;1H NMR (300 MHz, DMSO-d6) δ 9.05-8.80 (m, 1H), 8.57 (d, 1H), 7.85 (d, 1H), 7.59-7.41 (m, 2H), 7.35 (d, 1H), 5.40-5.20 (m, 1H), 5.18-4.92 (m, 1H), 4.88 (d, 1H), 4.72 (d, 1H), 4.28 (d, 2H), 3.96-3.81 (m, 1H), 3.63-3.34 (m, overlapping with solvent), 2.00-1.58 (m, 5H), 1.52-1.28 (m, 1H). Example 60: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-fluoroazepan-1-yl)quinoline-4-carboxamide TEA (0.68 mL, 4.9 mmol) was added to (S)-6-(3-fluoroazepan-1-yl)quinoline-4-carboxylic acid Intermediate 132 (70 mg, 0.24 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (202 mg, 0.97 mmol), HOBt (372 mg, 2.43 mmol) and EDC (465 mg, 2.43 mmol) in DMF (10 mL) at 13° C. The resulting suspension was stirred at 30° C. for 5 h under N2(g). The reaction mixture was diluted with sat NaHCO3(50 mL), and extracted with EtOAc (9×50 mL). The organic layers were combined and washed with brine (5×200 mL). The organic layer was dried over Na2SO4, filtered and evaporated to afford crude product. The crude product was purified by preparative HPLC, PrepMethod P, to give the title compound (58 mg, 54%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H25FN5O2S: 442.1708, found: 442.1698; 1H NMR (300 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.56 (d, 1H), 7.83 (d, 1H), 7.58-7.41 (m, 2H), 7.34 (d, 1H), 5.33-4.80 (m, 3H), 4.76-4.50 (m, 1H), 4.36-4.15 (m, 2H), 4.07-3.73 (m, 2H), 3.70-3.30 (m, overlapping with solvent), 1.89-1.32 (m, 6H). Example 61: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxamide DIPEA (0.54 mL, 3.1 mmol) was added to a stirred suspension of (R)-6-(7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylic acid Intermediate 134 (205 mg, 0.31 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (96 mg, 0.46 mmol) and TBTU (350 mg, 0.92 mmol) in EtOAc (5 mL) and MeCN (5 mL) at 4° C. The resulting solution was stirred at 4° C. overnight. The solvent was removed under reduced pressure. The residue was dissolved in a mixture of sat NaHCO3(70 mL) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (3×100 mL). The organic layers were combined and washed with water (4×25 mL). The aqueous layers were combined and extracted with EtOAc (4×50 mL). All organic layers were combined, dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod F, to give the title compound (90 mg, 66%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1746;1H NMR (300 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.55 (d, 1H), 7.84 (d1H), 7.55-7.40 (m, 2H), 7.32 (d, 1H), 5.39-5.19 (m, 1H), 4.89 (d, 1H), 4.70 (d, 1H), 4.37-4.16 (m, 2H), 4.01-3.81 (m, 2H), 3.79-3.34 (m, overlapping with solvent), 2.16-1.95 (m, 1H), 1.74-1.51 (m, 1H), 1.05 (d, 3H). Example 62: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxamide DIPEA (1.14 mL, 6.51 mmol) was added to a stirred suspension of (S)-6-(7-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylic acid Intermediate 136 (305 mg, 0.33 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (135 mg, 0.65 mmol), HOBt (249 mg, 1.63 mmol) and EDC (312 mg, 1.63 mmol) in EtOAc (5 mL) and MeCN (5 mL) at 6° C. The resulting solution was stirred at 4° C. overnight and then at 40° C. for a further 4 h. The solvent was removed under reduced pressure. The residue was dissolved in a mixture of sat NaHCO3(50 mL) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (3×100 mL). The organic layers were combined and washed with water (4×25 mL). All organic layers were combined, dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod F, to give the title compound (100 mg, 70%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1746;1H NMR (300 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.56 (d, 1H), 7.85 (d, 1H), 7.59-7.43 (m, 2H), 7.33 (d, 1H), 5.40-5.20 (m, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.33-4.19 (m, 2H), 4.05-3.80 (m, 2H), 3.78-3.23 (m, overlapping with solvent), 2.17-1.98 (m, 1H), 1.75-1.50 (m, 1H), 1.07 (d, 3H). Example 63: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxamide DIPEA (0.79 mL, 4.5 mmol) was added dropwise to a mixture of (R)-6-(3-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylic acid Intermediate 138 (65 mg, 0.23 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (94 mg, 0.45 mmol), HOBt (348 mg, 2.27 mmol) and EDC (435 mg, 2.27 mmol) in MeCN (3 mL) and EtOAc (3 mL) at 10° C. The resulting solution was stirred at 10° C. for overnight under N2(g). The solvent was removed under reduced pressure. The residue was diluted with sat NaHCO3(100 mL), and extracted with EtOAc (6×100 mL). The organic layers were combined and washed with brine (5×200 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, to afford the title compound (45 mg, 45%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1742;1H NMR (300 MHz, DMSO-d6) δ 8.94 (brs, 0.5H), 8.52 (d, 1H), 8.32 (brs, 0.5H), 7.84 (d, 1H), 7.54-7.35 (m, 2H), 7.31 (d, 1H), 5.31 (dd, 1H), 4.87 (d, 1H), 4.68 (d, 1H), 4.37-4.10 (m, 3H), 4.00 (dd, 1H), 3.97-3.72 (m, 2H), 3.61-3.30 (m, overlapping with solvent), 1.93-1.64 (m, 2H), 1.07 (d, 3H). Example 64: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxamide DIPEA (0.37 mL, 2.1 mmol) was added to a mixture of (R)-6-(2-methyl-1,4-oxazepan-4-yl)quinoline-4-carboxylic acid Intermediate 140 (100 mg, 0.35 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (90 mg, 0.52 mmol) and HATU (266 mg, 0.70 mmol) in MeCN (5 mL) and EtOAc (5 mL). The mixture was stirred under an atmosphere of air at 25° C. for 3 h. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH, 19:1), followed by preparative HPLC, PrepMethod F, (gradient 15-25%) to give the title compound (30 mg, 20%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1724;1H NMR (400 MHz, DMSO-d6) δ 9.18 (t, 1H), 8.74 (d, 1H), 7.98 (d, 1H), 7.74 (dd, 1H), 7.67-7.50 (m, 2H), 5.38-5.18 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.39-4.25 (m, 2H), 4.23-4.01 (m, 2H), 3.93-3.75 (m, 2H), 3.60-3.31 (m, overlapping with solvent), 3.29-3.06 (m, 2H), 2.19-1.98 (m, 1H), 1.95-1.80 (m, 1H), 1.20 (d, 3H). Example 65: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methylpyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (0.41 mL, 2.3 mmol) was added to a mixture of (S)-6-(2-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 128 (100 mg, 0.39 mmol), (R)-3-glycyl-thiazolidine-4-carbonitrile hydrochloride Intermediate 4 (122 mg, 0.59 mmol) and HATU (297 mg, 0.78 mmol) in MeCN (5 mL) and EtOAc (5 mL). The mixture was stirred at 25° C. for 3 h. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH, 19:1), and further purified by preparative HPLC, PrepMethod F, (gradient 20-30%) to give the title compound (35 mg, 22%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O2S: 410.1646, found: 410.1636;1H NMR (400 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.55 (d, 1H), 7.86 (d, 1H), 7.39-7.18 (m, 3H), 5.40-5.25 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.37-4.19 (m, 2H), 4.15-4.00 (m, 1H), 3.57-3.18 (m, overlapping with solvent), 2.16-1.94 (m, 3H), 1.80-1.60 (m, 1H), 1.17 (d, 3H). Example 66: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxyazetidin-1-yl)quinoline-4-carboxamide HATU (159 mg, 0.42 mmol) was added to a stirred mixture of crude 6-(3-methoxyazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 142 (102 mg, 0.35 mmol) and DIPEA (0.303 mL, 1.74 mmol) in a mixture of MeCN (1.5 mL) and EtOAc (1.5 mL) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (87 mg, 0.42 mmol) was added and the reaction mixture was stirred for 1.5 h at rt. The reaction was diluted with EtOAc (8 mL) and washed with 8% NaHCO3(aq, 6 mL). The aqueous layer was extracted with EtOAc and the combined organic layers was dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-45%) to give the title compound (0.071 g, 49%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C20H22N5O3S: 412.1438 found: 412.1442;1H NMR (500 MHz, DMSO-d6) δ 9.14 (t, 1H). 8.79 (d, 1H), 7.98 (d, 1H), 7.65 (d, 1H), 7.33-7.27 (m, 2H), 5.34 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.42-4.29 (m, 3H), 4.27-4.18 (m, 2H), 3.83 (dd, 2H), 3.41 (dd, 1H), 3.36 (dd, 1H), 3.27 (s, 3H). Example 67: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholinoquinoline-4-carboxamide DIPEA (0.15 mL, 0.87 mmol) was added to a suspension of 6-morpholinoquinoline-4-carboxylic acid Intermediate 144 (75 mg, 0.29 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (121 mg, 0.58 mmol), HOBt (53 mg, 0.35 mmol) and EDC (84 mg, 0.44 mmol) in EtOAc (1 mL) and MeCN (1 mL). Gives a clear yellow solution which was stirred at rt overnight. The mixture was diluted with EtOAc, washed with sat NaHCO3and brine. The organic phase was dried, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod E, (gradient 5-65%) to give the title compound (26 mg, 21%); HRMS (ESI) m/z [M+H]+calcd for C20H22N5O3S: 412.1438 found: 412.1437;1H NMR (400 MHz, CD3CN) δ 8.71 (d, 1H), 8.05 (s, 1H), 7.96 (d, 1H), 7.69 (d, 1H), 7.60 (dd, 1H), 7.43 (d, 1H), 5.24 (dd, 1H), 4.79-4.60 (m, 2H), 4.30 (d, 2H), 3.88-3.79 (m, 4H), 3.35-3.28 (m, 6H). Example 68: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6S)-2,6-dimethyl-morpholino)quinoline-4-carboxamide A vial was charged with tert-butyl 6-((2R,6S)-2,6-dimethylmorpholino)quinoline-4-carboxylate Intermediate 149 (121 mg, 0.35 mmol) and 90% TFA (aq, 0.5 mL). The vial was heated at 50° C. for 1 h 40 min. The reaction mixture was concentrated and co-evaporated from a mixture of H2O and MeCN. MeCN (1.5 mL), EtOAc (1.5 mL) and DIPEA (0.305 mL, 1.75 mmol) were added to the residue followed by HATU (0.16 g, 0.42 mmol) and the mixture was stirred at for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.087 g, 0.42 mmol) was added. The mixture was stirred at rt for 3 h. The reaction mixture was partitioned between EtOAc (25 mL) and 8% NaHCO3(aq, 10 mL). The aqueous layer was extracted with EtOAc (10 mL) and the combined organic layers were concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient of 5-45%) to give the title compound (85 mg, 55%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750 found: 440.1756;1H NMR (500 MHz, DMSO-d6) δ 9.08 (t, 1H), 8.72 (d, 1H), 7.93 (d, 1H), 7.77-7.71 (m, 2H), 7.45 (d, 1H), 5.31 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.36-4.27 (m, 2H), 3.89 (dd, 2H), 3.79-3.69 (m, 2H), 3.42 (dd, 1H), 3.36 (dd, 1H), 2.42 (t, 2H), 1.25-1.17 (m, 6H). Example 69: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(fluoromethyl)-morpholino)quinoline-4-carboxamide HATU (0.730 g, 1.92 mmol) was added to a stirred mixture of the crude (R)-6-(2-(fluoromethyl)morpholino)quinoline-4-carboxylic acid Intermediate 146 (1.60 mmol) and DIPEA (1.68 mL, 9.60 mmol) in a mixture of MeCN (7 mL) and EtOAc (7 mL) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.399 g, 1.92 mmol) was added and the reaction mixture was stirred for 1 h at rt. The reaction mixture was diluted with EtOAc (100 mL) and washed sequentially with 8% NaHCO3(aq, 2×20) mL) and H2O (2×10 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH 20:1). The product was further purified twice by preparative HPLC, PrepMethod G, (gradients: 0-30% and 5-35%). The product was further purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH, 20:1) to give the title compound (0.314 g, 44%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H23FN5O3S: 444.1500 found: 444.1506;1H NMR (500 MHz, DMSO-d6) δ 9.05 (t, 1H), 8.71 (d, 1H), 7.93 (d, 1H), 7.76 (d, 1H), 7.70 (dd, 1H), 7.42 (d, 1H), 5.32 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.68-4.48 (m, 2H), 4.37-4.25 (m, 2H), 4.08-4.01 (m, 1H), 3.95-3.82 (m, 2H), 3.82-3.68 (m, 2H), 3.43 (dd, 1H), 3.36 (dd, 1H), 2.82 (td, 1H), 2.68 (t, 1H). Example 70: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,6R)-2,6-dimethyl-morpholino)quinoline-4-carboxamide HATU (0.119 g, 0.31 mmol) was added to a stirred mixture of crude 6-((2R,6R)-2,6-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 148 (0.26 mmol) and DIPEA (0.227 mL, 1.30 mmol) in a mixture of MeCN (1.2 mL) and EtOAc (1.2 mL) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.065 g, 0.31 mmol) was added. The reaction was stirred for 4.5 h at rt. The reaction was diluted with EtOAc (5 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc (3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 15-55%). The compound was dissolved in EtOAc and the organic layer was washed twice with H2O, concentrated and freeze-dried to give the title compound (0.042 g, 37%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750 found: 440.1746;1H NMR (500 MHz, DMSO-d6) δ 9.10 (t, 1H), 8.73 (d, 1H), 7.93 (d, 1H), 7.7-7.8 (m, 2H), 7.49 (d, 1H), 5.31 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.25-4.39 (m, 2H), 4.07-4.16 (m, 2H), 3.43 (td, 3H), 3.36 (dd, 1H), 3.12 (dd, 2H), 1.24 (d, 6H). Example 71: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(fluoromethyl)-morpholino)quinoline-4-carboxamide A vial was charged with tert-butyl (S)-6-(2-(fluoromethyl)morpholino)quinoline-4-carboxylate Intermediate 150 (121 mg, 0.35 mmol) and 90% TFA (aq, 0.5 mL) and the reaction mixture was heated at 50° C. for 1 h 40 min. The reaction mixture was concentrated and the residue was co-evaporated from a mixture of H2O and MeCN. MeCN (1.5 mL), EtOAc (1.5 mL) and DIPEA (0.305 mL, 1.75 mmol) were added to the residue followed by HATU (0.160 g, 0.42 mmol). The mixture was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.087 g, 0.42 mmol) was added. The mixture was stirred at rt for 3 h. The reaction mixture was partitioned between EtOAc (25 mL) and 8% NaHCO3(aq, 10 mL). The aqueous layer was extracted with EtOAc (10 mL) and the combined organic layers were concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient 5-45%) to give the title compound (65 mg, 42%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H23FN5O3S: 444.1500 found: 444.1496;1H NMR (500 MHz, DMSO-d6) δ 9.06 (t, 1H), 8.72 (d, 1H), 7.94 (d, 1H), 7.78 (d, 1H), 7.71 (dd, 1H), 7.44 (d, 1H), 5.34 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.68-4.47 (m, 2H), 4.37-4.25 (m, 2H), 4.08-4.01 (m, 1H), 3.93-3.82 (m, 2H), 3.82-3.68 (m, 2H), 3.42 (dd, 1H), 3.36 (dd, 1H), 2.85 (td, 1H), 2.68 (t, 1H). Example 72: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methyl-morpholino)quinoline-4-carboxamide HATU (0.160 g, 0.42 mmol) was added to a stirred mixture of crude (R)-6-(2-methylmorpholino)quinoline-4-carboxylic acid Intermediate 152 (0.134 g) and DIPEA (0.306 mL, 1.75 mmol) in a mixture of MeCN/EtOAc (3 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.087 g, 0.42 mmol) was added. The reaction was stirred for 45 min at rt. The reaction was diluted with EtOAc (6 mL) and washed with 8% NaHCO3(aq, 6 mL). The aqueous layer was extracted with EtOAc (2×3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient of 5-55%) to give the title compound (56 mg, 38%) as a red/orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594 found: 426.1582;1H NMR (500 MHz, DMSO-d6) δ 9.10 (t, 1H), 8.75 (d, 1H), 7.95 (d, 1H), 7.79-7.73 (m, 2H), 7.50 (d, 1H), 5.32 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.39-4.25 (m, 2H), 4.01-3.94 (m, 1H), 3.88 (d, 1H), 3.78 (d, 1H), 3.74-3.62 (m, 3H), 3.42 (dd, 1H), 3.36 (dd, 1H), 2.83 (td, 1H), 1.22 (d, 3H). Example 73: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide HATU (0.137 g, 0.36 mmol) was added to a stirred mixture of crude (R)-6-(2-(trifluoromethyl)morpholino)quinoline-4-carboxylic acid Intermediate 154 (0.30 mmol) and DIPEA (0.262 mL, 1.50 mmol) in a mixture of MeCN/EtOAc (2.8 mL, 1:1) at rt. The reaction was stirred for ˜1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.075 g, 0.36 mmol) was added. The reaction mixture was stirred for 4.5 h at rt, diluted with EtOAc (5 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc (3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-55%). The purified compound was dissolved in EtOAc and washed sequentially with 8% NaHCO3(aq) and H2O (2×). The organic layer was concentrated and freeze-dried from MeCN/H2O to give the title compound (0.066 g, 46%) as a solid; HRMS (ESI) m/z [M+H]+calcd for C21H21F3N5O3S: 480.1312 found: 480.1308;1H NMR (500 MHz, DMSO-d) δ 9.06 (t, 1H), 8.74 (d, 1H), 7.95 (d, 1H), 7.82 (d, 1H), 7.76 (dd, 1H), 7.44 (d, 1H), 5.26 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.45-4.39 (m, 1H), 4.37-4.25 (m, 2H), 4.14 (d, 1H), 3.99 (d, 1H), 3.89-3.8 (m, 2H), 3.43 (dd, 1H), 3.37 (dd, 1H), 2.96-2.84 (m, 2H). Example 74: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(trifluoromethyl)-morpholino)quinoline-4-carboxamide HATU (0.137 g, 0.36 mmol) was added to a stirred mixture of crude (S)-6-(2-(trifluoromethyl)morpholino)quinoline-4-carboxylic acid Intermediate 156 (0.30 mmol) and DIPEA (0.262 mL, 1.50 mmol) in MeCN (1.4 mL) and EtOAc (1.4 mL) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.075 g, 0.36 mmol) was added and the mixture was stirred for 4.5 h at rt. The reaction mixture was diluted with EtOAc (5 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc (3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-55%). The product was partitioned between EtOAc and 8% NaHCO3(aq). The organic layer was washed with small portions of H2O (3×), concentrated and freeze-dried from MeCN/H2O to give the title compound (0.043 g, 30%); HRMS (ESI) m/z [M+H]+calcd for C21H21F3N5O3S: 480.1312, found: 480.1302;1H NMR (500 MHz, DMSO-d6) δ 9.04 (t, 1H), 8.74 (d, 1H), 7.95 (d, 1H), 7.84 (d, 1H), 7.76 (dd, 1H), 7.44 (d, 1H), 5.29-5.24 (m, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.44-4.38 (m, 1H), 4.31 (dd, 2H), 4.15 (d, 1H), 3.96 (d, 1H), 3.89-3.79 (m, 2H), 3.43 (d, 1H), 3.37 (dd, 1H), 2.99-2.88 (m, 2H). Example 75: 6-((1S,4S)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)-N-(2-((R)-4-cyano-thiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide HATU (0.141 g, 0.37 mmol) was added to a stirred mixture of crude 6-((1S,4S)-2-oxa-5-azabicyclo[2.2.1]heptan-5-yl)quinoline-4-carboxylic acid Intermediate 158 (0.31 mmol) and DIPEA (0.271 mL, 1.55 mmol) in a mixture of MeCN/EtOAc (2.8 mL, 1:1) at rt and the reaction was stirred for 1 min. (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.077 g, 0.37 mmol) was added and the reaction mixture was stirred for 45 min at rt. The reaction mixture was diluted with EtOAc (5 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc (3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient of 5-45%) to give an orange solid which was dissolved in EtOAc. The organic layer was washed with 8% NaHCO3(aq, 2×) and H2O (2×). The organic layer was concentrated and freeze-dried from a mixture of MeCN and H2O to give the title compound (0.035 g, 27%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438 found: 424.1442;1H NMR (500 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.59 (d, 1H), 7.87 (d, 1H), 7.45 (d, 1H), 7.36 (dd, 2H), 5.31 (dd, 1H), 4.89 (d, 1H), 4.77 (brs, 1H), 4.70 (d, 1H), 4.66 (brs, 1H), 4.36-4.23 (m, 2H), 3.84 (d, 1H), 3.74 (d, 1H), 3.62 (d, 1H), 3.42-3.34 (m, 2H), 3.14 (d, 1H), 1.98 (dd, 1H), 1.89 (d, 1H). Example 76: 6-((1R,4R)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)-N-(2-((R)-4-cyano-thiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide HATU (0.128 g, 0.34 mmol) was added to a stirred mixture of crude 6-((1R,4R)-2-oxa-5-azabicyclo[2.2.1]heptan-5-yl)quinoline-4-carboxylic acid Intermediate 160 (0.28 mmol) and DIPEA (0.245 mL, 1.40 mmol) in a mixture of MeCN/EtOAc (2.6 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.070 g, 0.34 mmol) was added. The reaction was stirred for 3 h at rt. The reaction mixture was diluted with EtOAc (5 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc (3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH, 6:1). The compound was further purified by preparative HPLC, PrepMethod G, (gradient of 5-45%). The compound was partitioned between EtOAc and 8% NaHCO3(aq). The organic layer was washed with 8% NaHCO3(aq) followed by H2O (2×). The organic layer was concentrated and the washing sequence was repeated. The crude compound was purified by straight phase flash chromatography (EtOAc:MeOH, 6:1) and freeze-dried from MeCN/H2O to give the title compound (0.026 g, 22%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438 found: 424.1430;1H NMR (500 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.59 (d, 1H), 7.87 (d, 1H), 7.47 (d, 1H), 7.38-7.32 (m, 2H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.79 (brs, 1H), 4.70 (d, 1H), 4.68-4.64 (m, 1H), 4.29 (d, 2H), 3.82 (d, 1H), 3.75 (d, 1H), 3.62 (d, 1H), 3.41 (dd, 1H), 3.38-3.36 (m, 1H), 2.02-1.96 (m, 1H), 3.14 (d, 1H), 1.88 (m, 1H). Example 77: 6-(6-Oxa-3-azabicyclo[3.1.1]heptan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide HATU (0.137 g, 0.36 mmol) was added to a stirred mixture of the crude 6-(6-oxa-3-azabicyclo[3.1.1]heptan-3-yl)quinoline-4-carboxylic acid Intermediate 162 (0.30 mmol) and DIPEA (0.262 mL, 1.50 mmol) in a mixture of MeCN/EtOAc (2.8 mL, 1:1) at rt. The reaction mixture was stirred for 5 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.075 g, 0.36 mmol) was added. The reaction mixture was stirred for 4.5 h at rt, diluted with EtOAc (5 mL) and washed with 5 mL 8% NaHCO3(aq). The aqueous layer was extracted with EtOAc (3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-45%) followed by straight phase flash chromatography on silica (EtOAc:MeOH, 9:1). The appropriate fractions were combined, concentrated, and the residue was partitioned between EtOAc and NaHCO3(aq). The organic layer was washed with H2O (3×), concentrated and freeze-dried from MeCN/H2O to give the title compound (0.028 g, 22%) as a solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438 found: 424.1454;1H NMR (500 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.62 (d, 1H), 7.95 (d, 1H), 7.55-7.48 (m, 2H), 7.40 (d, 1H), 5.31 (dd, 1H), 4.89 (d, 1H), 4.76 (d, 2H), 4.71 (d, 1H), 4.37-4.23 (m, 2H), 3.71 (t, 2H), 3.56 (t, 2H), 3.43-3.34 (m, 2H), 3.16 (q, 1H), 1.94 (d, 1H). Example 78: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,6S)-2,6-dimethyl-morpholino)quinoline-4-carboxamide HATU (0.114 g, 0.30 mmol) was added to a stirred mixture of crude 6-((2S,6S)-2,6-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 164 (0.25 mmol) and DIPEA (0.218 mL, 1.25 mmol) in a mixture of MeCN/EtOAc (2.2 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol) was added and the reaction was stirred for 2 h 45 min at rt. The reaction mixture was diluted with EtOAc (5 mL) and washed twice with 8% NaHCO3(aq, 5+3 mL) followed by H2O (2×5 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by normal phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH, 10:1). The compound was further purified twice by preparative HPLC, PrepMethod G, (gradients: 15-55% and 5-45%) to give the title compound (0.041 g, 38%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750 found: 440.1754;1H NMR (500 MHz, DMSO-d6) δ 9.03 (t, 1H), 8.67 (d, 1H), 7.90 (d, 1H), 7.70 (d, 1H), 7.65 (dd, 1H), 7.39 (d, 1H), 5.30 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.31 (d, 2H), 4.15-4.06 (m, 2H), 3.44-3.37 (m, overlapping with solvent), 3.08 (dd, 2H), 1.25 (d, 6H). Example 79: 6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide HATU (0.137 g, 0.36 mmol) was added to a stirred mixture of crude 6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)quinoline-4-carboxylic acid Intermediate 166 (0.30 mmol) and DIPEA (0.262 mL, 1.50 mmol) in a mixture of MeCN/EtOAc (2.8 mL, 1:1) at rt. The reaction mixture was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.075 g, 0.36 mmol) was added and the reaction was stirred for 3 h at rt. The reaction mixture was diluted with EtOAc (5 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc (3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The crude compound was purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH, 6:1) and further purified by preparative HPLC, PrepMethod G, (gradient: 5-50%). The compound was dissolved in EtOAc and the organic layer was washed with 8% NaHCO3(aq, 2×) and H2O (2×), concentrated and freeze-dried to give the title compound (0.051 g, 39%) a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O3S: 438.1594 found: 438.1608;1H NMR (500 MHz, DMSO-d6) δ 9.00 (t, 1H), 8.66 (d, 1H), 7.89 (d, 1H), 7.64-7.58 (m, 2H), 7.39 (d, 1H), 5.32 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.48 (s, 2H), 4.36-4.23 (m, 2H), 3.61 (d, 2H), 3.41 (dd, 1H), 3.36 (dd, 1H), 2.95 (ddd, 2H), 1.86 (s, 4H). Example 80: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-methyl-morpholino)quinoline-4-carboxamide HATU (0.137 g, 0.36 mmol) was added to a stirred mixture of crude (S)-6-(2-methylmorpholino)quinoline-4-carboxylic acid Intermediate 168 (0.30 mmol) and DIPEA (0.262 mL, 1.50 mmol) in a mixture of MeCN/EtOAc (2.8 mL, 1:1) at rt. The reaction mixture was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.075 g, 0.36 mmol) was added and the reaction was stirred for 45 min at rt. The reaction mixture was diluted with EtOAc (5 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc (3 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient of 5-45%). The compound was dissolved in EtOAc and the organic layer was washed with 8% NaHCO3(aq) and H2O (3×), concentrated and freeze-dried. The compound was further purified by normal phase flash chromatography on silica gel (EtOAc followed by EtOAc:MeOH, 20:1), and freeze-dried from MeCN/H2O to give the title compound (0.041 g, 32%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594 found: 426.1588;1H NMR (500 MHz, DMSO-d6) δ 9.04 (t, 1H), 8.69 (d, 1H), 7.91 (d, 1H), 7.74-7.66 (m, 2H), 7.40 (d, 1H), 5.30 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.31 (d, 2H), 3.96 (dd, 1H), 3.87 (d, 1H), 3.75 (d, 1H), 3.72-3.63 (m, 2H), 3.44-3.34 (m, 2H), 2.78 (td, 1H), 2.47 (d, 1H), 1.22 (d, 3H). Example 81: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3S)-2,3-dimethyl-morpholino)quinoline-4-carboxamide HATU (0.160 g, 0.42 mmol) was added to a stirred mixture of crude 6-((2R,3S)-2,3-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 170 (0.35 mmol) and DIPEA (0.306 mL, 1.75 mmol) in a mixture of MeCN/EtOAc (3.2 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.087 g, 0.42 mmol) was added and the reaction was stirred for 2.5 h at rt. The reaction mixture was diluted with EtOAc (20 mL) and washed with 8% NaHCO3(aq, 10 mL) followed by H2O (2 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH, 20:1). The compound was further purified by preparative HPLC, PrepMethod G, (gradient: 0-30% over 30 min) followed by straight phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH, 20:1). The compound was freeze-dried from MeCN/H2O to give the title compound (0.073 g, 47%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1752;1H NMR (500 MHz, DMSO-d6) δ 9.01 (t, 1H), 8.65 (d, 1H), 7.90 (d, 1H), 7.68-7.60 (m, 2H), 7.38 (d, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.70 (d, 1H), 4.29 (d, 2H), 4.14-4.08 (dd, 1H), 3.98 (dd, 1H), 3.86-3.78 (m, 1H), 3.63 (td, 1H), 3.47-3.34 (m, 3H), 3.06 (td, 1H), 1.14 (d, 3H), 0.93 (d, 3H). Example 82: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3S)-2,3-dimethyl-morpholino)quinoline-4-carboxamide HATU (0.114 g, 0.30 mmol) was added to a stirred mixture of crude 6-((2S,3S)-2,3-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 172 (0.25 mmol) and DIPEA (0.218 mL, 1.25 mmol) in a mixture of MeCN/EtOAc (2.2 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol) was added and the reaction was stirred for 1.5 h at rt. The reaction mixture was diluted with EtOAc (15 mL) and washed with 8% NaHCO3(aq, 6+3 mL) and H2O (2×2 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc/MeOH, 20:1). The compound was further purified by preparative HPLC, PrepMethod G, (gradient: 0-30% in 30 min) to give the title compound (0.056 g, 51%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750 found: 440.1750;1H NMR (500 MHz, DMSO-d6) δ 9.03 (t, 1H), 8.71 (d, 1H), 7.92 (d, 1H), 7.77 (d, 1H), 7.63 (dd, 1H), 7.41 (d, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.30 (d, 2H), 3.92-3.84 (m, 1H), 3.75-3.64 (m, 3H), 3.45-3.35 (m, 2H), 3.27-3.18 (m, 2H), 1.34 (d, 3H), 1.06 (d, 3H). Example 83: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3R)-2,3-dimethyl-morpholino)quinoline-4-carboxamide HATU (0.110 g, 0.29 mmol) was added to a stirred mixture of crude 6-((2R,3R)-2,3-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 174 (0.24 mmol) and DIPEA (0.210 mL, 1.20 mmol) in a mixture of MeCN/EtOAc (2.2 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.060 g, 0.29 mmol) was added and the reaction was stirred for 1.5 h at rt. The reaction mixture was diluted with EtOAc (15 mL) and washed with 8% NaHCO3(aq, 6+3 mL) followed by H2O (2×2 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH, 10:1). The compound was further purified by preparative HPLC, PrepMethod G, (gradient: 5-35%) to give the title compound (0.048 g, 46%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750 found: 440.1756;1H NMR (500 MHz, DMSO-d6) δ 9.03 (t, 1H); 8.71 (d, 1H), 7.92 (d, 1H), 7.73 (d, 1H), 7.63 (dd, 1H), 7.42 (d, 1H), 5.30 (dd, 1H), 4.90 (d, 1H), 4.70 (d, 1H), 4.34 (dd, 1H), 4.26 (dd, 1H), 3.93-3.84 (m, 1H), 3.75-3.65 (m, 3H), 3.43-3.34 (m, 2H), 3.30-3.17 (m, 2H), 1.34 (d, 3H), 1.07 (d, 3H). Example 84: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-(trifluoromethyl)-morpholino)quinoline-4-carboxamide Isomer 1 HATU (0.119 g, 0.31 mmol) was added to a stirred solution of crude rel-(R)-6-(3-(trifluoromethyl)morpholino)quinoline-4-carboxylic acid Isomer 1 Intermediate 178 (0.26 mmol) and DIPEA (0.227 mL, 1.30 mmol) in a mixture of MeCN/EtOAc (2.4 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.065 g, 0.31 mmol) was added and the reaction was stirred for 45 min at rt. The reaction was diluted with EtOAc (8 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-55%) to give the title compound (0.082 g, 66%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H21F3N5O3S: 480.1312, found: 480.1308;1H NMR (500 MHz, DMSO-d6) δ 9.07 (t, 1H), 8.74 (d, 1H), 7.94 (d, 1H), 7.85-7.78 (m, 2H), 7.45 (d, 1H), 5.34 (dd, 1H), 5.11-5.01 (m, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.38-4.27 (m, 2H), 4.24 (d, 1H), 4.06 (dd, 1H), 3.89-3.82 (m, 1H), 3.64 (td, 1H), 3.55-3.33 (m, overlapping with solvent). Example 85: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-(trifluoromethyl)-morpholino)quinoline-4-carboxamide Isomer 2 HATU (0.128 g, 0.34 mmol) was added to a stirred solution of crude rel-(R)-6-(3-(trifluoromethyl)morpholino)quinoline-4-carboxylic acid Isomer 2 Intermediate 179 (0.150 g, 0.28 mmol) and DIPEA (0.245 mL, 1.40 mmol) in a mixture of MeCN/EtOAc (2.4 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.070 g, 0.34 mmol) was added and the reaction was stirred for 1.5 h at rt. The reaction was diluted with EtOAc (8 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-55%) to give the title compound (0.085 g, 63%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H21F3N5O3S: 480.1312, found: 480.1308;1H NMR (500 MHz, DMSO-d6) δ 9.07 (t, 1H), 8.73 (d, 1H), 7.94 (d, 1H), 7.83-7.76 (m, 2H), 7.45 (d, 1H), 5.29 (dd, 1H), 5.11-5.03 (dd, 1H), 4.90 (d, 1H), 4.70 (d, 1H), 4.37 (dd, 1H), 4.30-4.20 (m, 2H), 4.06 (dd, 1H), 3.89-3.81 (m, 1H), 3.64 (td, 1H), 3.56-3.32 (m, overlapping with solvent). Example 86: 6-(3-Oxa-9-azabicyclo[3.3.1]nonan-9-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide HATU (0.068 g, 0.18 mmol) was added to a stirred solution of the crude 6-(3-Oxa-9-azabicyclo[3.3.1]nonan-9-yl)quinoline-4-carboxylic acid Intermediate 181 (0.070 g, 0.15 mmol) and DIPEA (0.131 mL, 0.75 mmol) in a mixture of MeCN/EtOAc (1.2 mL, 1:1) at rt and the reaction was stirred for 1 min. (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.037 g, 0.18 mmol) was added and the reaction was stirred for 2 h at rt. The reaction mixture was diluted with EtOAc (8 mL) and washed with 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-55%) to give the title compound (0.040 g, 60%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O3S: 452.1750, found: 452.1756;1H NMR (500 MHz, DMSO-d6) δ 9.01 (t, 1H), 8.62 (d, 1H), 7.89 (d, 1H), 7.73 (d, 1H), 7.61 (dd, 1H), 7.37 (d, 1H), 5.29 (dd, 1H), 4.89 (d, 1H), 4.69 (d, 1H), 4.35-4.22 (m, 2H), 4.11 (s, 2H), 3.99 (d, 2H), 3.85 (d, 2H), 3.44-3.35 (m, overlapping with solvent), 2.55-2.52 (m, overlapping with solvent), 1.98-1.83 (m, 2H), 1.83-1.70 (m, 2H), 1.58-1.48 (m, 1H). Example 87: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,5R)-2,5-dimethyl-morpholino)quinoline-4-carboxamide HATU (0.160 g, 0.42 mmol) was added to a stirred mixture of the crude 6-((2R,5R)-2,5-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 183 (0.35 mmol) and DIPEA (0.305 mL, 1.75 mmol) in a mixture of MeCN/EtOAc (3 mL, 1:1) and the mixture was stirred for 1 min. (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.087 g, 0.42 mmol) was added and the mixture was stirred at rt for 3 h. The reaction mixture was partitioned between EtOAc (25 mL) and 8% NaHCO3(aq, 10 mL). The aqueous layer was extracted with EtOAc (10 mL) and the combined organic layers were concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-45%) to give the title compound (0.072 g, 47%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1748;1H NMR (500 MHz, DMSO-d6) δ 9.03 (t, 1H), 8.66 (d, 1H), 7.91 (d, 1H), 7.70-7.64 (m, 2H), 7.39 (d, 1H), 5.31 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.34 (dd, 1H), 4.26 (dd, 1H), 4.20-4.14 (m, 1H), 3.85-3.73 (m, 2H), 3.69-3.53 (m, 2H), 3.45-3.33 (m, overlapping with solvent), 2.75 (dd, 1H), 1.27 (d, 3H), 1.05 (d, 3H). Example 88: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,2-dimethyl-morpholino)quinoline-4-carboxamide HATU (0.160 g, 0.42 mmol) was added to a stirred mixture of the crude 6-(2,2-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 185 (0.35 mmol) and DIPEA (0.305 mL, 1.75 mmol) in a mixture of MeCN/EtOAc (3 mL, 1:1) and the mixture was stirred for 1 min. (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.087 g, 0.42 mmol) was added and the mixture was stirred at rt for 3 h. The reaction mixture was partitioned between EtOAc (25 mL) and 8% NaHCO3(aq, 10 mL). The aqueous layer was extracted with EtOAc (10 mL) and the combined organic layers were concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient: 5-55%) to give the title compound (0.076 g, 49%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1768;1H NMR (500 MHz, DMSO-d6) δ 9.05 (t, 1H), 8.69 (d, 1H), 7.91 (d, 1H), 7.74 (d, 1H), 7.68 (dd, 1H), 7.41 (d, 1H), 5.30 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.36-4.26 (m, 2H), 3.81 (t, 2H), 3.47-3.15 (m, overlapping with solvent), 1.28 (s, 6H). Example 89: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(methoxymethyl)-morpholino)quinoline-4-carboxamide 4 M HCl in dioxane (2 mL, 8 mmol) was added to a vial containing tert-butyl (S)-6-(3-(methoxymethyl)morpholino)quinoline-4-carboxylate Intermediate 186 (146 mg, 0.41 mmol). The reaction was heated at 60° C. for 1 h. The volatiles were removed under reduced pressure and the residue was suspended in EtOAc and concentrated (×2). A mixture of MeCN/EtOAc (4.8 mL, 1:1) was added to the residue at rt, followed by DIPEA (0.430 mL, 2.46 mmol) and HATU (0.187 g, 0.49 mmol). The mixture was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.102 g, 0.49 mmol) was added and the reaction was stirred for 1.5 h at rt. The reaction mixture was diluted with EtOAc and washed with 8% NaHCO3(aq). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-D, to give the title compound (0.062 g, 33%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O4S: 456.1700, found: 456.1698;1H NMR (500 MHz, DMSO-d6) δ 9.00 (t, 1H), 8.67 (d, 1H), 7.91 (d, 1H), 7.69 (dd, 1H), 7.64 (d, 1H), 7.40 (d, 1H), 5.31 (dd, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.35-4.23 (m, 2H), 4.12 (s, 1H), 4.02-3.95 (m, 2H), 3.70-3.64 (m, 2H), 3.58 (td, 1H), 3.45-3.42 (m, overlapping with solvent), 3.24-3.13 (m, overlapping with solvent). Example 90: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5R)-3,5-dimethyl-morpholino)quinoline-4-carboxamide HATU (0.091 g, 0.24 mmol) was added to a stirred solution of the crude 6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 188 (0.2 mmol) and DIPEA (0.210 mL, 1.20 mmol) in a mixture of MeCN/H2O (2.4 mL, 1:1) at rt. The reaction mixture was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.050 g, 0.24 mmol) was added. The reaction mixture was stirred for 3 h at rt, diluted with EtOAc and washed with 0.4 M NaOH (aq, 4.5 mL). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-D, to give the title compound (0.039 g, 44%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1744;1H NMR (500 MHz, DMSO-d6) δ 9.01 (t, 1H), 8.69 (d, 1H), 7.95 (d, 1H), 7.72 (d, 1H), 7.59 (dd, 1H), 7.41 (d, 1H), 5.31 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.36-4.23 (m, 2H), 3.87-3.81 (m, 2H), 3.75-3.66 (m, 4H), 1.09 (d, 6H). Example 91: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,4-oxazepan-4-yl)-quinoline-4-carboxamide A vial was charged with tert-butyl 6-(1,4-oxazepan-4-yl)quinoline-4-carboxylate Intermediate 189 (0.085 g, 0.25 mmol) and 90% TFA (aq, 0.5 mL) and the reaction mixture was heated at 50° C. for 3 h. The reaction mixture was concentrated, a mixture of heptane and DCM (3 mL, 2:1) was added to the residue and the mixture was concentrated. A mixture of MeCN/EtOAc (3 mL, 1:1) and DIPEA (0.261 mL, 1.50 mmol) was added to the residue followed by HATU (0.114 g, 0.30 mmol). The mixture was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol) was added. The mixture was stirred at rt for 4 h and then partitioned between EtOAc (4 mL) and 8% NaHCO3(aq, 5 mL). The aqueous layer was extracted with EtOAc (2×1 mL) and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-A, followed by preparative HPLC, PrepMethod V, (gradient: 5-95%) to give the title compound (30 mg, 27%); HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594 found: 426.1576;1H NMR (600 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.53 (d, 1H), 7.82 (d, 1H), 7.50-7.45 (m, 2H), 7.30 (d, 1H), 5.26 (dd, 1H), 4.84 (d, 1H), 4.66 (d, 1H), 4.30-4.18 (m, 2H), 3.76-3.66 (m, overlapping with solvent), 3.57-3.51 (m, overlapping with solvent), 3.38-3.29 (m, overlapping with solvent), 1.95-1.88 (m, 2H). Example 92: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-((methylsulfonyl)-methyl)morpholino)quinoline-4-carboxamide The compound was synthesized and purified analogous to the procedure of Example 91 starting from tert-butyl (R)-6-(2-((methylsulfonyl)methyl)morpholino)quinoline-4-carboxylate Intermediate 193 (0.102 g, 0.25 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol) to give the title compound (26 mg, 20%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O5S2: 504.1370 found: 504.1372;1H NMR (600 MHz, DMSO-d6) δ 9.00 (m, 1H), 8.68 (d, 1H), 7.91 (d, 1H), 7.68 (d, 1H), 7.62 (dd, 1H), 7.40 (d, 1H), 5.31-5.27 (m, 1H), 4.86 (d, 1H), 4.67 (d, 1H), 4.31 (dd, 1H), 4.23 (dd, 1H), 4.10-3.99 (m, 2H), 3.88-3.81 (m, 1H), 3.75-3.65 (m, 2H), 3.53 (dd, overlapping with solvent), 3.37-3.27 (m, overlapping with solvent), 3.01 (s, 3H), 2.91-2.82 (m, 1H), 2.73-2.68 (m, 1H). Example 93: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(methoxymethyl)-morpholino)quinoline-4-carboxamide The compound was synthesized and purified analogous to the procedure of Example 91 starting from tert-butyl (S)-6-(2-(methoxymethyl)morpholino)quinoline-4-carboxylate Intermediate 194 (76 mg, 0.25 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol) to give the title compound (0.026 g, 22%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O4S: 456.1700 found: 456.1712;1H NMR (600 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.66 (d, 1H), 7.88 (d, 1H), 7.69 (d, 1H), 7.64 (dd, 1H), 7.38 (d, 1H), 5.29 (dd, 1H), 4.85 (d, 1H), 4.68 (d, 1H), 4.32-4.22 (m, 2H), 3.99-3.93 (m, 1H), 3.78-3.68 (m, 3H), 3.64 (td, 1H), 3.48-3.42 (m, overlapping with solvent), 3.35-3.31 (m, overlapping with solvent), 3.27 (s, 3H), 2.80 (td, 1H), 2.61-2.56 (m, overlapping with solvent). Example 94: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-((methylsulfonyl)-methyl)morpholino)quinoline-4-carboxamide The compound was synthesized analogous to the procedure of Example 91 starting from tert-butyl (S)-6-(2-((methylsulfonyl)methyl)morpholino)quinoline-4-carboxylate Intermediate 198 (0.102 g, 0.25 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol). The compound was purified by preparative SFC, PrepMethod SFC-A, followed by PrepMethod SFC-D, to give the title compound (0.023 g, 18%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O5S2: 504.1370, found: 504.1364;1H NMR (600 MHz, DMSO-d6) δ 9.00 (t, 1H), 8.68 (d, 1H), 7.91 (d, 1H), 7.71 (d, 1H), 7.62 (dd, 1H), 7.39 (d, 1H), 5.31-5.26 (m, 1H), 4.85 (d, 1H), 4.67 (d, 1H), 4.32-4.22 (m, 2H), 4.09-3.98 (m, 2H), 3.86 (d, 1H), 3.70 (t, 2H), 3.52 (dd, overlapping with solvent), 3.34-3.27 (m, overlapping with solvent), 3.01 (s, 3H), 2.93-2.84 (m, 1H), 2.74-2.67 (m, 1H). Example 95: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-(2-methoxyethyl)-morpholino)quinoline-4-carboxamide The compound was synthesized analogous to the procedure of Example 91 starting from tert-butyl (R)-6-(3-(2-methoxyethyl)morpholino)quinoline-4-carboxylate Intermediate 201 (0.093 g, 0.25 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol). The compound was purified by preparative SFC, PrepMethod SFC-A followed by preparative HPLC, PrepMethod F, to give the title compound (0.031 g, 25%); HRMS (ESI) m/z [M+H]+calcd for C23H28N5O4S: 470.1856, found: 470.1846;1H NMR (600 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.62 (d, 1H), 7.87 (d, 1H), 7.59-7.56 (m, 2H), 7.36 (d, 1H), 5.26 (dd, 1H), 4.85 (d, 1H), 4.67 (d, 1H), 4.30 (dd, 1H), 4.22 (dd, 1H), 4.05-4.00 (m, 1H), 3.93 (dd, 1H), 3.84 (d, 1H), 3.66-3.63 (m, 2H), 3.58-3.48 (m, overlapping with solvent), 3.46-3.42 (m, overlapping with solvent), 3.39-3.29 (m, overlapping with solvent), 3.28-3.21 (m, 1H), 3.17-3.10 (m, overlapping with solvent), 3.09 (s, 3H), 1.97-1.88 (m, 1H), 1.65-1.57 (m, 1H). Example 96: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2S,3S)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxamide The compound was synthesized and purified analogous to the procedure of Example 91 starting from tert-butyl 6-((2S,3S)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxylate Intermediate 204 (0.093 g, 0.25 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol) to give the title compound (0.030 g, 25%); HRMS (ESI) m/z [M+H]+calcd for C23H28N5O4S: 470.1856 found: 470.1848;1H NMR (600 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.63 (d, 1H), 7.87 (d, 1H), 7.63-7.58 (m, 2H), 7.36 (d, 1H), 5.26 (dd, 1H), 4.85 (d, 1H), 4.67 (d, 1H), 4.29 (dd, 1H), 4.22 (dd, 1H), 4.05-3.98 (m, 1H), 3.88 (td, 1H), 3.83-3.80 (m, 1H), 3.67-3.61 (m, overlapping with solvent), 3.42-3.28 (m, overlapping with solvent), 3.17 (dd, 1H), 3.15-3.12 (m, 5H), 1.35 (d, 3H). Example 97: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((2R,3R)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxamide The compound was synthesized analogous to the procedure of Example 91 starting from tert-butyl 6-((2R,3R)-3-(methoxymethyl)-2-methylmorpholino)quinoline-4-carboxylate Intermediate 207 (0.093 g, 0.25 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol). The compound was purified by preparative SFC, PrepMethod SFC-A, followed by PrepMethod SFC-D, to give the title compound (0.045 g, 36%); HRMS (ESI) m/z [M+H]+calcd for C23H28N5O4S: 470.1856 found: 470.1856;1H NMR (600 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.63 (d, 1H), 7.87 (d, 1H), 7.65 (d, 1H), 7.61 (dd, 1H), 7.37 (d, 1H), 5.27 (dd, 1H), 4.85 (d, 1H), 4.67 (d, 1H), 4.26 (d, 2H), 4.05-3.98 (m, 1H), 3.87 (td, 1H), 3.84-3.78 (m, 1H), 3.69-3.61 (m, 2H), 3.43-3.27 (m, overlapping with solvent), 3.19 (td, 1H), 3.15 (s, 3H), 1.35 (d, 3H). Example 98: 6-(3-Oxa-8-azabicyclo[3.2.1]octan-8-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide The compound was synthesized and purified analogous to the procedure of Example 91 starting from tert-butyl 6-(3-oxa-8-azabicyclo[3.2.1]octan-8-yl)quinoline-4-carboxylate Intermediate 208 (0.085 g, 0.25 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol) to give the title compound (0.054 g, 49%); HRMS (ESI) m/z [M+H]+calcd for C22H24N5O3S: 438.1594, found: 438.1606;1H NMR (500 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.62 (d, 1H), 7.88 (d, 1H), 7.71 (d, 1H), 7.57 (dd, 1H), 7.35 (d, 1H), 5.29 (dd, 1H), 4.89 (d, 1H), 4.69 (d, 1H), 4.43-4.36 (m, 2H), 4.29 (d, 2H), 3.78 (dd, 2H), 3.50 (dd, 2H), 3.43-3.34 (m, overlapping with solvent), 2.04-1.93 (m, 4H). Example 99: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,9-dioxa-4-azaspiro[5.5]undecan-4-yl)quinoline-4-carboxamide The compound was synthesized and purified analogous to the procedure of Example 91 starting from tert-butyl 6-(1,9-dioxa-4-azaspiro[5.5]undecan-4-yl)quinoline-4-carboxylat Intermediate 209 (0.096 g, 0.25 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.062 g, 0.30 mmol) to give the title compound (0.029 g, 24%); HRMS (ESI) m/z [M+H]+calcd for C24H28N5O4S: 482.1856, found: 482.1846;1H NMR (600 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.64 (d, 1H), 7.87 (d, 1H), 7.70 (d, 1H), 7.64 (dd, 1H), 7.36 (d, 1H), 5.26 (dd, 1H), 4.85 (d, 1H), 4.67 (d, 1H), 4.32-4.22 (m, 2H), 3.80 (t, overlapping with solvent), 3.62-3.54 (m, overlapping with solvent), 3.39-3.31 (m, overlapping with solvent), 3.28-3.18 (m, overlapping with solvent), 1.81-1.72 (m, 2H), 1.71-1.62 (m, 2H). Example 100: 7-Bromo-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5R)-3,5-dimethylmorpholino)quinoline-4-carboxamide HATU (13.5 mg, 0.04 mmol) was added to a solution of the crude 7-bromo-6-((3R,5S)-3,5-dimethylmorpholino)quinoline-4-carboxylic acid Intermediate 212 (0.020 g, 0.03 mmol) and DIPEA (25.9 μL, 0.15 mmol) in a mixture of MeCN/EtOAc (1 mL, 1:1) at rt. The reaction was stirred for 1.5 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (7 mg, 0.04 mmol) was added and the reaction was stirred for 2 h at rt. The reaction mixture was diluted with EtOAc and washed with 8% NaHCO3(aq). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-A, to give the title compound (5.7 mg, 37%); HRMS (ESI) m/z [M+H]+calcd for C22H25BrN5O3S: 518.0856, found: 518.0858;1H NMR (600 MHz, DMSO-d6) δ 9.14 (t, 1H), 8.96 (d, 1H), 8.41 (s, 1H), 8.38 (s, 1H), 7.59 (d, 1H), 5.30 (dd, 1H), 4.85 (d, 1H), 4.69 (d, 1H), 4.31 (d, 2H), 3.80 (d, overlapping with solvent), 3.37-3.31 (m, 2H), 3.17-3.08 (m, overlapping with solvent), 0.58 (d, 6H). Example 101: (R)-5-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholino-quinoline-4-carboxamide Step a) 2 M NaOH (aq, 1.025 mL, 2.05 mmol) was added to a suspension of ethyl 5-chloro-6-morpholinoquinoline-4-carboxylate hydrochloride Intermediate 213 (8 mg, 0.23 mmol) in MeOH (1.4 mL) and the reaction was heated at 80° C. for 20 min. Aq NaOH (3.8 M, 0.54 mL, 2.1 mmol) was added and the reaction was heated at 120° C. for 50 min. After cooling to rt, aq HCl (3.8 M, 1.2 mL, 4.6 mmol) was added dropwise and the resulting mixture was concentrated. The residue was co-evaporated once from H2O and twice from EtOAc to give crude 5-chloro-6-morpholinoquinoline-4-carboxylic acid as a solid; MS (ESI) m/z [M+H]+293.1. Step b) A mixture of MeCN/EtOAc (3 mL, 1:1) and DIPEA was added to the crude 5-chloro-6-morpholinoquinoline-4-carboxylic acid, followed by HATU (96 mg, 0.25 mmol). The reaction mixture was stirred for 2 min at rt after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (52 mg, 0.25 mmol) was added and the reaction mixture was stirred for 4.5 h at rt. The reaction mixture was diluted with EtOAc and washed with 8% NaHCO3(aq). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-D, to give the title compound (0.042 g, 41%); HRMS (ESI) m/z [M+H]+calcd for C20H21ClN5O3S: 446.1048, found: 446.1044;1H NMR (600 MHz, DMSO-d6) δ 8.91 (t, 1H), 8.85 (d, 1H), 8.03 (d, 1H), 7.73 (d, 1H), 7.46 (d, 1H), 5.30 (dd, 1H), 4.85 (d, 1H), 4.68 (d, 1H), 4.45-4.05 (m, 2H), 3.75 (t, 4H), 3.36 (dd, 1H), 3.30 (dd, 1H), 3.09 (brs, 4H). Example 102: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methyl-morpholino)quinoline-4-carboxamide A solution of tert-butyl (S)-6-(3-methylmorpholino)quinoline-4-carboxylate Intermediate 214 (48 mg, 0.15 mmol) in 90% TFA (aq, 0.5 mL) was stirred at rt for 5 h. The volatiles were removed under reduced pressure and the residue was suspended in a mixture of heptane/DCM, concentrated (2×) and dried under vacuum. HATU (0.068 g, 0.18 mmol) was added to a solution of the residue in a mixture of MeCN/EtOAc (2 mL, 1:1) and DIPEA (0.131 ml, 0.75 mmol). The reaction mixture was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.037 g, 0.18 mmol) was added. The resulting solution was stirred for 1 h at rt, diluted with EtOAc and washed with 8% NaHCO3(aq). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-C, to give the title compound (0.026 g, 41%); HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1594;1H NMR (600 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.66 (d, 1H), 7.89 (d, 1H), 7.63 (dd, 2H), 7.40 (d, 1H), 5.28 (dd, 1H), 4.86 (d, 1H), 4.68 (d, 1H), 4.27 (d, 2H), 4.16-4.11 (m, 1H), 3.96 (dd, 1H), 3.72 (d, 2H), 3.54 (dd, 1H), 3.43-3.35 (m, overlapping with solvent), 3.33 (dd, 1H), 3.16-3.12 (m, 1H), 1.04 (d, 3H). Example 103: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,5S)-3,5-dimethyl-morpholino)quinoline-4-carboxamide A solution of tert-butyl 6-((3S,5S)-3,5-dimethylmorpholino)quinoline-4-carboxylate Intermediate 215 (54 mg, 0.16 mmol) in 90% TFA (aq, 0.5 mL) was stirred at rt for 5 h. The volatiles were removed under reduced pressure and the residue was suspended in a mixture of heptane/DCM, concentrated (2×) and dried under vacuum. HATU (0.073 g, 0.19 mmol) was added to a stirred solution of the residue in a mixture of MeCN/EtOAc (2 mL, 1:1) at rt. The reaction was stirred for 1 min, and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.040 g, 0.19 mmol) was added. The resulting solution was stirred for 45 min at rt, diluted with EtOAc and washed with 8% NaHCO3(aq). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-D, to give the title compound (0.029 g, 41%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1738;1H NMR (600 MHz, DMSO-d6) δ 9.02 (t, 1H), 8.76 (d, 1H), 7.91 (d, 1H), 7.79 (d, 1H), 7.58 (dd, 1H), 7.44 (d, 1H), 5.29 (dd, 1H), 4.85 (d, 1H), 4.68 (d, 1H), 4.31-4.24 (m, 2H), 3.84 (dd, overlapping with solvent), 3.72-3.67 (m, overlapping with solvent), 3.42 (dd, 2H), 3.38-3.31 (m, 2H), 0.87 (d, 6H). Example 104: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(8-oxa-5-azaspiro[3.5]nonan-5-yl)quinoline-4-carboxamide A solution of tert-butyl 6-(8-oxa-5-azaspiro[3.5]nonan-5-yl)quinoline-4-carboxylate Intermediate 216 (103 mg, 0.29 mmol) in 90% TFA (aq, 1 mL) was stirred at 50° C. for 20 min. After cooling to rt the solution was concentrated under reduced pressure and the residue was suspended in a mixture of heptane/DCM and concentrated. This was repeated 3× and the residue was dried under vacuum overnight. HATU (0.132 g, 0.35 mmol) was added to a stirred solution of the residue (0.153 g, 0.29 mmol) and DIPEA (0.253 mL, 1.45 mmol) in a mixture of MeCN/EtOAc (3 mL, 1:1) at rt. The reaction was stirred for 1 min and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.072 g, 0.35 mmol) was added. The resulting solution was stirred at rt for 80 min, diluted with EtOAc and washed with 8% NaHCO3(aq). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-C, to give the title compound (0.042 g, 32%); HRMS (ESI) m/z [M+H]+calcd for C23H26N5O3S: 452.1750, found: 452.1754;1H NMR (600 MHz, DMSO-d6) δ 9.00 (t, 1H), 8.73 (d, 1H), 7.88 (d, 1H), 7.56 (d, 1H), 7.48-7.41 (m, 2H), 5.28 (dd, 1H), 4.86 (d, 1H), 4.68 (d, 1H), 4.31 (dd, 1H), 4.21 (dd, 1H), 3.84-3.76 (m, 2H), 3.54-3.47 (m, overlapping with solvent), 3.37-3.29 (m, overlapping with solvent), 2.16-2.07 (m, 2H), 2.05-1.96 (m, 2H), 1.70-1.62 (m, 1H), 1.54-1.46 (m, 1H). Example 105: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,5R)-3,5-dimethyl-morpholino)quinoline-4-carboxamide A solution of tert-butyl 6-((3R,5R)-3,5-dimethylmorpholino)quinoline-4-carboxylate Intermediate 217 (84 mg, 0.25 mmol) in 90% TFA (aq, 2 mL) was stirred at rt for 5 h. After cooling to rt the solution was concentrated under reduced pressure and the residue was co-evaporated with a mixture of heptane/DCM (×2) and dried under vacuum overnight. HATU (132 mg, 0.35 mmol) was added to a stirred solution of the residue in a mixture of MeCN/EtOAc (3 mL, 1:1) and DIPEA (253 μL, 1.45 mmol) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (72 mg, 0.35 mmol) was added. The resulting solution was stirred for 1 h at rt, diluted with EtOAc and washed with 8% NaHCO3(aq). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative SFC, PrepMethod SFC-D, to give the title compound (0.043 g, 39%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1746;1H NMR (600 MHz, DMSO-d6) δ 9.02 (t, 1H), 8.77 (d, 1H), 7.92 (d, 1H), 7.72 (d, 1H), 7.58 (dd, 1H), 7.46 (d, 1H), 5.29 (dd, 1H), 4.86 (d, 1H), 4.69 (d, 1H), 4.33 (dd, 1H), 4.23 (dd, 1H), 3.84 (dd, 2H), 3.70-3.62 (m, 2H), 3.43 (dd, 2H), 3.40-3.29 (m, 2H), 0.87 (d, 6H). Example 106: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-ethyl-morpholino)quinoline-4-carboxamide HATU (103 mg, 0.27 mmol) was added to a stirred solution of the crude (S)-6-(3-ethylmorpholino)quinoline-4-carboxylic acid Intermediate 219 (116 mg) and DIPEA (197 μL, 1.13 mmol) in DMF (2 mL) at rt. The reaction was stirred for ˜1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (56.2 mg, 0.27 mmol) was added. The resulting solution was stirred for 2 h at rt. The reaction was diluted with DCM and washed with 8% NaHCO3(aq). The organic layer was concentrated and co-evaporated with heptane (×4) until most of the DMF was removed. The residue was purified by preparative SFC, PrepMethod SFC-D, to give the title compound (0.046 g, 47%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1748;1H NMR (600 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.63 (d, 1H), 7.87 (d, 1H), 7.64-7.58 (m, 2H), 7.37 (d, 1H), 5.28 (dd, 1H), 4.85 (d, 1H), 4.67 (d, 1H), 4.26 (dd, 2H), 3.93 (dd, 1H), 3.87 (d, 1H), 3.85-3.79 (m, 1H), 3.61 (dd, 1H), 3.56-3.51 (m, 1H), 3.44 (d, overlapping with solvent), 3.38-3.30 (m, overlapping with solvent), 3.18-3.12 (m, 1H), 1.77-1.66 (m, 1H), 1.36 (m, 1H), 0.82 (t, 3H). Example 107: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,3-dimethyl-morpholino)quinoline-4-carboxamide A solution of tert-butyl 6-(3,3-dimethylmorpholino)quinoline-4-carboxylate Intermediate 220 (51 mg, 0.15 mmol) in 90% TFA (aq, 1 mL) was stirred at rt for 5 h. The solution was concentrated under reduced pressure and the residue was suspended in a mixture of DCM/heptane and then concentrated (×2). The residue was dissolved in DMF (1 mL) and DIPEA (0.13 mL, 0.76 mmol), and HATU (69 mg, 0.18 mmol) was added at rt and the reaction mixture was stirred for 1 min, after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (38 mg, 0.18 mmol) was added. The resulting solution was stirred for 3 h at rt, diluted with DCM and washed with 8% NaHCO3(aq). The organic layer was concentrated and the residue was co-evaporated with heptane (×4) until most of the DMF was removed. The residue was purified by preparative SFC, PrepMethod SFC-A, to give the title compound (0.027 g, 41%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1748;1H NMR (600 MHz, DMSO-d6) δ 9.01 (t, 1H), 8.81 (d, 1H), 7.98 (d, 1H), 7.90 (d, 1H), 7.58 (dd, 1H), 7.47 (d, 1H), 5.30 (dd, 1H), 4.85 (d, 1H), 4.69 (d, 1H), 4.32-4.24 (m, 2H), 3.72 (t, 2H), 3.40 (s, 2H), 3.36 (dd, 1H), 3.32 (dd, 1H), 3.20-3.14 (m, 2H), 1.05 (s, 3H), 1.04 (s, 3H). Example 108: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methyl-morpholino)quinoline-4-carboxamide A solution of tert-butyl (R)-6-(3-methylmorpholino)quinoline-4-carboxylate Intermediate 221 (67 mg, 0.20 mmol) in 90% TFA (aq, 3 mL) was stirred at rt for 5 h. The solution was concentrated under reduced pressure and the residue was co-evaporated twice with heptane. The residue was dissolved in DMF (1 mL) and DIPEA (157 μL, 0.90 mmol), and HATU (82 mg, 0.22 mmol) was added at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (45 mg, 0.22 mmol) was added. The resulting solution was stirred for 2 h at rt, diluted with DCM and washed with 8% NaHCO3(aq). The organic layer was concentrated and the residue was co-evaporated with heptane (4×) until most of the DMF had been removed. The residue was purified by preparative SFC, PrepMethod SFC-C, to give the title compound (0.028 g, 37%); HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1602;1H NMR (600 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.63 (d, 1H), 7.88 (d, 1H), 7.63-7.58 (m, 2H), 7.36 (d, 1H), 5.27 (dd, 1H), 4.86 (d, 1H), 4.67 (d, 1H), 4.30 (dd, 1H), 4.22 (dd, 1H), 4.16-4.09 (m, 1H), 3.95 (dd, 1H), 3.71 (d, 2H), 3.55 (td, 1H), 3.42-3.30 (m, overlapping with solvent), 3.09 (td, 1H), 1.04 (d, 3H). Example 109: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-2-methyl-6-morpholino-quinoline-4-carboxamide Ethyl 2-methyl-6-morpholinoquinoline-4-carboxylate Intermediate 222 (89 mg, 0.30 mmol) was dissolved in MeOH (1 mL). 2 M NaOH (aq, 0.148 mL, 0.30 mmol) was added and the reaction mixture was stirred at rt for 2 h and was then evaporated to dryness. HATU (141 mg, 0.37 mmol) and DIPEA (0.155 mL, 0.89 mmol) were added to a suspension of the residue and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (61 mg, 0.30 mmol) in EtOAc (1 mL), MeCN (1.0 mL) and DMF (1.0 mL). The solution was stirred at rt overnight. The reaction mixture was evaporated and the residue was purified by preparative HPLC, PrepMethod Q, to give the title compound (0.6 mg, 0.5%); HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1594. Example 110: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxo-1-oxa-3-azaspiro[5.5]undecan-3-yl)quinoline-4-carboxamide A solution of tert-butyl 6-(2-oxo-1-oxa-3-azaspiro[5.5]undecan-3-yl)quinoline-4-carboxylate Intermediate 223 (115 mg, 0.29 mmol) in 90% TFA (2 mL) was stirred at 50° C. for 15 min. The reaction solution was concentrated and the residue was co-evaporated twice from water. A mixture of MeCN/EtOAc (2.6 mL, 1:1) and DIPEA (0.253 mL, 1.45 mmol) was added to the residue followed by HATU (0.132 g, 0.35 mmol). The reaction mixture was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.072 g, 0.35 mmol) was added. The reaction mixture was stirred for 1 h 15 min, diluted with EtOAc (20 mL) and washed with 8% NaHCO3(aq, 10 mL) followed by H2O (2 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc:MeOH, 20:1). The compound was further purified by preparative HPLC, PrepMethod G, (gradient: 10-50%) and preparative SFC, PrepMethod SFC-D, to give the title compound (0.046 g, 32%); HRMS (ESI) m/z [M+H]+calcd for C25H28N5O4S: 494.1856, found: 494.1838;1H NMR (500 MHz, DMSO-d6) δ 9.17 (t, 1H), 8.96 (d, 1H), 8.49 (d, 1H), 8.05 (d, 1H), 7.88 (dd, 1H), 7.56 (d, 1H), 5.32 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.34 (d, 2H), 3.93-3.79 (m, 2H), 3.43 (dd, 1H), 3.36 (dd, 1H), 2.09 (t, 2H), 1.91-1.82 (m, 2H), 1.72-1.60 (m, 4H), 1.58-1.50 (m, 3H), 1.45-1.32 (m, 1H). Example 111: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(7-oxo-6-oxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxamide A solution of tert-butyl 6-(7-oxo-6-oxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxylate Intermediate 225 (105 mg, 0.27 mmol) in 90% TFA (aq, 1 mL) was stirred at 50° C. for 15 min. The reaction solution was concentrated and the residue was dissolved in a mixture of water/MeCN and freeze-dried. A mixture of MeCN/EtOAc (2.4 mL, 1:1) was added to the residue followed by DIPEA (0.236 mL, 1.35 mmol) and HATU (0.123 g, 0.32 mmol). The reaction was stirred at rt for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.067 g, 0.32 mmol) was added. The reaction mixture was stirred for 2 h, diluted with EtOAc (20 mL) and washed with of 8% NaHCO3(aq, 10 mL) followed by water (2 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by straight phase flash chromatography on silica (EtOAc followed by EtOAc/MeOH 10:1). The compound was further purified by preparative HPLC, PrepMethod G, (gradients: 0-30% and 0-45%) and preparative SFC to give the title compound (0.031 g, 24%) as a an off-white solid; HRMS (ESI) m/z [M+H]+calcd for C24H26N5O4S: 480.1700, found: 480.1718;1H NMR (500 MHz, DMSO-d6) δ 9.17 (t, 1H), 8.96 (d, 1H), 8.49 (d, 1H), 8.05 (d, 1H), 7.89 (dd, 1H), 7.57 (d, 1H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.34 (d, 2H), 3.93-3.82 (m, 2H), 3.46-3.36 (m, 2H), 2.18 (t, 2H), 2.03-1.95 (m, 2H), 1.87-1.66 (m, 6H). Example 112: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6,6-dimethyl-2-oxo-1,3-oxazinan-3-yl)quinoline-4-carboxamide TFA (35 μL, 0.45 mmol) was added to a solution of tert-butyl 6-(6,6-dimethyl-2-oxo-1,3-oxazinan-3-yl)quinoline-4-carboxylate Intermediate 226 (40 mg, 0.11 mmol) in DCM (2 mL). The resulting solution was stirred at 25° C. for 6 h. The solvent was removed by distillation under vacuum. The residue was dissolved in DMF (2 mL) and (R)-3-glycyl-thiazolidine-4-carbonitrile hydrochloride Intermediate 4 (55 mg, 0.27 mmol), T3P (170 mg, 0.53 mmol) and DIPEA (69 mg, 0.53 mmol) were added. The resulting solution was stirred at 25° C. for 6 h. The solvent was removed under reduced pressure and the residue was purified by preparative TLC (DCM:MeOH 18:1) followed by preparative HPLC, PrepMethod B, (gradient: 11-41%) to give the title compound (0.028 g, 46%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O4S: 454.1544, found: 454.1554;1H NMR (300 MHz, DMSO-d6) δ 9.16 (t, 1H), 8.95 (d, 1H), 8.49 (d, 1H), 8.04 (d, 1H), 7.86 (dd, 1H), 7.55 (d, 1H), 5.31 (dd, 1H), 4.88 (d, 1H), 4.69 (d, 1H), 4.33 (d, 2H), 3.86 (t, 2H), 3.46-3.32 (m, overlapping with solvent), 2.07 (t, 2H), 1.44 (s, 6H). Example 113: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(fluoromethyl)-azetidin-1-yl)quinoline-4-carboxamide DIPEA (1.37 mL, 7.81 mmol) was added to a mixture crude 6-(3-(fluoromethyl)-azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 228 (0.436 g, 0.724 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (263 mg, 1.27 mmol), HOBt (428 mg, 3.16 mmol) and EDC (607 mg, 3.16 mmol) in EtOAc (5 mL) and MeCN (5 mL) at 13° C. The resulting solution was stirred at 13° C. overnight under N2(g). The solvent was removed under reduced pressure. The reaction mixture was diluted with sat NaHCO3(aq, 100 mL), and extracted with EtOAc (5×100 mL). The organic layers were combined and washed with sat NaCl (aq, 3×50 mL), dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod C, (gradient: 10-30%) and further purified by straight phase flash chromatography on silica (EtOAc:MeOH, 9:1) to give the title compound (0.103 g, 34%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H21FN5O2S: 414.1394, found: 414.1412;1H NMR (500 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.63 (d, 1H), 7.89 (d, 1H), 7.41 (d, 1H), 7.19 (d, 1H), 7.13 (dd, 1H), 5.34 (dd, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.70 (d, 1H), 4.60 (d, 1H), 4.30 (d, 2H), 4.07 (t, 2H), 3.78 (dd, 2H), 3.44-3.36 (m, 2H), 3.20-3.06 (m, 1H). Example 114: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,5,6,7-tetrahydro-1H-indazol-1-yl)quinoline-4-carboxamide DIPEA (0.429 mL, 2.45 mmol) was added to a stirred suspension of crude 6-(4,5,6,7-tetrahydro-1H-indazol-1-yl)quinoline-4-carboxylic acid Intermediate 240 (50 mg, 0.08 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (26 mg, 0.12 mmol), HOBt (55 mg, 0.41 mmol) and EDC (78 mg, 0.41 mmol) in MeCN (3 mL) and EtOAc (3 mL) at 18° C. The resulting solution was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The residue was partitioned between sat NaHCO3(20 mL) and EtOAc (50 mL). The aqueous layer was extracted with EtOAc (5×50 mL). The organic layers were combined and washed with H2O (3×20 mL). The aqueous layers were combined and extracted with EtOAc (3×15 mL). All organic layers were combined and dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod C, (gradient: 35-50%), to give the title compound (0.035 g, 96%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C23H23N6O2S: 447.1598, found: 447.1618;1H NMR (300 MHz, DMSO-d6) δ 9.30-9.10 (m, 1H), 9.01 (d, 1H), 8.42 (d, 1H), 8.28-8.04 (m, 2H), 7.72-7.50 (m, 2H), 5.33 (dd, 1H), 4.91 (d, 1H), 4.72 (d, 1H), 4.48-4.23 (m, 2H), 3.48-3.34 (m, overlapping with solvent), 2.99-2.79 (m, 2H), 2.65-2.50 (m, overlapping with solvent), 1.89-1.63 (m, 4H). Example 115: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,5,6,7-tetrahydro-2H-indazol-2-yl)quinoline-4-carboxamide DIPEA (4.21 mL, 24.1 mmol) was added to a stirred suspension of the crude 6-(4,5,6,7-tetrahydro-2H-indazol-2-yl)quinoline-4-carboxylic acid Intermediate 241 (465 mg, 0.59 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (185 mg, 0.89 mmol), HOBt (401 mg, 2.97 mmol) and EDC (569 mg, 2.97 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 18° C. The resulting solution was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The residue was partitioned between sat NaHCO3(aq, 50 mL) and EtOAc (100 mL). The aqueous layer was extracted with EtOAc (5×100 mL). The organic layers were combined and washed with H2O (3×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod C, (gradient: 35-45%), to give the title compound (0.122 g, 46%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C23H23N6O2S: 447.1598, found: 447.1594;1H NMR (300 MHz, DMSO-d6) δ 9.28-9.08 (m, 1H), 8.94 (d, 1H), 8.77 (s, 1H), 8.43-8.30 (m, 2H), 8.16 (d, 1H), 7.60 (d, 1H), 5.45-5.28 (m, 1H), 4.93 (d, 1H), 4.74 (d, 1H), 4.39 (d, 2H), 3.50-3.34 (m, overlapping with solvent), 2.80-2.52 (m, overlapping with solvent), 1.77 (m, 4H). Example 116: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-methyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxamide DIPEA (1.56 mL, 8.92 mmol) was added to a stirred suspension of crude 6-(5-methyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxylic acid Intermediate 242 (199 mg, 0.29 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (90 mg, 0.44 mmol), HOBt (196 mg, 1.45 mmol) and EDC (278 mg, 1.45 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 15° C. The resulting solution was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was partitioned between sat NaHCO3(aq, 50 mL) and EtOAc (100 mL). The aqueous layer was extracted with EtOAc (4×100 mL). The organic layers were combined and washed with H2O (3×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod C, (gradient: 40-52%) to give the title compound (0.090 g, 65%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C21H18F3N6O2S: 475.1158, found: 475.1138;1H NMR (300 MHz, DMSO-d6) δ 9.23 (t, 1H), 9.09 (d, 1H), 8.54 (d, 1H), 8.26 (d, 1H), 8.04 (dd, 1H), 7.71 (d, 1H), 6.83 (s, 1H), 5.40-5.20 (m, 1H), 4.87 (d, 1H), 4.69 (d, 1H), 4.36-4.28 (m, 2H), 3.42-3.32 (m, overlapping with solvent), 2.46 (s, overlapping with solvent). Example 117: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6,6-dimethyl-5,6-dihydrocyclopenta[c]pyrazol-2(4H)-yl)quinoline-4-carboxamide DIPEA (3.29 mL, 18.8 mmol) was added to a stirred suspension of crude 6-(6,6-dimethyl-5,6-dihydrocyclopenta[c]pyrazol-2(4H)-yl)quinoline-4-carboxylic acid Intermediate 243 (482 mg, 0.62 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (194 mg, 0.94 mmol), HOBt (420 mg, 3.14 mmol) and EDC (596 mg, 3.14 mmol) in MeCN (7 mL) and EtOAc (7 mL) at 15° C. The resulting solution was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure and the residue was partitioned between sat NaHCO3(aq, 60 mL) and EtOAc (100 mL). The aqueous layer was extracted with EtOAc (5×100 mL). The organic layers were combined and washed with H2O (3×50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod F, (gradient 42-52%) to give the title compound (0.14 g, 49%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C24H25N6O2S: 461.1754, found: 461.1742;1H NMR (300 MHz, DMSO-d6) δ 9.14 (t, 1H), 8.92 (d, 1H), 8.70 (d, 1H), 8.31 (dd, 1H), 8.22 (s, 1H), 8.13 (d, 1H), 7.58 (d, 1H), 5.44-5.24 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.36 (d, 2H), 3.46-3.30 (m, overlapping with solvent), 2.66 (t, 2H), 2.19 (t, 2H), 1.30 (s, 6H). Example 118: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxamide DIPEA (0.443 mL, 2.54 mmol) was added to a stirred suspension of 6-(3-(trifluoromethyl)-1H-pyrazol-1-yl)quinoline-4-carboxylic acid Intermediate 234 (78 mg, 0.25 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (79 mg, 0.38 mmol), HOBt (172 mg, 1.27 mmol) and EDC (243 mg, 1.27 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 10° C. The resulting suspension was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure and the residue was partitioned between sat NaHCO3(30 mL) and EtOAc (60 mL). The aqueous layer was extracted with EtOAc (4×75 mL). The organic layers were combined and washed with H2O (3×20 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod F, (gradient: 40-50%) to give the title compound (0.052 g, 43%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C20H16F3N6O2S: 461.1002, found: 461.1018;1H NMR (300 MHz, DMSO-d6) δ 9.22 (t, 1H), 9.04 (d, 1H), 8.94 (d, 1H), 8.88 (d, 1H), 8.38 (dd, 1H), 8.26 (d, 1H), 7.69 (d, 1H), 7.15-7.00 (m, 1H), 5.35 (dd, 1H), 4.91 (d, 1H) 4.73 (d, 1H), 4.44-4.25 (m, 2H), 3.70-3.34 (m, overlapping with solvent). Example 119: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,6-difluoro-1H-indol-1-yl)quinoline-4-carboxamide A solution of 6-(4,6-difluoro-1H-indol-1-yl)quinoline-4-carboxylic acid Intermediate 235 (180 mg, 0.56 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (173 mg, 0.83 mmol), EDC (213 mg, 1.11 mmol), HOBt (150 mg, 1.11 mmol) and DIPEA (485 μL, 2.78 mmol) in EtOAc (6 mL) and MeCN (6 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The reaction mixture was diluted with EtOAc (25 mL) and washed with H2O (3×10 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative TLC (DCM:MeOH 10:1) followed by preparative HPLC, PrepMethod U, (gradient 40-50%) to give the title compound (0.105 g, 40%); HRMS (ESI) m/z [M+H]+calcd for C24H18F2N5O2S: 478.1144, found: 478.1134;1H NMR (300 MHz, DMSO-d6) δ 9.22 (t, 1H), 9.06 (d, 1H), 8.57 (d, 1H), 8.29 (d, 1H), 8.10 (dd, 1H), 7.86 (d, 1H), 7.71 (d, 1H), 7.42-7.32 (m, 1H), 7.03 (td, 1H), 6.85 (d, 1H), 5.81-5.20 (m, 1H), 4.86 (d, 1H), 4.69 (d, 1H), 4.55-4.26 (m, 2H), 3.36-3.25 (m, overlapping with solvent). Example 120: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-fluoro-1H-indol-1-yl)quinoline-4-carboxamide TEA (137 μL, 0.98 mmol) was added to a stirred suspension of 6-(5-fluoro-1H-indol-1-yl)quinoline-4-carboxylic acid Intermediate 236 (30 mg, 0.10 mmol), (R)-3-glycyl-thiazolidine-4-carbonitrile hydrochloride Intermediate 4 (31 mg, 0.15 mmol), HOBt (66 mg, 0.49 mmol) and EDC (94 mg, 0.49 mmol) in MeCN (3 mL) and EtOAc (3 mL) at 10° C. The resulting suspension was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The residue was dissolved in a mixture of NaHCO3(aq, 30 mL) and EtOAc (60 mL). The aqueous layer was extracted with EtOAc (4×75 mL). The organic layers were combined and washed with water (3×25 mL). The aqueous layers were combined and extracted with EtOAc (3×25 mL). All organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod F, (gradient: 40-50%) to give the title compound (0.018 g, 40%) as a grey solid; HRMS (ESI) m/z [M+H]+calcd for C24H19FN5O2S: 460.1238, found: 460.1236;1H NMR (300 MHz, DMSO-d6) δ 9.19 (t, 1H), 9.03 (d, 1H), 8.54 (d, 1H), 8.28 (d, 1H), 8.10 (dd, 1H), 7.89 (d, 1H), 7.75 (dd, 1H), 7.68 (d, 1H), 7.47 (dd, 1H), 7.16-7.05 (m, 1H), 6.77 (d, 1H), 5.39-5.19 (m, 1H), 4.88 (d, 1H), 4.71 (d, 1H), 4.32 (d, 2H), 3.38-3.34 (m, overlapping with solvent). Example 121: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methyl-1H-pyrrol-1-yl)quinoline-4-carboxamide TEA (249 μL, 1.78 mmol) was added to a stirred suspension of 6-(3-methyl-1H-pyrrol-1-yl)quinoline-4-carboxylic acid Intermediate 237 (45 mg, 0.18 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (56 mg, 0.27 mmol), HOBt (121 mg, 0.89 mmol) and EDC (171 mg, 0.89 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 10° C. The resulting suspension was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The residue was partitioned between sat NaHCO3(30 mL) and EtOAc (60 mL). The aqueous layer was extracted with EtOAc (4×60 mL). The organic layers were combined and washed with water (3×30 mL). The aqueous layers were combined and extracted with EtOAc (2×20 mL). All the organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod F, (gradient: 35-47%) to give the title compound (0.021 g, 29%) as a pale yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H20N5O2S: 406.1332, found: 406.1326;1H NMR (300 MHz, DMSO-d6) δ 9.16 (t, 1H), 8.89 (d, 1H), 8.60-8.40 (m, 1H), 8.17-8.01 (m, 2H), 7.55 (d, 1H), 7.53-7.30 (m, 1H), 6.21-6.12 (m, 1H), 5.36 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.35 (d, 2H), 3.41-3.37 (m, overlapping with solvent), 2.11 (s, 3H). Example 122: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-morpholinoazetidin-1-yl)quinoline-4-carboxamide HATU (114 mg, 0.30 mmol) was added to a stirred mixture of crude 6-(3-morpholinoazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 239 (104 mg) and DIPEA (0.217 mL, 1.24 mmol) in a mixture of MeCN/EtOAc (2.4 mL, 1:1) at rt. The reaction was stirred for 1 min after which (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (62 mg, 0.30 mmol) was added and the reaction mixture was stirred for 2 h at rt. The reaction mixture was diluted with EtOAc (8 mL) and washed with 8% NaHCO3(aq, 6 mL). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by preparative HPLC, PrepMethod G, (gradient 0-30%), followed by straight phase flash chromatography on silica (EtOAc:MeOH, 6:1) to give the title compound (0.037 g, 32%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H27N6O3S: 467.1860, found: 467.1860;1H NMR (500 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.62 (d, 1H), 7.88 (d, 1H), 7.39 (d, 1H), 7.23 (d, 1H), 7.12 (dd, 1H), 5.31 (dd, 1H), 4.89 (d, 1H), 4.70 (d, 1H), 4.29 (d, 2H), 4.06 (t, 2H), 3.81-3.72 (m, 2H), 3.62-3.56 (m, 4H), 3.40 (dd, 1H), 3.36 (d, 1H), 3.32-3.26 (m, 1H), 2.41-2.30 (m, 4H). Example 123: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,5-dimethyl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-4-carboxamide (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (57 mg, 0.27 mmol) was added to a mixture of 6-(5,5-dimethyl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-4-carboxylic acid Intermediate 244 (70 mg, 0.23 mmol), HATU (130 mg, 0.34 mmol) and DIPEA (119 μL, 0.68 mmol) in DMF (2 mL) and the reaction mixture was stirred at rt overnight. EtOAc (10 mL) and NaHCO3(5 mL, aq) were added and the reaction mixture was stirred, and the phases were separated. The organic layer was washed with water and brine, and the combined aqueous phase was extracted with EtOAc. The combined organic layer was dried over MgSO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative SFC, PrepMethod SFC-A, (gradient: 27-32%) to give the title compound (39 mg, 37%); HRMS (ESI) m/z [M+H]+calcd for C24H25N6O2S: 461.1754 found: 461.1748;1H NMR (600 MHz, DMSO-d6) δ 9.09 (t, 1H), 8.84 (d, 1H), 8.37 (d, 1H), 8.19 (s, 1H), 8.03 (d, 1H), 7.96 (dd, 1H), 7.48 (d, 1H), 5.31 (d, 1H), 4.79 (dd, 2H), 4.30 (qd, 2H), 3.87 (s, 2H), 3.49-3.31 (m, 2H), 3.13 (s, 2H), 1.18-1.13 (m, 6H). Example 124: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-fluoropyridin-4-yl)quinoline-4-carboxamide (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (5 mg, 0.02 mmol) was added to a mixture of 6-(2-fluoropyridin-4-yl)quinoline-4-carboxylic acid Intermediate 245 (5 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol) and DIPEA (9.8 μL, 0.06 mmol) in DMF (0.2 mL), and the reaction mixture was stirred at rt overnight. DCM (5 mL) and NaHCO3(aq) were added, and the reaction mixture was stirred, filtered through a phase separator, and the filtrate was evaporated at reduced pressure. The crude product was purified by preparative SFC, PrepMethod SFC-A, (gradient: 27-32%) to give the title compound (2 mg, 29%); HRMS (ESI) m/z [M+H]+calcd for C21H17FN5O2S: 422.1082, found: 422.1082. Example 125: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-fluoropyridin-2-yl)quinoline-4-carboxamide (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (6 mg, 0.03 mmol) was added to a mixture of 6-(5-fluoropyridin-2-yl)quinoline-4-carboxylic acid Intermediate 246 (6 mg, 0.02 mmol), HATU (13 mg, 0.03 mmol) and DIPEA (12 μL, 0.07 mmol) in MeCN (0.2 mL) and EtOAc (0.2 mL) and the reaction mixture was stirred at rt overnight. EtOAc (3 mL) and NaHCO3(3 mL, aq) were added, and the reaction mixture was stirred, and the phases were separated. The organic layer was filtered through a phase separator, and the filtrate was evaporated at reduced pressure. The crude product was purified by preparative SFC, PrepMethod SFC-A, (gradient: 27-32%) to give the title compound (2.5 mg, 26%); HRMS (ESI) m/z [M+H]+calcd for C21H17FN5O2S: 422.1082, found: 422.1092. Example 126: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(pyridin-3-yl)quinoline-4-carboxamide Step a) 6-(Pyridin-3-yl)quinoline-4-carboxylic acid A mixture of 6-bromoquinoline-4-carboxylic acid (60 mg, 0.24 mmol), pyridin-3-ylboronic acid (32 mg, 0.26 mmol), Cs2CO3(194 mg, 0.60 mmol) and Pd(dtbpf)Cl2(16 mg, 0.02 mmol) in 1,4-dioxane (2 mL) and water (0.5 mL) was stirred at rt under an atmosphere of argon overnight. Water was added to the reaction mixture and the aqueous phase was washed with EtOAc. The water phase was concentrated under reduced pressure to give the title compound. Step b) (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(pyridin-3-yl)quinoline-4-carboxamide A mixture of crude 6-(pyridin-3-yl)quinoline-4-carboxylic acid, (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (60 mg, 0.29 mmol), HATU (109 mg, 0.29 mmol) and DIPEA (0.167 mL, 0.96 mmol) in DMF (2 mL) was stirred at rt overnight. The reaction mixture was diluted with DCM (15 mL) and washed with sat NaHCO3(8 mL, aq). The organic phase was filtered through a phase separator and the filtrate was evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod V, (gradient 0-50%), to give the title compound (28 mg, 29%); HRMS (ESI) m/z [M+H]+calcd for C21H18N5O2S: 404.1176 found: 404.1140;1H NMR (600 MHz, DMSO-d6) δ 9.20 (t, 1H), 9.09 (d, 1H), 9.02 (d, 1H), 8.82 (d, 1H), 8.65 (dd, 1H), 8.36-8.19 (m, 3H), 7.62 (d, 1H), 7.57 (dd, 1H), 5.37 (dd, 1H), 4.82 (dd, 2H), 4.37 (d, 2H), 3.44-3.36 (m, overlapping with solvent). Example 127: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(pyrimidin-5-yl)-quinoline-4-carboxamide Step a) 6-(Pyrimidin-5-yl)quinoline-4-carboxylic acid A mixture of 6-bromoquinoline-4-carboxylic acid (60 mg, 0.24 mmol), pyrimidin-5-ylboronic acid (32 mg, 0.26 mmol), Cs2CO3(194 mg, 0.60 mmol) and Pd(dtbpf)Cl2(16 mg, 0.02 mmol) in 1,4-dioxane (2 mL) and water (0.5 mL) was stirred at rt under an atmosphere of argon overnight. Water was added to the reaction mixture and the aqueous phase was washed with EtOAc. The water phase was acidified to pH 3 with aq HCl (2 M), washed with DCM, and concentrated at reduced pressure to give the title compound. Step b) (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(pyrimidin-5-yl)-quinoline-4-carboxamide A mixture of crude 6-(pyrimidin-5-yl)quinoline-4-carboxylic acid, (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (60 mg, 0.29 mmol), HATU (109 mg, 0.29 mmol) and DIPEA (1.04 mL, 5.97 mmol) in DMF (2 mL) was stirred at rt overnight. The reaction mixture was diluted with DCM (15 mL) and washed with sat NaHCO3(8 mL, aq). The organic phase was filtered through a phase separator, and the filtrate was evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod V, (gradient: 0-50%) to give the title compound (40 mg, 42%); HRMS (ESI) m/z [M+H]+calcd for C20H17N6O2S: 405.1128 found: 405.1136;1H NMR (600 MHz, DMSO-d6) δ 9.33 (s, 2H), 9.26 (s, 1H), 9.23 (t, 1H) 9.05 (d, 1H), 8.87 (d, 1H), 8.33-8.23 (m, 2H), 7.65 (d, 1H), 5.36 (dd, 1H), 4.82 (dd, 2H), 4.37 (dd, 2H), 3.44-3.36 (m, overlapping with solvent). Example 128: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-methylpyridin-3-yl)-quinoline-4-carboxamide Step a) 6-(2-Methylpyridin-3-yl)quinoline-4-carboxylic acid The title compound was prepared as described in Example 127 Step a) from 6-bromoquinoline-4-carboxylic acid (60 mg, 0.24 mmol) and (2-methylpyridin-3-yl)boronic acid (36 mg, 0.26 mmol) to give the title compound as a crude product. Step b) (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-methylpyridin-3-yl)-quinoline-4-carboxamide The title compound was prepared as described for Example 127 Step b) from (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (57 mg, 0.27 mmol) and crude 6-(2-methylpyridin-3-yl)quinoline-4-carboxylic acid to give the title compound (38 mg, 40%); HRMS (ESI) m/z [M+H]+calcd for C22H20N5O2S: 418.1332 found: 418.1350;1H NMR (600 MHz, DMSO-d6) δ 9.16 (t, 1H), 9.04 (d, 1H), 8.55 (dd, 1H), 8.37 (d, 1H), 8.19 (d, 1H), 7.90 (dd, 1H), 7.80 (d, 1H), 7.64 (d, 1H), 7.41 (dd, 1H), 5.31 (dd, 1H), 4.77 (dd, 2H), 4.31 (d, 2H), 3.40-3.35 (m, overlapping with solvent), 2.52 (s, 3H). Example 129: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-methyl-1H-pyrazol-4-yl)quinoline-4-carboxamide Step a) 6-(1-Methyl-1H-pyrazol-4-yl)quinoline-4-carboxylic acid The title compound was prepared as described in Example 127 step a) from 6-bromoquinoline-4-carboxylic acid (60 mg, 0.24 mmol) and (1-methyl-1H-pyrazol-4-yl)boronic acid (33 mg, 0.26 mmol) to give the title compound as a crude product. Step b) (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-methyl-1H-pyrazol-4-yl)quinoline-4-carboxamide The title compound was prepared as described for Example 127 Step b) from (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (59 mg, 0.28 mmol) and crude 6-(1-methyl-1H-pyrazol-4-yl)quinoline-4-carboxylic acid to give the title compound (39 mg, 41%); HRMS (ESI) m/z [M+H]+calcd for C20H19N6O2S: 407.1284 found: 407.1286;1H NMR (600 MHz, DMSO-d6) δ 9.14 (t, 1H), 8.88 (d, 1H), 8.74 (d, 1H), 8.37 (s, 1H), 8.11-8.03 (m, 3H), 7.50 (d, 1H), 5.42 (dd, 1H), 4.83 (dd, 2H), 4.37 (d, 2H), 3.92 (s, 3H), 3.51-3.37 (m, 2H). Example 130: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-((trifluoromethoxy)-methyl)azetidin-1-yl)quinoline-4-carboxamide DIPEA (415 μL, 2.38 mmol) was added to a stirred suspension of 6-(3-((trifluoromethoxy)methyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 248 (155 mg, 0.48 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (197 mg, 0.95 mmol), HOBt (193 mg, 1.43 mmol) and EDC (273 mg, 1.43 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 20° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(40 mL, aq) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (4×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 32-52%) to give the title compound (0.090 g, 39%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H21F3N5O3S: 480.1312 found: 480.1292;1H NMR (300 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.62 (d, 1H), 7.87 (d, 1H), 7.40 (d, 1H), 7.18-7.07 (m, 2H), 5.35-5.26 (m, 1H), 4.87 (d, 1H), 4.69 (d, 1H), 4.34 (d, 2H), 4.29 (d, 2H), 4.06 (t, 2H), 3.79-3.69 (m, 2H), 3.39-3.35 (m, overlapping with solvent), 3.21-3.04 (m, 2H). Example 131: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methyl-3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxamide DIPEA (404 μL, 2.31 mmol) was added to a stirred suspension of 6-(3-methyl-3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 250 (150 mg, 0.46 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (192 mg, 0.93 mmol), HOBt (187 mg, 1.39 mmol) and EDC (266 mg, 1.39 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 20° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure and the residue was dissolved in NaHCO3(40 mL, aq) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (4×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 33-55%) to give the title compound (0.080 g, 36%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H23F3N5O2S: 478.1518 found: 478.15241H NMR (300 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.61 (d, 1H), 7.86 (d, 1H), 7.39 (d, 1H), 7.18-7.07 (m, 2H), 5.29 (dd, 1H), 4.87 (d, 1H), 4.69 (d, 1H), 4.28 (d, 2H), 3.84 (d, 2H), 3.72 (d, 2H), 3.45-3.34 (m, overlapping with solvent), 2.72 (q, 2H), 1.46 (s, 3H). Example 132: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(trifluoromethoxy)-azetidin-1-yl)quinoline-4-carboxamide DIPEA (783 μL, 4.48 mmol) was added to a stirred suspension of 6-(3-(trifluoromethoxy)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 252 (280 mg, 0.90 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (372 mg, 1.79 mmol), HOBt (364 mg, 2.69 mmol) and EDC (516 mg, 2.69 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 25° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(50 mL, aq) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (4×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 31-51%) to give the title compound (0.27 g, 65%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H19F3N5O3S: 466.1156 found: 466.11521H NMR (300 MHz, DMSO-d6) δ 9.00 (t, 1H), 8.68 (d, 1H), 7.92 (d, 1H), 7.43 (d, 1H), 7.33 (d, 1H), 7.19 (dd, 1H), 5.45-5.29 (m, 2H), 4.89 (d, 1H), 4.71 (d, 1H), 4.40 (dd, 2H), 4.31 (d, 2H), 4.05 (dd, 2H), 3.46-3.34 (m, overlapping with solvent). Example 133: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoroethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide DIPEA (356 μL, 2.04 mmol) was added to a stirred suspension of 6-(3-(2,2-difluoroethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 254 (125 mg, 0.41 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (127 mg, 0.61 mmol), HOBt (165 mg, 1.22 mmol) and EDC (235 mg, 1.22 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 25° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(40 mL, aq) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (4×100 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 29-49%) to give the title compound (0.11 g, 59%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24F2N5O2S: 460.1614 found: 460.15821H NMR (300 MHz, DMSO-d6) δ 8.95 (brs, 1H), 8.64 (d, 1H), 7.89 (d, 1H), 7.42 (d, 1H), 7.26-7.00 (m, 2H), 6.25 (t, 1H), 5.45-5.27 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.31 (d, 2H), 3.84 (d, 2H), 3.69 (d, 2H), 3.40-3.34 (m, overlapping with solvent), 2.24 (td, 2H), 1.43 (s, 3H). Example 134: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropyl-3-methylazetidin-1-yl)quinoline-4-carboxamide DIPEA (247 μL, 1.42 mmol) was added to a stirred suspension of 6-(3-cyclopropyl-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 256 (80 mg, 0.28 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (118 mg, 0.57 mmol), HOBt (115 mg, 0.85 mmol) and EDC (163 mg, 0.85 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 20° C. and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure and the residue was dissolved in NaHCO3(40 mL, aq) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (4×50 mL). The combined organic layer was washed with water (3×25 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod I, (gradient: 55-65%) to give the title compound (0.050 g, 40%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O2S: 436.1802 found: 436.1806;1H NMR (400 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.62 (d, 1H), 7.88 (d, 1H), 7.40 (d, 1H), 7.18 (d, 1H), 7.09 (dd, 1H), 5.83-5.30 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.31 (d, 2H), 3.57 (s, 4H), 3.42-3.33 (m, overlapping with solvent), 1.33 (s, 3H), 1.10-0.98 (m, 1H), 0.49-0.39 (m, 2H), 0.35-0.20 (m, 2H). Example 135: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxamide DIPEA (284 μL, 1.63 mmol) was added to a stirred suspension of 6-(3-(difluoromethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 258 (95 mg, 0.33 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (135 mg, 0.65 mmol), HOBt (132 mg, 0.98 mmol) and EDC (187 mg, 0.98 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 20° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(40 mL, aq) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (4×75 mL). The combined organic layer was washed with water (3×25 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod A, (gradient: 32-62%) to give the title compound (0.083 g, 57%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22F2N5O2S: 446.1456 found: 446.1438;1H NMR (300 MHz, DMSO-d6) δ 8.98 (t, 1H), 8.65 (d, 1H), 7.90 (d, 1H), 7.42 (d, 1H), 7.26-7.10 (m, 2H), 6.29 (t, 1H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.30 (d, 2H), 4.01 (d, 2H), 3.73 (d, 2H), 3.45-3.35 (m, 2H), 1.41 (s, 3H). Example 136: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(difluoromethoxy)-azetidin-1-yl)quinoline-4-carboxamide DIPEA (564 μL, 3.23 mmol) was added to a stirred suspension of 6-(3-(difluoromethoxy)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 260 (190 mg, 0.65 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (201 mg, 0.97 mmol), HOBt (262 mg, 1.94 mmol) and EDC (371 mg, 1.94 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 30° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(40 mL, aq) and EtOAc (100 mL). The phases were separated and the aqueous layer was extracted with EtOAc (5×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 24-44%) to give the title compound (0.242 g, 84%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O3S: 448.1250 found: 448.1246;1H NMR (300 MHz, DMSO-d6) δ 8.98 (t, 1H), 8.66 (d, 1H), 7.91 (d, 1H), 7.43 (d, 1H), 7.27 (d, 1H), 7.17 (dd, 1H), 6.81 (t, 1H), 5.34 (dd, 1H), 5.20-5.07 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.46-4.16 (m, 4H), 3.93 (dd, 2H), 3.45-3.36 (m, overlapping with solvent). Example 137: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-methyl-azetidin-1-yl)quinoline-4-carboxamide DIPEA (372 μL, 2.13 mmol) was added to a stirred suspension of 6-(3-ethyl-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 262 (115 mg, 0.43 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (177 mg, 0.85 mmol), HOBt (172 mg, 1.28 mmol) and EDC (245 mg, 1.28 mmol) in MeCN (6 mL) and EtOAc (6 mL) at 20° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(30 mL, aq) and EtOAc (80 mL). The phases were separated and the aqueous layer was extracted with EtOAc (4×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod I, (gradient: 60-78%) to give the title compound (0.110 g, 61%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O2S: 424.1802 found: 424.1802;1H NMR (400 MHz, DMSO-d6) δ 9.02-8.90 (m, 1H), 8.61 (d, 1H), 7.87 (d, 1H), 7.39 (d, 1H), 7.17 (d, 1H), 7.10 (dd, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.30 (d, 2H), 3.70 (dd, 2H), 3.62 (d, 2H), 3.43-3.34 (m, overlapping with solvent), 1.63 (q, 2H), 1.27 (s, 3H), 0.90 (t, 3H). Example 138: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-fluoro-azetidin-1-yl)quinoline-4-carboxamide A solution of T3P (1.48 g, 2.33 mmol, 50% in EtOAc) in MeCN (8 mL) was added to a stirred solution of 6-(3-ethyl-3-fluoroazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 264 (160 mg, 0.58 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (121 mg, 0.87 mmol) and DIPEA (306 μL, 1.75 mmol) in EtOAc (8 mL) at 20° C., and the reaction mixture was stirred at 25° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (75 mL), and washed sequentially with water (25 mL) and sat brine (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 28-48%) to give the title compound (0.050 g, 20%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H23FN5O2S: 428.1550 found: 428.1550;1H NMR (400 MHz, DMSO-d6) δ 8.98 (t, 1H), 8.65 (d, 1H), 7.91 (d, 1H), 7.41 (d, 1H), 7.33 (d, 1H), 7.17 (dd, 1H), 5.34 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.30 (d, 2H), 4.15-3.97 (m, 4H), 3.45-3.34 (m, 2H), 2.05-1.80 (m, 2H), 0.98 (t, 3H). Example 139: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide A solution of T3P (1.35 g, 2.13 mmol, 50% in EtOAc) in EtOAc (8 mL) was added to a stirred solution of 6-(2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylic acid Intermediate 266 (150 mg, 0.53 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (110 mg, 0.53 mmol) and DIPEA (278 μL, 1.59 mmol) in MeCN (8 mL) at 25° C. and the reaction mixture was stirred at 25° C. for 2 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (75 mL), and washed sequentially with sat brine (25 mL) and water (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod 0, (gradient: 57-67%) to give the title compound (0.050 g, 22%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O2S: 436.1802, found: 436.1826;1H NMR (400 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.61 (d, 1H), 7.86 (d, 1H), 7.39 (d, 1H), 7.18 (d, 1H), 7.10 (dd, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.29 (d, 2H), 3.81 (s, 4H), 3.44-3.34 (m, overlapping with solvent), 1.89-1.71 (m, 4H), 1.69-1.50 (m, 4H). Example 140: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoropropyl)-azetidin-1-yl)quinoline-4-carboxamide A solution of T3P (831 mg, 1.31 mmol, 50% in EtOAc) in EtOAc (8 mL) was added to a stirred solution of 6-(3-(2,2-difluoropropyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 268 (100 mg, 0.33 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (68 mg, 0.49 mmol) and DIPEA (171 μL, 0.98 mmol) in MeCN (8 mL) at 25° C., and the reaction mixture was stirred at 25° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (100 mL), and washed sequentially with water (25 mL) and sat brine (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 29-49%) to give the title compound (0.050 g, 33%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24F2N5O2S: 460.1614, found: 460.1600;1H NMR (400 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.62 (d, 1H), 7.87 (d, 1H), 7.41 (d, 1H), 7.17-7.07 (m, 2H), 5.31 (dd, 1H), 4.88 (d, 1H), 4.71 (d, 1H), 4.29 (d, 2H), 4.16 (t, 2H), 3.66 (t, 2H), 3.44-3.34 (m, overlapping with solvent), 3.12-2.95 (m, 1H), 2.28 (td, 2H), 1.63 (t, 3H). Example 141: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,5-difluoro-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide A solution of T3P (639 mg, 2.01 mmol, 50% in EtOAc) in EtOAc (8 mL) was added to a stirred solution of 6-(5,5-difluoro-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylic acid Intermediate 270 (160 mg, 0.50 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (104 mg, 0.75 mmol) and DIPEA (263 μL, 1.51 mmol) in MeCN (8.0 mL) at 20° C., and the reaction mixture was stirred at 25° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (125 mL), and washed sequentially with water (75 mL) and sat brine (75 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 31-51%) to give the title compound (0.050 g, 21%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H24F2N5O2S: 472.1614, found: 472.1612;1H NMR (400 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.64 (d, 1H), 7.90 (d, 1H), 7.40 (d, 1H), 7.31 (d, 1H), 7.16 (dd, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.30 (d, 2H), 4.12-4.00 (m, 2H), 3.84 (d, 2H), 3.48-3.34 (m, overlapping with solvent), 2.22-2.02 (m, 4H), 1.83-1.65 (m, 2H). Example 142: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(3,3,3-trifluoropropyl)azetidin-1-yl)quinoline-4-carboxamide A solution of 6-(3-(3,3,3-trifluoropropyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 272 (110 mg, 0.34 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (106 mg, 0.51 mmol), EDC (98 mg, 0.51 mmol), HOBt (78 mg, 0.51 mmol) and DIPEA (178 μL, 1.02 mmol) in MeCN (4 mL) and EtOAc (4 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 28-38%) to give the title compound (0.110 g, 68%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H23F3N5O2S: 478.1518, found: 478.1520;1H NMR (300 MHz, CD3OD) δ 8.56 (d, 1H), 7.88 (d, 1H), 7.48 (d, 1H), 7.24 (s, 1H), 7.15 (dd, 1H), 5.33 (dd, 1H), 4.85-4.70 (m, overlapping with solvent), 4.38 (s, 2H), 4.25-4.10 (m, 2H), 3.70 (dd, 2H), 3.49-3.34 (m, 2H), 2.90-2.75 (m, 1H), 2.31-2.12 (m, 2H), 1.94 (q, 2H). Example 143: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(trifluoromethyl)azetidin-1-yl)quinoline-4-carboxamide A solution of 6-(3-fluoro-3-(trifluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 274 (100 mg, 0.32 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (99 mg, 0.48 mmol), EDC (92 mg, 0.48 mmol), HOBt (73 mg, 0.48 mmol) and DIPEA (167 μL, 0.95 mmol) in MeCN (6 mL) and EtOAc (6 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with EtOAc, and washed sequentially with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 31-41%) to give the title compound (110 mg, 74%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H18F4N5O2S: 468.1112, found: 468.1124;1H NMR (300 MHz, CD3OD) δ 8.66 (d, 1H), 7.97 (d, 1H), 7.56-7.49 (m, 2H), 7.26 (dd, 1H), 5.34 (dd, 1H), 4.85-4.66 (m, overlapping with solvent), 4.61-4.46 (m, 2H), 4.44-4.25 (m, 4H), 3.52-3.33 (m, 2H). Example 144: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2-difluoroethyl)-azetidin-1-yl)quinoline-4-carboxamide A solution of 6-(3-(2,2-difluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 276 (110 mg, 0.38 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (117 mg, 0.56 mmol), EDC (108 mg, 0.56 mmol), HOBt (76 mg, 0.56 mmol) and DIPEA (197 μL, 1.13 mmol) in MeCN (4 mL) and EtOAc (4 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with EtOAc, and washed sequentially with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 21-31%) to give the title compound (128 mg, 76%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22F2N5O2S: 446.1456, found: 446.1442;1H NMR (300 MHz, CD3OD) δ 8.57 (d, 1H), 7.88 (d, 1H), 7.49 (d, 1H), 7.30-7.10 (m, 2H), 5.98 (tt, 1H), 5.40-5.25 (m, 1H), 4.85-4.70 (m, 2H), 4.45-4.30 (m, 2H), 4.22 (t, 2H), 3.82-3.72 (m, 2H), 3.51-3.34 (m, 2H), 3.08-2.95 (m, 1H), 2.35-2.12 (m, 2H). Example 145: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-cyclopropylazetidin-1-yl)quinoline-4-carboxamide A solution of 6-(3-cyclopropylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 278 (110 mg, 0.41 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (128 mg, 0.61 mmol), EDC (118 mg, 0.61 mmol), HOBt (83 mg, 0.61 mmol) and DIPEA (215 μL, 1.23 mmol) in MeCN (6 mL) and EtOAc (6 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, the residue was diluted with EtOAc, and washed sequentially with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, gradient: 19-40%) to give the title compound (130 mg, 75%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O2S: 422.1646, found: 422.1638;1H NMR (300 MHz, CD3OD) δ 8.55 (d, 1H), 7.87 (d, 1H), 7.49 (d, 1H), 7.22-7.09 (m, 2H), 5.39-5.31 (m, 1H), 4.85-4.70 (m, 2H), 4.38 (d, 2H), 4.12 (t, 2H), 3.72 (dd, 2H), 3.45-3.34 (m, 2H), 2.48-2.35 (m, 1H), 1.18-0.99 (m, 1H), 0.59-0.46 (m, 2H), 0.28-0.17 (m, 2H). Example 146: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2-fluoroethyl)-azetidin-1-yl)quinoline-4-carboxamide A solution of 6-(3-(2-fluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 280 (100 mg, 0.36 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (114 mg, 0.55 mmol), EDC (105 mg, 0.55 mmol), HOBt (84 mg, 0.55 mmol) and DIPEA (191 μL, 1.09 mmol) in MeCN (8 mL) and EtOAc (8 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with EtOAc, and washed sequentially with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 18-28%) to give the title compound (120 mg, 77%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H23FN5O2S: 428.1550, found: 428.1558;1H NMR (300 MHz, CD3OD) δ 8.56 (d, 1H), 7.88 (d, 1H), 7.49 (d, 1H), 7.25-7.10 (m, 2H), 5.37-5.29 (m, 1H), 4.86-4.73 (m, 2H), 4.60 (t, 1H), 4.49-4.35 (m, 3H), 4.20 (t, 2H), 3.74 (t, 2H), 3.55-3.36 (m, 2H), 3.00-2.85 (m, 1H), 2.11 (q, 1H), 2.03 (q, 1H). Example 147: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(1,1-difluoroethyl)-azetidin-1-yl)quinoline-4-carboxamide A solution of 6-(3-(1,1-difluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 282 (100 mg, 0.34 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (107 mg, 0.51 mmol), EDC (98 mg, 0.51 mmol), HOBt (79 mg, 0.51 mmol) and DIPEA (179 μL, 1.03 mmol) in MeCN (8 mL) and EtOAc (8 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with EtOAc, and washed with water (3×10 mL). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was triturated with MeOH (20 mL), the precipitate was collected by filtration, and dried under vacuum to give the title compound (115 mg, 75%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22F2N5O2S: 446.1456, found: 446.1448;1H NMR (300 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.62 (d, 1H), 7.88 (d, 1H), 7.39 (d, 1H), 7.24 (d, 1H), 7.12 (dd, 1H), 5.35-5.25 (m, 1H), 4.88 (d, 1H), 4.69 (d, 1H), 4.28 (d, 2H), 4.15-4.00 (m, 2H), 3.98-3.80 (m, 2H), 3.41-3.33 (m, overlapping with solvent), 1.66 (t, 3H). Example 148: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-isopropylazetidin-1-yl)quinoline-4-carboxamide DIPEA (3.1 mL, 18 mmol) was added to a solution of 6-(3-isopropylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 284 (237 mg, 0.88 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (273 mg, 1.31 mmol), HOBt (1.18 g, 8.75 mmol) and EDC (1.68 mg, 8.75 mmol) in MeCN (6 mL) and EtOAc (6 mL) at 25° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with sat NaHCO3(200 mL, aq), and extracted with EtOAc (3×200 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, gradient: 22-38%) to give the title compound (183 mg, 49%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O2S: 424.1802, found: 424.1798;1H NMR (300 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.61 (d, 1H), 7.87 (d, 1H), 7.40 (d, 1H), 7.18 (d, 1H), 7.10 (dd, 1H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.30 (d, 2H), 4.10-3.98 (m, 2H), 3.70-3.52 (m, 2H), 3.48-3.38 (m, overlapping with solvent), 2.48-2.30 (m, 1H), 1.82-1.60 (m, 1H), 0.89 (d, 6H). Example 149: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methyl-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide DIPEA (4.2 mL, 24 mmol) was added to a solution of 6-(6-methyl-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid Intermediate 286 (337 mg, 1.19 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (372 mg, 1.79 mmol), HOBt (1.61 g, 11.9 mmol) and EDC (2.29 g, 11.9 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 25° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with sat NaHCO3(250 mL, aq), and extracted with EtOAc (3×250 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 27-39%) to give the title compound (81 mg, 15%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O2S: 436.1802, found: 436.1808;1H NMR (400 MHz, DMSO-d6) δ 8.92 (t, 1H), 8.61 (d, 1H), 7.86 (d, 1H), 7.39 (d, 1H), 7.16 (d, 1H), 7.07 (dd, 1H), 5.33 (dd, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.29 (d, 2H), 3.96 (s, 2H), 3.87 (s, 2H), 3.45-3.37 (m, overlapping with solvent), 2.41-2.19 (m, 3H), 1.86-1.70 (m, 2H), 1.05 (d, 3H). Example 150: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(trifluoromethyl)-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide DIPEA (2.39 mL, 13.7 mmol) was added to 6-(6-(trifluoromethyl)-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid Intermediate 288 (230 mg, 0.68 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (213 mg, 1.03 mmol), HOBt (924 mg, 6.84 mmol) and EDC (1.31 g, 6.84 mmol) in MeCN (6 mL) and EtOAc (6 mL) at 25° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with sat NaHCO3(300 mL, aq), and extracted with EtOAc (3×300 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 26-40%) to give the title compound (185 mg, 55%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C23H23F3N5O2S: 490.1518, found: 490.1510;1H NMR (300 MHz, DMSO-d6) δ 8.96 (t, 1H), 8.63 (d, 1H), 7.88 (d, 1H), 7.41 (d, 1H), 7.22-7.13 (m, 1H), 7.09 (dd, 1H), 5.34 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.31 (d, 2H), 4.00 (s, 2H), 3.94 (s, 2H), 3.55-3.34 (m, overlapping with solvent), 3.21-3.05 (m, 1H), 2.60-2.40 (m, overlapping with solvent), 2.34-2.24 (m, 2H). Example 151: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methoxy-3-methyl-azetidin-1-yl)quinoline-4-carboxamide TEA (3.13 mL, 22.5 mmol) was added to a solution of 6-(3-methoxy-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 290 (306 mg, 1.12 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (350 mg, 1.69 mmol), HOBt (1.52 g, 11.2 mmol) and EDC (2.15 g, 11.2 mmol) in EtOAc (6 mL) and MeCN (6 mL) at 25° C., and the reaction mixture was stirred at 25° C. for 16 h. The solvent was removed under reduced pressure, and the residue was diluted with sat NaHCO3(250 mL, aq), and extracted with EtOAc (3×250 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 13-23%) to give the title compound (126 mg, 26%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1596;1H NMR (300 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.63 (d, 1H), 7.89 (d, 1H), 7.41 (d, 1H), 7.25 (d, 1H), 7.14 (dd, 1H), 5.34 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.30 (d, 2H), 3.93-3.78 (m, 4H), 3.45-3.34 (m, overlapping with solvent), 3.22 (s, 3H), 1.51 (s, 3H). Example 152: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethoxy-3-methyl-azetidin-1-yl)quinoline-4-carboxamide DIPEA (3.98 mL, 22.8 mmol) was added to a solution of 6-(3-ethoxy-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 292 (326 mg, 1.14 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (355 mg, 1.71 mmol), HOBt (1.54 g, 11.4 mmol) and EDC (2.19 g, 11.4 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 25° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with sat NaHCO3(250 mL, aq), and extracted with EtOAc (3×250 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 13-28%) to give the title compound (251 mg, 49%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1754;1H NMR (400 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.63 (d, 1H), 7.88 (d, 1H), 7.40 (d, 1H), 7.24 (d, 1H), 7.13 (dd, 1H), 5.33 (dd, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.30 (d, 2H), 3.90-3.80 (m, 4H), 3.50-3.33 (m, overlapping with solvent), 1.51 (s, 3H), 1.13 (t, 3H). Example 153: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide DIPEA (620 μL, 3.55 mmol) was added to a stirred suspension of 6-(1-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylic acid Intermediate 294 (192 mg, 0.71 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (251 mg, 1.21 mmol), HOBt (288 mg, 2.13 mmol) and EDC (409 mg, 2.13 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 30° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in a mixture of NaHCO3(40 mL, aq) and EtOAc (100 mL). The aqueous layer was extracted with EtOAc (4×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 15-35%) to give the title compound (90 mg, 30%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438, found: 424.1448;1H NMR (400 MHz, DMSO-d6) δ 9.10-8.90 (m, 1H), 8.65 (d, 1H), 7.89 (d, 1H), 7.42 (d, 1H), 7.24 (d, 1H), 7.13 (dd, 1H), 5.45-5.33 (m, 1H), 4.91 (d, 1H), 4.73 (d, 1H), 4.47 (t, 2H), 4.31 (d, 2H), 4.24 (d, 2H), 4.03 (d, 2H), 3.60-3.37 (m, 2H), 2.90 (t, 2H). Example 154: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-ethyl-3-hydroxy-azetidin-1-yl)quinoline-4-carboxamide T3P (1.40 g, 2.20 mmol, 50% in EtOAc) was added to a solution of 6-(3-ethyl-3-hydroxyazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 296 (150 mg, 0.55 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (114 mg, 0.55 mmol) and DIPEA (289 μL, 1.65 mmol) in MeCN (8 mL) and EtOAc (8 mL) at 25° C., and the reaction mixture was stirred at 25° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (75 mL), and washed sequentially with sat brine (15 mL) and water (15 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 15-35%) to give the title compound (50 mg, 21%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1586;1H NMR (400 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.61 (d, 1H), 7.87 (d, 1H), 7.39 (d, 1H), 7.17 (d, 1H), 7.12 (dd, 1H), 5.33 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.29 (d, 2H), 4.00-3.85 (m, 2H), 3.71 (d, 2H), 3.50-3.34 (m, overlapping with solvent), 1.74 (q, 2H), 0.94 (t, 3H). Example 155: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-fluoro-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxamide DIPEA (3.36 mL, 19.2 mmol) was added to a solution of 6-(6-fluoro-2-azaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid Intermediate 298 (275 mg, 0.96 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (299 mg, 1.44 mmol), HOBt (1.30 g, 9.61 mmol) and EDC (1.84 g, 9.61 mmol) in MeCN (6 mL) and EtOAc (6 mL) at 25° C., and the reaction mixture was stirred at 50° C. for 2 h. The reaction mixture was concentrated under reduced pressure, diluted with sat NaHCO3(250 mL, aq), and extracted with EtOAc (3×250 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 16-30%) to give the title compound (142 mg, 34%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C22H23FN5O2S: 440.1550, found: 440.1556;1H NMR (300 MHz, DMSO-d6) δ 8.94 (t, 1H), 8.63 (d, 1H), 7.88 (d, 1H), 7.41 (d, 1H), 7.23-7.02 (m, 2H), 5.34 (dd, 1H), 5.20-4.80 (m, 2H), 4.75-4.59 (m, 1H), 4.30 (d, 2H), 3.96 (d, 4H), 3.46-3.39 (m, 2H), 2.73-2.58 (m, 2H), 2.45-2.25 (m, 2H). Example 156: 6-((1RS,5RS)-6-Azabicyclo[3.2.0]heptan-6-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide A solution of 6-(6-azabicyclo[3.2.0]heptan-6-yl)quinoline-4-carboxylic acid Intermediate 300 (100 mg, 0.37 mmol) in DMF (5 mL) was added to a mixture of (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (155 mg, 0.75 mmol), HATU (283 mg, 0.75 mmol) and DIPEA (193 mg, 1.49 mmol), and the reaction mixture was stirred at 25° C. for 6 h. The solvent was removed under reduced pressure, and the crude product was purified by preparative TLC (DCM:MeOH, 18:1), followed by preparative HPLC, PrepMethod T, (gradient: 30-50%) to give the title compound (86 mg, 55%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O2S: 422.1646, found: 422.1640;1H NMR (300 MHz, DMSO-d6) δ 8.91 (t, 1H), 8.54 (d, 1H), 7.82 (d, 1H), 7.35 (d, 1H), 7.12-6.99 (m, 2H), 5.40-5.25 (m, 1H), 4.87 (d, 1H), 4.77-4.47 (m, 2H), 4.27 (d, 2H), 3.95 (t, 1H), 3.50 (dd, 1H), 3.41-3.33 (m, overlapping with solvent), 3.15-2.95 (m, 1H), 2.14-1.97 (m, 1H), 1.91-1.68 (m, 3H), 1.62-1.33 (m, 2H). Example 157: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxamide DIPEA (565 μL, 3.23 mmol) was added to a stirred suspension of 6-(3-fluoro-3-(fluoromethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 303 (90 mg, 0.32 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (101 mg, 0.49 mmol), HOBt (248 mg, 1.62 mmol) and EDC (310 mg, 1.62 mmol) in MeCN (5 mL) and EtOAc (5 mL), and the reaction mixture was stirred at 18° C. for 15 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(30 mL, aq) and EtOAc (80 mL). The aqueous layer was extracted with EtOAc (5×75 mL) and the combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 22-42%) to give the title compound (35 mg, 25%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O2S: 432.1300, found: 432.1294;1H NMR (300 MHz, DMSO-d6) δ 9.10-8.90 (m, 1H), 8.68 (d, 1H), 7.94 (d, 1H), 7.44 (d, 1H), 7.35 (d, 1H), 7.22 (dd, 1H), 5.40-5.24 (m, 1H), 5.03-4.68 (m, 4H), 4.35-3.98 (m, 6H), 3.41 (m, overlapping with solvent). Example 158: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(2,2,2-trifluoroethyl)-azetidin-1-yl)quinoline-4-carboxamide DIPEA (1.80 mL, 10.3 mmol) was added to a stirred suspension of 6-(3-(2,2,2-trifluoroethyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 305 (320 mg, 1.03 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (321 mg, 1.55 mmol), HOBt (790 mg, 5.16 mmol) and EDC (989 mg, 5.16 mmol) in MeCN (8 mL) and EtOAc (8 mL) at 28° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure and the residue was dissolved in NaHCO3(60 mL, aq) and EtOAc (100 mL). The aqueous layer was extracted with EtOAc (6×100 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 32-45%) to give the title compound (250 mg, 52%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H21F3N5O2S: 464.1362, found: 464.1360;1H NMR (300 MHz, DMSO-d6) δ 8.94 (t, 1H), 8.62 (d, 1H), 7.87 (d, 1H), 7.40 (d, 1H), 7.18-7.07 (m, 2H), 5.30 (dd, 1H), 4.87 (d, 1H), 4.70 (d, 1H), 4.28 (d, 2H), 4.15 (t, 2H), 3.71 (t, 2H), 3.43-3.33 (m, 2H), 3.15-2.95 (m, 1H), 2.80-2.58 (m, 2H). Example 159: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3,3-difluoro-2-methylazetidin-1-yl)quinoline-4-carboxamide DIPEA (2.36 mL, 13.5 mmol) was added to a mixture of 6-(3,3-difluoro-2-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 307 (188 mg, 0.68 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (210 mg, 1.01 mmol), HOBt (1.04 g, 6.76 mmol) and EDC (1.30 g, 6.76 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 20° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the reaction mixture was diluted with sat NaHCO3(250 mL, aq), and extracted with EtOAc (4×250 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 25-50%) to give the title compound (169 mg, 58%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O2S: 432.1300, found: 432.1306;1H NMR (300 MHz, DMSO-d6) δ 9.04 (brs, 1H), 8.72 (d, 1H), 7.96 (d, 1H), 7.56-7.50 (m, 1H), 7.45 (d, 1H), 7.27 (dd, 1H), 5.45-5.25 (m, 1H), 4.91 (d, 1H), 4.78-4.49 (m, 3H), 4.37-4.15 (m, 3H), 3.42-3.32 (m, overlapping with solvent), 1.52 (d, 3H). Example 160: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6,6-difluoro-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxamide DIPEA (957 μL, 5.48 mmol) was added to a stirred suspension of 6-(6,6-difluoro-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylic acid Intermediate 309 (153 mg, 0.27 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (114 mg, 0.55 mmol), HOBt (210 mg, 1.37 mmol) and EDC (263 mg, 1.37 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 12° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(30 mL, aq) and EtOAc (80 mL). The aqueous layer was extracted with EtOAc (5×75 mL). The combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 10-40%) to give the title compound (150 mg, 86%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H20F2N5O2S: 444.1300, found: 444.1282;1H NMR (300 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.59 (d, 1H), 7.88 (d, 1H), 7.37 (d, 1H), 7.33-7.20 (m, 2H), 5.36-5.22 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.31-4.24 (m, 2H), 3.85-3.58 (m, 4H), 3.42-3.34 (m, overlapping with solvent), 2.80-2.60 (m, 2H). Example 161: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-methoxy-pyrrolidin-1-yl)quinoline-4-carboxamide HATU (348 mg, 0.88 mmol), DIPEA (411 μL, 2.35 mmol) were added to a stirred solution of (R)-6-(3-methoxypyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 311 (160 mg, 0.59 mmol) and), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (122 mg, 0.59 mmol), in DMF (5 mL), and the reaction mixture was stirred at 25° C. for 2 h. The solvent was removed under reduced pressure, and the crude product was purified by preparative TLC (DCM:MeOH, 18:1), followed by preparative HPLC, PrepMethod F, (gradient: 1-35%) to give the title compound (108 mg, 42%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1584;1H NMR (300 MHz, DMSO-d6) δ 8.92 (t, 1H), 8.55 (d, 1H), 7.86 (d, 1H), 7.44-7.19 (m, 3H), 5.30 (dd, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.38-4.23 (m, 2H), 4.18-4.01 (m, 1H), 3.60-3.18 (m, overlapping with solvent), 2.20-2.00 (m, 2H). Example 162: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-oxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxamide A solution of 6-(3-oxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylic acid Intermediate 313 (100 mg, 0.31 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (95 mg, 0.46 mmol), EDC (88 mg, 0.46 mmol), HOBt (70 mg, 0.46 mmol) and DIPEA (161 μL, 0.92 mmol) in MeCN (6 mL) and EtOAc (6 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 30-40%) to give the title compound (75 mg, 51%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C25H30N5O3S: 480.2064, found: 480.2057;1H NMR (300 MHz, CD3OD) δ 8.62 (d, 1H), 7.90 (d, Hz, 1H), 7.78-7.62 (m, 2H), 7.48 (d, 1H), 5.35-5.27 (m, 1H), 4.90-4.80 (m, overlapping with solvent), 4.77 (d, 1H), 4.38 (s, 2H), 3.71 (t, 4H), 3.54-3.35 (m, 6H), 1.75 (t, 4H), 1.58 (t, 4H). Example 163: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,8-dioxa-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxamide (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (290 mg, 1.40 mmol), was added to a solution of 6-(5,8-dioxa-2-azaspiro[3.4]octan-2-yl)quinoline-4-carboxylic acid Intermediate 315 (200 mg, 0.70 mmol), HATU (531 mg, 1.40 mmol) and DIPEA (361 mg, 2.79 mmol) in DMF (5 mL), and the reaction mixture was stirred at 25° C. for 4 h. The solvent was evaporated at reduced pressure, and the residue was purified by preparative TLC (DCM:MeOH, 18:1), followed by preparative HPLC, PrepMethod B, (gradient: 18-38%) to give the title compound (50 mg, 16%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O4S: 440.1388, found: 440.1394;1H NMR (300 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.63 (d, 1H), 7.89 (d, 1H), 7.39 (d, 1H), 7.30 (d, 1H), 7.14 (dd, 1H), 5.32 (dd, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.28 (d, 2H), 4.09 (s, 4H), 3.95 (s, 4H), 3.43-3.33 (m, 2H). Example 164: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5-azaspiro[2.3]hexan-5-yl)quinoline-4-carboxamide DIPEA (3.98 mL, 22.8 mmol) was added to a mixture of 6-(5-azaspiro[2.3]hexan-5-yl)quinoline-4-carboxylic acid Intermediate 317 (290 mg, 1.14 mmol), (R)-3-glycyl-thiazolidine-4-carbonitrile hydrochloride Intermediate 4 (474 mg, 2.28 mmol), HOBt (1746 mg, 11.40 mmol) and EDC (2186 mg, 11.40 mmol) in MeCN (3 mL) and EtOAc (3 mL) at 15° C., and the reaction mixture was stirred at 15° C. overnight. The solvent was removed under reduced pressure. The residue was diluted with NaHCO3(100 mL, aq), and extracted with EtOAc (5×100 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 20-35%) to give the title compound (92 mg, 20%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O2S: 408.1488, found: 408.1500;1H NMR (300 MHz, DMSO-d6) δ 9.05-8.90 (m, 1H), 8.63 (d, 1H), 7.89 (d, 1H), 7.41 (d, 1H), 7.25-7.16 (m, 1H), 7.12 (dd, 1H), 5.32 (dd, 1H), 4.88 (d, 2H), 4.70 (d, 1H), 4.29 (d, 2H), 4.06 (s, 4H), 3.41-3.35 (m, overlapping with solvent), 1.27-1.21 (m, 1H), 0.69 (s, 4H). Example 165: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-hydroxy-3-methyl-azetidin-1-yl)quinoline-4-carboxamide DIPEA (575 μL, 3.29 mmol) was added to a stirred suspension of 6-(3-hydroxy-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 319 (85 mg, 0.33 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (137 mg, 0.66 mmol), HOBt (252 mg, 1.65 mmol) and EDC (315 mg, 1.65 mmol) in MeCN (3 mL) and EtOAc (3 mL) at 15° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was dissolved in NaHCO3(20 mL, aq) and EtOAc (60 mL). The aqueous layer was extracted with EtOAc (5×75 mL) and the combined organic layer was washed with water (3×25 mL). The combined aqueous layer was extracted with EtOAc (3×20 mL). The combined organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 15-35%) to give the title compound (20 mg, 15%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H22N5O3S: 412.1438, found: 412.1428;1H NMR (300 MHz, DMSO-d6) δ 8.95 (t, 1H), 8.61 (d, 1H), 7.87 (d, 1H), 7.39 (d, 1H), 7.22-7.06 (m, 2H), 5.60 (s, 1H), 5.39-5.24 (m, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.29 (d, 2H), 3.89 (d, 2H), 3.76 (d, 2H), 3.48-3.33 (d, overlapping with solvent), 1.48 (s, 3H). Example 166: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide DIPEA (746 μL, 4.27 mmol) was added to a stirred suspension of 6-(2-oxa-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylic acid Intermediate 321 (231 mg, 0.85 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (266 mg, 1.28 mmol), HOBt (346 mg, 2.56 mmol) and EDC (492 mg, 2.56 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 25° C., and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The residue was dissolved in NaHCO3(40 mL, aq) and EtOAc (100 mL). The aqueous layer was extracted with EtOAc (6×75 mL) and the combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 20-45%) to give the title compound (80 mg, 22%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438, found: 424.1428;1H NMR (300 MHz, DMSO-d6) δ 8.94 (t, 1H), 8.63 (d, 1H), 7.88 (d, 1H), 7.40 (d, 1H), 7.20 (d, 1H), 7.11 (dd, 1H), 5.36 (dd, 1H), 4.89 (d, 1H), 4.84-4.64 (m, 5H), 4.30 (d, 2H), 4.13 (s, 4H), 3.41-3.36 (m, overlapping with solvent). Example 167: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide A solution of (R)-6-(3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 323 (150 mg, 0.55 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (149 mg, 0.72 mmol), EDC (211 mg, 1.10 mmol), HOBt (169 mg, 1.10 mmol) and DIPEA (481 μL, 2.75 mmol) in EtOAc (5 mL) and MeCN (5 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 9-19%) to give the title compound (120 mg, 50%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1604;1H NMR (300 MHz, CD3OD) δ 8.49 (d, 1H), 7.87 (d, 1H), 7.46 (d, 1H), 7.31 (dd, 1H), 7.24 (d, 1H), 5.44-5.24 (m, 1H), 4.87-4.67 (m, overlapping with solvent), 4.38 (d, 2H), 3.72-3.51 (m, 2H), 3.49-3.33 (m, 4H), 2.12-1.97 (m, 2H), 1.48 (s, 3H). Example 168: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide A solution of (S)-6-(3-hydroxy-3-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 325 (150 mg, 0.55 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (149 mg, 0.72 mmol), EDC (211 mg, 1.10 mmol), HOBt (169 mg, 1.10 mmol) and DIPEA (481 μL, 2.75 mmol) in EtOAc (5 mL) and MeCN (5 mL) was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure, and the residue was diluted with EtOAc, and washed with water. The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient (9-20%), to give the title compound (125 mg, 52%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1581;1H NMR (300 MHz, CD3OD) δ 8.49 (d, 1H), 7.87 (d, 1H), 7.46 (d, 1H), 7.31 (dd, 1H), 7.24 (d, 1H), 5.44-5.24 (m, 1H), 4.87-4.67 (m, overlapping with solvent), 4.38 (d, 2H), 3.72-3.51 (m, 2H), 3.49-3.33 (m, 4H), 2.12-1.97 (m, 2H), 1.48 (s, 3H). Example 169: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-(difluoromethyl)-pyridin-3-yl)quinoline-4-carboxamide (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (104 mg, 0.50 mmol) and DIPEA (0.13 mL, 0.75 mmol) were added to a suspension of 6-(6-(difluoromethyl)pyridin-3-yl)quinoline-4-carboxylic acid Intermediate 326 (75 mg, 0.25 mmol), HOBT (46 mg, 0.30 mmol) and EDC (0.072 g, 0.37 mmol) in EtOAc (1.18 mL) and MeCN (1.18 mL), and the reaction mixture was stirred at rt overnight. The reaction mixture was diluted with EtOAc and washed sequentially with sat NaHCO3(aq) and brine. The organic phase was dried using a phase separator, filtered and evaporated at reduced pressure. The crude product was purified by preparative SFC, PrepMethod SFC-G, (gradient: 20-25%), to give the title compound (17 mg, 15%); HRMS (ESI) m/z [M+H]+calcd for C22H18F2N5O2S: 454.1144, found: 454.1140;1H NMR (600 MHz, DMSO-d6) δ 9.29-9.17 (m, 2H), 9.05 (d, 1H), 8.92 (d, 1H), 8.56 (dd, 1H), 8.33 (dd, 1H), 8.25 (d, 1H), 7.88 (d, 1H), 7.65 (d, 1H), 7.05 (t, 1H), 5.39 (dd, 1H), 4.91 (d, 1H), 4.74 (d, 1H), 4.38 (t, 2H). Example 170: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-cyclopropyl-1H-pyrazol-4-yl)quinoline-4-carboxamide DIPEA (1.42 mL, 8.13 mmol) was added to a suspension of 6-(1-cyclopropyl-1H-pyrazol-4-yl)quinoline-4-carboxylic acid Intermediate 327 (0.568 g, 2.03 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (0.422 g, 2.03 mmol) and HATU (0.773 g, 2.03 mmol) in DCM (16 mL) and MeCN (4 mL), and the reaction mixture was stirred at rt overnight. The reaction mixture was diluted with EtOAc, and washed sequentially with sat NaHCO3(aq) and brine. The organic phase was dried, filtered and evaporated at reduced pressure, and the crude product was purified by preparative SFC, PrepMethod SFC-H, (gradient: 25-30%) to give the title compound (220 mg, 25%); HRMS (ESI) m/z [M+H]+calcd for C22H21N6O2S: 433.1442, found: 433.1454;1H NMR (600 MHz, DMSO-d6) δ 9.10 (t, 1H), 8.95 (d, 1H), 8.53 (s, 1H), 8.36 (d, 1H), 8.27 (d, 1H), 8.13 (d, 1H), 7.98 (dd, 1H), 7.48 (d, 1H), 5.38 (dd, 1H), 4.90 (d, 1H), 4.73 (d, 1H), 4.33 (d, 2H), 3.79 (tt, 1H), 3.45-3.36 (m, overlapping with solvent), 1.15-1.11 (m, 2H), 1.05-0.99 (m, 2H). Example 171: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,3-dimethyl-1H-pyrazol-4-yl)quinoline-4-carboxamide HATU (267 mg, 0.70 mmol) and DIPEA (0.294 mL, 1.68 mmol) were added to a suspension of 6-(1,3-dimethyl-1H-pyrazol-4-yl)quinoline-4-carboxylic acid Intermediate 328 (150 mg, 0.56 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (117 mg, 0.56 mmol) in EtOAc (3 mL) and MeCN (3 mL), and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated under reduced pressure and the crude product was purified by preparative HPLC, PrepMethod V, (gradient: 0-50%), to give the title compound (90 mg, 38%); HRMS (ESI) m/z [M+H]+calcd for C21H21N6O2S: 421.1442, found: 421.1456;1H NMR (600 MHz, DMSO-d6) δ 9.12 (t, 1H), 8.92 (d, 1H), 8.45 (d, 1H), 8.14-8.06 (m, 2H), 7.94 (dd, 1H), 7.55 (d, 1H), 5.36 (dd, 1H), 4.91 (d, 1H), 4.73 (d, 1H), 4.34 (qd, 2H), 3.84 (s, 3H), 3.45-3.36 (m, overlapping with solvent), 2.40 (s, 3H). Example 172: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,5-dimethyl-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxamide HATU (54 mg, 0.14 mmol) and DIPEA (60 μL, 0.34 mmol) were added to a suspension of 6-(3,5-dimethyl-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxylic acid Intermediate 329 (40 mg, 0.11 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (24 mg, 0.11 mmol) in EtOAc (0.75 mL) and MeCN (0.75 mL), and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated under reduced pressure, and the residue was purified by preparative HPLC, PrepMethod F, (gradient: 5-95%), to give the title compound (15 mg, 26%); HRMS (ESI) m/z [M+H]+calcd for C26H29N6O3S: 505.2016, found: 505.2020;1H NMR (600 MHz, DMSO-d6) δ 9.12 (t, 1H), 8.96 (d, 1H), 8.27 (d, 1H), 8.11 (d, 1H), 7.78 (dd, 1H), 7.56 (d, 1H), 5.31 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.51-4.26 (m, 3H), 3.99 (dd, 2H), 3.49 (tt, 2H), 3.44-3.39 (m, overlapping with solvent), 2.35 (s, 3H), 2.23 (s, 3H), 2.08 (qt, 2H), 1.83 (tdd, 2H). Example 173: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxamide HATU (35 mg, 0.09 mmol) and DIPEA (0.039 mL, 0.22 mmol) were added to a suspension of 6-(1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-4-yl)quinoline-4-carboxylic acid Intermediate 330 (24 mg, 0.07 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (15 mg, 0.07 mmol) in EtOAc (0.75 mL) and MeCN (0.75 mL), and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated under reduced pressure and the residue was purified by preparative SFC, PrepMethod SFC-G, (gradient: 0-50%) to give the title compound (11 mg, 31%); HRMS (ESI) m/z [M+H]+calcd for C24H25N6O3S: 477.1704, found: 477.1706;1H NMR (600 MHz, DMSO-d6) δ 9.17 (t, 1H), 8.88 (d, 1H), 8.77 (d, 1H), 8.53 (s, 1H), 8.15-8.10 (m, 2H), 8.06 (d, 1H), 7.50 (d, 1H), 5.44 (dd, 1H), 4.94 (d, 1H), 4.75 (d, 1H), 4.52-4.44 (m, 1H), 4.38 (d, 2H), 4.05-3.95 (m, 2H), 3.54-3.39 (m, overlapping with solvent), 2.12-1.94 (m, 4H). Example 174: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-methyl-1H-pyrazol-5-yl)quinoline-4-carboxamide HATU (52 mg, 0.14 mmol) and DIPEA (0.058 mL, 0.33 mmol) were added to a suspension of 6-(1-methyl-1H-pyrazol-5-yl)quinoline-4-carboxylic acid Intermediate 331 (28 mg, 0.11 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (23 mg, 0.11 mmol) in EtOAc (0.7 mL) and MeCN (0.7 mL), and the reaction mixture was stirred at rt for 36 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by preparative SFC, PrepMethod SFC-H, (gradient: 22-27%) to give the title compound (16 mg, 36%); HRMS (ESI) m/z [M+H]+calcd for C20H19N6O2S: 407.1284, found: 407.1278;1H NMR (600 MHz, DMSO-d6) δ 9.21 (t, 1H), 9.04 (d, 1H), 8.54 (brs, 1H), 8.20 (d, 1H), 8.00 (d, 1H), 7.64 (d, 1H), 7.54 (brs, 1H), 6.61 (brs, 1H), 5.36 (d, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.34 (qd, 2H), 3.99 (s, 3H), 3.43-3.35 (m, overlapping with solvent). Example 175: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(5,6,7,8-tetrahydroimidazo[1,2-a]pyridin-3-yl)quinoline-4-carboxamide HATU (40 mg, 0.11 mmol) and DIPEA (0.045 mL, 0.26 mmol) were added to a suspension of 6-(5,6,7,8-tetrahydroimidazo[1,2-a]pyridin-3-yl)quinoline-4-carboxylic acid Intermediate 333 (25 mg, 0.09 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (18 mg, 0.09 mmol) in EtOAc (0.5 mL) and MeCN (0.5 mL), and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated under reduced pressure, and the residue was purified by preparative SFC, PrepMethod SFC-H, (gradient: 27-32%), followed by preparative HPLC, PrepMethod N, (gradient: 5-45%) to give the title compound (5 mg, 13%): HRMS (ESI) m/z [M+H]+calcd for C23H23N6O2S: 447.1598, found: 447.1619;1H NMR (500 MHz, CD3OD) δ 9.00 (d, 1H), 8.65 (s, 1H), 8.26 (s, 1H), 8.21 (d, 1H), 7.99 (d, 1H), 7.70 (d, 1H), 7.55 (s, 1H), 5.34-5.29 (m, 1H), 4.80-4.74 (m, overlapping with solvent), 4.51-4.21 (m, 4H), 3.52-3.33 (m, 2H), 3.07 (d, 2H), 2.18-2.02 (m, 4H). Example 176: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-((RS)-tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)quinoline-4-carboxamide Step a) rac-(R)-6-(1-(Tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)quinoline-4-carboxylic acid A mixture of 1-(tetrahydro-2H-pyran-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (77 mg, 0.28 mmol), 6-bromoquinoline-4-carboxylic acid (70 mg, 0.28 mmol), Cs2CO3(181 mg, 0.56 mmol) and Pd(dtbpf)C12(18 mg, 0.03 mmol) in 1,4-dioxane (1.5 mL) and water (0.37 mL) was stirred overnight. The reaction mixture was concentrated under reduced pressure to give the title compound (90 mg); MS (ESI) m/z [M+H]+324. Step b) N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)quinoline-4-carboxamide HATU (132 mg, 0.35 mmol) and DIPEA (0.146 mL, 0.84 mmol) were added to a suspension of crude 6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)quinoline-4-carboxylic acid (90 mg) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (58 mg, 0.28 mmol) in EtOAc (1 mL) and MeCN (1 mL), and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated under reduced pressure and the crude compound was purified by preparative SFC, PrepMethod SFC-C, (gradient: 27-32%), to give the title compound (74 mg, 56%); HRMS (ESI) m/z [M+H]+calcd for C24H25N6O3S: 477.1704, found: 477.1722;1H NMR (600 MHz, DMSO-d6) δ 9.17 (t, 1H), 9.06 (d, 1H), 8.49 (s, 1H), 8.22 (d, 1H), 8.00-7.95 (m, 1H), 7.68-7.63 (m, 2H), 6.64 (dd, 1H), 5.35-5.32 (m, 1H), 4.89 (dd, 1H), 4.72 (d, 1H), 4.40-4.27 (m, 2H), 4.04-3.97 (m, 1H), 3.58 (dt, 1H), 3.47-3.36 (m, overlapping with solvent), 2.48-2.37 (m, 1H), 1.94 (s, 1H), 1.85 (t, 1H), 1.63-1.47 (m, 3H). Example 177: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methylpyridin-3-yl)-quinoline-4-carboxamide Step a) 6-(6-Methylpyridin-3-yl)quinoline-4-carboxylic acid A mixture of 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (47 mg, 0.21 mmol), 6-bromoquinoline-4-carboxylic acid (54 mg, 0.21 mmol), Cs2CO3(209 mg, 0.64 mmol) and Pd(dtbpf)Cl2(21 mg, 0.03 mmol) in 1,4-dioxane (1 mL) and water (0.25 mL) was stirred overnight. The reaction mixture was concentrated under reduced pressure to give the title compound (56 mg) as a crude; MS (ESI) m/z [M+H]+263. Step b) (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methylpyridin-3-yl)-quinoline-4-carboxamide HATU (101 mg, 0.26 mmol) and DIPEA (0.111 mL, 0.64 mmol) were added to a suspension of crude 6-(6-methylpyridin-3-yl)quinoline-4-carboxylic acid (56 mg) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (44 mg, 0. mmol) in EtOAc (1 mL) and MeCN (1 mL) and the solution was stirred at rt overnight. The reaction mixture was concentrated under reduced pressure and the crude product was purified by preparative HPLC, PrepMethod F, (gradient: 0-30%), to give the title compound (30 mg, 34%); HRMS (ESI) m/z [M+H]+calcd for C22H20N5O2S: 418.1332, found: 418.1328;1H NMR (600 MHz, DMSO-d6) δ 9.20 (t, 1H), 9.01 (d, 1H), 8.96 (d, 1H), 8.78 (d, 1H), 8.26-8.17 (m, 3H), 7.62 (d, 1H), 7.42 (d, 1H), 5.38 (dd, 1H), 4.91 (d, 1H), 4.74 (d, 1H), 4.37 (d, 2H), 3.49-3.38 (m, overlapping with solvent), 2.55 (s, 3H). Example 178: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methoxypyridin-3-yl)quinoline-4-carboxamide Step a) 6-(6-Methoxypyridin-3-yl)quinoline-4-carboxylic acid A mixture of 2-methoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (49 mg, 0. mmol), 6-bromoquinoline-4-carboxylic acid (53 mg, 0.21 mmol), Cs2CO3(206 mg, 0.63 mmol) and Pd(dtbpf)Cl2(21 mg, 0.03 mmol) in 1,4-dioxane (2 mL) and water (0.5 mL) was stirred overnight. The reaction mixture was evaporated at reduced pressure to give the crude title compound (58 mg); MS (ESI) m/z [M+H]+281. Step b) (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(6-methoxypyridin-3-yl)quinoline-4-carboxamide HATU (98 mg, 0.26 mmol) and DIPEA (0.108 mL, 0.62 mmol) were added to a suspension of crude 6-(6-methoxypyridin-3-yl)quinoline-4-carboxylic acid (58 mg) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (43 mg, 0.21 mmol) in EtOAc (1 mL) and MeCN (1 mL), and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated under reduced pressure, and the crude product was purified by preparative SFC, PrepMethod SFC-C, (gradient: 25-30%), followed by PrepMethod SFC-E, (gradient: 20-25%), to give the title compound (23 mg, 25%); HRMS (ESI) m/z [M+H]+calcd for C22H20N5O3S: 434.1282, found: 434.1268;1H NMR (600 MHz, DMSO-d6) δ 9.18 (t, 1H), 8.99 (d, 1H), 8.76 (d, 1H), 8.69 (d, 1H), 8.30 (dd, 1H), 8.26-8.15 (m, 2H), 7.60 (d, 1H), 7.00 (d, 1H), 5.38 (dd, 1H), 4.91 (d, 1H), 4.74 (d, 1H), 4.41-4.33 (m, 2H), 3.94 (s, 3H), 3.48-3.39 (m, overlapping with solvent). Example 179: (R)-7-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholino-quinoline-4-carboxamide DIPEA (107 μL, 0.61 mmol) was added to 7-chloro-6-morpholinoquinoline-4-carboxylic acid Intermediate 336 (60 mg, 0.20 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (43 mg, 0.20 mmol) and T3P (521 mg, 0.82 mmol, 50% in EtOAc) in MeCN (5 mL) and EtOAc (5 ml), and the reaction mixture was stirred at 20° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (50 mL), and washed sequentially with water (15 mL) and sat brine (15 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod F, (gradient: 29-39%) to give the title compound (0.050 g, 55%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H21ClN5O3S: 446.1048, found: 446.1046;1H NMR (400 MHz, DMSO-d6) δ 9.23-9.05 (m, 1H), 8.88 (d, 1H), 8.16 (s, 1H), 8.09 (s, 1H), 7.55 (d, 1H), 5.34 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.41-4.27 (m, 2H), 3.81 (t, 4H), 3.45-3.34 (m, overlapping with solvent people), 3.20-3.05 (m, 4H). Example 180: (R)-8-Chloro-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-morpholino-quinoline-4-carboxamide A solution of T3P (608 mg, 0.96 mmol, 50% in EtOAc) in EtOAc (10 mL) was added to a stirred mixture of 8-chloro-6-morpholinoquinoline-4-carboxylic acid Intermediate 339 (70 mg, 0.24 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride, Intermediate 4 (50 mg, 0.24 mmol) in MeCN (10 mL) at 20° C., and the reaction mixture was stirred at 25° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with EtOAc (50 mL), and washed sequentially with sat brine (20 mL) and water (15 mL). The organic layer was dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 19-39%) to give the title compound (0.060 g, 56%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H21ClN5O3S: 446.1048, found: 446.1026;1H NMR (400 MHz, DMSO-d6) δ 9.07 (t, 1H), 8.79 (d, 1H), 7.91 (d, 1H), 7.71 (d, 1H), 7.50 (d, 1H), 5.31 (dd, 1H), 4.89 (d, 1H), 4.70 (d, 1H), 4.39-4.22 (m, 2H), 3.78 (t, 4H), 3.45-3.34 (m, overlapping with solvent). Example 181: (R)-6-(3-(Acetamidomethyl)-3-methylazetidin-1-yl)-N-(2-(4-cyano-thiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (32 mg, 0.15 mmol) was added to a mixture of 6-(3-(acetamidomethyl)-3-methylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 341 (40 mg), HATU (73 mg, 0.19 mmol) and DIPEA (89 μL, 0.51 mmol) in MeCN (1.5 mL) and EtOAc (1.5 mL), and the reaction mixture was stirred at rt overnight. DCM (10 mL) and NaHCO3(7 mL, aq) were added and the reaction mixture was filtered through a phase separator and evaporated under reduced pressure. The crude compound was purified by preparative HPLC, PrepMethod F, (gradient 5-95%) to give the title compound (3 mg, 6%); HRMS (ESI) m/z [M+H]+calcd for C23H27N6O3S: 467.1860, found: 467.1818;1H NMR (600 MHz, DMSO-d6) δ 8.89 (t, 1H), 8.57 (d, 1H), 7.98 (t, 1H), 7.83 (d, 1H), 7.37 (d, 1H), 7.07-7.01 (m, 2H), 5.33-5.25 (m, 1H), 4.84 (d, 1H), 4.68 (d, 1H), 4.31-4.21 (m, 2H), 3.74 (dd, 2H), 3.52 (d, 2H), 3.45-3.30 (m, overlapping with solvent), 3.25 (d, 2H), 1.79 (s, 3H), 1.23 (s, 3H). Example 182: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-fluoro-3-phenyl-azetidin-1-yl)quinoline-4-carboxamide 6-(3-Fluoro-3-phenylazetidin-1-yl)quinoline-4-carboxylic acid Intermediate 343 (22 mg, 0.07 mmol), HATU (39 mg, 0.10 mmol) and DIPEA (48 μL, 0.27 mmol) were mixed in MeCN (1 mL) and EtOAc (1 mL). (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (17 mg, 0.08 mmol) was added and the reaction mixture was stirred at rt for 1 h. DCM (8 mL) and NaHCO3(5 mL, aq) were added, and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The crude compound was purified by preparative SFC, PrepMethod SFC-E, (gradient: 30-35%), to give the title compound (2.4 mg, 7%); HRMS (ESI) m/z [M+H]+calcd for C25H23FN5O2S: 476.1550, found: 476.1560. Example 183: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(p-tolyl)azetidin-1-yl)quinoline-4-carboxamide 6-(3-(p-Tolyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 345 (18 mg, 0.06 mmol), HATU (32 mg, 0.08 mmol) and DIPEA (40 μL, 0.23 mmol) were mixed in MeCN (0.5 mL) and EtOAc (0.5 mL). (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (14 mg, 0.07 mmol) was added and the reaction mixture was stirred at rt for 1 h. DCM (5 mL) and NaHCO3(5 mL, aq) were added, and the reaction mixture was stirred, filtered through a phase separator, and evaporated under reduced pressure. The compound was purified by preparative HPLC, PrepMethod F, (gradient 5-95%) to give the title compound (85 mg, 32%); HRMS (ESI) m/z [M+H]+calcd for C26H26N5O2S: 472.1802, found: 472.1800;1H NMR (600 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.60 (d, 1H), 7.87 (d, 1H), 7.37 (d, 1H), 7.27-7.22 (m, 3H), 7.16-7.11 (m, 3H), 5.28 (dd, 1H), 4.84 (d, 1H), 4.67 (d, 1H), 4.37 (t, 2H), 4.25 (d, 2H), 3.99-3.85 (m, 3H), 3.35-3.25 (m, overlapping with water), 2.25 (s, 3H). Example 184: (R)-6-(6-Acetyl-2,6-diazaspiro[3.3]heptan-2-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide 6-(6-Acetyl-2,6-diazaspiro[3.3]heptan-2-yl)quinoline-4-carboxylic acid Intermediate 347 (90 mg, 0.29 mmol), HATU (165 mg, 0.43 mmol) and DIPEA (202 μL, 1.16 mmol) were mixed in MeCN (2 mL) and EtOAc (2 mL). (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (72 mg, 0.35 mmol) was added and the reaction mixture was stirred at rt for 3 h. DCM (15 mL) and NaHCO3(15 mL, aq) were added and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The crude compound was purified by preparative HPLC, PrepMethod F, (gradient 5-95%), to give the title compound (37 mg, 28%); HRMS (ESI) m/z [M+H]+calcd for C23H25N6O3S: 465.1704, found: 465.1692;1H NMR (600 MHz, DMSO-d6) δ 8.90 (t, 1H), 8.60 (d, 1H), 7.85 (d, 1H), 7.38 (d, 1H), 7.16 (d, 1H), 7.08 (dd, 1H), 5.31 (dd, 1H), 4.85 (d, 1H), 4.68 (d, 1H), 4.27 (s, 4H), 4.06 (s, 4H), 4.00 (s, 2H), 3.39-3.30 (m, 2H), δ 1.72 (s, 3H). Example 185: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(4-fluorophenyl)-azetidin-1-yl)quinoline-4-carboxamide 6-(3-(4-Fluorophenyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 349 (80 mg, 0.25 mmol), HATU (142 mg, 0.37 mmol) and DIPEA (173 μL, 0.99 mmol) were mixed in MeCN (2 mL) and EtOAc (2 mL). (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (62 mg, 0.30 mmol) was added and the reaction mixture was stirred at rt for 3 h. DCM (15 mL) and NaHCO3(15 mL, aq) were added, and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The crude compound was purified by preparative HPLC, PrepMethod F, (gradient 5-95%) to give the title compound (60 mg, 51%); HRMS (ESI) m/z [M+H]+calcd for C25H23FN5O2S: 476.1550, found: 476.1550;1H NMR (600 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.64 (d, 1H), 7.90 (d, 1H), 7.46-7.38 (m, 3H), 7.28 (d, 1H), 7.20-7.11 (m, 3H), 5.28 (dd, 1H), 4.84 (d, 1H), 4.67 (d, 1H), 4.39 (t, 2H), 4.27 (d, 2H), 4.00 (p, 1H), 3.91 (dq, 2H), 3.37-3.27 (m, 2H). Example 186: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-(m-tolyl)azetidin-1-yl)quinoline-4-carboxamide 6-(3-(m-Tolyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 351 (90 mg, 0.28 mmol), HATU (161 mg, 0.42 mmol) and DIPEA (197 μL, 1.13 mmol) were mixed in MeCN (2 mL) and EtOAc (2 mL). (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (70 mg, 0.34 mmol) was added and the reaction mixture was stirred at rt for 3 h. DCM (15 mL) and NaHCO3(15 mL, aq) were added, and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The compound was purified by preparative HPLC, PrepMethod F, (gradient 5-95%) to give the title compound (58 mg, 51%); HRMS (ESI) m/z [M+H]+calcd for C26H26N5O2S: 472.1802, found: 472.1792: 471.18;1H NMR (600 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.64 (d, 1H), 7.89 (d, 1H), 7.43 (d, 1H), 7.27 (d, 1H), 7.23-7.12 (m, 4H), 7.03 (d, 1H), 5.28 (dd, 1H), 4.84 (d, 1H), 4.67 (d, 1H), 4.38 (t, 2H), 4.27 (d, 2H), 3.99-3.88 (m, 3H), 3.37-3.26 (m, 2H), 2.27 (s, 3H). Example 187: (R)-6-(3-(4-Chlorobenzyl)azetidin-1-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide 6-(3-(4-Chlorobenzyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 353 (80 mg, 0.23 mmol), HATU (129 mg, 0.34 mmol) and DIPEA (158 μL, 0.91 mmol) were mixed in MeCN (2 mL) and EtOAc (2 mL). (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (56 mg, 0.27 mmol) was added and the reaction mixture was stirred at rt for 3 h. DCM (15 mL) and NaHCO3(15 mL, aq) were added, and the reaction mixture was stirred, filtered through a phase separator and evaporated under reduced pressure. The crude compound was purified by preparative HPLC, PrepMethod F, (gradient 5-95%) to give the title compound (18 mg, 16%); HRMS (ESI) m/z [M+H]+calcd for C26H25C1N5O2S: 506.1412, found: 506.1428;1H NMR (600 MHz, DMSO-d6) δ 8.89 (t, 1H), 8.57 (d, 1H), 7.82 (d, 1H), 7.36 (d, 1H), 7.33-7.29 (m, 2H), 7.25 (d, 2H), 7.11-7.03 (m, 2H), 5.27-5.21 (m, 1H), 4.82 (d, 1H), 4.66 (d, 1H), 4.24 (d, 2H), 3.99 (q, 2H), 3.66-3.59 (m, 2H), 3.35-3.28 (m, 2H), 2.99 (dt, 1H), 2.90 (d, 2H). Example 188: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-methyl-3-((methylsulfonyl)methyl)azetidin-1-yl)quinoline-4-carboxamide 6-(3-Methyl-3-((methylsulfonyl)methyl)azetidin-1-yl)quinoline-4-carboxylic acid Intermediate 355 (55 mg, 0.16 mmol), HATU (94 mg, 0.25 mmol) and DIPEA (115 μL, 0.66 mmol) were mixed in MeCN (1.5 mL) and EtOAc (1.5 mL). (R)-3-Glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (41 mg, 0.20 mmol) was added and the reaction mixture was stirred at rt overnight. DCM (10 mL) and NaHCO3(7 mL, aq) were added, and the reaction mixture was stirred, filtered through a phase separator, and evaporated under reduced pressure. The crude compound was purified by preparative SFC, PrepMethod SFC-E, (gradient 35-40%), to give the title compound (19 mg, 24%); HRMS (ESI) m/z [M+H]+calcd for C22H26N5O4S2: 488.1420, found: 488.1442;1H NMR (600 MHz, DMSO-d6) δ 8.89 (t, 1H), 8.59 (d, 1H), 7.84 (d, 1H), 7.38 (d, 1H), 7.08 (dd, 2H), 5.28 (dd, 1H), 4.83 (d, 1H), 4.67 (d, 1H), 4.29-4.21 (m, 2H), 3.94 (d, 2H), 3.71 (d, 2H), 3.59 (s, 2H), 2.98 (s, 3H), 3.35-3.25 (m, overlapping with solvent), 1.56 (s, 3H). Example 189: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-ethyl-4-hydroxy-piperidin-1-yl)quinoline-4-carboxamide DIPEA (262 μL, 1.50 mmol) was added to a stirred suspension of 6-(4-ethyl-4-hydroxypiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 357 (90 mg, 0.30 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (124 mg, 0.60 mmol), HOBt (121 mg, 0.90 mmol) and EDC (172 mg, 0.90 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 28° C. and the reaction mixture was stirred at 50° C. for 2 h. The reaction mixture was concentrated under reduced pressure and further diluted with sat NaHCO3(30 mL, aq) and EtOAc (100 mL). The aq layer was extracted with EtOAc (4×50 mL) and the combined organic layers was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod F (gradient: 13-23%) to give the title compound (0.089 g, 64%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H28N5O3S: 454.1908, found: 454.1926;1H NMR (300 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.64 (d, 1H), 7.86 (d, 1H), 7.70-7.60 (m, 2H), 7.39 (d, 1H), 5.40-5.23 (m, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.29 (d, 2H), 4.13 (brs, 1H), 3.67-3.51 (m, 2H), 3.50-3.15 (m, overlapping with solvent), 1.63-1.54 (m, 4H), 1.41 (q, 2H), 0.86 (t, 3H). Example 190: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-hydroxy-4-methyl-piperidin-1-yl)quinoline-4-carboxamide DIPEA (183 μL, 1.05 mmol) was added to a stirred suspension of 6-(4-hydroxy-4-methylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 359 (90 mg, 0.30 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride (109 mg, 0.52 mmol) Intermediate 4, HOBt (80 mg, 0.52 mmol) and EDC (100 mg, 0.52 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 28° C. and the reaction mixture was stirred at 50° C. for 2 h. The reaction mixture was concentrated under reduced pressure and was further diluted with EtOAc, and washed sequentially with H2O. The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod C (gradient: 10-16%) to give the title compound (0.075 g, 44%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1760;1H NMR (300 MHz, CD3OD) δ 8.61 (d, 1H), 7.90 (d, 1H), 7.75-7.63 (m, 2H), 7.50 (d, 1H), 5.34 (dd, 1H), 4.86-4.73 (m, 2H), 4.38 (d, 2H), 3.68-3.56 (m, 2H), 3.52-3.34 (m, overlapping with solvent), 1.85-1.65 (m, 4H), 1.26 (s, 3H). Example 191: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-ethyl-4-methoxy-piperidin-1-yl)quinoline-4-carboxamide DIPEA (142 μL, 0.81 mmol) was added to a stirred suspension of 6-(4-ethyl-4-methoxypiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 361 (85 mg, 0.27 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride (84 mg, 0.41 mmol) Intermediate 4, HOBt (62 mg, 0.41 mmol) and EDC (78 mg, 0.41 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 28° C. and the reaction mixture was stirred at 50° C. for 2 h. The reaction mixture was concentrated under reduced pressure and was further diluted with EtOAc (20 mL), and washed sequentially with H2O (2×15 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod C, (gradient 20-31%) to give the title compound (0.075 g, 59%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C24H30N5O3S: 468.2064, found: 468.2066;1H NMR (400 MHz, CD3OD) δ 8.62 (d, 1H), 7.90 (d, 1H), 7.75 (d, 1H), 7.68 (dd, 1H), 7.49 (d, 1H), 5.33 (dd, 1H), 4.85 (d, 1H), 4.77 (d, 1H), 4.39 (d, 2H), 3.75-3.63 (m, 2H), 3.51-3.32 (m, overlapping with solvent), 3.25-3.10 (m, 4H), 1.98-1.88 (m, 2H), 1.70-1.50 (m, 4H), 0.88 (t, 3H). Example 192: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-hydroxy-4-isopropyl-piperidin-1-yl)quinoline-4-carboxamide DIPEA (389 μL, 2.23 mmol) was added to a stirred suspension of 6-(4-hydroxy-4-isopropylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 363 (140 mg, 0.45 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride (185 mg, 0.89 mmol) Intermediate 4, HOBt (181 mg, 1.34 mmol) and EDC (256 mg, 1.34 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 28° C. and the reaction mixture was stirred at 50° C. for 2 h. The reaction mixture was concentrated under reduced pressure and was further diluted with EtOAc (100 mL) and sat NaHCO3(30 mL, aq). The aq layer was extracted with EtOAc (4×50 mL). The organic layers were combined and washed with H2O (3×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod F (gradient 18-28%) to give the title compound (0.12 g, 58%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C24H30N5O3S: 468.2064, found: 468.2068;1H NMR (300 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.65 (d, 1H), 7.86 (d, 1H), 7.70-7.59 (m, 2H), 7.39 (d, 1H), 5.40-5.25 (m, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.30 (d, 2H), 4.00 (brs, 1H), 3.80-3.60 (m, 2H), 3.50-3.34 (m, overlapping with solvent), 3.21-3.08 (m, 2H), 1.68-1.45 (m, 5H), 0.86 (d, 6H). Example 193: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4S,5S)-4-hydroxy-3,4,5-trimethylpiperidin-1-yl)quinoline-4-carboxamide DIPEA (194 μL, 1.11 mmol) was added to a stirred suspension of 6-((3R,4s,5S)-4-hydroxy-3,4,5-trimethylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 367 (70 mg, 0.22 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride (92 mg, 0.45 mmol) Intermediate 4, HOBt (90 mg, 0.67 mmol) and EDC (128 mg, 0.67 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 25° C. and the reaction mixture was stirred at 25° C. for 15 h. The reaction mixture was concentrated under reduced pressure and was further diluted with EtOAc (100 mL) and sat NaHCO3(20 mL, aq). The aq layer was extracted with EtOAc (4×75 mL). The organic layers were combined and washed with H2O (3×25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod C, (gradient 15-25%) to give the title compound (0.078 g, 75%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C24H30N5O3S: 468.2064, found: 468.2072;1H NMR (300 MHz, DMSO-d6) δ 8.98 (t, 1H), 8.61 (d, 1H), 7.83 (d, 1H), 7.68-7.58 (m, 2H), 7.34 (d, 1H), 5.80-5.22 (m, 1H), 4.87 (d, 1H), 4.70 (d, 1H), 4.27 (d, 2H), 4.00 (s, 1H), 3.65-3.45 (m, 2H), 3.44-3.34 (m, overlapping with solvent), 2.79 (t, 2H), 1.74-1.50 (m, 2H), 1.07 (s, 3H), 1.00-0.85 (m, 6H). Example 194: 6-((1R,5S)-9-Oxa-3-azabicyclo[3.3.1]nonan-3-yl)-N-(2-((R)-4-cyano-thiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide HATU (0.110 g, 0.29 mmol) was added to a stirred mixture of 6-((1R,5S)-9-oxa-3-azabicyclo[3.3.1]nonan-3-yl)quinoline-4-carboxylic acid Intermediate 369 (0.098 g, 0.24 mmol) and DIPEA (0.210 mL, 1.20 mmol) in a mixture of MeCN (1.1 mL) and EtOAc (1.1 mL) at rt. The reaction was stirred for 1 min after which Intermediate 4 (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride (0.060 g, 0.29 mmol) was added. The reaction was stirred for 1 h at rt. The reaction mixture was diluted with EtOAc (15 mL) and washed with sat NaHCO3(8 mL, aq) followed by H2O (2×2 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by flash chromatography (EtOAc:MeOH, gradient: 0% then 10%). The residue was further purified by preparative HPLC, PrepMethod G (gradient 5-35%) to give the title compound (0.053 g, 49%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O3S: 452.1750 found: 452.1746;1H NMR (500 MHz, DMSO-d6) δ 9.03 (t, 1H), 8.66 (d, 1H), 7.91 (d, 1H), 7.73-7.65 (m, 2H), 7.39 (d, 1H), 5.28-5.32 (m, 1H), 4.90 (d, 1H), 4.70 (d, 1H), 4.37-4.25 (m, 2H), 4.08-4.02 (m, 2H), 3.89 (d, 2H), 3.44-3.37 (m, 2H), 3.18-3.10 (m, 2H), 2.37-2.23 (m, 1H), 2.00-1.89 (m, 2H), 1.84-1.75 (m, 2H), 1.58-1.44 (m, 1H). Example 195: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-1-methyl-piperidin-4-yl)quinoline-4-carboxamide DIPEA (200 μL, 1.14 mmol) was added to a stirred suspension of 6-(4-fluoro-1-methylpiperidin-4-yl)quinoline-4-carboxylic acid Intermediate 373 (110 mg, 0.38 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride (119 mg, 0.57 mmol) Intermediate 4, HOBt (88 mg, 0.57 mmol) and EDC (110 mg, 0.57 mmol) in MeCN (6 mL) and EtOAc (6 mL) at 25° C. and the reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure. The reaction mixture was diluted with EtOAc and washed sequentially with H2O. The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod I (gradient 24-33%). The residue was further purified by preparative SFC, PrepMethod SFC-B to give the title compound (0.025 g, 15%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C22H25FN5O2S: 442.1708 found: 442.1698;1H NMR (400 MHz, CD3OD) δ 8.97 (d, 1H), 8.52 (s, 1H), 8.15 (d, 1H), 7.95 (dd, 1H), 7.71 (d, 1H), 5.68-5.38 (m, 1H), 4.87-4.72 (m, 2H), 4.52-4.36 (m, 2H), 3.57-3.35 (m, 2H), 3.00-2.83 (m, 2H), 2.65-2.50 (t, 2H), 2.49-2.36 (m, 5H), 2.25-2.06 (m, 2H). Example 196: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1H-pyrazol-5-yl)-quinoline-4-carboxamide To a suspension of 6-(1H-pyrazol-5-yl)quinoline-4-carboxylic acid Intermediate 374 (32 mg, 0.13 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (28 mg, 0.13 mmol) in EtOAc (0.7 mL) and MeCN (0.7 mL) was added HATU (64 mg, 0.17 mmol) and DIPEA (0.070 mL, 0.40 mmol). The solution was stirred at rt overnight and further stirred for 24 h. The reaction mixture was evaporated and the residue was purified by preparative SFC, PrepMethod SFC-C (gradient 27-32%) to give the title compound (8.9 mg, 17%); HRMS (ESI) m/z [M+H]+calcd for C19H17N6O2S: 393.1128 found: 393.1130;1H NMR (500 MHz, CD3OD) δ 8.91 (d, 1H), 8.87 (s, 1H), 8.34 (d, 1H), 8.13 (d, 1H), 7.72 (d, 1H), 7.66 (d, 1H), 7.06-7.02 (m, 1H), 5.43-5.38 (m, 1H), 4.89-4.70 (m, overlapping with solvent), 4.52-4.39 (m, 2H), 3.47-3.35 (m, 2H). Example 197: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-isopropyl-1H-pyrazol-5-yl)quinoline-4-carboxamide To a suspension of 6-(1-isopropyl-1H-pyrazol-5-yl)quinoline-4-carboxylic acid Intermediate 375 (56 mg, 0.20 mmol) and (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (41 mg, 0.20 mmol) in EtOAc (0.7 mL) and MeCN (0.7 mL) was added HATU (95 mg, 0.25 mmol) and DIPEA (0.104 mL, 0.60 mmol). The solution was stirred at rt overnight and further stirred for 24 h. The reaction mixture was evaporated and the residue was purified by preparative SFC, PrepMethod SFC-C (gradient 27-32%) to give the title compound (29 mg, 33%); HRMS (ESI) m/z [M+H]+calcd for C22H23N6O2S: 435.1598 found: 435.1590;1H NMR (600 MHz, DMSO-d6) δ 9.15 (t, 1H), 8.87 (d, 1H), 8.73 (d, 1H), 8.46 (s, 1H), 8.13-8.08 (m, 2H), 8.05 (d, 1H), 7.49 (d, 1H), 5.42 (dd, 1H), 4.93 (d, 1H), 4.74 (d, 1H), 4.62-4.52 (m, 1H), 4.37 (d, 2H), 3.45-3.32 (m, overlapping with solvent), 1.52-1.46 (m, 6H). Example 198: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3-fluoro-3-methylpyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (614 μL, 3.51 mmol) was added to a solution of rac-(R)-6-(3-fluoro-3-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 377 (439 mg, 0.35 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (146 mg, 0.70 mmol) and HATU (401 mg, 1.05 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 10° C. overnight and then concentrated under reduced pressure. The reaction mixture was diluted with sat NaHCO3(50 mL, aq), and extracted with EtOAc (6×50 mL). The organic layers were combined and washed with brine (5×200 mL). The combined organic layers was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod F (gradient: 15-25%) to give the title compound (0.064 g, 42%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H23FN5O2S: 428.1550 found: 428.1538;1H NMR (300 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.61 (d, 1H), 7.91 (d, 1H), 7.40 (d, 1H), 7.36-7.24 (m, 2H), 5.33 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.37-4.26 (m, 2H), 3.68-3.34 (m, overlapping with solvent), 2.34-2.02 (m, 2H), 1.70-1.57 (m, 3H). Example 199: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-2-cyclopropyl-pyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 Example 200: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-2-cyclopropyl-pyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 DIPEA (1.54 mL, 8.81 mmol) was added to a stirred suspension of rac-(R)-6-(2-cyclopropylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 421 (335 mg, 0.44 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (275 mg, 1.32 mmol), HOBt (472 mg, 3.08 mmol) and EDC (591 mg, 3.08 mmol in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 10° C. overnight and then concentrated under reduced pressure. The residue was dissolved with a mixture of sat NaHCO3(80 mL, aq) and EtOAc (100 mL). The aq layer was extracted with EtOAc (4×100 mL). The organic layers were combined and washed with H2O (3×75 mL). The aq layers were combined and extracted with EtOAc (3×25 mL). All organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod C, (gradient: 23-35%). The isomers were separated by preparative chiral HPLC on a CHIRALPAK® IA column (5 μm, 250×20 ID mm) using an isocratic run of 30% MeOH in Hexane/DCM (0.5% 2M NH3in MeOH) 3/1 as mobile phase, and with a flow rate of 20 mL/min; the first eluting compound gave the title compound Isomer 1 Example 199 (0.030 g, 44%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O2S: 436.1802 found: 436.1798;1H NMR (300 MHz, DMSO-d6) δ 8.89 (t, 1H), 8.54 (d, 1H), 7.83 (d, 1H), 7.43-7.24 (m, 3H), 5.40-5.25 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.27 (qd, 2H), 3.90-3.70 (m, 1H), 3.62-3.45 (m, 1H), 3.43-3.10 (m, overlapping with solvent), 2.19-1.80 (m, 4H), 1.02-0.87 (m, 1H), 0.60-0.41 (m, 2H), 0.40-0.24 (m, 1H), 0.22-0.08 (m, 1H); and the second eluting compound gave the title compound Isomer 2 Example 200 (0.025 g, 37%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O2S: 436.1802 found: 436.1796;1H NMR (300 MHz, DMSO-d6) δ 8.90 (t, 1H), 8.54 (d, 1H), 7.83 (d, 1H), 7.50-7.20 (m, 3H), 5.82-5.25 (m, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.35-4.19 (m, 2H), 3.90-3.74 (m, 1H), 3.53 (t, 1H), 3.45-3.10 (m, overlapping with solvent), 2.17-1.71 (m, 4H), 1.05-0.85 (m, 1H), 0.58-0.40 (m, 2H), 0.40-0.27 (m, 1H), 0.23-0.08 (m, 1H). Example 201: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 Example 202: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 DIPEA (1874 μL, 10.73 mmol) was added to a stirred suspension of rac-(R)-6-(3-methyl-2-oxopyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 379 (290 mg, 1.07 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (446 mg, 2.15 mmol), HOBt (822 mg, 5.36 mmol) and EDC (1028 mg, 5.36 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 15° C. The reaction mixture was stirred at 50° C. for 2 h and then concentrated under reduced pressure. The residue was dissolved with a mixture of sat NaHCO3(60 mL, aq) and EtOAc (100 mL). The aq layer was extracted with EtOAc (5×100 mL). The organic layers were combined and washed with H2O (3×50 mL). The aq layers were combined and extracted with EtOAc (4×25 mL). All organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod F (gradient: 15-35%). The isomers were separated by preparative chiral HPLC on a CHIRALPAK® IE Colum (5 μm, 250×20 ID mm) using an isocratic run of 50% MeOH in Hexane/DCM (0.5% 2M NH3in MeOH) 3/1 as mobile phase, and with a flow rate of 20 mL/min; the first eluting compound gave the title compound Isomer 1 Example 201 (0.052 g, 49%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438 found: 424.1438;1H NMR (400 MHz, DMSO-d6) δ 9.10 (t, 1H), 8.90 (d, 1H), 8.55 (dd, 1H), 8.30 (d, 1H), 8.08 (d, 1H), 7.56 (d, 1H), 5.40-5.30 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.33 (d, 2H), 3.98-3.74 (m, 2H), 3.44-3.35 (m, overlapping with solvent), 2.78-2.65 (m, 1H), 2.42-2.31 (m, 1H), 1.85-1.65 (m, 1H), 1.20 (d, 3H); and the second eluting compound gave the title compound Isomer 2 Example 202 (0.048 g, 45%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438 found: 424.1418;1H NMR (400 MHz, DMSO-d6) δ 9.10 (t, 1H), 8.90 (d, 1H), 8.55 (dd, 1H), 8.32 (d, 1H), 8.08 (d, 1H), 7.56 (d, 1H), 5.42-5.29 (m, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.54-4.17 (m, 2H), 3.99-3.82 (m, 2H), 3.51-3.34 (m, overlapping with solvent), 2.78-2.61 (m, 1H), 2.43-2 25 (m, 1H), 1.85-1.65 (m, 1H), 1.20 (d, 3H). Example 203: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide Isomer 1 Example 204: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R*)-3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxamide Isomer 2 DIPEA (509 mg, 3.94 mmol) was added to a stirred suspension of rac-(R)-6-(3-methyl-2-oxopiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 381 (280 mg, 0.98 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (337 mg, 1.97 mmol), T3P (50% in EtOAc, 1253 mg, 3.94 mmol) in DMF (5 mL) under air. The reaction mixture was stirred at 25° C. for 6 h. The solvent was removed under reduced pressure. The residue was purified by preparative TLC (DCM:MeOH; 18:1). The residue was further purified by preparative HPLC, PrepMethod B (gradient: 18-38%). The isomers were separated by preparative chiral HPLC on a CHIRALPAK® IE column (5 μm, 250×20 ID mm) using an isocratic run of 50% MeOH in Hexane/DCM (0.5% 2 M NH3in MeOH) 3/1 as mobile phase, and with a flow rate of 20 mL/min; the first eluting compound gave the title compound Isomer 1 Example 203 (0.025 g, 25%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O3S: 438.1594 found: 438.1592;1H NMR (300 MHz, DMSO-d6) δ 9.11 (t, 1H), 8.94 (d, 1H), 8.35-8.20 (m, 1H), 8.01 (d, 1H), 7.75 (dd, 1H), 7.55 (d, 1H), 5.42-5.22 (m, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.31 (d, 2H), 3.88-3.68 (m, 2H), 3.44-3.34 (m, overlapping with solvent), 2.63-2.53 (m, overlapping with solvent), 2.06-1.89 (m, 3H), 1.71-1.51 (m, 1H), 1.18 (d, 3H); and the second eluting compound gave the title compound Isomer 2 Example 204 (0.071 g, 71%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O3S: 438.1594 found: 438.1588;1H NMR (300 MHz, DMSO-d6) δ 9.12 (t, 1H), 8.94 (d, 1H), 8.40-8.20 (m, 1H), 8.01 (d, 1H), 7.75 (dd, 1H), 7.55 (d, 1H), 5.45-5.25 (m, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.31 (d, 2H), 3.94-3.63 (m, 2H), 3.44-3.33 (m, overlapping with solvent), 2.61-2.52 (m, overlapping with solvent), 2.15-1.80 (m, 3H), 1.71-1.53 (m, 1H), 1.18 (d, 3H). Example 205: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoropiperidin-1-yl)-quinoline-4-carboxamide DIPEA (6.11 mL, 35.0 mmol) was added to a solution of 6-(4-fluoropiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 383 (600 mg, 0.87 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (363 mg, 1.75 mmol), EDC (839 mg, 4.37 mmol) and HOBt (670 mg, 4.37 mmol) in MeCN (10 mL) and EtOAc (10 mL). The reaction mixture was stirred at 15° C. for 15 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (100 mL) and sat NaHCO3(60 mL, aq), and extracted with EtOAc (5×100 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod I (gradient: 40-52%) to give the title compound (0.21 g, 56%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H23FN5O2S: 428.1550, found: 428.1548;1H NMR (300 MHz, DMSO-d6) δ 9.00 (t, 1H), 8.66 (d, 1H), 7.88 (d, 1H), 7.77-7.62 (m, 2H), 7.38 (d, 1H), 5.31 (dd, 1H), 5.01-4.54 (m, 3H), 4.43-4.16 (m, 2H), 3.70-3.33 (m, overlapping solvent), 2.13-1.90 (m, 2H), 1.89-1.71 (m, 2H). Example 206: 6-(3-Azabicyclo[3.1.0]hexan-3-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide DIPEA (927 μL, 5.31 mmol) was added to a solution of 6-(3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylic acid Intermediate 385 (368 mg, 0.53 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (221 mg, 1.06 mmol) and HATU (604 mg, 1.59 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 10° C. overnight and then concentrated under reduced pressure. The reaction mixture was diluted with sat NaHCO3(50 mL, aq), and extracted with EtOAc (6×50 mL). The organic layers were combined and washed with brine (5×200 mL). The combined organic layers was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient: 15-25%) to give the title compound (0.132 g, 61%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O2S: 408.1488, found: 408.1486;1H NMR (300 MHz, DMSO-d6) δ 8.92 (t, 1H), 8.56 (d, 1H), 7.83 (d, 1H), 7.35 (d, 1H), 7.29 (dd, 1H), 7.23 (d, 1H), 5.74-5.31 (m, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.27 (d, 2H), 3.75-3.55 (m, 2H), 3.39-3.32 (m, overlapping with solvent), 1.82-1.60 (m, 2H), 0.85-0.65 (m, 1H), 0.35-0.15 (m, 1H). Example 207: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-methoxy-pyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (411 μL, 2.35 mmol) was added to a solution of (S)-6-(3-methoxypyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 387 (160 mg, 0.59 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (101 mg, 0.59 mmol) and HATU (335 mg, 0.88 mmol) in DMF (5 mL) The reaction mixture was stirred at 25° C. for 2 h under and then concentrated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH; 18:1) and further purified by preparative HPLC, PrepMethod D (gradient: 25-33%) to give the title compound (0.049 g, 19%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1584;1H NMR (300 MHz, DMSO-d6) δ 8.93 (t, 1H), 8.56 (d, 1H), 7.86 (d, 1H), 7.39-7.19 (m, 3H), 5.31 (dd, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.28 (d, 2H), 4.21-4.01 (m, 1H), 3.60-3.34 (m, overlapping with solvent), 2.20-1.99 (m, 2H). Example 208: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((1R,5S,6R)-6-(trifluoromethyl)-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxamide DIPEA (742 μL, 4.25 mmol) was added to a solution of 6-((1R,5S,6r)-6-(trifluoromethyl)-3-azabicyclo[3.1.0]hexan-3-yl)quinoline-4-carboxylic acid Intermediate 389 (349 mg, 0.42 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (176 mg, 0.85 mmol), EDC (407 mg, 2.12 mmol) and HOBt (325 mg, 2.12 mmol) in MeCN (9 mL) and EtOAc (9 mL). The reaction mixture was stirred at 40° C. for 3 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (100 mL) and washed with sat NaHCO3(3×200 mL, aq) and brine (3×200 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 20-55%) to give the title compound (0.201 g, 99%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H21F3N5O2S: 476.1362, found: 476.1356;1H NMR (300 MHz, DMSO-d6) δ 8.92 (t, 1H), 8.60 (d, 1H), 7.88 (m, 1H), 7.40-7.28 (m, 3H), 5.40-5.20 (m, 1H), 4.89 (d, 1H), 4.72 (d, 1H), 4.28 (d, 2H), 3.79 (d, 2H), 3.43-3.34 (m, overlapping with solvent), 2.33-2.10 (m, 2H), 1.98-1.73 (m, 1H). Example 209: (R)-6-(7-Azabicyclo[2.2.1]heptan-7-yl)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide DIPEA (560 μL, 3.21 mmol) was added to a suspension of 6-(7-azabicyclo[2.2.1]heptan-7-yl)quinoline-4-carboxylic acid Intermediate 391 (43 mg, 0.16 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (67 mg, 0.32 mmol), EDC (307 mg, 1.60 mmol) and HOBt (245 mg, 1.60 mmol) in MeCN (3 mL) and EtOAc (3 mL). The reaction mixture was stirred at 10° C. for overnight and then concentrated under reduced pressure. The reaction mixture was diluted sat NaHCO3(100 mL, aq) and extracted with EtOAc (6×100 mL) and washed with brine (5×200 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 10-35%) to give the title compound (0.025 g, 37%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O2S: 422.1646, found: 422.1658;1H NMR (300 MHz, DMSO-d6) δ 9.10-8.90 (m, 1H), 8.64 (d, 1H), 7.84 (d, 1H), 7.75-7.60 (m, 1H), 7.56 (dd, 1H), 7.36 (d, 1H), 5.34-5.28 (m, 1H), 4.88 (d, 1H), 4.70 (d, 1H), 4.43 (brs, 2H), 4.28 (d, 2H), 3.41-3.36 (m, overlapping with solvent), 1.85-1.64 (m, 4H), 1.58-1.35 (m, 4H). Example 210: 6-((1RS,4SR)-2-Azabicyclo[2.2.1]heptan-2-yl)-N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide TEA (291 μL, 2.08 mmol) was added to a suspension of 6-(2-azabicyclo[2.2.1]heptan-2-yl)quinoline-4-carboxylic acid Intermediate 393 (75 mg, 0.10 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (43 mg, 0.21 mmol), EDC (100 mg, 0.52 mmol) and HOBt (80 mg, 0.52 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 10° C. for overnight and then concentrated under reduced pressure. The reaction mixture was partitioned between sat NaHCO3(50 mL, aq) and EtOAc (100 mL), extracted with EtOAc (4×100 mL) and washed with brine (5×200 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 10-40%) to give the title compound (0.13 g) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H24N5O2S: 422.1646, found: 422.1658;1H NMR (300 MHz, DMSO-d) δ 8.89 (t, 1H), 8.51 (d, 1H), 7.81 (d, 1H), 7.40-7.10 (m, 3H), 5.40-5.20 (m, 1H), 4.87 (d, 1H), 4.70 (d, 1H), 4.45-4.15 (m, 3H), 3.60-3.34 (m, overlapping with solvent), 2.85 (d, 1H), 2.61 (s, 1H), 1.80-1.42 (m, 5H), 1.40-1.15 (m, 1H). Example 211: N-(2-((R)-4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-2-methylpyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (454 mg, 3.51 mmol) was added to a solution of (R)-6-(2-methylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 395 (150 mg, 0.59 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (150 mg, 0.88 mmol) and HATU (445 mg, 1.17 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 25° C. for 3 h and then concentrated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH; 19:1) and further purified by preparative HPLC, PrepMethod F (gradient: 15-32%) to give the title compound (0.050 g, 21%) as an orange solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O2S: 410.1646, found: 410.1634;1H NMR (400 MHz, DMSO-d6) δ 8.92 (t, 1H), 8.55 (d, 1H), 7.86 (d, 1H), 7.39-7.29 (m, 2H), 7.25 (d, 1H), 5.40-5.20 (m, 1H), 4.89 (d, 1H), 4.73 (d, 1H), 4.29 (d, 2H), 4.15-4.00 (m, 1H), 3.55-3.20 (m, overlapping with solvent), 2.16-1.90 (m, 3H), 1.82-1.65 (m, 1H), 1.17 (d, 3H). Example 212: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-2-(methoxymethyl)-pyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (549 μL, 3.14 mmol) was added to a suspension of (S)-6-(2-(methoxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 397 (150 mg, 0.52 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (163 mg, 0.79 mmol), EDC (201 mg, 1.05 mmol) and HOBt (160 mg, 1.05 mmol) in MeCN (4 mL) and EtOAc (4 mL). The reaction mixture was stirred at 50° C. for 3 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc and washed with H2O. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod P, (gradient: 15-25%) to give the title compound (0.205 g, 89%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O3S: 440.1750, found: 440.1772;1H NMR (300 MHz, DMSO-d6) δ 8.91 (t, 1H), 8.55 (d, 1H), 7.85 (d, 1H), 7.41-7.31 (m, 2H), 7.27 (d, 1H), 5.30 (dd, 1H), 4.87 (d, 1H), 4.70 (d, 1H), 4.27 (d, 2H), 4.06 (brs, 1H), 3.54-3.10 (m, overlapping with solvent), 2.13-1.93 (m, 4H). Example 213: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3S,4S)-3,4-difluoro-pyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (279 mg, 2.16 mmol) was added to a solution of 6-((3S,4S)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 399 (100 mg, 0.36 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (92 mg, 0.54 mmol) and HATU (273 mg, 0.72 mmol) in MeCN (2 mL) and EtOAc (2 mL). The reaction mixture was stirred at 25° C. for 3 h and then concentrated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH; 20:1) and further purified by preparative HPLC, PrepMethod F (gradient: 17-35%) to give the title compound (0.063 g, 40%) as a red solid; HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O2S: 432.1300, found: 432.1310;1H NMR (400 MHz, DMSO-d6) δ 9.10 (t, 1H), 8.75 (d, 1H), 8.00 (d, 1H), 7.50-7.25 (m, 3H), 5.70-5.25 (m, 3H), 4.91 (d, 1H), 4.72 (d, 1H), 4.45-4.25 (m, 2H), 3.95-3.65 (m, 4H), 3.47-3.33 (m, overlapping with solvent). Example 214: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R,4R)-3,4-difluoro-pyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (557 mg, 4.31 mmol) was added to a solution of rac-6-((3R,4R)-3,4-difluoropyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 401 (200 mg, 0.72 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (185 mg, 1.08 mmol) and HATU (547 mg, 1.44 mmol) in MeCN (4 mL) and EtOAc (4 mL). The reaction mixture was stirred at 25° C. for 3 h and then concentrated under reduced pressure. The crude product was purified by preparative TLC (DCM:MeOH; 20:1) and the isomers were separated by preparative chiral HPLC on a CHIRAL ART Cellulose-SB column (5 μm, 250×20 ID mm) using an isocratic run of 30% MeOH in Hexane/DCM (0.5% 2 M NH3in MeOH) 3/1 as mobile phase, and with a flow rate of 20 mL/min;the first eluting compound gave the title compound Example 214 (0.080 g, 26%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O2S: 432.1300, found: 432.1286;1H NMR (400 MHz, DMSO-d6) δ 8.97 (t, 1H), 8.63 (d, 1H), 7.93 (d, 1H), 7.50-7.28 (m, 3H), 5.65-5.38 (m, 2H), 5.34 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.37-4.27 (m, 2H), 3.92-3.68 (m, 4H), 3.49-3.34 (m, overlapping with solvent). Example 215: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2,4-dimethyloxazol-5-yl)quinoline-4-carboxamide DIPEA (0.078 mL, 0.45 mmol) was added to a solution of 6-(2,4-dimethyloxazol-5-yl)quinoline-4-carboxylic acid Intermediate 402 (30 mg, 0.11 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (23 mg, 0.11 mmol) and HATU (47 mg, 0.12 mmol) in DCM (4 mL). The reaction mixture was stirred at rt overnight. (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (23.22 mg, 0.11 mmol), HATU (14 mg, 0.04 mmol) and DIPEA (0.025 mL, 0.15 mmol) was added. The reaction mixture was stirred for 2 h and diluted with DMSO. The residue was purified by preparative HPLC, PrepMethod SFC-C (gradient: 20-25%) to give the title compound (37 mg, 79%). HRMS (ESI) m/z [M+H]+calcd for C21H20N5O3S: 422.1282, found: 422.1286;1H NMR (600 MHz, DMSO-d6) δ 9.17 (t, 1H), 8.98 (d, 1H), 8.57 (d, 1H), 8.16 (d, 1H), 8.04 (dd, 1H), 7.62 (d, 1H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.41-4.26 (m, 2H), 3.46-3.32 (m, overlapping with solvent), 2.48 (s, 3H), 2.40 (s, 3H). Example 216: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3,5-dimethylisoxazol-4-yl)quinoline-4-carboxamide DIPEA (0.98 mL, 5.6 mmol) was added to a solution of 6-(3,5-dimethylisoxazol-4-yl)quinoline-4-carboxylic acid Intermediate 403 (60 mg, 0.22 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (56 mg, 0.27 mmol) and HATU (102 mg, 0.27 mmol) in DMF (2 mL). The reaction mixture was stirred at rt overnight. The reaction mixture was diluted with DCM (15 mL) and washed with sat NaHCO3(8 mL, aq), passed through a phase separator and concentrated. The residue was purified by preparative HPLC, PrepMethod F (gradient: 5-95%) to give the title compound (57 mg, 60% over two-steps). HRMS (ESI) m/z [M+H]+calcd for C21H20N5O3S: 422.1282, found: 422.1298;1H NMR (600 MHz, DMSO-d6) δ 9.17 (t, 1H), 9.01 (d, 1H), 8.43 (d, 1H), 8.17 (d, 1H), 7.87 (dd, 1H), 7.60 (d, 1H), 5.33 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.32 (qd, 2H), 3.42-3.34 (m, overlapping with solvent), 2.54 (s, 6H). Example 217: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-phenyl-1H-imidazol-1-yl)quinoline-4-carboxamide TEA (133 μL, 0.95 mmol) was added to a suspension of 6-(2-phenyl-1H-imidazol-1-yl)quinoline-4-carboxylic acid Intermediate 404 (30 mg, 0.10 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (30 mg, 0.14 mmol, EDC (91 mg, 0.48 mmol) and HOBt (64 mg, 0.48 mmol) in MeCN (3 mL) and EtOAc (3 mL). The reaction mixture was stirred at 50° C. for 2 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (60 mL) and sat NaHCO3(30 mL, aq), extracted with EtOAc (5×75 mL). The pooled organic layers were washed with H2O (3×25 mL), dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 5-35%) to give the title compound (0.023 g, 52%) as a grey solid; HRMS (ESI) m/z [M+H]+calcd for C25H21N6O2S: 469.1442, found: 469.1450;1H NMR (300 MHz, DMSO-d6) δ 9.20-9.00 (m, 2H), 8.41 (d, 1H), 8.14-8.04 (m, 1H), 7.68 (d, 1H), 7.61-7.51 (m, 2H), 7.38-7.20 (m, 6H), 5.32 (dd, 1H), 4.85 (d, 1H), 4.68 (d, 1H), 4.28 (d, 2H), 3.37-3.33 (m, overlapping with solvent). Example 218: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(3-phenyl-1H-pyrrol-1-yl)quinoline-4-carboxamide DIPEA (417 μL, 2.39 mmol) was added to a suspension of 6-(3-phenyl-1H-pyrrol-1-yl)quinoline-4-carboxylic acid Intermediate 405 (150 mg, 0.48 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (149 mg, 0.72 mmol), EDC (183 mg, 0.95 mmol) and HOBt (129 mg, 0.95 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 50° C. for 2 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (25 mL) and washed with H2O (3×10 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative TLC (DCM:MeOH; 10:1) and further purified by preparative HPLC, PrepMethod C, (gradient: 40-60%) to give the title compound (0.105 g, 47%) as a grey solid; HRMS (ESI) m/z [M+H]+calcd for C26H22N5O2S: 468.1488, found: 468.1496;1H NMR (300 MHz, DMSO-d6) δ 9.24 (t, 1H), 8.93 (d, 1H), 8.77-8.65 (m, 1H), 8.31-8.14 (m, 3H), 7.76-7.68 (m, 3H), 7.58 (d, 1H), 7.49-7.25 (m, 2H), 7.19 (t, 1H), 6.84-6.76 (m, 1H), 5.40 (dd, 1H), 4.94 (d, 1H), 4.74 (d, 1H), 4.55-4.23 (m, 2H), 3.52-3.37 (m, overlapping with solvent). Example 219: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4,5,6,7-tetrahydro-1H-indol-1-yl)quinoline-4-carboxamide DIPEA (394 μL, 2.26 mmol) was added to a suspension of 6-(4,5,6,7-tetrahydro-1H-indol-1-yl)quinoline-4-carboxylic acid Intermediate 406 (50 mg, 0.15 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (47 mg, 0.23 mmol), EDC (144 mg, 0.75 mmol) and HOBt (102 mg, 0.75 mmol) in MeCN (4 mL) and EtOAc (4 mL). The reaction mixture was stirred at 50° C. for 2 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (60 mL) and sat NaHCO3(30 mL, aq), extracted with EtOAc (5×50 mL). The pooled organic layers were washed with H2O (3×20 mL), the aqueous layers were combined and extracted with EtOAc (3×20 mL) and all organic layers were pooled, dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 40-60%) to give the title compound (0.020 g, 30%) as a white solid; HRMS (ESI) m/z [M+H]+calcd for C24H24N5O2S: 446.1646, found: 446.1636;1H NMR (300 MHz, DMSO-d6) δ 9.25-9.10 (m, 1H), 8.97 (d, 1H), 8.25 (d, 1H), 8.15 (d, 1H), 7.87 (dd, 1H), 7.61 (d, 1H), 6.99 (d, 1H), 6.04 (d, 1H), 5.30 (dd, 1H), 4.87 (d, 1H), 4.71 (d, 1H), 4.40-4.20 (m, 2H), 3.38-3.35 (m, overlapping with solvent), 2.80-2.52 (m, overlapping with solvent), 1.78-1.66 (m, 4H). Example 220: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((R)-3-(hydroxymethyl)-pyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (192 μL, 1.10 mmol) was added to a suspension of (R)-6-(3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 408 (100 mg, 0.37 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (76 mg, 0.37 mmol), T3P (50% in EtOAc, 934 mg, 1.47 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 20° C. for 5 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (100 mL), washed with H2O (50 mL) and brine (50 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 1-25%) to give the title compound (0.027 g, 17%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1580;1H NMR (400 MHz, DMSO-d6) δ 8.92 (t, 1H), 8.56 (d, 1H), 7.86 (d, 1H), 7.37 (d, 1H), 7.29 (dd, 1H), 7.20 (d, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.77-4.68 (m, 2H), 4.30 (d, 2H), 3.53-3.32 (m, overlapping with solvent), 3.15 (dd, 1H), 2.50-2.42 (m, overlapping with solvent), 2.14-2.02 (m, 1H), 1.86-1.73 (m, 1H). Example 221: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((S)-3-(hydroxymethyl)-pyrrolidin-1-yl)quinoline-4-carboxamide DIPEA (231 μL, 1.32 mmol) was added to a suspension of (S)-6-(3-(hydroxymethyl)pyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 410 (120 mg, 0.44 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (92 mg, 0.44 mmol), T3P (50% in EtOAc, 1.12 g, 1.76 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 20° C. for 5 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (100 mL), washed with H2O (25 mL) and brine (25 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 5-23%) to give the title compound (0.032 g, 17%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H24N5O3S: 426.1594, found: 426.1596;1H NMR (400 MHz, DMSO-d6) δ 8.92 (t, 1H), 8.56 (d, 1H), 7.86 (d, 1H), 7.37 (d, 1H), 7.29 (dd, 1H), 7.20 (d, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.80-4.65 (m, 2H), 4.30 (d, 2H), 3.50-3.32 (m, overlapping with solvent), 3.15 (dd, 1H), 2.50-2.42 (m, overlapping with solvent), 2.14-2.02 (m, 1H), 1.86-1.73 (m, 1H). Example 222: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1-thia-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxamide DIPEA (198 μL, 1.13 mmol) was added to a suspension of 6-(1-thia-6-azaspiro[3.3]heptan-6-yl)quinoline-4-carboxylic acid Intermediate 412 (65 mg, 0.23 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (94 mg, 0.45 mmol), EDC (131 mg, 0.68 mmol) and HOBt (92 mg, 0.68 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 50° C. for 2 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (80 mL) and sat NaHCO3(30 mL, aq), extracted with EtOAc (5×75 mL). The pooled organic layers were washed with H2O (3×50 mL), dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod B (gradient: 26-46%) to give the title compound (0.055 g, 55%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O2S2: 440.1210, found: 440.1218;1H NMR (300 MHz, DMSO-d6) δ 8.94 (t, 1H), 8.62 (d, 1H), 7.86 (d, 1H), 7.39 (d, 1H), 7.20 (d, 1H), 7.09 (dd, 1H), 5.51-5.29 (m, 1H), 4.88 (d, 1H), 4.71 (d, 1H), 4.33-4.18 (m, 4H), 4.08 (d, 2H), 3.41-3.36 (m, overlapping with solvent), 3.22-3.00 (m, 4H). Example 223: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-fluoro-4-phenyl-piperidin-1-yl)quinoline-4-carboxamide DIPEA (274 μL, 1.57 mmol) was added to a suspension of 6-(4-fluoro-4-phenylpiperidin-1-yl)quinoline-4-carboxylic acid Intermediate 415 (110 mg, 0.31 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (98 mg, 0.47 mmol), EDC (120 mg, 0.63 mmol) and HOBt (85 mg, 0.63 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 50° C. for 2 h and then concentrated under reduced pressure. The reaction mixture was diluted with EtOAc (25 mL), washed with H2O (2×15 mL), dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod B (gradient: 59-66%) to give the title compound (0.060 g, 38%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C27H27FN5O2S: 504.1864, found: 504.1872;1H NMR (300 MHz, CD3OD) δ 8.67 (d, 1H), 7.97 (d, 1H), 7.88 (d, 1H), 7.78 (dd, 1H), 7.53 (d, 1H), 7.50-7.25 (m, 5H), 5.33 (dd, 1H), 4.88-4.68 (m, overlapping with solvent), 4.41 (d, 2H), 4.17-3.99 (m, 2H), 3.58-3.33 (m, overlapping with solvent), 2.48-2.03 (m, 4H). Example 224: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((RS)-3,3-difluoro-4-hydroxypyrrolidin-1-yl)quinoline-4-carboxamide TEA (142 μL, 1.02 mmol) was added to a suspension of rac-(R)-6-(3,3-difluoro-4-hydroxypyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 417 (100 mg, 0.34 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (71 mg, 0.34 mmol), EDC (98 mg, 0.51 mmol) and HOBt (69 mg, 0.51 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 20° C. for 16 h, diluted with EtOAc (75 mL), washed with sat NaHCO3(3×25 mL, aq), H2O (20 mL), brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient: 5-30%) to give the title compound (0.055 g, 35%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C20H20F2N5O3S: 448.1250, found: 448.1242;1H NMR (400 MHz, DMSO-d6) δ 9.00 (t, 1H), 8.64 (d, 1H), 7.93 (d, 1H), 7.40 (d, 1H), 7.38-7.30 (m, 2H), 6.17 (d, 1H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.40 (brs, 1H), 4.34-4.27 (d, 2H), 3.98-3.75 (m, 3H), 3.49-3.33 (m, overlapping with solvent). Example 225: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R*,4R*)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 1 Example 226: N-(2-((R)-4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-((3R*,4R*)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxamide Isomer 2 DIPEA (1.17 mL, 6.70 mmol) was added to a stirred suspension of rac-6-((3R,4R)-3,4-dimethylpyrrolidin-1-yl)quinoline-4-carboxylic acid Intermediate 419 (410 mg, 0.67 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (209 mg, 1.00 mmol) and HATU (762 mg, 2.01 mmol) in MeCN (5 mL) and EtOAc (5 mL). The reaction mixture was stirred at 4° C. for 15 h and then concentrated under reduced pressure. The residue was dissolved with a mixture of sat NaHCO3(50 mL, aq) and EtOAc (100 mL). The aq layer was extracted with EtOAc (3×100 mL). The organic layers were combined and washed with H2O (3×75 mL). The aq layers were combined and extracted with EtOAc (4×25 mL). All organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by preparative HPLC, PrepMethod F (gradient: 25-40%). The isomers were separated by preparative chiral HPLC on a CHIRAL ART Cellulose-SB column (5 μm, 250×20 ID mm) using an isocratic run of 50% MeOH in MTBE (0.5% 2 M NH3in MeOH) as mobile phase, and with a flow rate of 20 mL/min; the first eluting compound gave the title compound Isomer 1 Example 225 (0.088 g, 37%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O2S: 424.1802, found: 424.1798;1H NMR (300 MHz, DMSO-d6) δ 8.90 (t, 1H), 8.53 (d, 1H), 7.83 (d, 1H), 7.34 (d, 1H), 7.30-7.06 (m, 2H), 5.40-5.22 (m, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.28 (d, 2H), 3.62 (dd, 2H), 3.42-3.33 (m, overlapping with solvent), 2.98 (t, 2H), 2.00-1.78 (m, 2H), 1.08 (d, 6H); and the second eluting compound gave the title compound Isomer 2 Example 226 (0.088 g, 37%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C22H26N5O2S: 424.1802, found: 424.1808;1H NMR (300 MHz, DMSO-d6) δ 8.91 (t, 1H), 8.53 (d, 1H), 7.83 (d, 1H), 7.34 (d, 1H), 7.29-7.09 (m, 2H), 5.40-5.27 (m, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.40-4.20 (m, 2H), 3.70-3.53 (m, 2H), 3.45-3.34 (m, overlapping with solvent), 2.98 (t, 2H), 2.00-1.78 (m, 2H), 1.08 (d, 6H). Example 227: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,5-dioxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxamide DIPEA (586 μL, 3.36 mmol) was added to a solution of 6-(1,5-dioxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylic acid Intermediate 424 (220 mg, 0.67 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (209 mg, 1.01 mmol), HOBt (272 mg, 2.01 mmol) and EDC (386 mg, 2.01 mmol) in MeCN (10 mL) and EtOAc (10 mL) at 15° C. The reaction mixture was stirred at 40° C. for 3 h. The solvent was removed under reduced pressure and the residue was diluted with sat NaHCO3(aq, 200 mL). The aqueous phase was extracted with EtOAc (3×200 mL), and the combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod T, (gradient: 14-47%) to give the title compound (0.157 g, 49%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C24H28N5O4S: 482.1856 found: 482.1854;1H NMR (400 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.66 (d, 1H), 7.87 (d, 1H), 7.74-7.63 (m, 2H), 7.39 (d, 1H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.72 (d, 1H), 4.30 (dd, 2H), 3.87 (t, 4H), 3.45-3.34 (m overlapping with solvent), 1.98-1.90 (m, 4H), 1.63 (p, 2H). Example 228: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(1,4-dioxa-8-azaspiro[4.5]decan-8-yl)quinoline-4-carboxamide DIPEA (351 μL, 2.01 mmol) was added to a solution of 6-(1,4-dioxa-8-azaspiro[4.5]-decan-8-yl)quinoline-4-carboxylic acid Intermediate 426 (126 mg, 0.40 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (125 mg, 0.60 mmol), HOBt (544 mg, 4.02 mmol) and EDC (771 mg, 4.02 mmol) in MeCN (6.0 mL) and EtOAc (6.0 mL) at 15° C. The reaction mixture was stirred at 15° C. for 16 h under an atmosphere of N2(g). The solvent was removed under reduced pressure, and the residue was diluted with sat NaHCO3(aq, 250 mL). The aqueous phase was extracted with EtOAc (3×250 mL), and the combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod C, (gradient 12-22%) to give the title compound (0.065 g, 34%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O4S: 468.1705 found: 468.1723;1H NMR (300 MHz, DMSO-d6) δ 9.03 (t, 1H), 8.67 (d, 1H), 7.89 (d, 1H), 7.78-7.64 (m, 2H), 7.40 (d, 1H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.30 (d, 2H), 3.92 (s, 4H), 3.56-3.45 (m, 4H), 3.41-3.25 (m, overlapping with solvent), 1.76 (dd, 4H). Example 229: (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxa-7-azaspiro[3.5]-nonan-7-yl)quinoline-4-carboxamide Step a) 6-(2-Oxa-7-azaspiro[3.5]nonan-7-yl)quinoline-4-carboxylic acid NaOH (88 mg, 2.19 mmol) was added to a solution of methyl 6-(2-oxa-7-azaspiro[3.5]nonan-7-yl)quinoline-4-carboxylate Intermediate 427 (137 mg, 0.44 mmol) in MeOH (9.0 mL) and water (3.0 mL), and the reaction mixture was stirred at 15° C. for 2 h. The solvent was removed under reduced pressure. The residue was diluted with water (20 mL) and the pH was adjusted to 3 with aq HCl (1 M). The aqueous phase was extracted with EtOAc (10×50 mL), and the combined organic layer was dried over Na2SO4, filtered and evaporated to give the title compound (0.55 g) as a crude orange solid; MS (ESI) m/z [M+H]+299.05. Step b) (R)-N-(2-(4-Cyanothiazolidin-3-yl)-2-oxoethyl)-6-(2-oxa-7-azaspiro[3.5]nonan-7-yl)quinoline-4-carboxamide DIPEA (754 μL, 4.32 mmol) was added to 6-(2-oxa-7-azaspiro[3.5]nonan-7-yl)quinoline-4-carboxylic acid from Step a) (541 mg, 0.43 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (134 mg, 0.65 mmol), HOBt (583 mg, 4.32 mmol) and EDC (827 mg, 4.32 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 15° C. The reaction mixture was stirred at 15° C. for 16 h. The solvent was removed under reduced pressure, and the residue was diluted with sat NaHCO3(aq, 250 mL), The aqueous phase was extracted with EtOAc (3×250 mL), and the combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC, PrepMethod R, (gradient: 15-35%) to give the title compound (0.171 g, 88%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C23H26N5O3S: 452.1750, found: 452.1724;1H NMR (400 MHz, DMSO-d6) δ 8.99 (t, 1H), 8.67 (d, 1H), 7.87 (d, 1H), 7.71-7.62 (m, 2H), 7.39 (d, 1H), 5.33 (dd, 1H), 4.90 (d, 1H), 4.71 (d, 1H), 4.31 (s, 4H), 4.30 (dd, 2H), 3.40-3.27 (m, overlapping with solvent), 1.96-1.89 (m, 4H). Example 230: (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-6-(4-oxopiperidin-1-yl)quinoline-4-carboxamide DIPEA (189 μL, 1.08 mmol) was added to a solution of 6-(1,5-dioxa-9-azaspiro[5.5]undecan-9-yl)quinoline-4-carboxylic acid Intermediate 424 (71 mg, 0.22 mmol), (R)-3-glycylthiazolidine-4-carbonitrile hydrochloride Intermediate 4 (67 mg, 0.32 mmol), HOBt (292 mg, 2.16 mmol) and EDC (415 mg, 2.16 mmol) in MeCN (5 mL) and EtOAc (5 mL) at 15° C. The reaction mixture was stirred at 50° C. for 2 h. The solvent was removed under reduced pressure and the residue was diluted with sat NaHCO3(aq, 200 mL). The aqueous phase was extracted with EtOAc (3×200 mL), and the combined organic layer was washed with water (3×50 mL), dried over Na2SO4, filtered and evaporated at reduced pressure. The crude product was purified by preparative HPLC, PrepMethod X, (gradient: 14-24%). The product containing fractions were collected and evaporated and the residue was lyophilized from a mixture of water/MeCN (×3) to give the title compound (57 mg, 61%) as a yellow solid; HRMS (ESI) m/z [M+H]+calcd for C21H22N5O3S: 424.1438 found: 424.1450;1H NMR (500 MHz, DMSO-d6) δ 9.02 (t, 1H), 8.68 (d, 1H), 7.94 (d, 1H), 7.85 (d, 1H), 7.75 (dd, 1H), 7.40 (d, 1H), 5.32 (dd, 1H), 4.89 (d, 1H), 4.71 (d, 1H), 4.37-4.24 (m, 2H), 3.82 (t, 4H), 3.50-3.34 (m, overlapping with solvent), 2.49-2.46 (m, overlapping with solvent). D. Biological Data The hFAP protein used in the Examples was either commercially sourced or produced in insect cells as recombinant hFAP (Gp67-6HN-TEV-FAP(M39-A757), MW 89086.7 Da, or cd33-FAP (27-757)-6His, MW85926 Da). Recombinant hFAP protein was secreted from Sf21 cells in media, purified with affinity (batchmode, Ni excel resin, ÄKTA, GE Healthcare) and size exclusion chromatography (Superdex200, ÄKTA, GE Healthcare), concentrated to 19.5 mg/mL, snapfrozen in liquid N2and stored in −80° C. Example 231: FAP Inhibition and Binding Assays A. hFAP Inhibition Assay Compounds were tested in a biochemical inhibition assay using hFAP enzyme at 0.24 nM FAC (Proteros, 38-760 (PR-0071)) and the substrate Ala-Pro-AMC (ARI-3144) at 20 μM FAC. 384 low volume black plates (Greiner #784076) were used. 4 μL, 0.48 nM enzyme solution (100 mM Tris HCl, 100 mM NaCl, 0.05% Chaps, pH 7.4) was added to 40 nL compounds (in DMSO) at 10 CR, 3 fold dilution series from 50 μM FAC. Plates were incubated for 15 min at rt in dark. 4 μL, 40 μM substrate solution (100 mM Tris HCl, 100 mM NaCl, 0.05% Chaps, pH 7.4) was added to each well. Plates were centrifuged at 1000 rpm and incubated for 30 min at rt in dark. The plates were read on a PHERAstar® reader with excitation 340 nm and emission 460 nm. Data were analyzed in Genedata Screener®. IC50values were determined by plotting % inhibition versus log compound concentration and using a one site dose response model. Raw data signals were normalized using 0.5% DMSO as 0% control and Reference Compound A (i.e., (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-1-yl)-2-oxoethyl)quinoline-4-carboxamide as reported inJ. Med. Chem.2014, 57, 3053) at 50 μM as 100% inhibitor control. Data for the compounds tested are reported in Table 1. B hFAP Inhibition Assay (Tight Binders) Compounds were tested in a biochemical inhibition assay using human Fibroblast activation protein alpha (hFAP) enzyme at 2.4 μM FAC (Proteros, 38-760 (PR-0071) and the substrate Ala-Pro-AMC (ARI-3144) at 20 μM FAC. 384 low volume black plates (Greiner #784076) were used. 4 μL, 4.8 μM enzyme solution (100 mM Tris HCl, 100 mM NaCl, 0.05% Chaps, pH 7.4) was added to 40 nL compounds (in DMSO) at 10 CR, 3 fold dilution series from 50 nM FAC. Plates were incubated for 15 min at rt in dark. 4 μL, 40 μM substrate solution (100 mM Tris HCl, 100 mM NaCl, 0.05% Chaps, pH 7.4) was added to each well. Plates were centrifuged at 1000 rpm and incubated for 2.5 h at rt in dark. The plates were read on a PHERAstar® reader with excitation 340 nm and emission 460 nm. Data were analyzed in Genedata Screener®. IC50values were determined by plotting % inhibition versus log compound concentration and using a one site dose response model. Raw data signals were normalized using 0.5% DMSO as 0% control and Reference Compound A (i.e., (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-1-yl)-2-oxoethyl)quinoline-4-carboxamide as reported inJ. Med. Chem.2014, 57, 3053) at 50 μM as 100% inhibitor control. Data for the compounds tested are reported in Table 1. C. hFAP Binding Assay Compounds were tested in a direct binding assay using 8K surface plasmon resonance biosensor (GE Healthcare) at 20° C. Immobilization of hFAP (M39-A757) on a CMD200M sensor chip (Xantec) was performed using standard amine coupling procedure in immobilization buffer (10 mM HEPES, 150 mM NaCl, 0.05% Tween20, pH 7.4). The surface was washed with 10 mM NaOH, 1M NaCl before being activated with EDC/NHS (GE Healthcare), followed by immobilization of hFAP (in 10 mM Acetate pH 5.0). Finally, the surface was deactivated by ethanolamine. Immobilization levels of hFAP were around 4000-6000 RU. The reference spot was treated as described, omitting the injection of hFAP. Compound concentration series were injected over the immobilized protein in increasing concentrations (2-500 nM) using single cycle kinetics in running buffer (20 mM TRIS, 150 mM NaCl, 0.05% Tween20, 1% DMSO, pH 7.4). Interaction models were fitted globally to the experimental traces, enabling determination of kon, koffand Kd. Data for the compounds tested are reported in Table 1. TABLE 1hFAP inhibitionhFAP inhibition assayhFAP Bindingassay(tight binders)hFAP Binding assayhFAP Binding assayassayExampleIC50(nM)1IC50(nM)2Kd(nM)3K(on)(M−1s−1)3K(off)(1/s)310.0990.187300000.0001420.529.54100000.003830.240.130.09541000000.0004040.0580.07912000000.00009450.0600.049NVNV60.510.38400000000001870.160.107400000.00007580.0630.03213000000.00003490.250.6612000000.00076100.0540.153700000.000056110.0570.044NVNV120.0650.03819000000.000064130.0410.01492000000.000076140.0800.05013000000.000064150.120.114900000.000046160.110.0547400000.000041170.220.0850.01583000000.00012180.190.0470.06814000000.000059190.660.380.154300000.000066200.740.430.03042000000.00012210.290.110.06630000000.00020220.340.120.0509400000.000047230.310.200.08521000000.00018240.270.200.0094250000000.00048250.410.300.07614000000.00011260.310.100.05613000000.000065270.290.110.02411000000.000026280.260.100.1111000000.00011290.130.0550.05711000000.000061300.280.120.04252000000.00022310.220.100.03197000000.00024320.300.160.02213000000.000029330.340.270.05911000000.000052340.270.220.05722000000.000082350.470.170.109000000.00010360.240.0840.05330000000.000090370.220.190.02810000000.000030380.100.115200000.000078390.280.100.04049000000.00016400.240.0650.04313000000.000046410.330.160.0936800000.000064420.0530.03018000000.000054430.0450.006132000000.000019440.220.0740.04217000000.000072450.200.140.04854000000.00018460.290.0820.02030000000.000062470.220.200.03156000000.00016480.290.110.06714000000.000089490.190.0410.02088000000.00017500.340.0690.071NVNV510.210.0640.02156000000.00011520.200.0450.04468000000.00019530.270.180.0746800000.000050540.220.0620.04562000000.00023550.250.0830.03667000000.00025560.440.190.145400000.00011570.260.0980.02280000000.00018580.230.0760.01688000000000.091590.270.0610.08726000000.00019600.220.140.09917000000.00017610.210.0600.062NVNV620.250.0580.127100000.000085630.330.190.03041000000.000059640.280.0850.1213000000.00015650.280.170.05567000000.00047660.0770.035260000000.00022670.370.0840.01249000000.000046680.370.100.1110000000.00011690.290.0700.07014000000.000093700.120.02254000000.00014710.220.0680.050NVNV720.0790.02652000000.00016730.130.06624000000.00016740.0940.1411000000.00016750.0500.006323000000.000024760.0800.05110000000.000052770.0510.01448000000.000066780.0970.128800000.00010790.0430.018160000000.00012800.100.03624000000.000088810.130.04414000000.000063820.110.05214000000.000071830.0820.03917000000.000064840.0740.1212000000.00011850.0870.03240000000.00011860.0740.0817300000.000058870.400.110.03928000000.00011880.310.0740.08242000000.00034890.0960.05336000000.00015900.750.350.384800000.00010910.0880.07232000000.00021920.0740.04722000000.00011930.0910.04718000000.000080940.0710.07419000000.000099950.160.02429000000.000056960.170.06740000000.00022970.230.116900000.000076980.270.110.008816000000.000015990.120.1121000000.000201000.500.270.7330000000.00221010.380.350.0347400000.0000251020.210.0660.04620000000.0000801030.310.110.03330000000.000101040.210.0810.02030000000.0000611050.370.0880.0828900000.0000681060.230.0800.07518000000.000141070.640.210.08319000000.000161080.280.120.03814000000.0000501091.28.512000000.00991100.0540.01922000000.0000501110.0640.05518000000.0000961120.200.314400000.000141130.250.120.03115000000.0000451140.0720.062NV, NVNV, NV1150.0400.11NV, NVNV, NV1160.160.1579000000.00121170.0610.060NV, NVNV, NV1180.100.17NVNV1190.160.396100000.000311200.150.13NVNV1210.150.01211000000.0000161220.0490.03313000000.0000431230.250.0830.1430000000.000421240.240.0550.09430000000.000281250.210.0460.04830000000.000141260.320.0810.1118000000.000211270.380.160.3460000000.00201280.450.240.1554000000.000661290.200.0410.08714000000.000121300.190.184400000.0000781310.120.03318000000.000141320.410.522700000.000141330.100.06612000000.0000731340.130.136200000.0000801350.110.01317000000.0000231360.0610.03620000000.0000501370.110.01395000000.0000981380.0470.02825000000.0000751390.320.015NVNV1400.100.06426000000.000161410.0590.06214000000.0000781420.0880.02818000000.0000551430.0690.03616000000.0000571440.0810.04991000000.000511450.0700.02945000000.000131460.150.1214000000.000171470.0670.04528000000.000121480.0590.03166000000.000201490.0970.008748000000.0000391500.500.162800000.0000431510.130.03915000000.0000531520.0620.01310000000.0000141530.110.01553000000.0000851540.0920.0599100000.0000541550.0860.01631000000.0000501560.220.05612000000.0000661570.370.06519000000.000121580.100.1031000000.000291590.0730.03921000000.0000921600.0500.06811000000.0000741610.0520.022140000000.000281620.0530.01819000000.0000281630.0790.034NVNV1640.0810.02714000000.0000361650.320.05112000000.0000631660.340.220.03022000000.0000491670.0911680.160.03216000000.0000481690.220.0570.1662000000.000961700.240.0630.017100000000.000181710.280.130.08610000000.0000921720.550.260.03624000000.0000761730.270.0780.511100000.0000541740.440.260.1319000000.000241750.340.120.08614000000.000121760.540.0871.91200000.000221770.290.110.561000000.0000581780.200.0420.05331000000.000171790.0480.05232000000.000161800.201.35600000.000951810.940.380.161800000.0000271820.410.190.0932500000.0000421830.330.190.086NVNV1840.400.220.0159500000.0000141850.230.120.043NVNV1860.270.100.045NVNV1870.0980.0720.07031000000.000221880.280.190.01126000000.0000221890.0560.02026000000.0000511900.0690.02018000000.0000361910.0380.02215000000.0000331920.0510.007713000000.0000141930.240.167400000.000121940.0460.06611000000.0000681950.390.331900000.0000611960.190.0660.08032000000.000261970.720.420.75880000.0000661980.0560.03829000000.000111990.300.1023000000.000262000.240.09025000000.00023201<0.0250.05918000000.000102020.0440.04316000000.0000702030.260.154400000.0000632040.0670.0688500000.0000582050.270.0670.02621000000.0000462060.220.0650.033NVNV2070.170.01146000000.0000532080.220.110.04656000000.000262090.440.150.02032000000.0000422100.440.180.02638000000.0000982110.200.100.04959000000.000202120.290.260.1033000000.000342130.180.0390.03944000000.000172140.200.0790.04329000000.0000922150.410.120.09518000000.000172162.41.70.2752000000.000902170.670.7118000000.00132180.0680.486700000.000322190.110.08324000000.000202200.0910.03816000000.0000502210.170.004885000000.0000462220.0870.022NVNV2230.0760.01025000000.0000262240.0730.02330000000.0000682250.110.077NVNV2260.200.044NVNV2280.0872290.0910.01024000000.0000262300.0781IC50is reported after single measurement (n = 1) or as geometric mean for multiple measurements (n = 2-3).2IC50is reported after single measurement (n = 1) or as geometric mean for multiple measurements (n = 2-6).3Kdis reported after single measurement (n = 1) or as geometric mean for multiple measurements (n = 2-4).K(on)and k(off)are reported after single measurement (n = 1) or as an average for multiple measurements (n = 2-4).NV is not valid. D. FAP Plasma Inhibition Assay This assay was adapted from the method described in Example 237 for detection of FAP target engagement enzyme activity in plasma. Plasma (anticoagulant K2EDTA) was used as the enzyme source: Human plasma (Pooled from AZ Biobank), Mouse plasma (AZ AST Biobank), and Cynomolgus plasma (BioIVT, #NHP00PLK2FNN, lot CYN222895). 384-Well black fluotrack PS plates (Greiner 781076) were used. 20 μL diluted plasma (Cynomolgus and Human plasma dilution 1:40, Mouse plasma dilution 1:67) in buffer (PBS, 0.1% BSA) was added to 0.6 μL compounds (in DMSO). Compounds were tested using 10 CR, 3 fold dilution series from 500 nM FAC. Two replicates for each assay point were run on the same plate. A fluorescence blank read was taken before substrate addition. Substrate, Ala-Pro-AMC (ARI-3144) stock solution (20 mM in DMSO) was diluted in buffer (PBS, 0.1% BSA) to 150 μM concentration and 20 μL added giving 75 μM FAC. Plates were incubated for 40 min at rt in the dark. The plates were read on a Beckman Paradigm® reader with excitation 360 nm and emission 465 nm. Data were analyzed in Excel (IDBS XLfit Add-In) using a one site dose response model (4-parameter logistic fit). IC50values were determined by plotting % inhibition versus log compound concentration. Raw data signals were normalized using 1.5% DMSO in diluted plasma as 0% control and 1.5% DMSO in buffer (no plasma) as 100% inhibitor control. Data for the compounds tested are reported in Table 2. TABLE 2FAP Human PlasmaFAP Mouse PlasmaFAP Cyno PlasmaExampleIC50(nM)1IC50(nM)2IC50(nM)328.17.39.280.100.200.2090.600.600.60160.200.400.20240.200.400.30270.200.500.30280.200.500.30500.300.500.30670.360.520.40680.300.500.30690.200.300.20700.200.300.20710.200.200.20720.200.200.20740.200.300.20820.300.400.30880.200.300.20980.200.400.301020.200.200.201120.600.800.501130.200.500.401470.100.200.101510.200.400.301570.300.600.401630.100.200.201660.300.600.301700.300.501710.200.301720.200.601730.100.201740.200.401750.200.401780.100.101810.300.900.501840.200.600.301950.901.411960.100.202030.400.500.402040.200.300.202050.300.300.301IC50is reported after single measurement (n = 1) or as geometric mean for multiple measurements (n = 4).2IC50is reported after single measurement (n = 1) or as geometric mean for multiple measurements (n = 4).3IC50is reported after single measurement (n = 1). Example 232: hPrep Inhibition Assay Compounds were tested in a biochemical inhibition assay using Prolyl endopeptidase, Prolyl Oligopeptidase (hPREP) enzyme at 0.6 nM FAC (R&D Systems, 4308-SE) and the substrate Z-Gly-Pro-amino-methylcoumarin (Bachem, 1-1145) at 50 μM FAC. 384 Low volume black plates (Greiner #784076) were used. 4 μL, 1.2 nM enzyme solution (25 mM Tris HCl, 250 mM NaCl, 0.01% Triton X-100, 5 mM Glutathione, pH 7.5) was added to 40 nL compounds (in DMSO) at 10 CR, 3 fold dilution series from 50 μM FAC. Plates were incubated for 15 min at rt in dark. 4 μL, 100 μM substrate solution (25 mM Tris HCl, 250 mM NaCl, 0.01% Triton X-100, 5 mM Glutathione, pH 7.5) was added to each well. Plates were centrifuged at 1000 rpm and incubated for 20 min at rt in dark. The plates were read on a PHERAstar® reader with excitation 340 nm and emission 460 nm. Data were analyzed in Genedata Screener®. IC50values were determined by plotting % inhibition versus log compound concentration and using a one site dose response model. Raw data signals were normalized using 0.5% DMSO as 0% control and Reference Compound B (i.e., (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-7-methylquinoline-4-carboxamide) at 50 μM as 100% inhibitor control. Data for the compounds tested are reported in Table 3. TABLE 3PREPExampleIC50(nM)11410217003690411005220620075108140099801094011140125101338014120015210016160017160185701916020430219322470231602451025370265802766028580295003051031760324503322034430351700364003727038480395004028041640428804310004454045190461104727048170493405054051130524005317054300554405622057160581805930608661390622806340064610651006649067120068170069200070110071140072190073180074230075490765107747078890796308015008117008293083640847108572086360877608812008913009011009126092240093190094180095740968809789098790991100100170101380102680103580104650105590106520107110010810001094400110110011112001123600113490114230115130116230117170118360119761202512188122840123740124220125120126260127130128110129330130540131360132370013337013462013550013630013734013847013910001408401414601425301434601444601452501464501475701484201494701502000151100015269015354015459015538015631015712001585601592801602101614701622801639801642401656701661700167100016863016944017026017127017254017394017487175760176560177360178120179470180590181160018221018314018411001851301861501873818843018944019069019155019220019336019446019512001968419719001984101991702001302016102025502033500204920205640206330207430208210209540210220211110212330213470214280215250216190217310218390219230220390221410222490223862246702254702266602276002281200229320230600B25.2C37.11IC50is reported after single measurement (n = 1) or as geometric mean for multiple measurements (n = 2-9).2Reference Compound B: (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-7-methylquinoline-4-carboxamide3Reference Compound C: (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)-quinoline-4-carboxamide Example 233: hDPP Inhibition Assays A. hDPP7 Inhibition Assay Compounds were tested in a biochemical inhibition assay using human dipeptidylpeptidase 7 (hDPP7) enzyme at 15 nM FAC (BPS Bioscience, #80070) and the substrate Ala-Pro-amino-methylcoumarin (BPS Bioscience, #80305) at 5 μM FAC. The enzymatic reactions were conducted in duplicate at rt for 30 min in 50 μL DPP assay buffer (BPS Bioscience, #80300). Compound solutions (in DMSO) at 10 CR, 3 fold dilution series were prepared in assay buffer ten-fold higher than the final concentration, and 5 μL of the dilution was added to a 50 μL reaction so that the highest compound concentration was 100 μM FAC and the concentration of DMSO was 1% in all wells. The plates were read on a Tecan Infinite M1000 microplate reader with excitation 340 nm and emission 460 nm. Data were analyzed in Graph Pad Prism. IC50values were determined by plotting % inhibition versus log compound concentration and using a one site dose response model. Raw data signals were normalized using 1% DMSO as 0% control and no enzyme as 100% inhibitor control. Data for the compounds tested are reported in Table 4. B. hDPP8 Inhibition Assay Compounds were tested in a biochemical inhibition assay using human dipeptidylpeptidase 8 (hDPP8) enzyme at 1.5 nM FAC (BPS Bioscience, #80080) and the substrate Ala-Pro-amino-methylcoumarin (BPS Bioscience #80305) at 5 μM FAC. The enzymatic reactions were conducted in duplicate at rt for 30 min in 50 μL DPP assay buffer (BPS Bioscience, #80300). Compound solutions (in DMSO) at 10 CR, 3 fold dilution series were prepared in assay buffer ten-fold higher than the final concentration, and 5 μL of the dilution was added to a 50 μL reaction so that the highest compound concentration was 100 μM FAC and the concentration of DMSO was 1% in all wells. The plates were read on a Tecan Infinite M1000 microplate reader with excitation 340 nm and emission 460 nm. Data were analyzed in Graph Pad Prism. IC50values were determined by plotting % inhibition versus log compound concentration and using a one site dose response model. Raw data signals were normalized using 1% DMSO as 0% control and no enzyme as 100% inhibitor control. Data for the compounds tested are reported in Table 4. C. hDPP9 Inhibition Assay Compounds were tested in a biochemical inhibition assay using human dipeptidylpeptidase 9 (hDPP9) enzyme at 0.4 nM FAC (BPS Bioscience, #80090) and the substrate Ala-Pro-amino-methylcoumarin (BPS Bioscience #80305) at 5 μM FAC. The enzymatic reactions were conducted in duplicate at rt for 30 min in 50 μL DPP assay buffer (BPS Bioscience, #80300). Compound solutions (in DMSO) at 10 CR, 3 fold dilution series were prepared in assay buffer ten-fold higher than the final concentration, and 5 μL of the dilution was added to a 50 μL reaction so that the highest compound concentration was 100 μM FAC and the concentration of DMSO was 1% in all wells. The plates were read on a Tecan Infinite M1000 microplate reader with excitation 340 nm and emission 460 nm. Data were analyzed in Graph Pad Prism. IC50values were determined by plotting % inhibition versus log compound concentration and using a one site dose response model. Raw data signals were normalized using 1% DMSO as 0% control and no enzyme as 100% inhibitor control. Data for the compounds tested are reported in Table 4. TABLE 4hDPP7hDPP8hDPP9ExampleIC50(nM)1IC50(nM)1IC50(nM)18>10000033004009>100000510064027>10000043001100673300013000190068>1000006600150069>100000750095070>1000004700140071>1000005900190072>1000006600250080>10000035001400110>100000430250111>100000950410151>10000038002000163>10000039001500171>10000030006301IC50is reported after single measurement (n = 1). Example 234: Metabolic Stability Assays A. Aldehyde Oxidase (AO) Metabolism Assay 1 AO-mediated metabolism was measured essentially as described in Drug Metab. Disp. 2010, 38,1322. Human liver cytosol (Corning life sciences, UltraPool Human Cytosol 150, Product 452115) in phosphate buffer, pH 7.4, was pre-incubated for 5 min at 37° C. shaking at 900 rpm. The reactions were initiated by addition of pre-diluted test compounds including positive control, zaleplon, and incubated at 37° C. with final conditions 2.5 mg/mL human liver cytosolic fraction, 1 μM test compound, 0.01% DMSO and 0.09% MeCN. The samples were incubated for 120 min with time points taken at 0, 10, 30, 60, 90 and 120 min. The aliquots (25 μL) were precipitated with 100 μL MeCN containing internal standard (4-((2′-(1H-tetrazol-5-yl)-[1,1′-biphenyl]-4-yl)methoxy)-2-ethylquinoline, (J Med Chem1992, 35, 4027), centrifuged at 3500 rpm for 10 min and the supernatant diluted 1 in 7 (v/v) with ultra-pure HPLC water before analysis by LC-MS/MS. All incubations were carried out in duplicate. The in vitro elimination rate constant corresponding to parent compound depletion was determined for each reaction using the 1storder decay calculation in Microsoft Excel Sheet. In some cases the experiment was conducted additionally in presence of an aldehyde oxidase inhibitor: The cytosol mix was pre-incubated with 3 μM raloxifene shaking at 900 rpm for 5 min at 37° C. prior to addition of test compound. Data for the compounds tested are reported in Table 5. B. Aldehyde Oxidase (AO) Metabolism Assay 2 AO-mediated metabolism was measured essentially as described in Drug Metab. Disp. 2010, 38,1322. Human liver cytosol (BioreclamationIVT, stored at −80° C. prior to use, protein concentration 2.5 mg/mL) and 0.1 M phosphate buffer (with 0.1 mM EDTA) pH 7.4 is pre-incubated at 37° C. The reaction was initiated by addition of test compound (final substrate concentration 1 μM, final DMSO concentration 0.3% and final incubation volume 500 μL). Phthalazine (known to be metabolized by AO) was used as a control compound. Test compounds were incubated for 0, 5, 15, 30, 60 and 120 min. The reactions were stopped by removing an aliquot of incubate into organic solvent containing internal standard at the appropriate time points. The termination plates were centrifuged at 2500 rpm for 30 min at 4° C. to precipitate the protein. Sample supernatants were combined in cassettes of up to four compounds and analyzed using generic LC MS/MS conditions. From a plot of ln peak area ratio (compound peak area/internal standard peak area) against time, the gradient of the line was determined. Subsequently, half-life and intrinsic clearance were calculated using the equations below: Elimination rate constant(k)=(−gradient) Half-life(t1/2)(min)=0.693/k Intrinsic clearance(CLint)(μL/min/mg protein)=V×0.693/t1/2where V=Incubation volume (μL)/protein (mg) The percentage of the parent compound remaining at each time point, along with the intrinsic clearance value (CLint), half-life and standard error of the CLintwere reported. Data for the compounds tested are reported in Table 5. TABLE 5AO-assay 1AO-assay 2ExampleCLint(μL/min/mg)1CLint(μL/min/mg)180.509<0.50220.07270.12401.1471.367<0.500.2568<0.5069<0.50710.5610701290.761510.321630.30170<0.500.18171<0.50172<0.501730.581750.60196<0.502050.68Compound A23.8Compound C35.31CLintis reported after single measurement (n = 1).2Compound A ((S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-1-yl)-2-oxoethyl)quinoline-4-carboxamide,J Med Chem2014, 57, 3053)3Compound C (R)-N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)quinoline-4-carboxamide C. Human Liver Microsomes (HLM) Metabolic stability in HLM was measured as described inJ Comput Aided Mol Des2015, 29, 795. Data for the compounds tested are reported in Table 6. D. Rat Hepatocytes (rHep) Metabolic stability in rat hepatocytes was measured as described inJ Comput Aided Mol Des2015, 29, 795. Data for the compounds tested are reported in Table 6. E. Human Hepatocytes (hHep) Metabolic stability in human hepatocytes was measured as described inXenobiotica2010, 40, 637. Data for the compounds tested are reported in Table 6. TABLE 6HLMrHephHepCLint1CLint2CLint3Example(μL/min/mg)(μL/min/1E6)(μL/min/1E6)1144.9<12223<13752845014593326NV1007312.68<32.7<19214.3<1101304111<32.5126.4<113125.414<3<115431116<3<1174816182263.7191906320290120211517<12221142.9232527241420<125231426317.627168.3<1.428<3132.629184.5<130<37.2<13145143240194.133924434383335636436481937663038>300>3003912031402315<141301242217.62.743369.1442201604526016046190724732334889514958485013201.7511424522221531207154574955556356>30016057>300>300581407259>300916021012061472662314663180306456296515011066<3<167<5.23.4<168125.2<1695.62.6<170185.927174<1729.72.7<1733211742813<17596.7764.23.5777.15.47823121.279216.2807.52<181178.182391183709.28464268526035861604487521288228.6<189167.1<190100204.691<3892141.7<193<32.6947.12.2<19563139671159732199826131.899168.11.9100NV2801018.62.310212101.2103542310413028105180181064721107428.12.81085312109110393.311120<1112<3<1<1113<311<1.5114170361151202011698151171702911841211196827120100271214313122<3<1123327.11.61241214125147.41261610127118.21282711129<31.413054111319716132388.5133408.813421074135236.8136<34.8137701713820331397213140327.71415411142436.714340131441471454010146114.3147172.91485312149567150495151104.5<1152238.2153<3<1154<32.7155193.21561401201577.3<1<1158174.7159614716045331616.78.5162443.41635.6<1<1164228.1165<3<1166<3<1<1167<3<1168169188.81704.94.21.317173.1<1172223.7<1173122.2174127.2175109.71769215177137.4178167.2179140471801103181<32<1182100261834919184<3<1<1185581518670241871901401883.11.8<118911<1190<3<1191945.3192282.919353111947844195<31.8196<34.7197155.31982022199>300250200>300>300201<3<12027.7<1203122.920481.82052617<1.120646130207<31.4208721220917052210220250211200190212110572133119214271521583.92165817217>30037218371821916075220<34.4221<33.8222404.22235751224<3<1225110372269444227172.8228<39<1229<315230<32.81CLintis reported after single measurement (n = 1) or as an average for multiple measurements (n = 2-3).2CLintis reported after single measurement (n = 1) or as geometric mean for multiple measurements (n = 2-3).3CLintis reported after single measurement (n = 1) or as geometric mean for multiple measurements (n = 2). Example 235: Caco-2 Cell Permeability Caco-2 cell permeability was measured as described inMol Pharm2017, 14, 1601. Data for the compounds tested are reported in Table 7. TABLE 7Caco2Caco2Caco2BidirectionalBidirectionalCaco2AB(ABBA)(ABBA)BidirectionalIntrinsicA to BB to A(ABBA)Papp1Papp1Papp1EffluxExample(1E−6 · cm/s)(1E−6 · cm/s)(1E−6 · cm/s)Ratio14.7296.12230.98313241.6281851.9321770.3420598171.828169181.92312110.349.829120.291965140.152.416150.301345160.164.22618130.903134213.7236.222416.9263.8243.5339.327304.9295.928211.6201229263.7371030153.1309.6328.7283.3337.5192.540203289.34116422.83312500.8327326761191968162.5218.3697.60.792228701.42618710.782330721.32419741.85329750.771721760.642031770.571629781.22721791.13329800.8125318112524871.4292188131.32923895.40.381847901.21916910.903033920.124.235930.331750940.154.429970.522345988.60.711825990.46214610290.9721221070.7825321081.422161110.171061112<0.240.5136113201.923121220.201050123140.17291701240.6539591280.5726451290.3921541354.4296.51385.5325.81472.740151519.52.630111523.2257.81530.8223281540.3212371554.2276.5157229151638.70.9922221650.5116620.5613241670.139.574170101.424171713.80.2722811720.2621841730.18241301740.5425461750.526.4121771.32217181<0.200.243.3131840.100.250.783.11880.280.183.7211890.2913461900.327.6241920.5139761950.404.8121960.6310171983.436112014.82025.22030.408202040.448.519205253.4236.92072.330132150.4829592160.6240652200.2916562241.22280.925270.92290.722320.72302341721Pappis reported after single measurement (n = 1) or as an average for multiple measurements (n = 2-4). Example 236: Kinetic Solubility Kinetic solubility was measured as described inComput Aided Mol Des2015, 29, 795. Data for the compounds tested are reported in Table 8. TABLE 8SolubilityExample(μM)11>1000283370045551161207>1000868093101027011170128301327148301589016170172501893194802090021390224602363245002513026152772028732956030>100031180322403385347703560036<9837210383339224029041620428504352441404534046190475748184913503351565257532.654135511565.957150585.85940601606167062640636906413065496621067>8606818069>1000705807199072>100073310741807593076>1000778507819079610809808190082970838408494085800866408791088>9808991090>100091>10009281093300949209596096>10009795098>10009980010036101>1000102>1000103>1000104930105>1000106700107>1000108880109110950111330112670113711144.51150.59116490117251182011911200.9212144122>10001232801243812538126400127120128960129581301613114013213133<38013471135310136211377413826013950140391411401421.814333144181455.314624147401483.414913015077151850152810153930154850155400156100157621588.91595501601.81618416214016316016464165>1000166>910167240168169261701117132017210017332174800175640176800177401786179110180170181>100018223018319184930185718613018719188990189840190>1000191400192530193940194480195830196700197619838199852008120190202140203540204480205270206262072802081.620997021015021111021216213100214230215350216710217410218<0.162192.5220460221360222120223<1.6224502252.22264.5227852228879229>1000230>10001Solubility is reported after single measurement (n = 1) or as an average for multiple measurements (n = 2-3). Example 237: FAP Target Engagement Enzyme Activity in Mouse Plasma The effect of test compound on FAP enzyme activity in mouse plasma was evaluated in an enzymatic assay using the Fibroblast Activation Protein alpha (FAP) specific fluorogenic substrate dipeptide-Coumarin, Ala-Pro-AMC, (ARI-3144). In this assay, FAP cleaves Ala-Pro-AMC to release free AMC which is measured as a fluorescent signal that correlates with enzyme activity. Male C57Bl/6 mice (Charles River, Germany), 8 weeks of age, were single housed in a temperature-controlled room with a 12-hour light/dark cycle (06:00-18:00 light). The mice had ad libitum access to water and rodent chow diet (R70, Lactamin, Kimstad, Sweden), and were acclimated for 5 days upon arrival. After acclimation, all mice received a single oral dose of test compound (3 or 10 mg/kg). Blood samples for whole blood compound exposure measurements were taken at 0.25, 0.5, 1, 2, 4, 8 and 24 h post oral dosing. Samples were collected in EDTA capillary tubes (20 μL, K2E, REF 19.447) and were transferred to a 96-deep well plate (NUNC, Thermo Discher Scientific) and stored at −20° C. until further analyses were performed. Blood samples for plasma FAP enzyme activity measurements were taken at 0, 0.25, 0.5, 1, 2, 8 and 24 h post dosing. 25 μL of whole blood was collected in EDTA Microvette® CB 300 (K2E, REF 16.444.100) tubes, and were centrifuged at 4,000×g for 5 min. 10 μL of plasma was then transferred to PCR tubes and stored at −20° C. until further analysis was performed. All blood samples were taken by vena saphena puncture. Recombinant human FAP (PB-17-1837, construct PL-17-0278, cd33-FAP (27-757)-6His, Mw85926 Da) was used as a standard for this assay. Protein was secreted from Sf21 cells (insect cells) in media, purified with affinity (batch mode, Ni excel resin) and size exclusion chromatography (Superdex200), concentrated and aliquoted to be frozen in liquid N2for storage at −80° C. Recombinant FAP was diluted in protein buffer (25 mM Tris/HCl, pH 7.6, 150 mM NaCl, 5% glycerol, 1 mM EDTA, 0.25 mM TCEP) and 5 μL aliquots (0.1 mg/mL, 1.15 μM) were stored at −80° C. Standards were prepared using 2-fold dilution steps, 8 concentrations, 4 replicates (FAC: 1.2 nM, 0.6 nM, 0.3 nM . . . ). The plates were read on a Beckman Paradigm reader with excitation 360 nm and emission 465 nm. Fluorescence measurements were performed with kinetic read every 5 minutes for 60 min at room temperature. Data were analyzed in Excel (IDBS XLfit Add-In) using a Linear Regression (y=k*x+m) model to prepare a human recombinant FAP standard curve. On the day of the assay, plasma was diluted (1:2) to 20 μL volume in buffer (PBS, 0.1% BSA) and 7.5 μL was transferred to the assay plate (384-well black, fluotrack PS, Greiner 781076). Ala-Pro-AMC (stock solution in 10 mM DMSO) was diluted in buffer (PBS, 0.1% BSA) to 150 μM concentration (180 μL stock solution to 12 mL buffer) and 7.5 μL added to the assay plate followed by a pipetting mix. The plates were read on a Beckman Paradigm reader with excitation 360 nm and emission 465 nm. Fluorescence measurements were performed with kinetic read every 5 minutes for 60 minutes at room temperature. As noted above, FAP cleaves Ala-Pro-AMC to release free AMC which is measured as a fluorescent signal. The in vivo potency IC50of each test compound was then estimated by relating the plasma exposure C of the compound to target engagement E in plasma using the following equation: E=E0(1-Imax⁢CIC50+C) where E0is the FAP baseline in plasma prior to dosing and Imaxis the maximum effect of the compound. Data from each target engagement experiment were considered separately and therefore slightly different estimates of FAP baseline for each compound were obtained. Full inhibition was achieved for all tested compounds at the earlier timepoints and therefore the parameter Imaxwas fixed to 1 for all compounds. The parameter estimation was done in Phoenix WinNonlin Certara build 8.1.0.3530 with the algorithm ‘Naïve pooled’ as parameter estimation method. In vivo IC50estimates for the test compounds are reported in Table 9. TABLE 9ExampleIn vivo Mouse IC50(nM)1271.4670.186819692.1712.8727.2809.01132.01513.31633.01691.41708.71712.52051.2 Although specific embodiments and examples have been described above, these embodiments and examples are only illustrative and do not limit the scope of the disclosure. Changes and modifications can be made in accordance with ordinary skill in the art without departing from the disclosure in its broader aspects as defined in the following claims. For example, any embodiment described herein can be combined with any other suitable embodiment described herein to provide additional embodiments. As will be understood by the skilled artisan, all numbers, including those expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth, are approximations and understood as being modified in all instances by the term “about.” These values can vary depending upon the desired properties sought to be obtained by those skilled in the art utilizing the present teachings of the present disclosure. It is also understood that such values inherently contain variability necessarily resulting from the standard deviations found in their respective testing measurements. One skilled in the art will also readily recognize that where members are grouped together in a common manner, such as in a Markush group, the present disclosure encompasses not only the entire group listed as a whole, but each member of the group individually and all possible subgroups of the main group. Additionally, for all purposes, the present disclosure encompasses not only the main group, but also the main group absent one or more of the group members. The present disclosure also envisages the explicit exclusion or disclaimer of one or more of any of the group members in the claimed disclosure. As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof as well as the individual values making up the range, particularly integer values. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. For example, the range C(1-6), includes the subranges C(2-6), C(3-6), C(3-5), C(4-6), etc., as well as C1(methyl), C2(ethyl), C3(propyl), C4(butyl), C5(pentyl) and C6(hexyl) individually. As will also be understood by one skilled in the art, all language such as “up to,” “at least,” “greater than,” “less than,” “more than,” “or more” and the like include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. In the same manner, all ratios disclosed herein also include all subratios falling within the broader ratio. Reference to a “step” in this disclosure is used for convenience purposes only and does not categorize, define or limit the disclosure as set forth herein.
883,086
11858925
DEFINITIONS Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments described herein, some preferred methods, compositions, devices, and materials are described herein. However, before the present materials and methods are described, it is to be understood that this invention is not limited to the particular molecules, compositions, methodologies or protocols herein described, as these may vary in accordance with routine experimentation and optimization. It is also to be understood that the terminology used in the description is for the purpose of describing the particular versions or embodiments only, and is not intended to limit the scope of the embodiments described herein. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. However, in case of conflict, the present specification, including definitions, will control. Accordingly, in the context of the embodiments described herein, the following definitions apply. As used herein and in the appended claims, the singular forms “a”, “an” and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to “a GAS41 inhibitor” is a reference to one or more GAS41 inhibitors, and so forth. As used herein, the term “comprise” and linguistic variations thereof denote the presence of recited feature(s), element(s), method step(s), etc. without the exclusion of the presence of additional feature(s), element(s), method step(s), etc. Conversely, the term “consisting of” and linguistic variations thereof, denotes the presence of recited feature(s), element(s), method step(s), etc. and excludes any unrecited feature(s), element(s), method step(s), etc., except for ordinarily-associated impurities. The phrase “consisting essentially of” denotes the recited feature(s), element(s), method step(s), etc. and any additional feature(s), element(s), method step(s), etc. that do not materially affect the basic nature of the composition, system, or method. Many embodiments herein are described using open “comprising” language. Such embodiments encompass multiple closed “consisting of” and/or “consisting essentially of” embodiments, which may alternatively be claimed or described using such language. All chemical names of substituents should be interpreted in light of IUPAC and/or the modified nomenclature and with reference to the chemical structures depicted and/or described herein. For compounds described herein, groups and substituents thereof may be selected in accordance with permitted valence of the atoms and the substituents, and such that the selections and substitutions result in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. In accordance with a convention used in the art, the group: is used in structural formulae herein to depict the bond that is the point of attachment of the moiety or substituent to the core or backbone structure. As used herein, the term “subject” broadly refers to any animal, including but not limited to, human and non-human animals (e.g., dogs, cats, cows, horses, sheep, poultry, fish, crustaceans, etc.). As used herein, the term “patient” typically refers to a subject that is being treated for a disease or condition. As used herein, the term “subject at risk for cancer” refers to a subject with one or more risk factors for developing cancer. Risk factors may include, but are not limited to, gender, age, genetic predisposition, environmental exposures, infections, and previous incidents of diseases, lifestyle, etc. As used herein, the term “effective amount” refers to the amount of a compound or composition sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route. As used herein, the terms “administration” and “administering” refer to the act of giving a drug, prodrug, or other agent, or therapeutic treatment to a subject or in vivo, in vitro, or ex vivo cells, tissues, and organs. Exemplary routes of administration to the human body can be through space under the arachnoid membrane of the brain or spinal cord (intrathecal), the eyes (ophthalmic), mouth (oral), skin (topical or transdermal), nose (nasal), lungs (inhalant), oral mucosa (buccal), ear, rectal, vaginal, by injection (e.g., intravenously, subcutaneously, intratumorally, intraperitoneally, etc.) and the like. As used herein, the terms “co-administration” and “co-administering” refer to the administration of at least two agent(s) (e.g., a GAS41 inhibitor and one or more additional therapeutics) or therapies to a subject. In some embodiments, the co-administration of two or more agents or therapies is concurrent. In other embodiments, a first agent/therapy is administered prior to a second agent/therapy. Those of skill in the art understand that the formulations and/or routes of administration of the various agents or therapies used may vary. The appropriate dosage for co-administration can be readily determined by one skilled in the art. In some embodiments, when agents or therapies are co-administered, the respective agents or therapies are administered at lower dosages than appropriate for their administration alone. Thus, co-administration is especially desirable in embodiments where the co-administration of the agents or therapies lowers the requisite dosage of a potentially harmful (e.g., toxic) agent(s), and/or when co-administration of two or more agents results in sensitization of a subject to beneficial effects of one of the agents via co-administration of the other agent. As used herein, the term “pharmaceutical composition” refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for therapeutic use in vitro, in vivo or ex vivo. The terms “pharmaceutically acceptable” or “pharmacologically acceptable,” as used herein, refer to compositions that do not substantially produce adverse reactions, e.g., toxic, allergic, or immunological reactions, when administered to a subject. As used herein, the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers including, but not limited to, phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents, any and all solvents, dispersion media, coatings, sodium lauryl sulfate, isotonic and absorption delaying agents, disintegrants (e.g., potato starch or sodium starch glycolate), and the like. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see, e.g., Martin, Remington's Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, Pa. (1975), incorporated herein by reference in its entirety. As used herein, the term “pharmaceutically acceptable salt” refers to any pharmaceutically acceptable salt (e.g., acid or base) of a compound of the present invention which, upon administration to a subject, is capable of providing a compound of this invention or an active metabolite or residue thereof. As is known to those of skill in the art, “salts” of the compounds of the present invention may be derived from inorganic or organic acids and bases. Examples of acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts. Examples of bases include, but are not limited to, alkali metals (e.g., sodium) hydroxides, alkaline earth metals (e.g., magnesium), hydroxides, ammonia, and compounds of formula NR4+, wherein each R is independently C1-4alkyl, and the like. Examples of salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, undecanoate, and the like. Other examples of salts include anions of the compounds of the present invention compounded with a suitable cation such as Na+, NH4+, and NR4+(wherein each R is independently a C1-4alkyl group), and the like. For therapeutic use, salts of the compounds herein are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound. As used herein, the term “instructions for administering said compound to a subject,” and grammatical equivalents thereof, includes instructions for using the compositions contained in a kit for the treatment of conditions (e.g., providing dosing, route of administration, decision trees for treating physicians for correlating patient-specific characteristics with therapeutic courses of action). “Amino” refers to a —NH2moiety. “Carbonyl” refers to a moiety of formula —C(═O)—. “Carboxy” or “carboxyl” refers to the —CO2H moiety. “Cyano” refers to the —CN moiety. “Hydroxy” or “hydroxyl” refers to the —OH moiety. “Imino” refers to the ═NH moiety. Unless stated otherwise specifically in the specification, an imino group is optionally substituted. “Nitro” refers to the —NO2moiety. “Oxo” refers to the ═O moiety. “Thioxo” refers to the ═S moiety. “Acyl” refers to the group —C(═O)R, where R is selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, and heteroalkyl. Unless stated otherwise specifically in the specification, an acyl group is optionally substituted. “Alkyl” refers to a straight or branched saturated hydrocarbon chain having from 1 to thirty carbon atoms, for example from 1 to 16 carbon atoms (C1-C16alkyl), 1 to 12 carbon atoms (C1-C12alkyl), 1 to 8 carbon atoms (C1-C8alkyl), 1 to 6 carbon atoms (C1-C6alkyl), or 1 to 4 carbon atoms (C1-C4alkyl), e.g., methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, n-heptyl, n-octyl, n-nonyl, n-decyl, n-undecyl, and n-dodecyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted. “Alkenyl” refers to a straight or branched refers to a straight or branched hydrocarbon chain containing from 2 to 30 carbon atoms, for example from 2 to 16 carbon atoms (C2-C16alkenyl), 2 to 12 carbon atoms (C2-C12alkenyl), 2 to 8 carbon atoms (C2-C8alkenyl), 2 to 6 carbon atoms (C2-C6alkenyl), or 2 to 4 carbon atoms (C2-C4alkenyl), and containing at least one carbon-carbon double bond. Representative examples of alkenyl include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, 1,4-pentadienyl, 5-hexenyl, 2-heptenyl, 2-methyl-1-heptenyl, and 3-decenyl. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted. “Alkynyl” refers to a straight or branched hydrocarbon chain containing from 2 to 30 carbon atoms, for example from 2 to 16 carbon atoms (C2-C16alkynyl), 2 to 12 carbon atoms (C2-C12alkynyl), 2 to 8 carbon atoms (C2-C8alkynyl), 2 to 6 carbon atoms (C2-C6alkynyl), or 2 to 4 carbon atoms (C2-C4alkynyl), and containing at least one carbon-carbon triple bond. Representative examples of alkynyl include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, and hexynyl. Unless stated otherwise specifically in the specification, an alkynyl group is optionally substituted. “Alkylene” refers to a divalent group derived from a straight or branched chain hydrocarbon of 1 to 30 carbon atoms (C1-C30alkylene), for example, of 1 to 6 carbon atoms (C1-C6alkylene). Representative examples of alkylene include, but are not limited to, —CH2—, —CH2CH2—, —CH(CH3)—, —CH2CH2CH2—, —CH2CH(CH3)—, —CH2CH2CH2CH2—, —CH2CH(CH3)CH2—, —CH2CH2CH(CH3)—, —CH2CH2CH2CH2CH2—, —CH2CH(CH3)CH2CH2—, —CH(CH3)CH2CH2CH2—, —CH2CH2CH2CH2CH2CH2—, —CH2CH2CH(CH3)CH2CH2—, —CH2CH(CH3)CH2CH2CH2—, and —CH(CH3)CH2CH2CH2CH2—. Unless stated otherwise specifically in the specification, an alkylene group is optionally substituted. “Alkoxy” refers to a moiety of the formula —OR where R is an alkyl group as defined herein, e.g., an alkyl group containing 1 to 12 carbon atoms. Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy and tert-butoxy. Unless stated otherwise specifically in the specification, an alkoxy group is optionally substituted. “Alkenyloxy” refers to a moiety of the formula —OR where R is an alkenyl group as defined herein, e.g., an alkenyl group containing 2 to 12 carbon atoms. Unless stated otherwise specifically in the specification, an alkenyloxy group is optionally substituted. “Alkynyloxy” refers to a moiety of the formula —OR where R is an alkynyl group as defined herein, e.g., an alkynyl group containing 2 to 12 carbon atoms. Unless stated otherwise specifically in the specification, an alkynyloxy group is optionally substituted. “Alkylamino” refers to a moiety of the formula —NHR where R is an alkyl group as defined herein. Unless stated otherwise specifically in the specification, an alkylamino or dialkylamino group is optionally substituted. “Alkylaminoalkyl” refers to an alkyl moiety comprising at least one alkylamino substituent. Unless stated otherwise specifically in the specification, an alkylaminoalkyl group is optionally substituted. “Amide” or “amido” refers to a moiety with formula —C(═O)NRR′ or —NRC(═O)R′, where R and R′ are each independently selected from the group consisting of hydrogen, alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroaryl (bonded through a ring carbon), heteroarylalkyl, heterocyclyl, and heterocyclylalkyl. When the amido moiety is —C(═O)NRR′, R and R′ may optionally be taken together with the nitrogen to which they are attached to form a 4-, 5-, 6-, or 7-membered ring. Unless stated otherwise specifically in the specification, an amido group is optionally substituted. “Amidoalkyl” refers to an alkyl moiety, as defined herein, in which at least one hydrogen atom is replaced with an amido group, as defined herein. Unless stated otherwise specifically in the specification, an amidoalkyl group is optionally substituted. “Aminoalkyl” refers to an alkyl moiety, as defined herein, in which at least one hydrogen atom is replaced with an amino group, as defined herein. The amino group can be substituted on a tertiary, secondary or primary carbon. Unless stated otherwise specifically in the specification, an aminoalkyl group is optionally substituted. “Aryl” refers to an aromatic carbocyclic ring system having a single ring (monocyclic) or multiple rings (bicyclic or tricyclic) including fused ring systems, and zero heteroatoms. As used herein, aryl contains 6-20 carbon atoms (C6-C20aryl), 6 to 14 ring carbon atoms (C6-C14aryl), 6 to 12 ring carbon atoms (C6-C12aryl), or 6 to 10 ring carbon atoms (C6-C10aryl). Representative examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, and phenanthrenyl. Unless stated otherwise specifically in the specification, the term “aryl” is meant to include aryl groups that are optionally substituted. “Arylalkyl” refers to an alkyl group, as defined herein, wherein at least one hydrogen atom is replaced with an aryl group, as defined herein. Exemplary arylalkyl groups include, but are not limited to, benzyl and phenethyl. Unless stated otherwise specifically in the specification, the term “arylalkyl” is meant to include groups that are optionally substituted on the aryl moiety and/or on the alkyl moiety. “Arylene” refers to a divalent aryl group (e.g., phenylene). Unless stated specifically otherwise, an arylene is optionally substituted. “Aryloxy” refers to an —O-aryl moiety. Unless stated otherwise specifically in the specification, an aryloxy is optionally substituted. “Arylamino” refers to a —NRa-aryl moiety, where Rais H or alkyl. Unless stated otherwise specifically in the specification, an arylamino is optionally substituted. “Cycloalkyl” refers to a saturated carbocyclic ring system containing three to ten carbon atoms per ring. The cycloalkyl may be monocyclic, bicyclic, tricyclic, bridged, fused, and/or spirocyclic. Representative examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, adamantyl, bicyclo[2.2.1]heptanyl, bicyclo[3.2.1]octanyl, and bicyclo[5.2.0]nonanyl. Unless stated otherwise specifically in the specification, the term “cycloalkyl” is meant to include cycloalkyl groups that are optionally substituted. “Cycloalkenyl” refers to a non-aromatic monocyclic or multicyclic ring system containing at least one carbon-carbon double bond and preferably having from 5-10 carbon atoms per ring. Exemplary monocyclic cycloalkenyl rings include cyclopentenyl, cyclohexenyl, and cycloheptenyl. Unless stated otherwise specifically in the specification, the term “cycloalkenyl” is meant to include cycloalkenyl groups that are optionally substituted. “Cycloalkylalkyl” refers to an alkyl group, as defined herein, wherein at least one hydrogen atom is replaced with a cycloalkyl group, as defined herein. Unless stated otherwise specifically in the specification, the term “cycloalkylalkyl” is meant to include groups that are optionally substituted on the cycloalkyl moiety and/or on the alkyl moiety. “Cycloalkylalkylamino” refers to a cycloalkylalkyl-NRa— moiety, where Rais H or alkyl and where the cycloalkylalkyl moiety is attached via a carbon atom to nitrogen, wherein the nitrogen functions as a linker to attach the moiety to the remainder of the molecule. Unless stated otherwise specifically in the specification, a cycloalkylalkylamino is optionally substituted. “Cycloalkylalkyloxy” refers to a —O-cycloalkylalkyl moiety, where the cycloalkylalkyl moiety is attached via a carbon atom to oxygen, wherein the oxygen functions as a linker to attach the moiety to the remainder of the molecule. Unless stated otherwise specifically in the specification, a cycloalkylalkyloxy is optionally substituted. “Cycloalkylamino” refers to a —NRa-cycloalkyl moiety, where Rais H or alkyl. Unless stated otherwise specifically in the specification, a cycloalkylamino is optionally substituted. “Cycloalkyloxy” refers to an —O-cycloalkyl moiety. Unless stated otherwise specifically in the specification, a cycloalkyloxy is optionally substituted. “Dialkylamino” refers to a moiety of the formula —NRR where R and R′ are each independently an alkyl group as defined herein. Unless stated otherwise specifically in the specification, an alkylamino or dialkylamino group is optionally substituted. “Dialkylaminoalkyl” refers to an alkyl moiety comprising at least one dialkylamino substituent. Unless stated otherwise specifically in the specification, an alkylaminoalkyl group is optionally substituted. “Halo” or “halogen” refers to fluoro, chloro, bromo, or iodo. “Haloalkyl” refers to an alkyl group, as defined herein, that is substituted by one or more halo atoms, as defined herein, e.g., trifluoromethyl, difluoromethyl, fluoromethyl, trichloromethyl, —CH2CF3, —CH2CHF2, —CH2CH2F, —CHFCF3, —CHFCHF2, —CHFCH2F, —CHFCH3, —CF2CF3, —CF2CHF2, —CF2CH2F, —CF2CH3, —CH2CF2CH3, —CH2CHFCH3, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group is optionally substituted. “Haloalkoxy” refers to an alkoxy group, as defined herein, that is substituted with one or more halo atoms, as defined herein. As used herein, the term “heteroatom” or “ring heteroatom” is meant to include any element other than carbon or hydrogen. Suitable heteroatoms are oxygen (O), nitrogen (N), sulfur (S), and phosphorus (P). “Heteroalkyl” means an alkyl group, as defined herein, in which one or more of the carbon atoms (and any associated hydrogen atoms) are each independently replaced with a heteroatom group such as —NR—, —O—, —S—, —S(O)—, —S(O)2—, and the like, where R is H, alkyl, aryl, cycloalkyl, heteroalkyl, heteroaryl or heterocyclyl, each of which may be optionally substituted. By way of example, 1, 2 or 3 carbon atoms may be independently replaced with the same or different heteroatomic group. Examples of heteroalkyl groups include, but are not limited to, —OCH3, —CH2OCH3, —SCH3, —CH2SCH3, —NRCH3, and —CH2NRCH3, where R is hydrogen, alkyl, aryl, arylalkyl, heteroalkyl, or heteroaryl, each of which may be optionally substituted. Heteroalkyl also includes groups in which a carbon atom of the alkyl is oxidized (i.e., is —C(O)—). “Heteroalkylene” refers to an alkylene group, as defined herein, in which one or more of the carbon atoms (and any associated hydrogen atoms) are each independently replaced with a heteroatom group such as —NR—, —O—, —S—, —S(O)—, —S(O)2—, and the like, where R is H, alkyl, aryl, cycloalkyl, heteroalkyl, heteroaryl or heterocyclyl, each of which may be optionally substituted. By way of example, 1, 2 or 3 carbon atoms may be independently replaced with the same or different heteroatomic group. Heteroalkylene also includes groups in which a carbon atom of the alkyl is oxidized (i.e., is —C(O)—). Examples of heteroalkylene groups include, but are not limited to, —CH2—O—CH2—, —CH2—S—CH2—, —CH2—NR—CH2—, —CH2—NH—C(O)—CH2—, and the like, as well as polyethylene oxide chains, polypropylene oxide chains, and polyethyleneimine chains. “Heteroaryl” refers to an aromatic group having a single ring (monocyclic) or multiple rings (bicyclic or tricyclic), having one or more ring heteroatoms independently selected from O, N, and S. The aromatic monocyclic rings are five- or six-membered rings containing at least one heteroatom independently selected from O, N, and S (e.g. 1, 2, 3, or 4 heteroatoms independently selected from O, N, and S). The five-membered aromatic monocyclic rings have two double bonds, and the six-membered aromatic monocyclic rings have three double bonds. The bicyclic heteroaryl groups are exemplified by a monocyclic heteroaryl ring appended fused to a monocyclic aryl group, as defined herein, or a monocyclic heteroaryl group, as defined herein. The tricyclic heteroaryl groups are exemplified by a monocyclic heteroaryl ring fused to two rings independently selected from a monocyclic aryl group, as defined herein, and a monocyclic heteroaryl group as defined herein. Representative examples of monocyclic heteroaryl include, but are not limited to, pyridinyl (including pyridin-2-yl, pyridin-3-yl, pyridin-4-yl), pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, benzopyrazolyl, 1,2,3-triazolyl, 1,3,4-thiadiazolyl, 1,2,4-thiadiazolyl, 1,3,4-oxadiazolyl, 1,2,4-oxadiazolyl, imidazolyl, thiazolyl, isothiazolyl, thienyl, furanyl, oxazolyl, isoxazolyl, 1,2,4-triazinyl, and 1,3,5-triazinyl. Representative examples of bicyclic heteroaryl include, but are not limited to, benzimidazolyl, benzodioxolyl, benzofuranyl, benzooxadiazolyl, benzopyrazolyl, benzothiazolyl, benzothienyl, benzotriazolyl, benzoxadiazolyl, benzoxazolyl, chromenyl, imidazopyridine, imidazothiazolyl, indazolyl, indolyl, isobenzofuranyl, isoindolyl, isoquinolinyl, naphthyridinyl, purinyl, pyridoimidazolyl, quinazolinyl, quinolinyl, quinoxalinyl, thiazolopyridinyl, thiazolopyrimidinyl, thienopyrrolyl, and thienothienyl. Representative examples of tricyclic heteroaryl include, but are not limited to, dibenzofuranyl and dibenzothienyl. The monocyclic, bicyclic, and tricyclic heteroaryls are connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the rings. Unless stated otherwise specifically in the specification, a heteroaryl group is optionally substituted. “Heteroarylalkyl” refers to an alkyl group, as defined herein, wherein at least one hydrogen atom is replaced with a heteroaryl group, as defined herein. Unless stated otherwise specifically in the specification, a heteroarylalkyl group is optionally substituted. “Heteroarylalkylamino” refers to a heteroarylalkyl-NRa— moiety, where Rais H or alkyl. Unless stated otherwise specifically in the specification, an heteroarylalkylamino is optionally substituted. “Heteroarylalkyloxy” refers to an heteroarylalkyl-O— moiety. Unless stated otherwise specifically in the specification, a heteroarylalkyloxy is optionally substituted. “Heteroarylamino” refers to a —NRa-heteroaryl moiety, where Rais H or alkyl. Unless stated otherwise specifically in the specification, a heteroarylamino is optionally substituted. “Heteroaryloxy” refers to an —O-heteroaryl moiety. Unless stated otherwise specifically in the specification, an heteroaryloxy is optionally substituted. “Heteroarylene” refers to a divalent heteroaryl group. Unless stated specifically otherwise, a heteroarylene is optionally substituted. “Heterocycle” or “heterocyclic” refers to a saturated or partially unsaturated non-aromatic cyclic group having one or more ring heteroatoms independently selected from O, N, and S. means a monocyclic heterocycle, a bicyclic heterocycle, or a tricyclic heterocycle. The monocyclic heterocycle is a three-, four-, five-, six-, seven-, or eight-membered ring containing at least one heteroatom independently selected from O, N, and S. The three- or four-membered ring contains zero or one double bond, and one heteroatom selected from O, N, and S. The five-membered ring contains zero or one double bond and one, two or three heteroatoms selected from O, N and S. The six-membered ring contains zero, one, or two double bonds and one, two, or three heteroatoms selected from O, N, and S. The seven- and eight-membered rings contains zero, one, two, or three double bonds and one, two, or three heteroatoms selected from O, N, and S. Representative examples of monocyclic heterocycles include, but are not limited to, azetidinyl, azepanyl, aziridinyl, diazepanyl, 1,3-dioxanyl, 1,3-dioxolanyl, 1,3-dithiolanyl, 1,3-dithianyl, imidazolinyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl, oxadiazolidinyl, oxazolinyl, oxazolidinyl, oxetanyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl, pyrrolinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydrothienyl, thiadiazolinyl, thiadiazolidinyl, 1,2-thiazinanyl, 1,3-thiazinanyl, thiazolinyl, thiazolidinyl, thiomorpholinyl, 1,1-dioxidothiomorpholinyl (thiomorpholine sulfone), thiopyranyl, and trithianyl. The bicyclic heterocycle is a monocyclic heterocycle fused to a phenyl group, or a monocyclic heterocycle fused to a monocyclic cycloalkyl, or a monocyclic heterocycle fused to a monocyclic cycloalkenyl, or a monocyclic heterocycle fused to a monocyclic heterocycle, or a spiro heterocycle group, or a bridged monocyclic heterocycle ring system in which two non-adjacent atoms of the ring are linked by an alkylene bridge of 1, 2, 3, or 4 carbon atoms, or an alkenylene bridge of two, three, or four carbon atoms. Representative examples of bicyclic heterocycles include, but are not limited to, benzopyranyl, benzothiopyranyl, chromanyl, 2,3-dihydrobenzofuranyl, 2,3-dihydrobenzothienyl, 2,3-dihydroisoquinoline, 2-azaspiro[3.3]heptan-2-yl, azabicyclo[2.2.1]heptyl (including 2-azabicyclo[2.2.1]hept-2-yl), 2,3-dihydro-1H-indolyl, isoindolinyl, octahydrocyclopenta[c]pyrrolyl, octahydropyrrolopyridinyl, and tetrahydroisoquinolinyl. Tricyclic heterocycles are exemplified by a bicyclic heterocycle fused to a phenyl group, or a bicyclic heterocycle fused to a monocyclic cycloalkyl, or a bicyclic heterocycle fused to a monocyclic cycloalkenyl, or a bicyclic heterocycle fused to a monocyclic heterocycle, or a bicyclic heterocycle in which two non-adjacent atoms of the bicyclic ring are linked by an alkylene bridge of 1, 2, 3, or 4 carbon atoms, or an alkenylene bridge of two, three, or four carbon atoms. Examples of tricyclic heterocycles include, but are not limited to, octahydro-2,5-epoxypentalene, hexahydro-2H-2,5-methanocyclopenta[b]furan, hexahydro-1H-1,4-methanocyclopenta[c]furan, aza-adamantane (1-azatricyclo[3.3.1.13,7]decane), and oxa-adamantane (2-oxatricyclo[3.3.1.13,7]decane). The monocyclic, bicyclic, and tricyclic heterocycles are connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the rings. Unless stated otherwise specifically in the specification, a heterocyclyl group is optionally substituted. “Heterocyclylalkyl” refers to an alkyl group, as defined herein, wherein at least one hydrogen atom is replaced with a heterocyclyl group, as defined herein. Unless stated otherwise specifically in the specification, a heterocyclylalkyl group is optionally substituted. “Heterocyclylalkylamino” refers to a heterocyclylalkyl-NRa— moiety, where Rais H or alkyl and where the heterocyclylalkyl moiety is attached via a carbon atom to nitrogen, wherein the nitrogen functions as a linker to attach the moiety to the remainder of the molecule. Unless stated otherwise specifically in the specification, a heterocyclylalkylamino is optionally substituted. “Heterocyclylalkyloxy” refers to a —O-heterocycloalkyl moiety, where the heterocyclylalkyl moiety is attached via a carbon atom to oxygen, wherein the oxygen functions as a linker to attach the moiety to the remainder of the molecule. Unless stated otherwise specifically in the specification, a heterocyclylalkyloxy is optionally substituted. “Heterocyclylamino” refers to a —NRa-heterocyclyl moiety, where Rais H or alkyl and where the heterocyclyl moiety is attached via a carbon atom to nitrogen, wherein the nitrogen functions as a linker to attach the moiety to the remainder of the molecule. Unless stated otherwise specifically in the specification, a heterocyclylamino is optionally substituted. “Heterocyclyloxy” refers to an —O-heterocyclyl moiety, where the heterocyclyl moiety is attached via a carbon atom to oxygen, wherein the oxygen functions as a linker to attach the moiety to the remainder of the molecule. Unless stated otherwise specifically in the specification, a heterocyclyloxy is optionally substituted. “Hydroxyalkyl” refers to an alkyl group comprising at least one hydroxyl substituent. The —OH substituent may be on a primary, secondary, or tertiary carbon. Unless stated otherwise specifically in the specification, a hydroxylalkyl group is optionally substituted. “Sulfonamido” refers to a moiety of the formula —SO2NRR′, wherein where R and R′ are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, and heteroalkyl. R and R′ may optionally be taken together with the nitrogen to which they are attached to form a 4-, 5-, 6-, or 7-membered ring. Unless stated otherwise specifically in the specification, a sulfonamido group is optionally substituted. “Sulfonamidoalkyl” refers to an alkyl group, as defined herein, wherein at least one hydrogen atom is replaced with a sulfonamido group, as defined herein. Unless stated otherwise specifically in the specification, a sulfonamidoalkyl group is optionally substituted. “Thioalkyl” refers to a moiety of the formula —SR where R is an alkyl moiety as defined herein containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group is optionally substituted. “Thiourea” refers to a moiety of the formula —NH—C(S)—NHR where R is selected from hydrogen, alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl, each of which may be optionally substituted. “Thioureaalkyl” refers to an alkyl group, as defined herein, wherein at least one hydrogen atom is replaced with a thiourea group, as defined herein. Unless stated otherwise specifically in the specification, a thioureaalkyl group is optionally substituted. “Urea” refers to a moiety of the formula —NH—C(O)—NHR where R is selected from hydrogen, alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl, each of which may be optionally substituted. “Ureaalkyl” refers to an alkyl group, as defined herein, wherein at least one hydrogen atom is replaced with a urea group, as defined herein. Unless stated otherwise specifically in the specification, a ureaalkyl group is optionally substituted. The term “substituted” used herein refers to replacement of at least one hydrogen atom with any of the above groups (e.g., amino, carboxy, hydroxy, imino, acyl, alkyl, alkoxy, alkylamino, alkylaminoalkyl, amido, aminoalkyl, aminocarbonyl, aryl, arylalkyl, arylalkylamino, arylalkyloxy, arylamino, aryloxy, carboxyalkyl, cyano, cyanoalkyl, cycloalkyl, cycloalkyl, cycloalkylamino, cycloalkylalkyloxy, cycloalkylamino, cycloalkyloxy, halo, haloalkyl, heteroatom, heteroalkyl, heteroaryl, heteroarylalkyl, heteroarylalkylamino, heteroarylalkyloxy, heteroarylamino, heteroaryloxy, heterobicycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclylalkylamino, heterocyclylalkyloxy, heterocyclylamino, heterocyclyloxy, hydroxyalkyl, thioalkyl, alkylene, alkylenecarbonyl, alkenylene, alkenylenecarbonyl, arylene, heteroalkylene, heteroalkylenecarbonyl, heteroarylene, heteroarylenecarbonyl, heterocyclylalkylene, and/or heterocyclylalkylenecarbonyl), wherein the at least one hydrogen atom is replaced by a bond to a non-hydrogen atom such as, but not limited to: a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups such as alkyl sulfone groups, sulfonyl groups such as sulfonamide groups and sulfonylalkyl groups such as sulfonylmethane, and sulfoxide groups such as alkyl sulfoxide groups; a nitrogen atom in groups such as amino, amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; a phosphorus atom in groups such as dialkylphosphine oxide groups; and other heteroatoms in various other groups. “Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a carbon atom or a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. “Substituted” includes any of the above groups in which one or more hydrogen atoms are replaced with —NRgRh, —NRgC(═O)Rh, —NRgC(═O)NRgRh, —NRgC(═O)ORh, —NRgSO2Rh, —OC(═O)NRgRh, —ORg, —SRg, —SORg, —SO2Rg, —OSO2Rg, —SO2ORg, ═NSO2Rg, —SO2NRgRh, —C(═O)Rg, —C(═O)ORg, —C(═O)NRgRh, —CH2SO2Rg, or —CH2SO2NRgRh, where Rgand Rhare independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heteroalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl. “Substituted” further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, carbonyl, carboxy, cyano, hydroxyl, imino, nitro, oxo, thioxo, acyl, alkyl, alkoxy, alkylamino, alkylaminoalkyl, amide, aminoalkyl, aminocarbonyl, aryl, arylalkyl, arylalkylamino, arylalkyloxy, arylamino, aryloxy, bicycloalkyl, carboxyalkyl, cyanoalkyl, cycloalkyl, cycloalkylalkyl, cycloalkylamino, cycloalkyloxy, cycloalkylamino, cycloalkyloxy, halo, haloalkyl, heteroatom, heteroalkyl, heteroaryl, heteroarylalkyl, heteroarylalkylamino, heteroarylalkyloxy, heteroarylamino, heteroaryloxy, heterobicycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclylalkylamino, heterocyclylalkyloxy, heterocyclylamino, heterocyclyloxy, hydroxyalkyl, N-heteroaryl, N-heterocyclyl, thioalkyl, alkylene, alkylenecarbonyl, alkenylene, alkenylenecarbonyl, arylene, heteroalkylene, heteroalkylenecarbonyl, heteroarylene, heteroarylenecarbonyl, heterocyclylalkylene, heterocyclylalkylenecarbonyl, methylidene, trimethylsilanyl, dialkylphosphine oxide, —OR, —SR, —OC(O)—R, —N(R)2, —C(O)R, —C(O)OR, —C(O)N(R)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)S(O)tR (where t is 1 or 2), —S(O)tOR (where t is 1 or 2), —S(O)tN(R)2(where t is 1 or 2), —PO(R)2, or —PO(OR)2group, where each R is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl group. In addition, each of the foregoing substituents is optionally substituted with one or more of the above substituents. The term “optionally substituted,” as used herein, means that the referenced group (e.g., alkyl, cycloalkyl, etc.) may or may not be substituted with one or more substituents. DETAILED DESCRIPTION Provided herein are small molecules that bind to GAS41 and inhibit GAS41 activity, and methods of use thereof for the treatment of cancer. Proteins recognizing post-transcriptional modifications in histone proteins play a key role in transcriptional regulation (Allis 2016). The YEATS domain containing proteins belong to a family of epigenetic reader proteins and include four human paralogs: ENL, YEATS2, AF9 and GAS41. Biochemical studies have revealed that YEATS domains bind to chromatin by recognizing histones with acetylated or crotonylated lysine side chains. Previous studies reported molecular details of GAS41 YEATS-mediated histone acetyl- or crotonyl-lysine recognition events (Cho et al.ACS Chem. Biol.13, 2739-2746 (2018)). GAS41 YEATS demonstrates site-specific recognition of acetylated- and crotonylated-histone H3 peptides, albeit with modest mid μM affinities (id.). Structural analysis revealed that acylated lysine binds in a channel on GAS41 YEATS domain that may constitute a site for targeting with small molecule inhibitors. Compounds disclosed herein are shown to be low and sub-μM GAS41 YEATS domain inhibitors. GAS41 is dimeric in cells and can recognize di-acylated histone peptides with enhanced affinity via bivalent binding mode. Accordingly, some of the compounds disclosed herein are dimeric GAS41 inhibitors that exhibit enhanced potency and demonstrate activity in non-small cell lung cancer (NSCLC) cells. In some embodiments, the compounds described herein find use in the treatment or prevention of cancer (e.g., brain cancer, sarcoma, colorectal cancer, lung cancer, or gastric cancer) and/or the alleviation of symptoms associated therewith. In some embodiments, provided herein are pharmaceutical compositions comprising a compound described and/or within the scope herein. In some embodiments, pharmaceutical compositions comprising a compound described and/or within the scope herein are administered to a subject to treat cancer (e.g., brain cancer, sarcoma, colorectal cancer, lung cancer, or gastric cancer). Provided herein are compounds of formula (I): or a pharmaceutically acceptable salt thereof, wherein:R1is selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, alkynyl, hydroxy, alkoxy, thioalkyl, halogen, haloalkyl, carboxy, acyl, amido, cyano, sulfonyl, and hydrogen;X is —C(O)—, —C(S)—, —CH2—, or —SO2—, or is absent;Y is —NRa— or —O—;Rais selected from hydrogen, alkyl, haloalkyl, heteroalkyl, cycloalkyl, hydroxyalkyl, and aminoalkyl, or Rais taken together with the nitrogen atom to which it is attached to form a fused ring with A, or Raand R1together with the atoms to which they are attached together form an optionally substituted heterocyclic ring;Z is absent or is —CRbRc—;Rband Rcare each independently selected from hydrogen and alkyl;A is a five-membered heteroaryl;Q is a four-, five-, or six-membered heterocyclyl;R2is selected from hydrogen, halo, alkyl, amino, and hydroxy;R3is selected from hydrogen, halo, —ORd, —NReRf, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, and a group of formula: wherein B is aryl or heteroaryl; J is absent or is —CH2—, —O—, —S—, or —NH—; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, 4, or 5; and Rgand Rhare each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;or R2and R3are taken together with the carbon atom(s) to which they are attached to form a ring selected from aryl, heteroaryl, cycloalkyl, and heterocycle; or R2and R3are taken together with the carbon atom to which they are attached to form an alkenyl group; andRd, Re, and Rfare each independently selected from hydrogen, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, heteroalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl;wherein each alkyl, alkenyl, alkynyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, heterocyclyl, and heterocyclylalkyl is independently optionally substituted with 1, 2, 3, 4, or 5 substituents,with the proviso that when Z is —CRbRc—, R1is not cycloalkyl. In some embodiments, provided herein are compounds of formula (I): or a pharmaceutically acceptable salt thereof, wherein:R1is selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, alkynyl, hydroxy, alkoxy, thioalkyl, halogen, haloalkyl, carboxy, acyl, amido, cyano, sulfonyl, and hydrogen;X is —C(O)—, —C(S)—, —CH2—, or —SO2—, or is absent;Y is —NRa— or —O—;Rais selected from hydrogen, alkyl, haloalkyl, heteroalkyl, cycloalkyl, hydroxyalkyl, and aminoalkyl, or Rais taken together with the nitrogen atom to which it is attached to form a fused ring with A;Z is absent or is —CRbRc—;Rband Rcare each independently selected from hydrogen and alkyl;A is a five-membered heteroaryl;Q is a four-, five-, or six-membered heterocyclyl;R2is selected from hydrogen, halo, alkyl, amino, and hydroxy;R3is selected from hydrogen, halo, —ORd, —NReRf, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, and a group of formula: wherein B is aryl or heteroaryl; J is absent or is —CH2—, —O—, —S—, or —NH—; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, 4, or 5; and Rgand Rhare each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;or R2and R3are taken together with the carbon atom(s) to which they are attached to form a ring selected from aryl, heteroaryl, cycloalkyl, and heterocycle; or R2and R3are taken together with the carbon atom to which they are attached to form an alkenyl group; andRd, Re, and Rfare each independently selected from hydrogen, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, heteroalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl;wherein each alkyl, alkenyl, alkynyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, heterocyclyl, and heterocyclylalkyl is independently optionally substituted with 1, 2, 3, 4, or 5 substituents,with the proviso that when Z is —CRbRc—, R1is not cycloalkyl. In some embodiments, R1is selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, and alkyl. In some embodiments, R1is selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, and C1-C6alkyl. In some embodiments, R1is selected from heterocyclyl, aryl, arylalkyl, heteroarylalkyl, and C1-C6alkyl. In some embodiments, R1is selected from heterocyclyl (e.g., a monocyclic or bicyclic heterocyclyl having 1 or 2 heteroatoms independently selected from N, O, and S), aryl (e.g., phenyl), arylalkyl (e.g., phenethyl), heteroarylalkyl (e.g., wherein the heteroaryl is a monocyclic heteroaryl having 1 or 2 nitrogen atoms), and C1-C4alkyl (e.g., methyl, ethyl, or n-propyl). In some embodiments, R1is a monocyclic heterocyclyl having 1 or 2 heteroatoms independently selected from N, O, and S. In some embodiments, R1is a monocyclic heterocyclyl having 1 or 2 nitrogen atoms. In some embodiments, R1is pyrrolidinyl. R1may be unsubstituted or substituted with 1, 2, 3, 4, or 5 substituents. For example, in some embodiments, R1is unsubstituted or substituted with 1 or 2 substituents independently selected from C1-C6alkyl, C1-C6alkoxy, halo, hydroxy, amino, amino-C1-C6-alkyl, aryloxy, alkynyloxy, and methylidene. In some embodiments, R1is unsubstituted. In some embodiments, R1is unsubstituted pyrrolidinyl. In some embodiments, R1is selected from: In some embodiments, R1is: In some embodiments, X is selected from —C(O)—, —CH2—, and —SO2—, or is absent. In some embodiments, X is selected from —C(O)—, —CH2—, and —SO2—. In some embodiments, X is selected from —C(O)— and —CH2—. In some embodiments, X is —C(O)—. In some embodiments, Y is —NRa—. In some embodiments, Y is —NRa—, and Rais selected from hydrogen and C1-C6alkyl. In some embodiments, Y is —NRa—, and Rais selected from hydrogen, methyl, and ethyl. In some embodiments, Y is —NRa—, and Rais hydrogen. In some embodiments, Y is O. In some embodiments, Y is —NRa— wherein Rais taken together with the nitrogen atom to which it is attached to form a fused ring with A (e.g., a five-membered or six-membered ring fused with ring A). In some embodiments, Y is —NRa- wherein Rais taken together with the nitrogen atom to which it is attached to form a fused ring with R1(e.g., a bicyclic ring system, such as a 1,7-diazaspiro[4.4]nonane ring system), which ring is optionally substituted (e.g., with an oxo group). In some embodiments, Z is absent, or is selected from —CH2—, —CH(CH3)—, and —C(CH3)2—. In some embodiments, Z is absent or is —CH2—. In some embodiments, Z is absent. In some embodiments, A is a five-membered heteroaryl having 1, 2, or 3 heteroatoms independently selected from N, O, and S. In some embodiments, A is a five-membered heteroaryl having 1 or 2 heteroatoms independently selected from N, O, and S. In some embodiments, A is a five-membered heteroaryl having 1 or 2 heteroatoms independently selected from N and S. In some embodiments, A is selected from thiophene and thiazole. In some embodiments, A is thiophene. In some embodiments, A has formula: wherein E is selected from N and CH. In some embodiments, E is CH. In some embodiments, E is N. In some embodiments, A has formula: In some embodiments, Q is a four-, five-, or six-membered heterocyclyl having one nitrogen atom (i.e. the nitrogen atom indicated in formula (I)), wherein the heterocyclyl is optionally substituted. In some embodiments, Q is selected from azetidinyl, pyrrolidinyl, and piperidinyl. In some embodiments, Q is selected from azetidinyl and pyrrolidinyl. In some embodiments, Q is azetidinyl. In some embodiments, Q is pyrrolidinyl. In some embodiments, R2is selected from hydrogen, halo, amino, and hydroxy. In some embodiments, R2is selected from hydrogen, halo, and hydroxy. In some embodiments, R2is hydrogen. In some embodiments, R3is selected from hydrogen, —ORd, —NReRf, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, and a group of formula: wherein Rdis selected from C1-C6alkyl, C1-C6haloalkyl, phenyl, benzyl, and heteroaryl; Reis hydrogen; and Rfis selected from hydrogen, C1-C6alkyl, and heteroaryl; B is a monocyclic heteroaryl; J is absent; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0 or 1; n is 0, 1, 2, or 3; and Rgand Rhare each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl. In some embodiments, R3is selected from hydrogen, —ORd, —NReRf, phenyl, benzyl, heteroaryl, heteroarylalkyl, and heterocyclyl; wherein Rdis selected from C1-C6alkyl, C1-C6haloalkyl, phenyl, benzyl, and heteroaryl; Reis hydrogen; and Rfis selected from hydrogen, C1-C6alkyl, and heteroaryl. In some embodiments, R3is selected from hydrogen, —ORd, —NReRf, phenyl, benzyl, heteroaryl, heteroarylalkyl, and heterocyclyl; wherein Rdis selected from C1-C6alkyl, C1-C6haloalkyl, phenyl, benzyl, and heteroaryl; Reis hydrogen; and Rfis selected from hydrogen, C1-C6alkyl, and heteroaryl; wherein each heteroaryl is independently a monocyclic or bicyclic heteroaryl having 1 or 2 heteroatoms independently selected from N, S, and O, and wherein each heterocyclyl is independently a monocyclic or bicyclic heterocyclyl having 1 or 2 heteroatoms independently selected from N, S, and O. In some embodiments, R3is a group of formula: In some embodiments, R3is a group of formula: wherein B is a 5-membered monocyclic heteroaryl having 1 or 2 heteroatoms independently selected from N and S; J is absent; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0 or 1; n is 0, 1, 2, or 3; Rgis C1-C6alkyl; and each Rhis independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl. In some embodiments, B is thiazole or thiophene. In some embodiments, m is 0. In some embodiments, J is absent. In some embodiments, C is selected from phenyl and a monocyclic heteroaryl having 1 or 2 nitrogen atoms. In some embodiments, C is selected from phenyl and pyridyl. In some embodiments, at least one Rhis amido or amidoalkyl having formula —(CH2)rC(O)NRiRjor —(CH2)sNRkC(O)Rm, wherein:r and s are each independently selected from 0, 1, and 2;Riand Rkare each independently selected from hydrogen and C1-C6alkyl;Rjis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl;Rmis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl, amino, C1-C6-alkylamino, arylamino, and aryl-C1-C6-alkylamino;wherein each alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl is independently unsubstituted or substituted with 1 or 2 substituents independently selected from halo, C1-C6-alkyl, C1-C6-alkoxy, hydroxy, amino and oxo. In some embodiments, R2and R3, together with the carbon atom(s) to which they are attached, form a ring selected from aryl, heteroaryl, cycloalkyl, and heterocycle, any of which can be optionally substituted (e.g., with 1, 2, or 3 substituents independently selected from alkyl, halo, amino, alkylamino, dialkylamino, alkoxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, aminoalkyl, and amidoalkyl). In some embodiments, R2and R3are substituted on adjacent carbon atoms of ring Q, and are taken together with the carbon atoms to which they are attached to form a phenyl ring that is fused to ring Q, wherein the phenyl ring is optionally substituted. In some embodiments, the phenyl ring is unsubstituted. In some embodiments, R2and R3are substituted on the same carbon atom of ring Q, and are taken together with the carbon atom to which they are attached to form a spiro ring, which is optionally substituted. In some embodiments, R2and R3are substituted on the same carbon atom of ring Q, and are taken together with the carbon atom to which they are attached to form a 4-membered spiro ring selected from cyclobutyl and azetidinyl, each of which is optionally substituted with one substituent selected from —OR′ and heteroaryl, wherein R′ is selected from C1-C6alkyl, aryl and heteroaryl. In some embodiments, the spiro ring is substituted with one substituent selected from —OR′, wherein R′ is selected from methyl, phenyl, and a monocyclic 5- or 6-membered heteroaryl having 1 or 2 heteroatoms independently selected from N and S (e.g., pyridyl). In some embodiments, the spiro ring is substituted with a monocyclic 5- or 6-membered heteroaryl having 1 or 2 heteroatoms independently selected from N and S (e.g., pyridyl or thiazolyl). In some embodiments, R2and R3are substituted on the same carbon atom of ring Q, and are taken together with the carbon atom to which they are attached to form an alkenyl group (e.g., a methylidene group or a substituted version thereof). In some embodiments, the group has a formula selected from: wherein Rx, Ry, and Rzare substituents that are each independently selected from —ORv, aryl, and heteroaryl, wherein Rvis selected from C1-C6alkyl, aryl and heteroaryl. In some embodiments, Rx, Ry, and Rzare independently selected from —ORv, phenyl, and a monocyclic 5- or 6-membered heteroaryl having 1 or 2 heteroatoms independently selected from N and S (e.g., pyridyl or thiazolyl), wherein Rvis selected from C1-C6alkyl (e.g., methyl), aryl (e.g., phenyl), and a monocyclic 5- or 6-membered heteroaryl having 1 or 2 heteroatoms independently selected from N and S (e.g., pyridyl or thiazolyl). In some embodiments, Rxis aryl (e.g., phenyl). In some embodiments, Ryis selected from —ORvand a monocyclic 5- or 6-membered heteroaryl having 1 or 2 heteroatoms independently selected from N and S (e.g., pyridyl or thiazolyl), wherein Rvis selected from C1-C6alkyl (e.g., methyl), aryl (e.g., phenyl), and a monocyclic 5- or 6-membered heteroaryl having 1 or 2 heteroatoms independently selected from N and S (e.g., pyridyl or thiazolyl). In some embodiments, Rzis a monocyclic 5- or 6-membered heteroaryl having 1 or 2 heteroatoms independently selected from N and S, such as a monocyclic 6-membered heteroaryl (e.g., pyridyl). In some embodiments, the group has the formula: wherein B, J, C, Rg, Rh, m, and n are as defined herein. In some embodiments, the group has the formula: wherein Rhand n are as defined herein. For example, in some embodiments, n is 0, 1, 2 or 3; and each Rhis independently selected from C1-C6alkyl, halo, halo-C1-C6-alkyl, amino, amino-C1-C6-alkyl, hydroxy, hydroxy-C1-C6-alkyl, C1-C6alkoxy, amido, amido-C1-C6-alkyl, acyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl. In some embodiments, at least one Rhhas formula —(CH2)rC(O)NRiRjor —(CH2)sNRkC(O)Rm, wherein:r and s are each independently selected from 0, 1, and 2;Riand Rkare each independently selected from hydrogen and C1-C6alkyl;Rjis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl;Rmis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl, amino, C1-C6-alkylamino, arylamino, aryl-C1-C6-alkylamino;wherein each alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl is independently unsubstituted or substituted with 1 or 2 substituents independently selected from halo, C1-C6-alkyl, C1-C6-alkoxy, hydroxy, amino and oxo. In some embodiments, the group is selected from: In some embodiments, the compound of formula (I) is a compound of formula (Ia): or a pharmaceutically acceptable salt thereof, wherein R1and R3have any of the meanings disclosed herein. In some embodiments, the compound of formula (I) is a compound of formula (Ib): or a pharmaceutically acceptable salt thereof, wherein B, J, C, Rg, Rh, m, and n have any of the meanings disclosed herein. In some embodiments, the compound of formula (I) is a compound of formula (Ic): or a pharmaceutically acceptable salt thereof, wherein Rhand c have any of the meanings disclosed herein. Herein, when reference is made to a compound of formula (I) (e.g., to a pharmaceutical composition comprising a compound of formula (I) or a method of treatment using a compound of formula (I)), such reference also includes compounds of formula (Ia), (Ib), and (Ic). In some embodiments, the compound is selected from the compounds shown in Table 1 herein, or a pharmaceutically acceptable salt thereof. Also disclosed herein is a compound of formula (IIa): or a pharmaceutically acceptable salt thereof, wherein:R1and R1′are each independently selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, alkynyl, hydroxy, alkoxy, thioalkyl, halogen, haloalkyl, carboxy, acyl, amido, cyano, sulfonyl, and hydrogen;X and X′ are each independently absent or selected from —C(O)—, —C(S)—, —CH2—, and —SO2—;Y and Y′ are each independently —NRa— or —O—;Rais selected from hydrogen, alkyl, haloalkyl, heteroalkyl, cycloalkyl, hydroxyalkyl, and aminoalkyl, or Rais taken together with the nitrogen atom to which it is attached to form a fused ring with A, or Raand R1together with the atoms to which they are attached together form an optionally substituted heterocyclic ring;Z and Z′ are each independently absent or —CRbRc—;Rband Rcare each independently selected from hydrogen and alkyl;A and A′ are each independently a five-membered heteroaryl ring;Q and Q′ are each independently a four-, five-, or six-membered heterocycle;R2and R2′ are each independently selected from hydrogen, halo, alkyl, amino, and hydroxy;R3and R3′ are each independently selected from aryl, heteroaryl, cycloalkyl, heterocyclyl, and a group of formula: wherein B is aryl or heteroaryl; J is absent or is —CH2—, —O—, —S—, or —NH—; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, or 4; and Rgand Rhare each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;Rd, Re, and Rfare each independently selected from hydrogen, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, heteroalkyl, aryl, arylalkyl, and heteroaryl; andL is a linker;wherein each alkyl, alkenyl, alkynyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, heterocyclyl, and heterocyclylalkyl is independently optionally substituted with 1, 2, 3, 4, or 5 substituents. In some embodiments, disclosed herein is a compound of formula (IIa): or a pharmaceutically acceptable salt thereof, wherein:R1and R1′are each independently selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, alkynyl, hydroxy, alkoxy, thioalkyl, halogen, haloalkyl, carboxy, acyl, amido, cyano, sulfonyl, and hydrogen;X and X′ are each independently absent or selected from —C(O)—, —C(S)—, —CH2—, and —SO2—;Y and Y′ are each independently —NRa— or —O—;Rais selected from hydrogen, alkyl, haloalkyl, heteroalkyl, cycloalkyl, hydroxyalkyl, and aminoalkyl, or Rais taken together with the nitrogen atom to which it is attached to form a fused ring with AZ and Z′ are each independently absent or —CRbRc—;Rband Rcare each independently selected from hydrogen and alkyl;A and A′ are each independently a five-membered heteroaryl ring;Q and Q′ are each independently a four-, five-, or six-membered heterocycle;R2and R2′ are each independently selected from hydrogen, halo, alkyl, amino, and hydroxy;R3and R3′ are each independently selected from aryl, heteroaryl, cycloalkyl, heterocyclyl, and a group of formula: wherein B is aryl or heteroaryl; J is absent or is —CH2—, —O—, —S—, or —NH—; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, or 4; and Rgand Rhare each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;Rd, Re, and Rfare each independently selected from hydrogen, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, heteroalkyl, aryl, arylalkyl, and heteroaryl; andL is a linker;wherein each alkyl, alkenyl, alkynyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, heterocyclyl, and heterocyclylalkyl is independently optionally substituted with 1, 2, 3, 4, or 5 substituents. In some embodiments, R1and R1′ are the same, R2and R2′ are the same, R3and R3′ are the same, X and X′ are the same, Y and Y′ are the same, Z and Z′ are the same, A and A′ are the same, and Q and Q′ are the same. In some embodiments, R1and R1′ are heterocyclyl, which is optionally substituted. In some embodiments, R1and R1′ are monocyclic 4- to 6-membered heterocyclyl having 1 or 2 nitrogen atoms, which is optionally substituted. In some embodiments, R1and R1′ are each a 4- or 5-membered monocyclic heterocyclyl, such as a 4- or 5-membered heterocyclyl having 1 nitrogen atom, which is optionally substituted. In some embodiments, R1and R1′ are pyrrolidine, which is optionally substituted. In some embodiments, R1and R1′ are unsubstituted pyrrolidine. In some embodiments, X and X′ are —C(O)—. In some embodiments, Y and Y′ are —NRa—, and Rais selected from hydrogen and C1-C6alkyl. In some embodiments, Y and Y′ are —NRa—, and Rais selected from hydrogen and methyl. In some embodiments, Y and Y′ are —NRa—, and Rais hydrogen. In some embodiments, Z and Z′ are each absent. In some embodiments, A and A′ are each a five-membered monocyclic heteroaryl having 1 or 2 heteroatoms independently selected from S and N. In some embodiments, A and A′ are selected from thiophene and thiazole. In some embodiments, A and A′ are thiophene. In some embodiments, Q and Q′ are each a four-, five-, or six-membered heterocyclyl having one nitrogen atom (i.e. the nitrogen atom indicated in formula (IIa)). In other words, in some embodiments, Q and Q′ are selected from azetidine, pyrrolidine, and piperidine. In some embodiments, Q and Q′ are selected from azetidine and pyrrolidine. In some embodiments, Q and Q′ are azetidine. In some embodiments, Q and Q′ are pyrrolidine. In some embodiments, R2and R2′ are hydrogen. In some embodiments, R3and R3′ are selected from aryl, heteroaryl, and a group of formula: In some embodiments, R3and R3′are each a group of formula: wherein B is a 5-membered monocyclic heteroaryl having 1 or 2 heteroatoms independently selected from N, S or O; J is absent; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0 or 1; Rgis C1-C6alkyl; n is 0, 1, or 2; and each Rhis independently selected from C1-C6alkyl, halo, C1-C6haloalkyl, amino, amino-C1-C6-alkyl, amido-C1-C6-alkyl, and heterocyclyl. In some embodiments, B is selected from thiazole and thiophene. In some embodiments, B is thiazole. In some embodiments, C is selected from aryl and monocyclic heteroaryl. In some embodiments, C is selected from phenyl and pyridyl. In some embodiments, at least one Rhhas formula —(CH2)rC(O)NRiRjor —(CH2)sNRkC(O)Rm, wherein:r and s are each independently selected from 0, 1, and 2;Riand Rkare each independently selected from hydrogen and C1-C6alkyl;Rjis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl;Rmis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl, amino, C1-C6-alkylamino, arylamino, aryl-C1-C6-alkylamino;wherein each alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl is independently unsubstituted or substituted with 1 or 2 substituents independently selected from halo, C1-C6-alkyl, C1-C6-alkoxy, hydroxy, amino and oxo. In some embodiments, L is a linker comprising one or more groups independently selected from methylene (—CH2—), vinylene (—CH═CH—), acetylene (—C≡C—), ether (—O—), amine (—NH—), alkylamine (—NR—, wherein R is an optionally substituted C1-C6alkyl group), amide (—C(O)NH—), ester (—C(O)O—), carbamate (—OC(O)NH—), sulfonamide (—S(O)2NH—), phenylene (—C6H4—), heteroarylene, heterocyclylene, and any combination thereof. In some embodiments, L is selected from: wherein a, a1, and a2 are each independently selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; b, b1, and b2 are each independently selected from 0, 1, 2, 3, 4, 5, and 6; c, c1, and c2 are each independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; d and e are each independently selected form 0, 1, and 2; each G is independently selected from CH and N; X1and X2are each independently 0 or —NRx, wherein Rxis hydrogen or optionally substituted alkyl; and Y1and Z1are each independently selected from —CH2—, —NH—, and —O—. In some embodiments, the compound is selected from the compounds shown in Table 2 herein, or a pharmaceutically acceptable salt thereof. Also disclosed herein is a compound of formula (IIb) or a pharmaceutically acceptable salt thereof, wherein:R1and R1′are each independently selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, and alkynyl;X and X′ are each independently absent or selected from —C(O)—, —C(S)—, —CH2—, and —SO2—;Y and Y′ are each independently selected from —NRa— or —O—;Rais selected from hydrogen, alkyl, haloalkyl, heteroalkyl, cycloalkyl, hydroxyalkyl, and aminoalkyl, or Rais taken together with the nitrogen atom to which it is attached to form a fused ring with A;Z and Z′ are each independently absent or —CRbRc—;Rband Rcare each independently selected from hydrogen and alkyl;A and A′ are each independently a five-membered heteroaryl ring;Q and Q′ are each independently a four-, five-, or six-membered heterocyclyl;R2and R2′ are each independently selected from hydrogen, halo, alkyl, amino, and hydroxy;R3and R3′ are each independently selected from hydrogen, halo, —ORd, —NReRf, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, and a group of formula: wherein B is aryl or heteroaryl; J is absent or is —CH2—, —O—, —S—, or —NH—; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, or 4; and Rgand Rhare each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;or R2and R3are taken together with the carbon atom(s) to which they are attached to form a ring selected from aryl, heteroaryl, cycloalkyl, and heterocycle; or R2and R3are taken together with the carbon atom to which they are attached to form an alkenyl group;Rd, Re, and Rfare each independently selected from hydrogen, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, heteroalkyl, aryl, arylalkyl, and heteroaryl; andL is a linker;wherein each alkyl, alkenyl, alkynyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, heterocyclyl, and heterocyclylalkyl is independently optionally substituted with 1, 2, 3, 4, or 5 substituents. In some embodiments, R1and R1′ are the same, R2and R2′ are the same, R3and R3′ are the same, X and X′ are the same, Y and Y′ are the same, Z and Z′ are the same, A and A′ are the same, and Q and Q′ are the same. In some embodiments, R1and R1′ are heterocyclyl, which is optionally substituted. In some embodiments, R1and R1′ are monocyclic 4- to 6-membered heterocyclyl having 1 or 2 nitrogen atoms, which is optionally substituted. In some embodiments, R1and R1′ are each a 4- or 5-membered monocyclic heterocyclyl, such as a 4- or 5-membered heterocyclyl having 1 nitrogen atom, which is optionally substituted. In some embodiments, R1and R1′ are pyrrolidine, which is optionally substituted. In some embodiments, R1and R1′ are unsubstituted pyrrolidine. In some embodiments, X and X′ are —C(O)—. In some embodiments, Y and Y′ are —NRa—, and Rais selected from hydrogen and C1-C6alkyl. In some embodiments, Y and Y′ are —NRa—, and Rais selected from hydrogen and methyl. In some embodiments, Y and Y′ are —NRa—, and Rais hydrogen. In some embodiments, Z and Z′ are each absent. In some embodiments, A and A′ are each a five-membered monocyclic heteroaryl having 1 or 2 heteroatoms independently selected from S and N. In some embodiments, A and A′ are selected from thiophene and thiazole. In some embodiments, A and A′ are thiophene. In some embodiments, Q and Q′ are each a four-, five-, or six-membered heterocyclyl having one nitrogen atom (i.e. the nitrogen atom indicated in formula (IIa)). In other words, in some embodiments, Q and Q′ are selected from azetidine, pyrrolidine, and piperidine. In some embodiments, Q and Q′ are selected from azetidine and pyrrolidine. In some embodiments, Q and Q′ are azetidine. In some embodiments, Q and Q′ are pyrrolidine. In some embodiments, R2and R2′ are hydrogen. In some embodiments, R3and R3′are selected from aryl, heteroaryl, and a group of formula: In some embodiments, R3′and R3′are selected from monocyclic and bicyclic heteroaryl having 1, 2, or 3 heteroatoms independently selected from N and S. In some embodiments, R3and R3′are each a group of formula: wherein B is a 5-membered monocyclic heteroaryl having 1 or 2 heteroatoms independently selected from N, S or O; J is absent; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0 or 1; Rgis C1-C6alkyl; n is 0, 1, or 2; and each Rhis independently selected from C1-C6alkyl, halo, C1-C6haloalkyl, amino, amino-C1-C6-alkyl, amido-C1-C6-alkyl, and heterocyclyl. In some embodiments, B is selected from thiazole and thiophene. In some embodiments, B is thiazole. In some embodiments, C is selected from aryl and monocyclic heteroaryl. In some embodiments, C is selected from phenyl and pyridyl. In some embodiments, at least one Rhhas formula —(CH2)rC(O)NRiRjor —(CH2)sNRkC(O)Rm, wherein:r and s are each independently selected from 0, 1, and 2;Riand Rkare each independently selected from hydrogen and C1-C6alkyl;Rjis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl;Rmis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl, amino, C1-C6-alkylamino, arylamino, aryl-C1-C6-alkylamino;wherein each alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl is independently unsubstituted or substituted with 1 or 2 substituents independently selected from halo, C1-C6-alkyl, C1-C6-alkoxy, hydroxy, amino and oxo. In some embodiments, L is a linker comprising one or more groups independently selected from methylene (—CH2—), vinylene (—CH═CH—), acetylene (—C≡C—), ether (—O—), amine (—NH—), alkylamine (—NR—, wherein R is an optionally substituted C1-C6alkyl group), amide (—C(O)NH—), ester (—C(O)O—), carbamate (—OC(O)NH—), sulfonamide (—S(O)2NH—), phenylene (—C6H4—), heteroarylene, heterocyclylene, and any combination thereof. In some embodiments, L is selected from: wherein a, a1, and a2 are each independently selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; b, b1, and b2 are each independently selected from 0, 1, 2, 3, 4, 5, and 6; e, c1, and c2 are each independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; d and e are each independently selected form 0, 1, and 2; each G is independently selected from CH and N; X1and X2are each independently O or —NRx, wherein Rxis hydrogen or optionally substituted alkyl; and Y1and Z1are each independently selected from —CH2—, —NH—, and —O—. In some embodiments, the compound is selected from the compounds shown in Table 2 herein, or a pharmaceutically acceptable salt thereof. Also disclosed herein is a compound of formula (IIc) or a pharmaceutically acceptable salt thereof, wherein:R1is selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, alkynyl, hydroxy, alkoxy, thioalkyl, halogen, haloalkyl, carboxy, acyl, amido, cyano, sulfonyl, and hydrogen;R1′is selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, and alkynyl;X and X′ are each independently absent or selected from —C(O)—, —C(S)—, —CH2—, and —SO2—;Y and Y′ are each independently selected from —NRa— or —O—;Rais selected from hydrogen, alkyl, haloalkyl, heteroalkyl, cycloalkyl, hydroxyalkyl, and aminoalkyl, or Rais taken together with the nitrogen atom to which it is attached to form a fused ring with A, or Raand R1together with the atoms to which they are attached together form an optionally substituted heterocyclic ring;Z and Z′ are each independently absent or —CRbRc—;Rband Rcare each independently selected from hydrogen and alkyl;A and A′ are each independently a five-membered heteroaryl ring;Q and Q′ are each independently a four-, five-, or six-membered heterocycle;R2and R2′ are each independently selected from hydrogen, halo, alkyl, amino, and hydroxy;R3is selected from aryl, heteroaryl, heterocyclyl, and a group of formula: wherein B is aryl or heteroaryl; J is absent or is —CH2—, —O—, —S—, or —NH—; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, or 4; and Rgand Rhare each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;R3′ is selected from hydrogen, halo, —ORd′, —NRe′Rf′, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, and a group of formula: wherein B′ is aryl or heteroaryl; J′ is absent or is —CH2—, —O—, —S—, or —NH—; C′ is selected from aryl, heteroaryl, and heterocyclyl; m′ is 0, 1, 2, 3, or 4; n′ is 0, 1, 2, 3, or 4; and Rg′and Rh′are each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;Rd′, Re′, and Rf′are each independently selected from hydrogen, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, heteroalkyl, aryl, arylalkyl, and heteroaryl; andL is a linker;wherein each alkyl, alkenyl, alkynyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, heterocyclyl, and heterocyclylalkyl is independently optionally substituted with 1, 2, 3, 4, or 5 substituents. Also disclosed herein is a compound of formula (IIc) or a pharmaceutically acceptable salt thereof, wherein:R1is selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, alkynyl, hydroxy, alkoxy, thioalkyl, halogen, haloalkyl, carboxy, acyl, amido, cyano, sulfonyl, and hydrogen;R1′is selected from heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkyl, alkenyl, and alkynyl;X and X′ are each independently absent or selected from —C(O)—, —C(S)—, —CH2—, and —SO2—;Y and Y′ are each independently selected from —NRa— or —O—;Rais selected from hydrogen, alkyl, haloalkyl, heteroalkyl, cycloalkyl, hydroxyalkyl, and aminoalkyl, or Rais taken together with the nitrogen atom to which it is attached to form a fused ring with A;Z and Z′ are each independently absent or —CRbRc—;Rband Rcare each independently selected from hydrogen and alkyl;A and A′ are each independently a five-membered heteroaryl ring;Q and Q′ are each independently a four-, five-, or six-membered heterocycle;R2and R2′ are each independently selected from hydrogen, halo, alkyl, amino, and hydroxy;R3is selected from aryl, heteroaryl, heterocyclyl, and a group of formula: wherein B is aryl or heteroaryl; J is absent or is —CH2—, —O—, —S—, or —NH—; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, or 4; and Rgand Rhare each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;R3′ is selected from hydrogen, halo, —ORd′, —NRe′Rf′, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, and a group of formula: wherein B′ is aryl or heteroaryl; J′ is absent or is —CH2—, —O—, —S—, or —NH—; C′ is selected from aryl, heteroaryl, and heterocyclyl; m′ is 0, 1, 2, 3, or 4; n′ is 0, 1, 2, 3, or 4; and Rg′and Rh′are each independently selected from alkyl, alkenyl, alkynyl, halo, haloalkyl, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, amido, amidoalkyl, sulfonamido, sulfonamidoalkyl, urea, ureaalkyl, thiourea, thioureaalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkoxy, thioalkyl, acyl, carboxy, nitro, oxo, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, cycloalkyl, and cycloalkylalkyl;Rd′, Re′, and Rf′are each independently selected from hydrogen, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, heteroalkyl, aryl, arylalkyl, and heteroaryl; andL is a linker;wherein each alkyl, alkenyl, alkynyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, heterocyclyl, and heterocyclylalkyl is independently optionally substituted with 1, 2, 3, 4, or 5 substituents. In some embodiments, R1and R1′ are the same, R2and R2′ are the same, R3and R3′ are the same, X and X′ are the same, Y and Y′ are the same, Z and Z′ are the same, A and A′ are the same, and Q and Q′ are the same. In some embodiments, R1and R1′ are heterocyclyl, which is optionally substituted. In some embodiments, R1and R1′ are monocyclic 4- to 6-membered heterocyclyl having 1 or 2 nitrogen atoms, which is optionally substituted. In some embodiments, R1and R1′ are each a 4- or 5-membered monocyclic heterocyclyl, such as a 4- or 5-membered heterocyclyl having 1 nitrogen atom, which is optionally substituted. In some embodiments, R1and R1′ are pyrrolidine, which is optionally substituted. In some embodiments, R1and R1′ are unsubstituted pyrrolidine. In some embodiments, X and X′ are —C(O)—. In some embodiments, Y and Y′ are —NRa—, and Rais selected from hydrogen and C1-C6alkyl. In some embodiments, Y and Y′ are —NRa—, and Rais selected from hydrogen and methyl. In some embodiments, Y and Y′ are —NRa—, and Rais hydrogen. In some embodiments, Z and Z′ are each absent. In some embodiments, A and A′ are each a five-membered monocyclic heteroaryl having 1 or 2 heteroatoms independently selected from S and N. In some embodiments, A and A′ are selected from thiophene and thiazole. In some embodiments, A and A′ are thiophene. In some embodiments, Q and Q′ are each a four-, five-, or six-membered heterocyclyl having one nitrogen atom (i.e. the nitrogen atom indicated in formula (IIa)). In other words, in some embodiments, Q and Q′ are selected from azetidine, pyrrolidine, and piperidine. In some embodiments, Q and Q′ are selected from azetidine and pyrrolidine. In some embodiments, Q and Q′ are azetidine. In some embodiments, Q and Q′ are pyrrolidine. In some embodiments, R2and R2′ are hydrogen. In some embodiments, R3is selected from aryl, heteroaryl, and a group of formula: In some embodiments, R3is a group of formula: wherein B is a 5-membered monocyclic heteroaryl having 1 or 2 heteroatoms independently selected from N and S; J is absent; C is selected from aryl, heteroaryl, and heterocyclyl; m is 0 or 1; Rgis C1-C6alkyl; n is 0, 1, or 2; and each Rhis independently selected from C1-C6alkyl, halo, C1-C6haloalkyl, amino, amino-C1-C6-alkyl, amido-C1-C6-alkyl, and heterocyclyl. In some embodiments, B is selected from thiazole and thiophene. In some embodiments, B is thiazole. In some embodiments, C is selected from aryl and monocyclic heteroaryl. In some embodiments, C is selected from phenyl and pyridyl. In some embodiments, C is phenyl. In some embodiments, R3′is selected from hydrogen, aryl, heteroaryl, and a group of formula: In some embodiments, R3′is selected from a monocyclic and bicyclic heteroaryl having 1, 2, or 3 heteroatoms independently selected from N and S. In some embodiments, R3′is a group of formula: wherein B′ is a 5-membered monocyclic heteroaryl having 1 or 2 heteroatoms independently selected from N and S; J′ is absent; C′ is selected from aryl, heteroaryl, and heterocyclyl; m′ is 0 or 1; Rg′is C1-C6alkyl; n′ is 0, 1, or 2; and each Rh′is independently selected from C1-C6alkyl, halo, C1-C6haloalkyl, amino, amino-C1-C6-alkyl, amido-C1-C6-alkyl, and heterocyclyl. In some embodiments, B is selected from thiazole and thiophene. In some embodiments, B is thiazole. In some embodiments, C is selected from aryl and monocyclic heteroaryl. In some embodiments, C is selected from phenyl and pyridyl. In some embodiments, C is phenyl. In some embodiments, at least one Rhhas formula —(CH2)rC(O)NRiRjor —(CH2)sNRkC(O)Rm, wherein:r and s are each independently selected from 0, 1, and 2;Riand Rkare each independently selected from hydrogen and C1-C6alkyl;Rjis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl;Rmis selected from C1-C6-alkyl, aryl, aryl-C1-C6-alkyl, heteroaryl, heteroaryl-C1-C6-alkyl, heterocyclyl, heterocyclyl-C1-C6-alkyl, cycloalkyl, and cycloalkyl-C1-C6-alkyl, amino, C1-C6-alkylamino, arylamino, aryl-C1-C6-alkylamino;wherein each alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl is independently unsubstituted or substituted with 1 or 2 substituents independently selected from halo, C1-C6-alkyl, C1-C6-alkoxy, hydroxy, amino and oxo. In some embodiments, L is a linker comprising one or more groups independently selected from methylene (—CH2—), vinylene (—CH═CH—), acetylene (—C≡C—), ether (—O—), amine (—NH—), alkylamine (—NR—, wherein R is an optionally substituted C1-C6alkyl group), amide (—C(O)NH—), ester (—C(O)O—), carbamate (—OC(O)NH—), sulfonamide (—S(O)2NH—), phenylene (—C6H4—), heteroarylene, heterocyclylene, and any combination thereof. In some embodiments, L is selected from: wherein a, a1, and a2 are each independently selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; b, b1, and b2 are each independently selected from 0, 1, 2, 3, 4, 5, and 6; c, c1, and c2 are each independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; d and e are each independently selected form 0, 1, and 2; each G is independently selected from CH and N; X1and X2are each independently O or —NRx, wherein Rxis hydrogen or optionally substituted alkyl; and Y1and Z1are each independently selected from —CH2—, —NH—, and —O—. The compounds can be synthesized in a variety of ways. For example, compounds of Formula (I) can be synthesized as shown in Schemes 1 and 2. Generally, the compounds can be synthesized by coupling appropriate amines with acids using suitable coupling agents, such as HATU. (In Schemes 1 and 2, HATU refers to (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate, DIPEA is N,N-diisopropylethylamine, DCM is dichloromethane, and Mt is a metal selected from Li, Na, K, or the like.) The compounds and intermediates may be isolated and purified by methods well-known to those skilled in the art of organic synthesis. Examples of conventional methods for isolating and purifying compounds can include, but are not limited to, chromatography on solid supports such as silica gel, alumina, or silica derivatized with alkylsilane groups, by recrystallization at high or low temperature with an optional pretreatment with activated carbon, thin-layer chromatography, distillation at various pressures, sublimation under vacuum, and trituration, as described for instance in “Vogel's Textbook of Practical Organic Chemistry” 5th edition (1989), by Furniss, Hannaford, Smith, and Tatchell, pub. Longman Scientific & Technical, Essex CM20 2JE, England. Reaction conditions and reaction times for each individual step can vary depending on the particular reactants employed and substituents present in the reactants used. Specific procedures are provided in the Examples section. Reactions can be worked up in the conventional manner, e.g. by eliminating the solvent from the residue and further purified according to methodologies generally known in the art such as, but not limited to, crystallization, distillation, extraction, trituration and chromatography. Unless otherwise described, the starting materials and reagents are either commercially available or can be prepared by one skilled in the art from commercially available materials using methods described in the chemical literature. Starting materials, if not commercially available, can be prepared by procedures selected from standard organic chemical techniques, techniques that are analogous to the synthesis of known, structurally similar compounds, or techniques that are analogous to the above described schemes or the procedures described in the synthetic examples section. Routine experimentations, including appropriate manipulation of the reaction conditions, reagents and sequence of the synthetic route, protection of any chemical functionality that cannot be compatible with the reaction conditions, and deprotection at a suitable point in the reaction sequence of the method are included in the scope of the disclosure. Suitable protecting groups and the methods for protecting and deprotecting different substituents using such suitable protecting groups are well known to those skilled in the art; examples of which can be found in P G M Wuts and T W Greene, in Greene's book titled Protective Groups in Organic Synthesis (4thed.), John Wiley & Sons, NY (2006), which is incorporated herein by reference in its entirety. Synthesis of the compounds of the disclosure can be accomplished by methods analogous to those described in the synthetic schemes described above and in specific examples described below. The compounds described herein may in some cases exist as diastereomers, enantiomers, or other stereoisomeric forms. The compounds presented herein include all diastereomeric, enantiomeric, and stereoisomeric forms as well as the appropriate mixtures thereof. Separation of stereoisomers may be performed by chromatography or by the forming diastereomeric and separation by recrystallization, or chromatography, or any combination thereof. (Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley And Sons, Inc., 1981, herein incorporated by reference for this disclosure). Stereoisomers may also be obtained by stereoselective synthesis. In some embodiments, compounds may exist as tautomers. All tautomers are included within the formulas described herein. Unless specified otherwise, divalent variables or groups described herein may be attached in the orientation in which they are depicted or they may be attached in the reverse orientation. The methods and compositions described herein include the use of amorphous forms as well as crystalline forms (also known as polymorphs). The compounds described herein may be in the form of pharmaceutically acceptable salts. As well, active metabolites of these compounds having the same type of activity are included in the scope of the present disclosure. In addition, the compounds described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, etc. The solvated forms of the compounds presented herein are also considered to be disclosed herein. In some embodiments, compounds or salts described herein may be prodrugs. A “prodrug” refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug. An example, without limitation, of a prodrug would be a compound described herein, which is administered as an ester (the “prodrug”) to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial. A further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound. In certain embodiments, a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound. To produce a prodrug, a pharmaceutically active compound is modified such that the active compound will be regenerated upon in vivo administration. The prodrug can be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug. In some embodiments, by virtue of knowledge of pharmacodynamic processes and drug metabolism in vivo, once a pharmaceutically active compound is determined, prodrugs of the compound are designed. (see, for example, Nogrady (1985)Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392; Silverman (1992), The Organic Chemistry of Drug Design and Drug Action, Academic Press, Inc., San Diego, pages 352-401, Saulnier et al., (1994),Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985; Rooseboom et al.,Pharmacological Reviews,56:53-102, 2004; Miller et al.,J. Med. Chem. Vol. 46, no. 24, 5097-5116, 2003; Aesop Cho, “Recent Advances in Oral Prodrug Discovery”,Annual Reports in Medicinal Chemistry, Vol. 41, 395-407, 2006). The compounds described herein may be labeled isotopically (e.g. with a radioisotope) or by other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, photoactivatable or chemiluminescent labels, affinity labels (e.g. biotin), etc. Compounds and salts described herein include isotopically-labeled compounds. In general, isotopically-labeled compounds are identical to those recited in the various formulae and structures presented herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number most common in nature. Examples of isotopes that can be incorporated into the present compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, for example,2H,3H,13C,14C,15N,18O,17O,35S,18F, or36Cl. Certain isotopically-labeled compounds described herein, for example those into which radioactive isotopes such as3H and14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Further, substitution with isotopes such as deuterium, i.e.,2H, can afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. In additional or further embodiments, the compounds described herein are metabolized upon administration to an organism in need to produce a metabolite that is then used to produce a desired effect, including a desired therapeutic effect. Compounds described herein may be formed as, and/or used as, pharmaceutically acceptable salts. The type of pharmaceutical acceptable salts, include, but are not limited to: (1) acid addition salts, formed by reacting the free base form of the compound with a pharmaceutically acceptable: inorganic acid, such as, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, metaphosphoric acid, and the like; or with an organic acid, such as, for example, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, trifluoroacetic acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4,4′-methylenebis-(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, butyric acid, phenylacetic acid, phenylbutyric acid, valproic acid, and the like; (2) salts formed when an acidic proton present in the parent compound is replaced by a metal ion, e.g., an alkali metal ion (e.g. lithium, sodium, potassium), an alkaline earth ion (e.g. magnesium, or calcium), or an aluminum ion. In some cases, compounds described herein may coordinate with an organic base, such as, but not limited to, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine. In other cases, compounds described herein may form salts with amino acids such as, but not limited to, arginine, lysine, and the like. Acceptable inorganic bases used to form salts with compounds that include an acidic proton, include, but are not limited to, aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms or crystal forms thereof, particularly solvates or polymorphs. Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds described herein can be conveniently prepared or formed during the processes described herein. In addition, the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein. In some embodiments, compounds described herein are in various forms, including but not limited to, amorphous forms, milled forms and nano-particulate forms. In addition, compounds described herein include crystalline forms, also known as polymorphs. Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, melting points, density, hardness, crystal shape, optical properties, stability, and solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate. The screening and characterization of the pharmaceutically acceptable salts, polymorphs and/or solvates may be accomplished using a variety of techniques including, but not limited to, thermal analysis, x-ray diffraction, spectroscopy, vapor sorption, and microscopy. Thermal analysis methods address thermo chemical degradation or thermo physical processes including, but not limited to, polymorphic transitions, and such methods are used to analyze the relationships between polymorphic forms, determine weight loss, to find the glass transition temperature, or for excipient compatibility studies. Such methods include, but are not limited to, Differential scanning calorimetry (DSC), Modulated Differential Scanning Calorimetry (MDCS), Thermogravimetric analysis (TGA), and Thermogravi-metric and Infrared analysis (TG/IR). X-ray diffraction methods include, but are not limited to, single crystal and powder diffractometers and synchrotron sources. The various spectroscopic techniques used include, but are not limited to, Raman, FTIR, UV-VIS, and NMR (liquid and solid state). The various microscopy techniques include, but are not limited to, polarized light microscopy, Scanning Electron Microscopy (SEM) with Energy Dispersive X-Ray Analysis (EDX), Environmental Scanning Electron Microscopy with EDX (in gas or water vapor atmosphere), IR microscopy, and Raman microscopy. Pharmaceutical Compositions In certain embodiments, a compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc)), or a pharmaceutically acceptable salt thereof, is combined with one or more additional agents to form a pharmaceutical composition. Pharmaceutical compositions may be formulated in a conventional manner using one or more physiologically acceptable carriers including excipients and auxiliaries that facilitate processing of the active compound into a preparation, which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Additional details about suitable excipients for pharmaceutical compositions described herein may be found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999), herein incorporated by reference for such disclosure. A pharmaceutical composition, as used herein, refers to a mixture of a compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc)), or a pharmaceutically acceptable salt thereof, with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. The pharmaceutical composition facilitates administration of the compound to a subject. In practicing the methods of treatment or use provided herein, therapeutically effective amounts of compounds described herein are administered in a pharmaceutical composition to a subject having a disease, disorder, or condition to be treated (e.g., cancer). In some embodiments, the subject is a human. A therapeutically effective amount can vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. The compound or pharmaceutically acceptable salt thereof, can be used singly or in combination with one or more therapeutic agents as components of mixtures (as in combination therapy). The pharmaceutical formulations described herein can be administered to a subject by multiple administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical, rectal, or transdermal administration routes. Moreover, the pharmaceutical compositions described herein, which include a compound disclosed herein (e.g., a compound of formula (Ia), (IIa), (IIb), or (IIc)), or a pharmaceutically acceptable salt thereof, can be formulated into any suitable dosage form, including but not limited to, aqueous oral dispersions, liquids, gels, syrups, elixirs, slurries, suspensions, aerosols, fast melt formulations, effervescent formulations, lyophilized formulations, tablets, powders, pills, dragees, and capsules. One may administer the compounds and/or compositions in a local rather than systemic manner, for example, via injection of the compound directly into an organ or tissue, often in a depot preparation or sustained release formulation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with organ-specific antibody. The liposomes will be targeted to and taken up selectively by the organ. In addition, the drug may be provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation. Pharmaceutical compositions including a compound described herein may be manufactured in a conventional manner, such as, by way of example only, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes. In certain embodiments, compositions provided herein may also include one or more preservatives to inhibit microbial activity. Suitable preservatives include quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride. Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipients with one or more of the compounds disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc)), or a pharmaceutically acceptable salt thereof, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets, pills, or capsules. Suitable excipients include, for example, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. If desired, disintegrating agents may be added, such as the cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. Pharmaceutical preparations that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. In some embodiments, the solid dosage forms disclosed herein may be in the form of a tablet, (including a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet), a pill, a powder (including a sterile packaged powder, a dispensable powder, or an effervescent powder), a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC, or “sprinkle capsules”), solid dispersion, solid solution, bioerodible dosage form, multiparticulate dosage forms, pellets, granules, or an aerosol. In other embodiments, the pharmaceutical formulation is in the form of a powder. In still other embodiments, the pharmaceutical formulation is in the form of a tablet, including but not limited to, a fast-melt tablet. Additionally, pharmaceutical formulations of the compounds described herein may be administered as a single capsule or in multiple capsule dosage form. In some embodiments, the pharmaceutical formulation is administered in two, or three, or four, capsules or tablets. In some embodiments, solid dosage forms, e.g., tablets, effervescent tablets, and capsules, are prepared by mixing particles of a compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc)), or a pharmaceutically acceptable salt thereof, with one or more pharmaceutical excipients to form a bulk blend composition. When referring to these bulk blend compositions as homogeneous, it is meant that the particles of the compound are dispersed evenly throughout the composition so that the composition may be subdivided into equally effective unit dosage forms, such as tablets, pills, and capsules. The individual unit dosages may also include film coatings, which disintegrate upon oral ingestion or upon contact with diluent. These formulations can be manufactured by conventional pharmacological techniques. The pharmaceutical solid dosage forms described herein can include a compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc)), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable additives such as a compatible carrier, binder, filling agent, suspending agent, flavoring agent, sweetening agent, disintegrating agent, dispersing agent, surfactant, lubricant, colorant, diluent, solubilizer, moistening agent, plasticizer, stabilizer, penetration enhancer, wetting agent, anti-foaming agent, antioxidant, preservative, or one or more combination thereof. In still other aspects, using standard coating procedures, such as those described in Remington's Pharmaceutical Sciences, 20th Edition (2000), a film coating is provided around the formulation of the compound described herein. In one embodiment, some or all of the particles of the compound described herein are coated. In another embodiment, some or all of the particles of the compound described herein are microencapsulated. In still another embodiment, the particles of the compound described herein are not microencapsulated and are uncoated. Suitable carriers for use in the solid dosage forms described herein include, but are not limited to, acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, sodium caseinate, soy lecithin, sodium chloride, tricalcium phosphate, dipotassium phosphate, sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, hydroxypropylmethylcellulose, hydroxypropylmethylcellulose acetate stearate, sucrose, microcrystalline cellulose, lactose, mannitol and the like. Suitable filling agents for use in the solid dosage forms described herein include, but are not limited to, lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, hydroxypropylmethycellulose (HPMC), hydroxypropylmethycellulose phthalate, hydroxypropylmethylcellulose acetate stearate (HPMCAS), sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like. In order to release the compound from a solid dosage form matrix as efficiently as possible, disintegrants are often used in the formulation, especially when the dosage forms are compressed with binder. Disintegrants help rupturing the dosage form matrix by swelling or capillary action when moisture is absorbed into the dosage form. Suitable disintegrants for use in the solid dosage forms described herein include, but are not limited to, natural starch such as corn starch or potato starch, a pregelatinized starch such as National 1551 or Amijel®, or sodium starch glycolate such as Promogel® or Explotab®, a cellulose such as a wood product, methylcrystalline cellulose, e.g., Avicel®, Avicel® PH101, Avicel® PH102, Avicel® PH105, Elcema® P100, Emcocel®, Vivacel®, Ming Tia®, and Solka-Floc®, methylcellulose, croscarmellose, or a cross-linked cellulose, such as cross-linked sodium carboxymethylcellulose (Ac-Di-Sol®), cross-linked carboxymethylcellulose, or cross-linked croscarmellose, a cross-linked starch such as sodium starch glycolate, a cross-linked polymer such as crospovidone, a cross-linked polyvinylpyrrolidone, alginate such as alginic acid or a salt of alginic acid such as sodium alginate, a clay such as Veegum® HV (magnesium aluminum silicate), a gum such as agar, guar, locust bean, Karaya, pectin, or tragacanth, sodium starch glycolate, bentonite, a natural sponge, a surfactant, a resin such as a cation-exchange resin, citrus pulp, sodium lauryl sulfate, sodium lauryl sulfate in combination starch, and the like. Binders impart cohesiveness to solid oral dosage form formulations: for powder filled capsule formulation, they aid in plug formation that can be filled into soft or hard shell capsules and for tablet formulation, they ensure the tablet remaining intact after compression and help assure blend uniformity prior to a compression or fill step. Materials suitable for use as binders in the solid dosage forms described herein include, but are not limited to, carboxymethylcellulose, methylcellulose (e.g., Methocel®), hydroxypropylmethylcellulose (e.g. Hypromellose USP Pharmacoat-603, hydroxypropylmethylcellulose acetate stearate (Aqoate HS-LF and HS), hydroxyethylcellulose, hydroxypropylcellulose (e.g., Klucel®), ethylcellulose (e.g., Ethocel®), and microcrystalline cellulose (e.g., Avicel®), microcrystalline dextrose, amylose, magnesium aluminum silicate, polysaccharide acids, bentonites, gelatin, polyvinylpyrrolidone/vinyl acetate copolymer, crospovidone, povidone, starch, pregelatinized starch, tragacanth, dextrin, a sugar, such as sucrose (e.g., Dipac®), glucose, dextrose, molasses, mannitol, sorbitol, xylitol (e.g., Xylitab®), lactose, a natural or synthetic gum such as acacia, tragacanth, ghatti gum, mucilage of isapol husks, starch, polyvinylpyrrolidone (e.g., Povidone® CL, Kollidon® CL, Polyplasdone® XL-10, and Povidone® K-12), larch arabogalactan, Veegum®, polyethylene glycol, waxes, sodium alginate, and the like. In general, binder levels of 20-70% are used in powder-filled gelatin capsule formulations. Binder usage level in tablet formulations varies whether direct compression, wet granulation, roller compaction, or usage of other excipients such as fillers which itself can act as moderate binder. In some embodiments, formulators determine the binder level for the formulations, but binder usage level of up to 70% in tablet formulations is common. Suitable lubricants or glidants for use in the solid dosage forms described herein include, but are not limited to, stearic acid, calcium hydroxide, talc, corn starch, sodium stearyl fumerate, alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, magnesium stearate, zinc stearate, waxes, Stearowet®, boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol or a methoxypolyethylene glycol such as Carbowax™, PEG 4000, PEG 5000, PEG 6000, propylene glycol, sodium oleate, glyceryl behenate, glyceryl palmitostearate, glyceryl benzoate, magnesium or sodium lauryl sulfate, and the like. Suitable diluents for use in the solid dosage forms described herein include, but are not limited to, sugars (including lactose, sucrose, and dextrose), polysaccharides (including dextrates and maltodextrin), polyols (including mannitol, xylitol, and sorbitol), cyclodextrins and the like. Suitable wetting agents for use in the solid dosage forms described herein include, for example, oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, quaternary ammonium compounds (e.g., Polyquat 10®), sodium oleate, sodium lauryl sulfate, magnesium stearate, sodium docusate, triacetin, vitamin E TPGS and the like. Suitable surfactants for use in the solid dosage forms described herein include, for example, sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), and the like. Suitable suspending agents for use in the solid dosage forms described here include, but are not limited to, polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30, polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 5400 to about 7000, vinyl pyrrolidone/vinyl acetate copolymer (S630), sodium carboxymethylcellulose, methylcellulose, hydroxy-propylmethylcellulose, polysorbate-80, hydroxyethylcellulose, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gum, sugars, cellulosics, such as, e.g., sodium carboxymethylcellulose, methylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, hydroxyethylcellulose, polysorbate-80, sodium alginate, polyethoxylated sorbitan monolaurate, polyethoxylated sorbitan monolaurate, povidone and the like. Suitable antioxidants for use in the solid dosage forms described herein include, for example, e.g., butylated hydroxytoluene (BHT), sodium ascorbate, and tocopherol. There is considerable overlap between additives used in the solid dosage forms described herein. Thus, the above-listed additives should be taken as merely exemplary, and not limiting, of the types of additives that can be included in solid dosage forms of the pharmaceutical compositions described herein. In other embodiments, one or more layers of the pharmaceutical formulation are plasticized. Illustratively, a plasticizer is generally a high boiling point solid or liquid. Suitable plasticizers can be added from about 0.01% to about 50% by weight (w/w) of the coating composition. Plasticizers include, but are not limited to, diethyl phthalate, citrate esters, polyethylene glycol, glycerol, acetylated glycerides, triacetin, polypropylene glycol, polyethylene glycol, triethyl citrate, dibutyl sebacate, stearic acid, stearol, stearate, and castor oil. Compressed tablets are solid dosage forms prepared by compacting the bulk blend of the formulations described above. In various embodiments, compressed tablets which are designed to dissolve in the mouth will include one or more flavoring agents. In other embodiments, the compressed tablets will include a film surrounding the final compressed tablet. In some embodiments, the film coating aids in patient compliance (e.g., Opadry® coatings or sugar coating). Film coatings including Opadry® typically range from about 1% to about 3% of the tablet weight. In other embodiments, the compressed tablets include one or more excipients. A capsule may be prepared, for example, by placing the bulk blend of the formulation of the compound described above, inside of a capsule. In some embodiments, the formulations (non-aqueous suspensions and solutions) are placed in a soft gelatin capsule. In other embodiments, the formulations are placed in standard gelatin capsules or non-gelatin capsules such as capsules comprising HPMC. In other embodiments, the formulation is placed in a sprinkle capsule, wherein the capsule may be swallowed whole or the capsule may be opened and the contents sprinkled on food prior to eating. In some embodiments, the therapeutic dose is split into multiple (e.g., two, three, or four) capsules. In some embodiments, the entire dose of the formulation is delivered in a capsule form. In various embodiments, the particles of the compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof), and one or more excipients are dry blended and compressed into a mass, such as a tablet, having a hardness sufficient to provide a pharmaceutical composition that substantially disintegrates within less than about 30 minutes, less than about 35 minutes, less than about 40 minutes, less than about 45 minutes, less than about 50 minutes, less than about 55 minutes, or less than about 60 minutes, after oral administration, thereby releasing the formulation into the gastrointestinal fluid. In another aspect, dosage forms may include microencapsulated formulations. In some embodiments, one or more other compatible materials are present in the microencapsulation material. Exemplary materials include, but are not limited to, pH modifiers, erosion facilitators, anti-foaming agents, antioxidants, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, and diluents. Materials useful for the microencapsulation described herein include materials compatible with compounds described herein, which sufficiently isolate the compound from other non-compatible excipients. In still other embodiments, effervescent powders are also prepared in accordance with the present disclosure. Effervescent salts have been used to disperse medicines in water for oral administration. Effervescent salts are granules or coarse powders containing a medicinal agent in a dry mixture, usually composed of sodium bicarbonate, citric acid and/or tartaric acid. When such salts are added to water, the acids and the base react to liberate carbon dioxide gas, thereby causing “effervescence.” Examples of effervescent salts include, e.g., the following ingredients: sodium bicarbonate or a mixture of sodium bicarbonate and sodium carbonate, citric acid and/or tartaric acid. Any acid-base combination that results in the liberation of carbon dioxide can be used in place of the combination of sodium bicarbonate and citric and tartaric acids, as long as the ingredients were suitable for pharmaceutical use and result in a pH of about 6.0 or higher. In other embodiments, the formulations described herein are solid dispersions. Methods of producing such solid dispersions include, but are not limited to, for example, U.S. Pat. Nos. 4,343,789, 5,340,591, 5,456,923, 5,700,485, 5,723,269, and U.S. patent publication no. 2004/0013734. In still other embodiments, the formulations described herein are solid solutions. Solid solutions incorporate a substance together with the active agent and other excipients such that heating the mixture results in dissolution of the drug and the resulting composition is then cooled to provide a solid blend which can be further formulated or directly added to a capsule or compressed into a tablet. Methods of producing such solid solutions include, but are not limited to, for example, U.S. Pat. Nos. 4,151,273, 5,281,420, and 6,083,518. In some embodiments, pharmaceutical formulations are provided that include particles of the compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc)), or a pharmaceutically acceptable salt thereof, and at least one dispersing agent or suspending agent for oral administration to a subject. The formulations may be a powder and/or granules for suspension, and upon admixture with water, a substantially uniform suspension is obtained. Liquid formulation dosage forms for oral administration can be aqueous suspensions selected from the group including, but not limited to, pharmaceutically acceptable aqueous oral dispersions, emulsions, solutions, elixirs, gels, and syrups. See, e.g., Singh et al., Encyclopedia of Pharmaceutical Technology, 2nd Ed., pp. 754-757 (2002). The aqueous suspensions and dispersions described herein can remain in a homogenous state, as defined in The USP Pharmacists' Pharmacopeia (2005 edition, chapter 905), for at least 4 hours. The homogeneity should be determined by a sampling method consistent with regard to determining homogeneity of the entire composition. In one embodiment, an aqueous suspension can be re-suspended into a homogenous suspension by physical agitation lasting less than 1 minute. In another embodiment, an aqueous suspension can be re-suspended into a homogenous suspension by physical agitation lasting less than 45 seconds. In yet another embodiment, an aqueous suspension can be re-suspended into a homogenous suspension by physical agitation lasting less than 30 seconds. In still another embodiment, no agitation is necessary to maintain a homogeneous aqueous dispersion. The pharmaceutical compositions described herein may include sweetening agents such as, but not limited to, acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream, berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream, chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate (MagnaSweet®), maltol, mannitol, maple, marshmallow, menthol, mint cream, mixed berry, neohesperidine DC, neotame, orange, pear, peach, peppermint, peppermint cream, Prosweet® Powder, raspberry, root beer, rum, saccharin, safrole, sorbitol, spearmint, spearmint cream, strawberry, strawberry cream, stevia, sucralose, sucrose, sodium saccharin, saccharin, aspartame, acesulfame potassium, mannitol, talin, sucralose, sorbitol, swiss cream, tagatose, tangerine, thaumatin, tutti fruitti, vanilla, walnut, watermelon, wild cherry, wintergreen, xylitol, or any combination of these flavoring ingredients, e.g., anise-menthol, cherry-anise, cinnamon-orange, cherry-cinnamon, chocolate-mint, honey-lemon, lemon-lime, lemon-mint, menthol-eucalyptus, orange-cream, vanilla-mint, and mixtures thereof. In some embodiments, the pharmaceutical formulations described herein can be self-emulsifying drug delivery systems (SEDDS). Emulsions are dispersions of one immiscible phase in another, usually in the form of droplets. Generally, emulsions are created by vigorous mechanical dispersion. SEDDS, as opposed to emulsions or microemulsions, spontaneously form emulsions when added to an excess of water without any external mechanical dispersion or agitation. An advantage of SEDDS is that only gentle mixing is required to distribute the droplets throughout the solution. Additionally, water or the aqueous phase can be added just prior to administration, which ensures stability of an unstable or hydrophobic active ingredient. Thus, the SEDDS provides an effective delivery system for oral and parenteral delivery of hydrophobic active ingredients. SEDDS may provide improvements in the bioavailability of hydrophobic active ingredients. Methods of producing self-emulsifying dosage forms include, but are not limited to, for example, U.S. Pat. Nos. 5,858,401, 6,667,048, and 6,960,563. There is overlap between the above-listed additives used in the aqueous dispersions or suspensions described herein, since a given additive is often classified differently by different practitioners in the field, or is commonly used for any of several different functions. Thus, the above-listed additives should be taken as merely exemplary, and not limiting, of the types of additives that can be included in formulations described herein. Potential excipients for intranasal formulations include, for example, U.S. Pat. Nos. 4,476,116, 5,116,817 and 6,391,452. Formulations solutions in saline, employing benzyl alcohol or other suitable preservatives, fluorocarbons, and/or other solubilizing or dispersing agents. See, for example, Ansel, H. C. et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, Sixth Ed. (1995). Preferably these compositions and formulations are prepared with suitable nontoxic pharmaceutically acceptable ingredients. The choice of suitable carriers is highly dependent upon the exact nature of the nasal dosage form desired, e.g., solutions, suspensions, ointments, or gels. Nasal dosage forms generally contain large amounts of water in addition to the active ingredient. Minor amounts of other ingredients such as pH adjusters, emulsifiers or dispersing agents, preservatives, surfactants, gelling agents, or buffering and other stabilizing and solubilizing agents may also be present. Preferably, the nasal dosage form should be isotonic with nasal secretions. For administration by inhalation, the compounds described herein may be in a form as an aerosol, a mist or a powder. Pharmaceutical compositions described herein are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound described herein and a suitable powder base such as lactose or starch. Buccal formulations that include compounds described herein may be administered using a variety of formulations which include, but are not limited to, U.S. Pat. Nos. 4,229,447, 4,596,795, 4,755,386, and 5,739,136. In addition, the buccal dosage forms described herein can further include a bioerodible (hydrolysable) polymeric carrier that also serves to adhere the dosage form to the buccal mucosa. The buccal dosage form is fabricated so as to erode gradually over a predetermined time period, wherein the delivery of the compound is provided essentially throughout. Buccal drug delivery avoids the disadvantages encountered with oral drug administration, e.g., slow absorption, degradation of the active agent by fluids present in the gastrointestinal tract and/or first-pass inactivation in the liver. With regard to the bioerodible (hydrolysable) polymeric carrier, virtually any such carrier can be used, so long as the desired drug release profile is not compromised, and the carrier is compatible with the compounds described herein, and any other components that may be present in the buccal dosage unit. Generally, the polymeric carrier comprises hydrophilic (water-soluble and water-swellable) polymers that adhere to the wet surface of the buccal mucosa. Examples of polymeric carriers useful herein include acrylic acid polymers and co, e.g., those known as “carbomers” (Carbopol®, which may be obtained from B.F. Goodrich, is one such polymer). Other components may also be incorporated into the buccal dosage forms described herein include, but are not limited to, disintegrants, diluents, binders, lubricants, flavoring, colorants, preservatives, and the like. For buccal or sublingual administration, the compositions may take the form of tablets, lozenges, or gels formulated in a conventional manner. Transdermal formulations described herein may be administered using a variety of devices including but not limited to, U.S. Pat. Nos. 3,598,122, 3,598,123, 3,710,795, 3,731,683, 3,742,951, 3,814,097, 3,921,636, 3,972,995, 3,993,072, 3,993,073, 3,996,934, 4,031,894, 4,060,084, 4,069,307, 4,077,407, 4,201,211, 4,230,105, 4,292,299, 4,292,303, 5,336,168, 5,665,378, 5,837,280, 5,869,090, 6,923,983, 6,929,801 and 6,946,144. The transdermal dosage forms described herein may incorporate certain pharmaceutically acceptable excipients which are conventional in the art. In one embodiment, the transdermal formulations described herein include at least three components: (1) a formulation of a compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc)), or a pharmaceutically acceptable salt thereof; (2) a penetration enhancer; and (3) an aqueous adjuvant. In addition, transdermal formulations can include additional components such as, but not limited to, gelling agents, creams and ointment bases, and the like. In some embodiments, the transdermal formulation can further include a woven or non-woven backing material to enhance absorption and prevent the removal of the transdermal formulation from the skin. In other embodiments, the transdermal formulations described herein can maintain a saturated or supersaturated state to promote diffusion into the skin. Formulations suitable for transdermal administration of compounds described herein may employ transdermal delivery devices and transdermal delivery patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. Still further, transdermal delivery of the compounds described herein can be accomplished by means of iontophoretic patches and the like. Additionally, transdermal patches can provide controlled delivery of the compounds described herein. The rate of absorption can be slowed by using rate-controlling membranes or by trapping the compound within a polymer matrix or gel. Conversely, absorption enhancers can be used to increase absorption. An absorption enhancer or carrier can include absorbable pharmaceutically acceptable solvents to assist passage through the skin. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. Formulations suitable for intramuscular, subcutaneous, or intravenous injection may include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and non-aqueous carriers, diluents, solvents, or vehicles including water, ethanol, polyols (propyleneglycol, polyethylene-glycol, glycerol, cremophor and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. Formulations suitable for subcutaneous injection may also contain additives such as preserving, wetting, emulsifying, and dispensing agents. Prevention of the growth of microorganisms can be ensured by various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, such as aluminum monostearate and gelatin. For intravenous injections, compounds described herein may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally recognized in the field. For other parenteral injections, appropriate formulations may include aqueous or nonaqueous solutions, preferably with physiologically compatible buffers or excipients. Such excipients are generally recognized in the field. Parenteral injections may involve bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The pharmaceutical composition described herein may be in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. In certain embodiments, delivery systems for pharmaceutical compounds may be employed, such as, for example, liposomes and emulsions. In certain embodiments, compositions provided herein also include an mucoadhesive polymer, selected from among, for example, carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran. In some embodiments, the compounds described herein may be administered topically and are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments. Such pharmaceutical compounds can contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives. The compounds described herein may also be formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like. In suppository forms of the compositions, a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter is first melted. Generally, an agent, such as a compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc) or a pharmaceutically acceptable salt thereof), is administered in an amount effective for amelioration of, or prevention of the development of symptoms of, the disease or disorder (i.e., a therapeutically effective amount). Thus, a therapeutically effective amount can be an amount that is capable of at least partially preventing or reversing a disease or disorder. The dose required to obtain an effective amount may vary depending on the agent, formulation, disease or disorder, and individual to whom the agent is administered. Determination of effective amounts may also involve in vitro assays in which varying doses of agent are administered to cells in culture and the concentration of agent effective for ameliorating some or all symptoms is determined in order to calculate the concentration required in vivo. Effective amounts may also be based in in vivo animal studies. An agent can be administered prior to, concurrently with and subsequent to the appearance of symptoms of a disease or disorder. In some embodiments, an agent is administered to a subject with a family history of the disease or disorder, or who has a phenotype that may indicate a predisposition to a disease or disorder, or who has a genotype which predisposes the subject to the disease or disorder. In some embodiments, the compositions described herein are provided as pharmaceutical and/or therapeutic compositions. The pharmaceutical and/or therapeutic compositions of the present invention can be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration can be topical (including ophthalmic and to mucous membranes including vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Compositions and formulations for topical administration can include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional carriers; aqueous, powder, or oily bases; thickeners; and the like can be necessary or desirable. Compositions and formulations for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders can be desirable. Compositions and formulations for parenteral, intrathecal or intraventricular administration can include sterile aqueous solutions that can also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients. Pharmaceutical and/or therapeutic compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome containing formulations. These compositions can be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids and self-emulsifying semisolids. The pharmaceutical and/or therapeutic formulations, which can conveniently be presented in unit dosage form, can be prepared according to conventional techniques well known in the pharmaceutical/nutriceutical industries. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. The compositions of the present invention can be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, liquid syrups, soft gels, suppositories, and enemas. The compositions of the present invention can also be formulated as suspensions in aqueous, non-aqueous, oil-based, or mixed media. Suspensions can further contain substances that increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension can also contain stabilizers. In one embodiment of the present invention the pharmaceutical compositions can be formulated and used as foams. Pharmaceutical foams include formulations such as, but not limited to, emulsions, microemulsions, creams, jellies and liposomes. While basically similar in nature these formulations vary in the components and the consistency of the final product. The pharmaceutical composition described herein may be in unit dosage forms suitable for single administration of precise dosages. In unit dosage form, the formulation is divided into unit doses containing appropriate quantities of one or more compound. The unit dosage may be in the form of a package containing discrete quantities of the formulation. Non-limiting examples are packaged tablets or capsules, and powders in vials or ampoules. Aqueous suspension compositions can be packaged in single-dose non-reclosable containers. Alternatively, multiple-dose reclosable containers can be used, in which case it is typical to include a preservative in the composition. By way of example only, formulations for parenteral injection may be presented in unit dosage form, which include, but are not limited to ampoules, or in multi-dose containers, with an added preservative. Dosing and administration regimes are tailored by the clinician, or others skilled in the pharmacological arts, based upon well-known pharmacological and therapeutic considerations including, but not limited to, the desired level of therapeutic effect, and the practical level of therapeutic effect obtainable. Generally, it is advisable to follow well-known pharmacological principles for administrating chemotherapeutic agents (e.g., it is generally advisable to not change dosages by more than 50% at time and no more than every 3-4 agent half-lives). For compositions that have relatively little or no dose-related toxicity considerations, and where maximum efficacy is desired, doses in excess of the average required dose are not uncommon. This approach to dosing is commonly referred to as the “maximal dose” strategy. In certain embodiments, the compounds are administered to a subject at a dose of about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg. When the compounds described herein are co-administered with another agent (e.g., as sensitizing agents), the effective amount may be less than when the agent is used alone. Dosing may be once per day or multiple times per day for one or more consecutive days. Methods of Use/Treatment The present disclosure provides methods of using the compounds and compositions described herein (e.g., compounds of formula (I), (IIa), (IIb), and (IIc), or pharmaceutically acceptable salts thereof). The methods include methods of inhibiting GAS41 and methods of treating diseases such as cancer. In certain embodiments, the disclosure provides a method of inhibiting GAS41 activity in a sample, comprising contacting the sample with an effective amount a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof). The sample may be an in vitro or ex vivo sample (e.g., a sample comprising cells, tissue, or an organ). In some embodiments, the disclosure provides a method of inhibiting GAS41 activity by contacting the GAS41 with an effective amount of a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof), e.g., by contacting a cell, tissue, or organ that expresses GAS41 with the compound or the salt thereof. In some embodiments, the disclosure provides a method of inhibiting GAS41 activity in subject (including but not limited to rodents and mammals, e.g., humans), by administering into the subject an effective amount of a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable thereof). In some embodiments, the percentage inhibition exceeds 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%. In some embodiments, the disclosure provides methods of inhibiting GAS41 activity in a cell, comprising contacting the cell with an amount of a compound described herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof) sufficient to inhibit the activity. In some embodiments, the disclosure provides methods of inhibiting GAS41 activity in a tissue by contacting the tissue with an amount of a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof), sufficient to inhibit the GAS41 activity in the tissue. In some embodiments, the disclosure provides methods of inhibiting GAS41 activity in an organism (e.g., mammal, human, etc.) by contacting the organism with an amount of a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof), sufficient to inhibit GAS41 activity in the organism. Inhibition of GAS41 activity may be assessed and demonstrated by a wide variety of ways known in the art. Non-limiting examples include measure (a) a direct decrease in GAS41 activity; (b) a decrease in cell proliferation and/or cell viability; (c) an increase in cell differentiation; (d) a decrease in the levels of downstream targets of GAS41 activity; and (e) decrease in tumor volume and/or tumor volume growth rate. Kits and commercially available assays can be utilized for determining one or more of the above. The disclosure also provides methods for treating cancer in a subject in need thereof (e.g., a subject suffering from cancer), comprising administering a compound or pharmaceutical composition described herein (e.g., a compound of formula (I), (IIa) or (IIb) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of formula (I), (IIa) or (IIb) or a pharmaceutically acceptable salt thereof) to the subject. In certain embodiments, the cancer is associated with GAS41 expression (e.g., aberrant expression, overexpression, etc.) and/or activity. In certain embodiments, the cancer is brain cancer (e.g., an astrocytoma or glioblastoma), a sarcoma, colorectal cancer, lung cancer (e.g., non-small cell lung cancer), or gastric cancer. In certain embodiments, the disclosure provides a method of treating cancer in a subject, wherein the method comprises determining if the subject has a GAS41-mediated cancer, and administering to the subject a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof). Determining whether a tumor or cancer expresses (e.g., overexpresses, aberrantly expresses, etc.) GAS41 can be undertaken by assessing the nucleotide sequence encoding GAS41 or by assessing the amino acid sequence of GAS41. Methods for detecting a GAS41 nucleotide sequence are known by those of skill in the art. These methods include, but are not limited to, polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assays, polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP) assays, real-time PCR assays, PCR sequencing, mutant allele-specific PCR amplification (MASA) assays, direct sequencing, primer extension reactions, electrophoresis, oligonucleotide ligation assays, hybridization assays, TaqMan assays, SNP genotyping assays, high resolution melting assays, and microarray analyses. Methods for detecting a GAS41 protein are known by those of skill in the art. These methods include, but are not limited to, detection using a binding agent, e.g., an antibody specific for GAS41, protein electrophoresis and Western blotting, and direct peptide sequencing. Methods for determining whether a tumor or cancer expresses (e.g., overexpresses, aberrantly expresses, etc.) GAS41 or is mediated by GAS41 activity can use a variety of samples. In some embodiments, the sample is taken from a subject having a cancer or tumor. In some embodiments, the sample is a fresh tumor/cancer sample. In some embodiments, the sample is a frozen tumor/cancer sample. In some embodiments, the sample is a formalin-fixed paraffin-embedded sample. In some embodiments, the sample is processed to a cell lysate. In some embodiments, the sample is processed to DNA or RNA. The disclosure also relates to a method of treating a hyperproliferative disorder in a mammal that comprises administering to the mammal a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof). In some embodiments, the method relates to the treatment of cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers, e.g., Lymphoma and Kaposi's Sarcoma, anal cancer, angiosarcoma, appendix cancer, astrocytomas, atypical teratoid rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, carcinoid tumor, chondrosarcoma, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, epithelioid sarcoma, esophageal cancer, esthesioneuroblastoma, Ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, glioblastoma, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, leiomyosarcoma, lip and oral cavity cancer, liposarcoma, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer, multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, myxofibrosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, synovial sarcoma, T-Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or Viral-Induced cancer. In some embodiments, the method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin, e.g., psoriasis, restenosis, or prostate, e.g., benign prostatic hypertrophy (BPH). In some embodiments, the method relates to the treatment of brain cancer (e.g., astrocytoma or glioblastoma), a sarcoma, colorectal cancer, lung cancer (e.g., non-small cell lung cancer), or gastric cancer. Subjects that can be treated with compounds of the disclosure according to the methods of this disclosure include, for example, subjects that have been diagnosed as having acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers, e.g., Lymphoma and Kaposi's Sarcoma, anal cancer, angiosarcoma, appendix cancer, astrocytomas, atypical teratoid rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, carcinoid tumor, chondrosarcoma, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, epithelioid sarcoma, esophageal cancer, esthesioneuroblastoma, Ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, glioblastoma, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, leiomyosarcoma, lip and oral cavity cancer, liposarcoma, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer, multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, myxofibrosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, synovial sarcoma, T-Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or Viral-Induced cancer. In some embodiments, the method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin, e.g., psoriasis, restenosis, or prostate, e.g., benign prostatic hypertrophy (BPH). In some embodiments, the subject has been diagnosed with brain cancer (e.g., astrocytoma or glioblastoma), a sarcoma, colorectal cancer, lung cancer (e.g., non-small cell lung cancer), or gastric cancer The compositions containing the compounds or salts thereof described herein can be administered for prophylactic and/or therapeutic treatments. In therapeutic applications, the compounds or compositions are administered to a patient already suffering from a disease, in an amount sufficient to cure or at least partially arrest the symptoms of the disease. Amounts effective for this use will depend on the severity and course of the disease, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating clinician. In prophylactic applications, compositions containing the compounds or salts thereof described herein are administered to a patient susceptible to or otherwise at risk of a particular disease, disorder or condition. Such an amount is defined to be a “prophylactically effective amount or dose.” In this use, the precise amounts also depend on the patient's state of health, weight, and the like. When used in a patient, effective amounts for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating clinician. In the case wherein the patient's condition does not improve, upon the clinician's discretion the administration of the compounds may be administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease. In the case wherein the patient's status does improve, upon the clinician's discretion the administration of the compounds may be given continuously; alternatively, the dose of drug being administered may be temporarily reduced or temporarily suspended for a certain length of time (i.e., a “drug holiday”). The length of the drug holiday can vary between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday may be from about 10% to about 100%, including, by way of example only, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%. Once improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, can be reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained. Patients can, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms. The amount of a given agent that will correspond to such an amount will vary depending upon factors such as the particular compound, disease and its severity, the identity (e.g., weight) of the subject or host in need of treatment, but can nevertheless be determined in a manner recognized in the field according to the particular circumstances surrounding the case, including, e.g., the specific agent being administered, the route of administration, the condition being treated, and the subject or host being treated. In general, however, doses employed for adult human treatment will typically be in the range of about 0.02-about 5000 mg per day, in some embodiments, about 1-about 1500 mg per day. The desired dose may conveniently be presented in a single dose or as divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day. Toxicity and therapeutic efficacy of such therapeutic regimens can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD50(the dose lethal to 50% of the population) and the ED50(the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50and ED50. Compounds exhibiting high therapeutic indices are preferred. The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50with minimal toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. Combination Therapies Provided herein are methods for combination therapies in which an agent known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes are used in combination with a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof). In one aspect, such therapy includes but is not limited to the combination of one or more compounds of the disclosure with chemotherapeutic agents, targeted agents, therapeutic antibodies, and/or radiation treatment, to provide a synergistic or additive therapeutic effect. In general, the compounds and compositions described herein and, in embodiments where combinational therapy is employed, other agents do not have to be administered in the same pharmaceutical composition, and may, because of different physical and chemical characteristics, have to be administered by different routes. The determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the clinician. The initial administration can be made according to established protocols recognized in the field, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the clinician. In certain instances, it may be appropriate to administer at least one compound described herein in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving a compound described herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof), is nausea, then it may be appropriate to administer an anti-nausea agent in combination with the initial therapeutic agent. Or, by way of example only, the therapeutic effectiveness of one of the compounds described herein may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). Or, by way of example only, the benefit experienced by a patient may be increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit. In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit. The particular choice of compounds used will depend upon the diagnosis and judgment of the condition of the patient and the appropriate treatment protocol. The compounds may be administered concurrently (e.g., simultaneously, essentially simultaneously or within the same treatment protocol) or sequentially, depending upon the nature of the disease, disorder, or condition, the condition of the patient, and the actual choice of compounds used. The determination of the order of administration, and the number of repetitions of administration of each therapeutic agent during a treatment protocol, is well within the knowledge of the clinician after evaluation of the disease being treated and the condition of the patient. Therapeutically-effective dosages can vary when the drugs are used in treatment combinations. Methods for experimentally determining therapeutically-effective dosages of drugs and other agents for use in combination treatment regimens are described in the literature. For example, the use of metronomic dosing, i.e., providing more frequent, lower doses in order to minimize toxic side effects, has been described extensively in the literature. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient. For combination therapies described herein, dosages of the co-administered compounds will of course vary depending on the type of co-drug employed, on the specific drug employed, on the disease being treated and so forth. In addition, when co-administered with one or more biologically active agents, the compound provided herein may be administered either simultaneously with the biologically active agent(s), or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering protein in combination with the biologically active agent(s). In any case, the multiple therapeutic agents (one of which is a compound described herein or a pharmaceutically acceptable salt thereof (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof)), may be administered in any order or even simultaneously. If simultaneously, the multiple therapeutic agents may be provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). One of the therapeutic agents may be given in multiple doses, or both may be given as multiple doses. If not simultaneous, the timing between the multiple doses may vary from more than zero weeks to less than four weeks. In addition, the combination methods, compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations are also envisioned. It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, can be modified in accordance with a variety of factors. These factors include the disorder or condition from which the subject suffers, as well as the age, weight, sex, diet, and medical condition of the subject. Thus, the dosage regimen actually employed can vary widely and therefore can deviate from the dosage regimens set forth herein. The pharmaceutical agents which make up the combination therapy disclosed herein may be a combined dosage form or in separate dosage forms intended for substantially simultaneous administration. The pharmaceutical agents that make up the combination therapy may also be administered sequentially, with either therapeutic compound being administered by a regimen calling for two-step administration. The two-step administration regimen may call for sequential administration of the active agents or spaced-apart administration of the separate active agents. The time period between the multiple administration steps may range from, a few minutes to several hours, depending upon the properties of each pharmaceutical agent, such as potency, solubility, bioavailability, plasma half-life and kinetic profile of the pharmaceutical agent. Circadian variation of the target molecule concentration may also determine the optimal dose interval. In addition, the compounds described herein also may be used in combination with procedures that may provide additional or synergistic benefit to the patient. By way of example only, patients are expected to find therapeutic and/or prophylactic benefit in the methods described herein, wherein pharmaceutical composition of a compound disclosed herein and/or combinations with other therapeutics are combined with genetic testing to determine whether that individual is a carrier of a mutant gene that is known to be correlated with certain diseases or conditions. The compounds described herein and combination therapies can be administered before, during or after the occurrence of a disease, and the timing of administering the composition containing a compound can vary. Thus, for example, the compounds can be used as a prophylactic and can be administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease. The compounds and compositions can be administered to a subject during or as soon as possible after the onset of the symptoms. The administration of the compounds can be initiated within the first 48 hours of the onset of the symptoms, preferably within the first 48 hours of the onset of the symptoms, more preferably within the first 6 hours of the onset of the symptoms, and most preferably within 3 hours of the onset of the symptoms. The initial administration can be via any route practical, such as, for example, an intravenous injection, a bolus injection, infusion over about 5 minutes to about 5 hours, a pill, a capsule, transdermal patch, buccal delivery, and the like, or combination thereof. A compound is preferably administered as soon as is practicable after the onset of a disease is detected or suspected, and for a length of time necessary for the treatment of the disease, such as, for example, from 1 day to about 3 months. The length of treatment can vary for each subject, and the length can be determined using the known criteria. For example, the compound or a formulation containing the compound can be administered for at least 2 weeks, preferably about 1 month to about 5 years. Compounds and pharmaceutical compositions disclosed herein may be co-administered with one or more chemotherapeutics. Many chemotherapeutics are presently known in the art and can be used in combination with the compounds herein. In some embodiments, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, topoisomerase inhibitors, protein-protein interaction inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens. Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-peptide small molecules such as Gleevec® (Imatinib Mesylate), Velcade® (bortezomib), Casodex (bicalutamide), Iressa® (gefitinib), and Adriamycin as well as a host of chemotherapeutic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN™); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, Casodex™, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK®; razoxane; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxanes, e.g., paclitaxel (TAXOL™, Bristol-Myers Squibb Oncology, Princeton, N.J.) and docetaxel (TAXOTERE™, Rhone-Poulenc Rorer, Antony, France); retinoic acid; esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included as suitable chemotherapeutic cell conditioners are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, (Nolvadex™), raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; camptothecin-11 (CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO). Where desired, the compounds or pharmaceutical composition of the present invention can be used in combination with commonly prescribed anti-cancer drugs such as Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, ABVD, AVICINE, Abagovomab, Acridine carboxamide, Adecatumumab, 17-N-Allylamino-17-demethoxygeldanamycin, Alpharadin, Alvocidib, 3-Aminopyridine-2-carboxaldehyde thiosemicarbazone, Amonafide, Anthracenedione, Anti-CD22 immunotoxins, Antineoplastic, Antitumorigenic herbs, Apaziquone, Atiprimod, Azathioprine, Belotecan, Bendamustine, BIBW 2992, Biricodar, Brostallicin, Bryostatin, Buthionine sulfoximine, CBV (chemotherapy), Calyculin, cell-cycle nonspecific antineoplastic agents, Dichloroacetic acid, Discodermolide, Elsamitrucin, Enocitabine, Epothilone, Eribulin, Everolimus, Exatecan, Exisulind, Ferruginol, Forodesine, Fosfestrol, ICE chemotherapy regimen, IT-101, Imexon, Imiquimod, Indolocarbazole, Irofulven, Laniquidar, Larotaxel, Lenalidomide, Lucanthone, Lurtotecan, Mafosfamide, Mitozolomide, Nafoxidine, Nedaplatin, Olaparib, Ortataxel, PAC-1, Pawpaw, Pixantrone, Proteasome inhibitor, Rebeccamycin, Resiquimod, Rubitecan, SN-38, Salinosporamide A, Sapacitabine, Stanford V, Swainsonine, Talaporfin, Tariquidar, Tegafur-uracil, Temodar, Tesetaxel, Triplatin tetranitrate, Tris(2-chloroethyl)amine, Troxacitabine, Uramustine, Vadimezan, Vinflunine, ZD6126 or Zosuquidar. Embodiments herein further relate to methods for using a compound disclosed herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof), or a pharmaceutical composition provided herein, in combination with radiation therapy for inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the invention in this combination therapy can be determined as described herein. Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy. The term “brachytherapy,” as used herein, refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended without limitation to include exposure to radioactive isotopes (e.g., At-211, I-131, I-125, Y-90, Re-186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids. By way of non-limiting example, the radiation source can be a radionuclide, such as I-125, I-131, Yb-169, Ir-192 as a solid source, I-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays. The radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of I-125 or I-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive micro spheres. The compounds or pharmaceutical compositions herein are also used in combination with an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, antiproliferative agents, glycolysis inhibitors, or autophagy inhibitors. Anti-angiogenesis agents, such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloproteinase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the disclosure and pharmaceutical compositions described herein. Anti-angiogenesis agents include, for example, rapamycin, temsirolimus (CCI-779), everolimus (RAD001), sorafenib, sunitinib, and bevacizumab. Examples of useful COX-II inhibitors include CELEBREX™ (alecoxib), valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published Oct. 24, 1996), WO 96/27583 (published Mar. 7, 1996), European Patent Application No. 97304971.1 (filed Jul. 8, 1997), European Patent Application No. 99308617.2 (filed Oct. 29, 1999), WO 98/07697 (published Feb. 26, 1998), WO 98/03516 (published Jan. 29, 1998), WO 98/34918 (published Aug. 13, 1998), WO 98/34915 (published Aug. 13, 1998), WO 98/33768 (published Aug. 6, 1998), WO 98/30566 (published Jul. 16, 1998), European Patent Publication 606,046 (published Jul. 13, 1994), European Patent Publication 931, 788 (published Jul. 28, 1999), WO 90/05719 (published May 31, 1990), WO 99/52910 (published Oct. 21, 1999), WO 99/52889 (published Oct. 21, 1999), WO 99/29667 (published Jun. 17, 1999), PCT International Application No. PCT/IB98/01113 (filed Jul. 21, 1998), European Patent Application No. 99302232.1 (filed Mar. 25, 1999), Great Britain Patent Application No. 9912961.1 (filed Jun. 3, 1999), U.S. Provisional Application No. 60/148,464 (filed Aug. 12, 1999), U.S. Pat. No. 5,863,949 (issued Jan. 26, 1999), U.S. Pat. No. 5,861,510 (issued Jan. 19, 1999), and European Patent Publication 780,386 (published Jun. 25, 1997), all of which are incorporated herein in their entireties by reference. Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (e.g., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-ll, MMP-12, and MMP-13). Some specific examples of MMP inhibitors useful in the invention are AG-3340, RO 32-3555, and RS 13-0830. Autophagy inhibitors include, but are not limited to chloroquine, 3-methyladenine, hydroxychloroquine (Plaquenil™), bafilomycin A1, 5-amino-4-imidazole carboxamide riboside (AICAR), okadaic acid, autophagy-suppressive algal toxins which inhibit protein phosphatases of type 2A or type 1, analogues of cAMP, and drugs which elevate cAMP levels such as adenosine, LY204002, N6-mercaptopurine riboside, and vinblastine. In addition, antisense or siRNA that inhibits expression of proteins including but not limited to ATG5 (which are implicated in autophagy), may also be used. In some embodiments, the compounds described herein are formulated or administered in conjunction with liquid or solid tissue barriers also known as lubricants. Examples of tissue barriers include, but are not limited to, polysaccharides, polyglycans, seprafilm, interceed and hyaluronic acid. In some embodiments, medicaments which are administered in conjunction with the compounds described herein include any suitable drugs usefully delivered by inhalation for example, analgesics, e.g., codeine, dihydromorphine, ergotamine, fentanyl or morphine; anginal preparations, e.g., diltiazem; antiallergics, e.g., cromoglycate, ketotifen or nedocromil; anti-infectives, e.g., cephalosporins, penicillins, streptomycin, sulphonamides, tetracyclines or pentamidine; antihistamines, e.g., methapyrilene; anti-inflammatories, e.g., beclomethasone, flunisolide, budesonide, tipredane, triamcinolone acetonide or fluticasone; antitussives, e.g., noscapine; bronchodilators, e.g., ephedrine, adrenaline, fenoterol, formoterol, isoprenaline, metaproterenol, phenylephrine, phenylpropanolamine, pirbuterol, reproterol, rimiterol, salbutamol, salmeterol, terbutalin, isoetharine, tulobuterol, orciprenaline or (−)-4-amino-3,5-dichloro-α-[[[6-[2-(2-pyridinyl)ethoxy]hexyl]-amino]methyl]benzenemethanol; diuretics, e.g., amiloride; anticholinergics e.g., ipratropium, atropine or oxitropium; hormones, e.g., cortisone, hydrocortisone or prednisolone; xanthines e.g., aminophylline, choline theophyllinate, lysine theophyllinate or theophylline; and therapeutic proteins and peptides, e.g., insulin or glucagon. It will be clear to a person skilled in the art that, where appropriate, the medicaments are used in the form of salts (e.g., as alkali metal or amine salts or as acid addition salts) or as esters (e.g., lower alkyl esters) or as solvates (e.g., hydrates) to optimize the activity and/or stability of the medicament. Other exemplary therapeutic agents useful for a combination therapy include but are not limited to agents as described above, radiation therapy, hormone antagonists, hormones and their releasing factors, thyroid and antithyroid drugs, estrogens and progestins, androgens, adrenocorticotropic hormone; adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of adrenocortical hormones, insulin, oral hypoglycemic agents, and the pharmacology of the endocrine pancreas, agents affecting calcification and bone turnover: calcium, phosphate, parathyroid hormone, vitamin D, calcitonin, vitamins such as water-soluble vitamins, vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K, and E, growth factors, cytokines, chemokines, muscarinic receptor agonists and antagonists; anticholinesterase agents; agents acting at the neuromuscular junction and/or autonomic ganglia; catecholamines, sympathomimetic drugs, and adrenergic receptor agonists or antagonists; and 5-hydroxytryptamine (5-HT, serotonin) receptor agonists and antagonists. Other suitable therapeutic agents for coadministration with compounds herein also include agents for pain and inflammation such as histamine and histamine antagonists, bradykinin and bradykinin antagonists, 5-hydroxytryptamine (serotonin), lipid substances that are generated by biotransformation of the products of the selective hydrolysis of membrane phospholipids, eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, nonsteroidal anti-inflammatory agents, analgesic-antipyretic agents, agents that inhibit the synthesis of prostaglandins and thromboxanes, selective inhibitors of the inducible cyclooxygenase, selective inhibitors of the inducible cyclooxygenase-2, autacoids, paracrine hormones, somatostatin, gastrin, cytokines that mediate interactions involved in humoral and cellular immune responses, lipid-derived autacoids, eicosanoids, β-adrenergic agonists, ipratropium, glucocorticoids, methylxanthines, sodium channel blockers, opioid receptor agonists, calcium channel blockers, membrane stabilizers and leukotriene inhibitors. Additional therapeutic agents contemplated for co-administration with compounds and compositions herein include diuretics, vasopressin, agents affecting the renal conservation of water, rennin, angiotensin, agents useful in the treatment of myocardial ischemia, anti-hypertensive agents, angiotensin converting enzyme inhibitors, β-adrenergic receptor antagonists, agents for the treatment of hypercholesterolemia, and agents for the treatment of dyslipidemia. Other therapeutic agents contemplated for co-administration with compounds and compositions herein include drugs used for control of gastric acidity, agents for the treatment of peptic ulcers, agents for the treatment of gastroesophageal reflux disease, prokinetic agents, antiemetics, agents used in irritable bowel syndrome, agents used for diarrhea, agents used for constipation, agents used for inflammatory bowel disease, agents used for biliary disease, agents used for pancreatic disease. Therapeutic agents used to treat protozoan infections, drugs used to treat Malaria, Amebiasis, Giardiasis, Trichomoniasis, Trypanosomiasis, and/or Leishmaniasis, and/or drugs used in the chemotherapy of helminthiasis. Other therapeutic agents include antimicrobial agents, sulfonamides, trimethoprim-sulfamethoxazole quinolones, and agents for urinary tract infections, penicillins, cephalosporins, and other, β-lactam antibiotics, an agent comprising an aminoglycoside, protein synthesis inhibitors, drugs used in the chemotherapy of tuberculosis,Mycobacterium aviumcomplex disease, and leprosy, antifungal agents, antiviral agents including nonretroviral agents and antiretroviral agents. Examples of therapeutic antibodies that can be combined with a compound herein include but are not limited to anti-receptor tyrosine kinase antibodies (cetuximab, panitumumab, trastuzumab), anti CD20 antibodies (rituximab, tositumomab), and other antibodies such as alemtuzumab, bevacizumab, and gemtuzumab. Moreover, therapeutic agents used for immunomodulation, such as immunomodulators, immunosuppressive agents, tolerogens, and immunostimulants are contemplated by the methods herein. In addition, therapeutic agents acting on the blood and the blood-forming organs, hematopoietic agents, growth factors, minerals, and vitamins, anticoagulant, thrombolytic, and antiplatelet drugs. Further therapeutic agents that can be combined with a compound herein are found in Goodman and Gilman's “The Pharmacological Basis of Therapeutics” Tenth Edition edited by Hardman, Limbird and Gilman or the Physician's Desk Reference, both of which are incorporated herein by reference in their entirety. In some embodiments, a compound described herein is co-administered with another therapeutic agent effective in treating brain cancer, such as glioblastoma or astrocytoma. In some embodiments, the other therapeutic agent may be bevacizumab, carmustine (e.g., carmustine wafer), cisplatin, everolimus, lomustine, procarbazine, temozolomide, vincristine, or any combination thereof (e.g., a combination of procarbazine hydrochloride, lomustine, and vincristine sulfate). In some embodiments, a compound described herein is co-administered with one or more therapeutic agents approved for the treatment of a sarcoma, such as adriamycin, bevacizumab, carboplatin, cisplatin, cyclophosphamide, dacarbazine, dactinomycin, docetaxel, doxorubicin (e.g., doxorubicin hydrochloride liposome), epirubicin, eribulin, etoposide, gemcitabine, ifosfamide, imatinib, ixabepilone, methotrexate, paclitaxel, pazopanib, pomalidomide, recombinant interferon alfa-2b, tazemetostat, temozolomide, topotecan, trabectedin, vinblastine, vincristine, vinorelbine, or any combination thereof. In some embodiments, a compound described herein is co-administered with one or more therapeutic agents approved for the treatment of colorectal cancer, such as 5-fluorouracil, bevacizumab, capecitabine, cetuximab, ipilmumab, irinotecan, leucovorin, nivolumab, oxaliplatin, panitumumab, pembrolizumab, ramucirumab, regorafenib, tipiracil, trifluridine, ziv-afibercept, or any combination thereof. In some embodiments, a compound described herein is co-administered with one or more therapeutic agents approved for the treatment of lung cancer, such as non-small cell lung cancer. In such embodiments, the other therapeutic agent may be afatinib, alectinib, atezolizumab, bevacizumab, brigatinib, capmatinib, carboplatin, ceritinib, cisplatin, crizotinib, dabrafenib, dacomitinib, docetaxel, doxorubicin, durvalumab, entrectinib, erlotinib, everolimus, gefitinib, gemcitabine, ipilimumab, lorlatinib, mechlorethamine, methotrexate, necitumumab, nivolumab, osimertinib, paclitaxel, pembrolizumab, pemetrexed, ramucirumab, selpercatinib, trametinib, vinorelbine, or any combination thereof. In some embodiments, a compound described herein is co-administered with one or more therapeutic agents approved for the treatment of gastric cancer, such as 5-fluorouracil, capecitabine, carboplatin, cisplatin, docetaxel, epirubicin, irinotecan, oxaliplatin, paclitaxel, trifluridine, tipiracil, trastuzumab, or any combination thereof. In some embodiments, a compound described herein is co-administered with one or more alkylating agents (e.g., for the treatment of cancer) selected from, for example, nitrogen mustard N-oxide, cyclophosphamide, ifosfamide, thiotepa, ranimustine, nimustine, temozolomide, altretamine, apaziquone, brostallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, mafosfamide, bendamustin, mitolactol, cisplatin, carboplatin, eptaplatin, lobaplatin, nedaplatin, oxaliplatin, and satraplatin. In some embodiments, a compound described herein is co-administered with one or more anti-metabolites (e.g., for the treatment of cancer) selected from, for example, methotrexate, 6-mercaptopurineriboside, mercaptopurine, 5-fluorouracil, tegafur, doxifluridine, carmofur, cytarabine, cytarabine ocfosfate, enocitabine, gemcitabine, fludarabin, 5-azacitidine, capecitabine, cladribine, clofarabine, decitabine, eflornithine, ethynylcytidine, cytosine arabinoside, hydroxyurea, melphalan, nelarabine, nolatrexed, ocfosf[iota]te, disodium premetrexed, pentostatin, pelitrexol, raltitrexed, triapine, trimetrexate, vidarabine, vincristine, and vinorelbine; In some embodiments, a compound described herein is co-administered with one or more hormonal therapy agents (e.g., for the treatment of cancer) selected from, for example, exemestane, Lupron, anastrozole, doxercalciferol, fadrozole, formestane, abiraterone acetate, finasteride, epristeride, tamoxifen citrate, fulvestrant, Trelstar, toremifene, raloxifene, lasofoxifene, letrozole, sagopilone, ixabepilone, epothilone B, vinblastine, vinflunine, docetaxel, and paclitaxel; In some embodiments, a compound described herein is co-administered with one or more cytotoxic topoisomerase inhibiting agents (e.g., for the treatment of cancer) selected from, for example, aclarubicin, doxorubicin, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflomotecan, irinotecan, topotecan, edotecarin, epimbicin, etoposide, exatecan, gimatecan, lurtotecan, mitoxantrone, pirambicin, pixantrone, rubitecan, sobuzoxane, tafluposide, etc. In some embodiments, a compound described herein is co-administered with one or more anti-angiogenic compounds (e.g., for the treatment of cancer) selected from, for example, acitretin, aflibercept, angiostatin, aplidine, asentar, axitinib, recentin, bevacizumab, brivanib alaninat, cilengtide, combretastatin, DAST, endostatin, fenretinide, halofuginone, pazopanib, ranibizumab, rebimastat, removab, revlimid, sorafenib, vatalanib, squalamine, sunitinib, telatinib, thalidomide, ukrain, and vitaxin. In some embodiments, a compound described herein is co-administered with one or more antibodies (e.g., for the treatment of cancer) selected from, for example, trastuzumab, cetuximab, bevacizumab, rituximab, ticilimumab, ipilimumab, lumiliximab, catumaxomab, atacicept, oregovomab, and alemtuzumab. In some embodiments, a compound described herein is co-administered with one or more VEGF inhibitors (e.g., for the treatment of cancer) selected from, for example, sorafenib, DAST, bevacizumab, sunitinib, recentin, axitinib, aflibercept, telatinib, brivanib alaninate, vatalanib, pazopanib, and ranibizumab. In some embodiments, a compound described herein is co-administered with one or more EGFR inhibitors (e.g., for the treatment of cancer) selected from, for example, cetuximab, panitumumab, vectibix, gefitinib, erlotinib, and Zactima. In some embodiments, a compound described herein is co-administered with one or more HER2 inhibitors (e.g., for the treatment of cancer) selected from, for example, lapatinib, tratuzumab, and pertuzumab; CDK inhibitor is selected from roscovitine and flavopiridol; In some embodiments, a compound described herein is co-administered with one or more proteasome inhibitors (e.g., for the treatment of cancer) selected from, for example, bortezomib and carfilzomib. In some embodiments, a compound described herein is co-administered with one or more serine/threonine kinase inhibitors (e.g., for the treatment of cancer), for example, MEK inhibitors and Raf inhibitors such as sorafenib. In some embodiments, a compound described herein is co-administered with one or more tyrosine kinase inhibitors (e.g., for the treatment of cancer) selected from, for example, dasatinib, nilotibib, DAST, bosutinib, sorafenib, bevacizumab, sunitinib, AZD2171, axitinib, aflibercept, telatinib, imatinib mesylate, brivanib alaninate, pazopanib, ranibizumab, vatalanib, cetuximab, panitumumab, vectibix, gefitinib, erlotinib, lapatinib, tratuzumab and pertuzumab. In some embodiments, a compound described herein is co-administered with one or more androgen receptor antagonists (e.g., for the treatment of cancer) selected from, for example, nandrolone decanoate, fluoxymesterone, Android, Prostaid, andromustine, bicalutamide, flutamide, apocyproterone, apoflutamide, chlormadinone acetate, Androcur, Tabi, cyproterone acetate, and nilutamide. In some embodiments, a compound described herein is co-administered with one or more aromatase inhibitors (e.g., for the treatment of cancer) selected from, for example, anastrozole, letrozole, testolactone, exemestane, aminoglutethimide, and formestane. In some embodiments, a compound described herein is co-administered with one or more other anti-cancer agents including, e.g., alitretinoin, ampligen, atrasentan bexarotene, borte-zomib, bosentan, calcitriol, exisulind, fotemustine, ibandronic acid, miltefosine, mitoxantrone, 1-asparaginase, procarbazine, dacarbazine, hydroxycarbamide, pegaspargase, pentostatin, tazaroten, velcade, gallium nitrate, canfosfamide, darinaparsin, and tretinoin. In a preferred embodiment, the compounds of the present disclosure may be used in combination with chemotherapy (e.g., cytotoxic agents), anti-hormones and/or targeted therapies such as other kinase inhibitors, mTOR inhibitors and angiogenesis inhibitors. In embodiments in which the compounds and pharmaceutical compositions herein are used for the treatment or prevention of non-cancer diseases and/or conditions, the compounds and pharmaceutical compositions herein may be co-administered with therapeutics and/or therapies known in the field to be appropriate for the treatment of such diseases and/or conditions. Kits For use in the therapeutic applications described herein, kits and articles of manufacture are also provided, which include a compound or pharmaceutical composition described herein (e.g., a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof). In some embodiments, such kits comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers are formed from a variety of materials such as glass or plastic. The articles of manufacture provided herein contain packaging materials. Packaging materials for use in packaging pharmaceutical products include those found in, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment. For example, in some embodiments the container(s) includes a compound of formula (I), (IIa), (IIb), or (IIc), or a pharmaceutically acceptable salt thereof, optionally in a composition or in combination with another agent as disclosed herein. The container(s) optionally have a sterile access port (for example the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). Such kits optionally comprising a compound with an identifying description or label or instructions relating to its use in the methods described herein. For example, a kit typically includes one or more additional containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein. Non-limiting examples of such materials include, but not limited to, buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included. A label is optionally on or associated with the container. For example, a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself, a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In addition, a label is used to indicate that the contents are to be used for a specific therapeutic application. In addition, the label indicates directions for use of the contents, such as in the methods described herein. In certain embodiments, the pharmaceutical composition is presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein. The pack, for example, contains metal or plastic foil, such as a blister pack. Or, the pack or dispenser device is accompanied by instructions for administration. Or, the pack or dispenser is accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, is the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. In some embodiments, compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. EXAMPLES Abbreviations used in the following examples include the following: ACN is acetonitrile; Boc is tert-butyloxycarbonyl; DCM is dichloromethane; DIPEA is N,N-diisopropylethylamine; DMA is dimethylacetamide; dppf is 1,1′-bis(diphenylphosphino)ferrocene; EtOAc is ethyl acetate; HATU is (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate; MeOH is methanol; RPH refers to reversed phase chromatography; RT is room temperature; and TFA is trifluoroacetic acid. General Synthesis Methods General Procedure A: To a screw cap 10 dr vial were added carboxylic acid (1.1 eq.), DIPEA (2 eq., in case of amine hydrochloride 3 eq.), HATU (1.2 eq) and anhydrous DCM (5 ml). The mixture was stirred for 15 min at RT, then corresponding amine or its hydrochloride (1.2 eq) was added, the vial was sealed and the reaction mixture was heated at 45° C. overnight. After cooling to the RT mixture was diluted with DCM (20 mL), and sequentially washed with water, sat. aqueous NaHCO3, and brine. Organic phase was dried over Na2SO4, concentrated in vacuo and purified by silica gel column chromatography (DCM: EtOAc or DCM: MeOH) and then (if needed) by RPH chromatography (10-100% gradient of MeOH in water) providing a titled compound. General Procedure B: To a solution of corresponding methyl ester (1 mmol, 1 eq.) in MeOH (2 mL/mmol) was added 1M LiOH solution (2 eq.) and the mixture was stirred at RT for 4 h (for proline esters) or overnight (for aromatic esters). The mixture was concentrated in vacuo and the crude was diluted with water (5 mL) and acidified to pH 4 with 1M HCl. Obtained mixture was extracted with EtOAc (3*10 mL), and combined organics were washed with brine (3*10 mL), dried over Na2SO4and concentrated providing the product that either was used directly in the next step without further purification or was purified by silica gel column chromatography. General Procedure C: To a de-gassed suspension of zinc powder (217 mg, 3.338 mmol, 1.8 eq.) in DMA (2 mL) in the screw cap vial was added drop-wise a mixture of chlorotrimethylsilane (67.3 μL, 57.6 mg, 0.53 mmol, 0.3 eq.) and 1,2-dibromoethane (45.9 μL, 99.6 mg, 0.53 mmol, 0.3 eq.) and the resultant mixture was stirred at room temperature under Ar for 15 minutes. To this mixture was then added dropwise neat 3-iodoazetidine-1-carboxylic acid tert-butyl ester (753 mg, 2.661 mmol, 1.4 eq.) and the resultant mixture was stirred at room temperature for 15 minutes. In a separate vial, PdCl2(dppf)*DCM (65.2 mg, 0.08 mmol, 0.04 eq.), and copper iodide (30 mg, 0.157 mmol, 0.08 mmol) were added to a degassed solution of corresponding het(aryl)bromide (1.862 mmol, 1 eq.) in DMA (1 mL). After stirring for 30 minutes, the zinc suspension above was added to the solution suspension of het(aryl)bromide, PdCl2(dppf)*DCM and copper iodide and the reaction mixture was allowed to stir at 80° C. for 2 hours under argon. The resultant mixture was cooled to RT diluted with EtOAc and filtered through the pad with Celite, the pad was washed with EtOAc, and collected organics were washed with mixture of saturated ammonium chloride solution and ammonium hydroxide (15:1). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel flash chromatography (0-100% ethyl acetate in hexanes), fractions with the corresponding pick in the LC-MS were combined, concentrated and used in the next step without further purification. General Procedure D: To a solution of Boc-protected substituted azetidine (1 mmol) in the 3 mL of 1,4-dioxane 4M HCl in 1,4-dioxane (3 mL) was added dropwise, and the mixture was stirred overnight at RT. After this all volatiles were removed under reduced pressure, the residue was triturated with dry ACN, and ACN was decanted, remained solid was dried in vacuo providing the corresponding dihydrochloride as a white solid with quantitative yield. General Procedure E: To a degassed suspension of boronic acid or boronic acid pinacol ester (1.3 eq., 0.65 mmol), bromide (1 eq., 0.5 mmol), NaHCO3(3 eq., 1.5 mmol) in mixture 1,4-dioxane:water=10:1 PdCl2(dppf)*DCM (0.025 mmol) was added in one portion. Obtained suspension was degassed one time more, refilled with Ar and allowed to stir at 80° C. overnight under argon. The resultant mixture was cooled to RT diluted with EtOAc and filtered through the pad with Celite, the pad was washed with EtOAc, and collected organics were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel flash chromatography (0-100% ethyl acetate in hexanes then 0-20% methanol in dichloromethane), fractions with the corresponding pick in the LC-MS were combined, concentrated and the residue was re-dissolved in the DCM (5 mL). To this mixture TFA (30 eq.) was added dropwise at 0° C., and the mixture was stirred at 0° C. for 60 min. The mixture was concentrated in vacuo and the crude was triturated with 7N ammonia in methanol and concentrated again. Obtained residue was purified by silica gel column chromatography (0-100% Hexane/EtOAc to DCM/MeOH) then RPH (0-100% MeOH/Water) to afford the titled compound as an off white solid. General Procedure F: To a screw cap 10 dr vial were added amine (1 eq.), DIPEA (2 eq.) and anhydrous DCM (5 ml). The mixture was stirred for 15 min at 0° C., then corresponding acylchloride was added, the vial was sealed and the reaction mixture was stirred at RT overnight. After cooling to the RT mixture was diluted with DCM (20 mL), and sequentially washed with water, sat. aqueous NaHCO3, and brine. Organic phase was dried over sodium sulfate, concentrated in vacuo and the residue was re-dissolved in the DCM (2 mL) at 0° C. and TFA (30 eq.) was added dropwise, and the mixture was stirred at 0° C. for 60 min. The mixture was concentrated in vacuo and the crude was triturated with 7N ammonia in methanol and concentrated again. Obtained residue was purified by silica gel column chromatography (0-100% Hexane/EtOAc to DCM/MeOH) then RPH (0-100% MeOH/Water) to afford the titled compound as an off white solid. General Procedure G: The Boc-protected compound was dissolved in the DCM (2 mL) at 0° C. and TFA (30 eq.) was added dropwise. The mixture was stirred at 0° C. for 60 min. and then was concentrated in vacuo and the crude was triturated with 7N ammonia in methanol and concentrated again. Obtained residue was purified by silica gel column chromatography (0-100% Hexane/EtOAc to DCM/MeOH) then RPH (0-100% MeOH/Water) to afford the titled compound as an off white solid. Example 1 Compound Syntheses—Monomers Compound 14: N-((5-(pyrrolidine-1-carbonyl)thiophen-2-yl)methyl)azetidine-3-carboxamide Synthesized according to general procedure A with using (5-aminothiophen-2-yl)(pyrrolidin-1-yl)methanone and 1-Boc-azetidine-3-carboxylic acid, and general procedure G to afford Compound 14 (10 mg, 72%) as an off white solid.1H NMR (600 MHz, CD3OD): δ 7.46 (d, J=3.8 Hz, 1H), 7.00 (d, J=3.8 Hz, 1H), 4.56 (s, 2H), 3.96 (br.s, 2H), 3.78 (br.s, 4H), 3.59 (br.s, 3H), 2.02 (br.s, 2H), 1.95 (br.s, 2H).13C NMR (125 MHz, CD3OD): δ 174.9, 163.6, 148.3, 139.1, 131.4, 127.1, 118.1, 68.1, 50.3, 49.6, 39.0, 27.6, 24.9. HR-MS (ESI): [M+H+] calculated 294.1271, found 294.1270. Compound 37: N-(5-(pyrrolidine-1-carbonyl)thiophen-2-yl)azetidine-2-carboxamide Synthesized according to general procedure A with using (5-aminothiophen-2-yl)(pyrrolidin-1-yl)methanone and (rac)-1-Boc-azetidine-2-carboxylic acid, and general procedure G to afford Compound 20 (13 mg, 79%) as an off white solid.1H NMR (600 MHz, CD3OD): δ 7.44 (d, J=4.2 Hz, 1H), 6.79 (d, J=4.2 Hz, 1H), 4.46 (dd, J=9.1, 7.1 Hz, 1H), 3.82 (s, 2H), 3.69 (q, J=7.9 Hz, 1H), 3.61 (s, 2H), 3.49 (td, J=8.5, 5.1 Hz, 1H), 2.70 (ddt, J=8.9, 6.2, 4.4 Hz, 1H), 2.48-2.36 (m, 1H), 2.04 (s, 2H), 1.96 (s, 2H).13C NMR (125 MHz, CD3OD): δ 173.0, 164.3, 145.5, 130.6, 129.9, 113.4, 60.0, 50.2, 47.5 (from HSQC) 44.6, 27.6, 26.8, 24.9. HR-MS (ESI): [M+H+] calculated 280.1114, found 280.1117. Compound 44: (S)—N-(5-(pyrrolidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide Synthesized according to general procedure A, from corresponding amine and N-Boc-L-proline, and general procedure G to afford the titled compound as an off white solid (18 mg, 83%).1H NMR MeOD (600 MHz): δ 7.44 (d, J=4.2 Hz, 1H), 6.78 (d, J=4.2 Hz, 1H), 3.89-3.77 (m, 3H), 3.60 (s, 2H), 3.06 (dt, J=10.5, 6.5 Hz, 1H), 2.97 (dt, J=10.5, 6.5 Hz, 1H), 2.19 (dt, J=12.7, 6.8 Hz, 1H), 2.04 (s, 2H), 1.95 (s, 2H), 1.91-1.85 (m, 1H), 1.82-1.75 (m, 2H);13C NMR MeOD (150 MHz), mixture of rotamers (1:1): δ 174.1, 164.4, 145.5, 130.5, 129.9, 113.3, 61.6, 49.8, 48.1, 32.0, 27.6, 27.0, 24.9; HR-ESI-MS: C14H20N3O2S [M+H]+m/z calculated 294.1271, found 294.1275. Compound 85. (S)—N-(5-(3-(thiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide Step 1. tert-butyl (S)-2-((5-(methoxycarbonyl)thiophen-2-yl)carbamoyl)pyrrolidine-1-carboxylate Synthesized according to general procedure A, from methyl 5-amino-2-thiophenecarboxylate and N-Boc-L-proline. Obtained residue was purified by silica gel column chromatography (0-100% DCM/EtOAc) to afford the titled compound as a semi-solid (182 mg, 73%).1H NMR DMSO-d6(600 MHz) (mixture of rotamers 2:1): δ 11.66 (s, 1H), 7.60 (d, J=4.1 Hz, 1H), 6.75 (d, J=4.1 Hz, 1H), 4.34-4.18 (m, 1H), 3.77 (s, 3H), 3.50-3.40 (m, 1H), 3.40-3.32 (m, 1H), 2.30-2.13 (m, 1H), 1.95-1.76 (m, 3H), 1.40 (s, 3H), 1.23 (s, 6H);13C NMR DMSO-d6(150 MHz) (mixture of rotamers 2:1): δ 170.7, 170.2, 162.5, 153.6, 152.9, 146.3, 146.2, 132.0, 131.9, 121.9, 121.8, 112.0, 111.9, 78.9, 78.7, 59.8, 59.4, 51.7, 46.7, 46.8, 30.8, 30.6, 30.0, 28.1, 27.9, 27.8, 24.0, 23.4; HR-ESI-MS: C16H23N2O5S [M+H]+m/z calculated 355.1322, found 355.1336. Step 2. (S)-5-(1-(tert-butoxycarbonyl)pyrrolidine-2-carboxamido)thiophene-2-carboxylic Acid Synthesized according to general procedure B, from tert-butyl (S)-2-((5-(methoxycarbonyl)thiophen-2-yl)carbamoyl)pyrrolidine-1-carboxylate. Obtained residue was purified by silica gel column chromatography (0-100% DCM/EtOAc) to afford the titled compound as a semi-solid (120 mg, 93%).1H NMR DMSO-d6(600 MHz) (mixture of rotamers 2:1): δ 12.54 (s, 1H), 11.56 (s, 1H), 7.51 (d, J=4.1 Hz, 1H), 6.72 (d, J=4.1 Hz, 1H), 4.28 (dd, J=8.2, 4.8 Hz, 0.3H), 4.21 (dd, J=8.2, 4.8 Hz, 0.7H), 3.51-3.40 (m, 1H), 3.40-3.31 (m, 1H, overlapped with HDO pick), 2.29-2.14 (m, 1H), 1.96-1.79 (m, 3H), 1.40 (s, 3H), 1.24 (s, 6H);13C NMR DMSO-d6(150 MHz) (mixture of rotamers 2:1 based on1H NMR): δ 170.5, 170.1, 163.6, 153.6, 152.9, 145.8, 145.7, 131.4, 131.4, 123.8, 123.7, 111.8, 111.8, 78.9, 78.7, 59.8, 59.4, 46.7, 46.5, 30.9, 30.1, 28.1, 27.8, 24.0, 23.4; HR-ESI-MS: C15H21N2O5S [M+H]+m/z calculated 341.1166, found 341.1179. Step 3. (S)—N-(5-(3-(thiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide (Compound 85) Synthesized according to general procedure A, from (S)-5-(1-(tert-butoxycarbonyl)pyrrolidine-2-carboxamido)thiophene-2-carboxylic acid and 2-(azetidin-3-yl)thiazole dihydrochloride. Obtained residue was used in the next step without further purification. To a solution of the residue from the previous step in the DCM (2 mL) at 0° C. TFA (0.5 mL) was added dropwise, and the mixture was stirred at 0° C. for 30 min. After this all volatiles were removed under reduced pressure, the residue was triturated with 7N ammonia solution in methanol, re-concentrated and purified by silica gel column chromatography (0-100% DCM/MeOH+0.5% ammonia (v/v)) to afford the titled compound as an off-white solid (9 mg, 75%).1H NMR MeOD (600 MHz) δ: 7.80 (d, J=3.3 Hz, 1H), 7.56 (d, J=3.3 Hz, 1H), 7.40 (d, J=4.2 Hz, 1H), 6.80 (d, J=4.2 Hz, 1H), 4.96 (s, 1H), 4.67 (s, 2H), 4.41 (m, 2H), 3.83 (dd, J=8.7, 5.9 Hz, 1H), 3.05 (dt, J=10.5, 6.5 Hz, 1H), 2.97 (dt, J=10.5, 6.5 Hz, 1H), 2.20 (td, J=15.6, 12.7, 7.3 Hz, 1H), 1.88 (td, J=15.6, 12.7, 7.3 Hz, 1H), 1.80 (p, J=6.9 Hz, 2H);13C NMR MeOD (150 MHz) δ: 174.3, 172.1, 165.7, 146.3, 143.8, 130.4, 127.5, 120.9, 113.6, 61.6, 60.3 (azetidine CH2, identified from HSQC spectrum), 56.8 (azetidine CH2, identified from HSQC spectrum), 48.1, 33.3, 32.0, 27.0. HR-ESI-MS: C16H19N4O2S2[M+H]+m/z calculated 363.0944, found 363.0952. Compound 90. ((S)—N-(5-(3-(5-phenylthiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide Step 1. 2-(azetidin-3-yl)-5-phenylthiazole Hydrochloride Synthesized according to general procedures C and D to afford 2-(azetidin-3-yl)-5-phenylthiazole hydrochloride (70 mg, 15%) as an off white solid.1H NMR (600 MHz, DMSO-d6) δ 9.51 (s, 1H), 9.22 (s, 1H), 8.24 (s, 1H), 7.70-7.63 (m, 2H), 7.46 (t, J=7.7 Hz, 2H), 7.41-7.34 (m, 1H), 4.50 (p, J=8.3 Hz, 1H), 4.38-4.28 (m, 2H), 4.26-4.18 (m, 2H).13C NMR (150 MHz, DMSO-d6): δ 166.6, 139.4, 138.5, 130.6, 129.3, 128.5, 126.4, 50.6, 33.5. MS (m/z) [M+H+]: calculated 217, found 217. (S)—N-(5-(3-(5-phenylthiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide (Compound 91) Synthesized according to general procedure A with using (S)-5-(1-(tert-butoxycarbonyl)pyrrolidine-2-carboxamido)thiophene-2-carboxylic acid and 2-(azetidin-3-yl)-5-phenylthiazole hydrochloride, and general procedure G to afford Compound 90 (8 mg, 79%) as an off white solid.1H NMR (600 MHz, CD3OD) δ 7.91 (s, 1H), 7.55 (d, J=9.6 Hz, 1H), 7.41-7.37 (m, 3H), 7.33 (t, J=7.4 Hz, 1H), 6.75 (d, J=4.2 Hz, 1H), 4.94 (s, 1H), 4.81 (s, 2H), 4.48 (s, 2H), 4.33 (tt, J=8.8, 5.8 Hz, 1H), 3.86-3.80 (m, 1H), 3.07 (dt, J=11.1, 6.5 Hz, 1H), 3.04-2.96 (m, 1H), 2.28-2.16 (m, 1H), 1.92 (dq, J=12.9, 6.6 Hz, 1H), 1.81 (p, J=6.9 Hz, 2H). HR-MS (ESI): [M+H+] calculated 439.1257, found 439.1259. Compound 123. (S)—N-(5-(3-(5-(4-(acetamidomethyl)phenyl)thiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide Synthesized according to general procedure F with using (S)—N-(5-(3-(5-(4-(aminomethyl)phenyl)thiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide (obtained by general procedure E Boc-intermediate was used directly) and acetylchloride to afford Compound 123 (8 mg, 86%) as an off white solid.1H NMR (600 MHz, CD3OD+30% DMSO-d6) δ 8.07 (s, 1H), 7.64-7.58 (m, 2H), 7.39-7.33 (m, 3H), 6.84 (d, J=4.2 Hz, 1H), 4.90 (br.s, 1H), 4.65 (br.s, 2H), 4.40-4.36 (m, 1H), 4.34 (s, 3H), 3.90 (dd, J=8.8, 5.9 Hz, 1H), 3.02 (dtd, J=17.1, 10.5, 6.7 Hz, 2H), 2.19 (dq, J=12.6, 7.5 Hz, 1H), 1.96 (s, 3H), 1.93-1.84 (m, 1H), 1.78 (p, J=7.0 Hz, 2H).13C NMR (150 MHz, CD3OD+30% DMSO-d6): δ 173.2, 172.2, 170.7, 165.0, 146.0, 141.1, 140.6, 139.5, 131.1, 130.0, 129.5, 128.0, 127.8, 113.8, 61.5, 60.0, 56.3, 48.0, 43.5, 33.6, 31.7, 26.8, 23.1. HR-MS (ESI): [M+H+] calculated 510.1628, found 510.1628. Compound 125. (S)—N-(5-(3-(5-(4-(aminomethyl)phenyl)thiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide Step 1. tert-Butyl 3-(5-bromothiazol-2-yl)azetidine-1-carboxylate Corresponding Boc-intermediate was synthesized according to general procedure C and was used in the next step without further purification. To a solution of tert-butyl 3-(thiazol-2-yl)azetidine-1-carboxylate (1 g, 4.17 mmol, 1 eq.) in 20 mL of anhydrous DMF NBS (890 mg, 5 mmol, 1.2 eq.) was added portion wise at RT. The mixture was stirred 12 h at RT, poured on ice and extracted with EtOAc (3*50 mL). Collected organics were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel flash chromatography (0-60% ethyl acetate in hexanes) to afford tert-butyl 3-(5-bromothiazol-2-yl)azetidine-1-carboxylate (665 mg, 50%) as a clear oil.1H NMR (600 MHz, CDCl3) δ 7.62 (s, 1H), 4.33 (t, J=8.6 Hz, 2H), 4.14 (dd, J=8.6, 5.9 Hz, 2H), 4.02 (tt, J=8.7, 5.9 Hz, 1H), 1.45 (s, 9H).13C NMR (150 MHz, CDCl3): δ 172.3, 156.3, 144.1, 108.6, 80.1, 55.5, 32.4, 28.5. MS (m/z) [M+H+]: calculated 262, 264, found 262, 264. Step 2. tert-butyl (S)-2-((5-(3-(5-bromothiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)carbamoyl)pyrrolidine-1-carboxylate Synthesized according to general procedure A with using (S)-5-(1-(tert-butoxycarbonyl)pyrrolidine-2-carboxamido)thiophene-2-carboxylic acid and 3-(5-bromothiazol-2-yl)azetidin-1-ium trifluoroacetate (obtained by treating of tert-butyl 3-(5-bromothiazol-2-yl)azetidine-1-carboxylate solution in DCM with TFA (30 eq.) at 0° C., stirring at 0° C. for 60 min and removing of all volatiles in vacuo. Obtained residue was used in the HATU-assisted coupling reaction without further purification) to afford tert-butyl (S)-2-((5-(3-(5-bromothiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)carbamoyl)pyrrolidine-1-carboxylate (58 mg, 57%) as an off white solid.1H NMR (600 MHz, CDCl3, mixture of rotamers) δ 10.72 (s, 1H), 7.61 (s, 1H), 7.31 (br.s, 1H), 6.54 (br.s, 0.5H), 6.45 (br.s, 0.5H), 4.71 (br.s, 2H), 4.49 (br.s, 2H), 4.20 (tt, J=8.8, 5.9 Hz, 1H), 3.78-3.68 (m, 1H), 3.46 (s, 1H), 3.36 (s, 1H), 3.24-3.14 (m, 1H), 1.98 (s, 1H), 1.91 (s, 2H), 1.48 (s, 9H). MS (m/z) [M+H+]: calculated 541 and 543, found 541 and 543. Step 3. (S)—N-(5-(3-(5-(4-(aminomethyl)phenyl)thiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide (Compound 125) Synthesized according to general procedure E with using tert-butyl (S)-2-((5-(3-(5-bromothiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)carbamoyl)pyrrolidine-1-carboxylate and (4-aminomethylphenyl)boronic acid hydrochloride, obtained Boc-analog was further re-dissolved in DCM (2 mL) at 0° C. and treated with TFA (0.5 mL) the mixture was stirred at 0° C. for 30 min. After this, all volatiles were removed under reduced pressure, the residue was triturated with 7N ammonia solution in methanol, re-concentrated and purified by silica gel column chromatography (0-100% DCM/MeOH+0.5% ammonia (v/v)) to afford the titled compound as an off-white solid (8 mg, 53%).1H NMR (600 MHz, CD3OD) δ 8.09 (s, 1H), 7.71 (d, J=7.8 Hz, 2H), 7.55 (d, J=7.9 Hz, 2H), 7.41 (d, J=4.1 Hz, 1H), 6.86 (d, J=4.0 Hz, 1H), 4.99 (s, 1H), 2H are in the pick of water, present in HSQC at 59.8, 4.65 (s, 1H), 4.57-4.49 (m, 1H), 4.49-4.25 (m, 1H), 4.16 (s, 2H), 3.48 (q, J=6.5, 5.9 Hz, 1H), 3.44 (q, J=5.6, 5.0 Hz, 1H), 2.65-2.48 (m, 1H), 2.20-2.08 (m, 3H).13C NMR (150 MHz, CD3OD): δ 171.5, 166.7, 165.3, 145.4, 140.3, 139.9, 134.7, 133.1, 131.0, 130.4, 128.6, 128.3, 114.6, 61.3, 60.2, 56.5, 47.5, 43.9, 33.6, 30.8, 25.0. HR-MS (ESI): [M+H+] calculated 468.1522, found 468.1499. Other Compounds Additional compounds were synthesized according to similar procedures using appropriate starting materials. Compound structures and HR-MS data are shown in Table 1. TABLE 1Exemplary CompoundsHRMSHRMS(M − H+),(M − H+),NumberStructurecalculatedfound1325.1580325.15902359.1424359.14553352.0784352.07954387.1737387.17485421.1580421.15906414.0941414.09487315.1162315.11648225.1056226.10449329.1318329.131710253.1005253.100611394.1220394.121912394.1220394.122713281.1318281.131714294.1271294.127015333.1267333.126716400.0784400.078517351.0832351.083618319.1223319.122419408.1376408.137820280.1114280.111421393.0783393.078522266.1322266.132323309.1380309.138324356.1427356.143925308.1427308.143926308.1427308.143727282.1271282.127828282.1271282.127829358.1584358.158830358.1584358.158831385.1693385.170032312.1177312.118333342.1271342.128034308.1427308.143335394.1220394.122836428.0830428.083137280.1114280.111738294.1271294.127539398.1645398.165340357.1380357.138941423.1485423.149142421.1096421.109843386.1533386.154344294.1271294.127545294.1271294.127546357.1380357.138947391.0990391.100048373.1329373.133449356.1427356.143950372.1489372.149851308.1427308.143452386.1533386.154053310.1220310.122654310.1220310.122055322.1584322.159256424.0892424.089857338.1533338.153958320.1427320.143259441.0549441.055360348.1376348.138561441.1391441.139962412.1148412.115663371.1536371.154364378.1094378.110765306.1271306.127966372.1376372.138367374.1333374.133968447.0711447.071469439.1257439.126070400.1689400.169571396.1489396.149372388.1490388.149873473.0867473.087174308.1427308.143475308.1427308.143776371.1536371.154377371.1536371.154578342.1271342.127579389.1100389.110980403.1257403.126481424.1802424.181182483.1883483.189283368.1427368.142884364.0896364.090385363.0944363.095286357.1380357.138987457.1096457.110088413.1100413.110689447.0711447.071590439.1257439.125991455.1303455.130592439.1257439.125993438.1304438.131194469.1363469.137095469.1363469.137296440.1209440.121497473.0867473.087198471.1519471.152099397.1693397.1700100377.1100377.1090101412.1148412.1154102350.1533350.1538103427.1257427.1269104467.157467.158105454.1366454.137106454.1366454.1374107454.1366454.1369108446.0774446.0780109478.1366478.1376110429.1162429.1176111429.1162429.1173112444.1522444.1523113494.1679494.1698114468.1522468.1532115419.1206419.1219116547.1944547.1956117479.1318479.1342118494.1679494.1681119494.1679494.1679120412.1689412.1691121403.1257403.1267122397.1693397.1698123510.1628510.1628124480.1522480.1533125468.1522468.1499126538.1941538.1950127508.1835508.1839128522.1992522.1993129551.2257551.2265130572.1785572.1786131564.2098564.2105132522.1992522.1994133551.2257551.2263134238.1260238.1278135519.1631136508.1835137494.1679138494.1679139490.1366140593.1458141495.1268142453.1413143507.1131144552.2098145508.1835146522.1628147495.1268148539.1894539.1901149551.1894551.1905150535.1581151565.2050152565.2050153551.1894154498.1740155598.2053156453.1413157467.1570158454.1366159455.1209160425.1100161439.1257162578.2254163592.2411164595.2156165609.2312166596.2108167610.2265168610.2265169624.2421170586.1941171600.2098172592.1505173606.1662174565.2050175562.1690176611.1894177611.1894178606.1395179606.1395180606.1395181602.1890182602.1890183602.1890184581.1999185594.2316186587.1894187601.2050188565.2050189586.1941190572.1785191564.2098192578.2254193581.1999194595.2156195572.1785196586.1941197579.1301198593.1458199551.1894200565.1145201567.1843202573.1737203587.1894204536.6885536.1793205544.1238544.1243206544.1238544.1246207536.1785536.1789208310.1220310.1225209546.1298234519.1631519.1639235524.1785524.1794236565.2050565.2056237560.1785560.1788238580.2411580.2417239465.1413465.1416240602.2254602.2258241510.1628510.1633242579.2207579.2212243607.2520607.2529244536.1785536.1789245593.2363593.2370246579.2207579.2211247605.2363248565.2050249635.2833251565.2050 Example 2 Compound Syntheses—Dimers Compound 223. (2S,2′S,4R,4′R)-4,4′-(hexane-1,6-diylbis(oxy))bis(N-(5-(3-(thiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide) Step 1. 1-di-tert-butyl 2-dimethyl 4,4′-(hexane-1,6-diylbis(oxy))(2S,2′S,4R,4′R)-bis(pyrrolidine-1,2-dicarboxylate) To a suspension of NaH (60% dispersion in mineral oil, 400 mg, 0.01 mol) in dry DMF (10 mL) at 0° C. was added solution of 1-(tert-butyl) 2-methyl (2S,4R)-4-hydroxypyrrolidine-1,2-dicarboxylate (2.45 g, 0.01 mol) dropwise. The mixture was stirred for 30 min at 0° C. before adding 1,6-diiodohexane (1.54 g, 750 μL, 4.55 mmol). The temperature was allowed to increase slowly up to rt and the reaction mixture was stirred during 16 h before treatment with saturated aqueous NH4Cl. The aqueous layer was extracted with ethyl acetate and the combined organic layers were washed with brine, dried over MgSO4and concentrated under reduced pressure. The crude mixture was purified by silica gel chromatography (gradient of EtOAc in hexanes from 10 to 100%) providing the targeted 1-di-tert-butyl 2-dimethyl 4,4′-(hexane-1,6-diylbis(oxy))(2S,2′S,4R,4′R)-bis(pyrrolidine-1,2-dicarboxylate) as a clear oil. Yield 521 mg (20%).1H NMR (600 MHz, Methanol-d4, mixture of rotamers) δ 4.40-4.25 (m, 2H), 4.14-4.01 (m, 2H), 3.78-3.67 (m, 6H), 3.62-3.34 (m, 8H), 2.41-2.20 (m, 2H), 2.07-1.99 (m, 2H), 1.60-1.53 (m, 2H), 1.50-1.40 (m, 18H), 1.40-1.27 (m, 6H).13C NMR (125 MHz, Methanol-d4, mixture of rotamers) δ 175.2, 174.9, 174.3, 174.1, 172.9, 156.3, 156.2, 155.9, 155.7, 81.7, 81.6, 81.5, 78.7, 78.5, 77.9, 77.8, 70.1, 70.0, 70.0, 69.9, 69.8, 61.5, 59.5, 59.2, 59.1, 58.8, 53.4, 53.2, 52.8, 52.7, 52.7, 52.7, 52.6, 52.6, 52.6, 37.4, 37.4, 36.9, 36.6, 36.6, 36.0, 32.7, 30.9, 30.8, 30.8, 30.8, 30.7, 30.1, 28.7, 28.7, 28.6, 28.6, 27.1, 27.0, 26.9, 26.9, 23.7, 20.9, 14.5, 14.4; HR-ESI-MS: C28H49N2O10[M+H]+m/z calculated 573.3382, found 573.3356. Step 2. (2′S,4R,4′R)-4,4′-(hexane-1,6-diylbis(oxy))bis(1-(tert-butoxycarbonyl)-L-proline) Synthesized according to general procedure B from 1-di-tert-butyl 2-dimethyl 4,4′-(hexane-1,6-diylbis(oxy))(2S,2′S,4R,4′R)-bis(pyrrolidine-1,2-dicarboxylate). Clear oil. Yield 485 mg (98%).1H NMR (600 MHz, Methanol-d4, mixture of rotamers) δ=4.37-4.20 (m, 2H), 4.17-4.02 (m, 2H), 3.65-3.34 (m, 8H), 2.42-2.18 (m, 2H), 2.11-2.01 (m, 2H), 1.59-1.50 (m, 4H), 1.49-1.41 (m, 18H), 1.40-1.30 (m, 4H).13C NMR (125 MHz, Methanol-d4, mixture of rotamers) δ=176.6, 176.2, 175.7, 175.6, 175.4, 175.4, 156.4, 156.3, 156.0, 155.9, 81.8, 81.8, 81.5, 81.4, 78.7, 78.5, 77.9, 77.7, 77.7, 70.1, 70.1, 70.1, 70.0, 70.0, 70.0, 69.9, 59.5, 59.1, 58.9, 58.7, 53.5, 53.2, 53.2, 52.7, 52.7, 37.5, 37.5, 36.8, 36.8, 36.8, 36.7, 35.9, 30.8, 30.8, 30.7, 30.7, 29.9, 28.8, 28.7, 28.6, 28.6, 27.0, 27.0, 26.9, 26.9, 24.2. HR-ESI-MS: C26H45N2O10[M+H]+m/z calculated 545.3069, found 545.3075. Step 3. 5,5′-(((2S,2′S,4R,4′R)-4,4′-(hexane-1,6-diylbis(oxy))bis(1-(tert-butoxycarbonyl)pyrrolidine-4,2-diyl-2-carbonyl))bis(azanediyl))bis(thiophene-2-carboxylic Acid) Synthesized according to general procedure A, from (2′S,4R,4′R)-4,4′-(hexane-1,6-diylbis(oxy))bis(1-(tert-butoxycarbonyl)-L-proline) and methyl 5-amino-2-thiophenecarboxylate. Obtained crude was directly submitted to hydrolysis (general procedure B) and then purified by silica gel column chromatography (0-100% EtOAc in DCM) providing titled compound as off white solid. Yield 87 mg (61% on 2 steps).1H NMR (600 MHz, Methanol-d4, mixture of rotamers) δ 7.60-7.54 (m, 2H), 6.76-6.70 (m, 2H), 4.49-4.33 (m, 2H), 4.17-4.00 (m, 2H), 3.65-3.55 (m, 4H), 3.52-3.41 (m, 4H), 2.50-2.35 (m, 2H), 2.14-1.97 (m, 2H), 1.64-1.54 (m, 4H), 1.50-1.42 (m, 4H), 1.40-1.31 (m, 18H).13C NMR (125 MHz, Methanol-d4, mixture of rotamers) δ 172.4, 166.7, 155.8, 147.0, 132.6, 126.8, 113.7, 113.6, 113.5, 82.0, 82.0, 82.0, 78.7, 78.1, 78.0, 70.0, 70.0, 69.9, 60.8, 60.8, 60.3, 53.3, 53.3, 38.0, 37.9, 31.0, 30.8, 30.8, 28.7, 28.6, 28.5, 27.1, 27.1, 26.9. HR-ESI-MS: C36H51N4O12S2[M+H]+m/z calculated 795.2939, found 795.2948. Step 4. (2S,2′S,4R,4′R)-4,4′-(hexane-1,6-diylbis(oxy))bis(N-(5-(3-(thiazol-2-yl)azetidine-1-carbonyl)thiophen-2-yl)pyrrolidine-2-carboxamide) (Compound 223) Synthesized according to general procedure A, from 5,5′-(((2S,2′S,4R,4′R)-4,4′-(hexane-1,6-diylbis(oxy))bis(1-(tert-butoxycarbonyl)pyrrolidine-4,2-diyl-2-carbonyl))bis(azanediyl))bis(thiophene-2-carboxylic acid) and 2-(azetidin-3-yl)thiazole dihydrochloride. Obtained residue was used in the next step without further purification. To a solution of the residue from the previous step in the DCM (2 mL) at 0° C. TFA (0.5 mL) was added dropwise, and the mixture was stirred at 0° C. for 30 min. After this all volatiles were removed under reduced pressure, the residue was triturated with 7N ammonia solution in methanol, re-concentrated and purified by silica gel column chromatography (0-100% DCM/MeOH+0.5% ammonia (v/v)) to afford the titled compound in form of free base, which was re-dissolved in 1 mL of DCM and treated with 200 uL of TFA at 0° C. After stirring for 10 min, mixture was concentrated under reduced pressure, the residue was re-dissolved in MeOH and passed through a pad with Amberlite IRA402 Cl-form, obtained solution was re-concentrated providing the titled compound in form of dihydrochloride (23 mg, 52% in 2 steps).1H NMR (600 MHz, Methanol-d4) δ 7.80 (d, J=3.3 Hz, 2H), 7.56 (d, J=3.3 Hz, 2H), 7.38 (d, J=4.2 Hz, 2H), 6.84 (d, J=4.2 Hz, 2H), 4.97 (s, 2H), 4.76-4.56 (br.s., 4H), 4.53 (dd, J=10.4, 7.4 Hz, 2H), 4.46-4.37 (m, 3H), 4.36-4.28 (m, 3H), 3.59-3.48 (m, 4H), 3.46 (s, 4H), 2.70 (dd, J=13.7, 7.4 Hz, 2H), 2.11 (ddd, J=14.2, 10.4, 4.3 Hz, 2H), 1.63 (t, J=6.9 Hz, 4H), 1.51-1.40 (m, 4H).13C NMR (151 MHz, Methanol-d4) δ 171.9, 167.5, 165.3, 145.3, 143.8, 130.2, 128.8, 121.0, 114.5, 79.4, 70.2, 60.4 (2 overlapped carbons, one is azetidine CH2, identified from HSQC spectrum), 56.7 (azetidine CH2, identified from HSQC spectrum), 52.8, 49.8, 37.0, 33.3, 30.8, 27.1. HR-ESI-MS: C38H47N8O6S4[M+H]+m/z calculated 839.2496, found 839.2498. Other Compounds Additional dimer compounds were synthesized according to similar procedures using appropriate starting materials. Compound structures and HR-MS data are shown in Table 2. TABLE 2Exemplary CompoundsHRMSHRMS(M − H+),(M − H+),NumberStructurecalc'dfound210919.2547919.2579211918.2707918.2713212932.2863932.28772131059.3860  also [M − H + H]2+/ 2530.19721059.3843  also [M − H + H]2+/ 2530.1957214693.2594693.2528215701.3150701.3159216759.2953759.2961217867.2809867.2818218939.2809939.2818219967.3122967.3128220867.2809867.2809221911.2496911.2496222859.2183859.2193223839.2496839.24982241045.33402251089.36022261133.3864227992.30742281036.33372291113.25602301113.25602311101.39662321105.35512331129.40272521003.3234  also M − H+2/ 2502.16541003.3239  also M − H+2/ 2502.1658 Example 3 Fluorescence Polarization Assay A fluorescence polarization anisotropy (FP) assay was developed, employing di-crotonylated histone H3 derived peptide conjugated with a fluorophore (FAM-H3K23crK27cr), which binds to GST-fused GAS41 YEATS domain with sub-micromolar affinity (KD=0.9 μM). 5′ 6-Fluorescein (FAM)-labeled di-crotonylated Histone H3 peptide probe H3K23crK27cr was synthesized and used for competition experiments with 1 μM GST-GAS41(1-148) incubated with a competitor (e.g. compounds of the disclosure) at 1% DMSO in assay buffer containing 50 mM TRIS pH 7.5, 150 mM sodium chloride, 1 mM TCEP, 0.01% BSA, and 0.01% Tween-20 for 1 hour. 25 nM FAM-H3K23crK27cr peptide was added and the plate was incubated for an additional hour before fluorescence polarization data was measured at 525 nM on a Pherastar plate reader (BMG Labtech). This assay was validated by testing competition with H3K27ac peptide and determined IC50=243 μM, which is consistent with relatively weak affinity of mono-acetylated peptide (Cho 2018). In this assay, the Compound 134 ((5-(tert-butyl)thiophen-2-yl)(pyrrolidin-1-yl)methanone) exhibits comparable activity to H3K27ac, with IC50=210 μM. IC50values for selected compounds of the disclosure were determined using the fluorescence polarization assay with GAS41-YEATS and FAM-H3K23crK27cr. Table 3 shows biological activities (IC50values for inhibition of GAS41 YEATS) for selected compounds from Table 1 in a fluorescence polarization assay. Compound numbers correspond to the numbers and structures provided in Table 1. TABLE 3Less than2 μM to less than10 μM to less thanGreater than2 μM10 μM50 μM50 μMGAS4196, 98, 105,73, 76, 81, 85, 86,36, 37, 38, 40, 41,1, 2, 3, 4, 5, 6, 7,YEATS106, 107,87, 88, 89, 90, 91,42, 44, 46, 47, 49,8, 9, 10, 11, 12,inhibitor109, 110,92, 93, 94, 95, 97,51, 52, 53, 54, 56,13, 14, 15, 16,IC50(μM)111, 113,99, 100, 108, 112,58, 59, 60, 61, 62,17, 18, 19, 20,116, 118,114, 117, 120, 121,63, 64, 65, 67, 68,21, 22, 23, 24,119, 123,122, 125, 127, 128,69, 70, 71, 72, 75,25, 26, 27, 28,124, 126,237, 238, 239, 24077, 78, 79, 80, 82,29, 30, 31, 32,129, 130,84, 101, 102, 103,33, 34, 35, 39,131, 132,104, 115, 20843, 45, 48, 50,133, 234,55, 57, 66, 74,235, 236,83, 134241, 242,243, 244,245, 246 Example 4 AlphaScreen Assay An AlphaScreen competition assay was also developed using His6-tagged full-length GAS41 and biotinylated-, di-crotonylated-H3 peptide (biotin-H3K23crK27cr). For full-length protein competition experiments, 100 nM MOCR-his6-Gas41 protein was incubated with 100× competitor in 50 mM HEPES pH 7.5, 100 mM NaCl, 1 mM TCEP, 0.05% BSA, 0.01% Tween-20 for 1 hour at 1% DMSO in a 96-well ½-Area AlphaPlate. H3K23crK27cr-biotin was added to a final concentration of 25 nM and incubated for 1 hour. Nickel Chelate Acceptor AlphaScreen beads were added to a final concentration of 10 μg/mL and incubated for 1 hour. Streptavidin Donor AlphaScreen beads were added to a final concentration of 10 μg/mL and incubated for 2 hours. Alpha signal was measured on a Pherastar plate reader. We found IC50=73 μM for the compound (5-(tert-butyl)thiophen-2-yl)(pyrrolidin-1-yl)methanone (Compound 134 from Table 1), and IC50=24 μM for H3K27ac. Table 4 shows biological activities (IC50values for inhibition of GAS41) for selected compounds from Table 2 in the AlphaScreen assay. Compound numbers correspond to the numbers and structures provided in Table 2. TABLE 4Less than 500500 nM to lessnMthan 10 μMGAS41210, 211, 212,214, 215, 216YEATS213, 217, 218,inhibitor219, 220, 221,IC50(μM)222, 223 Example 5 Crystal Structure The crystal structure of the complex of Compound 85 with GAS41 YEATS at 2.10 Å resolution was determined (FIG.1). Compound 85 binds in a channel that constitutes a recognition site for acetyl-lysine (Cho 2018) and is comprised of side chains of H43, H71, S73, Y74, W93, and F96 and backbone of G92, G94 and E95 (FIG.1). Example 6 Dimeric Compounds Induce Dimerization of GAS41 YEATS Domain An AlphaScreen assay based on His-tagged and biotin-labeled Avi-tagged GAS41 YEATS domain constructs was developed. For dimerization experiments, 500 nM his6-Gas41(13-158) and 250 nM avi-Gas41-YEATS were incubated in 50 mM HEPES pH 7.5, 100 mM NaCl, 1 mM TCEP, 0.05% BSA, 0.01% Tween-20 in a 96-well ½-Area AlphaPlate incubated for 30 minutes. Compounds 221 and 223 were added to a final concentration of 250 nM at 1% DMSO. Nickel Chelate Acceptor AlphaScreen beads were added to a final concentration of 10 μg/mL and incubated for 1 hour. Streptavidin Donor AlphaScreen beads were added to a final concentration of 10 μg/mL and incubated for 2 hours. For competition experiments with the dimeric complex, 500 nM his-Gas41(13-158) and 250 nM avi-Gas41-YEATS were incubated in assay buffer with 250 nM dimeric inhibitor for 30 minutes before the addition of monomeric competitor. AlphaScreen beads were added as in previous experiments. Alpha signal was measured on a Pherastar plate reader. Titration of his6-Gas41(13-158) and avi-Gas41-YEATS with either Compound 223 or 221 resulted in an increase of luminescence signal reflecting formation of the dimeric complex (FIG.2A). The signal was further decreased at highest compound concentrations, and indicates saturation of YEATS domain via independent inhibitor molecules (Hook effect). Binding of Compound 85 and Compound 223 to15N-labeled GAS41 YEATS domain were also compared by NMR; only dimeric Compound 223 but not monomeric Compound 85 induces very substantial broadening of signals, suggesting formation of a larger dimeric complex (FIGS.2B,C). Example 7 Inhibition of GAS41 Interactions in Cells A NanoBiT assay (Promega Corporation, Madison, WI) was developed to detect inhibition of protein-protein interaction in HEK293T cells by compounds. GAS41-WT and GAS41-W93A mutant were cloned into pBiT1.1-C[TK/LgBiT] vector. SmBiT-H3.3 was purchased from Promega. HEK293T cells (4×10E5) were plated into 6-well plates (DMEM with 10% FBS) and incubated for 5 h. The LgBiT-GAS41 and SmBiT-H3.3 plasmids were co-transfected using FuGENE HD for 42 h. 5×104 cells were transferred into 96-well white plates (DMEM with 10% FBS and 1% Penicillin and Streptomycin) and treated with compounds for 24 h. After the Nano-Glo Live Cell Reagent was added to each well, the luminescence was measured immediately using PHERAstar FS instrument. Co-expression of both proteins resulted in strong luciferase signal reflecting the interaction of GAS41 with acetylated H3.3 in cells. Introduction of a point mutation W93A in LgBit GAS41 abolishing histone recognition (Hsu 2018) largely diminished luminescence signal and validates the NanoBit assay. Subsequently, activity of dimeric Compound 221 was tested in the NanoBit assay and found dose-dependent inhibition of the luminescence signal and estimated IC50=6 μM (FIG.3). Importantly, treatment with Compound 221 did not reduce the signal for W93A GAS41 mutant, further supporting specific activity (FIG.3). Example 8 Activity in NSCLS Cells To investigate cellular activity of GAS41 inhibitors, H1299 cells were treated with monomeric Compound 88 and dimeric Compound 221 for 4 days. Only the dimer Compound 221 induces dose dependent growth inhibition with GI50˜3 μM (FIG.4A). To determine whether growth inhibition is dependent on presence of GAS41 we developed A549 GAS41 knocked-out cells using the CRISPR/CAS9 system. We found GAS41-KO were viable but grew more slowly compare with parental A549 cells and had reduced growth by ˜70% at day 14 (FIG.4B). Treatment with Compound 221 partially inhibited growth of A549 cells by ˜40% at 12 μM concentration, but had no effect on the GAS41-KO cells (FIG.4B), validating specific growth inhibition. Next, we evaluated the effect of Compound 221 on growth of two NSCLC cell lines H1299 and H1933 with GAS41 amplification. Treatment with Compound 221 reduced growth of both cell lines with GI50˜6 μM at day 14 (FIG.4C). Such effect correlates closely with activity of Compound 221 in NanoBit assay (FIG.3). To further validate on-target activity of Compound 221, we tested expression of GAS41 target genes in H1299 (Hsu 2018). Treatment with Compound 221 resulted in statistically significant decrease in the expression of E2F2, FOXM1, and MCM6 (FIG.4D). Altogether, dimeric inhibitor Compound 221 reduces binding of GAS41 to acetylated H3.3 in cells and induces on-target growth inhibition in NSCLCs lines.
242,493
11858926
DETAILED DESCRIPTION The compounds or pharmaceutically acceptable salts thereof as described herein, can have activity as Btk modulators. In particular, compounds or pharmaceutically acceptable salts thereof as described herein, can be Btk inhibitors. In a second embodiment of the present invention, the compound is represented by formula (I), or a pharmaceutically acceptable salt thereof, wherein Q1, Q2and Q3are each independently CH—R3and the definitions for the other variables are as defined in the first embodiment. In a third embodiment of the present invention, the compound is represented by formula (I), or a pharmaceutically acceptable salt thereof, wherein Q2is N(R2), Q1and Q3are each independently CH—R3, and the definitions for the other variables are as defined in the first embodiment. In a fourth embodiment of the present invention, the compound is represented by formula (I), or a pharmaceutically acceptable salt thereof, wherein Q3is N(R2), Q1and Q2are each independently CH—R3, and the definitions for the other variables are as defined in the first embodiment. In a fifth embodiment of the present invention, the compound is represented by formula (I), or a pharmaceutically acceptable salt thereof, wherein Q1is O, Q2and Q3are each independently CH—R3; and the definitions for the other variables are as defined in the first embodiment. In a sixth embodiment of the present invention, the compound is represented by formula (I), or a pharmaceutically acceptable salt thereof, W is CH; and the definitions for the other variables are as defined in the first, second, third, fourth or fifth embodiment. In a seventh embodiment of the present invention, the compound is represented by formula (I), or a pharmaceutically acceptable salt thereof, wherein Y is N; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth or sixth embodiment. In an eighth embodiment of the present invention, the compound of the present invention is represented by any one of the following formulas: or a pharmaceutically acceptable salt thereof; and the definitions for the variables are as defined in the first embodiment. In a ninth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein ring A is selected from 1,2,3-oxadiazole, 1,3,4-oxadiazole, 1,2,4-oxadizole, 1,2,3-thiadiazole, 1,3,4-thiadiazole, 1,2,4-thiadiazole, 1,2,3-triazole, and 1,2,4-triazole, each of which is optionally substituted with one or two R1; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh or eighth embodiment. In a tenth embodiment of the present invention, the compounds is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein ring A is represented by one of the following formula: and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh or eighth embodiment. In a eleventh embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein:R1in each occurrence is independently C1-6alkyl or C3-5cycloalkyl; wherein said C1-6alkyl and C3-5cycloalkyl are optionally substituted with one to three R10;R10in each occurrence is independently selected from halo, —CN and C1-6alkyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth embodiment. In a twelfth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein:R1in each occurrence is independently C1-4alkyl, cyclopropyl, or cyclobutyl; wherein said C1-4alkyl, cyclopropyl and cyclobutyl are optionally substituted with one to three R10;R10in each occurrence is independently selected from halo, —CN and C1-3alkyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth embodiment. In a thirteenth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein:R1in each occurrence is independently selected from —C(CH3)3, —CH(CH3)2, —C(CH3)2CHF2, —C(CH3)2CF3, —C(CH3)2CH2F, —C(CH3)2CN, 1-methylcyclopropyl, cyclobutyl, and 3,3-difluorocyclobutyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth embodiment. In a fourteenth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R1is —C(CH3)3; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth embodiment. In a fifteenth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein:R2is selected from H, C1-6alkyl, C4-6cycloalkyl, saturated 4- to 6-membered monocyclic heterocyclyl, —C(O)R2a, —C(O)2R2a, and —S(O)2R2a, wherein said C1-6alkyl, C4-6cycloalkyl, and saturated 4- to 6-membered monocyclic heterocyclyl are optionally substituted with one to three R20;R2ain each occurrence is independently selected from H, C1-6alkyl, C4-6alkyl, and saturated 4- to 6-membered monocyclic heterocyclyl, wherein said C1-6alkyl, 4- to 6-membered monocyclic carbocyclyl, and saturated 4- to 6-membered monocyclic heterocyclyl in each occurrence are optionally and independently substituted with one or more R20;R20in each occurrence is independently selected from C1-6alkyl, C4-6cycloalkyl, saturated 4- to 6-membered monocyclic heterocyclyl, halo, —CN, —N(R20a)2, and —OR20a;R20ain each occurrence is independently H or C1-6alkyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth or fourteen embodiment. In a sixteenth embodiment, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein: R2is selected from H, C1-6alkyl, C4-6cycloalkyl selected from cyclobutyl, cyclopentyl and cyclohexyl, saturated 4- to 6-membered monocyclic heterocyclyl selected from azetidinyl, oxetanyl, pyrrolidinyl, tetrahydrofuranyl, thiolanyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, dioxolanyl, dithiolanyl, oxathiolanyl, piperidinyl, tetrahydropyranyl, thianyl, piperazinyl, morpholinyl, thiomorpholinyl, and dioxanyl, —C(O)R2a, —C(O)2R2a, and —S(O)2R2a, wherein said C1-6alkyl, C4-6cycloalkyl and saturated 4- to 6-membered monocyclic heterocyclyl are optionally substituted with one to three R20;R2ais C1-6alkyl optionally and independently substituted with one to three R20;R20in each occurrence is independently selected from C1-3alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, halo, —N(R20a)2, and —OR20a;R20ain each occurrence is independently H or C1-3alkyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen or fifteenth embodiment. In a seventeenth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein:R2is selected from H, C1-6alkyl, cyclobutyl, cyclopentyl and cyclohexyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, —C(O)R2a, —C(O)2R2a, and —S(O)2R2a, wherein said C1-6alkyl, cyclobutyl, cyclopentyl and cyclohexyl, oxetanyl, tetrahydrofuranyl, and tetrahydropyranyl are optionally substituted with one to three R20;R2ais C1-6alkyl optionally substituted with one R20;R20in each occurrence is independently selected from C1-3alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, halo, —N(R20a)2, and —OR20a;R20ain each occurrence is independently H or methyl; the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen or fifteenth embodiment. In a eighteenth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R2is selected from —H, —SO2CH3, —C(═O)OC(CH3)3, —C(═O)CH2N(CH3)2, —CH3, —CH2CH3, —CH2CH2OH, —CH2CH2OCH3, —CH2CH2OCH2CH3, —CH2CH(CH3)OH, —CH2C(CH3)2OH, —CH2CH2CH2OH, —CH2CHF2, —CH2CF3, and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen or fifteenth embodiment. In a nineteenth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R2is selected from —CH2CHF2, —CH2CF3, —CH2CH2OH, —CH2CH(CH3)OH, —CH2CH2OCH3, In a twentieth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein:R3is selected from H, C1-6alkyl, C4-6cycloalkyl, saturated 4- to 6-membered monocyclic heterocyclyl, halo, —OR3a, —OC(O)R3a, —OC(O)N(R3a)2, and —SR3a, wherein said C1-6alkyl, C4-6cycloalkyl, and saturated 4- to 6-membered monocyclic heterocyclyl are optionally substituted with one to three R30;R3ain each occurrence is independently H or C1-6alkyl, wherein said C1-6alkyl in each occurrence is optionally and independently substituted with one R30;R30in each occurrence is independently selected from C1-6alkyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, or nineteenth embodiment. In a twenty-first embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R3is selected from H, halo, and —OR3a; R3ais independently H or C1-3alkyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, or nineteenth embodiment. In a twenty-second embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R3is selected from H, —F and —OH; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, or nineteenth embodiment. In a twenty-third embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R4is H or C1-3alkyl optionally substituted with one to three fluoro; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, nineteenth, twentieth, twenty-first or twenty-second embodiment. In a twenty-fourth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R4is H or methyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, nineteenth, twentieth, twenty-first or twenty-second embodiment. In a twenty-fifth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R5is H or C1-3alkyl optionally substituted with one to three fluoro; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, nineteenth, twentieth, twenty-first, twenty-second, twenty-third or twenty-fourth embodiment. In a twenty-sixth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R5is H, methyl, ethyl or isopropyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, nineteenth, twentieth, twenty-first, twenty-second, twenty-third or twenty-fourth embodiment. In a twenty-seventh embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R6is H or C1-3alkyl optionally substituted with one to three fluoro; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, nineteenth, twentieth, twenty-first, twenty-second, twenty-third, twenty-fourth, twenty-fifth or twenty-sixth embodiment. In a twenty-eighth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R6is H, methyl or triflouromethyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, nineteenth, twentieth, twenty-first, twenty-second, twenty-third, twenty-fourth, twenty-fifth or twenty-sixth embodiment. In a twenty-ninth embodiment of the present invention, the compound is represented by formula (I), (II), (III), (IV), (V), (II′), (III′), (IV′) or (V′), or a pharmaceutically acceptable salt thereof, wherein R5and R6together with the atoms to which they are attached, form a 5- to 6-membered saturated heterocyclic ring containing one or two heteroatoms selected from O, N, and S, wherein the ring is optionally substituted with one R50; R50is a C1-3alkyl; and the definitions for the other variables are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteen, fifteenth, seventeenth, eighteenth, nineteenth, twentieth, twenty-first, twenty-second, twenty-third, or twenty-fourth embodiment. In a more specific embodiment, the 5- to 6-membered saturated heterocyclic ring heterocyclic ring is pyrrolindine, piperazine or N-methylpiperazine. In a thirtieth embodiment of the present invention, the compound is represented by the following formula: or a pharmaceutically acceptable salt thereof, wherein:ring A is represented by one of the following formula: R1is C1-6alkyl;R2is C1-6alkyl, oxetanyl, tetrahydrofuranyl, or tetrahydropyranyl, wherein said C1-6alkyl is optionally substituted with one to three R20;R20for each occurrence is independently halo or —OR20a;R20aH or C1-3alkyl;R3is H; andR5is H or C1-3alkyl. In a thirty-first embodiment of the present invention, the compound is represented by formula (IIA), (IVA), (VA), (IIA′), (IVA′), or (VA′), or a pharmaceutically acceptable salt thereof, wherein R1is tert-butyl; and the definitions for the other variables are as defined in the thirtieth embodiment. In a thirty-second embodiment of the present invention, the compound is represented by formula (IIA) or (IIA′), or a pharmaceutically acceptable salt thereof, wherein R2is —CH2CHF2, —CH2CF3, —CH2CH2OH, —CH2CH2OCH3, or —CH2C(CH3)OH; and the definitions for the other variables are as defined in the thirtieth or thirty-first embodiment. In a thirty-third embodiment of the present invention, the compound is represented by formula (IVA) to (IVA′), or a pharmaceutically acceptable salt thereof, wherein R3is H; and the definitions for the other variables are as defined in the thirtieth, or thirty-first embodiment. In a thirty-fourth embodiment of the present invention, the compound is represented by formula (IIA), (IVA), (VA), (IIA′), (IVA′), or (VA′), or a pharmaceutically acceptable salt thereof, wherein R5is methyl or isopropyl; and the definitions for the other variables are as defined in the thirtieth, thirty-first, thirty-second or thirty-third embodiment. In a thirty-fifth embodiment of the present invention, the compound of the present invention is selected from:5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide, 5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide, 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide(R)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide(R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide5-(tert-butyl)-N—((S)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide5-(tert-butyl)-N—((S)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide(S)-1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide, (S)-1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-((5R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(2-(2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N—((R)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N—((R)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N—((S)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N—((S)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(2-(2-methoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(2-(2-methoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-(2-methoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(2-(2,2-difluoroethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(2,2-difluoroethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(2,2-difluoroethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide(R)—N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide,(S)—N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,3-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-5-carboxamide,(R)-3-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-5-carboxamide,(S)-3-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-5-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,2,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,2,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,2,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide,1-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(3-(2-hydroxyethyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(3-(2-hydroxyethyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(3-(2-hydroxyethyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide5-(tert-butyl)-N—((R)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide5-(tert-butyl)-N—((S)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide5-(tert-butyl)-N—((S)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide5-(tert-butyl)-N-(3-(2-hydroxy-2-methylpropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(3-(2-hydroxy-2-methylpropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(3-(2-hydroxy-2-methylpropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,1-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(3-methyl-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(3-methyl-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(3-methyl-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(3-(2-hydroxyethyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(3-(2-hydroxyethyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(3-(2-hydroxyethyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide 5-(tert-butyl)-N-(2-(2-(dimethylamino)acetyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-(dimethylamino)acetyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-(dimethylamino)acetyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1,1,1-trifluoro-2-methylpropan-2-yl)-1,3,4-oxadiazole-2-carboxamide,(R)—N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1,1,1-trifluoro-2-methylpropan-2-yl)-1,3,4-oxadiazole-2-carboxamide,(S)—N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1,1,1-trifluoro-2-methylpropan-2-yl)-1,3,4-oxadiazole-2-carboxamide,N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1,1,1-trifluoro-2-methylpropan-2-yl)-1,3,4-oxadiazole-2-carboxamide,(R)—N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1,1,1-trifluoro-2-methylpropan-2-yl)-1,3,4-oxadiazole-2-carboxamide,(S)—N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1,1,1-trifluoro-2-methylpropan-2-yl)-1,3,4-oxadiazole-2-carboxamide,5-(1,1-difluoro-2-methylpropan-2-yl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(1,1-difluoro-2-methylpropan-2-yl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(1,1-difluoro-2-methylpropan-2-yl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(1,1-difluoro-2-methylpropan-2-yl)-N-(2-(2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(1,1-difluoro-2-methylpropan-2-yl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(1,1-difluoro-2-methylpropan-2-yl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(1,1-difluoro-2-methylpropan-2-yl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(1,1-difluoro-2-methylpropan-2-yl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(1,1-difluoro-2-methylpropan-2-yl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(1-fluoro-2-methylpropan-2-yl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(1-fluoro-2-methylpropan-2-yl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide(S)-5-(1-fluoro-2-methylpropan-2-yl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(1-fluoro-2-methylpropan-2-yl)-N-(2-(2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(1-fluoro-2-methylpropan-2-yl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(1-fluoro-2-methylpropan-2-yl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(1-fluoro-2-methylpropan-2-yl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(1-fluoro-2-methylpropan-2-yl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(1-fluoro-2-methylpropan-2-yl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-cyclobutyl-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-cyclobutyl-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-cyclobutyl-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(2-cyanopropan-2-yl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(2-cyanopropan-2-yl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(2-cyanopropan-2-yl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(2-cyanopropan-2-yl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(2-cyanopropan-2-yl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(2-cyanopropan-2-yl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,1-isopropyl-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-isopropyl-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-isopropyl-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-cyclobutyl-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-cyclobutyl-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-cyclobutyl-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-thiadiazole-2-carboxamide(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-thiadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-thiadiazole-2-carboxamide,3-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-5-carboxamide,(R)-3-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-5-carboxamide,(S)-3-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-5-carboxamide,1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(2-ethyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(2-ethyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-ethyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide(R)-1-(tert-butyl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(2-(2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide5-(tert-butyl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(2-(2-methoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-methoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-methoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-ethyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-ethyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-ethyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(2-hydroxy-2-methylpropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-tert-butyl-1,3,4-oxadiazole-2-carboxylic acid {(R)-2-(2-hydroxy-2-methyl-propyl)-8-[2-(1-methyl-1H-pyrazol-4-ylamino)-pyrimidin-4-yl]-2,3,4,5-tetrahydro-1H-2-benzazepin-5-yl}-amide,(S)-5-(tert-butyl)-N-(2-(2-hydroxy-2-methylpropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(3-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(3-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(3-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamideN-(8-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide,(R)—N-(8-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide,(S)—N-(8-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,1-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide,(R)—N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide,(S)—N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide, (R)-5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,1-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide, (S)-5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,1-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide, (S)-1-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,1-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(2-(cyclopropylmethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(cyclopropylmethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(cyclopropylmethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1,3-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,N-(2-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamide,(R)—N-(2-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamide,(S)—N-(2-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(2-(2-((5-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((5-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((5-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,N-(2-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide,(R)—N-(2-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide,(S)—N-(2-(2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide,1-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-4-methyl-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-4H-1,2,4-triazole-3-carboxamide,(R)-5-(tert-butyl)-4-methyl-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-4H-1,2,4-triazole-3-carboxamide,(S)-5-(tert-butyl)-4-methyl-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-4H-1,2,4-triazole-3-carboxamide,2-isopropyl-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-2H-1,2,3-triazole-4-carboxamide,(R)-2-isopropyl-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-2H-1,2,3-triazole-4-carboxamide,(S)-2-isopropyl-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-2H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-((8S)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-((8R)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-((5R,8S)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-((5S,8R)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-((5R,8R)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-((5S,8S)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,N-(8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide,N-((8S)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide,N-((8R)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamideN-((5R,8S)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamideN-((5S,8R)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamideN-((5S,8S)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamideN-((5S,8R)-8-fluoro-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamideN-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide,(R)—N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide,(S)—N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-cyclobutyl-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-cyclobutyl-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-cyclobutyl-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(2-ethoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-ethoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-ethoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,1-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1H-1,2,3-triazole-4-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-thiadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-thiadiazole-2-carboxamide,5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-thiadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-thiadiazole-2-carboxamide,5-(tert-butyl)-N—((S)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-thiadiazole-2-carboxamide,5-(tert-butyl)-N-(3-methyl-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(3-methyl-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(3-methyl-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-thiadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-thiadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-thiadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,tert-butyl 1-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,2,4,5-tetrahydro-3H-benzo[d]azepine-3-carboxylate,(R)-tert-butyl 1-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,2,4,5-tetrahydro-3H-benzo[d]azepine-3-carboxylate,(S)-tert-butyl 1-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,2,4,5-tetrahydro-3H-benzo[d]azepine-3-carboxylate,5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(3-(2-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(3-((S)-2-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(3-((S)-2-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(3-((S)-2-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(3-((R)-2-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(3-((R)-2-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(3-((R)-2-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(3-(3-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(3-(3-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(3-(3-hydroxypropyl)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,3,4-oxadiazole-2-carboxamide,5-(3,3-difluorocyclobutyl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(3,3-difluorocyclobutyl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(3,3-difluorocyclobutyl)-N-(2-methyl-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(3,3-difluorocyclobutyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(3,3-difluorocyclobutyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(3,3-difluorocyclobutyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(8-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(8-hydroxy-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,(R)-5-(tert-butyl)-N-(8-hydroxy-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,(S)-5-(tert-butyl)-N-(8-hydroxy-2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide,5-(tert-butyl)-N-(2-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(2-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,5-(tert-butyl)-N-(2-(2-((1-methyl-5-(trifluoromethyl)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(R)-5-(tert-butyl)-N-(2-(2-((1-methyl-5-(trifluoromethyl)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,(S)-5-(tert-butyl)-N-(2-(2-((1-methyl-5-(trifluoromethyl)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide,1-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1H-1,2,3-triazole-4-carboxamide,(R)-1-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1H-1,2,3-triazole-4-carboxamide,(S)-1-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1H-1,2,3-triazole-4-carboxamide,N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide,(R)—N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide, and(S)—N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-5-(1-methylcyclopropyl)-1,3,4-oxadiazole-2-carboxamide, or a pharmaceutically acceptable salt thereof. The present invention also provides crystalline forms of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (compound 27): As used herein, the term “crystalline” refers to a solid form having a crystal structure wherein the individual molecules have a highly homogeneous regular locked-in chemical configuration. Form A In one embodiment, the present invention provides crystalline Form A of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide. In one aspect, crystalline Form A is characterized by at least three, at least four, or at least five powder X-ray diffraction (PXRD) peaks at 2θ angles selected from 5.7°, 7.9°, 9.7°, 18.2°, 19.0° and 22.4°. In one embodiment, crystalline Form A is characterized by powder X-ray diffraction peaks at 2θ angles selected from 5.7°, 7.9°, 9.7°, 18.2°, 19.0° and 22.4°. In some embodiments, the peaks described above for crystalline Form A have a relative intensity of at least 5%, at least 10%, or at least 15%. In another embodiment, crystalline Form A is characterized by at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least sixteen, at least seventeen, or at least nineteen PXRD peaks at 2θ angles selected from 4.3° 5.7° 7.9° 8.7°, 97° 11.9°, 13.1°, 14.8°, 15.2°, 16.1°, 17.0°, 17.8°, 18.2°, 19.0°, 20.5°, 21.2°, 22.4°, 22.8°, 23.8°, and 25.60. As used herein, the term “relative intensity” refers to a ratio of the peak intensity for the peak of interest versus the peak intensity for the largest peak. In another aspect, crystalline Form A has a PXRD pattern that is substantially the same as PXRD pattern shown inFIG.1. In one aspect, crystalline Form A has a differential scanning calorimetry (DSC) profile that is substantially the same as the DSC profile shown inFIG.2. In particular, crystalline Form A is characterized by an onset temperature at 175.6° C.±2° C. in the DSC profile. In one embodiment, crystalline Form A has a melting temperature of 186° C.±2° C. In one aspect, crystalline Form A has a TGA profile that is substantially the same as the TGA profile shown inFIG.2. In particular, the TGA profile indicates that crystalline Form A is a hydrate. As used herein, “hydrate” refers to refers to a crystalline solid adduct containing (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide and either stoichiometric or nonstoichiometric amounts of a water incorporated within the crystal structure. Techniques known in the art to determine the to determine the amount of water present include, for example, TGA and Karl Fisher (KF) analysis. In another aspect, crystalline Form A has a solid state13C NMR spectrum that is substantially the same as that shown inFIG.3B. In one embodiment, crystalline Form A is characterized by chemical shifts at 143.7 ppm and/or 134.4 ppm in solid state13C NMR spectrum. The spectrum of Form A exhibit broader signals without showing clear duplicated signals. The Form A spectrum also suggests that there may be two independent molecules with different geometry. In another embodiments, crystalline Form A is characterized by chemical shifts in solid state13C NMR spectrum as shown in Table 3. In some embodiments, crystalline Form A is characterized by, for example, PXRD, DSC, TGA or13NMR described above or any combination thereof. In one embodiment, crystalline Form A is characterized by PXRD alone or PXRD in combination with one or more of DSC, TGA and13NMR described above. In some embodiments, crystalline Form A is at least 70%, 80%, 85%, 90%, 95%, 97%, 99%, 99.5% or 99.9% pure. The purity of Form A is determined by dividing the weight of crystalline Form A in a composition comprising compound (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide over the total weight of the compound in the composition. In one embodiment, the present invention provides a composition comprising compound (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide, wherein at least 70%, 80%, 85%, 90%, 95%, 97%, 99%, 99.5% or 99.9% by weight of the compound in the composition is crystalline Form A of the compound. In one embodiment, the present invention provides a method for preparing crystalline Form A of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide. Such method includes, e.g., forming crystalline Form A from a slurry comprising (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide and ethanol (EtOH). In one embodiment, the method comprises stirring the slurry containing (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide and EtOH at room temperature for 1 hour to 1 week, e.g., 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 10 hours, 15 hours, 24 hours, or 48 hours. Form G In one embodiment, the present invention provides crystalline Form G of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide. In one aspect, crystalline Form G is characterized by at least three, at least four or at least five PXRD peaks at 2θ angles selected from 3.6°, 8.9°, 10.9°, 12.6°, 20.2° and 21.8°. In one embodiment, crystalline Form G is characterized by PXRD peaks at 2θ angles selected from 3.6°, 8.9°, 10.9°, 12.6°, 20.2° and 21.8°. In some embodiments, the peaks described above for crystalline Form G have a relative intensity of at least 5%, at least 10%, or at least 15%. In another embodiment, crystalline Form G is characterized by at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, or at least thirteen PXRD peaks at 2θ angles selected from 3.6°, 8.9°, 11.0°, 12.6°, 14.5°, 15.4°, 16.3°, 18.4°, 20.2°, 21.8°, 23.4°, 25.4°, 26.8°, and 34.2°. In another embodiment, crystalline Form A is characterized by PXRD peaks at 2θ angles selected from 3.6°, 8.9°, 11.0°, 12.6°, 14.5°, 15.4°, 16.3°, 18.4°, 20.2°, 21.8°, 23.4°, 25.4°, 26.8°, and 34.2°. In another aspect, crystalline Form G has a PXRD pattern that is substantially the same as PXRD pattern shown inFIG.4. In one aspect, crystalline Form G has a DSC profile that is substantially the same as the DSC profile shown inFIG.5. In particular, crystalline Form G is characterized by an onset temperature at 215.4° C.±2° C. in the DSC profile. In another embodiment, crystalline Form G has a melting temperature of 217° C.±2° C. In one aspect, crystalline Form G has a TGA profile that is substantially the same as the TGA profile shown inFIG.5. In particular, the TGA profile indicates that crystalline Form G is an anhydrate. “Anhydrate” as used herein, means that the crystalline form comprises substantially no water in the crystal lattice e.g., less than 1% by weight as determined by, for example, TGA analysis or other quantitative analysis. In another aspect, crystalline Form G has a solid state13C NMR spectrum that is substantially the same as that shown inFIG.6B. In one embodiment, crystalline Form G is characterized by chemical shifts at 147.0 ppm, 146.0 ppm and/or 140.6 ppm in solid state13C NMR spectrum. The spectrum of Form G shows peak splitting (duplicate signals) in aromatic regions when compared to the solution13C NMR spectrum suggesting there are two independent molecules in the asymmetric unit. In another embodiment, crystalline Form G is characterized by chemical shifts in solid state13C NMR spectrum as shown in Table 3. In some embodiments, crystalline Form G is at least 70%, 80%, 85%, 90%, 95%, 97%, 99%, 99.5% or 99.9% pure. The purity of Form G is determined by dividing the weight of crystalline Form G in a composition comprising compound (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide over the total weight of the compound in the composition. In one embodiment, the present invention provides a composition comprising compound (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide, wherein at least 70%, 80%, 85%, 90%, 95%, 97%, 99%, 99.5% or 99.9% by weight of the compound in the composition is crystalline Form G of the compound. In one embodiment, the present invention provides a method for preparing crystalline Form G of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide. Such method includes, e.g., forming crystalline Form G from a slurry comprising (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide and isopropyl acetate (IPAc). In one embodiment, the method comprises stirring the slurry containing (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide and isopropyl acetate (IPAc) at an elevated temperature (e.g., between 30° C. and 70° C., between 40° C. and 60° C., between 45° C. and 55° C., or at 50° C.), for 1 hour to 1 week, e.g., 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 10 hours, 15 hours, 24 hours, or 48 hours. Alternatively, crystalline Form G can be prepared by a method comprising the steps of (i) removing at least a portion of dichloromethane by distillation from a mixture containing (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide and dichloromethane; (ii) adding isopropyl acetate (IPAc) to the mixture; (iii) heating the mixture containing IPAc to an elevated temperature (e.g., between 50° C. and 70° C., between 55° C. and 65° C. or 60° C.) followed by cooling to near room temperature (e.g., 20° C.) to form a slurry containing the compound and IPAc; and (iv) isolating crystalline Form G from the slurry. In one embodiment, steps (i) and (ii) can be repeated for one or more times (e.g. two, three, four, or five times). In one embodiment, steps (i) and (ii) are repeated until substantially all (e.g., at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% by volume) of dichloromethane is removed. In one embodiment, the heating and the cooling in step (iii) can be repeated for one or more times (e.g. two, three, four, five, ten, fifteen, twenty, or more times). It will be understood that the 20 values of the PXRD pattern for crystalline Form A or crystalline Form G may vary slightly from one instrument to another and may depend on variations in sample preparation. Therefore, the PXRD peak positions for crystalline Form A or crystalline Form G are not to be construed as absolute and can vary ±0.2°. As intended herein, “substantially the same PXRD pattern as shown inFIG.1”, “substantially the same PXRD pattern as shown inFIG.4”, “substantially the same as that shown inFIG.3B” or “substantially the same as that shown inFIG.6B” mean that for comparison purposes, at least 80%, at least 90%, or at least 95% of the peaks shown inFIG.1,FIG.4,FIG.3BandFIG.6Bare present. It is to be further understood that for comparison purposes some variability in peak position from those shown inFIG.1andFIG.4are allowed, such as ±0.2°. Similarly, for comparison purposes some variability in peak position from those shown inFIG.3BandFIG.6Bare allowed, such as ±0.5 ppm. As used herein, the term “alkyl” refers to a fully saturated branched or unbranched hydrocarbon moiety. Preferably the alkyl comprises 1 to 6 carbon atoms, or 1 to 4 carbon atoms. In some embodiments, an alkyl comprises from 6 to 20 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, or n-hexyl. “Alkenyl” refers to an unsaturated hydrocarbon group which may be linear or branched and has at least one carbon-carbon double bond. Alkenyl groups with 2-6 carbon atoms can be preferred. The alkenyl group may contain 1, 2 or 3 carbon-carbon double bonds, or more. Examples of alkenyl groups include ethenyl, n-propenyl, iso-propenyl, n-but-2-enyl, n-hex-3-enyl and the like. “Alkynyl” refers to an unsaturated hydrocarbon group which may be linear or branched and has at least one carbon-carbon triple bond. Alkynyl groups with 2-6 carbon atoms can be preferred. The alkynyl group may contain 1, 2 or 3 carbon-carbon triple bonds, or more. Examples of alkynyl groups include ethynyl, n-propynyl, n-but-2-ynyl, n-hex-3-ynyl and the like. The number of carbon atoms in a group is specified herein by the prefix “Cx-xx”, wherein x and xx are integers. For example, “C1-4alkyl” is an alkyl group which has from 1 to 4 carbon atoms. “Halogen” or “halo” may be fluoro, chloro, bromo or iodo. As used herein, the term “heterocyclyl” refers to a saturated or unsaturated, monocyclic or bicyclic (e.g., fused, bridged or spiro ring systems) ring system which has from 3- to 10-ring members, or in particular 3- to 8-ring members, 3- to 7-ring members, 3- to 6-ring members or 5- to 7-ring members or 4- to 7-ring members, at least one of which is a heteroatom, and up to 4 (e.g., 1, 2, 3, or 4) of which may be heteroatoms, wherein the heteroatoms are independently selected from O, S and N, and wherein C can be oxidized (e.g., C(O)), N can be oxidized (e.g., N(O)) or quaternized, and S can be optionally oxidized to sulfoxide and sulfone. Unsaturated heterocyclic rings include heteroaryl rings. As used herein, the term “heteroaryl” refers to an aromatic 5- or 6-membered monocyclic ring system, having 1 to 4 heteroatoms independently selected from O, S and N, and wherein N can be oxidized (e.g., N(O)) or quaternized, and S can be optionally oxidized to sulfoxide and sulfone. In one embodiment, a heterocyclyl is a 3- to 7-membered saturated monocyclic or a 3- to 6-membered saturated monocyclic or a 5- to 7-membered saturated monocyclic ring or a 4- to 6-membered saturated monocyclic ring. In one embodiment, a heterocyclyl is a 3- to 7-membered monocyclic or a 3- to 6-membered monocyclic or a 4- to 6-membered monocyclic ring or a 5- to 7-membered monocyclic ring. In another embodiment, a heterocyclyl is a 6 or -7-membered bicyclic ring. In yet another embodiment, a heterocyclyl is a 4- to 7-membered monocyclic non-aromatic ring. In another embodiment, a heterocyclyl is 6- to 8-membered spiro or bridged bicyclic ring. The heterocyclyl group can be attached at a heteroatom or a carbon atom. Examples of heterocyclyls include, but are not limited to, aziridinyl, oxiranyl, thiiranyl, oxaziridinyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, tetrahydrofuranyl, thiolanyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, dioxolanyl, dithiolanyl, oxathiolanyl, piperidinyl, tetrahydropyranyl, thianyl, piperazinyl, morpholinyl, thiomorpholinyl, dioxanyl, dithianyl, trioxanyl, trithianyl, azepanyl, oxepanyl, thiepanyl, dihydrofuranyl, imidazolinyl, dihydropyranyl, and heteroaryl rings including azetyl, thietyl, pyrrolyl, furanyl, thiophenyl (or thienyl), imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furazanyl, oxadiazolyl, thiadiazolyl, dithiazolyl, triazolyl, tetrazolyl, pyridinyl, pyranyl, thiopyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazinyl, thiazinyl, dioxinyl, dithiinyl, oxathianyl, triazinyl, tetrazinyl, azepinyl, oxepinyl, thiepinyl, diazepinyl, and thiazepinyl and the like. The term “fused ring system”, as used herein, is a ring system that has two rings each of which are independently selected from a carbocyclyl or a heterocyclyl, wherein the two ring structures share two adjacent ring atoms. A fused ring system may have from 9 to 12 ring members. The term “bridged ring system”, as used herein, is a ring system that has a carbocyclyl or heterocyclyl ring wherein two non-adjacent atoms of the ring are connected (bridged) by one or more (preferably from one to three) atoms selected from C, N, O, or S. A bridged ring system may have from 6 to 8 ring members. The term “spiro ring system,” as used herein, is a ring system that has two rings each of which are independently selected from a carbocyclyl or a heterocyclyl, wherein the two ring structures having one ring atom in common. Spiro ring systems have from 5 to 8 ring members. In one embodiment, a heterocyclyl is a 4- to 6-membered monocyclic heterocyclyl. Examples of 4- to 6-membered monocyclic heterocyclic ring systems include, but are not limited to azetidinyl, pyrrolidinyl, tetrahydrofuranyl, thiolanyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, dioxolanyl, dithiolanyl, oxathiolanyl, piperidinyl, tetrahydropyranyl, thianyl, piperazinyl, morpholinyl, thiomorpholinyl, dioxanyl, dithianyl, dihydrofuranyl, imidazolinyl, dihydropyranyl, pyrrolyl, furanyl, thiophenyl (or thienyl), imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furazanyl, oxadiazolyl, thiadiazolyl, dithiazolyl, triazolyl, tetrazolyl, pyridinyl, pyranyl, thiopyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazinyl, thiazinyl, dioxinyl, dithiinyl, oxathianyl, triazinyl, and tetrazinyl. In another embodiment, a heterocyclyl is a saturated 4- to 6-membered monocyclic heterocyclyl. Examples of saturated 4- to 6-membered monocyclic heterocyclic ring systems include, but are not limited to azetidinyl, pyrrolidinyl, tetrahydrofuranyl, thiolanyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, dioxolanyl, dithiolanyl, oxathiolanyl, piperidinyl, tetrahydropyranyl, thianyl, piperazinyl, morpholinyl, thiomorpholinyl, dioxanyl, and dithiinyl. In one embodiment, a saturated 4- to 6-membered monocyclic heterocyclyl is azetidinyl, oxetanyl, pyrrolidinyl, tetrahydrofuranyl, thiolanyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, dioxolanyl, dithiolanyl, oxathiolanyl, piperidinyl, tetrahydropyranyl, thianyl, piperazinyl, morpholinyl, thiomorpholinyl, or dioxinyl. In another embodiment, a saturated 4- to 6-membered monocyclic heterocyclyl is oxetanyl, tetrahydrofuranyl, or tetrahydropyranyl. As used herein, the term “carbocyclyl” refers to saturated or unsaturated monocyclic or bicyclic hydrocarbon groups of 3-7 carbon atoms, 3-5, 3-6, 4-6, or 5-7 carbon atoms. The term “carbocyclyl” encompasses cycloalkyl groups and aromatic groups. The term “cycloalkyl” refers to completely saturated monocyclic or bicyclic or spiro hydrocarbon groups of 3-7 carbon atoms, 3-6 carbon atoms, or 5-7 carbon atoms. Exemplary monocyclic carbocyclyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopropenyl, cyclobutenyl, cyclopenentyl, cyclohexenyl, cycloheptenyl, cyclobutadienyl, cyclopentadienyl, cyclohexadienyl, cycloheptadienyl, phenyl and cycloheptatrienyl. Exemplary bicyclic carbocyclyl groups include bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, tricyclo[2.2.1.02,6]heptanyl, 6,6-dimethylbicyclo[3.1.1]heptyl, or 2,6,6-trimethylbicyclo[3.1.1]heptyl, spiro[2.2]pentanyl, and spiro[3.3]heptanyl. In one embodiment, the carbocyclyl is a 4- to 6-membered monocyclic carbocyclyl. In another embodiment, the carbocyclyl is a 3- to 5-membered carbocyclyl. In one embodiment, the carbocyclyl is a C4-6≠cycloalkyl. In yet another embodiment, the carbocyclyl is cyclobutyl, cyclopentyl or cyclohexyl. In cases where a compound provided herein is sufficiently basic or acidic to form stable nontoxic acid or base salts, preparation and administration of the compounds as pharmaceutically acceptable salts may be appropriate. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, α-ketoglutarate, or α-glycerophosphate. Inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts. Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion. Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made. Pharmaceutically-acceptable base addition salts can be prepared from inorganic and organic bases. Salts from inorganic bases, can include but are not limited to, sodium, potassium, lithium, ammonium, calcium or magnesium salts. Salts derived from organic bases can include, but are not limited to, salts of primary, secondary or tertiary amines, such as alkyl amines, dialkyl amines, trialkyl amines, substituted alkyl amines, di(substituted alkyl) amines, tri(substituted alkyl) amines, alkenyl amines, dialkenyl amines, trialkenyl amines, substituted alkenyl amines, di(substituted alkenyl) amines, tri(substituted alkenyl) amines, cycloalkyl amines, di(cycloalkyl) amines, tri(cycloalkyl) amines, substituted cycloalkyl amines, disubstituted cycloalkyl amine, trisubstituted cycloalkyl amines, cycloalkenyl amines, di(cycloalkenyl) amines, tri(cycloalkenyl) amines, substituted cycloalkenyl amines, disubstituted cycloalkenyl amine, trisubstituted cycloalkenyl amines, aryl amines, diaryl amines, triaryl amines, heteroaryl amines, diheteroaryl amines, triheteroaryl amines, heterocycloalkyl amines, diheterocycloalkyl amines, triheterocycloalkyl amines, or mixed di- and tri-amines where at least two of the substituents on the amine can be different and can be alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, or heterocycloalkyl and the like. Also included are amines where the two or three substituents, together with the amino nitrogen, form a heterocycloalkyl or heteroaryl group. Non-limiting examples of amines can include, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, trimethamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, N-alkylglucamines, theobromine, purines, piperazine, piperidine, morpholine, or N-ethylpiperidine, and the like. Other carboxylic acid derivatives can be useful, for example, carboxylic acid amides, including carboxamides, lower alkyl carboxamides, or dialkyl carboxamides, and the like. The compounds or pharmaceutically acceptable salts thereof as described herein, can contain one or more asymmetric centers in the molecule. In accordance with the present disclosure any structure that does not designate the stereochemistry is to be understood as embracing all the various stereoisomers (e.g., diastereomers and enantiomers) in pure or substantially pure form, as well as mixtures thereof (such as a racemic mixture, or an enantiomerically enriched mixture). It is well known in the art how to prepare such optically active forms (for example, resolution of the racemic form by recrystallization techniques, synthesis from optically-active starting materials, by chiral synthesis, or chromatographic separation using a chiral stationary phase). When a particular stereoisomer of a compound is depicted by name or structure, the stereochemical purity of the compounds is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97%, 99%, 99.5% or 99.9%. “Stereochemical purity” means the weight percent of the desired stereoisomer relative to the combined weight of all stereoisomers. When a particular enantiomer of a compound is depicted by name or structure, the stereochemical purity of the compounds is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97%, 99%, 99.5% or 99.9%. “Stereochemical purity” means the weight percent of the desired enantiomer relative to the combined weight of all stereoisomers. When the stereochemistry of a disclosed compound is named or depicted by structure, and the named or depicted structure encompasses more than one stereoisomer (e.g., as in a diastereomeric pair), it is to be understood that one of the encompassed stereoisomers or any mixture of the encompassed stereoisomers are included. It is to be further understood that the stereoisomeric purity of the named or depicted stereoisomers at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97%, 99%, 99.5% or 99.9%. The stereoisomeric purity the weight percent of the desired stereoisomers encompassed by the name or structure relative to the combined weight of all of the stereoisomers. When a disclosed compound is named or depicted by structure without indicating the stereochemistry, and the compound has one chiral center, it is to be understood that the name or structure encompasses one enantiomer of compound in pure or substantially pure form, as well as mixtures thereof (such as a racemic mixture of the compound and mixtures enriched in one enantiomer relative to its corresponding optical isomer). When a disclosed compound is named or depicted by structure without indicating the stereochemistry and, e.g., the compound has at least two chiral centers, it is to be understood that the name or structure encompasses one stereoisomer in pure or substantially pure form, as well as mixtures thereof (such as mixtures of stereoisomers, and mixtures of stereoisomers in which one or more stereoisomers is enriched relative to the other stereoisomer(s)). The disclosed compounds may exist in tautomeric forms and mixtures and separate individual tautomers are contemplated. In addition, some compounds may exhibit polymorphism. In one embodiment, the compounds of the invention or a pharmaceutically acceptable salt thereof include deuterium. Another embodiment is a pharmaceutical composition comprising at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier. The compounds, or pharmaceutically acceptable salts thereof described herein may be used to decrease the activity of Btk, or to otherwise affect the properties and/or behavior of Btk, e.g., stability, phosphorylation, kinase activity, interactions with other proteins, etc. In some embodiments, the present invention provides methods of decreasing Btk enzymatic activity. In some embodiments, such methods include contacting a Btk with an effective amount of a Btk inhibitor. Therefore, the present invention further provides methods of inhibiting Btk enzymatic activity by contacting a Btk with a Btk inhibitor of the present invention. One embodiment of the invention includes a method of treating a disorder responsive to inhibition of Btk in a subject comprising administering to said subject an effective amount of at least one compound described herein, or a pharmaceutically acceptable salt thereof. In one embodiment, the present invention provides methods of treating autoimmune disorders, inflammatory disorders, and cancers in a subject in need thereof comprising administering to said subject an effective amount of at least one compound described herein, or a pharmaceutically acceptable salt thereof. The term “autoimmune disorders” includes diseases or disorders involving inappropriate immune response against native antigens, such as acute disseminated encephalomyelitis (ADEM), Addison's disease, alopecia areata, antiphospholipid antibody syndrome (APS), autoimmune hemolytic anemia, autoimmune hepatitis, bullous pemphigoid (BP), Coeliac disease, dermatomyositis, diabetes mellitus type 1, Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome (GBS), Hashimoto's disease, idiopathic thrombocytopenic purpura, lupus erythematosus, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, pemphigus vulgaris, pernicious anaemia, polymyositis, primary biliary cirrhosis, Sjogren's syndrome, temporal arteritis, and Wegener's granulomatosis. The term “inflammatory disorders” includes diseases or disorders involving acute or chronic inflammation such as allergies, asthma, prostatitis, glomerulonephritis, pelvic inflammatory disease (PID), inflammatory bowel disease (IBD, e.g., Crohn's disease, ulcerative colitis), reperfusion injury, rheumatoid arthritis, transplant rejection, and vasculitis. In some embodiments, the present invention provides a method of treating rheumatoid arthritis or lupus. In some embodiments, the present invention provides a method of treating multiple sclerosis. The term “cancer” includes diseases or disorders involving abnormal cell growth and/or proliferation, such as glioma, thyroid carcinoma, breast carcinoma, lung cancer (e.g. small-cell lung carcinoma, non-small-cell lung carcinoma), gastric carcinoma, gastrointestinal stromal tumors, pancreatic carcinoma, bile duct carcinoma, ovarian carcinoma, endometrial carcinoma, prostate carcinoma, renal cell carcinoma, lymphoma (e.g., anaplastic large-cell lymphoma), leukemia (e.g. acute myeloid leukemia, T-cell leukemia, chronic lymphocytic leukemia), multiple myeloma, malignant mesothelioma, malignant melanoma, and colon cancer (e.g. microsatellite instability-high colorectal cancer). In some embodiments, the present invention provides a method of treating leukemia or lymphoma. As used herein, the term “subject” and “patient” may be used interchangeably, and means a mammal in need of treatment, e.g., companion animals (e.g., dogs, cats, and the like), farm animals (e.g., cows, pigs, horses, sheep, goats and the like) and laboratory animals (e.g., rats, mice, guinea pigs and the like). Typically, the subject is a human in need of treatment. As used herein, the term “treating” or “treatment” refers to obtaining desired pharmacological and/or physiological effect. The effect can be therapeutic, which includes achieving, partially or substantially, one or more of the following results: partially or totally reducing the extent of the disease, disorder or syndrome; ameliorating or improving a clinical symptom or indicator associated with the disorder; or delaying, inhibiting or decreasing the likelihood of the progression of the disease, disorder or syndrome. The effective dose of a compound provided herein, or a pharmaceutically acceptable salt thereof, administered to a subject can be 10 μg-500 mg. Administering a compound described herein, or a pharmaceutically acceptable salt thereof, to a mammal comprises any suitable delivery method. Administering a compound described herein, or a pharmaceutically acceptable salt thereof, to a mammal includes administering a compound described herein, or a pharmaceutically acceptable salt thereof, topically, enterally, parenterally, transdermally, transmucosally, via inhalation, intracisternally, epidurally, intravaginally, intravenously, intramuscularly, subcutaneously, intradermally or intravitreally to the mammal. Administering a compound described herein, or a pharmaceutically acceptable salt thereof, to a mammal also includes administering topically, enterally, parenterally, transdermally, transmucosally, via inhalation, intracisternally, epidurally, intravaginally, intravenously, intramuscularly, subcutaneously, intradermally or intravitreally to a mammal a compound that metabolizes within or on a surface of the body of the mammal to a compound described herein, or a pharmaceutically acceptable salt thereof. Thus, a compound or pharmaceutically acceptable salt thereof as described herein, may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the compound or pharmaceutically acceptable salt thereof as described herein may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, or wafers, and the like. Such compositions and preparations should contain at least about 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions can be such that an effective dosage level will be obtained. The tablets, troches, pills, capsules, and the like can include the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; or a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent. The active compound may also be administered intravenously or intraperitoneally by infusion or injection. Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Exemplary pharmaceutical dosage forms for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions. In all cases, the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage. Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation can be vacuum drying and the freeze drying techniques, which can yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions. Exemplary solid carriers can include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the compounds or pharmaceutically acceptable salts thereof as described herein can be dissolved or dispersed at effective levels, optionally with the aid of nontoxic surfactants. Useful dosages of a compound or pharmaceutically acceptable salt thereof as described herein can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949, which is incorporated by reference in its entirety. The amount of a compound or pharmaceutically acceptable salt thereof as described herein, required for use in treatment can vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and can be ultimately at the discretion of the attendant physician or clinician. In general, however, a dose can be in the range of from about 0.1 to about 10 mg/kg of body weight per day. The a compound or pharmaceutically acceptable salt thereof as described herein can be conveniently administered in unit dosage form; for example, containing 0.01 to 10 mg, or 0.05 to 1 mg, of active ingredient per unit dosage form. In some embodiments, a dose of 5 mg/kg or less can be suitable. The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals. The disclosed method can include a kit comprising a compound or pharmaceutically acceptable salt thereof as described herein and instructional material which can describe administering a compound or pharmaceutically acceptable salt thereof as described herein or a composition comprising a compound or pharmaceutically acceptable salt thereof as described herein to a cell or a subject. This should be construed to include other embodiments of kits that are known to those skilled in the art, such as a kit comprising a (such as sterile) solvent for dissolving or suspending a compound or pharmaceutically acceptable salt thereof as described herein or composition prior to administering a compound or pharmaceutically acceptable salt thereof as described herein or composition to a cell or a subject. In some embodiments, the subject can be a human. EXEMPLIFICATIONS LCMS methods: Samples were analyzed on a Waters Acquity UPLC BEH C18 1.7 uM 2.1×50 mm, part number 186002350 machine, MS mode: MS:ESI+ scan range 100-1000 daltons. PDA detection 210-400 nm. The method utilized was 95% water/5% MeCN (initial conditions) linear gradient to 5% H2O/95% MeCN at 1 min, HOLD 5% H2O/95% MeCN to 1.3 min at 0.7 ml/min in 0.1% trifluoroacetic acid (0.1% v/v) and the injection volume was 0.5 uL. SFC separations: Each separation was run with conditions as specified in the examples below, including column name/part number, separation method, backpressure regulator setting on the SFC system, flowrate, detection wavelength, injection volume, sample concentration, and sample diluent. Example 1: 5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,2,4-oxadiazole-3-carboxamide 1. Synthesis of N-(3-bromophenethyl)-4-methylbenzenesulfonamide To a mixture of 2-(3-bromophenyl)ethanamine (2 g, 10 mmol) in CH2Cl2(10 mL), triethylamine (2.02 g, 20 mmol) and TsCl (2.18 g, 11.5 mmol) were added at 0° C. The mixture was stirred at rt for 2 h, diluted with NaOH (1N, 100 mL) and extracted with CH2Cl2(100 mL). The organic layer was washed with water (100 mL), brine (100 mL), dried (Na2SO4) and concentrated in vacuo to give N-(3-bromophenethyl)-4-methylbenzenesulfonamide (3.5 g, yield: 100%) as a yellow oil. ESI-MS (M+H)+: 354.0.1H NMR (400 MHz, CDCl3) δ: 7.69 (d, J=8.0 Hz, 2H), 7.34 (d, J=8.4 Hz, 1H), 7.30 (d, J=8.0 Hz, 2H), 7.17 (t, J=1.6 Hz, 1H), 7.13 (t, J=8.0 Hz, 1H), 7.03-7.02 (m, 1H), 4.52 (t, J=6.0 Hz, 1H), 3.22-3.17 (m, 2H), 2.73 (t, J=6.8 Hz, 2H), 2.45 (s, 3H). 2. Synthesis of ethyl 2-(N-(3-bromophenethyl)-4-methylphenylsulfonamido)acetate To a mixture of N-(3-bromophenethyl)-4-methylbenzenesulfonamide (7.2 g, 20 mmol) in (CH3)2CO (80 mL), K2CO3(19.3 g, 140 mmol) and ethyl 2-bromoacetate (3.67 g, 22 mmol) were added. The mixture was stirred at 60° C. for 12 h, cooled to rt and the salt was filtered out. The resulting filtrate was concentrated in vacuo to give ethyl 2-(N-(3-bromophenethyl)-4-methylphenylsulfonamido)acetate (8.78 g, yield: 100%) as a yellow oil. ESI-MS (M+H)+: 440.0.1H NMR (400 MHz, CDCl3) δ: 7.70 (d, J=8.4 Hz, 2H), 7.34 (d, J=8.4 Hz, 1H), 7.28 (d, J=8.0 Hz, 2H), 7.14 (t, J=7.6 Hz, 1H), 7.10-7.08 (m, 2H), 4.08 (q, J=7.6 Hz, 2H), 3.98 (s, 2H), 3.44 (t, J=7.6 Hz, 2H), 2.85 (t, J=7.2 Hz, 2H), 2.42 (s, 3H), 1.19 (t, J=7.2 Hz, 3H). 3. Synthesis of 2-(N-(3-bromophenethyl)-4-methylphenylsulfonamido)acetic acid To a solution of ethyl 2-(N-(3-bromophenethyl)-4-methylphenylsulfonamido)acetate (8.78 mg, 20 mmol) in EtOH (40 mL) and H2O (40 mL) was added NaOH (1.6 g, 40 mmol). The reaction mixture was stirred at rt for 12 h. Then the solvent was reduced and the residue was adjusted to pH=3 with HCl (1 N). The mixture was extracted with EtOAc (100 mL×3). The organic layers were dried over (Na2SO4) and concentrated in vacuo to give 2-(N-(3-bromophenethyl)-4-methylphenylsulfonamido)acetic acid as a yellow solid (8.2 g, yield: 100%). ESI-MS (M+H)+: 412.0.1H NMR (400 MHz, CDCl3) δ: 7.69 (d, J=8.0 Hz, 2H), 7.34 (d, J=7.6 Hz, 1H), 7.29 (d, J=8.4 Hz, 2H), 7.22 (s, 1H), 7.14 (t, J=8.0 Hz, 1H), 7.08-7.06 (m, 1H), 4.00 (s, 2H), 3.45 (t, J=7.6 Hz, 2H), 2.83 (t, J=7.6 Hz, 2H), 2.42 (s, 3H). 4. Synthesis of 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-one To a solution of 2-(N-(3-bromophenethyl)-4-methylphenylsulfonamido)acetic acid (8.2 g, 20 mmol) in CH2Cl2(100 mL) was added SOCl2(11.9 g, 100 mmol) and DMF (cat.). The reaction mixture was stirred at 40° C. for 1 h. Then the solvent was removed under reduced pressure and dried in vacuo for 2 h. The residue was dissolved in CH2Cl2(100 mL) and cooled in an ice bath. AlCl3(10.56 g, 80 mmol) was added and the mixture was stirred at 0° C.-rt for 12 h. The mixture was poured into conc. HCl (20 mL) and extracted with EtOAc (100 mL×2). The organic layers were washed with water (100 mL), brine (100 mL), dried (Na2SO4), and concentrated in vacuo to afford a residue which was purified by silica gel column (petroleum ether:EtOAc=4:1) to give 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-one as a yellow solid (1.88 g, yield: 24%). ESI-MS (M+H)+: 394.1.1H NMR (400 MHz, CDCl3) δ: 7.42 (d, J=8.4 Hz, 2H), 7.38 (dd, J=8.4, 1.6 Hz, 1H), 7.31-7.29 (m, 2H), 7.14 (d, J=8.0 Hz, 2H), 4.21 (s, 2H), 3.68 (t, J=6.8 Hz, 2H), 2.93 (t, J=7.2 Hz, 2H), 2.39 (s, 3H). 5. Synthesis of 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-amine Synthesis of 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-amine was similar to that of Example 2. The residue was purified by silica gel column (CH2Cl2:MeOH=20:1) to give 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-amine as a yellow solid (154 mg, yield: 64%). ESI-MS (M+H)+: 395.1.1H NMR (400 MHz, CDCl3) δ: 7.66 (d, J=8.4 Hz, 2H), 7.37 (d, J=8.4 Hz, 2H), 7.31 (dd, J=8.4, 1.6 Hz, 1H), 7.27 (d, J=1.6 Hz, 1H), 7.18 (d, J=8.4 Hz, 1H), 4.12-4.40 (m, 1H), 3.42-3.36 (m, 2H), 3.19-3.12 (m, 2H), 2.96-2.89 (m, 2H), 2.41 (s, 3H). 6. Synthesis of 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-amine A mixture of 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-amine (1.2 g, 3.04 mmol) in HBr/HOAc (33%, 20 mL) was stirred at 70° C. for 12 h. After cooling down, the mixture was diluted with EtOAc (60 mL) and the resulting precipitate was filtered and dried under vacuum to give 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-amine (870 mg, yield: 71%) as a white solid. ESI-MS (M+H)+: 241.1.1H NMR (400 MHz, CDCl3) δ: 7.65-7.63 (m, 2H), 7.25 (d, J=8.8 Hz, 1H), 5.17-5.14 (m, 1H), 3.84-3.80 (m, 1H), 3.69-3.65 (m, 1H), 3.44-3.40 (m, 2H), 3.27-3.14 (m, 2H). 7. Synthesis of tert-butyl-amino-7-bromo-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate To a mixture of 7-bromo-3-tosyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-amine (680 mg, 1.7 mmol) and triethylamine (515 mg, 5.1 mmol) in CH2Cl2(10 mL), Boc2O (333 mg, 1.0 mmol) was added. The mixture was stirred at rt for 2 h. After diluting with CH2Cl2(100 mL), the organic layer was washed with water (30 mL) and brine (30 mL), dried (Na2SO4), filtered and concentrated in vacuo to give tert-butyl 1-amino-7-bromo-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate (450 mg, yield: 77%) as a yellow oil. ESI-MS (M+H)+: 341.0.1H NMR (400 MHz, CDCl3) δ: 7.31 (d, J=8.0 Hz, 1H), 7.26 (s, 1H), 7.19-7.11 (m, 1H), 4.17-4.10 (m, 1H), 3.83-3.66 (m, 2H), 3.48-3.45 (m, 1H), 3.37-3.14 (m, 2H), 2.78-2.73 (m, 1H), 1.47 (s, 9H). 8. Synthesis of tert-butyl 7-bromo-1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate To a mixture of potassium 5-(tert-butyl)-1,2,4-oxadiazole-3-carboxylate (358 mg, 1.5 mmol) in CH2Cl2(10 mL), (COCl)2(567 mg, 4.5 mmol) and DMF (cat.) were added at 0° C. The mixture was stirred at room temperature for 1 h. The mixture was concentrated. The residue was dried in vacuo, then dissolved in CH2Cl2(10 mL), tert-butyl 1-amino-7-bromo-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate (408 mg, 1.5 mmol) and triethylamine (454 mg, 4.5 mmol) were added. The mixture was stirred at rt for 12 h and diluted with CH2Cl2(100 mL). The organic phase was washed with water (50 mL) and brine (50 mL) and concentrated. The residue was purified by silica gel column (PE (petroleum ether): EtOAc (ethyl acetate)=4:1) to give tert-butyl 7-bromo-1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate (290 mg, yield: 38%) as a white solid. ESI-MS (M+H−56)+: 437.0.1H NMR (400 MHz, CD3OD) δ: 7.42-7.38 (m, 2H), 7.26 (d, J=7.6 Hz, 1H), 5.39 (d, J=5.2 Hz, 1H), 4.14-4.08 (m, 1H), 4.01-3.87 (m, 1H), 3.75-3.70 (m, 2H), 3.56-3.46 (m, 1H), 3.24-3.15 (m, 1H), 3.00-2.91 (m, 1H), 1.48 (s, 9H), 1.38 (s, 9H). 9. Synthesis of 2-chloro-4-methoxypyrimidine To a solution of 2,4-dichloropyrimidine (7.5 g, 50 mmol,) in MeOH (80 mL) was added dropwise into a solution of NaOMe (2.84 g, 52.5 mmol) in MeOH (20 mL) at 0° C. The reaction mixture was stirred at 0° C. for 2 h. The reaction mixture was concentrated in vacuo to give crude product. The crude product was poured into 150 mL of water. The aqueous phase was extracted with EtOAc (100 mL). The organic layer was dried with Na2SO4and concentrated in vacuum to give 2-chloro-4-methoxypyrimidine (5.9 g, yield: 82%) as a light yellow solid. ESI-MS (M+H)+: 145.0.1H NMR (400 MHz, CDCl3) δ: 8.27 (d, J=6.0 Hz, 1H), 6.65 (d, J=6.0 Hz, 1H), 3.99 (s, 3H). 10. Synthesis of 4-methoxy-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine To a solution of 2-chloro-4-methoxypyrimidine (120 g, 0.83 mol) in dioxane (2 L) was added 1-methyl-1H-pyazol-4-amine hydrochloride (111 g, 0.83 mol), Cs2CO3(0.83 kg, 2.5 mol), S-Phos (13.3 g, 0.03 mol) and Pd2(dba)3(16.7 g, 0.02 mol). The reaction mixture was stirred at 120° C. under N2for 4 h. The reaction mixture was cooled to room temperature and water (4 L) was added. The layers were separated and the aqueous phase was extracted with EtOAc (3×2 L). The combined organic layers were washed with brine (3 L), dried (Na2SO4) and concentrated. The crude material was purified by silica gel chromatography (PE:EtOAc=5:1 to 1:1) to give 4-methoxy-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (73 g, yield: 43%) as a tan solid. ESI-MS (M+H)+: 205.8. 11. Synthesis of 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine To 4-methoxy-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (1.4 kg, 6.8 mol) was added HBr (11.2 L, 38% aqueous). The reaction mixture was heated to 100° C. and stirred at that temperature for 2 h. The reaction mixture was concentrated and then POCl3(11.2 L) was added. The reaction mixture was heated to 100° C. and stirred at that temperature for 16 h. The reaction mixture was cooled to room temperature and concentrated. Water (10 L) was added to the residue and the pH of the solution was adjusted to pH=14 with aqueous NaOH (4 M). The basic aqueous phase was extracted with EtOAc (3×10 L). The combined organic layers were washed with brine (9 L), dried (Na2SO4) and concentrated. The crude material was purified by silica gel chromatography (PE:EtOAc=5:1 to 2:1) to give 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine as a white solid (770 g, yield:54%). ESI-MS (M+H)+: 210.0. 12. Synthesis of tert-butyl 1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate To a mixture of tert-butyl 7-bromo-1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate (510 mg, 1.03 mmol) and PinB-BPin (263 mg, 1.0 mmol) in dry 1,4-dioxane (10 mL), KOAc (303 mg, 3.09 mmol) and Pd(dppf)Cl2·CH2Cl2(81 mg, 0.1 mmol) were added quickly under N2. The mixture was stirred at 100° C. for 12 h under N2. After cooling down, 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (237 mg, 1.13 mmol), K2CO3(213 mg, 1.54 mmol) and H2O (2.5 mL) were added. The mixture was stirred at 100° C. for 12 h under N2. After cooling down, the mixture was concentrated and purified by silica gel column (CH2Cl2:PE:EtOAc=1:1:1) to give tert-butyl 1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate (250 mg, yield: 41%) as a yellow solid. ESI-MS (M+H)+: 588.2.1H NMR (400 MHz, CD3OD) δ: 8.41 (d, J=4.8 Hz, 1H), 7.99-7.98 (m, 3H), 7.64 (s, 1H), 7.49 (d, J=8.4 Hz, 1H), 7.21 (d, J=5.2 Hz, 1H), 5.54-5.47 (m, 1H), 4.18-4.05 (m, 1H), 3.97-3.90 (m, 4H), 3.87-3.76 (m, 2H), 3.65-3.57 (m, 1H), 3.15-3.11 (m, 1H), 1.49 (s, 9H), 1.39 (s, 9H). 13. Synthesis of 5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,2,4-oxadiazole-3-carboxamide To a solution of TFA (1 mL) in CH2Cl2(2 mL) was added tert-butyl 1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate (230 mg, 0.39 mmol). The mixture was stirred at rt for 1 h, then concentrated and purified by prep-HPLC (CH3CN/water NH4HCO30.05% as mobile phrase) to give 5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,2,4-oxadiazole-3-carboxamide (120 mg, yield: 58%). ESI-MS (M+H)+: 488.3.1H NMR (400 MHz, CD3OD) δ: 8.37-8.35 (m, 1H), 7.95 (s, 1H), 7.91-7.88 (m, 2H), 7.63 (s, 1H), 7.46 (d, J=7.6 Hz, 1H), 7.15 (d, J=4.8 Hz, 1H), 5.34 (d, J=7.2 Hz, 1H), 3.95 (s, 3H), 3.24-3.19 (m, 1H), 3.11-3.08 (m, 3H), 2.99-2.94 (m, 2H), 1.49 (s, 9H). Example 2. (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 2) Method 1: 1. Preparation of 3-(3-Bromo-benzylamino)-propionic acid ethyl ester To a solution of ethyl 3-aminopropanoate (46.0 g, 0.3 mol) and 3-bromobenzaldehyde (55.5 g, 0.3 mol) in MeOH (1.2 L) were added triethylamine (60.7 g, 0.6 mol) and NaCNBH3(56.5 g, 0.9 mol) portion-wise. The resulting mixture was stirred at rt for 4 h. The reaction mixture was concentrated in vacuo and the residue was diluted with water (600 mL). The mixture was extracted with EtOAc (3×500 mL). The combined organic layer was washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 3-(3-bromo-benzylamino)-propionic acid ethyl ester (46.5 g, yield: 54%) as a light yellow oil.1H NMR (DMSO-d6, 300 MHz): δ 7.52 (s, 1H), 7.40 (d, J=7.5 Hz, 1H), 7.31-7.25 (m, 2H), 4.04 (q, J=7.2 Hz, 2H), 3.67 (s, 2H), 2.69 (t, J=7.2 Hz, 2H), 2.42 (t, J=6.9 Hz, 2H), 1.17 (t, J=6.9 Hz, 3H). 2. Preparation of 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionic acid ethyl ester To a solution of 3-(3-bromo-benzylamino)-propionic acid ethyl ester (45.6 g, 0.16 mol) in pyridine (500 mL) was added TosCl (61.0 g, 0.32 mol) at rt. The reaction mixture was stirred at 120° C. for 16 h. The solvent was removed in vacuo to give the crude product. The crude product was purified by column chromatography on silica gel (petroleum ether:EtOAc=10:1 to 5:1) to afford 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionic acid ethyl ester (61 g, yield: 88%) as a light yellow oil.1H NMR (DMSO-d6, 300 MHz): δ 7.74 (d, J=8.4 Hz, 2H), 7.49-7.41 (m, 4H), 7.31 (d, J=5.1 Hz, 2H), 4.33 (s, 2H), 3.93 (q, J=7.2 Hz, 2H), 3.32 (t, J=7.2 Hz, 2H), 2.41 (s, 3H), 2.36 (t, J=6.9 Hz, 2H), 1.10 (t, J=6.9 Hz, 3H). 3. Preparation of 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionic acid To a solution of 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionic acid ethyl ester (60.0 g, 0.14 mol) in a mixed solvent of EtOH (600 mL) and H2O (60 mL) was added NaOH (11.2 g, 0.28 mol) portion-wise, the reaction solution was stirred at 60° C. for 4 h. The reaction solution was cooled to 0° C. and acidified to pH=5 with concentrated HCl. The solvent was concentrated in vacuo to give a residue which was extracted with EtOAc (3×150 mL). The organic layer was dried with Na2SO4, filtered, and concentrated in vacuo to give 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionic acid (45.2 g, yield: 78.6%) as a white solid.1H NMR (DMSO-d6, 300 MHz): δ 12.28 (br, 1H), 7.74 (d, J=8.1 Hz, 2H), 7.49-7.41 (m, 4H), 7.32 (d, J=5.1 Hz, 2H), 4.33 (s, 2H), 3.29 (t, J=6.9 Hz, 2H), 2.41 (s, 3H), 2.27 (t, J=7.5 Hz, 2H). 4. Preparation of 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionyl chloride To a solution of 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionic acid (45.2 g, 0.11 mol) in CH2Cl2(1000 mL) were added dropwise DMF (1 mL) and oxalyl chloride (27.9 g, 0.22 mol) portion-wise. The reaction solution was stirred at 55° C. for 2 h. The mixture was concentrated in vacuo to give the crude 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionyl chloride (47.2 g, yield: 99%) as a black oil which was used in the next step without further purification. 5. Preparation of 8-Bromo-2-(toluene-4-sulfonyl)-1,2,3,4-tetrahydro-benzo[c]azepin-5-one To a solution of 3-[(3-Bromo-benzyl)-(toluene-4-sulfonyl)-amino]-propionyl chloride (47.0 g, 0.11 mol) in anhydrous CH2Cl2(1200 mL) was added AlCl3(29.3 g, 0.22 mol) portion-wise at rt. The reaction mixture was stirred at 55° C. for 2 h. The reaction mixture was poured into ice water (1.2 L) and extracted with (500 mL). The organic layer was concentrated in vacuo to give the crude product. The crude product was purified by column chromatography on silica gel (petroleum ether:EtOAc=5:1 to 2:1) to afford 8-bromo-2-(toluene-4-sulfonyl)-1,2,3,4-tetrahydro-benzo[c]azepin-5-one (35 g, yield: 81%) as a white solid.1H NMR (DMSO-d6, 300 MHz): δ 7.65 (d, J=8.4 Hz, 3H), 7.60-7.51 (m, 2H), 7.36 (d, J=8.1 Hz, 2H), 4.68 (s, 2H), 3.42 (t, J=9.2 Hz, 2H), 2.96 (t, J=6.3 Hz, 2H), 2.37 (s, 3H). 6. Preparation of [8-Bromo-2-(toluene-4-sulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl]-carbamic acid tert-butyl ester To a solution of 8-bromo-2-(toluene-4-sulfonyl)-1,2,3,4-tetrahydro-benzo[c]azepin-5-one (32.0 g, 0.08 mol) in EtOH (600 mL) were added NH4OAc (18.5 g, 0.24 mol) and NaCNBH3(14.9 g, 0.24 mol) portion-wise at rt. Then the reaction mixture was stirred at 95° C. for 16 h. The mixture was poured into ice water (500 mL) and then EtOH was removed in vacuo. The residue was extracted with CH2Cl2(3×500 mL). The combined solvent was concentrated. The residue was redissolved in CH2Cl2(300 mL) and were added triethylamine (12.2 g, 0.12 mol) and (Boc)2O (34.6 g, 0.12 mol) at rt. The mixture was stirred at rt for 4 h and then concentrated in vacuo to give the crude product. The crude product was purified by column chromatography on silica gel (peteroleum ether:EtOAc=8:1 to 2:1) to afford [8-bromo-2-(toluene-4-sulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl]-carbamic acid tert-butyl ester (16.7 g, yield: 42%) as a white solid.1H NMR (DMSO d6, 300 MHz): δ 7.62-7.51 (m, 2H), 7.47 (d, J=9.9 Hz, 1H), 7.41-7.34 (m, 3H), 7.10 (d, J=8.4 Hz, 1H), 4.81-4.74 (m, 1H), 4.53 (d, J=15.0 Hz, 1H), 4.28 (d, J=15.3 Hz, 1H), 3.64-3.57 (m, 1H), 3.41-3.30 (m, 1H), 2.35 (s, 3H), 1.85-1.77 (m, 1H), 1.69-1.63 (m, 1H), 1.36 (s, 9H). 7. Preparation of 8-Bromo-2-(toluene-4-sulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-ylamine A solution of [8-bromo-2-(toluene-4-sulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl]-carbamic acid tert-butyl ester (14.8 g, 0.03 mol) in HCl/EtOAc (150 mL) was stirred at 25° C. for 4 h. The resulting solid was filtered and washed with MeOH and Et2O to give the product 8-bromo-2-(toluene-4-sulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-ylamine (10.5 g, yield: 89%) as a white solid.1H NMR (DMSO-d6, 300 MHz): δ 8.79 (br, 3H), 7.64-7.58 (m, 3H), 7.53 (s, 1H), 7.36 (d, J=8.4 Hz, 2H), 7.15 (d, J=8.4 Hz, 1H), 4.71-4.61 (m, 2H), 4.31 (d, J=15.3 Hz, 1H), 3.82 (d, J=18.3 Hz, 1H), 2.38 (s, 3H), 2.14-2.07 (m, 1H), 1.77-1.71 (m, 1H). LC-MS: m/z 395.0/397.0 [M+H]+. 8. Synthesis of 8-bromo-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-amine A solution of 8-bromo-2-(toluene-4-sulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-ylamine (2.00 g, 5.06 mmol) in HBr (33% solution in acetic acid, 20 mL) was heated at 50° C. for 12 h. After cooling to rt, the mixture was diluted EtOAc (50 mL). The white solid was collected by filtration and dried in vacuo to afford crude product 8-bromo-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-amine (1.66 g, yield: 82%), which was used directly in the next step. ESI-MS (M+H)±241.1.1H NMR (400 MHz, CD3OD) δ: 7.72-7.55 (m, 2H), 7.18 (d, J=8.4 Hz, 1H), 4.99-4.98 (m, 1H), 4.51 (d, J=14.4 Hz, 1H), 4.39 (d, J=14.4 Hz, 1H), 3.62-3.49 (m, 2H), 2.38-2.24 (m, 1H), 2.16-2.00 (m, 1H). 9. Synthesis of tert-butyl 5-amino-8-bromo-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate To a solution of 8-bromo-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-amine (640 mg, 1.60 mmol) and triethylamine (490 mg, 4.8 mmol) in CH2Cl2(20 mL) was added (Boc)2O (314 mg, 1.44 mmol). The mixture was stirred at rt for 1 h. After diluting with CH2Cl2(100 mL), the mixture was washed with brine (20 mL×2). The organic phase was concentrated in vacuo and the residue was purified by prep-HPLC (CH3CN/H2O with 0.05% NH3·H2O as mobile phase) to give tert-butyl 5-amino-8-bromo-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate as a colorless oil (364 mg, yield: 67%). ESI-MS (M+H)+: 341.1. 10. The Preparation of tert-butyl 8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate Synthesis of tert-butyl 8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate was similar to that of tert-butyl 7-bromo-1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate in Example 1, Step 8. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give tert-butyl 8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate as a yellow solid (4.5 g, yield: 70%). ESI-MS (M+H)+: 493.3. 11. The Preparation of tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate Synthesis of tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate was similar to that of tert-butyl 1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate in Example 1, Step 12. The crude product was purified by silica gel column chromatograph (EtOAc/MeOH=15:1) to give tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate as yellow solid (1.2 g, yield: 28%). ESI-MS (M+H)+: 588.3.1H NMR (400 MHz, CD3OD) δ: 8.43-8.38 (m, 1H), 8.11-7.95 (m, 3H), 7.67-7.48 (m, 2H), 7.25-7.24 (m, 1H), 5.67-5.63 (m, 1H), 4.84-4.77 (m, 1H), 4.55-4.50 (m, 1H), 4.17-4.09 (m, 1H), 3.94-3.88 (m, 3H), 3.64-3.54 (m, 1H), 2.11-2.08 (m, 2H), 1.43-1.34 (m, 9H), 1.22 (s, 9H). 12. The Preparation of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide To a solution of tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate (600 mg, 1.0 mmol) in CH2Cl2(5 mL) was added TFA (5 mL), the mixture was stirred 1 h at rt. After concentration, the crude product (440 mg, yield: 85%) was used in the next step without further purification. ESI-MS (M+H)+: 488.3. 13. The Preparation of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide To a solution of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (180 mg, 0.36 mmol) and dihydrofuran-3(2H)-one (132 mg, 1.8 mmol) in MeOH (20 mL) were added NaBH3CN (66 mg, 1.08 mmol) and ZnCl2(246 mg, 1.8 mmol). The mixture was stirred at rt for 16 h. After concentration and diluting with water (30 mL), the mixture was extracted with EtOAc (80 mL×2). The combined organic layer was washed with H2O (60 mL) and concentrated. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (114 mg, yield: 49%). ESI-MS (M+H)+: 544.3.1H NMR (400 MHz, CD3OD) δ: 8.30 (d, J=5.2 Hz, 1H), 7.92-7.85 (m, 3H), 7.51 (s, 1H), 7.35 (d, J=8.4 Hz, 1H), 7.11 (d, J=5.2 Hz, 1H), 5.50 (d, J=9.6 Hz, 1H), 4.67-4.55 (m, 4H), 3.84-3.70 (m, 3H), 3.80 (s, 3H), 2.95-2.89 (m, 1H), 2.76-2.72 (m, 1H), 2.15-2.12 (m, 1H), 1.95-1.92 (m, 1H), 1.40 (s, 9H). 14. The Preparation of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide was subjected to SFC separation (OD-H (2×25 cm), 30% methanol/CO2, 100 bar, 60 mL/min, 220 nm, inj vol.: 1.5 mL, 9 mg/mL, methanol) and yielded 34.8 mg of peak-1 (chemical purity 99%, ee >99%) and 37.1 mg of peak-2 (chemical purity 99%, ee >99%). Peak 2 was assigned as 5-tert-butyl-1,2,4-oxadiazole-3-carboxylic acid {(R)-8-[2-(1-methyl-1H-pyrazol-4-ylamino)-pyrimidin-4-yl]-2-oxetan-3-yl-2,3,4,5-tetrahydro-1H-2-benzazepin-5-yl}-amide: LCMS: Rt 0.88 min, m/z 544.00. 1H NMR (400 MHz, METHANOL-d4) δ 8.40 (d, J=5.02 Hz, 1H), 7.87-8.09 (m, 3H), 7.63 (s, 1H), 7.45 (d, J=8.28 Hz, 1H), 7.20 (d, J=5.27 Hz, 1H), 5.60 (s, 1H), 4.55-4.77 (m, 4H), 3.89 (s, 3H), 3.75-3.85 (m, 3H), 2.75-3.10 (m, 2H), 1.89-2.42 (m, 2H), 1.51 (s, 9H). Method 2 1. Chiral Resolution of tert-butyl 5-amino-8-bromo-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate to give tert-butyl (R)-5-amino-8-bromo-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate compound with (11bS)-4-hydroxydinaphtho[2,1-d:1′,2′-f][1,3,2]dioxaphosphepine 4-oxide (1:1) To tert-butyl 5-amino-8-bromo-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate (800 g, 2.34 mol) was added MeOH (4.8 L) and (S)-(−)-1,1′-binaphthyl-2,2′-diyl hydrogenphosphate (816.6 g, 2.34 mol). The mixture was stirred at 25° C. for 30 min and formed a yellow slurry. The slurry was stirred at reflux (70° C.) to give a yellow solution. The mixture was concentrated to dryness and IPAc (3.44 L) was added. The mixture was heated to 70° C. and was stirred at that temperature for 3 h. The reaction mixture was cooled to room temperature and an additional portion of IPAc (3.44 L) was added. The reaction mixture continued to stir at room temperature for 16 h. The slurry was filtered on centrifuge and the cake was washed three times, each with 7 vol IPAc. The wet cake was briefly dried to give tert-butyl (R)-5-amino-8-bromo-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate compound with (11bS)-4-hydroxydinaphtho[2,1-d:1′,2′-f][1,3,2]dioxaphosphepine 4-oxide (1:1) (515 g. yield:64% ([assuming maximum recovery of 50%], 91.3% ee) as a white solid. The recrystallization process can be repeated to increase the ee to 97.2%. 2. The Preparation of tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate To a solution of potassium 5-(tert-butyl)-1,2,4-oxadiazole-3-carboxylate (800 mg, 4.0 mmol) and oxalyl chloride (2.0 g, 16 mmol) in CH2Cl2(10 mL) was added DMF (cat.). The mixture was stirred at room temperature for 2 h, and then was concentrated with CH2Cl2twice. The residue was diluted with CH2Cl2(20 mL) and tert-butyl (R)-5-amino-8-bromo-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate compound with (11bR)-4-hydroxydinaphtho[2,1-d:1′,2′-f][1,3,2]dioxaphosphepine 4-oxide (1:1) (2.0 g, 3.0 mmol) and triethylamine (900 mg, 9.0 mmol) were added. The mixture was stirred at room temperature for 2 h and concentrated. The crude product was purified by silica gel chromatograph y (PE:EtOAc=2:1) to give tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (1.2 g, yield: 80%). ESI-MS (M+Na)+: 515.1.1H NMR (400 MHz, CDCl3) δ: 7.48-7.36 (m, 2H), 7.24-7.18 (m, 1H), 5.61-5.50 (m, 1H), 4.70-4.51 (m, 1H), 4.38-4.29 (m, 1H), 4.05-3.85 (m, 1H), 3.53-3.48 (m, 1H), 2.24-2.16 (m, 2H), 1.48 (s, 9H), 1.40-1.37 (m, 9H). 3. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was similar to that of tert-butyl 1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate in Example 1, Step 12. The crude material was purified by silica gel chromatography (EtOAc:MeOH=15:1) to give tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (1.5 g, yield: 43%). ESI-MS (M+H)+: 588.3. 4. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide To a solution of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (1.4 g, 2.3 mmol) in CH2Cl2(10 mL) was added TFA (10 mL). The mixture was stirred for 1 h at room temperature. The reaction mixture was concentrated and the crude product (1.0 g, yield: 81%) was used in the next step without further purification. ESI-MS (M+H)+: 488.3. 5. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (I-RP33) Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide was similar to that of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide described above in method 1, step 13. The crude material was purified by silica gel chromatography (EtOAc:MeOH=20:1) to give (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (746 mg, Y: 67%). ESI-MS (M+H)+: 544.3.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J=5.2 Hz, 1H), 8.03-7.96 (m, 3H), 7.63 (s, 1H), 7.48 (d, J=8.0 Hz, 1H), 7.23 (d, J=5.2 Hz, 1H), 5.61 (d, J=9.6 Hz, 1H), 4.78-4.67 (m, 4H), 3.96-3.82 (m, 3H), 3.90 (s, 3H), 3.06-3.02 (m, 1H), 2.89-2.80 (m, 1H), 2.29-2.22 (m, 1H), 2.07-2.03 (m, 1H), 1.52 (s, 9H). Example 3. 5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 3) To a solution of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (200 mg, 0.41 mmol) in CH3CN (20 mL) were added 2-iodoethanol (141 mg, 0.82 mmol) and K2CO3(170 mg, 1.23 mmol). The mixture was stirred at 80° C. for 2 h. The mixture was diluted with EtOAc (100 mL), washed with water (60 mL) and concentrated. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (90 mg, yield: 32%). ESI-MS (M+H)+: 532.3.1H NMR (400 MHz, CD3OD) δ: 8.43 (d, J=5.2 Hz, 1H), 8.04-8.00 (m, 3H), 7.64 (s, 1H), 7.47 (d, J=8.0 Hz, 1H), 7.25 (d, J=5.0 Hz, 1H), 5.60 (d, J=9.6 Hz, 1H), 4.22-4.10 (m, 2H), 3.91 (s, 3H), 3.75 (t, J=6.0 Hz, 2H), 3.28-3.21 (m, 2H), 2.69-2.65 (m, 2H), 2.31-2.27 (m, 1H), 2.00-1.97 (m, 1H), 1.53 (s, 9H). Example 4. 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 4) Synthesis of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide was similar to that of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide in Example 15. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (98 mg, yield: 60%). ESI-MS (M+H)+: 566.2.1H NMR (400 MHz, CDCl3) δ: 8.45 (d, J=4.8 Hz, 1H), 8.02 (s, 1H), 7.94-7.92 (m, 2H), 7.57-7.48 (m, 3H), 7.07 (d, J=5.2 Hz, 1H), 6.96 (s, 1H), 5.72 (t, J=8.8 Hz, 1H), 4.85-4.81 (m, 1H), 4.57-4.53 (m, 1H), 4.03-3.97 (m, 1H), 3.93 (s, 3H), 3.66-3.63 (m, 1H), 2.74 (s, 3H), 2.39-2.21 (m, 2H), 1.49 (s, 9H). Example 5. 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 5) To a solution of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (180 mg, 0.36 mmol) and dihydrofuran-3(2H)-one (132 mg, 1.8 mmol) in MeOH (20 mL) were added NaBH3CN (66 mg, 1.08 mmol) and ZnCl2(246 mg, 1.8 mmol). The mixture was stirred at rt for 16 h. After concentration and diluting with water (30 mL), the mixture was extracted with EtOAc (80 mL×2). The combined organic layer was washed with H2O (60 mL) and concentrated. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (114 mg, yield: 49%). ESI-MS (M+H)+: 544.3.1H NMR (400 MHz, CD3OD) δ: 8.30 (d, J=5.2 Hz, 1H), 7.92-7.85 (m, 3H), 7.51 (s, 1H), 7.35 (d, J=8.4 Hz, 1H), 7.11 (d, J=5.2 Hz, 1H), 5.50 (d, J=9.6 Hz, 1H), 4.67-4.55 (m, 4H), 3.84-3.70 (m, 3H), 3.80 (s, 3H), 2.95-2.89 (m, 1H), 2.76-2.72 (m, 1H), 2.15-2.12 (m, 1H), 1.95-1.92 (m, 1H), 1.40 (s, 9H). Example 6. 5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 6) Synthesis of 5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide was similar to that of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide in Example 5. The crude product was purified by silica gel chromatography (EtOAc:MeOH=10:1) to give 5-(tert-butyl)-N-(2-(3-hydroxycyclobutyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (102 mg, yield: 53%). ESI-MS (M+H)+: 557.7.1H NMR (400 MHz, CD3OD) δ: 8.29 (d, J=5.2 Hz, 1H), 7.87 (s, 3H), 7.50 (s, 1H), 7.32 (d, J=8.8 Hz, 1H), 7.09 (d, J=5.6 Hz, 1H), 5.45 (d, J=10.0 Hz, 1H), 3.86-3.78 (m, 3H), 3.75 (s, 3H), 3.09-3.06 (m, 1H), 2.80-2.74 (m, 1H), 2.50-2.39 (m, 3H), 2.14-2.05 (m, 1H), 1.88-1.85 (m, 1H), 1.72-1.69 (m, 2H), 1.41 (s, 9H). Example 7. 5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 7) 1. Synthesis of 4-methoxy-N-(1-ethyl-1H-pyrazol-4-yl)pyrimidin-2-amine Synthesis of 4-methoxy-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine was similar to that of 4-methoxy-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine in Example 1, Step 10. The crude material was purified by silica gel chromatography (PE:EtOAc=1:1) to give 4-methoxy-N-(1-ethyl-1H-pyrazol-4-yl)pyrimidin-2-amine (420 mg, yield: 43%) as a tan solid. ESI-MS (M+H)+: 220.1. 2. Synthesis of 4-chloro-N-(1-eethyl-1H-pyrazol-4-yl)pyrimidin-2-amine To 4-methoxy-N-(1-ethyl-1H-pyrazol-4-yl)pyrimidin-2-amine (420 mg, 1.9 mmol) was added HBr (5 mL, 48% aqueous). The reaction mixture was heated to 100° C. and stirred at that temperature for 2 h. The reaction mixture was concentrated and then POCl3(5 mL) was added. The reaction mixture was heated to 100° C. and stirred at that temperature for 16 h. The reaction mixture was cooled to room temperature and poured into ice-water. The pH of the solution was adjusted to pH=8 with aqueous NaOH (5 M). The basic aqueous phase was extracted with EtOAc (2×30 mL). The combined organic layers were washed with brine (100 mL), dried (Na2SO4) and concentrated. The crude material was purified by silica gel chromatography (PE:EtOAc=1:1) to give 4-chloro-N-(1-ethyl-1H-pyrazol-4-yl)pyrimidin-2-amine as a white solid (220 mg, yield:51%). ESI-MS (M+H)+: 224.1. 3. Synthesis of tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate To a mixture of tert-butyl 8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (560 mg, 1.13 mmol) and PinB-BPin (288 mg, 1.10 mmol) in dry 1,4-dioxane (11 mL), KOAc (332 mg, 3.39 mmol) and Pd(dppf)Cl2·CH2Cl2(89 mg, 0.11 mmol) were added quickly under N2. The mixture was stirred at 100° C. for 12 h under N2. After cooling down, 4-chloro-N-(1-ethyl-1H-pyrazol-4-yl)pyrimidin-2-amine (301 mg, 1.35 mmol), K2CO3(312 mg, 2.26 mmol), Pd(dppf)Cl2·CH2Cl2(89 mg, 0.11 mmol) and H2O (2.5 mL) were added. The mixture was stirred at 80° C. for 2 h under N2. After cooling down, the mixture was concentrated and purified by silica gel column (PE:EtOAc=3:1) to give tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (200 mg, yield: 29%) as a yellow solid. ESI-MS (M+H)+: 602.2. 4. The Preparation of 5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide To a solution of tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (200 mg, 0.33 mmol) in CH2Cl2(5 mL) was added TFA (5 mL), the mixture was stirred 1 h at rt. After concentration, the crude product (166 mg, yield: 100%) was used in the next step without further purification. ESI-MS (M+H)+: 502.7. 5. Synthesis of 5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (I-RP1) To a solution of 5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (166 mg, 0.33 mmol) and dihydrofuran-3(2H)-one (119 mg, 1.65 mmol) in MeOH (18 mL) were added NaBH3CN (21 mg, 0.33 mmol) and ZnCl2(90 mg, 0.66 mmol). The mixture was stirred at rt for 3 h. After concentration and diluting with water (30 mL), the mixture was extracted with EtOAc (80 mL×2). The combined organic layer was washed with H2O (60 mL) and concentrated. The crude product was purified by prep-TLC (CH2Cl2:MeOH=20:1) to give 5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (60 mg, yield: 32%). ESI-MS (M+H)+: 557.7.1H NMR (400 MHz, CDCl3) δ: 8.42 (d, J=5.2 Hz, 2H), 7.90 (s, 1H), 7.85 (d, J=8.0 Hz, 1H), 7.79 (s, 1H), 7.57-7.53 (m, 2H), 7.04 (d, J=5.2 Hz, 1H), 5.71 (t, J=8.4 Hz, 1H), 4.82-4.66 (m, 4H), 4.17 (q, J=7.2 Hz, 2H), 3.91-3.72 (m, 3H), 2.99-2.92 (m, 1H), 2.63-2.58 (m, 1H), 2.40-2.34 (m, 1H), 2.15-2.07 (m, 1H), 1.51 (t, J=7.6 Hz, 3H), 1.45 (s, 9H). Example 8. 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 8) Synthesis of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide was similar to that of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide in Example 12, Step 4. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (20 mg, yield: 14%). ESI-MS (M+H)+: 572.3.1H NMR (400 MHz, CD3OD) δ: 8.30 (s, 1H), 7.91-7.86 (m, 3H), 7.53 (s, 1H), 7.36 (d, J=8.0 Hz, 1H), 7.10 (d, J=5.6 Hz, 1H), 5.48 (d, J=9.2 Hz, 1H), 4.19-3.88 (m, 4H), 3.78 (s, 3H), 3.30-3.21 (m, 2H), 3.16-3.03 (m, 2H), 2.67-2.62 (m, 1H), 2.16-2.11 (m, 1H), 1.96-1.83 (m, 3H), 1.64-1.52 (m, 2H), 1.38 (s, 9H). Example 9. (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 9) 1. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate A mixture of tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (1.2 g, 2.43 mmol), 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (650 mg, 2.56 mmol), KOAc (477 mg, 4.86 mmol) and Pd(dppf)Cl2·DCM (196 mg, 0.24 mmol) in 30 mL 1,4-dioxane was stirred at 90° C. for 2 h under nitrogen. After cooling to room temperature, the mixture was diluted with EtOAc (200 mL), washed with water (50 mL×2), dried with Na2SO4and concentrated to give tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate which was used for the next step without further purification. ESI-MS (M+H)+: 541.3. 2. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-chloropyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate A mixture of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (1.31 g, 2.43 mmol), 2,4-dichloropyrimidine (344 mg, 2.31 mmol), Pd(dppf)Cl2(196 mg, 0.24 mmol) and K2CO3(672 mg, 4.86 mmol) in 20 mL of dioxane/H2O (4:1) was stirred at 90° C. for 12 h under N2atmosphere. After removing the solvent, the residue was purified by silica-gel chromatography (CH2Cl2:MeOH=20:1) to give tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-chloropyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (900 mg, yield: 70% for two steps). ESI-MS (M+H)+: 527.2. 3. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate A mixture of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-chloropyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (900 mg, 1.71 mmol), 1,5-dimethyl-1H-pyrazol-4-amine (228 mg, 2.05 mmol), Cs2CO3(1.11 g, 3.42 mmol), Pd2(dba)3(157 mg, 0.17 mmol) and S-Phos (140 mg, 0.34 mmol) in 15 mL 1,4-dioxane was stirred at 100° C. for 2 h under nitrogen. The mixture was diluted with EtOAc (200 mL) and washed with water (60 mL×2). The organic phase was dried with Na2SO4and concentrated. The crude product was purified by silica gel chromatography (DCM/MeOH=20:1) to give tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (110 mg, yield: 11%). ESI-MS (M+H)+: 602.3.1H NMR (400 MHz, CDCl3) δ: 8.39-8.38 (m, 1H), 7.93-7.90 (m, 2H), 7.67 (s, 1H), 7.46-7.44 (m, 2H), 7.05 (d, J=4.2 Hz, 1H), 6.48-6.41 (m, 1H), 5.69-5.59 (m, 1H), 4.81-4.66 (m, 1H), 4.50-4.41 (m, 1H), 4.10-4.03 (m, 1H), 3.81 (s, 3H), 3.61-3.52 (m, 1H), 2.23 (s, 3H), 2.20-2.16 (m, 2H), 1.40 (s, 9H), 1.34-1.30 (m, 9H). 4. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide was like that of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide in Example 2. The crude material was carried forward without further purification. ESI-MS (M+H)+: 502.2. 5. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide was similar to that of 5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide in Example 3. The crude material was purified by silica gel chromatography (CH2Cl2:MeOH=10:1) to give (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2-hydroxyethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (43 mg, yield 42%). ESI-MS (M+H)+: 546.3.1H NMR (400 MHz, CD3OD) δ: 8.32 (d, J=5.2 Hz, 1H), 8.00-7.98 (m, 2H), 7.60 (s, 1H), 7.42 (d, J=8.0 Hz, 1H), 7.23 (d, J=5.2 Hz, 1H), 5.58-5.56 (m, 1H), 4.16-4.10 (m, 2H), 3.82 (s, 3H), 3.73 (t, J=6.0 Hz, 2H), 3.30-3.21 (m, 2H), 2.67-2.65 (m, 2H), 2.30-2.27 (m, 1H), 2.25 (s, 3H), 2.02-1.96 (m, 1H), 1.52 (s, 9H). Example 10. 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 10) Synthesis of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide was similar to that described in Example 7, Step 3. The crude material was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (97 mg, yield: 57%). ESI-MS (M+H)+: 558.3.1H NMR (400 MHz, CD3OD) δ: 8.43 (d, J=5.2 Hz, 1H), 8.04-7.99 (m, 3H), 7.64-7.63 (m, 1H), 7.48 (d, J=8.0 Hz, 1H), 7.24 (d, J=5.2 Hz, 1H), 5.61 (d, J=9.6 Hz, 1H), 4.10-3.91 (m, 4H), 3.90 (s, 3H), 3.78-3.71 (m, 2H), 3.36-3.10 (m, 3H), 2.33-2.20 (m, 2H), 2.06-1.96 (m, 2H), 1.52 (s, 9H). Example 11. (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 11) 1. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was similar to that of tert-butyl 1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate in Example 1, Step 12. The crude material was purified by silica gel chromatography (EtOAc:MeOH=15:1) to give tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (1.5 g, yield: 43%). ESI-MS (M+H)+: 588.3. 2. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide To a solution of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (1.4 g, 2.3 mmol) in CH2Cl2(10 mL) was added TFA (10 mL). The mixture was stirred for 1 h at room temperature. The reaction mixture was concentrated and the crude product (1.0 g, yield: 81%) was used in the next step without further purification. ESI-MS (M+H)+: 488.3. 3. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide To a solution of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (75 mg, 0.15 mmol) in CH3CN (5 mL) were added 2,2,2-trifluoroethyl trifluoromethanesulfonate (60 mg, 0.3 mmol) and DIPEA (34 mg, 0.3 mmol). The mixture was stirred at 80° C. for 2 h under microwave. The mixture was concentrated and purified by prep-TLC (DCM/MeOH=10:1) to give (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (45 mg, yield: 58%). ESI-MS (M+H)+: 570.2.1H NMR (400 MHz, CD3OD) δ: 8.30 (d, J=5.6 Hz, 1H), 7.92-7.86 (m, 3H), 7.50 (s, 1H), 7.35 (d, J=8.0 Hz, 1H), 7.10 (d, J=5.6 Hz, 1H), 5.48-5.50 (m, 1H), 4.26-4.22 (m, 1H), 4.02-3.98 (m, 1H), 3.78 (s, 3H), 3.32-3.27 (m, 1H), 3.21-3.17 (m, 1H), 3.05-2.98 (m, 2H), 2.17-2.07 (m, 1H), 1.91-1.82 (m, 1H), 1.40 (s, 9H). Example 12. 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 12) 1. The Preparation of tert-butyl 8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate To a solution of potassium 5-(tert-butyl)-1,3,4-oxadiazole-2-carboxylate (1.4 g, 6.7 mmol) and HATU (3.2 g, 8.4 mmol) in DMF (20 mL) were added triethylamine (1.69 g, 16.8 mmol) and tert-butyl 5-amino-8-bromo-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate (1.9 g, 5.6 mmol). The mixture was stirred at rt for 4 h. After diluting with water (80 mL), the mixture was extracted with EtOAc (100 mL×2). The combined organics were washed with brine (80 mL), dried, and concentrated. The crude product was purified by silica gel column chromatograph (PE/EtOAc=2:1) to give tert-butyl 8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate as yellow solid (1.8 g, yield: 62%). ESI-MS (M+H−56)+: 437.3.1H NMR (400 MHz, CD3OD) δ: 7.51-7.43 (m, 2H), 7.28-7.26 (m, 1H), 5.52-5.45 (m, 1H), 4.65-4.61 (m, 1H), 4.43-4.39 (m, 1H), 4.15-4.09 (m, 1H), 3.65-3.43 (m, 1H), 2.08-2.03 (m, 2H), 1.50 (s, 9H), 1.43-1.42 (m, 9H). 2. The Preparation of tert-butyl 5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate A mixture of tert-butyl 8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate (1.6 g, 3.2 mmol), PinB-BPin (802 mg, 3.84 mmol), KOAc (640 mg, 6.4 mmol) and Pd(dppf)Cl2·CH2Cl2(130 mg, 0.16 mmol) in 20 mL dry 1,4-dioxane was stirred at 80° C. for 16 h under nitrogen. After the mixture was cooled to rt, 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (802 mg, 3.84 mmol), Pd(dppf)Cl2·CH2Cl2(130 mg, 0.16 mmol), K2CO3(1.3 g, 9.6 mmol), and H2O (5 mL) were added and the resulting mixture was stirred at 80° C. for another 16 h. The mixture was diluted with EtOAc (200 mL), washed with water (80 mL×2), dried with Na2SO4, and concentrated. The crude product was purified by silica gel column chromatography (EtOAc/MeOH=20:1) to give tert-butyl 5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate as yellow solid (1.4 g, yield: 86%). ESI-MS (M+H)+: 588.3.1H NMR (400 MHz, CD3OD) δ: 8.42-8.38 (m, 1H), 8.11-7.95 (m, 3H), 7.66-7.50 (m, 2H), 7.24-7.21 (m, 1H), 5.64-5.62 (m, 1H), 4.85-4.47 (m, 2H), 3.93-3.88 (m, 1H), 3.57-3.54 (m, 1H), 2.12-2.10 (m, 2H), 1.51 (s, 9H), 1.43-1.34 (m, 9H) 3. The Preparation of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide To a solution of tert-butyl 5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate (600 mg, 1.02 mmol) in CH2Cl2(5 mL) was added TFA (5 mL). The mixture was stirred 1 h at rt. After removal of the solvent, the residue was dried in vacuo to give crude title product (460 mg, yield: 80%), which was used in the next step without further purification. ESI-MS (M+H)+: 488.3. 4. Synthesis of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide To a solution of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (170 mg, 0.35 mmol) and dihydro-pyran-4-one (175 mg, 1.75 mmol) in MeOH (30 mL) were added NaBH3CN (112 mg, 1.75 mmol) and CH3COOH (cat). The mixture was stirred at 50° C. for 4 h. After cooling to rt, the mixture was adjusted to pH=8 with conc. NH4OH. After concentration, the crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (24 mg, yield: 9%). ESI-MS (M+H)+: 572.3. H NMR (400 MHz, CD3OD) δ: 8.29 (d, J=5.2 Hz, 1H), 7.91-7.85 (m, 3H), 7.53 (s, 1H), 7.36 (d, J=8.0 Hz, 1H), 7.10 (d, J=5.6 Hz, 1H), 5.44 (d, J=9.2 Hz, 1H), 4.08-3.86 (m, 4H), 3.78 (s, 3H), 3.28-3.20 (m, 3H), 3.08-3.01 (m, 1H), 2.63-2.57 (m, 1H), 2.14-2.09 (m, 1H), 1.94-1.76 (m, 3H), 1.63-1.49 (m, 2H), 1.38 (s, 9H). Example 13. 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 13) Synthesis of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide was similar to that of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(tetrahydro-2H-pyran-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide in Example 8. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (59 mg, yield: 33%). ESI-MS (M+H)+: 544.2.1H NMR (400 MHz, CD3OD) δ: 8.29 (d, J=5.2 Hz, 1H), 7.91 (d, J=8.0 Hz, 1H), 7.86 (s, 1H), 7.83 (s, 1H), 7.51 (s, 1H), 7.36 (d, J=8.4 Hz, 1H), 7.09 (d, J=5.2 Hz, 1H), 5.46 (d, J=10.0 Hz, 1H), 4.65-5.55 (m, 4H), 3.83-3.67 (m, 3H), 3.77 (s, 3H), 2.96-2.93 (m, 1H), 2.79-2.73 (m, 1H), 2.13-2.10 (m, 1H), 1.93-1.90 (m, 1H), 1.38 (s, 9H). Example 14. (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 14a) and (S)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 14b) Method 1 5-tert-Butyl-1,3,4-oxadiazole-2-carboxylic acid {8-[2-(1-methyl-1H-pyrazol-4-ylamino)-pyrimidin-4-yl]-2-oxetan-3-yl-2,3,4,5-tetrahydro-1H-2-benzazepin-5-yl}-amide (264 mg) was subjected to SFC separation (OD-H (2×25 cm), 30% methanol (0.1% DEA)/CO2, 100 bar, 65 mL/min, 220 nm, inj vol.: 0.5 mL, 4 mg/mL, methanol) and yielded 93 mg of peak-1 (chemical purity>99%, ee>99%) and 97 mg of peak-2 (chemical purity>99%, ee>99%). Peak 1 was assigned as 5-tert-butyl-1,3,4-oxadiazole-2-carboxylic acid {(R)-8-[2-(1-methyl-1H-pyrazol-4-ylamino)-pyrimidin-4-yl]-2-oxetan-3-yl-2,3,4,5-tetrahydro-1H-2-benzazepin-5-yl}-amide: LCMS: Rt 0.84 min, m/z 544.2.1H NMR (400 MHz, METHANOL-d4) δ 8.41 (br. s., 1H), 7.84-8.14 (m, 3H), 7.63 (br. s., 1H), 7.47 (t, J=7.56 Hz, 1H), 7.21 (d, J=10.04 Hz, 1H), 5.57 (d, J=9.29 Hz, 1H), 4.61-4.80 (m, 4H), 3.66-4.07 (m, 6H), 3.05 (br. s., 1H), 2.88 (d, J=9.04 Hz, 1H), 2.23 (br. s., 1H), 2.05 (br. s., 1H), 1.50 (d, J=2.89 Hz, 9H). Peak 2 was assigned as 5-tert-butyl-1,3,4-oxadiazole-2-carboxylic acid {(S)-8-[2-(1-methyl-1H-pyrazol-4-ylamino)-pyrimidin-4-yl]-2-oxetan-3-yl-2,3,4,5-tetrahydro-1H-2-benzazepin-5-yl}-amide: LCMS: Rt 0.84 min, m/z 544.2.1H NMR (400 MHz, METHANOL-d4) δ 8.42 (d, J=5.02 Hz, 1H), 7.86-8.14 (m, 3H), 7.63 (s, 1H), 7.48 (d, J=8.09 Hz, 1H), 7.22 (d, J=5.27 Hz, 1H), 5.58 (d, J=9.73 Hz, 1H), 4.59-4.81 (m, 4H), 3.73-4.05 (m, 6H), 3.05 (br. s., 1H), 2.89 (br. s., 1H), 2.23 (br. s., 1H), 2.06 (d, J=5.52 Hz, 1H), 1.50 (s, 9H). Method 2 1. Synthesis of tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate To a solution of tert-butyl (R)-5-amino-8-bromo-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (3.1 g, 4.5 mmol) and triethylamine (910 mg, 9.0 mmol) in DCM (100 mL) were added HATU (2.6 g, 6.8 mmol) and potassium 5-(tert-butyl)-1,3,4-oxadiazole-2-carboxylate (1.12 g, 5.4 mmol). The mixture was stirred at rt for 2 h. Then water (100 mL) was added and the mixture was extracted with DCM (2×100 mL). The combined organics were dried and concentrated. The crude product was purified by silica gel column chromatography (petroleum ether/EtOAc=4:1) to give tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as white solid (1.8 g, yield: 82%). ESI-MS (M+H)+: 493.1. 2. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate A mixture of tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (1.8 g, 3.65 mmol), bis(pinocolato)diboron (975 mg, 3.84 mmol), KOAc (715 mg, 7.30 mmol) and Pd(dppf)Cl2·DCM (293 mg, 0.36 mmol) in 30 mL 1,4-dioxane was stirred at 90° C. for 2 h under nitrogen. After cooling to rt, the mixture was diluted with EtOAc (200 mL), washed with water (2×50 mL), dried with Na2SO4and concentrated. The crude product tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was used for next step without purification. ESI-MS (M+H)+: 541.3. 3. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate A mixture of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (4.85 g, 8.98 mmol), 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (1.88 g, 8.98 mmol), Pd(dppf)Cl2(734 mg, 0.9 mmol) and K2CO3(2.48 g, 18 mmol) in dioxane/H2O (4:1, 20 mL) was degassed and stirred at 100° C. for 2 h under a N2atmosphere. After concentration of the reaction mixture, the residue was purified by silica-gel chromatography (EtOAc:PE=2:1) to give tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (3.4 g, yield: 51%). ESI-MS (M+H)+: 588.3. 4. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-ol-1,3,4-oxadiazole-2-carboxamide To a solution of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (3.4 g, 5.79 mmol) in DCM (10 mL) was added TFA (4 mL). The resulting solution was stirred at room temperature for 2 h. After concentration of the reaction mixture, the residue was dissolved in MeOH (10 mL) and adjusted to pH=8 with aqueous ammonia. Then water (20 mL) was added and the mixture was extracted with DCM/MeOH solutions (20:1, 30 mL×3). The organic phase was dried and concentrated to give crude (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (2.47 g, yield: 88%), which was used to next step without further purification. ESI-MS (M+H)+: 488.2. 5. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (I-RP38) To a solution of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (1.5 g, 3.08 mmol) and oxetan-3-one (1.1 g, 15.4 mmol) in MeOH (30 mL) was added NaBH3CN (970 mg, 15.4 mmol) and ZnCl2(4.2 g, 30.8 mmol). The resulting mixture was stirred at room temperature for 4 h. After diluting with H2O (20 mL), the mixture was extracted with DCM/MeOH solutions (20:1, 20 mL×3). The combined organic layers were dried and concentrated. The residue was purified by silica-gel chromatography (DCM:MeOH=50:1 to 20:1) to give (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (1.1 g, yield: 58%). ESI-MS (M+H)+: 544.3.1H NMR (400 MHz, CDCl3) δ: 8.42 (d, J=5.2 Hz, 1H), 8.32 (br, 1H), 7.86-7.78 (m, 3H), 7.53-7.26 (m, 2H), 7.15 (s, 1H), 7.05-7.03 (m, 1H), 5.62 (t, J=8.4 Hz, 1H), 4.75-4.67 (m, 4H), 3.94-3.85 (m, 6H), 3.02-2.96 (m, 1H), 2.75-2.70 (m, 1H), 2.35-2.31 (m, 1H), 2.14-2.07 (m, 1H), 1.47 (s, 9H). Example 15. 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 15) To a solution of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (100 mg, 0.20 mmol) in CH2Cl2(20 mL) were added MsCl (34 mg, 0.3 mmol) and triethylamine (61 mg, 0.60 mmol). The mixture was stirred at rt for 2 h. The mixture was diluted with CH2Cl2(100 mL), washed with brine (60 mL), and concentrated. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(methylsulfonyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (130 mg, yield: 87%). ESI-MS (M+H)+: 566.3.1H NMR (400 MHz, DMSO-d6) δ: 10.01 (d, J=7.6 Hz, 1H), 9.57 (s, 1H), 8.49 (d, J=5.2 Hz, 1H), 8.07 (s, 1H), 8.04 (d, J=8.4 Hz, 1H), 7.99 (br, 1H), 7.50 (s, 1H), 7.47 (d, J=8.4 Hz, 1H), 7.27 (d, J=5.2 Hz, 1H), 5.56-5.51 (m, 1H), 4.75-4.70 (m, 1H), 4.60-4.56 (m, 1H), 3.86-3.84 (m, 1H), 3.83 (s, 3H), 3.61-3.58 (m, 1H), 2.80 (s, 3H), 2.15-2.09 (m, 2H), 1.43 (s, 9H). Example 16. (R)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 16) 1. Synthesis of tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate To a solution of tert-butyl (R)-5-amino-8-bromo-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (3.1 g, 4.5 mmol) and triethylamine (910 mg, 9.0 mmol) in DCM (100 mL) were added HATU (2.6 g, 6.8 mmol) and potassium 5-(tert-butyl)-1,3,4-oxadiazole-2-carboxylate (1.12 g, 5.4 mmol). The mixture was stirred at rt for 2 h. Then water (100 mL) was added and the mixture was extracted with DCM (2×100 mL). The combined organics were dried and concentrated. The crude product was purified by silica gel column chromatography (petroleum ether/EtOAc=4:1) to give tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as white solid (1.8 g, yield: 82%). ESI-MS (M+H)+: 493.1. 2. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate A mixture of tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (1.8 g, 3.65 mmol), bis(pinocolato)diboron (975 mg, 3.84 mmol), KOAc (715 mg, 7.30 mmol) and Pd(dppf)Cl2·DCM (293 mg, 0.36 mmol) in 30 mL 1,4-dioxane was stirred at 90° C. for 2 h under nitrogen. After cooling to rt, the mixture was diluted with EtOAc (200 mL), washed with water (2×50 mL), dried with Na2SO4and concentrated. The crude product tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was used for next step without purification. ESI-MS (M+H)+: 541.3. 3. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-chloropyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate A mixture of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate, 2,4-dichloropyrimidine (648 mg, 4.38 mmol), K2CO3(1.0 g, 7.30 mmol) and Pd(dppf)Cl2·DCM (293 mg, 0.36 mmol) in 30 mL 1,4-dioxane and 6 mL water was stirred at 90° C. for 12 h under nitrogen. The mixture was dilute with EtOAc (200 mL) and washed with water (2×60 mL). The organic phase was dried with Na2SO4and concentrated. The crude product was purified by silica gel column chromatography (DCM/MeOH=20:1) to give tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-chloropyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (1.3 g, yield: 67% for two steps). ESI-MS (M+H)+: 527.2. 4. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was similar to that of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (Example 9, Step 3). The crude product was purified by silica gel column chromatography (EtOAc/petroleum ether=3:1) to give tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (280 mg, yield: 78%). ESI-MS (M+H)+: 616.3. 5. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide was similar to that of (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Example 9, Step 4). Crude (R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as yellow solid (240 mg) was used to next step without further purification. ESI-MS (M+H)+: 516.3. 6. Synthesis of (R)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide To a solution of (R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (240 mg, 0.47 mmol) and 2-iodoethanol (200 mg, 1.17 mmol) in 10 mL of CH3CN was added K2CO3(195 mg, 1.41 mmol). The resulting mixture was stirred at 80° C. for 24 h. After diluting with H2O (20 mL), the mixture was extracted with DCM/MeOH solutions (20:1, 3×40 mL). The combined organic layers were dried (Na2SO4) and concentrated. The residue was purified by prep-TLC (DCM/MeOH=10:1) to give (R)-5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (108 mg, yield: 42%). ESI-MS (M+H)+: 560.3.1H NMR (400 MHz, CD3OD) δ: 8.37 (d, J=5.2 Hz, 1H), 8.03 (s, 1H), 7.98-7.95 (m, 2H), 7.66 (s, 1H), 7.44 (d, J=8.4 Hz, 1H), 7.17 (d, J=5.2 Hz, 1H), 5.55 (d, J=9.6 Hz, 1H), 4.54-4.37 (m, 1H), 4.17-4.04 (m, 2H), 3.71 (t, J=6.0 Hz, 2H), 3.30-3.17 (m, 2H), 2.29-2.25 (m, 2H), 2.29-2.25 (m, 1H), 1.99-1.95 (m, 1H), 1.52 (d, J=6.4 Hz, 6H), 1.49 (s, 9H). Example 17. (R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 17) A mixture of (R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (220 mg, 0.43 mmol), oxetan-3-one (157 mg, 2.15 mmol), NaBH3CN (135 mg, 2.15 mmol) and ZnCl2(585 mg, 4.30 mmol) in 10 mL MeOH was stirred at rt for 16 h. The mixture was dilute with EtOAc (150 mL) and washed with water (50 mL×2). The organic phase was dried with Na2SO4and concentrated. The crude product was purified by silica gel chromatography (DCM:MeOH=15:1) to give (R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (146 mg, yield: 60%). ESI-MS (M+H)+: 572.3.1H NMR (400 MHz, CDCl3) δ: 8.42 (d, J=5.2 Hz, 1H), 8.05-7.97 (m, 3H), 7.67 (s, 1H), 7.48 (d, J=8.0 Hz, 1H), 7.22 (d, J=8.4 Hz, 1H), 5.59-5.57 (m, 1H), 4.87-4.66 (m, 4H), 4.53-4.50 (m, 1H), 3.98-3.81 (m, 3H), 3.09-3.03 (m, 1H), 2.92-2.86 (m, 1H), 2.29-2.20 (m, 1H), 2.07-2.03 (m, 1H), 1.54-1.51 (m, 15H) Example 18. (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 18) 1. Synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate A mixture of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (4.85 g, 8.98 mmol), 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (1.88 g, 8.98 mmol), Pd(dppf)Cl2(734 mg, 0.9 mmol) and K2CO3(2.48 g, 18 mmol) in dioxane/H2O (4:1, 20 mL) was degassed and stirred at 100° C. for 2 h under a N2atmosphere. After concentration of the reaction mixture, the residue was purified by silica-gel chromatography (EtOAc:PE=2:1) to give tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (3.4 g, yield: 51%). ESI-MS (M+H)+: 588.3. 2. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide To a solution of tert-butyl (R)-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (3.4 g, 5.79 mmol) in DCM (10 mL) was added TFA (4 mL). The resulting solution was stirred at room temperature for 2 h. After concentration of the reaction mixture, the residue was dissolved in MeOH (10 mL) and adjusted to pH=8 with aqueous ammonia. Then water (20 mL) was added and the mixture was extracted with DCM/MeOH solutions (20:1, 30 mL×3). The organic phase was dried and concentrated to give crude (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (2.47 g, yield: 88%), which was used to next step without further purification. ESI-MS (M+H)+: 488.2. 3. Synthesis of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide To a solution of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (1.5 g, 3.1 mmol) and DIPEA (800 mg, 6.2 mmol) in CH3CN (30 mL) was added 2,2,2-trifluoroethyl trifluoromethanesulfonate (1.1 g, 4.7 mmol). The mixture was stirred at 50° C. for 12 h. After diluting with water (100 mL), the mixture was extracted with DCM (100 mL×3). The combined organic layers were washed with brine (100 mL), dried (Na2SO4), filtered and concentrated. The crude product was purified by silica gel chromatography (DCM:MeOH=10:1) to give (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (1.2 g, yield: 68%). ESI-MS (M+H)+: 570.2.1H NMR (400 MHz, CD3OD) δ: 8.31 (d, J=5.2 Hz, 1H), 7.94-7.88 (m, 3H), 7.51 (s, 1H), 7.38 (d, J=8.0 Hz, 1H), 7.12 (d, J=5.2 Hz, 1H), 5.49-5.47 (m, 1H), 4.27-4.23 (m, 1H), 4.04-4.00 (m, 1H), 3.78 (s, 3H), 3.33-3.24 (m, 2H), 3.04-2.98 (m, 2H), 2.14-2.05 (m, 1H), 1.87-1.84 (m, 1H), 1.40 (s, 9H). Example 19. 5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 19) Synthesis of 5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide was similar to that of 5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide in Example 3. The crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(2-(2-hydroxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (165 mg, yield: 84%). ESI-MS (M+H)+: 532.2.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J=5.2 Hz, 1H), 8.03-8.00 (m, 3H), 7.64 (s, 1H), 7.48 (d, J=8.0 Hz, 1H), 7.23-7.22 (m, 1H), 5.58 (d, J=9.6 Hz, 1H), 4.21-4.09 (m, 2H), 3.90 (s, 3H), 3.74 (t, J=5.6 Hz, 2H), 3.20-3.18 (m, 2H), 2.70-2.62 (m, 2H), 2.30-2.28 (m, 1H), 2.00-1.98 (m, 1H), 1.52 (s, 9H). Example 20. 5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 20) 1. Synthesis of (S)-tetrahydrofuran-3-yl trifluoromethanesulfonate To a solution of (S)-tetrahydrofuran-3-ol (500 mg, 5.7 mmol) and pyridine (538 mg, 6.8 mmol) in DCM (15 mL) at −10° C. was added Tf2O (1.8 g, 6.3 mmol). The mixture was stirred at −10° C. for 0.5 h. The mixture was quenched with 2N HCl solution. The organic layer were separated, dried over Na2SO4and filtered. The resulting DCM solution of (S)-tetrahydrofuran-3-yl trifluoromethanesulfonate was used for next step. ESI-MS (M+H)+: 221.0. 2. Synthesis of 5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide To a solution of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (1.7 g, 3.5 mmol) in THE (24 mL) and DMF (3 mL) was added KHMDS (5.2 mL, 5.2 mmol, 1M solution) dropwise at −78° C. The solution was stirred under nitrogen at −78° C. for 0.5 h. Then, the solution of (S)-tetrahydrofuran-3-yl trifluoromethanesulfonate (from previous step) was added dropwise. The mixture was stirred at room temperature for 16 h. After diluting with water (40 mL), the mixture was extracted with DCM (40 mL×3). The combined organic layers were washed with brine (60 mL), dried (Na2SO4), filtered and concentrated. The crude product was purified by silica gel chromatography (DCM:MeOH=10:1) to give 5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (448 mg, yield: 23%). ESI-MS (M+H)+: 558.3.1H NMR (400 MHz, DMSO-d6) δ: 9.82-9.80 (m, 1H), 9.51 (s, 1H), 8.47 (d, J=4.8 Hz, 1H), 7.99-7.95 (m, 3H), 7.55 (s, 1H), 7.40 (d, J=8.0 Hz, 1H), 7.28 (d, J=5.2 Hz, 1H), 5.43-5.38 (m, 1H), 4.06-3.99 (m, 2H), 3.83-380 (m, 5H), 3.63-3.50 (m, 2H), 3.10-3.05 (m, 3H), 2.22-2.02 (m, 2H), 1.85-1.82 (m, 2H), 1.42 (s, 9H). Example 21. 5-(tert-butyl)-N—((R)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 21) To a solution of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (101 mg, 0.21 mmol) in MeOH (7 mL) was added (S)-2-methyloxirane (29 μL, 0.42 mmol) and cesium carbonate (135 mg, 0.42 mmol). The mixture was stirred at 60° C. for 16 h. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated and the crude product was purified by silica gel chromatography (DCM:MeOH=10:1) to give 5-(tert-butyl)-N—((R)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (50 mg, yield: 44%). ESI-MS (M+H)+: 546.0.1H NMR (400 MHz, METHANOL-d4) δ: 8.41 (d, J=5.3 Hz, 1H), 8.07-8.02 (m, 2H), 7.97 (s, 1H), 7.63 (s, 1H), 7.48 (d, J=8.5 Hz, 1H), 7.22 (d, J=5.3 Hz, 1H), 5.62-5.53 (m, 1H), 4.33-4.10 (m, 2H), 4.04-3.95 (m, 1H), 3.89 (s, 3H), 3.33-3.27 (m, 2H), 2.58-2.46 (m, 2H), 2.38-2.23 (m, 1H), 2.07-1.95 (m, 1H), 1.48 (s, 9H), 1.15 (d, J=6.3 Hz, 3H). Example 22. 5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 22) 1. Synthesis of (R)-tetrahydrofuran-3-yl trifluoromethanesulfonate Synthesis of (R)-tetrahydrofuran-3-yl trifluoromethanesulfonate) was similar to that of (S)-tetrahydrofuran-3-yl trifluoromethanesulfonate) in Example 20, Step 1. The resulting DCM solution was used for the next step. ESI-MS (M+H)+: 221.0. 2. Synthesis of 5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (I-RP58) Synthesis of 5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide was similar to that of 5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide in Example 20, Step 2. The crude product was purified by silica gel chromatography (DCM:MeOH=10:1) to give 5-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (50 mg, yield: 38%). ESI-MS (M+H)+: 558.1.1H NMR (400 MHz, METHANOL-d4) δ: 8.38 (d, J=5.3 Hz, 1H), 8.03-7.95 (m, 3H), 7.60 (s, 1H), 7.45 (d, J=8.5 Hz, 1H), 7.19 (d, J=5.3 Hz, 1H), 5.56 (br d, J=9.3 Hz, 1H), 4.12-4.01 (m, 2H), 4.01-3.90 (m, 2H), 3.88 (s, 3H), 3.79-3.68 (m, 2H), 3.29-3.21 (m, 1H), 3.20-3.09 (m, 1H), 2.33-2.11 (m, 2H), 2.09-1.90 (m, 2H), 1.48 (s, 9H). Example 23. 1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 23) 1. The Preparation of tert-butyl 8-bromo-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate To a solution of tert-butyl 8-bromo-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (556 mg, 3.06 mmol) in CH2Cl2(10 mL) were added HATU (1.16 g, 3.06 mmol) and DIPEA (592 mg, 4.6 mmol). The mixture was stirred at rt for 1 h before 1-(tert-butyl)-1H-1,2,3-triazole-4-carboxylic acid (1.04 g, 3.06 mmol) was added. The mixture was stirred at rt for 12 h. The mixture was diluted with CH2Cl2(200 mL), washed with water (100 mL), brine (100 mL), dried and concentrated. The crude product was purified by silica gel column (PE:EtOAc=1:1) to give tert-butyl 8-bromo-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (1.1 g, yield: 73%) as a yellow solid. ESI-MS (M+H)+: 492.1.1H NMR (400 MHz, CDCl3) δ: 8.17 (s, 1H), 7.35-7.33 (m, 2H), 7.21 (d, J=8.0 Hz, 1H), 5.52-5.42 (m, 1H), 4.54-4.34 (m, 2H), 4.04-3.86 (m, 1H), 3.65-3.55 (m, 1H), 2.80 (s, 9H), 2.10-2.07 (m, 2H), 1.41 (s, 9H). 2. The Preparation of tert-butyl 5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate Synthesis of tert-butyl 5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was similar to that of tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate in Example 7. The crude product was purified by silica gel column chromatography (CH2Cl2:MeOH=20:1) to give tert-butyl 5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (170 mg, Y: 35%). ESI-MS (M+H)+: 601.2. 3. The Preparation of 1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide Synthesis of 1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide was similar to that of 5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide in Example 7. The crude product (135 mg, yield: 95%) was used in the next step without further purification. ESI-MS (M+H)+: 501.2. 4. The Preparation of 1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide Synthesis of 1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide was similar to that of 5-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide in Example 7. The crude product was purified by silica gel column chromatography (EtOAc:MeOH=10:1) to give 1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (67 mg, yield: 45%). ESI-MS (M+H)+: 556.7.1H NMR (400 MHz, DMSO-d6) δ: 9.48 (s, 1H), 9.02 (d, J=8.8 Hz, 1H), 8.76 (s, 1H), 8.46 (d, J=5.2 Hz, 1H), 7.97-7.91 (m, 3H), 7.56 (s, 1H), 7.37 (d, J=8.0 Hz, 1H), 7.25 (d, J=5.2 Hz, 1H), 5.47-5.43 (m, 1H), 4.61-1.47 (m, 4H), 4.13-4.08 (m, 2H), 3.88-3.65 (m, 3H), 2.90-2.78 (m, 2H), 2.15-2.06 (m, 1H), 1.88-1.82 (m, 1H), 1.65 (s, 9H), 1.36 (t, J=7.2 Hz, 3H). Example 24. (R)-1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 24a) and (S)-1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 24b) 1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (50 mg) was subjected to SFC separation (2.1×25.0 cm (S,S) Whelk0-1, 58% methanol with 0.5% isopropylamine/CO2, 120 bar, 85 mL/min, 230 nm, methanol) and yielded 23 mg of peak-1 (chemical purity 99%, ee >99%) and 24 mg of peak-2 (chemical purity 99%, ee=99%). Peak 1 is assigned as (R)-1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide: LCMS: Rt 4.0 min, m/z 557.20.1H NMR (400 MHz, METHANOL-d4) δ ppm 8.51 (s, 1H), 8.40 (d, J=5.27 Hz, 1H), 8.05-7.92 (m, 3H), 7.64 (s, 1H), 7.47 (d, J=8.28 Hz, 1H), 7.21 (d, J=5.27 Hz, 1H), 5.57 (br d, J=9.04 Hz, 1H), 4.78-4.71 (m, 1H), 4.71-4.65 (m, 3H), 4.17 (q, J=7.28 Hz, 2H), 4.01-3.91 (m, 1H), 3.91-3.78 (m, 2H), 3.15-2.98 (m, 1H), 2.88 (ddd, J=12.99 Hz, 9.60 Hz, 3.51 Hz, 1H), 2.36-2.14 (m, 1H), 2.11-1.87 (m, 1H), 1.73 (s, 9H), 1.46 (t, J=7.28 Hz, 3H). Peak 2 is assigned as (S)-1-(tert-butyl)-N-(8-(2-((1-ethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide: LCMS: Rt 5.3 min, m/z 557.00.1H NMR (400 MHz, METHANOL-d4) δ ppm 8.51 (s, 1H), 8.40 (d, J=5.27 Hz, 1H), 8.06-7.92 (m, 3H), 7.64 (s, 1H), 7.47 (d, J=8.03 Hz, 1H), 7.21 (d, J=5.27 Hz, 1H), 5.57 (br d, J=9.29 Hz, 1H), 4.77-4.70 (m, 1H), 4.70-4.63 (m, 2H), 4.17 (q, J=7.28 Hz, 2H), 4.05-3.91 (m, 1H), 3.91-3.76 (m, 2H), 3.15-2.98 (m, 1H), 2.89 (ddd, J=12.8 Hz, 9.7 Hz, 3.6 Hz, 1H), 2.40-2.13 (m, 1H), 2.12-1.87 (m, 1H), 1.74 (s, 9H), 1.47 (t, J=7.28 Hz, 3H). Example 25. (R)-1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 25) 1. Synthesis of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate Synthesis of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was similar to that of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (Example 9, Step 3). The crude material was purified by silica gel chromatography (EtOAc:PE=4:1) to give tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (520 mg, yield: 56%). ESI-MS (M+H)+: 615.3. 2. Synthesis of (R)-1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide Synthesis of (R)-1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide was similar to that of (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide in Example 9, Step 4. The crude product was isolated as a yellow solid and was used for the next step without further purification (420 mg, yield: 96%). ESI-MS (M+H)+: 515.3. 3. Synthesis of (R)-1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (I-RP35) Synthesis of (R)-1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide was similar to that of (R)-5-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide in Example 17. The crude material was purified by silica gel chromatography (DCM:MeOH=20:1) to give (R)-1-(tert-butyl)-N-(8-(2-((1-isopropyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (116 mg, yield: 51%). ESI-MS (M+H)+: 571.2.1H NMR (400 MHz, CDCl3) δ: 8.42 (d, J=4.8 Hz, 1H), 8.17 (s, 1H), 8.07 (d, J=8.8 Hz, 1H), 7.94 (s, 1H), 7.85 (d, J=8.0 Hz, 1H), 7.77 (s, 1H), 7.57 (s, 1H), 7.48 (d, J=8.0 Hz, 1H), 7.11 (s, 1H), 7.03 (d, J=5.2 Hz, 1H), 5.64 (t, J=8.0 Hz, 1H), 4.78-4.68 (m, 4H), 4.53-4.45 (m, 1H), 3.91-3.80 (m, 3H), 3.04-2.99 (m, 1H), 2.82-2.79 (m, 1H), 2.27-2.23 (m, 1H), 2.10-2.05 (m, 1H), 1.70 (s, 9H), 1.55 (s, 3H), 1.52 (s, 3H). Example 26. 1-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo [c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 26a) & 1-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 26b) 1. Synthesis of tert-butyl (R)-8-bromo-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate Synthesis of tert-butyl (R)-8-bromo-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was similar to that of tert-butyl 8-bromo-5-(5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamido)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate in Example 12, Step 1. The crude product was purified by silica gel column chromatography (EtOAc/petroleum ether=1:2) to give tert-butyl (R)-8-bromo-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as yellow solid (2.3 g, yield: 95%). ESI-MS (M+H)+: 492.2. 2. Synthesis of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate Synthesis of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate was similar to the synthesis of tert-butyl (R)-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate described in Example 9, Step 1. The crude product was used for next step without purification. ESI-MS (M+H)+: 540.3. 3. Synthesis of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate To a solution of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (5.4 g, 10.0 mmol) in dioxane/H2O (100 mL) was added 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (2.1 g, 10.0 mmol), K2CO3(2.8 g, 20.0 mmol) and Pd(dppf)Cl2(0.4 g, 0.5 mmol) were added. The mixture was stirred at 100° C. for 16 h under nitrogen. After cooling to rt, the mixture was concentrated and purified by silica gel column chromatography (petroleum ether/EtOAc=1:3) to give tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (3.2 g, yield: 55%). ESI-MS (M+H)+: 586.7.1H NMR (400 MHz, CDCl3) δ: 8.42 (s, 1H), 8.19 (s, 1H), 8.02-7.87 (m, 3H), 7.68 (s, 1H), 7.54-7.49 (m, 2H), 7.06 (d, J=5.2 Hz, 1H), 5.63-5.58 (m, 1H), 4.83-4.67 (m, 1H), 4.51-4.47 (m, 1H), 4.02-4.00 (m, 1H), 3.93 (s, 3H), 3.65-3.62 (m, 1H), 2.14-2.12 (m, 2H), 1.72 (s, 9H), 1.41-1.38 (m, 9H). 4. Synthesis of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide To a solution of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (3.2 g, 5.5 mmol) in DCM (30 mL) was added TFA (30 mL). The mixture was stirred at rt for 3 h. The solvent was removed. The crude was dissolved in MeOH (30 mL)/water (20 mL). The mixture was basified with NH4OH to pH=8-9 and extracted with DCM (3×50 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to give (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a gray solid (2.6 g, yield: 98%). ESI-MS (M+H)+: 486.7. 5. Synthesis of 1-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide & 1-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide To a solution of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (500 mg, 1.0 mmol) in MeOH (30 mL) were added dihydrofuran-3(2H)-one (258 mg, 3.0 mmol), ZnCl2(682 mg, 5.0 mmol) and NaBH3CN (189 mg, 3.0 mmol). The mixture was stirred at 50° C. for 16 h. The mixture was concentrated and purified by silica gel column chromatography (DCM/MeOH=20/1 to 15/1) to give the racemic product as a yellow solid (542 mg, yield: 79%). 1-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((S)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (154 mg) and 1-(tert-butyl)-N—((R)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-((R)-tetrahydrofuran-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (167 mg) were separated by chiral resolution. ESI-MS (M+H)+: 557.3. Isomer 1:1H NMR (400 MHz, CD3OD) δ: 8.53 (s, 1H), 8.42 (d, J=5.2 Hz, 1H), 8.05-7.99 (m, 3H), 7.64 (s, 1H), 7.48 (d, J=8.4 Hz, 1H), 7.23 (d, J=5.2 Hz, 1H), 5.58 (d, J=10.4 Hz, 1H), 4.16-4.09 (m, 2H), 4.02-3.96 (m, 2H), 3.90 (s, 3H), 3.79-3.71 (m, 2H), 3.31-3.24 (m, 2H), 3.15-3.10 (m, 1H), 2.31-2.17 (m, 2H), 2.07-1.97 (m, 2H), 1.74 (s, 9H). Isomer 2:1H NMR (400 MHz, CD3OD) δ: 8.43 (s, 1H), 8.30 (d, J=5.2 Hz, 1H), 7.95-7.86 (m, 3H), 7.67 (s, 1H), 7.37 (d, J=8.0 Hz, 1H), 7.13 (d, J=5.2 Hz, 1H), 5.47 (d, J=9.6 Hz, 1H), 4.00 (br, 2H), 3.93-3.85 (m, 2H), 3.85 (s, 3H), 3.69-3.58 (m, 2H), 3.28-3.22 (m, 1H), 3.17-2.97 (m, 2H), 2.21-2.01 (m, 2H), 1.99-1.80 (m, 2H), 1.63 (s, 9H). Example 27a. (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 27) To a solution of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (500 mg, 1.0 mmol) in MeOH (30 mL) were added oxetan-3-one (216 mg, 3.0 mmol), ZnCl2(682 mg, 5.0 mmol) and NaBH3CN (189 mg, 3.0 mmol). The mixture was stirred at 50° C. for 3 h. The mixture was concentrated and the crude material was purified by silica gel chromatography (CH2Cl2:MeOH grading from 20:1 to 15:1) to give (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (307 mg, yield: 55%). ESI-MS (M+H)+: 542.7.1H NMR (400 MHz, CD3OD) δ: 8.54 (s, 1H), 8.42 (d, J=5.6 Hz, 1H), 8.04-7.98 (m, 3H), 7.71 (s, 1H), 7.49 (d, J=8.0 Hz, 1H), 7.24 (d, J=5.2 Hz, 1H), 5.59 (d, J=9.2 Hz, 1H), 4.79-4.75 (m, 1H), 4.71-4.69 (m, 3H), 4.00-3.83 (m, 6H), 3.09-3.05 (m, 1H), 3.94-2.88 (m, 1H), 2.29-2.21 (m, 1H), 2.07-2.04 (m, 1H), 1.75 (s, 9H). Example 27b. (R)-1-(tert-butyl)-N-(8-(2-((1-(methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide 1. Synthesis of 1-(methyl-d3)-1H-pyrazol-4-amine A mixture of 4-nitro-1-pyrazole (5.0 g, 44 mmol) and d6-dimethyl sulfate (10.0 g, 75.7 mmol) in 1 M solution of NaOH in water (50.0 mL) was heated at 35° C. overnight. The solid formed was filtered, washed with water, and dried (Na2SO4) to give 1-d3-methyl-4-nitro-1-pyrazole as a white crystal (3.9 g, yield: 68%). LCMS: RT 0.36 min.; MH+131.1; 1H NMR (400 MHz, DMSO-d6) δ: 8.84 (s, 1H), 8.23 (s, 1H). 2. Synthesis of 1-(methyl-d3)-4-nitro-1H-pyrazole A solution of 1-d3-methyl-4-nitro-1-pyrazole (3.9 g, 30 mmol) in EtOH (50.0 mL) was degassed with nitrogen, followed by the addition of 10% palladium on carbon (0.32 g, 0.30 mmol). The mixture was placed under an atmosphere of hydrogen and stirred at rt for 2 h. The mixture was filtered and the filtrate was concentrated in vacuo to give 1-(d3-methyl-1H-pyrazol-4-amine as an oil (2.9 g, yield: 96%) which was used in the next step without further purification. 3. Synthesis of 4-methoxy-N-(1-(methyl-d3)-1H-pyrazol-4-yl)pyrimidin-2-amine To a solution of 2-chloro-4-methoxypyrimidine (9.4 g, 65.1 mmol) in 1,4-dioxane (0.3 L) was added 1-methyl-d3-1H-pyazol-4-amine (8.5 g, 85 mmol), Cs2CO3(63.6 g, 195 mmol), S-Phos (13.3 g, 0.03 mol) and Pd2(dba)3(16.7 g, 0.02 mol). The reaction mixture was stirred at reflux under N2for 16 h. The reaction mixture was cooled to room temperature and the mixture was filtered through a silica gel pad, and washed with EtOAc (500 mL). The combined filtrates were concentrated in vacuo. The crude material was purified by silica gel chromatography (heptane:EtOAc=100:0 to 0:100) to give 4-methoxy-N-(1-(methyl-d3)-1H-pyrazol-4-yl)pyrimidin-2-amine (9.2 g, yield: 68%) as a light yellow solid. ESI-MS (M+H)+: 209.1.1H NMR (400 MHz, CDCl3) δ: 8.10 (d, J=5.7 Hz, 1H), 7.77 (s, 1H), 7.51 (s, 1H), 6.13 (d, J=5.7 Hz, 1H), 3.94 (s, 3H). 4. Synthesis of 4-chloro-N-(1-(methyl-d3)-1H-pyrazol-4-yl)pyrimidin-2-amine To 4-methoxy-N-(1-(methyl-d3)-1H-pyrazol-4-yl)pyrimidin-2-amine (9.1 g, 43.7 mmol) was added HBr (90 mL, 38% aqueous). The reaction mixture was heated to 100° C. and stirred at that temperature for 3 h. The reaction mixture was cooled to room temperature and concentrated in vacuo, azetroped with toluene (3×100 mL) and dried at 50° C. overnight to afford the HBr salt (16 g) as a yellow/brown solid. The salt was then dissolved in POCl3(250 mL) and heated to 100° C. for 36 hrs. The reaction mixture was cooled to room temperature and concentrated in vacuo and azetroped with Toluene (3×100 mL). the resulting residue was diluted with EtOAc (500 mL) and water (100 mL) and the layer were separated. The aqueous layer was extracted with EtOAc (3×100 mL) and the combined organic layers were washed with brine (200 mL), dried (Na2SO4), filtered and concentrated in vacuo to afford a residue which was triturated with EtOAc/heptanes (1:1) to afford added to 4-chloro-N-(1-(methyl-d3)-1H-pyrazol-4-yl)pyrimidin-2-amine as a white solid (7.4 g, yield:80%). ESI-MS (M+H)+: 213.0. 5. Synthesis of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-(methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate To a solution of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (4.6 g, 8.5 mmol) in 1,4-dioxane (100 mL) and water (20 mL) was added 4-chloro-N-(1-(methyl-d3)-1H-pyrazol-4-yl)pyrimidin-2-amine (1.8 g, 8.5 mmol), K2CO3(2.4 g, 17 mmol) and Pd(dppf)Cl2(0.7 g, 0.85 mmol) were added. The mixture was stirred at 100° C. for 16 h under nitrogen. After cooling to rt, the mixture was diluted with EtOAc (300 mL) and washed with saturated brine (100 mL). The aqueous layer was extracted with EtOAc (3×100 mL) and the organics were combined, dried (Na2SO4), filtered, concentrated in vacuo to afford a residue. The crude material was purified by silica gel column chromatography (gradient heptanes/EtOAc=100:0-0:100) to give tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-(methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate as a yellow solid (3.2 g, yield: 55%). ESI-MS (M+H)+: 590.4. 6. Synthesis of (R)-1-(tert-butyl)-N-(8-(2-((1-(methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide To a solution of tert-butyl (R)-5-(1-(tert-butyl)-1H-1,2,3-triazole-4-carboxamido)-8-(2-((1-(methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate (3.2 g, 5.5 mmol) in CH2Cl2(40 mL) was added TFA (40 mL). The mixture was stirred at rt for 3 h. The solvent was removed and the crude material was re-dissolved in MeOH (30 mL)/water (20 mL). The mixture was basified with NH4OH to pH=8-9 and extracted with CH2Cl2(3×100 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated in vacuo to give (R)-1-(tert-butyl)-N-(8-(2-((1-(methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (3.0 g, yield: 86%). ESI-MS (M+H)+: 490.2. 7. (R)-1-(tert-butyl)-N-(8-(2-((1-(methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (2.6 g, 15.9 mmol) was dissolved in MeOH (160 mL) and treated with oxetan-3-one (1.15 g, 16.0 mmol), ZnCl2(3.6 g, 26.5 mmol) and NaBH3CN (1.0 mg, 15.9 mmol). The mixture was stirred at 50° C. for 16 h, concentrated in vacuo and the crude material was purified by silica gel chromatography (gradient CH2Cl2:MeOH 100:10) to give a yellow solid which was further washed purified by dissolving in MeOH (100 mL) and CH2Cl2(500 mL) and washed with water (100 mL) and saturated brine (100 mL), the organic layer was separated, dried (Na2SO4), filtered and concentrated in vacuo to afford (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-d3)-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (2.32 g, 67% yield: 55%). ESI-MS (M+H)+: 546.3. 1H NMR (400 MHz, DMSO-d6) δ: 9.46 (s, 1H), 8.98 (d, J=8.3 Hz, 1H), 8.74 (s, 1H), 8.46 (d, J=5.6 Hz, 1H), 7.95 (m, 3H), 7.54 (s, 1H), 7.38 (d, J=8.1 Hz, 1H), 7.25 (d, J=5.2 Hz, 1H), 5.45 (m, 1H), 4.61-4.47 (m, 4H), 3.86 (m, 1H), 3.78 (m, 1H), 3.67 (m, 1H), 2.93 (m, 1H), 2.77 (m, 1H), 2.12 (m, 1H), 1.86 (m, 1H), 1.66 (s, 9H). Example 27c. Preparation of Crystalline Form A and Crystalline Form G of Compound 27 Compound (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (1200 g, 2.47 mol) was added to a 20 L reactor at room temperature (25° C.), followed by addition of 24 L of 1,2-dichloroethane at 25° C. To the solution was added oxetan-3-one (534 g, 24.7 mol), NaBH(OAc)3(523 g, 2.47 mol), and AcOH (24 mL, 0.17 eq). An additional amount of NaBH(OAc)3(1046 g, 4.94 mol) was added in portion to the reactor at room temperature. The mixture was stirred at 25° C. for 16 h. Ice water (12 kg) was added slowly to reactor at room temperature. The organic layer was separated and the aqueous layer was extracted with dichloromethane three time (3×12 L). The combined organic layer was washed with brine (20 L), dried over Na2SO4, filtered and concentrated. The crude material was purified by silica gel chromatography (CH2Cl2:MeOH=20:1) to afford (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(oxetan-3-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (compound 27). The purified compound 27 (950 g) and EtOH (5 L) were vigorously stirred for 4 h and the slurry was filtered and washed with 1 L EtOH. The resulting wet cake was dried under vacuum at 45° C. for ˜ 24 h until reaching a constant weight to afford crystalline Form A (960 g, yield 85.7%, purity 99%). 300.2 mg of crystalline Form A was weighed into a 20 mL glass vial followed by addition of 6 mL of isopropyl acetate (IPAc) for suspension. The sample was magnetically stirred at 50° C. with a rate of ˜1000 rpm for three days. Solids were isolated by filtration after three days and then dried at room temperature under vacuum for about 5 h to yield crystalline Form G. Alternatively, to 2.1 g of crystalline Form A was charged 15 volumes of dichloromethane (mL/g). Distill the resulting mixture under atmospheric conditions using a Dean-Stark trap under the conditions of Tj−Tr=20 K and Tjmax=110° C., wherein Tj=jacket temperature, Tr=reaction/reactor temperature and Tjmax=max jacket temperature. 5 mL or 2 volumes of dichloromethane was removed, followed by addition of 2 volumes of isopropyl acetate (IPAc). Continue to distill under the conditions of Tj−Tr=40 K and Tjmax=110° C. until 10 mL or 5 volumes of the solvent(s) was removed. 5 volumes of IPAc was then added, followed by continued distillation to remove another 10 mL or 5 volumes of the solvent(s). 5 volumes of IPAc was added followed by an additional 30 mL of IPAc. The resulting mixture was stirred and temperature was cycled over the weekend between 20° C. to 60° C. to form a slurry and Form G was isolated from the slurry. Powder X-Ray Diffraction Crystallinity of the compound was studied using a XRD-D8 X-ray powder diffractometer using Cu Ka radiation (Bruker, Madison, WI). The instrument is equipped with a long fine focus X-ray tube. The tube voltage and amperage were set to 40 kV and 40 mA, respectively. The divergence and scattering slits were set at 1° and the receiving slit was set at 0.15 mm. Diffracted radiation was detected by a Lynxeye detector. A 0-20 continuous scan at 1.6°/min from 3 to 42° 2θ was used. The sample was prepared for analysis by placing it on a zero background plate. The powder X-ray diffraction (PXRD) pattern of crystalline Form A is shown inFIG.1and the main peaks are listed in Table 1. The PXRD pattern of crystalline Form G is shown inFIG.4and the main peaks are listed in Table 2. TABLE 1PXRD peak list for crystalline Form A2θ angleNet IntensityRelative Intensity4.3153.60.085.68700.91.007.94247.90.358.73136.10.199.65176.00.2511.89168.30.2413.0591.40.1314.79118.90.1715.1761.70.0916.08171.50.244.3153.60.085.68700.91.007.94247.90.358.73136.10.199.65176.00.2516.96164.80.2417.82132.60.1918.21488.10.7019.02208.40.3020.45143.80.2121.23116.40.1716.96164.80.2422.42248.80.3522.8079.20.1123.79136.40.1925.61103.50.15 TABLE 2PXRD peak list for crystalline Form G2θ angleNet IntensityRelative Intensity3.621071.50.588.9296.80.0510.96184.50.1012.59206.50.1114.5381.60.0415.41252.10.1416.33133.30.0718.44294.90.1620.18701.10.3821.791860.21.0023.36205.80.1125.40502.90.2726.7834.50.0234.1828.80.02 Differential Scanning Calorimetry (DSC) and Thermogravimetric Analysis (TGA) Thermal properties of the compound were examined using a Discovery Differential Scanning Calorimeter (DSC) (TA Instruments) and a Discovery Thermogravimetric Analyzer (TGA) (TA Instruments). Sample was enclosed in a closed aluminum DSC pan for DSC analysis and in an open aluminum pan for TGA analysis. The thermal analysis was performed with a linear gradient from 25° C. to 300° C. at 10° C. per minute for both DSC and TGA studies. The differential scanning calorimetry (DSC) analysis for crystalline Form A shows that Form A has an onset temperature at 175.6° C. and a melting temperature at 186° C. (FIG.2). The DSC analysis for crystalline Form G shows that Form G has an onset temperature at 215.4° C. and a melting temperature at 217.3° C. (FIG.5). The TGA analysis for Form A of compound 27 shows a 3.16% weight loss, indicating Form A is a hydrate (FIG.3). The TGA analysis of Form G of compound 27 shows no weight loss until the melt, indicating that Form G is anhydrous (FIG.5). Solid State NMR The13C CP/MAS (Cross polarization/magic angle spinning) solid-state NMR spectra were acquired on a 363 MHz Tecmag Redstone spectrometer by Spectral Data Services of Champaign, IL Each sample was packed into a 7 mm (OD) zirconia rotor closed with kel-F end caps for subsequent data acquisition. All three samples were about half full in the rotor. The13C CP/MAS NMR spectra were acquired on the Doty XC 7 mm CP/MAS probe at an observing frequency of 91 MHz (spin 7 kHz, 1H pulse width 5.0 s, spectral width 29.8 kHz, acquisition time 0.0344 sec, CP pulse width 2 ms, relaxation delay 5.0 sec, number of scans 1296). Spectra were referenced to the chemical shift of external sample of glycine carbonyl carbon at 176 ppm and processed using Nuts (line broadening of 10 Hz). The peak listing and overlay of spectra are processed using MNOVA. 13C CP/MAS solid state NMR for Form A and Form G are shown inFIG.3BandFIG.6B, respectively. Diagnostic chemical shifts are listed in Table 3. TABLE 3Diagnostic13C CP/MAS NMR chemical shiftsfor solid forms of compound 27Peak assignment*Form A (ppm)Form G (ppm)carbonyl163.2163.6163.2aromatic carbons159.5159.4157.9147.0156.2 (shoulder)146.0143.7141.5142.5 (shoulder)140.6135.8137.1134.4136.0132.3130.2130.5125.9129.6125.0126.9120.7124.7105.9123.6104.4106.1105.1aliphatic carbons77.477.775.976.860.775.559.561.455.960.951.658.250.354.439.551.737.949.935.240.030.137.329.130.0*This is a tentative assignment based on the chemical structure and ChemDraw chemical shift prediction and solution13C NMR spectra in DMSO Example 28. (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 28) To a solution of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (200 mg, 0.4 mmol) in CH3CN (5 mL) was added 2,2,2-trifluoroethyl trifluoromethanesulfonate (190 mg, 0.6 mmol). The mixture was stirred at 50° C. for 12 h. After concentration of the reaction mixture, the residue was purified by silica gel chromatography (PE:EtOAc=1:1) to give (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2-(2,2,2-trifluoroethyl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (85 mg, yield: 36%). ESI-MS (M+H)+: 569.3.1H NMR (400 MHz, CD3OD) δ: 8.54 (s, 1H), 8.42 (d, J=5.2 Hz, 1H), 8.03-7.99 (m, 3H), 7.61 (s, 1H), 7.49 (d, J=7.6 Hz, 1H), 7.22 (d, J=5.2 Hz, 1H), 5.60-5.57 (m, 1H), 4.39-4.35 (m, 1H), 4.17-4.12 (m, 1H), 3.89 (s, 3H) 3.43-3.32 (m, 2H), 3.16-3.09 (m, 2H), 2.24-2.20 (m, 1H), 1.98-1.94 (m, 1H), 1.74 (s, 9H). Example 29. 1-(tert-butyl)-N—((R)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 29) To a solution of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (250 mg, 0.51 mmol) in EtOH (5 mL) were added (R)-2-methyloxirane (58 mg, 1.0 mmol). The mixture was stirred at 50° C. for 24 h. After concentration, the residue purified by silica gel column (petroleum ether/EtOAc=1:2) to give 1-(tert-butyl)-N—((R)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a white solid (110 mg, yield: 40%). ESI-MS (M+H)+: 545.3.1H NMR (400 MHz, CD3OD) δ: 8.42 (s, 1H), 8.28 (d, J=5.2 Hz, 1H), 7.93-7.84 (m, 3H), 7.52 (s, 1H), 7.35 (d, J=8.8 Hz, 1H), 7.09 (d, J=5.2 Hz, 1H), 5.46 (d, J=10 Hz, 1H), 4.13-4.00 (m, 2H), 3.92-3.87 (m, 1H), 3.78 (s, 3H) 3.21-3.12 (m, 2H), 2.40-2.32 (m, 2H), 2.17-2.13 (m, 1H), 1.89-1.84 (m, 1H), 1.62 (s, 9H), 1.02 (d, J=6.0 Hz, 3H). Example 30. 1-(tert-butyl)-N—((R)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 30) Synthesis of 1-(tert-butyl)-N—((R)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide was similar to that of 1-(tert-butyl)-N—((R)-2-((R)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Example 29). The crude was purified by prep-TLC (DCM/MeOH=10:1) to give 1-(tert-butyl)-N—((R)-2-((S)-2-hydroxypropyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (97 mg, yield: 44%). ESI-MS (M+H)+: 545.3.1H NMR (400 MHz, CD3OD) δ: 8.54 (s, 1H), 8.41 (d, J=5.2 Hz, 1H), 8.01-7.97 (m, 3H), 7.63 (s, 1H), 7.46 (d, J=8.0 Hz, 1H), 7.22 (d, J=5.6 Hz, 1H), 5.57 (d, J=9.6 Hz, 1H), 4.23-4.20 (m, 1H), 4.12-4.07 (m, 1H), 4.02-3.97 (m, 1H), 3.90 (s, 3H), 3.30-3.28 (m, 1H), 3.26-3.19 (m, 1H), 2.47-2.45 (m, 2H), 2.31-2.22 (m, 1H), 2.01-1.97 (m, 1H), 1.74 (s, 9H), 1.16 (d, J=6.4 Hz, 3H). Example 31. (R)-1-(tert-butyl)-N-(2-(2-methoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 31) To a solution of (R)-1-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide (140 mg, 0.29 mmol) in MeOH (10 mL) were added 1-bromo-2-methoxyethane (121 mg, 0.87 mmol) and K2CO3(120 mg, 0.87 mmol). The mixture was stirred at 80° C. for 16 h. After diluting with water (20 mL), the mixture was extracted with CH2Cl2(30 mL×2). The combined organic layers were washed with H2O (20 mL×2), dried (Na2SO4), filtered and concentrated. The crude material was purified by prep-TLC (DCM:MeOH=20:1) to give (R)-1-(tert-butyl)-N-(2-(2-methoxyethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (66 mg, yield: 42%). ESI-MS (M+H)+: 545.1.1H NMR (400 MHz, CDCl3) δ: 8.41 (d, J=5.2 Hz, 1H), 8.18 (s, 1H), 7.98 (d, J=8.4 Hz, 1H), 7.90 (s, 1H), 7.84 (d, J=8.0 Hz, 1H), 7.80 (s, 1H), 7.53 (s, 1H), 7.46 (d, J=8.0 Hz, 1H), 7.04 (d, J=5.2 Hz, 1H), 6.97 (s, 1H), 5.60 (t, J=8.8 Hz, 1H), 4.19-4.07 (m, 2H), 3.91 (s, 3H), 3.57 (t, J=5.6 Hz, 2H), 3.37 (s, 3H), 3.32-3.26 (m, 1H), 3.19-3.15 (m, 1H), 2.78-2.69 (m, 2H), 2.24-2.20 (m, 1H), 2.02-1.99 (m, 1H), 1.70 (s, 9H). Example 32. (R)-5-(tert-butyl)-N-(2-(2,2-difluoroethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 32) To a solution of (R)-5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (105 mg, 0.21 mmol) in CH3CN (8 mL) was added 1,1-difluoro-2-iodoethane (23 μL, 0.26 mmol) and potassium carbonate (89 mg, 0.64 mmol). The mixture was stirred at 80° C. for 18 h. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated and the crude product was purified by prep-HPLC (CH3CN/H2O with 0.05% TFA as mobile phase) to give (R)-5-(tert-butyl)-N-(2-(2,2-difluoroethyl)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (30 mg, yield: 25%). ESI-MS (M+H)+: 552.0.1H NMR (400 MHz, METHANOL-d4) δ: 8.42 (d, J=5.3 Hz, 1H), 8.23 (d, J=8.4 Hz, 1H), 8.19 (s, 1H), 7.95 (s, 1H), 7.68-7.65 (m, 1H), 7.62 (d, J=7.8 Hz, 1H), 7.30 (d, J=5.5 Hz, 1H), 6.40 (tt, J=53.5 Hz, 3.6 Hz, 1H), 5.70 (dd, J=9.8 Hz, 2.5 Hz, 1H), 4.83 (br d, J=14.3 Hz, 1H), 4.67 (br d, J=14.3 Hz, 1H), 3.90 (s, 3H), 3.83-3.67 (m, 2H), 3.59 (dt, J=15.0 Hz, 3.4 Hz, 2H), 2.52-2.31 (m, 2H), 1.49 (s, 9H) Example 33. 5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 33) 1. Synthesis of (E)-5-(3-bromophenyl)pent-4-enoic acid To a solution of (3-carboxypropyl)triphenylphosphonium bromide (12.87 g, 30 mmol, 1.0 equiv) in dry DMSO (50 mL) was added NaH (3 g, 75 mmol, 2.5 equiv) by portions at 0° C. The reaction was stirred at room temperature for 30 min before 3-bromobenzaldehyde (5.5 g, 30 mmol, 1.0 equiv) was dropwise added. The mixture was stirred at room temperature for another 2 h and then poured into water (200 mL) and extracted with EtOAc (100 mL). The aqueous solution was acidified with concentrated HCl and extracted with EtOAc (200 mL×3). The combined organic layer was washed with brine (100 mL×3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/EtOAc=2:1) to give (E)-5-(3-bromophenyl)pent-4-enoic acid (4.4 g, yield: 58%) as a yellow oil. ESI-MS (M+1)+: 254.9.1H NMR (400 MHz, CDCl3) δ: 7.48 (s, 1H), 7.33 (d, J=7.6 Hz, 1H), 7.23 (d, J=8.0 Hz, 1H), 7.15 (t, J=8.0 Hz, 1H), 6.39-6.35 (m, 1H), 6.23-6.19 (m, 1H), 2.55-2.53 (m, 4H). 2. Synthesis of 5-(3-bromophenyl)pentanoic acid To a solution of (E)-5-(3-bromophenyl)pent-4-enoic acid (2.4 g, 9.4 mmol, 1.0 equiv) in ethanol (20 mL) was added PtO2(200 mg, 10%). The mixture was stirred for 1 h under hydrogen atmosphere. The catalyst was filtered out and the resulting filtrate was concentrated to give target compound 5-(3-bromophenyl)pentanoic acid (2.1 g, yield: 87%) as a yellow solid, which was used to next step without further purification. ESI-MS (M+1)+: 256.9.1H NMR (400 MHz, CD3OD) δ: 7.24 (s, 1H), 7.21-7.18 (m, 1H), 7.06-7.03 (m, 2H), 2.50 (t, J=6.8 Hz, 2H), 2.20 (t, J=6.8 Hz, 2H), 1.53-1.51 (m, 4H). 3. Synthesis of 2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-one A mixture of 5-(3-bromophenyl)pentanoic acid (2.1 g, 8.2 mmol, 1.0 equiv) in PPA (5 ml) was stirred at 130° C. for 1 h. After cooling down, the mixture was basified to pH=7-8 with NaOH (1 N). The mixture was extracted with EtOAc (200 mL×2). The combined organic layers was concentrated and purified by prep-HPLC (Gradient: 5% B increase to 95% B, A: 0.5% NH3in water, B: CH3CN) to give 2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-one (1.1 g, yield: 56%) as a colorless oil. ESI-MS (M+H)+: 239.0.1H NMR (400 MHz, CDCl3) δ: 7.59 (d, J=8.4 Hz, 1H), 7.43 (dd, J=8.4, 2.0 Hz, 1H), 7.38 (s, 1H), 2.89 (t, J=6.8 Hz, 2H), 2.72 (t, J=6.0 Hz, 2H), 1.90-1.79 (m, 4H). 4. Synthesis of 2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-ol To a solution of 2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-one (600 mg, 2.5 mmol, 1.0 equiv) in MeOH (10 mL) was added NaBH4(144 mg, 3.8 mmol, 1.5 equiv) and then stirred at room temperature for 1 h. After evaporation of the solvent, the residue was purified by silica gel column (EtOAc/hexane=1:5) to give 2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-ol (600 mg, yield: 99%) as a white solid. ESI-MS (M+H−17)+: 222.9.1H NMR (400 MHz, CDCl3) δ: 7.34-7.30 (m, 2H), 7.24 (s, 1H), 4.88-4.86 (m, 1H), 2.88-8.82 (m, 1H), 2.70-2.63 (m, 1H), 2.08-2.00 (m, 2H), 1.81-1.72 (m, 4H). 5. Synthesis of 5-azido-2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulene A solution of 2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-ol (600 mg, 2.5 mmol, 1.0 equiv) in toluene (10 mL) was cooled in an ice bath under N2and treated with DPPA (2.06 g, 7.5 mmol, 3.0 equiv) in one portion followed by DBU (1.14 g, 7.5 mmol, 3.0 equiv). The reaction temperature was kept at 0° C. for 1 h and then was warmed to room temperature for 12 h. The mixture was diluted with EtOAc (100 mL), washed with 2N HCl (2×50 mL), brine and the organic layer was dried over Na2SO4, filtered then concentrated. The crude product was purified by silica gel column (eluted with PE) to give 5-azido-2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulene (350 mg, yield: 45%) as a yellow oil. ESI-MS (M+H-N3)+: 223.0.1H NMR (400 MHz, CDCl3) δ: 7.31-7.29 (m, 2H), 7.15 (d, J=8.0 Hz, 1H), 4.72 (t, J=5.2 Hz, 1H), 2.99-2.92 (m, 1H), 2.70-2.64 (m, 1H), 2.08-2.00 (m, 1H), 1.90-1.59 (m, 5H). 6. Synthesis of 2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-amine To a mixture of 5-azido-2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulene (375 mg, 1.4 mmol, 1.0 equiv) in THF (5 mL) and H2O (0.5 mL) was added PPh3(741 mg, 2.8 mmol, 2.0 equiv). The mixture was stirred at room temperature for 12 h. The mixture was acidified to pH=1 with HCl (1 N) and extracted with EtOAc (100 mL). The separated aqueous layer was basified to pH=10 with NaOH (1 N). The resulting precipitate was collected and dried to give 2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-amine (360 mg, yield: 100%) as a white solid. ESI-MS (M+H−17)+: 222.9. 7. Synthesis of tert-butyl (2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate To a mixture of tert-butyl (2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (360 mg, 1.5 mmol, 1.0 equiv) in CH2Cl2(5 mL) and triethylamine (303 mg, 3.0 mmol, 2.0 equiv) was added Boc2O (394 mg, 1.8 mmol, 1.2 equiv). The mixture was stirred at room temperature for 2 h. After diluted with EtOAc (100 mL), the mixture was washed with water (100 mL×2). The organic layer was concentrated and purified by silica gel column (PE:EtOAc=30:1) to give tert-butyl (2-bromo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (310 mg, yield: 61%) as a white solid. ESI-MS (M−55): 284.0.1H NMR (400 MHz, CDCl3) δ: 7.29-7.23 (m, 2H), 7.10 (d, J=8.0 Hz, 1H), 4.92-4.82 (m, 2H), 2.84-2.75 (m, 2H), 1.88-1.83 (m, 5H), 1.44 (s, 9H). 8. Synthesis of tert-butyl (2-(2-chloropyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate Synthesis of tert-butyl (2-(2-chloropyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate was similar to that of tert-butyl 4-(2-chloropyrimidin-4-yl)-2-methylbenzylcarbamate. The mixture was concentrated and purified by silica gel column (PE:EtOAc=4:1) to give tert-butyl (2-(2-chloropyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (200 mg, yield: 66%) as a white solid. ESI-MS (M+H)+: 374.1. 9. Synthesis of tert-butyl (2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate A mixture of tert-butyl (2-(2-chloropyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (500 mg, 1.34 mmol), 1-methyl-1H-pyrazol-4-amine (195 mg, 2.01 mmol), Pd2(dba)3(120 mg, 0.13 mmol), S-phos (111 mg, 0.27 mmol) and Cs2CO3(870 mg, 2.68 mmol) in 1,4-dioxane (12 ml) was stirred at 100° C. for 6 h under N2. The mixture was filtrated through a Celite pad and the filtrate was concentrated. The residue was purified by silica-gel-column (EtOAc) to give tert-butyl (2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate as a white solid (480 mg, yield: 83%). ESI-MS (M+H)+: 435.3.1H NMR (400 MHz, CDCl3) δ: 8.46 (d, J=5.2 Hz, 1H), 7.88-7.80 (m, 3H), 7.38-6.85 (m, 4H), 4.97-4.94 (m, 2H), 3.85 (s, 3H), 2.96-2.94 (m, 2H), 1.93-1.64 (m, 6H), 1.47 (s, 9H). 10. Synthesis of 4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine A mixture of tert-butyl (2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (240 mg, 0.55 mmol) in TFA (4.0 mL) and CH2Cl2(4.0 mL) was stirred at rt for 2 h. Then the reaction mixture was concentrated to give compound 4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (250 mg, crude) as a yellow oil, which was used in the next step without further purification. ESI-MS (M+H)+: 335.3. 11. Synthesis of 5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide Synthesis of 5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide was similar to that of tert-butyl 8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate in Example 2. The residue was purified by prep-HPLC (CH3CN/H2O with 0.05% NH4HCO3as mobile phase) to give 5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (120 mg, yield: 42%). ESI-MS (M+H)+: 487.3.1H NMR (400 MHz, CD3OD) δ: 8.43 (d, J=5.2 Hz, 1H), 7.98-7.96 (m, 2H), 7.51-7.30 (m, 3H), 6.74 (d, J=2.4 Hz, 1H), 5.44 (d, J=10.0 Hz, 1H), 3.83 (s, 3H), 3.09-3.03 (m, 2H), 2.10-1.43 (m, 6H), 1.54 (s, 9H). Example 34. (R)—N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide (Compound 34) 1. Synthesis of (R,Z)-5-((1-phenylethyl)imino)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-ol A slurry mixture of 2-hydroxy-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-one (Herdman C. A. et al., Structural interrogation of benzosuberen-based inhibitors of tubulin polymerization, Bioorganic & Medical Chemistry, 23(24), 7497-7520, 2015) (10 g, 57 mmol), (R)-(+)-α-methylbenzylamine (9.0 g, 74 mmol), and p-toluenesulfonic acid (0.48 g, 2.84 mmol) in 150 mL toluene was heated at reflux with a Dean Stark apparatus. After 16 hr, the Dean Stark apparatus was removed and reflux was continued until ˜80 mL toluene was distilled, during which solid crystallized. The mixture was cooled to 0° C., kept at 0° C. for 2 hr, and then filtered to give (R,Z)-5-((1-phenylethyl)imino)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-ol as a tan solid (after drying—13.5 g, yield: 85%). The crude product was used for next step without further purification. 2. Synthesis of (R)-5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-ol acetate A slurry of (R,Z)-5-((1-phenylethyl)imino)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-ol (13.5 g, 48 mmol) in 10:1 EtOAc/MeOH (148 mL) was cooled to −5° C. Solid NaBH4(5.45 g, 144 mmol) was added in portions, while maintaining the temperature under 5° C. The mixture was stirred at −5° C. for 40 min. After 1 hr, water (135 mL) was added, followed by 5N HCl until pH ˜6. The layers were separated, and the aqueous layer was extracted with 135 mL EtOAc. The combined organics were washed with brine (100 mL) and dried (Na2SO4) and filtered to give a solution of (R)-5-(((R)-1-phenylethyl)amino)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-ol which was carried forward. To the solution of (R)-5-(((R)-1-phenylethyl)amino)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-ol (48 mmol) in EtOAc (˜185 mL) was added acetic acid (5.8 g, 96 mmol). The solution was then concentrated and carried forward. MeOH (200 mL) was added, followed by 10% Pd/C (1.4 g, 10% wt %). The mixture was stirred under an atmosphere of H2(50 psi) at 55° C. for 16 hr. The mixture was filtered and the filtrate was concentrated. Methyl tert-butyl ether (100 mL) was added, followed by petroleum ether (100 mL). The mixture was stirred at 25° C. for 2 hr, and filtered. After drying at 50° C., (R)-5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-ol acetate was isolated (9.9 g, yield: 87% for three steps. 3. Synthesis of tert-butyl (R)-(2-hydroxy-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate To a solution of (R)-5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-ol acetate (9.9 g, 42 mmol) in dioxane (50 mL) was added aqueous NaHCO3(1N, 100 mL, 100 mmol) followed by Boc2O (13.7 g, 63 mmol. The mixture was stirred at rt overnight. EtOAc (150 mL) was added and the layers were separated. The aqueous layer was extracted with EtOAc (150 mL). The combined organic layers were dried (Na2SO4), filtered, and concentrated. MTBE (150 mL) was added, followed by petroleum ether (150 mL). After stirring for 2 hr, the mixture was filtered and the filtrate was concentrated to give an oil. The crude material was purified by silica gel chromatography to give tert-butyl (R)-(2-hydroxy-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate as a white solid (7.8 g, yield: 67%, ee: 98.5%).1H NMR (400 MHz, DMSO-d6) δ: 9.06 (s, 1H), 7.29-7.27 (m, 1H), 6.94 (d, J=8.8 Hz, 1H), 6.49 (s, 2H), 4.48 (m, 1H), 2.65 (br s, 2H), 1.99-1.64 (m, 4H), 1.48-1.25 (m, 11H). 4. Synthesis of (R)-5-((tert-butoxycarbonyl)amino)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl trifluoromethanesulfonate To a solution of tert-butyl (R)-(2-hydroxy-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (1.0 g, 3.6 mmol) and pyridine (570 mg, 7.2 mmol) in DCM (30 mL) was added Tf2O (1.5 g, 5.4 mmol) at 0° C. The mixture was stirred at 0° C. for 1 h. After diluting with water (20 mL), the mixture was extracted with DCM (3×40 mL). The combined organic layers were washed by aq. NaHSO4 (0.5 N, 20 mL) to adjust water layer to pH=5-6, dried (Na2SO4), filtered and concentrated. The crude product was used for next step without further purification. ESI-MS (M+H)+: 410.1.1H NMR (400 MHz, CDCl3) δ: 7.31 (d, J=8.8 Hz, 1H), 7.07-7.00 (m, 2H), 4.93-4.88 (m, 2H), 2.89-2.82 (m, 2H), 1.91-1.68 (m, 6H), 1.45 (s, 9H). 5. Synthesis of tert-butyl (R)-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate Synthesis of tert-butyl (R)-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate was similar to that of tert-butyl 5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[c]azepine-2(3H)-carboxylate (Example 2, Step 11). The crude product was purified by silica gel column chromatograph (DCM/MeOH=20:1) to give tert-butyl (R)-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate as a yellow solid (420 mg, yield: 68%). ESI-MS (M+H)+: 435.2.1H NMR (400 MHz, CDCl3) δ: 8.41-8.37 (m, 1H), 7.88-7.77 (m, 3H), 7.55-7.36 (m, 4H), 7.05 (d, J=5.2 Hz, 1H), 5.09-5.01 (m, 1H), 3.90 (s, 3H), 2.93-2.91 (m, 2H), 2.18-2.17 (m, 1H), 1.91-1.82 (m, 5H), 1.28-1.24 (m, 9H). 6. Synthesis of (R)-4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine Synthesis of (R)-4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine was similar to 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide described in Example 2, Step 12. The crude product was used for next step without further purification. ESI-MS (M+H)+: 335.2. 7. Synthesis of ethyl (Z)-2-amino-2-(((1-methylcyclopropane-1-carbonyl)oxy)imino)acetate To a solution of 1-methylcyclopropane-1-carboxylic acid (2 g, 20 mmol) in DCM (50 mL) was added (COCl)2(5 g, 40 mmol). The mixture was stirred at rt for 16 h and then concentrated to give the intermediate acyl chloride. To a solution of ethyl (Z)-2-amino-2-(hydroxyimino)acetate (2.6 g, 20 mmol) and triethylamine (6 g, 60 mmol) in DCM (20 mL) was added a solution of the intermediate acyl chloride in DCM (20 mL). The reaction was stirred at rt for 2 h, washed by water, dried by Na2SO4, concentrated to give ethyl (Z)-2-amino-2-(((1-methylcyclopropane-1-carbonyl)oxy)imino)acetate as a white solid (3.0 g, yield: 63%). ESI-MS (M+H)+: 215.1. 8. Synthesis of ethyl 5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxylate A solution of ethyl (Z)-2-amino-2-(((1-methylcyclopropane-1-carbonyl)oxy)imino)acetate (3.0 g, 14 mmol) in AcOH (20 mL) was stirred at 100° C. for 2 h and then concentrated. The residue was diluted with DCM (20 mL), washed by saturated NaHCO3solution (3×10 mL), dried over Na2SO4, and concentrated. The residue was purified by silica gel column (petroleum ether:EtOAc=10:1) to give ethyl 5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxylate as a yellow solid (1.1 g, yield: 52%). ESI-MS (M+H)+: 197.1. 9. Synthesis of potassium 5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxylate Ethyl 5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxylate (1.0 g, 5.0 mmol) and KOH (308 mg, 5.5 mmol) was dissolved in EtOH/H2O (4:1, 20 mL). The reaction was stirred at rt for 12 h and then concentrated to give potassium 5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxylate as a yellow solid (1.1 g, yield: 95%). ESI-MS (M+H)+: 169.1. 10. Synthesis of N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide (I-IM_6) To a solution of potassium 5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxylate (123 mg, 0.6 mmol), DIPEA (232 mg, 0.75 mmol) and HATU (342 mg, 0.9 mmol) in DCM (5 mL) was added (R)-4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (150 mg, 0.50 mmol). The mixture was stirred at rt for 1 h. After diluted with water (60 mL), the mixture was extracted with DCM (80 mL×2). The combined organic layers were dried and concentrated. The crude product was purified by prep-HPLC to give (R)—N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide as a white solid (60 mg, yield: 30%). ESI-MS (M+H)+: 485.2.1H NMR (400 MHz, CD3OD) δ: 8.40 (d, J=5.2 Hz, 1H), 7.97 (s, 1H), 7.98-7.93 (m, 2H), 7.66 (s, 1H), 7.39 (d, J=8.4 Hz, 1H), 7.21 (d, J=5.2 Hz, 1H), 5.43 (d, J=10 Hz, 1H), 3.90 (s, 3H), 3.09-3.02 (m, 2H), 2.09-1.86 (m, 5H), 1.64 (s, 3H), 1.54-1.45 (m, 3H), 1.21-1.18 (m, 2H). Example 35. (R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 35) 1. Synthesis of tert-butyl (R)-(2-(2-chloropyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate Synthesis of tert-butyl (R)-(2-(2-chloropyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate was similar to that of tert-butyl 1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate (Example 1, Step 12). The crude was purified by silica gel column chromatography (MeOH/EtOAc=1/50) to give tert-butyl (R)-(2-(2-chloropyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate as a yellow solid (350 mg, yield: 44%). ESI-MS (M+H)+: 373.2. 2. Synthesis of tert-butyl (R)-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate To a solution of tert-butyl (R)-(2-(2-chloropyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (400 mg, 1.1 mmol) in dioxane (20 mL) were added 1-methyl-1H-pyrazol-4-amine (210 mg, 2.1 mmol), Pd2(dba)3(100 mg, 0.11 mmol), SPhos (45 mg, 0.11 mmol) and Cs2CO3(680 mg, 2.1 mmol). The mixture was heated to 100° C. for 2 h and concentrated. The crude was purified by silica gel column chromatography (MeOH/EtOAc=1/50) to give tert-butyl (R)-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (330 mg, yield: 71%) as a yellow solid. ESI-MS (M+H)+: 434.2. 3. Synthesis of (R)-4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyridin-2-amine Synthesis of (R)-4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyridin-2-amine was similar to that of 5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,2,4-oxadiazole-3-carboxamide (Example 1, Step 13). The crude product (R)-4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyridin-2-amine was obtained as yellow solid (200 mg, yield: 79%). ESI-MS (M+H)+: 334.2. 4. Synthesis of (R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide (I-IM_65) Synthesis of (R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide was similar to N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-5-(1-methylcyclopropyl)-1,2,4-oxadiazole-3-carboxamide described in Example 34, Step 10. The mixture was purified by prep-HPLC (CH3CN/H2O with 0.05% NH3·H2O as mobile phase) to give (R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-3-carboxamide as yellow solid (100 mg, yield: 88%). ESI-MS (M+H)+: 486.3.1H NMR (400 MHz, CDCl3) δ: 8.17-8.16 (m, 1H), 7.61 (s, 1H), 7.48 (s, 1H), 7.43-7.41 (m, 1H), 7.36-7.30 (m, 3H), 6.87-6.86 (m, 1H), 6.71 (s, 1H), 6.22-6.18 (m, 1H), 5.44-5.40 (m, 1H), 3.91 (s, 3H), 3.01-2.89 (m, 2H), 2.05-1.93 (m, 4H), 1.82-1.78 (m, 2H), 1.49 (s, 9H). Example 36. (R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 36a) 1. Synthesis of (R)-1-(tert-butyl)-N-(8-(2-chloropyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide Synthesis of (R)-1-(tert-butyl)-N-(8-(2-chloropyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1H-1,2,3-triazole-4-carboxamide was similar to that of (R)-5-(tert-butyl)-N-(8-(2-((1,5-dimethyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-5-yl)-1,2,4-oxadiazole-3-carboxamide (Example 9, Step 4). The crude product was used for next step without purification. ESI-MS (M+H)+: 427.2. 2. Synthesis of (R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 36a) and (S)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 36b) 3-tert-Butyl-1,2,4-oxadiazole-5-carboxylic acid {2-[2-(1-methyl-1H-pyrazol-4-ylamino)-pyrimidin-4-yl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-yl}-amide (76 mg) was subjected to SFC separation (IA 2×(2×15 cm), 30% ethanol/CO2, 100 bar, 70 mL/min, 220 nm, inj vol.: 1 mL, 5 mg/mL, ethanol) and yielded 43 mg of peak-1 (chemical purity 99%, ee >99%) and 36 mg of peak-2 (chemical purity 99%, ee >99%). Peak 1 is assigned as (R)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide: LCMS: Rt 1.23 min, m/z 487.00.1H NMR (400 MHz, METHANOL-d4) δ 8.41 (d, J=5.27 Hz, 1H), 7.96 (d, J=11.80 Hz, 3H), 7.66 (s, 1H), 7.43 (d, J=8.53 Hz, 1H), 7.22 (d, J=5.27 Hz, 1H), 5.43 (d, J=9.29 Hz, 1H), 3.90 (s, 3H), 2.96-3.24 (m, 2H), 1.74-2.25 (m, 5H), 1.51 (s, 9H), 1.28-1.43 (m, 1H). Peak 2 is assigned as (S)-5-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,3,4-oxadiazole-2-carboxamide: LCMS: Rt 1.23 min, m/z 487.00.1H NMR (400 MHz, METHANOL-d4) δ 8.40 (d, J=5.27 Hz, 1H), 7.96 (d, J=12.05 Hz, 3H), 7.66 (s, 1H), 7.43 (d, J=8.53 Hz, 1H), 7.22 (d, J=5.27 Hz, 1H), 5.44 (s, 1H), 3.90 (s, 3H), 2.93-3.21 (m, 2H), 1.78-2.23 (m, 5H), 1.51 (s, 9H), 1.31 (s, 1H). Example 37. 3-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-5-carboxamide (Compound 37) To a solution of 3-tert-butyl-1,2,4-oxadiazole-5-carboxylic acid (1.00 mg, 0.60 mmol) and N,N,N′,N′-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (273 mg, 0.72 mmol) and 4-(5-amino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (200 mg, 0.6 mmol) in N,N-dimethylformamide (2.3 mL) was added N,N-diisopropylethylamine (0.42 mL, 2.4 mmol). The reaction was stirred at rt overnight and was quenched with MeOH. After workup, prep HPLC gave 3-(tert-butyl)-N-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)-1,2,4-oxadiazole-5-carboxamide as a solid (186 mg; yield: 64%). LCMS: Rt 1.38 min.; m/z 487.1;1H NMR (400 MHz, METHANOL-d4) δ: 8.29 (br. s., 1H), 7.97-8.09 (m, 2H), 7.94 (s, 1H), 7.68 (br. s., 1H), 7.43 (d, J=8.78 Hz, 2H), 5.41 (d, J=9.79 Hz, 1H), 3.92 (s, 3H), 2.87-3.18 (m, 2H), 1.70-2.29 (m, 5H), 1.45 (s, 10H). Example 38: 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 38) 1. Synthesis of methyl 4-(3-bromophenoxy)butanoate To a solution of 3-bromophenol (3.44 g, 20.0 mmol) and methyl 4-bromobutanoate (4.32 g, 24.0 mmol) in DMF (20 mL) was added K2CO3(5.52 g, 40.0 mmol). The mixture was stirred at rt for 0.5 h and then heated with stirring at 90° C. for 1 h. After diluting with EtOAc (200 mL), the mixture was washed with water (3×50 mL), dried and concentrated. The crude product was purified by silica gel column chromatograph (petroleum/EtOAc=10:1) to give methyl 4-(3-bromophenoxy)butanoate as a white liquid (5.2 g, yield: 96%). ESI-MS (M+H)+: 273.1. 2. Synthesis of 4-(3-bromophenoxy)butanoic acid To a solution of methyl 4-(3-bromophenoxy)butanoate (4.9 g, 19 mmol) in MeOH (40 mL) and H2O (40 mL) was added NaOH (2.3 g, 57 mmol). The reaction mixture was stirred at 65° C. for 1 h. Then the solvent was concentrated under reduced pressure. The residue was adjusted to pH=3 with HCl (1 N). The mixture was extracted with EtOAc (3×100 mL×3). The organic layers were dried over sodium sulfate and concentrated under reduced pressure to give 4-(3-bromophenoxy)butanoic acid (4.8 g, yield: 98%). The crude product was used in next step without further purification. ESI-MS (M+H)+: 259.1.1H NMR (400 MHz, CDCl3) δ: 7.24-7.20 (m, 1H), 7.10-7.08 (m, 2H), 6.93 (d, J=9.6 Hz, 1H), 3.97 (t, J=6.4 Hz, 2H), 2.35 (t, J=7.2 Hz, 2H), 1.92-1.88 (m, 2H). 3. Synthesis of 8-bromo-3,4-dihydrobenzo[b]oxepin-5(2H)-one To a solution of 4-(3-bromophenoxy)butanoic acid (1.82 g, 7.04 mmol) in DCM (35 mL) was added SOCl2(1.67 g, 14 mmol) and DMF (cat.). The reaction mixture was stirred at 40° C. for 1 h. Then the solvent was removed under reduced pressure, dried in vacuo for 2 h. The residue was dissolved in DCM (35 mL) and cooling down with an ice bath. AlCl3(1.02 g, 80 mmol) was added and the mixture was stirred at 0° C.—rt for 12 h. The mixture was poured into con. HCl (10 mL) and extracted with EtOAc (2×50 mL). The organic layers were washed with water (50 mL), brine (50 mL) and dried over sodium sulfate. After concentration under reduced pressure, the crude product was purified by silica gel column chromatograph (petroleum ether/EtOAc=4:1) to give 8-bromo-3,4-dihydrobenzo[b]oxepin-5(2H)-one as a white solid (1.2 g, yield: 71%). ESI-MS (M+H)+: 241.1.1H NMR (400 MHz, CDCl3) δ: 7.64 (d, J=8.8 Hz, 1H), 7.27-7.23 (m, 2H), 4.25 (t, J=6.8 Hz, 2H), 2.89 (t, J=7.2 Hz, 2H), 2.25-2.18 (m, 2H). 4. Synthesis of 8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-amine To a mixture of 8-bromo-3,4-dihydrobenzo[b]oxepin-5(2H)-one (1.2 g, 5.0 mmol) in i-PrOH (50 mL), NH4OAc (7.63 g, 100 mmol) and NaBH3CN (3.15 g, 50 mol) was added. The mixture was stirred at 80° C. for 4 h. After cooling down, the mixture was basified to pH >12 with NaOH (1 N). The mixture was extracted with DCM (250 mL×2). The combined organic layers were dried and concentrated. The resulting residue was purified by silica gel column (DCM:MeOH=20:1) to give 8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-amine as a white solid (900 mg, yield: 75%) was obtained. ESI-MS (M+H—NH3)+: 225.1. 5. Synthesis of N-(8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide To a solution of 8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-amine (500 mg, 2.23 mmol, 1.0 eq) in DMF (20 mL), potassium 5-tert-butyl-1,3,4-oxadiazole-2-carboxylate (557 mg, 2.68 mmol, 1.2 eq), HATU (1.3 g, 3.35 mmol, 1.5 eq) and triethylamine (863 mg, 6.69 mmol, 3.0 eq) were added. The mixture was stirred at rt for 2 h. The solution was diluted with EtOAc (150 mL) and washed with water (50 mL), brine (2×50 mL). The organic layer was dried (Na2SO4), filtered, and concentrated via rotary evaporator. The residue was purified by reverse phase chromatography (CH3CN/H2O with 0.05% NH3·H2O as mobile phase) to give N-(8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide (210 mg, yield: 30%) as a slight yellow solid. ESI-MS (M+H)+: 394.1. 6. The Preparation of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,3,4-oxadiazole-2-carboxamide To a mixture of N-(8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-5-(tert-butyl)-1,3,4-oxadiazole-2-carboxamide (393 mg, 1.00 mmol) and PinB-BPin (263 mg, 1.1 mmol) in dry 1,4-dioxane (10 mL), KOAc (196 mg, 2.0 mmol), Pd(dppf)Cl2·DCM (81 mg, 0.1 mmol) was added under N2. The mixture was stirred at 100° C. for 2 h under N2. After cooling down, 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (209 mg, 1.0 mmol), K2CO3(276 mg, 2.0 mmol) and H2O (2.5 mL) was added. The mixture was stirred at 100° C. for 2 h under N2. After cooling down, the mixture was concentrated and purified silica gel column chromatograph (DCM/MeOH=20:1) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,3,4-oxadiazole-2-carboxamide as yellow solid (50 mg, yield: 21%). ESI-MS (M+H)+: 489.2.1H NMR (400 MHz, CD3OD) δ:8.26 (d, J=5.2 Hz, 1H), 7.82 (s, 1H), 7.69-7.64 (m, 2H), 7.51 (s, 1H), 7.29 (d, J=8.0 Hz, 1H), 7.02 (d, J=5.2 Hz, 1H), 5.35-5.32 (m, 1H), 4.14-4.10 (m, 1H), 3.83-3.80 (m, 1H), 3.76 (s, 3H), 2.00-1.98 (m, 4H), 1.36 (s, 9H). Example 39. Synthesis of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,2,4-oxadiazole-2-carboxamide (Compound 39) 1. Synthesis of N-(8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-5-(tert-butyl)-1,2,4-oxadiazole-2-carboxamide Synthesis of N-(8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamide was similar to that of tert-butyl (R)-8-bromo-5-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-1,3,4,5-tetrahydro-2H-benzo[c]azepine-2-carboxylate in Example 2, Method 2, Step 2. The crude material was purified by silica gel chromatography (PE:EtOAc=3:1) to give N-(8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-5-(tert-butyl)-1,2,4-oxadiazole-2-carboxamide (610 mg, yield: 85%) as a slight yellow solid. ESI-MS (M+H)+: 394.1. 2. The Preparation of 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,2,4-oxadiazole-2-carboxamide To a mixture of N-(8-bromo-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-5-(tert-butyl)-1,2,4-oxadiazole-2-carboxamide (135 mg, 0.34 mmol) and PinB-BPin (96 mg, 0.38 mmol) in dry 1,4-dioxane (3 mL), KOAc (68 mg, 0.69 mmol), Pd(dppf)Cl2·DCM (24 mg, 0.03 mmol) was added under N2. The mixture was stirred at 100° C. for 2 h under N2. After cooling down, 4-chloro-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (71 mg, 0.34 mmol), K2CO3(95 mg, 0.69 mmol) and H2O (0.8 mL) was added. The mixture was stirred at 100° C. for 2 h under N2. After cooling down, the mixture was concentrated and purified by silica gel chromatography (DCM:MeOH=20:1) to give 5-(tert-butyl)-N-(8-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydrobenzo[b]oxepin-5-yl)-1,2,4-oxadiazole-2-carboxamide as yellow solid (77 mg, yield: 46%). ESI-MS (M+H)+: 489.2.1H NMR (400 MHz, CD3OD) δ: 8.35 (d, J=5.2 Hz, 1H), 7.91 (s, 1H), 7.78-7.73 (m, 2H), 7.62 (s, 1H), 7.38 (d, J=8.4 Hz, 1H), 7.10 (d, J=5.6 Hz, 1H), 5.46-5.44 (m, 1H), 4.18-4.15 (m, 1H), 3.98-3.95 (m, 1H), 3.86 (s, 3H), 2.10-2.02 (m, 4H), 1.48 (s, 9H). Example 40. 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 40) 1. Synthesis of 3-chlorocyclohept-2-en-1-one To a solution of cycloheptane-1,3-dione (20.0 g, 0.16 mol) and DMF (11.6 g, 0.16 mol) in DCM (500 mL) was added oxalyl chloride (24.4 g, 0.19 mol) dropwise at 0° C. After stirring at 0° C. for 30 min, the reaction mixture was washed with water (3×500 mL). The aqueous phase was then extracted with diethyl ether (4×300 mL). The combined DCM and diethyl ether phases were dried over MgSO4and concentrated to yield 3-chlorocyclohept-2-en-1-one (crude 22.8 g, used for next step) as a brown oil. ESI-MS (M+H)+: 145.1. 2. Synthesis of 2-cyano-2-(3-oxocyclohept-1-en-1-yl)acetamide To a solution of 2-cyanoacetamide (26.9 g, 0.32 mol) in DMF (300 mL) was added NaH (60 percent in mineral oil, 14.1 g, 0.35 mol) in one portion at 0° C. After stirring at 0° C. for 30 min, a solution of 3-chlorocyclohept-2-en-1-one (22.8 g, 0.16 mol) in DMF (100 mL) was added dropwise. The reaction mixture was stirred at room temperature for 30 min and DMF was removed under reduced pressure. The residue was dissolved in water (350 mL). The solution was washed with ethyl acetate (4×150 mL), adjusted with 3.0 N aqueous HCl to pH 2-3 and extracted with 10% MeOH/DCM (6×300 mL). The latter combined extracts were dried over MgSO4and concentrated. The crude product was purified by silica gel column chromatography (EtOAc/petroleum ether=4:1) to give 2-cyano-2-(3-oxocyclohept-1-en-1-yl)acetamide as yellow oil (22.0 g, yield: 73% for two steps). ESI-MS (M+H)+: 193.1. 3. Synthesis of 3,9-dioxo-3,5,6,7,8,9-hexahydro-2H-cyclohepta[c]pyridine-4-carbonitrile To a solution of 2-cyano-2-(3-oxocyclohept-1-en-1-yl)acetamide (22.0 g, 0.11 mol) in DMF (150 mL) was added DMF-DMA (22.8 mL, 0.17 mol) dropwise over 0.5 h. The reaction mixture was stirred at 50° C. for 4 h and concentrated under reduced pressure. The resulting brown oil was dissolved in aqueous NaOH (1.0 N, 200 mL), washed with chloroform (5×150 mL) and acidified with HCl (6.0 N) slowly at 0° C. to pH 2-3. The brown solid, 3,9-dioxo-3,5,6,7,8,9-hexahydro-2H-cyclohepta[c]pyridine-4-carbonitrile (17.0 g, yield: 74%), was collected by filtration and dried in vacuo. ESI-MS (M+H)+: 203.1.1H NMR (400 MHz, CDCl3) δ: 8.16 (s, 1H), 3.17-3.14 (m, 2H), 2.77-2.74 (m, 2H), 2.04-2.00 (m, 2H), 1.90-1.87 (m, 2H). 4. Synthesis of 5,6,7,8-tetrahydro-2H-cyclohepta[c]pyridine-3,9-dione A solution of 3,9-dioxo-3,5,6,7,8,9-hexahydro-2H-cyclohepta[c]pyridine-4-carbonitrile (17.0 g, 0.084 mol) in 50 percent conc. sulfuric acid (100 mL) was stirred at 140° C. for 12 h. The reaction mixture was neutralized with 50 percent sodium hydroxide slowly at 0° C. to pH 7-8. The water was removed under reduced pressure. The residue was dissolved into warm chloroform and an insoluble solid was removed by filtration. The filtrate was concentrated and purified by silica gel column chromatograph (DCM/MeOH=20:1) to give 5,6,7,8-tetrahydro-2H-cyclohepta[c]pyridine-3,9-dione as a yellow solid (9.5 g, yield: 63%). ESI-MS (M+H)+: 178.1. 5. Synthesis of 9-amino-2,5,6,7,8,9-hexahydro-3H-cyclohepta[c]pyridin-3-one A mixture of 5,6,7,8-tetrahydro-2H-cyclohepta[c]pyridine-3,9-dione (7.0 g, 39.5 mmol), NH4OAc (60.8 g, 790.0 mmol), and NaBH3CN (7.4 g, 118.5 mmol) in i-PrOH (150 mL) was heated to 80° C. for 8 h and cooled to rt. The solution was used for next step without purification. ESI-MS (M+H)+: 179.2. 6. Synthesis of tert-butyl (3-oxo-3,5,6,7,8,9-hexahydro-2H-cyclohepta[c]pyridin-9-yl)carbamate To the previous solution was added NaHCO3(aq, 50 mL), THE (50 mL) and Boc2O (17.2 g, 79.0 mmol). The mixture was stirred at rt for 6 h. After concentration and diluting with water (100 mL), the mixture was extracted with DCM (3×200 mL). The combined organic layers were washed with brine (200 mL), dried (Na2SO4), filtered and concentrated. The crude product was purified by silica gel column chromatograph (DCM/MeOH=20:1) to give tert-butyl (3-oxo-3,5,6,7,8,9-hexahydro-2H-cyclohepta[c]pyridin-9-yl)carbamate as yellow solid (6.2 g, yield: 56% for two steps). ESI-MS (M+H)+: 279.2.1H NMR (400 MHz, CDCl3) δ: 7.25 (s, 1H), 6.35 (s, 1H), 5.30 (brs, 1H), 2.65-2.63 (m, 2H), 1.86-1.76 (m, 4H), 1.45 (s, 9H), 1.39-1.35 (m, 2H). 7. Synthesis of 9-((tert-butoxycarbonyl)amino)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-3-yl trifluoromethanesulfonate To a solution of tert-butyl (3-oxo-3,5,6,7,8,9-hexahydro-2H-cyclohepta[c]pyridin-9-yl)carbamate (6.2 g, 22.3 mmol) and triethylamine (6.8 g, 66.9 mmol) in DCM (150 mL) at 0° C. was added Tf2O (9.4 g, 33.5 mmol) dropwise. The mixture was stirred for 1 h. The solution was diluted with water (150 mL), extracted with DCM (2×200 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated. The crude product was purified by silica gel column chromatograph (petroleum ether/EtOAc=4:1) to give 9-((tert-butoxycarbonyl)amino)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-3-yl trifluoromethanesulfonate as a yellow solid (5.7 g, Y: 63%). ESI-MS (M+H)+: 411.1. 8. Synthesis of tert-butyl (3-(1-ethoxyvinyl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate A mixture of 9-((tert-butoxycarbonyl)amino)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-3-yl trifluoromethanesulfonate (2.4 g, 5.85 mmol), tributyl(1-ethoxyvinyl)stannane (4.3 g, 11.7 mmol), triethylamine (1.8 g, 17.6 mmol), Pd(OAc)2(131 mg, 0.58 mmol) and dppp (903 mg, 2.34 mmol) in 50 mL DMF was stirred at 70° C. for 2 h under nitrogen in sealed tube. The mixture was diluted with EtOAc (200 mL) and washed with water (3×100 mL). The organic phase was dried with Na2SO4and concentrated. The crude product was purified by silica gel column chromatograph (petroleum ether/EtOAc=4:1) to give tert-butyl (3-(1-ethoxyvinyl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate as a yellow solid (1.0 g, yield: 51%). ESI-MS (M+H)+: 333.2. 9. Synthesis of 1-(9-amino-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-3-yl)ethan-1-one To a solution of tert-butyl (3-(1-ethoxyvinyl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate (1.0 g, 3.0 mmol) in THF (30 mL) was added HCl (1 mL, 6.0 mmol) dropwise. The mixture was stirred at rt for 1 h. The solution was used for next step without purification. ESI-MS (M+H)+: 205.1. 10. Synthesis of tert-butyl (3-acetyl-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate Synthesis of tert-butyl (3-acetyl-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate was similar to that of tert-butyl (3-oxo-3,5,6,7,8,9-hexahydro-2H-cyclohepta[c]pyridin-9-yl)carbamate (Example 40, Step 6). The crude product was purified by silica gel column chromatograph (petroleum ether/EtOAc=4:1) to give tert-butyl (3-acetyl-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate as a yellow solid (550 mg, yield: 60% for two steps). ESI-MS (M+H)+: 305.2.1H NMR (400 MHz, CDCl3) δ: 8.54 (s, 1H), 7.78 (s, 1H), 4.99-4.97 (m, 2H), 2.91-2.90 (m, 2H), 2.70 (s, 3H), 1.94-1.74 (m, 4H), 1.62-1.60 (m, 2H), 1.45 (s, 9H). 11. Synthesis of tert-butyl (3-(3-(dimethylamino)acryloyl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate A solution of tert-butyl (3-acetyl-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate (550 mg, 1.8 mmol) in DMF-DMA (10 mL) was stirred at 100° C. for 2 h. After concentration, the crude product was purified by reversed phase HPLC (CH3CN/0.05% NH3·H2O in water) to give tert-butyl (3-(3-(dimethylamino)acryloyl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate (380 mg, yield: 61%). ESI-MS (M+H)+: 360.2. 12. Synthesis of 1-(1-methyl-1H-pyrazol-4-yl)guanidine hydrochloride To a solution of 1-methyl-1H-pyrazol-4-amine (500 mg, 5 mmol, 1.0 eq) in dioxane (10 mL) was added cyanamide (273 g, 6.5 mmol, 1.3 eq) and conc. HCl (1 mL). The reaction was stirred at 100° C. for 12 h. The solvent was removed under reduced pressure. The residue was recrystallized from the co-solvent of MeOH and Et2O. 1-(1-methyl-1H-pyrazol-4-yl)guanidine hydrochloride (600 mg, yield: 55%) was obtained as a yellow solid. ESI-MS (M+H)+: 140.1.1H NMR (400 MHz, CD3OD) δ: 7.78 (s, 1H), 7.48 (s, 1H), 3.91 (s, 3H). 13. Synthesis of tert-butyl (3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate A mixture of 1-(1-methyl-1H-pyrazol-4-yl)guanidine hydrochloride (588 mg, 4.23 mmol), K2CO3(1.2 g, 8.46 mmol), and triethylamine (855 mg, 8.46 mmol) in EtOH (10 mL) was stirred at rt for 1 h, then tert-butyl (3-(3-(dimethylamino)acryloyl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate (380 mg, 1.06 mmol) was added and the mixture was heated to 90° C. for 12 h. After concentration, the crude product was purified by reversed phase HPLC (CH3CN/0.05% NH3·H2O in water) to give tert-butyl (3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate (240 mg, yield: 52%). ESI-MS (M+H)+: 436.2.1H NMR (400 MHz, CDCl3) δ: 8.57 (s, 1H), 8.50 (d, J=5.2 Hz, 1H), 8.08 (s, 1H), 7.85 (s, 1H), 7.66 (d, J=5.6 Hz, 1H), 7.60 (s, 1H), 6.92 (s, 1H), 5.00 (br, 2H), 3.92 (s, 3H), 2.97-2.90 (m, 2H), 2.01-1.82 (m, 5H), 1.51-1.46 (m, 10H). 14. Synthesis of 3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-amine To a solution of tert-butyl (3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate (240 mg, 0.55 mmol) in DCM (5 mL) was added TFA (5 mL). The mixture was stirred at rt for 1 h. After concentration, the residue was basified with NaHCO3(aq) and extracted with DCM (3×30 mL), dried and concentrated to afford 3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-amine as yellow solid (170 mg, Y: 92%, used for next step without further purification). ESI-MS (M+H)+: 336.2. 15. Synthesis of 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide (I-IM_29) To a solution of 3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-amine (80 mg, 0.24 mmol) and triethylamine (49 mg, 0.48 mmol) in DCM (5 mL) were added HATU (182 mg, 0.48 mmol) and potassium 5-(tert-butyl)-1,3,4-oxadiazole-2-carboxylate (75 mg, 0.36 mmol). The mixture was stirred at rt for 3 h. Then water (30 mL) was added and the mixture was extracted with DCM (2×50 mL). The combined organics were dried and concentrated. The crude product was purified by silica gel column chromatograph (DCM/MeOH=10:1) to give 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide as a yellow solid (60 mg, yield: 51%). ESI-MS (M+H)+: 487.9.1H NMR (400 MHz, CD3OD) δ: 8.52 (s, 1H), 8.49 (d, J=5.2 Hz, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.66 (s, 1H), 7.54 (d, J=5.2 Hz, 1H), 5.47-5.45 (m, 1H), 3.90 (s, 3H), 3.08-3.06 (m, 2H), 2.13-1.98 (m, 5H), 1.51-1.49 (m, 10H). Example 41: (R)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide (Compound 41) 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide (216 mg, 0.44 mmol) was subjected to chiral SFC separation (CHIRALPAK AS-H 30×250 mm, 5 um; Co-solvent: 20% Methanol+0.1% DEA in CO2(flow rate: 100 g/min); System backpressure: 120 bar) to afford (R)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide (93 mg, yield: 43%) as a white solid. ESI-MS (M+H)+: 488.1.1H NMR (400 MHz, CD3OD) δ: ppm 8.52 (s, 1H), 8.49 (d, J=5.1 Hz, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.65 (s, 1H), 7.55 (d, J=5.0 Hz, 1H), 5.46 (br d, J=9.5 Hz, 1H), 3.90 (s, 3H), 3.07 (br t, J=4.4 Hz, 2H), 2.12 (br d, J=8.8 Hz, 2H), 1.91-2.05 (m, 3H), 1.44-1.57 (m, 10H). Example 42: (R)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 42a) & (S)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 42b) 1. Synthesis of 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide Synthesis of 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide was similar to that of 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide (Example 40, Step 15). The filtrate was purified by silica gel column chromatograph (DCM/MeOH=10:1) to give 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide as a yellow solid (55 mg, yield: 47%). ESI-MS (M+H)+: 487.9.1H NMR (400 MHz, CD3OD) δ: 8.50 (s, 1H), 8.49 (d, J=5.6 Hz, 1H), 8.21 (s, 1H), 7.97 (s, 1H), 7.66 (s, 1H), 7.54 (d, J=5.2 Hz, 1H), 5.49-5.47 (m, 1H), 3.90 (s, 3H), 3.08-3.06 (m, 2H), 2.12-1.97 (m, 5H), 1.52-1.50 (m, 10H). 2. Synthesis of (R)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide (I-IM_31) & (S)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide (Compound 42) 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide (40 mg, 0.08 mmol) was subjected to chiral SFC separation (CHIRALPAK AS-H 30×250 mm, Sum; Co-solvent: 30% Methanol+0.1% DEA in CO2(flow rate: 100 g/min); System backpressure: 120 bar) to afford (R)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide (11 mg, yield: 27%) & (S)-5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,2,4-oxadiazole-3-carboxamide (10 mg, 26%) as white solids. (R): ESI-MS (M+H)+: 488.1.1H NMR (400 MHz, CD3OD) δ: 8.51 (s, 1H), 8.49 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.65 (s, 1H), 7.55 (d, J=5.1 Hz, 1H), 5.48 (br d, J=9.3 Hz, 1H), 3.90 (s, 3H), 3.01-3.11 (m, 2H), 1.90-2.14 (m, 5H), 1.50-1.53 (m, 9H). (S): ESI-MS (M+H)+: 488.1.1H NMR (400 MHz, CD3OD) δ: 8.50-8.51 (m, 1H), 8.48 (s, 1H), 8.21 (s, 1H), 7.96 (s, 1H), 7.65 (s, 1H), 7.53-7.56 (m, 1H), 5.47 (br d, J=9.5 Hz, 1H), 3.89 (s, 3H), 3.02-3.13 (m, 2H), 1.91-2.16 (m, 5H), 1.50-1.52 (m, 10H). Example 43. 1-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 43) 1. Synthesis of tert-butyl (3-(2-chloropyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate Synthesis of tert-butyl (3-(2-chloropyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate was similar to that of tert-butyl 1-(5-(tert-butyl)-1,2,4-oxadiazole-3-carboxamido)-7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-4,5-dihydro-1H-benzo[d]azepine-3(2H)-carboxylate (Example 1, Step 12). The crude product was purified by silica gel chromatography (petroleum ether/EtOAc=3:1) to give tert-butyl (3-(2-chloropyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate as a yellow solid (170 mg, yield: 42%). ESI-MS (M+H)+: 374.2. 2. Synthesis of tert-butyl (3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate Synthesis of tert-butyl (3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate was similar to that of tert-butyl (R)-(2-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulen-5-yl)carbamate (Example 35, Step 2). The crude product was purified by silica gel column chromatograph (PE/EtOAc=1:1) to give tert-butyl (3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)carbamate as a yellow solid (140 mg, Y: 71%). ESI-MS (M+H)+: 435.2. 3. Synthesis of 3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-amine Synthesis of 3-(2-((1-methyl-H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-amine was similar to that of 5-(tert-butyl)-N-(7-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl)-1,2,4-oxadiazole-3-carboxamide (Example 1, Step 13). The crude product 3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-amine was obtained as yellow solid (100 mg, yield: 94%). ESI-MS (M+H)+: 335.2. 4. Synthesis of 1-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1H-1,2,3-triazole-4-carboxamide Synthesis of 1-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1H-1,2,3-triazole-4-carboxamide was similar to that of 5-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1,3,4-oxadiazole-2-carboxamide in Example 40, Step 15. The crude product was purified by silica gel chromatography (DCM/MeOH=15:1) to give 1-(tert-butyl)-N-(3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyridin-4-yl)-6,7,8,9-tetrahydro-5H-cyclohepta[c]pyridin-9-yl)-1H-1,2,3-triazole-4-carboxamide as a yellow solid (46 mg, yield: 32%). ESI-MS (M+H)+: 486.3.1H NMR (400 MHz, CD3OD) δ: 8.53-8.49 (m, 2H), 8.16 (d, J=5.2 Hz, 1H), 7.92 (s, 1H), 7.69 (s, 1H), 7.51 (s, 1H), 7.20-7.17 (m, 2H), 5.45-5.43 (m, 1H), 3.89 (s, 3H), 3.08-3.06 (m, 2H), 2.14-2.00 (m, 5H), 1.74 (s, 9H), 1.59-1.50 (m, 1H). Examples 44-154 The following compounds were synthesized according to procedures similar to those described in Examples 1-43. CompdNo.Chemical NameStructure1H-NMR and MS44(S)-5-(tert-butyl)-N-(7- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,2,4-oxadiazole-3- carboxamideLCMS: Rt 0.89 min, m/z 488.00.1H NMR (400 MHz, METHANOL-d4) δ: 8.38 (d, J = 5.27 Hz, 1H), 7.83- 8.04 (m, 3H), 7.64 (s, 1H), 7.47 (d, J = 7.97 Hz, 1H), 7.19 (s, 1H),5.34 (d, J = 6.46 Hz, 1H), 3.89 (s, 3H), 2.81- 3.28 (m, 6H), 1.43-1.54 (m, 9H).45(R)-5-(tert-butyl)-N-(7- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,2,4-oxadiazole-3- carboxamideLCMS: Rt = 0.89 min, m/z 488.00.1H NMR (400 MHz, METHANOL-d4) δ: 8.32-8.46 (m, 1H), 7.89-8.05 (m, 3H), 7.64 (s, 1H), 7.48 (d, J = 7.97 Hz, 1H), 7.19 (d, J = 5.33 Hz, 1H), 5.34 (d, J = 6.46 Hz, 1H), 3.89 (s, 3H), 2.91- 3.28 (m, 6H), 1.45-1.55 (m, 9H).465-(tert-butyl)-N-(3-(2- hydroxyethyl)-7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamide1H NMR (400 MHz, CD3OD) δ: 8.28-8.26 (m, 1H), 7.85-7.76 (m, 3H), 7.52 (s, 1H), 7.40-7.37 (m, 1H), 7.08-7.05 (m, 1H), 5.15-5.12 (m, 1H), 3.77 (s, 3H), 3.65-3.62 (m, 2H), 3.14-3.12 (m, 2H), 2.94-2.91 (m, 2H), 2.74-2.65 (m, 3H), 2.61-2.57 (m, 1H), 1.34 (s, 9H). ESI-MS (M + H)+: 532.3.475-(tert-butyl)-N-(7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-3-(methylsulfonyl)- 2,3,4,5-tetrahydro-1H- benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamide1H NMR (400 MHz, CD3OD) δ: 8.41 (d, J = 5.6 Hz, 1H), 7.99- 7.93 (m, 3H), 7.65 (s, 1H), 7.56 (d, J = 7.6 Hz, 1H), 7.16 (d, J = 5.2 Hz, 1H), 5.47-5.45 (m, 1H), 4.00-3.95 (m, 1H), 3.90 (s, 3H), 3.82-3.78 (m, 1H), 3.65-3.61 (m, 1H), 3.44-3.37 (m, 2H), 3.22-3.17 (m, 1H), 2.90 (s, 3H), 1.47 (s, 9H). ESI-MS (M + H)+: 566.2.485-(tert-butyl)-N-(7-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-3-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamide1H NMR (400 MHz, CD3OD) δ: 8.29 (d, J = 5.2 Hz, 1H), 7.85-7.80 (m, 3H), 7.52 (s, 1H), 7.41 (d, J = 8.0 Hz, 1H), 7.08 (d, J = 4.8 Hz, 1H), 5.16-5.15 (m, 1H), 4.62-4.58 (m, 3H), 4.55-4.51 (m, 1H), 3.78 (s, 3H), 3.70-3.66 (m, 1H), 3.20-3.15 (m, 1H), 2.98-2.93 (m, 1H), 2.88-2.83 (m, 1H), 2.70-2.66 (m, 1H), 2.47- 2.44 (m, 1H), 2.35-2.29 (m, 1H), 1.35 (s, 9H). ESI-MS (M + H)+: 544.3.495-(tert-butyl)-N-(7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-3-(tetrahydrofuran-3- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamide1H NMR (400 MHz, CD3OD) δ: 8.39 (d, J = 5.6 Hz, 1H), 7.96 (s, 1H), 7.94 (dd, J = 8.0, 2.0 Hz, 1H), 7.89 (s, 1H), 7.64 (s, 1H), 7.51 (d, J = 8.0 Hz, 1H), 7.18 (d, J = 5.2 Hz, 1H), 5.24-5.22 (m, 1H), 4.01-3.96 (m, 1H), 3.89 (s, 3H), 3.87-3.83 (m, 1H), 3.80-3.70 (m, 2H), 3.53-3.49 (m, 1H), 3.29-3.20 (m, 2H), 3.15-2.97 (m, 2H), 2.82-2.62 (m, 2H), 2.15- 2.10 (m, 1H), 1.98-1.91 (m, 1H), 1.46 (s, 9H). ESI-MS (M + H)+: 558.3.505-(tert-butyl)-N-(3-(2- hydroxy-2- methylpropyl)-7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamide1H NMR (400 MHz, CD3OD) δ: 8.40 (d, J = 4.8 Hz, 1H), 7.97-7.89 (m, 3H), 7.64 (s, 1H), 7.51 (d, J = 8.0, 2.0 Hz, 1H), 7.20-7.19 (m, 1H), 5.22-5.20 (m, 1H), 3.89 (s, 3H), 3.29-3.21 (m, 3H), 3.05-3.00 (m, 1H), 2.93-2.90 (m, 1H), 2.77-2.72 (m, 1H), 2.60 (s, 2H), 1.46 (s, 9H), 1.32-1.31 (m, 6H). ESI-MS (M + H)+: 560.3.511-(tert-butyl)-N-(7-(2- ((1-methyl-1H-pyrazol- 4-yl)amino)pyrimidin- 4-yl)-2,3,4,5- tetrahydro-1H- benzo[d]azepin-1-yl)- 1H-1,2,3-triazole-4- carboxamide1H NMR (400 MHz, CD3OD) δ: 8.48 (s, 1H), 8.40 (d, J = 5.6 Hz, 1H), 7.98-7.93 (m, 3H), 7.65 (br, 1H), 7.50 (d, J = 7.6 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 5.35-5.34 (m, 1H), 3.90 (s, 3H), 3.28-2.99 (m, 6H), 1.73 (s, 9H). ESI-MS (M + H)+: 487.2.521-(tert-butyl)-N-(3- methyl-7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1- yl)-1H-1,2,3-triazole-4- carboxamide1H NMR (400 MHz, CD3OD) δ: 8.45 (s, 1H), 8.41 (d, J = 5.2 Hz, 1H), 7.98-7.92 (m, 3H), 7.65 (s, 1H), 7.52 (d, J = 8.0 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 5.33-5.31 (m, 1H), 3.90 (s, 3H), 3.31-3.26 (m, 1H), 3.11-3.05 (m, 2H), 2.92-2.59 (m, 3H), 2.51 (s, 3H), 1.72 (s, 9H). ESI-MS (M + H)+: 501.3.531-(tert-butyl)-N-(3-(2- hydroxyethyl)-7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1- yl)-1H-1,2,3-triazole-4- carboxamide1H NMR (400 MHz, CD3OD) δ: 8.44 (s, 1H), 8.41 (d, J = 5.2 Hz, 1H), 7.98-7.91 (m, 3H), 7.65 (s, 1H), 7.54 (d, J = 7.6 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 5.21-5.25 (m, 1H), 3.90 (s, 3H), 3.79-3.76 (m, 2H), 3.19-3.02 (m, 4H), 2.86-2.64 (m, 4H), 1.71 (s, 9H). ESI-MS (M + H)+: 531.3.545-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2,2,2- trifluoroethyl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 570.2.1H NMR (400 MHz, CD3OD) δ: 8.31 (d, J = 4.8 Hz, 1H), 7.93-7.88 (m, 3H), 7.50 (s, 1H), 7.37 (d, J = 8.4 Hz, 1H), 7.12 (d, J = 4.8 Hz, 1H), 5.51 (d, J = 10.0 Hz, 1H), 4.26-4.22 (m, 1H), 4.02-3.98 (m, 1H), 3.48 (s, 3H), 3.30-3.27 (m, 2H), 3.05-2.98 (m, 2H), 2.13-2.08 (m, 1H), 1.85-1.82 (m, 1H), 1.41 (s, 9H).555-(tert-butyl)-N-(2-(2- (dimethylamino)acetyl)- 8-(2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 573.3.1H NMR (400 MHz, CD3OD) δ: 8.33-8.29 (m, 1H), 8.10 (s, 1H), 8.02-7.86 (m, 2H), 7.56-7.34 (m, 2H), 7.15-7.12 (m, 1H), 5.61-5.50 (m, 1H), 5.03-4.94 (m, 2H), 4.77-4.36 (m, 1H), 4.16- 4.13 (m, 1H), 4.04-3.99 (m, 3H), 3.83-3.35 (m, 2H), 3.10-3.05 (m, 2H), 2.17-2.12 (m, 6H), 1.42 (s, 9H).56N-(2-(2-hydroxyethyl)- 8-(2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5- yl)-5-(1,1,1-trifluoro-2- methylpropan-2-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 586.3.1H NMR (400 MHz, CD3OD) δ: 8.43 (d, J = 5.2 Hz, 1H), 8.05 (s, 1H), 8.03-8.00 (m, 2H), 7.65 (s, 1H), 7.49 (d, J = 7.6 Hz, 1H), 7.24 (d, J = 5.2 Hz, 1H), 5.59-5.57 (m, 1H), 4.64 (br, 1H), 4.17-4.15 (m, 2H), 3.91 (s, 3H), 3.75 (t, J = 6.0 Hz, 2H), 3.26-3.16 (m, 1H), 2.72-2.61 (m, 2H), 2.34- 2.25 (m, 1H), 2.02-1.97 (m, 1H), 1.76 (s, 6H).57N-(2-methyl-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 5-(1,1,1-trifluoro-2- methylpropan-2-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 556.2.1H NMR (400 MHz, CDCl3) δ: 8.52 (br, 1H), 8.42 (d, J = 5.2 Hz, 1H), 7.87 (s, 1H), 7.85-7.81 (m, 2H), 7.55 (s, 1H), 7.47 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 5.2 Hz, 1H), 6.91 (s, 1H), 5.61-5.56 (m, 1H), 3.94-3.92 (m, 5H), 3.14- 3.07 (m, 1H), 2.85-2.80 (m, 1H), 2.52 (s, 3H), 2.36-2.28 (m, 1H), 2.11-2.03 (m, 1H), 1.71 (s, 6H).58(R)-5-(1,1-difluoro-2- methylpropan-2-yl)-N-(8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 580.2.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.99-7.98 (m, 2H), 7.64 (s, 1H), 7.48 (d, J = 8.0 Hz, 1H), 7.23 (d, J = 5.2 Hz, 1H), 6.15 (t, J = 55.6 Hz, 1H), 5.60-5.56 (m, 1H), 4.78-4.75 (m, 1H), 4.70-4.66 (m, 3H), 3.98- 3.94 (m, 1H), 3.90 (s, 3H), 3.88-3.79 (m, 2H), 3.09-3.06 (m, 1H), 2.93- 2.87 (m, 1H), 2.30-2.20 (m, 1H), 2.07-2.04 (m, 1H), 1.60 (s, 6H).595-(1,1-difluoro-2- methylpropan-2-yl)-N-(2- ((S)-2-hydroxypropyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 582.3.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.03-8.00 (m, 3H), 7.64 (s, 1H), 7.48-7.46 (m, 1H), 7.23 (d, J = 5.2 Hz, 1H), 6.15 (t, J = 55.6 Hz, 1H), 5.57 (d, J = 9.6 Hz, 1H), 4.23- 4.19 (m, 1H), 4.13-4.06 (m, 1H), 4.00-3.96 (m, 1H), 3.91 (s, 3H), 3.26-3.20 (m, 2H), 2.44 (d, J = 6.0 Hz, 2H), 2.31-225 (m, 1H), 1.99-1.95 (m, 1H), 1.60 (s, 6H), 1.16 (d, J = 6.0 Hz, 3H).605-(1-fluoro-2- methylpropan-2-yl)-N-(8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 562.3.1H NMR (400 MHz, CD3OD) δ: 8.29 (d, J = 5.6 Hz, 1H), 8.21 (s, 1H), 7.91-7.89 (m, 1H), 7.86-7.83 (m, 2H), 7.35 (d, J = 8.0 Hz, 1H), 7.09 (d, J = 5.6 Hz, 1H), 5.46 (d, J = 9.6 Hz, 1H), 4.75- 4.57 (m, 1H), 4.56-4.55 (m, 4H), 4.43 (s, 1H), 3.85-3.66 (m, 6H), 2.96-2.92 (m, 1H), 1.94-1.91 (m, 1H), 1.41 (d, J = 2.0 Hz, 6H), 1.19 (m, 2H).615-(1-fluoro-2- methylpropan-2-yl)-N-(2- ((S)-2-hydroxypropyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 564.3.1H NMR (400 MHz, MeOD) δ: 8.31-8.29 (m, 1H), 7.89-7.88 (m, 3H), 7.55 (s, 1H), 7.38-7.36 (m, 1H), 7.11-7.10 (m, 1H), 5.48-5.46 (m, 1H), 4.53 (d, J = 47.2 Hz, 2H), 4.11-3.86 (m, 3H), 3.81 (s, 3H), 3.21-3.14 (m, 2H), 2.35-2.17 (m, 3H), 1.89-1.87 (m, 1H), 1.44 (s, 6H), 1.07-1.03 (m, 3H).625-cyclobutyl-N-(2- methyl-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5- yl)-1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 500.3.1H NMR (400 MHz, CD3OD) δ: 8.31-8.28 (m, 1H), 7.92-7.86 (m, 3H), 7.52 (s, 1H), 7.35-7.33 (m, 1H), 7.12-7.08 (m, 1H), 5.43 (d, J = 10.0 Hz, 1H), 4.00- 3.98 (m, 1H), 3.82-3.79 (m, 2H), 3.77 (s, 3H), 3.08-2.95 (m, 2H), 2.45-2.38 (m, 4H), 2.28 (s, 3H), 2.15-1.89 (m, 4H).635-(2-cyanopropan-2- yl)-N-(2-methyl-8-(2- ((1-methyl-1H-pyrazol- 4-yl)amino)pyrimidin- 4-yl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 513.6.1H NMR (400 MHz, CDCl3) δ: 8.59-8.58 (m, 1H), 8.41 (d, J = 5.2 Hz, 1H), 7.85- 7.81 (m, 3H), 7.53 (s, 1H), 7.45 (d, J = 7.6 Hz, 1H), 7.04 (d, J = 5.2 Hz, 1H), 5.58 (t, J = 8.0 Hz, 1H), 3.93- 3.92 (m, 1H), 3.90 (s, 3H), 3.13-3.06 (m, 1H), 2.86-2.80 (m, 1H), 2.51 (s, 3H), 2.35-2.28 (m, 1H), 2.10-2.03 (m, 2H), 1.91 (s, 6H).645-(2-cyanopropan-2-yl)- N-(2-(2-hydroxyethyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 543.6.1H NMR (400 MHz, CD3OD) δ: 8.40 (d, J = 5.2 Hz, 1H), 8.01-7.98 (m, 3H), 7.64 (s, 1H), 7.47 (d, J = 8.0 Hz, 1H), 7.20 (d, J = 5.2 Hz, 1H), 5.57 (d, J = 9.6 Hz, 1H), 4.20-4.11 (m, 2H), 3.90 (s, 3H), 3.75-3.72 (m, 2H), 3.28-3.20 (m, 2H), 2.68-2.63 (m, 2H), 2.31- 2.27 (m, 1H), 2.01-1.97 (m, 1H), 1.94 (s, 6H).651-isopropyl-N-(8-(2- ((1-methyl-1H-pyrazol- 4-yl)amino)pyrimidin- 4-yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 529.2.1H NMR (400 MHz, DMSO-d6) δ: 9.49 (s, 1H), 9.05 (d, J = 8.0 Hz, 1H), 8.74 (s, 1H), 8.46 (d, J = 5.2 Hz, 1H), 8.03-7.81 (m, 3H), 7.54 (s, 1H), 7.37 (d, J = 8.0 Hz, 1H), 7.25 (d, J = 5.2 Hz, 1H), 5.44 (t, J = 9.6 Hz, 1H), 5.00-4.80 (m, 1H), 4.59 (t, J = 6.4 Hz, 1H), 4.56-4.41 (m, 3H), 3.93- 3.59 (m, 6H), 2.98-2.70 (m, 2H), 2.19-2.03 (m, 1H), 1.90-1.77 (m, 1H), 1.54 (d, J = 6.8 Hz, 6H).661-cyclobutyl-N-(8-(2- ((1-methyl-1H-pyrazol- 4-yl)amino)pyrimidin- 4-yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 540.7.1H NMR (400 MHz, DMSO-de) δ: 9.49 (s, 1H), 9.07 (d, J = 8.4 Hz, 1H), 8.81 (s, 1H), 8.47 (d, J = 5.2 Hz, 1H), 7.97-7.94 (m, 2H), 7.90 (s, 1H), 7.53 (s, 1H), 7.37 (d, J = 8.0 Hz, 1H), 7.26 (d, J = 5.2 Hz, 1H), 5.44 (t, J = 9.6 Hz, 1H), 5.25-5.16 (m, 1H), 4.59 (t, J = 6.4 Hz, 1H), 4.54-4.47 (m, 3H), 3.88-3.62 (m, 2H), 3.68 (s, 3H), 2.93-2.90 (m, 1H), 2.81-2.76 (m, 1H), 2.74-2.72 (m, 1H), 2.60-2.48 (m, 4H), 2.14-2.06 (m, 1H), 1.92- 1.83 (m, 3H).675-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-thiadiazole-2- carboxamideESI-MS (M + H)+: 560.0.1H NMR (400 MHz, CDCl3) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.33 (d, J = 8.4 Hz, 1H), 7.87 (s, 1H), 7.86 (dd, J = 8.0 Hz, 1.6 Hz, 1H), 7.77 (d, J = 1.6 Hz, 1H), 7.53 (s, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.04 (d, J = 5.2 Hz, 1H), 6.95 (s, 1H), 5.62-5.58 (m, 1H), 4.77-4.68 (m, 4H), 3.92-3.86 (m, 6H), 3.04-2.97 (m, 1H), 2.81-2.75 (m, 1H), 2.32-2.26 (m, 1H), 2.13- 2.06 (m, 1H), 1.51 (s, 9H).683-(tert-butyl)-N-(2-(3- hydroxycyclobutyl)-8-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-5- carboxamideESI-MS (M + H)+: 558.3.1H NMR (400 MHz, CDCl3) δ: 8.61 (br, 1H), 8.43 (d, J = 4.8 Hz, 1H), 7.85-7.82 (m, 3H), 7.55 (s, 1H), 7.49 (d, J = 7.6 Hz, 1H), 7.05 (d, J = 4.2 Hz, 1H), 7.02 (s, 1H), 5.61 (t, J = 8.0 Hz, 1H), 4.07-3.73 (m, 6H), 3.15-3.06 (m, 1H), 2.73-2.49 (m, 3H), 2.37- 2.30 (m, 1H), 2.12-1.85 (m, 4H), 1.39 (s, 9H).691-(tert-butyl)-N-(8-(2- ((1-methyl-1H-pyrazol- 4-yl)amino)pyrimidin- 4-yl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 487.2.1H NMR (400 MHz, CDCl3) δ: 8.40 (d, J = 5.2 Hz, 1H), 8.19 (s, 1H), 7.98 (d, J = 8.8 Hz, 1H), 7.89 (s, 1H), 7.84-7.81 (m, 2H), 7.51-7.47 (m, 2H), 7.03 (d, J = 5.2 Hz, 1H), 5.63 (t, J = 9.2 Hz, 1H), 4.16-4.14 (m, 2H), 3.90 (s, 3H), 3.42-3.32 (m, 2H), 2.10-2.08 (m, 2H), 1.71 (s, 9H).701-(tert-butyl)-N-(2-ethyl- 8-(2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 514.8.1H NMR (400 MHz, CDCl3) δ: 8.41 (d, J = 5.2 Hz, 1H), 8.17 (s, 1H), 7.91 (s, 1H), 7.84-7.24 (m, 2H), 7.53 (s, 1H), 7.48 (d, J = 8.8 Hz, 1H), 7.04 (d, J = 5.2 Hz, 2H), 6.95 (s, 1H), 5.61 (t, J = 9.2 Hz, 1H), 4.02 (s, 2H), 3.91 (s, 3H), 3.31-3.22 (m, 1H), 3.02-2.98 (m, 1H), 2.66-2.60 (m, 2H), 2.28-2.21 (m, 1H), 2.06-2.02 (m, 1H), 1.67 (s, 9H), 1.20 (t, J = 7.2 Hz, 3H).711-(tert-butyl)-N-(8-(2- ((1-methyl-1H-pyrazol- 4-yl)amino)pyrimidin- 4-yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 542.7.1H NMR (400 MHz, CD3OD) δ: 8.52 (s, 1H), 8.42 (d, J = 5.6 Hz, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.98-7.97 (m, 2H), 7.64 (s, 1H), 7.48 (d, J = 8.0 Hz, 1H), 7.22 (d, J = 4.4 Hz, 1H), 5.60-5.57 (m, 1H), 4.76 (t, J = 6.4 Hz, 1H), 4.71-4.69 (m, 3H), 3.99-3.82 (m, 6H), 3.10-3.05 (m, 1H), 2.92-2.86 (m, 1H), 2.27-2.18 (m, 1H), 2.07- 2.02 (m, 1H), 1.74 (s, 9H).721-(tert-butyl)-N-(2-(3- hydroxycyclobutyl)-8-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 556.7.1H NMR (400 MHz, CDCl3) δ: 8.41 (d, J = 5.2 Hz, 1H), 8.17 (s, 1H), 7.88 (s, 1H), 7.81-7.78 (m, 2H), 7.54 (s, 1H), 7.48 (d, J = 8.4 Hz, 1H), 7.04-7.02 (s, 2H), 5.59 (t, J = 8.4 Hz, 1H), 4.03- 3.79 (m, 6H), 3.19-3.13 (m, 1H), 2.76-2.63 (m, 4H), 2.31-2.25 (m, 1H), 2.15-1.93 (m, 3H), 1.68 (s, 9H).731-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(tetrahydrofuran-3- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 556.7.1H NMR (400 MHz, CD3OD) δ: 8.47 (s, 1H), 8.38 (d, J = 5.2 Hz, 1H), 7.98-7.95 (m, 3H), 7.62 (d, J = 5.2 Hz, 1H), 7.47 (d, J = 7.6 Hz, 1H), 7.17 (d, J = 5.2 Hz, 1H), 5.57-5.55 (m, 1H), 4.09-3.94 (m, 4H), 3.89 (s, 3H), 3.78-3.70 (m, 2H), 3.28-3.06 (m, 3H), 2.32-1.94 (m, 4H), 1.74 (s, 9H).741-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(tetrahydro-2H- pyran-4-yl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)-1H- 1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 570.7.1H NMR (400 MHz, CDCl3) δ: 8.41 (d, J = 4.8 Hz, 1H), 8.22 (d, J = 10.0 Hz, 1H), 8.16 (s, 1H), 7.88 (s, 1H), 7.83-7.80 (m, 2H), 7.50 (s, 1H), 7.48 (d, J = 7.6 Hz, 1H), 7.04 (d, J = 5.2 Hz, 1H), 6.93 (s, 1H), 5.65 (t, J = 8.4 Hz, 1H), 4.10-4.05 (m, 4H), 3.91 (s, 3H), 3.44-3.37 (m, 2H), 3.19-3.16 (m, 1H), 3.10-3.05 (m, 1H), 2.86-2.81 (m, 1H), 2.31-2.26 (m, 1H), 2.10- 2.04 (m, 1H), 1.95-1.70 (m, 4H), 1.69 (s, 9H).751-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2,2,2- trifluoroethyl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)-1H- 1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 568.7.1H NMR (400 MHz, CDCl3) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.18 (s, 1H), 7.91 (s, 1H), 7.87 (d, J = 7.6 Hz, 1H), 7.83 (s, 1H), 7.78 (d, J = 8.8 Hz, 1H), 7.51 (s, 1H), 7.48 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 5.2 Hz, 1H), 7.02 (s, 1H), 5.61 (t, J = 8.4 Hz, 1H), 4.33- 4.12 (m, 2H), 3.91 (s, 3H), 3.41-3.35 (m, 2H), 3.10-3.03 (m, 2H), 2.22- 2.14 (m, 1H), 2.03-1.97 (m, 1H), 1.71 (s, 9H).761-(tert-butyl)-N-(2-(2- hydroxyethyl)-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 531.3.1H NMR (400 MHz, CDCl3) δ: 8.50 (br, 1H), 8.41 (d, J = 5.2 Hz, 1H), 8.16 (s, 1H), 7.88 (s, 1H), 7.83-7.80 (m, 2H), 7.54 (s, 1H), 7.49 (d, J = 7.6 Hz, 1H), 7.03 (d, J = 5.2 Hz, 1H), 6.91 (s, 1H), 5.61 (t, J = 8.8 Hz, 1H), 4.14-4.03 (m, 2H), 3.92 (s, 3H), 3.80-3.79 (m, 2H), 3.29-3.26 (m, 1H), 3.99-3.96 (m, 1H), 2.80-2.77 (m, 2H), 2.30-2.27 (m, 1H), 2.08- 2.01 (m, 1H), 1.70 (s, 9H).771-(tert-butyl)-N-(2-((R)- 2-hydroxypropyl)-8-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 544.3.1H NMR (400 MHz, DMSO-d6) δ: 9.50 (s, 1H), 9.00 (d, J = 7.2 Hz, 1H), 8.75 (s, 1H), 8.45 (d, J = 5.2 Hz, 1H), 7.95 (s, 3H), 7.53 (s, 1H), 7.36 (d, J = 8.0 Hz, 1H), 7.25 (d, J = 5.2 Hz, 1H), 5.46-5.42 (m, 1H), 4.35-3.94 (m, 3H), 3.82-3.76 (m, 4H), 3.31- 3.13 (m, 2H), 2.36-2.11 (m, 3H), 1.79-1.73 (m, 1H), 1.65 (s, 9H), 1.03 (t, J = 5.2 Hz, 3H).785-(tert-butyl)-N-(2- ((R)-2-hydroxypropyl)- 8-(2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5- yl)-1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 546.3.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.03-7.99 (m, 3H), 7.64 (s, 1H), 7.46 (d, J = 7.2 Hz, 1H),7.24-7.22 (m, 1H), 5.61-5.58 (m, 1H), 4.22-3.98 (m, 3H), 3.91 (s, 3H), 3.28-3.21 (m, 2H), 2.47-2.26 (m, 3H), 1.97-1.94 (m, 1H), 1.52 (s, 9H), 1.16-1.28 (m, 3H).795-(tert-butyl)-N-(2-(2- methoxyethyl)-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 545.7.1H NMR (400 MHz, CD3OD) δ: 8.43 (d, J = 5.2 Hz, 1H), 8.05-8.03 (m, 2H), 7.99 (s, 1H), 7.65 (s, 1H), 7.46 (d, J = 8.8 Hz, 1H), 7.24 (d, J = 5.6 Hz, 1H), 5.59 (d, J = 10.0 Hz, 1H), 4.22-4.11 (m, 2H), 3.90 (s, 3H), 3.61 (t, J = 5.6 Hz, 2H), 3.36 (s, 3H), 3.27-3.15 (m, 2H), 2.77-2.69 (m, 2H), 2.31-2.25 (m, 1H), 2.00-1.97 (m, 1H), 1.53 (s, 9H).805-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(tetrahydrofuran-3- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 558.3.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.04-7.99 (m, 3H), 7.64-7.63 (m, 1H), 7.50 (d, J = 8.0 Hz, 1H), 7.24 (d, J = 5.2 Hz, 1H), 5.59 (d, J = 10.0 Hz, 1H), 4.10-3.91 (m, 4H), 3.90 (s, 3H), 3.79-3.70 (m, 2H), 3.32-3.12 (m, 3H), 2.30-2.18 (m, 2H), 2.05-1.97 (m, 2H), 1.51 (s, 9H).815-(tert-butyl)-N-(2-(3- hydroxycyclobutyl)-8-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 558.2.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.03-8.01 (m, 3H), 7.63 (s, 1H), 7.47 (d, J = 8.4 Hz, 1H), 7.23 (d, J = 5.2 Hz, 1H), 5.56-5.54 (m, 1H), 3.98-3.87 (m, 6H), 3.25- 3.21 (m, 1H), 2.96-2.91 (m, 1H), 2.64-2.18 (m, 4H), 2.02-1.83 (m, 3H), 1.51 (s, 9H).82(R)-5-(tert-butyl)-N-(2- (3-hydroxycyclobutyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 2.9 min, m/z 558.00.1H NMR (400 MHz, METHANOL-d4) δ: 8.41 (d, J = 5.3 Hz, 1H), 8.07-7.94 (m, 3H), 7.65-7.59 (m, 1H), 7.50- 7.43 (m, 1H), 7.22 (d, J = 5.3 Hz, 1H), 5.54 (br d, J = 9.5 Hz, 1H), 4.58 (s, 1H), 4.07-3.94 (m, 2H), 3.92-3.82 (m, 4H), 3.29-3.18 (m, 1H), 3.04- 2.78 (m, 1H), 2.66-2.50 (m, 2H), 2.37-2.10 (m, 1H), 2.10-1.94 (m, 1H), 1.88-1.79 (m, 2H), 1.49 (s, 9H), 0.98-0.79 (m, 1H).835-(tert-butyl)-N-(2-ethyl- 8-(2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 516.2.1H NMR (400 MHz, CD3OD) δ: 8.41 (d, J = 5.2 Hz, 1H), 8.04-7.98 (m, 3H), 7.64 (s, 1H), 7.47 (d, J = 7.6 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 5.56-5.54 (m, 1H), 4.07 (s, 2H), 3.90 (s, 3H), 3.30-3.07 (m, 2H), 2.61-2.55 (m, 2H), 2.28-2.00 (m, 2H), 1.51 (s, 9H), 1.19 (t, J = 7.2 Hz, 3H).84(R)-5-(tert-butyl)-N-(2- ethyl-8-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 2.7 min, m/z 516.10.1H NMR (400 MHz, METHANOL-d4) δ: 8.38 (d, J = 5.3 Hz, 1H), 8.02-7.98 (m, 2H), 7.96 (s, 1H), 7.62 (s, 1H), 7.44 (d, J = 7.8 Hz, 1H), 7.19 (d, J = 5.3 Hz, 1H), 5.53 (br d, J = 9.3 Hz, 1H), 4.05 (s, 2H), 3.87 (s, 3H), 3.29- 3.22 (m, 1H), 3.09 (ddd, J = 13.3 Hz, 10.4 Hz, 2.9 Hz, 1H), 2.64-2.50 (m, 2H), 2.31-2.17 (m, 1H), 2.09-1.91 (m, 1H), 1.49 (s, 9H), 1.17 (t, J = 7.2 Hz, 3H).85(S)-5-(tert-butyl)-N-(2- ethyl-8-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 3.9 min, m/z 516.10.1H NMR (400 MHz, METHANOL-d4) δ: 8.39 (d, J = 5.3 Hz, 1H), 8.04-7.98 (m, 2H), 7.96 (s, 1H), 7.62 (s, 1H), 7.45 (d, J = 8.0 Hz, 1H), 7.20 (d, J = 5.3 Hz, 1H), 5.54 (d, J = 9.3 Hz, 1H), 4.07 (s, 2H), 3.88 (s, 3H), 3.29- 3.20 (m, 1H), 3.10 (ddd, J = 13.2 Hz, 10.5 Hz, 2.6 Hz, 1H), 2.66-2.50 (m, 2H), 2.25 (dtd, J = 14.0 Hz, 10.4 Hz, 3.4 Hz, 1H), 2.01 (dt, J = 14.4 Hz, 2.3 Hz, 1H), 1.49 (s, 9 H), 1.18 (t, J = 7.2 Hz, 3H).865-(tert-butyl)-N-(2- ((S)-2-hydroxypropyl)- 8-(2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5- yl)-1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 546.3.1H NMR (400 MHz, CD3OD) δ: 8.33 (d, J = 5.2 Hz, 1H), 7.94-7.90 (m, 3H), 7.61 (d, J = 2.0 Hz, 1H), 7.42 (d, J = 8.4 Hz, 1H), 7.11 (dd, J = 5.2, 1.6 Hz, 1H), 5.55-5.52 (m, 1H), 4.12-3.93 (m, 3H), 3.87 (s, 3H), 3.23-3.18 (m, 2H), 2.42-2.37 (m, 2H), 2.24-2.22 (m, 1H), 1.93-1.91 (m, 1H), 1.48 (s, 9H), 1.13-1.09 (m, 3H).875-(tert-butyl)-N-(2-((R)- 2-hydroxypropyl)-8-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 545.7.1H NMR (400 MHz, CD3OD) δ: 8.30-8.28 (m, 1H), 7.91-7.69 (m, 3H), 7.52 (s, 1H), 7.36 (d, J = 8.4 Hz, 1H), 7.11 (s, 1H), 5.46 (d, J = 10.0 Hz, 1H), 4.11- 3.85 (m, 3H), 3.79 (s, 3H), 3.10-3.06 (m, 2H), 2.33-2.14 (m, 3H), 1.86- 1.83 (m, 1H), 1.39 (s, 9H), 1.05-1.00 (m, 3H).885-(tert-butyl)-N-(2-(2- hydroxy-2- methylpropyl)-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 559.7.1H NMR (400 MHz, CD3OD) δ: 8.27-8.25 (m, 1H), 7.84-7.81 (m, 3H), 7.52 (s, 1H), 7.34-7.33 (m, 1H), 7.07-7.05 (m, 1H), 5.46 (d, J = 10.0 Hz, 1H), 4.11- 3.97 (m, 2H), 3.77 (s, 3H), 3.21-3.17 (m, 2H), 2.37-2.27 (m, 2H), 2.13- 2.12 (m, 1H), 1.84-1.80 (m, 1H), 1.38 (s, 9H), 1.09 (s, 6H).89a5-tert-butyl-1,3,4- oxadiazole-2-carboxylic acid {(R)-2-(2-hydroxy- 2-methyl-propyl)-8-[2-(1- methyl-1H-pyrazol-4- ylamino)-pyrimidin-4- yl]-2,3,4,5-tetrahydro- 1H-2-benzazepin-5-yl}- amideLCMS: Rt 0.87 min, m/z 560.3.1H NMR (400 MHz, METHANOL-d4) δ: 8.42 (br. s., 1H), 7.90-8.13 (m, 3H), 7.64 (s, 1H), 7.47 (d, J = 8.03 Hz, 1H), 7.22 (br. s., 1H), 5.58 (d, J = 9.79 Hz, 1H), 4.07-4.34 (m, 2H), 3.90 (s, 3H), 2.45 (q, J = 14.31Hz, 2H), 2.18-2.32 (m, 3H), 1.95 (d, J = 13.80 Hz, 1H), 1.50 (s, 9H), 1.22 (br. s., 6H).89b(S)-5-(tert-butyl)-N-(2- (2-hydroxy-2- methylpropyl)-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 0.87 min, m/z 560.3.1H NMR (400 MHz, METHANOL-d4) δ: 8.43 (br. s., 1H), 8.02 (s, 3H), 7.64 (s, 1H), 7.47 (d, J = 8.03 Hz, 1H), 7.23 (br. s., 1H), 5.58 (d, J = 9.54 Hz, 1H), 4.05-4.38 (m, 2H), 3.90 (s, 3H), 2.45 (q, J = 14.06 Hz, 2H), 2.27 (d, J = 2.51Hz, 1H), 1.95 (d, J = 13.80 Hz, 1H), 1.50 (s, 9H), 1.21 (s, 6H).905-(tert-butyl)-N-(2-(3- hydroxypropyl)-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 545.7.1H NMR (400 MHz, CD3OD) δ: 8.29 (d, J = 5.2 Hz, 1H), 7.91-7.88 (m, 2H), 7.85 (s, 1H), 7.53 (s, 1H), 7.35 (d, J = 8.0 Hz, 1H), 7.10 (d, J = 5.2 Hz, 1H), 5.46-5.42 (m, 1H), 4.02-3.93 (m, 2H), 3.78 (s, 3H), 3.51 (t, J = 6.0 Hz, 2H), 3.20-3.15 (m, 1H), 3.04-3.00 (m, 1H), 2.53-2.47 (m, 2H), 2.19- 2.11 (m, 1H), 1.90-1.87 (m, 1H), 1.74-1.68 (m, 2H), 1.38 (s, 9H).915-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2,2,2- trifluoroethyl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 570.2.1H NMR (400 MHz, CDCl3) δ: 8.37 (d, J = 5.2 Hz, 1H), 7.83-7.76 (m, 4H), 7.46 (s, 1H), 7.42 (d, J = 8.0 Hz, 1H), 6.99 (d, J = 5.2 Hz, 1H), 6.85 (s, 1H), 5.56-5.52 (m, 1H), 4.23-4.19 (m, 1H), 4.08-4.04 (m, 1H), 3.85 (s, 3H), 3.28-3.04 (m, 4H), 2.23-2.18 (m, 1H), 2.02-1.95 (m, 1H), 1.40 (s, 9H).92N-(8-(2-((1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)-5- (tert-butyl)-1,3,4- oxadiazole-2- carboxamideESI-MS (M + H)+: 530.3.1H NMR (400 MHz, DMSO-d6) δ: 9.83 (d, J = 8.4 Hz, 1H), 9.47 (s, 1H), 8.47 (d, J = 8.8 Hz, 1H), 7.99 (d, J = 8.8 Hz, 1H), 7.96 (s, 1H), 7.91 (s, 1H), 7.65 (s, 1H), 7.39 (d, J = 8.0 Hz, 1H), 7.27 (d, J = 5.2 Hz, 2H), 5.40 (t, J = 9.6 Hz, 1H), 4.58 (t, J = 6.4 Hz, 1H), 4.52-4.45 (m, 3H), 3.90-3.62 (m, 3H), 2.95-2.92 (m, 1H), 2.83-2.77 (m, 1H), 2.13-2.05 (m, 1H), 1.88- 1.85 (m, 1H), 1.42 (s, 9H).93(R)-5-(tert-butyl)-N-(8- (2-((5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3- yl)amino)pyrimidin-4- yl)-2-(2-hydroxyethyl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 557.7.1H NMR (400 MHz, CD3OD) δ: 8.36 (d, J = 5.2 Hz, 1H), 8.04-8.02 (m, 2H), 7.69 (s, 1H), 7.48 (d, J = 8.0 Hz, 1H), 7.22 (d, J = 5.2 Hz, 1H), 5.60-5.57 (m, 1H), 4.29-4.13 (m, 4H), 3.77 (t, J = 6.0 Hz, 2H), 3.35-3.33 (m, 2H), 2.95 (t, J = 7.2 Hz, 2H), 2.78-2.76 (m, 2H), 2.66-2.62 (m, 2H), 2.34- 2.28 (m, 1H), 2.08-2.01 (m, 1H), 1.50 (s, 9H).94(R)-1-(tert-butyl)-N-(8- (2-((5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol- 3-yl)amino)pyrimidin- 4-yl)-2-(2- hydroxyethyl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 556.7.1H NMR (400 MHz, CD3OD) δ: 8.45 (s, 1H), 8.27 (d, J = 5.2 Hz, 1H), 7.91-7.87 (m, 2H), 7.59 (s, 1H), 7.37 (d, J = 8.0 Hz, 1H), 7.14 (d, J = 5.6 Hz, 1H), 5.49 (d, J = 9.6 Hz, 1H), 4.13- 3.99 (m, 4H), 3.65 (t, J = 6.0 Hz, 2H), 3.20-3.05 (m, 2H), 2.86 (t, J = 7.2 Hz, 2H), 2.62-2.52 (m, 4H), 2.21-2.17 (m, 1H), 1.93-1.89 (m, 1H), 1.65 (s, 9H).95N-(2-methyl-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 5-(1-methylcyclopropyl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 530.2.1H NMR (400 MHz, CD3OD) δ: 8.39 (d, J = 5.2 Hz, 1H), 8.00-7.97 (m, 3H), 7.63 (s, 1H), 7.44 (d, J = 8.0 Hz, 1H), 7.19 (d, J = 5.6 Hz, 1H), 5.53 (d, J = 10.0 Hz, 1H), 4.18-4.06 (m, 2H), 3.90 (s, 3H), 3.73 (t, J = 6.0 Hz, 2H), 3.28-3.19 (m, 2H), 2.67-2.62 (m, 2H), 2.28-2.24 (m, 1H), 1.98-1.95 (m, 1H), 1.61 (s, 3H), 1.45-1.42 (m, 2H), 1.13-1.10 (m, 2H).965-(tert-butyl)-N-(8-(2-((1- ethyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(tetrahydrofuran-3- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 572.3.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.05-8.03 (m, 3H), 7.65 (s, 1H), 7.48 (d, J = 7.6 Hz, 1H), 7.24 (d, J = 5.2 Hz, 1H), 5.60-5.57 (m, 1H), 4.21-3.97 (m, 6H), 3.78- 3.70 (m, 2H), 3.32-3.14 (m, 3H), 2.33-1.96 (m, 4H), 1.51-1.47 (m, 12H).975-(tert-butyl)-N-((R)-8- (2-((1-ethyl-1H-pyrazol- 4-yl)amino)pyrimidin-4- yl)-2-((R*)- tetrahydrofuran-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 0.90 min, m/z 572.10.1H NMR (300 MHz, METHANOL-d4) δ: 8.41 (d, J = 5.3 Hz, 1H), 8.07-7.98 (m, 3H), 7.65 (s, 1H), 7.48 (d, J = 7.6 Hz, 1H), 7.22 (d, J = 5.3 Hz, 1H), 5.56 (br d, J = 9.1Hz, 1H), 4.58 (s, 1H), 4.23-4.07 (m, 4H),4.13-3.90 (m, 2H), 3.82-3.63 (m, 2H), 3.27- 3.05 (m, 2H), 2.42-2.11 (m, 2H), 2.11-1.90 (m,2H), 1.51-1.43 (m, 12H).985-(tert-butyl)-N-(8-(2-((1- ethyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2-hydroxyethyl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 545.7.1H NMR (400 MHz, CD3OD) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.04-8.02 (m, 3H), 7.66 (s, 1H), 7.47 (d, J = 8.0 Hz, 1H), 7.23 (d, J = 5.2 Hz, 1H), 5.58-5.56 (m, 1H), 4.23-4.10 (m, 4H), 3.74 (t, J = 6.0 Hz, 1H), 3.25-3.22 (m, 2H), 2.71-2.63 (m, 2H), 2.32-2.27 (m, 1H), 2.02-1.98 (m, 1H), 1.40-1.47 (m, 12H).995-(tert-butyl)-N-(2- methyl-8-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 502.2.1H NMR (400 MHz, CD3OD) δ: 8.43 (d, J = 5.2 Hz, 1H), 8.07-8.05 (m, 2H), 7.99 (s, 1H), 7.65 (s, 1H), 7.49 (d, J = 8.4 Hz, 1H), 7.25 (d, J = 5.6 Hz, 1H), 5.56 (d, J = 9.6 Hz, 1H), 4.13-4.09 (m, 1H), 3.97-3.93 (m, 1H), 3.90 (s, 3H), 3.21-3.18 (m, 1H), 3.10-3.05 (m, 1H), 2.42 (s, 3H), 2.30-2.26 (m, 1H), 2.05-2.01 (m, 1H), 1.52 (s, 9H).100(R)-5-(tert-butyl)-N-(8- (2-((1,5-dimethyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2-hydroxyethyl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 545.7.1H NMR (400 MHz, CD3OD) δ: 8.32 (d, J = 5.2 Hz, 1H), 8.01-7.99 (m, 2H), 7.60 (s, 1H), 7.44 (d, J = 8.0 Hz, 1H), 7.23 (d, J = 5.6 Hz, 1H), 5.57-5.54 (m, 1H), 4.21-4.07 (m, 2H), 3.82 (s, 3H), 3.73 (t, J = 6.0 Hz, 2H), 3.30- 3.21 (m, 2H), 2.68-2.63 (m, 2H), 2.30-2.27 (m, 1H), 2.24 (s, 3H), 2.01-1.96 (m, 1H), 1.51 (s, 9H).101(R)-1-(tert-butyl)-N-(8- (2-((1,5-dimethyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2-hydroxyethyl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 544.7.1H NMR (400 MHz, CDCl3) δ: 8.47 (br, 1H), 8.36 (d, J = 5.2 Hz, 1H), 8.16 (s, 1H), 7.80-7.78 (m, 2H), 7.67 (s, 1H), 7.45 (d, J = 7.6 Hz, 1H), 7.02 (d, J = 5.2 Hz, 1H), 6.45 (s, 1H), 5.59 (t, J = 8.8 Hz, 1H), 4.12-4.02 (m, 2H), 3.81 (s, 3H), 3.77-3.68 (m, 2H), 3.28-3.24 (m, 1H), 2.99-2.96 (m, 1H), 2.79- 2.76 (m, 2H), 2.33-2.26 (m, 1H), 2.22 (s, 3H), 2.08-2.01 (m, 1H), 1.70 (s, 9H).102(R)-5-(tert-butyl)-N-(8- (2-((1,3-dimethyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 558.3.1H NMR (400 MHz, CD3OD) δ: 8.38 (d, J = 5.6 Hz, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.95 (s, 1H), 7.84 (s, 1H), 7.46 (d, J = 8.0 Hz, 1H), 7.24 (d, J = 5.2 Hz, 1H), 5.61 (d, J = 9.2 Hz, 1H), 4.76 (t, J = 6.4 Hz, 1H), 4.70-4.66 (m, 3H), 3.96-3.81 (m, 3H), 3.84 (s, 3H), 3.08-3.02 (m, 1H), 2.87-2.81 (m, 1H), 2.27-2.24 (m, 1H), 2.22 (s, 3H), 2.07-2.03 (m, 1H), 1.15 (s, 9H).103(R)-1-(tert-butyl)-N-(8- (2-((1,3-dimethyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)-1H- 1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 557.3.1H NMR (400 MHz, CD3OD) δ: 8.53 (s, 1H), 8.37 (d, J = 5.2 Hz, 1H), 8.00 (d, J = 7.6 Hz, 1H), 7.95 (s, 1H), 7.84 (s, 1H), 7.47 (d, J = 8.0 Hz, 1H), 7.24 (d, J = 5.6 Hz, 1H), 5.59 (d, J = 9.6 Hz, 1H), 4.76 (t, J = 6.4 Hz, 1H), 4.70-4.67 (m, 3H), 3.98-3.81 (m, 3H), 3.84 (s, 3H), 3.08-3.05 (m, 1H), 2.92-2.87 (m, 1H), 2.27-2.21 (m, 1H), 2.19 (s, 3H), 2.06-2.03 (m, 1H), 1.74 (s, 9H).104(R)-5-(tert-butyl)-N-(8- (2-((1,3-dimethyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2-hydroxyethyl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 546.3.1H NMR (400 MHz, CD3OD) δ: 8.37 (d, J = 4.8 Hz, 1H), 8.01 (s, 1H), 8.00 (d, J = 8.4 Hz, 1H), 7.85 (s, 1H), 7.44 (d, J = 8.0 Hz, 1H), 7.25 (d, J = 5.6 Hz, 1H), 5.59 (d, J = 9.6 Hz, 1H), 4.21- 4.08 (m, 2H), 3.85 (s, 3H), 3.74 (t, J = 6.0 Hz, 2H), 3.27-3.17 (m, 2H), 2.71-2.63 (m, 2H), 2.30-2.25 (m, 1H), 2.22 (s, 3H), 1.99-1.96 (m, 1H), 1.52 (s, 9H).105(R)-1-(tert-butyl)-N-(2- (2-hydroxyethyl)-8-(2- ((1-isopropyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 559.3.1H NMR (400 MHz, CDCl3) δ: 8.51 (br, 1H), 8.41 (d, J = 4.8 Hz, 1H), 8.17 (s, 1H), 7.95 (s, 1H), 7.83-7.82 (m, 2H), 7.56 (s, 1H), 7.48 (d, J = 8.4 Hz, 1H), 7.15 (s, 1H), 7.02 (d, J = 5.2 Hz, 1H), 5.61 (t, J = 8.0 Hz, 1H), 4.53-4.46 (m, 1H), 4.14-4.02 (m, 2H), 3.79-3.70 (m, 2H), 3.29-3.24 (m, 1H), 2.98-2.95 (m, 1H), 2.79- 2.77 (m, 2H), 2.32-2.27 (m, 1H), 2.06-2.03 (m, 1H), 1.68 (s, 9H), 1.55 (d, J = 6.4 Hz, 6H).1065-(tert-butyl)-N-((R)-2- ((R)-2-hydroxypropyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 546.3.1H NMR (400 MHz, CD3OD) δ: 8.39 (d, J = 5.2 Hz, 1H), 8.00-7.97 (m, 3H), 7.64 (s, 1H), 7.45 (d, J = 8.0 Hz, 1H), 7.19 (d, J = 5.2 Hz, 1H), 5.57-5.55 (m, 1H), 4.21-4.09 (m, 2H), 3.99- 3.97 (m, 1H), 3.90 (s, 3H), 3.28-3.23 (m, 2H), 2.40-2.37 (m, 2H), 2.28- 2.25 (m, 1H), 1.97-1.94 (m, 1H), 1.48 (s, 9H), 1.13-1.12 (m, 3H).107(R)-5-(tert-butyl)-N-(2- (cyclopropylmethyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5- yl)-1,3,4-oxadiazole-2- carboxamideESI-MS (M+H)+: 542.1.1H NMR (300 MHz, METHANOL-d4) δ: 8.45 (d, J = 5.3 Hz, 1H), 8.28-8.23 (m, 2H), 7.92 (s, 1H), 7.70-7.61 (m, 2H), 7.28 (d, J = 5.7 Hz, 1H), 5.72 (br s, 1H), 4.83-4.72 (m, 2H), 3.89 (s, 3H), 3.87-3.67 (m, 2H), 3.25-3.14 (m, 1H), 2.46 (br s, 2H), 1.50 (s, 10H), 1.32-1.17 (m, 1H), 0.85 (br s, 2H), 0.52 (br s, 2H).1081-(tert-butyl)-N-((S)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-((S*)- tetrahydrofuran-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)-1H- 1,2,3-triazole-4- carboxamideLCMS: Rt 0.82 min, m/z 557.00.1H NMR (400 MHz, METHANOL-d4) δ: 8.51 (s, 1H), 8.41 (d, J = 5.4 Hz, 1H), 8.11-8.00 (m, 2H), 7.97 (s, 1H), 7.67-7.58 (m, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.23 (d, J = 5.3 Hz, 1H), 5.66-5.54 (m, 1H), 4.58 (br s, 1H), 4.28-4.108 (m, 2H), 4.03 (td, J = 8.7 Hz, 3.8 Hz, 1H), 3.99-3.93 (m, 1H), 3.89 (s, 3H), 3.86-3.70 (m, 2H), 3.47-3.38 (m, 1H), 3.27-3.17 (m, 1H), 2.37-2.17 (m, 2H), 2.16-1.96 (m, 2H), 1.73 (m, 9H).1091-(tert-butyl)-N-((S)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-((R*)- tetrahydrofuran-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideLCMS: Rt 0.82 min, m/z 557.10.1H NMR (400 MHz, METHANOL-d4) δ: 8.51 (s, 1H), 8.46-8.37 (m, 1H), 8.09-8.00 (m, 2H), 7.99 (s, 1H), 7.65-7.58 (m, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.22 (d, J = 5.3 Hz, 1H), 5.59 (br d, J = 9.5 Hz, 1H), 4.58 (s, 1H), 4.26-4.12 (m, 2H), 4.07-3.94 (m, 2H), 3.89 (s, 3H), 3.86-3.71 (m, 2H), 3.46-3.37 (m, 1H), 3.26-3.19 (m, 1H), 2.36-2.16 (m, 2H), 2.14- 1.95 (m,2H), 1.73 (s, 9H).110(R)-5-(tert-butyl)-N-(8- (2-((1,3-dimethyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2,2,2- trifluoroethyl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 584.3.1H NMR (400 MHz, CD3OD) δ: 8.38 (d, J = 5.2 Hz, 1H), 8.03 (d, J = 8.4 Hz, 1H), 7.98 (s, 1H), 7.84 (s, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.25 (d, J = 5.6 Hz, 1H), 5.59 (d, J = 9.6 Hz, 1H), 4.37-4.34 (m, 1H), 4.13-4.10 (m, 1H), 3.84 (s, 3H), 3.46-3.41 (m, 2H), 3.15-3.08 (m, 2H), 2.25-2.17 (m, 1H), 2.22 (s, 3H), 1.97-1.93 (m, 1H), 1.51 (s, 9H).1115-(tert-butyl)-N-(2-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,3,4-oxadiazole-2- carboxamideLCMS: Rt 1.23 min.; (M + H)+487.0;1H NMR (400 MHz, METHANOL- d4) δ: 9.70 (br. s., 1H), 8.34 (d, J = 5.77 Hz, 1H), 8.02 (s, 2H), 7.97 (s, 1H), 7.69 (s, 1H), 7.47 (d, J = 8.53 Hz, 1H), 7.41 (s, 1H), 5.44 (t, J = 8.28 Hz, 1H), 3.93 (s, 3H), 2.87- 3.22 (m, 2H), 1.77-2.28 (m, 4H), 1.51 (s, 11H).112N-(2-(2-((1H-pyrazol-4 yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-5-(tert-butyl)-1,2,4- oxadiazole-3- carboxamideESI-MS (M + H)+: 473.1.1H NMR (400 MHz, CD3OD) δ: 8.41 (d, J = 5.2 Hz, 1H), 8.07 (br, 1H), 7.96-7.95 (m, 2H), 7.76 (br, 1H), 7.40 (d, J = 8.4 Hz, 1H), 7.22 (d, J = 5.6 Hz, 1H), 5.44 (d, J = 9.6 Hz, 1H), 3.06- 3.03 (m, 2H), 2.10-1.91 (m, 5H), 1.53 (s, 9H), 1.49-1.46 (m, 1H).1135-(tert-butyl)-N-(2-(2-((5- methyl-4,5,6,7- tetrahydropyrazolo[1,5- a]pyrazin-3- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 542.2.1H NMR (400 MHz, CD3OD) δ: 8.34 (d, J = 5.2 Hz, 1H), 7.94-7.91 (m, 2H), 7.70 (s, 1H), 7.38 (d, J = 8.0 Hz, 1H), 7.25 (d, J = 5.6 Hz, 1H), 5.45-5.42 (m, 1H), 4.23-4.21 (m, 2H), 3.67 (s, 2H), 3.02-2.99 (m, 4H), 2.49 (s, 3H), 2.06-1.92 (m, 5H), 1.54 (s, 9H), 1.51-1.42 (m, 1H).114N-(2-(2-((1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-5-(tert-butyl)-1,3,4- oxadiazole-2- carboxamideESI-MS (M + H)+: 472.7.1H NMR (400 MHz, DMSO-d6) δ: 12.46 (s, 1H), 9.89 (d, J = 8.0 Hz, 1H), 9.45 (s, 1H), 8.45 (d, J = 5.2 Hz, 1H), 7.96-7.91 (m, 3H), 7.63-7.62 (m, 1H), 7.37 (d, J = 8.0 Hz, 1H), 7.24 (d, J = 5.2 Hz, 1H), 5.28-5.24 (m, 1H), 2.97-2.96 (m, 2H), 2.00-1.77 (m, 5H), 1.42 (s, 9H), 1.33-1.30 (m, 1H).1151-(tert-butyl)-N-(2-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 486.2.1H NMR (400 MHz, CDCl3) δ: 8.39 (d, J = 5.2 Hz, 1H), 8.16 (s, 1H), 7.90 (s, 1H), 7.81 (s, 1H), 7.79 (s, 1H), 7.68 (d, J = 8.4 Hz, 1H), 7.54 (s, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 5.2 Hz, 1H), 6.91 (s, 1H), 5.46-5.42 (m, 1H), 3.91 (s, 3H), 3.04-3.01 (m, 2H), 2.05-1.88 (m, 6H), 1.72 (s, 9H).1165-(tert-butyl)-4-methyl- N-(2-(2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-4H-1,2,4-triazole-3- carboxamideESI-MS (M + H)+: 500.3.1H NMR (400 MHz, CD3OD) δ: 8.27 (d, J = 5.2 Hz, 1H), 7.85-7.81 (m, 3H), 7.53 (s, 1H), 7.30 (d, J = 8.8 Hz, 1H), 7.08 (d, J = 5.2 Hz, 1H), 5.26-5.24 (m, 1H), 4.04 (s, 3H), 3.77 (s, 3H), 2.95-2.93 (m, 2H), 1.97-1.79 (m, 5H), 1.36-1.33 (m, 1H), 1.30 (s, 9H).1172-isopropyl-N-(2-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-2H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 472.2.1H NMR (400 MHz, CDCl3) δ: 8.41 (d, J = 5.2 Hz, 1H), 8.06 (s, 1H), 7.87-7.81 (m, 3H), 7.55 (s, 1H), 7.40 (d, J = 7.6 Hz, 1H), 7.20-7.18 (m, 1H), 7.06 (d, J = 5.2 Hz, 1H), 6.99 (s, 1H), 5.45- 5.41 (m, 1H), 4.91-4.85 (m, 1H), 3.91 (s, 3H), 3.08-2.96 (m, 2H), 2.03-1.86 (m, 5H), 1.67-1.66 (m, 1H), 1.63 (d, J = 6.8 Hz, 6H).118a**Racemic mixture of 5- (tert-butyl)-N-((5R,8S)-8- fluoro-2-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,3,4-oxadiazole-2- carboxamide and 5-(tert- butyl)-N-((5S,8R)-8- fluoro-2-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 505.2.1H NMR (400 MHz, CDCl3) δ: 8.44 (d, J = 5.2 Hz, 1H), 7.90-7.88 (m, 3H), 7.60 (d, J = 7.6 Hz, 1H), 7.55 (s, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.08 (d, J = 5.2 Hz, 1H), 6.91 (s, 1H), 5.38 (d, J = 8.0 Hz, 1H), 4.91 (d, J = 47.2 Hz, 1H), 3.92 (s, 3H), 3.45-3.29 (m, 2H), 2.32-2.20 (m, 3H), 2.00-1.93 (m, 1H), 1.48 (s, 9H).118b**Racemic mixture of 5- (tert-butyl)-N-((5R,8R)- 8-fluoro-2-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,3,4-oxadiazole-2- carboxamide and 5-(tert- butyl)-N-((5S,8S)-8- fluoro-2-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,3,4-oxadiazole-2- carboxamide1H NMR (400 MHz, CDCl3) δ: 8.44 (d, J = 5.2 Hz, 1H), 7.92-7.88 (m, 3H), 7.56-7.54 (m, 2H), 7.40 (d, J = 8.0 Hz, 1H), 7.07 (d, J = 5.2 Hz, 1H), 6.89 (s, 1H), 5.40 (d, J = 9.6 Hz, 1H), 4.70 (d, J = 48.0 Hz, 1H), 3.92 (s, 3H), 3.45-3.28 (m, 2H), 2.37-2.30 (m, 1H), 2.22-2.17 (m, 2H), 1.95-1.92 (m, 1H), 1.49 (s, 9H).119**Racemic mixture of N- ((5R,8S)-8-fluoro-2-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-5-(1- methylcyclopropyl)- 1,2,4-oxadiazole-3- carboxamide and N- ((5S,8R)-8-fluoro-2-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-5-(1- methylcyclopropyl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 503.2.1H NMR (400 MHz, CDCl3) δ: 8.44 (d, J = 5.2 Hz, 1H), 7.90-7.88 (m, 3H), 7.55 (s, 1H), 7.40-7.35 (m, 2H), 7.07 (d, J = 5.2 Hz, 1H), 6.87 (s, 1H), 5.42 (d, J = 8.0 Hz, 1H), 4.90 (d, J = 48.8 Hz, 1H), 3.92 (s, 3H), 3.39-3.33 (m, 2H), 2.33-2.17 (m, 3H), 1.95-1.92 (m, 1H), 1.61 (s, 3H), 1.53-1.50 (m, 2H), 1.12-1.09 (m, 2H).120**Racemic mixture of N- ((5S,8S)-8-fluoro-2-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-5-(1- methylcyclopropyl)- 1,2,4-oxadiazole-3- carboxamide and N- ((5R,8R)-8-fluoro-2-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-5-(1- methylcyclopropyl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 503.2.1H NMR (400 MHz, CDCl3) δ: 8.44 (d, J = 5.6 Hz, 1H), 7.91-7.87 (m, 3H), 7.55 (s, 1H), 7.36-7.30 (m, 2H), 7.07 (d, J = 5.2 Hz, 1H), 6.87 (s, 1H), 5.43 (d, J = 8.4 Hz, 1H), 4.67 (d, J = 47.2 Hz, 1H), 3.92 (s, 3H), 3.45-3.26 (m, 2H), 2.34-2.17 (m, 3H), 1.94-1.91 (m, 1H), 1.63 (s, 3H), 1.55-1.53 (m, 2H), 1.13-1.12 (m, 2H).1211-(tert-butyl)-N-((5R)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(tetrahydrofuran-3- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 557.3.1H NMR (400 MHz, CD3OD) δ: 8.53 (s, 1H), 8.42 (d, J = 5.2 Hz, 1H), 8.05-7.99 (m, 3H), 7.64 (s, 1H), 7.48 (d, J = 8.4 Hz, 1H), 7.23 (d, J = 5.2 Hz, 1H), 5.58 (d, J = 10.4 Hz, 1H), 4.16- 4.09 (m, 2H), 4.02-3.96 (m, 2H), 3.90 (s, 3H), 3.79-3.71 (m, 2H), 3.31-3.24 (m, 2H), 3.15-3.10 (m, 1H), 2.31-2.17 (m, 2H), 2.07-1.97 (m, 2H), 1.74 (s, 9H).122(R)-N-(2-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-5-(1- methylcyclopropyl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 485.2.1H NMR (400 MHz, CD3OD) δ: 8.39 (d, J = 5.2 Hz, 1H), 7.97-7.92 (m, 3H), 7.65 (s, 1H), 7.40 (d, J = 8.8 Hz, 1H), 7.20 (d, J = 5.2 Hz, 1H), 5.42-5.39 (m, 1H), 3.89 (s, 3H), 3.07-3.02 (m, 2H), 2.10-1.86 (m, 5H), 1.62 (s, 3H), 1.47-1.43 (m, 3H), 1.13-1.10 (m, 2H).1235-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 488.3.1H NMR (400 MHz, CD3OD) δ: 8.40 (d, J = 5.6 Hz, 1H), 8.02-7.98 (m, 3H), 7.64 (s, 1H), 7.47 (d, J = 8.4 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 5.60-5.58 (m, 1H), 4.12 (s, 2H), 3.89 (s, 3H), 3.39-3.37 (m, 1H), 3.28-3.21 (m, 2H), 2.15-2.09 (m, 2H), 1.51 (s, 9H).124*5-(tert-butyl)-N-((R)-8- (2-((1-ethyl-1H-pyrazol- 4-yl)amino)pyrimidin-4- yl)-2-((S*)- tetrahydrofuran-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 0.90 min, m/z 572.10.1H NMR (300 MHz, METHANOL-d4) δ: 8.41 (d, J = 5.3 Hz, 1H), 8.07-7.98 (m, 3H), 7.65 (s, 1H), 7.48 (d, J = 7.6 Hz, 1H), 7.22 (d, J = 5.3 Hz, 1H), 5.56 (br d, J = 9.1Hz, 1H), 4.58 (s, 1H), 4.23-4.09 (m, 4H), 4.00-3.92 (m, 2H), 3.78-3.69 (m, 2H), 3.27- 3.05 (m, 2H), 2.36-2.20 (m, 2H), 2.16-1.91 (m, 2H), 1.51-1.43 (m, 12H).125N-(2-(2-hydroxyethyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 5-(1-methylcyclopropyl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 530.2.1H NMR (400 MHz, CD3OD) δ: 8.39 (d, J = 5.2 Hz, 1H), 8.00-7.97 (m, 3H), 7.63 (s, 1H), 7.44 (d, J = 8.0 Hz, 1H), 7.19 (d, J = 5.6 Hz, 1H), 5.53 (d, J = 10.0 Hz, 1H), 4.18-4.06 (m, 2H), 3.90 (s, 3H), 3.73 (t, J = 6.0 Hz, 2H), 3.28-3.19 (m, 2H), 2.67-2.62 (m, 2H), 2.28-2.24 (m, 1H), 1.98-1.95 (m, 1H), 1.61 (s, 3H), 1.45-1.42 (m, 2H), 1.13-1.10 (m, 2H).1265-cyclobutyl-N-(2-(2- hydroxyethyl)-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 529.7.1H NMR (400 MHz, CDCl3) δ: 8.80 (br, 1H), 8.43 (d, J = 5.2 Hz, 1H), 7.87 (s, 1H), 7.84-7.81 (m, 2H), 7.55 (s, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 5.2 Hz, 1H), 6.90 (s, 1H), 5.59 (t, J = 8.0 Hz, 1H), 4.13-4.04 (m, 2H), 3.91 (s, 3H), 3.81-3.69 (m, 3H), 3.27-3.20 (m, 1H), 2.96-2.91 (m, 1H), 2.81 (t, J = 4.8 Hz, 2H), 2.52-2.32 (m, 5H), 2.17-2.02 (m, 3H), 1.60 (s, 9H).127(R)-5-(tert-butyl)-N-(2- (2-ethoxyethyl)-8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 560.1.1H NMR (300 MHz, METHANOL-d4) δ: 8.39 (d, J = 5.3 Hz, 1H), 8.02-7.95 (m, 3H), 7.62 (s, 1H), 7.45 (d, J = 7.9 Hz, 1H), 7.19 (d, J = 5.3 Hz, 1H), 5.54 (br d, J = 9.1Hz, 1H), 4.15 (d, J = 4.9 Hz, 2H), 3.88 (s, 3H), 3.62 (t, J = 5.7 Hz, 2H), 3.50 (q, J = 6.8 Hz, 2H), 3.26- 3.14 (m, 2H), 2.76-2.63 (m, 2H), 2.34-2.19 (m, 1H), 2.05-1.92 (m, 1H), 1.50-1.47 (m, 9H), 1.16 (t, J = 7.0 Hz, 3H).128(R)-5-(tert-butyl)-N-(7- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-3-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 0.87 min, m/z 544.2.1H NMR (400 MHz, METHANOL-d4) δ: 8.39 (br. s., 1H), 7.81-8.04 (m, 3H), 7.64 (br. s., 1H), 7.50 (d, J = 8.03 Hz, 1H), 7.17 (s, 1H), 5.25 (d, J = 6.53 Hz, 1H), 4.53-4.78 (m, 4H), 3.90 (br. s., 3H), 3.67-3.83 (m, 1H), 2.20-3.28 (m, 6H), 1.37-1.66 (m, 9H).1295-(tert-butyl)-N-(7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tctrahydio- 1H-benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 488.00.1H NMR (400 MHz, DMSO-d6) δ: 10.15 (d, J = 8.0 Hz, 1H), 9.53 (s, 1H), 9.21 (br s, 2H), 8.49 (d, J = 5.3 Hz, 1H), 8.09- 7.99 (m, 2H), 7.91 (s, 1H), 7.57 (br s, 1H), 7.48 (d, J = 8.3 Hz, 1H), 7.27 (d, J = 5.3 Hz, 1H), 5.62 (br t, J = 8.4 Hz, 1H), 3.82 (s, 3H), 3.61-3.47 (m, 2H), 3.44-3.21 (m, 3H), 3.13 (br d, J = 9.0 Hz, 1H), 1.43 (s, 9H).1301-(tert-butyl)-N-(7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-3-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[d]azepin-1-yl)-1H- 1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 543.3.1H NMR (400 MHz, CD3OD) δ: 8.44 (s, 1H), 8.41 (d, J = 5.2 Hz, 1H), 7.98-7.92 (m, 3H), 7.65 (s, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 5.29-5.27 (m, 1H), 4.81-4.65 (m, 4H), 3.90 (s, 3H), 3.86-3.79 (m, 1H), 3.10-2.87 (m, 4H), 2.57-2.37 (m, 2H), 1.72 (s, 9H).1315-(tert-butyl)-N-(8-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(tetrahydrofuran-3- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-thiadiazole-2- carboxamideESI-MS (M + H)+: 574.0.1H NMR (400 MHz, CDCl3) δ: 8.42 (d, J = 5.2 Hz, 1H), 8.38 (brs, 1H), 7.89-7.81 (m, 3H), 7.53 (s, 1H), 7.48 (d, J = 7.6 Hz, 1H), 7.05 (d, J = 5.2 Hz, 1H), 6.96 (s, 1H), 5.62-5.56 (m, 1H), 4.09-3.92 (m, 4H), 3.91 (s, 3H), 3.81-3.73 (m, 2H), 3.48-3.40 (m, 1H), 3.23-3.08 (m, 1H), 2.99-2.95 (m, 1H), 2.35-2.28 (m, 1H), 2.19- 2.06 (m, 2H), 2.00-1.95 (m, 1H), 1.51 (s, 9H).1325-(tert-butyl)-N-(3- methyl-7-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 502.1.133(S)-5-(tert-butyl)-N-(8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 544.0.1H NMR (400 MHz, METHANOL-d4) δ: 8.40 (d, J = 5.02 Hz, 1H), 7.87-8.09 (m, 3H), 7.63 (s, 1H), 7.45 (d, J = 8.28 Hz, 1H), 7.20 (d, J = 5.27 Hz, 1H), 5.60 (s, 1H), 4.55-4.77 (m, 4H), 3.89 (s, 3H), 3.75-3.85 (m, 3H), 2.75-3.10 (m, 2H), 1.89-2.42 (m, 2H), 1.51 (s, 9H).134(S)-5-(tert-butyl)-N-(2- (3-hydroxycyclobutyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 4.5 min, m/z 558.00.1H NMR (400 MHz, METHANOL-d4) δ: 8.41 (d, J = 5.3 Hz, 1H), 8.03-7.98 (m, 3H), 7.62 (s, 1H), 7.47-7.45 (m, 1H), 7.22 (d, J = 5.3 Hz, 1H), 5.58- 5.48 (m, 1H), 4.58 (br s, 1H), 3.99 (br s, 1H), 3.89 (s, 3H), 3.25-3.21 (m, 1H), 2.97-2.91 (m, 1H), 2.63- 2.52 (m, 2H), 2.25-2.15 (m, 2H), 2.05-1.98 (m, 1H), 1.87-1.79 (m, 2H), 1.49 (s, 9H), 0.97-0.86 (m, 1H).1355-(tert-butyl)-N-(2-(2-((1 methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,3,4-thiadiazole-2- carboxamideESI-MS (M + H)+: 503.2.1H NMR (400 MHz, CDCl3) δ: 8.40 (d, J = 5.2 Hz, 1H), 7.89-7.80 (m, 4H), 7.54 (s, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.06 (d, J = 5.2 Hz, 1H), 6.92 (s, 1H), 5.41-5.39 (m, 1H), 3.91 (s,3H), 3.03-3.00 (m, 2H), 2.00-1.84 (m, 6H), 1.53 (s, 9H).1365-(tert-butyl)-N-((S)-2- ((S)-2-hydroxypropyl)-8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 0.85 min, m/z 546.3.1H NMR (400 MHz, METHANOL-d4) δ: 8.41 (br. s., 1H),7.91-8.12 (m, 3H), 7.64 (br. s., 1H), 7.46 (d, J = 7.53 Hz, 1H), 7.20 (br. s., 1H), 5.57 (d, J = 9.79 Hz, 1H), 4.08-4.36 (m, 2H), 4.00 (br. s., 1H), 3.83-3.92 (m, 3H), 3.47-3.75 (m, 4H), 3.28 (br. s., 2H), 2.37-2.62 (m, 2H), 2.26 (br. s., 1H), 1.84-2.07 (m, 1H), 1.50 (s, 9H), 0.95-1.24 (m, 4H).137(S)-5-(tert-butyl)-N-(8- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-2-(2,2,2- trifluoroethyl)-2,3,4,5- tetrahydro-1H- benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-3- carboxamideLCMS: Rt 1.36 min, m/z 570.3.1H NMR (400 MHz, METHANOL-d4) δ: 8.40 (br. s., 1H), 7.98 (d, J = 5.27 Hz, 3H), 7.62 (br. s., 1H), 7.46 (br. s., 1H), 7.18 (br. s., 1H), 5.60 (d, J = 8.78 Hz, 1H), 4.26-4.45 (m, 1H), 4.02-4.20 (m, 1H), 3.89 (br. s., 3H), 3.39 (br. s., 1H), 3.11 (br. s., 2H), 2.24 (br. s., 1H), 1.97 (br. s., 1H), 1.52 (s, 9H).138*5-(tert-butyl)-N-((S)-8- (2-((1-ethyl-1H-pyrazol- 4-yl)amino)pyrimidin-4- yl)-2-((R*)- tetrahydrofuran-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 0.90 min, m/z 572.0.1H NMR (300 MHz, METHANOL-d4) δ: 8.41 (d, J = 5.3 Hz, 1H), 8.07-7.97 (m, 3H), 7.65 (s, 1H), 7.48 (d, J = 7.6 Hz, 1H), 7.22 (d, J = 5.3 Hz, 1H), 5.56 (br d, J = 8.7 Hz, 1H), 4.58 (s, 1H), 4.22-4.08 (m, 4H), 4.03-3.92 (m, 2H), 3.78-3.69 (m, 2H), 3.27- 3.07 (m, 2H), 2.36-2.14 (m, 2H), 2.06-1.91 (m, 2H), 1.53-1.43 (m, 12H).139*5-(tert-butyl)-N-((S)-8- (2-((1-ethyl-1H-pyrazol- 4-yl)amino)pyrimidin-4- yl)-2-((S*)- tetrahydrofuran-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 0.90 min, m/z 572.0.1H NMR (300 MHz, METHANOL-d4) δ: 8.41 (d, J = 5.3 Hz, 1H), 8.04- 8.01 (m, 3H), 7.63 (s, 1H), 7.47 (d, J = 7.6 Hz, 1H), 7.23 (d, J = 5.3 Hz, 1H), 5.57 (br d, J = 8.7 Hz, 1H), 4.58 (s, 1H), 4.23-4.10 (m, 4H), 4.04- 3.93 (m, 2H), 3.78-3.70 (m, 2H), 3.25-3.07 (m, 2H), 2.35-2.13 (m, 2H), 2.07-1.90 (m, 2H), 1.51-1.43 (m, 12H).140tert-butyl 1-(5-(tert- butyl)-1,3,4-oxadiazole- 2-carboxamido)-7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-1,2,4,5-tetrahydro- 3H-benzo[d]azepine-3- carboxylateESI-MS (M + H)+: 588.10.1H NMR (400 MHz, CHLOROFORM-d) δ: 8.44 (d, J = 5.3 Hz, 1H), 7.94-7.79 (m, 3H), 7.62-7.50 (m, 2H), 7.06 (d, J = 5.3 Hz, 1H), 6.92 (s, 1H), 5.46-5.30 (m, 1H), 4.48 (br s, 1H), 4.37-4.18 (m, 1H), 3.92 (s, 3H), 3.44 (br d, J = 13.3 Hz, 1H), 3.31-3.11 (m, 2H), 3.05 (br dd, J = 6.5, 13.6 Hz, 1H), 1.56-1.33 (m, 18H).141(S)-5-(tert-butyl)-N-(7- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-3-(oxetan-3-yl)- 2,3,4,5-tetrahydro-1H- benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideLCMS: Rt 0.87 min, m/z 544.3.1H NMR (400 MHz, METHANOL-d4) δ: 8.39 (d, J = 5.02 Hz, 1H), 7.85- 8.03 (m, 3H), 7.64 (s, 1H), 7.51 (d, J = 8.03 Hz, 1H), 7.18 (d, J = 5.27 Hz, 1H), 5.26 (d, J = 6.78 Hz, 1H), 4.72 (d, J = 7.28 Hz, 4H), 3.89 (s, 3H), 3.79 (d, J = 6.27 Hz, 1H), 2.21-3.29 (m, 6H), 1.47 (s, 9H).1425-(tert-butyl)-N-(7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-3-(2,2,2- trifluoroethyl)-2,3,4,5- tetrahydro-1H- benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 570.3.1H NMR (400 MHz, CD3OD) δ: 8.29 (d, J = 5.2 Hz, 1H), 7.85-7.79 (m, 3H), 7.52 (s, 1H), 7.41 (d, J = 8.0 Hz, 1H), 7.08 (d, J = 5.2 Hz, 1H), 5.14-5.13 (m, 1H), 3.78 (s, 3H), 3.36-3.23 (m, 3H), 3.15-3.09 (m, 2H), 3.01-2.92 (m, 2H), 2.86-2.81 (m, 1H), 1.35 (s, 9H).1435-(tert-butyl)-N-(3-((S)- 2-hydroxypropyl)-7-(2- ((1-methyl-1H-pylazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 546.3.1H NMR (400 MHz, CDCl3) δ: 8.95-8.79 (m, 1H), 8.41 (d, J = 5.2 Hz, 1H), 7.87- 7.78 (m, 3H), 7.56-7.54 (m, 2H), 7.04 (d, J = 5.2 Hz, 1H), 6.97 (s, 1H), 5.18-5.14 (m, 1H), 3.92-3.91 (m, 1H), 3.90 (s, 3H), 3.39-2.43 (m, 8H), 1.43 (s, 9H), 1.23-1.21 (m, 3H).1445-(tert-butyl)-N-(3-((R)- 2-hydroxypropyl)-7-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 546.3.1H NMR (400 MHz, CD3OD) δ: 8.40 (d, J = 4.8 Hz, 1H), 7.97-7.90 (m, 3H), 7.64 (s, 1H), 7.51 (d, J = 8.0, 2.0 Hz, 1H), 7.20-7.18 (m, 1H), 5.26-5.24 (m, 1H), 4.01-3.98 (m, 1H), 3.90 (s, 3H), 3.27-3.21 (m, 2H), 3.14-3.03 (m, 2H), 2.85-2.80 (m, 1H), 2.70-2.56 (m, 3H), 1.46 (s, 9H), 1.26-1.23 (m, 3H).1455-(tert-butyl)-N-(7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-3-(tetrahydro-2H- pyran-4-yl)-2,3,4,5- tetrahydro-1H- benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 572.3.1H NMR (400 MHz, CDCl3) δ: 9.02 (d, J = 7.2 Hz, 1H), 8.41 (d, J = 5.2 Hz, 1H), 7.87 (s, 1H), 7.81 (dd, J = 8.0, 1.6 Hz, 1H), 7.76 (d, J = 1.6 Hz, 1H), 7.58 (d, J = 8.0 Hz, 1H), 7.53 (s, 1H), 7.03 (d, J = 5.2 Hz, 1H), 6.98 (s, 1H), 5.11-5.08 (m, 1H), 4.05-4.02 (m, 2H), 3.91 (s, 3H), 3.41-3.25 (m, 5H), 2.92-2.67 (m, 4H), 1.76-1.63 (m, 4H), 1.42 (s, 9H).1465-(tert-butyl)-N-(3-(3- hydroxypropyl)-7-(2-((1- methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[d]azepin-1-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 545.7.1H NMR (400 MHz, CDCl3) δ: 9.02 (d, J = 6.8 Hz, 1H), 8.40 (d, J = 5.2 Hz, 1H), 7.89-7.76 (m, 3H), 7.57-7.52 (m, 3H), 7.03 (d, J = 5.2 Hz, 1H), 5.13- 5.10 (m, 1H), 3.94-3.84 (m, 5H), 3.32-3.15 (m, 3H), 2.90-2.67 (m, 5H), 2.45-2.42 (m, 1H), 1.83-1.81 (m, 2H), 1.43 (s, 9H).1475-(3,3- difluorocyclobutyl)-N-(2- methyl-8-(2-((1-methyl- 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 536.2.1H NMR (400 MHz, CDCl3) δ: 8.43-8.42 (m, 2H), 7.87 (s, 1H), 7.85-7.82 (m, 2H), 7.55 (s, 1H), 7.46 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 5.2 Hz, 1H), 6.94 (s, 1H), 5.60-5.56 (m, 1H), 3.98-3.88 (m, 5H), 3.69-3.61 (m, 1H), 3.13- 3.05 (m, 5H), 2.87-2.81 (m, 1H), 2.52 (s, 3H), 2.36-2.28 (m, 1H), 2.10-2.03 (m, 1H).1485-(3,3- difluorocyclobutyl)-N-(2- (2-hydroxyethyl)-8-(2- ((1-methyl-1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-2,3,4,5-tetrahydro- 1H-benzo[c]azepin-5-yl)- 1,3,4-oxadiazole-2- carboxamideESI-MS (M + H)+: 566.2.1H NMR (400 MHz, CDCl3) δ: 8.88 (br, 1H), 8.42 (d, J = 5.2 Hz, 1H),7.85 (s, 1H), 7.84-7.81 (m, 2H), 7.54 (s, 1H), 7.47 (d, J = 8.0 Hz, 1H), 7.05-7.02 (m, 2H), 5.61-5.57 (m, 1H), 4.12- 4.03 (m, 2H), 3.91 (s, 3H), 3.81-3.60 (m, 3H), 3.38-3.04 (m, 6H), 2.95- 2.78 (m, 3H), 2.39-2.31 (m, 1H), 2.12-2.05 (m, 1H).149(R)-5-(tert-butyl)-N-(8- (2-((5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3- yl)amino)pyrimidin-4- yl)-2-(2-hydroxyethyl)- 2,3,4,5-tetrahydro-1H- benzo[c]azepin-5-yl)- 1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 558.2.1H NMR (400 MHz, CD3OD) δ: 8.35 (d, J = 5.2 Hz, 1H), 7.99-7.97 (m, 2H), 7.69 (s, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 5.59-5.55 (m, 1H), 4.16-4.10 (m, 4H), 3.74 (t, J = 6.0 Hz, 2H), 3.27-3.13 (m, 2H), 2.96 (t, J = 7.2 Hz, 2H), 2.67-2.62 (m, 4H), 2.33-2.24 (m, 1H), 2.00- 1.96 (m, 1H), 1.52 (s, 9H).1505-(tert-butyl)-N-(8- hydroxy-2-(2-((1-methyl 1H-pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,3,4-oxadiazole-2- carboxamldcESI-MS (M + H)+: 503.2.1H NMR (400 MHz, CDCl3) δ: 8.74 (d, J = 4.8 Hz, 1H), 7.88-7.85 (m, 3H), 7.76- 7.59 (m, 1H), 7.54 (s, 1H), 7.39-7.37 (m, 1H), 7.05-7.03 (m, 2H), 5.41- 5.37 (m, 1H), 4.14-4.09 (m, 1H), 3.91 (s, 3H), 3.89-3.84 (m, 1H), 3.26-3.19 (m, 2H), 2.24-1.96 (m, 4H), 1.49-1.44 (m, 9H).151(R)-5-(tert-butyl)-N-(2- (2-((1,5-dimethyl-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 501.3.1H NMR (400 MHz, CD3OD) δ: 8.31 (d, J = 5.2 Hz, 1H), 7.93-7.91 (m, 2H), 7.60 (s, 1H), 7.38 (d, J = 8.0 Hz, 1H), 7.23 (d, J = 5.2 Hz, 1H), 5.44 (d, J = 10.0 Hz, 1H), 3.82 (s, 3H), 3.10-2.97 (m, 2H), 2.25 (s, 3H), 2.09-1.86 (m, 5H), 1.54 (s, 9H), 1.49-1.43 (m, 1H).152(R)-5-(tert-butyl)-N-(2- (2-((5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 513.3.1H NMR (500 MHz, DMSO-d6) δ: 9.57 (d, J = 7.94 Hz, 1H), 9.06 (s, 1H), 8.40 (d, J = 5.49 Hz, 1H), 7.93-7.84 (m, 2H), 7.62 (br s, 1H), 7.32 (d, J = 7.94 Hz, 1H), 7.21 (d, J = 4.88 Hz, 1H), 5.26 (t, J = 8.85 Hz, 1H), 4.05 (t, J = 7.33 Hz, 2H), 3.17 (s, 4H), 2.97- 2.91 (m, 2H), 2.91-2.83 (m, 2H), 2.54-2.51 (m, 2H), 1.99-1.89 (m, 3H), 1.88-1.68 (m, 2H), 1.46 (s, 9H), 1.36-1.26 (m, 1H).153(R)-5-(tert-butyl)-N-(2- (2-((1-methyl-5- (trifluoromethyl)-1H- pyrazol-4- yl)amino)pyrimidin-4- yl)-6,7,8,9-tetrahydro- 5H-benzo[7]annulen-5- yl)-1,2,4-oxadiazole-3- carboxamideESI-MS (M + H)+: 555.3.1H NMR (500 MHz, DMSO-d6) δ: 9.56 (d, J = 8.55 Hz, 1H), 8.93 (s, 1H), 8.43 (d, J = 5.49 Hz, 1H), 7.89 (s, 1H), 7.86 (dd, J = 8.2, 1.5 Hz, 1H), 7.79 (s, 1H), 7.37-7.28 (m, 2H), 5.25 (br t, J = 8.85 Hz, 1H), 3.95 (s, 3 H), 2.99- 2.86 (m, 2H), 1.99-1.88 (m, 3H), 1.86-1.71 (m,2H), 1.46 (s, 9H), 1.35-1.25 (m, 1H).154(R)-1-(tert-butyl)-N-(2- (2-((1-methyl-1H- pyrazol-4- yl)amino)pyridin-4-yl)- 6,7,8,9-tetrahydro-5H- benzo[7]annulen-5-yl)- 1H-1,2,3-triazole-4- carboxamideESI-MS (M + H)+: 485.3.1H NMR (400 MHz, CDCl3) δ: 8.17-8.15 (m, 1H), 7.66 (d, J = 8.4 Hz, 1H), 7.61 (s, 1H), 7.47 (s, 1H), 7.37-7.30 (m, 3H), 6.87-6.86 (m, 1H), 6.70 (s, 1H), 6.13-6.09 (m, 1H), 5.41-5.38 (m, 1H), 3.91 (s, 3H), 3.03-2.92 (m, 2H), 2.03-1.93 (m, 4H), 1.87-1.84 (m, 1H), 1.71 (s, 9H), 1.63-1.55 (m, 1H).*indicates that the stereochemistry at the chiral center is arbitrarily assigned.**indicates that the relative stereochemistry at the two chiral centers for the racemic mixture is arbitrarily assigned, i.e., the trans- or cis-configuration at one chiral center relative to the other chiral center is arbitrarily assigned. Example 155. In Vitro BTK Kinase Assay: BTK-POLYGAT-LS ASSAY The purpose of the BTK in vitro assay is to determine compound potency against BTK through the measurement of IC50. Compound inhibition is measured after monitoring the amount of phosphorylation of a fluorescein-labeled polyGAT peptide (Invitrogen PV3611) in the presence of active BTK enzyme (Upstate 14-552), ATP, and inhibitor. The BTK kinase reaction was done in a black 96 well plate (costar 3694). For a typical assay, a 24 pL aliquot of a ATP/peptide master mix (final concentration; ATP 10 μM, polyGAT 100 nM) in kinase buffer (10 mM Tri s-HCl pH 7.5, 10 mM MgCl2, 200 μM Na3PO4, 5 mM DTT, 0.01% Triton X-100, and 0.2 mg/ml casein) is added to each well. Next, I pL of a 4-fold, 40×compound titration in 100% DMSO solvent is added, followed by adding 15 uL of BTK enzyme mix in 1×kinase buffer (with a final concentration of 0.25 nM). The assay is incubated for 30 minutes before being stopped with 28 pL of a 50 mM EDTA solution. Aliquots (5 uL) of the kinase reaction are transferred to a low volume white 384 well plate (Corning 3674), and 5 pL of a 2×detection buffer (Invitrogen PV3574, with 4 nM Tb-PY20 antibody, Invitrogen PV3552) is added. The plate is covered and incubated for 45 minutes at room temperature. Time resolved fluorescence (TRF) on Molecular Devices M5 (332 nm excitation; 488 nm emission; 518 nm fluorescein emission) is measured. IC50values are calculated using a four parameter fit with 100% enzyme activity determined from the DMSO control and 0% activity from the EDTA control. Table 1 shows the activity of selected compounds of this invention in the in vitro Btk kinase assay, wherein each compound number corresponds to the compound numbering set forth in Examples 1-154 herein. “†” represents an IC50of equal to or less than 1000 nM and greater than 10 nM; “††” represents an IC50of equal to or less than 10 nM and greater than 1 nM; and “†††” represents an IC50of equal to or less than 1 nM. TABLE 1IC50(nM)Compound No.†††1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14a, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24a,25, 26a, 26b, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36a, 37, 38, 39, 40, 41, 42a, 44,54, 55, 58, 59, 68, 70, 71, 72, 73, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 86, 87, 88,89a, 90, 91, 92, 93, 96, 97, 98, 99, 101, 102, 105, 106, 107, 111, 112, 114, 117,119a, 116, 121, 122, 124, 125, 127, 135, 149, 150, 152, 153,††43, 45, 46, 47, 49, 48, 50, 51, 52, 53, 56, 57, 60, 61, 62, 63, 64, 65, 66, 67, 69, 74,94, 95, 100, 103, 110, 113, 115, 123, 126, 128, 129, 130, 131,†14b, 24b, 36b, 85, 89b, 108, 109, 132, 133, 134, 136, 137, 138, 139, 141 Example 156. In Vitro PD Assay in Human Whole Blood Human heparinized venous blood was purchased from Bioreclamation, Inc. or SeraCare Life Sciences and shipped overnight. Whole blood was aliquoted into 96-well plate and “spiked” with serial dilutions of test compound in DMSO or with DMSO without drug. The final concentration of DMSO in all wells was 0.1%. The plate was incubated at 37° C. for 30 min. Lysis buffer containing protease and phosphatase inhibitors was added to the drug-containing samples and one of the DMSO-only samples (+PPi, high control), while lysis buffer containing protease inhibitors was added to the other DMSO-only samples (−PPi, low control). All of the lysed whole blood samples were subjected to the total BTK capture and phosphotyrosine detection method described in US20160311802, incorporated herein by reference. ECL values were graphed in Prism and a best-fit curve with restrictions on the maximum and minimum defined by the +PPi high and −PPi low controls was used to estimate the test compound concentration that results in 50% inhibition of ECL signal by interpolation. Table 2 shows the activity of selected compounds of this invention in the pBTK assay, wherein each compound number corresponds to the compound numbering set forth in Examples 1-154 described herein. “†” represents an IC50of equal to or less than 10,000 nM but greater than 500 nM, “††” represents an IC50of equal to or less than 500 nM but greater than 100 nM; and “†††” represents an IC50of equal to or less than 100 nM. TABLE 2IC50(nM)Compound No.†††2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14a, 15, 16, 17, 18, 19, 20, 21, 22, 23,24a, 25, 26a, 26b, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36a, 37, 39, 40, 41, 42a,44, 56, 58, 60, 62, 63, 64, 65, 67, 68, 71, 72, 73, 75, 76, 77, 78, 79, 80, 81,82, 83, 84, 86, 87, 88, 89a, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101,104, 105, 106, 107, 110, 111, 112, 113, 114, 119a, 120, 121, 122, 123, 124,125, 126, 152,††38, 42b, 45, 46, 47, 48, 49, 51, 52, 53, 57, 59, 61, 66, 69, 70, 74, 102, 103,115, 117, 118a, 118b, 127, 128, 129, 130, 131, 132†14b, 24b, 36b, 43, 50, 85, 89b, 108, 109, 116, 133, 134, 135, 136, 137, 138,139, 140, 141, 153,
358,973
11858927
DETAILED DESCRIPTION OF THE INVENTION In an embodiment, R5is not halogen. In a further embodiment, R1, R4, and R6, independently of each other, are H or alkyl, and in particular are H. In a still further embodiment, R10is H, alkyl or phenylalkyl wherein the phenyl group is optionally substituted with halogen, and in particular R10is H or alkyl. In a still further embodiment, A is —CO—. In a still further embodiment, Q is phenyl which is substituted as defined above. In a still further embodiment, the invention relates to a compound of the formula I and the pharmaceutically acceptable salts, prodrugs, esters, solvates and optical isomers thereof, wherein R1, R4to R6, R10, A and Q are as defined above in any combination. In a still further embodiment, the invention relates to a compound of formula (Ia), (Ib), (Ic) and (Id) and the pharmaceutically acceptable salts, prodrugs, esters, solvates and optical isomers thereof, wherein the variables are as defined in the embodiments above. In a further embodiment, at least one or at least two of Rx, Ryor Rzare halogen, and the other of Rx, Ryor Rzis H, halogen or alkyl, in particular alkyl or halogen. Halogen is preferably F or Cl. In a further embodiment, R1, R4and R6are H. In a further embodiment, R12is methyl, ethyl or propyl. In an embodiment, the invention relates to MKK4 inhibitors of formula (I) and (Ia) to (Id) and the pharmaceutically acceptable salts, prodrugs, solvates and optical isomers thereof, and in particular to MKK4 inhibitors which selectively inhibit protein kinase MKK4 over protein kinases JNK1 and MKK7. Further, the invention also relates to the compounds of the invention for use in inhibiting protein kinase MKK4 and in particular for use in selectively inhibiting protein kinase MKK4 over protein kinases JNK1 and MKK7. Further, the invention also relates to said compounds for use in promoting liver regeneration or reducing or preventing hepatocyte death and, at the same time, increasing hepatocyte proliferation. The invention also includes the pharmaceutically acceptable salts of the compounds mentioned above. The pharmaceutically acceptable salts are especially acid or base addition salts with pharmaceutically acceptable acids or bases. Examples of suitable pharmaceutically acceptable organic and inorganic acids are hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, sulfamic acid, C1-C4-alkylsulfonic acids, such as methanesulfonic acid, cycloaliphatic sulfonic acids, such as S-(+)-10-camphor sulfonic acid, aromatic sulfonic acids, such as benzenesulfonic acid and toluenesulfonic acid, di- and tricarboxylic acids and hydroxycarboxylic acids having 2 to 10 carbon atoms, such as oxalic acid, malonic acid, maleic acid, fumaric acid, lactic acid, tartaric acid, citric acid, glycolic acid, adipic acid and benzoic acid. Other utilizable acids are described, e.g., in Fortschritte der Arzneimittelforschung [Advances in drug research], Volume 10, pages 224 ff., Birkhäuser Verlag, Basel and Stuttgart, 1966. Examples of suitable pharmaceutically acceptable organic and inorganic bases are alkali metal hydroxides, such as sodium hydroxide or potassium hydroxide, alkaline earth metal hydroxides such as calcium or magnesium hydroxide, ammonium hydroxide, organic nitrogen bases such as dimethylamine, trimethylamine, ethanolamine, diethanolamine, triethanolamine, choline, 2-amino-2-hydroxymethyl-propane-1,3-diol, meglumine, procaine etc. L-arginine, L-lysine, ethylenediamine, or hydroxyethylpyrrolidine. The invention also includes any tautomeric, crystal and polymorphic form of the compounds and salts of the present invention and mixtures thereof. The invention also includes solvates such as hydrates. The compounds of the invention may contain one or more chiral centers, and exist in different optically active forms such enantiomers and diastereomers. As used herein, the term “pro-drug” refers to an agent which is converted into the parent drug in vivo by some physiological chemical process. An example, without limitation, of a pro-drug would be a compound of the present invention in the form of an ester. Pro-drugs have many useful properties. For example, a pro-drug may be more water soluble than the ultimate drug, thereby facilitating intravenous administration of the drug. A pro-drug may also have a higher level of oral bioavailability than the ultimate drug. After administration, the prodrug is enzymatically or chemically cleaved to deliver the ultimate drug in the blood or tissue. Exemplary pro-drugs include, but are not limited to, compounds with carboxylic acid substituents wherein the free hydrogen is replaced by (C1-C4)alkyl, (C1-C12)alkanoyloxy-methyl, (C4-C9)1-(alkanoyloxy)ethyl, 1-methyl-1-(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1-(alkoxycarbonyl-oxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1-(alkoxycarbonyloxy)-ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N-(alkoxycarbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crotono-lactonyl, gamma-butyrolacton-4-yl, di-N,N—(C1-C2)alkylamino(C2-C3)alkyl (such as β-dimethylaminoethyl), carbamoyl-(C1-C2)alkyl, N,N-di(C1-C2)-alkylcarbamoyl-(C1-C2)alkyl and piperidino-, pyrrolidino- or morpholino(C2-C3)alkyl. Other exemplary pro-drugs release an alcohol of Formula (I) wherein the free hydrogen of the hydroxyl substituent (e.g., R group contains hydroxyl) is replaced by (C1-C6)alkanoyloxy-methyl, 1-((C1-C6)alkanoyloxy)-ethyl, 1-methyl-1-((C1-C6)alkanoyloxy)ethyl, (C1-C12)alkoxy-carbonyloxy-methyl, N—(C1-C6)-alkoxy-carbonylaminomethyl, succinoyl, (C1-C6)alkanoyl, α-amino(C1-C4)alkanoyl, arylactyl and α-aminoacyl, or α-aminoacyl-α-aminoacyl wherein said α-aminoacyl moieties are independently any of the naturally occurring L-amino acids found in proteins, P(O)(OH)2, —P(O)(O(C1-C6)alkyl)2or glycosyl (the radical resulting from detachment of the hydroxyl of the hemiacetal of a carbohydrate). The expression MKK4 inhibitor means that the kinase activity of MKK4 is inhibited with an IC50of <10 μmol/l, preferably <1 μmol/l, and in particular <0.5 μmol/l. The expression “selectively inhibit protein kinase MKK4 over protein kinases JNK1 and MKK7” as used herein means that the ratio of MKK7 inhibiting activity to MKK4 inhibiting activity or the ratio of JNK1 inhibiting activity to MKK4 inhibiting activity, expressed as either percent of control or Kd, is ≥10, as measured with KINOMEscan™. The expression “promoting liver regeneration or reducing or preventing hepatocyte death” as used herein means an increase in the relative number of proliferating hepatocytes by at least 30%, preferably at least 50%, as compared to the number of proliferating cells at the beginning of therapy. In particular, the expression means an increase by 100% when compared to the number of proliferating cells at the beginning of therapy. In this context, the experimental determination and quantification will be performed using standard methods, e.g. the quantification of the protein Ki67, which is strictly associated with cell proliferation. For quantification of proliferating hepatocytes in a tissue slide, several immunohistochemical standard methods are available, which use a primary anti-Ki67 antibody followed by visualization of anti-Ki67-binding by using, for example, a horseradish peroxidase conjugated secondary antibody. The amount of peroxidase activity, which is visualized by enzymatic conversion of chromogenic substrates, correlates with the amount of Ki67 protein and the number of proliferating cells. In the experiments described below, hepatocyte proliferation was quantified by Ki67-staining using the primary polyclonal rabbit anti-Ki67 antibody from Abcam (article no. ab15580, Abcam, Cambridge, USA) and the fluorophore tetramethylrhodamine containing secondary goat polyclonal antibody from Invitrogen (article no. 16101, Invitrogen/ThermoFisher). Based on data obtained from several preclinical mouse models it was found that shRNA (small hairpin RNA) mediated suppression of MKK4 in a chronic CCl4(carbon tetrachloride) mediated liver damage mouse model increased hepatocyte proliferation from 13% to 27% (compared to a control shRNA) and was associated with decreased liver damage (transaminases) and decreased liver fibrosis. According to the definition in the previous chapter, the relative increase of proliferating cells was 108%. In a model of alcohol induced steatohepatitis (ASH), shRNA mediated silencing of MKK4 resulted in a hepatocyte proliferation rate of 4% as compared to 2% when a control shRNA was used (relative increase: 100%). The duplication of hepatocyte proliferation was associated with decreased steatosis (fat deposition) and decreased liver damage as measured by transaminases. Along the same lines, shRNA mediated MKK4 silencing increased hepatocyte proliferation from 16% (control shRNA) to 33% (relative increase: 106%) in a model of partial hepatectomy (48 hrs after surgical removal of two thirds of the liver). Again, increased hepatocyte proliferation was associated with improved liver regeneration and a faster restoration of liver mass. In conclusion, these studies validate MKK4 as a therapeutic target for treatment of acute and chronic liver diseases. Furthermore, WO 2018/134254 discloses new compounds, which inhibit MKK4 selectively over MKK7 and JNK1. In experimental in vitro and in vivo models of liver regeneration, these compounds were effective in the prevention of acute liver failure induced by administration of a Jo2 antibody and induced the proliferation of isolated primary mouse hepatocytes. The new compounds disclosed in the present application are potent MKK4 inhibitors with selectivity against MKK7 and JNK1 and therefore, in analogy to the compounds disclosed in WO 2018/134254 can be used for treatment of liver disease and for promoting liver regeneration or reducing or preventing hepatocyte death. The organic moieties mentioned in the above definitions of the variables are—like the term halogen—collective terms for individual listings of the individual group members. The prefix Cn-Cmindicates in each case the possible number of carbon atoms in the group. The term halogen denotes in each case fluorine, bromine, chlorine or iodine, in particular fluorine or chlorine and preferably fluorine. Alkyl is a straight-chain or branched alkyl group which is preferably a C1-C6-alkyl group, i.e. an alkyl group having from 1 to 6 carbon atoms, and more preferably a C1-C4-alkyl group and in particular a C1-C3-alkyl group. Examples of an alkyl group are methyl, ethyl, n-propyl, iso-propyl, n-butyl, 2-butyl, iso-butyl, tert-butyl, pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 1-methylpentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, 1-ethylbutyl, 2-ethylbutyl, 1,1,2-trimethylpropyl, 1,2,2-trimethylpropyl, 1-ethyl-1-methylpropyl and 1-ethyl-2-methylpropyl. The definition of alkyl is likewise applicable to any group which includes an alkyl group. Haloalkyl is a halogenated alkyl group as defined above, wherein at least one, e.g. 1, 2, 3, 4 or all of the hydrogen atoms are replaced by 1, 2, 3, 4 or a corresponding number of identical or different halogen atoms, such as trifluoromethyl, chloromethyl, bromomethyl, difluoromethyl, fluoromethyl, difluoroethyl, etc. Particular examples include the fluorinated C1-C4alkyl groups as defined, such as trifluoromethyl, difluoromethyl, fluoromethyl, or difluoroethyl. Cycloalkyl is a cycloaliphatic radical which is preferably C3-C5-cycloalkyl, i.e. a cycloalkyl group having from 3 to 8 carbon atoms. In particular, 3 to 6 carbon atoms form the cyclic structure, such as cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The cyclic structure may be unsubstituted or may carry 1, 2, 3 or 4 C1-C4alkyl radicals, preferably one or more methyl radicals. Carbonyl is >C═O. Aminocarbonyl is NH2C(O)—. Alkenyl is a singly unsaturated hydrocarbon radical which is preferably a C2-C6-alkenyl group, i.e. an alkenyl group having 2, 3, 4, 5 or 6 carbon atoms, e.g. vinyl, allyl (2-propen-1-yl), 1-propen-1-yl, 2-propen-2-yl, methallyl (2-methylprop-2-en-1-yl) and the like. C3-C5-Alkenyl is, in particular, allyl, 1-methylprop-2-en-1-yl, 2-buten-1-yl, 3-buten-1-yl, methallyl, 2-penten-1-yl, 3-penten-1-yl, 4-penten-1-yl, 1-methylbut-2-en-1-yl or 2-ethylprop-2-en-1-yl, 2-hexen-1-yl. Alkinyl is a singly unsaturated hydrocarbon radical which is preferably a C2-C6-alkinyl group, i.e. an alkinyl group having 2, 3, 4, 5 or 6 carbon atoms, e.g. ethynyl, 2-propyn-1-yl, 1-propyn-1-yl, 2-propyn-2-yl and the like. C3-C5-Alkinyl is, in particular, 2-propyn-1-yl, 2-butyn-1-yl, 3-butyn-1-yl, 2-pentyn-1-yl, 3-pentyn-1-yl, 4-pentyn-1-yl. Alkylene is straight-chain or branched alkylene group which is preferably a C1-C5-alkylene group, i.e. an alkylene group having from 1 to 5 carbon atoms. Alkylene groups having 2 to 4 and in particular 2 to 3 carbon atoms are especially preferred. Examples include methylene, ethylene and 1-methylethylene. A further example is propylene. Another further example is butylene. The definition of alkylene is likewise applicable to any group which includes an alkylene group. Heteroalkylene is a straight-chain or branched alkyl group having 1, 2 or 3 heteroatoms which are selected from oxygen, nitrogen and sulfur. Examples for heteroalkylene are alkyloxyalkyl, alkylaminoalkyl, dialkylaminoalkyl or alkylthioalkyl. Any alkyl or alkylene group is as defined above. Alkyloxyalkyl is preferred. Alkenylene is straight-chain or branched alkenylene group which is preferably a C2-C4-alkenylene group, i.e. an alkenylene group having from 2 to 4 carbon atoms. Examples include vinyl and propenyl. Alkinylene is straight-chain or branched alkinylene group which is preferably a C2-C4-alkinylene group, i.e. an alkinylene group having from 2 to 4 carbon atoms. Examples include propynylene. Aryl (or aromatic group) is a 6- to 12-membered, in particular 6- to 10-membered, aromatic cyclic radical which can be a monocyclic aromatic ring, for example, phenyl etc., or a fused polycyclic aromatic ring comprising a first monocyclic aromatic ring and one or more carbocycles which are saturated, partially unsaturated or aromatic, for example, naphthyl, indenyl, tetrahydronaphthyl, indanyl. A heteroaromatic (or heteroaryl) group is a 5- or 6-membered monocyclic or 9- or 10-membered bicyclic aromatic group having 1, 2 or 3 heteroatoms selected from O, N or S. The heteroaryl or heteroaromatic group may be bound to the neighboring group via a carbon atom (C-bound) or via a nitrogen heteroatom (N-bound). The heterocyclic radicals may be bound via a carbon atom (C-bound) or a nitrogen atom (N-bound). Preferred heteroaromatic radicals comprise 1 nitrogen atom as ring member atom and optionally 1 or 2 further heteroatoms as ring members, which are selected, independently of each other from O, S and N. Examples are: C-bound, 5-membered, heteroaromatic rings: 2-furyl, 3-furyl, 5-furyl, 2-thienyl, 3-thienyl, 5-thienyl, pyrrol-2-yl, pyrrol-3-yl, pyrrol-5-yl, pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl, isothiazol-3-yl, isothiazol-4-yl, isothiazol-5-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, 1,2,3-oxadiazol-imidazol-4-yl,4-yl, 1,2,3-oxadiazol-5-yl, 1,2,4-oxadiazol-3-yl, 1,2,4,-oxadiazol-5-yl, 1,3,4-oxadiazol-2-yl, 1,2,3-thiadiazol-4-yl, 1,2,3-thiadiazol-5-yl, 1,2,4-thiadiazol-3-yl, 1,2,4-thiadiazol-5-yl, 1,3,4-thiadiazolyl-2-yl, 1,2,3-triazol-4-yl, 1,2,4-triazol-3-yl, tetrazol-5-yl; C-bound, 6-membered, heteroaromatic rings: pyridin-2-yl, pyridin-3-yl (3-pyridyl), pyridin-4-yl (4-pyridyl), pyridin-5-yl, pyridazin-3-yl, pyridazin-4-yl, pyridazin-6-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyrazin-2-yl, pyrazin-5-yl, 1,3,5-triazin-2-yl, 1,2,4-triazin-3-yl, 1,2,4-triazin-5-yl, 1,2,4-triazin-6-yl, 1,2,4,5-tetrazin-3-yl; N-bound, 5-membered, heteroaromatic rings: pyrrol-1-yl, pyrazol-1-yl, imidazol-1-yl, 1,2,3-triazol-1-yl, 1,2,4-triazol-1-yl. Bicyclic heteroaromatic groups include one of the described 5- or 6-membered heteroaromatic rings and a further anellated, saturated or unsaturated or aromatic carbocycle, such as a benzene, cyclohexane, cyclohexene or cyclohexadiene ring. Examples are quinolinyl, isoquinolinyl, indolyl, indolizinyl, isoindolyl, 4-, 5-, 6- or 7-azaindole, indazolyl, benzofuryl, benzthienyl, benzo[b]thiazolyl, benzoxazolyl, benzthiazolyl, benzimidazolyl, imidazo[b]thiazolyl, thieno[b]pyridyl, imidazo[a]pyridyl, pyrazo[a]pyridyl and pyrrol[d]pyrimidyl. Examples of 5- or 6-membered heteroaromatic compounds comprising an anellated cycloalkenyl ring include dihydroindolyl, dihydroindolizinyl, dihydroisoindolyl, dihydroquinolinyl, dihydroisoquinolinyl, dihydrobenzofuryl, chromenyl, chromanyl, dihydropyrrol[a]imidazolyl and tetrahydrobenzothiazolyl. A non-aromatic 5- or 6-membered group (heterocyclic group) may be saturated or partially unsaturated and includes 1, 2 or 3 heteroatoms selected from O, N and S. The heterocyclic radicals may be bound via a carbon atom (C-bound) or a nitrogen atom (N-bound). Preferred heterocyclic groups comprise 1 nitrogen atom as ring member atom and optionally 1 or 2 further heteroatoms as ring members, which are selected, independently of each other from O, S and N. Examples are: C-bound, 5-membered, saturated rings, such as tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, tetrahydropyrrol-2-yl, tetrahydropyrrol-3-yl, tetrahydropyrazol-3-yl, tetrahydro-pyrazol-4-yl, tetrahydroisoxazol-3-yl, tetrahydroisoxazol-4-yl, tetrahydroisoxazol-5-yl, 1,2-oxathiolan-3-yl, 1,2-oxathiolan-4-yl, 1,2-oxathiolan-5-yl, tetrahydroisothiazol-3-yl, tetrahydroisothiazol-4-yl, tetrahydroisothiazol-5-yl, 1,2-dithiolan-3-yl, 1,2-dithiolan-4-yl, tetrahydroimidazol-2-yl, tetrahydroimidazol-4-yl, tetrahydrooxazol-2-yl, tetrahydrooxazol-4-yl, tetrahydrooxazol-5-yl, tetrahydrothiazol-2-yl, tetrahydrothiazol-4-yl, tetrahydrothiazol-5-yl, 1,3-dioxolan-2-yl, 1,3-dioxolan-4-yl, 1,3-oxathiolan-2-yl, 1,3-oxathiolan-4-yl, 1,3-oxathiolan-5-yl, 1,3-dithiolan-2-yl, 1,3-dithiolan-4-yl, 1,3,2-dioxathiolan-4-yl; C-bound, 6-membered, saturated rings, such as tetrahydropyran-2-yl, tetrahydropyran-3-yl, tetrahydropyran-4-yl, piperidin-2-yl, piperidin-3-yl, piperidin-4-yl, tetrahydrothiopyran-2-yl, tetrahydrothiopyran-3-yl, tetrahydrothiopyran-4-yl, 1,3-dioxan-2-yl, 1,3-dioxan-4-yl, 1,3-dioxan-5-yl, 1,4-dioxan-2-yl, 1,3-dithian-2-yl, 1,3-dithian-4-yl, 1,3-dithian-5-yl, 1,4-dithian-2-yl, 1,3-oxathian-2-yl, 1,3-oxathian-4-yl, 1,3-oxathian-5-yl, 1,3-oxathian-6-yl, 1,4-oxathian-2-yl, 1,4-oxathian-3-yl, 1,2-dithian-3-yl, 1,2-dithian-4-yl, hexahydropyrimidin-2-yl, hexahydropyrimidin-4-yl, hexahydropyrimidin-5-yl, hexahydropyrazin-2-yl, hexahydropyridazin-3-yl, hexahydropyridazin-4-yl, tetrahydro-1,3-oxazin-2-yl, tetrahydro-1,3-oxazin-4-yl, tetrahydro-1,3-oxazin-5-yl, tetrahydro-1,3-oxazin-6-yl, tetrahydro-1,3-thiazin-2-yl, tetrahydro-1,3-thiazin-4-yl, tetrahydro-1,3-thiazin-5-yl, tetrahydro-1,3-thiazin-6-yl, tetrahydro-1,4-thiazin-2-yl, tetrahydro-1,4-thiazin-3-yl, tetrahydro-1,4-oxazin-2-yl, tetrahydro-1,4-oxazin-3-yl, tetrahydro-1,2-oxazin-3-yl, tetrahydro-1,2-oxazin-4-yl, tetrahydro-1,2-oxazin-5-yl, tetrahydro-1,2-oxazin-6-yl; N-bound, 5-membered, saturated rings, such as tetrahydropyrrol-1-yl (pyrrolidin-1-yl), tetrahydropyrazol-1-yl, tetrahydroisoxazol-2-yl, tetrahydroisothiazol-2-yl, tetrahydroimidazol-1-yl, tetrahydrooxazol-3-yl, tetrahydrothiazol-3-yl; N-bound, 6-membered, saturated rings, such as piperidin-1-yl, hexahydropyrimidin-1-yl, hexahydropyrazin-1-yl (piperazin-1-yl), hexahydro-pyridazin-1-yl, tetrahydro-1,3-oxazin-3-yl, tetrahydro-1,3-thiazin-3-yl, tetrahydro-1,4-thiazin-4-yl, tetrahydro-1,4-oxazin-4-yl (morpholin-1-yl), tetrahydro-1,2-oxazin-2-yl; C-bound, 5-membered, partially unsaturated rings, such as 2,3-dihydrofuran-2-yl, 2,3-dihydrofuran-3-yl, 2,5-dihydrofuran-2-yl, 2,5-di-hydrofuran-3-yl, 4,5-dihydrofuran-2-yl, 4,5-dihydrofuran-3-yl, 2,3-dihydro-thien-2-yl, 2,3-dihydrothien-3-yl, 2,5-dihydrothien-2-yl, 2,5-dihydrothien-3-yl, 4,5-dihydrothien-2-yl, 4,5-dihydrothien-3-yl, 2,3-dihydro-1H-pyrrol-2-yl, 2,3-dihydro-1H-pyrrol-3-yl, 2,5-dihydro-1H-pyrrol-2-yl, 2,5-dihydro-1H-pyrrol-3-yl, 4,5-dihydro-1H-pyrrol-2-yl, 4,5-dihydro-1H-pyrrol-3-yl, 3,4-dihydro-2H-pyrrol-2-yl, 3,4-dihydro-2H-pyrrol-3-yl, 3,4-dihydro-5H-pyrrol-2-yl, 3,4-dihydro-5H-pyrrol-3-yl, 4,5-dihydro-1H-pyrazol-3-yl, 4,5-dihydro-1H-pyrazol-4-yl, 4,5-dihydro-1H-pyrazol-5-yl, 2,5-dihydro-1H-pyrazol-3-yl, 2,5-dihydro-1H-pyrazol-4-yl, 2,5-dihydro-1H-pyrazol-5-yl, 4,5-dihydroisoxazol-3-yl, 4,5-dihydroisoxazol-4-yl, 4,5-dihydroisoxazol-5-yl, 2,5-dihydroisoxazol-3-yl, 2,5-dihydroisoxazol-4-yl, 2,5-dihydroisoxazol-5-yl, 2,3-dihydroisoxazol-3-yl, 2,3-dihydroisoxazol-4-yl, 2,3-dihydroisoxazol-5-yl, 4,5-dihydroisothiazol-3-yl, 4,5-dihydroisothiazol-4-yl, 4,5-dihydroisothiazol-5-yl, 2,5-dihydroisothiazol-3-yl, 2,5-dihydroisothiazol-4-yl, 2,5-dihydroisothiazol-5-yl, 2,3-dihydroisothiazol-3-yl, 2,3-dihydroisothiazol-4-yl, 2,3-dihydroisothiazol-5-yl, 4,5-dihydro-1H-imidazol-2-yl, 4,5-dihydro-1H-imidazol-4-yl, 4,5-dihydro-1H-imidazol-5-yl, 2,5-dihydro-1H-imidazol-2-yl, 2,5-dihydro-1H-imidazol-4-yl, 2,5-dihydro-1H-imidazol-5-yl, 2,3-dihydro-1H-imidazol-2-yl, 2,3-dihydro-1H-imidazol-4-yl, 4,5-dihydro-oxazol-2-yl, 4,5-dihydrooxazol-4-yl, 4,5-dihydrooxazol-5-yl, 2,5-dihydrooxazol-2-yl, 2,5-dihydrooxazol-4-yl, 2,5-dihydrooxazol-5-yl, 2,3-dihydrooxazol-2-yl, 2,3-dihydrooxazol-4-yl, 2,3-dihydrooxazol-5-yl, 4,5-dihydrothiazol-2-yl, 4,5-dihydrothiazol-4-yl, 4,5-dihydrothiazol-5-yl, 2,5-dihydrothiazol-2-yl, 2,5-dihydrothiazol-4-yl, 2,5-dihydrothiazol-5-yl, 2,3-dihydrothiazol-2-yl, 2,3-dihydrothiazol-4-yl, 2,3-dihydrothiazol-5-yl, 1,3-dioxol-2-yl, 1,3-dioxol-4-yl, 1,3-dithiol-2-yl, 1,3-dithiol-4-yl, 1,3-oxathiol-2-yl, 1,3-oxathiol-4-yl, 1,3-oxathiol-5-yl; C-bound, 6-membered, partially unsaturated rings, such as 2H-3,4-dihydropyran-6-yl, 2H-3,4-dihydropyran-5-yl, 2H-3,4-dihydropyran-4-yl, 2H-3,4-dihydropyran-3-yl, 2H-3,4-dihydropyran-2-yl, 2H-3,4-dihydrothiopyran-6-yl, 2H-3,4-dihydrothiopyran-5-yl, 2H-3,4-dihydrothiopyran-4-yl, 2H-3,4-dihydrothiopyran-3-yl, 2H-3,4-dihydrothiopyran-2-yl, 1,2,3,4-tetrahydropyridin-6-yl, 1,2,3,4-tetrahydropyridin-5-yl, 1,2,3,4-tetrahydropyridin-4-yl, 1,2,3,4-tetra-hydropyridin-3-yl, 1,2,3,4-tetrahydropyridin-2-yl, 2H-5,6-dihydropyran-2-yl, 2H-5,6-dihydropyran-3-yl, 2H-5,6-dihydropyran-4-yl, 2H-5,6-dihydropyran-5-yl, 2H-5,6-dihydropyran-6-yl, 2H-5,6-dihydrothiopyran-2-yl, 2H-5,6-dihydrothiopyran-3-yl, 2H-5,6-dihydrothiopyran-4-yl, 2H-5,6-dihydrothiopyran-5-yl, 2H-5,6-dihydrothiopyran-6-yl, 1,2,5,6-tetrahydropyridin-2-yl, 1,2,5,6-tetrahydropyridin-3-yl, 1,2,5,6-tetrahydropyridin-4-yl, 1,2,5,6-tetrahydropyridin-5-yl, 1,2,5,6-tetrahydropyridin-6-yl, 2,3,4,5-tetrahydropyridin-2-yl, 2,3,4,5-tetrahydropyridin-3-yl, 2,3,4,5-tetrahydropyridin-4-yl, 2,3,4,5-tetrahydropyridin-5-yl, 2,3,4,5-tetrahydropyridin-6-yl, 4H-pyran-2-yl, 4H-pyran-3-yl-, 4H-pyran-4-yl, 4H-thiopyran-2-yl, 4H-thiopyran-3-yl, 4H-thiopyran-4-yl, 1,4-dihydropyridin-2-yl, 1,4-dihydropyridin-3-yl, 1,4-dihydropyridin-4-yl, 2H-pyran-2-yl, 2H-pyran-3-yl, 2H-pyran-4-yl, 2H-pyran-5-yl, 2H-pyran-6-yl, 2H-thiopyran-2-yl, 2H-thiopyran-3-yl, 2H-thiopyran-4-yl, 2H-thiopyran-5-yl, 2H-thiopyran-6-yl, 1,2-dihydropyridin-2-yl, 1,2-dihydro-pyridin-3-yl, 1,2-dihydropyridin-4-yl, 1,2-dihydropyridin-5-yl, 1,2-dihydro-pyridin-6-yl, 3,4-dihydropyridin-2-yl, 3,4-dihydropyridin-3-yl, 3,4-dihydro-pyridin-4-yl, 3,4-dihydropyridin-5-yl, 3,4-dihydropyridin-6-yl, 2,5-dihydropyridin-2-yl, 2,5-dihydropyridin-3-yl, 2,5-dihydropyridin-4-yl, 2,5-dihydropyridin-5-yl, 2,5-dihydropyridin-6-yl, 2,3-dihydropyridin-2-yl, 2,3-dihydropyridin-3-yl, 2,3-dihydropyridin-4-yl, 2,3-dihydro-pyridin-5-yl, 2,3-dihydropyridin-6-yl, 2H-5,6-dihydro-1,2-oxazin-3-yl, 2H-5,6-dihydro-1,2-oxazin-4-yl, 2H-5,6-dihydro-1,2-oxazin-5-yl, 2H-5,6-dihydro-1,2-oxazin-6-yl, 2H-5,6-dihydro-1,2-thiazin-3-yl, 2H-5,6-dihydro-1,2-thiazin-4-yl, 2H-5,6-dihydro-1,2-thiazin-5-yl, 2H-5,6-dihydro-1,2-thiazin-6-yl, 4H-5,6-dihydro-1,2-oxazin-3-yl, 4H-5,6-dihydro-1,2-oxazin-4-yl, 4H-5,6-dihydro-1,2-oxazin-5-yl, 4H-5,6-dihydro-1,2-oxazin-6-yl, 4H-5,6-dihydro-1,2-thiazin-3-yl, 4H-5,6-dihydro-1,2-thiazin-4-yl, 4H-5,6-dihydro-1,2-thiazin-5-yl, 4H-5,6-dihydro-1,2-thiazin-6-yl, 2H-3,6-dihydro-1,2-oxazin-3-yl, 2H-3,6-dihydro-1,2-oxazin-4-yl, 2H-3,6-dihydro-1,2-oxazin-5-yl, 2H-3,6-dihydro-1,2-oxazin-6-yl, 2H-3,6-dihydro-1,2-thiazin-3-yl, 2H-3,6-dihydro-1,2-thiazin-4-yl, 2H-3,6-dihydro-1,2-thiazin-5-yl, 2H-3,6-dihydro-1,2-thiazin-6-yl, 2H-3,4-dihydro-1,2-oxazin-3-yl, 2H-3,4-dihydro-1,2-oxazin-4-yl, 2H-3,4-dihydro-1,2-oxazin-5-yl, 2H-3,4-dihydro-1,2-oxazin-6-yl, 2H-3,4-dihydro-1,2-thiazin-3-yl, 2H-3,4-dihydro-1,2-thiazin-4-yl, 2H-3,4-dihydro-1,2-thiazin-5-yl, 2H-3,4-dihydro-1,2-thiazin-6-yl, 2,3,4,5-tetrahydropyridazin-3-yl, 2,3,4,5-tetrahydropyridazin-4-yl, 2,3,4,5-tetrahydropyridazin-5-yl, 2,3,4,5-tetrahydro-pyridazin-6-yl, 3,4,5,6-tetrahydropyridazin-3-yl, 3,4,5,6-tetrahydropyridazin-4-yl, 1,2,5,6-tetrahydropyridazin-3-yl, 1,2,5,6-tetrahydropyridazin-4-yl, 1,2,5,6-tetra-hydropyridazin-5-yl, 1,2,5,6-tetrahydropyridazin-6-yl, 1,2,3,6-tetrahydro-pyridazin-3-yl, 1,2,3,6-tetrahydropyridazin-4-yl, 4H-5,6-dihydro-1,3-oxazin-2-yl, 4H-5,6-dihydro-1,3-oxazin-4-yl, 4H-5,6-dihydro-1,3-oxazin-5-yl, 4H-5,6-dihydro-1,3-oxazin-6-yl, 4H-5,6-dihydro-1,3-thiazin-2-yl, 4H-5,6-dihydro-1,3-thiazin-4-yl, 4H-5,6-dihydro-1,3-thiazin-5-yl, 4H-5,6-dihydro-1,3-thiazin-6-yl, 3,4,5-6-tetrahydropyrimidin-2-yl, 3,4,5,6-tetrahydropyrimidin-4-yl, 3,4,5,6-tetrahydropyrimidin-5-yl, 3,4,5,6-tetrahydropyrimidin-6-yl, 1,2,3,4-tetrahydropyrazin-2-yl, 1,2,3,4-tetrahydropyrazin-5-yl, 1,2,3,4-tetrahydro-pyrimidin-2-yl, 1,2,3,4-tetrahydropyrimidin-4-yl, 1,2,3,4-tetrahydropyrimidin-5-yl, 1,2,3,4-tetrahydropyrimidin-6-yl, 2,3-dihydro-1,4-thiazin-2-yl, 2,3-dihydro-1,4-thiazin-3-yl, 2,3-dihydro-1,4-thiazin-5-yl, 2,3-dihydro-1,4-thiazin-6-yl, 2H-1,3-oxazin-2-yl, 2H-1,3-oxazin-4-yl, 2H-1,3-oxazin-5-yl, 2H-1,3-oxazin-6-yl, 2H-1,3-thiazin-2-yl, 2H-1,3-thiazin-4-yl, 2H-1,3-thiazin-5-yl, 2H-1,3-thiazin-6-yl, 4H-1,3-oxazin-2-yl, 4H-1,3-oxazin-4-yl, 4H-1,3-oxazin-5-yl, 4H-1,3-oxazin-6-yl, 4H-1,3-thiazin-2-yl, 4H-1,3-thiazin-4-yl, 4H-1,3-thiazin-5-yl, 4H-1,3-thiazin-6-yl, 6H-1,3-oxazin-2-yl, 6H-1,3-oxazin-4-yl, 6H-1,3-oxazin-5-yl, 6H-1,3-oxazin-6-yl, 6H-1,3-thiazin-2-yl, 6H-1,3-oxazin-4-yl, 6H-1,3-oxazin-5-yl, 6H-1,3-thiazin-6-yl, 2H-1,4-oxazin-2-yl, 2H-1,4-oxazin-3-yl, 2H-1,4-oxazin-5-yl, 2H-1,4-oxazin-6-yl, 2H-1,4-thiazin-2-yl, 2H-1,4-thiazin-3-yl, 2H-1,4-thiazin-5-yl, 2H-1,4-thiazin-6-yl, 4H-1,4-oxazin-2-yl, 4H-1,4-oxazin-3-yl, 4H-1,4-thiazin-2-yl, 4H-1,4-thiazin-3-yl, 1,4-dihydropyridazin-3-yl, 1,4-dihydropyridazin-4-yl, 1,4-dihydropyridazin-5-yl, 1,4-dihydropyridazin-6-yl, 1,4-dihydropyrazin-2-yl, 1,2-dihydropyrazin-2-yl, 1,2-dihydropyrazin-3-yl, 1,2-dihydropyrazin-5-yl, 1,2-dihydropyrazin-6-yl, 1,4-dihydropyrimidin-2-yl, 1,4-dihydropyrimidin-4-yl, 1,4-dihydropyrimidin-5-yl, 1,4-dihydropyrimidin-6-yl, 3,4-dihydropyrimidin-2-yl, 3,4-dihydropyrimidin-4-yl, 3,4-dihydropyrimidin-5-yl or 3,4-dihydropyrimidin-6-yl; N-bound, 5-membered, partially unsaturated rings, such as 2,3-dihydro-1H-pyrrol-1-yl, 2,5-dihydro-1H-pyrrol-1-yl, 4,5-dihydro-1H-pyrazol-1-yl, 2,5-dihydro-1H-pyrazol-1-yl, 2,3-dihydro-1H-pyrazol-1-yl, 2,5-dihydroisoxazol-2-yl, 2,3-dihydroisoxazol-2-yl, 2,5-dihydroisothiazol-2-yl, 2,3-dihydroisoxazol-2-yl, 4,5-dihydro-1H-imidazol-1-yl, 2,5-dihydro-1H-imidazol-1-yl, 2,3-dihydro-1H-imidazol-1-yl, 2,3-dihydrooxazol-3-yl, 2,3-dihydrothiazol-3-yl; N-bound, 6-membered, partially unsaturated rings, such as 1,2,3,4-tetrahydropyridin-1-yl, 1,2,5,6-tetrahydropyridin-1-yl, 1,4-dihydro-pyridin-1-yl, 1,2-dihydropyridin-1-yl, 2H-5,6-dihydro-1,2-oxazin-2-yl, 2H-5,6-dihydro-1,2-thiazin-2-yl, 2H-3,6-dihydro-1,2-oxazin-2-yl, 2H-3,6-dihydro-1,2-thiazin-2-yl, 2H-3,4-dihydro-1,2-oxazin-2-yl, 2H-3,4-dihydro-1,2-thiazin-2-yl, 2,3,4,5-tetrahydropyridazin-2-yl, 1,2,5,6-tetrahydropyridazin-1-yl, 1,2,5,6-tetrahydropyridazin-2-yl, 1,2,3,6-tetrahydropyridazin-1-yl, 3,4,5,6-tetrahydropyrimidin-3-yl, 1,2,3,4-tetrahydropyrazin-1-yl, 1,2,3,4-tetrahydropyrimidin-1-yl, 1,2,3,4-tetrahydropyrimidin-3-yl, 2,3-dihydro-1,4-thiazin-4-yl, 2H-1,2-oxazin-2-yl, 2H-1,2-thiazin-2-yl, 4H-1,4-oxazin-4-yl, 4H-1,4-thiazin-4-yl, 1,4-dihydropyridazin-1-yl, 1,4-dihydropyrazin-1-yl, 1,2-dihydropyrazin-1-yl, 1,4-dihydropyrimidin-1-yl or 3,4-dihydropyrimidin-3-yl. Any group containing heteroatoms may contain 1, 2 or 3 heteroatoms which may be the same or different. The compounds of the invention can be prepared as disclosed in WO 2010/111527 which is incorporated herein in its entirety by reference or according to analogous procedures. The acid or base addition salts are prepared in a customary manner by mixing the free base with a corresponding acid or by mixing the free acid with the desired base. Optionally, the reaction is carried out in solution in an organic solvent, for example a lower alcohol, such as MeOH, ethanol or propanol, an ether, such as methyl tert-butyl ether or diisopropyl ether, a ketone, such as acetone or methyl ethyl ketone, or an ester, such as EtOAc. The compounds of the invention are useful for promoting liver regeneration or reducing or preventing hepatocyte death and, at the same time, increasing hepatocyte proliferation. The compounds are therefore useful in treating, modulating, improving or preventing diseases which involve acute or chronic damages to the liver that may be caused by infection, injury, exposure to toxic compounds, an abnormal build-up of normal substances in the blood, an autoimmune process, a genetic defect or unknown causes. Such liver diseases comprise all diseases where increased liver regeneration and reduction or prevention of hepatocyte death may be helpful to achieve a potential therapeutic effect, i.e. partial or complete restoration of liver functions. Such diseases compriseacute and chronic or acute on chronic liver diseases such as acute and chronic viral hepatitis like hepatitis B, C, E, hepatitis caused by Epstein-Barr virus, cytomegalovirus, herpes simplex virus and other viruses, all types of autoimmune hepatitis, primary sclerosing hepatitis, alcoholic hepatitis;metabolic liver diseases such as metabolic syndrome, fatty liver like non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), Morbus Wilson, Hemochromatosis, alpha1-antitrypsin deficiency, glycogen storage diseases;all types of liver cirrhosis, such as primary biliary cirrhosis, ethyl toxic liver cirrhosis, cryptogenic cirrhosis;acute (fulminant) or chronic liver failure such as toxic liver failure like acetaminophen (paracetamol) induced liver failure, alpha-amanitin induced liver failure, drug induced hepatotoxicity, liver failure caused, for example, by antibiotics, nonsteroidal anti-inflammatory drugs and anticonvulsants, acute liver failure induced by herbal supplements (kava, ephedra, skullcap, pennyroyal etc), liver disease and failure due to vascular diseases such as Budd-Chiari syndrome, acute liver failure of unknown origin, chronic liver disease due to right heart failure;galactosemia, cystic fibrosis, porphyria, hepatic ischemia perfusion injury, small for size syndrome after liver transplantation, primary sclerosing cholangitis or hepatic encephalopathy. For promoting liver regeneration or reducing or preventing hepatocyte death the compounds of the invention are administered to a patient in need thereof in a therapeutically effective amount. Various diagnostic methods are available to detect the presence of a liver disease. Blood levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), above clinically accepted normal ranges, are known to be indicative of on-going liver damage. Blood bilirubin levels or other liver enzymes may be used as detection or diagnostic criteria. Routine monitoring of liver disease patients for blood levels of ALT and AST is used to measure progress of the liver disease while on medical treatment. Reduction of elevated ALT and AST levels to within the accepted normal range is taken as clinical evidence reflecting a reduction in the severity of the patients' liver damage. Commercial assays such as FibroTest/FibroSURE, HepaScore®, FibroMeter or Cirrhometer evaluate the combined results of five and more biochemical parameters for the detection of liver steatosis, fibrosis and cirrhosis. Furthermore, non-invasive, innovative physical imaging techniques such as magnetic resonance imaging, sonography and, in particular, elastography techniques are available to detect and monitor the status and progression of liver diseases. It has further been found that shRNA mediated MKK4 suppression attenuate TNF-α-driven cartilage matrix degradation in osteoarthritis (Cell Death and Disease (2017) 8, e3140). Therefore, inhibition of the activity of MKK4 using the compounds of the invention are further useful for treating osteoarthritis and rheumatoid arthritis. Furthermore, MKK4 inhibitors may also be useful for treatment of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Grueninger et al. found that in human neuroblastoma cells, MKK4 plays a key role in the phosphorylation of Tau protein at serine 422 which promotes Tau aggregation (Mol Cell Biochem (2011) 357:199-207). Inhibitors of Tau phosphorylation which prevents the aggregation of Tau are being considered useful for prevention or treatment of Alzheimer's disease. Recently, a MKK4 inhibitor has been described with potent neuroprotective effects in vitro and in vivo. In hippocampal cultures, the incubation with an MKK4-inhibitor prevented glutamate-induced cell death and caspase-3 activation, and also inhibited N-Methyl-4-phenylpyridinium iodide- and amyloid (31-42-induced cell death in SH-SY5Y cells. The same compound also alleviated 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced degeneration of nigrostriatal dopaminergic neurons in mice (Biochemical Pharmacology (2018), doi: doi.org/10.1016/j.bcp.2018.10.008). The compounds of the invention are customarily administered in the form of pharmaceutical compositions which comprise at least one compound according to the invention, optionally together with an inert carrier (e.g. a pharmaceutically acceptable excipient) and, where appropriate, other drugs. These compositions can, for example, be administered orally, rectally, transdermally, subcutaneously, intraperitoneally, intravenously, intramuscularly or intranasally. Examples of suitable pharmaceutical compositions are solid medicinal forms, such as powders, granules, tablets, in particular film tablets, lozenges, sachets, cachets, sugar-coated tablets, capsules, such as hard gelatin capsules and soft gelatin capsules, or suppositories, semisolid medicinal forms, such as ointments, creams, hydrogels, pastes or plasters, and also liquid medicinal forms, such as solutions, emulsions, in particular oil-in-water emulsions, suspensions, for example lotions, injection preparations and infusion preparations. In addition, it is also possible to use liposomes or microspheres. When producing the compositions, the compounds according to the invention are optionally mixed or diluted with one or more carriers (excipients). Carriers (excipients) can be solid, semisolid or liquid materials which serve as vehicles, carriers or medium for the active compound. Suitable carriers (excipients) are listed in the specialist medicinal monographs. In addition, the formulations can comprise pharmaceutically acceptable auxiliary substances, such as wetting agents; emulsifying and suspending agents; preservatives; antioxidants; antiirritants; chelating agents; coating auxiliaries; emulsion stabilizers; film formers; gel formers; odor masking agents; taste corrigents; resins; hydrocolloids; solvents; solubilizers; neutralizing agents; diffusion accelerators; pigments; quaternary ammonium compounds; refatting and overfatting agents; raw materials for ointments, creams or oils; silicone derivatives; spreading auxiliaries; stabilizers; sterilants; suppository bases; tablet auxiliaries, such as binders, fillers, glidants, disintegrants or coatings; propellants; drying agents; opacifiers; thickeners; waxes; plasticizers and white mineral oils. A formulation in this regard is based on specialist knowledge as described, for example, in Fiedler, H. P., Lexikon der Hilfsstoffe für Pharmazie, Kosmetik und angrenzende Gebiete [Encyclopedia of auxiliary substances for pharmacy, cosmetics and related fields], 4thedition, Aulendorf: ECV-Editio-Cantor-Verlag, 1996. The compounds of the invention may also be suitable for combination with other therapeutic agents. The invention therefore further relates to a combination comprising a compound of the invention with one or more further therapeutic agents, in particular for use in promoting liver regeneration or reducing or preventing hepatocyte death. The combination therapies of the invention may be administered adjunctively. By adjunctive administration is meant the coterminous or overlapping administration of each of the components in the form of separate pharmaceutical compositions or devices. This regime of therapeutic administration of two or more therapeutic agents is referred to generally by those skilled in the art and herein as adjunctive therapeutic administration; it is also known as add-on therapeutic administration. Any and all treatment regimes in which a patient receives separate but coterminous or overlapping therapeutic administration of the compounds of the invention and at least one further therapeutic agent are within the scope of the current invention. In one embodiment of adjunctive therapeutic administration as described herein, a patient is typically stabilized on a therapeutic administration of one or more of the components for a period of time and then receives administration of another component. The combination therapies of the invention may also be administered simultaneously. By simultaneous administration is meant a treatment regime wherein the individual components are administered together, either in the form of a single pharmaceutical composition or device comprising or containing both components, or as separate compositions or devices, each comprising one of the components, administered simultaneously. Such combinations of the separate individual components for simultaneous combination may be provided in the form of a kit-of-parts. Suitable agents for use in combination with the compounds of the inventions include for example:ACC inhibitors such as TOFA (5-(tetradecyloxy)-2-furoic acid), PF-05221304,GS 0976, and ACC inhibitors as disclosed in WO 2016/112305,angiotensin II receptor antagonists,angiotensin converting enzyme (ACE) inhibitors, such as enalapril,caspase inhibitors, such as emricasan,cathepsin B inhibitors, such as a mixed cathepsin B/hepatitis C virus NS3 protease inhibitor. like VBY-376,CCR2 chemokine antagonists, such as a mixed CCR2/CCR5 chemokine antagonist like cenicriviroc,CCR5 chemokine antagonists,chloride channel stimulators, such as cobiprostone,cholesterol solubilizers,diacylglycerol 0-acyltransferase 1 (DGAT1) inhibitors, such as LCQ908,diacylglycerol O-Acyltransferase 2 (DGAT2) Inhibitor, such as PF-06865571,ketohexokinase (KHK) Inhibitor, such as PF-06835919,dipeptidyl peptidase IV (DPPIV) inhibitors, such as linagliptin,farnesoid X receptor (FXR) agonists, such as INT-747 (obeticholic acid), LJN452 (tropifexor) and analogues disclosed in Tully et al (J. Med. Chem., 2017 60 (24), 9960-9973), or GS-9674 (PX-102),FXR/TGR5 dual agonists, such as INT-767,galectin-3 inhibitors, such as GR-MD-02,glucagon-like peptide 1 (GLP1) agonists, such as liraglutide or exenatide,glutathione precursors,hepatitis C virus NS3 protease inhibitors, such as a mixed cathepsin B/hepatitis C virus NS3 protease inhibitor like VBY-376,HMG CoA reductase inhibitors, such as a statin like atorvastatin,11ß-hydroxysteroid dehydrogenase (11ß-HSD1) inhibitors, such as R05093151,IL-1ß antagonists,IL-6 antagonists, such as a mixed IL-6/IL-1ß/TNFα ligand inhibitor like BLX-1002,IL-10 agonists, such as peg-ilodecakin,IL-17 antagonists, such as KD-025,ileal sodium bile acid cotransporter inhibitors, such as SHP-626,leptin analogs, such as metreleptin,5-lipoxygenase inhibitors, such as a mixed 5-lipoxygenase/PDE3/PDE4/PLC inhibitor like tipelukast,LPL gene stimulators, such as alipogene tiparvovec,lysyl oxidase homolog 2 (LOXL2) inhibitors, such as an anti-LOXL2 antibody like GS-6624, PDE3 inhibitors, such as a mixed 5-ipoxygenase/PDE3/PDE4/PLC inhibitor like tipelukast, PDE4 inhibitors, such as ASP-9831 or a mixed 5-ipoxygenase/PDE3/PDE4/PLC inhibitor like tipelukast,phospholipase C (PLC) inhibitors, such as a mixed 5-ipoxygenase/PDE3/PDE4/PLC inhibitor like tipelukast,PPARα agonists, such as a mixed PPARα/δ agonist like GFT505,PPARγ agonists, such as pioglitazone,PPARδ agonists,Rho associated protein kinase 2 (ROCK2) inhibitors, such as KD-025,sodium glucose transporter-2 (SGLT2) inhibitors, such as remogliflozin etabonate,stearoyl CoA desaturase-1 inhibitors, such as aramchol or CVT-12805,thyroid hormone receptor ß agonists, such as MGL-3196,tumor necrosis factor α (TNFα) ligand inhibitors,transglutaminase inhibitors and transglutaminase inhibitor precursors, such as mercaptamine,PTPIb inhibitors, such as A119505, A220435, A321842, CPT633, ISIS-404173, JTT-551, MX-7014, MX-7091, MX-7102, NNC-521246, OTX-001, OTX-002, orTTP814 and ASK1 inhibitors such as GS4977. In some embodiments, the one or more further therapeutic agents are selected from acetylsalicylic acid, alipogene tiparvovec, aramchol, atorvastatin, BLX-1002, cenicriviroc, cobiprostone, colesevelam, emncasan, enalapril, GFT-505, GR-MD-02, hydrochlorothiazide, icosapent ethyl ester (ethyl eicosapentaenoic acid), IMM-124E, KD-025, linagliptin, liraglutide, mercaptamine, MGL-3196, obeticholic acid, olesoxime, peg-ilodecakin, pioglitazone, GS-9674, remogliflozin etabonate, SHP-626, solithromycin, tipelukast, TRX-318, ursodeoxycholic acid, and VBY-376. In some embodiments, one of the one or more further therapeutic agents is selected from acetylsalicylic acid, alipogene tiparvovec, aramchol, atorvastatin, BLX-1 002, and cenicriviroc. In an embodiment the invention relates to a method ofinhibiting protein kinase MKK4,selectively inhibiting protein kinase MKK4 over protein kinases JNK1 and MKK7, promoting liver regeneration or preventing hepatocyte death,treating acute, acute-on-chronic or chronic liver disease,treating acute and chronic or acute on chronic liver diseases such as acute and chronic viral hepatitis like hepatitis B, C, E, hepatitis caused by Epstein-Barr virus, cytomegalovirus, herpes simplex virus and other viruses, all types of autoimmune hepatitis, primary sclerosing hepatitis, alcoholic hepatitis;treating metabolic liver diseases such as metabolic syndrome, fatty liver like non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), Morbus Wilson, hemochromatosis, alpha1-antitrypsin deficiency, glycogen storage diseases;treating all types of liver cirrhosis, such as primary biliary cirrhosis, ethyl toxic liver cirrhosis, cryptogenic cirrhosis;treating acute (fulminant) or chronic liver failure such as toxic liver failure like acetaminophen (paracetamol) induced liver failure, alpha-amanitin induced liver failure, drug induced hepatotoxicity and liver failure caused, for example, by antibiotics, nonsteroidal anti-inflammatory drugs, anticonvulsants, acute liver failure induced by herbal supplements (kava, ephedra, skullcap, pennyroyal etc.), liver disease and failure due to vascular diseases such as Budd-Chiari syndrome, acute liver failure of unknown origin, chronic liver disease due to right heart failure;treating galactosemia, cystic fibrosis, porphyria, hepatic ischemia perfusion injury, small for size syndrome after liver transplantation, primary sclerosing cholangitis or hepatic encephalopathy,treating osteoarthritis, rheumatoid arthritis, or CNS-related diseases such as Alzheimer disease and Parkinson disease,which comprises administering an effective amount of a compound or a composition as defined above to a subject in need thereof. In an embodiment, the compounds of the invention are administered in a dosage of 0.2 to 15 mg/kg or 0.5 to 12 mg/kg of the subject being treated. The compounds can be administered once or several times a day. The compounds are administered over 4 to 12 weeks. The following examples illustrate the invention without limiting it. EXAMPLES Abbreviations Boc2O di-tert.-butyloxycarbonateCPME cyclopentylmethyl etherDCM dichloromethane4-DMAP 4-dimethylaminopyridineDME dimethyl etherDMF dimethylformamideDMSO dimethylsulfoxideEtOAc ethyl acetateHPLC high performance liquid chromatographyLDA lithium diisopropylamideMeCN acetonitrileMeOH methanolNaHCO3sodium bicarbonateNH4Cl ammonium chlorideNa2SO4sodium sulfatePd2(dba3) tris(dibenzylideneacetone)dipalladium(0)PE petroletherRT room temperatureSol. SolutionTHE tetrahydrofuraneTLC thin layer chromatographyXantphos 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene Example 1: Synthesis of N-(3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl)propane-1-sulfonamide (VI) Step 1: Synthesis of 5-Bromo-3-iodo-1H-pyrazolo[3,4-b]pyridine (II) To a stirred mixture of 5-bromo-1H-pyrazolo[3,4-b]pyridine ((I), 6.81 g, 34.4 mmol) and KOH (6.75 g, 120.4 mmol) in DMF (45 mL) was added iodine (9.60 g, 37.8 mmol) in one portion at RT. After a short induction period the exothermic reaction began. After 1 h, an additional 1 g portion of iodine was added and the mixture stirred at 45° C. for 1 h. The mixture was poured into 300 mL of a dilute solution of Na2SO3and the mixture was acidified with 2N HCl. The solids were collected by suction filtration, washed with water and dried in an oven at 110° C. Yield: 10.92 g, HPLC purity: 95%,1H NMR (200 MHz, DMSO) δ 14.29 (s, 1H), 8.62 (s, 1H), 8.17 (s, 1H);13C NMR (50 MHz, DMSO) δ 150.53, 150.17, 131.86, 120.58, 112.43, 91.95; [M−H]−=322.0/324.0. Step 2: Synthesis of 5-Bromo-1H-pyrazolo[3,4-b]pyridine-3-carboxylic acid (Ill) 5-Bromo-3-iodo-1H-pyrazolo[3,4-b]pyridine ((II), 10.44 g, 32.2 mmol) was combined with DMF, MeOH and triethylamine (75 mL each). The vessel was evacuated and flushed with argon (4×). XantPhos (1.12 g, 1.93 mmol) and Pd(OAc)2(217 mg, 0.97 mmol) were added and carbon monoxide (generated from formic acid and sulfuric acid) was bubbled through the solution while heating to 60° C. The mixture was stirred under an atmosphere of carbon monoxide (balloon) for 8 h. Every 1.5 h carbon monoxide was bubbled through the solution for 5 minutes. The mixture was concentrated under reduced pressure and the residue was triturated with 2N HCl. The solids were heated at 95° C. in about 100 mL 1N NaOH overnight. After cooling to RT, the mixture was acidified with conc. HCl and the precipitate collected by suction filtration and washed with water. The solids were dried in an oven at 110° C. to constant mass. The solids were sonicated in 100 mL of toluene for 5 minutes and stirred for 30 minutes. The product was filtered, washed with an additional 20 mL of toluene and dried at 110° C. Yield: 7.92 g HPLC purity: >99%,1H NMR (200 MHz, DMSO) δ 8.64 (d, J=7.9 Hz, 2H), 5.69 (bs, 1H);13C NMR (50 MHz, DMSO) δ 163.27, 150.97, 149.67, 136.69, 132.65, 115.73, 113.6; [M−H]−=239.9/241.9. Step 3: Synthesis of 5-bromo-N-methoxy-N-methyl-1H-pyrazolo[3,4-b]pyridine-3-carboxamide (IV) 5-Bromo-1H-pyrazolo[3,4-b]pyridine-3-carboxylic acid ((III), 7.91 g, 32.7 mmol) and 1,1′-carbonyldiimidazole (5.83 g, 35.9 mmol) were stirred in 200 mL of DMF at 60° C. for 45 minutes. To the resulting suspension was added N,O-dimethylhydroxylamine hydrochloride (3.51 g, 35.9 mmol) and the mixture was stirred for 4 h at 65° C. Most of the solvent was removed under vacuum and to the residue half sat. NaHCO3-solution was added. The solids were collected by suction filtration, washed with water and dried at 110° C. Yield: 7.94 g, HPLC purity: 96%,1H NMR (200 MHz, DMSO) δ 14.46 (s, 1H), 8.62 (d, J=20.4 Hz, 2H), 3.76 (s, 3H), 3.44 (s, 3H), [M−H]−=283.0/285.0. Step 4: Synthesis of (3-amino-2,6-difluorophenyl)(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (V) 2,4-Difluoroaniline (6.25 g, 48.4 mmol) was dissolved in 50 mL dry THE and cooled to −78° C. under an atmosphere of argon. 2.5 M n-butyllithium in hexane (19.4 mL, 48.4 mmol) was added dropwise. After 15 minutes 1,2-bis(chlorodimethylsilyl)ethane (10.9 g, 49.5 mmol) in 15 mL dry THE was added dropwise and the mixture was stirred for 30 minutes. 2.5 M n-butyllithium in hexane (19.4 mL, 48.4 mmol) was added dropwise and the mixture was allowed to reach RT within 1 h. After cooling to −78° C. 2.5 M n-butyllithium in hexane (19.4 mL, 48.4 mmol) was added dropwise and stirred for 1 h at −78° C. (This is considered solution A). 5-bromo-N-methoxy-N-methyl-1H-pyrazolo[3,4-b]pyridine-3-carboxamide ((IV), 6.00 g, 21.1 mmol) was suspended in 50 mL dry THE and cooled to 0° C. under an atmosphere of argon. NaH (60% in mineral oil, 0.88 g, 22.1 mmol) was added portionwise and the solution was stirred at RT for 1 h. (This is considered solution B). Solution B was added dropwise to solution A at −78° C. After complete addition, the mixture was warmed to RT within 30 minutes. 12 mL conc. HCl were added carefully and the mixture was stirred for 30 minutes. Solid NaHCO3was added to neutralize the solution, the solids were filtered off and washed with THF. The filtrate was evaporated and the residue triturated with MeOH and water and dried at 110° C. Yield: 4.03 g; HPLC purity: 97%,1H NMR (200 MHz, DMSO) δ 14.91 (s, 1H), 8.77 (dd, J=5.4, 2.1 Hz, 2H), 7.18-6.59 (m, 2H), 5.25 (s, 2H);13C NMR (50 MHz, DMSO) δ 183.95, 151.04, 150.79, 150.27 (dd, J=161.0, 6.8 Hz), 145.50 (dd, J=167.3, 6.8 Hz), 141.34, 133.35 (dd, J=12.8, 2.6 Hz), 132.28, 117.45 (dd, J=8.4, 6.5 Hz), 116.24 (dd, J=22.7, 19.1 Hz), 115.55, 114.81, 111.26 (dd, J=21.7, 3.5 Hz); [M−H]−=351.1/353.1. Step 5: Synthesis of N-(3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl)propane-1-sulfonamide (VI) (3-Amino-2,6-difluorophenyl)(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone ((V), 2.00 g, 5.66 mmol) and 4-DMAP (35 mg, 0.28 mmol) were heated in 9 mL pyridine to 65° C. and 1-propanesulfonyl chloride (1.21 g, 0.96 mL, 8.50 mmol) was added. After 2 h, another 0.19 mL 1-propanesulfonyl chloride were added. The warm solution was added to about 80 mL 2N HCl, the solid collected and washed with water. The solid was taken up in EtOAc, washed with 2N HCl and brine and dried over Na2SO4. The solvent was evaporated and the product purified by flash chromatography (SiO2, DCM/EtOAc gradient, from 0% to 20% EtOAc) and triturated with n-hexane. Yield: 1.68 g, HPLC purity: 97%,1H NMR (200 MHz, DMSO) δ 9.86 (s, 1H), 8.79 (dd, J=5.3, 2.0 Hz, 3H), 7.64 (td, J=9.0, 6.0 Hz, 1H), 7.30 (t, J=8.9 Hz, 1H), 3.17-2.96 (m, 3H), 1.87-1.62 (m, 2H), 0.96 (t, J=7.4 Hz, 4H);13C NMR (50 MHz, DMSO) δ 182.75, 157.39 (dd, J=177.1, 7.4 Hz), 152.42 (dd, J=180.3, 7.3 Hz), 151.56, 151.34, 141.39, 132.59, 130.78-130.05 (m), 122.20 (dd, J=13.5, 3.6 Hz), 117.19 (dd, J=23.0, 20.9 Hz), 116.14, 115.18, 112.59 (dd, J=22.2, 3.8 Hz), 54.14, 17.22 12.97; [M−H]−=457.1/459.1. Example 2: Synthesis of N-(2,4-difluoro-3-(5-phenyl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl)propane-1-sulfonamide A microwave vessel was charged with a magnetic stir bar, N-(3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl)propane-1-sulfonamide ((VI), 50 mg, 0.11 mmol), phenylboronic acid (15 mg, 0.12 mmol), Pd(PPh3)4(6 mg, 5 mol %) and purged with argon. Degassed 1,4-dioxane (0.3 mL) and degassed aqueous 1.5 M K2CO3(0.25 mL, 0.38 mmol) were added and the mixture was heated to 120° C. under microwave irradiation for 30 minutes. After cooling, the mixture was diluted with EtOAc and neutralized with sat. NH4Cl solution. The solvents were removed and the product isolated by flash chromatography and dried at 100° C. in a vacuum oven. Yield: 27 mg, HPLC purity: 97,1H NMR (200 MHz, DMSO) δ 7.84 (d, J=7.2 Hz, 2H), 7.71-7.40 (m, 4H), 7.31 (t, J=8.7 Hz, 1H), 3.18-3.04 (m, 2H), 1.87-1.63 (m, 2H), 0.97 (t, J=7.4 Hz, 3H). [M−H]−=455.1. General procedure for Suzuki coupling: A microwave vessel is charged with a magnetic stir bar, N-(3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl)propane-1-sulfonamide, the appropriate boronic acid or boronic acid pinacole ester (1.1 eq.) and Pd(PPh3)4[(tBu)3P Pd G4 and/or XPhos Pd G4 (G4: 4thgeneration; commercially available) for nitrophenylboronic acids, 4-hydroxyphenylboronic acid and 4-dimethylaminophenyl-boronic acid pinacol ester] (0.05 eq.) and purged with argon. Degassed 1,4-dioxane (0.4 M) and degassed aqueous 1.5 M K2CO3(3.5 eq.) are added and the mixture is heated to 120° C. under microwave irradiation until complete conversion (usually 30 minutes). After cooling, the mixture is diluted with EtOAc and neutralized with sat. NH4Cl solution. The solvents are removed and the product isolated by flash chromatography and dried at 100° C. in a vacuum oven. In analogy, the compounds given in the following table 1 were prepared. TABLE 1Boronicacid/Pina-Ex.col esterStructureMWIUPAC1H-NMR/MS3490.91N-(3-(5-(4-chlorophenyl)-1H- pyrazolo[3,4-b]pyri-dine-3- carbonyl)-2,4-difluorophenyl)- propane-1-sulfonamide1H NMR (200 MHz, DMSO) δ 9.04 (s, 1H), 8.78 (s, 1H), 7.89 (d, J = 8.5 Hz, 2H), 7.71-7.54 (m, 3H), 7.31 (t, J = 8.7 Hz, 1H), 3.19-3.05 (m, 2H), 1.75 (dd, J = 14.8, 7.6 Hz, 2H), 0.97 (t, J = 7.2 Hz, 3H); [M − H]−= 489.24488.49N-(2,4-difluoro-3-(5-(4-fluoro- 2-methylphenyl)-1H-pyrazo- lo[3,4-b]-pyri-dine-3-car- bonyl)-phenyl)-propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 14.95 (s, 1H), 9.85 (s, 1H), 8.70 (d, J = 1.8 Hz, 1H), 8.48 (d, J = 1.8 Hz, 1H), 7.64 (td, J = 9.1, 6.0 Hz, 1H), 7.48-7.10 (m, 4H), 3.21-3.03 (m, 2H), 2.28 (s, 3H), 1.85-1.65 (m, 2H), 0.97 (t, J = 7.3 Hz, 3H); [M − H]−= 486.95520.94N-(3-(5-(2-chloro-4-methoxy- phenyl)-1H-pyrazolo[3,4-b]- pyridine-3-carbonyl)-2,4- difluorophenyl)propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 14.93 (s, 1H), 9.83 (s, 1H), 8.74 (s, 1H), 8.58 (s, 1H), 7.75-7.47 (m, 2H), 7.37- 7.03 (m, 3H), 3.86 (s, 3H), 3.18-3.02 (m, 2H), 1.88- 1.61 (m, 2H), 0.97 (t, J = 7.2 Hz, 3H); [M − H]−= 519.16486.49N-(2,4-difluoro-3-(5-(4- methoxyphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.87 (s, 1H), 9.79 (s, 1H), 9.00 (d, J = 2.2 Hz, 1H), 8.69 (d, J = 2.2 Hz, 1H), 7.78 (d, J = 8.8 Hz, 1H), 7.63 (td, J = 9.0, 5.9 Hz, 1H), 7.31 (td, J = 9.0, 1.3 Hz, 1H), 7.11 (d, J = 8.7 Hz, 1H), 3.83 (s, 1H), 3.22-3.03 (m, 1H), 1.86-1.63 (m, 1H), 0.97 (t, J = 7.4 Hz, 1H); [M − H]−= 485.17490.91N-(3-(5-(3-chlorophenyl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluorophenyl)- propane-1-sulfonamide1H NMR (200 MHz, DMSO) δ 9.05 (s, 1H), 8.80 (s, 1H), 8.06-7.19 (m, 6H), 3.18-3.03 (m, 2H), 1.91-1.61 (m, 2H), 0.97 (t, J = 7.2 Hz, 3H); [M − H]−= 489.08499.54N-(3-(5-(4-(dimethylamino)- phenyl)-1H-pyrazolo[3,4- b]pyridine-3-carbonyl)-2,4- difluorophenyl)-propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 14.83 (s, 1H), 9.82 (s, 1H), 8.98 (d, J = 2.2 Hz, 1H), 8.63 (d, J = 2.2 Hz, 1H), 7.73- 7.54 (m, 3H), 7.31 (td, J = 8.9, 1.5 Hz, 1H), 6.87 (d, J = 8.9 Hz, 2H), 3.19-3.05 (m, 2H), 2.97 (s, 6H), 2.97 (s, 6H), 1.86-1.64 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 497.99490.91N-(3-(5-(2-chlorophenyl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluorophenyl)- propane-1-sulfonamide1H NMR (200 MHz, DMSO) δ 8.79 (d, J = 2.1 Hz, 1H), 8.62 (d, J = 2.1 Hz, 1H), 7.75-7.43 (m, 5H), 7.31 (td, J = 9.0, 1.4 Hz, 1H), 3.11 (dd, J = 9.0, 6.3 Hz, 2H), 1.87- 1.64 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 488.910512.58N-(3-(5-(4-tert-butylphenyl)- 1H-pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluoro- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.92 (s, 1H), 9.83 (s, 1H), 9.03 (d, J = 1.9 Hz, 1H), 8.73 (d, J = 1.7 Hz, 1H), 7.84- 7.50 (m, 5H), 7.31 (t, J = 8.7 Hz, 1H), 3.22-3.01 (m, 2H), 1.85-1.63 (m, 2H), 1.34 (s, 9H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 511.111498.55N-(2,4-difluoro-3-(5-(4- isopropylphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl) phenyl)propane-1-sulfon- amide1H NMR (400 MHz, DMSO) δ 14.92 (s, 1H), 9.83 (s, 1H), 9.02 (s, 1H), 8.73 (s, 1H), 7.75 (d, J = 7.9 Hz, 2H), 7.69- 7.59 (m, 1H), 7.42 (d, J = 8.0 Hz, 2H), 7.31 (t, J = 8.8 Hz, 1H), 3.19-3.07 (m, 2H), 2.96 (dt, J = 13.6, 6.8 Hz, 1H), 1.82-1.67 (m, 2H), 1.25 (d, J = 6.9 Hz, 6H), 0.97 (t, J = 7.4 Hz, 3H); [M + Na]+= 521.012484.52N-(3-(5-(3,4-dimethyl- phenyl)-1H-pyrazolo[3,4- b]pyridine-3-carbonyl)-2,4- difluorophenyl)-propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 14.44 (s, 1H), 9.37 (s, 1H), 8.54 (d, J = 2.1 Hz, 1H), 8.25 (d, J = 2.1 Hz, 1H), 7.25- 7.04 (m, 3H), 6.93-6.76 (m, 2H), 2.73-2.56 (m, 2H), 1.87 (s, 3H), 1.83 (s, 3H), 1.41-1.17 (m, 2H), 0.51 (t, J = 7.4 Hz, 3H); [M − H]−= 483.113470.49N-(2,4-difluoro-3-(5-o-tolyl- 1H-pyrazolo[3,4-b]pyridine-3- carbonyl)-phenyl)propane-1- sulfon-amide1H NMR (200 MHz, DMSO) δ 8.72 (d, J = 2.0 Hz, 1H), 8.49 (d, J = 2.1 Hz, 1H), 7.64 (td, J = 9.1, 5.8 Hz, 1H), 7.44-7.23 (m, 5H), 3.22-2.96 (m, 2H), 2.28 (s, 3H), 1.86-1.63 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 469.014486.49N-(2,4-difluoro-3-(5-(2- methoxyphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.90 (s, 1H), 9.83 (s, 1H), 8.81 (d, J = 2.0 Hz, 1H), 8.61 (d, J = 2.0 Hz, 1H), 7.63 (td, J = 9.0, 5.9 Hz, 1H), 7.45 (dd, J = 12.8, 4.6 Hz, 2H), 7.38-7.05 (m, 3H), 3.82 (s, 3H), 3.17-3.04 (m, 2H), 1.87-1.61 (m, 2H), 0.97 (t, J = 7.5 Hz, 3H); [M − H]−= 485.115535.544-(3-(2,6-difluoro-3-(propyl- sulfonamido)benzoyl)-1H- pyrazolo[3,4-b]pyridin-5- yl)benzene-sulfonamide1H NMR (200 MHz, DMSO) δ 14.53 (s, 1H), 9.37 (s, 1H), 8.64 (d, J = 2.2 Hz, 1H), 8.40 (d, J = 2.1 Hz, 1H), 7.62 (d, J = 8.5 Hz, 2H), 7.51 (d, J = 8.5 Hz, 2H), 7.28-7.06 (m, 2H), 7.02 (s, 2H), 6.86 (td, J = 8.8, 1.3 Hz, 1H), 2.76- 2.53 (m, 2H), 1.40-1.18 (m, 2H), 0.51 (t, J = 7.4 Hz, 3H); [M − H]−= 534.016474.46N-(2,4-difluoro-3-(5-(3- fluorophenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.95 (s, 1H), 9.81 (s, 1H), 9.06 (d, J = 1.9 Hz, 1H), 8.81 (d, J = 1.7 Hz, 1H), 7.81- 7.51 (m, 4H), 7.41-7.23 (m, 2H), 3.18-3.04 (m, 2H), 1.85-1.63 (m, 2H), 0.97 (t, J = 7.5 Hz, 3H); [M − H]−= 472.917486.49N-(2,4-difluoro-3-(5-(3- methoxyphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.94 (s, 1H), 9.83 (s, 1H), 9.04 (d, J = 2.2 Hz, 1H), 8.76 (d, J = 2.2 Hz, 1H), 7.64 (td, J = 9.1, 6.0 Hz, 1H), 7.51-7.26 (m, 4H), 7.09-6.99 (m, 1H), 3.87 (s, 3H), 3.20-2.99 (m, 2H), 1.86-1.61 (m, 2H), 0.97 (t, J = 7.5 Hz, 3H); [M − H]−= 485.018471.48N-(3-(5-(4-aminophenyl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluorophenyl)- propane-1-sulfonamide1H NMR (200 MHz, DMSO) δ 8.94 (s, 1H), 8.59 (s, 1H), 7.77-7.41 (m, 3H), 7.30 (t, J = 9.0 Hz, 1H), 6.72 (d, J = 7.9 Hz, 2H), 5.40 (s, 2H), 3.23-3.00 (m, 2H), 1.96- 1.56 (m, 2H), 0.97 (t, J = 7.5 Hz, 3H); [M + Na]+= 494.019472.47N-(2,4-difluoro-3-(5-(4- hydroxyphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.40 (s, 1H), 9.36 (s, 1H), 9.27 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 8.19 (d, J = 2.0 Hz, 1H), 7.26-7.06 (m, 3H), 6.84 (t, J = 8.8 Hz, 1H), 6.47 (d, J = 8.5 Hz, 2H), 2.73-2.54 (m, 2H), 1.39-1.17 (m, 2H), 0.51 (t, J = 7.4 Hz, 3H); [M − H]−= 471.120470.49N-(2,4-difluoro-3-(5-p-tolyl- 1H-pyrazolo[3,4-b]pyridine-3- carbonyl)phenyl)propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 14.91 (s, 1H), 9.85 (s, 1H), 9.01 (d, J = 1.9 Hz, 2H), 8.72 (d, J = 2.0 Hz, 2H), 7.82- 7.55 (m, 5H), 7.45-7.23 (m, 4H), 3.21-3.04 (m, 3H), 2.38 (s, 4H), 1.86-1.64 (m, 3H), 0.97 (t, J = 7.3 Hz, 4H); [M − H]−= 469.121472.47N-(2,4-difluoro-3-(5-(2- hydroxyphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.89 (s, 1H), 9.88 (s, 2H), 8.87 (d, J = 2.1 Hz, 1H), 8.71 (d, J = 2.1 Hz, 1H), 7.63 (td, J = 9.0, 6.0 Hz, 1H), 7.45 (dd, J = 7.3, 1.4 Hz, 1H), 7.38-7.21 (m, 2H), 7.10-6.89 (m, 2H), 3.20-3.02 (m, 2H), 1.86-1.64 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 470.922525.35N-(3-(5-(3,5-dichloro-phenyl)- 1H-pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluoro- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 9.06 (d, J = 2.2 Hz, 1H), 8.86 (d, J = 2.2 Hz, 1H), 7.96 (d, J = 1.8 Hz, 2H), 7.72- 7.55 (m, 2H), 7.31 (td, J = 9.0, 1.5 Hz, 1H), 3.20-3.00 (m, 2H), 1.87-1.64 (m, 2H), 0.97 (t, J = 7.5 Hz, 3H); [M − H]−= 523.023501.46N-(2,4-difluoro-3-(5-(4- nitrophenyl)-1H-pyrazolo- [3,4-b]pyridine-3-carbonyl)- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 24.19 (s, 1H), 18.97 (s, 1H), 18.29 (d, J = 2.0 Hz, 1H), 18.06 (d, J = 2.1 Hz, 1H), 17.53 (d, J = 8.7 Hz, 2H), 17.32 (d, J = 8.7 Hz, 2H), 16.79 (td, J = 9.0, 5.8 Hz, 1H), 16.47 (t, J = 8.7 Hz, 1H), 12.35-12.17 (m, 2H), 11.01-10.78 (m, 2H), 10.12 (t, J = 7.4 Hz, 3H); [M − H]−= 500.024534.55N-(2,4-difluoro-3-(5-(4- (methylsulfonyl)phenyl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)phenyl)propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 15.03 (s, 1H), 9.83 (s, 1H), 9.12 (d, J = 2.2 Hz, 1H), 8.88 (d, J = 2.2 Hz, 1H), 8.12 (q, J = 8.7 Hz, 4H), 7.72-7.55 (m, 1H), 7.32 (td, J = 9.0, 1.5 Hz, 1H), 3.30 (s, 3H), 3.21-3.03 (m, 2H), 1.85- 1.64 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 532.925474.46N-(2,4-difluoro-3-(5-(4- fluorophenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.91 (s, 1H), 9.89 (s, 1H), 9.02 (d, J = 2.1 Hz, 1H), 8.74 (d, J = 2.1 Hz, 1H), 7.89 (dd, J = 8.6, 5.4 Hz, 2H), 7.64 (td, J = 9.0, 5.9 Hz, 1H), 7.45-7.21 (m, 3H), 3.21-3.02 (m, 2H), 1.87-1.61 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 473.026471.48N-(3-(5-(2-aminophenyl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluorophenyl)- propane-1-sulfonamide1H NMR (200 MHz, DMSO) δ 14.39 (s, 1H), 9.41 (s, 1H), 8.24 (d, J = 2.1 Hz, 1H), 8.11 (d, J = 2.0 Hz, 1H), 7.17 (td, J = 9.1, 6.0 Hz, 1H), 6.85 (t, J = 8.7 Hz, 1H), 6.74- 6.57 (m, 3H), 6.36 (d, J = 7.8 Hz, 1H), 6.23 (t, J = 7.5 Hz, 1H), 4.56 (s, 2H), 2.72-2.57 (m, 3H), 1.39-1.12 (m, 3H), 0.51 (t, J = 7.4 Hz, 4H); [M − H]−= 470.027471.48N-(3-(5-(3-aminophenyl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluorophenyl)- propane-1-sulfonamide1H NMR (400 MHz, DMSO) δ 14.91 (s, 1H), 9.80 (s, 1H), 8.94 (s, 1H), 8.66 (s, 1H), 7.64 (d, J = 5.7 Hz, 1H), 7.39- 7.12 (m, 2H), 7.09-6.89 (m, 2H), 6.66 (d, J = 6.2 Hz, 1H), 5.30 (s, 2H), 3.12 (s, 2H), 1.75 (d, J = 5.9 Hz, 2H), 0.97 (s, 3H); [M + Na]+= 494.028499.54N-(3-(5-(3-(dimethyl- amino)phenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- 2,4-difluoro-phenyl)propane- 1-sulfonamide1H NMR (200 MHz, DMSO) δ 9.11-8.84 (m, 1H), 8.70 (d, J = 2.1 Hz, 1H), 7.64 (td, J = 9.0, 6.0 Hz, 1H), 7.32 (q, J = 8.1 Hz, 2H), 7.11-6.99 (m, 2H), 6.81 (dd, J = 8.1, 1.8 Hz, 1H), 3.18-3.04 (m, 2H), 2.99 (s, 6H), 1.87- 1.63 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 498.029470.49N-(2,4-difluoro-3-(5-m-tolyl- 1H-pyrazolo[3,4-b]pyridine-3- carbonyl)phenyl)propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 14.92 (s, 1H), 9.86 (s, 1H), 9.05 (d, J = 2.1 Hz, 1H), 8.77 (d, J = 2.1 Hz, 1H), 7.74- 7.56 (m, 3H), 7.55-7.23 (m, 3H), 3.22-3.00 (m, 2H), 2.45 (s, 3H), 1.92-1.58 (m, 2H), 1.00 (t, J = 7.4 Hz, 3H); [M − H]−= 468.930524.47N-(2,4-difluoro-3-(5-(3- (trifluoromethyl)phenyl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)phenyl)propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 14.99 (s, 1H), 9.83 (s, 1H), 9.10 (d, J = 2.2 Hz, 1H), 8.86 (d, J = 2.2 Hz, 1H), 8.28- 8.03 (m, 2H), 7.88-7.53 (m, 3H), 7.32 (td, J = 8.7, 1.4 Hz, 1H), 3.17-3.05 (m, 2H), 1.88-1.64 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 522.931474.46N-(2,4-difluoro-3-(5-(2- fluorophenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl) phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 15.00 (s, 1H), 9.80 (s, 1H), 8.90 (t, J = 1.9 Hz, 1H), 8.73 (s, 1H), 7.82-7.22 (m, 6H), 3.22-3.01 (m, 2H), 1.91-1.62 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 473.032501.46N-(2,4-difluoro-3-(5-(3- nitrophenyl)-1H-pyrazolo[3,4- b]pyridine-3-carbonyl) phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 14.99 (s, 1H), 9.84 (s, 1H), 9.13 (d, J = 2.2 Hz, 1H), 8.91 (d, J = 2.2 Hz, 1H), 8.64 (t, J = 1.9 Hz, 1H), 8.37-8.25 (m, 1H), 7.84 (t, J = 8.0 Hz, 1H), 7.64 (td, J = 9.0, 6.0 Hz, 1H), 7.32 (td, J = 9.0, 1.3 Hz, 1H), 3.21-3.04 (m, 1H), 1.88-1.61 (m, 1H), 0.97 (t, J = 7.4 Hz, 1H); [M − H]−= 500.033525.35N-(3-(5-(3,4-dichloro-phenyl)- 1H-pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluoro- phenyl)-propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 9.05 (s, 1H), 8.83 (s, 1H), 8.17 (s, 1H), 8.00-7.52 (m, 3H), 7.31 (t, J = 8.8 Hz, 1H), 3.21-2.98 (m, 2H), 2.00-1.58 (m, 2H), 0.97 (t, J = 7.3 Hz, 3H); [M − H]−= 522.934524.47N-(2,4-difluoro-3-(5-(4- (trifluoromethyl)phenyl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)-phenyl)propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 15.03 (s, 1H), 9.85 (s, 1H), 9.10 (d, J = 2.2 Hz, 1H), 8.86 (d, J = 2.2 Hz, 1H), 8.10 (d, J = 8.0 Hz, 2H), 7.90 (d, J = 8.4 Hz, 2H), 7.64 (td, J = 9.0, 5.9 Hz, 1H), 7.32 (td, J = 8.9, 1.4 Hz, 1H), 3.24- 2.96 (m, 2H), 1.89-1.60 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 522.935525.35N-(3-(5-(2,4-dichlorophenyl)- 1H-pyrazolo[3,4-b]pyridine-3- carbonyl)-2,4-difluoro- phenyl)propane-1- sulfonamide1H NMR (200 MHz, DMSO) δ 15.03 (s, 1H), 9.83 (s, 1H), 8.78 (d, J = 2.1 Hz, 1H), 8.64 (d, J = 1.9 Hz, 1H), 7.84 (d, J = 1.7 Hz, 1H), 7.77-7.54 (m, 3H), 7.46-7.18 (m, 1H), 3.24-3.00 (m, 2H), 1.91-1.56 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M + Na]+= 546.836558.91N-(3-(5-(4-chloro-3- (trifluoromethyl)phenyl)-1H- pyrazolo[3,4-13]-pyridine-3- carbonyl)-2,4-difluorophenyl)- propane-1-sulfonamide1H NMR (200 MHz, DMSO) δ 15.00 (s, 1H), 9.83 (s, 1H), 9.10 (d, J = 2.2 Hz, 1H), 8.89 (d, J = 2.0 Hz, 1H), 8.33- 8.10 (m, 2H), 7.89 (d, J = 8.3 Hz, 1H), 7.64 (td, J = 9.0, 6.0 Hz, 1H), 7.31 (td, J = 8.8, 1.2 Hz, 1H), 3.21-3.02 (m, 2H), 1.87-1.62 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H); [M − H]−= 557.037472.47N-(2,4-difluoro-3-(5-(3- hydroxyphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl) phenyl)propane-1-sulfon- amide1H NMR (200 MHz, DMSO) δ 15.21 (s, 1H), 10.04 (s, 2H), 9.32 (d, J = 2.1 Hz, 1H), 9.03 (d, J = 2.1 Hz, 1H), 7.98 (td, J = 9.1, 6.0 Hz, 1H), 7.76-7.43 (m, 4H), 7.29- 7.14 (m, 1H), 3.54-3.34 (m, 2H), 2.19-1.96 (m, 2H), 1.31 (t, J = 7.4 Hz, 3H); [M − H]−= 471.038474.89N-(3-(5-(2-chloro-4- methoxyphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- 2-fluorophenyl)-methane- sulfonamide1H NMR (400 MHz, DMSO-d6) δ = 14.76 (brs, 1H), 9.78 (brs, 1H), 8.69 (d, J = 2.4 Hz, 1H), 8.53 (d, J = 2.0 Hz, 1H), 7.65-7.55 (m, 2H), 7.50 (d, J = 8.8 Hz, 1H), 7.33 (t, J = 8.56 Hz, 1H), 7.21 (d, J = 2.4 Hz, 1H), 7.06 (dd, J = 2.4, 8.8 Hz, 1H), 3.82 (s, 3H), 3.05 (s, 3H). [M + H]+= 475.0439442.44N-(2-fluoro-3-(5-(4-fluoro-2- methylphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)methanesulfonamide1H NMR (400 MHz, DMSO-d6) δ = 14.81 (brs, 1H), 9.82 (brs, 1H), 8.68 (d, J = 2.0 Hz, 1H), 8.47 (d, J = 2.4 Hz, 1H), 7.67-7.56 (m, 2H), 7.43-7.34 (m, 2H), 7.26 (dd, J = 2.4, 9.8 Hz, 1H), 7.17 (dt, J = 2.7, 8.4 Hz, 1H), 3.09 (s, 3H), 2.27 (s, 3H). [M + H]+= 443.05.40468.46N-(3-(5-(2,3-dihydro-benzo- [b][1,4]dioxin-6-yl)-1H- pyrazolo[3,4-b]pyridine-3- carbonyl)-2-fluoro-phenyl)- methanesulfonamide1H NMR (400 MHz, DMSO-d6) δ = 14.73 (s, 1H), 9.84 (s, 1H), 8.94 (d, J = 2.4 Hz, 1H), 8.75-8.59 (m, 1H), 7.69-7.57 (m, 2H), 7.39-7.31 (m, 2H), 7.30-7.24 (m, 1H), 7.02 (d, J = 8.3 Hz, 1H), 4.31 (s, 4H), 3.09 (s, 3H). [M + H]+= 468.90.41516.97N-(3-(5-(2-chloro-4-methoxy- phenyl)-1H-pyrazolo[3,4- b]pyridine-3-carbonyl)-2- fluorophenyl)-butane-1- sulfonamide1H NMR (400 MHz, DMSO-d6) δ = 14.76 (brs, 1H), 9.80 (brs, 1H), 8.67 (d, J = 2.2 Hz, 1H), 8.52 (d, J = 2.0 Hz, 2H), 7.63-7.53 (m, 2H), 7.49 (d, J = 8.6 Hz, 1H), 7.31 (t, , J = 7.9 Hz, 1H), 7.21 (d, J = 2.4 Hz, 1H), 7.06 (dd, J = 2.4, 8.6 Hz, 2H), 3.82 (s, 3H), 3.14-3.07 (m, 2H), 1.70-1.62 (m, 2H), 1.40-1.26 (m, 2H), 0.82 (t, J = 7.3 Hz, 3H). [M + H]+= 51742484.52N-(2-fluoro-3-(5-(4-fluoro-2- methylphenyl)-1H-pyrazo- lo[3,4-b]pyridine-3-carbonyl)- phenyl)butane-1-sulfonamide1H NMR (400 MHz, DMSO-d6) δ = 14.76 (brs, 1H), 9.84 (brs, 1H), 8.68 (d, J = 2.0 Hz, 1H), 8.47 (d, J = 2.0 Hz, 1H), 7.67-7.54 (m, 2H), 7.43-7.31 (m, 2H), 7.30-7.22 (m, 1H), 7.17 (dt, J = 2.5, 8.4 Hz, 1H), 3.23-3.05 (m, 2H), 2.28 (s, 3H), 1.77-1.64 (m, 2H), 1.33-145 (m, 2H), 0.86 (t, J = 7.4 Hz, 3H). [M + H]+= 485.10.43510.54N-(3-(5-(2,3-dihydro-benzo- [b][1,4]dioxin-6-yl)-1H- pyrazolo[3,4-b]-pyridine-3- carbonyl)-2-fluorophenyl)- butane-1-sulfonamide1H NMR (400 MHz, DMSO-d6) δ = 14.68 (brs, 1H), 9.82 (brs, 1H), 8.94 (d, J = 2.0 Hz, 1H), 8.65 (d, J = 2.0 Hz, 1H), 7.66-7.61 (m, 2H), 7.30-7.39 (m, 2H), 7.30-7.25 (m, 1H), 7.02 (d, J = 8.3 Hz, 1H), 4.31 (s, 4H), 3.23- 3.10 (m, 2H), 1.79-1.65 (m, 2H), 1.49-1.32 (m, 2H), 0.86 (t, J = 7.3 Hz, 3H). [M + H]+= 511.10. Example 44: Synthesis of N-(5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)-2,6-difluoro-3-(propylsulfonamido)benzamide Synthesis of 2,6-difluoro-3-(propylsulfonamido)benzoic acid (Int. E) Synthesis of N-(5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)-2,6-difluoro-3-(propylsulfonamido) benzamide Example 45: Synthesis of 5-(4-chlorophenyl)-N-(2,6-difluoro-3-(propylsulfonamido)phenyl)-1H-pyrazolo[3,4-b]pyridine-3-carboxamide Synthesis of N-(3-amino-2,4-difluorophenyl)propane-1-sulfonamide (Intermediate F) Synthesis of 5-(4-chlorophenyl)-N-(2,6-difluoro-3-(propylsulfonamido)phenyl)-1H-pyrazolo[3,4-b]pyridine-3-carboxamide Example 46: Synthesis of N-(3-((5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)thio)-2,4-difluorophenyl)propane-1-sulfonamide Synthesis of N-(2,4-difluoro-3-mercaptophenyl)propane-1-sulfonamide (Intermediate C) Synthesis of N-(3-((5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)thio)-2,4-difluorophenyl)propane-1-sulfonamide Example 47 and 48: Synthesis of N-(3-((5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)sulfinyl)-2,4-difluorophenyl)propane-1-sulfonamide (Expl. 47) and N-(3-((5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)sulfonyl)-2,4-difluorophenyl)propane-1-sulfonamide (Expl. 48) Example 49: Synthesis of N-(3-((5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)(hydroxy)methyl)-2,4-difluorophenyl)propane-1-sulfonamide Synthesis of N-(2,4-difluoro-3-formylphenyl)propane-1-sulfonamide (Intermediate A) Synthesis of N-(3-((5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)(hydroxy)methyl)-2,4-difluorophenyl)propane-1-sulfonamide Example 50: Synthesis of N-(3-((5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)methyl)-2,4-difluorophenyl)propane-1-sulfonamide Example 51 The compounds of Example 51a-51m were prepared according to the procedure, illustrated in scheme 1. (3-Amino-2,6-difluorophenyl)(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (1) was prepared as described in Example 1 (Steps 1-4). The subsequent conversion with a sulfonyl chloride, leading to intermediate 2 was performed in analogy to Example 1, Step 5. The Suzuki coupling reactions to the final products 51a-51m were performed as follows (general procedure): A microwave vessel is charged with a magnetic stir bar, 5-bromo-1H-pyrazolopyridin derivatives, appropriate boronic acid or boronic acid pinacole ester (1.1 eq.) and XPhos Pd G3 or Pd G4 (0.05 eq.) and purged with argon. Degassed 1,4-dioxane (0.4 M) and degassed aqueous 1.5 M K2CO3(3.5 eq., +1 eq. for every acidic functional group) are added and the mixture is heated to 120° C. (100° C. for amides) under microwave irradiation until complete conversion (30 to 60 minutes). After cooling, the mixture is diluted with EtOAc and neutralized with sat. NH4Cl solution (or acidified with 2N HCl for acidic functional groups). The solvents are removed and the product isolated by flash chromatography (using mixtures of DCM, EtOAc and/or MeOH), triturated if necessary and dried at 100° C. in a vacuum oven. Example 51a: 4-(3-(2,6-difluoro-3-((phenylmethyl)sulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)benzenesulfonamide Step 1: N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl]-1-phenylmethanesulfonamide (3-amino-2,6-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (350 mg, 0.991 mmol) and N,N-dimethylpyridin-4-amine (6.05 mg, 0.0496 mmol) were dissolved in pyridine (1.98 mL) and heated to 65° C. Phenylmethanesulfonyl chloride (283 mg, 1.49 mmol) was added and the mixture stirred for 2 h. The mixture was poured into aqueous 2N HCl and extracted with EtOAc. The organic phase was washed with 2N HCl and brine, dried over Na2SO4, filtered and the solvent removed under removed pressure. The residue was purified by flash chromatography (n-hexane+EtOAc) 0% to 50% and triturated with n-hexane. N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl]-1-phenylmethanesulfonamide (315 mg, 0.6210 mmol, 63% yield). 1H NMR (200 MHz, CDCl3) δ 14.22 (s, 1H), 8.96 (s, 1H), 8.79 (d, J=2.2 Hz, 1H), 8.59 (d, J=2.1 Hz, 1H), 7.52-7.25 (m, 6H), 6.87 (td, J=9.1, 1.6 Hz, 1H), 4.32 (s, 2H); 13C NMR (101 MHz, CDCl3) δ 182.7, 156.6 (dd, J=251, 6.8 Hz), 153.03 (d, J=7.7 Hz), 151.2, 150.6, 150.5, 141.6, 132.9, 130.8, 127.75 (dd, J=151.3, 7.9 Hz), 126.9, 121.87 (dd, J=13.1, 3.9 Hz), 117.12 (dd, J=22.8, 20.7 Hz), 115.7, 115.5, 111.61 (dd, J=22.5, 3.8 Hz), 58.8. MS: [M−1]−=504.7 Step 2: Suzuki Coupling According to General Procedure (See Example 2 Above) Analytical Data:1H NMR (200 MHz, DMSO) δ 15.01 (s, 1H), 9.89 (s, 1H), 9.11 (d, J=1.7 Hz, 1H), 8.87 (d, J=1.7 Hz, 1H), 8.17-7.81 (m, 4H), 7.70-7.14 (m, 9H), 4.54 (s, 2H);13C NMR (101 MHz, DMSO) δ 182.6, 155.95 (dd, J=248.0, 6.2 Hz), 152.4, 152.14 (dd, J=251.0, 7.4 Hz), 149.8, 143.5, 142.0, 140.4, 132.0, 131.9, 131.5, 131.5, 131.4, 130.9, 129.2, 128.7, 128.6, 128.3, 128.3, 127.9, 127.5, 126.4, 126.3, 122.10 (dd, J=13.1, 3.5 Hz), 117.2, 117.0, 113.4, 111.90 (dd, J=22.0, 3.9 Hz) 58.5; MS: [M−1]−=581.8. Example 51b: 4-(3-(2,6-difluoro-3-(methylsulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl) benzenesulfonamide Step 1: N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl] methane-sulfonamide (3-amino-2,6-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (350 mg, 0.991 mmol) and N,N-dimethylpyridin-4-amine (6.05 mg, 0.0496 mmol) were dissolved in pyridine (1.98 mL) and heated to 65° C. Methanesulfonyl chloride (0.357 mL, 1.49 mmol) was added and the mixture stirred for 2 h. The mixture was poured into aqueous 2N HCl and extracted with EtOAc. The organic phase was washed with 2N HCl and brine, dried over Na2SO4, filtered and the solvent removed under removed pressure. The residue was purified by flash chromatography (DCM+EtOAc) 0% to 30% and triturated with n-hexane. N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl]methanesulfonamide (164 mg, 0.3800 mmol, 38% yield) Analytical Data:1H NMR (200 MHz, DMSO) δ 15.07 (s, 1H), 9.82 (s, 1H), 8.80 (dd, J=4.8, 2.1 Hz, 2H), 7.65 (td, J=9.0, 5.9 Hz, 1H), 7.32 (td, J=8.9, 1.5 Hz, 1H), 3.07 (s, 3H);13C NMR (101 MHz, DMSO) δ 182.3, 156.4 (dd, J=248.8, 6.6 Hz), 152.92 (dd, J=251.8, 8.1 Hz), 151.0, 150.9, 141.0, 132.2, 130.1, 130.0, 121.80 (dd, J=13.2, 3.7 Hz), 116.84 (dd, J=23.0, 20.9 Hz), 115.7, 114.7, 112.16 (dd, J=22.4, 3.7 Hz), 40.4; MS: [M−1]−=428.7. Step 2: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (200 MHz, DMSO) δ 14.99 (s, 1H), 9.85 (s, 1H), 9.10 (d, J=2.1 Hz, 1H), 8.86 (d, J=2.1 Hz, 1H), 8.03 (dd, J=20.8, 8.4 Hz, 4H), 7.65 (td, J=9.1, 6.1 Hz, 1H), 7.48 (s, 2H), 7.33 (t, J=8.8 Hz, 1H), 3.08 (s, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 156.40 (dd, J=248.5, 6.7 Hz), 153.0 (dd, J=251.5, 8.0 Hz), 152.4, 149.8, 143.5, 142.0, 140.3, 131.5, 130.0, 129.9, 128.2, 127.9, 126.4, 121.8 (dd, J=13.3, 3.5 Hz), 117.13 (dd, J=23.0, 21.5 Hz), 113.4, 112.15 (dd, J=22.1, 3.1 Hz), 40.4; MS: [M−1]−=505.9. Example 51c: 4-(3-(3-(butylsulfonamido)-2,6-difluorobenzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl) benzenesulfonamide Step 1: N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl]butane-1-sulfonamide (3-amino-2,6-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (350 mg, 0.991 mmol) and N,N-dimethylpyridin-4-amine (6.05 mg, 0.0496 mmol) were dissolved in pyridine (1.98 mL) and heated to 65° C. Butane-1-sulfonyl chloride (0.357 mL, 1.49 mmol) was added and the mixture stirred for overnight. 0.25 eq. butane-1-sulfonyl chloride were added and stirring continued for 2 h at 65° C. The mixture was poured into aqueous 2N HCl and extracted with EtOAc. The organic phase was washed with 2N HCl and brine, dried over Na2SO4, filtered and the solvent removed under removed pressure. The residue was purified by flash chromatography (hexane+EtOAc) 10% to 50% and triturated with n-hexane. N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl]butane-1-sulfonamide (180 mg, 0.3800 mmol, 38% yield) Analytical Data:1H NMR (200 MHz, DMSO) δ 15.03 (s, 1H), 9.83 (s, 1H), 9.02-8.62 (m, 2H), 3.21-3.03 (m, 2H), 1.70 (dt, J=15.0, 7.5 Hz, 2H), 1.37 (dq, J=14.5, 7.3 Hz, 2H), 0.84 (t, J=7.2 Hz, 3H);13C NMR (50 MHz, DMSO) δ 182.4, 151.1, 151.0, 141.1, 132.3, 115.8, 114.8, 51.8, 25.1, 20.7, 13.4; MS: [M−1]−=470.8. Step 2: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (200 MHz, DMSO) δ 15.00 (s, 1H), 9.82 (s, 1H), 9.10 (d, J=2.1 Hz, 1H), 8.86 (d, J=2.0 Hz, 1H), 8.13-7.93 (m, 4H), 7.64 (td, J=9.0, 6.2 Hz, 1H), 7.48 (s, 2H), 7.32 (td, J=8.9, 1.2 Hz, 1H), 3.21-3.07 (m, 2H), 1.82-1.61 (m, 2H), 1.50-1.27 (m, 2H), 0.85 (t, J=7.2 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 156.3 (dd, J=248.2, 6.3 Hz), 152.7 (dd, J=251.1, 8.8 Hz), 152.4, 149.8, 143.5, 142.0, 140.3, 131.5, 129.9, 129.8, 128.2, 127.9, 127.5, 126.4, 126.3, 121.8 (dd, J=13.0, 3.7 Hz), 117.08 (dd, J=23.1, 21.2 Hz), 113.4, 112.2, 112.2, 112.0, 51.8, 25.1, 20.7, 13.4; MS: [M−1]−=547.9. Example 51d: 4-(3-(2,6-difluoro-3-(propysulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)benzoic acid Suzuki Coupling According to General Procedure Analytical Data:1H NMR (200 MHz, DMSO) δ 14.99 (s, 1H), 13.14 (bs, 1H), 9.83 (s, 1H), 9.10 (d, J=2.1 Hz, 1H), 8.84 (d, J=2.2 Hz, 1H), 8.04 (dd, J=22.3, 8.5 Hz, 4H), 7.64 (td, J=9.0, 5.9 Hz, 1H), 7.31 (td, J=8.9, 1.5 Hz, 1H), 3.21-3.03 (m, 2H), 1.86-1.61 (m, 2H), 0.97 (t, J=7.4 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 167.0, 152.4, 149.8, 142.0, 141.3, 131.8, 130.2, 130.1, 128.1, 127.6, 113.5, 53.8, 16.8, 12.5; MS: [M−1]−=499.6. Example 51e: N-[2,4-difluoro-3-[5-[4-(1H-tetrazol-5-yl)phenyl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl] phenyl]propane-1-sulfonamide Suzuki Coupling According to General Procedure Analytical Data:1H NMR (200 MHz, DMSO) δ 14.99 (s, 1H), 9.83 (s, 1H), 9.14 (d, J=2.1 Hz, 1H), 8.88 (d, J=2.2 Hz, 1H), 8.17 (dd, J=19.9, 8.4 Hz, 4H), 7.65 (td, J=9.0, 6.1 Hz, 1H), 7.32 (td, J=9.0, 1.4 Hz, 1H), 3.20-3.05 (m, 2H), 1.90-1.59 (m, 2H), 0.97 (t, J=7.4 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 156.29 (dd, J=248.3, 6.4 Hz), 152.8 (dd, J=251.2, 8.2 Hz), 152.4, 149.8, 142.0, 139.7, 131.7, 129.9, 129.8, 128.3, 127.9, 127.7, 127.6, 123.8, 121.79 (dd, J=13.4, 3.4 Hz), 117.1, 113.5, 112.12 (dd, J=22.6, 4.3 Hz), 53.8, 16.8, 12.5; MS: [M−1]−=522.9. Example 51f: 4-(3-(2,6-difluoro-3-(propysulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl) benzamide Suzuki Coupling According to General Procedure Analytical Data:1H NMR (400 MHz, DMSO) δ 14.97 (s, 1H), 9.83 (s, 1H), 9.10 (d, J=1.3 Hz, 1H), 8.83 (d, J=1.3 Hz, 1H), 8.13-7.92 (m, 5H), 7.64 (td, J=8.9, 6.2 Hz, 1H), 7.46 (s, 1H), 7.32 (t, J=8.7 Hz, 1H), 3.18-3.06 (m, 2H), 1.83-1.68 (m, 2H), 0.97 (t, J=7.5 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.6, 167.4, 152.4, 149.8, 142.0, 139.8, 133.7, 132.0, 128.4, 127.9, 127.2, 113.5, 53.8, 16.8, 12.6; MS: [M−1]−=498.0. Example 51g: N-(2,4-difluoro-3-(5-(4-(methylsulfonamido)phenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl)propane-1-sulfonamide Step 1: N-(4-bromophenyl)methanesulfonamide (2) 4-bromoaniline (1, 2.08 g, 12.1 mmol) and 4-Dimethylaminopyridine (0.0739 g, 0.605 mmol) were dissolved in pyridine (12.1 mL) and methanesulfonyl chloride (1.03 mL, 13.3 mmol) was added at RT leading to an exothermic reaction. After RT was reached again (30 min) the mixture was poured into 2N HCl. The product was extracted with EtOAc, the extract washed with 2N HCl and brine, dried over Na2SO4, filtered and the solvent removed under reduced pressure and the residue triturated with n-hexane to yield N-(4-bromophenyl)methanesulfonamide (2.40 g, 9.6 mmol, 79% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 9.91 (s, 1H), 7.60-7.42 (m, 2H), 7.22-7.09 (m, 2H), 3.00 (s, 3H);13C NMR (50 MHz, DMSO) δ 137.9, 132.2, 121.5, 115.9, 39.3; [M−1]−=247.7. Step 2: N-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methanesulfonamide (3) A vessel was charged with N-(4-bromophenyl)methanesulfonamide (2, 252 mg, 1.01 mmol), Bis(pinacolato)diboron (281 mg, 1.11 mmol), Potassium acetate (297 mg, 3.02 mmol) and degassed dry 1,4-dioxane (5.04 mL). The vessel was evacuated and backfilled with argon (3×), XPhos Pd G4 (8.67 mg, 0.0101 mmol) was added and the mixture stirred at 85° C. for 4 h. After cooling to RT, the mixture was diluted with EtOAc and Acetic Acid (0.173 mL, 3.02 mmol), stirred for 30 minutes and filtered over Celite. The solvent was removed under reduced pressure. The residue was dissolved in a minimum amount of EtOAc, precipitated with n-heptane and the solids collected by suction filtration to yield N-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]methanesulfonamide (281 mg, 0.9460 mmol, 94% yield) which was used without further purification. Analytical Data:1H NMR (200 MHz, DMSO) δ 7.60 (d, J=8.5 Hz, 2H), 7.17 (d, J=8.5 Hz, 2H), 2.99 (s, 3H), 1.27 (s, 12H);13C NMR (50 MHz, DMSO) δ 142.4, 135.8, 117.9, 83.5, 24.7. Step 3: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (200 MHz, DMSO) δ 14.93 (s, 1H), 9.98 (s, 1H), 9.83 (s, 1H), 9.02 (d, J=2.2 Hz, 1H), 8.73 (d, J=2.1 Hz, 1H), 7.84 (d, J=8.6 Hz, 2H), 7.64 (td, J=9.0, 6.1 Hz, 1H), 7.43-7.26 (m, 3H), 3.19-3.03 (m, 5H), 1.86-1.63 (m, 2H), 0.97 (t, J=7.4 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 152.1, 149.6, 141.8, 138.5, 132.4, 128.3, 127.0, 120.0, 113.5, 53.8, 16.8, 12.5; [M−1]−=547.8. Example 51h: 4-(3-(2,6-difluoro-3-(propysulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)-N-methylbenzenesulfonamide Step 1: 4-bromo-N-methylbenzenesulfonamide (2) To a solution of 4-bromobenzenesulfonyl chloride (1, 2.52 g, 9.86 mmol) in tetrahydrofuran (49.3 mL) was added methylamine (14.8 mL, 29.6 mmol) 2M in THE. The reaction mixture was stirred at RT for 15 min, poured into NH4Cl solution and extracted with EtOAc. The organic layer was dried over Na2SO4, concentrated and triturated with n-hexane to furnish 4-bromo-N-methylbenzenesulfonamide (2.15 g, 8.6 mmol, 87% yield) Analytical Data:1H NMR (200 MHz, CDCl3) δ 7.77-7.59 (m, 1H), 5.03 (dd, J=10.0, 4.9 Hz, 1H), 2.62 (d, J=5.2 Hz, 1H);13C NMR (50 MHz, CDCl3) δ 137.8, 132.5, 128.9, 127.8, 29.3; [M−1]−=247.8. Step 2: [4-(methylsulfamoyl)phenyl]boronic acid (3) To a solution of 4-bromo-N-methylbenzenesulfonamide (2, 1.05 g, 4.20 mmol) and triisopropyl borate (1.45 mL, 6.30 mmol) in tetrahydrofuran (8.40 mL) was added n-Butyl Lithium (4.20 mL, 10.5 mmol) at −70° C. The mixture was slowly warmed to 0° C., then 10% HCl solution was added until pH 3-4. The resulting mixture was extracted with EtOAc. The organic layer was extracted with NaOH (2M) and the aqueous phase washed with diethyl ether. The aqueous phase was acidified to pH 3, extracted with EtOAc (impurities and unconsumed reactant were still present) was dried over Na2SO4, and evaporated under reduced pressure. The residue was triturated with diethyl ether to give [4-(methylsulfamoyl)phenyl]boronic acid (187 mg, 0.8700 mmol, 21% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 7.96 (d, J=8.1 Hz, 2H), 7.72 (d, J=8.0 Hz, 2H), 7.42 (q, J=4.9 Hz, 1H), 2.39 (d, J=5.0 Hz, 3H);13C NMR (50 MHz, DMSO) δ 140.6, 134.8, 125.7, 28.8. Step 3: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (400 MHz, DMSO) δ 15.01 (s, 1H), 9.83 (s, 1H), 9.11 (d, J=2.1 Hz, 1H), 8.87 (d, J=2.0 Hz, 1H), 8.11 (d, J=8.3 Hz, 2H), 7.93 (d, J=8.3 Hz, 2H), 7.64 (td, J=9.0, 5.9 Hz, 1H), 7.58 (q, J=5.0 Hz, 1H), 7.32 (t, J=8.7 Hz, 1H), 3.19-3.02 (m, 2H), 1.80-1.70 (m, 2H), 0.97 (t, J=7.5 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.6, 152.5, 149.9, 142.0, 141.0, 138.8, 131.4, 128.4, 128.2, 127.5, 113.4, 53.8, 28.6, 16.8, 12.5; MS: [M−1]−=547.9. Example 51i: 4-(3-(2,6-difluoro-3-(propysulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)-N-ethylbenzenesulfonamide Step 1: 4-bromo-N-ethylbenzenesulfonamide (2) 4-bromobenzenesulfonyl chloride (1, 2.57 g, 10.1 mmol) was dissolved in DCM (25.1 mL). Triethylamine (3.50 mL, 25.1 mmol) and ethylamine hydrochloride (1.07 g, 13.1 mmol) were added and the mixture stirred at RT for 1 h. The mixture was diluted with half sat. NH4Cl solution and extracted with EtOAc. The organic phase was washed with brine, dried over Na2SO4, filtered and the solvent removed under reduced pressure. The residue was triturated with n-hexane to yield 4-bromo-N-ethylbenzenesulfonamide (2.40 g, 9.09 mmol, 90% yield). Analytical Data:1H NMR (200 MHz, CDCl3) δ 7.92-7.47 (m, 4H), 5.17 (s, 1H), 3.08-2.81 (m, 2H), 1.08 (t, J=7.2 Hz, 3H);13C NMR (50 MHz, CDCl3) δ 139.1, 132.4, 128.7, 127.6, 38.3, 15.1; [M−1]−=261.7. Step 2: [4-(ethylsulfamoyl)phenyl]boronic acid (3) To a solution of 4-bromo-N-ethylbenzenesulfonamide (2, 1.06 g, 4.01 mmol) and triisopropyl borate (1.39 mL, 6.02 mmol) in tetrahydrofuran (20.1 mL) was added n-Butyl Lithium (4.01 mL, 10.0 mmol) at −70° C. The mixture was slowly warmed to 0° C., then 10% HCl solution was added until pH 3-4. The resulting mixture was extracted with EtOAc and the extract washed with brine, dried over Na2SO4, filtered and the solvent removed under reduced pressure. The residue was triturated with diethyl ether to yield [4-(ethylsulfamoyl)phenyl]boronic acid (368 mg, 1.61 mmol, 40% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 8.41 (s, 2H), 7.94 (d, J=8.1 Hz, 2H), 7.74 (d, J=8.1 Hz, 2H), 7.52 (t, J=5.7 Hz, 1H), 2.87-2.67 (m, 2H), 0.93 (t, J=7.2 Hz, 3H);13C NMR (50 MHz, DMSO) δ 142.1, 135.0, 125.7, 37.9, 15.0. Step 3: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (400 MHz, DMSO) δ 14.97 (s, 1H), 9.82 (s, 1H), 9.10 (d, J=2.1 Hz, 1H), 8.86 (d, J=2.1 Hz, 1H), 8.09 (d, J=8.3 Hz, 2H), 7.94 (d, J=8.5 Hz, 2H), 7.73-7.58 (m, 2H), 7.31 (t, J=8.7 Hz, 1H), 3.16-3.06 (m, 2H), 2.89-2.80 (m, 2H), 1.81-1.69 (m, 2H), 1.05-0.94 (m, 6H);13C NMR (101 MHz, DMSO) δ 182.5, 152.5, 149.8, 142.0, 140.8, 140.0, 131.4, 128.3, 128.2, 127.3, 113.4, 53.8, 37.6, 16.8, 14.8, 12.5; MS: [M−1]−=561.9. Example 51j: 4-(3-(2,6-difluoro-3-(propysulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)-N-(2,3-dihydroxypropyl)benzenesulfonamide Step 1: 4-bromo-N-(2,3-dihydroxypropyl)benzenesulfonamide (2) 4-bromobenzenesulfonyl chloride (1, 2.10 g, 8.22 mmol) and triethylamine (2.29 mL, 16.4 mmol) were combined in DCM (41.1 mL). 3-aminopropane-1,2-diol (0.952 mL, 12.3 mmol) was added and the mixture stirred for 1 h at RT. After evaporation of the solvent, the residue was dissolved in EtOAc, washed with 1N HCl, water and brine. The extract was dried over Na2SO4, filtered and the solvent removed under reduced pressure. The product was washed with water and diethyl ether and dried to yield 4-bromo-N-(2,3-dihydroxypropyl)benzenesulfonamide (0.980 g, 3.16 mmol, 38% yield) Analytical Data:1H NMR (200 MHz, DMSO) δ 7.77 (dd, J=17.1, 8.5 Hz, 5H), 4.79 (s, 1H), 4.56 (s, 1H), 3.46 (s, 1H) under water peak, 3.27 (s, 2H), 2.89 (dd, J=12.6, 4.2 Hz, 1H), 2.61 (dd, J=12.5, 7.1 Hz, 1H);13C NMR (50 MHz, DMSO) δ 139.9, 132.3, 128.7, 126.1, 70.3, 63.5, 46.1; [M−1]−=307.8. Step 2: N-(2,3-dihydroxypropyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzenesulfonamide (3) A vessel was charged with 4-bromo-N-(2,3-dihydroxypropyl)benzenesulfonamide (2, 205 mg, 0.661 mmol), Potassium acetate (195 mg, 1.98 mmol), Bis(pinacolato)diboron (185 mg, 0.727 mmol) and degassed dry 1,4-dioxane (6.61 mL). The vessel was evacuated and backfilled with argon (3×), XPhos Pd G4 (5.69 mg, 0.00661 mmol) was added and the mixture stirred at 85° C. overnight. After cooling to RT, the mixture was diluted with EtOAc and stirred for 30 minutes, filtered over Celite and the solvent was removed under reduced pressure. The residue was dissolved in a minimum amount of EtOAc, precipitated with n-heptane and the solids collected by suction filtration to yield N-(2,3-dihydroxypropyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzenesulfonamide (202 mg, 0.5650 mmol, 86% yield) which was used without further purification. Analytical Data:1H NMR (200 MHz, DMSO) δ 7.80 (q, J=8.0 Hz, 3H), 7.44 (s, 2H), 3.52-2.56 (m, 5H), 1.35-1.22 (m, 7H), 1.15 (s, 5H), 1.07 (s, 2H). Step 3: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (400 MHz, DMSO) δ 15.00 (s, 1H), 9.82 (s, 1H), 9.11 (d, J=2.1 Hz, 1H), 8.87 (d, J=2.1 Hz, 1H), 8.09 (d, J=8.5 Hz, 2H), 7.95 (d, J=8.5 Hz, 2H), 7.68-7.60 (m, 2H), 7.32 (t, J=8.3 Hz, 1H), 4.82 (d, J=5.2 Hz, 1H), 4.58 (t, J=5.7 Hz, 1H), 3.50 (dq, J=10.6, 5.3 Hz, 1H), 3.32-3.22 (m, 2H), 3.14-3.08 (m, 2H), 2.94 (ddd, J=11.7, 6.6, 4.9 Hz, 1H), 2.66 (ddd, J=12.8, 7.0, 5.8 Hz, 1H), 1.83-1.69 (m, 2H), 0.97 (t, J=7.5 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 152.5, 149.8, 142.0, 140.8, 140.0, 131.4, 128.3, 128.1, 127.3, 113.4, 70.3, 63.5, 53.8, 46.1, 16.8, 12.5; MS: [M−1]−=607.8. Example 51k: N-((4-(3-(2,6-difluoro-3-(propylsulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)phenyl)sulfonyl)acetamide Step 1: N-(4-bromophenyl)sulfonylacetamide (2) 4-bromobenzenesulfonyl chloride (1, 2.32 g, 9.08 mmol) and acetamide (1.34 g, 22.7 mmol) (washed with diethyl ether prior to use) were dissolved in tetrahydrofuran (30.3 mL). Sodium Hydride (0.908 g, 22.7 mmol) (60%) was added portion wise at 0° C. and the mixture stirred for 2 h. The mixture was acidified with conc. HCl and water was added until phase separation occurred. The phases were separated and the aqueous extracted with EtOAc. The organic phase was washed with water and brine, dried over Na2SO4, filtered and the solvent removed under reduced pressure. The residue was triturated with water and dried in vacuo to yield N-(4-bromophenyl)sulfonylacetamide (1.33 g, 4.78 mmol, 53% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 12.22 (s, 1H), 7.84 (s, 4H), 1.93 (s, 3H);13C NMR (50 MHz, DMSO) δ 169.0, 138.6, 132.3, 129.6, 127.7, 23.3; [M−1]−=275.8. Step 2: N-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]sulfonylacetamide (3) A vessel was charged with N-(4-bromophenyl)sulfonylacetamide (2, 206 mg, 0.741 mmol), Potassium acetate (218 mg, 2.22 mmol), Bis(pinacolato)diboron (207 mg, 0.815 mmol) and degassed dry 1,4-dioxane (7.41 mL). The vessel was evacuated and backfilled with argon (3×), XPhos Pd G4 (6.37 mg, 0.00741 mmol) was added and the mixture stirred at 85° C. overnight. Additional XPhos Pd G4 and Bis(pinacolato)diboron were added and the mixture stirred for another 2 h. After cooling to RT, the mixture was diluted with EtOAc and stirred for 30 minutes and filtered over Celite. The filtrate was discarded and the filter washed with 2N HCl and EtOAc. The solvent was removed under reduced pressure and the residue triturated with n-heptane to yield N-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]sulfonylacetamide (163 mg, 0.5010 mmol, 68% yield) which was used without further purification. Analytical Data:1H NMR (200 MHz, DMSO) δ 12.14 (s, 1H), 8.02-7.81 (m, 4H), 1.92 (s, 3H), 1.30 (s, 12H);13C NMR (50 MHz, DMSO) δ 168.8, 141.8, 134.9, 126.8, 84.3, 24.6, 23.2. Step 3: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (400 MHz, DMSO) δ 15.01 (s, 1H), 12.21 (s, 1H), 9.83 (s, 1H), 9.11 (d, J=2.1 Hz, 1H), 8.87 (d, J=2.1 Hz, 1H), 8.12 (d, J=8.5 Hz, 2H), 8.05 (d, J=8.5 Hz, 2H), 7.64 (td, J=9.0, 5.9 Hz, 1H), 7.32 (t, J=8.5 Hz, 1H), 3.11 (dd, J=5.7, 3.8 Hz, 2H), 1.96 (s, 3H), 1.83-1.69 (m, 2H), 0.97 (t, J=7.5 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 168.9, 152.5, 149.9, 142.1, 138.8, 131.2, 128.6, 128.3, 128.1, 113.4, 53.8, 23.2, 16.8, 12.5; MS: [M−1]−=575.7. Example 51l: 4-(3-(2,6-difluoro-3-(propylsulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)-N-(2,3-dihydroxypropyl)benzamide Step 1: 4-bromo-N-(2,3-dihydroxypropyl)benzamide (2) Methyl 4-bromobenzoate (2.41 g, 11.2 mmol) and 3-aminopropane-1,2-diol (1.12 g, 12.3 mmol) were heated to 125° C. for 4 h. The mixture was diluted with MeOH, evaporated over Celite and purified over a short column. (DCM+MeOH+formic acid (99+0+1 to 90+9+1). After evaporation of the solvents, the oily residue was dried at 0.05 mbar until crystallization occurred. It was then triturated with diethyl ether to yield 4-bromo-N-(2,3-dihydroxypropyl)benzamide (1.90 g, 6.93 mmol, 62% yield) as white solid.1H NMR (200 MHz, DMSO) δ 8.49 (s, 1H), 7.73 (d, J=22.1 Hz, 4H), 4.72 (d, J=48.1 Hz, 2H), 3.83-2.99 (m, 5H);13C NMR (50 MHz, DMSO) δ 165.8, 133.7, 131.3, 129.5, 124.9, 70.4, 64.0, 43.2; [M−1]−=271.7. Step 2: N-(2,3-dihydroxypropyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzamide (3) A vessel was charged with 4-bromo-N-(2,3-dihydroxypropyl)benzamide (656 mg, 2.39 mmol), Bis(pinacolato)diboron (669 mg, 2.63 mmol), Potassium acetate (705 mg, 7.18 mmol) and degassed dry 1,4-dioxane (12.0 mL). The vessel was evacuated and backfilled with argon (3×), XPhos Pd G3 (10.3 mg, 0.0120 mmol) was added and the mixture stirred at 85° C. 2 h. After cooling to RT, the mixture was diluted with EtOAc, stirred for 30 minutes and filtered over Celite. The solvent was removed under reduced pressure. The residue was dissolved in a minimum amount of EtOAc, added dropwise to n-heptane with stirring and the solids collected by suction filtration to yield N-(2,3-dihydroxypropyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzamide (410 mg, 1.28 mmol, 53% yield) which was used without further purification. Analytical Data:1H NMR (200 MHz, DMSO) δ 7.91-7.65 (m, 4H), 1.30 (s, 12H), 1.15 (s, 4H), 1.07 (s, 2H);13C NMR (101 MHz, DMSO) δ 134.2, 126.5, 83.8, 81.3, 73.5, 70.3, 64.0, 39.5, 24.9, 24.6, 24.4. Step 3: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (200 MHz, DMSO) δ 9.10 (d, J=2.1 Hz, 1H), 8.83 (d, J=2.2 Hz, 1H), 8.54 (t, J=5.7 Hz, 1H), 8.00 (q, J=8.5 Hz, 4H), 7.64 (td, J=9.0, 5.8 Hz, 1H), 7.32 (td, J=9.1, 1.3 Hz, 1H), 4.87 (d, J=5.1 Hz, 1H), 4.62 (t, J=5.7 Hz, 1H), 3.80-3.58 (m, 1H), 3.26-3.02 (m, 3H), 1.75 (dq, J=14.9, 7.4 Hz, 2H), 0.97 (t, J=7.4 Hz, 3H);13C NMR (50 MHz, CDCl3) δ 182.6, 166.1, 152.5, 142.0, 139.7, 133.9, 132.0, 128.2, 127.3, 113.6, 70.4, 64.0, 53.8, 43.1, 16.9, 12.6; MS: [M−1]−=571.7. Example 51m: 4-(3-(2,6-difluoro-3-(propylsulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)-3-fluorobenzenesulfonamide Step 1: 4-bromo-3-fluorobenzenesulfonamide (2) To an ice cooled solution of 4-bromo-3-fluorobenzenesulfonyl chloride (1, 1.03 g, 3.77 mmol) in acetonitrile (1.88 mL) was added 25% ammonia solution (1.46 mL, 9.41 mmol) dropwise. The mixture was stirred for 10 min at RT, diluted with water and extracted with diethyl ether. The extract was washed with brine, dried over Na2SO4, filtered and the solvent removed under reduced pressure to yield 4-bromo-3-fluorobenzenesulfonamide (0.780 g, 3.07 mmol, 82% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 7.95 (dd, J=8.2, 6.9 Hz, 1H), 7.75 (dd, J=8.4, 2.1 Hz, 1H), 7.64-7.57 (m, 3H);13C NMR (50 MHz, DMSO) δ 157.9 (d, J=249.0 Hz), 145.5 (d, J=6.1 Hz), 134.6, 123.2 (d, J=3.8 Hz), 114.1 (d, J=25.3 Hz), 112.3 (d, J=20.8 Hz). Step 2: 3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzenesulfonamide (3) A vessel was charged with 4-bromo-3-fluorobenzenesulfonamide (2, 255 mg, 1.00 mmol), Bis(pinacolato)diboron (280 mg, 1.10 mmol), Potassium acetate (295 mg, 3.01 mmol) and degassed dry 1,4-dioxane (5.02 mL). The vessel was evacuated and backfilled with argon (3×), XPhos Pd G4 (4.32 mg, 0.00502 mmol) was added and the mixture stirred at 85° C. for overnight. After cooling to RT, the mixture was diluted with EtOAc and Acetic Acid (0.172 mL, 3.01 mmol), stirred for 30 minutes and filtered over Celite. The solvent was removed under reduced pressure. The residue was dissolved in a minimum amount of EtOAc, precipitated with n-heptane and the solids collected by suction filtration to yield 3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzenesulfonamide (220 mg, 0.7310 mmol, 73% yield) which was used without further purification. Analytical Data:1H NMR (200 MHz, DMSO) δ 7.91-7.09 (m, 5H), 1.42-0.95 (m, 12H);13C NMR (50 MHz, DMSO) δ 148.6 (d, J=8.2 Hz), 137.5 (d, J=8.3 Hz), 121.05 (d, J=2.1 Hz), 112.48 (d, J=27.6 Hz), 83.9, 73.6, 25.0, 24.7. Step 3: Suzuki Coupling According to General Procedure Analytical Data:1H NMR (200 MHz, DMSO) δ 15.06 (s, 1H), 9.83 (s, 1H), 8.95 (s, 1H), 8.81 (s, 1H), 7.99 (t, J=8.0 Hz, 1H), 7.88-7.74 (m, 2H), 7.73-7.55 (m, 3H), 7.40-7.23 (m, 1H), 3.22-2.98 (m, 2H), 1.88-1.59 (m, 2H), 0.97 (t, J=7.4 Hz, 3H). MS: [M−1]−=551.7. Example 52 The compounds of Examples 52a-52c were prepared according to the procedure, illustrated in scheme 2. Step 1: N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4-difluorophenyl]propane-1-sulfonamide (2) To a suspensions of N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,4-difluorophenyl] propane-1-sulfonamide (1, 0.333 g, 0.725 mmol) in DCM (2.90 mL) was added dihydropyran (0.132 mL, 1.45 mmol) and p-toluenesulfonic acid monohydrate (0.0276 g, 0.145 mmol) and the mixture was heated to reflux temperature for 45 minutes. After cooling, the mixture was washed with sat. NaHCO3-solution, dried over Na2SO4, filtered and the solvent was removed under reduced pressure. The residue was dissolved in minimum amount of DCM and added dropwise to n-hexane with stirring. After 5 minutes the solids were collected by suction filtration and dried to yield N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4-difluorophenyl] propane-1-sulfonamide (0.297 g, 0.5470 mmol, 75% yield), which was used without further purification. Analytical Data:1H NMR (200 MHz, CDCl3) δ 8.83 (s, 1H), 8.65 (s, 1H), 7.70 (dd, J=13.6, 8.1 Hz, 1H), 7.02 (d, J=9.6 Hz, 2H), 6.14 (d, J=9.2 Hz, 1H), 4.14-3.64 (m, 2H), 3.22-2.90 (m, 2H), 2.61-2.31 (m, 1H), 2.15-1.16 (m, 10H);13C NMR (50 MHz, CDCl3) δ 182.4, 151.0, 149.7, 140.7, 133.7, 127.33 (d, J=8.6 Hz), 121.36 (dd, J=13.1, 3.8 Hz), 116.8, 116.6, 112.44 (dd, J=22.6, 3.7 Hz), 83.3, 77.2, 68.2, 54.1, 28.9, 24.8, 22.4, 17.3, 12.9; MS: [M−1]−=540.7. Step 2: N-[2,4-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (3) A vessel was charged with N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4-difluorophenyl]propane-1-sulfonamide (2, 271 mg, 0.499 mmol), Bis(pinacolato)diboron (139 mg, 0.549 mmol) and anhydrous Potassium acetate (147 mg, 1.50 mmol). Degassed, dry 1,4-dioxane (4.99 mL) was added and the vessel evacuated and refilled with argon (3×). 1,1′-Bis(diphenylphosphino)ferrocene-dichloropalladium (1:1) (9.12 mg, 0.0125 mmol) was added and the mixture stirred at 85° C. for overnight. After cooling to RT the mixture was diluted with EtOAc, filtered over Celite and the solvent was removed. The residue was dissolved in DCM, petrol ether (60/90) was added and DCM removed under reduced pressure. After cooling for 1 h at 4° C. the solids were collected by suction filtration and dried to yield N-[2,4-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (267 mg, 0.4520 mmol, 91% yield) which was used without further purification. Analytical Data:1H NMR (200 MHz, CDCl3) δ 9.11 (s, 1H), 8.94 (s, 1H), 7.68 (d, J=5.9 Hz, 1H), 7.12-6.83 (m, 2H), 6.22 (d, J=9.0 Hz, 1H), 4.19-3.60 (m, 2H), 3.07 (s, 2H), 2.57-1.11 (m, 23H);13C NMR (50 MHz, CDCl3) δ 182.5, 155.5, 152.4, 141.9, 139.3, 127.3 (d, J=9.0 Hz), 121.23 (dd, J=13.1, 4.0 Hz), 114.9, 112.33 (dd, J=23.2, 2.9 Hz), 84.4, 82.8, 77.2, 68.1, 54.1, 29.0, 24.9, 22.5, 17.2, 12.9; MS: [M−1]−=588.9. Example 52a: 3-chloro-4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide Step 1: 4-bromo-3-chlorobenzenesulfonamide (2) To an ice cooled solution of 4-bromo-3-chlorobenzenesulfonyl chloride (1, 0.450 g, 1.55 mmol) in acetonitrile (7.76 mL) was added 25% ammonia solution (0.602 mL, 3.88 mmol) dropwise. The mixture was stirred for 10 min at RT, diluted with water and extracted with EtOAc. The extract was washed with brine, dried over Na2SO4, filtered and the solvent removed under reduced pressure to yield 4-bromo-3-chlorobenzenesulfonamide (0.370 g, 1.37 mmol, 88% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 8.01 (d, J=8.1 Hz, 2H), 7.89-7.47 (m, 3H);13C NMR (50 MHz, DMSO) δ 144.9, 134.8, 133.8, 127.3, 125.8, 125.5; MS: [M−1]−=267.7. Step 2: 3-chloro-4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (3) A vessel was charged with N-[2,4-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (71.0 mg, 0.120 mmol), Tetrakis Pd (6.95 mg, 0.00601 mmol) and 4-bromo-3-chlorobenzenesulfonamide (2, 39.0 mg, 0.144 mmol) and purged with argon. Degassed 1,4-dioxane (0.401 mL) and degassed aqueous 1.5M potassium carbonate (0.240 mL, 0.361 mmol) were added and the vessel was evacuated and backfilled with argon (3×). The mixture was heated to 55° C. for 1 h. The mixture was cooled to RT, diluted with iPrOH, conc. HCl was added until the mixture was strongly acidic and stirring was continued at 70° C. overnight. After cooling to RT, solid NaHCO3was added to neutralize the mixture, the solvents were removed and the residue purified by flash chromatography (DCM+EtOAc 20% to 60%) to yield 3-chloro-4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (23.0 mg, 0.0378 mmol, 31% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 8.82 (d, J=2.0 Hz, 1H), 8.69 (d, J=1.8 Hz, 1H), 7.96-7.81 (m, 2H), 7.74-7.55 (m, 3H), 7.31 (t, J=8.7 Hz, 1H), 3.19-3.05 (m, 3H), 1.87-1.62 (m, 2H), 0.97 (t, J=7.5 Hz, 3H);13C NMR (101 MHz, DMSO) δ 183.1, 152.7, 151.3, 145.9, 142.5, 140.2, 133.4, 132.9, 131.2, 130.6, 127.3, 125.2, 122.4, 113.3, 54.4, 17.3, 13.1; MS: [M−1]−=567.9. Example 52b: 4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]-2-fluorobenzenesulfonamide Step 1: 4-bromo-2-fluorobenzenesulfonamide (2) To an ice cooled solution of 4-bromo-2-fluorobenzenesulfonyl chloride (1, 0.450 g, 1.55 mmol) in acetonitrile (10.6 mL) was added 25% ammonia solution (0.823 mL, 5.30 mmol) dropwise. The mixture was stirred for 10 min at RT, diluted with water and extracted with EtOAc. The extract was washed with brine, dried over Na2SO4, filtered and the solvent removed under reduced pressure to yield 4-bromo-2-fluorobenzenesulfonamide (0.520 g, 2.05 mmol, 97% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 7.87-7.67 (m, 4H), 7.60 (dd, J=8.6, 1.5 Hz, 1H);13C NMR (50 MHz, DMSO) δ 158 (d, J=258 Hz), 131.1 (d, J=15 Hz), 129.9, 127.9 (d, J=4 Hz), 126.4 (d, J=9 Hz), 120.5 (d, J=25 Hz); MS: [M−1]−=251.8. Step 2: 4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]-2-fluorobenzenesulfonamide (3) A vessel was charged with N-[2,4-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (74.0 mg, 0.125 mmol), 4-bromo-2-fluorobenzenesulfonamide (35.0 mg, 0.138 mmol) and XPhos Pd G3 (1.06 mg, 0.00125 mmol) and purged with argon. Degassed 1,4-dioxane (0.418 mL) and degassed aqueous 1.5M Potassium Carbonate (0.251 mL, 0.376 mmol) were added and the vessel evacuated and backfilled with argon (3×). The mixture was heated to 55° C. for 2 h. The mixture was cooled to RT, diluted with iPrOH (3 mL), conc. HCl was added until the mixture was strongly acidic and stirring was continued at 70° C. overnight. After cooling to RT, solid NaHCO3was added to neutralize the mixture, the solvents were removed and the residue purified by flash chromatography (DCM+EtOAc 20% to 60%) to yield 4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]-2-fluorobenzene-sulfonamide (26.0 mg, 0.0460 mmol, 37% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 15.01 (s, 1H), 9.81 (s, 1H), 9.13 (s, 1H), 8.91 (s, 1H), 8.14-7.53 (m, 6H), 7.43-7.21 (m, 1H), 3.21-3.06 (m, 2H), 1.90-1.63 (m, 2H), 0.97 (t, J=7.5 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 159.6, 157.1, 155.0, 155.0, 154.0, 153.9, 152.5, 149.8, 143.2, 143.2, 142.0, 130.8, 130.7, 130.3, 130.2, 129.9, 129.8, 129.0, 128.6, 123.3, 123.3, 121.8, 121.8, 121.7, 121.7, 116.0, 115.7, 113.3, 112.2, 112.2, 112.0, 53.8, 16.7, 12.5, all peaks reported; MS: [M−1]−=551.9. Example 52c: 4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]-3-methylbenzenesulfonamide Step 1: 4-bromo-3-methylbenzenesulfonamide (2) To an ice cooled solution of 4-bromo-3-methylbenzenesulfonyl chloride (1, 0.690 g, 2.76 mmol) in acetonitrile (1.48 mL) was added 25% ammonia solution (1.15 mL, 7.42 mmol) dropwise. The mixture was stirred for 10 min at RT, diluted with water and extracted with diethyl ether. The extract was washed with brine, dried over Na2SO4, filtered and the solvent removed under reduced pressure to yield 4-bromo-3-methylbenzenesulfonamide (0.690 g, 2.76 mmol, 93% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 7.86-7.72 (m, 2H), 7.56 (ddd, J=8.4, 2.3, 0.5 Hz, 1H), 7.43 (s, 2H), 2.41 (s, 3H);13C NMR (50 MHz, DMSO) δ 143.5, 138.5, 132.8, 127.9, 125.0, 22.6; MS: [M−1]−=247.7. Step 2: 4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]-3-methyl-benzenesulfonamide (3) A vessel was charged with N-[2,4-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (75.0 mg, 0.127 mmol), 4-bromo-3-methylbenzenesulfonamide (34.9 mg, 0.140 mmol) and XPhos Pd G3 (2.69 mg, 0.00318 mmol) and purged with argon. Degassed 1,4-dioxane (0.423 mL) and degassed aqueous 1.5M Potassium Carbonate (0.254 mL, 0.381 mmol) were added and the vessel evacuated and backfilled with argon (3×). The mixture was heated to 55° C. for 1.5 h. The solvent was removed under reduced pressure, the residue taken up THE 2 mL and 1.25M HCl in iPrOH and stirred at 70° C. overnight. The solvent was removed, the residue neutralized with NaHCO3and extracted with EtOAc. The solvent was removed and the residue purified by flash chromatography (DCM+EtOAc 10% to 50% %) and triturated with DCM to yield 4-[3-[2,6-difluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]-3-methylbenzenesulfonamide (41.0 mg, 0.0739 mmol, 58% yield). Analytical Data:1H NMR (400 MHz, DMSO) δ 15.00 (s, 1H), 9.82 (s, 1H), 8.76 (d, J=2.0 Hz, 1H), 8.55 (d, J=1.9 Hz, 1H), 7.84 (s, 1H), 7.78 (dd, J=8.0, 1.3 Hz, 1H), 7.67-7.56 (m, 2H), 7.44 (s, 2H), 7.31 (t, J=8.7 Hz, 1H), 3.16-3.07 (m, 2H), 1.80-1.69 (m, 2H), 0.97 (t, J=7.4 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.5, 152.0, 150.7, 143.7, 141.8, 141.0, 136.6, 132.4, 130.9, 129.9, 127.3, 123.3, 113.0, 53.8, 20.1, 16.8, 12.5; MS: [M−1]−=547.9. Example 53 The compounds of Example 53a-53c were prepared according to the procedure, illustrated in scheme 3. Step 1: 5-bromo-N-methoxy-N-methyl-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carboxamide To a suspensions of 5-bromo-N-methoxy-N-methyl-1H-pyrazolo[3,4-b]pyridine-3-carboxamide (4.43 g, 15.5 mmol) in DCM (62.2 mL) was added dihydropyran (2.84 mL, 31.1 mmol) and p-Toluenesulfonic acid monohydrate (0.591 g, 3.11 mmol) and the mixture was heated to reflux temperature for 30 minutes. After cooling, the mixture was washed with sat. NaHCO3-solution and brine, dried over Na2SO4, filtered and the solvent was removed under reduced pressure. The residue was purified by flash chromatography (hexane+EtOAc 10% to 50%) to yield 5-bromo-N-methoxy-N-methyl-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carboxamide (5.05 g, 13.7 mmol, 88% yield) as oil, which solidified after drying in high vacuum and standing. Analytical Data:1H NMR (200 MHz, CDCl3) δ 8.69 (d, J=2.0 Hz, 1H), 8.58 (d, J=2.2 Hz, 1H), 6.12 (dd, J=10.0, 2.4 Hz, 1H), 4.15-4.01 (m, 1H), 3.93-3.71 (m, 4H), 3.54 (s, 3H), 2.71-2.48 (m, 1H), 2.22-1.65 (m, 5H);13C NMR (50 MHz, CDCl3) δ 150.5, 149.0, 136.5, 134.2, 118.3, 115.1, 83.0, 68.2, 61.9, 29.2, 25.0, 22.8; MS: [M+H]+=390.8. Step 2a: (3-amino-2,6-difluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone To a solution of 2,4-difluoroaniline (0.815 g, 6.32 mmol) and Chlorotrimethylsilane (1.60 mL, 12.6 mmol) in tetrahydrofuran (5.74 mL) cooled to −78° C. was added 2M Lithium diisopropylamide (6.32 mL, 12.6 mmol) in THF/heptane/ethylbenzene dropwise. The mixture was warmed to RT and stirred for 30 minutes. 5-bromo-N-methoxy-N-methyl-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carboxamide (1.06 g, 2.87 mmol) was added and the mixture cooled to −30° C. Lithium diisopropylamide (3.16 mL, 6.32 mmol) in THE/heptane/ethylbenzene was added dropwise and the mixture stirred at −15° C. for 10 minutes. 2N HCl (20 mL) was added and stirring continued at RT for 20 minutes. The mixture was adjusted to pH ˜9 with 2N NaOH and extracted with EtOAc. The extract was washed with brine, dried over Na2SO4, filtered and the solvent was removed. The residue was purified by flash chromatography (hexane+EtOAc, 10% to 40%) to yield (3-amino-2,6-difluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone (0.732 g, 1.67 mmol, 58% yield) as yellow solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 8.86 (d, J=2.2 Hz, 1H), 8.79 (d, J=2.2 Hz, 1H), 7.31-6.69 (m, 2H), 6.14 (dd, J=9.8, 2.0 Hz, 1H), 5.28 (s, 2H), 4.02-3.85 (m, 1H), 3.81-3.62 (m, 1H), 2.42-2.14 (m, 1H), 2.04-1.46 (m, 5H);13C NMR (50 MHz, DMSO) δ 183.7, 151.0, 149.1, 140.3, 132.8, 116.3, 115.5, 82.9, 67.1, 28.4, 24.4, 21.6; MS: [M+Na]+=458.9. Step 2b: (3-amino-2,4,6-trifluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone To a solution of 2,4,6-trifluoroaniline (0.789 g, 5.36 mmol) and Chlorotrimethylsilane (1.36 mL, 10.7 mmol) in tetrahydrofuran (4.88 mL) cooled to −78° C. was added 2M Lithium diisopropylamide (5.36 mL, 10.7 mmol) in THF/heptane/ethylbenzene dropwise. The mixture was warmed to RT and stirred for 30 minutes. 5-bromo-N-methoxy-N-methyl-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carboxamide (0.900 g, 2.44 mmol) was added and the mixture cooled to −30° C. Lithium diisopropylamide (2.68 mL, 5.36 mmol) in THF/heptane/ethylbenzene was added dropwise and the mixture was stirred at −15° C. for 20 minutes. 2N HCl (20 mL) was added and stirring continued at RT for 10 minutes. The mixture pH was adjusted to pH 9 using 2N NaOH and extracted with EtOAc. The extracts were washed with brine, dried over Na2SO4und the solvent was removed. TLC-MS revealed almost no deprotection of TMS-groups. The residue was dissolved in 10 mL THF, 1 mL conc. HCl was added. Deprotection was complete immediately. After dilution with EtOAc solid K2CO3was added, the suspension filtered and the solvent removed. The residue was purified by flash chromatography (hexane+EtOAc 5% to 25%). Yield: 715 mg, 64%. Analytical Data:1H NMR (200 MHz, DMSO) δ 8.85 (d, J=2.1 Hz, 1H), 8.79 (d, J=2.2 Hz, 1H), 7.34-7.16 (m, 1H), 6.14 (d, J=8.4 Hz, 1H), 5.36 (s, 2H), 4.03-3.82 (m, 1H), 3.84-3.58 (m, 1H), 2.44-2.17 (m, 1H), 2.05-1.51 (m, 5H);13C NMR (50 MHz, DMSO) δ 182.7, 151.0, 149.2, 140.2, 132.8, 116.3, 115.5, 82.9, 67.0, 28.4, 24.4, 21.6; MS: [M+Na]+=476.9. Example 53a: 4-[3-[2,6-difluoro-3-(phenylsulfamoylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide Step 1: Preparation of sodium N-phenylsulfamate (2) Aniline (1, 3.95 g, 42.4 mmol) and Triethylamine (59.1 mL, 424 mmol) were dissolved in DCM (106 mL). Chlorosulfuric acid (2.82 mL, 42.4 mmol) was added drop wise at −5° C. and the mixture stirred for 10 minutes. The mixture was concentrated in vacuo and the solid dissolved in 1N Sodium hydroxide (84.8 mL, 84.8 mmol). The mixture was concentrated in vacuo to dryness. The product was suspended in about 500 mL boiling EtOH, filtered while hot and reduced to about 150 mL. After cooling to 7° C. overnight the product was collected by filtration and dried under vacuum to give sodium N-phenylsulfamate (3.94 g, 20.2 mmol, 48% yield) as a white solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 7.90 (s, 1H), 7.15-7.01 (m, 4H), 6.74-6.63 (m, 1H);13C NMR (50 MHz, DMSO) δ 143.7, 128.2, 118.5, 116.3. Step 2: Preparation of phenylsulfamoyl chloride (3) Sodium N-phenylsulfamate (2, 1.12 g, 5.74 mmol) and Phosphorus pentachloride (1.20 g, 5.74 mmol) were heated in toluene (19.1 mL) at 80° C. in an oil bath for 6 h. The reaction was filtered and concentrated in vacuo to yield N-phenylsulfamoyl chloride (0.990 g, 5.17 mmol, 90% yield) as oil which solidifies upon standing. Product was used without further purification and characterization. Step 3: [5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]-[2,6-difluoro-3-(phenylsulfamoylamino) phenyl]methanone (4) To a solution of (3-amino-2,6-difluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone (0.127 g, 0.290 mmol) and Triethylamine (0.0607 mL, 0.436 mmol) in DCM (1.45 mL) was added N-phenylsulfamoyl chloride (0.0724 g, 0.378 mmol) in DCM (1.45 mL) at 0° C. After stirring for 10 minutes at RT, the mixture was diluted with DCM, washed with water and NH4Cl solution, dried over Na2SO4, filtered and the solvent was removed under reduced pressure to yield [5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]-[2,6-difluoro-3-(phenylsulfamoylamino)phenyl]methanone (0.170 g, 0.2870 mmol, 99% yield) which was used without further purification. To the sticky oil was added 2 mL Et2O and the solvent was quickly removed in vacuo to obtain the product as foamed solid. Analytical Data:1H NMR (200 MHz, CDCl3) δ 8.89 (d, J=2.1 Hz, 1H), 8.71 (d, J=2.1 Hz, 1H), 7.73 (td, J=8.9, 5.4 Hz, 1H), 7.47-6.97 (m, 8H), 6.21 (dd, J=9.7, 2.1 Hz, 1H), 3.97 (dd, J=46.6, 10.1 Hz, 2H), 2.66-2.39 (m, 1H), 2.22-1.56 (m, 5H);13C NMR (50 MHz, CDCl3) δ 182.4, 151.0, 149.6, 140.7, 136.0, 133.7, 129.5, 125.8, 121.6, 116.8, 116.6, 83.4, 68.2, 29.0, 24.8, 22.4; MS: [M−1]−=589.8. Step 4: 4-[3-[2,6-difluoro-3-(phenylsulfamoylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (5) A vessel was charged with [5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]-[2,6-difluoro-3-(phenylsulfamoylamino)phenyl]methanone (4, 0.0880 g, 0.149 mmol), (4-sulfamoylphenyl)boronic acid (32.8 mg, 0.163 mmol) and XPhos Pd G3 (3.77 mg, 0.00446 mmol) and purged with argon. Degassed 1,4-dioxane (0.495 mL) and degassed 1.5M aqueous Potassium Carbonate (0.297 mL, 0.446 mmol) was added and the mixture was stirred at 65° C. for 1.5 h. Sat. NH4Cl solution and EtOAc were added and the phases were separated. The organic phase was dried over Na2SO4and evaporated. The residue was taken up in THE (3 mL) and TFA (300 μL) were added at RT. After stirring overnight another 300 μL TFA were added and stirring continued for 2 h (10-12 h). (Still no conversion) The mixture was concentrated and taken up in DCM (3 mL) and sonicated. Another 300 μL TFA were added and stirring continued at RT. After 3 h 3 mL TFA were added at RT and the mixture was stirred overnight and quenched into NaHCO3solution. The aqueous was extracted with EtOAc, the extract was dried over Na2SO4and the solvent was removed. The product was purified by flash chromatography (DCM+MeOH 3% to 13%) to furnish 4-[3-[2,6-difluoro-3-(phenylsulfamoylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (34.0 mg, 0.0547 mmol, 37% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 14.96 (s, 1H), 10.20 (s, 1H), 10.09 (s, 1H), 9.09 (d, J=1.9 Hz, 1H), 8.84 (d, J=1.9 Hz, 1H), 8.14-7.89 (m, 4H), 7.60-7.41 (m, 3H), 7.37-7.15 (m, 5H), 7.02 (t, J=6.9 Hz, 1H); MS: [M−1]−=583.0. Example 53b: 4-[3-[2,6-difluoro-3-(sulfamoylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide Step 1: Preparation of tert-butyl N-chlorosulfonylcarbamate (2) To a solution of chlorosulfonylisocyanate (1, 1.78 mL, 20.4 mmol) in toluene (8.17 mL) was slowly added a solution of 2-methylpropan-2-ol (1.66 g, 22.5 mmol) in toluene (1.16 mL). The mixture was stirred at RT for 1 h. Hexane (23.1 mL) was added and the solution was stirred for 30 minutes. The precipitate formed was filtered and washed with hexane to give tert-butyl N-chlorosulfonylcarbamate (3.33 g, 15.4 mmol, 76% yield) as white solid which was stored under nitrogen at −20° C. Analytical Data:1H NMR (200 MHz, CDCl3) δ 8.86 (s, 1H), 1.56 (s, 9H);13C NMR (50 MHz, CDCl3) δ 148.0, 87.3, 27.9. Step 2: tert-butylN-[[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4-difluorophenyl]sulfamoyl]carbamate (3) To a solution of (3-amino-2,6-difluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone (3, 0.126 g, 0.288 mmol) and Triethylamine (0.0602 mL, 0.432 mmol) in DCM (1.15 mL) was added tert-butyl N-chlorosulfonylcarbamate (0.0808 g, 0.375 mmol) in DCM (1 mL) at 0° C. After stirring for 10 minutes at RT, the mixture was diluted with DCM, washed with water and NH4Cl solution, dried over Na2SO4, filtered and the solvent was removed under reduced pressure to yield tert-butyl N-[[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4-difluorophenyl]sulfamoyl]carbamate (0.176 g, 0.2860 mmol, 99% yield) as yellow foam which was used without further purification. Analytical Data:1H NMR (200 MHz, CDCl3) δ 8.89 (d, J=1.8 Hz, 1H), 8.70 (d, J=1.8 Hz, 1H), 7.74 (td, J=8.9, 5.8 Hz, 1H), 7.06 (t, J=8.4 Hz, 1H), 6.19 (d, J=7.6 Hz, 1H), 4.08 (d, J=11.1 Hz, 1H), 3.91-3.72 (m, 1H), 2.66-2.39 (m, 1H), 2.22-1.90 (m, 2H), 1.88-1.57 (m, 3H), 1.45 (s, 9H);13C NMR (50 MHz, CDCl3) δ 182.4, 151.0, 150.4, 149.7, 140.7, 133.7, 116.8, 116.6, 84.2, 83.5, 77.2, 68.2, 46.0, 27.9, 24.8, 22.4. MS: [M−1]−=613.9. Step 3: 4-[3-[2,6-difluoro-3-(sulfamoylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (4) A vessel was charged with tert-butyl N-[[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4-difluorophenyl]sulfamoyl]carbamate (3, 129 mg, 0.209 mmol), (4-sulfamoylphenyl)boronic acid (46.3 mg, 0.230 mmol) and XPhos Pd G3 (5.31 mg, 0.00628 mmol) and purged with argon. Degassed 1,4-dioxane (0.698 mL) and degassed 1.5M aqueous Potassium Carbonate (0.419 mL, 0.628 mmol) was added and the mixture was stirred at 65° C. for 1.5 h. Sat. NH4Cl solution and EtOAc were added and the phases were separated. The organic phase was dried over Na2SO4and evaporated. The residue was taken up in DCM (3 mL) and TFA (300 μL) was added at RT. After stirring overnight the mixture was quenched into NaHCO3solution and extracted with EtOAc. The extract was dried over Na2SO4and the solvent was removed. The product was purified by flash chromatography (DCM+MeOH 1% to 11%) to furnish 4-[3-[2,6-difluoro-3-(sulfamoylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (37.0 mg, 0.0728 mmol, 35% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 14.96 (s, 1H), 9.32 (s, 1H), 9.10 (d, J=2.0 Hz, 1H), 8.86 (d, J=2.0 Hz, 1H), 8.02 (dd, J=20.7, 8.4 Hz, 4H), 7.68 (td, J=8.9, 5.9 Hz, 1H), 7.47 (s, 2H), 7.36-7.13 (m, 3H); MS: [M−1]−=507.0. Example 53c: 4-[3-[2,4,6-trifluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide Step 1: N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4,6-trifluorophenyl]-N-propylsulfonylpropane-1-sulfonamide (2) To (3-amino-2,4,6-trifluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone (1, 0.289 g, 0.635 mmol) and Triethylamine (0.133 mL, 0.952 mmol) in DCM (2.54 mL) was added 1-Propanesulfonyl chloride (0.0715 mL, 0.635 mmol) slowly at 0° C. After stirring at RT for 15 minutes, the mixture was diluted with DCM, washed with NH4Cl solution, dried over Na2SO4and the solvents were removed. The residue was purified by flash chromatography (hexane+EtOAc, 0 to 25%) to yield N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4,6-trifluorophenyl]-N-propylsulfonylpropane-1-sulfonamide (0.233 g, 0.3490 mmol, 55% yield). The resulting oil was dissolved in DCM, hexane was added, and the solvents were removed. Off white solid. Analytical Data:1H NMR (200 MHz, CDCl3) δ 8.81 (d, J=2.0 Hz, 1H), 8.65 (d, J=2.0 Hz, 1H), 6.97 (td, J=9.1, 1.8 Hz, 1H), 6.15 (dd, J=9.9, 2.2 Hz, 1H), 4.03 (d, J=11.1 Hz, 1H), 3.88-3.48 (m, 5H), 2.53 (dd, J=20.4, 11.9 Hz, 1H), 2.15-1.51 (m, 9H), 1.07 (td, J=7.4, 3.9 Hz, 6H);13C NMR (50 MHz, CDCl3) δ 180.6, 151.1, 149.7, 140.4, 133.5, 116.8, 116.5, 83.2, 68.1, 58.3, 28.7, 24.8, 22.4, 16.7, 12.9; MS: [M+Na]+=688.8. Step 2: N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4,6-trifluorophenyl]propane-1-sulfonamide (3) To a solution of N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4,6-trifluorophenyl]-N-propylsulfonylpropane-1-sulfonamide (2, 0.227 g, 0.340 mmol) in tetrahydrofuran (1.2 mL) and MeOH (0.40 mL) is added 1M aqueous NaOH (1.02 mL, 1.02 mmol). After stirring for 1 h, water (3 mL) was added and the organic solvents evaporated. 2N HCl was added to neutralize the mixture. The solids were collected by suction filtration and dried to yield N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4,6-trifluorophenyl]propane-1-sulfonamide (0.159 g, 0.2830 mmol, 83% yield) as off white solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 9.70 (s, 1H), 8.84 (d, J=10.9 Hz, 2H), 7.62 (t, J=9.3 Hz, 1H), 6.15 (d, J=8.6 Hz, 1H), 4.08-3.59 (m, 2H), 3.25-3.06 (m, 2H), 2.40-2.16 (m, 1H), 2.08-0.76 (m, 10H);13C NMR (50 MHz, DMSO) δ 151.2, 149.2, 139.8, 132.7, 116.6, 115.5, 82.9, 67.1, 54.8, 28.4, 24.4, 21.7, 16.9, 12.6; MS: [M−1]−=559.0. Step 3: 4-[3-[2,4,6-trifluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (4) A vessel was charged with N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,4,6-trifluorophenyl]propane-1-sulfonamide (3, 0.113 g, 0.201 mmol), 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzenesulfonamide (62.7 mg, 0.221 mmol), and XPhos Pd G3 (4.26 mg, 0.00503 mmol) and purged with argon. Degassed 1,4-dioxane (0.671 mL) and degassed 1.5M Potassium Carbonate (0.403 mL, 0.604 mmol) were added and the mixture was heated to 65° C. for 1 h. NH4Cl solution was added and the mixture extracted with EtOAc. The extracts were washed with brine, dried over Na2SO4and the solvent was removed. The residue was taken up in DCM (3 mL) and TFA (300 μL) was added and the mixture stirred at RT overnight. 3 mL TFA were added and stirring continued for 1 h. The reaction was quenched into NaHCO3solution, extracted with EtOAc, dried over Na2SO4and the solvent was removed. The residue was purified by flash chromatography (DCM+MeOH 1% to 11%) to furnish 4-[3-[2,4,6-trifluoro-3-(propylsulfonylamino)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (60.0 mg, 0.1070 mmol, 53% yield). Analytical Data:1H NMR (400 MHz, DMSO) δ 15.02 (s, 1H), 9.66 (s, 1H), 9.10 (d, J=2.1 Hz, 1H), 8.85 (d, J=2.1 Hz, 1H), 8.02 (dd, J=37.6, 8.5 Hz, 4H), 7.59 (t, J=9.4 Hz, 1H), 7.47 (s, 2H), 3.14 (dd, J=8.7, 6.6 Hz, 2H), 1.88-1.73 (m, 2H), 0.99 (t, J=7.4 Hz, 3H);13C NMR (101 MHz, DMSO) δ 181.5, 152.4, 149.9, 143.5, 141.8, 140.3, 131.5, 128.2, 127.9, 126.4, 113.4, 54.7, 16.8, 12.5; MS: [M−1]−=552.0. Example 53d: 4-[3-[3-(ethylsulfamoylamino)-2,6-difluorobenzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (5) Step 1: N-ethylsulfamoyl chloride (2) In analogy to literature procedure: A mixture of ethylamine hydrochloride (2.00 g, 24.5 mmol) and Sulfuryl chloride (7.93 mL, 98.1 mmol) in acetonitrile (4.91 mL) was heated to 75° C. overnight. The mixture was evaporated, treated with diethyl ether and filtered. The solvent was removed to yield N-ethylsulfamoyl chloride (3.28 g, 22.8 mmol, 93% yield), which was used without further characterization. Step 2: [5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]-[3-(ethylsulfamoylamino)-2,6-difluorophenyl]methanone (4) To a solution of (3-amino-2,6-difluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone (90.0 mg, 0.206 mmol) and Triethylamine (0.0373 mL, 0.268 mmol) in DCM (0.823 mL) was added N-ethylsulfamoyl chloride (35.5 mg, 0.247 mmol) in DCM (0.823 mL) at 0° C. After stirring for 10 minutes at RT, the mixture was diluted with DCM, washed with water and NH4Cl solution, dried over Na2SO4, filtered and the solvent was removed under reduced pressure to yield [5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]-[3-(ethylsulfamoylamino)-2,6-difluorophenyl]methanone (4, 0.110 g, 0,202 mmol, 98% yield) as yellow foam, which was used without further purification. Analytical Data: 1H NMR (200 MHz, CDCl3) δ 8.85 (s, 1H), 8.67 (s, 1H), 7.69 (td, J=8.9, 5.6 Hz, 1H), 7.01 (t, J=8.9 Hz, 1H), 6.73 (s, 1H), 6.15 (d, J=7.5 Hz, 1H), 4.75 (t, J=5.9 Hz, 1H), 4.13-3.97 (m, 1H), 3.90-3.67 (m, 1H), 3.24-3.04 (m, 2H), 2.63-2.31 (m, 1H), 2.21-1.51 (m, 5H), 1.16 (t, J=7.2 Hz, 3H). 13C NMR (50 MHz, CDCl3) δ 182.6, 156.8 (dd, J=252.0, 6.7 Hz), 151.5 (dd, J=250.7, 7.5 Hz), 151.1, 149.7, 140.8, 133.8, 125.8 (dd, J=9.8, 2.0 Hz), 122.0 (dd, J=12.7, 3.5 Hz), 116.8, 116.6, 112.29 (dd, J=22.2, 3.8 Hz), 83.4, 68.3, 38.6, 29.0, 24.8, 22.5, 15.1. Step 3: 4-[3-[3-(ethylsulfamoylamino)-2,6-difluorobenzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (5) A vessel was charged with [5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]-[3-(ethylsulfamoylamino)-2,6-difluorophenyl]methanone (4, 94.0 mg, 0.173 mmol), (4-sulfamoylphenyl)boronic acid (38.2 mg, 0.190 mmol) and XPhos Pd G3 (4.38 mg, 0.00518 mmol) and purged with argon. Degassed 1,4-dioxane (0.576 mL) and degassed 1.5 M aqueous Potassium Carbonate (0.345 mL, 0.518 mmol) was added and the mixture was stirred at 65° C. for 1.5 h. Sat. NH4Cl solution and EtOAc were added and the phases were separated. The organic phase was dried over Na2SO4and evaporated. The residue was taken up in DCM (3 mL) and TFA (300 μL) was added at RT. After 4 h the mixture was quenched into NaHCO3solution and extracted with EtOAc. The extract was dried over Na2SO4and the solvent was removed. The product was purified by flash chromatography (DCM+MeOH 1% to 11%) to furnish 4-[3-[3-(ethylsulfamoylamino)-2,6-difluorobenzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (5, 35.0 mg, 0.0652 mmol, 38% yield). Analytical Data:1H NMR (400 MHz, DMSO) δ 14.97 (s, 1H), 9.54 (s, 1H), 9.10 (d, J=2.0 Hz, 1H), 8.86 (d, J=1.9 Hz, 1H), 8.02 (dd, J=38.5, 8.3 Hz, 4H), 7.64 (td, J=9.0, 5.9 Hz, 1H), 7.51-7.41 (m, 3H), 7.30 (t, J=8.7 Hz, 1H), 3.04-2.88 (m, 2H), 1.04 (t, J=7.2 Hz, 3H);13C NMR (101 MHz, DMSO) δ 182.8, 162.8, 152.4, 149.8, 143.5, 142.0, 140.3, 131.4, 128.2, 127.9, 126.4, 113.4, 37.2, 14.6; MS: [M−1]−=535.0. Example 54: Synthesis of N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]methanesulfonamide (6) Step 1: N-(2,6-difluorophenyl)acetamide 2,6-difluoroaniline (1, 3.34 g, 25.9 mmol) and Acetic anhydride (2.56 mL, 27.2 mmol) were combined in DCM (34.5 mL) and stirred overnight at RT. The reaction mixture was diluted with DCM (50 mL), washed with water and saturated sodium bicarbonate, dried over Na2SO4and evaporated to give N-(2,6-difluorophenyl)acetamide (2, 3.88 g, 22.7 mmol, 88% yield) as white solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 9.69 (s, 1H), 7.43-7.23 (m, 1H), 7.13 (t, J=7.9 Hz, 2H), 2.08 (s, 3H);13C NMR (50 MHz, DMSO) δ 168.5, 160.4, 160.3, 155.5, 155.4, 128.0, 127.8, 127.6, 115.1, 114.7, 114.4, 112.1, 112.0, 111.9, 111.8, 111.6, 111.6, 22.4, all peaks reported; MS: [M+H]+=171.9. Step 2: 3-bromo-2,6-difluoroaniline (3) N-(2,6-difluorophenyl)acetamide (2, 8.77 g, 51.2 mmol) was dissolved in Sulfuric acid (41.0 mL, 769 mmol) and N-Bromosuccinimide (9.12 g, 51.2 mmol) was added portion wise at rt. The reaction was stirred overnight at RT and quenched slowly into ice water (400 mL) with stirring. The solids were collected by suction filtration and washed with water, hexane, hexane+EtOAc (8+2) and hexane again. The solids were taken up in ethanol (30.0 mL) and conc. HCl (30 mL) and heated to reflux for 3 h. The mixture was poured into ice (400 g), neutralized with solid NaOH and the solids were collected by suction filtration, washed with water and dried in vacuo to yield 3-bromo-2,6-difluoroaniline (3, 7.83 g, 37.6 mmol, 73% yield). Analytical Data:1H NMR (200 MHz, CDCl3) δ 6.95-6.59 (m, 2H), 3.84 (s, 2H);13C NMR (50 MHz, CDCl3) δ 152.2 (dd, J=133.8, 6.5 Hz), 147.5 (dd, J=132.7, 6.9 Hz), 125.4 (t, J=16.9 Hz), 122, 119.6 (d, J=8.4 Hz), 111.7 (dd, J=19.9, 3.1 Hz), 103.8 (dd, J=19.2, 3.8 Hz). Step 3: (3-amino-2,4-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (5) To 3-bromo-2,6-difluoroaniline (3, 12.9 g, 61.9 mmol) in tetrahydrofuran (76.9 mL) was added 2M Isopropylmagnesium chloride in THE (30.9 mL, 61.9 mmol) dropwise at 0° C. and stirred for 15 minutes at RT. After cooling to 0° C., chlorotrimethylsilane (7.85 mL, 61.9 mmol) was added, the mixture warmed to 25° C. and stirred for 20 minutes. The suspension was cooled to 0° C. and 2M Isopropylmagnesium chloride in THE (30.9 mL, 61.9 mmol) was added dropwise and stirred for 15 minutes at RT. After cooling to 0° C. chlorotrimethylsilane (7.85 mL, 61.9 mmol) was added stirring continued for 20 minutes at 25° C. The suspension was cooled to 0° C. again and 2M isopropylmagnesium chloride in THE (30.9 mL, 61.9 mmol) was added dropwise and the mixture was stirred for 10 minutes at 0° C. (Solution A). Meanwhile 2M isopropylmagnesium chloride in THE (13.5 mL, 26.9 mmol) was added dropwise to a suspension of 5-bromo-N-methoxy-N-methyl-1H-pyrazolo[3,4-b]pyridine-3-carboxamide (4, 7.67 g, 26.9 mmol) in tetrahydrofuran (76.9 mL) at 0° C. The resulting suspension was stirred for 5 minutes and transferred to solution A. The reaction was stirred overnight at RT, conc. HCl (26.9 mL, 323 mmol) was added and stirred for 10 minutes. Water was added until the phases became clear. The mixture was neutralized with 2N NaOH, saturated with NaCl and extracted with THE. The extracts were washed with brine, dried over Na2SO4and filtered. After evaporation of the solvent, the solids were stirred in 100 mL DCM collected by suction filtration and dried to yield 6.35 g a first crop as light yellow solid. The filtrate was evaporated, triturated with diethyl ether and DCM to obtain a second crop (0.41 g). Total yield (3-amino-2,4-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (5, 6.76 g, 19.1 mmol, 71% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 14.55 (s, 1H), 8.65 (dd, J=5.2, 2.1 Hz, 2H), 7.13-6.86 (m, 2H), 5.44 (s, 2H);13C NMR (50 MHz, DMSO) δ 186.1, 153.5 (dd, J=188, 9 Hz), 150.8, 150.3, 148.6 (dd, J=191, 9 Hz), 141.1, 132.5, 126.1 (t, J=17 Hz), 122.8 (dd, J=12, 3 Hz), 116.2 (dd, J=9, 3 Hz), 115.4, 115, 110.6 (dd, J=19, 3 Hz); MS: [M−1]−=351.2. Step 4: N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]methane-sulfonamide (6) To a suspension of (3-amino-2,4-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (5, 0.620 g, 1.76 mmol) in pyridine (2.83 mL, 35.1 mmol) was added methanesulfonyl chloride (0.544 mL, 7.02 mmol) at RT 50° C. for 2 h. The reaction was concentrated and taken up in 2 M Sodium hydroxide solution (13.2 mL, 26.3 mmol) and stirred for 15 minutes. The solution was poured into chilled 3N HCl 20 mL, extracted with EtOAc and the extracts were washed with 2N HCl, dried over Na2SO4and evaporated. The product was purified by flash chromatography (DCM+EtOAc 0% to 40%) to yield N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]methane-sulfonamide (0.390 g, 0.9040 mmol, 52% yield) as red solid. Analytical Data:1H NMR (400 MHz, DMSO) δ 14.87 (s, 1H), 9.75 (s, 1H), 8.71 (d, J=13.9 Hz, 2H), 7.85 (d, J=6.6 Hz, 1H), 7.39 (t, J=8.5 Hz, 1H), 3.12 (s, 3H);13C NMR (101 MHz, DMSO) δ 184.7, 160.8 (dd, J=255, 3 Hz), 157.06 (dd, J=257.4, 4.2 Hz), 150.8, 150.5, 140.9, 132.4, 130.6 (dd, J=10, 3 Hz), 123.3 (dd, J=13, 3 Hz), 115.4, 115.1, 114.3 (t, J=17 Hz), 111.9 (dd, J=21, 3 Hz), 41.5; MS: [M−1]−=429.1. Example 54a: N-[2,6-difluoro-3-[5-[4-(1H-tetrazol-5-yl)phenyl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide A microwave vessel was charged with N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]methanesulfonamide (60.0 mg, 0.139 mmol), XPhos Pd G3 (3.53 mg, 0.00417 mmol) and [4-(1H-tetrazol-5-yl)phenyl]boronic acid (31.7 mg, 0.167 mmol) and purged with argon. Degassed 1,4-dioxane (0.464 mL) and degassed aqueous 1.5 M Potassium Carbonate (0.325 mL, 0.487 mmol) were added and the mixture was heated to 110° C. under microwave irradiation for 60 minutes. After cooling, the mixture was diluted with EtOAc and neutralized with sat. NH4Cl solution. The organic phase was concentrated under reduced pressure and the product isolated by flash chromatography (DCM+MeOH (+1% formic acid) 5% to 15%), triturated with MeOH and dried at 100° C. in a vacuum oven to yield N-[2,6-difluoro-3-[5-[4-(1H-tetrazol-5-yl)phenyl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (21.0 mg, 0.0423 mmol, 30% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 14.85 (s, 1H), 9.75 (s, 1H), 9.10 (d, J=2.1 Hz, 1H), 8.86 (d, J=2.2 Hz, 1H), 8.25-8.00 (m, 4H), 7.97-7.81 (m, 1H), 7.41 (td, J=8.9, 1.3 Hz, 1H), 3.12 (s, 3H); MS: [M−1]−=495.2. Example 54b: 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzamide A microwave vessel was charged with N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]methanesulfonamide (60.0 mg, 0.139 mmol), XPhos Pd G3 (5.89 mg, 0.00696 mmol) and (4-Carbamoylphenyl)boronic acid (27.5 mg, 0.167 mmol) and purged with argon. Degassed 1,4-dioxane (0.464 mL) and degassed aqueous 1.5 M Potassium Carbonate (0.325 mL, 0.487 mmol) were added and the mixture was heated to 110° C. under microwave irradiation for 60 minutes. After cooling, the mixture was diluted with EtOAc and neutralized with sat. NH4Cl solution. The solvents were removed under reduced pressure and the product isolated by flash chromatography (DCM+MeOH, 5% to 15%) and dried at 100° C. in a vacuum oven to yield 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzamide (40.0 mg, 0.0789 mmol, 57% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 9.06 (d, J=1.9 Hz, 1H), 8.82 (d, J=2.0 Hz, 1H), 8.14-7.80 (m, 6H), 7.49-7.33 (m, 2H), 3.11 (s, 3H); MS: [M−1]−=470.3 Example 54c: 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide A microwave vessel was charged with N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]methanesulfonamide (60.0 mg, 0.139 mmol), XPhos Pd G3 (5.89 mg, 0.00696 mmol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzenesulfonamide (47.3 mg, 0.167 mmol) and purged with argon. Degassed 1,4-dioxane (0.464 mL) and degassed aqueous 1.5 M potassium carbonate (0.325 mL, 0.487 mmol) were added and the mixture was heated to 110° C. under microwave irradiation for 60 minutes. After cooling, the mixture was diluted with EtOAc and neutralized with sat. NH4Cl solution. The solvents were removed under reduced pressure and the product isolated by flash chromatography (DCM+EtOAc, 50% to 100%) and dried at 100° C. in a vacuum oven to yield 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzenesulfonamide (31.0 mg, 0.0574 mmol, 41% yield).1H NMR (200 MHz, DMSO) δ 9.07 (d, J=2.1 Hz, 1H), 8.84 (d, J=2.1 Hz, 1H), 8.12-7.78 (m, 4H), 7.50-7.32 (m, 2H), 3.11 (s, 3H); [M−1]−=506.2. Example 55: Synthesis of 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoic acid (4) and Ethyl 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoate (5) Step 1: N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]methanesulfonamide (2) To a suspension of N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]methanesulfonamide (380 mg, 0.881 mmol) and p-toluenesulfonic acid monohydrate (33.5 mg, 0.176 mmol) in DCM (2.94 mL) was added dihydropyran (0.161 mL, 1.76 mmol) and the reaction was heated to reflux temperature for 1 h. The reaction was diluted with DCM, washed with sat. NaHCO3solution, dried over Na2SO4and concentrated. The residue was triturated with n-hexane to yield N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]methane-sulfonamide (370 mg, 0.7180 mmol, 81% yield) as beige solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 9.78 (s, 1H), 8.80 (dd, J=13.4, 1.6 Hz, 2H), 7.88 (dd, J=14.6, 7.8 Hz, 1H), 7.43 (t, J=9.0 Hz, 1H), 6.14 (d, J=8.6 Hz, 1H), 3.94 (d, J=11.6 Hz, 1H), 3.72 (dd, J=14.6, 9.1 Hz, 1H), 3.13 (s, 3H), 2.46-2.19 (m, 1H), 2.00-1.18 (m, 5H);13C NMR (50 MHz, DMSO) δ 184.4, 150.8, 149.0, 139.9, 133.0, 116.1, 116.0, 82.7, 67.2, 41.5, 28.5, 24.5, 21.8; MS: [M−1]−=513.2. Step 2: 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoic acid (3) N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]methanesulfonamide (355 mg, 0.689 mmol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzoic acid (188 mg, 0.758 mmol) d XPhos Pd G3 (17.5 mg, 0.0207 mmol) were combined in degassed 1,4-dioxane (2.30 mL) and 1.5 M potassium carbonate (2.07 mL, 3.10 mmol). The reaction was evacuated and flushed with argon (3×). XPhos Pd G3 (17.5 mg, 0.0207 mmol) was added and the reaction was stirred at 60° C. oil bath temperature for 3 h. After cooling the mixture was acidified with 2N HCl and extracted with EtOAc. The extract was washed with brine, dried over Na2SO4and concentrated. The residue was purified by flash chromatography (DCM+MeOH 3% to 25%) and triturated with n-hexane to yield 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoic acid (276 mg, 0.4960 mmol, 72% yield). Analytical Data:1H NMR (400 MHz, DMSO) δ 9.77 (s, 1H), 9.10 (d, J=2.1 Hz, 1H), 8.83 (d, J=2.1 Hz, 1H), 8.09 (d, J=8.3 Hz, 2H), 7.96 (d, J=8.3 Hz, 2H), 7.90 (dd, J=14.8, 7.6 Hz, 1H), 7.44 (t, J=8.8 Hz, 1H), 6.21 (dd, J=9.9, 2.0 Hz, 1H), 4.01-3.91 (m, 1H), 3.80-3.68 (m, 1H), 3.59 (t, J=6.6 Hz, 2H), 3.13 (s, 3H), 2.46-2.33 (m, 1H), 2.06-1.93 (m, 2H), 1.84-1.68 (m, 2H); MS: [M−1]−=555.4. Step 3: 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoic acid (4) 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoic acid (203 mg, 0.365 mmol) was suspended in 3N HCl and heated to 70° C. with stirring overnight. The reaction was concentrated, the solids collected by suction filtration, washed with water and dried at 100° C. to yield 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoic acid (133 mg, 0.2820 mmol, 77% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 9.06 (d, J=2.1 Hz, 1H), 8.83 (d, J=2.1 Hz, 1H), 8.10 (d, J=8.3 Hz, 2H), 8.00-7.80 (m, 3H), 7.40 (td, J=9.0, 1.3 Hz, 1H), 3.12 (s, 3H); MS: [M−1]−=471.2. Step 4: Ethyl 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoate (5) 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoic acid (50.0 mg, 0.0898 mmol) was stirred in 1.25 M HCl in ethanol (0.719 mL, 0.898 mmol) at 75° C. in a sealed vessel. After 3 h 200 μL H2SO4were added and stirring continued at 75° C. overnight. The reaction was quenched into NaHCO3solution, the solids were collected by centrifugation and washed with water and diethyl ether and dried in vacuo to yield ethyl 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoate (22.0 mg, 0.0440 mmol, 49% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 14.86 (s, 1H), 9.74 (s, 1H), 9.06 (d, J=2.1 Hz, 1H), 8.83 (d, J=2.1 Hz, 1H), 8.18-7.80 (m, 5H), 7.41 (t, J=8.9 Hz, 1H), 4.36 (q, J=7.0 Hz, 2H), 3.12 (s, 3H), 1.35 (t, J=7.0 Hz, 3H); MS: [M−1]−=499.4. Example 56: Synthesis of [(2S)-2-amino-3-methoxy-3-oxopropyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoate hydrochloride Step 1: [(2S)-3-methoxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxopropyl] 4-bromobenzoate 4-Bromobenzoyl chloride (1.13 g, 5.15 mmol) was dissolved in 25.7 ml THE and cooled to 0° C. Triethylamine (0.861 mL, 6.18 mmol) was added and to the resulting suspension was added methyl (2S)-3-hydroxy-2-[(2-methylpropan-2yl)oxycarbonylamino]-propanoate (1.35 g, 6.18 mmol) and 4-DMAP (31.5-mg, 0.257 mmol). The mixture was stirred at RT for 60 minutes, then diluted with EtOAc and washed with water, sat. NaHCO3and sat. NH4Cl solution. After drying over Na2SO4, the solvent was removed in vacuo. The residue was dissolved in a small amount of MeOH and added to water with stirring. The solids were collected by suction filtration and dried to yield [(2S)-3-methoxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxopropyl] 4-bromobenzoate (1.70 g, 4.23 mmol, 82% yield). Analytical Data:1H NMR (200 MHz, CDCl3) δ 7.89-7.78 (m, 2H), 7.61-7.50 (m, 2H), 5.38 (d, J=8.0 Hz, 1H), 4.70 (dd, J=8.2, 4.0 Hz, 1H), 4.59 (d, J=4.0 Hz, 2H), 3.76 (d, J=4.3 Hz, 3H), 1.42 (d, J=4.2 Hz, 9H). 13C NMR (50 MHz, CDCl3) δ 170.4, 165.4, 155.2, 132.0, 131.3, 128.7, 128.5, 80.6, 65.3, 53.1, 52.9, 28.4. MS (ESI+): m/z 424.04 [M+1]+. Step 2: N-(2,6-difluoro-3-(1-(tetrahydro-2H-pyran-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl)methanesulfonamide A vessel was charged with N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]methanesulfonamide (0.630 g, 1.22 mmol), bis(pinacolato)diboron (341 mg, 1.34 mmol), anhydrous potassium acetate (360 mg, 3.67 mmol) and dry 1,4-dioxane (4.08 mL). The vessel was evacuated and filled with argon (3×). 1,1′-bis(diphenylphosphino) ferrocene-dichloropalladium (1:1) (17.9 mg, 0.0245 mmol) was added and the reaction was stirred at 80° C. overnight. After cooling, EtOAc was added, the suspension stirred for 30 minutes and filtered over celite. The solvent was concentrated, n-heptane was added and the solids were collected by suction filtration, washed with hexane and dried to furnish N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]-pyridine-3-carbonyl]phenyl]methanesulfonamide (0.690 g, 1.23 mmol, quant.). Analytical Data:1H NMR (200 MHz, DMSO) δ 8.94-8.71 (m, 2H), 7.19-6.88 (m, 2H), 6.18 (d, J=9.1 Hz, 1H), 4.05-3.61 (m, 2H), 2.64 (s, 3H), 2.44-1.10 (m, 18H). Step 3: [(2S)-3-methoxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxopropyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoate A vessel was charged with N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (101 mg, 0.180 mmol), [(2S)-3-methoxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxopropyl] 4-bromobenzoate (79.5 mg, 0.198 mmol), potassium fluoride (31.3 mg, 0.539 mmol), Pd(dppf)Cl2.DCM (7.33 mg, 0.00898 mmol) and degassed 1,4-dioxane/water (4+1) (0.6 mL) followed by evacuation and filling with argon (3×). The reaction mixture was heated to 65° C. overnight with stirring. The mixture was diluted with EtOAc, washed with brine and the solvents were removed. The product was isolated via flash chromatography (DCM/EtOAc gradient, from 0% to 20% EtOAc) to yield [(2S)-3-methoxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxopropyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoate (68.0 mg, 0.0897 mmol, 50% yield) as white solid. Analytical Data:1H NMR (400 MHz, acetone) δ 9.00 (d, J=2.0 Hz, 1H), 8.85 (d, J=2.1 Hz, 1H), 8.47 (s, 1H), 8.17 (d, J=8.3 Hz, 2H), 7.97-7.89 (m, 3H), 7.33 (t, J=8.5 Hz, 1H), 6.65 (d, J=7.6 Hz, 1H), 6.26 (dd, J=9.9, 2.3 Hz, 1H), 4.79-4.58 (m, 3H), 4.02 (d, J=11.4 Hz, 1H), 3.85-3.69 (m, 4H), 3.21 (s, 3H), 2.60-2.46 (m, 1H), 2.17-2.06 (m, 1H), 2.04-1.96 (m, 1H), 1.92-1.78 (m, 1H), 1.74-1.57 (m, 2H), 1.42 (s, 9H). 13C NMR (101 MHz, acetone) δ 185.7, 171.1, 166.2, 162.2 (dd, J=256, 3 Hz), 158.7 (dd, J=258, 5 Hz), 156.4, 151.9, 150.3, 143.3, 142.2, 133.6, 131.6 (dd, J=11, 4 Hz), 131.3, 130.3, 130.2, 128.6, 124.7 (dd, J=14, 4 Hz), 116.2, 115.8 (t, J=17 Hz), 112.7 (dd, J=21, 4 Hz), 83.9, 79.8, 68.3, 65.4, 54.0, 52.8, 42.1, 29.8, 29.8, 28.6, 25.7, 23.1. TLC-MS (ESI−): m/z 756.5 [M−1]−. Step 4: [(2S)-2-amino-3-methoxy-3-oxopropyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoate hydrochloride [(2S)-3-methoxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxopropyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoate (63.0 mg, 0.0831 mmol) was stirred in trifluoroacetic acid (1 mL) at RT for 3 h. The mixture was heated to 50° C. for 40 min. After cooling on ice, 4N HCl (1.04 mL, 4.16 mmol) in 1,4-dioxane was added to the solution, stirring continued for 5 minutes and diethyl ether (3 mL) was added. The product was collected by suction filtration, washed with diethyl ether and dried in vacuo to yield [(2S)-2-amino-3-methoxy-3-oxopropyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoate hydrochloride (44.0 mg, 0.0721 mmol, 87% yield) as off white solid. Analytical Data:1H NMR (400 MHz, DMSO) δ 14.92 (s, 1H), 9.76 (s, 1H), 9.10 (d, J=2.0 Hz, 1H), 8.97 (s, 2H), 8.86 (d, J=2.1 Hz, 1H), 8.20 (d, J=8.5 Hz, 2H), 8.03 (d, J=8.3 Hz, 2H), 7.88 (dd, J=14.7, 7.6 Hz, 1H), 7.41 (t, J=8.7 Hz, 1H), 4.73 (d, J=3.3 Hz, 2H), 4.67 (s, 1H), 3.81 (s, 3H), 3.12 (s, 3H). 13C NMR (101 MHz, DMSO) δ 185.1, 167.4, 164.8, 152.2, 149.5, 142.3, 141.8, 131.1, 130.5, 128.5, 128.0, 127.5, 114.0, 62.2, 53.1, 51.3, 41.5, 40.1. TLC-MS (ESI−): m/z 572.4 [M−1]. Example 57: Synthesis of (2S)-2-amino-3-[4-[3-[2,4-difluoro-3-(methanesulfonamido)-benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoyl]oxypropanoic acid hydrochloride Step 1: benzyl (2S)-3-hydroxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]propanoate A solution of di-tert-butyl dicarbonate (7.06 mL, 30.7 mmol) in 1,4-dioxane (14.5 mL) was added to a solution of (2S)-2-amino-3-hydroxypropanoic acid (2.69 g, 25.6 mmol) and potassium carbonate (3.54 g, 25.6 mmol) in water (14.5 mL). The solution was stirred for 16 h at RT. 1,4-Dioxane was evaporated and the aqueous solution was washed with 3× diethyl ether (10 mL). Water was evaporated in vacuo and remaining traces were azeotropically removed with EtOH. The resulting white powder was suspended in DMF (28.9 mL) and benzyl bromide (3.44 mL, 28.9 mmol) was added. The mixture was stirred at RT for 16 h. DMF was evaporated in vacuo and the residue was stirred with toluene (28.9 mL) and filtered. The phase was washed twice with water and brine and dried over Na2SO4. After filtration, the solvent was evaporated at 90° C. in vacuo. The oily benzyl (2S)-3-hydroxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]propanoate (4.77 g, 16.2 mmol, 63% yield) was used in the next step without further purification. Analytical Data:1H NMR (200 MHz, CDCl3) δ 7.34 (s, 5H), 5.56 (s, 1H), 5.20 (s, 2H), 4.40 (s, 1H), 3.92 (qd, J=11.2, 3.7 Hz, 2H), 2.46 (s, 1H), 1.43 (s, 9H). Step 2: [(2S)-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxo-3-phenylmethoxypropyl] 4-bromobenzoate To a solution of 4-bromobenzoyl chloride (0.500 g, 2.28 mmol) in THE (11.4 mL) was added triethylamine (0.381 mL, 2.73 mmol) and 4-DMAP (0.0139 g, 0.114 mmol). To the resulting suspension was added benzyl (2S)-3-hydroxy-2-[(2-methylpropan-2-yl)oxycarbonylamino]propanoate (0.807 g, 2.73 mmol) in 2 mL THE and the reaction was stirred at RT for 60 minutes. The reaction was diluted with EtOAc, washed with water, sat. NaHCO3and sat. NH4Cl solution. After drying over Na2SO4, the solvent was removed in vacuo. The product was purified by flash chromatography (n-hexane/EtOAc gradient, from 0% to 20% EtOAc) to yield [(2S)-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxo-3-phenylmethoxypropyl] 4-bromobenzoate (0.501 g, 1.05 mmol, 46% yield) as white solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 7.84 (d, J=8.5 Hz, 2H), 7.75-7.64 (m, 3H), 7.38-7.23 (m, 5H), 5.26-5.03 (m, 2H), 4.55 (dd, J=16.6, 7.0 Hz, 3H), 1.38 (s, J=14.3 Hz, 9H). 13C NMR (50 MHz, DMSO) δ 169.6, 164.7, 155.4, 135.7, 131.7, 131.3, 128.5, 128.3, 128.0, 127.7, 127.5, 78.6, 66.3, 52.7, 28.1. MS: [M+Na]+=500.3. Step 3: [(2S)-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxo-3-phenylmethoxypropyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoate A vessel was charged with [(2S)-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxo-3-phenylmethoxypropyl] 4-bromobenzoate (124 mg, 0.260 mmol), N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (146 mg, 0.260 mmol), Pd(dppf)Cl2.DCM (10.6 mg, 0.0130 mmol), potassium fluoride (45.2 mg, 0.779 mmol) and degassed 1,4-dioxane/water (4+1) (0.8 mL) and the vessel was evacuated and filled with argon (3×). The reaction was heated to 50° C. with stirring for 6 h. The mixture was diluted with EtOAc, washed with brine and the solvents were removed. The product was isolated via flash chromatography (3 times column volume hexane/EtOAc (80/20 v/v, followed by DCM/EtOAc 80/20 v/v to elute the product) to yield [(2S)-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxo-3-phenylmethoxy-propyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]-pyridin-5-yl]benzoate (147 mg, 0.1760 mmol, 68% yield) as white solid. Analytical Data:1H NMR (200 MHz, Acetone) δ 9.06 (s, 1H), 8.90 (s, 1H), 8.55 (s, 1H), 8.22-7.89 (m, 5H), 7.59-7.31 (m, 5H), 6.82 (d, J=7.6 Hz, 1H), 6.34 (d, J=9.2 Hz, 1H), 5.44-5.23 (m, 2H), 4.99-4.72 (m, 3H), 4.20-3.76 (m, 2H), 3.30 (s, 3H), 2.62 (dd, J=20.1, 9.7 Hz, 1H), 2.25-1.61 (m, 5H), 1.60-1.32 (m, 10H). 13C NMR (50 MHz, Acetone) δ 185.7, 170.6, 166.1, 156.4, 151.7, 150.3, 143.1, 142.1, 136.8, 133.5, 131.3, 130.1, 129.3, 129.0, 128.9, 128.4, 116.1, 83.8, 79.8, 68.3, 67.6, 65.4, 54.2, 42.0, 29.7, 28.5, 25.7, 23.1. TLC-MS (ESI−): m/z 833.0 [M−1]−. Step 4: (2S)-2-amino-3-[4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoyl]oxypropanoic acid hydrochloride [(2S)-2-[(2-methylpropan-2-yl)oxycarbonylamino]-3-oxo-3-phenylmethoxypropyl] 4-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]benzoate (142 mg, 0.170 mmol) was dissolved in EtOAc (1.70 mL) and 14 mg palladium on activated carbon (10%) was added. The reaction was stirred under H2atmosphere (3 bar) overnight. The reaction was degassed, 5 mg palladium on activated carbon (10%) was added and the reaction stirred under 5 bar H2atmosphere (for 6 h). The reaction was filtered through celite and the solvent was removed. The residue was taken up in 2 ml TFA and stirred at RT for 2 h. 4N HCl in dioxane (2 mL) was added with ice cooling. After 5 minutes, 5 ml THE was added and the solids collected by suction filtration, washed with THE and dried in vacuo to yield (2S)-2-amino-3-[4-[3-[2,4-difluoro-3-(methanesulfon-amido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]benzoyl]oxypropanoic acid hydrochloride (50.0 mg, 0.0814 mmol, 48% yield) as off white solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 14.92 (s, 1H), 9.76 (s, 1H), 9.10 (d, J=2.1 Hz, 1H), 8.86 (d, J=2.1 Hz, 1H), 8.78 (s, 3H), 8.22 (d, J=8.5 Hz, 2H), 8.03 (d, J=8.5 Hz, 2H), 7.88 (dd, J=15.0, 7.6 Hz, 1H), 7.41 (td, J=8.9, 1.2 Hz, 1H), 4.71 (s, 2H), 4.53 (s, 1H), 3.12 (s, 3H). 13C NMR (50 MHz, DMSO) δ 185.2, 168.4, 164.9, 152.3, 149.6, 142.4, 141.9, 131.1, 130.5, 128.6, 128.2, 127.6, 114.1, 66.3, 51.4, 41.5. TLC-MS (ESI−): m/z 558.2 [M−1]−. Example 58: Synthesis of 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylic acid hydrochloride Step 1: Methyl 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylate A vessel was charged with N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (132 mg, 0.235 mmol), methyl 5-bromopyridine-2-carboxylate (55.8 mg, 0.258 mmol), potassium fluoride (40.9 mg, 0.704 mmol), Pd(dppf)Cl2.DCM (9.58 mg, 0.0117 mmol) and degassed 1,4-dioxane/water (4+1) (0.6 mL) and the vessel was evacuated and filled with argon (3×). The reaction was heated to 50° C. overnight with stirring. The mixture was diluted with EtOAc, washed with brine and the solvents were removed. The product was isolated via flash chromatography (DCM/EtOAc gradient, from 20% to 60% EtOAc) to yield methyl 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylate (97.0 mg, 0.1700 mmol, 72% yield) as white solid. Analytical Data:1H NMR (400 MHz, acetone) δ 9.14 (d, J=1.9 Hz, 1H), 9.06 (d, J=2.1 Hz, 1H), 8.91 (d, J=2.1 Hz, 1H), 8.40 (dd, J=8.1, 2.3 Hz, 1H), 8.22 (d, J=8.1 Hz, 1H), 7.95 (dd, J=14.9, 7.5 Hz, 1H), 7.34 (dd, J=8.8, 8.0 Hz, 1H), 6.27 (dd, J=9.9, 2.4 Hz, 1H), 4.09-3.91 (m, 4H), 3.80 (td, J=11.2, 3.5 Hz, 1H), 3.21 (s, 3H), 2.54 (ddd, J=16.4, 13.3, 4.0 Hz, 1H), 2.19-2.06 (m, 1H), 2.05-1.97 (m, 1H), 1.94-1.77 (m, 1H), 1.76-1.56 (m, 2H). 13C NMR (101 MHz, acetone) δ 185.7, 166.1, 162.3 (dd, J=256, 3 Hz), 158.7 (dd, J=258, 5 Hz), 152.0, 150.3, 149.3, 148.5, 142.3, 137.3, 136.7, 131.63 (dd, J=10.9, 3.2 Hz), 130.7, 125.9, 124.7 (dd, J=14, 4 Hz), 116.2, 115.8 (t, J=17 Hz), 112.8 (dd, J=21, 4 Hz), 83.9, 68.4, 52.8, 42.1, 29.8, 25.8, 23.1. TLC-MS (ESI−): m/z 570.4 [M−1]−. Step 2: methyl 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylate To a solution of methyl 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylate (60.0 mg, 0.105 mmol) in MeOH (0.525 mL) was added methanesulfonic acid (0.0273 mL, 0.420 mmol) and the mixture was stirred at 65° C. for 1.5 h. The mixture was cooled to RT and added slowly into 15 ml diethyl ether, the solid was collected by suction filtration, washed with diethyl ether and dried in vacuo to yield methyl 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylate (36.0 mg, 0.0739 mmol, 70% yield) as white solid. Analytical Data:1H NMR (200 MHz, CDCl3) δ 14.90 (s, 1H), 9.74 (s, 1H), 9.16 (dd, J=12.4, 1.9 Hz, 2H), 8.93 (d, J=2.0 Hz, 1H), 8.47 (dd, J=8.2, 2.3 Hz, 1H), 8.19 (d, J=8.3 Hz, 1H), 7.88 (dd, J=14.8, 7.7 Hz, 1H), 7.41 (t, J=8.7 Hz, 1H), 3.93 (s, 3H), 3.12 (s, 3H). 13C NMR (50 MHz, CDCl3) δ 185.1, 165.0, 152.4, 149.6, 148.3, 146.5, 141.9, 136.1, 136.0, 129.2, 128.4, 125.0, 114.0, 52.5, 41.5. MS: [M+Na]+=510.4. Step 3: 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylic acid hydrochloride Methyl 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylate (46.0 mg, 0.0805 mmol) was stirred in 3N HCl (2.68 mL, 8.05 mmol) at 70° C. overnight in an open vessel. The solvent was removed, residual water was azeotropically removed with toluene. The residue was dissolved in MeOH (1 mL) and added dropwise to diethyl ether. The solid was collected by suction filtration, washed with diethyl ether and dried in vacuo to yield 5-[3-[2,4-difluoro-3-(methanesulfonamido)benzoyl]-1H-pyrazolo[3,4-b]pyridin-5-yl]pyridine-2-carboxylic acid hydrochloride (33.0 mg, 0.0647 mmol, 80% yield). Analytical Data:1H NMR (200 MHz, CDCl3) δ 14.96 (s, 1H), 9.77 (s, 1H), 9.18 (s, 1H), 9.13 (d, J=1.9 Hz, 1H), 8.93 (d, J=1.9 Hz, 1H), 8.48 (d, J=8.1 Hz, 1H), 8.19 (d, J=7.8 Hz, 1H), 7.88 (dd, J=14.7, 7.8 Hz, 1H), 7.41 (t, J=8.6 Hz, 1H), 3.12 (s, 3H). 13C NMR (101 MHz, DMSO) δ 185.0, 152.3, 149.5, 141.9, 136.1, 114.2, 114.0, 48.5, 41.5, 15.1. TLC-MS (ESI−): m/z 472.3 [M−1]−. Example 59: Synthesis of N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]methanesulfonamide hydrochloride Step 1: N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-pyridin-4-ylpyrazolo[3,4-b]pyridine-3-carbonyl]-phenyl]methanesulfonamide A vessel was charged with N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (86.0 mg, 0.153 mmol), 4-bromopyridine hydrochloride (29.7 mg, 0.153 mmol), Pd(dppf)Cl2.DCM (3.75 mg, 0.00459 mmol), degassed 1,4-dioxane (0.382 mL) and degassed 1.5 M aqueous potassium carbonate (0.408 mL, 0.612 mmol). The vessel was evacuated and filled with argon (3×) and heated to 60° C. for 2 h. After cooling, the reaction was diluted with EtOAc neutralized with NH4Cl solution, the aqueous phase was discarded, and the organics were concentrated. The product was isolated by flash chromatography (DCM/MeOH gradient, from 2% to 12% MeOH). N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-pyridin-4-ylpyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (63.0 mg, 0.1230 mmol, 80% yield). Analytical Data:1H NMR (200 MHz, acetone) δ 9.03 (d, J=2.3 Hz, 1H), 8.89 (d, J=2.1 Hz, 1H), 8.72 (d, J=5.9 Hz, 3H), 7.94 (ddd, J=8.8, 7.5, 6.2 Hz, 1H), 7.80 (dd, J=4.5, 1.6 Hz, 2H), 7.34 (td, J=9.0, 1.7 Hz, 1H), 6.25 (dd, J=9.9, 2.3 Hz, 1H), 4.12-3.95 (m, 1H), 3.90-3.68 (m, 1H), 3.21 (s, 3H), 2.51 (dt, J=10.1, 7.1 Hz, 1H), 2.16-1.57 (m, 5H). 13C NMR (50 MHz, acetone) δ 185.7, 152.1, 151.4, 150.1, 145.8, 142.2, 131.9, 131.6 (dd, J=10, 4 Hz), 130.3, 124.6 (dd, J=13, 4 Hz), 122.8, 116.1, 115.8, 112.8 (dd, J=21, 4 Hz), 83.8, 68.3, 42.0, 29.7, 25.7, 25.3, 23.1. TLC-MS (ESI−): m/z 512.6 [M−1]−. Step 2: N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]-methanesulfonamide hydrochloride N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-pyridin-4-ylpyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (60.0 mg, 0.117 mmol) was heated to reflux in MeOH (2 mL) and 4N HCl in dioxane (0.5 mL) for 1 h. The solvent was removed and the residue triturated with THF to yield N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]methanesulfonamide hydrochloride (37.0 mg, 0.0794 mmol, 68% yield) as white solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 15.09 (s, 1H), 9.77 (s, 1H), 9.28 (d, J=2.3 Hz, 1H), 9.13 (d, J=2.3 Hz, 1H), 8.99 (d, J=6.7 Hz, 2H), 8.52 (d, J=6.7 Hz, 2H), 7.90 (dd, J=15.0, 7.6 Hz, 1H), 7.42 (td, J=8.9, 1.2 Hz, 1H), 3.12 (s, 3H). 13C NMR (101 MHz, DMSO) δ 184.9, 160.7 (dd, J=255, 3 Hz), 157.1 (dd, J=257, 4 Hz), 153.0, 151.8, 149.7, 143.6, 142.3, 130.5, 127.3, 124.1, 123.4 (dd, J=13, 4 Hz), 114.3 (t, J=17 Hz), 112.0 (dd, J=21.3 Hz), 41.5. TLC-MS (ESI−): m/z 428.6 [M−1]−. Example 60: Synthesis of N-[2,6-difluoro-3-[5-[4-(methylsulfonimidoyl)phenyl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide Step 1: (4-bromophenyl)-imino-methyl-oxo-λ{circumflex over ( )}{6}-sulfane 1-bromo-4-methylsulfanylbenzene (0.970 g, 4.78 mmol), ammonium acetate (1.47 g, 19.1 mmol) and (diacetoxyiodo)benzene (3.85 g, 11.9 mmol) were combined in 9.55 mL MeOH at RT. The reaction was stirred for 1 h, diluted with water (20 mL) and extracted with EtOAc. The extracts were washed with water, dried over Na2SO4and the solvents were removed in vacuo. The product was isolated by flash chromatography (DCM/MeOH, 97/3% v/v) (4-bromophenyl)-imino-methyl-oxo-λ{circumflex over ( )}{6}-sulfane (0.570 g, 2.43 mmol, 51% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 7.91-7.76 (m, 4H), 4.24 (s, 1H), 3.08 (s, 1H). 13C NMR (50 MHz, DMSO) δ 143.5, 132.0, 129.4, 126.4, 45.6. MS: [M+H]+=234.1. Step 2: N-[2,6-difluoro-3-[5-[4-(methylsulfonimidoyl)phenyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide A vessel was charged with N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (93.0 mg, 0.165 mmol), (4-bromophenyl)-imino-methyl-oxo-λ{circumflex over ( )}{6}-sulfane (38.7 mg, 0.165 mmol), Pd(dppf)Cl2.DCM (4.05 mg, 0.00496 mmol), potassium fluoride (28.8 mg, 0.496 mmol) and degassed 1,4-dioxane/water (4:1, 0.5 mL). The vessel was evacuated and filled with argon (3×) and heated to 50° C. for 3 h. After cooling the reaction was diluted with EtOAc, washed with brine and the solvents were removed. The product was isolated by flash chromatography (DCM/MeOH gradient, from 1% to 8% MeOH). N-[2,6-difluoro-3-[5-[4-(methylsulfonimidoyl)phenyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]-methanesulfonamide (63.0 mg, 0.1070 mmol, 65% yield). Analytical Data:1H NMR (200 MHz, acetone) δ 8.97 (d, J=1.9 Hz, 1H), 8.82 (d, J=2.0 Hz, 1H), 8.63 (s, 1H), 8.13 (d, J=8.3 Hz, 2H), 8.03-7.87 (m, 3H), 7.33 (td, J=8.9, 1.3 Hz, 1H), 6.24 (dd, J=9.7, 1.9 Hz, 1H), 4.11-3.93 (m, 1H), 3.87-3.70 (m, 1H), 3.18 (d, J=13.0 Hz, 7H), 2.66-2.37 (m, 1H), 2.18-1.52 (m, 5H). 13C NMR (50 MHz, acetone) δ 151.8, 150.4, 142.7, 142.1, 130.3, 129.4, 129.0, 116.1, 83.8, 68.4, 46.6, 42.0, 35.2, 32.3, 29.8, 25.7, 23.1, 14.3. TLC-MS (ESI−): m/z 588.4 [M−1]−. Step 3: N-[2,6-difluoro-3-[5-[4-(methylsulfonimidoyl)phenyl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide To N-[2,6-difluoro-3-[5-[4-(methylsulfonimidoyl)phenyl]-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (63.0 mg, 0.107 mmol) in 2 mL MeOH was added 4N HCl in 1,4-dioxane (2 mL) and the reaction was stirred at reflux temperature for 1 h. After cooling, the mixture was concentrated, treated with dry THE/diethyl ether (1+1) and the solids were collected by suction filtration, washed with diethyl ether. The solids were taken up in water and neutralized with NaHCO3solution. The product was collected by suction filtration, washed with water and dried in vacuo to yield N-[2,6-difluoro-3-[5-[4-(methylsulfonimidoyl)phenyl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]methanesulfonamide (24.0 mg, 0.0451 mmol, 42% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 9.08 (s, 1H), 8.85 (s, 1H), 8.07 (s, 4H), 7.87 (dd, J=14.8, 8.0 Hz, 1H), 7.41 (t, J=8.5 Hz, 1H), 4.31 (s, 1H), 3.13 (d, J=3.4 Hz, 6H). 13C NMR (101 MHz, DMSO) δ 185.1, 152.3, 149.5, 143.5, 141.8, 141.1, 131.0, 128.6, 128.1, 127.9, 114.0, 45.8, 41.5. MS: [M+H]+=506.4. Example 61:_Synthesis of N-[3-[5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]propane-1-sulfonamide Step 1: N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]propane-1-sulfonamide To a suspension of (3-amino-2,4-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (805 mg, 2.28 mmol) and triethylamine (3.50 mL, 25.1 mmol) in DCM (11.4 mL) was added 1-propanesulfonyl chloride (898 μL, 7.98 mmol) in DCM (11.4 mL) slowly at −10° C. and the reaction was stirred at −10° C. for 15 minutes. The reaction was diluted with DCM, washed with 2N HCl and brine, dried over Na2SO4and evaporated. The residue was dissolved in THE (2 mL) and 2N KOH (2 mL) was added. After 10 minutes the reaction was diluted with water and THE was evaporated. The solution was added to 2N HCl (10 mL) with stirring, stirring was continued for 30 minutes and the solids were collected by suction filtration, washed with 2N HCl and water and dried at 100° C. to yield N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]propane-1-sulfonamide (801 mg, 1.69 mmol, 74% yield). Analytical Data:1H NMR (200 MHz, Acetone) δ 13.77 (s, 1H), 8.72 (d, J=12.1 Hz, 2H), 8.38 (s, 1H), 7.91 (dd, J=14.8, 7.6 Hz, 1H), 7.30 (t, J=8.7 Hz, 1H), 3.40-3.14 (m, 2H), 2.03-1.81 (m, 2H), 1.07 (t, J=7.4 Hz, 3H). 13C NMR (50 MHz, Acetone) δ 185.7, 162.1 (dd, J=255, 4 Hz), 153.7 (dd, J=222, 4 Hz), 152.0, 151.6, 142.5, 133.7, 131.5 (dd, J=11, 4 Hz), 124.5 (dd, J=14, 4 Hz), 116.6, 116.2, 115.7 (t, J=17 Hz), 112.8, 112.7, 112.4, 112.3, 56.2, 18.1, 13.1. MS: [M−1]−=457.3. Step 2: N-[3-[5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]-propane-1-sulfonamide A microwave vessel was charged with N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]propane-1-sulfonamide (60.0 mg, 0.131 mmol), (4-chlorophenyl)boronic acid (21.4 mg, 0.137 mmol) and Pd(dppf)Cl2.DCM (5.33 mg, 0.00653 mmol) and purged with argon. Degassed 1,4-dioxane (0.435 mL) and degassed aqueous 1.5M K2CO3(0.261 mL, 0.392 mmol) were added and the mixture stirred at 110° C. under microwave irradiation for 30 minutes. A spatula Pd(dppf)Cl2was added and heating continued for 30 minutes. After cooling, the mixture was neutralized with sat. NH4Cl solution and diluted with EtOAc. The aqueous phase was discarded, and the solvent removed under reduced pressure. The residue was purified by flash chromatography (DCM/EtOAc gradient, from 0 to 35% EtOAc) and triturated with DCM to yield N-[3-[5-(4-chlorophenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]propane-1-sulfonamide (30.0 mg, 0.0605 mmol, 46% yield) as white solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 14.81 (s, 1H), 9.66 (s, 1H), 9.01 (d, J=2.1 Hz, 1H), 8.77 (d, J=2.1 Hz, 1H), 7.98-7.78 (m, 3H), 7.60 (d, J=8.4 Hz, 2H), 7.40 (t, J=8.7 Hz, 1H), 3.22-3.09 (m, 2H), 1.92-1.70 (m, 2H), 1.00 (t, J=7.5 Hz, 3H). 13C NMR (101 MHz, DMSO) δ 185.2, 152.1, 149.4, 141.8, 136.2, 133.1, 131.3, 129.2, 128.1, 114.1, 55.0, 39.5, 16.9, 12.7. MS: [M−1]−=489.5. Examples 62-67 In analogy to the procedure of Example 61, step 2 the following compounds were prepared: ProductExpl.ReactantChemical structure/nameAnalytical data621H NMR (200 MHz, DMSO) δ 14.85 (s, 1H), 13.07 (s, 1H), 9.67 (s, 1H), 9.07 (d, J = 2.1 Hz, 1H), 8.84 (d, J = 2.0 Hz, 1H), 8.10 (d, J = 8.3 Hz, 2H), 8.01- 7.79 (m, 3H), 7.40 (td, J = 9.0, 1.2 Hz, 1H), 3.24-3.01 (m, 2H), 1.94-1.68 (m, 2H), 1.00 (t, J = 7.4 Hz, 3H).13C NMR (101 MHz, DMSO) δ 185.2, 167.1, 160.8 (dd, J = 255, 4 Hz), 157.1 (dd, J = 256, 4 Hz), 152.3, 149.5, 141.9, 141.5, 131.4, 130.5 (dd, J = 11, 4 Hz), 130.2, 128.5, 127.5, 123.6 (dd, J = 14, 4 Hz), 114.3 (d, J = 17 Hz), 114.1, 112.0 (dd, J = 22, 4 Hz), 54.9, 16.9, 12.6. TLC-MS (ESI−): m/z 499.7 [M − 1]−.4-(3-(2,4-difluoro-3-(propylsulfon-amido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)benzoic acid631H NMR (200 MHz, DMSO) δ 14.90 (s, 1H), 9.67 (s, 1H), 9.13 (s, 1H), 8.92 (s, 1H), 8.73 (s, 2H), 7.92 (d, J = 4.5 Hz, 3H), 7.40 (t, J = 8.7 Hz, 1H), 3.24-3.05 (m, 2H), 1.93-1.70 (m, 2H), 1.00 (t, J = 7.3 Hz, 3H).13C NMR (101 MHz, DMSO) δ 185.1, 160.8 (dd, J = 255, 3 Hz), 157.1 (dd, J = 257, 4 Hz), 152.7, 150.2, 149.4, 144.7, 142.0, 130.5 (dd, J = 10, 4 Hz), 129.5, 128.9, 123.6 (dd, J = 13, 3 Hz), 121.9, 114.3 (t, J = 17 Hz), 114.0, 112.0 (dd, J = 22, 3 Hz), 54.9, 16.9, 12.6. TLC-MS (ESI−): m/z 456.3 [M − 1]−N-(2,6-difluoro-3-(5-(pyridin-4-yl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl)propane-1-sulfonamide64TLC-MS (ESI−): m/z = 469.0, 489.0 [M − 1]−N-(3-(5-(2-chlorophenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl)propane-1-sulfonamide65TLC-MS (ESI−): m/z = 477.2, 497.2 [M − 1]−N-(2,6-difluoro-3-(5-(4-isopropyl-phenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl)propane-1-sulfonamide66TLC-MS (ESI−): m/z = 467.2, 487.1 [M − 1]−N-(2,6-difluoro-3-(5-(4-fluoro-2-methylphenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl)propane-1-sulfonamide67TLC-MS (ESI−): m/z = 514.1, 534.1 [M − 1]−4-(3-(2,4-difluoro-3-(propylsulfon-amido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)benzenesulfonamide Example 68: Synthesis of N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]-1-phenylmethanesulfonamide Step 1: N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]-1-phenylmethanesulfonamide To a solution of (3-amino-2,4-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methanone (830 mg, 2.35 mmol) and 4-DMAP (0.574 g, 4.70 mmol) in pyridine (4.74 mL, 58.8 mmol) was added phenylmethanesulfonyl chloride (583 mg, 3.06 mmol) at −10° C. and the mixture was stirred at −10° C. for 10 minutes until a homogeneous suspension was formed. Stirring was continued at RT for 10 minutes and the reaction was heated to 50° C. for 30 minutes. The reaction was concentrated under reduced pressure and taken up in 2N NaOH (3.53 mL, 7.05 mmol) and was stirred at RT for 10 minutes. The mixture was diluted with water and slowly added to 25 mL 2N HCl with stirring. After 10 minutes, the solids were collected by suction filtration, washed with water and dried at 100° C. to yield N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]-1-phenylmethanesulfonamide (1.02 g, 2.01 mmol, 86% yield) as off-white solid. Analytical Data:1H NMR (200 MHz, DMSO) δ 14.98 (s, 1H), 9.81 (s, 1H), 8.73 (dd, J=6.1, 2.2 Hz, 2H), 7.88 (dd, J=14.9, 7.6 Hz, 1H), 7.50-7.30 (m, 6H), 4.53 (s, 2H). 13C NMR (50 MHz, DMSO) δ 184.8, 150.9, 150.6, 140.9, 132.5, 131.0, 129.4, 128.4, 128.3, 115.5, 115.2, 59.4. MS: [M−1]−=505.2. Step 2: N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]-1-phenylmethanesulfonamide A microwave vessel was charged with N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]-1-phenylmethanesulfonamide (75.0 mg, 0.148 mmol), 4-pyridinylboronic acid (21.8 mg, 0.177 mmol) and Pd(dppf)Cl2.DCM (6.04 mg, 0.00739 mmol) and purged with argon. Degassed 1,4-dioxane (0.493 mL) and degassed aqueous 1.5M K2CO3(0.345 mL, 0.517 mmol) were added and the mixture was heated to 110° C. under microwave irradiation for 60 minutes. After cooling, the reaction was diluted with EtOAc and neutralized with NH4Cl solution. The aqueous phase was cut and the solvent evaporated. The residue was purified by flash chromatography (DCM+MeOH 2% to 10%) and triturated with DCM to yield N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]-1-phenylmethanesulfonamide (40.0 mg, 0.0783 mmol, 53% yield). Analytical Data:1H NMR (400 MHz, DMSO) δ 14.91 (s, 1H), 9.77 (s, 1H), 9.13 (d, J=2.3 Hz, 1H), 8.92 (d, J=2.3 Hz, 1H), 8.72 (dd, J=4.7, 1.4 Hz, 2H), 8.07-7.68 (m, 3H), 7.53-7.26 (m, 6H), 4.53 (s, 2H). [M−1]−=504.4. Example 69: Synthesis of N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]ethanesulfonamide Step 1: (3-amino-2,4-difluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone To 3-bromo-2,6-difluoroaniline (4.69 g, 22.6 mmol) in tetrahydrofuran (19.6 mL) was added 2M isopropylmagnesium chloride in THE (11.3 mL, 22.6 mmol) dropwise at 0° C. and stirred for 15 minutes without further cooling. After cooling again to 0° C., chlorotrimethylsilane (2.86 mL, 22.6 mmol) was added, the mixture warmed to 25° C. and stirred for 20 minutes. The solution was cooled to 0° C. and chlorotrimethylsilane (2.86 mL, 22.6 mmol) was added stirring continued for 20 minutes at 25° C. The solution was cooled to 0° C. again and 2M isopropylmagnesium chloride in THE (11.3 mL, 22.6 mmol) was added dropwise and the mixture was stirred for 10 minutes at 0° C. 5-bromo-N-methoxy-N-methyl-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carboxamide (3.62 g, 9.80 mmol) was dissolved in 10 mL THE and added to the mixture and stirring was continued for 1 h at RT. The reaction was quenched with sat. NH4Cl solution, water was added until the aqueous phase became clear and the phases were separated. The aqueous was extracted with EtOAc, the extracts were washed with brine, dried over Na2SO4and the solvent was removed. The oily residue was dissolved in 25 mL THE and 2 mL conc. HCl were added with stirring. After 5 minutes, the mixture was carefully neutralized with solid K2CO3, diluted with EtOAc and filtered. The solvents were removed and the residue purified by flash chromatography (n-hexane/EtOAc gradient, from 0% to 40% EtOAc) to yield (3-amino-2,4-difluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone (2.35 g, 5.37 mmol, 55% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 8.76 (dd, J=13.9, 2.1 Hz, 2H), 7.15-6.89 (m, 2H), 6.12 (d, J=7.8 Hz, 1H), 5.51 (s, 2H), 4.08-3.56 (m, 2H), 2.46-2.20 (m, 1H), 2.06-1.46 (m, 5H). 13C NMR (50 MHz, DMSO) δ 185.6, 153.6 (dd, J=190, 9 Hz), 150.6, 149.0, 146.8 (d, J=9 Hz), 140.2, 133.0, 126.3 (t, J=17 Hz), 122.3 (dd, J=11, 3 Hz), 116.4 (d, J=3 Hz), 116.2, 116.2, 115.7, 110.7 (dd, J=19, 3 Hz), 82.7, 67.0, 28.5, 24.4, 21.8. MS: [M+Na+MeOH]+=491.05. Step 2: N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]-ethanesulfonamide To a solution of (3-amino-2,4-difluorophenyl)-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridin-3-yl]methanone (286 mg, 0.654 mmol) and triethylamine (0.547 mL, 3.92 mmol) in DCM (3.27 mL) was added ethanesulfonyl chloride (143 μL, 1.50 mmol) at 0° C. The reaction was stirred at RT for 15 minutes and was washed with 2N HCl and brine. The solvent was removed and taken up in 2 mL THE and 2 mL 2N aqueous KOH and stirred for 10 minutes. The reaction was acidified with 2N HCl, diluted with water and extracted with EtOAc. The extract was washed with brine, dried over Na2SO4and evaporated. The residue was purified by flash chromatography (n-hexane/EtOAc gradient, from 5% to 40% EtOAc) to yield N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]ethanesulfonamide (0.267 g, 0.5040 mmol, 77% yield). Analytical Data:1H NMR (200 MHz, DMSO) δ 9.70 (s, 1H), 8.80 (dd, J=14.1, 2.1 Hz, 2H), 7.87 (dd, J=14.8, 7.7 Hz, 1H), 7.43 (td, J=8.9, 1.0 Hz, 1H), 6.14 (dd, J=9.6, 1.7 Hz, 1H), 4.07-3.87 (m, 1H), 3.83-3.62 (m, 1H), 3.20 (q, J=7.2 Hz, 2H), 2.34 (dd, J=22.5, 11.3 Hz, 1H), 2.07-1.46 (m, 5H), 1.33 (t, J=7.3 Hz, 3H). 13C NMR (50 MHz, DMSO) δ 184.4, 161.8 (dd, J=191, 4 Hz), 156.7 (dd, J=193, 4 Hz), 150.8, 149.0, 139.9, 133.0, 130.8 (dd, J=11, 4 Hz), 122.9 (dd, J=13, 4 Hz), 116.1, 116.0, 114.4 (t, J=17 Hz), 112.2 (dd, J=21, 4 Hz), 82.7, 67.1, 47.7, 28.5, 24.5, 21.8, 8.0. MS: [M−1]−=527.1. Step 3: N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]ethanesulfonamide A microwave vessel was charged with N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]ethanesulfonamide (76.0 mg, 0.144 mmol), 4-pyridinylboronic acid (19.4 mg, 0.158 mmol) and tri(tertbutyl)phosphine Pd G3 (6.08 mg, 0.00718 mmol) and was purged with argon. Degassed 1,4-dioxane (0.479 mL) and degassed 1.5M aqueous potassium carbonate (0.287 mL, 0.431 mmol) were added and the reaction was stirred at 70° C. for 45 minutes under microwave irradiation. The reaction was acidified with 2 mL 6N HCl diluted with 3 mL MeOH and heated to 70° C. under microwave irradiation for 60 minutes. The mixture was diluted with water, neutralized with 2N NaOH and extracted with THE. The extract was washed with brine, dried over Na2SO4and the solvent was removed. The residue was purified by flash chromatography (DCM/MeOH gradient, from 2% to 10% MeOH) to yield N-[2,6-difluoro-3-(5-pyridin-4-yl-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl]ethane-sulfonamide (42.0 mg, 0.0947 mmol, 66% yield). Analytical Data:1H NMR (400 MHz, DMSO) δ 14.89 (s, 1H), 9.66 (s, 1H), 9.12 (d, J=2.1 Hz, 1H), 8.91 (d, J=2.3 Hz, 1H), 8.72 (d, J=5.9 Hz, 2H), 7.91-7.83 (m, 3H), 7.40 (t, J=8.8 Hz, 1H), 3.18 (q, J=7.3 Hz, 2H), 1.33 (t, J=7.3 Hz, 3H). 13C NMR (101 MHz, DMSO) δ 185.0, 152.6, 150.3, 149.3, 144.4, 141.9, 129.5, 128.7, 121.8, 114.0, 47.7, 7.9. MS: [M−1]−=442.1. Example 70: Synthesis of 4-(3-(2,4-difluoro-3-(propysulfonamido)benzoyl)-1H-pyrazolo[3,4-b]pyridin-5-yl)-3-methylbenzenesulfonamide Step 1: N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]propane-1-sulfonamide To a suspension of N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]propane-1-sulfonamide (778 mg, 1.64 mmol) in DCM (8.47 mL) was added p-toluenesulfonic acid monohydrate (31.3 mg, 0.164 mmol) and dihydropyran (0.165 mL, 1.81 mmol) and the reaction was warmed to 40° C. for 1 h. The reaction was washed with NaHCO3solution and the solvent was removed. The residue was purified by flash chromatography (n-hexane+EtOAc, 5% to 45%) to yield N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]propane-1-sulfonamide (693 mg, 1.28 mmol, 78% yield). Analytical Data:1H NMR (200 MHz, acetone) δ 8.73 (dt, J=8.0, 1.8 Hz, 2H), 8.39 (s, 1H), 7.92 (ddd, J=8.8, 7.4, 6.2 Hz, 1H), 7.32 (td, J=8.9, 1.7 Hz, 1H), 6.18 (dd, J=9.8, 2.4 Hz, 1H), 4.03-3.67 (m, 2H), 3.33-3.20 (m, 2H), 2.61-2.38 (m, 1H), 2.01-1.54 (m, 5H), 1.07 (t, J=7.5 Hz, 3H). 13C NMR (50 MHz, acetone) δ 361.8, 339.4 (dd, J=178, 4 Hz), 334.3 (dd, J=181, 4 Hz), 327.9, 326.8, 317.5, 310.4, 308.0 (dd, J=11, 4 Hz), 300.7 (dd, J=13, 4 Hz), 293.8, 293.2, 292.2 (t, J=17 Hz), 289.0 (dd, J=22, 4 Hz), 260.2, 244.7, 232.6, 206.3, 206.0, 202.0, 199.4, 194.5, 189.5. TLC-MS (ESI−): m/z 541.0/543.0 [M−H]−. Step 2: N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide A vessel was charged with N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]propane-1-sulfonamide (875 mg, 1.61 mmol), bis(pinacolato)diboron (429 mg, 1.69 mmol), bis(triphenylphosphine)palladium(II) dichloride (22.6 mg, 0.0322 mmol), anhydrous potassium acetate (474 mg, 4.83 mmol) and dry 1,4-dioxane (8.05 mL). The vessel was evacuated and filled with argon (3×) and the reaction was heated to 80° C. for 3 h. After cooling, the reaction was diluted with EtOAc, filtered over celite and evaporated. The residue was taken up in EtOAc (ca. 20 mL) activated charcoal was added and the mixture was heated to reflux for 10 minutes. After cooling, the mixture was filtered over celite, the solvent was evaporated and the residue was sonicated with n-hexane. The solids were collected by suction filtration and in vacuo to yield N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (765 mg, 1.3 mmol, 80% yield) as colorless solid. Step 3 A vessel was charged with N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (100 mg, 0.169 mmol), potassium fluoride (29.5 mg, 0.508 mmol), 4-bromo-3-methylbenzenesulfonamide (46.6 mg, 0.186 mmol), Pd(dppf)Cl2.DCM (6.92 mg, 0.00847 mmol) and 0.5 mL 1,4-dioxane/water (4+1). The vessel was evacuated and filled with argon (3×) and heated to 60° C. for 2 h. The reaction was acidified with conc. HCl (0.3 mL), diluted with MeOH (0.2 mL) and heated to 60° C. overnight. After cooling, the reaction was diluted with EtOAc and water. The organic phase was evaporated and the product was purified by flash chromatography (DCM/EtOAc gradient, from 10% to 50% EtOAc) 4-[3-[2,4-difluoro-3-(propylsulfonyl-amino)benzoyl]-1H-pyrazolo[3,4-b]pyridine-5-yl]-3-methylbenzenesulfonamide (42.0 mg, 0.0764 mmol, 45% yield) Analytical Data: TLC-MS (ESI−): m/z=528.2, 548.1 [M−H]− Example 71: Synthesis of N-(2,6-difluoro-3-(5-(2-(trifluoromethyl)pyrimidin-5-yl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl)propane-1-sulfonamide A vessel was charged with N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (88.0 mg, 0.149 mmol), 5-bromo-2-(trifluoromethyl)pyrimidine (37.2 mg, 0.164 mmol), potassium fluoride (26.0 mg, 0.447 mmol), bis(triphenylphosphine)palladium(II) dichloride (5.23 mg, 0.00745 mmol) and 0.5 mL 1,4-dioxane/water (4+1). The vessel was evacuated and filled with argon (3×) and heated to 60° C. for 2 h. The reaction was acidified with conc. HCl (0.3 mL), diluted with MeOH (0.2 mL) and heated to 60° C. overnight. After cooling, the reaction was diluted with EtOAc and water. The organic phase was evaporated and the product was purified by flash chromatography (DCM/EtOAc gradient, from 5% to 35% EtOAc) N-[2,6-difluoro-3-[5-[2-(trifluoromethyl)pyrimidin-5-yl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (42.0 mg, 0.0798 mmol, 54% yield). Analytical Data: TLC-MS (ESI−): m/z=505.1, 525.1 [M−H]− Example 72: Synthesis of N-(3-(5-(4-chloro-2-methylphenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl)propane-1-sulfonamide A vessel was charged with N-[2,6-difluoro-3-[1-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (80.0 mg, 0.135 mmol), 1-bromo-4-chloro-2-methylbenzene (18.0 μL, 0.135 mmol), potassium fluoride (23.6 mg, 0.406 mmol), bis(triphenylphosphine)palladium(II) dichloride (4.76 mg, 0.00677 mmol) and 0.5 mL 1,4-dioxane/water (4+1). The vessel was evacuated and filled with argon (3×) and heated to 60° C. for 1 h. The reaction was acidified with conc. HCl (0.2 mL), diluted with MeOH (0.2 mL) and heated to 60° C. overnight. After cooling, the reaction was diluted with EtOAc and water. The organic phase was evaporated and the product was purified by flash chromatography (DCM/EtOAc gradient, 0%-40% EtOAc) to yield N-[3-[5-(4-chloro-2-methylphenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]propane-1-sulfonamide (32.0 mg, 0.0608 mmol, 45% yield). Analytical Data: TLC-MS (ESI−): m/z=483.3, 503.3 [M−H]− Example 73: Synthesis of N-(3-(5-(4-(1H-tetrazol-5-yl)phenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl)ethanesulfonamide Step 1: N-(3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl) ethanesulfonamide To a suspension of (3-amino-2,4-difluorophenyl)-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-yl)methanone (823 mg, 2.33 mmol) and triethylamine (3.57 mL, 25.6 mmol) in DCM (11.7 mL) was added ethanesulfonyl chloride (0.773 mL, 8.16 mmol) in DCM (11.7 mL) slowly at −10° C. and the reaction was stirred at −10° C. for 20 minutes. The reaction was washed with 2N HCl and brine and the solvent was removed. The residual gum was taken up in 10 mL THE and 2N NaOH (6.99 mL, 14.0 mmol) was added. After stirring for 30 minutes the solution acidified with 2N HCl. Water was added, THE was removed in vacuo and the solids were collected by suction filtration, washed with water and dried at 100° C. to yield N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]ethanesulfonamide (761 mg, 1.71 mmol, 73% yield). Step 2: N-(3-(5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl)ethanesulfonamide To a suspension of N-[3-(5-bromo-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl]ethanesulfonamide (529 mg, 1.19 mmol) and p-toluenesulfonic acid monohydrate (22.6 mg, 0.119 mmol) in DCM (5.94 mL) was added dihydropyran (0.130 mL, 1.43 mmol) and the reaction was warmed to 30° C. for 1 h. The reaction was diluted with EtOAc, washed with sat. NaHCO3solution and brine, and the solvent was removed. The product was purified by flash chromatography (n-hexane/EtOAc gradient, 0%-40% EtOAc) to yield N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]ethanesulfonamide (534 mg, 1.01 mmol, 85% yield). Step 3 Analytical Data: TLC-MS (ESI−): m/z=509.3 [M−H]− Example 74: Synthesis of N-(3-(5-(4-(1H-tetrazol-5-yl)phenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl)propane-1-sulfonamide A vessel was charged with N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]propane-1-sulfonamide (80.0 mg, 0.147 mmol), [4-(1H-tetrazol-5-yl)phenyl]boronic acid (33.6 mg, 0.177 mmol) and Pd(dppf)Cl2.DCM (6.01 mg, 0.00736 mmol) and purged with argon. Degassed 1,4-dioxane (0.491 mL) and degassed 1.5 M aqueous potassium carbonate (0.393 mL, 0.589 mmol) were added, the vessel was sealed and the reaction was heated to 80° C. for 2.5. The reaction was diluted with MeOH, acidified with conc. HCl and stirring was continued at 60° C. for 3 h. The reaction was poured into water and extracted with EtOAc. The extract was washed with brine, the solvent was removed and the product was purified by flash chromatography (DCM+MeOH (+1% formic acid), 5% to 10%), triturated with DCM and dried at 100° C. to yield N-[2,6-difluoro-3-[5-[4-(1H-tetrazol-5-yl)phenyl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (38.0 mg, 0.0724 mmol, 49% yield). Analytical Data: H NMR (200 MHz, DMSO) δ 14.99 (s, 1H), 9.83 (s, 1H), 9.14 (d, J=2.1 Hz, 1H), 8.88 (d, J=2.2 Hz, 1H), 8.17 (dd, J=19.9, 8.4 Hz, 4H), 7.65 (td, J=9.0, 6.1 Hz, 1H), 7.32 (td, J=9.0, 1.4 Hz, 1H), 3.20-3.05 (m, 2H), 1.90-1.59 (m, 2H), 0.97 (t, J=7.4 Hz, 3H). 13C NMR (101 MHz, DMSO) δ 182.5, 156.29 (dd, J=248.3, 6.4 Hz), 152.8 (dd, J=251.2, 8.2 Hz), 152.4, 149.8, 142.0, 139.7, 131.7, 129.9, 129.8, 128.3, 127.9, 127.7, 127.6, 123.8, 121.79 (dd, J=13.4, 3.4 Hz), 117.1, 113.5, 112.12 (dd, J=22.6, 4.3 Hz), 53.8, 16.8, 12.5. TLC-MS (ESI−): m/z=522.9 [M−H]−. Example 75: Synthesis of N-(3-(5-(4-(1H-tetrazol-5-yl)phenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)-2,6-difluorophenyl)-1-phenylmethanesulfonamide A vessel was charged with N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]-1-phenylmethanesulfonamide (76.0 mg, 0.129 mmol), [4-(1H-tetrazol-5-yl)phenyl]boronic acid (26.9 mg, 0.141 mmol) and Pd(dppf)Cl2.DCM (5.25 mg, 0.00643 mmol) and purged with argon. Degassed 1,4-dioxane (0.428 mL) and degassed 1.5 M aqueous potassium carbonate (0.343 mL, 0.514 mmol) were added, the vessel was sealed and the reaction was heated to 80° C. for 2.5 h. The reaction was diluted with MeOH (1 mL), acidified with conc. HCl (1 mL) and stirring was continued at 60° C. for 3 h. The reaction was poured into water and extracted with EtOAc. The extract was washed with brine, the solvent was removed and the product was purified by flash chromatography (DCM/MeOH (+1% formic acid) gradient, from 5% to 15% MeOH (1% formic acid)), triturated with DCM and dried at 100° C. to yield N-[2,6-difluoro-3-[5-[4-(1H-tetrazol-5-yl)phenyl]-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]-1-phenylmethanesulfonamide (40.0 mg, 0.0699 mmol, 54% yield). TLC-MS (ESI−): m/z 571.0 [M−H]− Example 76: Synthesis of N-(2,6-difluoro-3-(5-(4-hydroxyphenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl)phenyl)propane-1-sulfonamide A vessel was charged with N-[3-[5-bromo-1-(oxan-2-yl)pyrazolo[3,4-b]pyridine-3-carbonyl]-2,6-difluorophenyl]propane-1-sulfonamide (72.0 mg, 0.133 mmol), (4-hydroxyphenyl)boronic acid (20.1 mg, 0.146 mmol), Potassium fluoride (23.1 mg, 0.398 mmol) and Pd(dppf)Cl2DCM (5.41 mg, 0.00663 mmol) and purged with argon. Degassed 1,4-dioxane/water (4+1) was added and the mixture stirred at 80° C. 20 minutes. After cooling, the mixture was diluted with 1 mL MeOH, acidified with 0.3 mL conc. HCl and stirred at 60° C. for 2 h. The reaction was taken up in water and EtOAc, the organic phase was washed with brine, dried over Na2SO4and evaporated. The product was purified by flash chromatography (DCM 7 EtOAc gradient, from 20% to 60% EtOAc) and triturated with DCM to yield N-[2,6-difluoro-3-[5-(4-hydroxyphenyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonyl]phenyl]propane-1-sulfonamide (39.0 mg, 0.0817 mmol, 62% yield). Analytical Data:1H NMR (400 MHz, DMSO) δ 14.70 (s, 1H), 9.70 (s, 2H), 8.94 (d, J=1.9 Hz, 1H), 8.64 (d, J=2.0 Hz, 1H), 7.85 (dd, J=14.5, 7.8 Hz, 1H), 7.63 (d, J=8.5 Hz, 2H), 7.39 (t, J=8.8 Hz, 1H), 6.93 (d, J=8.5 Hz, 2H), 3.20-3.06 (m, 2H), 1.87-1.75 (m, 2H), 1.00 (t, J=7.4 Hz, 3H). 13C NMR (101 MHz, DMSO) δ 185.2, 160.7 (dd, J=254, 4 Hz), 157.6, 157.0 (dd, J=256, 4 Hz), 151.7, 149.2, 141.5, 132.7, 130.3 (dd, J=10, 5 Hz), 128.5, 127.9, 126.6, 123.7 (dd, J=14, 4 Hz), 116.1, 114.2, 111.9 (dd, J=22, 3 Hz), 54.9, 16.9, 12.6. TLC-MS (ESI−): m/z 450.9, 471.0 [M−H]−. Example 77: Biological Activity Example 77-1: Binding Assays The kinase activities of the compounds of the invention were measured using KINOMEscan™ Profiling Service at DiscoveRx Corporation, 42501 Albrae St. Fremont, CA 94538, USA which is based on a competition binding assay that quantitatively measures the ability of a compound to compete with an immobilized, active-site directed ligand. The assay was performed by combining three components: DNA-tagged kinase; immobilized ligand; and a test compound. The ability of the test compound to compete with the immobilized ligand was measured via quantitative PCR of the DNA tag. The technology is described in detail in Fabian, M. A. et al. A small molecule-kinase interaction map for clinical kinase inhibitors. Nat. Biotechnol., 23, 329-336 (2005) and in Karaman, M. W. et al. A quantitative analysis of kinase inhibitor selectivity, Nat. Biotechnol., 26, 127-132 (2008). For investigation of the affinity to MKK4, MKK7 and JNK1, the kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at RT to generate affinity resins for kinase assays. The liganded beads were blocked with excess biotin and washed with blocking buffer (SEABLOCK™ (Pierce), 1% BSA, 0.05% TWEEN®20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific binding. Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in 1× binding buffer (20% SEABLOCK™, 0.17×PBS, 0.05% TWEEN®20, 6 mM DTT). All reactions were performed in polystyrene 96-well plates in a final volume of 0.135 mL. The assay plates were incubated at RT with shaking for 1 hour and the affinity beads were washed with wash buffer (1×PBS, 0.05% TWEEN®20). The beads were then re-suspended in elution buffer (1×PBS, 0.05% TWEEN®20, 0.5 11M non-biotinylated affinity ligand) and incubated at RT with shaking for 30 minutes. The kinase concentration in the eluates was measured by qPCR. Average Z′ values and standard deviations were calculated for each kinase based on fourteen control wells per experiment in over 135 independent experiments spanning a period of sixteen months. Average Z′=0.71. Potency of Test Compounds: The compounds were screened at the indicated concentrations and results for binding interactions are reported as [% of control], where lower numbers indicate stronger binding, i.e. higher potency. Details regarding the kinases tested are given in table 2 below. The test compounds were provided as 10 mM stock solutions. The test solutions at indicated final concentrations were prepared at DiscoverX. The results are given in tables 3 to 6 below. TABLE 2MKK4MKK7JNK1GroupSTESTECMCGKinasePartial LengthFull LengthFull lengthConstructAccessionNP_003001.1NP_660186.1NP_002741.1NumberSpeciesHumanHumanHumanKinase FormWild TypeWild TypeWild TypeExpressionMammalianMammalianMammalianSystemAmino AcidS84/D399M1/R419M1/Q384Start/StopAverage Z′0.670.780.79Panel MKK4 Potency: Potency of Examples 2-43 against the protein kinase MKK4, expressed as residual percent of control binding (PoC), was determined at a concentration of 100 nM. The results are given in table 3 below (N/D means not determined). TABLE 3ExampleMKK42++3+4+5++6++7O8++9++10O11+12+13++14++15+++16++17++18+++19+++20++21++22O23++24++25++26+27++28+29+30O31++32+33O34+35+36+37++38+39+40++41+42+43+44O45O46O47O48O49O50O51a+++51b+++51c+++51d+++51e+++51f+++51g+++51h++51i++51j++51k+++51l+++51m++52a++52b++52c++53a++53bO53c+++53d+54aN/D54bN/D54cN/D55N/D56N/D57N/D58+++59N/D60N/D61+++62+++63+++64N/D65N/D66N/D67N/D68+++69+++70N/D71N/D72N/D73N/D74N/D75N/D76N/DPoC < 1 = “+++”;1 ≤ PoC < 10 = “++”;10 ≤ PoC < 30 = “+”;PoC ≥ 30 = “O”. Selectivity Against JNK1: Selectivity of Examples 2-67 against the off-target JNK1, determined by calculation of the ratio of residual percent of control binding (PoC) to JNK1 and MKK4, was determined at a concentration of 100 nM. The results are given in table 4 below. TABLE 4ExampleSelectivity vs. JNK12++3+4+5++6++7O8+++9++10O11+12+13++14++15+++16++17++18+++19+++20++21+22O23++24+++25+++26+27+++28+29+30O31+++32+33O34O35+36+37++38+39+40+++41+42+43+44O45O46O47O48O49O50O51a+++51b+++51c+++51d+++51e+++51f+++51g+++51h+++51i++51j+++51k+++51l+++51m+++52a++52b++52c+++53a+++53bO53c+++53dO54aN/D54bN/D54cN/D55N/D56N/D57N/D58+++59+++60N/D61+++62+++63+++64N/D65N/D66N/D67N/D68+++69+++70N/D71N/D72N/D73N/D74N/D75N/D76N/DPoC(JNK1)/PoC(MKK4) ≥ 30 = “+++”;30 > PoC(JNK1)/PoC(MKK4) ≥ 10 = “++”;10 > PoC(JNK1)/PoC(MKK4) ≥ 3 = “+”;PoC(JNK1)/PoC(MKK4) < 3 = “O”. MKK4 Potency and Selectivity Against MKK7: Selectivity of Examples 2-43 against the off-target MKK7, determined by calculation of the ratio of residual percent of control binding (P) to MKK7 and MKK4, was determined at a concentration of 100 nM. The results are given in table 5 below. TABLE 5ExampleSelectivity vs. MKK72+++3+4+5++6++7O8+++9++10O11+12+13++14+++15+++16++17++18+++19+++20++21++22O23++24+++25+++26+27+++28+29+30O31+++32+33O34+35+36+37+++38+39+40+++41+42+43+44O45O46O47O48O49O50O51a+++51b+++51c+++51d+++51e+++51f+++51g+++51h+++51i+++51j+++51k+++51l+++51m+++52a++52b++52c+++53a+++53bO53c+++53d+54aN/D54bN/D54cN/D55N/D56N/D57N/D58+++59+++60N/D61+++62+++63+++64N/D65N/D66N/D67N/D68+++69++70N/D71N/D72N/D73N/D74N/D75N/D76N/DPoC(MKK7)/PoC(MKK4) ≥ 30 = “+++”;30 > PoC(MKK7)/PoC(MKK4) ≥ 10 = “++”;10 > PoC(MKK7)/PoC(MKK4) ≥ 3 = “+”;PoC(MKK7)/PoC(MKK4) < 3 = “O”. MKK4 Potency and Selectivity Against BRaf: Selectivity of Examples 2-43 against the off-target BRaf, determined by calculation of the ratio of residual percent of control binding (PoC) to BRaf and MKK4, was determined at a concentration of 100 nM. The results are given in table 6 below. TABLE 6ExampleSelectivity vs. BRaf2++3O4+5++6O7O8O9+10O11+12O13+14+++15+16O17+18++19++20O21+22O23+24O25++26+27O28O29O30O31++32O33O34O35+36O37+38+39+40+++41+42+43+44O45O46O47O48O49O50O51a+++51b++51c+51d++51e+51f+51g+51hO51iO51jO51kO51lO51mO52a+52bO52c+53a+++53bO53c+++53dO54aN/D54bN/D54cN/D55N/D56N/D57N/D58+++59+++60N/D61+++62+++63+++64N/D65N/D66N/D67N/D68+++69+++70N/D71N/D72N/D73N/D74N/D75N/D76N/DPoC(BRaf)/PoC(MKK4) ≥ 30 = “+++”;30 > PoC(BRaf)/PoC(MKK4) ≥ 10 = “++”;10 > PoC(BRaf)/PoC(MKK4) ≥ 3 = “+”;PoC(BRaf)/PoC(MKK4) < 3 = “O”. Example 77-2: Functional Enzyme Assays (a) Material Recombinant kinase proteins (commercially available) MEKK2, recombinant, active: ProQinase product #0583-0000-1 MKK4, recombinant, activated: ProQinase product #0948-0000-1 MKK4, recombinant, non activated: ProQinase product #0948-0000-2 Substrate Proteins Casein (Sigma C-4765) JNK1 K55R/K56R, recombinant, inactive: ProQinase product #0524-0000-1 (b) Methods (b-1) MEKK2 Dependent MKK4 Activation MKK4 (non activated) is incubated with MEKK2 (active) in a ratio of 10:1 (w/w), corresponding to a molar ratio of 20:1, in the presence of compound or vehicle and 20 μM ATP for 30 min at 30° C. The activation step is done in 50 mM HEPES pH 7.5, 50 mM NaCl, 3.8 mM MgCl2, 2.5 mM DTT, 10% (v/v) glycerol. Final DMSO concentration is 1%. The activation mixture is pipetted in the following order:2.5 μl compound in 4% DMSO2.5 μl ATP/MgCl2 mix5 μl premixed kinase solution MKK4:MEKK2 10:1 (w/w) Protein concentrations in the activation mix are 1 μM MKK4 and 50 nM MEKK2. (b-2) Protein Kinase Assay A radiometric protein kinase assay was used for measuring the kinase activity of the respective protein kinases. All kinase assays were performed in 96-well polypropylene plates. After the reactions were stopped, the assay mixtures were transferred to 96-well MSFC filter-plates (Millipore). The reaction mix was passed through the filter membrane by aspiration, the membrane was washed 3 times with 150 mM H3PO4, once with ethanol, dried and liquid scintillation cocktail was added. Radioactivity was determined by counting of the samples in a Microbeta multiwell scintillation counter (Wallac). The reactions were pipetted in the following order:a) MEKK2-MKK4 activation mix20 μl standard assay buffer10 μl MEKK2-MKK4 activation mix5 μl radioactive33P-γ-ATP solution (typically 106 cpm/well)10 μl of substrate solutionb) Single Kinases20 μl standard assay buffer5 μl compound in 10% DMSO20 μl enzyme-substrate mix10 μl of substrate solution The assay contained 70 mM HEPES-NaOH, pH 7.5, 3 mM MgCl2, 3 mM MnCl2, 3 □M Na-orthovanadate, 1.2 mM DTT, ATP (variable amounts, corresponding to the apparent ATP-Kmof the respective kinase, see Table 1), [33P-γ-ATP (approx. 8×105cpm per well), protein kinase (variable amounts; see Table 1), and substrate (variable amounts; see Table 7 below). The results are given in table 8 below. TABLE 7Enzymes, substrates, and assay conditions (amounts/well)KinaseKinaseATPKinaseConc.Conc.Conc.Substrate#Nameng/50 μlnMμMNameμg/50 μlnM1MKK4-MEKK225100.2JNK11430mixKRKR2MKK4 active25100.2JNK11430KRKR3MEKK2150300.2Casein1870 TABLE 8Potency of Test CompoundsExplCascadeMKK4MEKK22++++◯3++++◯4++◯5++++◯8◯+◯9++++13+++++14+++◯15+++++17++◯18++++++19+++++21++++23◯◯◯24◯++◯25+++◯27++++++31+++++32+++◯37++++++39◯+◯40◯+◯41++◯42++◯51a+++++51b++++51c++++++51d+++++++51e+++++++51f+++++51g++++51j++++◯76N/D+++N/D51k++++++◯51l++++51m+++++52aN/D+++N/D52bN/D++N/D52cN/D++N/D53aN/D++N/D53bN/D++N/D53cN/D+++N/D53dN/D++N/D54aN/D+++N/D54bN/D+++N/D54cN/D+++N/D55N/D+++N/D57N/D+++N/D58N/D+++N/D59N/D+++N/D61N/D+++N/D62N/D+++N/D63N/D+++N/D64N/D+++N/D65++++66+++++67+++++++++68N/D+++N/D69N/D+++N/D70+++++++71+++++◯72++++73N/D+++N/D74+++++++75++++++*: potency derived from IC50-values (PoC) according to the followingclassification rule:IC50≥ 10 μM10 > IC50≥ 1 μM1 > IC50≥ 0.5 μMIC50< 0.5 μM◯++++++
199,219
11858928
DETAILED DESCRIPTION OF THE INVENTION The present invention relates to a process for preparation of compound of formula (1) or a salt thereof, the process comprising: a. Reacting compound of formula (3) and compound of formula (4) to provide compound of formula (2), in a solvent mixture comprising methanol and butanol; b. Adding water; c. Isolating solid crystalline form of compound (2) characterized by XRPD pattern having 2θ values 5.5°, 7.5°, 9.7°, 10.1°, 14.5°, 15.9°, 18.1°, 20.2° and 21.5° degrees 2 theta (±0.2 degrees 2 theta); d. Transforming compound of formula (2) into compound of formula (1). Compounds (3) and (4) are either commericially available or can be prepared by processes known in the prior art, for example described in Org. Process Dev. 2016, 20, 1191-1202, Org. Process Dev. 2016, 20, 1203-1216, Org. Process Dev. 2016, 20, 1217-1226. Prior art describes the reaction step a. done in sole butanol as a reaction solvent. We have surprisingly found that use of mixture of methanol and butanol, preferably a mixture comprising 30 to 99% (wt) of methanol, more preferably 30 to 65%, even more preferably 50-65% of methanol, allows both decrease of reaction temperature and decrease in reaction times. That result in a more economical process which is suitable for higher scales and in a decrease of impurities formed in the course of reaction. In the beginning of the reaction the reaction mixture is a suspension because compound (3) is poorly soluble in the used solvent mixture. When the mixture of methanol and butanol comprises between 30-65% of methanol, preferably between 50 and 65% of methanol, the reaction mixture is a suspension in the begining, during the course of the reaction it becomes a solution, because compound of formula (2) is soluble in the solvent mixture and the reaction mixture can be optionally filtered after the reaction is finished. When the solvent mixture comprises more than 65% of methanol it remains suspension, because compound (2) is not soluble in this solvent mixture. The molar ratio between compound (3) and compound (4) is preferably between 1:2 and 1:6, even more preferably it is between 1:2.5 and 1:3.5. The concentration of compound (3) in the mixture of methanol and butanol is preferably between 0.5 g/g and 3 g/g, more preferably it is between 0.8 g/g and 1.5 g/g. The reaction between compounds (3) and (4) is performed in the presence of Pd catalyst, for example Pd/C, Pd(OAc)2 or [1,1′-bis(diphenylphosphino)ferrocene]dichloro-palladium (II). The reaction can be optionally performed in the presence of a phosphine, for example bis-[2-(diphenylphoshpino)phenyl]ether (DPEPhos). The molar ratio of used Pd catalyst and compound of formula (3) is preferably between 1:30 and 1:45, more preferably between 1:35 and 1:41. The molar ratio between Pd catalyst and the phosphine is preferably between 1:1 and 1:1.5, more preferably it is 1:1.2. The reaction between compounds (3) and (4) is preferably performed at a temperature between 45 and 75° C., more preferably at a temperature between 55 and 65° C., even more preferably between 60° C. and 63° C. for 2 to 7 hours preferably for 3 to 5 hours. The reaction progress can be monitored by any suitable analytical method for example by HPLC or GC. When the reaction is finished, methanol can be added to the mixture so that the mixture at the end of methanol addition comprises 70-99% (wt) of methanol. To the mixture water is added (step b.). The ratio (wt:wt) between water and the solvent mixture of butanol and methanol is preferably between 1:2.5 and 1:6, more preferably between 1:3 and 1:5. Water is preferably added for a time period between 20 and 120 minutes. The mixture is cooled preferably to a temperature between −10° C. and 10° C., more preferably to a temperature between 0° C. and 5° C. and stirred at this temperature for 30 to 240 minutes, preferably for 60 minutes to precipitate compound of formula (2) from the mixture. Obtained solid form of compound of formula (2) is isolated (step c.) from the mixture by any suitable technique, for example by filtration. Isolated solid form of compound of formula (2) is characterized by XRPD pattern having 2θ values 5.5°, 7.5°, 9.7°, 10.1°, 14.5°, 15.9°, 18.1°, 20.2° and 21.5° degrees 2 theta (±0.2 degrees 2 theta). The solid form can be further characterized by XRPD pattern having 2θ values 5.5°, 7.5°, 9.7°, 10.1°, 11.6°, 14.5°, 15.0°, 15.4°, 15.9°, 17.0°, 18.1°, 18.7°, 20.2° and 21.5° degrees 2 theta (±0.2 degrees 2 theta). The solid form can be also characterized by a XRPD spectrum depicted inFIG.1. Org. Process Dev. 2016, 20, 1203-1216 describes three solid forms of compound (2), Forms A, B and C. The solid form of compound (2) prepared according to the presented invention not only shows a good crystallinity and stability but also a better purity with respect to the content of palladium in comparision with the Forms A, B or C prepared according to the prior art. Process Dev. 2016, 20, 1203-1216 discloses that prepared solid forms of compound of formula (2) contain 500 ppm of palladium, the content thereof can be further decreased to 200 ppm by using of 1,2-diaminopropane. Solid form of compound of formula (2) prepared according to the presented invention comprises typically 10-40 ppm of palladium, the content thereof can be decreased to 3-5 ppm by using of 1,2-diaminopropane. The compound of formula (2) can be further recrystallized by a process comprising: i. Mixing the solid form of compound (2) with a mixture comprising methanol and tetrahydrofuran; ii. Heating the mixture; iii. Adding water; iv. Isolating a solid form of compound of formula (2); The ratio (wt:wt) between methanol and tetrahydrofurane in step i. is preferably between 3:1 and 18:1, more preferably it is between 4.5:1 and 5.5:1, and even more preferred ratio is 5.2:1. The concentration of compound (2) in the solvent mixture can be between 0.03 g/ml and 0.09 g/ml. The mixture is heated (step ii.) to a temperature preferably between 40° C. and 75° C., more preferably between 50° C. and 60° C. to dissolve the compound of formula (2). The mixture is stirred at this temperature preferably for between 5 and 300 minutes. To the mixture water is added (step iii.). The ratio water:mixture of methanol and tetrahydrofurane (wt:wt) is preferably between 1:2.5 and 1:7, more preferably between 1:3 and 1:4. The mixture can be cooled to a temperature between −10° C. and 25° C. Solid form of compound of formula (2) can be isolated (step iv.) by any suitable technique, for example by filtration. The isolated solid form of compound of formula (2) is characterized by XRPD pattern having 2θ values 5.5°, 7.5°, 9.7°, 10.1°, 14.5°, 15.9°, 18.1°, 20.2° and 21.5° degrees 2 theta ( ±0.2 degrees 2 theta). The solid form can be further characterized by XRPD pattern having 2θ values 5.5°, 7.5°, 9.7°, 10.1°, 11.6°, 14.5°, 15.0°, 15.4°, 15.9°, 17.0°, 18.1°, 18.7°, 20.2° and 21.5° degrees 2 theta (±0.2 degrees 2 theta). The solid form can be also characterized by a XRPD spectrum depicted inFIG.1. The compound of formula (2) is further transformed into compound of formula (1) by processes known in the prior art. For example, compound of formula (2) is mixed with an acid in a solvent or a solvent mixture. Suitable acids are for example hydrochloric acid, nitric acid, phosphoric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, acetic acid, oxalic acid, valeric acid, oleic acid, palmitic acid, stearic acid, lauric acid, boric acid, benzoic acid, lactic acid, benzenesulfonic acid, citric acid, maleic acid, fumaric acid, succinic acid, tartaric acid, naphthalene dicarboxylic acid, methanesulfonic acid, lactic acid, lauryl sulfuric acid, isethionic acid. The molar ratio between compound of formula (2) and the acid can be between 1:2 and 1:8, preferably it is between 1:4 and 1:6. Suitable solvents are for example ketones, esters, ethers, amides, nitriles or organic acids, alcohols, aliphatic and aromatic hydrocarbons, chlorinated hydrocarbons, water or their mixtures. Aliphatic C1-C4alcohols, esters or their mixtures are preferred. The most commonly used solvents are methanol, ethanol, water or their mixtures. The concentration of compound of formula (2) in the solvent can be between 0.08 g/ml and 0.20 g/ml, preferably it is between 0.1 g/ml and 0.15 g/ml. The reaction mixture can be heated to a temperature between 40° C. and the reflux temperature of used solvent(s) and stirred at this temperature for 1 to 10 hours. The reaction progress can be monitored by any suitable analytical method for example by HPLC or GC. The mixture can be cooled to a temperature between −10° C. and the room temperature. A solid form of a salt of compound of formula (1) with the suitable acid can be optionally isolated by for example by filtration. Compound of formula (1) is obtained from a salt of compound of formula (1) by contacting a solution of the the salt with a base (for example NaOH, KOH, NaHCO3, Na2CO3, K2CO3, KHCO3) in a suitable solvent (for example a mixture of C1-C4alcohol and water). The final pH of the mixture should be higher that 8, preferably it is between 10-11. Obtained solid form of compound of formula (1) can be isolated by any suitable technique, for example by filtration. Obtained solid form of compound of formula (1) (palbociclib) preferably has the surface area between 6-16 m2/g, more preferably between 7-14 m2/g and is preferably in polymorph form A disclosed in WO2014128588. Obtained solid compound of formula (1) comprises less than 30 ppm of palladium, preferably less than 10 ppm of palladium. The invention will be further described with reference to the following non-limiting examples. EXAMPLES XRPD spectrum was obtained using the following measurement conditions: Panalytical Empyrean diffractometer with Θ/2Θ geometry (transmition mode), equipped with a PixCell 3D detector; Start angle (2θ):2.0°End angle (2θ):35.0°Step size:0.026°Scan speed:0.0955°/secondsRadiation type:CuRadiation wavelengths:1.5406 Å (Kα1), primarymonochromator usedDivergence slit:½°Antiscatter slit:½°Soller slit:0.02radDetector slit:7.5mmRotation speed:30rpm The specific surface area (SSA) is assessed with a Quantachrome NOVA touch 2LX. The N2is used as measuring gas and BET method is used to evaluate SSA. The following setup has been used for the measurement: Adsorbate: Nitrogen Sample cell: 9 mm large bulb cell, long; Measurement is performed with the corresponding filler rod Sample masses*: approximately ¾ of cell bulb Degassing conditions: 960 min at 30° C. under vacuum (ramping 10° C./min) Measured points of isotherm: 1 lequidistant points in the range 0.05-0.30 p/p0** Points analyzed using BET: 7 equidistant points from whole range 0.05-0.20 p/p0 Use data reduction adsorbate model: Yes (N2) Bath thermal delay: at least 600 s P0Option: continuous (measured each 30 minutes) Void volume mode: Helium measure Equilibration time: 160 s Equilibration timeout: 2400 s Tolerance: 0.05 * Used amount of sample depends on its SSA and other physical parameters. The real surface area of sample in measuring cell should be between 1 m2/g and 20 m2/g. ** Points above or below this range can be measured. Example 1 Preparation of a Solid Form of Compound of Formula (2) 100 g of compound of formula (3) was suspended in 375 g of dry 1-butanol and 625 g of dry methanol at 40° C. The suspension was placed under nitrogen and 50 g of compound of formula (4), 50 g of diisopropylethylamine, 1 g of Pd(OAc)2 and 3 g of DPEPhos were added. The reaction mixture was heated at 60° C. and stirred at this temperature for 3 hours. The mixture was filtered, to the fitrate 250 g of methanol and 300 g of water during 40 minutes were added. The suspension was cooled to 0-5° C., solid compound (2) was filtered off and washed with a mixture of 240 g of methanol and 40 g of water. The fitrer cake was dried at 65° C. 94 g of compound of formula (2) was obtained (yield 90% of the theoretical yield) in HPLC purity 99.1%. XRPD pattern of the obtained solid corresponds to XRPD pattern depicted inFIG.1. The content of palladium in obtained solid was 25 ppm. Example 2 Preparation of a Solid Form of Compound of Formula (2) 2 g of compound of formula (3) was suspended in 3.75 g of dry 1-butanol and 75 g of dry methanol at 40° C. The suspension was placed under nitrogen and 1.2 g of compound of formula (4), 1.2 g of diisopropylethylamine, 0.0625 g of Pd(OAc)2 and 0.1875 g of DPEPhos were added. The reaction mixture was heated at 55° C. and stirred at this temperature for 3 hours. The mixture was filtered, to the fitrate 36 g of water during 15 minutes were added. The suspension was cooled to 0-5° C., solid compound (2) was filtered off and washed with a mixture of 29 g of methanol and 5 g of water. The fitrer cake was dried at 65° C. 1.81 g of compound of formula (2) was obtained (yield 88% of the theoretical yield) in HPLC purity 98.9%. XRPD pattern of the obtained solid corresponds to XRPD pattern depicted inFIG.1. The content of palladium in obtained solid was 28 ppm. Example 3 Recrystallization of Compound of Formula (2) 5 g of compound of formula (2) prepared according to Example 1 were suspended in 75 g of methanol and 3 g of tetrahydrofuran. The suspension was heated to 55° C. to dissolve the compound of formula (2). To the mixture 25 g of water were added during 10 minutes. The suspension was cooled to 0-5° C. and stirred at this temperature for 10 minutes. The mixture was filtered off, the filter cake was washed with a mixture of 12 g of methanol and 2 g of water. 4.7 g of compound of formula (2) (94.6% of the theoretical yield) was obtained. XRPD pattern of the obtained solid corresponds to XRPD pattern depicted inFIG.1. The content of palladium in obtained solid was 13 ppm. Example 4 Process for Preparation of Compound of Formula (1) 100 g of compound of formula (2) prepared according to Example 3 was suspended in 790 g of methanol. Into the suspension 115 g of 26.2% solution of HCl in ethanol was added during 10 minutes. The mixture was heated at 55-57° C. and was stirred at this temperature for 2 hours. The suspension was cooled to 0-5° C. and filtered off. The cake was washed with 200 g of methanol. Filtrated solid was dissolved in a mixture of 400 g methanol and 500 g of water. The mixture was heated to 40° C. To the mixture 15.4 g of NaOH in 155 g of water was added during 60 minutes to set the pH of the mixture to 10-11. The suspension was stirred for 60 minutes, cooled to 25° C. and filtered. Then filtration cake was washed with 2×500 g of water and 157 g of acetone. The cake was dried to provide 69 g of compound of formula (1) with HPLC purity 99.9% (HPLC IN). The specific surface area of obtained compound of formula (1) was 9.5 m2/g and the content of palladium in the obtained solid was 10 ppm. The invention having been described, it will be readily apparent to those skilled in the art that further changes and modifications in actual implementation of the concepts and embodiments described herein can easily be made or may be learned by practice of the invention, without departing from the spirit and scope of the invention as defined by the following claims.
15,324
11858929
DETAILED DESCRIPTION The illustrative embodiments described in the detailed description and claims are not meant to be limiting. Other embodiments may be utilized, and other changes may be made, without departing from the spirit or scope of the subject matter presented here. Fascin is an actin-bundling protein. For cell migration to proceed, actin cytoskeleton must be reorganized by forming polymers and bundles to affect the dynamic changes of cell shapes (References 13-15). Individual actin filaments are flexible and elongation of individual filaments per se is insufficient for membrane protrusion which is necessary for cell migration. Bundling of actin filaments provides rigidity to actin filaments for protrusions in the form of lamellipodia and filopodia against the compressive force from the plasma membrane (Reference 16) (Reference 17). As noted, one of the critical actin-bundling proteins is fascin (Reference 18-22). Fascin is the primary actin cross-linker in filopodia and shows no sequence homology with other actin-binding proteins (Reference 23). It is required to maximally cross-link the actin filaments into straight, compact, and rigid bundles (Reference 24). Elevated levels of fascin have been found in many types of metastatic tumors (including breast, prostate, ovarian, lung, gastric, esophageal, and others) and are correlated with clinically aggressive phenotypes, poor prognosis, and shorter survival (Reference 25-29) (Reference 30, 31) (Reference 32-34). Fascin inhibitors may target tumor cell migration and invasion, and provide treatments for metastatic cancer. Definitions The technology is described herein using several definitions, as set forth throughout the specification. The use of the terms “a” and “an” and “the” and similar referents in the context of describing the elements (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. As used herein, “about” will be understood by persons of ordinary skill in the art and will vary to some extent depending upon the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art, given the context in which it is used, “about” will mean up to plus or minus 10% of the particular term. A dash (“-”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, —CONH2is attached through the carbon atom. By “optional” or “optionally” is meant that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, “optionally substituted alkyl” encompasses both “alkyl” and “substituted alkyl” as defined herein. It will be understood by those skilled in the art, with respect to any group containing one or more substituents, that such groups are not intended to introduce any substitution or substitution patterns that are sterically impractical, synthetically non-feasible and/or inherently unstable. “Alkyl” encompasses straight chain and branched chain having the indicated number of carbon atoms, usually from 1 to 20 carbon atoms, for example 1 to 8 carbon atoms, such as 1 to 6 carbon atoms. For example C1-C6alkyl encompasses both straight and branched chain alkyl of from 1 to 6 carbon atoms. Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl, 3-methylpentyl, and the like. Alkylene is another subset of alkyl, referring to the same residues as alkyl, but having two points of attachment. Alkylene groups will usually have from 2 to 20 carbon atoms, for example 2 to 8 carbon atoms, such as from 2 to 6 carbon atoms. For example, C0alkylene indicates a covalent bond and C1alkylene is a methylene group. When an alkyl residue having a specific number of carbons is named, all geometric isomers having that number of carbons are intended to be encompassed; thus, for example, “butyl” is meant to include n-butyl, sec-butyl, isobutyl and t-butyl; “propyl” includes n-propyl and isopropyl. “Lower alkyl” refers to an alkyl group having 1 to 4 carbons. “Alkenyl” refers to straight or branched hydrocarbyl groups having the indicated number of carbon atoms, usually from 1 to 8 carbon atoms, for example 2 to 4 carbon atoms, and at least 1 and preferably from 1 to 2 sites of vinyl (>C═C<) unsaturation. Such groups are exemplified, for example, by vinyl, allyl, and but-3-en-1-yl. Included within this term are the cis and trans isomers or mixtures of these isomers. “Lower alkenyl” refers to an alkenyl group having 1 to 4 carbons, which can be indicated by C2-C4alkenyl. “Cycloalkyl” indicates a non-aromatic partially saturated, or fully saturated carbocyclic ring having the indicated number of carbon ring atoms, for example, 3 to 10, or 3 to 8, or 3 to 6 ring carbon atoms. Cycloalkyl groups may be monocyclic or polycyclic (e.g., bicyclic, tricyclic). Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl and cyclohexyl, as well as bridged and caged ring groups (e.g., norbornane, bicyclo[2.2.2]octane). In addition, one ring of a polycyclic cycloalkyl group may be aromatic, provided the polycyclic cycloalkyl group is bound to the parent structure via a non-aromatic carbon. For example, a 1,2,3,4-tetrahydronaphthalen-1-yl group (wherein the moiety is bound to the parent structure via a non-aromatic carbon atom) is a cycloalkyl group, while 1,2,3,4-tetrahydronaphthalen-5-yl (wherein the moiety is bound to the parent structure via an aromatic carbon atom) is not considered a cycloalkyl group. Examples of polycyclic cycloalkyl groups consisting of a cycloalkyl group fused to an aromatic ring are described below. “Aryl” indicates an aromatic carbon ring having the indicated number of carbon atoms, for example, 6 to 12 or 6 to 10 carbon atoms, in the ring. Aryl groups may be monocyclic or polycyclic (e.g., bicyclic, tricyclic). In some instances, both rings of a polycyclic aryl group are aromatic (e.g., naphthyl). In other instances, polycyclic aryl groups may include a non-aromatic ring (e.g., cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl) fused to an aromatic ring, provided the polycyclic aryl group is bound to the parent structure via an atom in the aromatic ring. Thus, a 1,2,3,4-tetrahydronaphthalen-5-yl group (wherein the moiety is bound to the parent structure via an aromatic carbon atom) is considered an aryl group, while 1,2,3,4-tetrahydronaphthalen-1-yl (wherein the moiety is bound to the parent structure via a non-aromatic carbon atom) is not considered an aryl group. Similarly, a 1,2,3,4-tetrahydroquinolin-8-yl group (wherein the moiety is bound to the parent structure via an aromatic carbon atom) is considered an aryl group, while 1,2,3,4-tetrahydroquinolin-1-yl group (wherein the moiety is bound to the parent structure via a non-aromatic nitrogen atom) is not considered an aryl group. However, the term “aryl” does not encompass or overlap with “heteroaryl”, as defined herein, regardless of the point of attachment (e.g., both quinolin-5-yl and quinolin-2-yl are heteroaryl groups). In some instances, aryl is phenyl or naphthyl. In certain instances, aryl is phenyl. Additional examples of aryl groups comprising an aromatic carbon ring fused to a non-aromatic ring are described below. “Carboxy” or “carboxyl” refers to —COOH or a salt thereof. “Heteroaryl” indicates an aromatic ring containing the indicated number of ring atoms (e.g., 5 to 12, or 5 to 10 membered heteroaryl) made up of one or more heteroatoms (e.g., 1, 2, 3 or 4 heteroatoms) selected from N, O and S and with the remaining ring atoms being carbon. 5-Membered heteroaryl is a heteroaryl having 5 ring atoms. 6-Membered heteroaryl is a heteroaryl having 6 ring atoms. Heteroaryl groups do not contain adjacent S and O atoms. In some embodiments, the total number of S and O atoms in the heteroaryl group is not more than 2. In some embodiments, the total number of S and O atoms in the heteroaryl group is not more than 1. Unless otherwise indicated, heteroaryl groups may be bound to the parent structure by a carbon or nitrogen atom, as valency permits. For example, “pyridyl” includes 2-pyridyl, 3-pyridyl and 4-pyridyl groups, and “pyrrolyl” includes 1-pyrrolyl, 2-pyrrolyl and 3-pyrrolyl groups. When nitrogen is present in a heteroaryl ring, it may, where the nature of the adjacent atoms and groups permits, exist in an oxidized state (i.e., N+—O−). Additionally, when sulfur is present in a heteroaryl ring, it may, where the nature of the adjacent atoms and groups permits, exist in an oxidized state (i.e., S+—O−or SO2). Heteroaryl groups may be monocyclic or polycyclic (e.g., bicyclic, tricyclic). In some instances, a heteroaryl group is monocyclic. Examples include pyrrole, pyrazole, imidazole, triazole (e.g., 1,2,3-triazole, 1,2,4-triazole, 1,2,4-triazole), tetrazole, furan, isoxazole, oxazole, oxadiazole (e.g., 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,3,4-oxadiazole), thiophene, isothiazole, thiazole, thiadiazole (e.g., 1,2,3-thiadiazole, 1,2,4-thiadiazole, 1,3,4-thiadiazole), pyridine, pyridazine, pyrimidine, pyrazine, triazine (e.g., 1,2,4-triazine, 1,3,5-triazine) and tetrazine. In some instances, both rings of a polycyclic heteroaryl group are aromatic. Examples include indole, isoindole, indazole, benzoimidazole, benzotriazole, benzofuran, benzoxazole, benzoisoxazole, benzoxadiazole, benzothiophene, benzothiazole, benzoisothiazole, benzothiadiazole, 1H-pyrrolo[2,3-b]pyridine, 1H-pyrazolo[3,4-b]pyridine, 3H-imidazo[4,5-b]pyridine, 3H-[1,2,3]triazolo[4,5-b]pyridine, 1H-pyrrolo[3,2-b]pyridine, 1H-pyrazolo[4,3-b]pyridine, 1H-imidazo[4,5-b]pyridine, 1H-[1,2,3]triazolo[4,5-b]pyridine, 1H-pyrrolo[2,3-c]pyridine, 1H-pyrazolo[3,4-c]pyridine, 3H-imidazo[4,5-c]pyridine, 3H-[1,2,3]triazolo[4,5-c]pyridine, 1H-pyrrolo[3,2-c]pyridine, 1H-pyrazolo[4,3-c]pyridine, 1H-imidazo[4,5-c]pyridine, 1H-[1,2,3]triazolo[4,5-c]pyridine, furo[2,3-b]pyridine, oxazolo[5,4-b]pyridine, isoxazolo[5,4-b]pyridine, [1,2,3]oxadiazolo[5,4-b]pyridine, furo[3,2-b]pyridine, oxazolo[4,5-b]pyridine, isoxazolo[4,5-b]pyridine, [1,2,3]oxadiazolo[4,5-b]pyridine, furo[2,3-c]pyridine, oxazolo[5,4-c]pyridine, isoxazolo[5,4-c]pyridine, [1,2,3]oxadiazolo[5,4-c]pyridine, furo[3,2-c]pyridine, oxazolo[4,5-c]pyridine, isoxazolo[4,5-c]pyridine, [1,2,3]oxadiazolo[4,5-c]pyridine, thieno[2,3-b]pyridine, thiazolo[5,4-b]pyridine, isothiazolo[5,4-b]pyridine, [1,2,3]thiadiazolo[5,4-b]pyridine, thieno[3,2-b]pyridine, thiazolo[4,5-b]pyridine, isothiazolo[4,5-b]pyridine, [1,2,3]thiadiazolo[4,5-b]pyridine, thieno[2,3-c]pyridine, thiazolo[5,4-c]pyridine, isothiazolo[5,4-c]pyridine, [1,2,3]thiadiazolo[5,4-c]pyridine, thieno[3,2-c]pyridine, thiazolo[4,5-c]pyridine, isothiazolo[4,5-c]pyridine, [1,2,3]thiadiazolo[4,5-c]pyridine, quinoline, isoquinoline, cinnoline, quinazoline, quinoxaline, phthalazine, naphthyridine (e.g., 1,8-naphthyridine, 1,7-naphthyridine, 1,6-naphthyridine, 1,5-naphthyridine, 2,7-naphthyridine, 2,6-naphthyridine), imidazo[1,2-a]pyridine, 1H-pyrazolo[3,4-d]thiazole, 1H-pyrazolo[4,3-d]thiazole and imidazo[2,1-b]thiazole. In other instances, polycyclic heteroaryl groups may include a non-aromatic ring (e.g., cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl) fused to a heteroaryl ring, provided the polycyclic heteroaryl group is bound to the parent structure via an atom in the aromatic ring. For example, a 4,5,6,7-tetrahydrobenzo[d]thiazol-2-yl group (wherein the moiety is bound to the parent structure via an aromatic carbon atom) is considered a heteroaryl group, while 4,5,6,7-tetrahydrobenzo[d]thiazol-5-yl (wherein the moiety is bound to the parent structure via a non-aromatic carbon atom) is not considered a heteroaryl group. Examples of polycyclic heteroaryl groups consisting of a heteroaryl ring fused to a non-aromatic ring are described below. “Heterocycloalkyl” indicates a non-aromatic partially saturated, or fully saturated ring having the indicated number of ring atoms (e.g., 3 to 10, or 3 to 7, membered heterocycloalkyl) made up of one or more heteroatoms (e.g., 1, 2, 3 or 4 heteroatoms) selected from N, O and S and with the remaining ring atoms being carbon. 5-Membered heterocycloalkyl is a heterocycloalkyl having 5 ring atoms. 6-Membered heterocycloalkyl is a heterocycloalkyl having 6 ring atoms. Heterocycloalkyl groups may be monocyclic or polycyclic (e.g., bicyclic, tricyclic). Examples of heterocycloalkyl groups include oxiranyl, aziridinyl, azetidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, morpholinyl and thiomorpholinyl. When nitrogen is present in a heterocycloalkyl ring, it may, where the nature of the adjacent atoms and groups permits, exist in an oxidized state (i.e., N+—O−). Examples include piperidinyl N-oxide and morpholinyl-N-oxide. Additionally, when sulfur is present in a heterocycloalkyl ring, it may, where the nature of the adjacent atoms and groups permits, exist in an oxidized state (i.e., S+—O−or —SO2—). Examples include thiomorpholine S-oxide and thiomorpholine S,S-dioxide. In addition, one ring of a polycyclic heterocycloalkyl group may be aromatic (e.g., aryl or heteroaryl), provided the polycyclic heterocycloalkyl group is bound to the parent structure via a non-aromatic carbon or nitrogen atom. For example, a 1,2,3,4-tetrahydroquinolin-1-yl group (wherein the moiety is bound to the parent structure via a non-aromatic nitrogen atom) is considered a heterocycloalkyl group, while 1,2,3,4-tetrahydroquinolin-8-yl group (wherein the moiety is bound to the parent structure via an aromatic carbon atom) is not considered a heterocycloalkyl group. Examples of polycyclic heterocycloalkyl groups consisting of a heterocycloalkyl group fused to an aromatic ring are described below. By “alkoxy” is meant an alkyl group of the indicated number of carbon atoms attached through an oxygen bridge such as, for example, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentoxy, 2-pentyloxy, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3-hexoxy, 3-methylpentoxy, and the like. An alkoxy group is further meant to encompass a cycloalkyl group, as defined above, that is likewise attached through an oxygen bridge. Alkoxy groups will usually have from 1 to 6 carbon atoms attached through the oxygen bridge. “Lower alkoxy” refers to an alkoxy group having 1 to 4 carbons. The term “halo” includes fluoro, chloro, bromo, and iodo, and the term “halogen” includes fluorine, chlorine, bromine, and iodine. The term “substituted”, as used herein, means that any one or more hydrogens on the designated atom or group is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded. When a substituent is oxo (i.e., ═O) then 2 hydrogens on the atom are replaced. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds or useful synthetic intermediates. A stable compound or stable structure is meant to imply a compound that is sufficiently robust to survive isolation from a reaction mixture, and subsequent formulation as an agent having at least practical utility. Unless otherwise specified, substituents are named into the core structure. For example, it is to be understood that when (cycloalkyl)alkyl is listed as a possible substituent, the point of attachment of this substituent to the core structure is in the alkyl portion. “Haloalkyl” refers to alkyl groups substituted with 1 to 5, 1 to 3, or 1 to 2 halo groups, wherein alkyl and halo are as defined herein. Lower haloalkyl refers to a C1-C4alkyl substituted with 1 to 5, 1 to 3, or 1 to 2 halo groups. “Lower alkylphenyl” refers to C1-C4alkyl-phenyl. “Isomers” are different compounds that have the same molecular formula. “Stereoisomers” are isomers that differ only in the way the atoms are arranged in space. “Enantiomers” are stereoisomers that are non-superimposable mirror images of each other. A 1:1 mixture of a pair of enantiomers is a “racemic” mixture. The symbol “(±)” may be used to designate a racemic mixture where appropriate. “Diastereoisomers” are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other. A “meso compound” or “meso isomer” is a non-optically active member of a set of stereoisomers. Meso isomers contain two or more stereocenters but are not chiral (i.e., a plane of symmetry exists within the molecule). The absolute stereochemistry is specified according to the Cahn-Ingold-Prelog R-S system. When a compound is a pure enantiomer the stereochemistry at each chiral carbon can be specified by either R or S. Resolved compounds whose absolute configuration is unknown can be designated (+) or (−) depending on the direction (dextro- or levorotatory) which they rotate plane polarized light at the wavelength of the sodium D line. Certain of the compounds disclosed and/or described herein contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, meso isomers and other stereoisomeric forms. Unless otherwise indicated, compounds disclosed and/or described herein include all such possible enantiomers, diastereomers, meso isomers and other stereoisomeric forms, including racemic mixtures, optically pure forms and intermediate mixtures. Enantiomers, diastereomers, meso isomers and other stereoisomeric forms can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. Unless specified otherwise, when the compounds disclosed and/or described herein contain olefinic double bonds or other centers of geometric asymmetry, it is intended that the compounds include both E and Z isomers. “Tautomers” are structurally distinct isomers that interconvert by tautomerization. Tautomerization is a form of isomerization and includes prototropic or proton-shift tautomerization, which is considered a subset of acid-base chemistry. Prototropic tautomerization or proton-shift tautomerization involves the migration of a proton accompanied by changes in bond order, often the interchange of a single bond with an adjacent double bond. Where tautomerization is possible (e.g. in solution), a chemical equilibrium of tautomers can be reached. An example of tautomerization is keto-enol tautomerization. A specific example of keto-enol tautomerization is the interconverision of pentane-2,4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerization is phenol-keto tautomerization. A specific example of phenol-keto tautomerization is the interconversion of pyridin-4-ol and pyridin-4(1H)-one tautomers. When the compounds described herein contain moieties capable of tautomerization, and unless specified otherwise, it is intended that the compounds include all possible tautomers. Pharmaceutically acceptable forms of the compounds recited herein include pharmaceutically acceptable salts, and mixtures thereof. “Pharmaceutically acceptable salts” include, but are not limited to salts with inorganic acids, such as hydrochlorate, phosphate, diphosphate, hydrobromate, sulfate, sulfinate, nitrate, and like salts; as well as salts with an organic acid, such as malate, maleate, fumarate, tartrate, succinate, citrate, acetate, lactate, methanesulfonate, p-toluenesulfonate, 2-hydroxyethylsulfonate, benzoate, salicylate, stearate, and alkanoate such as acetate, HOOC—(CH2)n—COOH where n is 0-4, and like salts. Similarly, pharmaceutically acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium, and ammonium. In addition, if the compounds described herein are obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt. Conversely, if the product is a free base, an addition salt, particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds. Those skilled in the art will recognize various synthetic methodologies that may be used to prepare non-toxic pharmaceutically acceptable addition salts. The compounds disclosed and/or described herein can be enriched isotopic forms, e.g., enriched in the content of2H,3H,11C,13C and/or14C. In one embodiment, the compound contains at least one deuterium atom. Such deuterated forms can be made, for example, by the procedure described in U.S. Pat. Nos. 5,846,514 and 6,334,997. Such deuterated compounds may improve the efficacy and increase the duration of action of compounds disclosed and/or described herein. Deuterium substituted compounds can be synthesized using various methods, such as those described in: Dean, D., Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development,Curr. Pharm. Des.,2000; 6(10); Kabalka, G. et al., The Synthesis of Radiolabeled Compounds via Organometallic Intermediates,Tetrahedron,1989, 45(21), 6601-21; and Evans, E., Synthesis of radiolabeled compounds,J. Radioanal. Chem.,1981, 64(1-2), 9-32. As used herein the terms “group”, “radical” or “fragment” are synonymous and are intended to indicate functional groups or fragments of molecules attachable to a bond or other fragments of molecules. The term “active agent” is used to indicate a substance which has biological activity. In some embodiments, an “active agent” is a substance having pharmaceutical utility. For example an active agent may be an anti-metastasis therapeutic. The term “therapeutically effective amount” or “effective amount” means an amount effective, when administered to a human or non-human subject, to provide a therapeutic benefit such as amelioration of symptoms, slowing of disease progression, or prevention of disease, or to inhibit fascin activity in vitro or in vivo, e.g., a therapeutically effective amount may be an amount sufficient to decrease the symptoms of a disease responsive to inhibition of fascin activity. “Inhibition of fascin activity” refers to a decrease in fascin activity as a direct or indirect response to the presence of at least one compound, or pharmaceutically acceptable salt thereof, described herein, relative to the activity of fascin in the absence of the at least one compound, or pharmaceutically acceptable salt thereof, described herein. The decrease in activity may be due to the direct interaction of the at least one compound, or pharmaceutically acceptable salt thereof, described herein with fascin or with one or more other factors that in turn affect fascin activity. In some embodiments, the compound, or pharmaceutically acceptable salt thereof, described herein has an IC50(the concentration that inhibits 50% of fascin activity) value of about 500 micromolar, about 100 micromolar, about 10 micromolar, about 1 micromolar, about 500 nanomolar, about 400 nanomolar, about 300 nanomolar, about 200 nanomolar, about 100 nanomolar, about 50 nanomolar, about 10 nanomolar, of less than about 10 nanomolar, or a range between and including any two of these values. A “disease responsive to inhibition of fascin activity” is a disease in which inhibiting fascin provides a therapeutic benefit such as an amelioration of symptoms, decrease in disease progression, prevention or delay of disease onset, prevention or amelioration of an inflammatory response, or inhibition of aberrant activity and/or death of certain cell-types (such as cancer cells). “Treatment” or “treating” means any treatment of a disease in a patient, including:a) preventing the disease, that is, causing the clinical symptoms of the disease not to develop;b) inhibiting the progression of the disease;c) slowing or arresting the development of clinical symptoms; and/ord) relieving the disease, that is, causing the regression of clinical symptoms. “Subject” or “patient’ refers to an animal, such as a mammal, that has been or will be the object of treatment, observation or experiment. The methods described herein may be useful in both human therapy and veterinary applications. In some embodiments, the subject is a mammal; and in some embodiments the subject is human. As used herein, the term “cancer” includes solid mammalian tumors as well as hematological malignancies. The terms “tumor cell(s)” and “cancer cell(s)” are used interchangeably herein. “Solid mammalian tumors” include cancers of the head and neck, lung, mesothelioma, mediastinum, esophagus, stomach, pancreas, hepatobiliary system, small intestine, colon, colorectal, rectum, anus, kidney, urethra, bladder, prostate, urethra, penis, testis, gynecological organs, ovaries, breast, endocrine system, skin, central nervous system; sarcomas of the soft tissue and bone; and melanoma of cutaneous and intraocular origin. The term “hematological malignancies” includes childhood leukemia and lymphomas, Hodgkin's disease, lymphomas of lymphocytic and cutaneous origin, acute and chronic leukemia, plasma cell neoplasm and cancers associated with AIDS. Also, in these examples and elsewhere, abbreviations have the following meanings:° C.=degree CelsiusμL=microliterμM=micromolarDDT=dithiothreitolDMSO=dimethyl sulfoxideg=gramkg=kilogramhr or h=hourL=literM=molarnM=nanomolarmg=milligramMHz=mega Hertzmin=minutemL=millilitermm=millimetermM=millimolarmmol=millimolemol=molePMSF=phenylmethylsulfonyl fluorideN=normalEDTA=ethylenediaminetetraacetic acidμm=micrometerr.p.m=round per minuteS.D.=standard deviationv/v=volume/volumewt=weight Compounds The present technology provides compounds of Formula I, Ia or Ib: or tautomer thereof, and/or a pharmaceutically acceptable salt thereof, whereinA1, A2, A3, A4, A5and A6are independently CH, CR3or N, provided that no more than four of A1, A2, A3, A4, A5and A6are N;R1is phenyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein the phenyl, 5-membered heteroaryl or 6-membered heteroaryl is optionally substituted with 1 to 3 R6;L2is selected from the group consisting of —NR8—, —C(O)NR8—, —NR8C(O)—, —C(O)CR82—, —CR82C(O)—, —NR8CR82—, and —CR82NR8—;R2is hydrogen, lower alkyl, 6- to 10-membered aryl or 5- to 10-membered heteroaryl; wherein the 6- to 10-membered aryl or 5- to 10-membered heteroaryl is optionally substituted with 1 to 4 R4, wherein each R4is independently selected from the group consisting of lower alkyl, lower haloalkyl, phenyl (optionally substituted with lower alkyl, halo, lower haloalkyl, or —OH), —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;each R3is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;m is 0, 1, 2 or 3;q is 1, 2 or 3;each R6is independently selected from the group consisting of cyano, halo, lower alkyl (such as methyl or ethyl), lower haloalkyl, and —CH2OH;R7is lower alkyl (such as methyl or ethyl) or lower haloalkyl;R8is hydrogen or lower alkyl (such as methyl or ethyl);each R10is independently hydrogen or lower alkyl (such as methyl or ethyl), or two R10together with the atom(s) attached thereto form a 4- to 6-membered ring; andR11is hydrogen or R3;provided that the compound is not N-(1-(4-(trifluoromethyl)benzyl)-1H-indazol-3-yl)furan-2-carboxamide. The present technology provides compounds of Formula I, Ia or Ib: or tautomer thereof, and/or a pharmaceutically acceptable salt thereof, whereinA1, A2, A3, A4, A5and A6are independently CH, CR3or N, provided that no more than four of A1, A2, A3, A4, A5and A6are N;R1is phenyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein the phenyl, 5-membered heteroaryl or 6-membered heteroaryl is optionally substituted with 1 to 3 R6;L2is selected from the group consisting of —C(O)NR8—, —NR8C(O)—, —C(O)CR82—, —CR82C(O)—, —NR8CR82—, and —CR82NR8—;R2is 6- to 10-membered aryl or 5- to 10-membered heteroaryl; wherein the 6- to 10-membered aryl or 5- to 10-membered heteroaryl is optionally substituted with 1 to 4 R4, wherein each R4is independently selected from the group consisting of lower alkyl, lower haloalkyl, phenyl (optionally substituted with lower alkyl, halo, lower haloalkyl, or —OH), —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;each R3is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;m is 0, 1, 2 or 3;q is 1, 2 or 3;each R6is independently selected from the group consisting of cyano, halo, lower alkyl (such as methyl or ethyl), lower haloalkyl, and —CH2OH;R7is lower alkyl (such as methyl or ethyl) or lower haloalkyl;R8is hydrogen or lower alkyl (such as methyl or ethyl);each R10is independently hydrogen or lower alkyl (such as methyl or ethyl), or two R10together with the atom(s) attached thereto form a 4- to 6-membered ring; andR11is hydrogen or R3;provided that the compound is not N-(1-(4-(trifluoromethyl)benzyl)-1H-indazol-3-yl)furan-2-carboxamide. In some embodiments, R8is hydrogen. In some embodiments, q is 1. In some embodiments, provided is a compound of Formula II or tautomer thereof, and/or a pharmaceutically acceptable salt thereof, whereinR1is phenyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein the phenyl, 5-membered heteroaryl or 6-membered heteroaryl is optionally substituted with 1 to 3 R6;L2is selected from the group consisting of —C(O)NH—, —NHC(O)—, —C(O)CH2—, —CH2C(O)—, —NHCH2—, and —CH2NH—;R2is 6- to 10-membered aryl or 5- to 10-membered heteroaryl; wherein the 6- to 10-membered aryl or 5- to 10-membered heteroaryl is optionally substituted with 1 to 4 R4, wherein each R4is independently selected from the group consisting of lower alkyl, lower haloalkyl, phenyl (optionally substituted with lower alkyl, halo or lower haloalkyl, or —OH), —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7; each R3is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR10, —NR10CO2R10, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;m is 0, 1, 2 or 3;n is 0, 1, 2 or 3;q is 1, 2 or 3;each R6is independently selected from the group consisting of halo, cyano, lower alkyl (preferably methyl or ethyl) and lower haloalkyl;R7is lower alkyl (preferably methyl or ethyl) or lower haloalkyl; andeach R10is independently hydrogen or lower alkyl (preferably methyl or ethyl), or two R10together with the atom(s) attached thereto form a 4- to 6-membered ring;provided that the compound is not N-(1-(4-(trifluoromethyl)benzyl)-1H-indazol-3-yl)furan-2-carboxamide. In some embodiments, provided is a compound of Formula IIIa, IIIb, IIIc or IIId or tautomer thereof, and/or a pharmaceutically acceptable salt thereof; whereinR2is 6- to 10-membered aryl or 5- to 10-membered heteroaryl; wherein the 6- to 10-membered aryl or 5- to 10-membered heteroaryl is optionally substituted with 1 to 4 R4, wherein each R4is independently selected from the group consisting of lower alkyl, lower haloalkyl, phenyl (optionally substituted with lower alkyl, halo or lower haloalkyl, or —OH), —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR1OR1O, and —NR10SO2R7;each R3is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;m is 0, 1, 2 or 3;n is 0, 1, 2 or 3;each R6is independently selected from the group consisting of halo, cyano, lower alkyl (preferably methyl or ethyl) and lower haloalkyl;R7is lower alkyl (preferably methyl or ethyl); andeach R10is independently hydrogen or lower alkyl (preferably methyl or ethyl), or two R10together with the atom(s) attached thereto form a 4- to 6-membered ring;provided that the compound is not N-(1-(4-(trifluoromethyl)benzyl)-1H-indazol-3-yl)furan-2-carboxamide. In some embodiments, provided is a compound of Formula IVa, IVb, IVc or IVd: or tautomer thereof, and/or a pharmaceutically acceptable salt thereof, whereinR2is 6- to 10-membered aryl or 5- to 10-membered heteroaryl; wherein the 6- to 10-membered aryl or 5- to 10-membered heteroaryl is optionally substituted with 1 to 4 R4, wherein each R4is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR10, —NR10CO2R10, —SOR7, —SO2R7, —SO2NR10R10, phenyl (optionally substituted with lower alkyl, halo or lower haloalkyl, or —OH), and —NR10SO2R7;each R3is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR10, —NR10CO2R0, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;m is 0, 1, 2 or 3;R7is lower alkyl (preferably methyl or ethyl); andeach R10is independently hydrogen or lower alkyl (preferably methyl or ethyl), or two R10together with the atom(s) attached thereto form a 4- to 6-membered ring;provided that the compound is not N-(1-(4-(trifluoromethyl)benzyl)-1H-indazol-3-yl)furan-2-carboxamide. In another aspect, the present technology provides intermediate compounds for the preparation of compounds of Formula I or tautomer thereof, and/or a pharmaceutically acceptable salt thereof. In some embodiments, the intermediate compounds are of any of the following Formulas whereinA1, A2, A3, A4, A5and A6are independently CH, CR3or N, provided that no more than four of A1, A2, A3, A4, A5and A6are N;A7is NH or CH2;Y is F, Cl, Br or I;R1is phenyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein the phenyl, 5-membered heteroaryl or 6-membered heteroaryl is optionally substituted with 1 to 3 R6;R2is 6- to 10-membered aryl or 5- to 10-membered heteroaryl; wherein the 6- to 10-membered aryl or 5- to 10-membered heteroaryl is optionally substituted with 1 to 4 R4, wherein each R4is independently selected from the group consisting of lower alkyl, lower haloalkyl, phenyl (optionally substituted with lower alkyl, halo, lower haloalkyl, or —OH), —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;each R3is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7;m is 0, 1, 2 or 3;q is 1, 2 or 3;each R6is independently selected from the group consisting of cyano, halo, lower alkyl (such as methyl or ethyl) and lower haloalkyl;R7is lower alkyl (such as methyl or ethyl) or lower haloalkyl;R11is hydrogen or R3; andeach R10is independently hydrogen or lower alkyl (such as methyl or ethyl), or two R10together with the atom(s) attached thereto form a 4- to 6-membered ring. The present technology provides compounds of Formula VIII, VIIIa or VIIIb: or tautomer thereof, and/or a pharmaceutically acceptable salt thereof; whereinL2is selected from the group consisting of —NR8—, —C(O)NR8—, —NR8C(O)—, —C(O)CR82—, —CR82C(O)—, —NR8CR82—, and —CR82NR8—;R2ais hydrogen, or —NHC(O)R2, wherein R2is lower alkyl, 6-membered aryl or 5- to 10-membered heteroaryl; wherein the 6- to 10-membered aryl or 5- to 10-membered heteroaryl is optionally substituted with 1 to 4 R4, wherein each R4is independently selected from the group consisting of lower alkyl, lower haloalkyl, phenyl (optionally substituted with lower alkyl, halo, lower haloalkyl, or —OH), —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7; andeach R3is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR7, —SH, —SR7, —NR10R10, halo, cyano, nitro, —COH, —COR7, —CO2H, —CO2R7, —CONR10R10, —OCOR7, —OCO2R7, —OCONR10R10, —NR10COR7, —NR10CO2R7, —SOR7, —SO2R7, —SO2NR10R10, and —NR10SO2R7. In some embodiments, L2is —C(O)NH—, —C(O)CH2—, or —CH2NH—. In some embodiments, A1is N and A2, A3, A4, A5and A6are independently CH or CR3. In some embodiments, A2is N and A1, A3, A4, A5and A6are independently CH or CR3. In some embodiments, A3is N and A1, A2, A4, A5and A6are independently CH or CR3. In some embodiments, A4is N and A1, A2, A3, A5and A6are independently CH or CR3. In some embodiments, A5is N and A1, A2, A3, A4, and A6are independently CH or CR3. In some embodiments, A6is N and A1, A2, A3, A4, and A5are independently CH or CR3. In some embodiments, A1and A2are N. In some embodiments, A3, A4, A5and A6are independently CH or CR3. In some embodiments, A3is N, and A4, A5and A6are independently CH or CR3. In some embodiments, A4is N and A3, A5and A6are independently CH or CR3. In some embodiments, A5is N, and A3, A4and A6are independently CH or CR3. In some embodiments, A6is N, and A3, A4, and A5are independently CH or CR3. In some embodiments, A3and A4are N, and A5and A6are independently CH or CR3. In some embodiments, A3and A5are N, and A4and A6are independently CH or CR3. In some embodiments, A3and A6are N, and A4and A5are independently CH or CR3. In some embodiments, A4and A5are N, and A3and A6are independently CH or CR3. In some embodiments, A4and A6are N, and A3and A5are independently CH or CR3. In some embodiments, A5and A6are N, and A3and A4are independently CH or CR3. In some embodiments, A1and A3are N. In some embodiments, A2, A4, A5and A6are independently CH or CR3. In some embodiments, A4is N, and A2, A5and A6are independently CH or CR3. In some embodiments, A5is N and A2, A4and A6are independently CH or CR3. In some embodiments, A6is N, and A2, A4and A5are independently CH or CR3. In some embodiments, A2and A4are N, and A5and A6are independently CH or CR3. In some embodiments, A2and A5are N, and A4and A6are independently CH or CR3. In some embodiments, A2and A6are N, and A4and A5are independently CH or CR3. In some embodiments, A4and A5are N, and A2and A6are independently CH or CR3. In some embodiments, A4and A6are N, and A2and A5are independently CH or CR3. In some embodiments, A5and A6are N, and A2and A4are independently CH or CR3. In some embodiments, A1and A4are N. In some embodiments, A2, A3, A5and A6are independently CH or CR3. In some embodiments, A3is N, and A2, A5and A6are independently CH or CR3. In some embodiments, A5is N and A2, A3and A6are independently CH or CR3. In some embodiments, A6is N, and A2, A3and A5are independently CH or CR3. In some embodiments, A2and A3are N, and A5and A6are independently CH or CR3. In some embodiments, A2and A5are N, and A3and A6are independently CH or CR3. In some embodiments, A2and A6are N, and A3and A5are independently CH or CR3. In some embodiments, A3and A5are N, and A2and A6are independently CH or CR3. In some embodiments, A3and A6are N, and A2and A5are independently CH or CR3. In some embodiments, A5and A6are N, and A2and A3are independently CH or CR3. In some embodiments, A1and A5are N. In some embodiments, A2, A4, A3and A6are independently CH or CR3. In some embodiments, A4is N, and A2, A3and A6are independently CH or CR3. In some embodiments, A3is N and A2, A4and A6are independently CH or CR3. In some embodiments, A6is N, and A2, A4and A3are independently CH or CR3. In some embodiments, A2and A4are N, and A3and A6are independently CH or CR3. In some embodiments, A2and A3are N, and A4and A6are independently CH or CR3. In some embodiments, A2and A6are N, and A4and A3are independently CH or CR3. In some embodiments, A4and A3are N, and A2and A6are independently CH or CR3. In some embodiments, A4and A6are N, and A2and A3are independently CH or CR3. In some embodiments, A3and A6are N, and A2and A4are independently CH or CR3. In some embodiments, A1and A6are N. In some embodiments, A2, A4, A5and A3are independently CH or CR3. In some embodiments, A4is N, and A2, A5and A3are independently CH or CR3. In some embodiments, A5is N and A2, A4and A3are independently CH or CR3. In some embodiments, A3is N, and A2, A4and A5are independently CH or CR3. In some embodiments, A2and A4are N, and A5and A3are independently CH or CR3. In some embodiments, A2and A5are N, and A4and A3are independently CH or CR3. In some embodiments, A2and A3are N, and A4and A5are independently CH or CR3. In some embodiments, A4and A5are N, and A2and A3are independently CH or CR3. In some embodiments, A4and A3are N, and A2and A5are independently CH or CR3. In some embodiments, A5and A3are N, and A2and A4are independently CH or CR3. In some embodiments, A2is N. In some embodiments, A1is CH or CR3. In some embodiments, A3, A4, A5and A6are independently CH or CR3. In some embodiments, A3is N, and A4, A5and A6are independently CH or CR3. In some embodiments, A4is N and A3, A5and A6are independently CH or CR3. In some embodiments, A5is N, and A3, A4and A6are independently CH or CR3. In some embodiments, A6is N, and A3, A4, and A5are independently CH or CR3. In some embodiments, A3and A4are N, and A5and A6are independently CH or CR3. In some embodiments, A3and A5are N, and A4and A6are independently CH or CR3. In some embodiments, A3and A6are N, and A4and A5are independently CH or CR3. In some embodiments, A4and A5are N, and A3and A6are independently CH or CR3. In some embodiments, A4and A6are N, and A3and A5are independently CH or CR3. In some embodiments, A5and A6are N, and A3and A4are independently CH or CR3. In some embodiments, R1is phenyl. In some embodiments, R1is trifluoromethylphenyl. In some embodiments, R1is 4-trifluoromethylphenyl. In some embodiments, R1is 4-fluorophenyl. In some embodiments, R1is 4-chlorophenyl. In some embodiments, R1is 4-methylphenyl. In some embodiments, R1is pyridyl optionally substituted with 1 to 3 R6. In some embodiments, R2is phenyl optionally substituted with 1 to 4 R4. In some embodiments, R2is 5-membered heteroaryl optionally substituted with 1 to 4 R4. In some embodiments, R2is 6-membered heteroaryl optionally substituted with 1 to 4 R4. In some embodiments, R2is phenyl substituted with 2 R4. In some embodiments, R2is 5-membered heteroaryl substituted with 2 R4. In some embodiments, R2is 6-membered heteroaryl substituted with 2 R4. In some embodiments, R2is phenyl substituted with 1 R4. In some embodiments, R2is 5-membered heteroaryl substituted with 1 R4. In some embodiments, R2is 6-membered heteroaryl substituted with 1 R4. In some embodiments, R2is phenyl, chlorophenyl, methyl furan, In some embodiments, R2is selected from the group consisting of thiophene, thiazole, isoxazole, oxazole, 1,2,5-oxadiazole, pyrazole, pyrimidine and pyridazine, which are optionally substituted with methyl. In some embodiments, R2is pyridazine, isoxazole or oxazole. In some embodiments, R2is 5- or 6-membered heteroaryl optionally substituted with 1 to 4 R4, wherein the heteroaryl comprises two heteroatoms selected from N, O and S. In some embodiments, R2is 5- or 6-membered heteroaryl optionally substituted with 1 to 4 R4, wherein the heteroaryl comprises two heteroatoms selected from N and S. In some embodiments, R2is phenyl. In some embodiments, R2is selected from the group consisting of: In some embodiments, R2is In some embodiments, R2is In some embodiments, R2is In some embodiments of Formula VIIIa, VIIIb or VIIIc, R2is ethyl or isopropyl. In some embodiments of Formula VIIIa, VIIIb or VIIIc, R2is In some embodiments, R2is R5optionally substituted with 1 to 4 R4, wherein R5is selected from the group consisting of furan, benzofuran, pyridine, pyridazine, pyrimidine, pyrazine, thiophene, thiazole, isothiazole, oxazole, isoxazole, oxadiazole, imidazole, pyrrole, and pyrazole. In some embodiments, R2is R5substituted with 1 R4. In some embodiments, R2is R5substituted with 2 R4. In some embodiments, R2is R5substituted with 3 R4. In some embodiments, R2is R5substituted with 4 R4. In some embodiments, R4is selected from the group consisting of lower alkyl (such as methyl), halo, lower haloalkyl, —OH, —OR7, cyano and phenyl optionally substituted methyl, wherein R7is lower alkyl or lower haloalkyl. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments, R3is halo. In some embodiments, R3is lower alkyl. In some embodiments, n is 1. In some embodiments, R6is trifluoromethyl. In some embodiments, R6is fluoro. In some embodiments, R6is chloro. In some embodiments, R6is methyl. In some embodiments, R6is cyano. In some embodiments, R6is 4-trifluoromethyl. In some embodiments, R6is 4-fluoro. In some embodiments, R6is 4-chloro. In some embodiments, R6is 4-methyl. In some embodiments, R6is 4-cyano. In some embodiments, the compound is selected from or a tautomer, and/or pharmaceutically acceptable salt thereof. In some embodiments, the group in any of the above compounds is replaced with In some embodiments, the compound is selected from or a tautomer, and/or pharmaceutically acceptable salt thereof. In some embodiments, the group in any of the above compounds is replaced with In some embodiments, the present technology provides a compound selected from Table 2 or a tautomer, and/or pharmaceutically acceptable salt thereof: TABLE 2CompoundStructureName4N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)benzamide52-chloro-N-(1-(4- (trifluoromethyl)benzyl)-1H- indazol-3-yl)benzamide9N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)thiophene-2- carboxamide104-methyl-N-(1-(4- (trifluoromethyl)benzyl)-1H- indazol-3-yl)thiazole-5- carboxamide25N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)isoxazole-5- carboxamide284-methyl-N-(1-(4- (trifluoromethyl)benzyl)-1H- indazol-3-yl)isoxazole-5- carboxamide315-methyl-N-(1-(4- (trifluoromethyl)benzyl)-1H- indazol-3-yl)isoxazole-4- carboxamide33N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)-1,2,5-oxadiazole- 3-carboxamide344-methyl-N-(1-(4- (trifluoromethyl)benzyl)-1H- indazol-3-yl)oxazole-5- carboxamide353-methyl-N-(1-(4- (trifluoromethyl)benzyl)-1H- indazol-3-yl)isoxazole-4- carboxamide36N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)thiazole-5- carboxamide39N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)isothiazole-3- carboxamide40N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)oxazole-5- carboxamide432-methyl-N-(1-(4- (trifluoromethyl)benzyl)-1H- indazol-3-yl)furan-3-carboxamide443-methyl-N-(1-(4- (trifluoromethyl)benzyl)-1H- indazol-3-yl)-1H-pyrazole-4- carboxamide49N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)pyridazine-3- carboxamide56N-(1-(4-(trifluoromethyl)benzyl)- 1H-indazol-3-yl)pyrimidine-5- carboxamide64N-(1-(4-chlorobenzyl)-1H-indazol- 3-yl)furan-3-carboxamide65N-(1-(4-chlorobenzyl)-1H-indazol- 3-yl)-3-methylisoxazole-4- carboxamide66N-(1-(4-chlorobenzyl)-1H-indazol- 3-yl)isoxazole-5-carboxamide67N-(1-(4-chlorobenzyl)-1H-indazol- 3-yl)-2-methylfuran-3- carboxamide68N-(1-(4-chlorobenzyl)-1H-indazol- 3-yl)-4-methylthiazole-5- carboxamide69N-(1-(4-chlorobenzyl)-1H-indazol- 3-yl)pyridazine-3-carboxamide73N-(1-(4-fluorobenzyl)-1H-indazol- 3-yl)isoxazole-5-carboxamide80N-(1-(4-methylbenzyl)-1H- indazol-3-yl)isoxazole-5- carboxamide1374-chloro-1-(4- (trifluoromethyl)benzyl)-1H- pyrazolo[3,4-c]pyridin-3-amine138N-(4-chloro-1-(4- (trifluoromethyl)benzyl)-1H- pyrazolo[3,4-c]pyridin-3-yl)-4- methylthiazole-5-carboxamide139N-(4-chloro-1-(4- (trifluoromethyl)benzyl)-1H- pyrazolo[3,4-c]pyridin-3- yl)pyridazine-3-carboxamide151N-(4-chloro-1-(4- (trifluoromethyl)benzyl)-1H- pyrazolo[3,4-c]pyridin-3- yl)propionamide152N-(4-chloro-1-(4- (trifluoromethyl)benzyl)-1H- pyrazolo[3,4-c]pyridin-3- yl)isobutyramide In one aspect, the compound is not a compound of Formula I-j: or a tautomer thereof, and/or a pharmaceutically acceptable salt thereof, whereinR21is phenyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein the phenyl, 5-membered heteroaryl or 6-membered heteroaryl is optionally substituted with 1 to 3 R6;L11is selected from the group consisting —(C(R18)2)j—, —(C(R18)2)q—C(O)—(C(R18)2)r—, —(C(R18)2)q—C(O)N(R18)—(C(R18)2)r—, —(C(R18)2)q—N(R18)C(O)—(C(R8)2)r—, —(C(R18)2)q—N(R18)S(O)2—(C(R18)2)r—, —(CH2)q—S(O)2N(R18)—(CH2)r, —S—, —O— and —NR18—;q is 0 or 1;r is 0 or 1;L12is selected from the group consisting a covalent bond, —C(O)N(R18)—, —N(R18)C(O)—, —N(R18)S(O)2—, and —S(O)2N(R18)—;R15is phenyl, 5-membered heteroaryl, 6-membered heteroaryl, 5-membered heterocycloalkyl or 6-membered heterocycloalkyl; wherein the phenyl, 5-membered heteroaryl or 6-membered heteroaryl is optionally substituted with 1 to 4 R12, wherein each R12is independently selected from the group consisting of lower alkyl, lower haloalkyl, —OH, —OR17, —SH, —SR17, —NR20R20, halo, cyano, nitro, —COH, —COR17, —CO2H, —CO2R17, —CONR20R20, —OCOR17, —OCO2R17, —OCONR20R20, —NR20COR20, —NR20CO2R20, —SOR17, —SO2R17, —SO2NR20R20, and —NR20SO2R17;each R16is independently selected from the group consisting of halo and lower alkyl (preferably methyl or ethyl) optionally substituted with 1-3 halo; or two adjacent R16on a phenyl ring form a 5- or 6-membered cycloalkyl or heterocycloalkyl fused with the phenyl ring;R17is lower alkyl (preferably methyl or ethyl);R18is hydrogen or lower alkyl (preferably methyl or ethyl); andeach R20is independently hydrogen or lower alkyl (preferably methyl or ethyl), or two R20together with the atom(s) attached thereto form a 4- to 6-membered heterocycloalkyl ring. In one aspect, the compound is not a compound of Formula I-j: or a tautomer thereof, and/or a pharmaceutically acceptable salt thereof, whereinR21is phenyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein the phenyl, 5-membered heteroaryl or 6-membered heteroaryl is optionally substituted with 1 to 3 R6;L11is selected from the group consisting —C(R18)2—, —S—, —O— and —NR18—;L12is selected from the group consisting a covalent bond, —C(O)N(R18)—, —N(R18)C(O)—, —N(R18)S(O)2—, and —S(O)2N(R18)—;R15is phenyl, 5-membered heteroaryl, 6-membered heteroaryl, 5-membered heterocycloalkyl or 6-membered heterocycloalkyl; wherein the phenyl, 5-membered heteroaryl or 6-membered heteroaryl is optionally substituted with 1 to 4 R12, wherein each R12is independently selected from the group consisting of lower alkyl, lower haloalkyl, OH, OR17, SH, SR17, NR20R20, halo, cyano, nitro, COH, COR17, CO2H, —CO2R17, CONR20R20, OCOR17, OCO2R17, OCONR20R20, NR20COR20, —NR20CO2R20, —SOR17, SO2R17, —SO2NR20R20, and NR20SO2R17;each R16is independently selected from the group consisting of halo and lower alkyl (preferably methyl or ethyl) optionally substituted with 1-3 halo;R17is lower alkyl (preferably methyl or ethyl);R18is hydrogen or lower alkyl (preferably methyl or ethyl); andeach R20is independently hydrogen or lower alkyl (preferably methyl or ethyl), or two R20together with the atom(s) attached thereto form a 4- to 6-membered ring. Methods of Treatment In one aspect, the present technology provides a method of treating a condition or disorder mediated by fascin activity in a subject in need thereof which method comprises administering to the subject a therapeutically effective amount of a compound described herein. In one embodiment the present technology provides a method of inhibiting fascin activity, comprising administering an effective amount of a fascin inhibitor to a cell to thereby inhibit fascin activity in the cell, wherein the fascin inhibitor is a compound described herein. In some embodiments, the fascin inhibitor has a fascin inhibition IC50of no more than 100 μM. In some embodiments, the fascin inhibitor has a fascin inhibition IC50of no more than 50 μM. In some embodiments, the fascin inhibitor has a fascin inhibition IC50of no more than 20 μM. In some embodiments, the fascin inhibitor has a fascin inhibition IC50of no more than 8 μM. In some embodiments, the condition or disorder is a metastatic cancer, a neuronal disorder, neuronal degeneration, an inflammatory condition, a viral infection, a bacterial infection, lymphoid hyperplasia, Hodgkin's disease or ischemia-related tissue damage. In some embodiments, the condition or disorder is a metastatic cancer. In some embodiments, the cancer is a carcinoma, lymphoma, sarcoma, melanoma, astrocytoma, mesothelioma cells, ovarian carcinoma, colon carcinoma, pancreatic carcinoma, esophageal carcinoma, stomach carcinoma, lung carcinoma, urinary carcinoma, bladder carcinoma, breast cancer, gastric cancer, leukemia, lung cancer, colon cancer, central nervous system cancer, melanoma, ovarian cancer, renal cancer or prostate cancer. In some embodiments, the cancer is lung cancer, breast cancer or prostate cancer. In another aspect, the present technology provides is a method of inhibiting fascin activity, comprising administering an effective amount of a fascin inhibitor to a cell to thereby inhibit fascin activity in the cell, wherein the fascin inhibitor is a compound described herein. In some embodiments, the cell is in an animal. In some embodiments, the cell has been removed from an animal. In some embodiments, the animal is a human. In some embodiments, the human suffers from a disease or condition. In some embodiments, the condition or disorder is a metastatic cancer, a neuronal disorder, neuronal degeneration, an inflammatory condition, a viral infection, a bacterial infection, lymphoid hyperplasia, Hodgkin's disease or ischemia-related tissue damage. In some embodiments, the condition or disorder is a metastatic cancer. In some embodiments, the cancer is a carcinoma, lymphoma, sarcoma, melanoma, astrocytoma, mesothelioma cells, ovarian carcinoma, colon carcinoma, pancreatic carcinoma, esophageal carcinoma, stomach carcinoma, lung carcinoma, urinary carcinoma, bladder carcinoma, breast cancer, gastric cancer, leukemia, lung cancer, colon cancer, central nervous system cancer, melanoma, ovarian cancer, renal cancer or prostate cancer. In some embodiments, the cancer is lung cancer, breast cancer, or prostate cancer. Agents that modulate the activity of fascin can be used to treat a variety of diseases and conditions. For example, as illustrated herein, fascin promotes actin bundling and plays a key role in cell migration and metastasis of cancer cells. Hence, modulators and inhibitors of fascin can be used to treat and inhibit metastatic cancer. However, fascin also plays a role in other diseases and conditions. For example, neurite shape and trajectory is modulated by fascin (Kraft et al., Phenotypes of Drosophila brain neurons in primary culture reveal a role for fascin in neurite shape and trajectory, J. Neurosci, 26(34):8734-47 (2006)). Fascin is also involved in neuronal degeneration (Fulga et al., Abnormal bundling and accumulation of F-actin mediates tau-induced neuronal degeneration in vivo Nat Cell Biol. 9(2):139-48 (2007)). In addition, fascin plays a role in Hodgkin's disease (Pinkus et al., Fascin, a sensitive new marker for Reed-Sternberg cells of Hodgkin's disease, Am J Pathol. 150(2):543-562 (1997)). Fascin also plays a role in processing and presenting antigens, for example, on antigen presenting cells (Mosialos et al., Circulating human dendritic cells differentially express high levels of a 55-kd actin-bundling protein. Am. J. Pathol. 148(2):593-600 (1996); Said et al. The role of follicular and inter digitating dendritic cells in HIV-related lymphoid hyperplasia: localization of fascin. Mod Pathol. 10(5):421-27 (1997)). Moreover, fascin also plays a role in ischemic injury (Meller et al., Ubiquitin proteasome-mediated synaptic reorganization: a novel mechanism underlying rapid ischemic tolerance, J Neurosci. 28(1):50-9 (2008)). Provided herein are agents that modulate fascin activity and that can be used for methods of treating and inhibiting metastatic cancer, neuronal disorders, neuronal degeneration, inflammatory conditions, viral infections, bacterial infections, lymphoid hyperplasia, Hodgkin's disease, and ischemia-related tissue damage. Tumor metastasis is the major cause of death of cancer patients (Weiss 2000, Fidler 2003). Thus, inhibition or prevention of tumor metastasis will significantly increase the survival rate of cancer patients, allow more moderate radiation or chemotherapy with less side-effects, and control the progression of solid tumors. Tumor cell migration and invasion are critical steps in the process of tumor metastasis (Partin et al. 1989, Aznavoorian et al. 1993, Condeelis et al. 2005). For cell migration to proceed, the actin cytoskeleton must be reorganized by forming polymers and bundles to affect the dynamic changes of cell shapes (Jaffe et al. 2005, Matsudaira 1994, Otto 1994). Individual actin filaments are flexible and elongation of individual filaments per se is insufficient for membrane protrusion which is necessary for cell migration. Bundling of actin filaments provides rigidity to actin filaments for protrusion against the compressive force from the plasma membrane (Mogilner et al. 2005). One of the critical actin-bundling proteins is fascin. Fascin is the primary actin cross-linker in filopodia, which are membrane protrusions critical for the migration and metastasis of cancer cells. Fascin is required to maximally cross-link the actin filaments into straight, compact, and rigid bundles. Elevated expressions of fascin mRNA and protein in cancer cells have been correlated with aggressive clinical course, poor prognosis and shorter survival. Accordingly, metastatic cancer can be treated, prevented and/or inhibited by administering fascin inhibitors as described herein. In addition, a cancer at any stage of progression can be treated by the method of the present technology, such as primary, metastatic, and recurrent cancers. In some embodiments, cancers are treated before metastasis is detected, for example, to inhibit metastatic cancer from developing. In other embodiments, cancers are treated when metastasis is detected, for example, to inhibit further metastasis and progression of the cancer. Compounds described herein, or pharmaceutically acceptable salts thereof, can also be used to treat autoimmune deficiency syndrome-associated Kaposi's sarcoma, cancer of the adrenal cortex, cancer of the cervix, cancer of the endometrium, cancer of the esophagus, cancer of the head and neck, cancer of the liver, cancer of the pancreas, cancer of the prostate, cancer of the thymus, carcinoid tumors, chronic lymphocytic leukemia, Ewing's sarcoma, gestational trophoblastic tumors, hepatoblastoma, multiple myeloma, non-small cell lung cancer, retinoblastoma, or tumors in the ovaries. A cancer at any stage of progression can be treated or detected, such as primary, metastatic, and recurrent cancers. Information regarding numerous types of cancer can be found, e.g., from the American Cancer Society (www.cancer.org), or from, e.g., Wilson et al. (1991) Harrison's Principles of Internal Medicine, 12th Edition, McGraw-Hill, Inc. In some embodiments, method are provided for treating or inhibiting metastatic cancer in an animal, for example, for human and veterinary uses, which include administering to a subject animal (e.g., a human), a therapeutically effective amount of a compound described herein, or pharmaceutically acceptable salt thereof. In some embodiments, the cell has been removed from an animal. Treatment of, or treating, a disease or condition (e.g., cancer) is intended to include the alleviation of or diminishment of at least one symptom typically associated with the disease or condition. The treatment also includes alleviation or diminishment of more than one symptom of the disease or condition. The treatment may cure the disease or condition, for example, by eliminating the symptoms and/or the source of the disease or condition. For example, treatment can cure the cancer by substantially inhibiting metastasis of the cancer cells so that removal or killing of the primary tumor or cancer cell(s) substantially eliminates the cancer. Treatment can also arrest or inhibit the metastasis of the cancer and/or tumor cells without directly killing or promoting the apoptosis of cancer cells. Fascin functions in a variety of cellular functions that play critical roles in modulating the growth, movement and interaction of cells. However the actin bundling function of fascin is directly involved in tumor metastasis and invasive growth. The anti-metastatic activity of fascin (e.g., in the presence of various test agents or therapeutic agents like those described herein) can be evaluated against varieties of cancers using methods described herein and available to one of skill in the art. Anti-cancer activity, for example, can be determined by identifying the dose that inhibits 50% cancer cell metastasis (IC50) of a compound or composition as described herein. Also provided is a method for evaluating a therapeutically effective dosage for treating a cancer (e.g., inhibiting metastasis) with a compound described herein, or pharmaceutically acceptable salt thereof, that includes determining the IC50of the agent in vitro. Such a method permits calculation of the approximate amount of agent needed per volume to inhibit cancer cell migration. Such amounts can be determined, for example, by standard microdilution methods. In some embodiments, the compound or composition as described herein can be administered in multiple doses over an extended period of time, or intermittently. Compositions The compounds (e.g., fascin inhibitors) as described herein can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical, transdermally, intrathecally, ocularly, intranasally, intraperitoneally or subcutaneous routes. The compounds (e.g., fascin inhibitors) described herein may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained. The tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like. A syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. A material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and devices. The active compounds described herein may also be administered intravenously or intraperitoneally by infusion or injection. Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. In all cases, the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions. For topical administration, the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid. Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the present compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers. Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user. Examples of useful dermatological compositions which can be used to deliver the compounds described herein, or pharmaceutically acceptable salts thereof, to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508). Useful dosages of the compounds described herein, or pharmaceutically acceptable salts thereof, can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949. Generally, the concentration of the compounds described herein, or pharmaceutically acceptable salts thereof, in a liquid composition, such as a lotion, will be about 0.01 wt %, about 0.1 wt %, about 1.0 wt %, about 2.0 wt %, about 3.0 wt %, about 4.0 wt %, about 5.0 wt %, about 10.0 wt %, about 25.0 wt %, or a range between and including any two of these values. The concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.01 wt %, about 0.1 wt %, about 1.0 wt %, about 2.0 wt %, about 3.0 wt %, about 4.0 wt %, about 5.0 wt %, about 10.0 wt %, about 25.0 wt %, or a range between and including any two of these values. The amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician. In general, however, a suitable dose will be in the range of from about 1.0 to about 200 mg/kg, e.g., from about 1 to about 100 mg/kg of body weight per day, such as about 2.0 to about 100 mg/kg of body weight per day, such as about 3.0 to about 50 mg per kilogram body weight of the recipient per day, or in the range of about 5 to 20 mg/kg/day. Alternatively, the compositions can be administered five times a week on five consecutive days with a two day rest, or four times a week on four consecutive days with a three day rest, or every other day. Methods for extrapolating effective dosages in mice and other animals, to humans are known in the art (See, for example, U.S. Pat. No. 4,938,949). For example, in some embodiments, compounds described herein, or pharmaceutically acceptable salts thereof, (for example those useful for the treatment of colon and/or ovarian cancer) may be administered at dosage levels of about 0.01 mg/kg to about 300 mg/kg, from about 0.1 mg/kg to about 250 mg/kg, from about 1 mg/kg to about 200 mg/kg, from about 1 mg/kg to about 150 mg/kg, from about 1 mg/kg to about 100 mg/kg, from about 1 mg/kg to about 90 mg/kg, from about 1 mg/kg to about 80 mg/kg, from about 1 mg/kg to about 70 mg/kg, from about 1 mg/kg to about 60 mg/kg, from about 1 mg/kg to about 50 mg/kg, from about 1 mg/kg to about 40 mg/kg, from about 1 mg/kg to about 30 mg/kg, from about 1 mg/kg to about 20 mg/kg, from about 5 mg/kg to about 100 mg/kg, from about 5 mg/kg to about 90 mg/kg, from about 5 mg/kg to about 80 mg/kg, from about 5 mg/kg to about 70 mg/kg, from about 5 mg/kg to about 60 mg/kg, from about 5 mg/kg to about 50 mg/kg, from about 5 mg/kg to about 40 mg/kg, from about 5 mg/kg to about 30 mg/kg, from about 5 mg/kg to about 20 mg/kg, from about 10 mg/kg to about 100 mg/kg, from about 10 mg/kg to about 90 mg/kg, from about 10 mg/kg to about 80 mg/kg, from about 10 mg/kg to about 70 mg/kg, from about 10 mg/kg to about 60 mg/kg, from about 10 mg/kg to about 50 mg/kg, from about 10 mg/kg to about 40 mg/kg, from about 10 mg/kg to about 30 mg/kg, from about 10 mg/kg to about 20 mg/kg, from about 20 mg/kg to about 100 mg/kg, from about 20 mg/kg to about 90 mg/kg, from about 20 mg/kg to about 80 mg/kg, from about 20 mg/kg to about 70 mg/kg, from about 20 mg/kg to about 60 mg/kg, from about 20 mg/kg to about 50 mg/kg, from about 20 mg/kg to about 40 mg/kg, from about 20 mg/kg to about 30 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. In some embodiments, compounds may be administered at a dosage of about 1 mg/kg or greater, 5 mg/kg or greater; 10 mg/kg or greater, 15 mg/kg or greater, 20 mg/kg or greater, 25 mg/kg or greater, 30 mg/kg or greater, 35 mg/kg or greater, 40 mg/kg or greater, 45 mg/kg or greater, 50 mg/kg or greater, 60 mg/kg or greater, 70 mg/kg or greater, of body weight. It will also be appreciated that dosages smaller than 0.01 mg/kg or greater than 70 mg/kg (for example 70-200 mg/kg) can be administered to a subject. In some embodiments, the compounds described herein may be used in chemotherapy (i.e., to inhibit metastasis) and may be administered at higher dosage. For example, compounds to be used in chemotherapy may be administered from about 100 mg/kg to about 300 mg/kg, from about 120 mg/kg to about 280 mg/kg, from about 140 mg/kg to about 260 mg/kg, from about 150 mg/kg to about 250 mg/kg, from about 160 mg/kg to about 240 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. In certain other embodiments, the compounds described herein may be used in supportive therapy (e.g., as an adjuvant to surgery or irradiation in a range of common types of tumor) and may be administered at lower dosage. For example, compounds to be used in supportive therapy may be administered from about 1 mg/kg to about 30 mg/kg, from about 1 mg/kg to about 25 mg/kg, from about 5 mg/kg to about 20 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. In certain other embodiments, the compounds described herein may be used for treating metastatic cancer (e.g., ovarian and/or colon cancer) and may be administered at an intermediate dosage. For example, compounds to be used in supportive therapy may be administered from about 1 mg/kg to about 100 mg/kg, from about 1 mg/kg to about 80 mg/kg, from about 5 mg/kg to about 70 mg/kg, from about 10 mg/kg to about 70 mg/kg, from about 10 mg/kg to about 60 mg/kg, from about 20 mg/kg to about 70 mg/kg, from about 20 mg/kg to about 60 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. The compound is conveniently administered in unit dosage form; for example, containing 45 to 3000 mg, conveniently 90 to 2250 mg, most conveniently, 450 to 1500 mg of active ingredient per unit dosage form. In some embodiments, the compound is administered at dosages of about 1 to about 100 mg/kg. Ideally, the active ingredient should be administered to achieve peak plasma concentrations of the active compound of from about 0.5 nM to about 10 μM, or about 1 nM to 1 μM, or about 10 nM to about 0.5 μM. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the active ingredient, optionally in saline, or orally administered as a bolus containing about 20-2000 mg of the active ingredient. Desirable blood levels may be maintained by continuous infusion to provide about 0.2 to 1.0 mg/kg/hr or by intermittent infusions containing about 0.4 to 20 mg/kg of the active ingredient(s). The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye. Compounds described herein, or pharmaceutically acceptable salts thereof, are useful as therapeutic agents administered for inhibition of cell migration and treatment of metastatic cancer. Such cancers include but are not limited to, e.g., cancers involving the animal's head, neck, lung, mesothelioma, mediastinum, esophagus, stomach, pancreas, hepatobiliary system, small intestine, colon, colorectal, rectum, anus, kidney, ureter, bladder, prostate, urethra, penis, testis, gynecological organs, ovaries, breast, endocrine system, skin, or central nervous system. Thus, for example, the cancer can be a breast cancer, a leukemia, a lung cancer, a colon cancer, a central nervous system cancer, a melanoma, an ovarian cancer, a renal cancer, or a prostate cancer. Additionally, compounds described herein, or pharmaceutically acceptable salts thereof, such as the exemplary salts described herein, may be useful as pharmacological tools for the further investigation of the inhibition of cell migration. The compounds described herein, or pharmaceutically acceptable salts thereof, can also be administered in combination with other therapeutic agents that are effective for treating or controlling the spread of cancerous cells or tumor cells. Moreover, the compounds described herein, or pharmaceutically acceptable salts thereof, can be tested in appropriate animal models. For example, the compounds described herein, or pharmaceutically acceptable salts thereof, can be tested in animals with known tumors, or animals that have been injected with tumor cells into a localized area. The degree or number of secondary tumors that form over time is a measure of metastasis and the ability of the compounds to inhibit such metastasis can be evaluated relative to control animals that have the primary tumor but receive no test compounds. The compounds described herein, or pharmaceutically acceptable salts thereof, will also find use in treatment of brain disorders (Kraft et al., J. Neurosci. 2006 Aug. 23; 26(34):8734-47); Hodgkin's disease (Pinkus et al., Am J Pathol. 1997 February; 150(2):543-62); virus infection (Mosialos et al., Am J Pathol. 1996 February; 148(2):593-600); neuronal degeneration (Fulga et al., Nat Cell Biol. 2007 February: 9(2):139-48); lymphoid hyperplasia (Said et al., Mod Pathol. 1997 May; 10(5):421-7); and ischemia (Meller et al., J Neurosci. 2008 Jan. 2; 28(1):50-9.) General Synthetic Methods The compounds described herein are commercially available or can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures. Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. Suitable protecting groups for various functional groups as well as suitable conditions for protecting and deprotecting particular functional groups are well known in the art. For example, numerous protecting groups are described in T. W. Greene and G. M. Wuts,Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999, and references cited therein. Furthermore, the compounds described herein may contain one or more chiral centers. Accordingly, if desired, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures. All such stereoisomers (and enriched mixtures) are included within the scope of this invention, unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be prepared using, for example, optically active starting materials or stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such compounds can be separated using, for example, chiral column chromatography, chiral resolving agents and the like. The starting materials for the following reactions are generally known compounds or can be prepared by known procedures or obvious modifications thereof. For example, many of the starting materials are available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wisconsin, USA), Bachem (Torrance, California, USA), Emka-Chemce or Sigma (St. Louis, Missouri, USA). Others may be prepared by procedures, or obvious modifications thereof, described in standard reference texts such as Fieser and Fieser'sReagents for Organic Synthesis, Volumes 1-15 (John Wiley and Sons, 1991), Rodd'sChemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989),Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March'sAdvanced Organic Chemistry, (John Wiley and Sons, 4thEdition), and Larock'sComprehensive Organic Transformations(VCH Publishers Inc., 1989). The various starting materials, intermediates, and compounds described herein may be isolated and purified where appropriate using conventional techniques such as precipitation, filtration, crystallization, evaporation, distillation, and chromatography. Characterization of these compounds may be performed using conventional methods such as by melting point, mass spectrum, nuclear magnetic resonance, and various other spectroscopic analyses. Amide coupling reagents are known in the art and may include, but are not limited to, aminium and phosphonium based reagents. Aminium salts include N-[(dimethylamino)-1H-1,2,3-triazolo[4,5-b]pyridine-1-ylmethylene]-N-methylmethanaminium hexafluorophosphate N-oxide (HATU), N-[(1H-benzotriazol-1-yl)(dimethylamino)methylene]-N-methylmethanaminium hexafluorophosphate N-oxide (HBTU), N-[(1H-6-chlorobenzotriazol-1-yl)(dimethylamino)methylene]-N-methylmethanaminium hexafluorophosphate N-oxide (HCTU), N-[(1H-benzotriazol-1-yl)(dimethylamino)methylene]-N-methylmethanaminium tetrafluoroborate N-oxide (TBTU), and N-[(1H-6-chlorobenzotriazol-1-yl)(dimethylamino)methylene]-N-methylmethanaminium tetrafluoroborate N-oxide (TCTU). Phosphonium salts include 7-azabenzotriazol-1-yl-N-oxy-tris(pyrrolidino)phosphonium hexafluorophosphate (PyAOP) and benzotriazol-1-yl-N-oxy-tris(pyrrolidino)phosphonium hexafluorophosphate (PyBOP). Amide formation step may be conducted in a polar solvent such as dimethylformamide (DMF) and may also include an organic base such as diisopropylethylamine (DIEA) or dimethylaminopyridine (DMAP). Cross-coupling reactions are well known in the art and, for example, are reported in Anna Roglans, et al. Diazonium Salts as Substrates in Palladium-Catalyzed Cross-Coupling Reactions, Chem. Rev., 2006, 106 (11):4622-4643; Brad M. Rosen, et al., Nickel-Catalyzed Cross-Couplings Involving Carbon-Oxygen Bonds, Percec Chem. Rev., 2011, 111 (3):1346-1416; Jean-Pierre Corbet, et al., Selected Patented Cross-Coupling Reaction Technologies, Chem. Rev., 2006, 106 (7):2651-2710; Gwilherm Evano et al., Copper-Mediated Coupling Reactions and Their Applications in Natural Products and Designed Biomolecules Synthesis, Chem. Rev., 2008, 108 (8):3054-3131; Benny Bogoslavsky, et al., Formation of a Carbon-Carbon Triple Bond by Coupling Reactions In Aqueous Solution, Science 308 (5719): 234-235 (2005); and M. Lafrance, et al., Catalytic Intermolecular Direct Arylation of Perfluorobenzenes, J. Am. Chem. Soc. 128 (27): 8754-8756 (2006); Norio Miyaura, et al., “A new stereospecific cross-coupling by the palladium-catalyzed reaction of 1-alkenylboranes with 1-alkenyl or 1-alkynyl halides,” Tetrahedron Letters, 1979, 20(36): 3437-3440; P. E. Fanta, “The Ullmann Synthesis of Biaryls”, Synthesis, 1974, 1974: 9-21; M. Gomberg, and W. E. Bachmann, J. Am. Chem. Soc., 1924, 42(10):2339-2343; R. J. P. Corriu and Masse, J. P. “Activation of Grignard reagents by transition-metal complexes. A new and simple synthesis of trans-stilbenes and polyphenyls,” Journal of the Chemical Society, Chemical Communications, 1972, (3):144a. In some aspects, compounds of Formula I can be prepared according to Scheme 1 or other methods described herein. In some aspects, compounds of Formula IIIa wherein R3is hydrogen (Compound 2-3) can be prepared from 1H-indazol-3-amine (Compound 2-1, available from e.g., Enamine LLC) according to Scheme 2 or other methods described herein. In some aspects, compounds of Formula VIIIa wherein R3is 4-chloro (Compound 3-2 or 3-3) from 4-chloro-1H-pyrazolo[3,4-c]pyridin-3-amine (Compound 3-1, available from, e.g., Novasyn Organics PVT. Ltd.) can be prepared according to Scheme 3 or other methods described herein. Compounds of formula 2-4 are generally available from commercial sources or can prepared by methods known in the art. For example, 4-(bromomethyl)benzonitrile, 3-(bromomethyl)benzonitrile, 2-fluorobenzyl bromide, 3-fluorobenzyl bromide, 3-chlorobenzyl bromide, 4-chlorobenzyl bromide, 4-fluorobenzyl bromide, 4-methylbenzyl bromide, 3,4-difluorobenzyl bromide and 2,3-difluoro-4-methylbenzyl bromide, etc., are available from Sigma-Aldrich Co. LLC. All publications, patent applications, issued patents, and other documents referred to in this specification are herein incorporated by reference as if each individual publication, patent application, issued patent, or other document was specifically and individually indicated to be incorporated by reference in its entirety. Definitions that are contained in text incorporated by reference are excluded to the extent that they contradict definitions in this disclosure. The present technology, thus generally described, will be understood more readily by reference to the following Examples, which is provided by way of illustration and is not intended to be limiting of the present technology. Other compounds were or may be prepared similarly or by methods known in the art. EXAMPLES Example 1: Compound Preparation A Preparation of Intermediate 1: 1-(4-(trifluoromethyl)benzyl)-1H-indazol-3-amine A mixture of KOH (6.95 g, 124 mmol) in DMSO (165 mL) was stirred at room temperature for 5 min. 1H-indazol-3-amine (8.25 g, 62.0 mmol) was then added in one portion. The resulting mixture was stirred at room temperature for 5 min. A solution of 4-trifluoromethylbenzyl bromide (15.6 g, 65.1 mmol) in DMSO (83 ml) was then added dropwise over 30 min. When the addition was complete, the resulting mixture was stirred at room temperature for an additional 1 h. The mixture was quenched by the addition of water (200 mL). The mixture was then extracted with CH2Cl2(3×100 mL). The combined extracts were washed with H2O (2×100 mL), brine (1×100 mL), then dried over MgSO4, filtered and concentrated in vacuo. Purification by flash chromatography (Silica, 200 g, 10-100% EtOAc/Hexanes) gave 1-(trifluoromethylbenzyl)-1H-indazol-3-amine (21.79 g, 56.6 mmol, 91.3% yield) as an off-white crystalline solid. MS (ESI) m/z: 292 (M+H)+ Preparation of Compound 1: 4,5-dimethyl-N-(1-(4-(trifluoromethyl)benzyl)-1H-indazol-3-yl)furan-2-carboxamide To a solution of intermediate 1 (29.2 mg, 0.10 mmol), 4,5-dimethylfuran-2-carboxylic acid (15.4 mg, 0.11 mmol), and triethylamine (45.2 μL, 0.30 mmol) in dichloromethane (2 mL) was added 2,4,6-tripropyl-1,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (118.6 μL, 0.20 mmol). The resulting reaction mixture was stirred at room temp for 3 h and then the solvent was removed. The crude product was purified by Prep. HPLC (sunfire 5μ 100 mm column, MeOH/H2O as solvents). 26 mg of 4,5-dimethyl-N-(1-(4-(trifluoromethyl) benzyl)-1H-indazol-3-yl)furan-2-carboxamide obtained as a solid. MS (ESI) m/z: 414 (M+H)+ Preparation of Compounds 2 to 47 and 49 to 56 in Table 1 These compounds were prepared by following procedures similar to that described above in a similar yield. B Preparation of Intermediate 2: N-(1H-indazol-3-yl)furan-3-carboxamide To a solution of 1H-indazol-3-amine (1.33 g, 10 mmol), furan-2-carboxylic acid (1.23 g, 11 mmol), and triethylamine (452 μL, 30 mmol) in dichloromethane (20 mL) was added 2,4,6-tripropyl-1,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (1.12 mL, 20 mmol). The resulting reaction mixture was stirred at room temp for 3 h and then the solvent was removed. The crude product was purified by flash chromatography (Silica, 100 g, 10-100% EtOAc/DCM) gave N-(1H-indazol-3-yl)furan-3-carboxamide (1.27 g, 5.6 mmol, 56% yield) as a white crystalline solid. MS (ESI) m/z: 228 (M+H)+ C Preparation of Compound 48: N-(1-(3-(trifluoromethyl)benzyl)-1H-indazol-3-yl)furan-3-carboxamide A mixture of KOH (11.2 mg, 0.20 mmol) in DMSO (1 mL) was stirred at room temperature for 5 min. intermediate 2 (22.7 mg, 0.10 mmol) was then added in one portion. The resulting mixture was stirred at room temperature for 5 min. A solution of 3-trifluoromethylbenzyl bromide (23.9 mg, 0.10 mmol) in DMSO (1 mL) was then added dropwise. The resulting reaction mixture was stirred at room temperature overnight. The crude product was purified by Prep. HPLC (sunfire 5p 100 mm column, MeOH/H2O as solvents). 12 mg of N-(1-(3-(trifluoromethyl)benzyl)-1H-indazol-3-yl)furan-3-carboxamide obtained as a solid. MS (ESI) m/z: 386 (M+H)+ D: Preparation of Compounds 57 and 58 Compounds 57 and 58 were prepared by following procedures similar to that described for making compound 48 in a similar yield. E: Preparation of Compound 91: 1-((6-(trifluoromethyl)pyridine-3-yl)methyl)-1H-indazol-3-amine A mixture of KOH (6.95 g, 124 mmol) in DMSO (165 mL) was stirred at room temperature for 5 min. 1H-indazol-3-amine (8.25 g, 62.0 mmol) was then added in one portion. The resulting mixture was stirred at room temperature for 5 min. A solution of 5-(chloromethyl)-2-(trifluoromethyl)pyridine (12.7 g, 65.1 mmol) in DMSO (83 mL) was then added dropwise over 30 min. When the addition was complete, the resulting mixture was stirred at room temperature for an additional 1 h. The mixture was quenched by the addition of water (200 mL). The mixture was then extracted with CH2Cl2(3×100 mL). The combined extracts were washed with H2O (2×100 mL), brine (1×100 mL), then dried over MgSO4, filtered and concentrated in vacuo. Purification by flash chromatography (Silica, 200 g, 10-100% EtOAc/Hexanes) gave 1-((6-(trifluoromethyl)pyridine-3-yl)methyl)-1H-indazol-3-amine (14.9 g, 50.9 mmol, 82% yield) as an off-white crystalline solid. MS (ESI) m/z: 293 (M+H)+. F: Preparation of Compound 92: N-(1-((6-(trifluoromethyl)pyridine-3-yl)methyl)-1H-indazol-3-yl)furan-3-carboxamide To a solution of example 91 (29.3 mg, 0.10 mmol), furan-3-carboxylic acid (15.4 mg, 0.11 mmol), and triethylamine (45.2 μl, 0.30 mmol) in Dichloromethane (2 mL) was added 2,4,6-tripropyl-1,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (118.6 μl, 0.20 mmol). The resulting reaction mixture was stirred at room temp for 3 h and then the solvent was removed. The crude product was purified by Prep. HPLC (sunfire 5 u 100 mm column, MeOH/H2O as solvents). 26 mg of N-(1-((6-(trifluoromethyl)pyridine-3-yl)methyl)-1H-indazol-3-yl)furan-3-carboxamide was obtained as a solid. MS (ESI) m/z: 387 (M+H)+. G: Preparation of Compounds 93-142 Additional examples (93 to 142) were prepared by procedures similar to that described for making Example 91 and 92 in a similar yield. H: Preparation of Compound 149: 1-(4-(trifluoromethyl)benzyl)-4-chloro-1H-pyrazolo(3,4-c)pyridine-3-methylamine To a solution of example 137 (32.6 g, 0.10 mmol) in THF (2 mL) was added iodomethane (42.6 mg, 0.30 mmol), and triethylamine (30.5 mgl, 0.30 mmol). The resulting reaction mixture was sealed and heated at 100 degree for 3 days and then the solvent was removed. The crude product was purified by Prep. HPLC (sunfire 5 u 100 mm column, MeOH/H2O as solvents) to yield 1-(4-(trifluoromethyl)benzyl)-4-chloro-1H-pyrazolo(3,4-c)pyridine-3-methylamine (16 mg, 0.047 mmol, 47% yield) as a gum. MS (ESI) m/z: 341 (M+H)+. I: Preparation of Compound 150: N-(1-(4-(trifluoromethyl)benzyl)-4-chloro-1H-pyrazolo(3,4-c)pyridyl)-3-acetamide To a solution of example 137 (32.6 g, 0.10 mmol) in THF (2 mL) was added acetyl chloride (15.6 mg, 0.20 mmol), and triethylamine (30.5 mgl, 0.30 mmol). The resulting reaction mixture was stirred at room temperature for 5 hours and then the solvent was removed. The crude product was purified by Prep. HPLC (sunfire 5 u 100 mm column, MeOH/H2O as solvents) to yield N-(1-(4-(trifluoromethyl)benzyl)-4-chloro-1H-pyrazolo(3,4-c)pyridyl)-3-acetamide (23 mg, 0.0625 mmol, 62.5% yield) as a white solid MS (ESI) m/z: 369 (M+H)+. J. Preparation of Compounds 151 to 153 The examples 151 to 153 were prepared by procedures similar to that described for making Example 150 in a similar yield. K. Preparation of Compound 143: 3-Bromo-1-(4-(trifluoromethyl)benzyl)-1H-indazole A mixture of KOH (1.12 g, 20 mmol) in DMSO (50 mL) was stirred at room temperature for 5 min. 3-bromo-1H-indazole (1.97 g, 10 mmol) was then added in one portion. The resulting mixture was stirred at room temperature for 5 min. A solution of 1-(bromomethyl)-4-(trifluoromethyl)benzene (3.6 g, 15 mmol) in DMSO (5 mL) was then added dropwise over 10 min. When the addition was complete, the resulting mixture was stirred at room temperature for an additional 1 h. The mixture was quenched by the addition of water (200 mL). The mixture was then extracted with CH2Cl2 (3×100 mL). The combined extracts were washed with H2O (2×100 mL), brine (1×100 mL), then dried over MgSO4, filtered and concentrated in vacuo. Purification by flash chromatography (Silica, 200 g, 10-100% EtOAc/Hexanes) gave 3-Bromo-1-(4-(trifluoromethyl)benzyl)-1H-indazole (3.1 g, 8.7 mmol, 87% yield) as an off-white crystalline solid. MS (ESI) m/z: 355 and 357 (M+H)+. L: Preparation of Compound 144: 3-Phenyl-1-(4-(trifluoromethyl)benzyl)-1H-indazole Cesium carbonate (65 mg, 0.20 mmol), 3-Bromo-1-(4-(trifluoromethyl)benzyl)-1H-indazole (35.6 mg, 0.10 mmol), phenylboronic acid (18.2 mg, 0.15 mmol) and PdCl2(dppf) (7.2 mg, 0.01 mmol) were suspended in dioxane (5 mL) and degassed with argon for 5 minutes. The reaction was sealed and heated at 90 degree overnight. The crude product was purified by Prep. HPLC (sunfire 5 u 100 mm column, MeOH/H2O as solvents). 23 mg of 3-Phenyl-1-(4-(trifluoromethyl)benzyl)-1H-indazole was obtained as a solid. MS (ESI) m/z: 353 (M+H)+. M: Preparation of Compounds 145-148 The examples 145 to 148 were prepared by procedures similar to that described for making Example 144 in a similar yield. Example 2: Human Fascin-1 Expression and Purification Recombinant human fascin 1 was expressed as a GST fusion protein in BL21Escherichia coli. One liter of 2YT medium with ampicillin was inoculated overnight with 3 mL of BL21/DE3 culture transformed with pGEX4T-fascin 1 plasmid and grown at 37° C. until attenuance at 600 nm (D600) reached about 0.8. The culture was then transferred to 18° C. and induced by the addition of 0.1 mM isopropyl β-d-thiogalactoside (IPTG) for 12 h. Bacteria were harvested by centrifugation at 5,000 r.p.m. for 10 min. The pellets were suspended in 30 mL of PBS supplemented with 0.2 mM PMSF, 1 mM DTT, 1% (v/v) Triton X-100 and 1 mM EDTA. After sonication, the suspension was centrifuged at 15,000 r.p.m. for 30 min to remove the cell debris. The supernatant was then incubated for 2 h with 4 mL of glutathione beads (Sigma) at 4° C. After extensive washing with PBS, the beads were resuspended in 10 mL of thrombin cleavage buffer (20 mM Tris-HCl pH 8.0, 150 mM NaCl, 2 mM CaCl2), 1 mM DTT). Fascin was released from the beads by incubation overnight with 40-100 U of thrombin at 4° C. After centrifugation, 0.2 mM PMSF was added to the supernatant to inactivate the remnant thrombin activity. The fascin protein was further concentrated with a Centricon® (Boca Raton, FL) filter to about 50 mg/mL. Example 3: Quantification of Fascin Expression Levels The levels of fascin mRNA and protein can be determined by real-time PCR and Western blot, respectively. For quantitative real-time PCR, samples from cancer patients were used for RNA isolation. Oligonucleotide primers specific for fascin mRNA were used for PCR reactions. For Western blots, samples from cancer patients were assessed with anti-fascin antibody. The intensity of the bands representing fascin proteins was quantified by image documentation and quantification software. Example 4: Compound Analysis Representative compounds described herein were tested for fascin inhibition activity. Purified fascin protein (15 μL of 0.5 μM) in buffer (100 mM KCl, 20 mM Tris/HCl, pH 7.5, 2 mM MgCl2) was added into each well of a clear 384-well flat-bottom plate (Corning) using Thermo Multidrop Combi (Fisher). Compound solutions (5 mM stock, 180 nL) were pin transferred from stock 384-well plates into the 384-well assay plates and incubated for 30 min. Then 15 μL of 0.5 μM polymerized actin (in 100 mM KCl, 20 mM Tris/HCl, pH 7.5, 2 mM MgCl2, 1 mM DTT, 1 mM ATP) (Cytoskeleton Inc.) was added, resulting in 30 μM final concentration for chemical compounds. After another 30 min, 10 μL of Alexa Fluro 488 Phalloidin (25 times dilution from stocks in 100% methanol, Invitrogen) was added to stain F-actin and was incubated in the dark for one hour. Mixed solution (25 μL) was then transferred to one well in a black 384-well plate coated with poly-D-lysine, and stained actin bundles or F-actin would stick onto the poly-D-lysine plates. After the plates were thoroughly washed with 1×PBS for 3 times, the plate was imaged using an ImageXpress Micro High Content Screening System (Molecular devices). The images were processed and analyzed using MetaMorph software. The raw image data for each well was background-corrected by subtraction of the median intensities across all wells on the plate. The background-corrected data was used to compute the bundle length for each well. The negative control wells were employed for quality control: multiple DMSO-only control wells (16 wells/plate) were present on each assay plate. In confirmative screening of the compounds, a control with another actin-bundling protein, fimbrin, can used to eliminate compounds that are not specific to fascin. Also in confirmative screening, each compound can be tested in duplicate on the same plate. The % inhibition values of certain compounds are shown in Table 1 above. Example 5: Boyden-Chamber Cell Migration Assay Boyden chamber assays for cell migration can be used to show the activity of the compounds described herein in inhibiting the migration of tumor cells, such as breast tumor cells, prostate tumor cells, and lung tumor cells. Certain tumor cells with fascin expression are listed below. 4T1 breast tumor cellsMDA-MB-231 breast tumor cellsDU145 prostate tumor cellsPC-3 prostate tumor cellsLLC lung tumor cells Exemplifying procedure: MDA-MB-231 cells (5×104) or 4T1 Cells (1×105) were suspended in 100 μl starvation medium and added to the upper chamber of an insert (6.5 mm diameter, 8 μm pore size; Becton Dickson). The insert was placed in a 24-well plate containing 700 μL starvation medium with or without 10% FBS. When used, inhibitors were added to the lower chamber. Migration assays were performed for 6 h and cells are fixed with 3.7% formaldehyde. Cells were stained with crystal violet staining solution, and cells on the upper side of the insert were removed with a cotton swab. Three randomly selected fields (×10 objectives) on the lower side of the insert were photographed, and the migrated cells were counted. Migration was expressed as average number of migrated cells in a field. The following are the IC50data of selected compounds when tested using MDA-MB-231 human breast tumor cells.Compound 10: 31 μMCompound 25: 54 μMCompound 35: 18 μMCompound 43: 12 μMCompound 49: 13 μMCompound 65: 64 μMCompound 66: 161 μM In vitro data obtained in such assays are known to correlate with results obtained from in vivo models. See, e.g., Shan, D., et al., Synthetic analogues of migrastatin that inhibit mammary tumor metastasis in mice, Proc. Nat. Acad. Sci. 102: 3772-3776 (2005). Example 6: Tumor Metastasis in Mouse Models Tumor cell migration is essential for tumor metastasis. Representative compounds described herein were investigated their effects on tumor metastasis in an animal model. Tumor cells (4T1 breast tumor cells) were injected into the mammary fat-pad of mice. The metastasis of these breast tumor cells from the mammary gland to the lung was monitored by the clonogenic assay. Balb/c mice were purchased from Charles River. All animal procedures were approved by the Animal Care and Use Committees of the Weill Cornell Medical College and performed in accordance with institutional polices. For xenograft tumor metastasis studies, 5×1054T1 cells were suspended in 100 μL PBS and injected subcutaneously into the mammary glands of 6-8 week old female Balb/c mice. Tumor incidence was monitored for 21 days after injection. Tumor size was measured three times a week, and the volume was calculated using the formula length×width2×0.5. Compound treatment was initiated 7 days after tumor implantation; animals were administered daily with indicated dose for 2 weeks. On day 28, the mice were sacrificed. Numbers of metastatic 4T1 cells in lungs were determined by the clonogenic assay. In brief, lungs were removed from each mouse on day 28, finely minced and digested for 2 h at 37° C. in 5 mL of enzyme cocktail containing PBS and 1 mg/mL collagenase type IV on a rocker. After incubation, samples were filtered through 70-μm nylon cell strainers and washed twice with PBS. Resulting cells were suspended, plated with a series of dilutions in 10-cm tissue culture dishes in RPMI-1640 medium containing 60 μM thioguanine, metastasized tumor cells formed foci after 14 days, at which time they were fixed with methanol and stained with 0.03% methylene blue for counting. Data were expressed as mean±S.D. and analyzed by Student's t test with significance defined as p<0.05. When tested in this animal model at 100 mg/kg, Compounds 10 and 43 showed more than 90% inhibition of tumor metastasis. The compounds described herein are contemplated to be useful for treating a condition or disorder mediated by fascin activity and/or tumor metastasis. Example 7: In Vivo Mouse Model for Prostate Tumor Metastasis 5- to 6-week-old male severe combined immunodeficient mice (n=20) purchased from Charles River (Wilmington, MA) are randomly divided into two groups (n=10 animals per group). In both two groups, human prostate tumor cells PC-3Luc cells (stably transfected with luciferase gene) (2×105cells in 100 μl of Dulbecco phosphate-buffered saline [PBS] lacking Ca2+and Mg2+) are introduced into animals by intracardiac injection under 1.75% isoflurane/air anesthesia. Throughout the duration of the experiment, animals in group 1 receive daily testing compounds administered intraperitoneally (i.p.) in 0.2 mL of sterile physiological saline beginning 1 week before tumor cell inoculation. In group 2 (untreated control), animals receive a daily 0.2 mL i.p. injection of the vehicle, sterile physiological saline. Mice are serially imaged weekly for 5 weeks using an IVIS system (Xenogen Corp, Alameda, CA), and the results are analyzed using Living Image software (Xenogen). For imaging, mice are injected with luciferin (40 mg/mL) i.p., and ventral images are acquired 15 minutes after injection under 1.75% isoflurane/air anesthesia. At the end of the experiments, animals are killed, and tissue is collected for histopathologic confirmation of bone metastasis. It is contemplated that less bone metastasis is found in group 1 animals treated with a fascin inhibitory compound disclosed herein as compared with that found in group 2 animals. As such the test compounds are useful for treating cancer, in particular, prostate tumor metastasis. Example 8: In Vivo Mouse Model for Lung Tumor Metastasis 20 mice are divided into two groups, and 2×106A549 human lung tumor cells are injected into each mouse via the tail vein. One group is treated with a compound disclosed herein and another group is used as control. After 8 weeks, the lungs are harvested, fixed, and embedded in paraffin. The number of metastatic lung nodules is counted in serial histological sections stained with H&E. The areas of metastatic lung nodules are measured in scanned images of the H&E-stained tumor sections using Paint.NET software. It is contemplated that the number and area of metastatic lung nodules in the treated animals are smaller than that of the untreated control animals. As such the test compounds are useful for treating cancer, in particular, lung tumor metastasis. Example 9: Treatment of Tumor Metastasis in Human Human patients having metastatic breast cancer are administered intravenously with a fascin inhibitory compound disclosed herein or placebo in a randomized open-label trial. The patients are separated into 5 groups. Patients in each group are administered a daily dosage of 0 mg (placebo), 100 mg, 200 mg, 500 mg, or 1000 mg of the compound, respectively, in 3-week cycles. The time to disease progression, overall response rate (ORR), duration of response, and overall survival (OS) rate are measured at the end of each cycle with known techniques. It is contemplated that patients administered with the fascin inhibitory compound have a longer mean or average time to disease progression and/or duration of response, a higher mean or average overall response rate and/or overall survival rate, than patients administered with placebo. Fewer new tumors distant from the original tumor site are developed in patients administered with fascin inhibitory compound than in patients administered with placebo. In a preferred embodiment, one or more of the results are dose-responsive. Side effects are monitored and recorded. As such the test compounds are useful for treating tumor metastasis in human. REFERENCES 1. Hanahan, D., and Weinberg, R. A. (2000) The hallmarks of cancer,Cell100, 57-70.2. Christofori, G. (2006) New signals from the invasive front,Nature441, 444-450.3. Weiss, L. (2000) Metastasis of cancer: a conceptual history from antiquity to the 1990s,Cancer Metastasis Rev19, I-XI, 193-383.4. Fidler, I. J. (2003) The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited,Nat Rev Cancer3, 453-458.5. Valastyan, S., and Weinberg, R. A. (2011) Tumor metastasis: molecular insights and evolving paradigms,Cell147, 275-292.6. Fornier, M. N. (2011) Approved agents for metastatic breast cancer,Semin Oncol38 Suppl 2, S3-10.7. Davies, J. M., and Goldberg, R. M. (2011) Treatment of metastatic colorectal cancer,Semin Oncol38, 552-560.8. Sondak, V. K., Han, D., Deneve, J., and Kudchadkar, R. (2011) Current and planned multicenter trials for patients with primary or metastatic melanoma,J Surg Oncol104, 430-437.9. Partin, A. W., Schoeniger, J. S., Mohler, J. L., and Coffey, D. S. (1989) Fourier analysis of cell motility: correlation of motility with metastatic potential,Proc Natl Acad Sci USA86, 1254-1258.10. Aznavoorian, S., Murphy, A. N., Stetler-Stevenson, W. G., and Liotta, L. A. (1993) Molecular aspects of tumor cell invasion and metastasis,Cancer71, 1368-1383.11. Condeelis, J., Singer, R. H., and Segall, J. E. (2005) The great escape: when cancer cells hijack the genes for chemotaxis and motility,Annu Rev Cell Dev Biol21, 695-718.12. Roussos, E. T., Condeelis, J. S., and Patsialou, A. (2011) Chemotaxis in cancer,Nat Rev Cancer11, 573-587.13. Jaffe, A. B., and Hall, A. (2005) Rho GTPases: biochemistry and biology,Annu Rev Cell Dev Biol21, 247-269.14. Matsudaira, P. (1994) Actin crosslinking proteins at the leading edge,Semin Cell Biol5, 165-174.15. Otto, J. J. (1994) Actin-bundling proteins,Curr Opin Cell Biol6, 105-109.16. Mogilner, A., and Rubinstein, B. (2005) The physics of filopodial protrusion,Biophys J89, 782-795.17. Mattila, P. K., and Lappalainen, P. (2008) Filopodia: molecular architecture and cellular functions,Nat Rev Mol Cell Biol9, 446-454.18. Otto, J. J., Kane, R. E., and Bryan, J. (1979) Formation of filopodia in coelomocytes: localization of fascin, a 58,000 dalton actin cross-linking protein,Cell17, 285-293.19. Bryan, J., and Kane, R. E. (1978) Separation and interaction of the major components of sea urchin actin gel,J Mol Biol125, 207-224.20. Yamashiro-Matsumura, S., and Matsumura, F. (1985) Purification and characterization of an F-actin-bundling 55-kilodalton protein from HeLa cells,J Biol Chem260, 5087-5097.21. Vignjevic, D., Yarar, D., Welch, M. D., Peloquin, J., Svitkina, T., and Borisy, G. G. (2003) Formation of filopodia-like bundles in vitro from a dendritic network,J Cell Biol160, 951-962.22. Vignjevic, D., Kojima, S., Aratyn, Y., Danciu, O., Svitkina, T., and Borisy, G. G. (2006) Role of fascin in filopodial protrusion,J Cell Biol174, 863-875.23. Adams, J. C. (2004) Roles of fascin in cell adhesion and motility,Curr Opin Cell Biol16, 590-596.24. Tilney, L. G., Connelly, P. S., Vranich, K. A., Shaw, M. K., and Guild, G. M. (1998) Why are two different cross-linkers necessary for actin bundle formation in vivo and what does each cross-link contribute?,J Cell Biol143, 121-133.25. Darnel, A. D., Behmoaram, E., Vollmer, R. T., Corcos, J., Bijian, K., Sircar, K., Su, J., Jiao, J., Alaoui-Jamali, M. A., and Bismar, T. A. (2009) Fascin regulates prostate cancer cell invasion and is associated with metastasis and biochemical failure in prostate cancer,Clin Cancer Res15, 1376-1383.26. Pelosi, G., Pasini, F., Fraggetta, F., Pastorino, U., Iannucci, A., Maisonneuve, P., Arrigoni, G., De Manzoni, G., Bresaola, E., and Viale, G. (2003) Independent value of fascin immunoreactivity for predicting lymph node metastases in typical and atypical pulmonary carcinoids,Lung Cancer42, 203-213.27. Hashimoto, Y., Shimada, Y., Kawamura, J., Yamasaki, S., and Imamura, M. (2004) The prognostic relevance of fascin expression in human gastric carcinoma,Oncology67, 262-270.28. Cao, D., Ji, H., and Ronnett, B. M. (2005) Expression of mesothelin, fascin, and prostate stem cell antigen in primary ovarian mucinous tumors and their utility in differentiating primary ovarian mucinous tumors from metastatic pancreatic mucinous carcinomas in the ovary,Int J Gynecol Pathol24, 67-72.29. Rodriguez-Pinilla, S. M., Sarrio, D., Honrado, E., Hardisson, D., Calero, F., Benitez, J., and Palacios, J. (2006) Prognostic significance of basal-like phenotype and fascin expression in node-negative invasive breast carcinomas,Clin Cancer Res12, 1533-1539.30. Grothey, A., Hashizume, R., Sahin, A. A., and McCrea, P. D. (2000) Fascin, an actin-bundling protein associated with cell motility, is upregulated in hormone receptor negative breast cancer,Br J Cancer83, 870-873.31. Hashimoto, Y., Skacel, M., and Adams, J. C. (2005) Roles of fascin in human carcinoma motility and signaling: prospects for a novel biomarker?,Int J Biochem Cell Biol37, 1787-1804.32. Maitra, A., Iacobuzio-Donahue, C., Rahman, A., Sohn, T. A., Argani, P., Meyer, R., Yeo, C. J., Cameron, J. L., Goggins, M., Kern, S. E., Ashfaq, R., Hruban, R. H., and Wilentz, R. E. (2002) Immunohistochemical validation of a novel epithelial and a novel stromal marker of pancreatic ductal adenocarcinoma identified by global expression microarrays: sea urchin fascin homolog and heat shock protein 47, Am J Clin Pathol118, 52-59.33. Yoder, B. J., Tso, E., Skacel, M., Pettay, J., Tarr, S., Budd, T., Tubbs, R. R., Adams, J. C., and Hicks, D. G. (2005) The expression of fascin, an actin-bundling motility protein, correlates with hormone receptor-negative breast cancer and a more aggressive clinical course,Clin Cancer Res11, 186-192.34. Zigeuner, R., Droschl, N., Tauber, V., Rehak, P., and Langner, C. (2006) Biologic significance of fascin expression in clear cell renal cell carcinoma: systematic analysis of primary and metastatic tumor tissues using a tissue microarray technique,Urology68, 518-522. EQUIVALENTS The embodiments, illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms ‘comprising,’ ‘including,’ ‘containing,’ etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the claimed technology. Additionally, the phrase ‘consisting essentially of’ will be understood to include those elements specifically recited and those additional elements that do not materially affect the basic and novel characteristics of the claimed technology. The phrase ‘consisting of’ excludes any element not specified. The present disclosure is not to be limited in terms of the particular embodiments described in this application, which are intended as illustrations of various aspects. Many modifications and variations can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent compositions, apparatuses, and methods within the scope of the disclosure, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. The present disclosure is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. It is to be understood that this disclosure is not limited to particular methods, reagents, compounds compositions or biological systems, which can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group. As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as ‘up to,’ ‘at least,’ ‘greater than,’ ‘less than,’ and the like, include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. While certain embodiments have been illustrated and described, it should be understood that changes and modifications can be made therein in accordance with ordinary skill in the art without departing from the technology in its broader aspects as defined in the following claims.
116,969
11858930
DETAILED DESCRIPTION All publications, patents and patent applications, including any drawings and appendices therein are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, patent or patent application, drawing, or appendix was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. Definitions While the following terms are believed to be well understood by one of ordinary skill in the art, the following definitions are set forth to facilitate explanation of the presently disclosed subject matter. Throughout the present specification, the terms “about” and/or “approximately” may be used in conjunction with numerical values and/or ranges. The term “about” is understood to mean those values near to a recited value. Furthermore, the phrases “less than about [a value]” or “greater than about [a value]” should be understood in view of the definition of the term “about” provided herein. The terms “about” and “approximately” may be used interchangeably. Throughout the present specification, numerical ranges are provided for certain quantities. It is to be understood that these ranges comprise all subranges therein. Thus, the range “from 50 to 80” includes all possible ranges therein (e.g., 51-79, 52-78, 53-77, 54-76, 55-75, 60-70, etc.). Furthermore, all values within a given range may be an endpoint for the range encompassed thereby (e.g., the range 50-80 includes the ranges with endpoints such as 55-80, 50-75, etc.). The term “a” or “an” refers to one or more of that entity; for example, “a RAF inhibitor” refers to one or more RAF inhibitor or at least one RAF inhibitor. As such, the terms “a” (or “an”), “one or more” and “at least one” are used interchangeably herein. In addition, reference to “an inhibitor” by the indefinite article “a” or “an” does not exclude the possibility that more than one of the inhibitors is present, unless the context clearly requires that there is one and only one of the inhibitors. As used herein, the verb “comprise” as is used in this description and in the claims and its conjugations are used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. The present invention may suitably “comprise”, “consist of”, or “consist essentially of”, the steps, elements, and/or reagents described in the claims. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely”, “only” and the like in connection with the recitation of claim elements, or the use of a “negative” limitation. The term “pharmaceutically acceptable salts” includes both acid and base addition salts. Pharmaceutically acceptable salts include those obtained by reacting the active compound functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, camphorsulfonic acid, oxalic acid, maleic acid, succinic acid, citric acid, formic acid, hydrobromic acid, benzoic acid, tartaric acid, fumaric acid, salicylic acid, mandelic acid, carbonic acid, etc. Those skilled in the art will further recognize that acid addition salts may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. The term “treating” means one or more of relieving, alleviating, delaying, reducing, improving, or managing at least one symptom of a condition in a subject. The term “treating” may also mean one or more of arresting, delaying the onset (i.e., the period prior to clinical manifestation of the condition) or reducing the risk of developing or worsening a condition. The compounds of the invention, or their pharmaceutically acceptable salts contain at least one asymmetric center. The compounds of the invention with one asymmetric center give rise to enantiomers, where the absolute stereochemistry can be expressed as (R)- and (S)-, or (+) and (−). When the compounds of the invention have more than two asymmetric centers, then the compounds can exist as diastereomers or other stereoisomeric forms. The present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms whether or not they are specifically depicted herein. Optically active (+) and (−) or (R)- and (S)-isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposable mirror images of one another. A “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present disclosure includes tautomers of any said compounds. An “effective amount” means the amount of a formulation according to the invention that, when administered to a patient for treating a state, disorder or condition is sufficient to effect such treatment. The “effective amount” will vary depending on the active ingredient, the state, disorder, or condition to be treated and its severity, and the age, weight, physical condition and responsiveness of the mammal to be treated. The term “therapeutically effective” applied to dose or amount refers to that quantity of a compound or pharmaceutical formulation that is sufficient to result in a desired clinical benefit after administration to a patient in need thereof. As used herein, a “subject” can be a human, non-human primate, mammal, rat, mouse, cow, horse, pig, sheep, goat, dog, cat and the like. The subject can be suspected of having or at risk for having a cancer, including but not limited to colorectal cancer and melanoma. “Mammal” includes humans and both domestic animals such as laboratory animals (e.g., mice, rats, monkeys, dogs, etc.) and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like. All weight percentages (i.e., “% by weight” and “wt. %” and w/w) referenced herein, unless otherwise indicated, are measured relative to the total weight of the pharmaceutical composition. As used herein, “substantially” or “substantial” refers to the complete or nearly complete extent or degree of an action, characteristic, property, state, structure, item, or result. For example, an object that is “substantially” enclosed would mean that the object is either completely enclosed or nearly completely enclosed. The exact allowable degree of deviation from absolute completeness may in some cases depend on the specific context. However, generally speaking, the nearness of completion will be so as to have the same overall result as if absolute and total completion were obtained. The use of “substantially” is equally applicable when used in a negative connotation to refer to the complete or near complete lack of action, characteristic, property, state, structure, item, or result. For example, a composition that is “substantially free of” other active agents would either completely lack other active agents, or so nearly completely lack other active agents that the effect would be the same as if it completely lacked other active agents. In other words, a composition that is “substantially free of” an ingredient or element or another active agent may still contain such an item as long as there is no measurable effect thereof. The term “halo” refers to a halogen. In particular the term refers to fluorine, chlorine, bromine and iodine. “Alkyl” or “alkyl group” refers to a fully saturated, straight or branched hydrocarbon chain group, and which is attached to the rest of the molecule by a single bond. Alkyls comprising any number of carbon atoms, including but not limited to from 1 to 12 are included. An alkyl comprising up to 12 carbon atoms is a C1-C12alkyl, an alkyl comprising up to 10 carbon atoms is a C1-C10alkyl, an alkyl comprising up to 6 carbon atoms is a C1-C6alkyl and an alkyl comprising up to 5 carbon atoms is a C1-C5alkyl. A C1-C5alkyl includes C5alkyls, C4alkyls, C3alkyls, C2alkyls and C1alkyl (i.e., methyl). A C1-C6alkyl includes all moieties described above for C1-C5alkyls but also includes C6alkyls. A C1-C10alkyl includes all moieties described above for C1-C5alkyls and C1-C6alkyls, but also includes C7, C8, C9and C10alkyls. Similarly, a C1-C12alkyl includes all the foregoing moieties, but also includes C11and C12alkyls. Non-limiting examples of C1-C12alkyl include methyl, ethyl, n-propyl, i-propyl, sec-propyl, n-butyl, i-butyl, sec-butyl, t-butyl, n-pentyl, t-amyl, n-hexyl, n-heptyl, n-octyl, n-Nonyl, n-decyl, n-undecyl, and n-dodecyl. Unless stated otherwise specifically in the specification, an alkyl group can be optionally substituted. “Cycloalkyl” refers to a stable non-aromatic monocyclic or polycyclic fully saturated hydrocarbon group consisting solely of carbon and hydrogen atoms, which can include fused or bridged ring systems, having from three to twenty carbon atoms, preferably having from three to ten carbon atoms, and which is attached to the rest of the molecule by a single bond. Monocyclic cycloalkyl groups include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic cycloalkyl groups include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Unless otherwise stated specifically in the specification, a cycloalkyl group can be optionally substituted. “Haloalkyl” refers to an alkyl group, as defined above, that is substituted by one or more halo groups, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group can be optionally substituted. “Aryl” refers to a hydrocarbon ring system group comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring. For purposes of this invention, the aryl group can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems. Aryl groups include, but are not limited to, aryl groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, the term “aryl” is meant to include aryl groups that are optionally substituted. “Heterocyclyl,” “heterocyclic ring” or “heterocycle” refers to a stable 3- to 20-membered ring group which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Heterocyclycl or heterocyclic rings include heteroaryls as defined below. Unless stated otherwise specifically in the specification, the heterocyclyl group can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl group can be optionally oxidized; the nitrogen atom can be optionally quaternized; and the heterocyclyl group can be partially or fully saturated. Examples of such heterocyclyl groups include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocyclyl group can be optionally substituted. In embodiments, heterocyclyl, heterocyclic ring or heterocycle is a stable 3- to 20-membered non-aromatic ring group which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. “N-heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, a N-heterocyclyl group can be optionally substituted. “Heteroaryl” refers to a 5- to 20-membered ring system group comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl group can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl group can be optionally oxidized; the nitrogen atom can be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridinyl, 1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1-oxidopyridazinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteroaryl group can be optionally substituted. “N-heteroaryl” refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an N-heteroaryl group can be optionally substituted. The term “substituted” used herein means any of the above groups (i.e., alkyl, alkylene, alkenyl, alkenylene, alkynyl, alkynylene, alkoxy, alkylamino, alkylcarbonyl, thioalkyl, aryl, aralkyl, carbocyclyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, C1, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. “Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, “substituted” includes any of the above groups in which one or more hydrogen atoms are replaced with —NRgRh, —NRgC(═O)Rh, —NRgC(═O)NRgRh, —NRgC(═O)ORh, —NRgSO2Rh, —OC(═O)NRgRh, —ORg, —SRg, —SORg, —SO2Rg, —OSO2Rg, —SO2ORg, ═NSO2Rg, and —SO2NRgRh. “Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced with —C(═O)Rg, —C(═O)ORg, —C(═O)NRgRh, —CH2SO2Rg, —CH2SO2NRgRh. In the foregoing, Rgand Rhare the same or different and independently hydrogen, alkyl, alkenyl, alkynyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, haloalkenyl, haloalkynyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl. “Substituted” further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkenyl, alkynyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, haloalkenyl, haloalkynyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl group. In addition, each of the foregoing groups can also be optionally substituted with one or more of the above groups. Compounds of the Invention The present disclosure relates to pan-RAF inhibitors having the structure of formula (I), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein ring A is a 5-membered heterocycle or heteroaryl containing 1, 2, or 3, nitrogen atom as a ring member;wherein one of R1or R2is selected from substituted C1-8alkyl, unsubstituted C5-8alkyl, substituted or unsubstituted C1-8haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl, and the other R1or R2is H;or alternatively, R1and R2together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted;wherein when ring A is an imidazole, then the substituted aryl (e.g., R1or R2) is not andwherein when ring A is an imidazole, R1and R2together with the atoms to which they are attached do not form an unsubstituted phenyl ring. In embodiments of formula (I), ring A is imidazole, pyrazole, or triazole. In embodiments of formula (I), ring A is In embodiments, the present disclosure relates compounds of formula (I-A), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein one of R1or R2is selected from substituted or unsubstituted C5-8alkyl, substituted or unsubstituted C1-8haloalkyl, substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl, and the other R1or R2is H;or alternatively, R1and R2together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted;wherein the substituted aryl (e.g., R1or R2) is not andwherein R1and R2together with the atoms to which they are attached do not form an unsubstituted phenyl. In embodiments of formula (I) or (I-A), substituent is selected from halogen, —ORA, —NRARB, —SO2RC, —SORC, —CN, C1-6alkyl, C1-6haloalkyl, C3-6cycloalkyl, or —C(O)C1-6alkyl; wherein:RAand RBare each independently selected from H, C1-6alkyl and C1-6haloalkyl; andRCis selected from C1-6alkyl and C1-6haloalkyl; andwherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: —ORA, —CN, —SORC, —NRARB, or —NRDRE;RDand REtogether with the N atom to which they are attached forms a 5- or 6-membered saturated or partially unsaturated ring containing 1 or 2 heteroatoms selected from N, O, or S; wherein the saturated or partially unsaturated ring is optionally substituted with C1-6alkyl. In embodiments, the present disclosure relates compounds of formula (I-B), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein one of R1or R2is selected from substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl; andwherein the substituted aryl is not In embodiments of formula (I), (I-A), or (I-B), substituent is selected from halogen, —ORA, —NRARB, —SO2RC, —SORC, —CN, C1-6alkyl, C1-6haloalkyl, C3-6cycloalkyl, or —C(O)C1-6alkyl; wherein RAand RBare each independently selected from H, C1-6alkyl and C1-6haloalkyl; and RCis selected from C1-6alkyl and C1-6haloalkyl; wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: —ORA, —CN, —SORC, or —NRARB. In embodiments of formula (I), (I-A), or (I-B), substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, —CH2F, —CHF2, —CF3, —CH2CH2F, —CH2CHF2, —CH2CF3, —C(O)CH3, —CN, —OH, —NH2, —NH(C1-3alkyl), —N(C1-3alkyl)2, —CH2NH2, —CH2NH(C1-3alkyl), or —CH2N(C1-3alkyl)2. In embodiments of formula (I), (I-A), or (I-B), substituent on the substituted aryl or the substituted heteroaryl is selected from halogen, —ORA, —NRARB, —SO2RC, —SORC, —CN, C1-6alkyl, C1-6haloalkyl, C3-6cycloalkyl, or —C(O)C1-6alkyl; wherein RAand RBare each independently selected from H, C1-6alkyl and C1-6haloalkyl; and RCis selected from C1-6alkyl and C1-6haloalkyl. In embodiments, substituent is selected from methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, —CH2F, —CHF2, —CF3, —CH2CH2F, —CH2CHF2, —CH2CF3, —C(O)CH3, —CN, —OH, —NH2, —NH(C1-3alkyl), —N(C1-3alkyl)2, —CH2NH2, —CH2NH(C1-3alkyl), or —CH2N(C1-3alkyl)2. In embodiments of formula (I), (I-A), or (I-B), substituent on the substituted aryl is selected from C1-6alkyl, C1-6haloalkyl, C3-6cycloalkyl, —ORA, —NRARB, —CN, - or —C(O)C1-6alkyl. In embodiments of formula (I) or (I-A), R1is substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl. In embodiments, substituted aryl is substituted phenyl. In embodiments, substituted heteroaryl is substituted N-heteroaryl. In embodiments, substituted heteroaryl is substituted 5- or 6-membered N-heteroaryl. In embodiments of formula (I), (I-A), or (I-B), R1is substituted phenyl, substituted pyridyl, substituted pyrazole, substituted pyrimidinyl, or substituted thiophenyl. In embodiments of formula (I), (I-A), or (I-B), R1is substituted phenyl, substituted pyridyl, or substituted pyrazole. In embodiments of formula (I), (I-A), or (I-B), R1is substituted or unsubstituted heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R1is substituted or unsubstituted tetrahydropyran. In embodiments, R1is substituted or unsubstituted In embodiments of formula (I-B), R1is substituted or unsubstituted heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R1is substituted or unsubstituted 5- or 6-membered heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R1is substituted or unsubstituted saturated heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R1is substituted or unsubstituted saturated 5- or 6-membered heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R1is substituted or unsubstituted tetrahydropyran. In embodiments of formula (I) or (I-A), R1is substituted or unsubstituted C5-6alkyl. In embodiments, C5-6alkyl is linear or branched. In embodiments of formula (I), (I-A), or (I-B), R1is a monocyclic substituted aryl or a monocyclic substituted heteroaryl. In embodiments of formula (I), R1is a bicyclic substituted or unsubstituted aryl or a bicyclic substituted heteroaryl. In embodiments of formula (I), (I-A), or (I-B), R1is a bicyclic substituted aryl or a bicyclic substituted heteroaryl. In embodiments of formula (I), (I-A), or (I-B), R1is a fused bicyclic substituted aryl or a fused bicyclic substituted heteroaryl. In embodiments of formula (I), R1is substituted or unsubstituted indazole or substituted or unsubstituted benzoimidazole. In embodiments of formula (I), (I-A), or (I-B), R1is substituted indazole or substituted benzoimidazole. In embodiments of formula (I) or (I-A), R1or R2is substituted with 1, 2, or 3 substituents. In embodiments of formula (I) or (I-A), R1or R2is substituted with 1 or 2 substituents. In embodiments of formula (I-B), R1is substituted with 1, 2, or 3 substituents. In embodiments of formula (I-B), R1is substituted with 1 or 2 substituents. In embodiments of formula (I) or (I-A), R1and R2together with the atoms to which they are attached forms a 5- or 6-membered partially unsaturated or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted. In embodiments, R1and R2together with the atoms to which they are attached forms a 6-membered partially unsaturated or unsaturated ring containing 0 or 1 nitrogen atom in the ring, wherein the ring is substituted or unsubstituted. In embodiments of formula (I-A), R1and R2together with the atoms to which they are attached forms a phenyl ring, which is substituted or unsubstituted. In embodiments, R1and R2together with the atoms to which they are attached forms a phenyl ring thereby forming a benzoimidazole ring with the imidazole ring depicted in formula (I-A), which is substituted or unsubstituted. In embodiments of formula (I-A), R1and R2together with the atoms to which they are attached forms a pyridyl ring, which is substituted or unsubstituted. In embodiments, R1and R2together with the atoms to which they are attached forms a pyridyl ring thereby forming a imidazopyridine ring with the imidazole ring depicted in formula (I-A), which is substituted or unsubstituted. In embodiments of formula (I-A), R1and R2together with the atoms to which they are attached forms a tetrahydropyridyl ring, which is substituted or unsubstituted. In embodiments, R1and R2together with the atoms to which they are attached forms a tetrahydropyridyl ring thereby forming a tetrahydroimidazopyridine ring with the imidazole ring depicted in formula (I-A), which is substituted or unsubstituted. In embodiments of formula (I) or (I-A), R1and R2together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is unsubstituted. In embodiments of formula (I) or (I-A), R1and R2together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted with 1, 2, 3, or 4 substituents. In embodiments, the ring is substituted with 1 or 2 substituents. In embodiments, any substituents listed in embodiments of formula (I), (I-A), or (I-B) can be applicable to embodiments of formula (II) or (III). In embodiments of formula (I), (I-A), or (I-B), the compound is a racemate. In embodiments, the compound is an (R) stereoisomer. In embodiments, the compound is an (S) stereoisomer. In embodiments, the compound of formula (I), (I-A), or (I-B) is selected from Tables A-1, A-2, or A-3, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. TABLE A-1NoStructure123456789101112131415161718192021101103104105106107108109110111112113114115116117118119120121122123124125126127128129130131132 TABLE A-2NoStructure22232425262728293031323334133134135 TABLE A-3NoStructure35363738394041424344 In embodiments, the present disclosure relates compounds of formula (II), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: X1and X2are each N or CH; R1is selected from substituted C1-8alkyl, unsubstituted C5-8alkyl, substituted or unsubstituted C1-8haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl; and R4is —NRFC(O)R5, —NRFC(O)CH2R5, —NRFC(O)CH(CH3)R5, or —NRFR5; R5is substituted or unsubstituted group selected from carbocyclyl, aryl, heterocyclyl, or heteroaryl; and RFis selected from H or C1-3alkyl. In embodiments of the compounds of formula (II), one of X1and X2is N. In embodiments, X1is N and X2CH. In embodiments, X2is N and X1CH. In embodiments, X1and X2are both CH. In embodiments of the compounds of formula (II), R1is substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl. In embodiments, R1is substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrimidinyl, or substituted or unsubstituted thiophenyl. In embodiments, R1is substituted or unsubstituted phenyl. In embodiments, R1is substituted phenyl. In embodiments of the compounds of formula (II), R4is —NHC(O)R5, —NHC(O)CH2R5, —NHC(O)CH(CH3)R5, or —NHR5. In embodiments, R4is —NHC(O)R5, —NHC(O)CH2R5, or —NHR5. In embodiments of the compounds of formula (II), R5is substituted or unsubstituted group selected from alkyl, 3-6 membered carbocyclyl, phenyl, 3-6 membered heterocyclyl, or 5-6 membered heteroaryl. In embodiments, R5is substituted or unsubstituted group selected from 3-6 membered carbocyclyl, phenyl, 3-6 membered heterocyclyl, or 5-6 membered heteroaryl. In embodiments, R5is substituted or unsubstituted cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In embodiments, R5is substituted or unsubstituted heterocyclyl containing 1 or 2 heteroatoms selected from N, O, or S. In embodiments, R5is substituted or unsubstituted 5- or 6-membered heterocyclyl containing 1 or 2 heteroatoms selected from N, O, or S. In embodiments, R5is substituted or unsubstituted saturated heterocyclyl containing 1 or 2 heteroatoms selected from N, O, or S. In embodiments, R5is substituted or unsubstituted saturated 5- or 6-membered heterocyclyl containing 1 or 2 heteroatoms selected from N, O, or S. In embodiments, R5is substituted or unsubstituted azetidine, pyrrolidine, piperidine, piperazine, or morpholine moiety. In embodiments, R5is substituted or unsubstituted group selected from methyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, azetidine, pyrrolidine, piperidine, piperazine, or morpholine, pyridine, thiazole, imidazole, pyrazole, or triazole. In embodiments of the compounds of formula (II), R5is substituted or unsubstituted 5-6 membered heteroaryl. In embodiments, R5is substituted or unsubstituted 5-6 membered heteroaryl containing at least 1 nitrogen atom as a ring member. In embodiments, R5is substituted or unsubstituted pyridine, thiazole, imidazole, pyrazole, or triazole. In embodiments of the compounds of formula (II), RFis H or methyl. In embodiments, RFis H. In embodiments of formula (II), substituent is selected from halogen, —ORA, —NRARB, —SO2RC, —SORC, —CN, C1-6alkyl, C1-6haloalkyl, C3-6cycloalkyl, or —C(O)C1-6alkyl; wherein RAand RBare each independently selected from H, C1-6alkyl and C1-6haloalkyl; and RCis selected from C1-6alkyl and C1-6haloalkyl; wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: —ORA, —CN, —SORC, or —NRARB. In embodiments of formula (II), substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, —CH2F, —CHF2, —CF3, —CH2CH2F, —CH2CHF2, —CH2CF3, —C(O)CH3, —CN, —OH, —NH2, —NH(C1-3alkyl), —N(C1-3alkyl)2, —CH2NH2, —CH2NH(C1-3alkyl), or —CH2N(C1-3alkyl)2. In embodiments of formula (II), R5is substituted with a substituent selected from methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, —CH2F, —CHF2, —CF3, —CH2CH2F, —CH2CHF2, —CH2CF3, —C(O)CH3, —CN, —OH, —NH2, —NH(C1-3alkyl), —N(C1-3alkyl)2, —CH2NH2, —CH2NH(C1-3alkyl), or —CH2N(C1-3alkyl)2. In embodiments, R5is substituted with one or more substituent selected from halogen, methyl, ethyl, propyl, isopropyl, —CN, —OH, or —NH2. In embodiments of formula (II), R1is substituted with 1, 2, or 3 substituents. In embodiments of formula (II), R1is substituted with 1 or 2 substituents. In embodiments of formula (II), R1is unsubstituted. In embodiments of formula (II), R5is substituted with 1, 2, or 3 substituents. In embodiments of formula (II), R5is substituted with 1 or 2 substituents. In embodiments of formula (II), R5is unsubstituted. In embodiments, any substituents listed in embodiments of formula (II) can be applicable to embodiments of formula (I), (I-A), (I-B) or (III). In embodiments of formula (II), the compound is a racemate. In embodiments, the compound is an (R) stereoisomer. In embodiments, the compound is an (S) stereoisomer. In embodiments, the compound of formula (II) is selected from Table B, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. TABLE BNoStructureNoStructure45136148137149138150139151140152141153142154143155144156145157146158147 In embodiments, the present disclosure relates compounds of formula (III), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: R6is —C(O)NRFR5, —C(O)NRFCH2R5, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl;R5is substituted or unsubstituted group selected from carbocyclyl, aryl, heterocyclyl, or heteroaryl; andRFis selected from H or C1-3alkyl. In embodiments of the compounds of formula (III), R6is —C(O)NRFR5or —C(O)NRFCH2R5. In embodiments, R6is —C(O)NHR5or —C(O)NHCH2R5. In embodiments of the compounds of formula (III), R5is substituted or unsubstituted aryl. In embodiments, R5is substituted or unsubstituted phenyl. In embodiments of the compounds of formula (III), R6is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl. In embodiments, a monocyclic substituted aryl or a monocyclic substituted heteroaryl. In embodiments, R6is a bicyclic substituted aryl or a monocyclic substituted heteroaryl. In embodiments, R6is a fused bicyclic substituted aryl or a fused bicyclic substituted heteroaryl. In embodiments, R6is substituted or unsubstituted indazole or substituted or unsubstituted benzoimidazole. In embodiments of the compounds of formula (III), R6is substituted aryl or substituted heteroaryl, wherein the substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, —CH2F, —CHF2, —CF3, —CH2CH2F, —CH2CHF2, —CH2CF3, —C(O)CH3, —CN, —OH, —NH2, —NH(C1-3alkyl), —N(C1-3alkyl)2, —CH2NH2, —CH2NH(C1-3alkyl), —CH2N(C1-3alkyl)2, optionally substituted phenyl, optionally substituted heteroaryl. In embodiments of the compounds of formula (III), R6is substituted aryl or substituted heteroaryl, wherein the substituent is selected from optionally substituted phenyl or optionally substituted 5- or 6-membered heteroaryl. In embodiments, R6is substituted aryl or substituted heteroaryl, wherein the substituent is selected from phenyl or optionally substituted 5- or 6-membered N-heteroaryl. In embodiments, R6is substituted aryl or substituted heteroaryl, wherein the substituent is selected from phenyl or optionally substituted 5-membered N-heteroaryl. In embodiments, R6is substituted aryl or substituted heteroaryl, wherein the substituent is selected from phenyl or pyrazole substituted with one or two C1-3alkyl groups. In embodiments of formula (III), substituent is selected from halogen, —ORA, —NRARB, —SO2RC, —SORC, —CN, C1-6alkyl, C1-6haloalkyl, C3-6cycloalkyl, —C(O)C1-6alkyl, aryl, or heteroaryl; wherein RAand RBare each independently selected from H, C1-6alkyl and C1-6haloalkyl; and RCis selected from C1-6alkyl and C1-6haloalkyl; wherein the alkyl, haloalkyl, cycloalkyl, aryl, and heteroaryl groups are optionally substituted with 1 to 3 groups independently selected from: C1-6alkyl, —ORA, —CN, —SORC, or —NRARB. In embodiments of formula (II), substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, —CH2F, —CHF2, —CF3, —CH2CH2F, —CH2CHF2, —CH2CF3, —C(O)CH3, —CN, —OH, —NH2, —NH(C1-3alkyl), —N(C1-3alkyl)2, —CH2NH2, —CH2NH(C1-3alkyl), —CH2N(C1-3alkyl)2, optionally substituted phenyl, optionally substituted heteroaryl. In embodiments of formula (III), R5is substituted with 1, 2, or 3 substituents. In embodiments of formula (III), R5is substituted with 1 or 2 substituents. In embodiments of formula (III), R5is unsubstituted. In embodiments, any substituents listed in embodiments of formula (III) can be applicable to embodiments of formula (I), (I-A), (I-B) or (II). In embodiments, the compound of formula (III) is selected from Table C, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. TABLE CNoStructureNoStructure4649475048159162160163161 In embodiments, the present disclosure relates a compound in Table D, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. TABLE DNoStructure51 In embodiments, the compounds of the present disclosure have pERKA375 (1 hr) pIC50in the range of about 4 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK A375 (1 hr) pIC50in the range of about 5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK A375 (1 hr) pIC50value of about 5.0, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, about 6.0, about 6.1, about 6.2, about 6.3, about 6.4, about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 dimer (1 hr) pIC50in the range of about 4 to about 9 including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 dimer (1 hr) pIC50in the range of about 5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 dimer (1 hr) pIC50of about 5.0, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, about 6.0, about 6.1, about 6.2, about 6.3, about 6.4, about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2 about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0 including any values therebetween. In embodiments, the compounds of the present disclosure have a pERK A375 monomer/pERK HCT116 dimer ratio in the range of about 0.01 to about 2.5, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have a pERK A375 monomer/pERK HCT116 dimer ratio in the range of about 0.03 to about 2.0, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have a pERK monomer/dimer ratio of about 0.1, about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1.0, about 1.1, about 1.2, about 1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.8, about 1.9, or about 2.0, including any values therebetween. In embodiments, the compounds of the present disclosure have a pERK A375 monomer/pERK HCT116 dimer ratio of 2 or less. A pERK A375 monomer/pERK HCT116 dimer ratio is calculated using the potency in nM. In embodiments, the compounds of the present disclosure have pERK HCT116 (2 hr) absolute pIC50in the range of about 6 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 (2 hr) absolute pIC50in the range of about 6.5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 (2 hr) absolute pIC50of about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 (2 hr) absolute pIC50of about 6.5 or greater. In embodiments, the compounds of the present disclosure have pERK WiDr (2 hr) absolute pIC50in the range of about 6 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK WiDr (2 hr) absolute pIC50in the range of about 6.5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK WiDr (2 hr) absolute pIC50of about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pERK WiDr (2 hr) absolute pIC50of about 6.5 or greater. In embodiments, the compounds of the present disclosure have pGI50 3D HCT116 in the range of about 6 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D HCT116 in the range of about 6.5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D HCT116 of about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D HCT116 of about 6.5 or greater. In embodiments, the compounds of the present disclosure have pGI50 3D WiDr in the range of about 6 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D WiDr in the range of about 6.5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D WiDr of about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D WiDr of about 6.5 or greater. In embodiments, the compounds of the present disclosure have human liver microsome (HLM) intrinsic clearance (CLint) in the range of about 1 μL/min/mg to about 25 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have HLM CLint in the range of about 1 μL/min/mg to about 20 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have HLM CLint in the range of about 1 μL/min/mg to about 15 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have HLM CLint in the range of about 1 μL/min/mg to about 10 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have HLM CLint of about 1 μL/min/mg, about 2 μL/min/mg, about 3 μL/min/mg, about 4 μL/min/mg, about 5 μL/min/mg, about 6 μL/min/mg, about 7 μL/min/mg, about 8 μL/min/mg, about 9 μL/min/mg, about 10 μL/min/mg, about 11 μL/min/mg, about 12 μL/min/mg, about 13 μL/min/mg, about 14 μL/min/mg, or about 15 μL/min/mg, including any values therebetween. In embodiments, the compounds of the present disclosure have HLM CLint of less than about 15 μL/min/mg. In embodiments, the compounds of the present disclosure have HLM CLint of less than about 20 μL/min/mg. In embodiments, the compounds of the present disclosure have mouse liver microsome (MLM) intrinsic clearance (CLint) in the range of about 1 μL/min/mg to about 130 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have MLM CLint in the range of about 1 μL/min/mg to about 50 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have MLM CLint in the range of about 1 μL/min/mg to about 30 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have MLM CLint in the range of about 1 μL/min/mg to about 20 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have MLM CLint of about 1 μL/min/mg, about 2 μL/min/mg, about 3 μL/min/mg, about 4 μL/min/mg, about 5 μL/min/mg, about 6 μL/min/mg, about 7 μL/min/mg, about 8 μL/min/mg, about 9 μL/min/mg, about 10 μL/min/mg, about 11 μL/min/mg, about 12 μL/min/mg, about 13 μL/min/mg, about 14 μL/min/mg, about 15 μL/min/mg, about 16 μL/min/mg, about 17 μL/min/mg, about 18 μL/min/mg, about 19 μL/min/mg, or about 20 μL/min/mg, including any values therebetween. In embodiments, the compounds of the present disclosure have MLM CLint of less than about 30 μL/min/mg. In embodiments, the compounds of the present disclosure have MLM CLint of less than about 25 μL/min/mg. In embodiments, the compounds of the present disclosure have MLM CLint of less than about 20 μL/min/mg. In embodiments, the compounds of the present disclosure have human plasma protein binding (hPPB) percent free value in the range of about 0.01% to about 15%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have hPPB percent free value in the range of about 0.05% to about 10%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have hPPB percent free value in the range of about 0.1% to about 8%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have hPPB percent free value in the range of about 0.10% to about 5%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have mouse plasma protein binding (mPPB) percent free value in the range of about 0.01% to about 15%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have mPPB percent free value in the range of about 0.05% to about 10%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have mPPB percent free value in the range of about 0.1% to about 8%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have mPPB percent free value in the range of about 0.10% to about 5%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have fed state simulated intestinal fluid (FESSIF) solubility in the range of about 10 mg/L to about 1000 mg/L, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have FESSIF solubility in the range of about 20 mg/L to about 750 mg/L, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have FESSIF solubility in the range of about 25 mg/L to about 600 mg/L, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have FESSIF solubility of about 10 mg/L or greater. In embodiments, the compounds of the present disclosure have FESSIF solubility of about 20 mg/L or greater. In embodiments, the compounds of the present disclosure have FESSIF solubility of about 25 mg/L or greater. In embodiments, the compounds of the present disclosure have FESSIF solubility of about 30 mg/L or greater. Therapeutic Use The present disclosure also relates to method of using compounds of formula (I), (I-A), (I-B), (II), or (III), or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, for treating various diseases and conditions. In embodiments, compounds of formula (I), (I-A), (I-B), (II), or (III), or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, are useful for treating a disease or a condition implicated by abnormal activity of one or more Raf kinase. In embodiments, compounds of formula (I), (I-A), (I-B), (II), or (III), or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, are useful for treating a disease or a condition treatable by the inhibition of one or more Raf kinase. RAF kinase inhibition is relevant for the treatment of many different diseases associated with the abnormal activity of the MAPK pathway. In embodiments the condition treatable by the inhibition of RAF kinases, such as B-RAF or C-RAF. The present disclosure also relates to method of using compounds of Tables A1-A3 and B-D, or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, for treating various diseases and conditions. In embodiments, compounds of Tables A1-A3 and B-D, or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, are useful for treating a disease or a condition implicated by abnormal activity of one or more Raf kinase. In embodiments, compounds of Tables A1-A3 and B-D, or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, are useful for treating a disease or a condition treatable by the inhibition of one or more Raf kinase. RAF kinase inhibition is relevant for the treatment of many different diseases associated with the abnormal activity of the MAPK pathway. In embodiments the condition treatable by the inhibition of RAF kinases, such as B-RAF or C-RAF. In embodiments, the disease or the condition is cancer. In embodiments, the disease or the condition is selected from Barret's adenocarcinoma; biliary tract carcinomas; breast cancer; cervical cancer; cholangiocarcinoma; central nervous system tumors; primary CNS tumors; glioblastomas, astrocytomas; glioblastoma multiforme; ependymomas; secondary CNS tumors (metastases to the central nervous system of tumors originating outside of the central nervous system); brain tumors; brain metastases; colorectal cancer; large intestinal colon carcinoma; gastric cancer; carcinoma of the head and neck; squamous cell carcinoma of the head and neck; acute lymphoblastic leukemia; acute myelogenous leukemia (AML); myelodysplastic syndromes; chronic myelogenous leukemia; Hodgkin's lymphoma; non-Hodgkin's lymphoma; megakaryoblastic leukemia; multiple myeloma; erythroleukemia; hepatocellular carcinoma; lung cancer; small cell lung cancer; non-small cell lung cancer; ovarian cancer; endometrial cancer; pancreatic cancer; pituitary adenoma; prostate cancer; renal cancer; metastatic melanoma or thyroid cancers. In embodiments, the disease or the condition is melanoma, non-small cell cancer, colorectal cancer, ovarian cancer, thyroid cancer, breast cancer or cholangiocarcinoma. In embodiments, the disease or the condition is colorectal cancer. In embodiments, the disease or the condition is melanoma. In embodiments, the disease or the condition is cancer comprising a BRAFV600Emutation. In embodiments, the disease or the condition is modulated by BRAFV600EIn embodiments, the disease or the condition is BRAFV600Emelanoma, BRAFV600Ecolorectal cancer, BRAFV600Epapillary thyroid cancers, BRAFV600Elow grade serous ovarian cancers, BRAFV600Eglioma, BRAFV600Ehepatobiliary cancers, BRAFV600Ehairy cell leukaemia, BRAFV600Enon-small cell cancer, or BRAFV600Epilocytic astrocytoma. In embodiments, the disease or the condition is cardio-facio cutaneous syndrome and polycystic kidney disease. Pharmaceutical Compositions The present disclosure also relates to pharmaceutical compositions comprising the compounds of formula (I), (I-A), (I-B), (II), or (III), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, and a pharmaceutically acceptable carrier or excipient. The present disclosure also relates to pharmaceutical compositions comprising the compounds of Tables A1-A3 and B-D, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, and a pharmaceutically acceptable carrier or excipient. In embodiments, the pharmaceutical composition may further comprise an additional pharmaceutically active agent. The additional pharmaceutically active agent may be an anti-tumor agent. In embodiments, the additional pharmaceutically active agent is an antiproliferative/antineoplastic drug. In embodiments, antiproliferative/antineoplastic drug is alkylating agent (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, bendamustin, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolite (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, pemetrexed, cytosine arabinoside, and hydroxyurea); antibiotic (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agent (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like TAXOL® (paclitaxel) and taxotere and polokinase inhibitors); proteasome inhibitor, for example carfilzomib and bortezomib; interferon therapy; or topoisomerase inhibitor (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan, mitoxantrone and camptothecin). In embodiments, the additional pharmaceutically active agent is a cytostatic agent. In embodiments, cytostatic agent is antiestrogen (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogen (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonist or LHRH agonist (for example goserelin, leuprorelin and buserelin), progestogen (for example megestrol acetate), aromatase inhibitor (for example as anastrozole, letrozole, vorazole and exemestane) or inhibitor of 5a-reductase such as finasteride. In embodiments, the additional pharmaceutically active agent is an anti-invasion agent. In embodiments, the anti-invasion agent is dasatinib and bosutinib (SKI-606), metalloproteinase inhibitor, or inhibitor of urokinase plasminogen activator receptor function or antibody to Heparanase. In embodiments, the additional pharmaceutically active agent is an inhibitor of growth factor function. In embodiments, the inhibitor of growth factor function is growth factor antibody and growth factor receptor antibody, for example the anti-erbB2 antibody trastuzumab [Herceptin™], the anti-EGFR antibody panitumumab, the anti-erbB1 antibody cetuximab, tyrosine kinase inhibitor, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitor such as gefitinib, erlotinib and 6-acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3-morpholinopropoxy)-quinazolin-4-amine (CI 1033), erbB2 tyrosine kinase inhibitor such as lapatinib); inhibitor of the hepatocyte growth factor family; inhibitor of the insulin growth factor family; modulator of protein regulators of cell apoptosis (for example Bcl-2 inhibitors); inhibitor of the platelet-derived growth factor family such as imatinib and/or nilotinib (AMN107); inhibitor of serine/threonine kinases (for example Ras/RAF signalling inhibitors such as famesyl transferase inhibitor, for example sorafenib, tipifarnib and lonafarnib), inhibitor of cell signalling through MEK and/or AKT kinase, c-kit inhibitor, abl kinase inhibitor, Pl3 kinase inhibitor, Plt3 kinase inhibitor, CSF-1R kinase inhibitor, IGF receptor, kinase inhibitor; aurora kinase inhibitor or cyclin dependent kinase inhibitor such as CDK2 and/or CDK4 inhibitor. In embodiments, the additional pharmaceutically active agent is an antiangiogenic agent. In embodiments, the antiangiogenic agent inhibits the effects of vascular endothelial growth factor, for example the anti-vascular endothelial cell growth factor antibody bevacizumab (Avastin™); thalidomide; lenalidomide; and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib, vatalanib, sunitinib, axitinib and pazopanib. In embodiments, the additional pharmaceutically active agent is a cIn embodiments, the cytotoxic agent is fludaribine (fludara), cladribine, or pentostatin (Nipent™) In embodiments, the additional pharmaceutically active agent is a steroid. In embodiments, the steroid is corticosteroid, including glucocorticoid and mineralocorticoid, for example aclometasone, aclometasone dipropionate, aldosterone, amcinonide, beclomethasone, beclomethasone dipropionate, betamethasone, betamethasone dipropionate, betamethasone sodium phosphate, betamethasone valerate, budesonide, clobetasone, clobetasone butyrate, clobetasol propionate, cloprednol, cortisone, cortisone acetate, cortivazol, deoxycortone, desonide, desoximetasone, dexamethasone, dexamethasone sodium phosphate, dexamethasone isonicotinate, difluorocortolone, fluclorolone, flumethasone, flunisolide, fluocinolone, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluorocortisone, fluorocortolone, fluocortolone caproate, fluocortolone pivalate, fluorometholone, fluprednidene, fluprednidene acetate, flurandrenolone, fluticasone, fluticasone propionate, halcinonide, hydrocortisone, hydrocortisone acetate, hydrocortisone butyrate, hydrocortisone aceponate, hydrocortisone buteprate, hydrocortisone valerate, icomethasone, icomethasone enbutate, meprednisone, methylprednisolone, mometasone paramethasone, mometasone furoate monohydrate, prednicarbate, prednisolone, prednisone, tixocortol, tixocortol pivalate, triamcinolone, triamcinolone acetonide, triamcinolone alcohol and their respective pharmaceutically acceptable derivatives. A combination of steroids may be used, for example a combination of two or more steroids as described herein. In embodiments, the additional pharmaceutically active agent is a targeted therapy agent. In embodiments, the targeted therapy agent is a PI3 Kd inhibitor, for example idelalisib and perifosine. In embodiments, the additional pharmaceutically active agent is an immunotherapeutic agent. In embodiments, the immunotherapeutic agent is antibody therapy agent such as alemtuzumab, rituximab, ibritumomab tiuxetan (Zevalin®) and ofatumumab; interferon such as interferon α; interleukins such as IL-2 (aldesleukin); interleukin inhibitors for example IRAK4 inhibitors; cancer vaccine including prophylactic and treatment vaccines such as HPV vaccines, for example Gardasil, Cervarix, Oncophage and Sipuleucel-T (Provenge); toll-like receptor modulator for example TLR-7 or TLR-9 agonist; and PD-1 antagonist, PDL-1 antagonist, and IDO-1 antagonist. In embodiments, the pharmaceutical composition may be used in combination with another therapy. In embodiments, the other therapy is gene therapy, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2. In embodiments, the other therapy is immunotherapy approaches, including for example antibody therapy such as alemtuzumab, rituximab, ibritumomab tiuxetan (Zevalin®) and ofatumumab; interferons such as interferon α; interleukins such as IL-2 (aldesleukin); interleukin inhibitors for example IRAK4 inhibitors; cancer vaccines including prophylactic and treatment vaccines such as HPV vaccines, for example Gardasil, Cervarix, Oncophage and Sipuleucel-T (Provenge); toll-like receptor modulators for example TLR-7 or TLR-9 agonists; and PD-1 antagonists, PDL-1 antagonists, and IDO-1 antagonists. Compounds of the invention may exist in a single crystal form or in a mixture of crystal forms or they may be amorphous. Thus, compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, or spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose. For the above-mentioned compounds of the invention the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. For example, if the compound of the invention is administered orally, then the daily dosage of the compound of the invention may be in the range from 0.01 micrograms per kilogram body weight (μg/kg) to 100 milligrams per kilogram body weight (mg/kg). A compound of the invention, or pharmaceutically acceptable salt thereof, may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the compounds of the invention, or pharmaceutically acceptable salt thereof, is in association with a pharmaceutically acceptable adjuvant, diluent or carrier. Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, “Pharmaceuticals—The Science of Dosage Form Designs”, M. E. Aulton, Churchill Livingstone, 1988. Depending on the mode of administration of the compounds of the invention, the pharmaceutical composition which is used to administer the compounds of the invention will preferably comprise from 0.05 to 99% w (percent by weight) compounds of the invention, more preferably from 0.05 to 80% w compounds of the invention, still more preferably from 0.10 to 70% w compounds of the invention, and even more preferably from 0.10 to 50% w compounds of the invention, all percentages by weight being based on total composition. The pharmaceutical compositions may be administered topically (e.g. to the skin) in the form, e.g., of creams, gels, lotions, solutions, suspensions, or systemically, e.g. by oral administration in the form of tablets, capsules, syrups, powders or granules; or by parenteral administration in the form of a sterile solution, suspension or emulsion for injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion); by rectal administration in the form of suppositories; or by inhalation in the form of an aerosol. For oral administration the compounds of the invention may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets. If coated tablets are required, the cores, prepared as described above, may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide. Alternatively, the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent. For the preparation of soft gelatine capsules, the compounds of the invention may be admixed with, for example, a vegetable oil or polyethylene glycol. Hard gelatine capsules may contain granules of the compound using either the above-mentioned excipients for tablets. Also liquid or semisolid formulations of the compound of the invention may be filled into hard gelatine capsules. Liquid preparations for oral application may be in the form of syrups or suspensions, for example, solutions containing the compound of the invention, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol. Optionally such liquid preparations may contain colouring agents, flavouring agents, sweetening agents (such as saccharine), preservative agents and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in art. For intravenous (parenteral) administration the compounds of the invention may be administered as a sterile aqueous or oily solution. Pharmaceutical compositions can be prepared as liposome and encapsulation therapeutic agents. For various methods of preparing liposomes and encapsulation of therapeutic agents: see, for example, U.S. Pat. Nos. 3,932,657, 4,311,712, 4,743,449, 4,452,747, 4,830,858, 4,921,757, and 5,013,556. Known methods include the reverse phase evaporation method as described in U.S. Pat. No. 4,235,871. Also, U.S. Pat. No. 4,744,989 covers use of, and methods of preparing, liposomes for improving the efficiency or delivery of therapeutic compounds, drugs and other agents. Compounds of the invention can be passively or actively loaded into liposomes. Active loading is typically done using a pH (ion) gradient or using encapsulated metal ions, for example, pH gradient loading may be carried out according to methods described in U.S. Pat. Nos. 5,616,341, 5,736,155, 5,785,987, and 5,939,096. Also, liposome loading using metal ions may be carried out according to methods described in U.S. Pat. Nos. 7,238,367, and 7,744,921. Inclusion of cholesterol in liposomal membranes has been shown to reduce release of drug and/or increase stability after intravenous administration (for example, see: U.S. Pat. Nos. 4,756,910, 5,077,056, and 5,225,212). Inclusion of low cholesterol liposomal membranes continuing charged lipids has been shown to provide cryostability as well as increase circulation after intravenous administration (see: U.S. Pat. No. 8,518,437). Pharmaceutical compositions can comprise nanoparticles. The formation of nanoparticles has been achieved by various methods. Nanoparticles can be made by precipitating a molecule in a water-miscible solvent, and then drying and pulverizing the precipitate to form nanoparticles. (U.S. Pat. No. 4,726,955). Similar techniques for preparing nanoparticles for pharmaceutical preparations include wet grinding or milling. Other methods include mixing low concentrations of polymers dissolved in a water-miscible solution with an aqueous phase to alter the local charge of the solvent and form a precipitate through conventional mixing techniques. (U.S. Pat. No. 5,766,635). Other methods include the mixing of copolymers in organic solution with an aqueous phase containing a colloid protective agent or a surfactant for reducing surface tension. Other methods of incorporating additive therapeutic agents into nanoparticles for drug delivery require that nanoparticles be treated with a liposome or surfactant before drug administration (U.S. Pat. No. 6,117,454). Nanoparticles can also be made by flash nanoprecipitation (U.S. Pat. No. 8,137,699). U.S. Pat. No. 7,850,990 covers methods of screening combinations of agents and encapsulating the combinations in delivery vehicles such as liposomes or nanoparticles. The size of the dose for therapeutic purposes of compounds of the invention will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well-known principles of medicine. Dosage levels, dose frequency, and treatment durations of compounds of the invention are expected to differ depending on the formulation and clinical indication, age, and co-morbid medical conditions of the patient. The standard duration of treatment with compounds of the invention is expected to vary between one and seven days for most clinical indications. It may be necessary to extend the duration of treatment beyond seven days in instances of recurrent infections or infections associated with tissues or implanted materials to which there is poor blood supply including bones/joints, respiratory tract, endocardium, and dental tissues. EXAMPLES The disclosure now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention. As used herein the following terms have the meanings given: “Boc” refers to tert-butyloxycarbonyl; “Cbz” refers to carboxybenzyl; “dba” refers to dibenzylideneacetone; “DCM” refers to dichloromethane; “DIPEA” refers to N,N-diisopropylethylamine; “DMA” refers to dimethylacetamide; “DMF” refers to N,N-dimethylformamide; “DMSO” refers to dimethyl sulfoxide; “dppf” refers to 1,1′-bis(diphenylphosphino)ferrocene; “EtOAc” refers to ethyl acetate; “EtOH” refers to ethanol; “Et2O” refers to diethyl ether; “IPA” refers to isopropyl alcohol; “LiHMDS” refers to lithium bis(trimethylsilyl)amide; “mCPBA” refers to meta-chloroperoxybenzoic acid; “MeCN” refers to acetonitrile; “MeOH” refers to methanol; “min” refers to minutes; “NMR” refers to nuclear magnetic resonance; “PhMe” refers to toluene; “pTsOH” refers to p-toluenesulfonic acid; “py” refers to pyridine; “r.t.” refers to room temperature; “SCX” refers to strong cation exchange; “T3P” refers to propylphosphonic anhydride; “Tf2O” refers to trifluoromethanesulfonic anhydride; “THF” refers to tetrahydrofuran; “THP” refers to 2-tetrahydropyranyl; “(UP)LC-MS” refers to (ultra performance) liquid chromatography/mass spectrometry. Solvents, reagents and starting materials were purchased from commercial vendors and used as received unless otherwise described. All reactions were performed at room temperature unless otherwise stated. Compound identity and purity confirmations were performed by LC-MS UV using a Waters Acquity SQ Detector 2 (ACQ-SQD2 #LCA081). The diode array detector wavelength was 254 nM and the MS was in positive and negative electrospray mode (m/z: 150-800). A 2 μL aliquot was injected onto a guard column (0.2 μm×2 mm filters) and UPLC column (C18, 50×2.1 mm, <2 μm) in sequence maintained at 40° C. The samples were eluted at a flow rate of 0.6 mL/min with a mobile phase system composed of A (0.1% (v/v) formic acid in water) and B (0.10% (v/v) formic acid in MeCN) according to the gradients outlined below. Retention times RT are reported in minutes. Time (min)% A% BFinal purity09551.19556.159575957.59558955Short acidic09550.395525952.69553955 NMR was also used to characterise final compounds. NMR spectra were obtained on a Bruker AVIII 400 Nanobay with 5 mm BBFO probe. Optionally, compound Rf values on silica thin layer chromatography (TLC) plates were measured. Compound purification was performed by flash column chromatography on silica or by preparative LC-MS. LC-MS purification was performed using a Waters 3100 Mass detector in positive and negative electrospray mode (m z: 150-800) with a Waters 2489 UV/Vis detector. Samples were eluted at a flow rate of 20 mL/min on a Xbridgerm prep C18 5 μM OBD 19×100 mm column with a mobile phase system composed of A (0.1% (v/v) formic acid in water) and B (0.1% (v/v) formic acid in MeCN) according to the gradient outlined below: Time (min)% A% B090101.5901011.759513.7595149090159090 Chemical names in this document were generated using mol2nam—Structure to Name Conversion by OpenEye Scientific Software. Starting materials were purchased from commercial sources or synthesised according to literature procedures. Synthesis of Common Intermediates Example 1. Synthesis of 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one Step 1—tert-butyl N-(4-fluoro-2-pyridyl)carbamate 2-Amino-4-fluoropyridine (400 g, 3.57 mol) was charged into a 10 L fixed reactor vessel and then taken up in DCM (4 L) under a nitrogen atmosphere. DMAP (43.6 g, 357 mmol) was added and the reaction cooled to 10° C. Boc2O (934 g, 4.2 mol) was added as a solution in DCM (1 L) over 1.5 hours. The reaction was stirred at rt for 2 hours. The reaction was then treated with N,N-dimethylethylenediamine (390 mL, 3.57 mmol) and heated to 40° C. overnight. The reaction was cooled to rt, diluted with DCM (2 L) and washed with water. The aqueous layer was extracted with further DCM (2 L) and the organics washed with water, brine and dried over MgSO4. During evaporation of the solvent, a solid crashed out, which was filtered, washed with petroleum ether to give tert-butyl N-(4-fluoro-2-pyridyl)carbamate (505 g, 2.38 mol, 67% yield) as a cream solid product. UPLC-MS (ES+, Short acidic): 1.64 min, m/z 213.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.13 (1H, d, J=1.7 Hz), 8.26 (1H, dd, J=9.4 Hz, 5.7 Hz), 7.60 (1H, dd, J=12.3 Hz, 2.4 Hz), 6.94 (1H, ddd, J=8.2 Hz, 5.7 Hz, 2.4 Hz), 1.47 (9H, s). Step 2—tert-butyl N-(4-fluoro-3-iodo-2-pyridyl)carbamate Tert-butyl N-(4-fluoro-2-pyridyl)carbamate (126 g, 593.7 mmol) and N,N,N,N-tetramethylethylenediamine (223 mL, 1.48 mol) were taken up in dry THF (1.7 L) and then cooled to −78° C. under a nitrogen atmosphere. To this solution was added n-BuLi (2.5M in hexane—285 mL, 712.5 mmol) and then allowed to stir for a further 10 minutes. sec-BuLi (1.2M in cyclohexane—509 mL, 712.5 mmol) was then added keeping the reaction temperature below −70° C. and stirred for 1 hour. After this time, iodine (226 g, 890.6 mmol) in THF (300 mL) was added dropwise over 30 minutes and the temperature kept below −65° C. The reaction was stirred at −70° C. for another 10 minutes then quenched by the addition of sat. aq. NH4Cl (400 mL) and then a solution of sodium thiosulphate (134.1 g, 848.2 mmol) dissolved in water (600 mL) which raised the temperature to −25° C. The reaction was warmed to rt, transferred to a 5 L separator and extracted with EtOAc (2×1.5 L) and then washed with brine (500 ml). The organic phase was dried over MgSO4and the solvent removed in vacuo. The residue was taken up in DCM (500 mL) and passed through a 2 Kg silica pad, which was washed with DCM (10×1 L) and then the product was eluted from the column using as eluent a gradient 10-100% EtOAc in petroleum ether (1 L at each 10% increase) tert-butyl N-(4-fluoro-3-iodo-2-pyridyl)carbamate (154.6 g, 457.1 mmol, 77% yield) as a white solid. UPLC-MS (ES+, Short acidic): 1.60 min, m/z 339.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 9.47 (1H, s), 8.33 (1H, dd, J=8.7 Hz, 5.5 Hz), 7.19 (1H, dd, J=7.3 Hz, 5.5 Hz), 1.46 (9H, s). Step 3—5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one tert-butyl N-(4-fluoro-3-iodo-2-pyridyl)carbamate (263.3 g, 778.72 mmol) and 3,3-dimethoxyprop-1-ene (120 mL, 1.01 mol) and DIPEA (285 mL, 1.64 mol) were taken up in DMF (2.2 L) and water (440 mL) and degassed under a nitrogen atmosphere for 20 minutes. To this mixture, palladium (II) acetate (17.5 g, 77.9 mmol) was added, the reaction degassed for 15 minutes and then heated to 110° C. for 18 hours. The reaction was cooled to rt and filtered through celite. The solvent was removed under reduce pressure, the residue was taken up in water and acidified to pH-1-2 with 2N HCl solution. It was then basified to pH-9 with solid NaHCO3solution followed by extraction with DCM (2×2 L). The organic phases were combined, washed with water, brine and dried over MgSO4. EtOAc (2 L) was added to the solution and the organics were passed through a silica plug eluting with 40% EtOAc in DCM. Fractions containing the product were combined and the solvent removed in vacuo to give a solid, which was slurried in cold Et20 (300 mL) and filtered. The solid was washed with Et20 and then petroleum ether, pulled dry to give 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (5.7 mg, 343.1 mmol, 44% yield) as a pale yellow solid. UPLC-MS (ES+, Short acidic): 1.04 min, m/z 167.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.69 (1H, s), 8.29-7.90 (1H, m), 6.92 (1H, dd, J=8.8 Hz, 5.7 Hz, 1H), 2.88 (2H, dd, J=8.3 Hz, 7.1 Hz), 2.57-2.47 (2H, m). Example 2. Synthesis of 6-hydroxychromane-3-carboxylic Acid Step 1—2-hydroxy-5-tetrahydropyran-2-yloxy-benzaldehyde To a solution of 2,5-dihydroxybenzaldehyde (400 g, 2.90 mol) and pyridinium p-toluenesulfonate (36.4 g, 144.8 mmol) in DCM (7.5 L) was added 3,4-dihydro-2H-pyran (396.3 mL, 4.34 mol) dropwise over 10 minutes and the reaction stirred at 30° C. overnight. The reaction was washed with water (1.5 L), the organic layer separated and passed through a 1.5 Kg silica pad, which was washed with DCM (2.5 L), 25% EtOAc in petroleum ether (2.5 L) and finally 50% EtOAc in petroleum ether (2.5 L). Fractions containing the product were combined and the solvent removed in vacuo. The residue was slowly diluted with petroleum ether (1.75 L) and cooled to 10° C. to give a thick slurry. The product was filtered, washed with petroleum ether (2×150 mL) and dried to give 2-hydroxy-5-tetrahydropyran-2-yloxy-benzaldehyde (570.7 g, 2.57 mol, 89% yield) as a yellow solid. UPLC-MS (ES+, Short acidic): 1.64 min, m/z 223.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.35 (1H, s), 10.23 (1H, s), 7.32-7.19 (2H, m), 6.94 (1H, d, J=8.9 Hz), 5.36 (1H, t, J=3.3 Hz), 3.77 (1H, ddd, J=11.2 Hz, 8.8 Hz, 3.6 Hz), 3.59-3.49 (1H, m), 1.94-1.45 (6H, m). Step 2—tert-butyl 6-tetrahydropyran-2-yloxy-2H-chromene-3-carboxylate 2-hydroxy-5-tetrahydropyran-2-yloxy-benzaldehyde (107 g, 481.5 mmol) was dissolved in diglyme (750 mL) and K2CO3(133 g, 962.9 mmol) was added. The reaction was then heated to 140° C. and tert-butyl acrylate (155 mL, 1059.2 mmol) in DMF (75 mL) was added over 10 minutes at −110° C.; the reaction was stirred at 140° C. for a further 5 hour. The reaction was cooled to rt overnight, filtered and the solvent removed in vacuo. The reaction was suspended in EtOAc (2.5 L) and water (2.5 L) and the phases separated. The aqueous phase was extracted with EtOAc (2.5 L) and the combined organic layers were washed with brine (50%) and the solvent removed in vacuo. The crude was dissolved in DCM, loaded onto a 2 Kg silica pad, which was flushed with a gradient from 10-25% EtOAc in petroleum ether to give tert-butyl 6-tetrahydropyran-2-yloxy-2H-chromene-3-carboxylate (122.5 g, 368.5 mmol, 77% yield) as a yellow solid. UPLC-MS (ES+, Short acidic): 2.18 min, m/z nd.1H NMR (400 MHz, DMSO-d6) δ/ppm: 7.38 (1H, s), 7.05 (1H, d, J=2.9 Hz), 6.94 (1H, dd, J=8.8, 2.9 Hz), 6.79 (1H, dd, J=8.8, 0.7 Hz), 5.35 (1H, t, J=3.3 Hz), 4.82 (2H, d, J=1.4 Hz), 3.78 (1H, ddd, J=11.8, 8.6, 3.6 Hz), 3.58-3.49 (1H, m), 1.92-1.66 (3H, m), 1.66-1.52 (3H, m), 1.49 (s, 9H). Step 3—tert-butyl 6-hydroxy-2H-chromene-3-carboxylate tert-Butyl 6-tetrahydropyran-2-yloxy-2H-chromene-3-carboxylate (110.4 g, 332.1 mmol) was suspended in MeOH (1.2 L) at rt and pyridinium p-toluenesulfonate (8.35 g, 33.2 mmol) was added. The reaction was warmed to 34° C. for 3 hours. The solvent was removed in vacuo, the crude dissolved in EtOAc (1 L) and washed with water (750 mL). The organic layer was dried over MgSO4, filtered and the solvent removed under reduce pressure to give a yellow solid. This solid was suspended in petroleum ether and the solvent removed in vacuo to give tert-butyl 6-hydroxy-2H-chromene-3-carboxylate (82 g, 330.3 mmol, 99% yield) as a yellow solid. UPLC-MS (ES−, Short acidic): 1.71 min, m/z 247.2 [M−H]−. 1H NMR (400 MHz, DMSO-d6) δ/ppm: 9.17 (1H, s), 7.33 (1H, s), 6.76-6.64 (3H, m), 4.77 (2H, d, J=1.4 Hz), 1.49 (9H, s). Step 4—tert-butyl 6-hydroxychromane-3-carboxylate tert-Butyl 6-hydroxy-2H-chromene-3-carboxylate (177.3 g, 714.1 mmol) was suspended in MeOH (2.5 L) at rt and palladium, 10 wt. % on carbon powder, 50% wet (15.2 g, 142.8 mmol) added. The reaction was fitted with a H2balloon, extra H2added and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 5 hours. The crude was filtered over celite, washed with MeOH and the filtrate concentrated in vacuo to give tert-butyl 6-hydroxychromane-3-carboxylate (171 g, 683.2 mmol, 96% yield) as a pale cream solid. UPLC-MS (ES+, Short acidic): 1.64 min, m/z nd.1H NMR (400 MHz, DMSO-d6) δ/ppm: 8.81 (1H, s), 6.60-6.52 (1H, m), 6.47 (2H, d, J=7.7 Hz), 4.19 (1H, dd, J=10.6 Hz, 3.0 Hz), 3.96 (1H, dd, J=10.6 Hz, 7.4 Hz), 2.96-2.73 (3H, m), 1.40 (9H, s). Step 5-6-hydroxychromane-3-carboxylic Acid tert-Butyl 6-hydroxychromane-3-carboxylate (23 g, 91.9 mmol) was dissolved in DCM (250 mL) and TFA (48.3 mL, 630.35 mmol) added and the reaction stirred at rt overnight. Solvents were removed in vacuo and azeotroped with toluene (100 mL); the residue was slurried with Et20 and filtered to give 6-hydroxychromane-3-carboxylic acid (16.8 g, 86.5 mmol, 94% yield) as a pink solid. UPLC-MS (ES+, Short acidic): 1.08 min, m/z 194.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.60 (1H, s), 8.81 (1H, s), 6.60-6.53 (1H, m), 6.52-6.45 (2H, m), 4.22 (1H, dd, J=10.7 Hz, 3.0 Hz), 4.06-3.96 (1H, m), 2.97-2.77 (3H, m). Example 3. Synthesis of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic Acid 6-hydroxychromane-3-carboxylic acid (45.3 g, 233.3 mmol), 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (38.8 g, 233.3 mmol) were suspended in DMSO (230 mL) and K2CO3(119.7 g, 865.8 mmol) was added in portions. The reaction was heated to 98° C. for 48h. It was then cooled to 60° C., diluted with water (2 L) and extracted with EtOAc (750 mL). The aqueous layer was separated and slowly added to a solution of citric acid (179.3 g, 933.2 mmol) in water (400 mL) to give a cream solid, which was filtered, washed with water/acetone (1:1) and Et20 and dried to give 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (74.7 g, 219.5 mmol, 94% yield) as a cream solid. UPLC-MS (ES+, Short acidic): 1.26 min, m/z 341.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.69 (1H, s), 10.46 (1H, s), 7.95 (1H, d, J=5.8 Hz), 6.97 (1H, d, J=2.8 Hz), 6.89 (1H, dd, J=8.8 Hz, 2.8 Hz), 6.84 (1H, t, J=8.8 Hz), 6.24 (1H, d, J=5.8 Hz), 4.33 (1H, dd, J=10.9 Hz, 3.1 Hz), 4.20-4.10 (1H, m), 3.06-2.95 (3H, m), 2.92 (2H, t, J=8.4 Hz, 7.0 Hz), 2.59-2.51 (2H, m). Example 4. Synthesis of 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol and 5-[3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one Step 1—methyl 6-hydroxy-2H-chromene-3-carboxylate To 6-hydroxy-2H-chromene-3-carboxylic acid (16 g, 83.3 mmol) in MeOH (200 mL) was added sulfuric acid (0.44 mL, 8.33 mmol) and the reaction heated to 70° C. until completion. The solvent was removed in vacuo and the residue dissolved in DCM and washed with water. The organic layer was separated and passed through a 100 g silica pad eluting with DCM (500 mL) and then with 50% EtOAc in petroleum ether (2×500 mL). The solid obtained was slurried with petroleum ether, filtering and dried to give methyl 6-hydroxy-2H-chromene-3-carboxylate (14.1 g, 68.4 mmol, 82% yield) as a yellow solid. UPLC-MS (ES+, Short acidic): 1.38 min, m/z 207.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 9.19 (1H, s), 7.46 (1H, d, J=1.4 Hz), 6.79-6.65 (2H, m), 6.70 (1H, s), 4.82 (2H, d, J=1.4 Hz), 3.75 (3H, s). Step 2—methyl 6-hydroxychromane-3-carboxylate methyl 6-hydroxy-2H-chromene-3-carboxylate (53.3 g, 258.3 mmol) was suspended in MeOH (600 mL) at rt and palladium, 10 wt. % on carbon powder, 50% wet (2.75 g, 25.8 mmol) was added under a nitrogen atmosphere. The reaction was fitted with a H2balloon, extra H2added and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 3 hours. The crude was filtered over celite, washed with MeOH and the solvent removed in vacuo to give methyl 6-hydroxychromane-3-carboxylate (50.1 g, 233.9 mmol, 91% yield) as a cream solid. UPLC-MS (ES+, Short acidic): 1.27 min, m/z 209.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 8.83 (1H, s), 6.56 (1H, dd, J=8.1, 1.0 Hz), 6.48 (2H, dd, J=9.7 Hz, 1.8 Hz), 4.22 (1H, dd, J=10.8 Hz, 3.2 Hz), 4.04 (1H, dd, J=10.7 Hz, 7.6 Hz), 3.64 (3H, s), 3.10-2.98 (1H, m), 2.89 (2H, d, J=6.9 Hz). Step 3—methyl 6-benzyloxychromane-3-carboxylate Benzyl bromide (2.85 mL, 23.97 mmol) was slowly added to a stirred mixture of methyl 6-hydroxychromane-3-carboxylate (4.16 g, 19.98 mmol), K2CO3(8.28 g, 59.93 mmol) and DMF (100 mL) at rt under a nitrogen atmosphere. The reaction was heated to 50° C. and stirred for 1.5 hour, after which time it was cooled to rt and the solvent removed in vacuo. The residue was partitioned between water (300 mL) and DCM (300 mL). The organic layer was separated and the aqueous extracted with DCM (300 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-50% EtOAc in petroleum ether to give methyl 6-benzyloxychromane-3-carboxylate (5.9 g, 19.78 mmol, 99% yield) as a colourless oil which solidified to a white solid on standing. UPLC-MS (ES+, short acidic): 1.92 min, m/z 299.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.46-7.32 (5H, m), 6.79-6.77 (2H, m), 6.73 (1H, d, J=7.2 Hz), 5.02 (2H, s), 4.44-4.39 (1H, m), 4.14-4.08 (1H, m), 3.75 (3H, s), 3.14-2.97 (3H, m). Step 4-6-benzyloxychromane-3-carboxylic Acid Methyl 6-benzyloxychromane-3-carboxylate (5.9 g, 19.78 mmol) was stirred in a solution of aq. NaOH (100 mL, 100 mmol) and THF (100 mL) at rt for 18 hours. The organic solvent was removed in vacuo and the resulting mixture stirred, cooled in ice, followed by acidification to ˜pH 5 using conc. HCl. The resulting solid was filtered off and dried to give 6-benzyloxychromane-3-carboxylic acid (5.1 g, 17.94 mmol, 91% yield) as a white solid. UPLC-MS (ES−, short acidic): 1.71 min, m/z 283.2 [M−H]−.1H NMR (400 MHz, CDCl3) δ/ppm: 12.64 (1H, br s), 7.46-7.37 (4H, m), 7.35-7.30 (1H, m), 6.80 (1H, d, J=2.8 Hz), 6.74 (1H, dd, J=9.2 Hz, 2.8 Hz), 6.67 (1H, d, J=9.2 Hz), 5.02 (2H, s), 4.27-4.22 (1H, m), 4.09-4.04 (1H, m), 2.99-2.91 (3H, m). Step 5—6-benzyloxychroman-3-yl)methanol Di-methylsulfide borane (13.45 mL, 26.91 mmol) was added to a stirred solution of 6-benzyloxychromane-3-carboxylic acid (5.1 g, 17.94 mmol) and THF (200 mL) at 0° C. under a nitrogen atmosphere and stirred at rt for 3 hours. The reaction was cooled to 0° C. and quenched carefully with water (500 mL). This was extracted with EtOAc (2×500 mL). the combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-10% EtOAc in petroleum ether to give 6-benzyloxychroman-3-yl)methanol (4.5 g, 16.65 mmol, 93% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.69 min, m/z 271.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.46-7.31 (5H, m), 6.78-6.75 (2H, m), 6.72-6.69 (1H, m), 5.01 (2H, s), 4.27 (1H, ddd, J=10.8 Hz, 2.8 Hz, 1.2 Hz), 3.99 (1H, dd, J=10.8 Hz, 2.8 Hz), 3.77-3.65 (2H, m), 2.92-2.85 (1H, m), 2.59 (1H, dd, J=16.8 Hz, 7.6 Hz), 2.33-2.23 (1H, m). Exchangeable proton not seen. Step 6-6-benzyloxychromane-3-carbaldehyde A solution of DMSO (1.77 mL, 24.97 mmol) in DCM (200 mL) was stirred under a nitrogen atmosphere at −78° C. Oxalyl chloride (2.11 mL, 24.97 mmol) was then added and left stirring for 20 min; a solution of (6-benzyloxychroman-3-yl)methanol in DCM (50 mL) was then added dropwise whilst maintaining the temperature below −70° C. Upon completion of addition, the reaction was stirred for 30 minutes and triethylamine (5.8 mL, 41.62 mmol) was added dropwise. The reaction was left stirring at −78° C. for 20 min after which time it was allowed to warm to rt. The reaction was diluted with water (200 mL) and the organic layer separated, passed through a phase separator and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-50% EtOAc in petroleum ether to give 6-benzyloxychromane-3-carbaldehyde (4.4 g, 16.4 mmol, 99% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.82 min, m/z 268.2 [M]+.1H NMR (400 MHz, CDCl3) δ/ppm: 9.86 (1H, s), 7.47-7.32 (5H, m), 6.81-6.74 (3H, m), 5.02 (2H, s), 4.42-4.32 (2H, m), 3.17-3.09 (1H, m), 3.04-2.93 (2H, m). Step 7—2-(6-benzyloxychroman-3-yl)-1H-imidazole Ammonium Hydroxide (25 mL, 750 mmol) was added to a stirred solution of 6-benzyloxychromane-3-carbaldehyde (3.9 g, 14.54 mmol), glyoxal (9.96 mL, 87.21 mmol) and MeOH (25 mL) at rt. After 1 hour, the reaction was diluted with water (200 mL) and extracted with DCM (2×200 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo to give 2-(6-benzyloxychroman-3-yl)-1H-imidazole (2.4 g, 7.83 mmol, 54% yield) as a yellow solid. UPLC-MS (ES+, short acidic), 1.28 min, m/z 307.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 7.46-7.30 (6H, m), 7.08 (1H, d, J=9.2 Hz), 6.83-6.80 (1H, d), 6.79-6.75 (1H, d), 6.71 (1H, d, J=8.8 Hz), 5.03 (2H, s), 4.40-4.35 (1H, m), 3.97 (1H, t, J=10.0 Hz), 3.31-3.26 (1H, m), 3.15-3.10 (1H, m), 3.05-2.97 (1H, m). Exchangeable proton not seen. Step 8—2-[[2-(6-benzyloxychroman-3-yl)imidazol-1-yl]methoxy]ethyl-trimethyl-silane Sodium hydride (60% dispersed in mineral oil—895.6 mg, 22.39 mmol) was added to a stirred solution of 2-(6-benzyloxychroman-3-yl)-1H-imidazole (3.43 g, 11.2 mmol) in DMF (5 mL) at rt under a nitrogen atmosphere. After 20 minutes, 2-(trimethylsilyl)ethoxymethyl chloride (2.97 mL, 16.79 mmol) was added. The reaction was allowed to stir for 1 hour, then quenched with water (1 mL) and solvent removed in vacuo. The residue was partitioned between water (20 mL) and DCM (20 mL). The organic layer was separated and the aqueous extracted with DCM (20 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-5% MeOH in DCM to give 2-[[2-(6-benzyloxychroman-3-yl)imidazol-1-yl]methoxy]ethyl-trimethyl-silane (4.3 g, 9.85 mmol, 88% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.67 min, m/z 437.7 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.45-7.29 (5H, m), 7.03-7.01 (1H, m), 6.99-6.97 (1H, m), 6.80-6.78 (2H, m), 6.73-6.71 (1H, m), 5.31-5.28 (2H, m), 5.01 (2H, s), 4.44-4.38 (1H, m), 4.12 (1H, t, J=10.4 Hz), 3.55-3.50 (2H, m), 3.48-3.33 (2H, m), 2.99-2.92 (1H, m), 0.96-0.87 (2H, m), 0.00 (9H, s). Step 9—3-[1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol Palladium, 10 wt. % on carbon powder, dry (400 mgl) was added to a stirred solution of 2-[[2-(6-benzyloxychroman-3-yl)imidazol-1-yl]methoxy]ethyl-trimethyl-silane (4.3 g, 9.85 mmol) in MeOH (50 mL) at rt. The reaction was fitted with a H2balloon and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 2 hours. The crude was filtered over celite, washed with MeOH and the filtrate concentrated in vacuo to give 3-[1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (3.32 g, 9.58 mmol, 97% yield) as a yellow solid. The compound was used directly in the next step without further purification. UPLC-MS (ES+, short acidic): 1.36 min, m/z 347.7 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 6.98 (1H, d, J=1.6 Hz), 6.99 (1H, d, J=1.6 Hz), 6.69 (1H, d, J=8.8 Hz), 6.63 (1H, dd, J=8.8 Hz, 3.2 Hz), 6.56 (1H, d, J=3.2 Hz), 5.32 (1H, d, J=10.8 Hz), 5.28 (1H, d, J=10.8 Hz), 4.33 (1H, ddd, J=10.8 Hz, 3.2 Hz, 2.4 Hz), 4.10 (1H, t, J=10.4 Hz), 3.55-3.50 (3H, m), 3.33-3.24 (1H, m), 2.94-2.87 (1H, m), 0.91-0.95 (2H, m), 0.10 (9H, s). Exchangeable proton not seen. Step 10—[3-[1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl] acetate Acetyl chloride (0.94 mL, 13.29 mmol) was added to a stirred solution of 2-(1-benzylimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-ol (3.07 g, 8.86 mmol), Et3N (1.85 mL, 13.29 mmol) and DCM (100 mL) at rt under a nitrogen atmosphere. The reaction was stirred for 30 minutes, after which time water (100 mL) was added. The mixture was extracted with DCM (2×100 mL). The combined organic extracts were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-100% EtOAc in petroleum ether to give [3-[1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl] acetate (2.93 g, 7.53 mmol, 85% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.48 min, m/z 389.5 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.07-6.99 (2H, m), 6.88-6.81 (3H, m), 5.33-5.27 (2H, m), 4.46-4.41 (1H, m), 4.25-4.18 (1H, m), 3.57-3.50 (2H, m), 3.48-3.38 (2H, m), 3.03-2.95 (1H, m), 2.25 (3H, s), 0.96-0.89 (2H, m), 0.00 (9H, s). Step 11—[3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]acetate N-Bromosuccinimide (0.46 g, 2.57 mmol) was added to a stirred solution of [3-[1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl] acetate (1 g, 2.57 mmol) and DCM (100 mL) at 0° C. under a nitrogen atmosphere. The reaction was stirred for 30 minutes and then diluted with water (100 mL). The organic layer was separated, dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-50% EtOAc in petroleum ether to give [3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl] acetate (684 mg, 1.46 mmol, 57% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 2.07 min, m/z 467.2, 469.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 6.98 (1H, s), 6.85-6.80 (3H, m), 5.33 (2H, s), 4.46-4.41 (1H, m), 4.15-4.07 (2H, m), 3.62-3.56 (1H, m), 3.51-3.41 (1H, m), 3.38-3.30 (1H, m), 3.00-2.94 (1H, m), 2.27 (3H, s), 0.96-0.89 (2H, m), 0.00 (9H, s). Step 12—3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol [3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl] acetate (800 mg, 1.71 mmol) was stirred in a mixture of aq. 1M NaOH (10 mL, 10 mmol) and THF (10 mL) at rt. After 30 minutes, the reaction was taken to ˜pH 5 using aq. 1M HCl and then it was diluted with water (50 ml) and extracted with DCM (2×50 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo to give 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (728 mg, 1.71 mmol, 100% yield) as a yellow oil. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidi): 1.87 min, m/z 425.1, 427.1 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 6.99 (1H, s), 6.75-6.73 (1H, m), 6.64-6.60 (1H, m), 6.58-6.56 (1H, m), 5.34 (2H, s), 4.80-4.71 (1H, m), 4.42-4.36 (1H, m), 4.06 (1H, t, J=10.4 Hz), 3.61-3.57 (2H, m), 3.51-3.40 (1H, m), 3.37-3.28 (1H, m), 2.95-2.88 (1H, m), 0.96-0.89 (2H, m), 0.00 (9H, s). Step 13—5-[3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (2 g, 4.7 mmol), 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (781.2 mg, 4.7 mmol), K2CO3(2.6 g, 18.81 mmol) and DMSO (5 mL) were combined and stirred at 110° C. under a nitrogen atmosphere for 1 hour. The reaction was cooled to rt and poured into a solution of water (100 mL) and citric acid monohydrate (3.95 g, 18.81 mmol). The resultant mixture was extracted with DCM (2×100 mL) and the combined organic layers dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-10% MeOH in DCM to give 5-[3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (230 mg, 0.40 mmol, 9% yield) as a yellow solid. UPLC-MS (ES+, short acidic), 1.96 min, m/z 571.2, 573.2 [M+H]+. Example 5. Synthesis of 6-[(2-chloro-4-pyridyl)oxy]chromane-3-carboxylic Acid and 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-carboxylic Acid Step 1—6-[2-chloro-4-pyridyl)oxy]chromane-3-carboxylic Acid A solution of 6-hydroxychromane-3-carboxylic acid hydrochloride (1.4 g, 6.07 mmol), 2-chloro-4-fluoropyridine (550 μL, 6.09 mmol) and K2CO3(3.36 g, 24.28 mmol) in DMSO (7.6 mL) was heated to 110° C. under a nitrogen atmosphere for 1.5 hours. The reaction mixture was cooled to rt and poured into a solution of citric acid (4.67 g, 24.28 mmol) in water (100 mL). The resulting precipitate was filtered, washed with water and dried to give 6-[(2-chloro-4-pyridyl)oxy]chromane-3-carboxylic acid (1.74 g, 5.69 mmol, 94% yield) as a brown solid. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.55 min, m/z 306.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.69 (1H, br s), 8.27 (1H, d, J=5.7 Hz), 7.04 (1H, d, J=2.9 Hz), 6.97-6.89 (3H, m), 6.86 (1H, d, J=8.8 Hz), 4.34 (1H, dd, J=10.9, 3.2 Hz), 4.19-4.13 (1H, m), 3.05-2.95 (3H, m). Step 2—6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-carboxylic Acid To a solution of 6-[(2-chloro-4-pyridyl)oxy]chromane-3-carboxylic acid (500 mg, 1.64 mmol), cyclopropanecarboxamide (278.4 mg, 3.27 mmol) and Cs2CO3(1.07 g, 3.27 mmol) in 1,4-dioxane (16 mL) was added (+/−)-BINAP (203.7 mg, 0.33 mmol) and tris(dibenzylideneacetone)dipalladium (0) (149.7 mg, 0.16 mmol) under a nitrogen atmosphere. The mixture was heated at 100° C. for 20 hrs. After cooling to rt, the mixture was filtered through celite and washed with methanol. The solvent was removed under reduce pressure and the residue purified by column chromatography using as eluent a gradient 0-20% MeOH in DCM. Fractions containing the product were combined, solvent removed in vacuo, the residue suspended in DCM and filtered. The precipitate was washed with DCM and dried to give 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-carboxylic acid (272.3 mg, 0.77 mmol, 47% yield) as an off-white solid. UPLC-MS (ES+, Short acidic): 1.23 min, m/z 355.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.69 (1H, br s), 10.81 (1H, s), 8.15 (1H, d, J=5.7 Hz), 7.63 (1H, d, J=2.4 Hz), 6.97 (1H, d, J=2.7 Hz), 6.88 (1H, dd, J=8.8 Hz, 2.7 Hz), 6.83 (1H, d, J=8.8 Hz), 6.59 (1H, dd, J=5.7 Hz, 2.4 Hz), 4.34 (1H, dd, J=10.7 Hz, 2.8 Hz), 4.14 (1H, dd, J=10.7 Hz, 6.7 Hz), 3.04-2.92 (3H, m), 1.97 (1H, quint, J=6.2 Hz), 0.77 (4H, d, J=6.2 Hz). Example 6. Synthesis of 7-benzyloxy-1,2,3,4-tetrahydroisoquinoline Hydrochloride Step 1—acetic acid; 1,2,3,4-tetrahydroisoquinolin-7-ol Platinum(IV) oxide (Adam's Catalyst) (500 mg, 2.2 mmol) was added to 7-hydroxyisoquinoline (5. g, 34.45 mmol) in acetic acid (20 mL) at rt. The reaction was fitted with a H2balloon and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 18 hours. The crude was filtered over celite and the filtrate concentrated in vacuo. The residue was stirred in an acetone/petroleum ether mixture (1:2, 15 mL) for 1 hour, causing a solid to crash out which was filtered off and dried in vacuo to give acetic acid; 1,2,3,4-tetrahydroisoquinolin-7-ol (6.53 g, 31.2 mmol, 91% yield) as a light yellow solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 0.36 min, m/z 150.2 [M+H]+. Step 2—benzyl 7-hydroxy-3,4-dihydro-1H-isoquinoline-2-carboxylate Benzyl chloroformate (2.05 mL, 14.34 mmol) was added to a stirred solution of THF (15 mL), acetic acid; 1,2,3,4-tetrahydroisoquinolin-7-ol (2.5 g, 11.95 mmol) and NaOH (35.84 mL, 35.84 mmol) at rt. The reaction was stirred for 1 hour and then diluted with water (200 mL) and EtOAc (200 mL). The organic layer was separated, dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give benzyl 7-hydroxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (3.3 g, 11.65 mmol, 97% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.65 min, m/z 284.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.40-7.29 (5H, m), 6.98 (1H, d, J=8.4 Hz), 6.70-6.54 (2H, m), 5.18 (2H, s), 4.61-4.56 (2H, m), 3.75-3.66 (2H, m), 2.81-2.73 (2H, m). Exchangeable proton not seen. Step 3—benzyl 7-benzyloxy-3,4-dihydro-1H-isoquinoline-2-carboxylate Benzyl 7-hydroxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (569 mg, 1.45 mmol) was dissolved in DMF (8 mL) followed by addition of benzyl bromide (0.21 mL, 1.74 mmol) and K2CO3(600 mg, 4.34 mmol). The reaction was left stirring at 50° C. for 1.5 hour and then at rt overnight. The solvent was removed under reduce pressure. Water was added, followed by extraction with EtOAc (2×). The organic layers were combined, washed with brine (2×), dried over Na2SO4and the solvent removed in vacuo. The crude was purified by column chromatography using as eluent a gradient 0-25% EtOAc in petroleum ether to give benzyl 7-benzyloxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (428 mg, 1.15 mmol, 79% yield) as a colourless oil. UPLC-MS (ES+, short acidic): 2.12 min, m/z 374.2 [M+H]+.1H-NMR (400 MHz, CDCl3) δ/ppm: 7.50-7.28 (10H, m), 7.04 (1H, d, J=8.4 Hz), 6.81 (1H, dd, J=8.4 Hz, 2.4 Hz), 6.72 (1H, br d, J=12.0 Hz), 5.18 (2H, s), 5.04 (2H, s), 4.61 (2H, s), 3.71 (2H, br s), 2.78 (2H, br s). Step 4—7-benzyloxy-1,2,3,4-tetrahydroisoquinoline Hydrochloride To benzyl 7-benzyloxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (196 mg, 0.52 mmol) was added HCl (4N in 1,4-dioxane—6 mL, 24 mmol) and the reaction heated to 80° C. for 72 hours. The solvent was removed under reduce pressure to give 7-benzyloxy-1,2,3,4-tetrahydroisoquinoline hydrochloride (144.7 mg, 0.52 mmol, 100% yield) as a cream solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.27 min, m/z 240.2 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 9.18 (1H, br s), 7.52-7.37 (4H, m), 7.36-7.27 (1H, m), 7.14 (1H, d, J=8.4 Hz), 6.96-6.89 (2H, m), 5.10 (2H, s), 4.21 (2H, br s), 3.35-3.28 (2H, t, (under water)), 2.92 (2H, t, J=6.0 Hz). Example 7. Synthesis of 3-methyl-5-(trifluoromethyl)benzene-1,2-diamine 3-Bromo-4,5-diaminobenzotrifluoride (500 mg, 1.96 mmol), K2CO3(542 mg, 3.92 mmol), tetrakis(triphenylphosphine)palladium(0) (227 mg, 0.20 mmol) were mixed under N2followed by the addition of 2,4,6-Trimethylboroxin (0.27 mL, 1.96 mmol). The reaction was left stirring for 72 hours at 110° C., after which time a second portion of 2,4,6-trimethylboroxin (0.14 mL, 0.98 mmol), K2CO3(271 mg, 1.96 mmol) and tetrakis(triphenylphosphine)palladium(0) (113 mg, 0.10 mmol) were added. The reaction was stirred at 110° C. overnight, after which time water was added (20 mL) followed by extraction with EtOAc (2×). The organic phases were combined, washed with brine, dried over Na2SO4, filtered and the solvent removed in vacuo. The crude was purified by column chromatography using as eluent a gradient 0-5% MeOH in DCM to give 3-methyl-5-(trifluoromethyl)benzene-1,2-diamine (82 mg, 0.43 mmol, 22% yield) as a brown oil. UPLC-MS (ES+, short acidic): 1.37 min, m/z 191.1 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 6.70-6.67 (1H, m), 6.63-6.59 (1H, m), 4.81 (2H, s), 4.79 (2H, s), 2.07 (3H, s). Example 8. Synthesis of 6-(trifluoromethyl)pyridine-3,4-diamine Step 1—5-nitro-2-(trifluoromethyl)pyridin-4-ol To a cooled solution of 2-(trifluoromethyl)pyridin-4-ol (1000 mg, 6.13 mmol) at 0° C. in sulfuric acid (2.46 mL, 46.15 mmol) was added nitric acid, fuming, 90% (6.12 mL, 144.04 mmol) dropwise over 15 min, after which time the reaction was heated to 120° C. overnight. The reaction mixture was cooled to rt and poured into ice water. The mixture was brought to neutral pH by addition of a solution 32% of aq. NaOH and extracted with EtOAc (3×) and n-BuOH. The combined organic phases were dried over Na2SO4, filtered and the solvent removed in vacuo to give 5-nitro-2-(trifluoromethyl)pyridin-4-ol (1.27 g, 6.10 mmol, 100% yield) as a yellow solid. The compound was used without further purification in the following step. UPLC-MS (ES−, short acidic): 1.17 min, m/z 207.1 [M−H]−.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 9.00 (1H, s), 7.30 (1H, s). Step 2—4-chloro-5-nitro-2-(trifluoromethyl)pyridine 5-nitro-2-(trifluoromethyl)pyridin-4-ol (1.28 g, 6.13 mmol), phosphorus pentachloride (1.91 g, 9.19 mmol) and phosphorus oxychloride (0.86 mL, 9.19 mmol) were stirred at 80° C. overnight. The mixture was cooled to rt, diluted with DCM and washed with water, sat. aq. Na2CO3and brine. The organic phase was dried over Na2SO4, filtered and the solvent removed in vacuo. The compound was re-dissolved in DCM and poured into water-ice, followed by addition of aq. 1M NaOH. The organic phase was washed with brine, dried over Na2SO4, filtered and the solvent removed under reduce pressure to give 4-chloro-5-nitro-2-(trifluoromethyl)pyridine (804 mg, 3.55 mmol, 58% yield) as a yellow oil. UPLC-MS (ES−, short acidic): 1.68 min, m/z nd.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 9.43 (1H, s), 8.58 (1H, s). Step 3—5-nitro-2-(trifluoromethyl)pyridin-4-amine 4-chloro-5-nitro-2-(trifluoromethyl)pyridine (752 mg, 3.32 mmol) was dissolved with ammonia in MeOH (30.08 mL, 69.19 mmol) and left stirring at rt for 3 hours. The solvent was removed in vacuo to give 5-nitro-2-(trifluoromethyl)pyridin-4-amine (687 mg, 3.32 mmol, 100% yield) as a yellow solid. The product was used without further purification in the following step. UPLC-MS (ES+, short acidic): 1.36 min, m/z 208.1 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 9.07 (1H, s), 7.44 (1H, s). Step 4—6-(trifluoromethyl)pyridine-3,4-diamine 5-nitro-2-(trifluoromethyl)pyridin-4-amine (734 mg, 3.54 mmol) was dissolved in EtOAc (15 mL) and Ethanol (25 mL) followed by the addition of palladium, 10 wt. % on carbon powder, (238 mg) under a nitrogen atmosphere. The reaction was fitted with a H2balloon and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 72 hours. A second portion of palladium, 10 wt. % on carbon powder, dry (238 mg) was added and left stirring overnight. The crude was filtered over celite, washed with EtOAc and the solvent removed under reduce pressure to give 6-(trifluoromethyl)pyridine-3,4-diamine (571 mg, 3.22 mmol, 91% yield) as a colourless oil.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 7.69 (1H, s), 6.83 (1H, s), 5.79 (2H, br s), 5.21 (2H, br s). Example 9. Synthesis of 5-[(dimethylamino)methyl]-3-(trifluoromethyl)benzene-1,2-diamine Step 1—4-chloro-3-nitro-5-(trifluoromethyl)benzoic Acid To 4-chloro-3-(trifluoromethyl)benzoic acid (1.5 g, 6.68 mmol) was added sulfuric acid (5.34 mL, 100.20 mmol) and nitric acid, fuming, 90% (0.85 mL, 20.04 mmol) and the reaction mixture was heated at 90° C. for 1 hour. The crude was poured in ice-cold water (150 mL), filtered and dried in vacuo to give 4-chloro-3-nitro-5-(trifluoromethyl)benzoic acid (1.66 g, 6.15 mmol, 92% yield) as a white solid. UPLC-MS (ES−, short acidic): 1.66 min, m/z 268.1 [M−H]−.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 14.22 (1H, br s), 8.78 (1H, d, J=2.0 Hz), 8.44 (1H, d, J=1.6 Hz). Step 2—[4-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol 4-chloro-3-nitro-5-(trifluoromethyl)benzoic acid (1.66 g, 6.15 mmol) was dissolved in THF (20 mL) and cooled to 0° C., followed by dropwise addition of di-methylsulfide borane (9.24 mL, 18.46 mmol). The reaction was stirred at rt for 72 hours, after which time it was quenched by slow addition of a sat. aq. NaHCO3and extracted with EtOAc (×2). The organic layers were combined, dried over Na2SO4, filtered and the solvent removed under reduce pressure to give 4-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol (1.31 g, 5.11 mmol, 83% yield) as a pale yellow oil.1H-NMR (400 MHz, CDCl3) δ/ppm: 7.94 (1H, s), 7.91 (1H, s), 4.84 (2H, s). Exchangeable proton missing. Step 3—5-(bromomethyl)-2-chloro-1-nitro-3-(trifluoromethyl)benzene A solution of [4-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol (1.31 g, 5.11 mmol) in DCM (15 mL) was cooled to 0° C. followed by the addition of triphenylphosphine (1.61 g, 6.13 mmol) and tetrabromomethane (1864.1 mg, 5.62 mmol). The reaction was left stirring at rt overnight, after which time it was washed with sat. aq. NaHCO3. The organic layer was separated, dried over Na2SO4and the solvent removed in vacuo to give 5-(bromomethyl)-2-chloro-1-nitro-3-(trifluoromethyl)benzene (3.94 g, 5.07 mmol, 99% yield) as a yellow oil.1H-NMR (400 MHz, CDCl3) δ/ppm: 7.97 (1H, d, J=2.0 Hz), 7.95 (1H, d, J=2.0 Hz), 4.52 (2H, s). Step 4—1-[4-chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine To 5-(bromomethyl)-2-chloro-1-nitro-3-(trifluoromethyl)benzene (500. mg, 0.6300 mmol) was added dimethylamine (2M in THF-3.14 mL, 62.8 mmol) and the reaction was stirred at rt for 2 hours. The solvent was removed in vacuo and the crude purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether. Fractions containing the product were loaded into an SCX-2 column and flushed at first with MeOH (20 mL) and then NH3in MeOH (20 mL) to give 1-[4-chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (101 mg, 0.36 mmol, 57% yield) as a pale yellow oil. UPLC-MS (ES+, short acidic): 1.19 min, m/z 283.1 [M+H]+.1H-NMR (400 MHz, CDCl3) δ/ppm: 7.91-7.90 (1H, m), 7.89-7.87 (1H, m), 3.50 (2H, s), 2.27 (6H, s). Step 5—4-[(dimethylamino)methyl]-2-nitro-6-(trifluoromethyl)aniline 1-[4-Chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (101 mg, 0.36 mmol) was dissolved in 1,4-dioxane (1 mL) in a seal tube followed by addition of NH3(28% in H2O-0.99 mL, 7.15 mmol). The reaction was stirred at 120° C. overnight, after which time a second portion of NH3(28% in H2O-0.99 mL, 7.15 mmol) was added. The reaction was left stirring overnight at 120° C. The crude was filtered over a phase separator to give 4-[(dimethylamino)methyl]-2-nitro-6-(trifluoromethyl)aniline (94 mg, 0.36 mmol, 100% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 0.90 min, m/z 264.2 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 8.36 (1H, s), 7.95 (1H, s), 7.42 (2H, s), 3.78 (2H, s), 2.40 (6H, s). Step 6—5-[(dimethylamino)methyl]-3-(trifluoromethyl)benzene-1,2-diamine 4-[(Dimethylamino)methyl]-2-nitro-6-(trifluoromethyl)aniline (94 mg, 0.36 mmol) was dissolved in EtOAc (10 mL) and EtOH (17 mL) followed by the addition of palladium, 10 wt. % on carbon powder, (12.1 mg) under a nitrogen atmosphere. The reaction was fitted with a H2balloon and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere overnight. The crude was filtered over celite and the solvent removed under reduce pressure to give 5-[(dimethylamino)methyl]-3-(trifluoromethyl)benzene-1,2-diamine (77 mg, 0.33 mmol, 92% yield) as a yellow solid.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 6.81 (1H, s), 6.77 (1H, s), 5.15-5.00 (4H, m), 3.83 (2H, br s), 2.50 (6H, s, (under DMSO)). UPLC-MS (ES+, short acidic): 0.76 min, m/z 234.2 [M+H]+. Intermediates in the table below was made in an analogous manner, using 1-methylpiperazine in place of dimethylamine in step 4. StructureNameData5-[(4-methylpiperazin-1-yl)methyl]-3- (trifluoromethyl)benzene-1,2-diamineUPLC-MS (ES+, short acidic): 0.49 min, m/z 289.3 [M + H]+. Example 10. Synthesis of 3-[(dimethylamino)methyl]-5-(trifluoromethyl)benzene-1,2-diamine Step 1—2-chloro-3-nitro-5-(trifluoromethyl)benzoic Acid 2-(Trifluoromethyl)pyridin-4-ol (1 g, 6.13 mmol) was dissolved in sulfuric acid (10.68 mL, 200.39 mmol) following by the addition of nitric acid, fuming, 90% (1.7 mL, 40.08 mmol). The reaction mixture was then heated to 90° C. for 45 min, after which time the reaction mixture was cooled to rt and poured into ice-cold water (250 mL). The product was filtered, washed with ice-cold water (40 mL) and dried in vacuo to give 2-chloro-3-nitro-5-(trifluoromethyl)benzoic acid (3.36 g, 12.48 mmol, 93% yield) as a white solid in 93% yield. UPLC-MS (ES−, short acidic): 1.46 min, m/z 268.1 [M−H]−.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 8.73-8.70 (1H, m), 8.43-8.39 (1H, m). Exchangeable proton not seen. Step 2—[2-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol Isobutyl chloroformate (3.41 mL, 26.31 mmol) was added to a stirred solution of 4-methylmorpholine (4.82 mL, 43.85 mmol), 2-chloro-3-nitro-5-(trifluoromethyl)benzoic acid (2.36 g, 8.77 mmol) in DCM (70 mL) at 0° C. under a nitrogen atmosphere. The reaction was stirred at rt overnight after which time the solvent was evaporated under reduce pressure. The crude was dissolved in THF (50 mL) and slowly added to a solution of NaBH4(497.64 mg, 13.16 mmol) in H2O (50 mL) at 0° C. The reaction was stirred for 1 h at rt, after which time it was quenched by carefully adding to sat. aq. NH4Cl (100 mL), followed by extraction with DCM (2×100 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The crude was purified by column chromatography using an eluent of 0-50% EtOAc in petroleum ether to give [2-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol (1.21 g, 1.51 mmol, 17%) yield as a yellow oil. UPLC-MS (ES+/−, short acidic): 1.63 min, m/z nd.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 8.13 (1H, s), 8.00 (1H, s), 4.93 (2H, s). Step 3—1-(bromomethyl)-2-chloro-3-nitro-5-(trifluoromethyl)benzene A solution of [2-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol (1.21 g, 1.51 mmol) in DCM (10 mL) was cooled to 0° C. followed by the addition of triphenylphosphine (475 mg, 1.81 mmol) and tetrabromomethane (551 mg, 1.66 mmol). The reaction was left stirring at rt overnight, after which time a second portion of triphenylphosphine (475 mg, 1.81 mmol) and tetrabromomethane (551 mg, 1.66 mmol) were added and left stirring for 4h. The reaction was washed with a sat. aq. NaHCO3; the organic phase was separated, dried over Na2SO4and the solvent removed in vacuo to give 1-(bromomethyl)-2-chloro-3-nitro-5-(trifluoromethyl)benzene (2.71 g, 1.49 mmol, 99% yield) as a yellow oil. UPLC-MS (ES+/−, short acidic): 1.26 min, m/z nd. Step 4—1-[2-chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine To 1-(Bromomethyl)-2-chloro-3-nitro-5-(trifluoromethyl)benzene (1 g, 0.55 mmol) was added dimethylamine (2M in THF-2.74 mL, 54.95 mmol). The reaction was left stirring at rt for 5 min, after which time the solvent was removed in vacuo and the crude purified by column chromatography using as eluent a gradient 0-20% EtOAc in petroleum ether. Fractions containing the product were loaded into an SCX-2 column and flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to give 1-[2-chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (82 mg, 0.29 mmol, 53% yield) as an orange oil. UPLC-MS (ES+, short acidic): 1.07 min, m/z 283.1 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 8.08-8.05 (1H, m), 7.94-7.90 (1H, m), 3.65 (2H, s), 2.34 (6H, s). Step 5—2-[(dimethylamino)methyl]-6-nitro-4-(trifluoromethyl)aniline 1-[2-chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (82 mg, 0.29 mmol) was dissolved in 1,4-dioxane (0.8100 mL) in a seal tube followed by addition of NH3(28% in H2O-0.81 mL, 5.8 mmol) and the reaction was left stirring at 120° C. overnight. The crude was filtered over a phase separator to give 2-[(dimethylamino)methyl]-6-nitro-4-(trifluoromethyl)aniline (76 mg, 0.29 mmol, 100% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.11 min, m/z 264.2 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 8.24 (1H, s), 8.12 (2H, s), 7.70 (1H, s), 3.66 (2H, br s), 2.23 (6H, s). Step 6—3-[(dimethylamino)methyl]-5-(trifluoromethyl)benzene-1,2-diamine 2-[(Dimethylamino)methyl]-6-nitro-4-(trifluoromethyl)aniline (75 mg, 0.28 mmol) was dissolved in EtOAc (5 mL) and EtOH (9 mL) followed by the addition of palladium, 10 wt. % on carbon powder, (10 mg) under a nitrogen atmosphere. The reaction was fitted with a H2balloon and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 4 hours. The crude was filtered over celite and the solvent removed under reduce pressure to give 3-[(dimethylamino)methyl]-5-(trifluoromethyl)benzene-1,2-diamine (66 mg, 0.28 mmol, 99% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 0.91 min, m/z 234.2 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 6.78 (2H, s), 6.70 (2H, br s), 4.98 (4H, br s), 2.28 (6H, br s). Example 11. Synthesis of N-[[3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-(trifluoromethyl)phenyl]methyl]propan-2-amine Step 1—N-[[3-bromo-5-(trifluoromethyl)phenyl]methyl]propan-2-amine 2-Aminopropane (0.34 mL, 3.95 mmol) was added to a stirred solution of 3-Bromo-5-(trifluoromethyl)benzaldehyde (0.6 mL, 3.95 mmol) and MeOH (10 mL) at rt under a nitrogen atmosphere for 18 hours. NaBH4(224.3 mg, 5.93 mmol) was then added and the resulting mixture stirred for 10 minutes and quenched with water (5 mL). Organic solvent was then removed in vacuo and the remaining mixture partitioned between water (50 mL) and EtOAc (50 mL). The aqueous layer was further extracted with EtOAc (50 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-10% MeOH in DCM to give N-[[3-bromo-5-(trifluoromethyl)phenyl]methyl]propan-2-amine (665 mg, 2.25 mmol, 57% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.13 min, m/z 295.9, 297.9 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.72 (1H, s), 7.66 (1H, s), 7.56 (1H, s), 3.84 (2H, s), 2.86 (1H, septet, J=6.4 Hz), 1.12 (6H, d, J=6.4 Hz). Exchangeable proton not seen. Step 2—N-[[3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-(trifluoromethyl)phenyl]methyl]propan-2-amine N-[[3-bromo-5-(trifluoromethyl)phenyl]methyl]propan-2-amine (300 mg, 1.01 mmol), bis(pinacolato)diboron (283 mg, 1.11 mmol), KOAc (298.3 mg, 3.04 mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (82.7 mg, 0.10 mmol) and 1,4-dioxane (10 mL) were stirred at 90° C. under a nitrogen atmosphere for 2 hours, after which time the reaction was cooled to rt and solvent removed in vacuo. The residue was dissolved in DCM (10 mL), filtered over celite, which was washed with DCM (10 mL). The filtrate was concentrated in vacuo and the residue purified by column chromatography using an eluent of 0-5% MeOH in DCM to give N-[[3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-(trifluoromethyl)phenyl]methyl]propan-2-amine (185 mg, 0.54 mmol, 53% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.02 min, m/z 262.1 [M-pinacol+H]+; 1.41 min, m/z 344.3 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.96-7.93 (2H, m), 7.71 (1H, s), 3.85 (2H, s), 2.88 (1H, septet, J=6.4 Hz), 1.38 (12H, s), 1.13 (6H, d, J=6.4 Hz). Exchangeable proton not seen. Example 12. Synthesis of [3-[(dimethylamino)methyl]-5-(trifluoromethyl)phenyl]boronic Acid Step 1—[3-bromo-5-(trifluoromethyl)phenyl]methanol Sodium Borohydride (149.5 mg, 3.95 mmol) was added to a stirred solution of 3-bromo-5-(trifluoromethyl)benzaldehyde (500 mg, 1.98 mmol) and MeOH (10 mL) at rt for 1 hour. The reaction was poured into water (50 mL) and the resulting mixture was extracted with EtOAc (2×50 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo to give [3-bromo-5-(trifluoromethyl)phenyl]methanol (444 mg, 1.74 mmol, 88% yield) as a colourless oil. The product was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.68 min, m/z nd.1H NMR (400 MHz, CDCl3) δ/ppm: 7.74 (1H, s), 7.71 (1H, s), 7.59 (1H, s), 4.79 (2H, s). Exchangeable proton not seen. Step 2—1-bromo-3-(bromomethyl)-5-(trifluoromethyl)benzene [3-Bromo-5-(trifluoromethyl)phenyl]methanol (444 mg, 1.74 mmol) was added to a stirred solution of triphenylphosphine (685 mg, 2.61 mmol), tetrabromomethane (866 mg, 2.61 mmol) and THF (20 mL) at −10° C. under a nitrogen atmosphere. The reaction was allowed to warm to rt and stir for 2 hours. It was then concentrated in vacuo and partitioned between water (100 mL) and DCM (100 mL). The organic layer was separated, dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-50% EtOAc in petroleum ether to give 1-bromo-3-(bromomethyl)-5-(trifluoromethyl)benzene (469 mg, 1.48 mmol, 85% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 2.06 min, m/z nd.1H NMR (400 MHz, CDCl3) δ/ppm: 7.76-7.72 (2H, m), 7.60 (1H, s), 4.47 (2H, s). Step 3—1-[3-bromo-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine 1-Bromo-3-(bromomethyl)-5-(trifluoromethyl)benzene (469 mg, 1.48 mmol) was added to a stirred solution of dimethylamine (2M in THF-3 mL, 6 mmol) at rt under a nitrogen atmosphere and left stirring for 10 min. Solvent was removed in vacuo and the residue partitioned between water (20 mL) and DCM (20 mL). The organic layer was separated, dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-10% MeOH in DCM to give 1-[3-bromo-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (337 mg, 1.19 mmol, 81% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.13 min, m/z 281.9, 283.9 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.70-7.67 (2H, m), 7.54 (1H, s), 3.46 (2H, s), 2.27 (6H, s). Step 4—[3-[(dimethylamino)methyl]-5-(trifluoromethyl)phenyl]boronic acid 1-[3-Bromo-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (337 mg, 1.19 mmol), bis(pinacolato)diboron (334 mg, 1.31 mmol), KOAc (352 mg, 3.58 mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (98 mg, 0.1200 mmol) and 1,4-dioxane (10 mL) under a nitrogen atmosphere and stirred at 90° C. for 2 hours. The reaction was cooled to rt and solvent removed in vacuo. The residue was suspended in DCM (10 mL), filtered over celite, which was washed with DCM (10 mL). The filtrate was concentrated in vacuo and the residue purified by column chromatography using an eluent of 0-5% MeOH in DCM to give [3-[(dimethylamino)methyl]-5-(trifluoromethyl)phenyl]boronic acid (109 mg, 0.44 mmol, 37% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 0.93 min, m/z 248.1 [M+H]+. Example 13. Synthesis of 2-ethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine Step 1—2-ethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine [1,1′-Bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (0.09 g, 0.11 mmol) was added to a stirred solution of bis(pinacolato)diboron (0.3 g, 1.18 mmol), 4-bromo-2-ethylpyridine (0.2 g, 1.07 mmol), KOAc (0.16 g, 1.61 mmol) and 1,4-dioxane (20 mL) under a nitrogen atmosphere and the reaction was heated to 100° C. for 2 hours. The reaction was cooled to rt and solvent removed in vacuo. The residue was taken up in DCM (50 mL), filtered over celite, which was washed with DCM (10 mL). The filtrate was concentrated in vacuo to give 2-ethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (250 mg, 1.0725 mmol, 99.765% yield) as a black oil. The product was used in the next step without further purification. UPLCMS (ES+, short acidic): 0.83 min, m/z 151.9 [M-pinacol]+.1H NMR (400 MHz, CDCl3) δ/ppm: 8.57 (1H, d, J=9.2 Hz), 7.54 (1H, d, J=6.4 Hz), 7.47 (1H, d, J=9.2 Hz), 2.85 (2H, q, J=8.0 Hz), 1.39 (12H, s), 1.33 (3H, t, J=8.0 Hz). Intermediates in the table below was made in an analogous manner, using the appropriate (hetero)aryl bromide in place of 4-bromo-2-ethylpyridine: StructureNameData2-(difluoromethyl)-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridineUPLCMS (ES+, short acidic): 1.02 min, m/z 173.9 [M-pinacol]+.1-methyl-5-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)benzimidazoleUPLCMS (ES+, short acidic): 1.23 min, m/z 259.0 [M + H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 8.30 (1H, s), 7.88 (1H, s), 7.76 (1H, d, J =8.0 Hz), 7.38 (1H, d, J = 8.0 Hz), 3.71 (3H, s), 1.38 (12H, s).3-methyl-5-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-1- (2,2,2-trifluoroethyl)pyrazole *Reaction performed at 90° C.UPLC-MS (ES+, Short acidic): 1.16 min, m/z 208.7 [M + H]+. Example 14. Synthesis of 1-ethyl-3-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole n-Butyllithium solution (0.4 mL, 1 mmol) was added to a stirred solution of 1-ethyl-3-methyl-pyrazole (0.1 mL, 0.91 mmol) and THF (5 mL) at 0° C. under a nitrogen atmosphere. The mixture was allowed to warm to 25° C. for 1 hour and then cooled back to −78° C. 2-Isopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (0.22 mL, 1.09 mmol) was added; after 10 mins at −78° C. the reaction was allowed to warm to room temperature and stir for 2 hours. The reaction was quenched with water and the resulting mixture reduced in vacuo. The residue was azeotroped with toluene to give 1-ethyl-3-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (214 mg, 0.91 mmol, 100% yield) as an off white solid. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 0.83 min, m/z 154.6 [M+H]+. Example 15. Synthesis of 1-cyclopropyl-3-ethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole Step 1—5-bromo-1-cyclopropyl-3-ethyl-pyrazole 2-cyclopropyl-5-ethyl-pyrazol-3-ol (730 mg, 4.8 mmol) was suspended in PBr3(3.05 mL, 16.79 mmol) with MeCN (3 mL) in a sealable vial. The vial was sealed and the reaction was heated thermally to 150° C. for 1.5 hours. After cooling to rt, the reaction separates into two layers; the top layer (MeCN) was quenched into iced saturated aq. NaHCO3to pH ˜7-8. The aqueous layer was separated, extracted with EtOAc (3×); the organics were combined, dried through a phase separator and reduced in vacuo to give 5-bromo-1-cyclopropyl-3-ethyl-pyrazole (340 mg, 1.58 mmol, 33% yield) as an orange oil. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.75 min, m/z 214.9, 216.9 [M+H]+. Step 2—1-cyclopropyl-3-ethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole [1,1′-Bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (129.1 mg, 0.16 mmol) was added to a stirred solution of bis(pinacolato)diboron (401.4 mg, 1.58 mmol), 5-bromo-1-cyclopropyl-3-ethyl-pyrazole (340 mg, 1.58 mmol), potassium acetate (310.3 mg, 3.16 mmol) and 1,4-dioxane (10 mL) at room temperature under inert atmosphere. The reaction was heated to 85° C. for 1 hour, cooled to room temperature and solvent removed in vacuo. The residue was taken up in DCM (20 mL), filtered through celite and the filter cake washed with DCM (10 mL). The filtrate was concentrated in vacuo to give 1-cyclopropyl-3-ethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (414 mg, 1.58 mmol, 100% yield) as a black oil. The compound was used in the next step without further purification. UPLCMS (ES+, short acidic): 1.05 min, m/z 180.7 [M-pinacol]+. Intermediates in the table below were made in an analogous manner, using the appropriate starting material in step 1: StructureNameData3-methyl-5-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-1-(2,2,2- trifluoroethyl)pyrazoleUPLCMS (ES+, short acidic): 1.16 min, m/z 208.7 [M- pinacol]+.3-ethyl-1-methyl-5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazoleUPLCMS (ES+, short acidic): 0.92 min, m/z 154.6 [M- pinacol]+.3-cyclopropyl-1-methyl-5- (4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazoleUPLCMS (ES+, short acidic): 0.98 min, m/z 166.7 [M- pinacol]+. Synthesis of Compounds of the Disclosure Example 16. Synthesis of 5-[3-[4-[3-(trifluoromethyl)phenyl]-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 12) Step 1—[2-oxo-2-[3-(trifluoromethyl)phenyl]ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylate 6-[(7-Oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (200 mg, 0.59 mmol) and K2CO3(244 mg, 1.76 mmol) were dissolved in dry DMF (5 mL) and treated with 3-(trifluoromethyl)phenacyl bromide (173 mg, 0.65 mmol) portionwise. The reaction was stirred for 2.5 hours at rt, after which time, a second portion of 3-(trifluoromethyl)phenacyl bromide was added (172 mg, 0.65 mmol) and the reaction was left stirring for 1h. Water (15 mL) was added and the product was extracted with DCM (×3). The combined organic phases were dried over Na2SO4, filtered and the solvent removed under reduce pressure. The product was purified by column chromatography using as eluent a gradient from 0-100% EtOAc in petroleum to give 2-oxo-2-[3-(trifluoromethyl)phenyl]ethyl]6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylate (55 mg, 0.10 mmol, 18% yield) as yellow solid. UPLC-MS (ES+, short acidic): 1.78 min, m/z 527.4 [M+H]+. Step 2—5-[3-[4-[3-(trifluoromethyl)phenyl]-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one [2-oxo-2-[3-(trifluoromethyl)phenyl]ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylate (55 mg, 0.10 mmol) and ammonium acetate (1.21 g, 15.67 mmol) were mixed in a sealed vial and the reaction heated to 130° C. for 8 hours. Water was added and the product extracted with EtOAc (×3). The organic phase was dried over Na2SO4, filtered and the solvent removed under reduce pressure. The product was purified by column chromatography using a gradient 0-100% EtOAc in petroleum ether followed by 0-5% MeOH in DCM to give 5-[3-[4-[3-(trifluoromethyl)phenyl]-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 12) as a white solid. UPLC-MS (ES+, final purity) 3.17 min, m/z 507.3 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.51 (0.10H, br s), 12.26 (0.90H, br s), 10.46 (1H, s), 8.10-7.99 (2H, m), 7.95 (1H, d, J=6.0 Hz), 7.82 (1H, s), 7.60-7.48 (2H, m), 7.02 (1H, s), 6.94-6.87 (2H, m), 6.26 (1H, d, J=5.6 Hz), 4.55-4.45 (1H, m), 4.19-4.09 (1H, m), 3.46-3.38 (1H, m), 3.30-3.20 (1H, m), 3.17-3.08 (1H, m), 2.93 (2H, t, J=7.6 Hz), 2.56-2.53 (m, 2H). Example 17. Synthesis of 5-[3-[4-(3-methoxyphenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 13) Step 1—[2-(3-methoxyphenyl)-2-oxo-ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylate 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (200 mg, 0.59 mmol) and 2-bromo-1-(3-methoxyphenyl)ethanone (135 mg, 0.59 mmol) were dissolved in dry MeCN (10 mL) and cooled to 0° C. followed by dropwise addition of DIPEA (0.15 mL, 0.88 mmol). The reaction was stirred at rt for 2 hours. After this time, iced water was added followed by extraction with EtOAc (×3) and DCM (×2) The combined organic phases were dried over Na2SO4, filtered and the solvent evaporated under reduce pressure. The product was purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give [2-(3-methoxyphenyl)-2-oxo-ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylate (217 mg, 0.44 mmol, 76% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.69 min, m/z 489.3 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 10.46 (1H, s), 7.95 (1H, d, J=6.0 Hz), 7.59-7.55 (1H, m), 7.51-7.43 (2H, m), 7.27 (1H, ddd, J=8.2 Hz, 2.6 Hz, 0.8 Hz), 7.01 (1H, d, J=2.8 Hz), 6.93-6.85 (2H, m), 6.25 (1H, d, J=5.6 Hz), 5.57 (2H, s), 4.44 (1H, dd, J=10.8 Hz, 3.2 Hz), 4.24 (1H, dd, J=10.8 Hz, 8.0 Hz), 3.83 (3H, s), 3.31-3.25 (1H, m), 3.10-3.05 (2H, m), 2.92 (2H, t, J=8.0 Hz), 2.53 (2H, t, J=8.0 Hz, (partly under DMSO)). Step 2—5-[3-[4-(3-methoxyphenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one [2-(3-methoxyphenyl)-2-oxo-ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylate (98 mg, 0.20 mmol) and ammonium acetate (309 mg, 4.01 mmol) were mixed in a sealed vial followed by addition of xylenes (1 mL) and heated to 150° C. for 30 min. Water was added and the product extracted with EtOAc (×3). The organic phase was dried over Na2SO4, filtered and the solvent removed under reduce pressure. The residue was loaded into an SCX-2 column and flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to elute the product. Fractions containing the product were combined and purified by column chromatography using as eluent a gradient 0-5% MeOH in DCM. Fractions containing the product were re-purified by reverse column chromatography using as eluent a gradient 5-100% of acetonitrile+0.1% formic acid in water+0.1% formic acid to give 5-[3-[4-(3-methoxyphenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 13) (8 mg, 0.017 mmol, 9% yield) as a white solid. UPLC-MS (ES+, final purity) 2.76 min, m/z 469.3 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.29 (br s, 0.2H), 12.09 (br s, 0.8H), 10.46 (s, 1H). 7.95 (d, J=5.6 Hz, 1H), 7.61 (d, J=2.0 Hz, 0.8H), 7.35-7.28 (m, 2H), 7.25-7.19 (m, 1.2H), 7.01 (d, J=2.4 Hz, 1H), 6.94-6.85 (m, 2H), 6.84-6.78 (m, 0.2H), 6.74 (ddd, J=8.4 Hz, 2.8 Hz, 1.2 Hz, 0.8H), 6.27 (d, J=5.6 Hz, 1H), 4.53-4.47 (m, 1H), 4.15-4.09 (m, 1H), 3.80 (s, 0.6H), 3.77 (s, 2.4H), 3.45-3.35 (m, 1H), 3.30-3.19 (m, 1H), 3.16-3.06 (m, 1H), 2.93 (t, J=7.2 Hz, 2H), 2.54-2.52 (m, 2H). The compounds in the table below were made in an analogous manner to Example 17, using the appropriate 2-bromo-1-arylethanone in place of 2-bromo-1-(3-methoxyphenyl)ethanone in step 1: Comp.NoStructure and NameData9UPLC-MS (ES+, final purity): 2.84 min, m/z 453.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.24 (0.2H, br s), 12.03 (0.8H, br s), 10.46 (1H, s), 7.95 (1H, d, J = 6.0 Hz), 7.63 (1.6H, d, J = 8.0 Hz), 7.54-7.50 (1.2H, m), 7.23- 7.18 (0.6H, m), 7.13 (1.6H, d, J = 8.0 Hz), 7.02-6.99 (1H, m), 6.94-6.86 (2H, m), 6.275-[3-[4-(p-tolyl)-1H-imidazol-2-(1H, d, J = 5.6 Hz), 4.54-4.46 (1H, m), 4.16-yl]chroman-6-yl]oxy-3,4-dihydro-4.06 (1H, m), 3.40-3.36 (1H, m), 3.26-3.181H-1,8-naphthyridin-2-one(1H, m), 3.14-3.05 (1H, m), 2.93 (2H, t, J =7.6 Hz), 2.53 (2H, t, J = 7.6 Hz, (partly underDMSO)), 2.30 (0.6H, s), 2.28 (2.4H, s).11UPLC-MS (ES+, final purity): 2.95 min, m/z 473.2 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.22 (1H, br s), 10.47 (1H, s), 7.95 (1H, d, J = 5.6 Hz), 7.75 (2H, d, J = 8.4 Hz), 7.61 (1H, br s), 7.38 (2H, d, J= 8.4 Hz), 7.00 (1H, d, J = 2.4 Hz), 6.91 (1H, dd, J = 8.8 Hz, 2.4 Hz), 6.88 (1H, d, J = 8.8 Hz), 6.26 (1H, d,5-[3-[4-(4-chlorophenyl)-1H-J = 6.0 Hz), 4.54-4.47 (1H, m), 4.12 (1H, t,imidazol-2-yl]chroman-6-yl]oxy-3,4-J =10.0 Hz), 3.42-3.36 (1H, m), 3.23 (1H, dd,dihydro-1H-1,8-naphthyridin-2-oneJ = 16.8 Hz, 10.4 Hz), 3.11 (1H, dd, J =16.8 Hz, 5.6 Hz), 2.93 (2H, t, J = 7.6 Hz),2.53 (2H, t, J = 7.6 Hz, (partly underDMSO)).10UPLC-MS (ES+, final purity): 2.72 min, m/z 469.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.19 (0.25H, br s), 12.00 (0.75H, br s), 10.46 (1H, s), 7.95 (1H, d, J = 5.6 Hz), 7.73- 7.51 (2H, m), 7.45 (0.75H, br s), 7.14 (0.25H, br s), 7.00 (1H, d, J = 2.4 Hz), 6.94- 6.85 (4H, m), 6.27 (1H, d, J = 5.6 Hz), 4.53-5-[3-[4-(4-methoxyphenyl)-1H-4.46 (1H, m), 4.11 (1H, t, J = 10.0Hz), 3.75imidazol-2-yl]chroman-6-yl]oxy-3,4-(3H, s), 4.42-3.37 (1H, m, (under water)),dihydro-1H-1,8-naphthyridin-2-one3.28-3.18 (1H, m), 3.09 (1H, dd, J = 16.8 Hz,5.2Hz), 2.93 (2H, t, J = 7.6 Hz), 2.53 (2H, t,J = 7.6 Hz, (partly under DMSO)).1UPLC-MS (ES, final purity): 2.78 min, m/z 469.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.05-11.97 (1H, m), 10.46 (1H, br s), 8.17- 8.14 (0.2H, m), 8.06 (0.8H, dd, J = 7.6 Hz, 1.6 Hz), 7.95 (1H, d, J = 6.0 Hz), 7.62-7.60 (0.2H, m), 7.50 (0.8H, d, J = 2.0 Hz), 7.30- 6.87 (6H, m), 6.27 (1H, d, J = 6.0 Hz), 4.55- 4.44 (1H, m), 4.13 (1H, t, J = 10.4 Hz), 3.895-[3-[4-(2-methoxyphenyl)-1H-(3H, s), 3.47-3.34 (1H, m), 3.29-3.20 (1H,imidazol-2-yl]chroman-6-yl]oxy-3,4-m), 3.15-3.06 (1H, m), 2.93 (2H, t, J =dihydro-1H-1,8-naphthyridin-2-one7.6 Hz), 2.53 (2H, t, J = 7.6 Hz, (partly underDMSO)).14UPLC-MS (ES+, final purity): 2.85 min, m/z 453.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.28 (0.2H, br s), 12.07 (0.8H, br s), 10.46 (1H, s), 7.95 (1H, d, J = 5.6 Hz), 7.61-7.39 (3H, m), 7.32-7.16 (1H, m), 7.07-6.95 (2H, m), 6.94-6.86 (2H, m), 6.27 (1H, d, J = 5.6 Hz), 4.53-4.47 (1H, m), 4.12 (1H, t, J = 10.0 Hz), 3.46-3.36 (1H, m), 3.29-3.19 (1H,5-[3-[4-(m-tolyl)-1H-imidazol-2-m), 3.16-3.07 (1H, m), 2.93 (2H, t, J =yl]chroman-6-yl]oxy-3,4-dihydro-7.6 Hz), 2.53 (2H, t, J = 7.6 Hz, (partly under1H-1,8-naphthyridin-2-oneDMSO)), 2.32 (3H, s).2UPLC-MS (ES+, final purity): 2.75 min, m/z 453.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.17-12.08 (1H, m), 10.46 (1H, s), 7.97- 7.94 (1H, m), 7.82-7.77 (0.75H, m), 7.45- 7.42 (0.25H, m), 7.34-7.15 (3H, m), 7.14- 7.07 (0.75H, m), 7.03-6.98 (1.25H, m), 6.95-6.86 (2H, m), 6.27 (1H, d, J = 5.6 Hz), 4.55-4.48 (1H, m), 4.19-4.08 (1H, m), 3.46- 3.36 (1H, m), 3.29-3.19 (1H, m), 3.15-3.065-[3-[4-(o-tolyl)-1H-imidazol-2-(1H, m), 2.93 (2H, t, J = 7.6 Hz), 2.53 (2H, t,yl]chroman-6-yl]oxy-3,4-dihydro-J = 7.6 Hz, (partly under DMSO)), 2.431H-1,8-naphthyridin-2-one(2.25H, s), 2.38 (0.75H, s).3UPLC-MS (ES+, final purity): 2.78 min, m/z 457.3 [M + H]+.1H -NMR (400 MHz, CD3OD) δ/ppm: 7.98- 7.86 (2H, m), 7.39 (1H, d, J = 3.2 Hz), 7.30- 7.11 (4H, m), 6.97-6.86 (3H, m), 6.35 (1H, d, J = 6.0 Hz), 4.55-4.48 (1H, m), 4.23 (1H, t, J = 10.8 Hz), 3.61-3.46 (1H, m), 3.30-3.24 (1H, m, (under water)) 3.20-3.11 (1H, m), 3.07 (2H, t, J = 7.6 Hz), 2.66 (2H, t, J = 7.6 Hz). Exchangeable proton missing.5-[3-[4-(2-fluorophenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy -3,4-dihydro-1H-1,8-naphthyridin-2-one15UPLC-MS (ES+, final purity): 2.80 min, m/z 457.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.38 (0.15H, br s), 12.19 (0.85H, br s), 10.46 (1H, s), 7.95 (1H, d, J = 5.6 Hz), 7.70 (1H, s), 7.62-7.46 (2H, m), 7.40-7.32 (1H, m), 7.03-6.85 (4H, m), 6.26 (1H, d, J = 5.6 Hz), 4.53-4.45 (1H, m), 4.13 (1H, t, J = 10.0 Hz), 3.45-3.36 (1H, m), 3.28-3.19 (1H, m), 3.16-3.07 (1H, m), 2.93 (2H, t, J =5-[3-[4-(3-fluorophenyl)-1H-7.6 Hz), 2.53 (2H, t, J = 7.6 Hz, (partly underimidazol-2-yl]chroman-6-yl]oxy-3,4-DMSO)).dihydro-1H-1,8-naphthyridin-2-one4UPLC-MS (ES+, final purity): 2.88 min, m/z 507.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.35 (0.15H, br s), 12.25 (0.85H, br s), 10.45 (1H, s), 7.95 (1H, d, J = 5.6 Hz), 7.90- 7.82 (1H, m), 7.74 (1H, d, J = 7.6 Hz), 7.66 (1H, t, J = 7.6 Hz), 7.61-7.57 (0.15H, m), 7.45 (0.85H, t, J = 7.6 Hz), 7.30 (0.85H, br s), 7.00 (1H, d, J = 2.4 Hz), 6.98-6.86 (2.15H, m), 6.26 (1H, d, J = 5.6 Hz), 4.56-5-[3-[4-[2-(trifluoromethyl)phenyl]-4.49 (1H, m), 4.13 (1H, t, J = 10.0 Hz), 3.47-1H-imidazol-2-yl]chroman-6-yl]oxy-3.39 (1H, m, (partly under water)), 3.28-3,4-dihydro-1H-1,8-naphthyridin-2-3.19 (1H, m), 3.16-3.08 (1H, m), 2.93 (2H,onet, J = 7.6 Hz), 2.56-2.53 (2H, m, (partlyunder DMSO)).20UPLC-MS (ES, final purity): 3.21 min, m/z 507.2 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.30 (1H, br s), 10.46 (1H, s), 8.00-7.93 (3H, m), 7.81 (1H, s), 7.68 (2H, d, J = 8.4 Hz), 7.02 (1H, d, J = 2.4 Hz), 6.94-6.87 (2H, m), 6.27 (1H, d, J = 6.0 Hz), 4.56-4.49 (1H, m), 4.14 (1H, t, J = 10.0 Hz), 3.48-3.375-[3-[4-[4-(trifluoromethyl)phenyl]-(1H, m), 3.29-3.21 (1H, m), 3.17-3.09 (1H,1H-imidazol-2-yl]chroman-6-yl]oxy-m), 2.93 (2H, t, J = 7.6 Hz), 2.56-2.53 (2H,3,4-dihydro-1H-1,8-naphthyridin-2-m).one5UPLC-MS (ES+, final purity): 2.96 min, m/z 475.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.37 (1H, br s), 10.46 (1H, s), 7.95 (1H, d, J = 5.6 Hz), 7.75-7.68 (1H, m), 7.55-7.49 (1H, m), 7.31-7.23 (1H, m), 7.09-6.99 (2H, m), 6.94-6.86 (2H, m), 6.26 (1H, d, J = 5.6 Hz), 4.55-4.47 (1H, m), 4.15 (1H, t, J = 10.4Hz), 3.47-3.38 (1H, m), 3.19-3.07 (2H, m, (partly under water), 2.93 (2H, t, J =5-[3-[4-(2,5-difluorophenyl)-1H-7.6 Hz), 2.57-2.52 (2H, t, J = 7.6 Hz, (partlyimidazol-2-yl]chroman-6-yl]oxy-3,4-under DMSO).dihydro-1H-1,8-naphthyridin-2-one16UPLC-MS (ES+, final purity): 2.73 min, m/z 464.2 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.34 (1H, br s), 10.46 (1H, s), 8.15 (1H, s), 8.08 (1H, d, J = 8.0 Hz), 7.95 (1H, d, J = 5.6 Hz), 7.80 (1H, s), 7.61 (1H, d, J = 7.6 Hz), 7.54 (1H, t, J = 7.6 Hz), 7.02-6.99 (1H, m), 6.94-6.86 (2H, m), 6.26 (1H, d, J = 6.0 Hz), 4.54-4.47 (1H, m), 4.14 (1H, t, J = 10.0 Hz), 3.46-3.39 (1H, m), 3.26-3.19 (1H, m), 3.16-3-[2-[6-[(7-oxo-6,8-dihydro-5H-1,8-3.08 (1H, m), 2.93 (2H, t, J = 7.6 Hz), 2.57-naphthyridin-4-yl)oxy]chroman-3-5.53 (2H, m, (partly under DMSO)).yl]-1H-imidazol-4-yl]benzonitrile The compound in the table below was made in an analogous manner, using the appropriate 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-carboxylic acid (Example 5) in place of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic in step 1 and using 2-bromo-1-phenylethanone in place of 2-bromo-1-(3-methoxyphenyl)ethanone in step 2: Comp.NoStructure and NameData45UPLC-MS (ES+, final purity): 2.66 min, m/z 453.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.32 (0.25H, s), 12.10 (0.75H, s), 10.80 (1H, s), 8.16 (1H, d, J = 5.6 Hz), 7.75 (1.5H, d, J = 7.6 Hz), 7.65 (1H, d, J = 2.0 Hz), 7.63 (0.25H, br s), 7.59 (0.75H, d, J = 2.0 Hz), 7.43-7.20 (2.75H, m), 7.16 (0.75H, t, J = 7.6 Hz), 7.03 (1H, d, J = 2.4 Hz), 6.94-6.87 (2H, m), 6.62 (1H, dd, J = 5.6 Hz, 2.4 Hz), 4.56-4.49 (1H, m), 4.12N-[4-[3-(4-phenyl-1H-imidazol-(1H, t, J = 10.4 Hz), 3.44-3.36 (1H, m), 3.242-yl)chroman-6-yl]oxy-2-(1H, dd, J = 16.4 Hz, 10.4 Hz), 3.12 (1H, dd,pyridyl]cyclopropanecarboxamideJ = 16.4 Hz, 4.4 Hz), 1.07 (1H, q, J = 6.0 Hz),0.77 (4H, d, J = 6.0 Hz). Example 18. Synthesis of 5-[3-[4-(2-methoxy-4-pyridyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 19) Step 1—5-[3-[4-(2-methoxy-4-pyridyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one 5-[3-[4-Bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (100 mg, 0.18 mmol), 2-methoxypyridine-4-boronic acid (32.1 mg, 0.21 mmol), [1,1′-Bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (14.3 mg, 0.020 mmol), K2CO3(72.6 mg, 0.52 mmol), 1,4-dioxane (5 mL) and water (1 mL) were combined under a nitrogen atmosphere and heated to 90° C. for 1 hour. The reaction was cooled to rt and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-10% MeOH in DCM to give 5-[3-[4-(2-methoxy-4-pyridyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (96 mg, 0.16 mmol, 91% yield) as a orange oil. UPLC-MS (ES+, short acidic): 1.82 min, m/z 600.5 [M+H]+. Step 2—5-[3-[4-(2-methoxy-4-pyridyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one Trifluoroacetic acid (1 mL, 13.06 mmol) was added to a stirred solution of 5-[3-[4-(2-methoxy-4-pyridyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (97 mg, 0.16 mmol) in DCM (4 mL) and EtOH (0.1 mL) at rt and stirred for 48 hours, after which time solvent and TFA were removed in vacuo. The residue was loaded into an SCX-2 column and flushed at first with MeOH (20 mL) and then NH3in MeOH (20 mL) to elute the product. Product containing fractions were combined and concentrated in vacuo. The residue was purified by column chromatography using an eluent of 0-10% MeOH in DCM to give 5-[3-[4-(2-methoxy-4-pyridyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (21 mg, 0.045 mmol, 28% yield) as a white solid. UPLC-MS (ES+, short acidic): 2.66 min m/z 470.3 [M+H].1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.34 (1H, s), 10.47 (1H, s), 8.07 (1H, d, J=5.2 Hz), 7.96 (1H, d, J=5.6 Hz), 7.87 (1H, d, J=6. Hz), 7.34-7.31 (1H, 6), 7.10 (1H, d, J=8.0 Hz), 7.03-7.00 (1H, m), 6.95-6.87 (2H, m), 6.27 (1H, d, J=6.0 Hz), 4.54-4.48 (1H, n), 4.14 (1H, t, J=10.0 Hz), 3.88-3.83 (3H, in), 3.46-3.39 (1H, in), 3.28-3.09 (2H, in), 2.94 (2H, t, J=7.6 Hz), 2.62-2.57 (2H, m, (under DMSO peak). The compounds in the table below were made in an analogous manner to Example 18, using the appropriate boronic acids/pinacol esters in place of 2-methoxypyridine-4-boronic acid in step 1: Comp.No.Structure and NameData8UPLC-MS (ES+, final acidic): 3.28 min, m/z 508.3 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.60 (1H, s), 10.52 (1H, s), 8.72 (1H, s), 8.22-8.18 (2H, m), 8.05-8.04 (1H, m), 8.01 (1H, d, J = 6.0 Hz), 7.08 (1H, d, J = 7.6 Hz), 6.99 (1H, dd, J = 8.8 Hz, 2.4 Hz), 6.96 (1H, d, J = 8.8 Hz), 6.32 (1H, d, J = 6.0 Hz), 4.61-4.55 (1H, m), 4.22 (1H, t, J = 10.4 Hz), 3.55-3.45 (1H, m),5-[3-[4-[2-(trifluoromethyl)-4-3.35-3.17 (2H, m), 2.99 (2H, t, J = 7.6 Hz),pyridyl]-1H-imidazol-2-2.62-2.58 (2H, m, (partly under DMSO)).yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one103UPLC-MS (ES+, final purity): 3.14 min, m/z 508.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.42 (1H, s), 10.47 (1H, s), 9.28-9.26 (1H, m), 8.78-8.75 (1H, m), 8.42-8.38 (1H, m), 8.00- 7.94 (2H, m), 7.04-7.01 (1H, m), 6.95-6.88 (2H, m), 6.27 (1H, d, J = 6.0 Hz), 4.55-4.49 (1H, m), 4.19-4.13 (1H, m), 3.48-3.41 (1H, m), 3.29-3.23 (1H, m), 3.18-3.11 (1H, m),5-[3-[4-[5-(trifluoromethyl)-3-2.94 (2H, t, J = 7.6 Hz), 2.54 (2H, t, J = 7.6 Hzpyridyl]-1H-imidazol-2-(partly under DMSO)).yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one17UPLC-MS (ES+, short acidic): 2.51 min, m/z 470.3 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.47-12.26 (1H, m), 10.47 (1H, s), 8.58 (1H, d, J = 2.4 Hz), 8.10 (1H, d, J = 2.8 Hz), 7.96 (1H, d, J = 10.4 Hz), 7.80-7.77 (1H, m), 7.65- 7.62 (1H, m), 7.03-7.01 (1H, m), 6.92 (1H, dd, J = 8.8 Hz, 3.2 Hz), 6.89 (1H, d, J = 8.8 Hz), 6.27 (1H, d, J = 6.0 Hz), 4.55-4.50 (1H, m),5-[3-[4-(5-methoxy-3-pyridyl)-1H-4.15 (1H, t, J = 10.0 Hz), 3.90-3.85 (3H, m),imidazol-2-yl]chroman-6-yl]oxy-3.47-3.37 (1H, m), 3.30-3.21 (1H, m), 3.17-3,4-dihydro-1H-1,8-naphthyridin-2-3.10 (1H, m), 2.94 (2H, t, J = 8.0 Hz), 2.56-one2.50 (2H, m).117UPLC-MS (ES+, final purity): 2.48 min, m/z 454.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.33 (1H, s), 10.47 (1H, s), 8.35-8.34 (1H, m), 7.97-7.95 (1H, m), 7.85 (1H, s), 7.58 (1H, s), 7.51-7.47 (1H, m), 7.04-7.00 (1H, m), 6.95- 6.88 (2H, m), 6.28-6.27 (1H, m), 4.54-4.49 (1H, m), 4.16-4.11 (1H, m), 3.44-3.36 (1H, m), 3.29-3.21 (1H, m), 3.17-3.09 (1H, m),5-[3-[4-(2-methyl-4-pyridyl)-1H-2.94 (2H, t, J = 8.0Hz), 2.54 (2H, t, J = 8.0 Hz,imidazol-2-yl]chroman-6-yl]oxy-(partly under DMSO)), 2.46 (3H, s).3,4-dihydro-1H-1,8-naphthyridin-2-one18UPLC-MS (ES+, final compound acidic): 2.60 min, m/z 501.3 [M + H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 12.26 (1H, br s), 10.46 (1H, s), 8.81 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.43 (1H, s), 7.00 (1H, s), 6.94-6.85 (2H, m), 6.26 (1H, d, J = 6.0 Hz), 4.53-4.46 (1H, m), 4.14 (1H, t, J = 10.0 Hz), 4.04 (3H, s), 3.91 (3H, s), 3.45-3.405-[3-[4-(2,4-dimethoxypyrimidin-5-(1H, m, (partly under water)), 3.27-3.19 (1H,yl)-1H-imidazol-2-yl]chroman-6-m), 3.16-3.06 (1H, m), 2.93 (2H, t, J = 7.6 Hz),yl]oxy-3,4-dihydro-1H-1,8-2.56-2.53 (2H, m, (partly under DMSO)).naphthyridin-2-one6UPLC-MS (ES+, final acidic): 2.69 min, m/z 578.4 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.26 (1H, s), 10.47 (1H, s), 8.06-8.00 (1H, m), 7.98-7.93 (2H, m), 7.83-7.80 (1H, m), 7.56- 7.50 (1H, m), 7.03-7.00 (1H, m), 6.95-6.88 (2H, m), 6.27 (1H, d, J = 5.6 Hz), 4.55-4.59 (1H, m), 4.14 (1H, t, J = 10.4 Hz), 3.92-3.79 (2H, m), 3.47-3.37 (1H, m), 3.30-3.09 (3H,5-[3-[4-[3-m), 2.94 (2H, t, J = 8.0 Hz), 2.56-2.53 (2H, m,[(isopropylamino)methyl]-5-(partly under DMSO peak)), 1.11-1.03 (6H,(trifluoromethyl)phenyl]-1H-m). Exchangeable proton not seen.imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one7UPLC-MS (ES+, final acidic), 2.61 min, m/z 564.5 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.26 (1H, s), 10.47 (1H, s), 7.99-7.94 (3H, m), 7.85-7.83 (1H, m), 7.43-7.39 (2H, m), 7.04- 7.01 (1H, m), 6.95-6.88 (2H, m), 6.28 (1H, d, J = 5.6 Hz), 4.54-4.48 (1H, m), 4.14 (1H, t, J = 10.4 Hz), 3.51-3.47 (2H, m), 3.45-3.37 (1H, m), 3.30-3.22 (1H, m), 3.17-3.09 (1H, m),5-[3-[4-[3-2.94 (2H, t, J = 8.0 Hz), 2.55-2.50 (2H, m,[(dimethylamino)methyl]-5-(partly under DMSO)), 2.21-2.15 (6H, m).(trifluoromethyl)phenyl]-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one108UPLC-MS (ES+, final purity): 2.34 min, m/z 443.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.06 (0.3H, s), 11.88 (0.7H, s), 10.47 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.88 (0.3H, s), 7.81 (0.7H, s), 7.66 (0.3H, s), 7.58 (0.7H, s), 7.20 (0.7H, d, J = 2.0 Hz), 7.03-6.98 (1H, m), 6.94-6.87 (2.3H, m), 6.27 (1H, d, J = 6.0 Hz), 4.51-4.45 (1H, m), 4.08 (1H, t, J = 10.4 Hz), 3.86 (0.9H,5-[3-[4-(1-methylpyrazol-4-yl)-1H-s), 3.82 (2.1H, s), 3.39-3.34 (1H, m), 3.24-imidazol-2-yl]chroman-6-yl]oxy-3.16 (1H, m), 3.12-3.04 (1H, m), 2.93 (2H, t,3,4-dihydro-1H-1,8-naphthyridin-2-J = 7.6 Hz), 2.54 (2H, t, J = 7.6 Hz, (partlyoneunder DMSO)).125UPLC-MS (ES+, final purity): 2.80 min, m/z 483.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.26 (1H, s), 10.46 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.42 (1H, d, J = 2.0 Hz), 7.01-6.99 (1H, m), 6.91 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.87 (1H, d, J = 8.8 Hz), 6.26 (1H, d, J = 6.0 Hz), 6.05 (1H, s), 4.52-4.46 (1H, m), 4.17-4.10 (1H, m), 3.905-[3-[4-(5-cyclopropyl-2-methyl-(3H, s), 3.45-3.37 (1H, m), 3.26-3.18 (1H, m).pyrazol-3-yl)-1H-imidazol-2-3.16-3.09 (1H, m), 2.93 (2H, t, J = 8.0 Hz),yl]chroman-6-yl]oxy-3,4-dihydro-2.53 (2H, t, J = 7.6 Hz), 1.82-1.85 (1H, m),1H-1,8-naphthyridin-2-one0.89-0.79 (2H, m), 0.64-0.58 (2H, m).126UPLC-MS (ES+, final purity): 3.16 min, m/z 525.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.37 (1H, s), 10.46 (1H, s), 7.96 (1H, d, J = 5.8 Hz), 7.54 (1H, d, J = 1.8 Hz), 6.99 (1H, d, J = 2.8 Hz), 6.91 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.86 (1H, d, J = 8.8 Hz), 6.29 (1H, s), 6.26 (1H, d, J = 5.7 Hz), 5.57-5.44 (2H, m), 4.52-4.43 (1H,5-[3-[4-[5-methyl-2-(2,2,2-m), 4.17 (1H, dd, J = 10.7 Hz, 9.1 Hz), 3.48-trifluoroethyl)pyrazol-3-yl]-1H-3.38 (1H, m), 3.26-3.07 (2H, m), 2.98-2.87imidazol-2-yl]chroman-6-yl]oxy-(2H, m), 2.57-2.52 (2H, m), 2.16 (3H, s).3,4-dihydro-1H-1,8-naphthyridin-2-one*Deprotection performed at 40° C.111UPLC-MS (ES+, final purity): 2.73 min, m/z 471.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.64 (1H, s), 10.46 (1H, s), 7.96 (1H, d, J = 5.6 Hz), 7.45-7.43 (1H, m), 7.01-6.99 (1H, m), 6.91 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.88 (1H, d, J = 8.8 Hz), 6.26 (1H, d, J = 5.6 Hz), 6.18 (1H, s), 4.53-4.47 (1H, m), 4.14 (1H, t, J = 9.6 Hz),5-[3-[4-(5-ethyl-2-methyl-pyrazol-3.92 (3H, s), 3.46-3.37 (1H, m), 3.27-3.193-yl)-1-2)(1H, m). 3.16-3.08 (1H, m), 2.93 (2H, t, J =trimethylsilylethoxymethyl)imidazo7.6 Hz), 2.53 (2H, t, J = 7.6 Hz), 2.52 (2H, m,1-2-yl]chroman-6-yl]oxy-3,4-(under DMSO)), 1.16 (3H, t, J = 7.6 Hz).dihydro-1H-1,8-naphthyridin-2-one112UPLC-MS (ES+, final purity): 2.86 min, m/z 485.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.24 (1H, s), 10.46 (1H, s), 7.95 (1H, d, J = 6.0 Hz), 7.37 (1H, d, J = 2.0 Hz), 6.99 (1H, d, J = 2.4 Hz), 6.91 (1H, dd, J = 8.0 Hz, 2.4 Hz), 6.86 (1H, d, J = 8.0 Hz), 6.26 (1H, d, J = 6.0 Hz), 6.07 (1H, s), 5.24-5.17 (1H, m), 4.51-4.465-[3-[4-(2-isopropyl-5-methyl-(1H, m), 4.19-4.13 (1H, m), 3.46-3.37 (1H,pyrazol-3-yl)-1H-imidazol-2-m), 3.25-3.17 (1H, m), 3.16-3.08 (1H, m), 2.93yl]chroman-6-yl]oxy-3,4-dihydro-(2H, t, J = 7.6 Hz), 2.53 (2H, t, J = 7.6 Hz),1H-1,8-naphthyridin-2-one2.52 (3H, s, (under DMSO), 1.33 (3H, d, J =7.6 Hz), 1.31 (3H, d, J = 7.6 Hz).114UPLC-MS (ES+, final purity): 2.74 min, m/z 483.4 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.48 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.72 (1H, s), 7.02 (1H, d, J = 2.8 Hz), 6.94 (1H, dd, J = 8.4 Hz, 2.4 Hz), 6.90 (1H, d, J = 8.4 Hz), 6.35 (1H, s), 6.27 (1H, d, J = 6.0 Hz), 4.55-450 (1H, m), 4.29-4.22 (1H, m), 3.78-3.75 (1H, m), 3.65-3.56 (1H, m), 3.32-3.17 (2H, m), 2.935-[3-[4-(2-cyclopropyl-5-methyl-(2H, t, J = 7.6 Hz), 2.54 (2H, t, J = 7.6 Hz),pyrazol-3-yl)-1H-imidazol-2-2.14 (3H, s), 1.06-0.96 (4H, m). Oneyl]chroman-6-yl]oxy-3,4-dihydro-exchangeable proton not seen due to TFA salt.1H-1,8-naphthyridin-2-one113UPLC-MS (ES+, final purity): 2.90 min, m/z 497.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.48 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.76 (1H, s), 7.03 (1H, d, J = 2.4 Hz), 6.95 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.91 (1H, d, J = 8.8 Hz), 6.42 (1H, s), 6.27 (1H, d, J = 6.0 Hz), 4.58-450 (1H, m), 4.31-4.23 (1H, m), 3.78-3.72 (1H, m), 3.66-3.59 (1H, m), 3.34-3.18 (2H, m), 2.935-[3-[4-(2-cyclopropyl-5-ethyl-(2H, t, J = 7.6 Hz), 2.54 (2H, t, J = 7.6 Hz),pyrazol-3-yl)-1H-imidazol-2-2.52 (2H, m, (under DMSO), 1.17 (3H, t, J =yl]chroman-6-yl]oxy-3,4-dihydro-7.6 Hz), 1.03-0.99 (4H, m). One exchangeable1H-1,8-naphthyridin-2-oneproton not seen due to TFA salt.*1st step at 85° C., 2nd step at 40° C. Example 19. Synthesis of 5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3-4-dihydro-1H-1,8-naphthyridin-2-one (Compound 106) Step 1—3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (200 mg, 0.47 mmol), K2CO3(195 mg, 1.41 mmol), 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (104.4 mg, 0.47 mmol), 1,4-dioxane (4 mL) and water (1 mL) were combined and stirred at rt under a nitrogen atmosphere. [1,1′-Bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (38.4 mg, 0.050 mmol) was added and the reaction heated to 100° C. for 2 hours. The reaction was cooled to rt and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 25-100% EtOAc in petroleum ether to give 3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (143 mg, 0.32 mmol, 69% yield) as an orange oil. UPLC-MS (ES+, short acidic): 1.74 min, m/z 441.8 [M+H]+. Step 2—5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one Potassium tert-Butoxide (40 mg, 0.36 mmol) was added to a stirred solution of 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (53.9 mg, 0.32 mmol) and 3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (143 mg, 0.32 mmol) in DMF (2 mL) and the reaction heated at 80° C. for 18 hours. The reaction was cooled to rt, solvent was reduced in vacuo and the residue partitioned between EtOAc and water. The organic layer was washed with sat. brine, separated, dried over a phase separator and the solvent reduced in vacuo. The residue was purified by column chromatography using as eluent a gradient 25-100% EtOAc in petroleum ether to give 5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (83 mg, 0.14 mmol, 44% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.83 min, m/z 587.5 [M+H]+. Step 3—5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one Trifluoroacetic acid (0.54 mL, 7.08 mmol) was added to a stirred solution of 5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (83 mg, 0.14 mmol) and DCM (2 mL) in a sealed vial and heated to 40° C. for 18 hours. The solvent was removed under reduce pressure and the residue was loaded into an SCX-2 column and flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to elute the product. The residue was purified by column chromatography using as eluent a gradient 1-8% MeOH in DCM. Fractions containing the product were combined, the solvent removed in vacuo and the residue was triturated in MeCN/Et20 to give a white solid, which was filtered and dried to give 5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (33.5 mg, 0.073 mmol, 52% yield). UPLC-MS (ES+, final purity): 2.62 min, m/z 457.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.30 (0.13H, s), 12.26 (0.87H, s), 10.46 (1H, s), 7.96 (1H, d, J=6.0 Hz), 7.42 (0.87H, d, J=1.6 Hz), 7.13 (0.13H, d, J=1.6 Hz), 7.01-6.98 (1H, m), 6.94-6.81 (2H, m), 6.26-6.25 (1.13H, m), 6.14 (0.87H, s), 4.53-4.44 (1H, m), 4.13 (1H, t, J=10.4 Hz), 3.91 (2.61H, s), 3.80 (0.39H, s), 3.45-3.35 (1H, m), 3.27-3.06 (2H, m), 2.93 (2H, t, J=8.0 Hz), 2.53 (2H, t, J=8.0 Hz, (party under DMSO), 2.15 (2.61H, s), 2.11 (0.39H, s). The compounds in the table below were made in an analogous manner to Example 19, using the appropriate boronic acids/pinacol esters in place of 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole in step 1: Comp.NoStructure and NameData107UPLC-MS (ES+, final purity): 3.34 min, m/z 511.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.47 (1H, br s), 10.46 (1H, s), 7.96 (1H, d, J = 5.8 Hz), 7.66 (1H, br s), 7.01 (1H, d, J = 2.7 Hz), 6.92 (1H, dd, J = 8.8, 2.7 Hz), 6.88 (1H, d, J = 8.8 Hz), 6.83 (1H, s), 6.26 (1H, d, J = 5.8 Hz), 4.51 (1H, m), 4.16 (1H, dd, J = 10.7, 9.5 Hz), 4.11 (3H, s), 3.43 (1H, m),5-[3-[4-[2-methyl-5-3.28-3.09 (2H, m), 2.93 (2H, t, J = 8.0 Hz),(trifluoromethyl)pyrazol-3-yl]-1H-2.54 (2H, t, J = 8.0 Hz, (partly underimidazol-2-yl]chroman-6-yl]oxy-3,4-DMSO)).dihydro-1H-1,8-naphthyridin-2-one110UPLC-MS (ES+, final purity): 3.02 min, m/z 459.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.32 (0.16H, s), 12.07 (0.84H, s), 10.46 (1H, s), 7.96 (1H, d, J = 5.8 Hz), 7.36 (1H, d, J = 1.9 Hz), 7.07-6.83 (4H, m), 6.77 (0.16H, dd, J = 3.6 Hz, 1.3 Hz), 6.68 (0.84H, dd, J = 3.6 Hz, 1.3 Hz), 6.29-6.25 (1H, m), 4.52-4.445-[3-[4-(5-methyl-2-thienyl)-1H-(1H, m), 4.10 (1H, t, J = 10.3 Hz), 3.41-3.34imidazol-2-yl+chroman-6-yl+oxy-3,4-(1H, m), 3.25-3.05 (2H, m), 2.93 (2H, t, J =dihydro-1H-1,8-naphthyridin-2-one7.7 Hz), 2.54 (2H, t, J = 7.7 Hz, (partly underDMSO)), 2.44 (0.48H, d, J = 1.1 Hz), 2.41(2.52H, d, J = 1.1 Hz).118UPLC-MS (ES+, final purity): 3.07 min, m/z 468.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.33 (1H, s), 10.47 (1H, s), 8.40-8.36 (1H, m), 7.96 (1H, d, J = 5.6 Hz), 7.87 (1H, s), 7.58 (1H, s), 7.52-7.48 (1H, m), 7.04-7.00 (1H, m), 6.95-6.87 (2H, m), 6.27 (1H, d, J = 5.6 Hz), 4.54-4.49 (1H, m), 4.14 (1H, t, J =5-[3-[4-(2-ethyl-4-pyridyl)-1H-10.4 Hz), (3.47-3.38 (1H, m), 3.29-3.21 (1H,imidazol-2-yl]chroman-6-yl]oxy-3,4-m), 3.17-3.09 (1H, m), 2.94 (2H, t, J =dihydro-1H-1,8-naphthyridin-2-one7.6 Hz), 2.74 (2H, q, J = 7.6 Hz), 2.52 (2H, t,J =7.6 Hz), 1.25 (3H, t, J = 7.6 Hz).101UPLC-MS (ES+, final purity): 3.07 min, m/z 490.2 [M + H]+.1H NMR (400 MHz, DMSO d6) δ/ppm: 12.48 (1H, s), 10.46 (1H, s), 8.58 (1H, d, J = 5.2 Hz), 8.07-8.05 (1H, m), 8.01-7.99 (1H, m), 7.96 (1H, d, J = 6.0 Hz), 7.87-7.84 (1H, m), 7.03-7.01 (1H, m), 6.96-6.88 (2H, m), 6.93 (1H, t, J = 55.0 Hz), 6.27 (1H, d, J =5-[3-[4-[2-(difluoromethyl)-4-5.2Hz), 4.55 (1H, m), 4.16 (1H, t, J =pyridyl]-1H-imidazol-2-yl]chroman-10.4 Hz), 3.49-3.39 (1H, m), 3.28-3.22 (1H,6-yl]oxy-3,4-dihydro-1H-1,8-m), 3.18-3.10 (1H, m), 2.93 (2H, t, J =naphthyridin-2-one7.6 Hz), 2.54 (2H, t, J = 7.6 Hz).119UPLC-MS (ES+, final purity): 2.68 min, m/z 480.3 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.32 (1H, s), 10.46 (1H, s), 8.29 (1H, s), 7.96 (1H, dd, J = 5.6 Hz), 7.86 (1H, d, J = 2.0 Hz), 7.61 (1H, d, J = 5.2 Hz), 7.43 (1H, dd, J = 5.2 Hz, 1.6 Hz), 7.04-7.00 (1H, m), 6.96-6.89 (2H, m), 6.27 (1H, d, J = 5.6 Hz), 4.55-4.49 (1H, m), 4.14 (1H, t, J = 6.4 Hz),5-[3-[4-(2-cyclopropyl-4-pyridyl)-3.48-3.73 (1H, m), 3.28-3.21 (1H, m), 3.17-1H-imidazol-2-yl]chroman-6-yl]oxy-3.09 (1H, m), 2.93 (2H, t, J = 7.6 Hz), 2.543,4-dihydro-1H-1,8-naphthyridin-2-(2H, t, J = 7.6 Hz), 2.11-2.05 (1H, m), 0.92-one0.91 (4H, m).120UPLC-MS (ES+, final purity): 2.76 min, m/z 482.3 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.32 (1H, s), 10.46 (1H, s), 8.39 (1H, d, J = 5.2 Hz), 7.96 (1H, J = 5.6 Hz), 7.88 (1H, d, J = 7.2 Hz), 7.58-7.56 (1H, m), 7.52-7.49 (1H, m), 7.03-7.01 (1H, m), 6.95-6.88 (2H, m), 6.27 (1H, d, J = 7.2 Hz), 4.55-4.49 (1H, m), 4.14 (1H, t, J = 10.4 Hz), 3.47-3.38 (1H,5-[3-[4-(2-isopropyl-4-pyridyl)-1H-m), 3.28-3.21 (1H, m), 3.17-3.10 (1H, m),imidazol-2-yl]chroman-6-yl]oxy-3,4-3.04-2.99 (1H, m), 2.94 (2H, t, J = 8.0 Hz),dihydro-1H-1,8-naphthyridin-2-one2.54 (2H, t, J = 8.0 Hz), 1.25 (6H, d, J = 6.8Hz).121UPLC-MS (ES+, final purity): 2.84 min, m/z 496.3 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.32 (1H, s), 10.46 (1H, s), 8.41 (1H, d, J = 5.2 Hz), 7.96 (1H, d, J = 6.0 Hz), 7.90-7.89 (1H, m), 7.73-7.72 (1H, m), 7.50 (1H, dd, J = 4.8 Hz, 1.6 Hz), 7.03-7.01 (1H. m), 6.93 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.89 (1H, d, J = 8.8 Hz), 6.27 (1H, d, J = 5.6 Hz), 4.54-4.475-[3-[4-(2-tert-butyl-4-pyridyl)-1H-(1H, m), 4.15 (1H, t, J = 10.4 Hz), 3.47-3.38imidazol-2-yl]chroman-6-yl]oxy-3,4-(1H, m), 3.29-3.21 (1H, m), 3.17-3.10 (1H,dihydro-1H-1,8-naphthyridin-2-onem), 2.94 (2H, t, J = 7.6 Hz), 2.54 (2H, t, J =7.6 Hz), 1.34 (9H, s).128UPLC-MS (ES+, final purity): 2.79 min, m/z 508.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.40 (1H, s), 10.46 (1H, s), 8.60 (1H, dd, J = 4.8 Hz,1.6 Hz), 8.32 (1H, d, J = 7.6 Hz), 7.95 (1H, d, J = 6.0 Hz), 7.72 (1H, dd, J = 8.0 Hz, 4.8 Hz), 7.43 (1H, s), 7.02-7.00 (1H, m), 6.92 (1H, dd, J = 8.8 Hz, 6.4 Hz), 6.89 (1H, d, J = 8.8 Hz), 6.27 (1H, d, J = 6.0 Hz),5-[3-[4-[2-(trifluoromethyl)-3-4.55-4.50 (1H, m), 4.15 (1H, t, J = 10.4 Hz),pyridyl]-1H-imidazol-2-yl]chroman-3.49-3.41 (1H, m), 3.28-3.20 (1H, m), 3.18-6-yl]oxy-3,4-dihydro-1H-1,8-3.11 (1H, m), 2.93 (2H, t, J = 7.6 Hz), 2.54naphthyridin-2-one(2H, t, J = 7.6 Hz).127UPLC-MS (ES+, final purity): 3.24 min, m/z 508.1 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.44 (1H, s), 10.46 (1H, s), 9.13 (1H, d, J = 2.4 Hz), 8.33 (1H, dd, J = 8.0 Hz, 2.0 Hz), 7.98-7.95 (2H, m), 7.86 (1H, d, J = 8.4 Hz), 7.02 (1H, d, J = 2.4 Hz), 6.92 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.89 (1H, d, J = 8.8 Hz),5-[3-[4-[6-(trifluoromethyl)-3-6.27 (1H, d, J = 6.4 Hz), 4.56-4.50 (1H, m),pyridyl]-1H-imidazol-2-yl]chroman-4.16 (1H, t, J = 10.0 Hz), 3.50-3.40 (1H, m),6-yl]oxy-3,4-dihydro-1H-1,8-3.30-3.22 (1H, m), 3.18-3.11 (1H, m), 2.93naphthyridin-2-one(2H, t, J = 7.6 Hz), 2.54 (2H, t, J = 7.6 Hz).109UPLC-MS (ES+, final purity): 2.39 min, m/z 457.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 11.93-11.88 (1H, m), 10.46 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.48 (0.35H, s), 7.72 (0.65H, s), 7.08-7.07 (0.65H, m), 7.01-6.99 (1H, m), 6.91 (1H, dd, J = 8.4 Hz, 2.8 Hz), 6.88 (1H, d, J = 8,4 Hz), 6.82-6.81 (0.35, m), 6.27 (1H, d, J = 6.0 Hz), 4.51-4.45 (1H, m),5-[3-[4-(1,3-dimethylpyrazol-4-yl)-4.09 (1H, t, J = 10.4 Hz), 3.79 (1.05H, s),1H-imidazol-2-yl]chroman-6-yl]oxy-3.74 (1.95H, s), 3.39-3.34 (1H, m), 3.25-3,4-dihydro-1H-1,8-naphthyridin-2-3.17 (1H, m), 3.12-3.05 (1H, m), 2.93 (2H,onet, J = 8.4 Hz), 2.53 (2H, t, J = 8.4 Hz), 2.25(1.95H, s), 2.23 (1.05H, s).129UPLC-MS (ES+, final purity): 2.24 min, m/z 493.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.26 (0.3H, s), 12.01 (0.7H, s), 10.46 (1H, s), 8.18 (0.3H, s), 8.12 (0.7H, s), 8.01-7.93 (2H, m), 7.75-7.72 (0.6H, m), 7.60-7.58 (1.4H, m), 7.52-7.49 (0.7H, m), 7.26-7.25 (0.3H, m), 7.04-7.00 (1H, m), 6.92 (1H, dd,5-[3-[4-(1-methylbenzimidazol-5-J = 8.8 Hz, 2.8 Hz), 6.89 (1H, d, J = 8.8 Hz),yl)-1H-imidazol-2-yl]chroman-6-6.28 (1H, d, J = 6.0 Hz), 4.55 (1H, m), 4.19-yl]oxy-3,4-dihydro-1H-1,8-4.13 (1H, m), 3.85 (0.9H, s), 3.83 (2.1H, s),naphthyridin-2-one3.44-3.35 (1H, m), 3.30-3.25 (1H, m), 3.18-3.10 (1H, m), 2.94 (2H, t, J = 7.6 Hz), 2.54(2H, t, J = 7.6 Hz).130UPLC-MS (ES+, final purity): 2.66 min, m/z 493.1 [M + H]+.1H NMR (400 MHz, CD3OD) δ/ppm: 7.88- 7.74 (3H, m), 7.67-7.61 (1H, m), 7.45-7.35 (2H, m), 6.88-6.78 (3H, m), 6.23 (1H, d, J = 6.0 Hz), 4.46-4.41 (1H, m), 4.16 (1H, t, J = 10.4 Hz), 3.99 (3H, s), 3.49-3.39 (1H, m), 3.27-3.23 (1H, m), 3.12-3.02 (1H, m), 2.97 (2H, t, J = 7.2 Hz), 2.57 (2H, t, J = 7.2 Hz).5-[3-[4-(1-methylindazol-6-yl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one131UPLC-MS (ES+, final purity): 2.60 min, m/z 493.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.31 (0.2H, s), 12.05 (0.8H, s), 10.47 (1H, m), 8.11-8.07 (1H, m), 8.02-8.00 (0.8H, m), 7.99-7.95 (1.2H, m), 7.83 (0.8H, dd, J = 8.8 Hz, 1.6 Hz), 7.71-7.69 (0.4H, m), 7.61- 7.57 (1.6H, m), 7.27-7.25 (0.2H, m), 7.04- 7.01 (1H, m), 6.93 (1H, dd, J = 8.8 Hz,5-[3-[4-(1-methylindazol-5-yl)-1H-2.4 Hz), 6.89 (1H, d, J = 8.8 Hz), 6.28 (1H,imidazol-2-yl]chroman-6-yl]oxy-3,4-d, J = 6.0 Hz), 4.56 (1H, m), 4.15 (1H, t, J =dihydro-1H-1,8-naphthyridin-2-one10.0 Hz), 4.06 (0.6H, s), 4.04 (2.4, s), 3.45-3.36 (1H, m), 3.29-3.22 (1H, m), 3.17-3.10(1H, m), 2.94 (2H, t, J = 7.6 Hz), 2.54 (2H, t,J = 7.6 Hz).105UPLC-MS (ES+, final purity): 2.75 min, m/z 490.2 [M + H]+.1NMR (400 MHz, DMSO-d6) δ/ppm: 12.44 (1H, s), 10.47 (1H, s), 9.15-9.12 (1H, m), 8.61-8.58 (1H, m), 8.29-8.27 (1H, m), 7.97-7.87 (2H, m), 7.19 (1H, t, J = 55.2 Hz), 7.04-7.01 (1H, m), 6.96-6.88 (2H, m), 6.27 (1H, d, J = 5.2 Hz), 4.56-4.50 (1H, m), 4.175-[3-[4-[5-(difluoromethyl)-3-(1H, t, J = 10.0 Hz), 3.51-3.41 (1H, m), 3.29-pyridyl]-1H-imidazol-2-yl]chroman-3.22 (1H, m), 3.19-3.11 (1H, m), 2.94 (2H,6-yl]oxy-3,4-dihydro-1H-1,8-t, J = 7.2 Hz), 2.56-2.50 (2H, m).naphthyridin-2-one124UPLC-MS (ES+, final purity): 2.63 min, m/z 471.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.25 (1H, s), 10.46 (1H, s), 7.96 (1H, d, J = 5.8 Hz), 7.40 (1H, s), 7.00 (1H, d, J = 2.8 Hz), 6.91 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.87 (1H, d, J = 8.8 Hz), 6.26 (1H, d, J = 5.8 Hz), 6.12 (1H, s), 4.54-4.44 (1H, m), 4.40-4.28 (2H, m), 4.16 (1H, t, J = 10.0 Hz), 3.40 (1H,5-[3-[4-(2-ethyl-5-methyl-pyrazol-3-m), 3.22 (1H, dd, J =16.7 Hz, 9.5 Hz), 3.12yl)-1H-imidazol-2-yl]chroman-6-(1H, dd, J = 16.6 Hz, 5.8 Hz), 2.93 (2H, dd,yl]oxy-3,4-dihydro-1H-1,8-J = 8.4 Hz, 7.0 Hz), 2.57-2.53 (2H, m (partlynaphthyridin-2-oneunder DMSO)), 2.12 (3H, s), 1.25 (3H, t,J = 7.1 Hz). Example 20. Synthesis of 5-[3-(4-tetrahydropyran-4-yl-1H-imidazol-2-yl)chroman-6-yl]oxy-3,4-dihydro-TH-1,8-naphthyridin-2-one (Compound 132) Step 1—3-[4-(3,6-dihydro-2H-pyran-4-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol 3,6-Dihydro-2H-pyran-4-boronic acid pinacol ester (108.7 mg, 0.52 mmol), 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (220 mg, 0.52 mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (42.2 mg, 0.050 mmol), potassium carbonate (214.4 mg, 1.55 mmol), 1,4-dioxane (4 mL) and water (1 mL) were combined at room temperature under a nitrogen atmosphere in a sealable vial. The vial was sealed and the reaction was heated to 80° C. and allowed to stir for 2 hours. It was then cooled to room temperature and solvent removed in vacuo. The residue was taken up in DCM (20 mL) and filtered through celite and the filter cake washed with DCM (10 mL). The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 3-[4-(3,6-dihydro-2H-pyran-4-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (149 mg, 0.35 mmol, 67% yield) as a yellow solid. UPLCMS (ES+, short acidic): 1.53 min, m/z 429.7 [M+H]+. Step 2—3-[4-tetrahydropyran-4-yl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol 3-[4-(3,6-dihydro-2H-pyran-4-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (149. mg, 0.35 mmol) was stirred in EtOAc (10 mL) at room temperature under a nitrogen atmosphere. Palladium, 10 wt. % on carbon powder, dry (20 mg) was added and the reaction fitted with a hydrogen balloon and subjected to 3×vacuum/hydrogen cycles and then allowed to stir under a hydrogen atmosphere for 18 hours. The reaction was then filtered through a celite plug and the plug washed with MeOH (10 mL). The filtrate was concentrated in vacuo to give 3-[4-tetrahydropyran-4-yl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (95 mg, 0.22 mmol, 63% yield) as a yellow solid. The compound was used directly in the next step without further purification. UPLC-MS (ES+, short acidic): 1.49 min, m/z 431.7 [M+H]+. Step 3—5-[3-(4-tetrahydropyran-4-yl-1H-imidazol-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one Potassium tert-butoxide (75.4 mg, 0.67 mmol) was added to a stirred solution of 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (36.7 mg, 0.22 mmol) and 3-[4-tetrahydropyran-4-yl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (95 mg, 0.22 mmol) in DMF (2 mL) and the reaction was sealed in a tube and heated at 90° C. for 60 hours. Reaction was cooled to room temperature and the reaction was reduced in vacuo and partitioned between EtOAc and water. The organics were washed with saturated brine, separated, dried over a phase separator and reduced in vacuo. The residue was purified by column chromatography using as eluent a gradient 1-10% MeOH/DCM. Fractions containing the product were combined and re-purified by preparative HPLC (early method). Fractions containing the product were combined, solvent removed under reduce pressure and loaded onto a SCX column, which was flushed at first with MeOH and then 1.0 M MeOH/NH3to give 5-[3-(4-tetrahydropyran-4-yl-1H-imidazol-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (5 mg, 0.011 mmol, 5% yield) as a white solid. UPLC-MS (ES+, Final purity): 2.36 min, m/z 447.2 [M+H]+.1H NMR (400 MHz, DMSO-d6+CF3CO2D) δ/ppm: 14.11 (1H, s), 10.65 (1H, s), 8.00 (1H, d, J=6.0 Hz), 7.46 (1H, s), 7.03 (1H, d, J=2.8 Hz), 7.00-6.91 (2H, m), 6.30 (1H, d, J=6.0 Hz), 4.51 (1H, dd, J=10.9 Hz, 3.1 Hz), 4.34 (1H, dd, J=11.0 Hz, 8.1 Hz, 1H), 3.93 (2H, ddd, J=11.5 Hz, 4.5 Hz, 2.0 Hz), 3.83-3.73 (1H, m), 3.44 (2H, td, J=11.7 Hz, 2.1 Hz), 3.30-3.23 (2H, m), 3.01-2.89 (3H, m), 2.56 (2H, dd, J=8.4 Hz, 7.0 Hz), 1.92-1.82 (2H, m), 1.63 (2H, qd, J=12.1 Hz, 4.4 Hz). Example 21. Synthesis of 5-[3-[5-(3-methoxy-2-thienyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 122) Step 1—tert-butyl N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate and tert-butyl N-[2-(3-methoxy-2-thienyl)-2-oxo-ethyl]carbamate n-Butyllithium solution (3.5 mL, 8.76 mmol) was added to a stirred solution of 3-methoxythiophene (0.87 mL, 8.76 mmol) and THF (20 mL) at −78° C. under a nitrogen atmosphere. The mixture was stirred for 20 minutes at −78° C., allowed to warm to 0° C. for 20 minutes and then cooled back to −78° C. and tert-butyl N-[2-[methoxy(methyl)amino]-2-oxo-ethyl]carbamate (477.9 mg, 2.19 mmol) was added. After 1 hour at −78° C. the reaction was allowed to warm to room temperature and stir for 18 hours. The reaction was then poured into water (100 mL) and the resulting mixture extracted with EtOAc (2×100 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give tert-butyl N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate (93 mg, 0.34 mmol, 16% yield) and tert-butyl N-[2-(3-methoxy-2-thienyl)-2-oxo-ethyl]carbamate (170 mg, 0.63 mmol, 29% yield) as yellow oils. N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate: UPLCMS (ES+, short acidic): 1.62 min, m/z 294.0 [M+Na]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.37 (1H, d, J=1.6 Hz), 6.67 (1H, d, J=1.6 Hz), 5.39 (1H, br s), 4.53-4.52 (2H, m), 3.83 (3H, s), 1.45 (9H, s). N-[2-(3-methoxy-2-thienyl)-2-oxo-ethyl]carbamate: UPLCMS (ES+, short acidic): 1.60 min, m/z 272.0 [M+H]+, 294.0 [M+Na]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.59 (1H, d, J=5.6 Hz), 6.86 (1H, d, J=5.6 Hz), 5.56 (1H, br s), 4.52-4.50 (2H, m), 4.00 (3H, s), 1.47 (9H, s). Step 2—2-amino-1-(4-methoxy-2-thienyl)ethanone Hydrochloride Hydrogen Chloride (0.26 mL, 1.03 mmol-4M in dioxane) was added to a stirred solution of tert-butyl N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate (93 mg, 0.34 mmol) and DCM (5 mL) at room temperature. The reaction was allowed to stir for 18 hours after which time the solvent was removed in vacuo to give 2-amino-1-(4-methoxy-2-thienyl)ethanone hydrochloride (71 mg, 0.34 mmol, 100% yield) as a yellow solid, which was used in the next step without further purification. UPLCMS (ES+, short acidic), 0.47 min, m/z 171.9 [M+H]+. Step 3—N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide Propylphosphonic anhydride (0.31 mL, 0.51 mmol) was added to a stirred solution of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (116.4 mg, 0.34 mmol), 2-amino-1-(4-methoxy-2-thienyl)ethanone hydrochloride (71 mg, 0.34 mmol), triethylamine (0.1 mL, 0.68 mmol) and THF (5 mL). The reaction was heated to 65° C. and stirred for 18 hours. The reaction was then cooled to room temperature and solvent removed in vacuo. The residue was partitioned between water (50 mL) and DCM (50 mL). The organic layer was separated, dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-5% MeOH in DCM to give N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide (52 mg, 0.11 mmol, 31% yield) as a white solid. UPLCMS (ES+, short acidic): 1.47 min, m/z 494.1 [M+H]+. Step 4—5-[3-[5-(3-methoxy-2-thienyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide (52 mg, 0.11 mmol) and ammonium acetate (243.7 mg, 3.16 mmol) were taken up in butyric acid (2 mL, 21.88 mmol) in a sealable vial, which was sealed and heated at 175° C. for 2 hours. The reaction mixture was evaporated to dryness and then treated with aq. K2CO3solution and extracted with DCM (15 ml). The organic layer was passed through a phase separator and then evaporated to dryness. The residue was chromatographed via preparative LCMS to give 5-[3-[5-(4-methoxy-2-thienyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (8 mg, 0.016 mmol, 15% yield) as an off white solid product. UPLC-MS (ES+, final purity): 2.80 min, m/z 475.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.67 (1H, s), 8.05-7.91 (2H, m), 7.69 (1H, d, J=1.5 Hz), 7.25 (TH, d, J=1.7 Hz), 7.05 (1H, d, J=2.5 Hz), 7.02-6.88 (2H, in), 6.76 (1H, d, J=1.8 Hz), 6.32 (1H, d, J=6.0 Hz), 4.56 (1H, dd, J=10.9 Hz, 3.2 Hz), 4.35 (1H, dd, J=11.0 Hz, 8.6 Hz), 3.79 (3H, s), 3.87-3.69 (TH, in), 3.35-3.23 (2H, in), 2.95 (2H, t, J=7.7 Hz), 2.60-2.53 (2H, in). The compounds in the table below were made in an analogous manner to Example 21, using the appropriate reagent in place of tert-butyl N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate in step 2: Comp.NoStructure and NameData123UPLC-MS (ES+, final purity): 2.70 min, m/z 475.1 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 10.59 (1H, s), 7.99 (1H, d, J = 5.9 Hz), 7.74 (1H, s), 7.68 (1H, d, J = 5.5 Hz), 7.21 (1H, d, J = 5.5 Hz), 7.04 (1H, d, J = 2.7 Hz), 7.01-6.91 (2H, m), 6.30 (1H, d, J = 5.9 Hz), 4.55 (1H, ddd, J = 10.8 Hz, 3.3 Hz, 1.3 Hz), 4.33 (1H, dd, J = 10.8 Hz, 8.9 Hz), 3.95 (3H, s), 3.80 (1H, dq, J = 8.8 Hz, 2.9 Hz), 3.38-3.19 (2H, m), 2.94 (2H, dd, J = 8.4 Hz,7.0 Hz), 2.59-2.53 (2H, m),2.53 (1H, d, J = 3.9 Hz).116UPLC-MS (ES+, final purity): 2.83 min, m/z 433.2 [M + H]+.1H NMR (400 MHz, DMSO- d6+ CF3CO2D) δ/ppm: 14.08 (1H, m), 10.66 (1H, s), 7.99 (1H, d, J = 5.9 Hz), 7.41 (1H, s), 7.02 (1H, d, J = 2.8 Hz), 6.96 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.91 (1H, d, J = 8.8 Hz), 6.30 (1H, d, J = 6.0 Hz), 4.49 (1H, dd, J = 11.0 Hz, 3.1 Hz), 4.34 (1H, dd, J = 11.0 Hz, 8.0 Hz), 3.84-3.71 (1H, m), 3.33- 3.19 (2H, m), 2.94 (2H, t, J =7.7 Hz), 2.65-2.59 (2H, m),2.55 (2H, dd, J = 8.4 Hz, 7.0Hz), 1.63-1.40 (3H, m), 0.90(6H, d, J = 6.3 Hz). Example 22. Synthesis of 5-[3-[4-(3,3-dimethylbutyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 115) Step 1—3-[4-(3,3-dimethylbut-1-ynyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol Tetrakis(triphenylphosphine)palladium(0) (54.3 mg, 0.05 mmol) was added to a stirred mixture of 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (200 mg, 0.47 mmol), 3,3-dimethyl-1-butyne (0.17 mL, 1.41 mmol), copper(I) iodide (9.0 mg, 0.05 mmol), K2CO3(130 mg, 0.94 mmol), monoglyme (2 mL) and water (0.5 mL) in a sealable vial at room temperature under a nitrogen atmosphere. The vial was sealed and the reaction heated to 90° C. and stirred for 18 hours. After this time, the reaction mixture was cooled to room temperature and the solvent removed in vacuo. The residue was taken up in DCM, filtered through a celite plug and the filter cake washed with DCM (10 mL). The filtrate was diluted with DCM (50 mL) and the solution washed with water sat aq. NaHCO3(10 mL), brine (50 mL), dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-10% EtOAc in petroleum ether to give 3-[4-(3,3-dimethylbut-1-ynyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (156.5 mg, 0.37 mmol, 78% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 2.04 min, m/z 427.6 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.13 (1H, s), 6.73 (1H, d, J=9.2 Hz), 6.64-6.60 (1H, m), 6.59-6.56 (1H, m), 5.37 (2H, s), 4.69 (1H, s), 4.43-4.37 (1H, m), 4.07 (1H, t, J=10.0 Hz), 3.60-3.55 (2H, m), 3.46-3.27 (2H, m), 2.95-2.88 (1H, m), 1.32 (9H, s), 0.95-0.88 (2H, m), 0.00 (9H, s). Step 2—3-[4-(3,3-dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol 3-[4-(3,3-dimethylbut-1-ynyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (233. mg, 0.55 mmol) was stirred in MeOH (10 mL) at room temperature under a nitrogen atmosphere followed by the addition of palladium, 10 wt. % on carbon powder, dry (30 mg). The reaction was fitted with a H2balloon and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 18 hours. The crude was filtered over celite, which was washed with MeOH (10 mL) and the filtrate concentrated in vacuo to give 3-[4-(3,3-dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (170 mg, 0.39 mmol, 72% yield) as a yellow solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.76 min, m/z 431.9 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 6.77 (1H, s), 6.67 (1H, d, J=8.8 Hz), 6.61 (1H, dd, J=8.8 Hz, 2.4 Hz), 6.56 (1H, d, J=2.4 Hz), 5.24-5.18 (2H, m), 4.33-4.27 (1H, m), 4.05 (1H, t, J=10.8 Hz), 3.57-3.52 (2H, m), 3.43-3.36 (1H, m), 3.30-3.21 (1H, m), 2.86-2.79 (1H, m), 2.57-2.51 (2H, m), 1.59-1.53 (2H, m), 0.97 (9H, s), 0.96-0.91 (2H, m), 0.00 (9H, s). Exchangeable proton not seen. Step 3—5-[3-[4-(3,3-dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one Potassium tert-butoxide (132.9 mg, 1.18 mmol) was added to a stirred solution of 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (65.6 mg, 0.39 mmol), 3-[4-(3,3-dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (170 mg, 0.39 mmol) and DMF (2 mL) at room temperature under a nitrogen atmosphere in a sealable vial. The vial was sealed and the reaction was heated to 80° C. and stirred for 72 hours. The reaction was cooled to room temperature and solvent removed in vacuo. The residue was partitioned between water (20 mL) and DCM (20 mL). The organic layer was separated, passed through a phase separator and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-5% MeOH in DCM to give 5-[3-[4-(3,3-dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (116 mg, 0.20 mmol, 51% yield) as a yellow solid. UPLCMS (ES+, short acidic): 1.76 min, m/z 577.5 [M+H]+. Step 4—5-[3-[4-(3,3-dimethylbutyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one Trifluoroacetic acid (0.5 mL, 6.53 mmol) was added to a stirred solution of 5-[3-[4-(3,3-dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (116 mg, 0.20 mmol) and DCM (2 mL) in a sealable vial at room temperature under a nitrogen atmosphere. The vial was sealed and the reaction was heated to 40° C. and stirred for 18 hours. After this time the reaction was cooled to room temperature and solvent removed in vacuo and the residue loaded on to an SCX-2 column using the minimum amount of MeOH. MeOH (10 mL) was passed through the column followed by NH3in MeOH (10 mL) to elute the product. Product containing fractions were concentrated in vacuo and the residue purified by column chromatography using an eluent of 0-5% MeOH in DCM to give 5-[3-[4-(3,3-dimethylbutyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (10 mg, 0.022 mmol, 11% yield) as a white solid. UPLC-MS (ES+, final purity): 2.94 min, m/z 447.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 11.67 (0.5H, s), 11.58 (0.5H, s), 10.45 (1H, s), 7.95 (1H, d, J=5.6 Hz), 6.99-6.97 (1H, m), 6.93-6.85 (2H, m), 6.76 (0.5H, s), 6.49 (0.5H, s), 6.26 (1H, d, J=5.6 Hz), 4.46-4.40 (1H, m), 4.02 (1H, t, J=10.8 Hz), 3.29-3.23 (1H, m), 3.18-3.10 (1H, m), 3.06-2.99 (1H, m), 2.93 (2H, t, J=7.2 Hz), 2.55-2.50 (2H, m, (under DMSO)), 2.44-2.36 (2H, m), 1.50-1.43 (2H, m), 0.93-0.91 (9H, m). Example 23. Synthesis of 5-[3-[3-[2-(trifluoromethyl)phenyl]-1H-pyrazol-5-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 35) Step 1—N-methoxy-N-methyl-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide Propylphosphonic anhydride (50% in EtOAc-1.31 mL, 2.2 mmol) was added to a stirred solution of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (500 mg, 1.47 mmol), N,O-dimethylhydroxylamine hydrochloride (158 mg, 1.62 mmol), triethylamine (0.31 mL, 2.2 mmol) and THF (100 mL) at rt under a nitrogen atmosphere. The reaction was heated to 65° C. and stirred for 18 hours, then cooled to rt and solvent removed in vacuo. The residue was partitioned between water (100 mL) and DCM (100 mL). The organic layer was separated, dried over Na2SO4, filtered and solvent removed in vacuo. The crude was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give N-methoxy-N-methyl-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide (337 mg, 0.88 mmol, 60% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.39 min, m/z 384.3 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 8.13 (1H, s), 7.99 (1H, d, J=6.0 Hz), 6.92-6.89 (1H, m), 6.87-6.83 (2H, m), 6.32 (1H, d, J=6.0 Hz), 4.48-4.43 (1H, m), 4.06 (1H, t, J=10.4 Hz), 3.79 (3H, s), 3.45-3.35 (1H, m), 3.26 (3H, s), 3.23-3.14 (1H, m), 3.08 (2H, t, J=7.2 Hz), 2.93-2.86 (1H, m), 2.72 (2H, t, J=7.2 Hz). Step 2—5-[3-[3-[2-(trifluoromethyl)phenyl]prop-2-ynoyl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one n-Butyllithium solution (2.5M in hexane—0.35 mL, 0.88 mmol) was added dropwise to a stirred solution of 2-ethynyl-α,α,α-trifluorotoluene (0.12 mL, 0.88 mmol) in THF (20 mL) at −78° C. under a nitrogen atmosphere. After stirring for 30 minutes a solution of N-methoxy-N-methyl-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide (168 mg, 0.44 mmol) in THF (10 mL) was slowly added and the resultant reaction allowed to stir at −78° C. for 1 hour, after which time it was warmed to rt and quenched with sat. aq. NH4Cl (100 mL). This mixture was extracted with EtOAc (2×50 mL), the combined organic layers dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-5% MeOH in DCM to give 5-[3-[3-[2-(trifluoromethyl)phenyl]prop-2-ynoyl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (163 mg, 0.33 mmol, 76% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.85 min, m/z 493.3 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 8.00 (1H, s), 7.97 (1H, d, J=6.0 Hz), 7.81-7.77 (2H, m), 7.65-7.62 (2H, m), 6.92-6.83 (3H, m), 6.32 (1H, d, J=5.6 Hz), 4.62-4.57 (1H, m), 4.46-4.40 (1H, m), 3.33-3.24 (2H, m), 3.16-3.11 (1H, m), 3.08 (2H, t, J=8.0 Hz), 2.72 (2H, t, J=8.0 Hz). Step 3—5-[3-[3-[2-(trifluoromethyl)phenyl]-1H-pyrazol-5-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one Hydrazine Hydrate (0.03 mL, 0.66 mmol) was added to a stirred solution of 5-[3-[3-[2-(trifluoromethyl)phenyl]prop-2-ynoyl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (163. mg, 0.3300 mmol) and t-BuOH (2 mL) at rt. The reaction was heated to 85° C. for 1 hour, after which time it was poured into sat. aq. NH4Cl (100 mL) and extracted with DCM (2×50 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-5% MeOH in DCM to give 5-[3-[3-[2-(trifluoromethyl)phenyl]-1H-pyrazol-5-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (112 mg, 0.22 mmol, 67% yield) as a white solid. UPLC-MS (ES+, final purity): 3.94 min, m/z 507.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 13.05 (0.6H, s), 13.00 (0.4H, s), 10.46 (1H, s), 7.95 (1H, d, J=5.6 Hz), 7.90-7.54 (4H, br m), 7.01-6.98 (1H, m), 6.95-6.86 (2H, m), 6.39-6.33 (1H, m), 6.28-6.23 (1H, m), 4.50-4.41 (1H, in), 4.19-4.04 (1H, in), 3.52-3.42 (1H, in), 3.21-3.01 (2H, in), 2.93 (2H, t, J=7.6 Hz), 2.54 (2H, t, J=7.6 Hz). The compounds in the table below were made in an analogous manner to Example 23, using the appropriate arylacetylene in place of 2-ethynyl-α,α,α-trifluorotoluene in step 2: Comp.NoStructure and NameData38UPLC-MS (ES+, final purity): 4.13 min, m/z 507.3 |M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 13.37 (0.3H, s), 13.09 (0.7H, s), 10.47 (1H, s), 8.04-7.92 (3H, m), 7.86- 7.72 (2H, m), 7.01-6.73 (4H, m), 6.27 (1H, d, J = 6.0 Hz), 4.50-4.42 (1H, m), 4.19-4.08 (1H, m), 3.50-3.39 (1H, m), 3.22-3.05 (2H, m), 2.93 (2H, t, J = 7.2 Hz), 2.54 (2H, t, J = 7.2 Hz).41UPLC-MS (ES+, final purity): 3.72 min, m/z 469.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 13.11-12-85 (1H, m), 10.47 (1H, s), 7.99-7.93 (1H, m), 7.39-7.26 (2.5H, m), 7.01-6.80 (4H, m), 6.71-7.61 (1H, m), 6.30-6.23 (1H, m), 5.85 (0.5H, s), 4.50-4.31 (1H, m), 4.18-4.04 (1H, m), 3.84- 3.77 (3H, m), 3.48-3.38 (1H, m), 3.17-3.06 (2H, m), 2.93 (2H, t, J = 7.6 Hz), 2.59-2.53 (2H, m, (partly under DMSO peak)).40UPLC-MS (ES+, final purity): 3.80 min, m/z 469.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.89-12.72 (1H, m), 10.46 (1H, s), 7.95 (1H, d, J = 5.6 Hz), 7.90-7.86 (0.2H, m), 7.70-7.62 (0.8 Hz, m), 7.38-7.25 (1H, m), 7.17- 6.86 (5H, m), 6.68-6.60 (1H, m), 6.26 (1H, d, J = 5.6 Hz), 4.50 (1H, m), 4.15-4.05 (1H, m), 3.91-3.81 (3H, m), 3.45- 3.36 (1H, m), 3.16-3.06 (2H, m), 2.93 (2H, t, J = 8.0 Hz),2.93 (2H, t, J = 8.0 Hz, partlyunder DMSO)).43UPLC-MS (ES+, final purity): 3.67 min, m/z 469.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.96 (0.5H, s), 12.72 (0.5H, s), 10.46 (1H, s), 7.96 (1H, d, J = 5.6 Hz), 7.73-7.61 (2H, m), 7.05-6.85 (5H, m), 6.57-6.50 (1H, m), 6.27 (1H, d, J = 5.6 Hz), 4.49-4.40 (1H, m), 4.16-4.04 (1H, m), 3.82-3.75 (3H, m), 3.45-3.37 (1H, m), 3.18-3.06 (2H, m), 2.93 (2H, t, J = 7.6Hz), 2.53 (2H, t, J = 7.6 Hz,(partly under DMSO)).36UPLC-MS (ES+, final purity): 3.88 min, m/z 473.2, 475.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 13.06 (1H, m), 10.47 (1H, m), 7.95 (1H, d, J = 6.0 Hz), 7.80-7.31 (4H, m), 7.01-6.98 (1H, m), 6.96- 6.86 (2H, m), 6.64 (1H, s), 6.28-6.24 (1H, m), 4.52-4.42 (1H, m), 4.21-4.07 (1H, m), 3.53-3.35 (1H, m), 3.19-3.06 (2H, m), 2.94 (2H, t, J = 8.0 Hz), 2.57-2.53 (2H, m, (under DMSO peak)).37UPLC-MS (ES+, final purity): 4.00 min, m/z 473.2, 475.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 13.23 (0.4H, s), 13.00 (0.6H, s), 10.47 (1H, s), 7.96 (1H, d, J = 5.6 Hz), 7.84-7.67 (2H, m), 7.51-7.32 (2H, m), 7.00- 6.97 (1H, m), 6.95-6.86 (2H, m), 6.80-6.70 (1H, m), 6.26 (1H, d, J = 5.6 Hz), 4.49-4.41 (1H, m), 4.18-4.05 (1H, m), 3.48-3.36 (1H, m), 3.17-3.05 (2H, m), 2.93 (2H, t, J = 7.6Hz), 2.53 (2H, t, J = 7.6 Hz,(partly under DMSO)).39UPLC-MS (ES+, final purity): 4.01 min, m/z 473.2, 475.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 13.19 (0.4H, s), 12.94 (0.6H, s), 10.47 (1H, s), 7.96 (1H, d, J = 5.6 Hz), 7.82-7.71 (2H, m), 7.57-7.42 (2H, m), 7.00- 6.97 (1H, m), 6.95-6.86 (2H, m), 6.73-6.62 (1H, m), 6.26 (1H, d, J = 5.6 Hz), 4.48-4.41 (1H, m), 4.19-4.05 (1H, m), 3.49-3.38 (1H, m), 3.19-3.05(2H, m), 2.93 (2H, t, J = 7.6Hz), 2.53 (2H, t, J = 7.6 Hz,(partly under DMSO)).42UPLC-MS (ES+, final purity): 4.11 min, m/z 507.3 [M + H]+.1H NMR (400 MHz, DMSO- d6,) δ/ppm: 13.33 (0.3H, s), 13.05 (0.7H, s), 10.47 (1H, s), 8.13-8.00 (2H, m), 7.96 (1H, d, J = 5.6 Hz), 7.72-7.62 (2H, m), 7.01-6.98 (1H, m), 6.96-6.86 (2.3H, m), 6.81- 6.78 (0.7H, m), 6.26 (1H, d, J = 5.6 Hz), 4.50-4.43 (1H, m), 4.18-4.06 (1H, m), 3.48- 3.35 (1H, m), 3.18-3.06 (2H, m), 2.93 (2H, t, J = 7.6 Hz),2.53 (2H, t, J = 7.6 Hz, (partlyunder DMSO peak)). Example 24. Synthesis of 5-[3-[6-(trifluoromethyl)-1H-benzimidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 22) 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (246 mg, 0.65 mmol) was added to a stirred solution of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (200 mg, 0.59 mmol), 3,4-diaminobenzotrifluoride (114 mg, 0.65 mmol), DIPEA (0.31 mL, 1.76 mmol) and DMF (3 mL) at rt under a nitrogen atmosphere and stirred for 1 hour. Solvent was removed in vacuo and the residue partitioned between water (50 mL) and DCM (50 mL). The organic layer was separated, dried over Na2SO4, filtered and solvent removed in vacuo. The residue was then stirred at 80° C. in acetic acid (3 mL) for 18 hours, after which time the reaction was cooled to rt and solvent removed in vacuo. The residue was partitioned between sat. aq. NaHCO3(100 mL) and DCM (100 mL) and the organic layer separated, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography using an eluent of 0-5% MeOH in DCM to give 5-[3-[6-(trifluoromethyl)-1H-benzimidazol-2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (75 mg, 0.16 mmol, 26% yield) as a white solid. UPLC-MS (ES+, final purity): 3.41 min, m/z 481.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.92 (1H, br s), 10.48 (1H, s), 7.95 (1H, d, J=9.2 Hz), 7.89-7.76 (1H, m), 7.76-7.70 (1H, m), 7.50 (1H, d, J=8.8 Hz), 7.06-7.03 (1H, m), 6.93 (1H, dd, J=9.2 Hz, 2.8 Hz), 6.89 (1H, d, 8.8 Hz), 6.27 (1H, d, J=6.0 Hz), 4.65 (1H, m), 4.33 (1H, t, J=9.2 Hz), 3.71-3.62 (1H, m), 3.38-3.22 (2H, m), 2.93 (2H, t, J=8.4 Hz), 2.53 (2H, t, J=8.4 Hz, (partly under DMSO)). The compounds in the table below were made in an analogous manner, using the appropriate diamine (either commercially available or synthesised) in place of 3,4-diaminobenzotrifluoride: Comp.NoStructure and NameData23UPLC-MS (ES+, final purity): 3.17 min, m/z 449.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.68 (1H, s), 10.47 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.68-7.53 (2H, m), 7.02 (1H, d, J = 2.8 Hz), 6.92 (1H, dd, J = 8.4 Hz, 2.4 Hz), 6.88 (1H, d, J = 8.4 Hz), 6.26 (1H, d, J = 6.0 Hz), 4.61-4.56 (1H, m), 4.27 (1H, dd, J = 11.2 Hz, 9.6 Hz), 3.63-3.55 (1H, m), 3.30-3.17 (2H, m), 2.93 (2H, t, J = 7.6 Hz), 2.53 (2H, t, J = 7.6 Hz, (partly under DMSO)).24UPLC-MS (ES+, final purity): 3.11 min, m/z, 438.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 13.02 (1H, s), 10.47 (1H, s), 8.10 (1H, s), 7.97 (1H, d, J = 5.6 Hz), 7.70 (1H, d, J = 8.4 Hz), 7.57 (1H, dd, J = 8.4 Hz, 1.6 Hz), 7.04 (1H, d, 2.8 Hz), 6.93 (1H, dd, J = 8.4 Hz, 2.4 Hz), 6.89 (1H, d, J = 8.4 Hz), 6.27 (1H, d, 5.6 Hz), 4.64 (1H, m), 4.32 (1H, dd, J = 10.8 Hz, 8.8 Hz), 3.71-3.63 (1H, m), 3.31-3.21 (2H, m), 2.94 (2H, t, J = 8.0 Hz), 2.54 (2H, t, J = 8.0 Hz, (partly under DMSO)).25UPLC-MS (ES+, final purity): 3.13 min, m/z 447.2, 449.2 [M + H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.66 (1H, br s), 10.48 (1H, s), 7.96 (1H, d, J = 5.6 Hz), 7.65-7.50 (2H, m), 7.23-7.17 (1H, m), 7.03 (1H, d, J = 2.8 Hz), 6.93 (1H, dd, J = 8.8 Hz, 2.4 Hz), 6.89 (1H, d, J = 8.8 Hz), 6.27 (1H, d, J = 5.6 Hz), 4.62-4.57 (1H, m), 4.28 (1H, dd, J = 11.2 Hz, 9.6 Hz), 3.65-3.55 (1H, m), 3.31-3.18 (2H, m), 2.94 (2H, t, J = 7.6 Hz), 2.54 (2H, t, J = 7.6 Hz, (partly under DMSO)).26UPLC-MS (ES+, final purity): 2.52 min, m/z 456.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.14 (1H, br s), 10.47 (1H, s), 7.96 (1H, d, J = 5.2 Hz), 7.35 (1H, d, J = 8.8 Hz), 7.02 (1H, d, J = 2.8 Hz), 6.92 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.88 (1H, d, J = 8.8 Hz), 6.78-6.73 (2H, m), 6.27 (1H, d, J = 5.6 Hz), 4.59-4.54 (1H, m), 4.23 (1H, dd, J = 10.8 Hz, 9.6 Hz), 3.56-3.48 (1H, m), 3.30-3.15 (2H, m), 2.94 (2H, t, J = 7.2 Hz), 2.88 (6H, s), 2.54 (2H, t, J = 7.2 Hz, (partly under DMSO)).27UPLC-MS (ES+, final purity): 3.62 min, m/z 481.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 13.01-12.80 (1H, m), 10.48 (1H, s), 7.93-7.89 (1.4H, m), 7.82-7.78 (0.6H, m), 7.57-7.48 (1H, m), 7.38- 7.32 (1H, m), 7.07-7.04 (1H, m), 6.96-6.90 (2H, m), 6.29- 6.27 (1H, m), 4.66-4.59 (1H, m), 4.27 (1H, t, J = 10.4 Hz), 3.72-3.62 (1H, m), 3.40-3.35 (1H, m), 3.28-3.19 (1H, m), 2.94 (2H, t, J = 7.6 Hz), 2.56- 2.53 (2H, m, (partly under DMSO)).29UPLC-MS (ES+, final purity): 2.77 min, m/z 441.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.18 (1H, s), 10.47 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.33 (1H, s), 7.24 (1H, s), 7.02 (1H, d, J = 8.0 Hz), 6.92 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.88 (1H, d, J = 8.8 Hz), 6.27 (1H, d, J = 6.4 Hz), 4.61-4.55 (1H, m), 4.24 (1H, t, J = 10.4 Hz), 3.57-3.49 (1H, m), 3.30-3.15 (2H, m), 2.94 (2H, t, J = 7.2 Hz), 2.54 (2H, t, J = 7.2 Hz, (partially under DMSO)), 2.31 (3H, s), 2.85 (3H, s).30UPLC-MS (ES+, final purity): 4.03 min, m/z 515.2 [M + H]+.1H-NMR (400 MHz, DMSO- d6) δ/ppm: 13.29 (1H, br s), 10.47 (1H, s), 7.99-7.86 (2H, m), 7.66-7.58 (1H, m), 7.09- 7.00 (1H, m), 6.96-6.87 (2H, m), 6.30-6.23 (1H, m), 4.66- 4.58 (1H, m), 4.38-4.27 (1H, m), 3.74-3.64 (1H, m), 3.41- 3.37 (1H, m (partly under water)), 3.28-3.19 (1H, m), 2.96-2.89 (2H, m), 2.56-2.53 (2H, m, (partly under DMSO)).31UPLC-MS (ES+, final purity): 3.69 min, m/z 495.3 [M + H]+.1H-NMR (400 MHz, DMSO- d6) δ/ppm: 12.81 (1H, br s), 10.46 (1H, s), 7.96 (1H, d, J = 5.6 Hz), 7.75 (0.6H, br s), 7.64 (0.4H, br s), 7.31 (1H, br s), 7.04 (1H, d, J = 2.4 Hz), 6.95-6.88 (2H, m), 6.27 (1H, d, J = 5.6 Hz), 4.68-4.55 (1H, m), 4.29 (1H, t, J = 10.0 Hz), 3.70-3.58 (1H, m), 3.40-3.35 (1H, m), 3.23 (1H, dd, J = 16.4 Hz, 5.6 Hz), 2.93 (2H, t, J = 7.6 Hz), 2.58 (3H, s), 2.53 (2H, t, J = 7.6 Hz, (partly under DMSO peak)).32UPLC-MS (ES+, final purity): 3.26 min, m/z 482.3 [M + H]+.1H-NMR (400 MHz, DMSO- d6) δ/ppm: 13.42 (1H, br s), 10.46 (1H, s), 8.98 (1H, s), 8.03 (1H, s), 7.96 (1H, d, J = 5.6 Hz), 7.04 (1H, d, J = 2.8 Hz), 6.93 (1H, dd, J = 8.8 Hz, 2.8 Hz), 6.89 (1H, d, J = 8.8 Hz), 6.27 (1H, d, J = 5.6 Hz), 4.65-4.59 (1H, m), 4.37-4.30 (1H, m), 3.75-3.66 (1H, m), 3.39-3.35 (1H, m, (partly under water)), 3.29-3.22 (1H, m), 2.93 (2H, t, J = 7.6 Hz), 2.56-2.53 (2H, m, (party under DMSO)).34UPLC-MS (ES+, final purity): 2.72 min, m/z 538.2 [M + H]+.1H-NMR (400 MHz, DMSO- d6) δ/ppm: 12.93 (0.8H, s), 12.84 (0.2H, s), 10.46 (1H, s), 8.49 (0.2H, d, J = 4.4 Hz), 8.26 (0.2H, d, J = 8.0 Hz), 7.95 (0.8H, d, J = 6.0 Hz), 7.79 (0.2H, s), 7.69 (0.8H, s), 7.50 (0.2H, s), 7.45 (0.8H, s), 7.28 (0.2H, dd, J = 8.4 Hz, 4.4 Hz), 7.05 (0.8H, s), 6.96- 6.86 (1.8H, m), 6.27 (0.8H, d, J = 5.6 Hz), 6.23 (0.2H, d, J = 6.0 Hz), 4.64-4.56 (1H, m), 2.46 (1H, t, J = 10.4 Hz), 3.71-3.60 (3H, m), 3.41-3.37(1H, m (partly under water)),3.26-3.18 (1H, m), 2.93 (2H,t, J = 7.6 Hz), 2.53 (2H, t, J =7.6 Hz, (partly under DMSO)),2.22 (6H, s).28UPLC-MS (ES+, final purity): 2.75 min, m/z 593.4 [M + H]+.1H-NMR (400 MHz, DMSO- d6) δ/ppm: 12.89 (0.75H, br s), 12.81 (0.25H, br s), 10.46 (1H, s), 8.42 (0.25H, dd, J = 4.0 Hz, 1.2 Hz), 8.19 (0.25H, dd, J = 8.4 Hz, 1.2 Hz), 7.99-7.78 (0.75H, m), 7.81-7.58 (1H, m), 7.50-7.34 (1H, m), 7.22 (0.25H, dd, J = 8.4 Hz, 4.4 Hz), 7.04 (0.75H, s), 6.97-6.81 (1.75H, m), 6.27 (0.75H, d, J = 6.0 Hz), 6.23 (0.25H, d, J = 6.0 Hz), 4.64-4.57 (1H, m), 4.26 (1H, t, J = 10.0 Hz),3.70-3.58 (3H, m), 3.41-3.37(1H, m, (partly under water)),3.26-3.19 (1H, m), 2.93 (2H,t, J = 7.6 Hz), 2.55-2.53, (2H,m (partly under DMSO)), 2.46(8H, br s), 2.19 (3H, s).33UPLC-MS (ES+, final purity): 2.79 min, m/z 538.4 [M + H]+.1H-NMR (400 MHz, DMSO- d6) δ/ppm: 12.94 (0.3H, br s), 11.99 (0.7H, br s), 10.47 (1H, s), 7.96 (1H, d, J = 6.0 Hz), 7.83 (1H, br s), 7.45 (1H, br s), 7.04 (1H, d, J = 2.4 Hz), 6.95-6.88 (2H, m), 6.27 (1H, d, J = 6.0 Hz), 4.64-4.58 (1H, m), 4.28 (1H, t, J = 10.0 Hz), 3.80 (2H, br s), 3.73-3.62 (1H, m), 3.34 (1H, dd, J = 16.4 Hz, 10.0 Hz), 3.23 (1H, dd, J = 16.4 Hz, 5.6 Hz), 2.93 (2H, t, J = 7.6 Hz), 2.54 (2H, t, J = 7.6 Hz, (partly under DMSO)), 2.27 (6H, br s). Example 25. Synthesis of 5-[3-(3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 134) Step 1—N-(4-amino-3-pyridyl)-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (307.2 mg, 0.81 mmol) was added to a stirred solution of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (250 mg, 0.73 mmol), DIPEA (0.38 mL, 2.2 mmol), 3,4-diaminopyridine (88.2 mg, 0.81 mmol) and DMF (10 mL) under a nitrogen atmosphere and stirred at rt for 1h. Solvent was removed in vacuo and the residue partitioned between water (20 mL) and DCM+few drops of MeOH (20 mL), followed by extraction of the aqueous phase with EtOAc (20 mL). The organic layer was separated, dried over Na2SO4, filtered and the solvent removed in vacuo. The crude was therefore purified by column chromatography using as eluent a gradient 0-5% MeOH in DCM to give N-(4-amino-3-pyridyl)-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide (195 mg, 0.45 mmol, 62% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.13 min, m/z 432.2 [M+H]+. Step 2—5-[3-(3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one To N-(4-amino-3-pyridyl)-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxamide (144 mg, 0.33 mmol) in 1,4-dioxane (3 mL) was added HCl (4M in dioxane—0.67 mL, 2.68 mmol) in a sealed vial. The reaction was irradiated at 100° C. for 2 hours, after which time the solvent was removed under reduce pressure. The reaction was quenched with sat. aq. NaHCO3and extracted with DCM+MeOH. The organic phase was dried over Na2SO4, filtered and the solvent removed in vacuo. The crude was purified by automated column chromatography using as eluent a gradient 0-12% MeOH in DCM to give 5-[3-(3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (42.5 mg, 0.10 mmol, 31% yield) as a white solid. UPLC-MS (ES+, final purity): 2.30 min, m/z 414.2 [M+H]+.1H-NMR (400 MHz, DMSO-d6) δ/ppm: 13.00-12.79 (1H, m), 10.47 (1H, s), 8.87 (1H, s), 8.28 (1H, d, J=5.2 Hz), 7.96 (1H, d, J=5.6 Hz), 7.61-7.50 (1H, m), 7.03 (1H, d, J=2.4 Hz), 6.95-6.87 (2H, m), 6.27 (1H, d, J=5.6 Hz), 4.64-4.58 (1H, m), 4.31 (1H, t, J=10.0 Hz), 3.70-3.60 (1H, m), 3.36-3.19 (2H, m, (partly under water)), 2.93 (2H, t, J=7.6 Hz), 2.53 (2H, t, 8.0 Hz, (partly under DMSO)). Example 26. Synthesis of 5-[[2-(3-phenyl-1H-1,2,4-triazol-5-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one hydrochloride (Compound 44) Step 1—2-(5-bromo-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-ol 3,5-dibromo-1-tetrahydropyran-2-yl-1,2,4-triazole (1.042 g, 3.35 mmol), 1,2,3,4-tetrahydroisoquinolin-7-ol (500 mg, 3.35 mmol), L-proline (38.6 mg, 0.34 mmol), copper(I) iodide (63.8 mg, 0.34 mmol), K2CO3(1.39 g, 10.05 mmol) and DMF (3 mL) were combined in a sealed and heated to 120° C. for 18 hours. The reaction was allowed to cool to rt, solvent removed in vacuo and the residue partitioned between water (100 mL) and DCM (100 mL). The organic layer was separated, dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 2-(5-bromo-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-ol (508 mg, 1.34 mmol, 40% yield) as a brown solid. UPLC-MS (ES+, short acidic): 1.64 min, m/z 379.1, 381.1 [M+H]+. Step 2—2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-ol [1,1′-Bis(diphenylphosphino)ferrocene]Palladium(II) chloride dichloromethane complex (173.6 mg, 0.21 mmol) was added to a stirred mixture of 2-(5-bromo-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-ol (806 mg, 2.13 mmol), phenylboronic acid (311 mg, 2.55 mmol), K2CO3(881 mg, 6.38 mmol), 1,4-dioxane (9 mL) and water (1 mL) at rt under an inert atmosphere. The reaction was heated to 80° C. for 3 hours, after which time it was cooled to rt and solvent removed in vacuo. The residue was suspended in DCM (10 mL), filtered over celite, which was washed with DCM (10 mL). The residue was concentrated in vacuo and purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-ol (510 mg, 1.35 mmol, 64% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.85 min, m/z 399.3 [M+Na]+, 293.2 [M-THP]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 9.23 (1H, s), 7.98-7.93 (2H, m), 7.48-7.37 (3H, m), 6.98 (1H, d, 8.4 Hz), 6.61 (1H, dd, J=8.4 Hz, 2.8 Hz), 6.57 (1H, d, J=2.8 Hz), 5.31 (1H, dd, J=10.0 Hz, 2.0 Hz), 4.50-4.46 (1H, m), 4.35-4.31 (1H, m), 4.10-4.03 (1H, m), 3.72-3.64 (2H, m), 3.45-3.35 (1H, m), 3.10-3.01 (1H, m), 2.85-2.76 (1H, m), 2.06-1.95 (2H, m), 1.90-1.53 (4H, m). Step 3—5-[[2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one 2-(5-Phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-ol (510 mg, 1.35 mmol), 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (225.1 mg, 1.35 mmol), K2CO3(936 mg, 6.77 mmol) and DMSO (10 mL) were combined in a sealed vial and stirred at 110° C. for 18 hours. The reaction was cooled to rt and the solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 5-[[2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (192 mg, 0.37 mmol, 27% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.92 min, m/z 523.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.56 (1H, s), 8.00-7.93 (2H, m), 7.48-7.39 (3H, m), 7.31-7.28 (1H, m), 7.03 (1H, d, J=2.8 Hz), 6.99 (1H, dd, J=8.4 Hz, 2.8 Hz), 6.34 (1H, d, =5.6 Hz), 5.34 (1H, dd, J=10.0 Hz, 2.4 Hz), 4.56 (1H, d, J=16.0 Hz), 4.44 (1H, d, J=16.0 Hz), 4.08-4.00 (1H, m), 3.81-3.65 (2H, m), 3.50-3.42 (1H, m), 3.25-3.14 (1H, m), 3.00-2.88 (3H, m), 2.55-2.50 (3H, m), 1.90-1.54 (6H, m). Step 4—5-[[2-(3-phenyl-1H-1,2,4-triazol-5-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one hydrochloride HCl (4M in 1,4-dioxane—0.37 mL, 1.47 mmol) was added to 5-[[2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (192. mg, 0.37 mmol) in DCM (10 mL) at rt under a nitrogen atmosphere and the reaction was stirred for 18 hours. Solvent was removed in vacuo and the residue purified by column chromatography using an eluent of 0-10% MeOH in DCM to give 5-[[2-(3-phenyl-1H-1,2,4-triazol-5-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one hydrochloride (50 mg, 0.11 mmol, 29% yield) as a yellow solid. UPLC-MS (ES+, final purity): 3.39 min, m/z 439.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.63 (1H, s), 8.04-7.95 (3H, m), 7.52-7.44 (3H, m), 7.30 (1H, d, J=7.6 Hz), 7.05-6.99 (2H, m), 6.39 (1H, d, J=6.0 Hz), 4.72-4.67 (2H, m), 3.83-3.77 (2H, m), 2.99-2.91 (3H, m), 2.59-2.54 (3H, m). Broad peak at 5.4 ppm will include 1 exchangeable and HCl salt. Example 27. Synthesis of 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-N-phenyl-3,4-dihydro-1H-isoquinoline-2-carboxamide (Compound 46) Step 1—benzyl 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-3,4-dihydro-1H-isoquinoline-2-carboxylate Benzyl 7-hydroxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (500 mg, 1.76 mmol), 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (0.29 g, 1.76 mmol) and K2CO3(0.91 g, 6.55 mmol) were suspended in DMSO (50 mL) and heated to 110° C. 18 hours. The reaction was cooled to rt, carefully poured an aq. solution of citric acid (1.36 g, 7.06 mmol in 50 mL of H2O), which caused fizzing, and left stirring for 1 hour. Water (200 mL) was added and the resulting mixture extracted with DCM (2×100 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-75% EtOAc in petroleum ether to give benzyl 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-3,4-dihydro-1H-isoquinoline-2-carboxylate (510 mg, 1.19 mmol, 67% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.69 min, m/z 430.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.50 (1H, s), 7.97 (1H, d, J=5.6 Hz), 7.41-7.35 (5H, m), 7.25 (1H, d, J=8.4 Hz), 7.03 (1H, d, J=6.8 Hz), 6.96 (1H, dd, J=8.0 Hz, 2.4 Hz), 6.31 (1H, d, J=5.5 Hz), 5.13 (2H, s), 4.66-4.55 (2H, m), 3.70-3.62 (2H, m), 2.90 (2H, t, J=7.2 Hz), 2.82 (2H, J=5.6 Hz), 2.55-2.53 (2H, m (partly under DMSO)). Step 2—5-(1,2,3,4-tetrahydroisoquinolin-7-yloxy)-3,4-dihydro-1H-1,8-naphthyridin-2-one Palladium, 10 wt. % on carbon powder, dry (50 mg) was added to a stirred solution of benzyl 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-3,4-dihydro-1H-isoquinoline-2-carboxylate (510. mg, 1.19 mmol) in EtOAc (20 mL) and MeOH (5 mL) at rt under a nitrogen atmosphere. The reaction was fitted with a H2balloon and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 24 hours. The crude was filtered over celite, which was washed with MeOH (10 mL) and the filtrate concentrated in vacuo to give 5-(1,2,3,4-tetrahydroisoquinolin-7-yloxy)-3,4-dihydro-1H-1,8-naphthyridin-2-one (222 mg, 0.75 mmol, 63% yield) as a yellow solid. The product was used in the next step without further purification. UPLC-MS (ES+, short acidic): 0.91 min, m/z 296.2 [M+H]+.1H NMR (400 MHz, CDCl3) a/ppm: 8.17 (1H, s), 7.99 (1H, d, J=6.0 Hz), 7.15 (1H, d, J=8.4 Hz), 6.86 (1H, dd, J=8.4 Hz, 2.8 Hz), 6.76 (1H, d, J=2.8 Hz), 6.34 (1H, d, J=6.0 Hz), 4.03 (2H, s), 3.18 (2H, t, J=6.0 Hz), 3.08 (2H, t, J=8.0 Hz), 2.83 (2H, t, J=6.0 Hz), 2.72 (2H, t, J=8.0 Hz). Exchangeable proton not seen. Step 3—7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-N-phenyl-3,4-dihydro-1H-isoquinoline-2-carboxamide Phenyl isocyanate (0.02 mL, 0.19 mmol) was added to a stirred solution of 5-(1,2,3,4-tetrahydroisoquinolin-7-yloxy)-3,4-dihydro-1H-1,8-naphthyridin-2-one (50 mg, 0.17 mmol) and DIPEA (0.06 mL, 0.34 mmol) in DCM (5 mL) at rt under a nitrogen atmosphere. The reaction stirred for 1 hour, after which time it was diluted with water (20 mL) and DCM (15 mL). The organic layer was separated, passed through a phase separator and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-N-phenyl-3,4-dihydro-1H-isoquinoline-2-carboxamide (29 mg, 0.07 mmol, 41% yield) as a white solid. UPLC-MS (ES+, final purity): 3.35 min, m/z 415.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.51 (1H, s), 8.58 (1H, s), 7.98 (1H, d, J=5.6 Hz), 7.50-7.46 (2H, m), 7.30-7.22 (3H, m), 7.00-6.92 (3H, m), 6.34 (1H, d, J=5.6 Hz), 4.64 (2H, s), 3.73 (2H, t, J=6.0 Hz), 2.94-2.85 (4H, m), 2.55-2.53 (2H, m, (partly under DMSO peak)). The compounds in the table below were made in an analogous manner, using the appropriate aryl isocyanate in place of phenyl isocyanate in step 3: Comp.NoStructure and NameData49UPLC-MS (ES+, final purity): 3.63 min, m/z 483.4 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 10.51 (1H, s), 8.30 (1H, s), 7.98 (1H, d, J = 6.0 Hz), 7.71-7.62 (2H, m), 7.48- 7.39 (2H, m), 7.31-7.27 (1H, m), 7.00-6.96 (2H, m), 6.33 (1H, d, J = 6.0 Hz), 4.63 (2H, s), 3.72 (2H, t, J = 6.4 Hz), 2.94-2.85 (4H, m), 2.57-2.53 (2H, m, (under DMSO peak)).47UPLC-MS (ES+, final purity): 3.91 min, m/z 483.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 10.51 (1H, s), 8.99 (1H, s), 7.98 (1H, d, J = 6.0 Hz), 7.71 (2H, d, J = 8.8 Hz), 7.60 (2H, d, J = 8.8 Hz), 7.28 (1H, d, J = 8.4 Hz), 7.00-6.96 (2H, m), 6.33 (1H, d, J = 6.0 Hz), 4.67 (2H, s), 3.75 (2H, t, J = 5.6 Hz), 2.94-2.86 (4H, m), 2.54-2.52 (2H, m, (under DMSO peak)).50UPLC-MS (ES+, final purity): 3.90 min, m/z 483.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 10.51 (1H, s), 8.93 (1H, s), 7.98 (1H, d, J = 6.0 Hz), 7.94 (1H, d, J = 7.6 Hz), 7.78 (1H, d, J = 8.0 Hz), 7.48 (1H, t, J = 8.0 Hz), 7.31-7.26 (2H, m), 7.02-6.96 (2H, m), 6.34 (1H, d, J = 6.0 Hz), 4.66 (2H, s), 3.74 (2H, t, J = 6.0 Hz), 2.95-2.86 (4H, m), 2.56- 2.52 (2H, m, (under DMSO peak)).48UPLC-MS (ES+, final purity): 3.34 min, m/z 429.3 [M + H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 10.50 (1H, s), 7.97 (1H, d, J = 6.0 Hz), 7.32-7.15 (7H, m), 6.97-6.91 (2H, m), 6.32 (1H, d, J = 6.0 Hz), 4.53 (2H, s), 4.27 (2H, d, J = 6.0 Hz), 3.62 (2H, t, J = 6.0 Hz), 2.91 (2H, t, J = 7.6 Hz), 2.80 (2H, t, J = 6.0 Hz), 2.55 (2H, m, (under DMSO peak)). Exchangeable proton not seen. Example 28. Synthesis of 5-[[2-(1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 159) Step 1—2-(1H-benzimidazol-2-yl)-7-benzyloxy-3,4-dihydro-1H-isoquinoline 7-benzyloxy-1,2,3,4-tetrahydroisoquinoline (158 mg, 0.66 mmol), 2-chlorobenzimidazole (101 mg, 0.66 mmol) and 1,4-dioxane (1.2 mL) were mixed in a sealed vial and irradiated to 180° C. for 1 hour. The residue was dissolved in DCM, washed with sat. aq. NaHCO3and then dried through a phase separator. The solvent was removed in vacuo and the residue was purified by column chromatography using as eluent a gradient 0-50% EtOAc in petroleum ether to give 2-(1H-benzimidazol-2-yl)-7-benzyloxy-3,4-dihydro-1H-isoquinoline (175 mg, 0.49 mmol, 75% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.50 min, m/z 356.5 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 11.45 (1H, s), 7.49-7.27 (5H, m), 7.20 (2H, dd, J=14.1 Hz, 7.6 Hz), 7.09 (1H, d, J=8.3 Hz), 6.99-6.81 (4H, m), 5.10 (2H, s), 4.68 (2H, s), 3.78 (2H, t, J=5.9 Hz), 2.84 (2H, t, J=5.9 Hz). Step 2—2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane Sodium hydride (60% dispersed in mineral oil—38.3 mg, 0.96 mmol) was added to a stirred solution of 2-(1H-benzimidazol-2-yl)-7-benzyloxy-3,4-dihydro-1H-isoquinoline (170 mg, 0.48 mmol) and DMF (100 mL) at rt under a nitrogen atmosphere. After 20 minutes, 2-(trimethylsilyl)ethoxymethyl chloride (0.13 mL, 0.72 mmol) was added and the mixture was stirred for 18 hours. The reaction was quenched with water (1 mL) and solvent removed in vacuo. Water (20 mL) was added followed by extraction with DCM (2×20 mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-40% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (200 mg, 0.41 mmol, 86% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.93 min, m/z 486.9 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 7.64-7.55 (1H, m), 7.45-7.28 (6H, m), 7.24-7.15 (2H, m), 7.10 (1H, d, J=8.4 Hz), 6.85 (1H, dd, J=8.4 Hz, 2.7 Hz), 6.76 (1H, d, J=2.6 Hz), 5.34 (2H, s), 5.05 (2H, s), 4.60 (2H, s), 3.78-3.66 (4H, m), 3.02 (2H, t, J=5.9 Hz), 1.01-0.96 (2H, m), 0.00 (9H, s). Step 3—2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-ol 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (195 mg, 0.40 mmol) was dissolved in MeOH (4 mL) and Pd(OH)2(56 mg, 0.40 mmol) added under inert atmosphere. The reaction was fitted with a H2balloon and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere overnight. The crude was filtered over celite, flushed with MeOH and the filtrate concentrated in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-50% EtOAc in petroleum ether to give 2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-ol (120 mg, 0.30 mmol, 76% yield) as a colourless gum which crystallised on standing. UPLC-MS (ES+, short acidic): 1.58 min, m/z 396.6 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 8.45 (1H, s), 7.61-7.55 (1H, m), 7.33-7.28 (1H, m), 7.21-7.14 (2H, m), 6.93 (1H, d, J=8.3 Hz), 6.68 (1H, dd, J=8.3 Hz, 2.6 Hz), 6.63 (1H, d, J=2.5 Hz), 5.35 (2H, s), 4.57 (2H, s), 3.81-3.69 (4H, m), 2.92 (2H, t, J=5.8 Hz), 1.03-0.96 (2H, m), 0.00 (9H, s). Step 4—5-[[2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one Potassium tert-butoxide (37.4 mg, 0.33 mmol) was added to a stirred solution of 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (50.4 mg, 0.30 mmol) and 2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-ol (120 mg, 0.30 mmol) in DMF (2 mL) and the reaction was heated at 80° C. for 18 hours. The reaction was cooled to rt and the solvent removed in vacuo. The residue was partitioned between EtOAc and water; the organics were washed with saturated brine, dried over a phase separator and the solvent removed in vacuo. The residue was purified by column chromatography eluting with 25-100% EtOAc in petroleum ether to give 5-[[2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (94 mg, 0.17 mmol, 57% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.65 min, m/z 542.5 [M+H]+.1H NMR (400 MHz, CDCl3) δ/ppm: 8.69 (1H, s), 8.01 (1H, d, J=5.9 Hz), 7.63-7.58 (1H, m), 7.34-7.30 (1H, m), 7.25-7.16 (3H, m), 6.93-6.85 (2H, m), 6.35 (1H, d, J=5.9 Hz), 5.36 (2H, s), 4.65 (2H, s), 3.82-3.72 (4H, m), 3.13-3.02 (4H, m), 2.70 (2H, dd, J=8.4 Hz, 7.0 Hz), 1.03-0.96 (2H, m), 0.00 (9H, s). Step 5—5-[[2-(1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one Trifluoroacetic acid (0.64 mL, 8.31 mmol) was added to a stirred solution of 5-[[2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (90 mg, 0.17 mmol) in DCM (2 mL) in a sealed vial and heated to 40° C. for 18 hours, after which time the solvent was removed in vacuo. The residue was loaded into an SCX-2 column and flushed at first with MeOH (2×10 mL) and then NH3in MeOH (10 mL) to elute the product. The reside was then purified by column chromatography using as eluent a gradient 1-8% MeOH in DCM to give a colourless gum, which was triturated in MeCN/Et20 to give a white solid. The solid was filtered, washed with Et20 and dried to give 5-[[2-(1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (29 mg, 0.070 mmol, 42% yield) as a white solid. UPLC-MS (ES+, final purity): 2.52 min, m/z 412.4 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.21 (1H, s), 10.52 (1H, s), 7.99 (1H, d, J=5.8 Hz), 7.33-7.25 3H, m), 7.07-6.97 (4H, m), 6.35 (1H, d, J=5.8 Hz), 4.76 (2H, s), 3.85 (2H, t, J=5.9 Hz), 2.98 (2H, t, J=5.9 Hz), 2.92 (2H, t, J=7.9 Hz), 2.57-2.52 (2H, t, J=7.9 Hz, (partly under DMSO)). The compounds in the table below were made in an analogous manner, using the appropriate heteroaryl chloride in place of 2-chlorobenzimidazole in step 1: Comp.NoStructure and NameData160UPLC-MS (ES+, final purity): 2.88 min, m/z 480.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 11.89 (1H, s), 10.51 (1H, s), 7.99 (1H, dd, J = 5.7 Hz, 0.7 Hz), 7.48 (1H, s), 7.35 (1H, d, J = 8.1 Hz), 7.31-7.27 (2H, m), 7.05 (1H, d, J = 2.5 Hz), 6.99 (1H, dd, J = 8.3 Hz, 2.6 Hz), 6.35 (1H, d, J = 5.7 Hz), 4.78 (2H, s), 3.86 (2H, t, J = 5.9 Hz), 3.00- 2.88 (4H, m), 2.59-2.50 (2H, m). Example 29. Synthesis of 5-[[2-(5-phenyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 162) Step 1—5-bromo-1,3-dihydrobenzimidazol-2-one 1,1′-Carbonyldiimidazole (1.73 g, 10.69 mmol) was added to a stirred solution of 4-bromo-1,2-diaminobenzene (1 g, 5.35 mmol) in DMF (10 mL) at 25° C. under a nitrogen atmosphere and the reaction was stirred for 18 hours. The solvent was reduced in vacuo and the residue was diluted with water and EtOAc, causing a solid to crash out. The solid was filtered, slurried in water, sonicated, filtered, washed with further water and dried to give 5-bromo-1,3-dihydrobenzimidazol-2-one (912 mg, 4.28 mmol, 80% yield) as a brown solid. UPLC-MS (ES+, Short acidic): 1.28 min, m/z 212.9, 214.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.79-10.73 (2H, m), 7.08 (1H, dd, J=8.2 Hz, 2.0 Hz), 7.05 (1H, d, J=1.9 Hz), 6.87 (1H, d, J=8.2 Hz). Step 2—6-bromo-2-chloro-1H-benzimidazole 5-bromo-1,3-dihydrobenzimidazol-2-one (912 mg, 4.28 mmol) was dissolved in POCl3 (7.98 mL, 85.62 mmol) and the reaction was heated to 95° C. for 4 hours, after which time the reaction was reduced in vacuo and the residue was azeotroped with toluene. The residue was quenched with saturated NaHCO3solution causing a solid to crash out, which was sonicated, filtered, washed with water and dried to give 6-bromo-2-chloro-1H-benzimidazole (855 mg, 3.69 mmol, 86% yield) as a pale brown solid. The compound was used without further purification in the following step. UPLC-MS (ES+, Short acidic): 1.44 min, m/z 230.8, 232.8, 234.8 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 7.74 (1H, d, J=1.9 Hz), 7.49 (1H, d, J=8.6 Hz), 7.38 (1H, dd, J=8.6 Hz, 2.0 Hz). Exchangeable proton missing. Step 3—7-benzyloxy-2-(5-bromo-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinoline Eight separate microwave vials each with 7-benzyloxy-1,2,3,4-tetrahydroisoquinoline hydrochloride (50 mg, 0.18 mmol) and 6-bromo-2-chloro-1H-benzimidazole (46 mg, 0.2 mmol), and 1,4-Dioxane (2.5 mL) were sealed and heated to 180° C. in the microwave for 60 min. Each vial precipitated a solid, all the reaction mixtures were filtered and the combined solid washed with further dioxane and dried to give 7-benzyloxy-2-(5-bromo-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinoline (626 mg, 1.4 mmol, 99% yield) as a green/brown solid. UPLC-MS (ES+, Short acidic): 1.59 min, m/z 434.1; 436.0 [M+H]+ (67%) Step 4—2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-bromo-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane Sodium hydride (60% dispersed in mineral oil—168.9 mg, 4.22 mmol) was added to a stirred solution of 7-benzyloxy-2-(5-bromo-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinoline (692 mg, 1.12 mmol) and DMF (10 mL) at room temperature under a nitrogen atmosphere. After 20 minutes 2-(trimethylsilyl)ethoxymethyl chloride (0.3 mL, 1.67 mmol) was added and the reaction was allowed to stir for 2 hours. The reaction was quenched with water (1 mL) and solvent removed in vacuo. The residue was then partitioned between water (20 mL) and EtOAc (20 mL). The organic layer was separated and the aqueous extracted with DCM (20 mL). The combined organic layers were dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-40% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-bromo-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (442 mg, 0.78 mmol, 70% yield) as a pale yellow gum. UPLC-MS (ES+, Short acidic): 2.27 and 2.29 min, m/z 564.3, 566.2 [M+H]+. Presence of two SEM isomers. Step 5 (Method A)—2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-phenyl-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane Potassium carbonate (151.3 mg, 1.09 mmol), 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-bromo-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (206 mg, 0.36 mmol), phenylboronic acid (48.9 mg, 0.40 mmol), 1,4-dioxane (4 mL) and water (1 mL) were combined and stirred at room temperature under a nitrogen atmosphere, followed by addition of [1,1′-bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (29.8 mg, 0.04 mmol). The reaction was heated to 90° C. for 18 hours, after which time the reaction was cooled to room temperature and the solvent removed under reduce pressure. The residue was purified by column chromatography using an eluent of 0-35% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-phenyl-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (150 mg, 0.27 mmol, 73% yield) as a pale yellow oil. UPLC-MS (ES+, short acidic): 2.11 min, m/z 562.8 [M+H]+. Step 6—2-[5-phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-ol 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-phenyl-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (150 mg, 0.27 mmol) was dissolved in MeOH (4 mL) and palladium hydroxide (3.04 mg, 0.02 mmol) was added under an atmosphere of nitrogen. The reaction was fitted with a H2balloon, subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere overnight. The crude was filtered over celite, washed with MeOH and the solvent removed in vacuo to give 2-[5-phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-ol (117 mg, 0.25 mmol, 93% yield) as a colourless gum. UPLC-MS (ES+, short acidic):1.81 min, m/z 472.6 [M+H]+. Step 7—5-[[2-[5-phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one ol Potassium tert-butoxide (83.5 mg, 0.74 mmol) was added to a stirred solution of 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (41.2 mg, 0.25 mmol) in DMF (2 mL) in a sealable vial. The vial was sealed and and heated at 80° C. for 18 hours. The reaction was cooled to room temperature and the solvent reduced in vacuo. The crude was partitioned between EtOAc and water. The organic phase was separated, washed with saturated brine, dried over a phase separator and the solvent removed under reduce pressure. The residue was purified by column chromatography using as eluent a gradient 25-100% EtOAc in petroleum ether to give 5-[[2-[5-phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (90 mg, 0.15 mmol, 59% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.87 min, m/z 618.5 [M+H]+. Step 8—5-[[2-(5-phenyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one Trifluoroacetic acid (0.56 mL, 7.29 mmol) was added to a stirred solution of 5-[[2-[5-phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (90 mg, 0.15 mmol) in DCM (2 mL) in a sealable vial. The vial was sealed and and heated to 40° C. for 18 hours. The reaction was reduced in vacuo and loaded onto an SCX-2 column, which was flushed at first with methanol (2×), then NH3in MeOH followed by 20% DCM in MeOH/NH3to elute the product. The crude was purified by column chromatography using as eluent a gradient 1-8% MeOH in DCM to give a colourless gum, which was triturated in MeCN/Et20 to give a white solid. The solid was filtered, washed with Et20, followed by sonication in MeOH to precipitate a white solid. The whole suspension was evaporated and dried to give 5-[[2-(5-phenyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (27 mg, 0.053 mmol, 36% yield) as an off white solid. UPLC-MS (ES+, final purity): 3.04 min, m/z 488.4 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 11.57 (1H, s), 10.51 (1H, s), 7.99 (1H, dd, J=5.7 Hz, 0.7 Hz), 7.66-7.59 (2H, m), 7.52-7.38 (3H, m), 7.33-7.18 (3H, m), 7.05 (1H, d, J=2.5 Hz), 6.98 (1H, dd, J=8.3 Hz, 2.6 Hz), 6.35 (1H, d, J=5.8 Hz), 4.76 (2H, s), 3.85 (2H, t, J=5.9 Hz), 3.00-2.89 (4H, m), 2.58-2.52 (3H, m, (partly under DMSO)). As alternative, Step 5 could have been performed using Method B: 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-bromo-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (255 mg, 0.45 mmol), potassium phosphate tribasic (0.29 g, 1.35 mmol) and 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (150.5 mg, 0.68 mmol) were suspended in toluene (2 mL) and water (1 mL) and degassed under a nitrogen atmosphere in a sealable vial. Tricyclohexylphosphine (0.03 g, 0.09 mmol) and palladium (II) acetate (0.01 g, 0.03 mmol) were then added followed by further degassing, the vial was sealed and heated at 90° C. for 18 hours. The reaction was cooled and the water removed. The organics were reduced in vacuo onto silica and the product was purified by silica column chromatography using as eluent a gradient 0-80% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-(2,5-dimethylpyrazol-3-yl)benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (110 mg, 0.19 mmol, 42% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 2.02 and 2.05 min, m/z 580.5 [M+H]+(2 SEM-protected isomers). The compounds in the table below were made in an analogous manner using the appropriate boronic acid/ester and applying the appropriate method: Comp.NoStructure and NameData163UPLC-MS (ES+, final purity): 2.69 min, m/z 506.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 11.64 (1H, m), 10.50 (1H, s), 8.01-7.88 (1H, d, J = 5.8 Hz), 7.31-7.20 (3H, m), 7.10-6.89 (3H, m), 6.34 (1H, d, J = 5.8 Hz), 6.07 (1H, d, J = 5.7 Hz), 4.75 (2H, s), 3.84 (2H, t, J = 5.9 Hz), 3.75- 3.70 (3H, br s), 2.93 (4H, m), 2.56-2.52 (2H, m), 2.15 (3H, s). Example 30. Synthesis of 5-[[2-(5-propyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-nahphthyridin-2-one (Compound 161) Step 1—2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-cyclopropyl-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-bromo-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (205 mg, 0.36 mmol), cyclopropylboronic acid (0.08 g, 0.91 mmol) and potassium phosphate tribasic (0.23 g, 1.09 mmol) were suspended in toluene (2 mL) and water (1 mL) at room temperature and degassed under nitrogen atmosphere in a sealable vial. Tricyclohexylphosphine (0.02 g, 0.07 mmol) and palladium (II) acetate (0.01 g, 0.03 mmol) were then added, the vial sealed and heated 100° C. for 2 hours. The reaction was cooled and the water removed. The organics were reduced in vacuo onto silica and the product was purified by column chromatography using as eluent a gradient 0-35% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-cyclopropyl-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (155 mg, 0.29 mmol, 81% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.98 min, m/z 526.9 [M+H]+. Step 2—2-[5-propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-ol 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-cyclopropyl-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (155 mg, 0.29 mmol) was dissolved in methanol (4 mL) and palladium hydroxide (3.36 mg, 0.02 mmol) was added under an atmosphere of nitrogen. The reaction was fitted with a H2balloon, subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere overnight. The crude was filtered over celite, washed with MeOH and the solvent removed in vacuo to give 2-[5-propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-ol (107 mg, 0.24 mmol, 83% yield) as a colourless gum. UPLC-MS (ES+, short acidic): 1.78 min, m/z 438.6 [M+H]+. Step 3—5-[[2-[5-propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one Potassium tert-butoxide (83.5 mg, 0.74 mmol) was added to a stirred solution of 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (40.6 mg, 0.24 mmol) and 2-[5-propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-ol (107 mg, 0.24 mmol) in DMF (2 mL) in a sealable vial, which was sealed and heated at 100° C. for 72 hours. Reaction was cooled to room temperature and the solvent reduced in vacuo. The crude was partitioned between EtOAc and water. The organic layer was separated, washed with saturated brine, dried over a phase separator and the solvent removed under reduce pressure. The residue was purified by column chromatography using as eluent a gradient 25-100% EtOAc in petroleum ether to give 5-[[2-[5-propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (41 mg, 0.07 mmol, 29% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.81 min, m/z 584.5 [M+H]+. Step 3—5-[[2-(5-propyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one Trifluoroacetic acid (0.27 mL, 3.51 mmol) was added to a stirred solution of 5-[[2-[5-propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (41. mg, 0.07 mmol) in DCM (2 mL) in a sealable vial. The reaction was sealed and heated to 40° C. for 18 hours. The reaction was reduced in vacuo and loaded onto an SCX cartridge, which was flushed at first with MeOH and then MeOH/NH3followed by 20% DCM:MeOH/NH3to elute the product. The product was then purified by silica column chromatography using as eluent a gradient 1-8% MeOH/DCM to give a colourless gum. The compound was re-purified using reverse column chromatography using as eluent a gradient 0-30% (acetonitrile+0.1% formic acid) in (water+0.1% formic acid). Fractions containing the product were re-purified using preparative LCMS (early method). Fractions containing compound were loaded onto an SCX column which was flushed at first with MeOH and then MeOH/NH3to elute the product giving 5-[[2-(5-propyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (8 mg, 0.018 mmol, 25% yield) as an off white solid. UPLC-MS (ES+, final purity): 3.03 min, m/z 454.3 [M+H].1H NMR (400 MHz, DMSO-d6) δ/ppm: 11.80 (1H, s), 10.50 (1H, s), 7.98 (1H, d, J=5.8 Hz), 7.27 (1H, d, J=8.3 Hz), 7.13 (1H, d, J=8.0 Hz), 7.07-6.94 (3H, m), 6.81 (1H, dd, J=8.0 Hz, 1.6 Hz), 6.34 (1H, d, J=5.8 Hz), 4.72 (2H, s), 3.81 (2H, t, J=5.9 Hz), 2.98-2.87 (4H, m), 2.62-2.51 (4H, m), 1.65-1.51 (2H, m), 0.88 (3H, t, J=7.3 Hz). Example 31. Synthesis of N-[4-[3-[4-(4-fluorophenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide (Compound 155) Step 1—6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]-N-[2-(4-fluorophenyl)-2-oxo-ethyl]chromane-3-carboxamide To a solution of 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-carboxylic acid (65 mg, 0.18 mmol) and 2-amino-1-(4-fluorophenyl)ethanone hydrochloride (54.7 mg, 0.20 mmol) in THF (1.8 mL) were added T3P (164 μL, 0.28 mmol) and DIPEA (99 μL, 0.57 mmol) and the mixture was heated at 65° C. for 1 hour. After cooling to rt, the solvent was removed under reduce pressure and the residue dissolved in EtOAc (25 mL). The organic layer was washed with water (2×10 mL) and brine (10 mL), dried over Na2SO4, filtered and the solvent removed in vacuo to afford 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]-N-[2-(4-fluorophenyl)-2-oxo-ethyl]chromane-3-carboxamide (86 mg, 0.17 mmol, 95% yield) as a yellow solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.46 min, m/z 490.6 [M+H]+. Step 2—N-[4-[3-[4-(4-fluorophenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide To a suspension of 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]-N-[2-(4-fluorophenyl)-2-oxo-ethyl]chromane-3-carboxamide (85.5 mg, 0.17 mmol) in n-BuOH (1 mL) were added NH4OAc (134.6 mg, 1.75 mmol) and Et3N (26 μL, 0.18 mmol) and the mixture was irradiated at 150° C. for 1 hour. After cooling to rt, the solvent was removed under reduce pressure and the residue dissolved in EtOAc (25 mL). The organic layer was washed with sat. aq. NaHCO3(10 mL), water (2×10 mL) and brine (10 mL), dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by flash chromatography using as eluent a gradient 0-10% MeOH in DCM, followed by purification by preparative LC-MS. Fractions containing the product were dried and loaded into an SCX-2 column, which was flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to elute the product. The filtrate was concentrated to give N-[4-[3-[4-(4-fluorophenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide (10 mg, 0.022 mmol, 13% yield) as a white solid. UPLC-MS (ES+, final purity): 2.73 min, m/z 471.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.33 & 12.10 (0.2 & 0.8H, 2 s, mixture of tautomers), 10.80 (1H, s), 8.16 (1H, d, J=5.7 Hz), 7.80-7.42 (2H, m), 7.65 (1H, d, J=2.5 Hz), 7.57 (1H, br s), 7.25-7.10 (2H, m), 7.02 (1H, d, J=2.5 Hz), 6.94-6.86 (2H, m*), 6.62 (1H, dd, J=5.7 Hz, 2.4 Hz), 4.54-4.48 (1H, m), 4.16-4.08 (1H, m), 3.44-3.34 (1H, m), 3.28-3.18 (1H, m), 3.15-3.07 (1H, m), 1.97 (1H, quint, J=6.2 Hz), 0.77 (4H, d, J=6.2 Hz). Example 32. Synthesis of 4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-N-(2-pyridyl)pyridin-2-amine (Compound 140) Step 1—6-[(2-chloro-4-pyridyl)oxy]-N-phenacyl-chromane-3-carboxamide To a solution of 6-[(2-chloro-4-pyridyl)oxy]chromane-3-carboxylic acid (437 mg, 1.43 mmol) and 2-aminoacetophenone hydrochloride (270 mg, 1.57 mmol) in THF (14 mL) were added T3P (1.28 mL, 2.14 mmol) and DIPEA (0.8 mL, 4.58 mmol) and the mixture was heated at 65° C. for 1.5 hours. After cooling to rt, the solvent was removed under reduce pressure and the residue dissolved in EtOAc (60 mL). The organic layer was washed with water (2×20 mL) and brine (20 mL), dried over Na2SO4, filtered and the solvent removed in vacuo to give 6-[(2-chloro-4-pyridyl)oxy]-N-phenacyl-chromane-3-carboxamide (604.5 mg, 1.43 mmol, 100% yield) as a light beige solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.72 min, m/z 423.3 [M+H]+. Step 2—2-chloro-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine To a suspension of 6-[(2-chloro-4-pyridyl)oxy]-N-phenacyl-chromane-3-carboxamide (513 mg, 1.21 mmol) in n-BuOH (10 mL) were added NH4OAc (935.2 mg, 12.13 mmol) and Et3N (169 μL, 1.21 mmol). The vial was sealed and the mixture irradiated at 150° C. for 1 hr. After cooling to rt, the solvent was removed under reduce pressure and the residue dissolved in EtOAc (60 mL). The organic layer was washed with sat. aq. NaHCO3(20 mL), water (2×20 mL) and brine (20 mL), dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give 2-chloro-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine (314.9 mg, 0.78 mmol, 64% yield) as a beige solid. UPLC-MS (ES+, short acidic): 1.43 min, m/z 404.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 8.27 (1H, dd, J=5.6, 0.3 Hz), 7.77-7.73 (2H, m), 7.69 (1H, br s), 7.43-7.36 (2H, m), 7.28-7.22 (1H, m), 7.10 (1H, d, J=2.8 Hz), 7.00 (1H, dd, J=8.8, 2.8 Hz), 6.96-6.92 (3H, m), 4.57-4.51 (1H, m), 4.25-4.18 (1H, m), 3.53 (1H, br s), 3.32-3.25 (2H, m), 3.18 (1H, dd, J=16.6, 5.3 Hz), 0.89-0.79 (1H, m). Step 3—4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-N-(2-pyridyl)pyridin-2-amine A solution of 2-chloro-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine (50 mg, 0.12 mmol), 2-aminopyridine (17.5 mg, 0.19 mmol), XPhos Pd G2 (9.8 mg, 0.01 mmol), XPhos (5.9 mg, 0.01 mmol) and K2CO3(51.3 mg, 0.3700 mmol) in tert-butanol under a nitrogen atmosphere (0.6 mL) was irradiated at 130° C. for 1 hour. After this time, additional 2-aminopyridine (17.5 mg, 0.1900 mmol), K2CO3(51.3 mg, 0.37 mmol), XPhos Pd G2 (9.8 mg, 0.01 mmol) and XPhos (5.9 mg, 0.01 mmol) were added and the mixture was irradiated at 160° C. for 1 hour, after which time it was through a plug of celite and the filter cake washed with EtOAc. The filtrate was concentrated and the residue purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM, followed by purification by preparative LC-MS. Fractions containing the product were dried and loaded into an SCX-2 column, which was flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to elute the product. The filtrate was concentrated to give 4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-N-(2-pyridyl)pyridin-2-amine (11.5 mg, 0.025 mmol, 20% yield) as a white solid. UPLC-MS (ES+, final purity): 2.47 min, m/z 462.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.37 (1H, br s), 9.66 (1H, br s), 8.17-8.14 (1H, m), 8.08 (1H, d, J=5.8 Hz), 7.76-7.70 (2H, m), 7.69-7.60 (2H, m), 7.55 (1H, br s), 7.40-7.31 (3H, m), 7.21-7.15 (1H, m), 7.04 (1H, d, J=2.7 Hz), 6.94 (1H, dd, J=8.8 Hz, 2.7 Hz), 6.90 (1H, d, J=8.8 Hz), 6.87-6.82 (1H, m), 6.41 (1H, dd, J=5.8 Hz, 2.1 Hz), 4.56-4.49 (1H, m), 4.17-4.10 (1H, m), 3.45-3.36 (1H, m), 3.26 (1H, dd, J=16.7 Hz, 10.6 Hz), 3.18-3.09 (1H, m). The compound in the table below was made in an analogous manner, using the appropriate amine in step 3: Comp.NoStructure and NameData141UPLC-MS (ES+, final purity): 2.81 min, m/z 468.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.15 (1H, br s), 11.02 (1H, s), 8.16 (1H, d, J = 5.7 Hz), 7.73 (2H, d, J = 7.5 Hz), 7.57 (1H, br s), 7.39- 7.31 (3H, s), 7.22-7.15 (1H, m), 7.06 (1H, d, J = 2.6 Hz), 6.99-6.94 (2H, m), 6.92 (1H, d, J = 8.8 Hz), 6.53 (1H, d, J = 2.2 Hz), 6.52 (1H, dd, J = 5.7 Hz, 2.2 Hz), 4.56-4.50 (1H, m), 4.17-4.10 (1H, m),3.46-3.36 (1H, m), 3.31-3.21(1H, m), 3.19-3.10 (1H, m). Example 33. Synthesis of 2,2-difluoro-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide (Compound 137) Step 1—2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-4-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane and ˜2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-5-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane To a solution of 2-chloro-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine (553 mg, 1.37 mmol) in anhydrous THF (13.7 mL) at 0° C. under a nitrogen atmosphere was added NaH (60% in mineral oil—82.1 mg, 2.05 mmol) and the mixture was stirred at 0° C. for 45 minutes. 2-(trimethylsilyl)ethoxymethyl chloride (300 μL, 1.7 mmol) was added dropwise and the mixture was stirred for 2 hours at 0° C. The reaction mixture was quenched with water (15 mL) and extracted with EtOAc (3×10 mL). The combined organic layers were dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-50% EtOAc in petroleum ether to give 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-4-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane (444 mg, 0.83 mmol, 61% yield) and 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-5-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane (120.4 mg, 0.23 mmol, 16% yield), both as yellow gums. 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-4-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane: UPLC-MS (ES+, short acidic): 2.23 min, m/z 535.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 8.29-8.26 (1H, m), 7.77 (1H, s), 7.76-7.72 (2H, m), 7.37-7.32 (2H, m), 7.22-7.16 (1H, m), 7.08 (1H, d, J=2.8 Hz), 7.00 (1H, dd, J=8.8 Hz, 2.8 Hz), 6.97-6.92 (3H, m), 5.48 (1H, d, J=11.1 Hz), 5.42 (1H, d, J=11.1 Hz), 4.50-4.44 (1H, m), 4.11-4.04 (1H, m), 3.61-3.52 (3H, m), 3.32-3.25 (1H, m), 3.08-2.99 (1H, m), 0.93-0.86 (2H, m), −0.02 (9H, s). 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-5-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane: UPLC-MS (ES+, short acidic): 2.00 min, m/z 535.5 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 8.29-8.26 (1H, m), 7.51-7.38 (5H, m), 7.07 (1H, d, J=2.8 Hz), 7.02 (1H, s), 7.00 (1H, dd, J=8.8 Hz, 2.8 Hz), 6.97-6.92 (3H, m), 5.39 (1H, d, J=11.1 Hz), 5.33 (1H, d, J=11.1 Hz), 4.51-4.45 (1H, m), 4.12-4.05 (1H, m), 3.65-3.55 (1H, m), 3.44-3.38 (2H, m), 3.30-3.20 (1H, m), 3.09-3.01 (1H, m), 0.88-0.77 (2H, m), −0.08 (9H, s). Step 2—1,1-diphenyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]methanimine To a solution of 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-4-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane (444 mg, 0.83 mmol), benzophenone imine (210 μL, 1.25 mmol) and Cs2CO3(677.1 mg, 2.08 mmol) in dry 1,4-dioxane (4 mL) under a nitrogen atmosphere were added (+/−)-BINAP (103.5 mg, 0.17 mmol) and tris(dibenzylideneacetone)dipalladium (0) (76.1 mg, 0.08 mmol) and the mixture was heated at 110° C. for 18 hours. After cooling to rt, the mixture was filtered through celite and the filter cake washed with EtOAc. The solvent was removed in vacuo and the residue purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give 1,1-diphenyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]methanimine (564.3 mg, 0.83 mmol, 100% yield) as an orange solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 2.12 min, m/z 679.5 [M+H]+. Step 3—acetic acid; 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyridin-2-amine To a solution of 1,1-diphenyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]methanimine (564.3 mg, 0.83 mmol) in MeOH (8.3 mL) were added NaOH (204.6 mg, 2.49 mmol) and hydroxylamine hydrochloride (127.1 mg, 1.83 mmol) and the mixture was stirred at rt for 1 hour. The solvent was removed in vacuo and the residue purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM to give acetic acid; 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyridin-2-amine (366.4 mg, 0.64 mmol, 77% yield) as an orange solid. UPLC-MS (ES+, short acidic): 1.70 min, m/z 515.4 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 7.79-7.72 (4H, m), 7.38-7.32 (2H, m), 7.22-7.17 (1H, m), 7.00-6.97 (1H, m), 6.92-6.89 (2H, m), 6.13 (1H, dd, J=5.9 Hz, 2.3 Hz), 5.96 (2H, br s), 5.82 (1H, d, J=2.3 Hz), 5.48 (1H, d, J=11.1 Hz), 5.42 (1H, d, J=11.1 Hz), 4.49-4.43 (1H, m), 4.07-4.00 (1H, m), 3.60-3.48 (3H, m), 3.35-3.28 (1H, m, (partly under water peak)), 3.07-2.99 (1H, m), 1.91 (3H, s, AcOH), 0.94-0.83 (2H, m), −0.02 (9H, s). Step 4—2,2-difluoro-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide To a solution of 2,2-difluorocyclopropanecarboxylic acid (63.7 mg, 0.52 mmol) in anhydrous THF (0.7 mL) at 0° C. under a nitrogen atmosphere were added DMF (2 μL, 0.0300 mmol) and oxalyl chloride (44.6 μL, 0.53 mmol) and the reaction was stirred at 0° C. for 30 minutes. The mixture was then added to a solution of acetic acid; 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyridin-2-amine (60 mg, 0.10 mmol) in anhydrous Pyridine (0.70 mL) and stirred at rt for 1 hr. The mixture was diluted with EtOAc (30 mL) and washed with sat. aq. NaHCO3(10 mL), water (10 mL) and brine (10 mL). The organic layer was dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give 2,2-difluoro-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide (47.3 mg, 0.076 mmol, 73% yield) as a glassy solid. UPLC-MS (ES+, short acidic): 2.06 min, m/z 619.5 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 11.03 (1H, s), 8.20 (1H, d, J=5.7 Hz), 7.77 (1H, s), 7.76-7.72 (2H, m), 7.62 (1H, br s), 7.37-7.32 (2H, m), 7.22-7.16 (1H, m), 7.05 (1H, d, J=2.6 Hz), 6.96 (1H, dd, J=8.8 Hz, 2.6 Hz), 6.92 (1H, d, J=8.8 Hz), 6.67 (1H, ddd, J=5.7 Hz, 4.1 Hz, 2.3 Hz), 5.48 (1H, d, J=11.1 Hz), 5.42 (1H, d, J=11.1 Hz), 5.50-5.44 (1H, m), 4.05 (1H, td, J=10.7 Hz, 1.7 Hz), 3.60-3.54 (3H, m), 3.32-3.25 (1H, m), 3.07-2.90 (2H, m), 2.04-1.93 (2H, m), 0.93-0.85 (2H, m), −0.03 (9H, s). Step 5—2,2-difluoro-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide To a solution of 2,2-difluoro-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide (47.3 mg, 0.08 mmol) in DCM (1 mL) was added trifluoroacetic acid (0.5 mL, 6.53 mmol) and the mixture was stirred at r for 72h. The solvent was removed in vacuo and the residue loaded into loaded into an SCX-2 column, which was flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to elute the product. The residue was purified by column chromatography using as eluent a gradient 0-6% MeOH in DCM to give 2,2-difluoro-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide (28.8 mg, 0.059 mmol, 77% yield) as a white solid. UPLC-MS (ES+, final purity): 2.94 min m/z 489.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.32 (0.2H, br s), 12.10 (0.8H, br s), 11.03 (1H, s), 8.19 (1H, d, J=5.7 Hz), 7.79-7.56 (3.78H, m), 7.44-7.12 (3.22, N), 7.06-7.03 (1H, m), 6.94 (1H, dd, J=8.8 Hz, 2.8 Hz), 6.90 (1H, d, J=8.8 Hz), 6.67 (1H, ddd, J=5.7 Hz, 2.3 Hz, 0 Hz), 4.55-4.49 (1H, m), 4.17-4.09 (1H, d), 3.45-3.36 (1H, m), 3.29-3.19 (1H, m), 3.18-3.07 (1H, m), 3.01-2.90 (1H, m), 2.05-1.91 (2H, m). The compounds in the table below were made in an analogous manner, using the appropriate acid in step 4: Comp.NoStructure and NameData138UPLC-MS (ES+, final purity): 3.00 min, m/z 471.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.32 (0.2H, br s), 12.06 (0.8H, br s), 10.21 (1H, d, J = 1.3 Hz), 8.23 (1H, d, J = 5.7 Hz), 7.77-7.73 (1.6H, m), 7.65-7.61 (0.4H, m), 7.60- 7.56, (1.8H, m), 7.43-7.37 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.19- 7.13 (0.8H, m), 7.07-7.04 (1H, m), 6.95 (1H, dd, J = 8.8, 2.8 Hz), 6.90 (1H, d, J = 8.8 Hz),6.73 (1H, dd, J = 8.8, 2.4 Hz),4.55-4.49 (1H, m), 4.16-4.08(1H, m), 3.45-3.35 (1H, m),3.29-3.19 (1H, m), 3.16-3.07(1H, m), 1.46-1.27 (4H, m).139UPLC-MS (ES+, final purity): 2.99 min, m/z 503.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.32 (0.2H, br s), 12.10 (0.8H, br s), 10.70 (1H, s), 8.17 (1H, d, J = 5.7 Hz), 7.78-7.72 (1.6H, m), 7.72- 7.57 (2.2H, m), 7.43-7.37 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.19-7.13 (0.8H, m), 7.06- 7.02 (1H, m), 6.94 (1H, dd, J = 8.8, 2.6 Hz), 6.90 (1H, d, J = 8.8 Hz), 6.65 (1H, dd, J =5.7, 2.4 Hz), 4.56-4.50 (1H,m), 4.17-4.10 (1H, m), 3.46-3.36 (1H, m), 3.27-3.07 (3H,m), 2.82-2.69 (4H, m).157UPLC-MS (ES+, final purity): 2.79 & 2.81 min, m/z 467.3 [M + H]+(0.14/1 mixture of diastereoisomers). 1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.31 (0.2H, br s), 12.09 (0.8H, br s), 10.71 (1H, s), 8.15 (1H, d, J = 5.7 Hz), 7.77-7.73 (1.6H, m), 7.71- 7.57 (2.2H, m), 7.43-7.37 (0.4H, m), 7.36-7.29, (1.6H, m), 7.29-7.20 (0.4H, m), 7.19-7.13 (0.8H, m), 7.05- 7.00 (1H, m), 6.96-6.86 (2H,m), 6.61 & 6.57 (1H, dd, J =5.7, 2.3 Hz), 4.55-4.49 (1H,m), 4.16-4.08 (1H, m), 3.45-3.35 (1H, m), 3.28-3.18 (1H,m), 3.16-3.06 (1H, m), 1-76-1.70 (1H, m), 2.03-1.95 &1.27-1.14 (2H, m), 1.09-1.04(3H, m), 0.66-0.59 (1H, m). Example 34. Synthesis of N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]acetamide (Compound 156) Step 1—N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]acetamide To a solution of acetic acid; 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyridin-2-amine (69.5 mg, 0.12 mmol) in pyridine (0.6 mL) was slowly added a solution of acetic anhydride (23 μL, 0.24 mmol) in THF (0.6 mL) and the mixture was stirred at rt for 72 hrs. The mixture was diluted with EtOAc (30 mL) and washed with sat. aq. NaHCO3(10 mL), water (10 mL) and brine (10 mL). The organic layer was dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]acetamide (47.5 mg, 0.085 mmol, 71% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.90 min, m/z 557.5 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.50 (1H, s), 8.15 (1H, d, J=5.7 Hz), 7.77 (1H, s), 7.76-7.72 (2H, m), 7.66 (1H, d, J=2.1 Hz), 7.38-7.32 (2H, m), 7.22-7.17 (1H, m), 7.02 (1H, d, J=2.4 Hz), 6.97-6.90 (2H, m), 6.60 (1H, dd, J=5.7 Hz, 2.4 Hz), 5.49 (1H, d, J=11.1 Hz), 5.42 (1H, d, J=11.1 Hz), 4.50-4.43 (1H, m), 4.09-4.01 (1H, m), 3.61-3.50 (3H, m), 3.31-3.25 (1H, m), 3.07-2.98 (1H, m), 2.05 (3H, s), 0.93-0.86 (2H, m), −0.02 (9H, s). Step 2—N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]acetamide To a solution of N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]acetamide (39.4 mg, 0.07 mmol) in DCM (1 mL) was added TFA (0.5 mL, 6.53 mmol) and the mixture was stirred at rt for 22 hrs. The solvent was removed under reduce pressure and the residue was loaded into an SCX-2 column and flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to elute the product. The solvent was removed in vacuo and the residue was purified by column chromatography using as eluent a gradient 0-8% MeOH in DCM to give N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]acetamide (20.2 mg, 0.047 mmol, 70% yield) as a white solid. UPLC-MS (ES+, final purity): 2.45 min, m/z 427.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.32 (0.2, br s), 12.10 (0.8H, br s), 10.50 (1H, s), 8.15 (1H, d, J=5.7 Hz), 7.78-7.73 (1.6H, m), 7.68-7.57 (2.2H, m), 7.43-7.37 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.18-7.13 (0.8H, m), 7.05-7.01 (1H, m), 6.93 (1H, dd, J=8.8 Hz, 2.7 Hz), 6.89 (1H, d, J=8.8 Hz), 6.60 (1H, dd, J=5.7 Hz, 2.4 Hz), 4.55-4.49 (1H, m), 4.16-4.08 (1H, m), 3.45-3.35 (1H, m), 3.29-3.19 (1H, m), 3.16-3.07 (1H, m), 2.05 (3H, s). Example 35. Synthesis of N-cyclopropyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine-2-carboxamide (Compound 158) Step 1—4-chloro-N-cyclopropyl-pyridine-2-carboxamide To a suspension of 4-chloro-2-pyridinecarboxylic acid (500 mg, 3.17 mmol) and cyclopropylamine (242 μL, 3.49 mmol) in THF (32 mL) were added T3P (2.8 mL, 4.7 mmol) and DIPEA (1.1 mL, 6.32 mmol) and the mixture was heated at 65° C. for 2.5 hours. The solvent was removed under reduce pressure and the residue dissolved in EtOAc (100 mL). The organic layer was washed with sat. aq. NaHCO3(25 mL), water (2×25 mL) and brine (25 mL), dried over Na2SO4, filtered and the solvent removed in vacuo to give 4-chloro-N-cyclopropyl-pyridine-2-carboxamide (499.8 mg, 2.54 mmol, 80% yield) as an off-white solid. The compound used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.35 min, m/z 197.0 [M+H].1H NMR (400 MHz, DMSO-d6) δ/ppm: 8.82 (1H, d, J=4.4 Hz), 8.59 (1H, dd, J=5.3, 0.6 Hz), 8.01 (1H, dd, J=2.2 Hz, 0.6 Hz), 7.75 (1H, dd, J=5.3 Hz, 2.2 Hz), 2.95-2.87 (1H, m), 0.72-0.65 (4H, m). Step 2—6-[[2-(cyclopropylcarbamoyl)-4-pyridyl]oxy]chromane-3-carboxylic Acid To a solution of 4-chloro-N-cyclopropyl-pyridine-2-carboxamide (200 mg, 1.02 mmol) in DMF (10 mL) under a nitrogen atmosphere were added Cs2CO3(994.3 mg, 3.05 mmol) and 6-hydroxychromane-3-carboxylic acid (197.8 mg, 1.02 mmol) and the mixture was heated at 100° C. overnight. Additional 6-hydroxychromane-3-carboxylic acid (49.4 mg, 0.25 mmol) and Cs2CO3(165.7 mg, 0.51 mmol) were added and the mixture was heated at 100° C. for 72 hours. After cooling to rt, the mixture was poured into cold water and the aqueous was acidified to pH 2 with 1N aq. HCl followed by with EtOAc (4×50 mL). The combined organic layers were dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM to give 6-[[2-(cyclopropylcarbamoyl)-4-pyridyl]oxy]chromane-3-carboxylic acid (192.9 mg, 0.54 mmol, 54% yield) as a beige solid. UPLC-MS (ES+, short acidic): 1.47 min, m/z 355.4 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.69 (1H, s), 8.71 (1H, d, J=5.0 Hz), 8.47 (1H, dd, J=5.6 Hz, 0.4 Hz), 7.34 (1H, dd, J=2.7 Hz, 0.4 Hz), 7.12 (1H, dd, J=5.6 Hz, 2.7 Hz), 7.04 (1H, d, J=2.8 Hz), 6.94 (1H, dd, J=8.8 Hz, 2.8 Hz), 6.88 (1H, d, J=8.8 Hz), 4.36 (1H, dd, J=10.8 Hz, 3.1 Hz), 4.17 (1H, dd, J 10.8 Hz, 7.7 Hz), 3.06-2.95 (3H, m), 2.89-2.82 (1H, m), 0.69-0.63 (4H, m). Step 3—N-cyclopropyl-4-[3-(phenacylcarbamoyl)chroman-6-yl]oxy-pyridine-2-carboxamide To a solution of 6-[[2-(cyclopropylcarbamoyl)-4-pyridyl]oxy]chromane-3-carboxylic acid (190.6 mg, 0.54 mmol) and 2-aminoacetophenone hydrochloride (101.6 mg, 0.59 mmol) in THF (5.3 mL) were added T3P (480 μL, 0.81 mmol) and DIPEA (290 μL, 1.66 mmol) and the mixture was heated at 65° C. for 2 hours. After cooling to rt, the solvent was removed in vacuo and the residue dissolved in EtOAc (50 mL). The organic layer was washed with water (2×15 mL) and brine (15 mL), dried over Na2SO4, filtered and the solvent removed under reduce pressure. The residue was suspended in petroleum ether, filtered and dried to give N-cyclopropyl-4-[3-(phenacylcarbamoyl)chroman-6-yl]oxy-pyridine-2-carboxamide (228 mg, 0.48 mmol, 90% yield) as a cream solid. UPLC-MS (ES+, short acidic): 1.64 min, m/z 472.5 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 8.72 (1H, d, J=5.0 Hz), 8.58-8.53 (1H, m), 8.47 (1H, dd, J=5.6 Hz, 0.3 Hz), 8.02-7.97 (2H, m), 7.70-7.65 (1H, m), 7.58-7.52 (2H, m), 7.35 (1H, dd, J=2.6 Hz, 0.3 Hz), 7.13 (1H, dd, J=5.6 Hz, 2.6 Hz), 7.06 (1H, d, J=2.8 Hz), 6.96 (1H, dd, J=8.8 Hz, 2.8 Hz), 6.90 (1H, d, J=8.8 Hz), 4.67 (2H, d, J=5.6 Hz), 4.43-4.37 (1H, m), 4.03-3.95 (1H, m), 3.07-2.82 (4H, m), 0.69-0.62 (4H, m). Step 4—N-cyclopropyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine-2-carboxamide To a suspension of N-cyclopropyl-4-[3-(phenacylcarbamoyl)chroman-6-yl]oxy-pyridine-2-carboxamide (227.6 mg, 0.48 mmol) in 1-butanol (4.8 mL) were added ammonium acetate (372.1 mg, 4.82 mmol) and triethylamine (67 μL, 0.48 mmol) and the mixture was irradiated at 150° C. for 45 minutes. The solvent was removed under reduce pressure and the residue purified by column chromatography using as eluent a gradient 0-4% MeOH in DCM, followed by purification by reverse column chromatography using as eluent a gradient 0-50% (MeCN+0.1% formic acid) in (water+0.1% formic acid). Fractions containing product were combined, the solvent removed in vacuo and the residue loaded into a SCX-2 which was flushed at first with MeOH and then MeOH+NH3to elute the product. The solvent was removed in vacuo to give N-cyclopropyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine-2-carboxamide (75.7 mg, 0.17 mmol, 67% yield) as a white solid. UPLC-MS (ES+, final purity): 3.12 min, m/z 453.4 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.24 (1H, br s), 8.72 (1H, d, J=5.0 Hz), 8.47 (1H, dd, J=5.6 Hz, 0.3 Hz), 7.76-7.70 (2H, m), 7.55 (1H, br s), 7.39-7.32 (3H, m), 7.22-7.16 (1H, m), 7.15 (1H, dd, J=5.6 Hz, 2.6 Hz), 7.09 (1H, d, J=2.8 Hz), 6.99 (1H, dd, J=8.8 Hz, 2.8 Hz), 6.94 (1H, d, J=8.8 Hz), 4.57-4.51 (1H, m), 4.20-4.13 (1H, m), 3.49-3.39 (1H, m), 3.30-3.22 (1H, m), 3.19-3.10 (1H, m), 2.91-2.82 (1H, m), 0.71-0.62 (4H, m). Example 36. Synthesis of N-cyclobutyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridin-2-amine (Compound 142) Step 1—N-cyclobutyl-4-[3-[5-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-pyridin-2-amine A solution of 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-5-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane (125 mg, 0.23 mmol; Example 33, Step 1), tris(dibenzylideneacetone)dipalladium (0) (21.5 mg, 0.02 mmol), (+/−)-BINAP (29.2 mg, 0.05 mmol) and Cs2CO3(152.5 mg, 0.47 mmol) in 1,4-Dioxane (2.2 mL) was degassed under a nitrogen atmosphere for 10 minutes then cyclobutanamine (40 μL, 0.47 mmol) was added. The vial was sealed and the mixture heated at 100° C. for 20 hours. After cooling to rt, the mixture was filtered through diatomaceous earth and the filter cake was washed with EtOAc. The filtrate was concentrated and the residue purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM to give N-cyclobutyl-4-[3-[5-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-pyridin-2-amine (133.1 mg, 0.23 mmol, 100% yield) as an orange foam. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.83 min, m/z 569.3 [M+H]+. Step 2—N-cyclobutyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridin-2-amine To a solution of N-cyclobutyl-4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-pyridin-2-amine (133.1 mg, 0.23 mmol) in DCM (3.6 mL) was added TFA (1.8 mL, 23.51 mmol) and the mixture was stirred at 25° C. for 18 hours. The solvent was removed under reduce pressure, the residue loaded into an SCX-2 column and flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to elute the product. The residue was purified by column chromatography using as eluent a gradient 0-6% MeOH in DCM, followed by reverse column chromatography using as eluent a gradient 0-50% (MeCN+0.1% formic acid) in (water+0.1% formic acid). Fractions containing the product were loaded onto a SCX-2 column and flushed at first with MeOH (10 mL) and then NH3in MeOH (10 mL) to give N-cyclobutyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridin-2-amine (30 mg, 0.068 mmol, 29% yield) as a white solid. UPLC-MS (ES+, final purity): 2.45 min, m/z 439.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.12 (1H, br s), 7.82 (1H, d, J=5.8 Hz), 7.73 (2H, m), 7.55 (1H, br s), 7.38-7.30 (2H, m), 7.21-7.14 (1H, m), 6.99-6.96 (1H, m), 6.91-6.85 (2H, m), 6.77 (1H, d, J=6.6 Hz, broad), 6.10 (1H, dd, J=5.8, 2.2 Hz), 5.76 (1H, d, J=2.2 Hz), 4.54-4.48 (1H, m), 4.27-4.15 (1H, m), 4.15-4.06 (1H, m), 3.43-3.34 (1H, m), 3.28-3.19 (1H, m), 3.15-3.07 (1H, m), 2.25-2.16 (2H, m), 1.85-1.73 (2H, m), 1.69-1.53 (2H, m). Example 37. Synthesis of 1-methyl-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]piperidine-4-carboxamide (Compound 147) Step 1—1-methyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]piperidine-4-carboxamide A solution of 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyridin-2-amine (60 mg, 0.12 mmol), 1-methylpiperidine-4-carboxylic acid (18.4 mg, 0.13 mmol), propylphosphonic anhydride (104 μL, 0.17 mmol) and N,N-diisopropylethylamine (41 μL, 0.23 mmol) in THF (1.2 mL) was heated at 65° C., in a sealed tube, overnight. Additional 1-methylpiperidine-4-carboxylic acid (18.4 mg, 0.13 mmol), propylphosphonic anhydride (104 μL, 0.17 mmol) and N,N-diisopropylethylamine (41 μL, 0.23 mmol) were added in a sealable vial. The vial was sealed and the mixture heated at 65° C. for 2 hours. After cooling to room temperature, the mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-20% MeOH in DCM, followed by 20% NH32N in MeOH in DCM to give 1-methyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]piperidine-4-carboxamide (38.4 mg, 0.060 mmol, 51% yield) as an off-white solid. UPLC-MS (ES+, Short acidic): 1.71 min, m/z 640.3 [M+H]+. Step 2—1-methyl-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]piperidine-4-carboxamide To a solution of 1-methyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]piperidine-4-carboxamide (38.4 mg, 0.060 mmol) in DCM (1 mL) was added trifluoroacetic acid (0.5 mL, 6.53 mmol) in a sealable vial. The vial was sealed and the mixture stirred at 25° C. for 18 hours. The mixture was concentrated and the residue purified by reverse column chromatography using as eluent a gradient 0-50% of acetonitrile+0.1% formic acid in water+0.1% formic acid. Fraction containing the product were combined, the solvent removed in vacuo and loaded into an SCX-2 column. The column was washed at first with MeOH and then MeOH+NH3to give 1-methyl-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]piperidine-4-carboxamide (16 mg, 0.031 mmol, 52% yield) as a white solid. UPLC-MS (ES+, final purity): 2.26 min, m/z 510.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.31 (0.2H, br s), 12.09 (0.8H, br s), 10.45 (1H, s), 8.15 (1H, d, J=5.7 Hz), 7.77-7.73 (1.6H, m), 7.67 (1H, d, J=2.4 Hz), 7.66-7.61 (0.4H, m), 7.61-7.57 (0.8H, m), 7.43-7.37 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.19-7.13 (0.8H, m), 7.04-7.01 (1H, m), 6.93 (1H, dd, J=8.8, 2.6 Hz), 6.90 (1H, d, J=8.8 Hz), 6.62 (1H, dd, J=5.7, 2.4 Hz), 4.56-4.49 (1H, m), 4.17-4.09 (1H, m), 3.46-3.36 (1H, m), 3.29-3.20 (1H, m), 3.17-3.07 (1H, m), 2.85-2.75 (2H, m), 2.43-2.36 (1H, m), 2.16 (3H, s), 1.92-1.69 (2H, m), 1.74-1.65 (2H, m), 1.64-1.51 (2H, m). The compounds in the table below were made in an analogous manner, using the appropriate acid in step 1: Comp.NoStructure and NameData144UPLC-MS (ES+, final purity): 2.30 min, m/z 493.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.31 (1H, br s), 10.89 (1H, s), 8.20 (1H, d, J = 5.8 Hz), 7.76-7.69 (3H, m), 7.56 (1H, br s), 7.51 (1H, s), 7.37-7.31 (2H, m), 7.21-7.14 (2H, m), 7.02 (1H, d, J = 2.5 Hz), 6.95 (1H, br s), 6.91 (1H, dd, J = 8.8 Hz, 2.5 Hz), 6.88 (1H, d, J = 8.8 Hz), 6.69 (1H, dd, J = 5.8 Hz, 2.3 Hz), 4.96(2H, s), 4.54-4.47 (1H, m),4.16-4.08 (1H, m), 3.44-3.36(1H, m, (partly under waterpeak)), 3.28-3.16 (1H, m,(partly under water peak)),3.14-3.06 (1H, m).152UPLC-MS (ES+, final purity): 2.34 min, m/z 496.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.33 & 12.10 (0.2 & 0.8H, 2 × br s, mixture of tautomers), 10.02 (1H, br s), 8.16 (1H, d, J = 5.7 Hz), 7.79-7.71 (1.6H, m), 7.69- 7.56 (2H, m), 7.43-7.25 (2.4H, m), 7.24-7.12 (1H, m), 7.05 (1H, d, J = 2.6 Hz), 6.94 (1H, dd, J = 8.8 Hz, 2.6 Hz), 6.90 (1H, d, J = 8.8 Hz), 6.64(1H, dd, J = 5.7 Hz, 2.4 Hz),4.56-4.49 (1H, m), 4.17-4.09(1H, m), 3.45-3.35 (2H, m),3.29-3.20 (1H, m), 3.16-3.08(1H, m), 2.73-2.62 (4H, m),1.80-1.72 (4H, m). (1Hmissing (underneath waterand/or DMSO peak)).154UPLC-MS (ES+, final purity): 2.64 min, m/z 494.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.14 (1H, br s), 10.98 (1H, s), 8.50 (1H, s), 8.21 (1H, d, J = 5.7 Hz), 7.97 (1H, s), 7.76-7.69 (2H, m), 7.60-7.47 (2H, m), 7.38-7.30 (2H, m), 7.21-7.14 (1H, m), 7.02 (1H, d, J = 2.7 Hz), 6.92 (1H, dd, J = 8.8 Hz, 2.7 Hz), 6.88 (1H, d, J = 8.8 Hz), 6.70 (1H, dd, J = 5.7 Hz, 2.4 Hz),5.18 (2H, s), 4.54-4.47 (1H,m), 4.16-4.08 (1H, m), 3.44-3.35 (1H, m), 3.27-3.18 (1H,m), 3.15-3.06 (1H, m). Example 38. Synthesis of 2-(4-methylpiperazin-1-yl)-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]acetamide (Compound 146) Step 1—2-(4-methylpiperazin-1-yl)-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]acetamide To a solution of 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyridin-2-amine (100 mg, 0.19 mmol) and Et3N (68 μL, 0.49 mmol) in anhydrous THF (2 mL) at 0° C. under a nitrogen atmosphere was slowly added a solution of chloroacetyl chloride (16 μL, 0.20 mmol) in THF (0.5 mL) and the mixture was stirred at 25° C. for 1 hour. A solution of 1-methylpiperazine (26 μL, 0.23 mmol) in anhydrous THF (0.5 mL) was then added dropwise and the mixture was stirred at 25° C. for 18 hrs. Additional 1-methylpiperazine (26 μL, 0.23 mmol) was added and the mixture heated at 65° C. for 4 hours. After cooling to room temperature, the solvent was removed under reduce pressure and the residue purified by column chromatography using as eluent a gradient 0-20% MeOH in DCM to give 2-(4-methylpiperazin-1-yl)-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]acetamide (50.5 mg, 0.077 mmol, 40% yield) as a yellow solid. UPLC-MS (ES+, Short acidic): 1.68 min, m/z 655.4 [M+H]+. Step 2—2-(4-methylpiperazin-1-yl)-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]acetamide To a solution of 2-(4-methylpiperazin-1-yl)-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]acetamide (49.3 mg, 0.0800 mmol) in DCM (1.2 mL) was added trifluoroacetic acid (0.6 mL, 7.84 mmol) and the mixture was stirred at 25° C., in a sealed tube, for 22 hrs. The mixture was concentrated and the residue purified by flash chromatography (C18, 12 g column, gradient 0-50% MeCN in H2O+0.1% HCO2H). Pure fractions were loaded onto a SCX-2 column pre-equilibrated with methanol. The column was washed with methanol and the filtrate was discarded. The product was then eluted with NH32N in methanol and the filtrate was concentrated to afford 2-(4-methylpiperazin-1-yl)-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]acetamide (26 mg, 0.050 mmol, 66% yield) as a white solid. UPLC-MS (ES+, final purity): 2.29 min, m/z 525.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.31 (0.2H, br s), 12.09 (0.8H, br s), 9.89 (1H, s), 8.17 (1H, d, J=5.8 Hz), 7.77-7.73 (1.6H, m), 7.66 (1H, d, J=2.4 Hz), 7.65-7.61 (0.4H, m), 7.61-7.57 (0.8H, m), 7.43-7.37 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.19-7.13 (0.8H, m), 7.06-7.02 (1H, m), 6.94 (1H, dd, J=8.8, 2.7 Hz), 6.90 (1H, d, J=8.8 Hz), 6.66 (1H, dd, J=5.8, 2.4 Hz), 4.55-4.49 (1H, m), 4.17-4.09 (1H, m), 3.45-3.35 (1H, m), 3.26-3.19 (1H, m), 3.16-3.07 (3H, m), 2.37 (4H, m, seen as a br s), 2.18 (3H, s). 4H missing (underneath DMSO/water peaks). The compounds in the table below were made in an analogous manner, using the appropriate amine in step 1: Comp.NoStructure and NameData149UPLC-MS (ES+, final purity): 2.39 min, m/z 512.3 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.15 (1H, br s), 10.02 (1H, s), 8.17 (1H, d, J = 5.7 Hz), 7.76-7.70 (2H, m), 7.66 (1H, d, J = 2.3 Hz), 7.54 (1H, br s), 7.38-7.31 (2H, m), 7.21-7.15 (1H, m), 7.04 (1H, d, J = 2.7 Hz), 6.94 (1H, dd, J = 8.8 Hz, 2.7 Hz), 6.90 (1H, d, J = 8.8 Hz), 6.66 (1H, dd, J = 5.7 Hz, 2.3 Hz), 4.56-4.49 (1H, m), 4.17-4.10 (1H, m), 3.65-3.59 (4H, m), 3.46-3.36(1H, m), 3.30-3.08 (4H, m),2.59-2.51 (3H, m). (1Hmissing (underneath DMSOor water peak))145UPLC-MS (ES+, final purity): 2.30 min, m/z 512.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.33 & 12.10 (0.2 & 0.8H, 2 × br s, mixture of tautomers), 10.03 (1H, br s), 8.17 (1H, d, J = 5.7 Hz), 7.80-7.54 (3.8H, m), 7.42- 7.28 (2.2H, m), 7.22-7.12 (1H, m), 7.04 (1H, d, J = 2.7 Hz), 6.94 (1H, dd, J = 8.8 Hz, 2.7 Hz), 6.90 (1H, d, J = 8.8 Hz), 6.66 (1H, dd, J = 5.7 Hz, 2.4 Hz), 4.88 (1H, brs), 4.56-4.49 (1H, m), 4.27-4.20 (1H, m), 4.17-4.09 (1H,m), 3.45-3.35 (2H, m), 3.29-3.19 (1H, m), 3.16-3.07 (1H,m), 2.91-2.77 (2H, m), 2.65-2.54 (1H, m), 2.09-1.97 (1H,m), 1.69-1.58 (1H, m). (2Hmissing (underneath waterand/or DMSO peak))153UPLC-MS (ES+, final purity): 2.29 min, m/z 498.2 [M + H]+.1H NMR (400 MHz, DMSO- d6) δ/ppm: 12.32 & 12.10 (0.2 & 0.8H, 2 × br s), 9.96 (1H, s), 8.16 (1H, d, J = 5.7 Hz), 7.78-7.71 (1.6H, m), 7.66-7.56 (2H, m), 7.44-7.25 (2.4H, m), 7.21-7.12 (1H, m), 7.04 (1H, d, J = 2.7 Hz), 6.93 (1H, dd, J = 8.8 Hz, 2.7 Hz), 6.90 (1H, d, J = 8.8 Hz), 6.65 (1H, dd, J = 5.7 Hz, 2.4 Hz), 5.41 (1H, d, J = 6.5 Hz),4.56-4.48 (1H, m), 4.30-4.21(1H, m), 4.17-4.08 (1H, m),3.71-3.63 (2H, m), 3.45-3.35(1H, m), 3.29-3.19 (1H, m),3.16-3.07 (1H, m), 3.06-2.93(2H, m). (2H missing(underneath water and/orDMSO peak)) Example 39. Synthesis of 2-morpholino-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]propenamide (Compound 150) Step 1—2-morpholino-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]propanamide To a solution of 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyridin-2-amine (68 mg, 0.13 mmol) and triethylamine (55 μL, 0.40 mmol) in anhydrous THF (1.3 mL) at 0° C. under a nitrogen atmosphere slowly added solution of chloropropionyl chloride (16 μL, 0.16 mmol) in anhydrous THF (0.5 mL) and the mixture was stirred at room temperature for 2 hrs. To this mixture was added morpholine (23 μL, 0.26 mmol) and sodium iodide (40 mg, 0.26 mmol) and the reaction was heated at 60° C. for 72 hours. After cooling to room temperature, the mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-4% MeOH in DCM to give 2-morpholino-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]propanamide (43 mg, 0.065 mmol, 49% yield) as an off-white solid. UPLC-MS (ES+, Short acidic): 1.82 min, m/z 656.5 [M+H]+. Step 2—2-morpholino-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]propenamide To a solution of 2-morpholino-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2-pyridyl]propanamide (42 mg, 0.06 mmol) in DCM (1 mL) was added trifluoroacetic acid (490 μL, 6.4 mmol) and the mixture was stirred at 25° C. in a sealed tube for 22 hrs. The mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-50% (acetonitrile+0.1% formic acid):(water+0.1% formic acid). Fractions containing the product were loaded onto a SCX-2 column, which was flushed with MeOH and then MeOH+NH3to give 2-morpholino-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridyl]propanamide (23 mg, 0.043 mmol, 68% yield) as a white solid. UPLC-MS (ES+, final purity): 2.46 min, m/z 526.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.16 (1H, br s), 10.13 (1H, s), 8.17 (1H, d, J=5.7 Hz), 7.76-7.70 (2H, m), 7.68 (1H, d, J=2.3 Hz), 7.54 (1H, br s), 7.38-7.31 (2H, m), 7.21-7.15 (1H, m), 7.04 (1H, d, J=2.7 Hz), 6.94 (1H, dd, J=8.8 Hz, 2.7 Hz), 6.90 (1H, d, J=8.8 Hz), 6.66 (1H, dd, J=5.7 Hz, 2.3 Hz), 4.56-4.50 (1H, m), 4.18-4.10 (1H, m), 3.65-3.56 (4H, m), 3.46-3.35 (2H, m), 3.30-3.08 (2H, m), 1.15 (3H, d, J=6.7 Hz). 4H missing (underneath DMSO or water peak). Example 40. Synthesis of N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (Compound 148) Step 1—4-chloro-6-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-pyrimidin-2-amine A mixture of 4,6-dichloropyrimidin-2-amine (51.2 mg, 0.31 mmol), 3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (132 mg, 0.31 mmol) and Cs2CO3(152.6 mg, 0.47 mmol) in DMF (1.5 mL) was heated at 60° C. overnight. After cooling to rt, the mixture was poured into water (30 mL) and the aqueous was extracted with EtOAc (5×20 mL) then EtOAc:MeOH (10:1, 5×20 mL). The combined organic extracts were dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-4% MeOH in DCM to give 4-chloro-6-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-pyrimidin-2-amine (120 mg, 0.22 mmol, 70% yield) as a white foam. UPLC-MS (ES+, short acidic): 2.10 min, m/z 550.5 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 7.77 (1H, s), 7.76-7.72 (2H, m), 7.38-7.32 (2H, m), 7.22-7.17 (1H, m), 7.13 (2H, br s), 7.01 (1H, d, J=2.8 Hz), 6.95 (1H, dd, J=8.8, 2.8 Hz), 6.87 (1H, d, J=8.8 Hz), 6.13 (1H, s), 5.45 (2H, m), 4.49-4.42 (1H, m), 4.08-4.00 (1H, m), 3.63-3.46 (3H, m), 3.31-3.23 (1H, m), 3.07-2.97 (1H, m), 0.94-0.85 (2H, m), −0.02 (9H, s). Step 2—4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-amine To a mixture of 4-chloro-6-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-pyrimidin-2-amine (119 mg, 0.22 mmol) and triethylamine (33 μL, 0.24 mmol) in THF (7 mL) was added palladium, 10 wt. % on carbon powder, wet (57.5 mg, 0.05 mmol). The reaction was fitted with a H2balloon, extra H2added and subjected to 3×vacuum/H2cycles and then left to stir under a H2atmosphere for 72 hours. The mixture was filtered through a plug of diatomaceous earth and the filter cake was washed with THF and MeOH. The filtrate was concentrated and the residue partitioned between EtOAc (10 mL) and water (10 mL). The organic layer was separated and the aqueous extracted with EtOAc (3×10 mL). The combined organic extracts were washed with brine (10 mL), dried over Na2SO4, filtered and the solvent removed in vacuo to give 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-amine (111.5 mg, 0.22 mmol, 100% yield) as a white foam. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.75 min, m/z 516.5 [M+H]+. Step 3—N-(cyclopropanecarbonyl)-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide To a solution of 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-amine (111.5 mg, 0.22 mmol) in pyridine (1.6 mL) and DCM (1.1 mL) was added cyclopropanecarbonyl chloride (25 μL, 0.27 mmol) in DCM (0.1 mL) dropwise and the mixture was stirred at 25° C. under a nitrogen atmosphere for 18 hours. A solution of cyclopropanecarbonyl chloride (98 μL, 1.08 mmol) in DCM (0.1 mL) was slowly added and the mixture was stirred at 25° C. for 5 hours. The crude was diluted with DCM (30 mL), washed with water (15 mL) and brine (15 mL), dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-40% EtOAc in petroleum ether to give N-(cyclopropanecarbonyl)-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (101 mg, 0.16 mmol, 72% yield) as colourless oil, which solidified upon standing. UPLC-MS (ES+, short acidic): 2.14 min, m/z 652.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 8.76 (1H, d, J=5.8 Hz), 7.77 (1H, s), 7.77-7.72 (2H, m), 7.38-7.32 (2H, m), 7.23-7.16 (1H, m), 7.13 (1H, d, J=5.8 Hz), 7.07 (1H, d, J=2.8 Hz), 7.00 (1H, dd, J=8.8, 2.8 Hz), 6.91 (1H, d, J=8.8 Hz), 5.47 (1H, d, J=11.1 Hz), 5.43 (1H, d, J=11.1 Hz), 4.50-4.44 (1H, m), 4.09-4.02 (1H, m), 3.61-3.51 (3H, m), 3.31-3.25 (1H, m), 3.06-2.97 (1H, m), 1.95-1.87 (2H, m), 0.97-0.88 (10H, m), −0.02 (9H, s). Step 4—N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide N-(cyclopropanecarbonyl)-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (96 mg, 0.15 mmol) was dissolved in NH3(7M in MeOH—3 mL, 21 mmol) and the mixture was stirred at 25° C. for 1 hour. The solvent was removed under reduce pressure and the residue was purified by column chromatography using as eluent a gradient 0-80% EtOAc in petroleum ether to give N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (70 mg, 0.12 mmol, 82% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.97 min, m/z 584.4 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.62 (1H, s), 8.47 (1H, d, J=5.7 Hz), 7.77 (1H, s), 7.77-7.72 (2H, m), 7.38-7.32 (2H, m), 7.22-7.17 (1H, m), 7.10 (1H, d, J=2.8 Hz), 7.02 (1H, dd, J=8.8, 2.8 Hz), 6.89 (1H, d, J=8.8 Hz), 6.63 (1H, d, J=5.7 Hz), 5.47 (1H, d, J=11.1 Hz), 5.42 (1H, d, J=11.1 Hz), 4.50-4.43 (1H, m), 4.09-4.02 (1H, m), 3.61-3.54 (2H, m), 3.54-3.47 (1H, m), 3.31-3.24 (1H, m), 3.07-2.98 (1H, m), 2.24-2.16 (1H, m), 0.93-0.86 (2H, m), 0.80-0.68 (4H, m), −0.02 (9H, s). Step 5—N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide To a solution of N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (69 mg, 0.12 mmol) in DCM (1.8 mL) was added trifluoroacetic acid (0.9 mL, 11.75 mmol) and the mixture was stirred at 25° C. for 18 hours. The mixture was concentrated and the residue loaded onto a SCX-2 column, which was washed at first with MeOH and then NH3in MeOH to elute the product. The residue was purified by column chromatography using as eluent a gradient 0-8% MeOH in DCM. Like fractions were pooled and concentrated to afford N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (44 mg, 0.096 mmol, 82% yield) as a white solid. UPLC-MS (ES+, final purity): 2.65 min, m/z 454.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.32 (0.2H, br s), 12.10 (0.8H, br s), 10.62 (1H, s), 8.46 (1H, d, J=5.7 Hz), 7.79-7.72 (1.6H, m), 7.67-7.56 (1.2H, m), 7.45-7.36 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.20-7.12 (0.8H, m), 7.10 (1H, d, J=2.8 Hz), 7.00 (1H, dd, J=8.8, 2.8 Hz), 6.86 (1H, d, J=8.8 Hz), 6.63 (1H, d, J=5.7 Hz), 4.54-4.48 (1H, m), 4.15-4.06 (1H, m), 3.43-3.33 (1H, m), 3.29-3.18 (1H, m), 3.16-3.06 (1H, m), 2.23-2.15 (1H, m), 0.80-0.66 (4H, m). Example 41. Synthesis of 2-(4-methyl-1H-imidazol-2-yl)-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine (Compound 143) Step 1—tert-butyl 6-[[2-[4-methyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]-4-pyridyl]oxy]chromane-3-carboxylate A mixture of 2-[[2-(4-bromo-2-pyridyl)-4-methyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane (285 mg, 0.77 mmol), tert-butyl 6-hydroxychromane-3-carboxylate (213 mg, 0.85 mmol) and potassium phosphate tribasic (328.5 mg, 1.55 mmol) in anhydrous toluene (3.8 mL) was degassed under a nitrogen atmosphere for 10 minutes; then palladium (II) acetate (10.4 mg, 0.05 mmol) and 2-(di-tert-butylphosphino)biphenyl (27.7 mg, 0.09 mmol) were added. The vial was sealed and the mixture heated at 100° C. for 4 hours. After cooling to room temperature, the mixture was filtered through a plug of diatomaceous earth and the filter cake washed with DCM. The filtrate was concentrated and the residue purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give tert-butyl 6-[[2-[4-methyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]-4-pyridyl]oxy]chromane-3-carboxylate (330 mg, 0.61 mmol, 79% yield) as a dark orange oil. UPLC-MS (ES+, Short acidic): 1.92 & 1.98 min, m/z 538.8 [M+H]+.1H NMR (400 MHz, DMSO-d6) consistent with a ˜ 2.5/1 ratio of regioisomers. Step 2—formic acid; 6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]chromane-3-carboxylic Acid (0.4; 1) To a solution of tert-butyl 6-[[2-[4-methyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]-4-pyridyl]oxy]chromane-3-carboxylate (328 mg, 0.61 mmol) in DCM (4.6 mL) was added trifluoroacetic acid (2.3 mL, 30.04 mmol) and the mixture was stirred at room temperature, in a sealed vial, overnight. The mixture was concentrated and the residue azeotroped with toluene. The residue was purified by reverse column chromatography using as eluent a gradient 0-50% acetonitrile+0.1% formic acid in water+0.1% formic acid to give formic acid; 6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]chromane-3-carboxylic acid (0.4; 1) (148 mg, 0.40 mmol, 66% yield) as a white solid. UPLC-MS (ES+, Short acidic): 1.14 min, m/z 352.4 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 13.26 (1H, br s), 12.72 (1H, br s), 8.47 (1H, d, J=5.7 Hz), 8.13 (0.4H, s, formate), 7.37 (1H, d, J=2.3 Hz), 7.05 (1H, d, J=2.8 Hz), 6.98-6.93 (3H, m), 6.88 (1H, d, J=8.8 Hz), 4.36 (1H, dd, J=10.8 Hz, 3.1 Hz), 4.17 (1H, dd, J=10.8 Hz, 7.7 Hz), 3.07-2.95 (3H, m), 2.19 (3H, s). Step 3—6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]-N-phenacyl-chromane-3-carboxamide A mixture of formic acid; 6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]chromane-3-carboxylic acid (0.4; 1) (148 mg, 0.40 mmol), 2-aminoacetophenone hydrochloride (75.7 mg, 0.44 mmol), propylphosphonic anhydride (358 μL, 0.60 mmol) and N,N-diisopropylethylamine (245 μL, 1.41 mmol) in THF (4 mL) was heated at 65° C. for 2 hours. After cooling to room temperature, the mixture was concentrated and the residue taken up in EtOAc (30 mL). The organic layer was washed with water (2×10 mL) and brine (10 mL), dried over Na2SO4, filtered and the solvent removed in vacuo to give 6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]-N-phenacyl-chromane-3-carboxamide (188 mg, 0.40 mmol, 100% yield) as an orange solid. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.33 min, m/z 469.5 [M+H]+. Step 4—2-(4-methyl-1H-imidazol-2-yl)-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine To a suspension of 6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]-N-phenacyl-chromane-3-carboxamide (188 mg, 0.40 mmol) in 1-butanol (4 mL) were added ammonium acetate (309 mg, 4.01 mmol) and Et3N (56 μL, 0.40 mmol). The vial was sealed and the mixture irradiated at 150° C. for 45 minutes. After cooling to room temperature, the mixture was concentrated and the residue was purified by column chromatography using as eluent a gradient 0-20% MeOH. Fractions containing the product were combined and re-purified by reverse column chromatography using as eluent a gradient acetonitrile+0.1% formic acid. Fractions containing the product were loaded onto an SCX-2 column, which was flushed with MeOH followed by MeOH+NH3to give 2-(4-methyl-1H-imidazol-2-yl)-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-pyridine (42 mg, 0.093 mmol, 23% yield) as a white solid. UPLC-MS (ES+, final purity): 2.35 min, m/z 450.2 [M+H]+.1H NMR (400 MHz, DMSO-d6+few μL of CF3CO2D) δ/ppm: 8.65 (1H, d, J=5.7 Hz), 8.14 (1H, s), 7.84-7.80 (2H, m), 7.78 (1H, d, J=2.3 Hz), 7.58-7.51 (3H, m), 7.49-7.43 (1H, m), 7.15-7.10 (2H, m), 7.07 (1H, dd, J=8.8 Hz, 2.8 Hz), 7.01 (1H, d, J=8.8 Hz), 4.65-4.58 (1H, m), 4.40 (1H, dd, J=10.7 Hz, 9.0 Hz), 3.91-3.82 (1H, m), 3.44-3.27 (2H, m), 2.33 (3H, d, J=0.9 Hz). Example 42. Synthesis of 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N-phenacyl-chromane-3-carboxamide (Compound 151) Step 1—N-(6-chloropyrimidin-4-yl)cyclopropanecarboxamide To a solution of 6-chloropyrimidin-4-ylamine (800 mg, 6.18 mmol) and pyridine (1.25 mL, 15.46 mmol) in THF (25 mL) at 0° C., under a nitrogen atmosphere, was slowly added cyclopropanecarbonyl chloride (0.7 mL, 7.71 mmol) and the mixture was heated at 60° C. for 17 hours. After cooling to room temperature, the mixture was partitioned between water (50 mL) and EtOAc (50 mL). The two layers were separated and the aqueous extracted with EtOAc (2×50 mL). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography using as eluent a gradient 0-4% MeOH in DCM to give N-(6-chloropyrimidin-4-yl)cyclopropanecarboxamide (917 mg, 4.64 mmol, 75% yield) as a white solid. UPLC-MS (ES+, Short acidic): 1.35 min, m/z 197.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 11.54 (1H, s), 8.75 (1H, d, J=1.0 Hz), 8.10 (1H, d, J=1.0 Hz), 2.08-2.00 (1H, m), 0.93-0.86 (4H, m). Step 2—6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxychromane-3-carboxylic Acid A solution of 6-hydroxychromane-3-carboxylic acid (198 mg, 1.02 mmol), N-(6-chloropyrimidin-4-yl)cyclopropanecarboxamide (200 mg, 1.01 mmol) and potassium carbonate (560 mg, 4.05 mmol) in DMSO (1.25 mL) was heated 110° C. overnight. The reaction mixture was cooled to room temperature and poured into a solution of citric acid (777.8 mg, 4.05 mmol) in water (15 mL). The resulting precipitate was filtered, washed with water and dried to afford 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxychromane-3-carboxylic acid (316 mg, 0.89 mmol, 88% yield) as a beige solid. Compound used directly in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.44 min, m/z 356.2 [M+H]+(94%).1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.67 (1H, br s), 11.22 (1H, s), 8.48 (1H, d, J=0.9 Hz), 7.50 (1H, d, J=0.9 Hz), 6.98 (1H, d, J=2.8 Hz), 6.89 (1H, dd, J=8.8, 2.8 Hz), 6.80 (1H, d, J=8.8 Hz), 4.32 (1H, dd, J=10.8, 3.1 Hz), 4.15 (1H, dd, J=10.8, 7.5 Hz), 3.04-2.92 (3H, m), 2.06-1.98 (1H, m), 0.89-0.80 (4H, m). Step 3—6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N-phenacyl-chromane-3-carboxamide A mixture of 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxychromane-3-carboxylic acid (100 mg, 0.28 mmol), 2-aminoacetophenone hydrochloride (53 mg, 0.31 mmol), propylphosphonic anhydride (250 μL, 0.42 mmol) and N,N-diisopropylethylamine (152 μL, 0.87 mmol) in THF (2.8 mL) was heated at 65° C. for 1 hour. After cooling to room temperature, the mixture was concentrated and the residue dissolved in EtOAc (30 mL). The organic layer was washed with water (2×10 mL) and brine (10 mL), dried over Na2SO4, filtered and concentrated to afford 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N-phenacyl-chromane-3-carboxamide (131 mg, 0.28 mmol, 98% yield) as a beige solid. The compound used in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.61 min, m/z 473.2 [M+H]+. Step 4—6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N-phenacyl-chromane-3-carboxamide To a suspension of 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N-phenacyl-chromane-3-carboxamide (131 mg, 0.28 mmol) in 1-butanol (2.7 mL) were added ammonium acetate (213 mg, 2.77 mmol) and triethylamine (39 μL, 0.28 mmol). The vial was sealed and the mixture irradiated at 150° C. for 45 min. The mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-20% MeOH in DCM. Fractions containing the product was re-purified by reverse column chromatography using as eluent gradient 0-40% acetonitrile+0.10% formic acid in water+0.1% formic acid. Fractions containing the product were loaded onto a SCX-2 column, which was flushed at first with MeOH and the MeOH/NH3to give N-[6-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxypyrimidin-4-yl]cyclopropanecarboxamide (28 mg, 0.062 mmol, 22% yield) as a white solid. UPLC-MS (ES+, final purity): 2.94 min, m/z 454.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.30 (1H, br s), 11.23 (1H, s), 8.50 (1H, d, J=1.0 Hz), 7.76-7.70 (2H, m), 7.56 (1H, br s), 7.52 (1H, d, J=1.0 Hz), 7.38-7.32 (2H, m), 7.22-7.16 (1H, m), 7.03 (1H, d, J=2.8 Hz), 6.94 (1H, dd, J=8.8 Hz, 2.8 Hz), 6.86 (1H, d, J=8.8 Hz), 4.55-4.49 (1H, m), 4.18-4.10 (1H, m), 3.47-3.38 (1H, m), 3.29-3.19 (1H, m), 3.16-3.08 (1H, m), 2.07-1.99 (1H, m), 0.89-0.82 (4H, m). Example 43. Synthesis of 5-[3-(5-isopropyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 133) Step 1—tert-butyl 4-hydroxyiminopiperidine-1-carboxylate A mixture of 1-Boc-4-piperidone (2 g, 10.04 mmol), hydroxylamine hydrochloride (1.4 g, 20.08 mmol) and potassium acetate (1.97 g, 20.07 mmol) in EtOH (20 mL) was heated at 90° C. overnight. After cooling to room temperature, the mixture was concentrated and the residue taken up in water (20 mL). The aqueous layer was extracted with EtOAc (3×20 mL). The combined organic layers were washed with sat aq. NaHCO3(25 mL) and brine (25 mL), separated, dried over Na2SO4, filtered and concentrated to give tert-butyl 4-hydroxyiminopiperidine-1-carboxylate (2.067 g, 9.64 mmol, 96% yield) as a white solid. UPLC-MS (ES+, Short acidic): 1.36 min, m/z 215.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 10.44 (1H, s), 3.44-3.35 (4H, m), 2.46-2.41 (2H, m), 2.24-2.19 (2H, m), 1.40 (9H, s). Step 2—tert-butyl 3-amino-4,4-diethoxy-piperidine-1-carboxylate A mixture of tert-butyl 4-hydroxyiminopiperidine-1-carboxylate (2.06 g, 9.63 mmol), potassium carbonate (2.66 g, 19.28 mmol) and p-toluenesulfonyl chloride (1.84 g, 9.64 mmol) in THF (50 mL) was stirred at room temperature for 24 hours then heated at 40° C. for 20 hours. After cooling to room temperature, the mixture was filtered, and the solvent removed under reduce pressure. The residue was dissolved in EtOH (20 mL) and added dropwise to a mixture of potassium ethoxide (1.62 g, 19.27 mmol) and Na2SO4anhydrous (5.47 g, 38.54 mmol) in EtOH (20 mL) at 0° C. under a nitrogen atmosphere. The mixture was stirred at room temperature for 1.5 hours then heated at 60° C. for 1 hour. After cooling to room temperature, the mixture was filtered and the filter cake was washed with EtOH. The filtrate was concentrated and the residue purified by column chromatography using as eluent a gradient 0-8% MeOH in DCM to give tert-butyl 3-amino-4,4-diethoxy-piperidine-1-carboxylate (1.181 g, 4.10 mmol, 43% yield) as a thick yellow oil. UPLC-MS (ES+, Short acidic): 1.25 min, m/z 289.1 [M+H]+. Step 3—tert-butyl 4,4-diethoxy-3-[[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carbonyl]amino]piperidine-1-carboxylate A mixture of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (585 mg, 1.72 mmol), tert-butyl 3-amino-4,4-diethoxy-piperidine-1-carboxylate (741 mg, 2.57 mmol), propylphosphonic anhydride (2.05 mL, 3.44 mmol) and N,N-diisopropylethylamine (0.9 mL, 5.17 mmol) in THF (17.2 mL) was heated at 65° C. for 2 hours. The mixture was concentrated and the residue dissolved in EtOAc (100 mL). The organic layer was washed with water (2×30 mL) and brine (30 mL), dried over Na2SO4, filtered and concentrated to afford tert-butyl 4,4-diethoxy-3-[[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carbonyl]amino]piperidine-1-carboxylate (1049.7 mg, 1.72 mmol, 100% yield) as a light orange foam. The compound was used directly in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.79 min, m/z 611.7 [M+H]+. Step 4—tert-butyl 2-[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chroman-3-yl]-3,4,6,7-tetrahydroimidazo[4,5-c]pyridine-5-carboxylate A mixture of tert-butyl 4,4-diethoxy-3-[[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carbonyl]amino]piperidine-1-carboxylate (1.05 g, 1.72 mmol), ammonium acetate (2.65 g, 34.38 mmol) and p-toluenesulfonic acid monohydrate (65.4 mg, 0.34 mmol) in 1-butanol (11.5 mL) was irradiated at 150° C. for 1.5 hours. Additional ammonium acetate (2.65 g, 34.38 mmol) and p-toluenesulfonic acid monohydrate (65.4 mg, 0.34 mmol) were added and the mixture was further irradiated at 150° C. for 2 hours. The mixture was concentrated and the residue dissolved in EtOAc (100 mL). The organic layer was washed with sat. aq. NaHCO3(30 mL), water (30 mL) and brine (30 mL), dried over Na2SO4, filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM to give tert-butyl 2-[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chroman-3-yl]-3,4,6,7-tetrahydroimidazo[4,5-c]pyridine-5-carboxylate (214 mg, 0.41 mmol, 24% yield) as an off-white solid. UPLC-MS (ES+, Short acidic): 1.27 min, m/z 518.4 [M+H]+. Step 5—5-[3-(4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one To a mixture of tert-butyl 2-[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chroman-3-yl]-3,4,6,7-tetrahydroimidazo[4,5-c]pyridine-5-carboxylate (214 mg, 0.41 mmol) in DCM (2.5 mL) was added TFA (0.5 mL, 6.53 mmol) and stirred at 25° C. in a sealed vial for 1.5 hours. The mixture was concentrated and the residue azeotroped with toluene. The residue was dissolved in water and loaded onto an SCX-2 column. The column was washed with MeOH and then 2N NH3in methanol to elute the product. The filtrate was concentrated and dried to afford 5-[3-(4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (84 mg, 0.20 mmol, 49% yield) as a yellow solid. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 0.97 min, m/z 418.1 [M+H]+. Step 6—5-[3-(5-isopropyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one A mixture of 5-[3-(4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (65 mg, 0.16 mmol), acetone (22.9 μL, 0.31 mmol), sodium triacetoxyborohydride (66.1 mg, 0.31 mmol) and acetic acid (17.9 μL, 0.31 mmol) in DMF (1.5 mL) was stirred at 25° C. Additional acetone (22.9 μL, 0.31 mmol) and sodium triacetoxyborohydride (66.1 mg, 0.31 mmol) were added after 23 hours and 30 hours and the mixture was further stirred at 25° C. for 48 hours. The mixture was concentrated and the residue purified by reverse column chromatography using as eluent a gradient 0-40% (MeCN+0.1% formic acid) in (H2O+0.1% formic acid). Fractions containing the product were dried and loaded onto a SCX-2 column, which was flushed at first with MeOH and then 2N NH3in MeOH to elute 5-[3-(5-isopropyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (20.9 mg, 0.046 mmol, 29% yield) as a white solid. UPLC-MS (ES+, final purity): 2.05 min, m/z 460.2 [M+H]+.1H NMR (400 MHz, DMSO-d6+3drops of CF3CO2D) δ/ppm: 8.03 (1H, d, J=6.1 Hz), 7.04 (1H, d, J=2.7 Hz), 6.97 (1H, dd, J=8.8 Hz, 2.7 Hz), 6.91 (1H, d, J=8.8 Hz), 6.36 (1H, d, J=6.1 Hz), 4.50-4.44 (2H, m), 4.44-4.37 (2H, m), 3.92-3.84 (1H, m), 3.83-3.68 (2H, m), 3.45-3.20 (3H, m), 3.07-2.91 (4H, m), 2.61-2.55 (2H, m), 1.33 (6H, d, J=6.6 Hz). Exchangeable protons not seen. Example 44. Synthesis of 5-[3-(5-acetyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 135) Compound 135 was synthesised in analogy with Example 43 with the exception of Step 6 as follows: To a solution of 5-[3-(4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-TH-1,8-naphthyridin-2-one (36 mg, 0.09 mmol) and triethylamine (12 μL, 0.09 mmol) in DMF (0.8 mL) was added acetic anhydride (8.2 μL, 0.09 mmol) at 0° C. and the mixture was stirred at this temperature for 2 hours. The mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-40% (MeCN+0.1% formic acid) in (H2O+0.1% formic acid). Fractions containing the product were concentrated and loaded onto a SCX-2 column, which was flushed at first with MeOH and then NH32N in MeOH to give 5-[3-(5-acetyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-TH-1,8-naphthyridin-2-one (15.5 mg, 0.034 mmol, 39% yield) as a white solid. UPLC-MS (ES+, final purity): 2.22 and 2.25 min, m/z 460.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ/ppm: 12.08 (1H, br s), 10.45 (1H, s), 7.95 (1H, d, J=5.8 Hz), 6.97 (1H, d, J=2.6 Hz), 6.90 (1H, dd, J=8.8 Hz, 2.6 Hz), 6.86 (1H, d, J=8.8 Hz), 6.25 (1H, d, J=5.8 Hz), 4.46-4.31 (3H, m), 4.11-4.01 (1H, m), 3.75-3.63 (2H, m), 3.18-3.09 (1H, m), 3.09-3.00 (1H, m), 2.95-2.89 (2H, m), 2.66-2.61 (1H, m), 2.56-2.51 (3H, m), 2.08 & 2.05 (3H, 2s, acetamide rotamers). 1H missing (underneath water/DMSO peak). Example 45. Chiral Separation of Selected Compounds Conditions disclosed in table below can be used to separate stereoisomers of the racemic compound as shown: Comp.NoStructure and NameSeparation conditions32Instrument: MG II preparative SFC(SFC-14) Column: ChiralPak AD, 250 × 30 mm I.D., 10 μm Mobile phase: A for CO2 and B for Ethanol(0.1% NH3H2O) Gradient: B 40% Flow rate: 80 mL/min Back pressure: 100 bar Column temperature: 38° C. Wavelength: 220 nm Cycle time: ~6.8 min136Instrument: MG II preparative SFC(SFC-14) Column: ChiralPak AD, 250 × 30 mm I.D., 10 μm Mobile phase: A for CO2 and B for Ethanol Gradient: B 50% Flow rate: 80 mL/min Back pressure: 100 bar Column temperature: 38° C. Wavelength: 220 nm Cycle time: ~11 min Example 46. Biological Assay PGP-3 0R HCT-116AlphaLISA SureFire pERK1/2 Cellular Assay The human HCT-116 colorectal carcinoma cell line (ATCC CCL-247) endogenously expresses the KRASG13Dmutation, which leads to constitutive activation of the MAP kinase pathway and phosphorylation of ERK. To determine whether compounds inhibit constitutive ERK phosphorylation in HCT-116 cells, they were tested using AlphaLISA® SureFire® technology (Perkin Elmer p-ERK1/2 p-T202/Y204 assay kit ALSU-PERK-A10K). Assay read outs took place 2 or 24 hours after dosing with compounds. On the first day, HCT-116 cells were harvested, resuspended in growth medium (McCoys5A with Glutamax (Life Technologies 36600021) and 10% heat-inactivated fetal bovine serum (Sigma F9665)), and counted. Cells were plated in 100 μl per well in each well of a 96-well culture dish (Sigma CLS3598) to a final density of 30,000 (2 hr read) or 15,000 (24 hr read) cells per well and incubated over night at 37° C. and 5% CO2. On day 2, the growth medium was exchanged for dosing medium (McCoys5A with Glutamax (Life Technologies 36600021) and 1% heat-inactivated fetal bovine serum (Sigma F9665)) and the cells were dosed with compounds to produce a 10-point dose response, where the top concentration was 1 μM and subsequent concentrations were at ⅓ log dilution intervals. A matched DMSO control was included. The cells were subsequently incubated for either 2 or 24 hours at 37° C. and 5% CO2. After incubation, media was removed and the cells were incubated with lysis buffer containing phosphatase inhibitors for 15 minutes at room temperature. Cell lysates were transferred to a ½ area 96 well white Optiplate™ (Perkin Elmer 6005569) and incubated with anti-mouse IgG acceptor beads, a biotinylated anti-ERK1/2 rabbit antibody recognizing both phosphorylated and non-phosphorylated ERK1/2, a mouse antibody targeted to the Thr202/Tyr204 epitope and recognizing phosphorylated ERK proteins only, and streptavidin-coated donor beads. The biotinylated antibody binds to the streptavidin-coated donor beads and the phopsho-ERK1/2 antibody binds to the acceptor beads. Plates were read on an EnVision reader (Perkin Elmer) and excitation of the beads at 680 nm with a laser induced the release of singlet oxygen molecules from the donor beads that trigger energy transfer to the acceptor beads in close proximity, producing a signal that can be measured at 570 nm. Both antibodies bound to phosphorylated ERK proteins, bringing the donor and acceptor beads into close proximity. All data were analyzed using the Dotmatics or GraphPad Prism software packages. Inhibition of ERK phosphorylation was assessed by determination of the absolute IC50value, which is defined as the concentration of compound required to decrease the level of phosphorylated ERK proteins by 50% when compared to DMSO control. WiDr AlphaLISA SureFire pERK1/2 Cellular Assay The human WiDr colorectal adenocarcinoma cell line (ATCC CCL-218) endogenously expresses the BRAFV600Emutation, which leads to constitutive activation of the MAP kinase pathway and phosphorylation of ERK. To determine whether compounds inhibit constitutive ERK phosphorylation in WiDr cells, they were tested using AlphaLISA® SureFire® technology (Perkin Elmer p-ERK1/2 p-T202/Y204 assay kit ALSU-PERK-A10K). The main procedure is essentially the same as for HCT-116 cells (above), with the following adjustments to the growth medium (Eagle's Minimum Essential Medium (Sigma M2279) with 1× Glutamax (Life Technologies 35050038), 1× Sodium-Pyruvate (Sigma S8636), and 10% heat-inactivated fetal bovine serum (Sigma F9665)), the dosing medium (Eagle's Minimum Essential Medium (Sigma M2279) with 1× Glutamax (Life Technologies 35050038), 1× Sodium-Pyruvate (Sigma S8636), and 1% heat-inactivated fetal bovine serum (Sigma F9665)), and the seeding densities (2 hr: 50,000 cells per well; 24 hr: 35,000 cells per well). Moreover, the compounds were dosed in ½ log dilution intervals with the top concentration of 10 μM. HCT-116 AlphaLISA SureFire pERK1/2 Cellular Assay (Dimer) The human HCT-116 colorectal carcinoma cell line (ATCC CCL-247) endogenously expresses the KRASG13Dmutation, which leads to constitutive activation of the MAP kinase pathway and phosphorylation of ERK. First generation RAF inhibitors can promote RAF dimer formation in KRAS mutant tumours leading to a paradoxical activation of the pathway. To determine whether compounds can circumvent this problem and inhibit RAF dimers in HCT-116 cells, they were tested using AlphaLISA® SureFire® technology (Perkin Elmer p-ERK1/2 p-T202/Y204 assay kit ALSU-PERK-A10K). The main procedure is essentially the same as described above, with the following adjustments: Cells were seeded with the seeding density of 30,000 cells per well. On the second day (the day of dosing) no medium change was performed and the cells were dosed with 1 μM of Encorafenib for 1 hour (at 37° C. and 5% CO2) to induce RAF dimers and promote paradoxical dimer-dependent pERK signalling. After incubation, the cells were washed, 100 μl fresh growth medium was added, and cells were dosed with compounds of interest to produce a 10-point dose response, where the top concentration was 10 μM and subsequent concentrations are at ½ log dilution intervals. Cells were incubated for another hour at 37° C. and 5% CO2before lysis and processing with the pERK AlphaLISA® SureFire® kit as described above. A375 AlphaLISA SureFire pERK1/2 Cellular Assay (Monomer) The human A375 melanoma cell line (ATCC CRL-1619) endogenously expresses the BRAFV600Emutation, which leads to constitutive activation of the MAP kinase pathway and phosphorylation of ERK. In BRAFV600Emutant tumours, BRAF signals as a monomer to activate ERK. To determine whether compounds can inhibit BRAF monomers in A375 cells, they were tested using AlphaLISA® SureFire® technology (Perkin Elmer p-ERK1/2 p-T202/Y204 assay kit ALSU-PERK-A10K). The main procedure is essentially the same as described above for HCT-116 cells, with the following adjustments: The A375 cells were cultivated and dosed in Dulbecco's modified Eagle's medium containing 4.5 g/L D-glucose (Sigma D6546), 10% heat-inactivated fetal bovine serum (Sigma F9665), and 1% Sodium-Pyruvate (Sigma S8636), and seeded with a seeding density of 30,000 cells per well. No media exchange was performed before dosing with compounds to produce a 10-point dose response, where the top concentration was 10 μM and subsequent concentrations were at ½ log dilution intervals. Subsequently, the cells were incubated for 1 hour at 37° C. and 5% CO2before lysis. HCT-116 CellTiter-Glo 3D Cell Proliferation Assay The human HCT-116 colorectal carcinoma cell line (ATCC CCL-247) endogenously expresses the KRASG13D mutation, which leads to enhanced survival and proliferative signaling. To determine whether compounds inhibit the proliferation of HCT-116 cells, they are tested using the CellTiter-Glo® 3D Cell Viability Assay Kit (Promega G9683). On the first day, HCT-116 cells were harvested, resuspended in growth medium (McCoys5A with Glutamax (Life Technologies 36600021) with 10% heat-inactivated fetal bovine serum (Sigma F9665)), and counted. Cells were plated in 100 μl per well in each well of a Corning 7007 96-well clear round bottom Ultra-Low Attachment plate (VWR 444-1020) to a final density of 1000 cells per well. Cells were seeded for pre- and post-treatment readouts. The cells were then incubated at 37° C. and 5% CO2for 3 days (72 hours) to allow spheroid formation. After 72 hours, the plate seeded for a pre-treatment read was removed from the incubator to allow equilibration to room temperature for 30 minutes, before CellTitre-Glo® reagent was added to each well. The plates were incubated at room temperature for 5 minutes shaking at 300 rpm, followed by an incubation of 25 minutes on the benchtop before being read on the Envision reader (Perkin Elmer) as described below. On the same day, the cells plated for the post-treatment readout were dosed with compounds to produce a 9-point dose response, where the top concentration was 15 μM and following concentrations were at ½ log dilution intervals. These cells were subsequently incubated at 37° C. and 5% CO2for another 4 days (96 hours). After 4 days, the plate was removed from the incubator to allow equilibration to room temperature for 30 minutes and treated with CellTitre Glo® reagent as stated above. The method allows the quantification of ATP present in the wells, which is directly proportional to the amount of viable—hence metabolically active—cells in 3D cells cultures. The CellTitre Glo® reagent lyses the cells and contains luciferin and a luciferase (Ultra-Glo™ Recombinant Luciferase), which in the presence of ATP and oxygen can produce bioluminescence from luciferin. Therefore, plates were read on an EnVision reader (Perkin Elmer) and luminescence signals were recorded. Cell proliferation was determined on 4 days after dosing relative to the pre-treatment read. All data were analyzed using the Dotmatics or GraphPad Prism software packages. Inhibition of proliferation was assessed by determination of the GI50value, which was defined as the concentration of compound required to decrease the level of cell proliferation by 50% when compared to DMSO control. WiDr CellTiter-Glo 3D Cell Proliferation Assay The human WiDr colorectal adenocarcinoma cell line (ATCC CCL-218) endogenously expresses the BRAFV600Emutation, which leads to enhanced survival and proliferative signaling. To determine whether compounds inhibit the proliferation of WiDr cells, they were tested using the CellTiter-Glo® 3D Cell Viability Assay Kit (Promega G9683) as stated for HCT-116 cells, with the following adjustments to the growth medium: Eagle's Minimum Essential Medium (Sigma M2279) with 1× Glutamax (Life Technologies 35050038), 1× Sodium-Pyruvate (Sigma S8636) and 10% heat-inactivated fetal bovine serum (Sigma F9665). Microsomal Stability Assay The stability studies were performed manually using the substrate depletion approach. Test compounds were incubated at 37° C. with cryo-preserved mouse or human liver microsomes (Corning) at a protein concentration of 0.5 mg·mL−1and a final substrate concentration of 1 μM. Aliquots were removed from the incubation at defined timepoints and the reaction was terminated by adding to ice-cold organic solvent. Compound concentrations were determined by LC-MS/MS analysis. The natural log of the percentage of compound remaining was plotted against each time point and the slope determined. The half-life (t1/2) and CLint were calculated using Equations 1 and 2, respectively. Data analysis was performed using Excel (Microsoft, USA). t1/2(min)=0.693/−slope  (1) CLint(μL/min/mg)=(LN(2)/t1/2(min))*1000/microsomal protein (mg/mL)  (2) HLM (human liver microsomes) and MLM (mouse liver microsomes) stability assay results are described in Tables 1-2. Plasma Protein Binding Assay The plasma protein binding was determined by the equilibrium dialysis method. A known concentration of compound (5 μM) in previously frozen human or mouse plasma (Sera Labs) was dialysed against phosphate buffer using a RED device (Life Technologies) for 4 hours at 37° C. The concentration of compound in the protein containing (PC) and protein free (PF) sides of the dialysis membrane were determined by LC-MS/MS and the % free compound was determined by equation 4. Data analysis was performed using Excel (Microsoft, USA). % free=(1−((PC−PF)/PC))×100  (4) hPPB (human plasma protein binding) and mPPB (mouse plasma protein binding) results are described in Tables 1-2. FeSSIF Solubility Assay 1 mL of fed state simulated intestinal fluid (FeSSIF), prepared using FaSSIF/FeSSIF/FaSSGF powder (Biorelevant.com) and pH 5 acetate buffer, was added to 1.0 mg of compound and then incubated for 24 h (Bioshake iQ, 650 rpm, 37° C.). Following filtration under positive pressure, the concentration of compound in solution was assessed by LC-UV in comparison to the response for a calibration standard of known concentration (250 μM). FeSSIF solubility results are described in Tables 1-2. TABLE 1pERKpERKpERKpERKHCT116HCT116WiDrHLMMLMA375dimer(2 hr)(2 hr)(CLint)(CLint)Comp.(1 hr)(1 hr)AbsAbsμL/μL/hPPBmPPBSolNopIC50pIC50pIC50pIC50min/mgmin/mg(% free)(% free)mg/L17.557.17ndnd123.1108.80.20.5<1.227.907.23ndnd147.385.80.31<1.137.366.95ndnd30.334.40.20.5nd47.016.57ndnd261.4121.2<0.10.5<1.356.736.72ndnd33.928.50.30.4<1.266.016.35ndnd33.418.12.41.3<1.476.166.72ndnd67.939.21.90.9<1.486.506.576.766.753613.91.01.118.5 (FESSIF)96.486.73ndnd70.56.10.60.4<1.1106.086.55ndnd89.145.90.60.7<1.2116.596.67ndnd10.329.10.50.1<1.2127.376.646.596.9188.615.7<0.1<0.1<1.3137.456.91ndnd77.1114.10.71.2<1.2147.747.12ndnd60.751.30.10.5<1.1157.627.17ndnd39.933.90.21.3<1.1166.246.21ndndndndndnd<1.2176.036.46ndnd31.516.50.72.4<1.21855.44ndnd71.132.91.22.5nd195.796.466.505.9627.8231.91.519.6 (FESSIF)206.136.22ndnd16.930.10.1<1.3216.036.56ndnd15.623.20.71.6<1.2227.706.816.856.9235.925.1<0.1<0.1<1.2236.596.656.506.6429.930.50.50.5<1.1246.256.816.396.3012.741.61.81.9<1.1256.946.947.076.8950.148.50.31.51.8 (FESSIF)267.196.566.386.5277.589.30.71<1.1276.416.42ndnd101.7148.1<0.1<0.1<1.2285.74ndndnd68.131.10.10.5nd297.496.666.886.97118.677.60.20.6<1.1306.795.88ndndndndndnd<1.3316.906.06ndnd34.626<0.10.2<1.2326.606.406.646.6911.38.31.33.2<1.2336.426.18ndnd77.33723.17345.35ndndnd46.98.60.73.228.7355.616.28ndnd27742.1<0.1nv<1.3366.286.76ndnd23.518.4nv<0.1<1.2376.306.47ndndnd18.4nvnv<1.2385.075.40ndnd34.3nvnv<1.339ndndndndnd15.4nvnd<1.2406.606.49ndndnd27.1nvnd<1.2416.296.52ndndnd46.1nv1.2<1.2426.056.13ndnd10.622nv<0.1<1.343ndndndndnd23.2nvnd<1.2445.926.546.276.4514.2110.30.9<1.2456.797.057.567.1858.12770.20.2<1.246ndndndnd18.445.51.92.3<1476.647.197.166.9324.532.50.70.327.5 (FESSIF)485.045.14ndnd40.538.423<1.1495.115.05ndnd82.283.61.92.5<1.2506.577.197.337.0170.4791.21.6<1.251ndndndnd156.1123.40.52.61.2 TABLE 2pERKpERKpERKpERKHCT116HCT116WiDrHLMMLMA375dimer(2 hr)(2 hr)3D3D(CLint)(CLint)FESSIFComp.(1 hr)(1 hr)AbsAbsHCT116WiDrμL/μL/hPPBmPPBSolNopIC50plC50pIC50pIC50pGI50pGI50min/mgmin/mg(% free)(% free)mg/L86.506.576.766.756.866.723613.91.01.118.51016.697.206.886.747.076.5129.615.21.61.15.71036.416.976.626.576.546.2711.64.40.60.65.21045.796.466.505.966.355.9427.8231.91.519.61056.387.196.756.576.616.2224.82.11.00.868.61066.036.916.566.236.365.8110.517.12.43.028.11076.757.246.726.686.646.1130.312.50.70.31085.206.41<6109<6<1.11106.977.307.057.157.156.7048.355.50.20.18.51116.276.976.666.496.505.9948.126.91.911.73701125.756.59<66.0081.555.51.42.051.11135.336.29<65.8382.138.20.61.6139.61145.596.44<65.8738.512.61.22.0496.71157.866.987.348.097.177.422772770.30.3129.11167.337.067.177.407.186.902772770.130.23311.41176.146.966.976.236.676.1543.313.42.62.31.41186.206.836.576.406.405.9381.2401.31.01196.1474.235.80.60.9<1.21206.02189.6890.90.61.3121<6206.8137.30.40.438.21226.546.796.886.456.746.2980.370.80.230.21123.61237.557.237.207.477.397.2797.5125.80.230.25156.51245.976.576.466.186.145.8465.828.22.73.931255.356.866.456.015.954.8222.01266.106.616.305.994.82121.155.71.00.7<1.31276.076.806.3838.713.40.60.26.01286.086.586.410.91.21.6129<616.71306.006.786.856.226.215.7823.623.50.10.2131<612.21326.126.376.466.086.185.9420.79.32.610.141.9133<6661134<67.49<65.214.824.5913.672.46.19.5135<633.21366.797.057.567.187.326.7758.12770.20.217.11376.727.557.457.137.156.9032.62770.1<0.173.91385.796.096.56134.1277<0.1<0.11395.976.666.7099.8277<0.1<0.1106.61405.235.69<65.905.885.7248.781.1<0.10.2141<61.021.8<0.10.5142<6135.41436.106.246.516.526.135.8236.186.1<0.1<0.144.71446.697.227.347.027.006.4227.129.30.70.8317.51456.276.756.786.486.325.2057.2147.30.40.531466.436.776.756.406.426.0933.489.71.50.7403.91476.206.366.496.065.865.8610.41020.50.8527.61485.916.346.526.066.315.92132.22110.90.4104.31495.886.446.506.036.075.6563.7230.90.250.14200150<6235.41516.466.696.756.656.425.9418.31610.21<0.186.61526.226.566.686.666.085.2529258.70.480.47997.11536.306.906.706.546.375.2519.292.90.390.63361547.017.427.557.487.216.1822.895.20.750.7170.91556.937.147.226.997.006.2028.92770.10.1123.81566.757.287.376.887.056.5730.3151.50.50.4296.21576.286.766.956.566.586.1835.7277<0.1<0.1116.31586.1321.362.80.30.2159<619.834.41.00.5<1.01606.066.466.876.666.215.682.74.50.3<0.13.01616.25<1.1162<613.7163<6<1.3 The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. While the invention has been described in connection with proposed specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth and as follows in the scope of the appended claims.
313,693
11858931
DETAILED DESCRIPTION OF THE INVENTION The present disclosure provides, inter alia, compounds of formula (I), and variations thereof, or a salt thereof, pharmaceutical compositions comprising compounds of formula (I) or a salt thereof, and methods of using such compounds and compositions in treating fibrotic diseases. Definitions For use herein, unless clearly indicated otherwise, use of the terms “a”, “an” and the like refers to one or more. Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”. “Alkyl” as used herein refers to and includes, unless otherwise stated, a saturated linear (i.e., unbranched) or branched univalent hydrocarbon chain or combination thereof, having the number of carbon atoms designated (i.e., C1-C10means one to ten carbon atoms). Particular alkyl groups are those having 1 to 20 carbon atoms (a “C1-C20alkyl”), having 1 to 10 carbon atoms (a “C1-C10alkyl”), having 6 to 10 carbon atoms (a “C6-C10alkyl”), having 1 to 6 carbon atoms (a “C1-C6alkyl”), having 2 to 6 carbon atoms (a “C2-C6alkyl”), or having 1 to 4 carbon atoms (a “C1-C4alkyl”). Examples of alkyl groups include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, and the like. “Alkylene” as used herein refers to the same residues as alkyl, but having bivalency. Particular alkylene groups are those having 1 to 20 carbon atoms (a “C1-C20alkylene”), having 1 to 10 carbon atoms (a “C1-C10alkylene”), having 6 to 10 carbon atoms (a “C6-C10alkylene”), having 1 to 6 carbon atoms (a “C1-C6alkylene”), 1 to 5 carbon atoms (a “C1-C5alkylene”), 1 to 4 carbon atoms (a “C1-C4alkylene”) or 1 to 3 carbon atoms (a “C1-C3alkylene”). Examples of alkylene include, but are not limited to, groups such as methylene (—CH2—), ethylene (—CH2CH2—), propylene (—CH2CH2CH2—), isopropylene (—CH2CH(CH3)—), butylene (—CH2(CH2)2CH2—), isobutylene (—CH2CH(CH3)CH2—), pentylene (—CH2(CH2)3CH2—), hexylene (—CH2(CH2)4CH2—), heptylene (—CH2(CH2)5CH2—), octylene (—CH2(CH2)8CH2—), and the like. “Alkenyl” as used herein refers to and includes, unless otherwise stated, an unsaturated linear (i.e., unbranched) or branched univalent hydrocarbon chain or combination thereof, having at least one site of olefinic unsaturation (i.e., having at least one moiety of the formula C═C) and having the number of carbon atoms designated (i.e., C2-C10means two to ten carbon atoms). An alkenyl group may have “cis” or “trans” configurations, or alternatively have “E” or “Z” configurations. Particular alkenyl groups are those having 2 to 20 carbon atoms (a “C2-C20alkenyl”), having 6 to 10 carbon atoms (a “C6-C10alkenyl”), having 2 to 8 carbon atoms (a “C2-C6alkenyl”), having 2 to 6 carbon atoms (a “C2-C6alkenyl”), or having 2 to 4 carbon atoms (a “C2-C4alkenyl”). Examples of alkenyl group include, but are not limited to, groups such as ethenyl (or vinyl), prop-1-enyl, prop-2-enyl (or allyl), 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, buta-1,3-dienyl, 2-methylbuta-1,3-dienyl, pent-1-enyl, pent-2-enyl, hex-1-enyl, hex-2-enyl, hex-3-enyl, and the like. “Alkenylene” as used herein refers to the same residues as alkenyl, but having bivalency. Particular alkenylene groups are those having 2 to 20 carbon atoms (a “C2-C20alkenylene”), having 2 to 10 carbon atoms (a “C2-C10alkenylene”), having 6 to 10 carbon atoms (a “C6-C10alkenylene”), having 2 to 6 carbon atoms (a “C2-C6alkenylene”), 2 to 4 carbon atoms (a “C2-C4alkenylene”) or 2 to 3 carbon atoms (a “C2-C3alkenylene”). Examples of alkenylene include, but are not limited to, groups such as ethenylene (or vinylene) (—CH═CH—), propenylene (—CH═CHCH2—), 1,4-but-1-enylene (—CH═CH—CH2CH2—), 1,4-but-2-enylene (—CH2CH═CHCH2—), 1,6-hex-1-enylene (—CH═CH—(CH2)3CH2—), and the like. “Alkynyl” as used herein refers to and includes, unless otherwise stated, an unsaturated linear (i.e., unbranched) or branched univalent hydrocarbon chain or combination thereof, having at least one site of acetylenic unsaturation (i.e., having at least one moiety of the formula C≡C) and having the number of carbon atoms designated (i.e., C2-C10means two to ten carbon atoms). Particular alkynyl groups are those having 2 to 20 carbon atoms (a “C2-C20alkynyl”), having 6 to 10 carbon atoms (a “C6-C10alkynyl”), having 2 to 8 carbon atoms (a “C2-C8alkynyl”), having 2 to 6 carbon atoms (a “C2-C6alkynyl”), or having 2 to 4 carbon atoms (a “C2-C4alkynyl”). Examples of alkynyl group include, but are not limited to, groups such as ethynyl (or acetylenyl), prop-1-ynyl, prop-2-ynyl (or propargyl), but-1-ynyl, but-2-ynyl, but-3-ynyl, and the like. “Alkynylene” as used herein refers to the same residues as alkynyl, but having bivalency. Particular alkynylene groups are those having 2 to 20 carbon atoms (a “C2-C20alkynylene”), having 2 to 10 carbon atoms (a “C2-C10alkynylene”), having 6 to 10 carbon atoms (a “C6-C10alkynylene”), having 2 to 6 carbon atoms (a “C2-C6alkynylene”), 2 to 4 carbon atoms (a “C2-C4alkynylene”) or 2 to 3 carbon atoms (a “C2-C3alkynylene”). Examples of alkynylene include, but are not limited to, groups such as ethynylene (or acetylenylene) (—C≡C—), propynylene (—C≡CCH2—), and the like. “Cycloalkyl” as used herein refers to and includes, unless otherwise stated, saturated cyclic univalent hydrocarbon structures, having the number of carbon atoms designated (i.e., C3-C10means three to ten carbon atoms). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantyl. A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. Particular cycloalkyl groups are those having from 3 to 12 annular carbon atoms. A preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a “C3-C8cycloalkyl”), having 3 to 6 annular carbon atoms (a “C3-C6cycloalkyl”), or having from 3 to 4 annular carbon atoms (a “C3-C4cycloalkyl”). Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, and the like. “Cycloalkylene” as used herein refers to the same residues as cycloalkyl, but having bivalency. Cycloalkylene can consist of one ring or multiple rings which may be fused, spiro or bridged, or combinations thereof. Particular cycloalkylene groups are those having from 3 to 12 annular carbon atoms. A preferred cycloalkylene is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a “C3-C8cycloalkylene”), having 3 to 6 carbon atoms (a “C3-C6cycloalkylene”), or having from 3 to 4 annular carbon atoms (a “C3-C4cycloalkylene”). Examples of cycloalkylene include, but are not limited to, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, cycloheptylene, norbornylene, and the like. A cycloalkylene may attach to the remaining structures via the same ring carbon atom or different ring carbon atoms. When a cycloalkylene attaches to the remaining structures via two different ring carbon atoms, the connecting bonds may be cis- or trans- to each other. For example, cyclopropylene may include 1,1-cyclopropylene and 1,2-cyclopropylene (e.g., cis-1,2-cyclopropylene or trans-1,2-cyclopropylene), or a mixture thereof. “Cycloalkenyl” refers to and includes, unless otherwise stated, an unsaturated cyclic non-aromatic univalent hydrocarbon structure, having at least one site of olefinic unsaturation (i.e., having at least one moiety of the formula C═C) and having the number of carbon atoms designated (i.e., C3-C10means three to ten carbon atoms). Cycloalkenyl can consist of one ring, such as cyclohexenyl, or multiple rings, such as norbornenyl. A preferred cycloalkenyl is an unsaturated cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a “C3-C8cycloalkenyl”). Examples of cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, norbornenyl, and the like. “Cycloalkenylene” as used herein refers to the same residues as cycloalkenyl, but having bivalency. “Aryl” or “Ar” as used herein refers to an unsaturated aromatic carbocyclic group having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) which condensed rings may or may not be aromatic. Particular aryl groups are those having from 6 to 14 annular carbon atoms (a “C6-C14aryl”). An aryl group having more than one ring where at least one ring is non-aromatic may be connected to the parent structure at either an aromatic ring position or at a non-aromatic ring position. In one variation, an aryl group having more than one ring where at least one ring is non-aromatic is connected to the parent structure at an aromatic ring position. “Arylene” as used herein refers to the same residues as aryl, but having bivalency. Particular arylene groups are those having from 6 to 14 annular carbon atoms (a “C6-C14arylene”). “Heteroaryl” as used herein refers to an unsaturated aromatic cyclic group having from 1 to 14 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur. A heteroaryl group may have a single ring (e.g., pyridyl, furyl) or multiple condensed rings (e.g., indolizinyl, benzothienyl) which condensed rings may or may not be aromatic. Particular heteroaryl groups are 5 to 14-membered rings having 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 5 to 10-membered rings having 1 to 8 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, or 5, 6 or 7-membered rings having 1 to 5 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In one variation, particular heteroaryl groups are monocyclic aromatic 5-, 6- or 7-membered rings having from 1 to 6 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In another variation, particular heteroaryl groups are polycyclic aromatic rings having from 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. A heteroaryl group having more than one ring where at least one ring is non-aromatic may be connected to the parent structure at either an aromatic ring position or at a non-aromatic ring position. In one variation, a heteroaryl group having more than one ring where at least one ring is non-aromatic is connected to the parent structure at an aromatic ring position. A heteroaryl group may be connected to the parent structure at a ring carbon atom or a ring heteroatom. “Heteroarylene” as used herein refers to the same residues as heteroaryl, but having bivalency. “Heterocycle”, “heterocyclic”, or “heterocyclyl” as used herein refers to a saturated or an unsaturated non-aromatic cyclic group having a single ring or multiple condensed rings, and having from 1 to 14 annular carbon atoms and from 1 to 6 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like. A heterocycle comprising more than one ring may be fused, bridged or spiro, or any combination thereof. In fused ring systems, one or more of the fused rings can be cycloalkyl or aryl, but excludes heteroaryl groups. The heterocyclyl group may be optionally substituted independently with one or more substituents described herein. Particular heterocyclyl groups are 3 to 14-membered rings having 1 to 13 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 12-membered rings having 1 to 11 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 10-membered rings having 1 to 9 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 8-membered rings having 1 to 7 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, or 3 to 6-membered rings having 1 to 5 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In one variation, heterocyclyl includes monocyclic 3-, 4-, 5-, 6- or 7-membered rings having from 1 to 2, 1 to 3, 1 to 4, 1 to 5, or 1 to 6 annular carbon atoms and 1 to 2, 1 to 3, or 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In another variation, heterocyclyl includes polycyclic non-aromatic rings having from 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. “Heterocyclylene” as used herein refers to the same residues as heterocyclyl, but having bivalency. “Halo” or “halogen” refers to elements of the Group 17 series having atomic number 9 to 85. Preferred halo groups include the radicals of fluorine, chlorine, bromine and iodine. Where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached, e.g., dihaloaryl, dihaloalkyl, trihaloaryl etc. refer to aryl and alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be but are not necessarily the same halogen; thus 4-chloro-3-fluorophenyl is within the scope of dihaloaryl. An alkyl group in which each hydrogen is replaced with a halo group is referred to as a “perhaloalkyl.” A preferred perhaloalkyl group is trifluoromethyl (—CF3). Similarly, “perhaloalkoxy” refers to an alkoxy group in which a halogen takes the place of each H in the hydrocarbon making up the alkyl moiety of the alkoxy group. An example of a perhaloalkoxy group is trifluoromethoxy (—OCF3). “Carbonyl” refers to the group C═O. “Thiocarbonyl” refers to the group C═S. “Oxo” refers to the moiety ═O. “D” refers to deuterium (2H). “Optionally substituted” unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 1, 2, 3, 4 or 5) of the substituents listed for that group in which the substituents may be the same of different. In one embodiment, an optionally substituted group has one substituent. In another embodiment, an optionally substituted group has two substituents. In another embodiment, an optionally substituted group has three substituents. In another embodiment, an optionally substituted group has four substituents. In some embodiments, an optionally substituted group has 1 to 2, 1 to 3, 1 to 4, 1 to 5, 2 to 3, 2 to 4, or 2 to 5 substituents. In one embodiment, an optionally substituted group is unsubstituted. Unless clearly indicated otherwise, “an individual” as used herein intends a mammal, including but not limited to a primate, human, bovine, horse, feline, canine, or rodent. In one variation, the individual is a human. As used herein, “treatment” or “treating” is an approach for obtaining beneficial or desired results including clinical results. Beneficial or desired results include, but are not limited to, one or more of the following: decreasing one more symptoms resulting from the disease, diminishing the extent of the disease, stabilizing the disease (e.g., preventing or delaying the worsening of the disease), preventing or delaying the spread of the disease, delaying the occurrence or recurrence of the disease, delay or slowing the progression of the disease, ameliorating the disease state, providing a remission (whether partial or total) of the disease, decreasing the dose of one or more other medications required to treat the disease, enhancing effect of another medication, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival. Also encompassed by “treatment” is a reduction of pathological consequence of fibrosis. The methods of the invention contemplate any one or more of these aspects of treatment. As used herein, the term “effective amount” intends such amount of a compound of the invention which should be effective in a given therapeutic form. As is understood in the art, an effective amount may be in one or more doses, i.e., a single dose or multiple doses may be required to achieve the desired treatment endpoint. An effective amount may be considered in the context of administering one or more therapeutic agents (e.g., a compound, or pharmaceutically acceptable salt thereof), and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable or beneficial result may be or is achieved. Suitable doses of any of the co-administered compounds may optionally be lowered due to the combined action (e.g., additive or synergistic effects) of the compounds. A “therapeutically effective amount” refers to an amount of a compound or salt thereof sufficient to produce a desired therapeutic outcome. As used herein, “unit dosage form” refers to physically discrete units, suitable as unit dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Unit dosage forms may contain a single or a combination therapy. As used herein, the term “controlled release” refers to a drug-containing formulation or fraction thereof in which release of the drug is not immediate, i.e., with a “controlled release” formulation, administration does not result in immediate release of the drug into an absorption pool. The term encompasses depot formulations designed to gradually release the drug compound over an extended period of time. Controlled release formulations can include a wide variety of drug delivery systems, generally involving mixing the drug compound with carriers, polymers or other compounds having the desired release characteristics (e.g., pH-dependent or non-pH-dependent solubility, different degrees of water solubility, and the like) and formulating the mixture according to the desired route of delivery (e.g., coated capsules, implantable reservoirs, injectable solutions containing biodegradable capsules, and the like). As used herein, by “pharmaceutically acceptable” or “pharmacologically acceptable” is meant a material that is not biologically or otherwise undesirable, e.g., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained. Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration. “Pharmaceutically acceptable salts” are those salts which retain at least some of the biological activity of the free (non-salt) compound and which can be administered as drugs or pharmaceuticals to an individual. Such salts, for example, include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, oxalic acid, propionic acid, succinic acid, maleic acid, tartaric acid and the like; (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base. Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine and the like. Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. Pharmaceutically acceptable salts can be prepared in situ in the manufacturing process, or by separately reacting a purified compound of the invention in its free acid or base form with a suitable organic or inorganic base or acid, respectively, and isolating the salt thus formed during subsequent purification. The term “excipient” as used herein means an inert or inactive substance that may be used in the production of a drug or pharmaceutical, such as a tablet containing a compound of the invention as an active ingredient. Various substances may be embraced by the term excipient, including without limitation any substance used as a binder, disintegrant, coating, compression/encapsulation aid, cream or lotion, lubricant, solutions for parenteral administration, materials for chewable tablets, sweetener or flavoring, suspending/gelling agent, or wet granulation agent. Binders include, e.g., carbomers, povidone, xanthan gum, etc.; coatings include, e.g., cellulose acetate phthalate, ethylcellulose, gellan gum, maltodextrin, enteric coatings, etc.; compression/encapsulation aids include, e.g., calcium carbonate, dextrose, fructose dc (dc=“directly compressible”), honey dc, lactose (anhydrate or monohydrate; optionally in combination with aspartame, cellulose, or microcrystalline cellulose), starch dc, sucrose, etc.; disintegrants include, e.g., croscarmellose sodium, gellan gum, sodium starch glycolate, etc.; creams or lotions include, e.g., maltodextrin, carrageenans, etc.; lubricants include, e.g., magnesium stearate, stearic acid, sodium stearyl fumarate, etc.; materials for chewable tablets include, e.g., dextrose, fructose dc, lactose (monohydrate, optionally in combination with aspartame or cellulose), etc.; suspending/gelling agents include, e.g., carrageenan, sodium starch glycolate, xanthan gum, etc.; sweeteners include, e.g., aspartame, dextrose, fructose dc, sorbitol, sucrose dc, etc.; and wet granulation agents include, e.g., calcium carbonate, maltodextrin, microcrystalline cellulose, etc. Unless otherwise stated, “substantially pure” intends a composition that contains no more than 10% impurity, such as a composition comprising less than 9%, 7%, 5%, 3%, 1%, or 0.5% impurity. It is understood that aspects and embodiments described herein as “comprising” include “consisting of” and “consisting essentially of” embodiments. The following abbreviations may be used herein: AcOH for acetic acid; ACN, acetonitrile; anhyd, anhydrous; aq, aqueous; tBoc or BOC, tert-butoxycarbonyl; br, broad (spectral); ° C., degrees Celsius; calcd, calculated; CBZ, benzyloxycarbonyl; compd, compound; concd, concentrated; concn, concentration; δ, NMR chemical shift in ppm downfield of SiMe4; d, day(s); doublet (spectral); DCE, 1,2-dichloroethane; DCM, dichloromethane; DMA, dimethylacetamide; DMAP, 4-(N,N-dimethylamino)pyridine; DME, 1,2-dimethoxyethane; DMF, dimethylformamide; DMSO, dimethyl sulfoxide; EA, ethyl acetate; equiv, equivalent; Et, ethyl; g, gram(s); GC, gas chromatography; h, hour(s); Hz, hertz; IR, infrared; J, NMR coupling constant; K, kelvin(s); L, liter(s); μ, micro; m, multiplet (spectral); milli; M, molar (moles per liter), mega; M+, parent molecular ion; max, maximum; Me, methyl; MHz, megahertz; min, minute(s), minimum; mM, millimolar (millimoles per liter); mol, mole(s); MOM, methoxymethyl; mp, melting point; MS, mass spectrometry; MW, molecular weight; m/z, mass-to-charge ratio; N, normal (equivalents per liter); nm, nanometer(s); NMP, N-methylpyrrolidone; NMR, nuclear magnetic resonance; PE, petroleum ether; Ph, phenyl; ppm, part(s) per million; Pr, propyl; iPr, isopropyl; PSI, pounds per square inch; q, quartet (spectral); redox, reduction-oxidation; rel, relative; Rf; chromatography retention factor; rt, room temperature; s, singlet (spectral), second(s); t, triplet (spectral); TEA, triethylamine; THF, tetrahydrofuran; TLC, thin-layer chromatography; UV, ultraviolet; vis, visible; vol, volume; v/v, ratio of volume per unit volume; wt, and weight; w/w, ratio of weight per unit weight. Compounds Provided is a compound of formula (A): or a salt thereof, wherein:R1is hydrogen;R2is 5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl optionally substituted by R12, 1,2,3,4-tetrahydro-1,8-naphthyridin-2-yl optionally substituted by R12, 6-aminopyridin-2-yl optionally substituted by R12, or (pyridin-2-yl)amino optionally substituted by R12;G is —C(O)R3or R4;R3is —OR3a, —NR3bR3c, C1-C6alkyl optionally substituted by R3d, C3-C12cycloalkyl optionally substituted by R3e, 3- to 12-membered heterocyclyl optionally substituted by R3f, C3-C8cycloalkenyl optionally substituted by R3i;R4is C1-C6alkyl optionally substituted by R4a, C3-C8cycloalkyl optionally substituted by R4b, 3- to 12-membered heterocyclyl optionally substituted by R4c, C6-C14aryl optionally substituted by R4d, or 5- to 10-membered heteroaryl optionally substituted by R4e;R3ais C1-C6alkyl, C3-C8cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 12-membered heterocyclyl, wherein the C1-C6alkyl, C3-C8cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 12-membered heterocyclyl of R3aare independently optionally substituted by R3g;R3band R3care each independently hydrogen, deuterium, C1-C6alkyl, C3-C8cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 12-membered heterocyclyl, wherein the C1-C6alkyl, C3-C8cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 12-membered heterocyclyl of R3band R3care independently optionally substituted by R3h;R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, and R10bare each independently hydrogen, deuterium, or halogen;each R11aand R11bare independently hydrogen, deuterium, or halogen;n is 0, 1, or 2;each R3d, R3e, R3f, R3g, R3h, R3i, R4a, R4b, R4c, R4d, and R4eis independently oxo or R12;each R12is independently C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C8cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14aryl, halogen, deuterium, —CN, —OR13, —SR13, —NR14R15, —NO2, —C═NH(OR13), —C(O)R13, —OC(O)R13, —C(O)OR13, —C(O)NR14R15, —NR13C(O)R14, —NR13C(O)OR14, —NR13C(O)NR14R15, —S(O)R13, —S(O)2R13, —NR13S(O)R14, —NR13S(O)2R14, —S(O)NR14R15, —S(O)2NR14R15, or —P(O)(OR13)(OR14), wherein the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C8cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C6-C14aryl of R12are independently optionally substituted by R12a;each R12ais independently deuterium, halogen, oxo, —OR16, —NR16R17, —C(O)R16, —C(O)OR16, —NR16C(O)OR18, —CN, —S(O)R16, —S(O)2R16, —P(O)(OR16)(OR17), C3-C8cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14aryl, or C1-C6alkyl, wherein the 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14aryl, and C1-C6alkyl of R12aare independently optionally substituted by R12b;each R12bis independently deuterium, oxo, —OH, or halogen;each R13is independently hydrogen, deuterium, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 6-membered heterocyclyl of R13are each independently optionally substituted by R13a;each R13ais independently halogen, deuterium, oxo, —CN, —OR18, —NR19R20, —P(O)(OR19)(OR20), 3- to 12-membered heterocyclyl, or C1-C6alkyl optionally substituted by deuterium, halogen, —OH, or oxo;each R14is independently hydrogen, deuterium, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 6-membered heterocyclyl of R14and R15are independently optionally substituted by deuterium, halogen, oxo, —CN, —OR18, —NR19R20, or C1-C6alkyl optionally substituted by deuterium, halogen, —OH, or oxo;each R15is independently hydrogen, deuterium, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 6-membered heterocyclyl of R14and R15are independently optionally substituted by deuterium, halogen, oxo, —CN, —OR18, —NR19R20, or C1-C6alkyl optionally substituted by deuterium, halogen, —OH, or oxo;or R14and R15are taken together with the atom to which they attached to form a 3- to 6-membered heterocyclyl optionally substituted by deuterium, halogen, oxo, —OR18, —NR19R20, or C1-C6alkyl optionally substituted by deuterium, halogen, oxo, or —OH;each R16is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo;each R17is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo;each R18is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo;each R19is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo; andeach R20is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo;or R19and R20are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by deuterium, halogen, oxo or C1-C6alkyl optionally substituted by deuterium, oxo, or halogen; andR21is hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C3-C6cycloalkyl optionally substituted by deuterium, halogen, or oxo, provided that the compound is other than a compound in Table 1X or a salt thereof. In various embodiments of formula (A), R21is hydrogen or deuterium. R21is C1-C6alkyl optionally substituted by deuterium or halogen, C2-C6alkenyl optionally substituted by deuterium or halogen, or C2-C6alkynyl optionally substituted by deuterium or halogen. R21is C1-C6alkyl optionally substituted by deuterium, C2-C6alkenyl optionally substituted by deuterium, or C2-C6alkynyl optionally substituted by deuterium. R21is C1-C4alkyl, C1-C4alkenyl, or C1-C4alkynyl optionally substituted by deuterium. R21is C1-C2alkyl, C1-C2alkenyl, or C1-C2alkynyl optionally substituted by deuterium. R21is methyl optionally substituted by deuterium. The carbon to which R21is bonded is in the R configuration, or the S configuration. For example, R21is methyl, ethyl, 1-propyl, or 2-propyl, and the carbon to which R21is bonded is in the R configuration. R21is methyl, ethyl, 1-propyl, or 2-propyl, and the carbon to which R21is bonded is in the S configuration. In various embodiments, R21is hydrogen and R3is —OR3a, —NR3bR3c, C1-C6alkyl optionally substituted by R3d, C3-C8cycloalkyl optionally substituted by R3e, 3- to 12-membered heterocyclyl optionally substituted by R3f. In one embodiment, disclosed herein is a compound of formula (I): or a salt thereof, wherein:R1is hydrogen;R2is 5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl optionally substituted by R12, 1,2,3,4-tetrahydro-1,8-naphthyridin-2-yl optionally substituted by R12, 6-aminopyridin-2-yl optionally substituted by R12, or (pyridin-2-yl)amino optionally substituted by R12;G is —C(O)R3or R4;R3is —OR3a, —NR3bR3c, C1-C6alkyl optionally substituted by R3d, C3-C8cycloalkyl optionally substituted by R3e, or 3- to 12-membered heterocyclyl optionally substituted by R3f;R4is C1-C6alkyl optionally substituted by R4a, C3-C8cycloalkyl optionally substituted by R4b, 3- to 12-membered heterocyclyl optionally substituted by R4, C6-C14aryl optionally substituted by R4d, or 5- to 10-membered heteroaryl optionally substituted by R4e;R3ais C1-C6alkyl, C3-C8cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 12-membered heterocyclyl, wherein the C1-C6alkyl, C3-C8cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 12-membered heterocyclyl of R3aare independently optionally substituted by R3g;R3band R3care each independently hydrogen, deuterium, C1-C6alkyl, C3-C8cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 12-membered heterocyclyl, wherein the C1-C6alkyl, C3-C8cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 12-membered heterocyclyl of R3band R3care independently optionally substituted by R3b;R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, and R10bare each independently hydrogen, deuterium, or halogen;each R11aand R11bare independently hydrogen, deuterium, or halogen;n is 0, 1, or 2;each R3d, R3e, R3f, R3g, R3h, R4a, R4b, R4c, R4d, and R4eis independently oxo or R12;each R12is independently C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C8cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14aryl, halogen, deuterium, —CN, —OR13, —SR13, —NR14R15, —NO2, —C═NH(OR13), —C(O)R13, —OC(O)R13, —C(O)OR13, —C(O)NR14R15, —NR13C(O)R14, —NR13C(O)OR14, —NR13C(O)NR14R15, —S(O)R13, —S(O)2R13, —NR13S(O)R14, —NR13S(O)2R14, —S(O)NR14R15, —S(O)2NR14R15, or —P(O)(OR13)(OR14), wherein the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C8cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C6-C14aryl of R12are independently optionally substituted by R12a;each R12ais independently deuterium, halogen, oxo, —OR16, —NR16R17, —C(O)R16, —C(O)OR16, —NR16C(O)OR18, —CN, —S(O)R16, —S(O)2R16, —P(O)(OR16)(OR17), C3-C8cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14aryl, or C1-C6alkyl, wherein the 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14aryl, and C1-C6alkyl of R12aare independently optionally substituted by R12b;each R12bis independently deuterium, oxo, —OH, or halogen;each R13is independently hydrogen, deuterium, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 6-membered heterocyclyl of R3are each independently optionally substituted by R13a;each R13ais independently halogen, deuterium, oxo, —CN, —OR18, —NR19R20, —P(O)(OR19)(OR20), 3- to 12-membered heterocyclyl, or C1-C6alkyl optionally substituted by deuterium, halogen, —OH, or oxo;each R14is independently hydrogen, deuterium, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 6-membered heterocyclyl of R14and R15are independently optionally substituted by deuterium, halogen, oxo, —CN, —OR18, —NR19R20, or C1-C6alkyl optionally substituted by deuterium, halogen, —OH, or oxo;each R15is independently hydrogen, deuterium, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C6-C14aryl, 5- to 10-membered heteroaryl, and 3- to 6-membered heterocyclyl of R14and R15are independently optionally substituted by deuterium, halogen, oxo, —CN, —OR18, —NR19R20, or C1-C6alkyl optionally substituted by deuterium, halogen, —OH, or oxo;or R14and R15are taken together with the atom to which they attached to form a 3- to 6-membered heterocyclyl optionally substituted by deuterium, halogen, oxo, —OR18, —NR19R20, or C1-C6alkyl optionally substituted by deuterium, halogen, oxo, or —OH;each R16is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo;each R17is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo;each R18is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo;each R19is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo; andeach R20is independently hydrogen, deuterium, C1-C6alkyl optionally substituted by deuterium, halogen, or oxo, C2-C6alkenyl optionally substituted by deuterium, halogen, or oxo, or C2-C6alkynyl optionally substituted by deuterium, halogen, or oxo;or R19and R20are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by deuterium, halogen, oxo or C1-C6alkyl optionally substituted by deuterium, oxo, or halogen; provided that the compound is other than a compound in Table 1X or a salt thereof. In one variation is provided a compound of the formula (I), or a salt thereof, wherein the carbon bearing the CO2H and N(R1)G moieties is in the “S” configuration. In another variation is provided a compound of the formula (I), or a salt thereof, wherein the carbon bearing the CO2H and N(R1))G moieties is in the “R” configuration. Mixtures of a compound of the formula (I) are also embraced, including racemic or non-racemic mixtures of a given compound, and mixtures of two or more compounds of different chemical formulae. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In the descriptions herein, it is understood that every description, variation, embodiment or aspect of a moiety may be combined with every description, variation, embodiment or aspect of other moieties the same as if each and every combination of descriptions is specifically and individually listed. For example, every description, variation, embodiment or aspect provided herein with respect to R2of formula (I) may be combined with every description, variation, embodiment or aspect of G the same as if each and every combination were specifically and individually listed. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In some embodiments, the compound is other than a compound in Table 1X and salts thereof. In some embodiments, the compound herein, such as a compound of formula (I), is other than a compound selected from one or more of Compound Nos. 1x-4x in Table 1X. In some embodiments, the compounds of the disclosure, and methods of using the compounds detailed herein, encompass any of the compounds of formula (I), including those listed Table 1X and salts thereof. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In one variation, in any of the embodiments disclosed herein, the compounds can exclude compounds in Table 1X or salts thereof. TABLE 1XNo.StructureName1x(S)-2-(3-benzylureido)-9-(5,6,7,8- tetrahydro-1,8-naphthyridin-2- yl)nonanoic acid2x(S)-2-(((benzyloxy)carbonyl)amino)-9- (5,6,7,8-tetrahydro-1,8-naphthyridin-2- yl)nonanoic acid3x(S)-2-(2-phenylacetamido)-9-(5,6,7,8- tetrahydro-1,8-naphthyridin-2- yl)nonanoic acid4x(S)-2-acetamido-9-(5,6,7,8-tetrahydro- 1,8-naphthyridin-2-yl)nonanoic acid In some embodiments of the compound of formula (I), or a salt thereof, R3is C3-C8cycloalkyl optionally substituted by R3eor 3- to 12-membered heterocyclyl optionally substituted by R3f. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A), where additionally R3is C3-C12cycloalkyl optionally substituted by R3e. Also provided is a compound of formula (I), or a salt thereof, wherein:a. when G is —C(O)R3and R3is C1-C6alkyl optionally substituted by R3d, then:i. R3is C2-C6alkyl optionally substituted by R3d; orii. R3is C1alkyl substituted by 2-5 R3d; oriii. R3is C1alkyl substituted by at least one R3d, which is further substituted by R12a; andb. when G is —C(O)R3and R3is —OR3a, then R3ais unsubstituted C1-C6alkyl; andc. when G is —C(O)R3and R3is —NR3bR3c, then:i. R3band R3care other than hydrogen; orii. at least one of R3bor R3cis unsubstituted C1-C6alkyl. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In some embodiments of formula (I), or formula (A), G is —C(O)R3. In such embodiments, when R3is C1-C6alkyl optionally substituted by R3d, R3is one of: (1) C2-C6alkyl optionally substituted by R3d; (2) C1alkyl substituted by 2 or 3 R3d; or (3) C1alkyl substituted by at least one R3d, the at least one R3dbeing further substituted by at least one R12. Further in such embodiments, when R3is —NR3bR3c, R3band R3care any value described herein for R3band R3cother than hydrogen, or at least one of R3bor R3cis unsubstituted C1-C6alkyl. Also in such embodiments, when R3is —OR3a, R3ais unsubstituted C1-C6alkyl. In some such embodiments of the compound of formula (A) or (I), or a salt thereof: n is 1; R1, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10b, R11a, and R11bare each hydrogen; in the case of formula (A), R21is hydrogen; the carbon to which the depicted —CO2H group is bonded is in the S configuration; and R2is unsubstituted 5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl. In various embodiments of the compound of formula (A) or (I), or a salt thereof: n is 1; R1, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10b, R11a, and R11bare each hydrogen; in the case of formula (A), R21is hydrogen; the carbon to which the depicted —CO2H group is bonded is in the S configuration; R2is unsubstituted 5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl; and G is —C(O)R3. In such embodiments, when R3is C1-C6alkyl optionally substituted by R3d, R3is one of: (1) C2-C6alkyl optionally substituted by R3d; (2) C1alkyl substituted by 2 or 3 R3d; or (3) C1alkyl monosubstituted by a single R3d, wherein when the single R3dis phenyl, R3dis substituted with at least one R12. Further in such embodiments, when R3is —NR3bR3c, one of R3band R3cis C1-C6alkyl, and the other of R3band R3cis H, such that the one of R3band R3cthat is C1-C6alkyl is one of: (1) C2-C6alkyl optionally substituted by R3h; (2) C1alkyl substituted by 0, 2, or 3 R3h; or (3) C1alkyl monosubstituted by a single R3h, wherein when the single R3his phenyl, the single R3his substituted with at least one R12. In some embodiments, the at least one R12asubstituting the single R3his any value described herein for R12aother than deuterium or oxo. Also in such embodiments, when R3is —OR3aand R3ais C1-C6alkyl, R3ais one of: (1) C2-C6alkyl optionally substituted by R3g; (2) C1alkyl substituted by 0, 2, or 3 R3g; or (3) C1alkyl monosubstituted by a single R3g, wherein when the single R3gis phenyl, the single R3gis substituted with at least one R12a. In some embodiments, the at least one R12asubstituting the single R3gis any value described herein for R12aother than deuterium or oxo. In some embodiments of the preceding paragraph, where the single R3dis phenyl, the at least one R12is any value described herein for R12other than one of: methyl; C1-C2alkyl; C1-C3alkyl; C1-C4alkyl; C1-C5alkyl; or C1-C6alkyl. For example, the single R3dmay be phenyl substituted with at least one value described herein for R12other than methyl or ethyl. In several embodiments of the preceding paragraph where the single R3gis phenyl, or the single R3his phenyl, the at least one R12ais any value described herein for R12aother than one of: deuterium and methyl; deuterium and C1-C2alkyl; deuterium and C1-C3alkyl; deuterium and C1-C4alkyl; deuterium and C1-C5alkyl; or deuterium and C1-C6alkyl. For example, the single R3gmay be phenyl substituted by at least one value described for R12aherein other than deuterium, methyl, or ethyl. Further, for example, the single R3hmay be phenyl substituted by at least one value described for R12aherein other than deuterium, methyl, or ethyl. In various embodiments of the compound of formula (A) or (I), or a salt thereof: n is 1; R1, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10b, R11a, and R11bare each hydrogen; in the case of formula (A), R21is hydrogen; the carbon to which the depicted —CO2H group is bonded is in the S configuration; R2is unsubstituted 5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl; and G is —C(O)R3. In such embodiments, when R3is C1-C6alkyl optionally substituted by R3d, R3is one of: (1) C3-C6alkyl optionally substituted by R3d; (2) C1alkyl substituted by 2 R3din which at least one R3dis any value described for R3dherein other than methyl, or C1alkyl substituted by 3 R3d; or (3) C1alkyl monosubstituted by a single R3d, the single R3dbeing any value of R3ddescribed herein other than phenyl optionally substituted with at least one R12. Further in such embodiments, when R3is —NR3bR3c, one of R3band R3cis C1-C6alkyl, and the other of R3band R3cis H, such that the one of R3band R3cthat is C1-C6alkyl is one of: (1) C3-C6alkyl optionally substituted by R3h; (2) C1alkyl substituted by 0, 2, or 3 R3h; or (3) C1alkyl monosubstituted by a single R3h, the single R3hbeing any value described herein for R3hother than phenyl optionally substituted by R12a. Also in such embodiments, when R3is —OR3aand R3ais C1-C6alkyl, R3ais one of: (1) C2-C6alkyl optionally substituted by R3g; (2) C1alkyl substituted by 0, 2, or 3 R3g; or (3) C1alkyl monosubstituted by a single R3g, the single R3gbeing any value described herein for R3hother than phenyl optionally substituted by R12a. In various embodiments of the compound of formula (A) or (I), or a salt thereof: n is 1; R1, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10b, R11a, and R11bare each hydrogen; in the case of formula (A), R21is hydrogen; the carbon to which the depicted —CO2H group is bonded is in the S configuration; R2is unsubstituted 5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl; and G is —C(O)R3. In such embodiments, when R3is C1-C6alkyl optionally substituted by R3d, R3is one of: (1) C3-C6alkyl optionally substituted by R3d; (2) C1alkyl substituted by 2 R3din which at least one R3dis any value described herein for R3dother than methyl, or C1alkyl substituted by 3 R3d; or (3) C1alkyl monosubstituted by a single R3d, the single R3dbeing any value described herein for R3dother than C6-C14aryl optionally substituted with at least one R12. Further in such embodiments, when R3is —NR3bR3c, one of R3band R3cis C1-C6alkyl, and the other of R3band R3cis H, such that the one of R3band R3cthat is C1-C6alkyl is one of: (1) C2-C6alkyl optionally substituted by R3h; (2) C1alkyl substituted by 0, 2, or 3 R3h; or (3) C1alkyl monosubstituted by a single R3h, the single R3hbeing any value described herein for R3hother than C6-C14aryl optionally substituted by R12a. Also in such embodiments, when R3is —OR3aand R3ais C1-C6alkyl, R3ais one of: (1) C2-C6alkyl optionally substituted by R3g; (2) C1alkyl substituted by 0, 2, or 3 R3g; or (3) C1alkyl monosubstituted by a single R3g, the single R3gbeing any value described herein for R3gother than C6-C14aryl optionally substituted by R12a. In some embodiments of the compound of formula (I), or a salt thereof, n is 0. In some embodiments of the compound of formula (I), or a salt thereof, n is 1. In some embodiments of the compound of formula (I), or a salt thereof, n is 2. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In some embodiments of formula (A), or (I), or a salt thereof, R7aand R7bare each hydrogen. R7aand R7bare each deuterium. R7aand R7bare each halogen, e.g., R7aand R7bare each fluorine. R7aand R7bare each fluorine, and R5a, R5b, R6a, R6b, R8a, R8b, R9a, R9b, R10a, R10b, R11a, and R11bare each hydrogen. In some embodiments of formula (I), or a salt thereof, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10b, R11a, and R11bare each hydrogen. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In some embodiments of formula (I), or a salt thereof, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10b, R11a, and R11bare each deuterium. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In some embodiments of the compound of formula (I), or a salt thereof, at least one of R3b, R3c, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10b, R11a, R11b, R12, R12a, R12b, R13, R13a, R14, R15, R16, R17, R18, R19, or R20is deuterium. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A), where additionally R21is deuterium. In some embodiments of formula (I), or a salt thereof, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10bR11a, and R11bare each hydrogen, n is 1, and is represented by the compound of formula (I-A): wherein R2and G are as defined for formula (I). The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A), where additionally R21is hydrogen. For example, the compound is represented by formula (I-B) or (I-C): wherein R2, R3, and R4are as defined for formula (A) or (I). In some embodiments of formula (I-A), or a salt thereof, R5a, R5b, R6a, R6b, R7a, R7b, R8a, R8b, R9a, R9b, R10a, R10bR11a, and R11bare each hydrogen, n is 1, R2is 5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl and is represented by the compound of formula (II): wherein G is as defined for formula (I). The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A), where additionally R21is hydrogen. In some embodiments of the compound of formula (II), wherein G is —C(O)R3, the compound is of the formula (II-A): or a salt thereof, wherein R3is as defined for formula (I). The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In some embodiments of the compound of formula (II-A), the compound is of the formula (II-A-1): or a salt thereof, wherein R3fis as defined for formula (I), and m is 1, 2, or 3. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). In some embodiments of the compound represented by formula (II-A), the group represented by R3is —OR3aoptionally substituted, where possible, by up to four R3g, as described herein for formulas (I) or (A). For example, the compound is represented by any one of formulas (II-A-2) or (II-A-2a). Suitable values of R3gfor the depicted azetidinyl group include C1-C6alkyl, —C(O)OR13, —S(O)2R13, and the like, wherein R13is as described herein for formulas (I) or (A). In several embodiments, the depicted azetidinyl group is substituted with: C1-C3alkyl, —C(O)O—C1-C6alkyl, and/or —S(O)2—C1-C6alkyl. For example, the azetidinyl group is substituted with N-t-Boc. In some embodiments of the compound represented by formula (II-A), the group represented by R3is —NR3bR3c, wherein R3band R3care as described herein for formula (A) or (I) and R3band R3care further optionally substituted, where possible, by up to four R3hrepresenting, where possible, oxo or R12as described herein for formulas (I) or (A), such as embodiments of formulas (II-A-3) or (II-A-3a): Suitable values of R3band R3cinclude C1-C6alkyl and the like. For example, R3band R3ceach represent ethyl. In some embodiments of the compound represented by formula (II-A), the group represented by R3is C1-C6alkyl optionally substituted by R3d, representing, where possible, oxo or R12, of which R12is further optionally substituted, where possible, by or R12a, as described herein for formulas (I) or (A). For example, in various embodiments, the compound is represented by any one of formulas (II-A-4), (II-A-4a), (II-A-4b), or (II-A-4c). Suitable values for the depicted C1-6alkyl group include, e.g., methyl, ethyl, prop-1-yl, prop-2-yl, pentan-3-yl, t-butyl, and the like. Such alkyl groups are optionally substituted by one or more, up to four R12groups such as hydroxy, CH3SO2NH—, NH2SO2—, and the depicted phenyl, pyrrolidinyl, and pyridyl groups. Each of the depicted phenyl, pyrrolidinyl, and pyridyl groups are further substituted, where possible, by up to four R12agroups. For example, in some embodiments, the depicted C1-6alkyl represent methyl substituted with up to three of: OH, phenyl, 2-chlorophenyl, pyrrolidine-2-yl, N-tBOC-pyrrolidin-2-yl, and/or pyridin-4-yl. In several embodiments, the depicted C1-6alkyl represent ethyl, optionally substituted by up to four of: 2-CH3SO2NH, 2-NH2SO2, 2-OH, 1-OH, and/or 1-phenyl. In various embodiments, the depicted C1-6alkyl represent prop-1-yl or prop-2-yl optionally substituted, where possible, with up to four of: 3-methoxy, 3-CH3SO2, 2-(pyridin-3-yl), 2-(tetrahydropyran-4-yl), and/or phenyl. In some embodiments of the compound represented by formula (II-A), the group represented by R3is monocyclic, condensed bicyclic, or bridged bicyclic, and is, for example, C3-C12cycloalkyl, e.g., C3-C8cycloalkyl optionally substituted by R3e, representing, where possible, R12; or C3-C12, e.g., C3-C8cycloalkenyl optionally substituted by R3i, representing, where possible, oxo or R12, as described herein for formulas (I) or (A). Such groups include, for example cyclohexanyl, e.g., cyclohexan-1-yl; cyclohexenyl, e.g., cyclohexen-1-yl; bicyclopentanyl, e.g., bicyclo[1.1.1]pentan-1-yl; bicyclooctanyl, e.g., bicyclo[2.2.2]octan-1-yl; and adamantanyl, e.g., adamantan-1-yl. For example, in various embodiments, the compound is represented by any one of formulas (II-A-5a), (II-A-5b), (II-A-5c), (II-A-5d), or (II-A-5e). Such cycloalkyl or cycloalkenyl groups are substituted with any of the groups encompassed herein for R12, for example, in some embodiments: C1-C6alkyl, C6-C14aryl, —NR14R15, —NR13C(O)R14, and/or —NR13C(O)OR14groups, wherein R13, R14, and R15are as described herein for formulas (I) or (A). In several embodiments, such cycloalkyl or cycloalkenyl groups are substituted with: C1-C3alkyl, —NH2, —NHC(O)—C1-C6alkyl, and/or —NHC(O)O—C1-C6alkyl. In several embodiments, such cycloalkyl or cycloalkenyl groups include cyclohexanyl substituted with, where possible: 1-Me, 4-acetamido, 4-NH2, and/or 4-tBOC-NH. In various embodiments, such cycloalkyl or cycloalkenyl groups include, e.g., cyclohexenyl substituted with 2-phenyl. In some embodiments, such cycloalkyl or cycloalkenyl groups include, e.g., bicyclooctanyl substituted with 4-NH2or 4-tBOC-NH. For each generic structure herein where a point of attachment or a substituent of a multicyclic group, e.g., a bridged bicyclic or condensed bicyclic compound, is indicated generically in the chemical structure by a bond crossing one ring of the multicyclic group, it should be understood that attachment to any suitable ring atom of any ring of the multicyclic group is indicated. For example, in the indanyl group depicted above for II-A-6b, the indanyl group is considered as a cyclopentyl ring condensed with a phenyl ring. In various embodiments, the indanyl group is bonded to the depicted carbonyl group, where possible, to a position selected from the cyclopentyl ring, that is, one of positions 1, 2, or 3 of the indanyl group, or at the phenyl ring, that is, positions 4, 5, 6, or 7 of the indanyl group. Likewise, each R12is bonded, where possible, to a position selected from the cyclopentyl ring, that is, one of positions 1, 2, or 3 of the indanyl group, or at the phenyl ring, that is, positions 4, 5, 6, or 7 of the indanyl group. In some embodiments of the compound represented by formula (II-A), the group represented by R3is monocyclic, condensed bicyclic, or bridged bicyclic, and is, for example, saturated or unsaturated 3- to 12-membered heterocyclyl optionally substituted by R3frepresenting, where possible, R12, as described herein for formulas (I) or (A). For example, in various embodiments, R3represents: azetidinyl, e.g., azetidin-2-yl or azetidin-3-yl; pyrrolidinyl, e.g., pyrrolidin-1-yl, or pyrrolidin-2-yl; tetrahydrofuranyl, e.g., tetrahydrofuran-3-yl; thiazolidinyl, e.g., thiazolidin-4-yl; piperidinyl, e.g., piperidin-1-yl, piperidin-2-yl, piperidin-3-yl, or piperidin-4-yl; tetrahydropyranyl, e.g., tetrahydro-2H-pyran-3-yl or tetrahydro-2H-pyran-4-yl; piperazinyl, e.g., piperazin-1-yl; morpholinyl, e.g., morpholin-3-yl or morpholin-4-yl; dihydropyridinyl, e.g., 1,6-dihydropyridin-3-yl; chromanyl, e.g., chroman-4-yl; azabicyclononanyl, e.g., azabicyclo[3.3.1]nonan-9-yl; oxabicycloheptanyl, e.g., 7-oxabicyclo[2.2.1]heptan-2-yl; or oxabicyclooctanyl, e.g., 8-oxabicyclo[3.2.1]octan-3-yl. For example, in various embodiments, the compound is represented by any one of formulas (II-A-6a), (II-A-6b), (II-A-6c), (II-A-6d), (II-A-6e), (II-A-6f), (II-A-6g), (II-A-6h), (II-A-6i), (II-A-6j), (II-A-6k), (II-A-6l), or (II-A-6m). Such saturated or unsaturated 3- to 12-membered monocyclic heterocyclyl groups, are substituted with up to four of the groups encompassed herein for R12, for example, in some embodiments: C1-C6alkyl optionally substituted by halogen, —OR16, C6-C14aryl, 5- to 10-membered heteroaryl, —NR16R17, or —NR16C(O)OR18; —C(O)R13; —C(O)OR13; —S(O)2R13; cyano; halogen; C6-C14aryl; and/or 5- to 10-membered heteroaryl, wherein R13, R16, R17, and R18are as described herein for formulas (I) or (A). In several embodiments, such saturated or unsaturated 3- to 12-membered monocyclic heterocyclyl groups are substituted with up to four of the groups encompassed herein for R12, for example, in some embodiments: C1-C6alkyl optionally substituted by halogen, —OH, phenyl, 5- to 10-membered heteroaryl, —NH2, or —NH—C(O)O—C1-C6alkyl; —C(O)—C1-C6alkylene-O—C1-C6alkyl; —C(O)—O—C1-C6alkyl; —S(O)2—C1-C6alkyl; —S(O)2—(C6-C14aryl); cyano; halogen; C6-C14aryl; and/or 5- to 10-membered heteroaryl. In some embodiments, an azetidinyl group is substituted, where possible, with, e.g., N-benzyl, 3-methyl, and/or N-tBOC. In several embodiments, a pyrrolidinyl group is substituted, where possible, with up to four of: 2-Me, 3-Me, 5-Me, N-(3-methoxypropanoyl), N-phenyl, N-benzyl, N-pyridinyl, N-(pyridin-3-yl)methyl, N-(pyridin-4-yl)methyl, N-(pyrimidin-2-yl)methyl, N-(pyrimidin-4-yl)methyl, and/or N—SO2Ph. In various embodiments, a tetrahydrofuranyl group is substituted with up to three Me, e.g., 3-Me. In some embodiments, a thiazolidinyl group is substituted with, e.g., 5,5-di-Me and/or N—SO2Ph. In several embodiments, a piperidinyl group is substituted, where possible, with up to four of: 3-Me, 4-Me, 3,3-di-F, 2,6 di-Me, N-(3-methoxypropanoyl), N-acetyl, N-t-butylcarbonyl, N-tBOC, N—CH3SO2, 4-CF3, N-(3-fluoropropyl), N-(3,3,3-trifluoropropyl), 4-(2,2-difluoroethyl), and/or N-(1-methyl pyrazol-4-yl)methyl). In some embodiments, a tetrahydropyranyl group is substituted, where possible, with up to four of: 2-Me, 3-Me, 4-Me, 6-Me, 4-HOCH2—, 4-NH2CH2—, 4-CN, 4-CF3, 4-F, 4-phenyl, and/or 4-tBoc-NHmethyl. In several embodiments, a tetrahydropyran-4-yl group is substituted according to one of: 2-Me, 2,2-di-Me, 2,2,6,6-tetra-Me, 3-Me, 4-Me, 6-Me, 4-HOCH2—, 4-CF3, 4-F, or 4-phenyl. In various embodiments, a morpholino group is substituted with alkyl, e.g., methyl, such as 3,5-dimethyl, and/or tBOC, e.g., N-tBOC. In some embodiments of the compound of formula (II), wherein G is —R4, the compound is of the formula (II-B): or a salt thereof, wherein R4is as defined for formula (I). The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). For example, in various embodiments of the compound represented by formula (II-B), the group represented by R4is C1-C6alkyl, optionally substituted by up to four R4arepresenting, where possible, oxo or R12as described herein for formulas (I) or (A). Suitable alkyl groups include, e.g., methyl or ethyl. For example, the compound is represented by any one of formulas (II-B-1a) or (II-B-1 b). Suitable substituents for such alkyl groups include C6-C14aryl, halogen, 3- to 12-membered heterocyclyl, and 5- to 10-membered heteroaryl. For example, ethyl is substituted with 1-Ph, 1-(tetrahydropyran-4-yl), F, e.g., 2,2,2-tri-F, and/or 1-(pyridin-3-yl). Methyl is substituted with, e.g., Ph, 4-methytetrahydropyran-4-yl, 1H-pyrrolo[2,3-b]pyridin-3-yl, quinolin-4-yl, quinolin-6-yl, or quinolin-8-yl. In some embodiments of the compound represented by formula (II-B), the group represented by R4is C3-C8cycloalkyl, optionally substituted by up to four R4brepresenting, where possible, oxo or R12as described herein for formulas (I) or (A). Suitable cycloalkyl groups include, e.g., cyclopentyl or cyclohexyl. For example, the compound is represented by any one of formulas (II-B-2a) or (II-B-2b). Suitable substituents for such cycloalkyl groups include oxo, C6-C14aryl, halogen, 3- to 12-membered heterocyclyl, and 5- to 10-membered heteroaryl. For example, substituents include methyl, ethyl, Ph, tetrahydropyran-4-yl, F, Cl, pyridine-3-yl, 1H-pyrrolo[2,3-b]pyridin-3-yl, quinolin-4-yl, quinolin-6-yl, or quinolin-8-yl. In several embodiments of the compound represented by formula (II-B), the group represented by R4is 3- to 12-membered heterocyclyl, optionally substituted by up to four R4crepresenting, where possible, oxo or R12as described herein for formulas (I) or (A). For example, the compound is represented by any one of formulas (II-B-3a) or (II-B-3b). In some embodiments of the compound represented by formula (II-B), the group represented by R4is 5- to 10-membered heteroaryl, optionally substituted by up to four R4erepresenting, where possible, oxo or R12as described herein for formulas (I) or (A). Suitable heteroaryl groups include, e.g., pyrazolyl, e.g., pyrazol-4-yl; pyrimidinyl, e.g., pyrimidin-2-yl, pyrimidin-4-yl; quinazolinyl, e.g., quinazolin-4-yl; or pyrazolyl, e.g., pyrazol-4-yl. For example, the compound is represented by any one of formulas (II-B-4a), (II-B-4b), (II-B-4c), (II-B-4d), or (II-B-4e). Suitable substituents for such heteroaryl groups include C1-C6alkyl, —NR14R15, —S(O)2R13, halogen, 3- to 12-membered heterocyclyl, and/or 5- to 10-membered heteroaryl, wherein R13, R14, and R15are as described herein for formulas (I) or (A). In some embodiments, suitable substituents for such heteroaryl groups include C1-C3alkyl, —NH—C1-C6alkyl, —N(C1-C6alkyl)2, —S(O)2—C1-C6alkyl, halogen, 3- to 10-membered heterocyclyl, and/or 5- to 10-membered heteroaryl. For example, substituents for pyrazolyl include, e.g., 1-Me (i.e., N-Me), 3-Me, or 5-Me. Substituents for pyrimidinyl include, e.g., 6-NMe2, 6-SO2propyl, 6-(pyrrolidin-1-yl), 6-(morpholin-1-yl), 4-(4,4-difluoropiperidin-1-yl), 5-(pyridin-3-yl), and/or 5-(pyridin-4-yl). Suitable substituents for quinolinyl include halo, e.g., 8-Br. In various embodiments of the compound represented by formula (II-B), the group represented by R4is C6-C14aryl, optionally substituted by up to four R4drepresenting, where possible, oxo or R12as described herein for formulas (I) or (A). Suitable aryl groups include, e.g., phenyl, indanyl, or indenyl. For example, the compound is represented by any one of formulas (II-B-5a) or (II-B-5b). Suitable substituents for such aryl groups include oxo (e.g., for the saturated indanyl or indenyl carbons), C6-C14aryl, halogen, 3- to 12-membered heterocyclyl, and 5- to 10-membered heteroaryl. For example, substituents include methyl, ethyl, Ph, tetrahydropyran-4-yl, F, Cl, pyridine-3-yl, 1H-pyrrolo[2,3-b]pyridin-3-yl, quinolin-4-yl, quinolin-6-yl, or quinolin-8-yl. Also provided is a compound of formula (I), or a salt thereof, wherein G is —C(O)R3. In one variation, G is —C(O)R3, wherein R3is C1-C6alkyl substituted by 0-5 R3d(e.g., R3is unsubstituted C4-C5alkyl or C1-C3alkyl substituted by 0-5 R3d). In another variation, G is —C(O)R3, wherein R3is C1-C3alkyl substituted by 1-5 R3d. In another variation, G is —C(O)R3, wherein R3is C1-C3alkyl substituted by 1-5 R3d, wherein at least one of the R3dis —OR13(e.g., R13is hydrogen or C1-C6alkyl). In another variation, G is —C(O)R3, wherein R3is C1-C3alkyl substituted by 1-5 R3d, wherein at least one of the R3dis C6-C14aryl substituted by 0-5 halogen (e.g., R3dis unsubstituted phenyl or phenyl substituted by 1-4 halogen). In another variation, G is —C(O)R3, wherein R3is C1-C3alkyl substituted by 2-5 R3d, wherein at least one R3dis unsubstituted phenyl and at least one R3dis OR13. In another variation, G is —C(O)R3, wherein R3is C1-C3alkyl substituted by 1-5 R3d, wherein at least one of the R3dis 3- to 12-membered heterocyclyl substituted by 0-5 —C(O)OR16(e.g., R3dis pyrrolidinyl substituted by at least one —C(O)OR16). In another variation, G is —C(O)R3, wherein R3is C1-C3alkyl substituted by 1-5 R3d, wherein at least one of the R3dis 3- to 12-membered heterocyclyl substituted by 0-5 —C(O)OR16(e.g., R3dis pyrrolidinyl substituted by at least one —C(O)OR16), wherein R16is C1-C4alkyl. In another variation, G is —C(O)R3, wherein R3is C1-C3alkyl substituted by 1-5 R3d, wherein at least one of the R3dis 5- to 10-membered heteroaryl substituted by 0-5 R12a(e.g., R3dis unsubstituted pyridinyl). In another variation, G is —C(O)R3, wherein R3is C1-C3alkyl substituted by 1-5 R3dwherein at least one of the R3dis —S(O)2R13, —NR13S(O)2R14, or —S(O)2NR14R15. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided is a compound of formula (I), or a salt thereof, wherein G is —C(O)R3and R3is C3-C8cycloalkyl substituted by 0-5 R3e(e.g., R3is cyclohexanyl substituted by 0-5 C1-C3alkyl or R3is bicyclo[1.1.1]pentanyl). The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided in another embodiment is a compound of formula (I), or a salt thereof, wherein G is —C(O)R3and R3is 3- to 12-membered heterocyclyl (such as 4- to 6-membered heterocyclyl, e.g., azetidinyl, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, or morpholinyl), which is independently substituted by 0-5 R3f. In another aspect of the foregoing embodiment, R3is substituted by 1-5 R3f, wherein at least one R3fis C1-C6alkyl substituted by 0-5 moieties selected from the group consisting of halogen, —NR16R17, —NR16C(O)OR18, 5- to 10-membered heteroaryl, and C6-C14aryl, wherein the 5- to 10-membered heteroaryl and C6-C14aryl of R3fare independently substituted by 0-5 R12b. It is understood that in such embodiments wherein R3is substituted by 1-5 R3f, wherein at least one R3fis C1-C6alkyl substituted by 0-5 moieties selected from the group consisting of halogen, —NR16R17, —NR16C(O)OR18, 5- to 10-membered heteroaryl, and C6-C14aryl, when R3fis C1-C6alkyl substituted by 1-5 moieties selected from the group consisting of 5- to 10-membered heteroaryl and C6-C14aryl, such 5- to 10-membered heteroaryl and C6-C14aryl can be further independently substituted by 0-5 R12b. In one aspect of the foregoing embodiment, at least one R3fis C1-C2alkyl substituted by 0-5 fluoro, —NH2, —NHC(O)O-t-butyl, pyridinyl, pyrimidinyl, or phenyl. In another aspect of the foregoing embodiment, R3is substituted by 1-5 R3f, wherein at least one R3fis 5- to 10-membered heteroaryl or C6-C14aryl, each of which is independently substituted by 0-5 R12a. In one aspect of the foregoing embodiment, at least one R3fis unsubstituted 5- to 10-membered heteroaryl or unsubstituted C6-C14aryl. In another aspect of the foregoing embodiment, at least one R3fis 5- to 10-membered heteroaryl or C6-C14aryl, each of which is independently substituted by 1-5 R12a. In another aspect of the foregoing embodiment, at least one R3fis pyridinyl or phenyl, each of which is independently optionally substituted. In another aspect of the foregoing embodiment, at least one R3fis substituted pyridinyl or substituted phenyl. In another aspect of the foregoing embodiment, at least one R3fis unsubstituted pyridinyl or unsubstituted phenyl. In another aspect of the foregoing embodiment, R3is substituted by 1-5 R3f, wherein at least one R3fis —C(O)R13, —C(O)OR13, or —S(O)2R13. In one aspect of the foregoing embodiment, R13is independently C1-C6alkyl substituted by 0-5 —OR18, wherein R18is C1-C6alkyl substituted by 0-5 deuterium, halogen, or oxo. In another aspect of the foregoing embodiment, R3is substituted by two or more R3f, wherein each R3fis independently selected from the group consisting of C1-C6alkyl, —C(O)R13, and —C(O)OR13. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided in another embodiment is a compound of formula (I), or a salt thereof, wherein G is —C(O)R3and R3is —OR3a. In one aspect of the foregoing embodiment, R3ais C1-C6alkyl or 3- to 12-membered heterocyclyl, each of which is independently substituted by 0-5 R3g. In one aspect of the foregoing embodiment, R3ais C1-C4alkyl (e.g., t-butyl) or 4- to 6-membered heterocyclyl (e.g., azetidinyl), each of which is independently substituted by 0-5 R3g. In any of these aspects, in one variation, R3gis optionally substituted C1-C6alkyl or —C(O)OR13, wherein R13is C1-C6alkyl. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided in another embodiment is a compound of formula (I), or a salt thereof, wherein G is —C(O)R3and R3is —NR3bR3c. In one aspect of the foregoing embodiment, R3band R3care independently C1-C6alkyl. In another aspect of the foregoing embodiment, both R3band R3care C2alkyl. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided is a compound of formula (I), or a salt thereof, wherein G is —C(O)R3and R3is selected from the group consisting of: The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided is a compound of formula (A), or (I), or a salt thereof, wherein G is —C(O)R3and R3is selected from the group consisting of Also provided is a compound of formula (A), or (I), or a salt thereof, wherein G is —C(O)R3and R3is selected from the group consisting of all of the preceding structures depicted in this paragraph. Also provided are embodiments in any one or more hydrogen atom(s) in any of the preceding structures depicted in this paragraph is/are enriched, e.g., replaced with deuterium atom(s) or tritium atom(s). For example, in some embodiments, each hydrogen bonded to a ring carbon in the forgoing groups is replaced with a corresponding isotope, e.g., deuterium or tritium. Each hydrogen bonded to an acyclic carbon in the forgoing groups, e.g., methyl or methoxy carbons, is replaced with a corresponding isotope, e.g., deuterium or tritium. Further, for example, the forgoing groups are perdeuterated, in which every hydrogen is replaced with deuterium, or pertritiated, in which every hydrogen is replaced with tritium. In some embodiments, one or more ring carbons in the forgoing groups is/are replaced with13C. For example, in polycyclic rings among the forgoing groups, one or more ring carbons in the ring directly bonded to the rest of the compound is/are replaced with13C. In polycyclic rings among the forgoing groups, one or more ring carbons is/are replaced with13C in the ring that substitutes or is fused to the ring bonded to the rest of the compound. Further, for example, every ring carbon in the forgoing groups are replaced with13C. Also provided is a compound of formula (I), or a salt thereof, wherein G is R4and R4is C1-C6alkyl (such as C1-C2alkyl) substituted by 0-5 R4a; wherein when R4is substituted by 1-5 R4a, at least one R4ais 3- to 12-membered heterocyclyl (such as a 10-membered heterocyclyl, e.g. benzo-1,4-dioxanyl), 5- to 10-membered heteroaryl (such as 9- to 10-membered heteroaryl, e.g., quinolinyl or pyrrolopyridinyl), or C6-C14aryl (such as C6aryl, e.g., phenyl), each of which is independently substituted by 0-5 (e.g., 0 or 1) R12a. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided is a compound of formula (I), or a salt thereof, wherein G is R4and R45- to 10-membered heteroaryl (e.g., pyrimidinyl, such as pyrimidin-4-yl, or pyrimidin-2-yl) substituted by 0-5 (e.g., 0-3) R4e. In one variation, the 5- to 10-membered heteroaryl (e.g., pyrimidinyl, such as pyrimidin-4-yl, or pyrimidin-2-yl) of R4is unsubstituted. In one variation, 5- to 10-membered heteroaryl of R4is substituted by 1-5 R4e. In another variation, the 5- to 10-membered heteroaryl is substituted by 1-5 R4e, wherein at least one R4eis C1-C6alkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, —NR14R15, or —S(O)2R13, wherein the C1-C6alkyl, 3- to 12-membered heterocyclyl, and 5- to 10-membered heteroaryl of R4eare independently substituted by 0-5 R12a. In another variation, the 5- to 10-membered heteroaryl is substituted by 1-5 R4e, wherein at least one R4eis C1-C4alkyl, 5- to 6-membered heterocyclyl, or 6-membered heteroaryl, each of which is independently substituted by 0-5 halogen. In any of these aspects, R4e, in one variation, is methyl, difluoromethyl, trifluoromethyl, t-butyl, pyrrolidinyl, morpholinyl, or optionally substituted piperidinyl. In another variation, the 5- to 10-membered heteroaryl of R4is substituted by 2-5 R4e, wherein at least one R4eis methyl and at least one R4eis trifluoromethyl. In another variation, R4is quinazolinyl or pyrazolopyrimidinyl, each of which is independently substituted by 0-5 R4e(e.g., unsubstituted quinazolinyl, unsubstituted pyrazolopyrimidinyl, quinazolinyl substituted by 1-5 R4e, or pyrazolopyrimidinyl substituted by 1-5 R4e). In another variation, R4is quinazolinyl or pyrazolopyrimidinyl substituted by 1-5 R4e, wherein at least one R4eis C1-C6alkyl or halogen. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). When a moiety is contemplated, it is understood that the moiety can be attached to the rest of the structure at any available position. For example, 2-methylpyridinyl may be attached to the rest of the structure at the 3-, 4-, 5-, or 6-position (i.e., 2-methylpyridin-3-yl, 2-methylpyridin-4-yl, 2-methylpyridin-5-yl, or 2-methylpyridin-6-yl, respectively). Also provided is a compound of formula (I), or a salt thereof, wherein G is R4and R4is selected from the group consisting of: The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided is a compound of formula (A), or (I), or a salt thereof, wherein G is R4and R4is selected from the group consisting of: Also provided is a compound of formula (A), or (I), or a salt thereof, wherein G is R4and R4is selected from the group consisting of all of the preceding structures depicted in this paragraph. Also provided are embodiments in any one or more hydrogen atom(s) in any of the preceding structures depicted in this paragraph is/are enriched, e.g., replaced with deuterium atom(s) or tritium atom(s). For example, in some embodiments, each hydrogen bonded to a ring carbon in the forgoing groups is replaced with a corresponding isotope, e.g., deuterium or tritium. Each hydrogen bonded to an acyclic carbon in the forgoing groups, e.g., methyl or methoxy carbons, is replaced with a corresponding isotope, e.g., deuterium or tritium. Further, for example, the forgoing groups are perdeuterated, in which every hydrogen is replaced with deuterium, or pertritiated, in which every hydrogen is replaced with tritium. In some embodiments, one or more ring carbons in the forgoing groups is/are replaced with13C. For example, in polycyclic rings among the forgoing groups, one or more ring carbons in the ring directly bonded to the rest of the compound is/are replaced with13C. In polycyclic rings among the forgoing groups, one or more ring carbons is/are replaced with13C in the ring that substitutes or is fused to the ring bonded to the rest of the compound. Further, for example, every ring carbon in the forgoing groups is replaced with13C. Also provided is a compound of formula (I), or a salt thereof, wherein G is selected from the group consisting of: In the moieties listed above, R indicates the point of attachment to the N of the parent molecule. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Also provided are embodiments in any one or more hydrogen atom(s) in any of the preceding structures depicted in this paragraph is/are enriched, e.g., replaced with deuterium atom(s) or tritium atom(s). For example, in some embodiments, each hydrogen bonded to a ring carbon in the forgoing groups is replaced with a corresponding isotope, e.g., deuterium or tritium. Each hydrogen bonded to an acyclic carbon in the forgoing groups, e.g., methyl or methoxy carbons, is replaced with a corresponding isotope, e.g., deuterium or tritium. Further, for example, the forgoing groups are perdeuterated, in which every hydrogen is replaced with deuterium, or pertritiated, in which every hydrogen is replaced with tritium. In some embodiments, one or more ring carbons in the forgoing groups is/are replaced with13C. For example, in polycyclic rings among the forgoing groups, one or more ring carbons in the ring directly bonded to the rest of the compound is/are replaced with13C. In polycyclic rings among the forgoing groups, one or more ring carbons is/are replaced with13C in the ring that substitutes or is fused to the ring bonded to the rest of the compound. Further, for example, every ring carbon in the forgoing groups is replaced with13C. Representative compounds are listed in Table 1,FIG.1. Representative compounds are listed in Table 1,FIG.1, for example, in various embodiments, Compound Nos. 1-77, Compound Nos. 78-124, and Compound Nos. 1-124. In some embodiments, provided is a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof (including a mixture of two or more stereoisomers thereof), or a salt thereof. In some embodiments, the compound is a salt of a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof. In some embodiments, provided is a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof (including a mixture of two or more stereoisomers thereof), or a salt thereof. In some embodiments, the compound is a salt of a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof. In some embodiments, provided is a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof (including a mixture of two or more stereoisomers thereof), or a salt thereof. In some embodiments, the compound is a salt of a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof. In one variation, the compound detailed herein is selected from the group consisting of: (2-pivalamido-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(pyridin-2-yl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2-methyl-2-(pyridin-3-yl)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2-ethylbutanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(morpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-phenylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-benzylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2-methyl-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(pyrimidin-2-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(2-(Pyridin-4-yl)acetyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(pyrimidin-4-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(pyridin-3-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(tert-butoxycarbonyl)piperidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2-(2-chlorophenyl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(tert-butoxycarbonyl)-3-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(tert-butoxycarbonyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2-(1-(tert-butoxycarbonyl)pyrrolidin-2-yl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-benzylazetidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(3-methoxypropanoyl)-3-methylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(3-methoxypropanoyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-(methylsulfonyl)butanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2-hydroxy-2-phenylacetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(3-hydroxy-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(3,3-diethylureido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-methoxybutanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(tetrahydrofuran-3-carboxamido)nonanoic acid; 2-((((1-(tert-butoxycarbonyl)-3-methylazetidin-3-yl)oxy)carbonyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-[(1-tert-butoxycarbonylazetidin-3-yl)oxycarbonylamino]-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(tetrahydro-2H-pyran-4-carboxamido)nonanoic acid; 2-(1-acetylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(methylsulfonyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(3-sulfamoylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(methylsulfonyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(3-(methylsulfonamido)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(3-methyltetrahydrofuran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(4-(trifluoromethyl)tetrahydro-2H-pyran-4-carboxamido)nonanoic acid; 2-(8-oxabicyclo[3.2.1]octane-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-methylcyclohexanecarboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(bicyclo[1.1.1]pentane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(chromane-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(3-methyltetrahydro-2H-pyran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-(((tert-butoxycarbonyl)amino)methyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-Phenyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-(aminomethyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-fluorotetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((6-(propylsulfonyl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((1-methyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((5-(pyridin-3-yl)pyrimidin-2-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((6-(difluoromethyl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((5-(pyridin-4-yl)pyrimidin-2-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((6-morpholinopyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((6-(pyrrolidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((1-methyl-1H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((1H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((6-(4,4-difluoropiperidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((6-(dimethylamino)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(pyrimidin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((8-bromoquinazolin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(quinazolin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(((2,3-dihydrobenzo[b][1,4]dioxin-6-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(benzylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((quinolin-4-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((quinolin-6-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((quinolin-8-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((1-phenylethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(((1H-pyrrolo[2,3-b]pyridin-3-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-(tert-butoxycarbonyl)morpholine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(7-oxabicyclo[2.2.1]heptane-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2-methyl-2-(tetrahydro-2H-pyran-4-yl)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(tert-butoxycarbonyl)-3,3-difluoropiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2,6-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(tert-butoxycarbonyl)-4-(trifluoromethyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(2,2,6,6-tetramethyltetrahydro-2H-pyran-4-carboxamido)nonanoic acid; 2-(1-(tert-butoxycarbonyl)-4-(2,2-difluoroethyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(3,4,5,6-tetrahydro-[1,1′-biphenyl]-2-ylcarboxamido)nonanoic acid; 2-(2-(pyridin-4-yl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(phenylsulfonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(((4-methyltetrahydro-2H-pyran-4-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-((1-(pyridin-3-yl)ethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(((1,3,5-trimethyl-1H-pyrazol-4-yl)methyl)amino)nonanoic acid; 2-(2,6-Dimethylpiperidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(2,5-dimethylpyrrolidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(3,5-dimethylmorpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(2,4,6-trimethylpiperazine-1-carboxamido)nonanoic acid; 2-(3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(3-acetyl-3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-methyl-1-((1-methyl-1H-pyrazol-4-yl)methyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-((tert-butoxycarbonyl)amino)bicyclo[2.2.2]octane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(adamantane-1-carbonylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-((tert-butoxycarbonyl)amino)-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-amino-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-aminobicyclo[2.2.2]octane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-acetamido-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(5,5-dimethyl-3-(phenylsulfonyl)thiazolidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-methyl-1-(3,3,3-trifluoropropyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-[(1-acetyl-4-methyl-piperidine-4-carbonyl)amino]-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-methyl-1-pivaloylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(1-(3-fluoropropyl)-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 2-(4-(hydroxymethyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 5,5-difluoro-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 5,5-difluoro-2-(quinazolin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; 9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-[[2,2,2-trifluoro-1-tetrahydropyran-4-yl-ethyl]amino]nonanoic acid; and 2-(4-cyanotetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; or a salt thereof. In one variation, the compound detailed herein is selected from the group consisting of: (S)-2-pivalamido-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-(pyridin-2-yl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-1-(pyridin-2-yl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(2-methyl-2-(pyridin-3-yl)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(2-ethylbutanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(morpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-(2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-phenylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-benzylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(2-methyl-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-(pyrimidin-2-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-(2-(Pyridin-4-yl)acetyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-(pyrimidin-4-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-(pyridin-3-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-(tert-butoxycarbonyl)piperidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(2-(2-chlorophenyl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((3R,4R)-1-(tert-butoxycarbonyl)-3-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(1-(tert-butoxycarbonyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(2-((S)-1-(tert-butoxycarbonyl)pyrrolidin-2-yl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-benzylazetidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((2S,3S)-1-(3-methoxypropanoyl)-3-methylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-1-(3-methoxypropanoyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-(methylsulfonyl)butanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-2-hydroxy-2-phenylacetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-2-hydroxy-2-phenylacetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-3-hydroxy-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-3-hydroxy-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(3,3-diethylureido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-methoxybutanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-((R)-tetrahydrofuran-3-carboxamido)nonanoic acid; (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-((S)-tetrahydrofuran-3-carboxamido)nonanoic acid; (S)-2-((((1-(tert-butoxycarbonyl)-3-methylazetidin-3-yl)oxy)carbonyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-[(1-tert-butoxycarbonylazetidin-3-yl)oxycarbonylamino]-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(tetrahydro-2H-pyran-4-carboxamido)nonanoic acid; (S)-2-(1-acetylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-1-(methylsulfonyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-(methylsulfonyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(3-sulfamoylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(1-(methylsulfonyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(3-(methylsulfonamido)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-3-methyltetrahydrofuran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-3-methyltetrahydrofuran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(4-(trifluoromethyl)tetrahydro-2H-pyran-4-carboxamido)nonanoic acid; (S)-2-((1R,3s,5S)-8-oxabicyclo[3.2.1]octane-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((1R,3r,5S)-8-oxabicyclo[3.2.1]octane-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(1-methylcyclohexanecarboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(bicyclo[1.1.1]pentane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-chromane-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-chromane-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-3-methyltetrahydro-2H-pyran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-3-methyltetrahydro-2H-pyran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-(((tert-butoxycarbonyl)amino)methyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-Phenyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-(aminomethyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (R)-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-fluorotetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((6-(propylsulfonyl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((1-methyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((5-(pyridin-3-yl)pyrimidin-2-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((6-(difluoromethyl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((5-(pyridin-4-yl)pyrimidin-2-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((6-morpholinopyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((6-(pyrrolidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((1-methyl-1H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((1H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((6-(4,4-difluoropiperidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((6-(dimethylamino)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(pyrimidin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((8-bromoquinazolin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(quinazolin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(((2,3-dihydrobenzo[b][1,4]dioxin-6-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(benzylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((quinolin-4-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((quinolin-6-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((quinolin-8-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(((R)-1-phenylethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(((S)-1-phenylethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(((1H-pyrrolo[2,3-b]pyridin-3-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-4-(tert-butoxycarbonyl)morpholine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-(7-oxabicyclo[2.2.1]heptane-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-((2R)-7-oxabicyclo[2.2.1]heptane-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-((2S)-7-oxabicyclo[2.2.1]heptane-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(2-methyl-2-(tetrahydro-2H-pyran-4-yl)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-(1-(tert-butoxycarbonyl)-3,3-difluoropiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-((2R,6S)-2,6-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((R)-2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(1-(tert-butoxycarbonyl)-4-(trifluoromethyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(2,2,6,6-tetramethyltetrahydro-2H-pyran-4-carboxamido)nonanoic acid; (S)-2-(1-(tert-butoxycarbonyl)-4-(2,2-difluoroethyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(3,4,5,6-tetrahydro-[1,1′-biphenyl]-2-ylcarboxamido)nonanoic acid; (S)-2-(2-(pyridin-4-yl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-1-(phenylsulfonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(((4-methyltetrahydro-2H-pyran-4-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(((R)-1-(pyridin-3-yl)ethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(((S)-1-(pyridin-3-yl)ethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(((1,3,5-trimethyl-1H-pyrazol-4-yl)methyl)amino)nonanoic acid; (S)-2-((2S,6R)-2,6-Dimethylpiperidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((2S,5R)-2,5-dimethylpyrrolidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((2R,5R)-2,5-dimethylpyrrolidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((3R,5R)-3,5-dimethylmorpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((3R,5S)-3,5-dimethylmorpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-((2R,6S)-2,4,6-trimethylpiperazine-1-carboxamido)nonanoic acid; (2S)-2-(3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((1R,5S,9S)-3-acetyl-3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((1R,5S,9R)-3-acetyl-3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-methyl-1-((1-methyl-1H-pyrazol-4-yl)methyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-((tert-butoxycarbonyl)amino)bicyclo[2.2.2]octane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-(adamantane-1-carbonylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-((tert-butoxycarbonyl)amino)-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-amino-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-aminobicyclo[2.2.2]octane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-acetamido-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-((S)-5,5-dimethyl-3-(phenylsulfonyl)thiazolidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (R)-2-((S)-1-(phenylsulfonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-methyl-1-(3,3,3-trifluoropropyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-2-[(1-acetyl-4-methyl-piperidine-4-carbonyl)amino]-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-methyl-1-pivaloylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(1-(3-fluoropropyl)-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-2-(4-(hydroxymethyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-5,5-difluoro-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (R)-5,5-difluoro-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (S)-5,5-difluoro-2-(quinazolin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; (2S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-[[(1 S)-2,2,2-trifluoro-1-tetrahydropyran-4-yl-ethyl]amino]nonanoic acid; (2S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-[[(1R)-2,2,2-trifluoro-1-tetrahydropyran-4-yl-ethyl]amino]nonanoic acid; and (S)-2-(4-cyanotetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid; or a salt thereof. In some embodiments, a composition, such as a pharmaceutical composition, is provided wherein the composition comprises a compound selected from the group consisting of one or more of Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof (including a mixture of two or more stereoisomers thereof), or a salt thereof. In some embodiments, the composition comprises a compound selected from the group consisting of a salt of one or more of Compound Nos 1-77. In one aspect, the composition is a pharmaceutical composition that further comprises a pharmaceutically acceptable carrier. In some embodiments, a composition, such as a pharmaceutical composition, is provided wherein the composition comprises a compound selected from the group consisting of one or more of Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof (including a mixture of two or more stereoisomers thereof), or a salt thereof. In some embodiments, the composition comprises a compound selected from the group consisting of a salt of one or more of Compound Nos. 1-77,FIG.1. In some embodiments, a composition, such as a pharmaceutical composition, is provided wherein the composition comprises a compound selected from the group consisting of one or more of Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof (including a mixture of two or more stereoisomers thereof), or a salt thereof. In some embodiments, the composition comprises a compound selected from the group consisting of a salt of one or more of Compound Nos. 1-124 in Table 1,FIG.1. In one aspect, the composition is a pharmaceutical composition that further comprises a pharmaceutically acceptable carrier. The invention also includes all salts of compounds referred to herein, such as pharmaceutically acceptable salts. The invention also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms, and any tautomers or other forms of the compounds described. Unless stereochemistry is explicitly indicated in a chemical structure or name, the structure or name is intended to embrace all possible stereoisomers of a compound depicted. In addition, where a specific stereochemical form is depicted, it is understood that other stereochemical forms are also described and embraced by the invention. All forms of the compounds are also embraced by the invention, such as crystalline or non-crystalline forms of the compounds. It is also understood that prodrugs, solvates and metabolites of the compounds are embraced by this disclosure. Compositions comprising a compound of the invention are also intended, such as a composition of substantially pure compound, including a specific stereochemical form thereof. Compositions comprising a mixture of compounds of the invention in any ratio are also embraced by the invention, including mixtures of two or more stereochemical forms of a compound of the invention in any ratio, such that racemic, non-racemic, enantioenriched and scalemic mixtures of a compound are embraced. Where one or more tertiary amine moiety is present in the compound, the N-oxides are also provided and described. A chemical structure which can be depicted as different tautomers is considered aromatic if either tautomer would be considered aromatic. For example, the structure pyridin-2(1H)-one, is considered aromatic due to its tautomer 2-hydroxypyridine, Compounds described herein are αVβ6integrin inhibitors. In some instances, it is desirable for the compound to inhibit other integrins in addition to αVβ6integrin. In some embodiments, the compound inhibits αVβ6integrin and one or more of αVβ1, αVβ3, αVβ5, α2β1, α3β1, α6β1integrin, α7β1and αVβ1. In some embodiments, the compound inhibits αVβ6integrin and αVβ1integrin. In some embodiments, the compound inhibits αVβ6integrin, αVβ3integrin and αVβ5integrin. In some embodiments, the compound inhibits αVβ6integrin and α2β1integrin. In some embodiments, the compound inhibits αVβ6integrin, α2β1integrin and α3β1integrin. In some embodiments, the compound inhibits αVβ6integrin and α6β1integrin. In some embodiments, the compound inhibits αVβ6integrin and α7β1integrin. In some embodiments, the compound inhibits αVβ6integrin and α11β1integrin. In some instances, it is desirable to avoid inhibition of other integrins. In some embodiments, the compound is a selective αVβ6integrin inhibitor. In some embodiments, the compound does not inhibit substantially α4β1, αVβ8and/or α2β3integrin. In some embodiments, the compound inhibits αVβ6integrin but does not inhibit substantially α4β1integrin. In some embodiments, the compound inhibits αVβ6integrin but does not inhibit substantially αVβ8integrin. In some embodiments, the compound inhibits αVβ6integrin but does not inhibit substantially α2β3integrin. In some embodiments, the compound inhibits αVβ6integrin but does not inhibit substantially the αVβ8integrin and the α4β1integrin. The invention also intends isotopically-labeled and/or isotopically-enriched forms of compounds described herein. The compounds herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. In some embodiments, the compound is isotopically-labeled, such as an isotopically-labeled compound of the formula (I) or variations thereof described herein, where one or more atoms are replaced by an isotope of the same element. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, chlorine, such as2H,3H,11C,13C,14C,13N,15O,17O,32P,35S,18F,36Cl. Incorporation of heavier isotopes such as deuterium (2H or D) can afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, or reduced dosage requirements and, hence may be preferred in some instances. The description above of embodiments for formula (I) also apply equally to formula (A) to provide the corresponding embodiments of formula (A). Isotopically-labeled compounds of the present invention can generally be prepared by standard methods and techniques known to those skilled in the art or by procedures similar to those described in the accompanying Examples substituting appropriate isotopically-labeled reagents in place of the corresponding non-labeled reagent. The invention also includes any or all metabolites of any of the compounds described. The metabolites may include any chemical species generated by a biotransformation of any of the compounds described, such as intermediates and products of metabolism of the compound. Articles of manufacture comprising a compound of the invention, or a salt or solvate thereof, in a suitable container are provided. The container may be a vial, jar, ampoule, preloaded syringe, i.v. bag, and the like. Preferably, the compounds detailed herein are orally bioavailable. However, the compounds may also be formulated for parenteral (e.g., intravenous) administration. One or several compounds described herein can be used in the preparation of a medicament by combining the compound or compounds as an active ingredient with a pharmacologically acceptable carrier, which are known in the art. Depending on the therapeutic form of the medication, the carrier may be in various forms. General Synthetic Methods The compounds of the invention may be prepared by a number of processes as generally described below and more specifically in the Examples hereinafter (such as the Schemes provides in the Examples below). In the following process descriptions, the symbols when used in the formulae depicted are to be understood to represent those groups described above in relation to the formulae herein. Where it is desired to obtain a particular enantiomer of a compound, this may be accomplished from a corresponding mixture of enantiomers using any suitable conventional procedure for separating or resolving enantiomers. Thus, for example, diastereomeric derivatives may be produced by reaction of a mixture of enantiomers, e.g., a racemate, and an appropriate chiral compound. The diastereomers may then be separated by any convenient means, for example by crystallization, and the desired enantiomer recovered. In another resolution process, a racemate may be separated using chiral High Performance Liquid Chromatography. Alternatively, if desired a particular enantiomer may be obtained by using an appropriate chiral intermediate in one of the processes described. Chromatography, recrystallization and other conventional separation procedures may also be used with intermediates or final products where it is desired to obtain a particular isomer of a compound or to otherwise purify a product of a reaction. Solvates and/or polymorphs of a compound provided herein or a pharmaceutically acceptable salt thereof are also contemplated. Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are often formed during the process of crystallization. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and/or solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate. Compounds provided herein may be prepared according to Schemes, Procedures, and Examples. Reaction conditions for the transformations of Schemes listed below are provided in the Procedures that follow. The final product depicted below can be prepared according to Scheme A, wherein Rxis a carboxylic protecting group and R is R3as defined for formula (I). The final product depicted below can be prepared according to Scheme B, wherein Rxis a carboxylic protecting group and R is R4as defined for formula (I). The final product depicted below can be prepared according to Scheme C, wherein Rxis a carboxylic protecting group and Y refers to the portion of the molecule that links the —C(O)N(H)— portion of the molecule with the remainder of the R3moiety. The final product depicted below can be prepared according to Scheme D, wherein X is a halide and Rxis a carboxylic protecting group. It is understood the ring bearing the Het description can be any heteroaromatic ring. The final product depicted below can be prepared according to Scheme E, wherein Y refers to the portion of the molecule that links the —C(O)N(H)— portion of the molecule with the remainder of the R3moiety and Rxis a carboxylic protecting group. The final product depicted below can be prepared according to Scheme F, wherein X is a halide, Rxis a carboxylic protecting group, and R is R4eas defined for formula (I), or any applicable variations detailed herein. It is understood the ring bearing the Het description can be any heteroaromatic ring. The final product depicted below can be prepared according to Scheme G, wherein Rxis a carboxylic protecting group and R is R4aas defined for formula (I), or any applicable variations detailed herein. The final product depicted below can be prepared according to Scheme H, wherein Rxis a carboxylic protecting group and R is R4aas defined for formula (I), or any applicable variations detailed herein. The final product depicted below can be prepared according to Scheme I, wherein Rxis a carboxylic protecting group and R is R3as defined for formula (I), or any applicable variations detailed herein. The final product depicted below can be prepared according to Scheme J. The final product depicted below can be prepared according to Scheme K. It is understood that the Schemes above may be modified to arrive at various compounds of the invention by selection of appropriate reagents and starting materials. For a general description of protecting groups and their use, see P. G. M. Wuts and T. W. Greene, Greene's Protective Groups in Organic Synthesis 4thedition, Wiley-Interscience, New York, 2006. Additional methods of preparing compounds according to formula (I), and salts thereof, are provided in the Examples. As a skilled artisan would recognize, the methods of preparation taught herein may be adapted to provide additional compounds within the scope of formula (I), for example, by selecting starting materials which would provide a desired compound. The syntheses of the products depicted above in Schemes A-K for formula (I) can also be carried out for formula (A). Pharmaceutical Compositions and Formulations Pharmaceutical compositions of any of the compounds detailed herein, including compounds of the formula (I), (I-A), (II), (II-A), (II-A-1), and (II-B), or a salt thereof, or any of compounds of Table 1,FIG.1, or a salt thereof, or mixtures thereof, are embraced by this invention. Pharmaceutical compositions of any of the compounds detailed herein, including compounds of the formula (A), (1), (I-A), (II), (II-A), (II-A-1, 2, 3, or 4), (II-A-1a), (II-A-2a), (II-A-3a), (II-A-4a, 4b, or 4c), (II-A-5a, 5b, 5c, 5d, or 5e), (II-A-6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6l, or 6m), (II-B), (II-B-1a or 1b), (II-B-2a or 2b), (II-B-3a or 3b), (II-B-4a, 4b, 4c, 4d, or 4e), or (II-B-5a or 5b), or a salt thereof, or any of compounds of Table 1,FIG.1, or a salt thereof, or mixtures thereof, are embraced by this invention. Thus, the invention includes pharmaceutical compositions comprising a compound of the invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient. In one aspect, the pharmaceutically acceptable salt is an acid addition salt, such as a salt formed with an inorganic or organic acid. Pharmaceutical compositions according to the invention may take a form suitable for oral, buccal, parenteral, nasal, topical or rectal administration or a form suitable for administration by inhalation. In one embodiment, the pharmaceutical composition is a composition for controlled release of any of the compounds detailed herein. A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. In one embodiment, compositions may have no more than 35% impurity, wherein the impurity denotes a compound other than the compound comprising the majority of the composition or a salt thereof, for example, a composition of a compound selected from a compound of Table 1,FIG.1, may contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound of Table 1,FIG.1, or a salt thereof. In one embodiment, compositions may contain no more than 25% impurity. In one embodiment, compositions may contains no more than 20% impurity. In still further embodiments, compositions comprising a compound as detailed herein or a salt thereof are provided as compositions of substantially pure compounds. “Substantially pure” compositions comprise no more than 10% impurity, such as a composition comprising less than 9%, 7%, 5%, 3%, 1%, or 0.5% impurity. In some embodiments, a composition containing a compound as detailed herein or a salt thereof is in substantially pure form. In still another variation, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 10% impurity. In a further variation, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 9% impurity. In a further variation, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 7% impurity. In a further variation, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 5% impurity. In another variation, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 3% impurity. In still another variation, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 1% impurity. In a further variation, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 0.5% impurity. In yet other variations, a composition of substantially pure compound means that the composition contains no more than 10% or preferably no more than 5% or more preferably no more than 3% or even more preferably no more than 1% impurity or most preferably no more than 0.5% impurity, which impurity may be the compound in a different stereochemical form. For instance, a composition of substantially pure (S) compound means that the composition contains no more than 10% or no more than 5% or no more than 3% or no more than 1% or no more than 0.5% of the (R) form of the compound. In one variation, the compounds herein are synthetic compounds prepared for administration to an individual such as a human. In another variation, compositions are provided containing a compound in substantially pure form. In another variation, the invention embraces pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier or excipient. In another variation, methods of administering a compound are provided. The purified forms, pharmaceutical compositions and methods of administering the compounds are suitable for any compound or form thereof detailed herein. A compound detailed herein or salt thereof may be formulated for any available delivery route, including an oral, mucosal (e.g., nasal, sublingual, vaginal, buccal or rectal), parenteral (e.g., intramuscular, subcutaneous or intravenous), topical or transdermal delivery form. A compound or salt thereof may be formulated with suitable carriers to provide delivery forms that include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices), pastes, powders, dressings, creams, solutions, patches, aerosols (e.g., nasal spray or inhalers), gels, suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions or water-in-oil liquid emulsions), solutions and elixirs. One or several compounds described herein or a salt thereof can be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the compound or compounds, or a salt thereof, as an active ingredient with a pharmaceutically acceptable carrier, such as those mentioned above. Depending on the therapeutic form of the system (e.g., transdermal patch vs. oral tablet), the carrier may be in various forms. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants. Formulations comprising the compound may also contain other substances which have valuable therapeutic properties. Pharmaceutical formulations may be prepared by known pharmaceutical methods. Suitable formulations can be found, e.g., inRemington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, 21sted. (2005), which is incorporated herein by reference. Compounds as described herein may be administered to individuals (e.g., a human) in a form of generally accepted oral compositions, such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions. Examples of carriers, which may be used for the preparation of such compositions, are lactose, corn starch or its derivatives, talc, stearate or its salts, etc. Acceptable carriers for gel capsules with soft shell are, for instance, plant oils, wax, fats, semisolid and liquid polyols, and so on. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants. Any of the compounds described herein can be formulated in a tablet in any dosage form described, for example, a compound as described herein or a pharmaceutically acceptable salt thereof can be formulated as a 10 mg tablet. Compositions comprising a compound provided herein are also described. In one variation, the composition comprises a compound and a pharmaceutically acceptable carrier or excipient. In another variation, a composition of substantially pure compound is provided. In some embodiments, the composition is for use as a human or veterinary medicament. In some embodiments, the composition is for use in a method described herein. In some embodiments, the composition is for use in the treatment of a disease or disorder described herein. Methods of Use Compounds and compositions of the invention, such as a pharmaceutical composition containing a compound of any formula provided herein or a salt thereof and a pharmaceutically acceptable carrier or excipient, may be used in methods of administration and treatment as provided herein. The compounds and compositions may also be used in in vitro methods, such as in vitro methods of administering a compound or composition to cells for screening purposes and/or for conducting quality control assays. In one aspect, provided is a method of treating a fibrotic disease in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound of formula (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1), or (II-B), a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. In one aspect, provided is a method of treating a fibrotic disease in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound of formula (A), or (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1, 2, 3, or 4), (II-A-1a), (II-A-2a), (II-A-3a), (II-A-4a, 4b, or 4c), (II-A-5a, 5b, 5c, 5d, or 5e), (II-A-6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6l, or 6m), (II-B), (II-B-1a or 1 b), (II-B-2a or 2b), (II-B-3a or 3b), (II-B-4a, 4b, 4c, 4d, or 4e), or (II-B-5a or 5b), a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. In one aspect, the individual is a human. The individual, such as human, may be in need of treatment, such as a human who has or is suspected of having a fibrotic disease. In another aspect, provided is a method of delaying the onset and/or development of a fibrotic disease in an individual (such as a human) who is at risk for developing a fibrotic disease. It is appreciated that delayed development may encompass prevention in the event the individual does not develop the fibrotic disease. An individual at risk of developing a fibrotic disease in one aspect has or is suspected of having one or more risk factors for developing a fibrotic disease. Risk factors for fibrotic disease may include an individual's age (e.g., middle-age or older adults), the presence of inflammation, having one or more genetic component associated with development of a fibrotic disease, medical history such as treatment with a drug or procedure believed to be associated with an enhanced susceptibility to fibrosis (e.g., radiology) or a medical condition believed to be associated with fibrosis, a history of smoking, the presence of occupational and/or environmental factors such as exposure to pollutants associated with development of a fibrotic disease. In some embodiments, the individual at risk for developing a fibrotic disease is an individual who has or is suspected of having NAFLD, NASH, CKD, scleroderma, Crohn's Disease, NSIP, PSC, PBC, or is an individual who has had or is suspected of having had a myocardial infarction. In some embodiments, the fibrotic disease is fibrosis of a tissue such as the lung (pulmonary fibrosis), the liver, the skin, the heart (cardiac fibrosis), the kidney (renal fibrosis), or the gastrointestinal tract (gastrointestinal fibrosis). In some embodiments, the fibrotic disease is pulmonary fibrosis (such as IPF), liver fibrosis, skin fibrosis, scleroderma, cardiac fibrosis, renal fibrosis, gastrointestinal fibrosis, primary sclerosing cholangitis, or biliary fibrosis (such as PBC). In some embodiments, the fibrotic disease is a pulmonary fibrosis, e.g., idiopathic pulmonary fibrosis (IPF). In some embodiments, the fibrotic disease is a primary sclerosing cholangitis, or biliary fibrosis. In some embodiments, the fibrotic disease is fibrotic nonspecific interstitial pneumonia (NSIP). In some embodiments, the fibrotic disease is a liver fibrosis, e.g., infectious liver fibrosis (from pathogens such as HCV, HBV or parasites such as schistosomiasis), NASH, alcoholic steatosis induced liver fibrosis, and cirrhosis. In some embodiments, the fibrotic disease is biliary tract fibrosis. In some embodiments, the fibrotic disease is renal fibrosis, e.g., diabetic nephrosclerosis, hypertensive nephrosclerosis, focal segmental glomerulosclerosis (“FSGS”), and acute kidney injury from contrast induced nephropathy. In some embodiments, the fibrotic disease is systemic and local sclerosis or scleroderma, keloids and hypertrophic scars, or post surgical adhesions. In some embodiments, the fibrotic disease is atherosclerosis or restenosis. In some embodiments, the fibrotic disease is a gastrointestinal fibrosis, e.g., Crohn's disease. In some embodiments, the fibrotic disease is cardiac fibrosis, e.g., post myocardial infarction induced fibrosis and inherited cardiomyopathy. In one aspect, provided is a compound of formula (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1), or (II-B), a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, for use in the treatment of a fibrotic disease. In one aspect, provided is a compound of formula (A), or (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1, 2, 3, or 4), (II-A-1a), (II-A-2a), (II-A-3a), (II-A-4a, 4b, or 4c), (II-A-5a, 5b, 5c, 5d, or 5e), (II-A-6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6l, or 6m), (II-B), (II-B-1a or 1b), (II-B-2a or 2b), (II-B-3a or 3b), (II-B-4a, 4b, 4c, 4d, or 4e), or (II-B-5a or 5b), a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, for use in the treatment of a fibrotic disease. Also provided is use of a compound of formula (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1), or (II-B), a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a fibrotic disease. Also provided is use of a compound of formula (A), or (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1, 2, 3, or 4), (II-A-1a), (II-A-2a), (II-A-3a), (II-A-4a, 4b, or 4c), (II-A-5a, 5b, 5c, 5d, or 5e), (II-A-6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6l, or 6m), (II-B), (II-B-1a or 1 b), (II-B-2a or 2b), (II-B-3a or 3b), (II-B-4a, 4b, 4c, 4d, or 4e), or (II-B-5a or 5b), a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a fibrotic disease. In another aspect, provided is a method of inhibiting αVβ6integrin in an individual comprising administering a compound of formula (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1), or (II-B), a stereoisomer thereof, or a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a pharmaceutically acceptable salt thereof. In another aspect, provided is a method of inhibiting αVβ6integrin in an individual comprising administering a compound of formula (A), or (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1, 2, 3, or 4), (II-A-1a), (II-A-2a), (II-A-3a), (II-A-4a, 4b, or 4c), (II-A-5a, 5b, 5c, 5d, or 5e), (II-A-6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6l, or 6m), (II-B), (II-B-1a or 1b), (II-B-2a or 2b), (II-B-3a or 3b), (II-B-4a, 4b, 4c, 4d, or 4e), or (II-B-5a or 5b), a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. Also provided is a method of inhibiting TGFβ activation in a cell comprising administering to the cell a compound of formula (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1), or (II-B), a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. Also provided is a method of inhibiting TGFβ activation in a cell comprising administering to the cell a compound of formula (A), or (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1, 2, 3, or 4), (II-A-1a), (II-A-2a), (II-A-3a), (II-A-4a, 4b, or 4c), (II-A-5a, 5b, 5c, 5d, or 5e), (II-A-6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6l, or 6m), (II-B), (II-B-1a or 1 b), (II-B-2a or 2b), (II-B-3a or 3b), (II-B-4a, 4b, 4c, 4d, or 4e), or (II-B-5a or 5b), a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. Also provided is a method of inhibiting αVβ6integrin in an individual in need thereof, comprising administering to the individual a compound of formula (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1), or (II-B), a compound selected from Compound Nos. 1-77,FIG.1, in Table 1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. Also provided is a method of inhibiting αVβ6integrin in an individual in need thereof, comprising administering to the individual a compound of formula (A), or (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1, 2, 3, or 4), (II-A-1a), (II-A-2a), (II-A-3a), (II-A-4a, 4b, or 4c), (II-A-5a, 5b, 5c, 5d, or 5e), (II-A-6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6l, or 6m), (II-B), (II-B-1a or 1 b), (II-B-2a or 2b), (II-B-3a or 3b), (II-B-4a, 4b, 4c, 4d, or 4e), or (II-B-5a or 5b), a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. In one such method, the compound is a selective αVβ6integrin inhibitor. In another such method, the compound does not inhibit substantially α4β1, αVβ8and/or α2β3integrin. In yet another such method, the compound inhibits αVβ6integrin but does not inhibit substantially α4β1integrin. In still another such method, the compound inhibits αVβ6integrin but does not inhibit substantially αVβ8integrin. In a further such method, the compound inhibits αVβ6integrin but does not inhibit substantially α2β3integrin. In one embodiment is provided a method of inhibiting αVβ6integrin and one or more of αVβ1, αVβ3, αVβ5, α2β1, α3β1, α6β1integrin, α7β1and α11β1in an individual in need thereof. In another embodiment is provided a method of inhibiting αVβ6integrin and αVβ1integrin. In another embodiment is provided a method of inhibiting αVβ6integrin, αVβ3integrin and αVβ5integrin. In another embodiment is provided a method of inhibiting αVβ6integrin and α2β1integrin. In another embodiment is provided a method of inhibiting αVβ6integrin, α2β1integrin and α3β1integrin. In another embodiment is provided a method of inhibiting αVβ6integrin and α6β1integrin. In another embodiment is provided a method of inhibiting αVβ6integrin and α7β1integrin. In another embodiment is provided a method of inhibiting αVβ6integrin and α11β1integrin. In all such embodiments, in one aspect the method of inhibition is for an individual in need thereof, such as an individual who has or is suspected of having a fibrotic disease, and wherein the method comprises administering to the individual a compound of formula (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1), or (II-B), a compound selected from Compound Nos. 1-77 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. In all such embodiments, in one aspect the method of inhibition is for an individual in need thereof, such as an individual who has or is suspected of having a fibrotic disease, and wherein the method comprises administering to the individual a compound of formula (A), or (I), or any variation thereof, e.g., a compound of formula (I-A), (II), (II-A), (II-A-1, 2, 3, or 4), (II-A-1a), (II-A-2a), (II-A-3a), (II-A-4a, 4b, or 4c), (II-A-5a, 5b, 5c, 5d, or 5e), (II-A-6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6l, or 6m), (II-B), (II-B-1a or 1b), (II-B-2a or 2b), (II-B-3a or 3b), (II-B-4a, 4b, 4c, 4d, or 4e), or (II-B-5a or 5b), a compound selected from Compound Nos. 1-124 in Table 1,FIG.1, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. In any of the described methods, in one aspect the individual is a human, such as a human in need of the method. The individual may be a human who has been diagnosed with or is suspected of having a fibrotic disease. The individual may be a human who does not have detectable disease but who has one or more risk factors for developing a fibrotic disease. Kits The invention further provides kits for carrying out the methods of the invention, which comprises one or more compounds described herein, or a salt thereof, or a pharmacological composition comprising a compound described herein. The kits may employ any of the compounds disclosed herein. In one variation, the kit employs a compound described herein or a pharmaceutically acceptable salt thereof. The kits may be used for any one or more of the uses described herein, and, accordingly, may contain instructions for use in the treatment of a fibrotic disease. Kits generally comprise suitable packaging. The kits may comprise one or more containers comprising any compound described herein. Each component (if there is more than one component) can be packaged in separate containers or some components can be combined in one container where cross-reactivity and shelf life permit. One or more components of a kit may be sterile and/or may be contained within sterile packaging. The kits may be in unit dosage forms, bulk packages (e.g., multi-dose packages) or sub-unit doses. For example, kits may be provided that contain sufficient dosages of a compound as disclosed herein (e.g., a therapeutically effective amount) and/or a second pharmaceutically active compound useful for a disease detailed herein (e.g., fibrosis) to provide effective treatment of an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 7 months, 8 months, 9 months, or more. Kits may also include multiple unit doses of the compounds and instructions for use and be packaged in quantities sufficient for storage and use in pharmacies (e.g., hospital pharmacies and compounding pharmacies). The kits may optionally include a set of instructions, generally written instructions, although electronic storage media (e.g., magnetic diskette or optical disk) containing instructions are also acceptable, relating to the use of component(s) of the methods of the present invention. The instructions included with the kit generally include information as to the components and their administration to an individual. Procedures Compounds provided herein may be prepared according to Schemes, as exemplified by the Procedures and Examples. Minor variations in temperatures, concentrations, reaction times, and other parameters can be made when following the Procedures, which do not substantially affect the results of the Procedures. Procedure A To a solution of methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate in DMF was added DIPEA (10 equiv) followed by carboxylic acid (1.1 equiv) and HATU (1.1 equiv). The reaction was allowed to stir at rt while monitoring reaction progress by LCMS. When the starting material had been consumed, the reaction was diluted with 1 N NaOH and extracted with EA, washed with brine, dried over sodium sulfate, and concd. The crude residue was purified by silica gel chromatography to afford the depicted compound. In some embodiment, the R group attached to the amide moiety of the reaction product is R3as defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure B To a solution of methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate in a solvent such as IPA, DMF, or DMSO was added halogenated heteroarene and an excess of amine base such as triethylamine or diisopropylethylamine. The reaction mixture was then heated until completion as determined by LCMS. The reaction mixture was concd or used directly in the next step. Halogenated heterocyclyls can also be used to add a corresponding heterocyclic R group on the amine. In some embodiment, the R group attached to the amine moiety of the reaction product is R4as defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure C To a solution of the depicted ester in an appropriate solvent mixture such as THF/MeOH/H2O or THF/EtOH/H2O was added LiOH (3-5 equiv). The reaction was allowed to stir at rt while monitoring reaction progress by LCMS. Upon completion, the reaction was concd and purified by reverse phase preparative HPLC to afford the depicted carboxylic acid as the TFA salt. In some embodiment, the R group attached to the amine moiety of the starting material and reaction product is R4as defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material is a carboxylic acid protecting group. Procedure D To a solution of the depicted halogenated heterocycle in DMSO was added tosic acid monohydrate and the depicted amine. The reaction mixture was heated at 70° C. until the starting material had been consumed as determined by LCMS. The reaction was poured into water and extracted with EA. The organic layers were combined, washed with brine, dried over sodium sulfate, and concd by rotary evaporation to afford the depicted product as a crude mixture, which was used directly in the next reaction. In some embodiment, X is a halide. It is understood that the ring bearing the N description is any heteroaromatic ring containing at least one nitrogen atom. In some embodiments, the ring bearing the N description is R4as defined for formula (A). In some embodiments, one of the two R groups attached to nitrogen atom of the reaction product is R14, and the other R group attached to the nitrogen atom of the reaction product is R15, wherein R14and R15are as defined for formula (A). In some embodiments, the two R groups attached to the nitrogen atom are taken together with the nitrogen atom to which they are attached to form R12, wherein R12is a 3- to 12-membered heterocyclyl optionally substituted by R12a, wherein R12ais as defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure E To a solution of the depicted amine (1 equiv) in MeOH was added aldehyde (1.3 equiv), NaBH3CN (2.5 equiv), and acetic acid (1 equiv) at 0° C. The mixture was allowed to warm to rt and was stirred for 18 h or until LCMS indicated product formation was complete. The reaction mixture was then treated with sat aq sodium carbonate and extracted with DCM. The combined organic layers were washed with brine and concd by rotary evaporation to afford a crude residue, which was purified by reverse phase preparative TLC (PE:EA 1:1) to afford the depicted product. In some embodiments, the R group attached to the methylene moiety of the reaction product is R4aas defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure F A solution of the depicted amine (1 equiv) was prepared in DCE, and the reaction mixture was adjusted to pH 6 by the addition of AcOH before adding sodium triacetoxyborohydride (2.5 equiv). The reaction mixture was cooled to 0° C. before adding ketone (1.5 equiv). The reaction was allowed to warm to rt and stirred for 16 h, at which time LCMS indicated the presence of the depicted product. The reaction mixture was treated with sat aq sodium bicarbonate and extracted with DCM. The organic layer was washed with brine, dried over sodium sulfate, and concd to afford the crude residue, which was purified by preparative TLC (PE:EA, 1:1) to afford the depicted product. In some embodiments, the R group attached to the methylene moiety of the reaction product is R4aas defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure G To a solution of the depicted amine (1 equiv) in DCM was added DIPEA (10 equiv) followed by acid chloride (4 equiv). The reaction was stirred at rt for 1 h, concd, and used directly in the next step. In some embodiments, the R group attached to the amide moiety of the reaction product is R3as defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure H To a solution of the depicted BOC-protected amine in DCM or MeOH was added either TFA or HCl in 1,4-dioxane or diethyl ether in excess. The reaction was stirred at rt until LCMS indicated the starting material had been consumed. The reaction was then concd by rotary evaporation to afford the depicted product as a salt, which was used directly in the next reaction. In some embodiments, Y refers to the portion of the molecule that links the —C(O)N(H)— portion of the compound with the remainder of the R3moiety. In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure I To a solution of the depicted amine (1.0 equiv) in DCM was added DIPEA (4 equiv) followed by acid chloride or anhydride (2 equiv). The reaction was stirred at rt and monitored by LCMS for the consumption of starting material. The reaction mixture was then concd and purified by reverse phase preparatory HPLC to afford the depicted product. In some embodiments, Y refers to the portion of the molecule that links the —C(O)N(H)— portion of the compound with the remainder of the R3moiety. In some embodiments, the R group attached to the nitrogen atom of the heterocyclyl moiety is R3as defined for formula (A). In some embodiments, the R group attached to the nitrogen atom of the heterocyclyl moiety is R12aas defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure J A mixture of amine (1 equiv), aldehyde (1.5 equiv), and NaBH3CN (5 equiv) in MeOH was stirred at rt for 12 h until starting material had been consumed as determined by LCMS. The reaction mixture was diluted with EA and washed with brine, dried over sodium sulfate, filtered, and concd to afford the crude residue, which was purified by preparative TLC or column chromatography on silica gel to afford the depicted product. In some embodiments, Y refers to the portion of the molecule that links the —C(O)N(H)— portion of the compound with the remainder of the R3moiety. In some embodiments, the R group attached to the nitrogen atom of the heterocyclyl moiety is R3fas defined for formula (A). In some embodiments, the R group attached to the nitrogen atom of the heterocyclyl moiety is R12aas defined for formula (A). In some embodiments, the R group attached to the ester moiety of the starting material and reaction product is a carboxylic acid protecting group. Procedure K tert-Butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-methylpiperidine-1-carboxylate. To a solution of methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate hydrochloride (390 mg, 1.22 mmol) in DMF (1 mL) and THF (3 mL) was added 1-(tert-butoxycarbonyl)-4-methylpiperidine-4-carboxylic acid (326 mg, 1.34 mmol), diisopropylethylamine (0.85 mL, 4.9 mmol), and HATU (510 mg, 1.34 mmol). The reaction was allowed to stir at rt for 16 h before diluting with water, extracting with EtOAc, washing with brine, drying over sodium sulfate, and concentrating. The crude residue was purified by FCC eluting with 0-15% MeOH in DCM to afford tert-Butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-methylpiperidine-1-carboxylate (614 mg, 92% yield). LCMS theoretical m/z=545.4 [M+H]+, found 545.4. Procedure L (S)-methyl 2-((tert-butoxycarbonyl)amino)-5-oxonon-8-enoate: To a solution of (S)-1-tert-butyl 2-methyl 5-oxopyrrolidine-1,2-dicarboxylate (250 g, 1.0 mol, 1.0 equiv) in THF (2500 mL) was added but-3-en-1-ylmagnesium bromide (1.0 M, 1.2 L, 1.2 equiv) dropwise at −78° C. for 30 min, and then the solution was stirred at −78° C. for 1.5 h. TLC (PE:EA=5:1) showed that a new spot appeared. The mixture was quenched with sat NH4Cl (500 mL) and separated. The aqueous layer was extracted with EA. The combined organic layers were dried over Na2SO4and concd. The residue was purified by column chromatography (SiO2, PE:EA=15:1) to yield the title compound (180 g, 0.63 mol, 61% yield) as colorless oil. LCMS (ESI+): m/z=300.1 (M+H)+.1H NMR (400 MHz, CDCl3): δ ppm 5.74-5.84 (m, 1H) 4.88-5.21 (m, 3H) 4.27 (br d, J=4.63 Hz, 1H) 3.74 (s, 3H) 2.42-2.62 (m, 4H) 2.32 (q, J=7.06 Hz, 2H) 2.08-2.20 (m, 1H) 1.82-1.97 (m, 1H) 1.44 (s, 9H). (S)-methyl 2-((tert-butoxycarbonyl)amino)non-8-enoate: To a solution of (S)-methyl 2-((tert-butoxycarbonyl)amino)-5-oxonon-8-enoate (200 g, 670 mmol, 1.0 equiv) in AcOH (2 L) was added 4-methylbenzenesulfonohydrazide (147 g, 788 mmol, 1.18 equiv). The mixture was stirred at 15° C. for 2 h, then NaBH(OAc)3(566 g, 2.67 mol, 4.00 equiv) was added. The solution was stirred 15 h at 35° C. TLC (PE:EA=5:1) showed that a new spot had appeared and that the starting material was consumed. The mixture was concd and poured into cold water (12 L) and extracted with EA. The combined organic phases were washed with sat aq NaHCO3(1.2 L) and brine (1.2 L), dried over Na2SO4, filtered, and concd to afford a crude residue. The crude residue was purified by column chromatography (SiO2, PE:EA=15:1) to afford the title compound (105 g, 368 mmol, 55.1% yield) as a colorless oil.1H NMR (400 MHz, CDCl3): δ ppm 5.72-5.82 (m, 1H) 4.87-5.07 (m, 3H) 4.17-4.38 (m, 1H) 3.72 (s, 3H) 1.95-2.08 (m, 2H) 1.69-1.86 (m, 1H) 1.53-1.66 (m, 1H) 1.21-1.50 (m, 15H). (S,E)-methyl 2-((tert-butoxycarbonyl)amino)-10-oxoundec-8-enoate: To a solution of but-3-en-2-one (62.6 g, 893 mmol, 74.5 mL, 3.00 equiv) and Grubbs catalyst 2ndGeneration (12.6 g, 14.9 mmol, 0.0500 equiv) in DCM (800 mL) was added (S)-methyl 2-((tert-butoxycarbonyl)amino)non-8-enoate (85.0 g, 297 mmol, 1.00 equiv) at 40° C., and the mixture was stirred for 24 h. TLC (PE:EA=5:1) showed that a new spot appeared, and LCMS indicated that the starting material had been completely consumed. The solution was concd to give a crude residue. The crude residue was purified by column chromatography (SiO2, PE:EA=15:1) to afford the title compound (62.9 g, 192 mmol, 64.5% yield) as a colorless oil. LCMS (ESI+): m/z=228.4 (M+H−BOC)+;1H NMR (400 MHz, CDCl3): δ ppm 6.67-6.90 (m, 1H) 6.07 (dt, J=15.99, 1.38 Hz, 1H) 5.00 (br d, J=7.72 Hz, 1H) 4.22-4.37 (m, 1H) 3.75 (s, 3H) 2.15-2.28 (m, 5H) 1.75-1.85 (m, 1H) 1.57-1.66 (m, 1H) 1.43-1.50 (m, 11H) 1.31-1.39 (m, 4H); Chiral SFC method: column: Daicel CHIRALPAK® AD-3 (Chiral Technologies, Inc., West Chester, PA), 3 μm, 0.46×10 cm, 4.0 mL/min, 220 nm, phase A=CO2, Phase B=MeOH (0.05% IPA), Rt1=1.14 min, Rt2=1.29 min, 100% ee. (S)-methyl 2-((tert-butoxycarbonyl)amino)-10-oxoundecanoate: To a solution of (S,E)-methyl 2-((tert-butoxycarbonyl)amino)-10-oxoundec-8-enoate (100 g, 305 mmol, 1.00 equiv) in MeOH (400 mL) was added Pd/C (30 g, 10% purity), and the flask was evacuated and purged with H2gas (15 psi). The reaction flask was left under a H2balloon (15 psi) atmosphere for 14 h at 20° C. TLC (PE:EA=5:1) showed that starting material had been consumed and a new spot was detected. The solution was filtered through Celite and concd to afford (the title compound (300 g, 911 mmol, 99.4% yield) as a colorless oil.1H NMR (400 MHz, CDCl3): δ ppm 4.89-5.07 (m, 1H) 4.22-4.37 (m, 1H) 3.74 (s, 3H) 2.41 (t, J=7.40 Hz, 2H) 2.13 (s, 3H) 1.78 (br dd, J=12.96, 5.14 Hz, 1H) 1.51-1.66 (m, 3H) 1.45 (s, 9H) 1.23-1.36 (m, 8H). (S)-methyl 2-((tert-butoxycarbonyl)amino)-9-(1,8-naphthyridin-2-yl)nonanoate: To a solution of (S)-methyl 2-((tert-butoxycarbonyl)amino)-10-oxoundecanoate (20.0 g, 60.7 mmol, 1.00 equiv) in EtOH (200 mL) was added L-proline (3.49 g, 30.4 mmol, 0.500 equiv) and 2-aminonicotinaldehyde (7.41 g, 60.7 mmol, 1.00 equiv). The mixture was stirred at 65° C. for 13 h. TLC (PE:EA, 1:1) showed a new spot was detected with Rf=0.16. The reaction mixture was concd under reduced pressure to remove solvent. The residue was diluted with H2O (2000 mL) and extracted with EA. The combined organic layers were washed with brine (2000 mL), dried over anhyd Na2SO4, filtered, and concd under reduced pressure to give the crude residue. The residue was purified by column chromatography (SiO2, PE:EA=3:1 to 1:1) to afford the title compound (120 g, 289 mmol, 31.7% yield) as a yellow oil. LCMS (ESI+): m/z=416.2 (M+H)+;1H NMR (400 MHz, CDCl3): δ ppm 9.08 (dd, J=4.19, 1.98 Hz, 1H) 8.16 (dd, J=8.16, 1.98 Hz, 1H) 8.09 (d, J=8.38 Hz, 1H) 7.44 (dd, J=8.05, 4.30 Hz, 1H) 7.39 (d, J=8.38 Hz, 1H) 4.90-5.14 (m, 1H) 4.21-4.35 (m, 1H) 3.73 (s, 3H) 2.98-3.10 (m, 2H) 1.88 (quin, J=7.50 Hz, 2H) 1.70-1.82 (m, 1H) 1.53-1.67 (m, 1H) 1.44 (s, 9H) 1.29-1.48 (m, 8H); Chiral SFC method: column: Daicel CHIRALPAK® AD-3 (Chiral Technologies, Inc., West Chester, PA), 3 μm, 0.46×10 cm, 2.5 mL/min, 220 nm, phase A=CO2, Phase B=MeOH (0.05% IPA), Rt1=3.25 min, Rt2=3.45 min, 99.6% ee. (S)-methyl 2-((tert-butoxycarbonyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate: To a solution of (S)-methyl 2-((tert-butoxycarbonyl)amino)-9-(1,8-naphthyridin-2-yl)nonanoate (22.0 g, 52.9 mmol, 1.00 equiv) in MeOH (200 mL) was added Pd/C (6 g, 10% purity). The flask was evacuated and back-filled with H2(50 Psi), and held for 5 h at 25° C. LCMS showed that starting material had been completely consumed, and one main peak with the product mass was detected. The solution was concd to give a residue. The residue was purified by prep-HPLC (column: XTIMATE® (Welch Materials, Hurst, TX); C18 10 μm 250 mm×50 mm; mobile phase: [water (10 mM NH4HCO3)—I]; B %: 50%-73%, 20 min) to afford the title compound (77.9 g, 179 mmol, 48.3% yield) as a white solid. LCMS (ESI+): m/z=420.2 (M+H)+;1H NMR (400 MHz, CDCl3): δ ppm 7.05 (d, J=7.45 Hz, 1H) 6.34 (d, J=7.45 Hz, 1H) 5.01 (br d, J=8.33 Hz, 1H) 4.79 (br s, 1H) 4.21-4.36 (m, 1H) 3.73 (s, 3H) 3.35-3.46 (m, 2H) 2.69 (t, J=6.36 Hz, 2H) 2.42-2.59 (m, 2H) 1.86-1.95 (m, 2H) 1.72-1.84 (m, 1H) 1.55-1.67 (m, 3H) 1.44 (s, 9H) 1.30 (br s, 8H); Chiral SFC method: Daicel CHIRALPAK® AD-3 (Chiral Technologies, Inc., West Chester, PA), 3 μm, 0.46×10 cm, 2.5 mL/min, 220 nm, phase A=CO2, Phase B=MeOH (0.05% IPA), Rt1=3.04 min, Rt2=3.32 min, 99.5% ee. Methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (1.6 g, 3.8 mmol, 1.0 equiv) in 10 mL of DCM was added 4 N HCl in 1,4-dioxane (7.6 mL, 30 mmol, 8.0 equiv). The solution was stirred for 1 h until LCMS showed the starting material had been consumed. The reaction solution was concd via rotary evaporation to afford the title compound as a sticky, yellow solid, which was used without further purification. Procedure M (S)-2-(1-(3-fluoropropyl)-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. To a solution of methyl (S)-2-(1-(3-fluoropropyl)-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (113 mg, 0.225 mmol) in THF:MeOH:H2O (3:1:1) was added lithium hydroxide (22 mg, 0.90 mmol). The reaction mixture was stirred at RT for 12 h. The reaction mixture was diluted with AcOH:H2O (1:1) and purified by reverse phase prep HPLC to afford the title compound (44 mg, 40% yield) as a thin film. LCMS theoretical m/z=491.3 [M+H]+, found 491.3. Procedure N (S)-methyl 2-((3R,5S)-3,5-dimethylmorpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. Et3N (220 mg, 2.18 mmol) was added to a solution of methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (250 mg, 702 μmol, HCl) and CDI (125 mg, 772 μmol) in dry DMF (2.5 mL) and THF (5 mL) at 0° C., which was stirred for 30 min. A solution of (3R,5S)-3,5-dimethylmorpholine (117 mg, 772 μmol, HCl) in DMF (2.5 mL) was added to the mixture. The mixture was allowed to warm to 25° C. and stirred for 12 h. LCMS showed that the desired mass was detected. The mixture was diluted with H2O (5 mL), and extracted with EA (5 mL×3). The combined organic layers were washed with H2O (5 mL), dried and concd. The residue was purified by prep-TLC (SiO2, PE:EA=0:1) to yield the title compd (150 mg, 326 μmol, 46.4% yield) as yellow liquid. LCMS theoretical m/z=461.3 [M+H]+, found 461.2.400 MHz1H NMR, CDCl3, δ ppm 6.99 (d, J=7.06 Hz, 1H), 6.26 (d, J=7.28 Hz, 1H), 4.81 (br d, J=7.50 Hz, 1H), 4.39-4.55 (m, 1H), 3.72-3.89 (m, 2H), 3.61-3.70 (m, 5H), 3.48-3.57 (m, 2H), 3.32 (br d, J=4.41 Hz, 2H), 2.62 (t, J=6.17 Hz, 2H), 2.40-2.49 (m, 2H), 1.65-1.87 (m, 4H), 1.49-1.56 (m, 2H), 1.17-1.35 (m, 14H). Procedure O Methyl (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-2-methylhept-6-enoate. To a solution of (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-2-methylhept-6-enoic acid (9.00 g, 23.7 mmol) in DMF (90 mL) was added K2CO3(6.56 g, 47.4 mmol) and Mel (6.73 g, 47.4 mmol) at 0° C., then the reaction mixture was stirred at 20° C. for 2 h. The reaction mixture was poured into H2O (30 mL) and extracted with EA. The combined organic layers were dried over Na2SO4, filtered, and concd under reduced pressure to give a residue. The residue was purified by FCC (2% to 12% pet-ether in EA) to afford 8.6 g of title compd (92% yield) as a colorless oil. 400 MHz1H NMR, CDCl3, δ ppm 7.78 (d, J=7.58 Hz, 2H) 7.61 (d, J=7.34 Hz, 2H) 7.37-7.46 (m, 2H) 7.30-7.37 (m, 2H) 5.53-5.86 (m, 2H) 4.88-5.08 (m, 2H) 4.39 (br s, 2H) 4.20-4.27 (m, 1H) 3.77 (br s, 3H) 2.17 (br s, 1H) 2.04 (br s, 1H) 1.70-1.91 (m, 1H) 1.59 (br s, 3H) 1.31-1.47 (m, 1H) 1.07-1.24 (m, 1H). Procedure P tert-butyl (S)-3-(((1-ethoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)oxy)-3-methylazetidine-1-carboxylate. To a solution of tert-butyl 3-(2,5-dioxopyrrolidin-1-yl)oxycarbonyloxy-3-methyl-azetidine-1-carboxylate (120 mg, 0.38 mmol) and DIPEA (0.22 mL, 1.3 mmol) in DCM (10 mL) was added ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (120 mg, 0.38 mmol). The reaction was stirred at RT for 1 h and concentrated. The crude product was used without further purification. Procedure Q (S)—N—((S)-1-cyano-4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octyl)-2-methylpropane-2-sulfinamide. To a solution of 4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octanal (128 mg, 0.43 mmol) in THF (3 mL) was added (S)-2-methylpropane-2-sulfinamide (63 mg, 0.52 mmol) followed by Titanium(IV) ethoxide (247 mg, 1.085 mmol) at rt. The reaction mixture was refluxed for 30 h in which (S)—N-(4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octylidene)-2-methylpropane-2-sulfinamide was generated and used directly. In a separate reaction flask, diethylaluminium (1 M in toluene, 0.645 mL, 0.645 mmol) was added to a solution of i-PrOH (33 μL, 0.43 mmol) in THF (2 mL). After stirring for 10 min, the reaction mixture was cooled to −78° C., the previously generated intermediate in THF was added to the reaction mixture. The reaction mixture was allowed to warm up to rt slowly and stirred at ambient temperature for 10 h. The reaction was quenched with sat aq NH4Cl. The reaction mixture was diluted with EA and H2O, and filtered through a Celite pad. The aq phase was separated and extracted with EA, dried over Na2SO4, filtered, concd, and purified by prep-reverse phase HPLC to give (S)—N—((S)-1-cyano-4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octyl)-2-methylpropane-2-sulfinamide. LCMS (ESI+): m/z=427.2 [M+H]+. Procedure R (S)-2-amino-5,5-difluoro-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. To a solution of (S)—N—((S)-1-cyano-4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octyl)-2-methylpropane-2-sulfinamide (35 mg, 82 μmol) in 1,4-dioxane:H2O (1:1, 1 mL) was added H2SO4(45 μL, 0.82 mmol) at rt. The reaction mixture was refluxed for 20 h. Prep-reverse phase HPLC purification afforded (S)-2-amino-5,5-difluoro-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. LCMS (ESI+): m/z=342.1 [M+H]+. Procedure S tert-butyl 3-((((2,5-dioxopyrrolidin-1-yl)oxy)carbonyl)oxy)-3-methylazetidine-1-carboxylate. A solution of tert-butyl 3-hydroxy-3-methylazetidine-1-carboxylate (1.09 g, 5.81 mmol) in ACN (50 mL) was added bis(2,5-dioxopyrrolidin-1-yl) carbonate (2.98 g, 11.6 mmol) and DIPEA (2.02 mL, 11.6 mmol). The reaction was allowed to stir at rt for 18 h and was then concentrated and used without further purification. SYNTHETIC EXAMPLES The chemical reactions in the Synthetic Examples described can be readily adapted to prepare a number of other compounds of the invention, and alternative methods for preparing the compounds of this invention are deemed to be within the scope of this invention. For example, the synthesis of non-exemplified compounds according to the invention can be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention. Compound 1: (S)-2-pivalamido-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and pivalic acid using Procedures A and C. LCMS theoretical m/z=390.3 [M+H]+, found 390.1. Compound 2: (S)-2-((S)-1-(pyridin-2-yl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate using Procedure A with pyridin-2-yl-L-proline and Procedure C. LCMS theoretical m/z=480.3 [M+H]+, found 480.3. Compound 3: (S)-2-((R)-1-(pyridin-2-yl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate using Procedure A with pyridin-2-yl-D-proline, followed by Procedure C. LCMS theoretical m/z=480.3 [M+H]+, found 480.3. Compound 4: (S)-2-(2-methyl-2-(pyridin-3-yl)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate using Procedure A with 2-methyl-2-(pyridin-3-yl)propanoic acid, followed by Procedure C. LCMS theoretical m/z=453.3 [M+H]+, found 453.0. Ethyl (S)-2-(2-ethylbutanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a mixture of ethyl (2S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate bis hydrochloride salt (70 mg, 0.2 mmol, 1 equiv) in DCM (0.5 mL) at rt was added DIPEA (0.21 mL, 1.2 mmol, 6 equiv). The mixture was sonicated to aid dissolution. The mixture was treated by slowly adding 2-ethylbutanoyl chloride (0.04 mL, 0.3 mmol). The resulting mixture was stirred at rt for 2 d. LCMS of the reaction mixture showed the product mass, and the reaction was concd and used directly in the next reaction. Compound 5: (S)-2-(2-ethylbutanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Scheme I and Procedure C beginning with ethyl (S)-2-(2-ethylbutanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=404.3 [M+H]+, found 404.3. Ethyl (S)-2-(morpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a mixture of ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate bis hydrochloride (60 mg, 0.17 mmol, 1 equiv) in DCM (0.5 mL) at rt was added DIPEA (0.18 mL, 1.0 mmol, 6 equiv). The mixture was sonicated to aid dissolution. To the mixture was added morpholine-4-carbonyl chloride (0.03 mL, 0.26 mmol). The reaction was stirred at rt for 2 d until LCMS showed the mass of the title compound. The reaction mixture was concd and used directly in the next reaction. Compound 6: (S)-2-(morpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Scheme I with Procedure C starting with ethyl (S)-2-(morpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=419.3 [M+H]+, found 419.3. Compound 7: (2S)-2-(2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2,2-dimethyltetrahydropyran-4-carboxylic acid using Procedures A and C. LCMS theoretical m/z=446.3 [M+H]+, found 446.3. Compound 8: (S)-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-methyltetrahydro-2H-pyran-4-carboxylic acid using Procedures A and C. LCMS theoretical m/z=432.2 [M+H]+, found 432.3. Compound 9: (S)-2-((S)-1-phenylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate using Procedure A with (S)-1-phenylpyrrolidine-2-carboxylic acid and Procedure C. LCMS theoretical m/z=479.3 [M+H]+, found 479.3. Compound 10: (S)-2-((S)-1-benzylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (S)-1-benzylpyrrolidine-2-carboxylic acid using Procedures A and C. LCMS theoretical m/z=493.3 [M+H]+, found 493.0. Compound 11: (S)-2-(2-methyl-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate using Procedure A with 2-methyl-2-phenylpropanoic acid, followed by Procedure C. LCMS theoretical m/z=452.3 [M+H]+, found 452.3. Ethyl (S)-2-((S)-1-(pyrimidin-2-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a solution of ethyl (S)-2-((S)-pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (50 mg, 0.08 mmol, 1 equiv) in 0.5 mL MeOH was added pyrimidine-2-carbaldehyde (0.018 mL, 0.19 mol, 2.5 equiv). The mixture was heated at 40° C. for 10 min before adding sodium cyanoborohydride (12 mg, 0.19 mmol, 2.5 equiv) and continuing to heat for an additional 2 h. The crude mixture was used directly in the next step. Compound 12: (S)-2-((S)-1-(pyrimidin-2-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Scheme E with Procedure C employing a crude mixture of ethyl (S)-2-((S)-1-(pyrimidin-2-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=495.3 [M+H]+, found 495.3. Ethyl (S)-2-((S)-pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. (S)-2-((S)-1-(tert-butoxycarbonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid was prepared using Procedure A. (S)-2-((S)-1-(tert-butoxycarbonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid (334 mg, 0.63 mmol, 1.0 equiv) was treated with 4 N HCl in 1,4-dioxane (2.0 mL) at rt for 30 min. The reaction was concd and then azeotroped with EA and frozen to afford a yellow paste. The material was then purified by RP-HPLC to afford 201 mg (48% yield) of the title compound as the TFA salt, a yellowish, viscous oil, which was used directly in the next step. Ethyl (S)-2-((S)-1-(2-(Pyridin-4-yl)acetyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a solution of ethyl (S)-2-((S)-pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (50 mg, 0.08 mmol, 1 equiv) in 0.5 mL MeOH was added isonicotinaldehyde (0.01 mL, 0.15 mol, 2 equiv). The mixture was heated at 40° C. for 10 min before adding sodium cyanoborohydride (9.5 mg, 0.15 mmol, 2 equiv) and continuing to heat for an additional h. The crude mixture was used directly in the next step. Compound 13: (S)-2-((S)-1-(2-(Pyridin-4-yl)acetyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Scheme E with Procedure C employing a crude mixture of ethyl (S)-2-((S)-1-(pyridin-4-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=494.3 [M+H]+, found 494.3. Ethyl (S)-2-((S)-1-(pyrimidin-4-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a solution of ethyl (S)-2-((S)-pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (50 mg, 0.08 mmol, 1 equiv) in 0.5 mL MeOH was added pyrimidine-4-carbaldehyde (0.018 mL, 0.19 mol, 2.5 equiv). The mixture was heated at 40° C. for 10 min before adding sodium cyanoborohydride (12 mg, 0.19 mmol, 2.5 equiv) and continuing to heat for an additional 2 h. The crude mixture was used directly in the next step. Compound 14: (S)-2-((S)-1-(pyrimidin-4-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Scheme E with Procedure C employing a crude mixture of (S)-2-((S)-1-(pyrimidin-2-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. LCMS theoretical m/z=495.3 [M+H]+, found 495.3. Ethyl (S)-2-((S)-1-(pyridin-3-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a solution of ethyl (S)-2-((S)-pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (50 mg, 0.08 mmol, 1 equiv) in 0.5 mL MeOH was added 3-pyridinecarboxaldehyde (0.018 mL, 0.19 mol, 2.5 equiv). The mixture was heated at 40° C. for 10 min before adding sodium cyanoborohydride (12 mg, 0.19 mmol, 2.5 equiv) and continuing to heat for an additional 2 h. The crude mixture was used directly in the next step. Compound 15: (S)-2-((S)-1-(pyridin-3-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Scheme E with Procedure C employing a crude mixture of ethyl (S)-2-((S)-1-(pyridin-3-ylmethyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=494.3 [M+H]+, found 494.3. Compound 16: (S)-2-((S)-1-(tert-butoxycarbonyl)piperidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared with Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (S)-1-(tert-butoxycarbonyl)piperidine-2-carboxylic acid using Procedures A and C. LCMS theoretical m/z=517.3 [M+H]+, found 517.3. Compound 17: (S)-2-(2-(2-chlorophenyl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared with Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2-chlorophenylacetic acid using Procedures A and C. LCMS theoretical m/z=458.2 [M+H]+, found 458.2. Compound 18: (S)-2-((3R,4R)-1-(tert-butoxycarbonyl)-3-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared with Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (3S,4S)-1-tert-butoxycarbonyl-3-methyl-piperidine-4-carboxylic acid using Procedures A and C. LCMS theoretical m/z=531.3 [M+H]+, found 531.4. Compound 19: (S)-2-(1-(tert-butoxycarbonyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared with Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid using Procedures A and C. LCMS theoretical m/z=517.3 [M+H]+, found 517.3. Compound 20: (S)-2-(2-((S)-1-(tert-butoxycarbonyl)pyrrolidin-2-yl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared with Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate 2-[(2S)-1-tert-butoxycarbonylpyrrolidin-2-yl]acetic acid using Procedures A and C. LCMS theoretical m/z=517.3 [M+H]+, found 517.3. Compound 21: (S)-2-((S)-1-benzylazetidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Scheme E with Procedure A employing ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (S)-1-(tert-butoxycarbonyl)azetidine-2-carboxylic acid, Procedures H, J, and C. LCMS theoretical m/z=479.3 [M+H]+, found 479.2. Ethyl (S)-2-((2S,3S)-1-(3-methoxypropanoyl)-3-methylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. Ethyl (S)-2-((2S,3S)-3-methylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate was synthesized according to Procedure A using ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (2S,3S)-1-tert-butoxycarbonyl-3-methyl-pyrrolidine-2-carboxylic acid. To a mixture of ethyl (S)-2-((2S,3S)-3-methylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (17 mg, 0.038 mmol, 1 equiv) and 3-methoxypropionic acid (5 mg, 0.5 mmol, 1.2 equiv) in THF (0.5 mL) was added HATU (17 mg, 0.05 mmol, 1.2 equiv) followed by DIPEA (0.04 mL, 0.2 mmol, 6 equiv). The reaction was stirred at rt for 1 h before concentrating and purifying by reverse phase chromatography to afford the title compound as a white solid, which was used directly in the next reaction. Compound 22: (S)-2-((2S,3S)-1-(3-methoxypropanoyl)-3-methylpyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared with Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2-[(2S)-1-tert-butoxycarbonylpyrrolidin-2-yl]acetic acid using Procedure C. LCMS theoretical m/z=503.3 [M+H]+, found 503.3. Ethyl (S)-2-((R)-piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. Ethyl (S)-2-((R)-piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate was synthesized according to Procedure A employing ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (R)-1-(tert-butoxycarbonyl)piperidine-3-carboxylic acid. To a solution of ethyl (S)-2-((R)-piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (72 mg, 0.12 mmol) was added 4 N HCl in 1,4-dioxane (0.5 mL). The reaction was stirred for 1 h at rt before concentrating. The crude residue was used directly in the next reaction. Compound 23: (S)-2-((R)-1-(3-methoxypropanoyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme E and the above description as well as Procedure C, employing ethyl (S)-2-((R)-1-(3-methoxypropanoyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=503.3 [M+H]+, found 503.3. Compound 24: (S)-2-(4-(methylsulfonyl)butanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-methylsulfonylbutanoic acid using Procedures A and C. LCMS theoretical m/z=454.2 [M+H]+, found 454.3. Compound 25: (S)-2-((R)-2-hydroxy-2-phenylacetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (R)-(−)-mandelic acid using Procedures A and C. LCMS theoretical m/z=440.2 [M+H]+, found 440.3. Compound 26: (S)-2-((S)-2-hydroxy-2-phenylacetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (S)-(−)-mandelic acid using Procedures A and C. LCMS theoretical m/z=440.2 [M+H]+, found 440.3. Compound 27: (S)-2-((R)-3-hydroxy-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A using Procedure C employing ethyl (2S)-2-(3-hydroxy-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate to afford one of the (R)- and (S)-enantiomers by reverse phase column chromatography as the first eluting peak. Absolute stereochemistry at the benzylic center was unassigned, as indicated by the wavy bond for Compound 27 inFIG.1. LCMS theoretical m/z=454.3 [M+H]+, found 454.3. Compound 28: (S)-2-((S)-3-hydroxy-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A using Procedure C employing ethyl (2S)-2-(3-hydroxy-2-phenylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate to afford, compared to Compound 27, the other of the (R)- and (S)-enantiomers by reverse phase column chromatography as the second eluting peak. Absolute stereochemistry at the benzylic center was unassigned, as indicated by the wavy bond for Compound 28 inFIG.1. LCMS theoretical m/z=454.3 [M+H]+, found 454.3. Compound 29: (S)-2-(3,3-diethylureido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme I using Procedure G with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and diethylcarbamic chloride followed by Procedure C. LCMS theoretical m/z=405.3, [M+H]+, found 405.3. Compound 30: (S)-2-(4-methoxybutanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-methoxybutanoic acid using Procedures A and C. LCMS theoretical m/z=405.5. [M+H]+, found 406.4. Compound 31: (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-((R)-tetrahydrofuran-3-carboxamido)nonanoic acid and (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-((S)-tetrahydrofuran-3-carboxamido)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and tetrahydrofuran-3-carboxylic acid using Procedures A and C. LCMS theoretical m/z=403.5. [M+H]+, found 404.3. Compound 32: (S)-2-((((1-(tert-butoxycarbonyl)-3-methylazetidin-3-yl)oxy)carbonyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme I beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate, then generating tert-butyl 3-(2,5-dioxopyrrolidin-1-yl)oxycarbonyloxy-3-methyl-azetidine-1-carboxylate according to Procedure R using tert-butyl 3-(carboxyoxy)-3-methylazetidine-1-carboxylate, followed by Procedure C. LCMS theoretical m/z=519.3. [M+H]+, found 519.3. Compound 33: (2S)-2-[(1-tert-butoxycarbonylazetidin-3-yl)oxycarbonylamino]-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme I beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate then generating tert-butyl 3-(2,5-dioxopyrrolidin-1-yl)oxycarbonyloxy-3-methyl-azetidine-1-carboxylate according to Procedure R using tert-butyl 3-(carboxyoxy)-3-methylazetidine-1-carboxylate, followed by Procedure C. LCMS theoretical m/z=505.3. [M+H]+, found 505.3. Compound 34: (S)-2-(piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme C beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid using Procedure A, followed by Procedures H and C. LCMS theoretical m/z=417.3. [M+H]+, found 417.3. Compound 35: (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(tetrahydro-2H-pyran-4-carboxamido)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and tetrahydro-2H-pyran-4-carboxylic acid using Procedures A and C. LCMS theoretical m/z=418.3. [M+H]+, found 418.3. Compound 36: (S)-2-(1-acetylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme E beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid using Procedures A, H, I, and C. LCMS theoretical m/z=459.3. [M+H]+, found 459.2. Compound 37: (S)-2-((R)-1-(methylsulfonyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid and (S)-2-((S)-1-(methylsulfonyl)piperidine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(methylsulfonyl)piperidine-3-carboxylic acid using Procedures A and C. LCMS theoretical m/z=494.3. [M+H]+, found 495.3. Compound 38: (S)-2-(3-sulfamoylpropanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 3-sulfamoylpropanoic acid using Procedures A and C. LCMS theoretical m/z=441.2. [M+H]+, found 441.2. Compound 39: (S)-2-(1-(methylsulfonyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(methylsulfonyl) piperidine-4-carboxylic acid using Procedures A and C. LCMS theoretical m/z=495.3. [M+H]+, found 495.3. Compound 40: (S)-2-(3-(methylsulfonamido)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 3-(methylsulfonamido)propanoic acid using Procedures A and C. LCMS theoretical m/z=455.2. [M+H]+, found 455.3. Compound 41: (S)-2-((R)-3-methyltetrahydrofuran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid and (S)-2-((S)-3-methyltetrahydrofuran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 3-methyltetrahydrofuran-3-carboxylic acid using Procedures A and C to afford a 1:1 mixture of diastereomers. LCMS theoretical m/z=418.3 [M+H]+, found 418.3. Compound 42: (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(4-(trifluoromethyl)tetrahydro-2H-pyran-4-carboxamido)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-(trifluoromethyl)tetrahydropyran-4-carboxylic acid using Procedures A and C. LCMS theoretical m/z=486.3 [M+H]+, found 486.3. Compound 43: (S)-2-((1R,3s,5S)-8-oxabicyclo[3.2.1]octane-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid and (S)-2-((1R,3r,5S)-8-oxabicyclo[3.2.1]octane-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 8-oxabicyclo[3.2.1]octane-3-carboxylic acid using Procedures A and C to afford a mixture of diastereomers. LCMS theoretical m/z=444.3 [M+H]+, found 444.3. Compound 44: (S)-2-(1-methylcyclohexanecarboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-methylcyclohexane-1-carboxylic acid using Procedures A and C. Also prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-methylcyclohexanecarboxylic acid using Procedures K and M with methyl (S)-2-(1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=430.2 [M+H]+, found 430.3. Compound 45: (S)-2-(bicyclo[1.1.1]pentane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and bicyclo[1.1.1]pentane-1-carboxylic acid using Procedures A and C. Also prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and bicyclo[1.1.1]pentane-1-carboxylic acid using Procedures K and M. LCMS theoretical m/z=400.2 [M+H]+, found 400.2. Compound 46: (S)-2-((S)-chromane-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid and (S)-2-((R)-chromane-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and chromane-4-carboxylic acid using Procedures A and C to afford a 1:1 mixture of diastereomers. LCMS theoretical m/z=466.3 [M+H]+, found 466.3. Compound 47: (S)-2-((R)-3-methyltetrahydro-2H-pyran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid and (S)-2-((S)-3-methyltetrahydro-2H-pyran-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 3-methyltetrahydropyran-3-carboxylic acid using Procedures A and C to afford a 1:1 mixture of diastereomers. LCMS theoretical m/z=432.3 [M+H]+, found 432.3. Compound 48: (S)-2-(4-(((tert-butoxycarbonyl)amino)methyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme J using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-(((tert-butoxycarbonyl)amino)methyl)tetrahydro-2H-pyran-4-carboxylic acid, followed by Procedure C. LCMS theoretical m/z=547.3. [M+H]+, found 547.4. Compound 49: (S)-2-(4-Phenyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 3-methyltetrahydropyran-3-carboxylic acid using Procedures A and C. LCMS theoretical m/z=494.3 [M+H]+, found 494.3. Compound 50: (S)-2-(4-(aminomethyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme J using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-(((tert-butoxycarbonyl)amino)methyl)tetrahydro-2H-pyran-4-carboxylic acid. Final BOC removal was achieved using the following: (S)-2-(4-(((tert-butoxycarbonyl)amino)methyl) tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid (280 mg, 0.51 mmol, 1 equiv) was diluted with 1 mL DCM and treated with 2.55 mL of 2 M HCl in water (10 equiv) for 18 h. The reaction mixture was concd and azeotroped with hexanes. The product was then diluted in 1:1 ACN:H2O and placed under lyophilization to afford the title compound as a white foam (190 mg, 83% yield). LCMS theoretical m/z=447.3. [M+H]+, found 447.3. Compound 51: (R)-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Procedure L for the synthesis of (S)-methyl 2-((tert-butoxycarbonyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate except substituting 1-(tert-butyl) 2-methyl (R)-5-oxopyrrolidine-1,2-dicarboxylate for 1-(tert-butyl) 2-methyl (S)-5-oxopyrrolidine-1,2-dicarboxylate to afford methyl (R)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate dihydrochloride salt. The title compound was prepared according to Scheme A using Procedure B with methyl (R)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-methyltetrahydro-2H-pyran-4-carboxylic acid and Procedure C. LCMS theoretical m/z=432.3, [M+H]+, found 432.3. Methyl (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. Tert-Butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-methylpiperidine-1-carboxylate was synthesized according to Procedure A employing 1-tert-butoxycarbonyl-4-methyl-piperidine-4-carboxylic acid. To a crude solution of tert-butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-methylpiperidine-1-carboxylate (410 mg, 0.76 mmol, 1.0 equiv) in DCM (1.5 mL) was added TFA (1 mL). The reaction was stirred at rt for 12 h. LCMS showed no remaining starting material. The reaction was concd and purified by reverse phase preparative HPLC to afford 304 mg of the title compound as the TFA adduct (71% yield). Compound 52: (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme C beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-tert-butoxycarbonyl-4-methyl-piperidine-4-carboxylic acid using Procedure A. Procedure C was employed using used methyl (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=431.3 [M+H]+, found 431.3. Compound 53: (S)-2-(4-fluorotetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-fluorotetrahydro-2H-pyran-4-carboxylic acid using Procedures A and C. Also prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-fluorotetrahydro-2H-pyran-4-carboxylic acid using Procedures K and M. LCMS theoretical m/z=436.2 [M+H]+, found 436.2. Compound 54: (S)-2-((6-(propylsulfonyl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-chloro-6-(propylsulfonyl)pyrimidine using Procedures A and C. LCMS theoretical m/z=490.2 [M+H]+, found 490.0. Compound 55: (S)-2-((1-methyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate then using Procedure B with 7-chloro-1-methyl-1H-pyrazolo[4,3-d]pyrimidine and Procedure C with ethyl (S)-2-((1-methyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=438.3 [M+H]+, found 438.0. Compound 56: (S)-2-((5-(pyridin-3-yl)pyrimidin-2-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate, followed by Procedure B using 2-chloro-5-(pyridin-3-yl)pyrimidine and Procedure C using ethyl (S)-2-((5-(pyridin-3-yl)pyrimidin-2-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=461.3 [M+H]+, found 461.0. Compound 57: (S)-2-((1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate then using Procedure B with 7-chloro-1H-pyrazolo[4,3-d]pyrimidine and Procedure C with ethyl (S)-2-((1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=424.2 [M+H]+, found 424.0. 6-(difluoromethyl)pyrimidin-4-ol. To a mixture of ethyl 4,4-difluoro-3-oxobutanoate (5.00 g, 30.1 mmol), acetic acid (3.13 g, 30.1 mmol), and methanimidamide in MeOH (15.0 mL) was added MeONa (2.80 g, 71.9 mmol, 2.39 equiv) in one portion at 25° C. The mixture was stirred at 25° C. for 12 h until LCMS showed the consumption of starting material. The reaction mixture was diluted with acetic acid and H2O (90 mL, V:V=1:2) and extracted with EA (100 mL). The organic layer was washed with water (100 mL), dried over Na2SO4, and filtered. The filtrate was concd by rotary evaporation to afford 6-(difluoromethyl)pyrimidin-4-ol (2.75 g, 18.8 mmol, 62.5% yield) as yellow oil. The product was used to next step without further purification. 4-chloro-6-(difluoromethyl)pyrimidine. A mixture of 6-(difluoromethyl)pyrimidin-4-ol (2.57 g, 17.6 mmol) in POCl3(25.0 mL) was degassed and purged with N2, and then the mixture was stirred at 120° C. for 12 h under N2. TLC (PE:EA, 10:1, Rf=0.53) showed that the starting material had been consumed. The mixture was concd by rotary evaporation to remove POCl3. The mixture was diluted with dichloromethane and washed with aqueous NaHCO3), water, and aq NaCl. The organic layer was dried with Na2SO4and filtered, and the filtrate was concd by rotary evaporation to afford the title compound (800 mg, 4.86 mmol, 27.6% yield) as a brown oil. The product was used to next step without further purification. Compound 58: (S)-2-((6-(difluoromethyl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B using Procedure B with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-chloro-6-(difluoromethyl)pyrimidine and Procedure C. LCMS theoretical m/z=434.2 [M+H]+, found 434.2. Compound 59: (S)-2-((5-(pyridin-4-yl)pyrimidin-2-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate then using Procedure B with 2-chloro-5-(pyridin-4-yl)pyrimidine and Procedure C with ethyl (S)-2-((5-(pyridin-4-yl)pyrimidin-2-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=461.3 [M+H]+, found 461.0. Compound 60: (S)-2-((6-morpholinopyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme F beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4,6-dichloropyrimidine in Procedure B, using morpholine in Procedure D, and Procedure C to afford the title compound. LCMS theoretical m/z=469.3 [M+H]+, found 469.1. Compound 61: (S)-2-((6-(pyrrolidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme F beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4,6-dichloropyrimidine in Procedure B, using pyrrolidine in Procedure D, followed by Procedure C to afford the title compound. LCMS theoretical m/z=453.3 [M+H]+, found 453.2. Compound 62: (S)-2-((1-methyl-1H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and then using Procedure B with 4-chloro-1-methyl-1H-pyrazolo[3,4-d]pyrimidine and Procedure C. LCMS theoretical m/z=438.3 [M+H]+, found 438.2. Compound 63: (S)-2-((1H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate, then Procedure B using 4-chloro-1H-pyrazolo[3,4-d]pyrimidine, followed by Procedure C. LCMS theoretical m/z=424.2 [M+H]+, found 424.2. Compound 64: (S)-2-((1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate, then Procedure B using 7-chloro-1H-pyrazolo[4,3-d]pyrimidine, followed by Procedure C. LCMS theoretical m/z=424.2 [M+H]+, found 424.2. (S)-ethyl 2-((6-(4,4-difluoropiperidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. A solution of (S)-ethyl 2-((6-chloropyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (200.00 mg, 448.45 μmol, 1.00 equiv), 4,4-difluoropiperidine (706.71 mg, 4.48 mmol, 10.00 equiv, HCl) and TsOH·H2O (8.53 mg, 44.85 μmol, 0.10 equiv) in DMSO (2.00 mL) was stirred at 70° C. for 12 h. LCMS showed that the desired MS was detected. The reaction mixture was poured into water (15 mL), and extracted with EA. The organic layers were combined, washed with brine (30 mL), dried over sodium sulfate and evaporated under reduced pressure to yield the title compound (180.00 mg, 339.21 μmol, 75.64% yield) as yellow oil. LCMS theoretical m/z=531.3. [M+H]+, found 531.1. Compound 65: (S)-2-((6-(4,4-difluoropiperidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme F beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4,6-dichloropyrimidine in Procedure B, using the above description to afford (S)-ethyl 2-((6-(4,4-difluoropiperidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate, which was converted to the title compound by the following method: A solution of (S)-ethyl 2-((6-(4,4-difluoropiperidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (180 mg, 339 μmol, 1.00 equiv), HCl (12 M, 121 μL, 10.0 equiv), AcOH (20.4 mg, 0.339 mmol, 19.4 μL, 1.00 equiv) in ACN (5 mL) and H2O (5 mL) was stirred at 70° C. for 3 h. LCMS showed that the desired mass was detected. The solvent was removed in vacuo. The crude residue was purified by prep-HPLC (column: YMC-Actus Triart (YMC Co., Ltd., Kyoto, Japan) C18 150×30 mm 5 μm; mobile phase: [water (10 mM NH4HCO3)—I]; B %: 30%-50%, 12 min) to yield (S)-2-((6-(4,4-difluoropiperidin-1-yl)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid (2 mg, 0.004 mmol, 1% yield) as a yellow oil. LCMS theoretical m/z=503.3 [M+H]+, found 503.2. Compound 66: (S)-2-((6-(dimethylamino)pyrimidin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme F beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4,6-dichloropyrimidine in Procedure B, using dimethylamine in Procedure D, and Procedure C to afford the title compound. LCMS theoretical m/z=427.3. [M+H]+, found 427.2. Compound 67: (S)-2-(pyrimidin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B beginning with ethyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate then using Procedure B with 3-chloropyrimidine and Procedure C. LCMS theoretical m/z=384.2 [M+H]+, found 384.2. Compound 68: (S)-2-((8-bromoquinazolin-4-yl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B using Procedure B with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 8-bromo-4-chloroquinazoline followed by Procedure C. LCMS theoretical m/z=512.2, [M+H]+, found 513.2. Compound 69: (S)-2-(quinazolin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme B using Procedure B with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-chloroquinazoline followed by Procedure C. LCMS theoretical m/z=434.3, [M+H]+, found 434.3. (S)-methyl 2-(((2,3-dihydrobenzo[b][1,4]dioxin-6-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate: To a mixture of (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate hydrochloride (150 mg, 0.421 mmol, 1.00 equiv) in MeOH (3 mL) was added AcOH (25 mg, 0.42 mmol, 24 μL, 1.0 equiv), NaBH3CN (66 mg, 1.0 mmol, 2.5 equiv) at 0° C. under nitrogen. 2,3-Dihydrobenzo[b][1,4]dioxine-6-carbaldehyde (90 mg, 0.55, 55 μL, 1.3 equiv) was added into the mixture. The mixture was stirred at 20° C. for 18 h. LCMS showed the mass of the title compound. The mixture was treated with 6 mL NaHCO3solution and was extracted with DCM. The organic layer was washed with brine and Na2SO4and concd by rotary evaporation to give the crude residue, which was purified by preparative TLC (PE:EA, 1:1) to obtain (S)-methyl 2-(((2,3-dihydrobenzo[b][1,4]dioxin-6-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (130 mg, 0.23 mmol, 55% yield, 83% purity by HPLC) as a colorless oil. LCMS theoretical m/z=468.3 [M+H]+, found 468.5. Compound 70: (S)-2-(((2,3-dihydrobenzo[b][1,4]dioxin-6-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme G beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2,3-dihydrobenzo[b][1,4]dioxine-6-carbaldehyde using Procedures E and C. Also prepared according to Scheme G using Procedure E with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2,3-dihydrobenzo[b][1,4]dioxine-6-carbaldehyde and Procedure M with methyl (S)-2-(((2,3-dihydrobenzo[b][1,4]dioxin-6-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate to afford the title compd as a colorless oil. LCMS theoretical m/z=454.3 [M+H]+, found 454.2. 400 MHz1H NMR, methanol-d4, δ ppm 7.59 (d, J=7.28 Hz, 1H) 7.02 (s, 1H) 6.93-6.98 (m, 1H) 6.86-6.92 (m, 1H) 6.61 (d, J=7.50 Hz, 1H) 4.26 (s, 4H) 4.08-4.18 (m, 2H) 3.93 (t, J=6.06 Hz, 1H) 3.51 (t, J=5.62 Hz, 2H) 2.82 (t, J=6.17 Hz, 2H) 2.70 (t, J=7.83 Hz, 2H) 1.95 (dt, J=11.36, 5.79 Hz, 4H) 1.70 (br d, J=7.28 Hz, 2H) 1.39 (br s, 8H). Compound 71: (S)-2-(benzylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme G beginning with (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and benzaldehyde using Procedures E and C. Also prepared according to Scheme G beginning with Procedure E using methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and benzaldehyde using Procedures F and B. LCMS theoretical m/z=396.2 [M+H]+, found 396.2. Compound 72: (S)-2-((quinolin-4-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme H beginning with (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and quinoline-4-carbaldehyde using Procedures E and C. Also prepared according to Scheme G beginning with Procedure E using methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and quinoline-4-carbaldehyde and Procedure M with methyl (S)-2-((quinolin-4-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=447.2 [M+H]+, found 447.2. Compound 73: (S)-2-((quinolin-6-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme G beginning with (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and quinoline-6-carbaldehyde using Procedures E and C. Also prepared according to Scheme G beginning with Procedure E using methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and quinoline-6-carbaldehyde using Procedure M with methyl (S)-2-((quinolin-6-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=447.2 [M+H]+, found 447.2. Compound 74: (S)-2-((quinolin-8-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme G beginning with (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and quinoline-8-carbaldehyde using Procedures E and C. Also prepared according to Scheme G beginning with Procedure E using methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and quinoline-8-carbaldehyde and Procedure M with methyl (S)-2-((quinolin-8-ylmethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=447.2 [M+H]+, found 447.2. (2S)-methyl 2-((1-phenylethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate: To a mixture of (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (300 mg, 939.14 μmol, 1 equiv) in DCE (3 mL) was adjusted to pH=6 by AcOH. NaBH(OAc)3(497.61 mg, 2.35 mmol, 2.5 equiv) was added into the mixture at 0° C. under N2. Acetophenone (169.25 mg, 1.41 mmol, 164.32 μL, 1.5 equiv) was added into the mixture with stirring for 16 h at 20° C. LCMS indicated desired MS was detected. The mixture was quenched using NaHCO3solution and was extracted by DCM. The organic layer was dried by brine and Na2SO4, and concd under reduced pressure to give a residue. The crude product was purified by prep-TLC (PE:EA=0:1) to obtain the title compound (110 mg, 236.31 μmol, 25.16% yield, 91% purity) as a colorless oil. LCMS theoretical m/z=424.2 [M+H]+, found 424.2. Chiral purity: 41:58. Compound 75: (S)-2-(((R)-1-phenylethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid and (S)-2-(((S)-1-phenylethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme H beginning with (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and acetophenone using Procedures I and C. Also prepared according to Scheme G beginning with Procedure E using methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and acetophenone and Procedure M methyl (2S)-2-((1-phenylethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate to afford a mixture of diastereomers at the benzylic position. LCMS theoretical m/z=410.2 [M+H]+, found 410.2. Compound 76: (S)-2-(((1H-pyrrolo[2,3-b]pyridin-3-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme G beginning with (S)-methyl 2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1H-pyrrolo[2,3-b]pyridine-3-carbaldehyde using Procedures E and C. Also prepared according to Scheme G beginning with Procedure E with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1H-pyrrolo[2,3-b]pyridine-3-carbaldehyde using Procedure M with methyl (S)-2-(((1H-pyrrolo[2,3-b]pyridin-3-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=436.2 [M+H]+, found 436.2. Compound 77: (S)-2-((S)-4-(tert-butoxycarbonyl)morpholine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. May be prepared starting with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (S)-4-(tert-butoxycarbonyl)morpholine-3-carboxylic acid, employing an amide coupling reagent such as HATU in the presence of an amine base such as diisopropylethylamine to afford tert-butyl (S)-3-(((S)-1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)morpholine-4-carboxylate. tert-Butyl (S)-3-(((S)-1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)morpholine-4-carboxylate may then be converted to (S)-2-((S)-4-(tert-butoxycarbonyl)morpholine-3-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid by treatment with lithium hydroxide in a mixture of THF:MeOH:water 3:1:1 and purification by reverse-phase preparatory HPLC. Compound 78: (2S)-2-(7-oxabicyclo[2.2.1]heptane-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 7-oxabicyclo[2.2.1]heptane-2-carboxylic acid using Procedure K, methyl (2S)-2-(7-oxabicyclo[2.2.1]heptane-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate using Procedure M to afford a mixture of isomers. LCMS theoretical m/z=430.3 [M+H]+, found 430.4. Compound 79: (2S)-2-((2R)-7-oxabicyclo[2.2.1]heptane-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Compound 78 and separated by chiral SFC as follows: separation (column: Daicel CHIRALPAK® IC, Chiral Technologies, Inc., West Chester, PA (250 mm*30 mm, 5 μm); mobile phase: [0.1% NH3H2O ETOH]; B %: 42%-42%, 10 min) and prep-HPLC (neutral condition, column: XTIMATE® (Welch Materials, Hurst, TX); C18 150*25 mm*5 μm; mobile phase: [water (10 mM NH4HCO3)-ACN]; B %: 15%-40%, 10 min. column: HUAPU C8 Extreme BDS 150*30 5 μm (Dalian Institute of Chemical Physics, CAS 457, Zhongshan, China); mobile phase: [water (10 mM NH4HCO3)-ACN]; B %: 20%-40%, 10 min) to obtain the title compd as a white solid (6.74 mg, 15.7 μmol, 5.80% yield, 100% purity) as a 62:37 mixture of isomers of unassigned absolute stereochemistry at the oxobicycloheptane, as indicated by the wavy bond for Compound 79 inFIG.1. LCMS theoretical m/z=430.3 [M+H]+, found 430.4. Compound 80: (2S)-2-((2S)-7-oxabicyclo[2.2.1]heptane-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Compound 79, and isolated as a 47:53 mixture of isomers of unassigned absolute stereochemistry at the oxobicycloheptane, as indicated by the wavy bond for Compound 80 inFIG.1. LCMS theoretical m/z=430.3 [M+H]+, found 430.2. Compound 81: (S)-2-(2-methyl-2-(tetrahydro-2H-pyran-4-yl)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2-methyl-2-(tetrahydro-2H-pyran-4-yl)propanoic acid using Procedures K and M with methyl (S)-2-(2-methyl-2-(tetrahydro-2H-pyran-4-yl)propanamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=460.3 [M+H]+, found 460.3. Compound 82: (2S)-2-(1-(tert-butoxycarbonyl)-3,3-difluoropiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)-3,3-difluoropiperidine-4-carboxylic acid using Procedures K and M with tert-butyl 3,3-difluoro-4-(((S)-1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)piperidine-1-carboxylate. LCMS theoretical m/z=553.3 [M+H]+, found 553.3. Compound 83: (2S)-2-((2R,6S)-2,6-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (2R,6S)-2,6-dimethyltetrahydro-2H-pyran-4-carboxylic acid using Procedures A and O with methyl (2S)-2-((2R,6S)-2,6-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=446.3 [M+H]+, found 446.3. Compound 84: (S)-2-((S)-2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2,2-dimethyltetrahydro-2H-pyran-4-carboxylic acid using Procedures K and M with methyl (2S)-2-(2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate to afford the title compd as the first eluting isomer of unassigned absolute stereochemistry at the 4-position of the 2,2-dimethyltetrahydro-2H-pyran, as indicated by the wavy bond for Compound 84 inFIG.1. LCMS theoretical m/z=446.3. [M+H]+, found 446.3. Compound 85: (S)-2-((R)-2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2,2-dimethyltetrahydro-2H-pyran-4-carboxylic acid using Procedures K and M with methyl (2S)-2-(2,2-dimethyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate to afford the title compd as the second eluting isomer of unassigned absolute stereochemistry at the 4-position of the 2,2-dimethyltetrahydro-2H-pyran, as indicated by the wavy bond for Compound 85 inFIG.1. LCMS theoretical m/z=446.3. [M+H]+, found 446.3. Compound 86: (S)-2-(1-(tert-butoxycarbonyl)-4-(trifluoromethyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)-4-(trifluoromethyl)piperidine-4-carboxylic acid using Procedures K and M with tert-butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-(trifluoromethyl)piperidine-1-carboxylate. LCMS theoretical m/z=585.3. [M+H]+, found 585.3. Compound 87: (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(2,2,6,6-tetramethyltetrahydro-2H-pyran-4-carboxamido)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2,2,6,6-tetramethyltetrahydro-2H-pyran-4-carboxylic acid using Procedures K and M with methyl (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(2,2,6,6-tetramethyltetrahydro-2H-pyran-4-carboxamido)nonanoate. LCMS theoretical m/z=474.3. [M+H]+, found 474.6. Compound 88: (S)-2-(1-(tert-butoxycarbonyl)-4-(2,2-difluoroethyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with 1-(tert-butoxycarbonyl)-4-(2,2-difluoroethyl)piperidine-4-carboxylic acid and methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate using Procedures K and M and tert-butyl (S)-4-(2,2-difluoroethyl)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)piperidine-1-carboxylate. LCMS theoretical m/z=581.3 [M+H]+, found 581.3. Compound 89: (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(3,4,5,6-tetrahydro-[1,1′-biphenyl]-2-ylcarboxamido)nonanoic acid. Prepared according to Scheme A beginning with 3,4,5,6-tetrahydro-[1,1′-biphenyl]-2-carboxylic acid and methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate using Procedures K and M with methyl (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(3,4,5,6-tetrahydro-[1,1′-biphenyl]-2-carboxamido)nonanoate. LCMS theoretical m/z=490.3 [M+H]+, found 490.3. Compound 90: (S)-2-(2-(pyridin-4-yl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 2-(pyridin-4-yl)acetic acid using Procedures K and M with methyl (S)-2-(2-(pyridin-4-yl)acetamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=425.2 [M+H]+, found 425.2. Compound 91: (S)-2-((S)-1-(phenylsulfonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (S)-1-(phenylsulfonyl)pyrrolidine-2-carboxylic acid using Procedures K and M with methyl (S)-2-((S)-1-(phenylsulfonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=543.3. [M+H]+, found 543.3. Compound 92: (S)-2-(((4-methyltetrahydro-2H-pyran-4-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme G beginning with Procedure E using methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-methyltetrahydro-2H-pyran-4-carbaldehyde and Procedure M with methyl (S)-2-(((4-methyltetrahydro-2H-pyran-4-yl)methyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=418.3 [M+H]+, found 418.3. Compound 93: (S)-2-(((R)-1-(pyridin-3-yl)ethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme G beginning with Procedure E using methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(pyridin-3-yl)ethanone and Procedure M with methyl (2S)-2-((1-(pyridin-3-yl)ethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate to afford the title compd as a 74:26 mixture of diastereomers of unassigned absolute stereochemistry at the alpha-methyl pyridyl center, as indicated by the wavy bond for Compound 93 inFIG.1. LCMS theoretical m/z=411.3 [M+H]+, found 411.2. Compound 94: (S)-2-(((S)-1-(pyridin-3-yl)ethyl)amino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Compound 93 to afford the title compd as a 35:65 mixture of diastereomers of unassigned absolute stereochemistry at the alpha-methyl pyridyl center, as indicated by the wavy bond for Compound 94 inFIG.1. LCMS theoretical m/z=411.3 [M+H]+, found 411.2. Compound 95: (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(((1,3,5-trimethyl-1H-pyrazol-4-yl)methyl)amino)nonanoic acid. Prepared according to Scheme G beginning with Procedure E using methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1,3,5-trimethyl-1H-pyrazole-4-carbaldehyde and Procedure M using methyl (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-(((1,3,5-trimethyl-1H-pyrazol-4-yl)methyl)amino)nonanoate. LCMS theoretical m/z=428.3 [M+H]+, found 428.2. Compound 96: (S)-2-((2S,6R)-2,6-Dimethylpiperidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme K beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (2S,6R)-2,6-dimethylpiperidine using Procedures N and M with methyl (S)-2-((2S,6R)-2,6-dimethylpiperidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=445.3 [M+H]+, found 445.2. Compound 97: (S)-2-((2S,5R)-2,5-dimethylpyrrolidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme K beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (2S,5R)-2,5-dimethylpyrrolidine in Procedures N and M with methyl (S)-2-((2S,5R)-2,5-dimethylpyrrolidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=431.3. [M+H]+, found 431.2. Compound 98: (S)-2-((2R,5R)-2,5-dimethylpyrrolidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme K beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (2R,5R)-2,5-dimethylpyrrolidine using Procedures N and M with methyl (S)-2-((2R,5R)-2,5-dimethylpyrrolidine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=431.3. [M+H]+, found 431.3. Compound 99: (S)-2-((3R,5R)-3,5-dimethylmorpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme K beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (3R,5R)-3,5-dimethylmorpholine in Procedures N and M with methyl (S)-2-((3R,5R)-3,5-dimethylmorpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=447.3 [M+H]+, found 447.3. Compound 100: (S)-2-((3R,5S)-3,5-dimethylmorpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme K beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (3R,5R)-3,5-dimethylmorpholine in Procedures N and M with methyl (S)-2-((3R,5S)-3,5-dimethylmorpholine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=447.3 [M+H]+, found 447.3. tert-butyl (3R,5S)-4-(((S)-1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-3,5-dimethylpiperazine-1-carboxylate. To a mixture of tert-butyl (3S,5R)-3,5-dimethylpiperazine-1-carboxylate; hydrochloride (200 mg, 800 μmol) in THF (2 mL) and DMF (2 mL) was added CDI (130 mg, 800 μmol) and TEA (250 mg, 2.4 mmol) at 0° C. The reaction mixture was stirred at 30° C. under N2for 3 h. Methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (218 mg, 613 μmol) was added, and the reaction was stirred for 18 h. The aq phase was extracted with EA, dried over anhyd Na2SO4, concd, and purified by prep-TLC (EA:MeOH=10:1) to afford the title compd. LCMS theoretical m/z=560.4. [M+H]+, found 560.3. Methyl (S)-2-((2R,6S)-2,6-dimethylpiperazine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a mixture of tert-butyl (3R,5S)-4-(((S)-1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-3,5-dimethylpiperazine-1-carboxylate (247 mg, 441 μmol) in EA (1 mL) was added HCl/EA (4 M, 4.41 mL) at 0° C. The mixture was stirred at 0° C. for 1 h. The residue was concd in vacuum to yield 140 mg of the title compd as a crude yellow solid, which was used directly in the next reaction. LCMS theoretical m/z=460.3. [M+H]+, found 460.1. Methyl (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-((2R,6S)-2,4,6-trimethylpiperazine-1-carboxamido)nonanoate. To a mixture of methyl (S)-2-((2R,6S)-2,6-dimethylpiperazine-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (140 mg, 280 μmol, HCl) in DMF (2 mL) was added K2CO3(78 mg, 560 μmol) and methyl iodide (60 mg, 420 μmol) at 0° C. under N2. The mixture was stirred at 0° C. for 1 h. The aq phase was extracted with EA, dried with anhyd Na2SO4, filtered, and concd in vacuum. The residue was purified by prep-TLC (EA:MeOH=10:1) to afford 44 mg of the title compd as a colorless oil. LCMS theoretical m/z=474.3. [M+H]+, found 474.3. Compound 101: (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-((2R,6S)-2,4,6-trimethylpiperazine-1-carboxamido)nonanoic acid. Prepared according to Procedure M using methyl (S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-((2R,6S)-2,4,6-trimethylpiperazine-1-carboxamido)nonanoate to afford the title compd as a white solid. 400 MHz 1H NMR, methanol-d4, δ ppm 7.43 (d, J=7.34 Hz, 1H) 6.49 (d, J=7.34 Hz, 1H) 4.23 (t, J=5.81 Hz, 1H) 4.04-4.16 (m, 2H) 3.40-3.51 (m, 2H) 2.70-2.82 (m, 4H) 2.64 (t, J=7.64 Hz, 2H) 2.31 (s, 3H) 2.10-2.20 (m, 2H) 1.88-1.98 (m, 2H) 1.71-1.87 (m, 2H) 1.67 (br d, J=6.60 Hz, 2H) 1.29-1.41 (m, 14H). LCMS theoretical m/z=460.3. [M+H]+, found 460.3. Compound 102: (2S)-2-(3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme E beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 3-azabicyclo[3.3.1]nonane-9-carboxylic acid using Procedures K and M with methyl (2S)-2-(3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=457.6 [M+H]+, found 457.3. Compound 103: (S)-2-((1R,5S,9S)-3-acetyl-3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Procedure I using methyl (2S)-2-(3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate, and Procedure C with methyl (2S)-2-(3-acetyl-3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate to afford the title compd as the first eluting isomer; absolute stereochemistry at the amide carbon was not assigned, as indicated by the wavy bond for Compound 103 inFIG.1. LCMS theoretical m/z=499.3 m/z [M+H]+, found 499.3. 400 MHz 1H NMR, methanol-d4, δ ppm 7.40-7.48 (m, 1H) 6.52 (d, J=7.28 Hz, 1H) 4.54-4.64 (m, 1H) 4.30-4.40 (m, 1H) 4.02 (br d, J=13.45 Hz, 1H) 3.42-3.51 (m, 3H) 2.90-3.02 (m, 1H) 2.78 (t, J=6.17 Hz, 2H) 2.59-2.67 (m, 3H) 2.33 (br s, 2H) 2.12 (s, 3H) 2.10-2.14 (m, 1H) 1.55-2.03 (m, 11H) 1.55-2.03 (m, 1H) 1.30-1.46 (m, 9H). Compound 104: (S)-2-((1R,5S,9R)-3-acetyl-3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Procedure C with methyl (2S)-2-(cis-3-acetyl-3-azabicyclo[3.3.1]nonane-9-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate afforded the title compd as the second eluting isomer; absolute stereochemistry at the amide carbon was not assigned, as indicated by the wavy bond for Compound 104 inFIG.1. LCMS theoretical m/z=499.3 m/z [M+H]+, found 499.3. 400 MHz1H NMR, methanol-d4, δ ppm 7.45 (dd, J=7.28, 5.07 Hz, 1H) 6.51 (br d, J=4.63 Hz, 1H) 4.29 (br d, J=13.01 Hz, 2H) 3.67-3.85 (m, 2H) 3.42-3.50 (m, 2H) 3.13-3.28 (m, 1H) 2.78 (br t, J=6.06 Hz, 2H) 2.64 (br t, J=7.61 Hz, 2H) 2.57 (br s, 1H) 2.21-2.36 (m, 2H) 2.07 (d, J=2.65 Hz, 3H) 1.59-2.00 (m, 11H) 1.47-1.57 (m, 1H) 1.36 (br s, 8H). Compound 105: (S)-2-(4-methyl-1-((1-methyl-1H-pyrazol-4-yl)methyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme E using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)-4-methylpiperidine-4-carboxylic acid, Procedure H with (S)-2-(1-(tert-butoxycarbonyl)-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid, Procedure J with (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid and 1-methyl-1H-pyrazole-4-carbaldehyde, and Procedure C with methyl (S)-2-(4-methyl-1-((1-methyl-1H-pyrazol-4-yl)methyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=525.3 [M+H]+, found 525.4. Compound 106: (S)-2-(4-((tert-butoxycarbonyl)amino)bicyclo[2.2.2]octane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-((tert-butoxycarbonyl)amino)bicyclo[2.2.2]octane-1-carboxylic acid, and Procedure C with methyl (S)-2-(4-((tert-butoxycarbonyl)amino)bicyclo[2.2.2]octane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=557.4 [M+H]+, found 557.3. Compound 107: (2S)-2-(adamantane-1-carbonylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (3r,5r,7r)-adamantane-1-carboxylic acid, and Procedure C with methyl (S)-2-((3S,5S,7S)-adamantane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=468.3 [M+H]+, found 468.3. Compound 108: (S)-2-(4-((tert-butoxycarbonyl)amino)-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-((tert-butoxycarbonyl)amino)-1-methylcyclohexane-1-carboxylic acid, and Procedure C with methyl (S)-2-(4-((tert-butoxycarbonyl)amino)-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=545.4 [M+H]+, found 545.3. Compound 109: (S)-2-(4-amino-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Procedure H with (S)-2-(4-((tert-butoxycarbonyl)amino)-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. LCMS theoretical m/z=445.3 [M+H]+, found 445.3. Compound 110: (S)-2-(4-aminobicyclo[2.2.2]octane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Procedure H with (S)-2-(4-((tert-butoxycarbonyl)amino)bicyclo[2.2.2]octane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. LCMS theoretical m/z=457.3 [M+H]+, found 457.3. Compound 111: (S)-2-(4-acetamido-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared using Procedure I with (S)-2-(4-amino-1-methylcyclohexane-1-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. LCMS theoretical m/z=487.3 [M+H]+, found 487.3. Compound 112: (S)-2-((S)-5,5-dimethyl-3-(phenylsulfonyl)thiazolidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (S)-5,5-dimethyl-3-(phenylsulfonyl)thiazolidine-4-carboxylic acid, and using Procedure C with methyl (S)-2-((S)-5,5-dimethyl-3-(phenylsulfonyl)thiazolidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=589.2 [M+H]+, found 589.2. Compound 113: (R)-2-((S)-1-(phenylsulfonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and (phenylsulfonyl)-L-proline, and using Procedure C with methyl (S)-2-((S)-1-(phenylsulfonyl)pyrrolidine-2-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical=543.3 [M+H]+, found 543.3. Compound 114: (S)-2-(4-methyl-1-(3,3,3-trifluoropropyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme E using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)-4-methylpiperidine-4-carboxylic acid, Procedure H using tert-butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-methylpiperidine-1-carboxylate, Procedure J using methyl (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 3,3,3-trifluoropropanal, and Procedure C using methyl (S)-2-(4-methyl-1-(3,3,3-trifluoropropyl)piperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=527.3 [M+H]+, found 527.3. Compound 115: (2S)-2-[(1-acetyl-4-methyl-piperidine-4-carbonyl)amino]-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme E using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)-4-methylpiperidine-4-carboxylic acid, Procedure H using tert-butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-methylpiperidine-1-carboxylate, Procedure I using methyl (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and acetic anhydride, and Procedure C using methyl (S)-2-(1-acetyl-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=473.3 [M+H]+, found 473.3. Compound 116: (S)-2-(4-methyl-1-pivaloylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme E using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)-4-methylpiperidine-4-carboxylic acid, Procedure H using tert-butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-methylpiperidine-1-carboxylate, Procedure I using methyl (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and pivaloyl chloride, and Procedure C using methyl (S)-2-(4-methyl-1-pivaloylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=515.4 [M+H]+, found 515.3. methyl (S)-2-(1-(3-fluoropropyl)-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. To a solution of methyl (S)-2-(4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (99.99 mg, 0.225 mmol, 1.0 equiv) in ACN (0.5 mL) was added cesium carbonate (146.56 mg, 0.450 mmol, 2 equiv) and 1-bromo-3-fluoropropane (30 μL, 0.337 mmol, 1.5 equiv). The solution was stirred at rt for 24 h, at which time LCMS showed partial conversion. The reaction was heated to 50° C. for 1 h, at which time LCMS showed complete conversion. The reaction was diluted in water (10 mL) and extracted with EA (3×10 mL). The combined organic layers were dried over Na2SO4, filtered, and concd. The crude material was used directly into next reaction. Compound 117: (S)-2-(1-(3-fluoropropyl)-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme E using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 1-(tert-butoxycarbonyl)-4-methylpiperidine-4-carboxylic acid, Procedure H using tert-butyl (S)-4-((1-methoxy-1-oxo-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonan-2-yl)carbamoyl)-4-methylpiperidine-1-carboxylate, and Procedure C using methyl (S)-2-(1-(3-fluoropropyl)-4-methylpiperidine-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoat. LCMS theoretical m/z=491.3 [M+H]+, found 491.3. Compound 118: (S)-2-(4-(hydroxymethyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A using Procedure A with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-(hydroxymethyl)tetrahydro-2H-pyran-4-carboxylic acid, and using Procedure C with methyl (S)-2-(4-(hydroxymethyl)tetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate. LCMS theoretical m/z=448.3 [M+H]+, found 448.2. 7-Ethoxy-4,4-difluoro-7-oxoheptanoic acid. A solution of diester (5.00 g, 19.8 mmol) in ethanol was cooled to 0° C.; a solution of KOH (1.22 g, 21.8 mmol) in ethanol was added slowly to the reaction mixture. The resulting solution was warmed to rt and stirred for 10 h. The reaction mixture was concd, diluted with water, and extracted with hexanes:EA (3:1). The aq phase was acidified with 1N HCl and extracted by EA. The organic phases were combined and dried over Na2SO4, filtered, and concd to afford 2.88 g of the title compd as a white solid (65% yield). LCMS (ESI+): m/z=225.21 [M+H]+. Ethyl 4,4-difluoro-7-hydroxyheptanoate. To a cooled solution of acid (2.88 g, 12.8 mmol) in THF in an ice bath was added BH3/THF solution. After the addition, the reaction mixture was stirred at rt for 15 h. The reaction was treated with MeOH followed by water, extracted with EA, concd, and purified by FCC (hexanes:EA=2:1) to afford the title compd. LCMS (ESI+): m/z=211.127 [M+H]+. Ethyl 4,4-difluoro-7-oxoheptanoate. To a solution of alcohol (1.1 g, 5.2 mmol) in CH2C12(20 mL) at rt was added Dess-Martin Periodinane (2.7 g, 6.3 mmol), and the resulting mixture was stirred for an additional 2 h at rt. The reaction mixture was treated with a sat aq Na2S2O3solution followed by slow addition of sat aq solution of NaHCO3solution. The organic phase was separated, and the aq phase was extracted by DCM. The combined organic layers were washed with brine, dried over Na2SO4, filtered through a silica pad, and concd in vacuo to give the title compd as a light-yellow oil, which was used directly in the next reaction. 400 MHz1H NMR, CDCl3, δ 9.82 (t, J=1.0 Hz, 1H), 4.16 (q, J=7.2 Hz, 2H), 2.82-2.60 (m, 2H), 2.62-2.39 (m, 2H), 2.36-2.04 (m, 4H), 1.27 (t, J=7.1 Hz, 3H). Ethyl (E)-4,4-difluoro-9-oxodec-7-enoate. A mixture of ethyl 4,4-difluoro-7-oxoheptanoate (1.08 g, 5.20 mmol) and 1-(triphenylphosphoranylidene)-2-propanone (1.99 g, 6.24 mmol) in DMF (10 mL) was heated to 80° C. for 10 h. After cooling to rt, the reaction mixture was diluted with H2O and extracted with EA. The combined organic layers were washed with brine, dried with anhyd Na2SO4, filtered, and concd in vacuo. The residue was purified by FCC (hexanes:EA=3:1) to afford the title compd as a clear oil. LCMS (ESI+): m/z=249.2 [M+H]+. 400 MHz1H NMR, CDCl3, δ 6.79 (dt, J=16.0, 6.8 Hz, 1H), 6.11 (dt, J=15.9, 1.6 Hz, 1H), 4.25-4.04 (m, 2H), 2.59-2.49 (m, 2H), 2.46 (dtd, J=9.8, 6.6, 1.6 Hz, 2H), 2.33-2.14 (m, 5H), 2.12-1.91 (m, 2H), 1.36-1.16 (m, 3H). Ethyl 4,4-difluoro-9-oxodecanoate. A flask containing ethyl-4,4-difluoro-9-oxodec-7-enoate (2.17 g, 8.72 mmol) was charged 10 wt % Pd/C (244 mg), which was then diluted with MeOH (30 mL). The flask was evacuated and backfilled with H2for three cycles before stirring under an H2atmosphere overnight. The reaction mixture was filtered through a pad of Celite and concd in vacuo. The crude residue was purified by FCC to afford the title compd as a clear oil. LCMS (ESI+): m/z=251.1 [M+H]+. 400 MHz1H NMR, CDCl3, δ 4.15 (d, J=7.2 Hz, 2H), 2.48 (dt, J=20.4, 7.4 Hz, 4H), 2.28-2.08 (m, 5H), 1.95-1.73 (m, 2H), 1.67-1.54 (m, 2H), 1.53-1.39 (m, 2H), 1.26 (t, J=7.2 Hz, 3H). Ethyl 4,4-difluoro-8-(1,8-naphthyridin-2-yl)octanoate. To a mixture of ethyl 4,4-difluoro-9-oxodecanoate (2.18 g, 8.70 mmol) and 2-aminopyridine-3-carbaldehyde (1.17 g, 9.57 mmol) in EtOH (20 mL) was added L-proline (501 mg, 4.35 mmol). The mixture was refluxed at 85° C. for 12 h. The mixture was concd and purified by FCC (hexanes:EA=1:1 to 1:3) to give ethyl 4,4-difluoro-8-(1,8-naphthyridin-2-yl)octanoate (1.36 g, 46% yield) as a yellow solid. LCMS (ESI+): m/z=337.1 [M+H]+. 400 MHz1H NMR, CDCl3, δ 9.09 (dd, J=4.2, 2.0 Hz, 1H), 8.16 (dd, J=8.1, 2.0 Hz, 1H), 8.11 (d, J=8.3 Hz, 1H), 7.45 (dd, J=8.1, 4.2 Hz, 1H), 7.38 (d, J=8.2 Hz, 1H), 4.14 (q, J=7.2 Hz, 2H), 3.12-3.00 (m, 2H), 2.56-2.41 (m, 2H), 2.17 (tdd, J=16.6, 8.9, 6.8 Hz, 2H), 2.06-1.82 (m, 4H), 1.70-1.53 (m, 2H), 1.26 (t, J=7.1 Hz, 3H). Ethyl 4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octanoate. To a flask containing ethyl 4,4-difluoro-8-(1,8-naphthyridin-2-yl)octanoate (1.36 g, 4.04 mmol) was charged 20 wt % Pd(OH)2/C (57 mg. 0.40 mmol), and the reaction mixture was treated with MeOH (15 mL). The flask was evacuated and backfilled with H2for three cycles then stirred under an H2atmosphere overnight. The reaction mixture was filtered through a pad of Celite and concd in vacuo. The crude residue was purified by FCC (hexanes:EA) to give ethyl 4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octanoate as a clear oil. LCMS (ESI+): m/z=341.142 [M+H]+. 4,4-Difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octanal. To a cooled solution of ethyl 8-(8-acetyl-5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-4,4-difluorooctanoate (830 mg, 2.17 mmol) in THF (15 mL) in an ice bath was added LiBH4/THF solution (4.34 mmol). After addition, the reaction mixture was allowed to stir at 0° C. for 2 h before treating with sat aq NH4Cl. The mixture was filtered, and the filtrate was extracted with EA. The combined organic layers were dried over sodium sulfate and concd to provide the title compd. LCMS (ESI+): m/z=299.1 [M+H]+. 400 MHz 1H NMR, CDCl3, δ 7.08 (d, J=7.3 Hz, 1H), 6.33 (d, J=7.2 Hz, 1H), 3.67 (t, J=6.3 Hz, 4H), 3.48-3.35 (m, 2H), 2.71 (dt, J=12.6, 6.5 Hz, 2H), 2.58 (t, J=7.7 Hz, 2H), 1.99-1.79 (m, 6H), 1.72 (tt, J=16.1, 7.1 Hz, 3H), 1.61-1.46 (m, 3H), 1.23 (s, 9H). 4,4-Difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octanal. To a solution cooled to −78° C., of DMSO (116 μL, 1.63 mmol) in DCM (3 mL) was added oxalyl chloride (72 μL, 0.82 mmol) slowly, and the mixture was stirred for 15 min. Then a solution of 4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octan-1-ol (163 mg, 0.550 mmol) in DCM (1 mL) was added followed by Et3N (0.46 mL, 3.3 mmol). The reaction mixture was allowed to warm up to 0° C. and was treated with a sat aq NaHCO3solution. The organic phase was separated and the aq phase was extracted with EA. The combined organic layers were dried over sodium sulfate and concd to provide the title compd as a light yellow oil. 400 MHz1H NMR, CDCl3, δ 9.81 (s, 1H), 7.10 (d, J=7.3 Hz, 1H), 6.31 (d, J=7.4 Hz, 1H), 3.49-3.33 (m, 3H), 2.76-2.52 (m, 4H), 1.88 (tt, J=16.3, 7.5 Hz, 5H), 1.70 (q, J=7.7 Hz, 2H), 1.33-1.07 (m, 2H). Compound 119: (S)-5,5-difluoro-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. A solution of 2,5-dioxopyrrolidin-1-yl 4-methyltetrahydro-2H-pyran-4-carboxylate (6.0 mg, 22 μmol) generated using Procedure S, (S)-2-amino-5,5-difluoro-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid (5.0 mg, 15 μmol), and NaHCO3(7.0 mg, 73 μmol) in a mixed solvent of water:ACN (1:3, 1 mL) was heated to 50° C. for 2 h. The reaction mixture was cooled to rt and purified by prep-reverse phase HPLC to afford (S)-5,5-difluoro-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. LCMS (ESI+): m/z=468.2 [M+H]+. 400 MHz1H NMR, methanol-d4, δ 7.84 (d, J=8.0 Hz, 1H), 7.61 (dt, J=7.3, 1.3 Hz, 1H), 6.65 (d, J=7.4 Hz, 1H), 4.45 (td, J=8.3, 7.7, 4.8 Hz, 1H), 3.77 (dt, J=11.9, 4.3 Hz, 2H), 3.66-3.45 (m, 4H), 2.84 (t, J=6.3 Hz, 2H), 2.74 (t, J=7.7 Hz, 2H), 2.06 (s, 4H), 2.02-1.83 (m, 6H), 1.76 (p, J=7.7 Hz, 2H), 1.67-1.44 (m, 4H), 1.25 (s, 3H). Compound 120: (R)-5,5-difluoro-2-(4-methyltetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Procedure Q with 4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octanal, Procedure R with (R)—N—((R)-1-cyano-4,4-difluoro-8-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)octyl)-2-methylpropane-2-sulfinamide, and Procedure P with (S)-2-amino-5,5-difluoro-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid and 2,5-dioxopyrrolidin-1-yl 4-methyltetrahydro-2H-pyran-4-carboxylate generated from Procedure S using 4-methyltetrahydro-2H-pyran-4-carboxylic acid to afford the title compd. LCMS theoretical m/z=468.3 [M+H]+, found 468.3. 400 MHz1H NMR, methanol-d4, δ 7.83 (d, J=8.0 Hz, 1H), 7.61 (d, J=7.3 Hz, 1H), 6.64 (d, J=7.4 Hz, 1H), 4.45 (td, J=8.4, 7.8, 4.9 Hz, 1H), 3.77 (dt, J=11.7, 4.3 Hz, 2H), 3.66-3.45 (m, 4H), 2.84 (t, J=6.2 Hz, 2H), 2.74 (t, J=7.7 Hz, 2H), 2.20-2.04 (m, 4H), 1.95 (tq, J=14.6, 5.1, 3.7 Hz, 6H), 1.76 (p, J=7.7 Hz, 2H), 1.64-1.45 (m, 4H), 1.25 (s, 3H). Compound 121: (S)-5,5-difluoro-2-(quinazolin-4-ylamino)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. To a solution of (S)-2-amino-5,5-difluoro-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid (5 mg, 0.01 mmol) in IPA (0.5 mL) was added 4-chloroquinazoline (4 mg, 0.02 mmol). The reaction was stirred at 50 C for 1 h, concd, and purified by prep-HPLC to afford the title compd. LCMS theoretical m/z=470.3 [M+H]+, found 470.3. Compound 122 and 123: To a solution of methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate (66 mg, 0.21 mmol) and 2,2,2-trifluoro-1-(tetrahydro-2H-pyran-4-yl)ethan-1-one (45 mg, 0.25 mmol) in DCM (1 mL) was added NaBH3CN (16 mg, 0.25 mmol), and the reaction mixture was stirred overnight. Compound 122: (2S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-[[(1S)-2,2,2-trifluoro-1-tetrahydropyran-4-yl-ethyl]amino]nonanoic acid. The above reaction mixture was concd and purified by reverse phase prep-HPLC to afford the title compd as the first eluting isomer; absolute stereochemistry at the trifluoromethyl stereocenter was not assigned, as indicated by the wavy bond for compound 123 inFIG.1. LCMS theoretical m/z=472.3 [M+H]+, found 472.3. Compound 123: (2S)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-2-[[(1R)-2,2,2-trifluoro-1-tetrahydropyran-4-yl-ethyl]amino]nonanoic acid. The above reaction mixture was concd and purified by reverse phase prep-HPLC to afford the title compd as the second eluting isomer; absolute stereochemistry at the trifluoromethyl stereocenter was not assigned, as indicated by the wavy bond for compound 123 inFIG.1. LCMS theoretical m/z=472.3 [M+H]+, found 472.3. Compound 124: (S)-2-(4-cyanotetrahydro-2H-pyran-4-carboxamido)-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoic acid. Prepared according to Scheme A beginning with methyl (S)-2-amino-9-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)nonanoate and 4-cyanotetrahydro-2H-pyran-4-carboxylic acid using Procedures A and C. LCMS theoretical m/z=442.3. [M+H]+, found 443.2. BIOLOGICAL EXAMPLES Example B1—Solid Phase Integrin αVβ6Binding Assay Microplates were coated with recombinant human integrin αVβ6(2 μg/mL) in PBS (100 μL/well 25° C., overnight). The coating solution was removed, washed with wash buffer (0.05% Tween 20; 0.5 mM MnCl2; in 1×TBS). Plate was blocked with 200 μL/well of Block Buffer (1% BSA; 5% sucrose; 0.5 mM MnCl2; in 1×TBS) at 37° C. for 2 h. Dilutions of testing compounds and recombinant TGFβ1 LAP (0.67 μg/mL) in binding buffer (0.05% BSA; 2.5% sucrose; 0.5 mM MnCl2; in 1×TBS) were added. The plate was incubated for 2 hours at 25° C., washed, and incubated for 1 hour with Biotin-Anti-hLAP. Bound antibody was detected by peroxidase-conjugated streptavidin. The IC50values for testing compounds were calculated by a four-parameter logistic regression. The IC50values obtained for αVβ6integrin inhibition for a first series of exemplary compounds are shown in Table B-1. The compounds tested were compound samples prepared according to procedures described in the Synthetic Examples section, with the stereochemical purity as indicated in the Examples. TABLE B-1αvβ6Inhibition IC50Compound No.(nM) - range8<5024251-100030251-100032<503450-2503650-25037<5038251-100039<5042<5043<5044<5045<504650-2504750-250 Example B2—The Disclosed Compounds Potently Inhibit αVβ6in a Solid Phase Assay A second series of exemplary compounds was selected for testing in the solid phase integrin αVβ6binding assay. The compounds tested were compound samples prepared according to procedures described in the Synthetic Examples section, with the stereochemical purity as indicated in the Examples. As in Example B1, microplates were coated with recombinant human integrin αVβ6(2 μg/mL) in PBS (100 μL/well 25° C., overnight). The coating solution was removed, washed with wash buffer (0.05% Tween 20; 0.5 mM MnCl2; in 1×TBS). The plate was blocked with 200 μL/well of Block Buffer (1% BSA; 5% sucrose; 0.5 mM MnCl2; in 1×TBS) at 37° C. for 2 h. Dilutions of testing compounds and recombinant TGFβ1LAP (0.67 μg/mL) in binding buffer (0.05% BSA; 2.5% sucrose; 0.5 mM MnCl2; in 1×TBS) were added. The plate was incubated for 2 hours at 25° C., washed, and incubated for 1 hour with Biotin-Anti-hLAP. Bound antibody was detected by peroxidase-conjugated streptavidin. The IC50values for tested compounds were calculated by a four-parameter logistic regression. Example B3—The Disclosed Compounds Potently Inhibit αVβ1in a Solid Phase Assay The first and second series of exemplary compounds were tested in a solid phase integrin αVβ1binding assay. The compounds tested were compound samples prepared according to procedures described in the Synthetic Examples section, with the stereochemical purity as indicated in the Examples. Similar to Examples B1 and B2, microplates were coated with recombinant human integrin αVβ1(2 μg/mL) in PBS (100 μL/well 25° C., overnight). The coating solution was removed, washed with wash buffer (0.05% Tween 20; 0.5 mM MnCl2; in 1×TBS). The plate was blocked with 200 μL/well of Block Buffer (1% BSA; 5% sucrose; 0.5 mM MnCl2; in 1×TBS) at 37° C. for 2 h. Dilutions of testing compounds and recombinant TGFβ1LAP (0.67 μg/mL) in binding buffer (0.05% BSA; 2.5% sucrose; 0.5 mM MnCl2; in 1×TBS) were added. The plate was incubated for 2 hours at 25° C., washed, and incubated for 1 hour with Biotin-Anti-hLAP. Bound antibody was detected by peroxidase-conjugated streptavidin. The IC50values for tested compounds were calculated by a four-parameter logistic regression. Example B4—The Disclosed Compounds Potently Inhibit Human αVβ6Integrin The first and second series of exemplary compounds were tested for αVβ6integrin biochemical potency using the ALPHASCREEN® (Perkin Elmer, Waltham, MA) proximity-based assay (a bead-based, non-radioactive Amplified Luminescent Proximity Homogeneous Assay) as described previously (Ullman E F et al., Luminescent oxygen channeling immunoassay: Measurement of particle binding kinetics by chemiluminescence. Proc. Natl. Acad. Sci. USA, Vol. 91, pp. 5426-5430, June 1994). To gauge the potency of inhibitors of binding to human integrin αVβ6, inhibitor compounds and integrin were incubated together with recombinant TGFβ1LAP and biotinylated anti-LAP antibody plus acceptor and donor beads, following the manufacturer's recommendations. The donor beads were coated with streptavidin. The acceptor beads had a nitrilotriacetic acid Ni chelator, for binding to a 6×His-tag on human integrin αVβ6. All incubations occurred at room temperatures in 50 mM Tris-HCl, pH 7.5, 0.1% BSA supplemented with 1 mM each CaCl2and MgCl2. The order of reagent addition was as follows: 1. αVβ6integrin, test inhibitor compound, LAP, biotinylated anti-LAP antibody and acceptor beads were all added together. 2. After 2 hours, donor beads were added. After another 30 min incubation, samples were read. Integrin binding was evaluated by exciting donor beads at 680 nm, and measuring the fluorescent signal produced, between 520-620 nm, using a Biotek Instruments (Winooski, VT, USA) SynergyNeo2 multimode plate reader. Compound potency was assessed by determining inhibitor concentrations required to reduce fluorescent light output by 50%. Data analysis for IC50determinations was carried out by nonlinear four parameter logistic regression analysis using Dotmatics ELN Software (Core Informatics Inc., Branford, Ct). Example B5—The Disclosed Compounds Potently Inhibit Human αVβ1Integrin The first and second series of exemplary compounds were tested for αVβ1integrin biochemical potency using the ALPHASCREEN® proximity-based assay as described in Example B4. To gauge the potency of inhibitors of binding to human integrin αvβ1, inhibitor compounds and integrin were incubated together with biotinylated, purified human fibronectin plus acceptor and donor beads, following the manufacturer's recommendations. The donor beads were coated with streptavidin. The acceptor beads had a nitrilotriacetic acid Ni chelator, for binding to a 6×His-tag on human integrin αvβ1. All incubations occurred at room temperatures in 50 mM Tris-HCl, pH 7.5, 0.1% BSA supplemented with 1 mM each CaCl2) and MgCl2. The order of reagent addition was as follows: 1. αVβ1integrin, test inhibitor compound, fibronectin-biotinylated and acceptor beads were all added together. 2. After 2 hours, donor beads were added. After another 30 min incubation, samples were read. Integrin binding was evaluated by exciting donor beads at 680 nm, and measuring the fluorescent signal produced, between 520-620 nm, using a Biotek Instruments (Winooski, VT, USA) SynergyNeo2 multimode plate reader. Compound potency was assessed by determining inhibitor concentrations required to reduce fluorescent light output by 50%. Data analysis for IC50determinations was carried out by nonlinear four parameter logistic regression analysis using Dotmatics ELN Software (Core Informatics Inc., Branford, Ct). Combined Inhibition Results of Examples B1, B2, B3, B4, and B5 Table B-2,FIG.2, shows IC50data from Examples B1, B2, B3, B4, and B5 for inhibition of αVβ1and αVβ6integrin in the solid phase assays and inhibition of human αVβ1and αVβ6integrin in the proximity-based ALPHASCREEN® assays. The IC50data is shown in four ranges: below 50 nM; from 50 nM to below 250 nM; from above 250 nM to below 1000 nM; and 1000 nM and above. All references throughout, such as publications, patents, patent applications and published patent applications, are incorporated herein by reference in their entireties. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is apparent to those skilled in the art that certain minor changes and modifications will be practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention.
257,613
11858932
DETAILED DESCRIPTION While various embodiments of the invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions may occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed. Polycystic Ovary Syndrome (PCOS) Polycystic ovarian syndrome (PCOS) is a heterogeneous disorder with multiple phenotypes and is one of the most common endocrine metabolic disorders in reproductive aged women. PCOS was first diagnosed and described as a syndrome of oligo-amenorrhea and polycystic ovaries that was variably accompanied by hirsutism, acne, and obesity. There are various diagnostic criteria for adolescent and adult patients considering the core diagnostic features, such as hyperandrogenism, persistent ovulatory dysfunction, and polycystic ovarian morphology (PCOM). The exact etiology of this disorder is not entirely clear, however, both genetic and environmental factors seem to play roles in causing the disorder. Generally, PCOS presents as a phenotype reflecting a self-perpetuating vicious cycle involving neuroendocrine, metabolic, and ovarian dysfunction and is characterized by excessive ovarian and/or adrenal androgen secretion. For example, intrinsic ovarian factors such as altered steroidogenesis and factors external to the ovary such as hyperinsulinemia contribute to the excessive ovarian androgen production. The classic ovarian phenotype of enlarged ovaries with string-of-pearl morphology and theca interstitial hyperplasia reflects androgen exposure; this morphology has also been observed in women with congenital adrenal hyperplasia (CAH). In order to attempt to understand the pathophysiology of PCOS, it is important to understand the molecular basis of steroidogenesis and androgen source, production and physiology. Under normal circumstances in women, the ovaries and adrenal glands contribute about equally to testosterone production. Approximately half of testosterone originates from direct testosterone secretion by the ovaries and adrenal glands, whereas half is produced by peripheral conversion of circulating androstenedione, which itself arises from approximately equal ovarian and adrenal secretion. Androgen production is not under direct negative feedback regulation by the neuroendocrine system in females, as is the case for estradiol and cortisol secretion. Androgens are secreted by both the ovaries and adrenal glands in response to their respective tropic hormones, LH and ACTH. The zonareticularisof the adrenal gland resembles the theca cell compartment of the ovary in its expression of the core enzymatic pattern for androgen production. PCOS patients can be functionally categorized as distinct subtypes based on the source of androgen excess listed in Table 1 below. Androgen excess can be derived from the ovaries, the adrenal glands, both, or neither. Primary PCOS is the most common variety of PCOS without any known cause and a proposed theory suggests that functional ovarian hyperandrogenism (FOH) along with disturbance of hypothalamo-pituitary-ovarian (HPO) axis function may be the underlying cause. Further, functional adrenal hyperandrogenism (FAH) other than HPO axis disturbance may be explained for a certain population of PCOS patients. The PCOS patients with primary FAH may experience an average 50% increase in adrenal volume that correlates with hyperandrogenemia severity. TABLE 1Functional classification of PCOS based on source of androgen excessPCOSGrRHagDASTACTH testPrevalenceFunctionalTest 17OHPTestosteroneDHEAAmongTypeSource of AndrogenResponseResponseResponsePCOSPCOS-TPrimary FOR (TypicalHighaHigh inHigh in 28%67%*FOH)92.5%(associated FAH)PCOS-APrimary FOH (atypicalNormalaHighHigh in 30%20%FOH)(associated FAH)Primary FAH (isolatedNormalNormalHigh5%FAH) PCOS withoutNormalNormalNormal8%FOH or FAH (PCOS-Aof obesity or idiopathicPCOS-A) Test procedures to determine the source of androgen are listed below in Table 2. The ovarian hyperandrogenism of PCOS is demonstrated directly by the GnRHag test or the hCG test and in combination with the dexamethasone androgen-suppression test (DAST). The GnRHag test determines the coordinated function of the ovarian follicle. Leuprolide acetate 10 μg/kg sc (or a comparable dose of any other short-acting GnRHag) stimulates endogenous LH and FSH release that peaks at 3-4 hours and persists for 24 hours; this in turn stimulates increased secretion of sex steroids and their precursors, with serum levels peaking at 18-24 hours. In the absence of evidence of a steroidogenic block, an elevated 17OHP response is typical of PCOS. Ovarian steroidogenic enzyme deficiency, which is rare, can be detected by an abnormal pattern of steroid intermediates in response to the test. hCG is an LH analog: 5000 IU intramuscularly stimulates steroidogenic responses comparable with those of a GnRHag test at 24 hours. DAST indirectly tests for ovarian source of androgens by suppressing ACTH-dependent androgen production of androgens. In the presence of normal adrenocortical suppression an inappropriately elevated serum testosterone post-DAST indicates an ACTH-independent source of androgen, which is ordinarily of ovarian origin. Adrenal hyperandrogenism may be demonstrated by a rapid ACTH test: cosyntropin is administered iv and peak steroid responses occur at 15-60 minutes. An elevated DHEA post this test indicates an ACTH-dependent source of androgen, which is ordinarily of adrenal origin. The test may be performed using cosyntropin 250 μg, this is a supramaximal dose. Lower doses of cosyntropin may be used (10 μg/m2) which elicit a similar peak response. A low-dose ACTH test (1.0-μg cosyntropin) may be used and may be more physiologic. It usually elicits nearly as great a peak response that promptly wanes, and in PCOS, does not elicit such a wide spectrum of elevated steroid intermediates as do larger doses. TEST 2Methods to determine source of female androgen excessTestRationaleMethodOutcome MeasuresInterpretationaGARHagEndogenous LH and FSHLeuprolide somiste 10 μg/kgOvarian sieroky secretion17OHP >152 ng/dLrelease stimulates coordinatedsc (for maximum stimulation)peaks at 20-24 hwithout steroidonenicfunction of ovarian folliclesblock indicatestypical FOH (PCOS-T)hCGExogenous administration ofNCG 3000 IU/m2(forOvarian steroid secretion17OHP >152 ng/dLLH analog stiroulates theca-maximum stimulation)peaks at 24 hwithout steroidogenicinterstitial cellsblock indicatestypical FOH (PCOS-T)IDASTLong DAST: dexamethasoneDexametasona 0.5 mg QIDFree testosterone.Free testosterone ≥8profoundly suppresses adrenalper os × 4-5 dDHEAS. cortisol: samplepg/mL with DMEAS <70androgens over several daysearly morning d 5and characteristic of FOHSDASTShort DAST. dexamethistoneDexamethasone 0.25 mg/m2Total testosterone,Total testosterone >26rapidly suppresses adrenalper os at 12 nooncortisol: sample 4 PMng/mL, cortisol <5 μg/dLtestosterone and cortisol(4 h)suggests FOHACTHExogenous ACTH stimulatesCosyntropin ≥10 μg/m2(forDHEA, 17OHP, steroidDHEA 1500-3000 μg/dLadrenal steroidogenesismaximum stimulation)intermediates, cortisolwithout sterontogenicpeak at 30-60 minblock indicates FAH Corticotropin Releasing Factor Currently, no single universal treatment for PCOS is available. As a result, current treatments tend to be individualized and adapted to the actual needs of the individual patient. Furthermore, treatment may be symptom-oriented. For example, targets for pharmacological treatment may include androgen excess. For PCOS patients with elevated adrenal androgens, FOH+FAH and PCOS-FAH patients, in some embodiments, pharmacological treatment, such as corticotropin releasing factor (CRF) receptor antagonist targeting ACTH production may be used. Corticotropin releasing factor (CRF) is a 41 amino acid peptide that is the primary physiological regulator of proopiomelanocortin (POMC) derived peptide secretion from the anterior pituitary gland. In addition to its endocrine role at the pituitary gland, immunohistochemical localization of CRF has demonstrated that the hormone has a broad extrahypothalamic distribution in the central nervous system and produces a wide spectrum of autonomic, electrophysiological and behavioral effects consistent with a neurotransmitter or neuromodulator role in the brain. There is also evidence that CRF plays a significant role in integrating the response in the immune system to physiological, psychological, and immunological stressors. CRF has been implicated in psychiatric disorders and neurological diseases including depression and anxiety, as well as the following: Alzheimer's disease, Huntington's disease, progressive supranuclear palsy, amyotrophic lateral sclerosis, Parkinson's disease, epilepsy, migraine, alcohol and substance abuse and associated withdrawal symptoms, obesity, metabolic syndrome, congenital adrenal hyperplasia (CAH), Cushing's disease, hypertension, stroke, irritable bowel syndrome, stress-induced gastric ulceration, premenstrual syndrome, sexual dysfunction, premature labor, inflammatory disorders, allergies, multiple sclerosis, visceral pain, sleep disorders, pituitary tumors or ectopic pituitary derived tumors, chronic fatigue syndrome, and fibromyalgia. CRF is believed to be the major physiological regulator of the basal and stress-induced release of adrenocorticotropic hormone (“ACTH”), β-endorphin, and other proopiomelanocortin (“POMC”)-derived peptides from the anterior pituitary. Secretion of CRF causes release of ACTH from corticotrophs in the anterior pituitary via binding to the CRF1receptor, a member of the class B family of G-protein coupled receptors. Due to the physiological significance of CRF1, the development of biologically-active small molecules having significant CRF receptor binding activity and which are capable of antagonizing the CRF1receptor remains a desirable goal and has been the subject of ongoing research and development for the treatment of anxiety, depression, irritable bowel syndrome, post-traumatic stress disorder, and substance abuse, and congenital adrenal hyperplasia. The pituitary hormone ACTH, under the control of hypothalamic corticotropin-releasing factor (CRF), stimulates uptake of cholesterol and drives the synthesis of pregnenolone initiating steroidogenesis in the adrenal gland. The adrenal cortex is comprised of three zones, which produce distinct classes of hormones many of which are driven by ACTH mobilizing cholesterol through this pathway. The middle layer, thezona reticularisis responsible for the production of androgens such as DHEA, DHEAS and androstenedione, (A4) the precursor to testosterone (T) and dihydrotestosterone (DHT). This layer is also responsible for the production of the 11-oxyandrogens, 11β-hydroxyandrostenedione (11OHA4) and 11β-hydroxytestosterone (11OHT), which are major bioactive androgens, particularly in women. Due to an unknown cause in PCOS patients with elevated androgens, thezone reticularisdisplays a hyperresponsiveness to ACTH resulting excessive levels of DHEA, DHEAS, 11OHA4, 11OHT and A4. The excessive ACTH stimulation causes hypertrophy of thezona reticularisresulting in adrenal hyperplasia and clinically manifests with physical features typical of PCOS and hyperandrogenism. Reducing the stimulatory effect on the pituitary's ACTH secretion by inhibiting the CRF1 receptor is expected is reduce excessive androgen synthesis from the ACTH responsive zone of the adrenal gland in PCOS. The normalization of elevated androgen levels in the short term would be expected to ameliorate cardinal features of PCOS—hirsutism, acne and menstrual irregularities—over the long term. Certain Definitions Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments described herein, certain preferred methods, devices, and materials are now described. As used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to “an excipient” is a reference to one or more excipients and equivalents thereof known to those skilled in the art, and so forth. The term “about” is used to indicate that a value includes 10% level of error for the device or method being employed to determine the value. The use of the term “or” in the claims is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and to “and/or.” “Alkyl” refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, and preferably having from one to five carbon atoms (i.e. C1-C5alkyl). In some embodiments, an alkyl comprises one to four carbon atoms (i.e., C1-C4alkyl). ). In some embodiments, an alkyl comprises one to three carbon atoms (i.e., C1-C3alkyl). ). In some embodiments, an alkyl comprises one to two carbon atoms (i.e., C1-C2alkyl). In some embodiments, an alkyl comprises one carbon atom (i.e., C1alkyl). In certain embodiments, the alkyl group is selected from methyl, ethyl, 1-propyl (n-propyl), 1-methylethyl (iso-propyl), 1-butyl (n-butyl), 1-methylpropyl (sec-butyl), 2-methylpropyl (isobutyl), 1,1-dimethylethyl (tert-butyl), or 1-pentyl (n-pentyl). The alkyl is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more substituents such as those described herein. The terms “comprise,” “have” and “include” are open-ended linking verbs. Any forms or tenses of one or more of these verbs, such as “comprises,” “comprising,” “has,” “having,” “includes” and “including,” are also open-ended. For example, any method that “comprises,” “has” or “includes” one or more steps is not limited to possessing only those one or more steps and also covers other unlisted steps. “Administering” when used in conjunction with a therapeutic means to administer a therapeutic systemically or locally, as directly into or onto a target tissue, or to administer a therapeutic to a patient whereby the therapeutic positively impacts the tissue to which it is targeted. “Administering” a pharmaceutical composition may be accomplished by injection, topical administration, and oral administration or by other methods alone or in combination with other known techniques. By “pharmaceutically acceptable”, it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the composition and not deleterious to the recipient thereof. The term “pharmaceutical composition” means a composition comprising at least one active ingredient, such as a steroid or a pharmaceutically acceptable salt thereof or a CRF1antagonist or a pharmaceutically acceptable salt thereof, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human). Those of ordinary skill in the art will understand and appreciate the techniques appropriate for determining whether an active ingredient has a desired efficacious outcome based upon the needs of the artisan. The term “supraphysiologic amount” describes glucocorticoid dose levels that are above the daily glucocorticoid requirement (production rate) found in healthy individuals. The term “physiologic amount” describes glucocorticoid dose levels that meet the daily glucocorticoid requirement (production rate) found in healthy individuals. The term “hydrocortisone equivalents” as used herein is understood by a person skilled in the art to be the conversion calculations needed to be considered when substituting one glucocorticoid for another as the potency and duration of action of various glucocorticoids may vary. Hence, the term “hydrocortisone equivalents” is the standard used for comparison of glucocorticoid potency. A “therapeutically effective amount” or “effective amount” as used herein refers to the amount of active compound or pharmaceutical agent that elicits a biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes one or more of the following: (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease, (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology), and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology). The terms “treat,” “treated,” “treatment,” or “treating” as used herein refers to both therapeutic treatment in some embodiments and prophylactic or preventative measures in other embodiments, wherein the object is to prevent or slow (lessen) an undesired physiological condition, disorder or disease, or to obtain beneficial or desired clinical results. For the purposes described herein, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of the condition, disorder or disease; stabilization (i.e., not worsening) of the state of the condition, disorder or disease; delay in onset or slowing of the progression of the condition, disorder or disease; amelioration of the condition, disorder or disease state; and remission (whether partial or total), whether detectable or undetectable, or enhancement or improvement of the condition, disorder or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment. A prophylactic benefit of treatment includes prevention of a condition, retarding the progress of a condition, stabilization of a condition, or decreasing the likelihood of occurrence of a condition. As used herein, “treat,” “treated,” “treatment,” or “treating” includes prophylaxis in some embodiments. Compounds Disclosed herein are CRF1antagonists or pharmaceutically acceptable salt thereof such as Antalarmin hydrochloride, CP-154,526, CP-376395 hydrochloride, NBI 27914 hydrochloride, NBI 35965 hydrochloride, NGD 98-2 hydrochloride, Pexacerfont, R 121919 hydrochloride, SN003, and SSR125543 (crinecerfont). In one aspect, the CRF1antagonist or pharmaceutically acceptable salt thereof is selected from the group consisting of: n-butyl-N-ethyl-2,5,6-trimethyl-7-(2,4,6-trimethylphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine hydrochloride (Antalarmin hydrochloride), n-butyl-N-ethyl-2,5-dimethyl-7-(2,4,6-trimethylphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine hydrochloride (Pfizer CP154526), N-(1-Ethylpropyl)-3,6-dimethyl-2-(2,4,6-trimethylphenox y)-4-pyridinamine hydrochloride (Pfizer CP376395 hydrochloride), 5-Chloro-N-(cyclopropylmethyl)-2-methyl-n-propyl-N′-(2,4,6-trichlorophenyl)-4,6-pyrimidinediamine hydrochloride (NBI27914 hydrochloride), (7S)-6-(Cyclopropylmethyl)-2-(2,4-dichlorophenyl)-7-ethyl-7,8-dihydro-4-methyl-6H-1,3,6,8a-tetraazaacenaphthylene hydrochloride (NBI35965 hydrochloride), N-(1-Ethylpropyl)-3-methoxy-5-[2-methoxy-4-(trifluoromethoxy)phenyl]-6-methyl-2-pyrazinamine hydrochloride (NGD 98-2 hydrochloride), 8-(6-Methoxy-2-methyl-3-pyridinyl)-2,7-dimethyl-N-[(1R)-1-methylpropyl]pyrazolo[1,5-a]-1,3,5-triazin-4-amine (Pexacerfont), 3-[6-(Dimethylamino)-4-methyl-3-pyridinyl]-2,5-dimethyl-N,N-dipropylpyrazolo[1,5-a]pyrimidin-7-amine hydrochloride (R 121919 hydrochloride), and N-(4-Methoxy-2-methylphenyl)-1-[1-(methoxymethyl)propyl]-6-methyl-1H-1,2,3-triazolo[4,5-c]pyridine-4-amine (SN003), (S)-4-(2-chloro-4-methoxy-5-methylphenyl)-N-(2-cyclopropyl-1-(3-fluoro-4-methylphenyl)ethyl)-5-methyl-N-(prop-2-yn-1-yl)thiazol-2-amine (SSR125543). Disclosed herein is a compound of Formula (I): or a pharmaceutically acceptable salt thereof, wherein R1and R2are independently ethyl or n-propyl; R3is H, Cl, Br, methyl, trifluoromethyl, or methoxy; R4is H, Br, —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, and Raand Rbare independently hydrogen, C1-C3alkyl, —CH2CH2NH2, —CH2CH2NHC(O)OC(CH3)3, or —CH2CH2NHCH2CH2CH3. In some embodiments, R1and R2are independently ethyl or n-propyl. In some embodiments, R1is ethyl and R2is n-propyl. In some embodiments, R1is n-propyl and R2is ethyl. In some embodiments, R1and R2are both ethyl. In some embodiments, R1and R2are both n-propyl. In some embodiments, R3is hydrogen, Cl, Br, methyl, or trifluoromethyl. In some embodiments, R3is hydrogen, Cl, Br, or methyl. In some embodiments, R3is hydrogen, Cl, or Br. In some embodiments, R3is hydrogen. In some embodiments, R3is Cl. In some embodiments, R3is Br. In some embodiments, R4is hydrogen, Br, —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, In some embodiments, R4is Br, —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, In some embodiments, R4is —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, or In some embodiments, R4is —NRaRb, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, or In some embodiments, R4is —NRaRb, pyridin-4-yl, morpholin-4-yl, or In some embodiments, R4is morpholin-4-yl or In some embodiments, R4is morpholin-4-yl. In some embodiments, R4is In some embodiments, R4is —NRaRband Raand Rbare independently C1-C3alkyl. Disclosed herein is a compound of Formula (II): or a pharmaceutically acceptable salt thereof, wherein R3is H, Cl, Br, methyl, trifluoromethyl, or methoxy; R4is H, Br, —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, and Raand Rbare independently hydrogen, C1-C3alkyl, —CH2CH2NH2, —CH2CH2NHC(O)OC(CH3)3, or —CH2CH2NHCH2CH2CH3—CH2CH2NHCH2CH2CH3. Disclosed herein is a compound of Formula (III): or a pharmaceutically acceptable salt thereof, wherein R1and R2are n-propyl; R3is H, Cl, Br, methyl, trifluoromethyl, or methoxy; R4is H, Br, —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, and Raand Rbare independently hydrogen, C1-C3alkyl, —CH2CH2NH2, —CH2CH2NHC(O)OC(CH3)3, or —CH2CH2NHCH2CH2CH3. Disclosed herein is a compound of Formula (IV): or a pharmaceutically acceptable salt thereof, wherein R3is Cl, Br, methyl, trifluoromethyl, or methoxy; R4is H, Br, —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, and Raand Rbare independently hydrogen, C1-C3alkyl, —CH2CH2NH2, —CH2CH2NHC(O)OC(CH3)3, or —CH2CH2NHCH2CH2CH3. For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R3may be hydrogen, Cl, Br, methyl, trifluoromethyl, or methoxy. For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R3may be Cl, Br, methyl, trifluoromethyl, or methoxy. For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R3may be Cl, Br, methyl, or trifluoromethyl. For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R3may be Cl, Br, or methyl. For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R3may be Cl. For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R3may be Br. For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R3may be methyl. For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is Br, —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is Br, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is Br, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is Br, acetamido, pyridin-4-yl, morpholin-4-yl, For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is Br, pyridin-4-yl, morpholin-4-yl, For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is pyridin-4-yl, morpholin-4-yl, For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is morpholin-4-yl For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is For a compound or pharmaceutically acceptable salt of Formula (I), (II), (III), and (IV), R4is morpholin-4-yl. Disclosed herein is 3-[4-bromo-2-(2-methyl-2H-[1,2,4]triazol-3-yl)-thiazol-5-yl]-2,5-dimethyl-7-(1-propyl-butyl)-pyrazolo[1,5-a]pyrimidine (Compound 1) or a pharmaceutically acceptable salt thereof: Disclosed herein is 3-(4-bromo-2-(2-methyl-2H-[1,2,4]triazol-3-yl)-thiazol-5-yl)-7-(1-ethyl-propyl)-2,5-dimethyl-pyrazolo[1,5-a]pyrimidine (Compound 2) or a pharmaceutically acceptable salt thereof: Disclosed herein is 3-(4-Chloro-2-(morpholin-4-yl)thiazol-5-yl)-7-(1-ethylpropyl)-2,5-dimethylpyrazolo(1,5-a)pyrimidine (or alternatively 4-(4-chloro-5-(2,5-dimethyl-7-(pentan-3-yl)pyrazolo[1,5-a]pyrimidin-3-yl)thiazol-2-yl)morpholine) (Compound 3) or a pharmaceutically acceptable salt thereof: In some embodiments, 4-(4-chloro-5-(2,5-dimethyl-7-(pentan-3-yl)pyrazolo[1,5-a]pyrimidin-3-yl)thiazol-2-yl)morpholine is referred to as Compound 3. In some embodiments, 3-(4-Chloro-2-(morpholin-4-yl)thiazol-5-yl)-7-(1-ethylpropyl)-2,5-dimethylpyrazolo(1,5-a)pyrimidine is referred to as Compound 3. In one aspect, the CRF1antagonist or pharmaceutically acceptable salt thereof may be an astressin. An astressin generally refers to a nonselective corticotropin releasing hormone antagonist that reduces the synthesis of ACTH and cortisol. Pharmaceutical Compositions Disclosed herein are pharmaceutical compositions comprising a compound or pharmaceutically acceptable salt described herein. In certain embodiments, the composition comprises a steroid or a pharmaceutically acceptable salt thereof and a CRF1antagonist or pharmaceutically acceptable salt thereof as disclosed above. In some embodiments, the steroid or a pharmaceutically acceptable salt thereof is exogenous glucocorticoids (GC) or a pharmaceutically acceptable salt thereof. Dosage Form In some embodiments, the pharmaceutical compositions described herein are provided in unit dosage form. As used herein, a “unit dosage form” is a composition containing an amount of a compound or pharmaceutically acceptable salt described herein that is suitable for administration to an animal, preferably mammal, subject in a single dose, according to good medical practice. The preparation of a single or unit dosage form however, does not imply that the dosage form is administered once per day or once per course of therapy. Such dosage forms are contemplated to be administered once, twice, thrice or more per day and may be administered as infusion over a period of time (e.g., from about 30 minutes to about 2-6 hours), or administered as a continuous infusion, and may be given more than once during a course of therapy, though a single administration is not specifically excluded. Pharmaceutical compositions are administered in a manner appropriate to the disease to be treated (or prevented). An appropriate dose and a suitable duration and frequency of administration will be determined by such factors as the condition of the patient, the type and severity of the patient's disease, the particular form of the active ingredient, and the method of administration. In general, an appropriate dose and treatment regimen provides the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (e.g., an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity. Optimal doses are generally determined using experimental models and/or clinical trials. The optimal dose depends upon the body mass, weight, or blood volume of the patient. In some embodiments, the pharmaceutical compositions described herein are formulated as oral dosage forms. Suitable oral dosage forms include, for example, tablets, pills, sachets, or capsules. In some embodiments, the pharmaceutical composition comprises one or more additional pharmaceutically acceptable excipients. See, e.g., Remington: The Science and Practice of Pharmacy (Gennaro, 21st Ed. Mack Pub. Co., Easton, PA (2005) for a list of pharmaceutically acceptable excipients. Capsule In some embodiments, the pharmaceutical composition is formulated as a capsule. In some embodiments, the pharmaceutical composition is formulated as a hard gel capsule. In some embodiments, the pharmaceutical composition is formulated as a soft gel capsule. In some embodiments, the capsule is formed using materials which include, but are not limited to, natural or synthetic gelatin, pectin, casein, collagen, protein, modified starch, polyvinylpyrrolidone, acrylic polymers, cellulose derivatives, or any combinations thereof. In some embodiments, the capsule is formed using preservatives, coloring and opacifying agents, flavorings and sweeteners, sugars, gastroresistant substances, or any combinations thereof. In some embodiments, the capsule is coated. In some embodiments, the coating covering the capsule includes, but is not limited to, immediate release coatings, protective coatings, enteric or delayed release coatings, sustained release coatings, barrier coatings, seal coatings, or combinations thereof. In some embodiments, a capsule herein is hard or soft. In some embodiments, the capsule is seamless. In some embodiments, the capsule is broken such that the particulates are sprinkled on soft foods and swallowed without chewing. In some embodiments, the shape and size of the capsule also vary. Examples of capsule shapes include, but are not limited to, round, oval, tubular, oblong, twist off, or a non-standard shape. The size of the capsule may vary according to the volume of the particulates. In some embodiments, the size of the capsule is adjusted based on the volume of the particulates and powders. Hard or soft gelatin capsules may be manufactured in accordance with conventional methods as a single body unit comprising the standard capsule shape. A single-body soft gelatin capsule typically may be provided, for example, in sizes from 3 to 22 minims (1 minims being equal to 0.0616 ml) and in shapes of oval, oblong or others. The gelatin capsule may also be manufactured in accordance with conventional methods, for example, as a two-piece hard gelatin capsule, sealed or unsealed, typically in standard shape and various standard sizes, conventionally designated as (000), (00), (0), (1), (2), (3), (4), and (5). The largest number corresponds to the smallest size. In some embodiments, the pharmaceutical composition described herein (e.g., capsule) is swallowed as a whole. In some embodiments, the capsule comprises one or more pharmaceutically acceptable excipients. In some embodiments, the capsule is free of additional excipients. In some embodiments, a capsule is developed, manufactured and commercialized for a drug substance that is insoluble. In some embodiments, a drug substance is insoluble if solubility is less than 0.002 mg/mL in water. In some embodiments, the capsule has a dose strength of up to 200 mg. In some embodiments, drug substance in the capsule is immediately released in a dissolution medium using USP apparatus I. In some embodiments, drug substance in the capsule is immediately released in a dissolution medium using USP apparatus II. Tablet Poorly soluble drugs may be difficult to formulate using standard technologies such as high shear wet granulation. Optimum delivery of poorly soluble drugs may require complex technologies such as solid solutions amorphous dispersions (hot melt extrusion or spray drying), nano-formulations or lipid-based formulations. Hydrophobic drug substances may be considered poorly soluble according to USP criteria and may be known to be difficult to granulate with water and other excipients. This is likely due to most known excipients for immediate release formulations being water soluble or water-swellable. Making a tablet of a high dose drug substance that is poorly soluble may require a high concentration of the drug substance. However, as the drug concentration is increased above a certain level, formation of granules may become more and more difficult. Furthermore, at a certain drug load, it may become impossible. In some embodiments, the pharmaceutical composition is formulated as a tablet. In some embodiments, the tablet is made by compression, molding, or extrusion, optionally with one or more pharmaceutically acceptable excipient. In some embodiments, compressed tablets are prepared by compressing a compound or pharmaceutically acceptable salt described herein in a free-flowing form, optionally mixed with pharmaceutically acceptable excipients. In some embodiments, molded tablets are made by molding a mixture of the powdered a compound or pharmaceutically acceptable salt described herein, moistened with an inert liquid diluent. In some embodiments, the tablet is prepared by hot-melt extrusion. In some embodiments, extruded tablets are made by forcing a mixture comprising a compound or pharmaceutically acceptable salt described herein, through an orifice or die under controlled conditions. In some embodiments, the tablet is coated or scored. In some embodiments, the tablet is formulated so as to provide slow or controlled release of a compound or pharmaceutically acceptable salt described herein. In some embodiments, a tablet is developed, manufactured and commercialized for a drug substance that is insoluble. In some embodiments, a drug substance is insoluble if solubility is less than 0.002 mg/mL in water. In some embodiments, the tablet has a dose strength of up to 200 mg. In some embodiments, drug substance in the tablet is immediately released in a dissolution medium using USP apparatus I. In some embodiments, drug substance in the tablet is immediately released in a dissolution medium using USP apparatus II. In some embodiments, the tablet size is less than about 1000 mg, less than about 800 mg, less than about 600 mg, less than about 400 mg, less than about 200 mg, less than about 100 mg or less than 50 mg. In some embodiments, the tablet has a dose strength of more than about 10 mg, more than about 50 mg, more than about 100 mg, more than about 150 mg, more than about 200 mg, or more than about 250 mg. In some embodiments, the tablet size is less than about 1000 mg for a dose strength of more than about 50 mg. In some embodiments, the tablet size is less than 800 mg for a dose strength of more than about 100 mg. In some embodiments, the tablet size is less than 600 mg for a dose strength of more than about 150 mg. In some embodiment, the tablet size is less than 400 mg for a dose strength of more than about 200 mg. In some embodiments, the tablet size is less than 400 mg for a dose strength of 100 mg. In some embodiments, the tablet size is less than 200 mg for a dose strength of 50 mg. In some embodiments, the tablet size is less than 50 mg for a dose strength of 10 mg. In some embodiments, more than about 20% of the tablet is dissolved in conventional dissolution media. In some embodiments, more than about 40% of the tablet is dissolved in conventional dissolution media. In some embodiments, more than about 50% of the tablet is dissolved in conventional dissolution media. In some embodiments, more than about 60% of the tablet is dissolved in conventional dissolution media. In some embodiments, more than about 70% of the tablet is dissolved in conventional dissolution media. In some embodiments, more than about 80% of the tablet is dissolved in conventional dissolution media. In some embodiments, more than about 20% of the tablet is dissolved in less than 24 hours in conventional dissolution media. In some embodiments, more than about 20% of the tablet is dissolved in less than 12 hours in conventional dissolution media. In some embodiments, more than about 20% of the tablet is dissolved in less than 6 hours in conventional dissolution media. In some embodiments, more than about 20% of the tablet is dissolved in less than 3 hours in conventional dissolution media. In some embodiments, more than about 20% of the tablet is dissolved in less than 2 hours in conventional dissolution media. In some embodiments, more than about 20% of the tablet is dissolved in less than 60 minutes in conventional dissolution media. In some embodiments, more than about 40% of the tablet is dissolved in less than 60 minutes in conventional dissolution media. In some embodiments, more than about 50% of the tablet is dissolved in less than 60 minutes in conventional dissolution media. In some embodiments, more than about 60% of the tablet is dissolved in less than 60 minutes in conventional dissolution media. In some embodiments, more than about 70% of the tablet is dissolved in less than 60 minutes in conventional dissolution media. In some embodiments, more than about 80% of the tablet is dissolved in less than 60 minutes in conventional dissolution media. In some embodiments, more than about 90% of the tablet is dissolved in less than 60 minutes in conventional dissolution media. In some embodiments, more than about 100% of the tablet is dissolved in less than 60 minutes in conventional dissolution media. In some embodiments, the tablet is produced at a commercial scale. In some embodiments, the tablet comprises one or more pharmaceutically acceptable excipients. In some embodiments, the tablet is coated with a coating material, e.g., a sealant. In some embodiments, the coating material is water soluble. In some embodiments, the coating material comprises a polymer, plasticizer, a pigment, or any combination thereof. In some embodiments, the coating material is in the form of a film coating, e.g., a glossy film, a pH independent film coating, an aqueous film coating, a dry powder film coating (e.g., complete dry powder film coating), or any combination thereof. In some embodiments, the coating material is highly adhesive. In some embodiments, the coating material provides low level of water permeation. In some embodiments, the coating material provides oxygen barrier protection. In some embodiments, the coating material allows immediate disintegration for fast release of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the coating material is pigmented, clear, or white. In some embodiments, the coating is an enteric coating. Exemplary coating materials include, without limitation, polyvinylpyrrolidone, polyvinyl alcohol, an acrylate-methacrylic acid copolymer, a methacrylate-methacrylic acid copolymer, cellulose acetate phthalate, cellulose acetate succinate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, cellulose acetate trimellitate, sodium alginate, zein, and any combinations thereof. Pharmaceutically Acceptable Excipients In some embodiments, the pharmaceutical composition comprises a pharmaceutically acceptable excipient. In some embodiments, the composition is free of pharmaceutically acceptable excipients. The term “pharmaceutically acceptable excipient”, as used herein, means one or more compatible solid or encapsulating substances, which are suitable for administration to a mammal. The term “compatible”, as used herein, means that the components of the composition are capable of being commingled with the subject compound, and with each other, in a manner such that there is no interaction, which would substantially reduce the pharmaceutical efficacy of the composition under ordinary use situations. In some embodiments, the pharmaceutically acceptable excipient is of sufficiently high purity and sufficiently low toxicity to render them suitable for administration preferably to an animal, preferably mammal, being treated. Some examples of substances, which can serve as pharmaceutically acceptable excipients include:Amino acids such as alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine. In some embodiments, the amino acid is arginine. In some embodiments, the amino acid is L-arginine.Monosaccharides such as glucose (dextrose), arabinose, mannitol, fructose (levulose), and galactose.Cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose, and methyl cellulose.Solid lubricants such as talc, stearic acid, magnesium stearate, and sodium stearyl fumarate.Polyols such as propylene glycol, glycerin, sorbitol, mannitol, and polyethylene glycol.Emulsifiers such as the polysorbates.Wetting agents such as sodium lauryl sulfate, Tween*, Span, alkyl sulphates, and alkyl ethoxylate sulphates.Diluents such as calcium carbonate, microcrystalline cellulose, calcium phosphate, starch, pregelatinized starch, sodium carbonate, mannitol, and lactose.Binders such as starches (corn starch and potato starch), gelatin, sucrose, hydroxypropyl cellulose (HPC), polyvinylpyrrolidone (PVP), and hydroxypropyl methyl cellulose (HPMC).Disintegrants such as starch, and alginic acid.Super-disintegrants such as ac-di-sol, croscarmellose sodium, sodium starch glycolate and crospovidone.Glidants such as silicon dioxide.Coloring agents such as the FD&C dyes.Sweeteners and flavoring agents, such as aspartame, saccharin, menthol, peppermint, and fruit flavors.Preservatives such as benzalkonium chloride, PHMB, chlorobutanol, thimerosal, phenylmercuric, acetate, phenylmercuric nitrate, parabens, and sodium benzoate.Tonicity adjustors such as sodium chloride, potassium chloride, mannitol, and glycerin.Antioxidants such as sodium bisulfite, acetone sodium bisulfite, sodium formaldehyde, sulfoxylate, thiourea, and EDTA.pH adjuster such as NaOH, sodium carbonate, sodium acetate, HCl, and citric acid.Cryoprotectants such as sodium or potassium phosphates, citric acid, tartaric acid, gelatin, and carbohydrates such as dextrose, mannitol, and dextran.Cationic surfactants such as cetrimide, benzalkonium chloride and cetylpyridinium chloride.Anion surfactants such as alkyl sulphates, alkyl ethoxylate sulphates, soaps, carboxylates, sulfates, and sulfonates.Non-ionic surfactants such as polyoxyethylene derivatives, polyoxypropylene derivatives, polyol derivatives, polyol esters, polyoxyethylene esters, poloxamers, glyol esters, glycerol esters, sorbitan derivatives, polyethylene glycol (PEG-40, PEG-50, PEG-55), and ethers of fatty alcohols.Organic materials such as carbohydrate and modified carbohydrates, lactose, a-lactose monohydrate, spray dried lactose and anhydrous lactose, starch and pre-gelatinized starch, sucrose, manitol, sorbitol, cellulose, powdered cellulose and microcrystalline cellulose.Inorganic materials such as calcium phosphates (anhydrous dibasic calcium phosphate, dibasic calcium phosphate and tribasic calcium phosphate).Co-processed diluents.Surfactants such as sodium lauryl sulfate.Compression aids.Anti-tacking agents such as silicon dioxide and talc Amounts In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 500 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 400 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 300 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 200 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 100 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 90 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 80 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 70 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 60 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 50 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 40 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 30 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 20 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 1 mg and about 10 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 500 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 400 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 300 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 200 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 100 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 90 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 80 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 70 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 60 mg of a compound or pharmaceutically acceptable salt described herein f. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 50 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 40 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 30 mg of a compound or pharmaceutically acceptable salt described herein. In some embodiments, the pharmaceutical composition, in the form of a tablet or capsule, comprises between about 10 mg and about 20 mg of a compound or pharmaceutically acceptable salt described herein. Particle Size In some embodiments, the pharmaceutical composition, in the form of a tablet or a capsule, comprises a compound or pharmaceutically acceptable salt described herein, in the form of microparticles. In some embodiments, the microparticles have an average size from about 1 μm to about 100 μm. In some embodiments, the microparticles have an average size from about 1 μm to about 50 μm. In some embodiments, the microparticles have an average size from about 1 μm to about 30 μm. In some embodiments, the microparticles have an average size from about 1 μm to about 20 μm. In some embodiments, the microparticles have an average size from about 5 μm to about 15 μm. In some embodiments, the microparticles have an average size from about 1 μm to about 10 μm. In some embodiments, the microparticles have an average size from about 3 μm to about 10 μm. In some embodiments, the microparticles have an average size from about 4 μm to about 9 μm. Methods of Treatment Disclosed herein are methods of treating polycystic ovary syndrome (PCOS) in a subject in need thereof, comprising administering a compound or pharmaceutically acceptable salt described herein. In some embodiments, the subject in need thereof has PCOS-FAH. In some embodiments, the subject in need thereof has PCOS−FOH+FAH. In some embodiments, the methods described herein result in the reduction of a level of a hormone. Such hormones include deoxycorticosterone, 11-deoxycortisol, cortisol, corticosterone, pregnenolone, 17α-hydroxy pregnenolone, progesterone, 17-OHP, dehydroepiandrosterone (DHEA), dehydroepiandrosterone-sulfate (DHEAS), androstenediol, androstenedione (A4), testosterone (T), dihydrotestosterone (DHT), estrone, estradiol, estriol, 11β-hydroxyandrostenedione (11OHA4), 11β-hydroxytestosterone (11OHT), 11-ketoandrostenedione (11KA4), 11-ketotestosterone (11KT), 11β-hydroxy-5α-androstenedione (11OHDHA4), 11-keto-5α-androstenedione (11KDHA4), 11β-hydroxydihydrotestosterone (11OHDHT), 11-ketodihydrotestosterone (11KDHT) and ACTH. In some embodiments, the methods described herein result in the reduction of 17-OHP levels. In some embodiments, the methods described herein result in the reduction of A4 levels. In some embodiments, the methods described herein result in the reduction of ACTH levels. In some embodiments, the methods described herein result in the reduction of DHEA levels. In some embodiments, the methods described herein result in the reduction of DHEAS levels. In some embodiments, the methods described herein result in the reduction of testosterone (T) levels. In some embodiments, the methods described herein result in the reduction of DHT levels. In some embodiments, the methods described herein result in the reduction of 11OHA4 levels. In some embodiments, the methods described herein result in the reduction of 11OHT levels. In some embodiments, the methods described herein result in the reduction of 11KA4 levels. In some embodiments, the methods described herein result in the reduction of 11KT levels. Further, in some embodiments, the methods described herein result in the maintenance of the reduction of 17-OHP, A4, ACTH, DHEA, DHEAS, T, DHT, 11OHA4, 11OHT, 11KA4, and/or 11KT levels. In some embodiments, the reductions of 17-OHP, A4, ACTH, DHEA, DHEAS, T, DHT, 11OHA4, 11OHT, 11KA4 and/or 11KT levels may last for at least 24 hours, 36 hours, 48 hours, 60 hours, 72 hours, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, 24 weeks, 25 weeks, 26 weeks, 27 weeks, 28 weeks, 29 weeks, 30 weeks, 31 weeks, 32 weeks, 33 weeks, 34 weeks, 35 weeks, 36 weeks, 37 weeks, 38 weeks, 39 weeks, 40 weeks, 41 weeks, 42 weeks, 43 weeks, 44 weeks, 45 weeks, 46 weeks, 47 weeks, 48 weeks, 49 weeks, 50 weeks, 51 weeks, 52 weeks, 2 years, 5 years, 10 years, 15 years, 20 years, 25 years, 30 years, 35 years, 40 years, 45 years, 50 years, 55 years, 60 years, 65 years, 70 years, 75 years, 80 years, 85 years, 90 years, 95 years, or 100 years. In some embodiments, the methods described herein result in the reduction of DHEAS levels. In some embodiments, the DHEAS level is reduced by at least about 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% from baseline. The baseline level of the DHEAS may be measured in a subject before beginning the treatment disclosed herein. In some embodiments, the DHEAS level is reduced by at least 5% from baseline. In some embodiments, the reduced DHEAS level from baseline may be maintained for at least about 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 24 hours, 48 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, 24 weeks, 25 weeks, 26 weeks, 27 weeks, 28 weeks, 29 weeks, or 30 weeks. Further, in some embodiments, the methods described herein result in the maintenance of the reduction of 17-OHP, A4, ACTH, T, DHT, 11OHA4, 11OHT, 11KA4 and/or 11KT levels. In some embodiments, the reductions of 17-OHP, A4, ACTH T, DHT, 11OHA4, 11OHT, 11KA4 and/or 11KT levels may last for at least 24 hours, 36 hours, 48 hours, 60 hours, 72 hours, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, 24 weeks, 25 weeks, 26 weeks, 27 weeks, 28 weeks, 29 weeks, 30 weeks, 31 weeks, 32 weeks, 33 weeks, 34 weeks, 35 weeks, 36 weeks, 37 weeks, 38 weeks, 39 weeks, 40 weeks, 41 weeks, 42 weeks, 43 weeks, 44 weeks, 45 weeks, 46 weeks, 47 weeks, 48 weeks, 49 weeks, 50 weeks, 51 weeks, 52 weeks, 2 years, 5 years, 10 years, 15 years, 20 years, 25 years, 30 years, 35 years, 40 years, 45 years, 50 years, 55 years, 60 years, 65 years, 70 years, 75 years, 80 years, 85 years, 90 years, 95 years, or 100 years. In some embodiments, disclosed herein are methods for treating PCOS in a subject in need thereof, and the methods further comprise accessing a source of excessive androgen; and administering a CRF1antagonist or pharmaceutically acceptable salt thereof. In some embodiments, accessing a source of excessive androgen comprises measuring the primary source of excessive androgen produced in the subject suffering PCOS. In some embodiments, the primary source is the ovaries. In some embodiments, the primary source is the adrenal glands. In some embodiments, the primary source is both the ovaries and the adrenal glands. In some embodiments, the primary source is an organ that is able to produce androgen. In some embodiments, cosyntropin or any other suitable hormones or chemical compounds may be administered to a subject for ACTH response in order to test for adrenal hyperandrogenism. In some embodiments, the cosyntropin is administered at a dose at least about 0.25, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 200, 250 or μg/m2. In some embodiments, the cosyntropin is administered at a dose at about the range between the doses disclosed herein. In some embodiments, the cosyntropin is administered at a dose at most about 250, 200, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25, 20, 15, 10, 5, 4, 3, 2, 1 μg/m2. In some embodiments, the subject will be treated for a period of about 1 week to about 40 weeks. In some embodiments, the subject will be treated for a period of about 2 weeks to about 39 weeks. In some embodiments, the subject will be treated for a period of about 3 weeks to about 38 weeks. In some embodiments, the subject will be treated for a period of about 4 weeks to about 36 weeks. In some embodiments, the subject will be treated for a period of about 1 month to 12 months. In some embodiments, the subject will be treated for a period of about 10 months to 10 years. In some embodiments, the subject will be treated for a period of about 1 months to 10 years. In some embodiments, the subject will be treated for a period of about 10 months to 20 years. In some embodiments, the subject will be treated for a period of about 1 months to 20 years. In some embodiments, the subject will be treated for a period of about 10 months to 30 years. In some embodiments, the subject will be treated for a period of about 1 months to 30 years. In some embodiments, the subject will be treated for a period of about 10 months to 40 years. In some embodiments, the subject will be treated for a period of about 1 months to 40 years. In some embodiments, the subject will be treated for a period of about 10 months to 50 years. In some embodiments, the subject will be treated for a period of about 1 months to 50 years. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt thereof is selected from the group consisting of: Antalarmin hydrochloride, CP-154,526, CP-376395 hydrochloride, NBI 27914 hydrochloride, NBI 35965 hydrochloride, NGD 98-2 hydrochloride, Pexacerfont, R 121919 hydrochloride, SSR125543 (crinecerfont), AND SN003. In some embodiments, the CRF1antagonist is a compound of Formula (I): or a pharmaceutically acceptable salt thereof, wherein: R1and R2are independently ethyl or n-propyl; R3is hydrogen, F, Cl, Br, methyl, trifluoromethyl, or methoxy; and R4is hydrogen, Br, —NRaRb, methoxymethyl, n-butyl, acetamido, pyridin-4-yl, morpholin-4-yl, and Raand Rbare independently hydrogen, C1-C3alkyl, —CH2CH2NH2, —CH2CH2NHC(O)OC(CH3)3—CH2CH2NHC(O)OC(CH3)3, or —CH2CH2NHCH2CH2CH3—. In some embodiments, R3is F, Cl, Br, methyl, or trifluoromethyl. In some embodiments, R3is Cl, Br, or methyl. In some embodiments, R4is Br, —NRaRb—NRaRb, pyridin-4-yl, morpholin-4-yl, or In some embodiments, R4is morpholin-4-yl or In some embodiments, R4is hydrogen, Br, —NRaRband Raand Rbare independently C1-C3alkyl. In some embodiments, the compound is or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is or a pharmaceutically acceptable salt thereof. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 10 mg to about 200 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 200 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 150 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 100 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 50 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 40 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 30 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 25 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 20 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 15 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 10 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is administered in a dose of about 5 mg total daily dose to the subject. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt is in the form of microparticles. In some embodiments, the average size of the microparticles is between about 1 μm to about 20 μm. In some embodiments, the average size of the microparticles is between about 5 μm to about 15 μm. In some embodiments, the average size of the microparticles is less than about 10μ. In some embodiments, the CRF1antagonist or pharmaceutically acceptable salt thereof is administered as a pharmaceutical composition. In some embodiments, the steroid or a pharmaceutically acceptable salt thereof is administered as a pharmaceutical composition. In some embodiments, the pharmaceutical composition is in the form of a capsule or a tablet. In some embodiments, the capsule is a hard gelatin capsule. In some embodiments, the capsule is a soft gelatin capsule. In some embodiments, the capsule is formed using materials selected from the group consisting of natural gelatin, synthetic gelatin, pectin, casein, collagen, protein, modified starch, polyvinylpyrrolidone, acrylic polymers, cellulose derivatives, and any combinations thereof. In some embodiments, the pharmaceutical composition is free of additional excipients. In some embodiments, the pharmaceutical composition further comprises one or more pharmaceutically acceptable excipients. In some embodiments, the pharmaceutical composition is in the form of a tablet. In some embodiments, the pharmaceutical composition further comprises one or more pharmaceutically acceptable excipients. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 1 to about 8 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 2 to about 7 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 2 to about 6 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 3 to about 5 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 8 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 7 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 6 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 5 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 4 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 3 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 2 hours in a subject. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is formulated as a capsule or a tablet as to provide a Tmax of about 1 hour in a subject. In some embodiments, the methods described herein include administration of the pharmaceutical composition comprising CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof once a month, twice a month, three times a month, once a week, twice a week, three times a week, once every two days, once a day, twice a day, three times a day, or four times a day. In some embodiments, the methods described herein administer CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof once a day. In some embodiments, the methods described herein administer CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof twice a day. In some embodiments, the methods described herein include administration of about 1 mg to about 2000 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, per day. In some embodiments, CRF1antagonist or pharmaceutically acceptable salt is administered at a dose between about 50 mg/day and about 1600 mg/day. In some embodiments, Compound 3 is administered at a dose between about 50 mg/day and about 1500 mg/day. In some embodiments, Compound 3 is administered at a dose between about 50 mg/day and about 1400 mg/day. In some embodiments, Compound 3 is administered at a dose between about 50 mg/day and about 1300 mg/day. In some embodiments, Compound 3 is administered at a dose between about 50 mg/day and about 1200 mg/day. In some embodiments, Compound 3 is administered at a dose between about 50 mg/day and about 1100 mg/day. In some embodiments, Compound 3 is administered at a dose between about 50 mg/day and about 1000 mg/day. In some embodiments, Compound 3 is administered at a dose between about 50 mg/day and about 900 mg/day. In some embodiments, Compound 3 is administered at a dose between about 50 mg/day and about 800 mg/day. In some embodiments, Compound 3 is administered at a dose between about 60 mg/day and about 800 mg/day. In some embodiments, Compound 3 is administered at a dose between about 70 mg/day and about 800 mg/day. In some embodiments, Compound 3 is administered at a dose between about 80 mg/day and about 800 mg/day. In some embodiments, Compound 3 is administered at a dose between about 90 mg/day and about 800 mg/day. In some embodiments, Compound 3 is administered at a dose between about 100 mg/day and about 800 mg/day. In some embodiments, Compound 3 is administered at a dose between about 100 mg/day and about 700 mg/day. In some embodiments, Compound 3 is administered at a dose between about 100 mg/day and about 600 mg/day. In some embodiments, Compound 3 is administered at a dose between 150 mg/day and about 600 mg/day. In some embodiments, Compound 3 is administered at a dose between 200 mg/day and about 600 mg/day. In some embodiments, Compound 3 is administered at a dose between 200 mg/day and about 500 mg/day. In some embodiments, Compound 3 is administered at a dose between 200 mg/day and about 400 mg/day. In some embodiments, Compound 3 is administered at a dose between 25 mg/day and about 200 mg/day. In some embodiments, Compound 3 is administered at a dose of about 500 mg/day. In some embodiments, Compound 3 is administered at a dose of about 400 mg/day. In some embodiments, Compound 3 is administered at a dose of about 300 mg/day. In some embodiments, Compound 3 is administered at a dose of about 200 mg/day. In some embodiments, Compound 3 is administered at a dose of about 150 mg/day. In some embodiments, Compound 3 is administered at a dose of about 100 mg/day. In some embodiments, Compound 3 is administered at a dose of about 50 mg/day. In some embodiments, about 50 mg to about 1600 mg of CRF1 antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 100 mg to about 1600 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 200 mg to about 1600 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 200 mg to about 1200 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 200 mg to about 1000 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 200 mg to about 800 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 100 mg to about 800 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 200 mg to about 800 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 100 mg to about 600 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 200 mg to about 600 mg of CRF1antagonist or pharmaceutically acceptable salt is administered per day. In some embodiments, about 300 mg to about 600 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 100 mg to about 400 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 200 mg to about 400 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, about 300 mg to about 400 mg of CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered each day. In some embodiments, less than about 2000 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 1800 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 1600 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 1400 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 1200 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 1000 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 800 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 600 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 500 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 400 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 300 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, less than about 200 mg CRF1antagonist or pharmaceutically acceptable salt, or a pharmaceutically acceptable salt or solvate thereof, is administered per day. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein wherein the subject is in the fed state. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein wherein the subject is in the fasted state. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein at bedtime. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein less than about 4 hours before sleep. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein less than about 3 hours before sleep. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein less than about 2 hours before sleep. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein less than about 1 hour before sleep. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein less than about 30 mins before sleep. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein in the evening. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein at about 11 μm at night. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein at about 10 μm at night. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein at about 9 μm at night. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein at about 8 μm at night. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein in combination with eflornithine and/or retinoids. In some embodiments, the methods described herein include administration of the pharmaceutical compositions described herein in combination with contraceptive pills containing androgen receptor blockers such as cyproterone acetate, spironolactone, flutamide, or 5α-reductase inhibitors. In some embodiments, the levels of DHEA, DHEAS, A4, 11OHA4, T, 11OHT, DHT and/or ACTH in the subject are determined from a biological sample from the subject. In some embodiments, the biological sample is selected from the group of blood, blood fractions, plasma, serum, urine, other types of bodily secretions, and saliva. In some embodiments, the biological sample is obtained non-invasively. In some embodiments, the subject is a pediatric patient. In some embodiments, the subject is an adolescent. In some embodiments, the subject is from about 8 years old to about 18 years old. In some embodiments, the subject is an adult patient. In some embodiments, the subject is from about 18 years old to about 55 years old. In some embodiments, the subject is from about 18 years old to about 50 years old. In some embodiments, the steroid and the CRF1antagonist are administered concurrently. In some embodiments, the steroid and the CRF1antagonist are administered in one pharmaceutical composition. In some embodiments, the steroid and the CRF1antagonist are administered concurrently in separate pharmaceutical compositions. In some embodiments, the steroid and the CRF1antagonist are administered sequentially. EXAMPLES The following examples further illustrate the invention but should not be construed as in any way limiting its scope. In particular, the processing conditions are merely exemplary and can be readily varied by one of ordinary skill in the art. All methods described herein can be performed in a suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. Unless defined otherwise, technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs. Example 1—Reduction of DHEAS Level in a Subject Who has Polycystic Ovary Syndrome (PCOS) and Elevated Adrenal Androgens Study Compound 3 in a double-blind, proof-of-concept study, evaluating the safety and efficacy of repeated doses of Compound 3 in adults with elevated adrenal androgens, PCOS-FAH or PCOS−FAH+FOH. After screening, eligible subjects with elevated adrenal androgens documented by elevated DHEAS will be randomized to either a compound 3 dose escalation arm or matching placebo arm. The study is planned as a 3-period study with a duration of 4 weeks for Periods 1 and 2 and a duration of 8 weeks in Period 3 for a total treatment duration of 16 weeks. Subjects randomized to the compound 3 dose escalation arm will be treated with escalating doses of 50-mg, 100-mg or 200 mg based on achieving a DHEAS positive response criteria within each treatment period. Subjects not meeting the criteria will be dose escalated to the next highest dose in the preceding period. Subjects meeting the criteria will remain on their current dose level. Placebo subjects will remain on placebo through the full 16 weeks. The population are composed of approximately 40 patients, who receive Compound 3 or matching placebo daily for up to 16 weeks. Compound 3 is administered as an oral daily dose. Patients are undergo PK/PD or PD only assessments, approximately every 2 weeks. A follow-up outpatient visit occurs 30 days after the last dose. Study DesignStudy Type: InterventionalPrimary Purpose: TreatmentStudy Phase: Phase 2Interventional Study Model: RandomizedNumber of Arms: 2Masking: Masking of Study drugAllocation: Randomized (1:1)Enrollment: 40 Arms and Interventions:ArmsAssigned InterventionArm 1Drug: Compound 3dose escalation (50, 100 and 200-mg)Arm 2Drug: Matching Placebo Outcome Measures Primary Outcome Measures: 1. Safety and tolerability of Compound 3 in patients with elevated adrenal androgens (adverse effects, serious adverse effects, clinical laboratory parameters, etc.) [Time Frame: 16 weeks] Secondary Outcome Measures: 2. Proportion of subjects with:a. ≥30% change from baseline in dehydroepiandrosterone sulfate (DHEAS)b. DHEAS <upper limit of normal (ULN)c. DHEAS <75thpercentile of normal range (Q3) [Time Fame: 4 to up to 16 weeks] 3. Pharmacokinetics of compound 3 [Time Frame: 4 weeks] Exploratory Outcome Measure: 4. Changes in PD markers: Changes in cortisol, ACTH, DHEAS, DHEA, A4, T, 17-OHP, 11OHA4, 11OHT, 11KA4, 11KT [Time Frame: up to 16 weeks] EligibilityMinimum Age: 18 YearsMaximum Age: 30 yearsSex: Female onlyGender Based: YesAccepts Healthy Volunteers: NoCriteria: Inclusion Criteria: Inclusion Criteria:Female subjects 18 to 30 years old, inclusiveSubjects must have documented PCOS according to the NIH (1990) criteriaBMI <38 kg/m2DHEAS level >age matched reference ULN at ScreeningEvidence of DHEA hyperresponsiveness to ACTH stimulation during Screening Exclusion Criteria:Has a BMI >38 kg/m2Has PCOS—FOHHas HbAlc >6.5% or Fasting plasma glucose >126 mg/dLHas a history that includes bilateral adrenalectomy or hypopituitarismHas a history of allergy or hypersensitivity to tildacerfont or any other CRF1receptor antagonistClinically significant unstable medical condition, illness, or chronic diseaseClinically significant uncontrolled psychiatric disorderClinically significant abnormal laboratory finding or assessmentPregnant or nursing femalesUse of any other investigational drug within 30 daysUnable to understand and comply with the study procedures, understand the risks, and/or unwilling to provide written informed consent. While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. It is not intended that the invention be limited by the specific examples provided within the specification. While the invention has been described with reference to the aforementioned specification, the descriptions and illustrations of the embodiments herein are not meant to be construed in a limiting sense. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. Furthermore, it shall be understood that all aspects of the invention are not limited to the specific depictions, configurations or relative proportions set forth herein which depend upon a variety of conditions and variables. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is therefore contemplated that the invention shall also cover any such alternatives, modifications, variations or equivalents. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.
84,165
11858933
DETAILED DESCRIPTION OF THE INVENTION The invention is based on an unexpected discovery of a novel class of orally and/or topically available, selective and potent JAK1 therapeutics. The invention also provides pharmaceutical compositions of these compounds and methods of preparation and use thereof. The JAK1 inhibitors disclosed herein exhibited exceptional potency and selectivity profiles. More specifically, the novel JAK1 inhibitors disclosed herein enjoy improved potency as demonstrated by the superior binding affinities to JAK1 (e.g., IC50's of about 3-45 nM) and the potential for reduced hematopoietic side effects as demonstrated by their excellent specificity (e.g., JAK2 IC50's>20x JAK1). In one aspect, the invention generally relates to a compound having the structural formula (I): wherein R1is selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with NR′C(═O)RX, NR′C(═O)ORX, NR′C(═O)NRXRy, C(═O)NRXRy, NR′SO2RX, NR′SO2NRXRY, CR′R″SO2Rxor CR′R″SO2NRxRy; each R3is independently selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R4is a group selected from hydrogen, halogen, CN, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, and NHR′; each of RXand Ryis independently selected from H, alkyl (e.g., C1-C6alkyl), cycloalkyl (e.g., C3-C10cycloalkyl), heterocycloalkyl (e.g., C2-C9heterocycloalkyl), aryl (e.g., C4-C10aryl), heteroaryl (e.g., C3-C9heteroaryl) and RXand Rymay together form a 3- to 7-membered (e.g., 3-or 4-membered) ring, and each of RXand Ryis optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl); each of R′ and R″ is independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted and substituted alkyl and R′ and R″ may together form a 3- to 7-membered (e.g., 3- or 4-membered) ring; and n is 1 or 2, or a pharmaceutically acceptable form or an isotope derivative thereof. In certain embodiments of formula (I), R4is H and the compound has the structural formula (II): In certain embodiments of formula (II), n is 1, and the compound has the structural formula (III): In certain embodiments of formula (I), R′ is H, and the compound has the structural formula (IV): In certain embodiments of formulas (I), (II), (III) and (IV), both of R′ and R4is H. In certain embodiments of formulas (I), (II), (III) and (IV), R′ is methyl and R4is H. In certain embodiments of formulas (I), (II), (III) and (IV), n is 1. In certain embodiments, R3is H. In an exemplary embodiment where R′ is H, R4is H, n is 1, the compound has structural formula (V): In a further exemplary embodiment in formula (V), R3is H, and the compound has the structural formula (VI): R2may be selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with a group selected from NR′C(═O)RX, NR′C(═O)ORX, NR′C(═O)NRXRy, C(═O)NRXRy, NR′SO2RX, NR′SO2NRXRY, CR′R″SO2Rxor CR′R″SO2NRXRy, wherein each of RXand Ryis independently selected from H, alkyl (e.g., C1-C6alkyl), cycloalkyl (e.g., C3-C10cycloalkyl), heterocycloalkyl (e.g., C2-C9heterocycloalkyl), aryl (e.g., C4-C10aryl), heteroaryl (e.g., C3-C9heteroaryl), and each of RXand Ryis optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl), and RXand Rymay together form a 3- to 7-membered (e.g., 3- or 4-membered) ring. Each of R′ and R″ is independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted and substituted alkyl. R′ and R″ may together form a 3- to 7-membered ring. It is noted that the 3- to 7-membered (e.g., 3- or 4-membered) ring, optionally formed by R′ and R″ together or by RXand R together, may be a hetero 3- to 7-membered (e.g., 3- or 4-membered) ring, with 0 to 3 carbon atoms replaced by one or more heteroatoms selected from N, O, S and P. Heterocycloalkyl (e.g., C2-C9heterocycloalkyl) and heteroaryl (e.g., C3-C9heteroaryl) may have 1-4 carbon atoms replaced by one or more heteroatoms selected from N, O, S and P. In certain embodiments, R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with NR′C(═O)RX. In certain embodiments, R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with NR′C(═O)ORX. In certain embodiments, R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with NR′C(═O)NRXRy. In certain embodiments, R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with C(═O)NRXRy. In certain embodiments, R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with NR′SO2RX. In certain embodiments, R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with NR′SO2NRXRy. In certain embodiments, R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with CR′R″SO2RX. In certain embodiments, R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with CR′R″SO2NRXRY. In certain embodiments, R2comprises the following moiety: wherein each RLis independently (CH2)mand m is independently 0, 1, 2 or 3, wherein when an m is 0, the respective bridge is absent; provided that at least one m is not 0. In certain embodiments, no m is 0 (i.e., each m is independently 1, 2 or 3). In certain embodiments, not more than one m is 0 (i.e., not more than one RL's is absent). In certain embodiments, each m is independently 1, 2 or 3. In certain embodiments, all m's are the same integer selected from 1, 2 and 3. In certain embodiments, all m's are not the same integer selected from 1, 2 and 3. In certain embodiments, each m is 1 (i.e., forming a [1,1,1]-bicyclic moiety). In certain embodiments, each m is 2 (i.e., forming a [2,2,2]-bicyclic moiety). In certain embodiments, R2comprises: wherein R5is RXor NRXRY, wherein each of RXand Ryis independently selected from H, alkyl (e.g., C1-C6alkyl), cycloalkyl (e.g., C3-C10cycloalkyl), heterocycloalkyl (e.g., C2-C9heterocycloalkyl), aryl (e.g., C4-C10aryl), heteroaryl (e.g., C3-C9heteroaryl) and RXand Rymay together form a 3- to 7-membered (e.g., 3- or 4-membered) ring, and each of RXand Ryis and optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl); provided that when R5is RX, RXis not H (i.e., R5is not H). Each of R′ and R″ is independently selected from hydrogen and C1-C6unsubstituted and substituted alkyl and R′ and R″ may together form a 3- to 7-membered (e.g., 3- or 4-membered) ring. In certain embodiments, R5is RX. In certain embodiments, R5is a C1-C6alkyl (e.g., C1-C3alkyl), optionally substituted with one or more halogen (e.g., F, Cl), C1-C6(e.g., C1-C3) alkoxy, CN or amino groups. In certain embodiments, R5is a C1-C6alkyl (e.g., C1-C3alkyl). In certain embodiments, R5is a C1-C6alkyl (e.g., C1-C3alkyl), substituted with a halogen (e.g., F, Cl), C1-C6(e.g., C1-C3) alkoxy, or CN. In certain embodiments, R5is a C1-C6alkyl (e.g., C1-C3alkyl), substituted with a CN. In certain embodiments, RXis a linear or branched C1-C6alkyl. In certain embodiments, RXis a linear or branched C2-C4alkyl. In certain embodiments, RXis n-propyl or isopropyl. In certain embodiments, R5is NRXRY. In certain embodiments, one of RXand Ryis H. In certain embodiments, R5is NRXRY, wherein each of RXand Ryis independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted and substituted alkyl. In certain embodiments, R5is NRXRY, wherein the RXand Rytogether, along with the N in NRXRY, form a 3- to 7-membered (e.g., 3-, 4-, or 5-membered) heterocyclic group, optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl). In certain embodiments, the heterocyclic group is a 4-membered heterocyclic group. In certain embodiments, R5is NRXRY, wherein each of RXand Ryis independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted alkyl. In certain embodiments, R5is NRXRY, wherein RXand Rytogether form a 3- or 4-membered) cycloalkyl ring, substituted with a CN. In certain embodiments, each RLis CH2. In certain embodiments, each RLis (CH2)2. In another aspect, the invention generally relates to a compound having the structural formula (VII): wherein R1is selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; each R3is independently selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R4is a group selected from hydrogen (e.g., F, Cl), halogen, CN, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, and NHR′; R5is RXor NRXRY, wherein each of RXand Ryis independently selected from H, alkyl (e.g., C1-C6alkyl), cycloalkyl (e.g., C3-C10cycloalkyl), heterocycloalkyl (e.g., C2-C9heterocycloalkyl), aryl (e.g., C4-C10aryl), heteroaryl (e.g., C3-C9heteroaryl) and RXand Rymay together form a 3- to 7-membered (e.g., 3- or 4-membered) ring, and optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl); provided that when R5is RX, RXis not H (i.e., R5is not H); each RLis independently (CH2)mand m is independently 0, 1, 2 or 3, wherein when m is 0, the respective bridge is absent; each R′ and R″ is independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted and substituted alkyl and R′ and R″ may together form a 3- to 7-membered (e.g., 3- or 4-membered) ring; andn is 1 or 2, or a pharmaceutically acceptable form or an isotope derivative thereof. In certain embodiments of (VII), not more than one m is 0 (i.e., not more than one RL's is absent). In certain embodiments of (VII), no m is 0 (i.e., each m is independently 1, 2 or 3). In certain embodiments of (VII), all m's are the same integer selected from 1, 2 and 3. In certain embodiments of (VII), all m's are not the same integer selected from 1, 2 and 3. In certain embodiments of (VII), each m is 1 (i.e., forming a [1,1,1]-bicyclic moiety). In certain embodiments of (VII), each m is 2 (i.e., forming a [2,2,2]-bicyclic moiety). In certain embodiments of (VII), R4is H. In certain embodiments of (VII), R1is H. In certain embodiments of (VII), R1is methyl. In certain embodiments of (VII), n is 1. In certain embodiments of (VII), R4is H and n is 1, and the compound has the structural formula (VIII): In certain embodiments of (VIII), R′ is methyl and R3is H. In certain embodiments of (VIII), both R′ and R3is H, having the structural formula (IX): In certain embodiments of (VIII), each RLis CH2, and the compound has the structural formula (X): In certain embodiments of (X), R5is RX. In certain embodiments of (X), RXis a linear or branched C1-C6(e.g., C1-C3) alkyl, optionally substituted with one or more halogen (e.g., F, Cl), C1-C6(e.g., C1-C3) alkoxy, CN or amino groups. In certain embodiments of (X), RXis a linear or branched C2-C4alkyl. In certain embodiments of (X), RXis n-propyl or isopropyl. In certain embodiments of (X), R5is NRXRY. In certain embodiments of (X), one of RXand Ryis H. In certain embodiments, the RXand Rytogether, along with the N in NRXRY, form a 3-to 5-membered (e.g., 3-, 4- or 5-membered) heterocyclic group, optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl). In certain embodiments, the heterocyclic group is a 4-membered heterocyclic group. A list of non-limiting examples of compounds of the invention is provided in Table TABLE 1Exemplary Compounds123456789101112131415161718192021222324252627282930313233343536373839404142434445464748 In certain embodiments, the compound has the structural formula of compound 1. In certain embodiments, the compound has the structural formula of compound 4. In certain embodiments, the compound has the structural formula of compound 5. In certain embodiments, the compound has the structural formula of compound 6. In certain embodiments, the compound has the structural formula of compound 21. In certain embodiments, the compound has the structural formula of compound 23. In certain embodiments, the compound has the structural formula of compound 31. In certain embodiments, the compound has the structural formula of compound 32. In certain embodiments, the compound has the structural formula of compound 45. In certain embodiments, the compound has the structural formula of compound 46. In certain embodiments, the compound has the structural formula of compound 48. TABLE 1AExemplary CompoundsExample/CompoundNo.Compound NameCompound Structure1N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)- yl)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide43-Cyano-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin- 1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)azetidine-1- sulfonamide5N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)- yl)bicyclo[1.1.1]pentan-1-yl)-2-methylpropane-1- sulfonamide6N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)- yl)bicyclo[1.1.1]pentan-1-yl)-2- methoxyethanesulfonamide21N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)- yl)bicyclo[1.1.1]pentan-1-yl)-3-methoxyazetidine-1- sulfonamide233,3-Difluoro-N-(3-(imidazo[4,5-d]pyrrolo[2,3- b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)azetidine- 1-sulfonamide31N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)- yl)bicyclo[1.1.1]pentan-1-yl)butane-1-sulfonamide323,3,3-Trifluoro-N-(3-(imidazo[4,5-d]pyrrolo[2,3- b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane- 1-sulfonamide451-(((Cis-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)- yl)cyclobutyl)methyl) sulfonyl) azetidine-3-carbonitrile461-(3,3-Difluorocyclobutyl)-N-(3-(imidazo[4,5- d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan- 1-yl)methanesulfonamide48N-(3-(2-methylimidazo[4,5-d]pyrrolo[2,3-b]pyridin- 1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane-1- sulfonamide As discussed herein, isotope derivative compounds having one or more hydrogen atoms replaced with deuterium atoms are contemplated in the presented invention. In certain embodiments, a compound of the invention has one or more hydrogen atoms replaced with a deuterium atom. In certain embodiments, a compound of the invention has one hydrogen atom replaced with a deuterium atom. In certain embodiments, a compound of the invention has more than one hydrogen atom replaced with a deuterium atom. In yet another aspect, the invention generally relates to a pharmaceutical composition comprising a compound according to the herein disclosed invention, effective to treat or reduce one or more diseases or disorders, in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier, or diluent. In yet another aspect, the invention generally relates to a pharmaceutical composition comprising a compound having the structural formula (I): wherein R1is selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with NR′C(═O)RX, NR′C(═O)ORX, NR′C(═O)NRXRy, C(═O)NRXRy, NR′SO2Rx, NR′SO2NRxRY, CR′R″SO2Rxor CR′R″SO2NRxRy; each R3is independently selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R4is a group selected from hydrogen, halogen, CN, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, and NHR′; each of RXand Ryis independently selected from H, alkyl (e.g., C1-C6alkyl), cycloalkyl (e.g., C3-C10cycloalkyl), heterocycloalkyl (e.g., C2-C9heterocycloalkyl), aryl (e.g., C4-C10aryl), heteroaryl (e.g., C3-C9heteroaryl) and RXand Rymay together form a 3- to 7-membered (e.g., 3-or 4-membered) ring, and each of RXand Ryis optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl); each of R′ and R″ is independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted and substituted alkyl and R′ and R″ may together form a 3- to 7-membered (e.g., 3- or 4-membered) ring; and n is 1 or 2, or a pharmaceutically acceptable form or an isotope derivative thereof, and a pharmaceutically acceptable excipient, carrier, or diluent. In yet another aspect, the invention generally relates to a pharmaceutical composition comprising a compound having the structural formula (VII): wherein R1is selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; each R3is independently selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R4is a group selected from hydrogen (e.g., F, Cl), halogen, CN, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, and NHR′; R5is RXor NRXRY, wherein each of RXand Ryis independently selected from H, alkyl (e.g., C1-C6alkyl), cycloalkyl (e.g., C3-C10cycloalkyl), heterocycloalkyl (e.g., C2-C9heterocycloalkyl), aryl (e.g., C4-C10aryl), heteroaryl (e.g., C3-C9heteroaryl) and RXand Rymay together form a 3- to 7-membered (e.g., 3- or 4-membered) ring, and optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl); provided that when R5is RX, RXis not H (i.e., R5is not H); each RLis independently (CH2)mand m is independently 0, 1, 2 or 3, wherein when m is 0, the respective bridge is absent; each R′ and R″ is independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted and substituted alkyl and R′ and R″ may together form a 3- to 7-membered (e.g., 3- or 4-membered) ring; and n is 1 or 2, or a pharmaceutically acceptable form or an isotope derivative thereof, and a pharmaceutically acceptable excipient, carrier, or diluent. In certain embodiments, a pharmaceutical composition herein disclosed is suitable for oral administration. In certain embodiments, the pharmaceutical composition of the invention is suitable for topical administration. In certain embodiments, a pharmaceutical composition herein disclosed is useful to treat or reduce one or more of inflammatory diseases, immune-mediated diseases and cancer, or a related disease or disorder. In certain embodiments, a pharmaceutical composition herein disclosed is useful to treat or reduce one or more autoimmune diseases, or a related disease or disorder. In certain embodiments of the pharmaceutical composition, the disease or disorder is selected from: asthma, allergies, arthritis (e.g., rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis), juvenile arthritis, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), endocrinopathies (e.g., type 1 diabetes and Graves' disease), neurodegenerative diseases (e.g., multiple sclerosis (MS)), autistic spectrum disorder, depression, Alzheimer's disease, Guillain-Barre syndrome, obsessive-compulsive disorder, optic neuritis, retinal degeneration, dry eye syndrome DES, Sjögren's syndrome, amyotrophic lateral sclerosis (ALS), Parkinson's disease, Huntington's Disease, Guillain-Barre syndrome, myasthenia gravis, and chronic idiopathic demyelinating disease (CID), vascular diseases (e.g., autoimmune hearing loss, systemic vasculitis, and atherosclerosis), skin diseases (e.g., acne vulgaris dermatomyositis, pemphigus, systemic lupus erythematosus (SLE), discoid lupus erthematosus, scleroderma, psoriasis, plaque psoriasis, vasculitics, vitiligo and alopecias), Hashimoto's thyroiditis, pernicious anemia, Cushing's disease, Addison's disease, chronic active hepatitis, polycystic ovary syndrome (PCOS), celiac disease, pemphigus, transplant rejection (allograft transplant rejection), graft-versus-host disease (GVDH), or a related disease or disorder thereof. In yet another aspect, the invention generally relates to a unit dosage form comprising a pharmaceutical composition disclosed herein. In certain embodiments, the unit dosage form is a solid dosage form, for example, in the forms of capsules, tablets, pills, powders or granules. In certain embodiments, the unit dosage form is a tablet. In certain embodiments, the unit dosage form is a capsule. In certain embodiments, the unit dosage form is a liquid dosage form, for example, in the forms of emulsions, solutions, suspensions, syrups or elixirs. In yet another aspect, the invention generally relates to a method for treating or reducing a disease or disorder, comprising: administering to a subject in need thereof a pharmaceutical composition comprising a compound having the structural formula (I): wherein R1is selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R2is selected from C3-C10(e.g., C3-C6) cycloalkyl, bicycloalkyl, spirocyclic or bridgedcycloalkyl, substituted with NR′C(═O)RX, NR′C(═O)ORX, NR′C(═O)NRXRy, C(═O)NRXRy, NR′SO2Rx, NR′SO2NRxRY, CR′R″SO2Rxor CR′R″SO2NRxRy; each R3is independently selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R4is a group selected from hydrogen, halogen, CN, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, and NHR′; each of RXand Ryis independently selected from H, alkyl (e.g., C1-C6alkyl), cycloalkyl (e.g., C3-C10cycloalkyl), heterocycloalkyl (e.g., C2-C9heterocycloalkyl), aryl (e.g., C4-C10aryl), heteroaryl (e.g., C3-C9heteroaryl) and RXand Rymay together form a 3- to 7-membered (e.g., 3-or 4-membered) ring, and each of RXand Ryis optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl); each of R′ and R″ is independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted and substituted alkyl and R′ and R″ may together form a 3- to 7-membered (e.g., 3- or 4-membered) ring; and n is 1 or 2, or a pharmaceutically acceptable form or an isotope derivative thereof, effective to treat or reduce one or more of inflammatory diseases, immune-mediated diseases and cancer, or a related disease or disorder, in a mammal, including a human. In yet another aspect, the invention generally relates to a method for treating or reducing a disease or disorder, comprising: administering to a subject in need thereof a pharmaceutical composition comprising a compound having the structural formula (VII): wherein R1is selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; each R3is independently selected from hydrogen, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, COOR′ and CONR′R″; R4is a group selected from hydrogen (e.g., F, Cl), halogen, CN, C1-C6(e.g., C1-C3) unsubstituted or substituted alkyl, OR′, and NHR′; R5is RXor NRXRY, wherein each of RXand Ryis independently selected from H, alkyl (e.g., C1-C6alkyl), cycloalkyl (e.g., C3-C10cycloalkyl), heterocycloalkyl (e.g., C2-C9heterocycloalkyl), aryl (e.g., C4-C10aryl), heteroaryl (e.g., C3-C9heteroaryl) and RXand Rymay together form a 3- to 7-membered (e.g., 3- or 4-membered) ring, and optionally substituted with one or more of halogen (e.g., F, Cl), CN, OR′, NR′R″, alkyl (e.g., C1-C6alkyl), haloalkyl (e.g., CHF2, CF3), cyanoalkyl (e.g., CH2CN), hydroxyalkyl (e.g., CH2OH) and alkoxyalkyl (e.g., CH2O-alkyl); provided that when R5is RX, RXis not H (i.e., R5is not H); each RLis independently (CH2)mand m is independently 0, 1, 2 or 3, wherein when m is 0, the respective bridge is absent; each R′ and R″ is independently selected from hydrogen and C1-C6(e.g., C1-C3) unsubstituted and substituted alkyl and R′ and R″ may together form a 3- to 7-membered (e.g., 3- or 4-membered) ring; and n is 1 or 2, or a pharmaceutically acceptable form or an isotope derivative thereof, and a pharmaceutically acceptable excipient, carrier, or diluent, effective to treat or reduce one or more of inflammatory diseases, immune-mediated diseases and cancer, or a related disease or disorder, in a mammal, including a human. In yet another aspect, the invention generally relates to a method for treating or reducing a disease or disorder, comprising: administering to a subject in need thereof a pharmaceutical composition comprising a compound disclosed herein, wherein the disease or disorder is one or more of inflammatory diseases, immune-mediated diseases and cancer, or a related disease or disorder. In certain embodiments, the method of the invention is useful for treating or reducing an autoimmune disease. In certain embodiments, the disease or disorder is selected from: asthma, allergies, arthritis (e.g., rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis), juvenile arthritis, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), endocrinopathies (e.g., type 1 diabetes and Graves' disease), neurodegenerative diseases (e.g., multiple sclerosis (MS)), autistic spectrum disorder, depression, Alzheimer's disease, Guillain-Barre syndrome, obsessive-compulsive disorder, optic neuritis, retinal degeneration, dry eye syndrome DES, Sjögren's syndrome, amyotrophic lateral sclerosis (ALS), Parkinson's disease, Huntington's Disease, Guillain-Barre syndrome, myasthenia gravis, and chronic idiopathic demyelinating disease (CID), vascular diseases (e.g., autoimmune hearing loss, systemic vasculitis, and atherosclerosis), skin diseases (e.g., acne vulgaris dermatomyositis, pemphigus, systemic lupus erythematosus (SLE), discoid lupus erthematosus, scleroderma, psoriasis, plaque psoriasis, vasculitics, vitiligo and alopecias), Hashimoto's thyroiditis, pernicious anemia, Cushing's disease, Addison's disease, chronic active hepatitis, polycystic ovary syndrome (PCOS), celiac disease, pemphigus, transplant rejection (allograft transplant rejection), graft-versus-host disease (GVDH), or a related disease or disorder thereof. In certain embodiments, the method of the invention is useful for treating or reducing an inflammatory disease, or a related disease or disorder. In certain embodiments, the method of the invention is useful for treating or reducing an autoimmune disease, or a related disease or disorder. In certain embodiments, the method of the invention is useful for treating or reducing an immune-mediated disease, or a related disease or disorder. In certain embodiments, the method of the invention is useful for treating or reducing cancer, or a related disease or disorder. In certain embodiments, the method of the invention is useful for treating or reducing one or more of rheumatoid arthritis, ankylosing spondylitis, psoriasis, atopic dermatitis, inflammatory bowel disease, Crohn's, ulcerative colitis, DES, vitiligo, alopecia areata, alopecia totalis. In yet another aspect, the invention generally relates to use of a compound disclosed herein and a pharmaceutically acceptable excipient, carrier, or diluent, in preparation of a medicament for treating a disease or disorder. In certain embodiments of such use, the disease or disorder is one or more of inflammatory diseases, immune-mediated diseases and cancer. In certain embodiments of such use, the disease or disorder is an autoimmune disease. In certain embodiments of such use, the disease or disorder is selected from: asthma, allergies, arthritis (e.g., rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis), juvenile arthritis, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), endocrinopathies (e.g., type 1 diabetes and Graves' disease), neurodegenerative diseases (e.g., multiple sclerosis (MS), autistic spectrum disorder, depression, Alzheimer's disease, Guillain-Barre syndrome, obsessive-compulsive disorder, optic neuritis, retinal degeneration, dry eye syndrome DES, Sjögren's syndrome, amyotrophic lateral sclerosis (ALS), Parkinson's disease, Huntington's Disease, Guillain-Barre syndrome, myasthenia gravis, and chronic idiopathic demyelinating disease (CID), vascular diseases (e.g., autoimmune hearing loss, systemic vasculitis, and atherosclerosis), skin diseases (e.g., acne vulgaris dermatomyositis, pemphigus, systemic lupus erythematosus (SLE), discoid lupus erthematosus, scleroderma, psoriasis, plaque psoriasis, vasculitics, vitiligo and alopecias), Hashimoto's thyroiditis, pernicious anemia, Cushing's disease, Addison's disease, chronic active hepatitis, polycystic ovary syndrome (PCOS), celiac disease, pemphigus, transplant rejection (allograft transplant rejection), graft-versus-host disease (GVDH), or a related disease or disorder thereof. In certain embodiments of such use, the disease or disorder is one or more of rheumatoid arthritis, ankylosing spondylitis, psoriasis, atopic dermatitis, inflammatory bowel disease, Crohn's, ulcerative colitis, DES, vitiligo, alopecia areata, alopecia totalis. In certain embodiments of the use, the medicament is for oral administration. In certain embodiments of the use, the medicament is for topical administration. The term “inflammatory disease” refers to a disease or condition characterized by aberrant inflammation, e.g. an increased level of inflammation compared to a control such as a healthy person not suffering from a disease. Examples of inflammatory diseases that may be treated with a compound, pharmaceutical composition, or method described herein include autoimmune diseases, traumatic brain injury, arthritis, rheumatoid arthritis, psoriatic arthritis, juvenile idiopathic arthritis, multiple sclerosis, systemic lupus erythematosus (SLE), myasthenia gravis, juvenile onset diabetes, diabetes mellitus type 1, Guillain-Barre syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, ankylosing spondylitis, psoriasis, Sjögren's syndrome, vasculitis, glomerulonephritis, auto-immune thyroiditis, Behcet's disease, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, ichthyosis, Graves ophthalmopathy, inflammatory bowel disease, Addison's disease, Vitiligo, asthma, allergic asthma, acne vulgaris, celiac disease, chronic prostatitis, inflammatory bowel disease, pelvic inflammatory disease, reperfusion injury, ischemia reperfusion injury, stroke, sarcoidosis, transplant rejection, interstitial cystitis, atherosclerosis, scleroderma, and atopic dermatitis. Such conditions are frequently inextricably intertwined with other diseases, disorders and conditions. A non-limiting list of inflammatory-related diseases, disorders and conditions which may, for example, be caused by inflammatory cytokines, include, arthritis, kidney failure, lupus, asthma, psoriasis, colitis, pancreatitis, allergies, fibrosis, surgical complications (e.g., where inflammatory cytokines prevent healing), anemia, and fibromyalgia. Other diseases and disorders, which may be associated with chronic inflammation include Alzheimer's disease, congestive heart failure, stroke, aortic valve stenosis, arteriosclerosis, osteoporosis, Parkinson's disease, infections, inflammatory bowel disease (IBD), allergic contact dermatitis and other eczemas, systemic sclerosis, transplantation and multiple sclerosis. Some of the aforementioned diseases, disorders and conditions for which a compound of the present disclosure may be particularly efficacious (due to, for example, limitations of current therapies) are described in more detail hereafter. The term “autoimmune disease” refers to a disease or condition in which a subject's immune system has an aberrant immune response against a substance that does not normally elicit an immune response in a healthy subject. Examples of autoimmune diseases that may be treated with a compound, pharmaceutical composition, or method described herein include acne vulgaris, acute disseminated encephalomyelitis, acute necrotizing hemorrhagic leukoencephalitis, Addison's disease, agammaglobulinemia, Aicardi-Goutieres syndrome (AGS), alopecia areata, alopecia totalis, amyloidosis, ankylosing spondylitis, anti-GBM/anti-TBM nephritis, antiphospholipid syndrome, autoimmune angioedema, autoimmune aplastic anemia, autoimmune dysautonomia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease, autoimmune myocarditis, autoimmune oophoritis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura, autoimmune thyroid disease, autoimmune urticaria, axonal or neuronal neuropathies, balo disease, Behcet's disease, bullous pemphigoid, cardiomyopathy, Castleman disease, celiac disease, Chagas disease, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE), chronic active hepatitis, chronic fatigue syndrome, chronic inflammatory demyelinating polyneuropathy, chronic recurrent multifocal ostomyelitis, Churg-Strauss syndrome, cicatricial pemphigoid/benign mucosal pemphigoid, Crohn's disease, Cogans syndrome, cold agglutinin disease, congenital heart block, coxsackie myocarditis, CREST disease, Cushing's disease, demyelinating neuropathies, depression, dermatitis herpetiformis, dermatomyositis, Devic's disease (neuromyelitis optica), discoid lupus, Dressler's syndrome, dry eye syndrome DES (keratoconjunctivitis sicca), endometriosis, eosinophilic esophagitis, eosinophilic fasciitis, erythema nodosum, essential mixed cryoglobulinemia, experimental allergic encephalomyelitis, Evans syndrome, fibromyalgia, fibrosing alveolitis, giant cell arteritis (temporal arteritis), giant cell myocarditis, glomerulonephritis, Goodpasture's syndrome, granulomatosis with polyangiitis, graft-versus-host disease (GVDH), Graves' disease, Guillain-Barre syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, hemolytic anemia, Henoch-Schonlein purpura, herpes gestationis, hidradenitis suppurativa, hypogammaglobulinemia, idiopathic thrombocytopenic purpura, IgA nephropathy, IgG4-related sclerosing disease, inflammatory bowel disease (IBD), immunoregulatory lipoproteins, inclusion body myositis, interstitial cystitis, juvenile arthritis, juvenile diabetes (Type 1 diabetes), juvenile dermatomyositis (JDM), juvenile myositis, Kawasaki syndrome, Lambert-Eaton syndrome, leukocytoclastic vasculitis, lichen planus, lichen sclerosus, ligneous conjunctivitis, linear IgA disease, lupus, lyme disease, chronic, Meniere's disease, microscopic polyangiitis, mixed connective tissue disease, Mooren's ulcer, Mucha-Habermann disease, multiple sclerosis (MS), myasthenia gravis, myositis, narcolepsy, neuromyelitis optica, neutropenia, ocular cicatricial pemphigoid, optic neuritis, palindromic rheumatism, pediatric autoimmune neuropsychiatric disorders associated withstreptococcus, paraneoplastic cerebellar degeneration, paroxysmal nocturnal hemoglobinuria p, Parry Romberg syndrome, Parsonnage-Turner syndrome, Pars planitis (peripheral uveitis), pemphigus, peripheral neuropathy, perivenous encephalomyelitis, pernicious anemia, POEMS syndrome, polyarteritis nodosa, polycystic ovary syndrome (PCOS), Type I, II, & III autoimmune polyglandular syndromes, polymyalgia rheumatica, polymyositis, postmyocardial infarction syndrome, postpericardiotomy syndrome, progesterone dermatitis, primary biliary cirrhosis, primary sclerosing cholangitis, psoriasis, psoriatic arthritis, plaque psoriasis, idiopathic pulmonary fibrosis, pyoderma gangrenosum, pure red cell aplasia, Raynauds phenomenon, reactive Arthritis, reflex sympathetic dystrophy, Reiter's syndrome, relapsing polychondritis, restless legs syndrome, retroperitoneal fibrosis, rheumatic fever, rheumatoid arthritis, sarcoidosis, Schmidt syndrome, scleritis, scleroderma, Sjögren's syndrome, sperm & testicular autoimmunity, stiff person syndrome, stimulator of interferon genes (STING)-associated vasculopathy with onset during infancy (SAVI), subacute bacterial endocarditis, Susac's syndrome, sympathetic ophthalmia, systemic lupus erythematosus (SLE), Takayasu's arteritis, temporal arteritis/Giant cell arteritis, thrombocytopenic purpura, Tolosa-Hunt syndrome, transplant rejection (allograft transplant rejection), transverse myelitis, Type 1 diabetes, ulcerative colitis, undifferentiated connective tissue disease, uveitis, vasculitis, vesiculobullous dermatosis, vitiligo, or Wegener's granulomatosis. The term “immune-mediated disease” refers to chronic inflammatory diseases perpetuated by antibodies and cellular immunity. Immune-mediated diseases include, for example, but not limited to, asthma, allergies, arthritis (e.g., rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis), juvenile arthritis, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), endocrinopathies (e.g., type 1 diabetes and Graves' disease), neurodegenerative diseases (e.g., multiple sclerosis (MS)), autistic spectrum disorder, depression, Alzheimer's disease, Guillain-Barre syndrome, obsessive-compulsive disorder, optic neuritis, retinal degeneration, dry eye syndrome DES, Sjögren's syndrome, amyotrophic lateral sclerosis (ALS), Parkinson's disease, Huntington's Disease, Guillain-Barre syndrome, myasthenia gravis, and chronic idiopathic demyelinating disease (CID), vascular diseases (e.g., autoimmune hearing loss, systemic vasculitis, and atherosclerosis), and skin diseases (e.g., acne vulgaris dermatomyositis, pemphigus, systemic lupus erythematosus (SLE), discoid lupus erthematosus, scleroderma, psoriasis, plaque psoriasis, vasculitics, vitiligo and alopecias). Hashimoto's thyroiditis, pernicious anemia, Cushing's disease, Addison's disease, chronic active hepatitis, polycystic ovary syndrome (PCOS), celiac disease, pemphigus, transplant rejection (allograft transplant rejection), graft-versus-host disease (GVDH). The term “cancer” as used herein refers to all types of cancer, neoplasm or malignant tumors found in mammals, e.g., humans, including hematological cancers leukemia, and lymphomas, T-ALL, solid cancers such as carcinomas and sarcomas. Exemplary cancers include blood cancer, brain cancer, glioma, glioblastoma, neuroblastoma, prostate cancer, colorectal cancer, pancreatic cancer, cervical cancer, gastric cancer, ovarian cancer, lung cancer, and cancer of the head. Exemplary cancers include cancer of the thyroid, endocrine system, brain, breast, cervix, colon, head & neck, liver, kidney, lung, non-small cell lung, melanoma, mesothelioma, ovary, sarcoma, stomach, uterus, Medulloblastoma, colorectal cancer, pancreatic cancer. Additional examples include myeloproliferative neoplasms, thyroid carcinoma, cholangiocarcinoma, pancreatic adenocarcinoma, skin cutaneous melanoma, colon adenocarcinoma, rectum adenocarcinoma, stomach adenocarcinoma, esophageal carcinoma, head and neck squamous cell carcinoma, breast invasive carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, Hodgkin's Disease, Non-Hodgkin's Lymphoma, multiple myeloma, neuroblastoma, glioma, glioblastoma multiforme, ovarian cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, primary brain tumors, cancer, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, lymphomas, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, neoplasms of the endocrine or exocrine pancreas, medullary thyroid cancer, medullary thyroid carcinoma, melanoma, colorectal cancer, papillary thyroid cancer, hepatocellular carcinoma, or prostate cancer. Isotopically-labeled compounds are also within the scope of the present disclosure. As used herein, an “isotopically-labeled compound” or “isotope derivative” refers to a presently disclosed compound including pharmaceutical salts and prodrugs thereof, each as described herein, in which one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds presently disclosed include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as2H,3H,13C, 14C,15N18O,17O,31P,32p,35S,18F, and36Cl, respectively. By isotopically-labeling the presently disclosed compounds, the compounds may be useful in drug and/or substrate tissue distribution assays. Tritiated (3H) and carbon-14 (14C) labeled compounds are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (2H) can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labeled compounds presently disclosed, including pharmaceutical salts, esters, and prodrugs thereof, can be prepared by any means known in the art. Further, substitution of normally abundant hydrogen (H) with heavier isotopes such as deuterium can afford certain therapeutic advantages, e.g., resulting from improved absorption, distribution, metabolism and/or excretion (ADME) properties, creating drugs with improved efficacy, safety, and/or tolerability. Benefits may also be obtained from replacement of normally abundant12C with13C. (See, WO 2007/005643, WO 2007/005644, WO 2007/016361, and WO 2007/016431.) Stereoisomers (e.g., cis and trans isomers) and all optical isomers of a presently disclosed compound (e.g., R and S enantiomers), as well as racemic, diastereomeric and other mixtures of such isomers are within the scope of the present disclosure. Compounds of the present invention are, subsequent to their preparation, preferably isolated and purified to obtain a composition containing an amount by weight equal to or greater than 95% (“substantially pure”), which is then used or formulated as described herein. In certain embodiments, the compounds of the present invention are more than 99% pure. Solvates and polymorphs of the compounds of the invention are also contemplated herein. Solvates of the compounds of the present invention include, for example, hydrates. General considerations in formulation and/or manufacture of pharmaceutical compositions agents can be found, for example, in Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980), and Remington: The Science and Practice of Pharmacy, 21st Edition (Lippincott Williams & Wilkins, 2005). Pharmaceutical compositions described herein can be prepared by any method known in the art of pharmacology. In general, such preparatory methods include the steps of bringing a compound described herein (the “active ingredient”) into association with a carrier and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit. Pharmaceutical compositions can be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a “unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the compounds described herein or derivatives thereof are admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (i) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (ii) binders, as for example, carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose, and acacia, (iii) humectants, as for example, glycerol, (iv) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate, (v) solution retarders, as for example, paraffin, (vi) absorption accelerators, as for example, quaternary ammonium compounds, (vii) wetting agents, as for example, cetyl alcohol, and glycerol monostearate, (viii) adsorbents, as for example, kaolin and bentonite, and (ix) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethyleneglycols, and the like. Solid dosage forms such as tablets, dragées, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and others known in the art. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents, and emulsifiers, such as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols, and fatty acid esters of sorbitan, or mixtures of these substances, and the like. Besides such inert diluents, the composition can also include additional agents, such as wetting, emulsifying, suspending, sweetening, flavoring, or perfuming agents. Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition of the present disclosure will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100% (w/w) active ingredient. The exact amount of a compound required to achieve an effective amount will vary from subject to subject, depending, for example, on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular compound(s), mode of administration, and the like. The desired dosage can be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage can be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations). In certain embodiments, an effective amount of a compound for administration one or more times a day to an adult human of 70 Kg may comprise about 0.001 mg to about 3,000 mg (e.g., about 0.001 mg to about 2,000 mg, about 0.001 mg to about 1,000 mg, about 0.001 mg to about 500 mg, about 0.001 mg to about 100 mg, about 0.001 mg to about 50 mg, about 0.001 mg to about 10 mg, about 0.01 mg to about 1,000 mg, about 0.1 mg to about 1,000 mg, about 1 mg to about 1,000 mg, about 10 mg to about 1,000 mg, about 100 mg to about 1,000 mg, about 1 mg to about 500 mg, about 5 mg to about 250 mg) of a compound per unit dosage form. In certain embodiments, a compound described herein may be administered at dosage levels sufficient to deliver from about 0.001 mg/Kg to about 1,000 mg/Kg (e.g., from about 0.01 mg/Kg to about 1,000 mg/Kg, from about 0.1 mg/Kg to about 1,000 mg/Kg, from about 1 mg/Kg to about 1,000 mg/Kg, from about 0.001 mg/Kg to about 100 mg/Kg, from about 0.001 mg/Kg to about 10 mg/Kg, from about 0.001 mg/Kg to about 1 mg/Kg, from about 0.1 mg/Kg to about 40 mg/Kg, from about 0.5 mg/Kg to about 30 mg/Kg, from about 0.01 mg/Kg to about 10 mg/Kg, from about 0.1 mg/Kg to about 10 mg/Kg, or from about 1 mg/Kg to about 25 mg/Kg) of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. In some embodiments, the dosing regimen is continued for days, weeks, months, or years. It will be appreciated that dose ranges as described herein provide guidance for the administration of provided pharmaceutical compositions to an adult. The amount to be administered to, for example, a child or an adolescent can be determined by a medical practitioner or person skilled in the art and can be lower or the same as that administered to an adult. It will be also appreciated that a compound or pharmaceutical composition, as described herein, can be administered in combination with one or more additional therapeutically active agents. In certain embodiments, a compound or pharmaceutical composition provided herein is administered in combination with one or more additional therapeutically active agents that improve its bioavailability, reduce and/or modify its metabolism, inhibit its excretion, and/or modify its distribution within the body. It will also be appreciated that the therapy employed may achieve a desired effect for the same disorder, and/or it may achieve different effects. The compound or pharmaceutical composition can be administered concurrently with, prior to, or subsequent to, one or more additional therapeutically active agents. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent. In will further be appreciated that the additional therapeutically active agent utilized in this combination can be administered together in a single composition or administered separately in different compositions. The particular combination to employ in a regimen will consider compatibility of a provided compound with the additional therapeutically active agent and/or the desired therapeutic effect to be achieved. In general, it is expected that additional therapeutically active agents utilized in combination be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually. Exemplary additional therapeutically active agents include, but are not limited to, small organic molecules such as drug compounds, e.g., compounds approved by the U.S. Food and Drug Administration (FDA) as provided in the Code of Federal Regulations (CFR), peptides, proteins, carbohydrates, monosaccharides, oligosaccharides, polysaccharides, nucleoproteins, mucoproteins, lipoproteins, synthetic polypeptides or proteins, small molecules linked to proteins, glycoproteins, steroids, nucleic acids, DNAs, RNAs, nucleotides, nucleosides, oligonucleotides, antisense oligonucleotides, lipids, hormones, vitamins and cells. Materials, compositions, and components disclosed herein can be used for, can be used in conjunction with, can be used in preparation for, or are products of the disclosed methods and compositions. It is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutations of these compounds may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a method is disclosed and discussed and a number of modifications that can be made to a number of molecules including in the method are discussed, each and every combination and permutation of the method, and the modifications that are possible are specifically contemplated unless specifically indicated to the contrary. Likewise, any subset or combination of these is also specifically contemplated and disclosed. This concept applies to all aspects of this disclosure including, but not limited to, steps in methods using the disclosed compounds or compositions. Thus, if there are a variety of additional steps that can be performed, it is understood that each of these additional steps can be performed with any specific method steps or combination of method steps of the disclosed methods, and that each such combination or subset of combinations is specifically contemplated and should be considered disclosed. Certain compounds of the present invention may exist in particular geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis-and trans-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention. Isomeric mixtures containing any of a variety of isomer ratios may be utilized in accordance with the present invention. For example, where only two isomers are combined, mixtures containing 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98:2, 99:1, or 100:0 isomer ratios are contemplated by the present invention. Those of ordinary skill in the art will readily appreciate that analogous ratios are contemplated for more complex isomer mixtures. If, for instance, a particular enantiomer of a compound of the present invention is desired, it may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers. Alternatively, where the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic methods well known in the art, and subsequent recovery of the pure enantiomers. The following examples are meant to be illustrative of the practice of the invention and not limiting in any way. EXAMPLES Abbreviations Certain abbreviations are listed below.Methanol: MeOHDichloromethane: DCMPetroleum ether: PEEthyl acetate: EtOAcTriethylamine: TEASodium hydroxide: NaOHNitrogen: N2Diphenyl phosphoryl azide: DPPAThin-Layer Chromatography: TLCHigh Performance Liquid Chromatography: HPLCN,N-Diisopropylethylamine: DIPEAN,N-Dimethylformamide: DMF4-Methylbenzene-1-sulfonyl chloride: TsC1Room temperature: RTHours: hrs Representative methods of prep-HPLC: (flow rate and gradient may change) Exemplary methods for prep-HPLC are provided below. MethodA: NH4HCO3: (Column: XBrige Prep C18 5 m OBD 19*150 mm, PN 186002979; mobile phase: CH3CN in water (0.1% NH4HCO3) from 20% to 60%, flow rate: 15 mL/min). Method B: (Column: XBridge Prep C18 5 m OBD 19*150 mm, PN 186002979; mobile phase: CH3CN in water (0.1% formic acid) from 15% to 40%, flow rate: 15 mL/min) Representative methods of analytical-HPLC Method 1: Analysis was performed on an Agilent 1260 series HPLC-6120MS. UHPLC Long Gradient Equivalent 5% to 95% acetonitrile (containing 0.02% NH4OAc) in water run time of 6.5 minutes with a flow rate of 1.5 mL/min. A XBridge C18 column (5 m, 4.6*50 mm; PN 186003113) was used at a temperature of 40° C. Method 2: Analysis was performed on an Agilent 1200 series HPLC-6120MS. UHPLC Long Gradient Equivalent 5% to 95% acetonitrile (containing 0.1% trifluoroacetic acid) in water run time of 6.5 minutes with a flow rate of 1.5 mL/min. A XBridge C18 column (5 m, 4.6*50 mm; PN 186003113) was used at a temperature of 40° C. Method 3: Analysis was performed on an Agilent 1260 series HPLC-6120MS. UHPLC Long Gradient Equivalent 5% to 95% acetonitrile (containing 0.02% NH40Ac) in water run time of 6.5 minutes with a flow rate of 2 mL/min. A Diamonsil Plus C18 column (5 m, 4.6*30 mm Cat #99436) was used at a temperature of 40° C. Example 1 Step 1.4-Chloro-1-tosyl-1H-pyrrolo[2,3-b]pyridine (1b) Compound 1a (30 g, 0.2 mol) and TsC1 (45 g, 0.24 mol) were dissolved in a mixture of acetone and water (600 mL, V:V=5:1) followed by the addition of NaOH (11.8 g, 0.29 mmol) at 0° C. After stirring at RT for 1 h, the mixture was concentrated to 100 mL of solvent and cooled with ice-water. The formed solid was filtered and dried to afford title product as a white solid (52 g, 86% yield).1H NMR (400 MHz, CDCl3) δ 8.30 (d, J=5.6 Hz, 1H), 8.05 (d, J=8.4 Hz, 2H), 7.76 (d, J=4.0 Hz, 1H), 7.27 (d, J=8.4 Hz, 2H), 7.18 (d, J=5.2 Hz, 1H), 6.69 (d, J=4.0 Hz, 1H), 2.37 (s, 3H). Step 2.4-Chloro-5-nitro-1-tosyl-1H-pyrrolo[2,3-b]pyridine (1c) To a mixture of compound 1b (5.0 g, 16.3 mmol) and 75 mL of DCM was added tetrabutylammonium nitrate (2.9 g, 21.3 mmol) portion-wise at 0° C. followed by trifluoroacetic anhydride (3.14 mL, 22.2 mmol) slowly. After stirring for 16 hrs at RT, another portion of tetrabutylammonium nitrate (0.58 g, 4.23 mmol) and trifluoroacetic anhydride (0.8 mL, 5.7 mmol) were added at 0° C. After warmed up to room temperature, the reaction mixture was stirred for 4 hrs at RT. The reaction mixture was diluted with DCM (150 mL), washed with water (30 mL×2) and then concentrated to dryness. The residue was triturated in MeOH to afford title product as a white solid (3.15 g, 55% yield). LC-MS (Method 2): tR=1.76 min, m/z (M+H)+=351.8. Step 3. Tert-butyl 3-((5-nitro-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino)bicyclo[1.1.1]pentane-1-carboxylate (1d) Compound 1c (500 mg, 1.42 mmol), tert-butyl 3-aminobicyclo[1.1.1]pentane-1-carboxylate (313 mg, 1.71 mmol) and DIPEA (276 mg, 2.13 mmol) were dissolved in isopropanol (5 mL). The above solution was stirred at 120° C. for 2 hrs. After cooling, the formed solid was collected by filtering and dried to afford the title product as a brown solid (612 mg, 86% yield).1H NMR (400 MHz, CDCl3) δ 9.28 (s, 1H), 9.11 (s, 1H), 8.07 (d, J=8.0 Hz, 2H), 7.64 (d, J=5.6 Hz, 1H), 7.30 (d, J=8.0 Hz, 2H), 6.96 (d, J=5.6 Hz, 1H), 2.48 (s, 6H), 2.40 (s, 3H), 1.47 (s, 9H). Step 4. Tert-butyl 3-((5-amino-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino)bicyclo[1.1.1]pentane-1-carboxylate (1e) Compound 1d (600 mg, 1.22 mmol) was dissolved in MeOH (6 mL) followed by the addition of Pd/C (48 mg, 10% wt) in one portion. The mixture was hydrogenated (1 atm) at RT for 16 hrs. The mixture was filtered and the filtrate was concentrated. The residue was purified by prep. TLC (PE:EtOAc=1:1) to afford the title product as a white solid (258 mg, 46% yield). LC-MS (Method 2): tR=1.64 min, m/z (M+H)+=469.0. Step 5. Tert-butyl 3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentane-1-carboxylate (if) Compound 1e (258 mg, 0.55 mmol), triethyl orthoformate (204 mg, 1.37 mmol) and p-toluenesulfonic acid (10 mg, 0.05 mmol) were dissolved in toluene (6 mL). The mixture was stirred for 16 hrs at 120° C. After cooling, the mixture was concentrated to dryness. The residue was purified by chromatography on silica gel (elute: PE:EtOAc=1:1) to afford the title product as a brown solid (191 mg, 73% yield).1H NMR (400 MHz, CDCl3) δ 8.91 (s, 1H), 8.10 (d, J=8.0 Hz, 2H), 7.82 (d, J=8.0 Hz, 2H), 7.27-7.25 (m, 2H), 6.83 (d, J=4.4 Hz, 1H), 2.71 (s, 6H), 2.35 (s, 3H), 1.51 (s, 9H). Step 6.3-(6-Tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentane-1-carboxylic acid (1g) To a solution of compound if (191 mg, 0.40 mmol) in DCM (2 mL) was added TFA (1 mL). After stirring for 16 hrs at RT, the mixture was concentrated to dryness to afford crude title product as a brown solid (170 mg, 100% yield). LC-MS (Method 2): tR=1.47 min, m/z (M+H)+=423.0 Step 7. Tert-butyl (3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)carbamate (1h) To a mixture of compound 1g (153 mg, 0.36 mmol) in tert-butanol (7.2 mL) was added DPPA (130 mg, 0.47 mmol) and TEA (73 mg, 0.72 mmol) under N2. The mixture was stirred at RT for 30 minutes and then raised to 90° C. and stirred for another 16 hrs. After cooling, the mixture was concentrated to dryness. The residue was purified by chromatography on silica gel (elute: DCM:MeOH=50:1) to afford the title product as a brown solid (160 mg, 89% yield). LC-MS (Method 2): tR=1.71 min, m/z (M+H)+=494.0. Step 8. Tert-butyl (3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)carbamate (l1) To a solution of compound 1h (160 mg, 0.32 mmol) in MeOH (3 mL) and water (3 mL) was added NaOH (300 mg, 7.5 mmol). After stirring for 4 hrs at RT, the mixture was concentrated. The residue was diluted with water (20 mL) and extracted with EtOAc (30 mL*2). The combined organic layers were concentrated to dryness and the residue was purified by chromatography on silica gel (elute: DCM:MeOH=20:1) to afford the title product as a white solid (60 mg, 55% yield).1H NMR (400 MHz, CDCl3) δ 9.99 (s, 1H), 9.81 (s, 1H), 7.80 (s, 1H), 7.39 (d, J=4.4 Hz, 1H), 6.36 (d, J=4.4 Hz, 1H), 5.30 (br s, 1H), 2.80 (s, 6H), 1.50 (s, 9H). Step 9.3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-amine 2,2,2-trifluoroacetate (1j) To a solution of compound 1i (60 mg, 0.18 mmol) in DCM (2 mL) was added TFA (0.5 mL). After stirring for 1 hour at RT, the mixture was concentrated to dryness to afford crude title product as a brown solid (100 mg, 100% yield). LC-MS (Method 2): tR=0.309 min, m/z (M+H)+=240.0 Step 10. N-(3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (1) To a solution of compound 1j (40 mg, 0.16 mmol) and TEA (51 mg, 50 mmol) in DMF (1 mL) was added propane-1-sulfonyl chloride (28 mg, 0.5 mmol) at 0° C. After stirring for 3 hrs at RT, the mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL*3). The combined organic layers were concentrated to dryness. The residue was purified prep. HPLC (Method A) to afford the title product as a white solid (10 mg, 18% yield). LC-MS (Method 1): tR=2.71 min, m/z (M+H)+=346.0.1H NMR (400 MHz, DMSO-d6) δ 11.94 (s, 1H), 8.59 (d, J=1.6 Hz, 1H), 8.40 (s, 1H), 8.13 (s, 1H), 7.51 (s, 1H), 6.70 (d, J=1.6 Hz, 1H), 3.08 (d, J=8.8 Hz, 2H), 2.70 (s, 6H), 1.74-1.72 (m, 2H), 1.73 (d, J=6.0 Hz, 3H). Example 2 Step 1. Tert-butyl (cis-3-((5-nitro-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino)cyclobutyl)carbamate (2a) Compound 2a (380 mg) was synthesized in 89% yield by utilizing similar preparative procedure of the third step of example 1 with compound 1c (300 mg, 0.85 mmol) and tert-butyl (cis-3-aminocyclobutyl)carbamate (191 mg, 1.02 mmol) as starting materials.1H NMR (400 MHz, CDCl3) δ 9.09 (s. 1H), 9.04 (d, J=6.8 Hz, 1H), 8.06 (d, J=8.4 Hz, 2H), 7.60 (d, J=4.4 Hz, 1H), 7.30 (d, J=8.4 Hz, 2H), 6.75 (d, J=4.4 Hz, 1H), 4.73 (br s, 1H), 4.07 (br s, 1H), 3.04-2.92 (m, 2H), 2.41 (s, 3H), 2.03-1.94 (m, 2H), 1.40 (s, 9H). Step 2. Tert-butyl (cis-3-((5-amino-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino)cyclobutyl)carbamate (2b) Compound 2b (300 mg) was synthesized in 84% yield by utilizing similar preparative procedure of the fourth step of example 1 with compound 2a (380 mg, 0.76 mmol) as starting materials. LC-MS (Method 1): tR=1.63 min, m/z (M+H)+=472.2 Step 3. Tert-butyl (cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)carbamate (2c) Compound 2c (260 mg) was synthesized in 85% yield by utilizing similar preparative procedure of the fifth step of example 1 with compound 2b (300 mg, 0.64 mmol) and triethoxymethane (236 mg, 1.59 mmol) as starting materials.1H NMR (400 MHz, CDCl3) δ 9.90 (s, 1H), 8.10 (d, J=8.4 Hz, 2H), 8.05 (s, 1H), 7.80 (d, J=4.0 Hz, 1H), 7.25 (d, J=8.4 Hz, 2H), 6.77 (d, J=4.0 Hz, 1H), 4.73 (br s, 1H), 4.73-4.69 (m, 1H), 4.16-4.14 (m, 1H), 3.18-3.12 (m, 2H), 2.47-2.44 (m, 2H), 2.34 (s, 3H), 1.45 (s, 9H). Step 4. Tert-butyl (cis-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)carbamate (2d) Compound 2d (165 mg) was synthesized in 93% yield by utilizing similar preparative procedure of the eighth step of example 1 with compound 2c (260 mg, 0.54 mmol) as starting materials. LC-MS (Method 1): tR=1.47 min, m/z (M+H)+=328.1. Step 5. Cis-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutanamine 2,2,2-trifluoroacetate (2e) Compound 2e (199 mg crude) was synthesized in 87% yield by utilizing similar preparative procedure of the ninth step of example 1 with compound 2d (165 mg, 0.50 mmol) as starting materials. LC-MS (Method 1): tR=0.22 min, m/z (M+H)+=228.0. Step 6. N-(Cis-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)propane-1-sulfonamide 2 Example 2 (28.8 mg) was synthesized in 25% yield by utilizing similar preparative procedure of the final step of example 1 with compound 2e (160 mg crude, 0.35 mmol) and propane-1-sulfonyl chloride (60 mg, 0.42 mmol) as starting materials. LC-MS (Method 1): tR2.84 min, m/z (M+H)+=334.1.1H NMR (400 MHz, DMSO-d6) δ 11.85 (s, 1H), 8.57 (s, 1H), 8.35 (s, 1H), 7.58 (d, J=8.8 Hz, 1H), 7.46 (t, J=2.8 Hz, 1H), 7.58 (dd, J=3.6, 2.0 Hz, 1H), 4.95-4.90 (m, 1H), 3.86-3.80 (m, 1H), 3.10-3.24 (m, 2H), 3.00-2.96 (m, 2H), 2.54-2.47 (m, 2H), 1.73-1.67 (m, 2H), 0.99 (t, J=7.2 Hz, 3H). Example 3 Step 1. Tert-butyl (trans-3-((5-nitro-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino)cyclobutyl)carbamate (3a) Compound 3a (0.64 g) was synthesized in 89% yield by utilizing similar preparative procedure of the first step of example 2 with compound 1c (500 mg, 1.42 mmol) and tert-butyl (trans-3-aminocyclobutyl)carbamate (318 mg, 1.71 mmol) as starting materials.1H NMR (400 MHz, CDCl3) δ 9.16 (d, J=5.2 Hz, 1H), 9.11 (s, 1H), 8.06 (d, J=8.4 Hz, 2H), 7.57 (d, J=4.0 Hz, 1H), 7.31 (d, J=8.4 Hz, 2H), 6.61 (d, J=4.0 Hz, 1H), 4.81 (br s, 1H), 4.50 (br s, 1H), 4.33 (br s, 1H), 2.57-2.46 (m, 4H), 2.40 (s, 3H), 1.45 (s, 9H). Step 2. Tert-butyl (trans-3-((5-amino-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino)cyclobutyl)carbamate (3b) Compound 3b (0.45 g) was synthesized in 75% yield by utilizing similar preparative procedure of the second step of example 2 with compound 3a (0.64 g, 1.28 mmol) as starting materials. LC-MS (Method 1): tR=1.61 min, m/z (M+H)+=472.2. Step 3. Tert-butyl (trans-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)carbamate (3c) Compound 3c (160 mg) was synthesized in 35% yield by utilizing similar preparative procedure of the third step of example 2 with compound 3b (0.45 g, 0.95 mmol) and triethoxymethane (432 mg, 2.91 mmol) as starting materials.1H NMR (400 MHz, CDCl3) δ 9.91 (s, 1H), 8.11-8.07 (m, 3H), 7.79 (d, J=4.0 Hz, 1H), 7.26-7.22 (m, 2H), 6.70 (d, J=4.0 Hz, 1H), 5.21-5.14 (m, 1H), 4.90 (br s, 1H), 4.37 (br s, 1H), 2.93-2.86 (m, 2H), 2.79-2.73 (m, 2H), 2.35 (s, 3H), 1.46 (s, 9H). Step 4. Tert-butyl (trans-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)carbamate (3d) Compound 3d (100 mg) was synthesized in 92% yield by utilizing similar preparative procedure of the fourth step of example 2 with compound 3c (160 mg, 0.33 mmol) as starting materials. LC-MS (Method 1): tR=1.32 min, m/z (M+H)+=328.2. Step 5. Trans-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutanamine 2,2,2-trifluoroacetate 3e Compound 3e (60 mg crude) was synthesized in 43% yield by utilizing similar preparative procedure of the fifth step of example 2 with compound 3d (100 mg, 0.31 mmol) as starting materials. The crude product was used for next step directly without further purification. Step 6. N-(trans-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)propane-1-sulfonamide (3) Example 3 (29.6 mg) was synthesized in 34% yield by utilizing similar preparative procedure of the final step of example 2 with compound 3e (60 mg, 0.26 mmol) and propane-1-sulfonyl (45 mg, 0.31 mmol) chloride as starting materials. LC-MS (Method 1): tR=2.68 min, m/z (M+H)+=334.0.1H NMR (400 MHz, DMSO-d6) δ 11.84 (s, 1H), 8.58 (s, 1H), 8.51 (s, 1H), 7.84 (d, J=8.0 Hz, 1H), 7.46 (t, J=2.8 Hz, 1H), 6.72 (dd, J=3.2, 2.0 Hz, 1H), 5.34-5.27 (m, 1H), 4.10-4.05 (m, 1H), 3.02-2.90 (m, 4H), 2.73-2.66 (m, 2H), 1.73-1.64 (m, 2H), 0.98 (t, J=7.2 Hz, 3H). Example 4 3-Cyano-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)azetidine-1-sulfonamide (4) Example 4 (22.6 mg) was synthesized in 28% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and 3-cyanoazetidine-1-sulfonyl chloride (45 mg, 0.25 mmol) as starting materials. LC-MS (Method 1): tR=3.01 min, m/z (M+H)+=384.1.1H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 8.75 (s, 1H), 8.59 (s, 1H), 8.12 (s, 1H), 7.50 (t, J=2.8 Hz, 1H), 6.68 (dd, J=1.6, 3.2 Hz, 1H), 4.07 (t, J=8.4 Hz, 2H), 3.94 (t, J=6.0 Hz, 2H), 3.81-3.77 (m, 1H), 2.70 (s, 6H). Example 5 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-2 -methylpropane-1-sulfonamide (5) Example 5 (6.9 mg) was synthesized in 11% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (43 mg, 0.18 mmol) and 2-methylpropane-1-sulfonyl chloride (34 mg, 0.22 mmol) as starting materials. LC-MS (Method 1): tR=3.10 min, m/z (M+H)+=360.1.1H NMR (400 MHz, DMSO-d6) δ 11.97 (s, 1H), 8.62 (s, 1H), 8.44 (s, 1H), 8.16 (s, 1H), 7.54 (t, J=3.6 Hz, 1H), 6.72 (d, J=1.6 Hz, 1H), 3.04 (d, J=8.4 Hz, 2H), 2.73 (s, 6H), 2.22-2.13 (m, 1H), 1.10 (d, J=8.8 Hz, 6H). Example 6 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-2 -methoxyethanesulfonamide (6) Example 6 (21.1 mg) was synthesized in 31% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (45 mg, 0.19 mmol) and 2-methoxyethanesulfonyl chloride (36 mg, 0.23 mmol) as starting materials. LC-MS (Method 1): tR=2.48 min, m/z (M+H)+=362.1.1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.59 (s, 1H), 8.42 (s, 1H), 8.11 (s, 1H), 7.50 (t, J=2.8 Hz, 1H), 6.69 (dd, J=1.6, 3.2 Hz, 1H), 3.72 (t, J=6.4 Hz, 2H), 3.38 (t, J=6.4 Hz, 2H), 3.37 (s, 3H), 2.70 (s, 6H). Example 7 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)cyclopropanesulfonamide (7) Example 7 (15.5 mg) was synthesized in 25% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (43 mg, 0.18 mmol) and cyclopropanesulfonyl chloride (30 mg, 0.21 mmol) as starting materials. LC-MS (Method 1): tR=3.02 min, m/z (M+H)+=344.1.1H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 8.59 (s, 1H), 8.38 (s, 1H), 8.12 (s, 1H), 7.50 (t, J=2.8 Hz, 1H), 6.69 (dd, J=1.6, 3.2 Hz, 1H), 2.71 (s, 6H), 2.69-2.67 (m, 1H), 1.05-1.00 (m, 4H). Example 8 N-(3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-3-methylbutanamide (8) Example 8 (17.3 mg) was synthesized in 26% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and 3-methylbutanoyl chloride (38 mg, 0.31 mmol) as starting materials. LC-MS (Method 1): tR=2.43 min, m/z (M+H)=324.2.1H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 8.65 (s, 1H), 8.59 (s, 1H), 8.11 (s, 1H), 7.49 (t, J=3.2 Hz, 1H), 6.69 (dd, J=2.0, 3.6 Hz, 1H), 2.72 (s, 6H), 2.00-1.99 (m, 3H), 0.90 (d, J=6.4 Hz, 6H). Example 9 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)butyramide (9) Example 9 (12.4 mg) was synthesized in 14% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and butyryl chloride (53 mg, 0.32 mmol) as starting materials. The final compound was purified by prep-HPLC (Method B). LC-MS (Method 1): tR=3.06 min, m/z (M+H)+=310.2.1H NMR (400 MHz, DMSO-d6) δ 12.28 (s, 1H), 8.77 (s, 1H), 8.69 (s, 1H), 8.51 (s, 1H), 7.63 (s, 1H), 6.82 (s, 1H), 2.75 (s, 6H), 2.09 (t, J=7.6 Hz, 2H), 1.58-1.49 (m, 2H), 0.87 (t, J=7.2 Hz, 3H). Example 10 Isobutyl (3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)carbamate (10) Example 10 (2.3 mg) was synthesized in 3% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and isobutyl carbonochloridate (31 mg, 0.230 mmol) as starting materials. LC-MS (Method 1): tR=3.38 min, m/z (M+H)+=340.2.1H NMR (400 MHz, CD3OD) δ 8.49 (s, 1H), 8.03 (s, 1H), 7.38 (d, J=3.6 Hz, 1H), 6.76 (s, 1H), 3.77 (s, 2H), 2.72 (s, 6H), 1.91-1.78 (m, 1H), 0.88 (d, J=5.2 Hz, 6H). Example 11 Step 1.3-(6-(Triisopropylsilyl)imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo [1.1.1]pentan-1-amine (11a) To a solution of compound 1j (200 mg, 0.84 mmol) in DMF (2 mL) was added NaH (100 mg, 2.56 mmol, 60% in mineral oil) at 0° C. The mixture was stirred at 0° C. for 1 h. TIPSCI (240 mg, 1.28 mmol) was added to the reaction mixture at 0° C. After stirring for 4 hrs, the mixture was diluted with H2O (30 mL) and extracted with EtOAc (50 mL). The separated organic layer was concentrated and the residue was purified by prep-HPLC (Method A) to afford the title product as colorless oil (150 mg, 45% yield). LC-MS (Method 3): tR=1.94 min, m/z (M+H)+=396.2. Step 2.2-Cyano-N-(3-(6-(triisopropylsilyl)imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)acetamide (11b) To a solution of compound 11a (150 mg, 0.39 mmol), 2-cyanoacetic acid (65 mg, 0.76 mmol) in DMF (2 mL) was added HATU (433 mg, 1.14 mmol) and DIPEA (147 mg, 1.14 mmol) at RT. The mixture was stirred at RT for 2 h. The mixture was concentrated and the residue was purified by Prep-HPLC (Method A) to afford the title compound (150 mg, 83% yield) as a white solid. LC-MS (Method 3): tR=1.87 min, m/z (M+H)+=463.2. Step 3.2-Cyano-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)acetamide (11) To a solution of Compound 11b (150 mg, 0.32 mmol) in THE (4 mL) was added TBAF (0.49 ml, 0.49 mmol) at RT. The mixture was stirred at RT for 1 h. The mixture was concentrated and the residue was purified by prep-HPLC (Method A) to afford the title product as a white solid (33 mg, 34% yield). LC-MS (Method 1): tR=2.89 min, m/z (M+H)+=307.1.1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 9.18 (s, 1H), 8.59 (s, 1H), 8.12 (s, 1H), 7.50 (t, J=2.8 Hz, 1H), 6.69 (dd, J=2.0, 3.6 Hz, 1H), 3.70 (s, 2H), 2.75 (s, 6H). Example 12 N-(3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)cyclopropanecarboxamide (12) Example 12 (6 mg) was synthesized in 9% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and cyclopropanecarbonyl chloride (33 mg, 0.31 mmol) as starting materials. LC-MS (Method 1): tR=3.23 min, m/z (M+H)+=308.1.1H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 8.97 (s, 1H), 8.59 (s, 1H), 8.12 (s, 1H), 7.49 (t, J=3.2 Hz, 1H), 6.70 (dd, J=1.6, 3.2 Hz, 1H), 2.73 (s, 6H), 1.58-1.54 (m, 1H), 0.74-0.69 (m, 4H). Example 13 2-Cyclopropyl-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)acetamide (13) To a solution of 2-cyclopropylacetic acid (200 mg, 2.0 mmol) in DCM (2 mL) was dropwise added DMF (1 drop) and oxalyl chloride (508 mg, 4.0 mmol) at 0° C. The mixture was stirred at RT for 1 h. The mixture was concentrated in vacuo to give 2-cyclopropylacetyl chloride (236 mg, crude) as a white solid. To a solution of compound 1j (50 mg, 0.21 mmol) and DIPEA (270 mg, 2.1 mmol) in DMF (1 mL) was added 2-cyclopropylacetyl chloride (37.2 mg, 0.315 mmol) at 0° C. The mixture was stirred at RT for 3 hrs. The mixture was purified by prep-HPLC (Method B) to give the title compound (30 mg, 44% yield) as a white solid. LC-MS (Method 1): tR=2.96 min, m/z (M+H)+=322.2;1H NMR (400 MHz, DMSO-d6) δ 12.23 (s, 1H), 8.74 (s, 1H), 8.63 (s, 1H), 8.45 (s, 1H), 7.61 (t, J=2.8 Hz, 1H), 6.81 (s, 1H), 2.76 (s, 6H), 2.03 (d, J=7.2 Hz, 2H), 1.01-0.95 (m, 1H), 0.48-0.44 (m, 2H), 0.13 (dd, J1=5.2 Hz, J210.0 Hz, 2H). Example 14 3-Cyano-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propanamide (14) Example 14 (8.8 mg) was synthesized in 13% yield by utilizing similar preparative procedure of example 13 with compound 1j (50 mg, 0.21 mmol) and 3-cyanopropanoic acid (69 mg, 0.69 mmol) as starting materials. LC-MS (Method 1): tR=2.84 min, m/z (M+H)=321.1.1H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 8.93 (s, 1H), 8.59 (s, 1H), 8.13 (s, 1H), 7.50 (t, J=2.4 Hz, 1H), 6.70 (d, J=1.6 Hz, 1H), 2.74 (s, 6H), 2.66 (t, J=7.2 Hz, 2H), 2.44-2.39 (m, 2H). Example 15 4-Chloro-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)benzamide (15) Example 15 (15 mg) was synthesized in 19% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and 4-chlorobenzoyl chloride (54 mg, 0.31 mmol) as starting materials. LC-MS (Method 1): tR=3.48 min, m/z (M+H)=378.1.1H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 9.41 (s, 1H), 8.60 (s, 1H), 8.17 (s, 1H), 7.93 (d, J=8.4 Hz, 2H), 7.58 (d, J=8.4 Hz, 2H), 7.51 (t, J=3.2 Hz, 1H), 6.73 (dd, J=1.6, 3.2 Hz, 1H), 2.84 (s, 6H). Example 16 Isopropyl (3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)carbamate (16) Example 16 (15.3 mg) was synthesized in 21% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (55 mg, 0.23 mmol) and isopropyl chloroformate (0.23 mL, 1 mol/L, 0.23 mmol) as starting materials. LC-MS (Method 1): tR=2.79 min, m/z (M+H)+=326.2.1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.58 (s, 1H), 8.11 (s, 1H), 8.07 (br s, 1H), 7.49 (t, J=3.2 Hz, 1H), 6.68 (dd, J=1.6, 3.2 Hz, 1H), 4.83-4.77 (m, 1H), 2.67 (s, 6H), 1.21 (d, J=5.6 Hz, 6H). Example 17 3,3-Difluoro-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)cyclobutanecarboxamide (17) Example 17 (3.7 mg) was synthesized in 4% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and 3,3-difluorocyclobutanecarbonyl chloride (49 mg, 0.32 mmol) as starting materials. The title compound was purification by Prep-HPLC (Method B). LC-MS (Method 1): tR=3.11 min, m/z (M+H)=358.2.1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.95 (s, 1H), 8.59 (s, 1H), 8.12 (s, 1H), 7.49 (t, J=2.8 Hz, 1H), 6.70 (dd, J=1.6, 3.2 Hz, 1H), 2.89-2.87 (m, 1H), 2.76-2.68 (m, 10H). Example 18 4,4,4-Trifluoro-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)butanamide (18) Example 18 (29.9 mg) was synthesized in 39% yield by utilizing similar preparative procedure of example 13 with compound 1j (50 mg, 0.21 mmol) and 4,4,4-trifluorobutanoic acid (60 mg, 0.42 mmol) as starting materials. LC-MS (Method 1): tR=2.82 min, m/z (M+H)+=364.1.1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.91 (s, 1H), 8.59 (s, 1H), 8.12 (s, 1H), 7.49 (t, J=2.8 Hz, 1H), 6.69 (dd, J=2.0 Hz, 3.6 Hz, 1H), 2.73 (s, 6H), 2.57-2.52 (m, 2H), 2.42-2.39 (m, 2H). Example 19 Cyclopropylmethyl (3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)carbamate (19) Cyclopropylmethanol (30 mg, 0.42 mmol) and TEA (63 mg, 0.63 mmol) were dissolved in THE (1 mL). The resulting solution was cooled down to −30° C. followed by dropwise added a solution of bis(trichloromethyl) carbonate (124 mg, 0.42 mmol) in THE (1 mL) at the same temperature. The reaction was warmed to RT and stirred for 30 mins. Then a solution of compound 1j (50 mg, 0.21 mmol) and TEA (63 mg, 0.63 mmol) in THF/DMSO (1 mL/0.5 mL) was added to the above reaction mixture. After stirring at RT for 1 h, the mixture was diluted with DCM (10 mL) and washed with brine (5 mL). The organic layer was separated and concentrated to give a residue which was purified by Prep-HPLC (Method A) to afford the title product as a white solid (2.4 mg, 3% yield). LC-MS (Method 1): tR=8.49 min, m/z (M+H)+=338.2.1H NMR (400 MHz, CD3OD) δ 8.58 (s, 1H), 8.12 (s, 1H), 7.47 (d, J=3.6 Hz, 1H), 6.86 (d, J=3.2 Hz, 1H), 3.92 (s, 2H), 2.81 (s, 6H), 1.17-1.15 (m, 1H), 0.59-0.57 (m, 2H), 0.32-0.30 (m, 2H). Example 20 3-Cyano-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)pyrrolidine-1-sulfonamide (20) Compound 20a (100 mg, 0.76 mmol) and TEA (267 mg, 2.64 mmol) were dissolved in DCM (2 mL) followed by dropwise added a solution of SO2Cl2(122 mg, 0.91 mmol) in 6.0 mL DCM at −78° C. The mixture was stirred at −78° C. for 30 mins and warmed to RT. Aq. HCl (1 N, 10 mL) and brine (10 mL) were added to the above solution. The separated organic layer was dried over Na2SO4and filtered. The filtrate was concentrated to dryness to afford a brown oil. Then it was dissolved in DCM (0.5 ml). The resulting solution was added to a mixture of compound 1j (80 mg, 0.335 mmol) and DIPEA (130 mg, 1.005 mmol) in THF (2 mL) and DMSO (0.4 mL) at RT. The reaction was stirred at room temperature for 18 hrs. The mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL*3). The combined organic layers were dried over Na2SO4and filtered. The filtrate was concentrated to dryness to give a residue which was purified by prep-HPLC (Method A) to afford the title product as a yellow solid (12 mg, 9% yield). LC-MS (Method 1): tR=2.96 min, m/z (M+H)+=398.1.1H NMR (400 MHz, DMSO-d6) δ 11.94 (s, 1H), 8.63 (s, 1H), 8.59 (s, 1H), 8.12 (s, 1H), 7.51-7.49 (m, 1H), 6.68-6.67 (m, 1H), 3.52-3.49 (m, 2H), 3.44-3.41 (m, 1H), 3.29-3.27 (m, 2H), 2.71-2.68 (m, 6H), 2.34-2.29 (m, 1H), 2.22-2.16 (m, 1H). Example 21 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-3-methoxyazetidine-1-sulfonamide (21) Example 21 (13 mg) was synthesized in 16% yield by utilizing similar preparative procedure of example 20 with compound 1j (50 mg, 0.21 mmol) and compound 21a (150 mg, 1.21 mmol) as starting materials. LC-MS (Method 1): tR=2.87 min, m/z (M+H)+=389.2.1H NMR (400 MHz, DMSO-d6) δ 11.93 (s, 1H), 8.60 (s, 1H), 8.52 (s, 1H), 8.13 (s, 1H), 7.51 (s, 1H), 6.67 (s, 1H), 4.18 (s, 1H), 3.95 (d, J=6.4 Hz, 2H), 3.66-2.65 (m, 2H), 3.23 (s, 3H), 2.69 (s, 6H). Example 22 3-Fluoro-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)azetidine-1-sulfonamide (22) Example 22 (13 mg) was synthesized in 16% yield by utilizing similar preparative procedure of example 20 with compound 1j (50 mg, 0.21 mmol) and compound 22a (200 mg, 1.79 mmol) as starting materials. LC-MS (Method 1): tR=2.88 min, m/z (M+H)+=377.1.1H NMR (400 MHz, DMSO-d6) δ 11.94 (s, 1H), 8.69 (s, 1H), 8.59 (s, 1H), 8.13 (s, 1H), 7.51 (t, J=3.2 Hz, 1H), 6.67 (d, J=0.8 Hz, 1H), 5.47-5.44 (m, 0.5H), 5.32-5.30 (m, 0.5H), 4.15-4.06 (m, 2H), 3.93-3.84 (m, 2H), 2.69 (s, 6H). Example 23 3,3-difluoro-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)azetidine-1-sulfonamide (23) Example 23 (8.0 mg) was synthesized in 10% yield by utilizing similar preparative procedure of example 20 with compound 1j (50 mg, 0.21 mmol) and compound 23a (200 mg, 1.54 mmol) as starting materials. LC-MS (Method 1): tR=3.15 min, m/z (M+H)+=395.1.1H NMR (400 MHz, DMSO-d6) δ 11.93 (s, 1H), 8.93 (s, 1H), 8.60 (s, 1H), 8.13 (s, 1H), 7.51 (t, J=2.8 Hz, 1H), 6.67 (dd, J=1.6, 3.2 Hz, 1H), 4.29 (t, J=12.8 Hz, 4H), 2.68 (s, 6H). Example 24 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-N′-dimethyl-1-sulfonamide (24) To a solution of compound 1j (60 mg, 0.25 mmol) and DIPEA (130 mg, 1.00 mmol) in THE (1.5 mL) and DMSO (0.5 mL) was dropwise added a solution of dimethylsulfamoyl chloride (36 mg, 0.25 mmol) in 0.5 mL THE at 0° C. The mixture was stirred at 25° C. for 14 hrs. The mixture was diluted with H2O (15 mL) and extracted with DCM (20 mL*2). The combined organic layers were concentrated and the residue was purified by prep-HPLC (Method A) to afford the title product as a white solid (15 mg, 17% yield). LC-MS (Method 1): tR=2.59 min, m/z (M+H)+=347.1.1H NMR (400 MHz, CD3OD) δ 8.48 (s, 1H), 8.02 (s, 1H), 7.38 (d, J=3.6 Hz, 1H), 6.71 (d, J=3.2 Hz, 1H), 2.73 (s, 6H), 2.68 (s, 6H). Example 25 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-N′-methyl-N’-ethyl-1-sulfonamide (25) To a mixture consisting of compound 1j (70 mg, 0.29 mmol), Et3N (148 mg, 1.46 mmol), K2CO3(404 mg, 2.93 mmol) and ACN (3 mL) was dropwise added a solution of ethyl(methyl)sulfamoyl chloride (46.0 mg, 0.29 mmol) in 0.5 mL ACN at 0° C. The mixture was stirred at 30° C. for 14 hrs. The mixture was diluted with H2O (20 mL) and extracted with DCM (40 mL*2). The combined organic layers were concentrated to dryness and the residue was purified by prep-HPLC (Method A) to afford the title product as a white solid (8.0 mg, 9% yield). LC-MS (Method 1): tR=2.96 min, m/z (M+H)+=361.1.1H NMR (400 MHz, CD3OD) δ 8.48 (s, 1H), 8.02 (s, 1H), 7.38 (d, J=3.6 Hz, 1H), 6.71 (d, J=3.6 Hz, 1H), 3.17 (q, J=7.2 Hz, 2H), 2.75 (s, 3H), 2.68 (s, 6H), 1.14 (t, J=7.2 Hz, 3H). Example 26 N-(3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-3-methoxypropane-1-sulfonamide (26) Example 26 (15 mg) was synthesized in 12% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (80 mg, 0.33 mmol) and 3-methoxypropane-1-sulfonyl chloride (58 mg, 0.33 mmol) as starting materials. LC-MS (Method 1): tR=3.11 min, m/z (M+H)=376.1.1H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 8.59 (s, 1H), 8.11 (s, 1H), 7.50 (s, 1H), 6.69 (s, 1H), 3.45 (s, 4H), 3.09 (s, 3H), 2.68 (s, 6H), 1.93 (s, 2H). Example 27 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)ethanesulfonamide (27) Example 27 (11 mg) was synthesized in 16% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and ethanesulfonyl chloride (35 mg, 0.27 mmol) as starting materials. LC-MS (Method 1): tR=3.36 min, m/z (M+H)f=332.1.1H NMR (400 MHz, CD3OD) δ 8.87 (s, 1H), 8.68 (s, 1H), 7.73 (d, J=3.6 Hz, 1H), 7.10 (d, J=3.6 Hz, 1H), 3.16 (q, J=7.2 Hz, 2H), 2.90 (s, 6H), 1.41 (t, J=7.2 Hz, 3H). Example 28 4-Chloro-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)benzenesulfonamide (28) Example 28 (12.4 mg) was synthesized in 14% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and 4-chlorobenzene-1-sulfonyl chloride (58 mg, 0.27 mmol) as starting materials. LC-MS (Method 1): tR=2.94 min, m/z (M+H)+=414.0.1H NMR (400 MHz, CD3OD) δ 8.45 (s, 1H), 7.94 (s, 1H), 7.85 (d, J=8.4 Hz, 2H), 7.54 (d, J=8.4 Hz, 2H), 7.36 (d, J=3.6 Hz, 1H), 6.57 (d, J=3.6 Hz, 1H), 2.52 (s, 6H). Example 29 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-1-methyl-1H-pyrazole-4-sulfonamide (29) Example 29 (5.5 mg) was synthesized in 7% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and 1-methyl-1H-pyrazole-4-sulfonyl chloride (49 mg, 0.27 mmol) as starting materials. LC-MS (Method 1): tR=3.41 min, m/z (M+H)+=384.1. H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.72 (s, 1H), 8.57 (s, 1H), 8.34 (s, 1H), 8.07 (s, 1H), 7.81 (s, 1H), 7.49 (t, J=3.2 Hz, 1H), 6.59-6.58 (m, 1H), 3.91 (s, 3H), 2.56 (s, 6H). Example 30 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane-2-sulfonamide (30) Example 30 (3.4 mg) was synthesized in 5% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and propane-2-sulfonyl chloride (39 mg, 0.27 mmol) as starting materials. LC-MS (Method 1): tR=2.50 min, m/z (M+H)+=346.1.1H NMR (300 MHz, DMSO-d6) δ 11.96 (s, 1H), 8.62 (s, 1H), 8.37 (s, 1H), 8.14 (s, 1H), 7.53 (t, J=2.7 Hz, 1H), 6.72 (d, J=1.2 Hz, 1H), 3.30-3.21 (m, 1H), 2.72 (s, 6H), 1.32 (d, J=6.9 Hz, 6H). Example 31 N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)butane-1-sulfonamide (31) Example 31 (10 mg) was synthesized in 13% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and butane-1-sulfonyl chloride (66 mg, 0.42 mmol) as starting materials. LC-MS (Method 1): tR=3.13 min, m/z (M+H)+=360.1.1H NMR (400 MHz, CD3OD) δ 8.60 (s, 1H), 8.15 (s, 1H), 7.50 (d, J=3.6 Hz, 1H), 6.85 (d, J=3.2 Hz, 1H), 3.18-3.14 (m, 2H), 2.84 (s, 6H), 1.86-1.80 (m, 2H), 1.58-1.52 (m, 2H), 1.02 (t, J=7.2 Hz, 3H). Example 32 3,3,3-Trifluoro-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (32) Example 32 (4.7 mg) was synthesized in 6% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and 3,3,3-trifluoropropane-1-sulfonyl chloride (62 mg, 0.32 mmol) as starting materials. The final compound was purified by prep-HPLC (Method B) to afford the title compound. LC-MS (Method 1): tR=3.17 min, m/z (M+H)+=400.1.1H NMR (400 MHz, CD3OD) δ 8.67 (s, 1H), 8.41 (s, 1H), 7.55 (d, J=3.6 Hz, 1H), 6.92 (d, J=3.6 Hz, 1H), 3.31-3.27 (m, 2H), 2.79 (s, 6H), 2.67-2.61 (m, 2H).19F NMR (376 MHz, CD3OD) δ-67.55. Example 33 1-Cyano-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)methanesulfonamide (33) Example 33 (4.5 mg) was synthesized in 6% yield by utilizing similar preparative procedure of the final step of example 1 with compound 1j (50 mg, 0.21 mmol) and cyanomethanesulfonyl chloride (45 mg, 0.32 mmol) as starting materials. LC-MS (Method 1): tR=3.32 min, m/z (M+H)+=343.1.1H NMR (400 MHz, DMSO-d6) δ 11.94 (s, 1H), 8.59 (s, 1H), 8.12 (s, 1H), 7.50 (t, J=2.8 Hz, 1H), 6.75-6.74 (m, 1H), 4.87 (s, 2H), 2.75 (s, 6H). Example 34 1-(3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-3-propylurea (34) To a well stirred solution consisting of compound 1j (50 mg, 0.21 mmol), THE (0.6 mL) and DMSO (0.2 mL) was added 1-isocyanatopropane (36 mg, 0.42 mmol). The mixture was stirred at 30° C. for 3 hrs. The mixture was concentrated to dryness to give a residue which was purified by prep-HPLC (Method A) to give the title product (27.3 mg, 40% yield) as a light yellow solid. LC-MS (Method 1): tR=2.80 min, m/z (M+H)+=325.2.1H NMR (400 MHz, DMSO-d6) δ 11.89 (s, 1H), 8.58 (s, 1H), 8.10 (s, 1H), 7.48 (t, J=2.8 Hz, 1H), 6.75-6.70 (m, 2H), 5.91 (t, J=5.6 Hz, 1H), 2.99-2.94 (m, 2H), 2.66 (s, 6H), 1.42-1.37 (m, 2H), 0.85 (t, J=7.2 Hz, 3H). Example 35 1-Cyclopropyl-3-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)urea (35) Example 35 (29.6 mg) was synthesized in 44% yield by utilizing similar preparative procedure of example 34 with compound 1j (50 mg, 0.21 mmol) and isocyanatocyclopropane (35 mg, 0.42 mmol) as starting materials. LC-MS (Method 1): tR=2.83 min, m/z (M+H)+=323.1.1H NMR (400 MHz, DMSO-d6) δ 11.89 (s, 1H), 8.58 (s, 1H), 8.10 (s, 1H), 7.49 (t, J=3.2 Hz, 1H), 6.77 (s, 1H), 6.71 (dd, J=2.0 Hz, 3.6 Hz, 1H), 6.21 (d, J=2.4 Hz, 1H), 2.67 (s, 6H), 2.45-2.41 (m, 1H), 0.60-0.56 (m, 2H), 0.38-0.34 (m, 2H). Example 36 1-(3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-3-isobutylurea (36) Example 36 (17.4 mg) was synthesized in 25% yield by utilizing similar preparative procedure example 34 with compound 1j (50 mg, 0.21 mmol) and 1-isocyanato-2-methylpropane (41 mg, 0.42 mmol) as starting materials. LC-MS (Method 1): tR=3.23 min, m/z (M+H)+=339.2.1H NMR (400 MHz, DMSO-d6) δ 11.89 (s, 1H), 8.58 (s, 1H), 8.10 (s, 1H), 7.48 (t, J=3.2 Hz, 1H), 6.73 (s, 1H), 6.72 (dd, J=1.6, 3.2 Hz, 1H), 5.95 (t, J=5.6 Hz, 1H), 2.84 (t, J=6.4 Hz, 2H), 2.66 (s, 6H), 1.67-1.61 (m, 1H), 0.84 (d, J=6.8 Hz, 6H). Example 37 3,3-Difluoro-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)azetidine-1-carboxamide (37) To a well stirred solution consisting of compound 1j (50 mg, 0.21 mmol), TEA (74 mg, 0.73 mmol), DCM (1.0 mL) and DMSO (0.5 mL) was added CDI (68 mg, 0.42 mmol) in one portion. The mixture was stirred at RT for 2 hrs. The resulting reaction solution was added to a solution of 3,3-difluoroazetidine hydrochloride (95 mg, 0.73 mmol) and TEA (84 mg, 0.84 mmol) in DCM (1 mL). The mixture was stirred at RT overnight. The mixture was concentrated and the residue was purified by prep-HPLC (Method A) to afford the title product as a white solid (11.1 mg, 15% yield). LC-MS (Method 1): tR=2.79 min, m/z (M+H)+=359.2.1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.59 (s, 1H), 8.12 (s, 1H), 7.75 (s, 1H), 7.49 (s, 1H), 6.67 (s, 1H), 4.25 (t, J=12.8 Hz, 4H), 2.69 (s, 6H).19F NMR (376 MHz, DMSO-d6) δ −99.31. Example 38 N-(3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-3-methoxyazetidine-1-carboxamide (38) Example 38 (26.6 mg) was synthesized in 36% yield by utilizing similar preparative procedure of example 37 with compound 1j (50 mg, 0.21 mmol) and 3-methoxyazetidine hydrochloride (90 mg, 0.73 mmol) as starting materials. LC-MS (Method 1): tR=2.77 min, m/z (M+H)+=353.2.1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.58 (s, 1H), 8.11 (s, 1H), 7.49 (s, 1H), 7.31 (s, 1H), 6.67 (d, J=2.0 Hz, 1H), 4.14 (br s, 1H), 4.01-3.98 (m, 2H), 3.65-3.61 (m, 2H), 3.20 (s, 3H), 2.66 (s, 6H). Example 39 3-Cyano-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)pyrrolidine-1-carboxamide (39) Example 39 (12 mg) was synthesized in 16% yield by utilizing similar preparative procedure of example 37 with compound 1j (50 mg, 0.21 mmol) and pyrrolidine-3-carbonitrile hydrochloride (97 mg, 0.73 mmol) as starting materials. LC-MS (Method 1): tR=2.82 min, m/z (M+H)+=362.1.1H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 8.58 (s, 1H), 8.12 (s, 1H), 7.49 (t, J=3.2 Hz, 1H), 7.25 (s, 1H), 6.68 (dd, J=1.6, 3.2 Hz, 1H), 3.60-3.56 (m, 1H), 3.49-3.34 (m, 4H), 2.69 (s, 6H), 2.27-2.22 (m, 1H), 2.16-2.11 (m, 1H). Example 40 1-(2-Cyano-2-methylpropyl)-3-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)urea (40) Example 40 (21.4 mg) was synthesized in 28% yield by utilizing similar preparative procedure of example 37 with compound 1j (50 mg, 0.21 mmol) and 3-amino-2,2-dimethylpropanenitrile 4-methylbenzenesulfonate (198 mg, 0.73 mmol) as starting materials. LC-MS (Method 1): tR=3.26 min, m/z (M+H)+=364.2.1H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 8.58 (s, 1H), 8.11 (s, 1H), 7.49 (s, 1H), 6.96 (s, 1H), 6.72 (s, 1H), 6.38-6.35 (m, 1H), 3.22 (d, J=6.4 Hz, 2H), 2.68 (s, 6H), 1.26 (s, 6H). Example 41 1-(3-(Imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)-3-(2,2,2-trifluoroethyl)urea (41) Example 41 (19.6 mg) was synthesized in 21% yield by utilizing similar preparative procedure of example 37 with compound 1j (60 mg, 0.25 mmol) and 2,2,2-trifluoroethanamine (87 mg, 0.88 mmol) as starting materials. LC-MS (Method 1): tR=3.09 min, m/z (M+H)+=365.1.1H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 8.58 (s, 1H), 8.10 (s, 1H), 7.49 (t, J=2.8 Hz, 1H), 7.17 (s, 1H), 6.71 (dd, J=1.6, 3.2 Hz, 1H), 6.58 (t, J=6.4 Hz, 1H), 3.88-3.79 (m, 2H), 2.68 (t, J=8.0 Hz, 6H).19F NMR (376 MHz, DMSO-d6) δ −71.54. Example 42 2-Cyano-2-methylpropyl (3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)carbamate (42) Example 42 (2.2 mg) was synthesized in 3% yield by utilizing similar preparative procedure of example 19 with compound 1j (50 mg, 0.21 mmol) and 3-hydroxy-2,2-dimethylpropanenitrile (50 mg, 0.73 mmol) as starting materials. LC-MS (Method 1): tR=3.23 min, m/z (M+H)+=365.2.1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.59 (s, 1H), 8.47 (s, 1H), 8.12 (s, 1H), 7.49 (s, 1H), 6.69 (s, 1H), 4.04 (s, 2H), 2.71 (s, 6H), 1.35 (s, 6H). Example 43 1-(2-Cyanoethyl)-3-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)urea (43) Example 43 (12.8 mg) was synthesized in 18% yield by utilizing similar preparative procedure example 37 with compound 1j (50 mg, 0.21 mmol) and 3-aminopropanenitrile (51 mg, 0.73 mmol) as starting materials. LC-MS (Method 1): tR=2.60 min, m/z (M+H)+=336.2. H NMR (400 MHz, CD30D) δ 8.60 (s, 1H), 8.13 (s, 1H), 7.49 (d, J=3.6 Hz, 1H), 6.91 (d, J=3.6 Hz, 1H), 3.43 (t, J=6.4 Hz, 2H), 2.84 (s, 6H), 2.68 (t, J=6.8 Hz, 2H). Example 44 Step 1. Methyl cis-3-aminocyclobutane-1-carboxylate TFA (44b) To a solution of compound 44a (500 mg, 2.18 mmol) in 5 mL of DCM was added TFA (2.5 mL) at 0° C. The solution was stirred at 0° C. to 5° C. for 1.5 hrs. The solution was concentrated to afford the title product as colorless oil (530 mg, 100% yield).1H NMR (400 MHz, DMSO-d6) δ 8.05 (br s, 2H), 3.62-3.59 (m, 4H), 3.02-2.93 (m, 1H), 2.46-2.39 (m, 2H), 2.29-2.21 (m, 2H). Step 2. Methyl cis-3-((5-nitro-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino) cyclobutanecarboxylate (44c) Compound 44c (760 mg) was synthesized in 78% yield by utilizing similar preparative procedure of the third step of example 1 with 4-chloro-5-nitro-1-tosyl-1H-pyrrolo[2,3-b]pyridine (765 mg, 2.18 mmol) and compound 44b (530 mg, 2.18 mmol) as starting materials.1H NMR (400 MHz, DMSO-d6) δ 8.90-8.87 (m, 2H), 8.00 (d, J=8.4 Hz, 2H), 7.79 (d, J=4.4 Hz, 1H), 7.44 (d, J=8.4 Hz, 2H), 7.13 (d, J=4.0 Hz, 1H), 4.59-4.53 (m, 1H), 3.61 (s, 3H), 3.04-2.97 (m, 1H), 2.75-2.68 (m, 2H), 2.36 (s, 3H), 2.33-2.26 (m, 2H). Step 3. Methyl cis-3-((5-amino-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino) cyclobutanecarboxylate (44d) Compound 44d (640 mg) was synthesized in 92% yield by utilizing similar preparative procedure of the fourth step of example 1 with compound 44c (750 mg, 1.69 mmol) as starting materials. LC-MS (Method 3): tR=1.55 min, m/z (M+H)+=415.1. Step 4. Methyl cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl) cyclobutanecarboxylate (44e) Compound 44e (550 mg) was synthesized in 84% yield by utilizing similar preparative procedure of the fifth step of example 1 with compound 44d (640 mg, 1.54 mmol) and triethoxymethane (571 mg, 3.86 mmol) as starting materials.1H NMR (400 MHz, DMSO-d6) δ 8.73 (s, 1H), 8.58 (s, 1H), 8.01 (d, J=8.4 Hz, 2H), 7.97 (d, J=4.4 Hz, 1H), 7.40 (d, J=8.0 Hz, 2H), 7.29 (d, J=3.6 Hz, 1H), 5.22-5.17 (m, 1H), 3.34 (s, 3H), 3.19-3.12 (m, 1H), 2.89-2.82 (m, 2H), 2.74-2.66 (m, 2H), 2.32 (m, 3H). Step 5. (Cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)methanol (44f) To a solution of compound 44e (550 mg, 1.30 mmol) in 5 mL dry THE was added LiAlH4(73.9 mg, 1.94 mmol) at 0° C. The mixture was stirred for 1.5 hrs at RT. After cooling down to 0° C., the reaction mixture was quenched with 0.1 mL of water, 0.2 mL of 10% aq. NaOH, followed by 0.3 mL of water. The mixture was dried over Na2SO4and filtered. The filtrate was concentrated to afford the crude title product as a white solid (450 mg, 88% yield). LC-MS (Method 3): tR=1.43 min, m/z (M+H)+=397.1. Step 6. (Cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)methyl methanesulfonate (44g) Compound 44f (450 mg, 1.13 mmol) and Et3N (344 mg, 3.40 mmol) were dissolved in DCM (15 mL) followed by the addition of MsCl (196 mg, 1.70 mmol) at 0° C. After stirring for 1 hour at RT, the reaction mixture was diluted with water (100 mL) and extracted with EtOAc (150 mL). The organic layer was separated, washed with brine (40 mL), dried over Na2SO4and filtered. The filtrate was concentrated to dryness to afford the crude title compound (539 mg, 100% yield) as a yellow solid. LC-MS (Method 3): tR=1.35 min, m/z (M+H)+=475.1. Step 7. S-((cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)methyl) ethanethioate (44h) Compound 44g (535 mg, 1.13 mmol) and potassium thioacetate (386 mg, 3.38 mmol) were mixed in DMF (16 mL) and then heated at 50° C. for 5 hrs. The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (150 mL). The separated organic layer was concentrated. The residue was purified by reverse chromatography (ACN in water from 5-95%) to afford the crude title compound (338 mg, 66% yield) as a yellow solid. LC-MS (Method 3): tR=1.53 min, m/z (M+H)+=455.1. Step 8. (Cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl) methanesulfonic acid (44i) An aqueous hydrogen peroxide (0.55 mL, 30%) was added dropwise to a stirred suspension of compound 44h (330 mg, 0.73 mmol) in formic acid (4 mL). The resulting mixture was stirred at room temperature for 1 hour. Then the reaction mixture was concentrated to afford the title compound (334 mg, 100% yield) as a white solid. LC-MS (Method 3): tR=0.81 min, m/z (M+H)+=461.1. Step 9. (Cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl) methanesulfonyl chloride (44j) To a mixture of compound 44i (330 mg, 0.73 mmol) in DCM (50 mL) and DMF (1.0 mL) was added thionyl chloride (930 mg, 7.82 mmol). Then the reaction mixture was heated at 50° C. for 3 hours. The mixture was concentrated to afford title compound (334 mg, 100%) as a yellow solid. A small amount of the reaction solution was mixed with MeOH for analysis. LC-MS (Method 3): tR=1.60 min, m/z (M+H)+=475.1. Step 10. N-methyl-1-(cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)methanesulfonamide (44k) To a solution consisting of CH3NH2.HCl (34 mg, 0.50 mmol), TEA (127 mg, 1.25 mmol) and DCM (2 mL) was added compound 44j (200 mg, 0.42 mmol) at 0° C. The mixture was stirred at RT for 1.5 hrs. The mixture was diluted with water (30 mL) and extracted with DCM (40 mL). The separated organic layer was washed with water (30 mL*2) and concentrated to afford the crude title product as a white solid (25 mg, 13% yield). LC-MS (Method 3): tR=1.43 min, m/z (M+H)+=474.1. Step 11.1-(Cis-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)-N-methylmethanesulfonamide (44) Compound 44k (22 mg, 0.05 mmol) and LiOH H2O (10 mg, 0.05 mmol) were dissolved in a mixture of i-PrOH and H2O (2.5 ml, V:V=4:1). The above solution was stirred at 60° C. for 24 hrs. The reaction mixture was diluted with water (10 mL) and extracted with EtOAc (20 mL). The separated organic layer was concentrated and the residue was purified by prep-HPLC (Method A) to afford the title compound (3.7 mg, 25% yield) as a white solid. LC-MS (Method 1): tR=2.37 min, m/z (M+H)+=320.1.1H NMR (400 MHz, DMSO-d6) δ 8.59 (s, 1H), 8.42 (s, 1H), 7.46 (d, J=3.6 Hz, 1H), 6.88 (d, J=3.6 Hz, 1H), 5.26-4.86 (m, 1H), 3.38-3.33 (m, 2H), 3.09-3.02 (m, 2H), 2.86-2.80 (m, 1H), 2.76 (s, 3H), 2.54-2.47 (m, 2H). Example 45 Step 1.1-(((Cis-3-(6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)methyl) sulfonyl)azetidine-3-carbonitrile (45a) Compound 45a (80 mg) was synthesized in 64% yield by utilizing similar preparative procedure of the tenth step of example 44 with compound 44j (120 mg, 0.25 mmol) and azetidine-3-carbonitrile hydrochloride (32 mg, 0.26 mmol) as starting materials. LC-MS (Method 3): tR=1.50 min, m/z (M+H)+=525.1. Step 2.1-(((Cis-3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)cyclobutyl)methyl) sulfonyl) azetidine-3-carbonitrile (45) A mixture of compound 45a (40 mg, 0.08 mmol) and Mg powder (73 mg, 3.05 mmol) in MeOH (2 mL) was placed in an ultrasonic bath for 1.5 hours. The mixture was filtered and the filtrate was concentrated. The residue was purified by prep-HPLC (Method A) to afford the title product as an off-white solid (1.4 mg, 5% yield). LC-MS (Method 1): tR=3.04 min, m/z (M+H)+=371.1.1H NMR (400 MHz, CD3OD) δ 8.59 (s, 1H), 8.42 (s, 1H), 7.46 (d, J=3.6 Hz, 1H), 6.90 (d, J=3.6 Hz, 1H), 5.28-4.86 (m, 1H), 4.29-4.24 (m, 2H), 4.16-4.13 (m, 2H), 3.75-3.69 (m, 1H), 3.50-3.43 (m, 2H), 3.07-3.01 (m, 2H), 2.89-2.85 (m, 1H), 2.56-2.48 (m, 2H). Example 46 Step 1. (3,3-Difluorocyclobutyl)methyl 4-methylbenzenesulfonate (46b) To a mixture of (3,3-difluorocyclobutyl)methanol (1 g, 8.19 mmol), DMAP (100 mg, 0.82 mmol) and TEA (1.24 g, 12.29 mmol) in DCM (10 mL) was added TsC1 (889 mg, 9.83 mmol) at 0° C. and allowed to warm to RT. After stirring overnight at RT, the mixture was diluted with 20 mL of DCM and washed with water (10 mL), brine (10 mL) and dried over Na2SO4. The mixture was filtered and filtrate was concentrated to afford crude title compound (1.89 g crude, 79% yield) as yellow oil. Step 2. Benzyl ((3,3-difluorocyclobutyl)methyl)sulfane (46c) To a mixture of compound 46b (100 mg, 0.36 mmol) and benzyl carbamimidothioate hydrochloride (88 mg, 0.43 mmol) in DMF (0.5 mL) was added NaOH (36 mg, 0.90 mmol) in H2O (0.5 mL). The mixture was stirred at 60° C. overnight. The mixture was diluted with H2O (10 mL) and extracted with EtOAc (30 mL*3). The combined organic layers were dried Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (PE:EA=80:1) to give the crude title product as yellow oil (82 mg, 99% yield).1H NMR (400 MHz, CDCl3) δ 7.34-7.28 (m, 5H), 3.70 (s, 2H), 2.71-2.60 (m, 2H), 2.55 (d, J=7.2 Hz, 2H), 2.32-2.14 (m, 3H). Step 3.1-(3,3-Difluorocyclobutyl)-N-(3-(imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)methanesulfonamide (46) To a mixture consisting of compound 46c (82 mg, 0.36 mmol), DCM (1.5 mL) and H2O (0.4 mL) was added SO2Cl2(418 mg, 3.09 mmol) at −5° C. The mixture was stirred at 0° C. for 30 minutes. Then ice-water (15 mL) was added and the mixture was extracted with DCM (20 mL*2). The combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was dissolved into THE (0.5 mL) and the solution was added to a mixture of 1j (100 mg, 0.44 mmol) and DIPEA (170 mg, 1.31 mmol) in THE (1.5 mL) and DMSO (1 mL) at 0° C. The mixture was stirred at room temperature for 2 hours. The mixture was diluted with H2O (15 mL) and extracted with EtOAc (20 mL*2). The combined organic layers were concentrated. The residue was purified by prep-TLC (DCM: MeOH=25: 1) and prep-HPLC (Method A) to afford the title product as a yellow solid (14 mg, 8% yield). LC-MS (Method 1): tR=2.98 min, m/z (M+H)+=408.1.1H NMR (400 MHz, DMSO-d6) δ 11.93 (s, 1H), 8.60 (s, 1H), 8.50 (s, 1H), 8.12 (s, 1H), 7.51 (t, J=2.8 Hz, 1H), 6.71 (t, J=1.6 Hz, 1H), 3.39-3.38 (m, 2H), 2.82-2.75 (m, 2H), 2.72 (s, 6H), 2.62-2.53 (m, 3H). Example 47 Step 1. Tert-butyl (3-(propylsulfonamido)bicyclo[1.1.1]pentan-1-yl)carbamate (47b) To a solution of compound 47a (1.0 g, 5.04 mmol) and Et3N (1.5 g, 15.1 mmol) in DCM (1.5 mL) was added propane-1-sulfonyl chloride (1.0 g, 7.56 mmol) at 0° C. After stirring at RT for 3 hrs, the mixture was diluted with water (100 mL) and extracted with DCM (100 mL*2). The combined organic phases were washed with brine (100 mL*2), dried over Na2SO4, and filtered. The filtrate was concentrated to afford the title compound (1.45 g, 97% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 8.00 (s, 1H), 7.55 (s, 1H), 2.94-2.90 (m, 2H), 2.05 (s, 6H), 1.69-1.59 (m, 2H), 1.37 (s, 9H) 1.03-0.91 (m, 3H). Step 2. N-(3-aminobicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (47c) To a solution of compound 47b (1.45 g, 4.77 mmol) in EtOAc (20 mL) was added HCl(g) in EtOAc (2 M, 20 mL) at 0° C. The reaction mixture was stirred at RT for 3 hrs. The mixture was concentrated in vacuo to give the title compound (1.2 g, crude, yield ˜100%) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 9.02 (s, 3H), 8.25 (s, 1H), 2.98-2.95 (m, 2H), 2.13 (s, 6H), 1.73-1.61 (m, 2H), 1.03-0.96 (m, 3H). Step 3. N-(3-((5-nitro-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (47d) Compound 47d (2.4 g) was synthesized in 100% yield by utilizing similar preparative procedure of the third step of example 1 with compound 47c (1.2 g, 4.99 mmol) and compound 1c (1.6 g, 4.54 mmol) as starting materials. LC-MS (Method 3): tR=1.68 min, m/z (M+H)+=520.1 Step 4. N-(3-((5-amino-1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)amino)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (47e) To a solution consisting of 47d (2.4 g, 4.62 mmol), NH4Cl (1.2 g, 23.1 mmole), MeOH (900 mL) and H2O (300 mL) were added Fe powder (905 mg, 16.2 mmol) at RT. The reaction mixture was stirred at 80° C. for 2 hrs. After cooling to RT, the mixture was filtered and the filter cake was washed with MeOH (20 mL). The filtrate was concentrated in vacuo to afford the title compound (2.2 g, 95.6%, crude) as a brown solid. LC-MS (Method 3): tR=1.44 min, m/z (M+H)+=490.1. Step 5. (R)-N-(3-(2-(1-hydroxyethyl)-6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (47f) (R)-2-hydroxypropanamide (136 mg, 1.53 mmol) and triethyloxonium tetrafluoroborate (291 mg, 1.53 mmol) were dissolved in THE (5 mL) and the resulting mixture was stirred at RT for 30 mins under N2. Then 47e (150 mg, 0.31 mmol) in EtOH (5 mL) was added to the reaction mixture. The mixture was stirred for 2 hrs at 85° C. After cooling to RT, the mixture was diluted with water (100 mL) and extracted with EtOAc (100 mL*2). The combined organic layers were concentrated and the residue was purified by prep-TLC (DCM:MeOH=10:1) to afford the desired compound (55 mg, yield 50%) as a brown solid.1H NMR (400 MHz, DMSO-d6) δ 8.72 (s, 1H), 8.36 (s, 1H), 8.00 (s, 1H), 8.99 (d, J=8.0 Hz, 2H), 7.40 (d, J=8.0 Hz, 2H), 7.07 (d, J=4.0 Hz, 1H), 5.53 (d, J=7.2 Hz, 1H), 5.07-5.04 (m, 1H), 3.09-3.05 (m, 2H), 2.81 (s, 6H), 2.32 (s, 3H), 1.75-1.70 (m, 2H), 1.61 (d, J=6.0 Hz, 3H), 1.02 (t, J=7.2 Hz, 3H). Step 6. (R)-N-(3-(2-(1-hydroxyethyl)imidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (47) To a solution of compound 47f (55 mg, 0.10 mmol) in a mixture of MeOH and H2O (5.5 mL, V:V=1: 5) was added NaOH (12 mg, 0.30 mmol) in one portion. After stirring for 25 hours at 30° C., the reaction mixture was diluted with water (20 mL) and washed with DCM (20 mL*2). The separated aqueous layer was concentrated to dryness and the residue was purified by prep-HPLC (Method A) to afford title compound (10 mg, 16% yield) as a white solid. LC-MS (Method 1): tR=2.98 min, m/z (M+H) *=390.2.1H NMR (400 MHz, CD3OD) δ 8.58 (s, 1H), 7.48 (s, 1H), 6.88 (s, 1H), 5.26 (s, 1H), 3.13 (s, 2H), 3.00 (s, 6H), 1.89-1.88 (m, 2H), 1.78 (s, 3H), 1.12 (s, 3H). Example 48 Step 1. N-(3-(2-methyl-6-tosylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (48a) Compound 48a (89 mg) was synthesized in 57% yield by utilizing similar preparative procedure of the fifth step of Example 1 with compound 47e (150 mg, 0.31 mmol) and 1,1,1-triethoxyethane (124 mg, 0.76 mmol) as starting materials. LC-MS (Method 3): tR=1.52 min, m/z (M+H)+=514.1. Step 2. N-(3-(2-methylimidazo[4,5-d]pyrrolo[2,3-b]pyridin-1(6H)-yl)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (48) To a solution of compound 48a (85 mg, 0.17 mmol) in MeOH and H2O (3.3 mL, V: V=1: 10) was added NaOH (20 mg, 0.50 mmol) in one portion. After stirring at 30° C. for 20 hrs, the reaction mixture was diluted with water (20 mL) and washed with DCM (20 mL*2). The separated aqueous layers were concentrated to dryness and residue was purified by prep-HPLC (Method A) to afford the title compound (5 mg, 8% yield) as a white solid. LC-MS (Method 1): tR=3.22 min, m/z (M+H)+=360.2.1H NMR (400 MHz, CD3OD) δ 8.36 (s, 1H), 7.37 (d, J=3.6 Hz, 1H), 6.73 (d, J=3.2 Hz, 1H), 3.05-3.01 (m, 2H), 2.84 (s, 6H), 2.62 (s, 3H), 1.80-1.75 (m, 2H), 1.01 (t, J=7.6 Hz, 3H). Example 49 Step 1.4-Chloro-7-tosyl-7H-pyrrolo[2,3-d]pyrimidine (49b) Compound 49b (15 g) was synthesized in 92% yield by utilizing similar preparative procedure of the first step of compound 1 with 49a (10 g, 65 mmol) and TsC1 (14.8 g, 78 mmol) as starting materials.1H NMR (400 MHz, CDCl3) δ 8.76 (s, 1H), 8.09 (d, J=8.4 Hz, 2H), 7.77 (d, J=4.0 Hz, 1H), 7.32 (d, J=8.4 Hz, 2H), 6.70 (d, J=4.0 Hz, 1H), 2.40 (s, 3H). Step 2. Tert-butyl 3-((7-tosyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)bicyclo[1.1.1]pentane-1-carboxylate (49c) Compound 49b (294 mg, 0.95 mmol), tert-butyl 3-aminobicyclo[1.1.1]pentane-1-carboxylate (210 mg, 1.14 mmol) and DIPEA (247 mg, 1.91 mmol) were dissolved in NMP (1.5 mL). The above mixture was stirred for 6 hrs at 160° C. under microwave irradiation. After cooling, the mixture was diluted with water (40 mL) and extracted with EtOAc (30 mL×2). The combined organic layers were concentrated to dryness and the residue was purified by chromatography on silica gel (elute: PE:EtOAc=3:1) to afford the title product as a white solid (400 mg, 92% yield).1H NMR (400 MHz, CDCl3) δ 8.48 (s, 1H), 8.04 (d, J=8.4 Hz, 2H), 7.46 (d, J=4.0 Hz, 1H), 7.27 (d, J=8.4 Hz, 2H), 6.36 (d, J=4.0 Hz, 1H), 5.34 (s, 1H), 2.43 (s, 6H), 2.37 (s, 3H), 1.44 (s, 9H). Step 3. Tert-butyl 3-(methyl(7-tosyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)bicyclo[1.1.1]pentane-1-carboxylate (49d) To a solution of 49c (430 mg, 0.95 mmol) in dry THE (6 mL) was add LiHMDS (2.8 mL, 2.8 mmol, 1M in THF) at −50° C. After stirring for 30 minutes at 0° C., CH3I (268 mg, 1.89 mmol) was added to the above solution. The mixture was stirred for 1.5 hrs at 40° C. After cooling, the reaction mixture was quenched with sat. NH4Cl (20 mL) and water (20 mL). The mixture was extracted with EtOAc (30 mL×2). The combined organic layers were concentrated to dryness and the residue was purified by chromatography on silica gel (elute: PE:EtOAc=1:1) to afford the title product as a white solid (125 mg, 28% yield).1H NMR (400 MHz, CDCl3) δ 8.39 (s, 1H), 8.04 (d, J=8.0 Hz, 2H), 7.45 (d, J=4.0 Hz, 1H), 7.27 (d, J=8.4 Hz, 2H), 6.63 (d, J=4.0 Hz, 1H), 3.25 (s, 3H), 2.47. (s, 6H), 2.37 (s, 3H), 1.46 (s, 9H). Step 4.3-(Methyl(7-tosyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)bicyclo[1.1.1]pentane-1-carboxylic acid (49e) Compound 49e (154 mg) was synthesized in 100% yield by utilizing similar preparative procedure of the sixth step of compound 1 with 49d (175 mg, 0.37 mmol) as starting materials. LC-MS (Method 2): tR=1.62 min, m/z (M+H)+=412.9 Step 5. Tert-butyl (3-(methyl(7-tosyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)bicyclo[1.1.1]pentan-1-yl)carbamate (49f) Compound 49f (120 mg) was synthesized in 66% yield by utilizing similar preparative procedure of the seventh step of compound 1 with 49e (154 mg, 0.37 mmol) as starting materials. LC-MS (Method 2): tR=1.85 min, m/z (M+H)+=484.2. Step 6. Tert-butyl (3-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)bicyclo[1.1.1]pentan-1-yl)carbamate (49g) Compound 49g (40 mg) was synthesized in 49% yield by utilizing similar preparative procedure of the eighth step of compound 1 with 49f (120 mg, 0.25 mmol) as starting materials. LC-MS (Method 2): tR=1.50 min, m/z (M+H)+=330.2. Step 7. N-Methyl-N-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)bicyclo[1.1.1]pentane-1,3-diamine 2,2,2-trifluoroacetate (49h) Compound 49h (55 mg crude) was synthesized in 100% yield by utilizing similar preparative procedure of the ninth step of compound 1 with 49g (40 mg, 0.12 mmol) as starting materials. LC-MS (Method 2): tR=0.22 min, m/z (M+H)+=230.0. Step 8. N-(3-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)bicyclo[1.1.1]pentan-1-yl)propane-1-sulfonamide (49) Compound 49 (1.8 mg) was synthesized in 4% yield by utilizing similar preparative procedure of the final step of compound 1 with 49h (55 mg crude, 0.12 mmol) and propane-1-sulfonyl chloride (21 mg, 0.15 mmol) as starting materials. LC-MS (Method 1): tR=2.72 min, m/z (M+H)+=336.1.1H NMR (400 MHz, CD3OD) δ 8.15 (s, 1H), 7.07 (d, J=3.6 Hz, 1H), 6.62 (d, J=3.6 Hz, 1H), 3.40 (s, 3H), 3.06-3.02 (m, 2H), 2.53 (s, 6H), 1.86-1.79 (m, 2H), 1.09 (t, J=7.6 Hz, 3H). Example 50 Step 1. Benzyl (cis-3-(methyl(7-tosyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)cyclobutyl)carbamate (50a) 49b (420 mg, 1.36 mmol), benzyl ((cis)-3-(methylamino)cyclobutyl)carbamate hydrochloride (350 mg, 1.50 mmol) and DIPEA (614 mg, 4.76 mmol) were dissolved in i-PrOH (7 mL). The mixture was stirred at 75° C. for 7 hrs. Then the mixture was filtered. The filter cake was washed with i-PrOH and dried to afford the title product as a white solid (580 mg, 88% yield). LC-MS (Method 2): tR=1.79 min, m/z (M+H)+=506.2. Step 2. Cis-N1-methyl-N1-(7-tosyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)cyclobutane-1,3-diamine hydrobromide (50b) Compound 50a (250 mg, 0.49 mmol) were dissolved in HBr (7 mL, 33% in CH3COOH) and CH3COOH (2 mL). The solution was stirred at 90° C. for 0.5 hr. The mixture was concentrated to dryness to afford the crude title product as a brown solid (170 mg crude, 65% yield). LC-MS (Method 2): tR=1.34 min, m/z (M+H)+=372.1. Step 3. N-(cis-3-(methyl(7-tosyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)cyclobutyl)propane-1-sulfonamide (50c) To a mixture of 50b (170 mg crude, 0.46 mmol) and DIPEA (614 mg, 4.76 mmol) in DCM (8 mL) was added 3-cyanoazetidine-1-sulfonyl chloride (163 mg, 1.14 mmol) at 0° C. After stirring for 3 hrs at RT, the mixture was diluted with water (70 mL) and extracted with DCM (50 mL). The separated organic layer was concentrated to dryness to afford the crude title product as a brown solid (218 mg, 100% yield). LC-MS (Method 2): tR=1.58 min, m/z (M+H)+=478.1. Step 4. N-(cis-3-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)cyclobutyl)propane-1-sulfonamide (50) 50c (215 mg, 0.45 mmol) was dissolved in i-PrOH and H2O (5.8 mL, V:V=25:4) followed by the addition of LiOH H2O (95 mg, 2.25 mmol) in one portion. The mixture was stirred at 60° C. for 13 hrs, diluted with water (30 mL) and then extracted with EtOAc (50 mL). The separated organic layer was concentrated to dryness. The residue was purified by prep-HPLC (Method A) to afford the title product as a white solid (50.0 mg, 35% yield). LC-MS (Method 1): tR=2.80 min, m/z (M+H)+=324.1;1H NMR (400 MHz, DMSO-d6) δ 11.62 (s, 1H), 8.10 (s, 1H), 7.48 (d, J=9.2 Hz, 1H), 7.15-7.14 (m, 1H), 6.63 (d, J=1.2 Hz, 1H), 4.92-4.88 (m, 1H), 3.60-3.54 (m, 1H), 3.25 (s, 3H), 2.94 (t, J=7.6 Hz, 2H), 2.62-2.60 (m, 2H), 2.26-2.19 (m, 2H), 1.73-1.64 (m, 2H), 0.98 (t, J=7.6 Hz, 3H). Biochemical Assay JAK activity was determined in the reaction buffer 50 mM HEPES, 0.01% Brij35, 10 mM MgCl2, 2 mM DTT by a microfluidic assay. The phosphorylation of a FAM labeled peptide substrate was monitored in the Caliper EZ Reader II (Perkin Elmer). The assay condition for each batch of enzyme (Carna Biosciences) was optimized to obtain 10% conversion rate of peptide substrate. The test compounds were dissolved in DMSO to a stock concentration of 10 mM. Three-fold serially diluted compounds with top concentration of 5 μM were pre-incubated with JAK1, JAK2 or TYK2 for 10 min at ambient temperature. The final DMSO concentration of assay mixture was 1%. FAM labeled peptide substrate (final concentration 3 μM) and ATP (Km concentration or 1 mM) were sequentially added to initiate the kinase reaction at 28° C. The reaction was stopped by adding 50 mM EDTA. The well in the test plate without enzyme was defined as 100% inhibition. And the well without compound but with equivalent DMSO was defined as no inhibition. The percent inhibition was calculated by the following formula. % Inhibition=(Conversionmax−Conversionsample)/(Conversionmax−Conversionmmn)*100 Conversion max means the conversion rate in the positive well without addition of compound Conversion mill means the conversion rate in the well without addition of enzymeConversion sample means the conversion rate of test compounds The dose-response (percent inhibition) curve was plotted and IC50 values were determined by GraphPad software. The IC50 values of tested compounds were list in Table 2. TABLE 2JAK1JAK2TYK2(5 nM)(0.25 nM)(2.5 nM)Ex-(1 mM(1 mM(1 mMampleNameStructureATP)ATP)ATP)1N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)propane-1- sulfonamide5.95141.3119.02N-(Cis-3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)cyclobutyl) propane-1- sulfonamide69.221086.52849.03N-(trans-3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)cyclobutyl) propane-1- sulfonamide190.11538.03658.043-cyano-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)azetidine-1- sulfonamide3.2083.2111.05N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-2- methylpropane-1- sulfonamide5.53145.2111.06N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-2- methoxyethane-1- sulfonamide24.4597.11054.07N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)cyclopropane- sulfonamide45.3469.7574.08N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-3- methylbutanamide47.4328.3—9N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)butyramide58.7440.6—10isobutyl (3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)carbamate164.5867.8—112-cyano-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)acetamide11.7165.7—12N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)cyclopropane- carboxamide35.5——132-cyclopropyl-N- (3-(imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)acetamide14.8121.9400.5143-cyano-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)propanamide8.0673.1—154-chloro-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)benzamide82.6——16isopropyl (3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)carbamate64.0——173,3-difluoro-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)cyclobutane-1- carboxamide8.489.5—184,4,4-trifluoro-N- (3-(imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)butanamide18.7167.2—19cyclopropylmethyl (3-(imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)carbamate51.3577.7—203-cyano-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)pyrrolidine-1- sulfonamide8.3334.4200.321N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-3- methoxyazetidine- 1-sulfonamide14.2326.0—223-fluoro-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)azetidine-1- sulfonamide6.145.528.4233,3-difluoro-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)azetidine-1- sulfonamide3.450.833.524N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-N′- dimethyl-1- sulfonamide23.0——25N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-N′- methyl-N′-ethyl- 1-sulfonamide26.1——26N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-3- methoxypropane- 1-sulfonamide26.2849.4—27N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)ethane- sulfonamide20.2291.3—284-chloro-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)benzene- sulfonamide6.7258.8412.829N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-1- methyl-1H- pyrazole-4- sulfonamide90.5992.2—30N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)propane-2- sulfonamide25.5226.0—31N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)butane-1- sulfonamide4.8129.4169.7323,3,3-trifluoro-N- (3-(imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)propane-1- sulfonamide6.7155.0151.3331-cyano-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)methane- sulfonamide14.4146.4—341-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-3- propylurea25.4198.9—351-cyclopropyl-3- (3-(imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)urea234.8——361-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-3- isobutylurea49.5——373,3-difluoro-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)azetidine-1- carboxamide100.7677.7—38N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-3- methoxyazetidine- 1-carboxamide388.83305.0—393-cyano-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)pyrrolidine-1- carboxamide68.7525.8—401-(2-cyano-2- methylpropyl)-3- (3-(imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)urea49.4354.9—411-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)-3- (2,2,2- trifluoroethyl) urea8.594.6—422-cyano-2- methylpropyl (3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)carbamate141.7964.6—431-(2-cyanoethyl)- 3-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-yl)urea15.6223.3—441-((cis)-3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)cyclobutyl)-N- methylmethane- sulfonamide159.2604.5—451-((((cis)-3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)cyclobutyl) methyl)sulfonyl) azetidine-3- carbonitrile15.7170.0—461-(3,3- difluorocyclo- butyl)-N-(3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)methane- sulfonamide2.278.3108.047(R)-N-(3-(2-(1- hydroxyethyl) imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)propane-1- sulfonamide48.71108.0—48N-(3-(2- methylimidazo [4,5-d]pyrrolo [2,3-b]pyridin- 1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)propane-1- sulfonamide13.0244.1—49N-(3-(Methyl(7H- pyrrolo[2,3- d]pyrimidin-4- yl)amino)bicyclo [1.1.1]pentan-1- yl)propane-1- sulfonamide>5000>5000—50N-(cis-3- (methyl(7H- pyrrolo[2,3- d]pyrimidin-4- yl)amino)cyclo- butyl)propane-1- sulfonamide44.1708.91102.51j3-(imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1-amine277.9——1itert-butyl (3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)bicyclo[1.1.1] pentan-1- yl)carbamate43.0472.02884.02e(cis)-3- (imidazo[4,5- d]pyrrolo[2,3- b]pyridin-1(6H)- yl)cyclobutan-1- amine2570.0>5000— Anti-proliferative assay Dimerization domain of Tel protein fused with JAK kinase domain was permanently transduced into BaF3 cells, whose proliferation is dependent on JAK activity in the absence of IL-3 induction. These engineered BaF3-Tel-JAK cells were used to monitor JAK inhibitory activities of the compounds in the cellular. BaF3-Tel-JAK cells were cultured in RPMI-1640 (Corning) containing 10% fetal bovine serum. Cells were seeded at 2000/well of white flat bottom 96-well plates. The well containing medium only was used as background control. After 24h growth, cells were treated with compounds. The test compounds were dissolved in DMSO to a stock concentration of 10 mM. 3-fold serially diluted compounds for 9 concentrations with top concentration of 10 μM was added into the each well. The final DMSO concentration was 0.2%. The cells continued to grow at 37° C. in 5% CO2for 72 h after compound treatment. The viability was measured by cellular ATP determination using the Cell-Titer Glo luciferase reagent (Promega). The Luminescence value was recorded by a multi-label reader Envision (PerkinElmer). Values were transformed to percent inhibition using the following formula. % Inhibition=(Readoutmax−Readoutsample)/(Readoutmax−Readoutmin)*100 The well without compound but with equivalent DMSO was defined as Readout max The well with only medium and equivalent DMSO was defined as Readout min The dose-response (percent inhibition) curve was plotted and GI50 values (the concentration that causes 50% growth inhibition) were determined by GraphPad software. The GI50 of tested compounds are shown in Table 3. TABLE 3BaF3-TEL-BaF3-TEL-ExampleJAK1 (nM)JAK2 (nM)119.6354235223003109025365103801350.1107.51748.286.718120.3291.12254.0163.22341.6148.42851285.63127.8198.23233.6121.74199.6999.64614.2132.7 Human Liver Microsome Stability Study: Commercially available human liver microsome (vendor: Coming) were used for study the Phase I stability of test articles. Microsomes were pre-incubated with test compound or control compounds for 10 min at 37° C. in 100 mM potassium phosphate buffer, pH 7.4, 3.3 mM MgCl2. The reaction was initiated by addition of 80 μL of the NADPH regenerating system to 320 μL of each incubation mixture per time point. The final incubation condition was composed of 0.5 mg/mL microsomal protein, 1 μM test article/positive control, 1.3 mM NADP, 3.3 mM glucose-6-phosphate, and 0.6 U/mL glucose-6-phosphate dehydrogenase. The 0-minute samples were prepared by addition of an 80 μL aliquot of each incubation mixture to 400 μL quench reagent to precipitate proteins. And then a 20 μL aliquot of the NADPH regenerating system was added. At 10, 30, and 90 minutes, the reaction will be stopped by the addition of cold acetonitrile solution containing tolbutamide and propanolol served as internal standard. The samples taken at all time points were centrifuged at 4000×g for 15 minutes. 80 μL of supernatant are taken into 96-well assay plates pre-added with 160 μL ultrapure water, and then analyzed by LC/MS/MS (Shimadzu LC30AD & API4000/API5000)). Concentrations of test articles, control compounds in the samples were determined by using LC/MS/MS) method. Plotting of the chromatograms and peak area integrations are carried out by Analyst (AB Sciex). In the determination of the in vitro elimination constant, ke, of the control compounds, the analyte/internal standard peak area ratios will be converted to percentage remaining (% Remaining) with the following equation: %⁢Remaining=Peak⁢area⁢ratio⁢of⁢analyte⁢to⁢⁢IS⁢at⁢each⁢time⁢pointPeak⁢area⁢ratio⁢of⁢analyte⁢to⁢IS⁢at⁢t=0×1⁢0⁢0⁢% The CLint of microsomes was calculated using the formula: CLint (mic)=0.693/T1/2/mg microsome protein per mL. Exemplary results are summarized in Table 4. TABLE 4HLM T½HLM ClintExample(min)(uL/min/mg)1341.304.104655.422.11131377.091.01141788.640.77171339.121.04221114.821.2432509.782.725063.7021.76 Rat Pharmacokinetic Study: Pharmacokinetic profile of test articles were evaluated in fasted Sprague-Dawley rats. Typically, Rats were dosed with 1 mg/kg and 2 mg/kg by intravenous injection and oral gavage, respectively. After dosing, blood samples were collected at each time point. For IV injection group, time points were set at 5, 15, 30 min, and then 1, 2, 4, 8 and 24 hours after dosing. For oral gavage group, time points were set at 15, 30 min, and then 1, 2, 4, 8, and 24 hours. Blood was collected into appropriately labeled tubes containing K2EDTA as the anticoagulant. Plasma was obtained within 1 hours of blood collection by centrifugation at 8000×g and 4° C. for 6 minutes, and then stored at −20° C. until analyzed by LC/MS/MS for quantification. PK parameter values, including, but not necessarily limited to, the maximum plasma concentrations (Cmax), the time to reach the maximum concentrations (Tmax), and the area under the plasma concentration vs. time curve (AUC) from time zero to 24-hour (AUCO-24h) were determined using WinNonlin program. Exemplary results are summarized in Table 5. TABLE 5Administration RouteRat iv @1 mpk0.2 mg/mL in 5% DMSO + 15% Solutol HS 15 + 80% SalineAUCt½ClVdFormulation(h*ng/ml)(h)(mL/min/Kg)(L/Kg)Example 1458.100.4636.201.42Administration RouteRat iv @1 mpk0.2 mg/mL in 4% DMSO + 15% Solutol HS 15 + 81% SalineAUCt½ClVdFormulation(h*ng/ml)(h)(mL/min/Kg)(L/Kg)Example 4401.700.2641.900.93Administration RouteRat po @2 mpk0.2 mg/mL in 5% DMSO + 15% Solutol HS 15 + 80% SalineCmaxt½AUCFFormulation(ng/mL)(h)(h*ng/ml)(%)Example 192.82.06190.1021.90% Conclusion: Examples 1 and 4 have good Pharmacokinetic profile in rats. Applicant's disclosure is described herein in preferred embodiments with reference to the Figures, in which like numbers represent the same or similar elements. Reference throughout this specification to “one embodiment,” “an embodiment,” or similar language means that a particular feature, structure, or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention. Thus, appearances of the phrases “in one embodiment,” “in an embodiment,” and similar language throughout this specification may, but do not necessarily, all refer to the same embodiment. The described features, structures, or characteristics of Applicant's disclosure may be combined in any suitable manner in one or more embodiments. In the description, herein, numerous specific details are recited to provide a thorough understanding of embodiments of the invention. One skilled in the relevant art will recognize, however, that Applicant's composition and/or method may be practiced without one or more of the specific details, or with other methods, components, materials, and so forth. In other instances, well-known structures, materials, or operations are not shown or described in detail to avoid obscuring aspects of the disclosure. In this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural reference, unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure, the preferred methods and materials are now described. Methods recited herein may be carried out in any order that is logically possible, in addition to a particular order disclosed. INCORPORATION BY REFERENCE References and citations to other documents, such as patents, patent applications, patent publications, journals, books, papers, web contents, have been made in this disclosure. All such documents are hereby incorporated herein by reference in their entirety for all purposes. Any material, or portion thereof, that is said to be incorporated by reference herein, but which conflicts with existing definitions, statements, or other disclosure material explicitly set forth herein is only incorporated to the extent that no conflict arises between that incorporated material and the present disclosure material. In the event of a conflict, the conflict is to be resolved in favor of the present disclosure as the preferred disclosure. EQUIVALENTS The representative examples are intended to help illustrate the invention, and are not intended to, nor should they be construed to, limit the scope of the invention. Indeed, various modifications of the invention and many further embodiments thereof, in addition to those shown and described herein, will become apparent to those skilled in the art from the full contents of this document, including the examples and the references to the scientific and patent literature included herein. The examples contain important additional information, exemplification and guidance that can be adapted to the practice of this invention in its various embodiments and equivalents thereof.
136,326
11858934
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS The following definitions are provided for the purpose of understanding the present subject matter and for construing the appended patent claims. Definitions Throughout the application, where compositions are described as having, including, or comprising specific components, or where processes are described as having, including, or comprising specific process steps, it is contemplated that compositions of the present teachings can also consist essentially of, or consist of, the recited components, and that the processes of the present teachings can also consist essentially of, or consist of, the recited process steps. It is noted that, as used in this specification and the appended claims, the singular forms “a”, “an”, and “the” include plural references unless the context clearly dictates otherwise. In the application, where an element or component is said to be included in and/or selected from a list of recited elements or components, it should be understood that the element or component can be any one of the recited elements or components, or the element or component can be selected from a group consisting of two or more of the recited elements or components. Further, it should be understood that elements and/or features of a composition or a method described herein can be combined in a variety of ways without departing from the spirit and scope of the present teachings, whether explicit or implicit herein. The use of the terms “include,” “includes”, “including,” “have,” “has,” or “having” should be generally understood as open-ended and non-limiting unless specifically stated otherwise. The use of the singular herein includes the plural (and vice versa) unless specifically stated otherwise. In addition, where the use of the term “about” is before a quantitative value, the present teachings also include the specific quantitative value itself, unless specifically stated otherwise. As used herein, the term “about” refers to a ±10% variation from the nominal value unless otherwise indicated or inferred. As used herein, “halo” or “halogen” refers to fluoro, chloro, bromo, and iodo. As used herein, “alkyl” refers to a straight-chain or branched saturated hydrocarbon group. Examples of alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and z′-propyl), butyl (e.g., n-butyl, z′-butyl, sec-butyl, tert-butyl), pentyl groups (e.g., n-pentyl, z′-pentyl, -pentyl), hexyl groups, and the like. In various embodiments, an alkyl group can have 1 to 40 carbon atoms (i.e., C1-C40alkyl group), for example, 1-30 carbon atoms (i.e., C1-C30alkyl group). In some embodiments, an alkyl group can have 1 to 6 carbon atoms, and can be referred to as a “lower alkyl group” or a “C1-C6alkyl group”. Examples of lower alkyl groups include methyl, ethyl, propyl (e.g., n-propyl and z′-propyl), and butyl groups (e.g., n-butyl, z′-butyl, sec-butyl, tert-butyl). In some embodiments, alkyl groups can be substituted as described herein. An alkyl group is generally not substituted with another alkyl group, an alkenyl group, or an alkynyl group. As used herein, “alkenyl” refers to a straight-chain or branched alkyl group having one or more carbon-carbon double bonds. Examples of alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl groups, and the like. The one or more carbon-carbon double bonds can be internal (such as in 2-butene) or terminal (such as in 1-butene). In various embodiments, an alkenyl group can have 2 to 40 carbon atoms (i.e., C2-C40alkenyl group), for example, 2 to 20 carbon atoms (i.e., C2-C20alkenyl group) or 2 to 6 carbon atoms (i.e., C2-C6alkenyl group). In some embodiments, alkenyl groups can be substituted as described herein. An alkenyl group is generally not substituted with another alkenyl group, an alkyl group, or an alkynyl group. The term “substituted alkyl” as used herein refers to an alkyl group in which 1 or more (up to about 5, for example about 3) hydrogen atoms is replaced by a substituent independently selected from the group: —O, —S, acyl, acyloxy, optionally substituted alkoxy, optionally substituted amino (wherein the amino group may be a cyclic amine), azido, carboxyl, (optionally substituted alkoxy)carbonyl, amido, cyano, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, halogen, hydroxyl, nitro, sulfamoyl, sulfanyl, sulfinyl, sulfonyl, and sulfonic acid. Some of the optional substituents for alkyl are hydroxy, halogen exemplified by chloro and bromo, acyl exemplified by methylcarbonyl; alkoxy, and heterocyclyl exemplified by morpholino and piperidino. Other alkyl substituents as described herein may further be contemplated. The term “substituted alkenyl” refers to an alkenyl group in which 1 or more (up to about 5, for example about 3) hydrogen atoms is replaced by a substituent independently selected from those listed above with respect to a substituted alkyl. Other alkenyl substituents as described herein may further be contemplated. As used herein, “heteroatom” refers to an atom of any element other than carbon or hydrogen and includes, for example, nitrogen, oxygen, silicon, sulfur, phosphorus, and selenium. As used herein, “aryl” refers to an aromatic monocyclic hydrocarbon ring system or a polycyclic ring system in which two or more aromatic hydrocarbon rings are fused (i.e., having a bond in common with) together or at least one aromatic monocyclic hydrocarbon ring is fused to one or more cycloalkyl and/or cycloheteroalkyl rings. An aryl group can have 6 to 24 carbon atoms in its ring system (e.g., C6-C24aryl group), which can include multiple fused rings. In some embodiments, a polycyclic aryl group can have 8 to 24 carbon atoms. Any suitable ring position of the aryl group can be covalently linked to the defined chemical structure. Examples of aryl groups having only aromatic carbocyclic ring(s) include phenyl, 1-naphthyl (bicyclic), 2-naphthyl (bicyclic), anthracenyl (tricyclic), phenanthrenyl (tricyclic), pentacenyl (pentacyclic), and like groups. Examples of polycyclic ring systems in which at least one aromatic carbocyclic ring is fused to one or more cycloalkyl and/or cycloheteroalkyl rings include, among others, benzo derivatives of cyclopentane (i.e., an indanyl group, which is a 5,6-bicyclic cycloalkyl/aromatic ring system), cyclohexane (i.e., a tetrahydronaphthyl group, which is a 6,6-bicyclic cycloalkyl/aromatic ring system), imidazoline (i.e., a benzimidazolinyl group, which is a 5,6-bicyclic cycloheteroalkyl/aromatic ring system), and pyran (i.e., a chromenyl group, which is a 6,6-bicyclic cycloheteroalkyl/aromatic ring system). Other examples of aryl groups include benzodioxanyl, benzodioxolyl, chromanyl, indolinyl groups, and the like. In some embodiments, aryl groups can be substituted as described herein. In some embodiments, an aryl group can have one or more halogen substituents, and can be referred to as a “haloaryl” group. Perhaloaryl groups, i.e., aryl groups where all of the hydrogen atoms are replaced with halogen atoms (e.g., —C6F5), are included within the definition of “haloaryl”. In certain embodiments, an aryl group is substituted with another aryl group and can be referred to as a biaryl group. Each of the aryl groups in the biaryl group can be substituted as disclosed herein. As used herein, “heteroaryl” refers to an aromatic monocyclic ring system containing at least one ring heteroatom selected from oxygen (O), nitrogen (N), sulfur (S), silicon (Si), and selenium (Se) or a polycyclic ring system where at least one of the rings present in the ring system is aromatic and contains at least one ring heteroatom. Polycyclic heteroaryl groups include those having two or more heteroaryl rings fused together, as well as those having at least one monocyclic heteroaryl ring fused to one or more aromatic carbocyclic rings, non-aromatic carbocyclic rings, and/or non-aromatic cycloheteroalkyl rings. A heteroaryl group, as a whole, can have, for example, 5 to 24 ring atoms and contain 1-5 ring heteroatoms (i.e., 5-20 membered heteroaryl group). The heteroaryl group can be attached to the defined chemical structure at any heteroatom or carbon atom that results in a stable structure. Generally, heteroaryl rings do not contain O—O, S—S, or S-0 bonds. However, one or more N or S atoms in a heteroaryl group can be oxidized (e.g., pyridine N-oxide thiophene S-oxide, thiophene S,S-dioxide). Examples of heteroaryl groups include, for example, the 5- or 6-membered monocyclic and 5-6 bicyclic ring systems shown below: where T is O, S, NH, N-alkyl, N-aryl, N-(arylalkyl) (e.g., N-benzyl), SiH2, SiH(alkyl), Si(alkyl)2, SiH(arylalkyl), Si(arylalkyl)2, or Si(alkyl)(arylalkyl). Examples of such heteroaryl rings include pyrrolyl, furyl, thienyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazolyl, tetrazolyl, pyrazolyl, imidazolyl, isothiazolyl, thiazolyl, thiadiazolyl, isoxazolyl, oxazolyl, oxadiazolyl, indolyl, isoindolyl, benzofuryl, benzothienyl, quinolyl, 2-methylquinolyl, isoquinolyl, quinoxalyl, quinazolyl, benzotriazolyl, benzimidazolyl, benzothiazolyl, benzisothiazolyl, benzisoxazolyl, benzoxadiazolyl, benzoxazolyl, cinnolinyl, 1H-indazolyl, 2H-indazolyl, indolizinyl, isobenzofuyl, naphthyridinyl, phthalazinyl, pteridinyl, purinyl, oxazolopyridinyl, thiazolopyridinyl, imidazopyridinyl, furopyridinyl, thienopyridinyl, pyridopyrimidinyl, pyridopyrazinyl, pyridopyridazinyl, thienothiazolyl, thienoxazolyl, thienoimidazolyl groups, and the like. Further examples of heteroaryl groups include 4,5,6,7-tetrahydroindolyl, tetrahydroquinolinyl, benzothienopyridinyl, benzofuropyridinyl groups, and the like. In some embodiments, heteroaryl groups can be substituted as described herein. The term “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, “optionally substituted alkyl” means either “alkyl” or “substituted alkyl,” as defined herein. It will be understood by those skilled in the art with respect to any chemical group containing one or more substituents that such groups are not intended to introduce any substitution or substitution patterns that are sterically impractical and/or physically non-feasible. The term “isomers” or “stereoisomers” as used herein relates to compounds that have identical molecular formulae but that differ in the arrangement of their atoms in space. Stereoisomers that are not mirror images of one another are termed “diastereoisomers” and stereoisomers that are non-superimposable mirror images are termed “enantiomers,” or sometimes optical isomers. A carbon atom bonded to four non-identical substituents is termed a “chiral center.” Certain compounds herein have one or more chiral centers and therefore may exist as either individual stereoisomers or as a mixture of stereoisomers. Configurations of stereoisomers that owe their existence to hindered rotation about double bonds are differentiated by their prefixes cis and trans (or Z and E), which indicate that the groups are on the same side (cis or Z) or on opposite sides (trans or E) of the double bond in the molecule according to the Cahn-Ingold-Prelog rules. All possible stereoisomers are contemplated herein as individual stereoisomers or as a mixture of stereoisomers. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which the presently described subject matter pertains. Where a range of values is provided, for example, concentration ranges, percentage ranges, or ratio ranges, it is understood that each intervening value, to the tenth of the unit of the lower limit, unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the described subject matter. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and such embodiments are also encompassed within the described subject matter, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the described subject matter. Throughout the application, descriptions of various embodiments use “comprising” language. However, it will be understood by one of skill in the art, that in some specific instances, an embodiment can alternatively be described using the language “consisting essentially of” or “consisting of”. “Subject” as used herein refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, and pet companion animals such as household pets and other domesticated animals such as, but not limited to, cattle, sheep, ferrets, swine, horses, poultry, rabbits, goats, dogs, cats and the like. “Patient” as used herein refers to a subject in need of treatment of a condition, disorder, or disease, such as an acute or chronic airway disorder or disease. For purposes of better understanding the present teachings and in no way limiting the scope of the teachings, unless otherwise indicated, all numbers expressing quantities, percentages or proportions, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term “about”. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. In an embodiment, the present subject matter relates to a compound having the formula I: or a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof, wherein: R1represents an aryl group, a heteroaryl group, an aminoaryl group, an aminoheteroaryl group, an aminoalkyl group, an aminoalkylaryl group, an aminoalkylheteroaryl group, an aminocycloalkyl group, an aryloxy group, a heteroaryloxy group, or an alkoxy group, wherein each R1group may be optionally substituted with from one to three substituents independently selected from the group consisting of a linear or branched C1-C6alkyl group, a halogen atom, a linear or branched C1-C6alkoxy group, a linear or branched C1-C6trihaloalkyl group, a hydroxy group, an amide group, and a group of formula —NR4R5, wherein R4and R5, which may be the same or different, each independently represent a hydrogen atom or a linear or branched C1-C6alkyl group optionally substituted by a hydroxy group, or R4and R5, together with the nitrogen atom to which they are attached, form a nitrogen-containing heterocycle, such nitrogen-containing heterocycle being optionally substituted with from one to three substituents independently selected from the group consisting of a linear or branched C1-C6alkyl group, a halogen atom, a linear or branched C1-C6alkoxy group, a linear or branched C1-C6trihaloalkyl group, and a hydroxy group; and R2and R3taken independently can each represent hydrogen, a carboxyl group, an ester group, an amide group, a halogen atom, a linear or branched C1-C6alkyl group, an aryl group, a hydroxy group, a linear or branched C1-C6alkoxy group, an aryloxy group, an aminoalkyl group, an aminocycloalkyl group, an aminoaryl group, and an aminoheteroaryl group, wherein each of R2and R3taken independently may be optionally substituted with from one to three substituents independently selected from the group consisting of a linear or branched C1-C6alkyl group, a halogen atom, a linear or branched C1-C6alkoxy group, a linear or branched C1-C6trihaloalkyl group, a hydroxy group, and an amide group, and an amine group. In a further embodiment, the present subject matter relates to compounds of formula I, wherein R1is an optionally substituted aminophenyl group, an optionally substituted —NH(C1-C6)alkylphenyl group, an optionally substituted —NH(C1-C6alkyl) group, or an optionally substituted aminoheteroaryl group. In another embodiment, the present subject matter relates to compounds of formula I, wherein R1is aminophenyl substituted with one or more substituents independently selected from the group consisting of a straight or branched C1-C6alkyl, trihalomethyl, halogen, hydroxy, —NR4R5, and C1-C6alkoxy. In yet another embodiment, the present subject matter relates to compounds of formula I, wherein R1is aminophenyl substituted with one or more substituents independently selected from the group consisting of C1-C3alkyl, trifluoromethyl, chlorine, fluorine, hydroxy, dimethyl amine, and methoxy. In still yet another embodiment, the present subject matter relates to compounds of formula I, wherein R1is an aminoethyl group substituted with —NR4R5, wherein R4and R5are independently a linear C1-C6alkyl group, or R4and R5are taken together with the nitrogen atom to which they are attached to form an optionally substituted nitrogen-containing heterocycle. In this regard, R4and R5can both be ethyl, or R4and R5can be taken together with the nitrogen atom to which they are attached to form a 4-methylpiperazin-1-yl group. In one embodiment, the present subject matter relates to a compound of formula I, wherein R1is an aminomethylphenyl group substituted with a chlorine. In another embodiment, the present subject matter relates to a compound of formula I, wherein R1is a 2-methylpyridin-4-ylamine group. In a further embodiment, the present subject matter relates to a compound of formula I, wherein R2is —COOH and R3is hydrogen. In another embodiment, the present subject matter relates to a compound having the formula I: or a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof, wherein R1is aminophenyl substituted with one or more substituents independently selected from the group consisting of a straight or branched C1-C6alkyl, trihalomethyl, halogen, hydroxy, —NR4R5, and C1-C6alkoxy; an aminoethyl group substituted with —NR4R5, wherein R4and R5are independently a linear C1-C6alkyl group, or R4and R5are taken together with the nitrogen atom to which they are attached to form an optionally substituted nitrogen-containing heterocycle; an aminomethylphenyl group substituted with a chlorine; or a 2-methylpyridin-4-ylamine group; R2is —COOH; and R3is hydrogen. In an embodiment, the present subject matter relates to a compound selected from the group consisting of: 5-((3-chlorophenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (1), 5-((3-hydroxyphenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (2), 5-((3-methoxyphenyl)amino)pyrido[4′,3′:5 ,6]pyrimido[1,2-a]indole-9-carboxylic acid (3), 5-((3-(dimethylamino)phenyl)amino)pyrido[4′,3′:5 ,6]pyrimido[1,2-a]indole-9-carboxylic acid (4), 5 4(3-(trifluoromethyl)phenyl)amino)pyrido[4′,3′:5 ,6]pyrimido[1,2-a]indole-9-carboxylic acid (5), 5((4-chlorophenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (6), 5-((4-methoxyphenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (7), 5-((3,5-dimethylphenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (8), 5-((2-methylpyridin-4-yl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (9), 5-((3-chlorobenzyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (10), 5-((2-(diethylamino)ethyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (11), 5-((2-(4-methylpiperazin-1-yl)ethyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (12), and a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof. Said differently, the present subject matter can relate to compounds of formula I selected from the group consisting of: and a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof. It is to be understood that the present subject matter covers all combinations of substituent groups referred to herein. The present compounds may contain, e.g., when isolated in crystalline form, varying amounts of solvents. Accordingly, the present subject matter includes all solvates of the present compounds of formula I and pharmaceutically acceptable stereoisomers, esters, and/or salts thereof. Hydrates are one example of such solvates. Further, the present subject matter includes all mixtures of possible stereoisomers of the embodied compounds, independent of the ratio, including the racemates. Salts of the present compounds, or salts of the stereoisomers thereof, include all inorganic and organic acid addition salts and salts with bases, especially all pharmaceutically acceptable inorganic and organic acid addition salts and salts with bases, particularly all pharmaceutically acceptable inorganic and organic acid addition salts and salts with bases customarily used in pharmacy. Examples of acid addition salts include, but are not limited to, hydrochlorides, hydrobromides, phosphates, nitrates, sulfates, acetates, trifluoroacetates, citrates, D-gluconates, benzoates, 2-(4-hydroxy-benzoyl)benzoates, butyrates, subsalicylates, maleates, laurates, malates, lactates, fumarates, succinates, oxalates, tartrates, stearates, benzenesulfonates (besilates), toluenesulfonates (tosilates), methanesulfonates (mesilates) and 3-hydroxy-2-naphthoates. Examples of salts with bases include, but are not limited to, lithium, sodium, potassium, calcium, aluminum, magnesium, titanium, ammonium, meglumine and guanidinium salts. The salts include water-insoluble and, particularly, water-soluble salts. The present compounds, the salts, the stereoisomers and the salts of the stereoisomers thereof may contain, e.g., when isolated in crystalline form, varying amounts of solvents. Included within the present scope are, therefore, all solvates of the compounds of formula I, as well as the solvates of the salts, the stereoisomers and the salts of the stereoisomers of the compounds of formula I. The present compounds may be isolated and purified in a manner known per se, e.g., by distilling off the solvent in vacuo and recrystallizing the residue obtained from a suitable solvent or subjecting it to one of the customary purification methods, such as column chromatography on a suitable support material. Salts of the compounds of formula I and the stereoisomers thereof can be obtained by dissolving the free compound in a suitable solvent (by way of non-limiting example, a ketone such as acetone, methylethylketone or methylisobutylketone; an ether such as diethyl ether, tetrahydrofurane or dioxane; a chlorinated hydrocarbon such as methylene chloride or chloroform; a low molecular weight aliphatic alcohol such as methanol, ethanol or isopropanol; a low molecular weight aliphatic ester such as ethyl acetate or isopropyl acetate; or water) which contains the desired acid or base, or to which the desired acid or base is then added. The acid or base can be employed in salt preparation, depending on whether a mono- or polybasic acid or base is concerned and depending on which salt is desired, in an equimolar quantitative ratio or one differing therefrom. The salts are obtained by filtering, reprecipitating, precipitating with a non-solvent for the salt or by evaporating the solvent. Salts obtained can be converted into the free compounds which, in turn, can be converted into salts. In this manner, pharmaceutically unacceptable salts, which can be obtained, for example, as process products in the manufacturing on an industrial scale, can be converted into pharmaceutically acceptable salts by processes known to the person skilled in the art. Pure diastereomers and pure enantiomers of the present compounds can be obtained, e.g., by asymmetric synthesis, by using chiral starting compounds in synthesis and by splitting up enantiomeric and diastereomeric mixtures obtained in synthesis. Preferably, the pure diastereomeric and pure enantiomeric compounds are obtained by using chiral starting compounds in synthesis. Enantiomeric and diastereomeric mixtures can be split up into the pure enantiomers and pure diastereomers by methods known to a person skilled in the art. Preferably, diastereomeric mixtures are separated by crystallization, in particular fractional crystallization, or chromatography. Enantiomeric mixtures can be separated, e.g., by forming diastereomers with a chiral auxiliary agent, resolving the diastereomers obtained and removing the chiral auxiliary agent. As chiral auxiliary agents, for example, chiral acids can be used to separate enantiomeric bases and chiral bases can be used to separate enantiomeric acids via formation of diastereomeric salts. Furthermore, diastereomeric derivatives such as diastereomeric esters can be formed from enantiomeric mixtures of alcohols or enantiomeric mixtures of acids, respectively, using chiral acids or chiral alcohols, respectively, as chiral auxiliary agents. Additionally, diastereomeric complexes or diastereomeric clathrates may be used for separating enantiomeric mixtures. Alternatively, enantiomeric mixtures can be split up using chiral separating columns in chromatography. Another suitable method for the isolation of enantiomers is enzymatic separation. In one embodiment, the present compounds can be prepared according to the following general synthetic pathway. Specifically, synthesis commences with heating a mixture of dimethyl 1H-indole-2,5-dicarboxylate I, 3-bromisonicotinamide II, CuI, metformin hydrochloride, Cs2CO3, and DMF. After cooling to room temperature, dilution, filtering, and concentration, the resulting residue is purified to afford methyl 1-(4-carbamoylpyridin-3-yl)-1H-indole-5-carboxylate III as shown in Scheme 1. Next a solution of methyl 1-(4-carbamoylpyridin-3-yl)-1H-indole-5-carboxylate III and N-iodosuccinimide in CH2Cl2is stirred. After cooling, the reaction mixture is washed with a solution of Na2S2O3and extracted with CH2C12. The organic layers are combined, dried, filtrated, and evaporated. The resulting crude mixture is purified to provide methyl 6-oxo-5,6-dihydropyrido[4′,3′:5,6]pyrazino[1,2-a]indole-9-carboxylate, which is then chlorinated in CH2Cl2with SOCl2. The organic layer is washed, dried, filtrated, evaporated, and crystallized to provide the corresponding methyl 5-chloropyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylate IV as outlined in Scheme 2. The final step of the synthesis involves mixing the 5-chloro derivative IV and a variety of amines. The solid obtained is filtrated and washed. A mixture of methyl 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylate derivatives and KOH is then heated, cooled, filtrated, washed, and recrystallized to obtain the corresponding 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V (examples 1-12) as shown in Scheme 3. In another embodiment, the present subject matter is directed to pharmaceutical compositions comprising a therapeutically effective amount of the compounds as described herein together with one or more pharmaceutically acceptable carriers, excipients, or vehicles. In some embodiments, the present compositions can be used for combination therapy, where other therapeutic and/or prophylactic ingredients can be included therein. The present subject matter further relates to a pharmaceutical composition, which comprises at least one of the present compounds together with at least one pharmaceutically acceptable auxiliary. In an embodiment, the pharmaceutical composition comprises one or two of the present compounds, or one of the present compounds. Non-limiting examples of suitable excipients, carriers, or vehicles useful herein include liquids such as water, saline, glycerol, polyethyleneglycol, hyaluronic acid, ethanol, and the like. Suitable excipients for nonliquid formulations are also known to those of skill in the art. A thorough discussion of pharmaceutically acceptable excipients and salts useful herein is available in Remington's Pharmaceutical Sciences, 18th Edition. Easton, Pa., Mack Publishing Company, 1990, the entire contents of which are incorporated by reference herein. The present compounds are typically administered at a therapeutically or pharmaceutically effective dosage, e.g., a dosage sufficient to provide treatment for cancer. Administration of the compounds or pharmaceutical compositions thereof can be by any method that delivers the compounds systemically and/or locally. These methods include oral routes, parenteral routes, intraduodenal routes, and the like. While human dosage levels have yet to be optimized for the present compounds, generally, a daily dose is from about 0.01 to 10.0 mg/kg of body weight, for example about 0.1 to 5.0 mg/kg of body weight. The precise effective amount will vary from subject to subject and will depend upon the species, age, the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. The subject may be administered as many doses as is required to reduce and/or alleviate the signs, symptoms, or causes of the disease or disorder in question, or bring about any other desired alteration of a biological system. In employing the present compounds for treatment of cancer, any pharmaceutically acceptable mode of administration can be used with other pharmaceutically acceptable excipients, including solid, semi-solid, liquid or aerosol dosage forms, such as, for example, tablets, capsules, powders, liquids, suspensions, suppositories, aerosols or the like. The present compounds can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, and the like, for the prolonged administration of the compound at a predetermined rate, preferably in unit dosage forms suitable for single administration of precise dosages. The present compounds may also be administered as compositions prepared as foods for foods or animals, including medical foods, functional food, special nutrition foods and dietary supplements. A “medical food” is a product prescribed by a physician that is intended for the specific dietary management of a disorder or health condition for which distinctive nutritional requirements exist and may include formulations fed through a feeding tube (referred to as enteral administration or gavage administration). A “dietary supplement” shall mean a product that is intended to supplement the human diet and may be provided in the form of a pill, capsule, tablet, or like formulation. By way of non-limiting example, a dietary supplement may include one or more of the following dietary ingredients: vitamins, minerals, herbs, botanicals, amino acids, and dietary substances intended to supplement the diet by increasing total dietary intake, or a concentrate, metabolite, constituent, extract, or combinations of these ingredients, not intended as a conventional food or as the sole item of a meal or diet. Dietary supplements may also be incorporated into foodstuffs, such as functional foods designed to promote control of glucose levels. A “functional food” is an ordinary food that has one or more components or ingredients incorporated into it to give a specific medical or physiological benefit, other than a purely nutritional effect. “Special nutrition food” means ingredients designed for a particular diet related to conditions or to support treatment of nutritional deficiencies. Generally, depending on the intended mode of administration, the pharmaceutically acceptable composition will contain about 0.1% to 90%, for example about 0.5% to 50%, by weight of a compound or salt of the present compounds, the remainder being suitable pharmaceutical excipients, carriers, etc. One manner of administration for the conditions detailed above is oral, using a convenient daily dosage regimen which can be adjusted according to the degree of affliction. For such oral administration, a pharmaceutically acceptable, non-toxic composition is formed by the incorporation of any of the normally employed excipients, such as, for example, mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, sodium crosscarmellose, glucose, gelatin, sucrose, magnesium carbonate, and the like. Such compositions take the form of solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations and the like. The present compositions may take the form of a pill or tablet and thus the composition may contain, along with the active ingredient, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose and derivatives thereof, and the like. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc. an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, for example, sodium acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine acetate, triethanolamine oleate, etc. For oral administration, a pharmaceutically acceptable non-toxic composition may be formed by the incorporation of any normally employed excipients, such as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, cellulose derivatives, sodium crosscarmellose, glucose, sucrose, magnesium carbonate, sodium saccharin, talcum and the like. Such compositions take the form of solutions, suspensions, tablets, capsules, powders, sustained release formulations and the like. For a solid dosage form, a solution or suspension in, for example, propylene carbonate, vegetable oils or triglycerides, may be encapsulated in a gelatin capsule. Such diester solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545, the contents of each of which are incorporated herein by reference. For a liquid dosage form, the solution, e.g., in a polyethylene glycol, may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be easily measured for administration. Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and the like, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in U.S. Pat. Nos. Re. 28,819 and 4,358,603, the contents of each of which are hereby incorporated by reference. Another manner of administration is parenteral administration, generally characterized by injection, either subcutaneously, intramuscularly or intravenously. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like. In addition, if desired, the pharmaceutical compositions to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, solubility enhancers, and the like, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate, cyclodextrins, etc. Another approach for parenteral administration employs the implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject. However, percentages of active ingredient of 0.01% to 10% in solution are employable and will be higher if the composition is a solid which will be subsequently diluted to the above percentages. The composition may comprise 0.2% to 2% of the active agent in solution. Nasal solutions of the active compound alone or in combination with other pharmaceutically acceptable excipients can also be administered. Formulations of the active compound or a salt may also be administered to the respiratory tract as an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation have diameters of less than 50 microns, for example less than 10 microns. The present compounds have valuable pharmaceutical properties, which make them commercially utilizable. Accordingly, the present subject matter further relates to use of the present compounds for the treatment of diseases such as cancers. Similarly, the present compounds can be used to inhibit CK2 enzyme activity in a patient. In another embodiment of the present subject matter, the aforementioned compound derivatives demonstrated in vitro anticancer action against human cancer cell lines such as MCF7 and MDA-MB-231 (breast cancer), H1299 (lung cancer), PC3 (prostate cancer), HCT116 (colon cancer), A375 (melanoma), MIAPaCa2 (pancreatic cancer), and HL60 (leukemia). Accordingly, the present subject matter relates to methods of treating a cancer in a patient by administering one or more of the compounds presented herein to a patient in need thereof. In certain embodiments, the cancer treatable with the present compounds is one or more selected from the group consisting of leukemia, melanoma, colon cancer, prostate cancer, lung cancer, pancreatic cancer, and breast cancer. Accordingly, in an embodiment of the present subject matter, the pyrido[4′,3′:5,6]pyrimido[1,2-a]indole derivatives, or the 5-substituted aminopyrido[4′,3′:5,6] pyrimido [1,2-a]indole-9-carboxylic acid derivatives, as described herein engaged for in vitro study towards human cancer cell lines can display an IC50with a nano to micromolar concentration range when exposed to a period of at least 96 hrs. For example, a present compound engaged for in vitro study against PC3 (prostate) cancer cell lines can display an IC50concentration of 2.2 μM at an exposure period of at least 96 hrs. In another embodiment, a present compound engaged for in vitro study against HCT116 (colon) cancer cell lines can display an IC50concentration of 1.8 μM at an exposure period of at least 96 hrs. In a further embodiment, a present compound engaged for in vitro study against A375 (melanoma) cancer cell lines can display an IC50concentration of 3.5 μM at an exposure period of at least 96 hrs. In an embodiment, a present compound engaged for in vitro study against H1299 (lung) cancer cell lines can display an IC50concentration of 1.9 μM at an exposure period of at least 96 hrs. In another embodiment, a present compound engaged for in vitro study against MIAPaCa-2 (pancreas) cancer cell lines can display an IC50concentration of 1.4 μM at an exposure period of at least 96 hrs. In a further embodiment, a present compound engaged for in vitro study against HL60 (leukemia) cancer cell lines can display an IC50concentration of 3.3 μM at an exposure period of at least 96 hrs. In one embodiment, a present compound engaged for in vitro study against MCF7 (breast) cancer cell lines can display an IC50concentration of 6.9 μM at an exposure period of at least 96 hrs. In another embodiment, a present compound engaged for in vitro study against MDA-MB-231 (breast) cancer cell lines can display an IC50concentration of 5.5 μM at an exposure period of at least 96 hrs. The present subject matter further relates to a method of treating or preventing a disease comprising administering to a patient in need thereof a therapeutically effective amount of at least one of the compounds herein. In particular, the present subject matter relates to a method of treating one of the above-mentioned diseases or disorders comprising administering to a patient in need thereof a therapeutically effective amount of at least one of the compounds herein. In the above methods, the patient is preferably a mammal, more preferably a human. Furthermore, in the above methods, at least one of the present compounds can be used. In an embodiment, one or two of the present compounds are used, or one of the present compounds is used. Similarly, one or more of the present compounds can be used in combination therapy with one or more additional active agents. The following examples relate to various methods of manufacturing certain specific compounds as described herein. All compound numbers expressed herein are with reference to the synthetic pathway figures shown above. EXAMPLES Example 1 Preparation of methyl 1-(4-carbamoylpyridin-3-yl)-1H-indole-5-carboxylate III A mixture of dimethyl 1H-indole-2,5-dicarboxylate I (5 mmol), 3-bromoisonicotinamide II (5 mmol), CuI (0.5 mmol) metformin hydrochloride (1 mmol), Cs2CO3(10 mmol) and DMF (10 mL) was heated to 130° C. for 8 hours under N2atmosphere. After cooling to room temperature, the reaction mixture was diluted with EtOAc (20 mL). The solid was removed by filter, and the filtrate was washed with water and brine. The organic layer was dried over MgSO4, filtered, and concentrated under reduced pressure. The resulting residue was purified by column chromatography to afford the methyl 1-(4-carbamoylpyridin-3-yl)-1H-indole-5-carboxylate III. Elemental Analysis: Calculated C, 65.08; H, 4.44; N, 14.23; Found C, 64.91; H, 3.40; N, 14.38. Example 2 Preparation of methyl 5-chloropyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylate IV A solution of methyl 1-(4-carbamoylpyridin-3-yl)-1H-indole-5-carboxylate III (5 mmol) and N-iodosuccinimide (5 mmol,) in CH2Cl2(10 mL) was stirred at 80° C. for 4 hours. After cooling at room temperature, the reaction mixture was washed with a solution of saturated Na2S2O3and extracted with CH2Cl2. The organic layers were combined, dried over MgSO4, filtrated, evaporated under reduced pressure and purified by column chromatography to afford the product methyl 6-oxo-5,6-dihydropyrido[4′,3′:5,6]pyrazino[1,2-a]indole-9-carboxylate which was engaged in the next chlorination step. To a mixture of methyl 6-oxo-5,6-dihydropyrido[4′,3′:5,6]pyrazino[1,2-a]indole-9-carboxylate (2 mmol) in CH2Cl2(10 mL) were added SOCl2(3 mmol) and a drop of DMF. The reaction mixture was stirred at room temperature for 4 hours. The organic layer was washed with a saturated ammonium chloride solution. The organic layer was dried over MgSO4, filtrated, evaporated, and crystallized to provide the corresponding methyl 5-chloropyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylate IV. Elemental Analysis: Calculated C, 61.65; H, 3.23; N, 13.48; Found C, 61.62; H, 3.68; N, 13.51. Example 3 General Procedure for the Preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V A mixture of 5-chloro derivative IV (1 mmol) and a variety of amine (2 mmol) in mL 2-ethoxyethanol was heated at reflux for 6 hours. After cooling to room temperature, the solid obtained was filtrated, washed with diethyl ether and was engaged in the next step without further purification. A mixture of methyl 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylate derivatives (1 mmol) and KOH (3 mmol) in 5 mL THF was heated at reflux for 2 hours. After cooling to room temperature, a solution of 10% HCl was added and the resulting precipitate was filtrated, washed with water and recrystallized to furnish the corresponding 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V (compounds 1-12). Example 4 Preparation of 5-((3-chlorophenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (1) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 3-chloroaniline as the amine. Elemental Analysis: Calculated C, 64.87; H, 3.37; N, 14.41; Found C, 64.83; H, 3.36; N, 14.44. Example 5 Preparation of 5-((3-hydroxyphenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (2) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 3-hydroxyaniline as the amine. Elemental Analysis: Calculated C, 68.10; H, 3.81; N, 15.13; Found C, 68.03; H, 3.79; N, 15.09. Example 6 Preparation of 5-((3-methoxyphenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (3) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 3-methoxyaniline as the amine. Elemental Analysis: Calculated C, 68.74; H, 4.20; N, 14.58; Found C, 68.69; H, 4.17; N, 14.62. Example 7 Preparation of 5-((3-(dimethylamino)phenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (4) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 3-dimethylaminolaniline as the amine. Elemental Analysis: Calculated C, 69.51; H, 4.82; N, 17.62; Found C, 69.50; H, 4.74; N, 17.55. Example 8 Preparation of 5-((3-(trifluoromethyl)phenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (5) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 3-trifluoromethylaniline as the amine. Elemental Analysis: Calculated C, 62.56; H, 3.10; N, 13.27; Found C, 62.51; H, 3.16; N, 13.33. Example 9 Preparation of 5-((4-chlorophenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (6) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 4-chloroaminoaniline as the amine. Elemental Analysis: Calculated C, 64.87; H, 3.37; N, 14.41; Found C, 64.88; H, 3.41; N, 14.44. Example 10 Preparation of 5-((4-methoxyphenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (7) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 4-methoxyaniline as the amine. Elemental Analysis: Calculated C, 68.74; H, 4.20; N, 14.58; Found C, 68.80; H, 4.22; N, 14.66. Example 11 Preparation of 5-((3,5-dimethylphenyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (8) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 3,5-dimethylaniline as the amine. Elemental Analysis: Calculated C, 72.24; H, 4.74; N, 14.65; Found C, 72.19; H, 4.70; N, 14.64. Example 12 Preparation of 54(2-methylpyridin-4-yl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (9) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 2-methylpyridin-4-amine as the amine. Elemental Analysis: Calculated C, 68.28; H, 4.09; N, 18.96; Found C, 68.30; H, 4.02; N, 19.01. Example 13 Preparation of 5-((3-chlorobenzyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (10) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 3-chlorobenzylamine as the amine. Elemental Analysis: Calculated C, 65.60; H, 3.75; N, 13.91; Found C, 65.54; H, 3.76; N, 14.00. Example 14 Preparation of 5-((2-(diethylamino)ethyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (11) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using N1,N1-dimethylethane-1,2-diamine as the amine. Elemental Analysis: Calculated C, 66.83; H, 6.14; N, 18.55; Found C, 66.76; H, 6.11; N, 18.60. Example 15 Preparation of 5-((2-(4-methylpiperazin-1-yl)ethyl)amino)pyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid (12) The expected product was obtained in accordance with the general procedure for the preparation of 5-substituted aminopyrido[4′,3′:5,6]pyrimido[1,2-a]indole-9-carboxylic acid V of Example 3 using 3-(4-methylpiperazin-1-yl)ethan-1-amine as the amine. Elemental Analysis: Calculated C, 65.33; H, 5.98; N, 20.78; Found C, 65.29; H, 6.04; N, 20.86. Pharmacological Activity Example 16 In Vitro Cytotoxic Activity Assay Compounds 1-12 were screened for their in vitro cytotoxic activity utilizing a 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay against selected cancer human cell lines consisting of PC3 (prostate), HCT116 (colon), A375 (melanoma), H1299 (lung), MIAPaCa-2 (pancreas), HL60 (leukemia), MCF7 (breast), MDA-MB-231 (breast) (T. Mosmann, J. Immunol. Meth., 1983, 65, 55-63). The cells were cultured at 37° C. in RMPI1640 medium supplemented with 10% fetal bovine serum, 50 IU/mL penicillin, and 50 μg/mL streptomycin in a 5% CO2incubator. All cells were sub-cultured 3 times/week by trypsinisation. Viable cells were seeded and allowed to adhere for 12 h before a test drug was added in 96-well plates at an initial density of 1.0×105 cells/mL. Tumour cell lines were separately exposed to various concentrations of the tested compounds followed by incubation at a temperature of 37° C. during 96 h inside a medium of fresh RMPI 1640. Cells were subsequently incubated at 37° C. using MTT at 0.5 mg/mL during 4 h. After removal of supernatant, formazan crystals were dissolved in isopropanol and the optical density was measured at 570 nm. CX-4945 was used as a positive control. By way of example, the compound (1) displayed promising anti-proliferative activity against human cancer cells as reported in Table 1. TABLE 1Cytotoxicity activity of compound of Compound (1) and CX-4945on various human cancer cellsaCancer cell linesCompound (1)CX-4945PC3 (prostate)2.22.0HCT116 (colon)1.82.3A375 (melanoma)3.54.1H1299 (lung)1.92.3MIAPaCa-2 (pancreas)1.41.0HL60 (leukemia)3.33.7MCF7 (breast)6.99.1MDA-MB-231 (breast)5.56.2aCells were exposed for 96 hours and the number of viable cells was measured using the MTS reagent. IC50values were calculated as the concentration of compound eliciting a 50% inhibition of cell proliferation expressed in μM. The biological results demonstrated that the compound (1) displayed promising in vitro anti-proliferative activity against various human cancer cell lines similar to that of CX-4945 used as reference drug. Example 17 Evaluation of Inhibitory Activity on Protein Kinase CK2 CK2 Kinase Assay was conducted using the protocol as described in Pierre F. et al;J. Med. Chem.2011, 54, 635-654. The tested compounds in aqueous solution were added at a volume of 10 μL to a reaction mixture comprising 10 μL of assay dilution buffer (ADB; 20 mM MOPS, pH 7.2, 25 mM (β-glycerol phosphate, 5 mM EGTA, 1 mM sodium orthovanadate, and 1 mM dithiothreitol), 10 μL of substrate peptide (RRRDDDSDDD, dissolved in ADB at a concentration of 1 mM), 10 μL of recombinant human CK2 (RRfββ-holoenzyme, 25 ng dissolved in ADB; Millipore). Reactions were initiated by the addition of 10 μL of ATP solution (90% 75 mMMgCl2, 75 μM ATP (final ATP concentration: 15 μM) dissolved in ADB; 10% [γ-33P] ATP (stock 1 mCi/100 μL; 3000 Ci/mmol (Perkin-Elmer) and maintained for 10 min at 30° C. The reactions were quenched with 100 μL of 0.75% phosphoric acid and then transferred to and filtered through a phosphor cellulose filter plate (Millipore). After washing each well five times with 0.75% phosphoric acid, the plate was dried under vacuum for 5 min and, following the addition of 15 μL of scintillation fluid to each well, the residual radioactivity was measured using a Wallac luminescence counter. The IC50values were derived from eight concentrations of test inhibitors. The biological results demonstrated that the present compounds possessed favourable CK2 inhibition with IC50at a nanomolar concentration range. By way of example, the compound (1) displayed promising protein kinase CK2 activity with an IC50of 15 nM, while in the same experimental condition, the reference control CX-4945 inhibited protein kinase CK2 activity of 1 nM. It is to be understood that the pyrido[4′,3′:5,6]pyrimido[1,2-a]indole derivatives are not limited to the specific embodiments described above, but encompasses any and all embodiments within the scope of the generic language of the following claims enabled by the embodiments described herein, or otherwise shown in the drawings or described above in terms sufficient to enable one of ordinary skill in the art to make and use the claimed subject matter.
54,477
11858935
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS The following definitions are provided for the purpose of understanding the present subject matter and for construing the appended patent claims. Definitions Throughout the application, where compositions are described as having, including, or comprising specific components, or where processes are described as having, including, or comprising specific process steps, it is contemplated that compositions of the present teachings can also consist essentially of, or consist of, the recited components, and that the processes of the present teachings can also consist essentially of, or consist of, the recited process steps. It is noted that, as used in this specification and the appended claims, the singular forms “a”, “an”, and “the” include plural references unless the context clearly dictates otherwise. In the application, where an element or component is said to be included in and/or selected from a list of recited elements or components, it should be understood that the element or component can be any one of the recited elements or components, or the element or component can be selected from a group consisting of two or more of the recited elements or components. Further, it should be understood that elements and/or features of a composition or a method described herein can be combined in a variety of ways without departing from the spirit and scope of the present teachings, whether explicit or implicit herein. The use of the terms “include,” “includes”, “including,” “have,” “has,” or “having” should be generally understood as open-ended and non-limiting unless specifically stated otherwise. The use of the singular herein includes the plural (and vice versa) unless specifically stated otherwise. In addition, where the use of the term “about” is before a quantitative value, the present teachings also include the specific quantitative value itself, unless specifically stated otherwise. As used herein, the term “about” refers to a ±10% variation from the nominal value unless otherwise indicated or inferred. As used herein, “halo” or “halogen” refers to fluoro, chloro, bromo, and iodo. As used herein, “alkyl” refers to a straight-chain or branched saturated hydrocarbon group. Examples of alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and z′-propyl), butyl (e.g., n-butyl, z′-butyl, sec-butyl, tert-butyl), pentyl groups (e.g., n-pentyl, z′-pentyl, -pentyl), hexyl groups, and the like. In various embodiments, an alkyl group can have 1 to 40 carbon atoms (i.e., C1-C40alkyl group), for example, 1-30 carbon atoms (i.e., C1-C30alkyl group). In some embodiments, an alkyl group can have 1 to 6 carbon atoms, and can be referred to as a “lower alkyl group” or a “C1-C6alkyl group”. Examples of lower alkyl groups include methyl, ethyl, propyl (e.g., n-propyl and z′-propyl), and butyl groups (e.g., n-butyl, z′-butyl, sec-butyl, tert-butyl). In some embodiments, alkyl groups can be substituted as described herein. An alkyl group is generally not substituted with another alkyl group, an alkenyl group, or an alkynyl group. As used herein, “alkenyl” refers to a straight-chain or branched alkyl group having one or more carbon-carbon double bonds. Examples of alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl groups, and the like. The one or more carbon-carbon double bonds can be internal (such as in 2-butene) or terminal (such as in 1-butene). In various embodiments, an alkenyl group can have 2 to 40 carbon atoms (i.e., C2-C40alkenyl group), for example, 2 to 20 carbon atoms (i.e., C2-C20alkenyl group) or 2 to 6 carbon atoms (i.e., C2-C6alkenyl group). In some embodiments, alkenyl groups can be substituted as described herein. An alkenyl group is generally not substituted with another alkenyl group, an alkyl group, or an alkynyl group. The term “substituted alkyl” as used herein refers to an alkyl group in which 1 or more (up to about 5, for example about 3) hydrogen atoms is replaced by a substituent independently selected from the group: —O, —S, acyl, acyloxy, optionally substituted alkoxy, optionally substituted amino (wherein the amino group may be a cyclic amine), azido, carboxyl, (optionally substituted alkoxy)carbonyl, amido, cyano, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, halogen, hydroxyl, nitro, sulfamoyl, sulfanyl, sulfinyl, sulfonyl, and sulfonic acid. Some of the optional substituents for alkyl are hydroxy, halogen exemplified by chloro and bromo, acyl exemplified by methylcarbonyl; alkoxy, and heterocyclyl exemplified by morpholino and piperidino. Other alkyl substituents as described herein may further be contemplated. The term “substituted alkenyl” refers to an alkenyl group in which 1 or more (up to about 5, for example about 3) hydrogen atoms is replaced by a substituent independently selected from those listed above with respect to a substituted alkyl. Other alkenyl substituents as described herein may further be contemplated. As used herein, “heteroatom” refers to an atom of any element other than carbon or hydrogen and includes, for example, nitrogen, oxygen, silicon, sulfur, phosphorus, and selenium. As used herein, “aryl” refers to an aromatic monocyclic hydrocarbon ring system or a polycyclic ring system in which two or more aromatic hydrocarbon rings are fused (i.e., having a bond in common with) together or at least one aromatic monocyclic hydrocarbon ring is fused to one or more cycloalkyl and/or cycloheteroalkyl rings. An aryl group can have 6 to 24 carbon atoms in its ring system (e.g., C6-C24aryl group), which can include multiple fused rings. In some embodiments, a polycyclic aryl group can have 8 to 24 carbon atoms. Any suitable ring position of the aryl group can be covalently linked to the defined chemical structure. Examples of aryl groups having only aromatic carbocyclic ring(s) include phenyl, 1-naphthyl (bicyclic), 2-naphthyl (bicyclic), anthracenyl (tricyclic), phenanthrenyl (tricyclic), pentacenyl (pentacyclic), and like groups. Examples of polycyclic ring systems in which at least one aromatic carbocyclic ring is fused to one or more cycloalkyl and/or cycloheteroalkyl rings include, among others, benzo derivatives of cyclopentane (i.e., an indanyl group, which is a 5,6-bicyclic cycloalkyl/aromatic ring system), cyclohexane (i.e., a tetrahydronaphthyl group, which is a 6,6-bicyclic cycloalkyl/aromatic ring system), imidazoline (i.e., a benzimidazolinyl group, which is a 5,6-bicyclic cycloheteroalkyl/aromatic ring system), and pyran (i.e., a chromenyl group, which is a 6,6-bicyclic cycloheteroalkyl/aromatic ring system). Other examples of aryl groups include benzodioxanyl, benzodioxolyl, chromanyl, indolinyl groups, and the like. In some embodiments, aryl groups can be substituted as described herein. In some embodiments, an aryl group can have one or more halogen substituents, and can be referred to as a “haloaryl” group. Perhaloaryl groups, i.e., aryl groups where all of the hydrogen atoms are replaced with halogen atoms (e.g., —C6F5), are included within the definition of “haloaryl”. In certain embodiments, an aryl group is substituted with another aryl group and can be referred to as a biaryl group. Each of the aryl groups in the biaryl group can be substituted as disclosed herein. As used herein, “heteroaryl” refers to an aromatic monocyclic ring system containing at least one ring heteroatom selected from oxygen (O), nitrogen (N), sulfur (S), silicon (Si), and selenium (Se) or a polycyclic ring system where at least one of the rings present in the ring system is aromatic and contains at least one ring heteroatom. Polycyclic heteroaryl groups include those having two or more heteroaryl rings fused together, as well as those having at least one monocyclic heteroaryl ring fused to one or more aromatic carbocyclic rings, non-aromatic carbocyclic rings, and/or non-aromatic cycloheteroalkyl rings. A heteroaryl group, as a whole, can have, for example, 5 to 24 ring atoms and contain 1-5 ring heteroatoms (i.e., 5-20 membered heteroaryl group). The heteroaryl group can be attached to the defined chemical structure at any heteroatom or carbon atom that results in a stable structure. Generally, heteroaryl rings do not contain O—O, S—S, or S-0 bonds. However, one or more N or S atoms in a heteroaryl group can be oxidized (e.g., pyridine N-oxide thiophene S-oxide, thiophene S,S-dioxide). Examples of heteroaryl groups include, for example, the 5- or 6-membered monocyclic and 5-6 bicyclic ring systems shown below: where T is O, S, NH, N-alkyl, N-aryl, N-(arylalkyl) (e.g., N-benzyl), SiH2, SiH(alkyl), Si(alkyl)2, SiH(arylalkyl), Si(arylalkyl)2, or Si(alkyl)(arylalkyl). Examples of such heteroaryl rings include pyrrolyl, furyl, thienyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazolyl, tetrazolyl, pyrazolyl, imidazolyl, isothiazolyl, thiazolyl, thiadiazolyl, isoxazolyl, oxazolyl, oxadiazolyl, indolyl, isoindolyl, benzofuryl, benzothienyl, quinolyl, 2-methylquinolyl, isoquinolyl, quinoxalyl, quinazolyl, benzotriazolyl, benzimidazolyl, benzothiazolyl, benzisothiazolyl, benzisoxazolyl, benzoxadiazolyl, benzoxazolyl, cinnolinyl, 1H-indazolyl, 2H-indazolyl, indolizinyl, isobenzofuyl, naphthyridinyl, phthalazinyl, pteridinyl, purinyl, oxazolopyridinyl, thiazolopyridinyl, imidazopyridinyl, furopyridinyl, thienopyridinyl, pyridopyrimidinyl, pyridopyrazinyl, pyridopyridazinyl, thienothiazolyl, thienoxazolyl, thienoimidazolyl groups, and the like. Further examples of heteroaryl groups include 4,5,6,7-tetrahydroindolyl, tetrahydroquinolinyl, benzothienopyridinyl, benzofuropyridinyl groups, and the like. In some embodiments, heteroaryl groups can be substituted as described herein. The term “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, “optionally substituted alkyl” means either “alkyl” or “substituted alkyl,” as defined herein. It will be understood by those skilled in the art with respect to any chemical group containing one or more substituents that such groups are not intended to introduce any substitution or substitution patterns that are sterically impractical and/or physically non-feasible. The term “isomers” or “stereoisomers” as used herein relates to compounds that have identical molecular formulae but that differ in the arrangement of their atoms in space. Stereoisomers that are not mirror images of one another are termed “diastereoisomers” and stereoisomers that are non-superimposable mirror images are termed “enantiomers,” or sometimes optical isomers. A carbon atom bonded to four non-identical substituents is termed a “chiral center.” Certain compounds herein have one or more chiral centers and therefore may exist as either individual stereoisomers or as a mixture of stereoisomers. Configurations of stereoisomers that owe their existence to hindered rotation about double bonds are differentiated by their prefixes cis and trans (or Z and E), which indicate that the groups are on the same side (cis or Z) or on opposite sides (trans or E) of the double bond in the molecule according to the Cahn-Ingold-Prelog rules. All possible stereoisomers are contemplated herein as individual stereoisomers or as a mixture of stereoisomers. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which the presently described subject matter pertains. Where a range of values is provided, for example, concentration ranges, percentage ranges, or ratio ranges, it is understood that each intervening value, to the tenth of the unit of the lower limit, unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the described subject matter. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and such embodiments are also encompassed within the described subject matter, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the described subject matter. Throughout the application, descriptions of various embodiments use “comprising” language. However, it will be understood by one of skill in the art, that in some specific instances, an embodiment can alternatively be described using the language “consisting essentially of” or “consisting of”. “Subject” as used herein refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, and pet companion animals such as household pets and other domesticated animals such as, but not limited to, cattle, sheep, ferrets, swine, horses, poultry, rabbits, goats, dogs, cats and the like. “Patient” as used herein refers to a subject in need of treatment of a condition, disorder, or disease, such as cancer. For purposes of better understanding the present teachings and in no way limiting the scope of the teachings, unless otherwise indicated, all numbers expressing quantities, percentages or proportions, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term “about”. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. In an embodiment, the present subject matter relates to a compound having the formula I: or a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof, wherein:R is selected from the group consisting of —Ar, —NHAr, —NHCH2Ar, —NHCOAr, —COAr, —NHSO2Ar, —SO2Ar, —NHCH2CH2NHR3, —NHCH2CH2NR3R4, —NHCH2CH2CH2NHR3, —NHCH2CH2CH2NR3R4, —NHCOCH2CH2NHR3, —NHCOCH2CH2NR3R4, —NHCOCH2CH2CH2NHR3, —NHCOCH2CH2CH2NR3R4, —NHSO2CH2CH2NHR3, —NHSO2CH2CH2NR3R4, —NHSO2CH2CH2CH2NHR3, —NHSO2CH2CH2CH2NR3R4, —CH2Ar, —CH2CH2NHR3, —CH2CH2NR3R4, —CH2CH2CH2NHR3, —CH2CH2CH2NR3R4, —COCH2CH2NHR3, —COCH2CH2NR3R4, —COCH2CH2CH2NHR3, —COCH2CH2CH2NR3R4, —SO2CH2CH2NHR3, —SO2CH2CH2NR3R4, —SO2CH2CH2CH2NHR3, and —SO2CH2CH2CH2NR3R4;Ar is an aryl ring or a 5 or 6 membered heteroaryl ring, either of the aryl ring or the heteroaryl ring being optionally substituted with one or more substituents selected from the group consisting of halogen, cyano, —OH, —OR1, —NH2, —NHR1, —NR1R2, —CONH2, —CONHR1, —CONR1R2, —SO2NH2, —SO2NHR1, —SO2NR1R2, —NHCOR1, —NHCO2R1, —NHCONHR1, —NHSO2NHR1, —NHSO2R1, —COR1, —CO2R1, —CH2NHR1, —CH2NR1R2, —OCH2CO2R1, —OCH2CONHR1, —OCH2CONHR1R2, —OCH2COR1, —OCH2CONHR1, —OCH2CONR1R2, C1-C6straight chained alkyl, C1-C6branched alkyl, C3-C6cycloalkyl, halo-C1-C6straight chain alkyl, halo-C1-C6branched alkyl, halo-C3-C6cycloalkyl, cyano-C1-C6straight chain alkyl, cyano-C1-C6branched alkyl, and cyano-C3-C6cycloalkyl;R1and R2are each independently selected from the group consisting of hydrogen, C1-C6straight chained alkyl, C1-C6branched alkyl, C3-C6cycloalkyl, halo-C1-C6straight chain alkyl, halo-C1-C6branched alkyl, halo-C3-C6cycloalkyl, cyano-C1-C6straight chain alkyl, cyano-C1-C6branched alkyl, and cyano-C3-C6cycloalkyl; andR3and R4are each independently selected from the group consisting of hydrogen, C1-C6straight chained alkyl, C1-C6branched alkyl, C3-C6cycloalkyl, halo-C1-C6straight chain alkyl, halo-C1-C6branched alkyl, halo-C3-C6cycloalkyl, cyano-C1-C6straight chain alkyl, cyano-C1-C6branched alkyl, and cyano-C3-C6cycloalkyl, or wherein R3and R4, taken together with a nitrogen atom to which they are attached, form a nitrogen-containing heterocycle, such nitrogen-containing heterocycle being optionally substituted with from one to three substituents independently selected from the group consisting of a straight or branched C1-C6alkyl group, a halogen atom, a straight or branched C1-C6alkoxy group, a straight or branched C1-C6trihaloalkyl group, and a hydroxy group. In a further embodiment, the present subject matter relates to compounds of formula I, wherein R is an optionally substituted —NHAr group, an optionally substituted —NHCH2Ar group, or an optionally substituted —NHCH2CH2CH2NR3R4group. In yet another embodiment, the present subject matter relates to compounds of formula I, wherein R is —NHphenyl substituted with one or more substituents independently selected from the group consisting of —NR1R2, a straight or branched C1-C6alkyl, —CONR1R2, halogen, —SO2NR1R2, and —OR1. In a further embodiment, the present subject matter relates to compounds of formula I, wherein R is —NHphenyl substituted with one or more substituents independently selected from the group consisting of methyl, —CON(CH3)2, chlorine, —N(CH3)2, —SO2N(CH3)2, isopropyl, and methoxy. In this regard, in certain embodiments, R can be phenyl substituted with one or two chlorines, one or two methyls, one —CON(CH3)2, one —SO2N(CH3)2, one —N(CH3)2, one isopropyl, or one methoxy. In yet another embodiment, R may be a (pyridin-3-ylmethyl)amino group. In another embodiment, the present subject matter relates to a compound of formula I, wherein R is a (2-methylpyridin-4-yl)amino group. In still yet another embodiment, the present subject matter relates to compounds of formula I, wherein R is a —NHCH2CH2CH2NR3R4, wherein R3and R4are independently a straight chain C1-C6alkyl group, or wherein R3and R4are taken together with the nitrogen atom to which they are attached to form a nitrogen-containing heterocycle, such nitrogen-containing heterocycle being optionally substituted with a straight or branched C1-C6alkyl group. In this regard, R3and R4can both be methyl, or R3and R4can be taken together to form a 4-methylpiperazin-1-yl ring. In one embodiment, the present subject matter relates to a compound of formula I, wherein R is a —CH2phenyl group, wherein the phenyl in the —CH2phenyl group is substituted with a chlorine. In another embodiment, the present subject matter relates to a compound having the formula I: or a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof, wherein:R is —NHphenyl substituted with one or more substituents independently selected from the group consisting of —NR1R2, a straight or branched C1-C6alkyl, —CONR1R2, —SO2NR1R2, halogen, and —OR1, wherein R1and R2are independently a straight chain C1-C6alkyl group; a —NHCH2CH2CH2NR3R4group; a —NHCH2phenyl group substituted with a chlorine; a (2-methylpyridin-4-yl)amino group; and a (pyridin-3-ylmethyl)amino group; andR3and R4are independently a straight chain C1-C6alkyl group or wherein R3and R4are taken together with the nitrogen atom to which they are attached to form a nitrogen-containing heterocycle, such nitrogen-containing heterocycle being optionally substituted with a straight or branched C1-C6alkyl group. In an embodiment, the present subject matter relates to a compound selected from the group consisting of: 4-((3-chlorophenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (1), 5-(m-tolylamino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (2), 4-((3-methoxyphenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (3), 4-((3-(dimethylamino)phenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (4), 4-((4-chlorophenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (5), 4-(p-tolylamino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (6), 4-((4-methoxyphenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (7), 4-((4-(dimethylamino)phenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (8), 4-((3-(dimethylcarbamoyl)phenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (9), 4-((3-(N,N-dimethylsulfamoyl)phenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (10), 4-((3-isopropylphenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (11), 4-((2-methylpyridin-4-yl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (12), 4-((3-chlorobenzyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (13), 4-((pyridin-3-ylmethyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (14), 4-((3-(dimethylamino)propyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (15), 4-((3-(4-methylpiperazin-1-yl)propyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (16), and a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof. Said differently, the present subject matter can relate to compounds of formula I selected from the group consisting of: and a pharmaceutically acceptable salt, ester, stereoisomer, or solvate thereof. It is to be understood that the present subject matter covers all combinations of substituent groups referred to herein. The present compounds may contain, e.g., when isolated in crystalline form, varying amounts of solvents. Accordingly, the present subject matter includes all solvates of the present compounds of formula I and pharmaceutically acceptable stereoisomers, esters, and/or salts thereof. Hydrates are one example of such solvates. Further, the present subject matter includes all mixtures of possible stereoisomers of the embodied compounds, independent of the ratio, including the racemates. Salts of the present compounds, or salts of the stereoisomers thereof, include all inorganic and organic acid addition salts and salts with bases, especially all pharmaceutically acceptable inorganic and organic acid addition salts and salts with bases, particularly all pharmaceutically acceptable inorganic and organic acid addition salts and salts with bases customarily used in pharmacy. Examples of acid addition salts include, but are not limited to, hydrochlorides, hydrobromides, phosphates, nitrates, sulfates, acetates, trifluoroacetates, citrates, D-gluconates, benzoates, 2-(4-hydroxy-benzoyl)benzoates, butyrates, subsalicylates, maleates, laurates, malates, lactates, fumarates, succinates, oxalates, tartrates, stearates, benzenesulfonates (besilates), toluenesulfonates (tosilates), methanesulfonates (mesilates) and 3-hydroxy-2-naphthoates. Examples of salts with bases include, but are not limited to, lithium, sodium, potassium, calcium, aluminum, magnesium, titanium, ammonium, meglumine and guanidinium salts. The salts include water-insoluble and, particularly, water-soluble salts. The present compounds, the salts, the stereoisomers and the salts of the stereoisomers thereof may contain, e.g., when isolated in crystalline form, varying amounts of solvents. Included within the present scope are, therefore, all solvates of the compounds of formula I, as well as the solvates of the salts, the stereoisomers and the salts of the stereoisomers of the compounds of formula I. The present compounds may be isolated and purified in a manner known per se, e.g., by distilling off the solvent in vacuo and recrystallizing the residue obtained from a suitable solvent or subjecting it to one of the customary purification methods, such as column chromatography on a suitable support material. Salts of the compounds of formula I and the stereoisomers thereof can be obtained by dissolving the free compound in a suitable solvent (by way of non-limiting example, a ketone such as acetone, methylethylketone or methylisobutylketone; an ether such as diethyl ether, tetrahydrofurane or dioxane; a chlorinated hydrocarbon such as methylene chloride or chloroform; a low molecular weight aliphatic alcohol such as methanol, ethanol or isopropanol; a low molecular weight aliphatic ester such as ethyl acetate or isopropyl acetate; or water) which contains the desired acid or base, or to which the desired acid or base is then added. The acid or base can be employed in salt preparation, depending on whether a mono- or polybasic acid or base is concerned and depending on which salt is desired, in an equimolar quantitative ratio or one differing therefrom. The salts are obtained by filtering, reprecipitating, precipitating with a non-solvent for the salt or by evaporating the solvent. Salts obtained can be converted into the free compounds which, in turn, can be converted into salts. In this manner, pharmaceutically unacceptable salts, which can be obtained, for example, as process products in the manufacturing on an industrial scale, can be converted into pharmaceutically acceptable salts by processes known to the person skilled in the art. Pure diastereomers and pure enantiomers of the present compounds can be obtained, e.g., by asymmetric synthesis, by using chiral starting compounds in synthesis and by splitting up enantiomeric and diastereomeric mixtures obtained in synthesis. Preferably, the pure diastereomeric and pure enantiomeric compounds are obtained by using chiral starting compounds in synthesis. Enantiomeric and diastereomeric mixtures can be split up into the pure enantiomers and pure diastereomers by methods known to a person skilled in the art. Preferably, diastereomeric mixtures are separated by crystallization, in particular fractional crystallization, or chromatography. Enantiomeric mixtures can be separated, e.g., by forming diastereomers with a chiral auxiliary agent, resolving the diastereomers obtained and removing the chiral auxiliary agent. As chiral auxiliary agents, for example, chiral acids can be used to separate enantiomeric bases and chiral bases can be used to separate enantiomeric acids via formation of diastereomeric salts. Furthermore, diastereomeric derivatives such as diastereomeric esters can be formed from enantiomeric mixtures of alcohols or enantiomeric mixtures of acids, respectively, using chiral acids or chiral alcohols, respectively, as chiral auxiliary agents. Additionally, diastereomeric complexes or diastereomeric clathrates may be used for separating enantiomeric mixtures. Alternatively, enantiomeric mixtures can be split up using chiral separating columns in chromatography. Another suitable method for the isolation of enantiomers is enzymatic separation. In one embodiment, the present compounds can be prepared according to the following general synthetic pathway. Specifically, synthesis commences with adding, to a solution of N-bromosuccinimide in Dimethylformamide (DMF), a solution of diethyl 1H-pyrrole-2,5-dicarboxylate I in DMF. The reaction mixture is allowed to warm up to room temperature over at least about 12 hours before it is quenched with ice water and then extracted with ethyl acetate. The organic layer is washed with saturated NaHCO3and water, dried over MgSO4, and concentrated. The resulting residue is purified to give the intermediate diethyl 5-bromo-1H-pyrrole-2,4-dicarboxylate. To a solution of diethyl 5-bromo-1H-pyrrole-2,4-dicarboxylate in Tetrahydrofuran (THF) is added potassium tert-butoxide. After 10 minutes of stirring, di-tert-butyl dicarbonate is then added to the reaction mixture and stirred for an additional at least about 12 hours at room temperature. Then water is added cautiously, and the resulting mixture is extracted with ether. The combined organic layers are dried over MgSO4, evaporated under reduced pressure, and the residue is purified to afford the 1-(tert-butyl) 2,5-diethyl 3-bromo-1H-pyrrole-1,2,5-tricarboxylate II as outlined in Scheme 1. Next, to a solution of 1-(tert-butyl) 2,5-diethyl 3-bromo-1H-pyrrole-1,2,5-tricarboxylate II in DMF are added palladium tetrakis-triphenylphosphine, (3-nitropyridin-4-yl)boronic acid pinacol ester III, and sodium carbonate dissolved in minimal water. The reaction mixture is then heated to about 105° C. to about 115° C. for at least about 14 h under nitrogen atmosphere. After cooling to room temperature, the reaction is diluted with water and extracted with ethyl acetate. The combined organic layers are dried over MgSO4, filtered, evaporated under reduced pressure, and the residue is purified by to obtain diethyl 3-(3-nitropyridin-4-yl)-1H-pyrrole-2,5-dicarboxylate IV, as outlined in Scheme 2. Next, a mixture of diethyl 3-(3-nitropyridin-4-yl)-1H-pyrrole-2,5-dicarboxylate IV, Palladium on Carbon (Pd/C), and EtOH is stirred under hydrogen atmosphere for at least about 2 hours. The catalyst is removed by filtration through celite, and the filtrate is then transferred to a round bottom flask and heated at reflux for 12 hours. After removal of the solvent under reduced pressure, the resulting residue is treated with POCl3and heated at reflux for 2 hours. After cooling to room temperature, the reaction mixture is transferred to ice-water and neutralized by a 20% ammonia solution and extracted with CH2Cl2. The combined organic layers are dried over MgSO4, filtered, evaporated under reduced pressure, and the residue is purified to yield ethyl 4-chloro-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate V, as outlined in Scheme 3 The final step of the synthesis involves heating a mixture of ethyl 4-chloro-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate V and a variety of amines in 2-ethoxyethanol at reflux for at least about 6 hours. After cooling to room temperature, water is added and extracted with CH2Cl2. The combined organic layers are dried over MgSO4, filtrated, and evaporated to provide the crude product which is purified to provide the intermediate ethyl 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate which is engaged in the next step. A mixture of ethyl 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate and NaOH in THF is heated at reflux for at least about 2 hours. After cooling to room temperature, a solution of 10% HCl is added and the resulting precipitate is filtrated, washed with water, and recrystallized to furnish the corresponding 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI (compounds 1-16) as shown in Scheme 4. In another embodiment, the present subject matter is directed to pharmaceutical compositions comprising a therapeutically effective amount of the compounds as described herein together with one or more pharmaceutically acceptable carriers, excipients, or vehicles. In some embodiments, the present compositions can be used for combination therapy, where other therapeutic and/or prophylactic ingredients can be included therein. The present subject matter further relates to a pharmaceutical composition, which comprises at least one of the present compounds together with at least one pharmaceutically acceptable auxiliary. In an embodiment, the pharmaceutical composition comprises one or two of the present compounds, or one of the present compounds. Non-limiting examples of suitable excipients, carriers, or vehicles useful herein include liquids such as water, saline, glycerol, polyethylene glycol, hyaluronic acid, ethanol, and the like. Suitable excipients for nonliquid formulations are also known to those of skill in the art. A thorough discussion of pharmaceutically acceptable excipients and salts useful herein is available in Remington's Pharmaceutical Sciences, 18th Edition. Easton, Pa., Mack Publishing Company, 1990, the entire contents of which are incorporated by reference herein. The present compounds are typically administered at a therapeutically or pharmaceutically effective dosage, e.g., a dosage sufficient to provide treatment for cancer. Administration of the compounds or pharmaceutical compositions thereof can be by any method that delivers the compounds systemically and/or locally. These methods include oral routes, parenteral routes, intraduodenal routes, and the like. While human dosage levels have yet to be optimized for the present compounds, generally, a daily dose is from about 0.01 to 10.0 mg/kg of body weight, for example about 0.1 to 5.0 mg/kg of body weight. The precise effective amount will vary from subject to subject and will depend upon the species, age, the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. The subject may be administered as many doses as is required to reduce and/or alleviate the signs, symptoms, or causes of the disease or disorder in question, or bring about any other desired alteration of a biological system. In employing the present compounds for treatment of cancer, any pharmaceutically acceptable mode of administration can be used with other pharmaceutically acceptable excipients, including solid, semi-solid, liquid or aerosol dosage forms, such as, for example, tablets, capsules, powders, liquids, suspensions, suppositories, aerosols or the like. The present compounds can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, and the like, for the prolonged administration of the compound at a predetermined rate, preferably in unit dosage forms suitable for single administration of precise dosages. The present compounds may also be administered as compositions prepared as foods for humans or animals, including medical foods, functional food, special nutrition foods and dietary supplements. A “medical food” is a product prescribed by a physician that is intended for the specific dietary management of a disorder or health condition for which distinctive nutritional requirements exist and may include formulations fed through a feeding tube (referred to as enteral administration or gavage administration). A “dietary supplement” shall mean a product that is intended to supplement the human diet and may be provided in the form of a pill, capsule, tablet, or like formulation. By way of non-limiting example, a dietary supplement may include one or more of the following dietary ingredients: vitamins, minerals, herbs, botanicals, amino acids, and dietary substances intended to supplement the diet by increasing total dietary intake, or a concentrate, metabolite, constituent, extract, or combinations of these ingredients, not intended as a conventional food or as the sole item of a meal or diet. Dietary supplements may also be incorporated into foodstuffs, such as functional foods designed to promote control of glucose levels. A “functional food” is an ordinary food that has one or more components or ingredients incorporated into it to give a specific medical or physiological benefit, other than a purely nutritional effect. “Special nutrition food” means ingredients designed for a particular diet related to conditions or to support treatment of nutritional deficiencies. Generally, depending on the intended mode of administration, the pharmaceutically acceptable composition will contain about 0.1% to 90%, for example about 0.5% to 50%, by weight of a compound or salt of the present compounds, the remainder being suitable pharmaceutical excipients, carriers, etc. One manner of administration for the conditions detailed above is oral, using a convenient daily dosage regimen which can be adjusted according to the degree of affliction. For such oral administration, a pharmaceutically acceptable, non-toxic composition is formed by the incorporation of any of the normally employed excipients, such as, for example, mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, sodium croscarmellose, glucose, gelatin, sucrose, magnesium carbonate, and the like. Such compositions take the form of solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations and the like. The present compositions may take the form of a pill or tablet and thus the composition may contain, along with the active ingredient, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinyl pyrrolidine, gelatin, cellulose, and derivatives thereof, and the like. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc. an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, for example, sodium acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine acetate, triethanolamine oleate, etc. For oral administration, a pharmaceutically acceptable non-toxic composition may be formed by the incorporation of any normally employed excipients, such as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, cellulose derivatives, sodium croscarmellose, glucose, sucrose, magnesium carbonate, sodium saccharin, talcum and the like. Such compositions take the form of solutions, suspensions, tablets, capsules, powders, sustained release formulations and the like. For a solid dosage form, a solution or suspension in, for example, propylene carbonate, vegetable oils or triglycerides, may be encapsulated in a gelatin capsule. Such diester solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545, the contents of each of which are incorporated herein by reference. For a liquid dosage form, the solution, e.g., in a polyethylene glycol, may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be easily measured for administration. Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and the like, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in U.S. Pat. Nos. Re. 28,819 and 4,358,603, the contents of each of which are hereby incorporated by reference. Another manner of administration is parenteral administration, generally characterized by injection, either subcutaneously, intramuscularly, or intravenously. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like. In addition, if desired, the pharmaceutical compositions to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, solubility enhancers, and the like, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate, cyclodextrins, etc. Another approach for parenteral administration employs the implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject. However, percentages of active ingredient of 0.01% to 10% in solution are employable and will be higher if the composition is a solid which will be subsequently diluted to the above percentages. The composition may comprise 0.2% to 2% of the active agent in solution. Nasal solutions of the active compound alone or in combination with other pharmaceutically acceptable excipients can also be administered. Formulations of the active compound or a salt may also be administered to the respiratory tract as an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation have diameters of less than 50 microns, for example less than 10 microns. The present compounds have valuable pharmaceutical properties, which make them commercially utilizable. Accordingly, the present subject matter further relates to use of the present compounds for the treatment of diseases such as cancers. Similarly, the present compounds can be used to inhibit CK2 enzyme activity in a patient. In another embodiment of the present subject matter, the aforementioned compound derivatives demonstrated in vitro anticancer action against human cancer cell lines such as MCF7 and MDA-MB-231 (breast cancer), H1299 (lung cancer), PC3 (prostate cancer), HCT116 (colon cancer), A375 (melanoma), MIAPaCa2 (pancreatic cancer), and HL60 (leukemia). Accordingly, the present subject matter relates to methods of treating a cancer in a patient by administering one or more of the compounds presented herein to a patient in need thereof. In certain embodiments, the cancer treatable with the present compounds is one or more selected from the group consisting of leukemia, melanoma, colon cancer, prostate cancer, lung cancer, pancreatic cancer, and breast cancer. Accordingly, in an embodiment of the present subject matter, the Pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid compounds as described herein engaged for in vitro study towards human cancer cell lines can display an IC50with a nano to micromolar concentration range when exposed to a period of at least 96 hrs. For example, a present compound engaged for in vitro study against PC3 (prostate) cancer cell lines can display an IC50concentration of 1.5 μM at an exposure period of at least 96 hrs. In another embodiment, a present compound engaged for in vitro study against HCT116 (colon) cancer cell lines can display an IC50concentration of 1.8 μM at an exposure period of at least 96 hrs. In a further embodiment, a present compound engaged for in vitro study against A375 (melanoma) cancer cell lines can display an IC50concentration of 2.3 μM at an exposure period of at least 96 hrs. In an embodiment, a present compound engaged for in vitro study against H1299 (lung) cancer cell lines can display an IC50concentration of 1.4 μM at an exposure period of at least 96 hrs. In another embodiment, a present compound engaged for in vitro study against MIAPaCa-2 (pancreas) cancer cell lines can display an IC50concentration of 1.7 μM at an exposure period of at least 96 hrs. In a further embodiment, a present compound engaged for in vitro study against HL60 (leukemia) cancer cell lines can display an IC50concentration of 3.1 μM at an exposure period of at least 96 hrs. In one embodiment, a present compound engaged for in vitro study against MCF7 (breast) cancer cell lines can display an IC50concentration of 4.2 μM at an exposure period of at least 96 hrs. In another embodiment, a present compound engaged for in vitro study against MDA-MB-231 (breast) cancer cell lines can display an IC50concentration of 3.6 μM at an exposure period of at least 96 hrs. The present subject matter further relates to a method of treating or preventing a disease comprising administering to a patient in need thereof a therapeutically effective amount of at least one of the compounds herein. In particular, the present subject matter relates to a method of treating one of the above-mentioned diseases or disorders comprising administering to a patient in need thereof a therapeutically effective amount of at least one of the compounds herein. In the above methods, the patient is preferably a mammal, more preferably a human. Furthermore, in the above methods, at least one of the present compounds can be used. In an embodiment, one or two of the present compounds are used, or one of the present compounds is used. Similarly, one or more of the present compounds can be used in combination therapy with one or more additional active agents. The following examples relate to various methods of manufacturing certain specific compounds as described herein. All compound numbers expressed herein are with reference to the synthetic pathway figures shown above. EXAMPLES Example 1 Preparation of 1-(tert-butyl) 2,5-diethyl 3-bromo-1H-pyrrole-1,2,5-tricarboxylate II A solution of diethyl 1H-pyrrole-2,5-dicarboxylate I (10 mmol) in DMF (15 mL) was added to a solution of N-bromosuccinimide (11 mmol) in DMF (10 mmol) at 10° C. The reaction mixture was allowed to warm up to room temperature over 12 hours before it was quenched with ice-water, and then extracted with ethyl acetate. The organic layer was washed with saturated NaHCO3and water, dried over MgSO4and concentrated. The resulting residue was purified by column chromatography on silica gel to give the intermediate diethyl 5-bromo-1H-pyrrole-2,4-dicarboxylate. Next, to a solution of diethyl 5-bromo-1H-pyrrole-2,4-dicarboxylate (10 mmol) in THF (20 mL) was added potassium tert-butoxide (15 mmol). After 10 min. of stirring, di-tert-butyl dicarbonate (12 mmol) was then added to the reaction mixture and stirred for an additional 12 h at room temperature. Water (15 ml) was then added cautiously, and the resulting mixture was extracted with ether (3×50 ml). The combined organic layers were dried over MgSO4, evaporated under reduced pressure, and the residue was purified by column chromatography on silica gel to afford the 1-(tert-butyl) 2,5-diethyl 3-bromo-1H-pyrrole-1,2,5-tricarboxylate II. Elemental Analysis: Calculated C, 46.17; H, 5.17; N, 3.59. Found C, 46.15; H, 5.19; N, 3.57. Example 2 Preparation of diethyl 3-(3-nitropyridin-4-yl)-1H-pyrrole-2,5-dicarboxylate IV Palladium tetrakis-triphenylphosphine (0.25 mmol), (3-nitropyridin-4-yl)boronic acid pinacol ester III (10 mmol), and sodium carbonate dissolved in minimal water (40 mmol) were added to a solution of 1-(tert-butyl) 2,5-diethyl 3-bromo-1H-pyrrole-1,2,5-tricarboxylate II (5 mmol) in DMF (15 mL). The reaction mixture was then heated to 110° C. for 14 hours under nitrogen atmosphere. After cooling to room temperature, the reaction was diluted with water (50 mL) and extracted with ethyl acetate (3×20 mL). The combined organic layers were dried over MgSO4, filtered, evaporated under reduced pressure, and the residue was purified by column chromatography on silica gel to obtain diethyl 3-(3-nitropyridin-4-yl)-1H-pyrrole-2,5-dicarboxylate IV. Elemental Analysis: Calculated C, 54.05; H, 4.54; N, 12.61. Found C, 54.03; H, 4.52; N, 12.57. Example 3 Preparation of ethyl 4-chloro-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate V A mixture of diethyl 3-(3-nitropyridin-4-yl)-1H-pyrrole-2,5-dicarboxylate IV (3 mmol), Pd/C (5%, 200 mg), and EtOH (15 mL) was stirred under hydrogen atmosphere for 2 hours. The catalyst was removed by filtration through celite, and the filtrate was then transferred to a round bottom flask and heated at reflux for 12 hours. After removal of the solvent under reduced pressure, the resulting residue was treated with POCl3(5 mL) and heated at reflux for 2 hours. After cooling to room temperature, the reaction mixture was transferred to ice-water and neutralized by a 20% ammonia solution and extracted with CH2Cl2(3×15 mL). The combined organic layers were dried over MgSO4, filtered, evaporated under reduced pressure, and the residue was purified by column chromatography on silica gel to yield ethyl 4-chloro-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate V. Elemental Analysis: Calculated C, 56.64; H, 3.66; N, 15.24. Found C, 56.59; H, 3.62; N, 15.20. Example 4 General method of preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI (Examples 1-16) A mixture of ethyl 4-chloro-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate V (1 mmol) and a variety of amines (2 mmol) in 5 mL 2-ethoxyethanol was heated at reflux for 6 hours. After cooling to room temperature, water (10 mL) was added and extracted with CH2Cl2(3×10 mL). The combined organic layers were dried over MgSO4, filtrated and evaporated to provide the crude product which was purified by column chromatography to provide the intermediate ethyl 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate which was engaged in the next step. A mixture of ethyl 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylate (1 mmol) and NaOH (3 mmol) in 5 ml, THF was heated at reflux for 2 hours. After cooling to room temperature, a solution of 10% HCl was added and the resulting precipitate was filtrated, washed with water and recrystallized to furnish the corresponding 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI. Example 5 Preparation of 4-((3-chlorophenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (1) The expected product was obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-chloroaniline as the amine. Elemental Analysis: Elemental Analysis: Calculated C, 60.28; H, 3.27; N, 16.54. Found C, 60.24; H, 3.31; N, 16.55. Example 6 Preparation of 5-(m-tolylamino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (2) The expected product was obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using m-toluidine as the amine. Elemental Analysis: Calculated C, 67.92; H, 4.43; N, 17.60. Found C, 67.91; H, 4.41; N, 17.57. Example 7 Preparation of 4-((3-methoxyphenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (3) The expected product was obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-methoxyaniline as the amine. Elemental Analysis: Calculated C, 64.67; H, 4.22; N, 16.76. Found C, 64.63; H, 4.25; N, 16.74. Example 8 Preparation of 4-((3-(dimethylamino)phenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (4) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-dimethylaminoaniline as the amine. Elemental Analysis: Calculated C, 65.69; H, 4.93; N, 20.16. Found C, 65.65; H, 4.88; N, 20.14. Example 9 Preparation of 4-((4-chlorophenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (5) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 4-chloroaniline as the amine. Elemental Analysis: Calculated C, 60.28; H, 3.27; N, 16.54. Found C, 60.25; H, 3.24; N, 16.51. Example 10 Preparation of 4-(p-tolylamino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (6) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 9-toluidine as the amine. Elemental Analysis: Calculated C, 67.92; H, 4.43; N, 17.60. Found C, 67.86; H, 4.44; N, 17.58. Example 11 Preparation of 4-((4-methoxyphenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (7) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 4-methoxyaniline as the amine. Elemental Analysis: Calculated C, 64.67; H, 4.22; N, 16.76. Found C. 64.68; H, 4.20; N, 16.73. Example 12 Preparation of 4-((4-(dimethylamino)phenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (8) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 4-dimethylaminoaniline as the amine. Elemental Analysis: Calculated C, 65.69; H, 4.93; N, 20.16. Found C. 65.71; H, 4.90; N, 20.11. Example 13 Preparation of 4-((3-(dimethylcarbamoyl)phenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (9) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-amino-N,N-dimethylbenzamide as the amine. Elemental Analysis: Calculated C, 63.99; H, 4.56; N, 18.66. Found C, 63.98; H, 4.52; N, 18.63. Example 14 Preparation of 4-((3-(N,N-dimethylsulfamoyl)phenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (10) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-amino-N-methylbenzenesulfonamide as the amine. Elemental Analysis: Calculated C, 54.40; H, 3.80; N, 17.62. Found C, 54.35; H, 3.77; N, 17.58. Example 15 Preparation of 4-((3-isopropylphenyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (11) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-isopropylaniline as the amine. Elemental Analysis: Calculated C, 69.35; H, 5.24; N, 16.17. Found C, 69.32; H, 5.21; N, 16.19. Example 16 Preparation of 4-((2-methylpyridin-4-yl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (12) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 2-methylpyridin-4-amine as the amine. Elemental Analysis: Calculated C, 63.94; H, 4.10; N, 21.93. Found C, 63.93; H, 4.06; N, 21.88. Example 17 Preparation of 4-((3-chlorobenzyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (13) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-chlorobenzyl amine as the amine. Elemental Analysis: Calculated C, 61.28; H, 3.71; N, 15.88. Found C, 61.25; H, 3.68; N, 15.86. Example 18 Preparation of 4-((pyridin-3-ylmethyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (14) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using pyridin-3-ylmethanamine as the amine. Elemental Analysis: Calculated C, 63.94; H, 4.10; N, 21.93. Found C, 63.96; H, 4.06; N, 21.89. Example 19 Preparation of 4-((3-(dimethylamino)propyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (15) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-(dimethylamino)propyl)amine as the amine. Elemental Analysis: Calculated C, 61.33; H, 6.11; N, 22.35. Found C, 61.29; H, 6.12; N, 22.31. Example 20 Preparation of 4-((3-(4-methylpiperazin-1-yl)propyl)amino)-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid (16) The expected product is obtained in accordance with the general procedure for the preparation of 4-substituted amino-3H-pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid VI of Example 4 using 3-(4-methylpiperazin-1-yl)propyl)amine as the amine. Elemental Analysis: Calculated C, 61.94; H, 6.57; N, 22.81. Found C, 61.89; H, 6.56; N, 22.80. Pharmacological Activity Example 21 In Vitro Cytotoxic Activity Assay Compounds 1-16 were screened for their in vitro cytotoxic activity utilizing a 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay against selected cancer human cell lines consisting of PC3 (prostate), HCT116 (colon), A375 (melanoma), H1299 (lung), MIAPaCa-2 (pancreas), HL60 (leukemia), MCF7 (breast), MDA-MB-231 (breast) (T. Mosmann, J. Immunol. Meth., 1983, 65, 55-63). The cells were cultured at 37° C. in RMPI1640 medium supplemented with 10% fetal bovine serum, 50 IU/mL penicillin, and 50 μg/mL streptomycin in a 5% CO2incubator. All cells were sub-cultured 3 times/week by trypsinisation. Viable cells were seeded and allowed to adhere for 12 h before a test drug was added in 96-well plates at an initial density of 1.0×105cells/mL. Tumor cell lines were separately exposed to various concentrations of the tested compounds followed by incubation at a temperature of 37° C. during 96 h inside a medium of fresh RMPI 1640. Cells were subsequently incubated at 37° C. using MTT at 0.5 mg/mL during 4 h. After removal of supernatant, formazan crystals were dissolved in isopropanol and the optical density was measured at 570 nm. CX-4945 was used as a positive control. By way of example, the compound (1) displayed promising anti-proliferative activity against human cancer cells as reported in Table 1. TABLE 1Cytotoxicity activity of compound of Example 1 andCX-4945 on various human cancer cellsªCancer cell linesExample 1CX-4945PC3 (prostate)1.52.0HCT116 (colon)1.82.3A375 (melanoma)2.34.1H1299 (lung)1.42.3MIAPaCa-2 (pancreas)1.71.0HL60 (leukemia)3.13.7MCF7 (breast)4.29.1MDA-MB-231 (breast)3.66.2aCells were exposed for 96 hours and the number of viable cells was measured using the MTS reagent. IC50values were calculated as the concentration of compound eliciting a 50% inhibition of cell proliferation expressed in μM. The biological results demonstrated that the compound (1) displayed promising in vitro anti-proliferative activity against various human cancer cell lines similar to that of CX-4945 used as reference drug. Example 22 Evaluation of Inhibitory Activity on Protein Kinase CK2 CK2 Kinase Assay was conducted using the protocol as described in Pierre F. et al;J. Med. Chem.2011, 54, 635-654. The tested compounds in aqueous solution were added at a volume of 10 μL to a reaction mixture comprising 10 μL of assay dilution buffer (ADB; 20 mM MOPS, pH 7.2, 25 mM β-glycerol phosphate, 5 mM EGTA, 1 mM sodium orthovanadate, and 1 mM dithiothreitol), 10 μL of substrate peptide (RRRDDDSDDD, dissolved in ADB at a concentration of 1 mM), 10 μL of recombinant human CK2 (RRββ-holoenzyme, 25 ng dissolved in ADB; Millipore). Reactions were initiated by the addition of 10 μL of ATP solution (90% 75 mM MgCl2, 75 μM ATP (final ATP concentration:15 μM) dissolved in ADB; 10% [γ-33P] ATP (stock 1 mCi/100 μL; 3000 Ci/mmol (Perkin-Elmer) and maintained for 10 min at 30° C. The reactions were quenched with 100 μL of 0.75% phosphoric acid and then transferred to and filtered through a phosphor cellulose filter plate (Millipore). After washing each well five times with 0.75% phosphoric acid, the plate was dried under vacuum for 5 min and, following the addition of 15 μL of scintillation fluid to each well, the residual radioactivity was measured using a Wallac luminescence counter. The IC50values were derived from eight concentrations of test inhibitors. The biological results demonstrated that the present compounds possessed favourable CK2 inhibition with IC50at a nanomolar concentration range. By way of example, the compound (1) displayed promising protein kinase CK2 activity with an IC50of 26 nM, while in the same experimental condition, the reference control CX-4945 inhibited protein kinase CK2 activity of 1 nM. It is to be understood that the pyrrolo[2,3-c][1,7]naphthyridine-2-carboxylic acid compounds are not limited to the specific embodiments described above, but encompasses any and all embodiments within the scope of the generic language of the following claims enabled by the embodiments described herein, or otherwise shown in the drawings or described above in terms sufficient to enable one of ordinary skill in the art to make and use the claimed subject matter.
60,788
11858936
DETAILED DESCRIPTION OF EMBODIMENTS OF THE INVENTION Embodiments of the invention are directed to synthetic co-crystals of anhydrous guanine and at least one additional material and to anhydrous guanine prepared according to the process of this invention. According to some embodiments, the synthetic co-crystals of the invention have a plate morphology in contrast to the bulky prismatic crystals of pure anhydrous guanine. According to some embodiments, the synthetic co-crystals have a preferred morphology which provides high refractive index. In another embodiment, the preferred morphology is a plate morphology. According to some embodiments, in a plate morphology, the co-crystals or the anhydrous guanine have a high surface to volume ratio, wherein the thickness of the co-crystals or the anhydrous guanine, which determines the pearlescence and/or the whiteness of the material, may be controlled by altering, e.g., the additive concentration, the pH, and/or the temperature. The synthetic co-crystal comprises at least 50 mol % of guanine. In another embodiment, the synthesis co-crystal comprises at least 0.01 mol % of the additional material. According to some embodiments, the additive concentration may be altered between about 0.01-50 mol %. In another embodiment, the additive concentration is between 0.1 to 3 mol %. In another embodiment, the additive concentration is between 5 to 10 mol %. In another embodiment, the additive concentration is between 10 to 50 mol %. In another embodiment, the additive concentration is between 0.1 to 10 mol %. In another embodiment, the additive concentration is between 25 to 50 mol %. In some embodiment, the concentration of the guanine and the additional material used in the preparation of the co-crystal is not the same concentration obtained in the co-crystal itself. According to some embodiments, the structure of the guanine and the additional material is similar. According to some embodiments, the additional material is a purine or a derivative thereof. According to some embodiments, the purine or the derivative thereof is hypoxanthine, xanthine, uric acid, isoguanine or theobromine. According to some embodiments, the additional material is pteridine or a derivative thereof. According to some embodiments, the pteridine derivative is isoxanthopterin or xanthopterin. According to some embodiments, the additional material is pyrimidine or a derivative thereof. According to some embodiments, the pyrimidine derivative is thymine, cytosine or uracil. According to some embodiments, the additional material is a nucleoside of a purine, pteridine or pyrimidine or a derivative thereof, including, though not limited to guanosine, cytidine, thymidine, uridine, adenosine and inosine. According to some embodiments, the co-crystals may include two or more additional materials. In one embodiment, the co-crystal of thus invention includes a guanine and another material. In another embodiment, the other material includes a purine, pteridine, pyrimidine, nucleoside of a purine, pteridine or pyrimidine; or derivative thereof. In another embodiments the derivatives includes between one to six substituents of the additional materials selected from the group consisting from keto, halo, cyano, amino, nitro, hydroxy, alkoxy, alkyl, alkenyl, aryl cycloalkyl, heterocycloalkyl or combination thereof. In another embodiment, the co-crystal of this invention includes anhydrous guanine and between 1-5 different additional materials. Each represents a separate embodiment of this invention. In another embodiment, the co-crystal of this invention includes a guanine and hypoxanthine in a plate morphology (FIG.8C). In another embodiment, the co-crystal including the guanine and hypoxanthine in a plate morphology provides X-ray powder diffraction as presented inFIG.8B. According to some embodiments, the refractive index of the co-crystals is between about 1.65-1.95. In another embodiment, the refractive index of the co-crystals is between about 1.75-1.86. According to some embodiments, the crystals are about 5-250 μm long. According to some embodiments, the crystals are about 1-50 μm wide. According to some embodiments, the crystals are about 20-500 nm thick. According to some embodiments, the crystals have smooth surfaces, wherein smooth surfaces refers to a surface baying an even and regular surface or consistency; free from perceptible projections, lumps, or indentations. In another embodiment, smooth surfaces are considered to be flat surfaces within a range of 1-10 nm. The morphology of the crystals is such as having a plane surface within a range of 1-10 nm. The dimensions of the crystals may be controlled by various conditions, such as the initial concentration of reactants, the temperature gradient during the process for forming the crystals, the initial pH, the final pH, the rate of pH adjustment during the process, the speed of stirring, the time during which the crystals are maintained in suspension, and the like. Further embodiments of the invention are directed to a pH controlled process for preparing synthetic co-crystals of anhydrous guanine and at least one additional material, wherein said process comprises the steps of:preparing a basic or acidic aqueous solution of guanine and at least one additional material;maintaining the basic or acidic aqueous solution at a predetermined temperature range for a predetermined period of time;filtering the basic or acidic aqueous solution to provide a filtrate;adjusting the pH of the filtrate by adding a base or an acid to the filtrate over a predefined period of time until a predetermined pH value is obtained, thereby providing a synthetic co-crystal suspension comprising co-crystals;allowing the synthetic co-crystal suspension to mature over a predetermined period of time; andcollecting the synthetic co-crystals from the crystal suspension. Further embodiments of the invention are directed to a pH controlled process for preparing anhydrous guanine, wherein said process comprises the steps of:preparing a basic or acidic aqueous solution of guanine;maintaining the basic or acidic aqueous solution at a predetermined temperature range for a predetermined period of time;filtering the basic or acidic aqueous solution to provide a filtrate;adjusting the pH of the nitrate by adding a base or an acid to the filtrate over a predefined period of time until a predetermined pH value is obtained, thereby providing a guanine crystal suspension comprising crystals;allowing the suspension to mature over a predetermined period of time; andcollecting the anhydrous guanine crystals from the crystal suspension. In some embodiments, the anhydrous guanine prepared by the pH controlled process of this invention is a crystalline anhydrous guanine. According to some embodiments, the pH of the processes is controlled in the reaction vessel, such that the guanine is protonated and deprotonated, affecting the solubility thereof in the basic or acidic aqueous solution. According to some embodiments, once the guanine is dissolved, the pH of the basic or acidic aqueous solution may be adjusted to induce crystallization, wherein the morphology of the produced crystals may be controlled by the particular pH used throughout the process. According to some embodiments, the pH of the processes may be altered between about 7-13. According to some embodiments, the temperature may be altered between about 4-60° C. Further, as detailed herein, the thickness of the co-crystals may also be controlled by parameters influencing the crystal size, such us the initial concentration of reactants, temp the temperature gradient during the process for forming the crystals, the initial pH, the final pH, the rate of pH adjustments during the process, the time during which the crystals are maintained left in suspension, and the like, as detailed herein. According to some embodiments, the molar ratio between the additional material and the guanine introduced into the reaction vessel of the process for preparing synthetic co-crystals is about 3:1. According to some embodiments, the molar ratio between the additional material and the guanine introduced into the reaction vessel is between about 4:1-0.01:1. In another embodiment, the molar ratio between the additional material and the guanine introduced into the reaction vessel is between about 4:1-1:1. In another embodiment, the molar ratio between the additional material and the guanine introduced into the reaction vessel is between about 4:1-0.5:1. In another embodiment, the molar ratio between the additional material and the guanine introduced into the reaction vessel is between about 4:1-0.1:1. In another embodiment, the molar ratio between the additional material and the guanine introduced into the reaction vessel is between about 4:1-0.05:1. According to some embodiments, the higher the solubility of the additional material, the higher the molar ratio between the additional material and the guanine. According to some embodiments, the basic or acidic aqueous solution comprises about 0.003-0.2M of guanine. According to some embodiments, the basic or acidic aqueous solution comprises about 0.003-0.01M of guanine. According to some embodiments, the basic or acidic aqueous solution comprises about 0.01-0.05M of guanine. According to some embodiments, the basic or acidic aqueous solution comprises about 0.05-0.1M of guanine. According to some embodiments, the basic or acidic aqueous solution comprises about 0.1-0.2M of guanine. According to some embodiments, the low concentration of guanine and/or the slow adjustment of pH, e.g., dropwise, induces fewer nucleation events and therefore, large crystals, having an average size of about 5-1000 μm of anhydrous guanine, are formed. In another embodiment, large crystals of anhydrous guanine having an average size of about 5-50 μm, are formed. In another embodiment, large crystals having an average size of about 10-100 μm, are formed. In another embodiment, large crystals having an average size of about 100-1000 μm, are formed. In another embodiment, large crystals having an average size of above 5-70 μm provide a strong pearlescent effect. According to some embodiments, at high concentration of guanine, between about 0.1-0.2M, and when the pH is rapidly adjusted, e.g., by adding the acid/base in one dose, the formation of small crystals of anhydrous guanine, having an average size of about 0.01-10 μm, is induced. In another embodiment, small crystals having an average size of about 0.1-5 μm, are formed. In another embodiment, small crystals having an average size of about 1-10 μm, are formed. In another embodiment, small crystals having an average size of about 0.1-1 μm, are formed. In another embodiment, small crystals having an average size of less than 0.05-5 μm provide a whiteness with high coverage (non-interferential particles). In some embodiments, there is an overlap in the small and large crystal sizes because there isn't a clear cut in their optical properties; larger plate-like particles will have a tendency to cause interference and therefore pearlescence, small particles will have a tendency to produce whiteness. According to some embodiments, the low concentration of guanine and the additional material and/or the slow adjustment of pH, e.g., dropwise, induces fewer nucleation events and therefore, large co-crystals, having an average size of about 5-1000 μm of the co-crystal. In another embodiment, large co-crystals having an average size of about 5-50 μm, are formed. In another embodiment, large crystals having an average size of about 10-100 μm, are formed. In another embodiment, large crystals having an average size of about 100-1000 μm, are formed. In another embodiment, large crystals having an average size of above 5-70 μm provide a strong pearlescent effect. According to some embodiments, at high concentration of guanine and additional material, between about 0.1-0.2M, and when the pH is rapidly adjusted, e.g., by adding the acid/base in one dose, the formation of small crystals of co-crystals, having an average size of about 0.01-10 μm, is induced. In another embodiment, small crystals having an average size of about 0.1-5 μm, are formed. In another embodiment, small crystals having an average size of about 1-10 μm, are formed. In another embodiment, small crystals having an average size of about 0.1-1 μm, are formed. In another embodiment, small crystals having an average size of less than 0.05-5 μm provide a whiteness with high coverage (non-interferential particles). In some embodiments, there is an overlap in the small and large crystal sizes because there isn't a clear cut in their optical properties; larger plate-like particles will have a tendency to cause interference and therefore pearlescence, small particles will have a tendency to produce whiteness. According to some embodiments, the acid is any appropriate acid in an aqueous solution, such as HCl, H2SO4, H3PO4, HNO3, C6H8O7, H2CO3, H3BO3, or any combination thereof. It is noted that the concentrations of the acids may be determined by the required pH, and may be dependent on the Ka of the particular acid used, such that the stronger the acid, the lower the concentration thereof used. According to some embodiments, the pH of the acidic solution is between about 0-3. According to some embodiments, the pH of the acidic solution is between about 0-1. According to some embodiments, the pH of the acidic solution is between about 1-2. According to some embodiments, the pH of the acidic solution is between about 2-3. According to some embodiments, the pH of the acidic solution is between about 0.5-1.5. According to some embodiments, the pH of the acidic solution is about 1. According to some embodiments, the base is any appropriate base in aqueous solution, such as NaOH, NaHCO3, KOH, NH4OH, Ca(OH)2, or any combination thereof. It is noted that the concentrations of the bases may be determined by the required pH, and may be dependent on the Kb of the particular base used, such that the stronger the base, the lower the concentration thereof used. According to some embodiments, the pH of the basic solution is between about 12-14. According to some embodiments, the pH of the basic solution is between about 12-13. According to some embodiments, the pH of the basic solution is between about 13-14. According to some embodiments, the pH of the basic solution is between about 11.5-12.5. Accenting to some embodiments, the pH of the basic solution is about 12. According to some embodiments, the basic or acidic aqueous solution is maintained at a temperature of about 4-60° C. 10 min to 1 hour. According to some embodiments, the basic or acidic aqueous solution is maintained at a temperature of about 4-10° C. for a predetermined period of time. According to some embodiments, the basic or acidic aqueous solution is maintained at a temperature of about 10-20° C. for a predetermined period of time. According to some embodiments, the basic or acidic aqueous solution is maintained at a temperature of about 20-30° C. for a predetermined period of time. According to some embodiments, the basic or acidic aqueous solution is maintained at a temperature of about 30-40° C. for a predetermined period of time. According to some embodiments, the basic or acidic aqueous solution is maintained at a temperature of about 40-50° C. for a predetermined period of time. According to some embodiments, the basic or acidic aqueous solution is maintained at a temperature of about 50-60° C. for a predetermined period of time. According to some embodiments, the basic or acidic aqueous solution is maintained at a temperature of about 25° C. for a predetermined period of time. As detailed herein, after maintaining the basic or acidic aqueous solution at a predetermined temperature for a predetermined period of time, the basic or acidic aqueous solution is filtered to provide a filtrate. According to some embodiments, the basic or acidic aqueous solution is filtered using a polyvinylidene difluoride filter or any other appropriate filter that is resistant to high alkalinity and high acidity, e.g., a polytetrafluoroethylene (PTFE) filter. According to some embodiments, an acid or a base are added to the filtrate in order to ensure that the guanine and the additional material are dissolved. According to some embodiments, about 0.01 ml of a 0.1M basic or acidic solution, e.g., NaOH, NaHCO3, KOH, NH4OH, Ca(OH)2, or any combination thereof, or HCl, H2SO4, H3PO4, HNO3, C6H8O7, H2CO3, H3BO3, or any combination thereof, solution, respectively, are added to ensure dissolution. According to some embodiments, an acid or a base are added to the filtrate in order to ensure that the guanine is dissolved. According to some embodiments, about 0.01 ml of a 0.1M basic or acidic solution, e.g., NaOH, NaHCO3, KOH, NH4OH, Ca(OH)2, or any combination thereof, or HCl, H2SO4, H3PO4, HNO3, C6H8O7, H2CO3, H3BO3, or any combination thereof, solution, respectively, are added to ensure dissolution. According to some embodiments, the co-crystallization is induced by adjusting the pH of the filtrate, thereby providing a co-crystal suspension. According to some embodiments, the pH is adjusted by adding a base, e.g., NaOH, NaHCO3, KOH, NH4OH, Ca(OH)2, or any combination thereof, or an acid, e.g., HCl, to the solution at a predefined rate over a predefined period of time. According to some embodiments, the crystallization is induced by adjusting the pH of the filtrate, thereby providing a crystal suspension. According to some embodiments, the pH is adjusted by adding a base, e.g., NaOH, NaHCO3, KOH, NH4OH, Ca(OH)2, or any combination thereof, or an acid, e.g., HCl, the solution at a predefined rate over a predefined period of time. According to some embodiments the pH of the added base is about 12-14. According to some embodiments the pH of the added base is about 12-13. According to some embodiments the pH of the added base is about 13-14. According to some embodiments, the pH of the added acid is about 0-3. According to some embodiments, the pH of the added acid is about 0-1. According to some embodiments, the pH of the added acid is about 1-2. According to some embodiments, the pH of the added acid is about 2-3. According to some embodiments, the pH is adjusted by adding an acid or a base dropwise. According to some embodiments, the base or acid are added to the filtrate over a predefined period of time of about 1-20 minutes. According to some embodiments, the base or acid are added to the filtrate over a predefined period of time of about 1-5 minutes. According to some embodiments, the base or acid are added to the filtrate over a predefined period of time of about 5-10 minutes. According to some embodiments, the base or acid are added to the filtrate over a predefined period of time of about 10-15 minutes. According to some embodiments, the base or acid are added to the filtrate over a predefined period of time of about 15-20 minutes. According to some embodiments, the pH is adjusted by adding a predetermined amount of an acid or a base in one dose. In another embodiment, the pH of the reaction mixture is adjusted by evaporating a volatile material (for example ammonia, if used as a base). Any change in the concentration of the materials in the reaction mixture, affects the pH of the reaction mixture. According to some embodiments, the pH of the filtrate is adjusted until a predefined pH value is obtained. According to some embodiments, the pH of the filtrate is monitored throughout the adjustment of the pH using a pH meter. According to some embodiments, the predefined pH value is about 1-13. According to some embodiments, the predefined pH value is about 1-2. According to some embodiments, the predefined pH value is about 2-3. According to some embodiments, the predefined pH value is about 3-4. According to some embodiments, the predefined pH value is about 4-5. According to some embodiments, the predefined pH value is about 5-6. According to some embodiments, the predefined pH value is about 6-7. According to some embodiments, the predefined pH value is about 7-8. According to some embodiments, the predefined pH value is about 8-9. According to some embodiments, the predefined pH value is about 9-10. According to some embodiments, the predefined pH value is about 10-11. According to some embodiments, the predefined pH value is about 11-12. According to some embodiments, the predefined pH value is about 12-13. According to some embodiments, the predefined pH value is about 10.5-11.5. According to some embodiments, the predefined pH value is about 11. As detailed herein, the pH adjustment provides a co-crystal suspension. The co-crystal suspension may be allowed to mature for a predefined period of time. According to some embodiments, the co-crystal suspension is allowed to mature for about 1 minute to 1 week. According to some embodiments, the co-crystal suspension is allowed to mature for about 0.2-24 hours. According to some embodiments, the co-crystal suspension is allowed to mature for about 0.2-1 hours. According to some embodiments, the co-crystal suspension is allows to mature for about 1-6 hours. According to some embodiments, the co-crystal suspension is allowed to mature for about 6-12 hours. According to some embodiments, the co-crystal suspension is allowed to mature for about 12-24 hours. Once matured, the co-crystals may be collected from the crystal suspension by any appropriate means. According to some embodiments, a shorter maturing time provides smaller crystals than a longer maturing time, as long as no other parameters are changed. As detailed herein, the pH adjustment provides a crystal suspension. The crystal suspension may be allowed to mature for a predefined period of time. According to some embodiments, the crystal suspension is allowed to mature for about 1 minute to 1 week. According to some embodiments, the crystal suspension is allowed to mature for about 0.2-24 hours. According to some embodiments, the crystal suspension is allowed to mature for about 0.2-1 hours. According to some embodiments, the crystal suspension is allowed to mature for about 1-6 hours. According to some embodiments, the crystal suspension is allowed to mature for about 6-12 hours. According to some embodiments, the crystal suspension is allowed to mature for about 12-24 hours. Once matured, the crystals may be collected from the crystal suspension by any appropriate means. According to some embodiments, a shorter maturing time provides smaller crystals than a longer maturing time, as long as no other parameters are changed. Further embodiments of the invention are directed to a temperature controlled process for preparing synthetic co-crystals of anhydrous guanine and at least one additional material, wherein said process comprises the steps of:preparing a basic or acidic aqueous solution of guanine and at least one additional material;heating the basic or acidic aqueous solution to a predetermined temperature for the predetermined length of time;inducing co-crystallization by cooling the basic or acidic aqueous solution at a predetermined rate until reaching a predetermined temperature thereby providing a synthetic co-crystal suspension comprising crystals;allowing the synthetic co-crystal suspension to mature over a predetermined period of time; andcollecting the synthetic co-crystals from the crystal suspension. Further embodiments of the invention are directed to a temperature controlled process for preparing anhydrous guanine, wherein said process comprises the steps of:preparing a basic or acidic aqueous solution of guanine;heating the basic or acidic aqueous solution to a predetermined temperature for the predetermined length of time;inducing crystallization by cooling the basic or acidic aqueous solution at a predetermined rate until reaching a predetermined temperature thereby providing a suspension comprising anhydrous guanine crystals;allowing the anhydrous guanine suspension to mature over a predetermined period of time; andcollecting the anhydrous guanine from the crystal suspension. In some embodiments, the anhydrous guanine prepared by the temperature controlled process of this invention is a crystalline anhydrous guanine. It is noted that since the changes in the solubility of the guanine and the additional material obtained by changing the temperature in the temperature controlled process for preparing co crystals are smaller than the changes obtained by changing the pH, the concentration ranges in the temperature controlled process may be smaller. It is noted that since the changes in the solubility of the guanine obtained by changing the temperature in the temperature controlled process for preparing anhydrous guanine crystals are smaller than the changes obtained by changing the pH, the concentration ranges in the temperature controlled process may be smaller. According to some embodiments, the acid is any appropriate acid in an aqueous solution, such as HCl, H2SO4, H3PO4, HNO3, C6H8O7, H2CO3, H3BO3, or any combination thereof. It is noted that the concentrations of the acids may be determined by the required pH, and may be dependent on the Ka of the particular acid used, such that the stronger the acid, the lower the concentration thereof used. According to some embodiments, the pH of the acidic solution is between about 2-4. According to some embodiments, the pH of the acidic solution is between about 2-3. According to some embodiments, the pH of the acidic solution is between about 3-4. According to some embodiments, the pH of the acidic solution is between about 2.5-3.5. According to some embodiments, the pH of the acidic solution is about 3. According to some embodiments, the base is any appropriate base in aqueous solution, such as NaOH, NaHCO3, KOH, NH4OH, Ca(OH)2, or any combination thereof. It is noted that the concentrations of the bases may be determined by the required pH, and may be dependent on the Kb of the particular base used, such that the stronger the base, the lower the concentration thereof used. According to some embodiments, the pH of the basic solution is between about 11-14. According to some embodiments, the pH of the basic solution is between about 11-12. According to some embodiments, the pH of the basic solution is between about 12-13. According to some embodiments, the pH of the basic solution is between about 13-14. According to some embodiments, the pH of the basic solution is between about 11.5-12.5. According to some embodiments, the pH of the basic solution is about 12. According to some embodiments, the basic or acidic aqueous solution is heated to a temperature of between about 50-95° C. According to some embodiments, the basic or acidic aqueous solution is heated to a temperature of between about 50-60° C. According to some embodiments, the basic or acidic aqueous solution is heated to a temperature of between about 60-70° C. According to some embodiments, the basic or acidic aqueous solution is heated to a temperature of between about 70-80° C. According to some embodiments, the basic or acidic aqueous solution is heated to a temperature of between about 80-90° C. According to some embodiments, the basic or acidic aqueous solution is heated to a temperature of between about 90-95° C. According to some embodiments, co-crystallization of anhydrous guanine and at least one additional material is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 0.1-5.0 degrees/minute. According to some embodiments, co-crystallization of anhydrous guanine and at least one additional material is induced by cooling the basic or acidic aqueous solution at a predetermined rate of lie tween about 0.1-1.0 degrees/minute. According to some embodiments, co-crystallization of anhydrous guanine and at least one additional material is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 1.0-2.0 degrees/minute. According to some embodiments, co-crystallization of anhydrous guanine and at least one additional material is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 2.0-3.0 degrees/minute. According to some embodiments, co-crystallization of anhydrous guanine and at least one additional material is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 3.0-4.0 degrees/minute. According to some embodiments, co-crystallization of anhydrous guanine and at least one additional material is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 4.0-5.0 degrees/minute. According to some embodiments, crystallization of anhydrous guanine is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 0.1-5.0 degrees/minute. According to some embodiments, crystallization of anhydrous guanine is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 0.1-1.0 degrees/minute. According to some embodiments, crystallization of anhydrous guanine is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 1.0-2.0 degrees/minute. According to some embodiments, crystallization of anhydrous guanine is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 2.0-3.0 degrees/minute. According to some embodiments, crystallization of anhydrous guanine is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 3.0-4.0 degrees/minute. According to some embodiments, crystallization of anhydrous guanine is induced by cooling the basic or acidic aqueous solution at a predetermined rate of between about 4.0-5.0 degrees/minute. According to some embodiments, the basic or acidic aqueous solution is cooled to a predetermined temperature of about 4-20° C. According to some embodiments, the basic or acidic aqueous solution is cooled to a predetermined temperature of about 4-10° C. According to some embodiments, the basic or acidic aqueous solution is cooled to a predetermined temperature of about 10-15° C. According to some embodiments, the basic or acidic aqueous solution is cooled to a predetermined temperature of about 15-20° C. Embodiments of the invention are further directed to the preparation of various guanine phases, including anhydrous guanine and guanine monohydrate, as well as transforming guanine monohydrate to anhydrous guanine. As shown by the X-ray powder diffraction results presented inFIGS.2A-2C, the elongated acicular crystals are guanine monohydrate crystals (SeeFIG.2A, presenting the X-ray powder diffraction of the phase obtained at pH=2 and SEM pictures inFIGS.1A and1B. The phase presented inFIG.2Awas obtained by filtration of the monohydrate about 10 minutes after the crystals were obtained, such that the monohydrate would not be transformed into the anhydrous phase; however, the spectrum itself was obtained after 24 hours), while the crystals with prismatic bulky morphology are α and β polymorphs of anhydrous guanine (seeFIG.2B, presenting the X-ray powder diffraction of the phase obtained at pH=11, and SEM pictures inFIGS.1C and1D. The phase presented inFIG.2Bwas obtained by filtration of the anhydrous form about 16 hours after the crystals were obtained; however, the spectrum itself was obtained after 24 hours). It is noted that the differences in the filtration times of the monohydrate and the anhydrous forms, as presented inFIGS.2A and2B, respectively, stem from the differences in the rate of transformation from the monohydrate to the anhydrous form under different pH conditions.FIG.2Cpresents the evolution in time of the anhydrous guanine polymorphs in suspension, followed through the intensity of the (011) and (002) diffraction peaks in the α polymorph (solid lines) and in the β polymorph (dashed lines). Initially the suspension consists of the pure β form, which in time transforms into the α form. FTIR spectrum of guanine monohydrate is distinctly different from the spectrum of anhydrous guanine (FIG.4A). Most evident differences are: i) The broad peaks at 3420 and 3200 cm−1and the peak at 1596 cm−1, corresponding to the water stretching modes υ1 and υ3 and the bending mode υ2 respectively, which are present in the monohydrate but not in the anhydrous phase; ii) The C═O and NH2stretching vibrations appear as multiple peaks between 1633-1705 cm−1in the monohydrate phase, whereas they appear as two resolved peaks at 1695 and 1672 cm−1in the anhydrous phase. The Raman spectrum of guanine monohydrate is also distinctly different from the spectrum of anhydrous guanine (FIG.4B). Several vibrations are shifted: specifically, the C═O peak at 1675 cm−1in the anhydrous phase shifts to 1702 cm−1in the guanine monohydrate phase. According to some embodiments, when the solution is highly basic, e.g., the pH is approximately 14, disodium guanine heptahydrate salt is obtained (FIGS.7A-7C). According to some embodiments, guanine monohydrate is obtained from highly acidic solutions, e.g., when the pH is between about 1-3. According to some embodiments, when the pH is between about 4-6, a mixture of guanine monohydrate and anhydrous guanine is obtained. According to some embodiments, when the pH is between about 7-13 mainly anhydrous guanine is formed. According to some embodiments, the kinetically favored polymorph of anhydrous guanine is the β form, which in water suspension transforms with time into the α form (FIG.2C), wherein the higher the pH the slower the transformation is (FIG.6). It is noted that the transformation requires dissolution-reprecipitation, as demonstrated by the fact that material does not transform when kept dry. As noted herein, the high refractive index of guanine is provided by large, plate crystals, and therefore, the control of the crystal size may be essential. According to some embodiments, when performing pH induced crystallization, the interplay between the initial guanine concentration, final guanine concentration, initial pH, final pH and the rate of lowering the pH may allow substantial control over the crystallization process as well as the crystal size. According to some embodiments, the pH is adjusted slowly, e.g., dropwise. According to some embodiments, the concentration of the guanine is relatively low, e.g., between about 0.003-0.2M. According to some embodiments, the concentration of the guanine is about 0.013M. Possibly, the low concentration of guanine and/or the slow adjustment of pH induces fewer nucleation events and therefore, large crystals, having an average size between about 5-1000 μm, are formed. According to some embodiments, at high concentration of guanine, between about 0.1-0.2M, and when the pH is rapidly adjusted, e.g., by adding the acid/base in one dose, the formation of small crystals, having an average size between about 0.01-10 μm, is induced. Further embodiments of the invention are directed to a temperature controlled process for preparing crystalline anhydrous guanine, wherein said process comprises the steps of:heating a crystalline powder of guanine monohydrate to a temperature of between 90-250° C.; andcollecting the crystalline anhydrous guanine. According to some embodiments, crystalline anhydrous guanine is prepared by heating crystalline powder of guanine monohydrate. In another embodiment, without being bound by any mechanism or theory, it is suggested that the heating may remove the water molecules from the guanine monohydrate and subsequently induce phase transformation into anhydrous guanine. According to some embodiments, guanine monohydrate is heated to about 90-100° C., resulting in the deposition of aggregates of polycrystalline or crystalline anhydrous guanine, having acicular morphology. In another embodiment, guanine monohydrate is heated to about 100-150° C. In another embodiment, guanine monohydrate is heated to about 150-200° C. In another embodiment, guanine monohydrate is heated to about 200-250° C.FIG.3Apresents the TGA spectra, showing the transformation of guanine monohydrate to anhydrous guanine upon heating.FIG.3Bpresents the X-ray powder diffraction of the guanine before and after transformation from the monohydrate to the anhydrous form after heating the sample to 250° C. for 10 minutes.FIG.3Cpresents the FTIR spectra of the guanine before and after transformation from the monohydrate to the anhydrous form after heating the sample to 250° C. for 10 minutes. It is noted that although the samples presented inFIGS.3B and3Cwere heated to 250° C. it is possible, as detailed herein, to heat such samples to about 95° C. According to some embodiments, if the crystals are kept in the crystal suspension, the transformation from monohydrate to anhydrous crystals may occur at room temperature within a few hours. It is noted that the term “suspension” in this respect is directed to the solution in which the crystals are formed and suspended in. Possibly, the transformation within the suspension occurs through the dissolution of guanine monohydrate and the formation of anhydrous guanine. In acidic conditions the crystals of guanine monohydrate may grow rapidly and may be the first to form in the solution, while the crystals of anhydrous guanine take much longer to form. If the suspension is not filtered and dried, anhydrous crystals may continue to grow at the expense of the dissolving guanine monohydrate crystals. A suspension which initially contains almost exclusively guanine monohydrate crystals may become a mixture of both phases within 10 minutes and may contain almost only anhydrous guanine within a few hours, wherein the transformation rate is dependent, e.g., on the concentration of the solution as well as the pH thereof. For example, when the pH is highly acidic, e.g., pH=1-2, the transformation to the anhydrous form may be at a lower rate than when using the same concentrations at a higher pH. However, when stored dry at ambient conditions, guanine monohydrate is stable for at least several months and no solid to solid transformation occurs (FIG.5). In one embodiment, the process for the preparation of the co-crystals of this invention and the process for the preparation of the anhydrous guanine are carried out without the use of surfactant. Further embodiments of the invention are directed to the synthetic co-crystal and anhydrous guanine prepared according to the process of the invention for use in paints including, but not limited to, wall paints and car paints, coatings, including, but not limited to, plastic coatings, glass coatings, ceramic coatings, and hydrophobic coating, printing inks, plastics, cosmetic formulations including, but not limited to, nail varnish, lipstick, mascara, and eyeliner, food products, paper, agricultural products, and medicaments. Additional embodiments of the invention are directed to paints including, but not limited to, wall paints and car paints, coatings, including, but not limited to, plastic coatings, glass coatings, ceramic coatings, and hydrophobic coating, printing inks, plastics, cosmetic formulations including, but not limited to, nail varnish, lipstick, mascara, and eyeliner, food products, paper, agricultural products, and medicaments comprising the synthetic co-crystal of the invention and the anhydrous quinine prepared according to the process of this invention. The plate like co-crystals of this invention provide both pearlescent and/or whiteness. Using larger particles (preferably larger than 10 μm) will provide a stronger pearlescent effect and using smaller particles (smaller than 10 μm) will provide a whiteness with high coverage. The cosmetic compositions of this invention comprising the synthetic co-crystals of this invention and the anhydrous guanine prepared according to the process of this invention comprise white natural pigments having high coverage. The coverage measurements (contrast ratio) for different pigments can be calculated as described in WO2014097134 using the following equation: C⁢⁢R=mean⁡(Y⁢⁢black)mean⁡(Y⁢⁢white)·100 wherein Y is defined as luminance as in the CIE 1931 color space model. The greater the percentage of the contrast ratio, the greater the opaqueness of the samples. In one embodiment, the contrast ratio of the synthetic co-crystals of this invention and the anhydrous guanine prepared according to the process of this invention is between 20-60%. Throughout this document, the term “about” is defined to include ±10% of the disclosed value. In order to better understand how the present invention may be carried out, the following examples are provided, demonstrating a process according to the present disclosure. EXAMPLES Example 1 A Process of Preparing Guanine and Hypoxanthine Co-Crystal A solution of 95% guanine and 5% hypoxanthine (mol/mol) was prepared by dissolving 14.3 mg of guanine powder (Sigma Aldrich) together with 0.70 mg of hypoxanthine (Sigma Aldrich) in 10 ml solution of NaOH (0.1M, pH 13), to provide a solution having concentrations of 9.5×10−3M guanine and 5.1×10−4M of hypoxanthine. The solution was mixed for 15 minutes, at a temperature of 25° C. The solution was then filtered using a PVDF filter, and 0.01 ml of 0.1 M NaOH were added to the solution to ensure that all of the powder was dissolved. Next, 0.9 ml of 1M HCl solution was added dropwise while stirring at a rate of 0.5 ml\min. Further, ˜0.8 ml of 0.1 M HCl solution was added dropwise while stirring at a rate of 0.1 ml\min. until the pH of the solution was 11. The solution was matured for 20 hours, after which crystals were isolated therefrom by filtration with a PVDF membrane. The obtained crystals were the typical bulky 20-100 μm prismatic crystals obtained for anhydrous guanine. Example 2 A Process of Preparing Small (˜4 μm×0.5 μm) Anhydrous Guanine and Hypoxanthine Co-Crystals A solution of 25% guanine and 75% hypoxanthine (mol/mol) was prepared by dissolving 37.8 mg of guanine powder (Sigma Aldrich) together with 102.1 mg of hypoxanthine (Sigma Aldrich) in 10 ml solution of NaOH (0.1M, pH 13), to provide a solution having concentrations of 2.50×10−2M guanine and 7.50×10−2M of hypoxanthine. The solution was then mixed for 15 minutes, at a temperature of 25° C. The solutions were then filtered using a PVDF filter and 0.01 ml of 0.1 M NaOH were added to the solution to ensure that all of the powder was dissolved. Next, 0.95 ml of 1M HCl solution was added dropwise while stirring at a rate of 0.5 ml\min. Further, ˜0.8 ml of 0.1 M HCl solution was added dropwise while stirring at a rate of 0.1 ml\min. until the pH of the solution was 10.5. The solution was matured for 20 hours, after which crystals were isolated therefrom by filtration with a PVDF membrane. The obtained co-crystals have plate morphology as shown inFIG.8Cwith a refractive index of ˜1.8.FIG.8Bpresents the X-ray powder diffraction of the obtained co-crystals, showing a noticeable shift from pure anhydrous guanine. Moreover, the co-crystals have a preferred orientation, as is evident from the higher ratio of the intensity of the (100) to the (012) diffraction peaks. It is apparent from comparing the results of Examples 1 and 2 that the formation of guanine-hypoxanthine co-crystals is dependent on the concentration ratio of the guanine and the hypoxanthine. Example 3 A Process of Preparing Large (˜40 μm×5 μm) Anhydrous Guanine and Hypoxanthine Co-Crystals A solution of 25% guanine and 75% hypoxanthine (mol/mol) was prepared by dissolving 9.45 mg of guanine powder (Sigma Aldrich) together with 25.53 mg of hypoxanthine (Sigma Aldrich) in 10 ml solution of NaOH (0.1M, pH 13), to provide a solution having concentrations of 0.625×10−2M guanine and 1.875×10−2M of hypoxanthine. The solution was then mixed for 15 minutes, at a temperature of 25° C. The solutions were then filtered using a PVDF filter and 0.01 ml of 0.1 M NaOH were added to the solution to ensure that all of the powder was dissolved. Next, 0.8 ml of 1M HCl solution was added dropwise while stirring at a rate of 0.5 ml\min. Further, ˜0.5 ml of 0.1 M HCl solution was added dropwise while stirring at a rate of 0.1 ml\min. until the pH of the solution was 11. The solution was matured for 20 hours, after which crystals 40 μm long and 5 μm with excellent pearlescent properties were isolated therefrom by centrifugation. Example 4 A Process of Preparing Guanine and Guanosine Co-Crystal A solution of 90% guanine and 10% guanosine (mol/mol) was prepared by dissolving 378.0 mg of guanine powder (Sigma Aldrich) together with 78.5 mg of guanosine (Sigma Aldrich) in 100 ml solution of NaOH (0.1 M, pH 13), to provide a solution having concentrations of 2.50×10−2M guanine and 2.77×10−3M of guanosine. The solution was then mixed for 15 minutes, at a temperature of 40° C. The solutions were then filtered using a PVDF filler and 0.1 ml of 0.1 M NaOH were added to the solution to ensure that all of the powder was dissolved. Next, 9.0 ml of 1M HCl solution was added dropwise while stirring at a rate of 5.0 ml\min. Further, ˜8.0 ml of 0.1 M HCl solution was added dropwise while stirring at a rate of 0.5 ml\min. until the pH of the solution was 11. The solution was matured for 12 hours, after which crystals were isolated using centrifugation (10 min at 6400 RPM). The obtained co-crystals had plate morphology with a high refractive index of ˜1.8. Example 5 A Process of Preparing Guanine and Xanthine Co-Crystal A solution of 50% guanine and 50% xanthine (mol/mol) was prepared by dissolving 250.0 mg of guanine powder (Sigma Aldrich) together with 243.0 mg of xanthine (Sigma Aldrich) in 100 ml solution of NaOH (0.1 M, pH 13), to provide a solution having concentrations of 1.60×10−2M for both guanine and xanthine. The solution was then mixed for 30 minutes, at a temperature of 25° C. Next, 10.0 ml of 1M HCl solution was added dropwise while stirring at a rate of 5.0 ml\min. Further, ˜7.0 ml of 0.1 M HCl solution was added dropwise while stirring at a rate of 0.5 ml\min. until the pH of the solution was 10. The solution was matured for 8 hours, after which crystals were isolated using centrifugation (10 min at 6400 RPM). The obtained co-crystals had plate morphology with a high refractive index of ˜1.8. Example 6 A Process of Preparing Anhydrous Guanine Crystals A Guanine solution was prepared by dissolving 37.8 mg of guanine powder (Sigma Aldrich) in 10 ml solution of NaOH (0.1M, pH 13). The solution was then mixed for 15 minutes, at a temperature of 25° C. The solution was then filtered using a PVDF filter and 0.01 ml of 0.1 M NaOH were added to the solution to ensure that all of the powder was dissolved. Next, 0.95 ml of 1M HCl solution was added dropwise while stirring at a rate of 0.5 ml\min. Further, ˜0.8 ml of 0.1 M HCl solution was added dropwise while stirring at a rate of 0.1 ml\min. until the pH of the solution was 10.5. The solution was matured for 24 hours, after which crystals were isolated therefrom by centrifugation. The obtained crystals had a bulky prismatic morphology as shown inFIGS.1Cand D.FIG.2Bpresents the X-ray powder diffraction of the obtained crystals, showing a typical pattern of anhydrous guanine crystals. Example 7 A Nail Composition Comprising the Co-Crystal or the Anhydrous Guanine of This Invention 97% by weight of thixotropic nail varnish base 1348 (International Laquers S.A., comprising toluene, ethyl acetate, ethyl acetate, nitrocellulose, tosylamide-formaldehyde resin, dibutyl phthalate, isopropanol, stear-alkonium hectorite, camphor, acrylate copolymer, benzophenone) and 3% by weight of a dispersion of 11% by weight of plates of guanine-hypoxanthine co-crystals (prepared according to the procedure detailed in Example 2) or the anhydrous guanine (prepared according to the process of claim5) in castor oil are mixed by hand using a spatula and subsequently stirred at 1000 rpm for 10 min. A nail varnish having uniform luster is obtained. Example 8 A Lipstick Composition Comprising the Co-Crystal or the Anhydrous Guanine of This Invention A lipstick composition comprising the co-crystal of this invention (prepared according to Examples 1-5) or the anhydrous guanine (prepared according to the process of Example 6) is prepared as described in WO 2014097134. Example 9 A Thermal Process which Provides the Anhydrous Guanine of This Invention A crystalline powder of guanine monohydrate was heated to 250° C.FIG.3Apresents the TGA spectra, showing the transformation of crystalline guanine monohydrate to crystalline anhydrous guanine upon treating.FIG.3Bpresents the X-ray powder diffraction of the crystalline guanine before and after transformation from the monohydrate to the anhydrous form after heating the sample to 250° C. for 10 minutes.FIG.3Cpresents the FTIR spectra of the crystalline guanine before and after transformation from the monohydrate to the anhydrous form after heating the sample to 250° C. for 10 minutes. While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents may occur to those skilled in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.
49,712
11858937
DETAILED DESCRIPTION OF THE INVENTION In one aspect, the present invention relates to pyrazolo-triazine or pyrazolo-pyrimidine compounds which are defined by general formula I whereinX is, independently at each occurrence, selected from CH and N;Q is either absent or independently, at each occurrence, selected from the group consisting of —NH—, —NH(CH2)—, —NH(CH2)2—, —NH(C═O)—, —NHSO2—, —O—, —O(CH2)—, —(C═O)—, —(C═O)NH— and —(C═O)(CH2)—;Y is, independently at each occurrence, selected from the group consisting of C3-C8 cycloalkyl, aryl, heteroaryl, heterocyclyl, and C1-C6 alkyl, wherein C1-C6 alkyl is substituted with one or two of —OR5, —N(R5)R5, aryl, heteroaryl and heterocyclyl,C3-C8 cycloalkyl can be substituted with one or two of R3, R4and —(C═O)R5,heterocyclyl can be substituted with one or two of R3, R4and —(C═O)R5, andaryl or heteroaryl can be substituted with one or two of R3, C1-C6 alkyl, —OR5, —N(R5)R5, —(C═O)R5, halogen, heteroaryl and heterocyclyl,R1is, at each occurrence, independently selected from the group consisting of hydrogen and methyl;R2is, at each occurrence, independently selected from the group consisting of halogen, C1-C6 alkyl, C3-C10 cycloalkyl, —CN, —(C═O)CH3and C1-C3 haloalkyl, any of which is optionally substituted;R3is either absent or independently, at each occurrence, selected from the group consisting of hydrogen, —OR5, halogen, —N(R5)R5, —NH(C═)R5, —(C═O)NH2, aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and C1-C6 alkyl substituted with —OH or —NH2;R4is, independently, at each occurrence, selected from the group consisting of hydrogen, halogen, —OR5, —N(R5)R5, (═O), aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and C1-C6 alkyl substituted with —OH or —NH2′;R5is, at each occurrence, independently selected from the group consisting of hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C3 haloalkyl, heteroaryl, heterocyclyl, heteroaryl substituted with one or two of halogen, —OR11, —N(R11)R11, C1-C6 alkyl and C1-C6 alkyl substituted with —OH, —NH2, heterocyclyl substituted with one or two of halogen, —OR11, —N(R11)R11, C1-C6 alkyl and C1-C6 alkyl substituted with —OH or —NH2;Z is any structure of the following group A; Wherein n=1, 2, or 3; m=1, or 2;R6and R7are, at each occurrence, independently selected from the group consisting of hydrogen, —NH(C═O)R14, —NHR14, —OR14and any structure of the following group B, with the proviso that, when Z is one of R6and R7is not H; wherein o is, independently at each occurrence, selected from 1, 2 and 3;W is any structure of the following group C; L is absent or, at each occurrence, independently selected from the group consisting of —O— and —NH—; wherein n is, independently at each occurrence, selected from 1, 2 and 3;R8, R9and R10are, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —OR5, —CN and C1-C6 alkyl substituted with —OH, —OR5or —NHR5;R11is, at each occurrence, independently selected from the group consisting of hydrogen, C1-C6 alkyl, C3-C10 cycloalkyl and W, as defined above;R12is, at each occurrence, independently selected from hydrogen and W, as defined above; Wherein if R11is W, R12is hydrogen;R13is, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —NH2, —OR5, —CN and W, as defined above;Wherein if R13is W, R12is hydrogen;R14is any structure of group D; R15is, at each occurrence, independently selected from hydrogen and W, as defined above;R16is, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —NH2, —OR5, —CN and W, as defined above;Wherein if R16is W, R12is hydrogen;R17is, at each occurrence, independently selected from the group consisting of hydrogen, C1-C6 alkyl and C1-C3 haloalkyl;R18is, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —NH2, —OR5and —CN;R19is, at each occurrence, independently selected from the group consisting of hydrogen, C1-C6 alkyl, C3-C10 cycloalkyl and W, as defined above;Wherein if R19is W, R15is hydrogen;R20and R21are, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —OR5, heterocyclyl and —CN;R22is, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C3-C10 cycloalkyl, —N(R5)2, —NR19R20, —NR19CH2(CO)NH2, heterocyclyl, —OR5and —CN;or an enantiomer, stereoisomeric form, mixture of enantiomers, diastereomer, mixture of diastereomer, racemate of the above mentioned compounds or a pharmaceutically acceptable salt thereof. The present invention also relates to enantiomers, stereoisomeric forms, mixtures of enantiomers, diastereomers, mixtures of diastereomers, racemates of the above mentioned compounds and to pharmaceutically acceptable salts thereof. In one embodiment of the compounds of general formula I above, the compounds have the general formula Ia whereinX is, independently at each occurrence, selected from CH and N;Y1is, independently at each occurrence, selected from CH, C(OH) and N;Y2is, independently at each occurrence, selected from CH, C(OH) and N;Q is absent or, at each occurrence, independently selected from the group consisting of —NH—, —NH(CH2)—, —NH(C═O)—, —NHSO2—, —O—, —O(CH2)—, —(C═O)— and —(C═O)(CH2)—;R1is, at each occurrence, independently selected from the group consisting of hydrogen and methyl;R2is, at each occurrence, independently selected from the group consisting of halogen, C1-C6 alkyl, C3-C10 cycloalkyl, —CN, —(C═O)CH3and C1-C3 haloalkyl, any of which is optionally substituted;R3is, at each occurrence, independently selected from the group consisting of hydrogen, —OH, halogen, —N(R5)2, —NH(C═O)R5, —(C═O)NH2, C1-C6 alkyl and C1-C6 alkyl substituted with —OH or —NH2;R4is, at each occurrence, absent or independently selected from the group consisting of hydrogen, halogen, —OH, —OR5, —NH2, C1-C6 alkyl and C1-C6 alkyl substituted with —OH or —NH2;R5is, at each occurrence, independently selected from the group consisting of hydrogen, C1-C6 alkyl and C1-C3 haloalkyl;Z is any structure of the following group A; wherein n=1, 2, or 3; m=1, or 2;R6and R7are, at each occurrence, independently selected from the group consisting of hydrogen, —NH(C═O)R14, —NHR14, —OR14and any structure of the following group B, with the proviso that, when Z is one of R6 and R7 is not H; wherein o=1, 2 or 3;W is any structure of the following group C; L is absent or, at each occurrence, independently selected from the group consisting of —O— and —NH—;R8, R9and R10are, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —OR5, —CN and C1-C6 alkyl substituted with —OH, —OR5or —NHR5;R11is, at each occurrence, independently selected from the group consisting of hydrogen, C1-C6 alkyl, C3-C10 cycloalkyl and W, as defined above;R12is, at each occurrence, independently selected from hydrogen and W, as defined above;Wherein if R11is W, R12is hydrogen;R13is, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —NH2, —OR5, —CN and W, as defined above;Wherein if R13is W, R12is hydrogen;R14is any structure of group D; R15is, at each occurrence, independently selected from hydrogen and W, as defined above;R16is, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —NH2, —OR5, —CN and W, as defined above;Wherein if R16is W, R12is hydrogen;R17is, at each occurrence, independently selected from the group consisting of hydrogen, C1-C6 alkyl and C1-C3 haloalkyl;R18is, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —NH2, —OR5and —CN;R19is, at each occurrence, independently selected from the group consisting of hydrogen, C1-C6 alkyl, C3-C10 cycloalkyl and W, as defined above;Wherein if R19is W, R15is hydrogen;R20and R21are, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, —OR5, heterocycle and —CN;R22is, at each occurrence, independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C3-C10 cycloalkyl, —N(R5)2, —NR19R20, heterocycle, —OR5and —CN. In one embodiment of the compounds of general formula I or formula Ia above,at least one of Z, R6, R7, R11, R12, R13, R15, R16and R19is W, as defined above for general formula I, or is a structure containing W, as defined above for general formula I. In one embodiment of the compounds of general formula I or formula Ia aboveexactly one of Z, R6, R7, R11, R12, R13, R15, R16and R19is W, as defined above for general formula I, or is a structure containing W, as defined above for general formula I. In one embodiment, at least one of Z, R6, R7, R11, R12, R13, R15, R16and R19is W, as defined above, or is a structure containing W, as defined above for general formula Ia. In one embodiment, exactly one of Z, R6, R7, R11, R12, R13, R15, R16and R19is W, as defined above, or is a structure containing W, as defined above for general formula Ia. In one embodiment of the compounds of general formula I above,R1is hydrogen and the compound has the general formula II wherein X, Y, Z, R2and Q are as defined above for general formula I. In one embodiment of the compounds of general formula I above, the compounds have the general formula III wherein X, Z, R2and Q are as defined above for general formula I, andYais either absent or independently, at each occurrence, selected from the group consisting of aryl, heteroaryl, heterocyclyl, aryl substituted with one or two of C1-C6 alkyl, —OR5, —N(R5)R5, and halogen, heteroaryl substituted with one or two of C1-C6 alkyl, —OR5, —N(R5)R5and halogen, heterocyclyl substituted with one or two of R23and R24;R23is either absent or independently, at each occurrence, selected from the group consisting of hydrogen, —OR5, halogen, —N(R5)R5, —NH(C═O)R5, —(C═O)NH2, aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and C1-C6 alkyl substituted with —OH or —NH2;R24is, independently, at each occurrence, selected from the group consisting of hydrogen, halogen, —OR5, —N(R5)R5, (═O), aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and C1-C6 alkyl substituted with —OH or —NH2′;wherein R5is as defined in claim1;L′ is either absent or independently, at each occurrence, selected from the group consisting of —NH—, —NH(CH2)—, —NH(C═O)—, —NHSO2—, —O—, —O(CH2)—, —(C═O)—, —(C═O)NH— and —(C═O)(CH2)—;Y1is, independently at each occurrence, selected from CH, C(OH) and N;Y2is, independently at each occurrence, selected from CH, C(OH), O and N;q is, independently at each occurrence, selected from 0, 1 and 2;r is, independently at each occurrence, selected from 0, 1, 2 and 3; In one embodiment of the compounds of general formula I or general formula Ia aboveZ is Z1, and Z1is any structure of the following group E; wherein m is, independently at each occurrence, selected from 1 and 2; andn is as defined above for general formula I;R8, R9, R12and R13are as defined above for general formula I;R6is any structure of group B as defined above for general formula I. In one embodiment of the compounds of general formula Ia above,Z is Z1, and Z1is any structure of the following group E; wherein n=1, 2, or 3; m=1 or 2;R8, R9, R12and R13are as defined above; andR6is any structure of group B as defined above. In one embodiment of the compounds of general formula I or general formula Ia above,Z is p is, independently at each occurrence, selected from 0, 1, 2 and 3;X1is, independently at each occurrence, selected from CR8and N;R6is any structure of group B, as defined above for general formula I; andR8is as defined above for general formula I. In one embodiment of the compounds of general formula Ia above,Z is p is 0, 1, 2 or 3X1is, independently at each occurrence, selected from CR8and N;R6is any structure of group B, as defined above; andR8is as defined above. In one embodiment of the compounds of general formula I or general formula Ia above,Z is wherein R6-R8are as defined above for general formula I. In one embodiment of the compounds of general formula Ia above, Z is wherein R6-R8are as defined above. In one embodiment of the compounds of general formula I above,Z is wherein R6and R10are as defined above for general formula I. In one embodiment of the compounds of general formula I above, the compounds have the general formula IV wherein X, X1, R6, R8and Q are as defined above for general formula I, andX1is as defined above;wherein Ybis any structure of the following group F; R26and R27is either absent or independently, at each occurrence, selected from the group consisting of hydrogen, —OR5, halogen, —N(R5)R5, —NH(C═O)R5, —(C═O)NH2, aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and C1-C6 alkyl substituted with —OH or —NH2;wherein R5is as defined above for general formula I. In one embodiment of the compounds of general formula I above,R2is C1-C6 alkyl. In one embodiment of the compounds of general formula Ia above,R2is C1-C6 alkyl. In one embodiment of the compounds of general formula I above or of general formula Ia above,R6and R7are, at each occurrence, independently selected from —NH(C═O)R14, —NHR14, —OR14, any structure of the following group B′ wherein RNis selected from the group consisting of wherein R5, R8, R12, R16, R17, R18and W are as defined above. In one embodiment of the compounds of general formula I above or of general formula Ia above,W is selected from the group consisting of R20-R22and L being as defined above. In one embodiment of the compounds of general formula Ia above or of general formula III above,Y1is N, Y2is CH, and R3is —N(R5)2, R5being as defined above. In one embodiment, the present invention also relates to pharmaceutically acceptable salts of the compounds according to the present invention, as defined herein. In one embodiment, the compound according to the present invention is a compound selected from structures 1-88, as listed further below in table 7. In a further aspect, the present invention also relates to a pharmaceutical composition comprising a compound according to the present invention as defined herein, as an active ingredient, together with at least one pharmaceutically acceptable carrier, excipient and/or diluent. In one aspect, the present invention also relates to a compound according to the present invention as defined herein, for use as a pharmaceutical or pharmaceutically active agent, wherein said pharmaceutical or pharmaceutically active agent preferably has an inhibitory activity on cyclin-dependent kinase 7 (CDK7). In one aspect, the present invention also relates to a compound according to the present invention, as defined herein, for use in a method of prevention and/or treatment of a disease which is associated with inhibition of apoptosis, abnormal transcriptional activity and/or cell cycle arrest by aberrant activity and/or overexpression of one or several cyclin-dependent kinases (CDKs), in particular cyclin-dependent kinase 7 (CDK7), wherein the disease is selected from proliferative diseases, infectious diseases, including opportunistic diseases, immunological diseases, autoimmune diseases, and inflammatory diseases. In one embodiment, the disease associated with inhibition of apoptosis, abnormal transcriptional activity and/or cell cycle arrest by aberrant activity and/or overexpression of one or several cyclin-dependent kinases (CDKs), in particular cyclin-dependent kinase 7 (CDK7), is a disease associated with, accompanied by, caused by and/or induced by CDK7 dysfunction and/or hyperfunction. In one embodiment, the disease associated with inhibition of apoptosis, abnormal transcriptional activity and/or cell cycle arrest by aberrant activity and/or overexpression of one or several cyclin-dependent kinases (CDKs), in particular cyclin-dependent kinase 7 (CDK7), is a proliferative disease. In one embodiment said proliferative disease is a cancer. In one embodiment said cancer is selected from adenocarcinoma, choroidal melanoma, acute leukemia, acoustic neurinoma, ampullary carcinoma, anal carcinoma, astrocytoma, basal cell carcinoma, pancreatic cancer, Desmoid tumor, bladder cancer, bronchial carcinoma, estrogen dependent and independent breast cancer, Burkitt's lymphoma, corpus cancer, Carcinoma unknown primary tumor (CUP-syndrome), colorectal cancer, small intestine cancer, small intestinal tumors, ovarian cancer, endometrial carcinoma, ependymoma, epithelial cancer types, Ewing's tumors, gastrointestinal tumors, gastric cancer, gallbladder cancer, gall bladder carcinomas, uterine cancer, cervical cancer, cervix, glioblastomas, gynecologic tumors, ear, nose and throat tumors, hematologic tumor, hairy cell leukemia, urethral cancer, skin cancer, skin testis cancer, brain tumors (gliomas), brain metastases, testicle cancer, hypophysis tumor, carcinoids, Kaposi's sarcoma, laryngeal cancer, germ cell tumor, bone cancer, colorectal carcinoma, head and neck tumors (tumors of the ear, nose and throat area), colon carcinoma, craniopharyngiomas, oral cancer (cancer in the mouth area and on lips), cancer of the central nervous system, liver cancer, liver metastases, leukemia, eyelid tumor, lung cancer, lymphomas, stomach cancer, malignant melanoma, malignant neoplasia, malignant tumors gastrointestinal tract, breast carcinoma, rectal cancer, medulloblastomas, melanoma, meningiomas, Hodgkin's/Non-Hodgkin's lymphoma, mycosis fungoides, nasal cancer, neurinoma, neuroblastoma, kidney cancer, renal cell carcinomas, oligodendroglioma, esophageal carcinoma, osteolytic carcinomas and osteoplastic carcinomas, osteosarcomas, ovarian carcinoma, pancreatic carcinoma, penile cancer, plasmacytoma, prostate cancer, pharyngeal cancer, rectal carcinoma, retinoblastoma, vaginal cancer, thyroid carcinoma, esophageal cancer, T-cell lymphoma, thymoma, tube carcinoma, eye tumors, urethral cancer, urologic tumors, urothelial carcinoma, vulva cancer, wart appearance, soft tissue tumors, soft tissue sarcoma, Nephroblastoma, cervical carcinoma, tongue cancer, invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, lobular carcinoma in situ, small-cell lung carcinoma, non-small-cell lung carcinoma, bronchial adenoma, pleuropulmonary blastoma, mesothelioma, brain stem glioma, hypothalamic glioma, cerebellar astrocytoma, cerebral astrocytoma, neuroectodermal tumor, pineal tumors, sarcoma of the uterus, salivary gland cancers, anal gland adenocarcinomas, mast cell tumors, pelvis tumor, ureter tumor, hereditary papillary renal cancers, sporadic papillary renal cancers, intraocular melanoma, hepatocellular carcinoma, cholangiocarcinoma, mixed hepatocellular cholangiocarcinoma, squamous cell carcinoma, malignant melanoma, Merkel cell skin cancer, non-melanoma skin cancer, hypopharyngeal cancer, nasopharyngeal cancer, oropharyngeal cancer, oral cavity cancer, squamous cell cancer, oral melanoma, AIDS-related lymphoma, cutaneous T-cell lymphoma, lymphoma of the central nervous system, malignant fibrous histiocytoma, lymph sarcoma, rhabdomyo sarcoma, malignant histiocytosis, fibroblastic sarcoma, hemangio sarcoma, hemangiopericytoma, leiomyosarcoma (LMS), canine mammary carcinoma, and feline mammary carcinoma. In one embodiment, said infectious disease, including opportunistic diseases, is selected from AIDS, Adenovirus Infection, Alveolar Hydatid Disease (AHD), Amoebiasis, Angiostrongyliasis, Anisakiasis, Anthrax, Babesiosis, Balantidiasis,BaylisascarisInfection,Bilharzia(Schistosomiasis),Blastocystis hominisInfection, Lyme Borreliosis, Botulism, Brainerd Diarrhea, Brucellosis, Bovine Spongiform Encephalopathy (BSE), Candidiasis, Capillariasis, Chronic Fatigue Syndrome (CFS), Chagas Disease, Chickenpox,Chlamydia pneumoniaeInfection, Cholera, Chronic Fatigue Syndrome, Creutzfeldt-Jakob Disease (CJD), Clonorchiasis, Cutaneous Larva migrans (CLM), Coccidioidomycosis, Conjunctivitis, Coxsackievirus A16 (Cox A16), Cryptococcal disease, Cryptosporidiosis, West Nile fever, Cyclosporiasis, Neurocysticercosis, Cytomegalovirus Infection, Dengue Fever,Dipylidium caninumInfection, Ebola Hemorrhagic Fever (EHF), Alveolar Echinococcosis (AE), Encephalitis,Entamoeba coliInfection,Entamoeba disparInfection,Entamoeba hartmanniInfection,Entamoeba poleckiInfection, Pinworm Infection, Enterovirus Infection (Polio/Non-Polio), Epstein Barr Virus Infection,Escherichia coliInfection, Foodborne Infection, Aphthae epizooticae, Fungal Dermatitis, Fungal Infections, Gastroenteritis, Group A streptococcal Disease, Group B streptococcal Disease, Hansen's Disease (Leprosy), Hantavirus Pulmonary Syndrome, Head Lice Infestation (Pediculosis),Helicobacter pyloriInfection, Hematologic Disease, Hendra Virus Infection, Hepatitis (HCV, HBV), Herpes Zoster (Shingles), HIV Infection, Human Ehrlichiosis, Human Parainfluenza Virus Infection, Influenza, Isosporiasis, Lassa Fever, Leishmaniasis, Visceral leishmaniasis (VL), Malaria, Marburg Hemorrhagic Fever, Measles, Meningitis,Mycobacterium aviumComplex (MAC) Infection,NaegleriaInfection, Nosocomial Infections, Nonpathogenic Intestinal Amebae Infection, Onchocerciasis, Opisthorchiasis, Papilloma virus Infection, Parvovirus Infection, Plague,PneumocystisPneumonia (PCP), Polyomavirus Infection, Q Fever, Rabies, Respiratory Syncytial Virus (RSV) Infection, Rheumatic Fever, Rift Valley Fever, Rotavirus Infection, Roundworms Infection,Salmonellosis, Scabies, Shigellosis, Shingles, Sleeping Sickness, Smallpox, Streptococcal Infection, Tapeworm Infection, Tetanus, Toxic Shock Syndrome, Tuberculosis, duodenum,Vibrio parahaemolyticusInfection,Vibriosepticemia, Viral Hemorrhagic Fever, Warts, Waterborne infectious Diseases, Varicella-Zoster Virus infection, Pertussis and Yellow Fever. In one embodiment, the immunological disease and/or autoimmune disease is selected from asthma, diabetes, rheumatic diseases, rejection of transplanted organs and tissues, rhinitis, chronic obstructive pulmonary diseases, osteoporosis, ulcerative colitis, sinusitis, lupus erythematosus, recurrent infections, atopic dermatitis/eczema and occupational allergies, food allergies, drug allergies, severe anaphylactic reactions, anaphylaxis, manifestations of allergic diseases, primary immunodeficiencies, antibody deficiency states, cell mediated immunodeficiencies, severe combined immunodeficiency, DiGeorge syndrome, Hyper IgE syndrome (HIES), Wiskott-Aldrich syndrome (WAS), ataxia-telangiectasia, immune mediated cancers, white cell defects, autoimmune diseases, systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), multiple sclerosis (MS), immune-mediated or Type 1 Diabetes Mellitus, immune mediated glomerulonephritis, scleroderma, pernicious anemia, alopecia, pemphigus, pemphigus vulgaris, myasthenia gravis, inflammatory bowel diseases, Crohn's disease, psoriasis, autoimmune thyroid diseases, Hashimoto's disease, dermatomyositis, Goodpasture syndrome (GPS), myasthenia gravis (MG), Sympathetic ophthalmia, Phakogene Uveitis, chronical aggressive hepatitis, primary biliary cirrhosis, autoimmune hemolytic anemia, and Werlhof's disease. In one embodiment, the inflammatory disease is caused, induced, initiated and/or enhanced by bacteria, viruses, prions, parasites, fungi, and/or caused by irritative, traumatic, metabolic, allergic, autoimmune, or idiopathic agents. In one embodiment, the inflammatory disease is selected from the group comprising or consisting of inflammatory diseases of the central nervous system (CNS), inflammatory rheumatic diseases, inflammatory diseases of blood vessels, inflammatory diseases of the middle ear, inflammatory bowel diseases, inflammatory diseases of the skin, inflammatory disease uveitis, and inflammatory diseases of the larynx. In one embodiment, the inflammatory disease is selected from inflammatory diseases of the central nervous system (CNS), inflammatory rheumatic diseases, inflammatory diseases of blood vessels, inflammatory diseases of the middle ear, inflammatory bowel diseases, inflammatory diseases of the skin, inflammatory disease uveitis, inflammatory diseases of the larynx, wherein preferably said inflammatory diseases are selected from the group comprising abscessation,acanthamoebainfection, acne vulgaris, actinomycosis, acute inflammatory dermatoses, acute laryngeal infections of adults, acute multifocal placoid pigment epitheliopathy, acute (thermal) injury, acute retinal necrosis, acute suppurative otitis media, algal disorders, allergic contact dermatitis, amyloidosis angioedema, ankylosing spondylitis, aspergillosis, atopic dermatitis, pseudorabies, autoantibodies in vasculitis, bacterial disorders, bacterial laryngitis, bacterial meningitis, Behçet's disease (BD), birdshot choroidopathy, Gilchrist's disease, Borna disease, brucellosis, bullous myringitis, bursitis, candidiasis, canine distemper encephalomyelitis, canine distemper encephalomyelitis in immature animals, canine hemorrhagic fever, canine herpes virus encephalomyelitis, cholesteatoma, chronic granulomatous diseases (CGD), chronic inflammatory dermatoses, chronic relapsing encephalomyelitis, chronic suppurative otitis media, Ocular Cicatricial pemphigoid (OCP), common upper respiratory infection, granuloma, Crohn's disease, cryptococcal disease, dermatomyositis, diphtheria, discoid lupus erythematosus (DLE), drug-induced vasculitis, drug or hypersensitivity reaction, encephalitozoonosis, eosinophilic meningoencephalitis, Erythema multiforme (EM), feline leukemia virus, feline immunodeficiency virus, feline infectious peritonitis, feline Polioencephalitis, feline spongiform encephalopathy, fibromyalgia, Fuchs Heterochromic Uveitis, gastroesophageal (laryngopharyngeal) reflux disease, giant cell arteritis, glanders, glaucomatocyclitic crisis, gonorrhea granular myringitis, Granulomatous meningoencephalitis (GME), herpes simplex, histoplasmosis, idiopathic diseases, idiopathic inflammatory disorders, immune and idiopathic disorders, infections of the immunocompromised host, infectious canine hepatitis, inhalation laryngitis, interstitial nephritis, irritant contact dermatitis, juvenile rheumatoid arthritis, Kawasaki's disease, La Crosse virus encephalitis, laryngeal abscess, laryngotracheobronchitis, leishmaniasis, lens-induced uveitis, leprosy, leptospirosis, leukemia, lichen planus, lupus, lymphoma, meningitis, meningoencephalitis in greyhounds, miscellaneous meningitis/meningoencephalitis, microscopic polyangiitis, multifocal choroiditis, multifocal distemper encephalomyelitis in mature animals, multiple sclerosis, Muscle Tension Dysphonia (MTD), mycotic (fungal) diseases, mycotic diseases of the CNS, necrotizing encephalitis, neosporosis, old dog encephalitis, onchocerciasis, parasitic encephalomyelitis, parasitic infections, Pars planitis, parvovirus encephalitis, pediatric laryngitis, pollution and inhalant allergy, polymyositis, post-vaccinal canine distemper encephalitis, prion protein induced diseases, protothecosis, protozoal encephalitis-encephalomyelitis, psoriasis, psoriatic arthritis, pug dog encephalitis, radiation injury, radiation laryngitis, radionecrosis, relapsing polychondritis, Reiter's syndrome, retinitis pigmentosa, retinoblastoma, rheumatoid arthritis, Rickettsial disorders, rocky mountain spotted fever, salmon poisoning disease (SPD), Sarcocystosis, sarcoidosis, schistosomiasis, scleroderma, Rhinoscleroma, serpiginous choroiditis, shaker dog disease, Sjogren's syndrome, spasmodic croup, spirochetal (syphilis) diseases, spongiotic dermatitis, sporotrichosis, steroid responsive meningitis-arteritis, Stevens-Johnson syndrome (SJS, EM major), epiglottitis, sympathetic ophthalmia, Syngamosis, syphilis, systemic vasculitis in sarcoidosis, Takayasu's arteritis, tendinitis (tendonitis), Thromboangiitis obliterans (Buerger Disease), tick-borne encephalitis in dogs, toxic epidermal necrolysis (TEN), toxocariasis, toxoplasmosis, trauma, traumatic laryngitis, trichinosis, trypanosomiasis, tuberculosis, tularemia, ulcerative colitis, urticaria (hives), vasculitis, vasculitis and malignancy, vasculitis and rheumatoid arthritis, vasculitis in the idiopathic inflammatory myopathies, vasculitis of the central nervous system, vasculitis secondary to bacterial, fungal, and parasitic infection, viral disorders, viral laryngitis, vitiligo, vocal abuse, vocal-cord hemorrhage, Vogt-Koyanagi-Harada syndrome (VKH), Wegener's granulomatosis, and Whipple's disease. The present invention also relates to a method of treatment and/or prevention of a disease which is associated with inhibition of apoptosis, abnormal transcriptional activity and/or cell cycle arrest by aberrant activity and/or overexpression of one or several cyclin-dependent kinases (CDKs), in particular cyclin-dependent kinase 7 (CDK7), wherein the disease is selected from proliferative diseases, infectious diseases, including opportunistic diseases, immunological diseases, autoimmune diseases, and inflammatory diseases, wherein said method of treatment and/or prevention comprises administering a compound according to the present invention as defined herein, to a patient in need thereof. In one embodiment, the patient in need thereof is a mammal. In one embodiment, the patient in need thereof is a human being. In another embodiment, the patient in need thereof is a non-human animal. In one embodiment, the disease which is prevented or treated in said method is as defined herein. The present invention also relates to the use of a compound according to the present invention as defined herein in the manufacture of a medicament for the prevention and/or treatment of a disease which is associated with inhibition of apoptosis, abnormal transcriptional activity and/or cell cycle arrest by aberrant activity and/or overexpression of one or several cyclin-dependent kinases (CDKs), in particular cyclin-dependent kinase 7 (CDK7), wherein the disease is selected from proliferative diseases, infectious diseases, including opportunistic diseases, immunological diseases, autoimmune diseases, and inflammatory diseases, as defined herein. Further advantageous features, aspects and details of the invention are evident from the dependent claims, the description, the examples and the drawings. The compounds of the present invention are highly efficient inhibitors of CDK7 threonine/serine kinase and/or its complex, CDK7/MAT1/CycH. The inventive compounds are suitable for the use as a pharmaceutically active agent. The inventive compounds are suitable for the treatment of disorders associated with, accompanied by, caused by and/or induced by CDK7 and its complex, in particular a hyperfunction or dysfunction thereof. The inventive compounds are thus suitable for the treatment of CDK7-associated diseases or disorders and CDK7 complex induced disorders. The inventive compounds are also useful in the manufacture of a medicament or of a pharmaceutical composition for the treatment of disorders associated with, accompanied by, caused by and/or induced by CDK7 and its complex, in particular a hyperfunction or dysfunction thereof. The inventive compounds are further used in the manufacture of a medicament or of a pharmaceutical composition for the treatment and/or prevention of CDK7 and its complex induced disorders. The present inventors have found that in particular in those embodiments of the present invention wherein the compounds according to the present invention contain a W-group, as defined above, they are able to bind covalently to —SH-groups of cysteine residues within cyclin-dependent kinase(s), especially CDK7, thus forming a covalent bond and an adduct between the compound and the kinase and thus inhibiting the kinase(s). This concerns in particular those embodiments wherein at least one of Z, R6, R7, R11, R12, R13, R15, R16and R19is W, as defined above, or is a structure containing W, as defined above. Furthermore it concerns those embodiments wherein exactly one of Z, R6, R7, R11, R12, R13, R15, R16and R19is W, as defined above, or is a structure containing W, as defined above. This is because all W-structures as defined above contain a double or triple bond allowing a reaction with a sulfhydryl group within the kinase and allowing the formation of an adduct between the compound and the kinase. Through the covalent binding of a compound in accordance with the present invention, the kinase is inhibited. The term “exactly one”, as used in this context, means that it is only one (and no more) of the recited groups/residues which is W or a structure containing W, as defined above. The term “optionally substituted” as used herein is meant to indicate that a hydrogen atom where present and attached to a member atom within a group, or several such hydrogen atoms, may be replaced by a suitable group, such as halogen including fluorine, C1-C3alkyl, C1-C3haloalkyl, methylhydroxyl, COOMe, C(O)H, COOH, OMe, or OCF3; The term “alkyl” refers to a monovalent straight, branched or cyclic chain, saturated aliphatic hydrocarbon radical having a number of carbon atoms in the specified range. Thus, for example, “C1-C6alkyl” refers to any of the hexyl alkyl and pentyl alkyl isomers as well as n-, iso-, sec-, and t-butyl, n- and isopropyl, cyclic propyl, ethyl and methyl. The term “alkenyl” refers to a monovalent straight or branched chain aliphatic hydrocarbon radical containing one carbon-carbon double bond and having a number of carbon atoms in the specified range. Thus, for example, “C2-C6alkenyl” refers to all of the hexenyl and pentenyl isomers as well as 1-butenyl, 2-butenyl, 3-butenyl, isobutenyl, 1-propenyl, 2-propenyl, and ethenyl (or vinyl). The term “cycloalkyl”, alone or in combination with any other term, refers to a group, such as optionally substituted or non-substituted cyclic hydrocarbon, having from three to eight carbon atoms, unless otherwise defined. Thus, for example, “C3-C8cycloalkyl” refers to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. The term “haloalkyl” refers to an alkyl group, as defined herein that is substituted with at least one halogen. Examples of straight or branched chained “haloalkyl” groups useful in the present invention include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, and t-butyl substituted independently with one or more halogens. The term “haloalkyl” should be interpreted to include such substituents such as —CHF2, —CF3, —CH2—CH2—F, —CH2—CF3, and the like. The term “heteroalkyl” refers to an alkyl group where one or more carbon atoms have been replaced with a heteroatom, such as, O, N, or S. For example, if the carbon atom of alkyl group which is attached to the parent molecule is replaced with a heteroatom (e.g., O, N, or S) the resulting heteroalkyl groups are, respectively, an alkoxy group (e.g., —OCH3, etc.), an amine (e.g., —NHCH3, —N(CH3)2, etc.), or thioalkyl group (e.g., —SCH3, etc.). If a non-terminal carbon atom of the alkyl group which is not attached to the parent molecule is replaced with a heteroatom (e.g., O, N, or S) and the resulting heteroalkyl groups are, respectively, an alkyl ether (e.g., —CH2CH2—O—CH3, etc.), alkyl amine (e.g., —CH2NHCH3, —CH2N(CH3)2, etc.), or thioalkyl ether (e.g., —CH2—S—CH3). The term “halogen” refers to fluorine, chlorine, bromine, or iodine. The term “phenyl” as used herein is meant to indicate that optionally substituted or non-substituted phenyl group. The term “benzyl” as used herein is meant to indicate that optionally substituted or non-substituted benzyl group. The term “heteroaryl” refers to (i) optionally substituted 5- and 6-membered heteroaromatic rings and (ii) optionally substituted 9- and 10-membered bicyclic, fused ring systems in which at least one ring is aromatic, wherein the heteroaromatic ring or the bicyclic, fused ring system contains from 1 to 4 heteroatoms independently selected from N, O, and S, where each N is optionally in the form of an oxide and each S in a ring which is not aromatic is optionally S(O) or S(O)2. Suitable 5- and 6-membered heteroaromatic rings include, for example, pyridyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thienyl, furanyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isooxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, and thiadiazolyl. Suitable 9- and 10-membered heterobicyclic, fused ring systems include, for example, benzofuranyl, indolyl, indazolyl, naphthyridinyl, isobenzofuranyl, benzopiperidinyl, benzisoxazolyl, benzoxazolyl, chromenyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, isoindolyl, benzodioxolyl, benzofuranyl, imidazo[1,2-a]pyridinyl, benzotriazolyl, dihydroindolyl, dihydroisoindolyl, indazolyl, indolinyl, isoindolinyl, quinoxalinyl, quinazolinyl, 2,3-dihydrobenzo furanyl, and 2,3-dihydrobenzo-1,4-dioxinyl. The term “heterocyclyl” refers to (i) optionally substituted 4- to 8-membered, saturated and unsaturated but non-aromatic monocyclic rings containing at least one carbon atom and from 1 to 4 heteroatoms, (ii) optionally substituted bicyclic ring systems containing from 1 to 6 heteroatoms, and (iii) optionally substituted tricyclic ring systems, wherein each ring in (ii) or (iii) is independent of fused to, or bridged with the other ring or rings and each ring is saturated or unsaturated but nonaromatic, and wherein each heteroatom in (i), (ii), and (iii) is independently selected from N, O, and S, wherein each N is optionally in the form of an oxide and each S is optionally oxidized to S(O) or S(O)2. Suitable 4- to 8-membered saturated heterocyclyls include, for example, azetidinyl, piperidinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, isothiazolidinyl, oxazolidinyl, isoxazolidinyl, pyrrolidinyl, imidazolidinyl, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, pyrazolidinyl, hexahydropyrimidinyl, thiazinanyl, thiazepanyl, azepanyl, diazepanyl, tetrahydropyranyl, tetrahydrothiopyranyl, dioxanyl, and azacyclooctyl. Suitable unsaturated heterocyclic rings include those corresponding to the saturated heterocyclic rings listed in the above sentence in which a single bond is replaced with a double bond. It is understood that the specific rings and ring systems suitable for use in the present invention are not limited to those listed in this and the preceding paragraphs. These rings and ring systems are merely representative. Pharmaceutically Acceptable Salts Examples of pharmaceutically acceptable addition salts include, without limitation, the non-toxic inorganic and organic acid addition salts such as the acetate derived from acetic acid, the aconate derived from aconitic acid, the ascorbate derived from ascorbic acid, the benzenesulfonate derived from benzensulfonic acid, the benzoate derived from benzoic acid, the cinnamate derived from cinnamic acid, the citrate derived from citric acid, the embonate derived from embonic acid, the enantate derived from enanthic acid, the formate derived from formic acid, the fumarate derived from fumaric acid, the glutamate derived from glutamic acid, the glycolate derived from glycolic acid, the hydrochloride derived from hydrochloric acid, the hydrobromide derived from hydrobromic acid, the lactate derived from lactic acid, the maleate derived from maleic acid, the malonate derived from malonic acid, the mandelate derived from mandelic acid, the methanesulfonate derived from methane sulphonic acid, the naphthalene-2-sulphonate derived from naphtalene-2-sulphonic acid, the nitrate derived from nitric acid, the perchlorate derived from perchloric acid, the phosphate derived from phosphoric acid, the phthalate derived from phthalic acid, the salicylate derived from salicylic acid, the sorbate derived from sorbic acid, the stearate derived from stearic acid, the succinate derived from succinic acid, the sulphate derived from sulphuric acid, the tartrate derived from tartaric acid, the toluene-p-sulphonate derived from p-toluene sulphonic acid, and the like. Such salts may be formed by procedures well known and described in the art. Other acids such as oxalic acid, which may not be considered pharmaceutically acceptable, may be useful in the preparation of salts useful as intermediates in obtaining a chemical compound of the invention and its pharmaceutically acceptable acid addition salt. In another embodiment, the compounds of the invention are used in their respective free base form according to the present invention. Metal salts of a chemical compound of the invention include alkali metal salts, such as the sodium salt of a chemical compound of the invention containing a carboxy group. The chemical compounds of the invention may be provided in unsolvated or solvated forms together with a pharmaceutically acceptable solvent(s) such as water, ethanol, and the like. Solvated forms may also include hydrated forms such as the monohydrate, the dihydrate, the hemihydrate, the trihydrate, the tetrahydrate, and the like. In general, solvated forms are considered equivalent to unsolvated forms for the purposes of this invention. Further aspects of the present invention are illustrated and exemplified by the following schemes, examples, tables and procedural descriptions which are given merely to illustrate, not to limit the present invention. The scope of protection for the present invention is merely limited by the appended claims. Tables Reference is now made to tables, wherein Table 1 shows activity data in CDK1, CDK2, CDK5 and CDK7 enzymatic assay for selected compounds of the invention. Inhibition is indicated as IC50with the following key: A=IC50less than 100 nM; B=IC50greater than 100 nM, but less than 1,000 nM; C=IC50greater than 1,000 nM. Also table 1 shows selectivity data in CDK1/CDK7, CDK2/CDK7 and CDK5/CDK7 for selected compounds of the invention. Selectivity is indicated as CDK1/CDK7*, CDK2/CDK7** and CDK5/CDK7*** with the follow key: A=greater than 500 fold; B=less than 500 fold, but greater than 50 fold; C=less than 50 fold. Table 2 shows activity data of cellular H460 viability assay for selected compounds of the invention. Inhibition is indicated as IC50with the following key: A=IC50less than 1 uM; B=IC50greater than 1 uM, but less than 10 uM; C=IC50greater than 10 uM. Table 3 shows activity data of cellular MV4-11 viability assay for selected compounds of the invention. Inhibition is indicated as IC50with the following key: A=IC50less than 1 uM; B=IC50greater than 1 uM, but less than 10 uM; C=IC50greater than 10 uM. Table 4 shows activity data of A2780 viability assay for selected compounds of the invention. Inhibition is indicated as IC50with the following key: A=IC50less than 1 uM; B=IC50greater than 1 uM, but less than 10 uM; C=IC50greater than 10 uM. Table 5 shows activity data of OVCAR-3 viability assay for selected compounds of the invention. Inhibition is indicated as IC50with the following key: A=IC50less than 1 uM; B=IC50greater than 1 uM, but less than 10 uM; C=IC50greater than 10 uM. Table 6 summarizes compounds 1-88 in terms of their structures and corresponding characteristics. EXAMPLES The invention is now further described by reference to the following examples which are intended to illustrate, not to limit the scope of the invention. Example 1: Enzymatic Assay for CDK1, CDK2, CDK5 and CDK7 Enzymatic Binding Assay Protocol for CDK1, CDK2, CDK5 and CDK7 Inhibition activity of the respective compound on CDK kinase under Km value of ATP was tested in FRET based The LANCE® Ultra kinase assay (Perkin Elmer), which uses a ULight™-labeled peptide substrate and an appropriate Europium-labeled anti-phospho-antibody. Test compounds were made with DMSO solutions, and then 4-fold serial dilutions for 8 doses were prepared using automated liquid handler (POD™ 810, Labcyte) and 80 nL/well of diluted compound solutions were added into the 384-well plates (Greiner, Cat #784075). And then 68 nM of ULight-MBP peptide (Perkin Elmer, Cat #TRF0109-M) and 5 ul/well of ATP (Sigma, Cat #A7699) were added to the plate. After 1 min centrifugation at 1000 rpm, purified CDKs/Cyclin complex were added with the following concentrations respectively. 24 uM for CDK1/Cyclin B (Invitrogen, Cat #PR4768C), 22 uM for CDK2/Cyclin A (Invitrogen, Cat #PV6290), 10 uM for CDK5/p25 (Invitrogen, Cat #PR8543B) and 400 uM for CDK7/Cyclin H/MNAT1 (Invitrogen, Cat #PR6749B) were added to each corresponding plate for CDK1, CDK2, CDK5 and CDK7. Incubate at 23° C. for 60 min and then Eu-labeled anti-phospho-Myelin Basic Protein (PE, Cat #TRF0201-M) and EDTA (Invitrogen, Cat #15575038) mixture in Lance Detection Buffer (Perkin Elmer, Cat #CR97100) was added in each well. After additional incubation at 23° C. for 60 min, test articles were measured the fluorescence using Envision leader (Perkin Elmer, USA) [Laser as excitation light; APC 615 nm and Europium 665 as the first and the second emission filter]. Data were analyzed using XL Fit software. Example 2: Cellular H460, MV4-11, A2780 and OVCAR-3 Viability Assay Cell Culture Human T-cell acute lymphoblastic leukemia cell line, MV4-11_(ATCC, Cat #CRL-9591), NSCLC (Non-small cell lung cancer) cell line H460 (ATCC, Cat #HTB-177), A2780 (ECACC, Cat #93112519) and OVCAR-3 (ATCC, Cat #HTB-161) were obtained from ATCC. Cells were grown in RPMI-1640 media (Invitrogen, Cat #22400-089) supplemented with 10% FBS (Invitrogen, Cat #10099141) and 1% penicillin/streptomycin (Invitrogen, Cat #15070063) and cultured at 37° C., 5% CO2in a humidified chamber. All cell lines were routinely tested formycoplasma. Cell H460, MV4-11, A2780 and OVCAR-3 Viability Assay Protocol To effect of the CDK7 inhibitor to inhibit the growth of target cancer cells, viability assay were conducted a 72 hour time period. Briefly, the candidate cell line was plated in 96 well plate at the following density of cells respectively. 1×104cells/well for MV4-11, 5×103for H460 and OVCAR-3, and 1×103for A2780. After 24 hours, the cells were treated with various concentrations of the compound (ranging from 0.0015 uM to 10 uM). DMSO solvent without compound served as a control and final DMSO concentration lest than 0.1%. After 72 hours of incubation at 37° C., 5% CO2incubator, cells were analyzed for the viability using the CellTiter-Glo Luminescent Cell Viability Assay (Promega, Cat #G7570). All viability assays were performed in duplicate and Luminescence was read using an Envision (Perkin Elmer, USA). Data was analyzed using XLfit software. Example 3: Derivatization of the Pyrazolo-Triazine General Scaffold Presented compounds underwent derivation according to the methods outlined below (Scheme 1-37). Resulting derivatives were examined for enzymatic binding and cellular activity (H460, MV4-11, A2780 and OVCAR-3), using the assays described above (Examples 1 and 2) and the results are summarized in Table 1-5. The synthesized compounds 1-88 are shown in Table 6. The method to prepare compounds of formula I-2, II-3 and III-4 were shown in Scheme 1. Route I: Compound G10 can be treated with group A in presence of DIPEA to give compounds I-1. Compounds I-2 can be treated with HBr/AcOH to obtain the compounds of formula I-2. Route II: Compound G10 can be treated with group B in presence of DIPEA to give compounds II-1. Compound II-1 can be treated with group C which is defined claim1in presence of DIPEA and acyl chloride to give compounds II-2. Compounds II-2 can be treated with HBr/AcOH to obtain the compounds of formula II-3. Route III: Compound G10 can be treated with group D in presence of DIPEA to give compounds III-1. Compound III-1 can be treated with TFA to give compounds III-2. Compound III-2 can be treated with group C which was defined claim1in presence of DIPEA and acyl chloride to give compounds III-3. Compounds III-3 can be treated with HBr/AcOH to obtain the compounds of formula III-4. General Schemes of Group A Procedure for Synthesis of A3 To a solution of compound A2 (2.64 g), DIPEA (3.07 g, 23.7 mmol) in DCM (40.0 mL) was added 3-nitrobenzoyl chloride A1 (2.20 g, crude) at 0° C. The reaction mixture was stirred at 10° C. for 16 hours under N2atmosphere. TLC showed a new spot was formed. The reaction solution was washed with water (10 mL), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure. The residue was purified by Combi flash to give compound A3 (1.96 g) as an off-white powder. Procedure for Synthesis of A4 To a solution of compound A3 (1.76 g) in MeOH (40.0 mL) was added Zn powder (3.10 g, 47.40 mmol) and NH4Cl (2.54 g, 47.4 mmol). The reaction mixture was stirred at 10° C. for 1 hour. TLC showed the reaction was completed. The reaction mixture was poured into saturated aqueous NaHCO3solution (20 mL) and extracted with EtOAc (10 mL×2). The combined extract was washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a crude product. The crude product was purified by Combi flash to give compound A4 (875 mg) as a brown gum. Procedure for Synthesis of A6 To a solution of compound A4 (775 mg, 2.27 mmol) and DIPEA (601 mg, 4.65 mmol) in anhydrous THF (10 mL) was added a solution of compound A5 (500 mg, 2.72 mmol) in anhydrous THF (1.0 mL) dropwise at 15° C. After stirred at 15° C. for 10 minutes, dimethylamine (2 M in THF, 7.57 mL) was added. The reaction solution was stirred at 15° C. for 20 minutes. TLC showed the reaction was complete. The reaction mixture was poured into water (10 mL) and then extracted with EtOAc (10 mL×2). The organic layer was washed with water (10 mL) and brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give the crude product as a brown gum. The crude product was purified by Combi flash to give compound A6 (337 mg) as a light brown gum. Procedure for Synthesis of A7 To a solution of compound A6 (168 mg) in DCM (2 mL) was added TFA (500 uL). The reaction solution was stirred at 10° C. for 2 hours. TLC showed the reaction was completed. The mixture was concentrated under reduced pressure. The residue was partitioned between DCM (10 mL) and saturated aqueous NaHCO3solution (10 mL). The organic and aqueous layer was concentrated under reduced pressure to give 130 mg of crude compound A7. The crude product was used directly in the next step without further purification. Procedure for Synthesis of A10 To a solution of compound A8 (2.80 g, 12.5 mmol) in anhydrous DCM (100 mL) was added compound A9 (3.65 g, 18.8 mmol), Cu(OAc)2(3.42 g, 18.8 mmol) and TEA (3.81 g, 37.6 mmol, 5.22 mL). The reaction mixture was stirred at 15° C. for 1 day. TLC showed compound A13 was consumed. The reaction mixture was filtered. The filtrate was washed with water (20 mL) and concentrated under reduced pressure to give a light brown gum. The crude gum was purified by Combi flash to give compound A10 (629 mg) as a white powder. Procedure for Synthesis of A11 To a solution of compound A10 (625 mg, 1.68 mmol) in MeOH (6 mL) was added NaOH (1 M, 3.36 mL). The reaction mixture was stirred at 15° C. for 17 hour. TLC showed the reaction was completed. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in water (5 mL) and neutralized by aqueous HCl solution (1 M, 3.40 mL), then extracted with DCM (10 mL×2). The organic layer was washed with water (10 mL), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure to give compound A11 (483 mg) as a white powder. Procedure for Synthesis of A12 To a solution of compound A11 (383 mg, 1.12 mmol), benzyl alcohol (969 mg, 8.96 mmol, 931.67 uL), TEA (453 mg, 4.48 mmol, 621 uL) in dioxane (10 mL) was added DPPA (339 mg, 1.23 mmol, 267 uL). The reaction solution was heated to reflux for 2 hours. TLC showed the reaction was completed. The reaction mixture was partitioned between water (30 mL) and DCM (30 mL). The organic layer was washed with water (10 mL×2), brine (10 mL), dried over anhydrous Na2SO4, and concentrated under reduced pressure to give a residue. The crude product was purified by Combi flash to give compound A12 (1.20 g, crude) as a colorless oil. The crude product was used directly in the next step without further purification. Procedure for Synthesis of A13 To a solution of compound A12 (1.10 g, crude) in MeOH (50 mL) was added Pd/C (100 mg, 50% wet, 10% Pd). The reaction mixture was degassed under vacuum and purged with H2for 3 times, then stirred at 15° C. for 16 hours under H2atmosphere (15 psi). TLC showed the reaction was completed. The reaction mixture was filtered through a pad of Celite. The filtrate was concentrated under reduced pressure. The residue was purified by Combi flash to give compound A13 (210 mg) as a colorless oil. Procedure for Synthesis of A15 To a solution of compound A13 (140 mg, 0.445 mmol), TEA (64 mg, 0.63 mmol) in DCM (2 mL) was added acryl chloride A14 (61.0 mg, 0.674 mmol) dropwise at 20° C. The reaction solution was stirred at 20° C. for 2 hours under N2atmosphere. TLC showed the reaction was completed. The reaction was quenched with water (5 mL) and extracted with DCM (10 mL×2). The combined extract was washed with water (5 mL), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure to give the crude product as a brown gum. The crude product was purified by Combi flash to give compound A15 (131 mg) as a brown gum. Procedure for Synthesis of A16 The compound A15 (130 mg) was followed the same procedure of A7 to obtain 90 mg of compound A16 as a brown gum. Procedure for Synthesis of A17 To a solution of compound A13 (210 mg, 0.668 mmol) and DIPEA (177 mg, 1.37 mmol) in anhydrous THF (5 mL) was added a solution of compound A5 (147 mg, 0.801 mmol) in anhydrous THF (1.0 mL) dropwise at 15° C. After stirred at 15° C. for 30 minutes, LCMS showed the desired product. The reaction mixture was poured into water (10 mL) and then extracted with DCM (10 mL×2). The organic layer was washed with water (10 mL) and brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give the crude product as a brown gum. The crude product was purified by Combi flash to give compound A17 (183 mg) as a light brown gum. Procedure for Synthesis of A18 The compound A17 (180 mg) was followed the same procedure of A7 to obtain 133 mg of compound A18 as a brown gum. Procedure for Synthesis of 21 To a solution of compound A20 (300 mg, 1.68 mmol) and compound A19 (506 mg, 2.02 mmol) in dioxane (5 mL) and H2O (1 mL) was added Cs2CO3(547 mg, 1.68 mmol), Pd(dppf)Cl2(123 mg, 0.168 mmol) under N2. The resulting mixture was heated at 100° C. and stirred for 1.5 hours to give black suspension. LCMS and TLC showed the reaction was completed. The reaction mixture was quenched by addition H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by combi flash to give compound A21 (240 mg) as a yellow solid. Procedure for Synthesis of A22 The compound A21 (408 mg) was followed the same procedure of A6 to obtain 184 mg of compound A22 as a yellow oil. Procedure for Synthesis of A23 The compound A22 (184 mg) was followed the same procedure of A7 to obtain 160 mg of compound A23 as a yellow oil. General Schemes of Group B Procedure for Synthesis of B3 To a solution of compound B1 (2.48 g, 8.66 mmol), compound B2 (1.00 g, 7.87 mmol), pyridine-2-carboxylic acid (194 mg, 1.57 mmol) and K3PO4(3.34 g, 15.7 mmol) in DMSO (15 mL) was added CuI (150 mg, 0.787 mmol), the mixture was purged with N2for three times and stirred at 90° C. for 17 hours to give a dark solution. LCMS showed the reaction was completed. TLC showed the reaction was completed. The reaction mixture was poured into water (100 mL), extracted with EtOAc (100 mL×2), the combined extracts was washed with brine (30 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash to give compound B3 (860 mg) as light yellow oil. Procedure for Synthesis of B4 To a solution of compound B3 (860 mg, 2.59 mmol) in DCM (7 mL) was added TFA (3 mL), the reaction mixture was stirred at 25° C. under N2for 1 hour to give a brown solution. TLC showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to give compound B4 (800 mg) as brown oil. Procedure for Synthesis of B6 To a suspension of compound B5 (4.00 g, 25.2 mmol) and Cs2CO3(16.4 g, 50.5 mmol) in DMF (50 mL) was added 3-cyanophenol (3.16 g, 26.5 mmol), the mixture was stirred at 60° C. for 17 hours to give a brown suspension. Crude LCMS (RT: 1.384 min) showed the reaction was completed. The mixture was poured into water (400 mL), a lot of white solid appeared, filtered, the filter cake was washed with water (30 mL×3) to give the crude product. The crude product was purified by Combi flash to give 1.00 g of compound B6 as a yellow powder. Procedure for Synthesis of B7 To a solution of compound B6 (850 mg, 3.52 mmol) and Pd/C (170 mg, 10% Pd) in MeOH (10 mL) was added NH3·H2O (1.83 g, 5.21 mmol), the mixture was purged with N2for three times and stirred at 20° C. under H2balloon (15 psi) for 1 hour to give a brown suspension. The crude LCMS showed the reaction was completed. The mixture was filtered, the filtrate was concentrated under reduced pressure to give 700 mg of compound B7 as a yellow gum. Procedure for Synthesis of B10 To a solution of NaH (1.73 g, 43.4 mmol) in DMF (100 mL) was added 7-nitro-2H-indazole B8 (5.19 g, 31.8 mmol) in several portion at 20° C., the resulting mixture was stirred for 1 hour at 20° C., then 2-fluorobenzonitrile B9 (3.50 g, 28.9 mmol) was added to the mixture, the reaction mixture was stirred for another 12 hours at 130° C. to give black suspension. TLC showed the reaction was completed. The reaction mixture was quenched by addition H2O (500 mL) and extracted with EtOAc (500 mL×2). The combined organic layers were washed with brine (100 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by triturated to obtain compound B10 (1.80 g) as a black solid. Procedure for Synthesis of B11 To a solution of compound B10 (200 mg, 0.8 mmol) in MeOH (20 mL) and NH3·H2O (1.00 mL, 25%) was added Raney-Ni (64.8 mg, 0.8 mmol) under N2. The suspension was degassed under vacuum and purged with H2several times. The mixture was stirred under H2(40 psi) at 20° C. for 2 hours to give black suspension. TLC and LCMS showed the reaction was completed. The reaction mixture was filtered by celite pad and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash to obtain compound B11 (50 mg) as a yellow oil. Procedure for Synthesis of B13 To a solution of compound B12 (1.50 g, 9.25 mmol) in MeOH (50 mL) was added Zn (6.05 g, 92.5 mmol) and NH4Cl (4.95 g, 92.5 mmol). The resulting mixture was stirred at 20° C. for 12 hours to give black suspension. TLC showed the reaction was completed, one new spot was formed. The reaction mixture was filtered and concentrated under reduced pressure to give a residue to give 1.80 g compound B13 as a black solid and used in the next step without purification. Procedure for Synthesis of B14 To a solution of crude product compound B13 (1.80 g, 6.81 mmol) in MeOH (50 mL) was added Et3N (689 mg, 6.81 mmol) and Boc2O (2.23 g, 10.2 mmol). The resulting mixture was stirred at 20° C. for 12 hours to give black solution. TLC showed one new spot was formed. The reaction mixture was filtered under reduced pressure to give a residue. The residue was purified by Combi flash to obtain compound B14 (781 mg) as a white solid. Procedure for Synthesis of B16 To a mixture of compound B14 (810 mg, 3.23 mmol), compound B15 (500 mg, 2.15 mmol) in HOAc (20 mL) was added Pd(OAc)2(241 mg, 1.08 mmol), the mixture was stirred at 40-50° C. under O2(15 psi) atmosphere for 12 hours to give a black brown suspension. LCMS (Rt=1.436 min) showed the reaction was completed. AcOH was removed under reduced pressure, and the residue was dissolved in DCM (150 mL) and washed with saturated aqueous NaHCO3(100 mL×3). The organic layer was dried over Na2SO4, filtered, concentrated under reduced pressure to give black brown oil. The mixture was for Combi flash to give compound B16 (380 mg) as a yellow gum. Procedure for Synthesis of B17 To a mixture of compound B16 (380 mg, 0.869 mmol) in DCM (30 mL) was added TFA (8 mL). The mixture was stirred at 15° C. for 0.5 hour to give a yellow mixture. LCMS showed the reaction was completed. The mixture was combined and concentrated under reduced pressure to give compound B17 (350 mg) as yellow oil Procedure for Synthesis of B19 A solution of 2-chloroquinoline B18 (2.00 g, 12.2 mmol) in H2SO4(20 mL) was cooled to 0° C. HNO3(3.55 g, 36.7 mmol) was added dropwise. The reaction solution was stirred at 25° C. for 1 hour to give a black brown solution. TLC showed the reaction was completed. The reaction solution was poured into water (50 mL), neutralized to pH=7-8 with saturated Na2CO3. The resulting mixture was extracted with DCM (200 mL×2). The combined organic layer was washed with brine (200 mL), dried over anhydrous Na2SO4and concentrated under reduced pressure. The residue was purified by Combi Flash to give compound B19 (1.30 g) as a yellow solid. Procedure for Synthesis of B21 To a solution of compound B19 (560 mg, 2.68 mmol) and compound B20 (2.02 g, 8.04 mmol) in dioxane (10 mL)/H2O (3 mL) was added Cs2CO3(1.75 g, 5.36 mmol) and Pd(dppf)Cl2(98.1 mg, 0.134 mmol) under N2atmosphere. The reaction mixture was heated to 100° C. and stirred for 16 hours under N2atmosphere to give a black mixture. TLC showed the reaction was completed. The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (200 mL×2). The combined extracts were dried over anhydrous Na2SO4and concentrated under reduced pressure. The residue was purified by Combi Flash to give compound B21 (1.00 g) as brown solid. Procedure for Synthesis of B22 To a solution of compound B21 (500 mg, 1.32 mmol) and NH4Cl (706 mg, 13.2 mmol) in MeOH (10 mL) was added Zn (863 mg, 13.2 mmol) slowly. The reaction mixture was stirred at 25° C. for 16 hours to give a black brown mixture. TLC showed the reaction was completed. The reaction mixture was diluted with MeOH (50 mL) and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by Combi Flash to give compound B22 (500 mg) as a yellow solid. Procedure for Synthesis of B23 To a solution of compound B22 (500 mg, 1.43 mmol) in DCM (7 mL) was added TFA (3 mL). The reaction solution was stirred at 25° C. for 1 hour to give a red solution. LCMS showed the reaction was completed. The reaction was diluted with DCM (10 mL) and concentrated under reduced pressure to give compound B23 (300 mg) as black brown oil. Procedure for Synthesis of B25 Isoquinolin-6-amine B24 (2.00 g, 13.87 mmol) was dissolved in pyridine (20 mL), and 4-methylbenzenesulfonyl chloride (3.17 g, 16.64 mmol) was added. The reaction mixture was stirred at 20° C. for 12 hours. LCMS showed the reaction was completed. To the reaction mixture was added water (30 mL) under good stirring, the mixture was stirred at 20° C. for 0.5 hour, pale yellow solid precipitated out. The mixture was filtered and the solid was collected and washed with water (5 mL) to give compound B25 (2.2 g) as a yellow solid. Procedure for Synthesis of B26 Compound B25 (2.00 g, 6.70 mmol) was dissolved in CHCl3(30.00 mL). Under ice-cooling (0° C.), m-CPBA (1.71 g, 7.91 mmol) was added thereto, followed by stirring at 20° C. for 12 hours. TLC showed the reaction was completed. The solvent was evaporated and the resulting solid were washed with MTBE (50 mL). The filter cake was collected and dried in high vacuum to give compound B26 (1.96 g) as a light yellow solid. Procedure for Synthesis of B27 To a mixture of compound B26 (4.20 g, 13.4 mmol) in CHCl3(120 mL) was added POCl3(45.1 g, 293.9 mmol). The reaction mixture was heated to 61° C. and stirred for 16 hours to give a black brown solution. TLC showed the reaction was completed. The reaction solution was cooled to 20° C. and concentrated under reduced pressure. The residue was poured into water (200 mL) and neutralized to pH=8-9 with saturated Na2CO3. 300 mL EtOAc was added and the mixture was filtered. The filtrate was collected, separated and the aqueous phase was extracted with EtOAc (200 ml×2). The combined extracts were collected, dried over anhydrous Na2SO4and concentrated under reduced pressure to give compound B27 (4.00 g) as a yellow solid. Procedure for Synthesis of B28 A solution of compound B27 (4.00 g, 12.0 mmol) in H2SO4(50 mL) (90%) was stirred at 20° C. for 16 hours to give a black brown solution. LCMS showed the reaction was completed. The reaction solution was cooled to 0-10° C. with ice-water (100 mL), neutralized to pH=7-8 with Na2CO3(solid) and extracted with EtOAc (300 mL×2). The combined extracts were collected, dried over anhydrous Na2SO4and concentrated under reduced pressure to give compound B28 (1.80 g) as a red solid. Procedure for Synthesis of B30 To a solution of compound B28 (1.00 g, 5.60 mmol) and compound B29 (4.22 g, 16.8 mmol) in dioxane (15 mL)/H2O (5 mL) was added Cs2CO3(3.65 g, 11.2 mmol) and Pd(dppf)Cl2(205 mg, 0.280 mmol) under N2atmosphere. The reaction mixture was heated to 100° C. and stirred for 4 hours under N2atmosphere to give a black mixture. TLC showed the reaction was completed. The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (200 mL×2). The combined extracts were dried over anhydrous Na2SO4and concentrated under reduced pressure. The residue was purified by Combi Flash to give compound B30 (1.00 g) as black brown oil. Procedure for Synthesis of B31 To a solution of compound B30 (1.00 g, 2.86 mmol) in DCM (14 mL) was added TFA (6 mL) slowly. The reaction solution was stirred at 25° C. for 1 hour to give a red solution. LCMS showed the reaction was completed. The reaction solution was concentrated under reduced pressure to give compound B31 (1.00 g) as black brown oil, used for next step without further purification. Procedure for Synthesis of B33 To a solution of compound B19 (2.7 g, 12.9 mmol, 1 eq) and compound 32 (3.15 g, 12.9 mmol, 1.0 eq) in dioxane (50 mL) and H2O (10 mL) were added Na2CO3(2.74 g, 25.9 mmol, 2 eq) and Pd(PPh3)4(748 mg, 0.65 mmol, 0.05 eq) under N2. The resulting mixture was heated at 80° C. and stirred for 12 hrs to give yellow suspension. TLC showed the reaction was completed. The reaction mixture was filtered to give a residue as a light yellow solid. Then the residue was dissolved in DCM (200 mL) and H2O (200 mL), and extracted with DCM (200 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give compound B33 (3.25 g) as a light yellow solid. Procedure for Synthesis of B34 To a solution of compound B33 (2.5 g, 8.64 mmol, 1 eq) in DCM (100 mL) was added Pd/C (1.5 g, 1.75 mmol) under N2. The suspension was degassed under vacuum and purged with H2several times. The mixture was stirred under H2(15 psi) at 15° C. for 12 hours to give black solution. LCMS showed the reaction was completed. The reaction mixture was filtered and concentrated under reduced pressure to give compound B34 (1.96 g) as a yellow solid. Procedure for Synthesis of B35 To a suspension of LiAlH4(673 mg, 17.7 mmol, 2 eq) in THF (20 mL) was added a solution of compound B34 (2.3 g, 8.87 mmol, 1 eq) in THF (10 mL) at 0° C. The reaction mixture was stirred at 15° C. for 1 hour to give red solution. LCMS showed the reaction was completed. The mixture was quenched by saturated NH4Cl solution. The mixture was partitioned between EtOAc and H2O. The aqueous phase was extracted with EtOAc. The combined organic was washed with brine (200 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford compound B35 (1.53 g, crude) as a brown solid. The crude product was used in the next step directly. General Schemes of Group D Procedure for Synthesis of D2 To a solution of compound D1 (500 mg, 2.32 mmol) and 3-hydroxybenzonitrile (276 mg, 2.32 mmol) in THF (20 mL) was added PPh3(730 mg, 2.78 mmol) and DEAD (485 mg, 2.78 mmol). The resulting mixture was stirred at 20° C. for 12 hour to give yellow solution. LCMS and TLC showed the reaction completed. The reaction mixture was quenched by addition H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with brine (30 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash to obtain compound D2 (587 mg) as a yellow oil Procedure for Synthesis of D3 To a solution of compound D2 (587 mg, 1.86 mmol) in MeOH (20 mL) and NH3·H2O (1 mL) (28%) was added Raney-Ni (159 mg, 1.86 mmol) under N2. The suspension was degassed under vacuum and purged with H2several times. The mixture was stirred under H2(15 psi) at 20° C. for 2 hours to give black suspension. LCMS showed the reaction was completed. The reaction mixture was filtered by celite pad and concentrated under reduced pressure to give compound D3 (600 mg) as yellow oil, Procedure for Synthesis of D5 To tert-butyl 4-hydroxypiperidine-1-carboxylate (4.82 g, 23.95 mmol) in DMF (50 mL) was added portionwise NaH (1.44 g, 35.92 mmol) at 0° C. The reaction mixture was stirred at 20° C. for 2 hours. Then 2-fluorobenzonitrile D4 (2.90 g, 23.95 mmol) was added the above mixture and the reaction mixture was heated to 50° C. for 1 hour. TLC showed the reaction was completed. Saturated NH4Cl (100 mL) was added dropwise into the reaction mixture carefully at 0° C. to quench the reaction. The mixture was extracted with EtOAc (100 mL×2). The combined extracts were washed with water (50 mL×3), brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give compound D5 (9.10 g, crude) as a light yellow oil. Procedure for Synthesis of D6 To a mixture of Raney-Ni (425 mg, 4.96 mmol) and compound D5 (3.00 g, 9.92 mmol) in MeOH (90 mL) was added NH3·H2O (6 mL) under N2atmosphere. The suspension was degassed under vacuum and purged with H2three times. The mixture was stirred under H2(15 psi) at 20° C. for 4 hours. LCMS showed the reaction was completed. The reaction mixture was filtered, the filtrate was concentrated to remove most MeOH. The residue was diluted with DCM (100 mL), washed with brine (80 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give compound D6 (2.90 g) as a light yellow oil. The method to prepare compounds of formula IV-5 and IV-6 is shown in Scheme 14. Route IV: Compound G24 can be treated with group E in presence of DIPEA to give compounds IV-1. Compound IV-1 can be treated with mCPBA to give compounds IV-2. Compound IV-2 can be treated with group F in presence of DIPEA to give compounds IV-3. Compound IV-3 can be treated with Pd/C and H2, or HCl to give compounds IV-4. Compound IV-4 can be treated with group C which was defined claim1to give compounds IV-5. Compounds IV-5 can be treated with acid such as HCl to obtain the compounds of formula IV-6. General Schemes of Group E Procedure for Synthesis of E2 To a solution of E1 (93 g, 476 mmol) in DMF (45 mL) was added 1-tert-butoxy-N,N,N′,N′-tetramethyl-methanediamine (249 g, 1.43 mol, 295 mL). The reaction mixture was stirred at 140° C. for 3 hr to give a brown mixture. TLC showed new spot. The mixture was cooled to room temperature and stirred at 0° C. for 30 min. Solid was precipitated out. After filtration, the filter cake was washed with EtOAc/PE and dried under reduced pressure to give compound E2 (69 g) as a purple powder. Procedure for Synthesis of E3 A solution of E2 (63 g, 252 mmol), (2,4-dimethoxyphenyl)methanamine (63 g, 378 mmol, 57 mL) in toluene (150 mL) was stirred at 35° C. for 2 hours to give a yellow mixture. Then the reaction was stirred at 65° C. for 2 hours to give a yellow mixture. Then the reaction was stirred at 110° C. for 3 hours to give a yellow mixture. TLC showed new spot. The mixture was cooled to 20° C. The yellow solid was precipitated out. The mixture was filtered. The filter cake was washed with PE (50 mL) for twice and dried over high vacuum to give compound E3 (50 g) as yellow solid. Procedure for Synthesis of E4 A solution of E3 (55 g, 162 mmol) in TFA (330 mL) was stirred at 70° C. for 16 hours to give a purple mixture. TLC showed new spot. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was added PE (60 mL), and stirred at 25° C. for 2 hours to give a purple mixture. After filtration, the filter cake was washed with PE (50 mL), dried under reduced pressure to give compound E4 (80 g, crude) as purple solid. Procedure for Synthesis of E5 A solution of E4 (80 g, 421 mmol) in POCl3(341 mL) was stirred at 100° C. for 2 hours to give a brown mixture. TLC showed new spot. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (1000 mL). The organic layer was washed with saturated NaHCO3solution (1000 mL), solid was precipitated out. After filtration, the filter cake was washed with DCM (300 mL×2). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a crude product. The residue was purified by column chromatography to give compound E5 (35 g) as light yellow solid. Procedure for Synthesis of E7 To a solution of E6 (40 g, 183 mmol) in MeCN (400 mL) was added NBS (35.9 g, 202 mmol), BPO (444 mg, 1.83 mmol). The reaction mixture was stirred at 90° C. for 4 hours to give a brown mixture. LCMS showed the desired MS. The reaction mixture was diluted with water (200 mL) and extracted with EtOAc (100 mL*2). The organic layer was washed with water (100 mL*4), brine (100 mL*3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give E7 (57 g, crude) as brown oil. Procedure for Synthesis of E8 A mixture of E7 (57 g, 192 mmol) and NaN3 (18.8 g, 290 mmol) in DMSO (290 mL) was stirred at 25° C. for 16 hours to give a brown mixture. LCMS showed the desired MS. The reaction mixture was quenched with NaHCO3(500 mL) and extracted with EtOAc (200 mL*2). The organic layer was washed with water (300 mL*4), brine (300 mL*4), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give E8 (42 g, crude) as brown oil. Procedure for Synthesis of E9 To a solution of E8 (12 g, 46.3 mmol) in dioxane (60 mL) and H2O (15 mL) was added E5 (9.66 g, 46.3 mmol), Cs2CO3(30.2 g, 92.6 mmol, 2 eq) and Pd(dppf)Cl2(1.69 g, 2.32 mmol, 0.05 eq). The reaction mixture was stirred at 80° C. under N2atmosphere protect for 5 hours to give a black mixture. TLC showed new spot. The reaction mixture was diluted with water (300 mL) and extracted with EtOAc (200 mL*2). The organic layer was washed with water (100 mL*4), brine (100 mL*2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography to give E9 (8 g) as brown solid. Procedure for Synthesis of E10 To a solution of E9 (8 g, 26.2 mmol, 1 eq) in THF (80 mL)/H2O (40 mL) was added PPh3(10.3 g, 39.3 mmol, 1.5 eq). The reaction mixture was stirred at 60° C. for 16 hours to give a brown mixture. TLC showed the reaction completed. The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (100 mL*2). The organic layer was washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a E10 (7.32 g, crude) as brown gum. Procedure for Synthesis of E12 To a mixture of E11 (2 g, 8.93 mmol) in pyridine (20 mL) was added Tf2O (3.02 g, 10.71 mmol, 1.77 mL, 1.2 eq) at 0° C., the mixture was stirred at 0° C. for 2 hours to give a brown mixture. LCMS showed the reaction was completed. The reaction mixture was quenched with water (20 mL) and extracted with EtOAc (20 mL×2). The organic layer was washed with water (20 mL×2), brine (30 mL×4), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude product. The crude product was purified by combi flash to afford E12 (2.2 g) as a white solid. Procedure for Synthesis of E13 The compound E13 (2.2 g) was followed the same procedure of B33 to obtain 1.3 g of compound E13 as a yellow powder. Procedure for Synthesis of E14 To a mixture of E13 (600 mg, 1.77 mmol) and NH2Boc (248 mg, 2.12 mmol) in dioxane (10 mL) was added Pd(OAc)2(39.71 mg, 176.89 umol) and Xantphos (204 mg, 353.79 umol) and Cs2CO3(1.15 g, 3.54 mmol), the mixture was stirred at 100° C. for 16 hours to give a black mixture. TLC showed the reactant was consumed. The reaction mixture was quenched with water (20 mL) and extracted with EtOAc (20 mL×2). The organic layer was washed with water (20 mL×2), brine (30 mL×4), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude product. The crude product was purified by combi flash to afford E14 (190 mg) as a yellow solid. Procedure for Synthesis of E15 The compound E14 (400 mg) was followed the same procedure of B11 to obtain 385 mg of compound E15 as a yellow gum. Procedure for Synthesis of E16 The compound B12 (10.2 g) was followed the same procedure of B14 to obtain 15.2 g of compound E16 as a yellow powder. Procedure for Synthesis of E17 To a solution of compound E16 (7 g, 26.7 mmol) in THF (45 mL) was added LDA (2 M, 20 mL) at −78° C. under N2. The reaction was stirred at −78° C. for 30 min. Tributyl(chloro)stannane (10.4 g, 32 mmol, 1.2 eq) was added into the reaction at −78° C. The reaction was stirred at −78° C. for 30 min and the reaction was stirred at 15° C. for 17 hours to give a yellow mixture. TLC showed the starting material was not consumed completely. Saturated NH4Cl (20 mL) was added into the reaction mixture. The mixture was partitioned between EtOAc (300 mL) and H2O (300 mL). The organic extract was washed with saturated NH4Cl (300 mL), brine (300 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give yellow gum. The crude product was purified by combi flash to give compound E17 (9.8 g) as yellow oil. Procedure for Synthesis of E19 To a suspension of compound E18 (1 g, 5.10 mmol) in THF (10 mL) was added BH3·THF (1 M, 15.3 mL) under N2atmosphere at 0° C. The reaction was stirred at 15° C. for 30 min to give a yellow mixture. LCMS showed the starting material was not consumed completely. The reaction was stirred at 15° C. for 17 hours to give a yellow mixture. LCMS showed the starting material was consumed completely. Little mixture was purified by prep-TLC to give a sample. The reaction was quenched with MeOH (4 mL). The reaction mixture was concentrated under reduced pressure to give E19 (1.1 g, crude) as a yellow gum. Procedure for Synthesis of E20 The compound B19 (1.1 g) was followed the same procedure of B14 to obtain 890 mg of compound E20 as a colorless oil. Procedure for Synthesis of E21 The compound E20 (800 mg) and E17 (1.91 g) was followed the same procedure of B33 to obtain 385 mg of compound E21 as a yellow oil. Procedure for Synthesis of E22 The compound E21 (600 mg) was followed the same procedure of A7 to obtain 450 mg of compound E22 as a yellow powder. General Schemes of Group F Procedure for Synthesis of F2 To a mixture of compound F1 (3 g, 16.20 mmol) and trimethylsilylformonitrile (1.61 g, 16.20 mmol, 2.03 mL) was added AlCl3(60 mg, 449.97 umol, 24.59 uL, 2.78e-2 eq), and the mixture was heated to 50° C. for 20 hours. TLC showed the reaction was completed. The reaction was quenched by water (10 mL) and then extracted with EtOAc (10 mL*2). The combined organic phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give the compound F2 (4.6 g) as a yellow solid. Procedure for Synthesis of F3 To a solution of compound F2 (1 g, 3.52 mmol) in MeOH (10 mL) was added K2CO3(52.48 mg, 379.71 umol). The mixture was stirred at 20° C. for 2 hr. TLC indicated that material was disappeared and one major new spot with larger polarity was detected. 8.75 mL of HCl (0.1 M) was added to the reaction mixture, and then it was extracted with EtOAc (2*20 mL). The combined organic layer was dried over (Na2SO4) and evaporated to dryness to give compound F3 (0.74 g) as yellow oil. Procedure for Synthesis of F4 A mixture of compound F3 (2.42 g, 11.40 mmol), imidazole (993.59 mg, 14.59 mmol) and tert-butyl-chloro-dimethyl-silane (2.06 g, 13.68 mmol, 1.68 mL) in DMF (10 mL) was stirred at 20° C. for 14 hr. TLC indicated that material was consumed completely and new spots formed. The reaction mixture was diluted with water 50 mL and then it was extracted with EtOAc 100 mL (50 mL×2). The combined organic layers were washed with aqueous NaCl 100 mL (50 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography eluted with (PE:EtOAc=5:1) to give compound F4 (2.44 g) as colorless oil. Procedure for Synthesis of F5 To the solution of compound F4 (1 g, 3.06 mmol) in THF (12 mL) was added LAH (200 mg, 5.27 mmol) at 0° C., and it was stirred at 0° C. for 0.5 hr to give a white suspension. TLC indicated that material disappeared. The reaction mixture was diluted with THF (20 mL) and then quenched with water (0.2 mL), 15% aq. NaOH (0.2 mL) and water (0.6 mL) at 0° C. Then it was stirred at 0° C. for 0.5 hr. The mixture was dried over anhydrous Na2SO4and then filtered, and the filter cake was washed with EtOAc (20 mL). The combined filtrate was concentrated under reduced pressure to give compound F5 (870 mg, mixture of 2 trans-isomer) as colorless oil. Procedure for Synthesis of F7 To a mixture of F6 (1.7 g, 7.82 mmol), TEA (1.03 g, 10.2 mmol) in DCM (10 mL) was added TosCl (1.57 g, 8.21 mmol, 1.05) at 0° C. The reaction was stirred at 20° C. for 17 hours to give a yellow solution. LCMS showed the desired MS value was observed. The reaction was concentrated under reduced pressure. The concentrate was dissolved in EtOAc (150 mL) and the resulting solution was washed with aqueous HCl (100 mL, pH=4), aqueous NaOH (1 N, 100 mL), brine (150 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give F7 (2.49 g) as yellow oil. Procedure for Synthesis of F8 To a mixture of F7 (2.3 g, 6.19 mmol) in DMSO (20 mL) was added KCN (443 mg, 6.81 mmol, 291 uL) and KI (1.54 g, 9.29 mmol), the mixture was stirred at 80° C. for 3 hours and 100° C. for 2 hours to give a black mixture. TLC showed the reactant was consumed. The reaction mixture was quenched with water (50 mL) and extracted with EtOAc (50 mL×2). The organic layer was washed with water (50 mL×2), brine (30 mL×4), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude product. The crude product was purified by combi flash to afford F8 (1.19 g) as a yellow oil. Procedure for Synthesis of F9 A solution of F8 (600 mg, 2.65 mmol) in MeOH (20 mL) was added Raney-Ni (227 mg, 2.65 mmol), the suspension was degassed under vacuum and purged with H2several times, the mixture was stirred at 25° C. under H2(15 psi) for 16 hours to give a black mixture. TLC (PE/EA=1/1) showed the reaction was completed. The mixture was filtered and concentrated under reduced pressure to afford F9 (530 mg, crude) as a yellow oil. Commercial available reagents were used for group E such as benzyl piperidin-4-ylcarbamate, tert-butyl piperidin-4-ylcarbamate, tert-butyl piperazine-1-carboxylate, tert-butyl 2-(aminomethyl)morpholine-4-carboxylate, tert-butyl 2-(hydroxymethyl)morpholine-4-carboxylate, tert-butyl 4-aminopiperidine-1-carboxylate, 4-methoxypiperidine, piperidin-4-ylmethanamine, 4-methoxycyclohexan-1-amine, (tetrahydro-2H-pyran-4-yl)methanamine, morpholine, azepan-4-ol, pyrrolidin-3-ol, 4-aminocyclohexan-1-ol, N-methylpiperidin-4-amine, (1R,4R)-4-(aminomethyl)cyclohexan-1-ol, (1S,3 S)-3-aminocyclopentan-1-ol, piperidin-4-ol, 4-(aminomethyl)piperidin-2-one, piperidin-4-ylmethanol, 4-(trifluoromethoxy)piperidine, 4-ethoxypiperidine, 4-isopropoxypiperidine, 4-ethoxycyclohexan-1-amine, 4-methoxycyclohexan-1-amine, 4-isopropoxycyclohexan-1-amine and 3-aminopropan-1-ol. The method to prepare compounds of formula V-5 and V-6 is shown in Scheme 20. Route V: Compound G3 can be treated with NaOEt to give compounds I1. Compound I1 can be treated with POCl3to give compounds I2. Compound I2 can be treated with group E in presence of DIPEA to give compounds V-1. Compound V-1 can be treated with Boc2O to give compounds V-2. Compound V-2 can be treated with group F in presence of Pd2(dba)3to give compounds V-3. Compound V-3 can be treated with Pd/C and H2, or HCl to give compounds V-4. Compound V-4 can be treated with group C which was defined claim1to give compounds V-5. Compounds V-5 can be treated with acid such as HCl to obtain the compounds of formula V-6. Procedure for Synthesis of I1 Na (790 mg, 34.3 mmol) was added into anhydrous EtOH (125 mL), the mixture was stirred at 10° C. for an hour. Compound G3 (3.50 g, 27.9 mmol) and diethyl propanedioate (5.42 g, 33.8 mmol) were added into this solution. The mixture was stirred at 78° C. for 16 hours under N2atmosphere to give a yellow solution. TLC showed the reaction was completed. The mixture was concentrated under reduced pressure. The residue was dissolved in water (60 mL) and acidified to pH=3 with 3M HCl, then filtered to give compound I1 (3.70 g) as a white powder. Procedure for Synthesis of I2 To a mixture of compound I1 (1.30 g, 6.73 mmol) in POCl3(20.6 g, 134 mmol) was added N,N-diethylaniline (672 mg, 4.51 mmol). The mixture was stirred at 100° C. for 16 hours under N2atmosphere. TLC showed a new spot. Most of POCl3was removed under reduced pressure. Then the mixture was poured into H2O (40 mL), extracted with DCM (50 mL×3). The organic layer was washed with brine (50 mL×2), dried over anhydrous Na2SO4, then filtered, and concentrated under reduced pressure to give compound I2 (3.11 g, crude) as a yellow oil, without further purification for next step. Procedure for Synthesis of G2 To a mixture of diisopropylamine (6.69 g, 66.2 mmol) in anhydrous THF (20 mL) was added n-BuLi (2.5 M, 27.7 mL) at 0° C. and stirred at 0° C. for 0.5 hour, then the mixture was cooled to −70° C. and G1 (5.00 g, 60.2 mmol) in THF (20 mL) was added into the mixture at −70° C. and stirred at −70° C. for 0.5 hour, then the mixture was poured into a mixture of ethyl formate (4.90 g, 66.2 mmol) in THF (20 mL) at −70° C. under N2atmosphere and the resulting mixture was stirred at −70° C. for 0.5 hours, then warmed to 15° C. and stirred at 15° C. for 17 hours. TLC (silica gel, PE/EtOAc=2/1) showed the reaction was completed. The reaction mixture was poured into aqueous HCl (150 mL, 1M) at 0° C. and stirred at 0° C. for 0.5 hours, then the mixture was extracted with EtOAc (150 mL×3). The organic layer was washed with brine (250 ml), dried over anhydrous Na2SO4, filtered, the filtrate was concentrated under reduced pressure to give compound G2 (5.0 g) as a yellow oil. The crude product was used directly in next step without further purification. Procedure for Synthesis of G3 To a solution of compound G2 (8.82 g, 67.5 mmol) and AcOH (7.09 g, 118 mmol) in EtOH (5 mL) was added NH2—NH2·H2O (4.39 g, 87.7 mmol), the resulting mixture was stirred at 78° C. for 17 hours to give a pale yellow solution. TLC showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to give a residue, then the pH value of the residue was adjusted to 9 with aqueous NaOH (1M), diluted with water (50 mL) and extracted with EtOAc (100 mL×3). The organic layer was washed with brine (150 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give compound G3 (10.0 g, crude) as a yellow gum. The crude product was used directly in next step without further purification. Procedure for Synthesis of G4 To a mixture of compound G3 (5.00 g, 40.0 mmol) in anhydrous DCM (25 mL) was added a mixture of ethoxycarbonyl isothiocyanate (4.72 g, 36.0 mmol) in anhydrous DCM (25 mL) at −70° C. and stirred at −70° C. for 1 hour, a lot of white solid appeared. TLC showed the reaction was completed. Then the mixture was allowed to warm to −10° C. and filtered, and the filter cake was washed with DCM (15 mL) to give 4.50 g of desired compound as a white solid, the structure was confirmed by HNMR. The filtrate was purified by silica gel column to give compound G4 (1.80 g) as a white solid. Procedure for Synthesis of G5 To a mixture of compound G4 (6.30 g, 24.6 mmol) in MeCN (50 mL) was added K2CO3(6.79 g, 49.2 mmol), the mixture was stirred at 80° C. for 8 hours. Crude LCMS showed the reaction was completed. The mixture was cooled to room temperature, then AcOH (15 mL) was added into the mixture and stirred at 15° C. for 20 minutes, then the resulting mixture was concentrated under reduced pressure to give a residue, which was washed with water (50 mL×3) to give compound G5 (4.20 g) as a white solid. Procedure for Synthesis of G6 To a mixture of compound G5 (4.20 g, 20.0 mmol) in EtOH (40 mL) was added NaOH (2.00 g, 50.0 mmol) in H2O (20 mL) at 15° C., then MeI (2.84 g, 20.0 mmol) was added into above mixture and the resulting mixture was stirred at 15° C. for 2 hours. Crude LCMS showed the reaction was completed. The mixture was concentrated under reduced to give a residue, which was treated with ice cold water (50 mL) and aqueous HCl (20 mL, 6M) for 30 minutes, a lot of white solid appeared, filtered to give the crude product. The crude product was poured into MeCN (50 mL) to give a suspension, then the suspension was concentrated under reduced pressure to give compound G6 (3.60 g) as a white solid. Procedure for Synthesis of G7 To a mixture of compound G6 (1.00 g, 4.46 mmol) in DCM (30 mL) was added m-CPBA (3.07 g, 14.2 mmol) in portions at 20° C. The reaction mixture was stirred at 20° C. for 2 hours. LCMS showed the reaction was complete. The reaction mixture was diluted with a mixture solution of brine (20 mL) and NaOH/H2O (3M, 10 mL). The aqueous layer was separated and brought pH to 1 with HCl (3M). A lot of white powder was precipitated. The mixture was then extracted with EtOAc (50 mL×2). The organic layer was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure to give a residue. The residue was purified by Combi Flash to give compound G7 (1.03 g) as light brown gum. Procedure for Synthesis of G9 To a solution of compound G7 (6.62 g, 25.83 mmol) in NMP (100 mL) was added compound G8 (18.15 g, 77.5 mmol). The reaction mixture was stirred at 140° C. for 16 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between brine (500 mL) and EtOAc (400 mL). The organic layer was washed with water (100 mL×2), brine (100 mL), dried over anhydrous Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by Combi Flash to give a brown gum, which was triturated with CH3CN (50 mL) to give compound G9 (2.06 g) as an off-white powder. Procedure for Synthesis of G10 To a mixture of compound G9 (89 mg, 0.22 mmol) in POCl3(4.41 g, 28.7 mmol) was added N,N-diethylaniline (97 mg, 0.65 mmol) at 20° C. The reaction mixture was stirred at 80° C. for 2 hours. The reaction mixture was concentrated under reduced pressure to give compound G10 (93 mg) as an brown gum as the crude product. The crude product was used directly in next step without further purification. Procedure for Synthesis of G11 To a solution of compound G10 (127 mg, 0.297 mmol), DIPEA (95.9 mg, 0.742 mmol) in DMF (2 mL) was added compound A7 (131 mg, 0.371 mmol). The reaction solution was stirred at 10° C. for 1 hour. LCMS showed the desired MS value. The reaction mixture was partitioned between EtOAc (20 mL) and saturated aqueous NaHCO3solution (20 mL). The organic layer was washed with brine (10 mL), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure. The residue was purified by Combi flash to give compound G11 (103 mg) as a brown gum Procedure for Synthesis of Compound 1 To a solution of compound G11 (70 mg, 0.094 mmol) in AcOH (0.5 mL) was added HBr/AcOH (0.5 mL, 35% purity) at 15° C. This reaction solution was stirred at 15° C. for 1 hour to give a light brown solution. LCMS showed the reaction was complete. The reaction was diluted with 5 mL of MTBE to precipitate a grey powder, which was collected by filtration. The solid was dissolved in MeOH (4 mL), and then a drop of ammonia water (28%) was added to basify the solution. The crude product dissolved in MeOH was purified by prep-HPLC. The eluent containing the desired product was concentrated under reduced pressure and the residual solution was lyophilized to give compound 1 (9.9 mg) as a white powder. Procedure for Synthesis of G12 To a solution of compound A23 (183 mg, 0.385 mmol) in CH3CN (10 mL) was added DIPEA (226 mg, 1.75 mmol) and compound G10 (150 mg, 0.35 mmol). The resulting mixture was stirred at 20° C. for 1 hour to give white suspension. TLC showed the reaction was completed. The reaction mixture was quenched by addition H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with brine (10 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash to obtain compound G12 (75 mg) as an off-white solid. Procedure for Synthesis of Compound 9 A solution of compound G12 (30 mg, 0.4 mmol) in HBr/HOAC (35%) (1 mL) was stirred at 20° C. for 1 hour to give yellow solution. TLC showed the reaction was completed. The reaction was addition MTBE (50 mL) and lots of solid was precipitated, filtered under reduced pressure to give filter cake as an off-white solid. The filter cake was purified by prep-HPLC. The residue was concentrated most of solvent and lyophilized to obtain compound 9 (7.8 mg) as a white powder. Procedure for Synthesis of G13 A solution of compound B11 (50 mg, 0.2 mmol) and compound G10 (90 mg, 0.2 mmol) in CH3CN (5 mL) was added DIPEA (27.1 mg, 0.2 mmol). The resulting mixture was stirred at 0° C. for 30 min to give red solution. TLC showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by Combi flash to obtain compound G13 (100 mg) as a yellowish solid. Procedure for Synthesis of G14 To a solution of compound G13 (100 mg, 0.158 mmol) in THF (5 mL) was added compound A5 (58.2 mg, 0.317 mmol) and DIPEA (82 mg, 0.634 mmol), the resulting mixture was stirred at 20° C. for 1 hour to give red solution, LCMS showed the reaction was completed, dimethylamine (71.5 mg, 1.59 mmol) was added to the mixture, then stirred for another 3 hours at 20° C. to give red solution. TLC showed the reaction was completed. The reaction mixture was quenched by addition H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash to give 100 mg (impure), which was purified by prep-TLC to obtain compound G14 (50 mg) as a yellow solid. Procedure for Synthesis of Compound 16 A solution of compound G14 (50 mg, 67.4 umol) in HBr/HOAc (35%) (1 mL) was stirred at 20° C. for 30 min to give red solution. LCMS showed the reaction was completed. To the reaction mixture was added MTBE (3 mL) to precipitate a yellow gum. The yellow gum was collected by filtration and washed with MTBE (3 mL×2). The yellow gum was dissolved in MeOH (2 mL) and purified by cation exchange resin eluting with 5% NH3·H2O/MeOH, then lyophilized to obtain compound 16 (29.9 mg) as a yellow solid. Procedure for Synthesis of G15 To a mixture of compound B17 (403 mg, 0.866 mmol) in MeCN (5 mL) was added DIPEA (976 uL), compound G10 (338 mg, 0.787 mmol). The mixture was stirred at 15° C. for 0.5 hour to give a yellow mixture. TLC showed the reaction was completed. The mixture was partitioned between DCM (50 mL) and water (50 mL), the mixture was extracted with DCM (50 mL×2), the combined extracted was washed with brine (50 mL), dried over Na2SO4, filtered, concentrated under reduced pressure to give a yellow oil, which was purified by Combi flash to give compound G15 (250 mg) as yellow oil. Procedure for Synthesis of G16 To a mixture of compound G15 (250 mg, 0.397 mmol) in THF (5 mL) was added DIPEA (0.257 mg, 1.98 mmol), compound A5 (218 mg, 1.19 mmol). The yellow mixture was stirred at 15° C. for 1 hour. The color of mixture was become black. LCMS showed the reaction was completed. Dimethylamine (2 M, 992 uL) was added to the mixture and stirred at 15° C. for 1 hour to give a black brown mixture. LCMS showed the reaction was completed. The mixture was partitioned between DCM (50 mL) and water (30 mL), The aqueous was extracted with DCM (50 mL×2), the combined extracted phase was washed with brine (30 mL), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure to give a brown oil, which was purified by Combi flash to give compound G16 (250 mg) as a brown gum. Procedure for Synthesis of Compound 25 A mixture of compound G16 (250 mg, 0.337 mmol) in HBr/HOAc (3 mL) was stirred at 15° C. for 0.5 hour to give a yellow mixture. LCMS showed the reaction was complete. To the reaction mixture was added MTBE (10 mL) to precipitate yellow powder. The white powder was collected by filtration and washed with MTBE (5 mL×2), basified by saturated Na2CO3to pH=9-10, and extracted with DCM (50 mL×2), the combined organic phase was washed with water (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give yellow oil, which was purified by prep-HPLC (0.1% TFA), basified by saturated Na2CO3to pH=9-10, and extracted with DCM (50 mL×3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give compound 25 (30 mg) as a white powder. Procedure for Synthesis of G17 To a solution of compound B23 (230 mg, 0.633 mmol) in CH3CN (5 mL) was added DIPEA (818 mg, 6.33 mmol) and compound G10 (299 mg, 0.696 mmol) at 0-5° C. The reaction was stirred at 25° C. for 1 hour to give a black brown solution. LCMS showed desired MS value. The reaction was diluted with DCM (30 mL) and concentrated under reduced pressure. The residue was partitioned between water (50 mL) and DCM (100 mL). The separated organic layer was washed with water (80 mL), brine (80 mL), dried over anhydrous Na2SO4and concentrated under reduced pressure. The residue was purified by Combi Flash to give compound G17 (150 mg) as yellow oil. Procedure for Synthesis of G18 To a solution of compound G17 (200 mg, 0.312 mmol) in THF (5 mL) was added DIPEA (201 mg, 1.56 mmol) and compound A5 (172 mg, 0.935 mmol). The reaction solution was stirred at 25° C. for 30 minutes to give a black brown solution. Then, dimetnylamine (2 M, 1.56 mL) was added and stirred for another 16 hours to give a black brown solution LCMS and TLC showed the reaction was completed. The reaction mixture was concentrated under reduced pressure. The residue was partitioned between DCM (150 mL) and water (100 mL). The organic layer was washed with water (100 mL), brine (100 mL) and concentrated under reduced pressure. The residue was purified by Combi Flash to give compound G18 (100 mg) as a yellow oil. Procedure for Synthesis of Compound 20 A solution of compound G18 (150 mg, 0.199 mmol) in HBr/HOAc (2 mL, 35%) was stirred at 25° C. for 0.5 hour to give a red solution. LCMS showed the reaction was completed. 10 mL MTBE was added and the resulting white mixture was filtered. The filter cake was collected, washed with MTBE (20 mL), dissolved in MeOH (3 mL) and basified by cation exchange resin to give compound 20 (46.6 mg) as a yellow powder. Procedure for Synthesis of G19 To a solution of compound B31 (400 mg, 0.933 mmol) in CH3CN (10 mL) was added DIPEA (362 mg, 2.80 mmol) and compound G10 (500 mg, 1.38 mmol) at 0-10° C. The reaction was stirred for 1 hour at 25° C. to give a brown solution. Crude LCMS showed the reaction was completed. TLC showed new spot formed. The reaction solution was concentrated under reduced pressure. The residue was purified by Combi Flash to give compound F19 (380 mg) as a yellow gum. Procedure for Synthesis of G20 To a solution of compound G19 (380 mg, 0.600 mmol) in THF (5 mL) was added a solution of compound A5 (330 mg, 1.80 mmol) in THF (2 mL) at 0-5° C. in an ice-bath. The reaction solution was stirred at 0-25° C. for 0.5 hour. Dimethylamine (2M, 3 mL, 10 eq) was added and stirred for another 2 hours to give a black brown solution. Crude LCMS showed the reaction was completed. TLC showed the reaction was completed. The reaction solution was concentrated under reduced pressure. The residue was dissolved in DCM (200 mL), washed with saturated NH4Cl (100 mL), water (100 mL) and brine (100 mL), the organic layer was dried over anhydrous Na2SO4and concentrated under reduced pressure. The residue was purified by Combi Flash to give compound G20 (130 mg) as a yellow solid. Procedure for Synthesis of Compound 11 A solution of compound G20 (70 mg, 0.093 mmol) in HBr/HOAc (1 mL, 35%) was stirred at 25° C. for 0.5 hour to give a yellow solution. LCMS showed desired MS value. 10 mL MTBE was added and the resulting mixture was filtered to give a white solid. The solid was dissolved in MeOH (2 mL), purified by cation exchange resin and lyophilized to give a white powder which was further purified by prep-HPLC. The fraction was concentrated under reduced pressure. The residue was neutralized to pH=7-8 with K2CO3(solid). The resulting white mixture was extracted with EtOAc (50 mL×2). The organic layer was dried over anhydrous Na2SO4, concentrated and lyophilized to give compound 11 (23.1 mg) as a white powder Procedure for Synthesis of G21 To a solution of compound D3 (300 mg, 0.7 mmol) and compound G10 (448 mg, 1.40 mmol) in CH3CN (20 mL) was added DIPEA (181 mg, 1.40 mmol). The resulting mixture was stirred at 20° C. for 1 hour to give yellow solution. LCMS showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to give a residue, TLC. The residue was purified by Combi flash to compound G21 (240 mg) as a yellow oil. Procedure for Synthesis of G22 To a solution of compound G21 (240 mg, 0.34 mmol) in DCM (7 mL) was added TFA (4.62 g, 40.5 mmol). The resulting mixture was stirred at 20° C. for 1 hour to give yellow solution. TLC and LCMS showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to give compound G22 (268 mg) as a yellow oil. Procedure for Synthesis of G23 To a solution of compound G22 (268 mg, 0.37 mmol) in THF (20 mL) was added DIPEA (143 mg) and compound A5 (67.6 mg, 0.37 mmol), the resulting mixture was stirred at 20° C. for 1 hour, LCMS showed the reaction was completed, then dimethylamine (83.1 mg, 1.84 mmol) was added to the mixture and stirred for another 2 hours to give yellow suspension. TLC showed the reaction was completed. The reaction mixture was quenched by addition H2O (50 mL), extracted with EtOAc (50 mL). The organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash to give compound G23 (74 mg) as a yellow oil. Procedure for Synthesis of Compound 18 A solution of compound G23 (74.0 mg, 0.1 mmol) in HBr/HOAc (35%) (1 mL) was stirred at 20° C. for 1 hour to give yellow solution. LCMS showed the reaction was completed. To the reaction mixture was added MTBE (3 mL) to precipitate a white gum. The gum solid was collected by filtration and washed with MTBE (3 mL×2). The white gum was dissolved in MeOH (2 mL) and purified by cation exchange resin (PCX-SPE) eluting with 5% NH3·H2O/MeOH, the flows was concentrated and lyophilized to obtain compound 18 (42.9 mg) as a white powder. Procedure for Synthesis of G24 To a mixture of compound G6 (1.00 g, 4.46 mmol) in POCl3(14 mL) was added N,N-diethylaniline (2.00 g, 13.4 mmol) at 15° C., the mixture was stirred at 90° C. for 3 hours. The mixture was concentrated under reduced pressure to give compound G24 (3.20 g, crude) as a brown gum, which was used to next step without purification. Procedure for Synthesis of G25 To a solution of E10 (7.32 g, 26.2 mmol) in MeCN (70 mL) was added DIEA (6.77 g, 52.4 mmol, 9.13 mL), G24 (6.36 g, 26.2 mmol). The reaction mixture was stirred at 25° C. for 16 hours to give brown mixture. TLC showed new spot. The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (100 mL*2). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography to give G25 (11.3 g) as yellow solid. Procedure for Synthesis of G26 To a mixture of G25 (11.3 g, 23.2 mmol) in DCM (110 mL) was added m-CPBA (10.0 g, 46.5 mmol, 80% purity) at 0-10° C., the mixture was stirred at 0-10° C. for 2 hours to give a yellow mixture. LCMS showed the reaction was not completed. The mixture was stirred at 0-10° C. for another 1 hour to give a yellow mixture. LCMS showed the desired product was observed. The mixture was partitioned between DCM (100 mL) and saturated aqueous Na2SO3(100 mL). The aqueous phase was extracted with DCM (100 mL×2). The combined organic extract was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude product. The crude product was purified by combi flash to afford G26 (6.6 g) as a yellow solid. Procedure for Synthesis of G27 To a solution of 4-methoxycyclohexanamine (71.9 mg, 0.556 mmol) and G26 (180 mg, 0.348 mmol) in NMP (1 mL) was added DIEA (135 mg, 1.04 mmol). The resulting mixture was heated at 110° C. and stirred for 12 hrs to give brown solution. LCMS and TLC showed the reaction was completed. The reaction mixture was quenched by addition H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with brine (30 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1:1) to obtain compound G27 (105 mg) as an orange yellow solid. Procedure for Synthesis of G28 The compound G27 (105 mg) was followed the same procedure of B7 to obtain 110 mg of compound G28 as a yellow solid. Procedure for Synthesis of Compound 44 To a solution of G28 (110 mg, 0.172 mmol) in NMP (2 mL) was added (E)-4-(dimethylamino)but-2-enoyl chloride (3 M, 172 uL) at 0° C., the resulting mixture was stirred at 15° C. for 12 hour to give yellow solution, LCMS showed most of the starting material consumed. The reaction mixture was quenched by addition H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC, then concentrated to obtain compound 44 (21 mg) as a light yellow solid. Procedure for Synthesis of G29 To a solution of compound G26 (300 mg, 0.58 mmol) and tert-butyl piperazine-1-carboxylate (323.88 mg, 1.74 mmol) in NMP (3 mL) was added DIPEA (149.83 mg, 1.16 mmol, 201.93 uL). The resulting mixture was heated at 110° C. and stirred for 2 hrs to give yellow solution. LCMS and TLC showed the reaction was completed. The reaction mixture was quenched by addition H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with brine (30 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography to obtain compound G29 (285 mg) as a yellow solid. Procedure for Synthesis of G30 The compound G29 (285 mg) was followed the same procedure of B7 to obtain 258 mg of compound G30 as a yellow solid. Procedure for Synthesis of G31 To a solution of 2-fluoroprop-2-enoic acid (16.38 mg, 181.90 umol) in DMF (2 mL) were added TEA (46.02 mg, 454.76 umol, 63.30 uL) and HATU (69.17 mg, 181.90 umol), and then compound G30 (90 mg, 151.59 umol) was added to the mixture and stirred at 15° C. for 2 hours to give brown solution. LCMS showed part of the starting material remained, the reaction was stirred at 15° C. for another 2 hours to give brown solution. LCMS indicated that the reaction was almost complete. The reaction mixture was poured into water (15 mL), and the yellow suspension was filtered. The filter cake was washed with PE (10 mL) and dried to give compound G31 (100 mg) as a yellow solid. Procedure for Synthesis of Compound 79 A mixture of compound G31 (100 mg) in 4M HCl/MeOH (2 mL) was stirred at 15° C. for 1 hr to give a yellow solution. LCMS and HPLC indicated that the reaction worked well. The solvent was removed under reduced pressure to give the crude product. It was purified by prep-HPLC most of solvent was removed under reduced pressure, and the remaining solvent was removed by lyophilization to give compound 79 (34.7 mg) as a yellow solid. Procedure for Synthesis of G32 To a mixture of compound G26 (500 mg, 0.96 mmol) and tert-butyl N-(4-piperidyl)carbamate (386 mg, 1.93 mmol) in NMP (5 mL) was added DIEA (249 mg, 1.93 mmol, 336 uL), and the mixture was stirred at 110° C. for 16 hours to give a brown mixture. LCMS showed the reaction was completed. The reaction mixture was quenched with water (20 mL) and extracted with EtOAc (20 mL×2). The organic layer was washed with water (20 mL×2), brine (30 mL×4), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude product. The crude product was purified by combi flash to afford compound G32 (520 mg) as a red solid. Procedure for Synthesis of G33 The compound G32 (520 mg) was followed the same procedure of B7 to obtain 450 mg of compound G33 as a yellow solid. Procedure for Synthesis of G34 To a solution of compound G33 (50 mg, 82 umol, 1 eq) in THF (2 mL) was added a solution of A5 (30 mg, 164 umol, 2 eq) in THF (2 mL) at 0-5° C. in an ice-bath. The reaction solution was stirred at 0-25° C. for 1.5 hours. 2-(methylamino)acetamide (21 mg, 246 umol, 3 eq) and DIEA (10 mg, 82 umol, 14 uL, 1 eq) were added and it was stirred for another 16 hours to give a black brown solution. LCMS showed the desired product was observed. The reaction mixture was quenched with water (20 mL) and extracted with EtOAc (20 mL×2). The organic layer was washed with water (20 mL×2), brine (30 mL×4), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude product. The crude product was purified by prep-TLC to afford compound G34 (22 mg) as a white powder. Procedure for Synthesis of Compound 58 The compound G34 (22 mg) was followed the same procedure of compound 79 to obtain 3 mg of compound 58 as a yellow solid. Procedure for Synthesis of G35 To a solution of compound E10 (728 mg, 2.61 mmol) and compound 12 (500 mg, 2.17 mmol) in EtOH (10 mL) was added DIPEA (561 mg, 4.35 mmol, 0.757 mL, 2 eq). The resulting mixture was stirred at 75° C. for 12 hrs to give brown solution. TLC showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to give a residue. Then diluted with H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography to obtain compound G35 (948 mg) as a brown gum. Procedure for Synthesis of G36 The compound G35 (948 mg) was followed the same procedure of compound B14 to obtain 930 mg of compound G36 as a yellow solid. Procedure for Synthesis of G38 To a mixture of compound G36 (200 mg, 349 umol) and compound G37 (90.19 mg, 698 umol) in toluene (5 mL) were added Pd2(dba)3(31.9 mg, 34.9 umol), t-BuONa (50.3 mg, 523 umol, 1.5 eq) and BINAP (21.7 mg, 34.9 umol), and the mixture was stirred at 95° C. for 4 hours to give a yellow mixture. LCMS showed the reaction was not completed. The mixture was stirred at 95° C. for another 6 hours to give a red mixture. TLC showed the reaction was completed. The mixture was partitioned between EtOAc (15 ml) and water (10 mL). The aqueous phase was extracted with EtOAc (10 mL×2). The combined organic extract was washed with brine (130 mL), dried over Na2SO4, filtered, concentrated under reduced pressure to give crude product. The crude product was purified by combi flash to afford compound G38 (130 mg) as a yellow solid. Procedure for Synthesis of G39 The compound G38 (130 mg) was followed the same procedure of compound B7 to obtain 110 mg of compound G39 as a yellow solid. Procedure for Synthesis of G40 The compound G39 (100 mg) was followed the same procedure of compound G31 to obtain 70 mg of compound G40 as a yellow solid. Procedure for Synthesis of Compound 81 The compound G40 (70 mg) was followed the same procedure of compound 79 to obtain 18 mg of compound 81 as a yellow solid. Exceptional Synthetic Route Procedure for Synthesis of H2 To a mixture of compound H1 (214 mg, 1.75 mmol), DIPEA (1 mL) in MeCN (10 mL) was added compound G10 (500 mg, 1.17 mmol). The reaction solution was stirred at 20° C. for 1 hour to give a yellow mixture. LCMS showed the reaction was completed. The reaction mixture was partitioned with DCM (100 mL) and water (80 mL). The aqueous phase was extracted with DCM (80 mL×2). The combined extract was washed with brine (80 mL×2), dried over anhydrous Na2SO4and filtrated, then concentrated under reduced pressure to give crude product as yellow oil, which was purified by Combi flash to give compound H2 (500 mg) as a yellow gum. Procedure for Synthesis of H4 To a mixture of compound H2 (200 mg, 0.822 mmol) in DMF (5 mL) was added HATU (391 mg, 1.03 mmol), TEA (139 mg, 1.37 mmol), and stirred at 20° C. for 30 minutes. Then compound H3 (352 mg, 0.685 mmol) was added to the mixture, and stirred at 60° C. under N2atmosphere for 5 hours to give a yellow mixture. LCMS showed the reaction was completed. The mixture cooled to temperature and poured into water (50 mL). The yellow solid precipitated out from the mixture. The mixture was filtrated and the filter cake was washed with water (50 mL) to give a crude product as a yellow powder, which was purified by Combi flash to give compound H4 (302 mg) as a yellow gum. Procedure for Synthesis of H5 The compound H4 (300 mg) was followed the same procedure of A7 to obtain 290 mg of compound H5 as a yellow gum. Procedure for Synthesis of H6 The compound H5 (200 mg) was followed the same procedure of A6 to obtain 101 mg of compound H6 as a yellow oil. Procedure for Synthesis of Compound 22 To a mixture of compound H6 (100 mg, 0.133 mmol) in HBr/HOAc (2 mL, 35%) was stirred at 20° C. for 1 hour to give a yellow mixture. LCMS showed the reaction was completed. To the reaction mixture was added MTBE (10 mL) to precipitate an off-white powder. The white powder was collected by filtration and washed with MTBE (5 mL×2), which was purified by prep-HPLC (0.1% TFA) and then basified by cation exchange resin eluting with 5% NH3·H2O/MeOH to give compound 22 (15 mg) as a white powder. Procedure for Synthesis of H8 To a mixture of compound H2 (207 mg, 0.972 mmol), compound H7 (500 mg, 0.972 mmol) in DCM (8 mL) was added AcOH (58.3 mg, 0.972 mmol). The mixture was stirred at 20° C. for 1 hour and then added NaBH(OAc)3(309 mg, 1.46 mmol) to the mixture, and stirred at 20° C. for 24 hours. LCMS show desired MS value. The mixture was partitioned between DCM (50 mL) and saturated NaHCO3(50 mL). The DCM phase was washed with NaHCO3(50 mL×2), and dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure to give yellow oil, which was purified by Combi flash to give compound H8 (200 mg) as a yellow oil. Procedure for Synthesis of H9 To a mixture of compound H8 (200 mg, 0.281 mmol) in DCM (5 mL) was added TFA (1 mL). The mixture was stirred at 20° C. for 2 hours to give a yellow mixture. LCMS showed the reaction was completed. The mixture was combined and concentrated under reduced pressure to give compound H9 (200 mg) as a yellow oil Procedure for Synthesis of H10 To a mixture of compound H9 (200 mg, 0.276 mmol) in THF (5 mL) was added DIPEA (178 mg, 1.38 mmol), compound A5 (152 mg, 0.827 mmol). The yellow mixture was stirred at 25° C. for 1 hour and become black. LCMS showed the reaction was completed. Dimethylamine (2 M, 689 uL) was added to the mixture and stirred at 25° C. for 1 hour to give a brown mixture. LCMS showed desired MS value. The mixture was partitioned between DCM (50 mL) and water (30 mL). The aqueous phase was extracted with DCM (50 mL×2). The combined extracted phase was washed with brine (30 mL), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure to give brown oil, which was purified by Combi flash to give compound H10 (180 mg) as brown oil. Procedure for Synthesis of Compound 21 To a mixture of compound H10 (180 mg, 0.249 mmol) in HBr/HOAc (2.00 mL, 35% purity) was stirred at 20° C. for 1 hour to give a yellow mixture. TLC showed the reaction was completed. To the reaction mixture was added MTBE (10 mL) to precipitate an off-white powder. The white powder was collected by filtration and washed with MTBE (5 mL×2). The crude product was purified by prep-HPLC to give compound 21 (16.2 mg) as a white powder. Procedure for Synthesis of H12 A mixture of compound H11 (500 mg, 2.06 mmol) and DIPEA (400 mg, 3.09 mmol) in NMP (5 mL) was added (4-methoxyphenyl)methanamine (475 mg, 3.46 mmol). The reaction mixture was stirred at 135° C. for 24 hours under N2atmosphere. The mixture was cooled to room temperature, water (50 mL) was added to the reaction mixture and extracted with MTBE (30 mL×3), the combined organic phase was washed with water (20 mL) and brine (20 mL), dried with anhydrous Na2SO4and concentrated under reduced pressure to give the residue. The residue was purified by silica gel column to give compound H12 (910 mg) as a red solid. Procedure for Synthesis of H13 To a solution of compound H12 (500 mg, 1.46 mmol) in DMA (5 mL) was added Zn(CN)2(103 mg, 0.876 mmol), Pd2(dba)3(134 mg, 0.146 mmol), DPPF (162 mg, 0.292 mmol) and Zn (15.28 mg, 0.234 mmol). The reaction was stirred at 150° C. under microwave condition for 0.5 hour under N2atmosphere. LCMS showed the reaction was completed. The mixture was partitioned between with water (30 mL) and EtOAc (30 mL). The aqueous phase was extracted with EtOAc (20 mL). The combined extracts were washed with bine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give compound, which was purified by Combi flash to give compound H13 (300 mg) as a red gum. Procedure for Synthesis of H14 To a mixture of Raney-Ni (100 mg) in MeOH (30 mL) was added compound H13 (500 mg, 1.73 mmol) and NH3·H2O (2.5 mL, 25%). The suspension was degassed under vacuum and purged with H2several times, the mixture was stirred at 25° C. under H2(15 psi) for 3 hours to give a black mixture. TLC showed the reaction was completed. The mixture was filtered, the filtrate was concentrated under reduced pressure to give compound H14 (400 mg) as a yellow gum. Procedure for Synthesis of H15 To a mixture of compound H14 (90.0 mg, 0.307 mmol), DIPEA (165 mg, 1.28 mmol) in MeCN (1 mL) was added compound G10 (110 mg, 256 mmol). The reaction solution was stirred at 25° C. for 1 hour to give a yellow mixture. LCMS showed the reaction was completed. The mixture was partitioned with DCM (30 mL) and water (20 mL). The aqueous phase was extracted with DCM (30 mL×2). The combined extract was washed with brine (30 mL×2), dried over anhydrous Na2SO4and filtrated, then concentrated under reduced pressure to give crude product as yellow oil. The crude product was purified by Combi flash to give compound H15 (100 mg) as a yellow gum. Procedure for Synthesis of H16 To a mixture of compound H15 (100 mg, 0.146 mmol) in TFA (1 mL) was stirred at 25° C. for 3 hours, LCMS showed a lot of starting material was still remained, and heated to 60° C. for 12 hours LCMS showed desired MS value, then heated to 80° C. for 14 hours to give a yellow mixture. LCMS showed the reaction was completed. The mixture was partitioned with DCM (50 mL) and saturated solution of NaHCO3(30 mL). The aqueous phase was extracted with DCM (50 mL×2). The combined extract was washed with brine (30 mL×2), dried over anhydrous Na2SO4and filtrated, then concentrated under reduced pressure to give compound H16 (100 mg) as a yellow gum. Procedure for Synthesis of H17 To a solution of compound H16 (100 mg, crude) and di-tert-butyl dicarbonate (55.6 mg, 0.255 mmol) in DCM (1 mL) was added DIPEA (32.9 mg, 0.255 mmol). The resulting mixture was stirred at 25° C. for 2 hours to give colorless solution. LCMS showed the reaction was completed. The mixture was concentrated under reduced pressure to give compound H17 (102 mg) as a yellow gum. Procedure for Synthesis of H18 To a mixture of compound H17 (102 mg) in THF (2 mL) was added DIPEA (248 mg, 1.92 mmol) and compound A5 (106 mg, 0.576 mmol), the mixture was stirred at 25° C. for 1 hour. LCMS showed the reaction was completed, dimethylamine (2M, 383 uL) was added to the mixture. The mixture was stirred at 25° C. for 30 hours to give a brown mixture. LCMS showed the reaction was completed. The mixture was partitioned between DCM (50 mL) and water (30 mL), The aqueous was extracted with DCM (50 mL×2), the combined extracted phase was washed with brine (30 mL), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure to give a brown oil, which was purified by Combi flash to give compound H18 (80 mg) as a yellow oil. Procedure for Synthesis of Compound 12 To a mixture of compound H18 (80 mg, 0.083 mmol) in DCM (2 mL) was added TFA (770 mg, 0.5 mL), the mixture was stirred at 25° C. for 1 hour to give a yellow mixture. LCMS showed the reaction was completed. The mixture was concentrated under reduced pressure to give yellow oil, which was purified by prep-HPLC to give compound 12 (6.4 mg) as a white powder. Procedure for Synthesis of H20 To a mixture of compound G7 (650 mg, 2.54 mmol) and DIPEA (1.64 g, 12.7 mmol) in NMP (12 mL) was added compound H19 (488 mg, 3.80 mmol). The reaction mixture was stirred at 140° C. for 28 hours under N2atmosphere to give a brown solution. LCMS showed the reaction was completed. The reaction was conducted two pots in parallel. The reaction mixture was combined and concentrated under reduced pressure to give crude product as a brown gum, which was washed with MeOH (15 mL) to give compound H20 (1.09 g) as a yellow powder. Procedure for Synthesis of H21 To a mixture of compound H20 (400 mg, 1.31 mmol) in POCl3(5 mL) was added N, N-diethylaniline (588 mg, 3.94 mmol). The mixture was stirred at 90° C. for 1 hour to give a yellow mixture. The mixture was concentrated under reduced pressure to give compound H21 (420 mg) as a brown oil. Procedure for Synthesis of Compound 14 To a mixture of compound H21 (300 mg, 0.929 mmol), DIPEA (2.40 g, 18.6 mmol) in CH3CN (5 mL) was added compound A16 (533 mg, 1.39 mmol). The reaction solution was stirred at 25° C. for 1 hour to give a yellow mixture. LCMS showed the reaction was completed. The mixture was portioned between DCM (80 mL) and water (50 mL), the aqueous phase was extracted by DCM (80 mL×2), the combined extracted phase was washed with brine (80 mL×2), dried over anhydrous Na2SO4, filtered, concentrated under reduced pressure to give a yellow oil. Crude product was purified by Combi flash and then by prep-TLC. The product was lyophilized to give compound 14 (45.5 mg) as a yellow powder. Procedure for Synthesis of H23 To a solution of compound G26 (500 mg, 966.07 umol) and compound H22 (271 mg, 1.93 mmol) in DMF (10 mL) was added Cs2CO3(630 mg, 1.93 mmol). The resulting mixture was heated at 30-40° C. and stirred for 16 hr to give yellow solution. LCMS showed the reaction was completed. The mixture was filtered and was partitioned between EtOAc (50 mL). The aqueous layer was extracted with EtOAc (50 mL*3). The organic layer was washed with saturated brine (50 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give the crude product as a yellow oil. The crude product was purified by combi flash to give compound H23 (490 mg) as a yellow oil. Procedure for Synthesis of H24 The compound H23 (490 mg) was followed the same procedure of B7 to obtain 440 mg of compound H24 as a yellow powder. Procedure for Synthesis of H25 The compound H24 (390 mg) was followed the same procedure of E3 to obtain 310 mg of compound H25 as a yellow powder. Procedure for Synthesis of Compound 41 The compound H25 (150 mg) was followed the same procedure of compound 44 to obtain 23 mg of compound 41 as a yellow powder. Procedure for Synthesis of H28 To a solution of compound H26 (500 mg, 2.27 mmol), compound H27 (638 mg, 3.41 mmol) and PPh3(893 mg, 3.41 mmol) in THF (10 mL) was added DIAD (689 mg, 3.41 mmol, 662 u) at 0° C. The resulting mixture was stirred at 15° C. for 24 hr to give yellow solution. TLC showed most of the starting material consumed. The reaction mixture was quenched by addition H2O (30 mL) and extracted with EtOAc (30 mL×2). The combined organic layers were washed with brine (10 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography to give compound H28 (570 mg) as yellow oil. Procedure for Synthesis of H29 To a mixture of compound G26 (1 g, 1.93 mmol) in dioxane (10 mL) and H2O (10 mL) was added NaOH (309 mg, 7.73 mmol) and the mixture was stirred at 50° C. for 12 hours to give a yellow mixture. LCMS showed the desired product was observed. The mixture was concentrated under reduced pressure and adjusted to pH=3-4 and the mixture was filtered, and collected the white solid to afford compound H29 (620 mg) as a yellow solid. Procedure for Synthesis of H30 To a mixture of compound H29 (500 mg, 1.10 mmol) in toluene (10 mL) were added POBr3(944 mg, 3.29 mmol, 334 uL) and N,N-diethylaniline (16.3 mg, 109 umol, 17.56 uL) at 15° C., the mixture was stirred at 100° C. for 3 hours to give a yellow mixture. LCMS showed the desired product was observed. The mixture was concentrated under pressure to give the crude product. The crude product was triturated with (PE/EA=3/1) at 15° C. for 30 min to afford compound H30 (330 mg) as a black brown solid. Procedure for Synthesis of H31 To a mixture of compound H30 (330 mg, 636 umol) and compound H28 (247 mg, 636 umol) in dioxane (5 mL) and H2O (2 mL) were added Na2CO3(134 mg, 1.27 mmol, 2 eq) and Pd(dppf)Cl2(46.58 mg, 63.66 umol) and the mixture was stirred at 80° C. for 16 hours under N2to give a black mixture. LCMS showed the desired product was observed. The mixture was partitioned between EtOAc (15 ml) and water (10 mL). The aqueous phase was extracted with EtOAc (10 mL×2). The combined organic extract was washed with brine (10 mL), dried over Na2SO4, filtered, concentrated under reduced pressure to give crude product. The crude product was purified by combi flash (PE/EA=1/1) to afford compound H31 (50 mg) as a red solid. Procedure for Synthesis of H32 The compound H31 (50 mg) was followed the same procedure of compound B7 to obtain 43 mg of compound H32 as a yellow powder. Procedure for Synthesis of H33 The compound H32 (11.5 mg) was followed the same procedure of compound G31 to obtain 10 mg of compound H33 as a yellow powder. Procedure for Synthesis of Compound 87 The compound H32 (10 mg) was followed the same procedure of compound A7 to obtain 3 mg of compound 87 as a yellow powder. REFERENCES D. B. Bregman, R. G. Pestell and V. J. Kidd. Cell cycle regulation and RNA polymerase II.Front Biosci.2000 February; 1(5): D244-57.D. Desai, H. C. Wessling, R. P. Fisher, and D. O. Morgan. Effects of phosphorylation by CAK on cyclin binding by CDCl2and CDK2.Mol. Cell Biol.1995 January; 15(1): 345-350.S. Akhtar, M. Heidemann, J. R. Tietjen, D. W. Zhang, R. D. Chapman, D. Eick, and A. Z. Ansari. TFIIH Kinase Places Bivalent Marks on the Carboxy-Terminal Domain of RNA Polymerase II.Mol. Cell.2009 May; 15; 34(3):387-93.S. Larochelle, R. Amat, K. G. Cutter, M. Sank), C. Zhang, J. J. Allen, K. M. Shokat, D. L. Bentley and R. P. Fisher. Cyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase II.Nat. Struct. Mol. Biol.2012 November; 19(11):1108-15.G. I. Shapiro. Cyclin-Dependent Kinase Pathways as Targets for Cancer Treatment.J. Clin. Oncol.2006 April; 10; 24(11):1770-83.G. Lolli and L. N. Johnson. CAK-Cyclin-dependent Activating Kinase: a key kinase in cell cycle control and a target for drugs?Cell Cycle.2005 April; 4(4):572-7.T. I. Lee and R. A. Young. Transcriptional Regulation and its misregulation in Disease. Cell. 2013 March; 14; 152(6):1237-51.S. Nekhai, M. Zhou, A. Fernandez, W. S. Lane, Ned J. C. Lamb, J. Brady, A. Kumar.Biochem. J.2002 June; 15; 364(Pt 3):649-57.Y. K. Kim, C. F. Bourgeois, R. Pearson, M. Tyagi, M. J. West, J. Wong, S. Y. Wu, C. M. Chiang, and J. Karn. Recruitment of TFIIH to the HIV LTR is a rate-limiting step in the emergence of HIV from latency.EMBO. J.2006 August; 9; 25(15): 3596-3604.A. J. Kapasi and D. H. Spector. Inhibition of the Cyclin-Dependent Kinases at the Beginning of Human Cytomegalovirus Infection Specifically Alters the Levels and Localization of the RNA Polymerase II Carboxyl-Terminal Domain Kinases cdk9 and cdk7 at the Viral Transcriptosome.J. Virol.2008 January; 82(1): 394-407.Eickhoff et al, Pyrazolo-triazine derivatives as selective cyclin-dependent kinase inhibitors. PCT WO2013/128028A1 TABLE 1Enzymatic activity of CDKs (1, 2, 5 and 7) and selectivity of CDK7CDK1/CDK2/CDK5/# cpdsCDK1CDK2CDK5CDK7CDK7*CDK7**CDK7***1CBBABBC2BBBABBC3BBAACCC5CBBABBC6CCBAAAC7CCCAAAB8CCBAABB10CBBABBC11CCCAAAA12CCBAABC14CCBAABB15CBBAABB16CBBABBC18CBBAABB19BAAACCC20CCBABAB21CCBAABB22CBBAABC23CCBABCC24BBBABCC25BBAABCC26CCCAAAA27CCCABBB28CCCBBBB29CCCBBBB30CCCBBBB31CCCAAAA32CCCAAAA33CCCBBBB34CCCAAAA35CCCABBB36CCCAAAA37CCCBBBB38CCCBBBB39CCCAAAB40CCCAAAB41CCCABBB42CCCBBBB43CCCBBBB44CCCAAAA45CCCAAAA46CCCAAAA47CCCAAAB48CCCAAAA49CCCAAAA50CCCAAAA51CCCAAAA52CCCAAAA53CCCAAAA54CCCAAAA55CCCAAAB56CCCAAAA57CCCAAAA58CCCAAAA59CCCAAAA60CCCAAAA61CCCAAAA62CCCAAAA63CCCAAAA64CCCAAAA65CCCAAAA66CCCAAAA67CCCAAAA68CCCAAAA69CCCABBB70CCCAAAA71CCCABBB72CCCAAAA73CCCAAAB74CCBABBB75CCCBBBB76CCCAAAA77CCCAAAA78CCCAAAB79CCCAAAA80CCCAAAA81CCCBBBB82CCCAAAA83CCCAAAA84CCCBBBB85CCCBBBB86CCCCCCC87CCCAAAA88CCCBBBBActivity range: A indicates IC50≤ 100 nM, B indicates 100 < IC50≤ 1,000 nM, C indicates IC50> 1,000 nM*CDK1/CDK7: Selectivity of CDK7 inhibition over CDK1 inhibition [fold]**CDK2/CDK7: Selectivity of CDK7 inhibition over CDK2 inhibition [fold]***CDK5/CDK7: Selectivity of CDK7 inhibition over CDK5 inhibition [fold]Selectivity range: A indicates [fold] >500, B indicates 50 < [fold] ≤ 500, C indicates [fold] ≤50 TABLE 2H460 viability assay# cpdsIC501B2A3A5A6A7A8A9B10A11A12A13A14A15A16A18A19B20A21B22B23B24B25A26A27AActivity range: A indicates IC50≤ 1 uM, B indicates 1 < IC50≤ 10 uM, C indicates IC50> 10 uM TABLE 3MV4-11 viability assay# cpdsIC503A11A20A25A26A27A28A29A30A31A32A33A34A35A36A37A38A39A40A41A42A43A44A45A46A47A48A49A50A51A52A53A54A55A56A57A58A59A60A61A62A63A64A65A66A67A68A69A70B71A72A73A74A75A76A77A78A79AActivity range: A indicates IC50≤ 1 uM, B indicates 1 < IC50≤ 10 uM, C indicates IC50> 10 uM TABLE 4A2780 viability assay# cpdsIC503A11A20A25A26A27A28A29A30A31A32A33A34A35A36A37A38A39A40A41A42A43A44A45A46A47A48A49A50A51A52A53A54A55A56A57A58B59A60A61A62A63B64A65A66A67A68A69A70B71A72A73B74A75A76A77BActivity range: A indicates IC50≤ 1 uM, B indicates 1 < IC50≤ 10 uM, C indicates IC50> 10 uM TABLE 5OVCAR-3 viability assay# cpdsIC503A11A20A25A26A27A28A29A30A31A32A33A34A35A36A37A38A39A40A41A42A43A44A45A46A47A48A49A50A51A52A53A54A55A56A57A58B59A60A61A62A63B64A65A66A67A68A69A70B71A72A73B74A75A76A77BActivity range: A indicates IC50≤ 1 uM, B indicates 1 < IC50≤ 10 uM, C indicates IC50> 10 uM TABLE 6Summarized compounds 1-88 in terms of their structures and corresponding characteristics#cpdsStructureCharacterization Data1white powder;1H-NMR (DMSO-d6, 400 MHz): δ 10.28 (1H, brs), 10.25 (1H, brs), 8.86 (1H, brs), 8.13 (1H, s), 7.88 (1H, d, J = 8.0 Hz), 7.80 (1H, s), 7.72 (1H, s), 7.65 (1H, d, J = 7.6 Hz), 7.60 (1H, d, J = 7.6 Hz), 7.44 (1H, t, J = 8.0 Hz), 7.28 (1H, t, J = 7.6 Hz), 7.12 (1H, d, J = 7.2 Hz), 6.75 (1H, td, J = 15.6, 6.0 Hz,), 6.28 (1H, d, J = 15.6 Hz), 4.59 (2H, s), 4.42-4.54 (2H, m), 3.06 (2H, d, J = 5.2 Hz), 2.85- 2.98 (3H, m), 2.65-2.75 (1H, m), 2.18 (6H, s), 1.64-1.76 (2H, m), 1.23 (6H, d, J = 7.2 Hz), 1.03-1.15 (2H, m); LCMS: 95.8%, MS (ESI): m/z 611.3[M + H]+.2white powder;1H-NMR (DMSO-d6, 400 MHz): δ 10.12 (1H, brs), 8.85 (1H, brs), 7.70 (1H, s), 7.36-7.42 (2H, m), 7.24-7.36 (2H, m), 7.17 (1H, d, J = 7.6 Hz), 7.07 (1H, s), 6.90 (1H, dd, J = 8.0, 1.6 Hz), 6.63-6.74 (2H, m), 6.21 (1H, d, J = 15.2 Hz), 4.55 (2H, s), 4.39-4.50 (2H, m), 2.99-3.08 (2H, m), 2.81-2.97 (3H, m), 2.66-2.77 (1H, m), 2.16 (6H, s), 1.61-1.76 (2H, m), 1.22 (6H, d, J = 7.2 Hz), 0.99-1.13 (2H, m); LCMS: 100%, MS (ESI): m/z 584.2[M + H]+.3White powder;1H-NMR (DMSO-d6, 400 MHz): δ 8.65 (1H, brs), 7.89 (1H, brs), 7.62 (1H, s), 7.48-7.59 (3H, m), 7.31-7.43 (2H, m), 7.16-7.22 (1H, m), 7.09-7.15 (1H, m), 6.95-7.06 (1H, m), 6.61 (1H, t, J = 5.6 Hz), 6.56 (1H, s), 6.21 (1H, d, J = 15.2 Hz), 4.83 (2H, d, J = 5.2 Hz), 4.55-4.65 (2H, m), 3.10-3.20 (2H, m), 2.98- 3.08 (1H, m), 2.80-2.89 (3H, m), 2.30 (6H, s), 1.77-1.79 (2H, m), 1.28-1.32 (8H, m); LCMS: 100%, MS (ESI): m/z 629.4[M + Na]+4white powder;1H-NMR (CDCl3, 400 MHz): δ 7.56 (1H, s), 7.42-7.48 (2H, m), 7.26-7.32 (1H, m), 7.17-7.24 (1H, m), 7.14 (1H, brs), 7.07-7.12 (1H, m), 6.88-6.98 (2H, m), 6.60- 6.70 (2H, m), 6.07 (1H, d, J = 15.2 Hz), 4.77 (2H, d, J = 6.0 Hz), 4.68-4.76 (2H, m), 3.09 (2H, d, J = 5.6 Hz), 2.96-3.04 (1H, m), 2.86-2.96 (3H, m), 2.26 (6H, s), 1.76-1.90 (2H, m), 1.23-1.34 (8H, m); LCMS: 100%, MS (ESI): m/z 584.4[M + H]+.5white powder;1H-NMR (CDCl3, 400 MHz): δ 7.79 (1H, brs), 7.60 (1H, s), 7.40-7.50 (1H, m), 7.26-7.33 (2H, m), 7.19 (1H, s), 7.05-7.12 (2H, m), 6.93 (1H, d, J = 7.6 Hz), 6.75 (1H, d, J = 7.6 Hz), 6.71 (1H, brs), 6.41 (1H, d, J = 16.4 Hz), 6.24-6.34 (1H, m), 5.74 (1H, d, J = 10.0 Hz), 4.62-4.75 (4H, m), 2.96- 3.06 (1H, m), 2.83-2.95 (3H, m), 1.80-1.84 (2H, m), 1.21- 1.33 (8H, m); LCMS: 100%, MS (ESI): m/z 527.3[M + H]+.6white powder;1H-NMR (DMSO-d6, 400 MHz): δ 7.56 (1H, s), 7.33 (1H, d, J = 8.0 Hz), 7.24-7.28 (1H, m), 6.92 (1H, d, J = 6.8 Hz), 6.80-6.88 (2H, m), 6.63 (1H, t, J = 4.8 Hz), 6.53 (1H, d, J = 16.0 Hz), 4.76-4.83 (2H, m), 4.63-4.70 (2H, m), 3.65-3.80 (3H, m), 3.53-3.56 (1H, m), 3.21-3.25 (2H, m), 3.09-3.14 (1H, m), 2.91-3.03 (3H, m), 2.35 (6H, s), 1.94-1.97 (2H, m), 1.86-1.90 (4H, m), 1.45-1.48 (2H, m), 1.27 (6H, d, J = 7.2 Hz); LCMS: 100%, MS (ESI): m/z 576.3[M + H]+.7white powder;1H-NMR (CDCl3, 400 MHz): δ 7.49 (1H, s), 7.38-7.40 (2H, m), 7.14-7.22 (3H, m), 7.04-7.06 (2H, m), 6.87 (1H, d, J = 8.0 Hz), 6.58- 6.59 (1H, m), 6.53 (1H, t, J = 4.8 Hz), 6.33-6.37 (1H, d, J = 16.8 Hz), 6.12-6.14 (1H, m), 5.69 (1H, d, J = 10.8 Hz), 4.64- 4.71 (4H, m), 2.82-2.95 (4H, m), 1.79-1.82 (2H, m), 1.19-1.27 (8H, m); LCMS: 91.6%, MS (ESI): m/z 527.3[M + H]+.8white powder;1H-NMR (DMSO-d6, 400 MHz): δ 10.26-10.27 (1H, s), 10.12-10.14 (1H, s), 8.85-8.69 (1H, brs), 8.21 (1H, s), 7.92 (1H, d, J = 8.0 Hz), 7.72 (1H, s), 7.65-7.68 (1H, m), 7.37-7.49 (3H, m), 7.27-7.31 (1H, m), 7.19-7.23 (1H, m), 6.73-6.76 (1H, m), 6.28 (1H, d, J = 15.6 Hz), 4.63 (2H, s), 4.42-4.47 (2H, m), 3.06 (2H, d, J = 5.2 Hz), 2.82-2.94 (3H, m), 2.70-2.77 (1H, m), 2.17 (6H, s), 1.64-1.69 (2H, m), 1.22 (6H, d, J = 7.2 Hz), 1.02-1.11 (2H, m); LCMS: 100.0%, MS (ESI): m/z 611.3[M + H]+.9white powder; 1H-NMR (CDCl3, 400 MHz): δ 8.47-8.49 (2H, s), 7.97-7.99 (1H, m), 7.61-7.63 (1H, m), 7.55-7.58 (1H, m), 7.52 (1H, s), 7.38-7.49 (4H, m), 7.22-7.25 (1H, m), 7.04-7.11 (1H, m), 6.63-6.65 (1H, m), 6.35-6.40 (1H, m), 4.72-4.77 (1H, m), 4.78-4.55 (2H, m), 4.26-4.30 (1H, m), 3.19-3.21 (2H, m), 2.92-3.02 (2H, m), 2.76-2.83 (2H, m), 2.35 (6H, s), 1.79-7.82 (2H, m), 1.14-1.29 (8H, m); LCMS: 95.3%, MS (ESI): m/z 619.4[M + H]+.10white powder;1H-NMR (CDCl3, 400 MHz): δ 7.60 (1H, s), 7.44-7.53 (4H, m), 7.22-7.26 (1H, m), 7.08 (1H, t, J = 7.6 Hz), 6.93-7.00 (3H, m), 6.86 (1H, d, J = 7.6 Hz), 6.71 (1H, t, J = 6.2 Hz), 6.13 (1H, d, J = 15.6 Hz), 4.79 (2H, d, J = 6.4 Hz), 4.71-4.75 (2H, m), 3.12-3.15 (2H, m), 2.99-3.06 (1H, m), 2.89-2.96 (3H, m), 2.30 (6H, s), 1.86-1.89 (2H, m), 1.24-1.33 (8H, m); LCMS: 100.0%, MS (ESI): m/z 584.4[M + H]+.11white powder;1H-NMR (DMSO-d6, 400 MHz): δ 10.51 (1H, brs), 8.44-8.50 (3H, m), 7.75 (1H, d, J = 5.6 Hz), 7.64 (1H, s), 7.55-7.60 (1H, m), 7.46-7.53 (3H, m), 7.42 (1H, t, J = 8.0 Hz), 7.33 (1H, d, J = 7.2 Hz), 6.78-6.85 (1H, m), 6.36 (1H, d, J = 15.2 Hz), 4.25-4.43 (4H, m), 3.08 (2H, d, J = 5.2 Hz), 2.72-2.86 (4H, m), 2.19 (6H, s), 1.60-1.70 (2H, m), 1.19 (6H, d, J = 7.2 Hz), 1.05-1.10 (2H, m); LCMS: 98.9%, MS (ESI): m/z 619.4[M + H]+.12white powder;1H-NMR (CDCl3, 400 MHz): δ 9.36 (1H, brs), 8.51 (1H, d, J = 9.2 Hz), 8.14-8.17 (2H, m), 7.70 (1H, d, J = 6.8 Hz), 7.66 (1H, d, J = 6.8 Hz), 7.60 (1H, s), 7.34-7.38 (1H, m), 7.02-7.11 (1H, m), 6.32 (1H, d, J = 16.0 Hz), 5.16 (2H, d, J = 6.0 Hz), 4.78-4.84 (2H, m), 3.18 (2H, d, J = 6.4 Hz), 2.94-3.03 (4H, m), 2.33 (6H, s), 1.92-1.97 (2H, m), 1.30-1.45 (2H, m), 1.25 (6H, d, J = 6.8 Hz); LCMS: 95.9%, MS (ESI): m/z 565.3[M + Na]+.13white powder;1HNMR (DMSO-d6, 400 MHz): δ 9.93 (1H, brs), 8.89 (1H, brs), 7.81- 7.83 (1H, m), 7.72 (1H, s), 7.23-7.31 (2H, m), 7.15 (1H, d, J = 8.0 Hz), 7.06 (1H, s), 6.86 (1H, dd, J = 7.6, 2.0 Hz), 6.69-6.75 (2H, m), 6.46 (1H, d, J = 15.6), 4.50-4.55 (4H, m), 3.03 (2H, d, J = 4.8 Hz) 2.84-2.93 (4H, m), 2.16 (6H, s), 1.75-1.80 (2H, m), 1.18- 1.23 (8H, m); LCMS: 100%, MS (ESI): m/z 624.3[M + Na]+.14yellow powder;1H-NMR (CDCl3, 400 MHz): δ 7.74 (1H, brs), 7.60 (1H, s), 7.41 (1H, d, J = 8.0 Hz), 7.23-7.31 (3H, m), 7.05-7.13 (2H, m), 6.93 (1H, dd, J = 8.4, 1.6 Hz), 6.75 (1H, dd, J = 8.4, 1.6 Hz), 6.69 (1H, t, J = 5.6 Hz), 6.42 (1H, dd, J = 16.8, 1.6 Hz), 6.21-6.31 (1H, m), 5.75 (1H, d, J = 10.4), 4.78-4.88 (2H, m), 4.68 (2H, d, J = 6.0 Hz), 2.98-3.09 (1H, m), 2.72-2.85 (2H, m), 2.39-2.49 (1H, m), 2.31 (6H, s), 1.82-1.88 (2H, m), 1.34-1.47 (2H, m), 1.28 (6H, d, J = 6.8 Hz); LCMS: 94.2%, MS (ESI): m/z 55.3[M + H]+.15yellow powder;1H-NMR (CDCl3, 400 MHz): δ 8.18 (1H, s), 7.80 (1H, brs), 7.62 (1H, s), 7.27-7.32 (2H, m), 7.07-7.11 (2H, m), 6.92-7.05 (2H, m), 6.69-6.74 (2H, m), 6.24 (1H, d, J = 15.2 Hz), 4.67-4.76 (4H, m), 3.20-3.21 (2H, m), 3.01-3.04 (2H, m), 2.88-2.94 (2H, m), 2.33 (6H, s), 1.98-2.01 (2H, m), 1.36- 1.45 (2H, m), 1.28 (6H, d, J = 6.8 Hz); LCMS: 95.4%, MS (ESI): m/z 618.3[M + H]+.16yellowpowder;1H-NMR (CDCl3, 400 MHz): δ 9.22 (1H brs), 8.59 (1H, t, J = 5.8 Hz), 8.42 (1H, d, J = 7.6 Hz), 8.19 (1H, s), 7.70 (1H, s), 7.66 (1H, d, J = 6.8 Hz), 7.40-7.47 (4H, m), 7.16-7.20 (1H, m), 7.04-7.08 (1H, m), 6.61 (1H, d, J = 15.2 Hz), 4.83-4.88 (2H, m), 4.58 (2H, d, J = 6.0 Hz), 3.38-3.40 (1H, m), 3.27 (2H, d, J = 6.4 Hz), 2.98-3.05 (3H, m), 2.56 (6H, s), 2.21-2.38 (2H, m), 1.65-1.68 (2H, m), 1.25-1.29 (8H, m); LCMS: 95.6%, MS (ESI): m/z 608.3[M + H]+.17white powder;1H-NMR (DMSO-d6, 400 MHz): δ 11.13 (1H, brs), 10.76 (1H, brs), 9.08 (1H, brs), 8.24 (1H, d, J = 6.0 Hz), 8.00-8.22 (3H, m), 7.79 (1H, s), 7.73 (1H, s), 7.43 (1H, d, J = 8.0 Hz), 7.33 (1H, d, J = 7.2 Hz), 7.23 (1H, s), 7.05-7.15 (1H, m), 6.80-6.90 (1H, m), 6.73 (1H, d, J = 3.6 Hz), 6.56 (1H, d, J = 16.0 Hz), 4.55-4.70 (4H, m), 3.85-3.95 (2H, m), 2.85-3.00 (4H, m), 2.74 (6H, d, J = 3.6 Hz), 1.85-2.00 (2H, m), 1.30-1.50 (2H, m), 1.22 (6H, d, J = 6.4 Hz); LCMS: 100%, MS (ESI): m/z 607.3 [M + Na]+18Racemic mixture; white powder;1H-NMR (CDCl3, 400 MHz): δ 7.61 (1H, s), 7.24 (1H, t, J = 7.8 Hz), 6.88-6.96 (3H, m), 6.78- 6.81 (1H, m), 6.60-6.65 (1H, m), 6.40-6.45 (1H, m), 4.69-4.76 (4H, m), 4.48-4.51 (1H, m), 3.83- 3.90 (1H, m), 3.43-3.72 (3H, m), 2.90-3.11 (6H, m), 2.27 (6H, d, J = 5.6 Hz), 1.99-2.08 (4H, m), 1.73-1.91 (4H, m), 1.25-1.37 (8H, m); LCMS: 94.9%, MS (ESI): m/z 612.5[M + Na]+.19white powder;1H-NMR (DMSO-d6, 400 MHz): δ 8.72 (1H, s), 7.87 (1H, s), 7.58 (2H, s), 7.46-7.54 (2H, m), 7.33-7.40 (2H, m), 7.18-7.26 (2H, m), 6.90-7.01 (1H, m), 6.64 (1H, brs), 6.54 (1H, s), 6.13 (1H, d, J = 15.6 Hz), 4.82 (2H, s), 4.55-4.65 (2H, m), 3.11 (2H, d, J = 5.2 Hz), 2.98-3.05 (1H, m), 2.79-2.88 (3H, m), 2.28 (6H, s), 1.70-1.80 (2H, m), 1.29 (8H, d, J = 6.8 Hz); LCMS: 100%, MS (ESI): m/z 629.2 [M + Na]+20yellow powder;1H NMR (DMSO-d6, 400 MHz): δ 10.08 (1H, brs), 8.89 (1H, t, J = 5.6 Hz), 8.69 (1H, d, J = 7.6 Hz), , 8.50 (1H, d, J = 8.4 Hz), 7.88 (1H, d, J = 8.4 Hz), 7.60-7.85 (7H, m), 7.43-7.49 (2H, m), 6.73-6.80 (1H, m), 6.50 (1H, d, J = 16.0 Hz), 5.04 (2H, d, J = 6.4 Hz), 4.30-4.33 (2H, m), 3.11-3.17 (1H, m), 3.00 (2H, d, J = 6.4 Hz), 2.83-2.92 (1H, m), 2.62-2.67 (2H, m), 2.11 (6H, s), 1.71-1.75 (2H, m), 1.07-1.27 (8H, m); LCMS: 100%, MS (ESI): m/z 619.4[M + H]+.21Racemic mixture; white powder;1H-NMR (CDCl3, 400 MHz): δ 7.60 (1H, s), 7.12 (1H, t, J = 7.2 Hz), 6.82-6.93 (1H, m), 6.64-6.69 (1H, m), 6.60-6.63 (1H, m), 6.40-6.52 (3H, m), 4.70-4.80 (2H, m), 4.56-4.70 (2H, m), 3.37-3.83 (6H, m), 2.89-3.16 (6H, m), 2.25-2.31 (6H, m), 1.93-2.01 (3H, m), 1.59-1.76 (2H, m), 1.39-1.48 (2H, m), 1.23-1.33 (8H, m); LCMS: 100%, MS (ESI): m/z 611.5[M + Na]+.22Racemic mixture; white powder;1H-NMR (DMSO-d6, 400 MHz): δ 7.89 (1H, brs), 7.59 (1H, s), 7.42-7.56 (2H, m), 7.21-7.26 (1H, m), 7.06 (1H, t, J = 6.0 Hz), 6.83-6.92 (1H, m), 6.65-6.75 (1H, m), 6.40 (1H, d, J =15.6 Hz), 4.65-4.77 (4H, m), 3.86-4.03 (1H, m), 3.75-3.85 (1H, m), 3.63 (1H, t, J = 5.6 Hz), 3.42-3.53 (1H, m), 3.18- 3.36 (1H, m), 2.88-3.12 (6H, m), 2.32-2.46 (1H, m), 2.20-2.30 (7H, m), 1.83-1.92 (2H, m), 1.54-1.82 (2H, m), 1.19-1.40 (10H, m); LCMS: 100%, MS (ESI): m/z 617.5[M + H]+.23white powder;1H NMR (DMSO-d6, 400 MHz): δ 11.52 (1H, brs), 9.98 (1H, brs), 9.01 (1H, t, J = 5.4 Hz), 8.00 (1H, s), 7.91 (1H, s), 7.77 (1H, s), 7.73 (1H, d, J = 7.2 Hz), 7.39 (1H, t, J = 8.0 Hz), 7.31-7.34 (2H, m), 7.26 (1H, d, J = 8.0 Hz), 6.83 (1H, s), 6.67-6.73 (1H, m), 6.33 (1H, d, J = 15.2 Hz), 4.63-4.67 (4H, m), 3.13-3.19 (3H, m), 2.86-2.94 (3H, m), 2.26 (6H, s), 1.85-1.91 (2H, m), 1.29-1.36 (2H, m), 1.23 (6H, d, J = 6.8 Hz); LCMS: 100%, MS (ESI): m/z 607.3[M + H]+.24brown powder;1H-NMR (DMSO-d6, 400 MHz): δ 10.2 (1 2.72-2.83 (2H, m), 2.17-2.35 (6H, m), 1.81-1.90 (2H, m), 1.14-1.34 (8H, m); LCMS: 96.5%, MS (ESI): m/z = 608.3 [M + H]+.25white powder;1H-NMR (CDCl3, 400 MHz): δ 10.44 (1H, brs), 7.61 (1H, brs), 7.54- 7.63 (3H, m), 7.48 (1H, d, J = 7.6 Hz), 7.32-7.41 (2H, m), 6.93-7.07 (2H, m), 6.79 (1H, d, J = 7.2 Hz), 6.57-6.68 (2H, m), 6.21 (1H, d, J = 14.8 Hz), 4.90 (2H, d, J = 5.6 Hz), 4.58-4.68 (2H, m), 3.12 (2H, d, J = 5.6 Hz), 3.98-3.06 (1H, m), 2.80- 2.91 (3H, m), 2.29 (6H, s), 1.74-1.84 (2H, m), 1.29 (6H, d, J = 6.8 Hz), 1.20-1.26 (2H, m); LCMS: 100%, MS (ESI): m/z 629.3[M + Na]+.26(3R, 4R); pale-yellow powder;1H NMR (400 MHz, DMSO-d6): δ 10.91-11.17 (2H, m), 9.03 (1H, br s), 8.82 (1H, br s), 8.38- 8.58 (3H, m), 7.79-7.89 (1H, m), 7.70-7.77 (1H, m), 7.62-7.67 (1H, m), 7.50-7.57 (2H, m), 7.42-7.49 (1H, m), 7.37 (1H, br d, J = 7.2 Hz), 6.85-6.98 (1H, m), 6.63 (1H, d, J = 15.6 Hz), 5.67-5.96 (1H, 1H), 4.33-4.50 (2H, m), 3.96 (2H, d, J = 6.8 Hz), 3.42-3.57 (2H, m), 2.97- 3.25 (3H, m), 2.81-2.90 (1H, m), 2.77 (6H, s), 2.58-2.69 (1H, m), 1.39-1.90 (3H, m), 1.19 (6H, br d, J = 6.4 Hz); LCMS: 100%, MS (ESI): m/z 649.3[M + H]+27(1R, 4R); yellow powder;1H-NMR (400 MHz, CD3OD): δ 8.67 (1H, s), 8.42 (1H, br d, J = 6.4 Hz), 8.24 (1H, br d, J = 6.4 Hz), 7.84-7.93 (2H, m), 7.78 (1H, d, J = 9.2 Hz), 7.68 (1H, br s), 7.62 (3H, m), 6.95- 7.10 (1H, m), 6.72 (1 H, br d, J = 15.2 Hz), 4.70-5.10 (2H, m), 4.07 (2H, br d, J = 6.8 Hz) 3.47-3.65 (2H, m), 2.91-3.01 (6H, m), 2.77-2.87 (1H, m), 1.87-2.10 (3H, m), 1.65-1.72 (1H, m), 1.30-1.51 (4H, m), 1.12-1.23 (6H, m); LCMS: 100%, MS (ESI): m/z 634.3[M + H]+.28yellow powder;1H NMR (400 MHz, CD3OD): δ 8.42 (1H, s), 8.37 (1H, d, J = 6.0 Hz), 7.68- 7.77 (2H, m), 7.61-7.67 (1H, m), 7.46-7.60 (4H, m), 7.40 (1H, d, J = 7.6 Hz), 6.92-7.02 (1H, m), 6.64 (1 H, d, J = 15.2 Hz), 5.01-5.04 (1H, m), 4.39- 4.43 (1H, m), 4.06 (2H, d, J = 6.4 Hz), 3.65 (2H, t, J = 6.0 Hz), 3.33-3.41 (2H, m), 2.97 (6H, s), 2.82-2.91 (1H, m), 1.74-1.80 (2H, m), 1.15-1.25 (6H, m); LCMS: 100%, MS (ESI): m/z 594.3[M + H]+.29yellow solid;1H-NMR (400 MHz, CD3OD): δ 8.66 (1H, s), 6.42-6.44 (1H, m), 8.21-8.23 (1H, m), 7.80-7.87 (3H, m), 7.59-7.71 (4H, m), 7.00-7.07 (1H, m), 6.74-6.78 (1H, d, J = 15.6 Hz), 4.95-5.02 (1H, m), 4.71-4.81 (1H, m), 4.31- 3.41 (2H, m), 4.08 (2H, d, J = 7.2 Hz), 3.46-3.47 (2H, m), 3.08-3.13 (2H, m), 2.96- 2.97 (6H, m), 1.85-1.97 (2H, m), 1.29-1.38 (2H, m), 1.16- 1.19 (6H, m); LCMS: 100.0%, MS (ESI): 634.3 m/z [(M + H)]+.30Racemic mixture; off-white powder;1H NMR (400 MHz, CD3OD): δ 8.27-8.42 (2H, m) 7.25-7.75 (8H, m), 6.91-7.07 (1H, m), 6.33 (1H, d, J = 15.2 Hz), 4.80-4.87 (1H, m), 4.60- 4.67 (2H, m), 4.28-4.37 (1H, m), 3.47-3.56 (1H, m), 3.26 (2H, d, J = 6.4 Hz), 3.05-3.13 (1H, m), 2.85-2.92 (1H, m), 2.45-2.54 (2H, m), 2.36 (6H, s), 2.01-2.11 (1H, m), 1.65-1.75 (2H, m), 1.15-1.26 (6H, m); LCMS: 95.9%, MS (ESI): m/z 324.2 [M/2 + H]+.31light yellow powder;1H NMR (400 MHz, CD3OD): δ 8.32 (1H, d, J = 6.0 Hz), 8.29 (1H, s), 7.63-7.68 (1H, m), 7.53-7.61 (2H, m), 7.49 (1H, t, J = 6.8 Hz), 7.36-7.40 (2H, m), 7.31- 7.35 (2H, m), 6.91-7.00 (1H, m), 6.38-6.42 (1H, m), 4.34- 4.38 (1H, m), 4.13-4.25 (2H, m), 3.74-3.82 (1H, m), 3.49 (2H, d, J = 6.8 Hz), 2.98-3.13 (2H, m), 2.85 (1H, m), 2.53 (6H, s), 1.81 (2H, m), 1.23- 1.45(3H, m), 1.19 (6H, d, J = 6.8 Hz); LCMS: 100%, MS (ESI): m/z 620.2 [M + H]+.32(1S, 35); light yellow solid;1H-NMR (Me0D, 400 MHz): δ 8.69-8.71 (1H, m), 8.42-8.45 (1H, m), 8.25-8.27 (1H, m), 7.80-7.88 (3H, m), 7.58-7.72 (4H, m), 6.99-7.07 (1H, m), 6.73 (1H, d, J =15.2 Hz), 5.01- 5.06 (1H, m), 4.91-4.94 (1H, m), 4.71-4.78 (1H, m), 4.39- 4.44 (1H, m), 4.26-4.29 (1H, m), 4.07(2H, d, J = 7.2 Hz), 2.96 (6H, s), 2.84-2.88 (1H, m), 2.06-2.32 (2H, m), 1.95-1.99 (1H, m), 1.61-1.80 (3H, m), 1.18-1.21 (6H, m); LCMS: 99.7%, MS (ESI): 620.2 m/z [M + H]+.33(1R, 4R); white powder; 1H NMR (CD3OD, 400 MHz): δ 8.66 (1H, d, J = 2.0 Hz), 8.43 (1H, d, J = 6.8 Hz), 8.23 (1H, d, J = 6.8 Hz), 7.76-7.88 (3H, m), 7.66-7.74 (1H, m), 7.55- 7.63 (3H, m), 6.97-7.08 (1H, m), 6.71 (1H, d, J = 15.6 Hz), 4.89-4.90 (2H, m), 4.07 (2H, d, J = 6.4 Hz), 3.45-3.54 (1H, m), 3.17-3.26 (1H, m), 3.04 (1H, m), 2.96 (6H, s), 2.84 (1H, m), 1.88- 2.05 (2H, m), 1.67-1.83 (2H, m), 1.36-1.49 (1H, m), 1.28-1.35 (1H, m), 1.14-1.21 (7H, m), 0.84-1.11 (2H, m); LCMS: 99.7%, MS (ESI): m/z 648.2 [M + H]+; HPLC (254 nm): 100%.34yellow powder;1H NMR (400 MHz, CD3OD): δ 8.67 (1H, s), 8.38-8.45 (1H, br s), 8.18-8.26 (1H, m), 7.78-7.88 (3H, m), 7.55-7.73 (4H, m), 6.97-7.08 (1H, m), 6.74 (1H, d, J = 15.2 Hz), 4.95-5.03 (1H, m), 4.66- 4.76 (1H, m), 4.48-4.59 (2H, m), 4.07 (2H, d, J = 6.8 Hz), 3.39- 3.54 (1H, m), 3.03-3.24 (3H, m), 2.96 (6H, s), 2.76 (3H, s), 2.14- 2.31 (2H, m), 1.46-1.80 (2H, m), 1.19 (6H, d, J = 6.8 Hz); LCMS: 99.7%, MS (ESI): m/z 633.3 [M + H]+35light yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.69 (1H, s), 8.44 (1H, d, J = 6.4 Hz), 8.23 (1H, d, J = 6.8 Hz), 7.81-7.87 (3H, m), 7.68-7.72 (2H, m), 7.59-7.60 (2H, m), 6.99-7.07 (1H, m), 6.73 (1H, d, J = 15.2 Hz), 5.04-5.08 (1H, m), 4.70-4.74 (1H, m), 4.07 (2H, d, J = 6.4 Hz), 3.41-3.44 (2H, m), 2.96 (6H, s), 2.82- 2.86 (1H, m), 1.69-1.73 (2H, m), 1.28 (6H, s), 1.21 (6H, d, J = 6.8 Hz); LCMS: 98.2%, MS (ESI): 622.3 m/z [(M + H)]+.36White solid;1H NMR (400 MHz, DMSO-d6): δ 11.24- 11.45 (2H, m), 9.42-9.66 (1H, m), 8.61-8.66 (1H, m), 8.41- 8.47 (1H, m), 8.18-8.23 (1H, m), 7.72-7.75 (2H, m), 7.67- 7.69 (1H, m), 7.54-7.60 (1H, m), 7.41-7.52 (2H, m), 6.91- 7.01 (1H, m), 6.65 (1 H, m), 4.74-4.83 (1H, m), 4.50-4.61 (1H, m), 3.95-4.02 (2H, m), 2.72-2.81 (6H, m), 1.76-1.91 (1H, m), 1.44-1.68 (5H, m), 1.21-1.38 (2H, m), 1.10-1.16 (6H, m); LCMS: 100%, MS (ESI): m/z 634.3 [M + H]+.37Racemic mixture; off-white powder;1H NMR (400 MHz, CD3OD): δ 8.64-8.70 (1H, m), 8.40-8.51 (1H, m), 8.17-8.28 (1H, m), 7.81-7.92 (3H, m), 7.68-7.75 (2H, m), 7.58-7.66 (2H, m), 7.01-7.09 (1H, m), 6.77 (1H, d, J = 15.2 Hz), 5.11- 5.20 (1H, m), 4.53-4.72 (2H, m), 4.09 (2H, d, J = 7.2 Hz), 3.38-3.86 (4H, m), 3.08-3.21 (1H, m), 2.93 (6H, s), 2.03- 2.32 (2H, m), 1.14-1.26 (6H, m); HPLC: 100%, MS (ESI): m/z 606.2 [M + H]+ .38Racemic mixture; light yellow solid;1H-NMR (MeOD, 400 MHz): δ 8.67 (1H, s), 8.41-8.43 (1H, m), 8.18-8.23 (1H, m), 7.80-7.87 (3H, m), 7.59-7.68 (4H, m), 6.99-7.07 (1H, m), 6.74 (1H, d, J = 15.2 Hz), 4.95- 4.99 (1H, m), 4.72-4.81 (1H, m), 4.07 (2H, d, J = 7.2 Hz), 3.42- 3.93 (4H, m), 3.09-3.14 (1H, m), 2.96 (6H, s), 2.07-2.22 (1H, m), 1.43-1.98 (5H, m), 1.16-1.19 (6H, m); LCMS: 99.8%, MS (ESI): 634.2 m/z [(M + H)]+39Light yellow solid;1H NMR (400 MHz, CD3OD): δ 8.68 (1H, s), 8.42 (1H, d, J = 6.0 Hz), 8.21 (1H, d, J = 6.4 Hz), 7.82-7.89 (3H, m), 7.65-7.75 (2H, m), 7.59-7.64 (2H, m), 7.00-7.11 (1H, m), 6.74 (1H, d, J = 15.6 Hz), 4.98-5.07 (1H, m), 4.68-4.75 (1H, m), 4.54- 4.62 (2H, m), 4.07 (2H, d, J = 6.8 Hz), 3.59-3.69 (1H, m), 3.08-3.28 (3H, m), 2.98 (6H, s), 2.94 (6H, s), 2.21-2.29 (2H, m), 1.72-1.87 (2H, m), 1.29- 1.33 (2H, m), 1.19-1.24 (6H, m); LCMS: 97.9%, MS (ESI): m/z 647.3 [M + H]+40yellow powder;1H NMR (400 MHz, CD3OD): δ 8.58 (1H, s), 8.38 (1H, s), 8.10-8.19 (1H, m), 7.80-7.90 (2H, m), 7.66-7.74 (2H, m), 7.52-7.63 (3H, m), 6.02 (1H, s), 5.00-5.09 (1H, m), 4.65-4.72 (1H, m), 4.48-4.56 (2H, m), 3.46-3.59 (1H, m), 2.97-3.23 (3H, m), 2.33 (3H, s), 2.10-2.22 (2H, m), 2.01 (3H, s), 1.47-1.76 (2H, m), 1.21 (6H, d, J = 6.8 Hz); HPLC: 96.1%, MS (ESI): m/z 590.2[M + H]+41Yellow powder;1H NMR (CD3OD, 400 MHz): δ 8.51 (1H, s), 8.37 (1H, d, J = 2.8 Hz), 8.11 (1H, d, J = 6.8 Hz), 7.77-7.89 (4H, m), 7.62-7.71 (3H, m), 7.57 (1H, d, J = 6.8 Hz), 6.97-7.05 (1H, m), 6.58- 6.63 (2H, m), 5.03-5.10 (1H, m), 4.76-4.81 (1H, m), 4.74 (2H, s), 4.09 (2H, d, J = 7.2 Hz), 3.08-3.17 (1H, m), 2.99 (6H, s), 1.29-1.34 (6H, m); LCMS: 99.2%, MS (ESI): 617.2 m/z [M + H]+42off-white powder;1H NMR (400 MHz, CD3OD): δ 8.65 (1H, d, J = 1.6 Hz), 8.41 (1H, d, J = 6.8 Hz), 8.21 (1H, d, J = 6.4 Hz), 7.77-7.89 (3H, m), 7.55-7.73 (4H, m), 6.97- 7.12 (1H, m), 6.75 (1 H, d, J = 15.2 Hz), 4.97-5.03 (1H, m), 4.69-4.76 (1H, m), 4.07 (2H, d, J = 6.8 Hz), 3.66-3.85 (8H, m), 3.04-3.16 (1H, m), 2.96 (6 H, s), 1.16-1.21 (6H, m); HPLC: 99.4%, MS (ESI): m/z 606.2 [M + H]+43White powder;1H NMR (CD3OD, 400 MHz): δ 8.70 (1H, s), 8.46 (1H, d, J = 7.2 Hz), 8.22-8.28 (1H, m), 7.82-7.89 (3H, m), 7.57- 7.76 (4H, m), 7.00-7.09 (1H, m), 6.71 (1H, d, J = 15.2 Hz), 4.96-5.02 (1H, m), 4.75-4.83 (1H, m), 4.08 (2H, d, J = 6.4 Hz), 3.84-4.01 (2H, m), 3.32- 3.39 (2H, m), 3.23-3.28 (1H, m), 3.06-3.21 (1H, m), 2.98 (6H, s), 2.83-2.88 (1H, m), 1.73-1.80 (1H, m), 1.55- 1.64 (2H, m), 1.25-1.32 (1H, m), 1.18-1.24 (6H, m); LCMS: 100%, MS (ESI): m/z 634.2[M + H]+44Light yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.88- 6.89 (1H, m), 8.41-8.43 (1H, m), 8.26-8.28 (1H, m), 7.78- 7.88 (3H, m), 7.67-7.71 (1H, m), 7.55-7.60 (3H, m), 6.99- 7.07 (1H, m), 6.71 (1H, d, J = 15.2 Hz), 4.95-4.99 (1H, m), 4.78-4.82 (1H, m), 4.07 (2H, d, J = 7.2 Hz), 3.52-3.57 (1H, m), 3.40 (3H, s), 3.22- 3.28 (1H, m), 2.96 (6H, s), 2.80-2.87 (1H, m), 2.03-2.17 (3H, m), 1.71-1.73 (1H, m), 1.27-1.48 (4H, m), 1.16-1.19 (6H, m); LCMS: 100.0%, MS (ESI): 648.3 m/z [(M + H)]+45Light yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.68 (1H, s), 8.45 (1H, d, J = 6.4 Hz), 8.23 (1H, d, J = 6.8 Hz), 7.82-7.83 (3H, m), 7.68-7.72 (1H, m), 7.59-7.65 (3H, m), 6.99-7.05 (1H, m), 6.72 (1H, d, J = 15.2 Hz), 5.04-5.08 (1H, m), 4.77-4.81 (1H, m), 4.07 (2H, d, J = 6.8 Hz), 3.37-3.48 (3H, m), 3.18-3.27 (1H, m), 2.96-3.02 (8H, m), 2.83-2.88 (1H, m), 1.93-1.96 (m, 3H), 1.49-1.55 (2H, m), 1.18-1.21 (6H, m); LCMS: 100.0%, MS (ESI): 633.3 m/z [(M + H)]+46off-white powder;1H NMR (400 MHz, CD3OD): δ 8.59 (1 H, d, J = 2.0 Hz), 8.38 (1H, d, J = 6.4 Hz), 8.16 (1H, d, J = 6.4 Hz), 7.79-7.88 (2H, m), 7.66-7.78 (2H, m), 7.57-7.62 (2H, m), 7.53 (1H, s), 6.51-6.58 (2H, m), 5.94 (1H, dd, J = 8.0, 3.6 Hz), 4.98-5.09 (1H, m), 4.65-4.74 (1H, m), 4.49-4.57 (2H, m), 3.43-3.54 (1H, m), 2.95-3.21 (3H, m), 2.04-2.21 (2H, m), 1.44-1.76 (2H, m), 1.20 (6H, d, J = 6.8 Hz); HPLC: 100%, MS (ESI): m/z 562.2[M + H]+.47Light yellow powder;1H NMR (CD3OD, 400 MHz): δ 8.43 (1H, s), 8.39 (1H, d, J = 6.4 Hz), 8.17 (1H, d, J = 6.4 Hz), 7.78-7.84 (2H, m), 7.62-7.71 (2H, m), 7.56-7.62 (2H, m), 7.55 (1H, s), 4.91-5.05 (1H, m), 4.63-4.71 (1H, m), 4.47-4.551 (2H, m), 3.43-3.56 (1H, m), 3.10-3.29 (2H, m), 3.00-3.08 (1H, m), 2.02-2.21 (5H, m), 1.63-1.76 (1H, m), 1.51- 1.59 (1H, m), 1.21-1.27 (6H, m); LCMS: 96.8%, MS (ESI): m/z 574.2 [M + H]+48Pale yellow powder;1H NMR (CD3OD, 400 MHz): δ 8.55 (1H, d, J = 2.0 Hz), 8.37-8.42 (1H, m), 8.20 (1H, d, J = 6.4 Hz), 7.77- 7.92 (3H, m), 7.64-7.72 (1H, m), 7.54-7.62 (3H, m), 5.98 (1H, s), 5.71 (1H, d, J = 1.2 Hz), 4.99- 5.07 (1H, m), 4.67-4.73 (1H, m), 4.47-4.54 (1H, m), 3.44-3.56 (1H, m), 3.17-3.24 (1H, m), 3.03-3.09 (1H, m), 2.08-2.21 (2H, m), 2.06 (3H, s), 1.64-1.78 (1H, m), 1.55-1.60 (1H, m), 1.16-1.21 (6H, m); LCMS: 100%, MS (ESI): m/z 576.2 [M + H]+49White powder;1H NMR (CD3OD, 400 MHz): δ 8.48 (1H, d, J = 1.6 Hz), 8.40 (1H, d, J = 6.8 Hz), 8.19 (1H, d, J = 6.8 Hz), 7.80-7.89 (3H, m), 7.63-7.72 (2H, m), 7.55-7.62 (2H, m), 4.98-5.07 (1H, m), 4.68-4.75 (1H, m), 4.51-4.57 (2H, m), 3.46- 3.52 (1H, m), 3.13-3.23 (2H, m), 3.04-3.09 (1H, m), 2.08-2.17 (2H, m), 1.74-1.87 (4H, m), 1.55-1.72 (2H, m), 1.23-1.28 (6H, m); LCMS: 99.8%, MS (ESI):601.2m/z [M + H]+50Yellow powder;1H NMR (CD3OD, 400 MHz): δ 8.57 (1H, d, J = 2.0 Hz), 8.41 (1H, d, J = 6.4 Hz), 8.19 (1H, d, J = 6.8 Hz), 7.73-7.88 (3H, m), 7.66-7.71 (1H, m), 7.56-7.62 (3H, m), 7.06-7.17 (1H, m), 6.25 (1H, dd, J = 14.8, 2.0 Hz), 4.99-5.06 (1H, m), 4.66-4.73 (1H, m), 4.45-4.52 (2H, m), 3.47-3.58 (1H, m), 3.18-3.38 (2H, m, overlap with CD3OD signal), 3.0-3.09 (1H, m), 2.10-2.19 (2H, m), 1.98 (3H, dd, J = 6.8, 1.6 Hz), 1.65-1.80 (1H, m), 1.49-1.64 (1H, m), 1.18 (6H, d, J = 6.8 Hz);LCMS: 100%, MS (ESI): m/z576.3 [M + H]+51Racemic mixture; off-white powder;1H NMR (400 MHz, CD3OD): δ 8.71 (1H, s), 8.48 (1H, d, J = 6.4 Hz), 8.27 (1H, d, J = 6.0 Hz), 7.81-7.92 (3H, m), 7.56-7.75 (4H, m), 7.00- 7.13 (1H, m), 6.77 (1H, d, J = 15.2 Hz), 5.06-5.14 (1H, m), 4.72-4.79 (1H, m), 4.06- 4.21 (3H, m), 3.86-3.97 (2H, m), 3.43-3.59 (2H, m), 3.25-3.33 (3H, m), 3.14-3.26 (1H, m), 2.98 (6H, s), 2.83-2.94 (1H, m), 1.57-1.82 (2H, m), 1.21 (6H, d, J = 6.4 Hz); HPLC: 100%, MS (ESI): m/z 649.3 [M + H]+52yellow powder;1H NMR (400 MHz, CD3OD): δ 8.69 (1H, s), 8.47 (1H, s), 8.26 (1H, d, J = 5.2 Hz), 7.80-7.93 (3H, m), 7.56-7.76 (4H, m), 7.02-7.14 (1H, m), 6.76 (1H, d, J = 14.8 Hz), 5.08-5.18 (1H, m), 4.69- 4.78 (1H, m), 4.47-4.56 (2H, m), 4.03-4.08 (2H, m), 3.48- 3.68 (5H, m), 2.94-3.18 (3H, m), 2.11-2.23 (2H, m), 2.00- 2.07 (2H, m), 1.80-1.92 (4H, m), 1.51-1.67 (2H, m), 1.20 (6H, d, J = 6.0 Hz); HPLC: 97.1%, MS (ESI): m/z 659.3 [M + H]+53yellow powder;1H NMR (400 MHz, CD3OD): δ 8.69 (1H, s), 8.45 (1H, d, J = 5.2 Hz), 8.25 (1H, d, J = 6.4 Hz), 7.81-7.96 (3H, m), 7.59-7.76 (4H, m), 7.02-7.17 (1H, m), 6.79 (1H, d, J = 15.2 Hz), 5.01-5.08 (1H, m), 4.73-4.78 (1H, m), 4.48-4.55 (2H, m), 4.05-4.21 (4H, m), 3.79-3.92 (2H, m), 3.48-3.63 (4H, m), 3.12-3.28 4H, m), 2.11-2.27 (2H, m), 1.56-1.85 (2H, m), 1.20 (6H, d, J = 6.0 Hz); HPLC: 100%, MS (ESI): m/z 661.3[M + H]+54off-white powder;1H NMR (CD3OD, 400 MHz): δ 8.56 (1H, s), 8.31-8.36 (1H, m), 8.08-8.13 (1H, m), 7.77-7.90 (2H, m), 7.42-7.75 (5H, m), 7.06-7.13 (1H, m), 6.46 (1H, d, J = 15.2 Hz), 4.64-4.71 (1H, m), 4.52-4.58 (2H, m), 4.14- 4.23 (2H, m), 3.44-3.53 (4H, m), 2.87-3.19 (3H, m), 2.04- 2.13 (2H, m), 1.29-1.72 (3H,m), 1.20 (6H, d, J = 6.8 Hz);LCMS: 99.8%, MS (ESI):606.2 m/z[M + H]+55yellow powder;1H NMR (400 MHz, DMSO-d6): δ ppm 10.95 (1H, brs) 8.87 (1H, d, J = 4.8 Hz), 8.65-8.72 (1H, m), 8.64 (1H, s), 8.13 (2H, brs), 7.65- 7.75 (2H, m), 7.39-7.57 (3H, m), 7.24-7.38 (3H, m), 6.82- 6.95 (1H, m), 6.62 (1H, d, J = 14.8 Hz), 4.18-4.48 (4H, m), 3.76-3.88 (2H, m), 3.19-3.26 (1H, m), 2.83-2.94 (1H, m), 2.55-2.76 (9H, m), 1.86 (2H, m), 1.28-1.39 (2H, m), 1.20 (6H, d, J = 6.8 Hz); LCMS: 100%, MS (ESI): m/z 619.3 [M + H]+56yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.70- 8.72 (1H, m), 8.29-8.33 (1H, m), 7.78 (1H, s), 7.61-7.72 (3H, m), 7.42-7.48 (2H, m), 7.32-7.34 (1H, m), 6.86-6.92 (1H, m), 6.72-6.75 (1H, m), 4.32-4.36 (2H, m), 4.03 (2H, d, J = 7.2 Hz), 3.45-3.49 (1H, m), 3.02-3.15 (3H, m), 2.94 (6H, s), 2.61 (3H, s), 2.09- 2.12 (2H, m), 1.55-1.61 (2H, m), 1.26 (6H, d, J = 6.8 Hz); LCMS: 100.0%, MS (ESI): 633.3 m/z [(M + H)]+57off-white powder;1H NMR (400 MHz, CD3OD): δ 8.63 (1H, s), 8.32-8.37 (1H, m), 8.14 (1H, d, J = 6.4 Hz), 7.80- 7.87 (2H, m), 7.72-7.76 (1H, m), 7.68 (1H, t, J = 8.0 Hz), 7.54-7.63 (2H, m), 7.48 (1H, s), 6.96-7.15 (1H, m), 6.72 (1H, d, J = 15.20 Hz), 4.66- 4.73 (2H, m), 4.53-4.59 (2H, m), 4.16 (2H, d, J = 6.8 Hz), 3.69-3.74 (2H, m), 3.42-3.53 (1H, m), 2.93-3.25 (5H, m), 2.03-2.32 (6H, m), 1.43-1.69 (2H, m), 1.20 (6H, d, J = 7.2 Hz); HPLC: 100%, MS (ESI): m/z 645.3[M + H]+58yellow powder;1H NMR (400 MHz, CD3OD): δ 8.63 (1H, s), 8.37 (1H, d, J = 6.4 Hz), 8.16 (1H, d, J = 6.2 Hz), 7.73-7.88 (3H, m), 7.50-7.71 (4H, m), 6.95-7.08 (1H, m), 6.72 (1H, d, J = 15.2 Hz), 4.95-5.03 (1H, m), 4.65-4.71 (1H, m), 4.46- 4.54 (2H, m), 3.99-4.19 (4H, m), 3.42-3.50 (1H, m), 3.10- 3.17 (2H, m), 2.91-3.07 (4H, m), 2.05-2.13 (2H, m), 1.43- 1.74 (2H, m), 1.16 (6H, d, J = 6.8 Hz); HPLC: 100%, MS (ESI): m/z 662.3[M + H]+59pale yellow powder;1H NMR (CD3OD, 400 MHz): δ 8.61 (1H, d, J = 1.6 Hz), 8.33 (1H, d, J = 6.8 Hz), 8.11 (1H, d, J = 6.4 Hz), 7.77-7.85 (2H, m), 7.72-7.76 (1H, m), 7.64-7.70 (1H, m), 7.54-7.63 (2H, m), 7.48 (1H, s), 6.97-7.07 (1H, m), 6.72 (1H, d, J = 15.2 Hz), 4.96-5.03 (2H, m), 4.67-4.74 (1H, m), 4.05-4.09 (2H, m), 3.91-4.00 (4H, m), 3.26-3.32 (4H, m), 2.94-2.99 (7H, m), 1.20 (6H, d, J = 7.2 Hz); LCMS: 100%, MS (ESI): m/z 605.6 [M + H]+60(3R, 4R); off-white powder;1H NMR (400 MHz, CD3OD): δ 8.54 (1H, s), 8.33-8.37 (1H, m), 8.06-8.12 (1H, m), 7.67- 7.78 (2H, m), 7.51-7.64 (3H, m), 7.44-7.50 (2H, m), 6.38- 6.44 (2H, m), 5.80-5.86 (1H, m), 4.94-5.05 (2H, m), 4.52- 4.72 (2H, m), 3.49-3.75 (2H, m), 3.32-3.38 (1H, m), 2.64- 2.95 (3H, m), 1.85-2.01 (1H, m), 1.71-1.79 (1H, m), 1.42- 1.50 (1H, m), 1.09 (6H, d, J = 6.8 Hz); HPLC: 97.5%, MS (ESI): m/z 592.3 [M + H]+61pale yellow powder;1H NMR (400 MHz, CD3OD): δ 8.64 (1H, s), 8.37 (1H, d, J = 6.8 Hz), 8.16 (1H, d, J = 6.8 Hz), 7.76-7.86 (3H, m), 7.65-7.70 (1H, m), 7.54-7.64 (3H, m), 6.98-7.07 (1H, m), 6.72 (1H, d, J = 15.2 Hz), 4.95-5.01 (1H, m), 4.81-4.87 (1H, m), 4.68-4.74 (1H, m), 4.07 (2H, d, J = 7.2 Hz), 3.85-3.99 (2H, m), 3.52-3.64 (3H, m), 3.40 (3H, s), 3.00-3.09 (1H, m), 2.96 (6H, s), 2.91 (1H, s), 1.83-2.05 (3H, m), 1.57-1.69 (2H, m), 1.15-1.20 (6H, m); LCMS: 99.4%, MS (ESI): m/z 634.3 [M + H]+62Racemic mixture; yellow solid;1H -NMR (CD3OD, 400 MHz): δ 8.65-8.69 (1H, m), 8.43-8.47 (1H, m), 8.18-8.24 (1H, m), 7.78-7.88 (3H, m), 7.65-7.72 (2H, m), 7.57-7.61 (2H, m), 6.98-7.06 (1H, m), 6.73 (1H, d, J = 15.2 Hz), 4.99-5.11 (2H, m), 4.70-4.81 (1H, m), 3.84- 4.12 (5H, m), 3.38-3.65 (3H, m), 2.81-3.24 (9H, 1.16-1.22 (6H, m); LCMS: 100%, MS (ESI): 657.2 m/z [M + Na]+63yellow powder;1H NMR (CD3OD, 400 MHz): δ 8.63 (1H, s), 8.39-8.45 (1H, m), 8.11-8.17 (1H, m), 7.50-7.91 (7H, m), 6.96-7.13 (1H, m), 6.72 (1H, d, J = 15.4 Hz), 5.09-5.16 (1H, m), 3.97- 4.16 (3H, m), 3.37-3.62 (3H, m), 3.12-3.27 (1H, m), 2.77- 3.05 (8H, m), 2.24-2.31 (1H, m), 1.68-2.05 (3H, m), 1.12- 1.29 (6H, m); LCMS: 100%, MS (ESI): 619.3 m/z [M + H]+64(3R, 4R); yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.64-8.68 (1H, m), 8.51 (1H, dd, J = 6.4, 3.2 Hz), 8.27-8.31 (1H, m), 7.81-7.92 (3H, m), 7.65-7.72 (2H, m), 7.57-7.62 (2H, m), 5.87 (1H, dd, J = 46.0, 3.6 Hz), 5.46 (1H, dd, J = 14.8, 3.6 Hz), 5.06-5.13 (1H, m), 4.65-4.75 (1H, m), 3.69-3.76 (1H, m), 3.54-3.62 (1H, m), 3.34-3.47 (3H, m), 2.78-3.01 (3H, m), 1.98-2.05 (1H, m), 1.81-1.92 (1H, m), 1.53-1.65 (1H, m), 1.20 (6H, d, J = 6.8 Hz); LCMS: 99.4%, MS (ESI): 610.2 m/z [M + H]+65(3R, 4R); yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.58-8.61 (1H, m), 8.44 (1H, dd, J = 6.4, 2.4 Hz), 8.17-8.22 (1H, m), 7.79-7.91 (3H, m), 7.56-7.72 (4H, m), 6.92 (1H, d, J = 2.0 Hz), 5.05-5.13 (1H, m), 4.68-4.75 (1H, m), 3.69- 3.76 (1H, m), 3.37-3.59 (4H, m), 2.60-3.01 (7H, m), 1.96-2.07 (3H, m), 1.79-1.88 (1H, m), 1.52-1.63 (1H, m), 1.18 (6H, d, J = 6.8 Hz); LCMS: 98.3%, MS (ESI): 632.2 m/z [M + H]+66(3R, 4R); yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.52-8.59 (1H, m), 8.39-8.45 (1H, m), 8.04-8.13 (1H, m), 7.52-7.88 (7H, m), 5.94 (1H, s), 5.69 (1H, s), 5.08-5.17 (1H, m), 4.62-4.73 (1H, m), 3.34-3.76 (5H, m), 2.76-3.05 (3H, m), 1.52-2.10 (6H, m), 1.20 (6H, d, J = 6.8 Hz); LCMS: 98.9%, MS (ESI): 606.2 m/z [M + H]+67(3R, 4R); light yellow powder;1H-NMR (CD3OD, 400 MHz): δ 8.73 (1H, d, J = 8.0 Hz), 8.32 (1H, d, J = 8.8 Hz), 7.74 (1H, s), 7.59-7.69 (3H, m), 7.46- 7.48 (2H, m), 7.35-7.37 (1H, m), 6.49-6.56 (1H, m), 6.32- 6.36 (1H, m), 5.69-5.76 (1H, m), 4.91-4.95 (3H, m), 4.84- 4.86 (1H, m), 3.60-3.66 (1H, m), 3.34-3.39 (1H, m), 3.25- 3.28 (1H, m), 2.84-2.94 (2H, m), 2.69-2.75 (1H, m), 2.57 (3H, s), 1.88-1.94 (1H, m), 1.73-1.77 (1H, m), 1.43-1.47 (1H, m), 1.26 (6H, d, J = 6.8 Hz); LCMS: 100.0%, MS (ESI): 606.3 m/z [(M + H)]+68(3R, 4R); yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.59-8.63 (1H, m), 8.42-8.47 (1H, m), 8.17-8.22 (1H, m), 7.55-7.73 (7H, m), 7.06-7.12 (1H, m), 6.22 (1H, d, J = 15.2 Hz), 5.05-5.12 (1H, m), 4.64- 4.75 (1H, m), 3.69-3.76 (1H, m), 3.30-3.58 (4H, m), 2.89- 3.01 (1H, m), 2.75-2.84 (2H, m), 1.95-2.04 (4H, m), 1.80- 1.91 (1H, m), 1.52-1.63 (1H, m), 1.18 (6H, d,J = 6.8 Hz); LCMS: 100%, MS (ESI): 606.3 m/z [M + H]+69Racemic mixture; yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.54-8.61 (1H, m), 8.42 (1H, d, J = 6.8 Hz), 8.14-8.18 (1H, m), 7.55-7.87 (7H, m), 7.06- 7.14 (1H, m), 6.22 (1H, d, J = 15.2 Hz), 5.03-5.13 (1H, m), 4.67-4.72 (1H, m), 3.81-4.15 (3H, m), 3.34-3.61 (3H, m), 2.79-3.25 (3H, m), 1.97 (3H, d, J = 6.8 Hz), 1.16-1.21 (6H, m); LCMS: 100%, MS (ESI): 592.3 m/z [M + H]+70Yellow powder;1H-NMR (CD3OD, 400 MHz): δ 8.54 (1H, d, J = 7.6 Hz), 8.45 (1H, d, J = 8.8 Hz), 7.65-7.77 (4H, m), 7.45-7.52 (2H, m), 7.35- 7.37 (1H, m), 6.91-6.97 (1H, m), 6.22 (1H, d, J = 14.8 Hz), 4.93-4.94 (2H, m), 3.64-3.65 (1H, m), 3.32-3.36 (2H, m), 3.26-3.28 (2H, m), 2.85-2.92 (2H, m), 2.74-2.77 (1H, m), 2.62 (3H, s), 1.87-1.95 (3H, m), 1.70-1.77 (1H, m), 1.44- 1.52 (1H, m), 1.25 (6H, d, J = 6.8 Hz), 0.85-0.91 (1H, m); LCMS: 98.5%, MS (ESI): 620.2 m/z [(M + H)]+71yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.57 (1H, d, J = 2.0 Hz), 8.38 (1H, d, J = 6.8 Hz), 8.16 (1H, d, J = 6.8 Hz), 7.79-7.86 (2H, m), 7.66-7.74 (2H, m), 7.57- 7.62 (3H, m), 7.07-7.14 (1H, m), 6.25 (1H, dd, J = 15.2, 1.6 Hz), 5.01-5.05 (1H, m), 4.68- 4.72 (1H, m), 4.01-4.05 (4H, m), 3.33-3.36 (4H, m), 3.00- 3.07 (1H, m), 1.98 (3H, dd, J = 6.8, 1.6 Hz), 1.20 (6H, dd, J = 6.8, 1.6 Hz); LCMS: 100.0%, MS (ESI): 562.2 m/z [(M + H)]+72(3R, 4R); yellow powder;1H-NMR (CD3OD, 400 MHz): δ 8.46-8.52 (1H, m), 8.36-8.38 (1H, m), 8.05-8.08 (1H, m), 7.78-7.87 (2H, m), 7.65-7.69 (2H, m), 7.49-7.55 (3H, m), 7.05-7.12 (1H, m), 6.24 (1H, dd, J = 15.2, 1.2 Hz), 4.99-5.03 (1H, m), 4.65-4.72 (1H, m), 3.67-3.73 (1H, m), 3.38-3.48 (4H, m), 2.98-3.05 (1H, m), 2.81-2.91 (2H, m), 2.4-2.09 (1H, m), 1.97 (3H, dd, J = 6.8, 1.2 Hz), 1.81-1.86 (1H, m), 1.62-1.68 (1H, m), 1.17-1.22 (6H, m); LCMS: 99.4%, MS (ESI):605.3 m/z [(M + H)]+73(3R, 4R); off-white powder;1H NMR (CD3OD, 400 MHz): δ 7.85 (1H, s), 7.36-7.45 (3H, m), 7.34 (1H, d, J = 8.0 Hz), 7.19 (1H, d, J = 7.6 Hz), 6.97- 7.05 (1H, m), 6.60-6.74 (1H, m), 6.17 (1H, d, J = 15.2 Hz), 4.93-4.97 (2H, m), 3.55-3.61 (1H, m), 3.45-3.54 (1H, m), 3.35-3.43 (1H, m), 3.23-3.27 (1H, m), 3.03-3.09 (1H, m), 2.94-3.01 (1H, m), 2.57-2.68 (2H, m), 2.54 (3H, s), 1.88 (3H, d, J = 6.4 Hz), 1.64-1.84 (2H, m), 1.34-1.45 (1H, m), 1.30 (6H, d, J = 6.8 Hz); LCMS: 100%, MS (ESI): m/z 608.3 [M + H]+74(3R, 4R); white powder;1H NMR (CD3OD, 400 MHz): δ 7.93 (1H, s), 7.68-7.84 (2H, m), 7.59 (2H, m), 7.36 (1H, d, J = 7.2 Hz), 7.23-7.30 (1H, m), 6.89-7.04 (2H, m), 6.77-6.83 (1H, m), 6.48-6.56 (1H, m), 6.18 (1H, d, J = 14.8 Hz), 4.51-4.69 (2H, m), 3.60-3.69 (2H, m), 3.41-3.52 (1H, m), 3.13-3.19 (1H, m), 2.70-3.02 (4H, m), 1.89-1.99 (4H, m), 1.74-1.82 (1H, m), 1.41-1.57 (1H, m), 1.20-1.34 (6H, m); LCMS: 99.9%, MS (ESI): 594.2 m/z [M + H]+75white powder;1H NMR (400 MHz, CDCl3): δ 8.33-8.57 (3H, m), 7.53-7.65 (4H, m), 7.34- 7.48 (4H, m), 6.95-7.12 (2H, m), 6.29-6.52 (1H, m), 4.69- 4.88 (1H, m), 4.38-4.59 (3H, m), 4.17-4.36 (1H, m), 3.54- 3.73 (1H, m), 3.25-3.40 (2H, m), 2.83-3.05 (3H, m), 2.48 (6H, s), 1.85-2.0 (2H, m), 1.46 (9H, s), 1.24-1.26 (8H, m); LCMS: 100%, MS (ESI): m/z 719.3 [M + H]+76(3R, 4R); Yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.66-8.70 (1H, m), 8.45 (1H, dd, J = 6.8, 2.4 Hz), 8.19-8.24 (1H, m), 7.80-7.88 (3H, m), 7.64-7.71 (2H, m), 7.56-7.61 (2H, m), 7.01-7.08 (1H, m), 6.70 (1H, d, J = 15.2 Hz), 5.02-5.07 (1H, m), 4.69-4.79 (1H, m), 4.02 (2H, d, J = 7.2 Hz), 3.70-3.76 (1H, m), 3.53- 3.60 (3H, m), 3.31-3.46 (3H, m), 2.76-3.07 (5H, m), 1.96- 2.03 (3H, m), 1.78-1.91 (4H, m), 1.51-1.62 (2H, m), 1.19 (6H, d, J = 7.2 Hz); LCMS: 100%, MS (ESI): 689.3 m/z [M + H]+.77Mixture of 2 trans isomer; yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.55- 8.62 (1H, m), 8.41-8.46 (1H, m), 8.15-8.19 (1H, m), 7.56- 7.87 (7H, m), 7.06-7.18 (1H, m), 6.21 (1H, d, J = 16.0 Hz), 5.09-5.21 (1H, m), 4.67-4.82 (2H, m), 4.26-4.35 (1H, m), 3.41-3.62 (3H, m), 3.12-3.21 (2H, m), 2.80-2.86 (1H, m), 2.38-2.59 (1H, m), 1.97 (3H, dd, J = 7.2, 1.6 Hz), 1.18 (6H, d, J = 6.8 Hz). LCMS: 100%, MS (ESI): 592.2 m/z [M + H]+.78(3R, 4R); gray powder;1H NMR (MeOD, 400 MHz): δ ppm 7.85 1H, s), 7.34-7.48 (4H, m), 7.18 (1H, d, J = 7.6 Hz), 7.03-7.10 (1H, m), 6.49 (1H, s), 5.34-5.52 (1H, m), 5.21 (1H, dd, J = 15.2, 3.2 Hz), 4.92-4.95 (2H, m), 3.50- 3.62 (2H, m), 3.37-3.45 (1H, m), 3.20-3.27 (1H, m), 2.93- 3.05 (2H, m), 2.50-2.61 (5H, m), 1.67-1.83 (2 H, m), 1.33- 1.42 (1H, m), 1.30 (6H, d, J = 6.8 Hz); LCMS: 100%, MS (ESI): m/z 612.3 [M + H]+79Yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.56 (1H, d, J = 2.0 Hz), 8.36 (1H, d, J = 6.4 Hz), 8.15 (1H, d, J = 6.8 Hz), 7.76-7.83 (3H, m), 7.61-7.67 (1H, m), 7.53-7.60 (2H, m), 7.44 (1H, s), 5.85 (1H, dd, J = 46.0, 3.6 Hz), 5.44 (1H, dd, J = 15.2, 3.6 Hz), 5.00 (1H, d, J = 15.6 Hz), 4.63 (1H, d, J = 15.6 Hz), 3.89-4.00 (4H, m), 3.21-3.29 (4H, m, overlap with water signal), 2.87-2.95 (1H, m), 1.15-1.21 (6H, m); LCMS: 100%, MS (ESI): 566.2 m/z [M + H]+80Racemic mixture; Yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.62-8.65 (1H, m), 8.44-8.49 (1H, m), 8.20-8.24 (1H, m), 7.79-7.90 (3H, m), 7.65-7.71 (2H, m), 7.54-7.61 (2H, m), 5.85 (1H, dd, J = 46.4, 3.6 Hz), 5.44 (1H, dd, J = 14.8, 3.6 Hz), 5.03-5.13 (1H, m), 4.65-4.76 (1H, m), 3.81-4.12 (3H, m), 3.32-3.61 (3H, m), 2.78-3.21 (4H, m), 1.16-1.21 (6H, m); LCMS: 99.3%, MS (ESI): 596.2 m/z [M + H]+81off-white powder;1H NMR (400 MHz, CD3OD): δ 8.56 (1H, s), 8.44 (1H, d, J = 6.8 Hz), 8.24 (1H, d, J = 6.4 Hz), 7.92-8.00 (1H, m), 7.81-7.91 (2H, m), 7.73 (1H, t, J = 7.2 Hz), 7.62 (1H, d, J = 7.6 Hz), 7.52-7.56 (1H, m), 7.46 (1H, s), 5.85 (1H, dd, J = 46.4, 4.0 Hz), 5.48 (1H, dd, J = 15.2, 4.0 Hz), 4.90-5.02 (2H, m), 4.73-4.83 (1H, m), 3.41 (3H, s), 3.23-3.32 (2H, m), 2.85-2.94 (1H, m), 1.99-2.24 (3H, m), 1.65-1.76 (1H, m), 1.30-1.55 (4H, m), 1.15-1.24 (6H, m); HPLC: 98.5% MS (ESI): m/z 608.2[M + H]+82(3R, 4R); light yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.52-8.60 (1H, m), 8.44 (1H, J = 6.8 Hz), 8.22-8.26 (1H, m), 7.81-7.92 (3H, m), 7.67-7.72 (1H, m), 7.56-7.63 (2H, m), 7.48 (1H, s), 5.87 (1H, dd, J = 46.0, 2.8 Hz), 5.46 (1H, dd, J = 15.2, 4.0 Hz), 5.07- 5.12 (1H, m), 4.73-4.79 (1H, m), 3.69-3.75 (1H, m), 3.37- 3.56 (4H, m), 2.99-3.12 (1H, m), 2.81-2.97 (1H, m), 2.06- 2.12 (1H, m), 1.82-1.89 (1H, m), 1.64-1.73 (1H, m), 1.17- 1.24 (6H, m); LCMS: 99.7%; MS (ESI): 609.2 m/z [(M + H)]+83Mixture of 2 trans isomer; Yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.60-8.67 (1H, m), 8.48-8.52 (1H, m), 8.24-8.28 (1H, m), 7.81-7.93 (3H, m), 7.56-7.73 (4H, m), 5.85 (1H, dd, J = 46.0, 3.6 Hz), 5.44 (1H, dd, J = 14.8, 3.6 Hz), 5.09-5.22 (1H, m), 4.67-4.77 (1H, m), 4.36-4.35 (1H, m), 3.46-3.62 (3H, m), 3.10-3.21 (2H, m), 2.81-2.88 (1H, m), 2.41-2.62 (1H, m), 1.16-1.22 (6H, m); LCMS: 100%, MS (ESI): 596.2 m/z [M + H]+84Yellow solid;1H-NMR (CD3OD, 400 MHz): δ 8.63 (1H, s), 8.43 (1H, d, J = 6.8 Hz), 8.29 (1H, d, J = 6.8 Hz), 7.82-7.92 (3H, m), 7.65-7.71 (1H, m), 7.57- 7.62 (2H, m), 7.54 (1H, s), 5.85 (1H, dd, J = 46.4, 3.6 Hz), 5.45 (1H, dd, J = 14.8, 3.6 Hz), 4.92-4.99 (1H, m), 4.72-4.80 (1H, m), 3.51-3.58 (1H, m), 3.39 (3H, s), 3.22- 3.31 (1H, m, overlap with CD3OD signal), 2.76-2.83 (1H, m), 2.01-2.18 (3H, m), 1.69- 1.73 (1H, m), 1.27-1.46 (4H, m), 1.15-1.21 (6H, m); LCMS: 100%, MS (ESI): 609.2 m/z [M + H]+85White powder; H NMR (CDCl3, 400 MHz): δ 8.54 (1H, d, J = 5.6 Hz), 8.30 (1H, s), 8.04 (1H, brs), 7.59 (1H, d, J = 8.8 Hz), 7.55 (2H, d, J = 6.0 Hz), 7.44 (1H, s), 7.30-7.42 (3H, m), 7.25 (1H, d, J = 8.8 Hz), 6.92-6.96 (1H, m), 5.85 (1H, dd, J = 47.6, 3.6 Hz), 5.26 (1H, dd, J = 14.8, 3.6 Hz), 4.76-4.85 (1H, m), 4.12-4.31 (3H, m), 3.42-3.53 (3H, m), 3.09-3.18 (2H, m), 2.83-2.96 (1H, m), 1.80-1.87 (2H, m), 1.43-1.51 (2H, m, overlap with water signal), 1.15-1.21 (9H, m); LCMS: 98.8%, MS (ESI): m/z 609.3 [M + H]+86Off-white powder;1H NMR (CDCl3, 400 MHz): δ 8.62 (1H, d, J = 6.0 Hz), 8.41 (1H, s), 8.06-8.15 (1H, m), 7.61-7.75 (3H, m), 7.31-7.57 (5H, m), 7.02-7.23 (1H, m), 5.95 (1H, dd, J = 47.6, 3.2 Hz), 5.37 (1H, dd, J = 14.8, 3.2 Hz), 4.85-4.96 (1H, m), 4.46-4.54 (1H, m), 4.07-4.36 (2H, m), 3.47-3.60 (2H, m), 29.5-3.04 (1H, m), 1.92-2.05 (2H, m), 1.68-1.80 (2H, m), 1.27 (6H, d, J = 6.8 Hz); LCMS: 97.3%, MS (ESI): 649.1 m/z [M + H]+87Racemic mixture; off-white powder;1H NMR (400 MHz, CD3OD) δ 8.39 (1H, s), 7.97 (1H, d, J = 6.4 Hz), 7.83-7.92 (2H, m), 7.73-7.80 (4H, m), 7.61-7.72 (3H, m), 7.56 (1H, d, J = 7.6 Hz), 7.44 (1H, t, J = 8.0 Hz), 7.12-7.16 (1H, m), 5.85 (1H, dd, J = 40.0, 4.0 Hz), 5.79 (1H, dd, J = 14.8, 3.6 Hz), 5.35-5.38 (1H, m), 5.03-5.07 (1H, m), 3.64- 3.69 (1H, m), 3.18-3.27 (2H, m), 3.11-3 .17 (1H, m), 2.41- 2.48 (2H, m), 2.02-2.08 (1H, m), 1.58-1.64 (1H, m), 1.28- 1.34 (6H, m); HPLC: 96.9% MS (ESI): m/z 643.2[M + H]+88Yellow powder;1H NMR (CDCl3, 400 MHz): δ 8.47 (1H, d, J = 6.0 Hz), 8.28 (1H, d, J = 1.2 Hz), 8.04- 8.08 (1H, m), 7.59 (1H, d, J = 9.2 Hz), 7.52-7.57 (2H, m), 7.35-7.46 (3H, m), 7.26- 7.34 (2H, m), 5.85 (1H, dd, J = 47.6, 3.6 Hz), 5.27 (1H, dd, J = 15.2, 3.6 Hz), 4.78- 4.85 (1H, m), 4.08-4.24 (3H, m), 3.32-3.39 (4H, m), 3.19- 3.30 (2H, m), 2.85-2.96 (1H, m), 1.79-1.84 (2H, m), 1.45- 1.58 (2H, m, overlap with water signal), 1.17 (6H, d, J = 6.8 Hz); LCMS: 98%, MS (ESI): m/z 595.2 [M + H]+
173,816
11858938
DETAILED DESCRIPTION Compounds and methods in the fields of chemistry and medicine are disclosed. Some of the disclosed embodiments include compounds, compositions and methods of using heterocycle amines. Some of the disclosed embodiments include heterocycle amines useful to treat hematopoietic disorders. Definitions Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. All patents, applications, published applications and other publications referenced herein are incorporated by reference in their entirety unless stated otherwise. In the event that there are a plurality of definitions for a term herein, those in this section prevail unless stated otherwise. As used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are employed. The use of “or” or “and” means “and/or” unless stated otherwise. Furthermore, use of the term “including” as well as other forms, such as “include”, “includes,” and “included,” is not limiting. As used in this specification, whether in a transitional phrase or in the body of the claim, the terms “comprise(s)” and “comprising” are to be interpreted as having an open-ended meaning. That is, the terms are to be interpreted synonymously with the phrases “having at least” or “including at least.” When used in the context of a process, the term “comprising” means that the process includes at least the recited steps, but may include additional steps. When used in the context of a compound, composition, or device, the term “comprising” means that the compound, composition, or device includes at least the recited features or components, but may also include additional features or components. Unless specific definitions are provided, the nomenclatures utilized in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those known in the art. Standard chemical symbols are used interchangeably with the full names represented by such symbols. Thus, for example, the terms “hydrogen” and “H” are understood to have identical meaning. Standard techniques may be used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Reactions and purification techniques may be performed e.g., using kits according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference in its entirety for any purpose. The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. “Solvate” refers to the compound formed by the interaction of a solvent and a compound described herein or salt thereof. Suitable solvates are pharmaceutically acceptable solvates including hydrates. The term “pharmaceutically acceptable salt” refers to salts that retain the biological effectiveness and properties of a compound and, which are not biologically or otherwise undesirable for use in a pharmaceutical. In many cases, the compounds disclosed herein are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like; particularly preferred are the ammonium, potassium, sodium, calcium and magnesium salts. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. Many such salts are known in the art, as described in WO 87/05297, Johnston et al., published Sep. 11, 1987 (incorporated by reference herein in its entirety). As used herein, “Cato Cb” or “Ca-b” in which “a” and “b” are integers refer to the number of carbon atoms in the specified group. That is, the group can contain from “a” to “b”, inclusive, carbon atoms. Thus, for example, a “C1to C4alkyl” or “C1-4alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH3—, CH3CH2—, CH3CH2CH2—, (CH3)2CH—, CH3CH2CH2CH2—, CH3CH2CH(CH3)— and (CH3)3C—. The term “halogen” or “halo,” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, e.g., fluorine, chlorine, bromine, or iodine, with fluorine and chlorine being preferred. As used herein, “alkyl” refers to a straight or branched hydrocarbon chain that is fully saturated (i.e., contains no double or triple bonds). The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 9 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 4 carbon atoms. The alkyl group may be designated as “C1-4alkyl” or similar designations. By way of example only, “C1-4alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, and the like. As used herein, “alkoxy” refers to the formula —ORawherein Rais an alkyl as is defined above, such as “C1-9alkoxy”, including but not limited to methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, and tert-butoxy, and the like. The term “straight-chain alkoxy” refers to a group comprising the formula: —(CH2)pO— wherein p is any integer. Straight-chain alkoxy does not include substituted or branched alkoxy groups. As used herein, “alkenyl” refers to a straight or branched hydrocarbon chain containing one or more double bonds. The alkenyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term “alkenyl” where no numerical range is designated. The alkenyl group may also be a medium size alkenyl having 2 to 9 carbon atoms. The alkenyl group could also be a lower alkenyl having 2 to 4 carbon atoms. The alkenyl group may be designated as “C2-4alkenyl” or similar designations. By way of example only, “C2-4alkenyl” indicates that there are two to four carbon atoms in the alkenyl chain, i.e., the alkenyl chain is selected from the group consisting of ethenyl, propen-1-yl, propen-2-yl, propen-3-yl, buten-1-yl, buten-2-yl, buten-3-yl, buten-4-yl, 1-methyl-propen-1-yl, 2-methyl-propen-1-yl, 1-ethyl-ethen-1-yl, 2-methyl-propen-3-yl, buta-1,3-dienyl, buta-1,2,-dienyl, and buta-1,2-dien-4-yl. Typical alkenyl groups include, but are in no way limited to, ethenyl, propenyl, butenyl, pentenyl, and hexenyl, and the like. As used herein, “alkynyl” refers to a straight or branched hydrocarbon chain containing one or more triple bonds. The alkynyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term “alkynyl” where no numerical range is designated. The alkynyl group may also be a medium size alkynyl having 2 to 9 carbon atoms. The alkynyl group could also be a lower alkynyl having 2 to 4 carbon atoms. The alkynyl group may be designated as “C2-4alkynyl” or similar designations. By way of example only, “C2-4alkynyl” indicates that there are two to four carbon atoms in the alkynyl chain, i.e., the alkynyl chain is selected from the group consisting of ethynyl, propyn-1-yl, propyn-2-yl, butyn-1-yl, butyn-3-yl, butyn-4-yl, and 2-butynyl. Typical alkynyl groups include, but are in no way limited to, ethynyl, propynyl, butynyl, pentynyl, and hexynyl, and the like. The term “haloalkyl” refers to an alkyl in which at least one hydrogen atom is replaced with a halogen atom. In certain of the embodiments in which two or more hydrogen atoms are replaced with halogen atoms, the halogen atoms are all the same as one another. In certain of such embodiments, the halogen atoms are not all the same as one another. As used herein, “heteroalkyl” refers to a straight or branched hydrocarbon chain containing one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur. The heteroalkyl group may have 1 to 20 carbon atom, although the present definition also covers the occurrence of the term “heteroalkyl” where no numerical range is designated. The heteroalkyl group may be designated as “C1-4heteroalkyl” or similar designations. By way of example only, “C1-4heteroalkyl” indicates that there are one to four carbon atoms in the heteroalkyl chain and additionally one or more heteroatoms. In some embodiments, the term “heteroalkyl” refers to an alkyl group comprising two or more carbon atoms in which at least one —CH2— unit of the alkyl group is replaced with a substituent selected from —C═O, —NH—, —S— or —O—; or at least one unit is replaced with Examples of heteroalkyls include, but are not limited to, CH3C(═O)CH2—, CH3C(═O)CH2CH2—, CH3CH2C(═O)CH2CH2—, CH3C(═O)CH2CH2CH2—, CH3NHC(═O)CH2—, CH3C(═O)NHCH2—, CH3OCH2CH2—, CH3NHCH2—, and the like. The term “aromatic” refers to a ring or ring system having a conjugated pi electron system and includes both carbocyclic aromatic (e.g., phenyl) and heterocyclic aromatic groups (e.g., pyridine). The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of atoms) groups provided that the entire ring system is aromatic. As used herein, the term “carbocycle” or “carbocyclyl” refers to non-aromatic cyclic ring or ring system containing only carbon atoms in the ring system backbone. Carbocylic rings may be formed by three, four, five, six, seven, eight, nine, or more than nine carbon atoms. The carbocyclyl group may have 3 to 20 carbon atoms, although the present definition also covers the occurrence of the term “carbocyclyl” where no numerical range is designated. The carbocyclyl group may also be a medium size carbocyclyl having 3 to 10 carbon atoms. The carbocyclyl group could also be a carbocyclyl having 3 to 6 carbon atoms. The carbocyclyl group may be designated as “C3-6carbocyclyl” or similar designations. Examples of carbocyclyl rings include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,3-dihydro-indene, bicycle[2.2.2]octanyl, adamantyl, and spiro[4.4]nonanyl. As used herein, “cycloalkyl” means a fully saturated carbocyclyl ring or ring system. Examples include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. A “(carbocyclyl)alkyl” is a carbocyclyl group connected, as a substituent, via an alkylene group, such as “C4-10(carbocyclyl)alkyl” and the like, including but not limited to, cyclopropylmethyl, cyclobutylmethyl, cyclopropylethyl, cyclopropylbutyl, cyclobutylethyl, cyclopropylisopropyl, cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl, cyclohexylethyl, cycloheptylmethyl, and the like. In some cases, the alkylene group is a lower alkylene group. As used herein, “aryl” refers to an aromatic ring or ring system (i.e., two or more fused rings that share two adjacent carbon atoms) containing only carbon in the ring backbone. When the aryl is a ring system, every ring in the system is aromatic. The aryl group may have 6 to 18 carbon atoms, although the present definition also covers the occurrence of the term “aryl” where no numerical range is designated. In some embodiments, the aryl group has 6 to 10 carbon atoms. The aryl group may be designated as “C6-10aryl,” “C6or C10aryl,” or similar designations. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, azulenyl, and anthracenyl. An “aralkyl” or “arylalkyl” is an aryl group connected, as a substituent, via an alkylene group, such as “C7-14aralkyl” and the like, including but not limited to benzyl, 2-phenylethyl, 3-phenylpropyl, and naphthylalkyl. In some cases, the alkylene group is a lower alkylene group (i.e., a C1-4alkylene group). As used herein, “heteroaryl” refers to an aromatic ring or ring system (i.e., two or more fused rings that share two adjacent atoms) that contain(s) one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur, in the ring backbone. When the heteroaryl is a ring system, every ring in the system is aromatic. The heteroaryl group may have 5-18 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term “heteroaryl” where no numerical range is designated. In some embodiments, the heteroaryl group has 5 to 10 ring members or 5 to 7 ring members. The heteroaryl group may be designated as “5-7 membered heteroaryl,” “5-10 membered heteroaryl,” or similar designations. Examples of heteroaryl rings include, but are not limited to, furyl, thienyl, phthalazinyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, quinolinyl, isoquinlinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, indolyl, isoindolyl, and benzothienyl. A “heteroaralkyl” or “heteroarylalkyl” is heteroaryl group connected, as a substituent, via an alkylene group. Examples include but are not limited to 2-thienylmethyl, 3-thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazollylalkyl, and imidazolylalkyl. In some cases, the alkylene group is a lower alkylene group (i.e., a C1-4alkylene group). The term “heterocycle” or “heterocyclyl” means a non-aromatic cyclic ring or ring system containing at least one heteroatom in the ring backbone. Heterocyclyls may be joined together in a fused, bridged or spiro-connected fashion. Heterocyclyls may have any degree of saturation provided that at least one ring in the ring system is not aromatic. The heteroatom(s) may be present in either a non-aromatic or aromatic ring in the ring system. The heterocyclyl group may have 3 to 20 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term “heterocyclyl” where no numerical range is designated. The heterocyclyl group may also be a medium size heterocyclyl having 3 to 10 ring members. The heterocyclyl group could also be a heterocyclyl having 3 to 6 ring members. The heterocyclyl group may be designated as “3-6 membered heterocyclyl” or similar designations. In preferred six membered monocyclic heterocyclyls, the heteroatom(s) are selected from one up to three of O, N or S, and in preferred five membered monocyclic heterocyclyls, the heteroatom(s) are selected from one or two heteroatoms selected from O, N, or S. Examples of heterocyclyl rings include, but are not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, imidazolidinyl, morpholinyl, oxiranyl, oxepanyl, thiepanyl, piperidinyl, piperazinyl, dioxopiperazinyl, pyrrolidinyl, pyrrolidonyl, pyrrolidionyl, 4-piperidonyl, pyrazolinyl, pyrazolidinyl, 1,3-dioxinyl, 1,3-dioxanyl, 1,4-dioxinyl, 1,4-dioxanyl, 1,3-oxathianyl, 1,4-oxathiinyl, 1,4-oxathianyl, 2H-1,2-oxazinyl, trioxanyl, hexahydro-1,3,5-triazinyl, 1,3-dioxolyl, 1,3-dioxolanyl, 1,3-dithiolyl, 1,3-dithiolanyl, isoxazolinyl, isoxazolidinyl, oxazolinyl, oxazolidinyl, oxazolidinonyl, thiazolinyl, thiazolidinyl, 1,3-oxathiolanyl, indolinyl, isoindolinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydro-1,4-thiazinyl, thiamorpholinyl, dihydrobenzofuranyl, benzimidazolidinyl, and tetrahydroquinoline. Examples of heterocycles include, but are not limited to the following: wherein D, E, F, and G independently represent a heteroatom. Each of D, E, F, and G may be the same or different from one another. A “(heterocyclyl)alkyl” is a heterocyclyl group connected, as a substituent, via an alkylene group. Examples include, but are not limited to, imidazolinylmethyl and indolinylethyl. The term “heteroatom” refers to an atom other than carbon or hydrogen. Heteroatoms, typically, are independently selected from oxygen, sulfur, nitrogen, and phosphorus, but heteroatoms are not limited to those atoms. In embodiments in which two or more heteroatoms are present, the two or more heteroatoms may all be the same as one another, or some or all of the two or more heteroatoms may each be different from the others. The substituent “R” appearing by itself and without a number designation refers to a substituent selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein. Each of the substituent “RA” and “RB” appearing by itself and without a number designation refers to a substituent independently selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein. The term “O-carboxy” refers to the group consisting of formula RC(═O)O—. The term “C-carboxy” refers to the group consisting of formula —C(═O)OR. As used herein, “acyl” refers to —C(═O)R, wherein R is hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein. Non-limiting examples include formyl, acetyl, propanoyl, benzoyl, and acryl. The term “acetyl” refers to the group consisting of formula —C(═O)CH3. The term “trihalomethanesulfonyl” refers to the group consisting of formula X3CS(═O)2— where X is a halogen. The term “cyano” refers to the group consisting of formula —CN. The term “cyanato” refers to the group consisting of formula —OCN. The term “isocyanato” refers to the group consisting of formula —NCO. The term “thiocyanato” refers to the group consisting of formula —CNS. The term “isothiocyanato” refers to the group consisting of formula —NCS. The term “sulfinyl” refers to the group consisting of formula —S(═O)—R. The term “sulfonyl” refers to the group consisting of formula —S(O)2R. The term “S-sulfonamido” refers to the group consisting of formula —S(═O)2NRARB. The term “N-sulfonamido” refers to the group consisting of formula RAS(═O)2NRB—. The term “O-carbamyl” refers to the group consisting of formula —OC(═O)—NRARB. The term “N-carbamyl” refers to the group consisting of formula RBOC(═O)N(RA)—. The term “O-thiocarbamyl” refers to the group consisting of formula —OC(═S)—NRARB. The term “N-thiocarbamyl” refers to the group consisting of formula RBOC(═S)N(RA)—. The term “C-amido” refers to the group consisting of formula —C(═O)—NRARB. The term “N-amido” refers to the group consisting of formula RBC(═O)N(RA)—. The term “oxo” refers to the group consisting of formula ═O. The term “carbonyl” refers to the group consisting of formula —C(═O)—. The term “thiocarbonyl” refers to the group consisting of formula —C(═S)—. The term “ester” refers to a chemical moiety with formula —COOR, where R is selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein. An “amino” group refers to a “—NRARB” group in which RAand RBare each independently selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein. A non-limiting example includes free amino (i.e., —NH2). An “aminoalkyl” group refers to an amino group connected via an alkylene group. An “alkoxyalkyl” group refers to an alkoxy group connected via an alkylene group, such as a “C2-8alkoxyalkyl” and the like. An “O-carboxy” group refers to a “—OC(═O)R” group in which R is selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein. A “C-carboxy” group refers to a “—C(═O)OR” group in which R is selected from hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein. A non-limiting example includes carboxyl (i.e., —C(═O)OH). As used herein, a substituted group is derived from the unsubstituted parent group in which there has been an exchange of one or more hydrogen atoms for another atom or group. Unless otherwise indicated, when a group is deemed to be “substituted,” it is meant that the group is substituted with one or more substituents independently selected from C1-C6alkyl, C1-C6alkenyl, C1-C6alkynyl, C1-C6heteroalkyl, C3-C7carbocyclyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), C3-C7-carbocyclyl-C1-C6-alkyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), 5-10 membered heterocyclyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), 5-10 membered heterocyclyl-C1-C6-alkyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), aryl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), aryl(C1-C6)alkyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), 5-10 membered heteroaryl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), 5-10 membered heteroaryl(C1-C6)alkyl (optionally substituted with halo, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, and C1-C6haloalkoxy), halo, cyano, hydroxy, C1-C6alkoxy, C1-C6alkoxy(C1-C6)alkyl (i.e., ether), aryloxy, sulfhydryl (mercapto), halo(C1-C6)alkyl (e.g., —CF3), halo(C1-C6)alkoxy (e.g., —OCF3), C1-C6alkylthio, arylthio, amino, amino(C1-C6)alkyl, nitro, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, acyl, cyanato, isocyanato, thiocyanato, isothiocyanato, sulfinyl, sulfonyl, and oxo (═O). Wherever a group is described as “optionally substituted” that group can be substituted with the above substituents. It is to be understood that certain radical naming conventions can include either a mono-radical or a di-radical, depending on the context. For example, where a substituent requires two points of attachment to the rest of the molecule, it is understood that the substituent is a di-radical. For example, a substituent identified as alkyl that requires two points of attachment includes di-radicals such as —CH2—, —CH2CH2—, —CH2CH(CH3)CH2—, and the like. Other radical naming conventions clearly indicate that the radical is a di-radical such as “alkylene” or “alkenylene.” The term “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” as used herein, means one or more compatible solid or liquid filler diluents or encapsulating substances, which are suitable for administration to a mammal. The term “compatible”, as used herein, means that the components of the composition are capable of being commingled with the subject compound, and with each other, in a manner such that there is no interaction, which would substantially reduce the pharmaceutical efficacy of the composition under ordinary use situations. Pharmaceutically-acceptable carriers must, of course, be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration preferably to an animal, preferably mammal being treated. The term “pharmaceutical agent” refers to a chemical compound or composition capable of inducing a desired therapeutic effect in a patient. In certain embodiments, a pharmaceutical agent comprises an active agent, which is the agent that induces the desired therapeutic effect. In certain embodiments, a pharmaceutical agent comprises a prodrug. In certain embodiments, a pharmaceutical agent comprises inactive ingredients such as carriers, excipients, and the like. The term “therapeutically effective amount” as used herein, refer to an amount of a compound sufficient to cure, ameliorate, slow progression of, prevent, or reduce the likelihood of onset of the identified disease or condition, or to exhibit a detectable therapeutic, prophylactic, or inhibitory effect. The effect can be detected by, for example, the assays disclosed in the following examples. The precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration. Therapeutically and prophylactically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician. The term “prodrug” refers to a pharmaceutical agent that is converted from a less active form into a corresponding more active form in vivo. Isotopes may be present in the compounds described. Each chemical element as represented in a compound structure may include any isotope of said element. For example, in a compound structure a hydrogen atom may be explicitly disclosed or understood to be present in the compound. At any position of the compound that a hydrogen atom may be present, the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen-1 (protium) and hydrogen-2 (deuterium). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise. Where the compounds disclosed herein have at least one chiral center, they may exist as individual enantiomers and diastereomers or as mixtures of such isomers, including racemates. Separation of the individual isomers or selective synthesis of the individual isomers is accomplished by application of various methods which are well known to practitioners in the art. Unless otherwise indicated, all such isomers and mixtures thereof are included in the scope of the compounds disclosed herein. Furthermore, compounds disclosed herein may exist in one or more crystalline or amorphous forms. Unless otherwise indicated, all such forms are included in the scope of the compounds disclosed herein including any polymorphic forms. In addition, some of the compounds disclosed herein may form solvates with water (i.e., hydrates) or common organic solvents. Unless otherwise indicated, such solvates are included in the scope of the compounds disclosed herein. The term “pharmaceutically acceptable” refers to a formulation of a compound that does not significantly abrogate the biological activity, a pharmacological activity and/or other properties of the compound when the formulated compound is administered to a patient. In certain embodiments, a pharmaceutically acceptable formulation does not cause significant irritation to a patient. The term “co-administer” refers to administering more than one pharmaceutical agent to a patient. In certain embodiments, co-administered pharmaceutical agents are administered together in a single dosage unit. In certain embodiments, co-administered pharmaceutical agents are administered separately. In certain embodiments, co-administered pharmaceutical agents are administered at the same time. In certain embodiments, co-administered pharmaceutical agents are administered at different times. The term “patient” includes human and animal subjects. The term “substantially pure” means an object species (e.g., compound) is the predominant species present (i.e., on a molar basis it is more abundant than any other individual species in the composition). In certain embodiments, a substantially purified fraction is a composition wherein the object species comprises at least about 50 percent (on a molar basis) of all species present. In certain embodiments, a substantially pure composition will comprise more than about 80%, 85%, 90%, 95%, or 99% of all species present in the composition. In certain embodiments, the object species is purified to essential homogeneity (contaminant species cannot be detected in the composition by conventional detection methods) wherein the composition consists essentially of a single species. Compounds Some embodiments disclosed herein relate to a compound of formula (I) as described above or a pharmaceutically acceptable salt thereof. In some embodiments, X is N, X′ is CR4b, Y is CR5a, and Y′ is CR5b. In some such embodiments, R4bis hydrogen. In some such embodiments, R5ais hydrogen. In some such embodiments, R5bis hydrogen. In some embodiments, X′ is N, X is CR4a, Y is CR5a, and Y′ is CR5b. In some such embodiments, R4ais hydrogen. In some such embodiments, R5ais hydrogen. In some such embodiments, R5bis hydrogen. In some embodiments, Y is N, X is CR4a, X′ is CR4b, and Y′ is CR5b. In some such embodiments, R4ais hydrogen. In some such embodiments, R4bis hydrogen. In some such embodiments, R5bis hydrogen. In some embodiments, Y is N, X is CR4a, X′ is CR4b, and Y is CR5a. In some such embodiments, R4ais hydrogen. In some such embodiments, R4bis hydrogen. In some such embodiments, R5ais hydrogen. In some embodiments, the compound of Formula (I) is also represented by Formula (II): In some such embodiments, each R4aand R4bis hydrogen. In some embodiments, the compound of Formula (I) is also represented by Formula (III): In some such embodiments, each R4aand R5ais hydrogen. In some embodiments, R1is an optionally substituted (5 to 7 membered heterocyclyl)alkyl. In some such embodiments, R1is an optionally substituted (5 membered heterocyclyl)alkyl. In some such embodiments, R1is pyrrolidyl-CH2—. In one embodiment, R1is 1-pyrrolidinyl-CH2—. In some embodiments, R1is an optionally substituted (6 membered heterocyclyl)alkyl. In some such embodiments, R1is selected from piperidinyl-CH2— or morpholine-CH2—. In one embodiment, R1is 1-piperidinyl-CH2—. In another embodiment, R1is 1-morpholino-CH2—. In some embodiments, R1is an optionally substituted 5 to 7 membered heterocyclyl. In some sucy embodiments, R1is an optionally substituted 6 membered heterocyclyl. In some such embodiments, R1is selected from optionally substituted morpholinyl, optionally substituted piperazinyl, or optionally substituted piperidinyl. In one embodiment, R1is 1-morpholinyl. In another embodiment, R1is 4-substituted-piperazin-1-yl. In yet another embodiment, R1is and 1-substituted-piperidin-4-yl. In some embodiments, R2is hydrogen. In some embodiments, R3is hydrogen. In some further embodiments, both R2and R3are hydrogen. In some embodiments, R5ais hydrogen and R5bis selected from CN or an optionally substituted 5 to 10 membered heteroaryl. In some such embodiments, R5bis an optionally substituted 6 membered heteroaryl. In some such embodiments, R5bis selected from pyridyl, pyrimidyl, pyrazinyl, or pyridazinyl. In some other embodiments, R5bis an optionally substituted 5 membered heteroaryl. In one such embodiment, R5bis pyrazolyl. In some embodiments, the compound of formula (I) is selected from or pharmaceutically acceptable salts, esters, amides, or prodrugs thereof. Exemplary Synthetic Methods In certain embodiments, compounds of the present invention can by synthesized using the following Schemes. Scheme I describes general synthesis of the 2-(2-pyridinylamino)imidazo[1,2-a]pyrazines or 2-(2-pyridinylamino)imidazo[1,2-a]pyridazines of structure 6. An aminopyridazine of structure 1 is treated sequentially with TsCl and 2-bromoacetamide in the presence of bases to afford an intermediate o structure 2. Treatment of the intermediate of structure 2 with trifluoroacetic acid generates a bicyclic compound of structure 3. Palladium catalyzed coupling of structure 3 with a boronic acid affords a compound of structure 4. Deprotection of the trifluoroacetyl group followed by treatment of a 2-chloropyridine compound of structure 5 yield the final product of structure 6. One of skill in the art will recognize that analogous synthetic schemes may be used to synthesize similar compounds. One of skill will recognize that compounds of the present invention may be synthesized using other synthesis schemes. In certain embodiments, the invention provides a salt corresponding to any of the compounds provided herein. Certain Pharmaceutical Compositions Some embodiments include pharmaceutical compositions comprising: (a) a safe and therapeutically effective amount of a compound described herein (including enantiomers, diastereoisomers, tautomers, polymorphs, and solvates thereof), or pharmaceutically acceptable salts thereof; and (b) a pharmaceutically acceptable carrier, diluent, excipient or combination thereof. The compounds useful as described above can be formulated into pharmaceutical compositions for use in treatment of these conditions. Standard pharmaceutical formulation techniques are used, such as those disclosed in Remington's The Science and Practice of Pharmacy, 21st Ed., Lippincott Williams & Wilkins (2005), incorporated by reference in its entirety. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments is prepared using known techniques, including, but not limited to mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tabletting processes. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments is a liquid (e.g., a suspension, elixir and/or solution). In certain of such embodiments, a liquid pharmaceutical composition comprising one or more compounds of the present embodiments is prepared using ingredients known in the art, including, but not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments is a solid (e.g., a powder, tablet, and/or capsule). In certain of such embodiments, a solid pharmaceutical composition comprising one or more compounds of the present embodiments is prepared using ingredients known in the art, including, but not limited to, starches, sugars, diluents, granulating agents, lubricants, binders, and disintegrating agents. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments is formulated as a depot preparation. Certain such depot preparations are typically longer acting than non-depot preparations. In certain embodiments, such preparations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. In certain embodiments, depot preparations are prepared using suitable polymeric or hydrophobic materials (for example an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments comprises a delivery system. Examples of delivery systems include, but are not limited to, liposomes and emulsions. Certain delivery systems are useful for preparing certain pharmaceutical compositions including those comprising hydrophobic compounds. In certain embodiments, certain organic solvents such as dimethylsulfoxide are used. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments comprises a sustained-release system. A non-limiting example of such a sustained-release system is a semi-permeable matrix of solid hydrophobic polymers. In certain embodiments, sustained-release systems may, depending on their chemical nature, release compounds over a period of hours, days, weeks or months. Certain compounds used in the pharmaceutical composition of the present embodiments may be provided as pharmaceutically acceptable salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments comprises an active ingredient in a therapeutically effective amount. In certain embodiments, the therapeutically effective amount is sufficient to prevent, alleviate or ameliorate symptoms of a disease or to prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments is formulated as a prodrug. In certain embodiments, prodrugs are useful because they are easier to administer than the corresponding active form. For example, in certain instances, a prodrug may be more bioavailable (e.g., through oral administration) than is the corresponding active form. In certain instances, a prodrug may have improved solubility compared to the corresponding active form. In certain embodiments, a prodrug is an ester. In certain embodiments, such prodrugs are less water soluble than the corresponding active form. In certain instances, such prodrugs possess superior transmittal across cell membranes, where water solubility is detrimental to mobility. In certain embodiments, the ester in such prodrugs is metabolically hydrolyzed to carboxylic acid. In certain instances the carboxylic acid containing compound is the corresponding active form. In certain embodiments, a prodrug comprises a short peptide (polyaminoacid) bound to an acid group. In certain of such embodiments, the peptide is metabolized to form the corresponding active form. In certain embodiments, a pharmaceutical composition comprising one or more compounds of the present embodiments is useful for treating a condition or disorder in a mammal, such as a human. Suitable administration routes include, but are not limited to, oral, rectal, transmucosal, intestinal, enteral, topical, suppository, through inhalation, intrathecal, intraventricular, intraperitoneal, intranasal, intraocular and parenteral (e.g., intravenous, intramuscular, intramedullary, and subcutaneous). In certain embodiments, pharmaceutical intrathecals are administered to achieve local rather than systemic exposures. For example, pharmaceutical compositions may be injected directly in the area of desired effect (e.g., in the renal or cardiac area). In certain embodiments, a pharmaceutical composition comprising a compound of the present embodiments is prepared for oral administration. In certain of such embodiments, a pharmaceutical composition is formulated by combining one or more compounds of the present embodiments with one or more pharmaceutically acceptable carriers. Certain of such carriers enable compounds of the present embodiments to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient. In certain embodiments, pharmaceutical compositions for oral use are obtained by mixing one or more compounds of the present embodiments and one or more solid excipient. Suitable excipients include, but are not limited to, fillers, such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). In certain embodiments, such a mixture is optionally ground and auxiliaries are optionally added. In certain embodiments, pharmaceutical compositions are formed to obtain tablets or dragee cores. In certain embodiments, disintegrating agents (e.g., cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate) are added. In certain embodiments, dragee cores are provided with coatings. In certain of such embodiments, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to tablets or dragee coatings. In certain embodiments, pharmaceutical compositions for oral administration are push-fit capsules made of gelatin. Certain of such push-fit capsules comprise one or more compounds of the present embodiments in admixture with one or more filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In certain embodiments, pharmaceutical compositions for oral administration are soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. In certain soft capsules, one or more compounds of the present embodiments are be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. In certain embodiments, a pharmaceutical composition is prepared for administration by injection (e.g., intravenous, subcutaneous, intramuscular, etc.). In certain of such embodiments, a pharmaceutical composition comprises a carrier and is formulated in aqueous solution, such as water or physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. In certain embodiments, other ingredients are included (e.g., ingredients that aid in solubility or serve as preservatives). In certain embodiments, injectable suspensions are prepared using appropriate liquid carriers, suspending agents and the like. Certain pharmaceutical compositions for injection are presented in unit dosage form, e.g., in ampoules or in multi-dose containers. Certain pharmaceutical compositions for injection are suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Certain solvents suitable for use in pharmaceutical compositions for injection include, but are not limited to, lipophilic solvents and fatty oils, such as sesame oil, synthetic fatty acid esters, such as ethyl oleate or triglycerides, and liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, such suspensions may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. In certain embodiments, a pharmaceutical composition is prepared for rectal administration, such as a suppositories or retention enema. Certain of such pharmaceutical compositions comprise known ingredients, such as cocoa butter and/or other glycerides. In certain embodiments, a pharmaceutical composition is prepared for topical administration. Certain of such pharmaceutical compositions comprise bland moisturizing bases, such as ointments or creams. Exemplary suitable ointment bases include, but are not limited to, petrolatum, petrolatum plus volatile silicones, lanolin and water in oil emulsions such as Eucerin™, available from Beiersdorf (Cincinnati, Ohio). Exemplary suitable cream bases include, but are not limited to, Nivea™ Cream, available from Beiersdorf (Cincinnati, Ohio), cold cream (USP), Purpose Cream™, available from Johnson & Johnson (New Brunswick, New Jersey), hydrophilic ointment (USP) and Lubriderm™, available from Pfizer (Morris Plains, New Jersey). In certain embodiments, the formulation, route of administration and dosage for a pharmaceutical composition of the present embodiments can be chosen in view of a particular patient's condition. In certain embodiments, a pharmaceutical composition is administered as a single dose. In certain embodiments, a pharmaceutical composition is administered as a series of two or more doses administered over one or more days. In certain embodiments, a pharmaceutical composition of the present embodiments is administered for a period of continuous therapy. For example, a pharmaceutical composition of the present embodiments may be administered over a period of days, weeks, months, or years. In certain embodiments, a pharmaceutical composition may be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. Compositions comprising a compound of the present embodiments formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. In certain embodiments, a pharmaceutical composition is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. The compounds are administered at a therapeutically effective dosage, e.g., a dosage sufficient to provide treatment for the disease states previously described. While human dosage levels have yet to be optimized for the compounds of the preferred embodiments, generally, a daily dose for most of the compounds described herein is from about 0.25 mg/kg to about 120 mg/kg or more of body weight, from about 0.5 mg/kg or less to about 70 mg/kg, from about 1.0 mg/kg to about 50 mg/kg of body weight, or from about 1.5 mg/kg to about 10 mg/kg of body weight. Thus, for administration to a 70 kg person, the dosage range would be from about 17 mg per day to about 8000 mg per day, from about 35 mg per day or less to about 7000 mg per day or more, from about 70 mg per day to about 6000 mg per day, from about 100 mg per day to about 5000 mg per day, or from about 200 mg to about 3000 mg per day. The amount of active compound administered will, of course, be dependent on the subject and disease state being treated, the severity of the affliction, the manner and schedule of administration and the judgment of the prescribing physician. Administration of the compounds disclosed herein or the pharmaceutically acceptable salts thereof can be via any of the accepted modes of administration for agents that serve similar utilities including, but not limited to, orally, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarilly, vaginally, rectally, or intraocularly. Oral and parenteral administrations are customary in treating the indications that are the subject of the preferred embodiments. Certain Therapeutic Methods Some compounds and compositions provided herein, such as compounds and/or compositions comprising Formula I are useful for the treatment of a variety of diseases and disorders malignant or benign hematopoietic disorders. Examples of leukemia include acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL), acute myelogenous leukemia, chronic myelogenous leukemia (CML), chronic idiopathic myelofibrosis, chronic neutrophilic leukemia (CNL), and acute monocytic leukemia. In some embodiments, the compounds and compositions provided herein can be administered in combination with one or more additional anticancer agent(s) or treatment. Examples of other anticancer agents or treatment include HDAC inhibitors, chemotherapy, immunotherapy, VEGFR modulators, bone marrow transplant, and stem cell transplant. EXAMPLES The following examples, including experiments and results achieved, are provided for illustrative purposes only and are not to be construed as limiting the present invention. Where chemical structures depict atoms having an unfilled valency, it is to be understood that the valency is satisfied with one or more hydrogen atoms. Example 1 6-(4-Pyridinyl)-2-(4-(1-pyrrolidinylmethyl)-2-pyridinyl)imidazo[1,2-a]pyrazine (Compound 101) Compound 101 was prepared according to the general procedure described in Scheme I from 5-bromopyrazin-2-amine as follows. Preparation of 2-(5-bromo-2-(tosylimino)pyrazin-1(2H)-yl)acetamide (Compound 2A) A mixture of 5-bromopyrazin-2-amine (2.8 g, 16 mmol) and TsCl (3.7 g, 19 mmol) in pyridine (50 mL) was stirred at 90° C. for 5 h. The residue was concentrated under reduce pressure. The residue was diluted with 100 mL of water and the solid formed was collected, washed with water, and dried under vacuum to give the N-tosyl 5-bromopyrazin-2-amine (4.5 g, 85%). A mixture of the N-tosyl intermediate (4.5 g, 14 mmol), bromoacetamide (2.1 g, 15 mmol), and DIEA (1.9 g, 15 mmol) in DMF (60 mL) was stirred at room temperature for 48 h. The residue was poured into 300 mL water and the resulting solid was filtered and dried under vacuum to give Compound 2A (4.7 g, 87% yield). Preparation of 6-(pyridin-4-yl)imidazo[1,2-a]pyrazin-2-amine (Compound 4A) To a suspension of Compound 2A (4.7 g, 12 mmol) in DCM (60 mL) was added TFAA (10 mL) and the resulting mixture was stirred at room temperature for 3 hours. The mixture was concentrated and diluted with 100 mL of aqueous NaHCO3. The crude mixture was purified by column chromatography (PE/EA=5/1) to give Compound 3A as white solid (1.3 g, 35% yield). A mixture of Compound 3A (463 mg, 1.5 mmol), boronic acid (246 mg, 2 mmol), PdCl2(PPh3)2(210 mg, 0.3 mmol), and K2CO3(1.2 g, 4.5 mmol) in dioxane/water (5/1, 12 mL) was stirred at 100° C. in a microwave reactor for 2 hours. The mixture was concentrated and treated with K2CO3(1.4 g, 10 mmol) in dioxane/water (1/1, 20 mL) at 100° C. for 2 hours. After concentrated, the residue was stirred with 20 mL MeOH to form a crude solid that was purified by prep-HPLC (basic condition) to give Compound 4A (260 mg, 82%). Preparation of 6-(4-pyridinyl)-2-(4-(1-pyrrolidinylmethyl)-2-pyridinyl)imidazo[1,2-a]pyrazine (Compound 101) A mixture of Compound 4A (100 mg, 0.47 mmol) and Compound 5A (100 mg, 0.51 mmol), BINAP (20 mg, 0.032 mmol), Pd2(dba)3(20 mg, 0.022 mmol), Cs2CO3(307 mg, 0.95 mmol) in dioxane (10 mL) was stirred at 100° C. for 12 hours. The reaction mixture was concentrated and purified by prep-HPLC to give the desired compound as yellow solid (35 mg, 18% yield).1H-NMR (400 MHz, MeOD) 9.66 (d, J=1.2, 1H), 9.26 (s, 1H), 8.93 (d, J=6.4, 1H), 8.78 (d, J=6.8, 1H), 8.44 (d, J=6.4, 1H), 7.67 (s, 1H), 7.44 (dd, J=6.4 and 1.2, 1H), 4.62 (s, 2H), 3.80-3.55 (m, 4H), and 2.30-2.10 (m, 4H). Example 2 6-(4-Pyridinyl)-2-(4-(1-pyrrolidinylmethyl)-2-pyridinyl)imidazo[1,2-a]pyridazine (Compound 102) Preparation of 2-(5-bromo-2-(tosylimino)pyrazin-1(2H)-yl)acetamide (Compound 2B) A mixture of Compound 1B (6.8 g, 52 mmol) and TsCl (11 g, 57 mmol) in pyridine (60 mL) was stirred at 90° C. for 4 h. The mixture was concentrated under reduce pressure and diluted with 50 mL water to form the crude solid. The solid was washed with water and dried under vacuum to give the N-tosyl intermediate (10 g, 67%). A mixture of the N-tosyl intermediate (4.0 g, 14 mmol), bromoacetamide (2.2 g, 16 mmol), and DIEA (2.0 g, 16 mmol) in DMF (30 mL) was stirred at room temperature for 8 hours. The mixture was poured into 200 mL of ice water and stirred for 0.5 hour. The resulting solid was filtered and dried under vacuum to give Compound 2B (4.3 g, 89% yield). Preparation of 6-(pyridin-4-yl)imidazo[1,2-a]pyrazin-2-amine (Compound 4B) To a suspension of Compound 2B (2.0 g, 5.9 mmol) in DCM (30 mL) was added TFAA (10 mL). The mixture was stirred at room temperature for 3 hours. The reaction mixture was concentrated and diluted with 50 mL of aqueous NaHCO3to gave the crude solid, and further purification by column chromatography (PE/EA=5/1) afforded Compound 3B as white solid (1.3 g, 85% yield). A mixture of Compound 3B (529 mg, 2.0 mmol), 4-pyridineboronic acid (369 mg, 3.0 mmol), PdCl2(PPh3)2(280 mg, 0.4 mmol), and K2CO3(552 mg, 4.0 mmol) in dioxane/water (4/1, 15 mL) was stirred at 100° C. in a microwave reactor for 1.5 hours. The reaction mixture was concentrated and purified by column (PE/EA=1/1 then DCM/MeOH=20/1) to give N-trifluoroacetyl protected Compound 4B as a yellow solid (250 mg, 40% yield). Deprotection reaction with K2CO3(817 mg, 5.9 mmol) in dioxane/MeOH/H2O (1/1/1, 30 mL) at 100° C. for 1 hour to give Compound 4B as a solid (80 mg, 46% yield). Preparation of 6-(4-Pyridinyl)-2-(4-(1-pyrrolidinylmethyl)-2-pyridinyl)imidazo[1,2-a]pyridazine (Compound 102) A mixture of Compound 4B (80 mg, 0.38 mmol), Compound 5A (80 mg, 0.41 mmol), BINAP (15 mg, 0.024 mmol), Pd2(dba)3(15 mg, 0.016 mmol), Cs2CO3(246 mg, 0.76 mmol) in dioxane (5 mL) was stirred at 100° C. for 12 h. The reaction mixture was concentrated and purified by prep-HPLC to give Compound 102 as yellow solid (30 mg, 21% yield).1H-NMR (400 MHz, DMSO-d6) 11.85 (bs, 1H), 11.11 (s, 1H), 9.26 (s, 1H), 9.04 (d, J=4.4, 2H), 8.63 (t, J=6.4, 2H), 8.38 (d, J=5.6, 1H), 8.21 (d, J=9.6, 1H), 8.12 (d, J=9.2, 1H), 7.41 (d, J=5.6, 1H), 7.33 (s, 1H), 4.40 (d, J=5.6, 2H), 3.40 (bs, 2H), 3.06 (bs, 2H), and 2.02-1.91 (m, 4H). Example 3 The following inhibitory assay is useful for evaluating test compounds for inhibition of Flt3 kinase activity. The assay is performed using traditional radioisotope filtration binding. A base reaction buffer is prepared consisting of 20 mM Hepes (pH 7.5), 10 mM MgCl2, 1 mM EGTA, 0.02% Brij35, 0.02 mg/ml BSA, 0.1 mM Na3PO4, 2 mM DTT, 1% DMSO. The peptide substrate abltide [EAIYAAPFAKKK] is added to the base reaction buffer to yield a final concentration of 5 μM. Additional cofactors (1.5 mM CaCl2, 16 ug/ml Calmodulin, and 2 mM MnCl2) are added to the buffer/substrate solution. Recombinant human FLT3 (baculovirus expression system using a C-terminal His6-tag) is added to the solution and gently mixed. Test compounds are dissolved in DMSO and added to the buffer/kinase solution to yield a final concentration of 5 μM. Compounds were tested in a 10-dose IC50 in duplicate with 3-fold serial dilution starting at 1 μM to yield a full dose response curve. Staurosporine was used as positive control with a 10-dose IC50 with 3-fold serial dilution starting at 20 μM to yield a dose response curve. The reaction is initiated by the addition of33P-ATP (specific activity 500 Ci/μl) into the reaction mixture to a final concentration of 10 μM. The kinase reaction was incubated for 120 minutes at room temperature. The reactions are spotted onto P81 ion exchange paper (Whatman #3698-915) and the filter papers are extensively washed in 0.75% Phosphoric acid and beta emissions are measured. Flt3 inhibition IC50 values of the compounds are in the table below. NumberFlt3 inhibition IC50(nM)Compound 1010.11Compound 1020.25Staurosporine<1.0 All references cited herein, including but not limited to published and unpublished applications, patents, and literature references, are incorporated herein by reference in their entirety and are hereby made a part of this specification. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material. All numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth herein are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of any claims in any application claiming priority to the present application, each numerical parameter should be construed in light of the number of significant digits and ordinary rounding approaches. The above description discloses several methods and materials of the present invention. This invention is susceptible to modifications in the methods and materials, as well as alterations in the fabrication methods and equipment. Such modifications will become apparent to those skilled in the art from a consideration of this disclosure or practice of the invention disclosed herein. Consequently, it is not intended that this invention be limited to the specific embodiments disclosed herein, but that it cover all modifications and alternatives coming within the true scope and spirit of the invention.
59,612
11858939
DETAILED DESCRIPTION OF THE INVENTION 1. General Description of Certain Embodiments of the Invention This invention provides compounds that are inhibitors of HDAC2. The compounds accordingly are useful for treating, alleviating, or preventing a condition in a subject such as a neurological disorder, memory or cognitive function disorder or impairment, extinction learning disorder, fungal disease or infection, inflammatory disease, hematological disease, or neoplastic disease, or for improving memory or treating, alleviating, or preventing memory loss or impairment. In some embodiments the present invention provides a compound of formula I: or a pharmaceutically acceptable salt thereof, wherein: ring A is selected from R1is an optionally substituted monocyclic or bicyclic, non-aromatic heterocyclyl;R2is selected from optionally substituted C2-C6alkenyl, optionally substituted heteroaryl, optionally substituted partially unsaturated heterocyclyl, optionally substituted partially unsaturated carbocyclyl, and para-substituted phenyl wherein said phenyl can be optionally further substituted, and when ring A comprises two nitrogen atoms, R2is additionally selected from unsubstituted phenyl,wherein any two substituents on adjacent ring atoms in R2are optionally taken together with the adjacent ring atoms to form a ring that is an aryl, a carbocyclyl, a heteroaryl, or a heterocyclyl ring;R3, when present, is selected from chloro, fluoro, —CF3and —CHF2;n is 0 or 1;“1” represents a point of attachment between ring A and —NH—C(O)—R1;“2” represents a point of attachment between ring A and R2;“3” represents a point of attachment between ring A and —NH2. In one embodiment, R2in the compounds of formula I is selected from optionally substituted C2-C6alkenyl, optionally substituted heteroaryl, optionally substituted partially unsaturated heterocyclyl, optionally substituted partially unsaturated carbocyclyl, and para-substituted phenyl, and when ring A comprises two nitrogen atoms, R2is additionally selected from unsubstituted phenyl,wherein any two substituents on adjacent ring atoms in R2are optionally taken together with the adjacent ring atoms to form a ring that is an aryl, a carbocyclyl, a heteroaryl, or a heterocyclyl ring; In other embodiments the present invention provides a compound of formula II: or a pharmaceutically acceptable salt thereof, whereinring A′B′ is a fused bicyclic ring system containing at least two nitrogen atoms, wherein ring A′ is a 6-membered heterocyclyl and ring B′ is a 5-membered heteroaryl;X1is carbon or nitrogen;R3′and R4are each independently halo, (C1-C4)alkyl, halo(C1-C4)alkyl, (C1-C4)alkoxy, or halo(C1-C4)alkoxy;R5is halo, halo(C1-C4)alkyl, (C1-C4)alkoxy, halo(C1-C4)alkoxy, monocyclic heterocyclyl, or (C1-C4)alkyl optionally substituted with monocyclic heterocyclyl, wherein each of said heterocyclyl are optionally and independently substituted with 1 to 2 groups selected from halo, (C1-C4)alkyl, and halo(C1-C4)alkyl;n′ is 0 or 1; andp and t are each independently 0, 1, or 2. 2. Compounds and Definitions Compounds of this invention include those described generally for formula I and II, above, and are further illustrated by the classes, subclasses, and species disclosed herein. It will be appreciated that preferred subsets described for each variable herein can be used for any of the structural subsets as well. As used herein, the following definitions shall apply unless otherwise indicated. As described herein, compounds of the invention may be optionally substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted.” In general, the term “substituted”, whether preceded by the term “optionally” or not, means that a hydrogen radical of the designated moiety is replaced with the radical of a specified substituent, provided that the substitution results in a stable or chemically feasible compound. The term “substitutable”, when used in reference to a designated atom, means that attached to the atom is a hydrogen radical, which hydrogen atom can be replaced with the radical of a suitable substituent. Unless otherwise indicated, an “optionally substituted” group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. A stable compound or chemically feasible compound is one in which the chemical structure is not substantially altered when kept at a temperature from about −80° C. to about +40° C., in the absence of moisture or other chemically reactive conditions, for at least a week, or a compound which maintains its integrity long enough to be useful for therapeutic or prophylactic administration to a patient. The phrase “one or more substituents”, as used herein, refers to a number of substituents that equals from one to the maximum number of substituents possible based on the number of available bonding sites, provided that the above conditions of stability and chemical feasibility are met. As used herein, the term “independently selected” means that the same or different values may be selected for multiple instances of a given variable in a single compound. As used herein, the term “aromatic” includes aryl and heteroaryl groups as described generally below and herein. The term “aliphatic” or “aliphatic group”, as used herein, means a straight-chain or branched C1-12hydrocarbon which is completely saturated or which contains one or more units of unsaturation. For example, suitable aliphatic groups include linear or branched alkyl, alkenyl, alkynyl, alkylene, alkenylene, and alkynylene groups. Unless otherwise specified, in various embodiments, aliphatic groups have 1-12, 1-10, 1-8, 1-6, 1-4, 1-3, or 1-2 carbon atoms. Aliphatic groups can be unsubstituted or substituted (e.g., having 1, 2, 3, or 4 substituent groups as defined herein). The term “alkenyl”, used alone or as part of a larger moiety, refers to an optionally substituted straight or branched chain hydrocarbon group having at least one double bond and having 2-12, 2-10, 2-8, 2-6, 2-4, or 2-3 carbon atoms. The terms “cycloaliphatic”, “carbocycle”, “carbocyclyl”, “carbocyclo”, or “carbocyclic”, used alone or as part of a larger moiety, refer to an optionally substituted saturated or partially unsaturated cyclic aliphatic ring system having from 3 to about 14 ring carbon atoms, and which is not aromatic. In some embodiments, the cycloaliphatic group is an optionally substituted monocyclic hydrocarbon having 3-8 or 3-6 ring carbon atoms. Cycloaliphatic groups include, without limitation, optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, cyclooctyl, cyclooctenyl, or cyclooctadienyl. The terms “cycloaliphatic”, “carbocycle”, “carbocyclyl”, “carbocyclo”, or “carbocyclic” also include optionally substituted bridged or fused bicyclic rings having 6-12, 6-10, or 6-8 ring carbon atoms, wherein any individual ring in the bicyclic system has 3-8 ring carbon atoms. As used herein, the term “halogen” or “halo” means F, Cl, Br, or I. The terms “aryl” and “ar-”, used alone or as part of a larger moiety, e.g., “aralkyl”, “aralkoxy”, or “aryloxyalkyl”, refer to an optionally substituted C6-14aromatic hydrocarbon moiety comprising one to three aromatic rings. Preferably, the aryl group is a C6-10aryl group. Aryl groups include, without limitation, optionally substituted phenyl, naphthyl, or anthracenyl. The terms “aryl” and “ar-”, as used herein, also include groups in which an aryl ring is fused to one or more cycloaliphatic rings to form an optionally substituted cyclic structure such as a tetrahydronaphthyl, indenyl, or indanyl ring. The term “aryl” may be used interchangeably with the terms “aryl group”, “aryl ring”, and “aromatic ring”. The terms “heteroaryl” and “heteroar-”, used alone or as part of a larger moiety, e.g., “heteroaralkyl”, or “heteroaralkoxy”, refer to groups having 5 to 14 ring atoms, preferably 5, 6, 9, or 10 ring atoms; having 6, 10, or 14 π electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. A heteroaryl group may be mono-, bi-, tri-, or polycyclic, preferably mono-, bi-, or tricyclic, more preferably mono- or bicyclic, as long as each ring is aromatic. The term “heteroatom” refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. For example, a nitrogen atom of a heteroaryl may be a basic nitrogen atom and may also be optionally oxidized to the corresponding N-oxide. When a heteroaryl is substituted by a hydroxy group, it also includes its corresponding tautomer. The terms “heteroaryl” and “heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, and/or heterocyclic rings. Nonlimiting examples of heteroaryl groups include thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, pteridinyl, indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-1,4-oxazin-3(4H)-one. The term “heteroaryl” may be used interchangeably with the terms “heteroaryl ring”, “heteroaryl group”, or “heteroaromatic”, any of which terms include rings that are optionally substituted. The term “heteroaralkyl” refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted. As used herein, the terms “heterocycle”, “heterocyclyl”, “heterocyclic radical”, and “heterocyclic ring” are used interchangeably and refer to a stable 3- to 8-membered monocyclic or 7-10-membered bicyclic moiety that is either saturated or partially unsaturated in at least one ring, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothienyl, piperidinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and thiamorpholinyl. Unless otherwise specified, a heterocyclyl group may be mono-, bi-, tri-, or polycyclic, preferably mono-, bi-, or tricyclic, more preferably mono- or bicyclic. Additionally, a heterocyclic ring also includes groups in which the heterocyclic ring is fused to one or more aryl, heteroaryl and/or carbocyclic rings. As used herein, the term “partially unsaturated” refers to a ring moiety that includes at least one double or triple bond between ring atoms. The term “partially unsaturated” is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aromatic (e.g., aryl or heteroaryl) moieties, as herein defined. An aryl or heteroaryl group may contain one or more substituents (e.g., 1, 2, 3, or 4 substituents) and thus may be “optionally substituted”. In addition to the substituents defined above and herein, suitable substituents on the unsaturated carbon atom of an aryl or heteroaryl group also include and are generally selected from -halo, —NO2, —CN, —R+, —C(R+)═C(R+)2, —C≡C—R+, —OR+, —SR∘, —S(O)R∘, —SO2R∘, —SO3R+, —SO2N(R+)2, —N(R+)2, —NR+C(O)R+, —NR+C(S)R+, —NR+C(O)N(R+)2, —NR+C(S)N(R+)2, —N(R+)C(═NR+)—N(R+)2, —N(R+)C(═NR+)—R∘, —NR+CO2R+, —NR+SO2R∘, —NR+SO2N(R+)2, —O—C(O)R+, —O—CO2R+, —OC(O)N(R+)2, —C(O)R+, —C(S)R∘, —CO2R+, —C(O)—C(O)R+, —C(O)N(R+)2, —C(S)N(R+)2, —C(O)N(R+)—OR+, —C(O)N(R+)C(═NR+)—N(R+)2, —N(R+)C(═NR+)—N(R+)—C(O)R+, —C(═NR+)—N(R+)2, —C(═NR)—OR+, —N(R+)—N(R+)2, —C(═NR+)—N(R+)—OR+, —C(R∘)═N—OR+, —P(O)(R+)2, —P(O)(OR+)2, —O—P(O)—OR+, and —P(O)(NR+)—N(R+)2, wherein R+, independently, is hydrogen or an aliphatic, aryl, heteroaryl, cycloaliphatic, or heterocyclyl group, or two independent occurrences of R+that are bound to the same atom are taken together with their intervening atom(s) to form an optionally substituted 5-7-membered aryl, heteroaryl, cycloaliphatic, or heterocyclyl ring. When R+ is not hydrogen, R+ may be unsubstituted or substituted with 1, 2, 3, or 4 substituent groups. Each R∘is an aliphatic, aryl, heteroaryl, cycloaliphatic, or heterocyclyl group, wherein R∘is unsubstituted or substituted with 1, 2, 3, or 4 substituent groups. An alkenyl, a carbocyclic ring, or a heterocyclic ring may contain one or more substituents and thus may be “optionally substituted”. Unless otherwise defined above and herein, suitable substituents on any saturated carbon of an alkenyl, a carbocyclic ring, or a heterocyclic ring are selected from those listed above for the carbon atoms of an aryl or heteroaryl group and additionally include the following: ═O, ═S, ═C(R*)2, ═N—N(R*)2, ═N—OR*, ═N—NHC(O)R*, ═N—NHCO2R∘═N—NHSO2R∘or ═N—R* where R∘is defined above, and each R* is independently selected from hydrogen or an C1-6aliphatic group that is unsubstituted or substituted with 1, 2, 3, or 4 substituent groups. In addition to the substituents defined above and herein, optional substituents on the nitrogen of a non-aromatic heterocyclic ring also include and are generally selected from —R+, —N(R+)2, —C(O)R+, —C(O)OR+, —C(O)C(O)R+, —C(O)CH2C(O)R+, —S(O)2R+, —S(O)2N(R+)2, —C(S)N(R+)2, —C(═NH)—N(R+)2, or —N(R+)S(O)2R+; wherein each R+is defined above. A ring nitrogen atom of a heteroaryl or non-aromatic heterocyclic ring also may be oxidized to form the corresponding N-hydroxy or N-oxide compound. A nonlimiting example of such a heteroaryl having an oxidized ring nitrogen atom is N-oxidopyridyl. As detailed above, in some embodiments, two independent occurrences of R+(or any other variable similarly defined in the specification and claims herein) that are bound to the same atom, can be taken together with their intervening atom(s) to form a monocyclic or bicyclic ring selected from 3-13-membered cycloaliphatic, 3-12-membered heterocyclyl having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, 6-10-membered aryl, or 5-10-membered heteroaryl having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. With respect to the compounds defined by generic Formula I or II, unless otherwise specified, one or more hydrogens can be replaced by deuterium. Isotopic enrichments include e.g., at least 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 87%, 98%, 99.0%, 99.5% and 99.8%. In one embodiment, all hydrogen atoms represented in Formula I and II are present in natural abundance. With respect to specific compounds disclosed herein, such as those in Table 1 and in the Exemplification section, all hydrogen atoms are present in natural abundance unless otherwise specified. It is to be understood that, when a disclosed compound has at least one chiral center, the present invention encompasses one enantiomer free from the corresponding optical isomer, racemic mixture of the compound and mixtures enriched in one enantiomer relative to its corresponding optical isomer. When a mixture is enriched in one enantiomer relative to its optical isomers, the mixture contains, for example, an enantiomeric excess of at least 50%, 75%, 90%, 95% 99% or 99.5%. The enantiomers of the present invention may be resolved by methods known to those skilled in the art, for example by formation of diastereoisomeric salts which may be separated, for example, by crystallization; formation of diastereoisomeric derivatives or complexes which may be separated, for example, by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic esterification; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support for example silica with a bound chiral ligand or in the presence of a chiral solvent. Where the desired enantiomer is converted into another chemical entity by one of the separation procedures described above, a further step is required to liberate the desired enantiomeric form. Alternatively, specific enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation. When a disclosed compound has at least two chiral centers, the present invention encompasses a diastereomer free of other diastereomers, a pair of diastereomers free from other diastereomeric pairs, mixtures of diastereomers, mixtures of diastereomeric pairs, mixtures of diastereomers in which one diastereomer is enriched relative to the other diastereomer(s) and mixtures of diastereomeric pairs in which one diastereomeric pair is enriched relative to the other diastereomeric pair(s). When a mixture is enriched in one diastereomer or diastereomeric pair(s) relative to the other diastereomers or diastereomeric pair(s), the mixture is enriched with the depicted or referenced diastereomer or diastereomeric pair(s) relative to other diastereomers or diastereomeric pair(s) for the compound, for example, by a molar excess of at least 50%, 75%, 90%, 95%, 99% or 99.5%. The diastereoisomeric pairs may be separated by methods known to those skilled in the art, for example chromatography or crystallization and the individual enantiomers within each pair may be separated as described above. Specific procedures for chromatographically separating diastereomeric pairs of precursors used in the preparation of compounds disclosed herein are provided the examples herein. 3. Description of Exemplary Compounds As described generally above, in some embodiments the present invention provides a compound of formula I: or a pharmaceutically acceptable salt thereof, wherein:ring A is selected from R1is an optionally substituted monocyclic or bicyclic heterocyclyl;R2is selected from C2-C6alkenyl, heteroaryl, partially unsaturated heterocyclyl, partially unsaturated carbocyclyl, and para-substituted phenyl, and when ring A comprises two nitrogen atoms, R2is additionally selected from unsubstituted phenyl,wherein R2is optionally further substituted, and wherein any two substituents on adjacent ring atoms in R2are optionally taken together with the adjacent ring atoms to form a ring that is an aryl, a carbocyclyl, a heteroaryl, or a heterocyclyl ring;R3, when present, is selected from chloro, fluoro, —CF3and —CHF2;n is 0 or 1;“1” represents a point of attachment between ring A to —NH—C(O)—R1;“2” represents a point of attachment between ring A and R2;“3” represents a point of attachment between ring A and —NH2. In some embodiments, the compound is other than: In some embodiments, ring A is heteroaromatic. For example, ring A is selected from: In other embodiments, n is 0. In still other embodiments, nisi. In other embodiments, ring A is selected from: In further embodiments, ring A is selected from In some embodiments, ring A is selected from: In some embodiments, ring A is selected from any of the ring A moieties in the compounds set forth in Table 1. In some embodiments, R1is unsubstituted. In other embodiments, R1is substituted (e.g., R1comprises 1, 2, 3, or 4 additional substituents as described herein). In some embodiments, R1is selected from 5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-yl, pyrrolidin-1-yl, 1,3-dihydro-2H-pyrrolo[3,4-c]pyridin-2-yl, 6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-6-yl, tetrahydro-2H-pyran-4-yl, hexahydrocyclopenta[c]pyrrol-2(1H)-yl, 7-oxa-2-azaspiro[3.5]nonan-2-yl, isoindolin-2-yl, 4,5,6,7-tetrahydrothiazolo[4,5-c]pyridin-2-yl, 2-oxa-7-azaspiro[3.5]nonan-7-yl, 2-oxa-6-azaspiro[3.3]heptan-6-yl, and 2-oxa-6-azaspiro[3.4]octan-6-yl, wherein R1is optionally substituted with up to 3 independently selected substituents. In further embodiments, R1is selected from 5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-yl, pyrrolidin-1-yl, 1,3-dihydro-2H-pyrrolo[3,4-c]pyridin-2-yl, 6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-6-yl, tetrahydro-2H-pyran-4-yl, hexahydrocyclopenta[c]pyrrol-2(1H)-yl, 7-oxa-2-azaspiro[3.5]nonan-2-yl, isoindolin-2-yl, 4,6-difluoroisoindolin-2-yl, 4,7-difluoroisoindolin-2-yl, 4-fluoroisoindolin-2-yl, 5-fluoroisoindolin-2-yl, 4-chlorobsoindolin-2-yl, 4-methoxyisoindolin-2-yl, 5-methoxyisoindolin-2-yl, 5-chloroisoindolin-2-yl, 4-trifluoromethylisoindolin-2-yl, 5,6-difluoroisoindolin-2-yl, 5-trifluoromethylisoindolin-2-yl, 5-((4-methylpiperazin-1-yl)methyl)isoindolin-2-yl, 3-fluoro-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-6-yl, 5-(cyclopropylmethyl)-4,5,6,7-tetrahydrothiazolo[4,5-c]pyridin-2-yl, 4,5,6,7-tetrahydrothiazolo[4,5-c]pyridin-2-yl, 2-oxa-7-azaspiro[3.5]nonan-7-yl, 2-oxa-6-azaspiro[3.3]heptan-6-yl, 2-oxa-6-azaspiro[3.4]octan-6-yl, and 3-((4-methylpiperazin-1-yl)methyl)-6,7-dihydro-5H-cyclopenta[c]pyridin-6-yl. In some embodiments, R2is unsubstituted. In other embodiments, R2is substituted (e.g., R2comprises 1, 2, 3, or 4 additional substituents as described herein). In still other embodiments, R2is selected from —C2-C4alkenyl, phenyl, 4-substituted phenyl, pyridin-4-yl, isoxazol-5-yl, oxazol-5-yl, isothiazol-5-yl, thiazol-5-yl, 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl, 4,5,6,7-tetrahydropyrazolo[1,5-a]pyridin-3-yl, furan-3-yl, furan-2-yl, cyclopent-1-ene-1-yl, and 2,5-dihydrofuran-3-yl. In certain embodiments, R2is selected from —C(CH3)═CH2, phenyl, 4-fluorophenyl, 4-difluoromethoxyphenyl, 4-methylphenyl, 3,4-difluorophenyl, pyridin-4-yl, isoxazol-5-yl, oxazol-5-yl, isothiazol-5-yl, thiazol-5-yl, 1-methyl-1H-pyrazol-4-yl, 1-(2-methylpropyl)-1H-pyrazol-4-yl, 1-trifluoromethyl-1H-pyrazol-4-yl, 1,5-dimethyl-1H-pyrazol-4-yl, 1-cyclobutyl-1H-pyrazol-4-yl, 1-cyclopentyl-1H-pyrazol-4-yl, 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl, 4,5,6,7-tetrahydropyrazolo[1,5-a]pyridin-3-yl, furan-3-yl, furan-2-yl, 5-methylfuran-2-yl, 5-methylfuran-3-yl, cyclopent-1-ene-1-yl, and 2,5-dihydrofuran-3-yl. As described generally above, in some embodiments the present invention also provides a compound of formula II: or a pharmaceutically acceptable salt thereof, whereinring A′B′ is a fused bicyclic ring system containing at least two nitrogen atoms, wherein ring A′ is a 6-membered heterocyclyl and ring B′ is a 5-membered heteroaryl;X1is carbon or nitrogen;R3′and R4are each independently halo, (C1-C4)alkyl, halo(C1-C4)alkyl, (C1-C4)alkoxy, or halo(C1-C4)alkoxy;R5is halo, halo(C1-C4)alkyl, (C1-C4)alkoxy, halo(C1-C4)alkoxy, monocyclic heterocyclyl, or (C1-C4)alkyl optionally substituted with monocyclic heterocyclyl, wherein each of said heterocyclyl are optionally and independently substituted with 1 to 2 groups selected from halo, (C1-C4)alkyl, and halo(C1-C4)alkyl;n′ is 0 or 1; andp and t are each independently 0, 1, or 2. In certain embodiments, the compound of Formula II is of the Formula: or a pharmaceutically acceptable salt thereof. In other embodiments, the compound of Formula II is of the Formula: or a pharmaceutically acceptable salt thereof. In certain embodiments of Formula II and those described in preceding paragraphs 53 and 54, ring A′B′ is selected from wherein the remaining values are as described above for Formula II. Alternatively ring A′B′ is selected from wherein the remaining values are as described above for Formula II. In certain embodiments of Formula II and those described in preceding paragraphs 53 and 54, p is 0 or 1, wherein the remaining values are as described above for Formula II and the embodiment of paragraph 55. In certain embodiments of Formula II and those described in preceding paragraphs 53 and 54, R4, if present, is halo, wherein the remaining values are as described above for Formula II and the embodiment of paragraph 55 or 56. In certain embodiments of Formula II and those described in preceding paragraphs 53 and 54, t is 0 or 1, wherein the remaining values are as described above for Formula II and the embodiment of paragraph 55, 56, or 57. In certain embodiments of Formula II and those described in preceding paragraphs 53 and 54, R5, if present, is selected from halo(C1-C4)alkyl and (C1-C4)alkyl, wherein the remaining values are as described above for Formula II and the embodiment of paragraph 55, 56, 57, or 58. In certain embodiments of Formula II and those described in preceding paragraphs 53 and 54, ring A′B′ and (R5)ttaken together are selected from wherein the remaining values are as described above for Formula II and the embodiment of paragraph 55, 56, 57, 58, or 59. Although, as indicated above, various embodiments and aspects thereof for a variable in any of the formulas described herein (e.g., a compound of Formula I, II or any of compounds 100-128 or any of those in Tables 2 or 3) may be selected from a group of chemical moieties, the invention also encompasses as further embodiments and aspects thereof situations where such variable is: a) selected from any subset of chemical moieties in such a group; and b) any single member of such a group. Further, where various embodiments and aspects thereof are set forth individually for each variable in any of the formulas described herein (e.g., a compound of Formula I, II or any of compounds 100-128 or any of those in Tables 2 or 3), the invention encompasses all possible combinations of the different embodiments and aspects for each of the variables in the Formula. Exemplary compounds of and useful in the present invention are set forth in Table 1 below. In certain embodiments, the present invention provides a compound depicted in Table 1, or a pharmaceutically acceptable salt thereof. TABLE 1Exemplary CompoundsMSMSNo.StructureCalc.found1H NMR Data (400 MHz, DMSO-d6)100366367δ 8.61 (s, 1H), 8.50 (d, J = 5.2 Hz, 1H), 7.78 (s, 1H), 7.48-7.43 (m, 3H), 7.24 (t, J = 8.8 Hz, 2H), 7.18 (d, J = 8.8 Hz, 1H), 6.58 (d, J = 12.8 Hz, 1H), 5.39 (br, 2H), 4.80 (d, J = 6.4 Hz, 4H).101320321δ 8.61 (s, 1H), 8.50 (d, J = 5.2 Hz, 1H), 7.68 (s, 1H), 7.62 (d, J = 7.6 Hz, 2H), 7.50 (t, J = 7.6 Hz, 2H), 7.43 (d, J = 5.2 Hz, 1H), 7.39 (s, 1H), 7.33 (t, J = 7.6 Hz, 1H), 5.09 (s, 2H), 4.77 (d, J = 6.4 Hz, 4H).102338339δ 8.61 (s, 1H), 8.50 (d, J = 5.2 Hz, 1H), 7.68 (s, 1H), 7.65-7.62 (m, 2H), 7.43 (d, J = 5.2 Hz, 1H), 7.38 (s, 1H), 7.34 (t, J = 8.8 Hz, 2H), 5.01 (br, 2H), 4.76 (d, J = 6.0 Hz, 4H).103321322δ 8.62-8.61 (m, 3H), 8.50 (d, J = 5.2 Hz, 1H), 7.78 (d, J = 4.8 Hz, 2H), 7.74 (s, 1H), 7.53 (s, 1H), 7.44 (d, J = 4.0 Hz, 1H), 5.39 (br, 2H), 4.78 (d, J = 6.0 Hz, 4H)104340341δ 8.07 (d, J = 2.4 Hz, 1H), 7.69 (d, J = 2.0 Hz, 1H), 7.60-7.56 (m, 2H), 7.51 (s, 1H), 7.24 (t, J = 8.8 Hz, 2H), 5.84 (br, 2H), 3.64-3.59 (m, 2H), 3.18 (dd, J = 10.8 Hz, 4.0 Hz, 2H), 2.67-2.64 (m, 2H), 1.83-1.69 (m, 3H), 1.62-1.52 (m, 1H), 1.48- 1.43 (m, 2H).105300301δ 8.07 (s, 1H), 7.91-7.87 (m, 3H), 7.60 (s, 1H), 7.23 (t, J = 8.8 Hz, 2H), 5.18 (br, 2H), 3.42 (t, J = 6.8 Hz, 4H), 1.88 (t, J = 6.4 Hz, 4H).106315316δ 9.30 (br, 1H), 8.12 (s, 1H), 8.09 (s, 1H), 7.90-7.86 (m, 2H), 7.24 (t, J = 8.8 Hz, 2H), 5.29 (br, 2H), 3.94- 3.90 (m, 2H), 3.40-3.36 (m, 2H), 2.73-2.70 (m, 1H), 1.77-1.66 (m, 4H).107300301δ 8.21 (br, 1H), 799-7.95 (m, 2H), 7.54 (d, J = 8.4 Hz, 1H), 7.21 (t, J = 8.8 Hz, 2H), 7.17 (d, J = 8.0 Hz, 1H), 5.11 (br, 2H), 3.39-3.38 (m, 4H), 1.86 (s, 4H).108283284CD3OD δ 8.57 (d, J = 6.4 Hz, 2H), 8.20 (s, 1H), 7.99 (s, 1H), 7.90 (d, J = 6.4 Hz, 2H), 3.55 (t, J = 6.4 Hz, 4H), 2.02 (s, 4H).109315316δ 10.03 (br, 1H), 7.98-7.94 (m, 2H), 7.61 (d, J = 8.0 Hz, 1H), 7.24 (t, J = 8.8 Hz, 3H), 5.07 (br, 2H), 3.91 (dd, J = 11.6 Hz, 2.0 Hz, 2H), 3.38-3.32 (m, 2H), 2.80-2.74 (m, 1H), 1.78- 1.64 (m, 4H).110301302δ 8.71 (br, 1H), 8.27-8.2 (m, 2H), 8.20 (s, 1H), 7.26 (t, J = 8.8 Hz, 2H), 5.23 (br, 2H), 3.43 (s, 4H), 1.87 (s, 4H)111283284δ 8.56 (m, 2H), 8.30 (m, 1H), 7.90 (m, 2H), 7.25 (d, J = 3.6 Hz, 1H), 7.17 (d, J = 4.0 Hz, 1H), 5.40 (s, 2H), 3.40-3.37 (m, 4H), 1.86 (m, 4H).112317318δ 7.46-7.43 (m, 2H), 7.35 (br, 1H), 7.22 (t, J = 8.8 Hz, 2H), 7.06 (t, J = 8.4 Hz, 1H), 6.59 (d, J = 8.4 Hz, 1H), 5.25 (br, 2H), 3.35-3.34 (m, 4H), 1.85 (s, 4H).113367368δ 8.54 (s, 2H), 7.81 (s, 1H), 7.48- 7.45 (m, 2H), 7.24 (t, J = 8.8 Hz, 2H), 7.11 (t, J = 8.4 Hz, 1H), 6.61 (d, J = 8.4 Hz, 1H), 5.43 (br, 2H), 4.81 (s, 4H).114317318δ 7.45 (t, J = 6.4 Hz, 3H), 7.23 (t, J = 9.0 Hz, 2H), 7.14 (d, J = 8.8 Hz, 1H), 6.57 (d, J = 13.2 Hz, 1H), 5.26 (s, 2H), 3.36 (t, J = 6.6 Hz, 4H), 1.85 (t, J = 6.4 Hz, 4H).115332333δ 9.07 (s, 1H), 7.45 (t, J = 6.6 Hz, 2H), 7.30 (d, J = 8.4 Hz, 1H), 7.24 (t, J = 8.8 Hz, 2H), 6.59 (d, J = 13.2 Hz, 1H), 5.30 (s, 2H), 3.90 (d, J = 10.8 Hz, 2H), 3.38-3.35 (m, 2H), 2.66-2.59 (m, 1H), 1.75-1.61 (m, 4H).116335336δ 7.50-7.43 (m, 3H), 7.27 (bs, 1H), 7.18 (d, J = 8.8 Hz, 1H), 6.57 (d, J = 13.6 Hz, 1H), 5.35 (s, 2H), 3.36 (t, J = 6.4 Hz, 4H), 1.85 (t, J = 6.4 Hz, 4H).117335336δ 7.49-7.42 (m, 2H), 7.35 (br, 1H), 7.28-7.26 (m, 1H), 7.11 (t, J = 8.4 Hz, 1H), 6.59 (d, J = 8.4 Hz, 1H), 5.35 (br, 2H), 3.37 (s, 4H), 1.85 (s, 4H).118332333δ 9.02 (br, 1H), 7.46-7.43 (m, 2H), 7.23 (t, J = 8.4 Hz, 2H), 7.10 (t, J = 8.4 Hz, 1H), 6.61 (t, J = 8.4 Hz, 1H), 5.23 (br, 2H), 3.91-3.88 (m, 2H), 3.38-3.35 (m, 2H), 2.68-2.62 (m, 1H), 1.78-1.61 (m, 4H).119367368CD3OD δ 8.41 (s, 2H), 7.41-3.67 (m, 2H), 7.08 (d, J = 8.4 Hz, 1H), 7.04-6.98 (m, 2H), 6.52 (d, J = 12.4 Hz, 1H), 4.79 (s, 4H).120350351δ 8.66 (s, 1H), 8.55 (s, 2H), 8.00- 7.96 (m, 2H), 7.58 (d, J = 8.4 Hz, 1H), 7.23 (t, J = 8.8 Hz, 2H), 7.17 (d, J = 8.4 Hz, 1H), 5.21 (s, 2H), 4.85 (br, 4H).121351352δ 9.14 (br, 1H), 8.56 (s, 2H), 8.29- 8.25 (m, 3H), 7.27 (t, J = 8.8 Hz, 2H), 5.31 (br, 2H), 4.88 (br, 4H).122320321CD3OD δ 8.47 (s, 1H), 8.39 (d, J = 5.2 Hz, 1H), 7.65 (s, 1H), 7.53 (d, J = 8.0 Hz, 2H), 7.38 (d, J = 5.2 Hz, 1H), 7.32 (t, J = 8.0 Hz, 2H), 7.11 (t, J = 7.4 Hz, 1H), 4.82 (s, 4H). TABLE 2Exemplary CompoundsMSMSNo.StructureCalc.found1H NMR Data (400 MHz, DMSO-d6)123452453δ 8.49 (s, 1H), 7.73-7.58 (m, 3H), 7.52 (s, 1H), 7.26 (dd, J = 15.5, 6.7 Hz, 3H), 4.03 (s, 2H), 3.77 (s, 3H), 3.48-3.34 (m, 4H), 3.30-3.19 (m, 1H), 3.12 (d, J = 10.1 Hz, 1H), 2.45- 2.36 (m, 1H), 2.15 (d, J = 75.5 Hz, 3H), 1.81 (s, 1H), 1.53 (dd, J = 54.6, 29.6 Hz, 4H), 1.32 (s, 1H).124478479δ 8.51 (s, 1H), 7.66 (s, 1H), 7.65-7.60 (m, 2H), 7.24 (dd, J = 17.5, 8.6 Hz, 3H), 6.89 (d, J = 7.3 Hz, 2H), 6.83- 6.73 (m, 1H), 4.02 (s, 2H), 3.73 (s, 3H), 3.46 (m, 4H), 3.28 (d, J = 9.6 Hz, 1H), 3.16 (d, J = 10.3 Hz, 1H), 2.45- 2.35 (m, 1H), 2.19 (d, J = 54.6 Hz, 3H), 1.86 (s, 1H), 1.55 (m, 4H), 1.36 (s, 1H).125448449δ 8.97 (s, 1H), 7.68 (s, 1H), 7.67- 7.59 (m, 2H), 7.31-7.25 (m, 4H), 7.23-7.17 (m, 3H), 3.94 (s, 2H), 3.76-2.6 (m, 2H), 3.51-3.46 (m, 6H), 2.39-2.31 (m, 2H), 1.84- 1.40 (m, 1H), 1.25-1.01 (m, 1H).126299300δ 10.64 (s, 1H), 7.92-7.74 (m, 2H), 7.67 (dd, J = 8.6, 1.0 Hz, 2H), 7.52-7.36 (m, 2H), 7.20 (t, J = 7.4 Hz, 1H), 4.08 (s, 2H), 2.56 (d, J = 0.9 Hz, 3H).128436437δ 8.54 (s, 1H), 8.50 (s, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.38 (s, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.31-5.10 (m, 3H), 4.81 (s, 4H), 3.77 (s, 2H), 3.68-3.57 (m, 2H), 3.29-3.24 (m, 1H), 3.24- 3.17 (m, 1H).129327328δ 8.80 (s, 1H), 7.69 (s, 1H), 7.61 (d, J = 7.7 Hz, 2H), 7.40 (t, J = 8.0 Hz, 2H), 7.14 (t, J = 7.4 Hz, 1H), 4.05 (s, 2H), 3.70 (s, 4H), 3.61-3.46 (m, 4H), 1.80-1.63 (m, 4H).130341342δ 8.88 (s, 1H), 7.69 (s, 1H), 7.61 (d, J = 7.7 Hz, 2H), 7.40 (t, J = 8.0 Hz, 2H), 7.14 (t, J = 7.4 Hz, 1H), 3.94 (s, 2H), 3.76 (t, J = 7.1 Hz, 2H), 3.58-3.49 (m, 2H), 3.47 (s, 2H), 3.43-3.34 (m, 2H), 1.74 (t, J = 7.1 Hz, 2H), 1.49 (t, J = 5.5 Hz, 4H).131355356δ 8.86 (s, 1H), 7.71 (s, 1H), 7.70- 7.62 (m, 2H), 7.43-7.34 (m, 1H), 7.32-7.19 (m, 3H), 7.15 (t, J = 8.9 Hz, 1H), 4.81 (s, 4H), 4.05 (s, 2H).132356357CD3OD δ 8.41 (s, 1H), 7.73 (s, 1H), 7.71- 7.61 (m, 3H), 7.24-7.14 (m, 2H), 4.90 (s, 2H), 4.84 (s, 2H).133355356δ 8.82 (s, 1H), 7.71 (s, 1H), 7.69- 7.62 (m, 2H), 7.39 (dd, J = 8.3, 5.1 Hz, 1H), 7.25 (dd, J = 18.3, 9.4 Hz, 3H), 7.15 (t, J = 8.9 Hz, 1H), 4.74 (d, J = 12.1 Hz, 4H), 4.05 (s, 2H).134338339δ 8.89 (s, 1H), 8.48 (d, J = 3.7 Hz, 1H), 7.81 (d, J = 8.2 Hz, 1H), 7.71 (s, 1H), 7.70-7.61 (m, 2H), 7.33 (dd, J = 7.7, 4.9 Hz, 1H), 7.27 (t, J = 8.8 Hz, 2H), 4.77 (d, J = 9.6 Hz, 4H), 4.06 (s, 2H).135317318δ 8.86 (s, 1H), 7.67 (s, 1H), 7.63 (dd, J = 8.9, 4.7 Hz, 2H), 7.26 (t, J = 8.8 Hz, 2H), 4.66 (d, J = 14.3 Hz, 4H), 4.09 (d, J = 19.7 Hz, 4H), 4.02 (d, J = 11.6 Hz, 2H).136304305CD3OD δ 7.72 (s, 1H), 7.69-7.58 (m, 2H), 7.29-7.07 (m, 2H), 4.03 (dd, J = 10.2, 2.8 Hz, 2H), 3.52 (td, J = 11.4, 3.1 Hz, 2H), 2.83-2.65 (m, 1H), 1.96-1.75 (m, 4H).137345346δ 8.88 (s, 1H), 7.67 (s, 1H), 7.66- 7.58 (m, 2H), 7.25 (t, J = 8.8 Hz, 2H), 4.33 (s, 4H), 3.92 (s, 2H), 3.45- 3.35 (m, 4H), 1.81-1.70 (m, 4H).139339340δ 8.98 (s, 1H), 8.55 (s, 2H), 7.73 (s, 1H), 7.71-7.61 (m, 2H), 7.28 (t, J = 8.8 Hz, 2H), 4.78 (s, 4H), 4.06 (s, 2H).140423424CD3OD δ 7.80 (dd, J = 8.7, 5.5 Hz, 2H), 7.39 (d, J = 8.2 Hz, 1H), 7.18 (d, J = 8.2 Hz, 1H), 7.03 (t, J = 8.8 Hz, 2H), 3.65 (dd, J = 10.7, 8.1 Hz, 1H), 3.56 (dd, J = 10.6, 7.7 Hz, 1H), 3.39- 3.31 (m, 1H), 3.28-3.23 (m, 1H), 2.86-2.59 (m, 8H), 2.37-2.32 (m, 0.5H), 2.26-2.08 (m, 1.5H), 1.86-1.71 (m, 5H), 1.58-1.46 (m, 1H),1.17-1.09 (m, 1H).141337338δ 8.56 (s, 1H), 7.75-7.60 (m, 3H), 7.39-7.02 (m, 3H), 4.05 (s, 2H), 3.36 (s, 4H), 1.85 (s, 4H).142289290δ 8.54 (s, 1H), 7.74-7.54 (m, 3H), 7.25 (t, J = 8.8 Hz, 2H), 4.03 (s, 2H), 3.40-3.34 (m, 4H), 1.85 (s, 4H).143395396CDCl3δ 7.82 (s, 2H), 7.32 (s, 1H), 7.10 (t, J = 8.7 Hz, 3H), 6.81 (s, 1H), 4.57 (s, 2H), 3.74-3.70 (m, 2H), 3.54- 3.44 (m, 2H), 3.38 (s, 4H), 2.97 (s, 1H), 2.74 (s, 2H), 2.19-2.11 (m, 2H), 2.08-2.00 (m, 2H), 1.50- 1.38 (m, 2H).144420421δ 8.53 (s, 1H), 8.38 (s, 1H), 7.72 (s, 1H), 7.52 (d, J = 8.2 Hz, 1H), 7.48 (dd, J = 3.6, 1.1 Hz, 1H), 7.41 (dd, J = 5.0, 1.0 Hz, 1H), 7.12 (d, J = 8.2 Hz, 1H), 7.06 (dd, J = 5.0, 3.6 Hz, 1H), 5.18 (s, 2H), 4.77 (s, 4H), 3.62 (s, 2H), 2.44 (s, 4H), 1.70 (d, J = 3.3 Hz, 4H).145394395δ 8.54 (s, 1H), 8.37 (s, 1H), 7.71 (s, 1H), 7.52 (d, J = 8.2 Hz, 1H), 7.48 (dd, J = 3.6, 1.0 Hz, 1H), 7.41 (dd, J = 5.0, 1.0 Hz, 1H), 7.12 (d, J = 8.2 Hz, 1H), 7.06 (dd, J = 5.0, 3.7 Hz, 1H), 5.18 (s, 2H), 4.77 (s, 4H), 3.44 (s, 2H), 2.16 (s, 6H).146419420δ 8.58 (s, 1H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.54 (d, J = 8.2 Hz, 1H), 7.36 (d, J = 8.1 Hz, 2H), 7.29 (d, J = 8.0 Hz, 2H), 7.22 (t, J = 8.9 Hz, 2H), 7.16 (d, J = 8.2 Hz, 1H), 5.19 (s, 2H), 4.37 (t, J = 8.4 Hz, 2H), 3.97-3.89 (m, 2H), 3.85-3.80 (m, 1H), 3.43 (s, 2H), 2.18 (s, 6H).147404405δ 8.49 (s, 1H), 8.07 (s, 1H), 7.98 (dd, J = 8.8, 5.7 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 6.36 (s, 1H), 5.17 (s, 2H), 4.68 (s, 4H), 3.92 (t, J = 7.3 Hz, 4H), 2.36-2.25 (m, 2H).148444445δ 8.55 (d, J = 8.5 Hz, 2H), 8.03- 7.92 (m, 2H), 7.57 (d, J = 7.9 Hz, 2H), 7.22 (t, J = 8.7 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 6.70 (s, 1H), 5.19 (s, 2H), 4.81 (s, 4H), 3.06 (s, 2H), 2.58 (s, 4H), 2.29 (s, 3H).149446447CD3OD δ 8.43 (s, 1H), 7.81 (s, 2H), 7.42 (s, 1H), 7.30 (s, 1H), 7.21 (s, 1H), 7.03 (t, J = 8.7 Hz, 2H), 4.81 (s, 4H), 3.41-3.27 (m, 2H), 2.90 (s, 1H), 2.78 (s, 2H), 2.64 (s, 3H), 1.98 (m, 4H).150434435δ 8.50 (s, 1H), 8.15 (s, 1H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 6.86 (s, 1H), 5.18 (s, 2H), 4.70 (s, 4H), 3.71 (t, J = 4.0 Hz 4H), 3.43 (t, J = 4.0 Hz, 4H).151363364δ 8.54 (s, 1H), 8.46 (s, 1H), 7.98 (dd, J = 8.8, 5.7 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.29 (s, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.78 (s, 4H), 3.33- 3.32 (s, 3H).152410411CDCl3δ 7.83 (s, 2H), 7.36 (d, J = 7.4 Hz, 1H), 7.11 (dd, J = 17.7, 9.0 Hz, 3H), 6.77 (s, 1H), 4.59 (s, 2H), 3.77 (s, 2H), 3.58 (s, 2H), 3.44 (d, J = 9.9 Hz, 2H), 3.20-3.12 (m, 1H), 3.06 (s, 2H), 2.96 (s, 2H), 2.63 (s, 2H), 2.46 (d, J = 8.6 Hz, 2H), 2.42 (s, 3H).153383384δ 10.32 (s, 1H), 7.96 (dd, J = 8.8, 5.6 Hz, 2H), 7.66 (d, J = 8.3 Hz, 1H), 7.22 (t, J = 8.5 Hz, 3H), 5.31 (s, 2H), 3.62 (s, 2H), 2.97 (t, J = 5.3 Hz, 2H), 2.81-2.63 (m, 2H), 2.43 (s, 3H).154406407δ 8.56 (s, 1H), 8.38 (s, 1H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.72 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.78 (s, 4H), 3.45 (s, 2H), 2.17 (s, 6H).155432433δ 8.56 (s, 1H), 8.40 (s, 1H), 8.03- 7.94 (m, 2H), 7.73 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.78 (s, 4H), 3.64 (s, 2H), 2.47 (s, 4H), 1.71 (s, 4H).156338339δ 8.65 (s, 1H), 8.55 (s, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.49 (dd, J = 3.6, 1.0 Hz, 1H), 7.41 (dd, J = 5.0, 1.0 Hz, 1H), 7.12 (d, J = 8.2 Hz, 1H), 7.06 (dd, J = 5.0, 3.7 Hz, 1H), 5.20 (s, 2H), 4.84 (s, 4H).157404405δ 8.50 (s, 1H), 8.02-7.93 (m, 2H), 7.69 (d, J = 2.5 Hz, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 6.84 (d, J = 2.3 Hz, 1H), 5.17 (s, 2H), 4.68 (d, J = 21.5 Hz, 4H), 3.87 (t, J = 7.2 Hz, 4H), 2.37-2.33 (m, 2H).158434435δ 8.53 (s, 1H), 8.21 (d, J = 2.3 Hz, 1H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.38 (s, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.71 (d, J = 20.8 Hz, 4H), 3.76 (t, J = 4.0 Hz 4H), 3.17 (t, J = 4.0 Hz 4H).159447448δ 8.52 (s, 1H), 8.20 (d, J = 2.4 Hz, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.37 (s, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.17 (s, 2H), 4.70 (d, J = 21.9 Hz, 4H), 3.20 (s, 4H), 2.51 (s, 4H), 2.26 (s, 3H).160447448δ 8.50 (s, 1H), 8.12 (s, 1H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 6.85 (s, 1H), 5.18 (s, 2H), 4.65 (d, J = 30.1 Hz, 4H), 3.48 (s, 4H), 2.42 (s, 4H), 2.23 (s, 3H).161389390δ 9.15 (d, J = 1.8 Hz, 1H), 8.60 (s, 1H), 8.48 (dd, J = 4.7, 1.5 Hz, 1H), 8.37 (s, 1H), 8.28 (d, J = 8.1 Hz, 1H), 7.72 (s, 1H), 7.68 (d, J = 8.2 Hz, 1H), 7.42 (dd, J = 7.9, 4.6 Hz, 1H), 7.19 (d, J = 8.2 Hz, 1H), 5.31 (s, 2H), 4.79 (s, 4H), 3.44 (s, 2H), 2.16 (s, 6H).162406407δ 8.56 (s, 1H), 8.52 (s, 1H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.46 (s, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.82 (s, 4H), 3.65 (s, 2H), 2.27 (s, 6H).163355356δ 10.28 (s, 1H), 7.68 (d, J = 1.0 Hz, 1H), 7.46 (d, J = 8.3 Hz, 1H), 7.20 (d, J = 8.3 Hz, 1H), 6.74 (d, J = 2.9 Hz, 1H), 6.55 (dd, J = 3.3, 1.8 Hz, 1H), 5.34 (s, 2H), 3.62 (s, 2H), 2.96 (d, J = 5.5 Hz, 2H), 2.72 (t, J = 5.6 Hz, 2H), 2.43 (s, 3H).164398399δ 10.36 (s, 1H), 9.12 (d, J = 1.8 Hz, 1H), 8.49 (dd, J = 4.7, 1.5 Hz, 1H), 8.26 (d, J = 8.1 Hz, 1H), 7.77 (d, J = 8.3 Hz, 1H), 7.42 (dd, J = 8.0, 4.8 Hz, 1H), 7.26 (d, J = 8.3 Hz, 1H), 5.43 (s, 2H), 4.65 (dt, J = 47.6 Hz, J = 4.8 Hz, 2H), 3.80 (s, 2H), 3.03- 2.94 (m, 3H), 2.89 (t, J = 4.8 Hz, 3H).165415416δ 10.31 (s, 1H), 7.96 (dd, J = 8.8, 5.6 Hz, 2H), 7.67 (d, J = 8.3 Hz, 1H), 7.23 (t, J = 8.8 Hz, 3H), 5.31 (s, 2H), 4.65 (dt, J = 47.6 Hz, J = 4.8 Hz, 2H), 3.79 (s, 2H), 3.01- 2.93 (m, 3H), 2.89 (t, J = 4.8 Hz, 3H).166412413CD3OD δ 7.79 (dd, J = 8.7, 5.5 Hz, 2H), 7.39 (d, J = 7.9 Hz, 1H), 7.18 (d, J = 8.2 Hz, 1H), 7.03 (t, J = 8.8 Hz, 2H), 3.59 (dd, J = 10.8, 7.9 Hz, 2H), 3.42 (dd, J = 10.9, 2.5 Hz, 2H), 2.90 (t, J = 6.3 Hz, 4H), 2.76-2.67(m, 4H), 2.62-2.51 (m, 8H).167359360δ 8.39 (s, 1H), 8.35 (d, J = 2.4 Hz, 1H), 7.68 (d, J = 1.0 Hz, 1H), 7.49 (d, J = 8.3 Hz, 1H), 7.27 (d, J = 8.4 Hz, 1H), 6.51-6.44 (m, 1H), 5.03 (s, 2H), 4.52 (dt, J = 47.6 Hz, J = 4.8 Hz, 2H), 3.62 (dd, J = 10.7, 8.1 Hz, 2H), 3.33-3.31 (m, 2H), 2.81 (s, 2H), 2.73 (t, J = 4.9 Hz, 1H), 2.67- 2.60 (m, 3H), 2.51-2.50 (m, 2H).168462463δ 8.56 (s, 1H), 8.39 (s, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.72 (s, 1H), 7.58 (d, J = 8.2 Hz, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.78 (s, 4H), 3.54 (s, 2H), 2.60 (s, 4H), 2.49- 2.37 (m, 4H), 2.33 (s, 3H).169438439δ 8.29 (s, 1H), 8.01-7.92 (m, 2H), 7.52 (t, J = 7.4 Hz, 1H), 7.21 (dd, J = 12.3, 5.5 Hz, 2H), 7.16 (d, J = 8.2 Hz, 1H), 5.08 (s, 2H), 3.62 (dd, J = 10.6, 8.0 Hz, 2H), 3.29 (dd, J = 10.9, 2.9 Hz, 2H), 2.78 (s, 2H), 2.70 (d, J = 11.5 Hz, 2H), 2.65-2.56 (m, 2H), 2.45 (d, J = 2.5 Hz, 2H), 2.11 (s, 3H), 1.93-1.83 (m, 3H), 1.76 (d, J = 10.8 Hz, 2H), 1.41 1.34 (m, 2H).170376377δ 8.58 (s, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.54 (d, J = 8.2 Hz, 1H), 7.28 (d, J = 8.0 Hz, 2H), 7.20 (dt, J = 16.5, 8.6 Hz, 5H), 5.19 (s, 2H), 4.36 (t, J = 8.4 Hz, 2H), 3.98-3.86 (m, 2H), 3.80-3.77 (m, 1H), 2.30 (s, 3H).171352353δ 9.14 (d, J = 1.7 Hz, 1H), 8.48 (dd, J = 4.7, 1.6 Hz, 1H), 8.31-8.24 (m, 1H), 8.22 (s, 1H), 7.63 (d, J = 8.2 Hz, 1H), 7.42 (dd, J = 7.4, 4.7 Hz, 1H), 7.18 (d, J = 8.2 Hz, 1H), 5.23 (s, 2H), 3.52-3.39 (m, 4H), 3.13 (d, J = 10.0 Hz, 1H), 2.69 (s, 1H), 2.61 (d, J = 11.8 Hz, 2H), 2.53 (s, 1H), 1.85 (s, 1H), 1.65-1.62 (m, 1H), 1.60-1.50 (m, 1H), 1.48- 1.50 (m, 3H).172411412δ 8.26 (d, J = 6.8 Hz, 1H), 8.02- 7.93 (m, 2H), 7.54 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.16 (dd, J = 8.2, 3.9 Hz, 1H), 5.09 (s, 2H), 4.61 (dd, J = 12.3, 5.7 Hz, 2H), 4.31 (t, J = 6.3 Hz, 2H), 3.88 (p, J = 6.8 Hz, 1H), 3.51 (dd, J = 10.5, 7.5 Hz, 2H), 3.41-3.34 (m, 2H), 3.01- 2.88 (m, 1H), 2.55 (dd, J = 8.6, 5.4 Hz, 2H), 2.04-1.98 (m, 2H), 1.17- 1.10 (m, 2H).173361362δ 8.61 (s, 1H), 8.55 (s, 1H), 8.50 (d, J = 5.0 Hz, 1H), 7.88 (d, J = 8.9 Hz, 2H), 7.51 (d, J = 8.2 Hz, 1H), 7.44 (d, J = 4.9 Hz, 1H), 7.15 (d, J = 8.2 Hz, 1H), 6.96 (d, J = 8.9 Hz, 2H), 5.07 (s, 2H), 4.83 (s, 4H), 3.78 (s, 3H).174359360δ 8.36 (s, 1H), 7.67 (d, J = 0.8 Hz, 1H), 7.34 (d, J = 8.1 Hz, 1H), 7.13 (d, J = 8.2 Hz, 1H), 6.73 (d, J = 3.2 Hz, 1H), 6.55 (dd, J = 3.3, 1.8 Hz, 1H), 5.09 (s, 2H), 4.53 (dt, J = 48.0, J = 4.9 Hz, 2H), 3.59 (dd, J = 10.6, 8.0 Hz, 2H), 3.30 (d, J = 2.5 Hz, 2H), 2.80 (s, 2H), 2.73 (t, J = 4.9 Hz, 1H), 2.69-2.57 (m, 3H), 2.48 (s, 2H).175421422δ 8.36 (s, 1H), 7.67 (d, J = 0.9 Hz, 1H), 7.44 (t, J = 7.5 Hz, 1H), 7.37- 7.27 (m, 2H), 7.18 (t, J = 7.4 Hz, 2H), 7.13 (d, J = 8.2 Hz, 1H), 6.73 (d, J = 3.2 Hz, 1H), 6.55 (dd, J = 3.3, 1.8 Hz, 1H), 5.09 (s, 2H), 3.64 (s, 2H), 3.57 (dd, J = 14.0, 5.9 Hz, 2H), 3.31 (d, J = 13.8 Hz, 2H), 2.80 (s, 2H), 2.65-2.57 (m, 2H), 2.43 (d, J = 6.9 Hz, 2H).176449450δ 8.30 (s, 1H), 7.97 (dd, J = 8.9, 5.6 Hz, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.44 (t, J = 6.7 Hz, 1H), 7.33-7.26 (m, 1H), 7.24-7.14 (m, 5H), 5.08 (s, 2H), 3.72-3.53 (m, 4H), 3.30 (s, 2H), 2.81 (s, 2H), 2.76-2.61 (m, 2H), 2.46-2.44 (m, 2H).177421422δ 8.43-8.30 (m, 2H), 7.68 (d, J = 1.0 Hz, 1H), 7.51-7.38 (m, 2H), 7.35-7.24 (m, 2H), 7.17 (dd, J = 15.9, 8.0 Hz, 2H), 6.50-6.43 (m, 1H), 5.03 (s, 2H), 3.72-3.54 (m, 4H), 3.30 (s, 2H), 2.82 (s, 2H), 2.65- 2.56 (m, 2H), 2.46 (d, J = 9.1 Hz, 2H).178460461δ 8.50 (s, 1H), 7.99 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.24 (m, 6H), 5.18 (s, 2H), 4.82 (s, 4H), 3.45 (s, 2H), 2.36 (m, 7H), 2.20 (s, 4H).179438439CDCl3δ 7.84 (dd, J = 8.4, 5.6 Hz, 2H), 7.35 (d, J = 8.2 Hz, 1H), 7.14-7.00 (m, 3H), 6.73 (s, 1H), 4.59 (s, 2H), 3.76-3.64 (m, 2H), 3.48 (d, J = 8.6 Hz, 2H), 2.81-2.35 (m, 10H), 2.32 (s, 3H), 2.26-2.13 (m, 3H), 1.47 (d, J = 6.6 Hz, 2H).181425426CDCl3δ 7.84 (dd, J = 8.7, 5.5 Hz, 2H), 7.35 (d, J = 8.1 Hz, 1H), 7.11 (dd, J = 17.2, 8.4 Hz, 3H), 6.75 (s, 1H), 4.59 (s, 2H), 3.90-3.65 (m, 6H), 3.48 (d, J = 7.4 Hz, 2H), 2.75 (s, 2H), 2.69-2.60 (m, 1H), 2.49 (s, 4H), 2.28-2.10 (m, 2H), 1.46 (dd, J = 17.9, 12.2 Hz, 2H).182369370δ 8.35 (s, 1H), 7.66 (d, J = 0.9 Hz, 1H), 7.34 (d, J = 8.1 Hz, 1H), 7.13 (d, J = 8.2 Hz, 1H), 6.73 (d, J = 3.3 Hz, 1H), 6.55 (dd, J = 3.3, 1.8 Hz, 1H), 5.10 (s, 2H), 4.55 (q, J = 6.6 Hz, 2H), 4.46 (t, J = 5.9 Hz, 2H), 3.68-3.50 (m, 3H), 3.25 (d, J = 49.1 Hz, 2H), 2.83 (s, 2H), 2.53 (d, J = 9.5 Hz, 2H), 2.44-2.36 (m, 2H).183387388δ 8.32 (d, J = 14.9 Hz, 1H), 8.01- 7.93 (m, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.27-7.18 (m, 2H), 7.16 (d, J = 8.2 Hz, 1H), 5.08 (s, 2H), 4.53 (dt, J = 48.0, J = 4.9 Hz, 2H), 3.61 (dd, J = 10.7, 8.1 Hz, 2H), 3.34-3.31 (m, 2H), 2.81 (s, 2H), 2.73 (t, J = 5.0 Hz, 1H), 2.70-2.57 (m, 3H), 2.49 (s, 2H).184327328CD3OD δ 8.30 (d, J = 2.3 Hz, 1H), 7.58 (s, 1H), 7.45 (d, J = 8.4 Hz, 1H), 7.28 (d, J = 8.4 Hz, 1H), 6.37 (t, J = 2.4 Hz, 1H), 3.66-3.46 (m, 4H), 3.34 (s, 2H), 3.10 (s, 2H), 3.06-2.92 (m, 2H), 2.71 (s, 3H).185339340δ 8.67 (s, 1H), 8.49 (s, 1H), 8.35 (s, 1H), 7.82 (d, J = 9.2 Hz, 1H), 7.69 (s, 1H), 7.54 (d, J = 8.4 Hz, 1H), 7.29 (d, J = 8.4 Hz, 1H), 6.48 (s, 1H), 5.15 (s, 2H), 4.79 (d, J = 17.4 Hz, 4H).186367368δ 8.60 (s, 1H), 8.49 (s, 1H), 7.98 (dd, J = 8.6, 5.7 Hz, 2H), 7.81 (d, J = 7.1 Hz, 1H), 7.58 (d, J = 8.2 Hz, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.20 (s, 2H), 4.79 (d, J = 16.5 Hz, 4H).187379380δ 8.58 (s, 1H), 8.49 (s, 1H), 7.88 (d, J = 8.8 Hz, 2H), 7.81 (d, J = 6.8 Hz, 1H), 7.51 (d, J = 8.2 Hz, 1H), 7.15 (d, J = 8.2 Hz, 1H), 6.96 (d, J = 8.8 Hz, 2H), 5.08 (s, 2H), 4.79 (d, J = 17.6 Hz, 4H), 3.78 (s, 3H).188413414δ 8.21 (s, 1H), 8.03-7.90 (m, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.20 (dt, J = 18.7, 8.9 Hz, 3H), 5.25-5.00 (m, 1H), 5.09 (s, 2H), 3.66-3.45 (m, 4H), 3.34 (d, J = 3.9 Hz, 2H), 3.07- 2.94 (m, 2H), 2.79 (p, J = 6.7 Hz, 1H), 2.65 (s, 2H), 1.99-1.80 (m, 2H), 1.27-1.20 (m, 2H).189385386δ 8.25 (s, 1H), 7.70-7.63 (m, 1H), 7.34 (d, J = 8.1 Hz, 1H), 7.13 (d, J = 8.2 Hz, 1H), 6.73 (d, J = 3.3 Hz, 1H), 6.55 (dd, J = 3.3, 1.8 Hz, 1H), 5.20-5.03 (m, 3H), 3.61-3.47 (m, 4H), 3.33 (s, 1H), 3.31 (d, J = 3.7 Hz, 1H), 3.07-3.02 (m, 1H), 3.01- 2.96 (m, 1H), 2.79 (p, J = 6.8 Hz, 1H), 2.64 (s, 2H), 2.01-1.76 (m, 2H), 1.26-1.20 (m, 2H).190385386δ 8.32 (d, J = 17.9 Hz, 2H), 7.68 (s, 1H), 7.48 (d, J = 8.2 Hz, 1H), 7.27 (d, J = 8.4 Hz, 1H), 6.47 (s, 1H), 5.11 (m, 4H), 3.35 (s, 1H), 3.28- 3.17 (m, 1H), 3.02 (d, J = 24.8 Hz, 2H), 2.87-2.73 (m, 1H), 2.65 (s, 2H), 1.99-1.82 (m, 2H), 1.24 (m, 2H).191393394δ 8.26 (s, 1H), 7.94 (d, J = 7.3 Hz, 2H), 7.55 (d, J = 8.2 Hz, 1H), 7.40 (t, J = 7.6 Hz, 2H), 7.28 (t, J = 7.3 Hz, 1H), 7.17 (d, J = 8.2 Hz, 1H), 5.09 (s, 2H), 4.64-4.56 (m, 2H), 4.31 (t, J = 6.3 Hz, 2H), 3.94-3.83 (m, 1H), 3.57-3.46 (m, 2H), 3.42- 3.34 (m, 2H), 3.00-2.84 (m, 1H), 2.55 (s, 2H), 2.08-1.94 (m, 2H), 1.18-1.11 (m, 2H).192363364δ 10.15 (s, 1H), 8.07 (d, J = 4.0 Hz, 1H), 7.99-7.90 (m, 2H), 7.62 (d, J = 8.3 Hz, 1H), 7.56-7.46 (m, 1H), 7.28-7.16 (m, 3H), 6.64 (dd, J = 6.5, 5.2 Hz, 1H), 6.42 (d, J = 8.1 Hz, 1H), 5.20 (s, 2H), 4.05 (t, J = 6.8 Hz, 4H), 3.85 (s, 1H).193335336δ 10.22 (s, 1H), 8.07 (d, J = 3.9 Hz, 1H), 7.68 (d, J = 1.0 Hz, 1H), 7.55- 7.48 (m, 1H), 7.42 (d, J = 8.2 Hz, 1H), 7.19 (d, J = 8.3 Hz, 1H), 6.75 (d, J = 3.2 Hz, 1H), 6.64 (dd, J = 6.6, 5.3 Hz, 1H), 6.56 (dd, J = 3.3, 1.8 Hz, 1H), 6.41 (d, J = 8.3 Hz, 1H), 5.21 (s, 2H), 4.10-4.03 (m, 4H), 3.84 (s, 1H).194348349δ 9.85 (s, 1H), 7.92 (d, J = 7.2 Hz, 2H), 7.64 (d, J = 8.4 Hz, 1H), 7.55 (s, 1H), 7.40 (t, J = 7.6 Hz, 2H), 7.34-7.17 (m, 2H), 6.67 (s, 1H), 5.19 (s, 2H), 4.09 (t, J = 5.4 Hz, 2H), 3.83 (s, 2H), 3.48 (s, 2H), 3.10- 2.95 (m, 2H).195345346δ 10.15 (s, 1H), 8.07 (d, J = 3.9 Hz, 1H), 7.93 (d, J = 7.3 Hz, 2H), 7.64 (d, J = 8.3 Hz, 1H), 7.55-7.47 (m, 1H), 7.41 (t, J = 7.6 Hz, 2H), 7.30 (t, J = 7.3 Hz, 1H), 7.22 (d, J = 8.3 Hz, 1H), 6.68-6.57 (m, 1H), 6.42 (d, J = 8.3 Hz, 1H), 5.19 (s, 2H), 4.11- 4.04 (m, 4H), 3.86 (s, 1H).196353354CD3OD δ 7.94-7.86 (m, 2H), 7.62 (d, J = 8.3 Hz, 1H), 7.43 (t, J = 7.6 Hz, 2H), 7.39-7.31 (m, 2H), 3.70 (d, J = 18.4 Hz, 4H), 3.35 (d, J = 3.7 Hz, 2H), 2.71 (d, J = 21.9 Hz, 4H), 2.14 (s, 3H).197334335δ 8.88 (s, 1H), 7.93 (d, J = 7.7 Hz, 2H), 7.58 (d, J = 8.2 Hz, 1H), 7.39 (t, J = 7.6 Hz, 2H), 7.28 (t, J = 7.3 Hz, 1H), 7.19-7.10 (m, 2H), 6.91 (s, 1H), 5.09 (s, 2H), 4.71 (s, 2H), 4.07 (t, J = 5.0 Hz, 2H), 3.92 (t, J = 5.2 Hz, 2H).198348349δ 10.12 (s, 1H), 7.68 (d, J = 1.0 Hz, 1H), 7.41 (d, J = 8.0 Hz, 1H), 7.39- 6.96 (m, 6H), 6.74 (d, J = 3.2 Hz, 1H), 6.61-6.51 (m, 1H), 5.17 (s, 2H), 3.53 (d, J = 10.6 Hz, 3H), 3.38 (d, J = 17.4 Hz, 2H), 3.22 (s, 2H).199379380δ 8.32 (s, 1H), 7.96-7.90 (m, 2H), 7.56 (d, J = 8.2 Hz, 1H), 7.40 (t, J = 7.6 Hz, 2H), 7.28 (dd, J = 9.1, 5.5 Hz, 1H), 7.17 (d, J = 8.2 Hz, 1H), 5.09 (s, 2H), 4.56 (t, J = 6.5 Hz, 2H), 4.46 (t, J = 6.0 Hz, 2H), 3.65 (dd, J = 10.7, 8.1 Hz, 2H), 3.60- 3.51 (m, 1H), 3.32 (d, J = 3.3 Hz, 2H), 2.84 (s, 2H), 2.56-2.51 (m, 2H), 2.42 (dd, J = 9.0, 2.6 Hz, 2H).201355356δ 8.36 (s, 1H), 7.97 (dd, J = 8.7, 5.7 Hz, 2H), 7.53 (t, J = 8.6 Hz, 1H), 7.23 (q, J = 9.1 Hz, 2H), 7.16 (d, J = 8.2 Hz, 1H), 5.11 (s, 2H), 3.59 (dd, J = 10.6, 7.5 Hz, 2H), 3.40 (d, J = 10.7 Hz, 2H), 2.91 (s, 2H), 2.83 (s, 2H), 2.67 (s, 2H), 2.38 (d, J = 37.1 Hz, 3H).203397398CDCl3δ 7.83 (dd, J = 8.7, 5.5 Hz, 2H), 7.36 (d, J = 8.1 Hz, 1H), 7.17-7.06 (m, 3H), 6.77 (s, 1H), 4.71 (t, J = 6.6 Hz, 2H), 4.64 (t, J = 6.1 Hz, 2H), 4.59 (s, 2H), 3.85-3.76 (m, 2H), 3.67 (dd, J = 12.5, 6.2 Hz, 1H), 3.51-3.42 (m, 2H), 2.99 (s, 2H), 2.69-2.61 (m, 2H), 2.52 (d, J = 6.6 Hz, 2H).204300301δ 9.95 (s, 1H), 7.93 (dd, J = 8.4, 5.8 Hz, 2H), 7.58 (d, J = 8.3 Hz, 1H), 7.27-7.11 (m, 3H), 5.12 (s, 2H), 3.44-3.37(m, 3H), 3.12 (s, 2H), 2.16 (s, 3H).205348349δ 10.10 (s, 1H), 8.32 (s, 1H), 7.70 (s, 1H), 7.57 (d, J = 8.4 Hz, 1H), 7.40-7.20 (m, 6H), 6.48 (s, 1H), 5.11 (s, 2H), 3.63-3.39 (m, 5H), 3.26 (s, 2H).206348349δ 9.51 (s, 1H), 7.94 (d, J = 7.6 Hz, 2H), 7.82 (s, 1H), 7.65 (d, J = 8.3 Hz, 1H), 7.41 (t, J = 7.7 Hz, 2H), 7.29 (d, J = 7.2 Hz, 1H), 7.25 (d, J = 8.4 Hz, 1H), 5.33 (s, 2H), 4.08 (s, 2H), 3.63 (s, 2H), 2.83 (s, 2H), 2.43 (s, 3H).208334335δ 9.47 (s, 1H), 7.91 (d, J = 7.4 Hz, 2H), 7.77 (s, 1H), 7.63 (d, J = 8.3 Hz, 1H), 7.39 (t, J = 7.6 Hz, 2H), 7.29 (d, J = 7.2 Hz, 1H), 7.25 (d, J = 8.4 Hz, 1H), 5.33 (s, 2H), 3.96 (t, J = 5.3 Hz, 2H), 3.90 (s, 2H), 3.05 (s, 2H).210366367δ 9.12 (s, 1H), 8.45 (d, J = 4.5 Hz, 1H), 8.24 (s, 2H), 7.62 (d, J = 8.1 Hz, 1H), 7.39 (dd, J = 7.9, 4.8 Hz, 1H), 7.16 (d, J = 8.2 Hz, 1H), 5.21 (s, 2H), 3.46 (s, 2H), 3.27 (s, 2H), 2.53 (d, J = 24.3 Hz, 2H), 2.45- 2.33 (m, 2H), 2.29 (s, 3H), 1.74 (s, 2H), 1.58 (s, 4H).211366367δ 9.12 (d, J = 2.0 Hz, 1H), 8.45 (dd, J = 4.7, 1.5 Hz, 1H), 8.29-8.19 (m, 2H), 7.61 (d, J = 8.2 Hz, 1H), 7.39 (dd, J = 8.0, 4.7 Hz, 1H), 7.16 (d, J = 8.2 Hz, 1H), 5.21 (s, 2H), 3.52- 3.33 (m, 4H), 3.15 (d, J = 9.9 Hz, 1H), 2.31 (s, 2H), 2.15 (s, 3H), 2.07- 1.96 (m, 1H), 1.84 (d, J = 37.5 Hz, 1H), 1.68 (s, 1H), 1.61-1.38 (m, 3H), 1.31 (s, 1H).212334335δ 8.80 (s, 1H), 7.90 (d, J = 7.5 Hz, 2H), 7.59 (s, 1H), 7.55 (d, J = 8.2 Hz, 1H), 7.37 (t, J = 7.6 Hz, 2H), 7.25 (t, J = 7.2 Hz, 1H), 7.14 (d, J = 8.2 Hz, 1H), 6.74 (s, 1H), 5.05 (s, 2H), 4.71 (s, 2H), 4.10 (t, J = 5.2 Hz, 2H), 3.84 (t, J = 5.2 Hz, 2H).214333334δ 9.15 (s, 1H), 8.71 (s, 1H), 8.56 (s, 2H), 8.48 (d, J = 3.8 Hz, 1H), 8.28 (d, J = 8.0 Hz, 1H), 7.69 (d, J = 8.2 Hz, 1H), 7.43 (dd, J = 7.9, 4.7 Hz, 1H), 7.20 (d, J = 8.2 Hz, 1H), 5.33 (s, 2H), 4.85 (s, 4H).215446447CDCl3δ 9.09 (s, 1H), 8.55 (d, J = 4.3 Hz, 1H), 8.15 (d, J = 8.2 Hz, 1H), 7.46- 7.37 (m, 2H), 7.34-7.31 (m, 2H), 7.16 (d, J = 7.8 Hz, 1H), 6.75 (s, 1H), 4.70 (s, 2H), 3.88 (s, 3H), 3.59 (t, J = 6.8 Hz, 2H), 3.46 (s, 2H), 3.36 (s, 2H), 2.56 (s, 2H), 2.34 (s, 2H), 1.86 (s, 2H), 1.64 (d, J = 24.5 Hz, 4H).216338339δ 9.11 (d, J = 1.6 Hz, 1H), 8.62 (s, 1H), 8.45 (dd, J = 4.7, 1.6 Hz, 1H), 8.28-8.20 (m, 1H), 7.61 (d, J = 8.2 Hz, 1H), 7.39 (dd, J = 7.4, 4.7 Hz, 1H), 7.16 (d, J = 8.2 Hz, 1H), 5.07 (s, 2H), 4.21 (d, J = 11.8 Hz, 1H), 4.07 (d, J = 11.6 Hz, 1H), 2.93 (dt, J = 30.3, 9.8 Hz, 3H), 2.58-2.50 (m, 1H), 2.10-1.98 (m, 2H), 1.84 (s, 1H), 1.75 (d, J = 5.6 Hz, 1H), 1.67 (d, J = 8.1 Hz, 2H), 1.35-1.21 (m, 1H).217338339CD3OD δ 9.08 (s, 1H), 8.44 (d, J = 3.5 Hz, 1H), 8.32 (d, J = 7.8 Hz, 1H), 7.59 (d, J = 8.1 Hz, 1H), 7.46 (dd, J = 7.8, 4.9 Hz, 1H), 7.28 (d, J = 8.1 Hz, 1H), 3.65 (d, J = 7.2 Hz, 4H), 3.59-3.49 (m, 4H), 1.94-1.85 (m, 3H), 1.81 (d, J = 5.0 Hz, 1H).218350351δ 9.14 (d, J = 1.7 Hz, 1H), 8.63 (s, 1H), 8.48 (dd, J = 4.7, 1.5 Hz, 2H), 8.31-8.25 (m, 1H), 7.81 (dd, J = 9.0, 2.4 Hz, 1H), 7.68 (d, J = 8.2 Hz, 1H), 7.42 (dd, J = 8.0, 4.8 Hz, 1H), 7.19 (d, J = 8.2 Hz, 1H), 5.32 (s, 2H), 4.80 (d, J = 17.9 Hz, 4H).219335336δ 9.56 (s, 1H), 9.13 (d, J = 1.7 Hz, 1H), 8.50 (dd, J = 4.7, 1.6 Hz, 1H), 8.31-8.23 (m, 1H), 7.79 (d, J = 6.8 Hz, 1H), 7.73 (d, J = 8.3 Hz, 1H), 7.43 (dd, J = 7.7, 4.5 Hz, 1H), 7.29 (d, J = 8.3 Hz, 1H), 5.43 (s, 2H), 3.99 (t, J = 5.4 Hz, 2H), 3.92 (d, J = 11.7 Hz, 2H), 3.09 (t, J = 5.4 Hz, 2H).220338339δ 9.13 (d, J = 1.9 Hz, 1H), 8.51 (s, 1H), 8.48 (dd, J = 4.7, 1.5 Hz, 1H), 8.29-8.22 (m, 1H), 7.63 (d, J = 8.2 Hz, 1H), 7.42 (dd, J = 7.9, 4.8 Hz, 1H), 7.18 (d, J = 8.2 Hz, 1H), 5.28 (s, 2H), 3.76 (s, 4H), 3.69 (s, 4H), 2.67 (s, 1H), 1.64 (s, 4H).221283284δ 9.14 (d, J = 1.6 Hz, 1H), 8.48 (dd, J = 4.7, 1.6 Hz, 1H), 8.30-8.22 (m, 2H), 7.64 (d, J = 8.2 Hz, 1H), 7.42 (dd, J = 7.6, 4.4 Hz, 1H), 7.19 (d, J = 8.2 Hz, 1H), 5.23 (s, 2H), 3.40 (s, 4H), 1.87 (s, 4H).222489490δ 8.20 (s, 1H), 7.97 (dd, J = 8.9, 5.6 Hz, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.24-7.19 (m, 3H), 7.16 (d, J = 8.2 Hz, 1H), 6.89 (d, J = 8.0 Hz, 2H), 6.79 (dd, J = 7.4, 1.9 Hz, 1H), 5.10 (s, 2H), 3.46 (q, J = 13.7 Hz, 4H), 3.21 (d, J = 10.9 Hz, 1H), 2.49- 2.37 (m, 2H), 2.27 (s, 2H), 2.15 (s, 1H), 1.87 (s, 1H), 1.69 (s, 1H), 1.57- 1.46 (m, 3H), 1.38 (s, 1H).223463464δ 8.20 (s, 1H), 7.97 (dd, J = 8.5, 5.7 Hz, 2H), 7.56-7.48 (m, 2H), 7.29 (s, 1H), 7.22 (t, J = 8.8 Hz, 2H), 7.16 (d, J = 8.2 Hz, 1H), 5.11 (s, 2H), 3.77 (s, 3H), 3.52-3.35 (m, 4H), 3.18 (s, 1H), 2.47-2.38 (m, 1H), 2.24 (d, J = 31.9 Hz, 2H), 2.09 (s, 1H), 1.87 (d, J = 31.9 Hz, 1H), 1.63 (d, J = 31.9 Hz, 1H), 1.59- 1.40 (m, 3H), 1.33 (s, 1H), 1.23 (s, 1H).224355356δ 8.60 (s, 1H), 7.97 (dd, J = 8.9, 5.6 Hz, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.16 (d, J = 8.2 Hz, 1H), 4.98 (s, 2H), 4.24 (d, J = 11.5 Hz, 1H), 4.09 (d, J = 12.8 Hz, 1H), 3.05-2.84 (m, 3H), 2.57 (d, J = 10.7 Hz, 1H), 2.05 (dd, J = 17.8, 9.0 Hz, 2H), 1.86 (s, 1H), 1.78 (d, J = 5.4 Hz, 1H), 1.69 (d, J = 8.2 Hz, 2H), 1.39-1.24 (m, 1H).225159460δ 8.69 (s, 1H), 7.96 (dd, J = 8.8, 5.7 Hz, 2H), 7.54 (d, J = 8.2 Hz, 1H), 7.27 (dd, J = 15.7, 8.4 Hz, 3H), 7.23- 7.11 (m, 5H), 5.02 (s, 2H), 3.71 (d, J = 16.0 Hz, 2H), 3.52 (t, J = 15.3 Hz, 6H), 2.41-2.28 (m, 2H), 1.51-1.33 (m, 1H), 1.23 (dd, J = 12.1, 5.8 Hz, 1H).226328329δ 10.33 (s, 1H), 7.96 (dd, J = 8.4, 5.7 Hz, 2H), 7.82 (s, 1H), 7.67 (d, J = 8.3 Hz, 1H), 7.23 (t, J = 9.3 Hz, 3H), 5.32 (s, 2H), 2.55 (d, J = 13.6 Hz, 3H).228398399δ 8.66 (s, 1H), 8.55 (s, 2H), 8.00 (d, J = 8.8 Hz, 2H), 7.60 (d, J = 8.2 Hz, 1H), 7.27 (t, J = 74.0 Hz, 1H), 7.21 (d, J = 8.8 Hz, 2H), 7.17 (d, J = 8.4 Hz, 1H), 5.22 (s, 2H), 4.85 (s, 4H).229356357CD3OD δ 7.80 (dd, J = 8.8, 5.6 Hz, 2H), 7.39 (d, J = 8.2 Hz, 1H), 7.17 (d, J = 8.2 Hz, 1H), 7.02 (t, J = 8.8 Hz, 2H), 3.76 (s, 4H), 3.62-3.48 (m, 4H), 1.79-1.65 (m, 4H).230404405CD3OD δ 7.82 (d, J = 8.8 Hz, 2H), 7.41 (d, J = 8.2 Hz, 1H), 7.17 (d, J = 8.2 Hz, 1H), 7.07 (d, J = 8.7 Hz, 2H), 6.75 (t, J = 74.4 Hz, 1H), 3.75 (s, 4H), 3.59-3.49 (m, 4H), 1.79-1.64 (m, 4H).231286287δ 8.14 (s, 1H), 8.00 (s, 1H), 7.78 (s, 1H), 7.22 (d, J = 8.1 Hz, 1H), 7.09 (d, J = 8.1 Hz, 1H), 4.88 (s, 2H), 3.92-3.77 (s, 3H), 3.38-3.35 (s, 4H), 1.86 (s, 4H).232348349δ 8.21 (s, 1H), 8.02-7.95 (m, 2H), 7.55 (d, J = 8.2 Hz, 1H), 7.26 (t, J = 74.0 Hz, 1H), 7.20 (d, J = 8.8 Hz, 2H), 7.16 (d, J = 8.4 Hz, 1H), 5.12 (s, 2H), 3.40 (s, 4H), 1.87 (s, 4H).235439440CD3CN δ 7.97-7.93 (m, 2H), 7.46-7.44 (m, 1H), 7.18-7.12 (m, 4H), 4.64 (br, 2H), 3.87-3.83 (m, 2H), 3.69-3.64 (m, 2H), 3.40-3.28 (m, 4H), 2.84 (s, 2H), 2.53-2.46 (m, 4H), 2.26-2.24 (m, 2H), 1.68-1.63 (m, 3H), 1.18- 1.13 (m, 2H)236425426CD3CN δ 7.98-7.94 (m, 2H), 7.47-7.45 (m, 1H), 7.18-7.13 (m, 4H), 4.64 (br, 2H), 3.88-3.85 (m, 2H), 3.68-3.63 (m, 2H), 3.37-3.30 (m, 4H), 2.85 (s, 2H), 2.63-2.57 (m, 4H), 2.22-2.16 (m, 1H), 1.95-1.93 (m, 2H), 1.48- 1.38 (m, 2H)237439440CD3OD δ 7.83 (s, 2H), 7.36-7.34 (d, J = 8.0 Hz, 1H), 7.10-7.07 (m, 3H), 6.76 (s, 1H), 3.97-3.96 (t, J = 0.4 Hz, 1H), 3.78-3.70 (m, 5H), 3.51- 3.45 (m, 3H), 3.04-3.02 (d, J = 7.6 Hz, 1H), 2.72 (s, 2H), 2.24 (s, 3H), 2.13-2.09 (m, 2H), 2.01-1.91 (m, 2H), 1.54-1.47 (m, 2H), 1.26 (s, 2H).238342343CD3OD δ 7.91 (dd, J = 8.3, 5.6 Hz, 2H), 7.50 (d, J = 8.2 Hz, 1H), 7.29 (d, J = 8.2 Hz, 1H), 7.14 (t, J = 8.7 Hz, 2H), 3.77 (d, J = 10.3 Hz, 2H), 3.58 (d, J = 9.7 Hz, 2H), 3.50 (d, J = 6.8 Hz, 2H), 1.63 (s, 2H), 1.04-0.91 (m, 1H).239368369δ 10.21 (s, 1H), 8.01-7.90 (m, 2H), 7.59 (d, J = 8.2 Hz, 1H), 7.23 (dd, J = 16.9, 8.4 Hz, 3H), 5.09 (s, 2H), 4.55 (t, J = 6.5 Hz, 2H), 4.41 (t, J = 5.7 Hz, 2H), 3.75-3.63 (m, 1H), 3.05 (d, J = 8.9 Hz, 2H), 2.39 (d, J = 8.7 Hz, 3H), 1.93 (s, 2H).240433434δ 8.54 (d, J = 10.9 Hz, 2H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.34 (s, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.80 (s, 4H), 3.96 (d, J = 10.9 Hz, 2H), 3.51-3.40 (m, 2H), 2.97 (t, J = 11.5 Hz, 1H), 1.78 (dd, J = 10.6, 7.4 Hz, 4H).241381382δ 10.18 (s, 1H), 8.00-7.92 (m, 2H), 7.59 (d, J = 8.3 Hz, 1H), 7.22 (dd, J = 17.2, 8.5 Hz, 3H), 5.08 (s, 2H), 3.29 (t, J = 6.7 Hz, 2H), 3.17- 3.08 (m, 1H), 2.96 (d, J = 8.9 Hz, 2H), 2.78 (t, J = 6.4 Hz, 2H), 2.37 (d, J = 8.5 Hz, 2H), 2.31 (s, 1H), 2.18 (s, 3H), 1.89 (s, 2H).242355356δ 8.21 (s, 1H), 7.96 (dd, J = 8.8, 5.6 Hz, 2H), 7.53 (d, J = 8.1 Hz, 1H), 7.22 (t, J = 8.8 Hz, 2H), 7.15 (d, J = 8.2 Hz, 1H), 5.06 (s, 2H), 3.59 (d, J = 10.5 Hz, 2H), 3.41 (d, J = 9.7 Hz, 2H), 2.24 (s, 6H), 1.62 (s, 2H), 1.38 (s, 1H).243393394δ 8.55 (d, J = 8.1 Hz, 2H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.44 (s, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.19 (s, 2H), 4.83 (s, 4H), 4.53 (s, 2H), 3.38 (s, 3H).244421422δ 8.46 (s, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.31 (s, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.16 (s, 1H), 5.16 (s, 2H), 4.64 (s, 2H), 4.51 (s, 2H), 4.24 (d, J = 7.4 Hz, 2H), 3.31-3.22 (m, 2H), 3.03 (s, 2H), 2.87-2.74 (m, 1H), 2.25 (s, 3H).245421422δ 8.45 (s, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.61 (s, 1H), 7.56 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.15 (s, 2H), 4.51 (s, 4H), 4.29 (d, J = 7.3 Hz, 2H), 3.24 (t, J = 7.2 Hz, 2H), 2.98 (t, J = 6.1 Hz, 2H), 2.80- 2.73 (m, 1H), 2.22 (s, 3H).246407408δ 8.51 (s, 1H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.37 (s, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.15 (s, 2H), 4.91-4.84 (m, 1H), 4.81- 4.67 (m, 2H), 4.51 (s, 2H), 3.69 (t, J = 7.4 Hz, 2H), 3.32-3.23 (m, 2H), 2.32 (s, 3H).247407408δ 8.47 (s, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.73 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.16 (s, 2H), 4.99-4.89 (m, 1H), 4.55 (s, 4H), 3.70 (t, J = 7.4 Hz, 2H), 3.37 (t, J = 7.4 Hz, 2H), 2.32 (s, 3H).248425426δ 8.28 (s, 1H), 8.02-7.90 (m, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.38 (t, J = 7.9 Hz, 1H), 7.27-7.18 (m, 2H), 7.18-7.09 (m, 1H), 5.10 (s, 2H), 4.67-4.59 (m, 0.5H), 4.52-4.41 (m, 4H), 4.38-4.32 (m, 0.5H), 3.70- 3.63 (m, 1H), 3.52-3.47 (m, 2H), 3.38-3.35 (m, 1H), 2.67-2.52 (m, 3H), 2.05 (s, 3H), 1.95-1.84 (m, 2H), 1.27-1.19 (m, 2H).249417418δ 8.59 (s, 1H), 7.99 (dd, J = 8.9, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.25-7.16 (m, 3H), 7.12 (t, J = 7.7 Hz, 1H), 6.64 (d, J = 7.4 Hz, 1H), 6.48 (d, J = 8.2 Hz, 1H), 5.16 (s, 2H), 4.98 (s, 2H), 4.68 (s, 2H), 3.37 (t, J = 6.8 Hz, 4H), 1.92 (t, J = 6.4 Hz, 4H).250434435δ 8.53 (s, 1H), 8.38 (d, J = 1.7 Hz, 1H), 7.70 (s, 1H), 7.52 (d, J = 8.2 Hz, 1H), 7.48 (dd, J = 3.6, 1.1 Hz, 1H), 7.41 (dd, J = 5.0, 1.0 Hz, 1H), 7.12 (d, J = 8.2 Hz, 1H), 7.06 (dd, J = 5.0, 3.6 Hz, 1H), 5.17 (s, 2H), 4.74 (s, 4H), 2.87 (d, J = 11.5 Hz, 2H), 2.55 (dd, J = 10.5, 6.0 Hz, 1H), 2.12 (s, 3 H), 1.97 (td, J = 11.1, 3.2 Hz, 2H), 1.78-1.62 (m, 4H).251432433δ 8.59-8.52 (m, 2H), 7.85 (d, J = 1.6 Hz, 1H), 7.52 (d, J = 8.2 Hz, 1H), 7.48 (dd, J = 3.6, 1.1 Hz, 1H), 7.41 (dd, J = 5.1, 1.0 Hz, 1H), 7.12 (d, J = 8.2 Hz, 1H), 7.06 (dd, J = 5.0, 3.6 Hz, 1H), 6.28 (s, 1H), 5.18 (s, 2H), 4.77 (s, 4H), 3.05 (s, 2H), 2.60 (t, J = 5.4 Hz, 2H), 2.52 (s, 2H), 2.30 (s, 3H).252446447δ 8.55 (s, 1H), 8.38 (d, J = 1.7 Hz, 1H), 8.02-7.94 (m, 2H), 7.70 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.17 (d, J = 8.9 Hz, 2H), 4.75 (s, 4H), 2.89 (d, J = 11.1 Hz, 2H), 2.61-2.53 (m, 1H), 2.21 (s, 3H), 1.99 (t, J = 10.1 Hz, 2H), 1.78- 1.63 (m, 4H).253444445δ 8.57 (d, J = 2.7 Hz, 2H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.85 (s, 1H), 7.58 (d, J = 8.2 Hz, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 6.28 (s, 1H), 5.19 (s, 2H), 4.78 (s, 4H), 3.04 (s, 2H), 2.60 (t, J = 5.2 Hz, 2H), 2.52 (s, 2H), 2.30 (s, 3H).254404405δ 8.09 (d, J = 3.3 Hz, 2H), 7.94- 7.87 (m, 3H), 7.83 (s, 1H), 7.23 (t, J = 8.9 Hz, 2H), 6.37 (s, 1H), 5.25 (s, 2H), 4.71 (s, 4H), 3.92 (t, J = 7.3 Hz, 4H), 2.36-2.26 (m, 2H).255389390δ 8.53 (s, 1H), 8.40 (s, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.32 (s, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.77 (s, 4H), 2.17- 2.07 (m, 1H), 1.00-0.79 (m, 4H).256424425CDCl3δ 7.83 (dd, J = 8.8, 5.4 Hz, 2H), 7.35 (d, J = 8.1 Hz, 1H), 7.16-7.06 (m, 3H), 6.75 (s, 1H), 4.59 (s, 2H), 3.82-3.73 (m, 2H), 3.47 (s, 2H), 3.40 (d, J = 10.3 Hz, 2H), 2.90 (d, J = 17.3 Hz, 4H), 2.63 (d, J = 6.6 Hz, 5H), 2.48 (d, J = 6.4 Hz, 2H), 2.32 (s, 3H).257352353δ 8.45 (s, 1H), 7.98 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 2H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.16 (s, 2H), 4.51 (s, 4H), 3.85 (s, 3H).258352353δ 8.45 (s, 1H), 7.98 (dd, J = 8.7, 5.7 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.28 (s, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.16 (s, 2H), 4.62 (s, 2H), 4.52 (s, 2H), 3.78 (s, 3H).259366367δ 8.55 (s, 1H), 7.98 (dd, J = 8.9, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.42-7.36 (m, 1H), 7.25-7.13 (m, 5H), 5.19 (s, 2H), 4.84 (s, 4H).260379380δ 8.52 (s, 1H), 8.17 (s, 1H), 7.97 (dd, J = 8.8, 5.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.16 (d, J = 8.2 Hz, 1H), 6.84 (s, 1H), 5.18 (s, 2H), 4.75 (s, 4H), 3.86 (s, 3H).261420421δ 8.56 (s, 1H), 8.49 (s, 1H), 7.44 (s, 1H), 7.36 (dd, J = 8.5, 6.3 Hz, 1H), 7.17 (d, J = 8.1 Hz, 1H), 7.13-7.00 (m, 3H), 5.11 (s, 2H), 4.80 (s, 4H), 3.56 (s, 2H), 2.34 (s, 3H), 2.21 (s, 6H).262397398CDCl3δ 7.79 (s, 2H), 7.35 (s, 1H), 7.13 (d, J = 8.3 Hz, 3H), 4.72 (t, J = 6.5 Hz, 2H), 4.65 (t, J = 6.8 Hz, 2H), 3.92- 3.83 (m, 1H), 3.80-3.47 (m, 4H), 2.30 (s, 3H), 1.79 (s, 2H), 1.52-1.48 (m, 1H).263409410δ 10.17 (s, 1H), 7.95 (dd, J = 8.8, 5.6 Hz, 2H), 7.58 (d, J = 8.3 Hz, 1H), 7.22 (dd, J = 17.9, 8.7 Hz, 3H), 5.09 (s, 2H), 3.10 (d, J = 8.9 Hz, 2H), 2.68 (d, J = 8.5 Hz, 2H), 2.37 (d, J = 8.6 Hz, 2H), 2.28 (s, 1H), 2.12 (s, 3H), 2.00 (t, J = 7.6 Hz, 1H), 1.93-1.80 (m, 4H), 1.73 (d, J = 13.4 Hz, 2H), 1.32 (t, J = 13.5 Hz, 2H).264396397δ 10.17 (s, 1H), 7.96 (dd, J = 8.8, 5.6 Hz, 2H), 7.59 (d, J = 8.2 Hz, 1H), 7.22 (dd, J = 17.2, 8.5 Hz, 3H), 5.09 (s, 2H), 3.81 (d, J = 11.2 Hz, 2H), 3.28 (t, J = 11.2 Hz, 2H), 3.11 (d, J = 9.0 Hz, 2H), 2.39 (d, J = 8.5 Hz, 2H), 2.34-2.21 (m, 2H), 1.91 (s, 2H), 1.71 (d, J = 12.2 Hz, 2H), 1.30 (dd, J = 23.6, 13.4 Hz, 2H).265429430δ 9.15 (s, 1H), 8.59 (s, 1H), 8.48 (d, J = 3.6 Hz, 1H), 8.38 (s, 1H), 8.28 (d, J = 7.8 Hz, 1H), 7.70-7.67 (m, 2H), 7.42 (dd, J = 7.7, 4.6 Hz, 1H), 7.19 (d, J = 8.2 Hz, 1H), 5.30 (s, 2H), 4.77 (s, 4H), 2.87 (d, J = 10.9 Hz, 2H), 2.55 (s, 1H), 2.20 (s, 3H), 1.99-1.94 (m, 2H), 1.75-1.69 (m, 4H).266454455δ 8.55 (s, 1H), 8.52 (s, 1H), 8.01- 7.94 (m, 2H), 7.57 (d, J = 8.2 Hz, 1H), 7.42 (s, 1H), 7.26-7.19 (m, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.81 (s, 4H), 3.87 (s, 2H), 3.70 (t, J = 12.5 Hz, 4H).267383384δ 8.18 (s, 1H), 7.99-7.92 (m, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.14 (d, J = 8.2 Hz, 1H), 5.06 (s, 2H), 4.66-4.60 (m, 2H), 4.37 (t, J = 6.3 Hz, 2H), 3.97- 3.89 (m, 1H), 3.58 (d, J = 10.6 Hz, 2H), 3.40 (d, J = 10.2 Hz, 2H), 1.87 (s, 1H), 1.54 (s, 2H), 1.23 (s, 1H).268395396δ 10.20 (s, 1H), 8.01-7.91 (m, 2H), 7.58 (d, J = 8.2 Hz, 1H), 7.25- (m, 3H), 5.08 (s, 2H), 3.30-3.21 (m, 2H), 2.98 (d, J = 8.9 Hz, 2H), 2.69 (t, J = 6.3 Hz, 2H), 2.54 (t, J = 7.8 Hz, 2H), 2.46-2.37 (m, 1H), 2.32 (t, J = 8.8 Hz, 3H), 2.15 (s, 3H), 1.86 (s, 2H).269424425CDCl3δ 7.90-7.77 (m, 2H), 7.34 (d, J = 8.1 Hz, 1H), 7.10 (dd, J = 15.9, 8.4 Hz, 3H), 6.73 (s, 1H), 4.54 (s, 2H), 3.75 (d, J = 9.5 Hz, 2H), 3.60 (d, J = 9.3 Hz, 2H), 2.51 (s, 7H), 2.36 (d, J = 6.6 Hz, 2H), 2.31 (s, 4H), 1.51 (s, 2H), 0.87 (m, 1H)270381382CDCl3δ 7.82 (dd, J = 8.6, 5.5 Hz, 2H), 7.35 (d, J = 8.1 Hz, 1H), 7.10 (dd, J = 16.2, 7.9 Hz, 3H), 6.72 (s, 1H), 4.54 (s, 2H), 3.72 (d, J = 9.2 Hz, 2H), 3.59 (d, J = 9.3 Hz, 2H), 3.30 (t, J = 7.0 Hz, 4H), 2.40 (d, J = 6.8 Hz, 2H), 2.14 (p, J = 7.1 Hz, 2H), 1.54 (s, 2H), 0.85-0.70 (m, 1H).271454455δ 8.56 (s, 1H), 8.41 (s, 1H), 8.01- 7.94 (m, 2H), 7.74 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.78 (s, 4H), 3.78 (s, 2H), 3.63 (t, J = 12.5 Hz, 4H).272418419δ 8.55 (s, 1H), 8.36 (s, 1H), 8.02- 7.94 (m, 2H), 7.69 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.77 (s, 4H), 3.58 (s, 2H), 3.16 (t, J = 6.0 Hz, 4H), 2.04-1.95 (m, 2H).273468469δ 8.56 (s, 1H), 8.41 (s, 1H), 8.02- 7.95 (m, 2H), 7.74 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.18 (s, 2H), 4.79 (s, 4H), 3.68 (s, 2H), 2.89 (t, J = 13.3 Hz, 2H), 2.71 (t, J = 6.9 Hz, 2H), 2.34-2.19 (m, 2H).274405406CDCl3δ 8.33 (s, 1H), 7.87-7.64 (m, 2H), 7.46-7.36 (m, 2H), 7.21-7.06 (m, 3H), 6.96-6.65 (m, 1H), 5.00- 4.83 (m, 4H), 4.58 (s, 2H), 2.52 (d, J = 7.0 Hz, 2H), 1.95-1.82 (m, 1H), 0.94 (d, J = 6.6 Hz, 6H).275436437δ 8.56 (s, 1H), 8.38 (d, J = 1.5 Hz, 1H), 8.02-7.94 (m, 2H), 7.70 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.27- 7.19 (m, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.29-5.07 (m, 3H), 4.78 (s, 4H), 3.67 (s, 2H), 3.61-3.49 (m, 2H), 3.23-3.17 (m, 1H), 3.16- 3.10 (m, 1H).276356357δ 8.24 (s, 1H), 8.02-7.92 (m, 2H), 7.53 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 8.9 Hz, 2H), 7.15 (d, J = 8.2 Hz, 1H), 5.07 (s, 2H), 3.66 (d, J = 10.5 Hz, 2H), 3.42 (d, J = 10.1 Hz, 2H), 3.25 (d, J = 7.6 Hz, 5H), 1.53 (s, 2H), 0.90-0.85 (m, 1H).277350351δ 9.12 (s, 1H), 8.84 (s, 1H), 8.63 (s, 1H), 8.02-7.95 (m, 2H), 7.58 (d, J = 8.2 Hz, 1H), 7.23 (t, J = 8.9 Hz, 2H), 7.17 (d, J = 8.2 Hz, 1H), 5.19 (s, 2H), 4.83 (d, J = 7.4 Hz, 4H).Compounds 127, 138, 180, 200, 202, 207, 209, 213, 227, 233, and 234 were intentionally omitted. TABLE 3Exemplary CompoundsMSMSNo.StructureCalc.found1H NMR Data (400 MHz, DMSO-d6)350349350δ 8.83 (s, 1H), 7.75 (s, 1H), 7.65 (d, J = 7.60 Hz, 2H), 7.43 (t, J = 8.40 Hz, 2H), 7.31-7.24 (m, 3H), 7.16 (t, J = 7.20 Hz, 1H), 5.22 (t, J = 5.60 Hz, 1H), 4.75 (s, 4H), 4.52 (d, J = 5.60 Hz, 2H), 4.26 (s, 2H).351376377δ 8.79 (s, 1H), 7.72 (d, J = 2.80 Hz, 1H), 7.64 (d, J = 1.20 Hz, 2H), 7.41 (t, J = 0.80 Hz, 2H), 7.28 (t, J = 5.60 Hz, 2H), 7.17 (d, J = 1.20 Hz, 1H), 7.14 (t, J = 0.80 Hz, 1H), 4.74 (s, 4H), 4.05 (s, 2H), 3.39 (s, 2H), 2.15 (s, 6H).352417418δ 8.79 (s, 1H), 7.73 (s, 1H), 7.65 (d, J = 7.88 Hz, 2H), 7.43 (t, J = 8.16 Hz, 2H), 7.20-7.14 (m, 2H), 6.91 (d, J = 6.40 Hz, 2H), 4.69-4.66 (m, 1H), 4.06 (s, 2H), 3.14-3.12 (m, 4H), 2.47-2.46 (m, 4H), 2.23 (s, 3H).353431432δ 8.81 (s, 1H), 7.73 (s, 1H), 7.65 (d, J = 8.80 Hz, 2H), 7.42 (t, J = Hz, 2H), 7.27 (m, 3H), 7.18-7.12 (m, 1H), 4.74 (s, 4H), 4.05 (s, 2H), 3.47 (s, 2H), 2.68 (s, 3H), 2.37 (s, 5H), 2.16 (s, 3H).3542993001H-NMR (400 MHz, DMSO-d6): δ 10.18 (s, 1H), 7.72 (s, 1H), 7.61 (d, J = 7.60 Hz, 2H), 7.41 (t, J = 8.40 Hz, 2H), 7.16 (t, J = 7.20 Hz, 1H), 4.01 (s, 2H), 2.79 (d, J = 11.20 Hz, 2H), 2.15 (s, 3H), 1.89-1.82 (m, 3H), 1.74 (d, J = 10.00 Hz, 2H), 1.69-1.62 (m, 2H).356342343δ 8.90 (s, 1H), 7.69 (s, 1H), 7.61 (d, J = 7.60 Hz, 2H), 7.40 (t, J = 8.00 Hz, 2H), 7.14 (t, J = 7.60 Hz, 1H), 4.54 (t, J = 6.40 Hz, 2H), 4.45 (t, J = 6.00 Hz, 2H), 3.94 (s, 2H), 3.48 (s, 4H), 3.42 (t, J = 6.00 Hz, 1H), 2.26 (s, 4H).357285286δ 10.26 (s, 1H), 7.74 (s, 1H), 7.63 (d, J = 7.60 Hz, 2H), 7.42 (t, J = 7.60 Hz, 2H), 7.19 (d, J = 6.40 Hz, 1H), 4.04 (s, 1H), 3.13 (d, J = 10.80 Hz, 2H), 1.89 (s, 2H), 1.82 (d, J = 12.40 Hz, 2H), 1.65 (d, J = 13.20 Hz, 2H).358327328δ 10.28 (s, 1H), 7.74 (s, 1H), 7.63 (d, J = 8.00 Hz, 2H), 7.42 (t, J = 8.40 Hz, 2H), 7.18 (t, J = 7.60 Hz, 1H), 4.38 (d, J = 12.80 Hz, 1H), 4.02 (s, 1H), 3.86 (d, J = 14.00 Hz, 1H), 3.07 (t, J = 10.80 Hz, 1H), 2.67-2.64 (m, 3H), 2.01 (s, 3H), 1.82 (t, J = 14.80 Hz, 2H), 1.59 (d, J = 11.60 Hz, 1H), 1.44 (d, J = 12.40 Hz, 1H).359341342δ 10.20 (s, 1H), 7.72 (s, 1H), 7.62 (d, J = 8.00 Hz, 2H), 7.41 (t, J = 7.60 Hz, 2H), 7.16 (t, J = 7.60 Hz, 1H), 4.52 (t, J = 6.40 Hz, 2H), 4.42 (t, J = 6.00 Hz, 2H), 4.02 (s, 2H), 3.37 (t, J = 5.60 Hz, 1H), 2.73 (d, J = 11.20 Hz, 1H), 2.45-2.41 (m, 2H), 1.77 (t, J = 10.80 Hz, 4H), 1.69- 1.63 (m, 2H).360328329δ 8.98 (s, 1H), 7.70 (s, 1H), 7.61 (d, J = 8.40 Hz, 2H), 7.40 (t, J = 8.00 Hz, 2H), 7.15 (t, J = 8.00 Hz, 1H), 3.49 (s, 2H), 3.47-3.44 (m, 8H), 2.03 (s, 3H).361300301δ 8.88 (s, 1H), 7.69 (s, 1H), 7.61 (d, J = 8.40 Hz, 2H), 7.40 (t, J = 8.00 Hz, 2H), 7.14 (t, J = 7.60 Hz, 1H), 3.94 (s, 2H), 3.45 (s, 4H), 2.31 (s, 4H), 2.20 (s, 3H).362443444δ 8.48 (m, 1H), 7.99 (d, J = 5.60 Hz, 2H), 7.57 (td, J = 8.40, Hz, 1H), 7.41 (dt, J = 5.60, Hz, 2H), 7.32 (s, 1H), 7.21 (s, 3H), 6.18 (s, 1H), 5.18 (s, 2H), 4.79 (s, 4H), 3.02 (d, J = 2.80 Hz, 2H), 2.29 (s, 3H).363461462δ 8.48 (s, 1H), 7.99 (q, J = 5.72 Hz, 2H), 7.57 (d, J = 8.16 Hz, 1H), 7.31- 7.17 (m, 6H), 5.18 (s, 2H), 4.79 (s, 4H), 3.90-3.87 (m, 1H), 3.58 (s, 1H), 3.16 (s, 3H), 2.69-2.65 (m, 1H), 2.55 (d, J = 6.88 Hz, 1H), 2.44- 2.40 (m, 3H), 2.02-1.97 (m, 1H), 1.66 (t, J = 3.36 Hz, 1H).364336337δ 8.49 (s, 1H), 7.97 (q, J = 2.00 Hz, 2H), 7.57 (d, J = 8.40 Hz, 1H), 7.24 (t, J = 6.80 Hz, 2H), 7.16 (d, J = 8.00 Hz, 1H), 5.14 (s, 2H), 3.85 (t, J = 13.20 Hz, 2H), 3.65 (t, J = 7.60 Hz, 2H), 2.46 (t, J = 7.20 Hz, 2H).365377378δ 8.53 (s, 1H), 7.68 (t, J = 1.20 Hz, 1H), 7.38 (d, J = 8.00 Hz, 1H), 7.29 (t, J = 6.40 Hz, 2H), 7.23 (d, J = 8.00 Hz, 1H), 7.14 (d, J = 8.40 Hz, 1H), 6.74 (d, J = 3.20 Hz, 1H), 6.56 (q, J = 1.60 Hz, 1H), 5.20 (s, 2H), 4.77 (s, 4H), 3.41 (s, 2H), 2.16 (s, 6H).366378379δ 8.47 (s, 1H), 7.99 (q, J = 5.72 Hz, 2H), 7.58 (d, J = 8.32 Hz, 1H), 7.58 (s, 2H), 7.27-7.17 (m, 4H), 5.24- 5.19 (m, 3H), 4.79 (s, 4H), 4.53 (d, J = 5.40 Hz, 2H).367460461δ 8.47 (s, 1H), 7.99 (q, J = 5.72 Hz, 2H), 7.57 (d, J = 8.24 Hz, 1H), 7.32- 7.17 (m, 6H), 5.18 (s, 2H), 4.78 (s, 4H), 3.60 (s, 2H), 2.37-2.18 (m, 8H), 2.34 (s, 3H).368411412δ 9.98 (s, 1H), 7.97 (q, J = 6.00 Hz 2H), 7.61 (d, J = 8.40 Hz, 1H), 7.26- 7.21 (m, 3H), 5.05 (s, 2H), 2.92 (d, J = 11.20 Hz, 2H), 2.79 (d, J = 11.60 Hz, 2H), 2.17-2.11 (m, 5H), 1.91 (s, 2H), 1.84 (t, J = 12.40 Hz, 4H), 1.68-1.62 (m, 4H), 1.46-1.43 (m, 2H).369321322δ 8.63 (d, J = 5.88 Hz, 2H), 8.51 (d, J = 4.64 Hz, 1H), 8.35 (s, 1H), 7.70 (s, 1H), 7.54 (d, J = 7.76 Hz, 1H), 7.45 (d, J = 5.00 Hz, 1H), 7.29 (d, J = 8.52 Hz, 1H), 6.48 (s, 1H), 5.15 (s, 2H), 4.85 (s, 4H).370394395δ 10.05 (s, 1H), 8.31 (d, J = 2.40 Hz, 1H), 7.69 (s, 1H), 7.56 (d, J = 8.40 Hz, 1H), 7.43 (t, J = 6.00 Hz, 1H), 7.33 (t, J = 5.20 Hz, 2H), 7.21- 7.15 (m, 2H), 6.48 (t, J = 2.40 Hz, 1H), 4.99 (s, 2H), 3.54 (s, 2H), 2.88 (d, J = 11.20 Hz, 2H), 2.01 (t, J = 10.00 Hz, 2H), 1.82 (d, J = 11.60 Hz, 2H), 1.69-1.65 (m, 2H).371405406δ 8.48 (s, 1H), 8.01-7.97 (m, 2H), 7.57 (dd, J = 2.04, 8.18 Hz, 1H), 7.32-7.17 (m, 6H), 5.18 (s, 2H), 4.79 (s, 4H), 3.41 (s, 2H), 2.16 (s, 6H).372321322δ 8.62 (s, 2H), 8.51 (d, J = 5.04 Hz, 1H), 7.68 (t, J = 0.84 Hz, 1H), 7.44 (d, J = 4.84 Hz, 1H), 7.39 (d, J = 8.16 Hz, 1H), 7.14 (d, J = 8.24 Hz, 1H), 6.74 (d, J = 3.32 Hz, 1H), 6.56 (q, J = 1.76 Hz, 1H), 5.21 (s, 2H), 4.83 (s, 4H).373349350δ 8.62 (s, 1H), 8.56 (s, 1H), 8.51 (d, J = 5.08 Hz, 1H), 7.99 (q, J = 5.64 Hz, 2H), 7.58 (d, J = 8.24 Hz, 1H), 7.45 (s, 1H), 7.23 (t, J = 8.96 Hz, 2H), 7.17 (d, J = 8.28 Hz, 1H), 5.19 (s, 2H), 4.85 (s, 4H)374446447δ 8.44 (s, 1H), 7.99 (q, J = 5.64 Hz, 2H), 7.57 (d, J = 8.24 Hz, 1H), 7.25- 7.17 (m, 4H), 6.92 (d, J = 6.56 Hz, 2H), 5.17 (s, 2H), 4.71 (s, 4H), 3.34 (s, 4H), 3.14 (s, 4H), 2.25 (s, 3H)375341342(MeOD) δ 9.10 (s, 1H), 8.48 (d, J = 4.32 Hz, 1H), 8.35 (d, J = 8.08 Hz, 1H), 7.65 (d, J = 8.28 Hz, 1H), 7.49 (q, J = 4.88 Hz, 1H), 7.31 (d, J = 8.24 Hz, 1H), 4.10 (d, J = 6.00 Hz, 2H), 3.13 (d, J = 12.40 Hz, 2H), 2.49 (s, 3H), 2.40 (t, J = 10.40 Hz, 2H), 1.96-1.86 (m, 3H), 1.47 (t, J = 16.00 Hz, 2H).376314315δ 10.04 (s, 1H), 7.95 (q, J = 5.60 Hz, 2H), 7.59 (d, J = 8.40 Hz, 1H), 7.25-7.18 (m, 3H), 5.15 (s, 2H), 2.87-2.85 (m, 2H), 2.45 (m, 2H), 2.18 (s, 3H), 1.89 (s, 3H).377387388(MeOH) δ 8.53-8.51 (m, 1H), 8.05- 7.75 (m, 3H), 7.60 (d, J = 8.00 Hz, 2H), 7.41-7.30 (m, 5H), 3.79 (s, 2H), 3.11 (d, J = 11.60 Hz, 2H), 2.62 (s, 1H), 2.36 (t, J = 9.60 Hz, 2H), 2.03-1.91 (m, 4H)378405406δ 10.02 (s, 1H), 9.12 (d, J = 2.00 Hz, 1H), 8.50 (dd, J = 1.60, 4.80 Hz, 1H), 8.26 (dd, J = 2.00, 5.00 Hz, 1H), 7.71 (d, J = 8.40 Hz, 1H), 7.44 (t, J = 2.40 Hz, 2H), 7.42 (m, 1H), 7.31 (d, J = 1.60 Hz, 1H), 7.23 (q, J = 6.40 Hz, 2H), 5.16 (s, 2H), 3.54 (s, 2H), 2.89 (d, J = 11.60 Hz, 2H), 2.02 (t, J = 9.60 Hz, 2H), 1.91 (s, 1H), 1.83 (d, J = 10.80 Hz, 2H), 1.73-1.67 (m, 2H).379404405δ 10.01 (s, 1H), 7.93 (d, J = 7.00 Hz, 2H), 7.63 (d, J = 8.32 Hz, 1H), 7.45-7.39 (m, 3H), 7.34-7.28 (m, 2H), 7.24-7.15 (m, 3H), 5.05 (s, 2H), 3.54 (s, 2H), 2.89 (d, J = 11.60 Hz, 2H), 2.51 (s, 1H), 2.02 (t, J = 10.76 Hz, 2H), 1.83 (d, J = 13.16 Hz, 2H), 1.69 (d, J = 11.52 Hz, 2H).380338339δ 7.94 (t, J = 7.20 Hz, 2H), 7.63 (d, J = 8.40 Hz, 1H), 7.42 (t, J = 8.00 Hz, 2H), 7.30 (t, J = 7.20 Hz, 1H), 7.23 (d, J = 8.00 Hz, 1H), 5.06 (s, 2H), 4.40 (d, J = 13.60 Hz, 1H), 3.88 (d, J = 13.20 Hz, 1H), 3.07 (t, J = 12.00 Hz, 1H), 2.89-2.74 (m, 1H), 2.63-2.51 (m, 2H), 2.02 (s, 3H), 1.88 (t, J = 12.80 Hz, 2H), 1.62 (d, J = 11.60 Hz, 1H), 1.49 (d, J = 4.00 Hz, 1H).381352353δ 10.02 (s, 1H), 7.93 (d, J = 7.60 Hz, 2H), 7.63 (d, J = 8.40 Hz, 1H), 7.41 (t, J = 8.00 Hz, 2H), 7.31 (d, J = 7.60 Hz, 1H), 7.23 (d, J = 8.40 Hz, 1H), 5.06 (s, 1H), 4.68-4.61 (m, 2H), 4.53 (t, J = 6.40 Hz, 2H), 4.43 (t, J = 6.00 Hz, 2H), 4.35 (t, J = 6.40 Hz, 1H), 2.76 (d, J = 8.40 Hz, 2H), 1.86-1.67 (m, 6H).382313314δ 8.61 (s, 1H), 7.68 (s, 1H), 7.61 (d, J = 0.80 Hz, 2H), 7.39 (t, J = 0.40 Hz, 2H), 7.14 (t, J = 7.60 Hz, 1H), 3.99 (s, 2H), 3.82-3.79 (m, 2H), 3.60-3.56 (m, 2H), 3.54-3.51 (m, 2H), 3.35 (s, 2H), 2.93 (s, 2H).383416417δ 8.81 (s, 1H), 7.73 (s, 1H), 7.65 (d, J = 8.08 Hz, 2H), 7.43 (t, J = 7.80 Hz, 2H), 7.29-7.14 (m, 4H), 4.73 (s, 4H), 4.06 (s, 2H), 2.87 (d, J = 11.32 Hz, 2H), 2.20 (s, 3H), 1.96-1.91 (m, 2H), 1.72-1.68 (m, 4H).384311312δ 8.55 (s, 1H), 7.70 (s, 1H), 7.62 (d, J = 8.16 Hz, 2H), 7.41 (t, J = 7.84 Hz, 2H), 7.15 (t, J = 7.44 Hz, 1H), 4.02 (s, 2H), 3.58 (q, J = 8.00 Hz, 2H), 3.19 (dd, J = 3.48, 10.94 Hz, 2H), 2.68-2.65 (m, 2H), 1.80-0.71 (m, 3H), 1.58-1.57 (m, 1H), 1.45- 1.41 (m, 2H).385340341δ 8.21 (s, 1H), 7.96 (q, J = 2.00 Hz, 2H), 7.53 (d, J = 8.40 Hz, 1H), 7.24- 7.15 (m, 3H), 5.08 (s, 2H), 3.48 (d, J = 20.00 Hz, 2H), 3.22 (d, J = 16.80 Hz, 2H), 2.67 (s, 2H), 1.80- 1.75 (m, 3H), 1.73-1.70 (m, 1H), 1.48-1.44 (m, 2H).386467468δ 8.49 (s, 1H), 8.01-7.98 (m, 2H), 7.58 (d, J = 8.16 Hz, 1H), 7.32 (t, J = 7.36 Hz, 2H), 7.27-7.17 (m, 4H), 5.18 (s, 2H), 4.79 (s, 4H), 3.65 (s, 2H), 2.87 (t, J = 13.36 Hz, 2H), 2.70 (t, J = 6.92 Hz, 2H), 2.34-0.28 (m, 2H).387445446δ 8.46 (s, 1H), 7.99 (q, J = 5.60 Hz, 2H), 7.57 (d, J = 8.40 Hz, 1H), 7.29- 7.17 (m, 6H), 5.17 (s, 2H), 4.76 (s, 4H), 2.87 (d, J = 11.20 Hz, 2H), 2.20 (s, 3H), 1.96 (q, J = 8.40 Hz, 2H), 1.89 (s, 1H), 1.72-1.65 (m, 4H).388330331δ 8.28 (s, 1H), 7.98 (q, J = 2.00 Hz, 2H), 7.54 (d, J = 8.00 Hz, 1H), 7.25- 7.16 (m, 3H), 5.11 (s, 2H), 4.00 (s, 1H), 3.50 (s, 3H), 3.34 (s, 1H), 3.17 (s, 3H), 1.99 (s, 2H).389344345δ 8.56 (s, 1H), 8.00-7.96 (m, 2H), 7.54 (dd, J = 1.20, 8.00 Hz, 1H), 7.25-7.17 (m, 3H), 5.11 (s, 2H), 4.11 (s, 1H), 3.48-3.33 (m, 7H), 1.95-1.77 (m, 4H).390368369δ 8.43 (s, 1H), 7.99-7.96 (m, 2H), 7.56 (d, J = 8.40 Hz, 1H), 7.25-7.16 (m, 3H), 5.11 (s, 2H), 3.72 (q, J = 8.40 Hz, 1H), 3.59-3.48 (m, 3H), 3.32 (s, 1H), 2.22-2.21 (m, 1H), 2.06-2.03 (m, 1H).391447448(MeOD): δ 7.93 (t, J = 5.04 Hz, 2H), 7.56 (t, J = 11.36 Hz, 2H), 7.32 (d, J = 8.16 Hz, 1H), 7.15 (t, J = 8.72 Hz, 2H), 6.80 (s, 1H), 4.76 (d, J = 16.04 Hz, 4H), 3.61 (s, 4H), 2.61 (t, J = 4.76 Hz, 4H), 2.39 (s, 3H).392342343δ 8.31 (s, 1H), 7.97 (q, J = 6.00 Hz, 2H), 7.53 (d, J = 8.40 Hz, 1H), 7.24- 7.14 (m, 3H), 5.08 (s, 2H), 3.81 (q, J = 6.80 Hz, 2H), 3.64-3.54 (m, 4H), 3.38 (d, J = 2.40 Hz, 2H), 2.94 (d, J = 2.40 Hz, 2H).393375376δ 9.15 (s, 1H), 8.53-8.48 (m, 2H), 8.29 (d, J = 7.92 Hz, 1H), 7.68 (d, J = 8.12 Hz, 1H), 7.45-7.42 (m, 1H), 7.34 (t, J = 6.96 Hz, 2H), 7.27 (d, J = 8.00 Hz, 1H), 7.21 (d, J = 8.16 Hz, 1H), 5.31 (s, 2H), 4.80 (s, 4H), 4.44 (s, 2H), 3.31 (s, 3H).394392393δ 8.48 (s, 1H), 6.24 (q, J = 2805.60 Hz, 2H), 7.58 (d, J = 8.12 Hz, 1H), 7.34-7.17 (m, 6H), 5.18 (s, 3H), 4.77 (s, 5H), 1.34 (d, J = 6.36 Hz, 3H).395361362δ 9.15 (d, J = 2.00 Hz, 1H), 8.50- 8.47 (m, 2H), 8.28 (d, J = 8.00 Hz, 1H), 7.67 (d, J = 8.40 Hz, 1H), 7.42 (q, J = 4.40 Hz, 1H), 7.31 (d, J = 2.80 Hz, 2H), 7.26 (d, J = 8.00 Hz, 1H), 7.20 (d, J = 8.00 Hz, 1H), 5.30 (s, 2H), 5.22 (t, J = 5.60 Hz, 1H), 4.79 (s, 4H), 4.53 (d, J = 5.60 Hz, 2H).396392393δ 8.49 (s, 1H), 7.99 (q, J = 5.64 Hz, 2H), 7.58 (d, J = 8.16 Hz, 1H), 7.34 (t, J = 7.00 Hz, 2H), 7.28-7.17 (m, 4H), 5.19 (s, 2H), 4.80 (s, 4H), 4.44 (s, 2H), 3.31 (s, 3H).397349350δ 8.58 (s, 1H), 8.49 (d, J = 4.80 Hz, 1H), 7.99 (q, J = 6.00 Hz, 2H), 7.82 (d, J = 7.60 Hz, 1H), 7.59 (d, J = 8.40 Hz, 1H), 7.34 (q, J = 4.80 Hz, 1H), 7.23 (t, J = 8.80 Hz, 2H), 7.18 (d, J = 8.40 Hz, 1H), 5.20 (s, 2H), 4.81 (s, 4H).398433434δ 8.45 (s, 1H), 7.53-7.49 (m, 2H), 7.42 (d, J = 4.72 Hz, 1H), 7.30-7.25 (m, 3H), 7.13 (d, J = 8.04 Hz, 1H), 7.07 (s, 1H), 5.16 (s, 2H), 4.77 (s, 4H), 3.22 (d, J = 6.04 Hz, 1H), 2.32 (d, J = 8.96 Hz, 2H), 1.91 (s, 1H), 1.68 (s, 4H), 1.31 (d, J = 6.04 Hz, 3H).399445446(MeOD): δ 7.92 (q, J = 5.60 Hz, 2H), 7.53 (d, J = 8.00 Hz, 1H), 7.39- 7.31 (m, 4H), 7.14 (t, J = 8.80 Hz, 2H), 4.87-4.92 (m, 4H), 3.50-3.49 (m, 1H), 2.76 (s, 2H), 2.54 (s, 2H), 1.89-1.84 (m, 4H), 1.50 (d, J = 6.80 Hz, 3H).400455456δ 8.45 (s, 1H), 7.53-7.48 (m, 2H), 7.41 (d, J = 0.80 Hz, 1H), 7.32 (t, J = 7.20 Hz, 2H), 7.25 (d, J = 7.60 Hz, 1H), 7.13 (d, J = 8.00 Hz, 1H), 7.07 (t, J = 1.20 Hz, 1H), 5.16 (s, 2H), 4.78 (s, 4H), 3.65 (s, 2H), 2.87 (t, J = 13.20 Hz, 2H), 2.70 (t, J = 7.20 Hz, 2H), 2.28-2.22 (m, 2H).401427428δ 8.44 (s, 1H), 7.52 (d, J = 8.00 Hz, 1H), 7.35 (d, J = 4.00 Hz, 1H), 7.29 (t, J = 6.40 Hz, 2H), 7.23 (d, J = 8.00 Hz, 1H), 7.12 (d, J = 8.00 Hz, 1H), 7.06 (d, J = 3.60 Hz, 1H), 5.26 (s, 2H), 4.77 (s, 4H), 3.40 (s, 2H), 2.15 (s, 6H).402394395δ 8.80 (s, 1H), 7.71-7.66 (m, 3H), 7.31-7.22 (m, 5H), 4.75 (s, 4H), 4.06 (s, 2H), 3.40 (s, 2H), 2.16 (s, 6H).403432433δ 8.47 (s, 1H), 8.01-7.97 (m, 2H), 7.57 (d, J = 8.00 Hz, 1H), 7.29-7.17 (m, 6H), 5.17 (s, 2H), 4.77 (s, 4H), 3.96 (d, J = 10.80 Hz, 2H), 3.48- 3.41 (m, 2H), 2.81 (m, 1H), 1.72- 1.67 (m, 4H).404370371(MeOD): δ 8.20 (s, 1H), 7.97 (q, J = 6.00 Hz, 2H), 7.53 (d, J = 8.40 Hz, 1H), 7.24-7.15 (m, 2H), 5.10 (s, 2H), 3.64 (s, 2H), 3.55-3.51 (m, 2H), 3.51-3.32 (m, 2H), 1.81 (t, J = 6.80 Hz, 2H), 1.53 (t, J = 5.20 Hz, 4H).405431432δ 8.45 (s, 1H), 7.98 (q, J = 5.60 Hz, 2H), 7.56 (d, J = 8.40 Hz, 1H), 7.30- 7.16 (m, 6H), 5.17 (s, 2H), 4.77 (s, 4H), 3.58 (s, 2H), 2.43 (s, 4H), 1.69 (s, 4H).406341342δ 9.98 (s, 1H), 7.98-7.95 (m, 2H), 7.61 (d, J = 8.00 Hz, 1H), 7.26-7.21 (m, 3H), 5.05 (s, 2H), 4.35 (s, 2H), 3.05 (s, 1H), 1.84-1.64 (m, 6H), 1.62 (dd, J = 4.80, 12.60 Hz, 2H).407341342(MeOD): δ 7.74 (s, 1H), 7.63 (d, J = 8.00 Hz, 2H), 7.43 (t, J = 7.20 Hz, 2H), 7.23 (t, J = 6.40 Hz, 1H), 3.77 (m, 2H), 3.70 (m, 2H), 3.59 (m, 2H), 3.44 (s, 2H), 1.95 (t, J = 10.00 Hz, 2H), 1.66 (t, J = 4.80 Hz, 4H).408499500δ 8.45 (s, 1H), 7.98 (q, J = 6.00 Hz, 2H), 7.56 (d, J = 8.40 Hz, 1H), 7.32- 7.29 (m, 2H), 7.26-7.16 (m, 4H), 5.16 (s, 2H), 4.78 (s, 4H), 3.61 (q, J = 13.20 Hz, 2H), 3.06 (s, 1H), 2.72 (t, J = 9.20 Hz, 1H), 2.55-2.54 (m, 2H), 2.06-1.98 (m, 1H), 1.82-1.73 (m, 1H).409393394δ 8.43 (s, 1H), 7.51-7.48 (m, 2H), 7.41 (d, J = 1.20 Hz, 1H), 7.30-7.28 (m, 2H), 7.25 (d, J = 15.60 Hz, 1H), 7.12 (d, J = 8.00 Hz, 1H), 7.07-7.05 (m, 1H), 5.15 (s, 2H), 4.77 (s, 4H), 3.39 (s, 2H), 2.14 (s, 6H).410419420δ 8.44 (s, 1H), 7.50 (dd, J = 3.60, 11.60 Hz, 2H), 7.42 (d, J = 6.80 Hz, 1H), 7.29-7.25 (m, 3H), 7.17 (d, J = 20.00 Hz, 1H), 7.13-7.12 (m, 1H), 5.16 (s, 2H), 4.77 (s, 4H), 3.59 (s, 2H), 2.43 (s, 4H), 1.70 (s, 4H).411432433δ 8.56 (s, 1H), 8.02-7.97 (m, 2H), 7.78-7.76 (m, 1H), 7.59 (dd, J = 3.08, 8.08 Hz, 1H), 7.38-7.36 (m, 1H), 7.26-7.17 (m, 3H), 5.19 (s, 2H), 4.78 (s, 4H), 3.72 (s, 2H), 3.37 (s, 4H), 1.72 (s, 4H).Compounds 278-349, and 355 intentionally omitted. In some embodiments, the invention includes a pharmaceutical composition comprising a compound described herein (e.g., a compound according to Formula I, II, or any of Compounds 100-128 or any of those in Tables 2 or 3) or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier. In other embodiments, the invention features a method of inhibiting HDAC activity (e.g., HDAC2 activity) in a subject comprising the step of administering to a subject in need thereof an effective amount of a compound described herein (e.g., a compound according to Formula I, II or any Compounds 100-128 or any of those in Tables 2 or 3) or a pharmaceutically acceptable salt thereof, or a composition thereof. In other embodiments, the invention features a method of treating a condition in a subject selected from a neurological disorder, memory or cognitive function disorder or impairment, extinction learning disorder, fungal disease or infection, inflammatory disease, hematological disease, and neoplastic disease, comprising administering to a subject in need thereof an effective amount of a compound described herein (e.g., a compound according to Formula I, II, or any of Compounds 100-128 or any of those in Tables 2 or 3) or a pharmaceutically acceptable salt thereof, or a composition thereof. In still other embodiments, the invention features a method of improving memory in a normal subject or treating, alleviating, or preventing memory loss or impairment in a subject comprising administering to the subject in need thereof an effective amount of a compound described herein (e.g., a compound according to Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3) or a pharmaceutically acceptable salt thereof, or a composition thereof. In certain embodiments, the condition is:a. a cognitive function disorder or impairment associated with Alzheimer's disease, Huntington's disease, seizure induced memory loss, schizophrenia, Rubinstein Taybi syndrome, Rett Syndrome, Fragile X, Lewy body dementia, vascular dementia, frontotemporal dementia, ADHD, dyslexia, bipolar disorder and social, cognitive and learning disorders associated with autism, traumatic head injury, attention deficit disorder, anxiety disorder, conditioned fear response, panic disorder, obsessive compulsive disorder, posttraumatic stress disorder (PTSD), phobia, social anxiety disorder, substance dependence recovery, Age Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), ataxia, or Parkinson's disease; orb. a hematological disease selected from acute myeloid leukemia, acute promyelocytic leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia, myelodysplastic syndromes, and sickle cell anemia; orc. a neoplastic disease; ord. an extinction learning disorder selected from fear extinction and post-traumatic stress disorder. In further embodiments, the condition is Alzheimer's disease, Huntington's disease, frontotemporal dementia, Freidreich's ataxia, post-traumatic stress disorder (PTSD), Parkinson's disease, or substance dependence recovery. In still other embodiments, the method is a combination therapy further comprising:a. administering to the subject an effective amount of a pharmaceutically active ingredient; and/orb. exposing the subject to cognitive behavioral therapy (CBT), psychotherapy, behavioral exposure treatments, virtual reality exposure (VRE) and/or cognitive remediation therapy. In other embodiments, the method is a combination therapy for treating, alleviating, and/or preventing post-traumatic stress disorder or Alzheimer's disease and the pharmaceutically active ingredient administered is selected from Aricept®, memantine, galantamine and Excelon® (rivastigmine). In some embodiments, the invention features a method of increasing synaptic density or increasing synaptic plasticity or increasing dendritic density in a subject comprising administering to the subject in need of such increase an effective amount of a compound described herein (e.g., a compound according to Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3) or a pharmaceutically acceptable salt thereof, or a composition thereof. In still other embodiments, a compound described herein (e.g., a compound according to Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3), a pharmaceutically acceptable salt thereof, or a compound or salt (e.g., a compound according to Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3, or a pharmaceutically acceptable salt thereof) in the pharmaceutical composition selectively inhibits HDAC2. In certain embodiments, a compound described herein (e.g., a compound according to Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3), a pharmaceutically acceptable salt thereof, or a compound or salt (e.g., a compound according to Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3, or a pharmaceutically acceptable salt thereof) in the pharmaceutical composition compound has at least 2-, 5-, 10-, 15-, or 20-fold greater inhibition of HDAC2 as compared to one or more other HDAC isoforms. In further embodiments, the other HDAC isoform is HDAC1. General Synthetic Methods and Intermediates: The compounds of the present invention can be prepared by methods known to one of ordinary skill in the art and/or by reference to the schemes shown below and the synthetic examples that follow. Exemplary synthetic routes are set forth in the Schemes below, and in the Examples. One of ordinary skill in the art will recognize that numerous variations in reaction conditions including variations in solvent, reagents, catalysts, reaction temperatures and times are possible for each of the reactions described. Variation of order of synthetic steps and alternative synthetic routes are also possible. The compounds of the present invention can be prepared by methods known to one of ordinary skill in the art and/or by reference to the schemes shown below and the synthetic examples that follow. 4. Uses, Formulation and Administration Exemplary Uses Compounds of the invention are inhibitors of class I histone deacetylases (HDAC) and in particular HDAC2, and are useful for promoting cognitive function and enhancing learning and memory formation. As a result, these compounds are useful in treating, alleviating, and/or preventing various conditions, including e.g., neurological disorders, memory and cognitive function disorders/impairments, extinction learning disorders, fungal diseases, inflammatory diseases, hematological diseases, and neoplastic diseases in humans and animals. HDAC Inhibition The compounds of the present invention are useful in a variety of applications for human and animal health. The compounds of the invention are histone deacetylase (HDAC) inhibitors. A histone deacetylase inhibitor as used herein is a compound that inhibits, reduces, or otherwise modulates the activity of histone deacetylase. HDACs catalyze the removal of acetyl groups from lysine residues on proteins, including histones. HDAC inhibitors also show diverse biological functions including effecting gene expression, cell differentiation, cell cycle progression, growth arrest, and/or apoptosis. (J. Med. Chem. 2003, 46:5097 and Curr. Med. Chem. 2003, 10:2343). In various embodiments, the compounds of the invention reduce HDAC activity by at least about 50%, at least about 75%, or at least about 90% or more. In further embodiments, HDAC activity is reduced by at least about 95% or at least about 99% or more. One aspect of the invention provides a method of inhibiting histone deacetylase in a cell, comprising contacting a cell in which inhibition of histone deacetylase is desired with an inhibition effective amount of a compound of the invention or a composition thereof. Because compounds of the invention inhibit histone deacetylase(s), they are useful research tools for in vitro study of the role of histone deacetylase in biological processes. Accordingly, in one aspect of the invention, the step of contacting the cell is performed in vitro. The term an “inhibiting effective amount” is meant to denote a dosage sufficient to cause inhibition of activity of one or more histone deacetylase in a cell, which cell can be in a multicellular organism. The multicellular organism can be a plant, a fungus, or an animal, preferably a mammal, more preferably a human. The fungus may be infecting a plant or a mammal, preferably a human, and could therefore be located in and/or on the plant or mammal. If the histone deacetylase is in a multicellular organism, the method according to this aspect of the invention comprises administering to the organism a compound or composition of the invention. Measurement of the effect of a compound of the invention on the enzymatic activity of a histone deacetylase is achieved using known methodologies. For example, Bradner, J. et al. Nature Chemical Biology, Vol. 6, March 2010, 238-243. The potential of HDAC inhibitors is tremendous, but the development of clinical compounds will likely require the design of isoform selective compounds to minimize side effect issues e.g., fatigue, anorexia, hematological and GI-toxicity. Isoform specific HDAC inhibitors provide advantages by reducing toxicities associated with inhibition of other HDACs. Specific HDAC inhibitors provide a higher therapeutic index, resulting in better tolerance by patients during chronic or long term treatment. While several HDAC inhibitors are now in the clinic, most of these do not show significant selectivity for individual HDAC isoforms. The compounds of the present invention inhibit HDAC2. In some embodiments, the compound reduces the activity of other, but fewer than all histone deacetylases in the cell. In certain embodiments, the compound reduces the activity of HDAC2 to a greater extent than other histone deacetylases. In certain embodiments, the present invention relates to the aforementioned compound, wherein the compounds of the invention are selective HDAC2 inhibitors. In one embodiment, a compound of the invention is selective for HDAC2 and will have at least about 2-fold (e.g., at least about 5-fold, 10-fold, 15-fold, or 20-fold) greater activity to inhibit HDAC2 as compared to one or more other HDACs (e.g., one or more HDACs of class I or II). In one embodiment, a compound of the invention will have at least about 2-fold (e.g., at least about 5-fold, 10-fold, 15-fold, or 20-fold) greater activity to inhibit HDAC2 as compared to HDAC3. In one embodiment, a compound of the invention will have at least about 2-fold (e.g., at least about 5-fold, 10-fold, 15-fold, or 20-fold) greater activity to inhibit HDAC2 as compared to HDAC1. In one embodiment, a compound of the invention will have at least about 2-fold (e.g., at least about 5-fold, 10-fold, 15-fold, or 20-fold) greater activity to inhibit HDAC2 as compared to all other HDACs of a particular class of HDACs (e.g., one or more HDACs of class I or II). In one embodiment, a compound of the invention will have at least about 2-fold (e.g., at least about 5-fold, 10-fold, 15-fold, or 20-fold) greater activity to inhibit HDAC2 as compared to all other HDACs. In another embodiment, a compound selectively inhibits HDAC2 with an IC50value greater than 0.0000001 μM and less than or equal to 0.1 μM, 1 μM, 5 μM, or 30 μM. The compounds described herein (e.g., a Compound of Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3) provide an additional mechanism by which selectivity and an increased margin of safety may be obtained. In some embodiments, the compounds described herein (e.g., a Compound of Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3) partially inhibit the activity of HDAC2 within specific cells. Without being limited by theory, this partial inhibition is hypothesized to be the result of differential potency on HDAC2 when it resides within a multiple protein complex in the cell. The multiple protein complexes which contain HDAC2 vary between cells, with specific complexes within specific cell types. Accordingly, the compounds described herein (e.g., a Compound of Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3) compounds are proposed to incompletely inhibit the activity of HDAC2 in some complexes, sparing sufficient function of HDAC2 to provide an improved margin of safety while maintaining enough inhibition to result in the desired effect. Neurological Disorders In one aspect, the invention provides methods and compositions for treating, alleviating, and/or preventing neurological disorders. Recent reports have detailed the importance of histone acetylation in central nervous system (“CNS’) functions such as neuronal differentiation, memory formation, drug addiction, and depression (Citrome, Psychopharmacol. Bull. 2003, 37, Suppl. 2, 74-88; Johannessen, CNS Drug Rev. 2003, 9, 199-216; Tsankova et al., 2006, Nat. Neurosci. 9, 519-525). In one aspect, the invention provides methods and compositions for treating, alleviating, and/or preventing neurological disorders. The term “neurological disorder” as used herein includes neurological diseases, neurodegenerative diseases and neuropsychiatric disorders. A neurological disorder is a condition having as a component a central or peripheral nervous system malfunction. Neurological disorders may cause a disturbance in the structure or function of the nervous system resulting from developmental abnormalities, disease, genetic defects, injury or toxin. These disorders may affect the central nervous system (e.g., the brain, brainstem and cerebellum), the peripheral nervous system (e.g., the cranial nerves, spinal nerves, and sympathetic and parasympathetic nervous systems) and/or the autonomic nervous system (e.g., the part of the nervous system that regulates involuntary action and that is divided into the sympathetic and parasympathetic nervous systems). As used herein, the term “neurodegenerative disease” implies any disorder that might be reversed, deterred, managed, treated, improved, or eliminated with agents that stimulate the generation of new neurons. Examples of neurodegenerative disorders include: (i) chronic neurodegenerative diseases such as familial and sporadic amyotrophic lateral sclerosis (FALS and ALS, respectively), familial and sporadic Parkinson's disease, Huntington's disease, familial and sporadic Alzheimer's disease, multiple sclerosis, muscular dystrophy, olivopontocerebellar atrophy, multiple system atrophy, Wilson's disease, progressive supranuclear palsy, diffuse Lewy body disease, corticodentatonigral degeneration, progressive familial myoclonic epilepsy, strionigral degeneration, torsion dystonia, familial tremor, Down's Syndrome, Gilles de la Tourette syndrome, Hallervorden-Spatz disease, diabetic peripheral neuropathy, dementia pugilistica, AIDS Dementia, age related dementia, age associated memory impairment, and amyloidosis-related neurodegenerative diseases such as those caused by the prion protein (PrP) which is associated with transmissible spongiform encephalopathy (Creutzfeldt-Jakob disease, Gerstmann-Straussler-Scheinker syndrome, scrapie, and kuru), and those caused by excess cystatin C accumulation (hereditary cystatin C angiopathy); and (ii) acute neurodegenerative disorders such as traumatic brain injury (e.g., surgery-related brain injury), cerebral edema, peripheral nerve damage, spinal cord injury, Leigh's disease, Guillain-Barre syndrome, lysosomal storage disorders such as lipofuscinosis, Alper's disease, restless leg syndrome, vertigo as result of CNS degeneration; pathologies arising with chronic alcohol or drug abuse including, for example, the degeneration of neurons in locus coeruleus and cerebellum, drug-induced movement disorders; pathologies arising with aging including degeneration of cerebellar neurons and cortical neurons leading to cognitive and motor impairments; and pathologies arising with chronic amphetamine abuse to including degeneration of basal ganglia neurons leading to motor impairments; pathological changes resulting from focal trauma such as stroke, focal ischemia, vascular insufficiency, hypoxic-ischemic encephalopathy, hyperglycemia, hypoglycemia or direct trauma; pathologies arising as a negative side-effect of therapeutic drugs and treatments (e.g., degeneration of cingulate and entorhinal cortex neurons in response to anticonvulsant doses of antagonists of the NMDA class of glutamate receptor) and Wernicke-Korsakoff's related dementia. Neurodegenerative diseases affecting sensory neurons include Friedreich's ataxia, diabetes, peripheral neuropathy, and retinal neuronal degeneration. Other neurodegenerative diseases include nerve injury or trauma associated with spinal cord injury. Neurodegenerative diseases of limbic and cortical systems include cerebral amyloidosis, Pick's atrophy, and Rett syndrome. The foregoing examples are not meant to be comprehensive but serve merely as an illustration of the term “neurodegenerative disorder.” In some instances the neurological disorder is a neuropsychiatric disorder, which refers to conditions or disorders that relate to the functioning of the brain and the cognitive processes or behavior. Neuropsychiatric disorders may be further classified based on the type of neurological disturbance affecting the mental faculties. The term “neuropsychiatric disorder,” considered here as a subset of “neurological disorders,” refers to a disorder which may be generally characterized by one or more breakdowns in the adaptation process. Such disorders are therefore expressed primarily in abnormalities of thought, feeling and/or behavior producing either distress or impairment of function (i.e., impairment of mental function such with dementia or senility). Currently, individuals may be evaluated for various neuropsychiatric disorders using criteria set forth in the most recent version of the American Psychiatric Association's Diagnostic and Statistical Manual of Mental Health (DSM-IV). One group of neuropsychiatric disorders includes disorders of thinking and cognition, such as schizophrenia and delirium. A second group of neuropsychiatric disorders includes disorders of mood, such as affective disorders and anxiety. A third group of neuropsychiatric disorders includes disorders of social behavior, such as character defects and personality disorders. A fourth group of neuropsychiatric disorders includes disorders of learning, memory, and intelligence, such as mental retardation and dementia. Accordingly, neuropsychiatric disorders encompass schizophrenia, delirium, attention deficit disorder (ADD), schizoaffective disorder, Alzheimer's disease, Rubinstein-Taybi syndrome, depression, mania, attention deficit disorders, drug addiction, dementia, agitation, apathy, anxiety, psychoses, personality disorders, bipolar disorders, unipolar affective disorder, obsessive-compulsive disorders, eating disorders, post-traumatic stress disorders, irritability, adolescent conduct disorder and disinhibition. In one embodiment, the neurological disorder is Alzheimer's disease, Huntington's disease, seizure-induced memory loss, schizophrenia, Rubinstein Taybi syndrome, Rett Syndrome, Fragile X, Lewy body dementia, vascular dementia, ADHD, ADD, dyslexia, bipolar disorder and social, cognitive and learning disorders associated with autism, traumatic head injury, or attention deficit disorder. In another embodiment, the neurological disorder is an anxiety disorder, conditioned fear response, panic disorder, obsessive compulsive disorder, post-traumatic stress disorder, phobia, social anxiety disorder, or substance dependence recovery. In some embodiments neurological disorders are treated or prevented by decreasing the amount of DNA damage within the neuronal cell. In some embodiments neurological disorders are treated or prevented by increasing histone deacetylase activity within the neuronal cell. In some embodiments neurological disorders are treated or prevented by decreasing histone acetyl transferase activity within the neuronal cell. In some embodiments neurological disorders are treated or prevented by increasing the activity of class I histone deacetylases. Enhancing Cognitive Function In one aspect, the invention provides methods and compositions for promoting cognitive function and enhancing learning and memory formation in both normal subjects as well as those suffering from memory loss and cognitive function disorders/impairments. A normal subject, as used herein, is a subject that has not been diagnosed with a disorder associated with impaired cognitive function. “Cognitive function” refers to mental processes of a subject relating to information gathering and/or processing; the understanding, reasoning, and/or application of information and/or ideas; the abstraction or specification of ideas and/or information; acts of creativity, problem-solving, and possibly intuition; and mental processes such as learning, perception, and/or awareness of ideas and/or information. The mental processes are distinct from those of beliefs, desires, and the like. Memory Disorders/Impairment Transcription is thought to be a key step for long-term memory processes (Alberini, 2009, Physiol. Rev. 89, 121-145). Transcription is promoted by specific chromatin modifications, such as histone acetylation, which modulate histone-DNA interactions (Kouzarides, 2007, Cell, 128:693-705). Modifying enzymes, such as histone acetyltransferases (HATs) and histone deacetylases (HDACs), regulate the state of acetylation on histone tails. In general, histone acetylation promotes gene expression, whereas histone deacetylation leads to gene silencing. Numerous studies have shown that a potent HAT, cAMP response element-binding protein (CREB)-binding protein (CBP), is necessary for long-lasting forms of synaptic plasticity and long term memory (for review, see Barrett, 2008, Learn Mem 15:460-467). A “memory” as used herein refers to the ability to recover information about past events or knowledge. Memories include short-term memory (also referred to as working or recent memory) and long-term memory. Short-term memories involve recent events, while long-term memories relate to the recall of events of the more distant past. Methods of assessing the ability to recall a memory are known to those of skill in the art and may involve routine cognitive tests. Enhancing or retrieving memories is distinct from learning. However, in some instances in the art learning is referred to as memory. Learning, unlike memory enhancement, refers to the ability to create new memories that had not previously existed. Thus in order to test the ability of a compound to effect the ability of a subject to learn rather than recall old memories, the compound would be administered prior to or at the same time as the memory is created. In order to test the ability of a compound to affect recall of a previously created memory the compound is administered after the memory is created and preferably after the memory is lost. As used herein “age related memory loss” refers to any of a continuum of conditions characterized by a deterioration of neurological functioning that does not rise to the level of a dementia, as further defined herein and/or as defined by the Diagnostic and Statistical Manual of Mental Disorders: 4th Edition of the American Psychiatric Association (DSM-IV, 1994). Age related memory loss is characterized by objective loss of memory in an older subject compared to his or her younger years, but cognitive test performance that is within normal limits for the subject's age. Age related memory loss subjects score within a normal range on standardized diagnostic tests for dementias, as set forth by the DSM-IV. Moreover, the DSM-IV provides separate diagnostic criteria for a condition termed Age-Related Cognitive Decline. In the context of the present invention, as well as the terms “Age-Associated Memory Impairment” and “Age-Consistent Memory Decline” are understood to be synonymous with the age related memory loss. Age-related memory loss may include decreased brain weight, gyral atrophy, ventricular dilation, and selective loss of neurons within different brain regions. For purposes of some embodiments of the present invention, more progressive forms of memory loss are also included under the definition of age-related memory disorder. Thus persons having greater than age-normal memory loss and cognitive impairment, yet scoring below the diagnostic threshold for frank dementia, may be referred to as having a mild neurocognitive disorder, mild cognitive impairment, late-life forgetfulness, benign senescent forgetfulness, incipient dementia, provisional dementia, and the like. Such subjects may be slightly more susceptible to developing frank dementia in later life (See also US patent application 2006/008517 (Vasogen Ireland limited) which is incorporated by reference). Symptoms associated with age-related memory loss include but are not limited to alterations in biochemical markers associated with the aging brain, such as IL-1 beta, IFN-gamma, p-JNK, p-ERK, reduction in synaptic activity or function, such as synaptic plasticity, evidenced by reduction in long term potentiation, diminution of memory and learning. As used herein “injury related memory loss” refers to a loss of memory wherein there is damage to the brain, and there may have also been neurological damage. Sources of brain injury include traumatic brain injury such as concussive injuries or penetrating head wounds, brain tumors, alcoholism, Alzheimer's disease, stroke, heart attack and other conditions that deprive the brain of oxygen, meningitis, AIDS, viral encephalitis, and hydrocephalus. Methods for enhancing memories can include reestablishing access to memories as well as recapturing memories. The term re-establishing access as used herein refers to increasing retrieval of a memory. Although Applicants are not bound by a mechanism of action, it is believed that the compounds of the invention are effective in increasing retrieval of memories by re-establishing a synaptic network. The process of re-establishing a synaptic network may include an increase in the number of active brain synapses and or a reversal of neuronal loss. Neurogenesis, or the birth of new neuronal cells, was thought to occur only in developing organisms. However, recent research has demonstrated that neurogenesis does indeed continue into and throughout adult life. On going neurogenesis is thought to be an important mechanism underlying neuronal plasticity, enabling organisms to adapt to environmental changes and influencing learning and memory throughout life. In one aspect, the invention includes a method of increasing synaptic density in a subject comprising administering to the subject in need of such increase a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof. In one aspect, the invention includes a method of increasing synaptic plasticity in a subject comprising administering to the subject in need of such increase a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof. In one aspect, the invention includes a method of increasing dendritic density in neurons in a subject comprising administering to the subject in need of such increase a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof. The invention provides methods for enhancing memory in a subject having a memory disorder. Examples of types of memory disorders include Alzheimer's disease, absent-minded professor, absent-mindedness, amnesia, anterograde amnesia, blackout (alcohol-related amnesia), bromism, childhood amnesia, false memory syndrome, fugue state, hyperthymesia, Korsakoff s syndrome, lacunar amnesia, memory distrust syndrome, memory loss, post-traumatic amnesia, prosopamnesia, psychogenic amnesia, repressed memory, retrograde amnesia, Ribot's Law, selective memory loss, source amnesia, source-monitoring error, the seven sins of memory, tip of the tongue, transient epileptic amnesia, transient global amnesia, and twilight sleep. In one embodiment, Alzheimer's disease is the memory disorder. Such methods optionally involve administering the inhibitor and monitoring the subject to identify recapture of a memory that was previously lost. Subjects may be monitored by routine tests known in the art. In other embodiments the alzheimer's subject is one that has late stage Alzheimer's disease. Many of the drugs suggested for treating Alzheimer's disease are designed to treat the early stages of the disease by preventing plaque buildup. The compounds of the invention are useful for treating both early stages and late stages of dementia because they actually improve memory and cognition rather than preventing only plaque accumulation. Cognitive Function Disorders/Impairment The invention relates to methods of treating, alleviating, and/or preventing cognitive function disorders/impairments. Impaired cognitive function refers to cognitive function that is not as robust as that observed in an age-matched normal subject and includes states in which cognitive function is reduced. In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function measured in an age-matched normal subject. Cognitive function may be promoted to any detectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to carry out daily activities of normal life. In some embodiments, the cognitive function disorders or impairments are associated with, but not limited to, Alzheimer's disease, Huntington's disease, seizure induced memory loss, schizophrenia, Rubinstein Taybi syndrome, Rett Syndrome, Fragile X, Lewy body dementia, Vascular dementia, bipolar disorder and social, cognitive and learning disorders associated with autism, attention deficit hyperactivity disorder (ADHD), dyslexia, learning disorders, traumatic head injury, stroke induced cognitive and motor impairment, traumatic brain injury, neurodegeneration and neuronal loss mediated cognitive impairment, and attention deficit disorder. In some embodiments, the cognitive function disorders or impairments are associated with, but not limited to, anxiety disorders, conditioned fear response, panic disorders, obsessive compulsive disorders, post-traumatic stress disorder, phobias, social anxiety disorders, substance dependence recovery or Age Associated Memory Impairment (AAMI), and Age Related Cognitive Decline (ARCD). In some embodiments, the invention relates to methods of treating, alleviating, and/or preventing vascular dementia. Vascular dementia, also referred to as “multi-infarct dementia”, refers to a group of syndromes caused by different mechanisms all resulting in vascular lesions in the brain. The main subtypes of vascular dementia are, for example vascular mild cognitive impairment, multi-infarct dementia, vascular dementia due to a strategic single infarct (affecting the thalamus, the anterior cerebral artery, the parietal lobes or the cingulate gyms), vascular dementia due to hemorrhagic lesions, small vessel disease (including, e.g. vascular dementia due to lacunar lesions and Binswanger disease), and mixed Alzheimer's Disease with vascular dementia. In some embodiments, the invention relates to treating, alleviating, and/or preventing Huntington's disease. Huntington's disease is a neurological disease which results in cognitive decline associated with inexorable progression to death. Cognitive symptoms associated with Huntington's disease include loss of intellectual speed, attention, and short term memory and/or behavioral symptoms. Cognitive function may be assessed, and thus optionally defined, via one or more tests or assays for cognitive function. Non-limiting examples of a test or assay for cognitive function include CANTAB (see for example Fray et al. “CANTAB battery: proposed utility in neurotoxicology.” Neurotoxicol Teratol 1996; 18(4):499-504), Stroop Test, Trail Making, Wechsler Digit Span, or the CogState computerized cognitive test (see also Dehaene et al. “Reward-dependent learning in neuronal networks for planning and decision making.” Brain Res. 2000; 126:21729; Iverson et al. “Interpreting change on the WAIS-III/WMS-I11 in clinical samples.” Arch Clin Neuropsychol. 2001; 16(2): 183-91; and Weaver et al. “Mild memory impairment in healthy older adults is distinct from normal aging.” Cogn. 2006; 60(2): 146-55). The methods of the invention may be used to promote cognitive function in a normal subject or to treat, alleviate and/or prevent a subject from having a cognitive dysfunction. A normal subject, as used herein, is a subject that has not been diagnosed with a disorder associated with impaired cognitive function. Extinction Teaming Disorders In one aspect, the invention relates to methods of treating, alleviating, and/or preventing extinction learning disorders e.g., a fear extinction deficit. It has been demonstrated that administration of the HD AC inhibitors sodium butyrate or trichostatin A facilitates fear extinction in mice and this enhancement mirrors that caused by commonly used behavioral manipulation and is consistent with other studies demonstrating a role for the hippocampus in the extinction of contextual fear (Lattal, et al., 2007, Behav. Neurosci. 121, 5, 1125-1131). Compounds of the invention can be used to facilitate the psychological process of extinction learning and thus are useful for treating, alleviating, and/or preventing neuropsychiatric disorders and other related disorders. Unlike traditional anti-anxiety drugs that are administered on a chronic basis and address physiological symptoms of anxiety, the compounds of the invention can be used on a chronic or acute basis in conjunction with a second therapy e.g., psychotherapy. In one aspect, the present invention is directed to methods for treating, alleviating, and/or preventing a subject from having a neuropsychiatric disorder. The methods comprise subjecting the subject to one or more sessions of a combination therapy protocol, where the combination therapy protocol comprises an acute administration of a therapeutically effective amount of a compound of the invention that enhances learning or conditioning in combination with a session of psychotherapy. By “acute administration” is intended a single exposure of the subject to the therapeutically effective amount of the compound that enhances learning or conditioning. In one aspect, the exposure to the compound occurs within about 24 hours prior to initiating the session of psychotherapy, preferably within about 12 hours, and more preferably within about 6 hours prior to initiating the session of psychotherapy. A full course of treatment for the neuropsychiatric disorder entails at least one session of this combination therapy protocol. For purposes of the present invention, a subject may have a single disorder, or may have a constellation of disorders that are to be treated, alleviated, and/or prevented by the methods described herein. The neuropsychiatric disorders contemplated in the present invention include, but are not limited to, fear and anxiety disorders, addictive disorders including substance-abuse disorders, and mood disorders. Within the fear and anxiety disorder category, the invention encompasses the treatment or prevention of panic disorder, specific phobia, post-traumatic stress disorder (PTSD), obsessive-compulsive disorder, and movement disorders such as Tourette's syndrome. The disorders contemplated herein are defined in, for example, the DSM-IV (Diagnostic and Statistical Manual of Mental Disorders (4th ed., American Psychiatric Association, Washington D.C., 1994)), which is herein incorporated by reference. Anxiety-related disorders relate to those disorders characterized by fear, anxiety, addiction, and the like. Patients with anxiety-related disorders can have a single such disorder, or can have a constellation of disorders. The anxiety-related disorders contemplated in the present invention include, but are not limited to, anxiety disorders, addictive disorders including substance-abuse disorders, mood disorders (e.g., depression and/or bipolar disorder), movement disorders such as Tourette's syndrome, psychogenic erectile dysfunction (impotence resulting from a man's inability to obtain or maintain an erection of his penis), insomnia (e.g. chronic insomnia), and eating disorders (e.g. anorexia). Anxiety disorders include, but are not limited to, panic disorder, agoraphobia, social phobia, specific phobia, PTSD, obsessive-compulsive disorder, and generalized anxiety disorder. The disorders contemplated herein are defined in, for example, the DSM-IV (Diagnostic and Statistical Manual of Mental Disorders (4th ed., American Psychiatric Association, Washington D.C., 1994)). Movement disorders are neurological conditions that affect the speed, fluency, quality, and ease of movement. Representative movement disorders include but are not limited to ataxia, chorea, myoclonus, dystonia, Parkinson's disease, restless leg syndrome, tics, and Tourette's syndrome. Movement disorders typically occur as a result of damage or disease in the basal ganglia region of the brain. Movement disorders can result from age-related changes, medications, genetic disorders, metabolic disorders, disease, stroke, or injury. Recovery of movement after stroke or injury may be facilitated when treated according to the methods of the invention. Addictive disorders are disorders characterized by addiction to an activity or substance, and include, for example, alcohol addiction, drug addiction, and gambling addiction. Depression refers to the clinical condition known as major depressive disorder, and is characterized by a state of intense sadness, melancholia, or despair that has advanced to the point of being disruptive to an individual's social functioning and/or activities of daily living. Depression is alleviated if either (or both) the severity or frequency of a symptom of the depression is reduced. However, a subject can be treated for depression in accordance with the methods of the invention irrespective of whether the treatment actually was successful in alleviating the depression. Insomnia is defined herein as the inability to fall asleep or to stay asleep for a sufficient amount of time during regular sleeping hours. It includes acute insomnia, which occurs in either a transient or short term form, and chronic insomnia. It also includes initial insomnia, defined as difficulty in falling asleep; middle insomnia, defined as awakening in the middle of the night followed by eventually falling back to sleep, but with difficulty; and terminal insomnia, defined as awakening before one's usual waking time and being unable to return to sleep. As defined by the National Institute of Mental Health, Autism Spectrum Disorders (ASD), also widely known as Pervasive Developmental Disorders (PDDs), cause severe and pervasive impairment in thinking, feeling, language, and the ability to relate to others. These disorders are usually first diagnosed in early childhood and range from a severe form, called autistic disorder, through pervasive development disorder not otherwise specified (PDD-NOS), to a much milder form, Asperger syndrome. They also include two rare disorders, Rett syndrome and childhood disintegrative disorder. Attention-Deficit Hyperactivity Disorder (ADHD) is one of the most common mental disorders that develop in children. Children with ADHD typically have impaired functioning in multiple settings, including home, school, and in relationships with peers. Symptoms of ADHD include impulsiveness, hyperactivity, and inattention. Typical treatments encompassed by the present invention include combination therapies. For instance, the combination therapy may be a pharmacotherapy (i.e., a compound of the invention) and a behavioral therapy. Behavioral therapy comprises, but is not limited to, electroconvulsive seizure therapy, exercise, group therapy, talk therapy, or conditioning. In another embodiment, the behavioral therapy is cognitive-behavioral therapy. Examples of behavioral therapy that may be used in the ongoing methods are described, for example, in Cognitive-Behavioral Therapies by K. Dobson, ed., Guilford Publications, Inc., 2002; The new Handbook of Cognitive Therapy: Basics and Beyond by Judith S. S. Beck, Guilford Publications, Inc. 1995 herein incorporated by reference in their entireties. Any pharmaceutical active ingredient that is recognized by the skilled artisan as being a pharmacologic agent that enhances learning or conditioning can be used in the methods of the invention. For example, one such class of pharmaceutical active ingredients contemplated herein comprises compounds that increase the level of norepinephrine in the brain. Such compounds include those acting as norepinephrine reuptake inhibitors, for example tomoxetine, reboxetine, duloxetine, venlafaxine, and milnacipran, and those compounds that cause release of norepinephrine, for example amphetamine, dextroamphetamine, pemoline, and methylphenidate. Another class of such pharmaceutical active ingredients is those compounds that increase the level of acetylcholine in the brain, including, for example, compounds that block its breakdown. Examples of such compounds include, but are not limited to, donepezil HCl or Aricept™ and tacrine, which inhibit cholinesterase activity. Methods of the invention also encompass the use in combination with a compound of the invention of any type of psychotherapy that is suitable for the particular psychiatric disorder for which the subject is undergoing treatment. Suitable methods of psychotherapy include exposure based psychotherapy, cognitive psychotherapy, and psychodynamically oriented psychotherapy. Methods of the invention also encompass exposing the subject to cognitive behavioral therapy (CBT), behavioral exposure treatments, virtual reality exposure (VRE) or cognitive remediation therapy. Methods of the invention also encompass extinction training. The goal of extinction training is to pair a stimulus that previously provoked a deleterious, unwanted response with a new learning that will not lead to a negative outcome, thereby generating in a subject a new, more appropriate response to the stimulus to compete with and ideally replace the previous undesirable response. Extinction training frequently exposes a subject to a stimulus or situation in the absence of an aversive consequence, e.g., a subject that has deleterious, high anxiety responses to a given stimulus or situation is exposed to that stimulus or situation in the absence of an aversive consequence. A typical goal of extinction training is to produce new learning in the subject that results from the pairing of the original stimulus or situation with a non-deleterious outcome, thereby generating, in subsequent exposures to the stimulus, a more appropriate response in place of the unwanted response. An extinction learning event refers to a completed stimulus/response extinction training cycle. One form of extinction training entails psychotherapy. For example, the methods of the invention contemplate treating, alleviating, and/or preventing anxiety disorders by: (i) administering psychotherapy to treat, alleviate, and/or prevent an anxiety-related disorder in a suitable human subject, and (ii) administering a therapeutically effective dose a compound of the invention to said subject on an achronic, post-training, pre-sleep basis. Suitable methods of psychotherapy include but are not limited to exposure-based psychotherapy, cognitive psychotherapy, and psychodynamically oriented psychotherapy. One method of psychotherapy that is specifically contemplated is the use of virtual reality (VR) exposure therapy to treat, alleviate, and/or prevent an anxiety disorder using the methods of the invention. Another method of psychotherapy that is particularly beneficial when utilized in combination with a compound or composition of the present invention is cognitive behavioral therapy (“CBT”). CBT is a form of psychotherapy that combines cognitive therapy and behavior therapy, and emphasizes the critical role of thinking in causing people to act and feel as they do. Therefore, if an individual is experiencing unwanted feelings and behaviors, CBT teaches that it is important to identify the thinking that is causing the undesirable feelings and/or behaviors and to learn how to replace this deleterious thinking with thoughts that lead to more desirable reactions. CBT is widely used to help people who are experiencing a range of mental health difficulties, some of which do not conveniently fit definitions of a particular medical affliction. CBT has been used to treat anxiety disorders, mood disorders, addictive disorders, eating disorders, insomnia, chronic pain, schizophrenia, fibromyalgia, ADHD, and autism spectrum disorders, among others. Post-extinction training pre-sleep administration of a compound of the invention, subsequent to CBT treatment, can be used to augment the effectiveness of the CBT treatment for these medical conditions. In one embodiment, subjects suffering from social anxiety disorder undergo weekly cognitive behavioral therapy sessions to treat the affliction. After each therapy session, subjects are administered a therapeutically effective formulation of compounds of the invention on a post-extinction training pre-sleep basis. Relative to subjects treated only via cognitive behavioral therapy, or to subjects treated via cognitive behavioral therapy and a placebo, anxiety associated with social anxiety disorder is expected to be reduced to a greater extent in subjects treated with a combination of cognitive behavioral therapy and achronic administration of a compound of the invention on a post-extinction training pre-sleep basis. In another embodiment of the invention, a compound of the invention is administered after extinction training only if the extinction training yields positive results on that day. For example, a subject undergoing cognitive behavioral therapy for PTSD is administered a compound of the invention on a post-extinction training only if the cognitive behavioral therapy was deemed to be successful, as determined by the subject and/or therapist. In one aspect, the compound is administered on a post-extinction, pre-sleep basis. In another aspect, a subject undergoing cognitive behavioral therapy for PTSD is administered a compound of the invention on a pre-extinction training. In one aspect, the compound is administered on a pre-extinction, pre-sleep basis. This method may also be useful when applied to treatment of autism spectrum disorders or attention-deficit hyperactivity disorder. In some embodiments, the invention relates to treating a condition where the treating comprises re-writing memories. Memories of an event often have an associated emotional component. For example, memories of a traumatic event can cause feelings of grief, guilt, or loss, as well as negative emotional responses such as anger, rage or aggression. Conversely, memories of a positive events can cause joy and increase feelings of self-confidence and self-worth. During the period of time when a memory is recalled it can modified to alter the associations and reduce or alter the emotional reactions to it. In some embodiments, HDAC inhibitors in combination with cognitive behavioral therapy or virtual reality therapy may allow the emotional associations with a memory to be re-written producing a longer term or greater therapeutic benefit. In another embodiment of the invention, subjects afflicted with anxiety disorders such as PTSD receive extinction training using Eye Movement Desensitization and Reprocessing (EMDR), and subsequently are administered a therapeutically effective dose of a compound of the invention on a post-extinction training pre-sleep basis. Another form of extinction training is provided by biofeedback, which is particularly useful in enabling subjects to learn to control physiological processes that normally occur involuntarily, such as blood pressure, heart rate, muscle tension, and skin temperature. As used herein, “biofeedback” refers to a technique in which subjects are trained to improve their health by using signals from their own bodies to control their own physiological responses. In one embodiment of the invention, a subject suffering from chronic pain undergoes biofeedback sessions to help alleviate the pain. Upon the conclusion of each session wherein the subject has made progress in learning/developing responses that reduce the chronic pain, the subject is administered a compound of the invention on a post-extinction training pre-sleep basis in order to consolidate the desired learning. In another embodiment, a subject suffering from phantom limb syndrome undergoes thermal biofeedback sessions to reduce and hopefully eliminate the symptoms. After each session, the subject is administered a therapeutically effective formulation of a compound of the invention on a post-extinction training pre-sleep basis. In another embodiment, extinction training can be provided by physical therapy, or virtual reality physical therapy such as virtual reality gait therapy. For example, a stroke victim re-learning how to walk can undergo virtual reality gait therapy, and then be administered a compound of the invention on an achronic, post-extinction training pre-sleep basis. Another form of extinction training can be provided by pharmacotherapy. See, e.g., Davis et al.,NeuroRx: The Journal of the American Society for Experimental NeuroTherapeutics,93:82-96, 2006. For example, manipulation of the endogenous cannabinoid (eCB) is of interest both because of the potential for identifying new therapeutics to treat, e.g., mental illness and disorders, but also due to the dense expression of the CB1 receptor in regions associated with, e.g., anxiety and emotional learning. For example, studies have shown that both genetic CB1 knockout mice and mice subjected to pharmacological blockate of the CB1 receptor exhibited a similar effect in extinction (Davis, page 87). Studies using the CB1 antagonist rimonabant in rats showed that systemic administration of this drug led to significant and dose-dependent decreases in extinction. Together with other studies relating to the administration of CB1 agonist WIN 55,212-2 and of an inhibitor of CB1 reuptake and breakdown, the CB1 receptor can be important in extinction learning and modulation of the endocannabinoid system can be used to decrease or increase extinction. Extinction training does not always require intervention of a trained specialist. Individuals can carry out extinction training on themselves. Fungal Diseases or Infections In some aspects, the invention relates to a method for treating, alleviating, and/or preventing a fungal disease or infection comprising administering to a subject a compound of the invention. The invention provides a method for treating, alleviating, and/or preventing hospital-acquired fungal infections that attack immunocompromised patients including those with HIV and cancer. In one embodiment, the invention provides a method for treating, alleviating, and/or preventing a fungal disease in a subject not suffering from cancer. Inflammatory Disease In some aspects, the invention relates to a method for treating, alleviating, and/or preventing an inflammatory disease, including but not limited to stroke, rheumatoid arthritis, lupus erythematosus, ulcerative colitis and traumatic brain injuries (Leoni et al., PNAS, 99(5); 2995-3000 (2002); Suuronen et al. J. Neurochem. 87; 407-416 (2003) and Drug Discovery Today, 10: 197-204 (2005). Neoplastic Diseases In some aspects, the invention relates to methods of selectively inducing terminal differentiation, and arresting cell growth and/or apoptosis of neoplastic cells, thereby inhibiting proliferation of such cells. The compounds of the present invention are useful in treating, alleviating, and/or preventing cancer in a subject. The term “cancer” refers to any cancer caused by the proliferation of neoplastic cells, such as solid tumors, neoplasms, carcinomas, sarcomas, leukemias, lymphomas and the like. In particular, cancers that may be treated, alleviated and/or prevented by the compounds of the invention include, but are not limited to: cardiac cancer, lung cancer, gastrointestinal cancer, genitourinary tract cancer, liver cancer, nervous system cancer, gynecological cancer, hematologic cancer, skin cancer, and adrenal gland cancer. In some embodiments, the compounds of the invention relate to treating, alleviating, or preventing cardiac cancers selected from sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma. In some embodiments, the compounds of the invention relate to treating, alleviating, or preventing lung cancer selected from bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, and mesothelioma. In some embodiments, the compounds of the invention relate to treating, alleviating or preventing gastrointestinal cancer selected from esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), and large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma). In some embodiments, the compounds of the invention relate to treating, alleviating, and/or preventing genitourinary tract cancer selected from kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), and testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma). In some embodiments, the compounds of the invention relate to treating, alleviating, and/or preventing liver cancer selected from hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, and hemangioma. In some embodiments, the compounds of the invention relate to treating, alleviating, and/or preventing bone cancer selected from osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochondroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors. In some embodiments, the compounds of the invention relate to treating, alleviating, and/or preventing nervous system cancer selected from skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), and spinal cord (neurofibroma, meningioma, glioma, sarcoma). In some embodiments, the compounds of the invention relate to treating, alleviating, and/or preventing gynecological cancer selected from uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), and fallopian tubes (carcinoma). In some embodiments, the compounds of the invention relate to treating, alleviating, and/or preventing skin cancer selected from malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, and psoriasis. In some embodiments, the compounds of the invention relate to methods of treating, alleviating, and/or preventing adrenal gland cancer selected from neuroblastoma. In some embodiments, the instant compounds are useful in the treatment, alleviation, and/or preventing of cancers that include, but are not limited to: leukemias including acute leukemias and chronic leukemias such as acute lymphocytic leukemia (ALL), Acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML) and Hairy Cell Leukemia; lymphomas such as cutaneous T-cell lymphomas (CTCL), noncutaneous peripheral T-cell lymphomas, lymphomas associated with human T-cell lymphotropic virus (HTLV) such as adult T-cell leukemia/lymphoma (ATLL), Hodgkin's disease and non-Hodgkin's lymphomas, large-cell lymphomas, diffuse large B-cell lymphoma (DLBCL); Burkitt's lymphoma; mesothelioma, primary central nervous system (CNS) lymphoma; multiple myeloma; childhood solid tumors such as brain tumors, neuroblastoma, retinoblastoma, Wilm's tumor, bone tumors, and soft-tissue sarcomas, common solid tumors of adults such as head and neck cancers (e.g., oral, laryngeal and esophageal), genito urinary cancers (e.g., prostate, bladder, renal, uterine, ovarian, testicular, rectal and colon), lung cancer, breast cancer, pancreatic cancer, melanoma and other skin cancers, stomach cancer, brain tumors, liver cancer and thyroid cancer. Hematologic Diseases In some aspects, the invention relates to methods of treating, alleviating, or preventing hematological diseases. Hematologic diseases include abnormal growth of blood cells which can lead to dysplastic changes in blood cells and hematologic malignancies such as various leukemias. Examples of hematologic diseases include but are not limited to acute myeloid leukemia, acute promyelocytic leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia, the myelodysplastic syndromes, and sickle cell anemia. Acute myeloid leukemia (AML) is the most common type of acute leukemia that occurs in adults. Several inherited genetic disorders and immunodeficiency states are associated with an increased risk of AML. These include disorders with defects in DNA stability, leading to random chromosomal breakage, such as Bloom's syndrome, Fanconi's anemia, Li-Fraumeni kindreds, ataxia-telangiectasia, and X-linked agammaglobulinemia. Acute promyelocytic leukemia (APML) represents a distinct subgroup of AML. This subtype is characterized by promyelocytic blasts containing the 15; 17 chromosomal translocation. This translocation leads to the generation of the fusion transcript comprised of the retinoic acid receptor and a sequence PML. Acute lymphoblastic leukemia (ALL) is a heterogeneous disease with distinct clinical features displayed by various subtypes. Reoccurring cytogenetic abnormalities have been demonstrated in ALL. The most common cytogenetic abnormality is the 9;22 translocation. The resultant Philadelphia chromosome represents poor prognosis of the patient. Chronic myelogenous leukemia (CML) is a clonal myeloproliferative disorder of a pluripotent stem cell. CML is characterized by a specific chromosomal abnormality involving the translocation of chromosomes 9 and 22, creating the Philadelphia chromosome. Ionizing radiation is associated with the development of CML. The myelodysplastic syndromes (MDS) are heterogeneous clonal hematopoietic stem cell disorders grouped together because of the presence of dysplastic changes in one or more of the hematopoietic lineages including dysplastic changes in the myeloid, erythroid, and megakaryocytic series. These changes result in cytopenias in one or more of the three lineages. Patients afflicted with MDS typically develop complications related to anemia, neutropenia (infections), or thrombocytopenia (bleeding). Generally, from about 10% to about 70% of patients with MDS develop acute leukemia. Sickle cell disease is attributable to homozygous inheritance of a single amino acid substitution in the β-globin gene that leads to polymerization of deoxygenated hemoglobin, deformation of red blood cells, microvascular occlusion, hemolysis, and consequent disease manifestations, including pain, strokes, and pulmonary complications (Bunn H F, 1997, J. Med. 337:762-769). Abundant biochemical, epidemiological, and clinical evidence have shown that a high level of γ globin, the fetal form of β globin, inhibits the aberrant polymerization of sickle hemoglobin and ameliorates the disease phenotype. The only Food and Drug Administration (FDA)-approved drug for sickle cell disease, hydroxyurea, causes significant induction of fetal hemoglobin, decreased disease severity, and benefits overall mortality (Letvin et al., 1984, N Engl J Med 310:869-873; Platt O S, et al., 1984, J Clin Invest 74:652-656; Charache S, et al., 1995, N Engl J. Med 332: 317-1322; Steinberg M H, et al., 2003, JAMA 289:1645-1651). Nevertheless, hydroxyurea has bone marrow-suppressive effects and is ineffective in a significant portion of patients (Charache S, et al.; Steinberg M H, et al., 2003; Steinberg MH, 1999, N Engl J. Med 340:1021-1030). A drug that induces fetal hemoglobin more substantially with less myelosuppression would be expected to have greater therapeutic utility in sickle cell disease. Transcriptional regulation of the human globin gene locus has been investigated intensively. Gamma-globin gene expression is influenced by transcription factors (GATA-1, EKLF, NF-E4p22, Ikaros) and chromatin modifying enzymes [SWI/SNF complex, HATs, and histone deacetylase (HDACs)] as part of multiprotein complexes, and a unique, dynamic chromatin structure termed the β-globin active chromatin hub (βACH) (8-11). Polymorphisms in BCL11A, a transcriptional repressor, alter baseline fetal hemoglobin levels, and a multiprotein complex containing BCL11a binds to the β-globin locus, resulting in repression of γ-globin expression (Menzel S, et al., 2007, Nat Genet 39:1197-1199; Lettre G, et al., 2008, Proc Natl Acad Sci USA 105:11869-11874;Sankaran V G, et al., 2008, Science 322:1839-1842; Uda M, et al., 2008, Proc NATL Acad Sci USA 105:1620-1625; Sankaran V G, et al., 2009, Nature 460:1093-1097). Despite this granularity, discrete targets amenable to ligand discovery efforts have not been identified and functionally validated. The induction of fetal hemoglobin is a validated strategy to improve symptoms and complications of sickle cell disease. The development of targeted therapies has been limited by the absence of discrete draggable targets. Bradner et al., 2010, PNAS, 107:28, 12617-12622 has developed a unique bead-based strategy for the identification of inducers of fetal hemoglobin transcripts in primary human erythroid cells, which includes a small-molecule screen of bioactive compounds that have been identified to have remarkable class-associated activity among histone deacetylase (HDAC) inhibitors. Using a chemical genetic strategy combining focused libraries of biased chemical probes and reverse genetics by RNA interference, Bradner et al. identified HDAC1 and HDAC2 as molecular targets mediating fetal hemoglobin induction. Isoform-selective inhibitors of HDAC1 and HDAC2 are targets for the treatment of sickle cell disease. Pharmaceutical Compositions Accordingly, in another aspect of the present invention, pharmaceutical compositions are provided, wherein these compositions comprise any of the compounds as described herein, and optionally comprise a pharmaceutically acceptable carrier, adjuvant or vehicle. In certain embodiments, these compositions optionally further comprise one or more additional therapeutic agents. It will also be appreciated that certain of the compounds of present invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof. According to the present invention, a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof. As used herein, the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. A “pharmaceutically acceptable salt” means any non-toxic salt or salt of an ester of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof. As used herein, the term “inhibitorily active metabolite or residue thereof” means that a metabolite or residue thereof is also an inhibitor of an HD AC isoform as described herein. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1-4alkyl)4salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate. As described above, the pharmaceutically acceptable compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention. Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator. In yet another aspect, a method for treating a proliferative, inflammatory, or cardiovascular disorder is provided comprising administering an effective amount of a compound, or a pharmaceutical composition to a subject in need thereof. In certain embodiments of the present invention an “effective amount” of the compound or pharmaceutical composition is that amount effective for treating a proliferative, inflammatory, or cardiovascular disorder, or is that amount effective for treating cancer. In other embodiments, an “effective amount” of a compound is an amount which inhibits binding of PI3K and thereby blocks the resulting signaling cascades that lead to the abnormal activity of growth factors, receptor tyrosine kinases, protein serine/threonine kinases, G protein coupled receptors and phospholipid kinases and phosphatases. The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating the disease. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disorder, the particular agent, its mode of administration, and the like. The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disease being treated and the severity of the disease; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts. The term “patient”, as used herein, means an animal, preferably a mammal, and most preferably a human. The pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracistemally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use. In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues. Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. While one or more of the inventive compounds may be used in an application of monotherapy to treat a disorder, disease or symptom, they also may be used in combination therapy, in which the use of an inventive compound or composition (therapeutic agent) is combined with the use of one or more other therapeutic agents for treating the same and/or other types of disorders, symptoms and diseases. Combination therapy includes administration of the therapeutic agents concurrently or sequentially. Alternatively, the therapeutic agents can be combined into one composition which is administered to the patient. In one embodiment, the compounds of this invention are used in combination with other therapeutic agents. In other embodiments, a compound of the invention is administered in conjunction with a therapeutic agent selected from the group consisting of cytotoxic agents, radiotherapy, and immunotherapy. It is understood that other combinations may be undertaken while remaining within the scope of the invention. Those additional agents may be administered separately from a provided combination therapy, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a combination therapy, the two therapeutic agents may be submitted simultaneously, sequentially or within a period of time from one another normally within about one through twelve hours from one another. For example, one therapeutic agent can be administered within about one, two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve hours from the other therapeutic agent or agents used in the combination therapy. Combination therapy can be used for any of the therapeutic indications described herein. In one aspect, the invention provides a method, wherein the method is a combination therapy further comprising administering to the subject (1) a pharmaceutically active ingredient or exposing the subject to (2) cognitive behavioral therapy (CBT), (3) psychotherapy, (4) behavioral exposure treatments, (5) virtual reality exposure (VRE) or (6) cognitive remediation therapy or (7) any combination thereof. In one aspect, the invention provides a combination therapy for treating, alleviating, and/or preventing post-traumatic stress disorder (PTSD) or Alzheimer's disease in a subject comprising administering to the subject in need thereof an effective amount of (1) a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof and (2) a pharmaceutically active ingredient administered selected from Aricept®, memantine, and galantamine. In one aspect, the invention provides a method of treating extinction learning disorders in a subject in need thereof comprising administering to the subject an effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof. In one aspect, the extinction learning disorder is fear extinction deficit. In one aspect, the extinction learning disorder is post-traumatic stress disorder. In one aspect, the method is a combination therapy for treating extinction learning disorders in a subject in need thereof comprising administering to the subject (1) an effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof and (2) exposing the subject to cognitive behavioral therapy (CBT), psychotherapy, behavioral exposure treatments, virtual reality exposure (VRE) or cognitive remediation therapy. As used herein, the term “combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a combination of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Another aspect of the invention relates to inhibiting HDAC activity in a biological sample or a patient, which method comprises administering to the patient, or contacting said biological sample with a compound described herein, or a composition comprising said compound. The term “biological sample”, as used herein, generally includes in vivo, in vitro, and ex vivo materials, and also includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. Still another aspect of this invention is to provide a kit comprising separate containers in a single package, wherein the inventive pharmaceutical compounds, compositions and/or salts thereof are used in combination with pharmaceutically acceptable carriers to treat disorders, symptoms and diseases where HDAC plays a role. EXEMPLIFICATION As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein. General Information Spots were visualized by UV light (254 and 365 nm). Purification by column and flash chromatography was carried out using silica gel (200-300 mesh). Solvent systems are reported as the ratio of solvents. NMR spectra were recorded on a Bruker 400 (400 MHz) spectrometer.1H chemical shifts are reported in δ values in ppm with tetramethylsilane (TMS, =0.00 ppm) as the internal standard. LCMS spectra were obtained on an Agilent 1200 series 6110 or 6120 mass spectrometer with ESI (+) ionization mode. Example 1. Synthesis of Compound 100 Synthesis of 153-1. A solution of 153-0 (2.00 g, 8.4 mmol) and (NH4)2CO3(4.00 g, 42 mmol) in DMF (20 mL) was heated to 90° C. overnight. The mixture was cooled to room temperature and poured into water (100 mL). The precipitate was filtered off and washed with the mixture of diethyl ether (Et2O) and petroleum ether (PE; Et2O:PE=1:1) to give 153-1 (1.50 g, 76%) as a yellow solid. Synthesis of 153-2. To a mixture of 153-1 (1.40 g, 6.0 mmol), 4-fluorophenylboronic acid (920 mg, 7.7 mmol) and Cs2CO3(5.83 g, 18.0 mmol) in dioxane/H2O (30 mL/6 mL) was added Pd(PPh3)4(693 mg, 0.6 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 2 hours and then concentrated in vacuo. The residue was dissolved with ethyl acetate (EtOAc; 100 mL) and the solution was washed with brine (30 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=5:1˜3:1) to give 153-2 (1.10 g, 74%) as a yellow solid. Synthesis of 153-3. To a solution of 153-2 (260 mg, 1.0 mmol) and trimethylamine (TEA; 810 mg, 8.0 mmol) in dichloromethane (DCM; 15 mL) was added triphosgene (330 mg, 1.1 mmol) under ice bath. The solution was warmed to room temperature and continued to stir for 3 h. TEA (200 mg, 2.0 mmol) and SM0 (210 mg, 1.1 mmol) was then added. The resulting solution was heated at 50° C. for 2 hours. After the reaction was completed according to LCMS, the solution was diluted with DCM (15 mL) and the resulting solution was washed with brine (10 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜30:1) to give 153-3 (200 mg, 50%) as a yellow solid. Synthesis of 100. A mixture of 153-3 (200 mg, 0.5 mmol) and Pd/C (60 mg) in MeOH/THF (5 mL/5 mL) was stirred at room temperature for 2 hours under H2atmosphere. Pd/C was then removed by the filtration through the celite. The filtrate was concentrated and the residue was recrystallized with MTBE to give 100 (60 mg, 33%) as a yellow solid. Compounds 114, 116 and 119 were synthesized in a similar manner using the appropriately substituted amine variant of 100. Compound 114. 120 mg, 50%, a white solid. Compound 116. 160 mg, 54%, a white solid. Compound 119. 14 mg, 8%, a white solid. Example 2. Synthesis of Compound 101 Synthesis of 155-1. To a mixture of 155-0 (869 mg, 5.1 mmol) and phenylhydrazine (500 mg, 4.6 mmol) in AcOH/H2O (10 mL/2 mL) was added NaOAc·3H2O (1.40 g, 10.0 mmol). The reaction mixture was stirred at 130° C. for 30 min under microwave. The mixture was allowed to cool to room temperature and ice-water was added. The precipitate was collected by filtration and washed with the mixture of Et2O and PE (Et2O:PE=1:1) to give 155-1 (1.03 g, 96%) as a yellow solid. Synthesis of 155-1a. To a stirred solution of 155-1 (1.00 g, 4.3 mmol) in aq. HBr (48%, 5 mL) was added a solution of NaNO2(310 mg, 4.5 mmol) in H2O (3 mL) dropwise under ice bath. The solution was stirred at this temperature for 1.5 hours and a solution of CuBr (443 mg, 3.0 mmol) in aq. HBr (48%, 5 mL) was then added dropwise. The resulting mixture was stirred at 60° C. for another 1.5 hours. After the reaction was completed according to LCMS, the mixture was poured into water (40 mL) and the resultant was extracted with EtOAc (20 mL×3). The combined organic layer was washed with H2O (20 mL×3), dried over anhydrous Na2SO4and concentrated to give 155-1a (1.20 g, 94%) as a brown solid, which was used directly to next step without further purification. Synthesis of 155-2. To a solution of 155-1a (750 mg, 2.5 mmol), diphenylmethanimine (508 mg, 2.8 mmol), Xantphos (148 mg, 0.25 mmol) and Cs2CO3(2.50 g, 7.6 mmol) in dioxane (15 mL) was added Pd(OAc)2(115 mg, 0.51 mmol) under N2atmosphere in a seal tube. The mixture was heated at 130° C. for 3 hours under microwave. The resulting mixture was concentrated in vacuo. The residue was poured into H2O (50 mL) and the resultant was extracted with EtOAc (30 mL×3). The combined organic layer was washed with brine (30 mL×3), dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EA=10:1˜2:1) to give 155-2 (500 mg, 50%) as a yellow oil. Synthesis of 155-3. To a solution of 155-2 (150 mg, 0.38 mmol) in methanol (MeOH; 4 mL) was added aq. KOH (2 mL) dropwise under ice bath. The mixture was stirred at 60° C. for 4 hours and then poured into water (40 mL). The resultant was washed with EtOAc (20 mL) and the aqueous layer was adjusted to pH=5 with diluted HCl solution. The mixture was extracted with EtOAc (30 mL×3). The combined organic layer was washed with brine (30 mL×3), dried over anhydrous Na2SO4and concentrated in vacuo to give 155-3 (96 mg, 69%) as a yellow solid. Synthesis of 155-4. To a solution of 155-3 (560 mg, 1.5 mmol) in toluene (7 mL) was added TEA (0.64 mL, 4.6 mmol) and DPPA (840 mg, 3.0 mmol) successively. The resulting mixture was stirred at room temperature for 2 h. After the reaction was completed according to LCMS, the mixture was washed with water (7 mL) and dried over anhydrous Na2SO4. The resulting solution was concentrated to give 155-4 (510 mg, 85%) as a yellow solid, which was used directly to next step without further purification. Synthesis of 155-5. A solution of 155-4 (390 mg, 1.3 mmol) in toluene (7 mL) was heated at 80° C. for 4 hours. After cooling to room temperature, TEA (0.55 mL, 3.9 mmol) and SM0 (175 mg, 0.91 mmol) were added successively. The resulting mixture was stirred at 50° C. for 16 hours. After the reaction was completed according to LCMS, the mixture was diluted with EtOAc (30 mL). The resultant was washed with water (20 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=20:1) to give 155-5 (130 mg, 21%) as a yellow solid. Synthesis of 101. To a suspension of 155-5 (120 mg, 0.25 mmol) in EtOAc (5 mL) was added cone. HCl (0.1 mL). The mixture was stirred at room temperature for 5 min and then concentrated in vacuo. The residue was purified by Pre-HPLC to give 101 (30 mg, 38%) as a white solid. Example 3. Synthesis of Compound 102 Synthesis of 158-1. To a mixture of 155-0 (2.00 g, 11.8 mmol) and (4-fluorophenyl)hydrazine (1.35 g, 10.7 mmol) in AcOH/H2O (30 mL/6 mL) was added NaOAc·3H2O (3.03 g, 23.3 mmol). The reaction mixture was stirred at 130° C. for 30 minutes under microwave. After cooling to room temperature, ice-water was added. The precipitate was collected by filtration and washed with the mixture of Et2O and PE (Et2O:PE=1:1) to give 158-1 (2.00 g, 75%) as a yellow solid. Synthesis of 158-2. To a stirred solution of 158-1 (1.40 g, 5.6 mmol) in THF (20 mL) was added NaH (269 mg, 6.7 mmol) under ice bath. The solution was stirred at this temperature for 1 hour and a solution of benzyl chloroformate (CbzCl; 1.14 g, 6.7 mmol) in THF (8 mL) was then added dropwise. The resulting mixture was stirred at room temperature for another 2 h. After the reaction was completed according to LCMS, the mixture was diluted with water (40 mL). The resultant was extracted with EtOAc (30 mL×3). The combined organic layer was washed with brine (30 mL×3), dried over Na2SO4and concentrated in vacuo to give 158-2 as a crude product, which was used directly to next step without further purification. Synthesis of 158-3. A mixture of 158-2 (crude) and LiOH·H2O (470 mg, 11.2 mmol) in THF (30 mL) was stirred at 60° C. for 4 hours. The solvent was removed in vacuo. The residue was diluted with water (20 mL) and the resultant was washed with EtOAc (20 mL). The aqueous layer was adjusted to pH=5 with 2N HCl solution. The resulting mixture was extracted with EtOAc (30 mL×3). The combined organic layer was washed with brine (30 mL×3), dried over Na2SO4and concentrated to give 155-3 (0.90 g, 45% over two step) as a yellow solid. Synthesis of 158-4. A mixture of 158-3 (532 mg, 1.5 mmol) and TEA (454 mg, 4.5 mmol) in DCE (10 mL) was stirred at room temperature for 20 min. Diphenylphosphoryl azide (DPPA; 825 mg, 3 mmol) was then added dropwise. The mixture was stirred at 45° C. for 3 h. After cooling to room temperature, the resulting mixture was washed with water, dried over Na2SO4and concentrated (below 40° C.) to give 158-4 (600 mg) as a crude product, which was used directly for next step without further purification. Synthesis of 158-5. A solution of 158-4 (600 mg, 1.5 mmol) in toluene (10 mL) was stirred at 80° C. for 2 h. After cooling to room temperature, TEA (454 mg, 4.5 mmol) and SM0 (286 mg, 1.5 mmol) was added successively. The resulting mixture was stirred at 50° C. for 4 h. The solvent was removed and the residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜5:1) to give 158-5 (200 mg, 28%) as a yellow solid. Synthesis of 102. A mixture of 158-5 (200 mg, 0.42 mmol) and Pd/C (60 mg) in MeOH (15 mL) was stirred at room temperature overnight under H2atmosphere. Pd/C was then removed by the filtration through the celite. The filtrate was concentrated in vacuo and the residue was purified by Pre-HPLC to give (35 mg, 25%) as a yellow solid. Compound 103 was synthesized in a similar manner using the appropriately substituted amine variant of 102. Compound 103. 10 mg, 7%, a white solid. Example 4. Synthesis of Compound 104 Synthesis of 224-1. To a solution of 155-0 (20.0 g, 116.3 mmol) in pyridine (400 mL) was added ethyl carbonochloridate (15.1 g, 139.5 mmol) dropwise under ice bath. The reaction mixture was stirred at room temperature for 2 h. The solvent was removed in vacuo. The residue was dissolved in EtOAc (200 mL) and the resulting solution was washed with water (30 mL×5). The organic layer was dried over Na2SO4and concentrated to give 224-1 (20.0 g, 70%) as a yellow solid. Synthesis of 224-2. To a stirred solution of 224-1 (17.0 g, 69.7 mmol) in con·H2SO4(80 mL) was added con·HNO3(10 mL) under ice bath. The mixture was stirred 40° C. for 48 h. After cooling to room temperature, the mixture was poured into ice water (400 mL). The precipitate was collected by filtration and dried to give 224-2 (4.80 g, 24%) as a yellow solid. Synthesis of 224-3. A mixture of 224-2 (4.80 g, 16.7 mmol) and KOH (1.87 g, 33.4 mmol) in EtOH/H2O (50 mL/50 mL) was stirred at 95° C. for 2 h. The volatile solvent was removed in vacuo. The aqueous solution was washed with EtOAc (20 mL) and then adjusted to pH=5 with 2N HCl solution. The precipitate was collected by filtration and dried to give 224-3 (2.70 g, 75%) as a yellow solid. Synthesis of 224-4. To a mixture of 224-3 (1.50 g, 6.0 mmol), 4-fluorophenylboronic acid (1.01 g, 7.2 mmol) and Cs2CO3(3.91 g, 12.0 mmol) in dioxane/H2O (30 mL/6 mL) was added Pd(PPh3)4(693 mg, 0.6 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 2 hours and then concentrated in vacuo. The residue was dissolved in EtOAc (100 mL) and the resulting solution was washed with brine (30 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 224-4 (1.00 g, 72%) as a yellow solid. Synthesis of 224-5. To a solution of 224-4 (233 mg, 1.0 mmol) and TEA (810 mg, 8.0 mmol) in DCM (15 mL) was added triphosgene (330 mg, 1.1 mmol) under ice bath. The solution was warmed to room temperature and continued to stir for 3 h. TEA (200 mg, 2.0 mmol) and SM1 (122 mg, 1.1 mmol) was then added. The reaction mixture was heated at 50° C. for 2 h. After the reaction was completed according to LCMS, the solution was diluted with DCM (15 mL) and the resulting solution was washed with brine (10 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 224-5 (180 mg, 49%) as a yellow solid. Synthesis of 104. A mixture of 224-5 (180 mg, 0.5 mmol) and Pd/C (60 mg) in MeOH (5 mL) was stirred at room temperature for 2 hours under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was recrystallized with MTBE to give 104 (60 mg, 36%) as a yellow solid. Example 5. Synthesis of Compound 105 Synthesis of 323-1. To a mixture of 323-0 (1.00 g, 4.6 mmol), 4-fluorophenylboronic acid (773 mg, 5.5 mmol) and Cs2CO3(3.00 g, 9.2 mmol) in dioxane/H2O (20 mL/4 mL) was added Pd(PPh3)4(531 mg, 0.5 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 2 hours and then concentrated in vacuo. The residue was dissolved with EtOAc (60 mL) and the resulting solution was washed with brine (20 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 323-1 (1.00 g, 93%) as a yellow solid. Synthesis of 323-2. To a solution of 323-1 (300 mg, 1.3 mmol) and TEA (1.05 g, 10.4 mmol) in DCM (15 mL) was added triphosgene (425 mg, 1.4 mmol) under ice bath. The solution was warmed to room temperature and continued to stir for 3 h. TEA (263 mg, 2.6 mmol) and pyrrolidine (102 mg, 1.4 mmol) was then added. The reaction mixture was heated at 50° C. for 2 h. After the reaction was completed according to LCMS, the solution was diluted with DCM (15 mL) and the resulting solution was washed with brine (10 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 323-2 (130 mg, 30%) as a yellow solid. Synthesis of 105. A mixture of 323-2 (130 mg, 0.4 mmol) and Pd/C (50 mg) in MeOH (5 mL) was stirred at room temperature for 2 hours under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated in vacuo and the residue was purified by Pre-HPLC to give 105 (25 mg, 21%) as a yellow solid. Compound 108 was synthesized in a similar manner using the appropriately substituted amine variant of 105. Compound 108. 15 mg, 24%, a white solid. Example 6. Synthesis of Compound 106 Synthesis of 528-1. To a solution of 323-1 (100 mg, 0.4 mmol) in pyridine (3 mL) was added POCl3(91 mg, 0.6 mmol) dropwise under ice bath. The mixture was warmed to room temperature and stirred overnight. After the reaction was completed according to LCMS, the mixture was poured into ice water (10 mL). The precipitate was collected by filtration and dried to give 528-1 (60 mg, 40%) as a yellow solid. Synthesis of 106. A mixture of 528-1 (60 mg, 0.2 mmol) and Pd/C (20 mg) in MeOH (3 mL) was stirred at room temperature for 2 hours under H2atmosphere. Pd/C was removed by filtration through the celite. The filtrate was concentrated in vacuo and the residue was purified by Pre-HPLC to give 106 (13 mg, 24%) as a white solid. Example 7. Synthesis of Compound 107 Synthesis of 464-1. To a mixture of 464-0 (2.00 g, 11.6 mmol), 4-fluorophenylboronic acid (1.95 g, 13.9 mmol) and K2CO3(3.20 g, 23.2 mmol) in dioxane/H2O (40 mL/8 mL) was added Pd(PPh3)4(1.4 g, 1.2 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 2 hours and then concentrated in vacuo. The residue was dissolved with EtOAc (100 mL) and the resulting solution was washed with brine (30 mL×3). The organic layer was dried over Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 464-1 (1.70 g, 63%) as a yellow solid. Synthesis of 501-1. To a solution of 464-1 (90 mg, 0.4 mmol) in N,N-dimethylformamide (DMF; 3 mL) was added NaH (32 mg, 0.8 mmol) under ice bath. The solution was stirred for 1 hour at the same temperature and pyrrolidine-1-carbonyl chloride (67 mg, 0.5 mmol) was then added. The resulting solution was warmed to room temperature and continued to stir for 2 h. After the reaction was completed according to LCMS, the reaction mixture was poured into ice water (10 mL). The precipitate was collected by filtration and dried to give 501-1 (85 mg, 64%) as a yellow solid. Synthesis of 107. A mixture of 501-1 (85 mg, 0.3 mmol) and Pd/C (20 mg) in MeOH (5 mL) was stirred at room temperature for 2 hours under H2atmosphere. Pd/C was removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Pre-HPLC to give 107 (20 mg, 26%) as a white solid. Compounds 111 and 120 were synthesized in a similar manner using the appropriately substituted amine variant of 107. Compound 111. 20 mg, 28%, a white solid. Compound 120. 14 mg, 38%, a yellow solid. Example 8. Synthesis of Compound 109 Synthesis of 505-1. To a solution of 464-1 (300 mg, 1.3 mmol) in pyridine (5 mL) was added POCl3(297 mg, 2.0 mmol) dropwise under ice bath. The solution was warmed to room temperature and stirred overnight. After the reaction was completed according to LCMS, the mixture was poured into ice water (10 mL). The precipitate was collected by filtration and dried to give 505-1 (60 mg, 13%) as a yellow solid. Synthesis of 109. A mixture of 505-1 (60 mg, 0.2 mmol) and Pd/C (20 mg) in MeOH/THF (3 mL/3 mL) was stirred at room temperature for 2 hours under H2atmosphere. Pd/C was removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Pre-HPLC to give 109 (20 mg, 31%) as a white solid. Example 9. Synthesis of Compound 110 Synthesis of 553-1. To a mixture of 553-0 (2.00 g, 11.5 mmol), 4-fluorophenylboronic acid (1.93 mg, 13.8 mmol) and K2CO3(3.00 g, 9.2 mmol) in dioxane (40 mL) was added Pd(OAc)2(271 mg, 1.2 mmol) and 1,1′-Bis(di-tert-butylphosphino)ferrocene (D-t-BPF; 284 mg, 0.6 mmol) under N2atmosphere. The mixture was stirred at 100° C. for 2 hours and then concentrated in vacuo. The residue was dissolved with EtOAc (100 mL) and the resulting solution was washed with brine (30 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜30:1) to give 553-1 (500 mg, 19%) as a yellow solid. Synthesis of 553-2. To a solution of 553-1 (100 mg, 0.4 mmol) and TEA (323 mg, 3.2 mmol) in DCM (15 mL) was added triphosgene (149 mg, 0.5 mmol) under ice bath. The solution was warmed to room temperature and stirred for 3 h. TEA (81 mg, 0.8 mmol) and pyrrolidine (36 mg, 0.5 mmol) was then added. The resulting solution was heated at 50° C. for 2 h. After the reaction was completed, the mixture was diluted with DCM (15 mL) and the resulting solution was washed with brine (10 mL×3). The organic layer was dried over Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜10:1) to give 553-2 (60 mg, 45%) as a yellow solid. Synthesis of 110. A mixture of 553-2 (60 mg, 0.2 mmol) and Pd/C (20 mg) in MeOH (5 mL) was stirred at room temperature for 2 hours under H2atmosphere. Pd/C was removed by filtration through the celite. The filtrate was concentrated and the residue was recrystallized with MTBE to give 110 (40 mg, 73%) as a yellow solid. Compound 121 was synthesized in a similar manner using the appropriately substituted amine variant of 110. Compound 121. 24 mg, 43%, a yellow solid. Example 10. Synthesis of Compound 112 Synthesis of 728-1. To a solution of 728-0 (4.65 g, 35.7 mmol) in DML (30 mL) was added a solution of NBS (6.36 g, 35.7 mmol) in DML (20 mL) at 0° C. The resulting solution was stirred at the same temperature for 30 minutes, warmed to room temperature and continued to stir for 1 h. The solution was diluted with EtOAc (100 mL). The resulting solution was washed with brine (30 mL×3) and dried over anhydrous Na2SO4. The combined organic layer was concentrated in vacuo to give 728-1 (6.5 g, 93%) as a yellow solid. Synthesis of 728-2. A mixture of 728-1 (2.7 g, 11.5 mmol), 4-fluorobenzeneboronic acid (1.93 g, 13.8 mmol), Cs2CO3(11.2 g, 34.5 mmol), and Pd(PPh3)4in dioxane/H2O (45 mL/15 mL) was purged by N2and stirred at 95° C. for 3 h. The mixture was concentrated in vacuo. The residue was dissolved with EtOAc (50 mL) and the resulting solution was washed with brine (15 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=10:1˜4:1) give 728-2 (2.6 g, 90%) as a yellow solid. Synthesis of 728-3. A mixture of 728-2 (1.5 g, 6.0 mmol), (Boc)2O (2.87 g, 13.2 mmol), DMAP (732 mg, 6.0 mmol) in THF (50 mL) was stirred at 80° C. for 3 h. After the reaction was completed according to LCMS, the mixture was diluted with water (150 mL). The resulting mixture was extracted with EtOAc (50 mL×3). The combined organic layer was washed with brine, dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=20:1˜10:1) to give 728-3 (2.0 g, 74%) as a white solid. Synthesis of 728-4. A solution of 728-3 (2.0 g, 4.4 mmol) and Pd/C (200 mg) in MeOH (30 mL) was stirred at room temperature for 3 hours under H2atmosphere. Pd/C was removed by filtration through the celite. The filtrate was concentrated to give 728-4 (1.5 g, 81%) as a yellow solid. Synthesis of 728-5. To a solution of 728-4 (200 mg, 0.5 mmol), TEA (404 mg, 4.0 mmol) in DCM (10 mL) was added triphosgene (169 mg, 0.6 mmol) under ice bath. The solution was warmed to room temperature and continued to stir for 1 h. TEA (202 mg, 2.0 mmol) and pyrrolidine (50 mg, 0.70 mmol) was then added. The resulting solution was heated at 50° C. for 2 h. After cooling to room temperature, the mixture was diluted with DCM (10 mL). The resulting solution was washed with brine (10 mL×3), dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 728-5 (205 mg, 81%) as a gray solid. Synthesis of 112. A mixture of 728-5 (205 mg, 0.4 mmol) in HCl/EA (20 mL) was stirred at room temperature for 2 h. The solvent was removed under the reduced pressure. The residue was dissolved in water (3 mL) and adjusted to PH >7 by 1N NaOH solution. The resultant was extracted with EtOAc (5 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by Pre-HPLC to give 112 (90 mg, 71%) as a white solid. Compounds 113 and 117 were synthesized by a similar procedure using an appropriately phenyl-substituted derivative of 112. Compound 113. 130 mg, 45%, a white solid. Compound 117. 70 mg, 86%, a white solid. Example 11. Synthesis of Compound 115 Synthesis of 732-0. To a solution of SM2 (230 mg, 1.8 mmol) and a catalytic amount of DMF in THF (15 mL) was added (COCl)2(0.2 mL, 2.7 mmol) dropwise under ice bath. The resulting mixture was stirred at room temperature for 1 hour and then concentrated in vacuo to afford 732-0 (240 mg, 90%) as a yellow oil, which was used directly to next step without further purification. Synthesis of 732-1. To a solution of 153-2 (300 mg, 1.2 mmol) in DMF (12 mL) was added NaH (144 mg, 3.6 mmol) under ice bath. The mixture was warmed to room temperature and continued to stir for 30 min. After cooling to 0° C., a solution of 732-0 (265 mg, 1.8 mmol) in DMF (6 mL) was added dropwise. The resulting mixture was then warmed to room temperature and stirred for another 3 h. After the reaction was completed according to LCMS, the mixture was added to ice water (50 mL). The resultant was extracted with EtOAc (30 mL×3). The combined organic layer was washed with brine (20 mL×3), dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=10:1˜5:1) to give 732-1 (360 mg, 83%) as a yellow solid. Synthesis of 115. A mixture of 732-1 (360 mg, 1.0 mmol) and Pd/C (72 mg) in MeOH/THF (25 mL/5 mL) was stirred at room temperature for 2 hours under H2atmosphere. Pd/C was removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Pre-HPLC to give 115 (190 mg, 57%) as a white solid. Compound 118 was synthesized by a similar procedure using an appropriately phenyl-substituted derivative of 115. Compound 118. 72 mg, 52%, a white solid. Example 12. Synthesis of Compound 122 Synthesis of 156-1. To a solution of nitromethane (4.67 g, 77.0 mmol) in DMF (50 mL) was added DMF-DMA (6.72 g, 92.0 mmol). The mixture was stirred at 45° C. for 45 min. Then the mixture was cooled to room temperature and poured into EtOAc (100 mL). The resultant was washed with water (50 mL×3), dried over anhydrous Na2SO4and concentrated in vacuo to give 156-1 (3.50 g, 39%) as a yellow oil, which was used directly to next step without further purification. Synthesis of 156-2. A solution of NaNO2(7.24 g, 105 mmol) in H2O (15 ml) cooled to 0° C. was slowly added to a mixture of aniline (9.3 g, 100 mmol), H2O (100 ml) and cone. HCl (23 ml) with vigorous stirring. The prepared diazo solution was added drop by drop at 0-5° C. to a solution of ethyl 2-chloro-3-oxobutanoate (16.4 g, 100 mmol) and sodium acetate (12.3 g, 150 mmol) in 10 mL of ethanol containing minimal amount of water. The reaction mixture was continued to stir for 1 h. The obtained precipitate was filtered off, washed with water and dried in the open air to give 156-2 (11.3 g, 50%) as a yellow solid, which was used directly to next step without further purification. Synthesis of 156-3. To a stirred mixture of 156-1 (2.17 g, 19.0 mmol) and 156-2 (8.48 g, 37 mmol) in CHCl3(110 mL) was added TEA (3.79 g, 37 mmol). The resulting mixture was heated to reflux for 24 hours and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EA=10:1) to give 156-3 (3.7 g, 76%) as a brown solid. Synthesis of 156-4. To a solution of 156-3 (1.85 g, 7.1 mmol) in THF (33 mL) was added a solution of LiOH H2O (893 mg, 21 mmol) in water (11 mL) dropwise under ice bath. Then MeOH (11 mL) was added. The mixture was stirred at room temperature for 3 h. The volatile solvent was removed in vacuo and the aqueous layer was adjusted to pH=4 with 2N HCl solution. The resultant was extracted with EtOAc (30 mL×3). The combined organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜15:1) to give 156-4 (780 mg, 48%) as a brown solid. Synthesis of 156-5. To a solution of 156-4 (400 mg, 1.7 mmol) and TEA (0.45 mL, 3.3 mmol) in acetone (13 mL) was added ethyl carbonochloridate (370 mg, 3.4 mmol) dropwise under ice bath. The mixture was stirred at this temperature for 30 min. Then a solution of NaN3(212 mg, 3.3 mmol) in H2O (1 mL) was added dropwise and the reaction mixture was stirred at 0° C. for 2 h. After the reaction was completed according to LCMS, the solvent was removed in vacuo and the residue was diluted with DCM (30 mL). The resulting mixture was washed with water (20 mL). The organic layer was dried over Na2SO4and concentrated to give 156-5 (370 mg, 83%) as a brown solid, which was used directly to next step without further purification. Synthesis of 156-5a. A mixture of 156-5 (580 mg, 2.2 mmol), t-BuOH (20 mL) and dioxane (5 mL) was stirred at 90°cfor 2 h. Then the solvent was removed in vacuo to give 156-5a (530 mg, 77%) as a yellow solid. Synthesis of 156-5b. To a solution of 156-5a (680 mg, 2.2 mmol) in DCM (17 mL) was added TFA (3.3 mL, 45 mmol) dropwise under ice bath. The mixture was stirred at room temperature for 3 hours and the solvent was removed in vacuo. The residue was diluted with hexanes/EA/DCM (75 mL/15 mL/10 mL). The formed precipitate was filtered, washed with the above solvent mixture to give a yellow solid. The solid was diluted with DCM (20 mL) and the mixture was adjusted to PH=8 with TEA. The resultant was washed with water (20 mL). The organic layer was dried over anhydrous Na2SO4and concentrated to give 156-5b (410 mg, 90%) as a yellow solid. Synthesis of 156-6. To a solution of 156-5b (300 mg, 1.5 mmol) and TEA (1.65 mL, 12.0 mmol) in DCM (18 mL) was added triphosgene (480 mg, 1.6 mmol) successively under ice bath. The mixture was stirred at 35° C. for 3 h. After cooling to 0° C., a solution of SM-0 (312 mg, 1.6 mmol) and TEA (0.8 mL, 5.9 mmol) in DCM (6 mL) was added dropwise. The resulting mixture was heated to 50° C. and stirred for another 2 h. After cooling to room temperature, the mixture was diluted with DCM (10 mL). The resultant was washed with brine (10 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 156-6 (230 mg, 45%) as a yellow solid. Synthesis of 122 (T-156). A mixture of 156-6 (230 mg, 0.67 mmol) and Pd/C (45 mg) in MeOH (15 mL) was stirred at room temperature for 4 hours under H2atmosphere. Pd/C was removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Pre-HPLC to give 122 (T-156) (25 mg, 12%) as a white solid. Example 13. Synthesis of Compound 143. Synthesis of 143-A. To a solution of tert-butyl 5-oxohexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate (1.00 g, 4.7 mmol) in DCM (10 mL) was added TFA (10.8 g, 47.4 mmol) dropwise. Then the solution was stirred at room temperature for 1 h. The solution was concentrated in vacuo to give 1450-A (0.99 g, 94%) as a colorless oil. Synthesis of 143-B. A mixture of 6-chloro-3-nitropyridin-2-amine (10.0 g, 57.6 mmol), 4-fluorophenylboronic acid (8.87 g, 63.4 mmol) and Cs2CO3(37.56 g, 115.2 mmol) in dioxane/H2O (200 mL/20 mL) was added Pd(PPh3)4(2.44 g, 2.9 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 2 h and then concentrated in vacuo. The residue was dissolved with EtOAc (200 mL) and the solution was washed with brine (100 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=5:1˜3:1) to give 143-B (11.2 g, 83%) as a yellow solid Synthesis of 143-C. A stirred solution of 143-B (3.0 g, 13.0 mmol) in pyridine (60 mL) was added phenyl carbonochloridate (4.45 g, 28.5 mmol) dropwise. After the addition was completed, the mixture was hated to 50° C. for 4 h. The mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=8:1˜3:1) to give 143-C (5.2 g, 84%) as a yellow solid. Synthesis of 143-D. A mixture of 143-A (0.99 g, 4.2 mmol) and 143-C (0.9 g, 1.9 mmol) in acetonitrile (20 mL) was stirred at 50° C. for 30 min, then Na2CO3(1.82 g, 19.0 mmol) was added into above mixture and stirred at 50° C. for 2 h. The mixture was cooled to room temperature. Na2CO3was removed by filtered, the filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜10:1) to give 143-D (1.47 g, 91%) as a yellow solid. Synthesis of 143-E. To a mixture of 143-D (150 mg, 0.39 mmol) and azetidine hydrochloride (73 mg, 0.78 mmol) in DCE (5 mL) was added acetic acid (1 drop) and stirred at 50° C. for 2 h, then NaBH(OAc)3(165 mg, 0.78 mmol) was added into above mixture. Then the mixture was stirred at 50° C. for 2 h. When the mixture was cooled to room temperature. The mixture was diluted with water (15 mL) and extracted with DCM (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 143-E (80 mg, 48%) as a yellow solid. Synthesis of 143. A mixture of 143-E (80 mg, 0.19 mmol) and Pd/C (80 mg) in MeOH (5 mL) was stirred at room temperature for 30 min under H2atmosphere. Pd/C was removed by filtration through Celite. The filtrate was concentrated in vacuo and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 143 (40 mg, 53%) as a light yellow solid. Compounds 172, 179, 181, 237 and 238 was synthesized in a similar manner using the appropriately substituted amine variant of 143. Compound 172. 100 mg, 60%, a white solid. Compound 179. 60 mg, 43%, a white solid. Compound 181. 18 mg, 19%, a white solid. Compound 188. 40 mg, 41%, a white solid. Compound 237. 8 mg, 45%, a light yellow solid. Compound 248. 30 mg, 54%, a light yellow solid. Compound 189 was synthesized in a similar manner using furan-2-ylboronic acid and the appropriately substituted amine variant of 143. Compound 189. 27 mg, 19%, a yellow solid. Compound 191 was synthesized in a similar manner using phenylboronic acid and the appropriately substituted amine variant of 143. Compound 191. 80 mg, 67%, a yellow solid. Example 14. Synthesis of Compound 123 Synthesis of 123-A. To a solution of 4-nitro-1H-pyrazole-3-carboxylic acid (8.00 g, 50.1 mmol) in MeOH (160 mL) was added SOCl2(11.92 g, 100.2 mmol) dropwise under ice bath. Then the solution was stirred at room temperature overnight. The solution was concentrated in vacuo to give 123-A (8.70 g, 84%) as a white solid. Synthesis of 123-B. A mixture of 123-A (6.10 g, 29.4 mmol), 4-fluorophenylboronic acid (5.43 g, 38.8 mmol), pyridine (9.29 g, 117.6 mmol) and copper (II) acetate (8.03 g, 44.1 mmol) in DCM (120 mL) was stirred at room temperature overnight. The residue was diluted with DCM (100 mL) and the solution was washed with brine (40 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=50:1˜10:1) to give 123-B (4.80 g, 62%) as a yellow solid. Synthesis of 123-C. A mixture of 123-B (4.80 g, 18.1 mmol) and KOH (1.01 g, 18.1 mmol) in THF/H2O (40 mL/5 mL) was room temperature overnight. The solvent was removed in vacuo. The residue was dissolved with water (20 mL) and then adjusted to pH=3 with diluted HCl solution. The precipitate was collected by filtration and dried to give 123-B (4.00 g, 88%) as a white solid. Synthesis of 123-D. A mixture of 123-C (3.50 g, 13.9 mmol), DPPA (7.65 g, 27.8 mmol), TEA (7.02 g, 69.5 mmol) and t-BuOH (20.57 g, 278.0 mmol) in dioxane (70 mL) was heated to reflux for 4 h under N2atmosphere. The solvent was removed in vacuo. The residue was dissolved with EtOAc (100 mL) and the solution was washed with brine (40 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuon to give 123-D as a crude product. Synthesis of 123-E. To a solution of 123-D (crude product from last step) in DCM (14 mL) was added TFA (7 mL) dropwise under ice bath. Then the solution was stirred at room temperature 3 h. The solvent was removed in vacuo. The residue was dissolved with DCM (20 mL) and then adjusted to pH >10 by NaOH (1 N) solution. The mixture was extracted with DCM (50 mL×3). The combined organic layer was washed with brine (50 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜30:1) to give 123-E (2.30 g, 65% (two steps)) as a yellow solid. Synthesis of 123-F. To a solution of 123-E (222 mg, 1.0 mmol) and TEA (808 mg, 8.0 mmol) in DCM (8 mL) was added triphosgene (297 mg, 1.0 mmol) under ice bath. The solution was warmed to room temperature and continued to stir for 3 h. Then a solution of TEA (200 mg, 2.0 mmol) and SM5 (234 mg, 1.0 mmol) in DCM (4 mL) was added. The reaction mixture was heated at 50° C. for 2 h. After the reaction was completed according to LCMS, the solution was diluted with DCM (15 mL) and the resulting solution was washed with brine (10 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜10:1) to give 123-F (200 mg, 41%) as a yellow solid. Synthesis of 123. A mixture of 123-F (120 mg, 0.25 mmol) and Pd/C (30 mg) in EtOAc/MeOH (8 mL/2 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 123 (40 mg, 36%) as a white solid. Compounds 124, 125, 130, 131, 132, 133, 134, 135, 136, 137 and 140 were synthesized in a similar manner using the appropriately substituted amine variant of 123. Compound 124. 85 mg, 49%, a yellow solid. Compound 125. 53 mg, 32%, a white solid. Compound 130. 45 mg, 23%, a yellow solid. Compound 131. 18 mg, 10%, a yellow solid. Compound 132. 40 mg, 22%, a yellow solid. Compound 133. 70 mg, 33%, a white solid. Compound 134. 70 mg, 38%, a white solid. Compound 136. 50 mg, 31%, a yellow solid. Compound 137. 35 mg, 25%, a gray solid. Compound 140. 60 mg, 37%, a gray solid. Compounds 128, 129 and 142 were synthesized in a similar manner using phenylboronic acid and the appropriately substituted amine variant of 123. Compound 128. 56 mg, 50%, a white solid. Compound 129. 62 mg, 52%, a white solid. Compound 142. 48 mg, 23%, a white solid. Compounds 139 were synthesized in a similar manner using 4-(difluoromethoxy)phenyl boronic acid and the appropriately substituted amine variant of 123. Compound 139. 11 mg, 9%, a gray solid. Example 15. Synthesis of Compound 135 Synthesis of 135-A. To a solution of 123-E (300 mg, 1.35 mmol) in pyridine (6 mL) was added POCl3(1.04 g, 6.75 mmol) dropwise under ice bath. The mixture was warmed to room temperature and stirred 1 h. After the reaction was completed according to LCMS, the mixture was poured into ice water (10 mL). The precipitate was collected by filtration and dried to give 135-A (200 mg, 44%) as a yellow solid. Synthesis of 135. A mixture of 242-5 (200 mg, 0.6 mmol) and Pd/C (50 mg) in EtOAc (10 mL) was stirred at room temperature for 2 h under H2atmosphere. Pd/C was removed by filtration through Celite. The filtrate was concentrated in vacuo and the residue was purified by Pre-HPLC to give 135 (70 mg, 38%) as a white solid. Compounds 126 and 141 were synthesized in a similar manner using phenylboronic acid and the appropriately substituted acid variant of 135. Compound 126. 54 mg, 45%, a white solid. Compound 141. 47 mg, 17%, a yellow solid. Example 16. Synthesis of Compound 138 Synthesis of 138-A. To a solution of methoxymethyltriphenylphosphonium chloride (1.03 g, 3.0 mmol) in THF (15 mL) was added potassium hexamethyldisilane (1.8 mL, 1.7 M in toluene) dropwise at 0° C. The resulting mixture was stirred at 50° C. for 2 h. Then a solution of 143-D (576 mg, 1.50 mmol) in THF (5 mL) was added into above mixture dropwise. The mixture was stirred at 50° C. for 24 h. When the mixture was cooled to room temperature. The mixture was diluted with water (15 mL) and extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜20:1) to give 138-A (200 mg, 32%) as a yellow solid. Synthesis of 138-B. To a solution of 138-A (200 mg, 0.49 mmol) in THF (4 mL) was added 1 N HCl (2.5 mL) dropwise. Then the solution was stirred at room temperature for 16 h. The solution was concentrated in vacuo to give 138-B as a crude product used to next step directly. Synthesis of 138-C. To a mixture of 138-B (crude product from last step) and pyrrolidine (105 mg, 1.47 mmol) in DCE (5 mL) was added acetic acid (1 drop) and stirred at 50° C. for 1 h, then NaBH(OAc)3(312 mg, 1.47 mmol) was added into above mixture. Then the mixture was stirred at 50° C. for 2 h. When the mixture was cooled to room temperature. The mixture was diluted with water (15 mL) and extracted with DCM (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜10:1) to give 138-C (50 mg, 23% (two steps)) as a yellow solid. Synthesis of 138. A mixture of 138-C (50 mg, 0.11 mmol) and Pd/C (50 mg) in MeOH (4 mL) was stirred at room temperature for 30 min under H2atmosphere. Pd/C was removed by filtration through Celite. The filtrate was concentrated in vacuo and the residue was purified by Pre-TLC (DCM:MeOH=8:1) to give 138 (15 mg, 32%) as a light yellow solid. Example 17. Synthesis of Compound 146 Synthesis of 146-A. A mixture of 1-(bromomethyl)-4-iodobenzene (2.50 g, 8.4 mmol), dimethylamine hydrochloride (1.37 g, 16.8 mmol) and K2CO3(1.60 g, 33.6 mmol) in THF (50 mL) was stirred at room temperature for 12 h. The mixture was diluted with DCM (100 mL) and washed with brine (40 mL×3). The combined organics washed with brine (30 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=5:1˜2:1) to give 146-A (1.70 g, 77%) as a yellow oil. Synthesis of 146-B. To a mixture of Zn (628 mg, 9.6 mmol) in THF (3 mL) was added 1,2-dibromoethane (181 mg, 0.96 mmol) at room temperature under N2. The mixture was heated to 65° C. and stirred for 10 min. After cooling to room temperature, TMSCl (104 mg, 0.96 mmol) was added dropwise. The mixture was stirred for 30 min at room temperature, Zn powder turned dark and stickly. A solution of tert-butyl 3-iodoazetidine-1-carboxylate (1.70 g, 6.0 mmol) in THF (3 mL) was added dropwise during 1 h, but no obvious Zn powder consumption was observed. The mixture was heated to 65° C. for 10 min and the mixture turn hazy. After cooling to 25° C., the mixture was stirred for 1 h. The major Zn was consumed to give 146-B (used directly in the next step). Synthesis of 146-C. To a solution of 146-B was added Pd2(dba)3(34.5 mg, 0.06 mmol) and tri-2-furylphosphine (56 mg, 0.24 mmol), followed by 146-A (943 mg, 3.6 mmol) in THF (3 mL). The mixture was stirred at 65° C. for 16 h and then concentrated in vacuo. The residue was dissolved with DCM (50 mL) and the solution was washed with brine (20 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 146-C (200 mg, 20%) as a yellow solid. Synthesis of 146-D. To a solution of 146-C (200 mg, 0.69 mmol) in DCM (5 mL) was added TFA (2 mL) and stirred at room temperature for 1 h. when LCMS showed the reaction was finished. The solvent was removed in vacuo to give 146-D as a crude product and used to next step directly. Synthesis of 146-E. A mixture of 143-C (160 mg, 0.35 mmol) and 146-E (crude product from last step) in acetonitrile (5 mL) was stirred at 50° C. for 30 min. Then Na2CO3(370 mg, 2.5 mmol) was added into above mixture and stirred at 50° C. for 3 h. After the reaction was completed according to LCMS, the mixture was cooled to room temperature. The Na2CO3was removed by filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 146-E (50 mg, 35%) as a yellow solid. Synthesis of 146. A mixture of 146-E (50 mg, 0.11 mmol) and Ni (50 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Ni was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC to give 146 (12 mg, 26%) as a yellow solid. Synthesis of 170. A mixture of 146-E (40 mg, 0.09 mmol) and Pd/C (40 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 170 (12 mg, 48%) as a yellow solid. Example 18. Synthesis of Compound 147 Synthesis of 147-A. A solution of 6-chloro-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine (2.00 g, 12.98 mmol) and di-tert-butyl dicarbonate (4.24 g, 19.48 mmol) in DCM (40 mL) was added DIEA (3.34 g, 25.96 mmol) dropwise. The mixture was stirred at room temperature for 1 h. After the reaction was completed according to TLC. The mixture was diluted with DCM (20 mL) and washed with brine (20 mL×3). The combined organics washed with brine (20 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=2:1) to give 147-A (2.10 g, 66%) as a white solid. Synthesis of 147-B. A mixture of 147-A (600 mg, 2.4 mmol), azetidine hydrochloride (659 mg, 7.08 mmol), t-BuONa (680 mg, 7.08 mmol), Pd2(dba)3(216 mg, 0.24 mmol) and XPhos (225 mg, 0.47 mmol) in toluene (12 mL) was stirred at 80° C. for 12 h under Ar atmosphere. The mixture was diluted with EtOAc (30 mL) was added and the solution was washed with brine (40 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜10:1) to give 147-B (500 mg, 77%) as a yellow solid Synthesis of 147-C. A solution of 147-B (250 mg, 0.91 mmol) in DCM (5 mL) was added TFA (2 mL) and stirred at room temperature for 1 h. when LCMS showed the reaction was finished. The solvent was removed in vacuo to give 147-C as a crude product (used in the next step directly). Synthesis of 147-D. A mixture of 143-C (150 mg, 0.34 mmol) and 147-C (crude product from last step) in acetonitrile (5 mL) was stirred at 50° C. for 30 min. Then Na2CO3(230 mg, 2.40 mmol) was added into above mixture and stirred at 50° C. for 3 h. After the reaction was completed according to LCMS, the mixture was cooled to room temperature. The Na2CO3was removed by filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 147-D (80 mg, 58%) as a yellow solid. Synthesis of 147. A mixture of 147-D (80 mg, 0.18 mmol) and Pd/C (80 mg) in MeOH (3 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by pre-TLC (DCM:MeOH=15:1) to give 147 (30 mg, 41%) as a yellow solid Compounds 150, 157, 158, 159, 160, 254, 260 were synthesized in a similar manner using the appropriately substituted amine a variant of 147. Compound 150. 50 mg, 67%, a yellow solid. Compound 157. 15 mg, 16%, a yellow solid. Compound 158. 12 mg, 13%, a yellow solid. Compound 159. 80 mg, 57%, a yellow solid. Compound 160. 60 mg, 43%, a yellow solid. Compound 249. 20 mg, 25%, a white solid. Compound 254. 53 mg, 57%, a yellow solid. Example 19. Synthesis of Compound 148 Synthesis of 148-A. To a mixture of 147-A (600 mg, 2.36 mmol), SM-B (825 mg, 3.54 mmol) and K2CO3(980 mg, 7.08 mmol) in dioxane/H2O (15 mL/1.5 mL) was added Pd(PPh3)4(96 mg, 0.12 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 5 h and then concentrated in vacuo. The residue was dissolved with EtOAc (20 mL) and the solution was washed with brine (20 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=5:1˜2:1) to give 148-A (285 mg, 40%) as a yellow solid. Synthesis of 148-B. A solution of 148-A (280 mg, 0.88 mmol) in DCM (5 mL) was added TFA (2 mL) and stirred at room temperature for 1 h. when LCMS showed the reaction was finished. The solvent was removed in vacuo to give 148-B as a crude product (used in the next step directly). Synthesis of 148-C. A mixture of 143-C (208 mg, 0.44 mmol) and 148-A (crude product from last step) in acetonitrile (5 mL) was stirred at 50° C. for 30 min. Then Na2CO3(140 mg, 1.32 mmol) was added into above mixture and stirred at 50° C. for 3 h. After the reaction was completed according to LCMS, the mixture was cooled to room temperature. The Na2CO3was removed by filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 148-C (160 mg, 38%) as a yellow solid. Synthesis of 148. A mixture of 1422-4 (80 mg, 0.16 mmol) and Pd/C (80 mg) in MeOH (5 mL) was stirred at room temperature for 30 min under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=15:1) to give 148 (30 mg, 40%) as a white solid. Compounds 149, 240, 250, 251, 252, 253 and 255 were synthesized in a similar manner using the appropriately substituted amine variant of 148. Compound 149. 30 mg, 40%, a white solid. Compound 240. 14 mg, 29%, a white solid. Compound 250. 28 mg, 29%, a yellow solid. Compound 251. 43 mg, 31%, a yellow solid. Compound 252. 130 mg, 76%, a yellow solid. Compound 253. 40 mg, 43%, as a yellow solid. Compound 255. 10 mg, 32%, a white solid. Example 20. Synthesis of Compound 151 Synthesis of 151-A. To a mixture of 147-A (320 mg, 1.38 mmol), trimethylboroxine (522 mg, 4.14 mmol) and K2CO3(952 mg, 6.9 mmol) in dioxane (15 mL) was added Pd(dppf)2Cl2(57 mg, 0.07 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 24 h and then concentrated in vacuo. The residue was dissolved with EtOAc (20 mL) and the solution was washed with brine (20 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=5:1˜2:1) to give 151-A (160 mg, 50%) as a white solid. Synthesis of 151-B. A solution of 151-A (160 mg, 0.68 mmol) in DCM (5 mL) was added TFA (2 mL) and stirred at room temperature for 1 h. when LCMS showed the reaction was finished. The solvent was removed in vacuo to give 151-B as a crude product used in the next step directly. Synthesis of 151-C. A mixture of 143-C (215 mg, 0.45 mmol) and 151-B (crude product from last step) in DMSO (5 mL) was stirred at room temperature for 10 min. Then Na2CO3(382 mg, 3.6 mmol) was added into above mixture and stirred at room temperature for 2 h. After the reaction was completed according to LCMS, the mixture was diluted with water (30 mL) and extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM=MeOH=100:1˜50:1) 151-C (160 mg, 90%) as a yellow solid. Synthesis of 151. A mixture of 151-C (160 mg, 0.41 mmol) and Pd/C (160 mg) in MeOH (5 mL) was stirred at room temperature for 30 min under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 151 (103 mg, 69%) as a white solid. Example 21. Synthesis of Compound 153 Synthesis of 153-A. To a mixture of 4-chloropyridin-3-amine (30.0 g, 234.4 mmol) and TEA (47.3 g, 468.8 mmol) in THF (600 mL) was added methyl 2-chloro-2-oxoacetate (30.0 g, 246.2 mmol) dropwise at ice bath. The solution was stirred at room temperature for 2 h. The reaction mixture was diluted with water (100 mL) and then extracted with EtOAc (100 mL×3). The combined organics washed with brine (100 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=10:1˜5:1) to give 153-A (31.0 g, 62%) as a white solid. Synthesis of 153-B. A mixture of 153-A (15.0 g, 70.1 mmol) and Lawesson's reagent (19.8 g, 49.1 mmol) in toluene (300 mL) was heated to reflux overnight. The reaction mixture was diluted with water (100 mL) and then extracted with EtOAc (100 mL×3). The combined organics washed with brine (100 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=10:1˜3:1) to give 153-B (2.0 g, 15%) as a yellow solid. Synthesis of 153-C. A mixture of 153-B (2.0 g, 10.3 mmol) and PtO2(400 mg) in acetic acid (100 mL) was stirred at 70° C. overnight under H2atmosphere at 5 MPa. PtO2was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜20:1) to give 153-C (820 mg, 40%) as a white solid. Synthesis of 153-D. A mixture of 153-C (360 mg, 1.82 mmol) and formaldehyde solution (37% w/w, 0.7 mL) and acetic acid (2 drops) in MeOH (10 mL) was stirred at 40° C. for 1 h, then NaBH(OAc)3, (772 mg, 3.64 mmol) was added into above solution. The reaction mixture was stirred at 40° C. for 2 h. The solution was cooled to room temperature. The solution was diluted with water (10 mL), extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜20:1) to give 153-D (350 mg, 91%) as a white solid. Synthesis of 153-E. A mixture of 153-D (350 mg, 1.65 mmol) and LiOH·H2O (139 mg, 3.30 mmol) in MeOH/H2O (10 mL/4 mL) was stirred at room temperature overnight. After the reaction was completed according to LCMS, MeOH was removed in vacuo. The aqueous was adjusted to pH=6 with 1N HCl. Then the solution was concentrated to dryness to give 196-D as a white solid, which was used directly to next step without further purification. Synthesis of 154-F. To a solution of CDI (161 mg, 0.99 mmol) in DMF (5 mL) was added 154-E (0.83 mmol, crude product from last step) in portions and the solution was stirred at room temperature for 1 h to give solution A. At the same time, to a solution of 143-B (193 mg, 0.83 mmol) in DMF (5 mL) was added NaH (60% in mineral oil) (66 mg, 1.65 mmol) in portions and the mixture was stirred at room temperature for 1 h to give solution B. Then, the solution A was added into the solution B dropwise and the resulting mixture continue to stir at room temperature for 1 h. After the reaction was completed according to LCMS, the mixture was poured into water (10 mL). The precipitate was collected by filtered and concentrated to dryness to give 154-F (150 mg, 44%) as a yellow solid. Synthesis of 153. A mixture of 169-C (150 mg, 0.36 mmol) and Pd/C (150 mg) in EtOAc (5 mL) was stirred at room temperature for 30 min under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=8:1) to give 153 (100 mg, 70%) as a white solid. Compound 165 was synthesized in a similar manner using the appropriately substituted halogen variant of 153. Compound 165. 80 mg, 57%, a yellow solid. Compound 163 was synthesized in a similar manner using furan-2-ylboronic acid and the appropriately substituted halogen variant of 153. Compound 163. 35 mg, 38%, a white solid. Compound 164 was synthesized in a similar manner using pyridin-3-ylboronic acid and the appropriately substituted halogen variant of 153. Compound 164. 10 mg, 14%, a yellow solid. Example 22. Synthesis of Compound 155 Synthesis of 155-A. A mixture of potassium (bromomethyl)trifluoroborate (1.00 g, 4.98 mmol) and pyrrolidine (371 mg, 5.23 mmol) in THF (10 mL) was stirred at 80° C. for 4 h. The solvent was removed in vacuo. The residue was dissolved in acetone and the solution filtered to remove KCl. The filtrate was concentrated in vacuo, dissolved in a minimal amount of hot acetone (10 mL), and precipitated by the dropwise addition of Et2O (5 mL). Additional Et2O (150 mL) was added to facilitate filtering to give 155-A (750 mg, 98%) as a white solid. Synthesis of 155-B. A mixture of 155-A (750 mg, 4.90 mmol), SM-A (500 mg, 4.67 mmol), Cs2CO3(4.56 g, 14.0 mmol), Pd(OAc)2(52 mg, 0.23 mmol) and XPhos (224 mg, 0.47 mmol) in THF/H2O (20 mL/2 mL) was stirred 80° C. for 12 h under Ar. The mixture was cooled to room temperature and diluted with H2O (50 mL). The mixture was extracted with EtOAc (20 mL×3). The combined organics washed with brine (20 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=8:1˜3:1) to give 155-B (700 mg, 47%) as a yellow solid. Synthesis of 155-C. To a solution of 155-B (350 mg, 1.15 mmol) in DCM (8 mL) was added TFA (4 mL) and stirred at room temperature for 1 h. when LCMS showed the reaction was finished. The solvent was removed in vacuo to give 155-C as a crude product and used to next step directly. Synthesis of 155-D. A mixture of 143-C (200 mg, 0.42 mmol) and 155-C (crude product from last step) in acetonitrile (5 mL) was stirred at 50° C. for 30 min. Then Na2CO3(356 mg, 3.36 mmol) was added into above mixture and stirred at 50° C. for 3 h. After the reaction was completed according to LCMS, the mixture was cooled to room temperature. The Na2CO3was removed by filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 155-D (180 mg, 93%) as a yellow solid. Synthesis of 155. A mixture of 155-D (180 mg, 0.39 mmol) and Pd/C (180 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=8:1) to give 155 (125 mg, 74%) as a yellow solid Compound 144 was synthesized in a similar manner using thiophen-2-ylboronic acid variant of 155. Compound 144. 80 mg, 60%, a yellow solid. Example 23. Synthesis of Compound 156 Synthesis of 156-A. A mixture of 6-chloro-3-nitropyridin-2-amine (10.00 g, 57.6 mmol), thiophen-2-ylboronic acid (8.12 g, 63.4 mmol) and Cs2CO3(37.56 g, 115.2 mmol) in dioxane/H2O (200 mL/20 mL) was added Pd(PPh3)4(2.44 g, 2.88 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 2 h and then concentrated in vacuo. The residue was dissolved with EtOAc (200 mL) and the solution was washed with brine (100 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=5:1˜3:1) to give 156-A (10.0 g, 79%) as a yellow solid Synthesis of 156-B. A stirred solution of 156-A (1.30 g, 5.88 mmol) in pyridine (20 mL) was added phenyl carbonochloridate (2.29 g, 14.7 mmol) dropwise. After the addition was completed, the mixture was heated to 50° C. for 4 h. The mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=8:1˜3:1) to give 156-B (2.4 g, 89%) as a yellow solid Synthesis of 156-C. A mixture of 156-B (300 mg, 0.65 mmol) and 143-C (190 mg, 0.98 mmol) in DMSO (10 mL) was stirred at room temperature for 10 min, then Na2CO3(312 mg, 3.25 mmol) was added into above mixture and stirred at room temperature for 2 h. The mixture was diluted with water (30 mL) and extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 156-C (200 mg, 84%) as a yellow solid. Synthesis of 143. A mixture of 143-E (200 mg, 0.54 mmol) and Pd/C (200 mg) in MeOH (5 mL) was stirred at room temperature for 30 min under H2atmosphere. Pd/C was removed by filtration through Celite. The filtrate was concentrated in vacuo and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 156 (118 mg, 65%) as a light yellow solid. Compounds 173 and 187 were synthesized in a similar manner using 4-methoxyphenylboronic acid the appropriately substituted amine variant of 156. Compound 173. 6 mg, 7%, a yellow solid. Compound 187. 85 mg, 71%, a yellow solid. Compounds 186, 224, 229, 238, 259 and 260 were synthesized in a similar manner using 4-fluorophenylboronic acid the appropriately substituted amine variant of 156. Compound 186. 35 mg, 37%, a white solid. Compound 224. 40 mg, 26%, a yellow solid. Compound 225. 25 mg, 13%, a white solid. Compound 229. 12 mg, 43%, a white solid. Compound 238. 70 mg, 50%, a yellow solid Compound 259. 20 mg, 20%, a red solid. Compound 260. 50 mg, 54%, a yellow solid. Compounds 197 and 212 were synthesized in a similar manner using phenylboronic acid the appropriately substituted amine variant of 156. Compound 197. 16 mg, 43%, a yellow solid. Compound 212. 80 mg, 87%, a white solid. Compounds 214, 216, 218 and 221 were synthesized in a similar manner using pyridin-3-ylboronic acid and the appropriately substituted amine variant of 156. Compound 171. 15 mg, 25%, a yellow solid. Compound 214. 1 mg, 10%, a yellow solid. Compound 216. 30 mg, 41%, a yellow solid. Compound 217. 25 mg, 22%, a yellow solid. Compound 218. 30 mg, 33%, a yellow solid. Compound 220. 20 mg, 16%, a yellow solid. Compound 221. 165 mg, 66%, a white solid. Compounds 228, 230 and 232 were synthesized in a similar manner using 4-(difluoromethoxy)phenylboronic acid and the appropriately substituted amine variant of 156. Compound 228. 25 mg, 21%, a yellow solid. Compound 230. 20 mg, 71%, a white solid. Compound 232. 70 mg, 51%, a white solid. Compound 231 was synthesized in a similar manner using 1-methyl-1H-pyrazol-4-ylboronic acid and the appropriately substituted amine variant of 156. Compound 231. 35 mg, 28%, a yellow solid. Example 24. Synthesis of Compound 167 Synthesis of 167-A. A mixture of 6-chloro-3-nitropyridin-2-amine (6.00 g, 34.6 mmol), 1H-pyrazole (7.06 g, 103.76 mmol) and t-BuOK (11.64 g, 103.76 mmol) in dioxane (120 mL) was stirred at 120° C. under microwave for 1 h. The mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=2:1˜1:2) to give 167-A (4.0 g, 56%) as a yellow solid Synthesis of 167-B. A stirred solution of 167-A (2.00 g, 9.75 mmol) in pyridine (40 mL) was added phenyl carbonochloridate (3.36 g, 21.44 mmol) dropwise. After the addition was completed, the mixture was hated to 50° C. for 2 h. The mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=5:1˜2:1) to give 167-C (3.2 g, 74%) as a yellow solid. Synthesis of 167-D. A mixture of 167-C (258 mg, 0.58 mmol), 183-B (183 mg, 1.16 mmol) and Na2CO3(286 mg, 2.31 mmol) in acetonitrile was stirred at 50° C. for 3 h. The mixture was cooled to room temperature. The Na2CO3was removed by filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 167-D (160 mg, 71%) as a yellow solid. Synthesis of 167. A mixture of 167-D (160 mg, 0.41 mmol) and Pd/C (160 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by pre-TLC (DCM:MeOH=8:1) to give 167 (125 mg, 84%) as a white solid. Compounds 177, 184, 185 and 190 were synthesized in a similar manner using the appropriately substituted amine variant of 176. Compound 177. 75 mg, 65%, a yellow solid. Compound 184. 20 mg, 11%, a yellow solid. Compound 185. 30 mg, 33%, a white solid. Compound 190. 20 mg, 36%, a yellow solid. Example 25. Synthesis of Compound 168 Synthesis of 168-A. A mixture of potassium (bromomethyl)trifluoroborate (1.00 g, 4.98 mmol) and 1-methylpiperazine (524 mg, 5.23 mmol) in THF (10 mL) was stirred at 80° C. for 4 h. The solvent was removed in vacuo. The residue was dissolved in acetone and the solution filtered to remove KCl. The filtrate was concentrated in vacuo, dissolved in a minimal amount of hot acetone (10 mL), and precipitated by the dropwise addition of Et2O (5 mL). Additional Et2O (150 mL) was added to facilitate filtering to give 168-A (760 mg, 79%) as a white solid. Synthesis of 168-B. A mixture of 168-A (336 mg, 1.80 mmol), SM-A (500 mg, 1.70 mmol), Cs2CO3(1.60 g, 5.0 mmol), Pd(OAc)2(12 mg, 0.05 mmol) and XPhos (48 mg, 0.1 mmol) in THF/H2O (10 mL/1 mL) was stirred 80° C. for 12 h under Ar. The mixture was cooled to room temperature and diluted with H2O (50 mL). The mixture was extracted with DCM (20 mL×3). The combined organics washed with brine (20 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=8:1˜3:1) to give 168-B (320 mg, 60%) as a yellow solid. Synthesis of 168-C. To a solution of 168-B (200 mg, 0.62 mmol) in DCM (4 mL) was added TFA (2 mL) and stirred at room temperature for 1 h. when LCMS showed the reaction was finished. The solvent was removed in vacuo to give 168-C as a crude product and used to next step directly. Synthesis of 168-D. A mixture of 143-C (160 mg, 0.35 mmol) and 168-C (crude product from last step) in acetonitrile (5 mL) was stirred at 50° C. for 30 min. Then Na2CO3(370 mg, 2.5 mmol) was added into above mixture and stirred at 50° C. for 3 h. After the reaction was completed according to LCMS, the mixture was cooled to room temperature. The Na2CO3was removed by filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 168-D (130 mg, 75%) as a yellow solid. Synthesis of 168. A mixture of 168-D (130 mg, 0.26 mmol) and Pd/C (130 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=8:1) to give 168 (100 mg, 83%) as a yellow solid Compounds 154, 155, 162 and 178 were synthesized in a similar manner using the appropriately substituted amine variant of 168. Compound 154. 60 mg, 64%, a yellow solid Compound 155. 110 mg, 65%, a yellow solid Compound 162. 18 mg, 50%, a yellow solid Compound 178. 125 mg, 61%, a white solid. Compounds 144 and 145 were synthesized in a similar manner using thiophen-2-ylboronic acid and the appropriately substituted amine variant of 168. Compound 144. 80 mg, 60%, a yellow solid Compound 145. 50 mg, 15%, a yellow solid Compound 161 was synthesized in a similar manner using pyridin-3-ylboronic acid and the appropriately substituted amine variant of 168. Compound 161. 30 mg, 32%, a yellow solid. Compound 261 was synthesized in a similar manner using 4-fluoro-2-methylphenylboronic acid and the appropriately substituted amine variant of 168. Compound 261. 46 mg, 55%, a yellow solid Example 26. Synthesis of Compound 169 Synthesis of 169-A. A mixture of tert-butyl hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (750 mg, 3.54 mmol), 1-methylpiperidin-4-one (800 mg, 7.08 mmol) and acetic acid (2 drops) in DCE (15 mL) was stirred at 50° C. for 2 h. Then Sodium triacetoxyborohydride (1.50 g, 7.08 mmol) was added into above mixture and stirred at 50° C. for another 2 h. After the reaction was completed according to LCMS, the solvent was diluted with water (10 mL) and then extracted by DCM (10 mL×3). The combined organics washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 169-A (750 mg, 69%) as a yellow oil. Synthesis of 169-B. A solution of 169-A (400 mg, 1.29 mmol) in DCM (10 mL) was added TFA (5 mL) and stirred at room temperature for 1 h. when LCMS showed the reaction was finished. The solvent was removed in vacuo to give 169-B as a crude product and used to next step directly. Synthesis of 169-C. A mixture of 143-C (306 mg, 0.65 mmol) and 169-B (crude product from last step) in acetonitrile (6 mL) was stirred at 50° C. for 30 min. Then Na2CO3(624 mg, 6.50 mmol) was added into above mixture and stirred at 50° C. for 3 h. After the reaction was completed according to LCMS, the mixture was cooled to room temperature. The Na2CO3was removed by filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 169-C (230 mg, 76%) as a yellow solid. Synthesis of 169. A mixture of 169-C (230 mg, 0.49 mmol) and Pd/C (230 mg) in MeOH (10 mL) was stirred at room temperature for 30 min under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 169 (150 mg, 70%) as a white solid. Compounds 152, 182, 199, 201, 202, 203, 235, 236 and 256 were synthesized in a similar manner using the appropriately substituted aldehyde or ketone variant of 169. Compound 152. 50 mg, 36%, a light yellow solid. Compound 182. 70 mg, 38%, a red solid. Compound 199. 50 mg, 54%, a light yellow solid. Compound 201. 30 mg, 42%, as a yellow solid. Compound 202. 30 mg, 42%, a yellow solid. Compound 203. 30 mg, 18%, a yellow solid. Compound 235. 170 mg, 87%, a white solid. Compound 236. 70 mg, 50%, a white solid. Compound 256. 20 mg, 8%, a light yellow solid. Compounds 210, 211, 215, 222, 223, 242 and 262 were synthesized in a similar manner using the appropriately substituted amine variant of 169. Compound 210. 160 mg, 96%, a tan solid. Compound 211. 70 mg, 40%, a white solid Compound 215. 70 mg, 75%, a white solid. Compound 222. 30 mg, 42%, a yellow solid. Compound 223. 35 mg, 31%, a white solid. Compound 242. 50 mg, 34%, a white solid. Compound 262. 38 mg, 43%, a white solid. Example 27. Synthesis of Compound 183 Synthesis of 183-A. A solution of tert-butyl hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (2.00 g, 9.4 mmol), 1-bromo-2-fluoroethane (2.35 g, 18.8 mmol) and TEA (1.90 g, 18.8 mmol) in DCM (40 mL) was stirred at room temperature for 24 h. The mixture was diluted with DCM (40 mL) and washed with brine (20 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=20:1˜5:1) to give 183-A (1.50 g, 63%) as a colorless oil. Synthesis of 183-B. A solution of 183-A (200 mg, 0.77 mmol) in DCM (5 mL) was added TFA (2 mL) and stirred at room temperature for 1 h. when LCMS showed the reaction was finished. The solvent was removed in vacuo to give 183-B as a crude product and used to next step directly. Synthesis of 183-C. A mixture of 143-C (180 mg, 0.39 mmol), 183-B (0.77 mmol, a crude product from last step) and K2CO3(270 mg, 1.95 mmol) in acetonitrile was stirred at 50° C. for 3 h. The mixture was cooled to room temperature. The Na2CO3was removed by filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 183-C (130 mg, 85%) as a yellow solid. Synthesis of 183. A mixture of 183-c (130 mg, 0.31 mmol) and Pd/C (130 mg) in MeOH (10 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by pre-TLC (DCM:MeOH=10:1) to give 183 (90 mg, 75%) as a yellow solid Compounds 166 and 176 were synthesized in a similar manner using the appropriately substituted amine variant of 183. Compound 166. 12 mg, 28%, a white solid. Compound 176. 35 mg, 41%, a white solid. Compounds 174 and 175 were synthesized in a similar manner using furan-2-ylboronic acid and the appropriately substituted amine variant of 183 Compound 174. 65 mg, 50%, a yellow solid. Compound 175. 20 mg, 11%, a yellow solid. Example 28. Synthesis of Compound 192 Synthesis of 192-A. To a solution of azetidine-3-carboxylic acid (5.00 g, 49.5 mmol) in MeOH (100 mL) was added SOCl2(11.8 g, 99.0 mmol) dropwise at ice bath. The solution was stirred at room temperature overnight. The reaction mixture concentrated to dryness to give 192-A (6.8 g, 90%) as a white solid. Synthesis of 192-B. A mixture of 2,3-dichloropyridine (5.0 g, 31.6 mmol), 192-A (5.7 g, 37.9 mmol) and DIEA (12.2 g, 94.8 mmol) in DMSO (100 mL) was heated to 120° C. for 1 h. The reaction mixture was diluted with water (100 mL) and then extracted with EtOAc (100 mL×3). The combined organics washed with brine (100 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=10:1˜5:1) to give 192-B (1.0 g, 14%) as a yellow solid. Synthesis of 192-C. A mixture of 192-B (1.0 g, 4.42 mmol), TEA (1.3 g, 13.26 mmol) and Pd/C (1.0 g) in MeOH (40 mL) was stirred at 40° C. for 1 h under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was dissolved with DCM (50 mL), washed with brine (20 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo to give 192-C (770 mg, 91%) as a yellow solid. Synthesis of 192-D. A mixture of 192-C (700 mg, 3.65 mmol) and LiOH·H2O (460 mg, 10.95 mmol) in MeOH/H2O (10 mL/10 mL) was stirred at room temperature for 2 h. After the reaction was completed according to LCMS, the MeOH was removed in vacuo. The aqueous was adjusted to pH=6 with 1N HCl. Then the solution was concentrated to dryness to give 192-D as a white solid, which was used directly to next step without further purification. Synthesis of 192-E. To a solution of CDI (162 mg, 1.0 mmol) in DML (5 mL) was added 192-D (1.0 mmol, crude product from last step) in portions and the solution was stirred at room temperature for 1 h to give solution A. At the same time, to a solution of 143-B (233 mg, 1.0 mmol) in DML (5 mL) was added NaH (60% in mineral oil) (120 mg, 3.0 mmol) in portions and the mixture was stirred at room temperature for 1 h to give solution B. Then, the solution A was added into the solution B dropwise and the resulting mixture continue to stir at room temperature for 1 h. After the reaction was completed according to LCMS, the mixture was poured into water (10 mL). The precipitate was collected by filtered and concentrated to dryness to give 192-E (160 mg, 41%) as a yellow solid. Synthesis of 192. A mixture of 169-C (160 mg, 0.36 mmol) and Pd/C (160 mg) in MeOH (5 mL) was stirred at room temperature for 30 min under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 192 (60 mg, 39%) as a white solid. Compounds 193 and 195 were synthesized in a similar manner using the appropriately substituted amine variant of 192. Compound 193. 80 mg, 41%, a white solid. Compound 195. 60 mg, 43%, a gray solid. Example 29. Synthesis of Compound 196 Synthesis of 196-A. To a mixture of tert-butyl piperazine-1-carboxylate (200 mg, 1.1 mmol) and TEA (326 mg, 3.3 mmol) in DCM (20 mL) was added acetyl chloride (93 mg, 1.2 mmol) dropwise at ice bath. The reaction mixture was stirred at room temperature for 3 h. After the reaction was completed according to LCMS, the solution was diluted with DCM (15 mL) and the resulting solution was washed with brine (10 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜30:1) to give 196-A (150 mg, 60%) as a light yellow solid. Synthesis of 196-B. A solution of 196-A (150 mg, 0.66 mmol) in HCl/dioxane (2 N, 5 mL) was stirred at room temperature for 2 h. After the reaction was completed according to LCMS, the solution was concentrated to give 196-B as a crude product, used directly to next step without further purification. Synthesis of 196-C. A solution of 196-B (0.66 mmol, crude product from last step), methyl 2-bromoacetate (100 mg, 0.66 mmol) and DIEA (428 mg, 3.30 mmol) in MeCN (5 mL) stirred at room temperature for 3 h. After the reaction was completed according to LCMS, the mixture was diluted with water (5 mL) and extracted with EtOAc (5 mL×3). The combined organics washed with brine (50 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=50:1˜10:1) to give 196-C (120 mg, 91%) as a light yellow oil. Synthesis of 196-D. A solution of 196-C (120 mg, 0.60 mmol) and LiOH·H2O (101 mg, 2.40 mmol) in MeOH/H2O (10 mL/10 mL) was stirred at room temperature overnight. After the reaction was completed according to LCMS, the MeOH was removed in vacuo. The aqueous was adjusted to pH=6 by 1N HCl. Then the solution was concentrated to dryness to give 196-D as a crude product, which was used directly to next step without further purification. Synthesis of 196-E. To a solution of CDI (97 mg, 0.60 mmol) in DMF (2 mL) was added 196-D (0.60 mmol, crude product from last step) in portions and the solution was stirred at room temperature for 1 h to give solution A. At the same time, to a solution of 3-nitro-6-phenylpyridin-2-amine (129 mg, 0.60 mmol) in DMF (4 mL) was added NaH (60% in mineral oil) (48 mg, 1.2 mmol) in portions and the mixture was stirred at room temperature for 1 h to give solution B. Then, the solution A was added into the solution B in dropwise and the resulting mixture continue to stir at room temperature for 1 h. After the reaction was completed according to LCMS, the mixture was poured into water (10 mL). The precipitate was collected by filtered and concentrated to dryness to give 196-E (150 mg, 56%) as a yellow solid. Synthesis of 196. A mixture of 196-E (150 mg, 0.39 mmol) and Pd/C (150 mg) in EtOAc (10 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by the filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=8:1) to give 196 (120 mg, 87%) as a white solid. Compound 194 was synthesized in a similar manner using the appropriately substituted acid variant of 196. Compound 194. 16 mg, 43%, a yellow solid. Example 30. Synthesis of Compound 198 Synthesis of 198-A. A mixture of 6-bromo-3-nitropyridin-2-amine (5.0 g, 23.0 mmol), furan-2-ylboronic acid (3.1 g, 27.6 mmol) and Cs2CO3(22.5 g, 69.0 mmol) in dioxane/H2O (100 mL/10 mL) was added Pd(PPh3)4(2.44 g, 2.88 mmol) under N2atmosphere. The mixture was stirred at 95° C. for 3 h and then concentrated in vacuo. The residue was dissolved with EtOAc (200 mL) and the solution was washed with brine (100 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=10:1˜5:1) to give 198-A (2.0 g, 42%) as a yellow solid. Synthesis of 198-B. To a solution of CDI (395 mg, 2.44 mmol) in DMF (5 mL) was added 196-D (490 mg, 2.44 mmol) in portions and the solution was stirred at room temperature for 1 h to give solution A. At the same time, to a solution of 198-A (500 mg, 2.44 mmol) in DMF (5 mL) was added NaH (60% in mineral oil) (195 mg, 4.88 mmol) in portions and the mixture was stirred at room temperature for 1 h to give solution B. Then, the solution A was added into the solution B in dropwise and the resulting mixture continue to stir at room temperature for 1 h. After the reaction was completed according to LCMS, the solution was diluted with water (20 mL), extracted with EtOAc (10 mL×5). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=10:1˜2:1) to give 198-B (680 mg, 72%) as a yellow solid. Synthesis of 198-C. To a solution of 198-B (200 mg, 0.52 mmol) in DCM (5 mL) was added TFA (2 mL) dropwise under ice bath. Then the solution was stirred at room temperature 1 h. The solvent was removed in vacuo to give 198-C as a crude product. Synthesis of 198-D. A mixture of 198-C (0.52 mmol, crude product from last step), benzaldehyde (110 mg, 1.04 mmol) and acetic acid (2 drops) in MeOH (5 mL) was stirred at 40° C. for 1 h, then NaBH(OAc)3(221 g, 1.04 mmol) was added into above solution. The reaction mixture was stirred at 40° C. for 4 h. The solution was cooled to room temperature. The solution was diluted with water (10 mL), extracted with EtOAc (10 mL/3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=200:1˜50:1) to give 198-D (110 mg, 56% (two step)) as a yellow solid. Synthesis of 198. A mixture of 198-D (110 mg, 0.29 mmol), zinc powder (94 mg, 1.45 mmol) and ammonium formate (183 mg, 2.90 mmol) in MeOH (3 mL) was stirred at room temperature for 1 h. The zinc powder was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=15:1) to give 198 (20 mg, 20%) as a yellow solid. Compound 204, 239, 241, 263 and 264 was synthesized in a similar manner using 4-fluorophenylboronic acid and the appropriately substituted acid variant of 198. Compound 204. 60 mg, 73%, a white solid. Compound 239. 60 mg, 62%, a white solid. Compound 241. 69 mg, 58%, a white solid. Compound 263. 83 mg, 64%, a white solid. Compound 264. 50 mg, 79%, a white solid. Compound 205 was synthesized in a similar manner using 1H-pyrazole and the appropriately substituted acid variant of 205. Compound 205. 20 mg, 43%, a white solid. Example 31. Synthesis of Compound 206 Synthesis of 206-A. A mixture of ethyl 3-bromo-2-oxopropanoate (10.8 g, 55.3 mmol) and pyrazin-2-amine (5.0 g, 52.6 mmol) in DME (150 mL) was stirred at room temperature for 5 h. The precipitate was collected by filtered. Then the cake was dissolved in EtOH (100 mL) and stirred at 80° C. for 2 h. The solvent was removed in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=2:1˜1:2) to give 206-A (2.0 g, 20%) as a yellow solid. Synthesis of 206-B. A mixture of 206-A (1.50 g, 7.85 mmol), Pd/C (750 mg) and cone. HCl (15 mL) in EtOH (285 mL) was stirred at room temperature for 16 h under H2atmosphere at 40 psi. Pd/C was then removed by the filtration through the Celite. The filtrate was concentrated to give 206-B as a crude product, which was used directly to next step without further purification. Synthesis of 206-C. To a solution of 206-B (4.1 mmol, crude product from last step) and formaldehyde solution (37% w/w, 2 mL) and acetic acid (2 drops) in MeOH (20 mL) was stirred at 40° C. for 1 h, then NaBH(OAc)3(2.61 g, 12.3 mmol) was added into above solution. The reaction mixture was stirred at 40° C. for 2 h. The solution was cooled to room temperature. The solution was diluted with water (10 mL), extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜30:1) to give 206-C (500 mg, 58%) as a yellow solid. Synthesis of 206-D. A solution of 196-C (500 mg, 2.4 mmol) and LiOH·H2O (400 mg, 9.6 mmol) in MeOH/H2O (10 mL/10 mL) was stirred at room temperature overnight. After the reaction was completed according to LCMS, the MeOH was removed in vacuo. The aqueous was adjusted to pH=6 by 1N HCl. Then the solution was concentrated to dryness to give 206-D as a crude product, which was used directly to next step without further purification. Synthesis of 206-E. To a solution of 206-D (0.50 mmol, crude product from last step) and DMF (1 drop) in DCM (5 mL) was added (COCl)2(127 mg, 1.0 mmol) at ice bath. The resulting mixture was stirred at room temperature for 1 h and concentrated to give a white solid A. At the same time, to a solution of 3-nitro-6-phenylpyridin-2-amine (100 mg, 0.50 mmol) in DMF (5 mL) was added NaH (60% in mineral oil) (40 mg, 1.0 mmol) in portions and the mixture was stirred at room temperature for 1 h to give solution B. Then the solid A was added to solution B and the resulting mixture continue to stir at room temperature for 1 h. After the reaction was completed according to LCMS, the mixture was poured into water (20 mL). The precipitate was collected by filtered and concentrated to dryness to give 206-E (80 mg, 42%) as a yellow solid. Synthesis of 206. A mixture of 206-E (80 mg, 0.21 mmol) and Pd/C (80 mg) in MeOH (3 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified Pre-TLC (DCM:MeOH=10:1) to give 206 (25 mg, 34%) as a white solid. Compound 208 was synthesized in a similar manner using the appropriately substituted acid variant of 206. Compound 208. 28 mg, 38%, a light yellow solid. Compound 219 was synthesized in a similar manner using pyridin-3-ylboronic acid and the appropriately substituted acid variant of 206. Compound 219. 10 mg, 8%, a yellow solid. Compound 226 was synthesized in a similar manner using 4-fluorophenylboronic acid and the appropriately substituted acid variant of 206. Compound 226. 60 mg, 44%, a yellow solid. Example 32. Synthesis of Compound 243 Synthesis of 243-A. A mixture of tert-butyl 3-bromo-5H-pyrrolo[3,4-b]pyridine-6(7H)-carboxylate (820 mg, 2.75 mmol), potassium acetate (540 mg, 5.5 mmol), dppf (111 mg, 0.08 mmol) and palladium acetate (8.5 mg, 0.03 mmol) in ethanol (20 mL) was stirred at 100° C. for 12 h under CO atmosphere at 1.5 MPa. The reaction mixture was cooled to room temperature and filtered through Celite. The filtrate was concentrated in vacuo and the residue was dissolved with DCM (50 mL), washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=8:1˜3:1) to give 243-A (670 mg, 84%) as a white solid. Synthesis of 243-B. A mixture of 243-A (500 mg, 1.7 mmol) and NaBH4(390 mg, 10.2 mmol) in ethanol (50 mL) was stirred at room temperature for 12 h. The reaction mixture was concentrated in vacuo and the residue was dissolved with DCM (50 mL), washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 243-B (350 mg, 81%) as a white solid. Synthesis of 243-C. To a mixture of 243-B (150 mg, 0.6 mmol) in THF (5 mL) was added NaH (60% in mineral oil) (96 mg, 2.4 mmol) at room temperature. The resulting mixture was stirred at room temperature for 30 min. Then Mel (170 mg, 1.2 mmol) was added into above mixture dropwise. The resulting mixture was stirred at room temperature for 30 min. The solution was diluted with water (10 mL), extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=8:1˜3:1) to give 243-C (60 mg, 38%) as a yellow solid Synthesis of 243-D. To a solution of 243-C (60 mg, 0.23 mmol) in DCM (4 mL) was added TFA (2 mL) dropwise at ice bath. Then the solution was stirred at room temperature 1 h. The solvent was removed in vacuo to give 243-d as a crude product. Synthesis of 243-E. A mixture of 243-D (0.23 mmol, crude product from last step), 143-C (80 mg, 0.17 mmol) and Na2CO3(122 mg, 1.15 mmol) in acetonitrile was stirred at 50° C. for 3 h. After the reaction was completed according to LCMS. Na2CO3was removed by filtration, the filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 243-E (70 mg, 73%) as a yellow solid. Synthesis of 243. A mixture of 243-E (70 mg, 0.16 mmol) and Pd/C (70 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by pre-TLC (DCM:MeOH=15:1) to give 243 (48 mg, 78%) as a yellow solid Example 33. Synthesis of Compound 224 and 225 Synthesis of 244-A. To a solution of tert-butyl 4,6-dihydropyrrolo[3,4-c]pyrazole-5(2H)-carboxylate (1.40 g, 6.70 mmol) in DCM (10 mL) was added TFA (3 mL) dropwise under ice bath. Then the solution was stirred at room temperature 1 h. The solvent was removed in vacuo to give 244-A as a crude product. Synthesis of 244-B. A mixture of 244-A (crude product from last step) and 143-C (1.58 g, 3.35 mmol) in acetonitrile (30 mL) was stirred at 50° C. for 30 min, then Na2CO3(3.55 g, 33.5 mmol) was added into above mixture and stirred at 50° C. for 1 h. The mixture was cooled to room temperature. Na2CO3was removed by filtered, the filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 244-B (0.98 g, 78%) as a yellow solid. Synthesis of 244-C and 245-C. To a solution of 244-B (250 mg, 0.68 mmol) in DMF (5 mL) was added NaH (60% in mineral oil)(54 mg, 1.36 mmol) under ice bath and stirred at room temperature for 30 min. Then SM-A (204 mg, 0.82 mmol) was added into above mixture and stirred at for room temperature for 2 h. The mixture was quenched with water (15 mL), extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜30:1) to give 244-C and 245-C (300 mg, 82%) as a yellow solid. Synthesis of 244-D and 245-D. To a solution of 244-C and 245-C (300 mg, 0.56 mmol) in DCM (3 mL) was added TFA (1 mL) dropwise under ice bath. Then the solution was stirred at room temperature 1 h. The solvent was removed in vacuo to give 244-D and 245-D as a crude product. Synthesis of 244-E and 245-E. A mixture of 244-D and 245-D (crude product from last step), formaldehyde solution (37% w/w, 0.6 mL) and acetic acid (2 drops) in MeOH (6 mL) was stirred at 40° C. for 1 h, then NaBH(OAc)3(1.80 g, 8.48 mmol) was added into above solution. The reaction mixture was stirred at 40° C. for 16 h. The solution was cooled to room temperature. The solution was diluted with water (10 mL), extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=200:1) to give 244-E (30 mg, 12% (two step)) as a yellow solid and 245-E (90 mg, 36% (two step)) as a yellow solid. Synthesis of 244. A mixture of 244-E (30 mg, 0.07 mmol) and Pd/C (30 mg) in MeOH (2 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 244 (15 mg, 52%) as a yellow solid. Synthesis of 245. A mixture of 245-E (90 mg, 0.20 mmol) and Pd/C (90 mg) in MeOH (6 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=10:1) to give 245 (50 mg, 59%) as a yellow solid. Compounds 246 and 247 were synthesized in a similar manner using the appropriately substituted halogen variant of 244. Compound 246. 20 mg, 54%, a yellow solid. Compound 247. 6 mg, 43%, a yellow solid. Example 34. Synthesis of Compound 257 and 258 Synthesis of 257-A and 258-A. A mixture of tert-butyl 4,6-dihydropyrrolo[3,4-c]pyrazole-5(2H)-carboxylate (314 mg, 1.50 mmol), Cs2CO3(978 mg, 3.00 mmol) and iodomethane (320 mg, 2.25 mmol) in DMF (6 mL) was stirred at room temperature for 16 h. The mixture was diluted with water (18 mL), extracted with EtOAc (10 mL×3). The combined organic layer was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 257-A and 258-A (270 mg, 81%) as a yellow solid. Synthesis of 257-B and 258-B. To a solution of 257-A and 258-A (270 mg, 1.21 mmol) in DCM (6 mL) was added TFA (2 mL) dropwise under ice bath. Then the solution was stirred at room temperature 1 h. The solvent was removed in vacuo to give 257-B and 258-B as a crude product. Synthesis of 257-C and 258-C. A mixture of 257-B and 258-B (crude product from last step) and 143-C (286 mg, 0.61 mmol) in acetonitrile (10 mL) was stirred at 50° C. for 30 min, then Na2CO3(581 mg, 6.05 mmol) was added into above mixture and stirred at 50° C. for 1 h. The mixture was cooled to room temperature. Na2CO3was removed by filtered, the filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=200:1) to give 257-C (100 mg, 44%) as a yellow solid and 258-C (50 mg, 22%) as a yellow solid. Synthesis of 257. A mixture of 257-C (100 mg, 0.26 mmol) and Pd/C (100 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=15:1) to give 257 (50 mg, 55%) as a yellow solid. Synthesis of 258. A mixture of 258-C (50 mg, 0.13 mmol) and Pd/C (50 mg) in MeOH (3 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=15:1) to give 258 (25 mg, 55%) as a yellow solid. Example 35. Synthesis of Compound 271 Synthesis of 127-A. A mixture of tert-butyl 3-bromo-5H-pyrrolo[3,4-b]pyridine-6(7H)-carboxylate (820 mg, 2.75 mmol), potassium acetate (540 mg, 5.5 mmol), dppf (111 mg, 0.08 mmol) and palladium acetate (8.5 mg, 0.03 mmol) in ethanol (20 mL) was stirred at 100° C. for 12 h under CO atmosphere at 1.5 MPa. The reaction mixture was cooled to room temperature and filtered through Celite. The filtrate was concentrated in vacuo and the residue was dissolved with DCM (50 mL), washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=8:1˜3:1) to give 127-A (670 mg, 84%) as a white solid. Synthesis of 127-B. A mixture of 127-A (500 mg, 1.7 mmol) and NaBH4(390 mg, 10.2 mmol) in ethanol (50 mL) was stirred at room temperature for 12 h. The reaction mixture was concentrated in vacuo and the residue was dissolved with DCM (50 mL), washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜20:1) to give 127-B (350 mg, 81%) as a white solid. Synthesis of 127-C. To a mixture of 127-B (150 mg, 0.6 mmol) and TEA (121 mg, 1.2 mmol) in DCM (10 mL) was added MsCl (104 mg, 0.9 mmol) dropwise and the mixture was stirred at room temperature for 1 h. The solution was diluted with DCM (20 mL) and the solution was washed with brine (10 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo to give 127-C as crude product used to next step directly. Synthesis of 127-D. A mixture of 127-C (crude product from last step), 3-fluoroazetidine (59 mg, 0.78 mmol), K2CO3(166 mg, 1.2 mmol) in acetonitrile (5 mL) was heated to 40° C. for 12 h. The solvent was removed in vacuo. The residue was dissolved with DCM (100 mL) and the solution was washed with brine (30 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo to give 127-D (150 mg, 81%) as a yellow solid. Synthesis of 127-E. To a solution of 127-D (150 mg, 0.49 mmol) in DCM (4 mL) was added TFA (2 mL) dropwise at ice bath. Then the solution was stirred at room temperature 1 h. The solvent was removed in vacuo to give 271-E as a crude product used to next step directly. Synthesis of 127-F. A mixture of 127-E (crude product from last step), 143-C (118 mg, 0.25 mmol) and Na2CO3(212 mg, 2.0 mmol) in acetonitrile was stirred at 50° C. for 3 h. After the reaction was completed according to LCMS. Na2CO3was removed by filtration, the filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜50:1) to give 127-F (100 mg, 86%) as a yellow solid. Synthesis of 127. A mixture of 127-F (100 mg, 0.14 mmol) and Ni (100 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Ni was then removed by filtration through the Celite. The filtrate was concentrated and the residue was purified by Pre-TLC (DCM:MeOH=15:1) to give 271 (16 mg, 17%) as a yellow solid. Example 36. Synthesis of Compound 351 Synthesis of 351-A. To a solution of 4-nitro-1H-pyrazole-3-carboxylic acid (8.00 g, 50.1 mmol) in MeOH (160 mL) cooled in an ice bath was added SOCl2(11.92 g, 100.2 mmol) dropwise. The reaction mixture was slowly allowed to warm to room temperature and was stirred overnight. The MeOH and SOCl2were then distilled off and the residue was quenched with ice-water (50 mL). The product was extracted with DCM (100 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was triturated with diethyl ether (100 mL) to give 351-A (8.70 g, 84%) as an off white solid. Synthesis of 351-B. A mixture of 351-A (6.10 g, 29.4 mmol), 4-fluorophenylboronic acid (5.43 g, 38.8 mmol), pyridine (9.29 g, 117.6 mmol) and copper (II) acetate (8.03 g, 44.1 mmol) in DCM (120 mL) was stirred at room temperature under air for 48 h. The residue was diluted with DCM (100 mL) and the solution was washed with brine (40 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=50:1˜10:1) to give 351-B (4.80 g, 62%) as an off white solid. Synthesis of 351-C. A mixture of 351-B (4.80 g, 18.1 mmol) and KOH (1.01 g, 18.1 mmol) in THF/H2O (35 mL/15 mL) was stirred at room temperature overnight. The THF was removed in vacuo. The aqueous layer adjusted to pH=3 with diluted HCl solution. The precipitate was collected by filtration and dried to give 351-C (4.00 g, 88%) as a white solid. Synthesis of 351-D. A mixture of 351-C (3.50 g, 15.0 mmol), DPPA (7.65 g, 27.8 mmol) and TEA (7.02 g, 69.5 mmol) in t-BuOH (50 mL) was heated to reflux overnight under N2atmosphere. The solvent was removed in vacuo. The residue was dissolved with EtOAc (100 mL) and the solution was washed with brine (40 mL×3). The organic layer was dried over anhydrous Na2SO4and then concentrated in vacuon. The residue was purified by column chromatography on silica gel (PE:EtOAc=70:30˜50:50) to give 351-D (2 g, 44%) as a yellow solid. Synthesis of 351-E. To a solution of 351-D (2 g, 6.58 mmol) in DCM (14 mL) was added TFA (7 mL) drop wise under ice bath. Then the solution was stirred at room temperature 3 h. The solvent was removed in vacuo. The residue was dissolved with DCM (20 mL) and then adjusted to pH >10 by NaOH (1 N) solution. The mixture was extracted with DCM (50 mL×3). The combined organic layer was washed with brine (50 mL×3), dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1˜30:1) to give 351-E (1.2 g, 80%) as a yellow solid. Synthesis of 351-F. To a cooled (0° C.) suspension of NaH (118 mg, 50% in mineral oil, 2.45 mmol) in DMF (2 mL) was added 351-E (200 mg, 0.98 mmol) and stirred for 10 minutes. Diphenyl carbonate (385 mg, 1.96 mmol) was added to the mixture and stirred with slow warming to room temperature for 1 h. AM351 (312 mg, 1.47 mmol) was added at 0° C. and stirred with slow warming to room temperature for 1 h. The reaction mixture was quenched with cold water (10 mL) and extracted with DCM (20 mL×3). The combined organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1-25:1) to give 351-F (150 mg, 38%) as a brown solid. Synthesis of 351. A mixture of 351-F (150 mg, 0.37 mmol) and Pd/C (35 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Prep-HPLC to give 351 (45 mg, 32%) as an off white solid. Compounds 352, 353, 360, 361, 382, 383 and 384 were synthesized in a similar manner using the appropriately substituted amine variant of 351. Compound 352. 20 mg, 28%, an off white solid. Compound 353.17 mg, 20%, an off white solid. Compound 360. 31 mg, 34%, a brownish solid. Compound 361. 35 mg, 37%, a white solid. Compound 382. 15 mg, 14%, an off white solid. Compound 383. 12 mg, 13%, an off white solid. Compound 384. 25 mg, 21%, an off white solid. Example 37. Synthesis of Compound 364 Synthesis of 364-A. To a degassed mixture of 2-Amino-6-chloro-3-nitro pyridine (5 g, 28.80 mmol), 4-fluorophenyl boronic acid (8.06 g, 57.61 mmol) and Cs2CO3(23.89 g, 72.02 mmol) in 1,4-dioxane (50 mL) and water (15 mL) was added Pd(PPh3)2Cl2(0.5 g, 0.72 mmol) and the mixture was stirred at 100° C. overnight. The reaction mixture was concentrated in vacuum. The residue was dissolved with ethyl acetate (100 mL) and the solution was washed with brine (50 mL×3). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=75:25˜70:30) to give 364-A (4.8 g, 71.53%) as a yellow sold. Synthesis of 364-B. To a cooled (0° C.) suspension of NaH (82 mg, 50% in mineral oil, 1.71 mmol) in DMF (2 mL) was added 364-A (200 mg, 0.85 mmol) and stirred for 10 minutes. Diphenyl carbonate (458 mg, 2.14 mmol) was added to the mixture and stirred with slow warming to room temperature for 1 h. 3,3-difluoropyrrolidine (137.71 mg, 1.28 mmol) was added at 0° C. and stirred with slow warming to room temperature for 1 h. The reaction mixture quenched with cold water (10 mL) and extracted with DCM (20 mL×3). The combined organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1-25:1) to give 364-B (150 mg, 47%) as a yellow solid. Synthesis of 364. A mixture of 364-B (140 mg, 0.38 mmol) and Pd/C (35 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Prep-HPLC to give 364 (70 mg, 54%) as a pale yellow solid. Compounds 362, 363, 365, 367, 371, 372, 373, 374, 385, 386, 387, 388, 389, 390, 392, 396 and 397 were synthesized in a similar manner using the appropriately substituted boronic acid and amine variant of 364. Compound 362. 15 mg, 18%, an off white solid. Compound 363. 68 mg, 34%, an off white solid. Compound 365. 30 mg, 54%, an off white solid. Compound 367. 52 mg, 55%, an off white solid. Compound 371. 47 mg, 45%, an off white solid. Compound 372. 45 mg, 38%, an off white solid. Compound 373. 42 mg, 38%, an off white solid. Compound 374. 48 mg, 34%, an off white solid. Compound 385. 80 mg, 44%, an off white solid. Compound 386. 130 mg, 76%, an off white solid. Compound 387. 15 mg, 19%, an off white solid. Compound 388. 60 mg, 33%, an off white solid. Compound 389. 168 mg, 73%, an off white solid. Compound 390. 80 mg, 43%, an off white solid. Compound 392. 70 mg, 38%, an off white solid. Compound 396. 85 mg, 56%, a pale yellow solid. Compound 397. 27 mg, 27%, a pale yellow solid. Compound 398. 110 mg, 46%, a yellowish solid. Compound 399. 100 mg, 46%, an off white solid. Compound 400. 32 mg, 34%, an off white solid. Compound 401. 8 mg, 06%, an off white solid. Compound 403. 145 mg, 78%, an off white solid. Compound 404. 165 mg, 91%, an off white solid. Compound 405. 64 mg, 46%, an off white solid. Compound 408. 45 mg, 43%, an off white solid. Compound 409. 15 mg, 18%, an off white solid. Compound 410. 33 mg, 30%, an off white solid. Compound 411. 100 mg, 63%, an off white solid. Example 38. Synthesis of Compound 381 Synthesis of 381-A. To a cooled (0° C.) solution of N-boc piperidine-4-carboxylic acid (1 g, 4.36 mmol) in DCM (10 mL) was added CDI (0.84 gm, 5.23 mmol) and the reaction mixture was stirred at 0-4° C. for 8 h. The reaction mixture was diluted with diethyl ether (50 mL) and washed with water (20 mL), NaHCO3(10 mL, 10% in water) and brine (10 mL). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo to get 381-A (1 g, 70%) as a white solid. Synthesis of 381-B. To a cooled (0° C.) suspension of NaH (222 mg, 50% in mineral oil, 4.62 mmol) in DMF (5 mL) was added 364-A (0.5 g, 2.31 mmol) and stirred for 20 minutes. 381-A (0.37 g, 2.31 mmol) was added to the mixture and stirred with slow warming to room temperature for 2 h. Then the reaction mixture was quenched with cold water (25 mL) and extracted with DCM (25 mL×3). The combined organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1-50:1) to give 381-B (0.7 g, 71%) as a yellow solid. Synthesis of 381-C. To a cooled (0° C.) solution of 381-B (0.5 g, 1.17 mmol) in DCM (10 mL) was added TFA (1 mL). The reaction mixture was stirred at room temperature overnight. Then the reaction mixture was concentrated in vacuo. The residue was triturated with diethyl ether to give 381-C. TFA (0.35 g, 68%) as a yellow solid. Synthesis of 381-D. To a solution of 381-C. TFA (0.3 g, 0.92 mmol) and oxetan-3-one (0.36 g, 5.05 mmol) in DCM (7.5 mL) and MeOH (2.5 mL) was added AcOH (5.5 mg, 0.09 mmol) and stirred at room temperature for 4 h. NaBH3CN (0.17 g, 2.85 mmol) was added at 0° C. and stirred at room temperature overnight. The reaction mixture was quenched with water (15 mL) and extracted with DCM (25 mL×3). The combined organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:10-50:5) to give 381-D (200 mg, 57%) as a yellow solid. Synthesis of 381. A mixture of 381-D (150 mg, 0.39 mmol) and Pd/C (35 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Prep-HPLC to give 381 (45 mg, 34%) as a brown gum. Compounds 368, 377, 378 and 379 were synthesized in a similar manner using the appropriately substituted 364-A and carbonyl compound variant of 381. Compound 368. 35 mg, 47%, an off white solid. Compound 377. 45 mg, 35%, an off white solid. Compound 378. 75 mg, 20%, an off white solid. Compound 379. 75 mg, 53%, an off white solid. Example 39. Synthesis of Compound 357 Compound 357-C was synthesized in a similar manner using 364-A variant of 381-C. Synthesis of 357. A mixture of 357-C (150 mg, 0.48 mmol) and Pd/C (35 mg) in MeOH (5 mL) was stirred at room temperature for 1 h under H2atmosphere. Pd/C was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by Prep-HPLC to give 357 (20 mg, 15%) as an off white solid. Example 40. Synthesis of Compound 354 Compound 357-A (140 mg, 67%, as a yellow solid) was synthesized in a similar procedure used for 381-D from 381-C. Compound 354 (35 mg, 38%, as an off white solid) was synthesized in a similar procedure used for 357 from 357-C. Example 41. Synthesis of Compound 358 Synthesis of 358-A. To a solution of 357-C (150 mg, 0.48 mmol) and TEA (144 mg, 1.42 mmol) in DCM (10 ml) was added Ac2O (48 mg, 0.48 mmol) and the mixture was stirred at room temperature for 4 h. The reaction mixture was quenched with water (10 ml) and extracted with DCM (20 mL×3). The combined organic layer was dried over anhydrous Na2SO4and then concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1-50:1) to give 358-A (100 mg, 58%) as a yellow solid. Compound 358 (40 mg, 44%, as a brown solid) was synthesized in a similar procedure used for 357 from 357-C. Example 42. Synthesis of Compound 359 Compound 359-A (75 mg, 63%, as a brown solid) was synthesized in a similar procedure used for 381-D from 381-C. Compound 379 (15 mg, 23%, as a brownish solid) was synthesized in a similar procedure used for 357 from 357-C. Example 43. Synthesis of Compound 375 Compound 375-A (500 mg, 40%, as a yellow solid) was synthesized in a similar procedure used for 364-A. Compound 375-B (100 mg, 29%, as a yellow semi-solid) was synthesized in a similar manner using (1-methylpiperidine-4-yl) methanol instead of amine for 364-B. Compound 375 (10 mg, 11%, as a colorless semi-solid) was synthesized in a similar procedure used for 364 from 364-B. Example 44. Synthesis of Compound 366 Compounds 366-A (100 mg, 30%, a yellow solid) was synthesized in a similar manner using the appropriately substituted amine variant of 364-B. Synthesis of 366. To a cooled (0° C.) solution of 366-A in THF (5 mL) was added a solution of LAH in THF under nitrogen and the mixture was stirred with slow warming to 10° C. for 2 h. The reaction mixture was quenched with saturated aqueous solution of Na2SO4. The solid was filtered, washed with DCM (20 ml). The combined filtrate was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by preparative HPLC to give 366 (10 mg, 11%) as an off white solid. Compounds 350 and 395 were synthesized in a similar manner using the appropriately substituted ortho nitro amine and boronic acid variant of 366. Compound 350. 13 mg, 14%, an off white solid. Compound 395. 10 mg, 11%, a greenish solid. Example 45. Synthesis of Compound 394 Synthesis of 394-A. A mixture of 396-A (130 mg, 0.32 mmol) and MnO2(250 mg, 3.2 mmol) in DCM (10 mL) was stirred at room temperature overnight. The MnO2was then removed by filtration through the celite. The filtrate was concentrated to give 394-A (125 mg, 96%) as an yellowish solid. Synthesis of 394-B. To a cooled (−78° C.) solution of 394-A (115 mg, 0.28 mmol) in THF was added a solution of MeMgBr (0.37 mL, 3M in THF, 1.12 mmol) under nitrogen and the mixture was stirred with slow warming to room temperature overnight. The reaction mixture was quenched with saturated Na2SO4solution. The solid was filtered, washed with DCM (20 mL). The organic part was concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM:MeOH=100:1-100:5) to give 394-B (55 mg, 46%) as a yellow solid. Compound 394 (10 mg, 20%, as an off white solid) was synthesized in a similar procedure used for 364 from 364-B. Example 46. Synthesis of Compound 369 Synthesis of 369-A. To a suspension of 2-amino-6-chloro-3-nitro pyridine (1 g, 5.7 mmol) and K2CO3(1.99 g, 14.45 mmol) in acetonitrile (5 mL) was added 177-pyrazole (579.05 mg, 8.64 mmol) the mixture was heated at 70° C. overnight. K2CO3was then removed by filtration through the celite. The filtrate was concentrated and the residue was purified by trituration with diethyl ether to give 369-A (800 mg, 67%) as a yellow solid. Compound 369-B (110 mg, 43%, as a yellow solid) was synthesized in a similar procedure used for 364-B from 364-A. Compound 369 (43 mg, 47%, as an off white solid) was synthesized in a similar procedure used for 364 from 364-B. Example 47. Synthesis of Compound 391 Compound 391-A (200 mg, 45%, as a yellow solid) was synthesized in a similar manner with amine variant of 364-B. Synthesis of 391-B. To a degassed mixture of 391-A (200 mg, 0.48 mmol), 1-methylpiperazine (58 mg, 0.58 mmol), Xanthphos (14 mg, 0.024 mmol) and NaOBut(80 mg, 0.72 mmol) in toluene (10 mL) was added Pd2(dba)3(22 mg, 0.024 mmol) under N2and the mixture was stirred at 100° C. overnight. The reaction mixture was concentrated in vacuo. The residue was dissolved in ethyl acetate (50 mL) and washed with brine (30 mL). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuum. The residue was purified by column chromatography on silica gel (MeOH:CHCl3=100:1-100:5) to give 391-B (100 mg, 43%) as a yellow solid. Compound 391 (15 mg, 16%, as an off white solid) was synthesized in a similar procedure used for 364 from 364-B. Example 48. Synthesis of AM351 Synthesis of AM351-A. A degassed mixture of tert-butyl 5-bromoisoindoline-2-carboxylate (5.0 g, 16.76 mmol), Pd(OAc)2(564 mg, 2.51 mmol), DPPP (1.38 g, 3.35 mmol) and TEA (5.08 g, 50.30 mmol) in a mixture of MeOH (30 mL) and DMSO (30 mL) was heated at 80° C. under carbon monoxide atmosphere overnight. Pd(OAc)2was then removed by filtration through the celite. The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel (PE:EtOAc=75:25˜70:30) to get AM351-A (2.5 g, 53%) as an off white solid. Synthesis of AM351-B. To a cooled (0° C.) solution of AM351-A (2 g, 7.17 mmol) in THF (20 mL) was added a solution of LAH in THF (10.8 mL, 1M in THF, 10.8 mmol) under nitrogen and the mixture was stirred with slow warming to 10° C. for 2 h. The reaction mixture was quenched with saturated aqueous solution of Na2SO4. The solid was filtered, washed with DCM (100 mL). The combined filtrate was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=50:50-40:60) to get AM351-B (1.5 g, 80%) as a white solid. Synthesis of AM351-C. To a mixture of AM351-B (0.8 g, 3.21 mmol) and PPh3 (1.26 g, 4.82 mmol) in dichloromethane was added CBr4(1.59 g, 4.82 mmol) and the mixture was stirred at rt for 4 h. Then the mixture was concentrated and the crude was purified by column chromatography on silica gel (PE:EtOAc=80:20-70:30) to get AM351-C (570 mg, 57%) as a white solid. Synthesis of AM351-D. To a mixture of AM351-C (570 mg, 1.9 mmol) and K2CO3(655 mg, 4.75 mmol) in THF (10 mL) was added dimethylamine·HCl (387 mg, 4.75 mmol) in portions. Then the mixture was stirred at room temperature for 4 h. K2CO3was then removed by filtration through the celite and washed with DCM (20 mL). The combined filtrate was concentrated in vacuo to give AM351-D (480 mg, 95%) as a yellowish semi-solid. Synthesis of AM351. To a cooled (0° C.) solution of AM351-D (480 mg, 1.89 mmol) in DCM (5 mL) was added a solution of dry HCl in diethyl ether (10 mL, 2M) and the mixture was stirred at room temperature overnight. The reaction mixture was concentrated in vacuo and the residue was triturated with diethyl ether to give AM351 (350 mg, 95%) as a purple solid. Compounds AM353, AM355, AM363, AM386, AM408 and AM410 were synthesized in a similar manner using the appropriately substituted amine variant of AM351. Compound AM353. 210 mg, 87%, a grey solid. Compound AM355. 300 mg, 93%, a light pink solid. Compound AM363. 300 mg, 94%, an off white solid. Compound AM386. 300 mg, 92%, an off white solid. Compound AM408. 210 mg, 84%, a grey solid. Compound AM410. 210 mg, 89%, a grey solid. Compound AM411 (200 mg, 84%, a white solid) was synthesized in a similar manner using appropriate starting material and amine variant of AM351. Example 49. Synthesis of AM362 Synthesis of AM362-A. To a degassed mixture of tert-butyl 5-bromoisoindoline-2-carboxylate (1 g, 3.3 mmol), 1-methyl-1,2,3,6-tetrahydropyridine-4-boronic acid pinacol ester (1.12 g, 5.031 mmol) and Cs2CO3(3.2 g, 9.9 mmol) in 1,4-dioxane (10 mL) was added Pd(dppf)Cl2under N2atmosphere and the mixture was heated at 95° C. overnight. The reaction mixture was diluted with DCM (25 mL) and the catalyst was removed by filtration through the celite. The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel (PE:EtOAc=80:20˜60:40) to give AM362-A (0.8 g, 76%) as a brown gum. Compound AM362 (450 mg, 95%, a brown solid) was synthesized in a similar procedure used for AM351 from AM351-D. Compound AM403 (250 mg, 96%, a yellow solid) was synthesized in a similar manner using the appropriately substituted boronic acid pinacol ester variant of AM362. Example 50. Synthesis of AM393 Synthesis of AM393-A. To a cooled (0° C.) solution of NaH (190 mg, 50% in mineral oil, 4.0 mmol) in DMF (5 mL) was added AM351-A (400 mg, 1.6 mmol) and the mixture was stirred at room temperature for 20 min. The reaction mixture was cooled to 0° C. and methyl iodide was added. The reaction mixture was stirred with slow warming to room temperature for 1 h. The reaction mixture was quenched with ice-cold water (10 mL) and the product was extracted with DCM (20 mL×3). The combined organic layer was washed with brine (20 mL) and dried over anhydrous Na2SO4. The solvent was removed in vacuo and the residue was purified by column chromatography on silica gel (PE:EtOAc=90:10˜85:15) to give AM393-A (360 mg, 85%) as a yellowish gum. Compound AM393 (120 mg, 99%, a grey solid) was synthesized in a similar procedure used for AM351 from AM351-I) Example 51. Synthesis of AM374 Synthesis of AM374-A. To a degassed mixture of/erf-butyl 5-bromoisoindoline-2-carboxylate (1 g, 3.35 mmol), 1-methylpiperazine (403 mg, 4.03 mmol), Xanthphos (97 mg, 0.17 mmol) and NaOBut(482 mg, 5.03 mmol) in toluene (10 mL) was added Pd2(dba)3(153 mg, 0.17 mmol) under N2and the mixture was stirred at 100° C. overnight. The reaction mixture was concentrated in vacuo. The residue was dissolved in ethyl acetate (50 mL) and washed with brine (30 mL). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (McOH:CHCL=99:1˜95:5) to give AM374-A (800 mg, 75%) as a white solid. Compound AM374 (510 mg, 93%, an off white solid) was synthesized in a similar procedure used for AM351 from AM351-D. Example 52. Synthesis of AM398 Synthesis of AM398-A. To a degassed solution of tert-butyl 5-bromoisoindoline-2-carboxylate (1.0 g, 3.35 mmol) in DMF (10 mL) were added tri-n-butyl(1-ethoxyvinyl) stannane (1.33 g, 3.69 mmol) and Pd(PPh3)2Cl2(117 mg, 0.16 mmol) and the mixture was heated at 130° C. in Microwave for 2 h. The reaction mixture was concentrated in vacuo. The residue was dissolved with ethyl acetate (50 mL) and washed with brine (30 mL). The organic layer was dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=75:25˜70:30) to give AM398-A (400 mg, 46%) as a yellow sold. Synthesis of AM398-B. To a solution of AM398-A (400 mg, 1.53 mmol) in THF (10 mL) were added pyrrolidine (435 mg, 6.13 mmol) and Ti(IV)isoproxide (1.39 g, 6.13 mmol) and the mixture was stirred at room temperature overnight. EtOH (5 mL) was added to the reaction mixture and cooled to 0° C. NaBH4 (232 mg, 6.13 mmol) was added in portions and the mixture was stirred at room temperature for 30 min. Then the reaction mixture was quenched with ice-cold water (10 mL) and extracted with ethyl acetate (50 mL×3). The organic layer was washed with brine (30 mL), dried over anhydrous Na2SO4and concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc=60:40˜50:50) to give AM398-B (300 mg, 62%) as a pale yellow liquid. Compound AM398 (210 mg, 87%, a yellowish solid) was synthesized in a similar procedure used for AM351 from AM351-D. Example 53. Reporter Displacement Assay HDAC2 containing a C-terminal HIS-Tag (Proteros) and fluorescently-labeled anti-HIS-antibody are diluted in one vial in assay buffer 50 mM Tris, pH 8.0, 1 mM DTT, 150 mM NaCl, and 0.01% Tween20. Components are pre-incubated for 30 min, then a small volume of reporter probe (Proteros) is added from a highly concentrated stock and incubation is continued for another 30 min. Final concentrations after adding the reporter probe amount to 20 nM HDAC2, 4 nM antibody, and 180 nM probe. Ten μl/well of pre-formed complex are transferred into 384 well assay plates (Corning). Compounds to be profiled are serially diluted from 1×101-5.7×10−5mM in DMSO and 60 nl are added to assay plates by pintool transfer (CybiWell, Cybio). Fluorescence intensity signal is read after 4 hours in a Pherastar FS (BMG Labtech) at 337/665 nm. For Kddetermination, percent probe displacement values are calculated for each compound concentration and plotted against the compound concentration. IC50-like values (corresponding to 50% probe displacement) are calculated using standard fitting algorithms. Since the reporter probe is used at a concentration reflecting its own K4value, compound Kdvalues can be calculated according to the Cheng Prusoff equation. The results of this assay for compounds useful in this invention are reported in Table 4, below. In the table, “A” indicates a Kdvalue of less than 0.1 μM; “B” a Kdvalue of between 0.1 μM and 0.5 μM; “C” a Kdvalue of greater than 0.5 μM and less than or equal to 5.0 μM; and “D” a Kdvalue of greater than 5.0 μM. TABLE 4HDAC1 and HDAC2 KdValues for ExemplaryCompounds Useful in the InventionPatentHDAC2HDAC1CompoundHDAC2 Kd,residenceHDAC1 Kd,residenceNo.(μM)time (min)(μM)time (min)100B703B196101D0D0102D0D0103D0D0104D0D0105C634C851106D0D121107C1278C653108C1430C401109C264D14110D0C688111D540D151112B790B329113B609A350114D0D49115C185D0116C114C87117D42D0118C176C69119B1198B126120C1167B830121D1204C682122B2376B572 Example 54. Enzymatic and Cell Assay HDAC Enzymatic Assay All recombinant human HDACs were purchased from BPS Bioscience. The substrate, FAM-TSRHK(AC)KL-CONH, was synthesized at NanoSyn. Final assay reactions contained 100 mM HEPES (pH 7.5), 50 mM KCl, 0.1% BSA, 0.01% Triton X-100, 1% DMSO, 1 uM substrate and 5 nM HDAC enzyme. Enzyme and compounds were pre-incubated at 25° C. for 5 hours and reactions were initiated by addition of substrate. 10 uL reactions were incubated for 17 hours at 25° C. and terminated by the addition of 40 uL of buffer containing 100 mM HEPES (pH 7.5), 0.1% BSA, 0.01% Triton X-100 and 0.05% SDS. Substrate and product peptides present in each sample were separated electrophoretically using the LabChip 3000 capillary electrophoresis instrument. Change in the relative fluorescence intensity of the substrate and product peaks reflects enzyme activity. Reaction progress was determined as the product to sum ratio (PSR):P/(S+P), where P is the peak height of the product peptide and S is the peak height of the substrate peptide. Reactions were performed in duplicate at 12 concentrations, (3× serial dilutions starting at 30 uM). IC50values were calculated using a 4 Parameter Logistic Model. Cell Culture and Inhibitor Treatments SH-SY5Y cells (Sigma) were cultured in Eagle's Modified Essential Medium supplemented with 10% fetal bovine serum and pen/strep. Twenty-four hours prior to compound dosing 20 uL of cells were plated in white 384 well plates at a density of 1,500 cells/well. Compounds were serially diluted in neat DMSO and then diluted 1:100 v/v into media without FBS and mixed. Media was removed from the plated cells and the diluted compounds in serum free media (1% v/v final DMSO) were added and incubated at 37° C. for five hours. Ten uL of HDAC-Glo 2 reagent with 0.1% Triton X-100 was then added, the plate was mixed and allowed to develop at room temperature for 100 minutes. Plates were then read with a Spectramax L Max luminometer employing a 0.4 s integration time. Dose response curves were constructed with normalized data where CI-994 at 100 uM was defined as 100% inhibition and DMSO alone as 0% inhibition. TABLE 5IC50and EC50Values for ExemplaryCompounds Useful in the InventionPatent Comound No.IC50(μM)EC50(μM)123D124D125C126C137C143C144BB145BB146BC147B148BC149BB150B151B152C153B154BB155BB156BB157B158B159BC160B161BB162BB163BB164C165B166C167D168B169C170C171D172C173C174D175D176C177D178D179C181D182D183D184D185C186C187C188C189D190D191C192D193D194D195D196D197D198D199D201D203D204D205D206C208C210D211D212D214C215D218D219D220D221D222D223D224D225C226B228C232C235D236D237D238C239D240B241D242D243BC244C245B246D247B248C249D250BB251BB252BB253BB254C255B256C257BB258CC259B260B261D262D263CC264D265CC266CC267DD268DD269DD270DD271CC272BB273BB274BB275BB276CC277CC350C351B352C353C354D356D357D358D359D360D361D362B363B364C365B366BC367BC368C369C370D371BC372C373BC374B375D376D377D378D379D380D381D382D383B384D385C386B387C388C389D390C391BB392C393C394B395DC396B397BB401D402403404405BB406D407D408B409B410B“A” indicates a IC50or EC50value of less than 0.1 μM;“B” an IC50or EC50value of between 0.1 μM and 0.5 μM;“C” an IC50or EC50value of greater than 0.5 μM and less than or equal to 5.0 μM; and“D” an IC50or EC50value of greater than 5.0 μM. Morris Water Maze Task The compounds described herein (e.g., compounds according to Formula I, II or any of Compounds 100-128 or any of those in Tables 2 or 3) can be examined for its efficacy in the model behavior paradigm, the Morris water maze task as described below: The water maze task was originally designed by Morris et al. (J Neurosci Methods. 1984; 11: 47-60). Testing is performed in a large dark-colored tank (200 cm in diameter) filled with clear water at a temperature of 25.0±1.0° C. A submerged platform (square platform: 10×10 cm; 1.5 cm below water surface) is placed in the middle of the of the NW quadrant. The starting locations, which are labeled N, NE, E, SE, S, SW, W, NW, are located arbitrarily on the pool rim. The rats are lowered into the pool with their nose pointing toward the wall at one of the starting points. The release point adjacent to platform location (NW) is not used. At first, before the compound treatment is started, the visible platform pre-training is performed to determine whether any non-cognitive performance impairments (e.g. visual impairments and/or swimming difficulties) are present, which might affect performance on the place or probe trials. All rats receive 4 trials in one day with inter-trial interval of 15 min. On each trial, rats are placed in a fixed position in the swimming pool facing the wall and are allowed to swim to a platform with a rod (cue) 20 cm above water level randomly placed in middle of the pool. They are allowed 60 s to find the platform, which they stay on for 15 s before being removed from the pool. If a rat does not find the platform within 60 s, the rat will be gently guided to the platform and allowed to remain there for 15 s. The time for each rat to reach the cued platform, distance swam, thigmotaxis, and the swim speed are recorded. After the visible platform pre-training is completed, the data is analyzed and the rats are assigned to the different treatment groups based on their pre-training performance. This procedure is performed to ensure that each treatment group consist equally both good and poor performers in the cued version of the water maze task. Acquisition training—week 1: After completion of cued trials, acquisition (place) trials are executed to determine the rat's ability to learn the spatial relationship between distant cues and the escape platform (submerged, no cue rod), which remain in the same location for all place trials. The starting points are randomized (NW is not used). The rats receive four trials (15 min apart, 60 s maximum for each trial) each day for 4 days. Latency, path length, thigmotaxis and swim speed are recorded. Acquisition training—week 2: A second set of acquisition trials is executed to determine the rat's ability to learn the spatial relationship between distant cues and the escape platform (submerged, no cue rod), which remain in the same location for all place trials. The starting points are randomized (NW is not used). The rats receive four trials (15 min apart, 60 s maximum for each trial) each day for 4 days. Latency, path length, thigmotaxis and swim speed are recorded. Probe trial: A single probe trial is conducted 24 hours after the last place trials to evaluate memory retention capabilities. The platform is removed from the water maze and rat is started to swim in the quadrant opposite to one the platform was placed before. The rats are allowed to swim for 60 s during the probe trial. During the probe trial, the time spent in target quadrant and target platform annulus (36-cm-diameter circular area surrounding platform), and crosses over the target platform position are measured (memory retention). After completing the behavioral tests, the rats are sacrificed and tissue collected for further analysis. Blood was collected and processed to peripheral mononuclear cells and plasma. The cells can be further assayed for acetylation marks to demonstrate that compounds were inhibiting HDAC2 activity. The plasma can be frozen and later assayed by mass spectrometry for the presence of compound. Brain can be collected, dissected into cerebellum and hippocampus. Cerebellum can be frozen and later homogenized and the compound can be extracted and measured by mass spectrometry. Hippocampal brain tissue can be processed to extract RNA for gene expression analysis. Tissue can be washed with phosphate buffered saline (PBS). RNA can be isolated using the RNeasy isolation kit (Qiagen) according to manufacturer's instructions. The RNA is eluted in 30 μl RNAse free water. The concentration of the isolated RNA can be measured by nanodrop. The RNA can be converted into cDNA with the iScript kit (Biorad) according to manufacturer's instructions. 800 ng of RNA was used per sample. After cDNA synthesis the DNA was diluted 1:5 with milliQ water. Quantitative PCR was done with the SSo advanced supermix (Biorad). Reactions can be done in a white 96-well plate, each reaction contained 1 μl template, 0.75 μl primer mix (forward & reverse, both at 10 μM), 5.75 μl water and 7.5 μL SSo SYBR green advanced supermix. Detection can be done with a CFX Connect Instrument (Biorad). Gene-specific primers for the following genes can be used in these studies: GAPDH—glyceraldehyde-3-phosphate dehydrogenase: BDNF—Brain Derived Neurotrophic Factor: GRIN2A and GRIN2B Glutamate receptor N-methyl D-aspartate-associated proteins 1& 2: CDK5—cyclin-dependent kinase 5: HOMER1: GRIA1 and GRIA2—glutamate receptor, AMPA 1 & 2: EGR1—early growth response 1: NEFL—neurofilament, light polypeptide: SYT1—Synaptotagmin 1: SYP—synaptophysin. Values can be normalized to expression levels of GAPDH. Three replicates of each sample can be run in each assay and the mean of the replicates can be compared for statistical significant changes. Peripheral blood mononuclear cells can be isolated using a Ficoll-Paque Plus (GE Healthcare) and can be tested for acetylation marks following treatment with HDCA2 inhibitors. Blood cells can be lysed and proteins can be extracted using 13 RIPA buffer containing proteinase (complete, Roche) and phosphatase inhibitors (1 mMb-glycerophosphate, 10 mM NaF, 0.1 mM Na3VO4) and then can be transferred onto PVDF membranes (Biorad) and stripped using stripping buffer (Thermo Scientific). The following primary antibodies can be used: acetyl-K (Cell Signaling) and actin (Sigma). Secondary antibodies were horseradish peroxidase-linked (GE Healthcare). Signal intensities can be quantified using Image J 1.42q and normalized to values of actin. While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments, which utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments, which have been represented by way of example.
302,475
11858940
DETAILED DESCRIPTION The following is a detailed description provided to aid those skilled in the art in practicing the present invention. Those of ordinary skill in the art may make modifications and variations in the embodiments described herein without departing from the spirit or scope of the present disclosure. All publications, patent applications, patents, and other references mentioned herein are expressly incorporated by reference in their entirety. Presently described are compounds, compositions and methods that relate to the surprising and unexpected discovery that an E3 ubiquitin ligase (e.g., a cereblon E3 ubiquitin ligase) ubiquitinates the human LRRK2 protein once the E3 ubiquitin ligase and the LRRK2 protein are placed in proximity via a bifunctional compound that binds both the E3 ubiquitin ligase and the LRRK2 protein. Accordingly the present disclosure provides compounds and compositions comprising an E3 ubiquitin ligase binding moiety (“ULM”) coupled by a bond or chemical linking group (L) to a protein targeting moiety (“PTM”) that targets the LRRK2 protein, which results in the ubiquitination of the LRRK2 protein, and which leads to degradation of the LRRK2 protein by the proteasome (seeFIG.1). In one aspect, the description provides compounds in which the PTM preferably binds the LRRK2 protein. The present disclosure also provides a library of compositions and the use thereof to produce targeted degradation of the LRRK2 protein in a cell. In certain aspects, the present disclosure provides hetero-bifunctional compounds which comprise a ligand, e.g., a small molecule ligand (i.e., having a molecular weight of below 2,000, 1,000, 500, or 200 Daltons), which is capable of binding to an E3 ubiquitin ligase, such as cereblon. The compounds also comprise a small molecule moiety that is capable of binding to LRRK2 in such a way that the LRRK2 protein is placed in proximity to the ubiquitin ligase to effect ubiquitination and degradation (and/or inhibition) of the LRRK2 protein. “Small molecule” means, in addition to the above, that the molecule is non-peptidyl, that is, it is not considered a peptide, e.g., comprises fewer than 4, 3, or 2 amino acids. In accordance with the present description, each of the PTM, ULM and hetero-bifunctional molecule is a small molecule. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The terminology used in the description is for describing particular embodiments only and is not intended to be limiting of the disclosure. Where a range of values is provided, it is understood that each intervening value in the range, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise (such as in the case of a group containing a number of carbon atoms in which case each carbon atom number falling within the range is provided), between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the disclosure. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either/or both of those included limits are also included in the disclosure. The following terms are used to describe the present disclosure. In instances where a term is not specifically defined herein, that term is given an art-recognized meaning by those of ordinary skill applying that term in context to its use in describing the present disclosure. The articles “a” and “an” as used herein and in the appended claims are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article unless the context clearly indicates otherwise. By way of example, “an element” means one element or more than one element, unless otherwise indicated. In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of” and “consisting essentially of” shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03. It should also be understood that, in certain methods or processes described herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited unless the context indicates otherwise. The terms “co-administration” and “co-administering” or “combination therapy” refer to both concurrent administration (administration of two or more therapeutic agents at the same time) and time-varied administration (administration of one or more therapeutic agents at a time different from that of the administration of an additional therapeutic agent or agents), as long as the two or more therapeutic agents are present in the patient to some extent, preferably at effective amounts, at the same time. In certain preferred aspects, one or more of the hetero-bifunctional compounds described herein are coadministered with at least one additional bioactive agent, e.g., an anticancer agent. In particularly preferred aspects, the co-administration of such compounds results in synergistic activity and/or therapy such as, e.g., anticancer activity. The term “compound”, as used herein, unless otherwise indicated, refers to any specific hetero-bifunctional compound disclosed herein, pharmaceutically acceptable salts and solvates thereof, and deuterated forms of any of the aforementioned molecules, where applicable. Deuterated compounds contemplated are those in which one or more of the hydrogen atoms contained in the drug molecule have been replaced by deuterium. Such deuterated compounds preferably have one or more improved pharmacokinetic or pharmacodynamic properties (e.g., longer half-life) compared to the equivalent “undeuterated” compound. The term “ubiquitin ligase” refers to a family of proteins that facilitate the transfer of one or more ubiquitins to a specific substrate protein. Addition of a chain of several ubiquitins (poly-ubiquitination) targets the substrate protein for degradation. For example, cereblon is an E3 ubiquitin ligase that alone, or in combination with an E2 ubiquitin-conjugating enzyme, can ultimately cause the attachment of a chain of four ubiquitins to a lysine residue on the target protein, thereby targeting the protein for degradation by the proteasome. The ubiquitin ligase is involved in poly-ubiquitination such that a first ubiquitin is attached to a lysine on the target protein; a second ubiquitin is attached to the first; a third is attached to the second, and a fourth is attached to the third. Such poly-ubiquitination marks proteins for degradation by the proteasome. The term “patient” or “subject” is used throughout the specification to describe an animal, preferably a human or a domesticated animal, to whom treatment, including prophylactic treatment, with the compositions according to the present disclosure is provided. For treatment of those diseases, conditions or symptoms that are specific for a specific animal, such as a human patient, the term “patient” refers to that specific animal, including a domesticated animal such as a dog or cat, or a farm animal such as a horse, cow, sheep, etc. In general, in the present disclosure, the terms “patient” and “subject” refer to a human patient unless otherwise stated or implied from the context of the use of the term. The terms “effective” and “therapeutically effective” are used to describe an amount of a compound or composition which, when used within the context of its intended use, and either in a single dose or, more preferably after multiple doses within the context of a treatment regimen, effects an intended result such as an improvement in a disease or condition, or amelioration or reduction in one or more symptoms associated with a disease or condition. The terms “effective” and “therapeutically effective” subsume all other “effective amount” or “effective concentration” terms, which are otherwise described or used in the present application. Compounds and Compositions In one aspect, the description provides hetero-bifunctional compounds comprising an E3 ubiquitin ligase binding moiety (“ULM”) that is a cereblon E3 ubiquitin ligase binding moiety (a “CLM”), The CLM is covalently coupled to a protein targeting moiety (PTM) that binds to the protein, which coupling is either directly by a bond or via a chemical linking group (L) according to the structure: PTM-L-CLM  (A) wherein L is the bond or chemical linking group, and PTM is a protein targeting moiety that binds to the protein LRRK2, where the PTM is a LRRK2 targeting moiety. The term CLM is inclusive of all cereblon binding moieties. In any of the aspects or embodiments, the CLM demonstrates a half maximal inhibitory concentration (IC50) for the E3 ubiquitin ligase (e.g., cereblon E3 ubiquitin ligase) of less than about 200 μM. The IC50can be determined according to any suitable method known in the art, e.g., a fluorescent polarization assay. In certain embodiments, the hetero-bifunctional compounds described herein demonstrate an IC50or a half maximal degradation concentration (DC50) of less than about 100, 50, 10, 1, 0.5, 0.1, 0.05, 0.01, 0.005, 0.001 mM, or less than about 100, 50, 10, 1, 0.5, 0.1, 0.05, 0.01, 0.005, 0.001 μM, or less than about 100, 50, 10, 1, 0.5, 0.1, 0.05, 0.01, 0.005, 0.001 nM, or less than about 100, 50, 10, 1, 0.5, 0.1, 0.05, 0.01, 0.005, 0.001 μM. The term “alkyl” shall mean within its context a linear, branch-chained or cyclic fully saturated hydrocarbon radical, preferably a C1-C10, preferably a C1-C6, or more preferably a C1-C3alkyl group, which may be optionally substituted with any suitable functional group or groups. Examples of alkyl groups are methyl, ethyl, n-butyl, sec-butyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, isopropyl, 2-methylpropyl, cyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl, cyclopentylethyl, cyclohexylethyl and cyclohexyl, among others. In certain embodiments, the alkyl group is end-capped with a halogen group (At, Br, Cl, F, or I). The term “Alkenyl” refers to linear, branch-chained or cyclic C2-C10(preferably C2-C6) hydrocarbon radicals containing at least one C═C bond. The term “Alkynyl” refers to linear, branch-chained or cyclic C2-C10(preferably C2-C6) hydrocarbon radicals containing at least one C≡C bond. The term “alkylene” when used, refers to a —(CH2)n— group (n is an integer generally from 0-6), which may be optionally substituted. When substituted, the alkylene group preferably is substituted on one or more of the methylene groups with a C1-C6alkyl group (including a cyclopropyl group or a t-butyl group), but may also be substituted with one or more halo groups, preferably from 1 to 3 halo groups or one or two hydroxyl groups, O—(C1-C6alkyl) groups or amino acid sidechains as otherwise disclosed herein. In certain embodiments, an alkylene group may be substituted with a urethane or alkoxy group (or other suitable functional group) which may be further substituted with a polyethylene glycol chain (of from 1 to 10, preferably 1 to 6, or more preferably 1 to 4 ethylene glycol units) to which is substituted (preferably, but not exclusively on the distal end of the polyethylene glycol chain) an alkyl chain substituted with a single halogen group, preferably a chlorine group. In still other embodiments, the alkylene (e.g., methylene) group, may be substituted with an amino acid sidechain group such as a sidechain group of a natural or unnatural amino acid, for example, alanine, β-alanine, arginine, asparagine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan or tyrosine. The term “unsubstituted” shall mean substituted only with hydrogen atoms. A range of carbon atoms which includes C0means that carbon is absent and is replaced with H. Thus, a range of carbon atoms which is C0-C6includes carbons atoms of 1, 2, 3, 4, 5 and 6 and for C0, H stands in place of carbon. The term “substituted” or “optionally substituted” shall mean independently (i.e., where more than one substituent occurs, each substituent is selected independent of another substituent) one or more substituents (independently up to five substituents, preferably up to three substituents, more preferably 1 or 2 substituents on a moiety in a compound according to the present disclosure and may include substituents which themselves may be further substituted) at a carbon (or nitrogen) position anywhere on a molecule within context, and includes as possible substituents hydroxyl, thiol, carboxyl, cyano (C≡N), nitro (NO2), halogen (preferably, 1, 2 or 3 halogens, especially on an alkyl, especially a methyl group such as a trifluoromethyl), an alkyl group (preferably, C1-C10, more preferably, C1-C6), aryl (especially phenyl and substituted phenyl, for example benzyl or benzoyl), alkoxy group (preferably, C1-C6alkyl or aryl, including phenyl and substituted phenyl), thioether (preferably, C1-C6alkyl or aryl), acyl (preferably, C1-C6acyl), ester or thioester (preferably, C1-C6alkyl or aryl) including alkylene ester (such that attachment is on the alkylene group, rather than at the ester function which is preferably substituted with a C1-C6alkyl or aryl group), halogen (preferably, F or Cl), amine (including a five- or six-membered cyclic alkylene amine, further including a C1-C6alkyl amine or a C1-C6dialkyl amine which alkyl groups may be substituted with one or two hydroxyl groups) or an optionally substituted —N(C0-C6alkyl)C(O)(O—C1-C6alkyl) group (which may be optionally substituted with a polyethylene glycol chain to which is further bound an alkyl group containing a single halogen, preferably chlorine substituent), hydrazine, amido, which are preferably independently substituted with one or two C1-C6alkyl groups (including a carboxamide which is optionally substituted with one or two C1-C6alkyl groups), alkanol (preferably, C1-C6alkyl or aryl), or alkanoic acid (preferably, C1-C6alkyl or aryl). Substituents according to the present disclosure may include, for example —SiR1R2R3groups where each of R1and R2is as otherwise described herein and R3is H or a C1-C6alkyl group, preferably R1, R2, R3together is a C1-C3alkyl group (including an isopropyl or t-butyl group). Each of the above-described groups may be linked directly to the substituted moiety or alternatively, the substituent may be linked to the substituted moiety (preferably in the case of an aryl or heteroaryl moiety) through an optionally substituted —(CH2)m— or alternatively an optionally substituted —(OCH2)m—, —(OCH2CH2)m— or —(CH2CH2O)m— group, which may be substituted with any one or more of the above-described substituents. Alkylene groups —(CH2)m— or —(CH2)n— groups or other chains such as ethylene glycol chains, as identified above, may be substituted anywhere on the chain. Preferred substituents on alkylene groups include halogen or C1-C6(preferably C1-C3) alkyl groups, which may be optionally substituted with one or two hydroxyl groups, one or two ether groups (O—C1-C6groups), up to three halo groups (preferably F), or a side chain of an amino acid as otherwise described herein and optionally substituted amide (preferably carboxamide substituted as described above) or urethane groups (often with one or two C0-C6alkyl substituents, which group(s) may be further substituted). In certain embodiments, the alkylene group (often a single methylene group) is substituted with one or two optionally substituted C1-C6alkyl groups, preferably C1-C4alkyl group, most often methyl or O-methyl groups or a sidechain of an amino acid as otherwise described herein. In the present disclosure, a moiety in a molecule may be optionally substituted with up to five substituents, preferably up to three substituents. Most often, in the present disclosure moieties which are substituted are substituted with one or two substituents. The term “substituted” (each substituent being independent of any other substituent) shall also mean within its context of use C1-C6alkyl, C1-C6alkoxy, halogen, amido, carboxamido, sulfone, including sulfonamide, keto, carboxy, C1-C6ester (oxyester or carbonylester), C1-C6keto, urethane —O—C(O)—NR1R2or —N(R1)—C(O)—O—R1, nitro, cyano and amine (especially including a C1-C6alkylene-NR1R2, a mono- or di-C1-C6alkyl substituted amines which may be optionally substituted with one or two hydroxyl groups). Each of these groups contain unless otherwise indicated, within context, between 1 and 6 carbon atoms. In certain embodiments, preferred substituents will include for example, —NH—, —NHC(O)—, —O—, ═O, —(CH2)m— (here, m and n are in context, 1, 2, 3, 4, 5 or 6), —S—, —S(O)—, SO2— or —NH—C(O)—NH—, —(CH2)nOH, —(CH2)nSH, —(CH2)nCOOH, C1-C6alkyl, —(CH2)nO—(C1-C6alkyl), —(CH2)nC(O)—(C1-C6alkyl), —(CH2)nOC(O)—(C1-C6alkyl), —(CH2)nC(O)O—(C1-C6alkyl), —(CH2)nNHC(O)—R1, —(CH2)nC(O)—NR1R2, —(OCH2)nOH, —(CH2O)nCOOH, C1-C6alkyl, —(OCH2)nO—(C1-C6alkyl), —(CH2O)nC(O)—(C1-C6alkyl), —(OCH2)nNHC(O)—R1, —(CH2O)nC(O)—NR1R2, —S(O)2—RS, —S(O)—RS(RSis C1-C6alkyl or a —(CH2)m—NR1R2group), NO2, CN or halogen (F, Cl, Br, I, preferably F or Cl), depending on the context of the use of the substituent. R1and R2are each, within context, H or a C1-C6alkyl group (which may be optionally substituted with one or two hydroxyl groups or up to three halogen groups, preferably fluorine). The term “substituted” shall also mean, within the chemical context of the compound defined and substituent used, an optionally substituted aryl or heteroaryl group or an optionally substituted heterocyclic group as otherwise described herein. Alkylene groups may also be substituted as otherwise disclosed herein, preferably with optionally substituted C1-C6alkyl groups (methyl, ethyl or hydroxymethyl or hydroxyethyl is preferred, thus providing a chiral center), a sidechain of an amino acid group as otherwise described herein, an amido group as described hereinabove, or a urethane group O—C(O)—NR1R2group where R1and R2are as otherwise described herein, although numerous other groups may also be used as substituents. Various optionally substituted moieties may be substituted with 3 or more substituents, preferably no more than 3 substituents and preferably with 1 or 2 substituents. It is noted that in instances where, in a compound at a particular position of the molecule substitution is required (principally, because of valency), but no substitution is indicated, then that substituent is construed or understood to be H, unless the context of the substitution suggests otherwise. The term “aryl” or “aromatic”, in context, refers to a substituted (as otherwise described herein) or unsubstituted monovalent aromatic radical (e.g., a 5-16 membered ring) having a single ring (e.g., benzene, phenyl, benzyl, or 5, 6, 7 or 8 membered ring) or condensed rings (e.g., naphthyl, anthracenyl, phenanthrenyl, 10-16 membered ring, etc.) and can be bound to the compound according to the present disclosure at any available stable position on the ring(s) or as otherwise indicated in the chemical structure presented. Other examples of aryl groups, in context, may include heterocyclic aromatic ring systems, “heteroaryl” groups having one or more nitrogen, oxygen, or sulfur atoms in the ring (moncyclic) such as imidazole, furyl, pyrrole, furanyl, thiene, thiazole, pyridine, pyrimidine, pyrazine, triazole, oxazole or fused ring systems such as indole, quinoline, indolizine, azaindolizine, benzofurazan, etc., among others, which may be optionally substituted as described above. Among the heteroaryl groups which may be mentioned include nitrogen-containing heteroaryl groups such as pyrrole, pyridine, pyridone, pyridazine, pyrimidine, pyrazine, pyrazole, imidazole, triazole, triazine, tetrazole, indole, isoindole, indolizine, azaindolizine, purine, indazole, quinoline, dihydroquinoline, tetrahydroquinoline, isoquinoline, dihydroisoquinoline, tetrahydroisoquinoline, quinolizine, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, imidazopyridine, imidazotriazine, pyrazinopyridazine, acridine, phenanthridine, carbazole, carbazoline, pyrimidine, phenanthroline, phenacene, oxadiazole, benzimidazole, pyrrolopyridine, pyrrolopyrimidine and pyridopyrimidine; sulfur-containing aromatic heterocycles such as thiophene and benzothiophene; oxygen-containing aromatic heterocycles such as furan, pyran, cyclopentapyran, benzofuran and isobenzofuran; and aromatic heterocycles comprising 2 or more hetero atoms selected from among nitrogen, sulfur and oxygen, such as thiazole, thiadizole, isothiazole, benzoxazole, benzothiazole, benzothiadiazole, phenothiazine, isoxazole, furazan, phenoxazine, pyrazoloxazole, imidazothiazole, thienofuran, furopyrrole, pyridoxazine, furopyridine, furopyrimidine, thienopyrimidine and oxazole, among others, all of which may be optionally substituted. The term “substituted aryl” refers to an aromatic carbocyclic group comprised of at least one aromatic ring or of multiple condensed rings at least one of which being aromatic, wherein the ring(s) are substituted with one or more substituents. For example, an aryl group can comprise a substituent(s) selected from: —(CH2)nOH, —(CH2)n—O—(C1-C6)alkyl, —(CH2)n—O—(CH2)n—(C1-C6)alkyl, —(CH2)n—C(O)(C0-C6) alkyl, —(CH2)n—C(O)O(C0-C6)alkyl, —(CH2)n—OC(O)(C0-C6)alkyl, amine, mono- or di-(C1-C6alkyl) amine wherein the alkyl group on the amine is optionally substituted with 1 or 2 hydroxyl groups or up to three halo (preferably F, Cl) groups, OH, COOH, C1-C6alkyl, preferably CH3, CF3, OMe, OCF3, NO2, or CN group (each of which may be substituted in ortho-, meta- and/or para-positions of the phenyl ring, preferably para-), an optionally substituted phenyl group (the phenyl group itself is preferably connected to a PTM group, including a ULM group, via a linker group), and/or at least one of F, Cl, OH, COOH, CH3, CF3, OMe, OCF3, NO2, or CN group (in ortho-, meta- and/or para-positions of the phenyl ring, preferably para-), a naphthyl group, which may be optionally substituted, an optionally substituted heteroaryl, preferably an optionally substituted isoxazole including a methyl substituted isoxazole, an optionally substituted oxazole including a methyl substituted oxazole, an optionally substituted thiazole including a methyl substituted thiazole, an optionally substituted isothiazole including a methyl substituted isothiazole, an optionally substituted pyrrole including a methyl substituted pyrrole, an optionally substituted imidazole including a methylimidazole, an optionally substituted benzimidazole or methoxybenzylimidazole, an optionally substituted oximidazole or methyloximidazole, an optionally substituted diazole group, including a methyldiazole group, an optionally substituted triazole group, including a methyl substituted triazole group, an optionally substituted pyridine group, including a halo- (preferably, F) or methyl substituted pyridine group or an oxapyridine group (where the pyridine group is linked to the phenyl group by an oxygen), an optionally substituted furan, an optionally substituted benzofuran, an optionally substituted dihydrobenzofuran, an optionally substituted indole, indolizine or azaindolizine (2, 3, or 4-azaindolizine), an optionally substituted quinoline, and combinations thereof. “Carboxyl” denotes the group —C(O)OR, where R is hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl or substituted heteroaryl, whereas these generic substituents have meanings which are identical with definitions of the corresponding groups defined herein. The term “heteroaryl” or “hetaryl” can mean but is in no way limited to a 5-16 membered heteroaryl (e.g., 5, 6, 7 or 8 membered monocyclic ring or a 10-16 membered heteroaryl having multiple condensed rings), an optionally substituted quinoline (which may be attached to the pharmacophore or substituted on any carbon atom within the quinoline ring), an optionally substituted indole (including dihydroindole), an optionally substituted indolizine, an optionally substituted azaindolizine (2, 3 or 4-azaindolizine) an optionally substituted benzimidazole, benzodiazole, benzoxofuran, an optionally substituted imidazole, an optionally substituted isoxazole, an optionally substituted oxazole (preferably methyl substituted), an optionally substituted diazole, an optionally substituted triazole, a tetrazole, an optionally substituted benzofuran, an optionally substituted thiophene, an optionally substituted thiazole (preferably methyl and/or thiol substituted), an optionally substituted isothiazole, an optionally substituted triazole (preferably a 1,2,3-triazole substituted with a methyl group, a triisopropylsilyl group, an optionally substituted —(CH2)m—O—C1-C6alkyl group or an optionally substituted —(CH2)m—C(O)—O—C1-C6alkyl group), an optionally substituted pyridine (2-, 3, or 4-pyridine) or a group according to the chemical structure: wherein:Scis CHRSS, NRURE, or O;RHETis H, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group —C≡C—Rawhere Rais H or a C1-C6alkyl group (preferably C1-C3alkyl);RSSis H, CN, NO2, halo (preferably F or Cl), optionally substituted C1-C6alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O—(C1-C6alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted —C(O)(C1-C6alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);RUREis H, a C1-C6alkyl (preferably H or C1-C3alkyl) or a —C(O)(C1-C6alkyl), each of which groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted heterocycle, for example piperidine, morpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, piperazine, each of which is optionally substituted, andYCis N or C—RYC, where RYCis H, OH, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group —C≡C—Rawhere Rais H or a C1-C6alkyl group (preferably C1-C3alkyl). The terms “aralkyl” and “heteroarylalkyl” refer to groups that comprise both aryl or, respectively, heteroaryl as well as alkyl and/or heteroalkyl and/or carbocyclic and/or heterocycloalkyl ring systems according to the above definitions. The term “arylalkyl” as used herein refers to an aryl group as defined above appended to an alkyl group defined above. The arylalkyl group is attached to the parent moiety through an alkyl group wherein the alkyl group is one to six carbon atoms. The aryl group in the arylalkyl group may be substituted as defined above. The term “Heterocycle” refers to a cyclic group which contains at least one heteroatom, e.g., N, O or S, and may be aromatic (heteroaryl) or non-aromatic. Thus, the heteroaryl moieties are subsumed under the definition of heterocycle, depending on the context of its use. Exemplary heteroaryl groups are described hereinabove. Exemplary heterocyclics include: azetidinyl, benzimidazolyl, 1,4-benzodioxanyl, 1,3-benzodioxolyl, benzoxazolyl, benzothiazolyl, benzothienyl, dihydroimidazolyl, dihydropyranyl, dihydrofuranyl, dioxanyl, dioxolanyl, ethyleneurea, 1,3-dioxolane, 1,3-dioxane, 1,4-dioxane, furyl, homopiperidinyl, imidazolyl, imidazolinyl, imidazolidinyl, indolinyl, indolyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isoxazolidinyl, isoxazolyl, morpholinyl, naphthyridinyl, oxazolidinyl, oxazolyl, pyridone, 2-pyrrolidone, pyridine, piperazinyl, N-methylpiperazinyl, piperidinyl, phthalimide, succinimide, pyrazinyl, pyrazolinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, pyrrolyl, quinolinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydroquinoline, thiazolidinyl, thiazolyl, thienyl, tetrahydrothiophene, oxane, oxetanyl, oxathiolanyl, thiane among others. Heterocyclic groups can be optionally substituted with a member selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxy, carboxyalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, —SO-alkyl, —SO-substituted alkyl, —SOaryl, —SO— heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl, oxo (═O), and —SO2-heteroaryl. Such heterocyclic groups can have a single ring or multiple condensed rings. Examples of nitrogen heterocycles and heteroaryls include, but are not limited to, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, morpholino, piperidinyl, tetrahydrofuranyl, and the like as well as N-alkoxy-nitrogen containing heterocycles. The term “heterocyclic” also includes bicyclic groups in which any of the heterocyclic rings is fused to a benzene ring or a cyclohexane ring or another heterocyclic ring (for example, indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, and the like). The term “cycloalkyl” can mean but is in no way limited to univalent groups derived from monocyclic or polycyclic alkyl groups or cycloalkanes, as defined herein, e.g., saturated monocyclic hydrocarbon groups having from three to twenty carbon atoms in the ring, including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like. The term “substituted cycloalkyl” can mean but is in no way limited to a monocyclic or polycyclic alkyl group and being substituted by one or more substituents, for example, amino, halogen, alkyl, substituted alkyl, carbyloxy, carbylmercapto, aryl, nitro, mercapto or sulfo, whereas these generic substituent groups have meanings which are identical with definitions of the corresponding groups as defined in this legend. “Heterocycloalkyl” refers to a monocyclic or polycyclic alkyl group in which at least one ring carbon atom of its cyclic structure being replaced with a heteroatom selected from the group consisting of N, O, S or P. “Substituted heterocycloalkyl” refers to a monocyclic or polycyclic alkyl group in which at least one ring carbon atom of its cyclic structure being replaced with a heteroatom selected from the group consisting of N, O, S or P and the group is containing one or more substituents selected from the group consisting of halogen, alkyl, substituted alkyl, carbyloxy, carbylmercapto, aryl, nitro, mercapto or sulfo, whereas these generic substituent group have meanings which are identical with definitions of the corresponding groups as defined in this legend. The term “hydrocarbyl” shall mean a compound which contains carbon and hydrogen and which may be fully saturated, partially unsaturated or aromatic and includes aryl groups, alkyl groups, alkenyl groups and alkynyl groups. The term “independently” is used herein to indicate that the variable, which is independently applied, varies independently from application to application. The term “lower alkyl” refers to methyl, ethyl or propyl The term “lower alkoxy” refers to methoxy, ethoxy or propoxy. Exemplary CLMs Neo-Imide Compounds In any aspect or embodiment described herein, the description provides CLMs useful for binding and recruiting cereblon. In certain embodiments, the CLM is selected from the group consisting of chemical structures: wherein:W of Formulas (a1) through (e) [e.g., (a1), (a2), (a3), (a4), (b), (c), (d1), (d2), and/or (e)] is independently selected from the group CH2, O, CHR, C═O, SO2, NH, N, optionally substituted cyclopropyl group, optionally substituted cyclobutyl group, and N-alkyl;W3of Formulas (a1) through (e) is selected from C or N;X of Formulas (a1) through (e) is independently selected from the group absent, O, S and CH2;Y of Formulas (a1) through (e) is independently selected from the group CH2, —C═CR′, NH, N-alkyl, N-aryl, N-heteroaryl, N-cycloalkyl, N-heterocyclyl, O, and S;Z of Formulas (a1) through (e) is independently selected from the group absent, O, and S or CH2except that both X and Z cannot be CH2or absent;G and G′ of Formulas (a1) through (e) are independently selected from the group H, optionally substituted linear or branched alkyl, OH, R′OCOOR, R′OCONRR″, CH2-heterocyclyl optionally substituted with R′, and benzyl optionally substituted with R′;Q1-Q4 of Formulas (a1) through (e) represent a carbon C or N substituted with a group independently selected from H, R, N or N-oxide;A of Formulas (a1) through (e) is independently selected from the group H, optionally substituted linear or branched alkyl, cycloalkyl, Cl and F;n of Formulas (a1) through (e) represent an integer from 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);R of Formulas (a1) through (e) comprises, but is not limited to: a bond, H, —C(═O)R′ (e.g., a carboxy group), —CONR′R″ (e.g., an amide group), —OR′ (e.g., OH), —NR′R″ (e.g., an amine group), —SR′, —SO2R′, —SO2NR′R″, —CR′R″—, —CR′NR′R″—, (—CR′O)nR″, optionally substituted heterocyclyl, optionally substituted aryl, (e.g., an optionally substituted C5-C7 aryl), optionally substituted alkyl-aryl (e.g., an alkyl-aryl comprising at least one of an optionally substituted C1-C6 alkyl, an optionally substituted C5-C7 aryl, or combinations thereof), optionally substituted heteroaryl, optionally substituted alkyl (e.g., a C1-C6 linear or branched alkyl optionally substituted with one or more halogen, cycloalkyl (e.g., a C3-C6 cycloalkyl), or aryl (e.g., C5-C7 aryl)), optionally substituted alkoxyl group (e.g., a methoxy, ethoxy, butoxy, propoxy, pentoxy, or hexoxy; wherein the alkoxyl may be substituted with one or more halogen, alkyl, haloalky, fluoroalkyl, cycloalkyl (e.g., a C3-C6 cycloalkyl), or aryl (e.g., C5-C7 aryl)), optionally substituted cycloalkyl, optionally substituted heterocyclyl, —P(O)(OR′)R″, —P(O)R′R″, OP(O)(OR′)R″, —OP(O)R′R″, —Cl, —F, —Br, —I, —CF3, —CN, —NR′SO2NR′R″, —NR′CONR′R″, —CONR′COR″, —NR′C(═N—CN)NR′R″, —C(═N—CN)NR′R″, —NR′C(═N—CN)R″, —NR′C(═C—NO2)NR′R″, —SO2NR′COR″, —NO2, —CO2R′, —C(C═N—OR′)R″, —CR′═CR′R″, —CCR′, —S(C═O)(C═N—R′)R″, —SF5and —OCF3, wherein at least one W, X, Y, Z, G, G′, R, R′, R″, Q1-Q4, or A is modified to be covalently joined to a PTM, a chemical linking group (L), a ULM, CLM, or combination thereof;each of x, y, and z of Formulas (a1) through (e) are independently 0, 1, 2, 3, 4, 5, or 6;R′ and R″ of Formulas (a1) through (e) are independently selected from a bond, H, optionally substituted linear or branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclic, —C(═O)R, optionally substituted heterocyclyl;n′ of Formulas (a1) through (e) is an integer from 1-10 (e.g. 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);represents a single bond or a double bond; andof Formulas (a1) through (e) represents a bond that may be stereospecific ((R) or (S)) or non-stereospecific. In any aspect or embodiment described herein, the CLM comprises a chemical structure selected from the group consisting of: wherein:W of Formulas (a1) through (e) [e.g., (a1), (a2), (a3), (a4), (b), (c), (d1), (d2), and/or (e)] is independently selected from the group CH2, O, CHR, C═O, SO2, NH, N, optionally substituted cyclopropyl group, optionally substituted cyclobutyl group, and N-alkyl;W3of Formulas (a1) through (e) is selected from C or N;X of Formulas (a1) through (e) is independently selected from the group O, S and CH2;Y of Formulas (a1) through (e) is independently selected from the group CH2, —C═CR′, NH, N-alkyl, N-aryl, N-hetaryl, N-cycloalkyl, N-heterocyclyl, O, and S;Z of Formulas (a1) through (e) is independently selected from the group O, and S or CH2 except that both X and Z cannot be CH2or absent;G and G′ of Formulas (a1) through (e) are independently selected from the group H, optionally substituted linear or branched alkyl, OH, R′OCOOR, R′OCONRR″, CH2-heterocyclyl optionally substituted with R′, and benzyl optionally substituted with R′;Q1-Q4 of Formulas (a1) through (e) represent a carbon C or N substituted with a group independently selected from H, R, N or N-oxide;A of Formulas (a1) through (e) is independently selected from the group H, optionally substituted linear or branched alkyl, cycloalkyl, Cl and F;n of Formulas (a1) through (e) represent an integer from 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);R of Formulas (a1) through (e) comprises, but is not limited to: a bond, H, —C(═O)R′ (e.g., a carboxy group), —CONR′R″ (e.g., an amide group), —OR′ (e.g., OH), —NR′R″ (e.g. an amine group), —SR′, —SO2R′, —SO2NR′R″, —CR′R″—, —CR′NR′R″—, (—CR′O)nR″, optionally substituted aryl (e.g., an optionally substituted C5-C7 aryl), optionally substituted alkyl-aryl (e.g., an alkyl-aryl comprising at least one of an optionally substituted C1-C6 alkyl, an optionally substituted C5-C7 aryl, or combinations thereof), optionally substituted hetaryl, -optionally substituted linear or branched alkyl (e.g., a C1-C6 linear or branched alkyl optionally substituted with one or more halogen, cycloalkyl (e.g., a C3-C6 cycloalkyl), or aryl (e.g., C5-C7 aryl)), optionally substituted alkoxyl group (e.g., a methoxy, ethoxy, butoxy, propoxy, pentoxy, or hexoxy; wherein the alkoxyl may be substituted with one or more halogen, alkyl, haloalky, fluoroalkyl, cycloalkyl (e.g., a C3-C6 cycloalkyl), or aryl (e.g., C5-C7 aryl)), optionally substituted cycloalkyl, optionally substituted heterocyclyl, —P(O)(OR′)R″, —P(O)R′R″, OP(O)(OR′)R″, —OP(O)R′R″, —Cl, —F, —Br, —I, —CF3, —CN, —NR′SO2NR′R″, —NR′CONR′R″, —CONR′COR″, —NR′C(═N—CN)NR′R″, —C(═N—CN)NR′R″, —NR′C(═N—CN)R″, —NR′C(═C—NO2)NR′R″, —SO2NR′COR″, —NO2, —CO2R′, —C(C═N—OR′)R″, —CR′═CR′R″, —CCR′, —S(C═O)(C═N—R′)R″, —SF5 and —OCF3, wherein at least one of W, X, Y, Z, G, G′, R, R′, R″, Q1-Q4, or A is covalently joined (directly or indirectly, e.g., via a functional group or an atom, such as O, S, N) to a PTM, a chemical linking group (L), a ULM, CLM, or combination thereof;each of x, y, and z of Formulas (a1) through (e) are independently 0, 1, 2, 3, 4, 5, or 6;R′ and R″ of Formulas (a1) through (e) are independently selected from a bond, H, optionally substituted linear or branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclic, —C(═O)R, optionally substituted heterocyclyl;n′ of Formulas (a1) through (e) is an integer from 1-10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10); andof Formulas (a1) through (e) represents a bond that may be stereospecific ((R) or (S)) or non-stereospecific. In any aspect or embodiment described herein, the CLM or ULM is selected from the structure of Formula (g): wherein:W of Formula (g) is independently selected from the group CH2, O, C═O, NH, and N-alkyl;A of Formula (g) is selected from a H, methyl, or optionally substituted linear or branched alkyl;n is an integer from 1 to 4;R of Formula (g) is independently selected from a bond, H, O, OH, N, NH, NH2, Cl, —F, —Br, —I, methyl, optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl), optionally substituted linear or branched alkoxy (e.g., optionally substituted linear or branched C1-C6 alkoxy), -alkyl-aryl (e.g., an -alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxy), wherein at least one R or W is modified to be covalently joined to a PTM, a chemical linking group (L), a ULM, CLM, or combination thereof; andof Formula (g) represents a bond that may be stereospecific ((R) or (S)) or non-stereo specific. In any aspect or embodiment described herein, the CLM or ULM is selected from the group consisting of: wherein:W is C═O or CH2;N* is a nitrogen atom that is covalently linked to the PTM or linker, or that is shared with the PTM or linker (L) (e.g., a heteroatom shared with an optionally substituted heterocyclyl of the linker (L) or PTM); andindicates the point of attachment of the CLM or ULM to the linker (L) or PTM. In any aspect or embodiment described herein, R is selected from: H, O, OH, N, NH, NH2, C1-C6 alkyl, C1-C6 alkoxy, -alkyl-aryl (e.g., an -alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxy). In any aspect or embodiment described herein, at least one R (e.g. an R group selected from the following H, O, OH, N, NH, NH2, C1-C6 alkyl, C1-C6 alkoxy, -alkyl-aryl (e.g., an -alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxy) or W is modified to be covalently joined to a PTM, a chemical linker group (L), a ULM, a CLM, or a combination thereof In any aspect or embodiment described herein, the W, X, Y, Z, G, G′, R, R′, R″, Q1-Q4, and A of Formulas (a) through (g) can independently be covalently coupled to a linker and/or a linker to which is attached one or more PTM, ULM, or CLM groups. In any of the aspects or embodiments described herein, n is an integer from 1 to 4, and each R is independently selected functional groups or atoms, for example, O, OH, N, —Cl, —F, C1-C6 alkyl, C1-C6 alkoxy, -alkyl-aryl (e.g., an -alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxy, on the aryl or heteroaryl of the CLM, and optionally, one of which is modified to be covalently joined to a PTM, a chemical linker group (L), a ULM, CLM or combination thereof. More specifically, non-limiting examples of CLMs include those shown below as well as those “hybrid” molecules that arise from the combination of one or more of the different features shown in the molecules below wherein at least one R or W is modified to be covalently joined to a PTM, a chemical linking group (L), a ULM, CLM, or combination thereof. In any aspect or embodiment described herein, the CLM comprises a chemical structure selected from the group: wherein:W is independently selected from CH2, O, CHR, C═O, SO2, NH, N, optionally substituted cyclopropyl group, optionally substituted cyclobutyl group, and N-alkyl (e.g., CH2, CHR, C═O, SO2, NH, and N-alkyl);Q1, Q2, Q3, Q4, Q5are each independently represent a carbon C or N substituted with a group independently selected from R′, N or N-oxide;R1is selected from absent (i.e., a bond), H, OH, CN, C1-C3 alkyl, C═O;R2is selected from the group absent (i.e., a bond), H, OH, CN, C1-C3 alkyl, CHF2, CF3, CHO, C(═O)NH2;R3is selected from a bond, H, alkyl (e.g., C1-C6 or C1-C3 alkyl), substituted alkyl (e.g., substituted C1-C6 or C1-C3 alkyl), alkoxy (e.g., C1-C6 or C1-C3 alkoxyl), substituted alkoxy (e.g., substituted C1-C6 or C1-C3 alkoxyl);R4is selected from a bond, H, alkyl, substituted alkyl;R5and R6are each independently a bond, H, halogen, C(═O)R′, CN, OH, CF3;X is C, CH, C═O, or N;X1is C═O, N, CH, or CH2;R′ is selected from a bond, H, halogen, amine, alkyl (e.g., C1-C3 alkyl), substituted alkyl (e.g., substituted C1-C3 alkyl), alkoxy (e.g., C1-C3 alkoxyl), substituted alkoxy (e.g., substituted C1-C3 alkoxyl), NR2R3, C(═O)OR2, optionally substituted phenyl;n is 0-4;is a single or double bond; andthe CLM is covalently joined to a PTM, a chemical linker group (L), a ULM, CLM or combination thereof. In any aspect or embodiment described herein, the CLM is covalently joined to a PTM or a chemical linker group (L) via an R group (such as, R, R1, R2, R3, R4or R′), W, X, or a Q group (such as, Q1, Q2, Q3, Q4, or Q5). In any aspect or embodiment described herein, the CLM is covalently joined to a PTM or a chemical linker group (L) via W, X, R, R1, R2, R3, R4, R5, R′, Q1, Q2, Q3, Q4, and Q5. In any aspect or embodiment described herein, the W, X, R1, R2, R3, R4, R′, Q1, Q2, Q3, Q4, and Q5can independently be covalently coupled to a linker and/or a linker to which is attached to one or more PTM, ULM, CLM groups. More specifically, non-limiting examples of CLMs include those shown below as well as “hybrid” molecules or compounds that arise from combining one or more features of the following compounds: wherein:W is independently selected from the group CH2, CHR, C═O, SO2, NH, and N-alkyl;R1is selected from the group absent (i.e., a bond), H, CH, CN, C1-C3 alkyl;R2is selected from a bond, H or a C1-C3 alkyl;R3is selected from a bond, H, alkyl, substituted alkyl, alkoxy, substituted alkoxy;R4is selected from a bond, methyl or ethyl;R5is selected from a bond, H or halo;R6is selected from a bond, H or halo;n is an integer from 0-4;R and R′ are independently a bond, H, a functional group, or an atom (e.g., H, halogen (e.g., —Cl, —F), amine, C1-C3 alkyl, C1-C3 alkyl, C1-C3 alkoxyl, NR2R3, or C(═O)OR2); or an attachment point for a PTM or a chemical linker group (L);Q1and Q2are each independently C or N substituted with a group independently selected from H or C1-C3 alkyl;is a single or double bond. In any aspect or embodiment described herein, the W, R1, R2, Q1, Q2, Q3, Q4, R, and R′ can independently be covalently coupled to a linker and/or a linker to which is attached one or more PTM groups. In any aspect or embodiment described herein, the R1, R2, Q1, Q2, Q3, Q4, R, and R′ can independently be covalently coupled to a linker and/or a linker to which is attached one or more PTM groups. In any aspect or embodiment described herein, the Q1, Q2, Q3, Q4, R, and R′ can independently be covalently coupled to a linker and/or a linker to which is attached one or more PTM groups. In any aspect or embodiment described herein, R is modified to be covalently joined to the linker group (L), or a PTM, or combination thereof. In any aspect or embodiment described herein, the CLM is selected from: wherein R′ is a halogen and R1is as described herein. In any aspect or embodiment described herein, “CLM” can be an imide that binds to cereblon E3 ligase. These imides and linker attachment point can be, but not be limited to one of the following structures: In any aspect or embodiment described herein, the ULM is selected from the group consisting of: wherein:of the CLM indicates the point of attachment with a linker group or a PTM; andN* is a nitrogen atom that is shared with the chemical linker group or PTM In any aspect or embodiment described herein, the ULM is selected from the group consisting of: wherein:of the ULM indicates the point of attachment with a linker group or a PTM;N* is a nitrogen atom that is shared with the chemical linker group or PTM; andW, Q4, and Q5 are each defined as described in any aspect or embodiment described herein. Exemplary Tankers In any aspect or embodiment described herein, the compounds as described herein include a PTM chemically linked to a ULM (e.g., CLM) via a chemical linker (L). In certain embodiments, the linker group L comprises one or more covalently connected structural units (e.g., -AL1 . . .(AL)q- or -(AL)q-), wherein AL1is a group coupled to PTM, and (AL)qis a group coupled to ULM. In any aspect or embodiment described herein, the linker (L) to a ULM (e.g., CLM) connection is a stable L-ULM connection. For example, in certain embodiments, when a linker (L) and a ULM are connected via a heteroatom (e.g., N, O, S), any additional heteroatom, if present, is separated by at least a carbon atom (e.g., —CH2—), such as with an acetal or aminal group. By way of further example, in certain embodiments described herein, when a linker (L) and a ULM are connected via a heteroatom, the heteroatom is not part of an ester. In any aspect or embodiment described herein, the linker group L is a bond or a chemical linker group represented by the formula -(AL)q-, wherein A is a chemical moiety and q is an integer from 1-100 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80), and wherein L is covalently bound to both the PTM and the ULM, and provides for binding of the PTM to the protein target and the ULM to an E3 ubiquitin ligase to effectuate target protein ubiquitination. In any aspect or embodiment described herein, the linker group L is a bond or a chemical linker group represented by the formula -(AL)q-, wherein A is a chemical moiety and q is an integer from 6-30 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25), and wherein L is covalently bound to both the PTM and the ULM, and provides for binding of the PTM to the protein target and the ULM to an E3 ubiquitin ligase in sufficient proximity to result in target protein ubiquitination. In any aspect or embodiment described herein, the linker group L is -(AL)q-, wherein:(AL)qis a group which connects a ULM (e.g., CLM), to PTM;q of the linker is an integer greater than or equal to 1;each ALis independently selected from the group consisting of, a bond, CRL1RL2, O, S, SO, SO2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1═CRL2, C≡C, SiRL1RL2, P(O)RL1, P(O)ORL1, NRL3C(═NCN)NRL4, NRL3C(═NCN), NRL3C(═CNO2)NRL4, C3-11cycloalkyl optionally substituted with 1-6 RL1and/or RL2groups, C5-13spirocycloalkyl optionally substituted with 1-9 RL1and/or RL2groups, C3-11heterocyclyl optionally substituted with 1-6 RL1and/or RL2groups, C5-13spiroheterocyclyl optionally substituted with 1-8 RL1and/or RL2groups, aryl optionally substituted with 1-6 RL1and/or RL2groups, heteroaryl optionally substituted with 1-6 RL1and/or RL2groups, where RL1or RL2, each independently are optionally linked to other groups to form cycloalkyl and/or heterocyclyl moiety, optionally substituted with 1-4 R15groups; andRL1, RL2, RL3, RL4and RL5are, each independently, H, halo, C1-8alkyl, OC1-8alkyl, SC1-8alkyl, NHC1-8alkyl, N(C1-8alkyl)2, C3-11cycloalkyl, aryl, heteroaryl, C3-11heterocyclyl, OC3-8cycloalkyl, SC3-8cycloalkyl, NHC3-8cycloalkyl, N(C3-5cycloalkyl)2, N(C3-8cycloalkyl)(C1-8alkyl), OH, NH2, SH, SO2C1-8alkyl, P(O)(OC1-8alkyl)(C1-8alkyl), P(O)(OC1-8alkyl)2, CC—C1-8alkyl, CCH, CH═CH(C1-8alkyl), C(C1-8alkyl)═CH(C1-8alkyl), C(C1-8alkyl)═C(C1-8alkyl)2, Si(OH)3, Si(C1-8alkyl)3, Si(OH)(C1-8alkyl)2, COC1-8alkyl, CO2H, halogen, CN, CF3, CHF2, CH2F, NO2, SF5, SO2NHC1-8alkyl, SO2N(C1-8alkyl)2, SONHC1-8alkyl, SON(C1-8alkyl)2, CONHC1-8alkyl, CON(C1-8alkyl)2, N(C1-8alkyl)CONH(C1-8alkyl), N(C1-8alkyl)CON(C1-8alkyl)2, NHCONH(C1-8alkyl), NHCON(C1-8alkyl)2, NHCONH2, N(C1-8alkyl)SO2NH(C1-8alkyl), N(C1-8alkyl) SO2N(C1-8alkyl)2, NH SO2NH(C1-8alkyl), NH SO2N(C1-8alkyl)2, NH SO2NH2. In certain embodiments, q is an integer greater than or equal to 1. In certain embodiments, e.g., where q of the linker is greater than 2, (AL)qis a group which is AL1and (AL)qwherein the linker couples a PTM to a ULM. In certain embodiments, e.g., where q of the linker is 2, AL2is a group which is connected to AL1and to a ULM. In certain embodiments, e.g., where q of the linker is 1, the structure of the linker group L is -AL1-, and AL1is a group which connects a ULM moiety to a PTM moiety. In any aspect or embodiment described herein, the unit ALof linker (L) comprises a group represented by a general structure selected from the group consisting of:—NR(CH2)n-(lower alkyl)-, —NR(CH2)n-(lower alkoxyl)-, —NR(CH2)n-(lower alkoxyl)-OCH2—, —NR(CH2)n-(lower alkoxyl)-(lower alkyl)-OCH2—, —NR(CH2)n-(cycloalkyl)-(lower alkyl)-OCH2—, —NR(CH2)n-(heterocycloalkyl)-, —NR(CH2CH2O)n-(lower alkyl)-O—CH2—, —NR(CH2CH2O)n-(heterocycloalkyl)-O—CH2—, —NR(CH2CH2O)n-Aryl-O—CH2—, NR(CH2CH2O)n-(heteroaryl)-O—CH2—, —NR(CH2CH2O)n-(cyclo alkyl)-O-(heteroaryl)-O—CH2—, —NR(CH2CH2O)n-(cyclo alkyl)-O-Aryl-O—CH2—, —NR(CH2CH2O)n-(lower alkyl)-NH-Aryl-O—CH2—, —NR(CH2CH2O)n-(lower alkyl)-O-Aryl-CH2, —NR(CH2CH2O)n-cycloalkyl-O-Aryl-, —NR(CH2CH2O)n-cycloalkyl-O-(heteroaryl)l-, —NR(CH2CH2)n-(cycloalkyl)-O-(heterocyclyl)-CH2, —NR(CH2CH2)n-(heterocyclyl)-(heterocyclyl)-CH2, and —N(R1R2)-(heterocyclyl)-CH2; wheren of the linker can be 0 to 10;R of the linker can be H, or lower alkyl; andR1 and R2 of the linker can form a ring with the connecting N. In any aspect or embodiment described herein, the linker (L) includes an optionally substituted C1-C50alkyl (e.g., C1, C2, C3, C4, C5, C6, C7, C8, C9, C10, C11, C12, C13, C14, C15, C16, C17, C18, C19, C20, C21, C22, C23, C24, C25, C26, C27, C28, C29, C30, C31, C32, C33, C34, C35, C36, C37, C38, C39, C40, C41, C42, C43, Cn, C45, C46, C47, C48, C49, or C50alkyl, and including all implied subranges, e.g., C1-C10, C1-C20; C2-C10, C2-20; C10-C20, C10-C50 etc.), wherein each carbon is optionally independently substituted or replaced with (1) a heteroatom selected from N, O, S, P, or Si atoms that has an appropriate number of hydrogens, substitutions, or both to complete valency, (2) an optionally substituted cycloalkyl or bicyclic cycloalkyl, (3) an optionally substituted heterocyloalkyl or bicyclic heterocyloalkyl, (4) an optionally substituted aryl or bicyclic aryl, or (5) optionally substituted heteroaryl or bicyclic heteroaryl. In any aspect or embodiment described herein, the linker (L) does not have heteroatom-heteroatom bonding (e.g., no heteroatoms are covalently linked or adjacently located). In any aspect or embodiment described herein, the linker (L) includes an optionally substituted C1-C50alkyl (e.g., C1, C2, C3, C4, C5, C6, C7, C8, C9, C10, C11, C12, C13, C14, C15, C16, C17, C18, C19, C20, C21, C22, C23, C24, C25, C26, C27, C28, C29, C30, C31, C32, C33, C34, C35, C36, C37, C38, C39, C40, C41, C42, C43, C44, C45, C46, C47, C48, C49, or C50alkyl), wherein:each carbon is optionally independently substituted or replaced with CRL1RL2, O, S, SO, SO2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1═CRL2, C≡C, SiRL1RL2, P(O)RL1, P(O)ORL1, NRL3C(═NCN)NRL4, NRL3C(═NCN), NRL3C(═CNO2)NRL4, C3-11cycloalkyl optionally substituted with 1-6 RL1and/or RL2groups, C5-13spirocycloalkyl optionally substituted with 1-9 RL1and/or RL2groups, C3-11heterocyclyl optionally substituted with 1-6 RL1and/or RL2groups, C5-13spiroheterocyclyl optionally substituted with 1-8 RL1and/or RL2groups, aryl optionally substituted with 1-6 RL1and/or RL2groups, heteroaryl optionally substituted with 1-6 RL1and/or RL2groups, where RL1or RL2, each independently are optionally linked to other groups to form a cycloalkyl and/or a heterocyclyl moiety, optionally substituted with 1-4 RL5groups; andRL1, RL2, RL3, RL4and RL5are, each independently, H, halo, C1-8alkyl, OC1-8alkyl, SC1-8alkyl, NHC1-8alkyl, N(C1-5alkyl)2, C3-11cycloalkyl, aryl, heteroaryl, C3-11heterocyclyl, OC3-8cycloalkyl, SC3-8cycloalkyl, NHC3-8cycloalkyl, N(C3-5cycloalkyl)2, N(C3-8cycloalkyl)(C1-8alkyl), OH, NH2, SH, SO2C1-8alkyl, P(O)(OC1-8alkyl)(C1-8alkyl), P(O)(OC1-8alkyl)2, CC—C1-8alkyl, CCH, CH═CH(C1-8alkyl), C(C1-8alkyl)═CH(C1-8alkyl), C(C1-8alkyl)═C(C1-8alkyl)2, Si(OH)3, Si(C1-8alkyl)3, Si(OH)(C1-8alkyl)2, COC1-8alkyl, CO2H, halogen, CN, CF3, CHF2, CH2F, NO2, SF5, SO2NHC1-8alkyl, SO2N(C1-8alkyl)2, SONHC1-8alkyl, SON(C1-8alkyl)2, CONHC1-8alkyl, CON(C1-8alkyl)2, N(C1-8alkyl)CONH(C1-8alkyl), N(C1-8alkyl)CON(C1-8alkyl)2, NHCONH(C1-8alkyl), NHCON(C1-8alkyl)2, NHCONH2, N(C1-8alkyl)SO2NH(C1-8alkyl), N(C1-8alkyl) SO2N(C1-8alkyl)2, NH SO2NH(C1-8alkyl), NH SO2N(C1-8alkyl)2, NH SO2NH2. In any aspect or embodiment described herein, the linker group is optionally substituted an optionally substituted C1-C50alkyl (e.g., C1, C2, C3, C4, C5, C6, C7, C8, C9, C10, C11, C12, C13, C14, C15, C16, C17, C18, C19, C20, C21, C22, C23, C24, C25, C26, C27, C28, C29, C30, C31, C32, C33, C34, C35, C36, C37, C38, C39, C40, C41, C42, C43, C44, C45, C46, C47, C48, C49, or C50alkyl, and including all implied subranges, e.g., C1-C10, C1-C20; C2-C10, C2-20; C10-C20, C10-C50 etc.), wherein each carbon atom optionally substituted or replaced with: a O, N, S, P or Si atom that has an appropriate number of hydrogens, substitutions (e.g., OH, halo, alkyl, methyl, ethyl, haloalkyl, hydroxyalkyl, alkoxy, methoxy, etc.), or both to complete valency; an optionally substituted aryl (e.g., an optionally substituted C5 or C6 aryl) or bicyclic aryl (e.g, an optionally substituted C5-C20 bicyclic heteroaryl); an optionally substituted heteroaryl (e.g., an optionally substituted C5 or C6 heteroaryl) or bicyclic heteroaryl (e.g., an optionally substituted heteroaryl or bicyclic heteroaryl having one or more heteroatoms selected from N, O, S, P, and Si that has an appropriate number of hydrogens, substitutions (e.g., OH, halo, alkyl, methyl, ethyl, haloalkyl, hydroxyalkyl, alkoxy, methoxy, etc.), or both to complete valency); an optionally substituted C1-C6 alkyl; an optionally substituted C1-C6 alkenyl; an optionally substituted C1-C6 alkynyl; an optionally substituted cycloalkyl (e.g., an optionally substituted C3-C7 cycloalkyl) or bicyclic cycloalkyl (e.g., an optionally substituted C5-C20 bicyclic cycloalkyl); or an optionally substituted heterocycloalkyl (e.g., an optionally substituted 3-, 4-, 5-, 6-, or 7-membered heterocyclic group) or bicyclicheteroalkyl (e.g., an optionally substituted heterocyclo alkyl bicyclicheteroalkyl having one or more heteroatoms selected from N, O, S, P, or Si atoms that has an appropriate number of hydrogens, substitutions (e.g., OH, halo, alkyl, methyl, ethyl, haloalkyl, hydroxyalkyl, alkoxy, methoxy, etc.), or both to complete valency). In any aspect or embodiment described herein, the optionally substituted alkyl linker is optionally substituted with one or more OH, halo, linear or branched C1-C6 alkyl (such as methyl or ethyl), linear or branched C1-C6 haloalkyl, linear or branched C1-C6 hydroxyalkyl, or linear or branched C1-C6 alkoxy (e.g., methoxy). In any aspect or embodiment described herein, the linker (L) does not have heteroatom-heteroatom bonding (e.g., no heteroatoms are covalently linked or adjacently located). In any aspect or embodiment described herein, the linker (L) includes about 1 to about 50 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50) alkylene glycol units that are optionally substituted, wherein carbon or oxygen may be substituted with a heteroatom selected from N, S, P, or Si atoms with an appropriate number of hydrogens to complete valency. In any aspect or embodiment described herein, the L is selected from the group consisting of: wherein:N* is a nitrogen atom that is covalently linked to the CLM or PTM, or that is shared with the CLM or PTM;each m, n, o, and p is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; andthe chemical linker group is optionally substituted with 1, 2, 3, or 4 substitutions independently selected from a halogen (e.g., F or Cl) and a C1-4alkyl In any aspect or embodiment described herein, the until ALof the linker (L) comprises a structure selected from the group consisting of: wherein N* is a nitrogen atom that is covalently linked to the ULM or PTM, or that is shared with the ULM or PTM. In any aspect or embodiment described herein, the until ALof the linker (L) comprises a structure selected from the group consisting of: wherein N* is a nitrogen atom that is covalently linked to the ULM or PTM, or that is shared with the ULM or PTM. In any aspect or embodiment described herein, the unit ALof the linker (L) comprises a structure selected from the group consisting of: wherein:N* is a nitrogen atom that is covalently linked to the ULM or PTM, or that is shared with the ULM or PTM; andeach m, n, o, p, q, and r is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20. In any aspect or embodiment described herein, the unit ALof the linker (L) is selected from: wherein dashed line or * indicates the site that is covalently linked to the ULM or PTM, or that is shared with the ULM or PTM. In any aspect or embodiment described herein, the unit ALof the linker (L) is selected from: wherein dashed line or * indicates the site that is covalently linked to the CLM or the PTM, or that is shared with the CLM or the PTM. In any aspect or embodiment described herein, the unit ALof linker (L) comprises a group represented by a general structure selected from the group consisting of: wherein m, n, o, p, q, and r of the linker are independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; when m, n, o, p, q, and r are zero, N—O or O—O bond is absent, X of the linker is H or F where each n and m of the linker can independently be 0, 1, 2, 3, 4, 5, or 6. In any aspect or embodiment described herein, the unit ALof linker (L) is selected from the group consisting of: wherein each m and n is independently selected from 0, 1, 2, 3, 4, 5, or 6. In any aspect or embodiment described herein, the unit ALof linker (L) is selected from the group consisting of: wherein each m, n, o, p, q, r, and s is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20. In any aspect or embodiment described herein, the unit ALof linker (L) is selected from the group consisting of: In any aspect or embodiment described herein, the linker (L) comprises a structure selected from the structure shown below: wherein:WL1and WL2are each independently absent, a 4-8 membered ring with 0-4 heteroatoms, optionally substituted with RQ, each RQis independently a H, halo, OH, CN, CF3, optionally substituted linear or branched C1-C6alkyl, optionally substituted linear or branched C1-C6alkoxy, or 2 RQgroups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms;YL1is each independently a bond, optionally substituted linear or branched C1-C6alkyl and optionally one or more C atoms are replaced with O or NRYL1, optionally substituted C1-C6alkene and optionally one or more C atoms are replaced with O, optionally substituted C1-C6alkyne, and optionally one or more C atoms are replaced with O, or optionally substituted linear or branched C1-C6alkoxy;RYL1is H, or optionally substituted linear or branched C1-6alkyl;n is 0-10; andandindicates the attachment point to the PTM or ULM moieties. In any aspect or embodiment described herein, the linker (L) comprises a structure selected from the structure shown below: wherein:WL1and WL2are each independently absent, piperazine, piperidine, morpholine, optionally substituted with RQ, each RQis independently a H, —Cl—, —F—, OH, CN, CF3, optionally substituted linear or branched C1-C6alkyl (e.g. methyl, ethyl), optionally substituted linear or branched C1-C6alkoxy (e.g. methoxy, ethoxy);YL1is each independently a bond, optionally substituted linear or branched C1-C6alkyl and optionally one or more C atoms are replaced with O or NRYL1; optionally substituted C1-C6alkene and optionally one or more C atoms are replaced with O, optionally substituted C1-C6alkyne and optionally one or more C atoms are replaced with O, or optionally substituted linear or branched C1-C6alkoxy;RYL1is H, or optionally substituted linear or branched C1-6alkyl (e.g. methyl, ethyl);n is 0-10; andandindicates the attachment point to the PTM or ULM moieties. In any aspect or embodiment described herein, the linker (L) comprises a structure selected from the structure shown below: wherein:WL1and WL2are each independently absent, aryl, heteroaryl, cyclic, heterocyclic, C1-6alkyl and optionally one or more C atoms are replaced with O or NRYL1, C1-6alkene and optionally one or more C atoms are replaced with O, C1-6alkyne and optionally one or more C atoms are replaced with O, bicyclic, biaryl, biheteroaryl, or biheterocyclic, each optionally substituted with RQ, each RQis independently a H, halo, OH, CN, CF3, hydroxyl, nitro, C≡CH, C2-6alkenyl, C2-6alkynyl, optionally substituted linear or branched C1-C6alkyl, optionally substituted linear or branched C1-C6alkoxy, optionally substituted OC1-3alkyl (e.g., optionally substituted by 1 or more —F), OH, NH2, NRY1RY2, CN, or 2 RQgroups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms;YL1is each independently a bond, NRYL1, O, S, NRYL2, CRYL1RYL2, C═O, C═S, SO, SO2, optionally substituted linear or branched C1-C6alkyl and optionally one or more C atoms are replaced with O; optionally substituted linear or branched C1-C6alkoxy;QLis a 3-6 membered alicyclic, bicyclic, or aromatic ring with 0-4 heteroatoms, optionally bridged, optionally substituted with 0-6 RQ, each RQis independently H, optionally substitute linear or branched C1-6alkyl (e.g., optionally substituted by 1 or more halo, C1-6alkoxyl), or 2 RQgroups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms;RYL1, RYL2are each independently H, OH, optionally substituted linear or branched C1-6alkyl (e.g., optionally substituted by 1 or more halo, C1-6alkoxyl), or R1, R2together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms;n is 0-10; andandindicates the attachment point to the PTM or ULM moieties. In any aspect or embodiment described herein, the linker (L) comprises a structure selected from the structure shown below: wherein:WL1and WL2are each independently absent, cyclohexane, cyclopentane, piperazine, piperidine, morpholine, C1-6alkyl and optionally one or more C atoms are replaced with O or NRYL1, C1-6alkene and optionally one or more C atoms are replaced with O, or C1-6alkyne and optionally one or more C atoms are replaced with O, each optionally substituted with Rq, each RQis independently a H, Cl—, —F—, OH, CN, CF3, hydroxyl, optionally substituted linear or branched C1-C6alkyl (e.g. methyl, ethyl), optionally substituted linear or branched C1-C6alkoxy;YL1is each independently a bond, NRYL1, O, CRYL1RYL2, C═O, optionally substituted linear or branched C1-C6alkyl and optionally one or more C atoms are replaced with O or NRYL1, C1-6alkene and optionally one or more C atoms are replaced with O, C1-6alkyne and optionally one or more C atoms are replaced with O, or optionally substituted linear or branched C1-C6alkoxy;QLis a 3-6 membered heterocyclic, heterobicyclic, or heteroaryl ring, optionally substituted with 0-6 Rq, each RQis independently H, or optionally substituted linear or branched C1-6alkyl (e.g., methyl or ethyl, optionally substituted by 1 or more halo, C1-6alkoxyl);RYL1, RYL2are each independently H, optionally substituted linear or branched C1-6alkyl (e.g., optionally substituted by 1 or more halo, C1-6alkoxyl);n is 0-10; andandindicates the attachment point to the PTM or ULM moieties. Exemplary PTMs The term “protein target moiety” or PTM is used to describe a small molecule which binds to LRRK2, and can be used to target the PTM for ubiquitination and degradation. The compositions described below exemplify members of LRRK2 binding moieties that can be used according to the present invention. These binding moieties are linked to the ubiquitin ligase binding moiety preferably through a chemical linking group in order to present the LRRK2 protein in proximity to the ubiquitin ligase for ubiquitination and subsequent degradation. In certain contexts, the term “target protein” is used to refer to the LRRK2 protein, a member of the ROCO protein family, that serves as an upstream central integrator of multiple signaling pathways that are crucial for proper neuronal functioning, which is a target protein to be ubiquitinated and degraded. The compositions described herein exemplify the use of some of the members of these types of small molecule target protein binding moieties. In any aspect or embodiment described herein, the PTM is a small molecule that binds LRRK2. For example, in any aspect or embodiment described herein, the PTM is represented by the chemical structure PTM-I: wherein:X is CH or N;W is O or S;Q is selected from the group: wherein:A1, A2, and A3are each independently selected from N and CR6, wherein for (a) no more than two of A1, A2, and A3are simultaneously N, ring B is an optionally substituted C3-C8 aryl, optionally substituted C3-C8 heteroaryl, optionally substituted C3-C8 cycloalkyl, optionally substituted C3-C8 heterocycloalkyl, optionally substituted C3-C8 aryl, optionally substituted C3-C8 aryl,theof Q indicates the point of attachment with the nitrogen, andtheof Q indicates an optional covalent attachment to RPTM;R2is H or C1-4alkyl;R3Aand R3Bare each independently selected from H, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NRe1)NRc1Rd1, NRc1(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1; wherein said C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, and 4-10 membered heterocycloalkyl-C1-4alkyl of R1are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Cy2, Cy2-C1-4alkyl, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1;or R3Aand R3Btogether form a C3-7cycloalkyl or 4-10 membered heterocycloalkyl ring, each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Cy2, Cy2-C1-4alkyl, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1;R4is H, C1-4alkyl, halo, C1-4haloalkyl, or CN;R5is H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl —C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRc2Rd2; wherein said C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, and 4-10 membered heterocycloalkyl-C1-4alkyl of R1are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Cy3, Cy3-C1-4alkyl, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, or S(O)2NRc2Rd2;each R6is independently selected from H, halo, C1-6alkyl, C1-6haloalkyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, CN, NO2, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, OC(O)Rb3, OC(O)NRc3Rd3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)NRc3Rd3, NRc3C(O)ORa3, C(═NRe3)NRc3Rd3, NRc3C(═NRe3)NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, and S(O)2NRc3Rd3, wherein said C1-6alkyl, C1-6haloalkyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl of R6are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, OC(O)Rb3, OC(O)NRc3Rd3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)NRc3Rd3, NRc3C(O)ORa3, C(═NRe3)NRc3Rd3, NRc3C(═NRe3)NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, and S(O)2NRc3Rd3;each Cy1is independently selected from C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, and 4-14 membered heterocycloalkyl, each optionally substituted by 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa, SRa, C(O)Rb, C(O)NRcRd, C(O)ORa, OC(O)Rb, OC(O)NRcRd, NRcRd, NRcC(O)Rb, NRcC(O)ORa, NRcC(O)NRcRd, C(═NRe)Rb, C(═NRe)NRcRd, NRcC(═NRe)NRcRd, NRcS(O)Rb, NRcS(O)2Rb, NRcS(O)2NRcRd, S(O)Rb, S(O)NRcRd, S(O)2Rb, and S(O)2NRcRd;each Cy2is independently selected from C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, and 4-14 membered heterocycloalkyl, each optionally substituted by 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1;each Cy3is independently selected from C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, and 4-14 membered heterocycloalkyl, each optionally substituted by 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRc2Rd2;each Ra, Rb, Rc, Rd, Ra1, Rb1, Rc1, Rd1, Ra2, Rb2, Rc2, Rd2, Ra3, Rb3, Rc3, and Rd3is independently selected from H, C1-6alkyl, C1-6haloalkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-7cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, and 4-10 membered heterocycloalkyl-C1-4alkyl, wherein said C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-7cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, and 4-10 membered heterocycloalkyl-C1-4alkyl of Ra, Rb, Rc, Rd, Ra1, Rb1, Rc1, Rd1, Ra2, Rb2, Rc2, Rd2, RaRb3, Rc3, or Rd3is optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, Ci-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, CN, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, OC(O)Rb4, OC(O)NRc4Rd4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)NRc4Rd4, NRc4C(O)ORa4, C(═NRe4)NRc4Rd4, NRc4C(═NRe4)NRc4Rd4, S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, NRc4S(O)2Rm, NRc4S(O)2NRc4Rd4, and S(O)2NRc4Rd4;each Ra4, Rb4, Rc4, and Rd4are independently selected from H, C1-6alkyl, C1-6haloalkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, wherein said C1-6alkyl, C1-6haloalkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl are each optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino, halo, C1-6alkyl, C1-6alkoxy, C1-6haloalkyl, and C1-6haloalkoxy;each Re, Re1, Re2, Re3, and Re4is independently selected from H, C1-4alkyl, and CN;RPTMis H; halogen (e.g., Cl or F); —CN; —OH; —NO2; —NH2; optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl or optionally substituted linear or branched C1-C4 alkyl or C1-C8 alkyl optionally substituted with OH); optionally substituted cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl); O-optionally substituted linear or branched C1-C4 alkyl; an optionally substituted C1-C4 alkynyl; an optionally substituted C1-C4 alkyne; optionally substituted linear or branched hydroxyalkyl (e.g., optionally substituted linear or branched C1-C7 hydroxy alkyl); optionally substituted alkylcycloalkyl (e.g., includes optionally substituted C1-C6 alkyl, optionally substituted C3-C10 cycloalkyl; or both); optionally substituted alkyl-aryl (e.g., includes an optionally substituted linear or branched C1-C6 alkyl, an optionally substituted 5-10 member heteroaryl, or both); optionally substituted alkyl-heteroaryl (e.g., includes an optionally substituted linear or branched C1-C6 alkyl, an optionally substituted 5-10 member heteroaryl, or both); optionally substituted alkyl-heteroaryl (e.g., includes a C1-C6 alkyl, an optionally substituted 5 or 6 member heteroaryl, optionally substituted with a C1-C4 alkyl; the heteroaryl is selected from oxazol-4-yl, 1,3,4-triazol-2-yl, and imidazole-1-yl; or combination thereof); optionally substituted —NH-alkyl-heteroaryl (e.g., an optionally substituted linear or branched C1-C5 alkyl, an optionally substituted 5-8 member heteroaryl, optionally substituted with a C1-C4 alkyl, N—CH2-pyrazol-4-yl, or a combination thereof); optionally substituted alkoxy (e.g., an optionally substituted linear or branched C1-C6 alkyl or —OCH3); optionally substituted O-heterocyclyl (e.g., includes an optionally substituted 3-12 or 4-7 member heterocyclyl; an optionally substituted heterocycloalkyl; an optionally substituted C3-12monocyclic or bicyclic heterocycloalkyl; optionally substituted with at least one OH, C1-C5 alkyl (such as a methyl), ═O, NH2, or a combination thereof; or a combination thereof); optionally substituted S-heterocyclyl (e.g., includes an optionally substituted 4-7 member heterocyclyl; an optionally substituted heterocycloalkyl; optionally substituted with at least one C1-C4 alkyl (such as a methyl), ═O, or a combination thereof; or a combination thereof); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —(CH2)uCO(CH2)vCH3, —COCH3, or —CH2CH2COCH3, wherein each u and v is independently selected from 1, 2, 3, 4 or 5);optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —O(CH2)uCO(CH2)vCH3, —O(CH2)uCH((CH2)xCH3)(CH2)wCO(CH2)vCH3, —O—CH2COCH3, —O—CH2COCH2CH3, —O—CH(CH3)COCH3, —OCH2COCH3, or —OCH2(CH3)COCH3, wherein each u, v, w, and * is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —(CH2)uCO(CH2)vNRPTM1aRPTM2a, —CONRPTM1aRPTM2a, —CH2CONRPTM1aRPTM2a, —CH2CH2CONRPTM1aRPTM2a, —CONHCH3, or —CH2CONHCH3, wherein each u and v is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —O(CH2)uCO(CH2)vNRPTM1aRPTM2a, —O(CH2)uCH((CH2)xCH3)(CH2)wCO(CH2)vNRPTM1aRPTM2a, —O—CH(CH3)CONRPTM1aRPTM2a, —O—CH2CONRPTM1aRPTM2a, or —OCH2C(O)NHOCH3, wherein each u, v, w, and x is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —(CH2)uCHCH(CH2)wCO(CH2)vNRPTM1aRPTM2aor —CHCHCONRPTM1aRPTM2a, wherein each u, v, and w is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —NH—(CH2)uCO(CH2)vNRPTM1aRPTM2aor —NH—CH2CONRPTM1aRPTM2a, wherein each u and v is independently selected from 1, 2, 3, 4 or 5); fluoroalkoxy (e.g., a mono-, bi- and/or tri-fluoroalkoxy); optionally substituted monocyclic or bicyclic cycloalkyl (e.g., an optionally substituted 3-12 member cycloalkyl; optionally substituted with at least one of OH, ═O, linear or branched C1-C6 alkyl (such as a methyl, ethyl, or butyl), or NH2; or a combination thereof); optionally substituted hydroxycycloalkyl; optionally substituted aryl (e.g., an optionally substitute C5-C10 aryl, an optionally substituted 5-7 member aryl; optionally substituted with at least one halogen or C1-C3 alkyl (e.g., methyl or ethyl); or a combination thereof), optionally substituted heteroaryl (e.g., an optionally substituted 5-10 or member heteroaryl, an optionally substituted 5-7 member heteroaryl; an optionally substituted 5-member heteroaryl; optionally substituted with at least one halogen or C1-C3 alkyl (e.g., methyl or ethyl); or a combination thereof) optionally linked to Q via a C or N-atom of the heteroaryl (e.g., at least one of optionally linked to Q, optionally linked via an optionally substituted —(CH2)uO(CH2)vO(CH2)x—, or a combination thereof); optionally substituted monocyclic or bicyclic heterocyclyl (e.g., an optionally substituted 3-12 member heterocyclyl; an C3-C12 monocyclic or bicyclic heterocycloalkyl, azetidine1-yl, pyrrolidin-1-yl, piperidin-1yl, piperazin-1-yl, or morpholin-4-yl, or homopiperazin-1-yl, each optionally substituted with OH, a linear or branched C1-C5 alkyl (a methyl, ethyl, or butyl group) or NH2) optionally linked to Q via a C or N atom of the heterocyclyl (e.g., at least one of optionally linked to Q, optionally linked via an optionally substituted —(CH2)uO(CH2)vO(CH2)x—, or both);t1is selected from 1, 2, 3, 4, or 5;each t2is independently is independently selected from 0, 1, 2, 3, 4, or 5;RPTM1aand RPTM2aare independently H, optionally substituted C1-C4 alkyl (e.g., a CH3or CH2CH3), optionally substituted C1-C4 alkoxy (e.g., —OCH2or —CH2CH3), optionally substituted CH2OCH3or RPTM1aand RPTM2aare joined together form an optionally substituted 3-10 member ring;n is an integer from 0 to 10; andof the PTM indicates the point of attachment with a chemical linker group or a ULM. In any of the aspects or embodiments described herein, RPTMof PTM-I is modified to be covalently linked to a linker group (L) or a ULM (such as CLM). In any aspect or embodiment described herein, RPTMof PTM-I is modified to be covalently linked to a chemical linker group (L) or a ULM. In any aspect or embodiment described herein, W of PTM-I is O. In any aspect or embodiment described herein, Q of PTM-I is group (a) and ring B is selected from: whereinR1, R1A, R1B, R1Cand R1Dare each independently selected from H, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, and ORa, SRa, C(O)Rb, C(O)NRcRd, C(O)ORa, OC(O)Rb, OC(O)NRcRd, NRcRd, NRcC(O)Rb, NRcC(O)ORa, NRcC(O)NRcRd, C(═NRe)Rb, C(═NRe)NRcRd, NRcC(═NRe)NRcRd, NRcS(O)Rb, NRcS(O)2Rb, NRcS(O)2NRcRd, S(O)Rb, S(O)NRcRd, S(O)2Rb, S(O)2NRcRd, and RPTM, wherein said C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, wherein one of R1, R1A, R1B, R1C, and R1Dis optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Cy1, Cy1-C1-4alkyl, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa, SRa, C(O)Rb, C(O)NRcRd, C(O)ORa, OC(O)Rb, OC(O)NRcRd, NRcRd, NRcC(O)Rb, NRcC(O)ORa, NRcC(O)NRcRd, C(═NRe)Rb, C(═NRe)NRcRd, NRcC(═NRe)NRcRd, NRcS(O)Rb, NRcS(O)2Rb, NRcS(O)2NRcRd, S(O)Rb, S(O)NRcRd, S(O)2Rb, and S(O)2NRcRd;or R1Aand R1Btogether form a C3-7cycloalkyl or 4-10 membered heterocycloalkyl ring, each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Cy1, Cy1C1-4alkyl, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa, SRa, C(O)Rb, C(O)NRcRd, C(O)ORa, OC(O)Rb, OC(O)NRcRd, NRcRd, NRcC(O)Rb, NRcC(O)ORa, NRcC(O)NRcRd, C(═NRe)Rb, C(═NRe)NRcRd, NRcC(═NRe)NRcRd, NRcS(O)Rb, NRcS(O)2Rb, NRcS(O)2NRcRd, S(O)Rb, S(O)NRcRd, S(O)2Rb, S(O)2NRcRd, and RPTM, wherein ring B or a substituent of ring B is covalently coupled to at least one of RPTM, a linking group (L) or a CLM. In any aspect or embodiment described herein, Q if PTM-I is group (a) and ring B is selected from: wherein RPTMis as described herein, and theindicates a covalent attachment to a linking group (L) or CLM. In any of the aspects or embodiments, the Q of PTM-I is covalently coupled to the linker (L) or CLM. In any aspect or embodiment described herein, Q of PTM-I is group (a): wherein A1, A2, and A3are CR6; R6is independently selected from H, halo, C1-6alkyl, C1-6haloalkyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, CN, and NO2; ring B is a group as described herein, theof Q indicates the point of attachment with the nitrogen, and theof Q indicates the site of attachment to at least one of RPTM, a chemical linking group (L) or CLM. In any of the aspects or embodiments, the Q of PTM-I is covalently coupled to a chemical linking group (L) or CLM. In any aspect or embodiment described herein, Q of PTM-I is: wherein A1, A2and A3are defined in any aspect or embodiment described herein, theof Q indicates the point of attachment with the nitrogen, and theindicates the site of covalent attachment to at least one of RPTM, a chemical linking group (L) or CLM. In any of the aspects or embodiments, the Q of PTM-I is covalently coupled to the linker (L) or CLM. In any aspect or embodiment described herein, Q of PTM-I is: wherein A1, A2, and A3are defined in any aspect or embodiment described herein, theof Q indicates the point of attachment with the nitrogen, and theindicates the site of covalent attachment to at least one of RPTM, a chemical linking group (L) or CLM. In any of the aspects or embodiments, the Q of PTM-I is covalently coupled to the linker (L) or CLM. In any aspect or embodiment described herein, Q of PTM-I is: wherein:A1and A2are each CR6;each R6is independently selected from H, halo, C1-6alkyl, C1-6haloalkyl, Ono aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, CN, and NO2;theof Q indicates the point of attachment with the nitrogen; andtheindicates a covalent attachment to at least one of RPTM, a chemical linking group (L) or CLM. In any aspect or embodiment described herein, W PTM-I is O. In any aspect or embodiment described herein, R2PTM-I is H. In any aspect or embodiment described herein, R5of PTM-I is H or C1-6alkyl. In any aspect or embodiment described herein, R3Aand R3Bof PTM-I are each independently selected from H, halo, and C1-6alkyl. In any aspect or embodiment described herein, RPTMis selected from a bond, methyl, ethyl, propyl, butyl, pentyl, hexyl, H, O H, ethyl, whereinrepresents the site of attachment with PTM and the orindicates the site of covalent attachment with a chemical linking group (L) or CLM. In any of the aspects or embodiments, the RPTMis covalently coupled to the linker (L) or CLM. In any aspect or embodiment described herein, the PTM-I is selected from the group consisting of: wherein RPTMis a bond, an atom or chemical group as described herein, and theof the RPTMindicates the point of attachment of a chemical linking group (L) or a CLM. In any aspect or embodiment described herein, RPTMis a 5-7 membered aryl or heteroaryl (e.g., a 6-membered aryl or heteroaryl), wherein:the heteroaryl has 1, 2, or 3 heteroatoms selected from O, N, and S; andthe RPTMis optionally substituted with (a) Cy1or (b) a 3- to 10-membered cycloalkyl (e.g., 5- or 6-membered cycloalkyl), heterocycloalkyl (e.g., 5- or 6-membered heterocycloalkyl), spirocycloalky (e.g., 6- to 12-membered or 8- to 10-membered spirocycloalkyl), spiroheterocycloalkyl (e.g., 6- to 12-membered or 8- to 10-membered spiroheterocycloalkyl), bicyclic cycloalkyl (e.g., 6- to 10-membered bicyclic cycloalkyl), or bicyclic hetercycloalkyl (5- to 10-membered bicyclic heterocycloalkyl), wherein each of these is optionally substituted with 1, 2, 3, or 4 groups selected from the group consisting of H, C1-3alkyl, methyl, ethyl, and halogen. In any aspect or embodiment described herein, RPTMis a 5-7 membered aryl or heteroaryl (e.g., a 6-membered aryl or heteroaryl), wherein:the heteroaryl has 1, 2, or 3 heteroatoms selected from O, N, and S; andthe RPTMis optionally substituted with a 3- to 10-membered cycloalkyl (e.g., 5- or 6-membered cycloalkyl), heterocycloalkyl (e.g., 5- or 6-membered heterocycloalkyl), spirocycloalky (e.g., 6- to 12-membered spirocycloalkyl), spiroheterocycloalkyl (e.g., 6- to 12-membered spiroheterocycloalkyl), bicyclic cycloalkyl (e.g., 6- to 10-membered bicyclic cycloalkyl), or bicyclic hetercycloalkyl (5- to 10-membered bicyclic heterocycloalkyl), wherein each of these is optionally substituted with 1, 2, 3, or 4 groups selected from the group consisting of H, C1-3alkyl, methyl, ethyl, and halogen. In any aspect or embodiment described herein RPTM1aor RPTM2ais selected from: a bond, methyl, ethyl, propyl, butyl, pentyl, hexyl, H, O H, ethyl, whereinrepresents the site of attachment with PTM and theor * indicates the site of covalent attachment with a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the RPTMor the corresponding location of any PTM described herein (e.g. PTM-I and derivatives thereof) is a linear or branched C1-C8 alkyl optionally substituted with OH. In any aspect or embodiment described herein, the RPTMor the corresponding location of any PTM described herein (e.g. PTM-I derivatives thereof) is H, OH, CN, optionally substituted linear or branched C1-C4 alkyl, O-optionally substituted linear or branched C1-C4 alkyl, an optionally substituted C1-C4 alkynyl, an optionally substituted C1-C4 alkyne, an optionally substituted monocyclic or bicyclic C3-C12 heterocyclyl (e.g., an optionally substituted C3-C12 monocyclic or bicyclic heterocycloalkyl, such as an C3-C12 monocyclic or bicyclic heterocycloalkyl, azetidine1-yl, pyrrolidin-1-yl, piperidin-1yl, piperazin-1-yl, or morpholin-4-yl, or homopiperazin-1-yl, each optionally substituted with one or more of OH, a linear or branched C1-C5 alkyl or NH2), or an optionally substituted —O—C3-12monocyclic or bicyclic heterocyclyl (e.g., an optionally substituted —O—C3-12monocyclic or bicyclic heterocycloalkyl, such as —O—C3-12monocyclic or bicyclic heterocycloalkyl optionally substituted with at least one OH, a linear or branched C1-C5 alkyl or NH2), or an optionally substituted C3-C12 member ring (e.g., an optionally substituted C3-C12 non-aryl membered ring optionally substituted with one or more of OH, linear or branched C1-C5 alkyl, or NH2), wherein when RPTMis a ring structure it is optionally covalently linked to Q16via a C or N of the RPTMring. In any aspect or embodiment described herein, the PTM is represented by the chemical structure: wherein X, A1, A2, A3, R2, R3A, R3B, R4, R5, and RPTMare defined as described in any aspect or embodiment herein, and theindicates a covalent attachment to a chemical linking group (L) or CLM In any aspect or embodiment described herein, the PTM is represented by the chemical structure: wherein R2, R3A, R3B, R4, R5, and RPTMare defined as in any aspect or embodiment described herein, and theindicates a covalent attachment to a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is represented by a chemical structure selected from: wherein theindicates a covalent attachment of a chemical linking group (L) or CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) is selected from: wherein theof the PTM indicates the site of attachment of a chemical linking group (L) or a CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) has the chemical structure: and wherein theof the PTM indicates the covalent attachment of a chemical linking group or a CLM. In any aspect or embodiment described herein, the PTM (for e.g. PTM-I, PTM-IIa, PTM-IIb, PTM-IIIa, or PTM-IIIb, or the PTM of Formulas IIIa-IIIh and IVa-IVh) has the chemical structure: wherein theindicates a site of attachment of a linker or ULM, and wherein each PTM is coupled to at least one linker or ULM In any aspect or embodiment described herein, the hetero-bifunctional compound is represented by the chemical structure: wherein: R3A, R3B, R4and R5are defined as described herein;X is CH or N;R7is H, C1-C6 alkyl, C1-C6 alkoxy, or halogen;A is a 4-7 membered aryl, heteroaryl, cycloalkyl or heterocycloalkyl;L is a chemical linker group optionally coupled to the phthalimido group via an oxygen, amine group or methyl group;X is CH or N; andY is O or H2(i.e., absent). In any aspect or embodiment described herein, the hetero-bifunctional compound is represented by the chemical structure: wherein:Z1is an R group of a CLM as described in any aspect or embodiment described herein that is modified to be covalently linked to L, such a group selected from —C(═O)—, —CONR′—, —O—, —NR′—, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L;n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);R is selected from H, O, OH, N, NH, NH2, Cl, —F, —Br, —I, methyl, optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl), optionally substituted linear or branched alkoxy (e.g., optionally substituted linear or branched C1-C6 alkoxy), -alkyl-aryl (e.g., an -alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxy;L, R3A, R3B, R4, and R5are defined as in any aspect or embodiment described herein;each X is individually CH or N;R7is H, C1-C6 alkyl, C1-C6 alkoxy, or halogen;A is a 4-7 membered aryl, heteroaryl, cycloalkyl or heterocycloalkyl or a 6- to 12-membered (e.g., 8- to 10-membered) spirocycloalkyl or spiroheterocycloalkyl, each optionally substituted with 1, 2, 3, or 4 groups selected from the group consisting of H, C1-3alkyl, methyl, ethyl, and halogen;X is CH or N;A1, A2, and A3as defined in any aspect or embodiment described herein (e.g., A1and A3are individually N or CH, and A2is CR7or N); andY is O or H2(i.e., absent). In any aspect or embodiment described herein, the hetero-bifunctional compound is represented by the chemical structure: wherein:R3A, R3B, R4, and R5are defined as described herein;R7is H or halogen;A is a 4-7 membered heterocycloalkyl;L is a chemical linker group optionally coupled to the phthalimido group via an oxygen, amine group or methyl group;X is CH or N; andY is O or H2(i.e., absent). In any aspect or embodiment described herein, the hetero-bifunctional compound is represented by the chemical structure: wherein:Z1is an R group of a CLM as described in any aspect or embodiment described herein that is modified to be covalently linked to L, such a group selected from —C(═O)—, —CONR′—, —O—, —NR′—, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L;n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);R is selected from H, O, OH, N, NH, NH2, Cl, —F, —Br, —I, methyl, optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl), optionally substituted linear or branched alkoxy (e.g., optionally substituted linear or branched C1-C6 alkoxy), -alkyl-aryl (e.g., an -alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxy;L, R3A, R3B, R4, and R5are defined as in any aspect or embodiment described herein;R7is H or halogen;A is a 4-7 membered heterocycloalkyl or a 6- to 12-membered (e.g., 8- to 10-membered) spiroheterocycloalkyl, each optionally substituted with 1, 2, 3, or 4 groups selected from the group consisting of H, C1-3alkyl, methyl, ethyl, and halogen;each X is individually CH or N;A1, A2, and A3as defined in any aspect or embodiment described herein (e.g., A1and A3are individually N or CH, and A2is CR7or N); andY is O or H2(i.e., absent). In any aspect or embodiment described herein, the hetero-bifunctional compound has the chemical structure: PTM-L-CLM, or a pharmaceutically acceptable salt or solvate thereof, wherein: (a) the CLM is a small molecule E3 ubiquitin ligase binding moiety that binds a cereblon E3 ubiquitin ligase and is represented by the chemical structure: wherein:W is selected from the group consisting of CH2, O, CHR, C═O, SO2, NH, N, optionally substituted cyclopropyl group, optionally substituted cyclobutyl group, and N-alkyl;Q1, Q2, Q3, Q4, Q5each independently represent a carbon C or N substituted with a group independently selected from R′, N or N-oxide;R1is selected from absent (i.e., a bond), H, OH, CN, C1-C3 alkyl, C═O;R2is selected from the group absent (i.e., a bond), H, OH, CN, C1-C3 alkyl, CHF2, CF3, CHO, C(═O)NH2;R3is selected from a bond, H, alkyl (e.g., C1-C6 or C1-C3 alkyl), substituted alkyl (e.g., substituted C1-C6 or C1-C3 alkyl), alkoxy (e.g., C1-C6 or C1-C3 alkoxyl), substituted alkoxy (e.g., substituted C1-C6 or C1-C3 alkoxyl);R4is selected from a bond, H, alkyl, substituted alkyl;R5and R6are each independently a bond, H, halogen, C(═O)R′, CN, OH, CF3;X is C, CH, C═O, or N;X1is C═O, N, CH, or CH2;R′ is selected from a bond, H, halogen, amine, alkyl (e.g., C1-C3 alkyl), substituted alkyl (e.g., substituted C1-C3 alkyl), alkoxy (e.g., C1-C3 alkoxyl), substituted alkoxy (e.g., substituted C1-C3 alkoxyl), NR2R3, C(═O)OR2, optionally substituted phenyl;n is 0-4;is a single or double bond; andthe CLM is covalently joined to a PTM, a chemical linker group (L), a ULM, or CLM); (b) the PTM is a small molecule Leucine-rich repeat kinase 2 (LRRK2) targeting moiety that binds to the human LRRK2 or mutant thereof represented by the chemical structure: wherein:X is CH or N;W is O or S;Q is selected from one of the following: wherein:A1, A2, and A3are each independently selected from N and CR6, wherein for (a) no more than two of A1, A2, and A3are simultaneously N,ring B is an optionally substituted C3-C8 aryl, optionally substituted C3-C8 heteroaryl, optionally substituted C3-C8 cycloalkyl, optionally substituted C3-C8 heterocycloalkyl, optionally substituted C3-C8 aryl, optionally substituted C3-C8 aryl,theof Q indicates the point of attachment with the nitrogen, andtheof Q indicates an optional covalent attachment to RPTM;R2is H or C1-4alkyl;R3Aand R3Bare each independently selected from H, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRdRd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NRe1)NRc1Rd1, NRc1(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1; wherein said C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, and 4-10 membered heterocycloalkyl-C1-4alkyl of R1are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Cy2, Cy2-C1-4alkyl, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1;or R3Aand R3Btogether form a C3-7cycloalkyl or 4-10 membered heterocycloalkyl ring, each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Cy2, Cy2-C1-4alkyl, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1;R4is H, C1-4alkyl, halo, C1-4haloalkyl, or CN;R5is H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl —C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRc2Rd2; wherein said C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, and 4-10 membered heterocycloalkyl-C1-4alkyl of R1are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Cy3, Cy3-C1-4alkyl, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, or S(O)2NRc2Rd2;each R6is independently selected from H, halo, C1-6alkyl, C1-6haloalkyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, CN, NO2, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, OC(O)Rb3, OC(O)NRc3Rd3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)NRc3Rd3, NRc3C(O)ORa3, C(═NRe3)NRc3Rd3, NRc3C(═NRe3)NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, and S(O)2NRc3Rd3, wherein said C1-6alkyl, C1-6haloalkyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl of R6are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, CN, NO2, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, OC(O)Rb3, OC(O)NRc3Rd3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)NRc3Rd3, NRc3C(O)ORa3, C(═NRe3)NRc3Rd3, NRc3C(═NRe3)NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, and S(O)2NRc3Rd3;each Cy1is independently selected from C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, and 4-14 membered heterocycloalkyl, each optionally substituted by 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa, SRa, C(O)Rb, C(O)NRcRd, C(O)ORa, OC(O)Rb, OC(O)NRcRd, NRcRd, NRcC(O)Rb, NRcC(O)ORa, NRcC(O)NRcRd, C(═NRe)Rb, C(═NRe)NRcRd, NRcC(═NRe)NRcRd, NRcS(O)Rb, NRcS(O)2Rb, NRcS(O)2NRcRd, S(O)Rb, S(O)NRcRd, S(O)2Rb, and S(O)2NRcRd;each Cy2is independently selected from C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, and 4-14 membered heterocycloalkyl, each optionally substituted by 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1;each Cy3is independently selected from C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, and 4-14 membered heterocycloalkyl, each optionally substituted by 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6haloalkyl, C6-10aryl, C3-10cycloalkyl, 5-14 membered heteroaryl, 4-14 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, 4-10 membered heterocycloalkyl-C1-4alkyl, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRc2Rd2;each Ra, Rb, Rc, Rd, Ra1, Rb1, Rc1, Rd1, Ra2, Rb2, Rc2, Rd2, Ra3, Rb3, Rc3, and Rd3is independently selected from H, C1-6alkyl, C1-6haloalkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-7cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, and 4-10 membered heterocycloalkyl-C1-4alkyl, wherein said C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-7cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10aryl-C1-4alkyl, C3-7cycloalkyl-C1-4alkyl, 5-10 membered heteroaryl-C1-4alkyl, and 4-10 membered heterocycloalkyl-C1-4alkyl of R″, Rb, Rc, Rd, Ra1, Rb1, Rc1, Rd1, Ra2, Rb2, Rc2, Rd2, Ra3, Rb3, Rc3, or Rd3is optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from halo, C1-4alkyl, C 1-4 haloalkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, CN, ORa4, SRa4, C(O)Rm, C(O)NRc4Rd4, C(O)ORa4, OC(O)Rb4, OC(O)NRc4Rd4, NRc4Rd4, NRc4C(O)Rm, NRc4C(O)NRc4Rd4, NRc4C(O)ORa4, C(═NRe4)NRc4Rd4, NRc4C(═NRe4)NRc4Rd4, S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, and S(O)2NRc4Rd4;each Ra4, Rb4, Rc4, and Rd4are independently selected from H, C1-6alkyl, C1-6haloalkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, wherein said C1-6alkyl, C1-6haloalkyl, C2-6alkenyl, C2-6alkynyl, C6-10aryl, C3-7cycloalkyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl are each optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino, halo, C1-6alkyl, C1-6alkoxy, C1-6haloalkyl, and C1-6haloalkoxy;each Re, Re1, Re2, Re3, and Re4is independently selected from H, C1-4alkyl, and CN;RPTMis H; halogen (e.g., Cl or F); —CN; —OH; —NO2; —NH2; optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl or optionally substituted linear or branched C1-C4 alkyl or C1-C8 alkyl optionally substituted with OH); optionally substituted cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl); O-optionally substituted linear or branched C1-C4 alkyl; an optionally substituted C1-C4 alkynyl; an optionally substituted C1-C4 alkyne; optionally substituted linear or branched hydroxyalkyl (e.g., optionally substituted linear or branched C1-C7 hydroxyalkyl); optionally substituted alkylcycloalkyl (e.g., includes optionally substituted C1-C6 alkyl, optionally substituted C3-C10 cycloalkyl; or both); optionally substituted alkyl-aryl (e.g., includes an optionally substituted linear or branched C1-C6 alkyl, an optionally substituted 5-10 member heteroaryl, or both); optionally substituted alkyl-heteroaryl (e.g., includes an optionally substituted linear or branched C1-C6 alkyl, an optionally substituted 5-10 member heteroaryl, or both); optionally substituted alky 1-heteroaryl (e.g., includes a C1-C6 alkyl, an optionally substituted 5 or 6 member heteroaryl, optionally substituted with a C1-C4 alkyl; the heteroaryl is selected from oxazol-4-yl, 1,3,4-triazol-2-yl, and imidazole-1-yl; or combination thereof); optionally substituted —NH— alkyl-heteroaryl (e.g., an optionally substituted linear or branched C1-C5 alkyl, an optionally substituted 5-8 member heteroaryl, optionally substituted with a C1-C4 alkyl, N—CH2-pyrazol-4-yl, or a combination thereof); optionally substituted alkoxy (e.g., an optionally substituted linear or branched C1-C6 alkyl or —OCH3); optionally substituted O-heterocyclyl (e.g., includes an optionally substituted 3-12 or 4-7 member heterocyclyl; an optionally substituted heterocycloalkyl; an optionally substituted C3-12monocyclic or bicyclic heterocycloalkyl; optionally substituted with at least one OH, C1-C5 alkyl (such as a methyl), ═O, NH2, or a combination thereof; or a combination thereof); optionally substituted S-heterocyclyl (e.g., includes an optionally substituted 4-7 member heterocyclyl; an optionally substituted heterocycloalkyl; optionally substituted with at least one C1-C4 alkyl (such as a methyl), ═O, or a combination thereof; or a combination thereof); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —(CH2)uCO(CH2)vCH3, —COCH3, or —CH2CH2COCH3, wherein each u and v is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —O(CH2)uCO(CH2)vCH3, —O(CH2)uCH((CH2)xCH3)(CH2)wCO(CH2)vCH3, —O—CH2COCH3, —O—CH2COCH2CH3, —O—CH(CH3)COCH3, —OCH2COCH3, or —OCH2(CH3)COCH3, wherein each u, v, w, and x is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —(CH2)uCO(CH2)vNRPTM1aRPTM2a, —CONRPTM1aRPTM2a, —CH2CONRPTM1aRPTM2a, —CH2CH2CONRPTM1aRPTM2a, —CONHCH3, or —CH2CONHCH3, wherein each u and v is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —O(CH2)uCO(CH2)vNRPTM1aRPTM2a, —O(CH2)uCH((CH2)xCH3)(CH2)wCO(CH2)vNRPTM1aRPTM2a, —O—CH(CH3)CONRPTM1aRPTM2a, —O—CH2CONRPTM1aRPTM2a, Or —OCH2C(O)NHOCH3, wherein each u, v, w, and x is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —(CH2)uCHCH(CH2)wCO(CH2)vNRPTM1aRPTM2aor —CHCHCONRPTM1aRPTM2a, wherein each u, v, and w is independently selected from 1, 2, 3, 4 or 5); optionally substituted (e.g., optionally substituted with a linear or branched C1-C4 alkyl; —NH—(CH2)uCO(CH2)vNRPTM1aRPTM2aor —NH—CH2CONRPTM1aRPTM2a, wherein each u and v is independently selected from 1, 2, 3, 4 or 5); fluoroalkoxy (e.g., a mono-, bi- and/or tri-fluoroalkoxy); optionally substituted monocyclic or bicyclic cycloalkyl (e.g., an optionally substituted 3-12 member cycloalkyl; optionally substituted with at least one of OH, ═O, linear or branched C1-C6 alkyl (such as a methyl, ethyl, or butyl), or NH2; or a combination thereof); optionally substituted hydroxycycloalkyl; optionally substituted aryl (e.g., an optionally substitute C5-C10 aryl, an optionally substituted 5-7 member aryl; optionally substituted with at least one halogen or C1-C3 alkyl (e.g., methyl or ethyl); or a combination thereof), optionally substituted heteroaryl (e.g., an optionally substituted 5-10 or member heteroaryl, an optionally substituted 5-7 member heteroaryl; an optionally substituted 5-member heteroaryl; optionally substituted with at least one halogen or C1-C3 alkyl (e.g., methyl or ethyl); or a combination thereof) optionally linked to Q via a C or N-atom of the heteroaryl (e.g., at least one of optionally linked to Q, optionally linked via an optionally substituted —(CH2)uO(CH2)vO(CH2)x—, or a combination thereof); optionally substituted monocyclic or bicyclic heterocyclyl (e.g., an optionally substituted 3-12 member heterocyclyl; an C3-C12 monocyclic or bicyclic heterocycloalkyl, azetidine1-yl, pyrrolidin-1-yl, piperidin-1yl, piperazin-1-yl, or morpholin-4-yl, or homopiperazin-1-yl, each optionally substituted with OH, a linear or branched C1-C5 alkyl (a methyl, ethyl, or butyl group) or NH2) optionally linked to Q via a C or N atom of the heterocyclyl (e.g., at least one of optionally linked to Q, optionally linked via an optionally substituted —(CH2)uO(CH2)vO(CH2)x—, or both);t1is selected from 1, 2, 3, 4, or 5;each t2is independently is independently selected from 0, 1, 2, 3, 4, or 5;RPTM1aand RPTM2aare independently H, optionally substituted C1-C4 alkyl (e.g., a CH3or CH2CH3), optionally substituted C1-C4 alkoxy (e.g., —OCH2or —CH2CH3), optionally substituted CH2OCH3or RPTM1aand RPTM2aare joined together form an optionally substituted 3-10 member ring;n is an integer from 0 to 10; andof the PTM indicates the site of attachment with a chemical linking group or a CLM; and (c) the L is a bond or a chemical linking group that covalently couples the CLM to the PTM. Therapeutic Compositions The present invention further provides pharmaceutical compositions comprising therapeutically effective amounts of at least one bifunctional compound as described herein, in combination with a pharmaceutically acceptable carrier, additive or excipient. In an additional aspect, the description provides therapeutic compositions comprising an effective amount of a compound as described herein or salt form thereof, and a pharmaceutically acceptable carrier, additive or excipient, and optionally an additional bioactive agent. The therapeutic compositions effect targeted protein degradation in a patient or subject, for example, an animal such as a human, and can be used for treating or ameliorating disease states or conditions which are modulated by degrading the target protein. In certain embodiments, the therapeutic compositions as described herein may be used to effectuate the degradation of protein for the treatment or amelioration of LRRK2-mediated neurodegenerative disease, inflammatory diseases, autoimmune diseases or cancer. In certain additional embodiments, the disease is Parkinson's disease, Parkinson disease with dementia, Parkinson's disease at risk syndrome, dementia with Lewy bodies, Lewy body variant of Alzheimer's disease, combined Parkinson's disease and Alzheimer's disease, multiple system atrophy, striatonigral degeneration, olivopontocerebellar atrophy, and Shy-Drager syndrome. In alternative aspects, the present disclosure relates to a method for treating a disease state or ameliorating one or more symptoms of a disease or condition in a subject in need thereof by degrading the LRRK2 protein comprising administering to said patient or subject an effective amount, e.g., a therapeutically effective amount, of at least one compound as described herein, optionally in combination with a pharmaceutically acceptable carrier, additive or excipient, and optionally coadministered with an additional bioactive agent, wherein the composition is effective for treating or ameliorating the disease or disorder or one or more symptoms thereof in the subject. The method according to the present disclosure may be used to treat certain disease states or conditions including neurodegenerative diseases, by virtue of the administration of effective amounts of at least one compound described herein. The present disclosure further includes pharmaceutical compositions comprising a pharmaceutically acceptable salt, in particular, acid or base addition salts of compounds as described herein. The acids which are used to prepare the pharmaceutically acceptable acid addition salts of the aforementioned compounds useful according to this aspect are those which form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, such as the hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, acetate, lactate, citrate, acid citrate, tartrate, bitartrate, succinate, maleate, fumarate, gluconate, saccharate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate [i.e., 1,1′-methylene-bis-(2-hydroxy-3 naphthoate)]salts, among numerous others. Pharmaceutically acceptable base addition salts may also be used to produce pharmaceutically acceptable salt forms of the compounds according to the present disclosure. The chemical bases that may be used as reagents to prepare pharmaceutically acceptable base salts of the present compounds are those that form non-toxic base salts with such compounds. Such non-toxic base salts include, but are not limited to those derived from such pharmacologically acceptable cations such as alkali metal cations (e.g., potassium and sodium) and alkaline earth metal cations (e.g., calcium, zinc and magnesium), ammonium or water-soluble amine addition salts such as N-methylglucamine-(meglumine), and the lower alkanolammonium and other base salts of pharmaceutically acceptable organic amines, among others. The compounds as described herein may, in accordance with the disclosure, be administered in single or divided doses by the oral, parenteral or topical routes. Administration of the active compound may range from continuous (intravenous drip) to several oral administrations per day (for example, Q.I.D.) and may include oral, topical, parenteral, intramuscular, intravenous, sub-cutaneous, transdermal (which may include a penetration enhancement agent), buccal, sublingual, intra nasal, intra ocular, intrathecal, and suppository administration, among other routes of administration. Enteric coated oral tablets may also be used to enhance bioavailability of the compounds from an oral route of administration. The most effective dosage form will depend upon the pharmacokinetics of the particular agent chosen as well as the severity of disease in the patient. Administration of compounds according to the present disclosure as sprays, mists, or aerosols for intra-nasal, intra-tracheal or pulmonary administration may also be used. The present disclosure therefore also is directed to pharmaceutical compositions comprising an effective amount of compound as described herein, optionally in combination with a pharmaceutically acceptable carrier, additive or excipient. Compounds according to the present disclosure may be administered in immediate release, intermediate release or sustained or controlled release forms. Sustained or controlled release forms are preferably administered orally, but also in suppository and transdermal or other topical forms. Intramuscular injections in liposomal form or in depot formulation may also be used to control or sustain the release of compound at an injection site. The compositions as described herein may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers and may also be administered in controlled-release formulations. Pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as prolamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. The compositions as described herein may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term “parenteral” as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions as described herein may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as Ph. Helv or similar alcohol. The pharmaceutical compositions as described herein may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient may be combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. Alternatively, the pharmaceutical compositions as described herein may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient, which is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols. The pharmaceutical compositions as described herein may also be administered topically. Suitable topical formulations are readily prepared for each of these areas or organs. For topical applications, the pharmaceutical compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of this disclosure include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. In certain preferred aspects of the disclosure, the compounds may be coated onto a stent which is to be surgically implanted into a patient in order to inhibit or reduce the likelihood of occlusion occurring in the stent in the patient. Alternatively, the pharmaceutical compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. For ophthalmic use, the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutical compositions may be formulated in an ointment such as petrolatum. The pharmaceutical compositions as described herein may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. The amount of active pharmaceutical ingredient in a pharmaceutical composition as described herein that may be combined with the carrier materials to produce a single dosage form will vary depending upon the condition of the subject and disease treated, as well as the particular mode of administration. Preferably, the compositions should be formulated to contain between about 0.05 milligram and about 750 milligrams or more, more preferably about 1 milligram to about 600 milligrams, and even more preferably about 10 milligrams to about 500 milligrams of active ingredient, alone or in combination with another compound according to the present disclosure. It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity and bioavailability of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease or condition being treated. A patient or subject in need of therapy using compounds according to the methods described herein can be treated by administering to the patient (subject) an effective amount of the compound according to the present disclosure depending upon the pharmaceutically acceptable salt, solvate or polymorph, thereof optionally in a pharmaceutically acceptable carrier or diluent, either alone, or in combination with another known therapeutic agent. The active compound is combined with the pharmaceutically acceptable carrier or diluent in an amount sufficient to deliver to a patient a therapeutically effective amount for the desired indication, without causing serious toxic effects in the patient treated. A preferred dose of the active compound for all of the herein-mentioned conditions is in the range from about 10 nanograms per kilograms (ng/kg) to 300 milligrams per kilograms (mg/kg), preferably 0.1 to 100 mg/kg per day, more generally 0.5 to about 25 mg per kilogram body weight of the recipient/patient per day. A typical topical dosage will range from 0.01-5% wt/wt in a suitable carrier. The compound is conveniently administered in any suitable unit dosage form, including but not limited to a dosage form containing less than 1 milligrams (mg), 1 mg to 3000 mg, or 5 mg to 500 mg of active ingredient per unit dosage form. An oral dosage of about 25 mg-250 mg is often convenient. The active ingredient is preferably administered to achieve peak plasma concentrations of the active compound of about 0.00001-30 millimole (mM), preferably about 0.1-30 micromole (μM). This may be achieved, for example, by the intravenous injection of a solution or formulation of the active ingredient, optionally in saline, or an aqueous medium or administered as a bolus of the active ingredient. Oral administration may also be appropriate to generate effective plasma concentrations of active agent. The concentration of active compound in the drug composition will depend on absorption, distribution, inactivation, and excretion rates of the drug as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time. Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound or its prodrug derivative can be incorporated with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a dispersing agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or enteric agents. The active compound or pharmaceutically acceptable salt thereof can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors. The active compound or pharmaceutically acceptable salts thereof can also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as anti-cancer agents, as described herein among others. In certain preferred aspects of the disclosure, one or more compounds according to the present disclosure are coadministered with another bioactive agent, such as an anti-cancer agent or a wound healing agent, including an antibiotic, as otherwise described herein. Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. If administered intravenously, preferred carriers are physiological saline or phosphate buffered saline (PBS). In any aspect or embodiment described herein, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, poly anhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. Liposomal suspensions may also be pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811 (which is incorporated herein by reference in its entirety). For example, liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the active compound are then introduced into the container. The container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension. Therapeutic Methods In an additional aspect, the description provides therapeutic methods comprising administration of an effective amount of a compound as described herein or salt form thereof, and a pharmaceutically acceptable carrier. The therapeutic methods are useful to effect protein degradation in a patient or subject in need thereof, for example, an animal such as a human, for treating or ameliorating a disease state, condition or related symptom that me be treated through targeted protein degradation. The terms “treat”, “treating”, and “treatment”, etc., as used herein, refer to any action providing a benefit to a patient for which the present compounds may be administered, including the treatment of any disease state, condition, or symptom which is related to the protein to which the present compounds bind. Disease states or conditions, including cancer, which may be treated using compounds according to the present disclosure are set forth hereinabove. The description provides therapeutic methods for effectuating the degradation of proteins of interest for the treatment or amelioration of a disease, e.g., cancer. In any aspect or embodiment described herein, the disease is multiple myeloma. As such, in another aspect, the description provides a method of ubiquitinating/degrading a target protein in a cell. In certain embodiments, the method comprises administering a bifunctional compound of the invention. The control or reduction of specific protein levels in cells of a subject as afforded by the present disclosure provides treatment of a disease state, condition, or symptom. In any aspect or embodiment described herein, the method comprises administering an effective amount of a compound as described herein, optionally including a pharmaceutically acceptable excipient, carrier, adjuvant, another bioactive agent or combination thereof. In additional embodiments, the description provides methods for treating or ameliorating a disease, disorder or symptom thereof in a subject or a patient, e.g., an animal such as a human, comprising administering to a subject in need thereof a composition comprising an effective amount, e.g., a therapeutically effective amount, of a compound as described herein or salt form thereof, and a pharmaceutically acceptable excipient, carrier, adjuvant, another bioactive agent or combination thereof, wherein the composition is effective for treating or ameliorating the disease or disorder or symptom thereof in the subject. In another aspect, the description provides methods for identifying the effects of the degradation of proteins of interest in a biological system using compounds according to the present disclosure. In another aspect, the description provides a process for making a molecule that can cause degradation of LRRK2 in a cell, comprising the steps of: i. providing a small molecule that binds LRRK2; ii. providing and E3 ubiquitin ligase binding moiety (ULM), preferably a CLM such as thalidomide, pomalidomide, lenalidomide or an analog thereof; and iii. covalently coupling the small molecule of step (i) to the ULM of step (ii) via a chemical linking group (L) to form a compound which binds to both a cereblon E3 ubiquitin ligase and LRRK2 protein in the cell, such that the cereblon E3 ubiquitin ligase is in proximity to, and ubiquitinates LRRK2 protein bound thereto, such that the ubiquitinated LRRK2 is then degraded. In another aspect, the description provides a method for detecting whether a molecule can trigger degradation of a LRRK2 protein in a cell, the method comprising the steps of: (i) providing a molecule for which the ability to trigger degradation of LRRK2 protein in a cell is to be detected, said molecule comprising the structure: CLM-L-PTM, wherein CLM is a cereblon E3 ubiquitin ligase binding moiety capable of binding a cereblon E3 ubiquitin ligase in a cell, which CLM is thalidomide, pomalidomide, lenalidomide, or an analog thereof; PTM is a protein targeting moiety, which is a small molecule that binds to LRRK2, said LRRK2 having at least one lysine residue available to be ubiquitinated by a cereblon E3 ubiquitin ligase bound to the CLM of the molecule; and L is a chemical linking group that covalently links the CLM to the PTM to form the molecule; (ii) incubating a LRRK2 protein-expressing cell in the presence of the molecule of step (i); and (iii) detecting whether the LRRK2 protein in the cell has been degraded. In any of the aspects or embodiments described herein, the small molecule capable of binding LRRK2, is a small molecule as described herein. In another aspect of said treatment, the present disclosure provides a method of treating a human patient in need of said treatment of a disease state, condition, or symptom causally related to LRRK2 expression, over-expression, mutation, misfolding or dysregulation where the degradation of the LRRK2 protein will produce a therapeutic effect in the patient, the method comprising administering to the patient an effective amount of a compound according to the present disclosure, optionally in combination with another bioactive agent. The disease state, condition, or symptom may be caused by a microbial agent or other exogenous agent such as a vims, bacteria, fungus, protozoa or other microbe, or may be a disease state, which is caused by expression, overexpression, mutation, misfolding, or dysregulation of the protein, which leads to a disease state, condition, or symptom. In another aspect, the present disclosure provides a method of treating or ameliorating at least one symptom of a disease or condition in a subject, comprising the steps of: providing a subject identified as having a symptom of a disease or condition causally related to expression, overexpression, mutation, misfolding, or dysregulation of LRRK2 protein in the subject, and the symptom of the disease or condition is treated or ameliorated by degrading LRRK2 protein in cells of the subject; and administering to the subject therapeutically effective amount of a compound comprising a small molecule of the present invention such that the LRRK2 protein is degraded, thereby treating or ameliorating at least one symptom of a disease or condition in the subject. The term “disease state or condition” is used to describe any disease state or condition wherein protein expression overexpression, mutation, misfolding, or dysregulation (e.g., the amount of protein expressed in a patient is elevated) occurs and where degradation of the LRRK2 protein to reduce or stabilize the level of LRRK2 protein (whether mutated or not) in a patient provides beneficial therapy or relief of symptoms to a patient in need thereof. In certain instances, the disease state, condition, or symptom may be cured. Disease state, condition, or symptom which may be treated using compounds according to the present disclosure include, for example, inflammatory conditions, and immunological conditions, asthma, autoimmune diseases such as multiple sclerosis, neurodegenerative disease such as Parkinson's disease, various cancers, ciliopathies, diabetes, heart disease, hypertension, inflammatory bowel disease, mental retardation, mood disorder, obesity, refractive error, infertility, Angelman syndrome, Canavan disease, Coeliac disease, Charcot-Marie-Tooth disease, Cystic fibrosis, Duchenne muscular dystrophy, Haemochromatosis, Haemophilia, Klinefelter's syndrome, Neurofibromatosis, Phenylketonuria, Polycystic kidney disease, (PKD1) or 4 (PKD2) Prader-Willi syndrome, Sickle-cell disease, Tay-Sachs disease, and Turner syndrome. The term “neurodegenerative disease” (or Degenerative nerve diseases) is used throughout the specification to refer to the pathological process that affect many of body's activities, such as balance, movement, talking, breathing, and heart function. Many of these diseases are genetic, sometimes the cause is a medical condition such as alcoholism, a tumor, or a stroke. Other causes may include toxins, chemicals, and viruses, and sometimes the cause is unknown. Exemplary neurodegenerative disease which may be treated by the present compounds either alone or in combination with at least one additional anti-neurodegenerative disease therapeutic agent include therapeutic agents to treat Parkinson's disease, Parkinson disease with dementia, Parkinson's disease at risk syndrome, dementia with Lewy bodies, Lewy body variant of Alzheimer's disease, combined Parkinson's disease and Alzheimer's disease, multiple system atrophy, striatonigral degeneration, olivopontocerebellar atrophy, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis and Shy-Drager syndrome. For example, Carmustine, Bexarotene, Tamibarotene, Imatinib, Paclitaxel, azithromycin, erythromycin, Doxycycline, Rifampicin, acyclovir, penciclovir and foscamet. The term “bioactive agent” is used to describe an agent, other than a compound according to the present disclosure, which is used in combination with a present compound as an agent with biological activity to assist in effecting an intended therapy, inhibition and/or prevention/prophylaxis for which the present compounds are used. Preferred bioactive agents for use herein include those agents which have pharmacological activity similar to that for which the present compounds are used or administered and include for example, anti-cancer agents, antiviral agents, especially including anti-HIV agents and anti-HCV agents, antimicrobial agents, antifungal agents, etc. The term “additional anti-neurodegenerative disease agent” is used to describe an anti-neurodegenerative disease therapeutic agent, which may be combined with a compound according to the present disclosure to treat neurodegenerative disease. These agents include, for example, Carmustine, Bexarotene, Tamibarotene, Imatinib, Paclitaxel, azithromycin, erythromycin, Doxycycline, Rifampicin, acyclovir, penciclovir and foscamet. The term “pharmaceutically acceptable derivative” is used throughout the specification to describe any pharmaceutically acceptable prodrug form (such as an ester, amide other prodrug group), which, upon administration to a patient, provides directly or indirectly the present compound or an active metabolite of the present compound. EXAMPLES Abbreviations ACN AcetonitrileAcOH Acetic acidDCM DichloromethaneDMF DimethylformamideDMSO Dimethyl SulfoxideDIPEA N, N-DiisopropylethylamineEtOAc/EA Ethyl AcetateEtOH EthanolHATU Hexafluorophosphate Azabenzotriazole Tetramethyl UroniumHPLC High pressure liquid chromatographyHz HertzKOAc Potassium acetateLCMS Liquid Chromatography/Mass SpectrometryMHz MegahertzNMR Nuclear Magnetic ResonanceMeOH MethanolMS Mass SpectrometryPE Petroleum etherPsi Pound-force per square inchRT or r.t. Room temperatureTEA TriethylamineTHF TetrahydrofuranTFA Trifluoracetic acidTLC Thin layer chromatographyTMS Trimethylsilyl General Synthetic Approach The synthetic realization and optimization of the bifunctional molecules as described herein may be approached in a stepwise or modular fashion. For example, identification of compounds that bind to the target protein, i.e., LRRK2 can involve high or medium throughput screening campaigns if no suitable ligands are immediately available. It is not unusual for initial ligands to require iterative design and optimization cycles to improve suboptimal aspects as identified by data from suitable in vitro and pharmacological and/or ADMET assays. Part of the optimization/SAR campaign would be to probe positions of the ligand that are tolerant of substitution and that might be suitable places on which to attach the chemical linking group previously referred to herein. Where crystallographic or NMR structural data are available, these can be used to focus such a synthetic effort. In a very analogous way one can identify and optimize ligands for an E3 Ligase. With PTMs and ULMs (e.g. CLMs) in hand, one skilled in the art can use known synthetic methods for their combination with or without a chemical linking group(s). Chemical linking group(s) can be synthesized with a range of compositions, lengths and flexibility and functionalized such that the PTM and ULM groups can be attached sequentially to distal ends of the linker. Thus, a library of bifunctional molecules can be realized and profiled in in vitro and in vivo pharmacological and ADMET/PK studies. As with the PTM and ULM groups, the final bifunctional molecules can be subject to iterative design and optimization cycles in order to identify molecules with desirable properties. In some instances, protecting group strategies and/or functional group interconversions (FGIs) may be required to facilitate the preparation of the desired materials. Such chemical processes are well known to the synthetic organic chemist and many of these may be found in texts such as “Greene's Protective Groups in Organic Synthesis” Peter G. M. Wuts and Theodora W. Greene (Wiley), and “Organic Synthesis: The Disconnection Approach” Stuart Warren and Paul Wyatt (Wiley). Synthetic Procedures Synthesis of Intermediate 1: 4,5-Dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid Step 1: Ethyl 5-methyl-4,7-dihydrotetrazolo[1,5-a]pyrimidine-6-carboxylate To a solution of 1H-tetrazol-5-amine (5 gram (g), 48.50 millimole (mmol), 1 equivalent (eq), H2O) in water (100 milliliters (mL)) was added formaldehyde (5.46 g, 72.75 mmol, 5.01 mL, 1.5 eq) and ethyl acetoacetate (6.31 g, 48.50 mmol, 6.13 mL, 1 eq). The mixture was stirred at 100° C. for 9 hours (hr) to give white solution. Then the solution became suspension. LCMS (EB16-543-P1A1) showed the reaction was completed. The mixture was cooled to 20° C. and concentrated in reduced pressure at 20° C. The suspension was filtered and the filter cake was concentrated in vacuum to give ethyl 5-methyl-4,7-dihydrotetrazolo[1,5-a]pyrimidine-6-carboxylate (7.2 g, 34.42 mmol, 70.96% yield) as a white solid. Step 2: Ethyl 4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylate To a mixture of ethyl 5-methyl-4,7-dihydrotetrazolo[1,5-a]pyrimidine-6-carboxylate (7.2 g, 34.42 mmol, 1 eq) and Cs2CO3(12.33 g, 37.86 mmol, 1.1 eq) in MeCN (70 mL) was added Mel (6.06 g, 42.68 mmol, 2.66 mL, 1.24 eq) in one portion at 20° C. under N2. The mixture was stirred at 50° C. for 1 hour (h). LCMS (EB16-545-P1A1) showed the reaction was completed. The aqueous phase was extracted with ethyl acetate (50 mL*3). The combined organic phase was washed with brine (50 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum to give ethyl 4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylate (7.5 g, 33.60 mmol, 97.61% yield) as a yellow solid. Step 3: 4,5-Dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid To a mixture of ethyl 4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylate (4 g, 17.92 mmol, 1 eq) in THF (30 mL) was added LiOH (1.29 g, 53.76 mmol, 3 eq) and H2O (5 mL) in one portion at 20° C. under N2. The mixture was stirred at 60° C. for 16 hours. LCMS (EB16-548-P1A1) showed the reaction was completed. The mixture was cooled to 20° C. and concentrated in reduced pressure at 20° C. The residue was acidified to pH=1 with HCl. The solid formed and collected by filtration. The filter cake was filtered under vacuum to give 4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (2.9 g, 14.86 mmol, 82.92% yield) as a white solid. Synthesis of Intermediate 2: 4,5,7-Trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid. Synthesis of Intermediate 2 was made in a manner analogous to intermediate 1 using acetaldehyde in place of formaldehyde in step 1. Synthesis of Intermediate 3: 4,5,7,7-Tetramethyltetrazolo[1,5-a]pyrimidine-6-carboxylic acid. Synthesis of Intermediate 3 was made in a manner analogous to intermediate 1 using ethyl 2-acetyl-3-methyl-but-2-enoate, prepared by the condensation of acetone and ethyl acetoacetate, in step 1. Exemplary Synthesis of Exemplary Compound 1: N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1: Tert-butyl (3R,5S)-4-[(1-benzyloxycarbonyl-4-piperidyl)methyl]-3,5-dimethyl-piperazine-1-carboxylate To a mixture of tert-butyl (3R,5S)-3,5-dimethylpiperazine-1-carboxylate (1 g, 4.67 mmol, 1 eq) and benzyl 4-(p-tolylsulfonyloxymethyl)piperidine-1-carboxylate (1.88 g, 4.67 mmol, 1 eq) in MeCN (5 mL) was added KI (2.32 g, 14.00 mmol, 3 eq) and DIPEA (1.81 g, 14.00 mmol, 2.44 mL, 3 eq) in one portion at 20° C. under N2. The mixture was stirred at 100° C. for 16 hours. LCMS (EB16-606-P1A6) showed desired MS. The mixture was cooled to 20° C. and concentrated in reduced pressure at 20° C. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 mL*3). The combined organic phase was washed with brine (5 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (DCM:MeOH=10:1, Rf=0.59, 20 g, 0-14% (10 min) of Ethyl acetate in Petroleum ether, 14% (20 min) of Ethyl acetate in Petroleum ether) to give tert-butyl (3R,5S)-4-[(1-benzyloxycarbonyl-4-piperidyl)methyl]-3,5-dimethyl-piperazine-1-carboxylate (1.22 g, 2.74 mmol, 58.67% yield) as a yellow gum. Step 2: Benzyl 4-[[(2R,6S)-2,6-dimethylpiperazin-1-yl]methyl]piperidine-1-carboxylate To a solution of tert-butyl (3R,5S)-4-[(1-benzyloxycarbonyl-4-piperidyl)methyl]-3,5-dimethyl-piperazine-1-carboxylate (1.22 g, 2.74 mmol, 1 eq) in DCM (5 mL) was added TFA (312.18 mg, 2.74 mmol, 202.71 microliter (uL), 1 eq) in one portion at 20° C. under N2. The mixture was stirred at 20° C. for 30 min. LCMS (EB16-610-P1A1) showed the reaction was completed. The residue was basified to pH=7-8 with saturated NaHCO3. The aqueous phase was extracted with DCM (5 mL*3). The combined organic phase was washed with brine (5 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum to give benzyl 4-[[(2R,6S)-2,6-dimethylpiperazin-1-yl]methyl]piperidine-1-carboxylate (930 mg, 2.69 mmol, 98.32% yield) as a yellow gum. Step 3: Benzyl 4-[[(2R,6S)-4-(4-bromo-2-pyridyl)-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate To a mixture of benzyl 4-[[(2R,6S)-2,6-dimethylpiperazin-1-yl]methyl]piperidine-1-carboxylate (930 mg, 2.69 mmol, 1 eq) and 4-bromo-2-fluoro-pyridine (473.74 mg, 2.69 mmol, 1 eq) in DMF (5 mL) was added Cs2CO3(1.75 g, 5.38 mmol, 2 eq) in one portion at 25° C. The mixture was stirred at 100° C. for 12 hours. LCMS (EB16-612-P1A1) showed the reaction was completed. The mixture was cooled to 20° C. and concentrated in reduced pressure at 20° C. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 mL*3). The combined organic phase was washed with brine (5 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (12 g, 0-50%(10 min) of Ethyl acetate in Petroleum ether, 50%(10 min) of Ethyl acetate in Petroleum ether) to give benzyl 4-[[(2R,6S)-4-(4-bromo-2-pyridyl)-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate (580 mg, 1.16 mmol, 42.97% yield) as a yellow gum. Step 4: [2-[(3R,5S)-4-[(1-benzyloxycarbonyl-4-piperidyl)methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]boronic acid To a mixture of benzyl 4-[[(2R,6S)-4-(4-bromo-2-pyridyl)-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate (580 mg, 1.16 mmol, 1 eq) 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (440.57 mg, 1.73 mmol, 1.5 eq) and KOAc (340.53 mg, 3.47 mmol, 3 eq) in dioxane (2 mL) was added Pd(dppf)Cl2(42.32 mg, 57.83 umol, 0.05 eq) one portion at 25° C. under N2. The mixture was stirred at 100° C. for 2 hours. LCMS (EB16-615-P1A1) showed the reaction was completed. The mixture was cooled to 25° C., filtered and concentrated in vacuum to give [2-[(3R,5S)-4-[(1-benzyloxycarbonyl-4-piperidyl)methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]boronic acid (669 mg, crude) as a black brown solid. The crude product was used into the next step without further purification. Step 5: Benzyl 4-[[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]piperidine-1-carboxylate To a mixture of [2-[(3R,5S)-4-[(1-benzyloxycarbonyl-4-piperidyl)methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]boronic acid (669 mg, 1.43 mmol, 1 eq), 3-bromo-5-nitro-1H-indazole (381.90 mg, 1.58 mmol, 1.1 eq) and KOAc (422.34 mg, 4.30 mmol, 3 eq) in EtOH (10 mL) and H2O (2 mL) was added 4-ditert-butylphosphanyl-N,N-dimethyl-aniline; dichloropalladium (101.57 mg, 143.45 umol, 101.57 uL, 0.1 eq) in one portion at 25° C. under N2. The mixture was stirred at 100° C. for 12 hours. LCMS (EB16-616-P1A2) showed the reaction was completed. The mixture was cooled to 20° C. and concentrated in reduced pressure. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL*3). The combined organic phase was washed with brine (10 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (12 g, 100-200 mesh silica gel, 0-100% (15 min) of Ethyl acetate in Petroleum ether, 100% (15 min) of Ethyl acetate in Petroleum ether) to give benzyl 4-[[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]piperidine-1-carboxylate (609 mg, 980.78 umol, 68.37% yield, 94% purity) as a yellow gum. Step 6: Benzyl 4-[[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate To a mixture of benzyl 4-[[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]piperidine-1-carboxylate (609 mg, 1.04 mmol, 1 eq) and Fe (291.34 mg, 5.22 mmol, 5 eq) in H2O (1 mL) and EtOH (5 mL) was added NH4Cl (279.06 mg, 5.22 mmol, 5 eq) in one portion at 20° C. under N2. The mixture was stirred at 80° C. for 1.5 hours. LCMS (EB16-620-P1A1) showed the reaction was completed. The residue was filtered and solution was concentrated in vacuum to give benzyl 4-[[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate (550 mg, 814.53 umol, 78.07% yield, 82% purity) as a brown solid. Step 7: Benzyl 4-[[(2R,6S)-4-[4-[5-(tert-butoxycarbonylamino)-1H-indazol-3-yl]-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate To a mixture of benzyl 4-[[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate (400 mg, 722.42 umol, 1 eq) and Boc2O (173.43 mg, 794.66 umol, 182.56 uL, 1.1 eq) in THF (5 mL) was added DIPEA (140.05 mg, 1.08 mmol, 188.75 uL, 1.5 eq) in one portion at 20° C. under N2. The mixture was stirred at 20° C. for 16 hours. TLC showed the reaction was completed. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 mL*3). The combined organic phase was washed with brine (5 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (12 g, 0-50% (10 min) of Ethyl acetate in Petroleum ether, 50% (10 min) of Ethyl acetate in Petroleum ether) to give benzyl 4-[[(2R,6S)-4-[4-[5-(tert-butoxycarbonylamino)-1H-indazol-3-yl]-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate (274 mg, 410.70 umol, 56.85% yield, 98% purity) as an off-white solid. Step 8: Tert-butyl N-[3-[2-[(3R,5S)-3,5-dimethyl-4-(4-piperidylmethyl)piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]carbamate To a mixture of benzyl 4-[[(2R,6S)-4-[4-[5-(tert-butoxycarbonylamino)-1H-indazol-3-yl]-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]piperidine-1-carboxylate (274 mg, 419.08 umol, 1 eq) in THF (5 mL) was added Pd/C (50 mg, 419.08 umol, 10% purity, 1 eq) under N2. The suspension was degassed under vacuum and purged with H2several times. The mixture was stirred under H2(15 psi) at 25° C. for 16 hours. TLC showed the reaction was completed. The reaction mixture was filtered with MeOH (20 mL*3) and the filter was concentrated to give tert-butyl N-[3-[2-[(3R,5S)-3,5-dimethyl-4-(4-piperidylmethyl)piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]carbamate (160 mg, 252.16 umol, 60.17% yield, 81.9% purity) as a white solid. Step 9: Tert-butyl N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]carbamate To a mixture of tert-butyl N-[3-[2-[(3R,5S)-3,5-dimethyl-4-(4-piperidylmethyl)piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]carbamate (160 mg, 307.88 umol, 1 eq) and 1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperidine-4-carbaldehyde (170.58 mg, 461.82 umol, 1.5 eq) in MeOH (5 mL) was added AcOH (18.49 mg, 307.88 umol, 17.61 uL, 1 eq) and borane; 2-methylpyridine (65.86 mg, 615.76 umol, 2 eq) in one portion at 20° C. under N2. The mixture was stirred at 20° C. for 4 h. LCMS (EB16-636-P1A2) showed there was desired MS. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL*3). The combined organic phase was washed with brine (10 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (Dichloromethane:Methanol=10:1, Rf=0.09, 0-20% (15 min) of Methanol in Dichloromethane, 20% (5 min) of Methanol in Dichloromethane) to give tert-butyl N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]carbamate (220 mg, 231.83 umol, 75.30% yield, 92% purity) as a yellow gum. Step 10: 5-[4-[[4-[[(2R,6S)-4-[4-(5-Amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione To a mixture of tert-butyl N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]carbamate (220 mg, 251.99 umol, 1 eq) in DCM (3 mL) was added TFA (86.20 mg, 755.97 umol, 55.97 uL, 3 eq) in one portion at 20° C. under N2. The mixture was stirred at 20° C. for 30 min. TLC (DCM:MeOH=3:1, Rf=0.14) showed the reaction was completed. The mixture was concentrated under reduced pressure to give 5-[4-[[4-[[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (300 mg, crude, TFA) as a yellow gum. Step 11: N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (Compound 1) To a solution of 4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (Intermediate 1, 30.30 mg, 155.25 umol, 1.2 eq) and 5-[4-[[4-[[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (100 mg, 129.38 umol, 1 eq) in DMF (2 mL) was stirred at 0° C. for 10 min, then was added DIPEA (83.61 mg, 646.88 umol, 112.68 uL, 5 eq) and HATU (49.19 mg, 129.38 umol, 1 eq). Then the mixture was stirred at 25° C. for 16 h under N2. LCMS (EB16-642-P1A8) showed the reaction was completed. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL*3). The combined organic phase was washed with brine (10 mL*3), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The crude product was purified by reversed-phase HPLC (Column: Phenomenex luna C18 150*25 mm*10 um; Condition: water (0.2% FA)-ACN; Begin B: 0; End B: 40; FlowRate: 35 mL/min; Gradient Time: 35 min; 100% B Hold Time: 4 min) to give N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (29.3 mg, 30.53 umol, 23.60% yield, 99% purity) as a yellow solid. 1H NMR: (400 MHz, DMSO-d6) δ: 13.42 (s, 1H), 11.08 (s, 1H), 10.07 (s, 1H), 8.67 (s, 1H), 8.22 (d, J=5.1 Hz, 1H), 7.67-7.51 (m, 3H), 7.28 (d, J=13.8 Hz, 2H), 7.23 (d, J=8.8 Hz, 1H), 7.16 (d, J=5.3 Hz, 1H), 5.30 (s, 2H), 5.11-5.02 (m, 1H), 4.12 (d, J=11.1 Hz, 2H), 4.03 (d, J=12.9 Hz, 2H), 3.44 (s, 3H), 3.00-2.80 (m, 5H), 2.72-2.53 (m, 6H), 2.39-2.30 (m, 2H), 2.27 (s, 3H), 2.16 (d, J=5.8 Hz, 2H), 2.05-1.97 (m, 1H), 1.95-1.71 (m, 7H), 1.37 (s, 1H), 1.12 (d, J=5.8 Hz, 10H). Exemplary Synthesis of Exemplary Compound 2: N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide To a solution of 4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (32.48 mg, 155.25 umol, 1.2 eq) and 5-[4-[[4-[[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (100 mg, 129.38 umol, 1 eq) in DMF (2 mL) was stirred at 0° C. for 10 min, then was added DIPEA (83.60 mg, 646.88 umol, 112.67 uL, 5 eq) and HATU (49.19 mg, 129.38 umol, 1 eq). Then the mixture was stirred at 25° C. for 16 h under N2. LCMS (EB16-643-P1A1) showed the reaction was completed. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL*3). The combined organic phase was washed with brine (10 mL*3), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The crude product was purified by reversed-phase HPLC (Column: Phenomenex luna C18 150*25 mm*10 um; Condition: water (0.2% FA)-ACN; Begin B: 0; End B: 40; FlowRate: 35 mL/min; Gradient Time: 35 min; 100% B Hold Time: 4 min) to give N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (34.9 mg, 35.84 umol, 27.70% yield, 99% purity) as a yellow solid. 1H NMR: (400 MHz, DMSO) δ: 13.42 (s, 1H), 11.08 (s, 1H), 10.28 (s, 1H), 8.67 (s, 1H), 8.23 (d, J=5.3 Hz, 1H), 8.15 (s, 1H), 7.67-7.58 (m, 2H), 7.56-7.50 (m, 1H), 7.28 (d, J=13.6 Hz, 2H), 7.23 (d, J=8.8 Hz, 1H), 7.16 (d, J=5.4 Hz, 1H), 5.83-5.71 (m, 1H), 5.06 (dd, J=5.4, 12.9 Hz, 1H), 4.12 (d, J=12.0 Hz, 2H), 4.03 (d, J=12.9 Hz, 2H), 3.44 (s, 3H), 3.01-2.82 (m, 5H), 2.72-2.55 (m, 5H), 2.40-2.33 (m, 2H), 2.20 (s, 3H), 2.17 (s, 2H), 2.06-1.97 (m, 1H), 1.94-1.71 (m, 7H), 1.56 (d, J=6.4 Hz, 3H), 1.38 (s, 1H), 1.11 (d, J=5.8 Hz, 10H). Exemplary Synthesis of Exemplary Compound 3: N-[3-[2-[(3S,5R)-4-[2-[2-[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]ethoxy]ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1: (2R,6S)-4-(4-bromo-2-pyridyl)-1-[2-(2,2-dimethoxyethoxy)ethyl]-2,6-dimethyl-piperazine To a solution of (3R,5S)-1-(4-bromo-2-pyridyl)-3,5-dimethyl-piperazine (2 g, 7.40 mmol, 1 eq) and 2-(2,2-dimethoxyethoxy)ethyl 4-methylbenzenesulfonate (2.3 g, 7.56 mmol, 1.02 eq) in CH3CN (20 mL) was added DIEA (1.91 g, 14.81 mmol, 2.58 mL, 2 eq) and KI (2.46 g, 14.81 mmol, 2 eq). After addition, the reaction mixture was stirred at 120° C. for 16 h. LCMS (EB12-469-P1B) showed desired MS. TLC (dichloromethane:methanol=10:1) showed several new spots. After cooling, the reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 100% ethyl acetate in petroleum ether) to afford (2R,6S)-4-(4-bromo-2-pyridyl)-1-[2-(2,2-dimethoxyethoxy)ethyl]-2,6-dimethyl-piperazine (2.1 g, 4.79 mmol, 64.73% yield, 91.8% purity) as a yellow oil. Step 2: (2R,6S)-1-[2-(2,2-dimethoxyethoxy)ethyl]-2,6-dimethyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl]piperazine To a solution of (2R,6S)-4-(4-bromo-2-pyridyl)-1-[2-(2,2-dimethoxyethoxy)ethyl]-2,6-dimethyl-piperazine (2.1 g, 5.22 mmol, 1 eq) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (1.72 g, 6.79 mmol, 1.3 eq) in dioxane (40 mL) was added KOAc (1.54 g, 15.66 mmol, 3 eq) and Pd(dppf)Cl2(381.93 mg, 521.97 umol, 0.1 eq). After addition, the reaction mixture was stirred at 100° C. for 1 h under N2. LCMS (EB12-470-P1B1) showed desired MS. The reaction mixture was filtered and concentrated under reduced pressure to afford (2R,6S)-1-[2-(2,2-dimethoxyethoxy)ethyl]-2,6-dimethyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl]piperazine (3.8 g, 4.98 mmol, 95.40% yield) as a brown oil. Step 3: 3-[2-[(3R,5S)-4-[2-(2,2-dimethoxyethoxy)ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole A mixture of (2R,6S)-1-[2-(2,2-dimethoxyethoxy)ethyl]-2,6-dimethyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl]piperazine (3.8 g, 4.98 mmol, 1 eq), 3-bromo-5-nitro-1H-indazole (1.81 g, 7.47 mmol, 1.5 eq), 4-ditert-butylphosphanyl-N,N-dimethyl-aniline; dichloropalladium (352.66 mg, 498.05 umol, 352.66 uL, 0.1 eq) and KOAc (1.47 g, 14.94 mmol, 3 eq) in EtOH (30 mL) and H2O (6 mL) was stirred at 100° C. for 12 h under N2. LCMS (EB12-473-P1B) showed desired MS. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 15% methanol in dichloromethane) to afford 3-[2-[(3R,5S)-4-[2-(2,2-dimethoxyethoxy)ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole (2 g, 3.80 mmol, 76.24% yield, 92% purity) as a brown gum. Step 4: 2-[2-[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethoxy]acetaldehyde To a solution of 3-[2-[(3R,5S)-4-[2-(2,2-dimethoxyethoxy)ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole (700 mg, 1.44 mmol, 1 eq) in THF (5 mL) was added H2SO4(2 M, 4 mL) After addition, the reaction solution was stirred at 70° C. for 1 h. TLC (dichloromethane:methanol=10:1) showed starting material consumed and a new spot formed. After cooling, the reaction was diluted with water (10 mL) and washed with ethyl acetate (2×10 mL). The aqueous phase was raised to pH=14 with NaOH. The solid was collected by filtration. The solid was dried under reduced pressure to afford 2-[2-[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethoxy]acetaldehyde (500 mg, 1.04 mmol, 71.83% yield, 91% purity) as a yellow solid. The crude product was used for next step directly. Step 5: 5-[4-[2-[2-[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethoxy]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione To a solution of 2-[2-[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethoxy]acetaldehyde (250 mg, 570.15 umol, 1 eq) and AcOH (525.00 mg, 8.74 mmol, 0.5 mL, 15.33 eq) in DCE (10 mL) MeOH (10 mL) and DMSO (2 mL) was added 2-(2,6-dioxo-3-piperidyl)-5-piperazin-1-yl-isoindoline-1,3-dione (390.30 mg, 1.14 mmol, 2.00 eq) and NaOAc (140.32 mg, 1.71 mmol, 3 eq). The reaction solution was stirred at 25° C. for 30 min. Then NaBH3CN (107.49 mg, 1.71 mmol, 3 eq) was added and the reaction was stirred at 25° C. for 12 h. LCMS (EB12-480-P1D2) showed desired MS. TLC (dichloromethane:methanol=10:1) showed several new spots. The reaction solution was diluted with water (20 mL) and extracted with ethyl acetate (3×20 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 15% methanol in chloromethane) to afford 5-[4-[2-[2-[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethoxy]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (250 mg, 294.18 umol, 51.60% yield, 90% purity) as a yellow solid. Step 6: 5-[4-[2-[2-[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]ethoxy]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione To a mixture of 5-[4-[2-[2-[(2R,6S)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethoxy]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (250 mg, 326.87 umol, 1 eq) in EtOH (3 mL) and H2O (0.5 mL) was added Fe (91.27 mg, 1.63 mmol, 5 eq) and NH4Cl (87.42 mg, 1.63 mmol, 5 eq). After addition, the reaction mixture was stirred at 80° C. for 1 h. LCMS (EB12-481-P1C1) showed desired MS. The reaction mixture was filtered and filtrate was concentrated under reduced pressure. The resulting was dissolved with 10% methanol in dichloromethane (10 mL). The mixture was filtered and filtrate was concentrated under reduced pressure to afford 5-[4-[2-[2-[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]ethoxy]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (140 mg, 188.23 umol, 57.59% yield, 98.8% purity) as a yellow solid. Step 7: N-[3-[2-[(3S,5R)-4-[2-[2-[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]ethoxy]ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (Compound 3) To a solution of 5-[4-[2-[2-[(2S,6R)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]ethoxy]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (140 mg, 190.52 umol, 1 eq) and 4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (74.37 mg, 381.03 umol, 2 eq) in DMF (2 mL) was added DIEA (123.11 mg, 952.58 umol, 165.92 uL, 5 eq) and HATU (72.44 mg, 190.52 umol, 1 eq). After addition, the reaction solution was stirred at 25° C. for 12 h. LCMS (EB12-482-P1D) showed desired MS. The reaction solution was diluted with water (10 mL) and extracted with dichloromethane (3×10 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by prep.HPLC (column: Phenomenex luna C18 150*25 mm*10 um; mobile phase: [water (0.225% FA)-ACN]; B %: 0%-40%; 35 min) to afford N-[3-[2-[(3S,5R)-4-[2-[2-[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]ethoxy]ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (5.6 mg, 5.83 umol, 3.06% yield, 95% purity) as a yellow solid. 1H NMR: (400 MHz, CD3OD) δ: 8.65 (s, 1H), 8.40 (s, 1H), 8.17 (d, J=5.3 Hz, 1H), 7.52 (d, J=8.9 Hz, 1H), 7.38-7.32 (m, 2H), 7.24 (d, J=5.3 Hz, 1H), 7.19 (d, J=8.5 Hz, 1H), 6.93 (d, J=2.1 Hz, 1H), 6.75 (dd, J=2.1, 8.5 Hz, 1H), 5.30 (s, 2H), 5.02 (br dd, J=5.5, 12.8 Hz, 1H), 4.53 (br t, J=11.4 Hz, 2H), 3.92-3.81 (m, 4H), 3.67 (br t, J=4.8 Hz, 2H), 3.55 (br s, 2H), 3.51 (s, 3H), 3.26-3.20 (m, 4H), 3.04-2.94 (m, 2H), 2.92-2.81 (m, 1H), 2.76 (br d, J=2.8 Hz, 1H), 2.68-2.56 (m, 7H), 2.36 (s, 3H), 2.14-2.05 (m, 1H), 1.48 (br d, J=5.4 Hz, 6H) Exemplary Synthesis of Exemplary Compound 4: N-[3-[2-[(3S,5R)-4-[2-[2-[2-[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]ethoxy]ethoxy]ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide Compound 4 was prepared in a manner analogous to example 3. 1H NMR: (400 MHz, MeOD) δ: 8.71 (s, 1H), 8.27 (d, J=5.4 Hz, 1H), 7.55-7.48 (m, 2H), 7.41 (d, J=8.5 Hz, 1H), 7.38-7.31 (m, 2H), 6.99 (d, J=1.8 Hz, 1H), 6.87 (d, J=8.5 Hz, 1H), 5.29 (s, 2H), 5.05 (dd, J=5.4, 12.7 Hz, 1H), 4.55 (d, J=13.9 Hz, 2H), 3.85 (d, J=4.5 Hz, 2H), 3.79-3.53 (m, 10H), 3.51 (s, 3H), 3.22 (s, 4H), 3.05-2.93 (m, 2H), 2.91-2.51 (m, 9H), 2.34 (s, 3H), 2.16-2.07 (m, 1H), 1.49 (d, J=6.3 Hz, 6H). Exemplary Synthesis of Exemplary Compound 5: N-[3-[2-[(3S,5R)-4-[2-[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1: Tert-butyl 4-[2-[(2R,6S)-4-(4-bromo-2-pyridyl)-2,6-dimethyl-piperazin-1-yl]ethyl]piperazine-1-carboxylate To a solution of (3R,5S)-1-(4-bromo-2-pyridyl)-3,5-dimethyl-piperazine (1.5 g, 5.55 mmol, 1 eq) and tert-butyl 4-(2-chloroethyl)piperazine-1-carboxylate (2.07 g, 8.33 mmol, 3.58 mL, 1.5 eq) in NMP (20 mL) then was added Cs2CO3(5.43 g, 16.66 mmol, 3 eq) and KI (4.61 g, 27.76 mmol, 5.0 eq). Then the mixture was stirred at 140° C. for 16 h. TLC (Dichloromethane:Methanol=10:1, Rf=0.5) showed the reaction a new spot. The residue was diluted with H2O (50 mL) extracted with ethyl acetate (50 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 10% Methanol in Dichloromethane) to give tert-butyl 4-[2-[(2R,6S)-4-(4-bromo-2-pyridyl)-2,6-dimethyl-piperazin-1-yl]ethyl]piperazine-1-carboxylate (1.2 g, 2.26 mmol, 40.77% yield, 91% purity) as a yellow gum. Step 2: afford tert-butyl 4-[2-[(2R,6S)-2,6-dimethyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazine-1-carboxylate To a solution of tert-butyl 4-[2-[(2R,6S)-4-(4-bromo-2-pyridyl)-2,6-dimethyl-piperazin-1-yl]ethyl]piperazine-1-carboxylate (1.2 g, 2.49 mmol, 1 eq) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (1.26 g, 4.97 mmol, 2. eq) in dioxane (10 mL) was added KOAc (732.30 mg, 7.46 mmol, 3 eq), Pd(dppf)Cl2(182.00 mg, 248.73 umol, 0.1 eq). Then the mixture was stirred at 100° C. for 1 hr under N2. TLC (Dichloromethane:Methanol=10:1, Rf=0.3) showed the reaction a new spot. The reaction mixture was filtered and concentrated under reduced pressure to afford tert-butyl 4-[2-[(2R,6S)-2,6-dimethyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazine-1-carboxylate (1.3 g, crude) as a brown gum. Step 3: Tert-butyl 4-(2-((2R,6S)-2,6-dimethyl-4-(4-(5-nitro-1H-indazol-3-yl)pyridin-2-yl)piperazin-1-yl)ethyl)piperazine-1-carboxylate To a solution of tert-butyl 4-(2-((2R,6S)-2,6-dimethyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazin-1-yl)ethyl)piperazine-1-carboxylate (1.3 g, 2.46 mmol, 1 eq) and 3-bromo-5-nitro-1H-indazole (713.03 mg, 2.95 mmol, 1.2 eq) in EtOH (10 mL) and H2O (2 mL) was added KOAc (722.83 mg, 7.37 mmol, 3 eq), 4-ditert-butylphosphanyl-N,N-dimethyl-aniline; dichloropalladium (173.84 mg, 245.50 umol, 173.84 uL, 0.1 eq). Then the mixture was stirred at 100° C. for 2 hr under N2. TLC (Dichloromethane:Methanol=10:1, Rf=0.4) showed the reaction a new spot. The reaction mixture was poured into H2O (10 mL). The mixture was extracted with ethyl acetate (15 mL*3). The organic phase was washed with brine (10 mL), dried over anhydrous Na2SO4, concentrated in vacuum to give a residue. The residue was purified by silica gel column chromatography (0 to 15% Methanol in Dichloromethane) to give tert-butyl 4-(2-((2R,6S)-2,6-dimethyl-4-(4-(5-nitro-1H-indazol-3-yl)pyridin-2-yl)piperazin-1-yl)ethyl)piperazine-1-carboxylate (750 mg, 1.01 mmol, 41.12% yield, 76% purity) as a yellow solid. Step 4: 3-[2-[(3S,5R)-3,5-dimethyl-4-(2-piperazin-1-ylethyl)piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole To a solution of tert-butyl4-[2-[(2S,6R)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazine-1-carboxylate (750 mg, 1.33 mmol, 1 eq) in DCM (10 mL) was added TFA (12.32 g, 108.05 mmol, 8 mL, 81.35 eq). After addition, the reaction solution was stirred at 20° C. for 1 h. LCMS (EB12-483-P1B) showed desired MS. The reaction solution was diluted with water (50 mL) and washed with dichloromethane (2×30 mL). The aqueous phase was basified to pH˜14 with NaOH and extracted with ethyl acetate (3×50 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure to afford 3-[2-[(3S,5R)-3,5-dimethyl-4-(2-piperazin-1-ylethyl)piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole (450 mg, 852.42 umol, 64.18% yield, 88% purity) as a yellow solid. Step 5: 5-[4-[2-[(2S,6R)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione To a solution of 3-[2-[(3S,5R)-3,5-dimethyl-4-(2-piperazin-1-ylethyl)piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole (250 mg, 538.14 umol, 1 eq) and 2-(2,6-dioxo-3-piperidyl)-5-fluoro-isoindoline-1,3-dione (222.97 mg, 807.21 umol, 1.5 eq) in DMSO (3 mL) was added DIEA (208.65 mg, 1.61 mmol, 281.20 uL, 3 eq). After addition, the reaction was stirred at 100° C. for 12 h. LCMS (EB12-486-P1B1) showed desired MS. TLC (dichloromethane:methanol=10:1) showed several new spots. After cooling, the reaction solution was diluted with water (10 mL) and extracted with dichloromethane (3×10 mL). The organic layer was washed with brine (2×15 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 10% methanol in dichloromethane) to afford 5-[4-[2-[(2S,6R)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (150 mg, 168.57 umol, 31.32% yield, 81% purity) as a yellow solid. Step 6: 5-[4-[2-[(2S,6R)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione To a mixture of 5-[4-[2-[(2S,6R)-2,6-dimethyl-4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (150 mg, 208.11 umol, 1 eq) in EtOH (3 mL) and H2O (0.5 mL) was added Fe (58.11 mg, 1.04 mmol, 5 eq) and NH4Cl (55.66 mg, 1.04 mmol, 5 eq). After addition, the reaction was stirred at 80° C. for 1 h. LCMS (EB12-487-P1B1) showed the reaction was completed. After cooling, the reaction mixture was filtered and filtrate was concentrated under reduced pressure. The residue was triturated with 10% methanol in chloromethane at 20° C. for 15 min. The solid was removed by filtration and filtrate was concentrated under reduced pressure to afford 5-[4-[2-[(2S,6R)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (70 mg, 89.17 umol, 42.85% yield, 88% purity) as a yellow solid. Step 7: N-[3-[2-[(3S,5R)-4-[2-[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (Compound 5) To a solution of 5-[4-[2-[(2S,6R)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]ethyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (130 mg, 188.19 umol, 1 eq) and 4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylicacid (37.14 mg, 190.30 umol, 1.01 eq) in DMF (2 mL) was added DIEA (72.97 mg, 564.57 umol, 98.34 uL, 3 eq) and HATU (71.56 mg, 188.19 umol, 1 eq). After addition, the reaction solution was stirred at 25° C. for 18 h. LCMS (EB12-489-P1B72) showed desired MS. The reaction was diluted with water (5 mL) and extracted with ethyl acetate (3×5 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by prep.HPLC (column: PhenomenexLunaC18150*25 mm*10 um; mobile phase: [water (0.225% FA)-ACN]; B %: 0-30%; 35 min) to afford N-[3-[2-[(3S,5R)-4-[2-[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]ethyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5-dimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (10.7 mg, 12.26 umol, 6.51% yield, 99.42% purity) as a yellow solid. 1H NMR: (400 MHz, METHANOL-d4) δ: 8.69 (s, 1H), 8.41 (br s, 1H), 8.28 (d, J=5.3 Hz, 1H), 7.54-7.47 (m, 2H), 7.45 (s, 1H), 7.36 (d, J=6.6 Hz, 2H), 7.15 (s, 1H), 7.01 (br d, J=6.9 Hz, 1H), 5.29 (s, 2H), 5.08 (dd, J=5.5, 12.4 Hz, 1H), 4.59 (s, 3H), 4.43 (br s, 2H), 3.51 (s, 4H), 3.27 (br s, 4H), 3.02 (br t, J=12.6 Hz, 2H), 2.93-2.82 (m, 1H), 2.80-2.75 (m, 1H), 2.75-2.69 (m, 3H), 2.66 (br t, J=4.7 Hz, 4H), 2.35 (s, 3H), 2.19-2.05 (m, 1H), 1.44 (br d, J=4.9 Hz, 6H) Exemplary Synthesis of Exemplary Compound 6: N-(3-{2-[(3R,5S)-4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}-3,5-dimethylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7,7-tetramethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 6 was prepared in a manner analogous to Exemplary Compound 1 using Intermediate 3. Step 1 To a mixture of methyl 3-oxobutanoate (29 g, 249.75 mmol, 26.85 mL, 1 eq), ZnCl2 (5.11 g, 37.46 mmol, 1.75 mL, 0.15 eq) and acetone (21.76 g, 374.63 mmol, 27.54 mL, 1.5 eq) was added AC2O (33.15 g, 324.68 mmol, 30.41 mL, 1.3 eq) in one portion. The mixture was stirred at 50° C. for 48 hours to give yellow solution. TLC (Petroleum ether: Ethyl acetate=10:1, Rf=0.4) showed no start material and a new spot. The residue was diluted with H2O (200 mL) extracted with ethyl acetate (200 mL×3). The combined organic layers were washed with brine (75 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 10% Ethyl acetate in Petroleum ether) to give methyl 2-acetyl-3-methyl-but-2-enoate (18 g, 73.76 mmol, 29.53% yield, 64% purity) as a colorless oil. Step 2 To a mixture of methyl 2-acetyl-3-methyl-but-2-enoate (18 g, 115.25 mmol, 1 eq) and 1H-tetrazol-5-amine (9.80 g, 115.25 mmol, 1 eq) in EtOH (50 mL) and was added molecular sieves (10 g, 115.25 mmol, 1 eq) at 20° C. under N2. The mixture was stirred at 50° C. for 16 hours to give yellow solution. TLC (Petroleum ether: Ethyl acetate=10:1, Rf=0.1) showed no start material and a new spot. The reaction mixture was filtered and concentrated under reduced pressure. The crude product was poured into H2O (100 mL). The mixture was extracted with ethyl acetate (150 mL*3). The organic phase was washed with brine (80 mL), dried over anhydrous Na2SO4, concentrated in vacuum to give a residue. The residue was purified by silica gel column chromatography (0 to 30% Ethyl acetate in Petroleum ether) to give methyl 5,7,7-trimethyl-4H-tetrazolo[1,5-a]pyrimidine-6-carboxylate (11 g, 48.29 mmol, 41.90% yield, 98% purity) as a white solid. Step 3 To a mixture of methyl 5,7,7-trimethyl-4H-tetrazolo[1,5-a]pyrimidine-6-carboxylate (11 g, 49.28 mmol, 1 eq) in MeCN (100 mL) was added Mel (11.71 g, 82.50 mmol, 5.14 mL, 1.67 eq) in one portion and Cs2CO3(19.27 g, 59.13 mmol, 1.2 eq) at 20° C. under N2. The mixture was stirred at 50° C. for 1 hours to give yellow solution. The residue was diluted with H2O (200 mL) extracted with ethyl acetate (100 mL×3). The combined organic layers were washed with brine (85 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give methyl 4,5,7,7-tetramethyltetrazolo[1,5-a]pyrimidine-6-carboxylate (11 g, crude) as a yellow solid. Step 4 To a mixture of methyl 4,5,7,7-tetramethyltetrazolo[1,5-a]pyrimidine-6-carboxylate (640 mg, 2.70 mmol, 1 eq) in THF (5 mL) was added LiOH (129.33 mg, 5.40 mmol, 2 eq) in H2O (5 mL) at 20° C. The mixture was stirred at 50° C. for 16 hours to give yellow solution. TLC (Petroleum ether: Ethyl acetate=0:1, Rf=0.04) showed no start material. The THF was evaporated and the H2O solution was acidified for pH=3 with 1M HCl, extracted with EtOAc, dried over Na2SO4, filtered and concentrated under reduced pressure to give 4,5,7,7-tetramethyltetrazolo[1,5-a]pyrimidine-6-carboxylic acid (424 mg, crude) as a white solid. Step 5 To a mixture of 5-[4-[[4-[[(2R,6S)-4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-dimethyl-piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (97 mg, 125.50 umol, 1 eq) and 4,5,7,7-tetramethyltetrazolo[1,5-a]pyrimidine-6-carboxylic acid (45.68 mg, 188.24 umol, 92% purity, 1.5 eq) in DMF (5 mL) was added DIEA (48.66 mg, 376.49 umol, 65.58 uL, 3 eq) in one portion and HATU (47.72 mg, 125.50 umol, 1 eq) at 25° C. under N2. The mixture was stirred at 25° C. for 16 hours to give yellow solution. LCMS showed desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water (0.1M FANH4)-ACN]; B %: 0%-30%, 40 min) to afford N-[3-[2-[(3R,5S)-4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]-3,5-dimethyl-piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7,7-tetramethyl-tetrazolo[1,5-a]pyrimidine-6-carboxamide (36.1 mg, 36.18 umol, 28.83% yield, 98.04% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 8: (7R)—N-(3-{2-[(3S)-4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}-3-methylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 8 was prepared in a manner analogous to Exemplary Compound 1 using enantiomer 2 of 4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid. Step 1 The product 4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (9.5 g, 45.41 mmol, 1 eq) was separated by SFC (Column: DAICEL CHIRALPAK IC (250 mm*30 mm, 10 um); Condition: 0.1% NH3H2O ETOH; Begin B: 20; End B: 20; FlowRate: 100 mL/min) to give enantiomer 1 designated as (7S)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (4.68 g, 22.15 mmol, 48.77% yield, 99% purity) (Rt=3.080 min, 4.68 g) and enantiomer 2 designated as (7R)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (4.1 g, 19.40 mmol, 42.73% yield, 99% purity) (Rt=3.258 min, 4.1 g) both as an off-white solid. Enantiomer 1: Randomly assigned as (7S)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (4.68 g, 22.15 mmol, 48.77% yield, 99% purity) was obtained as an off-white solid (analytical chiral HPLC: ee %=100%, 4.11 min, α=−211). Enantiomer 2: Randomly assigned as (7R)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (4.1 g, 19.40 mmol, 42.73% yield, 99% purity) was obtained as a off-white solid (analytical chiral HPLC: ee %=100%, 4.68 min, α=266). Exemplary Synthesis of Exemplary Compound 9: (7S)—N-(3-{2-[(3S)-4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}-3-methylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 9 was prepared in a manner analogous to Exemplary Compound 8 using Enantiomer 1 of 4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid. Exemplary Synthesis of Exemplary Compound 10: N-(3-{2-[(3S)-4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}-3-methylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5-dimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 10 was prepared in a manner analogous to Exemplary Compound 1 using Intermediate 1. Exemplary Synthesis of Exemplary Compound 11: (7R)—N-{3-[2-(4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 11 was prepared in a manner analogous to Exemplary Compound 1 using Enantiomer 2 of 4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid. Exemplary Synthesis of Exemplary Compound 12: (7S)—N-{3-[2-(4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 12 was prepared in a manner analogous to Exemplary Compound 1 using Enantiomer 1 of 4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid. Exemplary Synthesis of Exemplary Compound 13: N-{3-[2-(4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5-dimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 13 was prepared in a manner analogous to Exemplary Compound 1 using Intermediate 1. Exemplary Synthesis of Exemplary Compound 14: (7R)—N-{3-[2-(4-{2-[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]ethyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a solution of 4-(2,2-dimethoxyethyl)piperidine (118.67 mg, 684.97 umol, 1.2 eq) and [1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl 4-methylbenzenesulfonate (300 mg, 570.81 umol, 1 eq) and DIEA (737.71 mg, 5.71 mmol, 994.22 uL, 10 eq) KI (947.54 mg, 5.71 mmol, 10 eq) in MeCN (10 mL). Then the mixture was stirred at 100° C. for 2 hours under N2. The residue was diluted with H2O 20 mL extracted with ethyl acetate (20 mL×3). The combined organic layers were washed with brine 15 mL, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 10% Methanol in Dichloromethane) to give 5-[4-[[4-(2,2-dimethoxyethyl)-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (280 mg, 515.74 umol, 90.35% yield, 97% purity) as a yellow solid. Step 2 To a solution of 5-[4-[[4-(2,2-dimethoxyethyl)-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (280 mg, 531.69 umol, 1 eq) in THF (2 mL) was added HCl (2 M, 1.51 mL, 5.69 eq) and stirred at 25° C. for 1 hour. The reaction mixture was poured into H2O (20 mL) and basified with aqueous NaHCO3till pH=8. The mixture was extracted with ethyl acetate (20 mL*5) and dried over anhydrous Na2SO4, concentrated in vacuum to give 2-[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]acetaldehyde (250 mg, crude) as a yellow solid. Step 3 To a solution of 2-[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]acetaldehyde (250 mg, 520.23 umol, 1 eq) and 5-nitro-3-(2-piperazin-1-yl-4-pyridyl)-1H-indazole (178.57 mg, 550.57 umol, 1.06 eq) in DCM (20 mL) and MeOH (30 mL) was added AcOH (3.12 mg, 52.02 umol, 2.98 uL, 0.1 eq) and the mixture was stirred at 20° C. for 20 minutes. Then the NaBH3CN (98.07 mg, 1.56 mmol, 3 eq) was added of the solution and was stirred at 20° C. for 16 hours. TLC (Dichloromethane:Methanol=5:1, Rf=0.2) showed no start material and a new spot. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 50% Methanol in Dichloromethane) to 2-(2,6-dioxo-3-piperidyl)-5-[4-[[4-[2-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]-1-piperidyl]methyl]-1-piperidyl]isoindoline-1,3-dione (400 mg, 344.79 umol, 66.28% yield, 68% purity) as a yellow solid. Step 4 To a solution of 2-(2,6-dioxo-3-piperidyl)-5-[4-[[4-[2-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]-1-piperidyl]methyl]-1-piperidyl]isoindoline-1,3-dione (400 mg, 507.04 umol, 1 eq) in EtOH (10. mL) and H2O (5 mL) was added NH4Cl (135.61 mg, 2.54 mmol, 5 eq), Fe (141.58 mg, 2.54 mmol, 5 eq). Then the mixture was stirred at 90° C. for 1 hour under N2. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (Phenomenex luna C18 100*40 mm*3 um: water (0.05% NH3H2O+10 mM NH4HCO3)-ACN; B %: 18%-48%, 10 min) to afford 5-[4-[[4-[2-[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (76 mg, 96.74 umol, 19.08% yield, 96.6% purity) as a yellow solid. Step 5 To a solution of 5-[4-[[4-[2-[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (45 mg, 59.30 umol, 1 eq) and (7R)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (12.40 mg, 59.30 umol, 1 eq) in DMF (2 mL) was stirred at 20° C. for 10 minutes, then was added DIEA (38.32 mg, 296.48 umol, 51.64 uL, 5 eq) and HATU (27.06 mg, 71.15 umol, 1.2 eq). Then the mixture was stirred at 70° C. for 3 hours under N2. LCMS showed desired product MS. The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: water (0.225% FA)-ACN; B %: 0%-30%, 35 min) to afford (7R)—N-[3-[2-[4-[2-[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]ethyl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (14.9 mg, 15.52 umol, 26.18% yield, 98.98% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 15: (7R)—N-(3-{2-[4-(3-{4-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperazin-1-yl}propyl)piperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a solution of 2-(2,6-dioxo-3-piperidyl)-5-piperazin-1-yl-isoindoline-1,3-dione (500 mg, 1.10 mmol, 1 eq, TFA) and 1-chloro-3-iodo-propane (1.12 g, 5.48 mmol, 589.42 uL, 5 eq) in CH3CN (5 mL) was added K2CO3(151.42 mg, 1.10 mmol, 1 eq). After addition, the reaction was stirred at 20° C. for 1 hour. TLC showed there was a new spot. The residue was poured into water (5 mL) and stirred for 5 minutes. The aqueous phase was extracted with ethyl acetate (5 mL*3). The combined organic phase was washed with brine (5 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (4 g, Rf=0.43, 100-200 mesh silica gel, 0-70% (10 min) of Ethyl acetate in Petroleum ether, 70% (10 min) Ethyl acetate in Petroleum ether) to give 5-[4-(3-chloropropyl)piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (260 mg, 564.85 umol, 51.56% yield, 91% purity) as a yellow gum. Step 2 To a mixture of (7R)-4,5,7-trimethyl-N-[3-(2-piperazin-1-yl-4-pyridyl)-1H-indazol-5-yl]-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (55 mg, 113.28 umol, 1 eq) and 5-[4-(3-chloropropyl)piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (47.45 mg, 113.28 umol, 1 eq) in MeCN (5 mL) was added KI (37.61 mg, 226.55 umol, 2 eq) and DIPEA (29.28 mg, 226.55 umol, 39.46 uL, 2 eq) in one portion at 20° C. under N2. The mixture was stirred at 80° C. for 10 hours. LCMS showed there was ˜78% of desired MS. The mixture was cooled to 20° C. and concentrated in reduced pressure at 20° C. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 mL*3). The combined organic phase was washed with brine (5 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The crude product was purified by reversed-phase HPLC (Column: 3_Phenomenex Luna C18 75*30 mm*3 um; Condition: water (0.2% LA)-ACN; Begin B: 0; End B: 30; FlowRate: 25 mL/min; Gradient Time: 35 minutes; 100% B Hold Time: 3 minutes) to give (7R)—N-[3-[2-[4-[3-[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]propyl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (19.5 mg, 21.34 umol, 18.84% yield, 95% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 17: (7R)—N-[3-(2-{4-[(1-{1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}azetidin-3-yl)methyl]piperazin-1-yl}pyridin-4-yl)-1H-indazol-5-yl]-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a solution of tert-butyl 3-(hydroxymethyl)azetidine-1-carboxylate (7.6 g, 40.59 mmol, 1 eq) in DCM (80 mL) was added HCl/MeOH (4 M, 38.00 mL, 3.74 eq), the mixture was stirred at 25° C. for 2 hours. TLC (Petroleum ether: Ethyl acetate=1:2, KMn04, Plate1) showed a new spot formed and the starting materials consumed completely. The reaction mixture was concentrated under reduced pressure to remove DCM and HCl/MeOH to give a residue. The crude product was used into the next step without further purification. Compound azetidin-3-ylmethanol (5.7 g, crude, HCl) was obtained as a light yellow liquid. Step 2 To a solution of azetidin-3-ylmethanol (2 g, 16.18 mmol, 1 eq, HCl) and tert-butyl 4-oxopiperidine-1-carboxylate (4.84 g, 24.28 mmol, 1.5 eq) in DCM (10 mL) and MeOH (2 mL) was added AcONa (6.64 g, 80.92 mmol, 5 eq) and HOAc (1.94 g, 32.37 mmol, 1.85 mL, 2 eq) for 30 minutes, after the mixture was added NaBH3CN (3.05 g, 48.55 mmol, 3 eq). Then the mixture was stirred 25° C. for 16 hours. TLC (Methanol:Dichloromethane=1:10, I2, Plate1) showed a new spot formed and the starting materials was remained. The reaction mixture was concentrated under reduced pressure to remove DCM. The residue was diluted with water 40 mL and extracted with EA (30 mL*4). The combined organic layers were washed with brine (60 mL), dried over Na2SO4filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-10% Methanol/Dichloromethane ether gradient @60 mL/min) to afford tert-butyl 4-[3-(hydroxymethyl)azetidin-1-yl]piperidine-1-carboxylate (2.2 g, 8.14 mmol, 50.28% yield) was obtained as a colorless gum. Step 3 To a solution of tert-butyl 4-[3-(hydroxymethyl)azetidin-1-yl]piperidine-1-carboxylate (2.2 g, 8.14 mmol, 1 eq) in MeOH (10 mL) was added HCl/dioxane (4 M, 10 mL). After addition, the reaction was stirred at 20° C. for 2 hours. LCMS showed desired mass (Rt=0.220 min). The reaction was concentrated under reduced pressure to afford [1-(4-piperidyl)azetidin-3-yl]methanol (1.8 g, crude, HCl) as a white solid. The crude product was used for next step directly. Step 4 To a solution of [1-(4-piperidyl)azetidin-3-yl]methanol (1.7 g, 8.22 mmol, 1 eq, HCl) in DMSO (20 mL) was added 2-(2,6-dioxo-3-piperidyl)-5-fluoro-isoindoline-1,3-dione (2.27 g, 8.22 mmol, 1 eq) and DIEA (3.19 g, 24.67 mmol, 4.30 mL, 3 eq). After addition, the reaction solution was stirred at 100° C. for 2 hours. LCMS showed desired mass. After cooling, the reaction was diluted with water (100 mL) and extracted with 10% methanol in dichloromethane (5×150 mL). The organic layer was dried over Na2SO4and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 20% methanol in dichloromethane) to afford 2-(2,6-dioxo-3-piperidyl)-5-[4-[3-(hydroxymethyl)azetidin-1-yl]-1-piperidyl]isoindoline-1,3-dione (1.4 g, 3.05 mmol, 37.12% yield, 93% purity) as a yellow solid. Step 5 To a solution of 2-(2,6-dioxo-3-piperidyl)-5-[4-[3-(hydroxymethyl)azetidin-1-yl]-1-piperidyl]isoindoline-1,3-dione (1.2 g, 2.81 mmol, 1 eq) in DCM (15 mL) was added TosCl (1.07 g, 5.63 mmol, 2 eq), DMAP (34.38 mg, 281.38 umol, 0.1 eq) and TEA (854.19 mg, 8.44 mmol, 1.17 mL, 3 eq). After addition, the reaction was stirred at 20° C. for 16 hours. LCMS showed desired MS. The reaction was diluted with water (30 mL) and extracted with dichloromethane (3×20 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 6% methanol in dichloromethane) to afford [1-[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]azetidin-3-yl]methyl 4-methylbenzenesulfonate (460 mg, 741.51 umol, 26.35% yield, 93.6% purity) as a yellow solid. Step 6 To a solution of [1-[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]azetidin-3-yl]methyl 4-methylbenzenesulfonate (77 mg, 132.61 umol, 1.07 eq) and (7R)-4,5,7-trimethyl-N-[3-(2-piperazin-1-yl-4-pyridyl)-1H-indazol-5-yl]-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (60 mg, 123.57 umol, 1 eq) in CH3CN (2 mL) was added KI (41.03 mg, 247.15 umol, 2 eq) and DIEA (31.94 mg, 247.15 umol, 43.05 uL, 2 eq). After addition, the reaction was stirred at 80° C. for 16 hours. LCMS showed desired mass. After cooling, the reaction mixture was filtered and the filter cake was washed with DMSO (2 mL). The filtrate was purified by prep.HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B %: 30%-60%; 35 min) to afford (7R)—N-[3-[2-[4-[[1-[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]azetidin-3-yl]methyl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (9.5 mg, 10.50 umol, 8.50% yield, 98.81% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 18: (7R)—N-(3-{2-[4-({1′-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]-[1,4′-bipiperidin]-4-yl}methyl)piperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a solution of 5-nitro-3-(2-piperazin-1-yl-4-pyridyl)-1H-indazole (1 g, 3.08 mmol, 1 eq) and tert-butyl 4-formylpiperidine-1-carboxylate (723.32 mg, 3.39 mmol, 1.1 eq) in MeOH (10 mL) and HOAc (1 mL) was added borane; 2-methylpyridine (659.57 mg, 6.17 mmol, 2 eq). After addition, the reaction mixture was stirred at 30° C. for 2 hours. LCMS showed desired mass. The reaction mixture was diluted with water (30 mL) and basified to pH 12 with solid NaOH. Then the mixture was filtered to afford filtrate cake. The residue was purified by silica gel column chromatography (0 to 10% methanol in dichloromethane) to afford tert-butyl 4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]piperidine-1-carboxylate (1.6 g, 2.99 mmol, 97.04% yield, 97.54% purity) as a yellow solid. Step 2 To a solution of tert-butyl 4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]piperidine-1-carboxylate (1.6 g, 3.07 mmol, 1 eq) in MeOH (10 mL) was added HCl/dioxane (4 M, 10 mL, 13.04 eq). After addition, the reaction solution was stirred at 20° C. for 16 hours. LCMS showed desired mass. The reaction was concentrated under reduced pressure. The resulting was dissolved in water (50 mL) and neutralized to pH 7 with solid NaOH. The solid was collected by filtration to afford 5-nitro-3-[2-[4-(4-piperidylmethyl)piperazin-1-yl]-4-pyridyl]-1H-indazole (860 mg, crude) as a yellow solid. The crude product was used for next step directly. Step 3 To a solution of 5-nitro-3-[2-[4-(4-piperidylmethyl)piperazin-1-yl]-4-pyridyl]-1H-indazole (860 mg, 2.04 mmol, 1 eq) and tert-butyl 4-oxopiperidine-1-carboxylate (2.03 g, 10.20 mmol, 5 eq) in MeOH (50 mL) and HOAc (5 mL) was added borane; 2-methylpyridine (654.72 mg, 6.12 mmol, 3 eq). After addition, the reaction solution was stirred at 35° C. for 12 hours. LCMS desired MS. The reaction was diluted with water (100 mL) and washed with ethyl acetate (100 mL). The aqueous phase was basified to pH 14 with solid KOH and extracted with ethyl acetate (3×50 mL). The aqueous phase was filtered to collect the solid. The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 50% methanol in dichloromethane) to afford tert-butyl 4-[4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]piperidine-1-carboxylate (750 mg, 1.14 mmol, 55.88% yield, 91.94% purity) as a yellow solid. Step 4 To a solution of tert-butyl 4-[4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]piperidine-1-carboxylate (750 mg, 1.24 mmol, 1 eq) in DCM (5 mL) was added TFA (3.08 g, 27.01 mmol, 2 mL, 21.78 eq). After addition, the reaction was stirred at 20° C. for 1 hour. LCMS showed desired MS. The reaction mixture was concentrated under reduced pressure to afford 5-nitro-3-[2-[4-[[1-(4-piperidyl)-4-piperidyl]methyl]piperazin-1-yl]-4-pyridyl]-1H-indazole (900 mg, crude, TFA) as a yellow gum. The crude product was used for next step directly. Step 5 To a solution of 5-nitro-3-[2-[4-[[1-(4-piperidyl)-4-piperidyl]methyl]piperazin-1-yl]-4-pyridyl]-1H-indazole (900 mg, 1.45 mmol, 1 eq, TFA) and 2-(2,6-dioxo-3-piperidyl)-5-fluoro-isoindoline-1,3-dione (602.76 mg, 2.18 mmol, 1.5 eq) in DMSO (10 mL) was added DIEA (940.08 mg, 7.27 mmol, 1.27 mL, 5 eq). After addition, the reaction was stirred at 100° C. for 12 hours. LCMS showed desired MS. After cooling, the reaction solution was diluted with water (100 mL). The solid was collected by filtration and dried under reduced pressure to afford 2-(2,6-dioxo-3-piperidyl)-5-[4-[4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]-1-piperidyl]isoindoline-1,3-dione (800 mg, 862.21 umol, 59.27% yield, 82% purity) as a yellow solid. The crude product was used for next step directly. Step 6 To a mixture of 2-(2,6-dioxo-3-piperidyl)-5-[4-[4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]-1-piperidyl]isoindoline-1,3-dione (400 mg, 525.74 umol, 1 eq) in EtOH (3 mL) and H2O (0.5 mL) was added Fe (146.80 mg, 2.63 mmol, 5 eq) and NH4Cl (140.61 mg, 2.63 mmol, 5 eq). After addition, the reaction was stirred at 80° C. for 2 hours. LCMS showed desired MS. After cooling, the reaction was filtered and filtrate was concentrated in vacuo. The resulting was dissolved in DMSO (10 mL) and then water (80 mL) was added. The solid was collected by filtration. The solid was dried under reduced pressure to afford 5-[4-[4-[[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (130 mg, 125.40 umol, 23.85% yield, 70.5% purity) as a yellow solid. Step 7 To a solution of (7R)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (37.21 mg, 177.87 umol, 1 eq) and 5-[4-[4-[[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (130 mg, 177.87 umol, 1 eq) in DMF (2 mL) was added DIEA (68.97 mg, 533.62 umol, 92.94 uL, 3 eq) and HATU (67.63 mg, 177.87 umol, 1 eq). After addition, the reaction solution was stirred at 25° C. for 16 hours. LCMS showed desired MS. The reaction was filtered and filtrate cake was washed with DMSO (1 mL). The filtrate was purified by prep.HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water (0.225% FA)-ACN]; B %: 0-30%; 35 min) to afford (7R)—N-[3-[2-[4-[[1-[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]-4-piperidyl]methyl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (10.7 mg, 11.45 umol, 6.44% yield, 98.66% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 20: (7R)—N-[3-(2-{4-[2-(2-{6-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]-2,6-diazaspiro[3.3]heptan-2-yl}ethoxy)ethyl]piperazin-1-yl}pyridin-4-yl)-1H-indazol-5-yl]-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 20 was prepared in a manner analogous to Exemplary Compound 3. Exemplary Synthesis of Exemplary Compound 21: (7S)—N-{3-[2-(4-{2-[4-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperazin-1-yl]ethyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a solution of 5-nitro-3-(2-piperazin-1-yl-4-pyridyl)-1H-indazole (500 mg, 1.54 mmol, 1 eq) and tert-butyl 4-(2-chloroethyl)piperazine-1-carboxylate (500 mg, 2.01 mmol, 1.3 eq) in CH3CN (10 mL) was added KI (511.82 mg, 3.08 mmol, 2 eq) and DIEA (398.48 mg, 3.08 mmol, 537.04 uL, 2 eq). After addition, the reaction was stirred at 80° C. for 3 hours. LCMS showed desired MS. After cooling, the reaction mixture was diluted with water (10 mL) and extracted with dichloromethane (3×10 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 10% methanol in dichloromethane) to afford tert-butyl 4-[2-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazine-1-carboxylate (303 mg, 558.99 umol, 36.26% yield, 99% purity) as a yellow solid. Step 2 To a solution of tert-butyl 4-[2-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazine-1-carboxylate (303 mg, 564.64 umol, 1 eq) in DCM (3 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 11.96 eq). After addition, the reaction was stirred at 20° C. for 1 hour. LCMS showed reactant consumed and desired MS was detected. The reaction mixture was concentrated in vacuo. The resulting was dissolved in DCM (2 mL) and treated with DIEA (1 mL). The mixture was concentrated under reduced pressure to afford 5-nitro-3-[2-[4-(2-piperazin-1-ylethyl)piperazin-1-yl]-4-pyridyl]-1H-indazole (240 mg, crude) as a yellow solid. The crude product was used for next step directly. Step 3 To a solution of 5-nitro-3-[2-[4-(2-piperazin-1-ylethyl)piperazin-1-yl]-4-pyridyl]-1H-indazole (240 mg, 549.82 umol, 1 eq) and 1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperidine-4-carbaldehyde (203.09 mg, 549.82 umol, 1 eq) in MeOH (10 mL) and HOAc (1 mL) was added borane; 2-methylpyridine (117.62 mg, 1.10 mmol, 2 eq). After addition, the reaction solution was stirred at 20° C. for 2 hour. LCMS showed starting material consumed and desired MS was detected. The reaction was diluted with water (20 mL) and extracted with dichloromethane (3×15 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 40% methanol in dichloromethane) to afford 2-(2,6-dioxo-3-piperidyl)-5-[4-[[4-[2-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazin-1-yl]methyl]-1-piperidyl]isoindoline-1,3-dione (445 mg, 523.38 umol, 95.19% yield, 92.9% purity) as a yellow solid. Step 4 To a solution of 2-(2,6-dioxo-3-piperidyl)-5-[4-[[4-[2-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazin-1-yl]methyl]-1-piperidyl]isoindoline-1,3-dione (445 mg, 563.38 umol, 1 eq) in EtOH (5 mL) and H2O (1 mL) was added Fe (157.31 mg, 2.82 mmol, 5 eq) and NH4Cl (150.68 mg, 2.82 mmol, 5 eq). After addition, the reaction was stirred at 80° C. for 1 hour. LCMS showed desired MS. After cooling, the reaction mixture was filtered and filtrate was concentrated in vacuo. The residue was triturated with 10% methanol in dichloromethane at 20° C. for 10 minutes. The resulting mixture was filtered and the filtrate was concentrated under reduced pressure to afford 5-[4-[[4-[2-[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazin-1-yl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (120 mg, 138.97 umol, 24.67% yield, 88% purity) as a yellow solid. Step 5 To a solution of 5-[4-[[4-[2-[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]piperazin-1-yl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (55 mg, 72.38 umol, 1 eq) and (7S)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (17 mg, 81.26 umol, 1.12 eq) in DMF (2 mF) was added DIEA (46.77 mg, 361.89 umol, 63.03 uL, 5 eq) and HATU (27.52 mg, 72.38 umol, 1 eq). After addition, the reaction was stirred at 25° C. for 12 hours. LCMS showed desired MS. The reaction was diluted with water (10 mF) and extracted with dichloromethane (3×15 mF). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by prep.HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water (0.225% FA)-ACN]; B %: 0-35%; 35 min) to afford (7S)—N-[3-[2-[4-[2-[4-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]piperazin-1-yl]ethyl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (12.8 mg, 13.11 umol, 18.11% yield, 97.40% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 22: (7R)—N-{3-[2-(4-{2-[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]-2,2-difluoroethyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 22 was prepared in a manner analogous to Exemplary Compound 1 using tert-butyl 4-[2-[4-(4-bromo-2-pyridyl)piperazin-1-yl]-1,1-difluoro-ethyl]piperidine-1-carboxylate. Step 1 To a solution of tert-butyl 4-(2-bromoacetyl)piperidine-1-carboxylate (2.78 g, 9.08 mmol, 1 eq) in MeCN (10 mL) was stirred at 20° C. Then the mixture was added benzyl piperazine-1-carboxylate (2 g, 9.08 mmol, 1.75 mL, 1 eq) and stirred at 20° C. for 16 hr under N2. TLC (Dichloromethane:Methanol=10:1, Rf=0.6) showed no start material and a new spot. The residue was diluted with H2O (200 mL) extracted with ethyl acetate (70 mL×3). The combined organic layers were washed with brine (80 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 10% Dichloromethane in Methanol) to give benzyl 4-[2-(1-tert-butoxycarbonyl-4-piperidyl)-2-oxo-ethyl]piperazine-1-carboxylate (1.9 g, 3.84 mmol, 42.27% yield, 90% purity) as a yellow gum. Step 2 To a solution of benzyl 4-[2-(1-tert-butoxycarbonyl-4-piperidyl)-2-oxo-ethyl]piperazine-1-carboxylate (1.9 g, 4.26 mmol, 1 eq) in DCM (30 mL) was stirred at 0° C. for 20 min. Then the mixture was added DAST (24.06 g, 149.25 mmol, 19.72 mL, 35 eq) and stirred at 20° C. for 16 hours under N2. TLC (Petroleum ether: Ethyl acetate=1:1, Rf=0.5) showed no start material and a new spot. The reaction was cooled to 0° C. and quenched with aqueous NaHCO3(200 mL) extracted with ethyl acetate (100 mL×2). The combined organic layers were washed with brine (45 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 50% Ethyl acetate in Petroleum ether) to give benzyl 4-[2-(1-tert-butoxycarbonyl-4-piperidyl)-2,2-difluoro-ethyl]piperazine-1-carboxylate (1.4 g, 2.55 mmol, 59.68% yield, 85% purity) as a Colorless gum. Step 3 To a solution of benzyl 4-[2-(1-tert-butoxycarbonyl-4-piperidyl)-2,2-difluoro-ethyl]piperazine-1-carboxylate (1.4 g, 2.99 mmol, 1 eq) in EtOH (10 mL) and EtOAc (10 mL). Then the mixture was added Pd/C (0.2 g, 299.43 umol, 10% purity, 0.1 eq) and stirred at 25° C. for 16 hours under H2(15 PSI). TLC (Petroleum ether: Ethyl acetate=1:1, Rf=0.01) showed no start material and a new spot. The reaction was filtered and concentrated under reduced pressure to give tert-butyl 4-(1,1-difluoro-2-piperazin-1-yl-ethyl)piperidine-1-carboxylate (900 mg, crude) as a colorless gum. Step 4 To a solution of tert-butyl 4-(1,1-difluoro-2-piperazin-1-yl-ethyl)piperidine-1-carboxylate (900 mg, 2.70 mmol, 1 eq) and 4-bromo-2-fluoro-pyridine (500 mg, 2.84 mmol, 1.05 eq) and K2CO3(746.15 mg, 5.40 mmol, 2 eq) in DMSO (15 mL). Then the mixture was stirred at 100° C. for 4 hours under N2. TLC (Petroleum ether: Ethyl acetate=3:1, Rf=0.5) showed no start material and a new spot. The residue was diluted with H2O (50 mL) extracted with ethyl acetate (60 mL×3). The combined organic layers were washed with brine 40 mL, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 30% Ethyl acetate in Petroleum ether) to give tert-butyl 4-[2-[4-(4-bromo-2-pyridyl)piperazin-1-yl]-1,1-difluoro-ethyl]piperidine-1-carboxylate (1 g, 1.94 mmol, 71.91% yield, 95% purity) as a white solid. Exemplary Synthesis of Exemplary Compound 23: (7R)—N-{3-[2-(4-{2-[4-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-1-yl]ethyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 23 was prepared in a manner analogous to Exemplary Compound 21 using enantiomer 2 of 4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid. Step 1 To a solution of tert-butyl 4-methylenepiperidine-1-carboxylate (6.24 g, 31.65 mmol, 1 eq) was added 9-BBN (0.5 M, 63.29 mL, 1 eq) at 25° C. The reaction mixture was stirred at 80° C. for 1 hour under N2. After cooling, 4-bromopyridine (5 g, 31.65 mmol, 1 eq), Pd(dppf)Cl2 (1.39 g, 1.90 mmol, 0.06 eq), K2CO3 (6.56 g, 47.47 mmol, 1.5 eq), DMF (50 mL) and H2O (5 mL) were added to the reaction. The resultant mixture was heated to 60° C. for 12 hours. LCMS showed desired MS. After cooling, the reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (3×100 mL). The organic layer was washed with brine (2×100 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 40% ethyl acetate in petroleum ether) to afford tert-butyl 4-(4-pyridylmethyl)piperidine-1-carboxylate (3.49 g, 11.66 mmol, 36.83% yield, 92.3% purity) as a pale yellow oil. Step 2 To a solution of tert-butyl 4-(4-pyridylmethyl)piperidine-1-carboxylate (3.49 g, 12.63 mmol, 1 eq) in EtOH (50 mL) and HOAc (758.33 mg, 12.63 mmol, 722.21 uL, 1 eq) was added PtO2(430.12 mg, 1.89 mmol, 0.15 eq) at 25° C. Then the mixture was stirred at 70° C. for 24 hours under H2(50 psi). TLC (PE:EA=1:1) showed starting material consumed and a new spot formed. After cooling, the reaction was filtered and filtrate was concentrated under reduced pressure to afford tert-butyl 4-(4-piperidylmethyl)piperidine-1-carboxylate (3.9 g, crude) as a brown oil. Step 3 To a solution of tert-butyl 4-(4-piperidylmethyl)piperidine-1-carboxylate (3.7 g, 13.10 mmol, 1.21 eq) and 2-(2,6-dioxo-3-piperidyl)-5-fluoro-isoindoline-1,3-dione (3 g, 10.86 mmol, 1 eq) in DMSO (40 mL) was added DIEA (5.61 g, 43.44 mmol, 7.57 mL, 4 eq). After addition, the reaction mixture was stirred at 100° C. for 2 hours. TLC (petroleum ether: ethyl acetate=1:1) showed a new spot. After cooling, the reaction was diluted with ethyl acetate (200 mL) and washed with brine (3×100 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 50% ethyl acetate in petroleum ether) to afford tert-butyl 4-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]piperidine-1-carboxylate (2.86 g, 4.41 mmol, 40.58% yield, 83% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 24: (7R)—N-[3-(2-{4-[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)azetidin-3-yl]piperazin-1-yl}pyridin-4-yl)-1H-indazol-5-yl]-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a solution of tert-butyl 3-oxoazetidine-1-carboxylate (170 mg, 993.03 umol, 1 eq) and 5-nitro-3-(2-piperazin-1-yl-4-pyridyl)-1H-indazole (305.97 mg, 943.38 umol, 0.95 eq) in HOAc (1 mL) and MeOH (10 mL) was stirred at 20° C. for 20 minutes, then was added borane; 2-methylpyridine (212.43 mg, 1.99 mmol, 2 eq). Then the mixture was stirred at 30° C. for 16 hours under N2. TLC (Dichloromethane:Methanol=10:1, Rf=0.3) showed no start material and a new spot. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 20% Dichloromethane in Methanol) to give tert-butyl 3-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]azetidine-1-carboxylate (460 mg, 774.13 umol, 77.96% yield, 80.7% purity) as a yellow solid. Step 2 To a solution of tert-butyl 3-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]azetidine-1-carboxylate (460 mg, 959.27 umol, 1 eq) in DCM (3 mL) was added TFA (4.25 g, 37.28 mmol, 2.76 mL, 38.86 eq) and then stirred at 20° C. for 1 hr. TLC (Dichloromethane:Methanol=5:1, Rf=0.01) showed no start material and a new spot. The residue was concentrated under reduced pressure to give 3-[2-[4-(azetidin-3-yl)piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole (360 mg, crude, TFA) as a yellow solid. Step 3 To a solution of 1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidine-4-carbaldehyde (292.06 mg, 790.69 umol, 1 eq) and 3-(2-(4-(azetidin-3-yl)piperazin-1-yl)pyridin-4-yl)-5-nitro-1H-indazole (300.00 mg, 790.69 umol, 1 eq) in HOAc (1 mL) and MeOH (20 mL) was stirred at 20° C. for 20 minutes, then was added borane; 2-methylpyridine (169.15 mg, 1.58 mmol, 2 eq). Then the mixture was stirred at 30° C. for 16 hours under N2. TLC (Dichloromethane:Methanol=5:1, Rf=0.1) showed no start material and a new spot. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 50% Dichloromethane in Methanol) to give 2-(2,6-dioxopiperidin-3-yl)-5-(4-((3-(4-(4-(5-nitro-1H-indazol-3-yl)pyridin-2-yl)piperazin-1-yl)azetidin-1-yl)methyl)piperidin-1-yl)isoindoline-1,3-dione (360 mg, 412.67 umol, 52.19% yield, 84% purity) as a yellow solid. Step 4 To a solution of 2-(2,6-dioxo-3-piperidyl)-5-[4-[[3-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]azetidin-1-yl]methyl]-1-piperidyl]isoindoline-1,3-dione (360 mg, 491.28 umol, 1 eq) in EtOH (10 mL) and H2O (5 mL) was added Fe (137.18 mg, 2.46 mmol, 5 eq), NH4Cl (131.39 mg, 2.46 mmol, 5 eq). Then the mixture was stirred at 90° C. for 1 hour under N2. LCMS showed desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (Phenomenex Gemini-NX 150*30 mm*5 um: water (0.05% HCl)-ACN; B %: 10%-40%, 10 min) to afford 5-[4-[[3-[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]azetidin-1-yl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (177 mg, 231.70 umol, 47.16% yield, 92% purity) as a yellow solid. Step 5 To a solution of 5-[4-[[3-[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]azetidin-1-yl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (78 mg, 110.98 umol, 1 eq) and (7R)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (23.22 mg, 110.98 umol, 1 eq) in DMF (4 mL) was stirred at 20° C. for 10 minutes, then was added DIEA (71.72 mg, 554.92 umol, 96.65 uL, 5 eq) and HATU (42.20 mg, 110.98 umol, 1 eq). Then the mixture was stirred at 30° C. for 16 hours under N2. LCMS showed desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water (0.225% FA)-ACN]; B %: 0%-30%, 35 min) to give (7R)—N-[3-[2-[4-[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]azetidin-3-yl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (19.1 mg, 21.21 umol, 19.11% yield, 99.27% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 25: (7R)—N-{3-[2-(4-{2-[4-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperazin-1-yl]ethyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a solution of tert-butyl 4-(2-chloroethyl)piperazine-1-carboxylate (200 mg, 804.02 umol, 1 eq) in DCM (2 mL) added TFA (3.08 g, 27.01 mmol, 2 mL, 33.60 eq) and then the mixture was stirred at 20° C. for 1 hour. TLC (Dichloromethane:Methanol=10:1, Rf=0.02) showed no start material and a new spot. The residue was concentrated under reduced pressure to give 1-(2-chloroethyl)piperazine (119 mg, crude, TFA) as a Colorless oil. Step 2 To a solution of 1-(2-chloroethyl)piperazine (119 mg, 800.63 umol, 1 eq) and 1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperidine-4-carbaldehyde (295.73 mg, 800.63 umol, 1 eq) in HOAC (1 mL) and MeOH (10 mL) was stirred at 20° C. for 20 min, then was added borane; 2-methylpyridine (171.27 mg, 1.60 mmol, 2 eq). Then the mixture was stirred at 30° C. for 16 hours under N2. TLC (Dichloromethane:Methanol=10:1, Rf=0.3) showed no start material and a new spot. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0 to 25% Dichloromethane in Methanol) to give 5-[4-[[4-(2-chloroethyl)piperazin-1-yl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (390 mg, 769.12 umol, 96.06% yield, 99% purity) as a yellow solid. Step 3 To a solution of (7R)-4,5,7-trimethyl-N-[3-(2-piperazin-1-yl-4-pyridyl)-1H-indazol-5-yl]-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (40 mg, 82.38 umol, 1 eq) and 5-[4-[[4-(2-chloroethyl)piperazin-1-yl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (45 mg, 89.64 umol, 1.09 eq) and DIEA (106.47 mg, 823.82 umol, 143.49 uL, 10 eq) and KI (136.75 mg, 823.82 umol, 10 eq) in MeCN (10 mL). Then the mixture was stirred at 80° C. for 4 hours under N2. LCMS showed desired product. The residue was diluted with H2O (20 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layers were washed with brine (15 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water (0.225% LA)-ACN]; B %: 0%-30%, 35 min) to afford (7R)—N-[3-[2-[4-[2-[4-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]piperazin-1-yl]ethyl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (8.8 mg, 9.04 umol, 10.97% yield, 97.66% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 26: (7R)—N-(3-{2-[6-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)-2,6-diazaspiro[3.3]heptan-2-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a mixture of 4-bromo-2-fluoro-pyridine (1 g, 5.68 mmol, 1 eq) and tert-butyl 2,6-diazaspiro[3.3]heptane-2-carboxylate; oxalic acid (1.38 g, 2.84 mmol, 0.5 eq) in DMSO (10 mL) was added K2CO3(2.36 g, 17.04 mmol, 70.14 mL, 3 eq) in one portion at 100° C. under N2. The mixture was stirred at 100° C. for 2 hours to give yellow solution. TLC and LCMS showed the reaction was completed. The mixture was cooled to 20° C. and concentrated in reduced pressure at 20° C. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL*4). The combined organic phase was washed with brine (10 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (Petroleum ether: Ethyl acetate=10:1, Rf=0.56, 12 g, 0-50% (10 min) of Ethyl acetate in Petroleum ether, 50% (5 min) of Ethyl acetate in Petroleum ether) to give tert-butyl 6-(4-bromo-2-pyridyl)-2,6-diazaspiro[3.3]heptane-2-carboxylate (1.3 g, 3.67 mmol, 64.61% yield) as a white solid. Step 2 To a mixture of 2-[(3-bromo-5-nitro-indazol-1-yl)methoxy]ethyl-trimethyl-silane (2 g, 5.37 mmol, 1 eq), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (2.05 g, 8.06 mmol, 1.5 eq) and Pd(dppf)Cl2(196.54 mg, 268.61 umol, 0.05 eq) in dioxane (20 mL) was added KOAc (1.58 g, 16.12 mmol, 3 eq) in one portion at 25° C. under N2. The mixture was stirred at 100° C. for 1 hour. TLC showed the reaction was completed. The mixture was cooled to 25° C., filtered and concentrated in vacuum to give [5-nitro-1-(2-trimethylsilylethoxymethyl)indazol-3-yl]boronic acid (1.8 g, crude) as a black brown solid. The crude product was used into the next step without purification. Step 3 To a mixture of tert-butyl 6-(4-bromo-2-pyridyl)-2,6-diazaspiro[3.3]heptane-2-carboxylate (1.3 g, 3.67 mmol, 1 eq), [5-nitro-1-(2-trimethylsilylethoxymethyl)indazol-3-yl]boronic acid (1.73 g, 5.14 mmol, 1.4 eq) and 4-ditert-butylphosphanyl-N,N-dimethyl-aniline; dichloropalladium (259.85 mg, 366.98 umol, 259.85 uL, 0.1 eq) in EtOH (10 mL) and H2O (4 mL) was added KOAc (1.08 g, 11.01 mmol, 3 eq) in one portion at 25° C. under N2. The mixture was stirred at 100° C. for 2 hours. LCMS showed the reaction was completed. The mixture was cooled to 20° C. and concentrated in reduced pressure. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL*3). The combined organic phase was washed with brine (10 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (20 g, 0-40% (10 min) of Ethyl acetate in Petroleum ether, 40% (10 min) of Ethyl acetate in Petroleum ether) to give tert-butyl 6-[4-[5-nitro-1-(2-trimethylsilylethoxymethyl)indazol-3-yl]-2-pyridyl]-2,6-diazaspiro[3.3]heptane-2-carboxylate (1.2 g, 2.12 mmol, 57.70% yield) as a yellow solid. Step 4 To a mixture of tert-butyl 6-[4-[5-nitro-1-(2-trimethylsilylethoxymethyl)indazol-3-yl]-2-pyridyl]-2,6-diazaspiro[3.3]heptane-2-carboxylate (600 mg, 1.06 mmol, 1 eq) in DCM (5 mL) was added TFA (362.16 mg, 3.18 mmol, 235.17 uL, 3 eq) in one portion at 20° C. The mixture was stirred at 20° C. for 2 hours. The mixture added dioxane (10 mL) and NH3.H2O (556.55 mg, 6.35 mmol, 611.60 uL, 40% purity, 6 eq) in one portion at 20° C., The mixture was stirred at 20° C. for 2 hours. LCMS showed the reaction was completed. The residue was poured into water (5 mL), the aqueous phase was extracted with DCM (5 mL*3). The combined organic phase was washed with brine (5 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum to give 3-[2-(2,6-diazaspiro[3.3]heptan-2-yl)-4-pyridyl]-5-nitro-1H-indazole (300 mg, 561.92 umol, 53.08% yield, 63% purity) as a yellow solid. Step 5 To a mixture of 3-[2-(2,6-diazaspiro[3.3]heptan-2-yl)-4-pyridyl]-5-nitro-1H-indazole (300 mg, 891.94 umol, 1 eq) and 1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperidine-4-carbaldehyde (329.45 mg, 891.94 umol, 1 eq) in MeOH (10 mL) was added CH3COOH (53.56 mg, 891.94 umol, 51.01 uL, 1 eq) and borane; 2-methylpyridine (190.80 mg, 1.78 mmol, 2 eq) in one portion at 20° C. under N2. The mixture was stirred at 30° C. for 16 hours. LCMS showed there was desired MS. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL*3). The combined organic phase was washed with brine (10 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (Dichloromethane:Methanol=10:1, Rf=0.32, 0-10% (10 min) of Methanol in Dichloromethane, 10% (5 min) of Methanol in Dichloromethane) to give 2-(2,6-dioxo-3-piperidyl)-5-[4-[[2-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]-2,6-diazaspiro[3.3]heptan-6-yl]methyl]-1-piperidyl]isoindoline-1,3-dione (600 mg, 565.45 umol, 63.40% yield, 65% purity) as a yellow gum. Step 6 To a mixture of 2-(2,6-dioxo-3-piperidyl)-5-[4-[[2-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]-2,6-diazaspiro[3.3]heptan-6-yl]methyl]-1-piperidyl]isoindoline-1,3-dione (600 mg, 869.92 umol, 1 eq) and Fe (242.90 mg, 4.35 mmol, 5 eq) in EtOH (10 mL), H2O (5 mL) and DCM (5 mL) was added NH4Cl (232.67 mg, 4.35 mmol, 5 eq) in one portion at 20° C. under N2. The mixture was stirred at 80° C. for 30 minutes. LCMS showed the reaction was completed. The residue was filtered with ethyl acetate (10 mL*5), and solution was concentrated in vacuum. The crude product was purified by reversed-phase HPLC (Column: YMC-Triart Prep C18 150*40 mm*7 um; Condition: water (0.05% HCl)-ACN; Begin B: 5; End B: 35; FlowRate: 60 mL/min; Gradient Time: 25 min; 100% B Hold Time: 2 min) to give 5-(4-((6-(4-(5-amino-1H-indazol-3-yl)pyridin-2-yl)-2,6-diazaspiro[3.3]heptan-2-yl)methyl)piperidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (110 mg, 166.07 umol, 19.09% yield, 99.6% purity) as a yellow solid. Step 7 To a solution of (7R)-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxylic acid (34.88 mg, 166.73 umol, 1 eq) and 5-[4-[[2-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]-2,6-diazaspiro[3.3]heptan-6-yl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (110 mg, 166.73 umol, 1 eq) in DMF (2 mL) was stirred at 0° C. for 10 minutes, then was added DIPEA (107.74 mg, 833.67 umol, 145.21 uL, 5 eq) and HATU (63.40 mg, 166.73 umol, 1 eq), Then the mixture was stirred at 25° C. for 16 hours under N2. LCMS showed desired MS. The residue was poured into water (2 mL). The aqueous phase was extracted with ethyl acetate (2 mL*3). The combined organic phase was washed with brine (2 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The crude product was purified by reversed-phase HPLC (Column: 3_Phenomenex Luna C18 75*30 mm*3 um; Condition: water (0.225% FA)-ACN; Begin B: 0; End B: 30; FlowRate: 25 mL/min; Gradient Time: 35 minutes; 100% B Hold Time: 3 minutes) to give (7R)—N-[3-[2-[6-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-2,6-diazaspiro[3.3]heptan-2-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7-trimethyl-7H-tetrazolo[1,5-a]pyrimidine-6-carboxamide (29 mg, 34.01 umol, 20.40% yield, 99.78% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 27: (7S)—N-{3-[2-(4-{2-[4-({4-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperazin-1-yl}methyl)piperidin-1-yl]ethyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamideuors Step 1 To a solution of 2-(2,6-dioxo-3-piperidyl)-5-piperazin-1-yl-isoindoline-1,3-dione (2 g, 4.38 mmol, 1 eq, TFA) and tert-butyl 4-formylpiperidine-1-carboxylate (1.40 g, 6.57 mmol, 1.5 eq) in MeOH (20 mL) was added NaOAc (1.08 g, 13.15 mmol, 3 eq), HOAc (131.59 mg, 2.19 mmol, 125.32 uL, 0.5 eq) and NaBH3CN (826.20 mg, 13.15 mmol, 3 eq) in one portion at 20° C. under N2. The solution was stirred at 20° C. for 1 hour. TLC (DCM:MeOH=10:1, Rf=0.43) showed the reaction was completed, and a main spot (Rf=0.43) was showed on TLC. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (3×20 mL). The combined organic phase was washed with brine (2×20 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (column: 40 g, 100-200 mesh silica gel, 0-50% (5 min) of Ethyl acetate in Petroleum ether, 50-100% (10 min) of Ethyl acetate in Petroleum ether) to give tert-butyl 4-[[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]methyl]piperidine-1-carboxylate (2.2 g, 3.99 mmol, 90.98% yield, 97.8% purity) as a yellow gum. Step 2 To a solution of tert-butyl 4-[[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]methyl]piperidine-1-carboxylate (2.2 g, 4.08 mmol, 1 eq) in DCM (2 mL) was added TFA (1.39 g, 12.23 mmol, 905.58 uL, 3 eq) in one portion at 20° C. under N2. The mixture was stirred at 20° C. for 30 minutes to give yellow solution. TLC (DCM:MeOH=10:1, Rf=0.43) showed the starting material was completed. The solution was concentrated in vacuum to give 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidylmethyl)piperazin-1-yl]isoindoline-1,3-dione (3 g, 3.73 mmol, 91.61% yield, 97.3% purity, 3TFA) as a yellow gum. Step 3 To a mixture of 5-nitro-3-(2-piperazin-1-yl-4-pyridyl)-1H-indazole (500 mg, 1.54 mmol, 1 eq) and 2-chloroacetaldehyde (520 mg, 2.65 mmol, 426.23 uL, 40% purity, 1.72 eq) in DCE (20 mL) and MeOH (20 mL) was added CH3COOH (0.1 mL) and NaBH3CN (290.63 mg, 4.62 mmol, 3 eq) in one portion at 20° C. under N2. The mixture was stirred at 20° C. for 2 hours. LCMS showed there was desired MS. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL*3). The combined organic phase was washed with brine (10 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (Dichloromethane:Methanol=10:1, Rf=0.27, 0-50% (15 min) of Ethyl acetate in Petroleum ether, 10% (5 min) of Ethyl acetate in Petroleum ether) to give 3-[2-[4-(2-chloroethyl)piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole (350 mg, 904.78 umol, 58.69% yield) as a yellow solid. Step 4 To a mixture of 3-[2-[4-(2-chloroethyl)piperazin-1-yl]-4-pyridyl]-5-nitro-1H-indazole (350 mg, 904.78 umol, 1 eq) and 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidylmethyl)piperazin-1-yl]isoindoline-1,3-dione (397.66 mg, 508.79 umol, 5.62e-1 eq, 3TFA) in MeCN (10 mL) was added KI (300.39 mg, 1.81 mmol, 2 eq) and DIPEA (233.87 mg, 1.81 mmol, 315.19 uL, 2 eq) in one portion at 20° C. under N2. The mixture was stirred at 80° C. for 16 hours. LCMS showed there was no starting material. The residue was added H2O (10 mL) and the solid formed was filtered under vacuum to give 2-(2,6-dioxo-3-piperidyl)-5-[4-[[1-[2-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]-4-piperidyl]methyl]piperazin-1-yl]isoindoline-1,3-dione (600 mg, crude) as a yellow solid. Step 5 To a mixture of 2-(2,6-dioxo-3-piperidyl)-5-[4-[[1-[2-[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]-4-piperidyl]methyl]piperazin-1-yl]isoindoline-1,3-dione (600 mg, 759.61 umol, 1 eq) and NH4Cl (203.16 mg, 3.80 mmol, 5 eq) in EtOH (10 mL), H2O (2 mL) and DCM (2 mL) was added Fe (212.10 mg, 3.80 mmol, 5 eq) in one portion at 20° C. under N2. The mixture was stirred at 80° C. for 1 hour. LCMS showed the reaction was completed. The residue was filtered with ethyl acetate (10 mL*5) and solution was concentrated in vacuum. The crude product was purified by reversed-phase HPLC (Column: 3_Phenomenex Luna C18 75*30 mm*3 um; Condition: water (0.05% HCl)-ACN; Begin B: 10.End B: 80; FlowRate: 25 mL/min; Gradient Time: 35 min; 100% B Hold Time: 1 minute) to give 5-[4-[[1-[2-[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]-4-piperidyl]methyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (180 mg, 232.14 umol, 30.56% yield, 98% purity) as a yellow solid. Step 6 To a mixture of 5-[4-[[1-[2-[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]ethyl]-4-piperidyl]methyl]piperazin-1-yl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (90 mg, 118.44 umol, 1 eq) and 4,5,7,7-tetramethyltetrazolo[1,5-a]pyrimidine-6-carboxylic acid (43.11 mg, 177.66 umol, 92% purity, 1.5 eq) in DMF (5 mL) was added DIEA (45.92 mg, 355.31 umol, 61.89 uL, 3 eq) in one portion and HATU (45.03 mg, 118.44 umol, 1 eq) at 25° C. under N2. The mixture was stirred at 25° C. for 16 hours to give yellow solution. LCMS showed desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water (0.1M FANH4)-ACN]; B %: 0%-30%, 40 min) to afford N-[3-[2-[4-[2-[4-[[4-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperazin-1-yl]methyl]-1-piperidyl]ethyl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7,7-tetramethyl-tetrazolo[1,5-a]pyrimidine-6-carboxamide (18.9 mg, 19.17 umol, 16.18% yield, 97.88% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 28: (7S)—N-(3-{2-[(3S)-4-{2-[4-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperazin-1-yl]ethyl}-3-methylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 28 was prepared in a manner analogous to Exemplary Compound 27. Exemplary Synthesis of Exemplary Compound 29: N-{3-[2-(4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7,7-tetramethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Step 1 To a solution of 5-nitro-3-(2-piperazin-1-yl-4-pyridyl)-1H-indazole (500 mg, 1.54 mmol, 1 eq) and tert-butyl 4-formylpiperidine-1-carboxylate (450 mg, 2.11 mmol, 1.37 eq) in HOAc (1 mL) and MeOH (10 mL) was added borane; 2-methylpyridine (329.78 mg, 3.08 mmol, 2 eq). After addition, the reaction solution was stirred at 20° C. for 2 hours. LCMS showed desired MS. The reaction was diluted with brine (100 mL). The solid was collected by filtration and dried under reduced pressure to afford tert-butyl 4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]piperidine-1-carboxylate (730 mg, 1.20 mmol, 78.07% yield, 86% purity) as a yellow solid. Step 2 To a solution of tert-butyl 4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]piperidine-1-carboxylate (730 mg, 1.40 mmol, 1 eq) in DCM (10 mL) was added TFA (7.70 g, 67.53 mmol, 5 mL, 48.25 eq). After addition, the reaction solution was stirred at 20° C. for 3 hours. LCMS showed starting material consumed and desired MS was detected. The reaction was concentrated under reduced pressure to afford 5-nitro-3-[2-[4-(4-piperidylmethyl)piperazin-1-yl]-4-pyridyl]-1H-indazole (589 mg, crude) as a yellow gum. The crude product was used for next step directly. Step 3 To a solution of 5-nitro-3-[2-[4-(4-piperidylmethyl)piperazin-1-yl]-4-pyridyl]-1H-indazole (589 mg, 1.40 mmol, 1 eq) and 1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]piperidine-4-carbaldehyde (619.39 mg, 1.68 mmol, 1.2 eq) in HOAc (5 mL) and MeOH (50 mL) was added borane; 2-methylpyridine (298.94 mg, 2.79 mmol, 2 eq). After addition, the reaction mixture was stirred at 20° C. for 12 hours. LCMS showed starting material consumed and desired MS detected. The reaction mixture was dilute with water (50 mL) and extracted with 10% methanol in dichloromethane (3×50 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0 to 40% methanol in dichloromethane) to afford 2-(2,6-dioxo-3-piperidyl)-5-[4-[[4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]isoindoline-1,3-dione (1 g, 1.25 mmol, 89.35% yield, 96.75% purity) as a yellow solid. Step 4 To a solution of 2-(2,6-dioxo-3-piperidyl)-5-[4-[[4-[[4-[4-(5-nitro-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]isoindoline-1,3-dione (500 mg, 645.27 umol, 1 eq) in EtOH (5 mL) and H2O (2 mL) was added Le (180.18 mg, 3.23 mmol, 5 eq) and NH4Cl (172.58 mg, 3.23 mmol, 5 eq). After addition, the reaction mixture was stirred at 80° C. for 1 hour. LCMS showed desired MS. After cooling, the reaction mixture was filtered and filtrate was concentrated under reduced pressure. The residue was triturated with 10% methanol in dichloromethane (20 mL) and filtered. The filtrate was concentrated under reduced pressure to afford 5-[4-[[4-[[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (110 mg, 143.24 umol, 22.20% yield, 97% purity) as a yellow solid. Step 5 To a mixture of 5-[4-[[4-[[4-[4-(5-amino-1H-indazol-3-yl)-2-pyridyl]piperazin-1-yl]methyl]-1-piperidyl]methyl]-1-piperidyl]-2-(2,6-dioxo-3-piperidyl)isoindoline-1,3-dione (100 mg, 134.25 umol, 1 eq) and 4,5,7,7-tetramethyltetrazolo[1,5-a]pyrimidine-6-carboxylic acid (56.19 mg, 201.37 umol, 80% purity, 1.5 eq) in DMF (5 mL) was added DIEA (52.05 mg, 402.75 umol, 70.15 uL, 3 eq) in one portion and HATU (51.05 mg, 134.25 umol, 1 eq) at 25° C. under N2. The mixture was stirred at 25° C. for 16 hours to give yellow solution. LCMS showed desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water (0.1M FANH4)-ACN]; B %: 0%-30%, 40 min) to afford N-[3-[2-[4-[[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]-4-piperidyl]methyl]piperazin-1-yl]-4-pyridyl]-1H-indazol-5-yl]-4,5,7,7-tetramethyl-tetrazolo[1,5-a]pyrimidine-6-carboxamide (12 mg, 12.25 umol, 9.13% yield, 97% purity) as a yellow solid. Exemplary Synthesis of Exemplary Compound 30: N-(3-{2-[(3S)-4-{2-[4-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperazin-1-yl]ethyl}-3-methylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7,7-tetramethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 30 was prepared in a manner analogous to Exemplary Compound 29. Exemplary Synthesis of Exemplary Compound 31: (7S)—N-(3-{2-[(3S)-4-{2-[4-({4-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperazin-1-yl}methyl)piperidin-1-yl]ethyl}-3-methylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7-trimethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 30 was prepared in a manner analogous to Exemplary Compound 29. Exemplary Synthesis of Exemplary Compound 32: N-(3-{2-[(2S)-2-{[4-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperazin-1-yl]methyl}morpholin-4-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7,7-tetramethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 30 was prepared in a manner analogous to Exemplary Compound 29. Step 1 To a mixture of tert-butyl (2R)-2-(hydroxymethyl)morpholine-4-carboxylate (2 g, 9.21 mmol, 1 eq) in DCM (20 mL) was added TFA (4.62 g, 40.52 mmol, 3 mL, 4.40 eq) in one portion at 20° C. under N2. The mixture was stirred at 20° C. for 30 minutes. TLC showed the reaction was completed. The solution was concentrated in vacuum to give [(2R)-morpholin-2-yl]methanol (2 g, 8.65 mmol, 93.98% yield, TFA) as a colourless oil. Step 2 To a mixture of 4-bromo-2-fluoro-pyridine (1.52 g, 8.65 mmol, 1 eq) and [(2R)-morpholin-2-yl]methanol (2 g, 8.65 mmol, 1 eq, TFA) in DMSO (10 mL) was added K2CO3(11.96 g, 86.52 mmol, 70.14 mL, 10 eq) in one portion at 100° C. under N2. The mixture was stirred at 100° C. for 2 hours to give yellow solution. TLC (Petroleum ether: Ethyl acetate=1:1, Rf=0.56) showed the reaction was completed. The mixture was cooled to 20° C. and concentrated in reduced pressure at 20° C. The residue was poured into water (50 mL). The aqueous phase was extracted with ethyl acetate (50 mL*4). The combined organic phase was washed with brine (50 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (Petroleum ether: Ethyl acetate=1:1, Rf=0.56, 20 g, 0-50% (10 min) of Ethyl acetate in Petroleum ether, 50% (10 min) of Ethyl acetate in Petroleum ether) to give [(2R)-4-(4-bromo-2-pyridyl)morpholin-2-yl]methanol (1.5 g, 5.49 mmol, 63.48% yield) as a yellow oil. Step 3 To a mixture of [(2R)-4-(4-bromo-2-pyridyl)morpholin-2-yl]methanol (1.5 g, 5.49 mmol, 1 eq) in DCM (15 mL) was added TEA (1.11 g, 10.98 mmol, 1.53 mL, 2 eq) and 4-methylbenzenesulfonyl chloride (1.57 g, 8.24 mmol, 1.5 eq) in one portion at 0° C. under N2. The mixture was stirred at 20° C. for 16 hours. LCMS showed the reaction was completed. The residue was poured into water (10 mL). The aqueous phase was extracted with DCM (20 mL*2). The combined organic phase was washed with brine (20 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (12 g, 0-100% (20 min) of Ethyl acetate in Petroleum ether) to give [(2R)-4-(4-bromo-2-pyridyl)morpholin-2-yl]methyl 4-methylbenzenesulfonate (2.1 g, 4.91 mmol, 89.52% yield) as a colourless oil Step 4 To a mixture of [(2R)-4-(4-bromo-2-pyridyl)morpholin-2-yl]methyl 4-methylbenzenesulfonate (2.1 g, 4.91 mmol, 1 eq) and tert-butyl piperazine-1-carboxylate (1.83 g, 9.83 mmol, 2 eq) in MeCN (20 mL) was added KI (1.63 g, 9.83 mmol, 2 eq) and DIPEA (1.27 g, 9.83 mmol, 1.71 mL, 2 eq) in one portion at 20° C. under N2. The mixture was stirred at 100° C. for 2 hours. TLC showed the reaction was completed. The mixture was cooled to 20° C. and concentrated in reduced pressure at 20° C. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 mL*3). The combined organic phase was washed with brine (20 mL*2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (12 g, 0-20% (5 min) of Ethyl acetate in Petroleum ether, 20% (15 minutes) of Ethyl acetate in Petroleum ether) to give tert-butyl 4-[[(2S)-4-(4-bromo-2-pyridyl)morpholin-2-yl]methyl]piperazine-1-carboxylate (1.1 g, 2.22 mmol, 45.14% yield, 89% purity) as a yellow gum. Step 5 To a mixture of tert-butyl 4-[[(2S)-4-(4-bromo-2-pyridyl)morpholin-2-yl]methyl]piperazine-1-carboxylate (1.1 g, 2.22 mmol, 89% purity, 1 eq), Pin2B2(1.13 g, 4.44 mmol, 2 eq) and KOAc (653.06 mg, 6.65 mmol, 3 eq) in dioxane (300 mL) was added Pd(dppf)Cl2(81.15 mg, 110.91 umol, 0.05 eq) in one portion at 25° C. under N2. The mixture was stirred at 100° C. for 1 hour. TLC showed the reaction was completed. The mixture was cooled to 25° C., filtered and concentrated in vacuum to give [2-[(2S)-2-[(4-tert-butoxycarbonylpiperazin-1-yl)methyl]morpholin-4-yl]-4-pyridyl]boronic acid (1 g, 1.75 mmol, 78.78% yield, 71% purity) as a black brown solid. Exemplary Synthesis of Exemplary Compound 33: N-{3-[2-(4-{2-[4-({4-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperazin-1-yl}methyl)piperidin-1-yl]ethyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7,7-tetramethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 33 was prepared in a manner analogous to Exemplary Compound 27. Exemplary Synthesis of Exemplary Compound 34: N-(3-{2-[(3S)-4-{[1-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperidin-4-yl]methyl}-3-methylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7,7-tetramethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 34 was prepared in a manner analogous to Exemplary Compound 1. Exemplary Synthesis of Exemplary Compound 35: N-{3-[2-(4-{2-[4-({1-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperidin-4-yl}methyl)piperazin-1-yl]ethyl}piperazin-1-yl)pyridin-4-yl]-1H-indazol-5-yl}-4,5,7,7-tetramethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 35 was prepared in a manner analogous to Exemplary Compound 27. Exemplary Synthesis of Exemplary Compound 36: N-(3-{2-[(3S)-4-{2-[4-({4-[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl]piperazin-1-yl}methyl)piperidin-1-yl]ethyl}-3-methylpiperazin-1-yl]pyridin-4-yl}-1H-indazol-5-yl)-4,5,7,7-tetramethyl-4H,7H-[1,2,3,4]tetrazolo[1,5-a]pyrimidine-6-carboxamide Exemplary Compound 35 was prepared in a manner analogous to Exemplary Compound 25. Protein Level Control This description also provides methods for the control of protein levels within a cell. The method is based on the use of compounds as described herein such that degradation of the target protein LRRK2 in vivo will result in the reducing the amount of the target protein in a biological system, preferably to provide a particular therapeutic benefit. The following examples are used to assist in describing the present disclosure, but should not be seen as limiting the present disclosure in any way. In certain embodiments, the description provides the following exemplary LRRK2-degrading bifunctional molecules (compounds of Table 1 or Compounds 1-36), including salts, polymorphs, analogs, derivatives, and deuterated forms thereof. Assay for Testing LRRK2 Degradation Driven by Compounds as Described Herein, The assay measures the degradation of wildtype and G2019S LRRK2 tagged with a HiBit tag on the C-terminus of the protein that was expressed from a mammalian expression vector, driven by the ubiquitin promoter in HEK293 cells. Each compound dose-response was repeated on three separate days, on three separate plates each day. Plasmid Preparation. Transfection mixes were assembled as follows and incubated for 30 minutes at room temperature. In a 15 mL tube, 5.25 mL Opti-MEM (no additions) was mixed with 17 μL Firefly Luciferase plasmid at 1 μg/μL and 158 μL WT plasmid DNA at 1 μg/μL (175 μg total DNA) were mixed by flicking. In a new 15 mL tube, 5.25 mL OptiMEM was mixed with 17 μL Firefly Luciferase plasmid at 1 μg/μL and 158 μL G2019S plasmid DNA at 1 μg/μL (175 μg total DNA) were mixed by flicking. X-tremeGene HP was mixed thoroughly using a vortex. Next, 175 μL was added to each tube and flicked to mix. Both tubes were left to incubate for 30 minutes at room temperature. While the transfection mixes were incubating, HEK293 cells acquired from ATCC (ATCC CRL-1573) were harvested with trypsin. Once cells are detached, the cells were resuspended in 12 mL OptiMEM+5% FBS and transferred to a 50 mL tube. The cells were mixed well and counted. Using OptiMEM+5% FBS, the cells were diluted in two 250 mL conical tubes at 0.71×106cells/mL in 70 mL. One tube was labeled “WT” and the other “G2019S”. The WT and G2019S transfection mixes were added dropwise to the corresponding 250 mL tubes. The tubes were mixed first by pipetting then by swirling. The tubes were incubated at room temperature for at least 5 minutes. Each tube was swirled before dispensing and after every three plates. Seventy microliters of cells were dispensed with WT or G2019S DNA to seven plates each. Three plates of each were tested with compound plate one (preparation described below) and three plates of each were tested with compound plate two (preparation described below). The first plate from each set served as a “prime” plate and was not used to test compounds. Each plate was incubated in the hood for 10 minutes before placing in the 37° C. incubator for 24 hours. Preparation of Compound and Assay Plates. Two compound plates were made using 96 well polypropylene plates. Compounds were made up at 10 mM and were diluted to 1 mM in 30 μL. Each dose response curve included a well of DMSO, as a negative control and for normalization, and a well of 0.5 μM of Exemplary Compound 4 as a positive control. In addition to seven test compounds, each plate also included a dose response of Exemplary Compound 4. The compound plates were spun down along at 1200 rpm for 2 minutes. The two compound plates were then mixed and 2 μL was diluted in intermediate plates having 248 μL of Opti-Mem in each well. Next, 10 μL diluted compounds from the intermediate plates were added to each test plate (three WT and three G2019S plates per compound plate for a total of 12 assay plates). The plates were incubated for 24 hours at 37° C. All assay plates and all Nano-Glo Dual-Luciferase Reporter Assay System components (except for the DLR substrate) were equilibrated to room temperature. Next, the luciferase buffer was mixed with the lyophilized amber bottle until fully dissolved, and 75 μL of the luciferase mixture was added to each well of each assay plate. The assay plates were incubated for 10 minutes at room temperature with shaking for at least 5 minutes, and then read on a plate reader. Developing Plates and Analyzing Data. One milliliter of DLR substrate and 1 mL LgBiT Protein were added to the Stop and Glo buffer, and 75 μL of the mixture was added to each well of each plate. Optically clear seals were added to each plate and each plate was incubated for 20 minutes with shaking for at least 10 minutes, and then read on a plate reader. As mentioned above, plates were fun in triplicate and assay repeated twice (total of 6 replicates per exemplary compound. Each cell was examined for firefly luciferase for cell number and viability and Nanoluc for the LRRK2-HiBit quantification. Ratio of (HiBit/luciferase)*1000 was determined and the data was_normalized to % of DMSO median value. Curve fitting was performed on each individual plate. The data for some of exemplary compounds of Table 1 below is shown below in Table 2 in the columns labeled *G2019S DC50 (nM) and **G2019S Dmax (%). Exemplary Assay for Testing LRRK2 Degradation Driven by Exemplary Hetero-Bifunctional Compounds Designed to Target LRRK2 The assay measures the degradation of wildtype and G2019S LRRK2 tagged with a HiBit tag on the C-terminus of the protein that was expressed from a mammalian expression vector, driven by the ubiquitin promoter in HEK293 cells. Each compound dose-response was repeated on two separate days, on three separate plates each day. Plasmid Preparation. Transfection mixes were assembled as follows and incubated for 30 minutes at room temperature. In a 15 mL tube, 5.25 mL Opti-MEM (no additions) was mixed with 17 μL Firefly Luciferase plasmid at 1 μg/μL and 158 μL WT plasmid DNA at 1 μg/μL (175 μg total DNA) were mixed by flicking. In a new 15 mL tube, 5.25 mL OptiMEM was mixed with 17 μL Firefly Luciferase plasmid at 1 μg/μL and 158 μL G2019S plasmid DNA at 1 μg/μL (175 μg total DNA) were mixed by flicking. X-tremeGene HP was mixed thoroughly using a vortex. Next, 175 μL was added to each tube and flicked to mix. Both tubes were left to incubate for 30 minutes at room temperature. While the transfection mixes were incubating, HEK293 cells (acquired from ATCC; ATCC CRL-1573) were harvested with trypsin. Once cells are detached, the cells were resuspended in 12 mL OptiMEM+5% FBS and transferred to a 50 mL tube. The cells were mixed well and counted. Using OptiMEM+5% FBS, the cells were diluted in two 250 mL conical tubes at 0.71×106cells/mL in 70 mL. One tube was labeled “WT” and the other “G2019S”. The WT and G2019S transfection mixes were added dropwise to the corresponding 250 mL tubes. The tubes were mixed first by pipetting then by swirling. The tubes were incubated at room temperature for at least 5 minutes. Each tube was swirled before dispensing and after every three plates. Seventy microliters of cells were dispensed with WT or G2019S DNA to seven plates each. Three plates of each were tested with compound plate one (preparation described below) and three plates of each were tested with compound plate two (preparation described below). The first plate from each set served as a “prime” plate and was not used to test compounds. Each plate was incubated in the hood for 10 minutes before placing in the 37° C. incubator for 24 hours. Preparation of Compound and Assay Plates. Two compound plates were made using 96 well polypropylene plates. Compounds were made up at 10 mM and were diluted to 1 mM in 30 μL. Each dose response curve included a well of DMSO, as a negative control and for normalization, and a well of 0.5 μM of Exemplary Compound 4 as a positive control. In addition to seven test compounds, each plate also included a dose response of Exemplary Compound 4. The compound plates were spun down along at 1200 rpm for 2 minutes. The two compound plates were then mixed and 2 μL was diluted in intermediate plates having 248 μL of Opti-Mem in each well. Next, 10 μL diluted compounds from the intermediate plates were added to each test plate (three WT and three G2019S plates per compound plate for a total of 12 assay plates). The plates were incubated for 24 hours at 37° C. All assay plates and all Nano-Glo Dual-Luciferase Reporter Assay System components (except for the DLR substrate) were equilibrated to room temperature. Next, the luciferase buffer was mixed with the lyophilized amber bottle until fully dissolved, and 75 μL of the luciferase mixture was added to each well of each assay plate. The assay plates were incubated for 10 minutes at room temperature with shaking for at least 5 minutes, and then read on a plate reader. Developing Plates and Analyzing Data. One milliliter of DLR substrate and 1 mL LgBiT Protein were added to the Stop and Glo buffer, and 75 μL of the mixture was added to each well of each plate. Optically clear seals were added to each plate and each plate was incubated for 20 minutes with shaking for at least 10 minutes, and then read on a plate reader. As mentioned above, plates were run in triplicate and assay repeated twice (total of 6 replicates per exemplary compound. Each cell was examined for firefly luciferase for cell number and viability and Nanoluc for the LRRK2-HiBit quantification. Ratio of (HiBit/luciferase)*1000 was determined and the data was_normalized to % of DMSO median value. Curve fitting was performed on each individual plate. The data for exemplary compounds of Table 1 below is shown below in Table 2 in the G2019S DC50, G2019S Dmax, WT DC50 and WT Dmax columns. Exemplary Assay for Testing LRRK2 Degradation Driven by Exemplary Hetero-Bifunctional Compounds Designed to Target LRRK2 The assay measures the degradation of LRRK2 in cells where the C-terminus (3′) of the endogenous gene has been tagged with a HiBit sequence in HEK293 cells. The cells also express firefly luciferase, expressed from a Cytomegalovirus promoter and introduced into the HiBit tagged cells and stably expressed. The Nano-Glo® Dual Luciferase Reporter Assay System (Promega™, Madison, Wis.) was utilized. Day 1—Preparation of Compound and Assay Plates. Two sets of plates were prepared: a triplicate set for the HiBit assay in white 384-well plates and a triplicate set of plate in black 384-well plates for the Alamar Blue cell viability assay. Briefly, the growth media (DMEM+Glutamax-10% fetal bovine serum-1% Penicillin-Streptomicin) from two T128 flasks was aspirated from the flasks. Cells were washed with Dulbecco's Phosphate Buffered Saline (dPBS) and aspirated. Trypsin (3 mL per flask) was added and the flasks were incubated for 2-3 minutes. Ten mL of OptiMEM-10% fetal bovine-1% penicillin-streptomycin (hereinafter, “OptiMEM media”) was added to the flask and the cells and transferred to a 50 mL conical tube. A cell count (25 ul of cell into Effendorf vial +25 ul of Trypan Blue Stain) was performed and the cell density adjusted to 15,000 cell/45 μl/well (3.33×10{circumflex over ( )}5/mL) in OptiMEM media. Forty-five microliters of the cell suspension (15,000 cells) was aliquoted to each well of the white 384-well plate. The plates incubated at room temperature for 10 minutes before being placed in the 37° C.+5% CO2incubator overnight Day 2—Compound Treatment. Exemplary compounds were prepared at a 1 mM starting concentration and 1:3 serial dilution for 11 points CRC prepared and stored in the freezer. The Master Compound Plate was thawed overnight at room temperature. DMSO (20 μL) was added into column 24 of the Master Compound Plate for negative control and 20 μL of 300 μM of Exemplary Compound 4 in column 23 as positive control. Intermediate Compound Plate with 4% DMSO in OptiMEM Media. DMSO was added to warm OptiMEM media to achieve a 4% DMSO solution (approximately 50 mL/plate). One-hundred microliters of the OptiMEM-4% DMSO was aliquoted to each well of 384-Well Deep Well Microplates. The Master Compound Plate and the Intermediate Compound Plate were spun down. One microliter of compound from the Master Compound Plate was transferred into the Intermediate plate (a 1:100 dilution). The diluted mixture was mixed and 5 μL transferred into the assay plate (a 1:10 dilution) for the final starting concentration of 1 μM. The Treated Assay plates were incubated for 24 hours at 37° C.+5% CO2. The Master Compound Plate was sealed and store at room temperature for a second run that was performed within a week. Day 3—HiBit Assay. Five microliters of Alamar Blue was added to each well of the black 384-well plates. The plates were incubated for 2 hours in the incubator (37° C.+5% CO2) and at room temperature for one hour. Fluorescence of each plate was read on a plated reader for the Alamar Blue viability assay. One set of white assay plates was warmed to room temperature (45 minute). The One Glo luciferase mixture was prepared. The media from white 384-well assay plates was aspirated. Twenty-five μL of the One Glo luciferase mixture was added to each well of the assay plates. The plates were incubated on the bench (room temperature) for 45 minutes, including 10 minutes of shaking at 700 rpm. The luminescence of each plate was read on a plate reader. 1:100 DLR substrate and 1:100 LgBiT Protein dilution were added to the Promega Stop and Glo buffer and mixed just before addition to assay plates. Twenty-five microliters of Stop and Glo mixture was added to each well. Assay plates incubated for at least 45 minutes, including 10 minutes of shaking at 700 rpm. The luminescence of each plate was read on a plate reader. Analysis of LRRK2 HiBit Screening assays. As mentioned above, plates were run in triplicate and the assay repeated twice (total of 6 replicate for exemplary compound). For each treatment, measurements were taken for firefly luciferase for cell number, cell viability (Alamar Blue), and Nanoluc for the LRRK2-HiBit quantification. The LRRK2 HiBit and alamar blue signal was normalized to % DMSO median value for each plate. Curve fitting was performed on each compound for replicates across three plates. The date for exemplary compounds of Table 1 below is shown below in Table 2 in the columns labeled *WT DC50 (nM) and **WT Dmax (%). TABLE 1Exemplary bifunctional compounds of the present disclosure.Ex.StructureMassName1949.48N-(3-(2-((3S,5R)-4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4-yl)methyl)- 3,5-dimethylpiperazin-1-yl)pyridin- 4-yl)-1H-indazol-5-yl)-4,5- dimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide2963.50N-(3-(2-((3S,5R)-4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4-yl)methyl)- 3,5-dimethylpiperazin-1-yl)pyridin- 4-yl)-1H-indazol-5-yl)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide3911.43N-(3-(2-((3R,5S)-4-(2-(2-(4-(2- (2,6-dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperazin-1- yl)ethoxy)ethyl)-3,5- dimethylpiperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5-dimethyl- 4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide4955.46N-(3-(2-((3R,5S)-4-(2-(2-(2-(4-(2- (2,6-dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperazin-1- yl)ethoxy)ethoxy)ethyl)-3,5- dimethylpiperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5-dimethyl- 4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide5867.40N-(3-(2-((3R,5S)-4-(2-(4-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperazin-1- yl)ethyl)-3,5-dimethylpiperazin-1- yl)pyridin-4-yl)-1H-indazol-5-yl)- 4,5-dimethyl-4,7- dihydrotetrazolo[1,5-a]pyrimidine- 6-carboxamide6977.51N-(3-(2-((3S,5R)-4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4-yl)methyl)- 3,5-dimethylpiperazin-1-yl)pyridin- 4-yl)-1H-indazol-5-yl)-4,5,7,7- tetramethyl-4,7- dihydrotetrazolo[1,5-a]pyrimidine- 6-carboxamide7963.50(S)-N-(3-(2-((3S,5R)-4-((1-((1-(2- (2,6-dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4-yl)methyl)- 3,5-dimethylpiperazin-1-yl)pyridin- 4-yl)-1H-indazol-5-yl)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide8949.48(R)-N-(3-(2-((S)-4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4-yl)methyl)-3- methylpiperazin-1-yl)pyridin-4-yl)- 1H-indazol-5-yl)-4,5,7-trimethyl- 4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide9949.48(S)-N-(3-(2-((S)-4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4-yl)methyl)-3- methylpiperazin-1-yl)pyridin-4-yl)- 1H-indazol-5-yl)-4,5,7-trimethyl- 4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide10935.47N-(3-(2-((S)-4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4-yl)methyl)-3- methylpiperazin-1-yl)pyridin-4-yl)- 1H-indazol-5-yl)-4,5-dimethyl-4,7- dihydrotetrazolo[1,5-a]pyrimidine- 6-carboxamide11935.47(7R)-N-(3-(2-(4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4- yl)methyl)piperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide12935.47(7S)-N-(3-(2-(4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4- yl)methyl)piperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide13921.45N-(3-(2-(4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4- yl)methyl)piperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5-dimethyl- 4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide14949.48(7R)-N-(3-(2-(4-(2-(1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4- yl)ethyl)piperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide15867.40(7R)-N-(3-(2-(4-(3-(4-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperazin-1- yl)propyl)piperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide16893.42(7R)-N-(3-(2-(4-((1-(1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)azetidin-3-yl)methyl)piperazin-1- yl)pyridin-4-yl)-1H-indazol-5-yl)- 4,5,7-trimethyl-4,7- dihydrotetrazolo[1,5-a]pyrimidine- 6-carboxamide17907.44(7R)-N-(3-(2-(4-((1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)azetidin-3- yl)methyl)piperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide18921.45(7R)-N-(3-(2-(4-((1-(1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)piperidin-4-yl)methyl)piperazin- 1-yl)pyridin-4-yl)-1H-indazol-5-yl)- 4,5,7-trimethyl-4,7- dihydrotetrazolo[1,5-a]pyrimidine- 6-carboxamide19850.3820909.41(7R)-N-[3-(2-{4-[2-(2-{6-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]-2,6- diazaspiro[3.3]heptan-2- yl}ethoxy)ethyl]piperazin-1- yl}pyridin-4-yl)-1H-indazol-5-yl]- 4,5,7-trimethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide21950.48(7R)-N-(3-(2-(4-(2-(4-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperazin-1- yl)ethyl)piperazin-1-yl)pyridin-4- yl)-1H-indazol-5-y1)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide22985.46(7R)-N-(3-(2-(4-(2-(1-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-4-yl)-2,2- difluoroethyl)piperazin-1- yl)pyridin-4-yl)-1H-indazol-5-yl)- 4,5,7-trimethyl-4,7- dihydrotetrazolo[1,5-a]pyrimidine- 6-carboxamide23949.48(7R)-N-(3-(2-(4-(2-(4-((1-(2-(2,6- dioxopiperidin-3-yl)-1,3- dioxoisoindolin-5-yl)piperidin-4- yl)methyl)piperidin-1- yl)ethyl)piperazin-1-yl)pyridin-4- yl)-1H-indazol-5-yl)-4,5,7- trimethyl-4,7-dihydrotetrazolo[1,5- a]pyrimidine-6-carboxamide24893.42(7R)-N-[3-(2-{4-[1-({1-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperidin- 4-yl} methyl)azetidin-3- yl]piperazin-1-yl}pyridin-4-yl)-1H- indazol-5-yl]-4,5,7-trimethyl- 4H,7H-[1,2,3,4]tetrazolo[1,5- a]pyrimidine-6-carboxamide25950.48(7R)-N-{3-[2-(4-{2-[4-({1-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperidin- 4-yl}methyl)piperazin-1- yl]ethyl}piperazin-1-yl)pyridin-4- yl]-1H-indazol-5-yl}-4,5,7- trimethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide26850.38(7R)-N-(3-{2-[6-({1-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperidin- 4-yl}methyl)-2,6- diazaspiro[3.3]heptan-2-yl]pyridin- 4-yl}-1H-indazol-5-yl)-4,5,7- trimethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide27950.48(7S)-N-{3-[2-(4-{2-[4-({4-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperazin- 1-yl}methyl)piperidin-1- yl]ethyl}piperazin-1-yl)pyridin-4- yl]-1H-indazol-5-yl}-4,5,7- trimethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide28964.49(7S)-N-(3-{2-[(3S)-4-{2-[4-({1-[2- (2,6-dioxopiperidin-3-yl)-1,3- dioxo-2,3-dihydro-1H-isoindol-5- yl]piperidin-4-yl}methyl)piperazin- 1-yl]ethyl}-3-methylpiperazin-1- yl]pyridin-4-yl}-1H-indazol-5-yl)- 4,5,7-trimethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide29949.48N-{3-[2-(4-{[1-({1-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperidin- 4-yl}methyl)piperidin-4- yl]methyl}piperazin-1-yl)pyridin-4- yl]-1H-indazol-5-yl}-4,5,7,7- tetramethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide30978.51N-(3-{2-[(3S)-4-{2-[4-({1-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperidin- 4-yl}methyl)piperazin-1-yl]ethyl}- 3-methylpiperazin-1-yl]pyridin-4- yl}-1H-indazol-5-yl)-4,5,7,7- tetramethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide31964.49(7S)-N-(3-{2-[(3S)-4-{2-[4-({4-[2- (2,6-dioxopiperidin-3-yl)-1,3- dioxo-2,3-dihydro-1H-isoindol-5- yl]piperazin-1-yl}methyl)piperidin- 1-yl]ethyl}-3-methylpiperazin-1- yl]pyridin-4-yl}-1H-indazol-5-yl)- 4,5,7-trimethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide32951.46N-(3-{2-[(2S)-2-{[4-({1-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperidin- 4-yl}methyl)piperazin-1- yl]methyl}morpholin-4-yl]pyridin- 4-yl}-1H-indazol-5-yl)-4,5,7,7- tetramethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide33964.49N-{3-[2-(4-{2-[4-({4-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperazin- 1-yl}methyl)piperidin-1- yl]ethyl}piperazin-1-yl)pyridin-4- yl]-1H-indazol-5-yl}-4,5,7,7- tetramethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide34963.50N-(3-{2-[(3S)-4-{[1-({1-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperidin- 4-yl}methyl)piperidin-4- yl]methyl}-3-methylpiperazin-1- yl]pyridin-4-yl}-1H-indazol-5-yl)- 4,5,7,7-tetramethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide35964.49N-{3-[2-(4-{2-[4-({1-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperidin- 4-yl}methyl)piperazin-1- yl]ethyl}piperazin-1-yl)pyridin-4- yl]-1H-indazol-5-yl}-4,5,7,7- tetramethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a ]pyrimidine- 6-carboxamide36978.51N-(3-{2-[(3S)-4-{2-[4-({4-[2-(2,6- dioxopiperidin-3-yl)-1,3-dioxo-2,3- dihydro-1H-isoindol-5-yl]piperazin- 1-yl}methyl)piperidin-1-yl]ethyl}- 3-methylpiperazin-1-yl]pyridin-4- yl}-1H-indazol-5-yl)-4,5,7,7- tetramethyl-4H,7H- [1,2,3,4]tetrazolo[1,5-a]pyrimidine- 6-carboxamide TABLE 2Characterization of exemplary bifunctional compounds of the present disclosure.Mean*G2019S**G2019*WT** WTObservedDC50S DmaxDC50DmaxEx.NMRMass(nM)(%)(nM)(%)11H NMR: (400 MHz, DMSO-d6) δ: 13.42 (s, 1H), 11.08 (s, 1H), 10.07 (s, 1H), 8.67950.60ABBB(s, 1H), 8.22 (d, J = 5.1 Hz, 1H), 7.67-7.51 (m, 3H), 7.28 (d, J = 13.8 Hz, 2H), 7.23(d, J = 8.8 Hz, 1H), 7.16 (d, J = 5.3 Hz, 1H), 5.30 (s, 2H), 5.11-5.02 (m, 1H), 4.12(d, J = 11.1 Hz, 2H), 4.03 (d, J = 12.9 Hz, 2H), 3.44 (s, 3H), 3.00-2.80 (m, 5H), 2.72-2.53 (m, 6H), 2.39-2.30 (m, 2H), 2.27 (s, 3H), 2.16 (d, J = 5.8 Hz, 2H), 2.05-1.97(m, 1H), 1.95-1.71 (m, 7H), 1.37 (s, 1H), 1.12 (d, J = 5.8 Hz, 10H).21H NMR: (400 MHz, DMSO) δ: 13.42 (s, 1H), 11.08 (s, 1H), 10.28 (s, 1H), 8.67 (s,964.60ABBB1H), 8.23 (d, J = 5.3 Hz, 1H), 8.15 (s, 1H), 7.67-7.58 (m, 2H), 7.56-7.50 (m, 1H),7.28 (d, J = 13.6 Hz, 2H), 7.23 (d, J = 8.8 Hz, 1H), 7.16 (d, J = 5.4 Hz, 1H), 5.83-5.71 (m, 1H), 5.06 (dd, J = 5.4, 12.9 Hz, 1H), 4.12 (d, J = 12.0 Hz, 2H), 4.03 (d,J = 12.9 Hz, 2H), 3.44 (s, 3H), 3.01-2.82 (m, 5H), 2.72-2.55 (m, 5H), 2.40-2.33(m, 2H), 2.20 (s, 3H), 2.17 (s, 2H), 2.06-1.97 (m, 1H), 1.94-1.71 (m, 7H), 1.56(d, J = 6.4 Hz, 3H), 1.38 (s, 1H), 1.11 (d, J = 5.8 Hz, 10H).31H NMR: (400 MHz, CD3OD) δ: 8.65 (s, 1H), 8.40 (s, 1H), 8.17 (d, J = 5.3 Hz, 1H),912.57D—D—7.52 (d, J = 8.9 Hz, 1H), 7.38-7.32 (m, 2H), 7.24 (d, J = 5.3 Hz, 1H), 7.19 (d, J = 8.5Hz, 1H), 6.93 (d, J = 2.1 Hz, 1H), 6.75 (dd, J = 2.1, 8.5 Hz, 1H), 5.30 (s, 2H), 5.02(br dd, J = 5.5, 12.8 Hz, 1H), 4.53 (br t, J = 11.4 Hz, 2H), 3.92-3.81 (m, 4H), 3.67(br t, J = 4.8 Hz, 2H), 3.55 (br s, 2H), 3.51 (s, 3H), 3.26-3.20 (m, 4H), 3.04-2.94(m, 2H), 2.92-2.81 (m, 1H), 2.76 (br d, J = 2.8 Hz, 1H), 2.68-2.56 (m, 7H), 2.36(s, 3H), 2.14-2.05 (m, 1H), 1.48 (br d, J = 5.4 Hz, 6H)41H NMR: (400 MHz, MeOD) δ: 8.71 (s, 1H), 8.27 (d, J = 5.4 Hz, 1H), 7.55-7.48956.59D—D—(m, 2H), 7.41 (d, J = 8.5 Hz, 1H), 7.38-7.31 (m, 2H), 6.99 (d, J = 1.8 Hz, 1H), 6.87(d, J = 8.5 Hz, 1H), 5.29 (s, 2H), 5.05 (dd, J = 5.4, 12.7 Hz, 1H), 4.55 (d, J = 13.9 Hz,2H), 3.85 (d, J = 4.5 Hz, 2H), 3.79-3.53 (m, 10H), 3.51 (s, 3H), 3.22 (s, 4H), 3.05-2.93 (m, 2H), 2.91-2.51 (m, 9H), 2.34 (s, 3H), 2.16-2.07 (m, 1H), 1.49 (d, J = 6.3Hz, 6H).51H NMR: (400 MHz, METHANOL-d4) δ: 8.69 (s, 1H), 8.41 (br s, 1H), 8.28 (d,868.54DCD—J = 5.3 Hz, 1H), 7.54-7.47 (m, 2H), 7.45 (s, 1H), 7.36 (d, J = 6.6 Hz, 2H), 7.15 (s,1H), 7.01 (br d, J = 6.9 Hz, 1H), 5.29 (s, 2H), 5.08 (dd, J = 5.5, 12.4 Hz, 1H), 4.59 (s,3H), 4.43 (br s, 2H), 3.51 (s, 4H), 3.27 (br s, 4H), 3.02 (br t, J = 12.6 Hz, 2H), 2.93-2.82 (m, 1H), 2.80-2.75 (m, 1H), 2.75-2.69 (m, 3H), 2.66 (br t, J = 4.7 Hz, 4H),2.35 (s, 3H), 2.19-2.05 (m, 1H), 1.44 (br d, J = 4.9 Hz, 6H)61H NMR (400 MHz, MeOD-d4) δ: 8.74 (s, 1H), 8.46 (s, 1H), 8.15 (d, J = 5.4 Hz,978.8BCDC1H), 7.68 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.9 Hz, 1H), 7.36 (d, J = 2.1 Hz, 1H), 7.31-7.20 (m, 4H), 5.07 (dd, J = 5.4, 12.3 Hz, 1H), 4.75 (br s, 1H), 4.71 (br s, 1H), 4.14-4.00 (m, 5H), 3.73 (s, 1H), 3.59 (br d, J = 11.0 Hz, 1H), 3.46 (s, 3H), 3.03 (br d,J = 14.3 Hz, 5H), 2.89-2.81 (m, 4H), 2.77 (br s, 2H), 2.74-2.67 (m, 2H), 2.63 (brd, J = 5.8 Hz, 2H), 2.20 (br d, J = 17.6 Hz, 2H), 2.14 (br s, 2H), 1.97-1.91 (m, 2H),1.88 (s, 3H), 1.57 (br d, J = 14.9 Hz, 2H), 1.50 (s, 3H), 1.45-1.37 (m, 2H), 1.33-1.25 (m, 2H), 1.19-1.14 (m, 5H)81H NMR (400 MHz, MeOD-d4)δ: 8.68 (s, 1H), 8.47 (br s, 1H), 8.22 (d, J = 5.6 Hz,950.4ABBB1H), 7.68 (d, J = 8.5 Hz, 1H), 7.61 (d, J = 9.0 Hz, 1H), 7.50 (d, J = 8.6 Hz, 1H), 7.38(br d, J = 5.9 Hz, 2H), 7.31-7.21 (m, 2H), 5.74 (br d, J = 5.8 Hz, 1H), 5.07 (dd,J = 5.4, 12.1 Hz, 1H), 4.12-4.07 (m, 2H), 4.01 (br d, J = 9.5 Hz, 2H), 3.50 (s, 5H),3.11-2.99 (m, 5H), 2.95 (br s, 2H), 2.89-2.80 (m, 2H), 2.79-2.69 (m, 3H), 2.62(br s, 1H), 2.43 (br s, 1H), 2.28 (s, 3H), 2.24-2.04 (m, 5H), 1.99-1.84 (m, 4H),1.69 (d, J = 6.4 Hz, 3H), 1.51-1.37 (m, 4H), 1.22-1.17 (m, 3H)91H NMR (400 MHz, MeOD-d4) δ: 8.68 (s, 1H), 8.42 (br s, 1H), 8.22 (d, J = 5.3 Hz,950.4CBDC1H), 7.68 (d, J = 8.5 Hz, 1H), 7.61 (d, J = 9.0 Hz, 1H), 7.49 (dd, J = 1.6, 8.9 Hz, 1H),7.41-7.34 (m, 2H), 7.30-7.21 (m, 2H), 5.74 (br d, J = 6.1 Hz, 1H), 5.07 (dd, J = 5.4,12.4 Hz, 1H), 4.09 (br d, J = 12.9 Hz, 2H), 4.01 (br d, J = 12.3 Hz, 2H), 3.56(br d, J = 9.0 Hz, 2H), 3.50 (s, 3H), 3.10-2.97 (m, 7H), 2.96-2.81 (m, 3H), 2.78-2.69 (m, 3H), 2.63 (br s, 1H), 2.44 (br t, J = 9.7 Hz, 1H), 2.28 (s, 3H), 2.25-2.15(m, 3H), 2.13-2.06 (m, 1H), 1.99 (br d, J = 14.4 Hz, 1H), 1.92 (br d, J = 12.9 Hz,3H), 1.68 (d, J = 6.4 Hz, 3H), 1.56-1.36 (m, 4H), 1.20 (d, J = 6.1 Hz, 3H)101H NMR (400 MHz, MeOD-d4) δ: 8.64 (s, 1H), 8.44 (s, 2H), 8.22 (d, J = 5.5 Hz,936.5ABBB1H), 7.68 (d, J = 8.5 Hz, 1H), 7.63-7.58 (m, 1H), 7.50 (d, J = 9.1 Hz, 1H), 7.40-7.34 (m, 2H), 7.31-7.22 (m, 2H), 5.29 (s, 2H), 5.07 (dd, J = 5.4, 12.6 Hz, 1H), 4.60(br s, 2H), 4.09 (br d, J = 11.6 Hz, 2H), 4.01 (br d, J = 11.9 Hz, 2H), 3.55 (br s, 2H),3.51 (s, 3H), 3.12-2.96 (m, 5H), 2.96-2.81 (m, 3H), 2.78-2.68 (m, 3H), 2.62 (brs, 1H), 2.43 (br t, J = 9.4 Hz, 1H), 2.34 (s, 3H), 2.20 (br s, 3H), 2.12-2.07 (m, 1H),2.02-1.89 (m, 4H), 1.57-1.35 (m, 4H), 1.20 (d, J = 6.1 Hz, 3H)111H NMR (400 MHz, MeOD-d4) δ: 8.64 (s, 1H), 8.23 (d, J = 5.1 Hz, 1H), 7.63 (dd,936.3BCCCJ = 8.6, 15.1 Hz, 2H), 7.55-7.49 (m, 1H), 7.38 (s, 1H), 7.30 (d, J = 5.1 Hz, 1H), 6.99(s, 1H), 6.83 (d, J = 7.4 Hz, 1H), 5.74 (d, J = 6.0 Hz, 1H), 5.06 (dd, J = 5.5, 12.8 Hz,1H), 3.72-3.56 (m, 9H), 3.49 (s, 3H), 3.45 (s, 1H), 3.22-3.11 (m, 4H), 2.97 (s,3H), 2.89-2.64 (m, 7H), 2.45-2.33 (m, 2H), 2.28 (s, 3H), 2.11 (d, J = 13.3 Hz,3H), 1.94 (d, J = 7.4 Hz, 3H), 1.87-1.79 (m, 1H), 1.68 (d, J = 6.3 Hz, 3H), 1.52 (d,J = 6.9 Hz, 1H)121H NMR (400 MHz, MeOD-d4) δ: 8.64 (s, 1H), 8.23 (d, J = 5.1 Hz, 1H), 7.63 (dd,936.4DCDJ = 8.6, 15.1 Hz, 2H), 7.55-7.49 (m, 1H), 7.38 (s, 1H), 7.30 (d, J = 5.1 Hz, 1H), 6.99(s, 1H), 6.83 (d, J = 7.4 Hz, 1H), 5.74 (d, J = 6.0 Hz, 1H), 5.06 (dd, J = 5.5, 12.8 Hz,1H), 3.72-3.56 (m, 9H), 3.49 (s, 3H), 3.45 (s, 1H), 3.22-3.11 (m, 4H), 2.97 (s,3H), 2.89-2.64 (m, 7H), 2.45-2.33 (m, 2H), 2.28 (s, 3H), 2.11 (d, J = 13.3 Hz,3H), 1.94 (d, J = 7.4 Hz, 3H), 1.87-1.79 (m, 1H), 1.68 (d, J = 6.3 Hz, 3H), 1.52 (d,J = 6.9 Hz, 1H)131H NMR (400 MHz, MeOD-d4) δ: 8.64 (s, 1H), 8.23 (d, J = 5.1 Hz, 1H), 7.63 (dd,922.4BBDCJ = 8.6, 15.1 Hz, 2H), 7.55-7.49 (m, 1H), 7.38 (s, 1H), 7.30 (d, J = 5.1 Hz, 1H), 6.99(s, 1H), 6.83 (d, J = 7.4 Hz, 1H), 5.74 (d, J = 6.0 Hz, 1H), 5.06 (dd, J = 5.5, 12.8 Hz,1H), 3.72-3.56 (m, 9H), 3.49 (s, 3H), 3.45 (s, 1H), 3.22-3.11 (m, 4H), 2.97 (s,3H), 2.89-2.64 (m, 7H), 2.45-2.33 (m, 2H), 2.28 (s, 3H), 2.11 (d, J = 13.3 Hz,3H), 1.94 (d, J = 7.4 Hz, 3H), 1.87-1.79 (m, 1H), 1.68 (d, J = 6.3 Hz, 3H), 1.52 (d,J = 6.9 Hz, 1H)141H NMR (400 MHz, MeOD-d4) δ: 8.75-8.61 (m, 1H), 8.38 (s, 2H), 8.28-8.18950.4CCDB(m, 1H), 7.63 (dd, J = 8.7, 13.6 Hz, 2H), 7.57-7.49 (m, 1H), 7.41 (s, 1H), 7.35-7.28 (m, 1H), 6.98 (s, 1H), 6.87-6.80 (m, 1H), 5.74 (br d, J = 6.0 Hz, 1H), 5.06 (dd,J = 5.6, 12.5 Hz, 1H), 4.62 (br s, 1H), 3.80-3.65 (m, 5H), 3.57 (br d, J = 9.0 Hz,3H), 3.51-3.42 (m, 4H), 3.23-3.10 (m, 3H), 3.06-2.92 (m, 2H), 2.88-2.79 (m,5H), 2.74 (br d, J = 15.9 Hz, 2H), 2.70-2.62 (m, 2H), 2.41 (br s, 1H), 2.28 (s, 3H),2.15-2.01 (m, 3H), 1.93 (br d, J = 8.4 Hz, 2H), 1.87-1.73 (m, 2H), 1.70-1.61 (m,5H), 1.52 (br d, J = 6.5 Hz, 2H)151H NMR (400 MHz, MeOD-d4) δ: 8.67 (s, 1H), 8.28 (d, J = 5.5 Hz, 1H), 7.69 (d,868.3BBDBJ = 8.5 Hz, 1H), 7.64-7.60 (m, 1H), 7.50 (dd, J = 1.8, 8.9 Hz, 1H), 7.45 (s, 1H), 7.39-7.33 (m, 2H), 7.26 (dd, J = 2.3, 8.4 Hz, 1H), 5.74 (d, J = 5.8 Hz, 1H), 5.07 (dd,J = 5.5, 12.4 Hz, 1H), 3.85 (s, 4H), 3.57-3.52 (m, 4H), 3.51-3.47 (m, 3H), 3.21-3.13 (m, 4H), 3.02 (t, J = 7.2 Hz, 2H), 2.88-2.80 (m, 5H), 2.78-2.69 (m, 4H), 2.28(d, J = 0.9 Hz, 3H), 2.14-2.07 (m, 1H), 2.04-1.97 (m, 2H), 1.68 (d, J = 6.4 Hz, 3H)171H NMR (400 MHz, MeOD-d4) δ: 8.56 (d, J = 8.9 Hz, 1H), 8.23 (d, J = 5.3 Hz, 1H),894.6CBDB7.66 (d, J = 8.5 Hz, 1H), 7.63-7.54 (m, 2H), 7.37 (s, 1H), 7.35 (d, J = 2.1 Hz, 1H),7.30 (d, J = 5.4 Hz, 1H), 7.22 (dd, J = 2.3, 8.6 Hz, 1H), 5.74 (q, J = 6.0 Hz, 1H), 5.06(dd, J = 5.4, 12.6 Hz, 1H), 4.02 (br d, J = 13.3 Hz, 2H), 3.68-3.62 (m, 4H), 3.59 (brt, J = 7.6 Hz, 2H), 3.50 (s, 3H), 3.02-2.97 (m, 3H), 2.91-2.79 (m, 2H), 2.78-2.69(m, 2H), 2.68-2.59 (m, 6H), 2.42 (br t, J = 10.4 Hz, 1H), 2.28 (d, J = 1.0 Hz, 3H),2.15-2.07 (m, 1H), 1.88 (br d, J = 12.6 Hz, 2H), 1.69 (d, J = 6.4 Hz, 3H), 1.29 (br s,3H)181H NMR (400 MHz, MeOD-d4) δ: 8.65 (s, 1H), 8.29 (br s, 2H), 8.23 (d, J = 5.4 Hz,922.6CBDC1H), 7.72 (d, J = 8.5 Hz, 1H), 7.64-7.59 (m, 1H), 7.51 (dd, J = 1.8, 9.0 Hz, 1H), 7.42(d, J = 2.1 Hz, 1H), 7.39 (s, 1H), 7.33-7.27 (m, 2H), 5.82-5.69 (m, 1H), 5.15-5.02 (m, 1H), 4.24 (br d, J = 13.5 Hz, 2H), 3.66 (br s, 4H), 3.58 (br d, J = 11.6 Hz,2H), 3.50 (s, 3H), 3.13-2.99 (m, 4H), 2.94-2.81 (m, 1H), 2.79-2.58 (m, 6H),2.56 (s, 1H), 2.38 (br d, J = 7.1 Hz, 1H), 2.28 (s, 3H), 2.26-2.07 (m, 5H), 1.99 (brs, 1H), 1.90-1.76 (m, 2H), 1.68 (d, J = 6.4 Hz, 3H), 1.57-1.42 (m, 3H)201H NMR (400 MHz, MeOD-d4) δ: 8.67 (s, 1H), 8.34 (br s, 2H), 8.27 (d, J = 5.3 Hz,910.5DD1H), 7.57 (dd, J = 8.6, 18.2 Hz, 2H), 7.46 (dd, J = 1.6, 9.0 Hz, 1H), 7.42 (s, 1H), 7.34(d, J = 5.3 Hz, 1H), 6.76 (d, J = 1.9 Hz, 1H), 6.60 (br d, J = 8.4 Hz, 1H), 5.74 (br d,J = 5.8 Hz, 1H), 5.04 (dd, J = 5.4, 12.7 Hz, 1H), 4.22 (s, 8H), 3.82-3.71 (m, 6H),3.67 (br t, J = 4.9 Hz, 2H), 3.48 (s, 3H), 3.28 (br s, 1H), 2.94-2.80 (m, 7H), 2.79-2.61 (m, 3H), 2.26 (s, 3H), 2.13-2.03 (m, 1H), 1.67 (d, J = 6.3 Hz, 3H)211H NMR (400 MHz, MeOD-d4) δ: 8.65 (s, 1H), 8.35 (s, 2H), 8.25 (d, J = 5.4 Hz,951.6DBD1H), 7.64 (dd, J = 8.9, 18.9 Hz, 2H), 7.53-7.47 (m, 1H), 7.40 (s, 1H), 7.36-7.30(m, 2H), 7.25-7.19 (m, 1H), 5.78-5.71 (m, 1H), 5.06 (dd, J = 5.7, 12.7 Hz, 1H),4.05 (br d, J = 13.5 Hz, 2H), 3.72 (br s, 4H), 3.52-3.48 (m, 4H), 3.11-2.96 (m,7H), 2.90-2.70 (m, 12H), 2.43 (br d, J = 4.4 Hz, 2H), 2.28 (d, J = 1.0 Hz, 3H), 2.11(br d, J = 5.3 Hz, 1H), 1.91 (br d, J = 9.8 Hz, 3H), 1.69 (d, J = 6.4 Hz, 3H), 1.31 (br d,J = 14.6 Hz, 3H)221H NMR (400 MHz, MeOD-d4) δ: 8.61 (s, 1H), 8.48 (br s, 1H), 8.23 (d, J = 5.3 Hz,986.4ABAB1H), 7.67 (d, J = 8.5 Hz, 1H), 7.63-7.59 (m, 1H), 7.55-7.50 (m, 1H), 7.39-7.34(m, 2H), 7.30 (d, J = 5.1 Hz, 1H), 7.22 (dd, J = 2.3, 8.6 Hz, 1H), 5.74 (br d, J = 6.1 Hz,1H), 5.19 (s, 1H), 4.11-4.03 (m, 2H), 3.65 (br s, 4H), 3.50 (s, 3H), 3.42-3.37 (m,1H), 3.02 (br t, J = 11.8 Hz, 2H), 2.92-2.80 (m, 4H), 2.79-2.76 (m, 4H), 2.74-2.64 (m, 4H), 2.55 (br s, 1H), 2.33 (br d, J = 7.6 Hz, 1H), 2.28 (d, J = 1.0 Hz, 3H),2.15-2.05 (m, 2H), 2.03-1.96 (m, 2H), 1.91 (br d, J = 11.5 Hz, 2H), 1.83-1.74(m, 2H), 1.68 (d, J = 6.3 Hz, 3H), 1.44-1.27 (m, 3H)231H NMR (400 MHz, MeOD-d4) δ: 8.69 (s, 1H), 8.44 (s, 1H), 8.27 (d, J = 5.3 Hz,950.6BBCB1H), 7.72-7.58 (m, 2H), 7.52 (dd, J = 1.7, 8.9 Hz, 1H), 7.42 (s, 1H), 7.37-7.31 (m,2H), 7.22 (dd, J = 2.3, 8.6 Hz, 1H), 5.76 (br d, J = 6.6 Hz, 1H), 5.08 (dd, J = 5.5, 12.5Hz, 1H), 4.06 (br d, J = 13.1 Hz, 2H), 3.71 (br s, 4H), 3.64 (br d, J = 15.3 Hz, 2H),3.52 (s, 3H), 3.11-2.95 (m, 5H), 2.87-2.69 (m, 11H), 2.30 (s, 3H), 2.17-2.09(m, 1H), 2.01 (br d, J = 12.9 Hz, 2H), 1.86 (br d, J = 11.9 Hz, 2H), 1.70 (d, J = 6.4 Hz,3H), 1.56-1.45 (m, 2H), 1.38-1.23 (m, 5H)241H NMR (400 MHz, MeOD-d4) δ: 8.65 (s, 1H), 8.47 (br s, 1H), 8.24 (d, J = 5.3 Hz,894.5BBBC1H), 7.67 (d, J = 8.5 Hz, 1H), 7.64-7.59 (m, 1H), 7.52 (d, J = 9.0 Hz, 1H), 7.41-7.34 (m, 2H), 7.32 (d, J = 5.3 Hz, 1H), 7.23 (dd, J = 2.1, 8.6 Hz, 1H), 5.78-5.70 (m,1H), 5.07 (dd, J = 5.4, 12.6 Hz, 1H), 4.08 (br d, J = 12.9 Hz, 4H), 3.82 (br s, 2H),3.69 (br s, 4H), 3.50 (s, 3H), 3.27 (br s, 1H), 3.07-2.96 (m, 4H), 2.92-2.81 (m,1H), 2.78-2.63 (m, 2H), 2.57 (br s, 4H), 2.28 (s, 3H), 2.15-2.06 (m, 1H), 1.95-1.81 (m, 3H), 1.68 (d, J = 6.3 Hz, 3H), 1.44-1.32 (m, 2H)251H NMR (400 MHz, MeOD-d4) δ: 8.64 (s, 1H), 8.45 (s, 1H), 8.25 (d, J = 5.3 Hz,951.6BACA1H), 7.66 (d, J = 8.6 Hz, 1H), 7.63-7.60 (m, 1H), 7.52 (dd, J = 1.7, 8.9 Hz, 1H), 7.40(s, 1H), 7.35-7.30 (m, 2H), 7.21 (dd, J = 2.2, 8.6 Hz, 1H), 5.74 (br d, J = 5.9 Hz,1H), 5.06 (br dd, J = 5.4, 12.5 Hz, 1H), 4.05 (br d, J = 13.5 Hz, 2H), 3.71 (br s, 4H),3.50 (s, 3H), 3.00 (br t, J = 11.4 Hz, 7H), 2.88-2.67 (m, 13H), 2.40 (br d, J = 6.3 Hz,2H), 2.28 (s, 3H), 2.15-2.06 (m, 1H), 1.90 (br d, J = 10.9 Hz, 3H), 1.68 (d, J = 6.4Hz, 3H), 1.37-1.25 (m, 3H)261H NMR: (400 MHz, DMSO) δ: 13.45 (s, 1H), 11.07 (s, 1H), 10.29 (s, 1H), 8.53851.3DAD(s, 1H), 8.21-8.18 (m, 1H), 7.66-7.58 (m, 3H), 7.29 (s, 1H), 7.24-7.17 (m, 2H),6.86-6.79 (m, 1H), 5.80-5.68 (m, 1H), 5.06 (dd, J = 5.4, 12.9 Hz, 1H), 4.08 (s,3H), 4.02 (d, J = 13.3 Hz, 2H), 3.35-3.27 (m, 8H), 2.99-2.79 (m, 4H), 2.69-2.53(m, 3H), 2.33 (s, 1H), 2.20 (s, 3H), 2.07-1.97 (m, 1H), 1.75 (d, J = 11.5 Hz, 2H),1.57 (d, J = 6.4 Hz, 3H), 1.23-1.11 (m, 2H)271H NMR (400 MHz, MeOD-d4) δ: 8.67 (s, 1H), 8.26 (d, J = 5.3 Hz, 1H), 7.70 (d,951.3DBDJ = 8.5 Hz, 1H), 7.62 (d, J = 8.9 Hz, 1H), 7.50 (d, J = 1.8 Hz, 1H), 7.40 (s, 1H), 7.37(d, J = 2.1 Hz, 1H), 7.32 (d, J = 5.5 Hz, 1H), 7.24 (dd, J = 2.3, 8.6 Hz, 1H), 5.75 (d,J = 5.8 Hz, 1H), 5.08 (dd, J = 5.5, 12.5 Hz, 1H), 3.70 (d, J = 4.6 Hz, 4H), 3.67-3.59(m, 3H), 3.51 (s, 3H), 3.48 (d, J = 3.8 Hz, 4H), 3.10-3.02 (m, 2H), 2.90-2.81 (m,3H), 2.80-2.72 (m, 6H), 2.68-2.59 (m, 5H), 2.35 (d, J = 7.3 Hz, 2H), 2.29 (d,J = 0.9 Hz, 3H), 2.16-2.06 (m, 3H), 1.95 (s, 1H), 1.69(d, J = 6.3 Hz, 3H), 1.59-1.48 (m, 2H)281H NMR (400 MHz, MeOD-d4) δ: 8.72 (s, 1H), 8.31 (s, 2H), 8.25 (d, J = 5.4 Hz,965.7DBD1H), 7.69-7.54 (m, 2H), 7.48 (dd, J = 1.8, 8.9 Hz, 1H), 7.43 (s, 1H), 7.37-7.29 (m,2H), 7.21 (dd, J = 2.3, 8.7 Hz, 1H), 5.74 (br d, J = 5.9 Hz, 1H), 5.06 (dd, J = 5.4, 12.4Hz, 1H), 4.06 (br t, J = 13.4 Hz, 4H), 3.52-3.49 (m, 3H), 3.27-3.15 (m, 4H), 3.11-2.90 (m, 9H), 2.87-2.66 (m, 9H), 2.42 (br d, J = 6.3 Hz, 2H), 2.28 (d, J = 0.9 Hz,3H), 2.16-2.05 (m, 1H), 1.95-1.83 (m, 3H), 1.68 (d, J = 6.4 Hz, 3H), 1.34-1.24(m, 5H)291H NMR (400 MHz, MeOD-d4) δ: 8.78 (s, 1H), 8.47 (s, 2H), 8.17 (d, J = 5.3 Hz,950.6BCDC1H), 7.68 (d, J = 8.6 Hz, 1H), 7.58 (d, J = 9.0 Hz, 1H), 7.37-7.32 (m, 2H), 7.29-7.22 (m, 3H), 5.07 (dd, J = 5.4, 12.4 Hz, 1H), 4.75 (br s, 1H), 4.71 (s, 1H), 4.09 (brd, J = 13.3 Hz, 2H), 3.70 (s, 1H), 3.63 (br d, J = 11.6 Hz, 2H), 3.58 (br s, 4H), 3.48(s, 3H), 3.16 (br s, 1H), 3.08-3.01 (m, 4H), 2.88-2.82 (m, 1H), 2.79-2.72 (m,2H), 2.68 (br d, J = 13.1 Hz, 1H), 2.60 (br s, 3H), 2.43 (br d, J = 7.0 Hz, 2H), 2.23-2.17 (m, 3H), 2.13-2.07 (m, 1H), 1.98-1.86 (m, 6H), 1.61 (br d, J = 11.4 Hz, 2H),1.51 (s, 3H), 1.46-1.39 (m, 2H), 1.33-1.28 (m, 1H)301H NMR (400 MHz, MeOD-d4) δ: 8.85 (s, 1H), 8.39 (s, 2H), 8.18 (d, J = 5.4 Hz,979.7BCDC1H), 7.65 (d, J = 8.6 Hz, 1H), 7.58 (d, J = 8.9 Hz, 1H), 7.37-7.30 (m, 2H), 7.29-7.22 (m, 2H), 7.19 (dd, J = 2.0, 8.6 Hz, 1H), 5.06 (dd, J = 5.4, 12.4 Hz, 1H), 4.75 (brs, 1H), 4.71 (s, 1H), 4.06-3.96 (m, 3H), 3.88-3.81 (m, 1H), 3.71 (s, 1H), 3.47 (d,J = 2.0 Hz, 4H), 3.25 (br d, J = 10.8 Hz, 1H), 3.22-3.14 (m, 5H), 3.12 (br d, J = 12.9Hz, 1H), 3.01-2.93 (m, 3H), 2.90 (br d, J = 4.8 Hz, 1H), 2.88-2.85 (m, 1H), 2.84-2.74 (m, 6H), 2.74-2.64 (m, 2H), 2.39 (br d, J = 4.3 Hz, 2H), 2.28-2.04 (m, 2H),1.91-1.83 (m, 6H), 1.51 (s, 2H), 1.33-1.19 (m, 6H)311H NMR (400 MHz, MeOD-d4) δ: 8.73-8.64 (m, 1H), 8.49 (br s, 1H), 8.23 (d,965.8DCDJ = 5.5 Hz, 1H), 7.68 (d, J = 8.5 Hz, 1H), 7.64-7.56 (m, 1H), 7.49 (br d, J = 9.0 Hz,1H), 7.41-7.31 (m, 2H), 7.30 (br d, J = 4.8 Hz, 1H), 7.26-7.19 (m, 1H), 5.73 (br d,J = 6.5 Hz, 1H), 5.07 (br dd, J = 5.5, 12.3 Hz, 1H), 3.98 (br d, J = 12.6 Hz, 2H), 3.57-3.37 (m, 9H), 3.23-3.04 (m, 2H), 2.96-2.68 (m, 12H), 2.61 (br s, 4H), 2.33 (br d,J = 7.4 Hz, 2H), 2.28 (s, 2H), 2.17-1.98 (m, 4H), 1.88 (br s, 1H), 1.68 (d, J = 6.4 Hz,2H), 1.61-1.42 (m, 2H), 1.31-1.14 (m, 4H)321H NMR (400 MHz, MeOD-d4) δ: 8.54 (dd, J = 1.3, 8.9 Hz, 1H), 8.51 (s, 1H), 8.22952.6BCD(d, J = 5.4 Hz, 1H), 7.65 (d, J = 8.5 Hz, 1H), 7.57 (d, J = 8.9 Hz, 1H), 7.37-7.32 (m,2H), 7.30 (s, 1H), 7.27 (d, J = 5.3 Hz, 1H), 7.20 (dd, J = 2.3, 8.6 Hz, 1H), 5.06 (dd,J = 5.4, 12.4 Hz, 1H), 4.75 (s, 1H), 4.70 (d, J = 1.5 Hz, 1H), 4.19 (br d, J = 12.5 Hz,1H), 4.08-4.00 (m, 4H), 3.88 (br s, 1H), 3.77-3.71 (m, 1H), 3.70 (s, 1H), 3.47 (d,J = 0.8 Hz, 3H), 3.05-2.93 (m, 6H), 2.89-2.81 (m, 3H), 2.81-2.75 (m, 4H), 2.74-2.63 (m, 4H), 2.44 (br d, J = 6.0 Hz, 2H), 2.14-2.07 (m, 1H), 1.92-1.87 (m, 6H),1.51 (s, 3H), 1.32 (br s, 2H)331H NMR (400 MHz, MeOD-d4) δ: 8.88 (s, 1H), 8.39 (s, 1H), 8.16 (d, J = 5.4 Hz,965.7DCDC1H), 7.68 (d, J = 8.6 Hz, 1H), 7.58 (d, J = 8.9 Hz, 1H), 7.38-7.33 (m, 2H), 7.30-7.20 (m, 3H), 5.07 (br dd, J = 5.4, 12.4 Hz, 1H), 4.76 (br s, 1H), 4.72 (br s, 1H),3.82 (br d, J = 9.6 Hz, 2H), 3.71 (s, 1H), 3.58 (br s, 3H), 3.54-3.51 (m, 1H), 3.50-3.46 (m, 6H), 3.21 (br t, J = 11.7 Hz, 2H), 2.88 (br t, J = 6.3 Hz, 2H), 2.83 (br d,J = 5.6 Hz, 1H), 2.78-2.75 (m, 1H), 2.73 (br s, 1H), 2.71-2.60 (m, 9H), 2.41-2.33(m, 2H), 2.22-2.06 (m, 4H), 2.03 (br d, J = 7.1 Hz, 1H), 1.90-1.86 (m, 3H), 1.68-1.57 (m, 2H), 1.52 (s, 3H), 1.34-1.27 (m, 1H)341H NMR (400 MHz, MeOD-d4) δ: 8.76 (br s, 1H), 8.53 (s, 1H), 8.15 (d, J = 5.3 Hz,964.7BCD1H), 7.68 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.9 Hz, 1H), 7.36 (d, J = 2.1 Hz, 1H), 7.31(s, 1H), 7.29-7.19 (m, 3H), 5.07 (dd, J = 5.5, 12.4 Hz, 1H), 4.75 (br s, 1H), 4.71 (brs, 1H), 4.09 (br d, J = 12.8 Hz, 2H), 3.92 (br d, J = 11.5 Hz, 1H), 3.80 (br d, J = 12.0Hz, 1H), 3.71 (s, 1H), 3.54 (br s, 2H), 3.47 (s, 3H), 3.07-2.97 (m, 6H), 2.91-2.81(m, 2H), 2.75-2.65 (m, 3H), 2.57 (br s, 1H), 2.28-2.19 (m, 3H), 2.15-2.09 (m,2H), 2.04 (br s, 1H), 1.93 (br d, J = 11.9 Hz, 2H), 1.88 (s, 3H), 1.58 (br d, J = 8.6 Hz,2H), 1.50 (s, 3H), 1.42 (br d, J = 10.9 Hz, 2H), 1.35-1.27 (m, 4H), 1.11 (br t, J = 6.4Hz, 3H)351H NMR (400 MHz, MeOD-d4) δ: 8.83 (s, 1H), 8.45 (s, 1H), 8.18 (d, J = 5.3 Hz,965.6BCD1H), 7.66 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.9 Hz, 1H), 7.35-7.31 (m, 2H), 7.29-7.19 (m, 3H), 5.06 (br dd, J = 5.4, 12.3 Hz, 1H), 4.75-4.75 (m, 1H), 4.71 (br s, 1H),4.06 (br s, 1H), 4.03 (br s, 1H), 3.70 (s, 1H), 3.62 (br s, 4H), 3.48 (s, 3H), 2.99 (brt, J = 12.0 Hz, 4H), 2.90 (br s, 2H), 2.86 (br d, J = 4.1 Hz, 1H), 2.87-2.85 (m, 1H),2.82 (br s, 4H), 2.76 (br s, 2H), 2.73 (br s, 2H), 2.72-2.65 (m, 1H), 2.70-2.65 (m,1H), 2.41 (br d, J = 5.5 Hz, 2H), 2.12-2.06 (m, 1H), 1.95-1.84 (m, 8H), 1.51 (s,3H), 1.30 (br d, J = 11.4 Hz, 4H)361H NMR (400 MHz, MeOD-d4) δ: 8.79 (br s, 1H), 8.55 (s, 1H), 8.16 (d, J = 5.4979.7DCDHz, 1H), 7.68 (d, J = 8.5 Hz, 1H), 7.57 (d, J = 9.0 Hz, 1H), 7.34 (br d, J = 12.4 Hz,2H), 7.28-7.18 (m, 3H), 5.07 (br dd, J = 5.3, 12.3 Hz, 1H), 4.75 (br s, 1H), 4.70(br s, 1H), 3.90 (br d, J = 11.4 Hz, 1H), 3.77 (br d, J = 11.6 Hz, 1H), 3.70 (s, 1H),3.50-3.44 (m, 8H), 3.20 (br d, J = 12.9 Hz, 2H), 2.99 (br d, J = 3.1 Hz, 1H), 2.91-2.81 (m, 3H), 2.71 (br d, J = 10.9 Hz, 4H), 2.60 (br s, 4H), 2.49 (br t, J = 9.1 Hz,1H), 2.33 (br d, J = 6.5 Hz, 2H), 2.24-2.13 (m, 1H), 2.13-2.08 (m, 1H), 2.03 (brd, J = 12.9 Hz, 2H), 1.90-1.86 (m, 4H), 1.51 (s, 4H), 1.36-1.23 (m, 4H), 1.16 (brt, J = 6.6 Hz, 3H)*DC50Ranges: A < 10; 10 ≤ B < 50; 50 ≤ C < 100; D ≥ 100.**DMaxRanges: A ≥ 70; 50 ≤ B < 70; C < 50 A novel bifunctional molecule, which contains a recruiting moiety that selectively or preferentially binds to a LRRK2 protein having at least one mutation that is G2019S and an E3 ubiquitin ligase recruiting moiety is described. The bifunctional molecules of the present disclosure actively ubiquitinate the mutated LRRK2, resulting in proteasomal degradation, leading to suppression of cellular proliferation and induction of apoptosis. The contents of all references, patents, pending patent applications and published patents, cited throughout this application are hereby expressly incorporated by reference. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the disclosure described herein. Such equivalents are intended to be encompassed by the following claims. It is understood that the detailed examples and embodiments described herein are given by way of example for illustrative purposes only, and are in no way considered to be limiting to the disclosure. Various modifications or changes in light thereof will be suggested to persons skilled in the art and are included within the spirit and purview of this application and are considered within the scope of the appended claims. For example, the relative quantities of the ingredients may be varied to optimize the desired effects, additional ingredients may be added, and/or similar ingredients may be substituted for one or more of the ingredients described. Additional advantageous features and functionalities associated with the systems, methods, and processes of the present disclosure will be apparent from the appended claims. Moreover, those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the disclosure described herein. Such equivalents are intended to be encompassed by the following claims.
303,559
11858941
DETAILED DESCRIPTION An aspect of the present description relates to compounds comprising, a compound of Formula (I): or a form thereof, wherein:the dashed lines represent one or more double bonds optionally present where allowed by available valences;W1is independently C—Ra, CH—Ra, N, N—Rb, O, or S where allowed by available valences;W2is independently C—Ra, CH—Ra, N, or N—Rbwhere allowed by available valences, and;W3is independently C, CH, or N where allowed by available valences;wherein at least one of W1, W2, or W3is N or N—Rb;W4and W5are independently C—Raor N,wherein when W1is S or O, W2is C—Ra, and W3is C;Rais, in each instance, independently selected from hydrogen, cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and hydroxy-C1-6alkyl;Rbis selected from hydrogen and C1-6alkyl;R1is selected from C3-10cycloalkyl and heterocyclyl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of C3-10cycloalkyl and heterocyclyl is optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or,wherein, alternatively, each instance of C3-10cycloalkyl and heterocyclyl is optionally substituted with one, two, three, or four R3substituents;R2is selected from phenyl, heterocyclyl, and heteroaryl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of phenyl, heterocyclyl, and heteroaryl is optionally substituted with one, two, or three R5substituents, and optionally, with one additional R6substituent;R3is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and hydroxy-C1-6alkyl;R4is selected from C3-10cycloalkyl, phenyl, heteroaryl, and heterocyclyl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of C3-10cycloalkyl, phenyl, heterocyclyl, and heteroaryl is optionally substituted with one, two, or three R7substituents;R5is, in each instance, independently selected from halogen, hydroxy, cyano, nitro, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, oxime, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, and C1-6alkyl-thio;R6is selected from phenyl and heteroaryl,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, and wherein, each instance of phenyl and heteroaryl is optionally substituted with one, two, three or four R8substituents;R7is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and C3-10cycloalkyl; andR8is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and C3-10cycloalkyl;wherein a form of the compound is selected from the group consisting of salt, hydrate, solvate, racemate, enantiomer, diastereomer, stereoisomer, and tautomer form thereof. ASPECTS OF THE DESCRIPTION One aspect of the present description includes a compound of Formula (I) comprising, a compound of Formula (I.1): or a form thereof, wherein:the dashed lines represent one or more double bonds optionally present where allowed by available valences;W1is independently C—Ra, CH—Ra, N, N—Rb, O, or S where allowed by available valences;W2is independently C—Ra, CH—Ra, N, or N—Rbwhere allowed by available valences, and;W3is independently C, CH, or N where allowed by available valences; wherein at least one of W1, W2, or W3is N or N—Rb;W4and W5are independently C—Raor N,wherein when W1is S or O, W2is C—Ra, and W3is C;Rais, in each instance, independently selected from hydrogen, cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and hydroxy-C1-6alkyl;Rbis selected from hydrogen and C1-6alkyl;R1is selected from C3-10cycloalkyl and heterocyclyl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of C3-10cycloalkyl and heterocyclyl is optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or,wherein, alternatively, each instance of C3-10cycloalkyl and heterocyclyl is optionally substituted with one, two, three, or four R3substituents;R2is selected from phenyl, heterocyclyl, and heteroaryl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of phenyl, heterocyclyl, and heteroaryl is optionally substituted with one, two, or three R5substituents, and optionally, with one additional R6substituent;R3is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and hydroxy-C1-6alkyl;R4is selected from C3-10cycloalkyl, phenyl, heteroaryl, and heterocyclyl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of C3-10cycloalkyl, phenyl, heterocyclyl, and heteroaryl is optionally substituted with one, two, or three R7substituents;R5is, in each instance, independently selected from halogen, hydroxy, cyano, nitro, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, oxime, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, and C1-6alkyl-thio;R6is selected from phenyl and heteroaryl,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of phenyl and heteroaryl is optionally substituted with one, two, three or four R8substituents;R7is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and C3-10cycloalkyl;R8is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, or C3-10cycloalkyl. One aspect includes a compound of Formula (I), wherein W1is N—Rband W4is N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is C—Ra, W3is C, W4is N and W5is C—Ra. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is CH—Ra, W3is CH, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W2is N—Rband W4is N. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is N—Rb, W3is CH, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W3and W4are N. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is C—Ra, W3is N, W4is N and W5is C—Ra. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is CH—Ra, W3is N, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W1, W2and W4are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is N, W3is CH, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W1and W2are N—Rband W4is N. Another aspect includes a compound of Formula (I), wherein W1and W2are N—Rb, W3is CH, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W1, W3and W4are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is C—Ra, W3is N, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W1is N—Rband W3and W4are N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is CH—Ra, W3is N, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W2, W3and W4are N. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is N, W3is N, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W2is N—Rband W3and W4are N. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is N—Rb, W3is N, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W1, W2, W3and W4are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is N, W3is N, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W1is S and W4is N. Another aspect includes a compound of Formula (I), wherein W1is S, W2is C—Ra, W3is C, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W1is O and W4is N. Another aspect includes a compound of Formula (I), wherein W1is O, W2is C—Ra, W3is C, W4is N and W5is C—Ra. One aspect includes a compound of Formula (I), wherein W1is N—Rband W5is N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is C—Ra, W3is C, W4is C—Raand W5is N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is CH—Ra, W3is CH, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W2is N—Rband W5is N. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is N—Rb, W3is CH, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W3and W5are N. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is C—Ra, W3is N, W4is C—Raand W5is N. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is CH—Ra, W3is N, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W1, W2and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is N, W3is CH, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W1and W2are N—Rband W5is N. Another aspect includes a compound of Formula (I), wherein W1and W2are N—Rb, W3is CH, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W1, W3and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is C—Ra, W3is N, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W1is N—Rband W3and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is CH—Ra, W3is N, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W2, W3and W5are N. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is N, W3is N, W4is C—Rband W5is N. One aspect includes a compound of Formula (I), wherein W2is N—Rband W3and W5are N. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is N—Rb, W3is N, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W1, W2, W3and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is N, W3is N, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W1is S and W5is N. Another aspect includes a compound of Formula (I), wherein W1is S, W2is C—Ra, W3is C, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W1is O and W5is N. Another aspect includes a compound of Formula (I), wherein W1is O, W2is C—Ra, W3is C, W4is C—Raand W5is N. One aspect includes a compound of Formula (I), wherein W1is N—Rb. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is C—Ra, W3is C and W4and W5are C—Ra. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is CH—Ra, W3is CH and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W2is N—Rb. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is N—Rb, W3is C and W4and W5are C—Ra. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is N—Rb, W3is CH and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W3is N. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is C—Ra, W3is N and W4and W5are C—Ra. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is CH—Ra, W3is N and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W1and W2are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is N, W3is CH and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W1and W2are N—Rb. Another aspect includes a compound of Formula (I), wherein W1and W2are N—Rb, W3is CH and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W1and W3are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is C—Ra, W3is N and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W1is N—Rband W3is N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is CH—Ra, W3is N and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W2and W3are N. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is N, W3is N and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W2is N—Rband W3is are N. Another aspect includes a compound of Formula (I), wherein W1is CH—Rb, W2is N—Rb, W3is N and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W1, W2and W3are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is N, W3is N and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W1is S. Another aspect includes a compound of Formula (I), wherein W1is S, W2is C—Ra, W3is C and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W1is O. Another aspect includes a compound of Formula (I), wherein W1is O, W2is C—Ra, W3is C and W4and W5are C—Ra. One aspect includes a compound of Formula (I), wherein W1is N—Rband W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is C—Ra, W3is C and W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is CH—Ra, W3is CH and W4and W5are N. One aspect includes a compound of Formula (I), wherein W2is N—Rband W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is N—Rb, W3is CH and W4and W5are N. One aspect includes a compound of Formula (I), wherein W3, W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is C—Ra, W3is N and W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is CH—Ra, W3is N and W4and W5are N. One aspect includes a compound of Formula (I), wherein W1, W2, W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is N, W3is CH and W4and W5are N. One aspect includes a compound of Formula (I), wherein W1and W2are N—Rband W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1and W2are N—Rb, W3is CH and W4and W5are N. One aspect includes a compound of Formula (I), wherein W1, W3, W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is C—Ra, W3is N and W4and W5are N. One aspect includes a compound of Formula (I), wherein W1is N—Rband W3, W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N—Rb, W2is CH—Ra, W3is N and W4and W5are N. One aspect includes a compound of Formula (I), wherein W2, W3, W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is C—Ra, W2is N, W3is N and W4and W5are N. One aspect includes a compound of Formula (I), wherein W2is N—Rband W3, W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is CH—Ra, W2is N—Rb, W3is N and W4and W5are N. One aspect includes a compound of Formula (I), wherein W1, W2, W3, and W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is N, W2is N, W3is N, and W4and W5are N. One aspect includes a compound of Formula (I), wherein W1is S and W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is S, W2is C—Ra, W3is C and W4and W5are N. One aspect includes a compound of Formula (I), wherein W1is O and W4and W5are N. Another aspect includes a compound of Formula (I), wherein W1is O, W2is C—Ra, W3is C and W4and W5are N. One aspect includes a compound of Formula (I), wherein Rais, in each instance, independently selected from hydrogen, cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and hydroxy-C1-6alkyl. Another aspect includes a compound of Formula (I), wherein Rais, in each instance, independently selected from hydrogen, hydroxy, C1-6alkyl, C1-6alkoxy, and C1-6alkyl-amino. Another aspect includes a compound of Formula (I), wherein Rais hydrogen. Another aspect includes a compound of Formula (I), wherein Rais hydroxy. Another aspect includes a compound of Formula (I), wherein Rais C1-6alkyl selected from methyl, ethyl, propyl, isopropyl, and tert-butyl. Another aspect includes a compound of Formula (I), wherein Rais methyl. Another aspect includes a compound of Formula (I), wherein Rais C1-6alkoxy selected from methoxy, ethoxy, propoxy, isopropoxy, and tert-butoxy. Another aspect includes a compound of Formula (I), wherein Rais methoxy. Another aspect includes a compound of Formula (I), wherein Rais C1-6alkyl-amino wherein C1-6alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, and tert-butyl. Another aspect includes a compound of Formula (I), wherein Rais C1-6alkyl-amino selected from methylamino and ethylamino. One aspect includes a compound of Formula (I), wherein Rbis selected from hydrogen and C1-6alkyl. Another aspect includes a compound of Formula (I), wherein Rbis hydrogen. One aspect includes a compound of Formula (I), wherein R1is selected from C3-10cycloalkyl and heterocyclyl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of C3-10cycloalkyl and heterocyclyl is optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or,wherein, alternatively, each instance of C3-10cycloalkyl and heterocyclyl is optionally substituted with one, two, three, or four R3substituents. Another aspect includes a compound of Formula (I), wherein R1is C3-10cycloalkyl, optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or alternatively, optionally substituted with one, two, three, or four R3substituents. Another aspect includes a compound of Formula (I), wherein R1is C3-10cycloalkyl selected from cyclopropyl, cylcobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, bicyclo[2.2.1]hexanyl, and adamantyl, optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or alternatively, optionally substituted with one, two, three, or four R3substituents. Another aspect includes a compound of Formula (I), wherein R1is C3-10cycloalkyl selected from cylcobutyl and cyclohexyl, optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or alternatively, optionally substituted with one, two, three, or four R3substituents. Another aspect includes a compound of Formula (I), wherein R1is heterocyclyl selected from azetidinyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl, 1,4-diazepanyl, 1,2,5,6-tetrahydropyridinyl, 1,2,3,6-tetrahydropyridinyl, 3-azabicyclo[3.1.0]hexyl, (1R,5S)-3-azabicyclo[3.1.0]hexyl, 8-azabicyclo[3.2.1]octyl, (1R,5S)-8-azabicyclo[3.2.1]octyl, 8-azabicyclo[3.2.1]oct-2-en-yl, (1R,5S)-8-azabicyclo[3.2.1]oct-2-en-yl, 9-azabicyclo[3.3.1]nonyl, (1R,5S)-9-azabicyclo[3.3.1]nonyl, 3-oxa-9-azabicyclo[3.3.1]nonyl, and 3-oxa-9-azabicyclo[3.3.1]non-6-en-yl, optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or, alternatively, optionally substituted with one, two, three or four R3substituents. Another aspect includes a compound of Formula (I), wherein R1is heterocyclyl selected from piperidinyl, piperazinyl, 1,2,3,6-tetrahydropyridinyl, 8-azabicyclo[3.2.1]octyl, (1R,5S)-8-azabicyclo[3.2.1]octyl, 8-azabicyclo[3.2.1]oct-2-en-yl, 3-oxa-9-azabicyclo[3.3.1]nonyl, and 3-oxa-9-azabicyclo[3.3.1]non-6-en-yl, optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or, alternatively, optionally substituted with one, two, three or four R3substituents. Another aspect includes a compound of Formula (I), wherein R1is heterocyclyl selected from azetidin-1-yl, tetrahydrofuran-3-yl, pyrrolidin-1-yl, piperidin-1-yl, piperidin-4-yl, piperazin-1-yl, 1,4-diazepan-1-yl, 1,2,5,6-tetrahydropyridin-5-yl, 1,2,3,6-tetrahydropyridin-4-yl, 8-azabicyclo[3.2.1]oct-3-yl, (1R,5S)-8-azabicyclo[3.2.1]oct-3-yl, 8-azabicyclo[3.2.1]oct-2-en-3-yl, (1R,5S)-8-azabicyclo[3.2.1]oct-2-en-3-yl, 9-azabicyclo[3.3.1]non-7-yl, (1R,5S)-9-azabicyclo[3.3.1]non-3-yl, 3-oxa-9-azabicyclo[3.3.1]non-7-yl, and 3-oxa-9-azabicyclo[3.3.1]non-6-en-7-yl, optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or, alternatively, optionally substituted with one, two, three or four R3substituents. Another aspect includes a compound of Formula (I), wherein R1is heterocyclyl selected from piperidin-4-yl, piperazin-1-yl, 1,2,3,6-tetrahydropyridin-4-yl, 8-azabicyclo[3.2.1]oct-3-yl, (1R,5S)-8-azabicyclo[3.2.1]oct-3-yl, 8-azabicyclo[3.2.1]oct-2-en-3-yl, 3-oxa-9-azabicyclo[3.3.1]non-7-yl, and 3-oxa-9-azabicyclo[3.3.1]non-6-en-7-yl, optionally substituted with one, two or three R3substituents and optionally, with one additional R4substituent, or, alternatively, optionally substituted with one, two, three or four R3substituents. One aspect includes a compound of Formula (I), wherein R2is selected from phenyl, heterocyclyl, and heteroaryl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of phenyl, heterocyclyl, and heteroaryl is optionally substituted with one, two, or three R5substituents, and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is phenyl, optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is heterocyclyl selected from azetidinyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl, 1,4-diazepanyl, 1,2,5,6-tetrahydropyridinyl, 1,2,3,6-tetrahydropyridinyl, and 2,3-dihydro-1H-indenyl, optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is 2,3-dihydro-1H-indenyl, optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is heterocyclyl selected from azetidin-1-yl, tetrahydrofuran-3-yl, pyrrolidin-1-yl, piperidin-1-yl, piperidin-4-yl, piperazin-1-yl, 1,4-diazepan-1-yl, 1,2,5,6-tetrahydropyridin-5-yl, 1,2,3,6-tetrahydropyridin-4-yl, and 2,3-dihydro-1H-inden-5-yl, optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is 2,3-dihydro-1H-inden-5-yl optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is heteroaryl selected from furanyl, 1H-pyrrolyl, 1H-pyrazolyl, 1H-imidazolyl, 1,3-oxazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, 1H-indolyl, 1H-indazolyl, benzofuranyl, 1H-benzimidazolyl, 1H-benzotriazolyl, and quinolinyl, optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is heteroaryl selected from pyridinyl, 1H-indazolyl, 1H-benzimidazolyl, 1H-benzotriazolyl, and quinolinyl, optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is heteroaryl selected from furan-3-yl, 1H-pyrrol-3-yl, 1H-pyrazol-1-yl, 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, 1H-imidazol-1-yl, 1H-imidazol-2-yl, 1H-imidazol-4-yl, 1,3-oxazol-4-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, pyridazin-3-yl, pyridazin-4-yl, pyridazin-5-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyrazin-2-yl, pyrazin-3-yl, 1H-indol-3-yl, 1H-indol-4-yl, 1H-indol-5-yl, 1H-indol-6-yl, 1H-indazol-5-yl, 1H-indazol-6-yl, benzofuran-2-yl, benzofuran-5-yl, 1H-benzimidazol-5-yl, 1H-benzimidazol-6-yl, 1H-benzotriazol-4-yl, 1H-benzotriazol-5-yl, 1H-benzotriazol-6-yl, 1H-benzotriazol-7-yl, and quinolin-7-yl, optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. Another aspect includes a compound of Formula (I), wherein R2is heteroaryl selected from pyridin-2-yl, 1H-indazol-6-yl, 1H-benzimidazol-6-yl, 1H-benzotriazol-7-yl, and quinolin-7-yl, optionally substituted with one, two or three R5substituents and optionally, with one additional R6substituent. One aspect includes a compound of Formula (I), wherein R3is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and hydroxy-C1-6alkyl. Another aspect includes a compound of Formula (I), wherein R3is, in each instance, independently selected from halogen, C1-6alkyl, and C1-6alkyl-amino. Another aspect includes a compound of Formula (I), wherein R3is halogen selected from bromo, chloro, fluoro, and iodo. Another aspect includes a compound of Formula (I), wherein R3is fluoro. Another aspect includes a compound of Formula (I), wherein R3is C1-6alkyl selected from methyl, ethyl, propyl, isopropyl, and tert-butyl. Another aspect includes a compound of Formula (I), wherein R3is methyl. Another aspect includes a compound of Formula (I), wherein R3is C1-6alkyl-amino wherein C1-6alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, and tert-butyl. Another aspect includes a compound of Formula (I), wherein R3is tert-butylamino. One aspect includes a compound of Formula (I), wherein R4is selected from C3-10cycloalkyl, phenyl, heteroaryl, and heterocyclyl,wherein heterocyclyl is a saturated or partially unsaturated 3-7 membered monocyclic, 6-10 membered bicyclic or 13-16 membered polycyclic ring system having 1, 2, or 3 heteroatom ring members independently selected from N, O, or S,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of C3-10cycloalkyl, phenyl, heterocyclyl, and heteroaryl is optionally substituted with one, two, or three R7substituents. One aspect includes a compound of Formula (I), wherein R5is, in each instance, independently selected from halogen, hydroxy, cyano, nitro, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, oxime, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, and C1-6alkyl-thio. Another aspect includes a compound of Formula (I), wherein R5is, in each instance, independently selected from halogen, hydroxy, C1-6alkyl, and oxime. Another aspect includes a compound of Formula (I), wherein R5is halogen selected from bromo, chloro, fluoro, and iodo. Another aspect includes a compound of Formula (I), wherein R5is halogen selected from chloro and fluoro. Another aspect includes a compound of Formula (I), wherein R5is hydroxy. Another aspect includes a compound of Formula (I), wherein R5is C1-6alkyl selected from methyl, ethyl, propyl, isopropyl, and tert-butyl. Another aspect includes a compound of Formula (I), wherein R5methyl. Another aspect includes a compound of Formula (I), wherein R5is oxime. One aspect includes a compound of Formula (I), wherein R6is selected from phenyl and heteroaryl,wherein heteroaryl is a 3-7 membered monocyclic or 6-10 membered bicyclic ring system having 1, 2, 3, or 4 heteroatom ring members independently selected from N, O, or S, andwherein, each instance of phenyl and heteroaryl is optionally substituted with one, two, three or four R8substituents. Another aspect includes a compound of Formula (I), wherein R6is phenyl, optionally substituted with one, two, three or four R8substituents. Another aspect includes a compound of Formula (I) wherein R6is heteroaryl selected from furanyl, thienyl, 1H-pyrrolyl, 1H-pyrazolyl, 1H-imidazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl, 4H-1,2,4-triazolyl, 1,3-thiazolyl, 1,2-oxazolyl, 1,3-oxazolyl, 1,2,4-thiadiazolyl, 1,3,4-thiadiazol-yl, pyridinyl, pyridin-2(1H)-on-yl, pyridazinyl, pyrimidinyl, pyrimidin-4(3H)-on-yl, pyrazinyl, 1,3,5-triazinyl, 1H-indolyl, 1H-indazolyl, benzofuranyl, 1H-benzimidazolyl, 1H-benzotriazolyl, 1H-pyrrolo[2,3-b]pyridinyl, 1H-pyrrolo[2,3-c]pyridinyl, imidazo[1,2-a]pyridinyl, imidazo[1,2-a]pyrimidinyl, imidazo[1,2-c]pyrimidinyl, imidazo[1,2-b]pyridazinyl, imidazo[1,2-a]pyrazinyl, imidazo[1,5-a]pyridinyl, [1,2,3]triazolo[1,5-a]pyridinyl, 1H-[1,2,3]triazolo[4,5-b]pyridinyl, 3H-[1,2,3]triazolo[4,5-c]pyridinyl, 3H-[1,2,3]triazolo[4,5-c]pyridazinyl, [1,2,4]triazolo[1,5-a]pyridinyl, [1,2,4]triazolo[4,3-b]pyridazinyl, and quinolinyl, optionally substituted with one, two, three or four R8substituents. Another aspect includes a compound of Formula (I) wherein R6is heteroaryl selected from furanyl, thienyl, 1H-pyrrolyl, 1H-pyrazolyl, 1H-imidazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl, 4H-1,2,4-triazolyl, 1,3-thiazolyl, 1,2-oxazolyl, 1,3-oxazolyl, 1,2,4-thiadiazolyl, 1,3,4-thiadiazol-yl, pyridinyl, pyridin-2(1H)-on-yl, pyridazinyl, pyrimidinyl, pyrimidin-4(3H)-on-yl, pyrazinyl, 1,3,5-triazinyl, 1H-benzotriazolyl, 1H-pyrrolo[2,3-b]pyridinyl, imidazo[1,2-a]pyridinyl, imidazo[1,2-a]pyrimidinyl, imidazo[1,2-b]pyridazinyl, imidazo[1,2-a]pyrazinyl, imidazo[1,5-a]pyridinyl, [1,2,3]triazolo[1,5-a]pyridinyl, 1H-[1,2,3]triazolo[4,5-b]pyridinyl, 3H-[1,2,3]triazolo[4,5-c]pyridinyl, [1,2,4]triazolo[1,5-a]pyridinyl, and [1,2,4]triazolo[4,3-b]pyridazinyl, optionally substituted with one, two, three or four R8substituents. Another aspect includes a compound of Formula (I), wherein R6is heteroaryl selected from furan-2-yl, furan-3-yl, thien-2-yl, thien-3-yl, 1H-pyrrol-3-yl, 1H-pyrazol-1-yl, 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl, 1H-imidazol-1-yl, 1H-imidazol-2-yl, 1H-imidazol-4-yl, 1H-1,2,3-triazol-1-yl, 1H-1,2,3-triazol-4-yl, 2H-1,2,3-triazol-2-yl, 2H-1,2,3-triazol-4-yl, 1H-1,2,4-triazol-1-yl, 4H-1,2,4-triazol-4-yl, 1,3-thiazol-2-yl, 1,3-thiazol-5-yl, 1,2-oxazol-4-yl, 1,3-oxazol-2-yl, 1,3-oxazol-3-yl, 1,3-oxazol-4-yl, 1,3-oxazol-5-yl, 1,2,4-thiadiazol-5-yl, 1,3,4-thiadiazol-2-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, pyridin-2(1H)-on-4-yl, pyridazin-3-yl, pyridazin-4-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyrimidin-4(3H)-on-6-yl, pyrazin-1-yl, pyrazin-2-yl, 1,3,5-triazin-2-yl, 1H-indol-3-yl, 1H-indol-4-yl, 1H-indol-5-yl, 1H-indol-6-yl, 1H-indazol-5-yl, 1H-indazol-6-yl, benzofuran-2-yl, benzofuran-5-yl, 1H-benzimidazol-2-yl, 1H-benzimidazol-5-yl, 1H-benzimidazol-6-yl, 1H-benzotriazol-4-yl, 1H-benzotriazol-5-yl, 1H-benzotriazol-6-yl, 1H-benzotriazol-7-yl, 1H-pyrrolo[2,3-b]pyridin-4-yl, 1H-pyrrolo[2,3-b]pyridin-5-yl, 1H-pyrrolo[2,3-c]pyridin-4-yl, pyrrolo[1,2-a]pyrimidin-7-yl, pyrrolo[1,2-a]pyrazin-7-yl, pyrrolo[1,2-b]pyridazin-2-yl, pyrazolo[1,5-a]pyridin-2-yl, pyrazolo[1,5-a]pyridin-5-yl, 2H-pyrazolo[4,3-b]pyridin-5-yl, 2H-pyrazolo[4,3-c]pyridin-5-yl, pyrazolo[1,5-a]pyrazin-2-yl, imidazo[1,2-a]pyridin-2-yl, imidazo[1,2-a]pyridin-6-yl, imidazo[1,2-a]pyridin-7-yl, imidazo[1,2-a]pyrimidin-2-yl, imidazo[1,2-a]pyrimidin-6-yl, imidazo[1,2-c]pyrimidin-2-yl, imidazo[1,2-b]pyridazin-2-yl, imidazo[1,2-b]pyridazin-6-yl, imidazo[1,2-a]pyrazin-2-yl, imidazo[1,2-a]pyrazin-3-yl, imidazo[1,2-a]pyrazin-6-yl, imidazo[1,5-a]pyridine-6-yl, imidazo[1,5-a]pyridin-7-yl, [1,2,3]triazolo[1,5-a]pyridin-5-yl, [1,2,3]triazolo[1,5-a]pyridin-7-yl, 1H-[1,2,3]triazolo[4,5-b]pyridin-5-yl, 1H-[1,2,3]triazolo[4,5-b]pyridin-6-yl, 3H-[1,2,3]triazolo[4,5-c]pyridin-6-yl, 3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl, [1,2,4]triazolo[1,5-a]pyridin-7-yl, [1,2,4]triazolo[4,3-b]pyridazin-6-yl, quinolin-6-yl, quinolin-7-yl, and quinolin-8-yl, optionally substituted with one, two, three or four R8substituents. Another aspect includes a compound of Formula (I), wherein R6is heteroaryl selected from furan-3-yl, thien-3-yl, 1H-pyrrol-3-yl, 1H-pyrazol-1-yl, 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl, 1H-imidazol-1-yl, 1H-imidazol-2-yl, 1H-imidazol-4-yl, 1H-1,2,3-triazol-1-yl, 1H-1,2,3-triazol-4-yl, 2H-1,2,3-triazol-2-yl, 2H-1,2,3-triazol-4-yl, 1H-1,2,4-triazol-1-yl, 4H-1,2,4-triazol-4-yl, 1,3-thiazol-2-yl, 1,3-thiazol-5-yl, 1,2-oxazol-4-yl, 1,3-oxazol-2-yl, 1,3-oxazol-5-yl, 1,2,4-thiadiazol-5-yl, 1,3,4-thiadiazol-2-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, pyridin-2(1H)-on-4-yl, pyridazin-3-yl, pyridazin-4-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyrimidin-4(3H)-on-6-yl, pyrazin-2-yl, 1,3,5-triazin-2-yl, 1H-benzotriazol-6-yl, 1H-pyrrolo[2,3-b]pyridin-4-yl, 1H-pyrrolo[2,3-c]pyridin-4-yl, imidazo[1,2-a]pyridin-7-yl, imidazo[1,2-a]pyrimidin-6-yl, imidazo[1,2-b]pyridazin-6-yl, imidazo[1,2-a]pyrazin-3-yl, imidazo[1,2-a]pyrazin-6-ylimidazo[1,5-a]pyridin-7-yl, [1,2,3]triazolo[1,5-a]pyridin-5-yl, [1,2,3]triazolo[1,5-a]pyridin-7-yl, 1H-[1,2,3]triazolo[4,5-b]pyridin-5-yl, 1H-[1,2,3]triazolo[4,5-b]pyridin-6-yl, 3H-[1,2,3]triazolo[4,5-c]pyridin-6-yl, and [1,2,4]triazolo[4,3-b]pyridazin-6-yl, optionally substituted with one, two, three or four R8substituents. One aspect includes a compound of Formula (I) wherein R7is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and C3-10cycloalkyl. One aspect includes a compound of Formula (I) wherein R8is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, C1-6alkoxy-C1-6alkyl, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, amino-C1-6alkyl, and C3-10cycloalkyl. One aspect includes a compound of Formula (I) wherein R8is, in each instance, independently selected from cyano, halogen, hydroxy, C1-6alkyl, deutero-C1-4alkyl, halo-C1-6alkyl, C1-6alkoxy, halo-C1-6alkoxy, amino, C1-6alkyl-amino, (C1-6alkyl)2-amino, and C3-10cycloalkyl. Another aspect includes a compound of Formula (I) wherein R8is cyano. Another aspect includes a compound of Formula (I), wherein R8is halogen selected from bromo, chloro, fluoro, and iodo. Another aspect includes a compound of Formula (I), wherein R8is halogen selected from bromo, chloro, and fluoro. Another aspect includes a compound of Formula (I) wherein R8is hydroxy. Another aspect includes a compound of Formula (I), wherein R8is C1-6alkyl selected from methyl, ethyl, propyl, isopropyl, and tert-butyl. Another aspect includes a compound of Formula (I), wherein R8is C1-6alkyl selected from methyl, ethyl, and propyl. Another aspect includes a compound of Formula (I) wherein R8is deutero-C1-4alkyl wherein C1-4alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, and tert-butyl partially or completely substituted with one or more deuterium atoms where allowed by available valences. Another aspect includes a compound of Formula (I) wherein R8is (2H3)methyl. Another aspect includes a compound of Formula (I), wherein R8is halo-C1-6alkyl, wherein C1-6alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, and tert-butyl partially or completely substituted with one or more halogens selected from bromo, chloro, fluoro, and iodo where allowed by available valences. Another aspect includes a compound of Formula (I), wherein R8is halo-C1-6alkyl selected from trifluromethyl and difluoromethyl. Another aspect includes a compound of Formula (I), wherein R8is C1-6alkoxy selected from methoxy, ethoxy, propoxy, isopropoxy, and tert-butoxy. Another aspect includes a compound of Formula (I), wherein R8is C1-6alkoxy selected from methoxy and ethoxy. Another aspect includes a compound of Formula (I), wherein R8is halo-C1-6alkoxy,wherein C1-6alkoxy is selected from methoxy, ethoxy, propoxy, isopropoxy, and tert-butoxy partially or completely substituted with one or more halogens selected from bromo, chloro, fluoro, and iodo where allowed by available valences. Another aspect includes a compound of Formula (I), wherein R8is difluoromethoxy. Another aspect includes a compound of Formula (I) wherein R8is amino. Another aspect includes a compound of Formula (I), wherein R8is C1-6alkyl-amino wherein C1-6alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, and tert-butyl. Another aspect includes a compound of Formula (I), wherein R8is methylamino. Another aspect includes a compound of Formula (I), wherein R8is (C1-6alkyl)2-amino wherein C1-6alkyl is independently selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, and tert-butyl. Another aspect includes a compound of Formula (I), wherein R8is dimethylamino. Another aspect includes a compound of Formula (I), wherein R8is C3-10cycloalkyl selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cyclohexyl. Another aspect includes a compound of Formula (I), wherein R8is cyclopropyl. One aspect of the compound of Formula (I) includes a compound selected from Formula (Ia), Formula (Ib), Formula (Id), Formula (e), Formula (If), Formula (Ig), Formula (Ih), Formula (Ii), Formula (Ij), Formula (Ik), Formula (Il), Formula (Im), Formula (In), Formula (o), Formula (Ip), Formula (Iq), Formula (Is), Formula (It), Formula (Iu), Formula (Iv), Formula (Iw), Formula (Ix), Formula (Iy), Formula (Iz), Formula (Iaa), Formula (Ibb), Formula (Icc), Formula (Idd), Formula (ee), Formula (Iff), Formula (Igg), Formula (Ihh), Formula (Iii), Formula (Ijj), Formula (Ikk), Formula (Ill), Formula (Imm), Formula (Inn), Formula (oo), Formula (Ipp), Formula (Iqq), Formula (Irr), Formula (Iss), Formula (Itt), Formula (Iuu), Formula (Iww), Formula (Ixx), Formula (Iyy), Formula (Izz), Formula (Iaaa), Formula (Ibbb), Formula (Iccc), Formula (Iddd), Formula (Ieee), Formula (Ifff), Formula (Iggg), or Formula (Ihhh): or a form thereof. Another aspect of the compound of Formula (I) includes a compound selected from Formula (Ie), Formula (i), Formula (Ik), Formula (Ip), Formula (Iq), Formula (It), Formula (Iu), Formula (Ix), Formula (Iz), Formula (Ibb), Formula (Icc), or Formula (Idd): or a form thereof. Another aspect of the compound of Formula (I) includes a compound selected from Formula (Ia1), Formula (I1), Formula (Id1), Formula (Ie1), Formula (If1), Formula (Ig1), Formula (Ih1), Formula (Ii1), Formula (I1j), Formula (Ik1), Formula (Il1), Formula (Im1), Formula (In1), Formula (Io1), Formula (Ip1), Formula (Iq1), Formula (Is1), Formula (It1), Formula (Iu1), Formula (Iv1), Formula (Iw 1), Formula (Ix1), Formula (Iy1), Formula (Iz1), Formula (Iaa1), Formula (Ibb1), Formula (Icc1), Formula (Idd1), Formula (Iee1), Formula (Iff1), Formula (Igg1), Formula (Ihh1), Formula (Iii1), Formula (Ijj1), Formula (Ikk1), Formula (Ill1), Formula (Imm1), Formula (Inn1), Formula (Ioo1), Formula (Ipp1), Formula (Iqq1), Formula (Irr1), Formula (Iss1), Formula (Itt1), Formula (Iuu1), Formula (Iww1), Formula (Ixx1), Formula (Iyy1), Formula (Izz1), Formula (Iaaa1), Formula (Ibbb1), Formula (Iccc1), Formula (Iddd1), Formula (Ieee1), Formula (Ifff1), Formula (Iggg1), or Formula (Ihhh1): or a form thereof. Another aspect of the compound of Formula (I) includes a compound selected from Formula (Ie1), Formula (Ii1), Formula (Ik1), Formula (Ip1), Formula (Iq1), Formula (It1), Formula (Iu1), Formula (Ix1), Formula (Iz1), Formula (Ibb1), Formula (Icc1), or Formula (Idd1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Ie1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Ii1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Ik1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Ip1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Iq1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (It1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Is1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Ix1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Iz1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Ibb1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Icc1): or a form thereof. Another aspect of the compound of Formula (I) includes the compound of Formula (Idd1): or a form thereof. An aspect of the compound of Formula (I) or a form thereof includes a compound selected from the group consisting of: wherein the form of the compound is selected from the group consisting of a salt, hydrate, solvate, racemate, enantiomer, diastereomer, stereoisomer, and tautomer form thereof. An aspect the compound of Formula (I) or a form thereof (wherein compound number (#1) indicates that the salt form was isolated) includes a compound selected from the group consisting of: CpdName115-(1H-pyrazol-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazin-5-yl]phenol215-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazine315-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]phenol413-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine512-[6-methyl-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol613-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-6-methyl-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine715-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol815-(1H-pyrazol-4-yl)-2-[5-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)-5H-pyrrolo[2,3-b]pyrazin-2-yl]phenol913-[2-hydroxy-4-(1H-pyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-6-ol1015-(1H-pyrazol-4-yl)-2-[5-(2,2,6,6-tetramethylpiperidin-4-yl)-5H-pyrrolo[2,3-b]pyrazin-2-yl]phenol1115-(1H-pyrazol-4-yl)-2-[7-(1,2,3,6-tetrahydropyridin-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]phenol1212-[7-(piperidin-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol1316-[2,3-difluoro-4-(1H-pyrazol-4-yl)phenyl]-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine1416-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine1515-(1H-pyrazol-4-yl)-2-[7-(1,2,3,6-tetrahydropyridin-4-yl)thieno[3,2-c]pyridazin-3-yl]phenol1612-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-5-(2,2,6,6-tetramethylpiperidin-4-yl)-5H-pyrrolo[2,3-b]pyrazine1715-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)thieno[3,2-c]pyridazin-3-yl]phenol1812-[7-(8-azabicyclo[3.2.1]oct-2-en-3-yl)thieno[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol1912-[1-(piperidin-4-yl)-1H-pyrazolo[3,4-b]pyrazin-5-yl]-5-(1H-pyrazol-4-yl)phenol2015-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]phenol2112-[1-(piperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]-5-(1H-pyrazol-4-yl)phenol2215-(1H-pyrazol-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol2315-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazin-3-yl]phenol2415-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)thieno[3,2-c]pyridazin-3-yl]phenol2512-[7-(3-oxa-9-azabicyclo[3.3.1]non-6-en-7-yl)thieno[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol2615-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazin-3-yl]phenol2712-[7-(8-azabicyclo[3.2.1]oct-2-en-3-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol2812-[7-(3-oxa-9-azabicyclo[3.3.1]non-6-en-7-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol2912-[7-(8-azabicyclo[3.2.1]oct-3-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol3012-[7-(3-oxa-9-azabicyclo[3.3.1]non-7-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol3115-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]phenol3213-[2,3-difluoro-4-(1H-pyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine3314-fluoro-2-(1H-pyrazol-4-yl)-5-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol345-(1H-pyrazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol3514-fluoro-5-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol3617-[(3-exo)-8-azabicyclo[3.2.1]oct-3-yl]-3-[2,3-difluoro-4-(1H-pyrazol-4-yl)phenyl]-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine374-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1-methylpyridin-2(1H)-one3814-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]biphenyl-3,4′-diol3915-(1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol4012-[6-methoxy-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol4212-[6-(methylamino)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol432-[7-(piperazin-1-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol465-(1-ethyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol475-(1-propyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol485-(1H-pyrazol-3-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol4912-[6-(ethylamino)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol505-(1-methyl-1H-pyrazol-5-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol515-(1-methyl-1H-pyrazol-3-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol5212-[3-(1,2,2,6,6-pentamethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-pyrazol-4-yl)phenol5316-fluoro-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl][1,1′-biphenyl]-3,4′-diol5412-fluoro-3-(1H-pyrazol-4-yl)-6-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol5514-{2-fluoro-5-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1-methylpyridin-2(1H)-one5612-[3-(2,2-dimethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-pyrazol-4-yl)phenol5712-{3-[(1R,5S)-1,5-dimethyl-8-azabicyclo[3.2.1]octan-3-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol5812-(1H-pyrazol-4-yl)-5-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]benzene-1,4-diol5913-fluoro-5-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol6015-(pyrazin-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol6115-(pyridin-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol6214-fluoro-5-(1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol6312-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-1,2,4-triazol-1-yl)phenol6412-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(4H-1,2,4-triazol-4-yl)phenol6515-(pyridin-3-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol6615-(pyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol6716-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyridin-3-ol6812-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyrimidin-5-ol6915-[1-(2H3)methyl-1H-pyrazol-4-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol7015-(1H-imidazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol7115-[1-(difluoromethyl)-1H-pyrazol-4-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol7212-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-1,2,3-triazol-1-yl)phenol7315-(2-methylpyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol7412-[3-(2,2,6,6,-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-[2-(trifluoromethyl)pyridin-4-yl]phenol7515-(pyrimidin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol7615-(pyridazin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol7715-(2-methoxypyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol785-(pyrimidin-5-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol7916-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyridazin-3-ol805-(1H-pyrrol-3-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol8116-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]quinolin-7-ol82(3E)-3-(hydroxyimino)-6-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-2,3-dihydro-1H-inden-5-ol8314-chloro-5-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol845-[6-(dimethylamino)pyridin-3-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol855-(imidazo[1,2-a]pyrazin-3-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol861-cyclopropyl-4-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyridin-2(1H)-one8714-fluoro-5-(pyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol885-(imidazo[1,5-a]pyridin-7-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol8912-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(thiophen-3-yl)phenol905-(imidazo[1,2-a]pyridin-7-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol915-(1H-imidazol-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol921-methyl-5-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-1H-benzimidazol-6-ol9314-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyridin-2(1H)-one945-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-1H-indazol-6-ol9515-(furan-3-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol9612-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1,3-thiazol-2-yl)phenol972-methyl-5-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-1H-benzimidazol-6-ol9815-(1-methyl-1H-pyrazol-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol995-(2-aminopyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol1005-[2-(dimethylamino)pyridin-4-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol1015-(3-fluoropyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol10215-[1-(2H3)methyl-1H-pyrazol-4-yl]-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol10315-[5-(difluoromethoxy)pyridin-2-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol1045-[2-(methylamino)pyridin-4-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol10515-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)furo[3,2-c]pyridazin-3-yl]phenol10615-(3-fluoro-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol10712-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1,3-thiazol-5-yl)phenol10815-(3-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol10914-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1H-pyrazole-3-carbonitrile11012-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1,3-thiazole-5-carbonitrile11115-(1,3-oxazol-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol1122-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-1,2,3-triazol-4-yl)phenol1135-(6-methoxypyrimidin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol11415-[2-(difluoromethoxy)pyridin-4-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol1155-(1H-imidazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol11612-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1,3,4-thiadiazol-2-yl)phenol11716-[4-(1H-pyrazol-4-yl)-1H-benzotriazol-7-yl]-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine11815-(1H-pyrrolo[2,3-b]pyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol11915-(2-methoxypyrimidin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol12015-(1,2-oxazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol12115-(5-fluoro-1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol12215-(1-ethyl-5-fluoro-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol12315-(2-ethoxypyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol12415-(6-ethoxypyrimidin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol12512-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-([1,2,3]triazolo[1,5-a]pyridin-5-yl)phenol12612-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-([1,2,4]triazolo[1,5-a]pyridin-7-yl)phenol12715-(3-chloro-1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol12816-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyrimidin-4(3H)-one12915-(3-chloro-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol13015-(3-fluoro-1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol13115-(3-methoxy-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol13214-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1-methyl-1H-pyrazole-3-carbonitrile13315-(5-methyl-1,3-thiazol-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol13412-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1,2,4-thiadiazol-5-yl)phenol13515-(4-fluoro-1H-benzotriazol-6-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol13615-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazin-3-yl]pyridin-3-ol13715-(3-bromo-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol13815-(1-methyl-1H-1,2,3-triazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol13912-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-[3-(trifluoromethyl)-1H-pyrazol-4-yl]phenol14015-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]pyridin-3-ol14115-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]pyridin-3-ol14215-(imidazo[1,2-a]pyrazin-6-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol14315-(4-fluoro-1H-imidazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol14415-(4-methyl-1H-imidazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol14512-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-[1,2,3]triazolo[4,5-b]pyridin-6-yl)phenol14612-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(3H-[1,2,3]triazolo[4,5-c]pyridin-6-yl)phenol14712-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-[1,2,3]triazolo[4,5-b]pyridin-5-yl)phenol1485-(3-fluoro-1-methyl-1H-pyrazol-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-y])-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol14915-(2,4-dimethyl-1H-imidazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol15015-(2-methyl-1,3-thiazol-5-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol15115-(2-methyl-2H-1,2,3-triazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol15212-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-([1,2,4]triazolo[4,3-b]pyridazin-6-yl)phenol15315-(3-methyl-1,2,4-thiadiazol-5-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol1545-(4-fluoro-2-methyl-1,3-thiazol-5-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol15515-(5-methyl-1H-pyrazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol1565-(4-methyl-1H-pyrazol-1-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol15715-(3-methyl-1H-pyrazol-1-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol1585-(2-methyl-1,3-oxazol-5-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol15915-(4-methoxy-1,3,5-triazin-2-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol16015-(imidazo[1,2-a]pyrimidin-6-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol16115-(3-fluoro-1H-pyrazol-4-yl)-2-(1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl)phenol16215-(imidazo[1,2-b]pyridazin-6-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol16312-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(2H-1,2,3-triazol-2-yl)phenol16412-{3-[(3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol16515-(pyridin-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol16615-(pyridin-3-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol16715-(pyrimidin-5-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol16812-{3-[(3S,4R)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol16915-(1-methyl-1H-pyrazol-3-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol17012-{3-[3-(tert-butylamino)cyclobutyl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol17114-(4-{3-[(3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-3-hydroxyphenyl)-1-methylpyridin-2(1H)-one17216-(4-{3-[(3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-3-hydroxyphenyl)-3-methylpyrimidin-4(3H)-one1735-(3-fluoro-1H-pyrazol-4-yl)-2-{3-[(3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}phenol17412-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]-5-(2H-1,2,3-triazol-2-yl)phenol17512-{3-[3-(tert-butylamino)cyclopentyl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol, and1762-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazin-3-yl]-5-(2H-1,2,3-triazol-2-yl)phenol; wherein the form of the compound is selected from the group consisting of a salt, hydrate, solvate, racemate, enantiomer, diastereomer, stereoisomer, and tautomer form thereof. Another aspect of the compound of Formula (I) or a form thereof is a compound salt selected from the group consisting of: CpdName15-(1H-pyrazol-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazin-5-yl]phenol hydrochloride25-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazine hydrochloride35-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]phenol hydrochloride43-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine hydrochloride52-[6-methyl-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride63-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-6-methyl-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine hydrochloride75-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride85-(1H-pyrazol-4-yl)-2-[5-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)-5H-pyrrolo[2,3-b]pyrazin-2-yl]phenol hydrochloride93-[2-hydroxy-4-(1H-pyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-6-ol hydrochloride105-(1H-pyrazol-4-yl)-2-[5-(2,2,6,6-tetramethylpiperidin-4-yl)-5H-pyrrolo[2,3-b]pyrazin-2-yl]phenol hydrochloride115-(1H-pyrazol-4-yl)-2-[7-(1,2,3,6-tetrahydropyridin-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]phenol hydrochloride122-[7-(piperidin-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenolhydrochloride136-[2,3-difluoro-4-(1H-pyrazol-4-yl)phenyl]-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine hydrochloride146-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine hydrochloride155-(1H-pyrazol-4-yl)-2-[7-(1,2,3,6-tetrahydropyridin-4-yl)thieno[3,2-c]pyridazin-3-yl]phenol hydrochloride162-[2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl]-5-(2,2,6,6-tetramethylpiperidin-4-yl)-5H-pyrrolo[2,3-b]pyrazine hydrochloride175-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)thieno[3,2-c]pyridazin-3-yl]phenol hydrochloride182-[7-(8-azabicyclo[3.2.1]oct-2-en-3-yl)thieno[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride192-[1-(piperidin-4-yl)-1H-pyrazolo[3,4-b]pyrazin-5-yl]-5-(1H-pyrazol-4-yl)phenolhydrochloride205-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]phenol hydrochloride212-[1-(piperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]-5-(1H-pyrazol-4-yl)phenolhydrochloride225-(1H-pyrazol-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol hydrochloride235-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazin-3-yl]phenol hydrochloride245-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)thieno[3,2-c]pyridazin-3-yl]phenol hydrochloride252-[7-(3-oxa-9-azabicyclo[3.3.1]non-6-en-7-yl)thieno[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride265-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazin-3-yl]phenol hydrochloride272-[7-(8-azabicyclo[3.2.1]oct-2-en-3-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride282-[7-(3-oxa-9-azabicyclo[3.3.1]non-6-en-7-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride292-[7-(8-azabicyclo[3.2.1]oct-3-y])-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride302-[7-(3-oxa-9-azabicyclo[3.3.1]non-7-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride315-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]phenol hydrochloride323-[2,3-difluoro-4-(1H-pyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine hydrochloride334-fluoro-2-(1H-pyrazol-4-yl)-5-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride354-fluoro-5-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrobromide367-[(3-exo)-8-azabicyclo[3.2.1]oct-3-yl]-3-[2,3-difluoro-4-(1H-pyrazol-4-yl)phenyl]-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine hydrochloride384-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]biphenyl-3,4′-diol hydrobromide395-(1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrobromide402-[6-methoxy-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride422-[6-(methylamino)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride492-[6-(ethylamino)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride522-[3-(1,2,2,6,6-pentamethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-pyrazol-4-yl)phenol dihydrochloride536-fluoro-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl][1,1′-biphenyl]-3,4′-diol hydrobromide542-fluoro-3-(1H-pyrazol-4-yl)-6-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride554-{2-fluoro-5-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1-methylpyridin-2(1H)-one hydrochloride562-[3-(2,2-dimethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride572-{3-[(1R,5S)-1,5-dimethyl-8-azabicyclo[3.2.1]octan-3-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol hydrochloride582-(1H-pyrazol-4-yl)-5-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]benzene-1,4-diol dihydrochloride593-fluoro-5-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride605-(pyrazin-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride615-(pyridin-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride624-fluoro-5-(1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride632-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-1,2,4-triazol-1-yl)phenol hydrochloride642-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(4H-1,2,4-triazol-4-yl)phenol hydrochloride655-(pyridin-3-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride665-(pyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride676-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyridin-3-ol dihydrochloride682-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyrimidin-5-ol dihydrochloride695-[1-(2H3)methyl-1H-pyrazol-4-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride705-(1H-imidazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride715-[1-(difluoromethyl)-1H-pyrazol-4-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride722-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-1,2,3-triazol-1-yl)phenol hydrochloride735-(2-methylpyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride742-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-[2-(trifluoromethyl)pyridin-4-yl]phenol dihydrochloride755-(pyrimidin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride765-(pyridazin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride775-(2-methoxypyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride796-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyridazin-3-ol hydrochloride816-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]quinolin-7-ol hydrobromide834-chloro-5-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrobromide874-fluoro-5-(pyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrobromide892-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(thiophen-3-yl)phenol hydrochloride934-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyridin-2(1H)-one hydrochloride955-(furan-3-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride962-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1,3-thiazol-2-yl)phenol hydrochloride985-(1-methyl-1H-pyrazol-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol dihydrochloride1025-[1-(2H3)methyl-1H-pyrazol-4-yl]-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol dihydrochloride1035-[5-(difluoromethoxy)pyridin-2-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol formate1055-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)furo[3,2-c]pyridazin-3-yl]phenol hydrochloride1065-(3-fluoro-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1072-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1,3-thiazol-5-yl)phenol hydrochloride1085-(3-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1094-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1H-pyrazole-3-carbonitrile hydrochloride1102-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1,3-thiazole-5-carbonitrile hydrochloride1115-(1,3-oxazol-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol formate1145-[2-(difluoromethoxy)pyridin-4-yl]-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1162-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1,3,4-thiadiazol-2-yl)phenol hydrochloride1176-[4-(1H-pyrazol-4-yl)-1H-benzotriazol-7-yl]-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine trifluoroacetate1185-(1H-pyrrolo[2,3-b]pyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1195-(2-methoxypyrimidin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1205-(1,2-oxazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1215-(5-fluoro-1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1225-(1-ethyl-5-fluoro-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1235-(2-ethoxypyridin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1245-(6-ethoxypyrimidin-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1252-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-([1,2,3]triazolo[1,5-a]pyridin-5-yl)phenol hydrochloride1262-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-([1,2,4]triazolo[1,5-a]pyridin-7-yl)phenol hydrochloride1275-(3-chloro-1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1286-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}pyrimidin-4(3H)-one hydrochloride1295-(3-chloro-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1305-(3-fluoro-1-methyl-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1315-(3-methoxy-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochlorid1324-{3-hydroxy-4-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenyl}-1-methyl-1H-pyrazole-3-carbonitrile hydrochloride1335-(5-methyl-1,3-thiazol-2-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-y])-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1342-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1,2,4-thiadiazol-5-yl)phenol hydrochloride1355-(4-fluoro-1H-benzotriazol-6-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1365-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazin-3-yl]pyridin-3-ol dihydrochloride1375-(3-bromo-1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1385-(1-methyl-1H-1,2,3-triazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1392-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-[3-(trifluoromethyl)-1H-pyrazol-4-yl]phenol hydrochloride1405-(1H-pyrazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]pyridin-3-ol hydrochloride1415-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl]pyridin-3-ol dihydrochloride1425-(imidazo[1,2-a]pyrazin-6-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1435-(4-fluoro-1H-imidazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride1445-(4-methyl-1H-imidazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride1452-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-[1,2,3]triazolo[4,5-b]pyridin-6-yl)phenol dihydrochloride1462-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(3H-[1,2,3]triazolo[4,5-c]pyridin-6-yl)phenol dihydrochloride1472-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(1H-[1,2,3]triazolo[4,5-b]pyridin-5-yl)phenol dihydrochloride1495-(2,4-dimethyl-1H-imidazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride1505-(2-methyl-1,3-thiazol-5-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride1515-(2-methyl-2H-1,2,3-triazol-4-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1522-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-([1,2,4]triazolo[4,3-b]pyridazin-6-yl)phenol hydrochloride1535-(3-methyl-1,2,4-thiadiazol-5-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol hydrochloride1555-(5-methyl-1H-pyrazol-1-yl)-2-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride1575-(3-methyl-1H-pyrazol-1-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol dihydrochloride1595-(4-methoxy-1,3,5-triazin-2-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol dihydrochloride1605-(imidazo[1,2-a]pyrimidin-6-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol hydrochloride1615-(3-fluoro-1H-pyrazol-4-yl)-2-(1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl)phenol hydrochloride1625-(imidazo[1,2-b]pyridazin-6-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol hydrochloride1632-[3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl]-5-(2H-1,2,3-triazol-2-yl)phenol hydrochloride1642-{3-[(3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol dihydrochloride1655-(pyridin-4-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol hydrochloride1665-(pyridin-3-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol hydrochloride1675-(pyrimidin-5-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol hydrochloride1682-{3-[(3S,4R)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol dihydrochloride1695-(1-methyl-1H-pyrazol-3-yl)-2-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]phenol hydrochloride1702-{3-[3-(tert-butylamino)cyclobutyl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-5-(1H-pyrazol-4-yl)phenol dihydrochloride1714-(4-{3-[(3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-3-hydroxyphenyl)-1-methylpyridin-2(1H)-one hydrochloride1726-(4-{3-[(35,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl}-3-hydroxyphenyl)-3-methylpyrimidin-4(3H)-one dihydrochloride1742-[1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl]-5-(2H-1,2,3-triazol-2-yl)phenol hydrochloride, and1752-{3-[3-(tert-butylamino)cyclopentyl]-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-y]}-5-(1H-pyrazol-4-yl)phenol dihydrochloride; wherein the form of the compound salt is selected from the group consisting of a hydrate, solvate, racemate, enantiomer, diastereomer, stereoisomer, and tautomer form thereof. An aspect of the present description includes a method of use of a compound of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound of Formula (I) or a form thereof to the subject. Another aspect of the present description includes a method of use of the compound salt of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound salt of Formula (I) or a form thereof to the subject. An aspect of the present description includes a use of the compound of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound of Formula (I) or a form thereof to the subject. Another aspect of the present description includes a use of the compound salt of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound salt of Formula (I) or a form thereof to the subject. Chemical Definitions The chemical terms used above and throughout the description herein, unless specifically defined otherwise, shall be understood by one of ordinary skill in the art to have the following indicated meanings. As used herein, the term “C1-6alkyl” generally refers to saturated hydrocarbon radicals having from one to eight carbon atoms in a straight or branched chain configuration, including, but not limited to, methyl, ethyl, n-propyl (also referred to as propyl or propanyl), isopropyl, n-butyl (also referred to as butyl or butanyl), isobutyl, sec-butyl, tert-butyl, n-pentyl (also referred to as pentyl or pentanyl), n-hexyl (also referred to as hexyl or hexanyl), and the like. In certain aspects, C1-6alkyl includes, but is not limited to C1-4alkyl and the like. A C1-6alkyl radical is optionally substituted with substituent species as described herein where allowed by available valences. As used herein, the term “C2-8alkenyl” generally refers to partially unsaturated hydrocarbon radicals having from two to eight carbon atoms in a straight or branched chain configuration and one or more carbon-carbon double bonds therein, including, but not limited to, ethenyl (also referred to as vinyl), allyl, propenyl and the like. In certain aspects, C2-8alkenyl includes, but is not limited to, C2-6alkenyl, C2-4alkenyl and the like. A C2-8alkenyl radical is optionally substituted with substituent species as described herein where allowed by available valences. As used herein, the term “C2-8alkynyl” generally refers to partially unsaturated hydrocarbon radicals having from two to eight carbon atoms in a straight or branched chain configuration and one or more carbon-carbon triple bonds therein, including, but not limited to, ethynyl, propynyl, butynyl and the like. In certain aspects, C2-8alkynyl includes, but is not limited to, C2-6alkynyl, C2-4alkynyl and the like. A C2-8alkynyl radical is optionally substituted with substituent species as described herein where allowed by available valences. As used herein, the term “C1-6alkoxy” generally refers to saturated hydrocarbon radicals having from one to eight carbon atoms in a straight or branched chain configuration of the formula: —O—C1-6alkyl, including, but not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, n-pentoxy, n-hexoxy and the like. In certain aspects, C1-6alkoxy includes, but is not limited to C1-4alkoxy and the like. A C1-6alkoxy radical is optionally substituted with substituent species as described herein where allowed by available valences. As used herein, the term “C3-10cycloalkyl” generally refers to a saturated or partially unsaturated monocyclic, bicyclic or polycyclic hydrocarbon radical, including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, 1H-indanyl, indenyl, tetrahydro-naphthalenyl and the like. In certain aspects, C3-10cycloalkyl includes, but is not limited to C3-8cycloalkyl, C5-8cycloalkyl, C3-10cycloalkyl and the like. A C3-10cycloalkyl radical is optionally substituted with substituent species as described herein where allowed by available valences. As used herein, the term “aryl” generally refers to a monocyclic, bicyclic or polycyclic aromatic carbon atom ring structure radical, including, but not limited to, phenyl, naphthyl, anthracenyl, fluorenyl, azulenyl, phenanthrenyl and the like. An aryl radical is optionally substituted with substituent species as described herein where allowed by available valences. As used herein, the term “heteroaryl” generally refers to a monocyclic, bicyclic or polycyclic aromatic carbon atom ring structure radical in which one or more carbon atom ring members have been replaced, where allowed by structural stability, with one or more heteroatoms, such as an O, S or N atom, including, but not limited to, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, isoxazolyl, isothiazolyl, oxazolyl, 1,3-thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, indolyl, indazolyl, indolizinyl, isoindolyl, benzofuranyl, benzothienyl, benzoimidazolyl, 1,3-benzothiazolyl, 1,3-benzoxazolyl, purinyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, 1,3-diazinyl, 1,2-diazinyl, 1,2-diazolyl, 1,4-diazanaphthalenyl, acridinyl, furo[3,2-b]pyridinyl, furo[3,2-c]pyridinyl, furo[2,3-c]pyridinyl, 6H-thieno[2,3-b]pyrrolyl, thieno[3,2-c]pyridinyl, thieno[2,3-d]pyrimidinyl, 1H-pyrrolo[2,3-b]pyridinyl, 1H-pyrrolo[2,3-c]pyridinyl, 1H-pyrrolo[3,2-b]pyridinyl, pyrrolo[1,2-a]pyrazinyl, pyrrolo[1,2-b]pyridazinyl, pyrazolo[1,5-a]pyridinyl, pyrazolo[1,5-a]pyrazinyl, imidazo[1,2-a]pyridinyl, 3H-imidazo[4,5-b]pyridinyl, imidazo[1,2-a]pyrimidinyl, imidazo[1,2-c]pyrimidinyl, imidazo[1,2-b]pyridazinyl, imidazo[1,2-a]pyrazinyl, imidazo[2,1-b][1,3]thiazolyl, imidazo[2,1-b][1,3,4]thiadiazolyl, [1,2,4]triazolo[1,5-a]pyridinyl, [1,2,4]triazolo[4,3-a]pyridinyl and the like. A heteroaryl radical is optionally substituted on a carbon or nitrogen atom ring member with substituent species as described herein where allowed by available valences. In certain aspects, the nomenclature for a heteroaryl radical may differ, such as in non-limiting examples where furanyl may also be referred to as furyl, thienyl may also be referred to as thiophenyl, pyridinyl may also be referred to as pyridyl, benzothienyl may also be referred to as benzothiophenyl and 1,3-benzoxazolyl may also be referred to as 1,3-benzooxazolyl. In certain other aspects, the term for a heteroaryl radical may also include other regioisomers, such as in non-limiting examples where the term pyrrolyl may also include 2H-pyrrolyl, 3H-pyrrolyl and the like, the term pyrazolyl may also include 1H-pyrazolyl and the like, the term imidazolyl may also include 1H-imidazolyl and the like, the term triazolyl may also include 1H-1,2,3-triazolyl and the like, the term oxadiazolyl may also include 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl and the like, the term tetrazolyl may also include 1H-tetrazolyl, 2H-tetrazolyl and the like, the term indolyl may also include 1H-indolyl and the like, the term indazolyl may also include 1H-indazolyl, 2H-indazolyl and the like, the term benzoimidazolyl may also include 1H-benzoimidazolyl and the term purinyl may also include 9H-purinyl and the like. As used herein, the term “heterocyclyl” generally refers to a saturated or partially unsaturated monocyclic, bicyclic or polycyclic carbon atom ring structure radical in which one or more carbon atom ring members have been replaced, where allowed by structural stability, with a heteroatom, such as an O, S or N atom, including, but not limited to, oxiranyl, oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolinyl, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, isoxazolinyl, isoxazolidinyl, isothiazolinyl, isothiazolidinyl, oxazolinyl, oxazolidinyl, thiazolinyl, thiazolidinyl, triazolinyl, triazolidinyl, oxadiazolinyl, oxadiazolidinyl, thiadiazolinyl, thiadiazolidinyl, tetrazolinyl, tetrazolidinyl, pyranyl, dihydro-2H-pyranyl, thiopyranyl, 1,3-dioxanyl, 1,2,5,6-tetrahydropyridinyl, 1,2,3,6-tetrahydropyridinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, 1,4-diazepanyl, 1,3-benzodioxolyl, 1,4-benzodioxanyl, 2,3-dihydro-1,4-benzodioxinyl, hexahydropyrrolo[3,4-b]pyrrol-(1H)-yl, (3aS,6aS)-hexahydropyrrolo[3,4-b]pyrrol-(1H)-yl, (3aR,6aR)-hexahydropyrrolo[3,4-b]pyrrol-(1H)-yl, hexahydropyrrolo[3,4-b]pyrrol-(2H)-yl, (3aS,6aS)-hexahydropyrrolo[3,4-b]pyrrol-(2H)-yl, (3aR,6aR)-hexahydropyrrolo[3,4-b]pyrrol-(2H)-yl, hexahydropyrrolo[3,4-c]pyrrol-(1H)-yl, (3aR,6aS)-hexahydropyrrolo[3,4-c]pyrrol-(1H)-yl, (3aR,6aR)-hexahydropyrrolo[3,4-c]pyrrol-(1H)-yl, octahydro-5H-pyrrolo[3,2-c]pyridinyl, octahydro-6H-pyrrolo[3,4-b]pyridinyl, (4aR,7aR)-octahydro-6H-pyrrolo[3,4-b]pyridinyl, (4aS,7aS)-octahydro-6H-pyrrolo[3,4-b]pyridinyl, hexahydropyrrolo[1,2-a]pyrazin-(1H)-yl, (7R,8aS)-hexahydropyrrolo[1,2-a]pyrazin-(1H)-yl, (8aS)-hexahydropyrrolo[1,2-a]pyrazin-(1H)-yl, (8aR)-hexahydropyrrolo[1,2-a]pyrazin-(1H)-yl, (8aS)-octahydropyrrolo[1,2-a]pyrazin-(1H)-yl, (8aR)-octahydropyrrolo[1,2-a]pyrazin-(1H)-yl, hexahydropyrrolo[1,2-a]pyrazin-(2H)-one, octahydro-2H-pyrido[1,2-a]pyrazinyl, 3-azabicyclo[3.1.0]hexyl, (1R,5S)-3-azabicyclo[3.1.0]hexyl, 8-azabicyclo[3.2.1]octyl, (1R,5S)-8-azabicyclo[3.2.1]octyl, 8-azabicyclo[3.2.1]oct-2-enyl, (1R,5S)-8-azabicyclo[3.2.1]oct-2-enyl, 9-azabicyclo[3.3.1]nonyl, (1R,5S)-9-azabicyclo[3.3.1]nonyl, 2,5-diazabicyclo[2.2.1]heptyl, (1S,4S)-2,5-diazabicyclo[2.2.1]heptyl, 2,5-diazabicyclo[2.2.2]octyl, 3,8-diazabicyclo[3.2.1]octyl, (1R,5S)-3,8-diazabicyclo[3.2.1]octyl, 1,4-diazabicyclo[3.2.2]nonyl, azaspiro[3.3]heptyl, 2,6-diazaspiro[3.3]heptyl, 2,7-diazaspiro[3.5]nonyl, 5,8-diazaspiro[3.5]nonyl, 2,7-diazaspiro[4.4]nonyl, 6,9-diazaspiro[4.5]decyl and the like. A heterocyclyl radical is optionally substituted on a carbon or nitrogen atom ring member with substituent species as described herein where allowed by available valences. In certain aspects, the nomenclature for a heterocyclyl radical may differ, such as in non-limiting examples where 1,3-benzodioxolyl may also be referred to as benzo[d][1,3]dioxolyl and 2,3-dihydro-1,4-benzodioxinyl may also be referred to as 2,3-dihydrobenzo[b][1,4]dioxinyl. As used herein, the term “deutero-C1-4alkyl,” refers to a radical of the formula: —C1-4alkyl-deutero, wherein C1-4alkyl is partially or completely substituted with one or more deuterium atoms where allowed by available valences. As used herein, the term “C1-6alkoxy-C1-6alkyl” refers to a radical of the formula: —C1-6alkyl-O—C1-6alkyl. As used herein, the term “C1-6alkyl-amino” refers to a radical of the formula: —NH—C1-6alkyl. As used herein, the term “(C1-6alkyl)2-amino” refers to a radical of the formula: —N(C1-6alkyl)2. As used herein, the term “C1-6alkyl-thio” refers to a radical of the formula: —S—C1-6alkyl. As used herein, the term “amino-C1-6alkyl” refers to a radical of the formula: —C1-6alkyl-NH2. As used herein, the term “halo” or “halogen” generally refers to a halogen atom radical, including fluoro, chloro, bromo and iodo. As used herein, the term “halo-C1-6alkoxy” refers to a radical of the formula: —O—C1-6alkyl-halo, wherein C1-6alkyl is partially or completely substituted with one or more halogen atoms where allowed by available valences. As used herein, the term “halo-C1-6alkyl” refers to a radical of the formula: —C1-6alkyl-halo, wherein C1-6alkyl is partially or completely substituted with one or more halogen atoms where allowed by available valences. As used herein, the term “hydroxy” refers to a radical of the formula: —OH. As used herein, the term “hydroxy-C1-6alkyl” refers to a radical of the formula: —C1-6alkyl-OH, wherein C1-6alkyl is partially or completely substituted with one or more hydroxy radicals where allowed by available valences. As used herein, the term “substituent” means positional variables on the atoms of a core molecule that are substituted at a designated atom position, replacing one or more hydrogens on the designated atom, provided that the designated atom's normal valency is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. A person of ordinary skill in the art should note that any carbon as well as heteroatom with valences that appear to be unsatisfied as described or shown herein is assumed to have a sufficient number of hydrogen atom(s) to satisfy the valences described or shown. In certain instances one or more substituents having a double bond (e.g., “oxo” or “═O”) as the point of attachment may be described, shown or listed herein within a substituent group, wherein the structure may only show a single bond as the point of attachment to the core structure of Formula (I). A person of ordinary skill in the art would understand that, while only a single bond is shown, a double bond is intended for those substituents. As used herein, the term “and the like,” with reference to the definitions of chemical terms provided herein, means that variations in chemical structures that could be expected by one skilled in the art include, without limitation, isomers (including chain, branching or positional structural isomers), hydration of ring systems (including saturation or partial unsaturation of monocyclic, bicyclic or polycyclic ring structures) and all other variations where allowed by available valences which result in a stable compound. For the purposes of this description, where one or more substituent variables for a compound of Formula (I) or a form thereof encompass functionalities incorporated into a compound of Formula (I), each functionality appearing at any location within the disclosed compound may be independently selected, and as appropriate, independently and/or optionally substituted. As used herein, the terms “independently selected,” or “each selected” refer to functional variables in a substituent list that may occur more than once on the structure of Formula (I), the pattern of substitution at each occurrence is independent of the pattern at any other occurrence. Further, the use of a generic substituent variable on any formula or structure for a compound described herein is understood to include the replacement of the generic substituent with species substituents that are included within the particular genus, e.g., aryl may be replaced with phenyl or naphthalenyl and the like, and that the resulting compound is to be included within the scope of the compounds described herein. As used herein, the terms “each instance of” or “in each instance, when present,” when used preceding a phrase such as “ . . . C3-14cycloalkyl, C3-14cycloalkyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, heteroaryl-C1-4alkyl, heterocyclyl and heterocyclyl-C1-4alkyl,” are intended to refer to the C3-14cycloalkyl, aryl, heteroaryl and heterocyclyl ring systems when each are present either alone or as a substituent. As used herein, the term “optionally substituted” means optional substitution with the specified substituent variables, groups, radicals or moieties. Compound Forms As used herein, the term “form” means a compound of Formula (I) having a form selected from the group consisting of a free acid, free base, salt, hydrate, solvate, racemate, enantiomer, diastereomer, stereoisomer, and tautomer form thereof. In certain aspects described herein, the form of the compound of Formula (I) is a free acid, free base or salt thereof. In certain aspects described herein, the form of the compound of Formula (I) is a salt thereof. In certain aspects described herein, the form of the compound of Formula (I) is an isotopologue thereof. In certain aspects described herein, the form of the compound of Formula (I) is a stereoisomer, racemate, enantiomer or diastereomer thereof. In certain aspects described herein, the form of the compound of Formula (I) is a tautomer thereof. In certain aspects described herein, the form of the compound of Formula (I) is a pharmaceutically acceptable form. In certain aspects described herein, the compound of Formula (I) or a form thereof is isolated for use. As used herein, the term “isolated” means the physical state of a compound of Formula (I) or a form thereof after being isolated and/or purified from a synthetic process (e.g., from a reaction mixture) or natural source or combination thereof according to an isolation or purification process or processes described herein or which are well known to the skilled artisan (e.g., chromatography, recrystallization and the like) in sufficient purity to be characterized by standard analytical techniques described herein or well known to the skilled artisan. As used herein, the term “protected” means that a functional group in a compound of Formula (I) or a form thereof is in a form modified to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene et al,Protective Groups in organic Synthesis(1991), Wiley, New York. Such functional groups include hydroxy, phenol, amino and carboxylic acid. Suitable protecting groups for hydroxy or phenol include trialkylsilyl or diarylalkylsilyl (e.g., t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, substituted benzyl, methyl, methoxymethanol, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters. In certain instances, the protecting group may also be a polymer resin, such as a Wang resin or a 2-chlorotrityl-chloride resin. Protecting groups may be added or removed in accordance with standard techniques, which are well-known to those skilled in the art and as described herein. It will also be appreciated by those skilled in the art, although such protected derivatives of compounds described herein may not possess pharmacological activity as such, they may be administered to a subject and thereafter metabolized in the body to form compounds described herein which are pharmacologically active. Such derivatives may therefore be described as “prodrugs”. All prodrugs of compounds described herein are included within the scope of the use described herein. As used herein, the term “prodrug” means a form of an instant compound (e.g., a drug precursor) that is transformed in vivo to yield an active compound of Formula (I) or a form thereof. The transformation may occur by various mechanisms (e.g., by metabolic and/or non-metabolic chemical processes), such as, for example, by hydrolysis and/or metabolism in blood, liver and/or other organs and tissues. A discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, “Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987. In one example, when a compound of Formula (I) or a form thereof contains a carboxylic acid functional group, a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a functional group such as alkyl and the like. In another example, when a compound of Formula (I) or a form thereof contains a hydroxyl functional group, a prodrug form can be prepared by replacing the hydrogen atom of the hydroxyl with another functional group such as alkyl, alkylcarbonyl or a phosphonate ester and the like. In another example, when a compound of Formula (I) or a form thereof contains an amine functional group, a prodrug form can be prepared by replacing one or more amine hydrogen atoms with a functional group such as alkyl or substituted carbonyl. Pharmaceutically acceptable prodrugs of compounds of Formula (I) or a form thereof include those compounds substituted with one or more of the following groups: carboxylic acid esters, sulfonate esters, amino acid esters, phosphonate esters and mono-, di- or triphosphate esters or alkyl substituents, where appropriate. As described herein, it is understood by a person of ordinary skill in the art that one or more of such substituents may be used to provide a compound of Formula (I) or a form thereof as a prodrug. One or more compounds described herein may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and the description herein is intended to embrace both solvated and unsolvated forms. As used herein, the term “solvate” means a physical association of a compound described herein with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. As used herein, “solvate” encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. As used herein, the term “hydrate” means a solvate wherein the solvent molecule is water. The compounds of Formula (I) can form salts, which are intended to be included within the scope of this description. Reference to a compound of Formula (I) or a form thereof herein is understood to include reference to salt forms thereof, unless otherwise indicated. The term “salt(s)”, as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases. In addition, when a compound of Formula (I) or a form thereof contains both a basic moiety, such as, without limitation an amine moiety, and an acidic moiety, such as, but not limited to a carboxylic acid, zwitterions (“inner salts”) may be formed and are included within the term “salt(s)” as used herein. The term “pharmaceutically acceptable salt(s)”, as used herein, means those salts of compounds described herein that are safe and effective (i.e., non-toxic, physiologically acceptable) for use in mammals and that possess biological activity, although other salts are also useful. Salts of the compounds of the Formula (I) may be formed, for example, by reacting a compound of Formula (I) or a form thereof with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization. Pharmaceutically acceptable salts include one or more salts of acidic or basic groups present in compounds described herein. Particular aspects of acid addition salts include, and are not limited to, acetate, ascorbate, benzoate, benzenesulfonate, bisulfate, bitartrate, borate, bromide, butyrate, chloride, citrate, camphorate, camphorsulfonate, ethanesulfonate, formate, fumarate, gentisinate, gluconate, glucaronate, glutamate, iodide, isonicotinate, lactate, maleate, methanesulfonate, naphthalenesulfonate, nitrate, oxalate, pamoate, pantothenate, phosphate, propionate, saccharate, salicylate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate (also known as tosylate), trifluoroacetate salts and the like. Certain particular aspects of acid addition salts include chloride, bromide or dichloride. Additionally, acids which are generally considered suitable for the formation of pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl et al, Camille G. (eds.) Handbook of Pharmaceutical Salts.Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al,Journal of Pharmaceutical Sciences(1977) 66(1) 1-19; P. Gould,International J. of Pharmaceutics(1986) 33, 201-217; Anderson et al,The Practice of Medicinal Chemistry(1996), Academic Press, New York; and inThe Orange Book(Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference thereto. Suitable basic salts include, but are not limited to, aluminum, ammonium, calcium, lithium, magnesium, potassium, sodium and zinc salts. All such acid salts and base salts are intended to be included within the scope of pharmaceutically acceptable salts as described herein. In addition, all such acid and base salts are considered equivalent to the free forms of the corresponding compounds for purposes of this description. Compounds of Formula (I) and forms thereof, may further exist in a tautomeric form. All such tautomeric forms are contemplated and intended to be included within the scope of the compounds of Formula (I) or a form thereof as described herein. The compounds of Formula (I) or a form thereof may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. The present description is intended to include all stereoisomeric forms of the compounds of Formula (I) as well as mixtures thereof, including racemic mixtures. The compounds described herein may include one or more chiral centers, and as such may exist as racemic mixtures (R/S) or as substantially pure enantiomers and diastereomers. The compounds may also exist as substantially pure (R) or (S) enantiomers (when one chiral center is present). In one particular aspect, the compounds described herein are (S) isomers and may exist as enantiomerically pure compositions substantially comprising only the (S) isomer. In another particular aspect, the compounds described herein are (R) isomers and may exist as enantiomerically pure compositions substantially comprising only the (R) isomer. As one of skill in the art will recognize, when more than one chiral center is present, the compounds described herein may also exist as a (R,R), (R,S), (S,R) or (S,S) isomer, as defined by IUPAC Nomenclature Recommendations. As used herein, the term “substantially pure” refers to compounds consisting substantially of a single isomer in an amount greater than or equal to 90%, in an amount greater than or equal to 92%, in an amount greater than or equal to 95%, in an amount greater than or equal to 98%, in an amount greater than or equal to 99%, or in an amount equal to 100% of the single isomer. In one aspect of the description, a compound of Formula (I) or a form thereof is a substantially pure (S) enantiomer form present in an amount greater than or equal to 90%, in an amount greater than or equal to 92%, in an amount greater than or equal to 95%, in an amount greater than or equal to 98%, in an amount greater than or equal to 99%, or in an amount equal to 100%. In one aspect of the description, a compound of Formula (I) or a form thereof is a substantially pure (R) enantiomer form present in an amount greater than or equal to 90%, in an amount greater than or equal to 92%, in an amount greater than or equal to 95%, in an amount greater than or equal to 98%, in an amount greater than or equal to 99%, or in an amount equal to 100%. As used herein, a “racemate” is any mixture of isometric forms that are not “enantiomerically pure”, including mixtures such as, without limitation, in a ratio of about 50/50, about 60/40, about 70/30, or about 80/20. In addition, the present description embraces all geometric and positional isomers. For example, if a compound of Formula (I) or a form thereof incorporates a double bond or a fused ring, both the cis- and trans-forms, as well as mixtures, are embraced within the scope of the description. Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by use of chiral HPLC column or other chromatographic methods known to those skilled in the art. Enantiomers can also be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Also, some of the compounds of Formula (I) may be atropisomers (e.g., substituted biaryls) and are considered as part of this description. All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds (including those of the salts, solvates, esters and prodrugs of the compounds as well as the salts, solvates and esters of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this description, as are positional isomers (such as, for example, 4-pyridyl and 3-pyridyl). Individual stereoisomers of the compounds described herein may, for example, be substantially free of other isomers, or may be present in a racemic mixture, as described supra. The use of the terms “salt”, “solvate”, “ester”, “prodrug” and the like, is intended to equally apply to the salt, solvate, ester and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or isotopologues of the instant compounds. The term “isotopologue” refers to isotopically-enriched compounds described herein which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds described herein include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as2H,3H,13C,14C,15N,18O,17O,31P,32P,35S,18F,35Cl and36Cl, respectively, each of which are also within the scope of this description. Certain isotopically-enriched compounds described herein (e.g., those labeled with3H and14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e.,3H) and carbon-14 (i.e.,14C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e.,2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Polymorphic crystalline and amorphous forms of the compounds of Formula (I) and of the salts, solvates, hydrates, esters and prodrugs of the compounds of Formula (I) are further intended to be included in the present description. Compound Uses An aspect of the present description relates to a method of use of a compound of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound or a form thereof to the subject. Another aspect of the present description relates to use of the compound of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof. Another aspect of the present description relates to use of the compound of Formula (I) or a form thereof having activity toward HD. An aspect of the present description relates to use of the compound of Formula (I) or a form thereof in a combination therapy to provide additive or synergistic activity, thus enabling the development of a combination product for treating or ameliorating HD. In addition to monotherapeutic use, the instant compounds are useful in a combination therapy with current standard of agents, having additive or synergistic activity with one or more known agents. A combination therapy comprising compounds described herein in combination with one or more known drugs may be used to treat HD regardless of whether HD is responsive to the known drug. Certain aspects of the present description include the use of a compound of Formula (I) or a form thereof in a combination therapy for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound of Formula (I) or a form thereof and an effective amount of one or more agent(s). Certain particular aspects of the present description include the use of a compound of Formula (I) or a form thereof in a combination therapy for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound of Formula (I) or a form thereof and an effective amount of one or more agent(s). In an aspect of a use or method provided herein, compounds of Formula (I) or a form thereof used in combination with one or more additional agents can be administered to a subject or contacted with a subject or patient cell(s) prior to, concurrently with, or subsequent to administering to the subject or patient or contacting the cell with an additional agent(s). A compound(s) of Formula (I) or a form thereof and an additional agent(s) can be administered to a subject or contacted with a cell in single composition or different compositions. In a specific aspect, a compound(s) of Formula (I) or a form thereof is used in combination with gene therapy to inhibit HTT expression (using, e.g., viral delivery vectors) or the administration of another small molecule HTT inhibitor. In another specific aspect, a compound(s) of Formula (I) or a form thereof are used in combination with cell replacement using differentiated non-mutant HTT stem cells. In another specific aspect, a compound(s) of Formula (I) or a form thereof are used in combination with cell replacement using differentiated HTT stem cells. In one aspect, provided herein is the use of compounds of Formula (I) or a form thereof in combination with supportive standard of care therapies, including palliative care. An aspect of the present description includes the use of a compound of Formula (I) or a form thereof in the preparation of a kit comprising the compound of Formula (I) or a form thereof and instructions for administering an effective amount of the compound of Formula (I) or a form thereof and an effective amount of one or more agent(s) in a combination therapy for treating or ameliorating HD in a subject in need thereof. Accordingly, the present description relates to use of a compound of Formula (I) or a form thereof for treating or ameliorating HD. In accordance with the use of the present description, compounds that are useful in selectively treating or ameliorating HD, have been identified and use of these compounds for treating or ameliorating HD has been provided. Another aspect of the use of the present description relates to use of a compound of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound of Formula (I) or a form thereof to the subject. Another aspect of the use of the present description relates to a method of use of a compound of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound to the subject. Another aspect of the use of the present description relates to a method of use of a compound of Formula (I) or a form thereof for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound to the subject. Another aspect of the use of the present description relates to use of a compound of Formula (I) or a form thereof in the manufacture of a medicament for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the medicament to the subject. Another aspect of the use of the present description relates to use of a compound of Formula (I) or a form thereof in the preparation of a kit comprising the compound of Formula (I) or a form thereof and instructions for administering the compound for treating or ameliorating HD in a subject in need thereof. In one respect, for each of such aspects, the subject is treatment naive. In another respect, for each of such aspects, the subject is not treatment naive. As used herein, the term “treating” refers to: (i) preventing a disease, disorder or condition from occurring in a subject that may be predisposed to the disease, disorder and/or condition but has not yet been diagnosed as having the disease, disorder and/or condition; (ii) inhibiting a disease, disorder or condition, i.e., arresting the development thereof; and/or (iii) relieving a disease, disorder or condition, i.e., causing regression of the disease, disorder and/or condition. As used herein, the term “subject” refers to an animal or any living organism having sensation and the power of voluntary movement, and which requires oxygen and organic food. Nonlimiting examples include members of the human, primate, equine, porcine, bovine, murine, rattus, canine and feline specie. In certain aspects, the subject is a mammal or a warm-blooded vertebrate animal. In other aspects, the subject is a human. As used herein, the term “patient” may be used interchangeably with “subject” and “human”. As used herein, the terms “effective amount” or “therapeutically effective amount” mean an amount of compound of Formula (I) or a form, composition or medicament thereof that achieves a target plasma concentration that is effective in treating or ameliorating HD as described herein and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect in a subject in need thereof. In one aspect, the effective amount may be the amount required to treat HD in a subject or patient, more specifically, in a human. In another aspect, the concentration-biological effect relationships observed with regard to a compound of Formula (I) or a form thereof indicate a target plasma concentration ranging from approximately 0.001 μg/mL to approximately 50 μg/mL, from approximately 0.01 μg/mL to approximately 20 μg/mL, from approximately 0.05 μg/mL to approximately 10 μg/mL, or from approximately 0.1 μg/mL to approximately 5 μg/mL. To achieve such plasma concentrations, the compounds described herein may be administered at doses that vary, such as, for example, without limitation, from 0.1 ng to 10,000 mg. In one aspect, the dose administered to achieve an effective target plasma concentration may be administered based upon subject or patient specific factors, wherein the doses administered on a weight basis may be in the range of from about 0.001 mg/kg/day to about 3500 mg/kg/day, or about 0.001 mg/kg/day to about 3000 mg/kg/day, or about 0.001 mg/kg/day to about 2500 mg/kg/day, or about 0.001 mg/kg/day to about 2000 mg/kg/day, or about 0.001 mg/kg/day to about 1500 mg/kg/day, or about 0.001 mg/kg/day to about 1000 mg/kg/day, or about 0.001 mg/kg/day to about 500 mg/kg/day, or about 0.001 mg/kg/day to about 250 mg/kg/day, or about 0.001 mg/kg/day to about 200 mg/kg/day, or about 0.001 mg/kg/day to about 150 mg/kg/day, or about 0.001 mg/kg/day to about 100 mg/kg/day, or about 0.001 mg/kg/day to about 75 mg/kg/day, or about 0.001 mg/kg/day to about 50 mg/kg/day, or about 0.001 mg/kg/day to about 25 mg/kg/day, or about 0.001 mg/kg/day to about 10 mg/kg/day, or about 0.001 mg/kg/day to about 5 mg/kg/day, or about 0.001 mg/kg/day to about 1 mg/kg/day, or about 0.001 mg/kg/day to about 0.5 mg/kg/day, or about 0.001 mg/kg/day to about 0.1 mg/kg/day, or from about 0.01 mg/kg/day to about 3500 mg/kg/day, or about 0.01 mg/kg/day to about 3000 mg/kg/day, or about 0.01 mg/kg/day to about 2500 mg/kg/day, or about 0.01 mg/kg/day to about 2000 mg/kg/day, or about 0.01 mg/kg/day to about 1500 mg/kg/day, or about 0.01 mg/kg/day to about 1000 mg/kg/day, or about 0.01 mg/kg/day to about 500 mg/kg/day, or about 0.01 mg/kg/day to about 250 mg/kg/day, or about 0.01 mg/kg/day to about 200 mg/kg/day, or about 0.01 mg/kg/day to about 150 mg/kg/day, or about 0.01 mg/kg/day to about 100 mg/kg/day, or about 0.01 mg/kg/day to about 75 mg/kg/day, or about 0.01 mg/kg/day to about 50 mg/kg/day, or about 0.01 mg/kg/day to about 25 mg/kg/day, or about 0.01 mg/kg/day to about 10 mg/kg/day, or about 0.01 mg/kg/day to about 5 mg/kg/day, or about 0.01 mg/kg/day to about 1 mg/kg/day, or about 0.01 mg/kg/day to about 0.5 mg/kg/day, or about 0.01 mg/kg/day to about 0.1 mg/kg/day, or from about 0.1 mg/kg/day to about 3500 mg/kg/day, or about 0.1 mg/kg/day to about 3000 mg/kg/day, or about 0.1 mg/kg/day to about 2500 mg/kg/day, or about 0.1 mg/kg/day to about 2000 mg/kg/day, or about 0.1 mg/kg/day to about 1500 mg/kg/day, or about 0.1 mg/kg/day to about 1000 mg/kg/day, or about 0.1 mg/kg/day to about 500 mg/kg/day, or about 0.1 mg/kg/day to about 250 mg/kg/day, or about 0.1 mg/kg/day to about 200 mg/kg/day, or about 0.1 mg/kg/day to about 150 mg/kg/day, or about 0.1 mg/kg/day to about 100 mg/kg/day, or about 0.1 mg/kg/day to about 75 mg/kg/day, or about 0.1 mg/kg/day to about 50 mg/kg/day, or about 0.1 mg/kg/day to about 25 mg/kg/day, or about 0.1 mg/kg/day to about 10 mg/kg/day, or about 0.1 mg/kg/day to about 5 mg/kg/day, or about 0.1 mg/kg/day to about 1 mg/kg/day, or about 0.1 mg/kg/day to about 0.5 mg/kg/day. Effective amounts for a given subject may be determined by routine experimentation that is within the skill and judgment of a clinician or a practitioner skilled in the art in light of factors related to the subject. Dosage and administration may be adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect. Factors which may be taken into account include genetic screening, severity of the disease state, status of disease progression, general health of the subject, ethnicity, age, weight, gender, diet, time of day and frequency of administration, drug combination(s), reaction sensitivities, experience with other therapies, and tolerance/response to therapy. The dose administered to achieve an effective target plasma concentration may be orally administered once (once in approximately a 24 hour period; i.e., “q.d.”), twice (once in approximately a 12 hour period; i.e., “b.i.d.” or “q.12h”), thrice (once in approximately an 8 hour period; i.e., “t.i.d.” or “q.8h”), or four times (once in approximately a 6 hour period; i.e., “q.d.s.”, “q.i.d.” or “q.6h”) daily. In certain aspects, the dose administered to achieve an effective target plasma concentration may also be administered in a single, divided, or continuous dose for a patient or subject having a weight in a range of between about 40 to about 200 kg (which dose may be adjusted for patients or subjects above or below this range, particularly children under 40 kg). The typical adult subject is expected to have a median weight in a range of about 70 kg. Long-acting pharmaceutical compositions may be administered every 2, 3 or 4 days, once every week, or once every two weeks depending on half-life and clearance rate of the particular formulation. The compounds and compositions described herein may be administered to the subject via any drug delivery route known in the art. Nonlimiting examples include oral, ocular, rectal, buccal, topical, nasal, sublingual, transdermal, subcutaneous, intramuscular, intravenous (bolus and infusion), intracerebral, and pulmonary routes of administration. In another aspect, the dose administered may be adjusted based upon a dosage form described herein formulated for delivery at about 0.02, 0.025, 0.03, 0.05, 0.06, 0.075, 0.08, 0.09, 0.10, 0.20, 0.25, 0.30, 0.50, 0.60, 0.75, 0.80, 0.90, 1.0, 1.10, 1.20, 1.25, 1.50, 1.75, 2.0, 3.0, 5.0, 10, 20, 30, 40, 50, 100, 150, 200, 250, 300, 400, 500, 1000, 1500, 2000, 2500, 3000 or 4000 mg/day. For any compound, the effective amount can be estimated initially either in cell culture assays or in relevant animal models, such as a mouse, guinea pig, chimpanzee, marmoset or tamarin animal model. Relevant animal models may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED50(the dose therapeutically effective in 50% of the population) and LD50(the dose lethal to 50% of the population). The dose ratio between therapeutic and toxic effects is therapeutic index, and can be expressed as the ratio, LD50/ED50. In certain aspects, the effective amount is such that a large therapeutic index is achieved. In further particular aspects, the dosage is within a range of circulating concentrations that include an ED50with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration. In one aspect, provided herein are methods for modulating the amount of HTT (huntingtin protein), comprising contacting a human cell with a compound of Formula (I) or a form thereof. In a specific aspect, provided herein are methods for modulating the amount of HTT, comprising contacting a human cell with a compound of Formula (I) or a form thereof that modulates the expression of HTT. The human cell can be contacted with a compound of Formula (I) or a form thereof in vitro, or in vivo, e.g., in a non-human animal or in a human. In a specific aspect, the human cell is from or in a human. In another specific aspect, the human cell is from or in a human with HD. In another specific aspect, the human cell is from or in a human with HD, caused by a CAG repeat in the Htt gene, resulting in a loss of HTT expression and/or function. In another aspect, the human cell is from a human with HD. In another aspect, the human cell is in a human with HD. In one aspect, the compound is a form of the compound of Formula (I). In a specific aspect, provided herein is a method for enhancing the inhibition of mutant HTT transcribed from the Htt gene, comprising contacting a human cell with a compound of Formula (I) or a form thereof. The human cell can be contacted with a compound of Formula (I) or a form thereof in vitro, or in vivo, e.g., in a non-human animal or in a human. In a specific aspect, the human cell is from or in a human. In another specific aspect, the human cell is from or in a human with HD. In another specific aspect, the human cell is from or in a human with HD, caused by a CAG repeat in the Htt gene, resulting in a loss of wild-type “normal” HTT expression and/or function. In another aspect, the human cell is from a human with HD. In another aspect, the human cell is in a human with HD. In one aspect, the compound is a form of the compound of Formula (I). In another aspect, provided herein is a method for modulating the inhibition of mutant HTT transcribed from the Htt gene, comprising administering to a non-human animal model for HD a compound of Formula (I) or a form thereof. In a specific aspect, provided herein is a method for modulating the inhibition of mutant HTT transcribed from the Htt gene, comprising administering to a non-human animal model for HD a compound of Formula (I) or a form thereof. In a specific aspect, the compound is a form of the compound of Formula (I). In another aspect, provided herein is a method for decreasing the amount of mutant HTT, comprising contacting a human cell with a compound of Formula (I) or a form thereof. In a specific aspect, provided herein is a method for decreasing the amount of mutant HTT, comprising contacting a human cell with a compound of Formula (I) that inhibits the transcription of mutant HTT (huntingtin mRNA) from the Htt gene. In another specific aspect, provided herein is a method for decreasing the amount of HTT, comprising contacting a human cell with a compound of Formula (I) that inhibits the expression of mutant HTT transcribed from the Htt gene. The human cell can be contacted with a compound of Formula (I) or a form thereof in vitro, or in vivo, e.g., in a non-human animal or in a human. In a specific aspect, the human cell is from or in a human. In another specific aspect, the human cell is from or in a human with HD. In another specific aspect, the human cell is from or in a human with HD, caused by a CAG repeat in the Htt gene, resulting in a loss of HTT expression and/or function. In another aspect, the human cell is from a human with HD. In another aspect, the human cell is in a human with HD. In one aspect, the compound is a form of the compound of Formula (I). In certain aspects, treating or ameliorating HD with a compound of Formula (I) or a form thereof (alone or in combination with an additional agent) has a therapeutic effect and/or beneficial effect. In a specific aspect, treating HD with a compound of Formula (I) or a form thereof (alone or in combination with an additional agent) results in one, two or more of the following effects: (i) reduces or ameliorates the severity of HD; (ii) delays onset of HD; (iii) inhibits the progression of HD; (iv) reduces hospitalization of a subject; (v) reduces hospitalization length for a subject; (vi) increases the survival of a subject; (vii) improves the quality of life for a subject; (viii) reduces the number of symptoms associated with HD; (ix) reduces or ameliorates the severity of a symptom(s) associated with HD; (x) reduces the duration of a symptom associated with HD; (xi) prevents the recurrence of a symptom associated with HD; (xii) inhibits the development or onset of a symptom of HD; and/or (xiii) inhibits of the progression of a symptom associated with HD. Metabolites Also included within the scope of the present description are the use of in vivo metabolic products of the compounds described herein. Such products may result, for example, from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the description includes the use of compounds produced by a process comprising contacting a compound described herein with a mammalian tissue or a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radio-labeled isotopologue (e.g.,14C or3H) of a compound described herein, administering the radio-labeled compound in a detectable dose (e.g., greater than about 0.5 mg/kg) to a mammal such as a rat, mouse, guinea pig, dog, monkey or human, allowing sufficient time for metabolism to occur (typically about 30 seconds to about 30 hours), and identifying the metabolic conversion products from urine, bile, blood or other biological samples. The conversion products are easily isolated since they are “radiolabeled” by virtue of being isotopically-enriched (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g., by MS or NMR analysis. In general, analysis of metabolites may be done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds described herein even if they possess no biological activity of their own. Pharmaceutical Compositions Aspects of the present description include the use of a compound of Formula (I) or a form thereof in a pharmaceutical composition for treating or ameliorating HD in a subject in need thereof, comprising administering an effective amount of the compound of Formula (I) or a form thereof in admixture with one or more pharmaceutically acceptable excipient(s). An aspect of the present description includes the use of a pharmaceutical composition of the compound of Formula (I) or a form thereof in the preparation of a kit comprising the pharmaceutical composition of the compound of Formula (I) or a form thereof and instructions for administering the compound for treating or ameliorating HD in a subject in need thereof. As used herein, the term “composition” means a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The pharmaceutical composition may be formulated to achieve a physiologically compatible pH, ranging from about pH 3 to about pH 11. In certain aspects, the pharmaceutical composition is formulated to achieve a pH of from about pH 3 to about pH 7. In other aspects, the pharmaceutical composition is formulated to achieve a pH of from about pH 5 to about pH 8. The term “pharmaceutically acceptable excipient” refers to an excipient for administration of a pharmaceutical agent, such as the compounds described herein. The term refers to any pharmaceutical excipient that may be administered without undue toxicity. Pharmaceutically acceptable excipients may be determined in part by the particular composition being administered, as well as by the particular mode of administration and/or dosage form. Nonlimiting examples of pharmaceutically acceptable excipients include carriers, solvents, stabilizers, adjuvants, diluents, etc. Accordingly, there exists a wide variety of suitable formulations of pharmaceutical compositions for the instant compounds described herein (see, e.g., Remington's Pharmaceutical Sciences). Suitable excipients may be carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive antibodies. Other exemplary excipients include antioxidants such as ascorbic acid; chelating agents such as EDTA; carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose (e.g., hydroxypropylmethylcellulose, also known as HPMC), stearic acid; liquids such as oils, water, saline, glycerol and ethanol; wetting or emulsifying agents; pH buffering substances; and the like. Liposomes are also included within the definition of pharmaceutically acceptable excipients. The pharmaceutical compositions described herein may be formulated in any form suitable for the intended use described herein. Suitable formulations for oral administration include solids, liquid solutions, emulsions and suspensions, while suitable inhalable formulations for pulmonary administration include liquids and powders. Alternative formulations include syrups, creams, ointments, tablets, and lyophilized solids which can be reconstituted with a physiologically compatible solvent prior to administration. When intended for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, non-aqueous solutions, dispersible powders or granules (including micronized particles or nanoparticles), emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents, and preserving agents, in order to provide a palatable preparation. Pharmaceutically acceptable excipients suitable for use in conjunction with tablets include, for example, inert diluents, such as celluloses, calcium or sodium carbonate, lactose, calcium or sodium phosphate; disintegrating agents, such as croscarmellose sodium, cross-linked povidone, maize starch, or alginic acid; binding agents, such as povidone, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid, or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed. Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example celluloses, lactose, calcium phosphate, or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with non-aqueous or oil medium, such as glycerin, propylene glycol, polyethylene glycol, peanut oil, liquid paraffin, or olive oil. In other aspects, pharmaceutical compositions described herein may be formulated as suspensions comprising a compound of Formula (I) or a form thereof in admixture with one or more pharmaceutically acceptable excipient(s) suitable for the manufacture of a suspension. In yet other aspects, pharmaceutical compositions described herein may be formulated as dispersible powders and granules suitable for preparation of a suspension by the addition of one or more excipient(s). Excipients suitable for use in connection with suspensions include suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth, gum acacia, dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycethanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate); and thickening agents, such as carbomer, beeswax, hard paraffin, or cetyl alcohol. The suspensions may also contain one or more preservatives such as acetic acid, methyl and/or n-propyl p-hydroxy-benzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin. The pharmaceutical compositions described herein may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth; naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids; hexitol anhydrides, such as sorbitan monooleate; and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent. Additionally, the pharmaceutical compositions described herein may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous emulsion or oleaginous suspension. Such emulsion or suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,2-propanediol. The sterile injectable preparation may also be prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables. The compounds described herein may be substantially insoluble in water and sparingly soluble in most pharmaceutically acceptable protic solvents and vegetable oils, but generally soluble in medium-chain fatty acids (e.g., caprylic and capric acids) or triglycerides and in propylene glycol esters of medium-chain fatty acids. Thus, contemplated in the description are compounds which have been modified by substitutions or additions of chemical or biochemical moieties which make them more suitable for delivery (e.g., increase solubility, bioactivity, palatability, decrease adverse reactions, etc.), for example by esterification, glycosylation, PEGylation, etc. In certain aspects, the compound described herein is formulated for oral administration in a lipid-based composition suitable for low solubility compounds. Lipid-based formulations can generally enhance the oral bioavailability of such compounds. As such, pharmaceutical compositions described herein may comprise a effective amount of a compound of Formula (I) or a form thereof, together with at least one pharmaceutically acceptable excipient selected from medium chain fatty acids or propylene glycol esters thereof (e.g., propylene glycol esters of edible fatty acids such as caprylic and capric fatty acids) and pharmaceutically acceptable surfactants, such as polysorbate 20 or 80 (also referred to as Tween® 20 or Tween® 80, respectively) or polyoxyl 40 hydrogenated castor oil. In other aspects, the bioavailability of low solubility compounds may be enhanced using particle size optimization techniques including the preparation of nanoparticles or nanosuspensions using techniques known to those skilled in the art. The compound forms present in such preparations include amorphous, partially amorphous, partially crystalline or crystalline forms. In alternative aspects, the pharmaceutical composition may further comprise one or more aqueous solubility enhancer(s), such as a cyclodextrin. Nonlimiting examples of cyclodextrin include hydroxypropyl, hydroxyethyl, glucosyl, maltosyl and maltotriosyl derivatives of α-, β-, and γ-cyclodextrin, and hydroxypropyl-β-cyclodextrin (HPBC). In certain aspects, the pharmaceutical composition further comprises HPBC in a range of from about 0.1% to about 20%, from about 1% to about 15%, or from about 2.5% to about 10%. The amount of solubility enhancer employed may depend on the amount of the compound in the composition. Preparation of Compounds General Synthetic Methods As disclosed herein, general methods for preparing the compounds of Formula (I) or a form thereof as described herein are available via standard, well-known synthetic methodology. Many of the starting materials are commercially available or, when not available, can be prepared using the routes described below using techniques known to those skilled in the art. The synthetic schemes provided herein comprise multiple reaction steps, each of which is intended to stand on its own and can be carried out with or without any preceding or succeeding step(s). In other words, each of the individual reaction steps of the synthetic schemes provided herein in isolation is contemplated. Scheme A: Compounds of Formula (I), wherein R1is C3-10cycloalkyl or heterocyclyl ring systems and R2is phenyl, heterocyclyl, or heteroaryl ring systems, may be prepared as described in Scheme A below. Compound A1 (where X1and X2are independently bromine, chlorine and the like; W2and W3, are independently CH or N) is converted to Compound A2 by a nucleophilic substitution with a primary amine in the presence of a suitable base (such as Et3N and the like) in a suitable solvent (such as decanol and the like). Alternatively, Compound A1 is converted to Compound A2 via cross coupling with a primary amine in the presence of a suitable catalyst (such as RuPhos Pd G2 and the like) and base (such as sodium tert-butoxide and the like) in an appropriate solvent such as 1,4-dioxane and the like). Compound A2 is converted to Compound A3 by a diazotization/cyclization sequence upon treatment with an appropriate reagent (such as sodium nitrite and the like) in an appropriate solvent (such acetic acid and the like). Compound A3 is converted to Compound A4 by a Suzuki coupling with an aryl- or heteroaryl-boronic acid (or pinacol boronic ester) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound A3 is converted to Compound A4 by a Stille coupling with an aryl- or heteroaryl-stannane in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as X-Phos and the like) and a base (such as CsF and the like) in a suitable solvent (such as 1,4-dioxane and the like). Any protection groups on R1and R2are removed upon treatment with a suitable reagent (such as HCl in dioxane for a Boc protecting group and the like) in a suitable solvent (such as dioxane and the like). Scheme B: Compounds of Formula (I), wherein R1is C3-10cycloalkyl or heterocyclyl ring systems and R2is phenyl, heterocyclyl, or heteroaryl ring systems, may be prepared as described in Scheme B below. Compound B1 (where X1and X2are independently bromine, chlorine and the like; W2and W3, are independently CH or N) is converted to Compound B2 through a nucleophilic substitution/cyclization sequence by treatment with hydrazine (R1NH2NH2) and a suitable base (such as Et3N and the like) in a suitable solvent (such as methanol and the like). Compound B2 is converted to Compound B3 by a Suzuki coupling with an aryl- or heteroaryl-boronic acid (or pinacol boronic ester) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound B2 is converted to Compound B3 by a Stille coupling with an aryl- or heteroaryl-stannane in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as X-Phos and the like) and a base (such as CsF and the like) in a suitable solvent (such as 1,4-dioxane and the like). Compound B3 is converted to Compound B4 by treatment with an activated triflate (such as Tf2O or Tf2NPh and the like) in the presence of a suitable base (such as Et3N and the like) in a suitable solvent (such as dichloromethane and the like). Compound B4 is converted to Compound B5 by hydrogenation using an appropriate hydrogen source (such as ammonium formate and the like) in the presence of a suitable catalyst (such as Pd(dppf)Cl2and the like) in a suitable solvent (such as tetrahydrofuran and the like). Any protection groups on R1and R2are removed upon treatment with a suitable reagent (such as HCl in dioxane for a Boc protecting group and the like) in a suitable solvent (such as dioxane and the like). Scheme C: Compounds of Formula (I), wherein R1is C3-10cycloalkyl or heterocyclyl ring systems and R2is phenyl, heterocyclyl, or heteroaryl ring systems, may be prepared as described in Scheme C below. Compound C1 (where X1and X2are independently bromine, chlorine and the like; W2and W3, are independently CH or N) is converted to Compound C2 by a Sonogashira coupling with a TMS protected acetylene in the presence of a suitable catalyst (such as Pd(PPh3)2Cl2and the like and CuI and the like) and suitable base (such as Et3N and the like) in a suitable solvent (such as acetonitrile and the like). Compound C2 is converted to Compound C3 by heating in a suitable solvent (such as DMF and the like) in the presence of a suitable base (such as K2CO3and the like). Compound C3 is converted to Compound C4 by a Suzuki coupling with an aryl- or heteroaryl-boronic acid (or pinacol boronic ester) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound C3 is converted to Compound C4 by a Stille coupling with an aryl- or heteroaryl-stannane in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as X-Phos and the like) and a base (such as CsF and the like) in a suitable solvent (such as 1,4-dioxane and the like). Compound C4 is converted to Compound C5 (where X3is iodine, bromine and the like) by halogenation with a suitable reagent (such as NIS the like) in a suitable solvent (such as DMF and the like). Compound C5 is converted to Compound C6 by a Suzuki coupling with an optionally substituted and appropriately protected amino-containing cycloalkyl/cycloalkenyl pinacol boronic ester in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound C5 is converted to Compound C6 by a Negishi coupling with an optionally substituted and appropriately protected amino-containing cycloalkyl zinc halide in the presence of a catalyst (such as Pd(dppf)Cl2and the like) in a suitable solvent (such as 1,4-dioxane and the like). Upon treatment with a deprotecting agent appropriate for the protecting group (such as HCl in dioxane for a Boc protecting group and the like), Compound C6 is converted to Compound C7. In cases where unsaturation exists in the ring containing the basic amino group, the compound may be converted to the fully saturated analog under an atmosphere of H2in a suitable solvent (such as methanol and the like) and in the presence of catalyst (such as 10% Pd/C and the like). Scheme D: Compounds of Formula (I), wherein R1is C3-10cycloalkyl or heterocyclyl ring systems and R2is phenyl, heterocyclyl, or heteroaryl ring systems, may be prepared as described in Scheme D below. Compound D1 (where X1, X2and X3are independently bromine, chlorine and the like; W2and W3, are independently CH or N) is converted to Compound D2 by a Suzuki coupling with a vinyl pinacol boronic ester in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Compound D2 is converted to Compound D3 by heating with a primary amine (R1NH2) in a suitable solvent (such as acetonitrile and the like). Compound D3 is converted to Compound D4 by a Suzuki coupling with an aryl- or heteroaryl-boronic acid (or pinacol boronic ester) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound D3 is converted to Compound D4 by a Stille coupling with an aryl- or heteroaryl-stannane in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as X-Phos and the like) and a base (such as CsF and the like) in a suitable solvent (such as 1,4-dioxane and the like). Compound D4 is converted to Compound D5 by treating with a suitable oxidizing agent (such as manganese dioxide and the like) in a suitable solvent (such as toluene and the like). Any protection groups on R1and R2are removed upon treatment with a suitable reagent (such as HCl in dioxane for a Boc protecting group and the like) in a suitable solvent (such as dioxane and the like). Scheme E: Compounds of Formula (I), wherein R1is C3-10cycloalkyl or heterocyclyl ring systems and R2is phenyl, heterocyclyl, or heteroaryl ring systems, may be prepared as described in Scheme E below. Compound E1 (where X1and X2are independently bromine, chlorine and the like; W2and W3, are independently CH or N) is converted to Compound E2 through a condensation/cyclization sequence in the presence of a suitable base (such as Et3N and the like) in a suitable solvent (such as acetonitrile and the like). Compound E2 is converted to Compound E3 by TIPS protection of the hydroxyl group by using an appropriate reagent (such as TIPSCl or TIPSOTf and the like) in the presence of a suitable base (such as imidazole and the like) in a suitable solvent (such as DMF and the like). Compound E3 is converted to Compound E4 by a Suzuki coupling with an aryl- or heteroaryl-boronic acid (or pinacol boronic ester) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound E3 is converted to Compound E4 by a Stille coupling with an aryl- or heteroaryl-stannane in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as X-Phos and the like) and a base (such as CsF and the like) in a suitable solvent (such as 1,4-dioxane and the like). Compound E4 is converted to Compound E5 through hydrolysis of methyl ester in the presence of a suitable base (such as aqueous NaOH and the like) in a suitable solvent (such as methanol and the like) followed by decarboxylation of the resulting carboxylic acid upon heating in the appropriate solvent (such as DMSO and the like). Compound E5 is converted to Compound E6 by treatment with an activated triflate (such as Tf2O or Tf2NPh and the like) in presence of a suitable base (such as Et3N and the like) in a suitable solvent (such as dichloromethane and the like). Compound E6 is converted to Compound E7 by a Suzuki coupling with an optionally substituted and appropriately protected amino-containing cycloalkyl/cycloalkenyl pinacol boronic ester in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound E6 is converted to Compound E7 by a Negishi coupling with an optionally substituted and appropriately protected amino-containing cycloalkyl zinc halide in the presence of a catalyst (such as Pd(dppf)Cl2and the like) in a suitable solvent (such as 1,4-dioxane and the like). In cases where unsaturation exists in the ring containing the basic amino group, the compound may be converted to the fully saturated analog under an atmosphere of H2in a suitable solvent (such as methanol and the like) and in the presence of catalyst (such as 10% Pd/C and the like). Any protection groups on R1and R2are removed upon treatment with a suitable reagent (such as HCl in dioxane for a Boc protecting group and the like) in a suitable solvent (such as dioxane and the like). Scheme F: Compounds of Formula (I), wherein R1is C3-10cycloalkyl or heterocyclyl ring systems and R2is phenyl, heterocyclyl, or heteroaryl ring systems, may be prepared as described in Scheme F below. Compound F1 (where X1and X2are independently bromine, chlorine and the like; W2and W3, are independently CH or N) is converted to Compound F2 through nucleophilic substitution with methyl 2-hydroxyacetate in the presence of a suitable base (such as NaH and the like) in a suitable solvent (such as THF and the like). Compound F2 is converted to Compound F3 by cyclization upon treatment with an appropriate base (such as NaOMe and the like) in a suitable solvent (such as THF and the like). Compound F3 is converted to Compound F4 by a Suzuki coupling with an aryl- or heteroaryl-boronic acid (or pinacol boronic ester) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound F3 is converted to Compound F4 by a Stille coupling with an aryl- or heteroaryl-stannane in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as X-Phos and the like) and a base (such as CsF and the like) in a suitable solvent (such as 1,4-dioxane and the like). Compound F4 is converted to Compound F5 through a hydrolysis/decarboxylation sequence in the presence of a suitable base (such as aqueous NaOH and the like) in a suitable solvent (such as DMSO and the like). Compound F5 is converted to Compound F6 by treatment with an activated triflate (such as Tf2or Tf2NPh and the like) in presence of a suitable base (such as Et3N and the like) in a suitable solvent (such as dichloromethane and the like). Compound F6 is converted to Compound F7 by a Suzuki coupling with an optionally substituted and appropriately protected amino-containing cycloalkyl/cycloalkenyl pinacol boronic ester in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound F6 is converted to Compound F7 by a Negishi coupling with an optionally substituted and appropriately protected amino-containing cycloalkyl zinc halide in the presence of a catalyst (such as Pd(dppf)Cl2and the like) in a suitable solvent (such as 1,4-dioxane and the like). In cases where unsaturation exists in the ring containing the basic amino group, the compound may be converted to the fully saturated analog under an atmosphere of H2in a suitable solvent (such as methanol and the like) and in the presence of catalyst (such as 10% Pd/C and the like). Any protection groups on R1and R2are removed upon treatment with a suitable reagent (such as HCl in dioxane for a Boc protecting group and the like) in a suitable solvent (such as dioxane and the like). Scheme G: Compounds of Formula (I), wherein R1is C3-10cycloalkyl or heterocyclyl ring systems and R2is phenyl, heterocyclyl, or heteroaryl ring systems, may be prepared as described in Scheme F below. Compound G1 (where X1and X2are independently bromine, chlorine and the like; W2and W3, are independently CH or N) is converted to Compound G2 by a nucleophilic substitution with a primary amine in the presence of a suitable base (such as Et3N and the like) in a suitable solvent (such as decanol and the like). Alternatively, Compound G1 is converted to Compound G2 via cross coupling with a primary amine in the presence of a suitable catalyst (such as RuPhos Pd G2 and the like) and base (such as sodium tert-butoxide and the like) in an appropriate solvent such as 1,4-dioxane and the like). Compound G2 is converted to Compound G3 (where R3is H, Me, Et and the like) via cyclization using an appropriate reagent (such as triethylorthoformate and the like) in the presence of an appropriate catalyst (such as HCl and the like). Compound G3 is converted to Compound G4 by a Suzuki coupling with an aryl- or heteroaryl-boronic acid (or pinacol boronic ester) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound G3 is converted to Compound G4 by a Stille coupling with an aryl- or heteroaryl-stannane in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as X-Phos and the like) and a base (such as CsF and the like) in a suitable solvent (such as 1,4-dioxane and the like). Any protection groups on R1and R2are removed upon treatment with a suitable reagent (such as HCl in dioxane for a Boc protecting group and the like) in a suitable solvent (such as dioxane and the like). Scheme H: Compounds of Formula (I), wherein R1is alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, R2is hydrogen, fluorine, chlorine, hydroxy, methoxy, aryl, or heteroaryl, and R3is monocyclic or bicyclic heterocyclyl or heteroaryl ring systems, may be prepared as described in Scheme H below. Compound H1 (where X1, X2and X3are independently bromine, chlorine and the like; W2and W3, are independently CH or N) is converted to Compound H2 by a nucleophilic substitution with sodium benzenesulfinate in a suitable solvent (such as THF, DMSO and the like). Compound H2 is converted to Compound H3 by a nucleophilic substitution with a primary amine in the presence of a suitable base (such as K2CO3and the like) in a suitable solvent (such as dioxane and the like). Alternatively, Compound H2 is converted to Compound H3 via cross coupling with a primary amine in the presence of a suitable catalyst (such as RuPhos Pd G2 and the like) and base (such as sodium tert-butoxide and the like) in an appropriate solvent such as 1,4-dioxane and the like). Compound H3 is converted to Compound H4 by treatment with sodium azide in an appropriate solvent (such as DMSO and the like). Compound H4 is converted to Compound H5 by a reduction upon treatment with an appropriate reagent (such as zinc metal and the like) in the presence of an appropriate acid (such as acetic acid and the like) in the appropriate solvent (such as CH2Cl2and the like). Compound H5 is converted to Compound H6 via a diazotization/cyclization sequence upon treatment with an appropriate reagent (such as sodium nitrite and the like) in an appropriate solvent (such acetic acid and the like). Compound H6 is converted to Compound H8 by a Suzuki coupling with an aryl-boronic acid (or pinacol boronic ester) H7 (where X4is bromine, chlorine and the like; R2is hydrogen, fluorine, chlorine, hydroxy, methoxy, aryl or heteroaryl; and P is a protecting group such as MOM and the like) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Compound H8 is converted to Compound H9 by a Suzuki coupling with an aryl- or heteroaryl-boronic acid (or pinacol boronic ester) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and a base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound H8 is converted to Compound H9 by a Stille coupling with an aryl- or heteroaryl-stannane in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as X-Phos and the like) and a base (such as CsF and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound H8 is converted to Compound H9 by treatment with pinacolatodiboron and a base (such as KOAc and the like) in the presence of a catalyst (such as Pd(dppf)Cl2and the like) in an appropriate solvent (such as 1,4-dioxane and the like), followed by addition of an aryl- or heteroaryl-halide in the presence of a catalyst (such as Pd(dppf)Cl2and the like) and a base (such as aqueous K2CO3and the like) in a suitable solvent (such as 1,4-dioxane and the like). Alternatively, Compound H8 is converted to Compound H9 by a Buchwald-Hartwig coupling with a heteroaryl or amine in the presence of a catalyst (such as Pd2(dba)3and the like), a ligand (such as tBuX-Phos and the like) and a base (such as K3PO4and the like) in a suitable solvent (such as 1,4-dioxane and the like). Compound H9 is converted to Compound H10 upon treatment with conditions appropriate to the removal of the protecting groups (such as HCl in dioxane for a MOM protecting group) in a suitable solvent (such as dioxane and the like). SPECIFIC SYNTHETIC EXAMPLES To describe in more detail and assist in understanding, the following non-limiting examples are offered to more fully illustrate the scope of compounds described herein and are not to be construed as specifically limiting the scope thereof. Such variations of the compounds described herein that may be now known or later developed, which would be within the purview of one skilled in the art to ascertain, are considered to fall within the scope of the compounds as described herein and hereinafter claimed. These examples illustrate the preparation of certain compounds. Those of skill in the art will understand that the techniques described in these examples represent techniques, as described by those of ordinary skill in the art, that function well in synthetic practice, and as such constitute preferred modes for the practice thereof. However, it should be appreciated that those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific methods that are disclosed and still obtain a like or similar result without departing from the spirit and scope of the present description. Other than in the following examples of the embodied compounds, unless indicated to the contrary, all numbers expressing quantities of ingredients, reaction conditions, experimental data, and so forth used in the specification and claims are to be understood as being modified by the term “about”. Accordingly, all such numbers represent approximations that may vary depending upon the desired properties sought to be obtained by a reaction or as a result of variable experimental conditions. Therefore, within an expected range of experimental reproducibility, the term “about” in the context of the resulting data, refers to a range for data provided that may vary according to a standard deviation from the mean. As well, for experimental results provided, the resulting data may be rounded up or down to present data consistently, without loss of significant figures. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should be construed in light of the number of significant digits and rounding techniques used by those of skill in the art. While the numerical ranges and parameters setting forth the broad scope of the present description are approximations, the numerical values set forth in the examples set forth below are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements. Compound Examples As used above, and throughout the present description, the following abbreviations, unless otherwise indicated, shall be understood to have the following meanings: AbbreviationMeaningΔheating (chemistry) or deletion (biology)AcOH or HOAcacetic acidAc2Oacetic anhydrideArargonACN or CH3CN oracetonitrileMeCNatmatmosphere(s)BBr3boron tribromideBnOHbenzyl alcoholBoctert-butoxy-carbonylBoc2Odi-tert-butyl dicarbonateB2pin2bis(pinacolato)diboronBuOHn-butanol(t-Bu)3P HBF4Tri-t-butylphosphonium tetrafluoroborate° C.degrees CentigradeCelite ® or Celitediatomaceous earth(COCl)2oxalyl chlorideCsClcesium chlorideCs2CO3cesium carbonateCsFcesium fluorideCuIcopper(I) iodided/h/hr/hrs/min/sday(d)/hour(h, hr or hrs)/minute(min)/second(s)DAST(diethylamino)sulfur trifluorideDCM or CH2Cl2dichloromethaneDIEA or DIPEAN,N-diisopropylethylamineDMAdimethylacetamideDMAP4-(dimethylamino)pyridineDME1,2-dimethoxyethaneDMFdimethylformamideDMSOdimethylsulfoxideEtIiodoethaneEt3NtriethylamineEtOAcethyl acetateEtOHethanolEt2Odiethyl etherH2hydrogenHClhydrochloric acidH2SO4sulfuric acidHCOOHformic acidK2CO3potassium carbonateKOAcpotassium acetateKOtBuPotassium t-butoxideKOHpotassium hydroxideKSCNpotassium thiocyanateLAHlithium aluminum hydrideLC/MS, LCMS orliquid chromatographic mass spectroscopyLC-MSLDAlithium diisopropylamineLiOHlithium hydroxideMeOHmethanolMeIiodomethaneMgSO4magnesium sulfateMOMmethoxy methylMOMClchloromethyl methyl etherMSmass spectroscopyNBSN-bromosuccinimideNCSN-chlorosuccinimideNH4Clammonium chlorideNH4OAcammonium acetateNH4OHammonium hydroxide or aqueous ammoniaNH2OH•HClhydroxylamine hydrochlorideNaBH4sodium borohydrideNa2CO3sodium carbonateNaHsodium hydrideNaHCO3sodium bicarbonateNaHsodium hydrideNaOAcsodium acetateNaOHsodium hydroxideNaOMesodium methoxideNa2SO4sodium sulfateN2nitrogenNH4Clammoniuim chlorideNMPN-methylpyrrolidoneNMRnuclear magnetic resonanceNOESYNuclear Overhauser Enhancement SpectroscopyPdpalladiumPd/Cpalladium on carbonPd2(dba)3or Pd2dba3tris(dibenzylideneacetone)dipalladium(0)Pd(dppf)Cl2or[1,1'-Pd(dppf)Cl2—CH2Cl2bis(diphenylphosphino)ferrocene]dichloro-palladium(II), complex with dichloromethanePd(PPh3)4ortetrakis(triphenylphosphine)palladium(0))Pd(Ph3P)4Pd(PPh3)2Cl2,bis(triphenylphosphine)palladium(II)PdCl2(PPh3)2ordichloridePdCl2(Ph3P)2PhMetoluenePsipounds per square inch pressurePt2Oplatinum(IV) oxidePyBOP(benzotriazol-1-yloxy)tripyrrolidinophos-phonium hexafluorophosphatePyBroP ®bromotripyrrolidinophosphonium hexa-fluorophosphateRTretention timeRuPhos Pd G2chloro(2-dicyclohexylphosphino-2′,6′-diisopropoxy-1,1′-biphenyl)[2-(2′-amino-1,1′-bipheny])]palladium(II)SOCl2thionly chlorideSO2Cl2sulfuryl chlorideTEA, Et3N or NEt3triethylamineTFAtrifluoroacetic acidTf2NPhN-phenyl-bis(trifluoromethanesulfon-imide) or 1,1,1-trifluoro-N-phenyl-N-[(trifluoromethyl)sulfonyl]methanesul-fonamide or N,N-bis(trifluoromethyl-sulfonyl)aniline or N-phenyl-trifluoro-methanesulfonimideTf2Otrifluoromethanesulfonic anhydrideTHFtetrahydrofuranTHPtetrahydropyranylTIPStiisopropylsilaneTIPSCltriisopropylsilyl chlorideTIPSOTftriisopropylsilyl trifluoromethanesul-fonate or trifluoromethanesulfonic acidtriisopropylsilyl ester or triisopropylsilyltriflateTLCthin layer chromatographyTMEDAtetramethylethylenediamineTMStrimethylsilaneTMSCltrimethylchlorosilane or trimethylsilyl chloridet-Butert-butylUPLCultra performance liquid chromatography Example 1 Preparation of Compound 11 Step 1: To suspension of 3,6-dichloropyridazin-4-amine (1.48 g, 9.02 mmol) in CH2Cl2(25 mL) was added di-tertbutyl dicarbonate (2.2 g, 10 mmol) in one portion followed by addition of a few crystals of DMAP. The reaction was stirred at room temperature for 4 h. Almost complete conversion was observed accompanied by formation of di-Boc material. The solvent was removed under reduced pressure and the product was isolated by silica gel column chromatography eluting with EtOAc/hexanes gradient (0-20% EtOAc) to afford tert-butyl N-(3,6-dichloropyridazin-4-yl)carbamate (1.71 g, 72%) as a white solid. Step 2: To a mixture of tert-butyl N-(3,6-dichloropyridazin-4-yl)carbamate (1.71 g, 6.47 mmol), CuI (75 mg, 0.39 mmol) and Pd(PPh3)2Cl2(140 mg, 0.20 mmol) in CH3CN (25 mL) under an argon atmosphere was added Et3N (4.50 mL, 32.3 mmol) followed by ethynyl(trimethyl)silane (1.10 mL, 7.78 mmol). The mixture was heated under an argon atmosphere for 1 h, after which, no starting material was detected by UPLC. The solvent was concentrated and the residue was treated with EtOAc. The solid was filtered, washed well with EtOAc and discarded. The mother liquor was concentrated and the residue was purified by silica gel column chromatography eluting with EtOAc/hexanes gradient (0-20% EtOAc) to afford tert-butyl N-[6-chloro-3-(2-trimethylsilylethynyl)pyridazin-4-yl]carbamate (1.12 g, 53%) as an oil which solidified on standing. Step 3: To a solution of tert-butyl N-[6-chloro-3-(2-trimethylsilylethynyl)pyridazin-4-yl]carbamate (1.1 g, 3.4 mmol) in DMF (10 mL) was added powdered K2CO3(1.00 g, 7.24 mmol). The mixture was heated at 60° C. for 30 min. The reaction was then diluted with water and extracted with EtOAc. Upon drying of the organic phase over Na2SO4and concentration of the solvent, the residue was purified by silica gel column chromatography eluting with EtOAc/hexanes gradient (0-50% EtOAc) to afford tert-butyl 3-chloropyrrolo[3,2-c]pyridazine-5-carboxylate (0.600 g, 70%) as a white solid. MS m/z 254.3 [M+H]+. Step 4: 1-Bromo-4-iodo-2-methoxybenzene (50 g, 160 mmol) was suspended in dichloromethane (75 mL) at −10° C. 1 N BBr3in CH2Cl2(250 mL, 250 mmol) was cannulated in over 30 minutes, with the internal temperature remaining below 0° C. throughout the addition. After the addition, the mixture was stirred at 0° C. for 1 h, and then at room temperature for an additional 16 h. The mixture was cooled in an ice bath. 10% Aqueous Na2CO3(250 mL) was added in portions. The mixture was then partitioned between H2O and dichloromethane. The dichloromethane layer was dried over MgSO4and then filtered. 2-Bromo-5-iodophenol (46 g, 96%) was obtained from the filtrate as a pinkish-white solid. 1H NMR (acetone-d6) δ: 9.24 (br s, 1H), 7.38 (d, J=2 Hz, 1H), 7.31 (d, J=8.5 Hz, 1H), 7.17 (dd, J=8.5 Hz, 2 Hz, 1H). Step 5: 2-Bromo-5-iodophenol (54.9 g, 184 mmol), was dissolved in DMF (240 mL) at 0° C. 2.5 M Sodium tert-pentoxide in THF (90 mL, 230 mmol) was then added dropwise. The reaction was stirred at 0° C. for 15 minutes after the addition was complete. Chloromethyl methyl ether (18 mL, 225 mmol) was added dropwise over 30 minutes. The mixture was warmed to ambient temperature and was stirred for 16 h. The mixture was diluted with H2O (1500 mL) and was extracted into EtOAc (2×400 mL). The combined organic layers were washed with H2O (300 mL), and then with brine. The organic layer was dried over MgSO4, filtered, and concentrated under vacuum. The crude product was flushed through a silica plug using CH2Cl2in hexanes (0-10%) to yield 1-bromo-4-iodo-2-(methoxymethoxy)benzene (61 g, 97%) as a clear liquid. 1H NMR (acetone-d6) δ: 7.56 (d, J=2 Hz, 1H), 7.38 (d, J=8 Hz, 1H), 7.33 (dd, J=8 Hz, 2 Hz, 1H), 5.35 (s, 2H), 3.50 (s, 3H). Step 6: 1-Bromo-4-iodo-2-(methoxymethoxy)benzene (49 g, 143 mmol), 1-(tetrahydro-2H-pyran-2-yl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (48.4 g, 174 mmol), PdCl2(dppf)-dichloromethane adduct (3.1 g, 3.6 mmol), dioxane (500 mL), and aqueous 1 N K2CO3(350 mL, 350 mmol) were heated at 90° C. for 2 h. The reaction mixture was then partitioned between H2O and EtOAc. The organic layer was dried over MgSO4, filtered, and concentrated under vacuum. Purification by silica gel chromatography (EtOAc in hexanes, 20-50%), followed by trituration with hexanes, yielded 4-(4-bromo-3-(methoxymethoxy)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (40.4 g, 77%) as an off-white solid. 1H NMR (acetone-d6) δ: 8.22 (s, 1H), 7.88 (s, 1H), 7.55 (d, J=8.5 Hz, 1H), 7.47 (d, J=2 Hz, 1H), 7.23 (dd, J=8.5 Hz, 2 Hz, 1H), 5.44 (dd, J=9.5 Hz, 2.5 Hz, 1H), 5.38 (S, 2H), 4.01 (m, 1H), 3.72 (m, 1H), 3.51 (s, 3H), 2.1-2.23 (m, 1H), 2.0-2.1 (m, 2H), 1.7-1.8 (m, 1H), 1.6-1.7 (m, 2H). Step 7: Potassium acetate (22 g, 224 mmol) was pumped dry at 180° C. for 2 h, and then the flask was filled with argon. 4-(4-Bromo-3-(methoxymethoxy)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (20 g, 54.5 mmol), PdCl2(dppf)-dichloromethane adduct (1.22 g, 1.47 mmol), bis(pinacolato)diboron (20.8 g, 81.9 mmol), and dry toluene (200 mL) was added. This mixture was heated at 110° C. for 2 days. The mixture was filtered through Celite®, eluting with ether. The filtrate was concentrated under vacuum, re-dissolved in ether, and was filtered again through Celite® to remove solid impurities. Purification by silica gel chromatography (EtOAc in hexanes, 20-50%) yielded crude product (12 g) that was mostly free of protodeboronated by-product. This was dissolved in ether (100 mL) and washed with aqueous NaHCO3(2×1.5 L) and brine, then dried over MgSO4, and then filtered. The filtrate was concentrated to provide pure product (7.05 g, 32%) as a glassy semi-solid. 1H NMR (500 MHz, acetone-d6): δ 8.24 (s, 1H), 7.90 (s, 1H), 7.65 (d, J=8 Hz, 1H), 7.33 (d, J=1.5 Hz, 1H), 7.29 (dd, J=8 Hz, 1.5 Hz, 1H), 5.45 (dd, J=10 Hz, 2.5 Hz, 1H), 5.25 (s, 2H), 4.01 (m, 1H), 3.69-3.74 (m, 1H), 3.52 (s, 3H), 2.15-2.2 (m, 1H), 2.0-2.1 (m, 2H), 1.7-1.8 (m, 1H), 1.6-1.68 (m, 2H), 1.35 (s, 12H). Step 8: A mixture of tert-butyl 3-chloropyrrolo[3,2-c]pyridazine-5-carboxylate (150 mg, 0.59 mmol), 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (0.300 g, 0.724 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(I) (25 mg, 0.033 mmol) and K2CO3(250 mg, 1.81 mmol) in a vial was evacuated and backfilled with argon. 1,4-Dioxane (2 mL) and water (0.5 mL) were added to the mixture and it was heated at 90° C. for 5 h. The mixture was cooled to room temperature, diluted with water, and the product was extracted with CH2Cl2(3 times). The combined organic layers were dried over Na2SO4and concentrated. The residue was purified by silica gel column chromatography eluting with EtOAc/hexanes gradient (70-100% EtOAc) to afford tert-butyl 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazine-5-carboxylate (0.15 g, 0.297 mmol, 100 mass %, 50.2%) as a white solid. MS m/z 506.6 [M+H]+;1H NMR (acetone-d6) δ: 8.67 (s, 1H), 8.30 (d, J=0.9 Hz, 1H), 8.10 (d, J=4.1 Hz, 1H), 8.02 (d, J=8.2 Hz, 1H), 7.97 (d, J=0.9 Hz, 1H), 7.58 (d, J=1.6 Hz, 1H), 7.48 (dd, J=7.9, 1.6 Hz, 1H), 7.08 (dd, J=3.8, 0.9 Hz, 1H), 5.49 (dd, J=9.8, 2.5 Hz, 1H), 5.40 (s, 2H), 3.98-4.06 (m, 1H), 3.70-3.79 (m, 1H), 3.45 (s, 3H), 2.17-2.26 (m, 1H), 2.03-2.09 (m, 2H), 1.81 (s, 1H), 1.74 (s, 9H), 1.57-1.68 (m, 2H). Step 9: A mixture of tert-butyl 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazine-5-carboxylate (140 mg, 0.28 mmol) in diphenyl ether (1.6 mL) was heated at 200° C. for 15 min and monitored by UPLC. Once complete, the reaction was cooled to room temperature and a precipitate was formed. The mixture was then diluted with pentane. The solid was filtered and washed with additional pentane. After drying, 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5H-pyrrolo[3,2-c]pyridazine (110 mg, 0.27 mmol) was dissolved in DMF (1.5 mL) and N-iodosuccinimide (68 mg, 0.30 mmol) was added. The reaction was stirred at room temperature for 15 min and a product precipitated out of the solution. The reaction was diluted with water and the solid was filtered and washed with water and dried. 7-Iodo-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5H-pyrrolo[3,2-c]pyridazine (140 mg, 97%) was obtained as tan solid. MS m/z 532.4 [M+H]+; Step 10: 7-Iodo-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5H-pyrrolo[3,2-c]pyridazine (140 mg, 0.26 mmol) was suspended in CH2Cl2(2 mL) and di-tertbutyl dicarbonate (80 mg, 0.37 mmol) was added followed by few crystals of DMAP. The reaction was stirred at room temperature and monitored by UPLC until complete consumption of the starting material was observed (20 min). The solvent was removed under reduce pressure and the residue was purified by silica gel column chromatography (60-100% EtOAc in hexanes) to afford tert-butyl 7-iodo-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazine-5-carboxylate (122 mg, 71%) as a pale yellow foam. MS m/z 632.5 [M+H]+;1H NMR (acetone-d6) δ: 8.66 (s, 1H), 8.31 (d, J=0.6 Hz, 1H), 8.27 (s, 1H), 8.03 (d, J=7.9 Hz, 1H), 7.98 (d, J=0.9 Hz, 1H), 7.58 (d, J=1.6 Hz, 1H), 7.49 (dd, J=7.9, 1.6 Hz, 1H), 5.49 (dd, J=9.8, 2.5 Hz, 1H), 5.41 (s, 2H), 3.98-4.06 (m, 1H), 3.69-3.80 (m, 1H), 3.45 (s, 3H), 2.14-2.28 (m, 1H), 2.02-2.08 (m, 2H), 1.77-1.84 (m, 1H), 1.75 (s, 9H), 1.60-1.69 (m, 2H). Step 11: An oven-dried flask was equipped with a magnetic stir bar and charged with tert-butyl 7-iodo-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazine-5-carboxylate (122 mg, 0.19 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylate (75 mg, 0.24 mmol), Pd(PPh3)4(25 mg, 0.022 mmol) and K2CO3(80 mg, 0.58 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (3×). 1,4-Dioxane (1.2 mL) and water (0.3 mL) were added and the reaction was heated to 90° C. for 6 h. The reaction was cooled to room temperature, diluted with water (5 mL), and then extracted with CH2Cl2(3×). The combined organic layers were dried over Na2SO4and concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with EtOAc/hexanes gradient (60-100% EtOAc) to afford tert-butyl 7-(1-tert-butoxycarbonyl-3,6-dihydro-2H-pyridin-4-yl)-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazine-5-carboxylate (38 mg, 29%) and tert-butyl 4-[3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5H-pyrrolo[3,2-c]pyridazin-7-yl]-3,6-dihydro-2H-pyridine-1-carboxylate (25 mg, 22%). Step 12: To tert-butyl 7-(1-tert-butoxycarbonyl-3,6-dihydro-2H-pyridin-4-yl)-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazine-5-carboxylate (38 mg, 0.055 mmol) was added 4N HCl in dioxane (1 mL, 4.0 mmol) followed by MeOH (1 mL). The reaction was heated at 55° C. for 8 h. The solvents were removed under reduced pressure and the residue was triturated in Et2O. The resultant solid was filtered, washed well with excess Et2O and dried under a nitrogen flow to afford 5-(1H-pyrazol-4-yl)-2-[7-(1,2,3,6-tetrahydropyridin-1-ium-4-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]phenol hydrochloride (15 mg, 71%) as a bright yellow solid. MS m/z 359.3 [M+H]+;1H NMR (DMSO-d6) δ:13.26 (br s, 1H), 9.49 (s, 2H), 8.60 (s, 1H), 8.50 (br. s., 1H), 8.20 (s, 2H), 7.82 (d, J=8.5 Hz, 1H), 7.41 (td, J=4.3, 1.9 Hz, 2H), 6.97-7.16 (m, 1H), 3.81-4.00 (m, 2H), 3.35-3.55 (m, 2H), 2.77-2.97 (m, 2H); 1H not observed (NH or OH). Using the procedure described for Example 1, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData20MS m/z 415.5 [M + H]+;1H NMR (methanol-d4) δ: 8.61 (s, 1H), 8.45 (s, 1H), 8.26 (s,2H), 7.77 (d, J = 7.9 Hz, 1H), 7.45 (dd, J = 7.9, 1.6 Hz, 1H), 7.36 (d, J = 1.6 Hz, 1H),7.06 (s, 1H), 3.73-3.79 (m, J = 6.6 Hz, 1H), 3.57-3.63 (m, J = 5.0 Hz, 1H), 1.71 (s, 6H),1.64 (s, 6H); 4 Hs not observed (3 NHs and OH).27MS m/z 385.4 [M + H]+;1H NMR (methanol-d4) δ: 8.60 (s, 1H), 8.42-8.49 (m, J = 2.5Hz, 2H), 8.39 (s, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.48 (dd, J = 8.0, 1.7 Hz, 1H), 7.40 (d,J = 1.6 Hz, 1H), 7.35 (d, J = 5.7 Hz, 1H), 4.54 (dd, J = 6.3, 5.4 Hz, 1H), 4.43 (dd, J = 7.3,4.1 Hz, 1H), 3.35-3.41 (m, 1H), 2.87 (d, J = 18.0 Hz, 1H), 2.42-2.52 (m, 1H), 2.37-2.42(m, 1H), 2.25-2.36 (m, 1H), 2.03-2.16 (m, 1H); 4 Hs not observed (3 NHs and OH).28MS m/z 401.4 [M + H]+;1H NMR (methanol-d4) δ: 8.62 (s, 1H), 8.50 (s, 2H), 8.47 (s,1H), 7.79 (d, J = 7.9 Hz, 1H), 7.49 (dd, J = 8.2, 1.6 Hz, 1H), 7.41 (d, J = 1.6 Hz, 1H),7.25 (d, J = 5.7 Hz, 1H), 4.33 (d, J = 6.0 Hz, 1H), 4.11-4.15 (m, 2H), 4.06 (dd, J = 12.6,1.9 Hz, 1H), 3.97 (d, J = 7.3 Hz, 1H), 3.93 (d, J = 12.6 Hz, 1H), 3.39 (dd, J = 18.6, 8.2Hz, 1H), 3.10 (dd, J = 18.3, 1.6 Hz, 1H); 4 Hs not observed (3 NHs and OH). Example 2 Preparation of Compound 30 Step 1: A solution of tert-butyl 7-[5-tert-butoxycarbonyl-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazin-7-yl]-3-oxa-9-azabicyclo[3.3.1]non-6-ene-9-carboxylate (prepared following the method described in Example 1, step 11) (100 mg, 0.14 mmol) in MeOH (2 mL) and EtOAc (0.2 mL) was hydrogenated over 10% Pd/C (20 mg, 0.02 mmol, 10 mass %) and 10% Pd(OH)2/C (20 mg, 0.014 mmol, 10 mass %) in a Parr shaker at 50 psi of H2over 72 h. The catalysts were filtered and washed with MeOH. The mother liquor was concentrated and the residue was purified by silica gel column chromatography eluting with a MeOH/CH2Cl2gradient (0-10% MeOH) to afford tert-butyl 7-[5-tert-butoxycarbonyl-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazin-7-yl]-3-oxa-9-azabicyclo[3.3.1]nonane-9-carboxylate (46 mg, 46%) as a pale yellow foam. MS m/z 631.4 [M+H]+. Step 2: To tert-butyl 7-[5-tert-butoxycarbonyl-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazin-7-yl]-3-oxa-9-azabicyclo[3.3.1]nonane-9-carboxylate (46 mg, 0.063 mmol) was added 4N HCl in dioxane (0.5 mL, 2 mmol) followed by MeOH (1 mL). The reaction was stirred at 50° C. for 16 h. Volatiles were then removed under reduced pressure, the residue was then triturated with Et2O, and the solid was filtered and dried in a nitrogen flow to afford 2-[7-(3-oxa-9-azoniabicyclo[3.3.1]nonan-7-yl)-5H-pyrrolo[3,2-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol hydrochloride (16 mg, 58%) as a yellow solid. Stereochemistry was assigned based on NOESY data. MS m/z 403.4 [M+H]+;1H NMR (methanol-d4) δ: 8.53 (s, 1H), 8.46 (s, 2H), 8.26 (d, J=0.6 Hz, 1H), 7.75 (d, J=8.2 Hz, 1H), 7.47 (dd, J=8.2, 1.9 Hz, 1H), 7.39 (d, J=1.9 Hz, 1H), 3.96 (dd, J=12.6, 1.9 Hz, 2H), 3.90 (d, J=12.6 Hz, 2H), 3.85 (dd, J=9.4, 3.2 Hz, 2H), 3.66-3.78 (m, 1H), 2.82 (ddd, J=14.5, 9.4, 6.0 Hz, 2H), 2.51 (ddd, J=14.5, 11.4, 3.2 Hz, 2H); 4 Hs not observed (3 NHs and OH). Using the procedure described for Example 2, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData12MS m/z 361.3 [M + H]+29MS m/z 387.4 [M + H]+;1H NMR (methanol-d4) δ: 8.53 (s, 1H), 8.49 (s, 2H), 8.20 (d,J = 0.9 Hz, 1H), 7.77 (d, J = 8.2 Hz, 1H), 7.48 (dd, J = 8.2, 1.6 Hz, 1H), 7.40 (d, J = 1.6Hz, 1H), 4.20-4.27 (m, 2H), 3.74-3.84 (m, 1H), 3.66-3.71 (m, 2H), 2.33-2.41 (m,J = 2.5 Hz, 3H), 2.25-2.32 (m, 3H); 4 Hs not observed (3 NHs and OH).31MS m/z 417.5 [M + H]+;1H NMR (D2O) δ: 8.15 (s, 1H), 7.89 (s, 2H), 7.72 (s, 1H), 7.41(d, J = 8.2 Hz, 1H), 6.99 (d, J = 8.8 Hz, 1H), 6.81-6.92 (m, 1H), 3.60-3.70 (m, 1H), 2.01(dd, J = 14.2, 2.8 Hz, 2H), 1.70 (t, J = 14.2 2H), 1.54 (s, 6H), 1.40 (s, 6H); 4 Hs notobserved (3 NHs and OH). Example 3 Preparation of Compound 43 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with tert-butyl 7-iodo-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazine-5-carboxylate (43 mg, 0.07 mmol), tert-butyl piperazine-1-carboxylate (15 mg, 0.08 mmol), tris(dibenzylideneacetone)dipalladium(0) (6.3 mg, 0.007 mmol), S-Phos (5.7 mg, 0.014 mmol) and Cs2CO3(45 mg, 0.14 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). DME (3 mL) was added and the reaction was heated to 80° C. for 2 h. The reaction was cooled to room temperature, filtered through Celite, and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with EtOAc/hexanes gradient (40-80% EtOAc) to afford tert-butyl 7-(4-(tert-butoxycarbonyl)piperazin-1-yl)-3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-5H-pyrrolo[3,2-c]pyridazine-5-carboxylate (18 mg, 38%) as a brownish solid. MS m/z 690.4 [M+H]+. Step 2: To a solution of tert-butyl 7-(4-tert-butoxycarbonylpiperazin-1-yl)-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrrolo[3,2-c]pyridazine-5-carboxylate (18 mg, 0.026 mmol) in CH2Cl2(0.5 mL) plus 1 drop of MeOH was added 4M HCl in 1,4-dioxane (0.03 mL, 0.12 mmol) and the reaction mixture was stirred for 16 h at room temperature. The reaction was concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with MeOH/CH2Cl2(0% to 30% MeOH) to provide 2-(7-piperazin-1-yl-5H-pyrrolo[3,2-c]pyridazin-3-yl)-5-(1H-pyrazol-4-yl)phenol (5 mg, 53%) as an orange solid. MS m/z 362.0 [M+H]+;1H NMR (methanol-d4) δ: 8.45 (s, 1H), 8.15 (br s, 2H), 7.80 (s, 1H), 7.70 (d, J=8.0 Hz, 1H), 7.42 (dd, J=8.0, 1.9 Hz, 1H), 7.33 (d, J=1.9 Hz, 1H), 3.69-3.76 (m, 4H), 3.50-3.56 (m, 4H); 4 Hs not observed (3 NHs and OH). Example 4 Preparation of Compound 8 Step 1: A mixture of 2-bromo-5H-pyrrolo[2,3-b]pyrazine (50 mg, 0.25 mmol), 4-[3-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (107 mg, 0.28 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (19 mg, 0.025 mmol) was purged with argon. 1,4-Dioxane (2 mL) and aqueous 2 M K2CO3(0.35 mL, 0.7 mmol) were added and the reaction was heated at 90° C. for 16 h. The reaction mixture was then cooled to room temperature and diluted with water. The aqueous layer was extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4and concentrated under reduced pressure. The residue was purified using silica gel chromatography, eluting with a EtOAc/hexanes gradient (50-100% EtOAc) to afford 2-[2-methoxy-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5H-pyrrolo[2,3-b]pyrazine (76 mg, 80%) as a yellow foam. MS m/z 376.3 [M+H]+;1H NMR (DMSO-d6) δ: 8.67 (s, 1H), 8.51 (s, 1H), 8.06 (s, 1H), 7.82-7.88 (m, 1H), 7.71-7.76 (m, 1H), 7.39-7.44 (m, 1H), 7.32-7.38 (m, 1H), 6.63-6.69 (m, 1H), 5.41-5.46 (m, 1H), 3.94-4.00 (m, 1H), 3.93 (s, 3H), 3.63-3.71 (m, 1H), 2.10-2.20 (m, 2H), 1.94-1.98 (m, 2H), 1.52-1.60 (m, 2H). Step 2: A mixture of 2-[2-methoxy-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5H-pyrrolo[2,3-b]pyrazine (76 mg, 0.20 mmol), (2,2,6,6-tetramethyl-1,3-dihydropyridin-4-yl) trifluoromethanesulfonate (120 mg, 0.42 mmol), aqueous 1 M K3PO4(0.1 mL, 0.1 mmol), XPhos (18 mg, 0.04 mmol), tris(dibenzylideneacetone)dipalladium(0) (19 mg, 0.02 mmol), and 1,4-dioxane (2 mL) were heated under an argon atmosphere at 90° C. for 16 h. The reaction mixture was cooled to room temperature and diluted with water. The aqueous layer was extracted with CH2Cl2(3×). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified using silica gel chromatography, eluting with a MeOH/EtOAc gradient (0-10% MeOH) to afford 5-[2-methoxy-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-1-(2,2,6,6-tetramethyl-1,3-dihydropyridin-4-yl)pyrrolo[3,2-b] pyridine (40 mg, 39%) as an orange foam. MS m/z 513.3 [M+H]+;1H NMR (CDCl3) δ: 8.73 (s, 1H), 7.87 (s, 1H), 7.81 (d, J=0.8 Hz, 1H), 7.75 (d, J=7.9 Hz, 1H), 7.51 (d, J=3.7 Hz, 1H), 7.18 (dd, J=7.9, 1.6 Hz, 1H), 7.06 (d, J=1.5 Hz, 1H), 6.69 (d, J=3.7 Hz, 1H), 6.06 (t, J=1.5 Hz, 1H), 5.37 (dd, J=9.1, 3.3 Hz, 1H), 4.02-4.05 (m, 2H), 3.85 (s, 3H), 3.63-3.73 (m, 1H), 2.56 (d, J=1.4 Hz, 2H), 2.00-2.16 (m, 3H), 1.54-1.71 (m, 3H), 1.29 (s, 6H), 1.26 (s, 6H). Step 3: A solution of 2-[2-methoxy-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5-(2,2,6,6-tetramethyl-1,3-dihydropyridin-4-yl)pyrrolo[2,3-b]pyrazine (40 mg, 0.08 mmol) and NaSEt (85 mg, 0.8 mmol) in NMP (2 mL) was heated to 180° C. in the microwave for 30 min. The reaction mixture was then cooled to room temperature and diluted with CH2Cl2(10 mL). The precipitate was filtered by vacuum filtration. The filtrate was concentrated and used without further purification. A mixture of crude 5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-[5-(2,2,6,6-tetramethyl-1,3-dihydropyridin-4-yl)pyrrolo[2,3-b]pyrazin-2-yl]phenol (0.039 g, 0.08 mmol) and 4 M HCl in dioxane (0.5 mL, 2 mmol) was stirred at room temperature for 1 h. The precipitate formed was collected by vacuum filtration and rinsed with CH2Cl2(10 mL) to afford 5-(1H-pyrazol-4-yl)-2-[5-(2,2,6,6-tetramethyl-1,3-dihydropyridin-4-yl)pyrrolo[2,3-b]pyrazin-2-yl]phenol hydrochloride (7 mg, 43%) as a yellow solid. MS m/z 415.4 [M+H]+;1H NMR (DMSO-d6) δ: 9.15-9.25 (m, 2H), 8.16-8.20 (m, 1H), 8.13-8.16 (m, 2H), 8.08-8.12 (m, 1H), 7.22-7.27 (m, 2H), 6.92-6.97 (m, 1H), 6.40-6.46 (m, 1H), 3.02-3.06 (m, 2H), 1.63 (s, 6H), 1.56 (s, 6H). Example 5 Preparation of Compound 10 Step 1: To a mixture of 2,2,6,6-tetramethylpiperidin-4-amine (4.2 g, 27 mmol) in acetonitrile (10 mL) was added 2,5-dichloropyrazine (4.0 g, 27 mmol). The reaction mixture was stirred at 120° C. for 16 h. The reaction mixture was then allowed to cooled to room temperature and the precipitate was collected by vacuum filtration to afford 5-chloro-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyrazin-2-amine as a light yellow solid (3.1 g, 43%). MS m/z 269.3 [M+H]+;1H NMR (DMSO-d6) δ: 8.02 (d, J=1.3 Hz, 1H), 7.68 (d, J=1.3 Hz, 1H), 7.09 (d, J=7.7 Hz, 1H), 4.09 (dd, J=7.7, 3.7 Hz, 1H), 1.77 (dd, J=12.4, 3.7 Hz, 2H), 1.16 (s, 6H), 1.03 (s, 6H), 0.95-0.99 (m, 2H). Step 2: To a suspension of 5-chloro-N-(2,2,6,6-tetramethyl-4-piperidyl)pyrazin-2-amine (0.7 g, 3 mmol) in acetic acid (5 mL) at room temperature was added NBS (0.5 g, 3 mmol). After stirring for 30 min, a yellow precipitate formed. The precipitate was collected by vacuum filtration to afford 3-bromo-5-chloro-N-(2,2,6,6-tetramethyl-4-piperidyl)pyrazin-2-amine (0.7 g, 80%) as a yellow solid. MS m/z 347.2, 349.2 [M+H]+;1H NMR (DMSO-d6) δ: 8.19 (s, 1H), 6.91-6.95 (m, 1H), 4.31-4.36 (m, 1H), 1.90-1.96 (m, 2H), 1.72-1.76 (m, 2H), 1.39-1.45 (m, 12H). Step 3: A mixture of 3-bromo-5-chloro-N-(2,2,6,6-tetramethyl-4-piperidyl)pyrazin-2-amine (1.4 g, 4.0 mmol), CuI (0.05 g, 0.3 mmol), and PdCl2(PPh3)2(0.17 g, 0.24 mmol) was purged with argon. THF (20 mL), Et3N (2.2 mL, 16 mmol) and ethynyl(trimethyl)silane (0.8 mL, 6 mmol) were added sequentially. The resulting mixture was stirred under an argon atmosphere at 60° C. for 1 h. The reaction mixture was then cooled to room temperature and concentrated. The crude residue was purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-10% MeOH) to afford 5-chloro-N-(2,2,6,6-tetramethyl-4-piperidyl)-3-(2-trimethylsilylethynyl)pyrazin-2-amine (1.42 g, 97%) as a dark yellow solid. MS m/z 365.3 [M+H]+;1H NMR (DMSO-d6) δ: 8.19 (s, 1H), 4.32-4.37 (m, 1H), 1.98-2.03 (m, 2H), 1.63-1.67 (m, 2H), 1.43 (s, 6H), 1.40 (s, 6H), 0.26 (s, 9H). Step 4: To a solution of 5-chloro-N-(2,2,6,6-tetramethyl-4-piperidyl)-3-(2-trimethylsilylethynyl)pyrazin-2-amine (1.42 g, 3.89 mmol) in THF (20 mL) was added 1M TBAF solution in THF (12 mL). The reaction mixture was stirred under argon at 60° C. for 2 h, then cooled to room temperature, and concentrated. The crude residue was purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-10% MeOH) to afford 2-chloro-5-(2,2,6,6-tetramethyl-4-piperidyl)pyrrolo[2,3-b]pyrazine (450 mg, 56%) as a dark red oil. MS m/z 293.3 [M+H]+;1H NMR (DMSO-d6) δ: 8.36 (s, 1H), 8.16-8.19 (m, 1H), 6.64-6.68 (m, 1H), 5.07-5.12 (m, 1H), 1.72-1.81 (m, 4H), 1.26 (s, 6H), 1.11 (s, 6H). Step 5: A mixture of 2-chloro-5-(2,2,6,6-tetramethyl-4-piperidyl)pyrrolo[2,3-b]pyrazine (50 mg, 0.1708 mmol), 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (85 mg, 0.2052 mmol), PdCl2(dppf) (13 mg, 0.01741 mmol) was purged with argon. Aqueous 2 N K2CO3(0.5 mL, 1 mmol), and 1,4-dioxane (2 mL) were added and the reaction was heated under argon at 100° C. for 16 h. The reaction mixture was then cooled to room temperature and diluted with CH2Cl2and then filtered through a phase separation column and then concentrated. The crude residue was purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-10% MeOH) to afford 2-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5-(2,2,6,6-tetramethyl-4-piperidyl)pyrrolo[2,3-b]pyrazine (55 mg, 59%) as a dark brown oil contaminated with ˜20% of 2-chloro-5-(2,2,6,6-tetramethyl-4-piperidyl)pyrrolo[2,3-b]pyrazine. MS m/z 545.5 [M+H]+. Step 6: To a solution of 2-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-5-(2,2,6,6-tetramethyl-4-piperidyl)pyrrolo[2,3-b]pyrazine (55 mg, 0.10 mmol) in CH2Cl2(1 mL) was added 4N HCl in dioxane (0.5 mL, 2 mmol) and the reaction was stirred at room temperature for 1 h. The yellow solid that precipitated was collected by vacuum filtration, rinsed with CH2Cl2and dried to afford 5-(1H-pyrazol-4-yl)-2-(5-(2,2,6,6-tetramethylpiperidin-4-yl)-5H-pyrrolo[2,3-b]pyrazin-2-yl)phenol hydrochloride (18 mg, 23%). MS m/z 417.4 [M+H]+;1H NMR (DMSO-d6) δ: 9.10 (s, 1H), 8.06-8.19 (m, 4H), 7.20-7.27 (m, 2H), 6.81-6.85 (m, 1H), 5.20-5.24 (m, 1H), 2.33-2.45 (m, 2H), 2.12-2.17 (m, 2H), 1.59 (s, 6H), 1.50 (s, 6H). Using the procedure described for Example 5, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData16MS m/z 437.2 [M + H]+,1H NMR (methanol-d4) δ: 8.82 (d, J = 2.2 Hz, 1H), 8.50 (d,J = 1.3 Hz, 2H), 8.11 (d, J = 3.8 Hz, 1H), 7.88 (dd, J = 11.3, 6.3 Hz, 1H), 7.78 (dd,J = 11.3, 6.6 Hz, 1H), 6.84 (d, J = 3.8 Hz, 1H), 5.34-5.46 (m, 1H), 2.49 (t, J = 13.9 Hz,2H), 2.29 (dd, J = 13.9, 3.8 Hz, 2H), 1.70 (s, 6H), 1.58 (s, 6H); 2 Hs not observed (2 NHs). Example 6 Preparation of Compound 23 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with 4-bromo-3,6-dichloropyridazine (0.227 g, 1.0 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (75.0 mg, 0.1 mmol), and 4 4,4,5,5-tetramethyl-2-vinyl-1,3,2-dioxaborolane (0.171 mL, 1.0 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). Dioxane (6 mL) and 2N aq. K2CO3(1.5 mL, 3.0 mmol) were added and the reaction was heated to 50° C. for 3 h. The reaction was cooled to room temperature, diluted with water (2 mL) and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a EtOAc/hexanes gradient (0-50% EtOAc) to provide 3,6-dichloro-4-vinylpyridazine (0.145 g, 82%). Step 2: A mixture of 3,6-dichloro-4-vinylpyridazine (0.34 g, 1.94 mmol) and 2,2,6,6-tetramethylpiperidin-4-amine (0.72 mL, 4.6 mmol) was dissolved in acetonitrile (5 mL) and the resulting solution was heated to 90° C. for 16 h. The reaction mixture was concentrated and the residue was purified by silica gel column chromatography eluting with a EtOAc/hexanes gradient (0-50% EtOAc) to provide 3-chloro-7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine (0.29 g, 62%). MS m/z 295.4 [M+H]+. Step 3: An oven-dried flask was equipped with a magnetic stir bar and charged with 3-chloro-7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine (0.15 g, 0.5 mmol), 4-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (0.32 mg, 0.76 mmol, prepared in Example 1, step 7), tetrakis(triphenylphosphine)palladium(0) (58 mg, 0.05 mmol), and Na2CO3(160 mg, 1.5 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (10 mL) and water (1.5 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water (2 mL), and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to afford 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine (0.145 mg, 52%) as an orange solid. MS m/z 547.3 [M+H]+. Step 4: 3-(2-(Methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine (20 mg, 0.037 mmol) was dissolved in 1 mL of methanol, then 4 N HCl in 1,4-dioxane (0.5 mL, 2 mmol) was added and the reaction stirred at room temperature for 2 h. The reaction was concentrated and then triturated with 20% MeOH/ether. The precipitate was filtered and dried to afford 5-(1H-pyrazol-4-yl)-2-(7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazin-3-yl)phenol hydrochloride (10 mg, 66%) as a yellow solid. MS m/z 419.5 [M+H]+;1H NMR (methanol-d4) δ: 8.17 (s, 2H), 8.07 (s, 1H), 7.66 (d, J=8.2 Hz, 1H), 7.36 (dd, J=8.2, 1.6 Hz, 1H), 7.27 (d, J=1.6 Hz, 1H), 4.49-4.58 (m, 1H), 3.94 (t, J=7.9 Hz, 2H), 3.42 (dd, J=8.5, 7.3 Hz, 2H), 2.12 (dd, J=13.9, 3.5 Hz, 2H), 2.00 (t, J=13.9 Hz, 2H), 1.61 (s, 6H), 1.55 (s, 6H); 3 Hs not observed (1 OH and 2 NHs). Using the procedure described for Example 6, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData32MS m/z 439.5 [M + H]+;1H NMR (methanol-d4) δ: 8.23 (s, 2H), 7.93 (s, 1H), 7.69-7.76(m, 1H), 7.55-7.64 (m, 1H), 4.55-4.67 (m, 1H), 4.01-4.07 (m, 2H), 3.44 (br s, 2H),2.12-2.19 (m, 2H), 2.02-2.08 (m, 2H), 1.64 (s, 6H), 1.57 (s, 6H); 2 Hs not observed (2 NHs).36MS m/z 409.3 [M + H]+;1H NMR (methanol-d4) δ: 8.17 (s, 2H), 7.89 (d, J = 1.9 Hz,1H), 7.66-7.74 (m, 1H), 7.55-7.61 (m, 1H), 4.48-4.58 (m, 1H), 4.21-4.26 (m, 2H),3.99 (t, J = 7.6 Hz, 2H), 3.39-3.43 (m, 2H), 2.22-2.31 (m, 6H), 2.08-2.16 (m, 2H); 2 Hsnot observed (2 NHs). Example 7 Preparation of Compound 26 Step 1: A mixture of 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-6,7-dihydro-5H-pyrrolo[2,3-c]pyridazine (0.28 g, 0.49 mmol, prepared in Example 6, step 3) and manganese dioxide (0.28 g, 3.21 mmol) in toluene (10 mL) was heated at 125° C. in a sealed tube for 24 h. The reaction mixture was cooled to room temperature, filtered over a small pad of Celite and concentrated. The crude compound was purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to provide 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazine (0.2 g, 71%) as a tan solid. MS m/z 545.4 [M+H]+. Step 2: To a solution of 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazine (0.2 g, 0.36 mmol) in 1,4-dioxane (4 mL) was added 4 N HCl in 1,4-dioxane (0.5 mL, 2 mmol) and the reaction was stirred at room temperature for 2 h. The reaction mixture was concentrated, triturated with 20% MeOH/ether, and the precipitate was filtered and dried to afford 5-(1H-pyrazol-4-yl)-2-(7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazin-3-yl)phenol hydrochloride (120 mg, 78%) as an orange solid. MS m/z 417.4 [M+H]+;1H NMR (methanol-d4) δ: 8.84 (s, 1H), 8.68 (d, J=3.5 Hz, 1H), 8.39 (br s, 2H), 7.74 (d, J=7.9 Hz, 1H), 7.45 (d, J=8.2 Hz, 1H), 7.33-7.39 (m, 1H), 7.14 (d, J=3.5 Hz, 1H), 5.42-5.58 (m, 1H), 2.53 (t, J=13.6 Hz, 2H), 2.42 (dd, J=13.6, 3.2 Hz, 2H), 1.72 (s, 6H), 1.63 (s, 6H); 3 Hs not observed (1 OH and 2 NHs). Using the procedure described for Example 7, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData176MS m/z 418.5 [M + H]+;1H NMR (methanol-d4) δ: 8.64 (s, 1H), 8.09-8.19 (m, 1H),8.02-8.06 (m, 1H), 7.97 (s, 2H), 7.70-7.76 (m, 2H), 6.79 (d, J = 1.8 Hz, 1H), 5.47-5.60(m, 1H), 2.51-2.59 (m, 2H), 2.37 (br d, J = 13.7 Hz, 2H), 1.74 (s,6H), 1.60 (s, 6H); 2Hs not observed (NH and OH). Example 8 Preparation of Compound 17 Step 1: To a solution of methyl 4,6-dichloropyridazine-3-carboxylate (2.05 g, 9.9 mmol) in CH3CN (26 mL) was added a solution of methyl 2-sulfanylacetate (0.90 mL, 10.0 mmol) in CH3CN (8.5 mL) dropwise at 0° C. Upon completion of addition, Et3N (1.40 mL, 10.0 mmol) was added dropwise. The reaction stirred at 0° C. for 15 min. After 15 min, an additional portion of Et3N (1.40 mL, 10.0 mmol) was added and the mixture was allowed to warm to room temperature and stirred overnight. The reaction was diluted with water and concentrated. The mixture was acidified with 4N HCl to pH 3 and the bright yellow solution turned colorless and a white precipitate was formed. The solid was collected by filtration and washed with water to afford methyl 3-chloro-7-hydroxy-thieno[3,2-c]pyridazine-6-carboxylate (2.26 g, 93.1% yield) as white solid. MS m/z 245.1 [M+H]+. Step 2: To a solution of methyl 3-chloro-7-hydroxy-thieno[3,2-c]pyridazine-6-carboxylate (2.26 g, 9.24 mmol) in DMF (30 mL) were added imidazole (1.0 g, 15 mmol) and TIPSCl (2.25 mL, 10 mmol). The mixture was stirred at room temperature for 15 min and then heated to 50° C. for 24 h. Upon completion, the reaction was cooled to room temperature and was then diluted with water and the product was extracted with EtOAc. The combined organic phases were dried over Na2SO4, concentrated and the residue was purified by silica gel column chromatography, eluting with a EtOAc/CH2Cl2gradient (0-5% EtOAc), to provide methyl 3-chloro-7-triisopropylsilyloxy-thieno[3,2-c]pyridazine-6-carboxylate (2.04 g, 55.1% yield) as an off-white solid. Step 3: A mixture of methyl 3-chloro-7-triisopropylsilyloxy-thieno[3,2-c]pyridazine-6-carboxylate (800 mg, 1.9 mmol), 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (prepared in example 1, step 7, 1.00 g, 2.4 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) complex with dichloromethane (85 mg, 0.10 mmol) and K2CO3(830 mg, 6.0 mmol) in a vial was evacuated and backfilled with N2 (repeated 3×), 1,4-dioxane (7 mL) and water (1.8 mL) were added and the mixture was heated at 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water and acidified with 4N HCl. The product was extracted with CH2Cl2(3×). The organic phases were combined, dried over Na2SO4and concentrated. Crude methyl 7-hydroxy-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]thieno[3,2-c]pyridazine-6-carboxylate was used directly in the next step without purification. Step 4: Crude methyl 7-hydroxy-3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]thieno[3,2-c]pyridazine-6-carboxylate obtained above was mixed with 5 N NaOH (3 mL, 15 mmol) and MeOH (15 mL) and heated at 95° C. until complete hydrolysis was observed (6 h). Once complete, the reaction was cooled to room temperature and carefully acidified with 4N HCl to pH 3-4. The intermediate carboxylic acid was extracted with CH2Cl2/MeOH. The organic phase was dried over Na2SO4. Volatiles were removed under reduced pressure and the residue was dissolved in DMSO (10 mL). The mixture was heated at 80° C. for 60 min after which complete decarboxylation of the intermediate α-ketoacid was observed. Upon cooling to room temperature, the mixture was diluted with water and the product was extracted with EtOAc. The organic phase was dried over Na2SO4and the solvent was removed. The residue was purified by silica gel column chromatography eluting with a MeOH/CH2Cl2gradient (0-5% MeOH) to afford 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]thieno[3,2-c]pyridazin-7-ol (0.64 g, 76% yield) as a yellow solid. MS m/z 439.4 [M+H]+. Step 5: To a solution of 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]thieno[3,2-c]pyridazin-7-ol (204 mg, 0.47 mmol) in CH2Cl2(2.5 mL) was added DIPEA (0.16 mL, 0.92 mmol). The reaction was cooled to 0° C. and triflic anhydride (0.09 mL, 0.53 mmol) was added dropwise. The reaction was stirred at 0° C. for 30 min, then diluted with water and the products were extracted with CH2Cl2. The organic phase was dried over Na2SO4, the volatiles were removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with a MeOH/EtOAc gradient (0-5% MeOH) to afford [3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]thieno[3,2-c]pyridazin-7-yl]trifluoromethanesulfonate (0.13 g, 48% yield) as white foam. MS m/z 571.3 [M+H]+. Step 6: An oven-dried flask was equipped with a magnetic stir bar and charged with [3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]thieno[3,2-c]pyridazin-7-yl]trifluoromethanesulfonate (127 mg, 0.22 mmol), 2,2,6,6-tetramethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-dihydropyridine (64 mg, 0.24 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) and (18 mg, 0.02 mmol), K2CO3(100 mg, 0.72 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). Dioxane (1.2 mL) and water (0.8 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water (5 mL), and then extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4and concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with a MeOH (with 2.5% NH4OH)/CH2Cl2gradient (0 to 10% MeOH/NH4OH) to afford 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)thieno[3,2-c]pyridazine (55.6 mg, 45% yield) as brownish foam. MS m/z 560.5 [M+H]+. Step 7: To 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethyl-1,3-dihydropyridin-4-yl)thieno[3,2-c]pyridazine (13.8 mg, 0.025 mmol) was added 4 N HCl in dioxane (0.5 mL, 2 mmol) and MeOH (0.5 mL). The reaction was stirred at room temperature for 30 min and then ˜2 h at 50° C. The volatiles were then removed under reduced pressure, the residue was triturated with Et2O, and the solid was filtered and dried to afford 5-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-1,3-dihydropyridin-4-yl)thieno[3,2-c]pyridazin-3-yl]phenol hydrochloride (7 mg, 60% yield) as a yellow solid. MS m/z 432.4 [M+H]+;1H NMR (methanol-d4) δ: 9.37 (s, 1H), 8.49 (s, 1H), 8.22 (s, 2H), 7.93 (d, J=8.2 Hz, 1H), 7.42 (dd, J=8.2, 1.6 Hz, 1H), 7.36 (d, J=1.6 Hz, 1H), 7.20 (s, 1H), 3.56-3.77 (m, 2H), 1.72 (s, 6H), 1.65 (s, 6H); 3 Hs not observed (2 NHs and OH). Using the procedure described for Example 8, above, additional compounds described herein may be prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData15MS m/z 376.3 [M + H]+;1H NMR (DMSO-d6) δ: 9.41 (br s, 2H), 9.38 (s, 1H), 8.40 (s,1H), 8.26 (s, 2H), 8.11 (d, J = 9.1 Hz, 1H), 7.58-7.68 (m, 1H), 7.40 (d, J = 1.9 Hz, 1H),7.37-7.39 (m, 1H), 3.84-4.03 (m, 2H), 3.41-3.51 (m, 2H), 2.87-3.02 (m, 2H); 1H notobserved (NH or OH).18MS m/z 402.3 [M + H]+;1H NMR (methanol-d4) δ: 9.47 (s, 1H), 8.55 (s, 1H), 8.36 (s,2H), 7.91 (d, J = 8.2 Hz, 1H), 7.59 (dt, J = 6.3, 1.6 Hz, 1H), 7.48 (dd, J = 8.2, 1.6 Hz,1H), 7.40 (d, J = 1.6 Hz, 1H), 4.57 (dd, J = 6.6, 6.0 Hz, 1H), 4.45 (dd, J = 6.6, 5.0 Hz,1H), 3.38-3.46 (m, 1H), 2.84-3.01 (m, 1H), 2.40-2.53 (m, 2H), 2.24-2.37 (m, 1H),2.06-2.19 (m, 1H); 3 Hs not observed (2 NHs and OH).25MS m/z 418.4 [M + H]+;1H NMR (methanol-d4) δ: 9.51 (s, 1H), 8.66 (s, 1H), 8.43 (s,2H), 7.91 (d, J = 8.2 Hz, 1H), 7.49 (dd, J = 8.2, 1.6 Hz, 1H), 7.44-7.47 (m, 1H), 7.41 (d,J = 1.6 Hz, 1H), 4.37 (d, J = 5.7 Hz, 1H), 4.16 (d, J = 12.6 Hz, 1H), 4.12 (dd, J = 12.6, 1.9Hz, 1H), 4.07 (dd, J = 12.6, 1.9 Hz, 1H), 3.98 (s, 2H), 3.39-3.48 (m, 1H), 3.18 (dd,J = 18.3, 1.9 Hz, 1H); 3 Hs not observed (2 NHs and OH). Example 9 Preparation of Compound 24 Step 1: A solution of 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethyl-1,3-dihydropyridin-4-yl)thieno[3,2-c]pyridazine (50 mg, 0.089 mmol, from Example 8, step 6) in MeOH (3 mL) was hydrogenated in a Parr shaker over Pt2O (100 mg, 0.44 mmol) for 72 h at 50 psi of H2. The catalyst was then filtered and washed with MeOH. The mother liquor was concentrated and the residue, crude 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethyl-4-piperidyl)thieno[3,2-c]pyridazine, was taken directly into the next step. Step 2: A solution of crude 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-7-(2,2,6,6-tetramethyl-4-piperidyl)thieno[3,2-c]pyridazine obtained from step 1 above in MeOH (1 mL) was treated with 4N HCl in dioxane (0.5 mL, 2 mmol). The reaction was stirred at room temperature for 30 min and then ˜2 h at 50° C. The volatiles were then removed under reduced pressure, the residue was triturated with Et2O, and the solid was filtered and washed with Et2O. The crude product was purified by prep HPLC using the polar method (20-65% CH3CN/H2O). Upon concentration of the desired fractions and treatment of the residue with 4N HCl in dioxane (0.5 mL, 2 mmol), 5-(1H-pyrazol-4-yl)-2-(7-(2,2,6,6-tetramethylpiperidin-4-yl)thieno[3,2-c]pyridazin-3-yl)phenol dihydrochloride (2.4 mg, 6% yield) was obtained as a yellow solid. MS m/z 434.3 [M+H]+;1H NMR (DMSO-d6) δ: 9.32 (br s 1H), 9.30 (s, 1H), 8.19 (s, 2H), 8.15 (br. s, 1H), 8.12 (s, 1H), 8.05 (d, J=8.8 Hz, 1H), 7.27-7.36 (m, 2H), 3.91-4.00 (m, 1H), 2.18 (dd, J=12.3, 2.5 Hz, 2H), 1.89-2.07 (m, 2H), 1.60 (s, 6H), 1.50 (s, 6H); 1H not observed (NH or OH). Example 10 Preparation of Compound 98 Step 1: Methyl 3,6-dichloropyridazine-4-carboxylate (500 mg, 2.42 mmol), (2,2,6,6-tetramethyl-4-piperidyl)hydrazine dihydrochloride (590 mg, 2.84 mmol), and DIPEA (1.3 mL, 7.26 mmol) were mixed in MeOH (2 mL) and heated to 70° C. overnight. UPLC showed 2 peaks with the desired mass in an approximately 2:3 ratio. The solvent was removed under reduced pressure and the residue was purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0% to 30%) to provide 5-chloro-2-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-ol (230 mg, 31%) as dark violet solid and 5-chloro-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-ol (200 mg, 13.4%) as a brownish-orange solid. MS m/z 310.8 [M+H]+;1H NMR (methanol-d4) δ: 7.84 (s, 1H), 5.24 (tt, J=12.3, 3.8 Hz, 1H), 2.30 (dd, J=14.2, 12.3 Hz, 2H), 2.05 (dd, J=14.2, 3.8 Hz, 2H), 1.66 (s, 6H), 1.54 (s, 6H); 2 Hs not observed (NH and OH). Step 2: An oven-dried flask was equipped with a magnetic stir bar and charged with 5-chloro-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-ol (500 mg, 1.61 mmol), triisopropyl-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy]silane (775 mg, 1.78 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (121 mg, 0.16 mmol) and K2CO3(451 mg, 3.23 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). Dioxane (2 mL) and water (0.5 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provided 5-[2-(methoxymethoxy)-4-triisopropylsilyloxy-phenyl]-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-ol (200 mg, 21%) as a brownish solid. MS m/z 584.4 [M+H]+. Step 3: To a suspension of 5-[2-(methoxymethoxy)-4-triisopropylsilyloxy-phenyl]-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-ol (200 mg, 0.34 mmol) in CH2Cl2(4 mL) were added N,N-bis(trifluoromethylsulfonyl)aniline (247 mg, 0.69 mmol) and Et3N (0.15 mL, 1.03 mmol) and the reaction was stirred at room temperature for 16 h. The reaction was concentrated under reduced pressure, and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide [5-[2-(methoxymethoxy)-4-triisopropylsilyloxy-phenyl]-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-yl] trifluoromethanesulfonate (236 mg, 96%) as a tan solid. MS m/z 716.6 [M+H]+. Step 4: To a mixture of [5-[2-(methoxymethoxy)-4-triisopropylsilyloxy-phenyl]-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-yl] trifluoromethanesulfonate (236 mg, 0.33 mmol), dppf (38 mg, 0.066 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (49 mg, 0.066 mmol), and ammonium formate (104 mg, 1.65 mmol) in dry THF (4 ml) was added Et3N (0.23 mL, 1.65 mmol). The mixture was purged with argon and then heated at 60° C. for 6 h in a sealed tube. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 15% MeOH) to provide triisopropyl-[3-(methoxymethoxy)-4-[1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-5-yl]phenoxy]silane (170 mg, 91%) as a grey solid. MS m/z 568.5 [M+H]+;1H NMR (methanol-d4) δ: 8.45 (s, 1H), 8.32 (s, 1H), 7.67 (d, J=8.2 Hz, 1H), 6.96 (d, J=2.5 Hz, 1H), 6.77 (dd, J=8.8, 2.5 Hz, 1H), 5.74 (tt, J=12.6, 3.8 Hz, 1H), 5.22 (s, 2H), 3.42 (s, 3H), 2.06-2.33 (m, 4H), 1.56 (s, 6H), 1.39 (s, 6H), 1.35 (spt, J=7.6 Hz, 3H), 1.19 (d, J=7.6 Hz, 18H); 1H not observed (NH). Step 5: To a solution of triisopropyl-[3-(methoxymethoxy)-4-[1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-5-yl]phenoxy]silane (273 mg, 0.48 mmol) in THF (4 mL) was added 1.0 M TBAF in THF (0.53 mmol, 0.53 mL) and the reaction was stirred at room temperature for 30 min. The solvent was removed under reduced pressure, and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide 3-(methoxymethoxy)-4-[1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-5-yl]phenol (101 mg, 51%) as a clear solid. MS m/z 412.4 [M+H]+. Step 6: To a suspension of 3-(methoxymethoxy)-4-[1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-5-yl]phenol (101 mg, 0.25 mmol) in CH2Cl2(2 mL) were added N,N-bis(trifluoromethylsulfonyl)aniline (133 mg, 0.37 mmol) and Et3N (0.104 mL, 0.74 mmol). The reaction mixture was stirred at room temperature for 16 h. UPLC showed complete conversion. The solvent was removed under reduced pressure and the residue was purified by column chromatography on silica gel, eluting with a EtOAc/hexanes gradient (20 to 100% EtOAc) to provide [3-(methoxymethoxy)-4-[1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-5-yl]phenyl] trifluoromethanesulfonate (121 mg, 91%) as a clear solid. MS m/z 544.4 [M+H]+;1H NMR (methanol-d4) δ: 8.53 (s, 1H), 8.38 (s, 1H), 7.94 (d, J=8.5 Hz, 1H), 7.40 (d, J=2.5 Hz, 1H), 7.24 (dd, J=8.5, 2.5 Hz, 1H), 5.79 (tt, J=12.6, 3.8 Hz, 1H), 5.31 (s, 2H), 3.44 (s, 3H), 2.39 (t, J=13.6 Hz, 2H), 2.28 (dd, J=13.6, 3.8 Hz, 2H), 1.66 (s, 6H), 1.50 (s, 6H); 1H not observed (NH). Step 7: An oven-dried flask was equipped with a magnetic stir bar and charged with [3-(methoxymethoxy)-4-[1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-5-yl]phenyl]trifluoromethanesulfonate (51 mg, 0.094 mmol), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (23 mg, 0.11 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(I) (7 mg, 0.009 mmol), and K2CO3(26 mg, 0.19 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (1 mL) and water (0.25 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide 5-[2-(methoxymethoxy)-4-(1-methylpyrazol-4-yl)phenyl]-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazine (40 mg, 89%) as an orange solid. MS m/z 476.5 [M+H]+. Step 8: To a solution of 5-[2-(methoxymethoxy)-4-(1-methylpyrazol-4-yl)phenyl]-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazine (40 mg, 0.084 mmol) in 1 mL of CH2Cl2and 2 drops of MeOH was added 4 N HCl in 1,4-dioxane (42 μL, 0.17 mmol) and the reaction was stirred for 5 h until UPLC showed complete consumption of the starting material. The solvent was removed under reduced pressure and the residue was triturated in Et2O, and the precipitate was collected by filtration. The solid was further washed with diethyl ether and dried to afford 5-(1-methylpyrazol-4-yl)-2-[1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-5-yl]phenol dihydrochloride (32 mg, 75%) as a yellow solid. MS m/z 432.5 [M+H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.66 (s, 1H), 8.16 (s, 1H), 7.99 (s, 1H), 7.86 (d, J=7.9 Hz, 1H), 7.35 (dd, J=8.2, 1.5 Hz, 1H), 7.29 (d, J=1.5 Hz, 1H), 5.74 (tt, J=12.6, 4.0 Hz, 1H), 4.00 (s, 3H), 2.52 (t, J=13.8 Hz, 2H), 2.44 (dd, J=13.8, 4.0 Hz, 2H), 1.76 (s, 6H), 1.62 (s, 6H); 2 NH not observed (NH and OH). Using the procedure described for Example 10, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData21MS m/z 362.4 [M + H]+;1H NMR (methanol-d4) δ: 9.12 (s, 1H), 8.68 (s, 1H), 8.42 (s,2H), 7.89 (d, J = 8.2 Hz, 1H), 7.44 (dd, J = 8.2, 1.9 Hz, 1H), 7.38 (d, J = 1.6 Hz, 1H),5.52 (tt, J = 10.4, 4.4 Hz, 1H), 3.67-3.72 (m, 2H), 3.43 (td, J = 12.3, 3.2 Hz, 2H), 2.58-2.70 (m, 2H), 2.46-2.55 (m, 2H); 3 Hs not observed (2 NHs and OH).102MS m/z 435.5 [M + H]+;1H NMR (methanol-d4) δ: 8.84 (s, 1H), 8.43 (s, 1H), 8.05 (s,1H), 7.97 (d, J = 8.2 Hz, 1H), 7.89 (s, 1H), 7.25 (d, J = 8.2 Hz, 1H), 7.23 (s, 1H), 5.78(tt, J = 12.4, 3.4 Hz, 1H), 2.51 (t, J = 13.7 Hz, 2H), 2.40 (dd, J = 13.7, 3.4 Hz, 2H), 1.76(s, 6H), 1.60 (s, 6H); 2 Hs not observed (NH and OH).148MS m/z 450.4 [M + H]+;1H NMR (methanol-d4) δ: 8.81 (s, 1H), 8.40 (s, 1H), 7.77-7.86(m, 2H), 7.25-7.32 (m, 1H), 6.91-7.00 (m, 1H), 5.56-5.72 (m, 1H), 3.26 (s, 3H), 2.42(t, J = 13.4 Hz, 2H), 2.28 (dd, J = 13.4, 2.8 Hz, 2H), 1.65 (s, 6H), 1.50 (s, 6H); 2 Hs notobserved (OH and NH).161MS m/z 436.5 [M + H]+;1H NMR (methanol-d4) δ: 8.81 (d, J = 8.5 Hz, 1H), 8.38 (s,1H), 7.86-8.07 (m, 2H), 7.23-7.31 (m, 1H), 7.04 (d, J = 8.6 Hz, 1H), 5.61-5.74 (m,1H), 1.98-2.18 (m, 4H), 1.48 (s, 6H), 1.30 (s, 6H), 3 Hs not observed (2 NHs andOH). Example 11 Preparation of Compound 22 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with 5-chloro-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-ol (186 mg, 0.60 mmol), 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (prepared in example 1, step 7, 298.5 mg, 0.72 mmol), [1,1-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (45 mg, 0.06 mmol), and K2CO3(252 mg, 1.80 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (2 mL) and water (0.5 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-30% MeOH) to provide 5-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-3-ol (229 mg, 68%) as a dark yellow solid. MS m/z 562.5 [M+H]+. Step 2: To a solution of 5-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-1-(2,2,6,6-tetramethyl-4-piperidyl)pyrazolo[3,4-c]pyridazin-3-ol (229 mg, 0.41 mmol) in CH2Cl2(4 mL) was added N,N-bis(trifluoromethylsulfonyl)aniline (294 mg, 0.82 mmol) and Et3N (0.17 mL, 1.22 mmol) and the reaction was stirred at room temperature for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with CH2Cl2(3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-25% MeOH) to provide 5-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-3-yl trifluoromethanesulfonate (166 mg, 59%) as a clear solid. MS m/z 694.3 [M+H]+;1H NMR (acetone-d6) δ: 8.50 (s, 1H), 8.17 (d, J=0.6 Hz, 1H), 7.91 (d, J=8.2 Hz, 1H), 7.83 (d, J=0.6 Hz, 1H), 7.47 (d, J=1.6 Hz, 1H), 7.37 (dd, J=8.2, 1.6 Hz, 1H), 5.73 (tt, J=12.3, 4.0 Hz, 1H), 5.34 (dd, J=9.8, 2.5 Hz, 1H), 5.27 (s, 2H), 3.85-3.92 (m, 1H), 3.54-3.64 (m, 1H), 3.30 (s, 3H), 2.15-2.27 (m, 4H), 1.60-1.68 (m, 2H), 1.54 (s, 6H), 1.45-1.52 (m, 4H), 1.37 (s, 6H), 1H not observed (NH). Step 3: To a solution of 5-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-3-yl trifluoromethanesulfonate (166 mg, 0.24 mmol), Pd(OAc)2(11 mg, 0.048 mmol), dppf (27 mg, 0.048 mmol), and Et3N (0.17 mL, 1.2 mmol) in dry THF (0.5 ml) was added ammonium formate (77 mg, 1.2 mmol). The mixture was purged with argon and heated at 60° C. for 2.5 h in a sealed tube. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 20% MeOH) to provide 5-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazine (115 mg, 88%) as a clear foam. MS m/z 546.5 [M+H]+. Step 4: To a solution of 5-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazine (115 mg, 0.21 mmol) in 1 ml CH2Cl2and 1 drop of MeOH was added 4N HCl in dioxane (0.11 mL, 0.44 mmol) and the reaction was stirred at room temperature for 2 h. The yellow solid that precipitated was collected by vacuum filtration, rinsed with CH2Cl2, Et2O and dried to afford 5-(1H-pyrazol-4-yl)-2-(1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-pyrazolo[3,4-c]pyridazin-5-yl)phenol hydrochloride (78 mg, 89%) as a yellow solid: MS m/z 418.5 [M+H]+;1H NMR (methanol-d4) δ: 9.20 (s, 1H), 8.75 (s, 1H), 8.62 (s, 2H), 7.91 (d, J=8.2 Hz, 1H), 7.49 (dd, J=8.2, 1.9 Hz, 1H), 7.43 (d, J=1.6 Hz, 1H), 5.75 (tt, J=12.0, 4.1 Hz, 1H), 2.53 (t, J=14.2 Hz, 2H), 2.46 (dd, J=14.2, 4.1 Hz, 2H), 1.76 (s, 6H), 1.63 (s, 6H); 3 Hs not observed (2 NHs and OH). Using the procedure described for Example 11, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData165MS m/z 429.3 [M + H]+;1H NMR (methanol-d4) δ: 8.90 (s, 1H), 8.61 (d, J = 4.6 Hz,2H), 8.41 (s, 1H), 8.06 (d, J = 8.5 Hz, 1H), 7.79 (br d, J = 4.9 Hz, 2H), 7.39 (s, 1H), 7.35(d, J = 8.2 Hz, 1H), 5.61-5.75 (m, 1H), 1.94-2.16 (m, 4H), 1.45 (s, 6H),1.26 (s, 6H); 2Hs not observed (NH and OH).166MS m/z 429.3 [M + H]+;1H NMR (methanol-d4) δ: 9.31 (br s, 1H), 9.01-9.10 (m, 2H),8.92 (br d, J = 3.1 Hz, 1H), 8.57 (s, 1H), 8.17-8.31 (m, 2H), 7.56 (br s, 2H), 5.71-5.88(m, 1H), 2.49-2.60 (m, 2H), 2.35-2.48 (m, 2H), 1.77 (s, 6H), 1.63 (s, 6H); 2 Hs notobserved (NH and OH).167MS m/z 430.3 [M + H]+;1H NMR (methanol-d4) δ: 9.19 (s, 1H), 9.16 (br s, 2H), 8.93(s, 1H), 8.46 (s, 1H), 8.17 (d, J = 8.5 Hz, 1H), 7.35-7.50 (m, 2H), 5.76-5.89 (m, 1H),2.49-2.56 (m, 2H), 2.42 (br dd, J = 13.6, 3.2 Hz, 2H), 1.77 (s, 6H), 1.61 (s, 6H); 2 Hsnot observed (NH and OH).169MS m/z 432.4 [M + H]+;1H NMR (methanol-d4) δ: 8.83 (s, 1H), 8.39 (s, 1H), 7.93-8.05(m, 1H), 7.65 (s, 1H), 7.35-7.54 (m, 2H),6.65-6.78 (m, 1H), 5.54-5.83 (m, 1H), 3.95(s, 3H), 2.12-2.46 (m, 4H), 1.61 (br s, 6H), 1.44 (br s, 6H); 2 Hs not observed (NHand OH).174MS m/z 419.4 [M + H]+;1H NMR (methanol-d4) δ: 8.77-8.95 (m, 1H), 8.38 (br s, 1H),8.01-8.14 (m, 1H), 7.97 (s, 2H), 7.55-7.78 (m, 2H), 5.58-5.73 (m, 1H), 1.94-2.22 (m,4H), 1.46 (s, 6H), 1.26 (s, 6H); 2Hs not observed (NH and OH). Example 12 Preparation of Compound 19 Step 1: 3,6-Dibromopyrazine-2-carbaldehyde (340 mg, 1.28 mmol) and benzyl 4-hydrazinopiperidine-1-carboxylate dihydrochloride (412 mg, 1.28 mmol) were mixed in DMF (3 mL) and stirred at room temperature for 5 min. The reaction mixture was partitioned between EtOAc and water. The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and the residue was purified by column chromatography on silica gel, eluting with a EtOAc/hexanes gradient (0% to 30% EtOAc) to provide benzyl 4-[(2E)-2-[(3,6-dibromopyrazin-2-yl)methylene]hydrazino]piperidine-1-carboxylate (550 mg, 87%) as a yellow solid. MS m/z 496.0, 498.0, 500.0 [M+H]+;1H NMR (acetone-d6) δ: 8.27-8.33 (m, 1H), 8.17 (d, J=4.7 Hz, 1H), 7.90 (s, 1H), 7.36-7.45 (m, 4H), 7.29-7.36 (m, 1H), 5.14 (s, 2H), 4.05-4.15 (m, 2H), 3.64-3.74 (m, 1H), 2.08-2.10 (m, 2H), 2.01-2.05 (m, 2H), 1.49-1.64 (m, 2H). Step 2: A solution of benzyl 4-[(2E)-2-[(3,6-dibromopyrazin-2-yl)methylene]hydrazino]piperidine-1-carboxylate (550 mg, 1.11 mmol) in CH3CN (2 mL) was heated in a microwave at 200° C. for 1 h. The solvent was removed under reduced pressure and the residue was purified by column chromatography on silica gel, eluting with a EtOAc/CH2Cl2gradient (0% to 60% EtOAc) to provide benzyl 4-(5-bromo-1H-pyrazolo[3,4-b]pyrazin-1-yl)piperidine-1-carboxylate (78 mg, 16%). MS m/z 416.0, 418.0 [M+H]+. Step 3: An oven-dried flask was equipped with a magnetic stir bar and charged with benzyl 4-(5-bromopyrazolo[3,4-b]pyrazin-1-yl)piperidine-1-carboxylate (78 mg, 0.19 mmol), 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (prepared in Example 1, step 7, 93 mg, 0.23 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(I) (14 mg, 0.019 mmol) and K2CO3(78 mg, 0.56 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (2 mL) and water (0.5 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a EtOAc/CH2Cl2gradient (0-60% EtOAc) to provide benzyl 4-(5-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazolo[3,4-b]pyrazin-1-yl)piperidine-1-carboxylate (69 mg, 59%) as brownish solid. MS m/z 624.3 [M+H]+; Step 4: To a solution of benzyl 4-[5-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]pyrazolo[3,4-b]pyrazin-1-yl]piperidine-1-carboxylate (69 mg, 0.11 mmol) in EtOH (3 mL) was added 10% Pd/C (10 mg) and the reaction mixture was hydrogenated under 1 atm of H2for 16 h. The catalyst was removed by filtration, the solvent was evaporated under reduced pressure, and the residue was purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-30% MeOH). 5-[2-(Methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-1-(4-piperidyl)pyrazolo[3,4-b]pyrazine (15 mg, 28%) was obtained as a white solid. MS m/z 490.4 [M+H]+; Step 5: To a solution of 5-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-1-(4-piperidyl)pyrazolo[3,4-b]pyrazine (15 mg, 0.031 mmol) in CH2Cl2(1 mL, with one drop of MeOH) was added 4 N HCl in 1,4-dioxane (15 μL, 0.06 mmol) and the reaction was stirred at room temperature for 2 h. The precipitate was collected by filtration, washed with Et2O (3×) and dried to provide 2-[1-(4-piperidyl)pyrazolo[3,4-b]pyrazin-5-yl]-5-(1H-pyrazol-4-yl)phenol; hydrochloride (8 mg, 66%) as a yellow solid. MS m/z 362.3 [M+H]+;1H NMR (DMSO-d6) δ:11.68 (br s, 1H), 10.09 (br s, 1H), 9.38 (d, J=1.6 Hz, 1H), 8.95 (br s, 1H), 8.68 (br s, 1H), 8.56 (s, 1H), 8.14 (br s, 1H), 8.05 (dd, J=8.8, 1.6 Hz, 1H), 7.27 (s, 2H), 5.18-5.28 (m, 1H), 3.58-3.66 (m, 1H), 3.42-3.53 (m, 1H), 3.17-3.39 (m, 2H), 2.76-2.88 (m, 1H), 2.34-2.45 (m, 1H), 2.14-2.31 (m, 2H). Example 13 Preparation of Compound 80 Step 1: To a suspension of 3,6-dichloropyridazin-4-amine (2.0 g, 12.2 mmol) in 1-decanol (3.5 mL) were added 2,2,6,6-tetramethylpiperidin-4-amine (2.3 mL, 1.1 equiv.) and N,N-diisopropylethylamine (3 mL, 1.4 equiv.) in a 60 mL sealed screw-cap tube. The reaction was stirred at 150° C. for 72 h after which the partially solidified reaction mixture was transferred to a round-bottom flask with the aid of methanol. The organic components were concentrated to afford a thick oil, which was rinsed with hexanes to remove 1-decanol (this may lead to solidification). The crude product was purified by column chromatography, eluting with a MeOH (with 2.5% NH4OH)/CH2Cl2gradient (5 to 30% MeOH/NH4OH). The first compound to elute was unreacted starting material 3,6-dichloropyridazin-4-amine. Then, a mixture of 6-chloro-N3-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,4-diamine and 6-chloro-N3-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,5-diamine elute (they co-elute), followed by N3,N6-bis(2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,4,6-triamine. Column chromatography generally results in a 50-60% yield of 6-chloro-N3-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,4-diamine and 6-chloro-N3-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,5-diamine (2:1 ratio). The mixture of 6-chloro-N3-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,4-diamine and 6-chloro-N3-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,5-diamine (2.7 g, 77%) obtained after chromatography was used in the next step without further purification. Step 2: To a solution of a mixture of 6-chloro-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,4-diamine and 6-chloro-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,5-diamine (2.7 g, 9.5 mmol) in AcOH (8 mL) was added NaNO2(1.3 g, 19 mmol) in portions and the mixture was stirred at room temperature for 1 h. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate slowly until reaching pH˜7. The aqueous layer was extracted with ethyl acetate three times. The organic layer was dried over Na2SO4, filtered, and concentrated in vacuo. The crude product was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to yield 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (1.22 g, 43%) as brownish solid: MS m/z 295.8 [M+H]+;1H NMR (500 MHz, methanol-d4) δ: 8.56 (s, 1H), 5.76 (tt, J=12.6, 4.1 Hz, 1H), 2.30 (dd, J=12.6, 4.1 Hz, 2H), 2.23 (t, J=12.6 Hz, 2H), 1.47 (s, 6H), 1.32 (s, 6H). Step 3: An oven-dried flask was equipped with a magnetic stir bar and charged with 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (1.11 g, 3.77 mmol), triisopropyl(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)silane (1.81 g, 4.15 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (138 mg, 0.19 mmol) and K2CO3(1.56 g, 11.31 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (18 mL) and water (4 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water (5 mL) and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4and concentrated under reduced pressure, to provide crude 6-(2-(methoxymethoxy)-4-((triisopropylsilyl)oxy)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (1.67 g, 78%) as a brown solid which was used in the next step without further purification. Step 4: To a solution of 6-(2-(methoxymethoxy)-4-((triisopropylsilyl)oxy)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (1.67 g, 2.94 mmol) in THF (10 mL) was added 1.0 M TBAF in THF (3.1 mL, 3.1 mmol). The reaction mixture was stirred at room temperature for 1 h until TLC showed complete consumption of the starting material. The solvent was removed under reduced pressure and the crude product was purified by column chromatography on silica gel, eluting with a EtOAc/hexanes gradient (10-80% EtOAc) to yield 3-(methoxymethoxy)-4-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol (985 mg, 81%) as a tan oil. To a solution of 3-(methoxymethoxy)-4-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol (985 mg, 2.39 mmol) in CH2Cl2(8 mL) was added N,N-bis(trifluoromethylsulfonyl)aniline (1.71 g, 4.78 mmol) and Et3N (1.0 mL, 7.2 mmol). The reaction was stirred for 5 h until UPLC showed complete conversion. The solvent was removed under reduced pressure and the residue was purified by column chromatography on silica gel, eluting with a EtOAc/hexanes gradient (0-60% EtOAc) to give 3-(methoxymethoxy)-4-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenyl trifluoromethanesulfonate (20 mg, 71%). MS m/z 545.6 [M+H]+;1H NMR (acetone-d6) δ: 8.68 (s, 1H), 8.04 (d, J=8.5 Hz, 1H), 7.35 (d, J=2.5 Hz, 1H), 7.21 (dd, J=8.8, 2.5 Hz, 1H), 5.67 (tt, J=11.2, 5.4 Hz, 1H), 5.31 (s, 2H), 3.35 (s, 3H), 2.04-2.27 (m, 4H), 1.36 (s, 6H), 1.16 (s, 6H), 1H not observed (NH). Step 5: An oven-dried flask was equipped with a magnetic stir bar and charged with [3-(methoxymethoxy)-4-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenyl]trifluoromethanesulfonate (50 mg, 0.092 mmol), tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrole-1-carboxylate (32 mg, 0.11 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(I) (7 mg, 0.009 mmol) and K2CO3(26 mg, 0.18 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (18 mL) and water (4 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water (5 mL), and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide tert-butyl 3-[3-(methoxymethoxy)-4-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenyl]pyrrole-1-carboxylate (40 mg, 78%) as an orange solid. MS m/z 562.3 [M+H]+. Step 6: To a solution of tert-butyl 3-[3-(methoxymethoxy)-4-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenyl]pyrrole-1-carboxylate (40 mg, 0.071 mmol) in CH2Cl2(2 mL) plus 2 drops of MeOH was added 4 N HCl in 1,4-dioxane (36 μL). The reaction was stirred for 5 h until UPLC showed complete consumption of the starting material. The solvents were removed under reduced pressure and the product was purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (with 2.5% NH4OH) (5 to 30% MeOH/NH4OH) to provide 5-(1H-pyrrol-3-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol (22 mg, 74%) as an orange solid. MS m/z 418.5 [M+H]+;1H NMR (methanol-d4) δ: 9.00 (s, 1H), 7.95 (d, J=8.2 Hz, 1H), 7.25 (d, J=1.6 Hz, 1H), 7.22-7.24 (m, 1H), 7.18 (d, J=1.6 Hz, 1H), 6.83 (dd, J=2.8, 1.9 Hz, 1H), 6.52 (dd, J=2.8, 1.6 Hz, 1H), 5.89 (tt, J=10.4, 5.5 Hz, 1H), 2.47-2.68 (m, 4H), 1.72 (s, 6H), 1.57 (s, 6H); 3 Hs not observed (2 NHs and OH). Using the procedure described for Example 13, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData37MS m/z 460.6 [M + H]+;1H NMR (methanol-d4) δ: 9.15 (s, 1H), 8.20 (d, J = 8.2 Hz,1H), 7.82 (d, J = 6.9 Hz, 1H), 7.41 (dd, J = 8.2, 1.9 Hz, 1H), 7.39 (d, J = 1.9 Hz, 1H),6.91 (d, J = 2.2 Hz, 1H), 6.84 (dd, J = 6.9, 2.2 Hz, 1H), 5.99 (tt, J = 10.4, 4.7 Hz, 1H),3.67 (s, 3H), 2.64-2.74 (m, 4H), 1.79 (s, 6H), 1.64 (s, 6H); 2 Hs not observed (NH andOH).38MS m/z 445.5 [M + H]+;1H NMR (DMSO-d6) δ: 9.17 (s, 1H), 8.97-9.06 (m, 1H), 8.15(br. s, 1H), 8.12 (d, J = 8.2 Hz, 1H), 7.49-7.61 (m, 2H), 7.29 (d, J = 1.6 Hz, 1H), 7.27 (s,1H), 6.80-6.96 (m, 2H), 5.93 (tt, J = 12.0, 4.4 Hz, 1H), 2.53-2.67 (m, 4H), 1.66 (s, 6H),1.53 (s, 6H); 1H not observed (NH).39MS m/z 433.5 [M + H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.14 (s, 1H), 8.08 (d,J = 7.9 Hz, 1H), 8.00 (s, 1H), 7.31 (dd, J = 8.8, 1.9 Hz, 1H), 7.28 (d, J = 1.9 Hz, 1H), 5.97(tt, J = 12.3, 4.0 Hz, 1H), 4.01 (s, 3H), 2.56-2.76 (m, 4H), 1.79 (s, 6H), 1.64 (s, 6H), 2Hs not observed (NH and OH).46MS m/z 447.6 [M + H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.21 (d, J = 0.9 Hz,1H), 8.07 (d, J = 8.2 Hz, 1H), 8.02 (s, 1H), 7.32 (dd, J = 8.2, 1.9 Hz, 1H), 7.29 (d, J = 1.9Hz, 1H), 5.97 (tt, J = 10.1, 6.3 Hz, 1H), 4.30 (q, J = 7.3 Hz, 2H), 2.55-2.76 (m, 4H), 1.79(s, 6H), 1.64 (s, 6H), 1.55 (t, J = 7.3 Hz, 3H), 2 Hs not observed (NH and OH).47MS m/z 461.6 [M + H]+;1H NMR (methanol-d4) δ: 9.03 (s, 1H), 8.11 (d, J = 0.9 Hz,1H), 8.05 (d, J = 8.5 Hz, 1H), 7.92 (d, J = 0.9 Hz, 1H), 7.29 (dd, J = 8.2, 1.9 Hz, 1H),7.26 (d, J = 1.9 Hz, 1H), 5.82 (tt, J = 12.3, 4.6 Hz, 1H), 4.19 (t, J = 7.3 Hz, 2H), 2.23-2.45(m, 4H), 1.95 (sxt, J = 7.3 Hz, 2H), 1.54 (s, 6H), 1.38 (s, 6H), 0.97 (t, J = 7.3 Hz, 3H), 2Hs not observed (NH and OH).48MS m/z 419.5 [M + H]+;1H NMR (methanol-d4) δ: 9.15 (s, 1H), 8.20 (d, J = 8.8 Hz,1H), 8.04 (s, 1H), 7.45-7.57 (m, 2H), 7.00 (s, 1H), 5.97 (tt, J = 12.1, 4.7 Hz, 1H), 2.59-2.76 (m, 4H), 1.79 (s, 6H), 1.65 (s, 6H), 3 Hs not observed (2 NHs and OH).50MS m/z 433.5 [M + H]+;1H NMR (methanol-d4) δ: 9.19 (s, 1H), 8.26 (d, J = 8.8 Hz,1H), 7.95-8.06 (m, 1H), 7.21-7.37 (m, 2H), 6.72-6.81 (m, 1H), 5.99 (tt, J = 11.3, 5.5Hz, 1H), 4.11 (s, 3H), 2.59-2.76 (m, 4H), 1.80 (s, 6H), 1.65 (s, 6H); 2 Hs not observed(NH and OH).51MS m/z 433.5 [M + H]+;1H NMR (methanol-d4) δ: 9.20 (s, 1H), 8.14 (dd, J = 7.6, 0.9Hz, 1H), 7.91 (d, J = 2.5 Hz, 1H), 7.54 (d, J = 1.9 Hz, 1H), 7.52 (s, 1H),6 .90 (d, J = 2.5Hz, 1H), 5.98 (tt, J = 10.7, 5.7 Hz, 1H), 4.09 (s, 3H), 2.59-2.78 (m, 4H), 1.79 (s, 6H),1.66 (s, 6H), 2 Hs not observed (NH and OH).65MS m/z 430.5 [M + H]+;1H NMR (methanol-d4) δ: 9.29 (s, 1H), 9.18 (s, 1H), 9.03 (s,1H), 8.90 (s, 1H), 8.30 (s, 1H), 8.23 (s, 1H), 7.54 (s, 2H), 5.96 (s, 1H), 2.52-2.68 (m,4H), 1.78 (s, 6H), 1.64 (s, 6H); 2 Hs not observed (NH and OH).66MS m/z 430.4 [M + H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.82 (d, J = 6.9 Hz,2H), 8.38 (d, J = 6.9 Hz, 2H), 8.24 (d, J = 7.9 Hz, 1H), 7.56-7.65 (m, 2H), 5.88-6.02(m, 1H), 2.53-2.63 (m, 4H), 1.68 (s, 6H), 1.54 (s, 6H); 2 Hs not observed (NH andOH).69MS m/z 436.5 [M + H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.17 (d, J = 0.6 Hz,1H), 8.07 (d, J = 8.2 Hz, 1H), 8.03 (d, J = 0.6 Hz, 1H), 7.31 (dd, J = 8.2, 1.9 Hz, 1H),7.28 (d, J = 1.9 Hz, 1H), 5.97 (tt, J = 11.0, 5.7 Hz, 1H), 2.61-2.74 (m, 4H), 1.79 (s, 6H),1.64 (s, 6H), 2 Hs not observed (NH and OH).71MS m/z 469.5 [M + H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.53 (s, 1H), 8.18 (s,1H), 8.11 (d, J = 8.2 Hz, 1H), 7.55 (t, J = 59.9 Hz, 1H), 7.37 (dd, J = 8.2, 1.6 Hz, 1H),7.34 (d, J = 1.6 Hz, 1H), 5.97 (tt, J = 10.1, 6.3 Hz, 1H), 2.56-2.73 (m, 4H), 1.78 (s, 6H),1.64 (s, 6H), 2Hs not observed (NH and OH).77MS m/z 460.5 [M + H]+;1H NMR (methanol-d4) δ: 9.22 (s, 1H), 8.40 (d, J = 6.3 Hz,1H), 8.32 (d, J = 7.9 Hz, 1H), 7.79 (dd, J = 6.3, 1.6 Hz, 1H), 7.75 (d, J = 0.9 Hz,1H),7.60-7.65 (m, 2H), 5.97-6.02 (m, 1H), 4.31 (s, 3H), 2.62-2.78 (m, 4H), 1.80 (s, 6H),1.65 (s, 6H); 2 Hs not observed (NH and OH).93MS m/z 446.4 [M + H]+;1H NMR (methanol-d4) δ: 9.17 (s, 1H), 8.24 (d, J = 7.9 Hz,1H), 7.83 (d, J = 6.3 Hz, 1H), 7.40-7.50 (m, 2H), 7.14 (d, J = 8.8 Hz, 1H), 7.08 (br s,1H), 5.89-6.05 (m, 1H), 2.58-2.76 (m, 4H), 1.79 (s, 6H), 1.65 (s, 6H); 3 Hs notobserved (NH and 2 OHs). Example 14 Preparation of Compound 67 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with [3-(methoxymethoxy)-4-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenyl]trifluoromethanesulfonate (prepared in example 13, step 4, 60 mg, 0.11 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (35 mg, 0.14 mmol), 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (5 mg, 0.006 mmol) and potassium acetate (33 mg, 0.33 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (1 mL) was added and the reaction was heated at 90° C. for 90 minutes, after which UPLC showed full conversion to the borylated product. The crude product was cooled to room temperature and used directly in the next step. Step 2: To crude 6-(2-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (˜0.11 mmol) was added 6-bromopyridin-3-ol (25 mg, 0.14 mmol), 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (9 mg, 0.011 mmol), and aqueous 2 M K2CO3(141 μL, 0.282 mmol). The mixture was purged with argon for 5 min, then heated to 85° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified using silica-gel chromatography eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide 6-[3-(methoxymethoxy)-4-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenyl]pyridin-3-ol (37.5 mg, 68%) as a light brown solid. MS m/z 490.3 [M+H]+. Step 3: 6-[3-(methoxymethoxy)-4-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenyl]pyridin-3-ol (37.5 mg, 0.077 mmol) was dissolved in 1 mL of methanol, then 4 N HCl in 1,4-dioxane (0.5 mL, 2 mmol) was added and the reaction stirred at room temperature for 2 h. The reaction was concentrated, triturated with 20% MeOH/ether, and the resultant precipitate was filtered to afford 6-[3-hydroxy-4-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenyl]pyridin-3-ol dihydrochloride (31 mg, 78%) as a light brown solid. MS m/z 446.4 [M+H]+;1H NMR (methanol-d4) δ: 9.23 (s, 1H), 8.36 (d, J=8.2 Hz, 1H), 8.34 (d, J=2.5 Hz, 1H), 8.30 (d, J=8.8 Hz, 1H), 8.12 (dd, J=9.1, 2.8 Hz, 1H), 7.54-7.59 (m, 2H), 5.95-6.04 (m, 1H), 2.62-2.77 (m, 4H), 1.75 (s, 6H), 1.65 (s, 6H); 3 Hs not observed (NH and 2 OHs). Using the procedure described for Example 14, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData60MS m/z 431.3 [M + H]+;1H NMR (methanol-d4) δ: 9.23 (s, 1H), 9.17 (s, 1H), 8.79 (s,1H), 8.64 (s, 1H), 8.24 (d, J = 8.2 Hz, 1H), 7.79-8.00 (m, 2H), 5.95-6.02 (m, 1H),2.55-2.75 (m, 4H), 1.79 (s, 6H), 1.65 (s, 6H); 2 Hs not observed (NH and OH).61MS m/z 430.5 [M + H]+;1H NMR (methanol-d4) δ: 9.25 (s, 1H), 8.91 (d, J = 5.4 Hz,1H), 8.76 (t, J = 7.6 Hz, 1H), 8.51 (d, J = 7.9 Hz, 1H), 8.42 (d, J = 7.9 Hz, 1H), 8.13 (t,J = 6.5 Hz, 1H), 7.63-7.71 (m, 2H), 6.00 (m, 1H), 2.59-2.79 (m, 4H), 1.80 (s, 6H),1.66 (s, 6H); 2 Hs not observed (NH and OH).68MS m/z 447.4 [M + H]+;1H NMR (methanol-d4) δ: 9.15 (s, 1H), 8.47 (s, 2H), 8.13-8.19(m, 1H), 7.98-8.02 (m, 2H), 5.92-6.04 (m, 1H), 2.65-2.74 (m, 4H), 1.79 (s, 6H), 1.64(s, 6H); 3 Hs not observed (NH and 2 OHs).73MS m/z 444.6 [M + H]+;1H NMR (methanol-d4) δ: 9.23 (s, 1H), 8.75 (d, J = 6.3 Hz,1H), 8.32-8.37 (m, 2H), 8.26 (dd, J = 6.1, 1.7 Hz, 1H), 7.63-7.71 (m, 2H), 5.98-6.02(m, 1H), 2.90 (s, 3H), 2.64-2.75 (m, 4H), 1.80 (s, 6H), 1.65 (s, 6H); 2 Hs not observed(NH and OH).74MS m/z 498.3 [M + H]+;1H NMR (methanol-d4) δ: 9.18 (s, 1H), 8.82 (d, J = 5.4 Hz,1H), 8.27 (d, J = 8.2 Hz, 1H), 8.15 (d, J = 1.9 Hz, 1H), 8.03 (dd, J = 5.0, 1.9 Hz, 1H),7.55 (dd, J = 8.2, 1.9 Hz, 1H), 7.52 (d, J = 1.9 Hz, 1H), 5.89-6.05 (m, 1H), 2.57-2.79 (m,4H), 1.79 (s, 6H), 1.64 (s, 6H); 2 Hs not observed (NH and OH).75MS m/z 431.5 [M + H]+;1H NMR (methanol-d4) δ: 9.42 (d, J = 1.3 Hz, 1H), 9.21 (s,1H), 8.97-9.03 (m, 1H), 8.35 (dd, J = 6.0, 1.3 Hz, 1H), 8.31 (d, J = 8.5 Hz, 1H), 8.04(s, 1H), 7.99 (dd, J = 8.2, 1.9 Hz, 1H), 5.98-6.02 (m, 1H), 2.63-2.76 (m, 4H), 1.79 (s,6H), 1.65 (s, 6H); 2 Hs not observed (NH and OH).76MS m/z 431.5 [M + H]+;1H NMR (methanol-d4) δ: 9.96 (dd, J = 2.4, 1.1 Hz, 1H), 9.57(dd, J = 6.0, 0.9 Hz, 1H), 9.24 (s, 1H), 8.71 (dd, J = 5.8, 2.4 Hz, 1H), 8.38 (d, J = 7.9Hz, 1H), 7.71-7.78 (m, 2H), 5.98-6.02 (m, 1H), 2.57-2.78 (m, 4H), 1.79 (s, 6H), 1.65(s, 6H); 2 Hs not observed (NH and OH).78MS m/z 431.4 [M + H]+;1H NMR (DMSO-d6) δ: 9.24 (s, 1H), 9.20 (s, 3H), 8.23 (d,J = 8.2 Hz, 1H), 7.48 (d, J = 8.2 Hz, 1H), 7.46 (s, 1H), 5.68 (tt, J = 12.5, 3.4 Hz, 1H), 2.21(dd, J = 12.5, 3.4 Hz, 2H), 2.11 (t, J = 12.5 Hz, 2H), 1.36 (s, 6H), 1.19 (s, 6H); 2 Hs notobserved (NH and OH).79MS m/z 447.5 [M + H]+;1H NMR (methanol-d4) δ: 9.12 (s, 1H), 8.64 (d, J = 1.3 Hz,1H), 8.28 (d, J = 8.8 Hz, 1H), 7.94 (d, J = 1.3 Hz, 1H), 7.62 (d, J = 2.2 Hz, 1H), 7.57 (dd,J = 8.5, 2.2 Hz, 1H), 5.74-5.90 (m, 1H), 2.27-2.44 (m, 4H), 1.52 (s, 6H), 1.36 (s, 6H); 3Hs not observed (2 OH and NH).84MS m/z 473.3 [M + H]+;1H NMR (DMSO-d6) δ: 9.19 (s, 1H), 8.50 (d, J = 2.4 Hz, 1H),8.14 (d, J = 8.8 Hz, 1H), 7.88 (dd, J = 8.9, 2.6 Hz, 1H), 7.18-7.36 (m, 2H), 6.75(d,J = 8.9 Hz, 1H), 5.67 (tt, J = 12.5, 3.9 Hz, 1H), 3.09 (s, 6H), 2.20 (dd, J = 12.5, 3.9 Hz,2H), 2.11 (t, J = 12.5 Hz, 2H), 1.28-1.42 (m, 6H), 1.18 (s, 6H); 2 Hs not observed (NHand OH).85MS m/z 470.3 [M + H]+;1H NMR (DMSO-d6) δ: 9.19 (d, J = 1.6 Hz, 1H), 9.17 (s, 1H),8.72 (dd, J = 4.9, 1.6 Hz, 1H), 8.26 (d, J = 8.8 Hz, 1H), 8.16 (s, 1H), 8.01 (d, J = 4.9 Hz,1H), 7.41-7.45 (m, 2H), 5.69 (tt, J = 12.2, 3.7 Hz, 1H), 2.22 (dd, J = 12.2, 3.7 Hz, 2H),2.13 (t, J = 12.2 Hz, 2H), 1.37 (s, 6H), 1.20 (s, 6H); 2 Hs not observed (NH and OH).86MS m/z 486.4 [M + H]+;1H NMR (DMSO-d6) δ: 9.13 (s, 1H), 8.14 (d, J = 7.8 Hz, 1H),7.65 (d, J = 7.3 Hz, 1H), 7.24-7.42 (m, 2H), 6.67 (d, J = 2.0 Hz,1H), 6.54 (dd, J = 7.3,2.0 Hz, 1H), 5.64-5.82 (m, 1H), 3.10-3.21 (m, 1H), 2.08-2.33 (m, 4H), 1.42 (s, 6H),1.26 (s, 6H), 0.98-1.07 (m, 2H), 0.83-0.95 (m, 2H); 2 Hs not observed (NH and OH).88MS m/z 469.4 [M + H]+;1H NMR (DMSO-d6) δ: 11.69 (br s, 2H), 9.18 (s, 1H), 8.45 (d,J = 7.3 Hz, 1H), 8.43 (s, 1H), 8.18 (d, J = 8.3 Hz, 1H), 7.97 (s, 1H), 7.48 (s, 1H), 7.44(dd, J = 8.2, 1.8 Hz, 1H), 7.41 (d, J = 1.7 Hz, 1H), 7.09 (dd, J = 7.5, 1.8 Hz, 1H), 5.68 (tt,J = 13.0, 3.9 Hz, 1H), 2.21 (dd, J = 12.5, 3.7 Hz, 2H), 2.11 (t, J = 12.5 Hz, 2H), 1.36 (s,6H), 1.19 (s, 6H).89MS m/z 435.5 [M + H]+;1H NMR (methanol-d4) δ: 8.94-9.13 (m, 1H), 8.07 (d, J = 8.2Hz, 1H), 7.75 (t, J = 2.2 Hz, 1H), 7.51 (d, J = 1.9 Hz, 2H), 7.38 (dd, J = 8.2, 1.6 Hz, 1H),7.34 (d, J = 1.9 Hz, 1H), 5.76-5.96 (m, 1H), 2.42-2.58 (m, 4H), 1.63 (s, 6H), 1.48 (s,6H); 2 Hs not observed (OH and NH).90MS m/z 469.4 [M + H]+;1H NMR (DMSO-d6) δ: 11.64 (br s, 1H), 9.18 (s, 1H), 8.67 (d,J = 7.5 Hz, 1H), 8.12-8.24 (m, 2H), 8.01 (s, 1H), 7.93 (s, 1H), 7.66 (s, 1H), 7.51 (dd,J = 8.3, 1.9 Hz, 1H), 7.47 (d, J = 1.9 Hz, 1H), 7.33 (dd, J = 7.1, 1.9 Hz, 1H), 5.75 (tt,J = 12.5, 4.0 Hz, 1H), 2.17-2.36 (m, 4H), 1.44 (s, 6H), 1.28 (s, 6H).91MS m/z 419.3 [M + H]+;1H NMR (DMSO-d6) δ: 12.64 (br s, 1H), 9.16 (s, 1H), 8.15 (d,J = 8.3 Hz, 1H), 7.67 (d, J = 1.9 Hz, 1H), 7.59 (dd, J = 8.3, 1.9 Hz, 1H), 7.30 (br s, 1H),7.08 (br s, 1H), 5.67 (tt, J = 12.2, 3.7 Hz, 1H), 2.20 (dd, J = 12.2, 3.7 Hz, 2H), 2.10 (t,J = 12.2 Hz, 2H), 1.35 (s, 6H), 1.18 (s, 6H); 2 Hs not observed (NH and OH).95MS m/z 419.5 [M + H]+;1H NMR (methanol-d4) δ: 9.10 (s, 1H), 8.08 (d, J = 8.2 Hz,1H), 8.01-8.03 (m, 1H), 7.61-7.63 (m, 1H), 7.30 (dd, J = 7.9, 1.9 Hz, 1H), 7.26 (d,J = 1.9 Hz, 1H), 6.86-6.89 (m, 1H), 5.92-6.03 (m, 1H), 2.65-2.70 (m, 4H), 1.78 (s, 6H),1.63 (s, 6H); 2 Hs not observed (OH and NH).96MS m/z 436.4 [M + H]+;1H NMR (methanol-d4) δ: 9.16 (s, 1H), 8.16-8.29 (m, 1H),7.96-8.07, (m, 1H), 7.80 (s, 1H), 7.68 (s, 2H), 5.84-6.06 (m, 1H), 2.67 (d, J = 4.4 Hz,4H), 1.77 (s, 6H), 1.62 (s, 6H); 2 Hs not observed (OH and NH).99MS m/z 445.1 [M + H]+;1H NMR (DMSO-d6) δ: 11.50 (br s, 2H), 9.12 (s, 1H), 8.15 (d,J = 8.5 Hz, 1H), 8.00 (d, J = 4.6 Hz, 1H), 7.19-7.40 (m, 2H), 6.81 (d,J = 4.6 Hz, 1H),6.75 (s, 1H), 6.05 (s, 2H), 5.60-5.77 (m, 1H), 2.20 (d, J = 12.2 Hz, 2H), 2.11 (t, J = 12.5Hz, 2H), 1.36 (s, 6H), 1.19 (s, 6H).100MS m/z 473.1 [M + H]+;1H NMR (DMSO-d6) δ: 9.17 (s, 1H), 8.08-8.22 (m, 2H), 7.39(s, 1H), 7.37 (d, J = 1.6 Hz, 1H), 6.87 (dd, J = 5.1, 1.6 Hz, 1H), 6.85 (s, 1H), 5.67 (tt,J = 12.2, 3.7 Hz, 1H), 3.10 (s, 6H), 2.20 (dd, J = 12.2, 3.7 Hz, 2H), 2.11 (t, J = 12.2 Hz,2H), 1.35 (s, 6H), 1.18 (s, 6H); 2Hs not observed (NH and OH).101MS m/z 448.1 [M + H]+;1H NMR (DMSO-d6) δ: 9.13 (s, 1H), 8.71 (d, J = 2.4 Hz, 1H),8.55 (d, J = 5.1 Hz, 1H), 8.19 (d, J = 8.3 Hz, 1H), 7.71 (dd, J = 7.1, 5.1 Hz, 1H), 7.38 (s,1H), 7.35 (d, J = 8.3 Hz, 1H), 5.69 (tt, J = 12.2, 4.2 Hz, 1H), 2.21 (dd, J = 12.2, 4.2 Hz,2H), 2.12 (t, J = 12.2 Hz, 2H), 1.36 (s, 6H), 1.19 (s, 6H); 2Hs not observed (NH andOH).103MS m/z 496.7[M + H]+;1H NMR (DMSO-d6) δ: 9.14 (s, 1H), 8.62 (d, J = 2.4 Hz, 1H),8.35 (s, 1H), 8.17 (d, J = 8.0 Hz, 1H), 8.10 (d, J = 8.8 Hz, 1H), 7.79-7.84 (m, 2H), 7.71(d, J = 6.4 Hz, 1H), 7.21-7.58 (m, 1H), 5.69-5.72 (m, 1H), 2.13-2.27 (m, 4H), 1.40 (s,6H), 1.23 (s, 6H); 1 H is not observed (NH or OH).104MS m/z 459.8[M + H]+;1H NMR (DMSO-d6) δ: 9.13 (s, 1H), 8.15 (d, J = 8.4 Hz, 1H),8.08 (d, J = 5.2 Hz, 1H), 7.30-7.34 (m, 2H), 6.81 (dd, J = 5.2, 1.2 Hz, 1H), 6.72 (s, 1H),6.58-6.62 (m, 1H), 5.63-5.72 (m, 1H), 2.83 (d, J = 4.8 Hz, 3H), 2.17-2.22 (m, 2H),2.06-2.14 (m, 2H), 1.35 (s, 6H), 1.18 (s, 6H).106MS m/z 437.5 [M + H]+;1H NMR (methanol-d4) δ: 9.10 (s, 1H), 8.09 (d, J = 9.2 Hz,1H), 8.03 (d, J = 1.7 Hz, 1H), 7.32 (d, J = 7.2 Hz, 2H), 5.93-6.01 (m, 1H), 2.65-2.70 (m,4H), 1.78 (s, 6H), 1.64 (s, 6H); 3Hs not observed (1 OH and 2 NHs).107MS m/z 436.5 [M + H]+;1H NMR (methanol-d4) δ: 9.58 (s, 1H), 9.17 (s, 1H), 8.57 (s,1H), 8.21 (d, J = 9.9 Hz, 1H), 7.44 (dd, J = 10.2, 3.2 Hz, 2H), 5.90-6.04 (m, 1H), 2.61-2.75 (m, 4H), 1.79 (s, 6H), 1.64 (s, 6H); 2Hs not observed (1 OH and 1 NH).108MS m/z 433.6 [M + H]+;1H NMR (DMSO-d6) δ: 9.41-9.58 (m, 1H), 9.14 (s, 1H), 8.23-8.37 (m, 1H), 8.8 (d, J = 6.6 Hz, 1H), 7.86-7.91 (m, 1H), 7.14-7.19 (m, 2H), 5.83-5.95(m, 1H), 2.56-2.65 (m, 2H), 2.46-2.56 (m, 2H), 2.44 (s, 3H), 1.66 (s, 6H),1.54 (s,6H); 1H not observed (NH or OH).109MS m/z 444.5 [M + H]+;1H NMR (DMSO-d6) δ: 9.17 (s, 1H), 8.55 (s, 1H), 8.18 (d,J = 10.5 Hz, 1H), 7.42 (d, J = 1.8 Hz, 1H), 7.34-7.38 (m, 1H), 5.65-5.83 (m, 1H), 2.11-2.34 (m, 4H), 1.41 (s, 6H), 1.24 (s, 6H); 3Hs not observed (2 NHs and OH).110MS m/z 461.5 [M + H]+;1H NMR (methanol-d4) δ: 9.16 (s, 1H), 8.57 (s, 1H), 8.23 (d,J = 9.2 Hz, 1H), 7.68-7.74 (m, 2H), 5.95-6.02 (m, 1H), 2.65-2.71 (m, 4H), 1.79 (s, 6H),1.64 (s, 6H); 2Hs not observed (OH and NH).111MS m/z 420.2 [M + H]+;1H NMR (DMSO-d6) δ: 9.10 (s, 1H), 8.30-8.39 (m, 2H), 8.28(s, 1H), 8.17 (d, J = 8.0 Hz, 1H), 7.71(d, J = 1.2 Hz, 1H), 7.63 (dd, J = 8.4, 1.6 Hz,1H), 7.44 (d, J = 0.8 Hz, 1H), 5.68-5.71 (m, 1H), 2.11-2.25 (m, 4H), 1.38 (s, 6H), 1.21(s, 6H).112MS m/z 420.1 [M + H]+;1H NMR (DMSO-d6) δ: 9.13 (s, 1H), 8.42 (s, 1H), 8.15 (d, J =8.4 Hz, 1H), 7.60 (d, J = 1.6 Hz, 1H), 7.53 (dd, J = 8.4, 1.6 Hz,1H), 5.66-5.69 (m,1H), 2.06-2.23 (m, 4H), 1.36 (s, 6H), 1.19 (s, 6H); 3 Hs not observed (2NHs and OH).113MS m/z 461.2 [M + H]+;1H NMR (DMSO-d6) δ: 9.14 (s, 1H), 8.89 (d, J = 1.2 Hz, 1H),8.17 (d, J = 8.4 Hz, 1H), 7.91 (d, J = 1.6 Hz, 1H), 7.80 (dd, J = 8.0, 1.6 Hz, 1H), 7.51(d, J = 0.8 Hz, 1H), 5.64-5.71 (m, 1H), 3.99 (s, 3H), 2.07-2.22 (m, 4H), 1.35 (s, 6H),1.18 (s, 6H); 2 Hs not observed (NH and OH).114MS m/z 496.4 [M + H]+;1H NMR (methanol-d4) δ: 9.15 (s, 1H), 8.32 (d, J = 5.5 Hz,1H), 8.22 (d, J = 8.2 Hz, 1H), 7.63-7.78 (m, 1H), 7.57 (dd, J = 5.5, 1.5 Hz, 1H), 7.43-7.50 (m, 2H), 7.32 (d, J = 0.9 Hz, 1H), 5.99 (tt, J = 10.9, 5.4 Hz, 1H), 2.63- 2.75 (m,4H), 1.79 (s, 6H), 1.64 (s, 6H); 2 Hs not observed (NH and OH).115MS m/z 419.1 [M + H]+;1H NMR (DMSO-d6) δ: 12.27 (s, 1H), 11.59 (m, 1H), 9.13 (d,J = 8.0 Hz, 1H), 8.07 (d, J = 8.0 Hz, 1H), 7.10-7.76 (m, 2H), 7.40-7.53 (m, 2H), 5.61-5.70 (m, 1H), 2.06-2.22 (m, 4H), 1.35 (s, 6H), 1.18 (s, 6H); 1 H not observed (NH orOH).116MS m/z 437.4 [M + H]+;1H NMR (methanol-d4) δ: 9.53 (s, 1H), 9.15 (s, 1H), 8.24 (d,J = 8.2 Hz, 1H), 7.74 (d, J = 1.8 Hz, 1H), 7.70 (dd, J = 7.9, 1.8 Hz, 1H), 5.87-6.00 (m,1H), 2.57 (d, J = 7.6 Hz, 4H), 1.70 (s, 6H), 1.55(s, 6H); 2 Hs not observed (OH andNH).118MS m/z 469.5 [M + H]+;1H NMR (methanol-d4) δ: 9.22 (s, 1H), 8.52 (d, J = 6.1 Hz,1H), 8.33-8.38 (m, 1H), 7.83 (d, J = 3.4 Hz, 1H), 7.79 (d, J = 6.1 Hz, 1H), 7.58-7.64(m, 2H), 7.13 (d, J = 3.4 Hz, 1H), 5.96-6.05 (m, 1H), 2.64-2.76 (m, 4H), 1.80 (s,6H), 1.65 (s, 6H); 3 Hs not observed (2NHs and OH).119MS m/z 461.5 [M + H]+;1H NMR (methanol-d4) δ: 9.24 (s, 1H), 8.70 (d, J = 6.4 Hz,1H), 8.34 (d, J = 8.2 Hz, 1H), 8.10 (d, J = 1.5 Hz, 1H), 8.04 (dd, J = 8.2, 1.5 Hz, 1H),7.20 (d, J = 6.7 Hz, 1H), 6.00 (tt, J = 11.0, 5.5 Hz, 1H), 4.33 (s, 3H), 2.61-2.77 (m,4H), 1.79 (s, 6H), 1.65 (s, 6H),;2 Hs not observed (NH and OH).120MS m/z 420.5 [M + H]+;1H NMR (methanol-d4) δ: 9.19-9.22 (m, 1H), 9.12-9.15 (m,1H), 8.91-8.95 (m, 1H), 8.14 (d, J = 9.2 Hz, 1H), 7.35 (dd, J = 14.2, 8.4 Hz, 2H), 5.94-6.02 (m, 1H), 2.64-2.71 (m, 4H), 1.78 (s, 6H), 1.63 (s, 6H); 2Hs not observed (NHand OH).121MS m/z 451.5 [M + H]+;1H NMR (methanol-d4) δ: 9.09 (s, 1H), 8.09 (d, J = 8.2 Hz,1H), 7.77-7.89 (m, 1H), 7.27 (s, 2H), 5.91-6.02 (m, 1H), 3.84 (s, 3H), 2.59-2.70 (m,4H), 1.78 (s, 6H), 1.63 (s, 6H); 2Hs not observed (NH and OH).122MS m/z 465.6 [M + H]+;1H NMR (methanol-d4) δ: 9.04 (s, 1H), 8.10-8,08 (d, J = 9.6Hz, 1H), 7.87 (d, J = 3.2 Hz, 1H), 7.24-7.31 (m, 2H), 5.74-5.88 (m, 1H), 4.20 (q, J = 7.2Hz, 2H), 2.24-2.41 (m, 4H), 1.51 (s, 6H), 1.50 (t, J = 7.5 Hz, 3H), 1.40 (s, 6H); 2Hs notobserved (NH and OH).123MS m/z 474.5 [M + H]+;1H NMR (methanol-d4) δ: 9.21 (s, 1H), 8.38 (d, J = 6.1 Hz,1H), 8.31 (d, J = 8.2 Hz, 1H), 7.77 (d, J = 6.4 Hz, 1H), 7.72 (s, 1H), 7.58-7.64 (m,2H), 5.99 (tt, J = 10.7, 5.6 Hz, 1H), 4.65 (q, J = 6.9 Hz, 2H), 2.63-2.75 (m, 4H), 1.79(s, 6H), 1.64 (s, 6H), 1.59 (t, J = 7.0 Hz, 3H); 2 Hs not observed (NH and OH).124MS m/z 475.5 [M + H]+;1H NMR (methanol-d4) δ: 9.23 (s, 1H), 9.16 (s, 1H), 8.36 (d,J = 8.2 Hz, 1H), 7.63-7.74 (m, 3H), 5.98-6.02 (m, 1H), 4.71-4.79 (q, J = 7.2 Hz,2H), 2.59-2.79 (m, 4H), 1.79 (s, 6H), 1.65 (s, 6H), 1.54 (t, J = 7.2 Hz, 3H); 2 Hs notobserved (NH and OH).125MS m/z 470.5 [M + H]+;1H NMR (methanol-d4) δ: 9.17 (s, 1H), 9.01 (d, J = 7.3 Hz,1H), 8.27 (d, J = 4.9 Hz, 2H), 8.23 (d, J = 8.2 Hz, 1H), 7.61 (dd, J = 7.3, 1.8 Hz, 1H),7.52 (dd, J = 8.2, 1.8 Hz, 1H), 7.50 (d, J = 1.8 Hz, 1H), 5.98 (ddd, J = 16.3, 10.2, 6.4Hz, 1H), 2.60-2.76 (m, 4H), 1.80 (s, 6H), 1.65 (s, 6H); 2 Hs not observed (NH andOH).126MS m/z 470.5 [M + H]+;1H NMR (methanol-d4) δ: 9.21 (s, 1H), 9.14 (d, J = 7.0 Hz,1H), 9.04 (s, 1H), 8.27-8.35 (m, 2H), 7.97 (dd, J = 7.0, 1.5 Hz, 1H), 7.59-7.64 (m,2H), 6.00 (tt, J = 10.9, 5.5 Hz, 1H), 2.64 -2.75 (m, 4H), 1.79 (s, 6H), 1.65 (s, 6H), 2 Hsnot observed (NH and OH).127MS m/z 467.5 [M + H]+;1H NMR (methanol-d4) δ: 9.12 (s, 1H), 8.08-8.10 (m, 1H),8.07-8.11 (m, 1H), 8.01 (s, 1H), 7.28-7.44 (m, 2H), 5.89-6.05 (m, 1H), 3.90-3.96 (m,3H), 2.63-2.71 (m, 4H), 1.76-1.80 (s,6H), 1.59-1.66 (s, 6H); 1H not observed (NH orOH).128MS m/z 447.5 [M + H]+;1H NMR (methanol-d4) δ: 9.17 (s, 1H), 8.54 (s, 1H), 8.22 (d,J = 8.2 Hz, 1H), 7.71 (d, J = 1.8 Hz, 1H), 7.67 (dd, J = 8.2, 1.8 Hz, 1H), 7.01 (s, 1H),5.99 (tt, J = 10.9, 5.5 Hz, 1H), 2.61-2.77 (m, 4H), 1.79 (s, 6H), 1.64 (s, 6H), 3 Hs notobserved (2NHs and OH).129MS m/z 453.5 [M + H]+;1H NMR (methanol-d4) δ: 9.08 (s, 1H), 8.01-8.11 (m, 2H),7.30-7.41 (m, 2H), 5.86-5.99 (m, 1H), 2.56-2.63 (m, 4H), 1.71 (s, 6H), 1.56 (s, 6H);3Hs not observed (2 NHs and OH).130MS m/z 451.5 [M + H]+;1H NMR (methanol-d4) δ: 9.00-9.11 (m, 1H), 8.07-8.07 (m,1H), 8.07-8.08 (m, 1H), 8.04-8.10 s, 1H), 7.97 (d, J = 2.0 Hz, 1H), 7.93-7.99 (s, 1H),7.22-7.29 (m, 2H), 5.67-5.94 (m, 1H), 3.85 (s, 3H), 2.28-2.42 (m, 4H), 1.53 (s, 6H),1.37 (s, 6H); 1 H not observed (NH or OH).131MS m/z 449.5 [M + H]+;1H NMR (methanol-d4) δ: 9.06 (s, 1H), 8.02 (d, J = 8.5 Hz,1H), 7.95 (s, 1H), 7.44-7.46 (m, 1H), 7.39 (d, J = 9.8 Hz, 1H), 5.90-6.01 (m, 1H), 4.05(s, 3H), 2.62-2.74 (m, 4H), 1.78 (s, 6H), 1.63 (s, 6H); 3Hs not observed (2 NHs andOH).132MS m/z 458.5 [M + H]+;1H NMR (methanol-d4) δ: 9.06-9.15 (m, 1H), 8.11-8.23 (m,1H), 7.98-8.07 (m, 1H), 7.41-7.49 (m, 2H), 5.78-5.93 (m, 1H), 4.09-4.18 (m, 3H),2.39-2.53 (m, 4H), 1.56 (s, 6H), 1.44 (s, 6H); 2Hs not observed (NH and OH).133MS m/z 450.5 [M + H]+;1H NMR (methanol-d4) δ: 9.20 (s, 1H), 8.28 (d, J = 9.2 Hz,1H), 7.68 (m, 2H), 7.55 (s, 1H), 5.94-6.05 (m, 1H), 2.61-2.73 (m, 4H), 2.61 (s, 3H),1.79 (s, 6H), 1.63 (s, 6H); 2Hs not observed (NH and OH).134MS m/z 437.5 [M + H]+;1H NMR (methanol-d4) δ: 9.16 (s, 1H), 8.83 (s, 1H), 8.25 (d,J = 7.9 Hz, 1H), 7.77 (s, 2H), 5.87-5.97 (m, 1H), 2.49-2.58 (m, 4H), 1.66 (s, 6H), 1.51(s, 6H); 2Hs not observed (NH and OH).135MS m/z 488.5 [M + H]+;1H NMR (methanol-d4) δ: 9.15 (s, 1H), 8.21 (d, J = 8.2 Hz,1H), 7.95 (s, 1H), 7.57 (d, J = 12.2 Hz, 1H), 7.46 (d, J = 8.2 Hz, 1H), 7.43 (s,1H),5.95-6.02 (m, 1H), 2.63-2.76 (m, 4H), 1.79 (s, 6H), 1.64 (s, 6H), 3 Hs not observed(2NHs and OH).137MS m/z 497.4, 499.4 [M + H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.10 (d, J = 8.2Hz, 1H), 8.00 (s, 1H), 7.40 (d, J = 1.4 Hz, 1H), 7.35 (dd, J = 8.3, 1.6 Hz, 1H), 5.92-6.01(m, 1H), 2.64-2.70 (m, 4H), 1.78 (s, 6H), 1.63 (s, 6H); 3Hs not observed (2NHs andOH).138MS m/z 434.5 [M + H]+;1H NMR (methanol-d4) δ: 9.17 (s, 1H), 8.48 (s, 1H), 8.15 (d,J = 7.8 Hz, 1H), 7.52-7.57 (m, 2H), 5.92-6.03 (m, 1H), 4.22-4.26 (m, 3H), 2.66-2.71(m, 4H), 1.79 (s, 6H), 1.64 (s, 6H); 2Hs not observed (NH and OH).139MS m/z 487.4 [M + H]+;1H NMR (methanol-d4) δ: 9.09 (s, 1H), 8.10 (d, J = 7.6 Hz,1H), 8.05 (s, 1H), 7.19 (s, 2H), 5.93 (s, 1H), 2.40-2.53 (m, 4H), 1.57-1.66 (m, 6H),1.42-1.50 (m, 6H); 3Hs not observed (NH and OH).142MS m/z 470.3 [M + H]+;1H NMR (methanol-d4) δ: 9.12-9.15 (m, 3H), 8.21 (d, J = 8.1Hz, 1H), 8.14 (s, 1H), 7.87 (d, J = 0.9 Hz, 1H), 7.79 (d, J = 1.7 Hz, 1H), 7.74 (dd,J = 8.2, 1.8 Hz, 1H), 5.83-5.95 (m, 1H), 2.23-2.69 (m, 4H), 1.63 (s, 6H), 1.49 (s, 6H);2Hs not observed (NH and OH).145MS m/z 471.3 [M + H]+;1H NMR (DMSO-d6) δ: 11.65 (br s, 1H), 9.23 (br s, 1H), 9.09(br s, 1H), 8.71 (br s, 1H), 8.23 (d, J = 7.9 Hz, 1H), 7.45-7.64 (m, 2H), 7.37 (br s,1H), 5.92 (br s, 1H), 2.56-2.72 (m, 4H), 1.67 (s, 6H), 1.55 (s, 6H); 1H not observed(NH or OH).146MS m/z 471.3 [M + H]+;1H NMR (methanol-d4) δ: 9.89 (s, 1H), 9.23 (s, 1H), 8.59 (d,J = 0.9 Hz, 1H), 8.36 (d, J = 7.9 Hz, 1H), 7.72 (d, J = 1.8 Hz, 1H), 7.69 (dd, J = 8.2, 2.1Hz, 1H), 6.00 (tt, J = 10.8, 5.6 Hz, 1H), 2.62-2.76 (m, 4H), 1.80 (s, 6H), 1.65 (s, 6H);3 Hs not observed (2NHs and OH).147MS m/z 471.5 [M + H]+;1H NMR (methanol-d4) δ: 9.18 (s, 1H), 8.48 (d, J = 8.9 Hz,1H), 8.23 (d, J = 8.2 Hz, 1H), 8.14 (d, J = 8.5 Hz, 1H), 7.92 (d, J = 1.8 Hz, 1H), 7.88(dd, J = 8.2, 1.8 Hz, 1H), 5.99 (tt, J = 10.8, 5.5 Hz, 1H), 2.61-2.76 (m, 4H), 1.80 (s,6H), 1.65 (s, 6H); 3 Hs not observed (2NHs and OH).150MS m/z 450.4 [M + H]+;1H NMR (DMSO-d6) δ: 11.51 (br s, 2H), 9.27 (d, J = 12.8 Hz,1H), 9.15 (s, 1H), 8.23 (d, J = 13.4 Hz, 1H), 8.12 (s, 1H), 8.12 (d, J = 8.2 Hz, 1H),7.35 (dd, J = 7.9, 1.8 Hz, 1H), 7.28 (d, J = 1.5 Hz, 1H), 5.92 (tt, J = 12.3, 3.9 Hz, 1H),2.72 (s, 3H), 2.52-2.66 (m, 4H), 1.66 (s, 6H), 1.54 (s, 6H).151MS m/z 434.3 [M + H]+;1H NMR (methanol-d4) δ: 9.09-9.13 (m, 1H), 8.14 (d, J = 8.7Hz, 1H), 8.08 (s, 1H), 7.49-7.60 (m, 2H), 5.91-6.05 (m, 1H), 4.26 (s, 3H), 2.65-2.70(m, 4H), 1.78 (s, 6H), 1.64 (s, 6H); 2Hs not observed (NH and OH).152MS m/z 471.3 [M + H]+;1H NMR (methanol-d4) δ: 9.56 (s, 1H), 9.17 (s, 1H), 8.37 (d,J = 9.3 Hz, 1H), 8.29 (d, J = 8.5 Hz, 1H), 8.03 (d, J = 9.7 Hz, 1H), 7.77-7.86 (m, 2H),5.83-5.91 (m, 1H), 2.42 (d, J = 2.9 Hz, 4H), 1.57 (s, 6H), 1.42 (s, 6H); 2Hs notobserved (NH and OH).153MS m/z 451.3 [M + H]+;1H NMR (methanol-d4) δ: 9.14 (s, 1H), 8.24 (d, J = 7.3 Hz,1H), 7.71 (d, J = 1.5 Hz, 2H), 5.82-5.90 (m, 1H), 2.73 (s, 3H), 2.35-2.44 (m, 4H), 1.55(s, 6H), 1.39 (s, 6H); 2Hs not observed (NH and OH).154MS m/z 468.4 [M + H]+;1H NMR (1:1 CDCl3: methanol-d4) δ: 8.88 (s, 1H), 7.95 (d, J =7.9 Hz, 1H), 7.19 (d, J = 1.8 Hz, 1H), 7.16 (dd, J = 8.2, 1.8 Hz, 1H), 5.66-5.77 (m,1H), 2.60 (s, 3H), 2.26-2.34 (m, 4H), 1.47 (s, 6H), 1.32 (s, 6H); 2 Hs not observed(NH and OH).158MS m/z 434.5 [M + H]+;1H NMR (methanol-d4) δ: 8.89 (s, 1H), 7.99 (d, J = 8.2 Hz,1H), 7.26-7.34 (m, 3H), 5.76 (tt, J = 12.5, 4.0 Hz, 1H), 2.56 (s, 3H), 2.47 (t, J = 12.5Hz, 2H), 2.39 (dd, J = 13.1, 4.0 Hz, 2H), 1.60 (s, 6H), 1.47 (s, 6H); 2 Hs not observed(NH and OH).159MS m/z 462.6 [M + H]+;1H NMR (methanol-d4) δ: 9.16 (s, 1H), 9.00 (s, 1H), 7.92-8.46(m, 3H), 5.89-6.08 (m, 1H), 4.20 (s, 3H), 2.54-2.79 (m, 4H), 1.79 (s, 6H), 1.65 (s,6H); 2 Hs not observed (NH and OH).160MS m/z 470.2 [M + H]+;1H NMR (methanol-d4) δ: 9.18 (s, 1H), 8.23-8.28 (m, 2H),8.14 (d, J = 9.5 Hz, 1H), 7.86 (d, J = 9.8 Hz, 1H), 7.79 (d, J = 8.2 Hz, 3H), 5.94-6.03 (m,1H), 2.63-2.71 (m, 4H), 1.79 (s, 6H); 1.63 (s, 6H), 2Hs not observed (NH and OH).162MS m/z 470.4 [M + H]+;1H NMR (methanol-d4) δ: 9.25-9.29 (m, 1H), 9.12 (s, 1H),8.98 (s, 1H), 8.24 (d, J = 8.9 Hz, 1H), 7.94 (s, 1H), 7.80 (s, 1H), 7.42-7.47 (m, 2H),5.80-5.85 (m, 1H), 2.34-2.42 (m, 4H), 1.55 (s, 6H), 1.39 (s, 6H); 2Hs not observed(NH and OH). Example 15 Preparation of Compound 70 Step 1: A mixture imidazole (0.1 g, 1.47 mmol), 1-bromo-4-iodo-2-methoxybenzene (0.5 g, 1.6 mmol), 2-(2-pyridyl)benzimidazole (58.0 mg, 0.3 mmol), cesium carbonate (1.2 g, 3.66 mmol), copper (I) iodide (56 mg 0.29 mmol) in DMF (2 mL) was heated at 100° C. for 48 h. The reaction mixture was cooled to room temperature, filtered through Celite, washed with EtOAc, and concentrated. The crude material was purified by silica gel chromatography eluting with a EtOAc/hexane gradient (0-80% EtOAc) to provide 1-(4-bromo-3-methoxyphenyl)-1H-imidazole (0.25 g, 62%). MS m/z 253.3, 255.3 [M+H]+;1H NMR (methanol-d4) δ: 8.19 (s, 1H), 7.66 (d, J=8.5 Hz, 1H), 7.62 (t, J=1.4 Hz, 1H), 7.25 (d, J=2.2 Hz, 1H), 7.14-7.17 (m, 1H), 7.08 (dd, J=8.5, 2.2 Hz, 1H), 3.97 (s, 3H). Step 2: An oven-dried flask was equipped with a magnetic stir bar and charged with 1-(4-bromo-3-methoxyphenyl)-1H-imidazole (127.0 mg, 0.5 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (254.0 mg, 1.0 mmol), 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (38.0 mg, 0.05 mmol) and potassium acetate (200.0 mg, 2.0 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). Dioxane (1 mL) was added and the reaction was heated at 90° C. for 90 min, after which UPLC showed full conversion to the borylated product. The crude mixture was cooled to room temperature and used directly in the next step. Step 3: To the crude mixture from the above reaction was added 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (38.0 mg, 0.05 mmol) and 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 100.0 mg, 0.34 mmol). The tube was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). Aqueous 2.0 M K2CO3(0.75 mL, 1.5 mmol) was added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide 6-(4-(1H-imidazol-1-yl)-2-methoxyphenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (85 mg, 39%). MS m/z 433.3 [M+H]+. Step 4: To a solution of 6-(4-(1H-imidazol-1-yl)-2-methoxyphenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (50 mg, 0.115 mmol) in dichloromethane (3 mL) was added 1 M BBr3in CH2Cl2(0.6 mL, 0.6 mmol). The mixture was stirred at room temperature for 3 h. Methanol (3 mL) was added and the reaction was stirred for an additional 3 h. The mixture was concentrated at reduced pressure. The residue was purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (with 2.5% NH4OH) (0% to 30% MeOH/NH4OH) to provide 5-(1H-imidazol-1-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol (33 mg, 70%) as a yellow solid. MS m/z 419.4 [M+H]+;1H NMR (methanol-d4) δ: 9.08 (s, 1H), 8.25 (t, J=1.3 Hz, 1H), 8.20 (d, J=8.2 Hz, 1H), 7.67 (t, J=1.3 Hz, 1H), 7.27 (d, J=1.6 Hz, 2H), 7.19 (t, J=0.9 Hz, 1H), 5.72-5.87 (m, 1H), 2.24-2.40 (m, 4H), 1.51 (s, 6H), 1.35 (s, 6H); 2 Hs not observed (OH and NH). Using the procedure described for Example 15, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData63MS m/z 420.4 [M + H]+;1H NMR (methanol-d4) δ: 9.20 (s, 1H), 9.10 (s, 1H), 8.24 (d,J = 8.5 Hz, 1H), 8.21 (s, 1H), 7.57 (d, J = 2.2 Hz, 1H), 7.54 (dd, J = 8.5, 2.2 Hz, 1H),5.75-5.91 (m, 1H), 2.30-2.47 (m, 4H), 1.55 (s, 6H), 1.40 (s, 6H); 2 Hs not observed(OH and NH).64MS m/z 420.5 [M + H]+;1H NMR (methanol-d4) δ: 9.17 (s, 1H), 9.12 (s, 2H), 8.30 (d,J = 9.1 Hz, 1H), 7.35-7.41 (m, 2H), 5.93-6.03 (m, 1H), 2.67 (s, 4H), 1.78 (s, 6H),1.64(s, 6H); 2 Hs not observed (OH and NH).72MS m/z 420.4 [M + H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.63 (d, J = 1.3 Hz,1H), 8.27 (d, J = 8.8 Hz, 1H), 7.94 (d, J = 1.3 Hz, 1H), 7.61 (d, J = 2.2 Hz, 1H), 7.57 (dd,J = 8.5, 2.2 Hz, 1H), 5.77-5.87 (m, 1H), 2.28-2.39 (m, 4H), 1.51 (s, 6H), 1.36 (s, 6H); 2Hs not observed (OH and NH).143MS m/z 437.3 [M + H]+;1H NMR (DMSO-d6) δ: 12.05 (s, 1H), 9.24 (s, 1H), 9.14 (d,J = 11.0 Hz, 1H), 8.22 (d, J = 8.5 Hz, 1H), 8.18 (d, J = 12.8 Hz, 1H), 8.13 (t, J = 1.5Hz, 1H), 7.63 (dd, J = 8.1, 1.7 Hz, 1H), 7.36 (dd, J = 8.4, 2.3 Hz, 1H), 7.32 (d, J = 2.4Hz, 1H), 5.92 (tt, J = 12.2, 4.0 Hz, 1H), 4.02-4.16 (m, 1H), 2.52-2.66 (m, 4H), 1.66 (s,6H), 1.53 (s, 6H).144MS m/z 433.4 [M + H]+;1H NMR (methanol-d4) δ: 9.50 (s, 1H), 9.21 (s, 1H), 8.33 (d,J = 9.5 Hz, 1H), 7.93 (s, 1H), 7.44 (br s, 2H), 5.93-6.05 (m, 1H), 2.62-2.78 (m, 4H),2.45-2.54 (m, 3H), 1.80 (s, 6H), 1.67 (s, 6H); 2 Hs not observed (NH and OH).149MS m/z 447.5 [M + H]+;1H NMR (methanol-d4) δ: 9.19 (s, 1H), 8.34 (d, J = 8.2 Hz,1H), 7.48 (d, J = 1.2 Hz, 1H), 7.21-7.29 (m, 2H), 5.95-6.05 (m, 1H), 2.67-2.75 (m,4H), 2.66 (s, 3H), 2.43 (d, J = 1.2 Hz, 3H), 1.79 (s, 6H), 1.64 (s, 6H); 2 Ns notobserved (NH and OH).155MS m/z 433.5 [M + H]+;1H NMR (methanol-d4) δ: 9.18 (s, 1H), 8.28 (d, J = 9.2 Hz,1H), 7.92 (d, J = 2.1 Hz, 1H), 7.27 (sxt, J = 2.1 Hz, 2H), 6.52-6.55 (m, 1H), 5.99 (spt,J = 5.5 Hz, 1H), 2.64-2.75 (m, 4H), 2.50 (s, 3H), 1.79 (s, 6H), 1.65(s, 6H); 2 Hs notobserved (NH and OH).156MS m/z 433.5 [M + H]+;1H NMR (DMSO-d6) δ: 11.70 (s, 1H), 9.16 (s, 1H), 8.92-9.04(m, 1H), 8.33 (s, 1H), 8.16 (d, J = 8.5 Hz, 1H), 7.61-7.64 (m, 1H), 7.54 (d, J = 2.1 Hz,1H), 7.46 (dd, J = 8.5, 2.1 Hz, 1H), 5.89-5.97 (m, 1H), 2.53-2.67 (m, 4H), 2.40 (s,3H), 1.65 (s, 6H), 1.52 (s, 6H).157MS m/z 433.6 [M + H]+;1H NMR (methanol-d4) δ: 9.14 (s, 1H), 8.23 (d, J = 2.4 Hz,1H), 8.18 (d, J = 8.8 Hz, 1H), 7.45 (s, 1H), 7.44 (dd, J = 10.7, 2.1 Hz, 1H), 6.42 (d, J =2.4 Hz, 1H), 5.97 (tt, J = 10.4, 5.8 Hz, 1H), 2.64-2.75 (m, 4H), 2.41 (s, 3H),1.79 (s,6H), 1.64 (s, 6H); 2 Hs not observed (NH and OH). Example 16 Preparation of Compound 52 Step 1: A mixture of 3,6-dichloropyridazin-4-amine (1.0 g, 6.0 mmol), 1,2,2,6,6-pentamethylpiperidin-4-amine (1.0 g, 6.1 mmol) and DIPEA (1.6 mL, 9.1 mmol) in decanol (10 mL) was heated at 150° C. for 7 days. Solvent was removed by blowing air and the residue was purified using silica-gel chromatography eluting with a MeOH/CH2Cl2gradient (2.5% NH4OH) (0% to 30% MeOH/NH4OH) to provide a mixture of 6-chloro-N3-(1,2,2,6,6-pentamethyl-4-piperidyl)pyridazine-3,5-diamine and 6-chloro-N3-(1,2,2,6,6-pentamethyl-4-piperidyl)pyridazine-3,4-diamine (0.85 g, 47%) as a brown solid which was used as is in the next step. Step 2: To a solution of 6-chloro-N3-(1,2,2,6,6-pentamethyl-4-piperidyl)pyridazine-3,4-diamine, prepared above (0.85 g, 2.9 mmol, ˜59% pure), in AcOH (4 mL) was added NaNO2(0.50 g, 1.21 mmol) and the mixture was stirred at room temperature for 1 h. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate slowly until pH 7. The aqueous layer was extracted with ethyl acetate three times. The organic layer was dried over Na2SO4, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to yield 6-chloro-3-(1,2,2,6,6-pentamethyl-4-piperidyl)triazolo[4,5-c]pyridazine (375 g, 43%) as a tan solid. MS m/z 309.1 [M+H]+. Step 3: An oven-dried flask was equipped with a magnetic stir bar and charged with [6-chloro-3-(1,2,2,6,6-pentamethyl-4-piperidyl)triazolo[4,5-c]pyridazine (72 mg, 0.23 mmol), 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (prepared in example 1, step 7, 116 mg, 0.28 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(I) (17 mg, 0.023 mmol), and K2CO3(65 mg, 0.47 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (2 mL) and water (0.5 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide 6-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-3-(1,2,2,6,6-pentamethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (75 mg, 57%). MS m/z 561.4 [M+H]+. Step 4: To a solution of 6-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-3-(1,2,2,6,6-pentamethyl-4-piperidyl)triazolo[4,5-c]pyridazine (61 mg, 0.11 mmol) in CH2Cl2(1 mL) was added 4 N HCl in 1,4-dioxane (0.14 mL, 0.54 mmol) and the reaction mixture was stirred for 16 h. The precipitate formed during this time was collected by filtration, washed with CH2Cl2(3×), and dried to provide 2-[3-(1,2,2,6,6-pentamethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]-5-(1H-pyrazol-4-yl)phenol dihydrochloride (41 mg, 75%) as a yellow solid. MS m/z 433.3 [M+H]+;1H NMR (methanol-d4) δ: 9.13 (s, 1H), 8.52 (s, 2H), 8.12 (d, J=8.2 Hz, 1H), 7.40 (dd, J=8.2, 1.9 Hz, 1H), 7.37 (d, J=1.9 Hz, 1H), 5.95 (tt, J=12.9, 3.5 Hz, 1H), 3.03 (s, 3H), 2.95 (t, J=14.2 Hz, 2H), 2.77 (dd, J=14.2, 3.5 Hz, 2H), 1.76 (s, 6H), 1.70 (s, 6H); 2 Hs not observed (NH and OH). Using the procedure described for Example 16, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData56MS m/z 391.5 [M + H]+;1H NMR (methanol-d4) δ: 9.09 (s, 1H), 8.37 (br s, 2H), 8.09(d, J = 8.2 Hz, 1H), 7.36 (d, J = 8.5 Hz, 1H), 7.34 (s, 1H), 5.70-5.90 (m, 1H), 3.57-3.64(m, 2H), 2.70 (br. s., 4H), 1.68 (s, 3H), 1.58 (s, 3H); 3 Hs not observed (OH and 2NHs).57MS m/z 417.6 [M + H]+;1H NMR (methanol-d4) δ: 9.09 (s, 1H), 8.26 (s, 2H), 8.08 (d,J = 8.5 Hz, 1H), 7.34 (dd, J = 8.2, 1.9 Hz, 1H), 7.32 (d, J = 1.6 Hz, 1H), 5.80-5.92 (m,1H), 2.76 (dd, J = 13.6, 12.6 Hz, 2H), 2.61-2.66 (m, 2H), 2.50 (d, J = 9.5 Hz, 2H), 2.17(d, J = 9.1 Hz, 2H), 1.64 (s, 6H); 3 Hs not observed (OH and 2 NHs). Example 17 Preparation of Compound 7 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with 6-chloro-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 75 mg, 0.25 mmol), 4-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-H-pyrazole (prepared in example 1, step 7, 150 mg, 0.36 mmol), and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(I) complex with dichloromethane (25 mg, 0.029 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (4 mL) and aqueous 2.0 M K2CO3(0.3 mL, 0.60 mmol) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water (2 mL), and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to provide 6-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (125 mg, 90%) as a yellow solid. MS m/z 547.4 [M+H]+. Step 2: To a solution of 6-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (125 mg, 0.23 mmol) in CH2Cl2(1 mL) was added 4N HCl in dioxane (3 mL, 12 mmol) and the reaction was stirred at room temperature for 2 h. The yellow solid that precipitated was collected by vacuum filtration, washed with CH2Cl2and Et2O and dried to afford 5-(1H-pyrazol-4-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol dihydrochloride (95 mg, 91%). MS m/z 419.5 [M+H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.34 (s, 2H), 8.11 (d, J=8.2 Hz, 1H), 7.37 (dd, J=8.2, 1.9 Hz, 1H), 7.35 (d, J=1.9 Hz, 1H), 5.97 (tt, J=11.0, 5.4 Hz, 1H), 2.62-2.74 (m, 4H), 1.79 (s, 6H), 1.64 (s, 6H); 3 Hs not observed (2NHs and OH). Example 18 Preparation of Compound 34 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with 6-chloro-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 50 mg, 0.17 mmol), 1-[3-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]pyrazole (61 mg, 0.20 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(I) (13 mg, 0.17 mmol), and K2CO3(71 mg, 0.51 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). Dioxane (4 mL) and water (0.5 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water (2 mL), and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to provide 6-(2-methoxy-4-pyrazol-1-yl-phenyl)-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (55 mg, 75%) as a yellow solid. MS m/z 433.5 [M+H]+. Step 2: To a solution of 6-(2-methoxy-4-pyrazol-1-yl-phenyl)-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (55 mg, 0.13 mmol) in CH2Cl2(1 mL) was added 1 M BBr3in CH2Cl2(0.64 mL, 0.64 mmol) and the reaction was stirred at room temperature for 16 h after which, UPLC showed complete consumption of the starting material. The reaction was quenched with MeOH (10 mL), concentrated under reduced pressure, and purified using silica gel chromatography, eluting with a MeOH/CH2Cl2gradient (2.5% NH4OH) (0% to 30% MeOH/NH4OH) to provide 5-pyrazol-1-yl-2-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenol (38 mg, 71%) as a yellow solid. MS m/z 419.5 [M+H]+;1H NMR (DMSO-d6) δ: 9.18 (s, 1H), 8.97-9.07 (m, 1H), 8.58 (d, J=2.2 Hz, 1H), 8.19 (d, J=8.8 Hz, 1H), 8.11-8.17 (m, 1H), 7.81 (d, J=1.3 Hz, 1H), 7.62 (d, J=2.5 Hz, 1H), 7.54 (dd, J=8.7, 2.4 Hz, 1H), 6.61 (dd, J=2.4, 1.7 Hz, 1H), 5.93 (tt, J=12.3, 4.1 Hz, 1H), 2.52-2.65 (m, 4H), 1.66 (s, 6H), 1.53 (s, 6H). Example 19 Preparation of Compound 14 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with 1,4-dibromo-2,5-difluoro-benzene (3.2 g, 12 mmol), 1-tetrahydropyran-2-yl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (2.95 g, 10.6 mmol) and Pd(dppf)Cl2—CH2Cl2(430 mg, 0.50 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (50 mL) and aqueous 2.0 M K2CO3(15 mL, 30 mmol) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a EtOAc/hexanes gradient (0-50% EtOAc) to provide 4-(4-bromo-2,5-difluoro-phenyl)-1-tetrahydropyran-2-yl-pyrazole (1.51 g, 42%) as a brown oil. MS m/z 343.0, 345.0 [M+H]+. Step 2: An oven-dried flask was equipped with a magnetic stir bar and charged with Pd(dppf)Cl2·CH2Cl2(200 mg, 0.23 mmol), 4-(4-bromo-2,5-difluoro-phenyl)-1-tetrahydropyran-2-yl-pyrazole (1.51 g, 4.40 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (2.90 g, 11.4 mmol), and KOAc (1.73 g, 17.6 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (22 mL) was added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a EtOAc/hexanes gradient (5-50% EtOAc) to provide 4-[2,5-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (1.54 g, 89%) as a brownish solid. 1H NMR (CDCl3) δ: 8.09 (d, J=1.9 Hz, 1H), 7.95 (s, 1H), 7.47 (dd, J=10.7, 4.7 Hz, 1H), 7.23 (dd, J=9.5, 5.7 Hz, 1H), 5.38-5.51 (m, 1H), 4.01-4.16 (m, 1H), 3.73-3.78 (m, 1H), 2.05-2.20 (m, 3H), 1.66-1.79 (m, 3H), 1.39 (s, 12H). Step 3: An oven-dried reaction tube was equipped with a magnetic stir bar and charged with 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 75 mg, 0.25 mmol), 4-[2,5-difluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (199 mg, 0.51 mmol), and Pd(dppf)Cl2—CH2Cl2(25 mg, 0.029 mmol). The tube was sealed with a rubber screw-cap, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (1 mL) and aqueous 2.0 M K2CO3(0.3 mL, 0.6 mmol) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide 6-[2,5-difluoro-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (96 mg, 72%) as a brown solid. MS m/z 523.4 [M+H]+; Step 4: To a solution of 6-[2,5-difluoro-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (96 mg, 0.18 mmol) in CH2Cl2(1 mL) was added 4N HCl in dioxane (2 mL, 8 mmol) and the reaction was stirred at room temperature for 1 h. The yellow solid that precipitated was collected by vacuum filtration, rinsed with CH2Cl2and Et2O and dried to afford 6-(2,5-difluoro-4-(1H-pyrazol-4-yl)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine dihydrochloride. MS m/z 439.5 [M+H]+;1H NMR (DMSO-d6) δ: 9.56 (d, J=15.8 Hz, 1H), 8.97 (d, J=1.6 Hz, 1H), 8.36 (d, J=12.9 Hz, 1H), 8.25 (s, 1H), 7.70-7.91 (m, 1H), 5.93 (tt, J=12.9, 4.1 Hz, 1H), 2.59-2.67 (m, 4H), 1.67 (s, 6H), 1.56 (s, 6H), 2 Hs not observed (2 NHs). Using the procedure described for Example 19, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData13MS m/z 439.5 [M + H]+;1H NMR (DMSO-d6) δ: 9.64 (d, J = 12.8 Hz, 1H), 8.95 (d,J = 1.3 Hz, 1H), 8.39 (d, J = 12.8 Hz, 1H), 8.26 (d, J = 2.3 Hz, 1H), 7.92-7.99 (m, 1H),5.93 (tt, J = 12.3, 3.8 Hz, 1H), 2.58-2.65 (m, 4H), 1.67 (s, 6H), 1.56 (s, 6H); 2 Hs notobserved (2 NHs).58MS m/z 435.5 [M + H]+;1H NMR (methanol-d4) δ: 9.02 (s, 1H), 8.79 (s, 2H), 7.61 (s,1H), 7.37 (s, 1H), 5.97 (tt, J = 11.7, 4.7 Hz, 1H), 2.60-2.77 (m, 4H), 1.79 (s, 6H), 1.66(s, 6H); 4 Hs not observed (2 NHs and 2 OHs). Example 20 Preparation of Compound 53 Step 1: An oven-dried flask equipped with a magnetic stir bar was charged with 1-bromo-4-chloro-2-fluoro-5-methoxy-benzene (100 mg, 0.42 mmol), (4-methoxyphenyl)boronic acid (69.8 mg, 0.46 mmol) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) complex with dichloromethane (17.1 mg, 0.021 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (1.0 mL) and aqueous 1 M K2CO3(0.5 mL, 0.5 mmol) were added and the reaction was heated to 90° C. for 2 h. The reaction was cooled to room temperature, diluted with water (5 mL), and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel, eluting with a EtOAc/hexanes gradient (0-10% EtOAc) to yield 1-chloro-5-fluoro-2-methoxy-4-(4-methoxyphenyl)benzene (89.2 mg, 80%) as an off white solid. MS m/z 267.8 [M+H]+;1H NMR (CDCl3) δ: 7.39 (dd, J=8.8, 1.6 Hz, 2H), 7.13 (d, J=9.5 Hz, 1H), 6.92 (d, J=8.8 Hz, 2H), 6.85 (d, J=6.9 Hz, 1H), 3.85 (s, 3H), 3.79 (s, 3H). Step 2: An oven-dried flask was equipped with a magnetic stir bar and charged with 1-chloro-5-fluoro-2-methoxy-4-(4-methoxyphenyl)benzene (56 mg, 0.21 mmol), bis(pinacolato)diboron (66.6 mg, 0.26 mmol), potassium acetate (61.8 mg, 0.63 mmol), and chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II) (8.3 mg, 0.011 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (1.4 mL) was added and the reaction was heated to 120° C. for 64 h. The reaction was cooled to room temperature and aq. 1 M K2CO3(0.7 mL, 0.7 mmol) was added followed by 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 40.9 mg, 0.14 mmol), and 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (8.6 mg, 0.011 mmol). The mixture was purged with argon and heated at 90° C. for 3 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0-30% MeOH) to yield 6-[5-fluoro-2-methoxy-4-(4-methoxyphenyl)phenyl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (28.5 mg, 29%). MS m/z 491.5 [M+H]+. Step 3: 6-[5-Fluoro-2-methoxy-4-(4-methoxyphenyl)phenyl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (28.5 mg, 0.058 mmol) was combined with dichloromethane (2 mL) and 1N BBr3in dichloromethane (0.6 mL, 0.6 mmol). The mixture was stirred at room temperature for 3 h. Methanol (0.5 mL) was added and the reaction was stirred for 3 h. The mixture was concentrated at reduced pressure and the residue was triturated with MeOH (3×2 mL) and dried under vacuum to yield 6-fluoro-4-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-[1,1′-biphenyl]-3,4′-diol hydrobromide as an off white solid (11.3 mg, 36%). MS m/z 463.5 [M+H]+;1H NMR (DMSO-d6) δ: 11.22 (s, 1H), 9.75 (s, 1H), 9.20 (s, 1H), 8.95 (d, J=12.0 Hz, 1H), 8.11 (d, J=12.0 Hz, 1H), 7.98 (d, J=11.7 Hz, 1H), 7.45 (dd, J=8.5, 1.6 Hz, 2H), 7.13 (d, J=6.9 Hz, 1H), 6.91 (d, J=8.5 Hz, 2H), 5.94 (tt, J=12.3, 4.1 Hz, 1H), 2.59 (d, J=12.0 Hz, 2H), 2.54 (d, J=10.1 Hz, 2H), 1.66 (s, 6H), 1.52 (s, 6H). Using the procedure described for Example 20, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData33MS m/z 437.5 [M + H]+;1H NMR (DMSO-d6) δ: 10.13-10.31 (m, 1H), 9.55-9.70 (m,1H), 8.82 (d, J = 1.3 Hz, 1H), 8.32-8.42 (m, 1H), 8.26 (s, 2H), 7.70 (d, J = 12.3 Hz, 1H),7.64 (d, J = 6.9 Hz, 1H), 5.91 (tt, J = 12.3, 3.8 Hz, 1H), 2.56-2.70 (m, 4H), 1.67 (s, 6H),1.56 (s, 6H).55MS m/z 478.2 [M + H]+;1H NMR (methanol-d4) δ: 9.18 (s, 1H), 8.03 (d, J = 12.0 Hz,1H), 7.79 (s, 1H), 7.22 (d, J = 6.6 Hz, 1H), 6.83 (s, 1H), 6.70 (dt, J = 6.9, 1.9 Hz, 1H),5.99 (tt, J = 11.3, 5.4 Hz, 1H), 3.66 (s, 3 H), 2.63-2.71 (m, 4H), 1.78 (s, 6H), 1.63 (s,6H); 2 Hs not observed (NH and OH).59MS m/z 437.5 [M + H]+;1H NMR (methanol-d4) δ: 8.75 (s, 1H), 8.06 (s, 2H), 7.04 -7.18 (m, 2H), 5.99 (tt, J = 12.0, 4.7 Hz, 1H), 2.62-2.74 (m, 4H), 1.78 (s, 6H), 1.63 (s,6H); 3 Hs not observed (2 NHs and OH).62MS m/z 451.5 [M + H]+;1H NMR (DMSO-d6) δ: 11.51 (br s, 2H), 9.48 (d, J = 12.3 Hz,1H), 9.23 (s, 1H), 8.31 (d, J = 12.0 Hz, 1H), 8.21 (d, J = 1.9 Hz, 1H), 8.03 (d, J = 12.3Hz, 1H), 7.92 (s, 1H), 7.35 (d, J = 6.9 Hz, 1H), 5.91 (tt, J = 12.6, 4.1 Hz, 1H), 3.94 (s,3H), 2.59 (d, J = 12.9 Hz, 2H), 2.50-2.54 (m, 2H), 1.67 (s, 6H), 1.55 (s, 6H).87MS m/z 448.3 [M + H]+;1H NMR (methanol-d4) δ: 9.26 (s, 1H), 8.96-9.03 (m, 2H),8.44 (dd, J = 6.9, 1.3 Hz, 2H), 8.21 (d, J = 12.6 Hz, 1H), 7.50 (d, J = 6.6 Hz, 1H), 6.01 (tt,J = 10.7, 5.5 Hz, 1H), 2.63-2.73 (m, 4H), 1.80 (s, 6H), 1.65 (s, 6H); 2 Hs not observed(NH, and OH). Example 21 Preparation of Compound 35 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with 4-bromo-5-fluoro-2-methoxy-aniline (1.0 g, 4.5 mmol), 1-tetrahydropyran-2-yl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (1.6 g, 5.5 mmol), Pd(dppf)Cl2(340 mg, 0.45 mmol) and K2CO3(1.9 g, 14 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (20 mL) and water (2 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a EtOAc/hexanes gradient (0-60% EtOAc) to provide 5-fluoro-2-methoxy-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)aniline (1.25 g, 94%) as a colorless oil. MS m/z 292.3 [M+H]. Step 2: To a well stirred suspension of 5-fluoro-2-methoxy-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)aniline (1.25 g, 4.29 mmol) in THF (40 mL) under a nitrogen flow were sequentially added CsI (1.67 g, 6.44 mmol), 2 (1.09 g, 4.29 mmol), CuI (0.41 g, 2.15 mmol) and tBuONO (1.33 mL, 10.7 mmol). The reaction mixture was stirred vigorously at 65-70° C. for 6 h. After cooling in an ice-water bath, the solid was filtered off. The filtrate was diluted with dichloromethane (500 mL), washed with 30% aq. ammonium hydroxide (150 mL), sodium thiosulphate (300 mL), brine, and dried over anhydrous Na2SO4and concentrated under reduced pressure. The residue was purified by column chromatography, eluting with a EtOAc/hexanes gradient (0-80% EtOAc) to provide 4-(2-fluoro-4-iodo-5-methoxy-phenyl)-1-tetrahydropyran-2-yl-pyrazole (0.92 g, 53%) as a brownish solid. MS m/z 403.1 [M+H]+. Step 3: An oven-dried flask was equipped with a magnetic stir bar and charged with 4-(2-fluoro-4-iodo-5-methoxy-phenyl)-1-tetrahydropyran-2-yl-pyrazole (0.92 g, 2.23 mmol), Pd(dppf)Cl2(171 mg, 0.23 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (1.17 g, 4.57 mmol), and KOAc (0.68 g, 6.85 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (10 mL) was added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a EtOAc/hexanes gradient (0-50% EtOAc) to provide 4-(2-fluoro-5-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (0.745 g, 81%) as a clear oil. MS m/z 403.3 [M+H]+. Step 4: An oven-dried reaction tube was equipped with a magnetic stir bar and charged with 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 98 mg, 0.33 mmol), 4-(2-fluoro-5-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (267 mg, 0.66 mmol), Pd(dppf)Cl2—CH2Cl2(50 mg, 0.066 mmol) and K2CO3(276 mg, 2.0 mmol). The tube was sealed with a rubber screw-cap, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (2 mL) and water (0.5 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0% to 25% MeOH) To provide 6-(5-fluoro-2-methoxy-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (150 mg, 42%) as a brownish solid. MS m/z 535.4 [M+H]+. Step 5: 6-(5-Fluoro-2-methoxy-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (80 mg, 0.15 mmol) was dissolved in dichloromethane (2 mL) and treated with 1 N BBr3in dichloromethane (0.74 mL, 0.74 mmol). The mixture was stirred at room temperature for 3 h. Methanol (0.5 mL) was added and the reaction was stirred for 1 h. The reaction was concentrated at reduced pressure. The residue was triturated in MeOH, the resultant solid was filtered, washed with Et2O and dried under vacuum to yield 4-fluoro-5-(1H-pyrazol-4-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenol dihydrobromide (33 mg, 41%) as an orange solid. MS m/z 437.5 [M+H]+;1H NMR (DMSO-d6) δ:11.51 (br s, 1H), 9.25 (s, 1H), 8.92-9.03 (m, 1H), 8.07-8.16 (m, 2H), 8.03 (d, J=12.3 Hz, 1H), 7.38 (d, J=6.8 Hz, 1H), 5.93 (tt, J=12.3, 4.8 Hz, 1H), 2.53-2.63 (m, 4H), 1.66 (s, 6H), 1.52 (s, 6H); 1H not observed (NH or OH). Using the procedure described for Example 21, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData83MS m/z 453.9 [M + H]+;1H NMR (methanol-d4) δ: 9.18 (s, 1H), 8.63 (s, 2H), 8.28 (s,1H), 7.34 (s, 1H), 5.98 (tt, J = 12.0, 4.5 Hz, 1H), 2.61-2.79 (m, 4H), 1.80 (s, 6H), 1.66(s, 6H); 3 Hs not observed (2 NHs and OH). Example 22 Preparation of Compound 54 Step 1: (4-Chloro-2-fluoro-3-methoxy-phenyl)boronic acid (200 mg, 0.98 mmol) was combined with 4-bromo-1-tetrahydropyran-2-yl-pyrazole (271 mg, 1.17 mmol) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) complex with dichloromethane (80.0 mg, 0.098 mmol), followed by addition of 1,4-dioxane (2.0 mL) and aqueous 1 M K2CO3(1.0 mL, 1.0 mmol). The mixture was stirred at 110° C. for 2 h. The mixture was then partitioned between EtOAc and H2O. The aqueous layer was extracted with EtOAc and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was chromatographed on silica gel, eluting with 0-20% EtOAc in hexanes to yield 4-(4-chloro-2-fluoro-3-methoxy-phenyl)-1-tetrahydropyran-2-yl-pyrazole (165.7 mg, 54%) as an off white solid. MS m/z 311.0 [M+H]+;1H NMR (CDCl3) δ: 7.93 (d, J=1.9 Hz, 1H), 7.82 (s, 1H), 7.13 (dd, J=8.5, 7.3 Hz, 1H), 7.07 (dd, J=8.5, 1.3 Hz, 1H), 5.37 (dd, J=9.0, 3.0 Hz, 1H), 4.02 (d, J=9.8 Hz, 1H), 3.91 (s, 3H), 3.67 (td, J=11.2, 2.8 Hz, 1H), 2.08-2.18 (m, 2H), 1.88-2.01 (m, 1H), 1.60-1.72 (m, 2H), 1.57 (d, J=2.5 Hz, 1H). Step 2: 4-(4-Chloro-2-fluoro-3-methoxy-phenyl)-1-tetrahydropyran-2-yl-pyrazole (48.0 mg, 0.15 mmol) was combined with bis(pinacolato)diboron (49.0 mg, 0.19 mmol), potassium acetate (45.5 mg, 0.46 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II) (5.6 mg, 0.008 mmol), and 1,4-dioxane (1.0 mL). The mixture was stirred at 120° C. for 24 h. The mixture was cooled to room temperature. To the mixture was added aqueous 1 M K2CO3(0.5 mL, 0.5 mmol), 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 30.0 mg, 0.102 mmol), and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) complex with dichloromethane (4.2 mg, 0.008 mmol). The mixture was stirred at 90° C. for 6 h. The mixture was then partitioned between EtOAc and H2O. The aqueous layer was extracted with EtOAc. The combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was chromatographed on silica gel, eluting with 0-30% MeOH in CH2Cl2to yield 6-[3-fluoro-2-methoxy-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (10.6 mg, 20%). MS m/z 535.5 [M+H]+. Step 3: 6-[3-Fluoro-2-methoxy-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (11 mg, 0.02 mmol) was combined with dichloromethane (0.5 mL) and 1 N BBr3in dichloromethane (0.10 mL, 0.10 mmol). The mixture was stirred at room temperature for 5 h. Methanol (0.5 mL) was added and the reaction was stirred for 16 h. The reaction was concentrated at reduced pressure. The residue was partitioned between EtOAc and saturated aqueous NaHCO3. The aqueous layer was extracted with EtOAc. The combined organic layers were dried over Na2SO4, filtered and concentrated. The material was chromatographed on silica gel, eluting with 0-30% MeOH in CH2Cl2, and then further purified by reverse phase chromatography on C18 silica gel, eluting with 10-100% MeCN in H2O, to provide 2-fluoro-3-(1H-pyrazol-4-yl)-6-[3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazin-6-yl]phenol dihydrochloride (1.1 mg, 11%). MS m/z 437.3 [M+H]+;1H NMR (methanol-d4) δ: 9.11 (s, 1H), 8.19 (br s, 1H), 8.10 (br s, 1H), 7.90 (dd, J=8.5, 1.3 Hz, 1H), 7.38 (dd, J=8.2, 6.9 Hz, 1H), 5.88-5.95 (m, 1H), 2.52-2.63 (m, 4H), 1.71 (s, 6H), 1.56 (s, 6H); 2 Hs not observed (NH and OH). Example 23 Preparation of Compound 81 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 85 mg, 0.29 mmol), (7-methoxy-6-quinolyl)boronic acid (70 mg, 0.35 mmol), [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(I) (22 mg, 0.029 mmol), and K2CO3(811 mg, 0.58 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). Dioxane (2 mL) and water (0.5 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 30% MeOH) to provide 7-methoxy-6-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)quinolone (110 mg, 91%) as a tan solid. MS m/z 418.4 [M+H]+. Step 2: 7-Methoxy-6-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)quinolone (110 mg, 0.26 mmol) was dissolved in CH2Cl2(2 mL) and 1 N BBr3in dichloromethane (1.3 mL, 1.3 mmol) was added dropwise. The mixture was stirred at room temperature for 16 h. Methanol (5 mL) was added and the reaction was stirred for 2 h. The reaction was concentrated at reduced pressure, the residue was triturated in Et2O, and the resultant precipitate was collected by vacuum filtration, washed with CH2Cl2, Et2O and dried to afford 6-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)quinolin-7-ol hydrobromide (97 mg, 76%) as an orange solid. MS m/z 404.5 [M+H]+;1H NMR (methanol-d4) δ: 9.21-9.25 (m, 2H), 9.11 (dd, J=5.7, 1.3 Hz, 1H), 9.03 (s, 1H), 7.94 (dd, J=8.2, 5.7 Hz, 1H), 7.73 (s, 1H), 6.00-6.08 (m, 1H), 2.60-2.80 (m, 4H), 1.81 (s, 6H), 1.66 (s, 6H); 2 Hs not observed (NH and OH). Example 24 Preparation of Compound 97 Step 1: To a solution of 5-bromo-6-methoxy-2-methyl-1H-benzo[d]imidazole (360 mg, 1.5 mmol) in DMF (5 mL) was added 60% NaH in mineral oil (90 mg, 2.25 mmol) at 0° C. under N2. The mixture was stirred at 0° C. for 15 min and then SEMCl (400 μL, 2.25 mmol) was added. The reaction was stirred at room temperature for 2 h and the mixture was quenched with ice-water (10 mL). The mixture was extracted with EtOAc (50 mL×2). The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated to give the residue, which was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (2% to 5% MeOH) to obtain the mixture of 5-bromo-6-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazole and (5-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazol-6-yl)bromonium as brown oil (416 mg, 75%). MS m/z 371 [M+H]+. Step 2: A mixture of 5-bromo-6-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazole and (5-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazol-6-yl)bromonium (370 mg, 1 mmol), B2(pin)2(280 mg, 1.1 mmol), Pd (dppf)Cl2(73 mg, 0.1 mmol) and KOAc (196 mg, 2 mmol) in 1,4-dioxane (5 mL) was stirred at 90° C. under N2 for 3 h. The solution was concentrated to give a crude mixture of 6-methoxy-2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazole and 2-(5-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazol-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolan-1-ium, which was used in the next step without purification. Step 3: A mixture of above crude 6-methoxy-2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazole and 2-(5-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazol-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolan-1-ium (150 mg, 0.51 mmol), 6-chloro-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (prepared in Example 13, step 2, 147 mg, 0.5 mmol), Pd (dppf)Cl2(73 mg, 0.1 mmol), and K2CO3(178 mg, 1.3 mmol) in 1,4-dioxane-H2O (4 mL, 3/1, v/v) was stirred at 90° C. under N2 for 3 h. The solution was concentrated and the residue was purified by column chromatography on silica gel, eluting with a CH2Cl2/MeOH gradient (0% to 5% MeOH) to give a mixture of 6-(6-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine and 6-(5-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazol-6-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine as brown oil (220 mg, 79%). MS m/z 551 [M+H]+. Step 4: To a solution of a mixture of 6-(6-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine and 6-(5-methoxy-2-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-benzo[d]imidazol-6-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (220 mg, 0.4 mmol) in CH2Cl2(1 mL) was added TFA (912 mg, 8 mmol). The mixture was stirred at room temperature for 16 h. The mixture was concentrated to give crude 6-(6-methoxy-2-methyl-1H-benzo[d]imidazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine, which was used in the next step without further purification (151 mg, 90%). MS m/z 421 [M+H]+. Step 5: To a solution of 6-(6-methoxy-2-methyl-1H-benzo[d]imidazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (150 mg, 0.36 mmol) in CH2Cl2(3 mL) was added 1.0M BBr3in CH2Cl2(3 mL, 3 mmol). The reaction was stirred at room temperature for 16 h, then quenched with MeOH (5 mL) and concentrated. The residue was dissolved in MeOH (with 2.5% NH4OH), filtered, concentrated and purified by prep-HPLC to obtain 2-methyl-5-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-1H-benzo[d]imidazol-6-ol as yellow solid (30 mg, 21%). MS m/z 407.3 [M+H]+;1H NMR (methanol-d4) δ 9.00 (s, 1H), 8.11 (s, 1H), 7.09 (s, 1H), 5.83-5.79 (m, 1H), 2.58 (s, 3H), 2.39-2.24 (m, 4H), 2.18 (s, 1H), 1.50 (s, 6H), 1.35 (s, 6H); 2 Hs not observed (NH and OH). Example 25 Preparation of Compound 92 Step 1: 5-Methoxy-2-nitroaniline (7.2 g, 43 mmol) and NBS (7.5 g, 43 mmol) were dissolved in acetonitrile (70 mL) and cooled to 0° C. Then TFA (3.2 mL, 43 mmol) was added dropwise into the mixture. The ice-bath was removed and the reaction was stirred for 4 h at room temperature. Water (100 mL) was added and the pH was adjusted to 8 by adding 2.5 M NaOH. The formed precipitate was recrystallized from methanol to give 4-bromo-5-methoxy-2-nitroaniline as a yellow solid (9.65 g, 82%). MS m/z 247, 249 [M+H]+ Step 2: To a solution of 4-bromo-5-methoxy-2-nitroaniline (4.92 g, 20 mmol) and TEA (5.6 mL, 40 mmol) in CH2Cl2(50 mL) was added TFAA (5.6 mL, 40 mmol) at 0° C. The reaction mixture was stirred at room temperature for 2 h. The solution was concentrated to give a crude intermediate, which was purified by column chromatography on silica gel, eluting with a EtOAc/hexanes gradient (4% to 10% EtOAc) to obtain N-(4-bromo-5-methoxy-2-nitrophenyl)-2,2,2-trifluoroacetamide as yellow solid (4.5 g, 66%). MS m/z 343,345 [M+H]+. Step 3: To a solution of N-(4-bromo-5-methoxy-2-nitrophenyl)-2,2,2-trifluoroacetamide (3.42 g, 10 mmol) and Cs2CO3(9.78 g, 3 mmol) in DMF (50 mL) was added Mel (3.8 mL, 25 mmol). The mixture was stirred at room temperature for 3 h. 1 M NaOH (10 mL) was added and the reaction was stirred for additional 1 h. The mixture was partitioned between EtOAc and H2O. The organic layer was washed with brine and concentrated to give 4-bromo-5-methoxy-N-methyl-2-nitroaniline as yellow oil without further purification (2.34 g, 90%). MS m/z 261,263 [M+H]+. Step 4: A mixture of 4-bromo-5-methoxy-N-methyl-2-nitroaniline (2.0 g, 7.7 mmol) and Fe (4.3 g, 77 mmol) in formic acid (20 mL) was stirred at 100° C. overnight. The mixture was diluted with MeOH (100 mL). The filtrate was concentrated and then partitioned between EtOAc and H2O. The organic layer was washed with brine, dried over anhydrous Na2SO4and concentrated to give 5-bromo-6-methoxy-1-methyl-1H-benzo[d]imidazole, which was used to the next step without further purification (1.63 g, 88%). 1H NMR (400 MHz, CDCl3) δ 7.97 (s, 1H), 7.76 (s, 1H), 6.83 (s, 1H), 3.96 (s, 3H), 3.81 (s, 3H). Step 5: A mixture of 5-bromo-6-methoxy-1-methyl-1H-benzo[d]imidazole (240 mg, 1 mmol), B2(pin)2(280 mg, 1.1 mmol), Pd(dppf)Cl2(73 mg, 0.1 mmol) and KOAc (196 mg, 2 mmol) in 1,4-dioxane (4 mL) was stirred at 90° C. under N2 for 3 hours. The solution was filtered through Celite and concentrated to give the crude 6-methoxy-1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-benzo[d]imidazole without purification for next step. MS m/z 289 [M+H]+. Step 6: A mixture of above crude 6-methoxy-1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-benzo[d]imidazole, 6-chloro-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 200 mg, 0.68 mmol), Pd (dppf)Cl2(50 mg, 0.068 mmol) and K2CO3(188 mg, 1.36 mmol) in 1,4-dioxane (4 mL) and water (1 mL) was stirred at 90° C. under N2 for 3 h. The solution was concentrated and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 5% MeOH) to give 6-(6-methoxy-1-methyl-1H-benzo[d]imidazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine as brown oil (228 mg, 80%). MS m/z 421 [M+H]+. Step 7: To a solution of 6-(6-methoxy-1-methyl-H-benzo[d]imidazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (100 mg, 0.24 mmol) in CH2Cl2(3 mL) was added 1 M BBr3in CH2Cl2(3 mL, 3 mmol). The reaction was stirred at room temperature for 16 h. The reaction was quenched with MeOH (5 mL) and concentrated. The residue was dissolved in MeOH (with 2.5% NH4OH), filtered and concentrated and purified by prep-TLC eluting with CH2Cl/30% MeOH (with 2.5% NH4OH) to obtain 1-methyl-5-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-1H-benzo[d]imidazol-6-ol, as yellow solid (48 mg, 50%). MS m/z 407.1 [M+H]+;1H NMR (methanol-d4) δ: 8.95 (s, 1H), 8.30 (s, 1H), 7.92 (s, 1H), 7.41 (s, 2H), 7.11 (s, 1H), 5.74 (tt, J=11.0, 5.5 Hz, 1H), 3.87 (s, 3H), 2.10-2.39 (m, 4H), 1.48 (s, 6H), 1.33 (s, 6H). Example 26 Preparation of Compound 94 Step 1: To a solution of 5-bromo-6-methoxy-1H-indazole (250 mg, 1.1 mmol) in DMF (5 mL) was added 60% NaH in mineral oil (66 mg, 1.65 mmol) at 0° C. under N2. The mixture was stirred at 0° C. for 15 min and then SEMCl (300 μL, 1.65 mmol) was added. The reaction was stirred at room temperature for 2 h and then was quenched with ice-water (10 mL). The mixture was extracted with EtOAc (50 mL×2). The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated to give the crude residue, which was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 3% MeOH) to give a mixture of 5-bromo-6-methoxy-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole and 5-bromo-6-methoxy-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-indazole as brown oil (315 mg, 80%). MS m/z 357,359 [M+H]+. Step 2: A mixture of 5-bromo-6-methoxy-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole and 5-bromo-6-methoxy-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-indazole (300 mg, 0.84 mmol), B2(pin)2(235 mg, 0.924 mmol), Pd (dppf)Cl2(61 mg, 0.084 mmol) and KOAc (165 mg, 1.68 mmol) in 1,4-dioxane (5 mL) was stirred at 90° C. under N2 for 3 h. The solution was concentrated to give a crude mixture of 6-methoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole and 6-methoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-indazole, which was used in the next step without further purification. MS m/z 405 [M+H]+. Step 3: A mixture of 6-methoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole and 6-methoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-indazole, 6-chloro-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 170 mg, 0.58 mmol), Pd(dppf)Cl2(42 mg, 0.058 mmol) and K2CO3(199 mg, 1.45 mmol) in 1,4-dioxane-H2O (4 mL) was stirred at 90° C. under N2 for 3 h. The solution was concentrated and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0% to 3% MeOH) to give a mixture of 6-(6-methoxy-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine and 6-(6-methoxy-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-indazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine as brown oil (217 mg, 70%). MS m/z 537 [M+H]+. Step 4: A solution of 6-(6-methoxy-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine and 6-(6-methoxy-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-indazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (217 mg, 0.4 mmol) in CH2Cl2(1 mL) was added TFA (912 mg, 8 mmol). The mixture was stirred at room temperature for 16 h. The mixture was concentrated to give the crude 6-(6-methoxy-1H-indazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine trifluoroacetic acid salt, which was used in the next step without further purification (150 mg, 90%). MS m/z 407 [M+H]+. Step 5: To a solution of 6-(6-methoxy-1H-indazol-5-yl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (150 mg, 0.37 mmol) in CH2Cl2(3 mL) was added 1 M BBr3in CH2Cl2(3 mL, 3 mmol). The reaction was stirred at room temperature for 16 h. The reaction was quenched with MeOH (5 mL) and concentrated. The residue was dissolved in MeOH (with 2.5% NH4OH), filtered and concentrated to give the crude product, which was purified by prep-TLC eluting with CH2C/30% OMeOH (with 2.5% NH4OH) to obtain 5-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-1H-indazol-6-ol as yellow solid (70 mg, 48%). MS m/z 393.8 [M+H]+;1H NMR (DMSO-d6) δ: 12.79 (br s, 1H), 11.00 (br s, 1H), 8.98 (s, 1H), 8.32 (s, 1H), 8.06 (s, 1H), 7.06 (s, 1H), 5.58-5.87 (m, 1H), 2.17-2.35 (m, 4H), 1.43 (s, 6H), 1.26 (s, 6H); 1H not observed (NH). Example 27 Preparation of Compound 82 Step 1: A mixture of 6-methoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,3-dihydro-1H-inden-1-one (1.04 g, 3.6 mmol), 6-chloro-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (prepared in example 13, step 2, 882 mg, 3 mmol), Pd (dppf)Cl2(220 mg, 0.3 mmol) and K2CO3(828 mg, 6 mmol) in 1,4-dioxane (12 mL) and water (3 mL) was stirred at 90° C. under N2 for 3 h. The solution was concentrated and the residue was purified by silica gel column chromatography eluting with a MeOH/CH2Cl2gradient (0 to 5% MeOH) to afford 6-methoxy-5-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-2,3-dihydro-1H-inden-1-one as an orange-yellow solid (1 g, 79%). MS m/z 421 [M+H]+;1H NMR (CDCl3) δ 8.75 (s, 1H), 8.18 (s, 1H), 7.40 (d, J=5.0 Hz, 1H), 5.85-5.67 (m, 1H), 3.95 (s, 3H), 3.26-3.13 (m, 2H), 2.88-2.72 (m, 2H), 2.28 (d, J=7.3 Hz, 4H), 1.27 (d, J=21.2 Hz, 12H). Step 2: A mixture of 6-methoxy-5-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-2,3-dihydro-1H-inden-1-one (210 mg, 0.5 mmol), NH2OH·HCl (69 mg, 1 mmol) and Et3N (0.17 mL, 1.25 mmol) in EtOH (4 mL) was stirred at 90° C. for 4 h. The reaction mixture was cooled to room temperature. The precipitate was collected by filtration, washed with Et2O and dried to afford 6-methoxy-5-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-2,3-dihydro-1H-inden-1-one oxime as white solid (174 mg, 80%). MS m/z 436 [M+H]+; Step 3: To a solution of 6-methoxy-5-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-2,3-dihydro-1H-inden-1-one oxime (66 mg, 0.15 mmol) in CH2Cl2(3 mL) was added 1 M BBr3in CH2Cl2(2 mL, 2 mmol). The reaction was stirred at room temperature for 16 h, then quenched with MeOH (5 mL) and concentrated. The residue was dissolved in MeOH (with 2.5% NH4OH), filtered, concentrated and then purified by prep-HPLC to afford 6-hydroxy-5-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-2,3-dihydro-1H-inden-1-one oxime as yellow solid (33 mg, 52%). MS m/z 421.9 [M+H]+;1H NMR (DMSO-d6) δ: 10.93-11.19 (m, 2H), 9.02 (s, 1H), 7.96 (s, 1H), 7.22 (s, 1H), 5.67 (tt, J=12.3, 3.4 Hz, 1H), 2.91-3.08 (m, 2H), 2.74-2.91 (m, 2H), 2.20 (dd, J=12.1, 3.3 Hz, 2H), 2.11 (t, J=12.3 Hz, 2H), 1.35 (s, 6H), 1.18 (s, 6H); 1H not observed (NH or OH). Example 28 Preparation of Compound 42 Step 1: A mixture of 6-chloro-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,4-diamine (prepared in example 13, step 1, 200 mg, 0.35 mmol) in triethylorthoformate (8 mL) and aq. 4N HCl (1 drop) was stirred at 100° C. for 24 h. The crude reaction mixture was diluted with MeOH to afford a clear solution and concentrated. The residue was purified by silica gel column chromatography eluting with a MeOH (2.5% NH4OH)/CH2Cl2gradient (0 to 20% MeOH/NH4OH) to afford 3-chloro-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazine (144 mg, 70%) as a clear oil that solidified under high vacuum. MS m/z 294.5 [M+H]+. Step 2: To a suspension of 3-chloro-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazine (95 mg, 0.32 mmol) in CHCl3(0.5 mL) and MeOH (0.5 mL) was added N-bromosuccinimide (178 mg, 0.98 mmol). The reaction was heated at 70° C. for 48 h. Solvents were removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with a MeOH (with 2.5% NH4OH)/CH2Cl2gradient (0 to 10% MeOH/NH4OH) to afford 6-bromo-3-chloro-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazine (80 mg, 66%) as a white solid. Step 3: To a solution of 6-bromo-3-chloro-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazine (25 mg, 0.07 mmol) in MeOH (1 mL) was added 8 M methylamine in MeOH (63 μL, 0.5 mmol). The reaction was stirred at 50° C. until complete conversion of starting material was obtained. The reaction was concentrated and purified by silica gel column chromatography eluting with a MeOH (with 2.5% NH4OH)/CH2Cl2gradient (0 to 30% MeOH/NH4OH) to afford 3-chloro-N-methyl-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazin-6-amine (20 mg, 65%) as a clear solid. MS m/z 323.2 [M+H]+. Step 4: A mixture of 3-chloro-N-methyl-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazin-6-amine (20 mg, 0.062 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(I) (3.3 mg, 0.004 mmol), and 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (prepared in example 1, step 7, 32 mg, 0.077 mmol) in 1,4-dioxane (1 mL) was purged with argon for 10 min. Then a solution of potassium carbonate (15 mg, 0.11 mmol) in water (0.2 mL) was added and the reaction mixture was heated to 90° C. for 3 h. The reaction was diluted with EtOAc and filtered through a small pad of Celite (washing with 20% MeOH/CH2Cl2). The organic solution was concentrated and purified by silica gel column chromatography eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to afford 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-N-methyl-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazin-6-amine (17 mg, 68%) as a light brown solid. MS m/z 575.4 [M+H]+. Step 5: To 3-[2-(methoxymethoxy)-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)phenyl]-N-methyl-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazin-6-amine (17 mg, 0.03 mmol) was added 4 N HCl in dioxane (1 mL, 4 mmol). The reaction was stirred for 4 h and then filtered to collect the solid precipitate. The solid was further washed with diethyl ether and dried to afford 2-[6-(methylamino)-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazin-3-yl]-5-(1H-pyrazol-4-yl)phenol dihydrochloride as a yellow solid (12.6 mg, 60%). MS m/z 447.9 [M+H]+;1H NMR (methanol-d4) δ: 8.79 (s, 1H), 8.50-8.62 (m, 2H), 7.44-7.55 (m, 2H), 7.40 (s, 1H), 5.26-5.41 (m, 1H), 3.85 (s, 3H), 2.50-2.64 (m, 2H), 2.32-2.49 (m, 2H), 1.71 (s, 6H), 1.63 (s, 6H); 4 Hs not observed (3 NHs and OH). Using the procedure described for Example 28, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData9MS m/z 434.4 [M + H]+;1H NMR (methanol-d4) δ: 8.15 (br s, 2H), 7.99 (s, 1H), 7.69(d, J = 8.2 Hz, 1H), 7.40 (dd, J = 7.6, 1.3 Hz, 1H), 7.31 (d, J = 1.9 Hz, 1H), 4.96-5.07 (m,1H), 2.71 (t, J = 13.6 Hz, 2H), 2.18 (dd, J = 13.6, 3.5 Hz, 2H), 1.66 (s, 6H), 1.58 (s, 6H);4 Hs not observed (2 OH and 2 NH).40MS m/z 448.2 [M + H]+;1H NMR (methanol-d4) δ : 9.13 (s, 1H), 8.26 (s, 2H), 7.55 (d,J = 8.2 Hz, 1H), 7.38 (d, J = 1.3 Hz, 1H), 7.31 (d, J = 1.3 Hz, 1H), 5.40-5.51 (m, 1H),4.79 (s, 3H), 2.54-2.70 (m, 4H), 1.73 (s, 6H), 1.65 (s, 6H); 3 Hs not observed (2 NHsand OH).49MS m/z 461.9 [M + H]+;1H NMR (methanol-d4) δ: 8.78 (s, 1H), 8.28-8.36 (m, 2H),7.34-7.50 (m, 2H), 7.30 (s, 1H), 5.25-5.41 (m, 1H), 4.11-4.15 (q, J = 6.0 Hz, 2H),2.51-2.65 (m, 2H), 2.33-2.49 (m, 2H), 1.71 (s, 6H), 1.63 (s, 6H), 1.38-1.41 (t, J = 6.0Hz, 3H); 4 Hs not observed (3 NHs and OH). Example 29 Preparation of Compound 3 Step 1: An oven-dried flask was equipped with a magnetic stir bar and charged with 3-chloro-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine (prepared in example 29, step 1, 60 mg, 0.15 mmol), tetrakis(triphenylphosphine)palladium(0) (25 mg, 0.015 mmol), and 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (prepared in example 1, step 7, 75 mg, 0.15 mmol), and Na2CO3(46 mg, 0.45 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (3 mL) and water (0.4 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water (2 mL), and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to provide 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine (60 mg, 75%) as a yellow solid. MS m/z 546.2 [M+H]+. Step 2: 3-(2-(Methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine (35 mg, 0.05 mmol) was dissolved in 1 mL of methanol. 4 N HCl in 1,4-dioxane (500 μL, 2 mmol) was added and the reaction stirred at room temperature for 2 h. The reaction was concentrated, triturated with 20% MeOH/ether, and the precipitate was filtered, and dried to afford 5-(1H-pyrazol-4-yl)-2-(7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl)phenol (25 mg, 86%). MS m/z 418.1 [M+H]+,1H NMR (methanol-d4) δ: 9.19 (s, 1H), 8.86 (s, 1H), 8.15 (s, 2H), 7.86 (d, J=8.2 Hz, 1H), 7.39 (dd, J=8.2, 1.9 Hz, 1H), 7.32 (d, J=1.6 Hz, 1H), 5.39-5.58 (m, 1H), 2.68 (t, J=13.9 Hz, 2H), 2.48 (dd, J=13.9, 3.5 Hz, 2H), 1.72 (s, 6H), 1.63 (s, 6H); 3 Hs not observed (OH and 2 NHs). Using the procedure described for Example 29, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData4MS m/z 438.3 [M + H]+;1H NMR (methanol-d4) δ: 9.29 (s, 1H), 8.74 (d, J = 0.9 Hz,1H), 8.41 (d, J = 1.3 Hz, 2H), 7.84-7.95 (m, 2H), 5.44-5.65 (m, 1H), 2.73 (t, J = 13.9 Hz,2H), 2.51 (dd, J = 13.9, 3.2 Hz, 2H), 1.74 (s, 6H), 1.65 (s, 6H); 2 Hs not observed (2NHs). Example 30 Preparation of Compound 5 Step 1: A mixture of 6-chloro-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,4-diamine (prepared in example 13, step 1, 180 mg, 0.63 mmol) in triethylorthoacetate (4 mL) and HCOOH (0.2 mL) was stirred at 100° C. for 24 h. The reaction was then cooled to room temperature and the precipitate was collected by filtration and dried under vacuum to afford 3-chloro-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazine (112 mg, 56%). MS m/z 308.2 [M+H]+. Step 2: An oven-dried flask was equipped with a magnetic stir bar and charged with 3-chloro-6-methyl-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazine (50 mg, 0.14 mmol), tetrakis(triphenylphosphine)palladium(0) (25 mg, 0.015 mmol), 4-[3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1-tetrahydropyran-2-yl-pyrazole (prepared in example 1, step 7, 75 mg, 0.15 mmol), and Na2CO3(46 mg, 0.45 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (1 mL) and water (0.25 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water, and extracted with EtOAc three times. The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to provide 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-6-methyl-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine (60 mg, 66%) as a yellow solid. MS m/z 560.5 [M+H]+. Step 3: To a solution of 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-6-methyl-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazine (45 mg, 0.05 mmol) in 1 mL of methanol was added 4 N HCl in 1,4-dioxane (500 μL, 2 mmol). The reaction was stirred at room temperature for 2 h. The reaction was concentrated, the residue was triturated in 20% MeOH/ether, and the precipitate was filtered and dried to afford 2-(6-methyl-7-(2,2,6,6-tetramethylpiperidin-4-yl)-7H-imidazo[4,5-c]pyridazin-3-yl)-5-(1H-pyrazol-4-yl)phenol hydrochloride (30 mg, 88%) as a yellow solid. MS m/z 432.5 [M+H]+;1H NMR (methanol-d4) δ: 8.74 (s, 1H), 8.31-8.44 (m, 2H), 7.79 (d, J=8.2 Hz, 1H), 7.45 (dd, J=8.2, 1.6 Hz, 1H), 7.37 (s, 1H), 5.05-5.20 (m, 1H), 3.02 (s, 3H), 2.96 (t, J=13.6 Hz, 2H), 2.30-2.40 (m, 2H), 1.71 (s, 6H), 1.63 (s, 6H); 3 Hs not observed (OH and 2 NHs). Using the procedure described for Example 30, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData6MS m/z 452.4 [M + H]+;1H NMR (methanol-d4) δ: 8.65 (s, 1H), 8.38 (d, J = 1.6 Hz,2H), 7.90 (dd, J = 11.3, 6.0 Hz, 1H), 7.85 (dd, J = 10.7, 6.0 Hz, 1H), 5.09-5.22 (m, 1H),3.05 (s, 3H), 3.02 (t, J = 13.6 Hz, 2H), 2.37 (dd, J = 13.6, 3.8 Hz, 2H), 1.72 (s, 6H), 1.63(s, 6H); 2 Hs not observed (2 NHs). Example 31 Preparation of Compound 1 Step 1: To a solution of 3,6-dibromopyrazin-2-amine (504 mg, 2 mmol) and 2,2,6,6-tetramethylpiperidin-4-amine (0.35 mL, 2 mmol) in EtOH (2 mL) was added DIEA (0.38 mL, 2 mmol). The reaction mixture was subjected to microwave irradiation at 180° C. for 3.5 h. The reaction mixture was cooled and concentrated. The residue was purified by silica gel column chromatography eluting with a MeOH (2.5% NH4OH)/CH2Cl2gradient (0-30% MeOH/NH4OH) to provide 5-bromo-N2-(2,2,6,6-tetramethylpiperidin-4-yl)pyrazine-2,3-diamine (0.35 g, 54%). MS m/z 328.0, 330.0 [M+H]+. Step-2: 5-Bromo-N2-(2,2,6,6-tetramethylpiperidin-4-yl)pyrazine-2,3-diamine (0.18 mg, 0.54 mmol) was dissolved in formic acid (0.36 mL) and the resulting solution was heated to 100° C. for 3 h. The solution was concentrated to give crude 5-bromo-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazine (0.18 g, 97%). MS m/z 338.1, 340.1 [M+H]+. Step-3: An oven-dried flask was equipped with a magnetic stir bar and charged with 5-bromo-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazine (50 mg, 0.15 mmol), 4-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (prepared in example 1, step 7, 62 mg, 0.15 mmol), tetrakis(triphenylphosphine)palladium(0) (25 mg, 0.015 mmol), and Na2CO3(46 mg, 0.45 mmol). The flask was sealed with a rubber septum, and then evacuated and backfilled with argon (repeated a total of 3×). 1,4-Dioxane (3 mL), water (0.4 mL) were added and the reaction was heated to 90° C. for 16 h. The reaction was cooled to room temperature, diluted with water (2 mL), and extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, concentrated under reduced pressure, and purified by column chromatography, eluting with a MeOH/CH2Cl2gradient (0-20% MeOH) to provide 5-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazine (60 mg, 75%) as a yellow solid. MS m/z 546.4 [M+H]+. Step-4: 5-(2-(Methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazine (30 mg, 0.05 mmol) was dissolved in methanol (1 mL), then 4 N HCl in 1,4-dioxane (500 μL, 2 mmol) was added and the reaction stirred at room temperature for 2 h. The reaction was concentrated, triturated with 20% MeOH/ether, and the precipitate was filtered and dried to afford 5-(1H-pyrazol-4-yl)-2-(1-(2,2,6,6-tetramethylpiperidin-4-yl)-1H-imidazo[4,5-b]pyrazin-5-yl)phenol hydrochloride (22 mg, 86%) as a yellow solid. MS m/z 418.5 [M+H]+;1H NMR (methanol-d4) δ: 9.29 (s, 1H), 8.96-9.07 (m, 1H), 8.32 (s, 2H), 8.08-8.15 (m, 1H), 7.21-7.39 (m, 2H), 5.29-5.44 (m, 1H), 2.59-2.72 (m, 2H), 2.39-2.49 (m, 2H), 1.70 (s, 6H), 1.59 (s, 6H); 3 Hs not observed (OH and 2 NHs). Using the procedure described for Example 31, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData2MS m/z 438.4 [M + H]+;1H NMR (methanol-d4) δ: 9.33 (s, 1H), 9.09 (d, J = 1.6 Hz,1H), 8.36 (d, J = 1.3 Hz, 2H), 7.97 (dd, J = 11.7, 6.3 Hz, 1H), 7.77 (dd, J = 12.0, 6.3 Hz,1H), 5.39-5.49 (m, 1H), 2.67 (t, J = 13.9 Hz, 2H), 2.46 (dd, J = 13.9, 3.5 Hz, 2H), 1.71(s, 6H), 1.61 (s, 6H). 2 Hs not observed (2 NHs). Example 32 Preparation of Compound 140 Step 1: To a solution of 5-bromo-2-chloro-pyridin-3-ol (5 g, 23.9 mmol) in DMF (50 mL) was added sodium hydride (1.2 g, 30 mmol, 60 mass % in mineral oil), and the reaction mixture was stirred for 30 min at room temperature. MOMCl (2.2 mL, 29.1 mmol) was added and the reaction was stirred for an additional hour. The reaction was quenched with water and partitioned between EtOAc and water. The combined organic layers were dried over MgSO4and concentrated. The crude product was purified by silica gel chromatography eluting with a EtOAc/hexanes gradient (0-25% EtOAc) to afford 5-bromo-2-chloro-3-(methoxymethoxy)pyridine (4.8 g) as white solid. 1H NMR (CDCl3) δ: 8.14 (d, J=2.1 Hz, 1H), 7.66 (d, J=2.0 Hz, 1H), 5.30 (s, 2H), 3.55 (s, 3H). Step 2: A mixture of 5-bromo-2-chloro-3-(methoxymethoxy)pyridine (1 g, 3.96 mmol), 1-(tetrahydro-2H-pyran-2-yl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (1.5 g, 5.40 mmol), 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichlormethane complex (0.33 g, 0.40 mmol) and potassium acetate (1.3 g, 13 mmol) was purged with argon. 1,4-Dioxane (12 mL) and water (3 mL) were added and the reaction mixture was heated to 90° C. for 2 h. The reaction was cooled to room temperature, filtered through Celite, and washed with MeOH. The organic layers were concentrated and the residue chromatographed on silica gel, eluting with a EtOAc/hexanes gradient (0-50% EtOAc) to afford 2-chloro-3-(methoxymethoxy)-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)pyridine (1.2 g, 93%). MS m/z 324.2 [M+H]+,1H NMR (CDCl3) δ: 8.20 (s, 1H), 7.92 (s, 1H), 7.82 (s, 1H), 7.56 (d, J=2.0 Hz, 1H), 5.42 (d, J=6.6 Hz, 1H), 5.32 (s, 2H), 4.08-4.16 (m, 1H), 3.74 (td, J=11.2, 2.8 Hz, 1H), 3.55 (s, 3H), 2.03-2.18 (m, 3H), 1.61-1.78 (m, 3H). Step 3: To a microwave vial were added 2-chloro-3-(methoxymethoxy)-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)pyridine (110 mg, 0.34 mmol), tributyl(tributylstannyl)stannane (410 mg, 0.71 mmol), tetrakis(triphenylphosphine)palladium(0) (40 mg, 0.03 mmol), and lithium chloride (90 mg, 2.0 mmol). The mixture was purged with argon. 1,4-Dioxane (2 mL) was added, and the reaction was heated in the microwave for 1.5 h at 150° C. The reaction was cooled to room temperature, filtered through Celite, and washed with MeOH. The organic layers were concentrated and the residue was purified using silica gel column chromatography eluting with a EtOAc/hexanes gradient (0-50% EtOAc) to afford 3-(methoxymethoxy)-5-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)-2-(tributylstannyl)pyridine (106 mg, 55%). MS m/z 580.6 [M+H]+,1H NMR (CDCl3) δ: 8.58 (s, 1H), 8.13 (s, 1H), 7.88 (s, 1H), 7.40 (s, 1H), 5.38-5.45 (m, 1H), 4.03 (br d, J=11.6 Hz, 1H), 3.93-4.07 (m, 1H), 3.66-3.77 (m, 1H), 3.63 (s, 1H), 2.16 (s, 3H), 1.26-1.40 (m, 18H), 1.11-1.18 (m, 6H), 0.90 (t, J=7.3 Hz, 9H). Step 4: To a microwave vial were added 3-(methoxymethoxy)-5-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)-2-(tributylstannyl)pyridine (0.11 g, 0.19 mmol), 6-chloro-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (300 mg, 0.10 mmol) tetrakis(triphenylphosphine)palladium(0) (12 mg, 0.01 mmol) and 1,4-dioxane (2 mL). The mixture was sparged with argon and heated in the microwave for 1.5 h at 150° C. The solvent was removed, and the crude mixture was purified by silica gel chromatography using a MeOH/CH2Cl2gradient (0-15% MeOH) to afford 6-(2-(methoxymethoxy)-4-(4-methyl-1H-imidazol-1-yl)phenyl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)-1,2,4-triazin-3-amine (0.04 g, 72%) with minor impurities. MS m/z 548.6 [M+H]+. The compound was used in the next step without additional purification. Step 5: To a solution of 6-(2-(methoxymethoxy)-4-(4-methyl-1H-imidazol-1-yl)phenyl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)-1,2,4-triazin-3-amine (0.02 g, 0.04 mmol) in MeOH (0.5 mL) was added 4.0 M HCl in dioxane (1 mL). The mixture was stirred for 1 h at room temperature. The solvent was removed, and the crude mixture was purified by silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-15% MeOH) to afford 5-(1H-pyrazol-4-yl)-2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)pyridin-3-ol (7 mg) as yellow solid. MS m/z 420.5 [M+H]+,1H NMR (methanol-d4) δ: 9.13-9.20 (m, 1H), 8.48 (s, 1H), 8.17 (d, J=8.9 Hz, 1H), 7.56 (m, 2H), 5.97 (dt, J=11.1, 5.5 Hz, 1H), 2.65-2.75 (m, 4H), 1.77-1.80 (m, 6H), 1.64 (s, 6H); 3Hs not observed (1 OH and 2 NH). Example 33 Preparation of Compound 136 Step 1: 1-Tetrahydropyran-2-yl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (5.57 g, 20.0 mmol), 5-bromo-2-chloro-3-fluoro-pyridine (4.01 g, 19.05 mmol), [1,1′-bis(diphenylphosphino)ferrocene(dichloropalladium(II) (493.4 mg, 0.60 mmol), 1,4-dioxane (20.0 mL), and aqueous potassium carbonate (2.0 M, 12.0 mL) were combined, purged with argon and stirred at 80° C. for 3 h. The reaction was concentrated, and the residue was purified by silica gel chromatography eluting with a EtOAc/hexanes gradient (0-60% EtOAc) to yield 2-chloro-3-fluoro-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)pyridine (3.89 g, 73%). MS m/z 282.3 [M+H]+. Step 2: Nickel(I) chloride (484.4 mg, 2.04 mmol), triphenylphosphine (2.14 g, 8.14 mmol), and N,N-dimethylformamide (11.0 mL) were combined, degassed with argon, then stirred at 50° C. for 45 min. 2-Chloro-3-fluoro-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)pyridine (286.7 mg, 1.02 mmol) and 3-chloro-7-(2,2,6,6,-tetramethyl-4-piperidyl)-5,6-dihydropyrrolo[2,3-c]pyridazine (300.0 mg, 1.02 mmol) were added, the reaction degassed with argon, and then stirred at 50° C. for 16 h. The reaction was partitioned between CH2Cl2, MeOH, brine, and aqueous ammonium hydroxide (30%) (roughly 9:1:5:5). The aqueous layer was extracted twice with CH2Cl2/MeOH (9:1) and the combined organic phases were washed with brine, dried over Na2SO4, and then filtered and concentrated. The residue was purified by silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-30% MeOH with 2.5% v/v 30% aqueous ammonium hydroxide additive) to yield 3-[3-fluoro-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-7-(2,2,6,6-tetramethyl-4-piperidyl)-5,6-dihydropyrrolo[2,3-c]pyridazine (735.0 mg, 19%). MS m/z 506.4 [M+H]+. Step 3: 3-[3-Fluoro-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-7-(2,2,6,6-tetramethyl-4-piperidyl)-5,6-dihydropyrrolo[2,3-c]pyridazine (413.7 mg, 0.82 mmol) was dissolved in methanolic sodium methoxide (25 wt %, 15.0 mL) and stirred at 50° C. for 3 h. The reaction was partitioned between CH2Cl2and H2O, the aqueous layer was extracted with CH2Cl2and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was combined with manganese dioxide (activated, 3.56 g, 41.11 mmol) in CH2Cl2(6.0 mL) and stirred at 50° C. in a sealed tube for 16 h. The reaction was filtered through Celite and rinsed with minimal CH2Cl2. The filtrate was combined with manganese dioxide (activated, 3.7 g, 42.5 mmol) and stirred at 60° C. in a sealed tube for 24 h. The reaction was filtered through Celite, rinsed with CH2Cl2/MeOH, and the filtrate was concentrated. The residue was purified by silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-100% MeOH with 2.5% v/v 30% aqueous ammonium hydroxide additive) to yield 3-[3-methoxy-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-7-(2,2,6,6-tetramethyl-4-piperidyl)pyrrolo[2,3-c]pyridazine (47.3 mg, 11%). MS m/z 516.3 [M+H]+. Step 4: 3-[3-Methoxy-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-7-(2,2,6,6-tetramethyl-4-piperidyl)pyrrolo[2,3-c]pyridazine (47.3 mg, 0.09 mmol) and boron tribromide (1.0 M in CH2Cl2, 2.0 mL, 2.0 mmol) were combined and stirred at room temperature under argon for 20 h. The reaction was reverse quenched into MeOH and concentrated. The residue was chromatographed on a reversed phase C18 column, eluting with a 0-100% CH3CN in H2O (0.1% v/v TFA additive) gradient to yield 5-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-4-piperidyl)pyrrolo[2,3-c]pyridazin-3-yl]pyridin-3-ol dihydrochloride (33.6 mg, 75%). MS m/z 418.4 [M+H]+,1H NMR (methanol-d4) δ: 9.53 (s, 1H), 8.79 (d, J=1.8 Hz, 1H), 8.71 (d, J=3.1 Hz, 1H), 8.67 (br s, 2H), 7.86 (s, 1H), 7.20 (d, J=3.4 Hz, 1H), 5.57 (tt, J=13.1, 2.8 Hz, 1H), 2.61 (t, J=13.1 Hz, 2H), 2.42 (dd, J=13.4, 2.7 Hz, 2H), 1.74 (s, 6H), 5.55 (s, 6H), 3 Hs not observed (2 NHs and OH). Example 34 Preparation of Compound 141 Step 1: Nickel(II) chloride hexahydrate (515.8 mg, 10.6 mmol), triphenylphosphine (11.1 g, 42.3 mmol), and N,N-dimethylformamide (52.5 mL) were combined, degassed with argon, and then stirred at 50° C. for 45 min. 2-Chloro-3-fluoro-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)pyridine (1.49 g, 5.3 mmol) and 6-chloro-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,4-diamine (1.5 g, 5.3 mmol) were added, the reaction was degassed with argon, and stirred at 50° C. for 16 h. The reaction was partitioned between CH2Cl2, MeOH, brine, and aqueous ammonium hydroxide (30%) (roughly 9:1:5:5). The aqueous layer was extracted twice with CH2Cl2/MeOH (9:1) and the combined organic phases were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was chromatographed on a reversed phase C18 column, eluting with 0-100% CH3CN in H2O to yield 6-[3-fluoro-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,4-diamine (540.0 mg, 21%). MS m/z 495.5 [M+H]+. Step 2: 6-[3-Fluoro-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,4-diamine (102.0 mg, 0.21 mmol) was dissolved in methanolic sodium methoxide (25 wt %, 4.0 mL) and stirred at 50° C. for 2 h. The reaction was partitioned between CH2Cl2and H2O, the aqueous layer extracted with CH2Cl2and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-100% MeOH with 2.5% v/v 30% aqueous ammonium hydroxide additive) to yield 6-[3-methoxy-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,4-diamine (48.1 mg, 46%). MS m/z 507.4 [M+H]+. Step 3: 6-[3-Methoxy-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-N3-(2,2,6,6-tetramethyl-4-piperidyl)pyridazine-3,4-diamine (48.1 mg, 0.095 mmol), N,N-dimethylformamide (3.0 mL), and Bredereck's reagent (0.2 mL, 0.99 mmol) were combined and stirred at 100° C. for 20 min. The reaction was concentrated to dryness. The residue was partitioned between brine and CH2Cl2, and the aqueous layer was extracted with CH2Cl2. The combined organic layers were dried over Na2SO4, filtered and concentrated to yield 3-[3-methoxy-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazine (50.9 mg, 104%). MS m/z 517.3 [M+H]+. Step 4: 3-[3-Methoxy-5-(1-tetrahydropyran-2-ylpyrazol-4-yl)-2-pyridyl]-7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazine (50.9 mg, 0.099 mmol) and boron tribromide (1.0 M in CH2Cl2, 2.0 mL, 2.0 mmol) were combined and stirred at room temperature under argon for 16 h. The reaction was reverse quenched into MeOH and concentrated. The residue was purified on a reverse phase C18 column, eluting with 0-100% CH3CN in H2O (0.1% v/v TFA additive), and subsequently chromatographed on silica gel, eluting with 0-100% MeOH (2.5% v/v 30% aqueous ammonium hydroxide additive) in CH2Cl2to yield 5-(1H-pyrazol-4-yl)-2-[7-(2,2,6,6-tetramethyl-4-piperidyl)imidazo[4,5-c]pyridazin-3-yl]pyridin-3-ol (12.3 mg, 25%). MS m/z 419.4 [M+H]+;1H NMR (methanol-d4) δ: 9.28 (s, 1H), 9.22 (s, 1H), 8.71 (s, 1H), 8.52 (s, 2H), 8.01 (s, 1H), 5.50 (br t, J=12.7 Hz, 1H), 2.67 (br t, J=13.0 Hz, 2H), 2.41 (br d, J=11.6 Hz, 2H), 1.64 (s, 6H), 1.56 (s, 6H), 3 Hs not observed (2 NHs and OH). Example 35 Preparation of Compound 117 Step 1: 1-Tetrahydropyran-2-yl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (105.0 mg, 0.38 mmol), 4,7-dibromo-1H-benzotriazole (195.7 mg, 0.71 mmol), [1,1′-bis(diphenylphosphino(ferrocene]dichloropalladium(I) (16.1 mg, 0.020 mmol), 1,4-dioxane (2.0 mL) and aqueous potassium carbonate (1.0 M, 1.0 mL) were combined and stirred at 80° C. for 16 h. The reaction was partitioned between EtOAc, H2O, and AcOH, and the aqueous layer was extracted once with EtOAc. The combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified using silica gel chromatography eluting with a EtOAc/hexanes gradient (0-100% EtOAc) to yield 7-bromo-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)-1H-benzotriazole (67.6 mg, 51%). MS m/z 348.2 [M+H]+. Step 2: 7-Bromo-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)-1H-benzotriazole (67.6 mg, 0.19 mmol), cesium carbonate (238.5 mg, 0.73 mmol), acetonitrile (2.0 mL), and 1-(chloromethyl)-4-methoxy-benzene (70.0 μL, 0.516 mmol) were combined and stirred at room temperature for 18 h. The reaction was concentrated and the residue was partitioned between EtOAc and H2O. The aqueous layer was extracted once with EtOAc, and the combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified using silica gel chromatography eluting with a EtOAC/hexanes gradient (0-100% EtOAc) to yield 7-bromo-1-[(4-methoxyphenyl)methyl]-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)benzotriazole (26.4 mg, 29%). MS m/z 490.3 [M+Na]+. Step 3: 7-Bromo-1-[(4-methoxyphenyl)methyl]-4-(1-tetrahydropyran-2-ylpyrazol-4-yl)benzotriazole (26.4 mg, 0.056 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II) (4.8 mg, 0.0065 mmol), bis(pinacolato)diboron (18.5 mg, 0.072 mmol), and potassium acetate (dried at 250° C. under vacuum immediately prior to using, 21.7 mg, 0.22 mmol), and 1,4-dioxane (1.0 mL) were combined, degassed with argon, and stirred at 110° C. for 1 h. 6-chloro-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (from Example 13, step 2, 16.6 mg, 0.0563 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)palladium(II) (4.1 mg, 0.006 mmol), and aqueous potassium carbonate (1.0 M, 0.5 mL) were added, the solution degassed with argon, and then stirred at 80° C. for 64 h. The reaction was partitioned between EtOAc and H2O, and the aqueous layer was extracted once with EtOAc. The combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-30% MeOH) to yield 6-[3-[(4-methoxyphenyl)methyl]-7-(1-tetrahydropyran-2-ylpyrazol-4-yl)benzotriazol-4-yl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (9.2 mg, 25%). MS m/z 648.7 [M+H]+. Step 4: 6-[3-[(4-Methoxyphenyl)methyl]-7-(1-tetrahydropyran-2-ylpyrazol-4-yl)benzotriazol-4-yl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine (9.2 mg, 0.014 mmol) was dissolved in trifluoroacetic acid (2.0 mL) and stirred at 60° C. for 2 h. The reaction was concentrated to dryness and the residue was chromatographed on a reverse phase C18 column, eluting with 0-100% CH3CN in H2O (0.1% v/v TFA additive) to yield 6-[7-(1H-pyrazol-4-yl)-3H-benzotriazol-4-yl]-3-(2,2,6,6-tetramethyl-4-piperidyl)triazolo[4,5-c]pyridazine; 2,2,2-trifluoroacetic acid (1.7 mg, 21%). MS m/z 444.5 [M+H]+;1H NMR (methanol-d4) δ: 9.24 (br s, 1H), 8.64 (s, 2H), 8.37-8.41 (m, 1H), 7.91 (d, J=7.6 Hz, 1H), 6.03 (ddd, J=16.1, 10.8, 5.5 Hz, 1H), 2.66-2.74 (m, 4H), 1.76-1.85 (m, 6H), 1.61-1.70 (m, 6H), 3 Hs not observed (NHs). Example 36 Preparation of Compound 105 Step 1: To a solution of methyl 2-hydroxyacetate (218 mg, 2.37 mmol) in THF (3 mL) was added NaH (93 mg, 2.33 mmol, 60 mass %) at 0° C. The reaction was stirred for 30 min at 0° C., and the resultant slurry was added slowly to a solution of methyl 4,6-dichloropyridazine-3-carboxylate (500 mg, 2.37 mmol) in THF (3 mL) at 0° C. The mixture was stirred for 30 min at room temperature. The reaction was quenched with sat. aq. NH4Cl and diluted with EtOAc and H2O. The organic phase was dried over Na2SO4and concentrated. The residue was purified using silica gel chromatography eluting with a EtOAc/hexanes gradient (20-50% EtOAc) to yield methyl 6-chloro-4-(2-methoxy-2-oxoethoxy)pyridazine-3-carboxylate (284 mg, 46%) as a white solid. MS m/z 261.4 [M+H]+. Step 2: To a solution of methyl 6-chloro-4-(2-methoxy-2-oxoethoxy)pyridazine-3-carboxylate (284 mg, 1.09 mmol) in THF (11.0 mL) was added sodium methoxide (0.22 mL, 1.2 mmol, 5.4 mol/L in MeOH) dropwise at room temperature. The reaction was stirred at room temperature for 15 min, then quenched with 1 M HCl. The mixture was partitioned between H2O and EtOAc, and the organic phases were collected and washed with brine and concentrated to afford methyl 3-chloro-7-hydroxyfuro[3,2-c]pyridazine-6-carboxylate (230 mg, 92%) as an off-white solid. MS m/z 229.2 [M+H]+. Step 3: A mixture of 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (18 mg, 0.022 mmol), 4-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (118 mg, 0.29 mmol), methyl 3-chloro-7-hydroxyfuro[3,2-c]pyridazine-6-carboxylate (50 mg, 0.22 mmol), and aqueous 2 M K2CO3(0.22 mL, 0.44 mmol) in dioxane (1 mL) was sparged with argon for 10 minutes, then heated to 90° C. for 3 h. The reaction was cooled to room temperature and filtered through Celite, washing with MeOH. The organic layers were concentrated and the residue was purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (5-20% MeOH) to yield methyl 7-hydroxy-3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)furo[3,2-c]pyridazine-6-carboxylate (75 mg, 71%) as a light brown solid. MS m/z 481.4 [M+H]+. Step 4: To a solution of methyl 7-hydroxy-3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)furo[3,2-c]pyridazine-6-carboxylate (280 mg, 0.58 mmol) in DMSO (7 mL) was added aq. 1M NaOH (1.5 mL, 1.5 mmol). The reaction was heated to 50° C. for 1 h, then cooled to room temperature. DMF (7 mL), Cs2CO3(360 mg, 1.1 mmol) and 1,1,1-trifluoro-N-phenyl-N-((trifluoromethyl)sulfonyl)methanesulfonamide (300 mg, 0.84 mmol) were added, and the mixture stirred at room temperature for 1 h. The reaction mixture was partitioned between CH2Cl2and H2O, and the aqueous layer was extracted once with CH2Cl2. The organic phase was dried over Na2SO4, concentrated, and the residue was purified using silica gel chromatography eluting with a EtOAc/hexanes gradient (20-60% EtOAc) to yield 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)furo[3,2-c]pyridazin-7-yl trifluoromethanesulfonate (43 mg, 13%) as a white solid. MS m/z 555.2 [M+H]+. Step 5: A mixture of 2,2,6,6-tetramethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,6-tetrahydropyridine (27 mg, 0.10 mmol), 1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (7 mg, 0.0085 mmol), 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)furo[3,2-c]pyridazin-7-yl trifluoromethanesulfonate (43 mg, 0.078 mmol), and aqueous 2 M K2CO3(0.12 mL, 0.24 mmol) in dioxane (0.5 mL) was sparged with argon for 10 minutes, then heated to 90° C. for 2 h. The reaction was cooled to room temperature and filtered over celite, washing with MeOH. The organic layers were collected and the residue was purified using silica gel chromatography eluting with a MeOH/CH2Cl2gradient (0-50% MeOH) to yield 3-(2-(methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)furo[3,2-c]pyridazine (7 mg, 16%) as a yellow film. MS m/z 544.5 [M+H]+. Step 6: 3-(2-(Methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)phenyl)-7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)furo[3,2-c]pyridazine (7 mg, 0.013 mmol) was dissolved in MeOH (0.5 mL). HCl (0.3 mL, 1.2 mmol, 4M in dioxane) was added and the reaction was stirred at 40° C. for 30 min. The reaction was concentrated and the residue was purified by reverse-phase column chromatography, eluting with 0-100% MeCN/H2O (0.1% TFA). The product was dissolved in HCl (2 mL, 1.25 M in MeOH) and concentrated to afford 5-(1H-pyrazol-4-yl)-2-(7-(2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridin-4-yl)furo[3,2-c]pyridazin-3-yl)phenol hydrochloride as a yellow film (2.0 mg, 34%). MS m/z 416.5 [M+H]+;1H NMR (methanol-d4) δ: 8.96 (s, 1H), 8.81 (s, 1H), 8.39 (br s, 2H), 7.92 (d, J=8.2 Hz, 1H), 7.47 (d, J=7.6 Hz, 1H), 7.40 (s, 1H), 7.19 (s, 1H), 2.85 (s, 2H), 1.72 (s, 6H), 1.60-1.66 (m, 6H), 3Hs not observed (2 NHs and OH). Example 37 Preparation of Compound 163 Step 1: A mixture of Pd2(dba)3(4.3 mg, 0.0047 mmol), (Me)4tButylXPhos (5.8 mg, 0.012 mmol), toluene (0.6 mL) and 1,4-dioxane (0.15 mL) was sparged with argon, then heated to 120° C. for 5 min and cooled to room temperature. Anhydrous K3PO4(41.0 mg, 0.19 mmol), triazole (9.5 mg, 0.11 mmol), and 3-(methoxymethoxy)-4-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)phenyl trifluoromethanesulfonate (Example 13, step 4, 50.0 mg, 0.091 mmol) were added and the reaction mixture was sparged with argon and stirred at 120° C. for 2 h. Upon completion, the mixture was cooled to room temperature, concentrated and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0-30% MeOH) to give 6-(2-(methoxymethoxy)-4-(2H-1,2,3-triazol-2-yl)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (30.0 mg, 71% yield) as a light yellow solid. MS m/z 464.4 [M+H]+. Step 2: 6-(2-(Methoxymethoxy)-4-(2H-1,2,3-triazol-2-yl)phenyl)-3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (30.0 mg, 0.065 mmol) was dissolved in methanol (0.5 mL) and HCl in dioxane (1.0 mL, 4.0 mmol, 4.0 mol/L) was added. The reaction mixture was stirred at 45° C. for 1 h and then concentrated. The residue was purified by column chromatography on silica gel, eluting with a MeOH/NH4OH/CH2Cl2gradient (0-30% MeOH/2.5% NH4OH) to yield 2-(3-(2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-5-(2H-1,2,3-triazol-2-yl)phenol (19.0 mg, 70% yield) as a yellow solid. MS m/z 420.5 [M+H]+;1H NMR (methanol-d4) δ: 9.09 (s, 1H), 8.19-8.25 (m, 1H), 7.98 (s, 2H), 7.71-7.83 (m, 2H), 5.74-5.94 (m, 1H), 2.31-2.41 (m, 4H), 1.53 (s, 6H), 1.37 (s, 6H); 2 Hs not observed (NH and OH). Example 38 Preparation of Compound 171 Step 1: To a solution of 3,4,6-trichloropyridazine (20.04 g, 106 mmol) in a mixture of THF and DMSO (5:1, 200 mL) was added sodium benzenesulfinate (18.6 g, 111.1 mmol) and the mixture was stirred vigorously at room temperature. Full conversion was observed in 40 minutes. After completion, the reaction mixture was diluted with EtOAc (100 mL) and washed with water and brine. The combined organic phases were dried over MgSO4. The solvent volume was reduced by evaporation. Recrystallization from EtOAc/hexanes yielded 3,6-dichloro-4-(phenylsulfonyl)pyridazine (28.5 g, 93% yield) as a white solid. MS m/z 289.0 [M+H]+;1H NMR (CDCl3) δ: 8.34 (s, 1H), 8.07-7.98 (m, 2H), 7.80-7.74 (m, 1H), 7.68-7.61 (m, 2H). Step 2: To a round bottom flask were added 3,6-dichloro-4-(phenylsulfonyl)pyridazine (1.0 g, 3.46 mmol), (3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-amine (1.3 g, 5.2 mmol), K2CO3(2.18 g, 15.6 mmol) and dioxane (14.0 mL). The mixture was stirred at 100° C. for 16 h, then cooled to room temperature. The reaction was partitioned between EtOAc and water. The organic phase was washed with brine, dried over MgSO4, and the solvent was removed in vacuo to provide crude 6-chloro-N-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-4-(phenylsulfonyl)pyridazin-3-amine which was used in the next step without further purification. MS m/z 427.2, 429.2 [M+H]+;1H NMR (CDCl3) δ: 8.00-7.93 (m, 2H), 7.77 (s, 1H), 7.76-7.73 (m, 1H), 7.64-7.61 (m, 2H), 6.73 (d, J=7.9 Hz, 1H), 5.04-4.88 (m, 1H), 4.34 (d, J=50.0 Hz, 1H), 1.81-1.72 (m, 1H), 1.61-1.49 (m, 1H), 1.29 (s, 6H), 1.22 (s, 6H); 1H (NH) not observed. Step 3: The crude mixture from Step 2 was dissolved in dioxane (8 mL) and DMSO (2 mL). NaN3(400.0 mg, 6.15 mmol) was added, and the mixture was stirred at 50° C. for 16 h, and then cooled to room temperature. The reaction mixture was diluted with EtOAc and washed with brine (4 times) to remove DMSO. The combined organic phases were dried over MgSO4and concentrated to provide crude 4-azido-6-chloro-N-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine as a dark brown oil which was used in the next step without further purification. MS m/z 328.2, 330.2 [M+H]+;1H NMR (CDCl3) δ: 6.88 (s, 1H), 4.90-4.69 (m, 2H), 4.33 (d, J=50.0 Hz, 1H), 1.73-1.63 (m, 1H), 1.46-1.35 (m, 1H), 1.23 (s, 6H), 1.11 (s, 6H); 1H (NH) not observed. Step 4: The crude product from Step 3 was dissolved in CH2Cl2(10 mL) and AcOH (2 mL) and the mixture was cooled at 0° C. Zinc mesh (640.0 mg, 9.8 mmol) was added portionwise and the mixture was stirred for 3 h at 0° C. Upon completion, the reaction was quenched with aqueous saturated NaHCO3. The organic phase was washed with brine, dried over MgSO4, and the solvent was removed in vacuo. The crude product was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0-30% MeOH) to yield 6-chloro-N3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,4-diamine (860.0 mg, 82% yield over 3 steps). MS m/z 302.2, 304.2 [M+H]+;1H NMR (DMSO-d6) δ: 6.46 (s, 2H), 6.41 (s, 1H), 6.02 (d, J=7.5 Hz, 1H), 4.76-4.62 (m, 1H), 4.45 (d, J=55.0 Hz, 1H), 1.61-1.54 (m, 2H), 1.23 (s, 3H), 1.22 (s, 3H), 1.13 (s, 3H), 1.09 (s, 3H); 1H (NH) not observed. Step 5: A solution of 6-chloro-N3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)pyridazine-3,4-diamine (860 mg, 2.8 mmol) in AcOH (6 mL) was cooled to 0° C. NaNO2(280 mg, 4.0 mmol) was dissolved in water (1 mL) and the solution was slowly added dropwise to the reaction mixture. The mixture was then gradually warmed to room temperature and stirred for 1 h at room temperature. After completion, the solvent was removed in vacuo and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0-10% MeOH) to provide 6-chloro-3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine bishydrochloride (450 mg, 50% yield). MS m/z 313.1, 315.1 [M+H]+;1H NMR (DMSO-d6) δ: 9.87 (br s, 1H), 8.96 (s, 1H), 8.59 (br s, 1H), 6.31-6.16 (m, 1H), 5.30 (d, J=45.0 Hz, 1H), 3.08 (t, J=13.6 Hz, 1H), 2.61-2.54 (m, 1H), 1.73 (s, 3H), 1.66 (s, 3H), 1.61 (s, 3H), 1.52 (s, 3H); extra 2Hs are due to bis HCl salt. Step 6: A dry screw cap vial was charged with 6-chloro-3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (200.0 mg, 0.64 mmol), Pd(PPh3)4(70.0 mg, 10 mol %) and 2-(4-chloro-2-(methoxymethoxy)phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (270.0 mg, 0.90 mmol). The vial was evacuated under vacuum and purged with argon, followed by the addition of dioxane (2.2 mL) and aqueous K2CO3solution (2.0 M, 0.8 mL, 1.92 mmol). The mixture was heated at 70° C. for 16 h. After completion, the solvent was removed in vacuo and the residue was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0-15% MeOH) to yield 6-(4-chloro-2-(methoxymethoxy)phenyl)-3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (279.0 mg, 97%) as a brownish solid. MS m/z 449.4, 451.4 [M+H]+. Step 7: A dry screw cap vial was charged with 6-(4-chloro-2-(methoxymethoxy)phenyl)-3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazine (40.0 mg, 0.9 mmol), bis(pinacolato)diboron (30.0 mg, 0.12 mmol), Pd X-Phos G3 (4.0 mg, 0.05 mmol) and KOAc (18.0 mg, 0.18 mmol). The vial was evacuated under vacuum and backfilled with argon. The argon/vacuum cycle was performed at least three times and then dioxane (0.5 mL) was added to the vial under Ar pressure. The reaction was then heated at 100° C. for 2 h. 3-((3S,4S)-3-Fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-6-(2-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3H-[1,2,3]triazolo[4,5-c]pyridazine was used for the next step without isolation. MS m/z 541.4 [M+H]+. Step 8: To the mixture from Step 7, was added 2 M aqueous K2CO3(0.1 mL), Pd X-Phos G3 (4.0 mg, 0.05 mmol) and 4-bromo-1-methylpyridin-2(1H)-one (20.0 mg, 0.11 mmol). The reaction was then heated at 100° C. for 12 h. The crude product was purified by column chromatography on silica gel, eluting with a MeOH/CH2Cl2gradient (0-30% MeOH) to yield 4-(4-(3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-3-(methoxymethoxy)phenyl)-1-methylpyridin-2(1H)-one (35 mg, 75% yield). MS m/z 522.4 [M+H]+. Step 9: To the solution of 4-(4-(3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-3-(methoxymethoxy)phenyl)-1-methylpyridin-2(1H)-one (35.0 mg, 0.067 mmol) in dichloromethane (1.0 mL) and MeOH (100 μL) was added 2.0 M HCl in Et2O (1.5 mL, 0.75 mmol) and the reaction mixture was stirred at room temperature for 5 h. The solvent volume was reduced by evaporation. The residue was purified by column chromatography on silica gel, eluting with a MeOH/NH4OH/CH2Cl2gradient (0-30% MeOH/2.5% NH4OH) to yield 4-(4-(3-((3S,4S)-3-fluoro-2,2,6,6-tetramethylpiperidin-4-yl)-3H-[1,2,3]triazolo[4,5-c]pyridazin-6-yl)-3-hydroxyphenyl)-1-methylpyridin-2(1H)-one (16.0 mg, 50% yield) as a tan solid. MS m/z 478.4 [M+H]+;1H NMR (methanol-d4) δ: 9.19 (s, 1H), 8.23 (d, J=7.9 Hz, 1H), 8.00 (br d, J=6.9 Hz, 1H), 7.49-7.36 (m, 2H), 7.15-7.04 (m, 2H), 6.30-6.21 (m, 1H), 5.42 (d, J=50 Hz 1H), 3.79 (s, 3H), 3.55-3.45 (m, 1H), 2.80-2.69 (m, 1H), 1.88 (s, 3H), 1.81 (s, 3H), 1.74 (s, 3H), 1.67 (s, 3H); 2Hs not observed (NH and OH). Using the procedure described for Example 38, above, additional compounds described herein were prepared by substituting the appropriate starting materials, suitable reagents and reaction conditions, obtaining compounds such as those selected from: CpdData164MS m/z 437.3 [M + H]+;1H NMR (DMSO-d6) δ: 10.74-10.49 (m, 1H), 9.22 (s, 1H),8.47 (br d, J = 12.2 Hz, 1H), 8.15 (s, 2H), 8.13-8.05 (m, 1H), 7.32 (br s, 2H), 6.32-6.16 (m, 1H), 5.37 (d, J = 45 Hz, 1H), 3.09 (t, J = 13.6 Hz, 1H), 2.67-2.57 (m,1H),1.79 (s, 3H), 1.71 (s, 3H), 1.68-1.61 (m, 3H), 1.61-1.49 (m, 3H); 1H not observed(OH).168MS m/z 437.3 [M + H]+;1H NMR (methanol-d4) δ: 9.19 (s, 1H), 8.70 (s, 2H), 8.16 (d,J = 8.1 Hz, 1H), 7.47-7.39 (m, 2H), 6.22-6.10 (m, 1H), 5.52 (dd, J = 50.0, 10.0 Hz,1H), 3.17-3.05 (m, 1H), 2.73-2.62 (m, 1H), 1.89-1.79 (m, 6H), 1.75 (s, 3H), 1.68(s, 3H); 3Hs not observed (OH and 2 NHs).170MS m/z 405.5 [M + H]+;1H NMR (methanol-d4) δ: 9.00 (s, 1H), 8.03 (br d, J = 8.2 Hz,3H), 7.16-7.37 (m, 2H), 5.44-5.61 (m, 1H), 3.52-3.65 (m, 1H), 3.13-3.19 (m, 2H),2.93 (br dd, J = 9.2, 2.5 Hz, 2H), 1.21 (s, 9H); 3 Hs not observed (2NHs and OH).172MS m/z 479.5 [M + H]+;1H NMR (methanol-d4) δ: 9.22 (s, 1H), 9.08 (s, 1H), 8.25 (d,J = 8.1 Hz, 1H), 7.68-7.55 (m, 2H), 7.04 (s, 1H), 6.36-6.15 (m, 1H), 5.42 (d, J =55.0 Hz, 1H), 3.72-3.65 (m, 1H), 3.68 (s, 4H), 2.75 (br d, J = 12.2 Hz, 1H), 1.88 (s,3H), 1.81(s, 3H), 1.74 (s, 3H), 1.67 (s, 3H); 2Hs not observed (NH and OH).173MS m/z 455.4 [M + H]+;1H NMR (DMSO-d6) δ: 12.69 (s, 1H), 11.55 (s, 1H), 9.15 (s,1H), 8.26 (s, 1H), 8.11 (d, J = 10 Hz, 1H), 7.29-7.26 (m, 2H), 6.10-5.90 (m, 1H),5.95 (d, J = 55 Hz, 1H), 2.67-2.57 (m, 1H), 2.19-2.25 (m, 1H), 1.41 (s, 3H), 1.34 (s,3H), 1.26 (m, 3H), 1.14 (m, 3H); 1H not observed.175MS m/z 419.5 [M + H]+;1H NMR (methanol-d4) δ: 9.00 (s, 1H), 8.02 (br d, J = 8.1 Hz,3H), 7.24-7.30 (m, 2H), 5.92-6.00 (m, 1H), 4.30 (br t, J = 8.0 Hz, 1H), 2.98-3.06 (m,1H), 2.75 (br dd, J = 8.6, 4.0 Hz, 1H), 2.55-2.67 (m, 3H), 1.99-2.11 (m, 1H), 1.45 (s,9H); 3 Hs not observed (2 NHs and OH). Biological Examples The following in vitro biological examples demonstrate the usefulness of the compounds of the present description for treating Huntington's disease. To describe in more detail and assist in understanding the present description, the following non-limiting biological examples are offered to more fully illustrate the scope of the description and are not to be construed as specifically limiting the scope thereof. Such variations of the present description that may be now known or later developed, which would be within the purview of one skilled in the art to ascertain, are considered to fall within the scope of the present description and as hereinafter claimed. Compounds of Formula (I) were tested using the Meso Scale Discovery (MSD) Assay provided in International Application No. PCT/US2016/066042, filed on Dec. 11, 2016 and claiming priority to United States Provisional Application U.S. 62/265,652 filed on Dec. 10, 2015, the entire contents of which are incorporated herein by reference. The Endogenous Huntingtin Protein assay used in Example 1 was developed using the ELISA-based MSD electrochemiluminescence assay platform. Example 1 Endogenous Huntingtin Protein Assay Meso Scale Discovery (MSD) 96-well or 384-well plates were coated overnight at 4° C. with MW1 (expanded polyglutamine) or MAB2166 monoclonal antibody (for capture) at a concentration of 1 μg/mL in PBS (30 μL per well). Plates were then washed three times with 300 μL wash buffer (0.05% Tween-20 in PBS) and blocked (100 μL blocking buffer; 5% BSA in PBS) for 4-5 hours at room temperature with rotational shaking and then washed three times with wash buffer. Samples (25 μL) were transferred to the antibody-coated MSD plate and incubated overnight at 4° C. After removal of the lysates, the plate was washed three times with wash buffer, and 25 μL of #5656S (Cell signaling; rabbit monoclonal) secondary antibody (diluted to 0.25 μg/mL in 0.05% Tween-20 in blocking buffer) was added to each well and incubated with shaking for 1 Hour at room temperature. Following incubation with the secondary antibody, the wells were rinsed with wash buffer after which 25 μL of goat anti-rabbit SULFO TAG secondary detection antibody (required aspect of the MSD system) (diluted to 0.25 μg/mL in 0.05% Tween-20 in blocking buffer) was added to each well and incubated with shaking for 1 hour at room temperature. After rinsing three times with wash buffer, 150 μL of read buffer T with surfactant (MSD) were added to each empty well, and the plate was imaged on a SI 6000 imager (MSD) according to manufacturers' instructions provided for 96- or 384-well plates. The resulting IC50values (μM) for compounds tested are shown in Table 1. As shown in Table 1, test compounds described herein had the following IC50values, an IC50value between >3 μM and ≤9 μM is indicated by a single star (*), an IC50value between >1 μM and ≤3 μM is indicated by two stars (**), an IC50value between >0.5 μM and ≤1 μM is indicated by three stars (***), an IC50value between >0.1 μM and ≤0.5 μM is indicated by four stars (****) and an IC50value of ≤0.1 μM is indicated by five stars (*****). TABLE 1CpdIC501****2****3*****4*****5*****6***7*****8**9**10*****11**12****13****14****15**16****17*****18*****19**20*****21**22*****23*****24*****25*****26*****27*****28**29*****30***31*****32****33***34****35*****36****37*****38*****39*****40*42****43***46***47**48****49**50****51****52***53*****54*****55****56****57*****58****59*****60**61**62*****63*****64**65***66*****67*****68*****69*****70*****71****72****73*****74****75****76****77*****78**79***80****81**82****83*****84**85**86**87****88****89****90****91**92**93*****94***95****96*****97*****98*****99*****100**101***102*****103****104*****105*****107*****108*****109*****110****111*****112*****113*****114**115****116*****117*118**119****120****121*****122**123*124*****125****126****127*****128*****129*****130*****131*****132***133****134*****135*****136*****137*****138****139****140*****141*****142*****143*****144*****145**146**147**148*****149**150*****151*****152****153*****154*****155*156****157****158*****159*****160****161*****162*****163*****164*****165*****166****167**168*****169*****170**171****172****173*****174*****175***176***** Without regard to whether a document cited herein was specifically and individually indicated as being incorporated by reference, all documents referred to herein are incorporated by reference into the present application for any and all purposes to the same extent as if each individual reference was fully set forth herein. Having now fully described the subject matter of the claims, it will be understood by those having ordinary skill in the art that the same can be performed within a wide range of equivalents without affecting the scope of the subject matter or particular aspects described herein. It is intended that the appended claims be interpreted to include all such equivalents.
371,526
11858942
DESCRIPTION OF THE INVENTION The present invention relates to triazolotriazine compounds of general formula (1), including hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof, wherein:R is hydrogen or optionally substituted C1-5alkyl; Any of said optionally substituted alkyls are substituted by halogen, cyano, hydroxyl, nitro, amino, alkylamino, cycloalkylamino, methyl, ethyl, methoxyl, ethoxyl, trifluoromethyl, trifluoroethyl, trifluoromethoxyl, or trifluoroethoxyl;Ar1is a 5-6 membered aromatic ring that is optionally substituted with halogen, oxo, cyano, methyl, methoxyl, trifluoromethyl, or trifluoromethoxyl;Ar2is a mono- or bicyclic aromatic ring that is optionally substituted with halogen, hydroxyl, cyano, methyl, methoxyl, trifluoromethyl, or trifluoromethoxyl; andQ is a mono- or bicyclic aromatic ring that is optionally substituted with X, an aminocarbonyl group that is optionally substituted with Y and Z on the nitrogen, an aminosulfonyl group that is optionally substituted with Y and Z on the nitrogen, a nitro group, or a cyano group. X is halogen, cyano, hydroxyl, nitro, amino, alkylamino, methoxyl, ethoxyl, trifluoromethoxyl, trifluoroethoxyl, optionally substituted C1-9alkyl, optionally substituted C1-9cycloalkyl, optionally substituted C1-9alkenyl, optionally substituted C1-9cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heteroaralkyl, optionally substituted heterocycloalkyl, or optionally substituted heterocycloalkenyl; Any of said optionally substituted groups are substituted by halogen, cyano, hydroxyl, nitro, amino, alkylamino, cycloalkylamino, aminocarbonyl, sulfonyl, aminosulfonyl, carbonylamino, sulfonylamino, methyl, ethyl, methoxyl, ethoxyl, trifluoromethyl, trifluoroethyl, trifluoromethoxyl, trifluoroethoxyl, heterocycloalkyl, aryl, heteroaryl, polyoxyethylene, polyoxypropylene, C1-3alkyl polyoxyethylene, or C1-3alkyl polyoxypropylene. Y and Z are each independently hydrogen, optionally substituted C1-9alkyl, optionally substituted mono- or bicyclic C1-9cycloalkyl, optionally substituted C1-9alkenyl, optionally substituted mono- or bicyclic C1-9cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heteroaralkyl, optionally substituted heterocycloalkyl, or optionally substituted heterocycloalkenyl; Any of said optionally substituted groups are substituted by halogen, cyano, hydroxyl, nitro, amino, alkylamino, cycloalkylamino, aminocarbonyl, sulfonyl, aminosulfonyl, carbonylamino, sulfonylamino, methyl, ethyl, methoxyl, ethoxyl, trifluoromethyl, trifluoroethyl, trifluoromethoxyl, trifluoroethoxyl, polyoxyethylene, polyoxypropylene, C1-3alkyl polyoxyethylene, or C1-3alkyl polyoxypropylene; Or Y and Z are joined to form an optionally substituted ring having from 3 to 10 ring atoms; Any of said optionally substituted ring is substituted by halogen, cyano, hydroxyl, oxo, nitro, amino, alkylamino, cycloalkylamino, aminocarbonyl, sulfonyl, aminosulfonyl, carbonylamino, sulfonylamino, methyl, ethyl, methoxyl, ethoxyl, trifluoromethyl, trifluoroethyl, trifluoromethoxyl, trifluoroethoxyl, polyoxyethylene, polyoxypropylene, C1-3alkyl polyoxyethylene, or C1-3alkyl polyoxypropylene. An embodiment of the present invention includes compounds wherein R in formula (1) is hydrogen, methyl, or trifluoromethyl. A preferred embodiment of the present invention includes compounds wherein R in formula (1) is hydrogen or methyl. A most preferred embodiment of the present invention includes compounds wherein R in formula (1) is hydrogen. Another embodiment of the present invention includes compounds wherein Ar1in formula (1) is optionally substituted imidazolyl, triazolyl, tetrazolyl, furanyl, thiophenyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, phenyl, pyridyl, pyridazinyl, pyrimidyl, pyrazinyl, or triazinyl; Any of said optionally substituted aromatic rings are substituted by halogen, oxo, cyano, methyl, methoxyl, trifluoromethyl, or trifluoromethoxyl. A preferred embodiment of the present invention includes compounds wherein Ar1in formula (1) is selected from the aromatic groups shown in Table (1). A most preferred embodiment of the present invention includes compounds wherein Ar1in formula (1) is 2-furanyl. TABLE 1Preferred structures of Ar1in formula (1) Another embodiment of the present invention includes compounds wherein Ar2in formula (1) is optionally substituted imidazolyl, triazolyl, tetrazolyl, furanyl, thiophenyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, phenyl, pyridyl, pyridazinyl, pyrimidyl, pyrazinyl, triazinyl, indolyl, isoindolyl, indazolyl, benzimidazolyl, azaindolyl, azaindazolyl, purinyl, benzofuranyl, isobenzofuranyl, benzothiophenyl, benzisoxazolyl, benzoisothiazolyl, benzoxazolyl, benzothiadiazolyl, quinolyl, isoquinolyl, quinoxalinyl, phthalazinyl, quinazolinyl, cinnolinyl, naphthyridinyl, pyridopyrimidinyl, pyridopyrazinyl, or pteridinyl; Any of said optionally substituted aromatic rings are substituted by halogen, hydroxyl, cyano, nitro, methyl, methoxyl, trifluoromethyl, or trifluoromethoxyl. A preferred embodiment of the present invention includes compounds wherein Ar2in formula (1) is optionally substituted phenyl, pyridyl, pyridazinyl, or pyrimidyl; Any of said optionally substituted aromatic rings are substituted by halogen, hydroxyl, cyano, methyl, methoxyl, trifluoromethyl, or trifluoromethoxyl. A most preferred embodiment of the present invention includes compounds wherein Ar2in formula (1) is phenyl or pyridyl that is optionally substituted with halogen or hydroxyl. Another embodiment of the present invention includes compounds wherein Q in formula (1) is a mono- or bicyclic aromatic ring that is optionally substituted with X. X is halogen, cyano, hydroxyl, nitro, amino, alkylamino, methoxyl, ethoxyl, trifluoromethoxyl, trifluoroethoxyl, optionally substituted C1-9alkyl, optionally substituted C1-9cycloalkyl, optionally substituted C1-9alkenyl, optionally substituted C1-9cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heteroaralkyl, optionally substituted heterocycloalkyl, or optionally substituted heterocycloalkenyl; Any of said optionally substituted groups are substituted by halogen, cyano, hydroxyl, nitro, amino, alkylamino, cycloalkylamino, aminocarbonyl, sulfonyl, aminosulfonyl, carbonylamino, sulfonylamino, methyl, ethyl, methoxyl, ethoxyl, trifluoromethyl, trifluoroethyl, trifluoromethoxyl, trifluoroethoxyl, heterocycloalkyl, aryl, heteroaryl, polyoxyethylene, polyoxypropylene, C1-3alkyl polyoxyethylene, or C1-3alkyl polyoxypropylene. A preferred embodiment of the present invention includes compounds wherein Q in formula (1) is a 5-6 membered aromatic ring that is optionally substituted with X. X is halogen, cyano, hydroxyl, nitro, amino, alkylamino, methoxyl, ethoxyl, trifluoromethoxyl, trifluoroethoxyl, optionally substituted Cis alkyl, optionally substituted C1-5cycloalkyl, optionally substituted C1-5alkenyl, optionally substituted C1-5cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heteroaralkyl, optionally substituted heterocycloalkyl, or optionally substituted heterocycloalkenyl; Any of said optionally substituted groups are substituted by halogen, cyano, hydroxyl, nitro, amino, alkylamino, cycloalkylamino, aminocarbonyl, sulfonyl, aminosulfonyl, carbonylamino, sulfonylamino, methyl, ethyl, methoxyl, ethoxyl, trifluoromethyl, trifluoroethyl, trifluoromethoxyl, trifluoroethoxyl, heterocycloalkyl, aryl, heteroaryl, polyoxyethylene, polyoxypropylene, C1-3alkyl polyoxyethylene, or C1-3alkyl polyoxypropylene. A most preferred embodiment of the present invention includes compounds wherein Q in formula (1) is a tetrazole ring that is optionally substituted with X. X is optionally substituted C1-3alkyl or optionally substituted heterocycloalkyl; Any of said optionally substituted groups are substituted by halogen, cyano, hydroxyl, methyl, ethyl, methoxyl, ethoxyl, trifluoromethyl, trifluoroethyl, trifluoromethoxyl, trifluoroethoxyl, heterocycloalkyl, aryl, or heteroaryl. The term “alkyl” includes both straight- and branched-chain saturated aliphatic hydrocarbon groups having a certain number of carbon atoms. For example, C4alkyl includes n-butyl, isobutyl, sec-butyl and t-butyl. The term “cycloalkyl” includes both mono- and bicyclic saturated aliphatic hydrocarbon groups having a certain number of carbon atoms. The term “alkenyl” includes both straight- and branched-chain aliphatic hydrocarbon groups containing at least one carbon-to-carbon double bond. Preferably, one carbon-to-carbon double bond is present. The term “aryl” includes both monocyclic and bicyclic aromatic rings comprising 5 to 14 ring atoms, preferably 6 to 10 ring atoms, unless it is specified otherwise. The aryl group can be optionally substituted with one or more substituents. The term “aryl” also includes both monocyclic and bicyclic heteroaryl rings comprising 5 to 14 ring atoms, preferably 6 to 10 ring atoms, unless it is specified otherwise. The term “heterocycloalkyl,” also known as “heterocyclyl,” includes saturated monocyclic and bicyclic ring systems comprising 3 to 14 ring atoms, preferably 4 to 10 ring atoms, in which one or more of the atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. Examples of heterocycloalkyl groups include, for example, azetidinyl, hexahydroazepinyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, tetrahydrofuranyl, thiomorpholinyl, and tetrahydrothienyl, and N-oxides thereof. The term “halogen” includes fluoro, chloro, bromo and iodo. The term “trifluoromethyl” refers to the group (—CF3). The term “hydroxyl” or “hydroxy” means an “—OH” group. The compounds of formula (1) of the present invention may exist in one or more geometrical, enantiomeric, diastereoisomeric or tautomeric forms. The compounds of formula (1) of the present invention include all such isomeric forms, including racemic and other mixtures thereof. In another aspect, the compounds of formula (1) of the present invention may exist in either solvated or unsolvated form. The term “solvated” is used herein to describe a compound complex that comprises a compound of the present invention and a number of pharmaceutically acceptable solvent molecules, such as water and ethanol molecules. The compounds of formula (1) of the present invention include all solvated or unsolvated forms thereof. In another aspect, the compounds of formula (1) of the present invention may exist in a form of pharmaceutically acceptable salts. The term “pharmaceutically acceptable salt” refers to a physiologically or toxicologically tolerable salt, and when appropriate, including pharmaceutically acceptable base addition salts and acid addition salts thereof. The compounds of formula (1) of the present invention include all pharmaceutically acceptable salts thereof. In another aspect, the compounds of formula (1) of the present invention may exist in a form of pharmaceutically acceptable nanoparticles. Nanoparticles containing a compound of formula (1) of the present invention can be designed to improve the pharmacokinetics and biodistribution of the drug. For example, a compound of formula (1) may be encased in liposomes, which may extend the life of the drug that is being distributed. Nanoparticles of suitable size may also have a better safety profile because the nanoparticles will preferentially leak out of the porous blood vessels around the tumor cells. This may further provide the benefit of lower doses of the drug. In another aspect, the compounds of formula (1) of the present invention may exist in the form of prodrugs. The term “prodrug” refers to a compound that is converted to a compound of the present invention by a metabolic process in vivo (for example, by hydrolysis, reduction or oxidation). The compounds of formula (1) of the present invention include all such prodrugs thereof. In another aspect, the compounds of formula (1) of the present invention also include pharmaceutically acceptable isotopic variations in which one or more atoms is replaced by atoms having the same atomic number but different atomic mass. The atoms suitable for such isotope replacement include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine. Certain isotopic variations of the compounds of formula (1), such as deuterium replaced compounds, may afford certain therapeutic advantages resulting from greater metabolic stability, and hence may be preferred in some circumstances. The isotopic variations of the compounds of formula (1) can be prepared by conventional techniques known to those skilled in the art. The Preparation of the Triazolotriazine Derivatives Another aspect of the present invention is the preparation of the triazolotriazine compounds as A2A receptor antagonists. The triazolotriazine compounds of the invention can be prepared by various synthetic methods. As an illustrative example, two general synthetic routes to the target compound are shown in Scheme (1). In the first approach, after the intermediate (1A) is prepared in a suitable manner, the methylsulfonyl group of intermediate (1A) is replaced by an alkyl amino group to give the triazolotriazine compound (1C). In the second approach, after the intermediate (1B) is prepared in a suitable manner, the phenoxy group of intermediate (1B) is replaced by an alkyl amino group to give the triazolotriazine compound (1C). The needed intermediates (1A) and (1B) in Scheme (1) can also be prepared by various synthetic methods. As an illustrative example, a general synthetic route to compound (1A) is shown in Scheme (2). In the first step, a suitable aryl hydrazide (2A) reacts with S-methylisothiourea (2B) in aqueous sodium hydroxide to give intermediate (2C). Vigorous heating of (2C) in aqueous medium in the following step provides intermediate (2D), which reacts with N-cyanodithioimino-carbonate (2E) to afford the sulfide intermediate (2F). Subsequent oxidation of intermediate (2F) with m-chloroperoxybenzoic acid provides the needed sulfone (1A). As is shown in Scheme (1), nucleophilic displacement of the methylsulfonyl group with a suitable alkyl amine gives the target compound (1C) (J. Chem. Soc., Perkin Trans. 1 1995, 801-808; Structural Chemistry. 2013, 24, 1241-1251). The intermediate (2D) in Scheme (2) can also be prepared by various synthetic methods. As illustrative examples, three alternative synthetic routes to intermediate (2D) are shown in Scheme (3). It is worth emphasizing that among the three approaches, the one starting from a methyl ester or ethyl ester (3F) usually improves efficiency and gives higher yield. The intermediate (1B) in Scheme (1) can be prepared as shown in Scheme (4). Starting from cyanuric chloride (4A), reflux in phenol provides 2,4,6-triphenoxy-1,3,5-triazine (4B). The following reaction with hydrazine hydrate gives 2-hydrazino-4,6-diphenoxy-1,3,5-triazine (4C), which upon reacting with suitable acid chloride gives the acyl hydrazides (4D). Cyclization of the hydrazide (4D) under dehydrative conditions provides the 2-substituted 5,7-diphenoxytriazolotriazine (4E), which can be converted into the key intermediate (1B) in refluxing methanolic ammonia (J. Chem. Soc., Perkin Trans. 1 1995, 801-808). The target compound (1C) is obtained by reacting compound (1B) with a suitable alkyl amine, as shown in Scheme (1). Cancer Treatment The triazolotriazine derivatives of the present invention are adenosine A2AR antagonists and can be used for the treatment or prevention of disorders related to hyperactive adenosine A2A receptors. For example, Parkinson's disease, cognitive or memory impairment disorder, and Alzheimer's disease are some of the disorders that can be treated with the triazolotriazine derivatives of the present invention. In particular, the A2AR antagonists of the present invention can be used for the treatment or prevention of cancer and related abnormal cell proliferations in a host, which is any multi-cellular vertebrate organism including both human and non-human mammals. The host is in particular human. The importance of lymphoid cells in tumor immunity is well appreciated in recent years. The immune response to tumors includes immunologic surveillance, by which cellular mechanisms associated with cell-mediated immunity destroy newly transformed tumor cells after recognizing tumor-associated antigens. The cytotoxic immune cells, which are mainly T-cells, have been found within neuroblastoma, malignant melanomas, sarcomas, and carcinomas of the colon, breast, cervix, endometrium, ovary, testis, nasopharynx, and kidney. Antibody-mediated protection against tumor growth is also known, although it generally plays a less significant role than cell-mediated immunity against cancer. The A2AR antagonists of the present invention can be used to increase the anti-tumor activity of immune cells in a host. The A2AR antagonists can reduce T cell anergy or the tolerance of T cells to cancer, can increase susceptibility of cancer cells to immune rejection, can inhibit the expansion of regulatory T cells, and can enhance the generation of memory T cells. The A2AR antagonists can improve both the natural immune response and various adaptive immunotherapy in a host. In a typical embodiment of the present invention, a method of treating or preventing abnormal cell proliferation comprises administering to a patient an effective dose of an A2AR antagonist of formula (1), or a pharmaceutically acceptable salt or solvate thereof. To be more effective, a synergistic effect may be achieved by combining the A2AR antagonists of the present invention with other modalities of cancer therapy, such as chemotherapy, tumor vaccines, and various immune checkpoint inhibitors. The term “combination therapy” refers to both concurrent and sequential administration of the active agents. As an example of the combination therapy, a method of treating or preventing abnormal cell proliferation in a host comprises administering to a patient an A2AR antagonist of formula (1) in combination or alternation with an immune checkpoint inhibitor. The immune checkpoint inhibitor can be a PD-1 inhibitor, a PD-L1 inhibitor, a PD-L2 inhibitor, a CTLA-4 inhibitor, a BTLA inhibitor, a LAG3 inhibitor, a TIM-3 inhibitor, a B7-H3 inhibitor, a B7-H4 inhibitor, a KIR inhibitor, a TIGIT inhibitor, or a VISTA inhibitor. In another example, a method of treating or preventing abnormal cell proliferation in a host comprises administering to a patient an A2AR antagonist of formula (1) in combination or alternation with a cell-based vaccine. The cell-based vaccine is based on cells that match the tumor to be prevented. For example, if a host is suffering from, or at risk of suffering from, a prostate cancer, the cell-based vaccine will be based on a prostate cancer tumor cell. In these instances, the cell is typically irradiated or otherwise prevented from replicating. Or, the cell is genetically modified to secrete a colony stimulating factor. In another example, a method of treating or preventing abnormal cell proliferation in a host comprises administering to a patient an A2AR antagonist of formula (1) in combination or alternation with Chimeric Antigen Receptor (CAR) T-Cell Therapy. In another example, a method of treating or preventing abnormal cell proliferation in a host comprises administering to a patient an A2AR antagonist of formula (1) in combination or alternation with an anti-cancer agent to treat abnormal cell proliferation. The anti-cancer agent can be an alkylating agent, an antimetabolite, an anthracycline derivative, a plant alkaloid, a topoisomerase inhibitor, an antitumor antibiotic, a kinase inhibitor, or a monoclonal antibody against tumor antigens. In other examples, a synergistic effect may be achieved by combining the A2AR antagonists of the present invention with two or more other modalities of cancer therapy, such as chemotherapy, tumor vaccines, and immune checkpoint inhibitors. The term “combination therapy” refers to both concurrent and sequential administration of the active agents. Pharmaceutical Compositions The triazolotriazine derivatives of the present invention can be formulated as pharmaceutical compositions when administered to a host. The pharmaceutical compositions are determined by the chosen route of administration, such as orally, parenterally, intravenously, intramuscularly, nasally, buccally, topically, transdermally or subcutaneously. The triazolotriazine derivatives included in the pharmaceutical compositions should be sufficient to deliver a therapeutically effective amount to treat cancer or other disorders characterized by abnormal cell proliferation without causing serious toxic effects to the host. The treatment can involve daily or multi-daily administration of the triazolotriazine derivatives over a period of a few days to weeks, months, or even years. A convenient mode of administration of the triazolotriazine derivatives of the present invention is oral. Oral compositions generally include an inert diluent or an edible carrier. The compositions may be enclosed in gelatin capsules or compressed into tablets. The tablets, pills, capsules, troches and the like can contain any of the following ingredients: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose; a disintegrating agent such as alginic acid or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate, or orange flavoring; a wetting or emulsifying agent; preservatives; and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate. When the pharmaceutical compositions are in a capsule, a liquid carrier such as fatty oil may also be included. In addition, the pharmaceutical compositions can contain various other materials, such as coatings of sugar, shellac, or other enteric agents. The triazolotriazine derivatives of the present invention can also be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like. A syrup may contain, in addition to the triazolotriazine derivatives, sucrose as a sweetening agent, preservatives, coloring agents and flavoring agents. Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include a sterile diluent such as water, saline solution, Ringer's solution, fixed oils, polyethylene glycols, glycerin, propylene glycol, fatty acids such as oleic acid and its glyceride derivatives, or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfate; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. The triazolotriazine derivatives of the present invention can also be placed in carriers that protect the derivatives against rapid elimination from the body. Various means to achieve controlled release, including implants and microencapsulated delivery systems, can also be used. The triazolotriazine derivatives of the present invention can also be administered through the use of nebulizer, a dry powder inhaler or a metered dose inhaler inhaled through the nasal aerosols. The compositions may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters, fluorocarbons, and other conventional solubilizing or dispersing agents. It is understandable that for any particular patient the specific dose and treatment regimen will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, and sex of the patient, diet, time of administration, rate of excretion, the pathological condition to be treated, the goal of treatment, as well as the judgment of the physician. The amount of active ingredient may also depend on what is the co-administered therapeutic agent if it is a combination therapy. EXAMPLES The following examples, which are for detailed illustration only, are not intended to limit the scope of the present invention. Example 1 2-(Furan-2-carboxamido) guanidine A mixture of furan-2-carbohydrazide (37.80 g, 300 mmol), S-methylisothiourea sulfate (41.70 g, 150 mmol) and an aqueous sodium hydroxide solution (2%, 1.2 L) was stirred at room temperature for 72 h. The precipitate was filtered, washed with ice-cold water, and used in the next step without further purification (25.25 g, 51.00% yield).1H NMR (500 MHz, DMSO-d6) δ: 10.95 (s, 1H), 7.56 (s, 1H), 6.91 (d, J=91.9 Hz, 4H), 6.64 (s, 1H), 6.45 (s, 1H). Example 2 3-(Furan-2-yl)-1H-1,2,4-triazol-5-amine A stirred solution of 2-(furan-2-carboxamido) guanidine (23.20 g, 138 mmol) in DMF (464 mL) was heated at 125° C. overnight. After it was cooled to room temperature, the solvent was removed under reduced pressure. To the residue was added DCM (200 ml) and it was stirred for 30 min. The precipitate was filtered and washed with DCM (20 mL×2) to afford the title compound as yellow solid (17.37 g, 84.00% yield).1H NMR (500 MHz, DMSO-d6) δ: 12.13 (s, 1H), 7.69 (s, 1H), 6.69 (d, J=1.8 Hz, 1H), 6.54 (dd, J=3.0, 1.7 Hz, 1H), 6.03 (s, 2H). Example 2A 3-(Furan-2-yl)-1H-1,2,4-triazol-5-amine To a stirred solution of CH3ONa (171.4 g, 3172 mol) and aminoguanidine hydrochloride (175.3 g, 1586 mmol) in methanol (1200 mL) at 0° C. was added slowly the solution of methyl furan-2-carboxylate (100 g, 793 mmol) in methanol (300 mL). The reaction mixture was then stirred at 75° C. overnight. The resulting mixture was filtered. The filtrate was concentrated in vacuo and the residue was dissolved in water (50 mL). 3N HCl was added to adjust pH to 4. The precipitated solid was collected by filtration and drying to afford the title compound as yellow solid (69.2 g, 58.1% yield).1H NMR (500 MHz, DMSO-d6) δ: 12.44 (s, 1H), 7.69 (d, 1H), 6.70 (d, 1H), 6.54 (dd, 1H), 6.03 (s, 2H). Example 3 2-(Furan-2-yl)-5-(methylthio)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine A mixture of 3-(furan-2-yl)-1H-1,2,4-triazol-5-amine (13.58 g, 90.46 mmol) and dimethyl cyanocarbonimidodithioate (13.23 g, 90.46 mmol) was stirred at 180° C. for 1.5 h. It was next cooled to room temperature. The residue was purified by column chromatography (PE:EA=1:1) afforded the title compound as white solid (7.00 g, 31.20% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.96 (s, 1H), 8.76 (s, 1H), 7.93 (d, J=0.9 Hz, 1H), 7.16 (d, J=3.3 Hz, 1H), 6.72 (dd, J=3.4, 1.7 Hz, 1H), 2.51 (s, 3H). Example 4 2-(Furan-2-yl)-5-(methylsulfonyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine A solution of m-CPBA (85% strength, 26.20 g, 128.90 mmol) in DCM (240 mL) was added dropwise to a stirred, ice-cold suspension of 2-(furan-2-yl)-5-(methylthio)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine (8.0 g, 32.2 mmol) in DCM (480 mL). The reaction was stirred at room temperature for 22 h before the solvent was removed under vacuum. The crude material was suspended in ethanol (120 mL) and stirred at room temperature for 30 min. The solid was filtered, washed with ethanol and dried to give the title compound as brown solid (7.82 g, 86.90% yield).1H NMR (500 MHz, DMSO-d6) δ: 9.81 (s, 1H), 9.48 (s, 1H), 7.99 (d, J=1.0 Hz, 1H), 7.27 (d, J=3.4 Hz, 1H), 6.76 (dd, J=3.4, 1.8 Hz, 1H), 3.36 (s, 3H). Example 5 2-(Furan-2-yl)-N5-(2-(pyridin-4-yl)ethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine A solution of 2-(pyridin-4-yl)ethanamine (100 mg, 0.86 mmol), 2-(furan-2-yl)-5-(methylsulfonyl)-[1,2,4]triazolo[1,5-α][1,3,5]triazin-7-amine (230 mg, 0.82 mmol) and TEA (250 mg, 2.46 mmol) in MeCN (5 mL) was stirred overnight. The reaction mixture was quenched with water (30 mL) and extracted with DCM (15 mL×3). The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography to afford the title compound as white solid (41.50 mg, 15.72% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.47 (d, J=4.5 Hz, 2H), 8.22 (s, 2H), 7.89 (d, J=14.3 Hz, 1H), 7.57 (d, J=46.0 Hz, 1H), 7.28 (s, 2H), 7.06 (s, 1H), 6.68 (s, 1H), 3.54 (d, J=5.6 Hz, 2H), 2.90 (s, 2H). Example 6 Methyl 4-(2-aminoethyl)benzoate A stirred mixture of methyl 4-(cyanomethyl)benzoate (2 g, 11.42 mmol), NH3/MeOH (7N, 4 mL) and Raney-Nickel (0.1 g) in MeOH (20 mL) was heated under H2at 55° C. overnight. The reaction mixture was filtered through Celite, the filtrate was concentrated, and purification by column chromatography (EtOAc/MeOH) afforded the title compound as brown oil (1.35 g, 66.0% yield).1H NMR (500 MHz, DMSO-d6) δ: 7.97-7.76 (m, 2H), 7.35 (d, J=8.2 Hz, 2H), 3.84 (s, 3H), 2.84-2.74 (m, 2H), 2.71 (t, J=6.9 Hz, 2H). Example 7 Methyl 4-(2-(tert-butoxycarbonyl)aminoethyl)benzoate To a stirred solution of methyl 4-(2-aminoethyl)benzoate (4.5 g, 25 mmol) in THF (15 mL) was added TEA (7.6 g, 75 mmol) and Boc2O (6.0 g, 27 mmol). The reaction mixture was stirred at room temperature overnight. TLC showed the reaction completed. The reaction mixture was concentrated in vacuo. The crude product was purified by column chromatography to afford the title compound as yellow solid (5.6 g, 80% yield).1H NMR (500 MHz, CDCl3) δ: 7.97 (d, 2H), 7.26 (d, 2H), 4.54 (dr, 1H), 3.90 (s, 3H), 3.38-3.40 (m, 2H), 2.86 (t, 2H), 1.43 (s, 9H). Example 8 4-(2-((tert-Butoxycarbonyl)amino)ethyl)benzoic acid To a stirred solution of methyl 4-(2-(tert-butoxycarbonyl)aminoethyl)benzoate (5.6 g, 0.20 mmol) in THF (50 mL) and H2O (50 mL) was added sodium hydroxide (8.0 g, 2.0 mmol). The reaction mixture was stirred at 50° C. overnight. TLC showed the reaction completed. The reaction mixture was neutralized with 2N HCl and extracted with EtOAc. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo to afford the title compound as white solid (5.1 g, 96% yield).1H NMR (500 MHz, DMSO-d6) δ: 7.84 (d, 2H), 7.26 (d, 2H), 6.88 (t, 1H), 3.14-3.17 (m, 2H), 2.74 (t, 2H), 1.36 (s, 9H). Example 9 tert-Butyl 4-((2-(pyrrolidin-1-yl)ethyl)carbamoyl)phenethylcarbamate A mixture of 4-(2-((tert-Butoxycarbonyl)amino)ethyl)benzoic acid (0.265 g, 1 mmol), HATU (0.494 g, 1.3 mmol) and TEA (0.202 g, 2 mmol) in dichloromethane (20 mL) was stirred at room temperature for 30 min. Then 2-(pyrrolidin-1-yl)ethanamine (0.126 g, 1.1 mmol) was added and the reaction was stirred at room temperature for 2 h. After more dichloromethane (80 mL) was added, the reaction mixture washed with water (2×20 mL). After drying with Na2SO4, removal of the solvent provided the crude product. Purification by column chromatography afforded the title compound as white solid (0.337 g, 93.2% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.47 (s, 1H), 7.78 (d, J=8.2 Hz, 2H), 7.29 (d, J=8.1 Hz, 2H), 6.88 (t, J=5.3 Hz, 1H), 3.51-3.42 (m, 2H), 3.18-3.13 (m, 2H), 2.94 (br, 5H), 2.74 (t, J=7.4 Hz, 2H), 2.69 (s, 1H), 1.81 (s, 4H), 1.36 (s, 9H). Example 10 4-(2-Aminoethyl)-N-(2-(pyrrolidin-1-yl)ethyl)benzamide Hydrochloric acid (20%, 2 mL) was added to a stirred solution of tert-butyl 4-((2-(pyrrolidin-1-yl)ethyl)carbamoyl)phenethylcarbamate (0.3 g, 0.83 mmol) in methanol (5 mL). After it was stirred at 30° C. for 17 h, the excess solvent was removed under reduced pressure to afford the product, which was used for the next step directly. Example 11 4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)-N-(2-(pyrrolidin-1-yl)ethyl)benzamide The reaction was carried out as in Example 5 to afford the title compound as light-yellow solid (13.1% yield). LC-MS m/z [M+H]+: 462;1H NMR (500 MHz, DMSO-d6) δ: 8.37 (s, 1H), 8.15 (s, 2H), 7.87 (s, 1H), 7.78 (d, J=7.7 Hz, 2H), 7.53 (d, J=40.6 Hz, 1H), 7.33 (d, J=7.4 Hz, 2H), 7.06 (d, J=2.9 Hz, 1H), 6.67 (s, 1H), 3.51 (d, J=5.1 Hz, 2H), 3.38 (d, J=6.0 Hz, 2H), 2.91 (d, J=6.7 Hz, 2H), 2.61 (s, 2H), 2.54 (s, 4H), 1.69 (s, 4H). Example 12 4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)-N-(2-morpholinoethyl)benzamide The title compound was synthesized in a similar way as the title compound in Example 11. LC-MS m/z [M+H]+: 478;1H NMR (500 MHz, DMSO-d6) δ: 8.33 (t, J=5.5 Hz, 1H), 8.18 (s, 2H), 7.87 (s, 1H), 7.77 (d, J=7.9 Hz, 2H), 7.54 (d, J=42.0 Hz, 1H), 7.34 (d, J=7.8 Hz, 2H), 7.06 (d, J=3.2 Hz, 1H), 6.67 (s, 1H), 3.59-3.48 (m, 6H), 3.37 (dd, J=13.0, 6.5 Hz, 2H), 2.91 (t, J=7.1 Hz, 2H), 2.44 (dd, J=18.0, 11.1 Hz, 6H). Example 13 4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)-N-(2-(4-methylpiperazin-1-yl)ethyl)benzamide The title compound was synthesized in a similar way as the title compound in Example 11. LC-MS m/z [M+H]+: 491;1H NMR (500 MHz, DMSO-d6) δ: 8.42 (s, 1H), 8.18 (s, 2H), 7.87 (s, 1H), 7.79 (d, J=8.1 Hz, 2H), 7.60-7.47 (m, 1H), 7.34 (d, J=7.9 Hz, 2H), 7.06 (d, J=3.3 Hz, 1H), 6.68 (d, J=1.6 Hz, 1H), 4.11 (s, 1H), 3.52 (d, J=5.6 Hz, 2H), 3.39 (s, 2H), 3.17 (s, 3H), 2.92 (dd, J=8.3, 6.3 Hz, 5H), 2.57 (s, 4H). Example 14 (4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yDamino)ethyl)phenyl)-(4-(2,2,2-trifluoroethy)piperazin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11. LC-MS m/z [M+H]+: 516;1H NMR (500 MHz, DMSO-d6) δ: 8.15 (br, 2H), 7.96-7.85 (m, 1H), 7.64-7.47 (m, 1H), 7.30 (d, J=8.9 Hz, 4H), 7.06 (d, J=3.1 Hz, 1H), 6.67 (s, 1H), 3.52 (d, J=6.1 Hz, 4H), 3.44-3.31 (m, 2H), 3.20 (q, J=10.1 Hz, 2H), 2.90 (t, J=7.0 Hz, 2H), 2.61 (br, 4H). Example 15 4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)-N-(3-(dimethylamino)propyl)benzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.44 (t, J=5.3 Hz, 1H), 8.15 (br, 2H), 7.87 (s, 1H), 7.77 (d, J=8.0 Hz, 2H), 7.51 (dd, J=25.9, 20.8 Hz, 1H), 7.34 (d, J=7.7 Hz, 2H), 7.06 (d, J=3.2 Hz, 1H), 6.67 (s, 1H), 3.52 (d, J=6.0 Hz, 2H), 3.27 (dd, J=12.8, 6.7 Hz, 2H), 2.92 (t, J=7.1 Hz, 2H), 2.29 (t, J=7.0 Hz, 2H), 2.16 (s, 6H), 1.74-1.59 (m, 2H). Example 16 (4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)phenyl)-(4,4-difluoropiperidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.31 (br, 2H), 7.87 (s, 1H), 7.56 (dt, J=41.0, 5.5 Hz, 1H), 7.40-7.29 (m, 4H), 7.05 (d, J=3.3 Hz, 1H), 6.68 (dd, J=3.1, 1.6 Hz, 1H), 3.82-3.38 (m, 6H), 2.90 (t, J=7.3 Hz, 2H), 2.03 (m, 4H). Example 17 (4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)phenyl)-(4-fluoropiperidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11. LC-MS m/z [M+H]+: 451;1H NMR (500 MHz, DMSO-d6) δ: 8.32 (br, J=115.2 Hz, 2H), 7.87 (s, 1H), 7.66-7.47 (m, 1H), 7.40-7.26 (m, 4H), 7.06 (d, J=3.3 Hz, 1H), 6.77-6.59 (m, 1H), 4.89 (dd, J=48.3, 3.0 Hz, 1H), 3.79-3.34 (m, 6H), 2.89 (dd, J=15.3, 8.0 Hz, 2H), 1.95-1.58 (m, 4H). Example 18 (4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)phenyl)-(morpholino)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.32 (br, 2H), 7.92-7.85 (m, 1H), 7.54 (dd, J=26.6, 20.9 Hz, 1H), 7.33 (s, 4H), 7.06 (d, J=3.1 Hz, 1H), 6.68 (s, 1H), 3.74-3.38 (m, 10H), 2.90 (t, J=7.2 Hz, 2H). Example 19 (4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)phenyl)-(pyrrolidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.31 (br, 2H), 7.87 (s, 1H), 7.52 (dd, J=26.3, 20.9 Hz, 1H), 7.44 (d, J=7.9 Hz, 2H), 7.30 (d, J=7.7 Hz, 2H), 7.06 (d, J=3.2 Hz, 1H), 6.68 (s, 1H), 3.52 (d, J=6.3 Hz, 2H), 3.44 (t, J=6.7 Hz, 2H), 3.35 (d, J=6.6 Hz, 2H), 2.90 (t, J=7.0 Hz, 2H), 1.89-1.72 (m, 4H). Example 20 (4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)phenyl)-(4-methylpiperazin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.26 (d, J=125.8 Hz, 2H), 7.86 (s, 1H), 7.52 (s, 1H), 7.30 (br, 4H), 7.05 (s, 1H), 6.67 (s, 1H), 3.53 (br, 4H), 3.31 (br, 2H), 2.89 (br, 2H), 2.27 (br, 4H), 2.16 (s, 3H). Example 21 (S)-(4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)-phenyl)(3-fluoropyrrolidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.28 (br, 2H), 7.86 (s, 1H), 7.63-7.43 (m, 3H), 7.32 (d, J=7.7 Hz, 2H), 7.06 (d, J=3.3 Hz, 1H), 6.67 (s, 1H), 5.31 (t, J=52.7 Hz, 1H), 3.82-3.63 (m, 2H), 3.58-3.41 (m, 4H), 2.91 (t, J=7.0 Hz, 2H), 2.19-1.99 (m, 2H). Example 22 4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)-N-(2-(azetidin-1-yl)ethyl)benzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.30-8.13 (m, 3H), 7.87 (s, 1H), 7.77 (d, J=8.0 Hz, 2H), 7.50 (dd, J=26.4, 21.0 Hz, 1H), 7.33 (d, J=7.7 Hz, 2H), 7.06 (d, J=3.3 Hz, 1H), 6.67 (s, 1H), 3.52 (d, J=6.1 Hz, 2H), 3.19 (dd, J=12.5, 6.4 Hz, 2H), 3.13 (t, J=6.9 Hz, 4H), 2.92 (t, J=7.2 Hz, 2H), 2.48 (d, J=6.7 Hz, 2H), 1.95 (p, J=6.9 Hz, 2H). Example 23 4-(2-((7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)-N-(tert-butyl)benzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.34 (br, 2H), 7.87 (s, 1H), 7.73 (d, J=8.1 Hz, 2H), 7.67 (s, 1H), 7.54 (dt, J=40.3, 5.6 Hz, 1H), 7.31 (t, J=7.8 Hz, 2H), 7.06 (d, J=3.3 Hz, 1H), 6.68 (dd, J=3.3, 1.7 Hz, 1H), 3.50 (dd, J=13.5, 6.5 Hz, 2H), 2.90 (t, J=7.3 Hz, 2H), 1.37 (s, 9H). Example 24 N5-(2-(1H-Benzo[d]imidazol-5-yl)ethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound (0.0231 g, 12.8% yield) as white solid. LC-MS m/z [M+H]+: 362;1H NMR (500 MHz, DMSO-d6) δ: 8.86 (d, J=15.7 Hz, 1H), 8.32 (br, 2H), 7.87 (s, 1H), 7.65 (t, J=7.6 Hz, 1H), 7.58 (s, 1H), 7.51 (t, J=5.4 Hz, 1H), 7.31 (d, J=8.2 Hz, 1H), 7.06 (d, J=3.2 Hz, 1H), 6.68 (s, 1H), 3.59-3.50 (m, 2H), 3.01 (t, J=7.1 Hz, 2H). Example 25 tert-Butyl (2-(isoquinolin-6-yl)ethyl)carbamate A mixture of 6-bromoisoquinoline (0.5 g, 2.4 mmol), potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (0.723 g, 2.88 mmol), cesium carbonate (2.346 g, 7.2 mmol) and Pd(dppf)Cl2(0.088 g, 0.12 mmol) in toluene (12 mL) and water (4 mL) was stirred under N2at 80° C. overnight. The reaction mixture was filtered through Celite and water (30 mL) was added to the filtrate. The filtrate was extracted with ethyl acetate (30 mL×2). The organic layer was washed with water (20 mL) and brine (20 mL). After drying with Na2SO4and removal of solvent, the residue purified by column chromatography to afford the title compound as light-yellow solid (0.43 g, 65.8% yield).1H NMR (500 MHz, DMSO-d6) δ: 9.24 (s, 1H), 8.45 (d, J=5.7 Hz, 1H), 7.90 (d, J=8.1 Hz, 2H), 7.78 (d, J=5.7 Hz, 1H), 7.65 (dd, J=8.5, 1.4 Hz, 1H), 6.95 (t, J=5.4 Hz, 1H), 3.27 (dd, J=13.5, 6.6 Hz, 2H), 2.91 (t, J=7.2 Hz, 2H), 1.34 (s, 9H). Example 26 2-(Isoquinolin-6-yl)ethanamine HCl (4 mL, 4N solution in 1,4-dioxane) was added to a stirred solution of tert-butyl (2-(isoquinolin-6-yl)ethyl)carbamate (0.25 g, 0.918 mmol) in MeOH (2 mL) at room temperature. After stirring for 1.5 h, the excess solvent was removed under reduced pressure to afford the crude product, which was used for the next step directly. Example 27 2-(Furan-2-yl)-N5-(2-(isoquinolin-6-yl)ethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diaminee The reaction was carried out as in Example 5 to afford the title compound as white solid (0.0212 g, 8.8% yield).1H NMR (500 MHz, DMSO-d6) δ: 9.27 (s, 1H), 8.37 (br, 3H), 7.92 (dd, J=27.4, 19.5 Hz, 3H), 7.76 (dd, J=35.7, 6.4 Hz, 2H), 7.61 (d, J=44.7 Hz, 1H), 7.06 (s, 1H), 6.68 (s, 1H), 3.63 (s, 2H), 3.10 (s, 2H). Example 28 2-(Furan-2-yl)-N5-(2-(quinolin-6-yl)ethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The title compound was synthesized in a similar way as the title compound in Example 27.1H NMR (500 MHz, DMSO-d6) δ: 8.85 (dd, J=4.1, 1.6 Hz, 1H), 8.51-8.06 (m, 3H), 7.97 (d, J=8.6 Hz, 1H), 7.85 (d, J=26.5 Hz, 2H), 7.70 (t, J=9.8 Hz, 1H), 7.67-7.54 (m, 1H), 7.50 (dd, J=8.3, 4.2 Hz, 1H), 7.07 (d, J=3.3 Hz, 1H), 6.68 (d, J=1.6 Hz, 1H), 3.62 (dd, J=13.2, 6.6 Hz, 2H), 3.08 (t, J=7.3 Hz, 2H). Example 29 tert-Butyl 4-(1-methyl-1H-1,2,4-triazol-3-yl)phenethylcarbamate A solution of 3-(4-bromophenyl)-1-methyl-1H-1,2,4-triazole (0.5 g, 2.1 mmol), potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (0.58 g, 2.3 mmol), cesium carbonate (2.05 g, 6.3 mmol) and Pd(dppf)Cl2(0.077 g, 0.105 mmol) in toluene (12 mL) and water (4 mL) was stirred under N2at 80° C. overnight. The reaction mixture was filtered through Celite. The filtrate was extracted with ethyl acetate (30 mL×2). The organic layer was washed with water (20 mL) and brine (20 mL). After drying with Na2SO4and removal of solvent, the residue purified by column chromatography to afford the title compound as light-yellow solid (0.416 g, 65.5% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.49 (s, 1H), 7.90 (d, J=8.1 Hz, 2H), 7.27 (d, J=8.2 Hz, 2H), 6.90 (t, J=5.4 Hz, 1H), 3.91 (s, 3H), 3.17 (dt, J=10.4, 5.5 Hz, 2H), 2.73 (t, J=7.4 Hz, 2H), 1.37 (s, 9H). Example 30 2-(4-(1-Methyl-1H-1,2,4-triazol-3-yl)phenyl)ethanamine HCl (4 mL, 4N solution in 1,4-dioxane) was added to a stirred solution of tert-butyl 4-(1-methyl-JH-1,2,4-triazol-3-yl)phenethylcarbamate (0.31 g, 1.02 mmol) in MeOH (2 mL) at room temperature. After stirring for 1 h, the excess solvent was removed under reduced pressure to afford the crude product, which was used for the next step directly. Example 31 2-(Furan-2-yl)-N5-(4-(1-methyl-1H-1,2,4-triazol-3-yl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (0.1076 g, 37.4% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.49 (s, 1H), 8.19 (s, 2H), 7.98-7.84 (m, 3H), 7.54 (dd, J=25.9, 20.4 Hz, 1H), 7.35 (t, J=7.4 Hz, 2H), 7.13-7.01 (m, 1H), 6.68 (d, J=1.6 Hz, 1H), 3.91 (s, 3H), 3.53 (dd, J=13.4, 6.7 Hz, 2H), 2.91 (t, J=7.3 Hz, 2H). Example 32 Methyl 4-(2-(7-amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-benzoate The reaction was carried out as in Example 5 to afford the title compound as yellow solid (41.1 mg, 10% yield).1H NMR (500 MHz, DMSO-d6): 8.09-8.21 (m, 2H), 7.89 (d, 2H), 7.86 (s, 1H), 7.49-7.59 (m, 1H), 7.40-7.43 (m, 2H), 7.05 (d, 1H), 6.67 (dr, 1H), 3.83 (s, 3H), 3.50-3.54 (m, 2H), 2.93-2.96 (m, 2H). Example 33 4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)benzoic acid A mixture of methyl 4-(2-(7-amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)benzoate (200 mg, 0.52 mmol) and NaOH (210 mg, 5.2 mmol) in THF (10 mL) and H2O (2 mL) was stirred at 40° C. overnight. TLC showed the reaction completed. The reaction mixture was neutralized by adding 10% HCl. It was next extracted with EtOAc. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Methanol=50:1) to afford the title compound as white solid (32 mg, 16.7% yield).1H NMR (500 MHz, DMSO-d6): 12.75 (dr, 1H), 8.09-8.21 (m, 2H), 7.89 (dr, 3H), 7.86 (s, 1H), 7.49-7.59 (m, 1H), 7.38 (dr, 2H), 7.05 (d, 1H), 6.67 (s, 1H), 3.52 (dr, 2H), 2.93 (dr, 2H). Example 34 4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-N,N-dimethylbenzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H LCMS [M+1]+: 393.23;1H NMR (500 MHz, DMSO-d6) δ: 8.03-8.24 (m, 2H), 7.86 (s, 1H), 7.49-7.58 (m, 1H), 7.32 (s, 4H), 7.05 (s, 1H), 6.67 (dr, 1H), 3.52 (dr, 2H), 2.89-2.95 (m, 8H). Example 35 4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-N-methylbenzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6) δ: 8.35 (d, 1H), 8.03-8.24 (m, 2H), 7.86 (s, 1H), 7.76 (d, 2H), 7.49-7.58 (m, 1H), 7.32-7.33 (m, 2H), 7.05 (d, 1H), 6.67 (dr, 1H), 3.51 (d, 2H), 2.89-2.91 (m, 2H), 2.77 (d, 3H). Example 36 4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)benzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6): 8.03-8.14 (m, 2H), 7.83 (d, 4H), 7.45-7.55 (m, 1H), 7.33 (dr, 1H), 7.23 (dr, 1H), 7.05 (dr, 1H), 6.67 (dr, 1H), 3.52 (dr, 2H), 2.91 (dr, 2H). Example 37 (4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-phenyl)(piperidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6): 8.09-8.21 (m, 2H), 7.87 (s, 1H), 7.51-7.61 (m, 1H), 7.27-7.31 (m, 4H), 7.06 (d, 1H), 6.67-6.68 (m, 1H), 3.49-3.53 (m, 4H), 3.24 (dr, 2H), 2.86-2.90 (m, 2H), 1.40-1.59 (m, 6H). Example 38 (4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-phenyl)(3,3-difluoropyrrolidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6): 8.09-8.41 (m, 2H), 7.87 (s, 1H), 7.51-7.61 (m, 1H), 7.48 (d, 2H), 7.34 (t, 1H), 7.05 (d, 1H), 6.67-6.68 (m, 1H), 3.88 (m, 2H), 3.63-3.70 (m, 2H), 3.49-3.53 (m, 2H), 2.88-2.92 (m, 2H), 2.39-2.43 (m, 2H). Example 39 (4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-phenyl)(3-fluoropiperidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6): 8.09-8.21 (m, 2H), 7.87 (s, 1H), 7.50-7.61 (m, 1H), 7.28-7.34 (m, 4H), 7.05 (d, 1H), 6.67-6.68 (m, 1H), 4.74 (t, 1H), 4.02-4.15 (m, 1H), 3.38-3.65 (m, 4H), 2.86-3.07 (m, 3H), 1.45-1.87 (m, 4H). Example 40 (4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-phenyl)((R)-3-fluoropyrrolidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6): 8.09-8.21 (m, 2H), 7.87 (s, 1H), 7.45-7.58 (m, 3H), 7.32 (d, 2H), 7.05 (d, 1H), 6.67 (d, 1H), 5.31 (t, 1H), 3.64-3.79 (m, 2H), 3.38-3.65 (m, 4H), 3.46-3.60 (m, 4H), 2.89-2.92 (m, 2H), 1.99-2.16 (m, 2H). Example 41 (4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-phenyl)(3,3-difluoropiperidin-1-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6): 8.09-8.21 (m, 2H), 7.87 (s, 1H), 7.50-7.59 (m, 1H), 7.30-7.35 (m, 4H), 7.05 (d, 1H), 6.67 (dr, 1H), 3.37-3.90 (m, 6H), 2.89-2.92 (m, 2H), 2.04-2.11 (m, 2H), 1.67 (dr, 2H). Example 42 4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-N-isopropyl-N-methylbenzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6): 8.09-8.21 (m, 2H), 7.86 (s, 1H), 7.48-7.56 (m, 1H), 7.27-7.31 (m, 4H), 7.05 (d, 1H), 6.67 (d, 1H), 4.68 (dr, 0.4H), 3.79 (dr, 0.6H), 3.51-3.52 (m, 2H), 2.87-2.90 (m, 2H), 2.77 dr, 3H), 1.07 (m, 6H). Example 43 4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-N-(1-hydroxy-2-methylpropan-2-yl)benzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H LCMS [M+1]+: 437.2;1H NMR (500 MHz, DMSO-d6): 8.09-8.21 (m, 2H), 7.87 (s, 1H), 7.73 (d, 2H), 7.49-7.53 (m, 1H), 7.47 (s, 1H), 7.32 (t, 1H), 7.05 (d, 2H), 6.67 (d, 1H), 4.91 (t, 1H), 3.48-3.52 (m, 4H), 2.89-2.92 (m, 2H), 1.30 (s, 6H). Example 44 4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-N-(1-hydroxy-2-methylpropan-2-yl)benzamide The title compound was synthesized in a similar way as the title compound in Example 11.1H NMR (500 MHz, DMSO-d6): 8.09-8.21 (m, 2H), 7.96 (d, 2H), 7.87 (s, 1H), 7.52-7.61 (m, 1H), 742 (d, 2H), 7.06 (s, 1H), 6.68 (s, 1H), 4.09 (t, 2H), 3.48-3.52 (m, 2H), 2.89-2.94 (m, 2H), 1.10 (s, 6H). Example 45 (4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)phenyl)-(7,8-dihydro-1,6-naphthyridin-6(5H)-yl)methanone The title compound was synthesized in a similar way as the title compound in Example 11.1H LCMS [M+1]+: 482.2;1H NMR (500 MHz, DMSO-d6): 8.38 (s, 1H), 8.09-8.21 (m, 2H), 7.86 (s, 1H), 7.51-7.69 (m, 3H), 7.34-7.40 (m, 4H), 7.23 (dr, 1H), 7.05 (d, 1H), 6.67 (s, 1H), 4.61-4.77 (m, 2H), 3.64-3.92 (m, 2H), 3.53-3.54 (m, 2H), 2.90-2.94 (m, 4H). Example 46 Benzyl 4-bromophenethylcarbamate To a solution of 2-(4-bromophenyl)ethanamine hydrochloride (10.0 g, 49.9 mmol) in THF (40 mL) and water (15 mL) was added Na2CO3(15.9 g, 150 mmol) and CbzCl (10.2 g, 59.9 mmol) at 0° C. The reaction mixture was stirred at room temperature for 2 h. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:2) to afford the title compound as white solid (16.7 g, 100%).1H NMR (500 MHz, DMSO-d6): 7.45 (d, 2H), 7.29-7.36 (m, 6H), 7.15 (d, 2H), 4.99 (s, 2H), 3.20-3.24 (m, 2H), 2.69 (t, 2H). Example 47 Benzyl 4-cyanophenethylcarbamate To a solution of benzyl 4-bromophenethylcarbamate (13.7 g, 41.1 mmol) in DMF (40 mL) was added Zn(CN)2(4.93 g, 41.2 mmol) and Pd(PPh3)4(4.75 g, 4.11 mmol). The reaction mixture was stirred at 100° C. under N2overnight. TLC showed the reaction completed. The reaction mixture was quenched with water and filtered. The residue was stirred in a mixed solvent of ethyl acetate and hexane (100 mL, EtOAc:Hexane=1:6) for 30 min. It was next filtered and dried to afford the title compound as white solid (8.9 g, 77.6%).1H NMR (500 MHz, DMSO-d6): 7.74 (d, 2H), 7.28-7.41 (m, 8H), 4.99 (s, 2H), 3.25-3.29 (m, 2H), 2.81 (t, 2H). Example 48 Benzyl 4-(2H-tetrazol-5-yl)phenethylcarbamate To a solution of benzyl 4-cyanophenethylcarbamate (5.0 g, 20.3 mmol) was added TMSN3(9.35 g, 81.2 mmol) and TBAF (monohydrate form, 2.65 g, 10.1 mmol). The reaction mixture was stirred at 90° C. under N2overnight. TLC showed the reaction completed. The reaction mixture was concentrated in vacuo. The crude product was purified by column chromatography to afford the title compound as white solid (5.0 g, 76% yield).1H NMR (500 MHz, DMSO-d6): 7.95 (d, 2H), 7.29-7.44 (m, 8H), 5.00 (s, 2H), 3.27-3.29 (m, 2H), 2.81 (t, 2H). Example 49 Benzyl 4-(2-methyl-2H-tetrazol-5-yl)phenethylcarbamate A4) and Benzyl 4-(1-methyl-1H-tetrazol-5-yl)phenethylcarbamate To a solution of benzyl 4-(2H-tetrazol-5-yl)phenethylcarbamate (1.97 g, 6.09 mmol) in THF (40 mL) was added 2.5 N (trimethylsilyl)diazomethane solution in hexane (10 mL) slowly at 0° C. The reaction mixture was stirred at 0° C. for 1 h. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:2) to afford benzyl 4-(2-methyl-2H-tetrazol-5-yl)phenethylcarbamate (1.37 g, 67%) and benzyl 4-(1-methyl-1H-tetrazol-5-yl)phenethylcarbamate (400 mg, 19%). Benzyl 4-(2-methyl-2H-tetrazol-5-yl)phenethylcarbamate.1H NMR (500 MHz, DMSO-d6): 7.96 (d, 2H), 7.30-7.39 (m, 8H), 5.00 (s, 2H), 4.42 (s, 3H), 3.27-3.29 (m, 2H), 2.80 (t, 2H). Benzyl 4-(1-methyl-1H-tetrazol-5-yl)phenethylcarbamate.1H NMR (500 MHz, DMSO-d6): 7.70 (d, 2H), 7.45 (d, 2H), 7.29-7.39 (m, 6H), 5.00 (s, 2H), 4.15 (s, 3H), 3.27-3.29 (m, 2H), 2.84 (t, 2H). Example 50 2-(4-(2-Methyl-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of benzyl 4-(2-methyl-2H-tetrazol-5-yl)phenethylcarbamate (1.37 g, 0.27 mmol) in MeOH (20 mL) was added Pd/C (10%). The reaction mixture was stirred at 40° C. under H2. for 2 h. TLC showed the reaction completed. The reaction mixture was filtered through diatomite. The filtrate was concentrated in vacuo to afford the title compound as white solid, which was used for the next step directly. Example 51 N5-(4-(2-Methyl-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (109.4 mg, 38% yield).1H NMR (500 MHz, DMSO-d6): 8.18-8.47 (m, 2H), 7.98 (d, 2H), 7.87 (s, 1H), 7.53-7.61 (m, 1H), 7.45 (t, 2H), 7.05 (d, 1H), 6.67 (dr, 1H), 4.41 (s, 3H), 3.52-3.56 (m, 2H), 2.89-2.96 (m, 2H). Example 52 2-(4-(1-Methyl-1H-tetrazol-5-yl)phenyl)ethanamine To a solution of benzyl 4-(1-methyl-1H-tetrazol-5-yl)phenethylcarbamate (400 mg, 1.18 mmol) in MeOH (10 mL) was added Pd/C (10%). The reaction mixture was stirred at 40° C. under H2for 2 h. TLC showed the reaction completed. The reaction mixture was filtered through diatomite. The filtrate was concentrated in vacuo to afford the title compound as white solid, which was used for the next step directly. Example 53 N5-(4-(1-Methyl-1H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (82.7 mg, 24% yield).1H NMR (500 MHz, DMSO-d6): 8.12-8.21 (m, 2H), 7.86 (s, 1H), 7.79 (d, 2H), 7.50-7.62 (m, 3H), 7.05 (d, 1H), 6.67 (dr, 1H), 4.15 (s, 3H), 3.55-3.56 (m, 2H), 2.96-2.99 (m, 2H). Example 54 5-(4-Bromophenyl)-2-methyl-2H-tetrazole To a solution of 5-(4-bromophenyl)-2H-tetrazole (10 g, 44.43 mmol) in THF (150 mL) was added 2.5 N (trimethylsilyl)diazomethane solution in hexane (50 mL) slowly at 0° C. The reaction mixture was stirred at 0° C. for 1 h. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:5) to afford the title compound as white solid (6.24 g, 59%).1H NMR (500 MHz, DMSO-d6): 7.84 (d, 2H), 7.62 (d, 2H), 4.28 (s, 3H). Example 55 tert-Butyl 4-(2-methyl-2H-tetrazol-5-yl)phenethylcarbamate To a solution of 5-(4-bromophenyl)-2-methyl-2H-tetrazole (6.24 g, 26.1 mmol) in toluene (40 mL) and water (10 mL) was added cesium carbonate (17 g, 52.2 mmol), Pd(dppf)Cl2(1.9 g, 2.61 mmol) and potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (7.21 g, 28.7 mmol). The reaction mixture was stirred at 80° C. under N2overnight. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=2:5) to afford the title compound as white solid (6.3 g, 79.7%).1H NMR (500 MHz, DMSO-d6): 7.96 (d, 2H), 7.38 (d, 2H), 6.92 (t, 1H), 4.42 (s, 3H), 3.17-3.21 (m, 2H), 2.77 (t, 2H), 1.36 (s, 9H). Example 56 2-(4-(2-Methyl-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of tert-butyl 4-(2-methyl-2H-tetrazol-5-yl)phenethylcarbamate (6.22 g, 20.5 mmol) in 1,4-dioxane (4 mL) was added 4N HCL/dioxane (4 mL). The reaction mixture was stirred at room temperature overnight. TLC showed the reaction completed. The excess HCl and 1,4-dioxane were removed to give the crude product (5.1 g, 90%), which was used for the next step directly (See Example 51). Example 57 Benzyl 4-(2-ethyl-2H-tetrazol-5-yl)phenethylcarbamate To a solution of benzyl 4-(2h-tetrazol-5-yl)phenethylcarbamate (300 mg, 0.93 mmol) in MeCN (15 mL) was added cesium carbonate (630 mg, 1.93 mmol) and iodoethane (800 mg, 5.12 mmol). The reaction mixture was stirred at 70° C. for 2 h. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:3) to afford the title compound as white solid (230 mg, 70.5%).1H NMR (500 MHz, DMSO-d6): 7.97 (d, 2H), 7.29-7.39 (m, 8H), 5.00 (s, 2H), 4.73-4.77 (m, 2H), 3.26-3.33 (m, 2H), 2.80 (t, 2H), 1.57 (t, 3H). Example 58 2-(4-(2-Ethyl-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of benzyl 4-(2-ethyl-2H-tetrazol-5-yl)phenethylcarbamate (230 mg, 0.65 mmol) in MeOH (20 mL) was added Pd/C (10%). The reaction mixture was stirred under H2at 40° C. for 2 h. TLC showed the reaction completed. The reaction mixture was filtered through diatomite. The filtrate was concentrated in vacuo to afford the title compound as white solid (120 mg, 86%), which was used for the next step directly. Example 59 N5-(4-(2-Ethyl-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (104 mg, 50% yield).1H NMR (500 MHz, DMSO-d6): 8.18-8.47 (m, 2H), 7.98 (d, 2H), 7.87 (s, 1H), 7.53-7.62 (m, 1H), 7.45 (t, 2H), 7.05 (d, 1H), 6.67 (dr, 1H), 4.42-4.47 (m, 2H), 3.52-3.56 (m, 2H), 2.93-2.96 (m, 2H), 1.57 (t, 3H). Example 60 Benzyl 4-(2-isopropyl-2H-tetrazol-5-yl)phenethylcarbamate To a solution of benzyl 4-(2h-tetrazol-5-yl)phenethylcarbamate (300 mg, 0.93 mmol) in MeCN (15 mL) was added cesium carbonate (630 mg, 1.93 mmol) and 2-iodopropane (473 mg, 2.98 mmol). The reaction mixture was stirred at 70° C. for 2 h. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:3) to afford the title compound as white solid (253 mg, 74.6%).1H NMR (500 MHz, DMSO-d6): 7.97 (d, 2H), 7.30-7.39 (m, 8H), 5.16-5.19 (m, 1H), 5.00 (s, 2H), 3.27-3.33 (m, 2H), 2.80 (t, 2H), 1.63 (dd, 6H). Example 61 2-(4-(2-Isopropyl-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of benzyl 4-(2-isopropyl-2H-tetrazol-5-yl)phenethylcarbamate (253 mg, 0.69 mmol) in MeOH (20 mL) was added Pd/C (10%). The reaction mixture was stirred under H2at 40° C. for 2 h. TLC showed the reaction completed. The reaction mixture was filtered through diatomite. The filtrate was concentrated in vacuo to afford the title compound as white solid (143 mg, 90%), which was used for the next step directly. Example 62 N5-(4-(2-Isopropyl-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (132.7 mg, 56% yield).1H NMR (500 MHz, DMSO-d6): 8.18-8.47 (m, 2H), 7.98 (d, 2H), 7.87 (s, 1H), 7.53-7.61 (m, 1H), 7.45 (t, 2H), 7.05 (d, 1H), 6.67 (dr, 1H), 5.15-5.18 (m, 1H), 3.53-3.56 (m, 2H), 2.89-2.96 (m, 2H), 1.61 (d, 6H). Example 63 Benzyl 4-(2-(2-methoxyethyl)-2H-tetrazol-5-yl)phenethylcarbamate To a solution of benzyl 4-(2h-tetrazol-5-yl)phenethylcarbamate (300 mg, 1.03 mmol) in MeCN (15 mL) was added cesium carbonate (674 mg, 2.07 mmol) and 1-bromo-2-methoxyethane (288 mg, 2.07 mmol). The reaction mixture was stirred at 70° C. for 2 h. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:2) to afford the title compound as yellow oil (248 mg, 63%). Example 64 2-(4-(2-(2-Methoxyethyl)-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of benzyl 4-(2-(2-methoxyethyl)-2H-tetrazol-5-yl)phenethylcarbamate (248 mg, 0.65 mmol) in MeOH (10 mL) was added Pd/C (10%). The reaction mixture was stirred under H2at 50° C. for 2 h. TLC showed the reaction completed. The reaction mixture was filtered through diatomite. The filtrate was concentrated in vacuo to afford the title compound as yellow oil (150 mg, 94%), which was used for the next reaction directly. Example 65 N5-(4-(2-(2-Methoxyethyl)-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (91.7 mg, 36% yield).1H NMR (500 MHz, DMSO-d6): 8.18-8.47 (m, 2H), 8.05 (d, 2H), 7.92 (s, 1H), 7.57-7.66 (m, 1H), 7.45 (t, 2H), 7.11 (d, 1H), 6.73 (dr, 1H), 4.95 (t, 2H), 3.96 (t, 2H), 3.59-3.61 (m, 2H), 3.29 (s, 3H), 2.99-3.02 (m, 2H). Example 66 Benzyl 4-(2-propyl-2H-tetrazol-5-yl)phenethylcarbamate To a solution of benzyl 4-(2h-tetrazol-5-yl)phenethylcarbamate (232 mg, 0.72 mmol) in MeCN (15 mL) was added cesium carbonate (700 mg, 3.58 mmol) and 1-bromopropane (441 mg, 3.58 mmol). The reaction mixture was stirred at 70° C. for 2 h. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:4) to afford the title compound as white solid (200 mg, 76.6%).1H NMR (500 MHz, DMSO-d6): 7.97 (d, 2H), 7.29-7.39 (m, 8H), 5.00 (s, 2H), 4.69 (t, 2H), 3.27-3.33 (m, 2H), 2.80 (t, 2H), 1.96-2.00 (m, 2H), 0.89 (t, 3H). Example 67 2-(4-(2-Propyl-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of benzyl 4-(2-propyl-2H-tetrazol-5-yl)phenethylcarbamate (200 mg, 0.54 mmol) in MeOH (20 mL) was added Pd/C (10%). The reaction mixture was stirred under H2at 40° C. for 2 h. TLC showed the reaction completed. The reaction mixture was filtered through diatomite. The filtrate was concentrated in vacuo to afford the title compound as white solid (100 mg, 80%), which was used for the next reaction directly. Example 68 N5-(4-(2-Propyl-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (95.4 mg, 66.7% yield).1H NMR (500 MHz, DMSO-d6): 8.18-8.47 (m, 2H), 7.98 (d, 2H), 7.87 (s, 1H), 7.53-7.61 (m, 1H), 7.45 (t, 2H), 7.05 (d, 1H), 6.67 (dr, 1H), 4.69 (t, 2H), 3.53-3.56 (m, 2H), 2.93-2.96 (m, 2H), 1.96-2.00 (m, 2H), 0.899 (t, 3H). Example 69 Benzyl 4-(2-(2,2,2-trifluoroethyl)-2H-tetrazol-5-yl)phenethylcarbamate To a solution of tert-butyl 4-(2H-tetrazol-5-yl)phenethylcarbamate (285 mg, 0.98 mmol) in DMF (15 mL) was added cesium carbonate (645 mg, 1.98 mmol) and 2,2,2-trifluoroethyl 4-methylbenzenesulfonate (321 mg, 1.97 mmol). The reaction mixture was stirred at 100° C. overnight. TLC showed the reaction completed. The reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:4) to afford the title compound as yellow oil (81 mg, 22.3%).1H NMR (500 MHz, DMSO-d6): 7.99 (d, 2H), 7.41 (d, 2H), 6.91 (t, 1H), 6.01-6.06 (m, 2H), 3.17-3.20 (m, 2H), 2.78 (t, 2H), 1.35 (s, 9H). Example 70 2-(4-(2-(2,2,2-Trifluoroethyl)-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of benzyl 4-(2-(2,2,2-trifluoroethyl)-2H-tetrazol-5-yl)phenethylcarbamate (81 mg, 0.22 mmol) in dioxane (4 mL) was added 4N HCl/dioxane (4 mL). The reaction mixture was stirred at room temperature overnight. TLC showed the reaction completed. The excess HCl and dioxane were removed to afford the crude product, which was used for the next step directly. Example 71 N5-(4-(2-(2,2,2-Trifluoroethyl)-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (24.9 mg, 25% yield).1H NMR (500 MHz, DMSO-d6): 8.18-8.47 (m, 2H), 8.03 (dr, 2H), 7.87 (s, 1H), 7.49-7.63 (m, 3H), 7.06 (s, 1H), 6.68 (s, 1H), 6.04 (dr, 2H), 3.55-3.56 (m, 2H), 2.95-2.96 (m, 2H). Example 72 tert-Butyl 4-(6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-6-carbonyl)phenethylcarbamate A mixture of 4-(2-((tert-butoxycarbonyl)amino)ethyl)benzoic acid (265 mg, 1 mmol), HATU (418 mg, 1.1 mmol) and DIPEA (387 mg, 3 mmol) in DCM (10 mL) was stirred at room temperature for 0.5 h. Then 6,7-dihydro-5H-pyrrolo[3,4-b]pyridine (193 mg, 1 mmol) was added and the reaction was stirred at room temperature overnight. TLC showed the reaction completed. The reaction mixture was concentrated and purified by column chromatography to afford the title compound as yellow solid (250 mg, 68.1% yield).1H NMR (500 MHz, DMSO-d6) δ 8.47 (dd, J=14.7, 4.5 Hz, 1H), 7.78 (dd, J=69.0, 7.6 Hz, 1H), 7.56 (dd, J=11.7, 8.0 Hz, 2H), 7.36-7.28 (m, 3H), 6.94 (d, J=5.6 Hz, 1H), 4.83 (t, J=29.2 Hz, 4H), 3.68-3.57 (m, 1H), 3.24-3.08 (m, 3H), 1.37 (s, 9H). Example 73 (4-(2-Aminoethyl)phenyl)(5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)methanone To a stirred mixture of tert-butyl 4-(6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-6-carbonyl)phenethylcarbamate (250 mg, 0.68 mmol) in dioxane (8 mL) was added 4N HCl in dioxane (3 mL) at room temperature. The reaction mixture was stirred at room temperature for 2 h. TLC showed the reaction completed. The reaction mixture was concentrated in vacuo to afford the crude product, which was used for the next step directly. Example 74 (4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-phenyl)(5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)methanone The reaction was carried out as in Example 5 to afford the title compound as white solid (88.7 mg, 28.0% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.53-8.01 (m, 3H), 7.90-7.49 (m, 5H), 7.41-7.26 (m, 3H), 7.06 (d, J=3.0 Hz, 1H), 6.68 (s, 1H), 4.93-4.73 (m, 4H), 3.54 (dd, J=13.1, 6.6 Hz, 2H), 2.93 (d, J=5.6 Hz, 2H); LC-MS (m/z): 468.2[M+H]+ Example 75 2-(Furan-2-yl)-N-(4-(methylsulfonyl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-amine The reaction was carried out as in Example 5 to afford the title compound as white solid (150 mg, 39.1% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.22 (s, 2H), 7.91-7.80 (m, 3H), 7.56 (dd, J=27.8, 18.4 Hz, 3H), 7.06 (d, J=3.3 Hz, 1H), 6.68 (d, J=1.6 Hz, 1H), 3.54 (dd, J=12.8, 6.5 Hz, 2H), 3.19 (s, 3H), 2.99 (t, J=7.1 Hz, 2H). Example 76 4-(2-((2-(Furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)amino)ethyl)benzene-sulfonamide The reaction was carried out as in Example 5 to afford the title compound as white solid (27.3 mg, 7.1% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.34 (d, J=120.0 Hz, 2H), 7.88 (s, 1H), 7.76 (d, J=8.0 Hz, 2H), 7.62-7.42 (m, 3H), 7.29 (s, 2H), 7.06 (d, J=3.2 Hz, 1H), 6.68 (d, J=1.5 Hz, 1H), 3.56-3.46 (m, 2H), 2.95 (t, J=7.2 Hz, 2H). Example 77 5-Bromo-2-(2-methyl-2H-tetrazol-5-yl)pyridine To a stirred mixture of 5-bromo-2-(2H-tetrazol-5-yl)pyridine (452 mg, 2 mmol) in THF (5 mL) was added (diazomethyl)trimethylsilane in hexane (2M, 5 mL, 10 mmol) at 0° C. The reaction mixture was stirred at room temperature for 3 h. TLC showed the reaction completed. The reaction mixture was quenched with water (30 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography to afford the title compound as white solid (180 mg, 28.5% yield).1H NMR (500 MHz, DMSO-d6) δ: 9.00-8.89 (m, 1H), 8.32 (dd, J=8.5, 2.4 Hz, 1H), 8.14 (d, J=8.4 Hz, 1H), 4.34 (s, 3H). Example 78 tert-Butyl 2-(6-(2-methyl-2H-tetrazol-5-yl)pyridin-3-yl)ethylcarbamate A mixture of 5-bromo-2-(2-methyl-2H-tetrazol-5-yl)pyridine (180 mg, 0.75 mmol), potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (207 mg, 0.83 mmol), cesium carbonate (733 mg, 2.25 mmol) and Pd(dppf)Cl2(15 mg) in toluene (6 mL) and water (2 mL) was stirred at 80° C. overnight. TLC showed the reaction completed. The reaction mixture was filtered. The filtrate was concentrated and purified by column chromatography to afford the title compound as white solid (150 mg, 65.8% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.65 (d, J=1.8 Hz, 1H), 8.17 (d, J=8.0 Hz, 1H), 7.92 (dd, J=8.1, 2.1 Hz, 1H), 6.97 (t, J=5.4 Hz, 1H), 4.41 (s, 3H), 3.25 (dd, J=12.8, 6.7 Hz, 2H), 2.84 (t, J=6.9 Hz, 2H), 1.34 (s, 9H). Example 79 2-(6-(2-Methyl-2H-tetrazol-5-yl)pyridin-3-yl)ethanamine To a stirred mixture of tert-butyl 4-(2-methyl-2H-tetrazol-5-yl)phenethylcarbamate (150 mg, 0.49 mmol) in dioxane (5 mL) was added 4N HCl in dioxane (3 mL) at room temperature. The reaction mixture was stirred at room temperature overnight. TLC showed the reaction completed. The reaction mixture was concentrated in vacuo to afford the crude product, which was used for the next step directly. Example 80 2-(Furan-2-yl)-N5-(2-(6-(2-methyl-2H-tetrazol-5-yl)pyridin-3-yl)ethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (39.0 mg, 19.7% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.72 (d, J=6.9 Hz, 1H), 8.27 (t, J=77.9 Hz, 3H), 7.99 (t, J=11.4 Hz, 1H), 7.87 (s, 1H), 7.60 (t, J=23.5 Hz, 1H), 7.05 (d, J=2.9 Hz, 1H), 6.68 (s, 1H), 4.40 (s, 3H), 3.59 (dd, J=12.5, 6.3 Hz, 2H), 3.02 (t, J=6.6 Hz, 2H). Example 81 3-(4-Bromophenyl)-4-methyl-4H-1,2,4-triazole To a stirred mixture of 3-(4-bromophenyl)-4H-1,2,4-triazole (448 mg, 2 mmol) in THF (5 mL) was added (diazomethyl)trimethylsilane (5 mL, 10 mmol) at 0° C. The reaction mixture was stirred at room temperature overnight. TLC showed the reaction completed. The reaction mixture was concentrated in vacuo. The crude product was purified by column chromatography to afford the title compound as yellow oil (100 mg, 21.2% yield). Example 82 tert-Butyl 4-(4-methyl-4H-1,2,4-triazol-3-yl)phenethylcarbamate A mixture of 3-(4-bromophenyl)-4-methyl-4H-1,2,4-triazole (100 mg, 0.42 mmol), potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (116 mg, 0.46 mmol), cesium carbonate (410 mg, 1.26 mmol) and Pd(dppf)Cl2(15 mg) in toluene (3 mL) and water (1 mL) was stirred at 80° C. overnight. TLC showed the reaction completed. The reaction mixture was filtered. The filtrate was concentrated and purified by column chromatography to afford the title compound as white solid (61 mg, 47.3% yield). Example 83 2-(4-(4-Methyl-4H-1,2,4-triazol-3-yl)phenyl)ethanamine To a stirred mixture of tert-butyl 4-(4-methyl-4H-1,2,4-triazol-3-yl)phenethylcarbamate (61 mg, 0.2 mmol) in dioxane (4 mL) was added 4N HCl in dioxane (2 mL) at room temperature. The reaction mixture was stirred at room temperature overnight. TLC showed the reaction completed. The reaction mixture was concentrated in vacuo to afford the crude product, which was used for the next step directly. Example 84 2-(Furan-2-yl)-N5-(4-(4-methyl-4H-1,2,4-triazol-3-yl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (23.3 mg, 29.0% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.31 (d, J=135.6 Hz, 2H), 7.98 (s, 1H), 7.87 (s, 1H), 7.72 (d, J=7.9 Hz, 2H), 7.55 (dd, J=26.5, 21.1 Hz, 1H), 7.45 (d, J=7.8 Hz, 2H), 7.06 (d, J=3.3 Hz, 1H), 6.68 (s, 1H), 3.96 (s, 3H), 3.56 (d, J=6.0 Hz, 2H), 2.96 (t, J=7.2 Hz, 2H). Example 85 5-(Pyridin-2-yl)-1H-1,2,4-triazol-3-amine To a stirred mixture of CH3ONa (31.49 g, 583 mmol) and hydrazinecarboximidamide (64.5 g, 583 mmol) in MeOH (450 mL) was added methyl picolinate (40 g, 292 mmol) in MeOH (120 mL) at 0° C. slowly. The reaction mixture was stirred at 75° C. overnight. TLC showed the reaction completed. The reaction mixture was filtered. The filtrate was concentrated afford the crude product. Then 150 mL of H2O was added and the pH was adjusted to pH 5 with 36% hydrochloric acid. The precipitate formed was filtered to afford the title compound as white solid (21 g, 55.3% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.70-8.48 (m, 1H), 8.06-7.82 (m, 2H), 7.41 (ddd, J=7.2, 4.8, 1.4 Hz, 1H). Example 86 5-(Methylthio)-2-(pyridin-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine A mixture of 5-(pyridin-2-yl)-1H-1,2,4-triazol-3-amine (5.0 g, 31 mmol) and dimethyl cyanocarbonimidodithioate (4.5 g, 31 mmol) was stirred at 180° C. for 1 h. TLC showed the reaction completed. The residue was purified by column chromatography to afford the title compound as white solid (4.0 g, 50.0% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.95 (d, J=57.3 Hz, 2H), 8.74 (d, J=4.3 Hz, 1H), 8.22 (d, J=7.8 Hz, 1H), 8.01 (td, J=7.7, 1.7 Hz, 1H), 7.55 (dd, J=6.6, 4.8 Hz, 1H), 2.54 (s, 3H). Example 87 5-Methylsulfonyl-2-(1-oxidopyridin-1-ium-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine To a stirred mixture of 5-(methylthio)-2-(pyridin-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine (4.0 g, 15.4 mmol) in DCM (240 mL) was added m-CPBA (12.5 g, 61.6 mmol) in DCM (120 mL) at 0° C. The reaction mixture was stirred at room temperature overnight. TLC showed the reaction completed. The reaction mixture was concentrated in vacuo to afford the crude product. Then EtOH (80 mL) was added and the mixture was stirred at room temperature 1 h. The precipitate was filtered to afford the title compound as white solid (4.36 g, 92.2% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.95 (d, J=57.3 Hz, 2H), 8.74 (d, J=4.3 Hz, 1H), 8.22 (d, J=7.8 Hz, 1H), 8.01 (td, J=7.7, 1.7 Hz, 1H), 7.55 (dd, J=6.6, 4.8 Hz, 1H), 2.54 (s, 3H); LC-MS (m/z): 308.1 [M+H]+ Example 88 N5-[2-[4-(2-methyltetrazol-5-yl)phenyl]ethyl]-2-(1-oxidopyridin-1-ium-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (120 mg, 27.9% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.59-8.12 (m, 3H), 7.99 (d, J=8.2 Hz, 2H), 7.90-7.81 (m, 1H), 7.62 (dd, J=28.5, 22.9 Hz, 1H), 7.56-7.51 (m, 1H), 7.45 (q, J=9.3 Hz, 3H), 4.42 (s, 3H), 3.60-3.53 (m, 2H), 2.96 (t, J=7.3 Hz, 2H); LC-MS (m/z): 431.2[M+H]+ Example 89 tert-Butyl 4-(pyrimidin-2-yl)phenethylcarbamate A mixture of 2-(4-bromophenyl)pyrimidine (470 mg, 2.00 mol), cesium carbonate (1300 mg, 4.00 mmol), Pd(dppf)Cl2(146 mg, 0.20 mmol) and potassium tert-butyl n-[2-(trifluoroboranuidyl)ethyl]carbamate (552 mg, 2.20 mmol) in toluene (7.5 ml) and water (2.5 mL) was stirred at 80° C. for 15 hours. The reaction mixture was diluted with ethyl acetate (100 mL) and washed with water and brine. The organic layer was dried with anhydrous sodium sulfate, concentrated and purified by column chromatography to afford the title compound as white solid (550 mg, 92.0% yield).1HNMR (500 MHz, DMSO-d6) δ: 8.88 (d, 2H), 8.31 (d, 2H), 7.41 (t, 1H), 7.35 (d, 2H), 6.91 (t, 1H), 3.19 (q, 2H), 2.77 (t, 2H), 1.37 (s, 9H). Example 90 2-(4-(Pyrimidin-2-yl)phenyl)ethanamine To a stirred solution of tert-butyl 4-(pyrimidin-2-yl)phenethylcarbamate (550 mg, 1.84 mol) in 1,4-dioxane (3 ml) was added 4M HCl in 1,4-dioxane (2 mL). After stirring at 35° C. for 3 hours, the reaction mixture was concentrated in vacuo to afford the title compound as white solid, which was used for the next step directly. Example 91 2-(Furan-2-yl)-N5-(4-(pyrimidin-2-yl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine To a stirred solution of 2-(4-(pyrimidin-2-yl)phenyl)ethanamine (366 mg, 1.84 mmol) and 2-(furan-2-yl)-5-(methylsulfonyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine (412 mg, 0.1.47 mmol) in MeCN (5 mL) was added TEA to adjust pH to 8. After stirring at room temperature for 15 h, the precipitated solid was collected and dried to afford the title compound as yellow solid (390 mg, 66.5% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.88 (d, 2H), 8.05-8.49 (d, 2H), 8.33 (d, 2H), 7.87 (s, 1H), 7.53 (d, 1H), 7.42 (q, 3H), 7.06 (d, 1H), 6.67 (d, 1H), 3.06 (t, 2H), 2.95 (t, 2H); LCMS m/z [M+H]+: 400.2 Example 92 5-(Methylsulfonyl)-2-phenyl-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine The title compound was prepared in a similar way as the title compound in Example 4.1H NMR (500 MHz, DMSO-d6) δ: 9.83 (s, 1H), 9.39 (s, 1H), 8.32-8.16 (m, 2H), 7.66-7.54 (m, 3H), 3.38 (s, 3H). Example 93 N5-(4-(2-Methyl-2H-tetrazol-5-yl)phenethyl)-2-phenyl-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (0.132 g, 43.7% yield). LC-MS m/z [M+H]+: 414;1H NMR (500 MHz, DMSO-d6) δ: 8.42-8.12 (d, J=6.4 Hz, 3H), 8.00 (d, J=8.1 Hz, 3H), 7.59-7.42 (m, 6H), 4.42 (s, 3H), 3.57 (d, J=6.4 Hz, 2H), 2.96 (t, J=7.2 Hz, 2H). Example 94 tert-Butyl 4-(pyridin-2-yl)phenethylcarbamate A solution of 2-(4-bromophenyl)pyridine (0.6 g, 2.56 mmol), potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (0.708 g, 2.82 mmol), cesium carbonate (2.5 g, 7.68 mmol), Pd(dppf)Cl2(0.095 g, 0.13 mmol) in PhMe (12 mL) and water (4 mL) was stirred at 80° C. overnight under N2. The reaction mixture was filtered through Celite, the filtrate was concentrated in vacuo, diluted with water (30 mL) and extracted with ethyl acetate (30 mL×2), the combined organic layers washed with water (20 mL) and brine (20 mL). After drying with Na2SO4and removal of solvent, the residue purified by column chromatography to afford the title compound as white solid (0.65 g, 85.1% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.71-8.59 (m, 1H), 8.01 (d, J=8.2 Hz, 2H), 7.95-7.82 (m, 2H), 7.32 (ddd, J=8.3, 4.8, 2.4 Hz, 3H), 6.91 (t, J=5.4 Hz, 1H), 3.25-3.12 (m, 2H), 2.76 (t, J=7.4 Hz, 2H), 1.38 (s, 9H). Example 95 2-(4-(Pyridin-2-yl)phenyl)ethanamine HCl (4 mL, 4N solution in 1,4-dioxane) was added to a stirred solution of tert-butyl 4-(pyridin-2-yl)phenethylcarbamate (0.31 g, 1.04 mmol) in MeOH (1 mL) at room temperature for 2 h, then the excess solvent was removed under reduced pressure to afford the crude product was used for next step reaction without further purification. Example 96 2-(Furan-2-yl)-N5-(4-(pyridin-2-yl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (0.1471 g, 44.4% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.65 (dd, J=4.7, 0.7 Hz, 1H), 8.33 (br, 2H), 8.03 (d, J=8.1 Hz, 2H), 7.94 (d, J=8.0 Hz, 1H), 7.91-7.81 (m, 2H), 7.55 (dd, J=26.4, 20.8 Hz, 1H), 7.39 (t, J=6.9 Hz, 2H), 7.33 (ddd, J=7.3, 4.8, 0.9 Hz, 1H), 7.07 (d, J=3.2 Hz, 1H), 6.74-6.62 (m, 1H), 3.54 (dd, J=13.4, 6.8 Hz, 2H), 2.93 (t, J=7.3 Hz, 2H). Example 97 tert-Butyl 4-(pyridin-3-yl)phenethylcarbamate A solution of 3-(4-bromophenyl)pyridine (0.6 g, 2.56 mmol), potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (0.708 g, 2.82 mmol), cesium carbonate (2.5 g, 7.68 mmol), Pd(dppf)Cl2(0.095 g, 0.13 mmol) in PhMe (12 mL) and water (4 mL) was stirred at 80° C. overnight under N2. The reaction mixture was filtered through Celite, the filtrate was concentrated in vacuo, diluted with water (30 mL) and extracted with ethyl acetate (30 mL×2), the combined organic layers washed with water (20 mL) and brine (20 mL). After drying with Na2SO4and removal of solvent, the residue purified by column chromatography to afford the title compound as light-yellow oil (0.201 g, 26.3% yield).1H NMR (500 MHz, DMSO-d6) 8.88 (d, J=1.9 Hz, 1H), 8.56 (dd, J=4.7, 1.6 Hz, 1H), 8.09-8.01 (m, 1H), 7.66 (d, J=8.2 Hz, 2H), 7.48 (ddd, J=8.0, 4.8, 0.6 Hz, 1H), 7.33 (d, J=8.1 Hz, 2H), 6.91 (t, J=5.4 Hz, 1H), 3.18 (d, J=5.2 Hz, 2H), 2.75 (t, J=7.4 Hz, 2H), 1.38 (s, 9H). Example 98 2-(4-(Pyridin-3-yl)phenyl)ethanamine HCl (4 mL, 4N solution in 1,4-dioxane) was added to a stirred solution of tert-Butyl 4-(pyridin-3-yl)phenethylcarbamate (0.201 g, 0.673 mmol) in MeOH (2 mL) at room temperature for 2 h, then the excess solvent was removed under reduced pressure to afford the crude product was used for next step reaction without further purification. Example 99 2-(Furan-2-yl)-N5-(4-(pyridin-3-yl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (0.0342 g, 14.2% yield). LC-MS m/z [M+H]+: 399;1H NMR (500 MHz, DMSO-d6) δ: 8.96-8.83 (m, 1H), 8.55 (dd, J=4.7, 1.5 Hz, 1H), 8.34 (br, 2H), 8.08-8.03 (m, 1H), 7.87 (s, 1H), 7.68 (d, J=8.0 Hz, 2H), 7.54 (dd, J=26.5, 20.9 Hz, 1H), 7.47 (dd, J=7.9, 4.8 Hz, 1H), 7.40 (t, J=7.1 Hz, 2H), 7.07 (dd, J=3.3, 0.6 Hz, 1H), 6.68 (dd, J=3.2, 1.7 Hz, 1H), 3.54 (dd, J=13.1, 6.5 Hz, 2H), 2.93 (t, J=7.3 Hz, 2H). Example 100 tert-Butyl 4-(pyridin-4-yl)phenethylcarbamate A solution of 4-(4-bromophenyl)pyridine (0.6 g, 2.56 mmol), potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (0.708 g, 2.82 mmol), cesium carbonate (2.5 g, 7.68 mmol), Pd(dppf)Cl2(0.095 g, 0.13 mmol) in PhMe (12 mL) and water (4 mL) was stirred at 80° C. overnight under N2. The reaction mixture was filtered through Celite, the filtrate was concentrated in vacuo, diluted with water (30 mL) and extracted with ethyl acetate (30 mL×2), the combined organic layers washed with water (20 mL) and brine (20 mL). After drying with Na2SO4and removal of solvent, the residue purified by column chromatography to afford the title compound as light-yellow solid (0.19 g, 24.9% yield).1H NMR (500 MHz, DMSO-d6) 8.62 (dd, J=4.5, 1.6 Hz, 2H), 7.79-7.62 (m, 4H), 7.35 (d, J=8.1 Hz, 2H), 6.92 (t, J=5.3 Hz, 1H), 3.19 (dd, J=13.6, 6.6 Hz, 2H), 2.76 (t, J=7.3 Hz, 2H), 1.37 (s, 9H). Example 101 2-(4-(pyridin-4-yl)phenyl)ethanamine HCl (4 mL, 4N solution in 1,4-dioxane) was added to a stirred solution of tert-Butyl 4-(pyridin-4-yl)phenethylcarbamate (0.19 g, 0.637 mmol) in MeOH (2 mL) at room temperature for 2 h, then the excess solvent was removed under reduced pressure to afford the crude product was used for next step reaction without further purification. Example 102 2-(Furan-2-yl)-N5-(4-(pyridin-4-yl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (0.0727 g, 30.2% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.62 (d, J=5.9 Hz, 2H), 8.32 (br, 2H), 7.87 (s, 1H), 7.75 (d, J=8.0 Hz, 2H), 7.70 (dd, J=4.6, 1.6 Hz, 2H), 7.54 (dd, J=26.4, 20.8 Hz, 1H), 7.41 (d, J=7.9 Hz, 2H), 7.07 (d, J=3.3 Hz, 1H), 6.72-6.63 (m, 1H), 3.65-3.48 (m, 2H), 2.94 (t, J=7.3 Hz, 2H). Example 103 2-Benzyl-5-(4-bromophenyl)-2H-tetrazole To a solution of 5-(4-bromophenyl)-2H-tetrazole (1.0 g, 4.44 mmol) in MeCN (30 mL) was added Cs2CO3(2.9 g, 8.88 mmol) and 1-(bromomethyl)benzene (1.14 g, 6.66 mmol). The reaction mixture was stirred at 70° C. for 3 h. TLC showed the reaction completed, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:6) to afford the title compound as white solid (1.4 g, 100%).1H NMR (500 MHz, DMSO-d6): 7.98 (d, 2H), 7.76 (d, 2H), 7.38-7.42 (m, 5H), 6.00 (s, 2H). Example 104 tert-Butyl 4-(2-benzyl-2H-tetrazol-5-yl)phenethylcarbamate To a solution of 2-benzyl-5-(4-bromophenyl)-2H-tetrazole 1 (700 mg, 2.22 mmol) in toluene (20 mL) and water (7 mL) was added Cs2CO3(1.4 g, 4.44 mmol), Pd(dppf)Cl2(163 mg, 0.22 mmol) and potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (613 mg, 2.44 mmol). The reaction mixture was stirred at 80° C. overnight under N2. TLC showed the reaction completed, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:4) to afford the title compound as white solid (515 mg, 61%).1H NMR (500 MHz, DMSO-d6): 7.96 (d, 2H), 7.31-7.41 (m, 7H), 7.53-7.62 (m, 1H), 6.90 (t, 1H), 5.99 (s, 2H), 3.16-3.20 (m, 2H), 2.76 (t, 2H), 1.36 (s, 9H). Example 105 2-(4-(2-Benzyl-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of tert-butyl 4-(2-benzyl-2H-tetrazol-5-yl)phenethylcarbamate (381 mg, 1.00 mmol) in 1,4-dioxane (5 mL) was added 4N HCl/dioxane (3 mL). The reaction mixture was stirred at room temperature 1 h. TLC showed the reaction completed. The excess HCl and dioxane were removed. The crude product was use for the next step directly. Example 106 N5-(4-(2-Benzyl-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (102.1 mg, 24%).1H NMR (500 MHz, DMSO-d6): 8.16-8.46 (m, 2H), 7.98 (d, 2H), 7.87 (s, 1H), 7.51-7.60 (m, 1H), 7.37-7.50 (m, 7H), 7.05 (d, 1H), 6.68 (dr, 1H), 5.99 (s, 2H), 3.53-3.55 (m, 2H), 2.94 (t, 2H). Example 107 4-((5-(4-Bromophenyl)-2H-tetrazol-2-yl)methyl)pyridine To a solution of 5-(4-bromophenyl)-2H-tetrazole (1.0 g, 4.44 mmol) in MeCN (35 mL) was added Cs2CO3(5.9 g, 18.1 mmol) and 4-(bromomethyl)pyridine hydrochloride (1.0 g, 6.1 mmol). The reaction mixture was stirred at 70° C. for 3 h. TLC showed the reaction completed, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc) to afford the title compound as white solid (800 mg, 57%).1H NMR (500 MHz, DMSO-d6): 8.60 (dd, 2H), 8.00 (d, 2H), 7.77 (d, 2H), 7.33 (d, 2H), 6.12 (s, 2H). Example 108 tert-Butyl 4-(2-(pyridin-4-ylmethyl)-2H-tetrazol-5-yl)phenethylcarbamate To a solution of 4-((5-(4-bromophenyl)-2H-tetrazol-2-yl)methyl)pyridine (800 mg, 2.53 mmol) in toluene (20 mL) and water (7 mL) was added Cs2CO3(1.65 g, 5.07 mmol), Pd(dppf)Cl2(186 mg, 0.25 mmol) and potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]-carbamate (700 mg, 2.79 mmol). The reaction mixture was stirred at 80° C. overnight under N2. TLC showed the reaction completed, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:4) to afford the title compound as white solid (637 mg, 66%).1H NMR (500 MHz, DMSO-d6): 8.60 (dd, 2H), 7.98 (d, 2H), 7.38 (d, 2H), 7.32 (d, 2H), 6.91 (t, 1H), 6.10 (s, 2H), 3.16-3.20 (m, 2H), 2.76 (t, 2H), 1.36 (s, 9H). Example 109 2-(4-(2-(Pyridin-4-ylmethyl)-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of tert-butyl 4-(2-(pyridin-4-ylmethyl)-2H-tetrazol-5-yl)phenethyl-carbamate (300 mg, 0.80 mmol) in 1,4-dioxane (5 mL) was added 4N HCl/dioxane (3 mL). The reaction mixture was stirred at room temperature 1.5 h. TLC showed the reaction completed. The excess HCl and dioxane were removed. The crude product was use for the next step directly. Example 110 N5-(4-(2-(Pyridin-4-ylmethyl)-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (205.6 mg, 59.7%).1H NMR (500 MHz, DMSO-d6): 8.59 (d, 2H), 8.11-8.45 (m, 2H), 8.00 (d, 2H), 7.87 (s, 1H), 7.52-7.60 (m, 1H), 7.44-7.47 (m, 2H), 7.32 (d, 2H), 7.05 (d, 1H), 6.68 (dr, 1H), 6.10 (s, 2H), 3.53-3.55 (m, 2H), 2.94 (t, 2H). Example 111 3-((5-(4-Bromophenyl)-2H-tetrazol-2-yl)methyl)pyridine To a solution of 5-(4-bromophenyl)-2H-tetrazole (1.0 g, 4.44 mmol) in MeCN (30 mL) was added Cs2CO3(4.3 g, 13.2 mmol) and 3-(bromomethyl)pyridine hydrochloride (1.46 g, 5.7 mmol). The reaction mixture was stirred at 70° C. for 2 h. TLC showed the reaction completed, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:3) to afford the title compound as white solid (393 mg, 24%).1H NMR (500 MHz, DMSO-d6): 8.72 (d, 1H), 8.60 (dd, 1H), 7.98 (d, 2H), 7.85-7.86 (m, 1H), 7.76 (d, 2H), 7.44-7.46 (m, 1H), 6.09 (s, 1H). Example 112 tert-Butyl 4-(2-(pyridin-3-ylmethyl)-2H-tetrazol-5-yl)phenethylcarbamate To a solution of 3-((5-(4-bromophenyl)-2H-tetrazol-2-yl)methyl)pyridine (393 mg, 1.24 mmol) in toluene (15 mL) and water (5 mL) was added Cs2CO3(808 g, 2.48 mmol), Pd(dppf)Cl2(91 mg, 0.12 mmol) and potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (345 mg, 1.37 mmol). The reaction mixture was stirred at 80° C. overnight under N2. TLC showed the reaction completed, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:4) to afford the title compound as yellow oil (280 mg, 59%).1H NMR (500 MHz, DMSO-d6): 8.71 (d, 1H), 8.60 (dd, 1H), 7.96 (d, 2H), 7.84 (dt, 1H), 7.43-7.46 (m, 1H), 7.37 (m, 2H), 6.90 (t, 1H), 6.07 (s, 2H), 3.16-3.20 (m, 2H), 2.76 (t, 2H), 1.36 (s, 9H). Example 113 2-(4-(2-(Pyridin-3-ylmethyl)-2H-tetrazol-5-yl)phenyl)ethanamine To a solution of tert-butyl 4-(2-(pyridin-3-ylmethyl)-2H-tetrazol-5-yl)phenethyl-carbamate (280 mg, 0.73 mmol) in 1,4-dioxane (5 mL) was added 4N HCl/dioxane (3 mL). The reaction mixture was stirred at room temperature 1.5 h. TLC showed the reaction completed. The excess HCl and dioxane were removed. The crude product was use for the next step directly. Example 114 N5-(4-(2-(pyridin-3-ylmethyl)-2H-tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (111.6 mg, 36%).1H NMR (500 MHz, DMSO-d6): 8.71 (d, 1H), 8.59 (dd, 1H), 8.11-8.45 (m, 2H), 7.98 (d, 2H), 7.83-7.87 (m, 3H), 7.52-7.60 (m, 1H), 7.43-7.46 (m, 3H), 7.32 (d, 2H), 7.05 (d, 1H), 6.68 (dr, 1H), 6.07 (s, 2H), 3.53-3.55 (m, 2H), 2.94 (t, 2H). Example 115 2-(5-(4-Bromophenyl)-2H-tetrazol-2-yl)acetonitrile To a solution of 5-(4-bromophenyl)-2H-tetrazole (1.0 g, 4.44 mmol) in MeCN (30 mL) was added Cs2CO3(2.9 g, 8.92 mmol) and 2-bromoacetonitrile (700 mg, 5.83 mmol). The reaction mixture was stirred at 60° C. for 3 h. TLC showed the reaction completed, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:5) to afford the title compound as white solid (600 mg, 51.2%).1H NMR (500 MHz, DMSO-d6): 8.08 (d, 2H), 7.86 (d, 2H), 6.35 (s, 2H). Example 116 tert-Butyl 4-(2-(cyanomethyl)-2H-tetrazol-5-yl)phenethylcarbamate To a solution of 2-(5-(4-bromophenyl)-2H-tetrazol-2-yl)acetonitrile (600 mg, 2.27 mmol) in toluene (20 mL) and water (6 mL) was added Cs2CO3(1.5 g, 4.5 mmol), Pd(dppf)Cl2(166 mg, 0.22 mmol) and potassium tert-butyl N-[2-(trifluoroboranuidyl)ethyl]carbamate (627 mg, 2.50 mmol). The reaction mixture was stirred at 80° C. overnight under N2. TLC showed the reaction completed, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (EtOAc:Hexane=1:4) to afford the title compound as white solid (220 mg, 29%).1H NMR (500 MHz, DMSO-d6): 8.00 (d, 2H), 7.41 (d, 2H), 6.93 (t, 1H), 6.29 (s, 2H), 3.18-3.20 (m, 2H), 2.78 (t, 2H), 1.36 (s, 9H). Example 117 2-(5-(4-(2-Aminoethyl)phenyl)-2H-tetrazol-2-yl)acetonitrile To a solution of tert-butyl 4-(2-(cyanomethyl)-2H-tetrazol-5-yl)phenethylcarbamate (220 mg, 0.67 mmol) in 1,4-dioxane (5 mL) was added 4N HCl/dioxane (3 mL). The reaction mixture was stirred at room temperature 1.5 h. TLC showed the reaction completed. The excess HCl and dioxane were removed. The crude product was use for the next step directly. Example 118 2-(5-(4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-phenyl)-2H-tetrazol-2-yl)acetonitrile The reaction was carried out as in Example 5 to afford the title compound as white solid (15.5 mg, 10%). LCMS [M+1]+: 429.21H NMR (500 MHz, DMSO-d6): 8.17-8.24 (m, 2H), 8.01 (d, 2H), 7.87 (s, 1H), 7.47-7.62 (m, 3H), 7.44-7.47 (m, 3H), 7.06 (d, 1H), 6.68 (dr, 1H), 6.29 (s, 2H), 3.54-3.57 (m, 2H), 2.96 (t, 2H). Example 119 4-(2-Aminoethyl)benzonitrile To a solution of benzyl 4-cyanophenethylcarbamate (550 mg, 1.96 mmol) in MeOH (20 mL) was added Pd/C (10%, 50 mg). The reaction mixture was stirred at 40° C. overnight under Hz. TLC showed the reaction completed, the reaction mixture was filtered by diatomite. The filtrate was concentrated to afford the title compound as yellow oil. Example 120 4-(2-(7-Amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5-ylamino)ethyl)-benzonitrile The reaction was carried out as in Example 5 to afford the title compound as white solid (54 mg, 12.2%).1H NMR (500 MHz, DMSO-d6): 8.04-8.45 (m, 2H), 7.87 (s, 1H), 7.76 (d, 2H), 7.45-7.59 (m, 3H), 7.06 (d, 1H), 6.68 (dr, 1H), 3.50-3.54 (m, 2H), 2.96 (t, 2H). Example 121 2-(3-Fluorophenyl)-5-(methylsulfonyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine The title compound was prepared in a similar way as the title compound in Example 4.1H NMR (500 MHz, DMSO-d6) δ: 9.87 (s, 1H), 9.41 (s, 1H), 8.04 (d, 1H), 7.91 (d, 1H), 7.65 (q, 1H), 7.43 (dt, 1H), 3.38 (s, 3H). Example 122 2-(3-Fluorophenyl)-N5-(4-(2-methyl-2H-tetrazol-5-yl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine To a stirred solution of 2-(4-(2-methyl-2H-tetrazol-5-yl)phenyl)ethanamine (203 mg, 1.00 mmol) and 2-(3-fluorophenyl)-5-(methylsulfonyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-7-amine (308 mg, 1.00 mmol) in MeCN (5 mL) was added TEA to adjust PH to 8. The reaction mixture was stirred at room temperature for 15 h. The precipitated solid was collected by filtration, washed with methanol and dried to afford the title compound as white solid (226 mg, 52.4% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.05-8.49 (m, 2H), 7.99 (d, 2H), 7.95 (d, 1H), 7.81 (d, 1H), 7.57 (m, 2H), 7.46 (t, 2H), 7.35 (dt, 1H), 4.41 (s, 3H), 3.57 (q, 2H), 2.96 (t, 2H). Example 123 2-(3-Fluorophenyl)-N5-(4-(2-methyl-2H-tetrazol-5-yl)phenethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid.1H NMR (500 MHz, DMSO-d6) δ: 8.05-8.51 (m, 4H), 7.87 (s, 1H), 7.72 (q, 1H), 7.54 (m, 3H), 7.05 (d, 1H), 6.68 (t, 1H), 3.56 (q, 2H), 3.02 (t, 2H). Example 124 2-(4-(2H-Tetrazol-5-yl)phenyl)ethanamine To a solution of benzyl 4-(2H-tetrazol-5-yl)phenethylcarbamate (100 mg, 0.30 mmol) in MeOH (20 mL) was added Pd/C (10%, 20 mg). The reaction mixture was stirred under H2at 50° C. for 3 h. TLC showed the reaction completed. The reaction mixture was filtered through diatomite. The filtrate was concentrated to afford the title compound as white solid (60 mg, 100%).1H NMR (500 MHz, DMSO-d6) δ: 7.92 (d, 2H), 7.74 (dr, 2H), 7.26 (d, 2H), 3.07 (t, 2H), 2.86 (t, 2H). Example 125 N5-(4-(2H-Tetrazol-5-yl)phenethyl)-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (18.7 mg, 12%). LCMS [M+1]+: 390.21H NMR (500 MHz, DMSO-d6): 7.95 (d, 2H), 7.68 (s, 1H), 7.35 (d, 2H), 7.11 (d, 1H), 6.60 (dr, 1H), 3.64-3.67 (m, 2H), 2.94-2.99 (m, 2H). Example 126 2-(Furan-2-yl)-N5-(2-(pyridin-3-yl)ethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (64.8 mg, 20.12% yield).1H NMR (500 MHz, CDCl3) 8.47 (d, J=9.3 Hz, 1H), 8.41 (dd, J=4.7, 1.3 Hz, 1H), 8.19 (s, 2H), 7.86 (s, 1H), 7.67 (t, J=8.7 Hz, 1H), 7.53 (dd, J=29.5, 24.0 Hz, 1H), 7.31 (dd, J=7.7, 4.8 Hz, 1H), 7.05 (d, J=3.3 Hz, 1H), 6.67 (d, J=1.5 Hz, 1H), 3.51 (dd, J=13.3, 6.7 Hz, 2H), 2.88 (t, J=7.1 Hz, 2H). Example 127 2-(Furan-2-yl)-N5-(2-(6-methoxypyridin-3-yl)ethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound as white solid (135 mg, 38.4% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.50-8.05 (d, 2H), 8.02 (d, 1H), 7.87 (s, 1H), 7.63-7.46 (m, 2H), 7.06 (d, 1H), 6.75 (d, 1H), 6.67 (d, 1H), 3.81 (s, 3H), 3.46 (q, 2H), 2.81 (t, 2H). Example 128 2-(Furan-2-yl)-N5-(2-(pyrimidin-4-yl)ethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 42. The precipitated solid was filtered, washed with MeCN and water, and dried to afford the title compound as yellow solid (135 mg, 41.8% yield).1H NMR (500 MHz, DMSO-d6) δ: 9.09 (s, 1H), 8.68 (d, 1H), 8.50-8.05 (d, 2H), 7.87 (s, 1H), 7.50-7.43 (m, 2H), 7.06 (d, 1H), 6.67 (s, 1H), 3.66 (m, 2H), 3.03 (t, 2H). Example 129 2-(Furan-2-yl)-N5-(2-(pyridin-2-yl)ethyl)-[1,2,4]triazolo[1,5-a][1,3,5]triazine-5,7-diamine The reaction was carried out as in Example 5 to afford the title compound (361 mg, 74.4% yield).1H NMR (500 MHz, DMSO-d6) δ: 8.51 (s, 1H), 8.04-8.40 (d, 2H), 7.87 (s, 1H), 7.72 (q, 1H), 7.47-7.54 (d, 1H), 7.29 (d, 1H), 7.21 (t, 1H), 7.06 (s, 1H), 6.67 (s, 1H), 3.63 (t, 2H), 3.02 (t, 2H). The above exemplary embodiments are used as illustrations of the invention. These embodiments are not intended to limit the scope of the invention. In fact, the invention is intended to cover all alternatives, modifications, and equivalents of these embodiments. It should not be understood that the present invention is only limited to the illustrated examples. Biological Activities of the A2A Receptor Antagonists The antagonistic activities of the triazolotriazine derivatives of the present invention were measured in a functional cAMP production assay. The assay consists of NECA stimulation of cAMP production and its inhibition by A2AR antagonists in A2AR-expressing HEK293 cells (hADORA2A-HEK293). All cells were cultured in complete medium at 37° C. in 5% CO2. The cells were detached with pancreatin and collected at 200 g for 5 min. After resuspending the cells in fresh complete medium, the cell viability is counted using the trypan blue exclusion method. The cAMP production assay was conducted only when cell viability was greater than 95%. After the cells were diluted with Hank's buffered saline solution containing HEPES (5 mM), BSA stabilizer (0.1%) and 20 Rolipram (10 μM), cells were loaded into white opaque 384-well plates (˜500 cells/well, 10 μl/well) and incubated with test compound at a suitable concentration range (11 concentrations) for 20 min at room temperature. Then the A2A receptor agonist NECA (final concentration=EC80, which was determined in the same experiment slightly earlier) was added to the sample and the mixture was incubated again for 30 min at 37° C. The amount of cAMP production was determined using Eu-cAMP tracer and Ulight-anti-cAMP by measuring the ratio of the TR-FRET emission at 665 nm and fluorescent emission at 615 nm. The inhibition rate (%) was calculated according to the following formula. The IC50values were calculated from concentration−inhibition (%) curves after log transformation. %⁢Inhibition=(1-Ratio565⁢nm/615⁢nm⁢high-Ratio565⁢nm/615⁢nm⁢cm⁢pdRatio665⁢nm/615⁢nm⁢high-Ratio665⁢nm/615⁢nm⁢low)×100⁢% Table (2) shows representative antagonistic activities of the triazolotriazine derivatives of the present invention. TABLE 2The Potency of A2AR antagonistsCompoundsA2AR Potency (nM)Example 1143.42Example 1227.10Example 1333.02Example 1623.99Example 177.54Example 279.08Example 348.71Example 3513.78Example 3613.32Example 376.00Example 4219.35Example 4317.14Example 4527.38Example 519.66Example 597.87Example 629.08Example 6513.88Example 12514.48Example 12863.81Example 12912.67ZM2413859.00
101,507
11858943
DETAILED DESCRIPTION OF THE INVENTION As mentioned above, the invention relates to compounds that antagonize vasopressin receptors, particularly the V1a receptor, to products comprising the same, and to methods for their use and synthesis. In one embodiment, compounds are provided having the structure of Formula (I), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein represents an optional double bond; A and B are independently nitrogen or oxygen, with the proviso that A and B are not both oxygen; G is nitrogen or carbon; X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1band R1care independently hydrogen, lower alkyl, or spiroalkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, —O-heterocyclyl-R6, —NHR5, or —NR5R5; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; Q is aryl or heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; each R5is independently cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. As used herein, “lower alkyl” means a straight chain or branched alkyl group having from 1 to 8 carbon atoms, in some embodiments from 1 to 6 carbon atoms, in some embodiments from 1 to 4 carbon atoms, and in some embodiments from 1 to 2 carbon atoms. Examples of straight chain lower alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, and n-octyl groups. Examples of branched lower alkyl groups include, but are not limited to, isopropyl, iso-butyl, sec-butyl, t-butyl, neopentyl, isopentyl, and 2,2-dimethylpropyl groups. “Halo” or “halogen” refers to fluorine, chlorine, bromine, and iodine. “Hydroxy” refers to —OH. “Cyano” refers to —CN. “Lower haloalkyl” refers to a lower alkyl as defined above with one or more hydrogen atoms replaced with halogen. Examples of lower haloalkyl groups include, but are not limited to, —CF3, —CH2CF3, and the like. “Lower alkoxy” refers to a lower alkyl as defined above joined by way of an oxygen atom (i.e., —O-(lower alkyl). Examples of lower alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, n-butoxy, isopropoxy, sec-butoxy, tert-butoxy, and the like. “Lower haloalkoxy” refers to a lower haloalkyl as defined above joined by way of an oxygen atom (i.e., —O-(lower haloalkyl). Examples of lower haloalkoxy groups include, but are not limited to, —OCF3, —OCH2CF3, and the like. “Cycloalkyl” refers to alkyl groups forming a ring structure, which can be substituted or unsubstituted, wherein the ring is either completely saturated, partially unsaturated, or fully unsaturated, wherein if there is unsaturation, the conjugation of the pi-electrons in the ring do not give rise to aromaticity. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups. In some embodiments, the cycloalkyl group has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms range from 3 to 5, 3 to 6, or 3 to 7. Cycloalkyl groups further include polycyclic cycloalkyl groups such as, but not limited to, norbornyl, adamantyl, bornyl, camphenyl, isocamphenyl, and carenyl groups, and fused rings such as, but not limited to, decalinyl, and the like “Cycloalkylalkyl” are alkyl groups as defined above in which a hydrogen or carbon bond of the alkyl group is replaced with a bond to a cycloalkyl group as defined above. “Aryl” groups are cyclic aromatic hydrocarbons that do not contain heteroatoms. Thus aryl groups include, but are not limited to, phenyl, azulenyl, heptalenyl, biphenyl, indacenyl, fluorenyl, phenanthrenyl, triphenylenyl, pyrenyl, naphthacenyl, chrysenyl, biphenylenyl, anthracenyl, and naphthyl groups. In some embodiments, aryl groups contain 6-14 carbons in the ring portions of the groups. The phrase “aryl groups” includes groups containing fused rings, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like). In one embodiment, aryl is phenyl or naphthyl, and in another embodiment aryl is phenyl. “Heterocyclyl” or “heterocyclic” refers to aromatic and non-aromatic ring moieties containing 3 or more ring members, of which one or more is a heteroatom such as, but not limited to, N, O, S, or P. In some embodiments, heterocyclyl include 3 to 20 ring members, whereas other such groups have 3 to 15 ring members. At least one ring contains a heteroatom, but every ring in a polycyclic system need not contain a heteroatom. For example, a dioxolanyl ring and a benzdioxolanyl ring system (methylenedioxyphenyl ring system) are both heterocyclyl groups within the meaning herein. Heterocyclyl groups also include fused ring species including those having fused aromatic and non-aromatic groups. A heterocyclyl group also includes polycyclic ring systems containing a heteroatom such as, but not limited to, quinuclidyl, and also includes heterocyclyl groups that have substituents, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring members. A heterocyclyl group as defined herein can be a heteroaryl group or a partially or completely saturated cyclic group including at least one ring heteroatom. Heterocyclyl groups include, but are not limited to, pyrrolidinyl, furanyl, tetrahydrofuranyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, dihydrobenzofuranyl, indolyl, dihydroindolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl groups. “Heteroaryl” refers to aromatic ring moieties containing 5 or more ring members, of which, one or more is a heteroatom such as, but not limited to, N, O, and S. Heteroaryl groups include, but are not limited to, groups such as pyrrolyl, pyrazolyl, pyridinyl, pyridazinyl, pyrimidyl, pyrazyl, pyrazinyl, pyrimidinyl, thienyl, triazolyl, tetrazolyl, triazinyl, thiazolyl, thiophenyl, oxazolyl, isoxazolyl, benzothiophenyl, benzofuranyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, and quinazolinyl groups. “Isomer” is used herein to encompass all chiral, diastereomeric or racemic forms of a structure, unless a particular stereochemistry or isomeric form is specifically indicated. Such compounds can be enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions, at any degree of enrichment. Both racemic and diastereomeric mixtures, as well as the individual optical isomers can be synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these are all within the scope of certain embodiments of the invention. The isomers resulting from the presence of a chiral center comprise a pair of non-superimposable isomers that are called “enantiomers.” Single enantiomers of a pure compound are optically active (i.e., they are capable of rotating the plane of plane polarized light and designated R or S). “Spiroalkyl” refers to a geminally substituted di-lower alkyl substituents of 1 to 3 carbon atoms that form a continuous ring of 3 to 7 carbon atoms, respectively. The number of carbon atoms of the bonded geminal substituents are independent of each other, e.g. one germinal substituent may be 2 carbon atoms and the second germinal substituent may be 3 carbon atoms, with terminal carbons bonded to make a fused spiroalkyl of 6 carbon atoms, counting the geminally substituted carbon atom. “Isolated optical isomer” means a compound which has been substantially purified from the corresponding optical isomer(s) of the same formula. For example, the isolated isomer may be at least about 80%, at least 80% or at least 85% pure. In other embodiments, the isolated isomer is at least 90% pure or at least 98% pure, or at least 99% pure by weight. “Substantially enantiomerically or diasteromerically” pure means a level of enantiomeric or diasteromeric enrichment of one enantiomer with respect to the other enantiomer or diasteromer of at least about 80%, and more specifically in excess of 80%, 85%, 90%, 95%, 98%, 99%, 99.5% or 99.9%. The terms “racemate” and “racemic mixture” refer to an equal mixture of two enantiomers. A racemate is labeled “(±)” because it is not optically active (i.e., will not rotate plane-polarized light in either direction since its constituent enantiomers cancel each other out). A “hydrate” is a compound that exists in combination with water molecules. The combination can include water in stoichiometric quantities, such as a monohydrate or a dehydrate, or can include water in random amounts. As the term is used herein a “hydrate” refers to a solid form; that is, a compound in a water solution, while it may be hydrated, is not a hydrate as the term is used herein. A “solvate” is similar to a hydrate except that a solvent other that water is present. For example, methanol or ethanol can form an “alcoholate”, which can again be stoichiometric or non-stoichiometric. As the term is used herein a “solvate” refers to a solid form; that is, a compound in a solvent solution, while it may be solvated, is not a solvate as the term is used herein. “Isotope” refers to atoms with the same number of protons but a different number of neutrons, and an isotope of a compound of Formula (I) includes any such compound wherein one or more atoms are replaced by an isotope of that atom. For example, carbon 12, the most common form of carbon, has six protons and six neutrons, whereas carbon 13 has six protons and seven neutrons, and carbon 14 has six protons and eight neutrons. Hydrogen has two stable isotopes, deuterium (one proton and one neutron) and tritium (one proton and two neutrons). While fluorine has a number of isotopes, fluorine 19 is longest-lived. Thus, an isotope of a compound having the structure of Formula (I) includes, but not limited to, compounds of Formula (I) wherein one or more carbon 12 atoms are replaced by carbon-13 and/or carbon-14 atoms, wherein one or more hydrogen atoms are replaced with deuterium and/or tritium, and/or wherein one or more fluorine atoms are replaced by fluorine-19. “Salt” generally refers to an organic compound, such as a carboxylic acid or an amine, in ionic form, in combination with a counter ion. For example, salts formed between acids in their anionic form and cations are referred to as “acid addition salts”. Conversely, salts formed between bases in the cationic form and anions are referred to as “base addition salts.” Co-crystal forms of compounds having the structure of Formula (I) are also included within the scope of this invention; namely, solids that are crystalline single phase materials composed of two or more different molecular and/or ionic compounds generally in a stoichiometric ratio which are neither solvates nor simple salts. The term “pharmaceutically acceptable” refers an agent that has been approved for human consumption and is generally non-toxic. For example, the term “pharmaceutically acceptable salt” refers to nontoxic inorganic or organic acid and/or base addition salts (see, e.g., Lit et al., Salt Selection for Basic Drugs,Int J. Pharm.,33, 201-217, 1986) (incorporated by reference herein). Pharmaceutically acceptable base addition salts of compounds of the invention include, for example, metallic salts including alkali metal, alkaline earth metal, and transition metal salts such as, for example, calcium, magnesium, potassium, sodium, and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N,N-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine. Pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, aromatic aliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, hippuric, malonic, oxalic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, panthothenic, trifluoromethanesulfonic, 2-hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, β-hydroxybutyric, salicylic, galactaric, and galacturonic acid. Although pharmaceutically unacceptable salts are not generally useful as medicaments, such salts may be useful, for example as intermediates in the synthesis of compounds having the structure of Formula I, for example in their purification by recrystallization. In one embodiment, compounds are provided having the structure of Formula (I), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein represents an optional double bond; A and B are independently nitrogen or oxygen, with the proviso that A and B are not both oxygen; G is nitrogen or carbon; X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl, lower alkoxyalkyl, cycloalkyl, heterocyclyl, lower haloalkyl, or —C(═O)R3; R1band R1care independently hydrogen or lower alkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, or cycloalkyl-R6; Q is aryl or heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower alkoxy, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In one embodiment, compounds are provided having the structure of Formula (I-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein represents an optional double bond; A and B are independently nitrogen or oxygen, with the proviso that A and B are not both oxygen; G is nitrogen or carbon; X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1band R1care independently hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In one embodiment, compounds are provided having the structure of Formula (I-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein represents an optional double bond; A and B are independently nitrogen or oxygen, with the proviso that A and B are not both oxygen; G is nitrogen or carbon; X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1band R1care independently hydrogen or lower alkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, R1cis hydrogen and compounds are provided having the structure of Formula (II), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof: wherein represents an optional double bond; A and B are independently nitrogen or oxygen, with the proviso that A and B are not both oxygen; G is nitrogen or carbon; X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring Q is aryl or heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, lower haloalkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (II-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein represents an optional double bond; A and B are independently nitrogen or oxygen, with the proviso that A and B are not both oxygen; G is nitrogen or carbon; X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (II-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein represents an optional double bond; A and B are independently nitrogen or oxygen, with the proviso that A and B are not both oxygen; G is nitrogen or carbon; X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (III), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (III-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In another embodiment, compounds are provided having the structure of Formula (III-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (IV), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (IV-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (IV-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (V), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; Q1, Q2, Q3, Q4, and Q5are independently N, CH, or CR4; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (V-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; Q1, Q2, Q3, Q4, and Q5are independently N, CH, or CR4; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (V-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; Q1, Q2, Q3, Q4, and Q5are independently N, CH, or CR4; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VI-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VI-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VII), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VII-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VIII), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VIII-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (VIII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (IX), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (IX-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (IX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (X), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (X-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (X-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and In a more specific embodiment, compounds are provided having the structure of Formula (XI-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XI-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In another embodiment, compounds are provided having the structure of Formula (XII), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XII-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, heterocyclyl, lower alkoxy, or —O-heterocyclyl R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XIII), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XIII-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XIII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R2is -Q-(R4)n, —S(═O)2R5, or —C(═O)R5; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XIV), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XIV-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XIV-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; Q is heteroaryl; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XV), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; Q1, Q2, Q3, Q4, and Q5are independently N, CH, or CR4; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XV-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; Q1, Q2, Q3, Q4, and Q5are independently N, CH, or CR4; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XV-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; Q1, Q2, Q3, Q4, and Q5are independently N, CH, or CR4; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVI-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVI-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVII), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVII-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVIII), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVIII-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XVIII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XIX), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XIX-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XIX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XX), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XX-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; each R4is independently halogen, hydroxy, lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxyalkyl, or cyano; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano; and n is 0, 1, or 2. In a more specific embodiment, compounds are provided having the structure of Formula (XXI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XXI-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XXI-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XXII), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, -alkyl-C(═O)R3, or —C(═O)R3; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R1or R1band R3, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XXII-A), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1is hydrogen, lower alkyl-R6, haloalkyl, lower alkoxyalkyl, -cycloalkyl-R6, -alkyl-cycloalkyl-R6, -aryl-R6, -alkyl-aryl-R6, -heterocyclyl-R6, -alkyl-heterocyclyl-R6, lower haloalkyl, or -alkyl-C(═O)R3; R1bis hydrogen or lower alkyl; or R1band R1, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In a more specific embodiment, compounds are provided having the structure of Formula (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof: wherein X is halogen, lower alkyl, lower haloalkyl, lower alkoxy, or cyano; R1bis hydrogen or lower alkyl; R3is lower alkyl, lower haloalkyl, lower alkoxy, lower haloalkoxy, -cycloalkyl-R6, —O-cycloalkyl-R6, or —O-heterocyclyl-R6; or R1band R3, together with the atoms to which they are attached, form a ring; R5is cycloalkyl, lower alkyl, lower haloalkyl, cycloalkylalkyl, lower alkoxy, heterocyclyl, or —O-heterocyclyl; and R6is hydrogen, halo, alkyl, lower haloalkyl, or cyano. In the following more specific embodiments, the various substituents (e.g., A, B, G, R1, R1b, R1c, R2and X) are set forth in more detail with respect to the compounds of each of Formulas (I) through (XXII-B) above, as applicable to the substituents being further defined. For example, reference to R1below is intended to further limit the compounds of Formulas (I), (I-A), (II), (II-A), (III), (III-A), (IV), (IV-A), (V), (V-A), (VI), (VI-A), (VII), (VII-A), (VIII), (VIII-A), (IX), (IX-A), (X), (X-A), (XI), (XI-A), (XII), (XII-A), (XIII), (XIII-A), (XIV), (XIV-A), (XV), (XV-A), (XVI), (XVI-A), (XVII), (XVII-A), (XVIII), (XVIII-A), (XIX), (XIX-A), (XX), (XX-A), (XXI), (XXI-A), (XXII), and (XXII-A) above, but not Formulas (I-B), (II-B), (III-B), (IV-B), (V-B), (VI-B), (VII-B), (VIII-B), (IX-B), (X-B), (XI-B), (XII-B), (XIII-B), (XIV-B), (XV-B), (XVI-B), (XVII-B), (XVIII-B), (XIX-B), (XX-B), (XXI), (XXI-B), (XXII), and (XXII-B) since the R1substituent has already been further defined in the same. Thus, reference to the substituents below is intended to further modify Formulas (I) through (XXII-B) to the extent such formulas recite that particular substituent as a variable. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is halogen. In more specific embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is Cl, F, or Br. In more specific embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is Cl. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is lower alkyl. In more specific embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is methyl, ethyl, or isopropyl. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is lower haloalkyl. In more specific embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is —CF3. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is lower alkoxy. In more specific embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is methoxy, ethoxy, isopropoxy, or t-butoxy. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein X is cyano. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1is hydrogen. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1is lower alkyl. In more specific embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1is methyl, ethyl, or isopropyl. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1is lower alkoxy. In more specific embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1is methoxy, ethoxy, isopropoxy, or t-butoxy. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1is cycloalkyl. In more specific embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1is cyclopropyl or cyclobutyl. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R2is -Q-(R4)n. In further embodiments, n is 0. In further embodiments, n is 1, 2, or 3. In further embodiments, n is 1. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is -Q-(R4)nand R4is halogen. In more specific embodiments, compounds are provided wherein R4is F or Cl. In further embodiments compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is -Q-(R4)nand R4is lower alkyl. In more specific embodiments, compounds are provided wherein R4is methyl or ethyl. In further embodiments compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is -Q-(R4)nand R4is lower alkoxy. In more specific embodiments, compounds are provided wherein R4is methoxy or ethoxy. In further embodiments compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is -Q-(R4)nand R4is cyano. In further embodiments compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is -Q-(R4)nand R4is hydroxy. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is —S(═O)2R5or —C(═O)R5and R5is lower alkyl. In more specific embodiments, compounds are provided wherein R5is methyl, ethyl, or isopropyl. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is —S(═O)2R5or —C(═O)R5and R5is lower alkoxy. In more specific embodiments, compounds are provided wherein R5is t-butoxy. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is —S(═O)2R5or —C(═O)R5and R5is heterocyclyl or cycloalkyl. In more specific embodiments, compounds are provided wherein R5is: In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is —S(═O)2R5or —C(═O)R5and R5is —O-heterocyclyl. In more specific embodiments, compounds are provided wherein R5is: In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is —S(═O)2R5or —C(═O)R5and R5is heteroaryl. In more specific embodiments, compounds are provided wherein R5is pyridinyl. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R2is —S(═O)2R5or —C(═O)R5and R5is cycloalkylalkyl. In more specific embodiments, compounds are provided wherein R5is: In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R1is alkyl, cycloalkyl, lower haloalkyl; R4is H, halo, lower alkyl, lower haloalkyl, lower alkoxy, lower alkoxyalkyl, lower haloalkoxyalkyl, cyano; and X is halo or lower alkoxy. In further embodiments, R1is methyl, cyclopropyl, or CF3, R4is H, F, methyl, —CF3, —O—CH3, or —O-isopropyl; and X is Cl or —O—CH3. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R3is alkyl, cycloalkyl, lower haloalkyl; R4is H, halo, lower alkyl, lower haloalkyl, lower alkoxyalkyl, lower alkoxhaloyalkyl, cyano; and X is halo, lower alkoxy. In further embodiments, R3is methyl, cyclopropyl, or CF3, R4is H, F, methyl, —CF3, —O—CH3, or —O-isopropyl; and X is Cl or —O—CH3. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R1is alkyl, haloalkyl, cycloalkyl; R5is cycloalkyl, lower alkyl, heterocyclyl and X is halo. In further embodiments, R1is cyclopropyl or —CH2—CF2-cyclopropyl. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R3is alkyl, cycloalkyl, lower haloalkyl; R5is cycloalkyl, lower alkyl, heterocyclyl and X is halo. In further embodiments, R3is methyl or cyclopropyl. In further embodiments, compounds are provided having the structure of any one of Formulas (I) through (XXII-B) wherein R1is Representative compounds of Formula (I), and Formulas (II) through (XXII-B) as applicable, include the compounds listed in Table 1 below, as well as pharmaceutically acceptable isomers, racemates, hydrates, solvates, isotopes, and salts thereof. TABLE 1Representative CompoundsCmpd.No.Structure1234567891011121314151617181920212223242526272829303132333435363738394041424344454647484950515253545556575859606162636465666768697071727374757677787980818283848586878889909192939495969798991001011021031041051061071081091101111121131141151161171181191201211221231241251261271281291301311321331341351361371381391401411421431441451461471481491501511521531541551561571581591601611621631641651661671681691701711721731741751761771781791801811821831841851861871881891901911921931941951961971981992002012022032042052062072082092102112122132142152162172182192202212222232242252262272282292302312322332342352362372382392402412422432442452462472482492502512522532542552562572582592602612622632642652662672682692702712722732742752762772782792802812822832842852862872882892902912922932942952962972982993003013023033043053063073083093103113123133143153163173183193203213223233243253263273283293303313323333343353363373383393403413423433443453463473483493503513523533543553563573583593603613623633643653663673683693703713723733743753763773783793803813823833843853863873883893903913923933943953963973983994004014024034044054064074084094104114124134144154164174184194204214224234244254264274284294304314324334344354364374384394404414424434444454464474484494504514524534544554564574584594604614624634644654664674684694704714724734744754764774784794804814824834844854864874884894904914924934944954964974984995005015025035045055065075085095105115125135145155165175185195205215225235245255265275285295305315325335345355365375385395405415425435445455465475485495505525535545555565575585595605615625635645655665675685695705715725735745755765775785795805815825835845855865875885895905915925935945955965975985996006016026036046056066076086096106116126136146156166176186196206216226236246256266276286296306316326336346356366376386396406416426436446456466476486496506516526536546556566576586596606616626636646656676686696706716726736746756766776786796806816826836846856866876886896906916926936946956966976986997007017027027037047057067077087097107117127137147157167177187197207217227237247257267277287297307317327337347357367377387397407417427437447457467477487497507517527537547557567577587597607617627637647657667677687697707717727737747757767777787797807817827837847857867877887897907917927937947957967977987998008018028038048058068078088098108118128138148158168178188198208218228238248258268278288298308318328338348358368378388398408418428438448458468478488498508518528538548558568578588598608618628638648658668678688698708718728738748758768778788798808818828838848858868878888898908918928938948958968978988999009019029039049059069079089099109119129139149159169179189199209219229239249259269279289299309319329339349359369379389399409419429439449459469479489499509519529539549559569579589599609619629639649659669679689699709719729739749759769779789799809819829839849859869879889899909919929939949959969979989991000100110021003100410051006100710081009101010111012101310141015101610171018101910201021102210231024102510261027102810291030103110321033103410351036103710381039104010411042104310441045104610471048104910501051105210531054105510561057105810591060106110621063106410651066106710681069107010711072 In certain embodiments, the invention provides a pharmaceutical composition comprising a compound of the invention together with at least one pharmaceutically acceptable carrier, diluent, or excipient. For example, the active compound will usually be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier which can be in the form of an ampoule, capsule, sachet, paper, or other container. When the active compound is mixed with a carrier, or when the carrier serves as a diluent, it can be solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active compound. The active compound can be adsorbed on a granular solid carrier, for example contained in a sachet. Some examples of suitable carriers are water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid, or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose, and polyvinylpyrrolidone. Similarly, the carrier or diluent can include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax. The formulations can be mixed with auxiliary agents which do not deleteriously react with the active compounds. Such additives can include wetting agents, emulsifying and suspending agents, salt for influencing osmotic pressure, buffers and/or coloring substances, preserving agents, sweetening agents, or flavoring agents. The compositions can also be sterilized if desired. The route of administration can be any route which effectively transports the active compound of the invention to the appropriate or desired site of action, such as oral, nasal, pulmonary, buccal, subdermal, intradermal, transdermal, or parenteral, e.g., rectal, depot, subcutaneous, intravenous, intraurethral, intramuscular, intranasal, ophthalmic solution, or an ointment, the oral route being preferred. For parenteral administration, the carrier will typically comprise sterile water, although other ingredients that aid solubility or serve as preservatives can also be included. Furthermore, injectable suspensions can also be prepared, in which case appropriate liquid carriers, suspending agents, and the like can be employed. For topical administration, the compounds of the present invention can be formulated using bland, moisturizing bases such as ointments or creams. If a solid carrier is used for oral administration, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form or it can be in the form of a troche or lozenge. If a liquid carrier is used, the preparation can be in the form of a syrup, emulsion, soft gelatin capsule, or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution. Injectable dosage forms generally include aqueous suspensions or oil suspensions which can be prepared using a suitable dispersant or wetting agent and a suspending agent Injectable forms can be in solution phase or in the form of a suspension, which is prepared with a solvent or diluent. Acceptable solvents or vehicles include sterilized water, Ringer's solution, or an isotonic aqueous saline solution. Alternatively, sterile oils can be employed as solvents or suspending agents. Preferably, the oil or fatty acid is non-volatile, including natural or synthetic oils, fatty acids, mono-, di-, or tri-glycerides. For injection, the formulation can also be a powder suitable for reconstitution with an appropriate solution as described above. Examples of these include, but are not limited to, freeze dried, rotary dried, or spray dried powders, amorphous powders, granules, precipitates, or particulates. For injection, the formulations can optionally contain stabilizers, pH modifiers, surfactants, bioavailability modifiers, and combinations of these. The compounds can be formulated for parenteral administration by injection such as by bolus injection or continuous infusion. A unit dosage form for injection can be in ampoules or in multi-dose containers. The formulations of the invention can be designed to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art. Thus, the formulations can also be formulated for controlled release or for slow release. Compositions contemplated by the present invention can include, for example, micelles or liposomes, or some other encapsulated form, or can be administered in an extended release form to provide a prolonged storage and/or delivery effect. Therefore, the formulations can be compressed into pellets or cylinders and implanted intramuscularly or subcutaneously as depot injections. Such implants can employ known inert materials such as silicones and biodegradable polymers, e.g., polylactide-polyglycolide. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). For nasal administration, the preparation can contain a compound of the invention, dissolved or suspended in a liquid carrier, preferably an aqueous carrier, for aerosol application. The carrier can contain additives such as solubilizing agents, e.g., propylene glycol, surfactants, absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin, or preservatives such as parabens. For parenteral application, particularly suitable are injectable solutions or suspensions, preferably aqueous solutions with the active compound dissolved in polyhydroxylated castor oil. Dosage forms can be administered once a day, or more than once a day, such as twice or thrice daily. Alternatively, dosage forms can be administered less frequently than daily, such as every other day, or weekly, if found to be advisable by a prescribing physician. Dosing regimens include, for example, dose titration to the extent necessary or useful for the indication to be treated, thus allowing the patient's body to adapt to the treatment and/or to minimize or avoid unwanted side effects associated with the treatment. Other dosage forms include delayed or controlled-release forms. Suitable dosage regimens and/or forms include those set out, for example, in the latest edition of thePhysicians' Desk Reference, incorporated herein by reference. When used to prevent the onset of a malcondition, the compounds provided herein will be administered to a subject at risk for developing the same, typically on the advice and under the supervision of a physician, at the dosage levels described above. Subjects at risk for developing a particular malcondition generally include those that have a family history of the same, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the malcondition. Chronic administration refers to administration of a compound or pharmaceutical composition thereof over an extended period of time, e.g., for example, over 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may be continued indefinitely, for example, for the rest of the subject's life. In certain embodiments, the chronic administration is intended to provide a constant level of the compound in the blood, e.g., within the therapeutic window over the extended period of time. In another embodiment, there are provided methods of making a composition of a compound described herein including formulating a compound of the invention with a pharmaceutically acceptable carrier or diluent. In some embodiments, the pharmaceutically acceptable carrier or diluent is suitable for oral administration. In some such embodiments, the methods can further include the step of formulating the composition into a tablet or capsule. In other embodiments, the pharmaceutically acceptable carrier or diluent is suitable for parenteral administration. In some such embodiments, the methods further include the step of lyophilizing the composition to form a lyophilized preparation. In another embodiment, a method is provided for antagonizing the V1a receptor, the method comprising contacting the receptor with an effective amount of a compound having the structure of Formula (I) through (XX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, or a pharmaceutical composition comprising the same. The term “antagonism” is used herein to encompass molecules that interact in some way with a receptor and thereby function as an antagonist, either by binding to the receptor at the binding site of its natural ligand or at locations other than the binding site. The phrase to “V1a antagonism” is used herein to encompass molecules that interact in some way with the V1a receptor and thereby function as an antagonist, either by binding to the Via receptor at the binding site of its natural ligand, or at a location other than the binding site (i.e., allosteric binding). In an embodiment, a method is provided for treatment of a malcondition in a subject for which antagonism of the V1a receptor is medically indicated. Such method comprises administering to the subject an effective amount of a compound having the structure of Formula (I) through (XX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for duration sufficient to provide a beneficial effect to the subject. As used herein, a “subject” means both mammals and non-mammals. Mammals include, for example: humans; non-human primates (e.g., apes and monkeys); cattle; horses; sheep; and goats. Non-mammals include, for example, fish and birds. “Treating” or “treatment” within the meaning herein refers to an alleviation of symptoms associated with a malcondition, or inhibition of further progression or worsening of those symptoms, or prevention or prophylaxis of the malcondition in certain instances. The expression “effective amount”, when used to describe use of a compound for treating a subject suffering from a malcondition for which antagonism of the V1a receptor is medically indicated, refers to an the amount of the compound sufficient to produce a beneficial therapeutic effect for the subject. The phrase “malcondition” is intended to broadly encompass any and all diseases, disorders, syndromes and/or symptoms wherein the V1a receptor plays a role in the same, such that a therapeutically beneficial effect can be achieved by antagonism of the Via receptor. In certain embodiments, the present invention provides a method for antagonizing the V1a receptor with a compound of Formula (I) through (XX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, by contacting the receptor with a suitable amount of the compound to antagonize the receptor. Such contacting can take place in vitro, for example in carrying out an assay to determine the V1a inhibition activity of a compound undergoing experimentation related to a submission for regulatory approval. In certain embodiments, the method for antagonizing the V1a receptor can also be carried out in vivo, that is, within the living body of the subject. The compound of Formula (I) through (XX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, can be supplied to the living organism via one of the routes as described above (e.g., orally) or can be provided locally within the body tissues. In the presence of the inventive compound, inhibition of the receptor takes place, and the effect thereof can be studied. In another embodiment, a compound of Formula (I) through (XX-B) is an imaging agent, wherein the compound contains an isotope, such as isotopes of F, O, N and C. In certain embodiments, the isotope is a fluorine isotope. The compounds may be used for therapeutic purposes, or to diagnose or assess the progression of a malcondition (a vasopressin-dependent condition) in a subject for which antagonism of the V1a receptor is medically indicated. In some embodiments, imaging and/or diagnostic methods are provided comprising administering to a subject in need thereof the imaging agent described herein and detecting the compound comprised in the imaging agent in the subject. In some aspects, the amount of the compound in the subject is quantified. In further aspects, a vasopressin-dependent condition in the subject is detected via a detection of the compound in the subject. In certain embodiments, the imaging is effected by a radiodiagnostic method. The radiodiagnostic method may be performed by any instrument capable of detecting radiation by the compounds. Exemplary radiodiagnostic methods include, but not limited to, Positron Emission Tomography (PET), PET-Time-Activity Curve (TAC) or PET-Magnetic Resonance Imaging (MRI). In particular aspect, the radiodiagnostic method is PET. In one embodiment, methods of treatment are provided comprising administering a compound of Formula (I) through (XX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, alone or in combination with another pharmacologically active agent or second medicament, to a subject having a malcondition for which antagonizing the V1a receptor is medically indicated. As mentioned above, V1a receptor antagonists provide significant promise for the treatment of malconditions which benefit from antagonism of the V1a receptor. As summarized in the review article by Szczepanska-Sadowaska et al.,Current Drug Metabolism18:306-345, 2017 (incorporated by reference herein in its entirety), vasopressin has been associated with a wide range of regulatory functions in numerous organs and/or tissues and implicated in or with: (1) the cardiovascular system, (2) renal effects, (3) circadian rhythm, (4) food intake and metabolic and endocrine regulation, (5) uterus, (6) endotoxemia, and (7) stress, depression and psychiatric disorders. AVP is also involved in the regulation of several functions, such as, hepatic, pancreatic, and platelet-aggregating effects, and effects on the central and peripheral nervous system. The effects of the AVP receptors depends on where they are located. In the cardiovascular system, vasopressin is associated with: (a) peripheral effects (e.g., it acts as a potent vasoconstrictor and plays a role in the regulation of carioca muscle differentiation, growth and contractility); (b) central cardiovascular control (e.g., buffering excessive increases and decreases in blood pressure); (c) regulation of cardiovascular reflexes (e.g., in the regulation of the baroreceptor reflex); (d) interaction with other factors (e.g., factors regulating blood pressure such as Ang II); (e) adaption to hemorrhage; and (f) cardiovascular diseases (e.g., hypertension and heart failure, intracranial hemorrhage and stroke). As for renal effects, vasopressin has antidiuretic action, and interacts with AngII in the regulation of urine excretion. Vasopressin also exerts a diposgenic action, manifested by reduction of the osmotic thirst threshold. In the context of circadian rhythm, vasopressin neurons in the suprachiasmatic nuclei (SCN) of the hypothalamus manifest a distinct circadian rhythmicity, and studies have shown distinct circadian rhythmicity of vasopressin concentration in the cerebrospinal fluid (CSF). It has been suggested that SCN vasopressin neurons belong to the group of autonomous pacemakers and play a role in the regulation of the circadian rhythm, and studies have shown that circadian rhythmicity of vasopressin release has repercussions in the diuranl rhythmicity of other functions, such as corticosterone release, locomotor activity and body temperature. With regard to food intake and metabolic and endocrine regulation, vasopressin has been associated with regulation of food intake and glucose homeostasis, and animal studies with V1a receptor knockout mice consuming high fat diet show that vasopressin acting on V1a receptor improves glucose tolerance and protects from the development of obesity. Studies have also shown that vasopressin plays a direct role in the regulation of glucagon and insulin release from the pancreatic cells. In the adrenal gland, vasopressin causes hypertrophy and hyperplasia of the adrenal cortex and stimulates secretion of aldosterone and glucocorticoids through stimulation of V1a receptors. Stimulation of the Via receptor by vasopressin also influences release of luteinizing hormone releasing hormone (LHRL) and is believed to play a role in initiating the preovulatory LH surge. The presence of V1a receptors has also been reported in the uterus, with the density of such receptors higher in the myometrium than in the endometrium, and they react with oxytocin (OT) receptors. Endotoxemia is associated with the increased expression of the vasopressin gene in the hypothalamic nuclei and elevated concentration of vasopressin in the blood. Vasopressin exerts various effects on the cardiovascular system during endotoxemia, including reducing renal medullary blood flow where aortic contractility is reduced. There is also evidence that vasopressin plays a role in the regulation of immunologic processes, and that it may play a role in the regeneration of the liver. With regard to stress, depression and psychiatric disorders, the role of vasopressin in the regulation of behavior has been studied for many decades, with early studies showing that it facilitates conditioned avoidance responses in rats. Experimental studies have shown that vasopressin has long-lasting effects on learning and new memory acquisition as well as emotional and social behaviors, and clinical observations have shown that depression and other psychiatric disorders are associated with significant changes in vasopressin secretion. Neurogenic stress has also been shown to stimulate vasopressin release in the blood and CSF. A strong association has been shown between chronic stress, inappropriate activation of the vasopressinergic system and depression. Studies in humans have shown that patients with major depression manifested an elevated plasma vasopressin level, and in patients with unipolar depression there was a significant positive correlation between peripheral plasma vasopressin and hypercortisolemia. There is also evidence that vasopressin is an anxiogenic agent, and direct administration of V1a receptor antagonist into the paraventricular nucleus (PVN) of rats attenuated anxiety and depression behavior. Aggression has also been associated with an increased release of vasopression into the CSF. Vasopressin plays a role in the regulation of pain, and its antinociceptive action has been shown in a number of studies. Inappropriate secretion of vasopressin has also been suggested in the disordered processing of psychosomatic stress which occurs in schizophrenia. Due to its wide and pivotal role for maintaining body homeostatis under a variety of conditions, vasopressin and its receptors, including V1a, have been recognized as an important target for diagnostic and therapeutic applications. To this end, vasopressin antagonists have shown efficacy in easing congestion symptoms and edema and increasing plasma sodium ion concentration in clinical trials. In addition, the compounds of the present invention have utility across a broad spectrum of malconditions, including the following: heart failure, hepatic cirrhosis, psychiatric disorders (e.g., major depressive disorder or generalized anxiety disorder), brain injury, circadian rhythm disorders (e.g., associated with shift work or jet lag, resulting in sleep drifting later each day, abnormal night sleep patterns, and/or difficulty staying awake during the day), bone growth, diabetes mellitus, ovarian function, septic shock (e.g. maintaining haemodynamic parameters and preventing organ damage), and cancer and metastisis (e.g., decreasing dissemination of tumor cells and the spread of metasteses by improving haemostasis and slowing of proliferation of carcinoma cells). The compounds of the present invention selectively block the effects of V1a receptors, are orally bioavailable/effective, and demonstrate central nervous system (CNS)-penetrating effects. These compounds, (when acting peripherally and/or centrally) are useful in the treatment of vasopressin-dependent conditions or in the conditions related to inappropriate secretion of vasopressin, particularly in the response to chronic stress and in circuits that are dysregulated in affective disorders. These compounds reduce measures of stress, fear, aggression, depression, and anxiety. In an embodiment, a method is provided for treatment or prevention of vasopressin-dependent conditions or in the conditions related to inappropriate secretion of vasopressin, comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for a duration sufficient to provide a beneficial effect to the subject. In an embodiment, a method is provided for treatment of a vasopressin-dependent condition, whether organic, stress-induced or iatrogenic. As used herein a “vasopressin-dependent condition” is defined as conditions related to inappropriate secretion of vasopressin, particularly in the response to chronic stress and in circuits that are dysregulated in affective disorders, such as disorders of stress, mood, and behavioral disorders, including stress-related affective disorders. Vasopressin-dependent conditions, include conditions such as cardiovascular conditions, for example hypertension, pulmonary hypertension, cardiac insufficiency, myocardial infarction or coronary vasospasm, in particular in smokers, Raynaud's syndrome, unstable angina and PTCA (percutaneous transluminal coronary angioplasty), cardiac ischemia, hemostasis disturbances or thrombosis; conditions of the central nervous system, such as migraine, cerebral vasospasm, cerebral hemorrhage, trauma and cerebral edema, depression, anxiety, stress, emotional disorders, obsessive-compulsive disorder, panic attacks, psychotic states, aggression, memory or sleep disorders, or cognitive disorders, for example disorders associated with impaired social cognition (e.g., schizophrenia, autism spectrum disorder); conditions of the renal system, such as renal vasospasm, necrosis of the renal cortex, nephrogenic diabetes insipidus or diabetic nephropathy; or conditions of the gastric system, such as gastric vasospasm, cirrhosis of the liver, ulcers or the pathology of vomiting, for example nausea, including nausea due to chemotherapy, or travel sickness; circadian rhythm-related disorders such as phase shift sleep disorders, jet-lag, sleep disorders and other chronobiological disorders. Additional examples of vasopressin-dependent conditions include but are not limited to neuropsychiatric disorders, neuropsychiatric symptoms in neurodegenerative diseases, PTSD, inappropriate aggression, anxiety, depressive disorders, major depression, obsessive compulsive disorder, autistic spectrum disorders, schizophrenia, and aggressive behavior, and other affective disorders. In an embodiment, a method is provided for treatment of an autism spectrum disorder, comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for a duration sufficient to provide a beneficial effect to the subject. Autism spectrum disorder (ASD), also referred to herein as autistic spectrum disorders, is a blanket term describing a complex developmental disorder that affects the brain's normal development of social and communication skills. Core symptoms of ASD include impaired social interactions such as social interaction difficulties, communication challenges including impaired verbal and nonverbal communication, problems processing information from the senses, and a tendency to engage in restricted and repetitive patterns of behavior. In one embodiment, the core symptoms of the autism spectrum disorder are impaired social interactions and communication challenges. In one embodiment, the core symptom of the autism spectrum disorder is impaired social interactions. In one embodiment, the core symptom of the autism spectrum disorder is impaired communication challenges. In one embodiment, the core symptom of the autism spectrum disorder is the tendency to engage in restricted and repetitive patterns of behavior. In an embodiment, a method is provided for treatment of an anxiety disorder, comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XX-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for a duration sufficient to provide a beneficial effect to the subject. Anxiety disorder is a blanket term covering several different forms of abnormal and pathological fear and anxiety. Current psychiatric diagnostic criteria recognize a wide variety of anxiety disorders, including generalized anxiety disorder, panic disorder, stress-related disorders, obsessive compulsive disorder, phobia, social anxiety disorder, separation anxiety disorder and post-traumatic stress disorder (PTSD). In one embodiment, the anxiety disorder is a social anxiety disorder. In one embodiment, the anxiety disorder is phobia. In one embodiment, the anxiety disorder is a stress-related disorder. In one embodiment, the anxiety related disorder is PTSD. Generalized anxiety disorder is a common chronic disorder characterized by long-lasting anxiety that is not focused on any one object or situation. A person suffering from generalized anxiety experience non-specific persistent fear and worry and become overly concerned with everyday matters. Generalized anxiety disorder is the most common anxiety disorder to affect older adults. In panic disorder, a person suffers from brief attacks of intense terror and apprehension, often marked by trembling, shaking, confusion, dizziness, nausea, difficulty breathing. These panic attacks, defined by the APA as fear or discomfort that abruptly arises and peaks in less than ten minutes, can last for several hours and can be triggered by stress, fear, or even exercise; although the specific cause is not always apparent. In addition to recurrent unexpected panic attacks, a diagnosis of panic disorder also requires that said attacks have chronic consequences: either worry over the attack's potential implications, persistent fear of future attacks, or significant changes in behavior related to the attacks. Accordingly, those suffering from panic disorder experience symptoms even outside of specific panic episodes. Often, normal changes in heartbeat are noticed by a panic sufferer, leading them to think something is wrong with their heart or they are about to have another panic attack. In some cases, a heightened awareness (hypervigilance) of body functioning occurs during panic attacks, wherein any perceived physiological change is interpreted as a possible life threatening illness (i.e. extreme hypochondriasis). Obsessive compulsive disorder is a type of anxiety disorder primarily characterized by repetitive obsessions (distressing, persistent, and intrusive thoughts or images) and compulsions (urges to perform specific acts or rituals). The OCD thought pattern may be likened to superstitions insofar as it involves a belief in a causative relationship where, in reality, one does not exist. Often the process is entirely illogical; for example, the compulsion of walking in a certain pattern may be employed to alleviate the obsession of impending harm. And in many cases, the compulsion is entirely inexplicable, simply an urge to complete a ritual triggered by nervousness. In a minority of cases, sufferers of OCD may only experience obsessions, with no overt compulsions; a much smaller number of sufferers experience only compulsions. The single largest category of anxiety disorders is that of Phobia, which includes all cases in which fear and anxiety is triggered by a specific stimulus or situation. Sufferers typically anticipate terrifying consequences from encountering the object of their fear, which can be anything from social phobia, specific phobia, agoraphobia, phobia of an animal to a location to a bodily fluid. Post-traumatic stress disorder or PTSD is an anxiety disorder which results from a traumatic experience. Post-traumatic stress can result from an extreme situation, such as combat, rape, hostage situations, or even serious accident. It can also result from long term (chronic) exposure to a severe stressor, for example soldiers who endure individual battles but cannot cope with continuous combat. Common symptoms include flashbacks, avoidant behaviors, and depression. In an embodiment, a method is provided for treatment of a depressive disorder, depression, or depressive illness, comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for duration sufficient to provide a beneficial effect to the subject. Examples of such disorders include major depression, MDD, drug-resistant depression, dysthymia and bipolar disorder. In an embodiment, a method is provided for treatment of a mood disorder, or an affective disorder comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for duration sufficient to provide a beneficial effect to the subject. Examples of a mood disorder or a affective disorder include major depressive disorder (MDD); bipolar disorder; anhedonia; dysthymia; major depression, Psychotic major depression (PMD), or psychotic depression; postpartum depression; seasonal affective disorder (SAD); and catatonic depression is a rare and severe form of major depression involving disturbances of motor behavior and other symptoms. The terms “anhedonia” and “anhedonic symptom” are used interchangeably and is defined as the inability to experience pleasure from activities usually found enjoyable, e.g. exercise, hobbies, music, sexual activities or social interactions. The terms “anhedonia” and “anhedonic symptom” are closely related to criterion of “depressive disorder with melancholic features” which is defined in DSM-5 as melancholic depression characterized by a loss of pleasure in most or all activities, a failure of reactivity to pleasurable stimuli, a quality of depressed mood more pronounced than that of grief or loss, a worsening of symptoms in the morning hours, early morning waking, psychomotor retardation, excessive weight loss, or excessive guilt. The term “treatment of depressive disorder with melancholic features” comprises treatment of both the depressive disorder and melancholic features associated herewith. In one embodiment, the mood disorder is anhedonia. In one embodiment, the mood disorder is major depression. In one embodiment, the mood disorder is seasonal affective disorder (SAD). In an embodiment, a method is provided for treatment of an affective disorder, comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for a duration sufficient to provide a beneficial effect to the subject. Affective disorders such as disorders of stress, mood, and behavioral disorders, including stress-related affective disorders, obsessive compulsive disorder, autistic spectrum disorders, Personality disorders, ADHD, panic attacks and the like. As used herein, “autistic spectrum disorders” and “Autism spectrum disorders” are used interchangeably and refer to autism, monogenetic causes of autism such as synaptophathies, e.g., Rett syndrome, Fragile X syndrome, Angelman syndrome and the like. In an embodiment, a method is provided for treatment of Anger, Aggression or Aggressive Disorder, or Impulse Control Disorders comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for a duration sufficient to provide a beneficial effect to the subject. Examples of Anger, Aggression or Aggressive Disorder, or Impulse Control Disorders include, but are not limited to, inappropriate aggression, aggressive behavior, aggression related to social isolation, for treatment of interpersonal violence co-occurring with such illness as ADHD, autism, bipolar disorder, emotional disorders, disorders of memory and/or cognition and cognitive disorders (such as Alzheimer's disease, Parkinson's disease, Huntington's disease and the like), and addictive disorder/substance abuse. In an embodiment, a method is provided for treatment of Intermittent Explosive Disorder (sometimes abbreviated as IED) comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for a duration sufficient to provide a beneficial effect to the subject. Intermittent explosive disorder is a behavioral disorder characterized by explosive outbursts of anger and violence, often to the point of rage, that are disproportionate to the situation at hand (e.g., impulsive screaming triggered by relatively inconsequential events). Impulsive aggression is unpremeditated, and is defined by a disproportionate reaction to any provocation, real or perceived. Some individuals have reported affective changes prior to an outburst (e.g., tension, mood changes, energy changes, etc.). The disorder is currently categorized in the Diagnostic and Statistical Manual of Mental Disorders (DSM-5) under the “Disruptive, Impulse-Control, and Conduct Disorders” category. The disorder itself is not easily characterized and often exhibits comorbidity with other mood disorders, particularly bipolar disorder. Individuals diagnosed with IED report their outbursts as being brief (lasting less than an hour), with a variety of bodily symptoms (sweating, stuttering, chest tightness, twitching, palpitations) reported by a third of one sample. Aggressive acts are frequently reported accompanied by a sensation of relief and in some cases pleasure, but often followed by later remorse. In other embodiments, a method is provided for treatment of a Schizophrenia spectrum disorders, comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for duration sufficient to provide a beneficial effect to the subject. Examples of Schizophrenia spectrum disorders include schizophrenia, schizoaffective disorder, psychotic states and memory disorders. In other embodiments, a method is provided for treatment of a circadian rhythm related disorders, comprising administering to a subject in need thereof an effective amount of a compound having the structure of Formulas (I) through (XXII-B), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, or a pharmaceutical composition comprising the same, at a frequency and for duration sufficient to provide a beneficial effect to the subject. Circadian rhythm sleep disorders are caused by desynchronization or misalignment between internal sleep-wake rhythms (body clock) and the external light-darkness cycle. Circadian rhythm disorders (sometimes also referred to as phase shift disorders) include sleep disorders associated with jet lag, shift work, or altered sleep phase types, resulting in sleep drifting later each day, abnormal nigh sleep patterns, and/or difficulty staying awake during the day. The cause may be internal (e.g., delayed or advanced sleep phase syndrome, or Non-24-h sleep-wake syndrome) or external (e.g., jet lag, shift work). If the cause is external, other circadian body rhythms, including temperature and hormone secretion, can become out of sync with the light-darkness cycle (external desynchronization) and with one another (internal desynchronization); in addition to insomnia and excessive sleepiness, these alterations may cause nausea, malaise, irritability, and depression. Risk of cardiovascular and metabolic disorders may also be increased. Compounds of the invention are useful for treating circadian rhythm-related disorders, such as depression, jet-lag, work-shift syndrome, sleep disorders, glaucoma, reproduction, cancer, premenstrual syndrome, immune disorders, inflammatory articular diseases and neuroendocrine disorders, Non-24 Hour Disorder. The compounds according to the invention may also be used in the treatment or prevention of Neuropsychiatric Disorders such as anorexia nervosa, bulimia, mood disorders, depression, anxiety, sleeping disorders, addictive disorders, panic attacks, phobias, obsession, pain-perception disorders (fibromyalgia), dependency on a substance, hemorrhagic stress, muscular spasms and hypoglycemia. Addictive disorder, including disorders related to substance abuse or addiction, and compulsive behavior. The compounds according to the invention may also be used in the treatment or prevention of chronic stress states such as immunodepression, fertility disorders and dysfunctions of the hypothalamopituitaryadrenal axis. The compounds according to the invention can also be used in the treatment of disorders such as primary or secondary dysmenorrhea, premature labor or endometriosis, male or female sexual dysfunction, hypertension, chronic heart failure, inappropriate secretion of vasopressin, liver cirrhosis and nephrotic syndrome. The compounds according to the invention can also be used in the treatment or prevention of any pathology resulting from stress, such as fatigue and its syndromes, ACTH-dependent disorders, cardiac disorders, pain, modifications in gastric emptying, in fecal excretion (colitis, irritable bowel syndrome or Crohn's disease) or in acid secretion, hyperglycemia, immunosuppression, inflammatory processes (rheumatoid arthritis and osteoarthritis), multiple infections, septic shock, cancers, asthma, psoriasis and allergies. The compounds according to the invention may also be used as psychostimulants, bringing about an increase in consciousness/alertness and/or in emotional reactivity towards the environment and making adaptation easier. The compounds according to the present invention can be used in healing, in analgesia, in anxiolysis, in the prevention of pain, in the prevention of anxiety, depression, schizophrenia, autism or obsessive-compulsive syndrome, in maternal behavior (facilitation of recognition and acceptance of the mother by the child) and social behavior, memory; regulation of food and drink intake, dependence on drugs, withdrawal and sexual motivation; hypertension, hyponatremia, cardiac insufficiency, atherosclerosis, angiogenesis, the proliferation of tumors, Kaposi's sarcoma, to regulate the storage of fat by the adipocyte, to control hyperlipidemia, triglyceridemia and metabolic syndrome. The compounds according to the invention can also be used in the treatment of cancers, such as small cell lung cancers or breast cancers; hyponatremic encephalopathy; pulmonary syndrome; Meniere's disease; ocular hypertension; glaucoma; cataracts; obesity; type-I and type-II diabetes; atherosclerosis; metabolic syndrome; hyperlipidemia; insulin resistance; or hypertriglyceridemia; in post-operative treatments, in particular after abdominal surgery; autism; hypercortisolemia; hyperaldosteronemia; pheochromocytoma; Cushing's syndrome; preeclampsia; disorders of micturition; or premature ejaculation. Compounds having the structure of Formula (I), as well as the sub-structures for Formulas (II) through (XXII-B), can be synthesized using standard synthetic techniques known to those of skill in the art. For examples, compounds of the present invention can be synthesized using the general synthetic procedures set forth in Schemes 1 and 2. To this end, the reactions, processes, and synthetic methods described herein are not limited to the specific conditions described in the following experimental section, but rather are intended as a guide to one with suitable skill in this field. For example, reactions may be carried out in any suitable solvent, or other reagents to perform the transformation[s] necessary. Generally, suitable solvents are protic or aprotic solvents which are substantially non-reactive with the reactants, the intermediates or products at the temperatures at which the reactions are carried out (i.e., temperatures which may range from the freezing to boiling temperatures). A given reaction may be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction, suitable solvents for a particular work-up following the reaction may be employed. Reagents and conditions: i) iPrOH; ii) TFA; iii) MIP-carbonate, MeOH; iv) R2Br, RuPhos, Pd(OAc)2, Cs2CO3dioxane; v) R2CI, Cs2CO3, DMF. Reagents and conditions: i) TFA, DCM; ii) dioxane; iii) R1Cl; iv) TFA, DCM; v) R2Br, RuPhos, Pd(OAc)2, Cs2CO3dioxane; vi) R2CI, Cs2CO3, DMF. Reagents and conditions: i) NH2OH·HCl, Na2CO3, MeOH then N-chlorosuccinimide, DMF; ii) NEt3, DCM; iii) K2CO3, THF; iv) Pd(OAc)2, pivalic acid, K2CO3, DMA; v) TFA, DCM; vi) R2Br, RuPhos, Pd(OAc)2, Cs2CO3, dioxane. EXAMPLES The invention is further illustrated by the following examples. The examples below are non-limiting are merely representative of various aspects of the invention. Solid and dotted wedges within the structures herein disclosed illustrate relative stereochemistry, with absolute stereochemistry depicted only when specifically stated or delineated. General Methods All the starting materials and reagents are commercially available and were used as is.1H Nuclear magnetic resonance (NMR) spectroscopy was carried out using a Bruker instrument operating at 400 MHz using the stated solvent at around room temperature unless otherwise stated. In all cases, NMR data were consistent with the proposed structures. Characteristic chemical shifts (6) are given in parts-per-million using conventional abbreviations for designation of major peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; dd, doublet of doublets; dt, doublet of triplets; m, multiplet; br, broad. Preparative HPLC purification was performed by reverse phase HPLC using a Waters Fractionlynx preparative HPLC system (2525 pump, 2996/2998 UV/VIS detector, 2767 liquid handler) or an equivalent HPLC system such as a Gilson Trilution UV directed system. The Waters 2767 liquid handler acted as both auto-sampler and fraction collector. The columns used for the preparative purification of the compounds were a Waters Sunfire OBD Phenomenex Luna Phenyl Hexyl (10 μm 21.2×150 mm, 10 μm) or Waters Xbridge Phenyl (10 μm 19×150 mm, 5 μm). Appropriate focused gradients were selected based on acetonitrile and methanol solvent systems under either acidic or basic conditions. The modifiers used under acidic/basic conditions were formic acid (0.1% V/V) and ammonium bicarbonate (10 mM) respectively. The purification was controlled by Waters Fractionlynx software through monitoring at 210-400 nm, and triggered a threshold collection value at 260 nm and, when using the Fractionlynx, the presence of target molecular ion as observed under APi conditions. Collected fractions were analysed by LCMS (Waters Acquity systems with Waters SQD). Normal phase flash column chromatography was performed utilizing a Biotage Isolera system. The silica gel columns were purchased from either Interchim or Biotage. The mobile phase was either ethyl acetate in hexanes or methanol in dichloromethane with various ratios, and the fraction collection was triggered by UV absorbance at 254 nm. Analytical high-performance liquid chromatography-mass spectrometry (HPLC-MS) was performed utilizing HP or Waters DAD+Micromass ZQ, single quadrapole LC-MS or Quattro Micro LC-MS-MS. Method 1: The RP-HPLC column was Phenomenex Luna 5 μm C18 (2), (100×4.6 mm). Mobile phase 5-95% acetonitrile in water (0.1% formic acid) gradient, flow rate 2.0 mL/min, and 6.5 min run time; Method 2: The RP-HPLC column was Waters Xterra MS 5 μm C18, 100×4.6 mm. Mobile phase 5-95% acetonitrile in water (10 mM ammonium bicarbonate (ammonium hydrogen carbonate); Method 3: method 1 with mobile phase 50-100% acetonitrile in water (0.1% formic acid) gradient, and 5 min run time; Method 4: method 1 with mobile phase 10-100% acetonitrile in water (0.1% formic acid) gradient, and 10 min run time; Method 5: method 1 with 20 min run time. Abbreviations The following abbreviations are used in the examples:Aq. Aqueous solutionBrettPhos: 2-(dicyclohexylphosphino)3,6-dimethoxy-2′,4′,6′-triisopropyl-1,1′-biphenylCDCl3: deuterochloroformDMSO: dimethyl sulfoxideDMA: N,N-DimethylacetamideESI electrospray Ionisationeq.: equivalentg: gramHATU: (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium-3-oxidhexafluoro-phosphateHPLC: high performance liquid chromatographyM: molarmg: milligramMHz: megahertzMl milliliterMmol: millimoleMP: macroporousMS: mass spectrometryNMP: N-methyl-2-pyrrolidoneNMR: nuclear magnetic resonanceRuPhos: 2-dicyclohexylphosphino-2′,6′-diisopropoxybiphenylSFC: supercritical fluid chromatographyTHF: tetrahydrofuranμL: microlitersDCM: dichloromethaneEtOAc: ethyl acetateNaHCO3: sodium hydrogencarbonateLiCl: lithium chlorideNEt3: triethylamineDMF: dimethylformamideMeOH: methanol Example 1 Step 1: Synthesis of tert-butyl 6-(hydrazinecarbonyl)-2-azaspiro[3.3]heptane-2-carboxylate To a stirred solution of 2-(tert-butoxycarbonyl)-2-azaspiro[3.3]heptane-6-carboxylic acid (0.26 g, 1 mmol, 1 eq.) in THE (5 mL) was added 1-1′-Carbonyldiimidazole (0.19 g, 1.2 mmol, 1.2 eq.) and the mixture was stirred at RT overnight. The resulting mixture was added to a solution of hydrazine monohydrate (0.07 mL, 1.4 mmol, 1.4 eq.) in THE (10 mL) and stirred at RT overnight. The mixture was diluted with brine and extracted with ethyl acetate. The organic phase was separated, washed with brine, dried (MgSO4), filtered and concentrated in vacuo to afford the title compound as a white solid (0.29 g, 100% yield). This material was used without further purification.1H NMR (400 MHz, CDCl3) δ 6.61-6.59 (m, 1H), 3.91-3.80 (m, 6H), 2.81-2.73 (m, 1H), 2.48-2.43 (m, 2H), 2.35-2.30 (m, 2H), 1.40 (s, 9H). Step 2: Synthesis of terttert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate To a solution of tert-butyl 6-(hydrazinecarbonyl)-2-azaspiro[3.3]heptane-2-carboxylate (0.27 g, 1.06 mmol, 1 eq.) in 2-propanol (20 mL) was added 7-chloro-4-methyl-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepin-2-imine (0.22 g, 1.06 mmol, 1 eq.) and the mixture was stirred at 80° C. over the weekend. The reaction mixture was concentrated under reduced pressure to afford an orange solid. Sat. aq. NaHCO3(20 mL) was added to the orange solid and the mixture was extracted with DCM (×2). The organic phases were combined, washed with brine, passed through a hydrophobic phase separator and concentrated in vacuo to give an orange solid. The crude material was purified by silica gel chromatography eluting with 0-10% methanol in DCM to afford the title compound as a yellow solid (0.257 g, 56% yield).1H NMR (400 MHz, CDCl3) δ 7.50 (d, J=7.3 Hz, 2H), 7.12 (d, J=8.9 Hz, 1H), 4.00 (s, 2H), 3.93 (s, 2H), 3.67 (s, 2H), 3.49-3.42 (m, 1H), 3.34 (s, 2H), 2.78 (dd, J=8.3, 12.7 Hz, 2H), 2.63-2.54 (m, 2H), 2.48 (s, 3H), 1.43 (s, 9H). Step 3: Synthesis of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine bis(2,2,2-trifluoroacetate) TFA (0.33 mL, 4.32 mmol, 74.4 eq.) was added dropwise to a solution of tert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (0.025 g, 0.058 mmol, 1.0 eq.) in DCM (1 mL) at RT and mixture was stirred at RT for 20 min. The mixture was concentrated in vacuo to give a yellow residue which was used without further purification (5.3 mg, 26% yield).1H NMR (400 MHz, DMSO) δ 8.80-8.75 (m, 2H), 7.90-7.86 (m, 2H), 7.60-7.58 (d, J=8.7 Hz, 1H), 4.61-4.15 (m, 4H), 4.11-4.08 (m, 2H), 4.01-3.98 (m, 2H), 3.74-3.68 (m, 1H), 2.96 (s, 3H), 2.71-2.53 (m, 4H). Step 4: Synthesis of 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 1) A mixture of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine bis(2,2,2-trifluoroacetate) (0.045 g, 0.08 mmol, 1 eq.), 2-bromo-5-fluoropyridine (0.029 g, 0.16 mmol, 2.04 eq.), RuPhos (0.013 g, 0.03 mmol, 0.345 eq.), palladium acetate (0.003 g, 0.01 mmol, 0.165 eq.), cesium carbonate (0.133 g, 0.41 mmol, 5.06 eq.) in NMP (1 mL) was degassed using N2, for 10 minutes and then heated to 80° C. for 20 minutes. The reaction was allowed to cool and allowed to stand at RT over the weekend. The mixture was diluted with EtOAc, 4% aq. sol. of LiCl and filtered through a layer of celite. The organic phase was separated, washed with aq. sol. of LiCl, dried (MgSO4), filtered and concentrated in vacuo to give a yellow oil. The crude material was purified by column chromatography on a Biotage® KP-NH cartridge eluting with 0-100% EtOAc in isohexane followed by 0-5% methanol in ethyl acetate to give a colourless glass. This was lyophilized to afford the title compound as a white solid (11.5 mg, 21% yield).1H NMR (400 MHz, CDCl3) δ 8.00 (d, J=2.9 Hz, 1H), 7.53-7.49 (m, 2H), 7.25-7.19 (m, 1H), 7.14 (d, J=8.3 Hz, 1H), 6.24 (dd, J=3.3, 8.8 Hz, 1H), 4.08 (s, 2H), 3.99 (s, 2H), 3.68 (s, 2H), 3.57-3.47 (m, 1H), 3.35 (s, 2H), 2.86 (dd, J=8.2, 12.9 Hz, 2H), 2.71-2.62 (m, 2H), 2.48 (s, 3H). m/z 425 (M+H)+. Compound Nos. 2 to 13 and 17 to 21 Compound Nos. 2 to 13 were prepared according to the methods set forth in Example 1. For example, Compound No. 2 of Table 2 lists the method of “Example 1”, indicating that this compound was prepared according to the procedure of Example 1 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 2. Compound Nos. 17 to 21 are prepared according to the procedure of Example 1 using appropriately substituted intermediates. TABLE 2Compound Nos. 2 to 13Compound No.Analytical DataSynthesis Method21H NMR (400 MHz, CDCl3) δ 8.14 (dd, J = 1.0, 5.1 Hz,Example 11H), 7.53-7.48 (m, 2H), 7.46-7.41 (m, 1H), 7.15 (d,J = 8.1 Hz, 1H), 6.61-6.58 (m, 1H), 6.27 (d, J = 8.3 Hz,1H), 4.12 (s, 2H), 4.02 (s, 2H), 3.68 (s, 2H), 3.57-3.47(m, 1H), 3.35 (s, 2H), 2.87 (dd, J = 8.2, 12.8 Hz, 2H), 2.71-2.63 (m, 2H), 2.49 (s, 3H). m/z 407 (M + H)+.31H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.5, 2.8 Hz,Example 11H), 7.84 (d, J = 2.5 Hz, 1H), 7.77 (d, J = 1.3 Hz, 1H), 7.51(d, J = 7.8 Hz, 2H), 7.14 (d, J = 8.6 Hz, 1H), 4.18 (s, 2H),4.12 (s, 2H), 3.68 (s, 2H), 3.58-3.48 (m, 1H), 3.35 (s,2H), 2.89 (dd, J = 8.1, 12.9 Hz, 2H), 2.74-2.65 (m, 2H),2.49 (s, 3H). m/z 408 (M + H)+.41H NMR (400 MHz, CDCl3) δ 7.92-7.89 (m, 1H), 7.53-Example 17.48 (m, 2H), 7.17-7.09 (m, 2H), 6.60-6.55 (m, 1H),4.23 (d, J = 1.8 Hz, 2H), 4.15 (d, J = 2.0 Hz, 2H), 3.68 (s,2H), 3.56-3.46 (m, 1H), 3.35 (s, 2H), 2.86 (dd, J = 8.5,12.8 Hz, 2H), 2.70-2.62 (m, 2H), 2.48 (s, 3H). m/z 425(M + H)+.51H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.3, 4.5 Hz,Example 11H), 7.85 (d, J = 2.8 Hz, 1H), 7.50 (d, J = 7.8 Hz, 2H), 7.16-7.07 (m, 2H), 6.73-6.69 (m, 1H), 4.00 (s, 2H), 3.94 (s,2H), 3.68 (s, 2H), 3.57-3.47 (m, 1H), 3.35 (s, 2H), 2.87(dd, J = 8.0, 12.8 Hz, 2H), 2.72-2.64 (m, 2H), 2.49 (s,3H). m/z 425 (M + H)+.61H NMR (400 MHz, CDCl3) δ 7.50 (d, J = 7.8 Hz, 2H),Example 17.33 (dd, J = 7.7, 7.7 Hz, 1H), 7.15 (d, J = 8.6 Hz, 1H), 6.46(d, J = 7.3 Hz, 1H), 6.08 (d, J = 8.1 Hz, 1H), 4.10 (s, 2H),3.99 (s, 2H), 3.67 (s, 2H), 3.56-3.46 (m, 1H), 3.35 (s,2H), 2.84 (dd, J = 8.3, 12.6 Hz, 2H), 2.69-2.60 (m, 2H),2.48 (s, 3H), 2.39 (s, 3H). m/z 421 (M + H)+.71H NMR (400 MHz, CDCl3) δ 7.51 (d, J = 7.9 Hz, 2H),Example 17.36 (dd, J = 7.8, 7.8 Hz, 1H), 7.15 (d, J = 8.3 Hz, 1H), 6.04(d, J = 7.9 Hz, 1H), 5.83 (d, J = 7.8 Hz, 1H), 4.08 (s, 2H),3.99 (s, 2H), 3.85 (s, 3H), 3.68 (s, 2H), 3.55-3.46 (m,1H), 3.35 (s, 2H), 2.85 (dd, J = 8.2, 12.7 Hz, 2H), 2.69-2.61 (m, 2H), 2.49 (s, 3H). m/z 437 (M + H)+.81H NMR (400 MHz, CDCl3) δ 7.53-7.47 (m, 3H), 7.14Example 1(d, J = 8.3 Hz, 1H), 6.15 (dd, J = 1.9, 7.7 Hz, 1H), 6.05 (dd,J = 2.0, 8.0 Hz, 1H), 4.11 (s, 2H), 4.02 (s, 2H), 3.68 (s,2H), 3.56-3.47 (m, 1H), 3.35 (s, 2H), 2.86 (dd, J = 8.2,12.8 Hz, 2H), 2.71-2.61 (m, 2H), 2.49 (s, 3H). m/z 425(M + H)+.91H NMR (400 MHz, CDCl3) d 7.49 (d, J = 7.4 Hz, 2H),Example 1;7.12 (d, J = 8.8 Hz, 1H), 4.00 (s, 2H), 3.93 (s, 2H), 3.66 (s,Product of2H), 3.49-3.39 (m, 1H), 3.34 (s, 2H), 2.78 (dd, J = 8.3,Step 212.7 Hz, 2H), 2.58 (dd, J = 10.6, 10.6 Hz, 2H), 2.48 (s,3H), 1.43 (s, 9H). m/z 430 (M + H)+.101H NMR (400 MHz, CDCl3) δ 7.85 (d, J = 2.3 Hz, 1H),Example 17.64 (dd, J = 1.9, 1.9 Hz, 1H), 7.51 (d, J = 7.3 Hz, 2H), 7.15-7.12 (m, 1H), 6.42-6.37 (m, 1H), 3.99 (d, J = 18.2 Hz,4H), 3.68 (s, 2H), 3.56-3.47 (m, 1H), 3.35 (s, 2H), 2.88(dd, J = 8.1, 12.9 Hz, 2H), 2.73-2.65 (m, 2H), 2.49 (s,3H). m/z 425 (M + H)+.111H NMR (400 MHz, CDCl3) δ 7.87 (d, J = 2.8 Hz, 1H),Example 17.53-7.48 (m, 2H), 7.17-7.11 (m, 2H), 6.28 (d, J = 9.1Hz, 1H), 4.05 (s, 2H), 3.95 (s, 2H), 3.77 (s, 3H), 3.67 (s,2H), 3.56-3.46 (m, 1H), 3.35 (s, 2H), 2.85 (dd, J = 8.3,12.6 Hz, 2H), 2.69-2.61 (m, 2H), 2.48 (s, 3H). m/z 437(M + H)+.121H NMR (400 MHz, CDCl3) δ7.96 (d, J = 2.3 Hz, 1H),Example 17.53-7.48 (m, 2H), 7.29 (d, J = 3.4 Hz, 1H), 7.15 (d,J = 8.1 Hz, 1H), 6.23 (d, J = 8.3 Hz, 1H), 4.07 (s, 2H), 3.97(s, 2H), 3.67 (s, 2H), 3.56-3.47 (m, 1H), 3.35 (s, 2H),2.85 (dd, J = 8.2, 12.8 Hz, 2H), 2.69-2.61 (m, 2H), 2.48(s, 3H), 2.18 (s, 3H). m/z 421 (M + H)+.131H NMR (400 MHz, CDCl3) δ 8.57 (dd, J = 1.3, 4.5 Hz,Example 11H), 7.54-7.49 (m, 2H), 7.19-7.13 (m, 2H), 6.52 (dd,J = 1.4, 9.0 Hz, 1H), 4.26 (s, 2H), 4.13 (s, 2H), 3.68 (s,2H), 3.59-3.49 (m, 1H), 3.35 (s, 2H), 2.89 (dd, J = 8.0,12.8 Hz, 2H), 2.75-2.67 (m, 2H), 2.49 (s, 3H). m/z 408(M + H)+. Example 2 Step 1: Synthesis of 1-(6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-yl)-2,2,2-trifluoroethan-1-one To a solution of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine bis(2,2,2-trifluoroacetate) from Example 1 step 3 (0.055 g, 0.1 mmol, 1 eq.), NEt3(69 μL, 0.49 mmol, 5.0 eq.) in DCM (2 mL) was added trifluoroacetic anhydride (21 μL, 0.15 mmol, 1.5 eq.) and the mixture was stirred at RT for 1 hour. The mixture was diluted with DCM/water and the organic layer was separated by passing through a hydrophobic phase separator. The organic phase was concentrated in vacuo to give the title compound as a yellow oil. This material was used without further purification (32 mg). Step 2: Synthesis of 1-(6-(8-chloro-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-yl)ethan-1-one (Compound No. 15) A suspension of 1-(6-(8-chloro-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4] diazepin-1-yl)-2-azaspiro[3.3]heptan-2-yl)-2,2,2-trifluoroethan-1-one (0.43 g, 0.1 mmol, 1 eq.) from step 1, K2CO3(0.056 g, 0.4 mmol, 4.0 eq.) and MeOH (2 mL) was stirred at RT for 1 hour. The mixture was concentrated under reduced pressure and the residue was treated with 20% MeOH in DCM, filtered and the filtrate was concentrated under reduced pressure to give a yellow gum. To a solution of the yellow gum in DCM (2 mL) was added NEt3(21.11 μL, 1.50 eq.), AcCl (10.81 μL, 0.15 mmol, 1.50 eq.) and the mixture was stirred at RT for 45 minutes. The mixture was concentrated under reduced pressure and the residue was treated with EtAc, sat. sol. of NaHCO3. The organic phase was separated, the aqueous phase was extracted with EtOAc. The organic phases were combined, dried (MgSO4), filtered and concentrated in vacuo to give a yellow residue. This was purified using reverse phase column chromatography on a C18 cartridge, eluting with 5-50% MeCN/H2O/0.1% formic acid. After lyophilisation this give the title compound as a white solid (4.5 mg, 10% yield)1H NMR (400 MHz, CDCl3) δ 7.53-7.49 (m, 2H), 7.14-7.09 (m, 1H), 4.19 (d, J=12.4 Hz, 2H), 4.06 (s, 1H), 3.99 (s, 1H), 3.67 (s, 2H), 3.53-3.43 (m, 1H), 3.34 (s, 2H), 2.87-2.78 (m, 2H), 2.69-2.57 (m, 2H), 2.48 (s, 3H), 1.85 (s, 3H). m/z 372 (M+H)+. Compound 16 Compound No. 16 was prepared according to the methods set forth in Example 2. using appropriately substituted intermediates. Analytical data (NMR, mass spectrum):1H NMR (400 MHz, DMSO) δ 7.76-7.75 (m, 1H), 7.65 (dd, J=2.4, 8.5 Hz, 1H), 7.50 (d, J=8.5 Hz, 1H), 4.59 (s, 2H), 4.42 (s, 2H), 4.10-4.08 (m, 4H), 3.70-3.62 (m, 1H), 2.61 (d, J=8.2 Hz, 4H), 2.56-2.53 (m, 2H), 1.52-1.45 (m, 1H), 1.10 (t, J=7.3 Hz, 3H), 0.71-0.68 (m, 4H). m/z 440 (M+H)+. Example 3 To a solution of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine bis(2,2,2-trifluoroacetate) from Example 1 Step 3 (0.202 g, 0.47 mmol, 1 eq.) in MeOH (10 mL) was added MP-carbonate (2.43 g, 7.05 mmol, 15.0 eq.) and the mixture was stirred at RT for 2 hours. The mixture was filtered and concentrated in vacuo to give a yellow oil. To a solution of the yellow oil (0.03 g, 0.09 mmol, 1.0 eq.) in DCM (1 mL) was added DIPEA (40 μL, 0.23 mmol, 2.5 eq.), picolinoyl chloride hydrochloride (0.19 g, 0.11 mmol, 1.2 eq.) and the mixture was stirred at RT for 15 minutes. The mixture was diluted with DCM, passed through a hydrophobic phase separator and concentrated in vacuo to give a residue. The residue was purified by preparative HPLC to give the title compound (23.7 mg, 60% yield).1H NMR (400 MHz, CDCl3) δ 8.56 (dd, J=4.5, 9.6 Hz, 1H), 8.09 (dd, J=8.0, 16.8 Hz, 1H), 7.83-7.76 (m, 1H), 7.54-7.48 (m, 2H), 7.37-7.33 (m, 1H), 7.13 (d, J=8.3 Hz, 1H), 4.81 (s, 1H), 4.72 (s, 1H), 4.30 (s, 1H), 4.22 (s, 1H), 3.67 (s, 2H), 3.53-3.44 (m, 1H), 3.35 (s, 2H), 2.90-2.81 (m, 2H), 2.71-2.61 (m, 2H), 2.48 (s, 3H). m/z 435 (M+H)+. Compounds 14 and 25 to 33 Compound Nos. 14 and 25 to 33 were prepared according to the methods set forth in Example 3. For example, Compound No. 14 of Table 3 lists the method of “Example 3”, indicating that this compound was prepared according to the procedure of Example 3 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 3. TABLE 3Compound Nos. 14 and 25 to 33Compound No.Analytical DataSynthesis Method141H NMR (400 MHz, CDCl3) δ 7.52-7.48 (m,Example 32H), 7.12-7.07 (m, 1H), 4.92-4.86 (m, 2H),(using oxetane-3-4.76-4.69 (m, 2H), 4.10-4.03 (m, 4H), 3.83-carbonyl chloride)3.74 (m, 1H), 3.69 (s, 2H), 3.52-3.43 (m, 1H),3.36 (s, 2H), 2.87-2.76 (m, 2H), 2.70-2.57 (m,2H), 2.49 (s, 3H). m/z 414 (M + H)+.251H NMR (400 MHz, CDCl3) δ 7.50 (d, J = 7.2Example 3Hz, 2H), 7.12-7.09 (m, 1H), 4.04 (d, J = 14.3 Hz,(using 4-4H), 3.69-3.62 (m, 6H), 3.49-3.39 (m, 1H),morpholinecarbonyl3.33 (dd, J = 4.8, 9.7 Hz, 6H), 2.78 (dd, J = 7.8,chloride)12.8 Hz, 2H), 2.66-2.58 (m, 2H), 2.48 (s, 3H).m/z 443 (M + H)+.261H NMR (400 MHz, CDCl3) δ 7.51-7.49 (m,Example 32H), 7.12-7.09 (m, 1H), 4.00 (d, J = 18.6 Hz,(using4H), 3.67 (s, 2H), 3.51-3.42 (m, 1H), 3.34 (s,cyclopropylsulfonyl2H), 2.81 (dd, J = 8.0, 13.0 Hz, 2H), 2.68-2.60chloride)(m, 2H), 2.48 (s, 3H), 2.35-2.27 (m, 1H), 1.17-1.12 (m, 2H), 1.01-0.97 (m, 2H). m/z 434(M + H)+.271H NMR (400 MHz, DMSO) δ 7.71 (d, J = 2.4Example 3Hz, 1H), 7.66-7.63 (m, 1H), 7.48 (dd, J = 2.3, 8.5(using tetrahydro-2H-Hz, 1H), 4.24-4.20 (m, 2H), 4.05 (s, 1H), 3.90-pyran-4-carbonyl3.80 (m, 3H), 3.76 (s, 2H), 3.51-3.36 (m, 3H),chloride)3.35 (s, 2H), 2.85-2.79 (m, 2H), 2.72-2.63(m, 2H), 2.48 (s, 3H), 2.41-2.35 (m, 1H), 1.89-1.75 (m, 2H), 1.53-1.46 (m, 2H). m/z 442(M + H)+.281H NMR (400 MHz, DMSO) δ 7.71 (d, J = 2.5Example 3Hz, 1H), 7.64 (dd, J = 2.4, 8.5 Hz, 1H), 7.49 (dd,(usingJ = 3.0, 8.5 Hz, 1H), 4.30 (s, 1H), 4.18 (s, 1H),cyclopropylcarbonyl3.91 (s, 1H), 3.77 (s, 1H), 3.68-3.61 (m, 1H),chloride)3.48 (s, 2H), 3.37 (s, 2H), 2.56-2.53 (m, 4H),2.33 (s, 3H), 1.51-1.43 (m, 1H), 0.70-0.64 (m,4H). m/z 398 (M + H)+.291H NMR (400 MHz, CDCl3) δ 7.52-7.49 (m,Example 32H), 7.13-7.10 (m, 1H), 4.85-4.77 (m, 1H),(using tetrahydro-2H-4.06 (s, 2H), 4.00 (s, 2H), 3.92-3.85 (m, 2H),pyran-4-yl3.68 (s, 2H), 3.56-3.43 (m, 3H), 3.35 (s, 2H),carbonochloridate)2.80 (dd, J = 8.2, 12.7 Hz, 2H), 2.65-2.58 (m,2H), 2.49 (s, 3H), 1.93-1.84 (m, 2H), 1.69-1.59 (m, 2H). m/z 458 (M + H)+.301H NMR (400 MHz, CDCl3) δ 7.54-7.47 (m,Example 32H), 7.12 (dd, J = 8.1, 8.1 Hz, 1H), 4.20 (d,(using isobutyrylJ = 13.9 Hz, 2H), 4.05 (s, 1H), 3.98 (s, 1H), 3.67chloride)(s, 2H), 3.51-3.43 (m, 1H), 3.35 (s, 2H), 2.87-2.76 (m, 2H), 2.70-2.59 (m, 2H), 2.48 (s, 4H),1.08 (d, J = 6.6 Hz, 6H). m/z 400 (M + H)+.311H NMR (400 MHz, DMSO) δ 7.65-7.59 (m,Example 32H), 7.45 (d, J = 8.4 Hz, 1H), 4.13 (s, 2H), 4.04 (s,(using 1-2H), 3.67-3.58 (m, 1H), 3.53 (s, 2H), 3.40 (s,methylcyclopropane-2H), 2.60-2.54 (m, 4H), 2.38 (s, 3H), 1.24 (s,1-carbonyl chloride)3H), 0.92-0.89 (m, 2H), 0.48-0.44 (m, 2H).m/z 412 (M + H)+.321H NMR (400 MHz, CDCl3) δ 7.54-7.47 (m,Example 32H), 7.14-7.08 (m, 1H), 4.16 (d, J = 12.6 Hz,(using 2-2H), 4.09-4.05 (m, 1H), 4.00 (s, 1H), 3.67 (s,cyclopropylacetyl2H), 3.48 (dd, J = 7.8, 7.8 Hz, 1H), 3.34 (s, 2H),chloride)2.86-2.76 (m, 2H), 2.69-2.60 (m, 2H), 2.47 (s,3H), 2.01 (d, J = 6.6 Hz, 2H), 1.03 (d, J = 1.5 Hz,1H), 0.54 (d, J = 6.3 Hz, 2H), 0.17-0.14 (m, 2H).m/z 412 (M + H)+.331H NMR (400 MHz, CDCl3) δ 7.53-7.48 (m,Example 32H), 7.11 (d, J = 8.7 Hz, 1H), 5.21-5.15 (m, 1H),4.71-4.57 (m, 2H), 4.42-4.25 (m, 2H), 4.15 (s,1H), 4.08 (s, 1H), 3.67 (s, 2H), 3.51-3.42 (m,1H), 3.34 (s, 2H), 2.94-2.77 (m, 4H), 2.69-2.59 (m, 2H), 2.48 (s, 3H). m/z 414 (M + H)+. Example 4 Step 1: Synthesis of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine To a solution of tert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate form Example 1 Step 2 (415 mg, 0.965 mmol, 1 eq.) in DCM (17 mL) was added TFA (6 mL) dropwise at RT. Mixture was stirred at RT for 15 minutes and concentrated in vacuo to give an off white solid. MeOH was added and the mixture was passed through an SCX (10 g, cartridge) eluting with MeOH followed by 2.3M ammonia solution in MeOH to give the title compound as an off white solid. (300 mg, 94% yield).1H NMR (400 MHz, CDCl3) δ 7.52-7.48 (m, 2H), 7.14 (d, J=8.4 Hz, 1H), 3.72 (s, 2H), 3.65 (d, J=11.9 Hz, 4H), 3.46-3.38 (s, 1H), 3.33 (s, 2H), 2.73 (dd, J=8.4, 12.5 Hz, 2H), 2.63-2.54 (m, 2H), 2.47 (s, 3H). Step 2: Synthesis of 8-chloro-5-methyl-1-(2-(methylsulfonyl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 34) To a solution of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine 30 mg, 0.091 mmol, 1 eq.), DIPEA (19 μL, 0.109 mmol, 1.2 eq.) in DCM 91 mL) was added methanesulfonyl chloride (8 μL, 0.10 mmol, 1.1 eq.) and the mixture was stirred at RT for 15 minutes. The mixture was diluted with DCM/H2O and passed through a phase separator. The organics were concentrated in vacuo and the residue purified by preparative HPLC to give the title compound as an off white solid. (4.4 mg, 12% yield).1H NMR (400 MHz, CDCl3) δ 7.51-7.49 (m, 2H), 7.11-7.08 (m, 1H), 3.99 (s, 2H), 3.95 (s, 2H), 3.67 (s, 2H), 3.52-3.43 (m, 1H), 3.34 (s, 2H), 2.84 (s, 3H), 2.83-2.79 (m, 2H), 2.68-2.60 (m, 2H), 2.48 (s, 3H). m/z 408 (M+H)+. Compounds 35 to 37 Compound Nos. 35 to 37 were prepared according to the methods set forth in Example 4, using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 4. TABLE 4Compound Nos. 35 to 37Compound No.Analytical DataSynthesis Method351H NMR (400 MHz, DMSO) δ 7.65-7.59 (m,Example 42H), 7.44 (d, J = 8.4 Hz, 1H), 3.99-3.97 (s, 2H),3.92-3.89 (s, 2H), 3.66-3.62 (m, 1H), 3.52 (s,2H), 3.38 (s, 2H), 3.10-3.01 (m, 1H), 2.58-2.55 (m, 4H), 2.34 (s, 3H), 2.19-2.05 (m, 4H),1.98-1.79 (m, 2H). m/z 412 (M + H)+.361H NMR (400 MHz, CDCl3) δ 7.52-7.49 (m,Example 42H), 7.12 (d, J = 7.9 Hz, 1H), 4.52 (d, J = 3.8 Hz,1H), 4.46 (d, J = 3.9 Hz, 1H), 4.15 (s, 1H), 4.07(s, 1H), 3.67 (s, 2H), 3.53-3.44 (m, 1H), 3.34 (s,2H), 2.84 (dd, J = 8.2, 12.9 Hz, 2H), 2.67-2.61(m, 2H), 2.48 (s, 3H), 1.36-1.30 (m, 2H), 1.26-1.16 (m, 2H). m/z 416 (M + H)+.371H NMR (400 MHz, DMSO) d 7.74 (s, 1H),Example 47.64 (dd, J = 2.4, 8.5 Hz, 1H), 7.49 (d, J = 8.5 Hz,1H), 4.59 (s, 2H), 4.41 (s, 2H), 3.99 (s, 2H), 3.90-3.87 (m, 2H), 3.68-3.60 (m, 1H), 3.10-3.01(m, 1H), 2.60 (d, J = 7.3 Hz, 4H) 2.56-2.54 (m,2H), 2.17-2.05 (m, 4H), 1.98-1.78 (m, 2H),1.10 (t, J = 7.4 Hz, 3H). m/z 454 (M + H)+. Example 5 To a solution of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine from ex4 step 1 (30 mg, 0.091 mmol, 1 eq.) in DMF (1 mL) was added Cs2CO3(36 mg, 0.19 mmol, 1.2 eq.), 2-chloropyrimidine (10 mg, 0.091 mmol, 1 eq.) and the mixture was heated to 80° C. overnight. The mixture was diluted with water and extracted with EtOAc (3×). The organic fractions were combined, washed with brine, dried (MgSO4), filtered, concentrated in vacuo to give a yellow film and purified using preparative HPLC to give the title compound as an off white solid. (11 mg, 29% yield).1H NMR (400 MHz, DMSO) δ 8.32 (d, J=4.8 Hz, 2H), 7.71 (d, J=2.4 Hz, 1H), 7.65 (dd, J=2.4, 8.5 Hz, 1H), 7.50 (d, J=8.4 Hz, 1H), 6.65 (dd, J=4.8, 4.8 Hz, 1H), 4.10 (s, 2H), 3.96 (s, 2H), 3.74-3.64 (m, 1H), 3.49 (s, 2H), 3.37 (s, 2H), 2.61-2.54 (m, 4H), 2.33 (s, 3H). m/z 408 (M+H)+. Example 6 Step 1: Synthesis of tert-butyl (4-chloro-2-(((2-methoxyethyl)amino)methyl)phenyl)carbamate To a solution of tert-butyl (4-chloro-2-formylphenyl)carbamate (0.4 g, 1.56 mmol, 1.0 eq.) in MeOH (10 mL) was added 2-methoxyethylamine (273 μL, 3.13 mmol, 2.0 eq.) and the mixture was heated to 65° C. for 45 minutes. The mixture was cooled in ice and sodium borohydride (0.12 g, 3.13 mmol, 2.0 eq.) was added portionwise. An additional amount of sodium borohydride (0.06 g, 1.56 mmol, 1 eq.) was added after 1 hour at RT. The mixture was stirred at RT for 16 hours, diluted with EtOAc/sat. NaHCO3and the organic layer separated. The aqueous phase was extracted with fresh EtOAc (×2), the organic phases were combined, washed with brine, dried (MgSO4), filtered and concentrated in vacuo to give the title compound as a colourless oil. (0.429 g, 98% yield).1H NMR (400 MHz, CDCl3) δ 9.70 (s, 1H), 7.95 (d, J=8.5 Hz, 1H), 7.20 (dd, J=2.5, 8.8 Hz, 1H), 7.06 (d, J=2.5 Hz, 1H), 3.82 (s, 2H), 3.50 (t, J=5.0 Hz, 2H), 3.37 (s, 3H), 2.75 (t, J=5.0 Hz, 2H), 1.53 (s, 9H). Step 2: Synthesis of tert-butyl (4-chloro-2-(((cyanomethyl)(2-methoxyethyl)amino)methyl)phenyl)carbamate A mixture of tert-butyl (4-chloro-2-(((2-methoxyethyl)amino)methyl)phenyl) carbamate (0.49 g, 1.56 mmol, 1 eq.), sodium bicarbonate (0.14 g, 1.72 mmol, 1.10 eq.), potassium iodide (0.05 g, 0.31 mmol, 0.20 eq.), chloroacetonitrile (0.109 mL, 1.72 mmol, 1.10 eq.) in EtOAc (5 mL) was heated to 75° C. overnight. The mixture was allowed to cool, diluted with EtOAc/NaHCO3(1:1) and the organic layer was separated, washed with brine, dried (MgSO4), filtered and concentrated in vacuo to give a yellow oil. The residue was triturated with MeOH to give the title compound as an off white solid. (0.320 g, 58% yield)1H NMR (400 MHz, CDCl3) δ 8.47 (s, 1H), 7.97 (d, J=8.8 Hz, 1H), 7.29-7.27 (m, 1H), 7.16 (d, J=2.5 Hz, 1H), 3.74 (s, 2H), 3.62-3.58 (m, 4H), 3.40 (s, 3H), 2.86 (t, J=5.1 Hz, 2H), 1.52 (s, 9H). Step 3: Synthesis of 7-chloro-4-(2-methoxyethyl)-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine Acetyl chloride (1.21 mL, 16.96 mmol, 20.0 eq.) was added to 2-propanol (7 mL) at RT over 20 minutes (reaction is exothermic) to give a suspension. This suspension was added to a solution of tert-butyl (4-chloro-2-(((cyanomethyl)(2-methoxyethyl)amino)methyl)phenyl) carbamate (0.30 g, 0.85 mmol, 1.0 eq.) in 2-propanol (8 mL) and the mixture was heated to 40° C. overnight. The mixture was allowed to cool, concentrated in vacuo, EtOAc/NaHCO3was added and the organic phase was separated. The aqueous layer was extracted with further EtOAc (×2), the organics were combined, washed with brine and concentrated in vacuo to give a yellow oil. This was purified using flash column chromatography (Biotage® KP-NH) eluting with 0-100% EtOAc in isohexane followed by 0-10% methanol in ethyl acetate to give a yellow glass. (0.196 g, 91% yield).1H NMR (400 MHz, CDCl3) δ 7.24-7.19 (m, 2H), 6.92 (d, J=6.9 Hz, 1H), 3.61-3.57 (m, 2H), 3.45 (s, 2H), 3.39 (s, 3H), 3.23 (s, 2H), 2.88-2.84 (m, 2H). Step 4: Synthesis of methyl 2-(pyridin-2-yl)-2-azaspiro[3.3]heptane-6-carboxylate To a solution of 2-(tert-butyl) 6-methyl 2-azaspiro[3.3]heptane-2,6-dicarboxylate (1.10 g, 4.31 mmol, 1.0 eq.) in DCM (15 mL) was added TFA (5 mL) and the mixture was stirred at RT for 1 hour. Toluene was added and the mixture was concentrated in vacuo to give a yellow oil. A mixture of the yellow oil (0.58 g, 2.15 mmol, 1.0 eq.), 2-bromopyridine (247 μL, 2.59 mmol, 1.2 eq.), RuPhos (0.201 g, 0.43 mmol, 0.20 eq.), palladium acetate (0.048 g, 0.22 mmol, 0.1 eq.), cesium carbonate (2.106 g, 6.46 mmol, 3.0 eq.) in dioxane (10 mL) was de-gassed using nitrogen for 10 minutes and heated to 80° C. overnight. The mixture was diluted with EtOAc, filtered through a layer of celite and the filtrate was concentrated in vacuo. The residue was purified by flash column chromatography (Biotage® KP-NH) eluting with 0-50% EtOAc in Isohexane to give the title compound as a pale yellow oil. (0.40 g, 80% yield).1H NMR (400 MHz, CDCl3) δ 8.13 (dd, J=1.0, 5.1 Hz, 1H), 7.45-7.40 (m, 1H), 6.58 (dd, J=5.2, 6.2 Hz, 1H), 6.26 (d, J=8.6 Hz, 1H), 4.04 (s, 2H), 3.97 (s, 2H), 3.70 (s, 3H), 3.09-3.02 (m, 1H), 2.53-2.47 (m, 4H). Step 5: Synthesis of 2-(pyridin-2-yl)-2-azaspiro[3.3]heptane-6-carbohydrazide To a solution of methyl 2-(pyridin-2-yl)-2-azaspiro[3.3]heptane-6-carboxylate 0.4 g, 1.72 mmol, 1.0 eq.) in MeOH (5 mL) was added hydrazine hydrate (501 μL, 10.33 mmol, 6.0 eq.) over 3 minutes. The mixture was stirred at RT overnight. The mixture was diluted with EtOAc, filtered through a layer of celite, the filtrate was washed with brine and the organic layer was separated. The aqueous phase was father extracted with fresh EtOAc (×2), DCM (×2), the organic phases were combine and concentrated in vacuo to give the title compound as a colourless oil. (0.324 g, 81% yield).1H NMR (400 MHz, CDCl3) δ 8.13 (dd, J=1.0, 5.1 Hz, 1H), 7.46-7.40 (m, 1H), 6.63 (s, 1H), 6.61-6.57 (m, 1H), 6.27 (d, J=8.3 Hz, 1H), 4.06 (s, 2H), 3.97 (s, 2H), 3.92-3.86 (m, 2H), 2.90-2.81 (m, 1H), 2.59-2.52 (m, 2H), 2.46-2.39 (m, 2H). Step 6: Synthesis of 8-chloro-5-(2-methoxyethyl)-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 39) A mixture of 7-chloro-4-(2-methoxyethyl)-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine from step 3 (0.036 g, 0.14 mmol, 1.0 eq.), 2-(pyridin-2-yl)-2-azaspiro[3.3]heptane-6-carbohydrazide from step 4 (0.035 g, 0.15 mmol, 1.05 eq.), AcOH (2 drops) in 2-propanol (1 mL) was heated to 80° C. for 90 minutes. The mixture was concentrated in vacuo, diluted with DCM/NaHCO3and passed through a through a phase separator and concentrated in vacuo. The residue was purified using preparative PLC to give the title compound. (0.031 g, 480 yield).1H NMR (400 MHz, CDCl3) δ 8.14-8.13 (m, 1H), 7.56-7.51 (m, 2H), 7.45-7.41 (m, 1H), 7.16 (d, J=8.6 Hz, 1H), 6.61-6.58 (m, 1H), 6.27 (d, J=8.3 Hz, 1H), 4.1 (s, 2H), 4.03 (s, 2H), 3.76-3.70 (m, 2H), 3.60-3.57 (s, 2H), 3.57-3.47 (m, 3H), 3.39 (s, 3H), 2.89-2.81 (m, 4H), 2.69-2.64 (m, 2H). m/z 451 (M+H)+. Compounds 40 to 56 Compound Nos. 40 to 56 were prepared according to the methods set forth in Example 6 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 5. TABLE 5Compound Nos. 40 to 56Compound No.Analytical DataSynthesis Method401H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.5,Example 62.5 Hz, 1H), 7.84 (d, J = 2.8 Hz, 1H), 7.77 (d,J = 1.5 Hz, 1H), 7.51-7.46 (m, 2H), 7.13 (d,J = 8.3 Hz, 1H), 4.17 (s, 2H), 4.11 (s, 2H), 3.63-3.47 (m, 3H), 3.28 (s, 2H), 3.18-3.09 (m, 1H),2.88 (dd, J = 8.1, 12.9 Hz, 2H), 2.73-2.65 (m,2H), 2.19-2.13 (m, 2H), 2.01-1.90 (m, 2H).m/z 448 (M + H)+.411H NMR (400 MHz, CDCl3) δ 8.13 (dd, J = 1.0,Example 65.1 Hz, 1H), 7.51-7.41 (m, 3H), 7.14 (d, J = 8.3Hz, 1H), 6.59 (dd, J = 5.2, 6.2 Hz, 1H), 6.27 (d,J = 8.3 Hz, 1H), 4.11 (s, 2H), 4.01 (s, 2H), 3.60-3.48 (m, 3H), 3.28 (s, 2H), 3.18-3.09 (m, 1H),2.86 (dd, J = 8.2, 12.8 Hz, 2H), 2.70-2.61 (m,2H), 2.19-2.11 (m, 2H), 2.01-1.89 (m, 2H),1.82-1.65 (m, 2H). m/z 447 (M + H)+.421H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.8Example 6Hz, 1H), 7.51-7.45 (m, 2H), 7.25-7.19 (m,1H), 7.13 (d, J = 8.3 Hz, 1H), 6.23 (dd, J = 3.3, 9.1Hz, 1H), 4.07 (s, 2H), 3.98 (s, 2H), 3.63-3.48(m, 3H), 3.27 (s, 2H), 3.18-3.08 (m, 1H), 2.85(dd, J = 8.1, 12.6 Hz, 2H), 2.69-2.61 (m, 2H),2.20-2.12 (m, 2H), 2.01-1.88 (m, 2H), 1.82-1.65 (m, 2H). m/z 464 (M + H)+.431H NMR (400 MHz, CDCl3) 8.01 (1H, dd, J = 1.5,Example 62.8 Hz), 7.84 (1H, d, J = 2.8 Hz), 7.77 (1H, d,J = 1.5 Hz), 7.53-7.48 (2H, m), 7.13 (1H, d,J = 8.3 Hz), 4.18 (2H, s), 4.12 (2H, s), 3.76 (2H,s), 3.60-3.47 (5H, m), 3.40 (3H, s), 2.92-2.80(4H, m), 2.73-2.65 (2H, m). m/z 452 (M + H)+.441H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 3.0Example 6Hz, 1H), 7.53-7.48 (m, 2H), 7.22 (ddd, J = 2.9,8.0, 9.0 Hz, 1H), 7.14 (d, J = 8.3 Hz, 1H), 6.23(dd, J = 3.5, 9.1 Hz, 1H), 4.08 (s, 2H), 3.98 (s,2H), 3.76 (s, 2H), 3.60-3.46 (m, 5H), 3.39 (s,3H), 2.89-2.80 (m, 4H), 2.70-2.62 (m, 2H).m/z 469 (M + H)+.451H NMR (400 MHz, CDCl3) δ 8.14 (dd, J = 1.0,Example 65.1 Hz, 1H), 7.50 (d, J = 7.3 Hz, 2H), 7.46-7.41(m, 1H), 7.17-7.13 (m, 1H), 6.59 (dd, J = 5.1, 6.3Hz, 1H), 6.27 (d, J = 8.3 Hz, 1H), 4.11 (s, 2H),4.01 (s, 2H), 3.70 (s, 2H), 3.57-3.47 (m, 1H),3.40 (s, 2H), 2.87 (dd, J = 8.2, 12.8 Hz, 2H), 2.71-2.62 (m, 4H), 1.19 (dd, J = 7.2, 7.2 Hz, 3H). m/z421 (M + H)+.461H NMR (400 MHz, CDCl3) δ 8.01 (1H, dd,Example 6J = 1.5, 2.8 Hz), 7.84 (1H, d, J = 2.8 Hz), 7.77 (1H,d, J = 1.3 Hz), 7.52-7.49 (2H, m), 7.15-7.12(1H, m), 4.18 (2H, s), 4.12 (2H, s), 3.70 (2H, s),3.58-3.48 (1H, m), 3.40 (2H, s), 2.89 (2H, dd,J = 8.1, 12.9 Hz), 2.73-2.62 (4H, m), 1.19 (3H,dd, J = 7.2, 7.2 Hz). m/z 422 (M + H)+.471H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.5,Example 62.8 Hz, 1H), 7.84 (d, J = 2.8 Hz, 1H), 7.77 (d,J = 1.5 Hz, 1H), 7.53-7.47 (m, 2H), 7.12 (d,J = 8.3 Hz, 1H), 4.18 (s, 2H), 4.12 (s, 2H), 3.81 (s,2H), 3.56-3.49 (m, 3H), 2.99-2.86 (m, 3H),2.73-2.66 (m, 2H), 1.19 (d, J = 6.6 Hz, 6H). m/z436 (M + H)+.481H NMR (400 MHz, CDCl3) δ 8.14 (1H, dd,Example 6J = 1.0, 5.1 Hz), 7.52-7.41 (3H, m), 7.13 (1H, d,J = 8.6 Hz), 6.59 (1H, dd, J = 5.2, 6.2 Hz), 6.27(1H, d, J = 8.3 Hz), 4.11 (2H, s), 4.02 (2H, s),3.81 (2H, s), 3.51 (3H, d, J = 9.1 Hz), 2.98-2.83(3H, m), 2.70-2.63 (2H, m), 1.19 (6H, d, J = 6.3Hz). m/z 435 (M + H)+.491H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Example 6Hz, 1H), 7.51-7.49 (m, 2H), 7.25-7.19 (m,1H), 7.16-7.13 (m, 1H), 6.24 (dd, J = 3.5, 9.0 Hz,1H), 4.08 (s, 2H), 3.99 (s, 2H), 3.70 (s, 2H), 3.55-3.47 (m, 1H), 3.40 (s, 2H), 2.86 (dd, J = 8.2,12.8 Hz, 2H), 2.70-2.62 (m, 4H), 1.19 (dd,J = 7.2, 7.2 Hz, 3H). m/z 439 (M + H)+.501H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 3.0Example 6Hz, 1H), 7.52-7.46 (m, 2H), 7.25-7.19 (m,1H), 7.12 (d, J = 8.4 Hz, 1H), 6.24 (dd, J = 3.5, 9.0Hz, 1H), 4.08 (s, 2H), 3.99 (s, 2H), 3.81 (s, 2H),3.51 (d, J = 8.0 Hz, 3H), 3.00-2.83 (m, 3H), 2.70-2.61 (m, 2H), 1.19 (d, J = 6.5 Hz, 6H). m/z 453(M + H)+.511H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.1,Example 64.7 Hz, 1H), 7.85 (d, J = 2.8 Hz, 1H), 7.53-7.46(m, 2H), 7.13-7.07 (m, 2H), 6.73-6.69 (m,1H), 4.00 (s, 2H), 3.94 (s, 2H), 3.81 (s, 2H), 3.51(d, J = 5.8 Hz, 3H), 2.99-2.83 (m, 3H), 2.72-2.64 (m, 2H), 1.19 (d, J = 6.6 Hz, 6H). m/z 435(M + H)+.521H NMR (400 MHz, CDCl3) δ 8.14 (dd, J = 1.0,Example 65.3 Hz, 1H), 7.56-7.50 (m, 2H), 7.49-7.43 (m,1H), 7.17 (d, J = 8.3 Hz, 1H), 6.61 (dd, J = 5.2, 6.2Hz, 1H), 6.28 (d, J = 8.3 Hz, 1H), 5.95 (tt, J = 4.1,40 Hz, 1H) 4.14 (s, 2H), 4.04 (s, 2H), 3.82 (s,2H), 3.56-3.49 (m, 3H), 3.07-2.95 (m, 2H),2.87 (dd, J = 8.3, 12.9 Hz, 2H), 2.72-2.64 (m,2H). m/z 457 (M + H)+.531H NMR (400 MHz, CDCl3) δ 7.92-7.89 (m,Example 61H), 7.53-7.48 (m, 2H), 7.16-7.09 (m, 2H),6.60-6.55 (m, 1H), 4.23 (d, J = 1.5 Hz, 2H), 4.15(d, J = 2.0 Hz, 2H), 3.74 (s, 2H), 3.60-3.46 (m,5H), 3.39 (s, 3H), 2.89-2.80 (m, 4H), 2.70-2.62 (m, 2H). m/z 469 (M + H)+.541H NMR (400 MHz, CDCl3) δ 7.91 (d, J = 4.9Example 6Hz, 1H), 7.52-7.47 (m, 2H), 7.16-7.10 (m,2H), 6.60-6.55 (m, 1H), 4.23 (d, J = 1.6 Hz, 2H),4.15 (d, J = 2.0 Hz, 2H), 3.81 (s, 2H), 3.54-3.48(m, 3H), 3.00-2.83 (m, 3H), 2.70-2.63 (m,2H), 1.19 (d, J = 6.4 Hz, 6H). m/z 453 (M + H)+.551H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.3,Example 64.8 Hz, 1H), 7.85 (d, J = 2.5 Hz, 1H), 7.53-7.47(m, 2H), 7.15-7.07 (m, 2H), 6.73-6.69 (m,1H), 4.00 (s, 2H), 3.94 (s, 2H), 3.75 (s, 2H), 3.60-3.48 (m, 5H), 3.39 (s, 3H), 2.90-2.80 (m, 4H),2.72-2.63 (m, 2H). m/z 451 (M + H)+.561H NMR (400 MHz, CDCl3) δ 8.14 (dd, J = 1.0,Example 65.1 Hz, 1H), 7.52-7.48 (m, 2H), 7.46-7.41 (m,1H), 7.17-7.14 (m, 1H), 6.59 (dd, J = 5.4, 6.7 Hz,1H), 6.27 (d, J = 8.3 Hz, 1H), 4.12 (s, 2H), 4.02(s, 2H), 3.82 (s, 2H), 3.56-3.49 (m, 3H), 2.87(dd, J = 8.2, 12.8 Hz, 2H), 2.71-2.62 (m, 2H),2.09-2.00 (m, 1H), 0.62-0.56 (m, 4H). m/z433 (M + H)+. Example 7 Step 1: Synthesis of tert-butyl 8-chloro-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate To a solution of tert-butyl 7-chloro-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (0.4 g, 1.28 mmol, 1.0 eq.), was added 2-(pyridin-2-yl)-2-azaspiro[3.3]heptane-6-carbohydrazide from Ex6 step 4 (0.320 g, 1.28 mmol, 1.0 eq.) in dioxane (10 mL) and the mixture was heated to 90° C. for 36 hours. The mixture was allowed to cool, saturated with nitrogen gas and was concentrated in vacuo. The residue was purified by flash column chromatography eluting with 50-100 EtOAc in isohexane then 0-10% MeOH in DCM to give the title product as an off white foam. (0.50 g, 81% yield). m/z 493 (M+H)+. Step 2: Synthesis of 8-chloro-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine hydrochloride To a solution of tert-butyl 8-chloro-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (0.10 g, 0.20 mmol, 1.0 eq.) in MeOH was added 4N HCl solution in dioxane (0.5 mL, 2.03 mmol, 10.0 eq.) and the mixture was stirred at RT for 6 hours. The mixture was concentrated in vacuo, EtOAc was added and the solution was washed with Sat. NaHCO3, the aqueous layer was re-extracted with EtOAc (×2). The organic phases were combined, dried (MgSO4), filtered and concentrated in vacuo to give the title compound as a colourless glass. (0.075 g, 93% yield).1H NMR (400 MHz, CDCl3) δ 8.14-8.13 (m, 1H), 7.52-7.41 (m, 3H), 7.15 (d, J=9.0 Hz, 1H), 6.61-6.53 (m, 1H), 6.27 (d, J=8.4 Hz, 1H), 4.11 (s, 2H), 4.01 (s, 2H), 3.71 (s, 2H), 3.57-3.50 (m, 1H), 3.50 (s, 2H), 2.85 (dd, J=8.2, 12.8 Hz, 2H), 2.70-2.61 (m, 2H) Step 3: Synthesis of 8-chloro-5-(oxetan-3-yl)-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 57) A solution of 8-chloro-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (0.025 g, 0.06 mmol, 1.0 eq.), 3-oxetanone (35 μL, 0.51 mmol, 8.0 eq.) in MeOH (0.5 mL) was heated to 70° C. for 6 hours. The mixture was cooled to 0° C. and sodium cyanoborohydride (0.012 g, 0.19 mmol, 3.0 eq.) was added and the mixture stirred at RT overnight. The mixture was concentrated in vacuo, diluted with DCM, passed through a phase separator and concentrated in vacuo. The residue was purified by preparative HPLC. (0.005 g, 17% yield).1H NMR (400 MHz, CDCl3) δ 8.16-8.11 (m, 2H), 7.56-7.51 (m, 1H), 7.50-7.45 (m, 1H), 7.16 (d, J=8.6 Hz, 1H), 6.64-6.60 (m, 1H), 6.29 (d, J=8.3 Hz, 1H), 4.77-4.65 (m, 4H), 4.16 (s, 2H), 4.04 (s, 2H), 3.97-3.90 (m, 1H), 3.63 (s, 2H), 3.57-3.47 (m, 1H), 3.36-3.24 (m, 2H), 2.86 (dd, J=8.3, 12.6 Hz, 2H), 2.71-2.62 (m, 2H). m/z 449 (M+H)+. Example 8 A mixture of 8-chloro-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine dihydrochloride (0.050 g, 0.11 mmol, 1.0 eq.), Et3N (30 μL, 0.21 mmol, 2.0 eq.), fluoroacetone (62 μL, 0.86 mmol, 8.0 eq.) in MeOH (1 mL) was heated to 70° C. for 2.5 hours. Mixture was cooled to 0° C. and sodium borohydride (0.02 g, 0.32 mmol, 3 eq.) was added, mixture was stirred at RT for 2 hours. Additional sodium borohydride (0.02 g, 0.32 mmol, 3 eq.), Et3N (30 μL, 0.21 mmol, 2.0 eq.), fluoroacetone (62 μL, 0.86 mmol, 8.0 eq.) were added and the mixture was stirred at RT for 3 days. The mixture was concentrated in vacuo, diluted with DCM, passed through a phase separator and concentrated in vacuo. The residue was purified by preparative HPLC. To give the title compound (0.0147 g, 30% yield).1H NMR (400 MHz, CDCl3) δ 8.13 (dd, J=1.0, 5.1 Hz, 1H), 7.54 (d, J=2.3 Hz, 1H), 7.49 (dd, J=2.4, 8.5 Hz, 1H), 7.46-7.41 (m, 1H), 7.14 (d, J=8.6 Hz, 1H), 6.60 (dd, J=5.4, 6.7 Hz, 1H), 6.27 (d, J=8.3 Hz, 1H), 4.62-4.41 (m, 2H), 4.12 (s, 2H), 4.02 (s, 2H), 3.87 (s, 2H), 3.62 (s, 2H), 3.57-3.48 (m, 1H), 3.22-3.11 (m, 1H), 2.87 (dd, J=8.2, 12.8 Hz, 2H), 2.71-2.63 (m, 2H), 1.22 (dd, J=1.5, 6.8 Hz, 3H). m/z 453 (M+H)+. Example 9 A mixture of 8-chloro-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine in MeCN (1 mL) was added K2CO3(0.018 g, 0.13 mmol, 2.0 eq.) followed by fluoro-2-iodoethane (6 μL, 0.08 mmol, 2.0 eq.) and stirred at RT for 2 hours. Mixture was then heated to 80° C. overnight. Mixture was diluted with NaHCO3/DCM, passed through a phase separator and concentrated in vacuo. The residue was purified by preparative HPLC. to give the title compound as white solid (0.0124 g, 44% yield).1H NMR (400 MHz, CDCl3) δ 8.14 (dd, J=1.0, 5.1 Hz, 1H), 7.53-7.49 (m, 2H), 7.46-7.41 (m, 1H), 7.18-7.14 (m, 1H), 6.59 (dd, J=5.1, 6.3 Hz, 1H), 6.27 (d, J=8.6 Hz, 1H), 4.70 (dd, J=4.7, 4.7 Hz, 1H), 4.58 (dd, J=4.8, 4.8 Hz, 1H), 4.12 (s, 2H), 4.02 (s, 2H), 3.77 (s, 2H), 3.56-3.51 (m, 3H), 3.00-2.83 (m, 4H), 2.71-2.63 (m, 2H). m/z 439 (M+H)+. Example 10 Step 1: Synthesis of 2-(4-fluoropyridin-2-yl)-2-azaspiro[3.3]heptane-6-carbohydrazide A solution of methyl 2-(4-fluoropyridin-2-yl)-2-azaspiro[3.3]heptane-6-carboxylate [prepared in a similar manner as Ex6 step 4 using 2-bromo-5-fluoropyridine instead of 2-bromopyridine] (510 mg, 2.11 mmol, 1.0 eq.) in EtOH (10 mL) was added dropwise to a mixture of hydrazine monohydrate (0.62 mL, 12.7 mmol, 6.0 eq.) in EtOH. Mixture was heated to 50° C. for 1 hours, diluted with EtOAc, washed with brine and concentrated in vacuo. To give the title compound as a pink solid. (365 mg, 69% yield).1H NMR (400 MHz, CDCl3) δ 7.97 (d, J=3.0 Hz, 1H), 7.93 (s, 1H), 7.25-7.18 (m, 1H), 6.23 (dd, J=3.5, 9.1 Hz, 1H), 4.00 (s, 2H), 3.94 (s, 2H), 3.85-3.76 (m, 2H), 2.96-2.86 (m, 1H), 2.57-2.50 (m, 2H), 2.44-2.37 (m, 2H). Step 2: Synthesis of tert-butyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate The title compound was prepared in the manner of Example 7 step 1 (189 mg, 67% yield).1H NMR (400 MHz, CDCl3) δ 8.00-7.99 (m, 1H), 7.58-7.45 (m, 2H), 7.26-7.17 (m, 2H), 6.23 (dd, J=5, 6.3 Hz, 1H), 4.66-4.20 (m, 4H), 4.02 (s, 2H), 3.98 (s, 2H), 3.52-3.48 (m, 1H), 2.88-2.85 (m, 2H), 2.66-2.55 (m, 2H), 1.62 (s, 9H). Step 3: Synthesis of 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 60) The title compound was prepared in the manner of Example 7 step 2 (5.2 mg, 97% yield).1H NMR (400 MHz, DMSO) δ 8.08 (dd, J=1.0, 5.0 Hz, 1H), 7.71 (s, 1H), 7.65 (dd, J=2.2, 8.1 Hz, 1H), 7.55-7.50 (m, 2H), 6.43 (dd, J=5.0, 6.1 Hz, 1H), 4.02 (s, 2H), 3.89 (s, 2H), 3.76 (s, 2H), 3.73-3.67 (m, 1H), 3.58 (s, 2H), 2.65-2.56 (m, 4H). m/z 411 (M+H)+. Example 11 To a cooled suspension of 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4,5-dihydro-6H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-one (prepared as in Example 7, step 2) (77 mg, 0.19 mmol, 1.0 eq.), Et3N (0.091 mL, 0.65 mmol, 3.5 eq.), at 0° C. in THE (5 mL) was added methyl chloroformate (0.022 mL, 0.28 mMol, 1.5 eq.) dropwise. The resulting thick suspension was stirred at RT for 1 hour, diluted with EtOAc, washed with an aq. sol. of sat. NaHCO3, dried, concentrated in vacuo. and the residue was purified by preparative HPLC to give the title compound (20.48 mg, 23% yield).1H NMR (400 MHz, DMSO) δ 8.03 (d, J=3.0 Hz, 1H), 7.69 (dd, J=2.5, 8.5 Hz, 2H), 7.54 (d, J=8.5 Hz, 1H), 7.50-7.44 (m, 1H), 6.39 (dd, J=3.3, 9.1 Hz, 1H), 4.50 (s, 2H), 4.33 (s, 2H), 3.98 (s, 2H), 3.85 (s, 2H), 3.71-3.67 (m, 4H), 2.58 (d, J=8.2 Hz, 4H). m/z 469 (M+H)+. Compounds 62 to 91 Compound Nos. 62 to 75, 78, 79, 81, 82, and 90 were prepared according to the methods set forth in Example 11 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 6. Compound Nos. 76, 77, 80, and 83 to 89 are prepared according to the procedure of Example 11 using appropriately substituted intermediates. TABLE 6Compound Nos. 62 to 75, 78, 79, 81, 82, and 90Compound No.Analytical DataSynthesis Method621H NMR (400 MHz, DMSO) δ 8.03-8.01 (m,Example 111H), 7.64-7.61 (m, 2H), 7.50-7.41 (m, 2H),6.56 (dd, J = 5.3, 6.8 Hz, 1H), 6.30 (d, J = 8.4 Hz,1H), 4.45 (s, 2H), 4.28 (s, 2H), 3.99 (s, 2H), 3.88(s, 2H), 3.67-3.63 (m, 1H), 2.60 (d, J = 8.0 Hz,4H), 1.47 (s, 9H). m/z 493 (M + H)+.631H NMR (400 MHz, DMSO) δ 8.00 (d, J = 3.0Example 11Hz, 1H), 7.76-7.76 (m, 1H), 7.66 (dd, J = 2.4, 8.5Hz, 1H), 7.52 (d, J = 8.5 Hz, 1H), 7.43-7.37 (m,1H), 6.37 (dd, J = 3.5, 9.0 Hz, 1H), 4.61 (s, 2H),4.45 (s, 2H), 4.01 (s, 2H), 3.90 (s, 2H), 3.71-3.70 (m, 1H), 3.10 (dd, J = 6.5, 6.5 Hz, 1H), 2.64(d, J = 8.0 Hz, 4H), 1.12 (d, J = 6.8 Hz, 6H). m/z481 (M + H)+.641H NMR (400 MHz, CDCl3) δ 8.00 (1H, d, J = 3.0Example 11Hz), 7.60-7.51 (2H, m), 7.25-7.18 (2H, m),6.24 (1H, dd, J = 3.4, 9.0 Hz), 4.70-4.61 (4H, m),4.08 (2H, s), 4.00 (2H, s), 3.55-3.46 (1H, m),2.87 (2H, dd, J = 8.1, 12.6 Hz), 2.71-2.66 (2H,m), 2.28 (1H, s), 2.20 (2H, s). m/z 453 (M + H)+.651H NMR (400 MHz, DMSO) δ 8.04-8.02 (m,Example 111H), 7.71 (s, 1H), 7.64 (dd, J = 2.4, 8.5 Hz, 1H),7.51 (d, J = 8.5 Hz, 1H), 7.46-7.42 (m, 1H), 6.56(dd, J = 5.0, 6.8 Hz, 1H), 6.30 (d, J = 8.3 Hz, 1H),4.68 (s, 2H), 4.53 (s, 2H), 4.00 (s, 2H), 3.89 (s,2H), 2.62 (d, J = 8.2 Hz, 4H), 2.54 (dd, J = 1.8, 1.8Hz, 1H), 2.11-2.07 (m, 1H), 0.84-0.82 (m,4H). m/z 461 (M + H)+.661H NMR (400 MHz, DMSO) δ 8.57 (d, J = 3.3Example 11Hz, 1H), 8.14 (dd, J = 2.9, 92.1 Hz, 1H), 7.77-7.70 (m, 1H), 7.63-7.58 (m, 1H), 7.39 (dd,J = 4.4, 9.0 Hz, 1H), 6.80 (dd, J = 1.3, 9.1 Hz, 1H),4.72 (s, 1H), 4.52-4.47 (m, 3H), 4.17 (s, 2H),4.04 (s, 2H), 3.81-3.71 (m, 1H), 2.67 (d, J = 7.8Hz, 4H), 2.28 (s, 2H), 2.17 (s, 1H). m/z 436(M + H)+.671H NMR (400 MHz, DMSO) δ 8.03-8.02 (m,Example 111H), 7.73 (s, 1H), 7.63 (dd, J = 2.4, 8.5 Hz, 1H),7.51-7.41 (m, 2H), 6.56 (dd, J = 5.3, 7.1 Hz, 1H),6.30 (d, J = 8.3 Hz, 1H), 4.57 (s, 2H), 4.40 (s,2H), 3.99 (s, 2H), 3.89 (s, 2H), 2.61 (d, J = 8.2Hz, 4H), 2.45 (d, J = 1.6 Hz, 1H), 1.09-1.06 (m,3H). m/z 489 (M + H)+.681H NMR (400 MHz, DMSO) δ 8.09 (dd, J = 1.0,Example 115.1 Hz, 1H), 7.80 (s, 1H), 7.74 (dd, J = 2.4, 8.5Hz, 1H), 7.60-7.50 (m, 2H), 6.65 (dd, J = 4.9, 6.2Hz, 1H), 6.39 (d, J = 8.3 Hz, 1H), 4.54 (s, 2H),4.43-4.40 (m, 2H), 4.18 (q, J = 7.1 Hz, 2H), 4.04(s, 2H), 3.91 (s, 2H), 3.78-3.69 (m, 1H), 2.67-2.59 (m, 4H), 1.32-1.29 (m, 3H). m/z 465(M + H)+.691H NMR (400 MHz, DMSO) δ 8.00 (d, J = 3.0Example 11Hz, 1H), 7.67-7.64 (m, 2H), 7.52-7.49 (m,1H), 7.43-7.37 (m, 1H), 6.37 (dd, J = 3.5, 9.0 Hz,1H), 4.53 (s, 2H), 4.36 (s, 2H), 4.18 (q, J = 7.0Hz, 2H), 4.00 (s, 2H), 3.90 (s, 2H), 3.04 (s, 1H),2.63 (d, J = 8.0 Hz, 4H), 1.29 (dd, J = 7.0, 7.0 Hz,3H). m/z 483 (M + H)+.701H NMR (400 MHz, DMSO) δ 8.03-8.02 (m,Example 111H), 7.65-7.62 (m, 2H), 7.50-7.41 (m, 2H),6.56 (dd, J = 5.1, 6.9 Hz, 1H), 6.30 (d, J = 8.3 Hz,1H), 4.50 (s, 2H), 4.33 (s, 2H), 3.99 (s, 2H), 3.88(s, 2H), 3.76 (s, 3H), 3.70-3.62 (m, 1H), 2.60(d, J = 8.2 Hz, 4H). m/z 451 (M + H)+.711H NMR (400 MHz, DMSO) δ 8.04 (d, J = 4.8Example 11Hz, 1H), 7.78-7.75 (m, 1H), 7.66 (dd, J = 2.3, 8.5Hz, 1H), 7.52 (d, J = 8.8 Hz, 1H), 7.49-7.44 (m,1H), 6.61-6.56 (m, 1H), 6.32 (d, J = 8.3 Hz, 1H),4.60 (s, 2H), 4.44 (s, 2H), 4.02 (s, 2H), 3.91 (s,2H), 3.73-3.69 (m, 1H), 3.09 (dd, J = 6.5, 6.5Hz, 1H), 2.66-2.61 (m, 4H), 1.12 (d, J = 6.7 Hz,6H). m/z 463 (M + H)+.721H NMR (400 MHz, DMSO) δ 8.00 (1H, d,Example 11J = 2.5 Hz), 7.76 (1H, s), 7.65 (1H, dd, J = 2.3, 8.6Hz), 7.51 (1H, d, J = 8.5 Hz), 7.43-7.37 (1H, m),6.36 (1H, dd, J = 3.5, 9.0 Hz), 4.59 (2H, s), 4.42(2H, s), 4.00 (2H, s), 3.90 (2H, s), 3.72-3.67(1H, m), 2.62 (4H, d, J = 8.0 Hz), 2.51-2.49 (m,2H), 1.10 (3H, dd, J = 7.3, 7.3 Hz). m/z 467(M + H)+.731H NMR (400 MHz, DMSO) δ 8.03-8.02 (m,Example 111H), 7.74-7.74 (m, 1H), 7.63 (dd, J = 2.4, 8.5 Hz,1H), 7.51-7.41 (m, 2H), 6.56 (dd, J = 5.1, 6.9 Hz,1H), 6.30 (d, J = 8.3 Hz, 1H), 4.57 (s, 2H), 4.40(s, 2H), 3.99 (s, 2H), 3.89 (s, 2H), 3.68-3.67 (m,1H), 2.61 (d, J = 8.2 Hz, 4H), 2.16 (s, 3H).). m/z435 (M + H)+.741H NMR (400 MHz, DMSO) δ 8.00 (d, J = 2.9Example 11Hz, 1H), 7.73 (s, 1H), 7.66 (dd, J = 2.3, 8.5 Hz,1H), 7.53 (d, J = 8.5 Hz, 1H), 7.43-7.37 (m, 1H),6.37 (dd, J = 3.6, 9.0 Hz, 1H), 4.70 (s, 2H), 4.55(s, 2H), 4.01 (s, 2H), 3.91 (s, 2H), 3.70-3.62 (m,1H), 2.64 (d, J = 8.0 Hz, 4H), 2.11 (dd, J = 6.1, 6.1Hz, 1H), 0.86 (d, J = 5.8 Hz, 4H). m/z 479(M + H)+.751H NMR (400 MHz, DMSO) δ 8.57 (dd, J = 1.5,Example 114.5 Hz, 1H), 7.82-7.77 (m, 1H), 7.74 (dd, J = 2.5,8.6 Hz, 1H), 7.59 (d, J = 8.6 Hz, 1H), 7.39 (dd,J = 4.4, 9.0 Hz, 1H), 6.79 (dd, J = 1.4, 9.0 Hz, 1H),4.55 (s, 2H), 4.41 (s, 2H), 4.16 (s, 2H), 4.03 (s,2H), 3.37 (d, J = 1.3 Hz, 3H), 2.66 (d, J = 8.1 Hz,4H), 2.13 (s, 1H). m/z 452 (M + H)+.781H NMR (400 MHz, DMSO) d 8.04 (d, J = 3.0Example 11Hz, 1H), 7.77-7.74 (m, 1H), 7.70 (dd, J = 2.4, 8.5Hz, 1H), 7.55 (d, J = 8.4 Hz, 1H), 7.51-7.45 (m,1H), 6.40 (dd, J = 3.5, 9.0 Hz, 1H), 5.06-4.95 (m,1H), 4.64-4.47 (m, 6H), 3.99 (s, 2H), 3.86 (s,2H), 3.74-3.64 (m, 1H), 2.60-2.55 (m, 4H),1.25 (d, J = 6.1 Hz, 3H). m/z 516 (M + H)+.791H NMR (400 MHz, CDCl3) d 8.00 (d, J = 2.9Example 11Hz, 1H), 7.58-7.54 (m, 2H), 7.25-7.19 (m,2H), 6.24 (dd, J = 3.1, 9.0 Hz, 1H), 5.24-5.10 (m,1H), 4.76-4.68 (m, 4H), 4.64-4.56 (m, 4H),4.08 (s, 2H), 4.00 (s, 2H), 3.55-3.46 (m, 1H),2.87 (dd, J = 8.1, 12.4 Hz, 2H), 2.68 (s, 2H). m/z534 (M + H)+.811H NMR (400 MHz, DMSO) d 8.03 (dd, J = 1.5,Example 112.8 Hz, 1H), 7.86-7.82 (m, 2H), 7.79 (s, 1H),7.73 (dd, J = 2.4, 8.6 Hz, 1H), 7.58 (d, J = 8.5 Hz,1H), 4.61-4.58 (m, 4H), 4.12 (s, 2H), 4.00 (s,2H), 3.78-3.69 (m, 1H), 2.63 (d, J = 7.9 Hz, 4H),1.46 (d, J = 19.8 Hz, 2H), 1.32-1.25 (m, 2H). m/z480 (M + H)+.821H NMR (400 MHz, DMSO) d 8.03 (dd, J = 1.5,Example 112.6 Hz, 1H), 7.87-7.82 (m, 3H), 7.73 (ddd,J = 2.4, 4.2, 8.6 Hz, 1H), 7.58 (d, J = 8.5 Hz, 1H),7.21-6.82 (m, 1H), 4.56-4.55 (m, 4H), 4.12 (s,2H), 4.00 (s, 2H), 3.76-3.69 (m, 1H), 2.63 (d,J = 7.5 Hz, 4H). m/z 472 (M + H)+.901H NMR (400 MHz, DMSO) d 7.95 (d, J = 2.8Example 11Hz, 1H), 7.67 (d, J = 1.8 Hz, 1H), 7.61 (dd, J = 2.4,8.5 Hz, 1H), 7.47-7.36 (m, 2H), 6.31 (dd, J = 3.5,9.1 Hz, 1H), 4.48-4.21 (m, 4H), 4.00 (d, J = 2.8Hz, 1H), 3.89 (s, 2H), 3.79 (s, 2H), 3.64-3.55(m, 1H), 2.43 (dd, J = 1.8, 1.8 Hz, 4H), 0.62-0.61(m, 4H). m/z 495 (M + H)+. Example 12 Step 1: Synthesis of tert-butyl (2-amino-5-chlorobenzyl)glycinate To a solution of 2-amino-5-chlorobenzylamine (3.0 g, 19.16 mmol, 1.0 eq.), tert-butylbromoacetate (2.97 mL, 20.11 mmol, 1.05 eq.) in THE (60 mL) was added Et3N (3.20 mL, 22.99 mmol, 1.2 eq.) and the mixture was stirred at room temperature overnight. The mixture was diluted with EtOAc, water and the layers separated. The aq. phase was extracted with EtOAc, the organic phases were combined, dried (MgSO4), filtered and concentrated in vacuo to afford the title compound as a yellow solid (5.09 g, 98% yield).1H NMR (400 MHz, CDCl3) δ 7.06-7.01 (m, 1H), 6.99 (d, J=2.4 Hz, 1H), 6.57 (d, J=8.4 Hz, 1H), 3.73 (s, 2H), 3.27 (s, 2H), 1.48 (s, 9H). Step 2: Synthesis of 7-chloro-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepin-2-one To a solution of tert-butyl (2-amino-5-chlorobenzyl)glycinate in THE (90 mL) was slowly added a solution of KOtBu (2.53 g, 22.56 mmol, 1.2 eq.) in THE (60 mL). The mixture was stirred at RT for 90 minutes, diluted with sat. sol. of NH4Cl and extracted with EtOAc (×3). The organic phases were combined, dried (MgSO4), filtered and concentrated in vacuo to afford the title compound as a yellow powder (3.11 g, 84% yield).1H NMR (400 MHz, CDCl3) δ 7.57 (s, 1H), 7.23-7.17 (m, 2H), 6.83 (d, J=8.4 Hz, 1H), 4.00 (s, 2H), 3.73 (s, 2H). Step 3: Synthesis of tert-butyl 7-chloro-2-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate To a cooled suspension of 7-chloro-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepin-2-one (3.11 g, 15.82 mmol, 1.0 eq.) in THE (75 mL) at 0° C. was added a solution of di-tert-butyl dicarbonate (4.14 g, 18.98 mmol, 1.2 eq.) in THE (35 mL) dropwise over 10 minutes. Mixture was allowed to warm to RT and stirred overnight. The mixture was concentrated in vacuo and triturated with diethyl ether to afford the title compound as a yellow powder (3.36 g, 72% yield).1H NMR (400 MHz, DMSO) δ 7.36-7.31 (m, 2H), 7.16 (d, J=8.6 Hz, 2H), 4.53-4.45 (m, 2H), 4.33 (s, 2H), 1.31 (s, 9H). Step 4: Synthesis of tert-butyl 7-chloro-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate To a suspension of tert-butyl 7-chloro-2-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (3.36 g, 11.32 mmol, 1.0 eq.) in THE (80 mL) was added 2,4-bis(4-methoxyphenyl)-2,4-dithioxo-1,3,2,4-dithiadiphosphetane (2.75 g, 6.79 mmol, 0.60 eq.) and the mixture was heated to reflux for 90 minutes. The mixture was allowed to cool, concentrated in vacuo, triturated with TBME and filtered. The solid was discarded and the filtrate was concentrated in vacuo to afford the title compound as a pale yellow solid (2.07 g, 58% yield).1H NMR (400 MHz, DMSO) δ 12.08-12.07 (m, 1H), 7.41-7.38 (m, 1H), 7.33 (dd, J=2.5, 8.6 Hz, 1H), 7.20-7.19 (m, 1H), 4.39 (s, 1H), 4.35 (s, 2H), 4.30 (s, 1H), 1.27 (d, J=22.0 Hz, 9H). Step 5: Synthesis of 7-chloro-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepine-2-thione To a solution of tert-butyl 7-chloro-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (1.08 g, 3.45 mmol, 1.0 eq.) in DCM (30 mL) was added TFA (10 mL) and the mixture was stirred at RT for 1 hour and concentrated in vacuo to afford the title compound as a pale yellow powder (0.737 g, quant. yield).1H NMR (400 MHz, DMSO) δ 12.02 (s, 1H), 7.46-7.40 (m, 2H), 7.27 (d, J=8.3 Hz, 1H), 3.87 (s, 2H), 3.69 (s, 2H), 3.51-3.50 (m, 1H). Step 6: Synthesis of tert-butyl 6-(8-chloro-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate The procedure of Example 1, step 2 was employed to afford the title compound as a foam (1.06 g, 94% yield).1H NMR (400 MHz, CDCl3) δ 7.72 (s, 1H), 7.51-7.47 (m, 1H), 7.12 (d, J=3.9 Hz, 2H), 3.99 (s, 2H), 3.94-3.90 (m, 4H), 3.68 (s, 2H), 3.49-3.40 (m, 1H), 2.76 (dd, J=8.2, 12.8 Hz, 2H), 2.63-2.54 (m, 2H), 1.43 (s, 9H) Step 7: Synthesis of methyl 1-(2-(tert-butoxycarbonyl)-2-azaspiro[3.3]heptan-6-yl)-8-chloro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate The procedure of Example 11 was employed to afford the title compound which was used without further purification.1H NMR (400 MHz, CDCl3) δ 7.59-7.56 (m, 1H), 7.53 (dd, J=2.4, 8.2 Hz, 1H), 7.15 (d, J=8.5 Hz, 1H), 4.01 (s, 4H), 4.00 (s, 2H), 3.93 (s, 2H), 3.79 (m, 3H), 3.42 (dd, J=8.3, 8.3 Hz, 1H), 2.78 (dd, J=8.1, 12.5 Hz, 2H), 2.62-2.59 (m, 2H), 1.43 (s, 9H). Step 8: Synthesis of methyl 8-chloro-1-(2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]-triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate The procedure of Example 1, step 3, was employed to afford the title compound (400, mg, 81% yield).1H NMR (400 MHz, CDCl3) δ 7.53-7.51 (m, 2H), 7.19-7.17 (m, 1H), 4.62-4.40 (m, 4H), 3.76 (s, 3H), 3.71 (s, 2H), 3.63 (s, 2H), 3.48-3.46 (m, 1H), 2.75-2.70 (m, 2H), 2.63-2.55 (m, 2H). Step 9: Synthesis of methyl 8-chloro-1-(2-(6-cyanopyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (Compound No. 91) The procedure of Example 1, step 4 using 6-bromo-2-pyridinecarbonitrile was employed to afford the title compound after preparative HPLC as a yellow film (0.0134 g, 21% yield).1H NMR (400 MHz, DMSO) δ 7.68-7.60 (m, 3H), 7.51 (d, J=8.3 Hz, 1H), 7.10 (d, J=7.2 Hz, 1H), 6.63 (d, J=8.5 Hz, 1H), 4.53 (s, 2H), 4.36 (s, 2H), 4.09 (s, 2H), 4.00 (s, 2H), 3.71 (s, 3H), 3.70-3.68 (m, 1H), 2.65 (d, J=8.0 Hz, 4H). m/z 476 (M+H)+. Compound Nos. 92 to 94 Compound Nos. 92 to 94 were prepared according to the methods set forth in Example 12. For example, Compound No. 92 of Table 7 lists the method of “Example 12”, indicating that this compound was prepared according to the procedure of Example 12 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 7. TABLE 7Compound Nos. 92 to 94Compound No.Analytical DataSynthesis Method921H NMR (400 MHz, DMSO) δ 8.34 (s, 2H),Example 127.68-7.64 (m, 2H), 7.50 (dd, J = 1.4, 8.0 Hz, 1H),4.53 (s, 2H), 4.36 (s, 2H), 4.12 (s, 2H), 4.01 (s,2H), 3.74 (s, 3H), 3.73-3.65 (m, 1H), 2.63 (d,J = 8.0 Hz, 4H). m/z 452 (M + H)+.931H NMR (400 MHz, DMSO) δ 8.01-7.99 (m,Example 121H), 7.82-7.79 (m, 3H), 7.66 (dd, J = 2.4, 8.5 Hz,1H), 7.52 (d, J = 8.5 Hz, 1H), 4.61 (s, 2H), 4.42(s, 2H), 4.13 (s, 2H), 4.03 (s, 2H), 3.72-3.68(m, 1H), 2.66 (d, J = 8.2 Hz, 4H), 2.19 (s, 3H).m/z 436 (M + H)+.941H NMR (400 MHz, DMSO) δ 7.98 (d, J = 2.4Example 12Hz, 1H), 7.33-7.19 (m, 4H), 6.24-6.20 (m,1H), 4.15 (s, 2H), 4.08 (s, 2H), 3.96 (s, 2H), 3.77(s, 2H), 3.75-3.65 (m, 1H), 2.64-2.59 (m, 4H),2.39 (s, 3H). m/z 460 (M + H)+. Example 13 A mixture of methyl 8-chloro-1-(2-azaspiro[3.3]heptan-6-yl)-4Hbenzo[f][1,2,4] triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (Example 2 step 8 above) (0.030 g, 0.0802 mmol, 1.0 eq.), 2-chloropyrimidine (0.0092 g, 0.0802 mmol, 1.0 eq.), cesium carbonate (0.031 g, 0.0963 mmol, 1.2 eq.) in DMF was heated 80° C. in a reaction tube for 4 hours. The mixture was diluted with water and extracted with EtOAc (×3). The organic phases were combined washed with brine, dried (MgSO4), filtered and concentrated in vacuo to afford the title compound (7.6 mg).1H NMR (400 MHz, DMSO) δ 8.30 (d, J=4.6 Hz, 2H), 7.68-7.64 (m, 2H), 7.51 (d, J=8.7 Hz, 1H), 6.61 (dd, J=4.8, 4.8 Hz, 1H), 4.53 (s, 2H), 4.36 (s, 2H), 4.12 (s, 2H), 4.01 (s, 2H), 3.74 (s, 3H), 3.73-3.65 (m, 1H), 2.64 (d, J=8.0 Hz, 4H). m/z 452 (M+H)+. Example 14 Step 1: Synthesis of isopropyl 7-chloro-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate To a cooled mixture of 7-chloro-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepine-2-thione as prepared by Example 12, step 5 (0.06 g, 0.28 mmol, 1.0 eq.), Et3N (59 μL, 0.42 mmol, 1.5 eq.) at 0° C. in DCM was added isopropyl chloroformate (0.31 mL, 0.31 mmol, 1.10 eq.) and the mixture was stirred at RT overnight. Mixture was diluted with DCM, Sat. NaHCO3and passed through a phase separator and concentrated in vacuo to afford the title compound as a yellow gum. (90 mg, quant. yield).1H NMR (400 MHz, CDCl3) δ 7.33-7.30 (m, 2H), 6.97 (d, J=8.1 Hz, 1H), 4.97-4.90 (m, 1H), 4.60-4.49 (m, 4H), 1.25-1.18 (m, 6H). Step 2: Synthesis of isopropyl 8-chloro-1-(2-(pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (Compound No. 96) The procedure of Example 7, step 2 was employed to afford the title compound as a yellow gum. (23 mg).1H NMR (400 MHz, DMSO) δ 8.06-8.04 (m, 1H), 7.68-7.64 (m, 2H), 7.53-7.44 (m, 2H), 6.59 (dd, J=5.0, 6.8 Hz, 1H), 6.33 (d, J=8.4 Hz, 1H), 4.95-4.85 (m, 1H), 4.52 (s, 2H), 4.34 (s, 2H), 4.02 (s, 2H), 3.91 (s, 2H), 3.73-3.65 (m, 1H), 2.63 (d, J=8.2 Hz, 4H), 1.30 (d, J=6.3 Hz, 6H). m/z 479 (M+H)+. Example 15 Step 1: Synthesis of tert-butyl (E)-6-(chloro(hydroxyimino)methyl)-2-azaspiro[3.3]heptane-2-carboxylate To a cooled solution of tert-butyl 6-formyl-2-azaspiro[3.3]heptane-2-carboxylate (0.8 g, 3.6 mmol, 1.0 eq.) in MeOH (4 mL), water (4 mL) at 0° C. was added Na2CO3(0.188 g, 1.8 mmol, 0.5 eq.) followed by hydroxylamine hydrochloride (0.283 mg, 4.1 mmol, 1.15 eq.) and stirred at °) C for 2 hours. The mixture was then stirred at RT for 3 days, concentrated in vacuo, partitioned between EtOAc and H2O. The organic phase was isolated, concentrated in vacuo the residue dissolved in DMF (8 mL), NCS (0.474 g, 3.6 mmol, 1.0 eq) was added and the mixture was stirred at RT overnight. The mixture was diluted with EtOAc and 4% aq. sol. of LiCl and stirred at RT overnight. The organic layer was separated and concentrated in vacuo to afford the title compound (1.29 g, quant.).1H NMR (400 MHz, CDCl3) δ 8.02 (s, 1H), 3.96 (s, 2H), 3.87 (s, 2H), 3.25-3.16 (m, 1H), 2.96-2.88 (m, 3H), 2.77 (s, 1H), 1.42 (s, 9H). Step 2: Synthesis of tert-butyl 6-(5-(bromomethyl)isoxazol-3-yl)-2-azaspiro[3.3]heptane-2-carboxylate To a cooled mixture of tert-butyl (E)-6-(chloro(hydroxyimino)methyl)-2-azaspiro[3.3]heptane-2-carboxylate (0.989 g, 3.6 mmol, 1.0 eq.), propargyl bromide (80% sol. in toluene, 481.2 μL, 4.3 mmol, 1.2 eq) at 0° C. in DCM (10 mL) was added Et3N (607.1 μL, 4.3 mmol, 1.2 eq.) in DCM dropwise. The mixture was allowed to warm to RT and stirred overnight. Mixture was diluted with DCM, washed with water and concentrated in vacuo to afford the title compound as a light brown oil (1.05 g, 81% yield). Step 3: Synthesis of 1-(5-chloro-2-iodophenyl)-N-methylmethanamine To a solution of 5-chloro-2-iodobenzaldehyde (1.0 g, 3.8 mmol, 1.0 eq.) in anhydrous MeOH (20 mL) was added NaHCO3(0.95 g, 11.3 mmol, 3.0 eq.), methylamine (33% sol. in EtOH) (0.93 mL, 7.5 mmol, 2.0 eq.) and the mixture was refluxed for 4 hours. Mixture was cooled to 0° C., sodium borohydride (0.17 g, 4.5 mmol, 1.20 eq.) was added portion wise, the mixture was allowed to warm to RT and stirred at RT overnight. The reaction was quenched with H2O (2 mL) and concentrated in vacuo to give a residue. This was partitioned between DCM and brine. The organic phase was separated and the aq. layer was extracted with DCM (×2). The organic phases were combined, dried (MgSO4), concentrated in vacuo to afford the title compound as a light yellow oil (0.97 g, 91% yield).1H NMR (400 MHz, CDCl3) δ 7.68 (d, J=8.3 Hz, 1H), 7.36 (d, J=2.8 Hz, 1H), 6.92 (dd, J=2.5, 8.3 Hz, 1H), 3.69 (s, 2H), 2.44 (s, 3H). Step 4: Synthesis of tert-butyl 6-(5-(((5-chloro-2-iodobenzyl)(methyl)amino)methyl)isoxazol-3-yl)-2-azaspiro[3.3]heptane-2-carboxylate To a solution of tert-butyl 6-(5-(bromomethyl)isoxazol-3-yl)-2-azaspiro[3.3]heptane-2-carboxylate (0.5 g, 1.4 mmol, 1.0 eq) in THE (5 mL) was added 1-(5-chloro-2-iodophenyl)-N-methylmethanamine (0.433 g, 2.1 mmol, 1.1 eq.), potassium carbonate (0.29 g, 2.1 mmol, 1.5 eq.) and stirred at RT for 48 hours. The mixture was diluted with DCM, washed with brine and concentrated in vacuo. The residue was purified using a Biotage® KP-NH cartridge, eluting with isohexane, isohexane:EtOAc (75%:25%) followed by 100% EtOAc, to afford the title compound (0.36 g, 46% yield).1H NMR (400 MHz, CDCl3) δ 7.74 (d, J=8.3 Hz, 1H), 7.46 (d, J=2.5 Hz, 1H), 6.97 (dd, J=2.5, 8.3 Hz, 1H), 6.03 (s, 1H), 4.02 (s, 2H), 3.88 (s, 2H), 3.75 (s, 2H), 3.57 (s, 2H), 3.48-3.39 (m, 1H), 2.63-2.55 (m, 2H), 2.48-2.39 (m, 2H), 2.33 (s, 3H). Step 5: Synthesis of tert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[c]isoxazolo[4,5-e]azepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate A mixture of tert-butyl 6-(5-(((5-chloro-2-iodobenzyl)(methyl)amino)-methyl)isoxazol-3-yl)-2-azaspiro[3.3]heptane-2-carboxylate (0.18 g, 0.3 mmol, 1.0 eq.), potassium carbonate (0.178 g, 1.3 mmol, 4.0 eq.), pivalic acid (0.0098 g, 0.1 mmol, 0.3 eq.), triphenylphosphine (0.0253 mg, 0.1 mmol, 0.3 eq.) in DMA (2 mL) was degassed using N2for 2 hours. Palladium acetate (0.0072 g, 0.0032 mmol, 0.1 eq.) was added and the reaction was heated to 80° C. for 2 hours, 100° C. for 2 hour, and 110° C. for 2 hours. The mixture was allowed to cool overnight. The mixture was filtered through a pad of celite, the filtrate was diluted with EtOAc, washed with 4% aq. sol. of LiCl (×4). The organic phase was concentrated in vacuo, the residue was purified using a Biotage® KP-NH cartridge, eluting with isohexane, isohexane:EtOAc (70%:30%) to afford the title compound (0.71 g, 51% yield).1H NMR (400 MHz, CDCl3) δ 7.31 (dd, J=2.3, 8.3 Hz, 1H), 7.26-7.23 (m, 1H), 7.20 (d, J=2.3 Hz, 1H), 4.17 (s, 2H), 4.03 (s, 2H), 3.89 (s, 2H), 3.73 (s, 2H), 3.61-3.53 (m, 1H), 2.67-2.59 (m, 4H), 2.38 (s, 3H), 1.43 (s, 9H). Step 6: Synthesis of 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5-methyl-5,6-dihydro-4H-benzo[c]isoxazolo[4,5-e]azepine (Compound No. 97) To a solution of tert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[c]isoxazolo[4,5-e]azepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (0.071 g, 0.2 mmol, 1.0 eq.) in DCM (1 mL) was added TFA (0.3 mL, 3.30 mmol, 20.0 eq.), the mixture was stirred at RT for 30 minutes and concentrated in vacuo. The procedure of Example 1, step 4 was employed to afford the title compound (0.0034 g, 40% yield).1H NMR (400 MHz, CDCl3) δ 7.78-7.73 (m, 1H), 7.65-7.59 (m, 2H), 7.52-7.50 (m, 1H), 7.10-7.08 (m, 1H), 6.64-6.62 (m, 1H), 4.18 (s, 2H), 4.11 (s, 2H), 3.96 (s, 2H), 3.74 (s, 2H), 3.70-3.60 (m, 1H), 2.72 (d, J=7.8 Hz, 4H), 2.19 (s, 3H). m/z 425 (M+H)+. Compound Nos 98 to 106 Compound Nos. 98 to 106 are prepared according to the methods set forth in Table 8 below. In particular, such compound may be prepared according to the procedures of Examples 1, 11, 15, and 16 using appropriately substituted intermediates. TABLE 8Compound Nos. 98 to 106Cpd. No.Synthesis Method98Examples 15 and 199Examples 15 and 11100Examples 15 and 11101Examples 15 and 11102Examples 15 and 16103Examples 15 and 16104Examples 15 and 16105Examples 15 and 16106Examples 15 and 16 Example 16 Step 1: Synthesis of tert-butyl 6-(8-chloro-5-propionyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate To a cooled solution of tert-butyl 6-(8-chloro-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (1.0 g, 2.4 mmol, 1.0 eq.), prepared in Example 12, step 6, in DCM (20 mL) at 0° C. was added Et3N (503 μL, 3.61 mmol, 1.5 eq.), propionyl chloride (231 μL, 2.64 mmol, 1.1 eq.) and the mixture was stirred at 0° C. for 30 minutes. The mixture was diluted with DCM and washed with sat·NaHCO3, the organic layer was isolated and concentrated in vacuo. The residue was purified using flash column chromatography, eluting with 0-10 MeOH in EtOAc to give the title compound (593 mg, 53% yield).1H NMR (400 MHz, CDCl3) δ 7.60-7.45 (m, 1H), 7.21-7.14 (m, 2H), 4.62-4.40 (m, 4H), 4.00 (s, 2H), 3.94 (s, 2H), 3.50-3.39 (m, 1H), 2.78 (dd, J=8.2, 12.5 Hz, 2H), 2.62-2.60 (m, 2H), 2.49-2.35 (m, 2H), 1.43 (s, 9H), 1.25-1.16 (m, 3H). Step 2: Synthesis of 1-(8-chloro-1-(2-(pyrimidin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-5(6H)-yl)propan-1-one (Compound No. 107) A solution of tert-butyl 6-(8-chloro-5-propionyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (40 mg, 0.11 mmol, 1.0 eq.), 2-chloropyrimidine (15 mg, 0.13 mmol, 1.2 eq.), cesium carbonate (53 mg, 0.16 mmol, 1.5 eq.) in DMF (1 mL) was heated to 80° C. for 3 hours. Mixture was diluted with DCM and concentrated in vacuo. The residue was purified by preparative HPLC to yield the title compound (2.9 mg, 6% yield).1H NMR (400 MHz, DMSO) δ 8.37 (d, J=4.8 Hz, 2H), 7.77-7.68 (m, 1H), 7.63-7.57 (m, 1H), 7.51 (d, J=8.1 Hz, 1H), 6.61 (dd, J=4.7, 4.7 Hz, 1H), 4.60 (s, 2H) 4.42 (s, 2H), 4.12 (s, 2H), 4.01 (s, 2H), 3.72-3.68 (m, 1H), 2.65-2.63 (m, 4H), 2.59-2.57 (m, 2H), 1.14-1.05 (m, 3H). m/z 450 (M+H)+. Compound Nos 108 to 123, 125, and 127 to 131 Compound Nos. 108 to 116, 122, and 125 were prepared according to the methods set forth in Example 16. For example, Compound No. 108 of Table 9 lists the method of “Example 16”, indicating that this compound was prepared according to the procedure of Example 16 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 9. Compound Nos. 117 to 121, 123, and 127 to 131 are prepared according to the procedure of Example 16 using appropriately substituted intermediates. TABLE 9Compound Nos. 108 to 116, 122, and 125Compound No.Analytical DataSynthesis Method1081H NMR (400 MHz, DMSO) δ 8.51-8.50 (m,Example 161H), 7.77-7.75 (m, 1H), 7.67-7.64 (m, 1H),7.52 (d, J = 8.3 Hz, 1H), 7.30-7.27 (m, 1H), 6.70-6.67 (m, 1H), 4.60 (s, 2H), 4.30 (s, 2H), 4.14(s, 2H), 4.04 (s, 2H), 3.73-3.69 (m, 1H), 2.57-2.53 (m, 2H), 2.67 (d, J = 8.1 Hz, 4H), 1.10 (t, J =7.9 Hz, 3H); m/z 450 (M + H)+.1091H NMR (400 MHz, DMSO) δ 7.71-7.64 (m,Example 161H), 7.67-7.64 (m, 1H), 7.51 (d, J = 7.9 Hz,1H), 7.37-7.33 (m, 1H), 6.46 (d, J = 7.3 Hz,1H), 6.12 (d, J = 8.1 Hz, 1H), 4.60 (s, 2H), 4.42(s, 2H), 3.96 (s, 2H), 3.89 (s, 2H), 3.72-3.63(m, 1H), 2.62-2.58 (m, 4H), 2.56-2.53 (m,2H), 2.29 (s, 3H), 1.07-0.98 (m, 3H). m/z 463(M + H)+.1101H NMR (400 MHz, DMSO) δ 8.06 (dd, J = 1.5,Example 162.5 Hz, 1H), 7.85-7.83 (m, 2H), 7.77-7.69 (m,1H), 7.66-7.63 (m, 1H), 7.51 (d, J = 8.3 Hz,1H), 4.62 (s, 2H), 4.48 (s, 2H), 4.15 (s, 2H), 4.02(s, 2H), 3.78-3.70 (m, 1H), 2.70-2.59 (m, 4H),2.56-2.53 (m, 2H), 1.13-1.02 (m, 3H). m/z450 (M + H)+.1111H NMR (400 MHz, DMSO) δ 8.01 (d, J = 2.3Example 16Hz, 1H), 7.67-7.64 (m, 2H), 7.50 (d, J = 8.2 Hz,1H), 7.42-7.36 (m, 1H), 6.37-6.34 (m, 1H),4.93-4.83 (m, 1H), 4.50 (s, 2H), 4.34 (s, 2H),4.00 (s, 2H), 3.99 (s, 2H), 3.72-3.64 (m, 1H),2.62 (d, J = 8.1 Hz, 4H), 1.29 (d, J = 6 Hz, 6H).m/z 497 (M + H)+.1121H NMR (400 MHz, DMSO) δ 7.77-7.75 (m,Example 161H), 7.66-7.63 (m, 1H), 7.52 (d, J = 8.4 Hz,1H), 7.27-7.24 (m, 1H), 6.98-6.94 (m, 1H),4.60 (s, 2H), 4.42 (s, 2H), 4.14 (s, 2H), 4.04 (s,2H), 3.75-3.67 (m, 1H), 2.70-2.65 (m, 4H),2.56-2.53 (m, 2H), 1.10 (t, J = 7.6 Hz, 3H). m/z468 (M + H)+.1131H NMR (400 MHz, DMSO) δ 7.67-7.56 (4H,Example 16m), 6.21-6.16 (2H, m), 4.60 (2H, s), 4.42 (2H,s), 4.03 (2H, s), 3.93 (2H, s), 3.75-3.66 (1H,m), 2.64 (4H, d, J = 12.4 Hz), 1.10 (3H, dd, J = 7.3,7.3 Hz); 2H obscured by DMSO peak). m/z 467(M + H)+.1141H NMR (400 MHz, DMSO) δ 7.99 (d, J = 3.0Example 16Hz, 1H), 7.72 (s, 1H), 7.66 (dd, J = 2.4, 8.5 Hz,1H), 7.51 (d, J = 8.5 Hz, 1H), 7.42-7.37 (m, 1H),6.36 (dd, J = 3.5, 9.0 Hz, 1H), 4.52 (s, 2H), 4.34(s, 2H), 4.01 (s, 2H), 3.90 (s, 2H), 3.71-3.67(m, 1H), 3.59-3.57 (m, 1H), 2.63 (d, J = 8.2 Hz,4H), 2.30-2.23 (m, 4H), 2.05-1.93 (m, 1H),1.90-1.82 (m, 1H). m/z 493 (M + H)+.1151H NMR (400 MHz, DMSO) δ 7.66 (2H, d,Example 16J = 7.9 Hz), 7.50 (1H, d, J = 8.0 Hz), 7.35 (1H, dd,J = 7.7, 7.7 Hz), 6.46 (1H, d, J = 7.3 Hz), 6.12 (1H,d, J = 8.3 Hz), 4.53 (2H, s), 4.36 (2H, s), 3.99(2H, s), 3.89 (2H, s), 3.74 (3H, s), 3.72-3.64(1H, m), 2.62 (4H, d, J = 8.0 Hz), 2.29 (3H, s);m/z 465 (M + H)+.1161H NMR (400 MHz, DMSO) δ 8.00 (1H, d,Example 16J = 3.0 Hz), 7.76-7.76 (1H, m), 7.70 (1H, dd,J = 2.4, 8.5 Hz), 7.54 (1H, d, J = 8.5 Hz), 7.43-7.37 (1H, m), 6.37 (1H, dd, J = 3.3, 9.0 Hz), 4.50(2H, s), 4.01 (2H, s), 3.91 (2H, s), 2.95 (1H, s),2.89 (1H, s), 2.64 (4H, d, J = 8.2 Hz), 2.54 (1H,s), 2.49-2.47 (1H, m); m/z 489 (M + H)+.1221H NMR (400 MHz, DMSO) d 8.05 (dd, J = 1.5,Example 164.9 Hz, 1H), 7.86 (d, J = 2.4 Hz, 1H), 7.73 (ddd,J = 2.5, 4.2, 8.6 Hz, 1H), 7.58 (d, J = 8.7 Hz, 1H),7.52-7.47 (m, 1H), 7.10-6.81 (m, 1H), 6.62(dd, J = 5.0, 6.8 Hz, 1H), 6.35 (d, J = 8.3 Hz, 1H),4.60-4.41 (m, 4H), 4.00 (s, 2H), 3.88 (s, 2H),3.75-3.66 (m, 1H), 2.64-2.55 (m, 4H). m/z 471(M + H)+.1251H NMR (400 MHz, DMSO) δ 7.76 (s, 1H),Example 177.67-7.62 (m, 2H), 7.51 (d, J = 8.0 Hz, 1H), 7.10-7.08 (m, 1H), 6.64-6.62 (m, 1H), 4.60 (s,2H), 4.42 (s, 2H), 4.09 (s, 2H), 3.99 (s, 2H), 3.72-3.68 (m, 1H), 2.65 (d, J = 8.1 Hz, 4H), 2.57-2.54 (m, 2H), 1.10 (t, J = 7.3 Hz, 3H). m/z 475(M + H)+. Example 17 To a solution of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine bis(2,2,2-trifluoroacetate) (30 mg, 0.09 mmol, 1.0 eq.) in DMF (1.5 mL) was added 2,2-difluorocyclopropane carboxylic acid (13 mg, 0.11 mmol, 1.2 eq.), HOBt (3 mg, 0102 mmol, 0.2 eq.), EDCl·HCl (23 mg, 0.12 mmol, 1.3 eq.), DIPEA (32 μL, 0.14 mmol, 1.5 eq.) and the mixture was stirred at RT overnight. Mixture was diluted with DCM, passed through a phase separator and concentrated in vacuo. The residue was purified by preparative HPLC to give the title compound (15 mg, 37% yield).1H NMR (400 MHz, CDCl3) δ 7.51 (d, J=8.0 Hz, 2H), 7.14-7.09 (m, 1H), 4.35-4.30 (m, 2H), 4.16-4.01 (m, 2H), 3.68 (s, 2H), 3.53-3.44 (m, 1H), 3.35 (s, 2H), 2.94-2.75 (m, 2H), 2.71-2.61 (m, 2H), 2.48 (s, 3H), 2.26-2.09 (m, 2H), 1.67-1.52 (m, 1H). m/z 434 (M+H)+. Compound Nos 126, 132 to 139, 144 to 155 Compound Nos. 126, 134, 136 to 138, 147, and 153 were prepared according to the methods set forth in Example 17. For example, Compound No. 126 of Table 10 lists the method of “Example 17”, indicating that this compound was prepared according to the procedure of Example 17 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 10. Compound Nos. to 132 to 133, 135, and 139 are prepared according to the methods set forth in Table 11 below. In particular, such compound may be prepared according to the procedures of Examples 11 and 17 using appropriately substituted intermediates. TABLE 10Compound Nos. 126, 134, and 136 to 138Compound No.Analytical DataSynthesis Method1261H NMR (400 MHz, CDCl3) δ 7.53-7.49 (m,Example 172H), 7.13-7.09 (m, 1H), 4.33-4.27 (m, 2H),4.12-4.05 (m, 2H), 3.96-3.94 (m, 2H), 3.67(s, 2H), 3.52-3.43 (m, 1H), 3.39 (s, 3H), 3.36 (s,2H), 2.86-2.78 (m, 2H), 2.69-2.58 (m, 2H),2.48 (s, 3H). m/z 402 (M + H)+.1341H NMR (400 MHz, CDCl3) d 7.62-7.57 (m,Example 171H), 7.55 (d, J = 2.1 Hz, 1H), 7.21 (dd, J = 6.5, 8.6Hz, 1H), 6.20 (t, J = 53 Hz, 1H), 4.53-4.49 (m,6H), 4.15-4.13 (m, 2H), 3.53-3.43 (m, 1H),2.86 (dd, J = 8.0, 12.1 Hz, 2H), 2.68 (s, 2H), 1.38-1.16 (m, 4H). m/z 480 (M + H)+.1361H NMR (400 MHz, DMSO) d 7.79-7.79 (m,Example 171H), 7.72 (dd, J = 2.4, 8.5 Hz, 1H), 7.56 (dd,J = 2.1, 8.5 Hz, 1H), 4.81-4.57 (m, 4H), 4.31 (s,1H), 4.19 (s, 1H), 3.92 (s, 1H), 3.79 (s, 1H), 3.74-3.65 (m, 1H), 2.63-2.54 (m, 4H), 1.52-1.40(m, 3H), 1.32-1.24 (m, 2H), 0.71-0.65 (m,4H). m/z 470 (M + H)+.1371H NMR (400 MHz, DMSO) d 7.72-7.68 (m,Example 172H), 7.53 (d, J = 8.8 Hz, 1H), 4.90-4.80 (m, 1H),4.70-4.60 (m, 1H), 4.50 (s, 2H), 4.38 (s, 2H),4.16 (s, 1H), 4.04 (s, 1H), 3.66 (dd, J = 8.2, 8.2Hz, 1H), 2.64-2.54 (m, 4H), 2.31-2.10 (m,1H), 0.87-0.79 (m, 4H). m/z 480 (M + H)+.1381H NMR (400 MHz, CDCl3) d 7.63-7.56 (m,Example 171H), 7.55 (d, J = 2.1 Hz, 1H), 7.25-7.18 (m, 1H),6.2 (t, J = 52 Hz, 1H), 4.90-4.75 (m, 2H), 4.74-4.62 (m, 2H), 4.36-4.29 (m, 2H), 4.08 (s, 1H),4.01 (s, 1H), 3.53-3.44 (m, 1H), 2.89-2.82 (m,2H), 2.68 (s, 2H), 1.43-1.34 (m, 1H), 0.97-0.91 (m, 2H), 0.77-0.70 (m, 2H). m/z 462(M + H)+.1471H NMR (400 MHz, DMSO) d 7.74 (d, J = 2.3Example 17Hz, 1H), 7.68 (d, J = 8.7 Hz, 1H), 7.51 (d, J = 8.5Hz, 1H), 4.90-4.80 (m, 1H), 4.61-4.30 (m,6H), 4.02 (s, 1H), 3.90 (s, 1H), 3.69-3.60 (m,1H), 2.52-2.48 (m, 4H), 1.28-1.16 (m, 10H).m/z 488 (M + H)+.1531H NMR (400 MHz, DMSO) d 7.74 (d, J = 2.3Example 17Hz, 1H), 7.68 (d, J = 8.7 Hz, 1H), 7.51 (d, J = 8.5Hz, 1H), 4.90-4.80 (m, 1H), 4.61-4.30 (m,6H), 4.02 (s, 1H), 3.90 (s, 1H), 3.69-3.60 (m,1H), 2.52-2.48 (m, 4H), 1.28-1.16 (m, 10H).m/z 471 (M + H)+. TABLE 11Compound Nos. to 132 to 133, 135, 139,144 to 146, 148 to 152, 154, 155Cpd. No.Synthesis Method132Example 17133Example 17135Example 17139Example 17144Examples 17 and 11145Examples 17 and 11146Examples 17 and 11148Examples 17 and 11149Examples 17 and 11150Examples 17 and 11151Examples 17 and 11152Examples 17 and 11154Examples 17 and 11155Examples 17 and 11 Example 18 The title compound was prepared by using 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine from Example 10 and treating in the manner of Example 17 (19.8 mg, 52% yield).1H NMR (400 MHz, DMSO) δ 8.00 (d, J=2.9 Hz, 1H), 7.70-7.67 (m, 2H), 7.56-7.53 (m, 1H), 7.42-7.37 (m, 1H), 6.36 (dd, J=3.5, 9.2 Hz, 1H), 4.71 (s, 2H), 4.55 (s, 2H), 4.01 (s, 2H), 3.91 (s, 2H), 3.75-3.67 (m, 1H), 2.64 (d, J=8.2 Hz, 4H), 1.47-1.27 (m, 4H). m/z 497 (M+H)+. Compounds 157 to 162 Compound Nos. 157 to 162 were prepared according to the methods set forth in Example 18. For example, Compound No. 157 of Table 12 lists the method of “Example 18”, indicating that this compound was prepared according to the procedure of Example 18 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 12. TABLE 12Compound Nos. 157 to 162Compound No.Analytical DataSynthesis Method1571H NMR (400 MHz, DMSO) δ 8.35 (2H, d,Example 18J = 2.6 Hz), 7.76 (1H, s), 7.66 (1H, dd, J = 3.3, 8.5Hz), 7.51 (1H, d, J = 9.8 Hz), 4.61 (2H, s), 4.43(2H, s), 4.13 (2H, s), 4.01 (2H, s), 3.70 (1H, ddd,J = 10.1, 10.1, 10.1 Hz), 2.64 (4H, d, J = 8.5 Hz),1.11 (3H, t, J = 7.5 Hz); (2H obscured by DMSOpeak). m/z 468 (M + H)+1581H NMR (400 MHz, DMSO) δ 8.00 (d, J = 3.0Example 18Hz, 1H), 7.75 (s, 1H), 7.67 (dd, J = 2.4, 8.5 Hz,1H), 7.53 (d, J = 8.7 Hz, 1H), 7.43-7.37 (m, 1H),6.37 (dd, J = 3.4, 8.9 Hz, 1H), 5.12-4.93 (m,1H), 4.78 (s, 1H), 4.67-4.63 (m, 2H), 4.48-4.45 (m, 1H), 4.01 (s, 2H), 3.91 (s, 2H), 3.72-3.68 (m, 1H), 2.69-2.55 (m, 4H), 2.35-2.32(m, 1H), 1.70-1.59 (m, 1H), 1.20-1.09 (m,1H). m/z 497 (M + H)+.1591H NMR (400 MHz, DMSO) δ 8.00 (d, J = 3.0Example 18Hz, 1H), 7.76 (s, 1H), 7.65 (dd, J = 2.2, 8.3 Hz,1H), 7.52 (d, J = 8.5 Hz, 1H), 7.40-7.37 (m, 1H),6.36 (dd, J = 3.2, 8.7 Hz, 1H), 4.94-4.79 (m,1H), 4.78-4.51 (m, 4H), 4.01 (s, 2H), 3.90 (s,2H), 3.74-3.66 (m, 1H), 2.80-2.69 (m, 1H),2.65-2.62 (m, 4H), 1.56-1.46 (m, 1H), 1.27-1.20 (m, 1H). m/z 497 (M + H)+.1601H NMR (400 MHz, DMSO) δ 7.99 (d, J = 3.0Example 18Hz, 1H), 7.74-7.74 (m, 1H), 7.66 (dd, J = 2.4, 8.5Hz, 1H), 7.51 (d, J = 8.7 Hz, 1H), 7.42-7.37 (m,1H), 6.36 (dd, J = 3.5, 9.0 Hz, 1H), 4.84-4.71 (m,4H), 4.52-4.49 (m, 2H), 4.36-4.25 (m, 3H),4.01 (s, 2H), 3.90 (s, 2H), 3.71-3.67 (m, 1H),2.63 (d, J = 8.0 Hz, 4H). m/z 495 (M + H)+.1611H NMR (400 MHz, DMSO) δ 8.35 (d, J = 2.6Example 18Hz, 2H), 7.76 (s, 1H), 7.66 (dd, J = 3.3, 8.5 Hz,1H), 7.51 (d, J = 9.8 Hz, 1H), 4.61 (s, 2H), 4.43(s, 2H), 4.13 (s, 2H), 4.01 (s, 2H), 3.71-3.67(m, 1H), 2.64 (d, J = 8.5 Hz, 4H), 2.57-2.53 (m,2H), 1.11 (t, J = 7.5 Hz, 3H). m/z 468 (M + H)+1621H NMR (400 MHz, DMSO) δ 7.68-7.64 (m,Example 182H), 7.49 (d, J = 8.3 Hz, 1H), 4.52 (s, 2H), 4.35(s, 2H), 4.09-4.01 (m, 4H), 3.73 (s, 3H), 3.68-3.63 (m, 1H), 2.60 (d, J = 8.0 Hz, 4H), 1.52-1.45(m, 1H), 0.71-0.68 (m, 4H); m/z 442 (M + H)+. Example 19 Step 1: Synthesis of methyl (5-chloro-2-nitrobenzyl)alaninate To a solution of 5-chloro-2-nitrobenzaldehyde (2 g, 10.78 mmol, 1.0 eq.) in DCM (30 mL) was added DL-Alanine methyl ester hydrochloride (1.5 g, 10.78 mmol, 1.0 eq.), trimethylamine (1.1 g, 10.78 mmol, 1.0 eq.) and sodium triacetoxyborohydride (4.6 g, 21.56 mmol, 2.0 eq.). The resulting solution was stirred at RT for 1 hour, diluted with DCM, washed with an aq. sol. of sat. NaHCO3, dried, concentrated in vacuo. The residue was purified by flash column chromatography eluting with 50-100% EtOAc in isohexane to give the title compound as a light yellow oil (1.23 g, 42% yield).1H NMR (400 MHz, CDCl3) δ 7.94-7.91 (m, 1H), 7.72 (d, J=2.3 Hz, 1H), 7.40-7.36 (m, 1H), 4.15-4.09 (m, 1H), 3.99-3.94 (m, 1H), 3.73-3.72 (m, 3H), 3.40-3.37 (m, 1H), 1.36-1.33 (m, 3H). Step 2: Synthesis of 7-chloro-3-methyl-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepin-2-one To a solution of methyl (5-chloro-2-nitrobenzyl)alaninate (1.23 g, 4.51 mmol, 1.0 eq.) in acetic acid (30 mL) was added iron (0.63 g, 11.28 mmol, 2.5 eq.). The resulting suspension was heated at 110° C. for 30 minutes, cooled to RT and filtered through celite rinsing with acetic acid. The filtrate was partitioned between aq. sol. of sat. NaHCO3and EtOAc and the organic phase was dried and concentrated in vacuo. The residue was triturated with diethyl ether to give the title compound as a light brown solid (0.70 g, 57% yield).1H NMR (400 MHz, CDCl3) δ 7.85-7.79 (m, 1H), 7.25-7.23 (m, 1H), 6.93-6.88 (m, 1H), 4.09 (d, J=13.4 Hz, 1H), 3.87 (d, J=13.1 Hz, 1H), 3.57 (q, J=6.6 Hz, 1H), 1.31 (d, J=6.6 Hz, 3H). Step 3: Synthesis of tert-butyl 7-chloro-3-methyl-2-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate To a cooled suspension of 7-chloro-3-methyl-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepin-2-one (2.07 g, 9.82 mmol, 1.0 eq.) in THE (20 mL) at 0° C. was added a solution of di-tert-butyl dicarbonate (3.21 g, 14.73 mmol, 1.5 eq.) in THE (10 mL) dropwise over 10 minutes. Mixture was allowed to warm to RT and stirred overnight. The mixture was concentrated in vacuo and triturated with diisopropyl ether to afford the title compound as a light brown solid (2.93 g, 96% yield).1H NMR (400 MHz, CDCl3) δ 8.02 (s, 1H), 7.23-7.17 (m, 1H), 6.84-6.80 (m, 1H), 5.21-4.19 (m, 3H), 1.54-1.50 (m, 3H), 1.41 (s, 9H). Step 4: Synthesis of tert-butyl 7-chloro-3-methyl-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate To a suspension of tert-butyl 7-chloro-3-methyl-2-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (0.95 g, 3.06 mmol, 1.0 eq.) in THF (10 mL) was added 2,4-bis(4-methoxyphenyl)-2,4-dithioxo-1,3,2,4-dithiadiphosphetane (0.74 g, 1.83 mmol, 0.60 eq.) and the mixture was heated to reflux for 90 minutes. The mixture was allowed to cool, concentrated in vacuo. The residue was purified by flash column chromatography eluting with 90-100% EtOAc in isohexane to give the title compound as a light yellow solid (1.02 g, 100% yield).1H NMR (400 MHz, CDCl3) δ 9.34-9.34 (m, 1H), 7.25-7.23 (m, 1H), 6.90-6.87 (m, 1H), 5.52-5.43 (m, 1H), 4.67-4.51 (m, 2H), 1.48-1.41 (m, 12H). Step 5: Synthesis of tert-butyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate To a solution of tert-butyl 7-chloro-3-methyl-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (0.3 g, 0.92 mmol, 1.0 eq.), was added 2-(4-fluoropyridin-2-yl)-2-azaspiro[3.3]heptane-6-carbohydrazide (0.25 g, 1.01 mmol, 1.1 eq.) in dioxane (5 mL) and the mixture was heated to 90° C. for 36 hours. The mixture was allowed to cool and was concentrated in vacuo. The residue was purified by flash column chromatography eluting with 90-100% EtOAc in isohexane then 0-10% MeOH in DCM to give the title product as a brown foam (0.31 g, 64% yield).1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=2.5 Hz, 1H), 7.56-7.48 (m, 2H), 7.26-7.16 (m, 2H), 6.23 (dd, J=3.4, 9.0 Hz, 1H), 5.57-5.51 (m, 1H), 5.06-4.75 (m, 1H), 4.14-3.97 (m, 4H), 3.81-3.61 (m, 1H), 3.56-3.45 (m, 1H), 2.93-2.47 (m, 4H), 1.49 (s, 9H), 1.16-1.03 (m, 3H). Step 6: Synthesis of methyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (Compound No. 142) To a solution of tert-butyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (1.10 g, 4.31 mmol, 1.0 eq.) in MeOH (3 mL) was added 4 M HCl in dioxane (1.5 mL, 5.90 mmol, 10.0 eq.) and the mixture was stirred at RT for 1 hour. The mixture was concentrated in vacuo to give a light brown solid. To a solution of the light brown solid (137 mg, 0.29 mmol, 1.0 eq.) and Et3N (0.15 mL, 1.04 mmol, 3.5 eq.) at 0° C. in THE (3 mL) was added methyl chloroformate (0.034 mL, 0.45 mmol, 1.5 eq.) dropwise. The resulting solution was stirred at RT for 1 hour, diluted with EtOAc, washed with an aq. sol. of sat. NaHCO3, and dried. The mixture was concentrated in vacuo and the residue was purified by preparative HPLC to give the title compound as an off-white solid (49 mg, 34% yield).1H NMR (400 MHz, DMSO-d6) δ 8.03 (d, J=3.0 Hz, 1H), 7.76-7.72 (m, 1H), 7.70 (dd, J=2.4, 8.5 Hz, 1H), 7.59 (d, J=8.5 Hz, 1H), 7.51-7.45 (m, 1H), 6.39 (dd, J=3.6, 9.1 Hz, 1H), 5.39-5.31 (m, 1H), 4.85-4.72 (m, 1H), 3.98 (m, 3H), 3.88-3.81 (m, 2H), 3.71 (m, 4H), 2.69-2.67 (m, 2H), 2.49-2.41 (m, 2H), 1.10-1.06 (m, 3H); m/z 483 (M+H)+. Compounds 140, 141, 143, and 163 to 173 Compound Nos. 163 to 173 were prepared according to the methods set forth in Example 19. For example, Compound No. 163 of Table 13 lists the method of “Example 19”, indicating that this compound was prepared according to the procedure of Example 19 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 13. Compound Nos. to 140, 141, and 143 are prepared according to the procedures of Examples 19 using appropriately substituted intermediates. TABLE 13Compound Nos. 163 to 173Compound No.Analytical DataSynthesis Method1631H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.8Example 19Hz, 1H), 7.57-7.54 (m, 1H), 7.52 (dd, J = 2.0,8.3 Hz, 1H), 7.25-7.17 (m, 2H), 6.24 (dd,J = 3.4, 9.0 Hz, 1H), 5.65-5.58 (m, 1H), 5.10-5.05 (m, 1H), 5.04-4.94 (m, 1H), 4.10-3.98 (m, 4H), 3.90-3.68 (m, 1H), 3.53-3.45(m, 1H), 2.95-2.77 (m, 3H), 2.55-2.52 (m,1H), 1.33-1.27 (m, 6H), 1.13-1.10 (m, 3H);m/z 511 (M + H)+.1641H NMR (400 MHz, CDCl3) δ 8.01 (dd,Example 19J = 1.5, 2.6 Hz, 1H), 7.85 (d, J = 2.8 Hz, 1H),7.77 (d, J = 1.5 Hz, 1H), 7.61-7.52 (m, 2H),7.18 (d, J = 8.5 Hz, 1H), 5.65-5.55 (m, 1H),5.11-4.94 (m, 2H), 4.20-4.12 (m, 4H), 3.77-3.72 (m, 1H), 3.55-3.46 (m, 1H), 2.98-2.81 (m, 3H), 2.60-2.51 (m, 1H), 1.34-1.27(m, 6H), 1.13-1.09 (m, 3H); m/z 494(M + H)+.1651H NMR (400 MHz, CDCl3) δ 8.01 (dd,Example 19J = 1.5, 2.8 Hz, 1H), 7.85 (d, J = 2.8 Hz, 1H),7.77 (d, J = 1.4 Hz, 1H), 7.53 (dd, J = 1.9, 8.3Hz, 2H), 7.18 (d, J = 8.5 Hz, 1H), 5.71-5.49(m, 1H), 5.17-4.91 (m, 1H), 4.20-4.12 (m,4H), 3.91-3.82 (m, 1H), 3.79 (s, 3H), 3.54-3.46 (m, 1H), 2.97-2.83 (m, 3H), 2.62 (s,1H), 1.15-1.13 (m, 3H); m/z 466 (M + H)+.1661H NMR (400 MHz, CDCl3) δ 8.30 (d, J = 4.8Example 19Hz, 2H), 7.57-7.51 (m, 2H), 7.20 (d, J = 8.5Hz, 1H), 6.53 (t, J = 4.8 Hz, 1H), 5.67-5.53(m, 1H), 5.11-4.93 (m, 1H), 4.24-4.13 (m,4H), 3.91-3.81 (m, 1H), 3.78 (s, 3H), 3.56-3.46 (m, 1H), 2.97-2.82 (m, 3H), 2.54-2.53(m, 1H), 1.15-1.12 (m, 3H); m/z 466(M + H)+.1671H NMR (400 MHz, CDCl3) δ 8.30 (d, J = 4.5Example 19Hz, 2H), 7.56-7.50 (m, 2H), 7.20 (d, J = 8.6Hz, 1H), 6.53 (dd, J = 4.7, 4.7 Hz, 1H), 5.67-5.61 (m, 1H), 5.19-4.91 (m, 2H), 4.27-4.17(m, 2H), 4.13 (s, 2H), 3.82-3.75 (m, 1H),3.56-3.46 (m, 1H), 2.97-2.79 (m, 3H), 2.57-2.49 (m, 1H), 1.32-1.29 (m, 6H), 1.19-1.05 (m, 3H); m/z 494 (M + H)+.1681H NMR (400 MHz, CDCl3) δ 8.32-8.28Example 19(m, 2H), 7.60-7.51 (m, 2H), 7.20 (d, J = 8.1Hz, 1H), 6.55-6.51 (m, 1H), 5.61-5.55 (m,1H), 5.10-5.01 (m, 1H), 4.27-4.11 (m, 4H),4.12 (s, 2H), 3.84-3.80 (m, 1H), 3.51-3.49(m, 1H), 2.97-2.80 (m, 3H), 2.56-2.53 (m,1H), 1.32-1.30 (m, 3H), 1.20-1.05 (m, 3H);m/z 480 (M + H)+.1691H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Example 19Hz, 1H), 7.55-7.50 (m, 2H), 7.25-7.17 (m,2H), 6.24 (dd, J = 3.3, 9.0 Hz, 1H), 5.61-5.56(m, 1H), 5.10-5.01 (m, 1H), 4.26-4.18 (m,2H), 4.10-3.98 (m, 4H), 3.91-3.68 (m, 1H),3.55-3.45 (m, 1H), 2.94-2.78 (m, 3H), 2.58-2.49 (m, 1H), 1.31 (t, J = 7.1 Hz, 3H), 1.18-1.08 (m, 3H); m/z 497 (M + H)+.1701H NMR (400 MHz, DMSO-d6) δ 8.43 (s,Example 192H), 7.76-7.72 (m, 1H), 7.69 (dd, J = 2.4, 8.5Hz, 1H), 7.59 (d, J = 8.5 Hz, 1H), 5.41-5.33(m, 1H), 4.89-4.68 (m, 1H), 4.14-4.06 (m,2H), 4.00-3.95 (m, 3H), 3.72-3.69 (m, 4H),2.73-2.68 (m, 2H), 2.51-2.39 (m, 2H), 1.15-1.05 (m, 3H); m/z 484 (M + H)+.1711H NMR (400 MHz, CDCl3) δ 8.31-8.30Example 19(m, 2H), 7.58-7.51 (m, 2H), 7.21-7.18 (m,1H), 6.53 (t, J = 4.8 Hz, 1H), 5.56-5.51 (m,1H), 5.06-4.66 (m, 1H), 4.24-4.12 (m, 5H),3.80-3.77 (m, 1H), 3.56-3.46 (m, 1H), 2.97-2.53 (m, 4H), 1.31-1.06 (m, 3H), 0.74 (d,J = 5.9 Hz, 4H); m/z 492 (M + H)+.1721H NMR (400 MHz, CDCl3) δ 8.20-8.19Example 19(m, 2H), 7.55-7.50 (m, 2H), 7.21-7.18 (m,1H), 5.66-5.60 (m, 1H), 5.02-4.77 (m, 1H),4.24-4.10 (m, 6H), 3.84-3.74 (m, 1H), 3.56-3.45 (m, 1H), 2.96-2.78 (m, 3H), 2.54-2.51 (m, 1H), 1.35-1.11 (m, 6H); m/z 498(M + H)+.1731H NMR (400 MHz, CDCl3) δ 8.19-8.19Example 19(m, 2H), 7.58-7.50 (m, 2H), 7.21-7.16 (m,1H), 5.58-5.52 (m, 1H), 5.07-4.72 (m, 1H),4.21-4.09 (m, 4H), 3.78-3.68 (m, 1H), 3.54-3.46 (m, 1H), 2.96-2.77 (m, 3H), 2.52-2.47 (m, 1H), 1.58 (s, 3H), 1.05-0.64 (m,7H); m/z 524 (M + H)+. Example 20 To a solution of tert-butyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (1.10 g, 4.31 mmol, 1.0 eq.) in MeOH (3 mL) was added 4 M HCl in dioxane (1.5 mL, 5.90 mmol, 10.0 eq.) and the mixture was stirred at RT for 1 hour. The mixture was concentrated in vacuo to give a light brown solid. To a solution of the light brown solid (137 mg, 0.29 mmol, 1.0 eq.) in DMF (3 mL) was added 1-fluorocyclopropane-1-carboxylic acid (37 mg, 0.36 mmol, 1.2 eq.), HOBt (8 mg, 0.059 mmol, 0.2 eq.), EDCl·HCl (74 mg, 0.39 mmol, 1.3 eq.), DIPEA (181 μL, 1.04 mmol, 3.5 eq.) and the mixture was stirred at RT overnight. The mixture was partitioned between aq. sol. of sat. NaHCO3and DCM and the organic phase was dried and concentrated in vacuo. The residue was purified by preparative HPLC to give the title compound as an off-white solid (61 mg, 40% yield).1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J=3.0 Hz, 1H), 7.79 (dd, J=2.3, 8.5 Hz, 2H), 7.69 (d, J=8.5 Hz, 1H), 7.56-7.50 (m, 1H), 6.45 (dd, J=3.6, 9.1 Hz, 1H), 5.66-5.61 (m, 1H), 5.11-5.07 (m, 1H), 4.28-4.24 (m, 1H), 4.08-3.99 (m, 2H), 3.91 (q, J=8.4 Hz, 2H), 3.82-3.74 (m, 1H), 2.85-2.80 (m, 2H), 2.53-2.43 (m, 2H), 1.56-1.45 (m, 2H), 1.41-1.32 (m, 2H), 1.07-1.03 (m, 3H); m/z 511 (M+H)+. Compound 175 Compound No. 18-2 was prepared according to the methods set forth in Example 20. For example, Compound No. 20-2 of Table 14 lists the method of “Example 20”, indicating that this compound was prepared according to the procedure of Example 20 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum):1H NMR (400 MHz, CDCl3) δ 8.30 (d, J=4.9 Hz, 2H), 7.60-7.55 (m, 2H), 7.26-7.23 (m, 1H), 6.54 (t, J=4.8 Hz, 1H), 6.01-5.88 (m, 1H), 5.16-5.13 (m, 1H), 4.27-4.13 (m, 4H), 4.08-3.92 (m, 1H), 3.58-3.50 (m, 1H), 2.97 (dd, J=8.3, 11.7 Hz, 1H), 2.83 (dd, J=8.7, 11.7 Hz, 2H), 2.62-2.49 (m, 1H), 1.53-1.24 (m, 4H), 1.20-1.06 (m, 3H); m/z 494 (M+H)+. Example 21 To a solution of tert-butyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (1.10 g, 4.31 mmol, 1.0 eq.) in MeOH (3 mL) was added 4 M HCl in dioxane (1.5 mL, 5.90 mmol, 10.0 eq.) and the mixture was stirred at RT for 1 hour. The mixture was concentrated in vacuo to give a light brown solid. To a solution of the light brown solid (60 mg, 0.14 mmol, 1.0 eq.) in DMF (1 mL) was added difluoroacetic acid (13 μL, 0.21 mmol, 1.5 eq.), HATU (80 mg, 0.21 mmol, 1.5 eq.), DIPEA (37 μL, 0.21 mmol, 1.5 eq.) and the mixture was stirred at RT overnight. The mixture was partitioned between aq. sol. of sat. NaHCO3and DCM and the organic phase was dried and concentrated in vacuo. The residue was purified by preparative HPLC to give the title compound as an off-white solid (11 mg, 15% yield).1H NMR (400 MHz, DMSO-d6) δ 8.04 (d, J=3.0 Hz, 1H), 7.88 (d, J=2.4 Hz, 1H), 7.75 (dd, J=2.4, 8.5 Hz, 1H), 7.63 (d, J=8.5 Hz, 1H), 7.51-7.45 (m, 1H), 7.04 (t, J=52.2 Hz, 1H), 6.40 (dd, J=3.5, 9.0 Hz, 1H), 5.63-5.57 (m, 1H), 4.84 (d, J=14.9 Hz, 1H), 4.15 (d, J=14.2 Hz, 1H), 4.04-3.94 (m, 2H), 3.90-3.82 (m, 2H), 3.78-3.68 (m, 1H), 2.80-2.68 (m, 2H), 2.53-2.48 (m, 1H), 2.39-2.34 (m, 1H), 0.96-0.95 (m, 3H); m/z 503 (M+H)+. Compound Nos. 177 to 178 Compound Nos. 177 to 178 were prepared according to the methods set forth in Example 21. For example, Compound No. 177 of Table 15 lists the method of “Example 21”, indicating that this compound was prepared according to the procedure of Example 21 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 15. TABLE 15Compound Nos. 177 to 178Compound No.Analytical DataSynthesis Method1771H NMR (400 MHz, CDCl3) δ 8.01 (dd,Example 21J = 1.5, 2.8 Hz, 1H), 7.85 (d, J = 2.8 Hz, 1H),7.78 (d, J = 1.5 Hz, 1H), 7.57 (d, J = 7.0 Hz,2H), 7.26-7.23 (m, 1H), 6.01-5.97 (m, 1H),5.19-5.07 (m, 1H), 4.22-4.13 (m, 4H), 4.10-4.01 (m, 1H), 3.57-3.50 (m, 1H), 3.00-2.81 (m, 3H), 2.60-2.52 (m, 1H), 1.60-1.26(m, 4H), 1.19-1.10 (m, 3H); m/z 494(M + H)+.1781H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 3.0Example 21Hz, 1H), 7.56 (d, J = 6.9 Hz, 2H), 7.25-7.19(m, 2H), 6.24 (dd, J = 3.5, 9.0 Hz, 1H), 5.99-5.94 (m, 1H), 5.35-5.29 (m, 1H), 4.12-3.99(m, 4H), 3.91-3.80 (m, 1H), 3.57-3.49 (m,1H), 2.97-2.76 (m, 3H), 2.51-2.49 (m, 1H),1.76-1.65 (m, 6H), 1.13-1.01 (m, 3H); m/z513 (M + H)+. Example 22 To a solution of 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (88 mg, 0.19 mmol, 1.0 eq.) in DCM (2 mL) was added a 37% formaldehyde aq. (0.043 mL, 0.57 mmol, 3.0 eq.), acetic acid (0.1 mL) and sodium triacetoxyborohydride (81 mg, 0.38 mmol, 2.0 eq.). The resulting solution was stirred at RT for 16 hours, diluted with DCM, washed with an aq. sol. of sat. NaHCO3, dried, concentrated in vacuo to give the title compound as an off-white solid (43 mg, 50% yield).1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=2.9 Hz, 1H), 7.49 (dd, J=2.3, 8.4 Hz, 1H), 7.43 (d, J=2.3 Hz, 1H), 7.25-7.19 (m, 1H), 7.15 (d, J=8.4 Hz, 1H), 6.23 (dd, J=3.5, 9.0 Hz, 1H), 4.10-4.03 (m, 2H), 4.00 (s, 2H), 3.51-3.44 (m, 3H), 3.31-3.27 (m, 1H), 2.90-2.76 (m, 3H), 2.55-2.48 (m, 1H), 2.41 (s, 3H); m/z 439 (M+H)+. Compound Nos. 22 and 23 Compound Nos. 22 and 23 are prepared according to the procedure of Example 22 using appropriately substituted intermediates. Example 23 To a solution of methyl 1-(2-(tert-butoxycarbonyl)-2-azaspiro[3.3]heptan-6-yl)-8-chloro-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (0.25 g, 0.51 mmol, 1.0 eq.) in DCM (15 mL) was added TFA (1.1 mL, 14.4 mmol, 28.1 eq.) and the mixture was stirred at RT for 1 hour. Toluene was added and the mixture was concentrated in vacuo to give a light yellow oil. A mixture of the light yellow oil (50 mg, 0.13 mmol, 1.0 eq.), 2-bromo-6-methylpyridine (0.015 mL, 0.13 mmol, 1.0 eq.), RuPhos (12 mg, 0.03 mmol, 0.20 eq.), palladium acetate (3 mg, 0.01 mmol, 0.10 eq.), cesium carbonate (0.126 g, 0.39 mmol, 3.0 eq.) in dioxane (3 mL) was degassed using N2, for 10 minutes and then heated to 80° C. for 2 hours. The mixture was allowed to cool to RT and partitioned between water and EtOAc. The organic phase was dried and concentrated in vacuo and the residue was purified by preparative HPLC to give the title compound as an off-white solid (8 mg, 13% yield).1H NMR (400 MHz, CDCl3) δ 7.55-7.50 (m, 2H), 7.37-7.31 (m, 1H), 7.21-7.17 (m, 1H), 6.46 (d, J=7.4 Hz, 1H), 6.11-6.07 (m, 1H), 5.62-5.54 (m, 1H), 5.01-4.78 (m, 1H), 4.16-4.00 (m, 4H), 3.88-3.79 (m, 4H), 3.53-3.44 (m, 1H), 2.92-2.77 (m, 3H), 2.55-2.49 (m, 1H), 2.39 (s, 3H), 1.13-1.07 (m, 3H); m/z 479 (M+H)+. Compound Nos 181 to 183 Compound Nos. 181 to 183 were prepared according to the methods set forth in Example 23. For example, Compound No. 181 of Table 16 lists the method of “Example 23”, indicating that this compound was prepared according to the procedure of Example 23 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 16. TABLE 16Compound Nos. 181 to 183Compound No.Analytical DataSynthesis Method1811H NMR (400 MHz, CDCl3) δ 8.15-8.13Example 23(m, 1H), 7.60-7.55 (m, 2H), 7.48-7.42 (m,1H), 7.26-7.20 (m, 1H), 6.61 (dd, J = 5.2, 6.7Hz, 1H), 6.28 (d, J = 8.4 Hz, 1H), 6.05-5.89(m, 1H), 5.17-5.11 (m, 1H), 4.13 (dd, J = 8.2,22.2 Hz, 2H), 4.06-4.01 (m, 2H), 3.85-3.79(m, 1H), 3.56-3.49 (m, 1H), 2.97-2.78 (m,3H), 2.57-2.48 (m, 1H), 1.61-1.27 (m, 4H),1.21-1.05 (m, 3H); m/z 493 (M + H)+.1821H NMR (400 MHz, CDCl3) δ 7.91 (d, J = 4.9Example 23Hz, 1H), 7.57-7.51 (m, 2H), 7.21-7.11 (m,2H), 6.61-6.55 (m, 1H), 5.61-5.59 (m, 1H),4.98-4.74 (m, 1H), 4.23-4.14 (m, 4H), 3.89-3.77 (m, 4H), 3.53-3.44 (m, 1H), 2.94-2.78 (m, 3H), 2.57-2.50 (m, 1H), 1.20-1.03(m, 3H); m/z 483 (M + H)+.1831H NMR (400 MHz, DMSO-d6) δ 8.30-8.28Example 23(m, 1H), 8.17 (d, J = 4.8 Hz, 1H), 7.76-7.68(m, 2H), 7.55-7.52 (m, 1H), 4.66-4.08 (m,8H), 3.72-3.68 (m, 4H), 2.65-2.57 (m, 4H);m/z 470 (M + H)+. Example 24 Step 1: Synthesis of methyl 7-chloro-3-methyl-2-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate To a solution 7-chloro-3-methyl-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepin-2-one, prepared in example 1 step 2, (1.0 g, 4.75 mmol, 1.0 eq.) and Et3N (1.3 mL, 9.49 mmol, 2.0 eq.) at 0° C. in THF (20 mL) was added methyl chloroformate (0.55 mL, 7.12 mmol, 1.5 eq.) dropwise. The resulting solution was stirred at RT for 2 hours, diluted with EtOAc, washed with an aq. sol. of sat. NaHCO3, and dried. The residue was purified by flash column chromatography eluting with 70-100% EtOAc in isohexane to give the title compound as a yellow solid (0.82 g, 64% yield).1H NMR (400 MHz, DMSO-d6) δ 8.57 (s, 1H), 7.26-7.19 (m, 2H), 6.90-6.87 (m, 1H), 5.22-4.95 (m, 1H), 4.60-4.53 (m, 1H), 4.32-4.21 (m, 1H), 3.70 (s, 3H), 1.55 (d, J=6.7 Hz, 3H). Step 2: Synthesis of methyl 7-chloro-3-methyl-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate To a suspension of methyl 7-chloro-3-methyl-2-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (0.82 g, 3.05 mmol, 1.0 eq.) in THE (10 mL) was added 2,4-bis(4-methoxyphenyl)-2,4-dithioxo-1,3,2,4-dithiadiphosphetane (0.74 g, 1.83 mmol, 0.60 eq.) and the mixture was heated to reflux for 1 hour. The mixture was allowed to cool and was concentrated in vacuo. The residue was purified by flash column chromatography eluting with 90-100% EtOAc in isohexane to give the title compound as a yellow solid (0.67 g, 77% yield).1H NMR (400 MHz, CDCl3) δ 9.46-9.43 (m, 1H), 7.31-7.28 (m, 2H), 6.93-6.90 (m, 1H), 5.60-5.52 (m, 1H), 4.71-4.55 (m, 2H), 3.73 (s, 3H), 1.42 (d, J=6.5 Hz, 3H). Step 3: Synthesis of methyl 1-(2-(tert-butoxycarbonyl)-2-azaspiro[3.3]heptan-6-yl)-8-chloro-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate To a solution of methyl 7-chloro-3-methyl-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (0.3 g, 1.05 mmol, 1.0 eq.), was added tert-butyl 6-(hydrazinecarbonyl)-2-azaspiro[3.3]heptane-2-carboxylate (0.320 g, 1.26 mmol, 1.2 eq.) in dioxane (5 mL) and the mixture was heated to 90° C. for 36 hours. The mixture was allowed to cool and was concentrated in vacuo. The residue was purified by flash column chromatography eluting with 50-100% EtOAc in isohexane then 0-10% MeOH in DCM to give the title product as an off-white solid (0.43 g, 83% yield).1H NMR (400 MHz, CDCl3) δ 7.54-7.49 (m, 2H), 7.16 (dd, J=2.6, 8.5 Hz, 1H), 5.59-5.52 (m, 1H), 5.01-4.58 (m, 1H), 4.04-3.88 (m, 5H), 3.77 (s, 3H), 3.46-3.38 (m, 1H), 2.85-2.70 (m, 3H), 2.51-2.42 (m, 1H), 1.61-1.58 (m, 9H), 1.19-1.05 (m, 3H). Step 4: Synthesis of methyl 8-chloro-4-methyl-1-(2-(pyrimidin-4-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (Compound No. 184) To a solution of methyl 1-(2-(tert-butoxycarbonyl)-2-azaspiro[3.3]heptan-6-yl)-8-chloro-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (0.30 g, 0.62 mmol, 1.0 eq.) in DCM (4 mL) was added TFA (1.3 mL, 17.3 mmol, 28.1 eq.) and the mixture was stirred at RT for 1 hour. The mixture was concentrated in vacuo and was purified using a 5 g SCX-2 cartridge eluting with MeOH then 2.3 M ammonia in MeOH to give a colourless oil. To a solution of the colourless oil (20 mg, 0.052 mmol, 1.0 eq.) in N,N-dimethylformamide (1 mL) was added cesium carbonate (50 mg, 0.155 mmol, 3.0 eq.) and 4-chloropyrimidine hydrochloride (12 mg, 0.077 mmol, 1.50 eq.). The mixture was heated to 80° C. for 16 hours, allowed to cool to RT and partitioned between water and EtOAc. The organic phase was dried and concentrated in vacuo and the residue was purified by preparative HPLC to give the title compound as a white solid (12 mg, 50% yield).1H NMR (400 MHz, CDCl3) δ 8.57 (s, 1H), 8.16 (d, J=6.1 Hz, 1H), 7.56-7.51 (m, 2H), 7.19-7.15 (m, 1H), 6.17-6.15 (m, 1H), 5.64-5.54 (m, 1H), 5.20-4.46 (m, 1H), 4.20-4.10 (m, 4H), 3.90-3.79 (m, 4H), 3.55-3.46 (m, 1H), 2.96-2.56 (m, 4H), 1.20-1.08 (m, 3H); m/z 466 (M+H)+. Compound Nos. 185 to 190 Compound Nos. 185 to 190 were prepared according to the methods set forth in Example 24. For example, Compound No. 185 of Table 17 lists the method of “Example 24”, indicating that this compound was prepared according to the procedure of Example 24 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 17. TABLE 17Compound Nos. 185 to 190Compound No.Analytical DataSynthesis Method1851H NMR (400 MHz, CDCl3) δ 8.34 (d, J = 3.1Example 24Hz, 1H), 7.99 (d, J = 4.4 Hz, 1H), 7.59-7.52(m, 2H), 7.19-7.16 (m, 1H), 5.24-3.60 (m,9H), 3.55-3.41 (m, 1H), 2.92-2.70 (m, 4H),1.33-1.26 (m, 6H); m/z 498 (M + H)+.1861H NMR (400 MHz, CDCl3) δ 8.58 (d, J = 1.0Example 24Hz, 1H), 8.18-8.13 (m, 1H), 7.59-7.52 (m,2H), 7.19-7.15 (m, 1H), 6.17 (d, J = 5.5 Hz,1H), 4.64-4.13 (m, 8H), 3.78 (s, 3H), 3.55-3.46 (m, 1H), 2.92-2.68 (m, 4H); m/z 452(M + H)+.1871H NMR (400 MHz, CDCl3) δ 8.58-8.56Example 24(m, 1H), 8.18-8.15 (m, 1H), 7.57-7.49 (m,2H), 7.18-7.15 (m, 1H), 6.17 (dd, J = 1.2, 6.0Hz, 1H), 5.66-5.55 (m, 1H), 5.09-4.94 (m,2H), 4.21-4.11 (m, 4H), 3.85-3.67 (m, 1H),3.55-3.46 (m, 1H), 2.96-2.57 (m, 4H), 1.33-1.09 (m, 9H); m/z 494 (M + H)+.1881H NMR (400 MHz, CDCl3) δ 8.34 (d, J = 3.1Example 24Hz, 1H), 7.99 (d, J = 4.4 Hz, 1H), 7.58-7.51(m, 2H), 7.19-7.15 (m, 1H), 5.59-5.54 (m,1H), 5.04-4.77 (m, 1H), 4.37-4.30 (m, 4H),3.88-3.79 (m, 4H), 3.52-3.44 (m, 1H), 2.97-2.82 (m, 3H), 2.63-2.54 (m, 1H), 1.15-1.10 (m, 3H); m/z 484 (M + H)+.1891H NMR (400 MHz, CDCl3) δ 8.45-8.43Example 24(m, 1H), 7.55-7.50 (m, 2H), 7.21-7.17 (m,1H), 6.79 (d, J = 4.9 Hz, 1H), 6.46-6.17 (m,1H), 5.66-5.57 (m, 1H), 5.07-4.67 (m, 1H),4.26-4.16 (m, 6H), 3.88-3.70 (m, 1H), 3.55-3.45 (m, 1H), 2.95-2.53 (m, 4H), 1.33-1.11 (m, 6H); m/z 530 (M + H)+.1901H NMR (400 MHz, CDCl3) δ 8.48 (d, J = 4.6Example 24Hz, 1H), 7.53 (dd, J = 2.2, 8.5 Hz, 2H), 7.20-7.17 (m, 1H), 6.80 (d, J = 4.9 Hz, 1H), 5.66-5.61 (m, 1H), 5.07-4.75 (m, 1H), 4.29-4.18(m, 6H), 3.86-3.75 (m, 1H), 3.55-3.45 (m,1H), 2.97-2.53 (m, 4H), 1.33-1.12 (m, 6H);m/z 548 (M + H)+. Example 25 Step 1: Synthesis of 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine To a solution of tert-butyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (0.28 g, 0.57 mmol, 1.0 eq.) in DCM (4 mL) was added TFA (1.2 mL, 15.4 mmol, 28.1 eq.) and the mixture was stirred at RT for 1 hour. The mixture was concentrated in vacuo and was purified using a 5 g SCX-2 cartridge eluting with MeOH then 2.3 M ammonia in MeOH to give the title compound as a pale yellow oil (0.28 g, 0.55 mmol, 1.0 eq.).1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=2.8 Hz, 1H), 7.50-7.46 (m, 2H), 7.28-7.19 (m, 1H), 7.12 (d, J=9.1 Hz, 1H), 6.23 (dd, J=3.2, 9.0 Hz, 1H), 3.98 (s, 2H), 3.79-3.70 (m, 2H), 3.69 (s, 2H), 3.60 (d, J=12.6 Hz, 1H), 3.54-3.45 (m, 1H), 2.92-2.75 (m, 3H), 2.54-2.46 (m, 1H), 1.63 (d, J=6.6 Hz, 3H). Step 2: Synthesis of cyclopropyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (Compound No. 191) To a solution of cyclopropanol (33 mg, 0.57 mmol, 7.0 eq.) in MeCN (1 mL) was added CDI (93 mg, 0.57 mmol, 7.0 eq.) and the mixture stirred for 6 hours. To the mixture was added 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (35 mg, 0.082 mmol, 1.0 eq.) and the resulting solution was heated to 80° C. for 16 hours. The mixture was partitioned between aq. sol. of sat. NaHCO3and DCM and the organic phase was dried and concentrated in vacuo. The residue was purified by preparative HPLC to give the title compound as an off-white solid (32 mg, 76% yield).1H NMR (400 MHz, CDCl3) δ 8.01-7.99 (m, 1H), 7.59-7.52 (m, 2H), 7.25-7.17 (m, 2H), 6.24 (dd, J=3.5, 9.0 Hz, 1H), 5.55-5.50 (m, 1H), 5.08-4.66 (m, 1H), 4.10-3.98 (m, 5H), 3.79-3.74 (m, 1H), 3.54-3.45 (m, 1H), 2.94-2.78 (m, 3H), 2.54-2.50 (m, 1H), 1.11-1.00 (m, 3H), 0.77-0.71 (m, 4H); m/z 509 (M+H)+. Compound 192 Compound No. 192 was prepared according to the methods set forth in Example 25. For example, Compound No. 192 lists the method of “Example 25”, indicating that this compound was prepared according to the procedure of Example 25 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum):1H NMR (400 MHz, CDCl3) δ 8.20-8.19 (m, 2H), 7.59-7.51 (m, 2H), 7.21-7.17 (m, 1H), 5.56-5.47 (m, 1H), 5.09-4.64 (m, 1H), 4.21-4.10 (m, 5H), 3.86-3.68 (m, 1H), 3.53-3.46 (m, 1H), 2.96-2.80 (m, 3H), 2.56-2.49 (m, 1H), 1.11-1.06 (m, 3H), 0.74 (d, J=5.8 Hz, 4H); m/z 510 (M+H)+. Example 26 Step 1: Synthesis of 1-((3,3-difluorocyclobutoxy)carbonyl)-3-methyl-1H-imidazol-3-ium To a solution of 3,3-difluorocyclobutan-1-ol (22 mg, 0.20 mmol, 1.0 eq.) in THE (1 mL) was added CDI (40 mg, 0.24 mmol, 1.20 eq.) and the mixture stirred for 16 hours. The mixture was partitioned between water and EtOAc. The organic phase was dried and concentrated in vacuo. To a solution of the residue in MeCN (1 mL) was added iodomethane (0.038 mL, 0.61 mmol, 3.0 eq.) and the mixture stirred for 16 hours. The mixture was concentrated in vacuo to give the title compound as an orange oil (70 mg, 100% yield).1H NMR (400 MHz, CDCl3) δ 9.40 (s, 1H), 7.53-7.50 (m, 1H), 7.46-7.43 (m, 1H), 5.37-5.24 (m, 1H), 4.10 (s, 3H), 3.27-2.88 (m, 4H). Step 2: Synthesis of 3,3-difluorocyclobutyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (Compound No. 193) To a solution of tert-butyl 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (0.28 g, 0.57 mmol, 1.0 eq.) in DCM (4 mL) was added TFA (1.2 mL, 15.4 mmol, 28.1 eq.) and the mixture was stirred at RT for 1 hour. The mixture was concentrated in vacuo and was purified using a 5 g SCX-2 cartridge eluting with MeOH then 2.3 M ammonia in MeOH to give a pale yellow oil. To a solution of the pale yellow oil (1.0 g, 4.75 mmol, 1.0 eq.) and Et3N (1.3 mL, 9.49 mmol, 2.0 eq.) in MeCN (3 mL) was added a solution of 1-((3,3-difluorocyclobutoxy)carbonyl)-3-methyl-1H-imidazol-3-ium (71 mg, 0.21 mmol, 1.75 eq.) in MeCN (1 mL). The resulting solution was stirred at RT for 2 hours, filtered through celite and concentrated in vacuo. The residue was purified by preparative HPLC to give the title compound as an off-white solid (7 mg, 10% yield).1H NMR (400 MHz, CDCl3) δ 8.00 (d, J=2.9 Hz, 1H), 7.58-7.52 (m, 2H), 7.25-7.18 (m, 2H), 6.24 (dd, J=3.4, 9.0 Hz, 1H), 5.63-5.55 (m, 1H), 5.01-4.56 (m, 2H), 4.11-3.79 (m, 5H), 3.52-3.46 (m, 1H), 3.07-2.53 (m, 8H), 1.13-1.10 (m, 3H); m/z 559 (M+H)+. Compound Nos. 194 to 196 and 201 to 220 Compound Nos. 194 to 196, 201 to 207, and 215 were prepared according to the methods set forth in Example 26. For example, Compound No. 194 of Table 18 lists the method of “Example 26”, indicating that this compound was prepared according to the procedure of Example 26 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 18. Compound Nos. 207 to 214 and 216 to 217 are prepared according to the procedure of Example 19 using appropriately substituted intermediates. TABLE 18Compound Nos. 194 to 196Compound No.Analytical DataSynthesis Method1941H NMR (400 MHz, CDCl3) d 8.20 (s, 2H),Example 267.58-7.50 (m, 2H), 7.23-7.20 (m, 1H), 5.72-5.63 (m, 1H), 5.26-5.03 (m, 2H), 4.73-4.59 (m, 4H), 4.20-4.11 (m, 4H), 3.87-3.74(m, 1H), 3.55-3.47 (m, 1H), 2.96-2.79 (m,3H), 2.57-2.51 (m, 1H), 1.17-1.06 (m, 3H);m/z 548 (M + H)+.1951H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 3.0Example 26Hz, 1H), 7.56-7.50 (m, 2H), 7.21 (d, J = 31.1Hz, 2H), 6.24 (dd, J = 3.1, 9.0 Hz, 1H), 5.68-5.57 (m, 1H), 5.16-4.84 (m, 2H), 4.12-3.76(m, 5H), 3.54-3.45 (m, 1H), 2.94-2.78 (m,3H), 2.56-2.53 (m, 1H), 2.39-2.34 (m, 2H),2.15-2.03 (m, 2H), 1.86-1.77 (m, 1H), 1.68-1.59 (m, 1H), 1.14-1.06 (m, 3H); m/z 523(M + H)+.1961H NMR (400 MHz, CDCl3) δ 8.30 (2H, d,Example 26J = 4.9 Hz), 7.58 (1H, s), 7.54 (1H, dd, J = 2.4,8.4 Hz), 7.20 (1H, d, J = 8.5 Hz), 6.53 (1H, dd,J = 4.8, 4.8 Hz), 4.55 (2H, s), 4.50 (2H, s), 4.43(2H, s), 4.22 (2H, s), 4.14 (2H, s), 3.56-3.47(1H, m), 2.89 (2H, dd, J = 8.6, 12.1 Hz), 2.67(2H, s), 1.54 (3H, s), 1.51 (3H, s); m/z 512(M + H)+.201Enantiomers separated and characterized inExample 26Example 29202Enantiomers separated and characterized inExample 26Example 29203Enantiomers separated and characterized inExample 26Example 29204Enantiomers separated and characterized inExample 26Example 29205Enantiomers separated and characterized inExample 26Example 29206Enantiomers separated and characterized inExample 26Example 29 Example 27 Step 1: Synthesis of methyl 2-((5-chloro-2-nitrobenzyl)amino)butanoate To a solution of 5-chloro-2-nitrobenzaldehyde (0.68 g, 3.66 mmol, 1.0 eq.) in DCM (18 mL) was added methyl-DL-alpha-aminobutyrate hydrochloride (0.69 g, 3.66 mmol, 1.0 eq.), triethylamine (0.51 mL, 3.66 mmol, 1.0 eq.) and sodium triacetoxyborohydride (1.55 g, 7.31 mmol, 2.0 eq.). The resulting solution was stirred at RT for 36 hours, diluted with DCM, washed with an aq. sol. of sat. NaHCO3, dried, concentrated in vacuo to give the title compound as a yellow oil (0.29 g, 28% yield).1H NMR (400 MHz, CDCl3) δ 7.92 (d, J=8.7 Hz, 1H), 7.72 (d, J=2.3 Hz, 1H), 7.37 (dd, J=2.3, 8.7 Hz, 1H), 4.14-4.09 (m, 1H), 3.94 (d, J=15.4 Hz, 1H), 3.72 (s, 3H), 3.19 (t, J=6.6 Hz, 1H), 1.78-1.58 (m, 2H), 0.96 (t, J=7.4 Hz, 3H). Step 2: Synthesis of 7-chloro-3-ethyl-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepin-2-one To a solution of methyl 2-((5-chloro-2-nitrobenzyl)amino)butanoate (0.29 g, 1.01 mmol, 1.0 eq.) in acetic acid (10 mL) was added iron (0.14 g, 2.53 mmol, 2.5 eq.). The resulting suspension was heated at 110° C. for 30 minutes, cooled to RT and filtered through celite rinsing with acetic acid. The filtrate was partitioned between aq. sol. of sat. NaHCO3and EtOAc and the organic phase was dried and concentrated in vacuo. The residue was triturated with diethyl ether to give the title compound as a light brown solid (80 mg, 35% yield).1H NMR (400 MHz, CDCl3) δ 7.45 (d, J=2.0 Hz, 1H), 7.26-7.23 (m, 1H), 6.91-6.86 (m, 1H), 4.08 (d, J=13.3 Hz, 1H), 3.87 (d, J=13.4 Hz, 1H), 3.33 (t, J=6.5 Hz, 1H), 1.95-1.85 (m, 1H), 1.65-1.54 (m, 1H), 0.96-0.91 (m, 3H). Step 3: Synthesis of methyl 7-chloro-3-ethyl-2-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4] diazepine-4-carboxylate To a solution 7-chloro-3-ethyl-1,3,4,5-tetrahydro-2H-benzo[e][1,4]diazepin-2-one (75 mg, 0.33 mmol, 1.0 eq.) and Et3N (0.07 mL, 0.50 mmol, 1.5 eq.) at 0° C. in THE (3 mL) was added methyl chloroformate (0.028 mL, 0.36 mmol, 1.1 eq.) dropwise. The resulting solution was stirred at RT for 20 minutes, partitioned between DCM and an aq. sol. of sat. NaHCO3. The organics were dried, concentrated in vacuo to give the title compound as an off-white solid (0.11 g, 100% yield).1H NMR (400 MHz, CDCl3) δ 7.99-7.88 (m, 1H), 7.21-7.17 (m, 1H), 6.84-6.79 (m, 1H), 5.07-4.49 (m, 2H), 4.29-4.23 (m, 1H), 3.71-3.68 (m, 3H), 2.24-1.76 (m, 2H), 1.01-0.94 (m, 3H). Step 4: Synthesis of methyl 7-chloro-3-ethyl-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate To a suspension of methyl 7-chloro-3-ethyl-2-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (0.11 g, 0.39 mmol, 1.0 eq.) in THE (4 mL) was added 2,4-bis(4-methoxyphenyl)-2,4-dithioxo-1,3,2,4-dithiadiphosphetane (94 mg, 0.23 mmol, 0.60 eq.) and the mixture was heated to reflux for 1 hour. The mixture was allowed to cool and was concentrated in vacuo to give the title compound as a yellow solid (0.12 g, 100% yield). m/z 299.1 (M+H)+. Step 5: Synthesis of methyl 8-chloro-4-ethyl-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxylate (Compound No. 197) To a solution of methyl 7-chloro-3-ethyl-2-thioxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine-4-carboxylate (58 mg, 0.19 mmol, 1.0 eq.), was added 2-(4-fluoropyridin-2-yl)-2-azaspiro[3.3]heptane-6-carbohydrazide (50 mg, 0.21 mmol, 1.1 eq.) in dioxane (1 mL) and the mixture was heated to 100° C. for 16 hours. The mixture was allowed to cool and was concentrated in vacuo. The residue was purified by preparative HPLC to give the title compound to give the title product as an off-white solid (4 mg, 4% yield).1H NMR (400 MHz, CDCl3) δ 8.01-7.99 (m, 1H), 7.56-7.50 (m, 2H), 7.25-7.17 (m, 2H), 6.24 (dd, J=3.3, 9.0 Hz, 1H), 5.46-5.40 (m, 1H), 5.06-4.83 (m, 1H), 4.12-3.97 (m, 4H), 3.85-3.78 (m, 4H), 3.55-3.47 (m, 1H), 2.96-2.76 (m, 3H), 2.55-2.46 (m, 1H), 1.62-1.53 (m, 1H), 1.03-0.86 (m, 1H), 0.70-0.59 (m, 3H); m/z 497 (M+H)+. Compound Nos. 198 to 199 Compound Nos. 198 to 199 were prepared according to the methods set forth in Example 27. For example, Compound No. 198 of Table 19 lists the method of “Example 27”, indicating that this compound was prepared according to the procedure of Example 27 using appropriately substituted intermediates. Analytical data (NMR, mass spectrum) is also presented in Table 19. TABLE 19Compound Nos. 198 to 199Compound No.Analytical DataSynthesis Method1981H NMR (400 MHz, CDCl3) δ 8.32-8.30Example 27(m, 2H), 7.61-7.58 (m, 2H), 7.19-7.16 (m,1H), 6.54 (t, J = 4.8 Hz, 1H), 5.37 (dd, J = 5.8,8.2 Hz, 1H), 4.74-4.62 (m, 3H), 4.24-4.15(m, 4H), 3.96 (d, J = 13.7 Hz, 1H), 3.59-3.49(m, 1H), 2.97-2.80 (m, 3H), 2.55-2.48 (m,1H); m/z 450 (M + H)+.1991H NMR (400 MHz, CDCl3) δ 8.30 (2H, d,Example 27J = 4.9 Hz), 7.52-7.47 (2H, m), 7.04 (1H, d,J = 8.3 Hz), 6.53 (1H, dd, J = 4.8, 4.8 Hz), 4.48(2H, s), 4.23 (2H, s), 4.14 (2H, s), 3.76 (3H,s), 3.62-3.52 (1H, m), 2.95-2.88 (2H, m),2.71-2.64 (2H, m), 1.31-1.26 (2H, m), 1.17-1.13 (2H, m); m/z 478 (M + H)+. Example 28 Step 1: Synthesis of methyl (5-chloro-2-nitrobenzyl)serinate To a solution of 2-(bromomethyl)-4-chloro-1-nitrobenzene (0.97 g, 3.86 mmol, 1.0 eq.) in DMF (10 mL) was added DL-Serine methyl ester hydrochloride (0.6 g, 3.86 mmol, 1.0 eq.) and potassium carbonate (1.07 g, 7.71 mmol, 2.0 eq.). The resulting mixture was stirred at RT for 4 hours, concentrated in vacuo, diluted with EtOAc and washed with an aq. sol. of sat. NaHCO3. The organics were dried, concentrated in vacuo and the residue was purified by flash column chromatography eluting with 5% EtOAc in isohexane to give the title compound as a light yellow oil (0.77 g, 69% yield).1H NMR (400 MHz, CDCl3) δ 7.96-7.93 (m, 1H), 7.64 (d, J=2.3 Hz, 1H), 7.41 (dd, J=2.4, 8.7 Hz, 1H), 4.20-4.05 (m, 2H), 3.86-3.80 (m, 1H), 3.75 (s, 3H), 3.68 (dd, J=6.2, 11.3 Hz, 1H), 3.44 (dd, J=4.4, 6.3 Hz, 1H). Step 2: Synthesis of methyl 3-(5-chloro-2-nitrobenzyl)-2-oxooxazolidine-4-carboxylate To a solution methyl (5-chloro-2-nitrobenzyl)serinate (0.77 g, 2.67 mmol, 1.0 eq.) and Et3N (0.56 mL, 4.0 mmol, 1.5 eq.) at 0° C. in DCM (10 mL) was added triphosgene (0.23 mL, 0.80 mmol, 0.3 eq.) dropwise. The resulting solution was stirred at RT for 30 minutes, partitioned between DCM and an aq. sol. of sat. NaHCO3. The organics were washed with brine, dried and concentrated in vacuo. The residue was purified by flash column chromatography eluting with 20% EtOAc in isohexane to give the title compound as a light yellow solid (0.51 g, 60% yield).1H NMR (400 MHz, CDCl3) δ 8.01-7.97 (m, 1H), 7.64 (d, J=1.8 Hz, 1H), 7.43 (ddd, J=2.2, 8.7, 19.5 Hz, 1H), 4.94 (d, J=16.4 Hz, 1H), 4.79 (d, J=16.2 Hz, 1H), 4.54 (t, J=10.2 Hz, 1H), 4.45-4.38 (m, 2H), 3.80 (s, 3H). Step 3: Synthesis of 7-chloro-1,5,10,11a-tetrahydro-3H,11H-benzo[e]oxazolo[3,4-a][1,4]diazepine-3,11-dione To a solution of methyl 3-(5-chloro-2-nitrobenzyl)-2-oxooxazolidine-4-carboxylate (0.51 g, 1.62 mmol, 1.0 eq.) in acetic acid (15 mL) was added iron (0.23 g, 4.05 mmol, 2.5 eq.). The resulting suspension was heated at 110° C. for 2 hours, cooled to RT and filtered through celite rinsing with acetic acid. The filtrate was partitioned between water and EtOAc and the organic phase was dried and concentrated in vacuo. The residue was triturated with diisopropyl ether to give the title compound as a light brown solid (0.27 g, 66% yield).1H NMR (400 MHz, CDCl3) δ 7.57 (s, 1H), 7.41-7.36 (m, 2H), 6.99 (d, J=8.3 Hz, 1H), 4.94 (dd, J=5.1, 8.3 Hz, 1H), 4.56 (d, J=13.6 Hz, 1H), 4.43-4.29 (m, 3H). Step 4: Synthesis of 7-chloro-11-thioxo-5,10,11,11a-tetrahydro-1H,3H-benzo[e]oxazolo[3,4-a][1,4]diazepin-3-one To a suspension of 7-chloro-1,5,10,11a-tetrahydro-3H,11H-benzo[e]oxazolo[3,4-a][1,4]diazepine-3,11-dione (0.27 g, 1.07 mmol, 1.0 eq.) in THF (5 mL) was added 2,4-bis(4-methoxyphenyl)-2,4-dithioxo-1,3,2,4-dithiadiphosphetane (0.26 g, 0.64 mmol, 0.60 eq.) and the mixture was heated to reflux for 2 hours. The mixture was allowed to cool and was concentrated in vacuo. The residue was purified by flash column chromatography eluting with 10-100% EtOAc in isohexane to give the title compound as a light yellow solid (0.31 g, 100% yield). 1H NMR (400 MHz, DMSO-d6) δ 9.47-9.43 (m, 1H), 7.42 (d, J=9.6 Hz, 2H), 7.06 (d, J=7.8 Hz, 1H), 5.33 (dd, J=5.3, 8.8 Hz, 1H), 4.59 (d, J=13.9 Hz, 1H), 4.46-4.31 (m, 3H). Step 5: Synthesis of 7-chloro-3-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-13,13a-dihydro-9H,11H-benzo[e]oxazolo[3,4-a][1,2,4]triazolo[3,4-c][1,4]diazepin-11-one (Compound No. 200) To a solution of 7-chloro-11-thioxo-5,10,11,11a-tetrahydro-1H,3H-benzo[e] oxazolo[3,4-a][1,4]diazepin-3-one (40 mg, 0.15 mmol, 1.0 eq.), was added 2-(4-fluoropyridin-2-yl)-2-azaspiro[3.3]heptane-6-carbohydrazide (41 mg, 0.16 mmol, 1.1 eq.) in dioxane (1 mL) and the mixture was heated to 100° C. for 16 hours. The mixture was allowed to cool and was concentrated in vacuo. The residue was purified by preparative HPLC to give the title compound to give the title product as an off-white solid (7 mg, 11% yield).1H NMR (400 MHz, CDCl3) δ 8.01-7.99 (m, 1H), 7.61-7.57 (m, 2H), 7.25-7.14 (m, 2H), 6.24 (dd, J=3.5, 9.0 Hz, 1H), 5.37 (dd, J=5.8, 8.3 Hz, 1H), 4.75-4.61 (m, 3H), 4.10-3.93 (m, 5H), 3.58-3.48 (m, 1H), 2.94-2.76 (m, 3H), 2.56-2.49 (m, 1H); m/z 467 (M+H)+. Example 29 Isomer Separation Homochiral isomer A and homochiral isomer B were isolated using Supercritical Fluid Chromatography (SFC) chiral separation of the corresponding racemates. Either a Waters Thar Prep100 preparative SFC system (P200 CO2 pump, 2545 modifier pump, 2998 UV/VIS detector, 2767 liquid handler with Stacked Injection Module) or Waters Thar Investigator semi preparative system (Waters Fluid Delivery Module, 2998 UV/VIS detector, Waters Fraction Collection Module) were used. Where the Waters 2767 liquid handler was used it acted as both auto-sampler and fraction collector. The compounds were purified using an appropriate column selected from the following: YMC Amylose-C, YMC Cellulose-C, YMC Cellulose-SC, Phenomenex LUX Cellulose-3 or Phenomenex LUX Cellulose-4. Appropriate isocratic methods were selected based on methanol, ethanol or isopropanol solvent systems under un-modified or basic conditions. The standard method used was modifier/CO2, 100 ml/min (or as appropriate), 120 Bar backpressure, 40° C. column temperature. The modifier used under basic conditions was diethyl amine (0.1% V/V). The purification was controlled either by Waters Fractionlynx or Waters Chromscope software through monitoring at 210-400 nm and triggered a threshold collection value at an appropriate wavelength. Collected fractions were analysed by SFC (Waters/Thar SFC systems with Waters SQD or Waters UPCC with Waters QDa). The fractions that contained the desired product were concentrated by vacuum centrifugation. The following compounds were purified using SFC chiral separation. TABLE 20Compound Separated by Chiral SFCRacemate Compound No.IsomerData142Isomer A1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.55-7.51 (m, 2H), 7.25-7.17 (m,2H), 6.23 (dd, J = 3.5, 9.0 Hz, 1H), 5.60-5.55(m, 1H), 5.05-4.98 (m, 1H), 4.11-4.03 (m,2H), 3.99 (s, 2H), 3.98-3.92 (m, 1H), 3.78 (s,3H), 3.54-3.45 (m, 1H), 2.94-2.79 (m, 3H),2.59-2.51 (m, 1H), 1.21-1.03 (m, 3H); m/z483 (M + H)+.142Isomer B1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.55-7.51 (m, 2H), 7.25-7.17 (m,2H), 6.24 (dd, J = 3.4, 9.2 Hz, 1H), 5.61-5.58(m, 1H), 5.06-4.98 (m, 1H), 4.11-4.03 (m,2H), 3.99 (s, 2H), 3.98-3.92 (m, 1H), 3.78 (s,3H), 3.54-3.45 (m, 1H), 2.94-2.79 (m, 3H),2.56-2.52 (m, 1H), 1.19-1.11 (m, 3H); m/z483 (M + H)+.174Isomer A1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 3.0Hz, 1H), 7.56 (d, J = 7.0 Hz, 2H), 7.25-7.20 (m,2H), 6.24 (dd, J = 3.4, 9.0 Hz, 1H), 6.00-5.95(m, 1H), 5.18-5.11 (m, 1H), 4.12-4.08 (m,2H), 4.02-3.99 (m, 2H), 3.85-3.71 (m, 1H),3.54-3.49 (m, 1H), 2.98-2.90 (m, 1H), 2.87-2.78 (m, 2H), 2.56-2.50 (m, 1H), 1.51-1.22(m, 4H), 1.21-1.09 (m, 3H); m/z 511 (M + H)+.184Isomer A1H NMR (400 MHz, CDCl3) δ 8.59-8.56 (m,1H), 8.21-8.15 (m, 1H), 7.56-7.50 (m, 2H),7.20-7.15 (m, 1H), 6.17 (d, J = 5.3 Hz, 1H),5.65-5.59 (m, 1H), 5.31-4.37 (m, 1H), 4.22-4.12 (m, 4H), 3.80-3.78 (m, 4H), 3.55-3.47(m, 1H), 2.98-2.54 (m, 4H), 1.26-1.14 (m,3H); m/z 466 (M + H)+.184Isomer B1H NMR (400 MHz, CDCl3) δ 8.57 (s, 1H),8.21-8.15 (m, 1H), 7.60-7.51 (m, 2H), 7.19-7.15 (m, 1H), 6.17 (dd, J = 1.2, 6.1 Hz, 1H), 5.59-5.57 (m, 1H), 5.03-4.74 (m, 1H), 4.21-4.12(m, 4H), 3.90-3.78 (m, 4H), 3.54-3.46 (m,1H), 2.97-2.57 (m, 4H), 1.20-1.05 (m, 3H);m/z 466 (M + H)+.191Isomer A1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.55-7.51 (m, 2H), 7.25-7.16 (m,2H), 6.23 (dd, J = 3.1, 9.0 Hz, 1H), 5.55-5.45(m, 1H), 5.27-4.33 (m, 1H), 4.09-3.98 (m,5H), 3.94-3.64 (m, 1H), 3.54-3.45 (m, 1H),2.93-2.78 (m, 3H), 2.56-2.53 (m, 1H), 1.11-1.04 (m, 3H), 0.74 (d, J = 6.0 Hz, 4H); m/z 510(M + H)+.191Isomer B1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.55-7.51 (m, 2H), 7.25-7.17 (m,2H), 6.24 (dd, J = 3.4, 9.0 Hz, 1H), 5.56-5.46(m, 1H), 5.07-4.68 (m, 1H), 4.15-3.79 (m,6H), 3.54-3.45 (m, 1H), 2.94-2.53 (m, 4H),1.14-0.97 (m, 3H), 0.74 (d, J = 6.0 Hz, 4H); m/z509 (M + H)+.179Isomer A1H NMR (400 MHz, CDCl3) δ 7.99 (d, J = 2.9Hz, 1H), 7.49 (dd, J = 2.4, 8.4 Hz, 1H), 7.43 (d,J = 2.4 Hz, 1H), 7.25-7.19 (m, 1H), 7.15 (d,J = 8.4 Hz, 1H), 6.23 (dd, J = 3.5, 9.0 Hz, 1H),4.10-4.03 (m, 2H), 3.99 (s, 2H), 3.51-3.44(m, 3H), 3.30-3.26 (m, 1H), 2.90-2.76 (m,3H), 2.56-2.48 (m, 1H), 2.41 (s, 3H); m/z 439(M + H)+.179Isomer B1H NMR (400 MHz, CDCl3) δ 7.99 (d, J = 2.9Hz, 1H), 7.52-7.48 (m, 1H), 7.43 (d, J = 2.4 Hz,1H), 7.25-7.19 (m, 1H), 7.15 (d, J = 8.4 Hz,1H), 6.23 (dd, J = 3.6, 8.8 Hz, 1H), 4.10-4.03(m, 2H), 3.99 (s, 2H), 3.51-3.44 (m, 3H), 3.31-3.27 (m, 1H), 2.90-2.76 (m, 3H), 2.55-2.48(m, 1H), 2.41 (s, 3H); m/z 439 (M + H)+.163Isomer A1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.8Hz, 1H), 7.58-7.55 (m, 1H), 7.52 (dd, J = 2.0,8.3 Hz, 1H), 7.25-7.17 (m, 2H), 6.24 (dd,J = 3.4, 9.0 Hz, 1H), 5.62-5.57 (m, 1H), 5.12-5.06 (m, 1H), 5.04-4.94 (m, 1H), 4.10-3.98(m, 4H), 3.90-3.68 (m, 1H), 3.53-3.45 (m,1H), 2.95-2.77 (m, 3H), 2.55-2.52 (m, 1H),1.33-1.27 (m, 6H), 1.17-1.10 (m, 3H); m/z511 (M + H)+.163Isomer B1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.8Hz, 1H), 7.58-7.55 (m, 1H), 7.52 (dd, J = 2.0,8.3 Hz, 1H), 7.25-7.17 (m, 2H), 6.24 (dd,J = 3.4, 9.0 Hz, 1H), 5.64-5.59 (m, 1H), 5.10-5.05 (m, 1H), 5.04-4.94 (m, 1H), 4.10-3.98(m, 4H), 3.90-3.68 (m, 1H), 3.53-3.45 (m,1H), 2.95-2.77 (m, 3H), 2.55-2.52 (m, 1H),1.33-1.27 (m, 6H), 1.16-1.11 (m, 3H); m/z511 (M + H)+.165Isomer A1H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.5,2.8 Hz, 1H), 7.85 (d, J = 2.8 Hz, 1H), 7.77 (d,J = 1.4 Hz, 1H), 7.52 (dd, J = 1.9, 8.3 Hz, 2H),7.18 (d, J = 8.5 Hz, 1H), 5.68-5.48 (m, 1H),5.17-4.91 (m, 1H), 4.20-4.12 (m, 4H), 3.91-3.82 (m, 1H), 3.79 (s, 3H), 3.54-3.46 (m, 1H),2.97-2.83 (m, 3H), 2.62 (s, 1H), 1.16-1.13(m, 3H); m/z 466 (M + H)+.165Isomer B1H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.5,2.8 Hz, 1H), 7.85 (d, J = 2.8 Hz, 1H), 7.77 (d,J = 1.4 Hz, 1H), 7.53 (dd, J = 1.9, 8.3 Hz, 2H),7.18 (d, J = 8.5 Hz, 1H), 5.71-5.49 (m, 1H),5.17-4.91 (m, 1H), 4.20-4.12 (m, 4H), 3.90-3.81 (m, 1H), 3.79 (s, 3H), 3.53-3.44 (m, 1H),2.97-2.83 (m, 3H), 2.62 (s, 1H), 1.17-1.14(m, 3H); m/z 466 (M + H)+.166Isomer A1H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 1.5,2.8 Hz, 1H), 7.84 (d, J = 2.8 Hz, 1H), 7.77 (d,J = 1.4 Hz, 1H), 7.53 (dd, J = 1.9, 8.3 Hz, 2H),7.18 (d, J = 8.5 Hz, 1H), 5.71-5.49 (m, 1H),5.17-4.91 (m, 1H), 4.20-4.12 (m, 4H), 3.90-3.82 (m, 1H), 3.79 (s, 3H), 3.52-3.45 (m, 1H),2.97-2.83 (m, 3H), 2.62 (s, 1H), 1.17-1.12(m, 3H); m/z 466 (M + H)+.166Isomer B1H NMR (400 MHz, CDCl3) δ 8.30 (d, J = 4.8Hz, 2H), 7.57-7.51 (m, 2H), 7.21 (d, J = 8.5 Hz,1H), 6.53 (t, J = 4.8 Hz, 1H), 5.67-5.53 (m,1H), 5.11-4.93 (m, 1H), 4.24-4.13 (m, 4H),3.92-3.83 (m, 1H), 3.78 (s, 3H), 3.56-3.46(m, 1H), 2.97-2.82 (m, 3H), 2.54-2.53 (m,1H), 1.15-1.11 (m, 3H); m/z 466 (M + H)+.167Isomer A1H NMR (400 MHz, CDCl3) δ 8.30 (d, J = 4.5Hz, 2H), 7.57-7.50 (m, 2H), 7.20 (d, J = 8.5 Hz,1H), 6.53 (t, J = 4.8 Hz, 1H), 5.66-5.62 (m,1H), 5.04-4.94 (m, 2H), 4.27-4.17 (m, 2H),4.13 (s, 2H), 3.81-3.75 (m, 1H), 3.56-3.47(m, 1H), 2.97-2.81 (m, 3H), 2.60-2.49 (m,1H), 1.33-1.27 (m, 6H), 1.14-1.08 (m, 3H);m/z 494 (M + H)+.167Isomer B1H NMR (400 MHz, CDCl3) δ 8.30 (d, J = 4.8Hz, 2H), 7.57-7.50 (m, 2H), 7.20 (d, J = 8.5 Hz,1H), 6.53 (t, J = 4.8 Hz, 1H), 5.69-5.55 (m,1H), 5.04-4.94 (m, 2H), 4.24-4.13 (m, 4H),3.81-3.74 (m, 1H), 3.56-3.47 (m, 1H), 2.97-2.81 (m, 3H), 2.60-2.48 (m, 1H), 1.33-1.27(m, 6H), 1.13-1.09 (m, 3H); m/z 494 (M + H)+.168Isomer A1H NMR (400 MHz, CDCl3) δ 8.32-8.28 (m,2H), 7.56-7.51 (m, 2H), 7.20 (d, J = 8.1 Hz,1H), 6.55-6.51 (m, 1H), 5.70-5.56 (m, 1H),5.16-4.95 (m, 1H), 4.24-4.13 (m, 4H), 4.11(s, 2H), 3.95-3.72 (m, 1H), 3.56-3.47 (m,1H), 2.97-2.81 (m, 3H), 2.60-2.47 (m, 1H),1.32-1.30 (m, 3H), 1.19-1.04 (m, 3H); m/z480 (M + H)+.168Isomer B1H NMR (400 MHz, CDCl3) δ 8.31-8.28 (m,2H), 7.56-7.51 (m, 2H), 7.20 (d, J = 8.1 Hz,1H), 6.55-6.51 (m, 1H), 5.68-5.57 (m, 1H),5.12-4.95 (m, 1H), 4.24-4.13 (m, 4H), 4.12(s, 2H), 3.85-3.80 (m, 1H), 3.56-3.47 (m,1H), 2.97-2.82 (m, 3H), 2.59-2.47 (m, 1H),1.32-1.30 (m, 3H), 1.20-1.05 (m, 3H); m/z480 (M + H)+.169Isomer A1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.55-7.50 (m, 2H), 7.25-7.17 (m,2H), 6.24 (dd, J = 3.3, 9.0 Hz, 1H), 5.60-5.54(m, 1H), 5.10-5.03 (m, 1H), 4.26-4.18 (m,2H), 4.10-3.99 (m, 4H), 3.91-3.67 (m, 1H),3.55-3.45 (m, 1H), 2.94-2.78 (m, 3H), 2.58-2.49 (m, 1H), 1.31 (t, J = 7.1 Hz, 3H), 1.20-1.04 (m, 3H); m/z 497 (M + H)+.169Isomer B1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.55-7.50 (m, 2H), 7.25-7.17 (m,2H), 6.24 (dd, J = 3.3, 9.0 Hz, 1H), 5.61-5.56(m, 1H), 5.10-5.01 (m, 1H), 4.26-4.18 (m,2H), 4.10-3.98 (m, 4H), 3.91-3.68 (m, 1H),3.55-3.45 (m, 1H), 2.94-2.78 (m, 3H), 2.58-2.49 (m, 1H), 1.31 (t, J = 7.1 Hz, 3H), 1.19-1.05 (m, 3H); m/z 497 (M + H)+.181Isomer A1H NMR (400 MHz, DMSO-d6) δ 7.96 (dd,J = 1.5, 5.0 Hz, 1H), 7.67-7.65 (m, 2H), 7.56(d, J = 8.7 Hz, 1H), 7.43-7.37 (m, 1H), 6.53(dd, J = 5.0, 6.8 Hz, 1H), 6.26 (d, J = 8.3 Hz, 1H),5.61-5.44 (m, 1H), 5.02-4.95 (m, 1H), 4.14-4.10 (m, 1H), 3.96-3.87 (m, 2H), 3.77 (q,J = 8.4 Hz, 2H), 3.70-3.61 (m, 1H), 2.70 (d,J = 8.0 Hz, 2H), 2.41-2.26 (m, 2H), 1.40-1.13(m, 4H), 1.00-0.82 (m, 3H); m/z 493 (M + H)+.181Isomer B1H NMR (400 MHz, DMSO-d6) δ 7.96 (dd,J = 1.5, 5.0 Hz, 1H), 7.67-7.65 (m, 2H), 7.56(d, J = 8.7 Hz, 1H), 7.43-7.38 (m, 1H), 6.53(dd, J = 5.0, 6.8 Hz, 1H), 6.26 (d, J = 8.3 Hz, 1H),5.62-5.46 (m, 1H), 5.01-4.95 (m, 1H), 4.14-4.10 (m, 1H), 3.96-3.87 (m, 2H), 3.78 (q,J = 8.4 Hz, 2H), 3.70-3.61 (m, 1H), 2.70 (d,J = 8.0 Hz, 2H), 2.41-2.26 (m, 2H), 1.41-1.15(m, 4H), 0.98-0.83 (m, 3H); m/z 493 (M + H)+.178Isomer A1H NMR (400 MHz, DMSO-d6) δ 7.95 (d,J = 2.9 Hz, 1H), 7.66 (dd, J = 2.1, 8.5 Hz, 1H),7.59-7.51 (m, 2H), 7.42-7.37 (m, 1H), 6.31(dd, J = 3.5, 9.0 Hz, 1H), 5.60-5.58 (m, 1H),5.13-5.06 (m, 1H), 4.02-3.95 (m, 1H), 3.90(dd, J = 8.3, 23.5 Hz, 2H), 3.77 (dd, J = 8.7, 18.3Hz, 2H), 3.71-3.62 (m, 1H), 2.73-2.70 (m,2H), 2.35-2.25 (m, 2H), 1.63-1.55 (m, 6H),0.85-0.79 (m, 3H); m/z 513 (M + H)+.178Isomer B1H NMR (400 MHz, DMSO-d6) δ 7.95 (d,J = 2.9 Hz, 1H), 7.66 (dd, J = 2.1, 8.5 Hz, 1H),7.58-7.51 (m, 2H), 7.42-7.37 (m, 1H), 6.31(dd, J = 3.4, 9.0 Hz, 1H), 5.59-5.57 (m, 1H),5.12-5.03 (m, 1H), 4.02-3.94 (m, 1H), 3.90(dd, J = 8.3, 23.5 Hz, 2H), 3.77 (dd, J = 8.7, 18.3Hz, 2H), 3.71-3.62 (m, 1H), 2.73-2.70 (m,2H), 2.35-2.25 (m, 2H), 1.63-1.55 (m, 6H),0.84-0.78 (m, 3H); m/z 513 (M + H)+.170Isomer A1H NMR (400 MHz, CDCl3) δ 8.20 (d, J = 0.9Hz, 2H), 7.53 (dd, J = 1.8, 8.3 Hz, 2H), 7.21-7.18 (m, 1H), 5.64-5.54 (m, 1H), 5.19-4.61(m, 1H), 4.21-4.10 (m, 4H), 4.01-3.64 (m,4H), 3.55-3.46 (m, 1H), 2.96-2.76 (m, 3H),2.56-2.49 (m, 1H), 1.18-1.08 (m, 3H); m/z484 (M + H)+.170Isomer B1H NMR (400 MHz, CDCl3) δ 8.20 (d, J = 0.8Hz, 2H), 7.54 (dd, J = 2.0, 8.3 Hz, 2H), 7.21-7.18 (m, 1H), 5.65-5.55 (m, 1H), 5.02-4.76(m, 1H), 4.21-4.10 (m, 4H), 3.88-3.79 (m,4H), 3.55-3.46 (m, 1H), 2.96-2.52 (m, 4H),1.23-1.03 (m, 3H); m/z 484 (M + H)+.192Isomer A1H NMR (400 MHz, CDCl3) δ 8.20-8.19 (m,2H), 7.56-7.51 (m, 2H), 7.21-7.17 (m, 1H),5.59-5.45 (m, 1H), 5.23-4.36 (m, 1H), 4.21-4.10 (m, 5H), 3.80 (s, 1H), 3.55-3.46 (m, 1H),2.96-2.78 (m, 3H), 2.57-2.48 (m, 1H), 1.10-1.02 (m, 3H), 0.74 (d, J = 5.9 Hz, 4H); m/z 510(M + H)+.192Isomer B1H NMR (400 MHz, CDCl3) δ 8.19 (s, 2H),7.53 (dd, J = 1.6, 8.5 Hz, 2H), 7.21-7.17 (m,1H), 5.54-5.45 (m, 1H), 5.06-4.65 (m, 1H),4.21-4.09 (m, 5H), 3.89-3.71 (m, 1H), 3.55-3.46 (m, 1H), 2.96-2.54 (m, 4H), 1.18-0.98(m, 3H), 0.75-0.71 (m, 4H); m/z 510 (M + H)+.187Isomer A1H NMR (400 MHz, CDCl3) δ 8.57 (s, 1H),8.16 (d, J = 5.9 Hz, 1H), 7.58-7.51 (m, 2H),7.18-7.15 (m, 1H), 6.17-6.15 (m, 1H), 5.66-5.54 (m, 1H), 5.11-4.96 (m, 2H), 4.20-4.10(m, 4H), 3.83-3.69 (m, 1H), 3.55-3.47 (m,1H), 2.97-2.58 (m, 4H), 1.35-1.08 (m, 9H);m/z 494 (M + H)+.187Isomer B1H NMR (400 MHz, CDCl3) δ 8.57-8.56 (m,1H), 8.18-8.15 (m, 1H), 7.57-7.49 (m, 2H),7.19-7.15 (m, 1H), 6.16 (dd, J = 1.2, 6.0 Hz,1H), 5.74-5.39 (m, 1H), 5.25-4.78 (m, 2H),4.20-4.10 (m, 4H), 3.98-3.60 (m, 1H), 3.55-3.47 (m, 1H), 2.98-2.49 (m, 4H), 1.34-1.26(m, 9H); m/z 494 (M + H)+.171Isomer A1H NMR (400 MHz, CDCl3) δ 8.30 (d, J = 4.8Hz, 2H), 7.58-7.50 (m, 2H), 7.22-7.17 (m,1H), 6.53 (t, J = 4.8 Hz, 1H), 5.56-5.51 (m,1H), 5.23-4.36 (m, 1H), 4.28-3.60 (m, 6H),3.56-3.47 (m, 1H), 2.97-2.81 (m, 3H), 2.53(d, J = 2.0 Hz, 1H), 1.22-0.87 (m, 3H), 0.77-0.71 (m, 4H); m/z 492 (M + H)+.171Isomer B1H NMR (400 MHz, CDCl3) δ 8.32-8.28 (m,2H), 7.58-7.50 (m, 2H), 7.23-7.17 (m, 1H),6.53 (t, J = 4.7 Hz, 1H), 5.56-5.51 (m, 1H),5.29-4.34 (m, 1H), 4.25-4.11 (m, 5H), 3.94-3.60 (m, 1H), 3.57-3.47 (m, 1H), 2.98-2.53(m, 4H), 1.11-0.99 (m, 3H), 0.78-0.70 (m,4H); m/z 492 (M + H)+.194Isomer A1H NMR (400 MHz, CDCl3) δ 8.20 (d, J = 0.9Hz, 2H), 7.59-7.50 (m, 2H), 7.23-7.19 (m,1H), 5.72-5.61 (m, 1H), 5.29-4.51 (m, 6H),4.21-4.10 (m, 4H), 3.89-3.70 (m, 1H), 3.55-3.47 (m, 1H), 2.97-2.81 (m, 3H), 2.65-2.44(m, 1H), 1.22-1.01 (m, 3H); m/z 548 (M + H)+.194Isomer B1H NMR (400 MHz, CDCl3) δ 8.20 (d, J = 0.8Hz, 2H), 7.58-7.50 (m, 2H), 7.23-7.19 (m,1H), 5.72-5.62 (m, 1H), 5.24-4.97 (m, 2H),4.73-4.59 (m, 4H), 4.22-4.10 (m, 4H), 3.86-3.73 (m, 1H), 3.54-3.47 (m, 1H), 2.97-2.51(m, 4H), 1.18-1.09 (m, 3H); m/z 548 (M + H)+.172Isomer A1H NMR (400 MHz, CDCl3) δ 8.20 (m, 2H),7.56-7.49 (m, 2H), 7.21-7.17 (m, 1H), 5.63-5.61 (m, 1H), 5.23-4.44 (m, 1H), 4.27-3.62(m, 7H), 3.54-3.46 (m, 1H), 2.96-2.78 (m,3H), 2.53-2.51 (m, 1H), 1.31 (t, J = 7.1 Hz,3H), 1.13 (m, 3H); m/z 498 (M + H)+.172Isomer B1H NMR (400 MHz, CDCl3) δ 8.25-8.19 (m,2H), 7.56-7.50 (m, 2H), 7.24-7.19 (m, 1H),5.72-5.56 (m, 1H), 5.06-4.78 (m, 1H), 4.23-3.81 (m, 7H), 3.55-3.47 (m, 1H), 2.98-2.54(m, 4H), 1.38-1.12 (m, 6H); m/z 498 (M + H)+.188Isomer A1H NMR (400 MHz, CDCl3) δ 8.35-8.32 (m,1H), 8.00-7.98 (m, 1H), 7.56-7.50 (m, 2H),7.19-7.15 (m, 1H), 5.63-5.55 (m, 1H), 5.29-4.55 (m, 1H), 4.37-4.30 (m, 4H), 3.95-3.69(m, 4H), 3.54-3.44 (m, 1H), 2.98-2.51 (m,4H), 1.23-1.04 (m, 3H); m/z 484 (M + H)+.188Isomer B1H NMR (400 MHz, CDCl3) δ 8.37-8.32 (m,1H), 7.99 (d, J = 4.3 Hz, 1H), 7.53 (d, J = 8.4 Hz,2H), 7.20-7.15 (m, 1H), 5.60-5.60 (m, 1H),5.09-4.74 (m, 1H), 4.35-4.31 (m, 4H), 3.81-3.77 (m, 4H), 3.50-3.48 (m, 1H), 2.96-2.82(m, 3H), 2.69-2.51 (m, 1H), 1.31-1.03 (m,3H); m/z 484 (M + H)+.201Isomer A1H NMR (400 MHz, CDCl3) δ 8.20-8.19 (m,2H), 7.57-7.50 (m, 2H), 7.22-7.16 (m, 1H),5.63-5.56 (m, 1H), 5.24-4.66 (m, 2H), 4.21-4.10 (m, 4H), 3.77-3.70 (m, 1H), 3.56-3.46(m, 1H), 2.97-2.79 (m, 3H), 2.55-2.52 (m,1H), 1.33-1.27 (m, 6H), 1.13-1.07 (m, 3H);m/z 512 (M + H)+.201Isomer B1H NMR (400 MHz, CDCl3) δ 8.20-8.19 (m,2H), 7.55-7.50 (m, 2H), 7.21-7.18 (m, 1H),5.67-5.54 (m, 1H), 5.09-4.94 (m, 2H), 4.21-4.10 (m, 4H), 3.86-3.65 (m, 1H), 3.56-3.46(m, 1H), 2.97-2.52 (m, 4H), 1.34-1.09 (m,9H); m/z 512 (M + H)+.202Isomer A1H NMR (400 MHz, DMSO-d6) δ 8.21 (d,J = 4.8 Hz, 2H), 7.60-7.55 (m, 2H), 7.50-7.45(m, 1H), 6.54 (t, J = 4.8 Hz, 1H), 5.28-4.51 (m,7H), 4.02-3.81 (m, 5H), 3.61-3.55 (m, 1H),2.65-2.53 (m, 2H), 2.36-2.25 (m, 2H), 1.11-0.81 (m, 3H); m/z 530 (M + H)+.202Isomer B1H NMR (400 MHz, DMSO-d6) δ 8.38-8.35(m, 2H), 7.79-7.71 (m, 2H), 7.66-7.61 (m,1H), 6.69 (t, J = 4.8 Hz, 1H), 5.57-4.90 (m,3H), 4.82-4.76 (m, 2H), 4.73-4.64 (m, 2H),4.57-3.85 (m, 5H), 3.79-3.70 (m, 1H), 2.73-2.70 (m, 2H), 2.50-2.40 (m, 2H), 1.22-1.02(m, 3H); m/z 530 (M + H)+.203Isomer A1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.58-7.50 (m, 2H), 7.23-7.18 (m,2H), 6.23 (dd, J = 3.1, 8.9 Hz, 1H), 5.77-5.56(m, 1H), 5.19-5.18 (m, 2H), 4.71-4.69 (m,2H), 4.61-4.57 (m, 2H), 4.10-3.99 (m, 4H),3.87-3.72 (m, 1H), 3.55-3.48 (m, 1H), 2.94-2.80 (m, 3H), 2.56-2.48 (m, 1H), 1.17-1.07(m, 3H); m/z 547 (M + H)+.203Isomer B1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 3.0Hz, 1H), 7.58-7.51 (m, 2H), 7.25-7.19 (m,2H), 6.24 (dd, J = 3.4, 9.0 Hz, 1H), 5.72-5.62(m, 1H), 5.24-5.17 (m, 2H), 4.72-4.57 (m,4H), 4.10-3.99 (m, 4H), 3.88-3.71 (m, 1H),3.55-3.46 (m, 1H), 2.94-2.79 (m, 3H), 2.57-2.48 (m, 1H), 1.20-1.04 (m, 3H); m/z 547(M + H)+.204Isomer A1H NMR (400 MHz, CDCl3) δ 7.57-7.44 (m,3H), 7.20-7.15 (m, 1H), 6.99-6.94 (m, 1H),6.43-6.39 (m, 1H), 5.66-5.56 (m, 1H), 5.26-4.57 (m, 2H), 4.29-3.58 (m, 5H), 3.55-3.46(m, 1H), 2.97-2.41 (m, 4H), 1.37-1.00 (m,9H); m/z 518 (M + H)+.204Isomer B1H NMR (400 MHz, CDCl3) δ 7.55-7.45 (m,3H), 7.19-7.16 (m, 1H), 6.97-6.95 (m, 1H),6.43-6.41 (m, 1H), 5.65-5.55 (m, 1H), 5.28-4.29 (m, 2H), 4.23-3.62 (m, 5H), 3.54-3.46(m, 1H), 2.95-2.79 (m, 3H), 2.60-2.51 (m,1H), 1.34-1.24 (m, 9H); m/z 518 (M + H)+.205Isomer A1H NMR (400 MHz, CDCl3) δ 8.31-8.29 (m,2H), 7.58-7.51 (m, 2H), 7.21-7.17 (m, 1H),6.53 (t, J = 4.8 Hz, 1H), 5.57-5.48 (m, 1H),5.30-4.38 (m, 1H), 4.24-4.12 (m, 4H), 3.73-3.69 (m, 1H), 3.55-3.46 (m, 1H), 2.97-2.51(m, 4H), 1.58 (s, 3H), 1.04-0.68 (m, 7H); m/z506 (M + H)+.205Isomer B1H NMR (400 MHz, CDCl3) δ 8.30 (dd, J = 1.4,4.8 Hz, 2H), 7.59-7.51 (m, 2H), 7.22-7.17(m, 1H), 6.55-6.52 (m, 1H), 5.59-5.52 (m,1H), 5.30-4.41 (m, 1H), 4.24-4.12 (m, 4H),3.97-3.62 (m, 1H), 3.54-3.47 (m, 1H), 2.97-2.77 (m, 3H), 2.58-2.52 (m, 1H), 1.59 (s, 3H),1.12-0.84 (m, 5H), 0.70-0.65 (m, 2H); m/z506 (M + H)+.206Isomer A1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.56-7.50 (m, 2H), 7.25-7.17 (m,2H), 6.23 (dd, J = 3.1, 9.0 Hz, 1H), 5.60-5.56(m, 1H), 5.04 (s, 1H), 4.52-4.45 (m, 2H), 4.09(d, J = 7.8 Hz, 1H), 4.05 (d, J = 7.9 Hz, 1H), 3.99(dd, J = 8.4, 10.4 Hz, 2H), 3.70 (s, 1H), 3.55-3.45 (m, 1H), 2.91 (t, J = 9.7 Hz, 1H), 2.81 (dd,J = 8.6, 11.9 Hz, 2H), 2.54-2.51 (m, 1H), 1.54(s, 3H), 1.52 (s, 3H), 1.11-1.08 (m, 3H); m/z543 (M + H)+.206Isomer B1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 2.9Hz, 1H), 7.56-7.50 (m, 2H), 7.25-7.17 (m,2H), 6.24 (dd, J = 3.1, 9.0 Hz, 1H), 5.54-5.53(m, 1H), 5.04 (s, 1H), 4.53-4.49 (m, 2H), 4.10(d, J = 7.7 Hz, 1H), 4.05 (d, J = 7.9 Hz, 1H), 3.99(dd, J = 8.4, 10.3 Hz, 2H), 3.70 (s, 1H), 3.55-3.45 (m, 1H), 2.91 (t, J = 9.8 Hz, 1H), 2.81 (dd,J = 7.7, 12.2 Hz, 2H), 2.53-2.51 (m, 1H), 1.54(s, 3H), 1.52 (s, 3H), 1.13-1.09 (m, 3H); m/z543 (M + H)+. Example 30 Step 1: Synthesis of tert-butyl 6-(hydrazinecarbonyl)-2-azaspiro[3.3]heptane-2-carboxylate To a stirred solution of 2-(tert-butoxycarbonyl)-2-azaspiro[3.3]heptane-6-carboxylic acid (0.26 g, 1 mmol, 1 eq.) in THE (5 mL) was added 1-1′-Carbonyldiimidazole (0.19 g, 1.2 mmol, 1.2 eq.) and the mixture was stirred at RT overnight. The resulting mixture was added to a solution of hydrazine monohydrate (0.07 mL, 1.4 mmol, 1.4 eq.) in THE (10 mL) and stirred at RT overnight. The mixture was diluted with brine and extracted with ethyl acetate. The organic phase was separated, washed with brine, dried (MgSO4), filtered and concentrated under reduced pressure to afford the title compound as a white solid (0.29 g, 100% yield). This material was used without further purification.1H NMR (400 MHz, CDCl3) δ 6.61-6.59 (m, 1H), 3.91-3.80 (m, 6H), 2.81-2.73 (m, 1H), 2.48-2.43 (m, 2H), 2.35-2.30 (m, 2H), 1.40 (s, 9H). Step 2: Synthesis of tert-butyl (4-chloro-2-((cyclopropylamino)methyl)phenyl)carbamate A mixture of tert-butyl (4-chloro-2-formylphenyl)carbamatee (5.122 g, 20.03 mmol, 1 eq.) and cyclopropanamine (2.08 mL, 30.05 mmol, 1.5 eq.) in MeOH (65.2 mL) was stirred for 5 h at 60° C., allowed to cool to RT, and then stirred 18 h. THF (30 mL) was added to the reaction mixture, followed by solid NaCNBH3(2.517 g, 40.06 mmol, 2 eq.). The reaction mixture was stirred at RT for 2 hours. Acetic acid (2.29 mL, 40.06 mmol, 2 eq) was added and stirring was continued for 20 min. The reaction mixture was concentrated under reduced pressure, and the resultant crude residue was diluted with EtOAc (150 mL) and washed with sat. aq. NaHCO3(100 mL). The aqueous later was collected, and the organic layer was further washed with sat. aq. NaHCO3(2×50 mL). The combined aqueous washes were back-extracted with EtOAc (1×100 mL) and the combined organic phases were dried over MgSO4, filtered to remove solid material, and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, 0-30% EtOAc/Hex—2% DCM additive) to afford tert-butyl (4-chloro-2-((cyclopropylamino)methyl)phenyl)carbamate (5.4 g, 91% yield) as a white solid. LCMS (ESI): m/z 297.1 (M+H); Retention time: 3.40 min (50-100% ACN/H2O, method 3). Step 3: Synthesis of tert-butyl (4-chloro-2-(((cyanomethyl)(cyclopropyl)amino)methyl)phenyl) carbamate A mixture of K2CO3(1.87 g, 13.5 mmol, 2 eq.), KI (0.671 g, 4.04 mmol, 0.6 eq.), 2-chloroacetonitrile (0.855 mL, 13.5 mmol, 2 eq.), and tert-butyl (4-chloro-2-((cyclopropylamino)methyl)phenyl)carbamate (2.00 g, 6.74 mmol, 1 eq.), added in sequence, was stirred in ACN (15 mL) under an atmosphere of N2for 18 h at 75° C. The reaction mixture was allowed to cool to RT, diluted with EtOAc (100 mL), and washed with sat aq. NaHCO3(100 mL). The organic phase was collected, and the aq. phase was extracted with EtOAc (2×100 mL). The combined organic extracts were washed with 15% aq. Na2S2O3(2×100 mL), brine (1×100 mL), and dried over MgSO4. Solids were removed by vacuum filtration and the filtrate was concentrated under reduced pressure to afford tert-butyl (4-chloro-2-(((cyanomethyl)(cyclopropyl)amino) methyl)phenyl) carbamate (2.548 g, 88% pure, 99% yield) as a brown oil. The crude material was taken on to the next step without further purification. LCMS (ESI): m/z 336.1 (M+H); Retention time: 3.80 min (50-100% ACN/H2O, method 3). Step 4: Synthesis of 7-chloro-4-cyclopropyl-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine A solution of acetyl chloride (9.64 mL, 134.6 mmol, 20 eq.) in isopropanol (55.6 mL) was stirred for 20 min prior to slow addition to a solution of tert-butyl (4-chloro-2-(((cyanomethyl)(cyclopropyl)amino) methyl)phenyl) carbamate (2.26 g, 6.730 mmol, 1 eq.) in isopropanol (67.3 mL) and stirred for 60 h at 50° C. The reaction mixture was concentrated under reduced pressure and the crude residue was diluted with EtOAc (125 mL). The crude mixture was washed with sat aq. NaHCO3(1×125 mL). The organic phase was collected, and the aqueous phase was extracted with EtOAc (3×50 mL). The combined organics were washed with brine (1×50 mL), dried over MgSO4, and solids were removed by vacuum filtration. The filtrate was concentrated under reduced pressure, and the crude residue was purified by column chromatography (SiO2, 0-100% EtOAc/Hex, 0-100% DCM/EtOAc, 0-12% MeOH/DCM) to afford 7-chloro-4-cyclopropyl-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine (0.721 g, 45.5% yield) as a light brown solid. LCMS (ESI): m/z 236.0 (M+H); Retention time: 1.37 min (50-100% ACN/H2O, method 3). Step 5: Synthesis of tert-butyl 6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo [4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate A mixture of 7-chloro-4-(2-methoxyethyl)-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine (0.200 g, 0.848 mmol, 1.0 eq.), tert-butyl 6-(hydrazinecarbonyl)-2-azaspiro[3.3]heptane-2-carboxylate (0.227 g, 0.890 mmol, 1.05 eq.) from Example 32, Step 1, and AcOH (0.097 mL, 1.70 mmol, 2.0 eq.) in 2-propanol (10 mL) was heated to 80° C. for 2 h. The reaction mixture was concentrated under reduced pressure, the crude residue was diluted with DCM (10 mL) and washed with sat. aq. NaHCO3(10 mL). The organic phase was collected, and the aq. phase was extracted with DCM (3×10 mL). The combined organics were dried over MgSO4, solids were removed by vacuum filtration, and the filtrate was concentrated under reduced pressure. The crude residue was purified by column chromatography (SiO2, 0-10% MeOH/DCM) to afford tert-butyl 6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (0.385 g, 99%) as a light brown solid. LCMS (ESI): m/z 456.1 (M+H); Retention time: 2.30 min (50-100% ACN/H2O, method 3). Step 6: Synthesis of 8-chloro-5-cyclopropyl-1-(2-azaspiro[3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium 2,2,2-trifluoroacetate A solution of tert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4] triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (0.421 g, 0.923 mmol, 1.0 eq.) in DCM (10 mL) was stirred at 0° C. Trifluoroacetic acid (2.11 mL, 27.7 mmol, 30 eq.) was added dropwise. The reaction mixture was allowed to warm to RT and stirred for 60 min. The mixture was concentrated under reduced pressure, and the crude residue was azeotroped with toluene (4×20 mL). After 6 h under high vacuum, 8-chloro-5-cyclopropyl-1-(2-azaspiro [3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium 2,2,2-trifluoroacetate (0.539 g, 100%) was isolated as a brown solid. LCMS (ESI): m/z 356.1 (M+H); Retention time: 1.41 min (50-100% ACN/H2O, method 3). Step 7: Synthesis of 8-chloro-5-cyclopropyl-1-(2-(4-methylpyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[/][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 217) A mixture of 8-chloro-5-cyclopropyl-1-(2-azaspiro[3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium 2,2,2-trifluoroacetate (0.045 g, 0.0771 mmol, 1.0 eq), 2-chloro-4-methylpyridine (0.020 g, 0.157 mmol, 2.0 eq.), NaOtBu (0.0371 g, 0.386 mmol, 5.0 eq.), and RuPhos Pd G3 (0.007 g, 0.0077 mmol, 0.1 eq.) was stirred in 1,4-dioxane (1.0 mL) under a N2atmosphere for 2.5 h at 155° C. The reaction mixture was diluted with EtOAc (3 mL) and filtered through a pad of celite. The filter cake was washed with EtOAc (2×2 mL) and the combined filtrates were concentrated under reduced pressure. The crude residue was purified by reverse phase chromatography (5-85% ACN/H2O, 20 min method). 8-chloro-5-cyclopropyl-1-(2-(4-methylpyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (0.012 g, >98% pure, 34.1%) was isolated as a white solid after lyophilization of pure fractions. LCMS (ESI): m/z 447.4 (M+H); Retention time: 4.82 min (5-95% ACN/H2O, method 5). Compound Nos. 218 to 225, 227 to 229, 232 to 235, 247 to 250, and 270 to 278 Compound Nos. 218 to 225, 227 to 229, 232 to 235, 247 to 250, and 270 to 278 were prepared according to the methods set forth in Example 30 using appropriately substituted intermediates. Analytical data (LCMS) is also presented in Table 21. Compound Nos. 279 to 1072 are prepared according to the procedure of Example 30 using appropriately substituted intermediates. TABLE 21Compound Nos. 218 to 225, 227 to 229,232 to 235, 247 to 250, and 270 to 278CompoundSynthesisNo.Analytical DataMethod218LCMS (ESI): m/z 447.4 (M + H);Examples 30Retention time: 4.67 min (5-95%ACN/H2O, method 5); 98%219LCMS (ESI): m/z 434.3 (M + H);Examples 30Retention time: 6.37 min (5-95%ACN/H2O, method 5); 98.5%220LCMS (ESI): m/z 466.3 (M + H);Examples 30Retention time: 7.97 min (5-95%ACN/H2O, method 5); 98%221LCMS (ESI): m/z 448.4 (M + H);Examples 30Retention time: 6.65 min (5-95%ACN/H2O, method 5); 98.5%222LCMS (ESI): m/z 448.4 (M + H);Examples 30Retention time: 6.65 min (5-95%ACN/H2O, method 5); 98%223LCMS (ESI): m/z 451.3 (M + H);Examples 30Retention time: 6.10 min (5-95%ACN/H2O, method 5); 98%224LCMS (ESI): m/z 452.3 (M + H);Examples 30Retention time: 7.43 min (5-95%ACN/H2O, method 5); 98%225LCMS (ESI): m/z 434.3 (M + H);Examples 30Retention time: 7.43 min (5-95%ACN/H2O, method 5); 98%226LCMS (ESI): m/z 447.3 (M + H);Examples 30Retention time: 4.78 min (5-95%ACN/H2O, method 5); 98%228LCMS (ESI): m/z 458.3 (M + H);Examples 30Retention time: 9.84 min (5-95%ACN/H2O, method 5); 98%229LCMS (ESI): m/z 465.3 (M + H);Examples 30Retention time: 6.52 min (5-95%ACN/H2O, method 5); 98%232LCMS (ESI): m/z 458.0 (M + H);Examples 30Retention time: 7.49 min (5-95%ACN/H2O, method 5); 98%233LCMS (ESI): m/z 434.0 (M + H);Examples X-Retention time: 3.69 min (5-95%32ACN/H2O, method 5); 98%234LCMS (ESI): m/z 447.9 (M + H);Examples 30Retention time: 6.16 min (5-95%ACN/H2O, method 5); 98%237LCMS (ESI): m/z 465.2 (M + H);Examples 30Retention time: 5.78 min (5-95%ACN/H2O, method 5); 98%247LCMS (ESI): m/z 452.1 (M + H);Examples 30Retention time: 5.03 min (5-95%ACN/H2O, method 5); 98%248LCMS (ESI): m/z 459.2 (M + H);Examples 30Retention time: 8.17 min (5-95%ACN/H2O, method 5); 98%249LCMS (ESI): m/z 481.2 (M + H);Examples 30Retention time: 5.07 min (5-95%ACN/H2O, method 5); 98%250LCMS (ESI): m/z 487.2 (M + H);Examples 30Retention time: 7.38 min (5-95%ACN/H2O, method 5); 98%270LCMS (ESI): m/z 448.2 (M + H);Examples 30Retention time: 4.01 min (5-95%ACN/H2O, method 5); 98%271LCMS (ESI): m/z 519.2 (M + H);Examples 30Retention time: 11.62 min (5-95%ACN/H2O, method 5); 98%272LCMS (ESI): m/z 517.2 (M + H);Examples 30Retention time: 9.23 min (5-95%ACN/H2O, method 5); 98%273LCMS (ESI): m/z 465.2 (M + H);Examples 30Retention time: 6.27 min (5-95%ACN/H2O, method 5); 96%274LCMS (ESI): m/z 477.2 (M + H);Examples 30Retention time: 5.50 min (5-95%ACN/H2O, method 5); 97.7%275LCMS (ESI): m/z 461.4 (M + H);Examples 30Retention time: 5.32 min (5-95%ACN/H2O, method 5); 97.6%276LCMS (ESI): m/z 501.3 (M + H);Examples 30Retention time: 12.95 min (5-95%ACN/H2O, method 5); 97.9%277LCMS (ESI): m/z 469.2 (M + H);Examples 30Retention time: 10.73 min (5-95%ACN/H2O, method 5); 97.1%278LCMS (ESI): m/z 519.3 (M + H);Examples 30Retention time: 13.60 min (5-95%ACN/H2O, method 5); 98.3% Example 31 A mixture of 8-chloro-5-cyclopropyl-1-(2-azaspiro[3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium 2,2,2-trifluoroacetate (0.060 g, 0.103 mmol, 1.0 eq) from Example X-32, Step 6, DIPEA (0.090 mL, 0.515 mmol, 5 eq.), and cyclopropanecarbonyl chloride (0.014 mL, 0.154 mmol, 1.5 eq.) in DCM (1.0 mL) was stirred for 60 h at RT. The reaction mixture was concentrated under reduced pressure and the crude residue was purified by reverse phase chromatography (5-85% ACN/H2O, 20 min method). (6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-yl)(cyclopropyl)methanone (0.010 g, 98% pure, 22.4% yield) was isolated as a white solid. LCMS (ESI): m/z 424.3 (M+H); Retention time: 6.34 min (5-95% ACN/H2O, method 5). Example 32 Step 1: 6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carbonitrile A mixture of 8-chloro-5-cyclopropyl-1-(2-azaspiro[3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium 2,2,2-trifluoroacetate (0.438 g, 0.750 mmol, 1.0 eq) from Example X-32, Step 7, DIPEA (0.654 mL, 3.76 mmol, 5 eq.), and cyanogen bromide (0.159 g, 1.50 mmol, 2 eq.) was stirred in DCM (2.5 mL) for 60 min at 0° C. The reaction mixture was diluted with EtOAc (10 mL) and washed with sat. aq. NaHCO3(2×10 mL), brine (1×10 mL) and dried over MgSO4. Solids were removed by vacuum filtration, and the filtrate was concentrated under reduced pressure. The crude residue was purified by column chromatography (SiO2, 0-10% MeOH/DCM) to afford 6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carbonitrile (0.385 g, 99%) as a light brown solid. LCMS (ESI): m/z 381.1 (M+H); Retention time: 1.66 min (50-100% ACN/H2O, method 3). Step 2: (E)-6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4] diazepin-1-yl)-N-hydroxy-2-azaspiro[3.3]heptane-2-carboximidamide A suspension of 6-(7-chloro-3,4-dihydro-2H-benzo[b][1,4]oxazine-4-carbonyl)-2,6-diazaspiro[3.3]heptane-2-carbonitrile (100 mg, 0.263 mmol, 1.0 eq.), TEA (0.0385 mL, 0.276 mmol, 1.05 eq.) and hydroxylamine hydrochloride (19.2 mg, 0.276 mmol, 1.05 eq.) in EtOH (1 mL) was heated at 80° C. for 1 hour. The mixture was concentrated under reduced pressure to afford (E)-6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4] diazepin-1-yl)-N-hydroxy-2-azaspiro[3.3]heptane-2-carboximidamide (0.109 g, 100%) as a light brown solid. The crude product was carried forward without further purification. LCMS (ESI): m/z 414.1 (M+H); Retention time: 1.46 min (50-100% ACN/H2O, method 3). Step 3: 3-(6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-yl)-5-methyl-1,2,4-oxadiazole (Compound No. 268) A mixture of (E)-6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-N-hydroxy-2-azaspiro[3.3]heptane-2-carboximidamide (0.055 g, 0.133 mmol, 1 eq.) and acetic anhydride (0.027 mL, 0.28 mmol, 1.05 eq.) in pyridine (1 mL) was stirred for 1 h at 80° C. The reaction mixture was concentrated under reduced pressure and the crude residue was purified by reverse phase chromatography (5-85% ACN/H2O, 20 min method). 3-(6-(8-chloro-5-cyclopropyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-yl)-5-methyl-1,2,4-oxadiazole (0.011 g, 95.2% pure, 18% yield) was isolated as a white solid. LCMS (ESI): m/z 438.3 (M+H); Retention time: 7.11 min (5-95% ACN/H2O, method 5). Compound No. 269 Compound No. 269 was prepared according to the methods set forth in Example 32 using appropriately substituted intermediates. Analytical data (LCMS) LCMS (ESI): m/z 464.4 (M+H); Retention time: 8.72 min (5-95% ACN/H2O, method 5); 97.0%. Example 33 Step 1: tert-butyl (4-chloro-2-(((cyanomethyl)(methyl)amino)methyl)phenyl)carbamate tert-butyl (4-chloro-2-formylphenyl)carbamate (3.00 g, 11.7 mmol, 1 eq.), 2-(methylamino)-acetonitrile hydrochloride (3.74 g, 35.1 mmol, 3 eq.), and DIPEA (6.16 g, 35.1 mmol, 3 eq.) were mixed in DCE (76 mL) and stirred for 15 min at 25° C. Acetic acid (2.1 mL, 35.1 mmol, 3 eq.)) and MgSO4(4.32 g, 35.1 mmol, 3 eq.) were added, and the suspension was stirred for 2 h at 60° C. After the reaction mixture was cooled to 25° C., sodium triacetoxyborohydride (6.21 g, 29.3 mmol, 2.5 eq.) was added in two portions, and the reaction was stirred for 18 h at 25° C. The reaction was quenched with methanol and sodium bicarbonate (sat. aq.) to pH 8, and the aqueous layer was extracted with ethyl acetate twice. The organic layers were combined, dried over MgSO4, and evaporated under reduced pressure. The residue was purified by column chromatography (SiO2, 0-30% EtOAc/Hex) to give tert-butyl (4-chloro-2-(((cyanomethyl)(methyl)amino)methyl)phenyl)carbamate (1.33 g, 37% yield) as a white solid. LCMS (ESI): m/z 310 (M+H); Retention time: 3.36 min (50-100% ACN/H2O, method 3). Step 2: 7-chloro-4-methyl-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine To a solution of tert-butyl (4-chloro-2-(((cyanomethyl)(methyl) amino)methyl)phenyl)carbamate (0.140 g, 0.45 mmol, 1 eq.) in dioxane (1.0 mL) was added hydrogen chloride solution in 1,4-dioxane (4.0 M, 2.25 mL, 9.0 mmol, 9 eq.) dropwise, and the reaction was stirred at 25° C. for 2 h. 2-Propanol (2.0 mL) was added. The solution was stirred for 18 h at 65° C., resulting in a light brown suspension. Na2CO3(2.0 M, aq.) was added to the suspension, and the final pH was 11. The mixture was extracted with EtOAc three times, and the organic layers were combined, dried over MgSO4, and evaporated under reduced pressure. 7-chloro-4-methyl-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine (125 mg, crude) was isolated as a brown solid, and used in the next step without further purification. Step 3: tert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]-diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate 7-chloro-4-methyl-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine (1.36 g, 6.5 mmol) and tert-butyl 6-(hydrazinecarbonyl)-2-azaspiro[3.3]heptane-2-carboxylate (1.65 mg, 6.5 mmol) were mixed in isopropanol (35 mL) and acetic acid (0.38 mL, 6.5 mmol), and the solution was stirred at 80° C. for 2 h. The reaction solution was diluted with EtOAc, washed by Na2CO3(twice) and brine, dried over MgSO4, and evaporated under reduced pressure. The brown residue was purified by column chromatography (SiO2, 0-15% MeOH/DCM) to afford tert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (1.80 g, 65% yield) as white foam solid. LCMS (ESI): m/z 430 (M+H); Retention time: 1.42 min (50-100% ACN/H2O, method 3). Step 4: 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]-triazolo[4,3-a][1,4]diazepine TFA salt To a solution of tert-butyl 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo-[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (1.53 g, 3.56 mmol) in DCM (21 mL) was added trifluoroacetic acid (5.5 mL, 71 mmol) at 0° C., and the solution was stirred at 25° C. for 2 h. The reaction mixture was evaporated under reduced pressure, and azeotropic evaporation was done with acetonitrile (twice) and toluene (once). 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]-diazepine TFA salt (4.03 g) was obtained as brown oil, and used without further treatment. LCMS (ESI): m/z 330 (M+H); Retention time: 1.40 min (50-100% ACN/H2O, method 3). Step 5: 8-chloro-5-methyl-1-(2-(4-methylpyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 241) 8-Chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4] triazolo[4,3-a][1,4]diazepine TFA salt (50 mg, 0.090 mmol, crude from Step 4), 2-bromo-4-methylpyridine (31 mg, 0.18 mmol), sodium tert-butoxide (43 mg, 0.45 mmol), RuPhos (4.2 mg, 9.0 umol) and RuPhos-Palladacycle-G3 (7.5 mg, 9.0 umol) were mixed in 1,4-dioxane (1.0 mL) and N-methylpyrrolidine (0.1 mL), and the mixture was stirred at 120° C. for 5 h. The reaction mixture was diluted with EtOAc, washed by NaHCO3(sat. aq.) and brine, dried over MgSO4, filtered and evaporated under reduced pressure. The residue was purified by RP-HPLC (5-85% ACN/H2O) to give 8-chloro-5-methyl-1-(2-(4-methylpyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (10.5 mg, 28% yield) as white solid. LCMS (ESI): m/z 421 (M+H); Retention time: 2.12 min (10-100% ACN/H2O, method 4). Compound Nos. 231, 237 to 240, 242 to 243, 251 to 252, and 256 to 258 Compound Nos. 231, 237 to 240, 242 to 243, 251 to 252, and 256 to 258 were prepared according to the methods set forth in Example 33 using appropriately substituted intermediates. Analytical data (LCMS) is also presented in Table 22. TABLE 22Compound Nos. 231, 237 to 240, 242to 243, 251 to 252, and 256 to 258CompoundSynthesisNo.Analytical DataMethod231LCMS (ESI): m/z 461.1 (M + H);Examples 33Retention time: 4.0 min (5-95%ACN/H2O, method 5)237LCMS (ESI): m/z 422 (M + H);Examples 33Retention time: 2.75 min (10-100%ACN/H2O, method 4)238LCMS (ESI): m/z 426 (M + H);Examples 33Retention time: 2.06 min (10-100%ACN/H2O, method 4)239LCMS (ESI): m/z 422 (M + H);Examples 33Retention time: 2.52 min (10-100%ACN/H2O, method 4)240LCMS (ESI): m/z 440 (M + H);Examples 33Retention time: 3.18 min (10-100%ACN/H2O, method 4)242LCMS (ESI): m/z 439 (M + H);Examples 33Retention time: 2.53 min (10-100%ACN/H2O, method 4)243LCMS (ESI): m/z 408 (M + H);Examples 33Retention time: 1.70 min (10-100%ACN/H2O, method 4)251LCMS (ESI): m/z 422 (M + H);Examples 33Retention time: 1.75 min (10-100%ACN/H2O, method 4252LCMS (ESI): m/z 439 (M + H);Examples 33Retention time: 2.49 min (10-100%ACN/H2O, method 4)256LCMS (ESI): m/z 447.1 (M + H);Examples 33Retention time: 6.8 min (5-95%ACN/H2O, method 5)257LCMS (ESI): m/z 461.2 (M + H);Examples 33Retention time: 1.8 min (5-95%ACN/H2O, method 5)258LCMS (ESI): m/z 461.2 (M + H);Examples 33Retention time: 1.0 min (5-95%ACN/H2O, method 5) Example 34 Step 1: 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carbonitrile To a solution of 8-chloro-5-methyl-1-(2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine TFA salt (100 mg, 0.18 mmol) and DIPEA (0.16 mL) in DCM (0.5 mL) was added cyanogen bromide (38 mg) at 0° C., and the reaction was stirred at 0° C. for 1 h. The reaction mixture was diluted with EtOAc, washed by NaHCO3(sat. aq.) and brine. The organic layer was dried over MgSO4and evaporated under reduced pressure. The residue was purified by column chromatography (SiO2, 0-10% MeOH/DCM) to give 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]-heptane-2-carbonitrile (36 mg, 33% yield) as yellow film. LCMS (ESI): m/z 355 (M+H); Retention time: 2.31 min (10-100% ACN/H2O, method 4). Step 2: 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-N-hydroxy-2-azaspiro[3.3]heptane-2-carboximidamide To a suspension of 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo-[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carbonitrile (147 mg, 0.41 mmol) and hydroxyamine hydrochloride (30 mg, 0.43 mmol) in ethanol (1.5 mL) was added triethylamine (60 uL, 0.43 mmol), and the reaction was stirred at 80° C. for 1 h, resulting in a yellow solution. Ethanol was evaporated under reduced pressure, and the crude 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-N-hydroxy-2-azaspiro[3.3]heptane-2-carboximidamide was diluted with pyridine (1.2 mL) and used without further treatment. LCMS (ESI): m/z 388 (M+H); Retention time: 1.44 min (50-100% ACN/H2O, method 3). Step 3: 3-(6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-yl)-5-methyl-1,2,4-oxadiazole (Compound No. 266) To a solution of 6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-N-hydroxy-2-azaspiro[3.3]heptane-2-carboximidamide (46 mg, 0.12 mmol) in pyridine (0.4 mL) was added acetic anhydride (13 uL, 0.14 mmol) at 0° C., and the reaction was stirred at 0° C. for 30 min, then at 80° C. for 1 h. The reaction solution was diluted with EtOAc and washed by NaHCO3(sat. aq.) and brine. The organic layer was dried over MgSO4and evaporated under reduced pressure. The residue was purified by RP-HPLC (5-85% ACN/H2O) to give 3-(6-(8-chloro-5-methyl-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-yl)-5-methyl-1,2,4-oxadiazole (4.8 mg, 9% yield) as white solid. LCMS (ESI): m/z 412; Retention time: 2.67 min (10-100% ACN/H2O, method 4). Compound No. 267 Compound No. 267 was prepared according to the methods set forth in Example 34 using appropriately substituted intermediates. Analytical data (LCMS): LCMS (ESI): m/z 438 (M+H); Retention time: 2.93 min (10-100% ACN/H2O, method 4). Example 35 8-chloro-5-methyl-1-(2-(pyrimidin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 236) To a solution of 7-chloro-4-methyl-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine (20 mg, 0.095 mmol) and 2-(5-fluoropyrimidin-2-yl)-2-azaspiro[3.3]heptane-6-carbohydrazide (25 mg, 0.11 mmol) in 2-propanol (1 mL) was added acetic acid (6 uL), and the reaction was stirred at 80° C. for 4 h. The reaction solution was diluted with EtOAc and washed by NaHCO3(sat. aq.) and brine. The organic layer was dried over MgSO4and evaporated under reduced pressure. The residue was purified by RP-HPLC (5-85% ACN/H2O) to give 8-chloro-5-methyl-1-(2-(5-fluoropyrimidin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (20 mg, 70% yield) as white solid. LCMS (ESI): m/z 426 (M+H); Retention time: 2.82 min (10-100% ACN/H2O, method 4). Example 36 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5-(2,2,2-trifluoroethyl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 259) To a mixture of 2,2,2-trifluoroethyl trifluoromethanesulfonate (56 mg, 0.244 mg, 2 eq.) and 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine from Example 10, step 3 (50 mg, 0.122 mmol, 1.0 eq.) in anhydrous MeCN (0.75 mL) was added K2CO3(35 mg, 0.13 mmol, 2.1 eq.) and the resultant mixture was stirred at room temperature for 0.5 hours and then at 75° C. for 3 hours. The mixture was cooled and partitioned between sodium bicarbonate solution and DCM. The organic fraction was collected, concentrated to a residue, and purified by reverse phase HPLC, eluting with a gradient of 5-85% CH3CN in water, to give the title compound as a white solid (23.2 mg, 38% yield). LCMS (ESI): m/z 493.1 (M+H), Retention time: 9.0 min (5-95% ACN/H2O with 0.1% formic acid, method 5). Example 37 8-chloro-N-cyclopropyl-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxamide (Compound No. 246) To a solution of 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine from Example 10, step 3 (35 mg, 0.085 mmol, 1.0 eq.) in 0.5 ml anhydrous THE and 30 μL DMF was added isocyanatocyclopropane (9 μL, 0.128 mmol, 1.5 eq.) and the mixture was heated at 45° C. in a sealed tube for 18 hours. The solution was diluted with methanol, concentrated under vacuum, and purified by reverse phase HPLC, eluting with a gradient of 5-85% CH3CN in water, to give 18.7 mg of white solid (44.5% yield). LCMS (ESI): m/z 494.1 (M+H), Retention time: 6.2 min (5-95% ACN/H2O with 0.1% formic acid, method 5). Example 38 Step 1: Synthesis of tert-butyl 6-(8-chloro-5-(dimethylcarbamoyl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate To a cooled 0° C. solution of 4-nitrophenyl chloroformate (29 mg, 0.143 mmol, 1.1 eq.) and triethylamine (36 μL, 0.26 mmol, 2 eq.) in anhydrous DCM (300 μL) was slowly added a solution of tert-butyl 6-(8-chloro-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate from Example 12, step 6 (54 mg, 0.13 mmol, 1.0 eq.) in DCM (1 mL). The mixture was stirred for three hours at 0° C., then allowed to warm to room temperature and stirred for 1 hour. A solution of 2M dimethylamine in THE (500 μL, 1.0 mmol, 7.7 eq) and the mixture was stirred at 45° C. for 72 hours. The reaction mixture was diluted with DCM and successively washed with water, 1M citric acid soln., and brine. The organic fraction was dried over sodium sulfate and concentrated to 52 mg of clear residue, which was used in the next step without further purification (82% yield). MS (ESI): m/z 487.2 (M+H) Step 2: 6-(8-chloro-5-(dimethylcarbamoyl)-5,6-dihydro-4H-benzo[/][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-ium 2,2,2-trifluoroacetate To a solution of tert-butyl 6-(8-chloro-5-(dimethylcarbamoyl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (52 mg, 0.10 mmol, 1 eq.) in 3 mL of DCM was added 1 mL of TFA, and the solution was stirred at room temperature for 2 hours. The solution was concentrated under vacuum, and the residue suspended in toluene and concentrated under vacuum to give the title compound as 55 mg of a clear oil (100% yield). MS (ESI): m/z 387.1 (M+H) Step 3: 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-N,N-dimethyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine-5(6H)-carboxamide (Compound No. 230) A vial was charged with 6-(8-chloro-5-(dimethylcarbamoyl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptan-2-ium 2,2,2-trifluoroacetate (55 mg, 0.11 mmol, 1.0 eq.), 2-bromo-5-fluoropyridine (39 mg, 0.22 mmol, 2 eq.), sodium tert-butoxide (53 mg, 0.55 mmol, 5 eq.), and 1 ml of a 9:1 mixture of 1,4-dioxane:NMP. The resultant suspension was degassed, placed under a nitrogen atmosphere, and heated at 85° C. for 5 hours. The mixture was cooled, filtered through celite (washing with DCM and EtOAc) and concentrated to a brown oil. The crude product was dissolved in methanol and purified by reverse phase chromatography, eluting from a C18 column with a gradient of 5-85% CH3CN in water to give the title compound as 9.7 mg of a white solid (18.3% yield). LCMS (ESI): m/z 482.5 (M+H), Retention time: 6.9 min (5-95% ACN/H2O with 0.1% formic acid, method 5). Example 39 Step 1: Synthesis of tert-butyl (4-chloro-2-(((1-methylcyclopropyl)amino)methyl)phenyl)-carbamate To a stirred solution of tert-butyl (4-chloro-2-formylphenyl)carbamate (0.5 g, 1.955 mmol, 1.0 eq) in MeOH (10 mL) was added 1-methylcyclopropan-1-amine (0.2781 g, 3.910 mmol, 2.0 eq) and the mixture was stirred at 43° C. for 16 h. THF (6.0 mL) was added to the reaction mixture, followed by the addition of sodium triacetoxyborohydride (2.08 g, 9.80 mmol, 5.0 eq) and acetic acid (0.35 g, 5.89 mmol, 3.0 eq), and stirred at RT for 20 h. The reaction mixture was diluted with EtOAc (20 mL) and sat. aq. NaHCO3(20 mL). The organic phase was collected, and the aqueous phase was extracted with EtOAc (2×10 mL). Combined organics were washed with brine, dried over MgSO4, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, 0-20% EtOAc/Hex) to afford tert-butyl (4-chloro-2-(((1-methylcyclopropyl)amino)methyl)phenyl)-carbamate (0.520 g, 85%) as a white solid. LCMS (ESI): m/z=311.4 (M+H), retention time; 1.40 min (50-100% ACN/H2O, method 3). Step 2: Synthesis of tert-butyl (4-chloro-2-(((cyanomethyl)(1-methylcyclopropyl)amino)methyl)-phenyl)carbamate To a mixture of tert-butyl (4-chloro-2-(((1-methylcyclopropyl)amino)methyl)-phenyl)carbamate (0.510 g, 1.64 mmol, 1.0 eq), K2CO3(0.453 g, 3.28 mmol, 2.0 eq), and KI (0.164 g, 0.988 mmol, 0.6 eq) in ACN (2.0 mL) and 1,4-dioxane (3.0 mL) was added 2-chloroacetonitrile (0.248 g, 3.28 mmol, 2.0 eq). The mixture was stirred at 80° C. for 22 h under an atmosphere of N2. The reaction mixture was then cooled to RT, diluted with (1:1) mixture of EtOAc and sat. aq. NaHCO3(20 mL). The organic phase was collected, and the aqueous phase was extracted with EtOAc (2×20 mL). The combined organics were washed with sat. aq Na2S2O3solution, brine, dried over MgSO4, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, 0-20% EtOAc/Hex) to afford tert-butyl (4-chloro-2-(((cyanomethyl)(1-methylcyclopropyl)amino)methyl)-phenyl)carbamate (0.50 g, 87%) as a white solid. LCMS (ESI): m/z=350.1 (M+1), retention time; 4.15 min (50-100% ACN/H2O, method 3). Step 3: Synthesis of 7-chloro-4-(1-methylcyclopropyl)-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine A solution of acetyl chloride (3.17 g, 40.4 mmol, 20.0 eq) in isopropanol (15 mL) was stirred for 20 min at RT prior to slow addition to a solution of tert-butyl (4-chloro-2-(((cyanomethyl)(1-methylcyclopropyl)amino)-methyl)-phenyl)carbamate (0.705 g, 2.02 mmol, 1.0 eq) in isopropanol (15 mL) and stirred for 60 h at 50° C. The reaction mixture was concentrated under reduced pressure and the crude was mixed vigorously with a (1:1) mixture of EtOAc/satd aq NaHCO3(60 mL). The organic phase was collected, and the aqueous phase was extracted with EtOAc (3×15 mL). Combined organics were washed with brine, dried over MgSO4, filtered, and concentrated under reduced pressure. The crude residue was purified by column chromatography (SiO2, 0-100% EtOAc/Hex, 0-100% DCM/EtOAc, 0-12% MeOH/DCM) to afford 7-chloro-4-(1-methylcyclopropyl)-4,5-dihydro-3H-benzo[e][1,4]diazepin-2-amine (0.433 g, 86% yield) as a light brown solid. LCMS (ESI): m/z 250.1 (M+H); Retention time: 1.39 min (50-100% ACN/H2O, method 3). Step 4: Synthesis of tert-butyl 6-(hydrazinecarbonyl)-2-azaspiro[3.3]heptane-2-carboxylate To a stirred solution of 2-(tert-butoxycarbonyl)-2-azaspiro[3.3]heptane-6-carboxylic acid (0.26 g, 1 mmol, 1 eq.) in THE (5 mL) was added 1-1′-Carbonyldiimidazole (0.19 g, 1.2 mmol, 1.2 eq.) and the mixture was stirred at RT overnight. The resulting mixture was added to a solution of hydrazine monohydrate (0.07 mL, 1.4 mmol, 1.4 eq.) in THE (10 mL) and stirred at RT overnight. The mixture was diluted with brine and extracted with ethyl acetate. The organic phase was separated, washed with brine, dried (MgSO4), filtered and concentrated in vacuo to afford tert-butyl 6-(hydrazinecarbonyl)-2-azaspiro[3.3]heptane-2-carboxylate as a white solid (0.29 g, 100% yield). This material was used without further purification. (TLC rf: 0.4; 10% MeOH in DCM). Step 5: Synthesis of tert-butyl 6-(8-chloro-5-(1-methylcyclopropyl)-5,6-dihydro-4H-benzo[f]-[1,2,4]triazolo[4,3-a][1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate A mixture of 7-chloro-4-(1-methylcyclopropyl)-4,5-dihydro-3H-benzo[e][1,4]-diazepin-2-amine (0.058 g, 0.232 mmol, 1.0 eq), 2-(5-fluoro-4-methylpyridin-2-yl)-2-azaspiro-[3.3]heptane-6-carbohydrazide (0.089 g, 0.348 mmol, 1.5 eq), and AcOH (2 drops) in 2-propanol (1 mL) was stirred for 20 h at 80° C. The reaction mixture was then concentrated under reduced pressure, and the crude was diluted with DCM (5 mL) and satd aq NaHCO3(5 mL). Organic phase was collected, and the aqueous phase was extracted with DCM (3×5 mL). Combined organics were washed with brine, dried over MgSO4, filtered, and concentrated under reduced pressure. The crude residue was purified by column chromatography (SiO2, 0-10% MeOH/DCM) to afford tert-butyl 6-(8-chloro-5-(1-methylcyclopropyl)-5,6-dihydro-4H-benzo[f]-[1,2,4]triazolo[4,3-a]-[1,4]diazepin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (0.082 g, 74%) as a light brown solid. LCMS (ESI): m/z 470.2 (M+H); Retention time: 2.48 min (50-100% ACN/H2O, method 3). Step 6: Synthesis of 8-chloro-5-(1-methylcyclopropyl)-1-(2-azaspiro[3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium-6-ylium 2,2,2-trifluoroacetate To a solution of tert-butyl 8-chloro-5-(1-methylcyclopropyl)-1-(2-azaspiro[3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium 2,2,2-trifluoroacetate (0.279 g, 0.594 mmol, 1.0 eq) in DCM (20 mL) at 0° C., TFA (6 mL) was added dropwise. The reaction mixture was allowed to warm to RT and stirred 60 min. The mixture was concentrated under reduced pressure, and the crude residue was azeotroped with toluene (4×20 mL). After 6 h under high vacuum, 8-chloro-5-(1-methylcyclopropyl)-1-(2-azaspiro[3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium-6-ylium 2,2,2-trifluoroacetate (0.353 g, 100%) was isolated as a brown solid. LCMS (ESI): m/z 370.1 (M+H); Retention time: 1.41 min (50-100% ACN/H2O, method 3). Step 7: Synthesis of 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5-(1-methylcyclopropyl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (Compound No. 255) A mixture of 8-chloro-5-(1-methylcyclopropyl)-1-(2-azaspiro[3.3]heptan-2-ium-6-yl)-5,6-dihydro-4H-benzo[1,2,4]triazolo[4,3-a][1,4]diazepin-5-ium-6-ylium 2,2,2-trifluoroacetate (0.025 g, 0.043 mmol, 1.0 eq), 2-chloro-5-fluoropyridine (0.012 g, 0.087 mmol, 2.0 eq), NaOtBu (0.021 g, 0.218 mmol, 5.0 eq.), and RuPhos Pd G3 (0.004 g, 0.004 mmol, 0.1 eq) was stirred in 1,4-dioxane (1.0 mL) and few drops of NMP under a N2atmosphere for 16 h at 90° C. The reaction mixture was diluted with EtOAc (3 mL) and filtered through a pad of celite. The filter cake was washed with EtOAc (2×2 mL) and the combined filtrates were concentrated under reduced pressure. The crude residue was purified by reverse phase chromatography (5-85% ACN/H2O, 20 min method). 8-chloro-1-(2-(5-fluoropyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)-5-(1-methylcyclopropyl)-5,6-dihydro-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine (0.0085 g, >97% pure, 41.1%) was isolated as a white solid after lyophilization of pure fractions. LCMS (ESI): m/z 465.2 (M+H); Retention time: 7.35 min (5-95% ACN/H2O, method 5). Compound Nos. 244 to 245, 253 to 254, and 260 to 265 Compound Nos. 244 to 245, 253 to 254, and 260 to 265 were prepared according to the methods set forth in Example 38 using appropriately substituted intermediates. Analytical data (LCMS) is also presented in Table 23. TABLE 23Compound Nos. 244 to 245, 253 to 254, and 260 to 265CompoundSynthesisNo.Analytical DataMethod244LCMS (ESI): m/z 462.0 (M + H);Example 39Retention time: 6.75 min (10-100%ACN/H2O, method 5)245LCMS (ESI): m/z 479.0 (M + H);Example 39Retention time: 6.03 min (10-100%ACN/H2O, method 5)253LCMS (ESI): m/z 462.3 (M + H);Example 39Retention time: 7.95 min (10-100%ACN/H2O, method 5)254LCMS (ESI): m/z 448.2 (M + H);Example 39Retention time: 4.10 min (10-100%ACN/H2O, method 5)260LCMS (ESI): m/z 479.1 (M + H);Example 39Retention time: 6.27 min (10-100%ACN/H2O, method 5)261LCMS (ESI): m/z 466.3 (M + H);Example 39Retention time: 8.48 min (10-100%ACN/H2O, method 5)262LCMS (ESI): m/z 472.2 (M + H);Example 39Retention time: 10.75 min (10-100%ACN/H2O, method 5)263LCMS (ESI): m/z 473.1 (M + H);Example 39Retention time: 9.41 min (10-100%ACN/H2O, method 5)264LCMS (ESI): m/z 448.1 (M + H);Example 39Retention time: 6.77 min (10-100%ACN/H2O, method 5)265LCMS (ESI): m/z 461.2 (M + H);Example 39Retention time: 5.10 min (10-100%ACN/H2O, method 5) In Vitro Activity Example B1 Vasopressin V1A Receptor Antagonist Assay The purpose of this assay was to determine the inhibitory effect of synthesized compounds on the Vasopressin V1a receptor. The assay as performed in Chinese Hamster Ovary (CHO) cells expressing the human Arginine Vasopressin Receptor 1a (AVPR1a). Arginine Vasopressin (AVP) evokes an increase in intracellular calcium in CHO-AVPR1a cells which is measured in a fluorescence assay on the FLIPRTETRAusing calcium sensitive dyes. Test compounds were assessed for their ability to affect the magnitude of the response to AVP, with antagonists showing a concentration-dependent reduction in the AVP-mediated fluorescence. Compounds were tested in duplicate in a 10-point, 1:3 dilution series starting at a nominal concentration of 3 μM in the assay. CHO-AVPR1a cells were maintained in routine culture in T175 Flasks at 37° C., 5% CO2. The growth medium consists of Ham's F12 media supplemented with 10% v/v fetal bovine serum, 1× non-essential amino acids, and 0.4 mg/ml Geneticin G418. On day one, cells were harvested from T175 flasks when they are 80-90% confluent by first washing the cell monolayer with PBS and then dissociated using trypsin 0.05%/EDTA (3 mL for a T175 Flask). The flasks were incubated at room temperature until the cells detached. To the cell suspension, 10 ml of growth media was added and the cell density determined using the Vi-Cell automated cell counter. The cells were spun at 1000 rpm for 3 minutes, then the supernatant was carefully removed and discarded. The cell pellet was re-suspended at 6.0e5cells/ml in growth media. 25 μL of cells in growth media was dispensed into each well (15,000 cells per well) of a poly-D-lysine coated black, clear bottomed, 384-well plate. The plates were incubated at 37° C., 5% CO2overnight. At the start of each assay day, the potency of AVP was assessed and an EC80concentration determined for subsequent compound profiling. Assays were performed using a two-step addition protocol on the FLIPRTETRA; first addition of 5 μl of control or test compound at 10× final in assay buffer with 15 min incubation at 37° C., 5% CO2followed by 10 μl of AVP at 6× final concentration in assay buffer. Changes in fluorescence were monitored for 3 min after both additions on the FLIPRTETRAusing 470-495 nm excitation and 515-575 nm emission wavelengths. The assay buffer consisted of HBSS (+Ca/+Mg) supplemented with 20 mM HEPES, and for the preparation of the AVP agonist only, 0.1% w/v bovine serum albumin. The assay was initiated by the removal of growth media from the cells and replacement with 45 μl of Calcium-6 dye (Molecular Devices) prepared at 1× in assay buffer. Cells are loaded with dye for 60-90 min at 37° C., 5% CO2before initiation of the FLIPRTETRAprotocol. For the AVP potency determination, the first addition consisted of assay buffer containing 3% v/v DMSO and the second addition, a 10-point dilution series of AVP (1:3 dilutions from 1 μM) in assay buffer supplemented with 0.1% BSA. For compound profiling, test compounds were first serially diluted in DMSO (10-point curve, 1:3 dilutions) then diluted 33.3-fold in assay buffer prior to addition to the dye loaded cells on the FLIPRTETRA. At the end of the incubation period, 10 μl of AVP in assay buffer containing 0.1% BSA was added at the previously determined EC80concentration. In-plate controls for the assay include Ro5028442 and PF-184563 concentration-response curves as reference V1a antagonists and an AVP concentration-response curve to confirm the reproducibility of the EC80used for the compound challenge. MAX-MIN raw data is normalised to in-plate assay controls comprising DMSO matched solutions of 300 nM SR49059 (100% inhibition) and AVP EC80(0% inhibition). Selectivity profiling of certain example compounds was determined against Vasopressin V1b and V2 receptors. Example B2 Vasopressin V1B Receptor Antagonist Assay The purpose of the assay is to determine the inhibitory effect of synthesized compounds on the Vasopressin V1b receptor. The assay was performed in Chinese Hamster Ovary (CHO) cells expressing the human Arginine Vasopressin Receptor 1b (AVPR1b). Arginine Vasopressin (AVP) evokes an increase in intracellular calcium in CHO-AVPR1b cells which is measured in a fluorescence assay on the FLIPRTETRAusing calcium sensitive dyes. Test compounds were assessed for their ability to affect the magnitude of the response to AVP, with antagonists showing a concentration-dependent reduction in the AVP-mediated fluorescence. CHO-AVPR1b cells were maintained in routine culture in T175 Flasks at 37° C., 5% CO2. The growth medium consisted of Ham's F12 media supplemented with 10% v/v fetal bovine serum, 1× non-essential amino acids, and 0.4 mg/ml Geneticin G418. Cells were harvested from T175 flasks when they were 80-90% confluent by first washing the cell monolayer with PBS and then dissociated using trypsin 0.05%/EDTA (3 mL for a T175 Flask). The flasks were incubated at room temperature until the cells detached. To the cell suspension, 10 ml of growth media was added and the cell density determined using the Vi-Cell automated cell counter. The cells were spun at 1000 rpm for 3 minutes, then the supernatant was carefully removed and discarded. The cell pellet was re-suspended at 6.0e5cells/ml in growth media. 25 μL of cells in growth media was dispensed into each well (15,000 cells per well) of a poly-D-lysine coated black, clear bottomed, 384-well plate. The plates were incubated at 37° C., 5% CO2overnight. At the start of each assay day, the potency of AVP is assessed and an EC80concentration determined for subsequent compound profiling. Assays were performed using a two-step addition protocol on the FLIPRTETRA; first addition of 5 μl of control or test compound at 10× final in assay buffer with 15 min incubation at 37° C., 5% CO2followed by 10 μl of AVP at 6× final concentration in assay buffer. Changes in fluorescence were monitored for 3 min after both additions on the FLIPRTETRAusing 470-495 nm excitation and 515-575 nm emission wavelengths. The assay buffer consisted of HBSS (+Ca/+Mg) supplemented with 20 mM HEPES, and for the preparation of the AVP agonist only, 0.1% w/v bovine serum albumin. The assay was initiated by the removal of growth media from the cells and replacement with 45 μl of Calcium-6 dye (Molecular Devices) prepared at 1× in assay buffer. Cells were loaded with dye for 60-90 min at 37° C., 5% CO2before initiation of the FLIPRTETRAprotocol. For the AVP potency determination, the first addition consisted of assay buffer containing 3% v/v DMSO and the second addition a 10-point dilution series of AVP (1:3 dilutions from 1 μM) in assay buffer supplemented with 0.1% BSA. For compound profiling, test compounds were first serially diluted in DMSO (10-point curve, 1:3 dilutions) then diluted 33.3-fold in assay buffer prior to addition to the dye loaded cells on the FLIPRTETRA. At the end of the incubation period 10 μl of AVP in assay buffer containing 0.1% BSA was added at the previously determined EC80concentration. In-plate controls for the assay include a Nelivaptan concentration-response curve as the reference V1b antagonist and an AVP concentration-response curve to confirm the reproducibility of the EC80used for the compound challenge. MAX-MIN raw data is normalised to in-plate assay controls comprising DMSO matched solutions of 3 μM nelivaptan (100% inhibition) and AVP EC80(0% inhibition). Example B3 Vasopressin V2 Receptor Antagonist Assay The purpose of the assay was to determine the inhibitory effect of synthesized compounds on the Vasopressin receptor 2. The assay was performed in commercially available 1321N1 cells expressing the human Arginine Vasopressin Receptor V2 (AVPR2) (Perkin Elmer #ES-363-CF). Arginine Vasopressin (AVP) evokes an increase in intracellular cAMP in these cells which is measured in a TR-FRET assay using a Europium cAMP tracer and ULight labelled antibody reagents contained in a LANCE Ultra cAMP kit (Perkin Elmer #TRF0263). Increases in cAMP in the assay result in a reduction in TR-FRET as the cAMP produced by the stimulated cells competes with the Eu-cAMP tracer for binding sites on the ULight labelled antibody. Test compounds were assessed for their ability to affect the magnitude of the response to AVP, with antagonists showing a concentration-dependent decrease in the AVP-mediated reduction in TR-FRET signal. cAMPZen V2 assay ready cells were thawed at 37° C. and resuspended directly from frozen in 9 ml growth medium consisting of DMEM supplemented with 10% v/v fetal bovine serum, 1× non-essential amino acids, and 1 mM sodium pyruvate. Cells were spun at 1000 rpm for 3 minutes and the supernatant was carefully removed and discarded. The pellet was resuspended in 5 ml stimulation buffer and the cell density determined using the Vi-Cell automated cell counter. The cell suspension was diluted to a 0.2×106/ml suspension ready for plating. To all wells of a white 384-well Optiplate (Perkin Elmer #6007299) 5 μL of cells in stimulation buffer were dispensed (1,000 cells per well). Stimulation buffer consisted of HBSS (+Ca/+Mg) supplemented with 5 mM HEPES, 0.1% BSA stabiliser and 0.5 mM IBMX. At the start of each assay day the potency of AVP was assessed and an EC80concentration determined for subsequent compound profiling. Assays were performed by first an addition of 2.5 μl of control or test compound at 4× final concentration in stimulation buffer followed by 2.5 μl of AVP at 4× final concentration in stimulation buffer. After a 1 hour reaction, detection reagents were added by first an addition of 5 μl EU-cAMP tracer, followed by 5 μl ULight-anti-cAMP both diluted as per the manufacturer's instructions. After a one hour incubation, plates were ready to be read (signals then remained stable for up to 24 hours). Changes in time resolved fluorescence were monitored with excitation via a laser (337 nm) measuring both 615 nm and 665 nm emission wavelengths. For the AVP potency determination the first addition consisted of stimulation buffer containing 3% v/v DMSO and the second addition a 10-point dilution series of AVP (1:3 dilutions from 0.1 nM) in stimulation buffer. For compound profiling, test compounds were dispensed by the Labcyte Echo (10-point curve, 1:3 dilutions) in a target 0.1 μl volume then diluted 750-fold in stimulation buffer containing 3% DMSO prior to addition to the cells. At the end of the incubation period, 2.5 μl of AVP in stimulation buffer was added at the previously determined EC80concentration. In-plate controls for the assay include a Tolvaptan concentration-response curve as the reference V2 antagonist and an AVP concentration-response curve to confirm the reproducibility of the EC80used for the compound challenge. Data for fluorescence at 665 nm is normalised to in-plate assay controls comprising DMSO matched solutions of assay buffer without agonist (100% inhibition) and AVP EC80(0% inhibition). Example B4 Oxytocin Receptor Antagonist Assay This assay was performed in CHEM-1 cells expressing the human Oxytocin Receptor (hOTR) to determine the inhibitory effect of the compounds of the invention on the human Oxytocin receptor. Oxytocin evokes an increase in intracellular calcium in CHEM-1-hOTR cells which is measured in a fluorescence assay on the FLIPRTETRAusing calcium sensitive dyes. Test compounds were assessed for their ability to affect the magnitude of the response to oxytocin, with antagonists showing a concentration-dependent reduction in the oxytocin-mediated fluorescence. Compounds displaying potency at the vasopressin V1a receptor of <100 nM were progressed to selectivity testing against hOTR and were tested in triplicate in a 10-point, 1:3 dilution series starting at a nominal concentration of 3 μM in the assay. CHEM-1-hOTR ready was used to assay frozen cells (Eurofins #HTS090RTA) which are supplied with a proprietary Media Component. Day 1 of the assay: Cells were thawed in a 37° C. water bath and diluted with the supplied Media Component to a final volume of 10 ml. The cell suspension was centrifuged at 1000 rpm for 3 min at room temperature and the supernatant was discarded. The cell pellet was resuspended in Media Component (10.5 ml) and the cells (25 μL) were dispensed into a poly-D-lysine coated black, clear bottomed, 384-well plate. The plates were incubated overnight at 37° C., 5% CO2. Day 2: At the start of each assay day the potency of oxytocin was assessed and an EC80concentration was determined for subsequent compound profiling. Assays were performed using a two-step addition protocol on the FLIPRTETRA; first addition of 5 μl of control or test compound at 10× final in assay buffer with 15 min incubation at 37° C., 5% CO2followed by 10 μl of oxytocin at 6× final concentration in assay buffer. Changes in fluorescence were monitored for 3 min after both additions on the FLIPRTETRAusing 470-495 nm excitation and 515-575 nm emission wavelengths. The assay buffer consisted of HBSS (+Ca/+Mg) supplemented with 20 mM HEPES, and for the preparation of the oxytocin agonist only, 0.1% w/v bovine serum albumin. The assay was initiated by the removal of growth media from the cells and replaced with 45 μl of Calcium-6 dye (Molecular Devices) prepared at 1× in assay buffer. Cells were loaded with dye for 60-90 min at 37° C., 5% CO2before initiation of the FLIPRTETRAprotocol. For the oxytocin potency determination the first addition consisted of assay buffer containing 3% v/v DMSO and the second addition involved a 10-point dilution series of oxytocin (1:3 dilutions from 1 μM) in assay buffer supplemented with 0.1% BSA. For compound profiling, test compounds were first serially diluted in DMSO (10-point curve, 1:3 dilutions) then diluted 33.3-fold in assay buffer prior to addition to the dye loaded cells on the FLIPRTETRA. At the end of the incubation period 10 μl of oxytocin in assay buffer containing 0.1% BSA was added at the previously determined EC80concentration. In-plate controls for the assay include a L-368,899 concentration-response curve as the reference OTR antagonist and an oxytocin concentration-response curve to confirm the reproducibility of the EC80used for the compound challenge. MAX-MIN raw data is normalised to in-plate assay controls comprising DMSO matched solutions of assay buffer without agonist (100% inhibition) and oxytocin EC80(0% inhibition). Activity expressed as IC50of representative compounds against the V1a, V1b, V2, and OT receptors is provided in Table 24 below. With respect to V1a, V1b, V2, and OT activity: “++++” denotes an IC50of less than 100 nM; “+++” denotes an IC50of from 100 nM to less than 500 nM; “++” denotes an IC50of from 500 nM to less than 1000 nM; and “+” denotes an IC50of 1000 nM or more. Reference compounds were assessed in the in vitro antagonist assay with the following results: balovaptan—V1A (++++), V1B (+), V2 (+), OT (+); relcovaptan—V1A (++++), V1B (−), V2 (−), OT (−); JNJ-17308616—V1A (++++), V1B (+), V2 (+++), OT (+). TABLE 24Activity of Representative CompoundsCpdV1aV1bV2OTNo.IC50IC50IC50IC501+++++++2+++++++3+++++++4+++++++5+++−−+6++++−−+7+++−−+8+++++++9+++−−++10+++−−+11++−−+12+++−−+13+++−−+14+−−+15+−−+16++++−−+24+−−−25+−−−26+++−−−27+−−−28+++++++29+−−−30+++−−−31+++−−−32+−−−33+−−−34++−−−35+++−−−36++++−−−37++++−−+38++++−−−39+++++++40++++−−−41+++++++++++42+++++++++++43++++−−−44++++−−−45+++++++++46+++++++47+++++++48++++++++49+++++++++50+++++++++51+++−−−52++++++++++53++++−−−54++++++++55+++−−−56++++++++++57+++++++58++++++++59++++++++60+++−−−61++++++++++62++++−−−63++++++++64+++++++++65++++−−+66+++−−−67+++++++68++++−−+69+++++++++++70++++++++++71+++++++72+++++++++73++++−−+74++++−−+75++++−−−78+++++++++++79++++++++++81+++++++82++++−−−90+++++++++91++++−−++92++++−−+93++++−−+94++++−−+95++++−−++96++++−−+++97++++−−−107++++−−+108++++−−+109++++−−+110++++−−+111++++−−+++112++++−−+113++++−−+114++++−−+115++++−−−116++++−−+122+++++++++124+++−−−125++++−−+126+−−−134+++++++136+++++++137++++−−−138++++−−+142++++++++142A+++++++142B++++++147++++−−−153+++++++156++++−−+157++++−−+158++++−−−159++++−−−160++++−−−161++++−−+162++++−−−163++++−−−163A+++++++++163B++−−−164+++++++++165++++++++165A++++++++165B++−−−166+++++++166A+++++++166B++−−−167+++++++167A+++++++167B+−−−168+++++++168A++++++++168B+++−−−169+++++++++169A+++++++++169B+++−−−170+++++++170A++++++++170B++−−−171+++++++171A+++++++171B+++++172+++++++172A++++−−−172B+−−−173+++++++++174++++++++174A+++++++++175+++++++176++++−−−177+++++++178++++−−−178A+++++++++++178B+++−−−179+++−−−179A+++++++179B+++−−−180+++++++++++181++++−−−181A+++++++++++181B+++−−−182+++++++++183+++−−+++184+++−−−184A++++−−−184B+−−−185++++−−++186+++−−−187+++−−−187A++++−−−187B+−−−188++++−−+++188A++++−−−188B+−−−189++++−−+190++++−−−191++++−−++191A+++++++++191B+−−−192++++−−−192A+++++++192B+−−−193++++−−+194+++++++194A++++−−−194B+−−−195+++++++++196+++++−+197+++−−−198+++−−−199+++++−+200+++−−−201A++++++−201B++−−−202A+++++++202B+−−+203A++++++++++203B+−−+204A+++++−+++204B++++−+205A+++++−++205B++−+206A+++++−+++206B++++−+217++++−−−218++++−−−219++++−−−220++++−−−221++++−−−222++++−−−223++++−+++++224++++−−−225++++−−−226++++−−−227++++−−−228++++−−−229++++−++++++230++++−−−231+++−−−232++++−−−233++++−−−234+++−−−235++++−−−236++++−−−237++++−−−238++++−−−239+++−−−240++++−−−241++++−−−242++++−−−243+++−−−244++++−−−245++++−−−246++++−−−247++++−−−248++++−−−249++++−−−250++++−−−251+++−−−252++++−−−253+++−−−254+++−−−255++++−++++256++++−−−257++−−−258+−−−259++++−−−260++++−−−261++++−−−262++++−−−263++++−−−264++++−−−265+++−−−266++−−−267+−−−268+++−−−269++−−−270+++−−−271++++−−−272++++−−−273++++−−−274++++−−−275++++−−−276++++−−−277+++−−−278++++−−− Example B5 MDCK-MDR1 Effective Efflux Ratio The MDR1-MDCK effective efflux assay was performed as described in the BioFocus Standard Operating Procedure, ADME-SOP-56. Both wild-type (WT) and MDR1-MDCK cells (Solvo Biotechnology) were seeded onto 24-well Transwell plates at 2.35×105 cells per well and used in confluent monolayers after a 3 day culture at 37° C. under 5% CO2. For both cell types, test and control compounds (propranolol, vinblastine) were added (10 μM, 0.1% DMSO final, n=2) to donor compartments of the Transwell plate assembly in assay buffer (Hanks balanced salt solution supplemented with 25 mM HEPES, adjusted to pH 7.4) for both apical to basolateral (A>B) and basolateral to apical (B>A) measurements. Incubations were performed at 37° C., with samples removed from both donor and acceptor chambers at T=0 and 1 hour and compound analysed by mass spectrometry (LC-MS/MS) including an analytical internal standard. Apparent permeability (Papp) values were determined from the relationship: Papp=[Compound AcceptorT=end]×VAcceptor/([Compound DonorT=0]×VDonor)/incubation time×VDonor/Area×60×10−6 cm/s Where V is the volume of each Transwell compartment (apical 125 μL, basolateral 600 μL), and concentrations are the relative MS responses for compound (normalized to internal standard) in the donor chamber before incubation and acceptor chamber at the end of the incubation. Area=area of cells exposed for drug transfer (0.33 cm2). Efflux ratios (Papp B>A/Papp A>B) were calculated for each compound from the mean Papp values in each direction for both wild-type and MDR1-MDCK cells. The MDR1-MDCK cell line has been engineered to over-express the efflux transporter, MDR1 (P-glycoprotein), and a finding of good permeability B>A, but poor permeability A>B, suggests that a compound is a substrate for this transporter. In order to confirm the involvement of MDR1 in any efflux seen, an “effective efflux ratio” (EER) was calculated by comparing compound efflux ratios (ER) in the two cell types, i.e. EER=ER (MDR1-MDCK)/ER (wild-type MDCK) This ratio illustrates the effect of the over-expressed MDR1 normalised for the background movement of compound through the wild-type cells. Lucifer Yellow (LY) was added to the apical buffer in all wells to assess viability of the cell layer. Compound recovery from the wells was determined from MS responses (normalized to internal standard) in donor and acceptor chambers at the end of incubation compared to response in the donor chamber pre-incubation. TABLE 25Papp (10{circumflex over ( )}−6 cm/sec), Efflux ratio, and Effective efflux ratioCompoundPapp ABMDR1MDR1Papp ABWTNo.MDR1EREERWTER1++++++++++8++++++++++++++++++16++++++19++++++++++36++++++++52+++++++++++++56++++++++++++++57++++++++++59++++++++++++61++++++++++++63+++++++++++64+++++++65+++++++++++67++++++++++69+++++++++++++++++70+++++++++++++71+++++++++++72++++++++++73+++++++74++++++++++75+++++++75++++++++++++78++++++++++++++++79++++++++++++81+++++++++++90+++++++++++++++91+++++++++++92+++++++++++93+++++94+++++++95++++++++++96++++++++++++++++++107+++++++110+++++++111+++++++++++++++++113++++++++++114+++++115++++++++++++116+++++++++++122++++++++++++125++++++++++136+++++++137+++++142+++++++++++++++142A++++++++++++++++147+++++153+++++++++156+++++++++++++160+++++162+++++163++++++++++++++++++163A++++++++++++++++++165+++++++++++165A++++++++++165B++++++++++166++++++++++166A++++++++++166B++++++++++167+++++++++++++++++167A++++++++-++++++++168+++++++++++168A++++++++-++++++++169+++++++++++++++++169A++++++++-++++++++170+++++++++++++171+++++++++++++171A++++++++++172++++++++++++++++172A+++++++++++++++173++++++++++++++++++174+++++++++++++++++174A++++++++++++++++++175A+++++++++++175B+++++++++++176++++++++++++177++++++++++178A++++++++-++++++++179++++++++++++++180+++++++-++++++++181A++++++++++++++++++182+++++++++++++++++184A++++++++++++185+++++++++++++++++187++++++-++++++++191A+++++++++++++++++192+++++++++++++++++192A++++++++++++++++193+++++++++++++++++194++++++++++++194A++++++++++++195+++++++++++++++++197+++++++++++++++200+++++++++201A++++++++++++++++++202+++++++++++202A++++++++++202B++++++++++203+++++++++++++++ MDCK II Cell Permeability Assay Procedure MDCK II cell culture media was prepared using Dulbecco's modified eagle medium (DMEM), fetal bovine serum (FBS) 10%, Glutamax 1% and PenStrep 1% and was sterile-filtered. Transwell 24-well plates of MDCK II_WT or MDCK II-MDR1 cells were prepared and the plates were fed every alternate day until the day of use. Plates were used on 5th day after cell plating. Preparation for changing the media of the basal plate was conducted by filling all wells of a 24 well sterile plate with 900 μl of culture media and placing it in an incubator until use. Then the apical section of plate was lifted out and lowered onto an empty basal plate, followed by aspiration of 200 μl of the culture media from the apical compartment and replacement with 200 μl of fresh culture media. This step was repeated twice for a total of 3 washes followed by removal of the basal plate from the incubator and placement of the plate in the hood. The apical section of plate was then added to the basal plate and returned to incubator. On the day of the assay, approximately 3 ml of 1000-fold diluted compound solution (required concentration for the assay) was prepared in transport buffer using the following volumes: 200 μl/insert/well (apical application) and 780 μl/insert/well (basal application). The basal assay plate was prepared by adding 750 μl of transport buffer (Hank's Balanced Salt Solution) to A-B wells, and 780 μl of diluted compound solution to B-A wells. Triplicate samples of 10 μl each were collected from basal compartments of B-A wells for T0, and then basal assay plates were placed in the incubator. MDCK plates were placed in the hood and the apical section of the plates were lifted out and lowered onto empty basal plates. 200 μl of the media was removed from the apical wells and replaced with 200 μl of fresh transport media, and this step was repeated twice for a total of 3 washes. 200 μl of the media was removed from the apical wells and replaced with 200 μl of the diluted compound (for A-B wells) or 200 μl of fresh transport buffer (for B-A wells). Triplicate samples were collected (10 μl each) from apical compartments of A-B wells for T0. Basal plates were removed from the incubator and transferred to the apical section of the plate to the basal plate and the assay plates were covered and returned to the incubator. The T0 samples were diluted with 40 μl transport buffer and 100 μl of room temperature quench solution was added to the diluted T0 samples. 50 μl of all T0 samples were mixed and transferred to T0 sample plates and diluted with 100 μl of MilliQ water for bioanalysis. At T-2 hrs, 3 replicate 10 μl samples from all apical compartments and B-A basal compartments were collected; and, 3 replicate 50 μl samples from A-B basal compartments were collected. The 10 μl samples were diluted with 40 μl transport buffer. 100 μl of quench solution was added to all T-2 hrs samples. 50 μl of all T-2 hrs samples were mixed and transferred to sample plates and diluted with 100 μl of MilliQ water for bioanalysis. Analyte levels (peak area ratios) were measured on apical (A) and basolateral (B) sides at T0 and T2 hrs. A-to-B and B-to-A fluxes were calculated (mean of n=3 measurements). Apparent permeability (Papp, 10{circumflex over ( )}−6 cm/sec) was calculated as dQ (flux)/(dt×Area×Concentration). The efflux ratio was (B-to-A)/(A-to-B) ratio [i.e., Papp(B-A)/Papp(B-A)]. A ratio >2-3 was determined as evidence of efflux, and compounds that demonstrate efflux ratios in or above this range PGP efflux can be confirmed by testing+/−pgp inhibitor (dosing solutions prepared with and without verapamil at a final assay concentration of 25 μM). The ability of a test compound to penetrate the blood brain barrier and avoid efflux by transporters expressed in the brain, can be roughly correlated with the Papp(A-B) and the efflux ratio (as defined above), respectively, the results are provided in Table 26. (+) denotes an apparent permeability <7 (10{circumflex over ( )}−6 cm/sec); (++) denotes >7 (10{circumflex over ( )}−6 cm/sec) but <10 (10{circumflex over ( )}−6 cm/sec; (+++) denotes >10 (10{circumflex over ( )}−6 cm/sec) but <20 (10{circumflex over ( )}−6 cm/sec); and (++++) denotes >20 (10{circumflex over ( )}−6 cm/sec). PGP efflux can be confirmed by testing+/−pgp inhibitor (dosing solutions prepared with and without verapamil at a final assay concentration of 25 μM). Reference compounds were assessed in the in vitro MDCK apparent permeability, efflux ratio, and effective efflux ratio assays (MDR1 and WT) with the following results: balovaptan—MDR1AB (++++), MDR1ER (++), MDR1EER (++), WTAB (+++), WTER (++++); JNJ-17308616—MDR1AB (+), MDR1ER (+), MDR1EER (+), WTAB (+++), WTER (+). TABLE 26Papp (10{circumflex over ( )}−6 cm/sec) and Efflux ratioCompoundNo.Papp AB MDR1MDR1 ER218++++++++220+++++++223+++++++224++++++++225+++++++226++242++++++255+++++++259++++++++ In Vivo Activity Example B6 Evaluation of Behavioral, Biochemical and/or Neurophysiological Characteristics in the Valproate Model Valproate (VPA) is an anticonvulsant drug commonly prescribed for patients with epilepsy. During pregnancy, administration of VPA elevates the risk of neurodevelopmental disorders in the offspring and this effect has been modeled similarly in rodents to better understand the mechanisms underlying the VPA-induced neurodevelopmental changes. V1a antagonists are assessed for preventative and/or restorative effects in rodents following the administration of a single injection of valproate acid (600 mg/kg) or vehicle (sham) to pregnant females dams on gestational day 13 (embryonic day 13). Pregnant dams are monitored on a daily basis for changes in weight and health, or in their feeding patterns. After birth, pups are monitored for any signs of physical abnormalities (e.g., weights, food and water intake, postnatal day of eye opening). Selective studies are conducted to evaluate behavioral, biochemical and/or neurophysiological characteristics of the valproate treated animals as compared to control animals. More specifically, the effects of V1 antagonists administered to VPA treated animals are assessed using standard methodology for behavioral changes such as anxiety (e.g., ultrasonic vocalizations, elevated plus maze), learning and memory (e.g., Morris water maze, novel object recognition), social interactions, sensorimotor gating and locomotor activity. Biochemical changes are measured by assessing synaptic proteins and mRNA (e.g., gamma-aminobutyric acid [GABA] synthesis, glutamic acid decarboxylase [GAD], brain derived neurotrophic factor [BDNF]). Neurophysiological characteristics are assessed by whole cell recordings of the electrophysiological properties of neurons from VPA- and sham-treated animals to identify differences in neuronal function with and without V1 antagonists. Activity and/or Telemetry Studies in Rodents and Non-Human Primates to Assess Sleep/Wake Cycles and Circadian Rhythms: The vasopressin system is important in regulating biological circadian rhythms and re-entrainment following environmental alterations. In these studies, animals are housed on a 12 hour light/dark cycle and activity is monitored using an infrared beam break system or by wheel running (rodents) or by activity monitors attached to the collar of the animal (non-human primates). Activity data is collected for up to 30 days to establish circadian rhythms and changes induced by phase shifting the light/dark cycle by e.g., 4, 8 or 12 hours is recorded and analyzed. V1a antagonist is administered to improve re-entrainment as measured by re-establishment of the regular activity patterns. Additional endpoints may include cognitive assessment (e.g., spatial working memory). Implantation of a telemetry device with electrodes to record electroencephalography/electromoyography/electroculography (EEG/EMG/EOG) for staging sleep/wake cycles is used. In this case, EEG/EMG electrodes and transmitters are implanted in fully anesthetized animals by trained surgeons. The transmitter module is implanted subcutaneously below the scapular region or into the abdomen. Biopotential leads are guided subcutaneously from the back to the head via a midline incision. Using a stereotaxic approach, stainless steel screws are implanted into the skull over areas of interest until the tips are on the surface of the dura mater. The biopotential leads are wrapped around the screws and referenced. The EMG or EOG leads are sutured into the temporalis muscle or intra-ocular muscle, respectively. Animals receive postoperative analgesia and antibiotics and recover for a minimum of 21-days before testing. Receiver boards are placed in close proximity to the animal to facilitate real-time EEG/EMG/EOG recordings during testing. Physiological Measures Vasopressin is an important regulator of water conservation and blood pressure in the body and its release into the peripheral blood supply can be induced by an increase in plasma osmolality. In healthy adults, a rise in plasma osmolality of 1-2% above basal level produces thirst that promotes water intake and normalization of osmolality. Intravenous administration of a hyperosmolic solution to rodents or humans increases the plasma vasopressin concentration and other measures (e.g., thirst, urine output and vasoconstriction). V1a antagonist is evaluated for its ability to alter plasma vasopressin concentrations, vasoconstriction and/or urine output following administration of a hyperosmolic solution. Example B7 Arginine-Vasopressin (AVP) Induced Phospho-Erk Measurement in Native Tissue When V1a receptors are coupled to phospholipase C (PLC), they increase intracellular Ca2+ concentrations and protein kinase C (PKC) activity, and transactivate the mitogen-activated protein kinases/extracellular signal-regulated kinase (MAPK/Erk) and PI3 kinase/Akt pathways upon activation (Chen et al., J Neuroendocrinol. 2010). Rat choroid plexus (RCP) cell lines express functional V1a receptors measured by increased calcium concentrations in response to V1a receptor agonists (Battle et al., Biochem Biophys Res Comm 2000). In these studies, RCP were stimulated with AVP and V1a receptor antagonists reference compounds relcovaptan and balovaptan and test compound 142A were evaluated. RCP P9(18) cells were seeded 30K/well, in 100p growth medium containing 10% FBS in polystyrene 96-well plates and incubated at 37° C., 5% CO2and incubated overnight. The following day, the growth medium was replaced with 50p pre-warmed HBSS containing 20 mM HEPES and the cells were incubated at 37° C., 5% CO2for 1.5 hrs. 1 mM AVP (Sigma V9879) was freshly prepared in distilled water in a glass vial, and diluted to 3× concentrations in HBSS containing 20 mM HEPES and 0.1% BSA in glass vials and kept on ice. Cells were treated with 25 μl 3× vehicle, 3× eBioscience Cell Stimulation Cocktail (Thermo Fisher Scientific 00-4970-93) or 3× AVP and incubated at 37° C., 5% CO2for 5, 10 or 20 min. Final concentrations of AVP: 10, 100 or 1000 nM. Final concentrations of components in Cell Stimulation Cocktail: 81 nM PMA, 1.34 μM ionomycin, 0.2% ethanol. Cells were lysed with 25 μl 4×CST lysis buffer containing protease and phosphatase inhibitors, PMSF and SDS and then stored at −80° C., for 48 h. The lysates were thawed, centrifuged at 2000 g for 30 min at 4° C. and 40 μl supernatants assayed for pERK1/2 (Thr202/Tyr204; Thr185/Tyr187) and total ERK1/2 using MSD kit K15107D. The MSD ECL data for the lysates were corrected for no cell blanks, then phospho-protein levels expressed as a ratio to the total ERK1/2 level. The ratios were expressed as fold-change from the vehicle-treated control at each timepoint. In study 1, the IC50values for the compounds tested, ralcovaptan, 142A and balovaptan were: 0.03 nM, 16.0 nM, 85.6 nM and 10.9 nM, respectively. In the second study, the IC50values for the compounds tested, ralcovaptan, 142A and balovaptan were: 0.08 nM, 20.0 nM, 56.7 nM and 13.6 nM, respectively. Ralcovaptan and balovaptan are purchased commercially. Example B8 Arginine-Vasopressin (AVP) Induced Behavior in Mouse Administration of Arginine-Vasopressin (AVP) intracerebroventricularly (i.c.v.) elicits characteristic scratching, digging and grooming behavior in mice that can be measured readily and is sensitive to blockade with vasopressin antagonists (Meisenberg, 1988; Bleickart et al., 2009). Male CD-1 mice (Charles River Germany) weighing 22-25 g upon the study in-life are used for this study. Animals are housed in groups of 4-5 per cage in standard temperature (22±1° C.) and light-controlled environment (lights on from 7 am to 8 μm), with ad libitum access to food and water. Prior to commencing any procedures to the mice, they are allowed to habituate in the vivarium for a minimum of 7 days. Anesthesia is induced in a plexiglass chamber for 2-3 min with 5% isoflurane, and maintained through a snout mask with 1-2% isoflurane thereafter. A homeothermic blanket system with a rectal probe is used to monitor and maintain the animal's body temperature at 37.0° C.±1.5° C. during the operation. Anesthetized mice are placed in a stereotaxic apparatus and skin between the ears shaved and disinfected with povidone-iodine solution (Betadine). A 10-μl Hamilton syringe with 28-gauge needle is used for the i.c.v. injections. All animals receive identical AVP injections (3.689 μM) or sterile saline (0.9% sodium chloride solution) into the right lateral ventricle at the following coordinates: AP=+0.5 mm; ML=+1.0 mm; DV=−2.5 mm (approximately from bregma). The actual coordinates are calculated by the distance from the point in midline between the eyes and no skin incision is made. After the needle is placed in the ventricle and the AVP is delivered, the needle is left in place for 3 minutes before withdrawal. Finally, the mouse is detached from the anesthesia mask and immediately placed in a clean cage to commence the observation. Mice are observed and video-recorded for 15 minutes following AVP/saline administration and behaviors are measured (in seconds) and a cumulative time is calculated. The following behaviors are considered as AVP-related: scratching of limbs or torso, digging, licking and face washing (swiping of face). Using this assay, balovaptan (100 and 300 mg/kg, po) and JNJ-17308616 (30, 100 mg/kg, po) were evaluated for antagonist activity to AVP-induced scratching behaviors. Balovaptan was effective at 100 mg/kg and JNJ-17308616 showed weak effects at 100 mg/kg. The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, including U.S. Provisional Application 62/515,473 filed Jun. 5, 2017, are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications and publications to provide yet further embodiments. These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure
339,632
11858945
DETAILED DESCRIPTION OF THE INVENTION In one embodiment of the present invention is a compound of Formula (I) as described above, or a pharmaceutically acceptable salt thereof. In one embodiment of the present invention, the compound of Formula (I) is represented by Formula (I-A) or Formula (I-B), or a pharmaceutically acceptable salt, ester or prodrug thereof: wherein A, B, Q1, R1, R2, R3, and R4are as previously defined. In a preferred embodiment, the compound of Formula (I) has the stereochemistry shown in Formula (I-A). In certain embodiments of the compounds of Formula (I), R1is hydrogen, optionally substituted —C1-C6alkyl; optionally substituted —C3-C6cycloalkyl; optionally substituted C3-C6cycloalkyl-C1-C2-alkyl; optionally substituted aryl; optionally substituted arylalkyl; optionally substituted heteroarylalkyl. In certain embodiments of the compounds of Formula (I), R2is hydrogen, optionally substituted —C1-C4alkyl; optionally substituted —C3-C6cycloalkyl; optionally substituted aryl; optionally substituted arylalkyl; or optionally substituted heteroarylalkyl. In certain embodiments of the compounds of Formula (I), R1is selected from the following groups: In certain embodiments of the compounds of Formula (I), R2is hydrogen, and R1is hydrogen, optionally substituted —C1-C6alkyl; optionally substituted —C3-C6cycloalkyl; optionally substituted C3-C6cycloalkyl-C1-C2-alkyl; optionally substituted aryl; optionally substituted arylalkyl; or optionally substituted heteroarylalkyl. In certain embodiments of the compounds of Formula (I), R2is hydrogen, and R1is selected from the following groups: In certain embodiments of the compounds of Formula (I) or Formula (Ia), R3is hydrogen or optionally substituted —C1-C4alkyl; and R4is hydrogen or optionally substituted —C1-C4alkyl. In certain embodiments of the compounds of Formula (I), R3is hydrogen, -Me, -Et, —Pr, -i-Pr, -allyl, —CF3, —CD3or cyclopropyl. In certain embodiments of the compounds of Formula (I), R4is hydrogen, -Me, -Et, —Pr, -i-Pr, -allyl, —CF3or cyclopropyl. In certain embodiments of the compounds of Formula (I), R3is hydrogen, and R4is hydrogen. In certain embodiments of the compounds of Formula (I), R2is hydrogen, R3is hydrogen or —CH3, R4is hydrogen, and R1is hydrogen, optionally substituted —C1-C6alkyl; optionally substituted —C3-C6cycloalkyl; optionally substituted C3-C6cycloalkyl-C1-C2-alkyl-optionally substituted aryl; optionally substituted arylalkyl; or optionally substituted heteroarylalkyl. In certain embodiments of the compounds of Formula (I), R2is hydrogen, R3is hydrogen or —CH3, R4is hydrogen, and R1is selected from the following groups: In certain embodiments of the compounds of Formula (I), Q1is hydrogen. In certain embodiments of the compounds of Formula (I), A is derived from one of the following by removal of a hydrogen atom and is optionally substituted: In certain embodiments of the compounds of Formula (I), A is selected from the following groups, and A is optionally substituted: preferably the substituents are independently selected from halogen, CN, NH2, optionally substituted —C1-C3alkoxy, optionally substituted —C1-C3alkyl, optionally substituted —C3-C6cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl. Preferably the number of substituents is 0 to 3. In certain embodiments of the compounds of Formula (I), A is selected from the following groups, and A is optionally substituted: preferably the substituents are independently selected from halogen, CN, NH2, optionally substituted —C1-C3alkoxy, optionally substituted —C1-C3alkyl, optionally substituted —C3-C6cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl. Preferably the number of substituents is 0 to 3. In certain embodiments of the compounds of Formula (I), A is selected from the following groups, and A is optionally substituted: In certain embodiments of the compounds of Formula (I), B is selected from the following groups, and B is optionally substituted: In certain embodiments, the compound of Formula (I), is represented by Formula (II), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, B, R1, R3, R4, and Q1are as previously defined. In certain embodiments, the compound of Formula (I), is represented by Formula (III), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, B, R1, R2, R3, and Q1are as previously defined. In certain embodiments, the compound of Formula (I), is represented by Formula (III-1), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, B, R1, and R3, are as previously defined. In certain embodiments, the compound of Formula (I), is represented by Formula (IV), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, B, R1, and Q1are as previously defined. In certain embodiments, the compound of Formula (I), is represented by Formula (V), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, B, and R1are as previously defined. In certain embodiments, the compound of Formula (I), is represented by Formula (V-A), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, B, and R1are as previously defined. In certain embodiments, the compound of Formula (I) is represented by Formula (VI), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, Q1, R1, R2, R3, and R4are as previously defined, each R9is independently selected from:1) Halogen;2) —CN;3) —OR13;4) —SR13;5) —NR13R14;6) —OC(O)NR13R14;7) Optionally substituted —C1-C6alkyl;8) Optionally substituted —C3-C8cycloalkyl;9) Optionally substituted 3- to 8-membered heterocycloalkyl;10) Optionally substituted aryl; and11) Optionally substituted heteroaryl; and n is 0, 1, 2, 3, or 4; preferably, n is 0, 1, or 2. In certain embodiments, the compound of Formula (I) is represented by Formula (VII), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, Q1, R1, R3, R4, R9, and n are as previously defined. In certain embodiments, the compound of Formula (I) is represented by Formula (VIII), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, Q1, R1, R2, R3, R9, and n are as previously defined. In certain embodiments, the compound of Formula (I) is represented by Formula (IX), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, Q1, R1, R3, R9, and n are as previously defined. In certain embodiments, the compound of Formula (I) is represented by Formula (X), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, R1, and R3are as previously defined. In certain embodiments, the compound of Formula (I) is represented by Formula (X-A), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein A, R1, and R3are as previously defined. In certain embodiments of the compounds of Formula (X), or Formula (X-A), wherein R3is hydrogen or —CH3; R1is selected from: and A is derived from one of the following by removal of a hydrogen atom and is optionally substituted: In certain embodiments, the compound of Formula (I) is represented by one of Formulae (XI-1)˜(XI-5), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein R1, R3, R4, R9, and n are as previously defined, each R10is independently selected from:1) Halogen;2) —CN;3) —OR13;4) —SR13;5) —NR13R14;6) —OC(O)NR13R14;7) Optionally substituted —C1-C6alkyl;8) Optionally substituted —C3-C8cycloalkyl;9) Optionally substituted 3- to 8-membered heterocycloalkyl;10) Optionally substituted aryl; and11) Optionally substituted heteroaryl; and m is 0, 1, 2, 3, or 4; preferably m is 0, 1 or 2. In certain embodiments, the compound of Formula (I) is represented by one of Formulae (XII-1) to (XII-5), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein R1, R3, R4, R10, and m are as previously defined. In certain embodiments, the compound of Formula (I) is represented by one of Formulae (XII-1A) to (XII-5A), or a pharmaceutically acceptable salt, ester, or prodrug thereof: wherein R1, R3, R4, R10, and m are as previously defined. In certain embodiments, the compound of Formula (I) is represented by one of Formulae (XII-1) to (XII-5), or Formulae (XII-1A) to (XII-5A), or a pharmaceutically acceptable salt, ester, or prodrug thereof, wherein m is 0, 1, 2 or 3; R10is selected from halogen, CN, NH2, optionally substituted —C1-C3alkoxy, optionally substituted —C1-C3alkyl, optionally substituted —C3-C6cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; R3is hydrogen or —CH3, and R1is selected from the groups below: Preferably, R3is hydrogen, and R1is It will be appreciated that the description of the present invention herein should be construed in congruity with the laws and principles of chemical bonding. In some instances, it may be necessary to remove a hydrogen atom in order to accommodate a substituent at any given location. It will be yet appreciated that the compounds of the present invention may contain one or more asymmetric carbon atoms and may exist in racemic, diastereoisomeric, and optically active forms. It will still be appreciated that certain compounds of the present invention may exist in different tautomeric forms. All tautomers are contemplated to be within the scope of the present invention. The compounds of the present invention and any other pharmaceutically active agent(s) may be administered together or separately and, when administered separately, administration may occur simultaneously or sequentially, in any order. The amounts of the compounds of the present invention and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. The administration in combination of a compound of the present invention and salts, solvates, or other pharmaceutically acceptable derivatives thereof with other treatment agents may be achieved by concomitant administration in: (1) a unitary pharmaceutical composition including both compounds; or (2) separate pharmaceutical compositions each including one of the compounds. In certain embodiments of the combination therapy, the additional therapeutic agent is administered at a lower dose and/or dosing frequency as compared to dose and/or dosing frequency of the additional therapeutic agent required to achieve similar results in treating or preventing coronavirus. It should be understood that the compounds encompassed by the present invention are those that are suitably stable for use as pharmaceutical agent. Definitions Listed below are definitions of various terms used to describe this invention. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group. The term “aryl,” as used herein, refers to a mono- or polycyclic carbocyclic ring system comprising at least one aromatic ring. Preferred aryl groups are C6-C12-aryl groups, including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, and indenyl. A polycyclic aryl is a polycyclic ring system that comprises at least one aromatic ring. Polycyclic aryls can comprise fused rings, covalently attached rings or a combination thereof. The term “heteroaryl,” as used herein, refers to a mono- or polycyclic aromatic radical having one or more ring atom selected from S, O and N; and the remaining ring atoms are carbon, wherein any N or S contained within the ring may be optionally oxidized. In certain embodiments, a heteroaryl group is a 5- to 10-membered heteroaryl, such as a 5- or 6-membered monocyclic heteroaryl or an 8- to 10-membered bicyclic heteroaryl. Heteroaryl groups include, but are not limited to, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, quinoxalinyl. A polycyclic heteroaryl can comprise fused rings, covalently attached rings or a combination thereof. A heteroaryl group can be C-attached or N-attached where possible. In accordance with the invention, aryl and heteroaryl groups can be substituted or unsubstituted. The term “bicyclic aryl” or “bicyclic heteroaryl” refers to a ring system consisting of two rings wherein at least one ring is aromatic; and the two rings can be fused or covalently attached. The term “alkyl” as used herein, refers to saturated, straight- or branched-chain hydrocarbon radicals. “C1-C4alkyl,” “C1-C6alkyl,” “C1-C8alkyl,” “C1-C12alkyl,” “C2-C4alkyl,” and “C3-C6alkyl,” refer to alkyl groups containing from 1 to 4, 1 to 6, 1 to 8, 1 to 12, 2 to 4 and 3 to 6 carbon atoms respectively. Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, neopentyl, n-hexyl, n-heptyl and n-octyl radicals. The term “alkenyl” as used herein, refers to straight- or branched-chain hydrocarbon radicals having at least one carbon-carbon double bond. “C2-C8alkenyl,” “C2-C12alkenyl,” “C2-C4alkenyl,” “C3-C4alkenyl,” and “C3-C6alkenyl,” refer to alkenyl groups containing from 2 to 8, 2 to 12, 2 to 4, 3 to 4 or 3 to 6 carbon atoms respectively. Alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, 2-methyl-2-buten-2-yl, heptenyl, octenyl, and the like. The term “alkynyl” as used herein, refers to straight- or branched-chain hydrocarbon radicals having at least one carbon-carbon triple bond. “C2-C8alkynyl,” “C2-C12alkynyl,” “C2-C4alkynyl,” “C3-C4alkynyl,” and “C3-C6alkynyl,” refer to alkynyl groups containing from 2 to 8, 2 to 12, 2 to 4, 3 to 4 or 3 to 6 carbon atoms respectively. Representative alkynyl groups include, but are not limited to, ethynyl, 2-propynyl, 2-butynyl, heptynyl, octynyl, and the like. The term “cycloalkyl”, as used herein, refers to a monocyclic or polycyclic saturated carbocyclic ring, such as a bi- or tri-cyclic fused, bridged or spiro system. The ring carbon atoms are optionally oxo-substituted or optionally substituted with an exocyclic olefinic double bond. Preferred cycloalkyl groups include C3-C12cycloalkyl, C3-C6cycloalkyl, C3-C8cycloalkyl and C4-C7cycloalkyl. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl, cyclooctyl, 4-methylene-cyclohexyl, bicyclo[2.2.1]heptyl, bicyclo[3.1.0]hexyl, spiro[2.5]octyl, 3-methylenebicyclo[3.2.1]octyl, spiro[4.4]nonanyl, and the like. The term “cycloalkenyl”, as used herein, refers to monocyclic or polycyclic carbocyclic ring, such as a bi- or tri-cyclic fused, bridged or spiro system having at least one carbon-carbon double bond. The ring carbon atoms are optionally oxo-substituted or optionally substituted with an exocyclic olefinic double bond. Preferred cycloalkenyl groups include C3-C12cycloalkenyl, C4-C12-cycloalkenyl, C3-C8cycloalkenyl, C4-C8cycloalkenyl and C5-C7cycloalkenyl groups. Examples of cycloalkenyl include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, bicyclo[2.2.1]hept-2-enyl, bicyclo[3.1.0]hex-2-enyl, spiro[2.5]oct-4-enyl, spiro[4.4]non-2-enyl, bicyclo[4.2.1]non-3-en-12-yl, and the like. As used herein, the term “arylalkyl” means a functional group wherein an alkylene chain is attached to an aryl group, e.g., —(CH2)n-phenyl, where n is 1 to 12, preferably 1 to 6 and more preferably 1 or 2. The term “substituted arylalkyl” means an arylalkyl functional group in which the aryl group is substituted. Similarly, the term “heteroarylalkyl” means a functional group wherein an alkylene chain, is attached to a heteroaryl group, e.g., —(CH2)n-heteroaryl, where n is 1 to 12, preferably 1 to 6 and more preferably 1 or 2. The term “substituted heteroarylalkyl” means a heteroarylalkyl functional group in which the heteroaryl group is substituted. As used herein, the term “alkoxy” refers to a radical in which an alkyl group having the designated number of carbon atoms is connected to the rest of the molecule via an oxygen atom. Alkoxy groups include C1-C12-alkoxy, C1-C8-alkoxy, C1-C6-alkoxy, C1-C4-alkoxy and C1-C3-alkoxy groups. Examples of alkoxy groups includes, but are not limited to, methoxy, ethoxy, n-propoxy, 2-propoxy (isopropoxy) and the higher homologs and isomers. Preferred alkoxy is C1-C3alkoxy. An “aliphatic” group is a non-aromatic moiety comprised of any combination of carbon atoms, hydrogen atoms, halogen atoms, oxygen, nitrogen or other atoms, and optionally contains one or more units of unsaturation, e.g., double and/or triple bonds. Examples of aliphatic groups are functional groups, such as alkyl, alkenyl, alkynyl, O, OH, NH, NH2, C(O), S(O)2, C(O)O, C(O)NH, OC(O)O, OC(O)NH, OC(O)NH2, S(O)2NH, S(O)2NH2, NHC(O)NH2, NHC(O)C(O)NH, NHS(O)2NH, NHS(O)2NH2, C(O)NHS(O)2, C(O)NHS(O)2NH or C(O)NHS(O)2NH2, and the like, groups comprising one or more functional groups, non-aromatic hydrocarbons (optionally substituted), and groups wherein one or more carbons of a non-aromatic hydrocarbon (optionally substituted) is replaced by a functional group. Carbon atoms of an aliphatic group can be optionally oxo-substituted. An aliphatic group may be straight chained, branched, cyclic, or a combination thereof and preferably contains between about 1 and about 24 carbon atoms, more typically between about 1 and about 12 carbon atoms. In addition to aliphatic hydrocarbon groups, as used herein, aliphatic groups expressly include, for example, alkoxyalkyls, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and polyimines, for example. Aliphatic groups may be optionally substituted. The terms “heterocyclic” and “heterocycloalkyl” can be used interchangeably and refer to a non-aromatic ring or a polycyclic ring system, such as a bi- or tri-cyclic fused, bridged or spiro system, where (i) each ring system contains at least one heteroatom independently selected from oxygen, sulfur and nitrogen, (ii) each ring system can be saturated or unsaturated (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, (v) any of the above rings may be fused to an aromatic ring, and (vi) the remaining ring atoms are carbon atoms which may be optionally oxo-substituted or optionally substituted with exocyclic olefinic double bond. Representative heterocycloalkyl groups include, but are not limited to, 1,3-dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, 2-azabicyclo[2.2.1]-heptyl, 8-azabicyclo[3.2.1]octyl, 5-azaspiro[2.5]octyl, 2-oxa-7-azaspiro[4.4]nonanyl, 7-oxooxepan-4-yl, and tetrahydrofuryl. Such heterocyclic or heterocycloalkyl groups may be further substituted. A heterocycloalkyl or heterocyclic group can be C-attached or N-attached where possible. It is understood that any alkyl, alkenyl, alkynyl, alicyclic, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclic, aliphatic moiety or the like described herein can also be a divalent or multivalent group when used as a linkage to connect two or more groups or substituents, which can be at the same or different atom(s). One of skill in the art can readily determine the valence of any such group from the context in which it occurs. The term “substituted” refers to substitution by independent replacement of one, two, or three or more of the hydrogen atoms with substituents including, but not limited to, —F, —Cl, —Br, —I, —OH, C1-C12-alkyl; C2-C12-alkenyl, C2-C12-alkynyl, —C3-C12-cycloalkyl, protected hydroxy, —NO2, —N3, —CN, —NH2, protected amino, oxo, thioxo, —NH—C1-C12-alkyl, —NH—C2-C8-alkenyl, —NH—C2-C8-alkynyl, —NH—C3-C12-cycloalkyl, —NH-aryl, —NH-heteroaryl, —NH-heterocycloalkyl, -dialkylamino, -diarylamino, -diheteroarylamino, —O—C1-C12-alkyl, —O—C2-C8-alkenyl, —O—C2-C8-alkynyl, —O—C3-C12-cycloalkyl, —O-aryl, —O-heteroaryl, —O-heterocycloalkyl, —C(O)—C1-C12-alkyl, —C(O)—C2-C8-alkenyl, —C(O)—C2-C8-alkynyl, —C(O)—C3-C12-cycloalkyl, —C(O)-aryl, —C(O)— heteroaryl, —C(O)-heterocycloalkyl, —CONH2, —CONH—C1-C12-alkyl, —CONH—C2-C8-alkenyl, —CONH—C2-C8-alkynyl, —CONH—C3-C12-cycloalkyl, —CONH-aryl, —CONH-heteroaryl, —CONH— heterocycloalkyl, —OCO2—C1-C12-alkyl, —OCO2—C2-C8-alkenyl, —OCO2—C2-C8-alkynyl, —OCO2—C3-C12-cycloalkyl, —OCO2-aryl, —OCO2-heteroaryl, —OCO2-heterocycloalkyl, —CO2—C1-C12alkyl, —CO2—C2-C8alkenyl, —CO2—C2-C8alkynyl, —CO2—C3-C12-cycloalkyl, —CO2-aryl, —CO2-heteroaryl, —CO2-heterocyloalkyl, —OCONH2, —OCONH—C1-C12-alkyl, —OCONH—C2-C8-alkenyl, —OCONH—C2-C8-alkynyl, —OCONH—C3-C12-cycloalkyl, —OCONH-aryl, —OCONH-heteroaryl, —OCONH— heterocycloalkyl, —NHC(O)H, —NHC(O)—C1-C12-alkyl, —NHC(O)—C2-C8-alkenyl, —NHC(O)—C2-C8-alkynyl, —NHC(O)—C3-C12-cycloalkyl, —NHC(O)-aryl, —NHC(O)-heteroaryl, —NHC(O)— heterocycloalkyl, —NHCO2—C1-C12-alkyl, —NHCO2—C2-C8-alkenyl, —NHCO2—C2-C8-alkynyl, —NHCO2—C3-C12-cycloalkyl, —NHCO2-aryl, —NHCO2-heteroaryl, —NHCO2— heterocycloalkyl, —NHC(O)NH2, —NHC(O)NH—C1-C12-alkyl, —NHC(O)NH—C2-C8-alkenyl, —NHC(O)NH—C2-C8-alkynyl, —NHC(O)NH—C3-C12-cycloalkyl, —NHC(O)NH-aryl, —NHC(O)NH-heteroaryl, —NHC(O)NH-heterocycloalkyl, —NHC(S)NH2, —NHC(S)NH—C1-C12-alkyl, —NHC(S)NH—C2-C8-alkenyl, —NHC(S)NH—C2-C8-alkynyl, —NHC(S)NH—C3-C12-cycloalkyl, —NHC(S)NH-aryl, —NHC(S)NH-heteroaryl, —NHC(S)NH-heterocycloalkyl, —NHC(NH)NH2, —NHC(NH)NH—C1-C12-alkyl, —NHC(NH)NH—C2-C8-alkenyl, —NHC(NH)NH—C2-C8-alkynyl, —NHC(NH)NH—C3-C12-cycloalkyl, —NHC(NH)NH-aryl, —NHC(NH)NH-heteroaryl, —NHC(NH)NH-heterocycloalkyl, —NHC(NH)—C1-C12-alkyl, —NHC(NH)—C2-C8-alkenyl, —NHC(NH)—C2-C8-alkynyl, —NHC(NH)—C3-C12-cycloalkyl, —NHC(NH)-aryl, —NHC(NH)-heteroaryl, —NHC(NH)-heterocycloalkyl, —C(NH)NH2, —C(NH)NH—C1-C12-alkyl, —C(NH)NH—C2-C8-alkenyl, —C(NH)NH—C2-C8-alkynyl, —C(NH)NH—C3-C12-cycloalkyl, —C(NH)NH-aryl, —C(NH)NH-heteroaryl, —C(NH)NH— heterocycloalkyl, —S(O)—C1-C12-alkyl, —S(O)—C2-C8-alkenyl, —S(O)—C2-C8-alkynyl, —S(O)—C3-C12-cycloalkyl, —S(O)-aryl, —S(O)-heteroaryl, —S(O)-heterocycloalkyl, —SO2NH2, —SO2NH—C1-C12-alkyl, —SO2NH—C2-C8-alkenyl, —SO2NH—C2-C8-alkynyl, —SO2—C1-C12-alkyl, —SO2—C2-C8-alkenyl, —SO2—C2-C8-alkynyl, —SO2—C3-C12-cycloalkyl, —SO2-aryl, —SO2-heteroaryl, —SO2-heterocycloalkyl, —SO2NH—C3-C12-cycloalkyl, —SO2NH-aryl, —SO2NH-heteroaryl, —SO2NH-heterocycloalkyl, —NHSO2—C1-C12-alkyl, —NHSO2—C2-C8-alkenyl, —NHSO2—C2-C8-alkynyl, —NHSO2—C3-C12-cycloalkyl, —NHSO2-aryl, —NHSO2-heteroaryl, —NHSO2-heterocycloalkyl, —CH2NH2, —CH2SO2CH3, -aryl, -arylalkyl, -heteroaryl, -heteroarylalkyl, -heterocycloalkyl, —C3-C12-cycloalkyl, polyalkoxyalkyl, polyalkoxy, -methoxymethoxy, -methoxyethoxy, —SH, —S—C1-C12-alkyl, —S—C2-C8-alkenyl, —S—C2-C8-alkynyl, —S—C3-C12-cycloalkyl, —S-aryl, —S-heteroaryl, —S— heterocycloalkyl, or methylthio-methyl. In certain embodiments, the substituents are independently selected from halo, preferably C1and F; C1-C4-alkyl, preferably methyl and ethyl; halo-C1-C4-alkyl, such as fluoromethyl, difluoromethyl, and trifluoromethyl; C2-C4-alkenyl; halo-C2-C4-alkenyl; C3-C6-cycloalkyl, such as cyclopropyl; C1-C4-alkoxy, such as methoxy and ethoxy; halo-C1-C4-alkoxy, such as fluoromethoxy, difluoromethoxy, and trifluoromethoxy; —CN; —OH; NH2; C1-C4-alkylamino; di(C1-C4-alkyl)amino; and NO2. It is understood that an aryl, heteroaryl, alkyl, alkenyl, alkynyl, cycloalkyl, or heterocycloalkyl in a substituent can be further substituted. In certain embodiments, a substituent in a substituted moiety is additionally optionally substituted with one or more groups, each group being independently selected from C1-C4-alkyl; —CF3, —OCH3, —OCF3, —F, —Cl, —Br, —I, —OH, —NO2, —CN, and —NH2. Preferably, a substituted alkyl group is substituted with one or more halogen atoms, more preferably one or more fluorine or chlorine atoms. The term “halo” or halogen” alone or as part of another substituent, as used herein, refers to a fluorine, chlorine, bromine, or iodine atom. The term “optionally substituted”, as used herein, means that the referenced group may be substituted or unsubstituted. In one embodiment, the referenced group is optionally substituted with zero substituents, i.e., the referenced group is unsubstituted. In another embodiment, the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from groups described herein. The term “hydrogen” includes hydrogen and deuterium. In addition, the recitation of an element includes all isotopes of that element so long as the resulting compound is pharmaceutically acceptable. In certain embodiments, the isotopes of an element are present at a particular position according to their natural abundance. In other embodiments, one or more isotopes of an element at a particular position are enriched beyond their natural abundance. The term “hydroxy activating group,” as used herein, refers to a labile chemical moiety which is known in the art to activate a hydroxyl group so that it will depart during synthetic procedures such as in a substitution or an elimination reaction. Examples of hydroxyl activating group include, but not limited to, mesylate, tosylate, triflate, p-nitrobenzoate, phosphonate and the like. The term “activated hydroxyl,” as used herein, refers to a hydroxy group activated with a hydroxyl activating group, as defined above, including, but not limited to mesylate, tosylate, triflate, p-nitrobenzoate, phosphonate groups. The term “hydroxy protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect a hydroxyl group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the hydroxy protecting group as described herein may be selectively removed. Hydroxy protecting groups as known in the art are described generally in P. G. M. Wuts,Greene's Protective Groups in Organic Synthesis,5th edition, John Wiley & Sons, Hoboken, NJ (2014). Examples of hydroxyl protecting groups include, but not limited to, benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, tert-butoxycarbonyl, isopropoxycarbonyl, diphenylmethoxycarbonyl, 2,2,2-trichloroethoxycarbonyl, allyloxycarbonyl, acetyl, formyl, chloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, benzoyl, methyl, t-butyl, 2,2,2-trichloroethyl, 2-trimethylsilyl ethyl, allyl, benzyl, triphenyl-methyl (trityl), methoxymethyl, methylthiomethyl, benzyloxymethyl, 2-(trimethylsilyl)-ethoxymethyl, methanesulfonyl, trimethylsilyl, triisopropylsilyl, and the like. The term “protected hydroxy,” as used herein, refers to a hydroxy group protected with a hydroxy protecting group, as defined above, including but not limited to, benzoyl, acetyl, trimethylsilyl, triethylsilyl, methoxymethyl groups, for example. The term “hydroxy prodrug group,” as used herein, refers to a promoiety group which is known in the art to change the physicochemical, and hence the biological properties of a parent drug in a transient manner by covering or masking the hydroxy group. After said synthetic procedure(s), the hydroxy prodrug group as described herein must be capable of reverting back to hydroxy group in vivo. Hydroxy prodrug groups as known in the art are described generally in Kenneth B. Sloan,Prodrugs, Topical and Ocular Drug Delivery, (Drugs and the Pharmaceutical Sciences; Volume 53), Marcel Dekker, Inc., New York (1992). The term “amino protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect an amino group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the amino protecting group as described herein may be selectively removed. Amino protecting groups as known in the art are described generally in P. G. M. Wuts,Greene's Protective Groups in Organic Synthesis,5th edition, John Wiley & Sons, Hoboken, NJ (2014). Examples of amino protecting groups include, but are not limited to, methoxycarbonyl, t-butoxycarbonyl, 12-fluorenyl-methoxycarbonyl, benzyloxycarbonyl, and the like. The term “protected amino,” as used herein, refers to an amino group protected with an amino protecting group as defined above. The term “leaving group” means a functional group or atom which can be displaced by another functional group or atom in a substitution reaction, such as a nucleophilic substitution reaction. By way of example, representative leaving groups include chloro, bromo and iodo groups; sulfonic ester groups, such as mesylate, tosylate, brosylate, nosylate and the like; and acyloxy groups, such as acetoxy, trifluoroacetoxy and the like. The term “aprotic solvent,” as used herein, refers to a solvent that is relatively inert to proton activity, i.e., not acting as a proton-donor. Examples include, but are not limited to, hydrocarbons, such as hexane and toluene, for example, halogenated hydrocarbons, such as, for example, methylene chloride, ethylene chloride, chloroform, and the like, heterocyclic compounds, such as, for example, tetrahydrofuran and N-methylpyrrolidinone, and ethers such as diethyl ether, bis-methoxymethyl ether. Such compounds are well known to those skilled in the art, and it will be obvious to those skilled in the art that individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of aprotic solvents may be found in organic chemistry textbooks or in specialized monographs, for example:Organic Solvents Physical Properties and Methods of Purification,4th ed., edited by John A. Riddick et al., Vol. II, in theTechniques of Chemistry Series, John Wiley & Sons, N Y, 1986. The term “protic solvent,” as used herein, refers to a solvent that tends to provide protons, such as an alcohol, for example, methanol, ethanol, propanol, isopropanol, butanol, t-butanol, and the like. Such solvents are well known to those skilled in the art, and it will be obvious to those skilled in the art that individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of protogenic solvents may be found in organic chemistry textbooks or in specialized monographs, for example:Organic Solvents Physical Properties and Methods of Purification,4th ed., edited by John A. Riddick et al., Vol. II, in theTechniques of Chemistry Series, John Wiley & Sons, N Y, 1986. Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term “stable,” as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject). The synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization. As can be appreciated by the skilled artisan, further methods of synthesizing the compounds of the Formula herein will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock,Comprehensive Organic Transformations,2ndEd. Wiley-VCH (1999); P. G. M. Wuts,Greene's Protective Groups in Organic Synthesis,5th edition, John Wiley & Sons, Hoboken, NJ (2014); L. Fieser and M. Fieser,Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed.,Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof. The term “subject,” as used herein, refers to an animal. Preferably, the animal is a mammal. More preferably, the mammal is a human. A subject also refers to, for example, a dog, cat, horse, cow, pig, guinea pig, fish, bird and the like. The compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and may include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion. The compounds described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-, or as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optical isomers may be prepared from their respective optically active precursors by the procedures described above, or by resolving the racemic mixtures. The resolution can be carried out in the presence of a resolving agent, by chromatography or by repeated crystallization or by some combination of these techniques which are known to those skilled in the art. Further details regarding resolutions can be found in Jacques, et al.,Enantiomers, Racemates, and Resolutions(John Wiley & Sons, 1981). When the compounds described herein contain olefinic double bonds, other unsaturation, or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers or cis- and trans-isomers. Likewise, all tautomeric forms are also intended to be included. Tautomers may be in cyclic or acyclic. The configuration of any carbon-carbon double bond appearing herein is selected for convenience only and is not intended to designate a particular configuration unless the text so states; thus a carbon-carbon double bond or carbon-heteroatom double bond depicted arbitrarily herein as trans may be cis, trans, or a mixture of the two in any proportion. Certain compounds of the present invention may also exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers. The present invention includes each conformational isomer of these compounds and mixtures thereof. As used herein, the term “pharmaceutically acceptable salt,” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 2-19 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable organic acid. Examples of pharmaceutically acceptable salts include, but are not limited to, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentane-propionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aryl sulfonate. As used herein, the term “pharmaceutically acceptable ester” refers to esters which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates. Pharmaceutical Compositions The pharmaceutical compositions of the present invention comprise a therapeutically effective amount of a compound of the present invention formulated together with one or more pharmaceutically acceptable carriers or excipients. As used herein, the term “pharmaceutically acceptable carrier or excipient” means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. Some examples of materials which can serve as pharmaceutically acceptable carriers are sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols such as propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator. The pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, preferably by oral administration or administration by injection. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intra-arterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectable. The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use. In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissues. Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulations, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention. The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Powders and sprays can contain, in addition to the compounds of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons. Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. For pulmonary delivery, a therapeutic composition of the invention is formulated and administered to the patient in solid or liquid particulate form by direct administration e.g., inhalation into the respiratory system. Solid or liquid particulate forms of the active compound prepared for practicing the present invention include particles of respirable size: that is, particles of a size sufficiently small to pass through the mouth and larynx upon inhalation and into the bronchi and alveoli of the lungs. Delivery of aerosolized therapeutics, particularly aerosolized antibiotics, is known in the art (see, for example U.S. Pat. No. 5,767,068 to Van Devanter et al., U.S. Pat. No. 5,508,269 to Smith et al., and WO 98/43650 by Montgomery, all of which are incorporated herein by reference). Antiviral Activity In certain embodiments, the present invention provides a method of treating or preventing a viral infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof. The viral infection is preferably a coronavirus infection. In certain embodiments, the coronavirus is SARS-CoV-1, SARS-CoV-2, or MERS-CoV. Preferably the coronavirus is SARS-CoV-2. A viral inhibitory amount or dose of the compounds of the present invention may range from about 0.01 mg/Kg to about 500 mg/Kg, alternatively from about 1 to about 50 mg/Kg. Inhibitory amounts or doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents. According to the methods of treatment of the present invention, viral infections are treated or prevented in a patient such as a human or another animal by administering to the patient a therapeutically effective amount of a compound of the invention, in such amounts and for such time as is necessary to achieve the desired result. By a “therapeutically effective amount” of a compound of the invention is meant an amount of the compound which confers a therapeutic effect on the treated subject, at a reasonable benefit/risk ratio applicable to any medical treatment. The therapeutic effect may be objective (i.e., measurable by some test or marker) or subjective (i.e., subject gives an indication of or feels an effect). A therapeutically effective amount of the compound described above may range, for example, from about 0.1 mg/Kg to about 500 mg/Kg, preferably from about 1 to about 50 mg/Kg. Effective doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or contemporaneously with the specific compound employed; and like factors well known in the medical arts. The total daily dose of the compounds of this invention administered to a human or other animal in single or in divided doses can be in amounts, for example, from 0.01 to 50 mg/kg body weight or more usually from 0.1 to 25 mg/kg body weight. Single dose compositions may contain such amounts or submultiples thereof to make up the daily dose. In general, treatment regimens according to the present invention comprise administration to a patient in need of such treatment from about 10 mg to about 1000 mg of the compound(s) of this invention per day in single or multiple doses. The compounds of the present invention described herein can, for example, be administered by injection, intravenously, intra-arterial, subdermally, intraperitoneally, intramuscularly, or subcutaneously; or orally, buccally, nasally, transmucosally, topically, in an ophthalmic preparation, or by inhalation, with a dosage ranging from about 0.1 to about 500 mg/kg of body weight, alternatively dosages between 1 mg and 1000 mg/dose, every 4 to 120 hours, or according to the requirements of the particular drug. The methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect. Typically, the pharmaceutical compositions of this invention will be administered from about 1 to about 6 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with pharmaceutically excipients or carriers to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5% to about 95% active compound (w/w). Alternatively, such preparations may contain from about 20% to about 80% active compound. Lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the disease, condition or symptoms, the patient's disposition to the disease, condition or symptoms, and the judgment of the treating physician. Upon improvement of a patient's condition, a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms. Combination and Alternation Therapy The compounds of the present invention may be used in combination with one or more antiviral therapeutic agents or anti-inflammatory agents useful in the prevention or treatment of viral diseases or associated pathophysiology. Thus, the compounds of the present invention and their salts, solvates, or other pharmaceutically acceptable derivatives thereof, may be employed alone or in combination with other antiviral or anti-inflammatory therapeutic agents. The compounds herein and pharmaceutically acceptable salts thereof may be used in combination with one or more other agents which may be useful in the prevention or treatment of respiratory disease, inflammatory disease, autoimmune disease, for example; anti-histamines, corticosteroids, (e.g., fluticasone propionate, fluticasone furoate, beclomethasone dipropionate, budesonide, ciclesonide, mometasone furoate, triamcinolone, flunisolide), NSAIDs, leukotriene modulators (e.g., montelukast, zafirlukast.pranlukast), tryptase inhibitors, IKK2 inhibitors, p38 inhibitors, Syk inhibitors, protease inhibitors such as elastase inhibitors, integrin antagonists (e.g., beta-2 integrin antagonists), adenosine A2a agonists, mediator release inhibitors such as sodium chromoglycate, 5-lipoxygenase inhibitors (zyflo), DP1 antagonists, DP2 antagonists, PI3K delta inhibitors, ITK inhibitors, LP (Iysophosphatidic) inhibitors or FLAP (5-lipoxygenase activating protein) inhibitors (e.g., sodium 3-(3-(tert-butylthio)-1-(4-(6-ethoxypyridin-3-yl)benzyl)-5-((5-ethylpyridin-2-yl)methoxy)-1H-indol-2-yl)-2,2-dimethylpropanoate), bronchodilators (e.g., muscarinic antagonists, beta-2 agonists), methotrexate, and similar agents; monoclonal antibody therapy such as anti-lgE, anti-TNF, anti-IL-5, anti-IL-6, anti-IL-12, anti-IL-1 and similar agents; cytokine receptor therapies e.g. etanercept and similar agents; antigen non-specific immunotherapies (e.g. interferon or other cytokines/chemokines, chemokine receptor modulators such as CCR3, CCR4 or CXCR2 antagonists, other cytokine/chemokine agonists or antagonists, TLR agonists and similar agents), suitable anti-infective agents including antibiotic agents, antifungal agents, antheimintic agents, antimalarial agents, antiprotozoal agents, antitubercuiosis agents, and antiviral agents, including those listed at https://www.drugs.com/drug-class/anti-infectives.html. In general, combination therapy is typically preferred over alternation therapy because it induces multiple simultaneous stresses on the virus. When the compositions of this invention comprise a combination of a compound of the Formula described herein and one or more additional therapeutic or prophylactic agents, both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen. The additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those agents may be part of a single dosage form, combined with a compound of this invention in a single composition. The “additional therapeutic or prophylactic agents” include but are not limited to, immune therapies (e.g. interferon), therapeutic vaccines, antifibrotic agents, anti-inflammatory agents such as corticosteroids or NSAIDs, bronchodilators such as beta-2 adrenergic agonists and xanthines (e.g. theophylline), mucolytic agents, anti-muscarinics, anti-leukotrienes, inhibitors of cell adhesion (e.g. ICAM antagonists), anti-oxidants (e.g. N-acetylcysteine), cytokine agonists, cytokine antagonists, lung surfactants and/or antimicrobial and anti-viral agents (e.g. ribavirin and amantidine). The compositions according to the invention may also be used in combination with gene replacement therapy. Although the invention has been described with respect to various preferred embodiments, it is not intended to be limited thereto, but rather those skilled in the art will recognize that variations and modifications may be made therein which are within the spirit of the invention and the scope of the appended claims. Abbreviations Abbreviations which may be used in the descriptions of the scheme and the examples that follow are: Ac for acetyl; AcOH for acetic acid; Boc2O for di-tert-butyl-dicarbonate; Boc for t-butoxycarbonyl; Bz for benzoyl; Bn for benzyl; t-BuOK for potassium tert-butoxide; Brine for sodium chloride solution in water; CDI for carbonyldiimidazole; DCM or CH2Cl2for dichloromethane; CH3for methyl; CH3CN for acetonitrile; Cs2CO3for cesium carbonate; CuCl for copper (I) chloride; CuI for copper (I) iodide; dba for dibenzylidene acetone; DBU for 1,8-diazabicyclo[5.4.0]-undec-7-ene; DEAD for diethylazodicarboxylate; DIAD for diisopropyl azodicarboxylate; DIPEA or (i-Pr)2EtN for N,N,-diisopropylethyl amine; DMP or Dess-Martin periodinane for 1,1,2-tris(acetyloxy)-1,2-dihydro-1,2-benziodoxol-3-(1H)-one; DMAP for 4-dimethylamino-pyridine; DME for 1,2-dimethoxyethane; DMF for N,N-dimethylformamide; DMSO for dimethyl sulfoxide; EtOAc for ethyl acetate; EtOH for ethanol; Et2O for diethyl ether; HATU for O-(7-azabenzotriazol-2-yl)-N,N,N′,N′,-tetramethyluronium Hexafluoro-phosphate; HCl for hydrogen chloride; K2CO3for potassium carbonate; n-BuLi for n-butyl lithium; DDQ for 2,3-dichloro-5,6-dicyano-1,4-benzoquinone; LDA for lithium diisopropylamide; LiTMP for lithium 2,2,6,6-tetramethyl-piperidinate; MeOH for methanol; Mg for magnesium; MOM for methoxymethyl; Ms for mesyl or —SO2—CH3; NaHMDS for sodium bis(trimethylsilyl)amide; NaCl for sodium chloride; NaH for sodium hydride; NaHCO3for sodium bicarbonate or sodium hydrogen carbonate; Na2CO3sodium carbonate; NaOH for sodium hydroxide; Na2SO4for sodium sulfate; NaHSO3for sodium bisulfite or sodium hydrogen sulfite; Na2S2O3for sodium thiosulfate; NH2NH2for hydrazine; NH4C1for ammonium chloride; Ni for nickel; OH for hydroxyl; OsO4for osmium tetroxide; OTf for triflate; PPA for polyphosphoric acid; PTSA for p-toluenesulfonic acid; PPTS for pyridinium p-toluenesulfonate; TBAF for tetrabutylammonium fluoride; TEA or Et3N for triethylamine; TES for triethylsilyl; TESCl for triethylsilyl chloride; TESOTf for triethylsilyl trifluoromethanesulfonate; TFA for trifluoroacetic acid; THE for tetrahydrofuran; TMEDA for N,N,N′,N′-tetramethylethylene-diamine; TPP or PPh3for triphenyl-phosphine; Tos or Ts for tosyl or —SO2—C6H4CH3; Ts2O for tolylsulfonic anhydride or tosyl-anhydride; TsOH for p-tolylsulfonic acid; Pd for palladium; Ph for phenyl; Pd2(dba)3for tris(diben-zylideneacetone) dipalladium (0); Pd(PPh3)4for tetrakis(triphenylphosphine)-palladium (0); PdCl2(PPh3)2for trans-dichlorobis-(triphenylphosphine)palladium (II); Pt for platinum; Rh for rhodium; rt for room temperature; Ru for ruthenium; TBS for tert-butyl dimethylsilyl; TMS for trimethylsilyl; and TMSCl for trimethylsilyl chloride. Synthetic Methods Scheme 1 illustrates a general method to prepare the compound of formula (Ia) from the amino ester compound (X-1), wherein B is as previously defined and PG1is C1-C4 alkyl or Bn. In Step 1, treatment of amine (X-1) with formaldehyde affords the cyclized amine (X-2), which is converted in Step 2 to (X-3) using appropriate protecting group PG2(e.g. Boc). In Step 3, treatment of (X-3) with NBS in solvents containing AcOH at low temperature provides the rearranged spiro proline derivative (X-4). Examples of this sequence of transformation has been reported in literature (Pellegrini C. et al. “Synthesis of the Oxindole Alkaloid (−)-Horsfiline” Tetrahedron Asymmetry, 1994, vol. 5, No. 10, pp 1979-1992; Efremov, I. V. et al. “Discovery and Optimization of a Novel Spiropyrrolidine Inhibitor of β-Secretase (BACE1) through Fragment-Based Drug Design” Journal of Medicinal Chemistry, 2012, 55, 9069-9088). In Step 4, the ester compound of formula (X-4), wherein B, PG1and PG2are previously defined, is reduced to the alcohol compound (X-5) employing reducing reagents such as, but not limited to, LiBH4, NaBH4, or DIBAL-H. In Step 5, the alcohol compound of formula (X-5) is oxidized to the aldehyde compound of formula (X-6) using agents such as, but not limited to, Dess-Martin periodinane, IBX, or pyridine-SO3. In Step 6, the aldehyde compound of formula (X-6) is reacted with a phosphonate ester such as a compound of formula (X-7), to produce the alkyne compound of formula (X-8). In Step 7, the protecting group of the compound of formula (X-8) is removed under acidic, basic, or reductive conditions to produce amine compound of formula (X-9). In Step 8, an amide bond is formed between the compound of formula (X-9) and an acid compound such as a compound of formula (X-10) using a reagent such as, but not limited to, HATU or DMAP, upon which, followed by a deprotection step under acidic, basic, or reductive conditions, will produce a compound of formula (X-11). In Step 9, an acidic compound of formula (X-12) is reacted with the amine of the compound of formula (X-11) in an amide bond forming reaction using a reagent such as, but not limited to, HATU or DMAP to produce the compound of formula (X-13). EXAMPLES The compounds and processes of the present invention will be better understood in connection with the following examples, which are intended as an illustration only and not limiting of the scope of the invention. Starting materials were either available from a commercial vendor or produced by methods well known to those skilled in the art. General Conditions: Mass spectra were run on LC-MS systems using electrospray ionization. These were Agilent 1290 Infinity II systems with an Agilent 6120 Quadrupole detector. Spectra were obtained using a ZORBAX Eclipse XDB-C18 column (4.6×30 mm, 1.8 micron). Spectra were obtained at 298K using a mobile phase of 0.1% formic acid in water (A) and 0.1% formic acid in acetonitrile (B). Spectra were obtained with the following solvent gradient: 5% (B) from 0-1.5 min, 5-95% (B) from 1.5-4.5 min, and 95% (B) from 4.5-6 min. The solvent flowrate was 1.2 mL/min. Compounds were detected at 210 nm and 254 nm wavelengths. [M+H]+refers to mono-isotopic molecular weights. NMR spectra were run on a Bruker 400 MHz spectrometer. Spectra were measured at 298K and referenced using the solvent peak. Chemical shifts for1H NMR are reported in parts per million (ppm). Compounds were purified via reverse-phase high-performance liquid chromatography (RPHPLC) using a Gilson GX-281 automated liquid handling system. Compounds were purified on a Phenomenex Kinetex EVO C18 column (250×21.2 mm, 5 micron), unless otherwise specified. Compounds were purified at 298K using a mobile phase of water (A) and acetonitrile (B) using gradient elution between 0% and 100% (B), unless otherwise specified. The solvent flowrate was 20 mL/min and compounds were detected at 254 nm wavelength. Alternatively, compounds were purified via normal-phase liquid chromatography (NPLC) using a Teledyne ISCO Combiflash purification system. Compounds were purified on a REDISEP silica gel cartridge. Compounds were purified at 298K and detected at 254 nm wavelength. Ex. 1: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-5-(methylsulfonyl)-1H-indole-2-carboxamide Step 1 methyl (S)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indole-3-carboxylate hydrochloride (500 mg, 1.875 mmol) was dissolved in CH2Cl2(10 ml). Triethylamine (523 μl, 3.75 mmol) and a 2.0 M solution of di-tert-butyl dicarbonate in DCM (1031 μl, 2.062 mmol) was added. The mixture was stirred at rt for 3 h, quenched with sat. NaHCO3, and extracted with DCM. The organic layer was washed with brine, dried over MgSO4, and concentrated in vacuo. Purification of the residue on silica gel with 0-30% EtOAc/cyclohexane provided compound 1-2 (578 mg, 1.749 mmol, 93% yield). Step 2 Compound 1-2 (578 mg) was dissolved in THE (15 ml), AcOH (10 ml), and water (10 ml). The solution was cooled to −15° C. A solution of NBS (328 mg, 1.843 mmol) in THE (5 mL) was added dropwise. The mixture was slowly warmed to 5° C. over 1 h. The reaction was quenched with Na2SO3and sat. NaHCO3, and extracted with DCM (2×). The organic layer was washed with brine, dried with MgSO4, and concentrated in vacuo. Purification of the residue on silica gel with 0-50% EtOAc/cyclohexane provided compound 1-3 (328 mg, 0.947 mmol, 53.9% yield). Step 3 To a solution of compound 1-3 (2.5 g, 7.22 mmol) in THE (24.06 mL) was added drowpise a solution of 2M LiBH4in THE (10.83 mL, 21.65 mmol). The mixture was stirred at rt for 2 hrs and the majority of THE was removed in vacuo. The reaction was quenched carefully with 1N HCl to pH=5-6 (˜22 mL) and extracted with EtOAc (3×40 mL). The combined organic layers were washed with sat NaHCO3, brine, dried and concentrated. Purification of the residue on silica gel with 0-50% EtOAc/Cyclohexane provided compound 1-4 (1.54 g, 67% yield). Step 4 In a 250 mL round-bottomed flask equipped with a stir bar, compound 1-4 (2.50 g, 7.85 mmol, 1.0 equiv) was dissolved in methylene chloride (52 mL, 0.15M) under a nitrogen atmosphere. The resulting solution was cooled using an ice and water bath. Dess-Martin periodinane (5.00 g, 11.8 mmol, 1.5 equiv) was then added in a single portion. The resulting mixture was allowed to warm to room temperature slowly and was stirred for 16 h. Upon completion, as judged by LCMS analysis of the reaction mixture, the reaction was filtered through a pad of celite using methylene chloride to rinse. Following concentration of the filtrate, the residue was purified directly by silica gel column chromatography (gradient elution, 0 to 100% ethyl acetate/cyclohexane) to afford compound 1-5 (2.31 g, 7.31 mmol, 93% yield). Step 5 In a 500 mL round-bottomed flask equipped with a stir bar, compound 1-5 (2.21 g, 6.99 mmol, 1.0 equiv) was dissolved in methanol (116 mL, 0.6M) at room temperature under a nitrogen atmosphere. The resulting solution was cooled using an ice and water bath, and dimethyl (1-diazo-2-oxopropyl)phosphonate (19.5 mL, 8.38 mmol, 10 wt % in acetonitrile, 1.2 equiv) was added, followed by potassium carbonate (1.93 g, 14.0 mmol, 2.0 equiv). The resulting mixture was allowed to warm to room temperature slowly and was stirred for 16 h. Upon completion, as judged by LCMS analysis of the reaction mixture, the solvent was removed under vacuum, and the residue was partitioned between ethyl acetate (125 mL) and water (75 mL). The aqueous phase was extracted with ethyl acetate (50 mL), and the combined organic layers were dried over magnesium sulfate. After concentration, the crude residue was purified by silica gel column chromatography (gradient elution, 0 to 50% ethyl acetate/cyclohexane) to afford compound 1-6 (2.02 g, 6.99 mmol, 93% yield). Step 6 To a 250 mL round-bottomed flask containing compound 1-6 (2.02 g, 6.48 mmol, 1.0 equiv) under a nitrogen atmosphere was charged 4M HCl in dioxane (32 mL, 20 equiv) at room temperature. After stirring for 2 h, LCMS analysis of the reaction mixture indicated complete consumption of the starting material, and the reaction mixture was concentrated under vacuum to afford compound 1-7 (1.61 g, 6.48 mmol) which was used directly in the next step without purification. Step 7 In a 250 mL round-bottomed flask equipped with a stir bar, compound 1-7 (1.61 g, 6.48 mmol, 1.0 equiv) and (S)-2-((tert-butoxycarbonyl)amino)-4-fluoro-4-methylpentanoic acid (1.62 g, 6.48 mmol, 1.0 equiv) were combined in a mixed solvent system of methylene chloride (20.7 mL) and N,N-dimethylformamide (5.2 mL) under a nitrogen atmosphere. The resulting mixture was cooled in an ice and water bath, and N-methylmorpholine (2.28 mL, 20.7 mmol, 3.2 equiv) was added, followed by HATU (2.46 g, 6.48 mmol, 1.0 equiv). The mixture was allowed to warm to room temperature. After 16 h, LCMS analysis indicated complete consumption of the starting material, and the reaction mixture was diluted with methylene chloride (125 mL). The organic phase was washed once with 1.2M HCl (40 mL) and once with saturated aqueous sodium chloride (40 mL) and then dried over magnesium sulfate. Upon concentration, the crude residue was purified by reversed-phase HPLC (MeCN/water, 0.1% formic acid) affording compound 1-8 (1.29 g, 2.91 mmol, 45% yield). Step 8 In a 40 mL vial equipped with a stir bar, compound 1-8 (356 mg, 0.80 mmol, 1.0 equiv) was treated with 4M HCl in dioxane (4.0 mL, 20 equiv) under an air atmosphere. After stirring for 2 h in the capped vial, LCMS analysis indicated full consumption of the starting material. The solvent was subsequently removed under vacuum to afford compound 1-9 (305 mg, 0.80 mmol) which was used in the next step without purification. Step 9 In a 4 mL vial equipped with a stir bar, compound 1-9 (35.0 mg, 0.09 mmol, 1.0 equiv) and 5-(methylsulfonyl)-1H-indole-2-carboxylic acid (22.0 mg, 0.09 mmol, 1.0 equiv) were combined in a mixed solvent system of methylene chloride (0.5 mL) and N,N-dimethylformamide (0.1 mL). The resulting solution was cooled in an ice and water bath and N-methylmorpholine (32 μL, 0.30 mmol, 3.2 equiv) was added, followed by HATU (35.0 mg, 0.09 mmol, 1.0 equiv). The vial was capped under an air atmosphere, and the reaction mixture was allowed to slowly warm to room temperature. After 19 h, LCMS analysis of the reaction mixture indicated complete consumption of the starting material. The reaction was quenched with formic acid (100 μL) and concentrated. Purification of the crude residue by reversed-phase HPLC (MeCN/water, 0.1% formic acid) afforded Ex. 1 (29.9 mg, 0.05 mmol, 57%). ESI MS m/z=563.1 [M−H]−. Ex. 2: Synthesis of 4-chloro-N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 4-chloro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 10.90 (s, 1H), 9.72 (s, 1H), 8.26 (d, J=8.8 Hz, 1H), 7.49-7.52 (m, 1H), 7.36-7.37 (m, 1H), 7.22-7.26 (m, 1H), 7.15-7.17 (m, 1H), 7.04-7.08 (m, 1H), 7.0 (d, J=7.5 Hz, 1H), 6.93 (d, J=7.7, 1H), 6.74 (td, J=7.6, 1.1 Hz, 1H), 5.05-5.20 (m, 2H), 4.35 (d, J=10.5, 1H), 3.98 (d, J=10.5 Hz, 1H), 2.93 (d, J=2.1 Hz, 1H), 2.53-2.64 (m, 2H), 2.21-2.42 (m, 2H), 1.50 (s, 3H), 1.45 (s, 3H); ESI MS m/z=519.0 [M−H]−. Ex. 3: Synthesis of 5-chloro-N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 5-chloro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 10.73 (s, 1H), 9.68 (s, 1H), 8.04 (d, J=8.7 Hz, 1H), 7.70 (d, J=2.0 Hz, 1H), 7.56 (d, J=8.7 Hz, 1H), 7.20-7.28 (m, 2H), 7.07 (td, J=7.7, 1.2 Hz, 1H), 6.94-6.99 (m, 2H), 6.74 (td, J=7.5, 1.1 Hz, 1H), 5.16-5.03 (m, 2H), 4.34 (d, J=10.4 Hz, 1H), 3.95 (d, J=10.4 Hz, 1H), 2.92 (d, J=2.1 Hz, 1H), 2.53-2.62 (m, 2H), 2.17-2.38 (m, 2H), 1.49 (s, 3H), 1.43 (s, 3H); ESI MS m/z=519.1 [M−H]−. Ex. 4: Synthesis of 6-chloro-N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 6-chloro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 10.72 (s, 1H), 9.68 (s, 1H), 8.00 (d, J=8.6 Hz, 1H), 7.68 (d, J=8.5 Hz, 1H), 7.58-7.59 (m, 1H), 7.27-7.29 (m, 1H), 7.12 (dd, J=8.6, 1.9 Hz, 1H), 7.07 (td, J=7.7, 1.3 Hz, 1H), 6.94-6.98 (m, 2H), 6.73 (td, J=7.5, 1.1 Hz, 1H), 5.01-5.17 (m, 2H), 4.34 (d, J=10.4 Hz, 1H), 3.94 (d, J=10.4 Hz, 1H), 2.91 (d, J=2.1 Hz, 1H), 2.52-2.61 (m, 2H), 2.15-2.38 (m, 2H), 1.49 (s, 3H), 1.43 (s, 3H); ESI MS m/z=519.2 [M−H]−. Ex. 5: Synthesis of 7-chloro-N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 7-chloro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 10.29 (s, 1H), 9.69 (s, 1H), 8.04 (d, J=8.7 Hz, 1H), 7.66 (d, J=8.1 Hz, 1H), 7.33-7.35 (m, 1H), 7.30-7.31 (m, 1H), 7.12-7.16 (m, 1H), 7.06 (td, J=7.7, 1.3 Hz, 1H), 6.95-6.99 (m, 2H), 6.74 (td, J=7.5, 1.1 Hz, 1H), 5.17-5.05 (m, 2H), 4.34 (d, J=10.4 Hz, 1H), 3.95 (d, J=10.4 Hz, 1H), 2.92 (d, J=2.1 Hz, 1H), 2.53-2.62 (m, 2H), 2.14-2.40 (m, 2H), 1.49 (s, 3H), 1.44 (s, 3H); ESI MS m/z=519.1 [M−H]−. Ex. 6: Synthesis of 4,6-dichloro-N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 4,6-dichloro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 10.98 (s, 1H), 9.69 (s, 1H), 8.32 (d, J=8.6 Hz, 1H), 7.53 (m, 1H), 7.36-7.37 (m, 1H), 7.21 (d, J=1.6 Hz, 1H), 7.07 (td, J=7.7, 1.2 Hz, 1H), 7.03-6.91 (m, 2H), 6.74 (td, J=7.6, 1.1 Hz, 1H), 5.18-5.05 (m, 2H), 4.33 (d, J=10.5 Hz, 1H), 3.97 (d, J=10.5 Hz, 1H), 2.93 (d, J=2.1 Hz, 1H), 2.54-2.63 (m, 2H), 2.21-2.40 (m, 2H), 1.50 (s, 3H), 1.44 (s, 3H); ESI MS m/z=553.0 [M−H]−. Ex. 7: Synthesis of 6,7-dichloro-N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 6,7-dichloro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9. ESI MS m/z=555.4 [M+H]+. Ex. 8: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-7-fluoro-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 7-fluoro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 10.82 (s, 1H), 9.67 (s, 1H), 7.97 (d, J=8.6 Hz, 1H), 7.50 (d, J=7.8 Hz, 1H), 7.31 (dd, J=3.2, 2.2 Hz, 1H), 6.94-7.11 (m, 5H), 6.77 (td, J=7.5, 1.1 Hz, 1H), 5.20-5.02 (m, 2H), 4.32 (d, J=10.5 Hz, 1H), 3.95 (d, J=10.4 Hz, 1H), 2.91 (d, J=2.1 Hz, 1H), 2.52-2.62 (m, 2H), 2.15-2.39 (m, 2H), 1.49 (s, 3H), 1.44 (s, 3H); ESI MS m/z=505.2 [M+H]+. Ex. 9: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-6-fluoro-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 6-fluoro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9. ESI MS m/z=503.2 [M−H]−. Ex. 10: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-4,6-difluoro-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 4,6-difluoro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 10.92 (s, 1H), 9.68 (s, 1H), 8.09 (d, J=8.7 Hz, 1H), 7.35 (m, 1H), 7.12 (dd, J=9.4, 1.6 Hz, 1H), 7.07 (td, J=7.7, 1.2 Hz, 1H), 6.94-6.99 (m, 2H), 6.68-6.82 (m, 2H), 5.05-5.14 (m, 2H), 4.34 (d, J=10.4 Hz, 1H), 3.95 (d, J=10.4 Hz, 1H), 2.92 (d, J=2.1 Hz, 1H), 2.53-2.62 (m, 2H), 2.16-2.39 (m, 2H), 1.49 (s, 3H), 1.43 (s, 3H); ESI MS m/z=521.1 [M−H]−. Ex. 11: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-4,7-difluoro-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 4,7-difluoro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9. ESI MS m/z=521.2 [M−H]−. Ex. 12: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-5,6,7-trifluoro-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 5,6,7-trifluoro-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9. ESI MS m/z=539.3 [M−H]−. Ex. 13: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-7-(trifluoromethyl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 7-(trifluoromethyl)-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 9.97 (s, 1H), 9.75 (s, 1H), 8.34 (d, J=8.5 Hz, 1H), 7.99 (d, J=8.2 Hz, 1H), 7.63-7.65 (m, 1H), 7.37 (d, J=2.1 Hz, 1H), 7.29-7.34 (m, 1H), 6.96-7.07 (m, 3H), 6.75 (td, J=7.5, 1.1 Hz, 1H), 5.04-5.23 (m, 2H), 4.38 (d, J=10.4 Hz, 1H), 3.98 (d, J=10.5 Hz, 1H), 2.95 (d, J=2.1 Hz, 1H), 2.55-2.65 (m, 2H), 2.19-2.41 (m, 2H), 1.50 (s, 3H), 1.44 (s, 3H); ESI MS m/z=553.3 [M−H]−. Ex. 14: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-5-(trifluoromethyl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 5-(trifluoromethyl)-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9. ESI MS m/z=553.2 [M−H]−. Ex 15. Synthesis of 7-cyano-N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-1H-indole-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 7-cyano-1H-indole-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9. ESI MS m/z=510.0 [M−H]−. Ex. 16: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-1H-pyrrolo[2,3-b]pyridine-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 1H-pyrrolo[2,3-b]pyridine-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 11.14 (br s, 1H), 9.66 (s, 1H), 8.43 (dd, J=4.7, 1.6 Hz, 1H), 8.14 (dd, J=7.9, 1.7 Hz, 1H), 8.03 (d, J=8.6 Hz, 1H), 7.26 (s, 1H), 7.19 (dd, J=7.9, 4.7 Hz, 1H), 6.99-7.10 (m, 2H), 6.89-6.96 (m, 1H), 6.78 (td, J=7.5, 1.1 Hz, 1H), 5.02-5.22 (m, 2H), 4.36 (d, J=10.4 Hz, 1H), 3.96 (d, J=10.5 Hz, 1H), 2.92 (d, J=2.1 Hz, 1H), 2.53-2.62 (m, 2H), 2.16-2.40 (m, 2H), 1.50 (s, 3H), 1.45 (s, 3H); ESI MS m/z=488.3 [M+H]+. Ex. 17: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-7-fluorobenzofuran-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 7-fluorobenzofuran-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 9.68 (s, 1H), 8.23 (d, J=8.5 Hz, 1H), 7.71-7.55 (m, 1H), 7.48 (d, J=2.9 Hz, 1H), 7.43-7.29 (m, 2H), 7.01-7.07 (m, 2H), 6.94 (d, J=7.7 Hz, 1H), 6.80 (td, J=7.6, 1.1 Hz, 1H), 5.06-5.15 (m, 2H), 4.28 (d, J=10.5 Hz, 1H), 3.95 (d, J=10.5 Hz, 1H), 2.92 (d, J=2.1 Hz, 1H), 2.58 (d, J=8.7 Hz, 2H), 2.22-2.44 (m, 2H), 1.51 (s, 3H), 1.45 (s, 3H); ESI MS m/z=506.2 [M+H]+. Ex. 18: N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-5-fluorobenzofuran-2-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 5-fluorobenzofuran-2-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 9.68 (s, 1H), 8.05 (d, J=8.5 Hz, 1H), 7.65-7.68 (m, 1H), 7.51-7.51 (m, 1H), 7.41 (d, J=0.9 Hz, 1H), 7.32 (td, J=9.2, 2.7 Hz, 1H), 7.08 (td, J=7.7, 1.2 Hz, 1H), 6.95-7.02 (m, 2H), 6.80 (td, J=7.6, 1.1 Hz, 1H), 5.06-5.13 (m, 2H), 4.28 (d, J=10.5 Hz, 1H), 3.95 (d, J=10.5 Hz, 1H), 2.92 (d, J=2.1 Hz, 1H), 2.58 (d, J=8.8 Hz, 2H), 2.23-2.43 (m, 2H), 1.50 (s, 3H), 1.45 (s, 3H); ESI MS m/z=506.2 [M+H]+. Ex. 19: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-8-fluoroquinoline-6-carboxamide The title compound was prepared according to the procedure for Ex. 1, except that 8-fluoroquinoline-6-carboxylic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 9.71 (s, 1H), 9.07 (dd, J=4.2, 1.7 Hz, 1H), 8.50 (dt, J=8.4, 1.6 Hz, 1H), 8.22-8.29 (m, 2H), 7.78 (dd, J=11.5, 1.8 Hz, 1H), 7.73 (dd, J=8.4, 4.2 Hz, 1H), 7.10 (td, J=7.7, 1.2 Hz, 1H), 6.94-7.03 (m, 2H), 6.81 (td, J=7.6, 1.1 Hz, 1H), 5.07-5.20 (m, 2H), 4.41 (d, J=10.4 Hz, 1H), 3.97 (d, J=10.5 Hz, 1H), 2.93 (d, J=2.1 Hz, 1H), 2.55-2.63 (m, 2H), 2.23-2.43 (m, 2H), 1.51 (d, J=2.8 Hz, 3H), 1.46 (d, J=2.8 Hz, 3H); ESI MS m/z=517.3 [M+H]+. Ex. 20: Synthesis of N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)-3-(trifluoromethoxy)benzamide The title compound was prepared according to the procedure for Ex. 1, except that 3-(trifluoromethoxy)benzoic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9.1HNMR (acetone-d6, 400 MHz, PPM): δ 9.69 (s, 1H), 8.09 (d, J=8.5 Hz, 1H), 7.86 (dt, J=7.7, 1.3 Hz, 1H), 7.70-7.75 (m, 1H), 7.62 (t, J=8.0 Hz, 1H), 7.52-7.55 (m, 1H), 7.16 (td, J=7.7, 1.3 Hz, 1H), 6.94-7.04 (m, 2H), 6.82 (td, J=7.6, 1.1 Hz, 1H). 5.04-5.13 (m, 2H), 4.37 (d, J=10.5 Hz, 1H), 3.94 (d, J=10.4 Hz, 1H), 2.91 (d, J=2.1 Hz, 1H), 2.53-2.62 (m, 2H), 2.18-2.38 (m, 2H), 1.49 (d, J=1.6 Hz, 3H), 1.44 (d, J=1.7 Hz, 3H); ESI MS m/z=530.2 [M−H]−. Ex. 21: Synthesis of 2,6-dichloro-N—((S)-1-((3R,5′S)-5′-ethynyl-2-oxospiro[indoline-3,3′-pyrrolidin]-1′-yl)-4-fluoro-4-methyl-1-oxopentan-2-yl)benzamide The title compound was prepared according to the procedure for Ex. 1, except that 2,6-dichlorobenzoic acid was used in place of 5-(methylsulfonyl)-1H-indole-2-carboxylic acid in Step 9. ESI MS m/z=518.2 [M+H]+. Biological Activity SARS-CoV-2 3C-like (3CL) protease fluorescence assay (FRET): Recombinant SARS-CoV-2 3CL-protease was expressed and purified. TAMRA-SITSAVLQSGFRKMK-Dabcyl-OH peptide 3CLpro substrate was synthesized. Black, low volume, round-bottom, 384 well microplates were used. In a typical assay, 0.85 μL of test compound was dissolved in DMSO then incubated with SARS-CoV-2 3CL-protease (10 nM) in 10 μL assay buffer (50 mM HEPES [pH 7.5], 1 mM DTT, 0.01% BSA, 0.01% Triton-X 100) for 30 min at RT. Next, 10 μL of 3CL-protease substrate (40 μM) in assay buffer was added and the assays were monitored continuously for 1 h in an Envision multimode plate reader operating in fluorescence kinetics mode with excitation at 540 nm and emission at 580 nm at RT. No compound (DMSO only) and no enzyme controls were routinely included in each plate. All experiments were run in duplicate. Data Analysis: SARS-CoV-2 3CL-protease enzyme activity was measured as initial velocity of the linear phase (RFU/s) and normalized to controlled samples DMSO (100% activity) and no enzyme (0% activity) to determine percent residual activity at various concentrations of test compounds (0-10 μM). Data were fitted to normalized activity (variable slope) versus concentration fit in GraphPad Prism 7 to determine IC50. All experiments were run in duplicate, and IC50ranges are reported as follows: A<0.1 μM; B 0.1-1 μM; C>1 μM. TABLE 1Summary of ActivitiesFRETFRETCompoundIC50CompoundIC501B2B3B4A5A6B7A8A9A10B11A12A13A14B15A16B17C18C19C20B21C All references cited herein, whether in print, electronic, computer readable storage media or other form, are expressly incorporated by reference in their entirety, including but not limited to, abstracts, articles, journals, publications, texts, treatises, internet web sites, databases, patents, and patent publications. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the chemical structures, substituents, derivatives, formulations and/or methods of the invention may be made without departing from the spirit of the invention and the scope of the appended claims. Although the invention has been described with respect to various preferred embodiments, it is not intended to be limited thereto, but rather those skilled in the art will recognize that variations and modifications may be made therein which are within the spirit of the invention and the scope of the appended claims.
87,346
11858946
DETAILED DESCRIPTION OF THE INVENTION Overview The canonical view about the importance of highly polar molecules with a molecular weight less than 600 Da for Gram-negative activity has not led to general strategies to convert Gram-positive-only compounds into broad-spectrum antibiotics. The seminal observation over 50 years ago that derivatizing penicillin G into ampicillin results in broad-spectrum activity has not been generalizable, and important classes of experimental therapeutics and FDA-approved antibiotics have coverage only against Gram-positive organisms despite intensive derivatization efforts. A systematic analysis of the accumulation of an unbiased and structurally diverse set of small molecules in Gram-negative bacteria has not been previously reported. According to the present disclosure, a diverse set of 100 compounds was assembled, and their capacity to accumulate inE. coli, and by extension other Gram-negative bacteria, was quantified. As there are many variables affecting small molecule accumulation (e.g., multiple porins, efflux pumps, and varying lipopolysaccharides of the cellular envelope), model systems were not utilized, and instead compounds were assessed in accumulation assays using whole cells. From these experiments, and from follow-on structure-activity relationship (SAR) studies and computational analyses, predictive guidelines of small molecule accumulation in Gram-negative bacteria have been developed. Described herein are novel compounds and methods of use thereof as antibiotics. In some embodiments, the compounds disclosed herein comprise an ionizable nitrogen atom. In some embodiments, the compounds disclosed herein comprise a terminal —N(R5)mmoiety, wherein R5is independently selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl, and m is is 2 or 3. In some embodiments, —N(R5)mis —NH2, —NHMe, —NHEt, —NMe2, —NEt2, or —NMe(Et); or a protonated form (quaternary amine) thereof. In some embodiments, —N(R5)mis —NH3+, —NH2Me+, or —NHMe2+. In some embodiments, the compounds disclosed herein comprise a terminal —NH2, a primary amine. In some embodiments, the compounds disclosed herein comprise a quaternary amine. In some embodiments, the compounds disclosed herein comprise a terminal —NH3+. In some embodiments, the compounds disclosed herein have at least one hydrophobic region. In some embodiments, the compounds disclosed herein are amphiphilic. In some embodiments, the compounds disclosed herein have at least one hydrophobic region and at least one hydrophilic region. In some embodiments, the compounds disclosed herein have an amphiphilic moment greater than 1, greater than 2, greater than 3, greater than 4, greater than 5, greater than 6, greater than 7, greater than 8, greater than 9, or greater than 10. In some embodiments, the compounds disclosed herein have an amphiphilic moment between 1 and 20. In some embodiments, the amphiphilic moment is between about 2 and about 10 or between about 2 and about 7. In some embodiments, the amphiphilic moment is selected from the group consisting of about 2.2, about 2.3, about 2.4, about 2.5, about 2.6, about 2.7, about 2.8, about 2.9, about 3.0, about 3.1, about 3.2, about 3.3, about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4.0, about 4.1, about 4.2, about 4.3, about 4.4, about 4.5, about 4.6, about 4.7, about 4.8, about 4.9, about 5.0, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, about 6.0, about 6.1, about 6.2, about 6.3, about 6.4, about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, about 8.0, about 8.1, about 8.2, about 8.3, about 8.4, about 8.5, about 8.6, about 8.7, about 8.8, about 8.9, and about 9.0. In some embodiments, the compounds disclosed herein are rigid. In some embodiments, the compounds disclosed herein are less flexible. In some embodiments, the compounds disclosed herein have 15 or fewer rotatable bonds (RBs). In some embodiments, the compounds disclosed herein have 12 or fewer rotatable bonds, 11 or fewer rotatable bonds, 10 or fewer rotatable bonds, 9 or fewer rotatable bonds, 8 or fewer rotatable bonds, 7 or fewer rotatable bonds, 6 or fewer rotatable bonds, 5 or fewer rotatable bonds, 4 or fewer rotatable bonds, 3 or fewer rotatable bonds, 2 or fewer rotatable bonds, or 1 or fewer rotatable bonds. In some embodiments, the number of rotatable bonds is selected from the group consisting of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12. In some embodiments, the number of rotatable bonds is selected from the group consisting of 0, 1, 2, 3, 4, and 5. In some embodiments, the compounds disclosed herein have RB of 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less. In some embodiments, the compounds disclosed herein have low 3-dimensionality. In some embodiments, the compounds disclosed herein comprise a moiety with a planar feature, such as a phenyl or heteroaromatic ring. In some embodiments, the compounds disclosed herein have low globularity. In some embodiments, the compounds disclosed herein have a globularity (Glob) less than 0.8, less than 0.75, less than 0.7, less than 0.65, less than 0.6, less than 0.55, less than 0.5, less than 0.45, less than 0.4, less than 0.3, less than 0.25, less than 0.2, less than 0.15, or less than 0.1. In some embodiments, the compounds disclosed herein have Glob of 0.4 or less, 0.3 or less, 0.2 or less, or 0.1 or less. In some embodiments, the compounds disclosed herein have Glob of 0.42 or less, 0.4 or less, 0.38 or less, 0.36 or less, 0.34 or less, 0.32 or less, 0.3 or less, 0.28 or less, 0.26 or less, 0.24 or less, 0.22 or less, 0.20 or less, 0.18 or less, 0.16 or less, 0.14 or less, 0.12 or less, 0.1 or less, 0.08 or less, 0.06 or less, 0.04 or less, or 0.02 or less. In some embodiments, the compounds disclosed herein have a globularity between 0 and 0.7. In some embodiments, the globularity is between about 0 and about 0.6, between about 0 and about 0.5, between about 0 and about 0.4, or between about 0 and about 0.3. In some embodiments, the globularity is selected from the group consisting of about 0.01, about 0.02, about 0.03, about 0.04, about 0.05, about 0.06, about 0.07, about 0.08, about 0.09, about 0.10, about 0.11, about 0.12, about 0.13, about 0.14, about 0.15, about 0.16, about 0.17, about 0.18, about 0.19, about 0.20, about 0.21, about 0.22, about 0.23, about 0.24, about 0.25, about 0.26, about 0.27, about 0.28, about 0.29, about 0.30, about 0.31, about 0.32, about 0.33, about 0.34, and about 0.35. In some embodiments, the compounds disclosed herein have a planar best fit (PBF) between about 0.2 and about 2, between about 0.4 and about 1.8, between about 0.5 and about 1.7, between about 0.6 and about 1.6, between about 0.8 and about 1.5, or between about 0.9 and about 1.4. In some embodiments, the PBF is selected from the group consisting of about 0.45, about 0.5, about 0.55, about 0.6, about 0.65, about 0.7, about 0.75, about 0.8, about 0.85, about 0.87, about 0.89, about 0.9, about 0.92, about 0.94, about 0.96, about 0.98, about 1.0, about 1.02, about 1.04, about 1.06, about 1.08, about 1.1, about 1.12, about 1.14, about 1.16, about 1.18, about 1.2, about 1.22, about 1.24, about 1.26, about 1.28, about 1.3, about 1.32, about 1.34, about 1.36, about 1.38, about 1.4, about 1.5, and about 1.5. In some embodiments, the PBF is about 0.89. In some embodiments, the compounds disclosed herein have a ratio of principal moment of inertia to molecular weight (PMI1/MW) between about 1 and about 20, between about 1 and about 18, between about 1 and about 15, between about 1 and about 12, between about 1 and about 10, or between about 2 and about 9. In some embodiments, the PMI1/MW is selected from the group consisting of about 2.2, about 2.3, about 2.4, about 2.5, about 2.6, about 2.7, about 2.8, about 2.9, about 3.0, about 3.1, about 3.2, about 3.3, about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4.0, about 4.1, about 4.2, about 4.3, about 4.4, about 4.5, about 4.6, about 4.7, about 4.8, about 4.9, about 5.0, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, about 6.0, about 6.1, about 6.2, about 6.3, about 6.4, about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, about 8.0, about 8.1, about 8.2, about 8.3, about 8.4, about 8.5, about 8.6, about 8.7, about 8.8, about 8.9, and about 9.0. In some embodiments, the compounds disclosed herein have a combination of any number of the foregoing properties. Exemplary Compounds Provided herein are compounds that are derivatives or analogs of an oxazolidinone, lincosamide, mutilin, pleuromutilin, fusidic acid, ETX0914, gepotidacin, iclaprim, or a deoxynybomycin. In some embodiments, the compound has RB of 6 or less, 5 or less, or 4 or less and a Glob of 0.4 or less, 0.3 or less, 0.2 or less, or 0.1 or less; or a combination of any of the foregoing. In certain embodiments, the compounds disclosed herein are derivatives or analogs of a compound selected from the group consisting of linezolid, eperezolid, lincomycin, clindamycin, retapamulin, pleuromutilin, fusidic acid, ETX0914, gepotidacin, iclaprim, PF-708093, AZD0914, and deoxynybomycin (FIG.1). Exemplary Oxazolidinone Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of an oxazolidinone antibiotic such as linezolid or eperezolid, wherein linezolid is and wherein eperzolid is The compounds disclosed herein exclude linezolid and eperezolid. In some embodiments, the oxazolidinone derivative or analog has RB of 4 or less and a Glob of 0.06 or less. In some embodiments, the lincosamide derivative or analog has RB of 4 or less, a Glob of 0.06 or less, PBF of about 0.89, and a PMI1/MW of about 2.7 or 2.9. In some embodiments, the compound is represented by Formula (I) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R1is selected from the group consisting of substituted and unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or R2and R3, taken together, form a 5-10-membered heterocyclic or heteroaromatic ring comprising one N heteroatom and optionally further comprising one or two heteroatoms independently selected from the group consisting of O, N, and S; R4is selected from the group consisting of hydrogen and (C1-C6)alkyl; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; n is an integer from 1-4; and m is 2 or 3; provided that at least one of R1, R2, or R3comprises a terminal —N(R5)m. In some embodiments, R1is selected from the group consisting of substituted and unsubstituted cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl. In some embodiments, R1is substituted or unsubstituted heterocycloalkyl, heteroaryl, or heteroaralkyl. In some embodiments, R1is substituted or unsubstituted heterocycloalkyl or substituted or unsubstituted heteroaryl. In some embodiments, R1is represented by wherein W is selected from the group consisting of substituted and unsubstituted cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl. In some embodiments, R1is represented by wherein W is heterocycloalkyl or heteroaryl. In some embodiments, the compound of formula (I) is represented by Formula (Ia) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R1is represented by wherein W is heterocycloalkyl or heteroaryl; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or R2and R3, taken together, form a 5-10-membered heterocyclic or heteroaromatic ring comprising one N heteroatom and optionally further comprising one or two heteroatoms independently selected from the group consisting of O, N, and S; R4is selected from the group consisting of hydrogen and (C1-C6)alkyl; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; n is an integer from 1-4; m is 2 or 3; and p is an integer from 0-4; provided that at least one of R2, R3, or R6comprises a terminal —N(R5)m. In some embodiments, R2and R3, taken together, form a 5-10-membered heterocyclic or heteroaromatic ring comprising one N heteroatom and optionally further comprising one or two heteroatoms independently selected from the group consisting of O, N, and S. In some embodiments, the compound of formula (I) is represented by Formula (Ib) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R1is selected from the group consisting of substituted and unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; X is selected from the group consisting of CH2, O, NH, and S; R4is selected from the group consisting of hydrogen and (C1-C6)alkyl; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; n is an integer from 1-3; m is 2 or 3; and q is an integer from 0-5; provided that at least one of R1or R2comprises a terminal —N(R5)m. In some embodiments, independently for each occurrence if present: R1is selected from the group consisting of substituted and unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, heterocycloalkyl, and heteroaryl; or R1is represented by wherein W is heterocycloalkyl or heteroaryl; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or R2and R3, taken together, form a 5-10-membered heterocyclic or heteroaromatic ring comprising one N heteroatom and optionally further comprising one or two heteroatoms independently selected from the group consisting of O, N, and S; R4is (C1-C6)alkyl; R5is hydrogen or (C1-C6)alkyl; R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkyl)N(R5)m, —C(O)((C1-C6)alkyl)N(R5)m, —OR5, and —N(R5)m; X is selected from the group consisting of CH2, O, NH, and S; n is 1 or 2; m is 2 or 3; p is O or 1; and q is 0 or 1. In some embodiments, R1is selected from the group consisting of substituted and unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, heterocycloalkyl, and heteroaryl; or R1is represented by wherein W is heterocycloalkyl or heteroaryl; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R1is selected from the group consisting of substituted and unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, heterocycloalkyl, and heteroaryl; or R1is represented by wherein W is heterocycloalkyl or heteroaryl; p is 0 or 1; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, the heterocycloalkyl or heteroaryl is selected from the group consisting of morpholinyl, piperazinyl, and pyridinyl. In some embodiments, R1comprises a terminal —N(R5)m. In some embodiments, R1is unsubstituted —((C1-C6)alkylene)N(R5)mor —C(O)((C1-C6)alkylene)N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments of the compounds of formula (I), (Ia), or (Ib), R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R2comprises a terminal —N(R5)m. In some embodiments R2is —N(R5)m. In some embodiments, R2is halogen. In some embodiments of the compounds of formula (I), (Ia), or (Ib), R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, and —((C1-C6)alkyene)N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R2comprises a terminal —N(R5)m. In some embodiments, R3is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments R3is —((C1-C6)alkyene)N(R5)m. In some embodiments R3is hydrogen. In some embodiments, R6comprises a terminal —N(R5)m. In some embodiments, R6is —((C1-C6)alkylene)N(R5)mor —C(O)((C1-C6)alkyl)N(R5)m; R5is hydrogen or (C1-C6)alkyl; and p is 0 or 1. In some embodiments of the compounds of formula (I), (Ia), or (Ib), m is 2. In some embodiments, m is 3. In some embodiments of the compounds of formula (I), (Ia), or (Ib), p is 0. In some embodiments, p is 1. In some embodiments of the compounds of formula (I), (Ia), or (Ib), q is 0. In some embodiments, the compound of formula (I) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of formula (I) is selected from the group consisting of: In some embodiments, the compound of formula (I) is selected from the group consisting of: Exemplary PF-708093 Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of PF-708093, wherein PF-708093 is The compounds disclosed herein exclude PF-708093. In some embodiments, the PF-708093 derivative or analog has RB of 5 or less and a Glob of 0.05 or less. In some embodiments, the compound is represented by Formula (II) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R1is selected from the group consisting of substituted and unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or R1and R2, taken together, form a substituted 3-10-membered cycloalkyl or aromatic ring or form a substituted 3-10-membered heterocyclic or heteroaromatic ring comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R7is selected from the group consisting of —C(O)N(R5)m, —((C1-C6)alkylene)N(R5)m, —((C1-C6)alkylene)N(R5)m(C(O))((C1-C6)alkyl), —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m, —C(O)(N(R5)m)(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; n is an integer from 1-4; and m is an integer from 1-3; provided that at least one of R1, R2, R3, or R7comprises a terminal —N(R5)m; provided that the compound of formula (II) is not In some embodiments, R1is selected from the group consisting of substituted and unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, aralkyl, and heteroaralkyl. In some embodiments, R1is substituted or unsubstituted —((C1-C6)alkylene)N(R5)mor —C(O)((C1-C6)alkylene)N(R5)m. In some embodiments, R1is substituted or unsubstituted aralkyl or heteroaralkyl. In some embodiments, R1is substituted or unsubstituted cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl. In some embodiments, R1is substituted or unsubstituted cycloalkyl, heterocycloalkyl, aryl, or heteroaryl. In some embodiments of the compound of formula (II), R1comprises a terminal —N(R5)m. In some embodiments, R1is selected from the group consisting of substituted cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl. In some embodiments, R1is represented by wherein W is selected from the group consisting of substituted and unsubstituted cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl and heteroaralkyl. In some embodiments, R1is represented by wherein W is cycloalkyl, heterocycloalkyl, aryl, or heteroaryl. In some embodiments, the compound of formula (II) is represented by Formula (IIa) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R1is represented by wherein W is cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or p is at least 2, and two R6groups taken together form an oxo; R7is selected from the group consisting of —C(O)N(R5)m, —((C1-C6)alkylene)N(R5)m, —((C1-C6)alkylene)N(R5)m(C(O))((C1-C6)alkyl), —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m, —C(O)(N(R5)m)(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; n is an integer from 1-4; m is an integer from 1-3; and p is an integer from 0-4; provided that at least one of R2, R3, R6, or R7comprises a terminal —N(R5)m. In some embodiments, independently for each occurrence: R1is represented by wherein W is cycloalkyl, heterocycloalkyl, or heteroaryl; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; R6is —((C1-C6)alkylene)N(R5)mor —N(R5)m; or p is at least 2, and two R6groups taken together form an oxo; and p is 1, 2, or 3. In some embodiments, the cycloalkyl, heterocycloalkyl, or heteroaryl is selected from the group consisting of cyclopropyl, tetrahydrothiopyranyl, and imidazolyl. In some embodiments, p is at least 2, and two R6groups taken together form an oxo. In some embodiments, R1is represented by which represents In some embodiments, R1is represented by which represents In some embodiments, R6comprises a terminal —N(R5)m. In some embodiments, R6is —((C1-C6)alkylene)N(R5)mor —C(O)((C1-C6)alkylene)N(R5)m. In some embodiments, R6is (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, or —OR5. In some embodiments, p is 2. In some embodiments, p is 3. In some embodiments, R1and R2, taken together, form a substituted 3-10-membered cycloalkyl or aromatic ring or form a substituted 3-10-membered heterocyclic or heteroaromatic ring comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S. In some embodiments, the compound of formula (II) is represented by Formula (IIb) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R7is selected from the group consisting of —C(O)N(R5)m, —((C1-C6)alkylene)N(R5)m, —((C1-C6)alkylene)N(R5)m(C(O))((C1-C6)alkyl), —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m, —C(O)(N(R5)m)(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R8is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or n is at least 2, and two R8groups taken together form an oxo; ring A is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; n is an integer from 1-3; m is an integer from 1-3; and o is an integer from 1-4; provided that at least one of R2, R3, R7, or R8comprises a terminal —N(R5)m. In some embodiments, independently for each occurrence if present: R1is represented by wherein W is cycloalkyl, heterocycloalkyl, or heteroaryl; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; or R1and R2, taken together, form a substituted 3-10-membered heterocyclic comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R5is hydrogen or (C1-C6)alkyl; R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or p is at least 2, and two R6groups taken together form an oxo; R7is selected from the group consisting of —C(O)N(R5)m, —((C1-C6)alkyl)N(R5)m, —((C1-C6)alkylene)N(R5)m(C(O))((C1-C6)alkyl), —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m, —C(O)(N(R5)m)(cycloalkyl)N(R5)m, and heteroaralkyl. R8is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or o is 2, and two R8groups taken together form an oxo; ring A is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; n is an integer from 1-3; m is an integer from 1-3; o is an integer from 1-4; and p is 1, 2, or 3. In some embodiments of formula (IIb), R8is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or o is at least 2, and two R8groups taken together form an oxo; ring A is heterocycloalkyl; and o is 1, 2, or 3. In some embodiments, the heterocycloalkyl is pyrrolidinyl or azepanyl. In some embodiments, R8is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m. In some embodiments, R8comprises a terminal —N(R5)m. In some embodiments, R8is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, and —N(R5)m. In some embodiments, R8is —((C1-C6)alkylene)N(R5)m. In some embodiments, o is 2. In some embodiments, o is 3. In some embodiments, o is at least 2, and two R8groups taken together form an oxo. In some embodiments, o is 3, two R8groups taken together form an oxo; and ring A, and the phenylene to which it is attached are represented by In some embodiments of the compounds of formula (II), (IIa), or (IIb), R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R2comprises a terminal —N(R5)m. In some embodiments R2is —N(R5)m. In some embodiments, R2is halogen. In some embodiments of the compounds of formula (II), (IIa), or (IIb), R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, and —((C1-C6)alkyene)N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R3comprises a terminal —N(R5)m. In some embodiments, R3is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments R3is —((C1-C6)alkyene)N(R5)m. In some embodiments R3is hydrogen. In some embodiments, R7is selected from the group consisting of —C(O)N(R5)m, —((C1-C6)alkylene)N(R5)m, —((C1-C6)alkylene)N(R5)m(C(O))((C1-C6)alkyl), —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m, —C(O)(N(R5)m)(cycloalkyl)N(R5)m, and heteroaralkyl. In some embodiments, R7comprises a terminal —N(R5)m. In some embodiments, R7is selected from the group consisting of —C(O)N(R5)m, —((C1-C6)alkylene)N(R5)m, —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m, and —C(O)(N(R5)m)(cycloalkyl)N(R5)m. In some embodiments of the compounds of formula (II), (IIa), or (IIb), m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, the compound of formula (II) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of formula (II) is selected from the group consisting of: In some embodiments, the compound of formula (II) is selected from the group consisting of: Exemplary Mutilin and Pleuromutilin Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of a mutilin or pleuromutilin, such as retapamulin or pleuromutilin, wherein retapamulin is and wherein pleuromutilin is The compounds disclosed herein exclude retapamulin and pleuromutilin. In some embodiments, the mutilin or pleuromutilin derivative or analog has RB of 6 or less and a Glob of 0.4 or less. In some embodiments, the mutilin or pleuromutilin derivative or analog has RB of 6 or less, a Glob of 0.36 or less, PBF of about 1.3, and a PMI1/MW of about 5.5. In some embodiments, the compound is represented by Formula (III) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R9is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R10is selected from the group consisting of (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R11is selected from the group consisting of (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; and m is 2 or 3; provided that at least one of R3, R9, or R11comprises a terminal —N(R5)m. In some embodiments, independently for each occurrence: R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R5is hydrogen or (C1-C6)alkyl; R9is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R10is (C1-C6)alkyl or cycloalkyl; R11is selected from the group consisting of (C2-C6)alkenyl, (C2-C6)alkynyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, heterocycloalkyl, and heteroaralkyl; and m is 2 or 3. In some embodiments, R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R3comprises a terminal —N(R5)m. In some embodiments, R3is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments, R3is —((C1-C6)alkyene)N(R5)m. In some embodiments, R3is hydrogen or —N(R5)m. In some embodiments, R3is hydrogen. In some embodiments, R9is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R9is —OR5or —N(R5)m. In some embodiments, R9comprises a terminal —N(R5)m. In some embodiments, R9is —((C1-C6)alkylene)N(R5)mor —N(R5)m. In some embodiments, R9is —N(R5)m. In some embodiments, R10is (C1-C6)alkyl or cycloalkyl. In some embodiments, R10is (C1-C6)alkyl. In some embodiments, R11is selected from the group consisting of (C2-C6)alkenyl, (C2-C6)alkynyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, heterocycloalkyl, and heteroaralkyl. In some embodiments, R11is (C1-C6)alkenyl or —((C1-C6)alkyl)N(R5)m. In some embodiments, R11comprises a terminal —N(R5)m. In some embodiments, R11is —((C1-C6)alkylene)N(R5)mor —C(O)((C1-C6)alkylene)N(R5)m. In some embodiments, R11is —((C1-C6)alkylene)N(R5)m. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, the compound of formula (III) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. Exemplary Lincosamide Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of a lincosamide antibiotic such as lincomycin or clindamycin, wherein lincomycin is wherein clindamycin is The compounds disclosed herein exclude lincomycin and clindamycin. In some embodiments, the lincosamide derivative or analog has RB of 7 or less and a Glob of 0.2 or less. In some embodiments, the lincosamide derivative or analog has RB of 7 or less, a Glob of 0.16 or less, PBF of about 1.13, and a PMI1/MW of about 5. In some embodiments, the compound is represented by Formula (IV) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R8is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or o is at least 2, and two R8groups taken together form an oxo; R12is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)OR5, —((C1-C6)alkylene)Z, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, -(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; or R8and R12, taken together, form an unsubstituted or substituted 5-10-membered heterocyclic or heteroaromatic ring comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S; X is selected from the group consisting of CH2, O, NH, and S; W is heterocycloalkyl or heteroaryl; Z is halogen; m is 2 or 3; o is 1 or 2; and p is an integer from 1-4; provided that at least one of R6, R8, or R12comprises a terminal —N(R5)m; provided that the compound of formula (IV) is not In some embodiments, independently for each occurrence: R5is hydrogen or (C1-C6)alkyl; R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6) alkylene)N(R5)m, —OR5, and —N(R5)m; R8is —OR5or —N(R5)m; or o is 2, and two R8groups taken together form an oxo; R12is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)OR5, —((C1-C6)alkylene)Z, —((C1-C6)alkylene)N(R5)m, -(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aralkyl, and heteroaralkyl; or R8and R12, taken together, form an unsubstituted or substituted 5-10-membered heteroaromatic ring comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S; X is selected from the group consisting of O, NH, and S; W is heterocycloalkyl or heteroaryl; m is 2 or 3; o is 1 or 2; and p is an integer from 1-4. In some embodiments, R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R6comprises a terminal —N(R5)m. In some embodiments, R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, and —N(R5)m. In some embodiments, the moiety is selected from the group consisting of In some embodiments, R8comprises a terminal —N(R5)m. In some embodiments, R8is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments, R8is —OR5or —N(R5)m; or o is 2, and two R8groups taken together form an oxo. In some embodiments, o is 2, and two R8groups taken together form an oxo. In some embodiments, R12is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)OR5, —((C1-C6)alkylene)Z, —((C1-C6)alkylene)N(R5)m, -(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aralkyl, and heteroaralkyl. In some embodiments, R12is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)OR5, —((C1-C6)alkylene)Z, 1-C6)alkylene)N(R5)m, -(cycloalkyl)N(R5)m, and cycloalkyl. In some embodiments, R12comprises a terminal —N(R5)m. In some embodiments, R12is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or -(cycloalkyl)N(R5)m. In some embodiments, the moiety is selected from the group consisting of In some embodiments, R8and R12, taken together, form an unsubstituted or substituted 5-10-membered heteroaromatic ring comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S. In some embodiments, the 5-10-membered heteroaromatic ring is selected from the group consisting of oxazolyl, oxadiazolyl, and triazolyl. In some embodiments, X is selected from the group consisting of O, NH, and S. In some embodiments, X is NH. In some embodiments, the moiety is selected from the group consisting of In some embodiments, W is heterocycloalkyl or heteroaryl. In some embodiments, W is heterocycloalkyl. In some embodiments, the heterocycloalkyl is selected from the group consisting of pyrrolidinyl, piperidinyl, and azepanyl. In some embodiments, the moiety is selected from the group consisting of In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, p is 1 or 2. In some embodiments, p is 1. In some embodiments, p is 2. In some embodiments, the compound of formula (IV) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. Exemplary Iclaprim Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of a diaminopyrimidine dihydrofolate reductase (DHFR)-inhibiting antibiotic such as iclaprim or trimethoprim iclaprim, wherein iclaprim is The compounds disclosed herein exclude iclaprim. In some embodiments, the compound has RB of 5 or less and a Glob of 0.2 or less. In some embodiments, the compound has RB of 5 or less, a Glob of 0.16 or less, PBF of about 1.05, and a PMI1/MW of about 5. In some embodiments, the compound is represented by Formula (V) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R12is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)OR5, —((C1-C6)alkylene)Z, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, -(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; and Z is halogen; m is 2 or 3; provided that at least one of R3or R12comprises a terminal —N(R5)m. In some embodiments, independently for each occurrence: R3is selected from the group consisting of hydrogen, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R5is hydrogen or (C1-C6)alkyl; R12is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)OR5, —((C1-C6)alkylene)Z, —((C1-C6)alkylene)N(R5)m, -(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aralkyl, and heteroaralkyl; and m is 2 or 3. In some embodiments, R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, hydrogen, —((C1-C6)alkyl)N(R5)m, —OR5, and —N(R5)m. In some embodiments, R3comprises a terminal —N(R5)m. In some embodiments, R3is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments, R3is —((C1-C6)alkyene)N(R5)m. In some embodiments, R3is hydrogen or —N(R5)m. In some embodiments, R3is hydrogen. In some embodiments, R12is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)OR5, —((C1-C6)alkylene)Z, —((C1-C6)alkylene)N(R5)m, -(cycloalkyl)N(R5)m, cycloalkyl, heterocycloalkyl, aralkyl, and heteroaralkyl; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R12is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, -(cycloalkyl)N(R5)m, and cycloalkyl. In some embodiments, R12comprises a terminal —N(R5)m. In some embodiments, R12is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)mor -(cycloalkyl)N(R5)m. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, the compound of formula (V) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. Exemplary AZD0914 Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of AZD0914, ETX0914, or QPT-1, wherein AZD0914 or ETX0914 is and wherein QPT-1 is The compounds disclosed herein exclude AZD0914, ETX0914, and QPT-1. In some embodiments, the compound has RB of 2 or less and a Glob of 0.1 or less. In some embodiments, the lincosamide derivative or analog has RB of 1 or less, a Glob of 0.09 or less, PBF of about 0.94, and a PMI1/MW of about 5.3. In some embodiments, the compound is represented by Formula (VI) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R13is selected from the group consisting of —CN, —NO2, substituted or unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —(N(R5)m)((C1-C6)alkylene)N(R5)m, —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R14is selected from the group consisting of halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or R13and R14, taken together, form a substituted 3-10-membered cycloalkyl or aromatic ring or form a substituted 3-10-membered heterocyclic or heteroaromatic ring comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S; m is an integer from 1-3; and r is an integer from 0-4; provided that at least one of R13or R14comprises a terminal —N(R5)mor a terminal —CN. In some embodiments, independently for each occurrence: R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R13is selected from the group consisting of —CN, —NO2, substituted or unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —(N(R5)m)((C1-C6)alkylene)N(R5)m, and —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m; R14is selected from the group consisting of halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or m is an integer from 1-3; and r is an integer from 0-4; provided that at least one of R13or R14comprises a terminal —N(R5)mor a terminal —CN. In some embodiments, independently for each occurrence: R5is hydrogen or (C1-C6)alkyl; R13is selected from the group consisting of —CN, —NO2, —((C1-C6)alkylene)N(R5)m, and —(N(R5)m)((C1-C6)alkylene)N(R5)m; R14is selected from the group consisting of halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; m is an integer from 1-3; and r is 0 or 1. In some embodiments, R13is selected from the group consisting of —CN, —NO2, substituted or unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —(N(R5)m)((C1-C6)alkylene)N(R5)m, —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl. In some embodiments, R13is substituted or unsubstituted cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl. In some embodiments, R13is selected from the group consisting of —CN, —NO2, substituted or unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —(N(R5)m)((C1-C6)alkylene)N(R5)m, and —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m. In some embodiments, R7comprises a terminal —N(R5)m. In some embodiments, R13is selected from the group consisting of substituted and unsubstituted —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —(N(R5)m)((C1-C6)alkylene)N(R5)m, and —C(O)(N(R5)m)((C1-C6)alkylene)N(R5)m. In some embodiments, R13and R14, taken together, form a substituted 3-10-membered cycloalkyl or aromatic ring or form a substituted 3-10-membered heterocyclic or heteroaromatic ring comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S. In some embodiments, R13and R14, taken together, form a substituted 3-10-membered heterocyclic or heteroaromatic ring comprising 1-3 heteroatoms independently selected from the group consisting of O, N, and S. In some embodiments, the compound of formula (VI) is represented by Formula (VIa) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or p is at least 2, and two R6groups taken together form an oxo; R14is selected from the group consisting of halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or R15is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, and —O((C1-C6)alkylene)N(R5)m; or R15is represented by L is selected from the group consisting of a covalent bond, —O—, —N(R5)m—, —C(O)—, and —C(O)N(R5)m—; Y is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; m is an integer from 1-3; p is an integer from 0-4; and r is an integer from 0-4; provided that at least one of R14or R15comprises a terminal —N(R5)m. In some embodiments, R14is selected from the group consisting of halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R14comprises a terminal —N(R5)m. In some embodiments, R14is —N(R5)m. In some embodiments, R15comprises a terminal —N(R5)m. In some embodiments, R15is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, and —O((C1-C6)alkylene)N(R5)m. In some embodiments, R15is —((C1-C6)alkylene)N(R5)mor —O((C1-C6)alkylene)N(R5)m. In some embodiments, R15is represented by In some embodiments, L is selected from the group consisting of a covalent bond, —(C1-C6)alkylene-, —O—, —N(R5)m—, —C(O)— and —C(O)N(R5)m—; Y is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; R5is hydrogen or (C1-C6)alkyl; R6is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or p is at least 2, and two R6groups taken together form an oxo; and p is an integer from 1-3. In some embodiments, Y is selected from the group consisting of cyclopropyl, cyclopentenyl, azetidinyl, oxazolindinyl, and phenyl. In some embodiments, p is at least 2, and two R6groups taken together form an oxo. In some embodiments, R15is represented by a moiety selected from the group consisting of: In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, the compound of formula (VI) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof, wherein R15is represented by a moiety selected from the group consisting of: In some embodiments, the compound of formula (VI) is selected from the group consisting of: a pharmaceutically acceptable salt thereof. In some embodiments, the compound of formula (VI) is wherein R15is represented by a moiety selected from the group consisting of: Exemplary Gepotidacin Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of gepotidacin, wherein gepotidacin is The compounds disclosed herein exclude gepotidacin. In some embodiments, the compound has RB of 5 or less and a Glob of 0.15 or less. In some embodiments, the compound has RB of 5 or less, a Glob of 0.12 or less, PBF of about 1.1, and a PMI1/MW of about 4.4. In some embodiments, the compound is represented by Formula (VII) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: represents a single or a double bond; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R17is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; or R3and R17, taken together, form a 5-10-membered heterocyclic or heteroaromatic ring comprising one N heteroatom and optionally further comprising one or two heteroatoms independently selected from the group consisting of O, N, and S; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; or n is at least 2, and two R2groups taken together form an oxo; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R6is selected from the group consisting of (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or p is at least 2, and two R6groups taken together form an oxo; and R16is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or t is at least 2, and two R16groups taken together form a C1-C5carbon bridge; and A is C or N; X is selected from the group consisting of CH2, O, NH, and S; m is 2 or 3; n is an integer from 1-4; p is an integer from 0-4; and t is an integer from 0-4; provided that at least one of R3, R6, or R16comprises a terminal —N(R5)m. In some embodiments, independently for each occurrence if present: R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R17is hydrogen or (C1-C6)alkyl; or R3and R17, taken together, form a 5-10-membered heterocyclic or heteroaromatic ring comprising one N heteroatom and optionally further comprising one or two heteroatoms independently selected from the group consisting of O, N, and S; R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; or n is at least 2, and two R2groups taken together form an oxo; R5is hydrogen or (C1-C6)alkyl; R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or p is at least 2, and two R6groups taken together form an oxo; R16is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or t is at least 2, and two R16groups taken together form a C1-C5 carbon bridge; and A is C or N; X is selected from the group consisting of CH2, O, NH, and S; m is 2 or 3; n is an integer from 1-4; p is an integer from 0-4; and t is an integer from 0-4. In some embodiments, R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, and —N(R5)m; and R17is hydrogen or (C1-C6)alkyl. In some embodiments, R3is hydrogen or —N(R5)m. In some embodiments, R3comprises a terminal —N(R5)m. In some embodiments, R3is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments R3is —N(R5)m. In some embodiments R3is hydrogen. In some embodiments, R3and R17, taken together, form a 5-10-membered heterocyclic or heteroaromatic ring comprising one N heteroatom and optionally further comprising one or two heteroatoms independently selected from the group consisting of O, N, and S. In some embodiments, the compound of formula (VII) is represented by Formula (VIIa) or a pharmaceutically acceptable salt thereof: In some embodiments, X is CH2. In some embodiments, the compound of formula (VII) or (VIIa) is represented by Formula (VIIb) or a pharmaceutically acceptable salt thereof: In some embodiments of the compounds of formula (VII), (VIIa), or (VIIb), R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; or p is at least 2, and two R6groups taken together form an oxo. In some embodiments, R6is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m. In some embodiments, R6comprises a terminal —N(R5)m. In some embodiments, R6is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkyl)N(R5)m, or —N(R5)m; R5is hydrogen or (C1-C6)alkyl; and p is 0 or 1. In some embodiments, p is at least 2, and two R6groups taken together form an oxo. In some embodiments, p is 0. In some embodiments of the compounds of formula (II), (IIa), or (IIb), R16is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m. In some embodiments, R16comprises a terminal —N(R5)m. In some embodiments, R16is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkyl)N(R5)m, or —N(R5)m. In some embodiments, R16is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkyl)N(R5)m, or —N(R5)m. In some embodiments, R16is —((C1-C6)alkylene)N(R5)mor —N(R5)m. In some embodiments of the compounds of formula (II), (IIa), or (IIb), A is N. In some embodiments, A is C. In some embodiments, t is at least 2, and two R16groups taken together form a C1-C5carbon bridge; and A in the bridged ring is C. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, the compound of formula (VII) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of formula (VII) is In some embodiments, the compound of formula (VII) is selected from the group consisting of: Exemplary Fusidic Acid Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of fusidic acid, wherein fusidic acid is The compounds disclosed herein exclude fusidic acid. In some embodiments, the compound has RB of 6 or less and a Glob of 0.15 or less. In some embodiments, the compound has RB of 6 or less, a Glob of 0.1 or less, PBF of about 1.05, and a PMI1/MW of about 3.17. In some embodiments, the compound is represented by Formula (VIII) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R9is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R10is selected from the group consisting of (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R18is selected from the group consisting of (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; or R18is represented by L is selected from the group consisting of a covalent bond, —((C1-C6)alkylene)-, —O—, —N(R5)m—, —C(O)—, and —C(O)N(R5)m—; Y is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; R19is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, -(cycloalkyl)((C1-C6)alkyl), -(heterocycloalkyl)((C1-C6)alkyl), -(aryl)((C1-C6)alkyl), -(heteroaryl)((C1-C6)alkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —OR5, and —N(R5)m; R20is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —((C1-C6)alkene)((C1-C6)alkyl), —((C1-C6)alkene)(cycloalkyl), —((C1-C6)alkene)(heterocycloalkyl), —OR5, and —N(R5)m; m is 2 or 3; and u is an integer from 0-4; provided that at least one of R3, R6, R9, R18, or R19comprises a terminal —N(R5)m. In some embodiments, independently for each occurrence: R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R5is hydrogen or (C1-C6)alkyl; R9is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R10is (C1-C6)alkyl or cycloalkyl; R18is selected from the group consisting of (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; or R18is represented by L is a covalent bond or —((C1-C6)alkylene)-; Y is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; R19is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, -(cycloalkyl)((C1-C6)alkyl), -(heterocycloalkyl)((C1-C6)alkyl), -(aryl)((C1-C6)alkyl), -(heteroaryl)((C1-C6)alkyl), —OR5, and —N(R5)m; R20is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —((C1-C6)alkene)((C1-C6)alkyl), —((C1-C6)alkene)(cycloalkyl), —((C1-C6)alkene)(heterocycloalkyl), —OR5, and —N(R5)m; m is 2 or 3; and u is an integer from 0-4; provided that at least one of R3, R6, R9, or R19comprises a terminal —N(R5)m. In some embodiments, R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m. In some embodiments, R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R3is selected from the group consisting of hydrogen, methyl, —OH, and —NH2. In some embodiments, R3comprises a terminal —N(R5)m. In some embodiments, R3is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments, R3is —((C1-C6)alkyene)N(R5)mor —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R3is hydrogen. In some embodiments, R9is selected from the group consisting of —((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R9is —OR5or —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R9comprises a terminal —N(R5)m. In some embodiments, R9is —((C1-C6)alkylene)N(R5)mor —N(R5)m. In some embodiments, R9is —OH. In some embodiments, R10is (C1-C6)alkyl or cycloalkyl. In some embodiments, R10is (C1-C6)alkyl. In some embodiments, R10is methyl. In some embodiments, R18is selected from the group consisting of (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl. In some embodiments, R18is selected from the group consisting of (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl. In some embodiments, R18is selected from the group consisting of (C1-C6)alkyl, (C2-C6)alkenyl, aryl, heteroaryl, aralkyl, and heteroaralkyl. In some embodiments, R18comprises a terminal —N(R5)m. In some embodiments, R18is represented by L is a covalent bond or —((C1-C6)alkylene)-; Y is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; R20is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —((C1-C6)alkene)((C1-C6)alkyl), —((C1-C6)alkene)(cycloalkyl), —((C1-C6)alkene)(heterocycloalkyl), —OR5, and —N(R5)m; and u is an integer from 0-4. In some embodiments, R20is —((C1-C6)alkene)((C1-C6)alkyl) or —((C1-C6)alkene)(cycloalkyl). In some embodiments, R20comprises a terminal —N(R5)m. In some embodiments, R20is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments, R19is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, -(cycloalkyl)((C1-C6)alkyl), -(heterocycloalkyl)((C1-C6)alkyl), -(aryl)((C1-C6)alkyl), -(heteroaryl)((C1-C6)alkyl), —OR5, and —N(R5)m. In some embodiments, R19is selected from the group consisting of (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, -(heterocycloalkyl)((C1-C6)alkyl), -(heteroaryl)((C1-C6)alkyl), and —N(R5)m. In some embodiments, R19comprises a terminal —N(R5)m. In some embodiments, R19is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, the compound of formula (VIII) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. Exemplary Deoxynybomycin Derivatives In some embodiments of the compounds disclosed herein, the compound is a derivative or analog of deoxynybomycin, wherein deoxynybomycin is The compounds disclosed herein exclude deoxynybomycin. In some embodiments, the compound has RB of 2 or less and a Glob of 0.12 or less. In some embodiments, the compound has RB of 2 or less and a Glob of 0.1 or less. In some embodiments, the compound has RB of 1 and a Glob of about 0.09. In some embodiments, the compound is represented by Formula (IX) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R2is selected from the group consisting of hydrogen, halogen, —CN, (C1-C6)alkyl optionally substituted with 1, 2, or 3 halogen atoms, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —C(O)OR5, —OR5, SR5, —S(O)R5, —SO2R5, —SO2NR5, —NO2, —N3, and —N(R5)m; R3is selected from the group consisting of hydrogen, halogen, —CN, (C1-C6)alkyl optionally substituted with 1, 2, or 3 halogen atoms, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —C(O)OR5, —OR5, —O((C1-C6)alkylene)N(R5)m, —SR5, —S(O)R5, —SO2R5, —SO2NR5, —NO2, —N3, and —N(R5)m; R5is selected from the group consisting of hydrogen, (C1-C6)alkyl optionally substituted with 1, 2, or 3 halogen atoms, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; R10is selected from the group consisting of (C1-C6)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl; and m is 2 or 3; and provided that at least one of R2or R3comprises a terminal —N(R5)m. In some embodiments, independently for each occurrence: R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, —OR5, and —N(R5)m; R5is hydrogen or (C1-C6)alkyl; R10is (C1-C6)alkyl or cycloalkyl; and m is 2 or 3. In some embodiments, R2is selected from the group consisting of hydrogen, halogen, —CN, (C1-C6)alkyl optionally substituted with 1, 2, or 3 halogen atoms, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —C(O)OR5, —OR5, SR5, —S(O)R5, —SO2R5, —SO2NR5, —NO2, —N3, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R2is selected from the group consisting of hydrogen, halogen, (C1-C6)alkyl, —OR5, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R2comprises a terminal —N(R5)m. In some embodiments, R2is —N(R5)m. In some embodiments, R2is hydrogen. In some embodiments, R3is selected from the group consisting of hydrogen, halogen, —CN, (C1-C6)alkyl optionally substituted with 1, 2, or 3 halogen atoms, —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, —C(O)OR5, —OR5, —O((C1-C6)alkylene)N(R5)m, —SR5, —S(O)R5, —SO2R5, —SO2NR5, —NO2, —N3, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R3is selected from the group consisting of hydrogen, (C1-C6)alkyl, —((C1-C6)alkylene)N(R5)m, and —N(R5)m; and R5is hydrogen or (C1-C6)alkyl. In some embodiments, R3is selected from the group consisting of hydrogen, methyl, ethyl, —CH2—NH2, or —CH2(NH)CH3. In some embodiments, R3comprises a terminal —N(R5)m. In some embodiments, R3is —((C1-C6)alkylene)N(R5)m, —C(O)((C1-C6)alkylene)N(R5)m, or —N(R5)m. In some embodiments, R3is —((C1-C6)alkyene)N(R5)m. In some embodiments, R5is hydrogen or (C1-C6)alkyl optionally substituted with 1, 2, or 3 halogen atoms. In some embodiments, the compound of formula (IX) is represented by Formula (IXa) or a pharmaceutically acceptable salt thereof: wherein, independently for each occurrence: R3is —((C1-C6)alkylene)N(R5)mor —N(R5)m; R5is hydrogen or (C1-C6)alkyl; R10is (C1-C6)alkyl or cycloalkyl; and m is 2 or 3. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, the compound of formula (IX) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of formula (IX) is selected from the group consisting of: or a pharmaceutically acceptable salt thereof. Exemplary Methods In some embodiments, the compounds disclosed herein accumulate in Gram-negative bacteria. In some embodiments, the compounds disclosed herein traverse a porin. In some embodiments, provided herein is a method of antimicrobial treatment, comprising, administering to a subject in need thereof a therapeutically effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, thereby killing or inhibiting the growth of at least a portion of a plurality of microorganisms in the subject. In some embodiments, the compound is a compound of any one of formulas (I), (II), (III), (IV), (V), (VI), (VII), (VIII), or (IX). In some embodiments, provided herein is a method of antimicrobial treatment, comprising: providing a sample comprising a plurality of microorganisms; contacting the sample with a compound disclosed herein; thereby killing or inhibiting the growth of at least a portion of the plurality of microorganisms in the sample. In some embodiments of the methods of antimicrobial treatment disclosed herein, at least a portion of the plurality of microorganisms is killed. In some embodiments of the methods of antimicrobial treatment disclosed herein, the growth of at least a portion of the plurality of microorganisms is inhibited. In some embodiments, the microorganism is a bacterium, a virus, a fungus, or a parasite. In some embodiments, the microorganism is drug resistant, such as antibiotic resistant. In some embodiments, the microorganism is multi-drug resistant. In some embodiments, the microorganism is a bacterium. In some embodiments, the microorganism is a Gram-negative bacterium. In some embodiments, the microorganism is a Gram-positive bacterium. In some embodiments, for example, the microorganism is at least one bacterium selected from the group consisting ofAcinetobacter, anthrax-causing bacteria, Bacilli,Bordetella, Borrelia, botulism,Brucella, Burkholderia, Canpylobacter, Chlamydia, cholera-causing bacteria,Clostridium, Conococcus, Corynebacterium, diptheria-causing bacteria,Enterobacter, Enterococcus, Erwinia, Escherichia, Francisella, Haemophilus, Heliobacter, Klebsiella, Legionella, Leptospira, leptospirosis-causing bacteria,Listeria, Lyme's disease-causing bacteria, meningococcus,Mycobacterium, Mvcoplasna, Neisseria, Pasteurella, Pelobacter, plague-causing bacteria,Pneunonococcus, Proteus, Pseudiomonas, Rickettsia, Salmonella, Serratia, Shigella, Staphylococcus, Streptococcus, tetanus-causing bacteria,Treponema, Vibrio, YersiniaandXanthomonas. In some embodiments, the microorganism is at least one bacterium selected from the group consisting ofAcinetobacter baumannii, Escherichia coli, Enterobacter cloacae, Klebsiella pneumoniae, Pseudomonas aeruginosa, andStaphylococcus aureus. In some embodiments, the microorganism is methicillin-resistantStaphylococcus aureus(MRSA). In some embodiments, the microorganism isPseudomonas aeruginosa. In some embodiments, for example, the microorganism is at least one virus selected from Adenoviridae, Papillomaviridae, Polyomaviridae, Herpesviridae, Poxviridae, Hepadnaviridae, Parvoviridae, Astroviridae, Caliciviridae, Picornaviridae, Coronoviridae, Flaviviridae, Retroviridae, Togaviridae, Arenaviridae, Bunyaviridae, Filoviridae, Orthonyxoviridae, Paramyxoviridae, Rhabdoiviridae, and Reoviridae. In certain embodiments, the virus may be arboviral encephalitis virus, adenovirus, herpes simplex type 1, herpes simplex type 2, Varicella-zoster virus, Epstein-barr virus, cytomegalovirus, herpesvirus type 8, papillomavirus, B3K virus, coronavirus, echovirus, JC virus, smallpox, Hepatitis B, bocavirus, parvovirus B19, astrovirus, Norwalk virus, coxsackievirus, Hepatitis A, poliovirus, rhinovirus, severe acute respiratory syndrome virus, Hepatitis C, yellow fever, dengue virus, West Nile virus, rubella, Hepatitis E, human immunodeficiency virus (HIV), human T-cell lymphotropic virus (HTLV), influenza, guanarito virus, Junin virus, Lassa virus, Machupo virus, Sabia virus, Crimean-Congo hemorrhagic fever virus, ebola virus, Marburg virus, measles virus, molluscum virus, mumps virus, parainfluenza, respiratory syncytial virus, human metapneumovirus, Hendra virus, Nipah virus, rabies, Hepatitis D, rotavirus, orbivirus, coltivirus, vaccinia virus, and Banna virus. In some embodiments, for example, the microorganism is at least one fungus selected fromAspergillus(fumigatus, niger, etc.),Basidiobolus(ranarum, etc.),Blastomyces dermatitidis, Candida(albicans, krusei, glabrata, tropicalis, etc.),Coccidioides immitis, Cryptococcus(neoformans, etc.),eumycetoma, Epidermophylon(floccosum, etc.),Histoplasma capsulatum, Hortaea werneckii, Lacazia loboi, Microsproum(audouinii, nanumetc.),Mucorales(mucor, absidia, rhizophus),Paracoccidioides brasiliensis, Rhinosporidiun seeberi, Sporothrix schenkii, andTrichophyton(schoeleinii,mentagrophytes, rubrum, verrucosum, etc.). In some embodiments, for example, the microorganism is at least one parasite selected fromAcanthamoeba, Babesia microti, Balantidium coli, Eniamoeba hystolytica, Giardia lamblia, Cryptospordiun muris, Trypanosonatida gambiense, Trypanosonatida rhodesiense, Trypanosoma brucei, Trypanosoma cruzi, Leishmania mexicana, Leishmania braziliensis, Leishmania tropica, Leishmania donovani, Toxoplasma gondii, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Plasmodium falciparum, Pneumocystis carinii, Trichomonas vaginalis, Histomonoas meleagridis, Secementea, Trichuris trichiura, Ascaris lumbricoides, Enterobius vermicularis, Ancylostoma duodenale, Naegleria fowleri, Necator americanus, Nippostrongylus brasiliensis, Strongyloides stereoralis, Wuchereria bancrofti, Dracunculus medinensis, blood flukes, liver flukes, intestinal flukes, lung flukes,Schistosoma mansoni, Schistosoma haematobium, Schistosoma japonicum, Fasciola hepatica, Fasciola gigantica, Heterophves heterophyes, andParagonimus westermani. For purposes of this disclosure, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 67th Ed., 1986-87, inside cover. Definitions For convenience, before further description of the present disclosure, certain terms employed in the specification, examples, and appended claims are collected here. The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element. The term “heteroatom” is art-recognized and refers to an atom of any element other than carbon or hydrogen. Illustrative heteroatoms include boron, nitrogen, oxygen, phosphorus, sulfur and selenium. The term “alkyl” refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. In preferred embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C1-C30for straight chain, C3-C30for branched chain), and more preferably 20 or fewer. For example, (C1-C6)alkyl. Likewise, preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure. Unless the number of carbons is otherwise specified, “lower alkyl” as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carbon atoms in its backbone structure. Likewise, “lower alkenyl” and “lower alkynyl” have similar chain lengths but with at least two carbon atoms. Preferred alkyl groups are lower alkyls. In preferred embodiments, a substituent designated herein as alkyl is a lower alkyl. The term “aralkyl”, as used herein, means an aryl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Representative examples of arylalkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, and 2-naphth-2-ylethyl. The term “alkoxy” means an alkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, and hexyloxy. The term “alkoxycarbonyl” means an alkoxy group, as defined herein, appended to the parent molecular moiety through a carbonyl group, represented by —C(═O)—, as defined herein. Representative examples of alkoxycarbonyl include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, and tert-butoxycarbonyl. The term “carboxy” as used herein, means a —CO2H group. The term “alkylthio” as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through a sulfur atom. Representative examples of alkylthio include, but are not limited, methylthio, ethylthio, tert-butylthio, and hexylthio. The terms “arylthio,” “alkenylthio” and “arylakylthio,” for example, are likewise defined. The term “amido” as used herein, means —NHC(═O)—, wherein the amido group is bound to the parent molecular moiety through the nitrogen. Examples of amido include alkylamido such as CH3C(═O)N(H)— and CH3CH2C(═O)N(H)—. The term “aryl” as used herein includes 5-, 6- and 7-membered aromatic groups that may include from zero to four heteroatoms, for example, benzene, naphthalene, anthracene, pyrene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as “aryl heterocycles” or “heteroaromatics”. The aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, —CF3, —CN, or the like. The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls. The abbreviations Me, Et, Ph, Tf, Nf, Ts, Ms, and dba represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, nonafluorobutanesulfonyl, p-toluenesulfonyl, methanesulfonyl, and dibenzylideneacetone, respectively. Also, “DCM” stands for dichloromethane; “rt” stands for room temperature, and may mean about 20° C., about 21° C., about 22° C., about 23° C., about 24° C., about 25° C., or about 26° C.; “THF” stands for tetrahydrofuran; “BINAP” stands for 2,2′-bis(diphenylphosphino)-1,1′-binaphthyl; “dppf” stands for 1,1′-bis(diphenylphosphino)ferrocene; “dppb” stands for 1,4-bis(diphenylphosphinobutane; “dppp” stands for 1,3-bis(diphenylphosphino)propane; “dppe” stands for 1,2-bis(diphenylphosphino)ethane. A more comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears in the first issue of each volume of theJournal of Organic Chemistry; this list is typically presented in a table entitled Standard List of Abbreviations. The abbreviations contained in said list, and all abbreviations utilized by organic chemists of ordinary skill in the art are hereby incorporated by reference. The terms ortho, meta and para apply to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively. For example, the names 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous. The terms “heterocyclyl”, “heterocycloalkyl”, or “heterocyclic group” refer to 3- to 10-membered ring structures, more preferably 3- to 7-membered rings, whose ring structures include one to four heteroatoms. Heterocycles can also be polycycles. Heterocyclyl groups include, for example, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxathiin, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, oxolane, thiolane, oxazole, piperidine, piperazine, morpholine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like. The heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, —CF3, —CN, or the like. The terms “polycyclyl” or “polycyclic group” refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are “fused rings”. Rings that are joined through non-adjacent atoms are termed “bridged” rings. Each of the rings of the polycycle can be substituted with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, —CF3, —CN, or the like. The term “heteroatom” as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, sulfur and phosphorous. As used herein, the term “nitro” means —NO2; the term “halogen” or “halo” designates —F, —Cl, —Br or —I; the term “sulfhydryl” means —SH; the term “hydroxyl” means —OH; the term “sulfonyl” means —SO2—; and the term “cyano” as used herein, means a —CN group. The term “haloalkyl” means at least one halogen, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Representative examples of haloalkyl include, but are not limited to, chloromethyl, 2-fluoroethyl, trifluoromethyl, pentafluoroethyl, and 2-chloro-3-fluoropentyl. The terms “amine” and “amino” are art recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that can be represented by the general formula: wherein R9, R10and R′10each independently represent a hydrogen, an alkyl, an alkenyl, —(CH2)m—R8, or R9and R10taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R8represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle or a polycycle; and m is zero or an integer in the range of 1 to 8. In preferred embodiments, only one of R9or R10can be a carbonyl, e.g., R9, R10and the nitrogen together do not form an imide. In even more preferred embodiments, R9and R10(and optionally R′10) each independently represent a hydrogen, an alkyl, an alkenyl, or —(CH2)m—R8. Thus, the term “alkylamine” as used herein means an amine group, as defined above, having a substituted or unsubstituted alkyl attached thereto, i.e., at least one of R9and R10is an alkyl group. The definition of each expression, e.g., alkyl, m, n, and the like, when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure. The terms triflyl (-Tf), tosyl (-Ts), mesyl (-Ms), and nonaflyl are art-recognized and refer to trifluoromethanesulfonyl, p-toluenesulfonyl, methanesulfonyl, and nonafluorobutanesulfonyl groups, respectively. The terms triflate (-OTf), tosylate (-OTs), mesylate (-OMs), and nonaflate are art-recognized and refer to trifluoromethanesulfonate ester, p-toluenesulfonate ester, methanesulfonate ester, and nonafluorobutanesulfonate ester functional groups and molecules that contain said groups, respectively. The phrase “protecting group” as used herein means temporary modifications of a potentially reactive functional group which protect it from undesired chemical transformations. Examples of such protecting groups include silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones, respectively. In embodiments of the disclosure, a carboxylate protecting group masks a carboxylic acid as an ester. In certain other embodiments, an amide is protected by an amide protecting group, masking the —NH2of the amide as, for example, —NH(alkyl), or —N(alkyl)2. The field of protecting group chemistry has been reviewed (Greene, T. W.; Wuts, P. G. M.Protective Groups in Organic Synthesis,2nded.; Wiley: New York, 1991). It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described hereinabove. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms, such as nitrogen, may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms. As used herein, the term “rotatable bonds” as used herein is a count of single bonds, not in a ring, bound to a nonterminal heavy atom. Excluded from the count are C—N amide bonds because of their high rotational energy barrier. Rotatable bonds are abbreviated as “RB” herein. The term “ClogD7.4” as used herein means the predicted octanol/water distribution coefficient at pH 7.4. The term “globularity” as used herein means the inverse condition number of the covariance matrix of atomic coordinates (a value of 1 indicates a perfect sphere while a value of 0 indicates a two- or one-dimensional object). Globularity is abbreviated as “Glob” herein. The phrase “plane of best fit” (PBF) as used herein means the average distance in angstroms of each heavy atom in the molecule to the plane that best fits atomic coordinates. The phrase “principal moment of inertia” (PMI1) as used herein means the first diagonal element of diagonalized moment of inertia tensor. See Sauer, W. H. & Schwarz, M. K. Molecular shape diversity of combinatorial libraries: a prerequisite for broad bioactivity. J J.Chem. Inf Comput. Sci.43, 987-1003, (2003). The phrase “PMI1/MW” as used herein represents the ratio of PMI 1 and molecular weight. This represents the normalized principal moment of inertia. The term “vsurf_A” as used herein means a vector pointing from the center of the hydrophobic domain to the center of the hydrophilic domain. The vector length is proportional to the strength of the amphiphilic moment. See Thanassi, D. G., Suh, G. S. & Nikaido, H. Role of outer membrane barrier in efflux-mediated tetracycline resistance ofEscherichia coli. J. Bacteriol.177, 998-1007, (1995). EXEMPLIFICATION The disclosure may be understood with reference to the following examples, which are presented for illustrative purposes only and which are non-limiting. The substrates utilized in these examples were either commercially available, or were prepared from commercially available reagents. Example 1—Accumulation Assay The method for assessment of accumulation was adapted from Bazile, S., Moreau, N., Bouzard, D. & Essiz, M. Relationships among antibacterial activity, inhibition of DNA gyrase, and intracellular accumulation of 11-fluoroquinolones.Antimicrob. Agents Chemother.36, 2622-2627, (1992); Davis, T. D., Gerry, C. J. & Tan, D. S. General platform for systematic quantitative evaluation of small-molecule permeability in bacteria.ACS Chem. Biol.9, 2535-2544, (2014); and Cai, H., Rose, K., Liang, L. H., Dunham, S. & Stover, C. Development of a liquid chromatography/mass spectrometry-based drug accumulation assay inPseudomonas aeruginosa. Anal. Biochem.385, 321-325, (2009); with LC-MS/MS used to quantify the accumulation of each compound.E. coliMG1655 was utilized for these experiments as this strain has been only minimally altered from its K-12 progenitor. To ensure the assay was reporting on compound accumulation, as opposed to non-specific affinity for the outer membrane, several control experiments were conducted. The assay method was evaluated with antibiotics that have known high or low levels of accumulation, based on published accumulation data and antibacterial activity againstE. coli. Known high accumulating antibacterials (tetracycline, ciprofloxacin, chloramphenicol) and tazobactam (a β-lactamase inhibitor active in Gram-negative bacteria), and low accumulating antibacterials (novobiocin, erythromycin, rifampicin, vancomycin, daptomycin, clindamycin, mupirocin, and fusidic acid) were assessed. Ampicillin was also used as a “low-accumulation” control as it is rapidly covalently appended to the reactive serine residue in the active site of penicillin-binding proteins, preventing measurement by LC-MS/MS. To account for the possibility of non-specific binding to the outer membrane, Gram-negative active antibiotics with various charged states at physiological pH were chosen: tetracycline (positively charged), tazobactam (negatively charged), ciprofloxacin (zwitterionic), and chloramphenicol (neutral). The results show a significantly higher level of accumulation inE. colifor the Gram-negative-active compounds as compared to compounds with low Gram-negative antibacterial activity and ampicillin, consistent with measuring accumulation as opposed to non-specific binding (FIG.2). To further ensure that variations in observed accumulation levels were not due to differences in non-specific affinity of the controls for the membrane, penetrance was perturbed by co-treatingE. coliwith the membrane permeabilizing agent colistin. Colistin is known to potentiate the activity of low-accumulating antibiotics by aiding the diffusion of lipophilic compounds across the membrane. For an assay measuring accumulation as opposed to non-specific binding, low-accumulating antibiotics inE. coliwould show an increased level of accumulation in this experiment. Indeed, in the assay an increase in accumulation for low-accumulating antibiotics (novobiocin, erythromycin, rifampicin, and fusidic acid) was observed upon co-treatment with colistin (data not shown). Towards understanding the physicochemical properties that influence small molecule accumulation inE. coli, a collection of diverse compounds was required. Given that the majority of antibacterial drugs are natural products or their derivatives, critical to this effort would be access to a collection of compounds whose members possess natural-product-like properties, but also are synthetically accessible so that various physicochemical traits are tunable, enabling SAR studies on accumulators. Thus, these compounds were produced using the “complexity-to-diversity” (CtD) strategy (FIG.3), whereby diverse compounds are constructed from readily available natural products. To begin the analysis, a set of 100 compounds, including positively charged, negatively charged, and neutral compounds (including two zwitterions) were synthesized and tested. As retrospective studies suggest that compound accumulation is related to ClogD7.4, after assessing accumulation inE. colifor all 100 compounds, the accumulation data were plotted vs ClogD7.4. This data is shown inFIG.4, with ionic state of the compounds indicated by the shape of the data points (diamonds=positive, triangles=neutral, squares=negative). These results strikingly differ from the conclusions gleaned from retrospective studies. Within this set of 100 compounds, charge is the primary factor dictating accumulation inE. coli. The positively-charged compounds are the most likely to accumulate, with 12 of 41 positively-charged compounds showing a significant level of accumulation compared to the low accumulating controls. In contrast, 0 of 39 neutral compounds and 0 of 20 negatively-charged compounds show a level of accumulation higher than the negative controls. Notably, even carboxylic acids with strongly negative ClogD7.4values (<−5) do not accumulate, whereas certain amines with relatively high ClogD7.4values (>0) do accumulate. All 12 accumulating compounds contain amines, and the majority of these compounds (8 out of 12) are primary amines. To further examine the importance of the primary amine, a SAR analysis was performed for multiple different classes of accumulating compounds. Replacement of the amine with a carboxylic acid, an amide, an ester, a nitrile, an azide, or an alcohol on multiple different scaffolds dramatically reduces accumulation. Even conversion of the primary amine to a more substituted amine has a deleterious effect on accumulation. Shown inFIG.5are three primary amines (1-3) alongside their methylated (4-6), di-methylated (7-9), tri-methylated (10-12), and acetylated derivatives (13-15). In all cases, the primary amine shows the highest level of accumulation, with only one of the derivatives (6) showing significant accumulation. An additional 54 primary amine-containing compounds were then obtained and their accumulation inE. coliwas assessed; these compounds are both CtD compounds and commercially available primary amines that more closely mimic the types of compounds found in commercial screening libraries. A graph of accumulation versus ClogD7.4for all 68 primary amines (8 accumulators from initial test set, 6 non-accumulators from initial test set, and 54 additional amines) is provided inFIG.6. Again, for this set of amines accumulation does not increase with lower ClogD7.4, and when MW is plotted versus accumulation for the 68 primary amines, there is also no correlation (FIG.7). SAR analysis on several of these compounds also showed the importance of the amine to accumulation (data not shown). While the presence of a primary amine is clearly important for accumulation inE. coli, it is not sufficient: although 36 of the primary amines within the test set do accumulate, 32 of them do not. Therefore, a chemoinformatic approach was implemented to understand which factors contribute to amine accumulation. For this expanded set of 68 primary amines, 297 molecular descriptors were calculated for conformer ensembles of each compound (e.g., MW, ClogD7.4, rotatable bonds, globularity, PBF, and PMI1/MW). For example, this set of primary amines included dihydrofolate reductase inhibitors (e.g., iclaprim), fluoroquinolone (e.g., ciprofloxacin and lomefloxacin), sulfa (e.g., sulfadiazine and sulfameter), tetracycline (e.g., doxycycline and tigecycline), macrolide (e.g., clarithromycin and erythromycin), oxazolidinone (e.g., ranbezolid), glycopeptide (e.g., dalbavancin and vancomycin), lincosamide (e.g., lincomycin), lipopeptide (e.g., daptomycin), ansamycin (e.g., rifampicin), and steptogramin (e.g., dalfopristin) antibiotics. The molecular descriptors were used to train a random forest classification model that predicts amine accumulation (data not shown). The random forest model offers many advantages for this application including resistance to over-fitting and the ability to measure descriptor importance. Through this analysis it was revealed that the flexibility and shape of a compound are important factors that govern accumulation. Flexibility was best captured by measuring the number of rotatable bonds (RB), whereas shape was best described by the term globularity (ratio of smallest eigenvalue and largest eigenvalue following principal component analysis of atomic coordinates). This globularity analysis (Glob), is routinely used to provide information on the three-dimensionality of compounds, where a completely flat compound (e.g. benzene) has a Glob of 0 and a spherical compound (e.g. adamantane) has a Glob of 1. Molecular Operating Environment (MOE), 2015.10 (1010 Sherbooke St. West, Suite #910, Montreal, QC, 381 Canada, H3 A 2R7, 2016). Case studies demonstrate the importance of the flexibility and globularity parameters for accumulation of the primary amines: as shown inFIG.8A, amine 16 with four RBs accumulates at a high level (1965 nmol/1012CFUs); however, amine 17, a compound of similar molecular weight and Glob but with 13 RBs, shows virtually no accumulation. Analogous results are observed inFIG.8Bwhen comparing a compound with low globularity (18, Glob=0.14, accumulation=922 nmol/1012CFUs) to a compound with similar functional groups, MW, and RBs but with high globularity (19, Glob=0.49, accumulation=335 nmol/1012CFUs); compound globularity is most easily observed in their three-dimensional models,FIG.8B. While in general primary amines with five or fewer RBs and a globularity of 0.25 or less have a markedly higher likelihood of accumulation, two additional factors related to the placement of the primary amine are important. First, the random forest model identified increased amphiphilic moment (vsurf_A), which measures the distance between hydrophobic and hydrophilic portions of a compound, as favoring accumulation. Strikingly, while mono-amine isomannide (20) does not accumulate, derivatives with increased vsurf_A do accumulate (21-23, 2 and 16) (FIG.9), and similar trends are observed for other compound classes (24-27) (FIG.9). Accordingly, some degree of hydrophobicity appears necessary for accumulation, although in practice most organic compounds possess this feature. Secondly, compounds with sterically encumbered primary amines are not high accumulators, for example 28 and 29 (FIG.10A), which both have low flexibility and globularity but contain a primary amine on a tetra-substituted carbon, do not show significant accumulation. This result is in accord with the superior accumulation of primary amines versus substituted amines (FIG.5), and is also consistent with the increased accumulation of compounds where the primary amine is systematically extended from a sterically congested ring system (30<1<31), as shown inFIG.10B. Based on these analyses the following guiding principles for compound accumulation inE. coliwere developed: compounds are most likely to accumulate if they contain a non-sterically encumbered primary amine, some non-polar functionality, are rigid, and have low globularity. As shown inFIG.11, the vast majority of compounds that meet these criteria in the test set accumulate inE. coli. It should be noted that side-by-side comparisons of compounds containing two primary amines to analogues with one amine, (6 paired sets of compounds), largely follow the same rules for accumulation with no clear impact of the second primary amine (FIGS.12A and12B). To further test the validity of these guidelines, the set of antibacterials assessed by O'Shea and Moser was evaluated for charge, flexibility, and globularity. For the charge analysis, antibacterial drugs containing an ionizable nitrogen were accepted. For Gram-negative antibacterials, only those predicted to enter through porins were included, and due to the sheer number of β-lactams they have been left off the graph inFIG.13. As is shown inFIG.13, for these drugs with ionizable nitrogens, compounds active against Gram-negative bacteria cleanly separate from those with Gram-positive-only activity based on these two physicochemical parameters. As is also apparent fromFIG.13, no Gram-positive-only antibacterial with an ionizable nitrogen has the correct rigidity and globularity for accumulation inE. coli. Although globularity was found to best predict accumulators and non-accumulators in combination with flexibility, other measures of three-dimensionality also exhibit the same trend (FIGS.14A-15B). The accumulation assay was performed in triplicate in batches of ten samples, with each batch containing either tetracycline or ciprofloxacin as a positive control. For each replicate, 2.5 mL of an overnight culture ofE. coliwas diluted into 250 mL of fresh Luria Bertani (LB) broth (Lennox) and grown at 37° C. with shaking to an OD600=0.55. The bacteria were pelleted at 3,220 rcf for 10 minutes at 4° C. and the supernatant was discarded. The pellets were re-suspended in 40 mL of phosphate buffered saline (PBS) and pelleted as before, and the supernatant was discarded. The pellets were re-suspended in 8.8 mL of fresh PBS and aliquoted into ten 1.5 mL eppendorf tubes (875 μL each). The number of colony forming units (CFUs) was determined via a calibration curve. The samples were equilibrated at 37° C. with shaking for 5 minutes, compound was added ([final]=50 μM), and then samples were incubated at 37° C. with shaking for 10 minutes. A 10-minute time point was chosen because it is longer than the predicted amount of time required to reach a steady-state concentration, but short enough to minimize metabolic and growth changes (no changes in OD600observed; CFUs were reduced by a factor of five after ciprofloxacin treatment for 10 minutes, but no other antibiotics had an effect). After incubation, 800 μL of the cultures were carefully layered on 700 μL of silicone oil (9:1 AR20/Sigma High Temperature, cooled to −78° C.). Bacteria were pelleted through the oil by centrifuging at 13,000 rcf for 2 minutes at room temperature (supernatant remains above the oil); the supernatant and oil were then removed by pipetting. To lyse the samples, each pellet was dissolved in 200 μL of water, and then they were subjected to three freeze-thaw cycle of three minutes in liquid nitrogen followed by three minutes in a water bath at 65° C. The lysates were pelleted at 13,000 rcf for 2 minutes at room temperature and the supernatant was collected (180 μL). The debris was re-suspended in 100 μL of methanol and pelleted as before. The supernatants were removed and combined with the previous supernatants collected. Finally, remaining debris was removed by centrifuging at 20,000 rcf for 10 minutes at room temperature. Supernatants were analyzed by LC-MS/MS. Samples were analyzed with the 5500 QTRAP LC/MS/MS system (AB Sciex, Foster City, CA) with a 1200 series HPLC system (Agilent Technologies, Santa Clara, CA) including a degasser, an autosampler, and a binary pump. The LC separation was performed on an Agilent SB-Aq column (4.6×50 mm, 5 μm) (Agilent Technologies, Santa Clara, CA) with mobile phase A (0.1% formic acid in water) and mobile phase B (0.1% formic acid in acetonitrile). The flow rate was 0.3 mL/min. The linear gradient was as follows: 0-3 min, 100% A; 10-15 min, 2% A; 15.5-21 min, 100% A. The autosampler was set at 5° C. The injection volume was 15 μL. Mass spectra were acquired with both positive electrospray ionization (ESI) at the ion spray voltage of 5500 V and negative ESI at the ion spray voltage of −4500 V. The source temperature was 450° C. The curtain gas, ion source gas 1, and ion source gas 2 were 33, 50, and 65, respectively. Multiple reaction monitoring (MRM) was used to quantify metabolites. Power analysis was performed using G*Power 3.1 to determine appropriate sample size. Based on data collected from control compounds (FIG.2), three replicates would be necessary in order to detect accumulation above 500 nmol/1012CFUs at 0.96 percent power. Error bars represent the standard error of the mean of three biological replicates. All compounds evaluated in biological assays were ≥95% pure. Assays measuring permeabilization by colistin were performed as above, with the addition of 6.0 μM colistin sulfate immediately before the compound of interest was added. Example 2—Calculation of Physiochemical Properties The findings presented here are congruent with what is known about β-lactams and explain why their spectrum could be broadened (e.g. penicillin G to ampicillin) where other classes could not. In both liposome swelling assays and whole cell studies, positive charge greatly accelerates penetration of β-lactams through porins, while negative charge and bulky substituents impede penetration. Indeed, early generation β-lactams lacking an amine, such as penicillin G, are inactive against Gram-negative bacteria. Our analysis shows penicillin G has flexibility/shape parameters (RB=4, Glob=0.17) that make it an outstanding candidate for conversion, and addition of an amine results in ampicillin (RB=4, Glob=0.12), which now meets all criteria for accumulation (structures inFIG.16). Although there are third and fourth generation β-lactams with Gram-negative activity that do not meet the guidelines outlined here, these β-lactams have greatly reduced accumulation in Gram-negative bacteria compared to their positively-charged analogues that meet the flexibility/shape parameters. However, the third and fourth generation 3-lactams are significantly more stable to β-lactamases than the early generation β-lactams, thus requiring lower levels of accumulation for antibacterial activity. As delineated herein, to favor Gram-negative accumulation, a primary amine should be embedded on a compound with proper flexibility and shape parameters; these results explain why simple addition of an amine has not been a generalizable strategy to increase Gram-negative accumulation and activity for other antibiotic classes. As one example from the macrolide class, there are no differences in the spectrum of activity observed for erythromycin versus 9(S)-erythromycylamine; as shown inFIG.16these compounds do not possess the appropriate RB and/or Glob parameters for accumulation. Datasets of chemical structures were created and managed using Canvas (Version 2.6, Schrödinger, LLC, New York, NY, 2015.). Initial structure preparation and 3D minimization was performed with LigPrep (Version 3.6, Schrödinger, LLC, New York, NY, 2015.) using OPLS_2005 force fields. Tautomeric and protonation states were determined using Epik (Version 3.4, Schrödinger, LLC, New York, NY, 2015) at pH 7.4. Generation of ensembles of conformations was performed using Conformational Search in MOE 2015.10 using the LowModeMD method with default settings. Physiochemical descriptors (297, both 2D- and 3D-based) were calculated using MOE for each conformation. Descriptors were averaged (unweighted mean) across all conformations for each molecule. Data with descriptors were used to train a random forest classification prediction model using the R package caret. Preprocessing of data removed descriptors with near zero variance or high co-correlation with other descriptors. An additional approximation of three-dimensionality, average distance to the plane of best fit (PBF), was calculated using a custom Python program. The PBF algorithm was implemented in Python with SDfile I/O and structure representation being handled by libraries from Schrodinger. For each compound, the PBF algorithm determines the plane that best fits a set of 3D coordinates that represent the positions of all heavy atoms in the molecule using single value decomposition. The distance of each heavy atom to this plane is measured in angstroms and averaged. This Python program was incorporated into the Maestro GUI for convenient use. ClogD7.4were calculated using the online compound property calculation software FAFdrugs. Example 3—Outer Membrane Protein Profiles As small molecules that traverse the outer membrane of Gram-negative bacteria predominately cross via porins, knocking out the major porins ofE. coliwould be expected to decrease compound accumulation. While there are many porins present inE. coli, the two major non-specific porins are OmpF and OmpC, which are differentially expressed (based on the extracellular osmolarity) by the EnvZ/OmpR two component regulatory system. A ΔompR strain ofE. colifrom the KEIO knockout collection was therefore chosen to effectively knockout both OmpF and OmpC (knockout was validated by analyzing the outer membrane proteins of the ΔompR strain compared to the parental strain, data not shown). For control high accumulators ciprofloxacin, tetracycline, and chloramphenicol, a significant decrease in accumulation is observed in the ΔompR strain ofE. colicompared to the parental strainE. coliBW25113 (FIG.17A), consistent with previous data demonstrating that these antibiotics enter through the OmpF and OmpC porins. Eight high accumulating compounds from the test set were also evaluated, and accumulation decreased in the ΔompR strain (FIG.17B), suggesting these porins are a major gateway to small molecule accumulation for the compounds tested. The accumulation of 6DNM-NH3 in the ΔompR strain was compared to accumulation in the WT strain ofE. coli. A decrease in accumulation is observed, suggesting that OmpF and/or OmpC contribute to 6DNM-NH3 accumulation (Extended DataFIG.17A). The method used to compare the outer membrane proteins ofE. coliBW25113 to outer membrane of the ΔompRE. colifrom the KEIO collection was adapted from Adler, M., Anjum, M., Andersson, D. I. & Sandegren, L. Influence of acquired beta-lactamases on the evolution of spontaneous carbapenem resistance inEscherichia coli. J. Antimicrob. Chemother.68, 51-59, (2013). Briefly, bacteria were grown to OD600=1.0 at 37° C. in LB broth. 4 mL were centrifuged for 10 min at 2,350 g at 4° C., washed with 1 mL of 100 mM Tris-HCl, pH 8.0, with 20% sucrose and incubated on ice for 10 min. Cells were pelleted as before and taken up in 1 mL of 100 mM Tris-HCl, pH 8.0, 20% sucrose containing 10 mM sodium ethylenediaminetetraacetate (EDTA). Lysozyme was added to a final concentration of 100 mg/mL and incubated on ice for 10 min. MgSO4was added to 20 mM final concentration and RNAseA and DNAseI were added to a final concentration of 10 mg/mL. Cells were disrupted with five freeze-thaw cycles in dry ice/ethanol and room temperature/water bath. A sixth freezing sample was left to thaw on ice for 2 hours. Membranes were pelleted for 25 min at 16,100 g at 4° C. The supernatants were discarded, and the pellet was washed and pelleted three times in 1 mL of 20 mM NaPO4, pH 7 and 0.5% sarkosyl. The protein extracts were taken up in 60 mL of Laemmli sample buffer (80 mM Tris-HCl, pH 6.8, 3% SDS, 10% glycerol, 5% β-mercaptoethanol, 0.02% bromophenol blue), boiled for 5 min and subjected to SDS-PAGE (12% polyacrylamide). Proteins were visualized via staining with Coomassie Blue-G. Example 4—Accumulation Analysis in Protoplasts A limitation to measuring accumulation in whole cells is that no distinction is made between periplasmic and cytoplasmic accumulation. To reach the cytoplasm ofE. colicompounds must also diffuse through the inner membrane, whose filtering properties may be different from the filtering properties of the outer membrane. To examine this, high accumulating compounds and some of their derivatives were tested for accumulation inE. coliprotoplasts, cells lacking the outer membrane and peptidoglycan. As shown inFIG.17B, minimal variation in accumulation was observed between compounds in this experiment, supporting the hypothesis that traversing the outer membrane is the main barrier to small molecule accumulation in Gram-negative bacteria. The method for preparing protoplasts was adapted from Weiss. 85 μL of an overnight culture ofE. coliMG1655 was diluted into 85 mL of fresh LB broth and grown at 37° C. with shaking to an OD600=1.0. The bacteria were pelleted at 3,220 rcf for 10 minutes at 4° C. and the supernatant was discarded. The pellet was washed 3 times with 10 mL of 10 mM Tris HCl buffer (pH 8), and the pellet was resuspended in 30 mL of 10 mM Tris HCl (pH 8) containing 0.5 M sucrose. Potassium ethylenediaminetetraacetate (EDTA, 0.5 M, pH 8.0) was added slowly over a period of 20 minutes to a final concentration of 0.01 M. The bacteria were shaken at 130 rpm for 20 minutes at 37° C., and then harvested as before. The supernatant was discarded and the pellets were washed two times with SMM buffer (0.5 M sucrose, 20 mM sodium maleate, 20 mM MgCl2, pH 6.5). The bacteria were then resuspended in 30 mL of SMM buffer, 30 mg of lysozyme was added, and the bacteria were shaken at 130 rpm for 1.5 hour at 37° C. The protoplasts were harvested by centrifuging at 2000 rcf for 20 minutes at 4° C. The protoplast pellet was resuspended in 20 mL of SMM buffer, and protoplast formation was confirmed by diluting an aliquot in water and observing a 3-fold decrease on OD600. To test accumulation, 500 μL aliquots containing 10 μM compound were shaken at 130 rpm for 5 minutes at 37° C. Samples were pelleted at 2000 rcf for 10 minutes at room temperature and the supernatants were discarded. The pellets were resuspended in 200 μL of water and incubated at room temperature for 5 minutes. The lysed protoplasts were pelleted by centrifuging at 21,130 rcf for 10 minutes, and the supernatant was analyzed by LC-MS/MS for compound concentration as before. Example 5—All-Atom Molecular Dynamics Simulations To better understand the observed accumulation trends, molecular modeling was performed on a subset of test set compounds and antibacterials as they traverse the bacterial porin OmpF. Steered molecular dynamics (SMD) simulations were performed such that molecules were pulled through the constriction site of OmpF. While it does not directly provide the free energy landscape, SMD is frequently employed to map the pathway for long time-scale processes and has been previously utilized to study OmpF. In repeated simulations molecules adopted similar pathways for traversing the porin while often hydrogen bonding with different hydrophilic residues (data not shown). The trajectory of high-accumulating compound 1 reveals a key interaction between the pendant amine and acidic residues (most often Asp113) that assisted in movement through the constriction site (FIG.19A). This finding is in accord with previous reports of the importance of Asp113 in producing the cation selectivity of OmpF. This interaction was absent in the trajectory of a low-accumulating analogue of 1 (amide 13) (FIG.18B), in agreement with accumulation data (FIG.5). Further, amide 13 induces greater distortions in constriction site residues as it is forced through the porin (data not shown). In addition, SMD simulations were performed with 6DNM and 6DNM-NH3 as these molecules move through the porin OmpF. Similar to SMD simulations inFIG.18A, the translocation of 6DNM-NH3 was assisted by a key interaction between the primary amine on 6DNM-NH3 and Asp113 (data not shown). 6DNM was incapable of this interaction and instead required distortion in Asp113 and neighboring residues to allow passage (data not shown). Trajectories from SMD clearly suggests 6DNM-NH3 and 6DNM proceed through significantly different pathways. The simulation model was constructed using CHARMM-GUI and comprised one OmpF monomer (PDB 3POX), 108 (90%) 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE) lipid molecules, 12 (10%) 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (POPG), and solvated with 8,234 water molecules in 150 mM NaCl (36 Na+and 15 Cl−) for a total of 45,402 atoms. Hexagonal periodic boundary conditions were applied with a distance of 77.3 Å in the XY-direction and 92.8 Å in the Z-direction. Electrostatic interactions were calculated using the Particle-Mesh Ewald (PME) method. Protein residues E296, D312, and D127 were protonated. The simulations were performed at constant pressure (1 atm) and temperature (303 K) with a time step of 2 fs. Each small molecule under investigation was manually placed directly above the pore. Restraints were initially applied to protein backbone and small molecule analyte atoms and then removed to equilibrate the system. For SMD production simulations, each small molecule analyte was pulled at the molecules center of mass (5 kcal/mol Å2) at a constant velocity (10 Å/ns) along the Z-axis for 4 ns. The all-atom CHARMM force field was used for protein and lipids. TIP3P was used for water. All the MD simulations were carried out using the NAMD 2.11 scalable molecular dynamics program and run on Stampede at TACC. CHARMM residue topology and parameter files for the small molecules were constructed using CGenFF. SMD trajectories were analyzed and visualized using VMD 1.9.2 and rendered using Pov-Ray 3.6. Example 6—Design of Novel Compounds The natural product deoxynybomycin (DNM) was selected as a good candidate for conversion to a broad-spectrum agent. DNM has antibacterial activity through inhibition of wild-type and mutant DNA gyrase, and DNM and its alkyl-chain derivatives are only active against Gram-positive bacteria. Chemoinformatic analysis shows that DNM has zero rotatable bonds and a Glob of 0.02, suggesting that the addition of an amine to a position that does not alter the DNM antibacterial activity would provide a derivative able to accumulate in and be active against Gram-negative pathogens. To facilitate construction of a derivative with a primary amine, an analogue of DNM was first synthesized where the five-membered ring in DNM is expanded to a six-membered ring, affording the compound 6DNM (structures in Table 1 ofFIG.19; all synthetic routes can be found in Example 6 below). Assessment with the accumulation assay reveals 6DNM has low accumulation inE. coli(298 nmol/1012CFUs) (Table 1 ofFIG.19). A derivative of 6DNM was then designed that maintains low RB and Glob, but that also contains a primary amine; this compound is named 6DNM-NH3 and its structure is shown in Table 1 ofFIG.19. Synthesis of 6DNM-NH3 followed by antibacterial evaluation reveals that this compound retains activity againstS. aureus, but it also accumulates inE. colito a high degree (1114 nmol/1012CFUs). Consistent with this enhanced accumulation, 6DNM-NH3 shows significant activity againstE. coliMG1655 (MIC=0.5 μg/mL). Similar to observed patterns of accumulation from the test compounds, when the amine of 6DNM-NH3 is acetylated or replaced with a carboxylic acid, the resulting compounds (6DNM-amide and 6DNM-acid) do not accumulate inE. coli(Table 1 ofFIG.19). Example 7—Antibiotic Susceptibility Tests 6DNM is active againstStaphylococcus aureus(MIC=0.06-1 μg/mL) but shows no activity versusE. coli(MIC>32 μg/mL, Table 1 ofFIG.19). Synthesis of 6DNM-NH3 followed by antibacterial evaluation reveals that this compound retains activity againstS. aureus, but it also accumulates inE. colito a high degree (1114 nmol/1012CFUs). Consistent with this enhanced accumulation, 6DNM-NH3 shows significant activity againstE. coliMG1655 (MIC=0.5 μg/mL). When the amine of 6DNM-NH3 is acetylated or replaced with a carboxylic acid, the resulting compounds (6DNM-amide and 6DNM-acid) show no activity againstE. coli(Table 1 ofFIG.19). 6DNM and its derivatives were further evaluated against an expanded panel of Gram-negative pathogens, laboratory strains and clinical isolates of ESKAPE pathogensE. coli, Acinetobacter baumannii, Klebsiella pneumoniae, Enterobacter cloacae, andPseudomonas aeruginosa(Table 1 ofFIG.19). While 6DNM possesses no notable activity against these Gram-negative organisms, 6DNM-NH3 has antibacterial activity against all of these Gram-negative pathogens exceptP. aeruginosa. Encouragingly, 6DNM-NH3 possesses activity against a multi-drug resistant New Delhi metallo-β-lactamase-1 producing strain ofE. coli(ATCC BAA-246948); this clinical isolate is highly resistant to ciprofloxacin (MIC>64 μg/mL, Table 1 ofFIG.19) and many other antibiotics but is killed by 6DNM-NH3 with an MIC of 4 μg/mL (Table 1 ofFIG.19). 6DNM-NH3 also maintains good activity against most of the other clinical isolates ofE. coli, A. baumannii, K. pneumonia, andE. cloacae(Table 1 ofFIG.19). Susceptibility testing was performed in triplicate, using the microdilution broth method as outlined by the Clinical and Laboratory Standards Institute. Mueller Hinton (MH) broth was used. Example 8—Synthesis of Novel Compounds General Reagent Information Chemical reagents were purchased from commercial sources and used without further purification. Anhydrous solvents used during these studies were dried after being passed through columns with activated alumina. Various 2-D NMR experiments were conducted as necessary.1H NMR and13C NMR experiments were recorded on Varian Unity spectrometers at either 400 MHz, 500 MHz, or 750 MHz, and 125 MHz or 188 MHz, respectively. Spectra were obtained in the following solvents (reference peaks also included for1H and13C NMRs): CDCl3(1H NMR: 7.26 ppm;13C NMR: 77.26 ppm), d6-acetone (1H NMR: 2.05 ppm;13C NMR: 206.26 ppm), CD3OD (1H NMR: 3.30 ppm;13C NMR: 49.00 ppm), d7-DMF (1H NMR: 2.75 ppm;13C NMR: 34.90 ppm) D2O (1H NMR: 4.79 ppm) d6-DMSO (1H NMR: 2.50 ppm;13C NMR: 39.52 ppm). NMR experiments were performed at room temperature unless otherwise indicated. Chemical shift values are reported in parts per million (ppm) for all1H NMR and13C NMR spectra.1H NMR multiplicities are reported as: s=singlet, d=doublet, t=triplet, q=quartet, m=multiplet, br=broad. Various novel compounds were synthesized. Exemplary Pleuromutilin Derivatives General Procedure A for Pleuromutilin Derivatives A solution of pleuromutilin (1.0 g, 2.6 mmol) in dichloromethane was cooled to −78° C. and a stream of ozone was passed through the reaction mixture until a blue color persisted (12 minutes). The reaction mixture was then purged with oxygen, dimethyl sulfide (0.57 mL, 7.8 mmol) was added, and the reaction was allowed to warm to room temperature while stirring for 24 hours. The crude reaction mixture was washed with brine, extracted with dichloromethane, and the combined organic layers were dried with magnesium sulfate and evaporated. Purification by flash chromatography (2:1 ethyl acetate:hexanes) provided precursor A to 4-16 (503 mg, 51%) as a white solid. Precursor A for 4-16 (450 mg, 1.18 mmol), hydroxylamine hydrochloride (164 mg, 2.36 mmol), and sodium acetate (387, 4.72 mmol) were dissolved in acetonitrile (11.8 mL) and water (2.95 mL) and heated to 50° C. in a sealed vial for 6 hours. The reaction mixture was then cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with magnesium sulfate, and evaporated. Purification by flash chromatography (2:1 ethyl acetate:hexanes) provided precursor B for 4-16 (425 mg, 91%) as a white solid. To a solution of precursor B for 4-16 (372 mg, 0.96) and ammonium acetate (296, 3.84 mmol) in methanol at room temperature was added sodium cyanoborohydride (241 mg, 3.84 mmol). The reaction mixture was then cooled to 0° C., a solution of titanium trichloride (˜10 wt. % TiCl3in 20-30 wt. % HCl, 0.96 mL) was then added dropwise, and the reaction was stirred for 24 hours while warming to room temperature. The reaction was quenched by addition of 2 M sodium hydroxide and extracted with dichloromethane. The combined organic layers were washed with brine, dried with magnesium sulfate, and evaporated. Purification by flash chromatography (15% methanol and 2% triethylamine in dichloromethane) provided amine 4-16 (134 mg, 36%) as a white solid. 1H NMR (CD3OD, 500 MHz): δ 5.60 (d, J=8.7 Hz, 1H), 4.06 (d, J=17.1 Hz, 1H), 3.98 (d, J=16.9 Hz, 1H), 3.53 (dd, J=15.8, 6.4 Hz, 1H), 3.24-3.16 (m, 1H), 2.78-2.73 (m, 1H), 2.45-2.37 (m, 1H), 1.96-1.86 (m, 2H), 1.82 (dq, J=13.8, 3.0 Hz, 1H), 1.71-1.47 (m, 3H), 1.43 (s, 3H), 1.40-1.35 (m, 1H), 1.27 (t, J=7.3 Hz, 1H), 1.22-1.14 (m, 2H), 1.13 (s, 3H), 1.12-1.07 (m, 2H), 0.98 (d, J=7.1 Hz, 3H), 0.97-0.93 (m, 2H), 0.72 (d, J=6.9 Hz, 3H). (33-nonexchangeable protons). 13C NMR (CD3OD, 125 MHz): δ 219.72, 173.80, 76.90, 70.38, 61.91, 58.32, 46.62, 45.50, 43.01, 42.85, 42.33, 37.99, 36.72, 31.54, 28.18, 25.99, 25.43, 18.38, 17.31, 15.16, 11.93. General Procedure B for Pleuromutilin Derivatives To a solution of pleuromutilin (757 mg, 2 mmol) and p-tosyl chloride (763 mg, 4 mmol) in methyl t-butyl ether (2 mL) and water (0.5 mL) was added a solution of 10 M sodium hydroxide (0.5 mL) dropwise. The reaction mixture was then heated to reflux for 1 hour, cooled to room temperature, diluted with water, and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with magnesium sulfate, and evaporated. Purification by flash chromatography (2:3 ethyl acetate:hexanes) provide the precursor to 4-57 (706 mg, 66%) as a white solid. To a solution of cysteamine hydrochloride (9 mg, 0.11 mmol) in ethanol (903 μL) at room temperature was added a solution of sodium ethoxide (97 μL, 0.26 mmol, 21% in ethanol) and the mixture was stirred for 30 minutes. The solution was then cooled to 0° C., the precursor to 4-57 (53 mg, 0.1 mmol) was added, and the reaction was stirred at 0° C. for 3.5 hours. The crude reaction mixture was then warmed to room temperature diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with magnesium sulfate, and evaporated. Purification by flash chromatography (10% methanol and 2% triethylamine in ethyl acetate) provided 4-57 (25 mg, 58%) as a white solid. 1H-NMR (CD3OD, 500 MHz): δ 6.32 (dd, J=17.5, 11.2 Hz, 1H), 5.74 (d, J=8.3 Hz, 1H), 5.35-5.03 (m, 2H), 3.50 (d, J=6.2 Hz, 1H), 3.29-3.19 (m, 2H), 2.97-2.89 (m, 2H), 2.78 (td, J=6.6, 3.1 Hz, 2H), 2.41-2.37 (m, 1H), 2.37-2.22 (m, 2H), 2.23-2.11 (m, 2H), 1.82 (dq, J=14.1, 2.9 Hz, 1H), 1.75-1.52 (m, 3H), 1.52-1.41 (m, 4H), 1.42-1.33 (m, 2H), 1.26-1.17 (m, 1H), 1.16 (s, 3H), 1.14-1.05 (m, 1H), 0.94 (d, J=7.0 Hz, 3H), 0.74 (d, J=6.7 Hz, 3H). (37 non-exchangeable protons). 13C-NMR (CD3OD, 125 MHz, 60° C.): δ 219.6, 170.7, 141.3, 116.5, 75.4, 71.3, 59.3, 46.8, 46.0, 45.3, 43.1, 40.6, 38.2, 37.7, 35.3, 34.9, 34.7, 31.5, 28.2, 28.0, 25.8, 17.1, 15.4, 11.8. HRMS(ESI): m/z calc. for C24H40NO4S [M+H]+: 438.2678, found: 438.2680. Exemplary Deoxynybomycin Derivatives General Procedure A for Deoxynybomycin Derivatives A flame-dried flask was charged with diazaanthracenol, prepared as previously described (45, 46), and N,N-dimethylformamide (35 mM) then warmed to 115° C. under an atmosphere of nitrogen. The resulting solution was treated sequentially with potassium carbonate (10.0 equiv) and appropriate dibromide (2.0 equiv). An additional equivalent of dibromide was added after 1 hour, 2 hours, and 3 hours. The reaction was monitored by thin layer chromatography (TLC) and upon completion (5-8 h) was cooled to room temperature then diluted with chloroform (30 mL) and water (50 mL). The aqueous layer was further extracted with chloroform (4×30 mL). The combined organic layers were dried over sodium sulfate and concentrated in vacuo. The resulting residue was purified by silica gel chromatography (90-100% ethyl acetate/hexanes). Synthesized from diazaanthrecol (30.6 mg, 0.113 mmol) and 1,2-dibromoethane according to General Procedure A to yield 6DNM as a white solid (26.4 mg, 79% yield). 1H NMR (CDCl3, 500 MHz): δ 7.52 (s, 1H), 6.52 (d, J=1.7 Hz, 2H), 4.38 (t, J=4.7 Hz, 2H), 4.27 (t, J=4.7 Hz, 2H), 3.91 (s, 3H), 2.49 (d, J=1.2 Hz, 3H), 2.5 (d, J=1.18 Hz, 3H). 13C NMR (CDCl3, 125 MHz): δ 163.36, 160.25, 146.52, 145.65, 131.11, 130.79, 127.72, 120.65, 119.72, 118.41, 116.77, 113.79, 63.59, 39.83, 35.29, 19.19, 18.97. HRMS (ESI): m/z calc. for C17H17N2O3 [M+H]+: 297.1239, found: 297.1236. Synthesized from diazaanthrecol (45 mg, 0.166 mmol) and benzyl (2,3-dibromopropyl)carbamate (according to General Procedure A to yield 6DNM-NHCbz as a white solid (30.1 mg, 39% yield). A flame-dried flask was charged with Pd(OAc)2(25.6 mg, 0.114 mmol) and dichloromethane (10 mL) under an atmosphere of nitrogen. The resulting solution was treated sequentially with Et3SiH (0.27 mL, 1.7 mmol) and triethylamine (32 μL, 0.23 mmol). After stirring for 15 min, 1.0 mL of the black solution was transferred to a solution of DNM-NHCbz (26.2 mg, 0.0570 mmol) in dichloromethane under an atmosphere of nitrogen. After stirring at room temperature for 24 hours, the reaction was quenched with methanol (2 mL) and saturated NH4OH (20 μL) and stirred for an additional 30 minutes. The solvent was removed in vacuo and the resulting residue was purified by silica gel chromatography (0-8% methanol/chloroform) to produce 6DNM-NH3as an off white solid (16.5 mg, 88%). A flame-dried flask was charged with 6DNM-NH3(10.0 mg, 0.0307 mmol) was dissolved in a mixture of dichloromethane (2 mL) and pyridine (2 mL) under an atmosphere of nitrogen. The resulting solution was treated with acetic anhydride (7.6 μL, 0.054 mmol.). After stirring at room temperature for 2 hours, the reaction as diluted with dichloromethane (25 mL) and 3 M HCl (25 mL). The layers were separated and the aqueous layer extracted with additional dichloromethane (2×25 mL). The combined organic layers were washed with brine (30 mL), dried over sodium sulfate and concentrated in vacuo. The resulting residue was purified by silica gel chromatography eluting (0-5% methanol/dichloromethane) to yield 6DNM-NHAc as an off-white solid (8.1 mg, 72%). Synthesized from diazaanthrecol (30.0 mg, 0.111 mmol) and ethyl 3,4-dibromobutanoate (53) according to General Procedure A to yield 6DNM-CO2Et as a white solid (31.0 mg, 73% yield). A flask was charged with 6DNM-CO2Et (31.0 mg, 0.0811 mmol) and methanol (6 mL). The resulting solution was treated with concentrated aqueous lithium hydroxide (0.60 mL) at room temperature. After stirring for 4 hours, the solvent was removed in vacuo and the resulting residue redissolved in water (30 mL) and passed through a syringe filter. The solution was acidified with 6 M HCl and extracted with chloroform (5×25 mL). The combined organic layers were dried over sodium sulfate and concentrated in vacuo to yield 6DNM-acid as a white solid (25.2 mg, 88%). INCORPORATION BY REFERENCE All of the U.S. patents and U.S. patent application publications cited herein are hereby incorporated by reference. EQUIVALENTS Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
130,280
11858947
REFERENCE NUMERALS 1: Substrate2: First Electrode3: Organic Material Layer4: Second Electrode5: Hole Injection Layer6: Hole Transfer Layer7: Electron Blocking Layer8: Light Emitting Layer9: Hole Blocking Layer10: Electron Injection and Transfer Layer DETAILED DESCRIPTION Hereinafter, the present specification will be described in more detail. One embodiment of the present specification provides a heterocyclic compound of Chemical Formula 1. In the present specification, a description of a certain part “including” certain constituents means capable of further including other constituents, and does not exclude other constituents unless particularly stated on the contrary. In the present specification, a description of one member being placed “on” another member includes not only a case of the one member adjoining the another member but a case of still another member being present between the two members. Examples of substituents in the present specification are described below, however, the substituents are not limited thereto. The term “substitution” means a hydrogen atom bonding to a carbon atom of a compound is changed to another substituent, and the position of substitution is not limited as long as it is a position at which a hydrogen atom is substituted, that is, a position at which a substituent can substitute, and when two or more substituents substitute, the two or more substituents can be the same as or different from each other. In the present specification, the term “substituted or unsubstituted” means being substituted with one, two or more substituents selected from the group consisting of deuterium; a nitrile group; a substituted or unsubstituted alkyl group; a substituted or unsubstituted cycloalkyl group; a substituted or unsubstituted silyl group; a substituted or unsubstituted aryl group; and a substituted or unsubstituted heterocyclic group, or being substituted with a substituent linking two or more substituents among the substituents illustrated above, or having no substituents. For example, “a substituent linking two or more substituents” can include an aryl group substituted with an aryl group, an aryl group substituted with a heteroaryl group, a heterocyclic group substituted with an aryl group, an aryl group substituted with an alkyl group, and the like. In the present specification, the alkyl group can be linear or branched, and although not particularly limited thereto, the number of carbon atoms is preferably from 1 to 30. Specific examples thereof include methyl, ethyl, propyl, n-propyl, isopropyl, butyl, n-butyl, isobutyl, tert-butyl, sec-butyl, 1-methyl-butyl, 1-ethyl-butyl, pentyl, n-pentyl, isopentyl, neopentyl, tert-pentyl, hexyl, n-hexyl, 1-methylpentyl, 2-methylpentyl, 4-methyl-2-pentyl, 3,3-dimethylbutyl, 2-ethylbutyl, heptyl, n-heptyl, 1-methyl-hexyl, cyclopentylmethyl, cyclohexylmethyl, octyl, n-octyl, tert-octyl, 1-methylheptyl, 2-ethylhexyl, 2-propylpentyl, n-nonyl, 2,2-dimethylheptyl, 1-ethyl-propyl, 1,1-dimethyl-propyl, isohexyl, 4-methylhexyl, 5-methyl-hexyl and the like, but are not limited thereto. In the present specification, specific examples of the silyl group can include a trimethylsilyl group, a triethylsilyl group, a t-butyldimethylsilyl group, a vinyldimethylsilyl group, a propyldimethylsilyl group, a triphenylsilyl group, a diphenylsilyl group, a phenylsilyl group and the like, but are not limited thereto. In the present specification, the aryl group is not particularly limited, but preferably has 6 to 30 carbon atoms, and the aryl group can be monocyclic or polycyclic. When the aryl group is a monocyclic aryl group, the number of carbon atoms is not particularly limited, but is preferably from 6 to 30. Specific examples of the monocyclic aryl group can include a phenyl group, a biphenyl group, a terphenyl group and the like, but are not limited thereto. When the aryl group is a polycyclic aryl group, the number of carbon atoms is not particularly limited, but is preferably from 10 to 30. Specific examples of the polycyclic aryl group can include a naphthyl group, an anthracenyl group, a phenanthryl group, a triphenylene group, a pyrenyl group, a phenalenyl group, a perylenyl group, a chrysenyl group, a fluorenyl group and the like, but are not limited thereto. In the present specification, the heterocyclic group includes one or more atoms that are not carbon, that is, heteroatoms, and specifically, the heteroatom can include one or more atoms selected from the group consisting of O, N, Se, S and the like. The number of carbon atoms is not particularly limited, but is preferably from 2 to 30, and the heterocyclic group can be monocyclic or polycyclic. Examples of the heterocyclic group can include a thiophene group, a furanyl group, a pyrrole group, an imidazolyl group, a triazolyl group, an oxazolyl group, an oxadiazolyl group, a pyridyl group, a bipyridyl group, a pyrimidyl group, a triazinyl group, a triazolyl group, an acridyl group, a pyridazinyl group, a pyrazinyl group, a quinolinyl group, a quinazolinyl group, a quinoxalinyl group, a phthalazinyl group, a pyridopyrimidyl group, a pyridopyrazinyl group, a pyrazinopyrazinyl group, an isoquinolinyl group, an indolyl group, a carbazolyl group, a benzoxazolyl group, a benzimidazolyl group, a benzothiazolyl group, a benzocarbazolyl group, a benzothiophene group, a dibenzothiophene group, a benzofuranyl group, a phenanthrolinyl group, an isoxazolyl group, a thiadiazolyl group, a phenothiazinyl group, a dibenzofuranyl group and the like, but are not limited thereto. According to one embodiment of the present specification, R1 to R4 are the same as or different from each other, and each independently is hydrogen, or bond to adjacent substituents to form an aromatic ring. According to one embodiment of the present specification, R1 to R4 bond to adjacent groups to form at least one substituted or unsubstituted aromatic ring, and substituents that do not form the ring are hydrogen. In the present specification, Chemical Formula 1 is any one of the following Chemical Formulae 1-1 to 1-3: In Chemical Formulae 1-1 to 1-3, R1 to R14, L and Ar have the same definitions as in Chemical Formula 1. According to one embodiment of the present specification, L is the following Chemical Formula 3 or Chemical Formula 4: In Chemical Formula 3 and Chemical Formula 4:X11 to X14 and X21 to X24 are the same as or different from each other, and are each N or C—Ar1;one of X11 to X14 bonds to N of Chemical Formula 1;one of X21 to X24 bonds to N of Chemical Formula 1;two or more of X11 to X14 are N;two or more of X21 to X24 are N;Y is O, S or C(CH3)2;Ar1 is a substituted or unsubstituted alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted heteroaryl group; andn and m are 0 or 1. According to one embodiment of the present specification, Chemical Formula 3 can be any one of the following Chemical Formula 3-1 to Chemical Formula 3-4: In Chemical Formulae 3-1 to 3-4, X11 to X14 have the same definitions as in Chemical Formula 3. According to one embodiment of the present specification, Chemical Formula 4 can be any one of the following Chemical Formula 4-1 or Chemical Formula 4-2: In Chemical Formulae 4-1 and 4-2, X21 to X24 and Y have the same definitions as in Chemical Formula 4. According to one embodiment of the present specification, L is any one selected from among the following substituents: One of the dotted lines bonds to N of Chemical Formula 1, and the other one bonds to Ar. According to one embodiment of the present specification, Ar is a substituted or unsubstituted alkyl group having 1 to 10 carbon atoms, a substituted or unsubstituted aryl group having 6 to 20 carbon atoms, or a substituted or unsubstituted heteroaryl group having 3 to 30 carbon atoms. According to one embodiment of the present specification, Ar is a substituted or unsubstituted aryl group having 6 to 20 carbon atoms, or a substituted or unsubstituted heteroaryl group having 3 to 30 carbon atoms. According to one embodiment of the present specification, Ar is a substituted or unsubstituted aryl group having 6 to 20 carbon atoms, or a heteroaryl group having 3 to 30 carbon atoms unsubstituted or substituted with an aryl group. According to one embodiment of the present specification, Ar is a phenyl group, a biphenyl group, a naphthyl group, a phenanthrene group, a triphenylene group, a dibenzofuran group, a dibenzothiophene group, or a carbazole group, and the phenyl group, the biphenyl group, the naphthyl group, the phenanthrene group, the triphenylene group, the dibenzofuran group, the dibenzothiophene group, or the carbazole group is unsubstituted or substituted with an alkyl group having 1 to 10 carbon atoms, an aryl group having 6 to 20 carbon atoms, or a heteroaryl group having 3 to 30 carbon atoms. According to one embodiment of the present specification, Ar is a phenyl group, a biphenyl group, a naphthyl group, a phenanthrene group, a triphenylene group, a dibenzofuran group, a dibenzothiophene group, or a carbazole group, and the phenyl group, the biphenyl group, the naphthyl group, the phenanthrene group, the triphenylene group, the dibenzofuran group, the dibenzothiophene group, or the carbazole group is unsubstituted or substituted with a phenyl group or a naphthyl group. According to another embodiment of the present specification, the heterocyclic compound of Chemical Formula 1 can be any one of the following compounds: One embodiment of the present specification provides an organic light emitting device including a first electrode; a second electrode provided opposite to the first electrode; and one, two or more organic material layers provided between the first electrode and the second electrode, wherein one or more layers of the organic material layers include the heterocyclic compound. According to one embodiment of the present disclosure, the organic material layer includes a light emitting layer, and the light emitting layer includes the heterocyclic compound. According to one embodiment of the present disclosure, the organic material layer includes a light emitting layer, and the light emitting layer includes the heterocyclic compound as a host. According to one embodiment of the present disclosure, the organic material layer includes a light emitting layer, and the light emitting layer includes the heterocyclic compound as a red host. According to one embodiment of the present disclosure, the light emitting layer further includes a dopant. According to one embodiment of the present disclosure, the light emitting layer includes a metal complex as the dopant. According to one embodiment of the present disclosure, the light emitting layer includes an iridium-based complex as the dopant. According to one embodiment of the present disclosure, the light emitting layer includes any one of the following compounds as the dopant: According to one embodiment of the present disclosure, the light emitting layer includes the host and the dopant in a weight ratio of 1:99 to 99:1. According to one embodiment of the present disclosure, the light emitting layer includes the host and the dopant in a weight ratio of 50:50 to 99:1. According to one embodiment of the present disclosure, the light emitting layer includes the host and the dopant in a weight ratio of 80:20 to 99:1. According to one embodiment of the present disclosure, the light emitting layer includes the host and the dopant in a weight ratio of 90:10 to 99:1. According to one embodiment of the present disclosure, the organic material layer includes an electron injection layer, an electron transfer layer, or an electron injection and transfer layer, and the electron injection layer, the electron transfer layer, or the electron injection and transfer layer includes the heterocyclic compound. According to one embodiment of the present disclosure, the organic material layer includes a hole injection layer, a hole transfer layer, or a hole injection and transfer layer, and the hole injection layer, the hole transfer layer, or the hole injection and transfer layer includes the heterocyclic compound. According to one embodiment of the present disclosure, the organic material layer includes an electron blocking layer or a hole blocking layer, and the electron blocking layer or the hole blocking layer includes the heterocyclic compound. For example, the organic light emitting device of the present disclosure can have structures as illustrated inFIGS.1and2, however, the structure is not limited thereto. FIG.1illustrates a structure of the organic light emitting device in which a first electrode (2), an organic material layer (3) and a second electrode (4) are consecutively laminated on a substrate (1). FIG.2illustrates a structure of the organic light emitting device in which a first electrode (2), a hole injection layer (5), a hole transfer layer (6), an electron blocking layer (7), a light emitting layer (8), a hole blocking layer (9), an electron injection and transfer layer (10) and a second electrode (4) are consecutively laminated on a substrate (1). FIG.2illustrates the organic light emitting device, and the structure is not limited thereto, and additional organic material layers can be further included between each layer. For example, the organic light emitting device according to the present disclosure can be manufactured by forming an anode on a substrate by depositing a metal, a metal oxide having conductivity, or an alloy thereof using a physical vapor deposition (PVD) method such as sputtering or e-beam evaporation, forming an organic material layer including a hole injection layer, a hole transfer layer, a light emitting layer or an electron transfer layer and an organic material layer including the heterocyclic compound of Chemical Formula 1 thereon, and then depositing a material usable as a cathode thereon. In addition to such a method, the organic light emitting device can also be manufactured by consecutively depositing a cathode material, an organic material layer and an anode material on a substrate. As the anode material, materials having large work function are normally preferred so that hole injection to an organic material layer is smooth. Specific examples of the anode material usable in the present disclosure include metals such as vanadium, chromium, copper, zinc and gold, or alloys thereof; metal oxides such as zinc oxide, indium oxide, indium tin oxide (ITO) and indium zinc oxide (IZO); combinations of metals and oxides such as ZnO:Al or SnO2:Sb; conductive polymers such as polypyrrole and polyaniline, but are not limited thereto. As the cathode material, materials having small work function are normally preferred so that electron injection to an organic material layer is smooth. Specific examples of the cathode material include metals such as magnesium, calcium, sodium, potassium, titanium, indium, yttrium, lithium, gadolinium, aluminum, silver, tin and lead, or alloys thereof; multilayer structure materials such as LiF/Al or LiO2/Al, and the like, but are not limited thereto. The hole injection material is a material favorably receiving holes from an anode at a low voltage, and the highest occupied molecular orbital (HOMO) of the hole injection material is preferably in between the work function of an anode material and the HOMO of surrounding organic material layers. Specific examples of the hole injection material include metal porphyrins, oligothiophene, arylamine-based organic materials, hexanitrile hexaazatriphenylene-based organic materials, quinacridone-based organic materials, perylene-based organic materials, anthraquinone, polyaniline, and polycompound-based conductive polymers, and the like, but are not limited thereto. The hole transfer material is a material capable of receiving holes from an anode or a hole injection layer, moving the holes to a light emitting layer, and materials having high mobility for the holes are suited. Specific examples thereof include arylamine-based organic materials, conductive polymers, block copolymers having conjugated parts and non-conjugated parts together, and the like, but are not limited thereto. The light emitting material is a material capable of emitting light in a visible region by receiving holes and electrons from a hole transfer layer and an electron transfer layer, respectively, and binding the holes and the electrons, and is preferably a material having favorable quantum efficiency for fluorescence or phosphorescence. Specific examples thereof include 8-hydroxy-quinoline aluminum complexes (Alq3); carbazole-based compounds; dimerized styryl compounds; BAlq; 10-hydroxybenzoquinoline-metal compounds; benzoxazole-, benzothiazole- and benzimidazole-based compounds; poly(p-phenylenevinylene) (PPV)-based polymers; spiro compounds; polyfluorene, rubrene, and the like, but are not limited thereto. When the organic light emitting device includes a plurality of organic material layers, the organic material layers can be formed with materials the same as or different from each other. The organic light emitting device of the present specification can be manufactured using materials and methods known in the art, except that one of more layers of the organic material layers are formed using the heterocyclic compound. The dopant material can include aromatic heterocyclic compounds, styrylamine compounds, boron complexes, fluoranthene compounds, metal complexes and the like. Specifically, the aromatic heterocyclic compound is a fused aromatic ring derivative having a substituted or unsubstituted arylamino group, and arylamino group-including pyrene, anthracene, chrysene, periflanthene and the like can be included. The styrylamine compound is a compound in which substituted or unsubstituted arylamine is substituted with at least one arylvinyl group, and one, two or more substituents selected from the group consisting of an aryl group, a silyl group, an alkyl group, a cycloalkyl group and an arylamino group can be substituted or unsubstituted. Specifically, styrylamine, styryldiamine, styryltriamine, styryltetramine and the like can be included, however, the styrylamine compound is not limited thereto. As the metal complex, iridium complexes, platinum complexes and the like can be included, however, the metal complex is not limited thereto. The electron transfer layer is a layer that receives electrons from an electron injection layer and transfers the electrons to a light emitting layer, and as the electron transfer material, materials capable of favorably receiving electrons from a cathode, moving the electrons to a light emitting layer, and having high mobility for the electrons are suited. Specific examples thereof include Al complexes of 8-hydroxyquinoline; complexes including Alq3; organic radical compounds; hydroxyflavon-metal complexes, and the like, but are not limited thereto. The electron transfer layer can be used together with any desired cathode material as used in the art. Particularly, examples of the suitable cathode material include common materials that have small work function, and in which an aluminum layer or a silver layer follows. Specifically, the cathode material includes cesium, barium, calcium, ytterbium and samarium, and in each case, an aluminum layer or a silver layer follows. The electron injection layer is a layer that injects electrons from an electrode, and as the electron injection material, compounds having an electron transferring ability, having an electron injection effect from a cathode, having an excellent electron injection effect for a light emitting layer or light emitting material, and preventing excitons generated in the light emitting layer from moving to a hole injection layer, and in addition thereto, having an excellent thin film forming ability are preferred. Specific examples thereof include fluorenone, anthraquinodimethane, diphenoquinone, thiopyran dioxide, oxazole, oxadiazole, triazole, imidazole, perylene tetracarboxylic acid, fluorenylidene methane, anthrone or the like, and derivatives thereof, metal complex compounds, nitrogen-containing 5-membered ring derivatives, and the like, but are not limited thereto. The metal complex compound includes 8-hydroxy-quinolinato lithium, bis(8-hydroxyquinolinato)zinc, bis(8-hydroxyquinolinato) copper, bis(8-hydroxyquinolinato)-manganese, tris(8-hydroxyquinolinato)aluminum, tris(2-methyl-8-hydroxyquinolinato)aluminum, tris(8-hydroxy-quinolinato)gallium, bis(10-hydroxybenzo[h]quinolinato)-beryllium, bis(10-hydroxybenzo[h]quinolinato)zinc, bis(2-methyl-8-quinolinato)chlorogallium, bis(2-methyl-8-quinolinato) (o-cresolato) gallium, bis(2-methyl-8-quinolinato) (1-naphtholato)aluminum, bis(2-methyl-8-quinolinato) (2-naphtholato)gallium and the like, but is not limited thereto. The hole blocking layer is a layer blocking holes from reaching a cathode, and can be generally formed under the same condition as the hole injection layer. Specific examples thereof can include oxadiazole derivatives, triazole derivatives, phenanthroline derivatives, BCP, aluminum complexes and the like, but are not limited thereto. The organic light emitting device according to the present specification can be a top-emission type, a bottom-emission type or a dual-emission type depending on the materials used. EXAMPLES The compounds of the present disclosure were prepared using, as a representative reaction, a Buchwald-Hartwig coupling reaction, a Suzuki coupling reaction or the like. The following is a preparation example for a representative structure in the disclosure of the present specification, and by varying substituents, all the compounds of the present specification can be prepared. Preparation Example 1 After dissolving 2-nitronaphthalen-1-yl trifluoromethane sulfonate (100.0 g, 1.0 eq.) and triphenylen-1-ylboronic acid (93.17 g, 1.1 eq.) in tetrahydrofuran (THF) (1000 ml), K2CO3(86.05 g, 2.0 eq.) dissolved in water (300 ml) was introduced thereto. Pd(t-Bu3P)2(1.59 g, 0.005 eq.) was introduced thereto, and the result was stirred under reflux. When the reaction was finished, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and treated with anhydrous magnesium sulfate. The result was vacuumed again to remove the solvent, and the result was column chromatographed to obtain Compound A-1 (88.28 g, yield 71%). [M+H]=400 Compound A-1 (88.28 g, 1.0 eq.) was introduced to triethylphosphite (200 mL), and the result was stirred under reflux. The reaction was terminated after 2 hours, and the reaction material was poured into ethanol (2 L) to precipitate solids. These solids were completely dissolved in CHCl3, washed with water, and treated with anhydrous magnesium sulfate. The solution was vacuum concentrated and purified using column chromatography to obtain Compound A (49.37 g, yield 61%). [M+H]=218 Preparation Example 2 Compound B was synthesized in the same manner as in the method for preparing Compound A except that 3-nitronaphthalen-2-yl trifluoromethane sulfonate was used instead of 2-nitronaphthalen-1-yl trifluoromethane sulfonate in Preparation Example 1. Preparation Example 3 Compound C was synthesized in the same manner as in the method for preparing Compound A except that 3-1-nitronaphthalen-2-yl trifluoromethane sulfonate was used instead of 2-nitronaphthalen-1-yl trifluoromethane sulfonate in Preparation Example 1. Synthesis Example Synthesis Example 1 Compound A (10.0 g, 1.0 eq.), 2-chloro-4-phenylbenzo[h]quinazoline (8.73 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 1 (12.35 g, yield 73%). [M+H]=622 Synthesis Example 2 Compound A (10.0 g, 1.0 eq.), 2-chloro-4-phenylbenzo[4,5]thieno[3,2-d]pyrimidine (8.88 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 2 (10.42 g, yield 61%). [M+H]=628 Synthesis Example 3 Compound A (10.0 g, 1.0 eq.), 2-([1,1′-biphenyl]-4-yl)-3-chloro-9,9-dimethyl-9H-indeno[1,2-b]pyrazine (11.46 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 3 (13.98 g, yield 72%). [M+H]=714 Synthesis Example 4 Compound A (10.0 g, 1.0 eq.), 3-chloro-1-(dibenzo[b,d]furan-1-yl)benzo[f]quinazoline (11.40 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 4 (13.17 g, yield 68%). [M+H]=712 Synthesis Example 5 Compound A (10.0 g, 1.0 eq.), 2-chloro-4-(dibenzo[b,d]furan-3-yl)-9,9-dimethyl-9H-indeno[2,1-d]pyrimidine (11.88 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 5 (12.87 g, yield 65%). [M+H]=728 Synthesis Example 6 Compound A (10.0 g, 1.0 eq.), 2-chloro-4-(dibenzo[b,d]thiophen-3-yl)benzofuro[3,2-d]pyrimidine (11.58 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 6 (13.08 g, yield 67%). [M+H]=718 Synthesis Example 7 Compound A (10.0 g, 1.0 eq.), 2-chloro-3-(9-phenyl-9H-carbazol-2-yl)benzofuro[2,3-b]pyrazine (13.34 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 7 (14.58 g, yield 69%). [M+H]=777 Synthesis Example 8 Compound B (10.0 g, 1.0 eq.), 2-chloro-3-(4-phenylnaphthalen-1-yl)benzofuro[2,3-b]pyrazine (12.18 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 8 (13.45 g, yield 67%). [M+H]=738 Synthesis Example 9 Compound B (10.0 g, 1.0 eq.), 2-chloro-4-(4-(naphthalen-1-yl)phenyl)benzo[h]quinazoline (12.48 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 9 (13.63 g, yield 61%). [M+H]=748 Synthesis Example 10 Compound B (10.0 g, 1.0 eq.), 2-chloro-3-(naphthalen-2-yl)benzo[f]quinoxaline (26.49 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 10 (13.34 g, yield 74%). [M+H]=672 Synthesis Example 11 Compound B (10.0 g, 1.0 eq.), 2-chloro-3-(phenanthren-9-yl)benzo[4,5]thieno[2,3-b]pyrazine (11.88 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 11 (14.46 g, yield 71%). [M+H]=728 Synthesis Example 12 Compound B (10.0 g, 1.0 eq.), 2-chloro-4-(dibenzo[b,d]-furan-3-yl)benzo[4,5]thieno[3,2-d]pyrimidine (11.58 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 12 (14.45 g, yield 74%). [M+H]=718 Synthesis Example 13 Compound B (10.0 g, 1.0 eq.), 2-chloro-4-(dibenzo[b,d]-thiophen-3-yl)-5,5-dimethyl-5H-indeno[1,2-d]pyrimidine (12.36 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 13 (14.77 g, yield 73%). [M+H]=744 Synthesis Example 14 Compound B (10.0 g, 1.0 eq.), 3-chloro-2-(dibenzo[b,d]-thiophen-2-yl)-9,9-dimethyl-9H-indeno[1,2-b]pyrazine (12.36 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 14 (14.17 g, yield 70%). [M+H]=744 Synthesis Example 15 Compound B (10.0 g, 1.0 eq.), 2-chloro-4-(dibenzo[b,d]-thiophen-4-yl)benzofuro[3,2-d]pyrimidine (11.58 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 15 (14.26 g, yield 73%). [M+H]=718 Synthesis Example 16 Compound B (10.0 g, 1.0 eq.), 2-chloro-5,5-dimethyl-4-(triphenylen-2-yl)-5H-indeno[1,2-d]pyrimidine (13.67 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 16 (15.65 g, yield 73%). [M+H]=788 Synthesis Example 17 Compound C (10.0 g, 1.0 eq.), 4-([1,1′:4′,1″-terphenyl]-4-yl)-2-chloro-9,9-dimethyl-9H-indeno[2,1-d]pyrimidine (13.74 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 17 (15.26 g, yield 71%). [M+H]=790 Synthesis Example 18 Compound C (10.0 g, 1.0 eq.), 2-chloro-4-(dibenzo[b,d]-furan-4-yl)benzo[4,5]thieno[2,3-d]pyrimidine (11.58 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 18 (12.11 g, yield 62%). [M+H]=718 Synthesis Example 19 Compound C (10.0 g, 1.0 eq.), 4-([1,1′:3′,1″-terphenyl]-5′-yl)-2-chlorobenzofuro[2,3-d]pyrimidine (12.95 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 19 (15.17 g, yield 73%). [M+H]=764 Synthesis Example 20 Compound C (10.0 g, 1.0 eq.), 3-chloro-1-(9-phenyl-9H-carbazol-4-yl)benzo[f]quinazoline (13.64 g, 1.1 eq.), Pd(t-Bu3P)2(0.13 g, 0.01 eq.) and K3PO4(11.55 g, 2.0 eq.) were introduced to xylene (250 ml), and the result was stirred under reflux. When the reaction was terminated after 2 hours, the result was vacuumed to remove the solvent. After that, the result was completely dissolved in CHCl3, washed with water, and vacuumed again to remove approximately 50% of the solvent. Under reflux again, crystals were precipitated while adding ethyl acetate thereto, and cooled and then filtered. The result was column chromatographed to obtain Compound 20 (14.99 g, yield 70%). [M+H]=787 Experimental Example Comparative Example 1 A glass substrate on which indium tin oxide (ITO) was coated as a thin film to a thickness of 1,000 Å was placed in distilled water in which detergent was dissolved and ultrasonically cleaned. A product of Fischer Co. was used as the detergent, and as the distilled water, distilled water filtered twice with a filter manufactured by Millipore Co. was used. After the ITO was cleaned for 30 minutes, ultrasonic cleaning was repeated twice using distilled water for 10 minutes. After the cleaning with distilled water was finished, the substrate was ultrasonically cleaned with solvents of isopropyl alcohol, acetone and methanol, then dried, and then transferred to a plasma cleaner. The substrate was cleaned for 5 minutes using oxygen plasma, and then transferred to a vacuum depositor. On the transparent ITO electrode prepared as above, the following HI-1 compound was formed to a thickness of 1150 Å as a hole injection layer, and the following D-1 compound was p-doped thereto in a concentration of 1.5%. On the hole injection layer, a hole transfer layer having a film thickness of 800 Å was formed by vacuum depositing the following HT-1 compound. Subsequently, an electron blocking layer was formed to a film thickness of 150 Å on the hole transfer layer by vacuum depositing the following EB-1 compound. Then, a red light emitting layer having a thickness of 400 Å was formed on the EB-1 deposited film by vacuum depositing the following RH-1 compound and the following Dp-7 compound in a weight ratio of 98:2. On the light emitting layer, a hole blocking layer was famed to a film thickness of 30 Å by vacuum depositing the following HB-1 compound. Subsequently, an electron injection and transfer layer was formed to a thickness of 300 Å on the hole blocking layer by vacuum depositing the following ET-1 compound and the following LiQ compound in a weight ratio of 2:1. On the electron injection and transfer layer, a cathode was formed by consecutively depositing lithium fluoride (LiF) to a thickness of 12 Å and aluminum to a thickness of 1,000 Å. In the above-mentioned process, the deposition rates of the organic materials were maintained at 0.4 Å/sec to 0.7 Å/sec, the deposition rates of the lithium fluoride and the aluminum of the cathode were maintained at 0.3 Å/sec and 2 Å/sec, respectively, and the degree of vacuum during the deposition was maintained at 2×10−7torr to 5×10−6torr to manufacture an organic light emitting device. Comparative Example Compound Example 1 to Example 20 Organic light emitting devices were manufactured in the same manner as in Comparative Example 1 except that compounds described in the following Table 1 were used instead of RH-1 in the organic light emitting device of Comparative Example 1. Comparative Example 2 to Comparative Example 16 Organic light emitting devices were manufactured in the same manner as in Comparative Example 1 except that compounds described in the following Table 1 were used instead of RH-1 in the organic light emitting device of Comparative Example 1. When applying a current to each of the organic light emitting devices manufactured in Example 1 to Example 20, and Comparative Example 1 to Comparative Example 16, voltage, efficiency and lifetime were measured, and the results are shown in the following Table 1. T95 means time taken for luminance decreasing to 95% with respect to initial luminance (10,000 nit). TABLE 1LightDrivingEfficiencyLifetimeEmissionCategoryMaterialVoltage (V)(cd/A)T95 (hr)ColorComparativeRH-14.4834.7174RedExample 1Example 1Compound 13.9537.5213RedExample 2Compound 23.9343.1295RedExample 3Compound 34.2338.8191RedExample 4Compound 44.1739.7234RedExample 5Compound 54.0337.6217RedExample 6Compound 64.0539.9203RedExample 7Compound 73.9738.5199RedExample 8Compound 84.1742.1223RedExample 9Compound 94.1038.9221RedExample 10Compound 104.2734.3195RedExample 11Compound 114.1538.0190RedExample 12Compound 124.0137.5214RedExample 13Compound 133.9239.4199RedExample 14Compound 143.8437.5205RedExample 15Compound 154.1338.1239RedExample 16Compound 163.8538.5193RedExample 17Compound 174.3137.3229RedExample 18Compound 184.2136.5231RedExample 19Compound 193.9336.7251RedExample 20Compound 204.0437.0217RedComparativeC-14.6131.7197RedExample 2ComparativeC-24.2334.579RedExample 3ComparativeC-34.3533.5188RedExample 4ComparativeC-44.5332.8134RedExample 5ComparativeC-54.5032.1142RedExample 6ComparativeC-64.6433.1195RedExample 7ComparativeC-74.5934.7177RedExample 8ComparativeC-84.6931.5181RedExample 9ComparativeC-94.3033.961RedExample 10ComparativeC-104.3533.593RedExample 11ComparativeC-114.5529.447RedExample 12ComparativeC-124.1531.553RedExample 13ComparativeC-134.0434.573RedExample 14ComparativeC-144.3732.367RedExample 15ComparativeC-154.5523.131RedExample 16 When applying a current to each of the organic light emitting devices manufactured in Examples 1 to 20 and Comparative Examples 1 to 16, results of Table 1 were obtained. In the red organic light emitting device of Comparative Example 1, materials widely used in the art were used. In Comparative Examples 2 to 16, organic light emitting devices were manufactured using C-1 to C-15 instead of RH-1. Based on the results of Table 1, a driving voltage decreased by up to almost 20% when using the compounds of the present disclosure as a host of a red light emitting layer compared to the materials of the comparative examples, and efficiency also increased by 20% or greater. Based on such results, it was seen that energy was favorably transferred from the host to the red dopant. In addition, it was seen that lifetime properties were significantly improved by 1.5 times or greater while maintaining high efficiency. This can be considered to be due to the fact that the compounds of the present disclosure have higher stability for electrons and holes compared to the compounds of the comparative examples, and electron migration and hole migration are well balanced in the red OLED device. In conclusion, it was identified that using the compounds of the present disclosure as a host of a red light emitting layer was capable of improving driving voltage, light emission efficiency and lifetime properties of an organic light emitting device.
44,213
11858948
DESCRIPTION OF THE INVENTION The invention relates to a novel and very efficient catalyst to be used in the very challenging hydrogenation, in particular for the hydrogenation of esters, hindered ketones or thermo sensitive ketones. So, a first object of the present invention is a process for the reduction by hydrogenation, using molecular H2, of a C3-C70substrate containing one or two ketones, aldehydes, esters, or lactones functional groups into the corresponding alcohol, or diol, characterized in that said process is carried out in the presence of a base and at least one catalyst or pre-catalyst containing a ruthenium and a tetradentate ligand of formula wherein one dotted line indicates a single bond and the other dotted line a single or a double bond, z is 1 when both dotted lines is a single bond (i.e. the nitrogen atom belongs to an amino group) or is 0 when one dotted line is a double bond and the other a single bond (i.e. the nitrogen atom belongs to an imino group);m is 0 or 1; n is a integer between 0 and 4;q is 0 when the dotted line between N and C(R9)(R10) indicates a double bond or is 1 when the dotted line between N and C(R9)(R10) indicates a single bond;q′ is 0 when the dotted line between N and C(R11)(R12) indicates a double bond or is 1 when the dotted line between N and C(R11)(R12) indicates a single bond;R1and R2, when taken separately, represent, simultaneously or independently, a linear C1to C8alkyl group optionally substituted, a linear C2to C8alkenyl group optionally substituted, a linear, branched or cyclic C3to C8alkyl or alkenyl group optionally substituted, a C6to C10aromatic group optionally substituted, or an OR1′or NR1′R2′group, R1′and R2′being a C1to C8alkyl group or a C2to C8alkenyl group; or R1and R2, when taken together, form a saturated or unsaturated ring optionally substituted, having 4 to 10 atoms and including the phosphorus atom to which said R1and R2groups are bonded;R3, R4, R5, R6, R7, R8, R9, R10, R11and R12, taken separately, represent, simultaneously or independently, a hydrogen atom, a C1-C10linear alkyl group optionally substituted, a C2-C10linear alkenyl group optionally substituted, a C3-C10linear, branched or cyclic alkyl or alkenyl group optionally substituted or a C6to C10aromatic group optionally substituted;or R3and R4and/or R4and R5and/or R5and R6and/or R6and R7and/or R7and R8and/or R8and R9and/or R9and R10and/or R9and R11and/or R11and R12, when taken together, form a saturated or unsaturated ring optionally substituted, having 4 to 10 atoms;R13and R14when taken separately, represent, simultaneously or independently, a linear C1to C8alkyl group optionally substituted, a linear C2to C8alkenyl group optionally substituted, a linear, branched or cyclic C3to C8alkyl or alkenyl group optionally substituted, a C6to C10aromatic group optionally substituted, or an OR1′or NR1′R2′group, R1′and R2′being a C1to C8alkyl group or a C2to C8alkenyl group; or R13andR14, when taken together, form a saturated or unsaturated ring optionally substituted, having 4 to 10 atoms and including the phosphorus atom to which said R13and R14groups are bonded; andR15, when taken separately, represent, simultaneously or independently, a hydrogen atom, a halogen atom, a linear C1to C8alkyl group optionally substituted, a linear C2to C8alkenyl group optionally substituted, a linear, branched or cyclic C3to C8alkyl or alkenyl group optionally substituted, or a halo- or perhalo-hydrocarbon, CN, SO3R3′, SO2R3′, NO2, OR3′, or CONR3′R4′group, R3′and R4′, independently from each other, being a hydrogen atom or a C1to C8alkyl group or a C2to C8alkenyl group; two adjacent R15groups can be bonded together to form a C5to C10ring optionally substituted. R15may be, relative to the phosphine substituent, an ortho, a meta, a para substituent of the aromatic ring. According to a particular embodiment of the invention, the substrate can be a compound of formula (I) Wherein p is 0 or 1; when p is 1, Raand Rbrepresent, simultaneously or independently, a linear, branched or cyclic C1-C30aromatic, alkyl or alkenyl group, optionally substituted; orwhen p is 0, Rarepresents a linear, branched or cyclic C1-C30aromatic, alkyl or alkenyl group, optionally substituted and Rbrepresents a hydrogen atom, a linear, branched or cyclic C1-C30aromatic, alkyl or alkenyl group, optionally substituted; orRaand Rbare bonded together and form a C4-C20saturated or unsaturated group, optionally substituted. When p is 1, the corresponding alcohols (i.e (II-a) and (II-b)), or the corresponding diol (II′), of said substrate (I), are of formula wherein Raand Rbare defined as in formula (I). A compound of formula (II) (i.e. II-a or II-b) will be obtained in the case where Raand Rbare not bonded together, while a compound of formula (II′) will be obtained in the case where Raand Rbare bonded together. When p is 0, the corresponding alcohols of said substrate (I) are of formula wherein Raand Rbare defined as in formula (I). It is understood that by “a linear, branched or cyclic . . . aromatic, alkyl, or alkenyl group” it is meant that said Raor Rbcan be in the form of, e.g., a linear alkyl group or can also be in the form of a mixture of said type of groups, e.g. a specific Ramay comprises a linear alkyl, a branched alkenyl, a (poly)cyclic alkyl and an aryl moiety, unless a specific limitation to only one type is mentioned. Similarly, in all the below embodiments of the invention when a group is mentioned as being in the form of more than one type of topology (e.g. linear, cyclic or branched) and/or unsaturation (e.g. alkyl, aromatic or alkenyl) it is meant also a group which may comprise moieties having any one of said topologies or unsaturations, as above explained. According to a further embodiment of the invention, the substrate is a ketone, an aldehyde, an ester, or a lactone that will provide an alcohol or a diol, which is useful in the pharmaceutical, agrochemical or perfumery industry as final product or as an intermediate. Particularly preferred substrate is a ketone, an aldehyde, an ester, or a lactone that will provide an alcohol or diol, which is useful in the perfumery industry as final product or as an intermediate. Even a more particularly preferred substrate is an ester, or a lactone that will provide an alcohol or diol, which is useful in the perfumery industry as final product or as an intermediate. A particular embodiment of the invention's process is shown in Scheme 1: According to any one of the above embodiments of the invention, p is 0 or 1. Preferably p is 1. According to any one of the above embodiments of the invention, the substrate is a C5-C30compound of formula (I), and in particular one may cite those wherein Raand Rbrepresent simultaneously or independently a linear C1-C30alkyl group optionally substituted, a branched or cyclic C3-C30alkyl or alkenyl group optionally substituted or a C5-C30aromatic group optionally substituted; or Raand Rbare bonded together and form a C4-C20saturated or unsaturated linear, branched, mono-, di- or tri-cyclic group, optionally substituted. According to a further embodiment of the invention the substrate is a C5-C20compound of formula (I), wherein Raand Rbrepresent simultaneously or independently a linear, branched or cyclic C5-C18aromatic or alkyl group, optionally substituted, or a cyclic C5-C18alkenyl group, optionally substituted; or Raand Rbare bonded together and form a C4-C20saturated or unsaturated linear, branched, mono-, di- or tri-cyclic group, optionally substituted. Furthermore, according to a yet further embodiment, when Raand/or Rbrepresent an alkenyl group then the carbon-carbon double bond is not terminal and is not conjugated. Possible substituents of Raand Rbare one, two or three halogen, ORc, NRc2or Rcgroups, in which Rcis a hydrogen atom, a halogenated C1-C2group or a C1to C10cyclic, linear or branched alkyl, or alkenyl group, preferably a C1to C4linear or branched alkyl or alkenyl group. As other possible substituents one may also cite a group COORc, which can also be reduced to the corresponding alcohol during the invention's process, according to the molar amount of H2used, as well known by a person skilled in the art. Non-limiting examples of substrates are alkyl cinnamates, sorbates or salycilates, alkyl esters of natural (fatty or not) acids, Sclareolide, spirolactones, allylic ester, di alkyl diesters, (un)substituted benzoic esters, and unsaturated esters such as 13-7 unsaturated esters. In particular, the substrate can be selected from the group consisting of sclareolide, C9-C15spirolactones and C1-C4alkyl esters of 4-methyl-6-(2,6,6-trimethyl-1-cyclohexen-1-yl)-3-hexenoic acid. One can also cite the di alkyl esters of 1,4-dicarboxylate-cyclohexane, the di C1-5alkyl esters of the C2-10alkanediyl-dicarboxylates, C1-5alkyl cyclopropanecarboxylates, mono-, di- or tri-methoxybenzoic esters. The process of the invention is characterized by the use, as catalyst or pre-catalyst (hereinafter referred to as complexes unless specified otherwise), of a ruthenium complex as described above. The complex can be in the form of an ionic or neutral species. According to an embodiment of the invention, the ruthenium complex can be of the general formula wherein L represents a tetradentate ligand as defined above; andeach Y represents, simultaneously or independently, CO, a hydrogen or halogen atom, a hydroxyl group, or a C1-C6alkyl, alkenyl, alkoxy or carboxylic radical, or also a BH4or AlH4group;X represents a C3-C30mono-phosphine or a solvent.Z represents a non-coordinated anion; andn is 0, 1 or 2. In a particular embodiment of the invention, in formula (1), (2) or (3), each Y represents, simultaneously or independently, a hydrogen or chlorine atom, a hydroxy radical, a C1to C6alkoxy radical, such as a methoxy, ethoxy or isopropoxy radical, or a C1to C6acyloxy radical such as a CH3COO, CH3CH2COO or (CH3)3CCOO radical. More preferably, each Y represents, simultaneously or independently, a hydrogen or chlorine atom, a methoxy, ethoxy or isopropoxy radical, or a CH3COO, CH3CH2COO or (CH3)3CCOO radical. In a particular embodiment of the invention, in formula (2), the tetradendate ligand L is partly coordinated to a metal; i.e. only 3 atoms are coordinated to the Metal. When complex of formula (2) is used, the complex of formula (1) is formed in situ under the reaction conditions. In a particular embodiment of the invention, in formula (2) or (3), X represents a mono-phosphine of formula PRd3, wherein Rdis a C1-C12group, such as linear, branched or cyclic alkyl, alkoxy or aryloxy group optionally substituted, substituted or unsubstituted phenyl, diphenyl or naphthyl or di-naphthyl group. More particularly Rdmay represent a substituted or unsubstituted phenyl, diphenyl or naphthyl or di-naphthyl group. Possible substituents are those cited below for the various groups R1to R15. Preferably, X is a triphenylphosphine. In formula (3), X may also be a solvent, the term “solvent” has to be understood according to the usual meaning in the art and includes compounds used as diluent in the preparation of the complex or during the invention's process, non-limiting examples are dimethylsulfoxide, acetonitrile, dimethylformamide, an alcohol (e.g. an C1-C4alcohol), or also THF, acetone, pyridine or a C3-C8ester or the substrate of the invention's process. In a particular embodiment of the invention, in formula (3), Z represents a halogen atom, a hydroxyl group, or a C1-C6alkoxy, phenoxy or carboxylic radical. The complex of formula (1) represents, in general for practical reasons, a preferred embodiment of the invention. Possible substituents of the various groups R1to R15are one or two halogen atoms, C1to C10alkoxy, polyalkyleneglycols, halo- or perhalo-hydrocarbon, COOR, or R groups, wherein R is a C1to C6alkyl, or a C5to C12cycloalkyl, aralkyl (such as benzyl, phenethyl etc.) or aromatic group, the latter being also optionally substituted by one, two or three halogen atoms or C1-C8alkyl, alkoxy, nitro, sulfonates, halo- or perhalo-hydrocarbon or ester groups. By “halo- or perhalo-hydrocarbon” it is meant groups such as CF3or CClH2for instance. Preferably, said substituents can be, and in particular when said groups are or contain phenyl groups, one or two halogen atoms, one or two C1to C5alkoxy or polyalkyleneglycols groups, COOR or R groups wherein R is a C1to C4alkyl, or a C5-6cycloalkyl, aralkyl or aromatic group, the latter being also optionally substituted as above defined. Alternatively, possible substituents of R3, R4, R5, R6, R7, R8, R9, R10, R11and R12are one or two halogen atoms or R16or OR16groups wherein R16being a C1to C6alkyl groups or a C1to C4alkyl groups. According to a particular embodiment of the invention, m is 1. In other words, L can be a compound of formula wherein the dotted lines, z, n, q, q′, R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14and R15have the same meaning as above. According to a particular embodiment of the invention, m is 0. In other words, L can be a compound of formula Wherein the dotted line, z, n, q, R1, R2, R3, R4, R5, R6, R9, R10, R11, R12, R13, R14and R15have the same meaning as above. According to a particular embodiment of the invention, L can be a compound of formula wherein the dotted line, z, n, R1, R2, R9, R13, R14and R15have the same meaning as above. According to a particular embodiment of the invention, L can be a compound of formula wherein the dotted line, z, n, R1, R2, R12, R13, R14and R15have the same meaning as above. According to any one of the above embodiments of the invention, R1and R2may represent, when taken separately, simultaneously or independently, a linear C1to C8alkyl group optionally substituted, a linear C2to C8alkenyl group optionally substituted, a branched or cyclic C3to C8alkyl or alkenyl group optionally substituted, a C6to C10aromatic group optionally substituted; or R1and R2, when taken together, may form a saturated or unsaturated ring optionally substituted, having 4 to 10 atoms and including the phosphorus atom to which said R1and R2groups are bonded. Preferably, R1and R2may represent, when taken separately, simultaneously or independently, a linear C1to C6alkyl group optionally substituted, a branched or cyclic C3to C6alkyl group optionally substituted, a phenyl group optionally substituted; or R1and R2, when taken together, may form a saturated or unsaturated ring optionally substituted, having 4, 5, 6 or 7 carbon atoms and including the phosphorus atom to which said R1and R2groups are bonded. Preferably, R1and R2may represent a linear C1to C6alkyl group optionally substituted, a branched or cyclic C3to C6alkyl group optionally substituted or a phenyl group optionally substituted. Preferably, R1and R2may represent a linear C1to C6alkyl group, a branched or cyclic C3to C6alkyl group or a phenyl group. Even more preferably, R1and R2may represent a cyclohexyl, a phenyl, a tert-butyl, an iso-propyl or an ethyl group. Even more preferably, R1and R2may represent a phenyl or a tert-butyl group. According to any one of the above embodiments of the invention, R13and R14, when taken separately, represent, simultaneously or independently, a C6to C10aromatic group optionally substituted or an OR1′or NR1′R2′group wherein R1′and R2′is a C1to C8alkyl group or a C2to C8alkenyl group. Preferably, R13and R14may represent phenyl group optionally substituted. Even more preferably, R13and R14may represent phenyl group substituted with at least one halogen atom, halo- or perhalo-hydrocarbon or R group wherein R is a C1to C4alkyl, or a C5-6cycloalkyl, aralkyl or aromatic group, the latter being also optionally substituted as above defined. According to any one of the above embodiments of the invention, R3, R4, R5, R6, R7, R8, R9, R10, R11and R12, taken separately, represent, simultaneously or independently, a hydrogen atom, a C1-C6linear alkyl group optionally substituted, a C2-C6linear alkenyl group optionally substituted, a C3-C6branched or cyclic alkyl or alkenyl group optionally substituted or a C6to C10aromatic group optionally substituted; or R4and R5and/or R5and R6and/or R8and R9, when taken together, form a saturated or unsaturated ring optionally substituted, having 4 to 10 atoms; or R9and R11, when taken together, form a saturated or unsaturated non aromatic ring optionally substituted, having 4 to 10 atoms. Preferably, R3, R4, R5, R6, R7, R8, R9, R10, R11, and R12, taken separately, may represent, simultaneously or independently, a hydrogen atom, a C1-C4linear alkyl group optionally substituted, a C5-C6branched or cyclic alkyl group optionally substituted or a phenyl group optionally substituted; R4and R5or R5and R6or R8and R9, when taken together, form a saturated or unsaturated ring optionally substituted, having 4 to 7 carbon atoms. Preferably, R3, R4, R5, R6, R7, R8, R9, R11and R12may represent a hydrogen atom, a methyl or a phenyl group. Even more preferably, R3, R4, R5, R6, R7, R8, R9, R11and R12may represent a hydrogen atom According to any one of the above embodiments of the invention, n may be 0, 1 or 2. Preferably, n is 0 or 1. When n is 1, R15may be, relative to the phosphine substituent, a meta or a para substituent of the aromatic ring. Preferably, R15may be, relative to the phosphine substituent, a para substituent of the aromatic ring. According to any one of the above embodiments of the invention, R15, when taken separately, may represent, simultaneously or independently, a halogen atom, a linear C1to C4alkyl group optionally substituted, a linear C2to C5alkenyl group optionally substituted, a linear, branched or cyclic C3to C8alkyl or alkenyl group optionally substituted, or a halo- or perhalo-hydrocarbon group. Preferably, may represent, simultaneously or independently, a halogen atom, or a halo- or perhalo-hydrocarbon group such as CF3. Examples of suitable ligands includes, but are not limited to, 1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)methanimine, 2-(diphenylphosphaneyl)-N-((6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methyl)aniline, 2-(diphenylphosphaneyl)-N-(1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)ethyl)aniline, 1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)ethan-1-imine, 1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)-1-phenylmethanimine, 2-(diphenylphosphaneyl)-N-((6-((diphenylphosphaneyl)methyl)pyridin-2-yl) (phenyl)methyl) aniline, 2-(diphenylphosphaneyl)-N-((6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methyl)-5-(trifluoromethyl)aniline, N-(2-(diphenylphosphaneyl)-5-(trifluoromethyl)phenyl)-1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methanimine, N-(5-chloro-2-(diphenylphosphaneyl)phenyl)-1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methanimine, 5-chloro-2-(diphenylphosphaneyl)-N-((6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methyl)aniline, 4-chloro-2-(diphenylphosphaneyl)-N-((6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methyl)aniline, N-(4-chloro-2-(diphenylphosphaneyl)phenyl)-1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methanimine, N-(2-(diphenylphosphaneyl)-4-(trifluoromethyl)phenyl)-1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methanimine, 2-(diphenylphosphaneyl)-N-((6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methyl)-4-(trifluoromethyl)aniline, N-((6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl)-4-(trifluoromethyl)aniline, 1-(6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)-4-(trifluoromethyl)phenyl)methanimine, N-(4-chloro-2-(diphenylphosphaneyl)phenyl)-1-(6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)methanimine, 4-chloro-N-((6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl)aniline, 5-chloro-N-((6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl)aniline, N-(5-chloro-2-(diphenylphosphaneyl)phenyl)-1-(6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)methanimine, 1-(6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)-5-(trifluoromethyl)phenyl)methanimine, ((6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl)-5-(trifluoromethyl)aniline, ((6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl)-5-(trifluoromethyl)aniline, 1-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)-5-(trifluoromethyl)phenyl)methanimine, N-(5-chloro-2-(diphenylphosphaneyl)phenyl)-1-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)methanimine, 5-chloro-N-((6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl)aniline, N-(4-chloro-2-(diphenylphosphaneyl)phenyl)-1-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)methanimine, 4-chloro-N-((6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl)aniline, N-((6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl)-4-(trifluoromethyl)aniline, 1-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)-4-(trifluoromethyl)phenyl)methanimine, 1-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)ethan-1-imine, N-(1-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)ethyl)-2-(diphenylphosphaneyl) aniline, N-((6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl) (phenyl)methyl)-2-(diphenylphosphaneyl)aniline, 1-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)-1-phenylmethanimine, 1-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)methanimine, N-((6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl) aniline, N-((6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl) aniline, 1-(6-((diisopropylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)methanimine, 1-(6-((diethylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)methanimine, N-((6-((diethylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl) aniline, 1-(6-((dicyclohexylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)phenyl)methanimine, N-((6-((dicyclohexylphosphaneyl)methyl)pyridin-2-yl)methyl)-2-(diphenylphosphaneyl) aniline, N-(2-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)ethyl)-1-(2-(diphenylphosphaneyl)phenyl)methanimine, N-(2-(diphenylphosphaneyl)benzyl)-2-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)ethan-1-amine, 2-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)-N-(2-(diphenylphosphaneyl)benzyl)ethan-1-amine or N-(2-(6-((di-tert-butylphosphaneyl)methyl)pyridin-2-yl)ethyl)-1-(2-(diphenylphosphaneyl)phenyl)methanimine. When the ligand is an imine, said ligand may be in a Z or E configuration, preferably in E. The ligands described above can be obtained by applying standard methods which are well known in the state of the art and by the person skilled in the art. Therefore, their preparation does not require a specific description. For example one may revert toOrg. Lett.,2015, 17 (3), 454-457. In general, the complexes of formula (1) can be prepared and isolated prior to their use in the process according to the general methods described in the literature. A method is described in the Example. Moreover, the complexes can be prepared in situ, by several methods, in the hydrogenation medium, without isolation or purification, just before their use. One of the possible procedures to advantageously prepare in situ a complex of the invention consists in reacting an appropriate Ru complex of formula [Ru(“diene”)(“allyl”)2], wherein “diene” represents a cyclic or linear hydrocarbon containing two carbon-carbon double bonds, conjugated or not, such as for example 1,5-cyclooctadiene (COD) or norbornadiene, and “allyl” represents a linear or branched C3to C8hydrocarbon radical containing one carbon-carbon double bond such as methylallyl or allyl, with a non-coordinating acid such as HBF4·Et2O, and then treating the resulting solution with the required amount of a ligands L, such as defined previously, to give a solution of a catalyst according to formula (3). Furthermore, the mixture thus obtained can also be treated with a base in the presence of a primary or secondary alcohol. Furthermore, the complexes of formula (1) or (2) can be prepared by reacting an appropriate Ru complex such as, [Ru(“diene”)(“allyl”)2], [RuCl2(PPh3)3], [RuCl2(COD)] or [RuCl2(arene)]2with the required amount of a ligands L, such as defined previously (COD representing a cyclooctadiene and arene being e.g. a benzene or naphthalene). It is also understood that the complex of the invention can also be obtained in situ from complexes which have a similar formula and which in presence of, for example an alcohol and a base, are converted into a invention's ruthenium complex, for example, from a complex wherein Y has other meaning. To carry out the processes of the invention it is required also to use a base. Said base can be the substrate itself, if the latter is basic, a corresponding alcoholate or any base having preferentially a pKaabove 11. According to a particular embodiment of the invention said base may have a pKaabove 14. It is also understood that preferably said base does not reduce itself a substrate of formula (I). As non-limiting examples one may cite the following type of base: alcoholate, hydroxides, alkaline or alkaline-earth carbonates, phosphazenes, alkylamidines, alkylguanidine amides, basic alox, siliconates (i.e. silicium derivatives having SiO−or SiRO−groups), hydrides such as NaBH4, NaH or KH. One can cite, as non-limiting examples, alkaline or alkaline-earth metal carbonates, such as cesium carbonate, an alkaline or alkaline-earth metal hydroxides, C1-10amidures, C10-26phosphazene or an alcoholate of formula (R17O)2M or R17O M′, wherein M is an alkaline-earth metal, M′ is an alkaline metal or an ammonium NR184+, R17stands for hydrogen or a C1to C6linear or branched alkyl radical and R18stands for a C1to C10linear or branched alkyl radical, such as sodium, lithium, cesium or potassium alcoholates. Of course, other suitable bases can be used. According to an embodiment of the invention, said base is an alkaline alcoholate of formula R17OM′. As previously mentioned the processes of the invention consist in the hydrogenation of a substrate using a ruthenium complex and a base. A typical process implies the mixture of the substrate with the ruthenium complex, a base and optionally a solvent, and then treating such a mixture with molecular hydrogen at a chosen pressure and temperature. The complex of the invention, an essential parameter of the process, can be added to the reaction medium in a large range of concentrations. As non-limiting examples, one can cite as complex concentration values those ranging from 1 ppm to 50000 ppm, relative to the amount of substrate. Preferably, the complex concentration will be comprised between 10 and 20000 ppm. Even more preferably, the complex concentration will be comprised between 10 and 5000 ppm. It goes without saying that the optimum concentration of complex will depend, as the person skilled in the art knows, on the nature of the latter, on the nature of the substrate and on the pressure of H2used during the process, as well as the desired time of reaction. Useful quantities of base, added to the reaction mixture, may be comprised in a relatively large range. One can cite, as non-limiting examples, ranges between 5 to 50000 molar equivalents, relative to the complex (e.g. base/com=5 to 50000), preferably 20 to 10000. The hydrogenation reaction can be carried out in the presence or absence of a solvent. When a solvent is required or used for practical reasons, then any solvent current in hydrogenation reactions can be used for the purposes of the invention. Non-limiting examples include aromatic solvents such as toluene, chlorobenzene or xylene, hydrocarbon solvents such as hexane or cyclohexane, ethers such as tetrahydrofuran, methyltetrahydrofuran or MTBE, polar solvents such as primary or secondary alcohols such as isopropanol or ethanol, or mixtures thereof. The choice of the solvent is a function of the nature of the complex and the person skilled in the art is well able to select the solvent most convenient in each case to optimize the hydrogenation reaction. In the hydrogenation process of the invention, the reaction can be carried out at a H2pressure comprised between 105Pa and 100×105Pa (1 to 100 bars) or even more if desired. Again, a person skilled in the art is well able to adjust the pressure as a function of the catalyst load and of the dilution of the substrate in the solvent. As examples, one can cite typical pressures of 1 to 50×105Pa (1 to 50 bars). The temperature at which the hydrogenation can be carried out is comprised between 0° C. and 120° C., more preferably in the range of between 20° C. and 100° C. Of course, a person skilled in the art is also able to select the preferred temperature as a function of the melting and boiling point of the starting and final products as well as the desired time of reaction or conversion. The ligand of formula (L) as defined above is also new. So another object of the present invention is the Ligand of formula (L). In addition, the catalyst of the present invention is also novel. So a last object of the present invention is a ruthenium complex of the general formula (1) as defined above. EXAMPLES The invention will now be described in further detail by way of the following examples, wherein the temperatures are indicated in degrees centigrade and the abbreviations have the usual meaning in the art. All the procedures described hereafter have been carried out under an inert atmosphere unless stated otherwise. Hydrogenations were carried out in open glass tubes placed inside a stainless steel autoclave. H2gas (99.99990%) was used as received. All substrates and solvents were distilled from appropriate drying agents under Ar. NMR spectra were recorded on a Bruker AM-400 (1H at 400.1 MHz,13C at 100.6 MHz, and31P at 161.9 MHz) spectrometer and normally measured at 300 K, in CDCl3unless indicated otherwise. Chemical shifts are listed in ppm. Example 1 Preparation of Ligand of the Invention a) N-(2-(diphenylphosphino)benzyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine (L-1) Preparation of 6-((diphenylphosphino)methyl)picolinaldehyde 6-((diphenylphosphino)methyl)picolinaldehyde was obtained according to some previously described procedure (Tan X., Wang Y., Liu Y., Wang F., Shi L., Lee K.-H., Lin Z., Lv H., Zhang X.,Org. Lett.,2015, 17 (3), 454-457). Initially isolated by filtration as the hydrochloride salt upon hydrolysis reaction of the acetal derivative under acidic aqueous conditions, basic treatment of the recovered white solid with sodium carbonate afforded desired product as a pale yellow solid. 1H NMR (500 MHz, CD2Cl2): δ (ppm) 3.71 (s, 2H, CH2), 7.17 (d, J=7.7 Hz, 1H, CH), 7.31 (m, 6H, 6CH), 7.44 (m, 4H, 4CH), 7.63 (t, J=7.7 Hz, 1H, CH), 7.68 (d, J=7.6 Hz, 1H, CH), 9.92 (s, 1H, CH). 13C NMR (125 MHz, CD2Cl2): δ (ppm) 38.7 (CH2), 119.2 (CH), 128.3 (CH), 128.8 (CH), 129.3 (CH), 133.3 (CH), 138.2 (C), 152.9 (C), 159.5 (C), 193.9 (CHO). 31P NMR (202 MHz, CD2Cl2): δ (ppm) −9.74 (s). (2-(diphenylphosphino)phenyl)methanamine (2-(diphenylphosphino)phenyl)methanamine may be synthesized in 2 steps from 2-bromobenzonitrile according to the following pathway Step 1: Pd(PPh3)4(0.8 mol. %), 2-bromobenzonitrile and degazed and dry toluene were loaded altogether in a round-bottomed flash equipped with a magnetic stirring bar and a dropping funnel. After purging with nitrogen, NEt3(1.04 eq.) was slowly added a room temperature. Upon addition completion, diphenylphosphine (1.03 eq.) was also slowly added a room temperature and reaction mixture was then heated to reflux for 12 h. After cooling down to room temperature, it was washed with degassed water, two times with 20 wt. % aqueous NH4Cl to bring pH down to neutrality and then with water. After azetropic water removal, toluene was fully concentrated to afford crude product as a yellow-orange sticky solid. It was recrystallized from MeOH to afford desired 2-(diphenylphosphino)benzonitrile as a pale yellow solid in 80% yield. 2-(diphenylphosphino)benzonitrile 1H NMR (400 MHz, CDCl3): δ (ppm) 7.04 (ddd, J=7.6, 4.5 and 1.1 Hz, 1H, CH), 7.27-7.34 (m, 7H, 7CH), 7.34-7.40 (m, 6H, 6CH), 7.42 (dd, J=7.6 and 1.1 Hz, 1H, CH), 7.47 (td, J=7.8 and 1.4 Hz, 1H, CH), 7.71 (ddd, J=7.8, 3.0 and 1.4 Hz, 1H, CH). 13C NMR (100 MHz, CDCl3): δ (ppm) 117.8 (C), 128.8 (CH), 128.9 (CH), 129.4 (CH), 132.4 (CH), 133.4 (CH), 133.7 (C), 133.9 (CH), 134.2 (CH), 134.7 (C), 143.2 (C). 31P NMR (162 MHz, CDCl3): δ (ppm) −7.79 (s, 1P). Step 2: Degassed and dry THF was added under nitrogen to a round-bottomed flask equipped with a magnetic stirring bar and containing pre-weighted LiAlH4 (1.2 eq.). The suspension was cooled down to 0° C. and 2-(diphenylphosphino)benzonitrile was added portionwise. After 2 additional hours at 0° C. it was stirred at room temperature overnight. It was then cooled down back to 0° C. and slowly quenched with aqueous sodium hydroxide. After THF removal, the remaining residue was dissolved in DCM, passed through a celite plug. The DCM solution was washed with water, dried other sodium sulfate and concentrated to dryness and further dried under high vacuum to afford desired product as a pale yellow solid. (2-(diphenylphosphino)phenyl)methanamine 1H NMR (400 MHz, CD2Cl2): δ (ppm) 1.35 (broad s, 2H, NH2), 3.97 (d, J=1.65 Hz, 2H, CH2), 6.87 (ddd, J=7.6, 4.5 and 1.2 Hz, 1H, CH), 7.12 (td, J=7.6 and 1.2 Hz, 1H, CH), 7.20-7.38 (m, 11H, 11CH), 7.42-7.48 (m, 1H, CH). 13C NMR (100 MHz, CD2Cl2): δ (ppm) 45.4 (CH2), 127.3 (CH), 128.2 (CH), 128.9 (CH), 129.1 (CH), 129.6 (CH), 133.8 (CH), 134.2 (CH), 135.4 (C), 137.1 (C), 148.2 (C). 31P NMR (162 MHz, CD2Cl2): δ (ppm) −15.55 (s, 1P). (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine (L-1) (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine was obtained by reaction at room temperature in THF of an equimolar mixture of 6-((diphenylphosphino)methyl)picolinaldehyde and ((2-(diphenylphosphino)phenyl)methanamine. Pure compound was obtained in quantitative yield as a pale yellow viscous oil upon solvent concentration and drying under high vacuum overnight. 1H NMR (500 MHz, CD2Cl2): δ (ppm) 3.61 (s, 2H, CH2), 5.04 (broad s, 2H, CH2), 6.90-6.96 (m, 2H), 7.20-7.50 (m, 25H), 8.25 (s, 1H, CH imine). 13C NMR (125 MHz, CD2Cl2): δ (ppm) 38.7 (CH2), 63.4 (CH2), 118.5 (CH), 124.9 (CH), 127.7 (CH), 128.7 (CH), 128.9 (CH), 129.0 (CH), 129.2 (CH), 129.5 (CH), 133.3 (CH), 134.0 (CH), 134.2 (CH), 134.4 (CH), 136.1 (C), 136.7 (CH), 137.1 (C), 138.7 (C), 144.1 (C), 154.7 (C), 158.1 (C), 163.7 (CH). 31P NMR (202 MHz, CD2Cl2): δ (ppm) −15.33 (s, 1P), −10.47 (s, 1P). b) Preparation of (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl)methanimine (L-2) Preparation of 6-((di-tert-butylphosphino)methyl)picolinaldehyde 6-((di-tert-butylphosphino)methyl)picolinaldehyde was obtained according to some similar multi-step synthesis as for 6-((diphenylphosphino)methyl)picolinaldehyde. It was fully characterized by 1H, 13C and 31P NMR analysis. 1H-NMR (300 MHz, CDCl3): δ 9.96 (1H, s, CHO), 7.71-7.65 (2H, m, 2×HPy), 7.62-7.58 (1H, m, HPy), 3.07 (2H, d, J=3.3 Hz, CH2P), 1.10 (18H, d, J=11.2 Hz, 6×CH3); 13C-NMR (75 MHz, CDCl3): δ 193.8 (CHO), 163.2 (d, J=14.7 Hz, CPy), 152.0 (CPy), 136.8 (CHPy), 128.3 (d, J=9.3 Hz, CHPy), 118.8 (d, J=1.3 Hz, CHPy), 32.0 (d, J=21.5 Hz, 2×CP), 31.5 (d, J=24.6 Hz, C H2P), 29.6 (d, J=13.3 Hz, 6×CH3); 31P-NMR (121 MHz, CDCl3): δ+37.7 s; Preparation of (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl)methanimine (L-2) (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl)methanimine was obtained by condensation at room temperature in THF of an equimolar mixture of 6-((di-tert-butylphosphino)methyl)picolinaldehyde and (2-(diphenylphosphino)phenyl)methanamine. It was obtained in quantitative yield as a pale yellow viscous oil upon solvent concentration and drying under high vacuum overnight and used directly for complex synthesis c) Preparation of (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl) ethan-1-imine (L-3) Preparation of 1-(6-((Di-tert-butylphosphino)methyl)pyridin-2-yl)ethan-1-one) 1-(6-((Di-tert-butylphosphino)methyl)pyridin-2-yl)ethan-1-one was obtained according to some procedure previously described in Angew. Chem. Int. Ed. 2016, 55, 6671-6675. It was fully characterized by 1H, 13C and 31P NMR analysis, with data corresponding the the previously reported ones. Preparation of (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl) ethan-1-imine (L-3) E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl) ethan-1-imine was obtained by condensation at room temperature in THF of an equimolar mixture of 1-(6-((Di-tert-butylphosphino)methyl)pyridin-2-yl)ethan-1-one and (2-(diphenylphosphino)phenyl)methanamine. It was obtained in quantitative yield as a pale yellow solid upon solvent concentration and drying under high vacuum overnight and used directly for complex synthesis Example 2 Preparation of Invention Complex a) Preparation of [RuCl2((E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine)] (Complex C1) Complex C1 was obtained by reaction of (PPh3)3RuCl2ruthenium complex with 1.05 equivalents of ligand L1 in toluene refluxing for 4 h. Upon cooling down to room temperature, toluene was partly concentrated under vacuum and Et2O was added for product precipitation. The suspension was filtered under nitrogen and the obtained purple solid was washed several times with a toluene/Et2O mixture and then pure Et2O. After drying under high vacuum overnight, product was obtained in 85% yield as a 5/2 stereoisomers mixture. 31P NMR (202 MHz, CD2Cl2): δ (ppm) 46.27 (d, J=22.8 Hz, 1P major isomer), 47.34 (d, J=28.6 Hz, 1P minor isomer), 48.49 (d, J=22.8 Hz, 1P major isomer), 59.49 (d, J=28.6 Hz, 1P minor isomer). b) Preparation of Complex [RuCl2((E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl)methanimine)] (Complex C2) Complex C2 was obtained by reaction of (PPh3)3RuCl2ruthenium complex with 1.05 equivalents of (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl)methanimine (L-2) in toluene refluxing for 4 h. Upon cooling down to room temperature, toluene was partly concentrated under vacuum and Et2O was added for product precipitation. The suspension was filtered under nitrogen and the obtained purple solid was washed several times with a toluene/Et2O mixture and then pure Et2O. After drying under high vacuum overnight, product was obtained in 75% yield as a 4/1 stereoisomers mixture. 31P NMR (202 MHz, CD2Cl2): δ 38.38 (d, J=18.5 Hz, 1P major isomer), 58.62 (broad s, 1P minor isomer), 61.82 (d, J=18.5 Hz, 1P major isomer), 62.35 (broad s, 1P minor isomer). c) Preparation of Complex [RuCl2((E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl)ethan-1-imine)] (Complex C3) Complex C3 was obtained by reaction of (PPh3)3RuCl2ruthenium complex with 1.05 equivalents of (E)-N-(2-(diphenylphosphino)benzyl)-1-(6-((di-tert-butylphosphino)methyl)pyridin-2-yl)ethan-1-imine (L-3) in toluene refluxing for 4 h. Upon cooling down to room temperature, toluene was partly concentrated under vacuum and Et2O was added for product precipitation. The suspension was filtered under nitrogen and the obtained purple solid was washed several times with a toluene/Et2O mixture and then pure Et2O. After drying under high vacuum overnight, product was obtained in 85% yield as a single isomer. 1H-NMR (500 MHz, CD2Cl2): δ 7.80-7.70 (m, 5H), 7.58 (t, J=1.80 Hz, 1H), 7.45-7.25 (m, 11H), 5.21 (t, J=3.2 Hz, 2H, CH2), 3.79 (d, J=8.8 Hz, 2H, CH2), 2.78 (s, 3H, CH3), 1.10 (s, 9H, 3 CH3), 1.07 (s, 9H, 3 CH3). 13C-NMR (125.76 MHz, CD2Cl2): δ 169.26 (C), 166.09 (C), 159.58 (C), 139.24 (C), 138.57 (C), 136.82 (C), 135.62 (CH), 135.10 (CH), 134.06 (CH), 132.09 (CH), 131.21 (CH), 129.78 (CH), 129.11 (CH), 128.88 (CH), 128.09 (CH), 128.00 (CH), 123.87 (CH), 122.47 (CH), 60.95 (CH2), 39.17 (CH2), 37.70 (C), 30.54 (CH3), 17.04 (CH3). 31P NMR (202 MHz, CD2Cl2): δ 35.84 (d, J=19.5 Hz, 1P), 63.47 (d, J=19.5 Hz, 1P). Example 3 Preparation of Comparative Complexes a) Preparation of [RuCl2(2-(diphenylphosphino)-N-((6-((diphenylphosphino)methyl)pyridin-2-yl)methyl)ethan-1-amine)] (Comparative Complex CC1) Complex [RuCl2(2-(diphenylphosphino)-N-((6-((diphenylphosphino)methyl)pyridin-2-yl)methyl)ethan-1-amine)] was obtained according to some previously described procedure (Tan X., Wang Y., Liu Y., Wang F., Shi L., Lee K.-H., Lin Z., Lv H., Zhang X.,Org. Lett.,2015, 17 (3), 454-457). b) Preparation of [RuCl2((E)-N-(2-(diphenylphosphino)ethyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine)] (comparative Complex CC2) (E)-N-(2-(diphenylphosphino)ethyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine ligand (E)-N-(2-(diphenylphosphaneyl)ethyl)-1-(6-((diphenylphosphaneyl)methyl)pyridin-2-yl)methanimine (E)-N-(2-(diphenylphosphino)ethyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine ligand was obtained by reaction at room temperature in THF of an equimolar mixture of 6-((diphenylphosphino)methyl)picolinaldehyde and 2-(diphenylphosphino)ethan-1-amine Pure compound was obtained in quantitative yield as a pale yellow viscous oil upon solvent concentration and drying under high vacuum overnight. 1H NMR (500 MHz, CD2Cl2): δ (ppm) 2.46 (tm, J=7.8 Hz, 2H, CH2), 3.62 (s, 2H, CH2), 3.75 (qm, J=7.8 Hz, 2H, CH2) 6.94 (d, J=7.8 Hz, 1H, CH), 7.31 (m, 12H, 12CH), 7.44 (m, 9H, 9CH), 7.63 (d, J=7.8 Hz, 1H, CH), 8.22 (s, 1H, CH). 13C NMR (125 MHz, CD2Cl2): δ (ppm) 30.0 (CH2), 38.7 (CH2), 58.4 (CH2), 118.5 (CH) 124.9 (CH), 128.7 (CH), 128.8 (CH), 128.9 (CH), 129.1 (CH) 133.1 (CH), 133.2 (CH), 136.8 (CH), 138.8 (C), 138.9 (C), 154.6 (C), 158.2 (C), 162.8 (CH). 31P NMR (202 MHz, CD2Cl2): δ (ppm) −18.73 (s), −10.41 (s). [RuCl2((E)-N-(2-(diphenylphosphino)ethyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine)] (Complex CC2) it was obtained by reaction of (PPh3)3RuCl2ruthenium complex with 1.05 equivalents of (E)-N-(2-(diphenylphosphino)ethyl)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)methanimine ligand in toluene refluxing for 4 h. Upon cooling down to room temperature, toluene was partly concentrated under vacuum and Et2O was added for product precipitation. The suspension was filtered under nitrogen and the obtained purple solid was washed several times with a toluene/Et2O mixture and then pure Et2O. After drying under high vacuum overnight, pure product was obtained in 85% yield. 1H NMR (500 MHz, CD2Cl2): δ (ppm) 3.21 (qm, J=7.8 Hz, 2H, CH2), 4.52 (d, J=10.4 Hz, 2H, CH2), 4.60-4.70 (m, 2H, CH2), 7.22-7.28 (m, 8H, 8CH), 7.30-7.38 (m, 4H, 4CH), 7.43-7.57 (m, 8H, 8CH), 7.74-7.86 (m, 3H, 3CH), 8.95 (dt, J=7.0 and 1.6 Hz, 1H, CH). 13C NMR (125 MHz, CD2Cl2): δ (ppm) 35.8 (CH2), 47.7 (CH2), 58.6 (CH2), 122.5 (CH), 125.4 (CH), 127.8 (CH), 127.9 (CH), 129.6 (CH), 129.8 (CH), 133.8 (CH), 134.1 (CH), 135.0 (CH), 135.6 (C), 136.2 (C), 158.4 (C), 160.8 (CH) 162.5 (C). 31P NMR (202 MHz, CD2Cl2): δ (ppm) 54.6 (d, J=19.60 Hz), 60.5 (d, J=19.60 Hz) c) Preparation of [RuCl2((E)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)-N-(3-(diphenylphosphino)propyl)methanimine)] (Comparative Complex CC3) (E)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)-N-(3-(diphenylphosphino)propyl) methanimine ligand it was obtained by reaction at room temperature in THF of an equimolar mixture of 6-((diphenylphosphino)methyl)picolinaldehyde and 3-(diphenylphosphino)propan-1-amine Pure compound was obtained in quantitative yield as a pale yellow viscous oil upon solvent concentration and drying under high vacuum overnight. 1H NMR (500 MHz, CD2Cl2): δ (ppm) 1.75-1.87 (m, 2H, CH2), 2.09-2.16 (m, 2H, CH2), 3.62 (s, 2H, CH2), 3.70 (td, J=6.8 and 1.2 Hz, 2H, CH2) 6.94 (dt, J=7.8 and 1.2 Hz, 1H, CH), 7.27-7.35 (m, 12H, 12CH), 7.38-7.46 (m, 8H, 8CH), 7.50 (t, J=7.8 Hz, 1H, CH), 7.63 (d, J=7.8 Hz, 1H, CH), 8.26 (s, 1H, CH). 13C NMR (125 MHz, CD2Cl2): δ (ppm) 25.8 (CH2), 27.7 (CH2), 38.7 (CH2), 62.4 (CH2), 118.4 (CH) 124.9 (CH), 128.7 (CH), 128.9 (CH), 129.1 (CH) 133.0 (CH), 133.2 (CH), 136.8 (CH), 138.7 (C), 139.3 (C), 154.8 (C), 158.2 (C), 162.8 (CH). 31P NMR (202 MHz, CD2Cl2): δ (ppm) −16.11 (s), −10.42 (s). [RuCl2((E)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)-N-(3-(diphenylphosphino)propyl)methanimine)] (Comparative Complex CC3) it was obtained by reaction of (PPh3)3RuCl2ruthenium complex with 1.05 equivalents of (E)-1-(6-((diphenylphosphino)methyl)pyridin-2-yl)-N-(3 (diphenylphosphino)propyl) methanimine ligand in toluene refluxing for 4 h. Upon cooling down to room temperature, toluene was partly concentrated under vacuum and Et2O was added for product precipitation. The suspension was filtered under nitrogen and the obtained purple solid was washed several times with a toluene/Et2O mixture and then pure Et2O. After drying under high vacuum overnight, pure product was obtained in 75% yield. 1H NMR (500 MHz, CD2Cl2): δ (ppm) 2.40-2.54 (m, 2H, CH2), 2.79-2.87 (m, 2H, CH2), 4.35-4.40 (m, 2H, CH2), 4.41 (d, J=10.8 Hz, 2H, CH2), 7.08-7.14 (m, 8H, 8CH), 7.16-7.22 (m, 4H, 4CH), 7.23-7.30 (m, 4H, 4CH), 7.32-7.38 (m, 4H, 4CH), 7.61-7.67 (m, 1H, CH, 7.75-7.81 (m, 2H, 2CH), 8.71 (dt, J=5.8 and 1.8 Hz, 1H, CH). 13C NMR (125 MHz, CD2Cl2): δ (ppm) 24.4 (CH2), 27.5 (CH2), 48.7 (CH2), 63.0 (CH2), 122.7 (CH) 125.4 (CH), 127.5 (CH), 127.6 (CH), 129.1 (CH), 129.6 (CH), 134.1 (CH), 134.2 (CH), 135.4 (CH), 135.8 (C), 138.0 (C), 155.6 (C), 162.4 (C), 165.0 (CH). 31P NMR (202 MHz, CD2Cl2): δ (ppm) 36.12 (d, J=30.4 Hz), 50.5 (d, J=30.4 Hz). d) [RuCl2(triphenylphosphine)(bis(2-(ethylthio)ethyl)amine)] (Comparative Complex CC4) Commercially available complex [RuCl2(triphenylphosphine)(bis(2-(ethylthio)ethyl)amine)] was purchased from Sigma-Aldrich. e) [RuCl2(N,N′-(ethane-1,2-diyl)bis(1-(2-(diphenylphosphaneyl)phenyl)methanimine))] (Comparative Complex CC5) Complex [RuCl2(N,N′-(ethane-1,2-diyl)bis(1-(2-(diphenylphosphaneyl)phenyl)methanimine))] was synthesized according to some previously described procedure (Saudan L., Dupau P., Riedhauser J.-J., Wyss P., WO200610648) f) [RuCl2(bis(2-(diphenylphosphino)ethylamine))] (Comparative Complex CC6) Commercially available complex [RuCl2(bis(2-(diphenylphosphino)ethylamine))] was purchased from Sigma-Aldrich (CAS number: [506417-41-0]). Example 4 General Hydrogenation Reaction Procedure: Ester, ruthenium catalyst, metal alkoxide co-catalyst (used as a solid or some alcoholic solution) and optionally solvent (see Table 1) were loaded altogether in an 100 mL or 1 L autoclave equipped with a mechanical stirring device, pressure and internal temperature sensors and a heating/cooling system for internal temperature regulation. Sealed autoclave was then purged under stirring with nitrogen (3 times 5 bars) and hydrogen (3 times 5 bars) before being pressurized to required hydrogen pressure via an hydrogen tank equipped with a way out pressure regulator and also an internal pressure sensor to follow and determine hydrogen consumption. Reaction mixture was then heated to required temperature and hydrogen pressure into the autoclave was maintained to the desired value during the whole reaction. Upon reaction completion also determined by GC analysis with complete disappearance of both starting material and mixed ester coming from transesterification reaction with product and eventually with metal alkoxide co-catalyst and/or alcoholic solvent, autoclave was then cooled down to 25° C. It was then depressurized and purged with nitrogen (3 times 5 bars) and reaction mixture was then transferred to a round-bottomed flask and lights compounds were removed under vacuum. Crude product was then flash distilled in order to determine the quantity of residues formed during the reaction and yield was calculated based on GC purity of distilled product. Example 5 Catalytic hydrogenation of different esters using different catalyst of the invention and comparative catalyst: The hydrogenation has been performed as reported in Example 4. TABLE 1Hydrogenation of different esters using different complexesTime forcompleteComplexNaOEtPTConver-EntryEster(mol %)(mol %)(bars)(° C.)sion1)(h)Yield1E1CC15501007>99%(0.00166)2E1CC25501007>99%(0.00166)3E1CC35501007>99%(0.00166)4E1C15501001>99%(0.00166)5E2CC15501004>99%(0.00166)6E2CC25501004>99%(0.00166)7E2C15501001.5>99%(0.00166)8E3CC15501003>98%2)(0.00333)9E3CC25501003>98%2)(0.00333)10E3C15501001>98%2)(0.00333)11E2C1550800.66>99%(0.01)12E2C2550800.8>99%(0.01)13E2C3550801>99%(0.01)14E2CC1550802.5>99%(0.01)15E2CC4550805>99%(0.01)16E2CC55508024>99%(0.01)17E2CC655080>36n.d.(0.01)1)Complete conversion was achieved with nearly no residues formation (<1 wt. %).2)Product was obtained with >99% GC selectivity The complete conversion was reached faster with the invention's catalyst compared to prior art catalysts. TABLE 2Structure and names of esters usedEsterStructureNameE1Ethyl benzoateE2Ethyl octanoateE3Butyl 3-(4,4-dimethylcyclohex-1-en-1- yl)propanoateE4Methyl octanoateE5Methyl 2-cyclohexylacetateE6Methyl 3,7-dimethyloct-6-enoateE7Ethyl 2-(cyclopent-1-en-1-yl)acetateE8Methyl (E)-4-methyl-6-(2,6,6- trimethylcyclohex-1-en-1-yl)hex-3-enoateE9Butyl (E)-4-methyl-6-(2,6,6- trimethylcyclohex-1-en-1-yl)hex-3-enoateE10Ethyl 2-methylhexanoateE11Methyl 2-methylhexanoateE12Ethyl (E)-5-cyclohexyl-2,4-dimethylpent- 4-enoateE13Ethyl (E)-2,4-dimethylpent-2-enoateE14Ethyl 2,5-dimethyl-2,3-dihydro-1H-indene- 2-carboxylateE15(+/−)-(3aR,5aS,9aS,9bR)-3a,6,6,9a- tetramethyldecahydronaphtho[2,1-b]furan- 2(1H)-one (racemic compound with displayed relative stereochemistry)E16(+)-(3aR,5aS,9aS,9bR)-3a,6,6,9a- tetramethyldecahydronaphtho[2,1-b]furan- 2(1H)-one (enantiomerically enriched) Example 6 Catalytic Hydrogenation of Ethyl Benzoate Using Different Catalysts of the Invention and Comparative Catalysts in Various Solvents: The hydrogenation has been performed as reported in Example 4. TABLE 3Hydrogenation of Ethyl benzoate using different complexes in various solventsTime forcompleteComplexNaOEtPTConversion2)EntrySolvent1)(mol %)(mol %)(bars)(° C.)(h)Yield1noneCC15501002>99%(0.00333)2noneCC25501002>99%(0.00333)3noneC15501000.5>99%(0.00333)4iPrOHCC15501004>99%(0.00333)5iPrOHCC25501004>99%(0.00333)6iPrOHC15501001>99%(0.00333)7EtOHCC15501004>99%(0.00333)8EtOHCC25501004>99%(0.00333)9EtOHC15501001>99%(0.00333)10THFCC15501005>99%(0.00333)11THFCC25501005>99%(0.00333)12THFC15501001.5>99%(0.00333)13TolueneCC15501008>99%(0.00333)14TolueneCC25501008>99%(0.00333)15TolueneC15501001.5>99%(0.00333)16ChlorobenzeneCC15501006>99%(0.00333)17ChlorobenzeneCC25501006>99%(0.00333)18chlorobenzeneC15501001.5>99%(0.00333)1)Reactions were run (when applicable) with 2 equivalents in volume of solvent.2)Complete conversion was achieved with nearly no residues formation (<1 wt. %). The invention's catalyst allows reaching complete conversion faster than the prior art catalyst regardless of the solvent used. Example 7 Catalytic Hydrogenation of Different Esters Using Complex C1 at Various Temperatures: The hydrogenation has been performed as reported in Example 4. TABLE 4Hydrogenation of ester using complex C1 at various temperatureTime forcompleteComplexNaOEtPTConver-EntryEster(mol %)(mol %)(bars)(° C.)sion1)(h)Yield1E1C1550404>99%(0.01)2E1C1550601>99%(0.01)3E1C1550603>99%(0.003333)4E1C1550801>99%(0.003333)5E1C1550804>99%(0.001666)6E1C15501001>99%(0.001666)7E2C15504012>99%(0.01)8E2C1550602>99%(0.01)9E2C1550606>99%(0.003333)10E2C1550802>99%(0.003333)11E2C1550806>99%(0.001666)12E2C15501001.5>99%(0.001666)1)Complete conversion was achieved with nearly no residues formation (<1 wt. %). Example 8 Catalytic Hydrogenation Under Neat Conditions of Ethyl Octanoate Using Complex C1 at Various Hydrogen Pressures: The hydrogenation has been performed as reported in Example 4. TABLE 5Hydrogenation of ethyl octanoate withC1 at various hydrogen pressuresTime forcompleteComplexNaOEtPTConver-EntryEster(mol %)(mol %)(bars)(° C.)sion1)(h)Yield1E2C1510809>99%(0.01)2E2C1520803>99%(0.01)3E2C1530802>99%(0.01)4E2C1550800.66>99%(0.01)1)Complete conversion was achieved with nearly no residues formation (<1 wt. %). Example 9 Catalytic Hydrogenation Under Neat Conditions of Ethyl Octanoate Using Complex C1 with Various Metal Alkoxides as a Base: The hydrogenation has been performed as reported in Example 4. TABLE 6Neat hydrogenation using various metal alkoxides as a baseTime forcompleteComplexBasePTConver-EntryEster(mol %)(mol %)(bars)(° C.)sion1)(h)Yield1E2C1NaOEt50802>99%(0.003333)(5)2E2C1KOEt50805>99%(0.003333)(5)3E2C1KOtBu50805>99%(0.003333)(5)4E2C1LiOEt508012>99%(0.003333)(5)5E2C1LiOtBu508012>99%(0.003333)(5)1)Complete conversion was achieved with nearly no residues formation (<1 wt. %). Example 10 Catalytic Hydrogenation Under Neat Conditions of Various Esters Using Complex C1: The hydrogenation has been performed as reported in Example 4. TABLE 7Neat hydrogenation of various estersTime forcompleteComplexNaOEtPTConver-EntryEster(mol %)(mol %)(bars)(° C.)sion1)(h)Yield1E4C17.5508012>99%(0.003333)2E5C1550804>99%(0.01)3E6C1550805>98%2)(0.01)4E7C1550802>98%2)(0.005)5E8C12.55080994%3)(0.005)6E9C12.55080494%3)(0.01)7E10C1550805>99%(0.01)8E11C15508016>99%(0.01)9E12C15501006>98%2)(0.005)10E13C155080494%3)(0.01)11E14C15501003>99%(0.005)1)Complete conversion was achieved with nearly no residues formation (<1 wt. %).2)Desired product was obtained with more than 99% GC selectivity at complete conversion.3)Desired product was obtained with 95% GC selectivity at complete conversion. Example 11 Catalytic Hydrogenation of (+/−)-(3aR,5aS,9aS,9bR)-3a,6,6,9a-tetramethyldecahydronaphtho[2,1-b]furan-2(1H)-one Using Different Catalyst of the Invention and Comparative Catalyst in Various Solvents The hydrogenation has been performed as reported in Example 4. TABLE 8Neat Hydrogenation of (+/−)-(3aR,5aS,9aS,9bR)-3a,6,6,9a-tetramethyldecahydronaphtho[2,1-b]furan-2(1H)-one in solventTime forcompleteComplexNaOEtPTConver-EntrySolvent(mol %)(mol %)(bars)(° C.)sion3)(h)Yield1iPrOH1)C15501002>98%(0.0025)2iPrOH1)CC655010024>98%(0.01)3Chlorobenzene2)C15301006>98%(0.005)4chlorobenzene2)CC653010024>98%(0.04)1)Reactions run with 1 equivalent in volume of solvent.2)Reactions run with 2 equivalents in volume of solvent.3)Complete conversion was achieved with nearly no residues formation (<1 wt. %). Same results were obtained from enantiomerically enriched compound E16.
55,174
11858949
DETAILED DESCRIPTION Reference will now be made in detail to embodiments, examples of which are illustrated in the accompanying drawings, wherein like reference numerals refer to like elements throughout. In this regard, the present embodiments may have different forms and should not be construed as being limited to the descriptions set forth herein. Accordingly, the embodiments are merely described below, by referring to the figures, to explain aspects. As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items. Expressions such as “at least one of,” when preceding a list of elements, modify the entire list of elements and do not modify the individual elements of the list. It will be understood that when an element is referred to as being “on” another element, it can be directly on the other element or intervening elements may be present therebetween. In contrast, when an element is referred to as being “directly on” another element, there are no intervening elements present It will be understood that, although the terms “first,” “second,” “third” etc. may be used herein to describe various elements, components, regions, layers and/or sections, these elements, components, regions, layers and/or sections should not be limited by these terms. These terms are only used to distinguish one element, component, region, layer or section from another element, component, region, layer or section. Thus, “a first element,” “component,” “region,” “layer” or “section” discussed below could be termed a second element, component, region, layer or section without departing from the teachings herein. The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. As used herein, “a,” “an,” “the,” and “at least one” do not denote a limitation of quantity, and are intended to cover both the singular and plural, unless the context clearly indicates otherwise. For example, “an element” has the same meaning as “at least one element,” unless the context clearly indicates otherwise. “Or” means “and/or.” As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items. It will be further understood that the terms “comprises” and/or “comprising,” or “includes” and/or “including” when used in this specification, specify the presence of stated features, regions, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, regions, integers, steps, operations, elements, components, and/or groups thereof. Furthermore, relative terms, such as “lower” or “bottom” and “upper” or “top,” may be used herein to describe one element's relationship to another element as illustrated in the Figures. It will be understood that relative terms are intended to encompass different orientations of the device in addition to the orientation depicted in the Figures. For example, if the device in one of the figures is turned over, elements described as being on the “lower” side of other elements would then be oriented on “upper” sides of the other elements. The exemplary term “lower,” can therefore, encompasses both an orientation of “lower” and “upper,” depending on the particular orientation of the FIGURE. Similarly, if the device in one of the figures is turned over, elements described as “below” or “beneath” other elements would then be oriented “above” the other elements. The exemplary terms “below” or “beneath” can, therefore, encompass both an orientation of above and below. “About” or “approximately” as used herein is inclusive of the stated value and means within an acceptable range of deviation for the particular value as determined by one of ordinary skill in the art, considering the measurement in question and the error associated with measurement of the particular quantity (i.e., the limitations of the measurement system). For example, “about” can mean within one or more standard deviations, or within ±30%, 20%, 10% or 5% of the stated value. Unless otherwise defined, all terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. It will be further understood that terms, such as those defined in commonly used dictionaries, should be interpreted as having a meaning that is consistent with their meaning in the context of the relevant art and the present disclosure, and will not be interpreted in an idealized or overly formal sense unless expressly so defined herein. Exemplary embodiments are described herein with reference to cross section illustrations that are schematic illustrations of idealized embodiments. As such, variations from the shapes of the illustrations as a result, for example, of manufacturing techniques and/or tolerances, are to be expected. Thus, embodiments described herein should not be construed as limited to the particular shapes of regions as illustrated herein but are to include deviations in shapes that result, for example, from manufacturing. For example, a region illustrated or described as flat may, typically, have rough and/or nonlinear features. Moreover, sharp angles that are illustrated may be rounded. Thus, the regions illustrated in the figures are schematic in nature and their shapes are not intended to illustrate the precise shape of a region and are not intended to limit the scope of the present claims. An organometallic compound according to one or more embodiments is represented by Formula 1 below: M1(L11)n11(L12)n12<Formula 1> M11in Formula 1 may be a Period 1 transition metal, a Period 2 transition metal, or a Period 3 transition metal. For example, M1in Formula 1 may be beryllium (Be), magnesium (Mg), aluminum (Al), calcium (Ca), titanium (Ti), manganese (Mn), cobalt (Co), copper (Cu), zinc (Zn), gallium (Ga), germanium (Ge), zirconium (Zr), ruthenium (Ru), rhodium (Rh), palladium (Pd), silver (Ag), rhenium (Re), platinum (Pt), or gold (Au). According to one or more embodiments, M1may be Pd, Pt or Au. According to one or more embodiments, M1in Formula 1 may be Pt or Pd. According to one or more embodiments, M1in Formula 1 may be Pt. L11in Formula 1 may be a ligand represented by Formula 1-1: *1 to *4 in Formula 1-1 may each independently indicate a binding site to M1. A20, A30, and A40in Formula 1-1 may each independently be a C5-C30carbocyclic group or a C1-C30heterocyclic group. According to one or more embodiments, A20, A30, and A40may each independently be a benzene group, a naphthalene group, an anthracene group, a phenanthrene group, a triphenylene group, a pyrene group, a chrysene group, a cyclopentadiene group, a 1,2,3,4-tetrahydronaphthalene group, a furan group, a thiophene group, a silole group, an indene group, a fluorene group, an indole group, a carbazole group, a benzofuran group, a dibenzofuran group, a benzothiophene group, a dibenzothiophene group, a benzosilole group, a dibenzosilole group, an azafluorene group, an azacarbazole group, an azadibenzofuran group, an azadibenzothiophene group, an azadibenzosilole group, a pyridine group, a pyrimidine group, a pyrazine group, a pyridazine group, a triazine group, a quinoline group, an isoquinoline group, a quinoxaline group, a quinazoline group, a phenanthroline group, a pyrrole group, a pyrazole group, an imidazole group, a triazole group, a tetrazole group, an oxazole group, an isooxazole group, a thiazole group, an isothiazole group, an oxadiazole group, a thiadiazole group, a benzopyrazole group, a benzimidazole group, an indazole group, a benzoxazole group, a benzothiazole group, a benzoxadiazole group, a benzothiadiazole group, a benzotriazole group, a diazaindene group, a triazaindene group, a 5,6,7,8-tetrahydroisoquinoline group, or a 5,6,7,8-tetrahydroquinoline group. T1in Formula 1-1 may be a single bond, *—N[(L1)a1-(R1)b1]—*′, *—B(R1)—*′, *—P(R1)—*′,*—C(R1)(R2)—*′, *—Si(R1)(R2)—*′, *—Ge(R1)(R2)—*′, *—S—*′, *—Se—*′, *—O—*′, *—C(═O)—*′, *—S(═O)—*′,*—S(═O)2—*′, *—C(R1)═C(R2)—*′, *—C(═S)—*′, or *—C≡C—*′, T2may be a single bond, *—N[(L2)a2-(R3)b3]—*′, *—B(R3)—*′, *—P(R3)—*′, *—C(R3)(R4)—*′,*—Si(R3)(R4)—*′, *—Ge(R3)(R4)—*′, *—S—*′, *—Se—*′, *—O—*′, *—C(═O)—*′, *—S(═O)—*′, *—S(═O)2—*′, *—C(R3)═C(R4)—*′, *—C(═S)—*′, or *—C≡C—*′. According to one or more embodiments, T1may be a single bond, *—N[(L1)a1-(R1)b1]—*′, *—B(R1)—*′, *—C(R1)(R2)—*′, *—Si(R1)(R2)—*′, *—O—*1or *—S—*′. According to one or more embodiments, T1may be *—N[(L1)a1-(R1)b1]—*′,*—C(R1)(R2)—*′, *—Si(R1)(R2)—*′, *—O—*′ or *—S—*′. According to one or more embodiments, T2may be a single bond, *—N[(L2)a2-(R3)b3]—*′, *—C(R3)(R4)—*′, *—Si(R3)(R4)—*′, *—O—*′ or *—S—*′. L1and L2in Formula 1-1 may each independently be a single bond, a substituted or unsubstituted C5-C30carbocyclic group, or a substituted or unsubstituted C1-C30heterocyclic group, a1 may be an integer from 1 to 3, and when a1 is two or more, two or more L1(s) may be identical to or different from each other, and a2 may be an integer from 1 to 3, and when a2 is 2 or more, two or more L2(s) may be identical to or different from each other. According to one or more embodiments, L1and L2may each independently be a phenylene group, a pentacenylene group, an indenylene group, a naphthylene group, an azulenylene group, a heptalenylene group, an acenaphthylene group, a fluorenylene group, a phenalenylene group, a phenanthrenylene group, an anthracenylene group, a fluoranthenylene group, a triphenylenylene group, a pyrenylene group, a chrysenylenylene group, a naphthacenylene group, a picenylene group, a perylenylene group, or a pentacenylene group; or a phenylene group, a pentalenylene group, an indenylene group, a naphthylene group, an azulenylene group, a heptalenylene group, an acenaphthylene group, a fluorenylene group, a phenalenylene group, a phenanthrenylene group, an anthracenylene group, a fluoranthenylene group, a triphenylenylene group, a pyrenylene group, a chrysenylenylene group, a naphthacenylene group, a picenylene group, a perylenylene group, or a pentacenylene group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, a C3-C10cycloalkyl group, a C3-C10cycloalkenyl group, a C1-C10heterocycloalkyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, a monovalent non-aromatic condensed heteropolycyclic group, or any combination thereof. X11in Formula 1-1 may be C(R11) or N. For example, X11may be C(R11). For example, X11may be N. X12in Formula 1-1 may be C(R12) or N. For example, X12may be C(R12). In one or more embodiments, X12may be N. According to one or more embodiments, X11may be C(R11) and X12may be C(R12), and as described later, R11and R12may optionally be linked to each other to form a substituted or unsubstituted C5-C30carbocyclic group or a substituted or unsubstituted C1-C30heterocyclic group. According to one or more embodiments, X11may be C(R11) and X12may be C(R11), and R11and R12may optionally be linked to each other to form a benzene group, a pyridine group, a pyrimidine group, a pyrazine group, or a pyridazine group. X20in Formula 1-1 may be C or N. X30in Formula 1-1 may be C or N. X40in Formula 1-1 may be C or N. X21, X22, X31, X32and X41in Formula 1-1 may each independently be C or N. According to one or more embodiments, a bond between M1and X20, a bond between M1and X30, and a bond between M1and X40may each independently be a coordinate bond or a covalent bond. In Formula 1, a bond between M1and moiety may be a coordination bond. In Formula 1, two bonds of a bond between M1and A20, a bond between M1and A30, and a bond between M1and A40may each be a covalent bond, and the other bond may be a coordination bond. Thus, the organometallic compound represented by Formula 1 may be electrically neutral. According to one or more embodiments, a bond between M1and A20may be a covalent bond, a bond between M1and A30may be a covalent bond, and a bond between M1and A40may be a coordinate bond. Ar1in Formula 1-1 may be a group represented by Formula Ar1-1: In Formula Ar1-1, E1may be deuterium, a cyano group, a substituted or unsubstituted C1-C60alkyl group, a substituted or unsubstituted C3-C10cycloalkyl group, a substituted or unsubstituted C3-C10cycloalkenyl group, or a substituted or unsubstituted phenyl group, E2to E5may each independently be hydrogen, deuterium, a cyano group, a substituted or unsubstituted C3-C10cycloalkyl group, a substituted or unsubstituted C3-C10cycloalkenyl group, or a substituted or unsubstituted phenyl group, E1and E2may be different from each other, and * indicates a binding site to a neighboring atom. Since E1and E2in Formula Ar1-1 are different from each other, a group represented by Formula Ar1-1, that is, Ar1may have an asymmetric structure. Accordingly, the organometallic compound represented by Formula 1 is less likely to be crystallized, and thus when applied to an organic light-emitting device, the obtained organic light-emitting device may have a longer lifespan. According to one or more embodiments, E1may be deuterium, a cyano group, a substituted or unsubstituted C1-C60alkyl group, or a substituted or unsubstituted C3-C10cycloalkyl group, or a substituted or unsubstituted phenyl group, and E2to E5may each independently be hydrogen, deuterium, a cyano group, a substituted or unsubstituted C3-C10cycloalkyl group, or a substituted or unsubstituted phenyl group. In one or more embodiments, E1may be: deuterium, a cyano group, a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, an n-hexyl group, an isohexyl group, a sec-hexyl group, a tert-hexyl group, an n-heptyl group, an isoheptyl group, a sec-heptyl group, a tert-heptyl group, an n-octyl group, an isooctyl group, a sec-octyl group, a tert-octyl group, an n-nonyl group, an isononyl group, a sec-nonyl group, a tert-nonyl group, an n-decyl group, an isodecyl group, a sec-decyl group, a tert-decyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, or a phenyl group; or a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, an n-hexyl group, an isohexyl group, a sec-hexyl group, a tert-hexyl group, an n-heptyl group, an isoheptyl group, a sec-heptyl group, a tert-heptyl group, an n-octyl group, an isooctyl group, a sec-octyl group, a tert-octyl group, an n-nonyl group, an isononyl group, a sec-nonyl group, a tert-nonyl group, an n-decyl group, an isodecyl group, a sec-decyl group, a tert-decyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, or a phenyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a C1-C10alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof. In one or more embodiments, E2to E5may each independently be: hydrogen, deuterium, a cyano group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, or a phenyl group; or a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, or a phenyl group, each substituted with at least one of deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a C1-C10alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof. In one or more embodiments, Ar1may be a group represented by one of Formulae 3-1 to 3-37: * in Formulae 3-1 to 3-37 indicates a binding site to a neighboring atom. R1to R4, R11to R14, R20, R30, and R40in Formula 1-1 may each independently be hydrogen, deuterium, —F, —Cl, —Br, —I, —SF5, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a substituted or unsubstituted C1-C60alkyl group, a substituted or unsubstituted C2-C60alkenyl group, a substituted or unsubstituted C2-C60alkynyl group, a substituted or unsubstituted C1-C60alkoxy group, a substituted or unsubstituted C1-C60alkylthio group, a substituted or unsubstituted C3-C10cycloalkyl group, a substituted or unsubstituted heterocycloalkyl group, a substituted or unsubstituted C3-C10cycloalkenyl group, a substituted or unsubstituted C2-C10heterocycloalkenyl group, a substituted or unsubstituted C6-C60aryl group, a substituted or unsubstituted C6-C60aryloxy group, a substituted or unsubstituted C6-C60arylthio group, a substituted or unsubstituted C1-C60heteroaryl group, a substituted or unsubstituted monovalent non-aromatic condensed polycyclic group, a substituted or unsubstituted monovalent non-aromatic condensed heteropolycyclic group, —N(Q1)(Q2), —Si(Q3)(Q4)(Q5), —B(Q6)(Q7), or —P(═O)(Q8)(Q9), and two or more neighboring R1to R4, R11to R14, R20, R30, and R40may optionally be linked together to form a substituted or unsubstituted C5-C30carbocyclic group or a substituted or unsubstituted C1-C30heterocyclic group. b1 and b3 in Formula 1-1 may each independently be an integer from 1 to 5, and when b1 is 2 or more, two or more of R1(s) may be identical to or different from each other, and when b3 is 2 or more, two or more of R3(s) may be identical to or different from each other. b20, b30, and b40 in Formula 1-1 may each independently be an integer from 1 to 10, and when b20 is 2 or more, two or more R20(s) may be identical to or different from each other, when b30 is 2 or more, two or more R30(s) may be identical to or different from each other, and when b40 is 2 or more, two or more R40(s) may be identical to or different from each other. R1to R4, R11to R14, R20, R30and R40in Formula 1-1 may each independently be hydrogen, deuterium, —F, —Cl, —Br, —I, —SF5, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a substituted or unsubstituted C1-C60alkyl group, a substituted or unsubstituted C2-C60alkenyl group, a substituted or unsubstituted C2-C60alkynyl group, a substituted or unsubstituted C1-C60alkoxy group, a substituted or unsubstituted C1-C60alkylthio group, a substituted or unsubstituted C3-C10cycloalkyl group, a substituted or unsubstituted heterocycloalkyl group, a substituted or unsubstituted C3-C10cycloalkenyl group, a substituted or unsubstituted C2-C10heterocycloalkenyl group, a substituted or unsubstituted C6-C60aryl group, a substituted or unsubstituted C6-C60aryloxy group, a substituted or unsubstituted C6-C60arylthio group, a substituted or unsubstituted C1-C60heteroaryl group, a substituted or unsubstituted monovalent non-aromatic condensed polycyclic group, a substituted or unsubstituted monovalent non-aromatic condensed heteropolycyclic group, —N(Q1)(Q2), —Si(Q3)(Q4)(Q5), —B(Q6)(Q7), or —P(═O)(Q8)(Q9), and two or more neighboring R1to R4, R11to R14, R20, R30, and R40may optionally be linked to form a substituted or unsubstituted C5-C30carbocyclic group or a substituted or unsubstituted C1-C30heterocyclic group. b1 and b3 in Formula 1-1 may each independently be an integer from 1 to 5, and when b1 is two or more, two or more R1(s) may be identical to or different from each other, and when b3 is two or more, two or more R3(s) may be identical to or different from each other. b20, b30, and b40 in Formula 1-1 may each independently be an integer from 1 to 10, and when b20 is two or more, two or more R20(s) may be identical to or different from each other, when b30 is two or more, two or more R30(s) may be identical to or different from each other, and when b40 is two or more, two or more R40(s) may be identical to or different from each other. According to one or more embodiments, R1to R4, R11to R14, R20, R30, and R40may each independently be:hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, —SF5, C1-C20alkyl group, or a C1-C20alkoxy group;a C1-C20alkyl group or a C1-C20alkoxy group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof;a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group;a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, an imidazopyrimidinyl group, or any combination thereof; or—N(Q1)(Q2), —Si(Q3)(Q4)(Q5), —B(Q6)(Q7), or —P(═O)(Q8)(Q9), andQ1to Q9may each independently be:—CH3, —CD3, —CD2H, —CDH2, —CH2CH3, —CH2CD3, —CH2CD2H, —CH2CDH2, —CHDCH3, —CHDCD2H, —CHDCDH2, —CHDCD3, —CD2CH3, —CD2CD3, —CD2CD2H, or —CD2CDH2,an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, a phenyl group, or a naphthyl group; oran n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, a phenyl group, or a naphthyl group, each substituted with at least one deuterium, a C1-C10alkyl group, a phenyl group, or any combination thereof. According to one or more embodiments, R1to R4, R11to R14, R20, R30and R40may each independently be hydrogen, deuterium, —F, a cyano group, a nitro group, —SF5, —CH3, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a group represented by one of Formulae 9-1 to 9-19, or a group represented by one of Formulae 10-1 to 10-194: In Formulae 9-1 to 9-19 and 10-1 to 10-194, * indicates a binding site to a neighboring atom, Ph is a phenyl group, and TMS is a trimethylsilyl group. In one or more embodiments, R20may not be hydrogen. In one or more embodiments, R20may be: deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, —SF5, a C1-C20alkyl group, or a C1-C20alkoxy group; a C1-C20alkyl group or a C1-C20alkoxy group, each substituted with at least one of deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof; a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group; a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, an imidazopyrimidinyl group, or any combination thereof; or —N(Q1)(Q2), —Si(Q3)(Q4)(Q5), —B(Q6)(Q7), or —P(═O)(Q8)(Q9), wherein Q1to Q9may each independently be: —CH3, —CD3, —CD2H, —CDH2, —CH2CH3, —CH2CD3, —CH2CD2H, —CH2CDH2, —CHDCH3, —CHDCD2H, —CHDCDH2, —CHDCD3, —CD2CH3, —CD2CD3, —CD2CD2H, or —CD2CDH2, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, a phenyl group, or a naphthyl group; or an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, a phenyl group, or a naphthyl group, each substituted with at least one deuterium, a C1-C10alkyl group, a phenyl group, or any combination thereof. In one or more embodiments, R20may be Formulae 9-1 to 9-12 and 10-1 to 10-194. In one or more embodiments, the organometallic compound represented by Formula 1 may be represented by one of Formulae 2-1 and 2-2: In Formulae 2-1 and 2-2, M1, Ar1and R11to R14are the same as described in the present specification, X1may be O or S, and X2may be a single bond, O, or S, R21to R23may each independently be the same as described in connection with R20, R31to R37may each independently be the same as described in connection with R30, R41to R44may each independently be the same as described in connection with R40, and two or more of neighboring R11to R14, R21to R23, R31to R37, and R41to R44may optionally be linked together to form a benzene ring or a naphthalene ring. R1to R4, R11to R14, R20, R30and R40in Formula 1 may each independently be hydrogen, deuterium, —F, —Cl, —Br, —I, —SF5, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a substituted or unsubstituted C1-C60alkyl group, a substituted or unsubstituted C2-C60alkenyl group, a substituted or unsubstituted C2-C60alkynyl group, a substituted or unsubstituted C1-C60alkoxy group, a substituted or unsubstituted C1-C60alkylthio group, a substituted or unsubstituted C3-C10cycloalkyl group, a substituted or unsubstituted heterocycloalkyl group, a substituted or unsubstituted C3-C10cycloalkenyl group, a substituted or unsubstituted C2-C10heterocycloalkenyl group, a substituted or unsubstituted C6-C60aryl group, a substituted or unsubstituted C6-C60aryloxy group, a substituted or unsubstituted C6-C60arylthio group, a substituted or unsubstituted C1-C60heteroaryl group, a substituted or unsubstituted monovalent non-aromatic condensed polycyclic group, a substituted or unsubstituted monovalent non-aromatic condensed heteropolycyclic group, —N(Q1)(Q2), —Si(Q3)(Q4)(Q5), —B(Q6)(Q7), or —P(═O)(Q8)(Q9), two or more neighboring R1to R4, R11to R14, R20, R30, and R40may optionally be linked to form a substituted or unsubstituted C6-C30carbocyclic group or a substituted or unsubstituted C1-C30heterocyclic group. According to one or more embodiments, R1to R4may optionally be linked, via a single bond, a double bond, or a first linking group, to form a C6-C30carbocyclic group unsubstituted or substituted with at least one R10a, or a C1-C30heterocyclic group unsubstituted or substituted with at least one R10a(for example, a fluorene group, an xanthene group, and an acridine group, each unsubstituted or substituted with at least one R10a). R10amay be the same as described in connection with R1. The first linking group may be *—N(R5)—*′, *—B(R5)—*′, *—P(R6)—*′, *—C(R5)(R6)—*′, *—Si(R5)(R6)—*′, *—Ge(R5)(R6)—*′, *—S—*′, *—O—*′, *—C(═O)—*′, *—S(═O)—*′, *—S(═O)2—*′, *—C(R5)═*′, *═C(R5)—*′, *—C(R5)═C(R6)—*′, *—C(═S)—*′, or *—C≡C—*′, and R5and R6are the same as described in connection with R1, and * and *′ each indicates a binding site to a neighboring atom. According to one or more embodiments, R1to R4, R11to R14, R20, R30, and R40may each independently be:hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, —SF5la C1-C20alkyl group, or a C1-C20alkoxy group;a C1-C20alkyl group or a C1-C20alkoxy group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof;a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group;a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, an imidazopyrimidinyl group, or any combination thereof; or—N(Q1)(Q2), —Si(Q3)(Q4)(Q5), —B(Q6)(Q7), or —P(═O)(Q8)(Q9);Q1to Q9may each independently be:CH3, —CD3, —CD2H, —CDH2, —CH2CH3, —CH2CD3, —CH2CD2H, —CH2CDH2, —CHDCH3, —CHDCD2H, —CHDCDH2, —CHDCD3, —CD2CH3, —CD2CD3, —CD2CD2H, or —CD2CDH2;an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, a phenyl group, or a naphthyl group; oran n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, a phenyl group, or a naphthyl group, each substituted with at least one deuterium, a C1-C10alkyl group, a phenyl group, or any combination thereof. For example, R1to R4may each independently be:a C1-C30alkyl group;a C1-C30alkyl group, substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a C1-C10alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof;a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group; ora phenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof. According to one or more embodiments, R1to R4, R11to R14, R20, R30, and R40may each independently be: hydrogen, deuterium, —F, a cyano group, a nitro group, —SF5, a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, an n-hexyl group, an isohexyl group, a sec-hexyl group, a tert-hexyl group, an n-heptyl group, an isoheptyl group, a sec-heptyl group, a tert-heptyl group, an n-octyl group, an isooctyl group, a sec-octyl group, a tert-octyl group, an n-nonyl group, an isononyl group, a sec-nonyl group, a tert-nonyl group, an n-decyl group, an isodecyl group, a sec-decyl group, a tert-decyl group, a methoxy group, an ethoxy group, a propoxy group, a butoxy group, a pentoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, a carbazolyl group, a dibenzofuranyl group, or a dibenzothiophenyl group; a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, an n-hexyl group, an isohexyl group, a sec-hexyl group, a tert-hexyl group, an n-heptyl group, an isoheptyl group, a sec-heptyl group, a tert-heptyl group, an n-octyl group, an isooctyl group, a sec-octyl group, a tert-octyl group, an n-nonyl group, an isononyl group, a sec-nonyl group, a tert-nonyl group, an n-decyl group, an isodecyl group, a sec-decyl group, a tert-decyl group, a methoxy group, an ethoxy group, a propoxy group, a butoxy group, a pentoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, a carbazolyl group, a dibenzofuranyl group, or a dibenzothiophenyl group, each substituted with at least one deuterium, —F, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a cyano group, a nitro group, a C1-C10alkyl group, a C1-C10alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, a carbazolyl group, a dibenzofuranyl group, a dibenzothiophenyl group, or any combination thereof; or —N(Q1)(Q2), —Si(Q3)(Q4)(Q5), —B(Q6)(Q7), or —P(═O)(Q8)(Q9), and Q1to Q9may each independently be; —CH3, —CD3, —CD2H, —CDH2, —CH2CH3, —CH2CD3, —CH2CD2H, —CH2CDH2, —CHDCH3, —CHDCD2H, —CHDCDH2, —CHDCD3, —CD2CH3, —CD2CD3, —CD2CD2H, or —CD2CDH2;an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, a phenyl group, or a naphthyl group; oran n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a sec-pentyl group, a tert-pentyl group, a phenyl group, or a naphthyl group, each substituted with at least one deuterium, a C1-C10alkyl group, a phenyl group, or any combination thereof. In one or more embodiments, two or more neighboring groups of R1to R4, R11to R14, R20, R30, and R40may optionally be linked to form a substituted or unsubstituted C5-C30carbocyclic group or a substituted or unsubstituted C1-C30heterocyclic group. For example, two or more neighboring groups of R1to R4, R11to R14, R20, R30, and R40in Formula 1 may be bonded to each other to form a cyclopentane group, a cyclopentadiene group, a furan group, a thiophene group, a pyrrole group, a silole group, an adamantane group, a norbornane group, a norbornene group, a cyclopentene group, a cyclohexane group, a cyclohexene group, a benzene group, a naphthalene group, an indene group, an indole group, a benzofuran group, a benzothiophene group, a benzosilole group, a fluorene group, a carbazole group, a dibenzofuran group, a dibenzothiophene group, or a dibenzosilole group, each unsubstituted or substituted with at least one R10a. At least one of substituents of the substituted C5-C30carbocyclic group, substituted C1-C30heterocyclic group, substituted phenyl group, substituted C1-C60alkyl group, substituted C2-C60alkenyl group, substituted C2-C60alkynyl group, substituted C1-C60alkoxy group, substituted C1-C60alkylthio group, substituted C3-C10cycloalkyl group, substituted C1-C10heterocycloalkyl group, substituted C3-C10cycloalkenyl group, substituted C2-C10heterocycloalkenyl group, substituted C6-C60aryl group, substituted C6-C60aryloxy group, substituted C6-C60arylthio group, substituted C1-C60heteroaryl group, substituted monovalent non-aromatic condensed polycyclic group, and substituted monovalent non-aromatic condensed heteropolycyclic group may be:deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, or any combination thereof;a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, or a C1-C60alkoxy group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, a monovalent non-aromatic condensed heteropolycyclic group, —N(Q11)(Q12), —Si(Q13)(Q14)(Q15), —B(Q16)(Q17), —P(═O)(Q18)(Q19), or any combination thereof;a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, or a monovalent non-aromatic condensed heteropolycyclic group;a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, or a monovalent non-aromatic condensed heteropolycyclic group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, a monovalent non-aromatic condensed heteropolycyclic group, —N(Q21)(Q22), —Si(Q23)(Q24)(Q25), —B(Q26)(Q27), —P(═O)(Q28)(Q29), or any combination thereof; or—N(Q31)(Q32), —Si(Q33)(Q34)(Q35), —B(Q36)(Q37), or —P(═O)(Q38)(Q39),wherein Q1to Q9, Q11to Q19, Q21to Q29, and Q31to Q39may each independently be hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryl group substituted with at least one a C1-C60alkyl group, a C6-C60aryl group, or any combination thereof, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, or a monovalent non-aromatic condensed heteropolycyclic group. L12in Formula 1 may be a monodentate ligand or a bidentate ligand. For example, L12in Formula 1 may be a ligand represented by one of Formulae 7-1 to 7-11, but embodiments are not limited thereto: In Formulae 7-1 to 7-11, A71and A72may each independently be a C5-C20carbocyclic group or a C1-C20heterocyclic group, X71and X72may each independently be C or N, X73may be N or C(Q73); X74may be N or C(Q74); X75may be N or C(Q75); X76may be N or C(Q76), and X77may be N or C(Q77), X78may be O, S or N(Q78); and X79may be O, S, or N(Q79), Y71and Y72may each independently be a single bond, a double bond, a substituted or unsubstituted C1-C5alkylene group, a substituted or unsubstituted C2-C5alkenylene group, or a substituted or unsubstituted C6-C10arylene group, Z71and Z72may each independently be N, O, N(R74), P(R75)(R76), or As(R75)(R76), Z73may be P or As, Z74may be CO or CH2, R71to R80and Q73to Q79may each independently be hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a substituted or unsubstituted C1-C60alkyl group, a substituted or unsubstituted C2-C60alkenyl group, a substituted or unsubstituted C2-C60alkynyl group, a substituted or unsubstituted C1-C60alkoxy group, a substituted or unsubstituted C3-C10cycloalkyl group, a substituted or unsubstituted heterocycloalkyl group, a substituted or unsubstituted C3-C10cycloalkenyl group, a substituted or unsubstituted C2-C10heterocycloalkenyl group, a substituted or unsubstituted C6-C60aryl group, a substituted or unsubstituted C6-C60aryloxy group, a substituted or unsubstituted C6-C60arylthio group, a substituted or unsubstituted C1-C60heteroaryl group, a substituted or unsubstituted monovalent non-aromatic condensed polycyclic group, or a substituted or unsubstituted monovalent non-aromatic condensed heteropolycyclic group; R71and R72may optionally be bonded to form a ring; R77and R78may optionally be bonded to form a ring; R78and R79may optionally be bonded to form a ring; and R79and R80may optionally be bonded to form a ring, b71 and b72 may each independently be 1, 2, and 3, and * and *′ each independently indicate a binding site to a neighboring atom. For example, A71and A72in Formulae 7-1, 7-9, and 7-11 may each independently be a benzene group, a naphthalene group, an imidazole group, a benzimidazole group, a pyridine group, a pyrazine group, a pyrimidine group, a triazine group, a quinoline group, or an isoquinoline group, but embodiments of the present disclosure are not limited thereto. For example, X72and X79in Formulae 7-1, 7-9, and 7-11 may be N, but embodiments of the present disclosure are not limited thereto. For example, in Formula 7-7, X73may be C(Q73); X74may be C(Q74); X75may be C(Q75); X76may be C(Q76); and X77may be C(Q77), but embodiments of the present disclosure are not limited thereto. For example, in Formula 7-8, X78may be N(Q79), and X79may be N(Q79), but embodiments of the present disclosure are not limited thereto. For example, Y71and Y72in Formulae 7-2, 7-3, 7-8, 7-9, and 7-11 may each independently be a substituted or unsubstituted methylene group or a substituted or unsubstituted phenylene group, but embodiments of the present disclosure are not limited thereto. For example, Z71and Z72in Formulae 7-1 and 7-2 may be 0, but embodiments of the present disclosure are not limited thereto. For example, Z73in Formula 7-4 may be P, but embodiments of the present disclosure are not limited thereto. For example, R71to R80and Q73to Q76in Formulae 7-1 to 7-11 may each independently be: hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, —SF5, C1-C20alkyl group, or a C1-C20alkoxy group; a C1-C20alkyl group or a C1-C20alkoxy group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof; a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a dibenzosilolyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group; a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a dibenzosilolyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a dibenzosilolyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, an imidazopyrimidinyl group, —Si(Q11)(Q12)(Q13), —B(Q11)(Q12), —N(Q11)(Q12), or any combination thereof; or —Si(Q1)(Q2)(Q3), —B(Q1)(Q2), or —N(Q1)(Q2), wherein Q1to Q3and Q11to Q13may each independently be: a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a 2-methylbutyl group, a sec-pentyl group, a tert-pentyl group, a neo-pentyl group, 3-pentyl group, 3-methyl-2-butyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, or a naphthyl group; or a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a 2-methylbutyl group, a sec-pentyl group, a tert-pentyl group, a neo-pentyl group, 3-pentyl group, 3-methyl-2-butyl group, a phenyl group, or a naphthyl group, each substituted with at least one deuterium, a phenyl group, or any combination thereof, but embodiments of the present disclosure are not limited thereto. L12in Formula 1 may be a ligand represented by one of Formulae 5-1 to 5-116 and 8-1 to 8-23, but embodiments of the present disclosure are not limited thereto: In Formulae 5-1 to 5-116 and 8-1 to 8-23, R51to R53may each independently be: hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, —SF5, C1-C20alkyl group, or a C1-C20alkoxy group; a C1-C20alkyl group or a C1-C20alkoxy group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, a naphthyl group, a pyridinyl group, a pyrimidinyl group, or any combination thereof; a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a dibenzosilolyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group; a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbomanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a dibenzosilolyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, or an imidazopyrimidinyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazino group, a hydrazono group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, an adamantanyl group, a norbornanyl group, a norbornenyl group, a cyclopentenyl group, a cyclohexenyl group, a cycloheptenyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, a naphthyl group, a fluorenyl group, a phenanthrenyl group, an anthracenyl group, a fluoranthenyl group, a triphenylenyl group, a pyrenyl group, a chrysenyl group, a pyrrolyl group, a thiophenyl group, a furanyl group, an imidazolyl group, a pyrazolyl group, a thiazolyl group, an isothiazolyl group, an oxazolyl group, an isoxazolyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an isoindolyl group, an indolyl group, an indazolyl group, a purinyl group, a quinolinyl group, an isoquinolinyl group, a benzoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group, a carbazolyl group, a phenanthrolinyl group, a benzimidazolyl group, a benzofuranyl group, a benzothiophenyl group, an isobenzothiazolyl group, a benzoxazolyl group, an isobenzoxazolyl group, a triazolyl group, a tetrazolyl group, an oxadiazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a dibenzosilolyl group, a benzocarbazolyl group, a dibenzocarbazolyl group, an imidazopyridinyl group, an imidazopyrimidinyl group, —Si(Q11)(Q12)(Q13), —B(Q11)(Q12), —N(Q11)(Q12), or any combination thereof; or —Si(Q1)(Q2)(Q3), —B(Q1)(Q2), or —N(Q1)(Q2), wherein Q1to Q3and Q11to Q13may each independently be: a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a 2-methylbutyl group, a sec-pentyl group, a tert-pentyl group, a neo-pentyl group, 3-pentyl group, 3-methyl-2-butyl group, a phenyl group, a biphenyl group, a C1-C20alkylphenyl group, or a naphthyl group; or a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an n-pentyl group, an isopentyl group, a 2-methylbutyl group, a sec-pentyl group, a tert-pentyl group, a neo-pentyl group, 3-pentyl group, 3-methyl-2-butyl group, a phenyl group, or a naphthyl group, each substituted with at least one deuterium, a phenyl group, or any combination thereof, b51 and b54 may each independently be 1 or 2, b53 and b55 may each independently be 1, 2, or 3, b52 may be 1, 2, 3, or 4, Ph indicates a phenyl group, Ph-d5 indicates a phenyl group in which all hydrogen are substituted with deuterium, and * and *′ each indicate a binding site to a neighboring atom. In Formula 1, n11 may be 1 and n12 may be 0, 1, or 2. In detail, in Formula 1, M1may be Pt, n11 may be 1, and n12 may be 0, but embodiments of the present disclosure are not limited thereto. In one or more embodiments, the organometallic compound may be one of Compounds 1 to 325: The organometallic compound represented by Formula 1 may satisfy the structure of Formula 1 described above, and due to the structure in which the benzimidazole moiety in L11ligand is N-substituted with an Ar1group, the organometallic compound may have improved photochemical stability, and may be suitable for deep blue light emission. An electronic device, for example, an organic light-emitting device, using the organometallic compound represented by Formula 1 may be excellent in luminescence efficiency, lifespan, and color purity. The Ar1group satisfies the structure of Formula Ar1-1, and due to the inductive effect of substituent E1in Formula Ar1-1, the efficiency of organic light-emitting device may be improved. For example, since E1is located at the meta position of Ar1group, compared to the substitution at the ortho position, the bending of the bond angle of a molecule may occur less, and thus, the bonding force may be sufficiently maintained, and accordingly, the lifespan of organic light-emitting device may be improved. In addition, regarding substituent Ar1, E2to E5in Formula Ar1-1 are not an alkyl group, and thus, deterioration of a compound by the alkyl group and the decrease in lifespan of the organic light-emitting device due to impurities generated therefrom can be prevented. In addition, since Ar1has an asymmetric structure, the organometallic compound may be less likely to crystallize, and thus, the lifespan of the organic light-emitting device may be improved. In one or more embodiments, the highest occupied molecular orbital (HOMO), lowest unoccupied molecular orbital (LUMO), triplet (T1) energy level, and spin density of compounds 1 and 2 and comparative compounds A to F are evaluated by using DFT method of Gaussian program (structurally optimized at the level of B3LYP, 6-31G(d,p)). Results thereof are shown in Table 1. TABLE 1T1CompoundHOMOLUMOenergySpinNo.(eV)(eV)level (eV)densityCompound 1−4.61−1.202.620.381Compound 2−4.66−1.242.640.379Comparative−4.67−1.212.670.356Example AComparative−4.64−1.202.640.377Example BComparative−4.61−1.202.620.385Example CComparative−4.64−1.192.650.378Example DComparative−4.61−1.192.630.377Example EComparative−4.65−1.222.650.367Example F From Table 1, it is confirmed that the organometallic compound represented by Formula 1 has such electric characteristics that are suitable for use as a material for an emission layer of an electronic device, for example, an organic light-emitting device. Also, the organometallic compound represented by Formula 1 provides high spin densities compared to the comparative compounds, and accordingly, the metal to ligand charge transfer (MLCT) effectively occur, and the efficiency and lifespan of an organic light-emitting device may be improved. Synthesis methods of the organometallic compound represented by Formula 1 may be recognizable by one of ordinary skill in the art by referring to Synthesis Examples provided below. Accordingly, since the organometallic compound represented by Formula 1 is suitable for use in an organic layer, for example, an emission layer of an organic light-emitting device, another aspect provides an organic light-emitting device including: a first electrode; a second electrode; and an organic layer between the first electrode and the second electrode and including an emission layer, wherein the organic layer includes at least one of the organometallic compounds represented by Formula 1. Since the organic light-emitting device includes the organic layer including the organometallic compounds represented by Formula 1 described above, the organic light-emitting device has a low driving voltage, high efficiency, high power efficiency, high quantum efficiency, a long lifespan, a low roll-off ratio, and excellent color purity. In one or more embodiments, in the organic light-emitting device, the first electrode is an anode, and the second electrode is a cathode, and the organic layer further includes a hole transport region between the first electrode and the emission layer and an electron transport region between the emission layer and the second electrode, and the hole transport region includes a hole injection layer, a hole transport layer, an electron blocking layer, or any combination thereof, and the electron transport region includes a hole blocking layer, an electron transport layer, an electron injection layer, or any combination thereof. In one or more embodiments, the organometallic compound represented by Formula 1 may be included in the emission layer. The organometallic compound included in the emission layer may act as an emitter. For example, the emission layer including the organometallic compound represented by Formula 1 may emit phosphorescence generated when the triplet exciton of the organometallic compound is transferred to the ground state. In one or more embodiments, the emission layer including the organometallic compound represented by Formula 1 may further include a host. The host may be any host, and may be understood by referring to the description provided herein in connection with the host. The amount of host in the emission layer may be greater than the amount of organometallic compound represented by Formula 1. In one or more embodiments, the emission layer may include a host and a dopant, the host may be any host, and the dopant may include the organometallic compound represented by Formula 1. The emission layer may emit phosphorescence generated when the triplet exciton of the organometallic compound, acting as a dopant, is transferred to a ground state. According to one or more embodiments, when the emission layer further includes a host, the amount of the host may be greater than the amount of the organometallic compound. In one or more embodiments, the emission layer may include a host and a dopant, the host may be any host, the dopant may include the organometallic compound represented by Formula 1, and the emission layer may further include a fluorescent dopant. The emission layer may emit fluorescence generated by transition of when the triplet exciton of the organometallic compound which has been delivered to the fluorescent dopant. According to an embodiment, the emission layer may emit blue light having a maximum emission wavelength of about 410 nm to about 490 nm. The expression “(an organic layer) includes at least one of organometallic compounds” used herein may include a case in which “(an organic layer) includes identical organometallic compounds represented by Formula 1” and a case in which “(an organic layer) includes two or more different organometallic compounds represented by Formula 1.” For example, the organic layer may include, as the organometallic compound, only Compound 1. In this regard, Compound 1 may exist in an emission layer of the organic light-emitting device. In one or more embodiments, the organic layer may include, as the organometallic compound, Compound 1 and Compound 2. In this regard, Compound 1 and Compound 2 may exist in an identical layer (for example, Compound 1 and Compound 2 all may exist in an emission layer). The term “organic layer” used herein refers to a single layer and/or a plurality of layers between the first electrode and the second electrode of the organic light-emitting device. The “organic layer” may include, in addition to an organic compound, an organometallic complex including metal. FIGURE is a schematic view of an organic light-emitting device10according to one embodiment. Hereinafter, the structure of an organic light-emitting device according to an exemplary embodiment and a method of manufacturing an organic light-emitting device according to an embodiment will be described in connection with FIGURE. The organic light-emitting device10includes a first electrode11, an organic layer15, and a second electrode19, which are sequentially stacked. A substrate may be additionally located under the first electrode11or above the second electrode19. For use as the substrate, any substrate that is used in organic light-emitting devices available in the art may be used, and the substrate may be a glass substrate or a transparent plastic substrate, each having excellent mechanical strength, thermal stability, transparency, surface smoothness, ease of handling, and water resistance. In one or more embodiments, the first electrode11may be formed by depositing or sputtering a material for forming the first electrode11on the substrate. The first electrode11may be an anode. The material for forming the first electrode11may be materials with a high work function to facilitate hole injection. The first electrode11may be a reflective electrode, a semi-transmissive electrode, or a transmissive electrode. The material for forming the first electrode11may be indium tin oxide (ITO), indium zinc oxide (IZO), tin oxide (SnO2), or zinc oxide (ZnO). In one or more embodiments, the material for forming the first electrode11may be metal, such as magnesium (Mg), aluminum (Al), aluminum-lithium (Al—Li), calcium (Ca), magnesium-indium (Mg—In), or magnesium-silver (Mg—Ag). The first electrode11may have a single-layered structure or a multi-layered structure including two or more layers. For example, the first electrode11may have a three-layered structure of ITO/Ag/ITO, but the structure of the first electrode11is not limited thereto. The organic layer15is located on the first electrode11. The organic layer15may include a hole transport region, an emission layer, and an electron transport region. The hole transport region may be between the first electrode11and the emission layer. The hole transport region may include a hole injection layer, a hole transport layer, an electron blocking layer, a buffer layer, or any combination thereof. The hole transport region may include only either a hole injection layer or a hole transport layer. In one or more embodiments, the hole transport region may have a hole injection layer/hole transport layer structure or a hole injection layer/hole transport layer/electron blocking layer structure, which are sequentially stacked in this stated order from the first electrode11. When the hole transport region includes a hole injection layer (HIL), the hole injection layer may be formed on the first electrode11by using one or more suitable methods, for example, vacuum deposition, spin coating, casting, and/or Langmuir-Blodgett (LB) deposition. When a hole injection layer is formed by vacuum deposition, the deposition conditions may vary according to a material that is used to form the hole injection layer, and the structure and thermal characteristics of the hole injection layer. For example, the deposition conditions may include a deposition temperature of about 100 to about 500° C., a vacuum pressure of about 10−8torr to about 10−3torr, and a deposition rate of about 0.01 Å/sec to about 100 Å/sec. However, the deposition conditions are not limited thereto. When the hole injection layer is formed using spin coating, coating conditions may vary according to the material used to form the hole injection layer, and the structure and thermal properties of the hole injection layer. For example, a coating speed may be from about 2,000 rpm to about 5,000 rpm, and a temperature at which a heat treatment is performed to remove a solvent after coating may be from about 80° C. to about 200° C. However, the coating conditions are not limited thereto. Conditions for forming a hole transport layer and an electron blocking layer may be understood by referring to conditions for forming the hole injection layer. The hole transport region may include at least one of m-MTDATA, TDATA, 2-TNATA, NPB, β-NPB, TPD, Spiro-TPD, Spiro-NPB, methylated-NPB, TAPC, HMTPD, 4,4′,4″-tris(N-carbazolyl)triphenylamine (TCTA), polyaniline/dodecylbenzenesulfonic acid (PAN I/DBSA), poly(3,4-ethylenedioxythiophene)/poly(4-styrenesulfonate) (PEDOT/PSS), polyaniline/camphor sulfonicacid (PANI/CSA), polyaniline/poly(4-styrenesulfonate) (PANI/PSS), a compound represented by Formula 201 below, a compound represented by Formula 202 below, or any combination thereof: Ar101and Ar102in Formula 201 may each independently be: a phenylene group, a pentalenylene group, an indenylene group, a naphthylene group, an azulenylene group, a heptalenylene group, an acenaphthylene group, a fluorenylene group, a phenalenylene group, a phenanthrenylene group, an anthracenylene group, a fluoranthenylene group, a triphenylenylene group, a pyrenylene group, a chrysenylenylene group, a naphthacenylene group, a picenylene group, a perylenylene group, or a pentacenylene group; or a phenylene group, a pentalenylene group, an indenylene group, a naphthylene group, an azulenylene group, a heptalenylene group, an acenaphthylene group, a fluorenylene group, a phenalenylene group, a phenanthrenylene group, an anthracenylene group, a fluoranthenylene group, a triphenylenylene group, a pyrenylene group, a chrysenylenylene group, a naphthacenylene group, a picenylene group, a perylenylene group, or a pentacenylene group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, a C3-C10cycloalkyl group, a C3-C10cycloalkenyl group, a C1-C10heterocycloalkyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryloxy group, a C8-C80arylthio group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, a monovalent non-aromatic condensed heteropolycyclic group, or any combination thereof. xa and xb in Formula 201 may each independently be an integer from 0 to 5, or 0, 1, or 2. For example, xa may be 1 and xb may be 0, but xa and xb are not limited thereto. R101to R108, R111to R119and R121to R124in Formulae 201 and 202 may each independently be: hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group (for example, a methyl group, an ethyl group, a propyl group, a butyl group, a pentyl group, a hexyl group, and so on), or a C1-C10alkoxy group (for example, a methoxy group, an ethoxy group, a propoxy group, a butoxy group, a pentoxy group, and so on); a C1-C10alkyl group or a C1-C10alkoxy group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, and a phosphoric acid group or a salt thereof, or any combination thereof; a phenyl group, a naphthyl group, an anthracenyl group, a fluorenyl group, or a pyrenyl group; or a phenyl group, a naphthyl group, an anthracenyl group, a fluorenyl group, or a pyrenyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C10alkyl group, a alkoxy group, or any combination thereof, but embodiments of the present disclosure are not limited thereto. R109in Formula 201 may be: a phenyl group, a naphthyl group, an anthracenyl group, ora pyridinyl group; or a phenyl group, a naphthyl group, an anthracenyl group, or a pyridinyl group, each substituted with at least one a deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a phenyl group, a naphthyl group, an anthracenyl group, a pyridinyl group, or any combination thereof. According to an embodiment, the compound represented by Formula 201 may be represented by Formula 201A below, but embodiments of the present disclosure are not limited thereto: R101, R111, R112, and R109in Formula 201A may be understood by referring to the description provided herein. For example, the compound represented by Formula 201 and the compound represented by Formula 202 may include compounds HT1 to HT20 illustrated below, but are not limited thereto: A thickness of the hole transport region may be in a range of about 100 Å to about 10,000 Å, for example, about 100 Å to about 1,000 Å. When the hole transport region includes at least one of a hole injection layer and a hole transport layer, a thickness of the hole injection layer may be in a range of about 100 Å to about 10,000 Å, for example, about 100 Å to about 1,000 Å, and a thickness of the hole transport layer may be in a range of about 50 Å to about 2,000 Å, for example about 100 Å to about 1,500 Å. When the thicknesses of the hole transport region, the hole injection layer and the hole transport layer are within these ranges, satisfactory hole transporting characteristics may be obtained without a substantial increase in driving voltage. The hole transport region may further include, in addition to these materials, a charge-generation material for the improvement of conductive properties. The charge-generation material may be homogeneously or non-homogeneously dispersed in the hole transport region. The charge-generation material may be, for example, a p-dopant. The p-dopant may be one of a quinone derivative, a metal oxide, and a cyano group-containing compound, but embodiments of the present disclosure are not limited thereto. Non-limiting examples of the p-dopant are a quinone derivative, such as tetracyanoquinonedimethane (TCNQ) or 2,3,5,6-tetrafluoro-tetracyano-1,4-benzoquinonedimethane (F4-TCNQ); a metal oxide, such as a tungsten oxide or a molybdenum oxide; and a cyano group-containing compound, such as Compound HT-D1 below, but are not limited thereto. The hole transport region may include a buffer layer. Also, the buffer layer may compensate for an optical resonance distance according to a wavelength of light emitted from the emission layer, and thus, efficiency of a formed organic light-emitting device may be improved. Then, an emission layer (EML) may be formed on the hole transport region by vacuum deposition, spin coating, casting, LB deposition, or the like. When the emission layer is formed by vacuum deposition or spin coating, the deposition or coating conditions may be similar to those applied in forming the hole injection layer although the deposition or coating conditions may vary according to a material that is used to form the emission layer. Meanwhile, when the hole transport region includes an electron blocking layer, a material for the electron blocking layer may be materials for the hole transport region described above and materials for a host to be explained later. However, the material for the electron blocking layer is not limited thereto. For example, when the hole transport region includes an electron blocking layer, a material for the electron blocking layer may be mCP, which will be explained later. The emission layer may include a host and a dopant, and the dopant may include the organometallic compound represented by Formula 1. The host may include at least one TPBi, TBADN, ADN (also referred to as “DNA”), CBP, CDBP, TCP, mCP, Compound H50, Compound H51, or any combination thereof: In one or more embodiments, the host may further include a compound represented by Formula 301 below. Ar111and Ar112in Formula 301 may each independently be: a phenylene group, a naphthylene group, a phenanthrenylene group, or a pyrenylene group; or a phenylene group, a naphthylene group, a phenanthrenylene group, or a pyrenylene group, each substituted with at least one a phenyl group, a naphthyl group, an anthracenyl group, or any combination thereof. Ar113to Ar116in Formula 301 may each independently be:a C1-C10alkyl group, a phenyl group, a naphthyl group, a phenanthrenyl group, or a pyrenyl group; ora phenyl group, a naphthyl group, a phenanthrenyl group, or a pyrenyl group, each substituted with at least one a phenyl group, a naphthyl group, an anthracenyl group, or any combination thereof. g, h, i, and j in Formula 301 may each independently be an integer from 0 to 4, and may be, for example, 0, 1, or 2. Ar113to Ar116in Formula 301 may each independently be:a C1-C10alkyl group, substituted with at least one a phenyl group, a naphthyl group, an anthracenyl group, or any combination thereof;a phenyl group, a naphthyl group, an anthracenyl group, a pyrenyl, a phenanthrenyl group, or a fluorenyl group;a phenyl group, a naphthyl group, an anthracenyl group, a pyrenyl group, a phenanthrenyl group, or a fluorenyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, a phenyl group, a naphthyl group, an anthracenyl group, a pyrenyl group, a phenanthrenyl group, a fluorenyl group, or any combination thereof; or but embodiments of the present disclosure are not limited thereto. In one or more embodiments, the host may include a compound represented by Formula 302 below: Ar122to Ar125in Formula 302 are the same as described in detail in connection with Ar113in Formula 301. Ar126and Ar127in Formula 302 may each independently be a C1-C10alkyl group (for example, a methyl group, an ethyl group, or a propyl group). k and l in Formula 302 may each independently be an integer from 0 to 4. For example, k and l may be 0, 1, or 2. When the organic light-emitting device is a full-color organic light-emitting device, the emission layer may be patterned into a red emission layer, a green emission layer, and a blue emission layer. In one or more embodiments, due to a stacked structure including a red emission layer, a green emission layer, and/or a blue emission layer, the emission layer may emit white light. When the emission layer includes a host and a dopant, an amount of the dopant may be in a range of about 0.01 parts by weight to about 15 parts by weight based on 100 parts by weight of the host, but embodiments of the present disclosure are not limited thereto. In one or more embodiments, the organic layer of the organic light-emitting device may further include a fluorescent dopant in addition to the organometallic compound represented by Formula 1. For example, the fluorescent dopant may be a condensed polycyclic compound or a styryl compound. For example, the fluorescent dopant may include one of a naphthalene-containing core, a fluorene-containing core, a spiro-bifluorene-containing core, a benzofluorene-containing core, a dibenzofluorene-containing core, a phenanthrene-containing core, an anthracene-containing core, a fluoranthene-containing core, a triphenylene-containing core, a pyrene-containing core, a chrysene-containing core, a naphthacene-containing core, a picene-containing core, a perylene-containing core, a pentaphene-containing core, an indenoanthracene-containing core, a tetracene-containing core, a bisanthracene-containing core, and cores represented by Formulae 501-1 to 501-18, but embodiments of the present disclosure are not limited thereto: In one or more embodiments, the fluorescent dopant may be a styryl-amine-based compound and a styryl-carbazole-based compound, but embodiments of the present disclosure are not limited thereto. In one or more embodiments, the fluorescent dopant may be compounds represented by Formula 501: In Formula 501, Ar501may be: a naphthalene group, a fluorene group, a spiro-bifluorene group, a benzofluorene group, a dibenzofluorene group, a phenanthrene group, an anthracene group, a fluoranthene group, a triphenylene group, a pyrene group, a chrysene group, a naphthacene group, a picene group, a perylene group, a pentaphene group, an indenoanthracene group, a tetracene group, a bisanthracene group, or groups represented by Formulae 501-1 to 501-18; or a naphthalene group, a fluorene group, a spiro-bifluorene group, a benzofluorene group, a dibenzofluorene group, a phenanthrene group, an anthracene group, a fluoranthene group, a triphenylene group, a pyrene group, a chrysene group, a naphthacene group, a picene group, a perylene group, a pentaphene group, an indenoanthracene group, a tetracene group, a bisanthracene group, or groups represented by Formulae 501-1 to 501-18, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, a monovalent non-aromatic condensed heteropolycyclic group, —Si(Q501)(Q502)(Q503) (wherein Q501to Q503may each independently be hydrogen, C1-C60alkyl group, a C1-C60alkoxy group, a C6-C60aryl group, a C1-C60heteroaryl group, a monovalent non-aromatic condensed polycyclic group, or a monovalent non-aromatic condensed heteropolycyclic group, or any combination thereof; L501to L503may each independently be a substituted or unsubstituted C3-C10cycloalkylene group, a substituted or unsubstituted heterocycloalkylene group, a substituted or unsubstituted C3-C10cycloalkenylene group, a substituted or unsubstituted C2-C10heterocycloalkenylene group, a substituted or unsubstituted C6-C60arylene group, a substituted or unsubstituted C1-C60heteroarylene group, a substituted or unsubstituted divalent non-aromatic condensed polycyclic group, or a substituted or unsubstituted divalent non-aromatic condensed heteropolycyclic group, and R501and R502may each independently be: a phenyl group, a biphenyl group, a terphenyl group, a naphthyl group, a fluorenyl group, a spiro-bifluorenyl group, a benzofluorenyl group, a dibenzofluorenyl group, a phenanthrenyl group, an anthracenyl group, a pyrenyl group, a chrysenyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, a quinolinyl group, an isoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a carbazole group, a triazinyl group, a dibenzofuranyl group, or a dibenzothiophenyl group; or a phenyl group, a biphenyl group, a terphenyl group, a naphthyl group, a fluorenyl group, a spiro-bifluorenyl group, a benzofluorenyl group, a dibenzofluorenyl group, a phenanthrenyl group, an anthracenyl group, a pyrenyl group, a chrysenyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, a quinolinyl group, an isoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a carbazolyl group, a triazinyl group, a dibenzofuranyl group, or a dibenzothiophenyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a phenyl group, a biphenyl group, a terphenyl group, a naphthyl group, a fluorenyl group, a spiro-bifluorenyl group, a benzofluorenyl group, a dibenzofluorenyl group, a phenanthrenyl group, an anthracenyl group, a pyrenyl group, a chrysenyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, a quinolinyl group, an isoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a carbazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, or any combination thereof, xd1 to xd3 may each independently be 0, 1, 2, or 3, and xd4 may be 0, 1, 2, 3, 4, 5, or 6. For example, in Formula 501, Ar501may be: a naphthalene group, a fluorene group, a spiro-bifluorene group, a benzofluorene group, a dibenzofluorene group, a phenanthrene group, an anthracene group, a fluoranthene group, a triphenylene group, a pyrene group, a chrysene group, a naphthacene group, a picene group, a perylene group, a pentaphene group, an indenoanthracene group, a tetracene group, a bisanthracene group, or groups represented by Formulae 501-1 to 501-18; or a naphthalene group, a fluorene group, a spiro-bifluorene group, a benzofluorene group, a dibenzofluorene group, a phenanthrene group, an anthracene group, a fluoranthene group, a triphenylene group, a pyrene group, a chrysene group, a naphthacene group, a picene group, a perylene group, a pentaphene group, an indenoanthracene group, a tetracene group, a bisanthracene group, or groups represented by Formula 501-1 to 501-18, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a phenyl group, a biphenyl group, a terphenyl group, a naphthyl group, a fluorenyl group, a dibenzofuranyl group, a dibenzothiophenyl group, a carbazolyl group, a pyridinyl group, a pyrimidinyl group, a triazinyl group, a quinolinyl group, an isoquinolinyl group, —Si(Q501)(Q502)(Q503) (Q501to Q503may each independently be hydrogen, a C1-C20alkyl group, a C1-C20alkoxy group, a phenyl group, a biphenyl group, a terphenyl group, or a naphthyl group), or any combination thereof, L501to L503are the same as described in connection with L1, xd1 to xd3 may each independently be 0, 1, or 2, and xd4 may be 0, 1, 2, or 3, but embodiments of the present disclosure are not limited thereto. In one or more embodiments, the fluorescent dopant may include a compound represented by one of Formulae 502-1 to 502-5: In Formulae 502-1 to 502-5, X51may be N or C-[(L501)xd1-R501], X52may be N or C-[(L502)xd2-R502], X53may be N or C-[(L503)xd3-R503], X54may be N or C-[(L504)xd4-R504], X55may be N or C-[(L505)xd5-R505], X56may be N or C-[(L506)xd6-R506], X57may be N or C-[(L507)xd7-R507], and X58may be N or C-[(L508)xd8-R508], L501to L508are each the same as described in connection with L501in Formula 501, xd1 to xd8 are each the same as described in connection with xd1 in Formula 501, R501to R508may each independently be:hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, or a C1-C20alkoxy group;a phenyl group, a biphenyl group, a terphenyl group, a naphthyl group, a fluorenyl group, a spiro-bifluorenyl group, a benzofluorenyl group, a dibenzofluorenyl group, a phenanthrenyl group, an anthracenyl group, a pyrenyl group, a chrysenyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, a quinolinyl group, an isoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a carbazole group, a triazinyl group, a dibenzofuranyl group, or a dibenzothiophenyl group; or a phenyl group, a biphenyl group, a terphenyl group, a naphthyl group, a fluorenyl group, a spiro-bifluorenyl group, a benzofluorenyl group, a dibenzofluorenyl group, a phenanthrenyl group, an anthracenyl group, a pyrenyl group, a chrysenyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, a quinolinyl group, an isoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a carbazolyl group, a triazinyl group, a dibenzofuranyl group, or a dibenzothiophenyl group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amino group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C20alkyl group, a C1-C20alkoxy group, a phenyl group, a biphenyl group, a terphenyl group, a naphthyl group, a fluorenyl group, a spiro-bifluorenyl group, a benzofluorenyl group, a dibenzofluorenyl group, a phenanthrenyl group, an anthracenyl group, a pyrenyl group, a chrysenyl group, a pyridinyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, a quinolinyl group, an isoquinolinyl group, a quinoxalinyl group, a quinazolinyl group, a carbazolyl group, a triazinyl group, a dibenzofuranyl group, a dibenzothiophenyl group, or any combination thereof,xd11 and xd12 may each independently be an integer from 0 to 5,two of R501to R504may optionally be linked together to form a saturated or unsaturated ring, andtwo of R505to R505may optionally be linked together to form a saturated or unsaturated ring. The fluorescent dopant may include at least one compound of the following compounds FD(1) to FD(16) and FD1 to FD13: A thickness of the emission layer may be in a range of about 100 Å to about 1,000 Å, for example, about 200 Å to about 600 Å. When the thickness of the emission layer is within this range, excellent light-emission characteristics may be obtained without a substantial increase in driving voltage. Then, an electron transport region may be located on the emission layer. The electron transport region may include a hole blocking layer, an electron transport layer, an electron injection layer, or any combination thereof. For example, the electron transport region may have a hole blocking layer/electron transport layer/electron injection layer structure or an electron transport layer/electron injection layer structure, but the structure of the electron transport region is not limited thereto. The electron transport layer may have a single-layered structure or a multi-layered structure including two or more different materials. Conditions for forming the hole blocking layer, the electron transport layer, and the electron injection layer which constitute the electron transport region may be understood by referring to the conditions for forming the hole injection layer. When the electron transport region includes a hole blocking layer, the hole blocking layer may include, for example, at least one of BCP, Bphen, BAlq, or any combination thereof, but embodiments of the present disclosure are not limited thereto. A thickness of the hole blocking layer may be in a range of about 20 Å to about 1,000 Å, for example, about 30 Å to about 300 Å. When the thickness of the hole blocking layer is within these ranges, the hole blocking layer may have excellent hole blocking characteristics without a substantial increase in driving voltage. The electron transport layer may include at least one BCP, Bphen, Alq3, BAlq, TAZ, NTAZ, or any combination thereof. In one or more embodiments, the electron transport layer may include at least one of ET1 to ET25, but are not limited thereto: A thickness of the electron transport layer may be in a range of about 100 Å to about 1,000 Å, for example, about 150 Å to about 500 Å. When the thickness of the electron transport layer is within the range described above, the electron transport layer may have satisfactory electron transport characteristics without a substantial increase in driving voltage. Also, the electron transport layer may further include, in addition to the materials described above, a metal-containing material. The metal-containing material may include a L1complex. The L1complex may include, for example, Compound ET-D1 (lithium 8-hydroxyquinolate, LiQ) or ET-D2. The electron transport region may include an electron injection layer (EIL) that promotes flow of electrons from the second electrode19thereinto. The electron injection layer may include at least one LiF, NaCl, CsF, Li2O, BaO, or any combination thereof. A thickness of the electron injection layer may be in a range of about 1 Å to about 100 Å, and, for example, about 3 Å to about 90 Å. When the thickness of the electron injection layer is within the range described above, the electron injection layer may have satisfactory electron injection characteristics without a substantial increase in driving voltage. The second electrode19is located on the organic layer15. The second electrode19may be a cathode. A material for forming the second electrode19may be metal, an alloy, an electrically conductive compound, or a combination thereof, which have a relatively low work function. For example, lithium (Li), magnesium (Mg), aluminum (Al), aluminum-lithium (Al—Li), calcium (Ca), magnesium-indium (Mg—In), or magnesium-silver (Mg—Ag) may be formed as the material for forming the second electrode19. To manufacture a top-emission type light-emitting device, a transmissive electrode formed using ITO or IZO may be used as the second electrode19. Hereinbefore, the organic light-emitting device has been described with reference to FIGURE, but embodiments of the present disclosure are not limited thereto. Another aspect provides a diagnostic composition including at least one organometallic compound represented by Formula 1. The organometallic compound represented by Formula 1 provides high luminescent efficiency. Accordingly, a diagnostic composition including the organometallic compound may have high diagnostic efficiency. The diagnostic composition may be used in various applications including a diagnosis kit, a diagnosis reagent, a biosensor, and a biomarker. The term “C1-C60alkyl group” as used herein refers to a linear or branched saturated aliphatic hydrocarbon monovalent group having 1 to 60 carbon atoms, and non-limiting examples thereof include a methyl group, an ethyl group, a propyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, a pentyl group, an isoamyl group, and a hexyl group. The term “C1-C60alkylene group” used herein refers to a divalent group having the same structure as that of the C1-C60alkyl group. The term “C1-C60alkoxy group” used herein refers to a monovalent group represented by —OA101(wherein A101is the C1-C60alkyl group), and examples thereof include a methoxy group, an ethoxy group, and an isopropyloxy group. The term “C1-C60alkylthio group” used herein refers to a monovalent group represented by —SA102(wherein A102is the C1-C60alkyl group), and examples thereof include a methylthio group, a ethylthio group, and an isopropylthio group. The term “C2-C60alkenyl group” as used herein refers to a hydrocarbon group formed by substituting at least one carbon-carbon double bond in the middle or at the terminus of the C2-C60alkyl group, and examples thereof include an ethenyl group, a propenyl group, and a butenyl group. The term “C2-C60alkenylene group” used herein refers to a divalent group having the same structure as that of the C2-C60alkenyl group. The term “C2-C60alkynyl group” as used herein refers to a hydrocarbon group formed by substituting at least one carbon-carbon triple bond in the middle or at the terminus of the C2-C60alkyl group, and examples thereof include an ethynyl group, and a propynyl group. The term “C2-C60alkynylene group” as used herein refers to a divalent group having the same structure as that of the C2-C60alkynyl group. The term “C3-C10cycloalkyl group” as used herein refers to a monovalent saturated hydrocarbon monocyclic group having 3 to 10 carbon atoms, and examples thereof include a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, a cyclohexyl group, and a cycloheptyl group. The term “C3-C10cycloalkylene group” as used herein refers to a divalent group having the same structure as that of the C3-C10cycloalkyl group. The term “C1-C10heterocycloalkyl group” as used herein refers to a monovalent saturated monocyclic group having at least one N, O, P, Si, B, Se, Ge, Te, S, or any combination thereof as a ring-forming atom and 1 to 10 carbon atoms, and non-limiting examples thereof include a tetrahydrofuranyl group, and a tetrahydrothiophenyl group. The term “C1-C10heterocycloalkylene group” as used herein refers to a divalent group having the same structure as the C1-C10heterocycloalkyl group. The term “C3-C10cycloalkenyl group” as used herein refers to a monovalent monocyclic group that has 3 to 10 carbon atoms and at least one carbon-carbon double bond in the ring thereof and no aromaticity, and non-limiting examples thereof include a cyclopentenyl group, a cyclohexenyl group, and a cycloheptenyl group. The term “C3-C10cycloalkenylene group” as used herein refers to a divalent group having the same structure as the C3-C10cycloalkenyl group. The term “C2-C10heterocycloalkenyl group” as used herein refers to a monovalent monocyclic group that has at least one N, O, P, Si, B, Se, Ge, Te, S, or any combination thereof as a ring-forming atom, 2 to 10 carbon atoms, and at least one carbon-carbon double bond in its ring. Examples of the C2-C10heterocycloalkenyl group are a 2,3-dihydrofuranyl group and a 2,3-dihydrothiophenyl group. The term “C2-C10heterocycloalkenylene group” as used herein refers to a divalent group having the same structure as the C2-C10heterocycloalkenyl group. The term “C6-C60aryl group” as used herein refers to a monovalent group having a carbocyclic aromatic system having 6 to 60 carbon atoms, and the term “C6-C60arylene group” as used herein refers to a divalent group having a carbocyclic aromatic system having 6 to 60 carbon atoms. Non-limiting examples of the C6-C60aryl group include a phenyl group, a naphthyl group, an anthracenyl group, a phenanthrenyl group, a pyrenyl group, and a chrysenyl group. When the C6-C60aryl group and the06-C60arylene group each include two or more rings, the rings may be fused to each other. The C7-C60alkylaryl group refers to a C6-C60aryl group substituted with at least one C1-C60alkyl group. The term “C1-C60heteroaryl group” as used herein refers to a monovalent group having a cyclic aromatic system that has at least one N, O, P, Si, B, Se, Ge, Te, S, or any combination thereof as a ring-forming atom, and 1 to 60 carbon atoms. The term “C1-C60heteroarylene group” as used herein refers to a divalent group having a cyclic aromatic system that has at least one N, O, P, B, Se, Ge, Te, S, or any combination thereof as a ring-forming atom, and 1 to 60 carbon atoms. Examples of the C1-C60heteroaryl group include a pyridinyl group, a pyrimidinyl group, a pyrazinyl group, a pyridazinyl group, a triazinyl group, a quinolinyl group, and an isoquinolinyl group. When the C1-C60heteroaryl group and the C1-C60heteroarylene group each include two or more rings, the rings may be fused to each other. The C2-C60alkylheteroaryl group refers to a C1-C60heteroaryl group substituted with at least one C1-C60alkyl group. The term “C6-C60aryloxy group” as used herein indicates —OA102(wherein A102is the C6-C60aryl group), and the term “C6-C60arylthio group” as used herein indicates —SA103(wherein A103is the C6-C60aryl group). The term “monovalent non-aromatic condensed polycyclic group” as used herein refers to a monovalent group (for example, having 8 to 60 carbon atoms) having two or more rings condensed to each other, only carbon atoms as ring-forming atoms, and no aromaticity in its entire molecular structure. Examples of the monovalent non-aromatic condensed polycyclic group include a fluorenyl group. The term “divalent non-aromatic condensed polycyclic group” as used herein refers to a divalent group having the same structure as the monovalent non-aromatic condensed polycyclic group. The term “monovalent non-aromatic condensed heteropolycyclic group” as used herein refers to a monovalent group (for example, having 2 to 60 carbon atoms) having two or more rings condensed to each other, a heteroatom of N, O, P, Si, B, Se, Ge, Te, S, or any combination thereof other than carbon atoms, as a ring-forming atom, and no aromaticity in its entire molecular structure. Non-limiting examples of the monovalent non-aromatic condensed heteropolycyclic group include a carbazolyl group. The term “divalent non-aromatic condensed heteropolycyclic group” as used herein refers to a divalent group having the same structure as the monovalent non-aromatic condensed heteropolycyclic group. The term “C5-C30carbocyclic group” as used herein refers to a saturated or unsaturated cyclic group having, as a ring-forming atom, 5 to 30 carbon atoms only. The C5-C30carbocyclic group may be a monocyclic group or a polycyclic group. The term “C1-C30heterocyclic group” as used herein refers to a saturated or unsaturated cyclic group having, as a ring-forming atom, at least one N, O, Si, P, B, Se, Ge, Te, S, or any combination thereof other than 1 to 30 carbon atoms. The C1-C30heterocyclic group may be a monocyclic group or a polycyclic group. At least one substituent of the substituted C5-C30carbocyclic group, the substituted C1-C30heterocyclic group, the substituted C1-C60alkyl group, the substituted C2-C60alkenyl group, the substituted C2-C60alkynyl group, the substituted C1-C60alkoxy group, the substituted C1-C60alkylthio group, the substituted C3-C10cycloalkyl group, the substituted C1-C10heterocycloalkyl group, the substituted C3-C10cycloalkenyl group, the substituted C2-C10heterocycloalkenyl group, the substituted C6-C60aryl group, the substituted C7-C60alkylaryl group, the substituted C6-C60aryloxy group, the substituted C6-C60arylthio group, the substituted C1-C60heteroaryl group, the substituted C2-C60alkyl heteroaryl group, the substituted monovalent non-aromatic condensed polycyclic group, and the substituted monovalent non-aromatic condensed heteropolycyclic group may be:deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, or any combination thereof;a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, or a C1-C60alkoxy group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C7-C60alkylaryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a C2-C60alkyl heteroaryl group, a monovalent non-aromatic condensed polycyclic group, a monovalent non-aromatic condensed heteropolycyclic group, —N(Q11)(Q12), —Si(Q13)(Q14)(Q15), —B(Q16)(Q17), —P(═O)(Q18)(Q19), or any combination thereof;a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C7-C60alkylaryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a C2-C60alkyl heteroaryl group, a monovalent non-aromatic condensed polycyclic group, or a monovalent non-aromatic condensed heteropolycyclic group;a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C7-C60alkylaryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a C2-C60alkyl heteroaryl group, a monovalent non-aromatic condensed polycyclic group, or a monovalent non-aromatic condensed heteropolycyclic group, each substituted with at least one deuterium, —F, —Cl, —Br, —I, —CD3, —CD2H, —CDH2, —CF3, —CF2H, —CFH2, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C7-C60alkylaryl group, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a C2-C60alkyl heteroaryl group, a monovalent non-aromatic condensed polycyclic group, a monovalent non-aromatic condensed heteropolycyclic group, —N(Q21)(Q22), —Si(Q23)(Q24)(Q25), —B(Q26)(Q27), —P(═O)(Q28)(Q29), or any combination thereof; or—N(Q31)(Q32), —Si(Q33)(Q34)(Q35), —B(Q36)(Q37), or —P(═O)(Q38)(Q39),wherein Q1to Q9, Q11to Q19, Q21to Q29, and Q31to Q39may each independently be hydrogen, deuterium, —F, —Cl, —Br, —I, a hydroxyl group, a cyano group, a nitro group, an amidino group, a hydrazine group, a hydrazone group, a carboxylic acid group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a C1-C60alkyl group, a C1-C60alkyl group substituted with at least one deuterium, a C1-C60alkyl group, a C6-C60aryl group, or a combination thereof, a C2-C60alkenyl group, a C2-C60alkynyl group, a C1-C60alkoxy group, a C3-C10cycloalkyl group, a C1-C10heterocycloalkyl group, a C3-C10cycloalkenyl group, a C2-C10heterocycloalkenyl group, a C6-C60aryl group, a C6-C60aryl group substituted with at least one deuterium, a C1-C60alkyl group, a C6-C60aryl group, or any combination thereof, a C6-C60aryloxy group, a C6-C60arylthio group, a C1-C60heteroaryl group, a C2-C60alkyl heteroaryl group, a monovalent non-aromatic condensed polycyclic group, a monovalent non-aromatic condensed heteropolycyclic group, or any combination thereof. Hereinafter, a compound and an organic light-emitting device according to embodiments are described in detail with reference to Synthesis Example and Examples. However, the organic light-emitting device is not limited thereto. The wording “B was used instead of A” used in describing Synthesis Examples means that an amount of A used was identical to an amount of B used, in terms of a molar equivalent. EXAMPLES Synthesis Example 1: Synthesis of Compound 1 (1) Synthesis of Intermediate 1(1) 127.0 mmol (15 g) 1H-benzo[d]imidazole, 152.4 mmol (45 g) 1,3-dibromo-5-(tert-butyl)benzene, 31.7 mmol (6.1 g) CuI, 38.1 mmol (6.9 g) 1,10-phenanthroline, and 253.9 mmol (83 g) Cs2CO3were added to 250 mL of dimethylformamide (DMF) and refluxed at a temperature of 130° C. for 12 hours. The reaction mixture obtained therefrom was cooled and diluted using a mixture including ethyl acetate and water. The organic layer was separated, washed three times with water and dried using magnesium sulfate, and then, a solvent was removed therefrom under reduced pressure, thereby obtaining a crude product. The crude product was subjected to silica gel column chromatography (eluent: ethyl acetate:hexane) to obtain Intermediate 1(1) (yield of 52%). MALDI-TOF (m/z): 328.04 [M]+ (2) Synthesis of Intermediate 1(2) 101.4 mmol (20.0 g) 2-methoxy-9H-carbazole and 152.1 mmol (32.6 g) 2-bromo-4-(tert-butyl)pyridine were dissolved in 340 ml of dioxane, and then, 50.7 mmol (9.7 g) CuI, 152.1 mmol (32.3 g) K3PO4, and 72.4 mmol (12.2 ml) trans-1,2-cyclohexanediamine were added thereto, followed by refluxing at a temperature of 120° C. for 12 hours. After completion of the reaction, the mixture was cooled to room temperature, and diluted with mixture including ethyl acetate and water. The organic layer was separated, washed three times with water and dried using magnesium sulfate, and then, a solvent was removed therefrom under reduced pressure, thereby obtaining a crude product. The crude product was subjected to silica gel column chromatography (eluent: ethyl acetate:hexane) to obtain Intermediate 1(2) (yield of 85%). MALDI-TOF (m/z): 331.16 [M]+ (3) Synthesis of Intermediate 1(3) 95.3 mmol (31.5 g) Intermediate 1(2) and 1.4 mol (165.3 g) pyridine hydrochloride were added and refluxed in a neat condition at a temperature of 180° C. for 20 hours. After completion of the reaction, the mixture was cooled to room temperature, and diluted using a mixture including dichloromethane and water. The organic layer was separated, washed three times with water and dried using magnesium sulfate, and then, a solvent was removed therefrom under reduced pressure, thereby obtaining a crude product. The crude product was subjected to silica gel column chromatography (eluent: ethyl acetate: dichloromethane:hexane) to obtain Intermediate 1(3) (yield of 65%). MALDI-TOF (m/z): 317.15 [M]+ (4) Synthesis of Intermediate 1(4) 15.2 mmol (5 g) Intermediate 1(1) and 12.7 mmol (4.0 g) Intermediate 1(3) were dissolved in 250 ml of dimethyl sulfoxide (DMSO), and then, 3.8 mmol (0.7 g) CuI and 50.8 mmol (10.8 g) K3PO4, and 19.1 mmol (2.3 g) picolinic acid were added thereto, and refluxed at a temperature of 100° C. for 12 hours. After completion of the reaction, the mixture was cooled to room temperature, and diluted using a mixture including ethyl acetate and water. The organic layer was washed three times with water and dried using magnesium sulfate, and then, a solvent was removed therefrom under reduced pressure, thereby obtaining a crude product. The crude product was subjected to silica gel column chromatography (eluent: ethyl acetate:hexane) to obtain Intermediate 1(4) (yield of 73%). MALDI-TOF (m/z): 564.29 [M]+ (5) Synthesis of Intermediates 1(5) and 1(6) 19.2 mmol (5.0 g) 1-(tert-butyl)-3-iodobenzene, 28.8 mmol (3.5 g) mesitylene compound, 24.0 mmol (4.1 g) 3-chloroperoxybenzoic acid (mCBPA) were dissolved in 50 ml of dichloromethane, and then, cooled in an ice bath at a temperature of 0° C. 48.0 mmol (4.2 ml) triflic acid was added dropwise thereto. After the temperature was raised to room temperature, the resultant mixture was stirred for 2 hours and then a solvent was completely removed therefrom. A small amount of diethyl ether was added thereto, followed by stirring and filtration. The obtained crude product, that is, Intermediate 1(5) was used for the next reaction. 7.6 mmol (4.0 g) Intermediate 1(5), 5.0 mmol (2.9 g) Intermediate 1(4), and 0.3 mmol (0.09 g) copper acetate (Cu(OAc)2) were added to 25 mL of dimethylformamide (DMF), and then, refluxed at a temperature of 130° C. for 12 hours. The crude product obtained by removing the solvent therefrom under reduced pressure was subjected to silica gel column chromatography (eluent: dichloromethane:acetone) to obtain Intermediate 1(6) (yield of 80%). MALDI-TOF (m/z): 697.38 [M]+ (6) Synthesis of Compound 1 3.5 mmol (1.3 g) Pt(COD)Cl2, 3.5 mmol (3.0 g) Intermediate 1(6), and 10.5 mmol (0.9 g) sodium acetate (NaOAc) were added to 180 mL of benzonitrile, and then, refluxed at a temperature of 180° C. for 12 hours. After completion of the reaction, the resultant mixture was cooled to room temperature and the solvent was removed therefrom under reduced pressure to obtain a crude product, which was then subjected to gel column chromatography (eluent: dichloromethane and hexane) to obtain compound 1 (yield of 58%). MALDI-TOF (m/z): 889.32 [M]+ Synthesis Example 2: Synthesis of Compound 2 (1) Synthesis of Intermediate 2(1) Intermediate 2(1) (yield: 60%) was synthesized in the same manner as used to synthesize Intermediate 1(1) of Synthesis Example 1, except that 1,3-dibromobenzene was used instead of 1,3-dibromo-5-(tert-butyl)benzene. MALDI-TOF (m/z): 272.99 [M]+ (2) Synthesis of Intermediate 2(2) Intermediate 2(2) (yield: 69%) was obtained in the same manner as used to synthesize Intermediate 1(4) of Synthesis Example 1, except that 1-(3-bromophenyl)-1H-benzo[d]imidazole was used instead of 1-(3-bromo-5-(tert-butyl)phenyl)-1H-benzo[d]imidazole. MALDI-TOF (m/z): 509.22 [M]+ (3) Synthesis of Intermediates 2(3) and 2(4) Intermediates 2(3) and 2(4) (yield: 75%) were synthesized in the same manner as used to synthesize Intermediates 1(5) and 1(6) of Synthesis Example 1, except that 3-(tert-butyl)-5-iodo-1,1′-biphenyl was used instead of 1-(tert-butyl)-3-iodobenzene, and the Intermediate 2(2) was used instead of the Intermediate 1(4). MALDI-TOF (m/z): 717.37 [M]+ (4) Synthesis of Compound 2 Compound 2 (yield: 52%) was synthesized in the same manner as used to synthesize compound 1 of Synthesis Example 1, except that Intermediate 2(4) was used instead of Intermediate 1(6). MALDI-TOF (m/z): 910.33 [M]+ Example 1 An ITO glass substrate was cut to a size of 50 mm×50 mm×0.5 mm and then, sonicated in acetone, isopropyl alcohol and pure water, each for 15 minutes, and then, washed by exposure to UV ozone for 30 minutes. Then, m-MTDATA was deposited on an ITO electrode (anode) of the glass substrate at a deposition rate of 1 Å/sec to form a hole injection layer having a thickness of 600 Å, and then, α-NPD was deposited on the hole injection layer at a deposition rate of 1 Å/sec to form a hole transport layer having a thickness of 250 Å. Compound 1 (dopant) and CBP (host) were co-deposited on the hole transport layer at a deposition rate of 0.1 Å/sec and a deposition rate of 1 Å/sec, respectively, to form an emission layer having a thickness of 400 Å. BAlq was deposited on the emission layer at a deposition rate of 1 Å/sec to form a hole blocking layer having a thickness of 50 Å, and Alq3was deposited on the hole blocking layer to form an electron transport layer having a thickness of 300 Å, and then, LiF was deposited on the electron transport layer to form an electron injection layer having a thickness of 10 Å, and then, Al was vacuum deposited on the electron injection layer to form a second electrode (cathode) having a thickness of 1,200 Å, thereby completing manufacturing of an organic light-emitting device having a structure of ITO m-MTDATA (600 Å)/α-NPD (250 Å)/CBP+Compound 1 (10%) (400 Å)/BAlq (50 Å)/Alq3(300 Å)/LiF (10 Å)/Al(1,200 Å). Example 2, and Comparative Examples 1 to 6 Organic light-emitting devices were manufactured in the same manner as in Example 1, except that in forming an emission layer, for use as a dopant, corresponding compounds shown in Table 2 were used instead of Compound 1. Evaluation Example 1: Characterization of Organic Light-Emitting Devices For each of the organic light-emitting devices manufactured according to Examples 1 and 2 and Comparative Examples 1 to 6, luminescence quantum efficiency (PLQY), external quantum efficiency (EQE), photochemical stability (PCS) and lifespan (T95) were evaluated as relative values. Results thereof are shown in Table 2. This evaluation was performed using a current-voltage meter (Keithley 2400) and a luminescence meter (Minolta Cs-1000A), and the lifespan (T95) was evaluated as a relative value (%) by measuring the amount of time that elapsed until luminance was reduced to 95% of the initial brightness of 100%. PCS values were calculated by the following Equation P. PCS (%)=I2/I1×10  <Equation P> In Equation P, With respect to a film formed by depositing a compound of which PCS was to be measured, a PL spectrum was evaluated at room temperature by using He—Cd laser (excitation wavelength=325 nm, and laser power density=100 mW/cm2) of KIMMON-KOHA Co., Ltd. in an Ar atmosphere in which external air was blocked immediately after the film was formed. The maximum light intensity of the PL spectrum was indicated as With respect to a film formed by depositing a compound of which PCS was to be measured, the PL spectrum thereof was evaluated at room temperature by using a He—Cd laser (excitation wavelength=325 nm, and laser power density=100 mW/cm2) of KIMMON-KOHA Co., Ltd., after 3 hours of exposure to light of a He—Cd laser (excitation wavelength=325 nm, and laser power density=100 mW/cm2) of KIMMON-KOHA Co., Ltd., which was a pumping laser used when 1, was evaluated, in an Ar atmosphere in which external air was blocked. The maximum light intensity of the PL spectrum herein was indicated as I2. TABLE 2LifespanMaximumDopantPLQYEQE(T95)luminescenceNo.compound(relative value)(relative value)PCS (%)(relative value)wavelength (nm)Example 1Compound 1878684120465Example 2Compound 265778053461ComparativeCompound A49767613459Example 1ComparativeCompound B71898144463Example 2ComparativeCompound C63738575466Example 3ComparativeCompound D101887754465Example 4ComparativeCompound E10010081100465Example 5Comparativecompound F74958041461Example 6 From Table 2, it can be seen that the organic light-emitting devices of Examples 1 and 2 have excellent light emission quantum efficiency and external quantum efficiency, and has a higher photochemical stability and a longer lifespan than the organic light-emitting device of Comparative Examples 1 to 6. Example 3 An organic light-emitting device was manufactured in the same method as in Example 1, except that compound CBP was used as a host in a weight ratio of 88.5%, and compound 1 and compound FD were used as a dopant in a weight ratio of 10%: 1.5%. Comparative Example 7 An organic light-emitting device was manufactured in the same manner as in Example 3, except that compound FD was used as a dopant when forming the emission layer. Evaluation Example 2: Characterization of Organic Light-Emitting Devices For each organic light-emitting device manufactured according to Example 3 and Comparative Example 7, the driving voltage, external quantum efficiency (EQE), maximum emission wavelength, and lifespan (T95) were evaluated. Results thereof are shown in Table 3. This evaluation was performed using a current-voltage meter (Keithley 2400) and a luminescence meter (Minolta Cs-1000A), and the lifespan (T95) (at 1200 nit) was evaluated by measuring the amount of time that elapsed until luminance was reduced to 95% of the initial brightness of 100%. TABLE 3LifespanMaximumDopantDrivingEQE(T95)luminescenceNo.compoundvoltage (V)(relative value)(relative value)wavelength (nm)Example 3Compound 14.70194928465+Compound FDComparativeCompound FD5.81100100461Example 7 From Table 3, it can be seen that the organic light-emitting device of Example 3 has a low driving voltage and significantly improved external quantum efficiency and lifespan characteristics compared to the organic light-emitting device of Comparative Example 7. The organometallic compound has excellent photochemical stability, and organic light-emitting devices using the organometallic compound have improved efficiency and lifespan characteristics. Such organometallic compounds have excellent phosphorescent luminescent characteristics, and thus, when these compounds are used, a diagnostic composition having a high diagnostic efficiency may be provided. It should be understood that embodiments described herein should be considered in a descriptive sense only and not for purposes of limitation. Descriptions of features or aspects within each embodiment should typically be considered as available for other similar features or aspects in other embodiments. While one or more embodiments have been described with reference to the figures, it will be understood by those of ordinary skill in the art that various changes in form and details may be made therein without departing from the spirit and scope as defined by the following claims.
140,012
11858950
DETAILED DESCRIPTION Exemplary embodiments will hereinafter be described in detail, and may be easily performed by a person skilled in the related art. However, this disclosure may be embodied in many different forms and is not to be construed as limited to the exemplary embodiments set forth herein. In the drawings, the thickness of layers, films, panels, regions, etc., are exaggerated for clarity. It will be understood that when an element such as a layer, film, region, or substrate is referred to as being “on” another element, it may be directly on the other element or intervening elements may also be present. In contrast, when an element is referred to as being “directly on” another element, there are no intervening elements present. It will be understood that, although the terms “first,” “second,” “third” etc. may be used herein to describe various elements, components, regions, layers and/or sections, these elements, components, regions, layers and/or sections should not be limited by these terms These terms are only used to distinguish one element, component, region, layer or section from another element, component, region, layer or section Thus, “a first element,” “component,” “region,” “layer” or “section” discussed below could be termed a second element, component, region, layer or section without departing from the teachings herein. The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. As used herein, “a,” “an,” “the,” and “at least one” do not denote a limitation of quantity, and are intended to cover both the singular and plural, unless the context clearly indicates otherwise. For example, “an element” has the same meaning as “at least one element,” unless the context clearly indicates otherwise. “Or” means “and/or.” As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items It will be further understood that the terms “comprises” and/or “comprising,” or “includes” and/or “including” when used in this specification, specify the presence of stated features, regions, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, regions, integers, steps, operations, elements, components, and/or groups thereof. Furthermore, relative terms, such as “lower” or “bottom” and “upper” or “top,” may be used herein to describe one element's relationship to another element as illustrated in the Figures It will be understood that relative terms are intended to encompass different orientations of the device in addition to the orientation depicted in the Figures For example, if the device in one of the figures is turned over, elements described as being on the “lower” side of other elements would then be oriented on “upper” sides of the other elements The exemplary term “lower,” can therefore, encompasses both an orientation of “lower” and “upper,” depending on the particular orientation of the figure Similarly, if the device in one of the figures is turned over, elements described as “below” or “beneath” other elements would then be oriented “above” the other elements The exemplary terms “below” or “beneath” can, therefore, encompass both an orientation of above and below. “About” or “approximately” as used herein is inclusive of the stated value and means within an acceptable range of deviation for the particular value as determined by one of ordinary skill in the art, considering the measurement in question and the error associated with measurement of the particular quantity (i.e., the limitations of the measurement system). For example, “about” can mean within one or more standard deviations, or within ±30%, 20%, 10% or 5% of the stated value. Unless otherwise defined, all terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs It will be further understood that terms, such as those defined in commonly used dictionaries, should be interpreted as having a meaning that is consistent with their meaning in the context of the relevant art and the present disclosure, and will not be interpreted in an idealized or overly formal sense unless expressly so defined herein. Exemplary embodiments are described herein with reference to cross section illustrations that are schematic illustrations of idealized embodiments As such, variations from the shapes of the illustrations as a result, for example, of manufacturing techniques and/or tolerances, are to be expected Thus, embodiments described herein should not be construed as limited to the particular shapes of regions as illustrated herein but are to include deviations in shapes that result, for example, from manufacturing. For example, a region illustrated or described as flat may, typically, have rough and/or nonlinear features Moreover, sharp angles that are illustrated may be rounded Thus, the regions illustrated in the figures are schematic in nature and their shapes are not intended to illustrate the precise shape of a region and are not intended to limit the scope of the present claims. Hereinafter, as used herein, when a definition is not otherwise provided, “substituted” refers to replacement of hydrogen of a compound or a group by a substituent of a halogen atom, a hydroxy group, a nitro group, a cyano group, an amino group, an azido group, an amidino group, a hydrazino group, a hydrazono group, a carbonyl group, a carbamyl group, a thiol group, an ester group, a carboxyl group or a salt thereof, a sulfonic acid group or a salt thereof, a phosphoric acid group or a salt thereof, a silyl group, a C1 to C20 alkyl group, a C2 to C20 alkenyl group, a C2 to C20 alkynyl group, a C6 to C30 aryl group, a C7 to C30 arylalkyl group, a C1 to C30 alkoxy group, a C1 to C20 heteroalkyl group, a C3 to C20 heterocyclic group, a C3 to C20 heteroarylalkyl group, a C3 to C30 cycloalkyl group, a C3 to C15 cycloalkenyl group, a C6 to C15 cycloalkynyl group, a C3 to C30 heterocycloalkyl group, or a combination thereof. As used herein, when specific definition is not otherwise provided, “hetero” refers to one including 1 to 4 of N, O, S, Se, Te, Si, P, or a combination thereof. As used herein, when a definition is not otherwise provided, the term “aromatic ring” refers to a cyclic functional group in which all ring-forming atoms have a p-orbital, wherein these p-orbitals are conjugated. Hereinafter, as used herein, when a definition is not otherwise provided, “aryl group” refers to a group including at least one aromatic hydrocarbon moiety. All the elements of the hydrocarbon aromatic moiety have p-orbitals which form conjugation, for example a phenyl group, a naphthyl group, and the like; two or more aromatic hydrocarbon moieties may be linked by a sigma bond, for example a biphenyl group, a terphenyl group, a quarterphenyl group, and the like; and two or more aromatic hydrocarbon moieties may be fused directly or indirectly to provide a non-aromatic fused ring, for example a fluorenyl group. The aryl group may include a monocyclic, polycyclic or fused ring polycyclic (i.e., rings sharing adjacent pairs of carbon atoms) functional group. Hereinafter, as used herein, when a definition is not otherwise provided, “heterocyclic group” refers to a group obtained by replacing carbon atoms in a ring of an aryl group, alicyclic hydrocarbon group, or a fused ring thereof with at least one of N, O, S, Se, Te, P, Si, or a combination thereof. Hereinafter, as used herein, when a definition is not otherwise provided, “ring” refers to an aromatic ring, a non-aromatic ring, a heteroaromatic ring, a hetero non-aromatic ring, a fused ring thereof, and/or a combination thereof. Hereinafter, as used herein, when a definition is not otherwise provided, the term “C1 to C30 alkyl group” as used herein refers to a linear or branched saturated aliphatic hydrocarbons monovalent group having 1 to 30 carbon atoms. Hereinafter, as used herein, when a definition is not otherwise provided, the term “C1 to C30 alkoxy group” used herein refers to a monovalent group represented by —OA101(wherein A101is the C1 to C30 alkyl group), and examples thereof are a methoxy group, an ethoxy group, a propoxy group, a butoxy group, and a pentoxy group. Hereinafter, as used herein, when a definition is not otherwise provided, the term “C2 to C30 alkenyl group” as used herein refers to a hydrocarbon group formed by substituting at least one carbon-carbon double bond in the middle or at the terminus of the C2 to C30 alkyl group, and examples thereof include an ethenyl group, a propenyl group, and a butenyl group. Hereinafter, as used herein, when a definition is not otherwise provided, the term “C2 to C30 alkynyl group” as used herein refers to a hydrocarbon group formed by substituting at least one carbon-carbon triple bond in the middle or at the terminus of the C2 to C30 alkyl group, and examples thereof include an ethynyl group, and a propynyl group. Hereinafter, as used herein, when a definition is not otherwise provided, the term “haloalkyl group” refers to an alkyl group where at least one hydrogen is replaced by F, Cl, Br, I, or a combination thereof. Specific examples of a haloalkyl group may be a fluoroalkyl group, for example a perfluoroalkyl group. Hereinafter, as used herein, when a definition is not otherwise provided, the term “haloaryl group” refers to an aryl group where at least one hydrogen is replaced by F, Cl, Br, I, or a combination thereof. Specific examples of a haloaryl group may be a fluoroaryl group, for example a perfluoroaryl group. Hereinafter, a compound according to an embodiment is described. The compound according to an embodiment may be represented by Chemical Formula 1. In Chemical Formula 1,Ar1and Ar2may independently be a substituted or unsubstituted benzene; a substituted or unsubstituted heterocycle including at least one N, O, S, Se, Te, or any combination thereof; or a fused ring having two or more of a substituted or unsubstituted benzene; a substituted or unsubstituted heterocycle, or a combination thereof,Z may be N or CRa, wherein Ramay be an electron withdrawing group,L1and L2may independently be a single bond, a substituted or unsubstituted C6 to C30 arylene group, a divalent substituted or unsubstituted C3 to C30 heterocyclic group, or a combination thereof,R1may be a substituted or unsubstituted C3 to C30 heterocyclic group or NRbRc, wherein Rband Rcmay independently be hydrogen, a substituted or unsubstituted C1 to C30 alkyl group, a substituted or unsubstituted C1 to C30 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group, a substituted or unsubstituted silyl group, a halogen, or a combination thereof, and Rband Rcmay independently be present or Rband Rcmay be linked with each other to form a ring,R2may be hydrogen, a substituted or unsubstituted C1 to C30 alkyl group, a substituted or unsubstituted C2 to C30 alkenyl group, a substituted or unsubstituted C2 to C30 alkynyl group, a substituted or unsubstituted C1 to C30 alkoxy group, a substituted or unsubstituted silyl group, a halogen, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group or NRdRe, wherein Rdand Remay independently be hydrogen, a substituted or unsubstituted C1 to C30 alkyl group, a substituted or unsubstituted C1 to C30 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group, a substituted or unsubstituted silyl group, a halogen, or a combination thereof, and Rdand Remay independently be present or Rdand Remay be linked with each other to form a ring,R3and R4may independently be a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group, or a combination thereof, andR5and R6together may be oxygen (═O), sulfur (═S), selenium (═Se), or tellurium (═Te), or R5and R6may independently be a substituted or unsubstituted C1 to C30 alkyl group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group, a halogen, a cyano group, or a combination thereof. The compound has a structure in which two pyrroles form a complex with a disubstituted boron atom. The compound may be configured to absorb light in the near infra-red wavelength spectrum and may exhibit good electrical properties, by combining a core and/or a substituent with a moiety that imparts electron donating properties. A peak absorption wavelength (λmax) of the compound may be for example greater than or equal to about 700 nm, for example greater than or equal to about 720 nm, greater than or equal to about 730 nm, greater than or equal to about 750 nm, greater than or equal to about 780 nm, greater than or equal to about 790 nm, greater than or equal to about 800 nm, greater than or equal to about 810 nm, greater than or equal to about 820 nm, greater than or equal to about 830 nm, greater than or equal to about 840 nm, greater than or equal to about 850 nm, greater than or equal to about 870 nm, greater than or equal to about 890 nm, greater than or equal to about 900 nm, or greater than or equal to about 910 nm. The peak absorption wavelength of the compound may be for example in a wavelength spectrum of about 700 nm to about 3000 nm, within the range, for example about 750 nm to about 2500 nm, about 780 nm to about 2200 nm, about 790 nm to about 2100 nm, about 800 nm to about 2000 nm, about 810 nm to about 2000 nm, about 820 nm to about 2000 nm, about 830 nm to about 2000 nm, about 850 nm to about 1900 nm, about 870 nm to about 1800 nm, about 900 nm to about 1600 nm, or about 910 nm to about 1500 nm. For example, Ar1and Ar2may independently be a substituted or unsubstituted benzene, a substituted or unsubstituted thiophene, a substituted or unsubstituted furan, a substituted or unsubstituted selenophene, a substituted or unsubstituted tellurophene, a fused ring having two or more of a substituted or unsubstituted benzene, a substituted or unsubstituted thiophene, a substituted or unsubstituted furan, a substituted or unsubstituted selenophene, a substituted or unsubstituted tellurophene, or a combination thereof, or a combination thereof. For example, Ar1and Ar2may independently be a substituted or unsubstituted benzene, a substituted or unsubstituted naphthalene, a substituted or unsubstituted thiophene, a substituted or unsubstituted benzothiophene, a substituted or unsubstituted dibenzothiophene, a substituted or unsubstituted furan, a substituted or unsubstituted benzofuran, a substituted or unsubstituted dibenzofuran, a substituted or unsubstituted selenophene, a substituted or unsubstituted benzoselenophene, a substituted or unsubstituted dibenzoselenophene, a substituted or unsubstituted tellurophene, a substituted or unsubstituted benzotellurophene, a substituted or unsubstituted dibenzotellurophene, or a combination thereof. For example, Ar1and Ar2may be the same. For example, Ar1and Ar2may be different. For example, Z may be nitrogen (N) or carbon substituted with an electron withdrawing group (Ra), wherein the electron withdrawing group may be for example a substituted or unsubstituted heterocyclic group including at least one nitrogen; a C1 to C30 haloalkyl group; a C6 to C30 haloaryl group; a halogen; a cyano group; or a combination thereof. For example, the electron withdrawing group (Ra) may be a substituted or unsubstituted pyridinyl group, a substituted or unsubstituted pyrimidinyl group, a substituted or unsubstituted triazinyl group, a substituted or unsubstituted pyrrolyl group, a substituted or unsubstituted pyrazolyl group, a substituted or unsubstituted triazolyl group, CH2F, CHF2, CF3, F, or CN. For example, L1and L2may independently be a single bond; a substituted or unsubstituted phenylene group; a substituted or unsubstituted biphenylene group; a substituted or unsubstituted naphthylene group; a substituted or unsubstituted terphenylene group; a divalent substituted or unsubstituted C3 to C30 heterocyclic group including at least one O, S, Se, Te, N, Si; or a combination thereof, a fused ring having two or more of a substituted or unsubstituted phenylene group; a substituted or unsubstituted biphenylene group; a substituted or unsubstituted naphthylene group; a substituted or unsubstituted terphenylene group; a divalent substituted or unsubstituted C3 to C30 heterocyclic group, or a combination thereof, or a combination thereof. For example, L1and L2may independently be a single bond, a substituted or unsubstituted p-phenylene group, a substituted or unsubstituted m-phenylene group, a substituted or unsubstituted o-phenylene group, a substituted or unsubstituted biphenylene group, a substituted or unsubstituted naphthylene group, a substituted or unsubstituted terphenylene group, a substituted or unsubstituted thiophenylene group, a substituted or unsubstituted benzothiophenylene group, a substituted or unsubstituted dibenzothiophenylene group, a substituted or unsubstituted furanylene group, a substituted or unsubstituted benzofuranylene group, a substituted or unsubstituted dibenzofuranylene group, a substituted or unsubstituted selenophenylene group, a substituted or unsubstituted benzoselenophenylene group, a substituted or unsubstituted dibenzoselenophenylene group, a substituted or unsubstituted tellurophenylene group, a substituted or unsubstituted benzotellurophenylene group, a substituted or unsubstituted dibenzotellurophenylene group, a substituted or unsubstituted pyrrolylene group, a substituted or unsubstituted benzopyrrolylene group, a substituted or unsubstituted Dibenzopyrrolylene group, or a combination thereof. For example, L1and L2may be the same. For example, L1and L2may be the different. For example, R1and R2may be the same or different substituted or unsubstituted amine group and may be, for example independently represented by one of Chemical Formulae A-1 to A-4. In Chemical Formulae A-1 to A-4,W1is a single bond, O, S, Se, Te, CRfRg, or SiRhRi,W2is O, S, Se, Te, CRjRk, or SiRlRm,R18to R25and Rfto Rmare independently hydrogen, a substituted or unsubstituted C1 to C30 alkyl group, a substituted or unsubstituted C2 to C30 alkenyl group, a substituted or unsubstituted C2 to C30 alkynyl group, a substituted or unsubstituted C1 to C30 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group, a substituted or unsubstituted amine group, a substituted or unsubstituted silyl group, a halogen, or a combination thereof,R18and R19may independently be present or R18and R19may be linked with each other to form a ring,R20and R21may independently be present or R20and R21may be linked with each other to form a ring,R22and R23may independently be present or R22and R23may be linked with each other to form a ring,R24and R25may independently be present or R24and R25may be linked with each other to form a ring,Rfand Rgmay independently be present or Rfand Rgmay be linked with each other to form a ring,Rhand Rimay independently be present or Rhand Rimay be linked with each other to form a ring,Rjand Rkmay independently be present or Rjand Rkmay be linked with each other to form a ring,Rland Rmmay independently be present or Rland Rmmay be linked with each other to form a ring, and* is a bond with Chemical Formula 1. For example, in Formula 1, R1and R2may be the same or different substituted or unsubstituted heterocyclic group and may be, for example, independently represented by one of Chemical Formulae B-1 to B-6. In Chemical Formulae B-1 to B-6,X1to X35may independently be N, O, S, Se, Te, C, or CRn, or a combination thereof, andR26to R33and Rnmay independently be hydrogen, a substituted or unsubstituted C1 to C30 alkyl group, a substituted or unsubstituted C2 to C30 alkenyl group, a substituted or unsubstituted C2 to C30 alkynyl group, a substituted or unsubstituted C1 to C30 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group, a substituted or unsubstituted amine group, a substituted or unsubstituted silyl group, a halogen, a bond with Chemical Formula 1, or a combination thereof, wherein one of R26to R33and Rnis a bond with L1and one of R26to R33and Rnis a bond with L2of Chemical Formula 1. For example, X1and X2may independently be O, S, Se, or Te. For example, X1and X2may independently be O or S. For example, X3, X6, X11, X18, X23, X26, and X33may independently be O, S, Se, Te, C, or CRn. For example, X3, X6, X11, X18, X23, X26, and X33may independently be O, S, or CRn. For example, X4, X5, X7to X10, X12to X17, X19to X22, X24, X25, X27to X32, X34, and X35may independently be N or CRn. For example, R1and R2of Formula 1 may be a group derived from one of compounds in Group 1, but are not limited thereto: wherein R1of Group 1 is attached to Chemical Formula 1 via a single bond between a carbon atom of a compound of Group 1 and L1and R2of Group 1 is attached to Chemical Formula 1 via a single bond between a carbon atom of a compound of Group 1 and L2. For example, in Formula 1, R3and R4may independently be a substituted or unsubstituted C6 to C30 aryl group; a substituted or unsubstituted heterocycle group including at least one N, O, S, Se, Te, or a combination thereof; or a combination thereof. For example, R3and R4may independently be a substituted or unsubstituted phenyl group, a substituted or unsubstituted pyrrolyl group, a substituted or unsubstituted thiophenyl group, a substituted or unsubstituted furanyl group, a substituted or unsubstituted selenophenyl group, a substituted or unsubstituted tellurophenyl group, a fused ring having two or more of a substituted or unsubstituted phenyl group, a substituted or unsubstituted pyrrolyl group, a substituted or unsubstituted thiophenyl group, a substituted or unsubstituted furanyl group, a substituted or unsubstituted selenophenyl group, a substituted or unsubstituted tellurophenyl group, or a combination thereof, or a combination thereof, and for example R3and R4may independently be a substituted or unsubstituted phenyl group, a substituted or unsubstituted naphthyl group, a substituted or unsubstituted thiophenyl group, a substituted or unsubstituted benzothiophenyl group, a substituted or unsubstituted dibenzothiophenyl group, a substituted or unsubstituted furanyl group, a substituted or unsubstituted benzofuranyl group, a substituted or unsubstituted dibenzofuranyl group, a substituted or unsubstituted selenophenyl group, a substituted or unsubstituted benzoselenophenyl group, a substituted or unsubstituted dibenzoselenophenyl group, a substituted or unsubstituted tellurophenyl group, a substituted or unsubstituted benzotellurophenyl group, a substituted or unsubstituted dibenzotellurophenyl group, a substituted or unsubstituted pyrrolyl group, a substituted or unsubstituted benzopyrrolyl group, a substituted or unsubstituted dibenzopyrrolyl group, or a combination thereof. Herein “substituted” may refer to replacement of at least one hydrogen with for example a C1 to C20 alkyl group, a C1 to C20 haloalkyl group, a C1 to C20 alkoxy group, a C6 to C30 aryl group, or a C3 to C30 heterocyclic group, or a combination thereof, but is not limited thereto. For example, R3and R4may be the same. For example, R3and R4may be the different. For example, R5and R6may independently be a halogen or a C1 to C20 haloalkyl group and R5and R6may be for example fluorine. For example, R5and R6may be the same. For example, R5and R6may be the different. For example, Z may be N. The compound of Formula 1 may be represented by one of Chemical Formulae 1a-1 to 1l-1. In Chemical Formulae 1a-1 to 1l-1,Rbto Remay independently be hydrogen, a substituted or unsubstituted C1 to C30 alkyl group, a substituted or unsubstituted C1 to C30 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group, a substituted or unsubstituted silyl group, a halogen, or a combination thereof,Rband Rcmay independently be present or Rband Rcmay be linked with each other to form a ring,Rdand Remay independently be present or Rdand Remay be linked with each other to form a ring,Y1to Y22may independently be N, O, S, Se, Te, or a combination thereof,X1aand X1bmay independently be N, O, S, Se, Te, or CRn, andR34to R67and Rnmay independently be hydrogen, a substituted or unsubstituted C1 to C30 alkyl group, a substituted or unsubstituted C2 to C30 alkenyl group, a substituted or unsubstituted C2 to C30 alkynyl group, a substituted or unsubstituted C1 to C30 alkoxy group, a substituted or unsubstituted C6 to C30 aryl group, a substituted or unsubstituted C3 to C30 heterocyclic group, a substituted or unsubstituted amine group, a substituted or unsubstituted silyl group, a halogen or a combination thereof. For example, Z may be CRa. The compound of Formula 1 may be represented by one of Chemical Formulae 1a-2 to 1l-2. In Chemical Formulae 1a-2 to 1l-2,Ramay be a substituted or unsubstituted pyridinyl group, a substituted or unsubstituted pyrimidinyl group, a substituted or unsubstituted triazinyl group, a substituted or unsubstituted pyrrolyl group, a substituted or unsubstituted pyrazolyl group, a substituted or unsubstituted triazolyl group, C1 to C30 haloalkyl group, C6 to C30 haloaryl group, a halogen, or a cyano group, Rbto Re, Y1to Y22, X1aand X1b, R34to R67, and Rnmay be the same as described above. For example, the compound of Formula 1 may be one of compounds of Group 2, but is not limited thereto. Group 2 The compound may be a light absorbing material, for example, a light absorbing material configured to absorb light in a near infra-red wavelength spectrum. For example, a peak absorption wavelength of the compound may be for example greater than or equal to about 700 nm, greater than or equal to about 720 nm, greater than or equal to about 730 nm, greater than or equal to about 750 nm, greater than or equal to about 780 nm, greater than or equal to about 790 nm, greater than or equal to about 800 nm, greater than or equal to about 810 nm, greater than or equal to about 820 nm, greater than or equal to about 830 nm, greater than or equal to about 840 nm, greater than or equal to about 850 nm, greater than or equal to about 870 nm, greater than or equal to about 890 nm, greater than or equal to about 900 nm, or greater than or equal to about 910 nm. The peak absorption wavelength of the compound may be in a wavelength spectrum of for example about 700 nm to about 3000 nm, within the range, for example about 750 nm to about 2500 nm, about 780 nm to about 2200 nm, about 790 nm to about 2100 nm, about 800 nm to about 2000 nm, about 810 nm to about 2000 nm, about 820 nm to about 2000 nm, about 830 nm to about 2000 nm, about 850 nm to about 1900 nm, about 870 nm to about 1800 nm, about 900 nm to about 1600 nm, or about 910 nm to about 1500 nm. The compound may exhibit good charge transfer characteristics and accordingly has good photoelectric conversion characteristics for absorbing light and converting the light into an electrical signal, so that the compound may be effectively used as a photoelectric conversion material of a photoelectric diode. The compound has good heat resistance, which may prevent or reduce thermal decomposition during deposition, and thus may be deposited repeatedly. The compound may be thermally or vacuum deposited and may be deposited, for example, by sublimation. For example, deposition by sublimation may be confirmed by thermogravimetric analysis (TGA), and at a thermogravimetric analysis at a pressure of less than or equal to about 10 Pa, a temperature at which a 10% weight loss relative to an initial weight occurs may be less than or equal to about 450° C. and a temperature at which a 50% weight loss relative to an initial weight occurs may be less than or equal to about 500° C. For example, at a thermogravimetric analysis of the compound at a pressure of less than or equal to about 10 Pa, for example temperature at which a 10% weight loss relative to an initial weight occurs may be about 230° C. to about 450° C. and a temperature at which a 50% weight loss relative to an initial weight occurs may be about 300° C. to about 500° C. The compound may be produced in the form of a film. The film may be applied to various fields where absorption characteristics of the near infra-red wavelength range are required, and may be used, for example, as a near infra-red absorbing/blocking film. Since the compound has both light absorption properties and photoelectric conversion properties in a near infra-red wavelength spectrum, the compound may be effectively used as a photoelectric conversion material. Hereinafter, examples of the photoelectric diode and the organic sensor to which a compound of Formula 1 is applied are described with reference to the accompanying drawings. FIG.1is a schematic view showing an example of a pixel array of an organic sensor according to some exemplary embodiment. Referring toFIG.1, an organic sensor200may include a plurality of pixels PX, and the plurality of pixels PX may have a matrix arrangement repeatedly arranged along columns and/or rows. The plurality of pixels PX may include, for example, a unit pixel group A such as a 2×2 pixel array. However, the arrangement of the pixels is not limited thereto and may be variously modified, and the unit pixel group A may be, for example, various pixel arrays such as a 3×3 pixel array and a 4×4 pixel array. FIG.2is a cross-sectional view showing an example of a photoelectric diode according to some exemplary embodiment. Referring toFIG.2, a photoelectric diode100according to some exemplary embodiment includes a first electrode10and a second electrode20facing each other and an organic layer30between the first electrode10and the second electrode20. A substrate (not shown) may be disposed at the side of the first electrode10or the second electrode20. The substrate may be for example made of an inorganic material such as glass; an organic material such as polycarbonate, polymethylmethacrylate, polyethyleneterephthalate, polyethylenenaphthalate, polyamide, polyethersulfone, or a combination thereof; or a silicon wafer. The substrate may be omitted. One of the first electrode10and the second electrode20is an anode and the other is a cathode. For example, the first electrode10may be an anode and the second electrode20may be a cathode. At least one of the first electrode10and the second electrode20may be a light-transmitting electrode and the light-transmitting electrode may be for example made of a conductive oxide such as an indium tin oxide (ITO), indium zinc oxide (IZO), zinc oxide (ZnO), tin oxide (SnO), aluminum tin oxide (AITO), and fluorine doped tin oxide (FTO), or a metal thin layer of a single layer or a multi-layer. When one of the first electrode10and the second electrode20is a non-light-transmitting electrode, the non-light-transmitting electrode may be made of for example an opaque conductor such as aluminum (Al), silver (Ag), or gold (Au). For example, both the first electrode10and the second electrode20may be light-transmitting electrodes. For example, the second electrode20may be a light receiving electrode disposed at a light receiving side. The organic layer30may include an active layer. The active layer is a layer including a p-type semiconductor and an n-type semiconductor to provide a pn junction, which is a layer producing excitons by receiving light from outside and then separating holes and electrons from the produced excitons. The p-type semiconductor and the n-type semiconductor may be independently a light-absorbing material configured to absorb light in at least one part of a wavelength spectrum and the compound of Formula 1 may be a p-type semiconductor or an n-type semiconductor. For example, the compound of Formula 1 may be used as a p-type semiconductor and fullerene or a fullerene derivative may be included as an n-type semiconductor, but is not limited thereto. The active layer may include an intrinsic layer (I layer) in which the aforementioned p-type semiconductor and an n-type semiconductor including fullerene derivative are co-deposited. Herein, the p-type semiconductor and the n-type semiconductor may be included in a volume ratio of about 1:9 to about 9:1, for example about 2:8 to about 8:2, about 3:7 to about 7:3, about 4:6 to about 6:4, or about 5:5. The active layer may further include a p-type layer and/or an n-type layer in addition to the intrinsic layer. The p-type layer may include the aforementioned p-type semiconductor and the n-type layer may include the aforementioned n-type semiconductor. For example, they may be included in various combinations of p-type layer/I layer, I layer/n-type layer, p-type layer/I layer/n-type layer, and the like. The organic layer30includes the compound of Formula 1, and thus may effectively be configured to absorb light in a near infra-red wavelength spectrum and perform photoelectric conversion. For example, a peak absorption wavelength of the organic layer30may be for example greater than or equal to about 700 nm, greater than or equal to about 720 nm, greater than or equal to about 730 nm, greater than or equal to about 750 nm, greater than or equal to about 780 nm, greater than or equal to about 790 nm, greater than or equal to about 800 nm, greater than or equal to about 810 nm, greater than or equal to about 820 nm, greater than or equal to about 830 nm, greater than or equal to about 840 nm, greater than or equal to about 850 nm, greater than or equal to about 870 nm, greater than or equal to about 890 nm, greater than or equal to about 900 nm, or greater than or equal to about 910 nm. The peak absorption wavelength of the organic layer30may be for example in a wavelength spectrum of about 700 nm to about 3000 nm, within the range, for example about 750 nm to about 2500 nm, about 780 nm to about 2200 nm, about 790 nm to about 2100 nm, about 800 nm to about 2000 nm, about 810 nm to about 2000 nm, about 820 nm to about 2000 nm, about 830 nm to about 2000 nm, about 850 nm to about 1900 nm, about 870 nm to about 1800 nm, about 900 nm to about 1600 nm, or about 910 nm to about 1500 nm. The organic layer30may further include a charge auxiliary layer (not shown) between the first electrode10and the active layer and/or the second electrode20and the active layer. The charge auxiliary layer may make holes and electrons separated in the active layer30be transported easily to improve efficiency. The charge auxiliary layer may include at least one selected from a hole injection layer (HIL) for facilitating hole injection, a hole transport layer (HTL) for facilitating hole transport, an electron blocking layer (EBL) for preventing electron transport, an electron injection layer (EIL) for facilitating electron injection, an electron transport layer (ETL) for facilitating electron transport, and a hole blocking layer (HBL) for preventing hole transport. The charge auxiliary layer may include for example an organic material, an inorganic material, or an organic/inorganic material. The organic material may be an organic material having hole or electron characteristics and the inorganic material may be for example a metal oxide such as a molybdenum oxide, a tungsten oxide, or a nickel oxide. The charge auxiliary layer may include for example a compound of Formula 1. The photoelectric diode100may further include an anti-reflection layer (not shown) on the first electrode10or the second electrode20. The anti-reflection layer may be disposed at a light incidence side and lower reflectance of light of incident light and thereby light absorbance is further improved. For example, when light enters through the first electrode10, the anti-reflection layer may be disposed on the first electrode10while when light enters through the second electrode20, the anti-reflection layer may be disposed under the second electrode20. The anti-reflection layer may include, for example a material having a refractive index of about 1.6 to about 2.5 and may include for example at least one of a metal oxide, a metal sulfide, and an organic material having a refractive index within the ranges. The anti-reflection layer may include, for example a metal oxide such as an aluminum-containing oxide, a molybdenum-containing oxide, a tungsten-containing oxide, a vanadium-containing oxide, a rhenium-containing oxide, a niobium-containing oxide, a tantalum-containing oxide, a titanium-containing oxide, a nickel-containing oxide, a copper-containing oxide, a cobalt-containing oxide, a manganese-containing oxide, a chromium-containing oxide, a tellurium-containing oxide, or a combination thereof; a metal sulfide such as zinc sulfide; or an organic material such as an amine derivative, but is not limited thereto. In the photoelectric diode100, when light enters through the first electrode10or the second electrode20and the organic layer30may be configured to absorb light in a predetermined wavelength spectrum, excitons may be generated thereinside. The excitons may be separated into holes and electrons in the organic layer30, and the separated holes may be transported to an anode that is one of the first electrode10and the second electrode20and the separated electrons may be transported to the cathode that is the other of the first electrode10and the second electrode20so as to flow a current. The photoelectric diode100may be applied to a solar cell, an image sensor, a photodetector, or a photosensor, but is not limited thereto. The photoelectric diode may be for example applied to an organic sensor. The organic sensor may be an organic CMOS sensor, for example an organic CMOS infra-red light sensor or an organic CMOS image sensor. FIG.3is a cross-sectional view showing an organic sensor according to an exemplary embodiment. The organic sensor300according to present embodiment includes a semiconductor substrate110, an insulation layer80, and a photoelectric diode100. The semiconductor substrate110may be a silicon substrate and is integrated with a transmission transistor (not shown) and a charge storage55. The charge storage55may be integrated in each pixel. The charge storage55is electrically connected to the photoelectric diode100that will be described later and information of the charge storage55may be transferred by the transmission transistor. A metal wire (not shown) and a pad (not shown) are formed on the semiconductor substrate110. In order to decrease signal delay, the metal wire and pad may be made of a metal having low resistivity, for example, aluminum (Al), copper (Cu), silver (Ag), and alloys thereof, but are not limited thereto. Further, it is not limited to the structure, and the metal wire and pad may be disposed under the semiconductor substrate110. The insulation layer80is formed on the metal wire and pad. The insulation layer80may be made of an inorganic insulating material such as a silicon oxide and/or a silicon nitride, or a low dielectric constant (low K) material such as SiC, SiCOH, SiCO, and SiOF. The insulation layer80has a trench85exposing the charge storage55. The trench85may be filled with fillers. The aforementioned photoelectric diode100is formed on the insulation layer80. As described above, the photoelectric diode100includes a first electrode10, an organic layer30, and a second electrode20. Even though a structure in which the first electrode10, the organic layer30and the second electrode20are sequentially stacked is shown as an example in the drawing, the present disclosure is not limited to this structure, and the second electrode20, the organic layer30, and the first electrode10may be arranged in this order. The first electrode10and the second electrode20may both be transparent electrodes, and the organic layer30is the same as described above. The organic layer30may selectively absorb light in a near infra-red wavelength spectrum. Incident light from the side of the second electrode20may be photoelectrically converted by mainly absorbing light in a near infra-red wavelength spectrum in the organic layer30. A focusing lens (not shown) may be further formed on the photoelectric diode100. The focusing lens may control a direction of incident light and gather the light in one region. The focusing lens may have a shape of, for example, a cylinder or a hemisphere, but is not limited thereto. The organic sensor according to the present embodiment may be an organic infra-red light sensor, for example an iris sensor or a depth sensor. The iris sensor identifies a person by using unique iris characteristics of every person and specifically, taking an image of an eye of a user within an appropriate distance, processing the image, and comparing it with his/her stored image. The depth sensor identifies a shape and a location of an object from its three-dimensional information by taking an image of the object within an appropriate distance with a user and processing the image. This depth sensor may be for example used as a face recognition sensor. FIG.4is a cross-sectional view showing another example of an organic sensor according to an exemplary embodiment. The organic sensor according to the present embodiment may include a plurality of sensors having different functions. For example, at least one of the plurality of sensors having different functions may be a biometric sensor, and the biometric sensor may be for example an iris sensor, a depth sensor, a fingerprint sensor, a blood vessel distribution sensor, and the like, but is not limited thereto. For example, one of the plurality of sensors having different functions may be an iris sensor and the other may be a depth sensor. For example, a plurality of sensors may include, for example a first infra-red light sensor configured to sense light in a near infra-red region having a first wavelength (λ1) in a infra-red wavelength spectrum and a second infra-red light sensor configured to sense light in a near infra-red region having a second wavelength (λ2) in an infra-red wavelength spectrum. The first wavelength (λ1) and the second wavelength (λ2) may be for example different in a wavelength spectrum of about 750 nm to about 3000 nm, and for example a difference between the first wavelength (λ1) and the second wavelength (λ2) may be greater than or equal to about 30 nm, greater than or equal to about 50 nm, greater than or equal to about 70 nm, greater than or equal to about 80 nm, or greater than or equal to about 90 nm. For example, one of the first wavelength (λ1) and the second wavelength (λ2) may belong to a wavelength spectrum of about 780 nm to about 900 nm and the other of the first wavelength (λ1) and the second wavelength (λ2) may belong to a wavelength spectrum of about 830 nm to about 1000 nm. For example, one of the first wavelength (λ1) and the second wavelength (λ2) may belong to a wavelength spectrum of about 780 nm to about 840 nm and the other of the first wavelength (λ1) and the second wavelength (λ2) may belong to a wavelength spectrum of about 910 nm to about 970 nm. For example, one of the first wavelength (λ1) and the second wavelength (λ2) may belong to a wavelength spectrum of about 800 nm to about 830 nm and the other of the first wavelength (λ1) and the second wavelength (λ2) may belong to a wavelength spectrum of about 930 nm to about 950 nm. For example, one of the first wavelength (λ1) and the second wavelength (λ2) may belong to a wavelength spectrum of about 805 nm to about 815 nm and the other of the first wavelength (λ1) and the second wavelength (λ2) may belong to a wavelength spectrum of about 935 nm to about 945 nm. For example, one of the first wavelength (λ1) and the second wavelength (λ2) may about 810 nm and the other of the first wavelength (λ1) and the second wavelength (λ2) may be about 940 nm. The organic sensor400according to the present embodiment includes a dual bandpass filter40, a first infra-red light sensor100A, an insulation layer80, and a semiconductor substrate110integrated with a second infra-red light sensor120. The first infra-red light sensor100A and the second infra-red light sensor120may be stacked. The dual bandpass filter40may be disposed on a front side of the organic sensor400and may selectively transmit infra-red light including the first wavelength (λ1) and infra-red light including the second wavelength (λ2) and may block and/or absorb other light. Herein, other light may include light in an ultraviolet (UV) and visible region. The first infra-red light sensor100A may be the photoelectric diode100according to the aforementioned embodiment and details thereof are omitted. The second infra-red light sensor120may be integrated in the semiconductor substrate110and may be a photo-sensing device. The semiconductor substrate110may be for example a silicon substrate and may be integrated with the second infra-red light sensor120, the charge storage55, and a transmission transistor (not shown). The second infra-red light sensor120may be a photodiode and may sense entered light, and sensed information is transferred by the transmission transistor. Herein, the light entered into the second infra-red light sensor120is light that passes the dual bandpass filter40and the first infra-red light sensor100A and may be infra-red light in a predetermined region including the second wavelength (λ2). All infra-red light in a predetermined region including the first wavelength (λ1) may be absorbed by the organic layer30and may not reach the second infra-red light sensor120. In this case, a separate filter for wavelength selectivity with respect to the light entered into the second infra-red light sensor120is not separately needed. However, for the time when all infra-red light in a predetermined region including the first wavelength (λ1) is not absorbed by organic layer30, a filter between the first infra-red light sensor100A and the second infra-red light sensor120may be further disposed. The organic sensor according to the present embodiment may include two infra-red light sensors respectively performing separately functions and thus may work as a combination sensor. In addition, two sensors performing separately functions are stacked in each pixel, and thus the number of pixel performing functioning of each sensor is twice increased while maintaining a size and resultantly, sensitivity may be much improved. FIG.5is a cross-sectional view showing another example of an organic sensor according to some exemplary embodiment. An organic sensor according to the present embodiment may be an organic CMOS image sensor. Referring toFIG.5, an organic sensor500according to an embodiment includes a semiconductor substrate110integrated with photo-sensing devices50a,50b, and50c, a transmission transistor (not shown), and a charge storage55, a lower insulation layer60, color filter layers70a,70b, and70c, an upper insulation layer80, and a photoelectric diode100. The semiconductor substrate110may be integrated with photo-sensing devices50a,50b, and50c, a transmission transistor (not shown), and a charge storage55. The photo-sensing devices50a,50b, and50cmay be photodiodes. The photo-sensing devices50a,50b, and50c, the transmission transistor, and/or the charge storage55may be integrated in each pixel. For example, the photo-sensing device50amay be included in a red pixel, the photo-sensing device50bmay be included in a green pixel, and the photo-sensing device50cmay be included in a blue pixel. The photo-sensing devices50a,50b, and50csense light, the information sensed by the photo-sensing devices may be transferred by the transmission transistor, the charge storage55is electrically connected to the photoelectric diode100that will be described later, and the information of the charge storage55may be transferred by the transmission transistor. A metal wire (not shown) and a pad (not shown) are formed on the semiconductor substrate110. In order to decrease signal delay, the metal wire and pad may be made of a metal having low resistivity, for example, aluminum (Al), copper (Cu), silver (Ag), and alloys thereof, but are not limited thereto. However, it is not limited to the structure, and the metal wire and pad may be disposed under the photo-sensing devices50a,50b, and50c. The lower insulation layer60is formed on the metal wire and the pad. Color filters70a,70b, and70care formed on the lower insulation layer60. The color filters70a,70b, and70cincludes a red filter70aformed in a red pixel, a green filter70bformed in a green pixel, and a blue filter70cformed in a blue pixel. The upper insulation layer80is formed on the color filters70a,70b, and70c. The upper insulation layer80eliminates steps caused by the color filters70a,70b, and70cand planarizes the surface. The photoelectric diode100is formed on the upper insulation layer80. As described above, the photoelectric diode100includes a first electrode10, an organic layer30, and a second electrode20. Even though a structure in which the first electrode10, the organic layer30and the second electrode20are sequentially stacked is shown as an example in the drawing, the present disclosure is not limited to this structure, and the second electrode20, the organic layer30, and the first electrode10may be arranged in this order. The first electrode10and the second electrode20may both be transparent electrodes, and the organic layer30is the same as described above. The organic layer30may be configured to selectively absorb light in a near infra-red wavelength spectrum. Incident light from the side of the second electrode20may be configured to be photoelectrically converted by mainly absorbing light in a near infra-red wavelength spectrum in the organic layer30. Light in the remaining wavelength spectrum may pass through the first electrode10and the color filters70a,70b, and70c, the light in a red wavelength spectrum passing through the color filter70amay be sensed by the photo-sensing device50a, the light in a green wavelength spectrum passing through the color filter70bmay be sensed by the photo-sensing device50b, and the light in a blue wavelength spectrum passing through the color filter70cmay be sensed by the photo-sensing device50c. FIG.6is a schematic cross-sectional view of another example of an organic sensor according to some exemplary embodiment. Referring toFIG.6, an organic sensor600according to an embodiment includes a semiconductor substrate110including photo-sensing devices50a,50b, and50c, a transmission transistor (not shown), and a charge storage55, a lower insulation layer60, a color filter layer70including color filters70a,70b, and70c, and a photoelectric diode100. Referring toFIG.6, the photoelectric diode100may be disposed between the semiconductor substrate110and the color filter layer70, such that the color filter layer70is far from the photo-sensing devices50a,50b, and50cwith respect to the photoelectric diode100. Other constituent elements are the same as the organic sensor ofFIG.5. For example, the color filter layer70may further include a mixed color filter configured to transmit a wavelength spectrum of light of mixed colors. For example, inFIG.6, the color filter70amay be configured to selectively filter light in a magenta wavelength spectrum, the color filter70bmay be configured to selectively filter light in a cyan wavelength spectrum, and the color filter70cmay be configured to selectively filter light in a yellow wavelength spectrum. Herein, the photo-sensing device50amay be configured to sense blue light and the photo-sensing device50bmay be configured to sense red light. FIG.7is a schematic cross-sectional view of another example of an organic sensor according to some exemplary embodiment. Referring toFIG.7, an organic image sensor700according to an embodiment includes a semiconductor substrate110including photo-sensing devices50a,50b, and50c, a transmission transistor (not shown), and a charge storage55; a lower insulation layer60, a color filter layer70including color filters70a,70b, and70c; an upper insulation layer80; and a photoelectric diode100under the semiconductor substrate110. As shown inFIG.7, the photoelectric diode100is disposed under the semiconductor substrate110, and thereby the color filter layer70is far from the photoelectric diode100with respect to the photo-sensing devices50a,50b, and50c. Other constituent elements are the same as the organic sensor ofFIG.5. FIG.8is a schematic cross-sectional view showing an organic sensor according to some exemplary embodiment. Referring toFIG.8, an organic sensor800according to an embodiment includes a semiconductor substrate110including photo-sensing devices50a,50b, and50c, a transmission transistor (not shown), and a charge storage55; an insulation layer60having a trench85, and a photoelectric diode100. In the organic sensor800, the photo-sensing devices50a,50b, and50care stacked in a vertical direction and the color filter layer70is omitted. The photo-sensing devices50a,50b, and50care electrically connected to charge storage (not shown) and their information may be transferred by the transmission transistor. The photo-sensing devices50a,50b, and50cmay selectively absorb light in each wavelength spectrum of light depending on a stack depth. Other constituent elements are the same as the organic sensor ofFIG.5. FIG.9is a schematic top view of an example of an organic sensor according to some exemplary embodiment andFIG.10is a schematic cross-sectional view of the organic sensor ofFIG.9. Referring toFIGS.9and10, an organic sensor900according to an embodiment includes a near infra-red photoelectric diode configured to selectively absorb light in a near infra-red wavelength spectrum, a red photoelectric diode configured to selectively absorb light in a red wavelength spectrum and convert it into electrical signals, a green photoelectric diode configured to selectively absorb light in a green wavelength spectrum and convert it into electrical signals, and a blue photoelectric diode configured to selectively absorb light in a blue wavelength spectrum and convert it into electrical signals. The near infra-red photoelectric diode, red photoelectric diode, green photoelectric diode, and blue photoelectric diode are arranged in parallel in a horizontal direction. Referring toFIG.10, the organic sensor900according to an embodiment includes a photoelectric diode100that includes a plurality of photoelectric diodes100a,100b,100c, and100don a semiconductor substrate110. The plurality of photoelectric diodes100a,100b,100c, and100dare configured to absorb light in one wavelength spectrum of the red wavelength spectrum, blue wavelength spectrum, green wavelength spectrum, and near infra-red wavelength spectrum and to convert it into electrical signals. Referring toFIG.10, an organic sensor900according to an embodiment includes a semiconductor substrate110integrated with photo-sensing devices55ato55d, and a transmission transistor (not shown); a lower insulation layer60; and photoelectric diodes100ato100d. The photoelectric diodes100ato100dare disposed in parallel on the semiconductor substrate110and are partially overlapped with each other. Each of the photoelectric diodes100ato100dare overlapped with each other in a parallel direction on the surface110aof the semiconductor substrate110. Each of the photoelectric diodes100ato100dmay be electrically connected to the charge storage55that is integrated into the semiconductor substrate110, via the trench85. One of photoelectric diodes100ato100dmay be the aforementioned photoelectric diode100. For example, the photoelectric diodes100ato100dmay share the same common electrode as the second electrode20. Other constituent elements are the same as the organic sensor ofFIG.5. FIG.11is a schematic cross-sectional view of an example of an organic sensor according to some exemplary embodiment. Referring toFIG.11, an organic sensor1100according to an embodiment includes a semiconductor substrate110integrated with a charge storage and a transmission transistor (not shown); a lower insulation layer60; a first photoelectric diode1190, a second photoelectric diode1190a, a third photoelectric diode1190b; and a fourth photoelectric diode1190c. The first photoelectric diode1190may be a photoelectric diode that absorbs light in a near infra-red wavelength spectrum and may be formed on the second to fourth photoelectric diodes1190ato1190c. The first photoelectric diode1190may be the aforementioned photoelectric diode100. The second to fourth photoelectric diodes1190ato1190cmay be configured to selectively absorb light in a different wavelength spectrum of a blue wavelength spectrum, a red wavelength spectrum, and a green wavelength spectrum. For example, the second to fourth photoelectric diodes1190ato1190cmay share a common electrode1120, and may include a separate pixel electrode1110and separate photoelectric conversion layers1130a,1130b, and1130c, respectively. Other constituent elements are the same as the organic sensor ofFIG.5. Referring toFIG.11, the first photoelectric diode1190are stacked on the second to fourth photovoltaic elements1190ato1190c, which overlap each other in a direction perpendicular to the surface of the semiconductor substrate110. The second to fourth photoelectric diodes1190ato1190cmay partially overlap in a direction parallel to the surface of the semiconductor substrate110. FIG.12is a schematic perspective view of an example of an organic sensor according to some exemplary embodiment andFIG.13is a schematic cross-sectional view according to one example of the organic sensor ofFIG.12. Referring toFIG.12, an organic sensor1200according to an embodiment includes a photoelectric diode configured to selectively absorb light in a near infra-red wavelength spectrum, a photoelectric diode configured to selectively absorb and to electrically convert light in a red wavelength spectrum, a photoelectric diode configured to selectively absorb and to electrically convert light in a green wavelength spectrum, and a photoelectric diode configured to selectively absorb and to electrically convert light in a blue wavelength spectrum. Referring toFIG.13, an organic sensor1200according to an embodiment includes a semiconductor substrate110integrated with charge storages55ato55d, and transmission transistors; a lower insulation layer80a; intermediate insulation layers80band80c; an upper insulation layer80d; a first photoelectric diode1200a; a second photoelectric diode1200b; a third photoelectric diode1200c; and a fourth photoelectric diode1200d. The first to fourth photoelectric diodes1200ato1200dare vertically stacked on the semiconductor substrate110. The first photoelectric diode1200aincludes a first electrode10aand a second electrode20afacing each other, and a photoelectric conversion layer30adisposed between the first electrode10aand the second electrode20a. The second photoelectric diode1200bincludes a first electrode10band a second electrode20bfacing each other, and a photoelectric conversion layer30bdisposed between the first electrode10band the second electrode20b. The third photoelectric diode1200cincludes a first electrode10cand a second electrode20cfacing each other, and a photoelectric conversion layer30cdisposed between the first electrode10cand the second electrode20c. The fourth photoelectric diode1200dincludes a first electrode10dand a second electrode20dfacing each other, and a photoelectric conversion layer30ddisposed between the first electrode10dand the second electrode20d. The photoelectric conversion layers30a,30b,30c, and30dmay selectively absorb light in one wavelength spectrum of the red wavelength spectrum, the green wavelength spectrum, the blue wavelength spectrum, and the near infra-red wavelength spectrum and may photoelectrically convert the light. One of the photoelectric conversion layers30a,30b,30c, and30dmay be the aforementioned organic layer30. The first electrodes10a,10b,10c, and10d, and the second electrodes20a,20b,20c, and20dare the same as the first electrode10and the second electrode20that are described above. Focusing lens1300may be further formed on the fourth photoelectric diode1200d. The focusing1300lens may control a direction of incident light and gather the light in one region. The focusing lens1300may have a shape of, for example, a cylinder or a hemisphere, but is not limited thereto. In the drawing, the first to fourth photoelectric diodes1200ato1200dare sequentially stacked, but the present disclosure is not limited thereto, and they may be stacked in various orders. FIG.14is a schematic diagram of an electronic device according to some exemplary embodiment. Referring toFIG.14, an electronic device1400may include a processor1420, a memory1430, and an organic sensor1440that are electrically coupled together via a bus1410. The organic sensor1440may be an organic sensor of any of the aforementioned embodiments The memory1430, which may be a non-transitory computer readable medium, may store a program of instructions. The processor1420may execute the stored program of instructions to perform one or more functions. For example, the processor1420may be configured to process electrical signals generated by the organic sensor1440. The processor1420may be configured to generate an output (e.g., an image to be displayed on a display interface) based on such as processing. The organic sensor may be applied to various electronic devices, for example and the electronic devices may include for example a camera, a camcorder, a mobile phone internally having them, a display device, a security device, or a medical device, but are not limited thereto. Hereinafter, the embodiments are illustrated in more detail with reference to examples. However, these examples are exemplary, and the present scope is not limited thereto. Simulation Evaluation of Light Absorption Properties The light absorption properties of compounds are evaluated using a Gaussian09 program, a wave function is expressed by using a DGDZVP basis-set, an optimal structure is obtained through B3LYP hybrid density functional calculation, and then, the structure is used for a B3LYP hybrid density functional or ωB97X-D range-separated density functional time-dependent DFT (density functional theory) calculation, and thus an adsorption wavelength is obtained (by calculating an energy difference between singlet exited state and ground state). The results are shown in Table 1. TABLE 1PeakabsorptionAbsorptionwavelengthintensity(λmax) (nm)(a.u.)Compound 328961.72Compound 348951.72Compound 358721.72Compound 369471.72Compound 389141.89Compound 409101.84Compound 418771.90Compound 429741.91Compound 628721.94Compound 658581.94Compound 669181.95Compound 688962.19Compound 708862.14Compound 718762.23Compound 729532.23 SYNTHESIS EXAMPLES Synthesis Example 1: Synthesis of Compound 35 Synthesis of Intermediate I-11 1 g of 4H-thieno[3,2-b]pyrrole-5-carboxylic acid is dissolved in 50 ml of ethyl alcohol. 10 ml of 0.1 N HCl is added thereto and then, refluxed and stirred at 100° C. for 24 hours and cooled down to room temperature. When a reaction is complete, after evaporating the ethyl alcohol under a reduced pressure, 200 ml of ethyl acetate is poured thereinto for dilution. An organic layer therefrom is washed with a saturated NaHCO3aqueous solution (50 ml×2) and a saturated NaCl aqueous solution (50 ml×2), dried with MgSO4, filtered, and evaporated under a reduced pressure. The residue is purified by column chromatography (ethyl acetate/hexane=1:5 v/v) to obtain 0.8 g of Intermediate I-11. A yield thereof is 69%. The produced compound is identified by using LC-MS. LC-MS m/z=196.24 (M+H)+ Synthesis of Intermediate I-12 1 g of ethyl4H-thieno[3,2-b]pyrrole-5-carboxylate (Intermediate I-11) was dissolved in 50 ml of dichloromethane. 0.2 g of bromine is slowly added thereto in a dropwise fashion and then, stirred at room temperature for 24 hours. When a reaction is complete, an organic layer therefrom is diluted with 100 ml of dichloromethane, washed with a NaHCO3aqueous solution (50 ml×2) and a saturated NaCl aqueous solution (50 ml×2), dried with MgSO4, and filtered, and evaporated under a reduced pressure. The residue is purified by column chromatography (ethyl acetate/hexane=1:5 v/v) to obtain 1.7 g of Intermediate I-12. A yield thereof is 94%. The produced compound is identified using LC-MS. LC-MS m/z=354.03 (M+H)+ Synthesis of Intermediate I-13 2.3 g (6.94 mmol) of ethyl2,6-dibromo-4H-thieno[3,2-b]pyrrole-5-carboxylate (Intermediate I-12) and 1.5 g (7.64 mmol) of (4-(dimethylamino)phenyl)boronic acid are put in a flask, and 150 ml of a mixed solvent of tetrahydrofuran/distilled water (a volume ratio of 4:1) is added thereto under a nitrogen flow. Then, 0.4 g (0.35 mmol) of tetrakis(triphenylphosphine)palladium (0) and 2.21 g (20.83 mmol) of potassium carbonate are sequentially added thereto and stirred at 110° C. for 24 hours. When a reaction is complete, after decreasing the temperature down to room temperature, 500 ml of ethyl acetate is added thereto for dilution. An organic layer therefrom is washed with a saturated NaHCO3aqueous solution (150 ml×2) and a saturated NaCl aqueous solution (100 ml×2), dried with MgSO4, filtered, and evaporated under a reduced pressure. The residue is purified by column chromatography (ethyl acetate/hexane=1:3 v/v) to obtain 2.3 g of Intermediate I-13. A yield thereof is 89%. The produced compound is identified using LC-MS. LC-MS m/z=394.30 (M+H)+ Synthesis of Intermediate I-14 5.3 g (7.64 mmol) of ethyl6-bromo-2-(4-(dimethylamino)phenyl)-4H-thieno[3,2-b]pyrrole-5-carboxylate (Intermediate I-13) and 1.5 g (6.94 mmol) of (4-(trifluoromethyl)phenyl)boronic acid are put in a flask, and 150 ml of a mixed solvent of toluene/ethanol/water (a volume ratio of 3:1:1) is added thereto under a nitrogen flow. Subsequently, 0.4 g (0.35 mmol) of tetrakis (triphenylphosphine)palladium (0) and 2.21 g (20.83 mmol) of potassium carbonate are sequentially added thereto and then, stirred at 120° C. for 24 hours. When a reaction is complete, after decreasing the temperature down to room temperature, 500 ml of ethyl acetate is added thereto for dilution. An organic layer therefrom is washed with a saturated NaHCO3aqueous solution (150 ml×2) and a saturated NaCl aqueous solution (100 ml×2), dried with MgSO4, filtered, and evaporated under a reduced pressure. The residue is purified by column chromatography (ethyl acetate/hexane=1:3 v/v) to obtain 2.3 g of Intermediate I-14. A yield thereof is 37%. The produced compound is identified by using LC-MS. LC-MS m/z=459.50 (M+H)+ Synthesis of Intermediate I-15 1.0 g (2.18 mmol) of ethyl-(4-(dimethylamino)phenyl)-6-(4-(trifluoromethyl)phenyl)-4H-thieno[3,2-b]pyrrole-5-carboxylate (Intermediate I-14) is added to 10 ml of ethylene glycol and then, stirred at room temperature. 6.1 g (108.73 mmol) of potassium hydroxide is added thereto and then, stirred at 130° C. for 24 hours. When a reaction is complete, after decreasing the temperature to room temperature, 500 ml of ethyl acetate is added thereto for dilution. An organic layer therefrom is washed with a saturated NaHCO3aqueous solution (150 ml×2) and a saturated NaCl aqueous solution (100 ml×2), dried with MgSO4, filtered, and evaporated under a reduced pressure. The residue is purified by column chromatography (ethyl acetate/hexane=1:3 v/v) to obtain 0.5 g of Intermediate I-15. A yield thereof is 59%. The produced compound is identified by using LC-MS. LC-MS m/z=387.11 (M+H)+ Synthesis of Intermediate I-16 0.80 g (2.07 mmol) of N,N-dimethyl-4-(6-(4-(trifluoromethyl)phenyl)-4H-thieno[3,2-b]pyrrol-2-yl)aniline (Intermediate I-15) is added to a mixed solvent of 2.0 ml/1.0 ml of acetic acid/acetic anhydride. After decreasing a temperature to 0° C., 1.60 g (2.31 mmol) of sodium nitrite (NaNO2) is added thereto and then, stirred for 15 minutes. Subsequently, 0.80 g (2.07 mmol) of N,N-dimethyl-4-(6-(4-(trifluoromethyl)phenyl)-4H-thieno[3,2-b]pyrrol-2-yl)aniline (Intermediate I-15) is additionally added thereto. Then, the reaction solution is heated up to 80° C. and then, stirred for 3 hours. After decreasing the temperature down to room temperature, a solid generated therein is filtered and washed with n-hexane to obtain 0.70 g of Intermediate I-16. A yield thereof is 43%. The produced compound is identified by using LC-MS. LC-MS m/z=784.19 (M+H)+ Synthesis of Compound 35 0.70 g (0.89 mmol) of (Z)-2-(4-(dimethylamino)phenyl)-N-(2-(4-(dimethylamino)phenyl)-6-(4-(trifluoromethyl)phenyl)-5H-thieno[3,2-b]pyrrol-5-yl-dene)-6-(4-(trifluoromethyl)phenyl)-4H-thieno[3,2-b]pyrrole-5-amine (Intermediate I-16) is put in a flask and dissolved in 3.0 ml of toluene under a nitrogen flow, and 1.0 ml of triethylamine is added thereto. 0.3 mL of borontrifluoride diethyl etherate is slowly added thereto in a dropwise fashion and then, stirred at 80° C. for 3 hours. When a reaction is complete, after decreasing the temperature down to room temperature, precipitates therein are gathered and washed with ethanol, and the residue is purified by column chromatograph y(dichloromethane/hexane=1:3 v/v) to obtain 0.2 g of Compound 35. A yield thereof is 27%. The produced compound is identified using LC-MS. LC-MS m/z=832.19 (M+H)+ Synthesis Example 2: Synthesis of Compound 99 Synthesis of Intermediate I-21 Intermediate I-21 is obtained according to the aforementioned synthesis method of Intermediate I-12. Synthesis of Intermediate I-22 1.0 g (2.83 mmol) of ethyl 2,6-dibromo-4H-thieno[3,2-b]pyrrole-5-carboxylate (Intermediate I-21) is dissolved in 50 ml of toluene and then, put in an ice bath, and a temperature thereof is decreased. 3.0 ml of tributyl(thiophen-2-yl)stannane is slowly added thereto in a dropwise fashion, and the obtained mixture is heated up to room temperature and 30 minutes later, refluxed and stirred at 120° C. for 24 hours. When a reaction is complete, after decreasing the temperature down to room temperature, 500 ml of ethyl acetate is poured thereinto for dilution. An organic layer therefrom is washed with a saturated NaHCO3aqueous solution (150 ml×2) and a saturated NaCl aqueous solution (100 ml×2), dried with MgSO4, filtered, and evaporated under a reduced pressure. The residue is purified by column chromatography (ethyl acetate/hexane=1:3 v/v) to obtain 0.5 g of Intermediate I-22. A yield thereof is 49%. The produced compound is identified using LC-MS. LC-MS m/z=360.49 (M+H)+ Synthesis of Intermediate I-23 1.0 g (2.78 mmol) of ethyl 2,6-di(thiophene-2-yl)-4H-thieno[3,2-b]pyrrole-5-carboxylate (Intermediate I-22) is put in 30 ml of ethylene glycol and then, stirred at room temperature. 7.8 g (139.0 mmol) of potassium hydroxide is added thereto and then, stirred at 130° C. for 24 hours. When a reaction is complete, after decreasing the temperature down to room temperature, 500 ml of ethyl acetate is poured thereto for dilution. An organic layer therefrom is washed with a saturated NaHCO3aqueous solution (150 ml×2) and a saturated NaCl aqueous solution (100 ml×2), dried with MgSO4, filtered, and evaporated under a reduced pressure. The residue is purified by column chromatography (ethyl acetate/hexane=1:3 v/v) to obtain 0.4 g of Intermediate I-23. A yield thereof is 50%. The produced compound is identified by using LC-MS. LC-MS m/z=287.99 (M+H) Synthesis of Intermediate I-24 0.5 g (1.74 mmol) of 2,6-di(thiophene-2-yl)-4H-thieno[3,2-b]pyrrole (Intermediate I-23) is added to a mixed solvent of 2.0 ml/1.0 ml of acetic acid/acetic anhydride. Subsequently, after decreasing a temperature down to 0° C., 1.32 g (1.91 mmol) of sodium nitrite (NaNO2) is added thereto and then, stirred for 15 minutes. 0.5 g (1.74 mmol) of 2,6-di(thiophene-2-yl)-4H-thieno[3,2-b]pyrrole (Intermediate I-23) is added thereto. Subsequently, the reaction solution is heated up to 80° C. and then, stirred for 3 hours. The reaction solution is cooled down to room temperature, and a solid generated therein is filtered and washed with ethanol to obtain 0.45 g of Intermediate I-24. A yield thereof is 44%. The produced compound is identified by using LC-MS. LC-MS m/z=585.98 (M+H)+ Synthesis of Compound 99 0.4 g (0.7 mmol) of (Z)—N-(2,6-di(thiophene-2-yl)-5H-thieno[3,2-b]pyrrole-5-yl-diene)-2,6-di(thiophene-2-yl)-4H-thieno[3,2-b]pyrrole-5-amine (Intermediate I-24) is put in a flask and dissolved in 3.0 ml of toluene under a nitrogen flow, and 1.0 ml of triethylamine is added thereto. 0.2 mL of borontrifluoride diethyl etherate is slowly added thereto in a dropwise fashion and then, stirred at 80° C. for 3 hours. When a reaction is complete, after decreasing the temperature down to room temperature, precipitates therein are gathered, washed with ethanol, and the residue is purified by column chromatography (dichloromethane/hexane=1:3 v/v) to obtain 0.1 g of Compound 99. A yield thereof is 23%. The produced compound is identified by using LC-MS. LC-MS m/z=634.63 (M+H)+ Evaluation I The compounds of Synthesis Examples are respectively dissolved in dichloromethane at a concentration of 1×10−5M to prepare solutions to evaluate light absorption properties of the compounds. The light absorption properties are evaluated by measuring a peak absorption wavelength (λmax) with a Shimadzu UV-3600 Plus UV-Vis-NIR (UV-Vis-NIR) spectrometer. The results are shown in Table 2. TABLE 2λmax(nm)Synthesis Example 1963Synthesis Example 2819 Referring to Table 2, absorption spectra of the compounds of Synthesis Examples show a peak absorption wavelength in a near infra-red wavelength spectrum. Evaluation II Deposition characteristics of the compounds of Synthesis Examples are evaluated. The deposition characteristics are evaluated by sublimating the compounds under high vacuum of 10 Pa or less and measuring a weight loss of the compounds depending on a temperature increase in a thermogravimetric analysis method. The results are shown in Table 3. TABLE 3Ts(° C.)Ts(° C.)(−10 wt %)(−50 wt %)Synthesis Example 1220° C.280° C.Synthesis Example 2200° C.260° C.* Ts(° C.) (−10 wt %): A temperature at which a specimen loses 10 wt % of a weight* Ts(° C.) (−50 wt %): A temperature at which a specimen loses 50 wt % of a weight Referring to Table 3, the compounds of Synthesis Examples have satisfactory heat resistance and are formed into a thin film through repetitive thermal deposition. While this disclosure has been described in connection with what is presently considered to be practical exemplary embodiments, it is to be understood that the invention is not limited to the disclosed embodiments. On the contrary, it is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.
75,515
11858951
DESCRIPTION OF THE INVENTION The present invention provides compounds of formula (I) wherein (I)LY denotes (CH2)m, wherein 1 to 4H atoms may be replaced by Hal, R3aand/or OR4a, and/or wherein one CH2group may be replaced by O, S, SO or SO2;X denotes a heterobicycle or heterotricycle of formula (xa), (xb), (xc), (xd), (xe), (xf), (xg), (xh) or (xi) each, independently from one another, unsubstituted or mono-, di- or trisubstituted by Hal, NO2, CN, R5a, OR5a, CONR5aR5b, NR5aCOR5b, SO2R5a, SOR5a, SO2NR5aR5b, NR5aSO2R5b, NR5aR5b, (CH2)q—R6, COR5aand/or SO2R5a, and wherein 1, 2 or 3 of the cyclic CH2groups may be replaced CR4aR4b, C═O, O, S, NR5a, SO and/or SO2; (optional substituents of (xa)-(xi) not shown)Y denotes P1, P2or P3;P1denotes a linear or branched C1-C6-alkyl or C3-C3-cycloalkyl, each, independently from one another, unsubstituted or mono-, di-, tri- or tetrasubstituted by Hal, CN, R3a, OR3a, and/or (CH2)q—R6;P2denotes phenyl or an aromatic monocyclic 5-, 6- or 7-membered heterocycle, each unsubstituted or mono-, di-, tri-, tetra- or pentasubstituted by Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6, wherein the heterocyclic system contains 1, 2 or 3 N, O and/or S atoms;P3denotes a bicyclic 8-, 9- or 10-membered hydrocarbon or heterocycle, each independently from one another unsubstituted or mono-, di-, tri-, tetra- or pentasubstituted by Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6, wherein at least one ring of the bicyclic hydrocarbon or heterocycle is aromatic, and wherein the heterocyclic system contains 1, 2 or 3 N, O and/or S atoms;Cy1, Cy2, Cy3, Cy4and Cy5denote each, independently from one another, Ar1or Het1;R1, R2denote each, independently from one another, H or C1-C6-alkyl, or R1and R2form together a residue according to formula (CE) R3a, R3bdenote each, independently from one another, linear or branched C1-C6-alkyl or C3-C8cycloalkyl, wherein 1 to 5H atoms may be replaced by Hal, CN, OH and/or OAlk;R4a, Rob denote each, independently from one another, H or R3a; orR4aand R4bform together a C3-C8alkylene group;R5a, R5bdenote each, independently from one another, H, R3a, Ar2or Het2;R6denotes OH or OR3a;T1, T2, T3, T4, T5, T6, T7, T8and T9denote each, independently from one another, O, SO, C═O;Alk denotes linear or branched C1-C6-alkyl;Ar1represents an aromatic 6-membered carbocycle;Het1represents a saturated, unsaturated or aromatic 5- or 6-membered heterocycle having 1 to 4 N, O and/or S atoms;Ar2denotes phenyl, which is unsubstituted or mono- or disubstituted by Hal, NO2, CN, R3a, OR3a, CONHR3a, NR3aCOR3b, SO2R3a, SOR3a, NH2, NHR3a, N(R3a)2and/or (CH2)q—R6;Het2denotes a saturated, unsaturated or aromatic 5- or 6-membered heterocycle having 1 to 4 N, O and/or S atoms, which is unsubstituted or mono- or disubstituted by Hal, NO2, CN, R3a, OH, OR3a, CONHR3a, NR3aCOR3b, SO2R3a, SOR3a, NH2, NHR3a, N(R3a)2, (CH2)q—R6and/or oxo (═O);q denotes 1, 2, 3, 4, 5 or 6;m denotes 0, 1 or 2;Hal denotes F, Cl, Br or I; and prodrugs, solvates, tautomers, oligomers, adducts and stereoisomers thereof as well as the pharmaceutically acceptable salts of each of the foregoing, including mixtures thereof in all ratios. Compounds of the present invention are inhibitors of the immunoproteasome subunit LMP7. They show a particularly high selectivity on LMP7 over Beta5 (cP) and good properties in terms of solubility, plasma-protein binding, CYP inhibition, PK profile and oral bioavailabiliy. It is known that boronic acid derivatives such as compounds of formula (I), wherein R1and R2denote H form oligomeres (Boronic Acids. Edited by Dennis G. Hall, Copyright (c) 2005 WILEY-VCH Verlag, GmbH & Co. KGaA, Weinheim, ISBN 3-527-30991-8). Such oligomeres (in particular but not limited to dimers or trimers) of compounds of formula (I) are included within this invention. Known cyclic trimers of boronic acids have for example following structure: It is also known that boronic acid derivatives such as compounds of formula (I), wherein R1and R2denote H form adducts by reaction with aliphatic or aromatic alcohols, diols, sugars, sugar alcohols, α-hydroxy acids or nucleophiles containing one, two or three N-/O-containing functional group (e.g. —NH2, —CONH2or C═NH, —OH, —COOH) wherein in case that three functional groups are present, one of the three heteroatoms might form a coordinative bond (“Boronic Acids” Edited by Dennis G. Hall, 2ndEdition, Copyright (c) 2011 WILEY-VCH Verlag, GmbH & Co. KGaA, Weinheim, ISBN 978-3-527-32598-6; WO2013128419; WO2009154737). The adduct formation is particularly fast with preorganized diols. The present invention includes such adducts (in particular esters or heterocyclic derivatives) of boronic acid compounds of formula (I). It is to be noted that the compounds of the present invention bear a stereogenic center at the carbon atom adjacent to the boronic acid residue; it has been denoted with an asterix (*) in formula (I)* below: The compounds according to formula (I) thus exhibit two different configurations at this stereogenic center, i.e. the (R)-configuration and the (S)-configuration. Hence, the compounds of the present invention may be present either enantiopure or as a racemic (1:1) mixture of the two enantiomers of formula (R)-(I) and (S)-(I). Compounds of formula (I) may also be present in a mixture in which one of the enantiomers (R)-(I) or (S)-(I) is present in an excess over the other one, e.g. 60:40, 70:30, 80:20, 90:10, 95:5 or the like. In a particular embodiment of the present invention the stereoisomer of formula (R)-(I) of the compound of formula (Ia) and the stereoisomer of formula (S)-(I) of the compound of formula (Ia) are present in a ratio of (R)-(I) to (S)-(I) of at least 90 parts of (R)-(I) to not more than 10 parts of (S)-(I), preferably of at least 95 (R)-(I) to not more than 5 (S)-(I), more preferably of at least 99 (R)-(I) to not more than 1 (S)-(I), even more preferably of at least 99.5 (R)-(I) to not more than 0.5 (S)-(I). In another particular embodiment of the present invention the stereoisomer of formula (S)-(I) of the compound of formula (Ia) and the stereoisomer of formula (R)-(I) of the compound of formula (Ia) are present in a ratio of (S)-(I) to (R)-(I) of at least 90 (S)-(I) to not more than 10 (R)-(I), preferably of at least 95 (S)-(I) to not more than 5 (R)-(I), more preferably of at least 99 (S)-(I) to not more than 1 (R)-(I), even more preferably of at least 99.5 (S)-(I) to not more than 0.5 (R)-(I). Enriched or pure stereoisomers of formulas (R)-(I) and (S)-(I) can be obtained by usual methods known in the art and the specific methods described hereinafter. A particular method for obtaining them is preparative column chromatography, such as HPLC or SFC, using chiral column material. Particular preferred embodiments of the present invention comprise compounds of formula (R)-(I), wherein the stereogenic center at the carbon atom adjacent to the boronic acid residue has an (R)-configuration: The compounds according to formula (I) might also carry further stereogenic centers located at carbon atoms other than the carbon atom adjacent to the boronic acid residue. All of these stereogenic centers may occur in (R)- or (S)-configuration. In particular, the compounds of the present invention bear further stereogenic centers at the carbon atom of substituent X, which is directly attached to the carbon atom of the amide group CONH shown in formula (I)) and at the carbon atoms adjacent to the bridge atom; these stereogenic centers are for example shown in formula (xa*) below, wherein they are denoted with an asterix (*): (optional substitutents of (xa*) not shown) The possible stereoisomers of (xa*) are shown below: The compounds according to formula (I) thus also exhibit two different configurations at these stereogenic centers, i.e. the (R)-configuration and the (S)-configuration. The compounds of the present invention may have either an (R)-configuration or (S)-configuration at each of these stereogenic centers or the compounds are present in a racemic (1:1) mixture of two stereoisomers. The compounds of formula (I) may also be present in a mixture in which one of the stereoisomers is present in an excess over the other one, e.g. 60:40, 70:30, 80:20, 90:10, 95:5 or the like. Above and below, in those cases, where a chemical structure with a stereogenic center is shown and no specific stereochemistry is indicated, the structure includes all possible stereoisomers as well as mixtures thereof. For example, the present invention includes the stereoisomers [(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]-heptan-2-yl]-formamido}ethyl]boronic acid, [(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1S)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]-boronic acid, [(1S)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]-boronic acid, [(1R)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1R)-2-[(3R) 2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1S)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]-boronic acid, [(1S)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1S)-2-[(3S) 2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]-heptan-2-yl]formamido}ethyl]boronic acid, [(1S)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]-boronic acid, [(1R)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1R)-2-[(3R) 2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1S)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]-boronic acid as well as [(1S)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]-boronic acid. Another exemplary set of stereoisomers, which are included in the present invention is represented by following stereoisomers [(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1S)-2-(1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1R)-2-(1-benzofuran-3-yl)-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1S)-2-(1-benzofuran-3-yl)-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1S)-2-(1-benzofuran-3-yl)-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid, [(1R)-2-(1-benzofuran-3-yl)-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid and [(1S)-2-(1-benzofuran-3-yl)-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid. The different stereoisomers of a given compound are useful for the analytical characterization of a specific sample (e.g. for quality control purposes) via NMR, HPLC, SFC or any other suitable analytical method. Thus, another aspect of the present invention relates to the use of stereoisomers of compounds according to formula (I) in analytical characterization methods. In general, all residues of compounds described herein which occur more than once may be identical or different, i.e. are independent of one another. Above and below, the residues and parameters have the meanings indicated for formula (I), unless expressly indicated otherwise. Accordingly, the invention relates, in particular, to the compounds of formula (I) in which at least one of the said residues has one of the preferred meanings indicated below. Furthermore, all specific embodiments described below shall include derivatives, prodrugs, solvates, tautomers or stereoisomers thereof as well as the physiologically acceptable salts of each of the foregoing, including mixtures thereof in all ratios. The heterobicycles or heterotricycles of formula (xa), (xb), (xc), (xd), (xe), (xf), (xg), (xh) and (xi) can be unsubstituted or mono-, di- or trisubstituted by Hal, NO2, CN, R5a, OR5, CONR5aR5b, NR5aCOR5b, SO2R5a, SOR5a, SO2NR5aR5b, NR5aSO2R5b, NR5aR5b, (CH2)q—R6, COR5aand/or SO2R5a. In case the heterobicycles or heterotricycles of formula (xe), (xf), (xg), (xh) and (xi) are substituted, the one or more substituents can be attached to the one of the bridged rings or one of the fused rings Cy1, Cy2, Cy3, Cy4and Cy5. This includes for example compounds wherein one substitutent is attached to the bridged ring and one substituent is attached to the fused ring Cy1, Cy2, Cy3, Cy4or Cy5. In case one of the fused rings Cy1, Cy2, Cy3, Cy4and Cy5contains one or more CH2groups these groups are understood to be part of the “cyclic CH2groups” of heterobicycles or heterotricycles of formula (xe), (xf), (xg), (xh) and (xi), which may be replaced by CR4aR4b, C═O, O, S, NR5a, SO or SO2. Thus, if 1, 2 or 3 of the cyclic CH2groups of heterobicycles or heterotricycles of formula (xe), (xf), (xg), (xh) and (xi) are be replaced by CR4aR4b, C═O, O, S, NR5a, SO or SO2these cyclic CH2may be part of the bridged ring and/or the fused rings Cy1, Cy2, Cy3, Cy4and Cy5. This includes for example compounds wherein one CH2group of the bridged ring is replaced and one CH2the fused ring Cy1, Cy2, Cy3, Cy4or Cy5is replaced. In case Y is P3, wherein at least one of the two rings of the bicyclic hydrocarbon or heterocycle is an aromatic ring, the other ring may be a saturated, unsaturated or aromatic ring. In specific examples of such embodiments P3and the adjacent group LY are attached to each other via the aromatic ring of P3. In other embodiments P3and the adjacent group LY are attached to each other via the saturated or unsaturated ring of P3. In case P3is a bicyclic heterocyle it preferably contains 1 or 2 heteroatoms selected from N, O and/or S. In case P2is an aromatic monocyclic heterocyle it preferably contains 1 or 2 heteroatoms selected from N, O and/or S. In case Y is P2and P2is phenyl, it is preferably unsubstituted or mono-, di- or trisubstituted by Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R5. Particular preferred are embodiments wherein P2denotes a di- or trisubstituted phenyl. In those embodiments wherein P2denotes a monosubstituted phenyl, the substituent is preferably in the 3-, or the 4-position. In those embodiments wherein P2denotes a disubstituted phenyl, the two substituents are preferably in 2,3-, 2,4-, 2,5- or 3,4-position (most preferably in 2,4- or 3,4-position). And in those embodiments, wherein P2denotes a trisubstituted phenyl, the three substituents are preferably in 2,3,4-position of the aromatic ring. In case P2denotes a monocyclic heterocycle this heterocycle can be saturated, unsaturated or aromatic. In embodiments wherein m denotes 0, LY is absent. In the context of the present invention “C1-C6-alkyl” means an alkyl moiety having 1, 2, 3, 4, 5 or 6 carbon atoms and being straight-chain or branched. The term “C3-C6-cycloalkyl” refers to saturated cyclic hydrocarbon groups having 3, 4, 5 or 6 carbon atoms. The term “unsubstituted” means that the corresponding radical, group or moiety has no substituents other than H; the term “substituted”, which applies to one or more hydrogens that are either explicit or implicit from the structure, means that the corresponding radical, group or moiety has one or more substituents other than H. Where a radical has a plurality of substituents, i.e. at least two, and a selection of various substituents is specified, the substituents are selected independently of one another and do not need to be identical. The term “carbocycle” means a ring system, wherein all ring members are carbon atoms. The term “heterocycle” means a rings system, wherein some of the ring members are heteroatoms such as N, O, or S. The group “NRR′”, is an amino group, wherein R and R′ are for example each independently from one another H or linear or branched C1-C6-alkyl residues (particularly methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl or tert-butyl, pentyl, hexyl). The group “SO” as e.g. included in the SOR5a, is group, wherein S and O are connected via a double bond (S═O). The group “CO” as e.g. included in the COR4a, is group, wherein C and O are connected via a double bond (C═O). The term “alkylene” refers to a divalent alkyl group. An “alkylene group” is a (poly)methylene group (—(CH2)x—). The oxo group (═O) is a substituent, which may which may occur e.g. in saturated cyclic residues or, to the extent possible, in (partially) unsaturated ring such as in particular Het1and Het2. In preferred embodiments the heterocycles Het1and Het2optionally carry one or two oxo groups. As used herein, the term “unsaturated”, means that a moiety has one or more units of unsaturation. As used herein with reference to any rings, cyclic systems, cyclic moieties, and the like, the term “partially unsaturated” refers to a cyclic moiety that includes at least one double or triple bond. The term “partially unsaturated” is intended to encompass cyclic moieties having more the one double or triple bond. In the context of the present invention notations like “O—CH3” and “OCH3” or “CH2CH2” and “—CH2—CH2—” have the same meaning and are used interchangeably. As used herein, in structural formulas arrows or bonds with vertical dotted lines are used to indicate the point of attachment to an adjacent group. For example, the arrow in (xa) shows the point of attachment to the adjacent C═O group. Particular important embodiments of the present invention include compounds of formula (I), whereinR1, R2denote each, independently from one another, H or C1-C4-alkyl or R1and R2form together a residue according to formula (CE); andLY denotes CH2or CH2CH2, wherein 1 to 2H atoms may be replaced by Hal, R3a, OR4a(preferably 1 to 2H atoms may be replaced by F, Cl, CH3, CH2CF3, CH2CHF2, CH2F, CHF2, CF3. OCH3and/or OCF3, and most preferably LY denotes CH2or CH2CH2). Specific embodiments include compounds of formula (I), wherein T1, T2, T3, T4, T5, T6, T7, T8and T9denote O. Other specific embodiments comprise compounds according to formula (I), whereinP1denotes a linear or branched C1-C6-alkyl or C3-C6-cycloalkyl, each, independently from one another, unsubstituted mono-, di- or trisubstituted by Hal, CN, R3a, OR3a, and/or (CH2)q—R6;P2denotes phenyl, pyridyl, pyrrolyl, furanyl, thiophenyl, pyrimidyl, pyranzinyl or pyridazinyl, each, independently from one another, unsubstituted mono-, di- or trisubstituted by Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6; andP3denotes a bicyclic residue of formula (ya), (yb), (yc), (yd), (ye), (yf), (yg), (yh), (yi), (yj), (yk), (yl), (ym), (yn), (yp) or (yp), each, independently from one another, unsubstituted mono-, di- or trisubstituted by Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6: (optional substituents of (ya)-(yp) not shown) whereinEadenotes O, S, N(Alk) or CH═CH;Ebdenotes O, S, N(Alk), CH2, CH2CH2, OCH2, SCH2or N(Alk)CH2. In further embodiments of the invention the residues of a compound of formula (I) are defined as follows:R3a, R3bdenote each, independently from one another, linear or branched C1-C4-alkyl or C3-C6cycloalkyl, wherein 1 to 3H atoms may be replaced by F, Cl and/or and wherein 1 or 2H atoms may be replaced by CN, OH, OCH3, and/or OC2H5. Further embodiments of the invention comprise compounds according to formula (I) wherein Y denotes P2or P3. Other specific embodiments comprise compounds according to formula (I), whereinX is a heterobicycle or heterotricycle of formula of formula (xa1), (xb1), (xc1), (xd1), (xe1), (xf1), (xg1), (xh1) or (xi1), each, independently from one another, unsubstituted or mono-, di- or trisubstituted by Hal, NO2, CN, R5a, OR5a, CONR5aR5b, NR5aCOR5b, SO2R5a, SOR5a, SO2NR5aR5b, NR5aSO2R5b, NR5aR5b, (CH2)q—R6, COR5aand/or SO2R5a, and wherein 1 of the cyclic CH2groups may be replaced CR4aR4b, C═O, O, S, NR5a, SO and/or SO2: (optional substituents of (xa1)-(xi1) not shown) Other specific embodiments comprise compounds according to formula (I), whereinX is a heterobicycle or heterotricycle of formula (xa), (xb), (xc), (xd), (xe), (xf), (xg), (xh) or (xi) each, independently from one another, unsubstituted or mono- or disubstituted by F, Cl, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3. N(CH3)2, CH2N(CH3)2and/or N(C2H5)2, and wherein 1 or 2 of the cyclic CH2groups may be replaced C(CH3)2, C(C2H5)2, C═O, O, S, NH, NR3a, SO and/or SO2(wherein R3ais preferably methyl, ethyl, propyl, isopropyl or cyclopropyl). Important embodiments comprise compounds, of formula (I), wherein X is a heterobicycle or heterotricycle of formula (xa1), (xb1), (xc1), (xd1), (xe1), (xf1), (xg1), (xh1) or (xi1), each, independently from one another, unsubstituted or mono-, disubstituted by F, Cl, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2and/or N(C2H5). Very specific embodiments comprise compounds, of formula (I), wherein X is a heterobicycle or heterotricycle selected from the following group: Other embodiments comprise compounds of formula (I), whereinP3denotes unsubstituted or mono- or disubstituted 1- or 2-naphthyl, wherein the optional substituents are selected from a group consisting of Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6,or(P3is) a residue according to formula (Ra) or (Rb): whereinGa, Gbdenote each, independently from one another, H, Hal, CN, R3a, OR3a, CONHR3a, CONR3bR3a, CONH2, NR3aCOR3b, SO2R3a, SOR3a, NHR3a, N(R3a)2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6;Ka, Kbdenote each, independently from one another, H, Hal, CN, R3a, OR3a, CONHR3a, CONR3bR3a, CONH2, NR3aCOR3b, SO2R3a, SOR3a, NHR3a, N(R3)2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6;Eadenotes O, S, N(Alk) or CH═CH;Ebdenotes O, S, N(Alk), CH2, CH2CH2, O—CH2, S—CH2or N(Alk)CH2. The residue according to formula (Rb) bears a stereogenic center at the carbon atom next to LY; it has been denoted with an asterix (*) in formula (Rb)* below: The residues according to formula (Rb) thus exhibit two different configurations at this stereogenic center, i.e. the (R)-configuration and the (S)-configuration. Hence, the compounds of the present invention may be present either enantiopure or as a racemic (1:1) mixture of the two enantiomers of formula (R)—(Rb) and (S)—(Rb). Compounds of formula (I) which include residues according to formula (Rb) may also be present in a mixture in which one of the enantiomers (R)—(Rb) or (S)—(Rb) is present in an excess over the other one, e.g. 60:40, 70:30, 80:20, 90:10, 95:5 or the like. In a particular embodiment of the present invention the stereoisomer of formula (R)—(Rb) of the compound of formula (I) and the stereoisomer of formula (S)—(Rb) of the compound of formula (I) are present in a ratio of (R)—(Rb) to (S)—(Rb) of at least 90 parts of (R)—(Rb) to not more than 10 parts of (S)—(Rb), preferably of at least 95 (R)—(Rb) to not more than 5 (S)—(Rb), more preferably of at least 99 (R)—(Rb) to not more than 1 (S)—(Rb), even more preferably of at least 99.5 (R)—(Rb) to not more than 0.5 (S)—(Rb). In another particular embodiment of the present invention the stereoisomer of formula (S)—(Rb) of the compound of formula (Rb) and the stereoisomer of formula (R)—(Rb) of the compound of formula (I) are present in a ratio of (S)—(Rb) to (R)—(Rb) of at least 90 (S)—(Rb) to not more than 10 (R)—(Rb), preferably of at least 95 (S)—(Rb) to not more than 5 (R)—(Rb), more preferably of at least 99 (S)—(Rb) to not more than 1 (R)—(Rb), even more preferably of at least 99.5 (S)—(Rb) to not more than 0.5 (R)—(Rb). Particular preferred embodiments of the present invention comprise compounds of formula (I), wherein P3is a residue of formula (S)—(Rb) (which has an (S)-configuration at the carbon attached to LY). Specific embodiments comprise compounds according to formula (I), wherein:P2denotes unsubstituted or mono- or disubstituted 2- or 3-thienyl or unsubstituted or 3-, 4-, 2,3-, 2,4-, 2,5-, 3,4- or 2,3,4-substituted phenyl, wherein in each case the optional substituents are independently selected from a group consisting of Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and (CH2)q—R6;P3denotes unsubstituted or mono- or disubstituted 1- or 2-naphthyl, wherein the optional substituents are selected from a group consisting of Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6,or(P3is) a residue according to formula (Ra) or (Rb) (preferably P3is a residue according to formula (Ra) or (S)—(Rb));Ga, Gbdenote each, independently from one another, H, Hal, CN, R3a, OR3a, CONHR3a, CONR3bR3a, CONH2, NR3aCOR3b, SO2R3a, SOR3a, NHR3a, N(R3a)2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6;Ka, Kbdenote each, independently from one another, H, Hal, CN, R3a, OR3a, CONHR3a, CONR3bR3a, CONH2, NR3aCOR3b, SO2R3a, SOR3a, NHR3a, N(R3a)2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6;Eadenotes O, S, N(Alk) or CH═CH;Ebdenotes O, S, N(Alk), CH2, CH2—CH2, OCH2, SCH2or N(Alk)CH2. Further specific embodiments comprise compounds of formula (I), wherein:P2denotes unsubstituted or mono- or disubstituted 2- or 3-thienyl or unsubstituted or 3-, 4-, 2,3-, 2,4-, 2,5-, 3,4- or 2,3,4-substituted phenyl, wherein in each case the optional substituents are independently selected from a group consisting of Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;P3is a residue according to formula (Re) or (Rb) (preferably P3is a residue according to formula (Re) or (S)—(Rb));Ga, Gbdenote each, independently from one another, H, Hal, CN, R7a, OR78, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;Ka, Kbdenote each, independently from one another, H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;R7a, R7bdenote each, independently from one another, linear or branched C1-C3-alkyl, wherein 1 to 3H atoms may be replaced by Hal; andR8denotes OH or OR7a; andp denotes 1 or 2. Even more specific embodiments comprise compounds according to formula (I), wherein:P2denotes unsubstituted or mono- or disubstituted 2- or 3-thienyl or unsubstituted or 3-, 4-, 2,3-, 2,4-, 2,5-, 3,4- or 2,3,4-substituted phenyl, wherein the optinal substituents are selected from a group consisting of F, Cl, CN, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2or N(C2H5)2;P3is a residue according to formula (Ra) or (Rb) (preferably P3is a residue according to formula (Ra) or (S)—(Rb));Ga, Gbdenote each, independently from one another, H, F, Cl, CN, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2or N(C2H5)2;Ka, Kbdenote each, independently from one another, H, F, Cl, CN, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2or N(C2H5)2. Particular embodiments comprise compounds of formula (I), wherein P3is a residue according to formula (Ra) or (Rb), and wherein Ea, Ebdenote each, independently from one another, O or S. Particular preferred embodiments comprise compounds of formula (I), wherein P3is a residue according to formula (Fa) or (Fb): In such embodiments, the stereogenic center at the carbon atom in position 3 of the dihydrofuranyl residue (Fb) shows preferably an (S)-configuration, i.e. the residue is an (optionally substituted) (3S)-2,3-dihydrobenzofuran-3-yl residue (S)—(Fb)*: (optional substituents not shown). Thus, further very specific embodiments of the invention the present invention comprise compounds according to formula (I), whereinP2denotes unsubstituted or mono- or disubstituted 2- or 3-thienyl or unsubstituted or 3-, 4-, 2,3-, 2,4-, 2,5-, 3,4- or 2,3,4-substituted phenyl, wherein the optinal substituents are selected from a group consisting of Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6;P3denotes a residue according to formula (Fa) or (S)—(Fb);Ga, Gbdenote each, independently from one another, H, Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6; andKa, Kbdenote each, independently from one another, H, Hal, CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6. Other very specific embodiments of the invention the present invention comprise compounds according to formula (I), wherein:P2denotes unsubstituted or mono- or disubstituted 2- or 3-thienyl or unsubstituted or 3-, 4-, 2,3-, 2,4-, 2,5-, 3,4- or 2,3,4-substituted phenyl, wherein the optinal substituents are selected from a group consisting of H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;P3denotes a residue according to formula (Fa) or (S)—(Fb); Ga, Gbdenote each, independently from one another, H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;Ka, Kbdenote each, independently from one another, H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;R7a, R7bdenote each, independently from one another, linear or branched C1-C3-alkyl, wherein 1 to 3H atoms may be replaced by Hal; andR8denotes OH or OR7a; andp denotes 1 or 2. Other very specific embodiments of the present invention comprise compounds according to formula (I), wherein:P2denotes unsubstituted or mono- or disubstituted 2- or 3-thienyl or unsubstituted or 3-, 4-, 2,3-, 2,4-, 2,5-, 3,4- or 2,3,4-substituted phenyl, wherein the optional substituents are selected from a group consisting of F, Cl, CN, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2or N(C2H5)2;P3denotes a residue according to formula (P) or (S)—(Fb),Ga, Gbdenote each, independently from one another, H, F, Cl, CN, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2or N(C2H5)2; andKa, Kbdenote each, independently from one another, H, F, Cl, CN, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2or N(C2H5)2. Particular embodiments of the present invention comprise compounds according to formula (I), wherein Y denotes P2or P3, preferrably Y denotes P3. Specific embodiments of the present invention comprise compounds according to formula (I) whereinLY denotes CH2or CH2CH2, wherein 1 to 2H atoms may be replaced by Hal, R7a, OH and/or OR7a, and/or wherein one CH2group may be replaced by O or S;X is a heterobicycle or heterotricycle of formula (xa), (xb), (xc), (xd), (xe), (xf), (xg), (xh) or (xi) each, independently from one another, unsubstituted or mono- or disubstituted by F, Cl, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2and/or N(C2H5)2, and wherein 1 of the cyclic CH2groups may be replaced C(CH3)2, C(C2H5)2, C═O, O, S, NCH3, SO or SO2;Y denotes P2or P3(preferably P3);P2denotes unsubstituted or mono- or disubstituted 2- or 3-thienyl or unsubstituted or 3-, 4-, 2,3-, 2,4-, 2,5-, 3,4- or 2,3,4-substituted phenyl, wherein the optinal substituents are selected from a group consisting of H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCORT, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8; P3denotes a residue according to formula (Fa) or (S)—(Fb); Ga, Gbdenote each, independently from one another, H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;Ka, Kbdenote each, independently from one another, H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R792, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;R7a, R7bdenote each, independently from one another, linear or branched C1-C3-alkyl, wherein 1 to 3H atoms may be replaced by Hal; andR8denotes OH or OR7a; andp denotes 1 or 2.Cy1, Cy2, Cy3, Cy4, Cy5denote each, independently from one another, Ar1or Het1;R1, R2denote each, independently from one another, H or C1-C6-alkyl, or R1and R2form together a residue according to formula (CE);T1, T2, T3, T4, T8, T6, T7, T8and T9denote each O; Hal denotes F, Cl or Br. Other very specific embodiments of the present invention comprise compounds according to formula (R)-(I)-(S)-(P) or (R)-(I)-(P): whereinGa, Gbdenote each, independently from one another, H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)p—N(R7a)2and/or (CH2)p—R8;Ka, Kbdenote each, independently from one another, H, Hal, CN, R7a, OR7a, CONHR7a, CONR7bR7a, CONH2, NR7aCOR7b, SO2R7a, SOR7a, NHR7a, N(R7a)2, (CH2)p—SR7a, (CH2)N—N(R7a)2and/or (CH2)p—R8;X is a heterobicycle or heterotricycle of formula ((xa1), (xb1), (xc1), (xd1), (xe1), (xf1), (xg1), (xh1) or (xi1) each, independently from one another, unsubstituted or mono- or disubstituted by F, Cl, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2and/or N(C2H5)2, wherein 1 of the cyclic CH2groups may be replaced C(CH3)2, C(C2H5)2, C═O, O, S, NCH3, SO or SO2;R1, R2denote H or C1-C4-alkyl or R1and R2form together a residue according to formula (CE);R7a, R7bdenote each, independently from one another, linear or branched C1-C3-alkyl, wherein 1 to 3H atoms may be replaced by Hal;R8denotes OH or OR7a; andp denotes 1 or 2. In general, the residues included in the compounds according to formula (I) as described above may have following meaning: LY denotes preferably —CH2— or —CH2—CH2— wherein 1 to 4H atoms may be replaced by Hal and/or 1H atom may be replaced by Hal, R3aand/or OR4a, and/or wherein 1 or 2 non-adjacent CH2groups may be replaced by O, SO and/or SO2. Most preferably LY denotes —CH2— or —CH2—CH2—, wherein 1 to 4H atom may be replaced by F or Cl and/or 1 or 2H atoms may be replaced by OH, methy, ethyl, isopropyl, CF3, CF2CF3, OCH3, OCH2CH3, OCH2CH2OH and/or CH2OCH3and/or wherein 1 CH2group of LY may be replaced by O. R1, R2denote preferably each, independently from one another H or methyl, ethyl, n-propyl or isopropyl or R1and R2form together a residue according to formula (CE) as described above. Most preferably R1, R2denote H, methyl or ethyl and particular preferably R1, R2denote H. In embodiments wherein R3aor R3brepresent a linear or branched C1-C6alkyl, they denote preferably each, independently from one another, linear or branched methyl, ethyl, n-propyl or isopropyl, wherein 1 to 5H atoms may be replaced by F, Cl, CN, OH and OAlk, wherein Alk is preferably methyl or ethyl. Most preferably R3aand R3bdenote each, independently from one another, methyl, ethyl, n-propyl or isopropyl, wherein 1, 2 or 3H atoms are replaced by F, Cl, OH, OCH3, OC2H5or OCH(CH3)2. In embodiments wherein R3aor R3brepresent independently from one another a cyclic alkyl group (cycloalkyl), they preferably denote independently from each other cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, each unsubstituted or mono-, di- or trisubstituted by Hal (preferably F or Cl), methyl, ethyl, n-propyl, OH, CN, OCH3or OC2H5. R4aand R4bdenote preferably each, independently from one another, preferably H, methyl, furthermore ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl or pentyl, wherein 1, 2 or 3H atoms are replaced by F, Cl, OH, OCH3, OC2H5or OCH(CH3)2or R4aand R4bfrom together a C3-C6alkylene group. Y can denote phenyl, 1- or 2-naphthyl, 4- or 5-indanyl, 1-, 2-, 3-, 4-, 5-, 6- or 7-indolyl, 1-, 2-, 4-, 5- or 6-azulenyl, 1- or 2-tetrahydronaphthalin 5- or 6-yl, 2- or 3-furyl, 2-, 3-, 4-, 5-, 6- or 7-benzofuryl, 2,3-dihydrobenzofuran-2- or 3-yl, 2- or 3-thienyl, 2- or 3-benzothienyl, 2-, 3-, 4-, 5-, 6- or 7-benzothiophenyl, methylenedioxyphenyl, benzodioxan-6- or 7-yl or 3,4-dihydro-1,5-benzodioxepin-6- or -7-yl, each independently from one another, unsubstituted, mono-, disubstituted or trisubstituted by Hal (preferably F or Cl), CN, R3a, OH, OR3a, CONR4aR4b, NR3aCOR3b, SO2R3a, SOR3a, NR4aR4b, Ar2, Het2, (CH2)q—SR3a, (CH2)q—N(R4a)2and/or (CH2)q—R6. In particular Y can denote phenyl, 1- or 2-naphthyl 2-, 3-, 4-, 5-, 6- or 7-benzofuryl 2,3-dihydrobenzofuran-2- or 3-yl, 2- or 3-thienyl, 2- or 3-benzothienyl or benzodioxan-6- or 7-yl, each independently from one another, unsubstituted, mono-, disubstituted or trisubstituted by F, Cl, CN, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3. N(CH3)2, CH2N(CH3)2or N(C2H5)2. In case Y denotes a disubstituted phenyl the substituents are preferably in 2,4-, 2,5- or 3,4-position, most preferably in 2,4- or 3,4-position. In case Y denotes a trisubstituted phenyl the substituents are preferably in 2,3,4-position. In particular Y can denote o-, m- or p-tolyl, o-, m- or p-ethylphenyl, o-, m- or p-propylphenyl, o-, m- or p-isopropylphenyl, o-, m- or p-tert-butylphenyl, o-, m- or p-acetamidophenyl, o-, m- or p-methoxyphenyl, o-, m- or p-ethoxyphenyl, o-, m- or p-fluorophenyl, o-, m- or p-bromophenyl, o-, m- or p-chlorophenyl, o-, m- or p-trifluormethyl-phenyl, o-, m- or p-trichloromethyl-phenyl, o-, m- or p-(methylsulfonyl)phenyl, o-, m- or p-phenoxyphenyl, o-, m- or p-methoxymethyl-phenyl further preferably 2,4-, 2,5-, 2,6- or 3,4-dimethylphenyl, 2,4-, 2,5- or 3,4-difluorophenyl, 2,4-, 2,5- or 3,4-dichloro-phenyl, 2,4-, 2,5- or 3,4-dibromophenyl, 2,5- or 3,4-dimethoxyphenyl, 2,3,4-, 2,3,5-, 2,3,6-, 2,4,6- or 3,4,5-trichlorophenyl, 2,3,4-, 2,3,5-, 2,3,6-, 2,4,6- or 3,4,5-trifluorophenyl, 2,3,4-, 2,3,5-, 2,3,6-, 2,4,6- or 3,4,5-trimethylphenyl, 2,3,4-, 2,3,5-, 2,3,6-, 2,4,6- or 3,4,5-tris trifluormethyl-phenyl, 2,3,4-, 2,3,5-, 2,3,6-, 2,4,6- or 3,4,5-tristrichlormethyl-phenyl, 2,3,4-, 2,3,5-, 2,3,6-, 2,4,6- or 3,4,5-trimethoxymethyl-phenyl, 2,4,6-trimethoxyphenyl, p-iodophenyl, 2-fluoro-3-chlorophenyl, 2-fluoro-3-bromophenyl, 2,3-difluoro-4-bromophenyl, 3-bromo-3-methoxyphenyl, 2-chloro-3-methoxyphenyl, 2-fluoro-3-methoxyphenyl, 2-chloro-3-acetamidophenyl, 2-fluoro-3-methoxyphenyl, 2-chloro-3-acetamidophenyl, 2,3-dimethyl-4-chlorophenyl, 2,3-dimethyl-4-fluorophenyl. Y can also denote 1- or 2-naphthyl, 4- or 5-indanyl, 1-, 2-, 4-, 5- or 6-azulenyl, 1- or 2-tetrahydronaphthalin 5- or 6-yl, 2- or 3-furyl, 2-, 3-, 4-, 5-, 6- or 7-benzofuryl, 2-, 3-, 4-, 5-, 6- or 7-benzothiophenyl, methylenedioxyphenyl, benzodioxan-6- or 7-yl or 3,4-dihydro-1,5-benzodioxepin-6- or -7-yl. Particular preferred substitutents of Y are selected from a group comprising, Cl, CN, CH3, C2H5, CF3, OCH3, OC2H5, COCF3, SCH3, SC2H5, CH2OCH3, N(CH3)2, CH2N(CH3)2or N(C2H5)2. Ar2denotes preferably phenyl, which is unsubstituted or mono- or disubstituted by Hal, CN, R3a, OR3a, CONHR3a, NH2, NHR3aand/or N(R3a)2. Thus, Ar2preferably denotes e.g. phenyl, o-, m- or p-tolyl, o-, m- or p-ethylphenyl, o-, m- or p-propylphenyl, o-, m- or p-isopropylphenyl, o-, m- or p-tert-butylphenyl, o-, m- or p-hydroxyphenyl, o-, m- or p-nitrophenyl, o-, m- or p-aminophenyl, o-, m- or p-(N-methylamino)phenyl, o-, m- or p-(N-methyl-aminocarbonyl)phenyl, o-, m- or p-acetamidophenyl, o-, m- or p-methoxyphenyl, o-, m- or p-ethoxyphenyl, o-, m- or p-(N,N-dimethylamino)phenyl, o-, m- or p-(N-ethylamino)phenyl, o-, m- or p-(N,N-diethylamino)phenyl, o-, m- or p-fluorophenyl, o-, m- or p-bromophenyl, o-, m- or p-chlorophenyl, o-, m- or p-cyanophenyl. Het2denotes preferably a saturated, unsaturated or aromatic 5- or 6-membered heterocycle having 1 to 4 N, O and/or S atoms, which is unsubstituted or mono- or disubstituted by Hal, CN, R3a, OR3a, CONHR3a, NH2, NHR3aand/or N(R3a)2. Thus, Het2may e.g. denote 2- or 3-furyl, 2- or 3-thienyl, 1-, 2- or 3-pyrrolyl, 1-, 2-, 4- or 5-imidazolyl, 1-, 3-, 4- or 5-pyrazolyl, 2-, 4- or 5-oxazolyl, 3-, 4- or 5-isoxazolyl, 2-, 4- or 5-thiazolyl, 3-, 4- or 5-isothiazolyl, 2-, 3- or 4-pyridyl, 2-, 4-, 5- or 6-pyrimidinyl, imidazolyl, morpholinyl or piperazinyl. Alk denotes preferably methy, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl or tert-butyl, pentyl or hexyl, most preferably methy, ethyl, propyl or isopropyl, most preferably methy, ethyl, n-propyl or isopropyl. Hal denotes preferably F, Cl or Br, most preferably F or C1. m denotes preferably 0, 1 or 2, more 1 or 2 and most preferably 1. q denotes preferably 0, 1, 2, 3 or 4 and even more preferably 0, 1 or 2. Particular embodiments of the present invention comprise the compounds selected from the group consisting of:[(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}-2-(thiophen-3-yl)ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1R,8S)-11-oxatricyclo[6.2.1.02,7]undeca-2(7),3,5-trien-1-yl]formamido}ethyl]boronic acid;[(1S)-2-(1-benzofuran-3-yl)-1-{[(1R,8S)-11-oxatricyclo[6.2.1.02,7]undeca-2(7),3,5-trien-1-yl]formamido}ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,8R)-11-oxatricyclo[6.2.1.02,7]undeca-2(7),3,5-trien-1-yl]formamido}ethyl]boronic acid;[(1S)-2-(1-benzofuran-3-yl)-1-{[(1S,8R)-11-oxatricyclo[6.2.1.02,7]undeca-2(7),3,5-trien-1-yl]formamido}ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-(1-benzofuran-3-yl)-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-(7-chloro-1-benzofuran-3-yl)-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-(7-chloro-1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3R)-7-methyl-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3S)-7-methyl-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,8S)-11-oxatricyclo[6.2.1.02,7]undeca-2(7),3,5-trien-1-yl]formamido}ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,6S,7R)-3-cyclopropyl-4-oxo-10-oxa-3-azatricyclo[5.2.1.01,5]dec-8-en-6-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,8R)-11-oxatricyclo[6.2.1.02,7]undeca-2(7),3,5-trien-1-yl]formamido}ethyl]boronic acid;[(1R)-2-(7-methyl-1-benzofuran-3-yl)-1-{[(1R,8S)-11-oxatricyclo[6.2.1.02,7]undeca-2,4,6-trien-1-yl]formamido}ethyl]boronic acid;[(1R)-2-(7-methyl-1-benzofuran-3-yl)-1-{[(1S,8R)-11-oxatricyclo[6.2.1.02,7]undeca-2,4,6-trien-1-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,8R)-8-methyl-11-oxatricyclo[6.2.1.02,7]undeca-2,4,6-trien-1-yl]formamido}ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1R,8S)-11-oxatricyclo[6.2.1.02,7]undeca-2(7),3,5-trien-9-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,8S)-8-methyl-11-oxatricyclo[6.2.1.02,7]undeca-2,4,6-trien-1-yl]formamido}ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,8R)-11-oxatricyclo[6.2.1.02,7]undeca-2(7),3,5-trien-9-yl]formamido}ethyl]boronic acid;[(1R)-2-(2,4-dimethylphenyl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-cyclohexyl-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}-3-phenylpropyl]boronic acid;[(1R)-3-methyl-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}butyl]boronic acid;[(1S)-2-(1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-(1-benzofuran-3-yl)-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-(1-benzofuran-3-yl)-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2S,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2R,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1R)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid;[(1S)-2-[(3R)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1R,2S,4S)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic acid; and prodrugs, solvates, tautomers, oligomers, adducts or stereoisomers thereof as well as the pharmaceutically acceptable salts of each of the foregoing, including mixtures thereof in all ratios. The term solvates of the compounds is taken to mean adductions of inert solvent molecules onto the compounds which form owing to their mutual attractive force. Solvates are, for example, mono- or dihydrates or alkoxides. It is understood, that the invention also relates to the solvates of the salts. The term pharmaceutically acceptable derivatives is taken to mean, for example, the salts of the compounds according to the invention and also so-called prodrug compounds. As used herein and unless otherwise indicated, the term “prodrug” means a derivative of a compound of formula (I) that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound, particularly a compound of formula (I). Examples of prodrugs include, but are not limited to, derivatives and metabolites of a compound of formula (I) that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. In certain embodiments, prodrugs of compounds with carboxyl functional groups are the lower alkyl esters of the carboxylic acid. The carboxylate esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule. Prodrugs can typically be prepared using well-known methods, such as those described by Burger's Medicinal Chemistry and Drug Discovery 6th ed. (Donald J. Abraham ed., 2001, Wiley) and Design and Application of Prodrugs (H. Bundgaard ed., 1985, Harwood Academic Publishers Gmfh). The expression “effective amount” denotes the amount of a medicament or of a pharmaceutical active ingredient which causes in a tissue, system, animal or human a biological or medical response which is sought or desired, for example, by a researcher or physician. In addition, the expression “therapeutically effective amount” denotes an amount which, compared with a corresponding subject who has not received this amount, has the following consequence: improved treatment, healing, prevention or elimination of a disease, syndrome, condition, complaint, disorder or side-effects or also the reduction in the advance of a disease, complaint or disorder. The expression “therapeutically effective amount” also encompasses the amounts which are effective for increasing normal physiological function. The invention also relates to the use of mixtures of the compounds of the formula (I), for example mixtures of two diastereomers, for example in the ratio 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:100 or 1:1000. “Tautomers” refers to isomeric forms of a compound that are in equilibrium with each other. The concentrations of the isomeric forms will depend on the environment the compound is found in and may be different depending upon, for example, whether the compound is a solid or is in an organic or aqueous solution. The invention further comprises a process for the preparation of a compound of the formula (I) as described above and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, characterized in that a compound of formula (III) is coupled with a compound of formula (VI) wherein all residues of formula (III) and formula (IV) are as defined above and wherein the obtained compound of formula (Ib) may subsequently converted into a compound of formula (Ia), by treatment with HCl, HBr, HI and/or TFA, in the presence or absence of an excess of a small molecular weight boronic acid The following abbreviations refer to the abbreviations used below: AcOH (acetic acid), ACN (acetonitrile), BINAP (2,2′-bis(disphenylphosphino)-1,1′-binaphthalene), dba (dibenzylidene acetone), tBu (tert-Butyl), tBuOK (potassium tert-butoxide), CDI (1,1′-Carbonyldiimidazole), DBU (1,8-dizabicyclo[5.4.0]undec-7-ene), DCC (dicyclohexylcarbodiimide), DCM (dichloromethane), DIAD (diisobutylazodicarboxylate), DIC (diisopropilcarbodiimide), DIEA (diisopropyl ethylamine), DMA (dimethyl acetamide), DMAP (4-dimethylaminopyridine), DMSO (dimethyl sulfoxide), DMF (N,N-dimethylformamide), EDC.HCl (1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride), EtOAc or EE (ethyl acetate), EtOH (ethanol), g (gram), cHex (cyclohexane), HATU (dimethylamino-([1,2,3]triazolo[4,5-b]pyridin-3-yloxy)-methylene]-dimethyl-ammonium hexafluorophosphate), HOBt (N-hydroxybenzotriazole), HPLC (high performance liquid chromatography), hr (hour), MHz (Megahertz), McOH (methanol), min (minute), mL (milliliter), mmol (millimole), mM (millimolar), mp (melting point), MS (mass spectrometry), MW (microwave), NMM (N-methyl morpholine), NMR (Nuclear Magnetic Resonance), NBS (N-bromo succinimide), PBS (phosphate buffered saline), PMB (para-methoxybenzyl), PyBOP (benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate), rt (room temperature), TBAF (tetra-butylammonium fluoride), TBTU (N,N,N′,N′-tetramethyl-O-(benzotriazol-1-yl)uronium tetrafluoroborate), T3P (propane phosphonic acid anhydride), TEA (triethyl amine), TFA (trifluoroacetic acid), THF (tetrahydrofuran), PetEther (petroleum ether), TBME (tert-butyl methyl ether), TLC (thin layer chromatography), TMS (trimethylsilyl), TMSI (trimethylsilyl iodide), UV (ultraviolet). Generally, compounds of formula (I), wherein all residues are defined as above, can be obtained from a compound of formula (III) as outlined in Scheme 1. The first step consists in the reaction of a compound of formula (III), wherein X is defined as above, with a compound of formula (IV), wherein R1, R2, LY and Y are defined as above. The reaction is performed using conditions and methods well known to those skilled in the art for the preparation of amides from a carboxylic acid with standard coupling agents, such as but not limited to HATU, TBTU, polymer-supported 1-alkyl-2-chloropyridinium salt (polymer-supported Mukaiyama's reagent), 1-methyl-2-chloropyridinium iodide (Mukaiyama's reagent), a carbodiimide (such as DCC, DIC, EDC) and HOBt, PyBOP® and other such reagents well known to those skilled in the art, preferably TBTU, in the presence or absence of bases such as TEA, DIEA, NMM, polymer-supported morpholine, preferably DIEA, in a suitable solvent such as DCM, THF or DMF, at a temperature between −10° C. to 50° C., preferably at 0° C., for a few hours, e.g. one hour to 24 h. Alternatively, the compounds of formula (III) could be converted to carboxylic acid derivatives such as acyl halides or anhydrides, by methods well known to those skilled in the art, such as but not limited to treatment with SOCl2, POCl3, PCl5, (COCl)2, in the presence or absence of catalytic amounts of DMF, in the presence or absence of a suitable solvent such as toluene, DCM, THF, at a temperature rising from 20° C. to 100° C., preferably at 50° C., for a few hours, e.g. one hour to 24 h. Conversion of the carboxylic acid derivatives to compounds of formula (I), can be achieved using conditions and methods well known to those skilled in the art for the preparation of amides from a carboxylic acid derivative (e.g. acyl chloride) with alkyl amines, in the presence of bases such as TEA, DIEA, NMM in a suitable solvent such as DCM, THF or DMF, at a temperature rising from 20° C. to 100° C., preferably at 50° C., for a few hours, e.g. one hour to 24 h. In the process described above the reaction between the compound of formula (III) and the compound of formula (IV) is preferably performed in the presence of a coupling agent selected from HATU, TBTU, polymer-supported 1-alkyl-2-chloropyridinium salt (polymer-supported Mukaiyama's reagent), 1-methyl-2-chloropyridinium iodide (Mukaiyama's reagent), a carbodiimide. Compounds of formula (Ia), wherein X, LY and Y are defined as above and wherein R1and R2are H, can be prepared starting from compounds of formula (Ib), using methods well known to those skilled in the art for the hydrolysis of boronic esters, such as but not limited to treatment with HCl, HBr, HI, TFA, in the presence or absence of an excess of a small molecular weight boronic acid, such as but not limited to iBuB(OH)2(Scheme 2). Compounds of formula (III) or (IV) are either commercially available or can be prepared by methods well known to those skilled in the art. In general, compounds of formula (IV) are for example accessible by the following scheme 3a: The synthesis of compounds of formula (IV) is further described in WO 2016/050356, WO 2016/050355, WO 2016/050359, and WO 2016/050358. Compounds of formula (III) are for example accessible by the routes described in Scheme 4, 5 and 6: By similar approaches also substituted 7-oxa-bicyclo[2.2.1]heptane-2-carboxylic acids can be synthesised. By similar approaches also substituted 11-oxatricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-9-carboxylic acids can be synthesised. By similar approaches starting from substituted furane-2-carboxylic acids, substituted 11-oxatricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic acids can be synthesised. Also substituted anthranilic acids can be used to synthesise corresponding compounds which carry additional substituents in the aromatic moiety. If the above set of general synthetic methods is not applicable to obtain compounds according to formula (I) and/or necessary intermediates for the synthesis of compounds of formula (I), suitable methods of preparation known by a person skilled in the art should be used. In general, the synthesis pathways for any individual compounds of formula (I) will depend on the specific substitutents of each molecule and upon the ready availability of Intermediates necessary; again such factors being appreciated by those of ordinary skill in the art. For all the protection and de-protection methods, see Philip J. Kocienski, in “Protecting Groups”, Georg Thieme Verlag Stuttgart, New York, 1994 and, Theodora W. Greene and Peter G. M. Wuts in “Protective Groups in Organic Synthesis”, Wiley Interscience, 3rd Edition 1999. Compounds of this invention can be isolated in association with solvent molecules by crystallization from evaporation of an appropriate solvent. The pharmaceutically acceptable acid addition salts of the compounds of formula (I), which contain a basic center, may be prepared in a conventional manner. For example, a solution of the free base may be treated with a suitable acid, either neat or in a suitable solution, and the resulting salt isolated either by filtration or by evaporation under vacuum of the reaction solvent. Pharmaceutically acceptable base addition salts may be obtained in an analogous manner by treating a solution of compounds of formula (I), which contain an acid center, with a suitable base. Both types of salts may be formed or interconverted using ion-exchange resin techniques. Depending on the conditions used, the reaction times are generally between a few minutes and 14 days, and the reaction temperature is between about −30° C. and 140° C., normally between −10° C. and 90° C., in particular between about 0° C. and about 70° C. Compounds of the formula (I) can furthermore be obtained by liberating compounds of the formula (I) from one of their functional derivatives by treatment with a solvolysing or hydrogenolysing agent. Preferred starting materials for the solvolysis or hydrogenolysis are those which conform to the formula (I), but contain corresponding protected amino and/or hydroxyl groups instead of one or more free amino and/or hydroxyl groups, preferably those which carry an amino-protecting group instead of an H atom bound to an N atom, in particular those which carry an R′—N group, in which R′ denotes an amino-protecting group, instead of an HN group, and/or those which carry a hydroxyl-protecting group instead of the H atom of a hydroxyl group, for example those which conform to the formula (I), but carry a —COOR″ group, in which R″ denotes a hydroxyl-protecting group, instead of a —COOH group. It is also possible for a plurality of—identical or different—protected amino and/or hydroxyl groups to be present in the molecule of the starting material. If the protecting groups present are different from one another, they can in many cases be cleaved off selectively. The term “amino-protecting group” is known in general terms and relates to groups which are suitable for protecting (blocking) an amino group against chemical reactions, but which are easy to remove after the desired chemical reaction has been carried out elsewhere in the molecule. Typical of such groups are, in particular, unsubstituted or substituted acyl, aryl, aralkoxymethyl or aralkyl groups. Since the amino-protecting groups are removed after the desired reaction (or reaction sequence), their type and size are furthermore not crucial; however, preference is given to those having 1-20, in particular 1-8, carbon atoms. The term “acyl group” is to be understood in the broadest sense in connection with the present process. It includes acyl groups derived from aliphatic, araliphatic, aromatic or heterocyclic carboxylic acids or sulfonic acids, and, in particular, alkoxy-carbonyl, aryloxycarbonyl and especially aralkoxycarbonyl groups. Examples of such acyl groups are alkanoyl, such as acetyl, propionyl and butyryl; aralkanoyl, such as phenylacetyl; aroyl, such as benzoyl and tolyl; aryloxyalkanoyl, such as POA; alkoxycarbonyl, such as methoxy-carbonyl, ethoxycarbonyl, 2,2,2-trichloroethoxycarbonyl, BOC (tert-butoxy-carbonyl) and 2-iodoethoxycarbonyl; aralkoxycarbonyl, such as CBZ (“carbo-benz-oxy”), 4-methoxybenzyloxycarbonyl and FMOC; and aryl-sulfonyl, such as Mtr. Preferred amino-protecting groups are BOC and Mtr, furthermore CBZ, Fmoc, benzyl and acetyl. The term “hydroxyl-protecting group” is likewise known in general terms and relates to groups which are suitable for protecting a hydroxyl group against chemical reactions, but are easy to remove after the desired chemical reaction has been carried out elsewhere in the molecule. Typical of such groups are the above-mentioned unsubstituted or substituted aryl, aralkyl or acyl groups, furthermore also alkyl groups. The nature and size of the hydroxyl-protecting groups are not crucial since they are removed again after the desired chemical reaction or reaction sequence; preference is given to groups having 1-20, in particular 1-10, carbon atoms. Examples of hydroxyl-protecting groups are, inter alia, benzyl, 4-methoxybenzyl, p-nitro-benzoyl, p-toluenesulfonyl, tert-butyl and acetyl, where benzyl and tert-butyl are particularly preferred. The term “solvates of the compounds” is taken to mean adductions of inert solvent molecules onto the compounds which form owing to their mutual attractive force. Solvates are, for example, mono- or dihydrates or alcoholates. The compounds of the formula (I) are liberated from their functional derivatives—depending on the protecting group used—for example using strong acids, advantageously using TFA or perchloric acid, but also using other strong inorganic acids, such as hydrochloric acid or sulfuric acid, strong organic carboxylic acids, such as trichloroacetic acid, or sulfonic acids, such as benzene- or p-toluenesulfonic acid. The presence of an additional inert solvent is possible, but is not always necessary. Suitable inert solvents are preferably organic, for example carboxylic acids, such as acetic acid, ethers, such as THE or dioxane, amides, such as DMF, halogenated hydrocarbons, such as DCM, furthermore also alcohols, such as methanol, ethanol or isopropanol, and water. Mixtures of the above-mentioned solvents are furthermore suitable. TFA is preferably used in excess without addition of a further solvent, and perchloric acid is preferably used in the form of a mixture of acetic acid and 70% perchloric acid in the ratio 9:1. The reaction temperatures for the cleavage are advantageously between about 0 and about 50° C., preferably between 15 and 30° C. (rt). The BOC, OBut and Mtr groups can, for example, preferably be cleaved off using TFA in DCM or using approximately 3 to 5N HCl in dioxane at 15-30° C., and the FMOC group can be cleaved off using an approximately 5 to 50% solution of dimethylamine, diethylamine or piperidine in DMF at 15-30° C. Protecting groups which can be removed hydrogenolytically (for example CBZ, benzyl or the liberation of the amidino group from the oxadiazole derivative thereof) can be cleaved off, for example, by treatment with hydrogen in the presence of a catalyst (for example a noble-metal catalyst, such as palladium, advantageously on a support, such as carbon). Suitable solvents here are those indicated above, in particular, for example, alcohols, such as methanol or ethanol, or amides, such as DMF. The hydrogenolysis is generally carried out at temperatures between about 0 and 100° C. and pressures between about 1 and 200 bar, preferably at 20-30° C. and 1-10 bar. Hydrogenolysis of the CBZ group succeeds well, for example, on 5 to 10% Pd/C in methanol or using ammonium formate (instead of hydrogen) on Pd/C in methanol/DMF at 20-30° C. Examples of suitable inert solvents are hydrocarbons, such as hexane, petroleum ether, benzene, toluene or xylene; chlorinated hydrocarbons, such as trichloroethylene, 1,2-dichloroethane, tetrachloromethane, tri-fluoro-methylbenzene, chloroform or DCM; alcohols, such as methanol, ethanol, isopropanol, n-propanol, n-butanol or tert-butanol; ethers, such as diethyl ether, diisopropyl ether, tetrahydrofurane (THF) or dioxane; glycol ethers, such as ethylene glycol monomethyl or monoethyl ether or ethylene glycol dimethyl ether (diglyme); ketones, such as acetone or butanone; amides, such as acetamide, dimethylacetamide, N-methylpyrrolidone (NMP) or dimethylformamide (DMF); nitriles, such as acetonitrile; sulfoxides, such as dimethyl sulfoxide (DMSO); carbon disulfide; carboxylic acids, such as formic acid or acetic acid; nitro compounds, such as nitromethane or nitrobenzene; esters, such as EtOAc, or mixtures of the said solvents. Esters can be saponified, for example, using LiOH, NaOH or KOH in water, water/THF, water/THF/ethanol or water/dioxane, at temperatures between 0 and 100° C. Furthermore, ester can be hydrolysed, for example, using acetic acid, TFA or HCl. Free amino groups can furthermore be acylated in a conventional manner using an acyl chloride or anhydride or alkylated using an unsubstituted or substituted alkyl halide or reacted with CH3—C(═NH)—OEt, advantageously in an inert solvent, such as DCM or THE and/or in the presence of a base, such as triethylamine or pyridine, at temperatures between −60° C. and +30° C. Throughout the specification, the term leaving group preferably denotes Cl Br, I or a reactively modified OH group, such as, for example, an activated ester, an imidazolide or alkylsulfonyloxy having 1 to 6 carbon atoms (preferably methylsulfonyloxy or trifluoromethylsulfonyloxy) or arylsulfonyloxy having 6 to 10 carbon atoms (preferably phenyl- or p tolylsulfonyloxy). Radicals of this type for activation of the carboxyl group in typical acylation reactions are described in the literature (for example in the standard works, such as Houben-Weyl, Methoden der organischen Chemie [Methods of Organic Chemistry], Georg-Thieme-Verlag, Stuttgart). Activated esters are advantageously formed in situ, for example through addition of HOBt or N-hydroxysuccinimide. Pharmaceutical Salts and Other Forms The said compounds of the formula (I) can be used in their final non-salt form. On the other hand, the present invention also relates to the use of these compounds in the form of their pharmaceutically acceptable salts, which can be derived from various organic and inorganic acids and bases by procedures known in the art. Pharmaceutically acceptable salt forms of the compounds of the formula (I) are for the most part prepared by conventional methods. If the compound of the formula (I) contains an acidic center, such as a carboxyl group, one of its suitable salts can be formed by reacting the compound with a suitable base to give the corresponding base-addition salt. Such bases are, for example, alkali metal hydroxides, including potassium hydroxide and sodium hydroxide; alkaline earth metal hydroxides, such as magnesium hydroxide and calcium hydroxide; and various organic bases, such as piperidine, diethanolamine and N-methyl-glucamine (meglumine), benzathine, choline, diethanolamine, ethylenediamine, benethamine, diethylamine, piperazine, lysine, L-arginine, ammonia, triethanolamine, betaine, ethanolamine, morpholine and tromethamine. In the case of certain compounds of the formula I, which contain a basic center, acid-addition salts can be formed by treating these compounds with pharmaceutically acceptable organic and inorganic acids, for example hydrogen halides, such as hydrogen chloride or hydrogen bromide, other mineral acids and corresponding salts thereof, such as sulfate, nitrate or phosphate and the like, and alkyl- and monoaryl-sulfonates, such as methanesulfonate, ethanesulfonate, toluenesulfonate and benzene-sulfonate, and other organic acids and corresponding salts thereof, such as carbonate, acetate, trifluoro-acetate, tartrate, maleate, succinate, citrate, benzoate, salicylate, ascorbate and the like. Accordingly, pharmaceutically acceptable acid-addition salts of the compounds of the formula (I) include the following: acetate, adipate, alginate, aspartate, benzoate, benzene-sulfonate (besylate), bisulfate, bisulfite, bromide, camphorate, camphor-sulfonate, caprate, caprylate, chloride, chlorobenzoate, citrate, cyclamate, cinnamate, digluconate, dihydrogen-phosphate, dinitrobenzoate, dodecyl-sulfate, ethanesulfonate, formate, glycolate, fumarate, galacterate (from mucic acid), galacturonate, glucoheptanoate, gluco-nate, glutamate, glycerophosphate, hemisuccinate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy-ethane-sulfonate, iodide, isethionate, isobutyrate, lactate, lactobionate, malate, maleate, malonate, mandelate, metaphosphate, methanesulfonate, methyl benzoate, mono-hydrogen-phosphate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, oleate, palmoate, pectinate, persulfate, phenylacetate, 3-phenylpropionate, phosphate, phosphonate, phthalate, but this does not represent a restriction. Both types of salts may be formed or interconverted preferably using ion-exchange resin techniques. Furthermore, the base salts of the compounds of the formula (I) include aluminium, ammonium, calcium, copper, iron (III), iron(II), lithium, magnesium, manganese(III), manganese(II), potassium, sodium and zink salts, but this is not intended to represent a restriction. Of the above-mentioned salts, preference is given to ammonium; the alkali metal salts sodium and potassium, and the alkaline earth metal salts calcium and magnesium. Salts of the compounds of the formula (I) which are derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines, also including naturally occurring substituted amines, cyclic amines, and basic ion exchanger resins, for example arginine, betaine, caffeine, chloroprocaine, choline, N,N′-dibenzyl-ethylen-ediamine (benzathine), dicyclohexylamine, diethanolamine, diethyl-amine, 2-diethyl-amino-ethanol, 2-dimethyl-amino-ethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethyl-piperidine, glucamine, glucosamine, histidine, hydrabamine, isopropyl-amine, lido-caine, lysine, meglumine (N-methyl-D-glucamine), morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethanolamine, triethylamine, trimethylamine, tripropyl-amine and tris(hydroxy-methyl)-methylamine (tromethamine), but this is not intended to represent a restriction. Compounds of the formula (I) of the present invention which contain basic nitrogen-containing groups can be quaternised using agents such as (C1-C4)-alkyl halides, for example methyl, ethyl, isopropyl and tert-butyl chloride, bromide and iodide; di(C1-C4)alkyl sulfates, for example dimethyl, diethyl and diamyl sulfate; (C10-C18)alkyl halides, for example decyl, do-decyl, lauryl, myristyl and stearyl chloride, bromide and iodide; and aryl-(C1-C4)alkyl halides, for example benzyl chloride and phenethyl bromide. Both water- and oil-soluble compounds of the formula (I) can be prepared using such salts. The above-mentioned pharmaceutical salts which are preferred include acetate, trifluoroacetate, besylate, citrate, fumarate, gluconate, hemisuccinate, hippurate, hydrochloride, hydrobromide, isethionate, mandelate, meglumine, nitrate, oleate, phosphonate, pivalate, sodium phosphate, stearate, sulfate, sulfosalicylate, tartrate, thiomalate, tosylate and tro-meth-amine, but this is not intended to represent a restriction. The acid-addition salts of basic compounds of the formula (I) are prepared by bringing the free base form into contact with a sufficient amount of the desired acid, causing the formation of the salt in a conventional manner. The free base can be regenerated by bringing the salt form into contact with a base and isolating the free base in a conventional manner. The free base forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solubility in polar solvents; for the purposes of the invention, however, the salts otherwise correspond to the respective free base forms thereof. As mentioned, the pharmaceutically acceptable base-addition salts of the compounds of the formula (I) are formed with metals or amines, such as alkali metals and alkaline earth metals or organic amines. Preferred metals are sodium, potassium, magnesium and calcium. Preferred organic amines are N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methyl-D-glucamine and procaine. The base-addition salts of acidic compounds of the formula (I) are prepared by bringing the free acid form into contact with a sufficient amount of the desired base, causing the formation of the salt in a conventional manner. The free acid can be regenerated by bringing the salt form into contact with an acid and isolating the free acid in a conventional manner. The free acid forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solubility in polar solvents; for the purposes of the invention, however, the salts otherwise correspond to the respective free acid forms thereof. If a compound of the formula (I) contains more than one group which is capable of forming pharmaceutically acceptable salts of this type, the formula (I) also encompasses multiple salts. Typical multiple salt forms include, for example, bitartrate, diacetate, difumarate, dimeglumine, di-phosphate, disodium and trihydrochloride, but this is not intended to represent a restriction. With regard to that stated above, it can be seen that the term “pharmaceutically acceptable salt” in the present connection is taken to mean an active ingredient which comprises a compound of the formula (I) in the form of one of its salts, in particular if this salt form imparts improved pharmacokinetic properties on the active ingredient compared with the free form of the active ingredient or any other salt form of the active ingredient used earlier. The pharmaceutically acceptable salt form of the active ingredient can also provide this active ingredient for the first time with a desired pharmacokinetic property which it did not have earlier and can even have a positive influence on the pharmacodynamics of this active ingredient with respect to its therapeutic efficacy in the body. Owing to their molecular structure, the compounds of the formula (I) are chiral and can accordingly occur in various enantiomeric forms. They can therefore exist in racemic or in optically active form. Since the pharmaceutical activity of the racemates or stereoisomers of the compounds according to the invention may differ, it may be desirable to use the enantiomers. In these cases, the end product or even the Intermediates can be separated into enantiomeric compounds by chemical or physical measures known to the person skilled in the art or even employed as such in the synthesis. In the case of racemic amines, diastereomers are formed from the mixture by reaction with an optically active resolving agent. Examples of suitable resolving agents are optically active acids, such as the (R)- and (S)-forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid, suitable N-protected amino acids (for example N-benzoylproline or N-benzenesulfonylproline), or the various optically active camphorsulfonic acids. Also advantageous is chromatographic enantiomer resolution with the aid of an optically active resolving agent (for example dinitrobenzoylphenylglycine, cellulose triacetate or other derivatives of carbohydrates or chirally derivatised methacrylate polymers immobilised on silica gel). Suitable eluents for this purpose are aqueous or alcoholic solvent mixtures, such as, for example, hexane/isopropanol/acetonitrile, for example in the ratio 82:15:3. Isotopes There is furthermore intended that a compound of the formula (I) includes isotope-labelled forms thereof. An isotope-labelled form of a compound of the formula (I) is identical to this compound apart from the fact that one or more atoms of the compound have been replaced by an atom or atoms having an atomic mass or mass number which differs from the atomic mass or mass number of the atom which usually occurs naturally. Examples of isotopes which are readily commercially available and which can be incorporated into a compound of the formula (I) by well-known methods include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, for example2H,3H,13C,14C,15N,18O,17O,31P,32P,35S,18F and38Cl, respectively. A compound of the formula (I), a prodrug, thereof or a pharmaceutically acceptable salt of either which contains one or more of the above-mentioned isotopes and/or other isotopes of other atoms is intended to be part of the present invention. An isotope-labelled compound of the formula (I) can be used in a number of beneficial ways. For example, an isotope-labelled compound of the formula (I) into which, for example, a radioisotope, such as3H or14C, has been incorporated is suitable for medicament and/or substrate tissue distribution assays. These radioisotopes, i.e. tritium (3H) and carbon-14 (14C), are particularly preferred owing to simple preparation and excellent detectability. Incorporation of heavier isotopes, for example deuterium (2H), into a compound of the formula (I) has therapeutic advantages owing to the higher metabolic stability of this isotope-labelled compound. Higher metabolic stability translates directly into an increased in vivo half-life or lower dosages, which under most circumstances would represent a preferred embodiment of the present invention. An isotope-labelled compound of the formula (I) can usually be prepared by carrying out the procedures disclosed in the synthesis schemes and the related description, in the example part and in the preparation part in the present text, replacing a non-isotope-labelled reactant by a readily available isotope-labelled reactant. Deuterium (2H) can also be incorporated into a compound of the formula (I) for the purpose in order to manipulate the oxidative metabolism of the compound by way of the primary kinetic isotope effect. The primary kinetic isotope effect is a change of the rate for a chemical reaction that results from exchange of isotopic nuclei, which in turn is caused by the change in ground state energies necessary for covalent bond formation after this isotopic exchange. Exchange of a heavier isotope usually results in a lowering of the ground state energy for a chemical bond and thus cause a reduction in the rate in rate-limiting bond breakage. If the bond breakage occurs in or in the vicinity of a saddle-point region along the coordinate of a multi-product reaction, the product distribution ratios can be altered substantially. For explanation: if deuterium is bonded to a carbon atom at a non-exchangeable position, rate differences of kM/kD=2-7 are typical. If this rate difference is successfully applied to a compound of the formula (I) that is susceptible to oxidation, the profile of this compound in vivo can be drastically modified and result in improved pharmacokinetic properties. When discovering and developing therapeutic agents, the person skilled in the art attempts to optimise pharmacokinetic parameters while retaining desirable in vitro properties. It is reasonable to assume that many compounds with poor pharmacokinetic profiles are susceptible to oxidative metabolism. In vitro liver microsomal assays currently available provide valuable information on the course of oxidative metabolism of this type, which in turn permits the rational design of deuterated compounds of the formula (I) with improved stability through resistance to such oxidative metabolism. Significant improvements in the pharmacokinetic profiles of compounds of the formula (I) are thereby obtained, and can be expressed quantitatively in terms of increases in the in vivo half-life (t½), concentration at maximum therapeutic effect (Cmax), area under the dose response curve (AUC), and F; and in terms of reduced clearance, dose and materials costs. The following is intended to illustrate the above: a compound of the formula (I) which has multiple potential sites of attack for oxidative metabolism, for example benzylic hydrogen atoms and hydrogen atoms bonded to a nitrogen atom, is prepared as a series of analogues in which various combinations of hydrogen atoms are replaced by deuterium atoms, so that some, most or all of these hydrogen atoms have been replaced by deuterium atoms. Half-life determinations enable favourable and accurate determination of the extent of the extent to which the improvement in resistance to oxidative metabolism has improved. In this way, it is determined that the half-life of the parent compound can be extended by up to 100% as the result of deuterium-hydrogen exchange of this type. Deuterium-hydrogen exchange in a compound of the formula (I) can also be used to achieve a favourable modification of the metabolite spectrum of the starting compound in order to diminish or eliminate undesired toxic metabolites. For example, if a toxic metabolite arises through oxidative carbon-hydrogen (C H) bond cleavage, it can reasonably be assumed that the deuterated analogue will greatly diminish or eliminate production of the unwanted metabolite, even if the particular oxidation is not a rate-determining step. Further information on the state of the art with respect to deuterium-hydrogen exchange may be found, for example in Hanzlik et al., J. Org. Chem. 55, 3992-3997, 1990, Reider et al., J. Org. Chem. 52, 3326-3334, 1987, Foster, Adv. Drug Res. 14, 1-40, 1985, Gillette et al, Biochemistry 33(10) 2927-2937, 1994, and Jarman et al. Carcinogenesis 16(4), 683-688, 1993. The present invention relates to a pharmaceutical formulation (preferably for use in the treatment of an immunoregulatory abnormality or a cancer) comprising at least one compound of formula (I) (particularly a therapeutically effective amount of a compound of formula (I)), and/or a prodrug, solvate, tautomer, oligomer, adduct or stereoisomer thereof as well as a pharmaceutically acceptable salt of each of the foregoing, including mixtures thereof in all ratios, as active ingredient, together with a pharmaceutically acceptable carrier. For the purpose of the present invention the term “pharmaceutical formulation” refers to a composition or product comprising one or more active ingredients, and one or more inert ingredients that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical formulations of the present invention encompass any composition made by admixing at least one compound of the present invention and a pharmaceutically acceptable carrier, excipient or vehicle. The pharmaceutical formulations of the present invention also encompass any composition that further comprises a second active ingredient and/or a prodrug or solvate thereof as well as a pharmaceutically acceptable salt of each of the foregoing, including mixtures thereof in all ratios, wherein that second active ingredient is other than a compound of formula (I) wherein all residues are defined above. The pharmaceutical formulations according to the present invention can be used as medicaments in human and veterinary medicine. For the purpose of the present invention an immunoregulatory abnormality is preferably an autoimmune or chronic inflammatory disease selected from the group consisting of: systemic lupus erythematosis, chronic rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, amyotrophic lateral sclerosis (ALS), atherosclerosis, scleroderma, autoimmune hepatitis, Sjogren Syndrome, lupus nephritis, glomerulonephritis, Rheumatoid Arthritis, Psoriasis, Myasthenia Gravis, Imunoglobuline A nephropathy, Vasculitis, Transplant rejection, Myositis, Henoch-Schonlein Purpura and asthma; cancer is preferably a hematological malignancy or a solid tumor, wherein the hematological malignancy is preferably a disease selected from the group of malignant B- and T/NK-cell non-Hodgkin lymphoma such as: multiple myeloma, mantle cell lymphoma, diffuse large B-cell lymphoma, plasmocytoma, follicular lymphoma, immunocytoma, acute lymphoblastic leukemia, chronic lymphocytic leukemia and myeloid leukemia; and wherein the solid tumor is preferably a disease selected from the group of: inflammatory breast, liver and colon cancer, lung cancer, head and neck cancer, prostate cancer, pancreas cancer, bladder cancer, renal cancer, hepatocellular cancer and gastric cancer. Pharmaceutical formulations can be administered in the form of dosage units, which comprise a predetermined amount of active ingredient per dosage unit. Such a unit can comprise, for example, 0.5 mg to 1 g, preferably 1 mg to 700 mg, particularly preferably 5 mg to 100 mg, of a compound according to the invention, depending on the disease condition treated, the method of administration and the age, weight and condition of the patient, or pharmaceutical formulations can be administered in the form of dosage units which comprise a predetermined amount of active ingredient per dosage unit. Preferred dosage unit formulations are those which comprise a daily dose or part-dose, as indicated above, or a corresponding fraction thereof of an active ingredient. Furthermore, pharmaceutical formulations of this type can be prepared using a process, which is generally known in the pharmaceutical art. Pharmaceutical formulations can be adapted for administration via any desired suitable method, for example by oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) methods. Such formulations can be prepared using all processes known in the pharmaceutical art by, for example, combining the active ingredient with the excipient(s) or adjuvant(s). Pharmaceutical formulations adapted for oral administration can be administered as separate units, such as, for example, capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or foam foods; or oil-in-water liquid emulsions or water-in-oil liquid emulsions. Thus, for example, in the case of oral administration in the form of a tablet or capsule, the active-ingredient component can be combined with an oral, non toxic and pharmaceutically acceptable inert excipient, such as, for example, ethanol, glycerol, water and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing it with a pharmaceutical excipient comminuted in a similar manner, such as, for example, an edible carbohydrate, such as, for example, starch or mannitol. A flavour, preservative, dispersant and dye may likewise be present. Capsules are produced by preparing a powder mixture as described above and filling shaped gelatine shells therewith. Glidants and lubricants, such as, for example, highly disperse silicic acid, talc, magnesium stearate, calcium stearate or polyethylene glycol in solid form, can be added to the powder mixture before the filling operation. A disintegrant or solubiliser, such as, for example, agar-agar, calcium carbonate or sodium carbonate, may likewise be added in order to improve the availability of the medica-ment after the capsule has been taken. In addition, if desired or necessary, suitable binders, lubricants and disintegrants as well as dyes can likewise be incorporated into the mixture. Suitable binders include starch, gelatine, natural sugars, such as, for example, glucose or beta-lactose, sweeteners made from maize, natural and synthetic rubber, such as, for example, acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. The lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. The disintegrants include, without being restricted thereto, starch, methylcellulose, agar, bentonite, xanthan gum and the like. The tablets are formulated by, for example, preparing a powder mixture, granulating or dry-pressing the mixture, adding a lubricant and a disintegrant and pressing the entire mixture to give tablets. A powder mixture is prepared by mixing the compound comminuted in a suitable manner with a diluent or a base, as described above, and optionally with a binder, such as, for example, carboxymethylcellulose, an alginate, gelatine or polyvinyl-pyrrolidone, a dissolution retardant, such as, for example, paraffin, an absorption accelerator, such as, for example, a quaternary salt, and/or an absorbant, such as, for example, bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting it with a binder, such as, for example, syrup, starch paste, acadia mucilage or solutions of cellulose or polymer materials and pressing it through a sieve. As an alternative to granulation, the powder mixture can be run through a tableting machine, giving lumps of non-uniform shape which are broken up to form granules. The granules can be lubricated by addition of stearic acid, a stearate salt, talc or mineral oil in order to prevent sticking to the tablet casting moulds. The lubricated mixture is then pressed to give tablets. The active ingredients can also be combined with a free-flowing inert excipient and then pressed directly to give tablets without carrying out the granulation or dry-pressing steps. A transparent or opaque protective layer consisting of a shellac sealing layer, a layer of sugar or polymer material and a gloss layer of wax may be present. Dyes can be added to these coatings in order to be able to differentiate between different dosage units. Oral liquids, such as, for example, solution, syrups and elixirs, can be prepared in the form of dosage units so that a given quantity comprises a pre-specified amount of the compounds. Syrups can be prepared by dissolving the compounds in an aqueous solution with a suitable flavour, while elixirs are prepared using a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersion of the compounds in a non-toxic vehicle. Solubilisers and emulsifiers, such as, for example, ethoxylated isostearyl alcohols and polyoxyethylene sorbitol ethers, preservatives, flavour additives, such as, for example, peppermint oil or natural sweeteners or saccharin, or other artificial sweeteners and the like, can likewise be added. The dosage unit formulations for oral administration can, if desired, be encapsulated in microcapsules. The formulation can also be prepared in such a way that the release is extended or retarded, such as, for example, by coating or embedding of particulate material in polymers, wax and the like. The compounds of the formula (I) and salts, solvates and physiologically functional derivatives thereof and the other active ingredients can also be administered in the form of liposome delivery systems, such as, for example, small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from various phospholipids, such as, for example, cholesterol, stearylamine or phosphatidylcholines. The compounds of the formula (I) and the salts, solvates and physiologically functional derivatives thereof and the other active ingredients can also be delivered using monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds can also be coupled to soluble polymers as targeted medicament carriers. Such polymers may encompass polyvinylpyrrolidone, pyran copolymer, polyhydroxypropyl-methacrylamidophenol, polyhydroxyethylaspartamido-phenol or polyethylene oxide polylysine, substituted by palmitoyl radicals. The compounds may furthermore be coupled to a class of biodegradable polymers which are suitable for achieving controlled release of a medicament, for example polylactic acid, poly-epsilon-caprolactone, polyhydroxybutyric acid, poly-orthoesters, polyacetals, polydihydroxypyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels. Pharmaceutical formulations adapted for transdermal administration can be administered as independent plasters for extended, close contact with the epidermis of the recipient. Thus, for example, the active ingredient can be delivered from the plaster by iontophoresis, as described in general terms in Pharmaceutical Research, 3(6), 318 (1986). Pharmaceutical compounds adapted for topical administration can be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. For the treatment of the eye or other external tissue, for example mouth and skin, the formulations are preferably applied as topical ointment or cream. In the case of formulation to give an ointment, the active ingredient can be employed either with a paraffinic or a water-miscible cream base. Alternatively, the active ingredient can be formulated to give a cream with an oil-in-water cream base or a water-in-oil base. Pharmaceutical formulations adapted for topical application to the eye include eye drops, in which the active ingredient is dissolved or sus-pended in a suitable carrier, in particular an aqueous solvent. Pharmaceutical formulations adapted for topical application in the mouth encompass lozenges, pastilles and mouthwashes. Pharmaceutical formulations adapted for rectal administration can be administered in the form of suppositories or enemas. Pharmaceutical formulations adapted for nasal administration in which the carrier substance is a solid comprise a coarse powder having a particle size, for example, in the range 20-500 microns, which is administered in the manner in which snuff is taken, i.e. by rapid inhalation via the nasal passages from a container containing the powder held close to the nose. Suitable formulations for administration as nasal spray or nose drops with a liquid as carrier substance encompass active-ingredient solutions in water or oil. Pharmaceutical formulations adapted for administration by inhalation encompass finely particulate dusts or mists, which can be generated by various types of pressurised dispensers with aerosols, nebulisers or insuf-flators. Pharmaceutical formulations adapted for vaginal administration can be administered as pessaries, tampons, creams, gels, pastes, foams or spray formulations. Pharmaceutical formulations adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions comprising antioxidants, buffers, bacteriostatics and solutes, by means of which the formulation is rendered isotonic with the blood of the recipient to be treated; and aqueous and non-aqueous sterile suspensions, which may comprise suspension media and thickeners. The formulations can be administered in single-dose or multidose containers, for example sealed ampoules and vials, and stored in freeze-dried (lyophilised) state, so that only the addition of the sterile carrier liquid, for example water for injection purposes, immediately before use is necessary. Injection solutions and suspensions prepared in accordance with the recipe can be prepared from sterile powders, granules and tablets. It goes without saying that, in addition to the above particularly mentioned constituents, the formulations may also comprise other agents usual in the art with respect to the particular type of formulation; thus, for example, formulations which are suitable for oral administration may comprise flavours. The compositions/formulations according to the invention can be used as medicaments in human and veterinary medicine. A therapeutically effective amount of a compound of the formula (I) and of the other active ingredient depends on a number of factors, including, for example, the age and weight of the animal, the precise disease condition which requires treatment, and its severity, the nature of the formulation and the method of administration, and is ultimately determined by the treating doctor or vet. However, an effective amount of a compound is generally in the range from 0.1 to 100 mg/kg of body weight of the recipient (mammal) per day and particularly typically in the range from 1 to 10 mg/kg of body weight per day. Thus, the actual amount per day for an adult mammal weighing 70 kg is usually between 70 and 700 mg, where this amount can be administered as an individual dose per day or usually in a series of part-doses (such as, for example, two, three, four, five or six) per day, so that the total daily dose is the same. An effective amount of a salt or solvate or of a physiologically functional derivative thereof can be determined as the fraction of the effective amount of the compound per se. The invention further relates to a compound according to formula (I) or any specific embodiment described above and/or its prodrugs, solvates, tautomers, oligomers, adducts or stereoisomers thereof as well as the pharmaceutically acceptable salts of each of the foregoing, including mixtures thereof in all ratios, for use in the prevention and/or treatment of medical conditions that are affected by inhibiting LMP7. The invention relates to a compound according to formula (I) or any specific embodiment described above and/or a prodrug, solvate, tautomers, oligomers, adducts or stereoisomers thereof as well as the pharmaceutically acceptable salts of each of the foregoing, including mixtures thereof in all ratios, for use in the treatment and/or prophylaxis (prevention) of an immunoregulatory abnormality or cancer (including in particular hematological malignancy and solid tumors). The present invention furthermore relates to a method of treating a subject suffering from an immunoregulatory abnormality or a cancer, comprising administering to said subject a compounds of formula (I) in an amount that is effective for treating said immunoregulatory abnormality or a cancer. The present invention preferably relates to a method of treating a subject suffering from an autoimmune or chronic inflammatory disease, a hematological malignancy or a solid tumor. The disclosed compounds of the formula (I) can be administered and/or used in combination with other known therapeutic agents (active ingredients), including anticancer agents. As used herein, the term “anticancer agent” relates to any agent which is administered to a patient with cancer for the purposes of treating the cancer. The anti-cancer treatment defined above may be applied as a monotherapy or may involve, in addition to the herein disclosed compounds of formula (I), conventional surgery or radiotherapy or medicinal therapy. Such medicinal therapy, e.g. a chemotherapy or a targeted therapy, may include one or more, but preferably one, of the following anti-tumor agents: Alkylating Agents such as altretamine, bendamustine, busulfan, carmustine, chlorambucil, chlormethine, cyclophosphamide, dacarbazine, ifosfamide, improsulfan, tosilate, lomustine, melphalan, mitobronitol, mitolactol, nimustine, ranimustine, temozolomide, thiotepa, treosulfan, mechloretamine, carboquone;apaziquone, fotemustine, glufosfamide, palifosfamide, pipobroman, trofosfamide, uramustine, TH-3024, VAL-0834; Platinum Compoundssuch as carboplatin, cisplatin, eptaplatin, miriplatine hydrate, oxaliplatin, lobaplatin, nedaplatin, picoplatin, satraplatin; DNA Altering Agentssuch as amrubicin, bisantrene, decitabine, mitoxantrone, procarbazine, trabectedin, clofarabine;amsacrine, brostallicin, pixantrone, laromustine1,3; Topoisomerase Inhibitorssuch as etoposide, irinotecan, razoxane, sobuzoxane, teniposide, topotecan; amonafide, belotecan, elliptinium acetate, voreloxin; Microtubule Modifierssuch as cabazitaxel, docetaxel, eribulin, ixabepilone, paclitaxel, vinblastine, vincristine, vinorelbine, vindesine, vinflunine;fosbretabulin, tesetaxel; Antimetabolitessuch as asparaginase3, azacitidine, calcium levofolinate, capecitabine, cladribine, cytarabine, enocitabine, floxuridine, fludarabine, fluorouracil, gemcitabine, mercaptopurine, methotrexate, nelarabine, pemetrexed, pralatrexate, azathioprine, thioguanine, carmofur; doxifluridine, elacytarabine, raltitrexed, sapacitabine, tegafur2,3, trimetrexate; Anticancer Antibioticssuch as bleomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, levamisole, miltefosine, mitomycin C, romidepsin, streptozocin, vairubicin, zinostatin, zorubicin, daunurobicin, plicamycin; aclarubicin, peplomycin, pirarubicin; Hormones/Antagonistssuch as abarelix, abiraterone, bicalutamide, buserelin, calusterone, chlorotrianisene, degarelix, dexamethasone, estradiol, fluocortolone, fluoxymesterone, flutamide, fulvestrant, goserelin, histrelin, leuprorelin, megestrol, mitotane, nafarelin, nandrolone, nilutamide, octreotide, prednisolone, raloxifene, tamoxifen, thyrotropin alfa, toremifene, trilostane, triptorelin, diethylstilbestrol;acolbifene, danazol, deslorelin, epitiostanol, orteronel, enzalutamide1,3; Aromatase Inhibitorssuch as aminoglutethimide, anastrozole, exemestane, fadrozole, letrozole, testolactone;formestane; Small Molecule Kinase Inhibitorssuch as crizotinib, dasatinib, erlotinib, imatinib, lapatinib, nilotinib, pazopanib, regorafenib, ruxolitinib, sorafenib, sunitinib, vandetanib, vemurafenib, bosutinib, gefitinib, axitinib;afatinib, alisertib, dabrafenib, dacomitinib, dinaciclib, dovitinib, enzastaurin, nintedanib, lenvatinib, linifanib, linsitinib, masitinib, midostaurin, motesanib, neratinib, orantinib, perifosine, ponatinib, radotinib, rigosertib, tipifamib, tivantinib, tivozanib, trametinib, pimasertib, brivanib alaninate, cediranib, apatinib4, cabozantinib S-malate1,3, ibrutinib1,3, icotinib4, buparlisib2, cipatinib4, cobimetinib1,3, idelalisib1,3, fedratinib1, XL-6474; Photosensitizerssuch as methoxsalen3;porfimer sodium, talaporfin, temoporfin; Antibodiessuch as alemtuzumab, besilesomab, brentuximab vedotin, cetuximab, denosumab, ipilimumab, ofatumumab, panitumumab, rituximab, tositumomab, trastuzumab, bevacizumab, pertuzumab2,3; catumaxomab, elotuzumab, epratuzumab, farletuzumab, mogamulizumab, necitumumab, nimotuzumab, obinutuzumab, ocaratuzumab, oregovomab, ramucirumab, rilotumumab, siltuximab, tocilizumab, zalutumumab, zanolimumab, matuzumab, dalotuzumab1,2,3, onartuzumab1,3, racotumomab1, tabalumab1,3, EMD-5257974, nivolumab1,3; Cytokinessuch as aldesleukin, interferon alfa2, interferon alfa2a3, interferon alfa2b2,3; celmoleukin, tasonermin, teceleukin, oprelvekin1,3, recombinant interferon beta-1a4; Drug Conjugatessuch as denileukin diftitox, ibritumomab tiuxetan, iobenguane I123, prednimustine, trastuzumab emtansine, estramustine, gemtuzumab, ozogamicin, aflibercept; cintredekin besudotox, edotreotide, inotuzumab ozogamicin, naptumomab estafenatox, oportuzumab monatox, technetium (99mTc) arcitumomab1,3, vintafolidet3; Vaccinessuch as sipuleuce1,3; vitespen3, emepepimut-S3, oncoVAX4, rindopepimut3, troVax4, MGN-16014, MGN-17034; Miscellaneousalitretinoin, bexarotene, bortezomib, everolimus, ibandronic acid, imiquimod, lenalidomide, lentinan, metirosine, mifamurtide, pamidronic acid, pegaspargase, pentostatin, sipuleuce1,3, sizofiran, tamibarotene, temsirolimus, thalidomide, tretinoin, vismodegib, zoledronic acid, vorinostat; celecoxib, cilengitide, entinostat, etanidazole, ganetespib, idronoxil, iniparib, ixazomib, lonidamine, nimorazole, panobinostat, peretinoin, plitidepsin, pomalidomide, procodazol, ridaforolimus, tasquinimod, telotristat, thymalfasin, tirapazamine, tosedostat, trabedersen, ubenimex, valspodar, gendicine4, picibanil4, reolysin4, retaspimycin hydrochloride1,3, trebananib2,3, virulizin4, carfilzomib1,3, endostatin4, immucothel4, belinostat3, MGN-17034;1Prop. INN (Proposed International Nonproprietary Name)2Rec. INN (Recommended International Nonproprietary Names)3USAN (United States Adopted Name)4no INN. The invention furthermore relates to the use of compounds of formula (I), and related formulae in combination with at least one further medicament active ingredient, preferably medicaments used in the treatment of multiple sclerosis such as cladribine or another co-agent, such as interferon, e.g. pegylated or non-pegylated interferons, preferably interferon beta and/or with compounds improving vascular function or in combination with immunomodulating agents for example Fingolimod; cyclosporins, rapamycins or ascomycins, or their immunosuppressive analogs, e.g. cyclosporin A, cyclosporin G, FK-506, ABT-281, ASM981, rapamycin, 40-O-(2-hydroxy)ethyl-rapamycin etc.; corticosteroids; cyclophosphamide; azathioprene: methotrexate; leflunomide; mizoribine; mycophenolic add; mycophenolate mofetil; 15-deoxyspergualine; diflucortolone valerate; difluprednate; Alclometasone dipropionate; amcinonide; amsacrine; asparaginase; azathioprine; basiliximab; beclometasone dipropionate; betamethasone; betamethasone acetate; betamethasone dipropionate; betamethasone phosphate sodique; betamethasone valerate; budesonide; captopril; chlormethine chlorhydrate; cladribine; clobetasol propionate; cortisone acetate; cortivazol; cyclophosphamide; cytarabine; daclizumab; dactinomycine; desonide; desoximetasone; dexamethasone; dexamethasone acetate; dexamethasone isonicotinate; dexamethasone metasulfobenzoate sodique; dexamethasone phosphate; dexamethasone tebutate; dichlorisone acetate; doxorubicine chlorhydrate; epirubicine chlorhydrate; fluclorolone acetonide; fludrocortisone acetate; fludroxycortide; flumetasone pivalate; flunisolide; fluocinolone acetonide; fluocinonide; fluocortolone; fluocortolone hexanoate; fluocortolone pivalate; fluorometholone; fluprednidene acetate; fluticasone propionate; gemcitabine chlorhydrate; halcinonide; hydrocortisone, hydrocortisone acetate, hydrocortisone butyrate, hydrocortisone hemisuccinate; melphalan; meprednisone; mercaptopurine; methylprednisolone; methylprednisolone acetate; methylprednisolone hemisuccinate; misoprostol; muromonab-cd3; mycophenolate mofetil; paramethasone acetate; prednazoline, prednisolone; prednisolone acetate; prednisolone caproate; prednisolone metasulfobenzoate sodique; prednisolone phosphate sodique; prednisone; prednylidene; rifampicine; rifampicine sodique; tacrolimus; teriflunomide; thalidomide; thiotepa; tixocortol pivalate; triamcinolone; triamcinolone acetonide hemisuccinate; triamcinolone benetonide; triamcinolone diacetate; triamcinolone hexacetonide; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD25, CD28, B7, CD40, CD45 or CD58 or their ligands; or other immunomodulatory compounds, e.g. CTLA41g, or other adhesion molecule inhibitors, e.g. mAbs or low molecular weight inhibitors including Selectin antagonists and VLA-4 antagonists. A preferred composition is with Cyclosporin A, FK506, rapamycin or 40-(2-hydroxy)ethyl-rapamycin and Fingolimod. These further medicaments, such as interferon beta, may be administered concomitantly or sequentially, e.g. by subcutaneous, intramuscular or oral routes. The invention furthermore relates to the use of compounds of formula (I), and related formulae in combination with at least one further medicament active ingredient, preferably medicaments used in the treatment of cancer (such as in particular the anticancer and/or antitumor agents described above). The present invention further relates to a set (kit) consisting of separate packs of(a) an effective amount of a compound of the formula (I) and/or a prodrug, solvate, tautomer, oligomer, adduct or stereoisomer thereof as well as a pharmaceutically acceptable salt of each of the foregoing, including mixtures thereof in all ratios,and(b) an effective amount of a further medicament active ingredient. The compounds of the present invention can be prepared according to the procedures of the following Schemes and Examples, using appropriate materials, and are further exemplified by the following specific examples. Moreover, by utilizing the procedures described herein, in conjunction with ordinary skills in the art, additional compounds of the present invention claimed herein can be readily prepared. The compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention. The examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds. The starting materials for the preparation of compounds of the present invention can prepared by methods as described in the examples or by methods known per se, as described in the literature of synthetic organic chemistry and known to the skilled artisan, or can be obtained commercially. The synthesis of compounds of formula (IV) is described in WO 2016/050356, WO 2016/050355, WO 2016/050359, and WO 2016/050358. Examples LCMS: Method A: Agilent 70108359—Chromolith Speed Rod RP18e 50-4.6 mm; polar.m; 2.4 mL/min; 220 nm; buffer A: 0.05% HCOOH/H2O, buffer B: 0.04% HCOOH/ACN; 0.0-2.8 min 4%-100% buffer B; 2.8-3.3 min 100% buffer B; 3.3-3.4 min 100%-4% buffer B. Method B: Waters XBrigde C8 3.5 μm; 4.6×50 mm; EliteLa Chrom 70173815; 8.1 min; 2 ml/min; 215 nm; buffer A: 0.05% TFA/H2O; buffer B: 0.04% TFA/ACN; 0.0-0.2 min 5% buffer B; 0.2-8.5 min 5%-100% buffer B; 8.5-10.0 min 99%-5% buffer. RT: Retention rime. The invention will be illustrated, but not limited, by reference to the specific embodiments described in the following examples. Unless otherwise indicated in the schemes, the variables have the same meaning as described above. Unless otherwise specified, all starting materials are obtained from commercial suppliers and used without further purifications. Unless otherwise specified, all temperatures are expressed in ° C. and all reactions are conducted at rt. Compounds can be purified by common means such as in particular silica chromatography or preparative HPLC. Unless stated otherwise all structures indicated below, where no specific stereochemistry is indicated, refer to mixtures of the stereoisomers. Intermediate 1 Step 1: 2-(2,4-Dimethyl-benzyl)-4,4,5,5-tetramethyl-[1,3,2]dioxaborolane To a solution of 1-Bromomethyl-2,4-dimethyl-benzene (25.00 g; 114.40 mmol; 1.00 eq.) in degased Dioxane (250.00 ml), Bis(pinacolato)diboron (35.21 g; 137.28 mmol; 1.20 eq.), dried K2CO3(47.91 g; 343.19 mmol; 3.00 eq.) and Tetrakis(triphenylphosphine)palladium(0) (6623 mg; 5.72 mmol; 0.05 eq.) are added. The reaction mixture is then heated at 100° C. under nitrogen atmosphere for 16 h. The reaction mixture is diluted with dichloromethane and passed through celite. The filtrate is concentrated. The residue is dissolved in ethyl acetate and washed with brine. The organic layer is dried over anhydrous Na2SO4, filtered and concentrated. The crude is purified by column chromatography using 1% ethyl acetate in petroleum ether to get 2-(2,4-Dimethyl-benzyl)-4,4,5,5-tetramethyl-[1,3,2]dioxaborolane (11.50 g; 37.84 mmol; 33.1%) as colorless liquid. 1H NMR (400 MHz, CDCl3) δ 7.04-7.02 (m, 1H), 6.95-6.93 (m, 1H), 6.92-6.90 (m, 1H), 2.28 (s, 3H), 2.25 (s, 3H), 2.23 (s, 2H), 1.24 (s, 12H). Step 2: (1S,2S,6R,8S)-4-(2,4-Dimethyl-benzyl)-2,9,9-Dimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane To an ice-cooled solution of 2-(2,4-Dimethyl-benzyl)-4,4,5,5-tetramethyl-[1,3,2]dioxaborolane (24.00 g; 79.3 mmol; 1.0 eq.) in Diethyl ether (240.00 ml) under nitrogen atmosphere, (1S,2S,3R,5S)-2,6,6-Trimethyl-bicyclo[3.1.1]heptane-2,3-diol (20.68 g; 119.07 mmol; 1.50 eq.) is added and the reaction mixture is stirred at rt for 14 h. TLC analysis showed completion of reaction. The reaction mixture is washed with brine. The organic layer is dried over anhydrous Na2SO4and concentrated. The crude is purified by flash column chromatography using 2% ethyl acetate in petroleum ether to get (1S,2S,6R,8S)-4-(2,4-Dimethyl-benzyl)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (28.00 g; 82.96 mmol; 90.0%) as colorless oil. 1H NMR (400 MHz, CDCl3): δ 7.05-7.03 (m, 1H), 6.95-6.94 (m, 1H), 6.92-6.90 (m, 1H), 4.27-4.25 (m, 1H), 2.33-2.30 (m, 9H), 2.27-2.17 (m, 1H), 2.05 (t, J=5.76 Hz, 1H), 1.90-1.89 (m, 1H), 1.84-1.80 (m, 1H), 1.38 (s, 3H), 1.28 (s, 3H), 1.11-1.09 (m, 1H), 0.91 (s, 3H) GCMS: m/z: 298.3. Step 3: (1S,2S,6R,8S)-4-[(S)-1-Chloro-2-(2,4-dimethyl-phenyl)-ethyl]-2,9,9-timethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane Dichloromethane (37.33 ml; 583.45 mmol; 3.00 eq.) in Tetrahydrofuran (140.00 ml) is taken in a RB-flask (round bottom flask) under a positive pressure of nitrogen and cooled to −99° C. using liquid nitrogen-ethanol mixture. To this n-butyl lithium (1.6 M in THF) (133.71 ml; 213.93 mmol) is added dropwise through the sides of the RB-flask (at a medium rate, addition took about 35 min.) so that the internal temperature is maintained between −92° C. and −102° C. After addition, the reaction mixture is stirred for 25 minutes. During the course of the reaction a white precipitate is formed (The internal temperature is maintained between −90° C. and −96° C.). Then a solution of (1S,2S,6R,8S)-4-(2,4-Dimethyl-benzyl)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (58.00 g; 194.5 mmol) in THF (300.00 ml) is added dropwise through the sides of the RB-flask (about 40 min) so that the internal temperature is maintained between −94° C. and 100° C. After addition the reaction mixture is stirred for 10 min. Then zinc chloride (0.5 M in THF) (388.97 ml; 194.48 mmol) is added dropwise through the sides of the RB-flask (at a medium rate, addition took about 35 min.) so that the internal temperature is maintained between −94° C. and −99° C. The reaction mixture is then slowly allowed to reach 20° C. and stirred at 20° C. for 2.5 h. An aliquot of the reaction mixture is worked-up and analysed by1H NMR which showed the completion of reaction. The reaction mixture is concentrated (temperature of the bath 30° C.). The residue is partitioned between diethyl ether and saturated NH4Cl solution. The organic layer is dried over anhydrous Na2SO4and concentrated (temperature of bath 30° C.) to afford (1S,2S,6R,8S)-4-[(S)-1-Chloro-2-(2,4-dimethyl-phenyl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bore-tricyclo[6.1.1.02,6]decane (75.70 g; 154.83 mmol; 79.6%) as white solid. 1H NMR (400 MHz, CDCl3): □ 7.12 (d, J=7.64 Hz, 1H), 6.98 (s, 1H), 6.96 (d, J=7.68 Hz, 1H), 4.38-4.36 (m, 1H), 3.67-3.62 (m, 1H), 3.18-3.11 (m, 2H), 2.40-2.36 (m, 2H), 2.32 (s, 3H), 2.30 (s, 3H), 2.23-2.20 (m, 1H), 2.08 (t, J=5.96 Hz, 1H), 1.93-1.87 (m, 2H), 1.36 (s, 3H), 1.30 (s, 3H), 1.14-1.11 (m, 1H), 0.84 (s, 3H). 7.18-7.08 (m, 5H), 4.37 (dd, J=1.32, 8.74 Hz, 1H), 3.77-3.75 (m, 1H), 3.67-3.63 (m, 1H), 3.19-3.17 (m, 1H), 3.10-3.08 (m, 1H), 2.36-2.31 (m, 5H), 2.09 (t, J=5.84 Hz, 1H), 1.93-1.86 (m, 4H), 1.39 (s, 3H), 1.30 (s, 3H), 1.13-1.10 (m, 1H), 0.84 (s, 3H). GCMS: m/z: 346.3. Step 4: (1S,2S,6R,8S)-4-[(R)-2-(2,4-Dimethyl-phenyl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane A solution of (1S,2S,6R,8S)-4-[(S)-1-Chloro-2-(2,4-dimethyl-phenyl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (75.70 g; 218.35 mmol; 1.00 eq.) in THF (400.00 ml) under a positive pressure of nitrogen atmosphere is cooled to −78° C. To this a solution of Lithium (bistrimethylsilyl)amide (1.0 M in THF) (262 ml; 262 mmol; 1.20 eq.) is added dropwise over a period of 30 minutes. The reaction mixture is allowed to attain rt and stirred at rt for 18 h. The reaction mixture is evaporated at a temperature 30° C. The residue is triturated with hexane and the solid formed is filtered. The filtrate is allowed to stand for some time under vacuum and any solid if formed is filtered again. The filtrate is concentrated at a temperature 30° C. to get (1S,2S,6R,8S)-4-[(R)-2-(2,4-Dimethyl-phenyl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (80.10 g; 169.84 mmol; 77.8%; brown oil). 1H NMR (400 MHz, CDCl3): δ: 7.06 (d, J=7.64 Hz, 1H), 6.94 (s, 1H), 6.90 (d, J=7.80 Hz, 1H), 4.29-4.27 (m, 1H), 3.15-3.10 (m, 1H), 2.87-2.83 (m, 1H), 2.58-2.53 (m, 1H), 2.34-2.32 (m, 2H), 2.30 (s, 3H), 2.28 (s, 3H), 2.15-2.13 (m, 1H), 2.03 (t, J=5.88 Hz, 1H), 1.90-1.88 (m, 1H), 1.81-1.77 (m, 1H), 1.39 (s, 3H), 1.32 (s, 3H), 1.01-0.98 (m, 1H), 0.93 (s, 3H), 0.85 (s, 3H), 0.09 (s, 18H). Step 5: (R)-2-(2,4-Dimethyl-phenyl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine Hydrochloride A stirred solution of (1S,2S,6R,8S)-4-[(R)-2-(2,4-Dimethyl-phenyl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-timethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,5]decane (80.10 g; 169.84 mmol; 1.00 eq.) in diethyl ether (400.00 ml) under nitrogen atmosphere is cooled to −10° C. To this 2M solution of Hydrochloric acid in diethylether (212.30 ml; 424.59 mmol; 2.50 eq.) is added dropwise. The reaction mixture is stirred at rt for 2 h. The reaction mixture is evaporated under reduced pressure to get (R)-2-(2,4-Dimethyl-phenyl)-1-((1S,2S,6R,8S)-2,9,9-timethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine hydrochloride (63.00 g; 72.61 mmol; 42.8%; brown solid). 1H NMR (400 MHz, DMSO-d6): δ 8.19 (s, 3H), 7.05 (d, J=7.68 Hz, 1H), 6.95 (s, 1H), 6.90 (d, J=8.16 Hz, 1H), 4.31 (dd, J=1.80, 8.76 Hz, 1H), 3.02-3.00 (m, 1H), 2.99-2.92 (m, 1H), 2.87-2.84 (m, 1H), 2.26-2.24 (m, 3H), 2.26 (s, 3H), 2.24 (s, 3H), 2.03-2.00 (m, 1H), 1.91 (t, J=5.68 Hz, 1H), 1.82-1.80 (m, 1H), 1.71-1.66 (m, 1H), 1.31 (s, 3H), 1.21 (s, 3H), 0.98-0.96 (m, 1H), 0.77 (s, 3H). By similar sequences described for intermediate 1 the following compounds can be prepared wherein the group Y denotes one of the following groups: Intermediate 2 Step 1: Benzofuran-3-ylmethanol A solution of 1-Benzofuran-3-carbaldehyde (5 g, 34.2 mmol) in methanol (50 mL) is cooled with ice and sodium borohydride (1.9 g, 51.3 mmol) is added portionwise. The reaction mixture is stirred at room temperature for 1 h. The reaction mixture is concentrated and the residue is partitioned between saturated ammonium chloride and ethylacetate. The organic layer is separated, dried over sodium sulfate and concentrated (5.0 g, colourless liquid, 98%). 1H NMR (400 MHz, CDCl3): δ 7.70-7.68 (m, 1H), 7.62 (s, 1H), 7.52-7.50 (m, 1H), 7.36-7.26 (m, 2H), 4.86 (s, 2H). Step 2: 3-(bromomethyl)benzofuran A cold (0° C.) solution of benzofuran-3-ylmethanol (5.0 g, 33.7 mmol) in diethyl ether (50 mL) is treated with phosphorus tribromide (1.1 mL, 11.2 mmol) and the reaction mixture is stirred at 0° C. for 30 min. The reaction mixture is then poured into ice and extracted with ether. The organic layer is dried over sodium sulfate and concentrated (7.1 g, yellow liquid, 100%). 1H NMR (400 MHz, CDCl3): δ 7.74-7.71 (m, 2H), 7.53 (s, 1H), 7.39-7.31 (m, 2H), 4.65 (s, 2H). Step 3: 2-(benzofuran-3-ylmethyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane A solution of 3-(bromomethyl)benzofuran (7.1 g, 33.8 mmol) in degassed 1,4-dioxane (70 ml) is treated with bis(pinacolato)diboron (10.3 g, 40.5 mmol), potassium carbonate (13.9 g, 101.0 mmol), tetrakis(triphenylphosphine) palladium(0) (1.9 g, 1.7 mmol) and the mixture heated at 100° C. for 12 h The contents of the flask are cooled to room temperature and filtered through a celite bed. Filtrate is concentrated and the crude is purified by flash column chromatography on silica gel, eluting with 2-5% of ethylacetate in petroleum ether to get the title compound (6.1 g, 69%) as yellow oil. 1H NMR (400 MHz, CDCl3) b 7.57-7.52 (m, 2H), 7.46-7.44 (m, 1H), 7.30-7.21 (m, 2H), 2.23 (s, 2H), 1.29 (s, 12H). Step 4: 2-(benzofuran-3-ylmethyl)boronic Acid (+)-pinanediol Ester A solution of 2-(benzofuran-3-ylmethyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (6.1 g, 23.6 mmol) in diethyl ether (60 ml) is treated with (1S,2S, 3R, 5S)-(+)-pinanediol (6.0 g, 35.4 mmol). The reaction mixture is stirred at room temperature for 12 h then the mixture is washed with water twice, then with brine and dried over anhydrous sodium sulphate, then concentrated. The crude product is purified by flash column chromatography on silica gel, eluting with 5% of ethyl acetate in petroleum ether, to afford the title compound (6.3 g. 82%). 1H NMR (400 MHz, CDCl3): δ 7.58-7.56 (m, 1H), 7.55-7.53 (m, 1H), 7.46-7.44 (m, 1H), 7.28-7.23 (m, 2H), 4.33 (dd, J=1.88, 8.76 Hz, 1H), 2.34-2.32 (m, 1H), 2.28 (s, 2H), 2.22-2.21 (m, 1H), 2.08 (t, J=5.88 Hz, 1H), 1.42 (s, 3H), 1.29 (s, 3H), 1.13 (d, J=10.92 Hz, 1H), 0.85 (s, 3H). GCMS: m/z: 310. Step 5: [(1S)-1-chloro-2-(benzofuran-3-ylmethyl)boronic Acid (+)-pinanediol Ester To a cooled (−100° C.) mixture of dichloromethane (6.3 ml, 60.9 mmol) and anhydrous tetrahydrofuran (36 ml) is added n-butyl lithium (1.6 M in hexanes, 14.0 ml, (22.3 mmol) over 20 min. After stirring for 20 min. at −100° C., a solution of 2-(benzofuran-3-ylmethyl)boronic acid (+)-pinanediol ester (6.3 g, 20.3 mmol) in anhydrous THF (22 ml) is added over 20 min. Then a solution of zinc chloride (0.5 M in THF, 36.5 mL, 18.2 mmol) is added at 100° C. over 30 min. The mixture is allowed to reach room temperature and stirred for 18 h and concentrated. To the resulting oil is added diethyl ether and saturated ammonium chloride. The organic layer is dried over anhydrous sodium sulphate and concentrated in vacuo (residue: 7.3 g, 99%). 1H NMR (400 MHz, DMSO-d6): δ 7.60-7.57 (m, 2H), 7.49-7.47 (m, 1H), 7.31-7.25 (m, 2H), 4.36-4.34 (m, 1H), 3.31-3.29 (m, 1H), 3.24-3.22 (m, 1H), 2.35-2.31 (m, 1H), 2.14-2.12 (m, 1H), 2.06 (t, J=5.84 Hz, 1H), 1.90-1.86 (m, 2H), 1.42 (s, 3H), 1.04 (d, J=11.04 Hz, 1H), 0.85 (s, 3H). GCMS: m/z: 358.2. Step 6: [(1R)-1-[bis(trimethylsilyl)amino]-2-(benzofuran-3-ylmethyl) Boronic Acid (+)-pinanediol Ester To a cooled (−78° C.) solution of [(1S)-1-chloro-2-(benzofuran-3-ylmethyl)boronic acid (+)-pinanediol ester (7.3 g, 20.3 mmol) in 40 ml of anhydrous tetrahydrofuran is added lithium bis(trimethylsilyl)amide (1M in THF, 25.5 ml, 25.5 mmol). The mixture is allowed to room temperature, stirred for 18 h and concentrated to dryness. To the resulting residue is added hexane, and then the precipitated solid is filtered off. The filtrate is concentrated to give the required crude product (6.7 g, 68%). 1H NMR (400 MHz, CDCl3): δ 7.60-7.59 (m, 1H), 7.50-7.45 (m, 2H), 7.28-7.24 (m, 2H), 4.31 (dd, J=1.56, 8.70 Hz, 1H), 3.18-3.14 (m, 1H), 2.92-2.90 (m, 1H), 2.75-2.72 (m, 1H), 2.34-2.30 (m, 1H), 2.15-2.14 (m, 1H), 2.03 (t, J=5.68 Hz, 1H), 1.88-1.80 (m, 2H), 1.39 (s, 3H), 1.30 (s, 3H), 1.01 (d, J=10.88 Hz, 1H), 0.84 (s, 3H), 0.09 (s, 18H). Step 7: [(1R)-1-amino-2-(benzofuran-3-ylmethyl)boronic Acid (+)-pinanediol Ester Trifluroacetate A cooled (0° C.) solution of [(1R)-1-[bis(trimethylsilyl)amino]-2-(benzofuran-3-ylmethyl)boronic acid (+)-pinanediol ester (6.7 g, 13.9 mmol) in diethyl ether (30 ml) is treated with trifluoroacetic acid (3.2 ml, 41.7 mmol) dropwise. The reaction mixture is then stirred at rt for 3 h. Precipitation is seen. The reaction mixture is cooled to 0° C. and filtered. The filtered solid is washed with cold ether and dried under vacuum to afford the title compound (2.3 g, white solid, 36%). 1H NMR (400 MHz, DMSO-d6): δ 7.66 (s, 1H), 7.61-7.60 (m, 1H), 7.47-7.45 (m, 1H), 7.29-7.20 (m, 2H), 4.30-4.28 (m, 1H), 3.27-3.16 (m, 3H), 2.25-2.13 (m, 3H), 1.94 (t, J=5.56 Hz, 1H), 1.86-1.81 (m, 2H), 1.25 (s, 6H), 1.01 (d, J=8.00 Hz, 1H), 0.75 (s, 3H). Intermediate 3: 2-(7-Methyl-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine Hydrochloride Step 1: 7-Methyl-benzofuran-3-carboxylic Acid Ethyl Ester To a solution of 2-Hydroxy-3-methyl-benzaldehyde (20.00 g; 139.55 mmol; 1.00 eq.) in dichloromethane (120 mL) is added Tetrafluoroboric acid diethylether complex (1.88 ml; 13.96 mmol; 0.10 eq.). To the resulting dark red mixture, Ethyldiazoacetate (31.70 ml; 300.04 mmol; 2.15 eq.) in dichloromethane (80 mL) is added drop wise slowly at 25-30° C. (internal temperature) for about 50 min. After 16 h, concentrated H2804is added. The reaction mixture is stirred for 30 min. The reaction mixture is then neutralized with solid NaHCO3, filtered through celite and the filtrate is concentrated to get a crude residue. The residue is purified by column chromatography using 2% ethyl acetate in petroleum ether to afford 7-Methyl-benzofuran-3-carboxylic acid ethyl ester (19.00 g; 86.83 mmol; 62.2%; yellow oil). HPLC (method A): RT 4.98 min (HPLC purity 93%) 1H NMR, 400 MHz, CDCl3: 8.27 (s, 1H), 7.88-7.90 (m, 1H), 7.25-7.29 (m, 1H), 7.17 (d, J=7.32 Hz, 1H), 4.39-4.45 (m, 2H), 2.55 (s, 3H), 1.44 (t, J=7.16 Hz, 3H). Step 2: (7-Methyl-benzofuran-3-yl)-methanol To a solution of 7-Methyl-benzofuran-3-carboxylic acid ethyl ester (19.00 g; 86.83 mmol; 1.00 eq.) in Dichloromethane (190.00 ml) under nitrogen is added Diisobutyl Aluminium Hydride (1.0 M in Toluene) (191.03 ml; 191.03 mmol; 2.20 eq.) drop wise at −78° C. The reaction mixture is allowed to come to rt and stirred for 1 h. The reaction mixture is cooled with ice bath and quenched with an aqueous solution of 1.5N HCl. The resultant mixture (which had sticky solid mass suspended in solvent) is diluted with ethylacetate and filtered through celite. The celite bed is washed thoroughly with ethylacetate and dichloromethane. The filtrate is evaporated to get a crude residue. The solid which remained in the celite bed is taken and triturated with ethylacetate and filtered. The filtrate is mixed together with the crude residue and evaporated. The residue thus obtained is taken in ethylacetate and washed with an aqueous solution of 1.5 N HCl and brine. The organic layer is dried over anhydrous Na2SO4and concentrated. The residue obtained is purified by flash column chromatography using 40-50% ethyl acetate in petroleum ether as eluent to get (7-Methyl-benzofuran-3-yl)-methanol (8.20 g; 48.40 mmol; 55.7%; light yellow oil). HPLC (method A): RT 3.33 min., (HPLC purity 95.7%). 1H NMR, 400 MHz, CDCl3: 7.64 (s, 1H), 7.50-7.52 (m, 1H), 7.17-7.21 (m, 1H), 7.14 (d, J=7.20 Hz, 1H), 4.86-4.86 (m, 2H), 2.54 (s, 3H). Step 3: 3-(bromomethyl)-7-methyl-benzofuran To an ice-cooled solution of (7-Methyl-benzofuran-3-yl)-methanol (8.20 g; 48.40 mmol; 1.00 eq.) in Diethyl ether (82.00 ml) under nitrogen atmosphere is added phosphorus tribromide (1.53 ml; 16.12 mmol; 0.33 eq.) drop wise and the reaction mixture is stirred at ice cold condition for 30 minutes. The reaction mixture is poured into ice and extracted with diethyl ether. The organic layer is dried over anhydrous Na2SO4and concentrated to afford 3-Bromomethyl-7-methyl-benzofuran (10.00 g; 44.43 mmol; 91.8%; colorless oil). 1H NMR, 400 MHz, CDCl3: 7.71 (s, 1H), 7.53-7.55 (m, 1H), 7.21-7.25 (m, 1H), 7.16 (d, J=7.32 Hz, 1H), 4.65 (s, 2H), 2.48 (s, 3H). Step 4: 7-Methyl-3-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-ylmethyl)-benzofuran To a solution of 3-Bromomethyl-7-methyl-benzofuran (10.00 g; 44.43 mmol; 1.00 eq.) in degased Dioxane-1,4 (100.00 ml) are added Bis(pinacolato)diboron (13.68 g; 53.31 mmol; 1.20 eq.), dried K2CO3(18.61 g; 133.28 mmol: 3.00 eq.) and tetrakis(triphenylphosphine)palladium(0) (2.57 g; 2.22 mmol; 0.05 eq.). The reaction mixture is then heated at 100° C. under nitrogen atmosphere for 16 h. The reaction mixture is diluted with dichloromethane and filtered through celite. The filtrate is concentrated. The residue is dissolved in ethyl acetate and washed with brine. The organic layer is dried over anhydrous Na2SO4and concentrated. The crude is purified by column chromatography using 2% ethyl acetate in petroleum ether to get 7-Methyl-3-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-ylmethyl)-benzofuran (5.00 g; 18.37 mmol; 41.4%; colorless liquid). 1H NMR, 400 MHz, DMSO-d6: 7.65 (s, 1H), 7.33-7.35 (m, 1H), 7.07-7.13 (m, 2H), 2.43 (s, 3H), 2.13 (s, 2H), 1.16 (s, 12H). Step 5: Trimethyl-4-(7-methyl-benzofuran-3-ylmethyl)-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane To an ice-cooled solution of 7-Methyl-3-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-ylmethyl)-benzofuran (5.00 g; 18.37 mmol; 1.00 eq.) in Et2O (50.00 ml) under nitrogen atmosphere is added 1S, 2S, 3R, 5S-(+)-2,3-pinane diol (4.69 g; 27.56 mmol; 1.50 eq.) and the reaction mixture is stirred at rt for 14 h. TLC analysis showed completion of reaction. The reaction mixture is washed with brine. The organic layer is dried over anhydrous Na2SO4and concentrated. The crude is purified by flash column chromatography using 2% ethyl acetate in petroleum ether to get (1S,2S,6R,8S)-2,9,9-Trimethyl-4-(7-methyl-benzofuran-3-ylmethyl)-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (5.00 g; 13.00 mmol; 70.7%; colorless liquid). GCMS: m/z: 324.2 1H NMR, 400 MHz, CDCl3: 7.53-7.55 (m, 1H), 7.39-7.40 (m, 1H), 7.12-7.27 (m, 1H), 7.06-7.08 (m, 1H), 4.31-4.34 (m, 1H), 2.53 (s, 3H), 2.30-2.37 (m, 1H), 2.26 (s, 2H), 2.18-2.23 (m, 1H), 2.07 (t, J=5.76 Hz, 1H), 1.84-1.93 (m, 2H), 1.42 (s, 3H), 1.29 (s, 3H), 1.12-1.15 (m, 1H), 0.85 (s, 3H). Step 6: (1S,2S,6R,8S)-4-[1-Chloro-2-(7-methyl-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane Dichloromethane (2.96 ml; 46.26 mmol; 3.00 eq.) in THE (40 mL) is taken in a RB-flask under a positive pressure of nitrogen and cooled to −95° C. using liquid nitrogen-ethanol mixture. To this n-butyl lithium (1.6 M in hexanes) (10.60 ml; 16.96 mmol; 1.10 eq.) is added drop wise through the sides of the RB-flask (at a medium rate, addition took about 30 min.) so that the internal temperature is maintained between −95° C. and −100° C. After addition, the reaction mixture is stirred for 20 minutes. During the course of the reaction a white precipitate is formed (The internal temperature is maintained between −95° C. and −100° C.). Then a solution of (1S,2S,6R,8S)-2,9,9-Trimethyl-4-(7-methyl-benzofuran-3-yl methyl)-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (5.00 g; 15.42 mmol; 1.00 eq.) in THF (20 mL) is added drop wise through the sides of the RB-flask (about 25 min) so that the internal temperature is maintained between −95° C. and −100° C. After addition, immediately zinc chloride (0.5 M in THF) (27.76 ml; 13.88 mmol; 0.90 eq.) is added drop wise through the sides of the RB-flask (at a medium rate, addition took about 45 min.) so that the internal temperature is maintained between −95° C. and −100° C. The reaction mixture is then slowly allowed to attain rt and stirred at rt for 16 h. The reaction mixture is concentrated (temperature of the bath 30° C.). The residue is partitioned between diethylether and saturated NH4Cl solution. The organic layer is separated, dried over anhydrous Na2SO4and concentrated (temperature of bath 30° C.) to afford (1S,2S,6R,8S)-4-[1-Chloro-2-(7-methyl-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (5.90 g; 15.83 mmol; 102.7%; brown liquid). 1H NMR, 400 MHz, CDCl3: 7.57 (s, 1H), 7.42-7.44 (m, 1H), 7.27 (s, 1H), 7.09-7.18 (m, 1H), 4.34-4.36 (m, 1H), 3.74-3.76 (m, 1H), 3.28-3.30 (m, 1H), 3.20-3.22 (m, 1H), 2.52 (s, 3H), 2.32-2.34 (m, 1H), 2.07 (t, J=5.88 Hz, 1H), 1.85-1.91 (m, 2H), 1.42 (s, 3H), 1.29 (s, 3H), 1.06-1.09 (m, 1H), 0.85 (s, 3H). Step 7: ((1S,2S,6R,8S)-4-[1-(1,1,1,3,3,3-Hexamethyl-disilazan-2-yl)-2-(7-methyl-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane A solution of (1S,2S,6R,8S)-4-[1-Chloro-2-(7-methyl-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (5.90 g; 15.83 mmol; 1.00 eq.) in THF (40.00 ml) under a positive pressure of nitrogen atmosphere is cooled to −78° C. To this a solution of lithium (bistrimethylsilyl)amide (1.0 M in THF) (17.41 ml; 17.41 mmol; 1.10 eq.) is added drop wise over a period of 30 minutes. The reaction mixture is allowed to attain rt and stirred at rt for 18 h. The reaction mixture is evaporated at 30° C. The residue is triturated with n-hexane and the solid formed is filtered. The filtrate is concentrated at 30° C. to get (1S,2S,6R,8S)-4-[1-(1,1,1,3,3,3-Hexamethyl-disilazan-2-yl)-2-(7-methyl-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (6.00 g; 12.06 mmol; 76.2%; brown dark oil). 1H NMR, 400 MHz, CDCl3: 7.50 (s, 1H), 7.41-7.43 (m, 1H), 7.12-7.16 (m, 1H), 7.06-7.08 (m, 1H), 4.29-4.32 (m, 1H), 3.17-3.09 (m, 1H), 2.70-2.89 (m, 1H), 2.52-2.70 (m, 1H), 2.52 (s, 3H), 2.28-2.31 (m, 1H), 2.14-2.14 (m, 1H), 2.03 (t, J=5.68 Hz, 1H), 1.78-1.89 (m, 2H), 1.39 (s, 3H), 1.31 (5, 3H), 1.01-1.04 (m, 1H), 0.90-0.92 (m, 2H), 0.88 (s, 3H), 0.12 (s, 18H). Step 8: 2-(7-Methyl-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.0206]dec-4-yl)-ethylamine Hydrochloride A stirred solution of (1S,2S,6R,8S)-4-[1-(1,1,1,3,3,3-Hexamethyl-disilazan-2-yl)-2-(7-methyl-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (6.00 g; 12.06 mmol; 1.00 eq.) in Diethyl ether (60.00 ml) under nitrogen atmosphere is cooled to −10° C. To this 2M solution of Hydrochloric acid in diethylether (15.07 ml; 30.14 mmol; 2.50 eq.) is added drop wise. The reaction mixture is stirred at rt for 2 h. The reaction mixture is evaporated at 30° C. To the residue diethyl ether (20 mL) is added and the solid formed is filtered off, washed with cold diethyl ether and dried under vacuum to get 2-(7-Methyl-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine hydrochloride (3.50 g; 8.98 mmol; 74.5%; brown orange solid). 1H NMR, 400 MHz, DMSO-d6: 8.09 (s, 3H), 7.83 (s, 1H), 7.52-7.53 (m, 1H), 7.12-7.19 (m, 2H), 4.39 (dd, J=1.84, 8.62 Hz, 1H), 3.07-3.13 (m, 1H), 3.03-3.07 (m, 2H), 2.43 (s, 4H), 2.28-2.30 (m, 1H), 2.07-2.08 (m, 1H), 1.92 (t, J=5.68 Hz, 1H), 1.82-1.84 (m, 1H), 1.71-1.75 (m, 1H), 1.19-1.25 (m, 8H), 1.00-1.08 (m, 1H), 0.78 (s, 3H). Intermediate 4: 2-(7-Chloro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.0′a]dec-4-yl)-ethylamine Hydrochloride Step 1: 7-Chloro-benzofuran-3-carboxylic Acid Ethyl Ester To a solution of 3-Chloro-2-hydroxy-benzaldehyde (25.00 g; 156.48 mmol; 1.00 eq.) in dichloromethane (250 ml), tetrafluoroboric acid diethylether complex (2.11 ml; 15.65 mmol; 0.10 eq.) is added. To the resulting dark red mixture, ethyldiazoacetate (35.55 ml; 336.44 mmol; 2.15 eq.) taken in dichloromethane (50 mL) is added dropwise slowly at 25-30° C. (internal temperature) for about 50 min. After 16 h, concentrated H2SO4is added. The reaction mixture is stirred for 15 minutes. The reaction mixture is then neutralized with solid NaHCO3, filtered through celite and the filtrate is concentrated to get a crude residue. The residue is purified by column chromatography using 2% ethyl acetate in petroleum ether to afford 7-chloro-benzofuran-3-carboxylic acid ethyl ester 2 (18.20 g; 81.02 mmol; 51.8%; colorless liquid). 1H NMR, 400 MHz, DMSO-d6: 8.88 (s, 1H), 7.95-7.93 (m, 1H), 7.57-7.55 (m, 1H), 7.42 (t, J=7.8 Hz, 1H), 4.38-4.33 (m, 2H), 1.35 (t, J=7.1 Hz, 3H). Step 2: (7-Chloro-benzofuran-3-yl)-methanol To a stirred solution of 7-chloro-benzofuran-3-carboxylic acid ethyl ester (450 g; 2.0089 mol; 1.00 eq.) in DCM (4500 ml) at −78° C. is added diisobutyl aluminium hydride 1.0 M in toluene (4017 ml; 4.0178 mol; 2.20 eq.). The reaction mixture is then slowly allowed to attain room temp. and stirred at rt for 2 h. After completion of reaction as confirmed by TLC, the reaction mixture is quenched with 1.5N HCL (500 mL), passed through celite, washed with DCM (2000 mL). The filtrate is washed with brine solution (1×2000 mL). The organic layer is separated, dried over Na2SO4, filtered and concentrated in vacuum. The crude product is subjected to column chromatography and eluted with 15% ethyl acetate in petroleum ether to afford (7-Chloro-benzofuran-3-yl)-methanol 3 (365 g; 2.0054 mol; 99.8%; white solid foam). 1H NMR, 400 MHz, DMSO-d6: 7.99 (s, 1H), 7.66 (dd, J=1.0, 7.8 Hz, 1H), 7.41 (dd, J=0.8, 7.8 Hz, 1H), 7.26 (t, J=7.8 Hz, 1H), 5.24 (t, J=5.6 Hz, 1H), 4.63-4.62 (m, 2H). Step 3: 3-Bromomethyl-7-chloro-benzofuran To an ice-cooled solution of (7-chloro-benzofuran-3-yl)-methanol (365 g; 2.0054 mol; 1.00 eq.) in diethyl ether (3650 ml) is added phosphorus tribromide (62.2 ml; 0.6618 mol; 0.33 eq.) dropwise under a nitrogen atmosphere. The reaction mixture is stirred under ice bath cooling for 30 minutes. Subsequently, the reaction mixture is poured into ice and extracted with diethyl ether. The organic layer is dried over anhydrous Na2SO4, filtered and concentrated to afford 3-bromomethyl-7-chloro-benzofuran (480 g; mol; 97.71%; white solid). 1H NMR, 400 MHz, DMSO-d6: 8.29 (s, 1H), 7.72 (dd, J=1.0, 7.8 Hz, 1H), 7.49 (dd, J=0.8, 7.8 Hz, 1H), 7.36 (t, J=7.8 Hz, 1H), 4.90 (s, 2H). Step 4: 7-Chloro-3-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-ylmethyl)-benzofuran To a solution of 3-bromomethyl-7-chloro-benzofuran (480 g; 1.9591 mol; 1.00 eq.) in degased dioxane-1,4 (4800 ml) are added bis(pinacolato)diboron (596.9 g; 2.3510 mol; 1.20 eq.), dried potassium acetate (576.8 g; 5.877 mol; 3.00 eq.) and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (70.339; 0.0979 mol; 0.05 eq.). The reaction mixture is then heated at 100° C. under nitrogen atmosphere for overnight. The reaction mixture is diluted with dichloromethane and passed through celite. The filtrate is concentrated. The residue is dissolved in ethyl acetate and washed with brine (1000 mL×1). The organic layer is dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude material is purified by column chromatography using 2% ethyl acetate in petroleum ether to obtain 7-chloro-3-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-ylmethyl)-benzofuran (480 g; 1.6438 mol; 83.9%; yellow semi solid). GCMS: m/z: 292 (Column: DB-5 ms (15 m×0.25 mm×0.25 μm); Carrier gas: helium, flow rate: 2.0 mL/min). 1H NMR, 400 MHz, DMSO-d6: 7.79 (s, 1H), 7.52 (dd, J=1.0, 7.8 Hz, 1H), 7.38 (dd, J=0.8, 7.8 Hz, 1H), 7.27-7.23 (m, 1H), 2.17 (s, 2H), 1.18 (s, 12H). Step 5: (1S,2S,6R,8S)-4-(7-Chloro-benzofuran-3-yl-methyl)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane To an ice-cooled solution of 7-chloro-3-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-ylmethyl)-benzofuran (480 g; 1.6438 mol; 1.00 eq.) in diethyl ether (5000 ml) and under a nitrogen atmosphere is added 1S, 2S, 3R, 5S-(+)-2,3-pinane diol (335.7 g; 1.9726 mol; 1.20 eq.). The reaction mixture is stirred at rt for 16 h. The reaction mixture is washed with water (2000 mL×1) and brine (1500 mL×1). The organic layer is dried over anhydrous Na2SO4, filtered and concentrated. The crude material is purified by flash chromatography using 1% ethyl acetate in petroleum ether to afford (1S,2S,6R,8S)-4-(7-chloro-benzofuran-3-yl-methyl)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (520 g; 1.510 mol; 91.9%; pale yellow semi-solid). GCMS: m/z: 344 (Column: HP-5 MS (12 m×0.20 D mm×0.33 μm); Carrier gas: helium, flow rate: 2.0 mL/min) 1H NMR, 400 MHz, DMSO-d6: 7.80 (s, 1H), 7.54 (dd, J=0.9, 7.8 Hz, 1H), 7.38 (dd, J=0.7, 7.8 Hz, 1H), 7.24 (t, J=7.8 Hz, 1H), 4.33 (t, J=6.9 Hz, 1H), 2.29-2.24 (m, 3H), 2.14-2.10 (m, 1H), 1.93 (t, J=5.4 Hz, 1H), 1.84-1.81 (m, 1H), 1.70-1.65 (m, 1H), 1.31 (s, 3H), 1.21 (s, 3H), 0.98 (d, J=10.72 Hz, 1H), 0.78 (s, 3H). Step 6: (1S,2S,6R,8S)-4-[(S)-1-Chloro-2-(7-chloro-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane Dichloromethane (95.7 ml; 1.499 mol; 3.00 eq.) in THF (1200 mL) is taken in a RB-flask under a positive pressure of nitrogen and cooled to −95° C. using liquid nitrogen-ethanol mixture. To this n-butyl lithium (1.6 M in THF) (343.6 ml; 0.549 mol; 1.10 eq.) is added dropwise through the side neck of the RB-flask (at a medium rate, addition took about 45 min.) so that the internal temperature is maintained between −95° C. and −100° C. After addition, the reaction mixture is stirred for 30 minutes. During the course of the reaction a white precipitate is formed (internal temperature is maintained between −95° C. and −100° C.). Then, a solution of (1S,2S,6R,8S)-4-(7-chloro-benzofuran-3-ylmethyl)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (172 g; 0.4999 mol; 1.00 eq.) in THF (500 mL) is added dropwise through the side neck of the RB-flask (about 25 min) so that the internal temperature is again maintained between −95° C. and −100° C. After finishing addition, Zinc chloride (0.5 M in THF) (1599.6 ml; 0.7998 mol; 1.6 eq.) is immediately added dropwise through the side neck of the RB-flask (at a medium rate, addition took about 40 min.) so that the internal temperature is maintained between −95° C. and −100° C. The reaction mixture is then slowly allowed to attain −5° C. and stirred at −5° C. for 1.5 h. The reaction mixture is quenched by adding saturated NH4Cl solution (500 mL). The reaction mixture is concentrated in vacuo (temperature of the bath 30° C.). The residue is partitioned between diethylether and saturated NH4Cl solution. The organic layer is separated, dried over anhydrous Na2SO4and concentrated (temperature of bath 30° C.) to afford (1S,2S,6R,8S)-4-[(S)-1-chloro-2-(7-chloro-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (205 g; 0.5214 mol; 104.5%; orange oil). GCMS: m/z: 392 (Column: ZB-1MS (10 m×0.101D mm×0.1 μm); Carrier gas: helium, flow rate: 2.0 mL/min) 1H NMR, 400 MHz, CDCl3: 7.64 (s, 1H), 7.50 (d, J=8.00 Hz, 1H), 7.33-7.31 (m, 1H), 7.23-7.21 (m, 1H), 4.36-4.34 (m, 1H), 3.29-3.27 (m, 1H), 3.22-3.20 (m, 1H), 2.34-2.32 (m, 1H), 2.15-2.14 (m, 1H), 2.06 (t, J=5.60 Hz, 1H), 1.91-1.83 (m, 7H), 1.36 (s, 3H), 1.29 (s, 3H), 1.05-1.02 (m, 1H), 0.85 (s, 3H). Step 7: (1S,2S,6R,8S)-4-[(R)-2-(7-Chloro-benzofuran-3-yl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bore-tricyclo[6.1.1.02,6]decane A solution of (1S,2S,6R,8S)-4-[(S)-1-chloro-2-(7-chloro-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (205 g; 0.5214 mol; 1.00 eq.) in THF (2050 ml) under a positive pressure of nitrogen atmosphere is cooled to −78° C. To this a solution of lithium (bis-trimethylsilyl)-amide (1.0 M in THF) (625 ml; 0.6257 mol; 1.20 eq.) is added dropwise over a period of 30 minutes. The reaction mixture is allowed to attain rt and stirred at rt for 18 h. The solvent of the reaction mixture is evaporated at 30° C. The residue is triturated with hexane and the solid formed is filtered. The filtrate is allowed to stand for some time under vacuum and any solid if formed is filtered again. The filtrate is concentrated at 30° C. to obtain (1S,2S,6R,8S)-4-[(R)-2-(7-chloro-benzofuran-3-yl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (180 g; 0.3481 mol; 66.7%; orange oil). 1H NMR, 400 MHz, CDCl3: 7.63 (s, 1H), 7.51-7.49 (m, 1H), 7.29-7.27 (m, 1H), 7.19-7.15 (m, 1H), 4.32-4.29 (m, 1H), 3.63-3.61 (m, 1H), 3.14-3.12 (m, 1H), 2.87-2.85 (m, 1H), 2.26-2.24 (m, 1H), 2.14-2.12 (m, 1H), 1.88-1.86 (m, 1H), 1.88-1.76 (m, 2H), 1.33 (s, 3H), 1.30 (s, 3H), 1.02-0.99 (m, 1H), 0.85 (s, 3H), 0.07 (s, 18H). Step 8: (R)-2-(7-Chloro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine Hydrochloride A stirred solution of (1S,2S,6R,8S)-4-[(R)-2-(7-chloro-benzofuran-3-yl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (180 g; 0.348 mol) in diethyl ether (1800 ml) under a nitrogen atmosphere is cooled to −10° C. To this solution, hydrochloric acid in diethyl ether (strength 2.0 M; 435.2 ml; 0.870 mol; 2.50 eq.) is added dropwise. The reaction mixture is stirred at rt for 2 h (precipitation of solid is observed during the course of the reaction). The reaction mixture is evaporated to dryness and the obtained solid is triturated with diethyl ether (500 mL) and subsequently filtered. The filter cake is washed with diethyl ether (3×300 mL) and dried under vacuum to afford (R)-2-(7-chloro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine hydrochloride (81.5 g; 0.1992 mol; 57.2%; off-white solid). 1H NMR, 400 MHz, CDCl3: 8.09 (s, 3H), 7.97 (s, 1H), 7.73 (dd, J=1.52 Hz, 7.76 Hz, 1H), 7.44 (d, J=7.76 Hz, 1H), 7.31 (d, J=7.80 Hz, 1H), 4.42-4.40 (m, 1H), 3.16-3.07 (m, 3H), 2.32-2.27 (m, 1H), 2.10-2.04 (m, 1H), 1.93 (t, J=5.56 Hz, 1H), 1.83-1.71 (m, 2H), 1.27 (s, 3H), 1.25 (s, 3H), 1.08-1.02 (m, 1H), 0.79 (s, 3H). Intermediate 5: (R)-2-(2,3-Dihydro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine Hydrochloride Step 1: (1S,2S,6R,8S)-4-(2,3-Dihydro-benzofuran-3-ylmethyl)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane To a solution of (1S,2S,6R,8S)-4-Benzofuran-3-ylmethyl-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (5.00 g; 10.72 mmol; 1.00 eq.) in methanol (100.00 ml) in a tiny clave is added palladium on carbon (10 wt %) (2.28 g; 2.14 mmol; 0.20 eq.). The contents are hydrogenated under a H2pressure of 5 Kg/cm2for 3 h. TLC analysis revealed complete conversion. The reaction mixture is filtered through celite and the filtrate is evaporated. The crude is purified by Biotage-isolera column chromatography (C18column; mobile phase: ACN/H2O; 50:50 isocratic) to get a (1S,2S,6R,8S)-4-(2,3-Dihydro-benzofuran-3-yl methyl)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (4.10 g; 13.13 mmol; 122.5%; pale yellow liquid). GCMS: m/z: 312.3. Step 2: (1S,2S,6R,8S)-4-(1-Chloro-2-(7-methyl-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane Dichloromethane (2.46 ml; 38.44 mmol; 3.00 eq.) in THF (40.00 ml) is taken in a RB-flask under a positive pressure of nitrogen and cooled to −95° C. using liquid nitrogen-ethanol mixture. To this n-butyl lithium (1.6 M in THF) (8.81 ml; 14.09 mmol; 1.10 eq.) is added drop wise through the sides of the RB-flask (at a medium rate, addition took about 20 min.) so that the internal temperature is maintained between −95° C. and −100° C. After addition, the reaction mixture is stirred for 25 minutes. During the course of the reaction a white precipitate is formed (The internal temperature is maintained between −95° C. and −100° C.). Then a solution of (1S,2S,6R,8S)-4-(2,3-Dihydro-benzofuran-3-ylmethyl)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (4.00 g; 12.81 mmol; 1.00 eq.) in THF (15.00 ml) is added drop wise through the sides of the RB-flask (about 25 min) so that the internal temperature is maintained between −95° C. and −100° C. After addition, immediately zinc chloride (0.5 M in THF) (25.62 ml; 12.81 mmol; 1.00 eq.) is added drop wise through the sides of the RB-flask (at a medium rate, addition took about 25 min.) so that the internal temperature is maintained between −95° C. and −100° C. The reaction mixture is then slowly allowed to attain rt and stirred at rt for 18 h. The reaction mixture is concentrated (temperature of the bath 30° C.). The residue is partitioned between diethylether and saturated NH4Cl solution. The organic layer is dried over anhydrous Na2SO4and concentrated (temperature of bath 30° C.) to afford (1S,2S,6R,8S)-4-[(S)-1-Chloro-2-(2,3-dihydro-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (4.60 g; 12.75 mmol; 99.5%; yellow oil). 1H NMR, 400 MHz, CDCl3: 7.29 (d, J=6.72 Hz, 1H), 7.21-7.10 (m, 1H), 6.90-6.77 (m, 2H), 4.68-4.65 (m, 1H), 4.32-4.29 (m, 2H), 3.65-3.60 (m, 1H), 2.40-2.08 (m, 4H), 1.94-1.85 (m, 2H), 1.42 (s, 3H), 1.33 (5, 3H), 1.22 (s, 3H), 1.17-1.15 (m, 1H), 0.86 (s, 3H). Step 3: (1S,2S,6R,8S)-4-[(R)-2-(2,3-Dihydro-benzofuran-3-yl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bore-tricyclo[6.1.1.02,6]decane A solution of (1S,2S,6R,8S)-4-[(S)-1-Chloro-2-(2,3-dihydro-benzofuran-3-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (4.60 g; 12.75 mmol; 1.00 eq.) in THF (45.00 ml) under a positive pressure of nitrogen atmosphere is cooled to −78° C. To this a solution of Lithium(bistrimethylsilyl)amide (1.0 M in THF) (16.58 ml; 16.58 mmol; 1.30 eq.) is added drop wise over a period of 30 minutes. The reaction mixture is allowed to attain rt and stirred at rt for 18 h. The reaction mixture is evaporated at 30° C. The residue is triturated with hexane and the solid formed is filtered. The filtrate is allowed to stand for some time under vacuum and any solid if formed is filtered again. The filtrate is concentrated at 30° C. to get (1S,2S,6R,8S)-4-[(R)-2-(2,3-Dihydro-benzofuran-3-yl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (3.77 g; 7.76 mmol; 60.9%; yellow oil). 1H NMR, 400 MHz, CDCl3: 7.22-7.10 (m, 2H), 6.90-6.79 (m, 2H), 4.62-4.59 (m, 1H), 4.33-4.27 (m, 1H), 2.34-2.20 (m, 2H), 2.07-2.05 (m, 1H), 1.94-1.84 (m, 2H), 1.40 (s, 3H), 1.30 (s, 3H), 1.15-1.13 (m, 1H), 0.86 (s, 3H), 0.10 (s, 18H). Step 4: (R)-2-(2,3-Dihydro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine Hydrochloride A stirred solution of (1S,2S,6R,8S)-4-[(R)-2-(2,3-Dihydro-benzofuran-3-yl)-1-(1,1,1,3,3,3-hexamethyl-disilazan-2-yl)-ethyl]-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]decane (3.77 g; 7.76 mmol; 1.00 eq.) in Et2O (35.00 ml) under nitrogen atmosphere is cooled to −10° C. To this 2 N HCl in diethylether (9.70 ml; 19.41 mmol; 2.50 eq.) is added drop wise. The reaction mixture is stirred at rt for 2 h. The reaction mixture is evaporated to dryness under reduced pressure to get a solid. The solid formed is triturated with diethylether, filtered, washed with diethylether and dried under vacuum get (R)-2-(2,3-Dihydro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine hydrochloride (2.30 g; 5.25 mmol; yield 67.7%; pale brown solid). Analysis showed the presence of isomers (˜65.50%+20.75%) at the indicated (*) position. LCMS: 4.73 min., 86.25% (max), 80.47% (220 nm), 342.20 (M+1). 1H NMR, 400 MHz, DMSO-d6: 8.11 (s, 3H), 7.23-7.19 (m, 1H), 7.13-7.10 (m, 1H). 6.85 (t, J=7.40 Hz, 1H), 6.77 (d, J=8.04 Hz, 1H), 4.61-4.57 (m, 1H), 4.48-4.45 (m, 1H), 4.25-4.22 (m, 1H), 3.68-3.62 (m, 1H), 2.90-2.85 (m, 1H), 2.34-2.32 (m, 1H), 2.19-2.17 (m, 1H), 2.02-1.99 (m, 2H), 1.89-1.77 (m, 3H), 1.39 (s, 3H), 1.25 (s, 3H), 1.17-1.14 (m, 1H), 0.82 (s, 3H). By similar sequences described for intermediates 2-4 other examples of the following moiety can be prepared such as in particular compounds wherein the group Y denotes one of the following groups: Acid Intermediate 1: (1S,2R,4R)-7-oxabicycio[2.2.1]heptane-2-carboxylic Acid Step 1: (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (R)-1-phenyl-ethyl Ester To a solution of 7-oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (4.680 g; 31.276 mmol, racemic) in dry dichloromethane (max. 0,005% H2O) SeccoSolv® (100 ml) under an atmosphere of argon are added (R)-1-phenyl-ethanol (4.623 ml; 37.531 mmol), 4-(dimethylamino)pyridine for synthesis (DMAP) (3.821 g; 31.276 mmol) and (3-dimethylamino-propyl)-ethyl-carbodiimide hydrochloride (EDCl) (6.730 g; 34.404 mmol) under stirring at 0° C. Subsequently, the clear reaction solution is stirred over night at room temperature. After completion of the ester formation, the reaction is quenched by adding sat. NH4Cl (aq) solution. Then, the mixture is extracted twice with CH2Cl2. The organic layer is washed trice with sat. NaHCO3(aq) and brine, dried over Na2SO4, filtrated and evaporated to dryness. The crude product is purified by flash chromatography (silica gel; n-heptane/ethyl acetate 0-30% ethyl acetate) to obtain 7.496 g (30.43 mmol, 97.3%) of a colorless oil (HPLC: 100% pure mixture of diastereomers). The mixture of diastereomers is separated by preparative, chirale HPLC (Chiralcel OD-H; n-heptane/2-propanol 95:5; 220 nm) to obtain (1R,2S,4S)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (R)-1-phenyl-ethyl ester (3.22 g, colorless oil, yield 41.8%, chiral HPLC 100%) and (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (R)-1-phenyl-ethyl ester (3.14 g, oil, yield 40.7% chiral HPLC 100%). Step 2: (1S,2R,4R)-7-oxabicyclo[2.2.1]heptane-2-carboxylic Acid To a solution of (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (R)-1-phenyl-ethyl ester (46.74 g; 182.75 mmol; 1.00 eq.) in THE (233.70 ml), palladium on carbon (10% w/w) (1.94 g; 1.83 mmol; 0.01 eq.) is added. The contents are hydrogenated under a H2atmosphere at 50° C. and 5 bar pressure for 16 h. After completion of the hydrogenation, the reaction mixture is filtered through celite, the filtrate is evaporated to dryness and taken up in pentane. The organic layer is extracted trice with water. Subsequently, the water layer is lyophilized to obtain (1S,2R,4R)-7-oxabicyclo[2.2.1]heptane-2-carboxylic acid (22.62 g; 159.09 mmol; yield 87.1%) as a colorless solid. TLC: Chloroform/methanol (9.5/0.5) Rf 0.5.1H-NMR 400 MHz, DMSO-d6: 12.16 (s, 1H), 4.66 (d, J=4.4 Hz, 1H), 4.54 (t, J=4.4 Hz, 1H), 2.57 (d, J=35.2 Hz, 1H), 1.91-1.86 (m, 1H), 1.65-1.37 (m, 4H), 1.34-1.33 (m, 1H). Optical rotation [□]20 D=+31.9°; □D20=+0.0644° (ethanol, 20.16 mg/10 ml). Acid Intermediate 2: (1R,2S,4S)-7-oxabicyclo[2.2.1]heptane-2-carboxylic Acid To a solution of (1R,2S,4S)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (R)-1-phenyl-ethyl ester (4.52 g; 17.98 mmol; 1.00 eq.) in THE (22.60 ml) Palladium on carbon (10% w/w) (0.19 g; 0.18 mmol; 0.01 eq.) is added. The contents are hydrogenated under a H2atmosphere at 50° C. for 12 h. TLC analysis revealed starting is completed. The reaction mixture is filtered through Celite and the filtrate is evaporated to get (1R,2S,4S)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (2.10 g; 14.77 mmol; 82.1%; off white solid) 1H-NMR 400 MHz, DMSO-d6: 12.16 (s, 1H), 4.66 (d, J=4.4 Hz, 1H), 4.54 (t, J=4.4 Hz, 1H), 2.57 (d, J=35.2 Hz, 1H), 1.91-1.86 (m, 1H), 1.65-1.37 (m, 4H), 1.34-1.33 (m, 1H). Acid Intermediate 3: (1S,8R)-8-Methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic Acid Lithium (Salt) Step 1: 8-Methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-1-carboxylic Acid Methyl Ester Isopentyl nitrite (3.64 ml; 27.12 mmol) is added to a solution of 3.72 g anthranilic acid (27.12 mmol) and trifluoroacetic acid (0.26 ml; 3.39 mmol) in 45 ml dried THF at 0° C. The resulting solution is stirred vigorously for a few minutes at 0° C. and then warmed up to rt. After stirring for 1 h at rt, the color of the suspension turned into yellow. The brown solid is filtered off and washed with dry THF before transferring it into a flask containing a solution of 5-methyl-furan-2-carboxylic acid methyl ester (2.00 g; 13.56 mmol) in ethylene glycol dimethyl ether for synthesis (45.00 ml). The resulting mixture is then gradually heated to 100° C. until decomposition is complete and stirred for another hour at 100° C. After evaporation of the solvent the reaction mixture is purified by flash chromatography (silica gel; EE/heptane gradient; 0-25% EE) to obtain 8-methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-1-carboxylic acid methyl ester (1.82 g; 53.7%; yellow gum) as a 1:1 mixture of stereoisomers. LCMS Method A: (M+H) 217.0; Rt 2.03 min. Step 2: (1S,8R)-8-Methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic Acid Methyl Ester and (1R,8S)-8-Methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic Acid Methyl Ester A solution 8-methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-1-carboxylic acid methyl ester (1.82 g, 7.28 mmol) in 18 ml EE is hydrogenated at rt and normal pressure using 500 mg Pd/C (54% water) until the reaction is complete. The reaction mixture is filtrated and the filtrate is concentrated. The residual sticky oil (mixture of stereoisomers) is separated using chiral preparative SFC (Chiral Cel 00-H; CO2/2-propanol 58.5: 1.5; 220 nm) to obtain (1S,8R)-8-methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic acid methyl ester (439 mg, yield 25.3%) and (1R,8S)-8-methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic acid methyl ester (449 mg, yield 25.9%), both as colourless oils. LCMS Method A: (M+H) not detected; Rt 2.09 min (same for both compounds). Step 3: (1S,8R)-8-Methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic Acid Lithium (Salt) (1S,8R)-8-Methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic acid methyl ester (0.439 g; 1.84 mmol) is taken up in 5 ml deionised water and 2.5 ml THF. LiOH (44 mg, 1.84 mmol) is added, the mixture is stirred under argon at rt for 1 h and evaporated to yield the title compound, which is used without further purification. LCMS Method A: (M−Li+H−18) 187; Rt 1.71 min. Acid Intermediate 4: (1R,8S)-8-Methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic Acid Lithium (1R,8S)-8-Methyl-11-oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-1-carboxylic acid methyl ester (purity 91%; 0.449 g; 1.88 mmol) is taken up in ml deionised water and 2.5 ml THF. LiOH (45 mg, 1.88 mmol) is added, the mixture is stirred under argon at rt for 1 h and evaporated to yield the title compound. LCMS Method A: (M−Li+H−18) 187; Rt 1.71 min. Acid Intermediate 5: (1S,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic Acid Step 1: Furan-3-carboxylic acid (R)-1-phenyl-ethyl Ester To a solution of 3-furancarboxylic acid (2.00 g; 17.84 mmol) in 40 ml dry dichloromethane are added (R)-1-phenyl-ethanol (2.64 ml; 21.41 mmol), 4-dimethylamino-pyridine (2.18 g; 17.85 mmol) and (3-dimethylamino-propyl)-ethyl-carbodiimide hydrochloride (3.84 g; 19.63 mmol) under argon-atmosphere at 0° C. The clear reaction solution is stirred without further cooling for 3 h. The reaction solution is quenched with sat. NH4Cl and extracted with dichloromethane. The organic layer is washed 3× with sat. NaHCO3solution and brine, dried over Na2SO4, and filtrated. After evaporation of the solvent the reaction mixture is purified by flash chromatography (silica gel; EE/heptane gradient; 0-30% EE) to obtain furan-3-carboxylic acid (R)-1-phenyl-ethyl ester (3.55 g; yield 90%; yellow oil). LCMS Method A: (M+H) not detected; Rt 2.46 min. Step 2: (1R,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-9-carboxylic acid (R)-1-phenyl-ethyl ester and (1S,8S)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-9-carboxylic acid (R)-1-phenyl-ethyl Ester Isopentyl nitrite (1.91 ml; 14.18 mmol) is added to a solution of anthranilic acid (1.94 g, 14.18 mmol) and trifluoroacetic acid (0.137 ml; 1.77 mmol) in 22 ml dried THE at 0° C. The resulting solution is stirred vigorously for a few minutes at 0° C. then warmed up to rt. After stirring for 1 h at rt the color of the suspension turned into yellow. The liquid is removed by decantation and the remaining brown solid is washed with dry THE before transferring it into a flask containing a solution of furan-3-carboxylic acid (R)-1-phenyl-ethyl ester (1.60 g; 7.09 mmol) in ethylene glycol dimethyl ether for synthesis (22 ml). The resulting mixture is then gradually heated to 100° C. until decomposition is complete and stirred for another hour at 100° C. After evaporation of the solvent the reaction mixture is purified by flash chromatography (silica gel; EE/heptane gradient; 0-35% EE) to obtain 677 mg 11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-9-carboxylic acid (R)-1-phenyl-ethyl ester (677 mg, colorless oil) as a mixture of diastereomers. This mixture is separated using chiral preparative HPLC (Chiral Pak AD; n-heptan/ethanol 1:1; 220 nm) to obtain (1R,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-9-carboxylic acid (R)-1-phenyl-ethyl ester (190 mg, chiral HPLC>97%; Rt 7.73 min) and (1S,8S)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-9-carboxylic acid (R)-1-phenyl-ethyl ester (180 mg; chiral HPLC>96%; Rt 12.53 min). Step 3: (1S,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic acid (R)-1-phenyl-ethyl Ester A solution (1S,8S)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-9-carboxylic acid (R)-1-phenyl-ethyl ester (0.470 g, 1.190 mmol) in 10 ml THE is hydrogenated at rt and normal pressure using 500 mg Pd/C (54% water) until the reaction is complete. The reaction mixture is filtrated, the filtrate is concentrated and purified by flash chromatography (silica gel; EE/heptane gradient; 0-60% EE) to obtain (1S,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic acid (R)-1-phenyl-ethyl ester (184 mg) as colourless oil. LCMS Method A: (M+H) not detected; Rt 2.51 min. Step 4: (1S,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic Acid A solution of (1S,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic acid (R)-1-phenyl-ethyl ester (0.184 g, 0.626 mmol) in 10 ml THF is hydrogenated at rt and normal pressure using 200 mg Pd/C (54% water) overnight. The reaction mixture is filtrated and the filtrate is evaporated to obtain 130 mg (1S,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic acid as white solid. LCMS Method A: (M−18) 174; Rt 1.42 min. Acid Intermediate 6: (1R,8S)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic Acid Step 1: (1R,8S)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic Acid (R)-1-phenyl-ethyl Ester A solution (1R,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6,9-tetraene-9-carboxylic acid (R)-1-phenyl-ethyl ester (0.470 g, purity 76%, 1.22 mmol) in 10 ml THF is hydrogenated at rt and normal pressure using 100 mg Pd/C (54% water) until the reaction is complete (5 min). The reaction mixture is filtrated, the filtrate is concentrated and purified by flash chromatography (silica gel; EE/heptane gradient; 0-50% EE) to obtain (1R,8S)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic acid (R)-1-phenyl-ethyl ester (107 mg, yield 30%) as colourless wax. LCMS Method A: (M+H) not detected; Rt 2.52 min. Step 2: (1R,8S)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic Acid A solution of (1S,8R)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic acid (R)-1-phenyl-ethyl ester (0.107 g, 0.364 mmol) in 10 ml THE is hydrogenated at rt and normal pressure using 100 mg Pd/C (54% water) overnight. The reaction mixture is filtrated and the filtrate is evaporated to obtain (1R,8S)-11-Oxa-tricyclo[6.2.1.02,7]undeca-2,4,6-triene-9-carboxylic acid (69 mg; 92% yield) as white solid. LCMS Method A: (M+H) not detected; Rt 1.36 min. Example 1: [(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic Acid (Compound No. 1) Step 1: (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic Acid [(R)-2-(S)-2,3-dihydro-benzofuran-3-yl-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-amide To a solution of (R)-2-(S)-2,3-Dihydro-benzofuran-3-yl-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine hydrochloride (0.250 g; 0.66 mmol) in 5 ml dried DMF is added at −15° C. and argon atmosphere (1S,2R,4R)-7-oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (0.113 g; 0.79 mmol), ethyl-diisopropyl-amine (0.34 ml; 1.99 mmol) and TBTU (0.70 g; 2.18 mmol). The yellow reaction solution is stirred 1 h at −10° C., than 1 h at rt. The reaction solution is cooled with ice and diluted with ethyl acetate. After separation the organic phase is washed with brine and sat. NaHCO3-solution, dried over sodium sulfate, filtered and concentrated in vacuo (bath-temp 30° C.). The obtained orange oil is first purified by flash chromatography (silica-gel; heptane/EE gradient, 0-100% EE) to yield a mixture of diastereomers, which is separated using chiral SFC (ChiralPak AD, CO2: Methanol (88:12)). 122 mg of the title compound (yield 39.6%) are obtained as colourless oil. LCMS Method A: (M+H) 466.2; Rt 2.49 min Step 2: [(1R)-2-[(3S)-2,3-dihydro-1-benzofuran-3-yl]-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic Acid (Compound No. 1) To a two phase system of (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid [(R)-2-(S)-2,3-dihydro-benzofuran-3-yl-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-am ide (0.206 g; 0.44 mmol) in 27 ml n-pentane and 20.6 ml methanol is added at 0° C. isobutylboronic acid (0.180 g; 1.77 mmol) and 2 N HCl (1.99 ml; 3.98 mmol). The reaction is stirred at rt overnight. The pentane phase is separated and the methanolic aqueous phase is washed 5× with pentane. The methanolic phase is concentrated in vacuo, diluted with ice water and alkalised with 1 N NaOH. Afterwards is extracted with DCM (2×). The aqueous phase is acidified 1 N HCl and extracted with DCM (5×). This organic phase is dried over Na2SO4, filtrated and evaporated. The residue is solved in acetonitrile/water and lyophilized to give 104 mg (yield: 71%) of the title compound as white solid. 1H NMR (500 MHz, DMSO-d6/D2O) d 7.23-7.20 (m, 1H), 7.13-7.09 (m, 1H), 6.86 (td, J=7.4, 1.0 Hz, 1H), 6.76 (d, J=7.8 Hz, 1H), 4.60 (d, J=4.7 Hz, 1H), 4.59-4.53 (m, 2H), 4.21 (dd, J=9.0, 6.7 Hz, 1H), 3.47-3.38 (m, 1H), 2.94-2.89 (m, 1H), 2.59 (dd, J=9.0, 4.9 Hz, 1H), 1.91-1.84 (m, 2H), 1.71 (dd, J=12.0, 9.1 Hz, 1H), 1.64-1.42 (m, 5H). LCMS Method A: (M+H) 314.2; Rt 1.57 min Example 2: [(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic Acid (Compound No. 9) Step 1: (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic Acid [(R)-2-(benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-amide To a solution of (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (1.87 g; 13.18 mmol), [Dimethylamino-([1,2,3]triazolo[4,5-b]pyridin-3-yloxy)-methylene]-dimethyl-ammonium hexafluoro phosphate (HATU) (4.62 g; 14.37 mmol) and 4-Methyl-morpholine (3.29 ml; 29.94 mmol) in 70 ml dry DMF is added under ice cooling and argon atmosphere (R)-2-(benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-boratricyclo[6.1.1.02,6]dec-4-yl)-ethylamine hydrochloride (4.50 g; 11.98 mmol). The yellow solution is stirred for 2.5 h at rt. The reaction mixture is poured into 500 ml ice cooled, saturated NaHCO3-solution and stirred for 15 min. The precipitate is collected by vacuum-filtration and washed with water. The obtained solid is triturated with acetonitrile, diluted with MTB-ether, and sucked off to yield (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid [(R)-2-(benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-amide (3.26 g, yield: 58.8%) as white solid (purity 100%). LCMS Method A: (M+H) 464.2; Rt 2.57 min Step 2: [(1R)-2-(1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic Acid To a two phase system of (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid [(R)-2-benzofuran-3-yl-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-amide (ee=97%, 3.45 mmol; 1.60 g) in 150 ml n-pentane and 50 ml methanol is added at 0° C. isobutylboronic acid (13.81 mmol; 1.41 g) and 1N Hydrochloric acid (15.54 mmol; 15.54 ml). The reaction is stirred at rt overnight. The pentane phase is separated and the methanolic phase is washed with pentane (3×80 mL). The methanolic phase is concentrated (bath temp below 30° C.) in vacuo, diluted with ice water and alkalized with 1 N NaOH (pH 11-12). This basic solution is extracted with DCM (3×80 mL). The aqueous phase is acidified with 1 N HCl (pH 2) and extracted with DCM (5×80 mL) again. The combined organic phases are dried over Na2SO4, filtrated and evaporated. The residue is solved in acetonitrile/water and lyophilized to give 0.697 g (yield 61.3%) of the title compound as white solid. Analytical data: see Table 2 Example 3: [(1R)-2-(7-chloro-1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic Acid (Compound No. 13) Step 1: (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic Acid [(R)-2-(7-chloro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.026]dec-4-yl)-ethyl]-amide To a solution of (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid (3.77 g; 26.55 mmol), [Dimethylamino-([1,2,3]triazolo[4,5-b]pyridin-3-yloxy)-methylene]-dimethyl-ammonium hexafluoro phosphate (HATU) (9.30 g; 28.97 mmol) and 4-Methyl-morpholine (6.63 ml; 60.34 mmol) in dry 148 ml DMF is added under ice cooling and argon atmosphere (R)-2-(7-Chloro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethylamine hydrochloride (9.90 g; 24.14 mmol). The yellow solution is stirred for 3 h at rt. The reaction mixture is poured into 1 l ice cooled, saturated NaHCO3-solution and stirred for 15 min. The precipitate is collected by vacuum-filtration and washed with water. The obtained solid is triturated with acetonitrile, diluted with MTB-ether, and sucked off to yield (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid [(R)-2-(7-chloro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-amide (6.9 g, yield: 56.6%) as white solid (purity 95%). LCMS Method A: (M+H) 498.2; Rt 2.70 min Step 2: [(1R)-2-(7-chloro-1-benzofuran-3-yl)-1-{[(1S,2R,4R)-7-oxabicyclo[2.2.1]heptan-2-yl]formamido}ethyl]boronic Acid (Compound No. 13) To a two phase system of (1S,2R,4R)-7-Oxa-bicyclo[2.2.1]heptane-2-carboxylic acid [(R)-2-(7-chloro-benzofuran-3-yl)-1-((1S,2S,6R,8S)-2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-amide (ee=99%, 12.39 mmol; 6.26 g) in 220 ml n-pentane and 125 ml methanol is added at 0° C. isobutylboronic acid (37.17 mmol; 3.79 g) and 1N Hydrochloric acid (55.760 mmol; 55.760 ml). The reaction is stirred at rt overnight. The pentane phase is separated and the methanolic phase is washed with pentane (3×200 mL). The methanolic phase is concentrated (bath temp below 30° C.) in vacuo, diluted with 200 mL ice water and alkalized with 1 N NaOH (pH 12-13). This basic solution is extracted with DCM (3×200 mL). The aqueous phase is acidified with 1 N HCl (pH 2-3) and extracted with DCM (5×220 mL) again. The combined organic phases are dried over Na2SO4, filtrated and evaporated. The residue is solved in acetonitrile/water and lyophilized to give 3.277 g of the title compound as white solid. 1H NMR (500 MHz, DMSO-d6/D2O) d 7.78 (s, 1H), 7.64-7.60 (m, 1H), 7.42-7.39 (m, 1H), 7.29 (t, J=7.8 Hz, 1H), 4.54-4.50 (m, 1H), 4.48-4.45 (m, 1H), 3.15-3.09 (m, 1H), 2.89 (dd, J=14.9, 5.8 Hz, 1H), 2.77 (dd, J=14.9, 8.4 Hz, 1H), 2.50 (dd, J=9.0, 4.9 Hz, 1H), 1.82-1.75 (m, 1H), 1.65 (dd, J=11.9, 9.0 Hz, 1H), 1.58-1.49 (m, 2H), 1.49-1.39 (m, 2H). Waters XBridge C8 3.5 μm 4.6×50 mm (A19/533—La Chrom Elite; 70173815); 8.1 min; 2 mL/min; 215 nm; buffer A: 0.05% TFA/H2O; buffer B: 0.04% TFA/ACN; 0.0-0.2 min 5% buffer B; 0.2-8.1 min 5%-100% buffer B; 8.1-10.0 min 100%-5% buffer B: %; Rt 4.24 min. UPLC MS Waters Acquity UPLC; CORTECS C18 1.6 μm 50-2.1 mm; A: H2O+0.05% HCOOH; B: MeCN+0.04% HCOOH; T: 30° C.; Flow: 0.9 ml/min; 2%≥ 100% B: 0≥1.0 min; 100% B: 1.0-≥1.3 min: 346.1 [M+H−H2O]; RT 0.61 min. Starting from commercial available acids (or acids synthetised by saponification of commercial available esters) or acids described in the literature the following compounds have been synthetised according to example 1 or 2 steps 1 and 2: TABLE 1List of exemplary compoundsCom-poundNo.StructureName1[(1R)-2-[(3S)- 2,3-dihydro-1- benzofuran-3- yl]-1-{[(1S,2R, 4R)-7-oxabi- cyclo[2.2.1] heptan-2-yl] formamido} ethyl]boronic acid2[(1R)-2-[(3S)- 2,3-dihydro-1- benzofuran-3- yl]-1-{[(1R,2S, 4S)-7-oxabi- cyclo[2.2.1] heptan-2-yl] formamido} ethyl]boronic acid3[(1R)-1- {[(1S,2R,4R)- 7-oxabicyclo [2.2.1]heptan- 2-yl]forma- mido}-2- (thiophen-3- yl)ethyl] boronic acid4[(1R)-2-(1- benzofuran- 3-yl)-1- {[(1R,8S)-11- oxatricyclo [6.2.1.02,7] undeca-2(7), 3,5-trien-1- yl]formamido} ethyl]boronic acid5[(1S)-2-(1- benzofuran- 3-yl)-1- {[(1R,8S)-11- oxatricyclo [6.2.1,02,7] undeca-2(7), 3,5-trien-1-yl] formamido} ethyl]boronic acid6[(1R)-2-(1- benzofuran- 3-yl)-1- {[(1S,8R)-11- oxatricyclo [6.2.1.02,7] undeca-2(7), 3,5-trien-1-yl] formamido} ethyl]boronic acid7[(1S)-2-(1- benzofuran- 3-yl)-1- {[(1S,8R)-11- oxatricyclo [6.2.1.02,7] undeca-2(7), 3,5-trien-1-yl] formamido} ethyl]boronic acid8[(1R)-2-(1- benzofuran- 3-yl)-1- {[(1R,2S,4S)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid9[(1R)-2-(1- benzofuran- 3-yl)-1- {[(1S,2R,4R)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid10[(1R)-2-(1- benzofuran- 3-yl)-1- {[(1R,2R,4S)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid11[(1S)-2-(1- benzofuran- 3-yl)-1- {[(1R,2R,4S)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid12[(1R)-2-(7- chloro-1- benzofuran- 3-yl)-1- {[(1R,2S,4S)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid13[(1R)-2-(7- chloro-1- benzofuran- 3-yl)-1- {[(1S,2R,4R)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid14[(1R)-2- [(3R)-7- methyl-2,3- dihydro-1- benzofuran- 3-yl]-1- {[(1S,2R,4R)- 7-oxabicycio [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid15[(1R)-2- [(3S)-7- methyl-2,3- dihydro-1- benzofuran- 3-yl]-1- {[(1S,2R,4R)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid16[(1R)-2- [(3S)-2,3- dihydro-1- benzofuran- 3-yl]-1- {[(1R,8S)-11- oxatricyclo [6.2.1.02,7] undeca-2(7), 3,5-trien-1- yl] formamido} ethyl]boronic acid17[(1R)-2-(1- benzofuran- 3-yl)-1- {[(1S,6S,7R)- 3-cyclopropyl- 4-oxo-10- oxa-3- azatricyclo [5.2.1.01,5] dec-8-en-6- yl]formamido} ethyl]boronic acid18[(1R)-2-[(3S)- 2,3-dihydro- 1-benzofuran- 3-yl]-1- {[(1S,8R)-11- oxatricyclo [6.2.1.02,7] undeca-2(7), 3,5-trien-1- yl]formamido} ethyl]boronic acid19[(1R)-2-(7- methyl-1- benzofuran- 3-yl)-1- {[(1R,8S)-11- oxatricyclo [6.2.1.02,7] undeca-2,4,6- trien-1- yl]formamido} ethyl]boronic acid20[(1R)-2-(7- methyl-1- benzofuran- 3-yl)-1- {[(1S,8R)-11- oxatricyclo [6.2.1.02,7] undeca-2,4,6- trien-1-yl] formamido} ethyl]boronic acid21[(1R)-2- [(3S)-2,3- dihydro-1- benzofuran- 3-yl]-1- {[(1S,8R)-8- methyl-11- oxatricyclo [6.2.1.02,7] undeca-2,4,6- trien-1-yl] formamido} ethyl]boronic acid22[(1R)-2-(1- benzofuran- 3-yl)-1- {[(1R,8S)-11- oxatricyclo [6.2,1.02,7] undeca-2(7), 3,5-trien-9- yl]formamido} ethyl]boronic acid23[(1R)-2- [(3S)-2,3- dihydro-1- benzofuran- 3-yl]-1- {[(1R,8S)-8- methyl-11- oxatricyclo [6.2.1.02,7] undeca-2,4,6- trien-1-yl] formamido} ethyl]boronic acid24[(1R)-2-(1- benzofuran- 3-yl)-1- {[(1S,8R)-11- oxatricyclo [6.2.1.02,7] undeca-2(7), 3,5-trien-9- yl]formamido} ethyl]boronic acid25[(1R)-2-(2,4- dimethyl- phenyl)-1- {[(1S,2R,4R)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} ethyl]boronic acid26[(1R)-2- cyclohexyl- 1-{[(1S,2R, 4R)-7- oxabicyclo [2.2.1] heptan-2- yl] formamido} ethyl]boronic acid27[(1R)-1- {[(1S,2R, 4R)-7- oxabicyclo [2.2.1]heptan- 2-yl] formamido}- 3-phenyl- propyl] boronic acid28[(1R)-3- methyl-1- {[(1S,2R,4R)- 7-oxabicyclo [2.2.1]heptan- 2-yl] formamido} butyl]boronic acid TABLE 2Analytical dataRET;observedMass;Compound(LCMSNo.Method)NMR Signals13.4 min;1H NMR (500 MHz, DMSO-d6/D2O) d314.18;7.23-7.20 (m, 1H), 7.13-7.09 (m, 1H),(Method B)6.86 (td, J = 7.4, 1.0 Hz, 1H), 6.76 (d. J =7.8 Hz, 1H), 4.60 (d, J = 4.7 Hz, 1H), 4.59-4.53 (m, 2H), 4.21 (dd, J = 9.0, 6.7 Hz,1H), 3.47-3.38 (m, 1H), 2.94-2.89 (m,1H), 2.59 (dd, J = 9.0, 4.9 Hz, 1H), 1.91-1.84 (m, 2H), 1.71 (dd, J = 12.0, 9.1 Hz,1H), 1.64-1.42 (m, 5H).23.4 min;1H NMR (400 MHz, DMSO-d6/D2O) d314.18;7.16 (d, J = 7.4 Hz, 1H), 7.09-7.03 (m,(Method B)1H), 6.81 (td, J = 74, 1.0 Hz, 1H), 6.70(d, J = 7.9 Hz, 1H), 4.56 (d, J = 4.4 Hz,1H), 4.54-4.48 (m, 2H), 4.15 (dd, J =9.0, 6.7 Hz 1H), 3.41-3.32 (m, 1H),2.83 (dd, J = 9.8, 5.3 Hz, 1H), 2.55-2.51(m, 1H), 1.86-1.76 (m, 2H), 1.67 (dd, J =12.0, 9.1 Hz, 1H), 1.60-1.37 (m, 5H).32.46 min;1H NMR (400 MHz, DMSO-d6/D2O) d278.16;7.37-7.33 (m, 1H), 7.06-7.03 (m, 1H),(Method B)6.92 (d, J = 5.0 Hz, 1H), 4.51-4.40 (m,2H), 3.08-2.99 (m, 1H), 2.83-2.74 (m,H), 2.69 (dd, J = 14.5, 8.2 Hz, 1H), 2.49-2.43 (m, 1H), 1.78-1.69 (m, 1H), 1.66-1.57 (m, 1H), 1.53-1.36 (m, 4H).4 + 55.42 min;—(mixture of360.2;diastereomers,(Method B)2:1 R:S atC*-B(OH)2)6 + 75.26 min;—(mixture of360.2;diastereomers,(Method B)2:1 R:Sat C*-B(OH)2)83.38 min;1H NMR (400 MHz, DMSO-d6/D2O) d312.16;7.60 (s, 1H), 7.58 (d, J = 7.5 Hz, 1H), 7.47(Method B)(d, J = 8.0 Hz, 1H), 7.27 (t, J = 7.6 Hz,1H), 7.21 (t, J = 7.4 Hz, 1H), 4.50-4.43(m, 2H), 3.11-3.05 (m, 1H), 2.84 (dd, J =14.9, 5.9 Hz, 1H), 2.73 (dd, J = 14.9, 8.1Hz, 1H), 2.45 (dd, J = 9.0, 5.0 Hz, 1H),1.77-1.69 (m, 1H), 1.61 (dd, J = 12.0,9.0 Hz, 1H), 1.53-1.44 (m, 2H), 1.44-1.34 (m, 2H).93.39 min;1H NMR (500 MHz, DMSO-d6/D2O) d312.16;7.61 (s, 1H), 7.59 (d, J = 7.7 Hz, 1H), 7.48(Method B)(d, J = 8.1 Hz, 1H), 7.29-7.25 (m, 1H),7.24-7.19 (m, 1H), 4.48-4.45 (m, 1H),4.42-4.40 (m, 1H), 3.12-3.08 (m, 1H),2.84 (dd, J = 14.9, 5.9 Hz, 1H), 2.73 (dd,J = 14.9, 8.3 Hz, 1H), 2.45 (dd, J = 9.1,4.9 Hz, 1H), 1.76-1.71 (m, 1H), 1.60(dd, J = 11.9, 9.1 Hz, 1H), 1.52-1.44 (m,2H), 1.43-1.34 (m, 2H),10 + 113.59 min;—(mixture of312.16;diastereomers,(Method B)2:1 R:Sat C*-B(OH)2)124.25 min;1H NMR (500 MHz, DMSO-d6/D2O) d346.6;7.78 (s 1H) 7.63-7.60 (m, 1H), 7.42-(Method B)7.39 (m, 1H), 7.29 (t, J = 7.8 Hz, 1H),4.56-4.48 (m, 2H), 3.13-3.07 (m, 1H),2.88 (dd, J = 14.7, 5.8 Hz, 1H), 2.78 (dd,J = 14.9, 8.3 Hz, 1H), 2.53-2.48 (m, 1H),1.81-1.74 (m, 1H), 1.66 (dd, J = 11.7,9.3 Hz, 1H), 1.59-1.50 (m, 2H), 1.49-1.40 (m, 2H).134.25 min;1H NMR (500 MHz, DMSO-d6/D2O) d346.6;7.78 (s, 1H), 7.64-7.60 (m, 1H), 7.42-(Method B)7.39 (m, 1H), 7.29 (1, J = 7.8 Hz, 1H),4.54-4.50 (m, 1H), 4.48-4.45 (rn, 1H),3.15-3.09 (m, 1H), 2.89 (dd, J = 14.9,5.8 Hz, 1H), 2.77 (dd, J = 14.9, 8.4 Hz.1H), 2.50 (dd, J = 9.0, 4.9 Hz, 1H), 1.82-1.75 (m, 1H), 1.65 (dd, J = 11.9, 9.0 Hz,1H), 1.58-1.49 (m, 2H), 1.49-1.39 (m,2H).143.8 min;1H NMR (400 MHz, DMSO-d6/D2O) d328.2;7.02 (d, J = 7.3 Hz, 1H), 6.89 (d, J = 7.5(Method B)Hz, 1H), 6.71 (t, J = 7.4 Hz, 1H), 4.58-4.49 (m, 3H), 4.09 (dd, J = 8.9, 6.6 Hz,1H), 3.41-3.30 (m, 1H), 2.81 (t, J = 7.6Hz, 1H), 2.57-2.50 (m, 1H), 2.07 (s, 3H),1.90-1.77 (m, 2H), 1.65 (dd, J = 12.0,9.1 Hz, 1H), 1.61-1.36 (m, 5H).153.8 min;1H NMR (400 MHz, DMSOd6/D2O) d328.2;6.97 (d, J = 7.3 Hz, 1H), 6.89 (d, J = 7.4(Method B)Hz, 1H), 6.72 (t. J = 7.4 Hz, 1H), 4.58-4.48 (m, 3H), 4.17 (dd, J = 9.0, 6.8 Hz,1H), 3.41-3.32(m, 1H), 2.99 (dd, J =10.2, 4.7 Hz, 1H), 2.64-2.50 (m, 1H),2.08 (s, 3H), 1.93-1.79 (m, 2H), 1.64-1.37 (m, 6H).165.0 min;1H NMR (400 MHz, DMSO-d6/D2O) d362.22,7.34-7.29 (m, 2H), 7.21-7.13 (m, 3H),(Method B)7.09-7.04 (m, 1H), 6.82 (td, J = 7.4, 1.0Hz, 1H), 6.71 (d, J = 7.9 Hz, 1H), 5.52 (d,J = 4.9 Hz, 1H), 4.57 (1, J = 8.9 Hz, 1H),4.21 (dd, J = 9.1, 6.9 Hz, 1H), 3.39-3.29(m, 2H), 2.19-2.08 (m, 1H), 2.01-1.87(m, 2H), 1.70-1.55(m, 2H), 1.37-1.29(m, 1H).174.02 min;1H NMR (400 MHz, DMSO-d6/D2O) d405.24;7.65-7.57 (m, 2H), 7.47 (d, J = 8.0 Hz,(Method B)1H), 7.30-7.24 (m, 1H), 7.24-7.19 (m,I H), 6.51 (dd, J = 6.8, 5.7 Hz, 1H), 6.36-6.32 (m, 1H), 4.85-4.75 (m, 1H), 3.91(dd, J = 12.0, 9.7 Hz, 1H), 3.42 (dd, J =12.0, 5.0 Hz, 1H), 3.17-3.07 (m, 1H),2.95-2.83 (m, 1H), 2.83-2.70 (m, 2H),2.57-2.52 (m, 1H), 2.44 (dd, J = 13.4,9.2 Hz, 1H), 0.65-0.46 (m, 4H).185.1 min;1H NMR (400 MHz, DMSO-d6/D2O) d362.22;7.33-7.27 (m, 2H), 7.17 (td, J = 7.3, 1.4(Method B)Hz, 1H), 7.13 (td, J = 7.4, 1.4 Hz, 1H),7.08-7.01 (m, 2H), 6.76 (t, J = 7.4 Hz,1H), 6.69 (d, J = 7.8 Hz, 1H), 5.53 (d, J =5.0 Hz, 1H), 4.55 (t, J = 9.0 Hz, 1H), 4.20(dd, J = 9.1, 6.9 Hz, 1H), 3.43 (dd, J =10.5, 4.1 Hz, 1H), 3.32-3.22 (m, 1H),2.19-2.08 (m, 1H). 1.98-1.84 (m, 2H),1.69-1.53 (m, 2H), 1.38-1.28 (m, 1H).195.44 min;1H NMR (500 MHz, DMSO-d6) d 7.61 (s,374.23;1H), 7.48-7.44 (m, 1H), 7.35-7.31 (m,(Method B)2H), 7.22 (td, J = 7.4, 1.4 Hz, 1H), 7.19(td, J = 7.4, 1.3 Hz, 1H), 7.16-7.11 (m,2H), 5.50 (d, J = 5.0 Hz, 1H), 3.58 (dd,J = 7.3, 5.5 Hz, 1H), 3.08 (dd, J = 14.8, 5.4Hz, 1H), 2.93 (dd, J = 14.8, 7,4 Hz, 1H),2.45 (s, 3H), 2.14-2.07 (m, 1H), 1.80 (td,J = 11.0, 3.9 Hz, 1H), 1.60-1.53 (m, 1H),1.38-1.32 (m, 1H).205.41 min;1H NMR (400 MHz, DMSO-d6) d 7.46 (s,374.23;1H), 7.36-7.32 (m, 2H), 7.27-7.16 (m,(Method B)3H), 7.06 (d, J = 7.3 Hz, 1H), 6.93 (t, J =7.5 Hz, 1H), 5.50 (d, J = 5.0 Hz, 1H), 3.68-3.63 (m, 1H), 3.07 (dd, J = 14.9, 5.5 Hz,1H), 2.91 (dd, J = 14.9, 7.1 Hz, 1H), 2.40(s, 3H), 2.19-2.10 (m, 1H), 1,94 (td, J =11.1, 3.8 Hz, 1H), 1.65-1.57 (m, 1H),1.40-1.32 (m, 1H).215.3 min;1H NMR (400 MHz, DMSO-d6/D2O) d376.25;7.29-7.23 (m, 2H), 7.20 (td, J = 7.3, 1.2(Method B)Hz, 1H) 7.14 (td, J = 7.3, 1.4 Hz, 1H),7.09-7.01 (m, 2H), 6.76 (td, J = 7.4, 1.0Hz, 1H), 6.69 (d, J = 7.9 Hz, 1H), 4.57 (t,J = 9.0 Hz, 1H), 4.22 (dd, J = 9.2, 6.9 Hz,H), 3.43 (dd, J = 10.6, 4.1 Hz, 1H), 3.33-3.22 (m, 1H), 2.11-2.02 (m, 1H), 1.94-1.84 (m, 2H), 1.80 (s, 3H), 1.70-1.58(m, 2H), 1.46-1.38 (m, 1H).224.23 min,1H NMR (400 MHz, DMSO-d6) d 7.52-360.2;7.45 (m, 2H), 7.42 (s, 1H), 7.30-7.18 (m,(Method B)3H), 7.11-7.06 (m, 1H), 7.03-6.99 (m,1H), 6.97-6.91 (m, 1H), 5.45 (d, J = 5.2Hz, 1H), 5.38-5.32 (m, 1H), 3.21-3.14(m, 1H), 3.09-3.03 (m, 1H), 2.70 (dd, J =15.4, 6.0 Hz, 1H), 2.61 (dd, J = 14.9, 8.0Hz, 1H), 2.09 (td, J = 11.4, 5.1 Hz, 1H),1.52 (dd, J = 11.7, 4.2 Hz, 1H).235.3 min;1H NMR (400 MHz, DMSO-d6/D2O d 7.33-376.25;7.30 (m, 1H), 7.27-7.13 (m, 4H), 7.10-(Method B)7.04 (m, 1H), 6.82 (1d, J = 7.4, 1.0 Hz,1H), 6.71 (d, J = 7.9 Hz, 1H), 4.58 (t, J =8.9 Hz, 1H), 4.23 (dd, J = 9.1, 6.9 Hz,1H), 3.41-3.31 (m, 2H), 2.07 (td, J =10.8, 3.8 Hz, 1H), 2.02-1.85 (m, 2H),1.80 (s, 3H), 1.72-1.60 (m, 2H), 1.47-1.38 (m, 1H).244.31 min;1H NMR (400 MHz, DMSO-d6/D2O) d360.2;7.63 (d, J = 7.6 Hz, 1H), 7.59-7.55 (m,(Method B)2H), 7.38-7.32 (m, 1H), 7.31-7.22 (m,2H), 7.07 (t, J = 7.4 Hz, 1H), 6.75 (t, J =7.4 Hz, 1H), 6.51 (d, J = 7.3 Hz, 1H), 5.40-5.36 (m, 2H), 3.25-3.18 (m, 1H), 3.07(dd, J = 9.3, 5.4 Hz, 1H), 2.82 (dd, J =14.8, 5.5 Hz, 1H), 2.66 (dd, J = 14.9, 9.2Hz, 1H), 2.14-2.06 (m, 1H), 1.59 (dd,J = 11.6, 4.3 Hz, 1H).253.87 min;1H NMR (400 MHz, DMSO-d6) d 7.34 (d,300.19;J = 5.9 Hz, 1H), 6.94 (d, J = 7.7 Hz, 1H),(Method B)6.91-6.87 (m, 1H), 6.87-6.82 (m, 1H),4.48-4.44 (m, 1H), 4.41 (d, J = 3.9 Hz,1H), 3.05-2.97 (m, 1H), 2.71 (dd, J =14.1,6.1 Hz, 1H), 2.59 (dd, J = 14.2,8.9Hz, 1H), 2.41 (dd, J = 9.1, 4.9 Hz, 1H),2.18 (s, 6H), 1.76-1.68 (m, 1H), 1.58(dd, J = 11.9, 9.0 Hz, 1H), 1.53-1.34 (m,4H).263.78 min;1H NMR (400 MHz, DMSO-d6+ 4-5 drops278.19;D2O) d 4.66-4.40 (m, 2H), 2.91 (t, J =(Method B)7.4 Hz, 1H), 2.51 (dd, J = 9.1, 5.0 Hz,1H), 1.91-1.78 (m, 1H), 1.76-1.41 (m,10H), 1.39-1.02 (m, 6H), 0.95-0.69 (m,2H).273.51 min;1H NMR (400 MHz, DMSO-d6) d 7.34-286.17;7.24 (m, 2H), 7.24-7.13 (m, 3H), 4,64-(Method B)4.49 (m, 2H), 2.82 (t, J = 7.0 Hz, 1H),2.66-2.48 (m, 3H), 1.97-1.82 (m, 1H),1.82-1.36 (m, 7H).282.64 min;1H NMR (400 MHz, DMSO-d6+ 4-5 drops238.12;D2O) d 4.58-4.37 (m, 21-1), 2.88 (dd, J =(Method B)9.1, 6.0 Hz, 1H), 2.45 (dd, J = 9.0, 4.9 Hz,1H), 1.90-1.73 (m, 1H), 1.60 (dd, J =11.8, 9.1 Hz, 1H), 1.56-1.16 (m, 7H),0.90-0.68 (m, 6H). Biological Activity Determination of LMP7 Activity: Measurement of LMP7 inhibition is performed in 384 well format based on fluorescence intensity assay. Purified human immuno proteasome (0.25 nM) and serial diluted compounds in DMSO (range of concentrations from 30 μM to 15 pM) or controls are incubated for 20 minutes or 120 minutes (long incubation) at 25° C. in assay buffer containing 50 mM Tris pH 7.4, 0.03% SDS, 1 mM EDTA and 1% DMSO. The reaction is initiated by the addition of the fluorogenic peptide substrate, Suc-LLVY-AMC (Bachem 1-1395), at a concentration of 40 μM. After 60 minutes of incubation at 37° C., fluorescence intensity is measured at λex=350 nm and λem=450 nm with a fluorescence reader (Perkin Elmer Envision reader or equivalent). The LMP7 activity of the compounds is summarized in Table 3. Unless indicated otherwise the results are obtained after incubation for 20 minutes. Determination of Beta5 Activity: Measurement of Beta5 inhibition is performed in 384 well format based on fluorescence intensity assay. Purified human constitutive proteasome (1.25 nM) and serial diluted compounds in DMSO (range of concentrations from 30 μM to 15 pM) or controls are incubated for 20 minutes or 120 minutes (long incubation) at 25° C. in assay buffer containing 50 mM Tris pH 7.4, 0.03% SDS, 1 mM EDTA and 1% DMSO. The reaction is initiated by the addition of the fluorogenic peptide substrate, Suc-LLVY-AMC (Bachem 1-1395), at a concentration of 40 μM. After 60 minutes of incubation at 37° C., fluorescence intensity is measured at λex=350 nm and λem=450 nm with a fluorescence reader (Perkin Elmer Envision reader or equivalent). Table 3 shows the Beta5 activity of compounds according to the invention and their selectivity to LMP7 versus Beta5. Unless indicated otherwise the results are obtained after incubation for 20 minutes. TABLE 3Beta5SelectivityLMP7IC50LMP7 vsCompound No.IC50 (M)(M)Beta51*****+++++2****+++3**n. m. a.−4 + 5******+++++(mixture ofdiastereomers,2:1 R:S at C*-B(OH)2)6 + 7*****+++mixture ofdiastereomers,2:1 R:S at C*-B(OH)2)8*****++++9******+++++10 + 11***n. m. a.−mixture ofdiastereomers,2:1 R:S at C*-B(OH)2)12*****+++++13******+++++14**n. m. a.−15*****+++++16*****+++++17*****++18*****+++++19*****+++++20*****++++21*****+++++22*****++23*****+++++24*******+25****++26**n. m. a.−(long)(long)27**n. m. a.−28**n. m. a.−*: 5 μM < IC50≤ 3.0*10−5M,**: 0.5 μM < IC50≤ 5 μM,***: 0.05 μM < IC50≤ 0.5 μM,****: IC50< 0.05 μM,+: Selectivity <100,++: 100 ≤ Selectivity < 300,+++: 300 ≤ Selectivity < 500,++++: 500 ≤ Selectivity < 700,+++++: Selectivity ≥700,n. m. a.: no measurable the given concentration range; in accordance with the method described above, “long incubation” means that the sample is incubated for 120 min. The following examples relate to medicaments: Example A: Injection Vials A solution of 100 g of an active ingredient of the formula (I) and 5 g of disodium hydrogenphosphate in 3 l of bidistilled water is adjusted to pH 6.5 using 2 N hydrochloric acid, sterile filtered, transferred into injection vials, lyophilised under sterile conditions and sealed under sterile conditions. Each injection vial contains 5 mg of active ingredient. Example B: Suppositories A mixture of 20 g of an active ingredient of the formula (I) with 100 g of soya lecithin and 1400 g of cocoa butter is melted, poured into moulds and allowed to cool. Each suppository contains 20 mg of active ingredient. Example C: Solution A solution is prepared from 1 g of an active ingredient of the formula I, 9.38 g of NaH2PO42H2O, 28.48 g of Na2HPO4. 12 H2O and 0.1 g of benzalkonium chloride in 940 ml of bidistilled water. The pH is adjusted to 6.8, and the solution is made up to 1 l and sterilised by irradiation. This solution can be used in the form of eye drops. Example D: Ointment 500 mg of an active ingredient of the formula (I) are mixed with 99.5 g of Vaseline under aseptic conditions. Example E: Tablets A mixture of 1 kg of active ingredient of the formula I, 4 kg of lactose, 1.2 kg of potato starch, 0.2 kg of talc and 0.1 kg of magnesium stearate is pressed in a conventional manner to give tablets in such a way that each tablet contains 10 mg of active ingredient. Example F: Dragees Tablets are pressed analogously to Example E and subsequently coated in a conventional manner with a coating of sucrose, potato starch, talc, tragacanth and dye. Example G: Capsules 2 kg of active ingredient of the formula (I) are introduced into hard gelatine capsules in a conventional manner in such a way that each capsule contains 20 mg of the active ingredient. Example H: Ampoules A solution of 1 kg of active ingredient of the formula (I) in 60 l of bidistilled water is sterile filtered, transferred into ampoules, lyophilised under sterile conditions and sealed under sterile conditions. Each ampoule contains 10 mg of active ingredient.
189,813
11858952
DETAILED DESCRIPTION OF THE INVENTION Hereinafter, the present application will be described in more detail. One embodiment of the present specification provides a compound represented by Chemical Formula 1. In the present specification, a certain part “including” certain constituents means capable of further including other constituents, and does not exclude other constituents unless particularly stated on the contrary. In the present specification, one member being placed “on” another member includes not only a case of the one member being in contact with the another member but a case of still another member being present between the two members. Examples of substituents in the present specification are described below, however, the substituents are not limited thereto. The term “substitution” means a hydrogen atom bonding to a carbon atom of a compound is changed to another substituent, and the position of substitution is not limited as long as it is a position at which the hydrogen atom is substituted, that is, a position at which a substituent can substitute, and when two or more substituents substitute, the two or more substituents may be the same as or different from each other. The term “substituted or unsubstituted” in the present specification means being substituted with one, two or more substituents selected from the group consisting of hydrogen; deuterium; a halogen group; a cyano group; a nitro group; a carbonyl group; an imide group; an amide group; an ester group; a hydroxyl group; an amine group; a substituted or unsubstituted alkyl group; a substituted or unsubstituted cycloalkyl group; a substituted or unsubstituted alkoxy group; a substituted or unsubstituted aryloxy group; a substituted or unsubstituted alkenyl group; a substituted or unsubstituted alkynyl group; a substituted or unsubstituted aryl group; a substituted or unsubstituted alkylthioxy group; a substituted or unsubstituted arylthioxy group; a substituted or unsubstituted alkylsulfoxy group; and a substituted or unsubstituted heteroaryl group, or being substituted with a substituent linking two or more substituents among the substituents illustrated above, or having no substituents. For example, “a substituent linking two or more substituents” may include a biphenyl group. In other words, a biphenyl group may be an aryl group, or interpreted as a substituent linking two phenyl groups. In the present specification, mean a site bonding to other substituents or bonding sites. In the present specification, examples of the halogen group may include fluorine, chlorine, bromine or iodine. In the present specification, the number of carbon atoms of the imide group is not particularly limited, but is preferably from 1 to 30. In the present specification, in the amide group, nitrogen of the amide group may be substituted with hydrogen, a linear, branched or cyclic alkyl group having 1 to 30 carbon atoms, or an aryl group having 6 to 30 carbon atoms. In the present specification, in the ester group, oxygen of the ester group may be substituted with a linear, branched or cyclic alkyl group having 1 to 25 carbon atoms; or a monocyclic or polycyclic aryl group having 6 to 30 carbon atoms. Specifically, compound having a structure such as —C(═O)ORa or —O(C═O)Ra may be included, and in this case, Ra is a substituted or unsubstituted alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted heteroaryl group. In the present specification, the number of carbon atoms of the carbonyl group is not particularly limited, but is preferably from 1 to 30. Specifically, compounds having a structure such as —C(═O)Rb may be included, and in this case, Rb is hydrogen or an alkyl group, however, the carbonyl group is not limited thereto. In the present specification, the alkyl group may be linear or branched, and although not particularly limited thereto, the number of carbon atoms is preferably from 1 to 30. Specific examples thereof may include methyl, ethyl, propyl, n-propyl, isopropyl, butyl, n-butyl, isobutyl, tert-butyl, sec-butyl, 1-methyl-butyl, 1-ethyl-butyl, pentyl, n-pentyl, isopentyl, neopentyl, tert-pentyl, hexyl, n-hexyl, 1-methylpentyl, 2-methylpentyl, 4-methyl-2-pentyl, 3,3-dimethylbutyl, 2-ethylbutyl, heptyl, n-heptyl, 1-methylhexyl, cyclopentylmethyl, cyclohexylmethyl, octyl, n-octyl, tert-octyl, 1-methylheptyl, 2-ethylhexyl, 2-propylpentyl, n-nonyl, 2,2-dimethylheptyl, 1-ethyl-propyl, 1,1-dimethyl-propyl, isohexyl, 2-methylpentyl, 4-methylhexyl, 5-methylhexyl and the like, but are not limited thereto. In the present specification, the cycloalkyl group is not particularly limited, but preferably has 3 to 30 carbon atoms, and specific examples thereof may include cyclopropyl, cyclobutyl, cyclopentyl, 3-methylcyclopentyl, 2,3-dimethylcyclopentyl, cyclohexyl, 3-methylcyclohexyl, 4-methylcyclohexyl, 2,3-dimethylcyclohexyl, 3,4,5-trimethylcyclohexyl, 4-tert-butylcyclohexyl, cycloheptyl, cyclooctyl and the like, but are not limited thereto. In the present specification, the alkoxy group may be linear, branched or cyclic. The number of carbon atoms of the alkoxy group is not particularly limited, but is preferably from 1 to 30. Specific examples thereof may include methoxy, ethoxy, n-propoxy, isopropoxy, i-propyloxy, n-butoxy, isobutoxy, tert-butoxy, sec-butoxy, n-pentyloxy, neopentyloxy, isopentyloxy, n-hexyloxy, 3,3-dimethylbutyloxy, 2-ethylbutyloxy, n-octyloxy, n-nonyloxy, n-decyloxy, benzyloxy, p-methylbenzyloxy and the like, but are not limited thereto. In the present specification, the alkenyl group may be linear or branched, and although not particularly limited thereto, the number of carbon atoms is preferably from 2 to 30. Specific examples thereof may include vinyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 3-methyl-1-butenyl, 1,3-butadienyl, allyl, 1-phenylvinyl-1-yl, 2-phenylvinyl-1-yl, 2,2-diphenylvinyl-1-yl, 2-phenyl-2-(naphthyl-1-yl)vinyl-1-yl, 2,2-bis(diphenyl-1-yl)vinyl-1-yl, a stilbenyl group, a styrenyl group and the like, but are not limited thereto. In the present specification, the alkynyl group may be linear or branched, and although not particularly limited thereto, the number of carbon atoms is preferably from 2 to 30. Specific examples thereof may include an alkynyl group such as ethynyl, propynyl, 2-methyl-2-propynyl, 2-butynyl or 2-pentynyl, but are not limited thereto. In the present specification, the amine group may be selected from the group consisting of —NH2; a monoalkylamine group; a dialkylamine group; an N-alkylarylamine group; a monoarylamine group; a diarylamine group; an N-arylheteroarylamine group; an N-alkylheteroarylamine group, a monoheteroarylamine group and a diheteroarylamine group, and although not particularly limited thereto, the number of carbon atoms is preferably from 1 to 30. Specific examples of the amine group may include a methylamine group, a dimethylamine group, an ethylamine group, a diethylamine group, a phenylamine group, a naphthylamine group, a biphenylamine group, an anthracenylamine group, a 9-methyl-anthracenylamine group, a diphenylamine group, a ditolylamine group, an N-phenyltolylamine group, a triphenylamine group, an N-phenylbiphenylamine group; an N-phenylnaphthylamine group; an N-biphenylnaphthylamine group; an N-naphthylfluorenylamine group; an N-phenylphenanthrenylamine group; an N-biphenylphenanthrenylamine group; an N-phenylfluorenylamine group; an N-phenylterphenylamine group; an N-phenanthrenylfluorenylamine group; an N-biphenylfluorenylamine group and the like, but are not limited thereto. In the present specification, the N-alkylarylamine group means an amine group in which N of the amine group is substituted with an alkyl group and an aryl group. In the present specification, the N-arylheteroarylamine group means an amine group in which N of the amine group is substituted with an aryl group and a heteroaryl group. In the present specification, the N-alkylheteroarylamine group means an amine group in which N of the amine group is substituted with an alkyl group and a heteroaryl group. In the present specification, the alkyl group in the alkylamine group, the N-alkylarylamine group, the alkylthioxy group, the alkylsulfoxy group and the N-alkylheteroarylamine group is the same as the examples of the alkyl group described above. Specific examples of the alkylthioxy group may include a methylthioxy group, an ethylthioxy group, a tert-butylthioxy group, a hexylthioxy group, an octylthioxy group and the like, and specific examples of the alkylsulfoxy group may include mesyl, an ethylsulfoxy group, a propylsulfoxy group, a butylsulfoxy group and the like, however, the alkylthoixy group and the alkylsulfoxy group are not limited thereto. In the present specification, the aryl group is not particularly limited, but preferably has 6 to 30 carbon atoms, and the aryl group may be monocyclic or polycyclic. When the aryl group is a monocyclic aryl group, the number of carbon atoms is not particularly limited, but is preferably from 6 to 30. Specific examples of the monocyclic aryl group may include a phenyl group, a biphenyl group, a terphenyl group and the like, but are not limited thereto. When the aryl group is a polycyclic aryl group, the number of carbon atoms is not particularly limited, but is preferably from 10 to 30. Specific examples of the polycyclic aryl group may include a naphthyl group, an anthracenyl group, a phenanthryl group, a triphenyl group, a pyrenyl group, a perylenyl group, a chrysenyl group, a fluorenyl group and the like, but are not limited thereto. In the present specification, the fluorenyl group may be substituted, and adjacent substituents may bond to each other to form a ring. When the fluorenyl group is substituted, and the like may be included. However, the structure is not limited thereto. In the present specification, the aryl group in the aryloxy group, the arylthioxy group, the N-alkylarylamine group and the N-arylheteroarylamine group is the same as the examples of the aryl group described above. Specific examples of the aryloxy group may include phenoxy, p-tolyloxy, m-tolyloxy, 3,5-dimethyl-phenoxy, 2,4,6-trimethylphenom p-tert-butylphenoxy, 3-biphenyloxy, 4-biphenyloxy, 1-naphthyloxy, 2-naphthyloxy, 4-methyl-1-naphthyloxy, 5-methyl-2-naphthyloxy, 1-anthryloxy, 2-anthryloxy, 9-anthryloxy, 1-phenanthryloxy, 3-phenanthryloxy, 9-phenanthryloxy and the like, and specific examples of the arylthioxy group may include a phenylthioxy group, a 2-methylphenylthioxy group, a 4-tert-butylphenylthioxy group and the like, however, the aryloxy group and the arylthioxy group are not limited thereto. In the present specification, the heteroaryl group is a group including one or more atoms that are not carbon, that is, heteroatoms, and specifically, the heteroatom may include one or more atoms selected from the group consisting of O, N, Se, S and the like. The number of carbon atoms is not particularly limited, but is preferably from 2 to 30, and the heteroaryl group may be monocyclic or polycyclic. Examples of the heteroaryl group may include a thiophene group, a furanyl group, a pyrrole group, an imidazolyl group, a thiazolyl group, an oxazolyl group, an oxadiazolyl group, a pyridine group, a bipyridine group, a pyrimidine group, a triazinyl group, a triazolyl group, an acridyl group, a pyridazinyl group, a pyrazinyl group, a quinolinyl group, a quinazolinyl group, a quinoxalinyl group, a phthalazinyl group, a pyridopyrimidyl group, a pyridopyrazinyl group, a pyrazinopyrazinyl group, an isoquinolinyl group, an indolyl group, a carbazolyl group, a benzoxazolyl group, a benzimidazolyl group, a benzothiazolyl group, a benzocarbazolyl group, a benzothiophene group, a dibenzothiophene group, a benzofuranyl group, a phenanthrolinyl group, an isoxazolyl group, a thiadiazolyl group, a phenothiazinyl group, a dibenzofuranyl group, a chromene group and the like, but are not limited thereto. In the present specification, the heteroaryl group may be monocyclic or polycyclic, may be aromatic or a fused ring of aromatic and aliphatic, and may be selected from among the examples of the heterocyclic group. In the present specification, an “adjacent” group may mean a substituent substituting an atom directly linked to an atom substituted by the corresponding substituent, a substituent sterically most closely positioned to the corresponding substituent, or another substituent substituting an atom substituted by the corresponding substituent. For example, two substituents substituting ortho positions in a benzene ring, and two substituents substituting the same carbon in an aliphatic ring may be interpreted as groups “adjacent” to each other. In the present specification, the meaning of “adjacent groups bond to each other to form a ring” among substituents means adjacent groups bonding to each other to form a substituted or unsubstituted hydrocarbon ring; or a substituted or unsubstituted heteroring. One embodiment of the present specification provides a compound represented by the following Chemical Formula 1. In Chemical Formula 1, X1 to X3 are the same as or different from each other, and each independently O or S, X4 and X5 are the same as or different from each other, and each independently a halogen group; CN; a substituted or unsubstituted alkoxy group; a substituted or unsubstituted alkenyl group; a substituted or unsubstituted alkynyl group; a substituted or unsubstituted aryl group; a substituted or unsubstituted aryloxy group; or a substituted or unsubstituted heteroaryl group, R1 and R6 are the same as or different from each other, and each independently hydrogen; deuterium; a halogen group; CN; a substituted or unsubstituted alkyl group; a substituted or unsubstituted cycloalkyl group; a substituted or unsubstituted alkoxy group; a substituted or unsubstituted aryloxy group; a substituted or unsubstituted aryl group; or a substituted or unsubstituted heteroaryl group, R2 and R5 are the same as or different from each other, and each independently a substituted or unsubstituted ester group; a substituted or unsubstituted aryl group; or a substituted or unsubstituted heteroaryl group, R3 and R4 are the same as or different from each other, and each independently a substituted or unsubstituted alkyl group; a substituted or unsubstituted cycloalkyl group; a substituted or unsubstituted aryl group; or a substituted or unsubstituted heteroaryl group, and R7 is a substituted or unsubstituted aryl group; or a substituted or unsubstituted heteroaryl group. According to one embodiment of the present specification, Chemical Formula 1 is represented by any one of the following Chemical Formulae 1-1 to 1-4. In Chemical Formulae 1-1 to 1-4, R1 to R7, X4 and X5 have the same definitions as in Chemical Formula 1. In one embodiment of the present specification, X1 to X3 are the same as or different from each other, and each independently O or S. In one embodiment of the present specification, X1 to X3 are O. In another embodiment, X1 is O, and X2 and X3 are S. In another embodiment, X1 to X3 are S. In another embodiment, X1 is S, and X2 and X3 are O. In one embodiment of the present specification, X4 and X5 are the same as or different from each other, and each independently a halogen group; CN; a substituted or unsubstituted alkoxy group; a substituted or unsubstituted alkenyl group; a substituted or unsubstituted alkynyl group; a substituted or unsubstituted aryl group; a substituted or unsubstituted aryloxy group; or a substituted or unsubstituted heteroaryl group. In one embodiment of the present specification, X4 and X5 are the same as or different from each other, and each independently a halogen group; CN; an alkoxy group unsubstituted or substituted with a halogen group; an alkynyl group unsubstituted or substituted with a substituted or unsubstituted aryl group; an aryl group unsubstituted or substituted with a nitro group; an aryloxy group; or a heteroaryl group. In one embodiment of the present specification, X4 and X5 are the same as or different from each other, and each independently fluorine; CN; an n-butoxy group substituted with a halogen group; an ethynyl group substituted with a substituted or unsubstituted aryl group; a phenyl group unsubstituted or substituted with a nitro group; a substituted or unsubstituted phenoxy group; or a pyridine group. In one embodiment of the present specification, X4 and X5 are the same as or different from each other, and each independently fluorine; CN; an n-butoxy group substituted with fluorine; an ethynyl group substituted with a phenyl group unsubstituted or substituted with an alkyl group; a phenyl group unsubstituted or substituted with NO2; a phenoxy group; or a pyridine group. In one embodiment of the present specification, R1 and R6 are the same as or different from each other, and each independently hydrogen; deuterium; a halogen group; CN; a substituted or unsubstituted alkyl group; a substituted or unsubstituted cycloalkyl group; a substituted or unsubstituted alkoxy group; a substituted or unsubstituted aryloxy group; a substituted or unsubstituted aryl group; or a substituted or unsubstituted heteroaryl group. In one embodiment of the present specification, R1 and R6 are the same as or different from each other, and each independently hydrogen; deuterium; a halogen group; CN; an alkyl group; a cycloalkyl group unsubstituted or substituted with an alkyl group; an alkoxy group; an aryloxy group unsubstituted or substituted with a halogen group, CN, CF3or an alkyl group; an aryl group unsubstituted or substituted with a halogen group, CN, CF3, an alkyl group or an alkoxy group; or a substituted or unsubstituted heteroaryl group. In one embodiment of the present specification, R1 and R6 are the same as or different from each other, and each independently hydrogen; deuterium; chlorine; bromine; CN; a methyl group; a cycloalkyl group having 3 to 30 carbon atoms unsubstituted or substituted with an alkyl group; a methoxy group; an isopropoxy group; an aryloxy group having 6 to 30 carbon atoms unsubstituted or substituted with a halogen group, CN, CF3or an alkyl group; an aryl group having 6 to 30 carbon atoms unsubstituted or substituted with a halogen group, CN, CF3, an alkyl group or an alkoxy group; a pyrrole group; a pyridine group; or a thiophene group. In one embodiment of the present specification, R1 and R6 are the same as or different from each other, and each independently hydrogen; deuterium; chlorine; bromine; CN; a methyl group; a cyclopropyl group; a cyclobutyl group; a cyclopentyl group; a cyclohexyl group unsubstituted or substituted with an alkyl group; an aryloxy group having 6 to 30 carbon atoms unsubstituted or substituted with fluorine, CN, CF3or a methyl group; an aryl group having 6 to 30 carbon atoms unsubstituted or substituted with fluorine, CN, CF3, a methyl group, a butyl group, a tert-butyl group or a methoxy group; a pyrrole group; a pyridine group; or a thiophene group. In one embodiment of the present specification, R2 and R5 are the same as or different from each other, and each independently a substituted or unsubstituted ester group; a substituted or unsubstituted aryl group; or a substituted or unsubstituted heteroaryl group. In one embodiment of the present specification, R2 and R5 are the same as or different from each other, and each independently —C(═O)ORa; an aryl group having 6 to 30 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of a halogen group, CN, CF3, —C(═O)ORa, a substituted or unsubstituted alkyl group, a substituted or unsubstituted alkoxy group, a substituted or unsubstituted amine group, a substituted or unsubstituted aryl group, and a substituted or unsubstituted heteroaryl group; or a heteroaryl group having 6 to 30 carbon atoms unsubstituted or substituted with an aryl group, and Ra is a substituted or unsubstituted alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted heteroaryl group. In one embodiment of the present specification, R2 and R5 are the same as or different from each other, and each independently —C(═O)ORa; an aryl group having 6 to 30 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of a halogen group, CN, CF3, —C(═O)ORa, an alkyl group unsubstituted or substituted with a halogen group, an alkoxy group, an amine group unsubstituted or substituted with an alkyl group, an aryl group having 6 to 30 carbon atoms, and a heteroaryl group having 2 to 30 carbon atoms unsubstituted or substituted with an ester group and ═O; a substituted or unsubstituted dibenzofuranyl group; a substituted or unsubstituted dibenzothiophene group; a substituted or unsubstituted carbazole group; or a substituted or unsubstituted phenanthrolinyl group, and Ra is a substituted or unsubstituted alkyl group having 1 to 30 carbon atoms, a substituted or unsubstituted aryl group having 6 to 30 carbon atoms, or a substituted or unsubstituted heteroaryl group having 6 to 30 carbon atoms. In one embodiment of the present specification, R2 and R5 are the same as or different from each other, and each independently —C(═O)ORa; an aryl group having 6 to 20 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of fluorine, chlorine, bromine, CN, CF3, —C(═O)ORa, a methyl group unsubstituted or substituted with a halogen group, a propyl group, an isopropyl group, a butyl group, a tert-butyl group, a pentyl group, a hexyl group, a methoxy group, NH2, a dialkylamine group, a naphthyl group, an anthracenyl group, a carbazole group, a dibenzofuranyl group, a pyridine group, and a chromene group substituted with an ester group and ═O; a dibenzofuranyl group unsubstituted or substituted with a phenyl group; a dibenzothiophene group unsubstituted or substituted with a phenyl group; a carbazole group unsubstituted or substituted with a phenyl group; or a phenanthrolinyl group, and Ra is a methyl group, a phenyl group unsubstituted or substituted with CN, or a chromene group substituted with ═O. In one embodiment of the present specification, R3 and R4 are the same as or different from each other, and each independently a substituted or unsubstituted alkyl group; a substituted or unsubstituted cycloalkyl group; a substituted or unsubstituted aryl group; or a substituted or unsubstituted heteroaryl group. In one embodiment of the present specification, R3 and R4 are the same as or different from each other, and each independently an alkyl group having 1 to 30 carbon atoms unsubstituted or substituted with CF3; a cycloalkyl group having 1 to 30 carbon atoms unsubstituted or substituted with an alkyl group; an aryl group having 6 to 30 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of a halogen group, CN, CF3, —C(═O)ORa, an amine group, an alkoxy group, an alkyl group having 1 to 30 carbon atoms and a heteroaryl group having 6 to 30 carbon atoms; or a substituted or unsubstituted heteroaryl group having 6 to 30 carbon atoms, and Ra is a substituted or unsubstituted alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted heteroaryl group. In one embodiment of the present specification, R3 and R4 are the same as or different from each other, and each independently an alkyl group having 1 to 30 carbon atoms unsubstituted or substituted with CF3; a cyclohexyl group unsubstituted or substituted with an alkyl group; an aryl group having 6 to 20 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of a halogen group, CN, CF3, —C(═O)ORa, NH2, a dialkylamine group, a diphenylamine group, an alkoxy group, an alkyl group having 1 to 30 carbon atoms, a pyridine group, a dibenzofuranyl group and a carbazole group; a dibenzofuranyl group unsubstituted or substituted with an aryl group; a dibenzothiophene group unsubstituted or substituted with an aryl group; a carbazole group unsubstituted or substituted with an aryl group; or a chromene group unsubstituted or substituted with ═O, and Ra is a substituted or unsubstituted alkyl group having 1 to 30 carbon atoms. In one embodiment of the present specification, R3 and R4 are the same as or different from each other, and each independently an alkyl group having 1 to 30 carbon atoms unsubstituted or substituted with CF3; a cyclohexyl group unsubstituted or substituted with an alkyl group; an aryl group having 6 to 20 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of fluorine, chlorine, CN, CF3, —C(═O)ORa, NH2, a dialkylamine group, a diphenylamine group, a methoxy group, a methyl group, an ethyl group, a propyl group, an isopropyl group, a butyl group, a tert-butyl group, a pentyl group, a hexyl group, a pyridine group, a dibenzofuranyl group and a carbazole group; a dibenzofuranyl group unsubstituted or substituted with a phenyl group; a dibenzothiophene group; a carbazole group unsubstituted or substituted with a phenyl group; or a chromene group substituted with ═O, and Ra is a methyl group. In one embodiment of the present specification, R3 and R4 are the same as each other, and an alkyl group having 1 to 30 carbon atoms unsubstituted or substituted with CF3; a cyclohexyl group unsubstituted or substituted with an alkyl group; an aryl group having 6 to 20 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of fluorine, chlorine, CN, CF3, —C(═O)ORa, NH2, a dialkylamine group, a diphenylamine group, a methoxy group, a methyl group, an ethyl group, a propyl group, an isopropyl group, a butyl group, a tert-butyl group, a pentyl group, a hexyl group, a pyridine group, a dibenzofuranyl group and a carbazole group; a dibenzofuranyl group unsubstituted or substituted with a phenyl group; a dibenzothiophene group; a carbazole group unsubstituted or substituted with a phenyl group; or a chromene group substituted with ═O, and Ra is a methyl group. In one embodiment of the present specification, R7 is a substituted or unsubstituted aryl group; or a substituted or unsubstituted heteroaryl group. In one embodiment of the present specification, R7 is an aryl group unsubstituted or substituted with one or more selected from the group consisting of a halogen group, CN, CF3, an alkoxy group, an alkyl group unsubstituted or substituted with a halogen group, a substituted or unsubstituted aryl group, and a heteroaryl group; or a heteroaryl group unsubstituted or substituted with O═. In one embodiment of the present specification, R7 is an aryl group having 6 to 30 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of fluorine, chlorine, CN, CF3, an alkoxy group, an alkyl group having 1 to 30 carbon atoms unsubstituted or substituted with a halogen group, an aryl group and a heteroaryl group; a pyridine group; a dibenzofuranyl group; a dibenzothiophene group; a carbazolyl group; or In one embodiment of the present specification, R7 is an aryl group having 6 to 20 carbon atoms unsubstituted or substituted with one or more selected from the group consisting of fluorine, chlorine, CN, CF3, a methoxy group, an alkyl group having 1 to 30 carbon atoms unsubstituted or substituted with fluorine or chlorine, a naphthyl group, a dibenzofuranyl group and a pyridine group; a pyridine group; a dibenzofuranyl group; a dibenzothiophene group; a carbazolyl group; or In one embodiment of the present specification, X1 to X5 of Chemical Formula 1 may be selected from the following Tables 1-1 to 1-4, R1, R6 and R7 of Chemical Formula 1 may be selected from the following Tables 2-1 to 2-9, and R2 to R5 of Chemical Formula 1 may be selected from the following Tables 3-1 to 3-14. TABLE 1-1X4X5X1X2X3A1FFOOOA2FCNOOOA3CNCNOOOA4OOOA5OOOA6OOOA7OOOA8OOOA9OOOA10OOO TABLE 1-2X4X5X1X2X3A11FFOSSA12FCNOSSA13CNCNOSSA14OSSA15OSSA16OSSA17OSSA18OSSA19OSSA20SSS TABLE 1-3X4X5X1X2X3A21FFSOOA22FCNSOOA23CNCNSOOA24SOOA25SOOA26SOOA27SOOA28SOOA29SOOA30SOO TABLE 1-4X4X5X1X2X3A31FFSSSA32FCNSSSA33CNCNSSSA34SSSA35SSSA36SSSA37SSSA38SSSA39SSSA40SSS TABLE 2-1R7R1R6HHB1B2B3B4ClHB5B6B7B8ClClB9B10B11B12BrBrB13B14B15B16HCNB17B18B19B20H*—B21B22B23B24*—*—B25B26B27B28HB29B30B31B32HB33B34B35B36HB37B38B39B40B41B42B43B44B45B46B47B48B49B50B51B52HB53B54B55B56HB57B58B59B60HB61B62B63B64HB65B66B67B68HB69B70B71B72HB73B74B75B76HB77B78B79B80B81B82B83B84B85B86B87B88B89B90B91B92B93B94B95B96B97B98B99B100B101B102B103B104B105B106B107B108HB109B110B111B112HB113B114B115B116B117B118B119B120B121B122B123B124HB125B126B127B128HB129B130B131B132HB133B134B135B136HB137B138B139B140HB141B142B143B144HB145B146B147B148B149B150B151B152B153B154B155B156B157B158B159B160B161B162B163B164B165B166B167B168B169B170B171B172B173B174B175B176 TABLE 2-2R7R1R6HHB177B178B179B180ClHB181B182B183B184ClClB185B186B187B188BrBrB189B190B191B192HCNB193B194B195B196H*—B197B198B199B200*—*—B201B202B203B204HB205B206B207B208HB209B210B211B212HB213B214B215B216HB217B218B219B220B221B222B223B224B225B226B227B228B229B230B231B232HB233B234B235B236HB237B238B239B240HB241B242B243B244HB245B246B247B248HB249B250B251B252HB253B254B255B256HB257B258B259B260B261B262B263B264B265B266B267B268B269B270B271B272B273B274B275B276B277B278B279B280B281B282B283B284B285B286B287B288HB289B290B291B292HB293B294B295B296B297B298B299B300B301B302B303B304HB305B306B307B308HB309B310B311B312HB313B314B315B316HB317B318B319B320HB321B322B323B324HB325B326B327B328B329B330B331B332B333B334B335B336B337B338B339B340B341B342B343B344B345B346B347B348B349B350B351B352 TABLE 2-3R7R1R6HHB353B354B355B356ClHB357B358B359B360ClClB361B362B363B364BrBrB365B366B367B368HCNB369B370B371B372H*—B373B374B375B376*—*—B377B378B379B380HB381B382B383B384HB385B386B387B388HB389B390B391B392B393B394B395B396B397B398B399B400B401B402B403B404HB405B046B407B408HB409B410B411B412HB413B414B415B416HB417B418B419B420HB421B422B423B424HB425B426B427B428HB429B430B431B432B433B434B435B436B437B438B439B440B441B442B443B444B445B446B447B448B449B450B451B452B453B454B455B456B457B458B459B460HB461B462B463B464HB465B466B467B468B469B470B471B472B473B474B475B476HB477B478B479B480HB481B482B483B484HB485B486B487B488HB489B490B491B492HB493B494B495B496HB497B498B499B500B501B502B503B504B505B506B507B508B509B510B511B512B513B514B515B516B517B518B519B520B521B522B523B524 TABLE 2-4R7R1R6HHB525B526B527B528ClHB529B530B531B532ClClB533B534B535B536BrBrB537B538B539B540HCNB541B542B543B544H*—B545B546B547B548*—*—B549B550B551B552HB553B554B555B556HB557B558B559B560HB561B562B563B564B565B566B567B568B569B570B571B572B573B574B575B576HB577B578B579B580HB581B582B583B584HB585B586B587B588HB589B590B591B592HB593B594B595B596HB597B598B599B600HB601B602B603B604HB605B606B607B608B609B610B611B612B613B614B615B616B617B618B619B620B621B622B623B624B625B626B627B628B629B630B631B632B633B634B635B636HB637B638B639B640HB641B642B643B644B645B646B647B648B649B650B651B652HB653B654B655B656HB657B658B659B660HB661B662B663B664HB665B666B667B668HB669B670B671B672HB673B674B675B676B677B678B679B680B681B682B683B684B685B686B687B688B689B690B691B692B693B694B695B696B697B698B699B700 TABLE 2-5R7R1R6HHB701B702B703B704ClHB705B706B707B708ClClB709B710B711B712BrBrB713B714B715B716HCNB717B718B719B720H*—B721B722B723B724*—*—B725B726B727B728HB729B730B731B732HB733B734B735B736HB737B738B739B740B741B742B743B744B745B746B747B748B749B750B751B752HB753B754B755B756HB757B758B759B760HB761B762B763B764HB765B766B767B768HB769B770B771B772HB773B774B775B776HB777B778B779B780B781B782B783B784B785B786B787B788B789B790B791B792B793B794B795B796B797B798B799B800B801B802B803B804B805B806B807B808HB809B810B811B812HB813B814B815B816B817B818B819B820B821B822B823B824HB825B826B827B828HB829B830B831B832HB833B834B835B836HB837B838B839B840HB841B842B843B844HB845B846B847B848B849B850B851B852B853B854B855B856B857B858B859B860B861B862B863B864B865B866B867B868B869B870B871B872B873B874B875B876 TABLE 2-6R7R1R6HHB877B878B879B880ClHB881B882B883B884ClClB885B886B887B888BrBrB889B890B891B892HCNB893B894B895B896H*—B897B898B899B900*—*—B901B902B903B904HB905B906B907B908HB909B910B911B912HB913B914B915B916B917B918B919B920B921B922B923B924B925B926B927B928HB929B930B931B932HB933B934B935B936HB937B938B939B940HB941B942B943B944HB945B946B947B948HB949B950B951B952HB953B954B955B956B957B958B959B960B961B962B963B964B965B966B967B968B969B970B971B972B973B974B975B976B977B978B979B980B981B982B983B983HB985B986B987B988HB989B990B991B992B993B994B995B996B997B998B999B1000HB1001B1002B1003B1004HB1005B1006B1007B1008HB1009B1010B1011B1012HB1013B1014B1015B1016HB1017B1018B1019B1020HB1021B1022B1023B1024B1025B1026B1027B1028B1029B1030B1031B1032B1033B1034B0135B1036B1037B1038B1039B1040B1041B1042B1043B1044B1045B1046B1047B1048B1049B1050B1051B1052 TABLE 2-7R7R1R6HHB1053B1054B1055B1056ClHB1057B1058B1059B1060ClClB1061B1062B1063B1064BrBrB1065B1066B1067B1068HCNB1069B1070B1071B1072H*—B1073B1074B1075B1076*—*—B1077B1078B1079B1080HB1081B1082B1083B1084HB1085B1086B1087B1088HB1089B1090B1091B1092B1093B1094B1095B1096B1097B1098B1099B1100B1101B1102B1103B1104HB1105B1106B1107B1108HB1109B1110B1111B1112HB1113B1114B1115B1116HB1117B1118B1119B1120HB1121B1122B1123B1123HB1124B1125B1126B1127HB1128B1129B1130B1131B11132B1133B1134B1135B1136B1137B1138B1139B1140B1141B1142B1143B1144B1145B1146B1147B1148B1149B1150B1151B1152B1153B1154B1155B1156B1157B1158B1169HB1160B1161B1162B1163HB1164B1165B1166B1167B1168B1169B1170B1171B1172B1173B1174B1175HB1176B1177B1178B1179HB1180B1181B1182B1183HB1184B1185B1186B1187HB1188B1189B1190B1191HB1192B1193B1194B1195HB1196B1197B1198B1199B1200B1201B1202B1203B1204B1205B1206B1207B1208B1209B1210B1211B1212B1213B121481215B1216B1217B1218B1219B1220B1221B1222B1223B1224B1225B1226B1227HB1228B1229B1230B1231 TABLE 2-8R7R1R6HHB1232B1233B1234B1235ClHB1236B1237B1237B1238ClClB1239B1240B1241B1242BrBrB1243B1244B1245B1246HCNB1247B1248B1249B1250H*—B1251B1252B1253B1254*—*—B1255B1256B1257B1258HB1259B1260B1261B1262HB1263B1264B1265B1266HB1267B1268B1269B1270HB1271B1272B1273B1274B1275B1276B1277B1278B1279B1280B1281B1282B1283B1284B1285B1286HB1287B1288B1289B1290HB1291B1292B1293B1294HB1295B1296B1297B1298HB1299B1300B1301B1302HB1303B1304B1035B1306HB1037B1308B1309B1310HB1311B1312B1313B1314HB1315B1316B1317B1318B1319B1320B1321B1322B1323B1324B1325B1326B1327B1328B1329B1330B1331B1332B1333B1334B1335B1336B1337B1338B1339B1340B1341B1342B1343B1344B1345B1346HB1347B1348B1349B1350HB1351B1352B1353B1354B1355B1356B1357B1358B1359B1360B1361B1362HB1363B1364B1365B1366HB1367B1368B1369B1370HB1371B1372B1373B1374HB1375B1376B1377B1378HB1379B1380B1381B1382HB1383B1384B1385B1386B1387B1388B1389B1390B1391B1392B1303B1394B1395B1396B1397B1393B1399B1400B1401B1402B1403B1404B1405B1406B1407B1408B1409B1410B1411B1412B1413B1414HB1415B1416B1417B1413 TABLE 2-9R7R1R6HHB1419B1420B1421ClHB1422B1423B1424ClClB1425B1426B1427BrBrB1428B1429B1430HCNB1431B1432B1433H*—B1434B1435B1436*—*—B1437B1438B1430HB1440B1441B1442HB1443B1444B1445HB1446B1447B1448HB1449B1450B1451B1452B1453B1454B1455B1456B1457B1458B1459B1460HB1461B1462B1463HB1464B1465B1466HB1467B1468B1469HB1470B1471B1472HB1473B1474B1475HB1476B1477B1478HB1479B1480B1481HB1482B1483B1484B1485B1486B1487B1488B1489B1490B1491B1492B1493B1494B1495B1496B1497B1498B1499B1500B1501B1502B1503B1504B1505HB1506B1507B1508HB1509B1510B1511B1512B1513B1514B1515B1516B1517HB1518B1519B1520HB1521B1522B1523HB1524B1526B1526HB1527B1528B1529HB1530B1531B1532HB1533B1534B1535HB1536B1537B1538B1539B1540B1541B1542B1543B1544B1545B1546B1547B1548B1549B1550B1551B1552B1553B1554B1555B1556B1557B1558B1559HB1560B1561B1562HB1563B1564B1565B1566B1567B1568HB1569B1570B1571B1572B1573B1574HB1575B1576B1577 TABLE 3-1R3, R4R2R5C1C2C3C4C5C6C7C8C9C10C11C12C13C14C15C16C17C18C19C20C21C22C23C24C25C26C27C28C29C30C31C32C33C34C35C36C37C38C39C40C41C42C43C44C45C46C47C48C49C50C51C52C53C54C55C56C57C58C59CC0C61C62C63C64C65C66C67C68C69C70C71C72C73C74C75C76C77C78C79C80C81C82C83C84C85C86C87C88C89C90C91C92C93C94C95C96C97C98C99C100C101C102C103C104C105C106C107C108C109C110C111C112C113C114C115C116C117C118C119C120C121C122C123C124C125C126C127C128C129C130C131C132C133C134C135C136C137C138C139C140 TABLE 3-2R3, R4R2R5C141C142C143C144C145C146C147C148C149C150C151C152C153C154C155C156C157C158C159C160C161C162C163C164C165C166C167C168C169C170C171C172C173C174C176C176C177C178C179C180C181C182C183C184C185C186C187C188C189C190C191C192C193C194C195C196C197C198C199C200C201C202C203C204C205C206C207C208C209C210C211C212C213C214C215C216C217C218C219C220C221C222C223C224C225C226C227C228C229C230C231C232C233C234C235C236C237C238C239C240C241C242C243C244C245C246C247C248 TABLE 3-3R3, R4R2R5C249C260C251C252C253C254C255C256C257C258C259C260C261C262C263C264C265C266C267C268C269C270C271C272C273C274C275C276C277C278C279C280C281C282C283C284C285C286C287C288C289C290C291C292C293C294C295C296C297C298C299C300C301C302C303C304C305C306C307C308C309C310C311C312C313C314C315C316C317C318C319C320C321C322C323C324C325C326C327C328C329 TABLE 3-4R3, R4R2R5C330C331C332C333C334C335C336C337C338C339C340C341C342C343C344C345C346C347C348C349C350C351C352C353C354C356C356C357C358C359C360C361C362C363C364C365C366C367C368C369C370C371C372C373C374C375C376C377C378C379C380C381C382C383C384C385C386C387C388C389C390C391C392C393C394C395C396C397C398C399C400C401C402C403C404C405C406C407C408C409C410C411C412C413C414C415C416C417C418C419C420C421C422C423C424C425C426C427C428C429 TABLE 3-5R3, R4R2R5C430C431C432C433C434C435C436C437C438C439C440C441C442C443C444C445C446C447C448C449C450C451C452C453C454C455C456C457C458C459C460C461C462C463C464C465C466C467C468C469C470C471C472C473C474C475C476C477C478C479C480C481C482C483C484C485C486C487C488C489C490C491C492C493C494C495C496C497C498C499C500C501C502C503C504 TABLE 3-6R3, R4R2R5C505C506C507C508C509C510C511C512C513C514C515C516C517C518C519C520C521C522C523C524C525C526C527C528C529C530C531C532C533C534C535C536C537C538C539C540C541C542C543C544C545C546C547C548C549C550C551C552C553C554C555C556C557C558C559C560C561C562C563C564C565C566C567C568C569C570C571C572C573C574C575C576C577C578C579C580C581C582C583C584C585 TABLE 3-7R3, R4R2R5C586C587C588C589C590C591C592C593C594C595C596C597C598C599C600C601C602C603C604C605C606C607C608C609C610C611C612C613C614C615C616C617C618C619C620C621C622C623C624C625C626C627C628C629C630C631C632C633C634C635C636C637C638C639C640C641C642C643C644C645C646C647C648C649C650C651C652C653C654C655C656C657C658C659C660 TABLE 3-8R3, R4R2R5C661C662C663C664C665C666C667C668C669C670C671C672C673C674C675C676C677C678C679C680C681C682C683C684C685C686C687C688C689C690C691C692C693C694C695C696C697C698C699C700C701C702C703C704C705C706C707C708C709C710C711C712C713C714C715C716C717C718C719C720C721C722C723C724C725C726C727C728C729C730C731C732C733C734C735C736C737C738C739C740C741C742C743C744 TABLE 3-9R3, R4R2R5C745C746C747C748C749C750C751C752C753C754C755C756C757C758C759C760C761C762C763C764C765C766C767C768C769C770C771C772C773C774C775C776C777C778C779C780C781C782C783C784C785C786C787C788C789C790C791C792C793C794C795C796C797C798C799C800C801C802C803C804C805C806C807C808C809C810C811C812C813C814C815C816C817C818C819C820C821C822C823C824C825C826C827C828C829C830C831C832C833C834C835C836C837C838C839C840 TABLE 3-10R3, R4R2R5C841C842C843C844C845C846C847C848C849C850C851C852C853C854C855C856C857C858C859C860C861C862C863C864C865C866C867C868C869C870C871C872C873C874C875C876C877C878C879C880C881C882C883C884C885C886C887C888C889C890C891C892C893C894C895C896C897C898C899C900C901C902C903C904C905C906C907C908C909C910C911C912C913C914C915 TABLE 3-11R3, R4R2R5C916C917C918C919C920C921C922C923C924C925C926C927C928C929C930C931C932C933C934C935C936C937C938C939C940C941C942C943C944C945C946C947C948C949C950C951C952C953C954C955C956C957C958C959C960C961C962C963C964C965C966C967C968C969C970C971C972C973C974C975C976C977C978C979C980C981C982C983C984C985C986C987C988C989C990C991C992C993C994C995C996C997C998C999C1000C1001C1002C1003C1004C1005C1006C1007C1008C1009C1010C1011C1012C1013C1014C1015C1016C1017C1018C1019 TABLE 3-12R3, R4R2R5C1020C1021C1022C1023C1024C1025C1026C1027C1028C1029C1030C1031C1032C1033C1034C1035C1036C1037C1038C1039C1040C1041C1042C1043C1044C1045C1046C1047C1048C1049C1050C1051C1052C1053C1054C1055C1056C1057C1058C1059C1060C1061C1062C1063C1064C1065C1066C1067C1068C1069C1070C1071C1072C1073C1074C1075C1076C1077C1078C1079C1080C1081C1082C1083C1084C1085C1086C1087C1088C1089C1090C1091C1092C1093C1094C1095C1096C1097C1098C1099C1100C1101C1102C1103C1104C1105C1106C1107C1108C1109C1110C1111C1112C1113C1114C1115C1116C1117C1118C1119C1120C1121C1122C1123C1124C1125C1126C1127C1128C1129C1130C1131C1132C1133C1134C1135C1136C1137C1138C1139C1140C1141C1142C1143C1144C1145C1146C1147C1148C1149C1150C1151C1152C1153C1154C1155C1156C1157C1158C1159C1160C1161C1162C1163C1164C1165C1166C1167 TABLE 3-13R3, R4R2R5C1168C1169C1170C1171C1172C1173C1174C1175C1176C1177C1178C1179C1180C1181C1182C1183C1184C1185C1186C1187C1188C1189C1190C1191C1192C1193C1194C1195C1196C1197C1198C1199C1200C1201C1202C1203C1204C1205C1206C1207C1208C1209C1210C1211C1212C1213C1214C1215C1216C1217C1218C1219C1220C1221C1222C1223C1224C1225C1226C1227C1228C1229C1230C1231C1232C1233C1234C1235C1236C1237C1238C1239C1240C1241C1242C1243C1244C1245C1246C1247C1248C1249C1250C1251C1252C1253C1254C1255C1256C1257C1258C1259C1260C1261C1262C1263C1264C1265C1266C1267C1268C1269C1270C1271C1272C1273C1274C1275C1276C1277C1278C1279C1280C1281C1282C1283C1284C1285C1286C1287C1288C1289C1290C1291C1292C1293C1294C1295C1296C1297C1298C1299C1300C1301C1302C1303C1304C1305C1306C1307C1308C1309C1310C1311C1312C1313C1314C1315C1316C1317C1318C1319C1320C1321C1322C1323C1324C1325C1326C1327C1328C1329C1330C1331C1332C1333C1334C1335C1336C1337C1338C1339C1340C1341C1342C1343C1344C1345C1346C1347 TABLE 3-14R3, R4R2R5C1348C1349C1350C1351C1352C1353C1354C1355C1356C1357C1358C1359C1360C1361C1362C1363C1364C1365C1366C1367C1368C1369C1370C1371C1372C1373C1374C1375C1376C1377C1378C1379C1380C1381C1382C1383C1384C1385C1386C1387C1388C1389C1390C1391C1392C1393C1394C1395C1396C1397C1398C1399C1400C1401C1402C1403C1404C1405C1406C1407C1408C1409C1410C1411C1412C1413C1414C1415C1416C1417C1418C1419C1420C1421C1422C1423C1424C1425C1426C1427C1428C1429C1430C1431C1432C1433C1434C1435 In Tables 1-1 to 1-4, 2-1 to 2-9, and 3-1 to 3-14 in one embodiment of the present specification, * means a position bonding to Chemical Formula 1. In one embodiment of the present specification, the compounds represented by Chemical Formula 1 are referred to as [1-1 to 1-4]-[2-1 to 2-9]-[3-1 to 3-14] according to the above-described Tables 1-1 to 1-4, 2-1 to 2-9, and 3-1 to 3-14, and specifically, for example, the compound of A1-B328-C437 has a structure as the following Structure 1, and the compound of A21-B423-C628 has a structure as the following Structure 2. According to one embodiment of the present specification, the compound represented by Chemical Formula 1 has a maximum light emission peak present in 500 nm to 550 nm in a film state. Such a compound emits green light. According to one embodiment of the present specification, the compound represented by Chemical Formula 1 has a maximum light emission peak present in 520 nm to 550 nm in a film state, and the light emission peak has a full width at half maximum of 50 nm or less. Having such a small full width at half maximum may further increase color gamut. Herein, it is the better that the light emission peak of the compound represented by Chemical Formula 1 has a smaller full width at half maximum. According to one embodiment of the present specification, the compound represented by Chemical Formula 1 has a maximum light emission peak present in 580 nm to 680 nm in a film state. Such a compound emits red light. According to one embodiment of the present specification, the compound represented by Chemical Formula 1 has a maximum light emission peak present in 580 nm to 680 nm in a film state, and the light emission peak has a full width at half maximum of 60 nm or less. Having such a small full width at half maximum may further increase color gamut. Herein, the light emission peak of the compound represented by Chemical Formula 1 may have a full width at half maximum of 5 nm or greater. According to one embodiment of the present specification, the compound represented by Chemical Formula 1 has quantum efficiency of 0.8 or greater. In the present specification, the “film state” means, instead of a solution state, a state prepared to a film form with the compound represented by Chemical Formula 1 alone or by mixing the compound represented by Chemical Formula 1 with other components that do not affect measurements of full width at half maximum and quantum efficiency. In the present specification, the full width at half maximum means a width of a light emission peak at a half of the maximum height in a maximum light emission peak of the light emitting from the compound represented by Chemical Formula 1. In the present specification, the quantum efficiency may be measured using methods known in the art, and for example, may be measured using an integrating sphere. According to one embodiment of the present specification, the core of the compound represented by Chemical Formula 1 may be prepared using a general preparation method of a reaction formula as below, however, the preparation method is not limited thereto. In the reaction formula, substituents have the same definitions as above. For example, X4 and X5 of the reaction formula may each have the same definition as in Chemical Formula 1 described above, and may be fluorine. One embodiment of the present specification provides a color conversion film including a resin matrix; and the compound represented by Chemical Formula 1 dispersed into the resin matrix. The content of the compound represented by Chemical Formula 1 in the color conversion film may be in a range of 0.001% by weight to 10% by weight. The color conversion film may include one type of the compound represented by Chemical Formula 1, or may include two or more types thereof. For example, the color conversion film may include one type of compound emitting green light among the compounds represented by Chemical Formula 1. As another example, the color conversion film may include one type of compound emitting red light among the compounds represented by Chemical Formula 1. As another example, the color conversion film may include one type of compound emitting green light and one type of compound emitting red light among the compounds represented by Chemical Formula 1. The color conversion film may further include additional fluorescent substances in addition to the compound represented by Chemical Formula 1. When using a light source emitting blue light, the color conversion film preferably includes both a green light emitting fluorescent substance and a red light emitting fluorescent substance. In addition, when using a light source emitting blue light and green light, the color conversion film may only include a red light emitting fluorescent substance. However, the color conversion film is not limited thereto, and even when using a light source emitting blue light, the color conversion film may only include a red light emitting compound when a separate film including a green light emitting fluorescent substance is laminated. On the other hand, even when using a light source emitting blue light, the color conversion film may only include a green light emitting compound when a separate film including a red light emitting fluorescent substance is laminated. The color conversion film may further include a resin matrix; and an additional layer including a compound dispersed into the resin matrix and emitting light in a wavelength different from the wavelength of the compound represented by Chemical Formula 1. The compound emitting light in a wavelength different from the wavelength of the compound represented by Chemical Formula 1 may also be the compound represented by Chemical Formula 1, or may be other known fluorescent substances. The resin matrix material is preferably a thermoplastic polymer or a thermocurable polymer. Specifically, a poly(meth)acryl-based such as polymethyl methacrylate (PMMA), a polycarbonate (PC)-based, a polystyrene (PS)-based, a polyarylene (PAR)-based, a polyurethane (TPU)-based, a styrene-acrylonitrile (SAN)-based, a polyvinylidene fluoride (PVDF)-based, a modified polyvinylidene fluoride (modified-PVDF)-based and the like may be used as the resin matrix material. According to one embodiment of the present specification, the color conversion film according to the embodiments described above additionally includes light diffusing particles. By dispersing light diffusing particles into the color conversion film instead of a light diffusing film used in the art for enhancing luminance, higher luminance may be exhibited compared to using a separate light diffusing film, and an adhering process may be skipped as well. As the light diffusing particles, particles having a high refractive index with the resin matrix may be used, and examples thereof may include TiO2, silica, borosilicate, alumina, sapphire, air or other gases, air- or gas-filled hollow beads or particles (for example, air/gas-filled glass or polymers); polystyrene, polycarbonate, polymethyl methacrylate, acryl, methyl methacrylate, styrene, melamine resin, formaldehyde resin, or polymer particles including melamine and formaldehyde resins, or any suitable combination thereof. The light diffusing particles may have particle diameters in a range of 0.1 μm to 5 μm, for example, in a range of 0.3 μm to 1 μm. The content of the light diffusing particles may be determined as necessary, and for example, may be in a range of approximately 1 part by weight to 30 parts by weight based on 100 parts by weight of the resin matrix. The color conversion film according to the embodiments described above may have a thickness of 2 μm to 200 μm. Particularly, the color conversion film may exhibit high luminance even with a small thickness of 2 μm to 20 μm. This is due to the fact that the content of the fluorescent substance molecules included in the unit volume is higher compared to quantum dots. The color conversion film according to the embodiments described above may have a substrate provided on one surface. This substrate may function as a support when preparing the color conversion film. Types of the substrate are not particularly limited, and the material or thickness is not limited as long as it is transparent and is capable of functioning as the support. Herein, being transparent means having visible light transmittance of 70% or higher. For example, a PET film may be used as the substrate. The color conversion film described above may be prepared by coating a resin solution in which the compound represented by Chemical Formula 1 described above is dissolved on a substrate and drying the result, or by extruding and filming the compound represented by Chemical Formula 1 described above together with a resin. The compound represented by Chemical Formula 1 is dissolved in the resin solution, and therefore, the compound represented by Chemical Formula 1 is uniformly distributed in the solution. This is different from a quantum dot film preparation process that requires a separate dispersion process. As for the resin solution in which the compound represented by Chemical Formula 1 is dissolved, the preparation method is not particularly limited as long as the compound represented by Chemical Formula 1 and the resin described above are dissolved in the solution. According to one example, the resin solution in which the compound represented by Chemical Formula 1 is dissolved may be prepared using a method of preparing a first solution by dissolving the compound represented by Chemical Formula 1 in a solvent, preparing a second solution by dissolving a resin in a solvent, and mixing the first solution and the second solution. When mixing the first solution and the second solution, it is preferable that these be uniformly mixed. However, the method is not limited thereto, and a method of simultaneously adding and dissolving the compound represented by Chemical Formula 1 and a resin in a solvent, a method of dissolving the compound represented by Chemical Formula 1 in a solvent and subsequently adding and dissolving a resin, a method of dissolving a resin in a solvent and then subsequently adding and dissolving the compound represented by Chemical Formula 1, and the like, may be used. As the resin included in the solution, the resin matrix material described above, a monomer curable to this resin matrix resin, or a mixture thereof, may be used. For example, the monomer curable to the resin matrix resin includes a (meth)acryl-based monomer, and this may be formed to a resin matrix material by UV curing. When using such a curable monomer, an initiator required for curing may be further added as necessary. The solvent is not particularly limited as long as it is capable of being removed by drying afterword while having no adverse effects on the coating process. Non-limiting examples of the solvent may include toluene, xylene, acetone, chloroform, various alcohol-based solvents, methylethyl ketone (MEK), methylisobutyl ketone (MIBK), ethyl acetate (EA), butyl acetate (BA), dimethylformamide (DMF), dimethylacetamide (DMAc), dimethyl sulfoxide (DMSO), N-methyl-pyrrolidone (NMP) and the like, and one type or a mixture of two or more types may be used. When the first solution and the second solution are used, solvents included in each of the solutions may be the same as or different from each other. Even when different types of solvents are used in the first solution and the second solution, these solvents preferably have compatibility so as to be mixed with each other. The process of coating the resin solution in which the compound represented by Chemical Formula 1 is dissolved on a substrate may use a roll-to-roll process. For example, a process of unwinding a substrate from a substrate-wound roll, coating the resin solution in which the compound represented by Chemical Formula 1 is dissolved on one surface of the substrate, drying the result, and then winding the result again on the roll may be used. When a roll-to-roll process is used, viscosity of the resin solution is preferably determined in a range capable of conducting the process, and for example, may be determined in a range of 200 cps to 2,000 cps. As the coating method, various known methods may be used, and for example, a die coater may be used, or various bar coating methods such as a comma coater and a reverse comma coater may be used. After the coating, a drying process is conducted. The drying process may be conducted under a condition required to remove a solvent. For example, a color conversion film including a fluorescent substance including the compound represented by Chemical Formula 1 having target thickness and concentration may be obtained on a substrate by carrying out the drying in an oven located close to a coater under a condition to sufficiently evaporate a solvent, in a direction of the substrate progressing during the coating process. When a monomer curable to the resin matrix resin is used as the resin included in the solution, curing, for example, UV curing, may be conducted prior to or at the same time as the drying. When the compound represented by Chemical Formula 1 is filmed by being extruded with a resin, extrusion methods known in the art may be used, and for example, the color conversion film may be prepared by extruding the compound represented by Chemical Formula 1 with a resin such as a polycarbonate (PC)-based, a poly(meth)acryl-based and a styrene-acrylonitrile (SAN)-based. According to one embodiment of the present specification, the color conversion film may have a protective film or a barrier film provided on at least one surface. As the protective film or the barrier film, those known in the art may be used. Another embodiment of the present specification provides a backlight unit including the color conversion film described above. The backlight unit may have backlight unit constitutions known in the art except for including the color conversion film. For example,FIG.1illustrates one example. According toFIG.1, the color conversion film according to the embodiments described above is provided on a surface opposite to a surface facing a reflecting plate of a light guide plate.FIG.1illustrates a constitution including a light source and a reflecting plate surrounding the light source, however, the constitution is not limited to such a structure, and may vary depending on the backlight unit structure known in the art. In addition, as the light source, a direct type as well as a side chain type may be used, and the reflecting plate or the reflective layer may not be included or may be replaced with other constituents as necessary, and when necessary, additional films such as a light diffusing film, a light concentrating film and a luminance enhancing film may be further provided. Preferably, a light concentrating film and a luminance enhancing film are further provided on the color conversion film. In the constitution of the backlight unit as inFIG.1, a scattering pattern may be provided as necessary on the upper surface or a lower surface of the light guide plate. Light introduced into the light guide plate has non-uniform light distribution due to repetition of optical processes such as reflection, total reflection, refraction or transmission, and the scattering pattern may be used to induce the non-uniform light distribution to uniform brightness. Another embodiment of the present application uses a display apparatus including the backlight unit described above. This display apparatus is not particularly limited as long as it includes the above-described backlight unit as a constituent. For example, the display apparatus includes a display module and a backlight unit.FIG.2illustrates a structure of the display apparatus. However, the structure is not limited thereto, and between the display module and the backlight unit, additional films such as a light diffusing film, a light concentrating film and a luminance enhancing film may be further provided as necessary. Hereinafter, the present specification will be described in detail with reference to examples. However, the examples according to the present specification may be modified to various other forms, and the scope of the present specification is not to be construed as being limited to the examples described below. Examples of the present specification are provided in order to more fully describe the present specification to those having average knowledge in the art. PREPARATION EXAMPLE The compound according to one embodiment of the present specification may be prepared using the following Synthesis Methods 1 to 14. [Synthesis Method 1] Chloro BODIPY (1 equivalent), R—OH (1 equivalent) and potassium carbonate (1.2 equivalents) were introduced into an acetonitrile (ACN) solvent, and the result was stirred while heating. After the reaction was finished, the result was extracted using water and chloroform, and the organic layer was dried with anhydrous magnesium sulfate. The solvent was dried through a vacuum distillation apparatus, and produced solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 2] After dissolving a starting material (1 equivalent) in an acetonitrile solvent, N-bromosuccinimide (NBS) was slowly introduced thereto at room temperature. When connecting 5 Brs, N-bromosuccinimide was used in 6 equivalents, and for 6 Brs, 10 equivalents were used. The reaction was conducted through stirring while heating, and when the reaction was finished, the result was cooled to room temperature, and then sufficiently stirred after introducing a sodium thiosulfate solution thereto. The organic layer was separated and dried with anhydrous magnesium sulfate, and the solvent was dried using a vacuum distillation apparatus. After the drying, solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 3] After dissolving a starting material (1 equivalent) in a dichloromethane (DCM) solvent, the result was stirred at −78° C. under the nitrogen atmosphere. Bromine (4 equivalents) diluted to 10 times in an acetonitrile solvent was slowly added dropwise thereto. During the dropwise addition, the temperature was continuously maintained so that the temperature did not rise. After the stepwise addition, the reaction progress was checked, and when the reaction was finished, a sodium thiosulfate solution and a potassium carbonate solution were introduced thereto, and the result was stirred for a sufficient period of time. The organic layer was separated, washed once more with water, and dried using anhydrous magnesium sulfate. After the drying, produced solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 4] After dissolving a starting material (1 equivalent) in an acetonitrile solvent, aryl alcohol/alkyl alcohol (3 equivalents) and potassium carbonate (5 equivalents) to use in the reaction were added thereto, and the result was stirred while heating. When the reaction was finished, the result was cooled to room temperature, and then extracted using water and chloroform. The organic layer was dried using anhydrous magnesium sulfate, and then the solvent was dried through a vacuum distillation apparatus. Produced solids were filtered using methanol to obtain a target material. [Synthesis Method 5] A starting material (1 equivalent) having halogen and a material having boronic acid were introduced using toluene and ethanol, potassium carbonate was dissolved in water, and these were stirred together while heating. For one Suzuki coupling, the boronic acid was used in 1.1 equivalents, and for two Suzuki couplings, 3 equivalents were used. Tetrakistriphenylphosphine palladium (Pd(PPh3)4) was used in 0.01 equivalents to conduct the reaction. After the reaction was finished, the result was cooled to room temperature, and extracted using water and ethyl acetate. The organic layer was dried using anhydrous magnesium sulfate, and the solvent was dried through a vacuum distillation apparatus. Produced solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 6] After dissolving a starting material (1 equivalent) in an acetonitrile solvent, N-chlorosuccinimide (NCS) was slowly added dropwise thereto. To make 5 Cls, the N-chlorosuccinimide was used in 7 equivalents, and for 6 Cls, 10 equivalents were used. After the dropwise addition was completed, the reaction was progressed through stirring while heating, and after the reaction was finished, the result was cooled to room temperature, and sufficiently stirred using a sodium thiosulfate solution. The organic layer was separated, and then dried using anhydrous magnesium sulfate, and the solvent was dried through a vacuum distillation apparatus. Produced solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 7] After dissolving a starting material in a dichloromethane solvent, the result was stirred at 0° C. under the nitrogen atmosphere. Trimethylsilyl cyanide (TMS-CN) and boron trifluoride ethyl ether (BF3OEt2) were slowly added dropwise thereto. For one cyanide substitution, the trimethylsilyl cyanide was used in 5 equivalents and the boron trifluoride ethyl ether was used in 2 equivalents, and for two cyanide substitution, 15 equivalents and 5 equivalents were respectively used. When the reaction was finished, the result was extracted using water and chloroform, and the organic layer was dried using anhydrous magnesium sulfate. The solvent was dried through a vacuum distillation apparatus, and produced solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 8] After dissolving a starting material (1 equivalent) in a dimethylformamide (DMF) solvent, a cycloalkyl-boron trifluoride potassium salt was introduced thereto, and manganese triacetate dihydrate (Mn(OAc)32H2O) was introduced thereto. For one cycloalkyl, the corresponding cycloalkyl was used in 1.5 equivalents and the manganese was used in 3 equivalents, and for two cycloalkyls, the cycloalkyl was used in 3 equivalents and the manganese was used in 5 equivalents. When the reaction was finished, water was introduced thereto, and produced solids were filtered through filtration. The solids were dissolved again in chloroform, and the result was dried using anhydrous magnesium sulfate. Produced solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 9] After introducing a dichloroethane (DCE) solvent to a flask at 0° C. under the nitrogen atmosphere, phosphorous oxychloride (POCl3) and dimethylformamide were introduced thereto in 1:1, and the result was stirred for approximately 1 hour. After introducing a starting material (1 equivalent) to the flask, the reaction was progressed through stirring while heating. To make one aldehyde, the phosphorous oxychloride was used in 3 equivalents to prepare a solution, and to make two aldehydes, 10 equivalents were used to prepare a solution. When checking the reaction progress, a small amount was taken out, washed with a sodium bicarbonate solution, and then checked. After the reaction was finished, the flask was immersed in ice water, and then the result was neutralized by slowly adding a sodium bicarbonate solution thereto. After finishing the neutralization, the organic layer was separated, dried using anhydrous magnesium sulfate, and produced solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 10] After dissolving a starting material in a tetrahydrofuran (THF) solvent, sulfamic acid corresponding to 3 equivalents per 1 equivalent of aldehyde to oxidize was dissolved in water, and these were stirred together. The temperature was lowered to 0° C. after 30 minutes, and sodium chloride (1.2 equivalents) dissolved in water was slowly introduced thereto. After the reaction was completed, a sodium thiosulfate solution was introduced thereto, and after stirring the result, the organic layer was separated. The separated organic layer was dried using anhydrous magnesium sulfate, and the solvent was removed through a vacuum distillation apparatus. Produced solids were filtered using a methanol solvent to obtain a target material. [Synthesis Method 11] A starting material including an acid and a starting material including an alcohol were dissolved in chloroform with 1.05 equivalent of the alcohol for 1 equivalent of the acid. Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and dimethylaminopyridine (DMAP) were introduced thereto in 1.1 equivalents each with respect to the acid, and the result was stirred while heating. After the reaction was finished, the result was extracted using water and chloroform, and the organic layer was dried using anhydrous magnesium sulfate. Produced solids were filtered using methanol to obtain a target material. [Synthesis Method 12] After stirring palladium acetate (Pd(OAc)2) and Xantphos (Sigma-Aldrich, CAS Number 161265-03-8/4,5-bis(diphenylphosphino)-9,9-dimethylxanthene) in a dimethylformamide solvent, the result was introduced to a starting material having halogen placed in a flask at room temperature under the nitrogen atmosphere. After approximately 5 minutes, the result was introduced to a flask in which an indium starting material and diisopropylethylamine (DIPEA) were stirred in a dimethylformamide solvent using a cannula (double ended needle), and the result was stirred while heating. After the reaction was finished, the result was extracted using a sodium bicarbonate solution and chloroform, and the organic layer was dried using anhydrous magnesium sulfate. Produced solids were filtered using methanol to obtain a target material. [Synthesis Method 13] After dissolving a starting material (1 equivalent) in dichloromethane, aluminum chloride (5 equivalents) was introduced thereto, and the result was stirred. Heptafluorobutanol (C3F7CH2OH) (3 equivalents) was introduced thereto, the result was stirred while heating, and when the reaction was finished, the result was extracted using water and chloroform. The organic layer was dried using anhydrous magnesium sulfate, and after removing the solvent through a vacuum distillation apparatus, produced solids were filtered using methanol to obtain a target material. [Synthesis Method 14] After dissolving a starting material (1 equivalent) and t-butyl ethynylbenzene (2.1 equivalents) in an anhydrous tetrahydrofuran solvent, the flask was maintained under the nitrogen atmosphere at −78° C. for approximately 1 hour. n-BuLi (2.05 equivalents) was slowly added dropwise thereto, and the temperature was raised to room temperature. When the reaction was finished, the result was extracted using water and chloroform, and the organic layer was dried using anhydrous magnesium sulfate. The solvent was removed through a vacuum distillation apparatus, and produced solids were filtered using methanol to obtain a target material. Preparation Example 1 Compound 1 (Synthesis of 1-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 7-hydroxycoumarin. Compound 1-1 was obtained in 6.6 g (yield 85%). (Synthesis of 1-2) Synthesis was progressed according to Synthesis Method 2 using Compound 1-1 and N-bromosuccinimide. Compound 1-2 was obtained in 9.1 g (yield 72%). (Synthesis of 1-3) Synthesis was progressed according to Synthesis Method 4 using Compound 1-2 and tetrakistrifluoromethylbiphenyl-ol. Compound 1-3 was obtained in 14.3 g (yield 81%). (Synthesis of 1-4) Synthesis was progressed according to Synthesis Method 5 using Compound 1-3 and t-butylphenylboronic acid. Compound 1-4 was obtained in 11.4 g (yield 76%). (Synthesis of Compound 1) Synthesis was progressed according to Synthesis Method 4 using Compound 1-4 and biphenylol. Final Compound 1 was obtained in 9.7 g (yield 84%) through column chromatography. HR LC/MS/MS m/z calculated for C82H51BF26N2O6(M+): 1664.3425; found: 1664.3428 Preparation Example 2 Compound 2 (Synthesis of 2-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 2,6-diisopropylphenol. Compound 2-1 was obtained in 14.1 g (yield 87%). (Synthesis of 2-2) Synthesis was progressed according to Synthesis Method 3 using Compound 2-1 and bromine. Compound 2-2 was obtained in 23.1 g (yield 89%). (Synthesis of 2-3) Synthesis was progressed according to Synthesis Method 4 using Compound 2-2 and 4-cyano-2,6-diisopropylphenol. Compound 2-3 was obtained in 10.5 g (yield 77%). (Synthesis of Compound 2) Synthesis was progressed according to Synthesis Method 5 using Compound 2-3 and 2,4-ditrifluoromethylboronic acid. Compound 2 was obtained in 11.1 g (yield 86%). HR LC/MS/MS m/z calculated for C63H57BF14N4O3(M+): 1194.4300; found: 1194.4296 Preparation Example 3 Compound 3 (Synthesis of 3-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 4-cyano-2,6-diisopropylphenol. Compound 3-1 was obtained in 15.5 g (yield 89%). (Synthesis of 3-2) Synthesis was progressed according to Synthesis Method 6 using Compound 3-1. Compound 3-2 was obtained in 9.8 g (yield 68%). (Synthesis of 3-3) Synthesis was progressed according to Synthesis Method 4 using Compound 3-2 and 2,6-dichlorophenol. Compound 3-3 was obtained in 7.4 g (yield 59%). (Synthesis of Compound 3) Synthesis was progressed according to Synthesis Method 5 using Compound 3-3 and 2,4-ditrifluoromethylboronic acid. Compound 3 was obtained in 8.0 g (yield 76%). HR LC/MS/MS m/z calculated for C50H29BCl5F14N3O3(M+): 1171.0521; found: 1171.0525 Preparation Example 4 Compound 4 (Synthesis of 4-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and phenol. Compound 4-1 was obtained in 5.6 g (yield 90%). (Synthesis of 4-2) Synthesis was progressed according to Synthesis Method 2 using Compound 4-1. Compound 4-2 was obtained in 9.7 g (yield 81%). (Synthesis of 4-3) Synthesis was progressed according to Synthesis Method 4 using Compound 4-2 and cyanobenzenethiol. Compound 4-3 was obtained in 9.4 g (yield 90%). (Synthesis of 4-4) Synthesis was progressed according to Synthesis Method 5 using Compound 4-3 and t-butylbenzeneboronic acid. Compound 4-4 was obtained in 8.4 g (yield 82%). (Synthesis of Compound 4) Synthesis was progressed according to Synthesis Method 5 using Compound 4-4 and benzeneboronic acid. Compound 4 was obtained in 6.3 g (yield 79%). HR LC/MS/MS m/z calculated for C55H45BF2N4OS2(M+): 890.3096; found: 890.3094 Preparation Example 5 Compound 5 (Synthesis of 5-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and cyanophenol. Compound 5-1 was obtained in 6.2 g (yield 91%). (Synthesis of 5-2) Synthesis was progressed according to Synthesis Method 2 using Compound 5-1. Compound 5-2 was obtained in 13.1 g (yield 86%). (Synthesis of 5-3) Synthesis was progressed according to Synthesis Method 4 using Compound 5-2 and dibenzofuran-4-thiol. Compound 5-3 was obtained in 14.8 g (yield 87%). (Synthesis of 5-4) Synthesis was progressed according to Synthesis Method 5 using Compound 5-3 and t-butylbenzeneboronic acid. Compound 5-4 was obtained in 11.2 g (yield 76%). (Synthesis of 5-5) Synthesis was progressed according to Synthesis Method 5 using Compound 5-4 and benzeneboronic acid. Compound 5-5 was obtained in 7.9 g (yield 76%). (Synthesis of Compound 5) Synthesis was progressed according to Synthesis Method 4 using Compound 5-5 and cyanophenol. Compound 5 was obtained in 6.4 g (yield 86%). HR LC/MS/MS m/z calculated for C70H44BF2N5O5S2(M+): 1147.2845; found: 1147.2850 Preparation Example 6 Compound 6 (Synthesis of 6-1) Synthesis was progressed according to Synthesis Method 4 using Compound 2-2 and dichlorobenzenethiol. Compound 6-1 was obtained in 5.0 g (yield 78%). (Synthesis of Compound 6) Synthesis was progressed according to Synthesis Method 5 using Compound 6-1 and 4-methoxyphenylboronic acid. Compound 6 was obtained in 4.8 g (yield 90%). HR LC/MS/MS m/z calculated for C47H39BCl4F2N2O3S2(M+): 932.1217; found: 932.1215 Preparation Example 7 Compound 7 (Synthesis of 7-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 4-cyano-2,6-diisopropylbenzenethiol. Compound 7-1 was obtained in 5.8 g (yield 64%). (Synthesis of 7-2) Synthesis was progressed according to Synthesis Method 2 using Compound 7-1. Compound 7-2 was obtained in 7.2 g (yield 73%). (Synthesis of 7-3) Synthesis was progressed according to Synthesis Method 4 using Compound 7-2 and 5′-fluoro-2,2″-bis(trifluoromethyl)terphenyl-2′-ol. Compound 7-3 was obtained in 9.5 g (yield 76%). (Synthesis of 7-4) Synthesis was progressed according to Synthesis Method 5 using Compound 7-3 and dibenzothiopheneboronic acid. Compound 7-4 was obtained in 7.0 g (yield 68%). (Synthesis of Compound 7) Synthesis was progressed according to Synthesis Method 5 using Compound 7-4 and 4-trifluoromethylphenylboronic acid. Compound 7 was obtained in 5.3 g (yield 73%). HR LC/MS/MS m/z calculated for C93H55BF19N3O2S3(M+): 1713.3246; found: 1713.3242 Preparation Example 8 Compound 8 (Synthesis of 8-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and dibenzofuran-4-thiol. Compound 8-1 was obtained in 6.8 g (yield 79%). (Synthesis of 8-2) Synthesis was progressed according to Synthesis Method 2 using Compound 8-1. Compound 8-2 was obtained in 11.2 g (yield 84%). (Synthesis of 8-3) Synthesis was progressed according to Synthesis Method 4 using Compound 8-2 and benzenethiol. Compound 8-3 was obtained in 8.6 g (yield 81%). (Synthesis of 8-4) Synthesis was progressed according to Synthesis Method 5 using Compound 8-3 and 2,4-difluorobenzeneboronic acid. Compound 8-4 was obtained in 6.5 g (yield 76%). (Synthesis of Compound 8) Synthesis was progressed according to Synthesis Method 5 using Compound 8-4 and 4-cyanobenzeneboronic acid. Compound 8 was obtained in 4.8 g (yield 77%). HR LC/MS/MS m/z calculated for C59H31BF6N4OS3(M+): 1032.1657; found: 1032.1653 Preparation Example 9 Compound 9 (Synthesis of 9-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 3,5-dimethoxyphenol. Compound 9-1 was obtained in 6.5 g (yield 86%). (Synthesis of 9-2) Synthesis was progressed according to Synthesis Method 6 using Compound 9-1. Compound 9-2 was obtained in 7.0 g (yield 73%). (Synthesis of 9-3) Synthesis was progressed according to Synthesis Method 4 using Compound 9-2 and 2,6-dimethylphenol. Compound 9-3 was obtained in 7.0 g (yield 76%). (Synthesis of 9-4) Synthesis was progressed according to Synthesis Method 5 using Compound 9-3 and 6-phenyl-dibenzofuranyl-4-boronic acid. Compound 9-4 was obtained in 6.1 g (yield 65%). (Synthesis of Compound 9) Synthesis was progressed according to Synthesis Method 7 using Compound 9-4. Compound 9 was obtained in 2.7 g (yield 45%). HR LC/MS/MS m/z calculated for C70H49BCl2FN3O7(M+): 1143.3025; found: 1143.3027 Preparation Example 10 Compound 10 (Synthesis of 10-1) Synthesis was progressed according to Synthesis Method 2 using Compound 4-1. Compound 10-1 was obtained in 11.5 g (yield 86%). (Synthesis of 10-2) Synthesis was progressed according to Synthesis Method 4 using Compound 10-1 and 2-(2′-trifluoromethylphenyl)-4,6-difluorobenzenethiol. Compound 10-2 was obtained in 13.5 g (yield 79%). (Synthesis of 10-3) Synthesis was progressed according to Synthesis Method 5 using Compound 10-2 and 4-cyanophenylboronic acid. Compound 10-3 was obtained in 10.8 g (yield 80%). (Synthesis of 10-4) Synthesis was progressed according to Synthesis Method 5 using Compound 10-3 and biphenyl-4-ol. Compound 10-4 was obtained in 8.3 g (yield 72%). (Synthesis of Compound 10) Synthesis was progressed according to Synthesis Method 7 using Compound 10-4. Compound 10 was obtained in 4.1 g (yield 51%). HR LC/MS/MS m/z calculated for C80H43BF11N5O3S2(M+): 1405.2725; found: 1405.2730 Preparation Example 11 Compound 11 (Synthesis of 11-1) Synthesis was progressed according to Synthesis Method 2 using Compound 7-1. Compound 11-1 was obtained in 9.1 g (yield 84%). (Synthesis of 11-2) Synthesis was progressed according to Synthesis Method 4 using Compound 11-1 and dibenzofuran-4-ol. Compound 11-2 was obtained in 8.7 g (yield 79%). (Synthesis of 11-3) Synthesis was progressed according to Synthesis Method 5 using Compound 11-2 and 2,4-di(trifluoromethyl)phenylboronic acid. Compound 11-3 was obtained in 7.2 g (yield 72%). (Synthesis of 11-4) Synthesis was progressed according to Synthesis Method 5 using Compound 11-3 and phenylboronic acid. Compound 11-4 was obtained in 4.7 g (yield 68%). (Synthesis of Compound 11) Synthesis was progressed according to Synthesis Method 7 using Compound 11-4. Compound 11 was obtained in 2.0 g (yield 49%). HR LC/MS/MS m/z calculated for C75H46BF13N4O4S (M+): 1356.3125; found: 1356.3129 Preparation Example 12 Compound 12 (Synthesis of 12-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 5′-methoxy-terphenyl-2′-thiol. Compound 12-1 was obtained in 8.1 g (yield 76%). (Synthesis of 12-2) Synthesis was progressed according to Synthesis Method 6 using Compound 12-1. Compound 12-2 was obtained in 7.9 g (yield 69%). (Synthesis of 12-3) Synthesis was progressed according to Synthesis Method 4 using Compound 12-2 and 2,4-di(trifluoromethyl)benzenethiol. Compound 12-3 was obtained in 7.2 g (yield 64%). (Synthesis of 12-4) Synthesis was progressed according to Synthesis Method 5 using Compound 12-3 and 4-methoxyphenylboronic acid. Compound 12-4 was obtained in 6.8 g (yield 86%). (Synthesis of Compound 12) Synthesis was progressed according to Synthesis Method 7 using Compound 12-4. Compound 12 was obtained in 3.0 g (yield 49%). HR LC/MS/MS m/z calculated for C59H35BCl2F13N3O3S3(M+): 1257.1103; found: 1257.1106 Preparation Example 13 Compound 13 (Synthesis of 13-1) Synthesis was progressed according to Synthesis Method 3 using Compound 3-1. Compound 13-1 was obtained in 7.9 g (yield 88%). (Synthesis of 13-2) Synthesis was progressed according to Synthesis Method 4 using Compound 13-1 and 4-trifluoromethylphenol. Compound 13-2 was obtained in 7.3 g (yield 85%). (Synthesis of 13-3) Synthesis was progressed according to Synthesis Method 5 using Compound 13-2 and dibenzofuranyl-4-boronic acid. Compound 13-3 was obtained in 5.2 g (yield 68%). (Synthesis of 13-4) Synthesis was progressed according to Synthesis Method 5 using Compound 13-3 and phenylboronic acid. Compound 13-4 was obtained in 4.3 g (yield 86%). (Synthesis of Compound 13) Synthesis was progressed according to Synthesis Method 7 using Compound 13-4. Compound 13 was obtained in 1.8 g (yield 44%). HR LC/MS/MS m/z calculated for C56H38BF6N5O4(M+): 696.2921; found: 696.2918 Preparation Example 14 Compound 14 (Synthesis of 14-1) Synthesis was progressed according to Synthesis Method 4 using Compound 2-2 and 3,5-dimethoxyphenol. Compound 14-1 was obtained in 4.8 g (yield 79%). (Synthesis of 14-2) Synthesis was progressed according to Synthesis Method 5 using Compound 14-1 and 4-cyanophenylboronic acid. Compound 14-2 was obtained in 3.8 g (yield 89%). (Synthesis of 14-3) Synthesis was progressed according to Synthesis Method 8 using Compound 14-2 and cyclopentyl potassium trifluoroborate. Compound 14-3 was obtained in 2.1 g (yield 61%). (Synthesis of Compound 14) Synthesis was progressed according to Synthesis Method 7 using Compound 14-3. Compound 14 was obtained in 1.6 g (yield 78%). HR LC/MS/MS m/z calculated for C63H61BN6O7(M+): 1024.4695; found: 1024.4693 Preparation Example 15 Compound 15 (Synthesis of 15-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 5′-fluoro-terphenyl-2′-ol. Compound 15-1 was obtained in 7.2 g (yield 72%). (Synthesis of 15-2) Synthesis was progressed according to Synthesis Method 2 using Compound 15-1. Compound 15-2 was obtained in 12.2 g (yield 85%). (Synthesis of 15-3) Synthesis was progressed according to Synthesis Method 4 using Compound 15-2 and 7-hydroxycoumarin. Compound 15-3 was obtained in 11.6 g (yield 82%). (Synthesis of 15-4) Synthesis was progressed according to Synthesis Method 5 using Compound 15-3 and 2,6-dimethylphenylboronic acid. Compound 15-4 was obtained in 9.1 g (yield 79%). (Synthesis of 15-5) Synthesis was progressed according to Synthesis Method 5 using Compound 15-4 and 3-fluorophenylboronic acid. Compound 15-5 was obtained in 6.3 g (yield 68%). (Synthesis of Compound 15) Synthesis was progressed according to Synthesis Method 7 using Compound 15-5. Compound 15 was obtained in 4.4 g (yield 73%). HR LC/MS/MS m/z calculated for C75H48BF3N4O7(M+): 1184.3568; found: 1184.3571 Preparation Example 16 Compound 16 (Synthesis of 16-1) Synthesis was progressed according to Synthesis Method 9 using Compound 2-1. Compound 16-1 was obtained in 4.3 g (yield 80%). (Synthesis of 16-2) Synthesis was progressed according to Synthesis Method 3 using Compound 16-1. Compound 16-2 was obtained in 5.0 g (yield 79%). (Synthesis of 16-3) Synthesis was progressed according to Synthesis Method 4 using Compound 16-2 and benzenethiol. Compound 16-3 was obtained in 4.7 g (yield 86%). (Synthesis of 16-4) Synthesis was progressed according to Synthesis Method 5 using Compound 16-3 and benzeneboronic acid. Compound 16-4 was obtained in 3.2 g (yield 81%). (Synthesis of 16-5) Synthesis was progressed according to Synthesis Method 10 using Compound 16-4. Compound 16-5 was obtained in 2.2 g (yield 72%). (Synthesis of 16-6) Synthesis was progressed according to Synthesis Method 11 using Compound 16-5 and 7-hydroxycoumarin. Compound 16-6 was obtained in 2.2 g (yield 91%). (Synthesis of 16-7) Synthesis was progressed according to Synthesis Method 8 using Compound 16-6 and cyclohexyl potassium trifluoroborate. Compound 16-7 was obtained in 1.5 g (yield 68%). (Synthesis of Compound 16) Synthesis was progressed according to Synthesis Method 7 using Compound 16-7. Compound 16 was obtained in 1.4 g (yield 91%). HR LC/MS/MS m/z calculated for C57H49BN4O5S2(M+): 944.3237; found: 944.3235 Preparation Example 17 Compound 17 (Synthesis of 17-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 2,4,6-trimethylphenol. Compound 17-1 was obtained in 6.2 g (yield 86%). (Synthesis of 17-2) Synthesis was progressed according to Synthesis Method 2 using Compound 17-1. Compound 17-2 was obtained in 12.4 g (yield 84%). (Synthesis of 17-3) Synthesis was progressed according to Synthesis Method 12 using Compound 17-2 and indium hexafluoropropane-2-thiolate. Compound 17-3 was obtained in 6.3 g (yield 42%). (Synthesis of 17-4) Synthesis was progressed according to Synthesis Method 5 using Compound 17-3 and 2-methoxyphenylboronic acid. Compound 17-4 was obtained in 5.4 g (yield 86%). (Synthesis of 17-5) Synthesis was progressed according to Synthesis Method 5 using Compound 17-4 and phenylboronic acid. Compound 17-5 was obtained in 3.6 g (yield 72%). (Synthesis of Compound 17) Synthesis was progressed according to Synthesis Method 7 using Compound 17-5. Compound 17 was obtained in 2.3 g (yield 76%). HR LC/MS/MS m/z calculated for C52H37BF12N4O3S2(M+): 1068.2209; found: 1068.2213 Preparation Example 18 Compound 18 (Synthesis of 18-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 5′-methoxyterphenyl-2′-ol. Compound 18-1 was obtained in 8.9 g (yield 86%). (Synthesis of 18-2) Synthesis was progressed according to Synthesis Method 2 using Compound 18-1. Compound 18-2 was obtained in 13.1 g (yield 81%). (Synthesis of 18-3) Synthesis was progressed according to Synthesis Method 4 using Compound 18-2 and 5′-cyanoterphenyl-2′-thiol. Compound 18-3 was obtained in 14.2 g (yield 76%). (Synthesis of 18-4) Synthesis was progressed according to Synthesis Method 5 using Compound 18-3 and benzeneboronic acid. Compound 18-4 was obtained in 11.6 g (yield 83%). (Synthesis of 18-5) Synthesis was progressed according to Synthesis Method 4 using Compound 18-4 and 4-cyanophenol. Compound 18-5 was obtained in 8.7 g (yield 75%). (Synthesis of Compound 18) Synthesis was progressed according to Synthesis Method 7 using Compound 18-5. Compound 18 was obtained in 5.8 g (yield 72%). HR LC/MS/MS m/z calculated for C94H57BN8O4S2(M+): 1436.4037; found: 1436.4040 Preparation Example 19 Compound 19 (Synthesis of 19-1) Synthesis was progressed according to Synthesis Method 9 using Compound 8-1. Compound 19-1 was obtained in 4.8 g (yield 84%). (Synthesis of 19-2) Synthesis was progressed according to Synthesis Method 3 using Compound 19-1. Compound 19-2 was obtained in 5.9 g (yield 86%). (Synthesis of 19-3) Synthesis was progressed according to Synthesis Method 4 using Compound 19-2 and 2-methylcyclohexanol. Compound 19-3 was obtained in 3.5 g (yield 64%). (Synthesis of 19-4) Synthesis was progressed according to Synthesis Method 10 using Compound 19-3. Compound 19-4 was obtained in 2.5 g (yield 79%). (Synthesis of 19-5) Synthesis was progressed according to Synthesis Method 11 using Compound 19-4 and 7-hydroxycoumarin. Compound 19-5 was obtained in 2.4 g (yield 91%). (Synthesis of 19-6) Synthesis was progressed according to Synthesis Method 4 using Compound 19-5 and 2-methylphenol. Compound 19-6 was obtained in 2.0 g (yield 96%). (Synthesis of Compound 19) Synthesis was progressed according to Synthesis Method 7 using Compound 19-6. Compound 19 was obtained in 1.6 g (yield 86%). HR LC/MS/MS m/z calculated for C71H57BN4O13S (M+): 1216.3736; found: 1216.3739 Preparation Example 20 Compound 20 (Synthesis of 20-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 4-mercaptobenzonitrile. Compound 20-1 was obtained in 6.2 g (yield 86%). (Synthesis of 20-2) Synthesis was progressed according to Synthesis Method 9 using Compound 20-1. Compound 20-2 was obtained in 5.3 g (yield 82%). (Synthesis of 20-3) Synthesis was progressed according to Synthesis Method 2 using Compound 20-2. Compound 20-3 was obtained in 7.8 g (yield 74%). (Synthesis of 20-4) Synthesis was progressed according to Synthesis Method 4 using Compound 20-3 and 4-cyano-2,6-diisopropylbenzenethiol. Compound 20-4 was obtained in 7.4 g (yield 77%). (Synthesis of 20-5) Synthesis was progressed according to Synthesis Method 5 using Compound 20-4 and benzeneboronic acid. Compound 20-5 was obtained in 5.0 g (yield 72%). (Synthesis of 20-6) Synthesis was progressed according to Synthesis Method 10 using Compound 20-5. Compound 20-6 was obtained in 3.5 g (yield 68%). (Synthesis of 20-7) Synthesis was progressed according to Synthesis Method 11 using Compound 20-6 and 7-hydroxycoumarin. Compound 20-7 was obtained in 3.1 g (yield 91%). (Synthesis of Compound 20) Synthesis was progressed according to Synthesis Method 7 using Compound 20-7. Compound 20 was obtained in 2.5 g (yield 84%). HR LC/MS/MS m/z calculated for C60H46BBr2N7O4S3(M+): 1193.1233; found: 1193.1230 Preparation Example 21 Compound 21 (Synthesis of 21-1) Synthesis was progressed according to Synthesis Method 4 using Compound 18-2 and 4-(9H-carbazol-9-yl)phenol. Compound 21-1 was obtained in 10.5 g (yield 76%). (Synthesis of 21-2) Synthesis was progressed according to Synthesis Method 5 using Compound 21-1 and benzeneboronic acid. Compound 21-2 was obtained in 6.8 g (yield 68%). (Synthesis of 21-3) Synthesis was progressed according to Synthesis Method 5 using Compound 21-2 and 4-trifluoromethylphenylboronic acid. Compound 21-3 was obtained in 4.7 g (yield 75%). (Synthesis of 21-4) Synthesis was progressed according to Synthesis Method 5 using Compound 21-3 and 2-methoxyphenylboronic acid. Compound 21-4 was obtained in 3.0 g (yield 81%). (Synthesis of Compound 21) Synthesis was progressed according to Synthesis Method 13 using Compound 21-4. Compound 21 was obtained in 1.3 g (yield 51%). HR LC/MS/MS m/z calculated for C86H55BF18N4O7(M+): 1608.3876; found: 1608.3872 Preparation Example 22 Compound 22 (Synthesis of 22-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 2-(pyridin-2-yl)phenol. Compound 22-1 was obtained in 5.0 g (yield 63%). (Synthesis of 22-2) Synthesis was progressed according to Synthesis Method 2 using Compound 22-1. Compound 22-2 was obtained in 7.6 g (yield 73%). (Synthesis of 22-3) Synthesis was progressed according to Synthesis Method 4 using Compound 22-2 and 5′-fluoroterphenyl-2′-thiol. Compound 22-3 was obtained in 8.6 g (yield 80%). (Synthesis of 22-4) Synthesis was progressed according to Synthesis Method 5 using Compound 22-3 and phenylboronic acid. Compound 22-4 was obtained in 6.2 g (yield 78%). (Synthesis of Compound 22) Synthesis was progressed according to Synthesis Method 13 using Compound 22-4. Compound 22 was obtained in 3.1 g (yield 39%). HR LC/MS/MS m/z calculated for C82H52BF16N3O3S2(M+): 1505.3288; found: 1505.3291 Preparation Example 23 Compound 23 (Synthesis of 23-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and terphenyl-2′-thiol. Compound 23-1 was obtained in 6.5 g (yield 65%). (Synthesis of 23-2) Synthesis was progressed according to Synthesis Method 2 using Compound 23-1. Compound 23-2 was obtained in 10.7 g (yield 87%). (Synthesis of 23-3) Synthesis was progressed according to Synthesis Method 4 using Compound 23-2 and 5′-(t-butyl)-terphenyl-2′-ol. Compound 23-3 was obtained in 10.8 g (yield 73%). (Synthesis of 23-4) Synthesis was progressed according to Synthesis Method 5 using Compound 23-3 and phenylboronic acid. Compound 23-4 was obtained in 6.4 g (yield 64%). (Synthesis of 23-5) Synthesis was progressed according to Synthesis Method 5 using Compound 23-4 and 4-aminophenylboronic acid. Compound 23-5 was obtained in 4.4 g (yield 73%). (Synthesis of 23-6) Synthesis was progressed according to Synthesis Method 4 using Compound 23-5 and 2-trifluoromethylphenol. Compound 23-6 was obtained in 3.6 g (yield 81%). (Synthesis of Compound 23) Synthesis was progressed according to Synthesis Method 13 using Compound 23-6. Compound 23 was obtained in 1.6 g (yield 43%). HR LC/MS/MS m/z calculated for C105H78BF20N3O6S (M+): 1899.5385; found: 1899.5382 Preparation Example 24 Compound 24 (Synthesis of 24-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 2,4,6-trimethylbenzenethiol. Compound 24-1 was obtained in 6.2 g (yield 82%). (Synthesis of 24-2) Synthesis was progressed according to Synthesis Method 3 using Compound 24-1. Compound 24-2 was obtained in 9.7 g (yield 84%). (Synthesis of 24-3) Synthesis was progressed according to Synthesis Method 4 using Compound 24-2 and 2,6-diisopropylbenzenethiol. Compound 24-3 was obtained in 9.8 g (yield 81%). (Synthesis of 24-4) Synthesis was progressed according to Synthesis Method 5 using Compound 24-3 and 2,4-difluorophenylboronic acid. Compound 24-4 was obtained in 8.6 g (yield 89%). (Synthesis of Compound 24) Synthesis was progressed according to Synthesis Method 13 using Compound 24-4. Compound 24 was obtained in 5.3 g (yield 48%). HR LC/MS/MS m/z calculated for C62H57BF18N2O2S3(M+): 1310.3388; found: 1310.3390 Preparation Example 25 Compound 25 (Synthesis of 25-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 4′-hydroxy-3′,5′-diisopropyl-biphenyl-4-carbonitrile. Compound 25-1 was obtained in 7.8 g (yield 75%). (Synthesis of 25-2) Synthesis was progressed according to Synthesis Method 2 using Compound 25-1. Compound 25-2 was obtained in 8.8 g (yield 68%). (Synthesis of 25-3) Synthesis was progressed according to Synthesis Method 4 using Compound 25-2 and 2,4-difluorophenol. Compound 25-3 was obtained in 7.1 g (yield 80%). (Synthesis of 25-4) Synthesis was progressed according to Synthesis Method 5 using Compound 25-3 and dibenzothiophenyl-4-boronic acid. Compound 25-4 was obtained in 4.6 g (yield 59%). (Synthesis of 25-5) Synthesis was progressed according to Synthesis Method 5 using Compound 25-4 and phenylboronic acid. Compound 25-5 was obtained in 2.5 g (yield 63%). (Synthesis of 25-6) Synthesis was progressed according to Synthesis Method 4 using Compound 25-5 and phenol. Compound 25-6 was obtained in 4.6 g (yield 78%). (Synthesis of Compound 25) Synthesis was progressed according to Synthesis Method 14 using Compound 25-6. Compound 25 was obtained in 0.8 g (yield 62%). HR LC/MS/MS m/z calculated for C88H70BF4N3O4S (M+): 1351.5116; found: 1351.5118 Preparation Example 26 Compound 26 (Synthesis of 26-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and terphenyl-2′-ol. Compound 26-1 was obtained in 8.3 g (yield 86%). (Synthesis of 26-2) Synthesis was progressed according to Synthesis Method 3 using Compound 26-1. Compound 26-2 was obtained in 11.2 g (yield 81%). (Synthesis of 26-3) Synthesis was progressed according to Synthesis Method 4 using Compound 26-2 and 2-(2-pyridinyl)-benzenethiol. Compound 26-3 was obtained in 10.2 g (yield 72%). (Synthesis of 26-4) Synthesis was progressed according to Synthesis Method 5 using Compound 26-3 and phenylboronic acid. Compound 26-4 was obtained in 7.7 g (yield 77%). (Synthesis of 26-5) Synthesis was progressed according to Synthesis Method 8 using Compound 26-4 and cyclopropyl potassium trifluoroborate. Compound 26-5 was obtained in 4.8 g (yield 63%). (Synthesis of Compound 26) Synthesis was progressed according to Synthesis Method 14 using Compound 26-5. Compound 26 was obtained in 2.8 g (yield 56%). HR LC/MS/MS m/z calculated for C91H75BN4OS2(M+): 1314.5475; found: 1314.5473 Preparation Example 27 Compound 27 (Synthesis of 27-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 2-trifluoromethylbenzenethiol. Compound 27-1 was obtained in 6.3 g (yield 78%). (Synthesis of 27-2) Synthesis was progressed according to Synthesis Method 3 using Compound 27-1. Compound 27-2 was obtained in 7.5 g (yield 85%). (Synthesis of 27-3) Synthesis was progressed according to Synthesis Method 4 using Compound 27-2 and 2′-hydroxy-2,2″-bistrifluoromethyl-terphenyl-5′-carbonitrile. Compound 27-3 was obtained in 12.8 g (yield 73%). (Synthesis of 27-4) Synthesis was progressed according to Synthesis Method 5 using Compound 27-3 and 4-(phenoxycarbonyl)phenylboronic acid. Compound 27-4 was obtained in 8.6 g (yield 61%). (Synthesis of 27-5) Synthesis was progressed according to Synthesis Method 8 using Compound 27-4 and cyclohexyl potassium trifluoroborate. Compound 27-5 was obtained in 5.0 g (yield 57%). (Synthesis of Compound 27) Synthesis was progressed according to Synthesis Method 14 using Compound 27-5. Compound 27 was obtained in 2.0 g (yield 34%). HR LC/MS/MS m/z calculated for C120H90BF15N4O6S (M+): 2010.6435; found: 2010.6439 Preparation Example 28 Compound 28 (Synthesis of 28-1) Synthesis was progressed according to Synthesis Method 1 using chloro BODIPY and 7-mercapto-2H-chromen-2-one. Compound 28-1 was obtained in 11.9 g (yield 73%). (Synthesis of 28-2) Synthesis was progressed according to Synthesis Method 2 using Compound 28-1. Compound 28-2 was obtained in 21.1 g (yield 84%). (Synthesis of 28-3) Synthesis was progressed according to Synthesis Method 4 using Compound 28-2 and 4-aminobenzenethiol. Compound 28-3 was obtained in 15.8 g (yield 68%). (Synthesis of 28-4) Synthesis was progressed according to Synthesis Method 5 using Compound 28-3 and 4-aminobenzeneboronic acid. Compound 28-4 was obtained in 11.2 g (yield 73%). (Synthesis of 28-5) Synthesis was progressed according to Synthesis Method 12 using Compound 28-4 and indium(III)propan-2-olate. Compound 28-5 was obtained in 4.9 g (yield 47%). (Synthesis of Compound 28) Synthesis was progressed according to Synthesis Method 14 using Compound 28-5. Compound 28 was obtained in 2.7 g (yield 52%). HR LC/MS/MS m/z calculated for C72H69BN6O4S3(M+): 1188.4635; found: 1188.4639 EXAMPLE Example 1 A first solution was prepared by dissolving Compound 1, an organic fluorescent substance, in a xylene solvent. A second solution was prepared by dissolving a thermoplastic resin SAN (styrene-acrylonitrile-based) in a xylene solvent. The first solution and the second solution were mixed so that the amount of the organic fluorescent substance was 0.5 parts by weight based on 100 parts by weight of the SAN, and the result was homogeneously mixed. The solid content in the mixed solution was 20% by weight and viscosity was 200 cps. This solution was coated on a PET substrate, and the result was dried to prepare a color conversion film. A luminance spectrum of the prepared color conversion film was measured using a spectroradiometer (SR series of TOPCON Corporation). Specifically, the prepared color conversion film was laminated on one surface of a light guide plate of a backlight unit including an LED blue backlight (maximum light emission wavelength 450 nm) and the light guide plate, and after laminating a prism sheet and a DBEF film on the color conversion film, a luminance spectrum of the film was measured. When measuring the luminance spectrum, an initial value was set so that the brightness of the blue LED light was 600 nit based on without the color conversion film. Example 2 An experiment was performed in the same manner as in Example 1 except that Compound 2 was used instead of Compound 1. Example 3 An experiment was performed in the same manner as in Example 1 except that Compound 4 was used instead of Compound 1. Example 4 An experiment was performed in the same manner as in Example 1 except that Compound 6 was used instead of Compound 1. Example 5 An experiment was performed in the same manner as in Example 1 except that Compound 11 was used instead of Compound 1. Example 6 An experiment was performed in the same manner as in Example 1 except that Compound 12 was used instead of Compound 1. Example 7 An experiment was performed in the same manner as in Example 1 except that Compound 18 was used instead of Compound 1. Example 8 An experiment was performed in the same manner as in Example 1 except that Compound 20 was used instead of Compound 1. Example 9 An experiment was performed in the same manner as in Example 1 except that Compound 21 was used instead of Compound 1. Example 10 An experiment was performed in the same manner as in Example 1 except that Compound 23 was used instead of Compound 1. Example 11 An experiment was performed in the same manner as in Example 1 except that Compound 27 was used instead of Compound 1. Example 12 An experiment was performed in the same manner as in Example 1 except that Compound 28 was used instead of Compound 1. Comparative Example 1 An experiment was performed in the same manner as in Example 1 except that diPh was used instead of Compound 1. Comparative Example 2 An experiment was performed in the same manner as in Example 1 except that diPhO was used instead of Compound 1. Comparative Example 3 An experiment was performed in the same manner as in Example 1 except that OdiPh was used instead of Compound 1. Comparative Example 4 An experiment was performed in the same manner as in Example 1 except that diPhS was used instead of Compound 1. Comparative Example 5 An experiment was performed in the same manner as in Example 1 except that SdiPh was used instead of Compound 1. Thin film light emission wavelength, PLQY (thin film quantum efficiency) and PL intensity (%) of each of the color conversion films according to Examples 1 to 12 and Comparative Examples 1 to 5 are as shown in the following Table 4. TABLE 4Thin FilmLight EmissionPLWavelength PLλmaxPLQYIntensity(nm)(%)(%)Example 1Compound 15209793Example 2Compound 25319494Example 3Compound 46209682Example 4Compound 66269586Example 5Compound 116249691Example 6Compound 126829788Example 7Compound 186139592Example 8Compound 206899491Example 9Compound 215559486Example 10Compound 235369384Example 11Compound 276299495Example 12Compound 287039293ComparativediPh6229054Example 1ComparativediPhO5503249Example 2ComparativeOdiPh5198552Example 3ComparativediPhS6165653Example 4ComparativeSdiPh6227848Example 5 When a color conversion film has low stability, there is a problem in that a wavelength of light finally appearing after passing through a light source and a film continuously changes over time. According to Table 4, it was identified that the color conversion films according to Examples 1 to 12 had small changes in the PL intensity compared to Comparative Examples 1 to 5 leading to small changes in the wavelength, and therefore, light emission efficiency was high and stability was excellent. The thin film light emission wavelength (PL λmax(nm)) was measured using FS-2 equipment of SCINCO Co., Ltd., and the thin film quantum efficiency (PLQY) was measured using Quantaurus-QY equipment of HAMAMATSU Photonics K.K. PL intensity (%) is a value obtained by, based on PL of a manufactured film, irradiating an LED light source for 1,000 hours on the corresponding film, measuring PL again, and calculating a difference in the intensity from the initial value.
103,829
11858953
DETAILED DESCRIPTION The present invention is based, in part, on the discovery that an organic molecule comprising at least one phosphate moiety can be used as a phosphorylation reagent. In one embodiment, the organic molecule comprising at least one phosphate moiety can be used as a phosphorylation reagent to synthesize a phosphorylated molecule through a reaction of the organic molecule comprising at least one phosphate moiety with an activated precursor molecule. The compositions and methods disclosed herein can be used to synthesize phosphorylated sugar molecules, phosphorylated proteins or peptides, phosphorylated lipids, or phosphorylated nucleic acid molecules. Thus, in various aspects the invention provides compositions and methods for synthesizing natural and modified phosphorylated molecules. The compositions and methods disclosed herein can be used to synthesize phosphorylated nucleosides. In one embodiment, the organic phosphate molecule of the invention can be an organic pyrophosphate molecule for use in the synthesis of nucleoside triphosphates from an activated nucleoside monophosphate molecule. In one embodiment, the organic phosphate molecule of the invention can be an organic monophosphate molecule for use in the synthesis of nucleoside diphosphates from an activated nucleoside monophosphate molecule. In one embodiment, the organic phosphate molecule of the invention can be an organic triphosphate molecule for use in the synthesis of nucleoside tetraphosphates from an activated nucleoside monophosphate molecule, in one embodiment, the organic phosphate molecule of the invention can be an organic tetraphosphate molecule for use in the synthesis of nucleoside pentaphosphates from an activated nucleoside monophosphate molecule. In one embodiment, the organic phosphate molecule of the invention can be an organic pentaphosphate molecule for use in the synthesis of nucleoside hexaphosphates from an activated nucleoside monophosphate molecule. Definitions Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are described. As used herein, each of the following terms has the meaning associated with it in this section. The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element. “About” as used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass non-limiting variations of ±40% or ±20% or ±10%, ±5%, ±1%, or ±0.1% from the specified value, as such variations are appropriate. “Acid catalyst,” as used herein, refers to any acidic compounds including the so-called Lewis acids, which catalyze the reaction between an activated nucleoside monophosphate and a pyrophosphate containing compound. Examples of acids used for this purpose include ZnCl2, H2SO4, HCl, H3PO4or BF3, or sulfonic acids or their salts. Examples of sulfonic acids comprise ortho-, meta- and para-toluenesulfonic acids, alkylbenzenesulfonic acids, secondary alkyl-sulfonic acids, sulfonic resins, alkylsulfates, alkylbenzenesulfonates, alkyl-sulfonates and sulfosuccinic acid. As used herein, the term “purified” or “to purify” refers to the removal of components (e.g., contaminants) from a sample. For example, nucleic acids are purified by removal of contaminating cellular proteins or other undesired nucleic acid species. The removal of contaminants results in an increase in the percentage of a desired compound in the sample. A “natural” nucleoside is one that occurs in nature. For the purposes of this invention the following nucleosides are defined as the natural nucleosides: adenosine, cytidine, guanosine, uridine, 2′-deoxyadenosine, 2′-deoxycytidine, T-deoxyguanosine, thymidine, and inosine. The term base, unless otherwise specified, refers to the base moiety of a nucleoside or nucleotide (a nucleobases). The base moiety is the heterocycle portion of a nucleoside or nucleotide. The base moiety may be a pyrimidine derivative or analog, a purine derivative or analog, or other heterocycle. The nucleoside base may contain two or more nitrogen atoms and may contain one or more peripheral substitutents. The nucleoside base is attached to the sugar moiety of the nucleotide mimic in such ways that both β-D- and β-L-nucleoside and nucleotide can be produced. The term sugar refers to the ribofuranose of deoxyribofuranose portion of a nucleoside or nucleotide. The sugar moiety may contain one or more substitutents at the C1-, C2-, C3-, C4-, and C5-position of the ribofuranose. Substituents may direct to either the α- or β-face of the ribofuranose. The nucleoside base that can be considered as a substitutent at the C-1 position of the ribofuranose directs to the β-face of the sugar. The β-face is the side of a ribofuranose on which a purine or pyrimidine base of natural β-D-nucleosides is present. The α-face is the side of the sugar opposite to the β-face. The sugar moiety of the present invention is not limited to a ribofuranose and its derivatives, instead, it may be a carbohydrate, a carbohydrate analog, a carbocyclic ring, or other ribofuranose analog. The term sugar-modified nucleoside refers to a nucleoside containing a modified sugar moiety. The term base-modified nucleoside refers to a nucleoside containing a modified base moiety, relative to a base moiety found in a natural nucleoside. As used herein, the term “nucleic acid” refers to both naturally-occurring molecules such as DNA and RNA, but also various derivatives and analogs. Generally, the probes, hairpin linkers, and target polynucleotides of the present teachings are nucleic acids, and typically comprise DNA. Additional derivatives and analogs can be employed as will be appreciated by one having ordinary skill in the art. The term “nucleotide base”, as used herein, refers to a substituted or unsubstituted aromatic ring or rings in certain embodiments, the aromatic ring or rings contain at least one nitrogen atom. In certain embodiments, the nucleotide base is capable of forming Watson-Crick and/or Hoogsteen hydrogen bonds with an appropriately complementary nucleotide base. Exemplary nucleotide bases and analogs thereof include, but are not limited to, naturally occurring nucleotide bases adenine, guanine, cytosine, 6 methyl-cytosine, uracil, thymine, and analogs of the naturally occurring nucleotide bases, e.g., 7-deazaadenine, 7-deazaguanine, 7-deaza-8-azaguanine, 7-deaza-8-azaadenine, N6 delta 2-isopentenyladenine (6iA), N6-delta 2-isopentenyl-2-methylthioadenine (2 ms6iA), N2-dimethylguanine (dmG), 7methylguanine (7mG), inosine, nebularine, 2-aminopurine, 2-amino-6-chloropurine, 2,6-diaminopurine, hypoxanthine, pseudouridine, pseudocytosine, pseudoisocytosine, 5-propynylcytosine, isocytosine, isoguanine, 7-deazaguanine, 2-thiopyrimidine, 6-thioguanine, 4-thiothymine, 4-thiouracil, 06-methylguanine, N6-methyladenine, 04-methylthymine, 5,6-dihydrothymine, 5,6-dihydrouracil, pyrazolo[3,4-D]pyrimidines (see, e.g., U.S. Pat. Nos. 6,143,877 and 6,127,121 and PCT published application WO 01/38584), ethenoadenine, indoles such as nitroindole and 4-methylindole, and pyrroles such as nitropyrrole. Certain exemplary nucleotide bases can be found, e.g., in Fasman, 1989, Practical Handbook of Biochemistry and Molecular Biology, pp. 385-394, CRC Press, Boca. Raton, Fla., and the references cited therein. The term “nucleotide”, as used herein, refers to a compound comprising a nucleotide base linked to the C-1′ carbon of a sugar, such as ribose, arabinose, xylose, and pyranose, and sugar analogs thereof. The term nucleotide also encompasses nucleotide analogs. The sugar may be substituted or unsubstituted. Substituted ribose sugars include, but are not limited to, those riboses in which one or more of the carbon atoms, for example the 2′-carbon atom, is substituted with one or more of the same or different Cl, F, —R, —OR, —NR2 or halogen groups, where each R is independently H, C1-C6 alkyl or C5-C14 aryl. Exemplary riboses include, but are not limited to, 2′-(C1-C6)alkoxyribose, 2′-(C5-C14)aryloxyribose, 2′,3′-didehydroribose, 2′-deoxy-3′-haloribose, 2′-deoxy-3′-fluororibose, 2′-deoxy-3′-chlororibose, 2′-deoxy-3′-aminoribose, 2′-deoxy-3′-(C1-C6)alkylribose, 2′-deoxy-3′-(C1-C6)alkoxyribose and 2′-deoxy-3′-(C5-C14)aryloxyribose, ribose, 2′-deoxyribose, 2′,3′-dideoxyribose, 2′-haloribose, 2′-fluororibose, 2′-chlororibose, and 2′-alkylribose, e.g., 2′-O-methyl, 4′-anomeric nucleotides, 1′-anomeric nucleotides, 2′-4′- and 3′-4′-linked and other “locked” or “LNA”, bicyclic sugar modifications (see, e.g., PCT published application nos. WO 98/22489, WO 98/39352; and WO 99/14226). The term “nucleic acid” typically refers to large polynucleotides. The term “nucleotide analogs” as used herein refers to modified or non-naturally occurring nucleotides including, but not limited to, analogs that have altered stacking interactions such as 7-deaza purines 7-deaza-dATP and 7-deaza-dGTP); base analogs with alternative hydrogen bonding configurations such as Iso-C and iso-G and other non-standard base pairs described in U.S. Pat. No. 6,001,983 to S. Benner and herein incorporated by reference); non-hydrogen bonding analogs non-polar, aromatic nucleoside analogs such as 2,4-difluorotoluene, described by B. A. Schweitzer and E. T. Kool, Org, Chem., 1994, 59, 7238-7242; B. A. Schweitzer and E. T. Kool, J. Am. Chem. Soc., 1995, 117, 1863-1872); “universal” bases such as 5-nitroindole and 3-nitropyrrole; and universal purines and pyrimidines (such as “K” and “P” nucleotides, respectively; Kong, et al., Nucleic Acids Res., 1989, 17, 10373-10383, P. Kong et al., Nucleic Acids Res., 1992, 20, 5149-5152). Nucleotide analogs include nucleotides having modification on the sugar moiety, such as dideoxy nucleotides and 2′-O-methyl nucleotides. Nucleoside analogue examples wherein the natural sugar moiety is modified include but are not limited to hexitol nucleic acid (HNA), cyclohexene nucleic acids (CeNA), locked nucleic acids (LNA), altritol nucleic acids (ANA) and peptide nucleic acids (PNA). Nucleotide analogs include modified forms of deoxyribo-nucleotides as well as ribonucleotides. The term “oligonucleotide” typically refers to short polynucleotides, generally, no greater than about 50 nucleotides. It will be understood that when a nucleotide sequence is represented by a DNA sequence (i.e., A, T, G, C), this also includes an RNA sequence (i.e., A, U, G, C) in which “U” replaces “T.” The term “polynucleotide” as used herein is defined as a chain of nucleotides. Furthermore, nucleic acids are polymers of nucleotides. Thus, nucleic acids and polynucleotides as used herein are interchangeable. One skilled in the art has the general knowledge that nucleic acids are polynucleotides, which can be hydrolyzed into the monomeric “nucleotides,” The monomeric nucleotides can be hydrolyzed into nucleosides. As used herein polynucleotides include, but are not limited to, all nucleic acid sequences which are obtained by any means available in the art, including, without limitation, recombinant means, i.e., the cloning of nucleic acid sequences from a recombinant library or a cell genome, using ordinary cloning and amplification technology, and the like, and by synthetic means. An “oligonucleotide” as used herein refers to a short polynucleotide, typically less than 100 bases in length. As used herein, the term “pharmaceutical composition” refers to a mixture of at least one compound useful within the invention with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration. A “therapeutic” treatment is a treatment administered to a subject who exhibits signs of pathology, for the purpose of diminishing or eliminating those signs. As used herein, the term “treatment” or “treating” is defined as the application or administration of a therapeutic agent, i.e., a compound of the invention (alone or in combination with another pharmaceutical agent), to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications), who has a condition contemplated herein, a sign or symptom of a condition contemplated herein or the potential to develop a condition contemplated herein, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect a condition contemplated herein, the symptoms of a condition contemplated herein or the potential to develop a condition contemplated herein. Such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics. As used herein, the terms “effective amount,” “pharmaceutically effective amount” and “therapeutically effective amount” refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction and/or alleviation of a sign, a symptom, or a cause of a disease or disorder, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation. As used herein, the term “pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material may be administered to an individual without causing an undesirable biological effect or interacting in a deleterious manner with any of the components of the composition in which it is contained. As used herein, the language “pharmaceutically acceptable salt” refers to a salt of the administered compound prepared from pharmaceutically acceptable non-toxic acids, including inorganic acids, organic acids, solvates, hydrates, or clathrates thereof. Examples of such inorganic acids are hydrochloric, hydrobromic, hydroiodic, sulfuric, phosphoric, acetic, hexafluorophosphoric, citric, gluconic, benzoic, propionic, butyric, sulfosalicylic, maleic, lauric, malic, fumaric, succinic, tartaric, amsonic, pamoic, p-tolunenesulfonic, and mesylic. Appropriate organic acids may be selected, for example, from aliphatic, aromatic, carboxylic and sulfonic classes of organic acids, examples of which are formic, acetic, propionic, succinic, camphorsulfonic, citric, fumaric, gluconic, isethionic, lactic, malic, mucic, tartaric, para-toluenesulfonic, glycolic, glucuronic, maleic, furoic, glutamic, benzoic, anthranilic, salicylic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, pantothenic, benzenesulfonic (besylate), stearic, sulfanilic, alginic, galacturonic, and the like. Furthermore, pharmaceutically acceptable salts include, by way of non-limiting example, alkaline earth metal salts (e.g., calcium or magnesium), alkali metal salts (e.g., sodium-dependent or potassium), and ammonium salts. As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent; diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function. Typically, such constructs are carried or transported from one organ, or portion of the body, to another organ; or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the invention, and not injurious to the patient. Some examples of materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations. As used herein, “pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound useful within the invention, and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions. The “pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound useful within the invention. Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the invention are known in the art and described, for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), Which is incorporated herein by reference. As used herein, the term “alkyl,” by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e. C1-6means one to six carbon atoms) and including straight, branched chain, or cyclic substituent groups. Examples include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, hexyl, and cyclopropylmethyl. As used herein, the term “substituted alkyl” means alkyl as defined above, substituted by one, two or three substituents selected from the group consisting of halogen, —OH, alkoxy, —NH2, amino, azido, —N(CH3)2, —C(═O)OH, trifluoromethyl, —C≡N, —C(═O)O(C1-C4)alkyl, —C(═O)NH2, —SO2NH2, —C(═NH)NH2, and —NO2. Examples of substituted alkyls include, but are not limited to, 2,2-difluoropropyl, 2-carboxycyclopentyl and 3-chloropropyl. As used herein, the term “heteroalkyl” by itself or in combination with another term means, unless otherwise stated, a stable straight or branched chain alkyl group consisting of the stated number of carbon atoms and one or two heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may be optionally oxidized and the nitrogen heteroatom may be optionally quaternized. The heteroatom(s) may be placed at any position of the heteroalkyl group, including between the rest of the heteroalkyl group and the fragment to which it is attached, as well as attached to the most distal carbon atom in the heteroalkyl group. Examples include: —O—CH2—CH2—CH3, —CH2—CH2—CH2—OH, —CH2—CH2—NH—CH3, —CH2—S—CH2—CH3, and —CH2CH2—S(═O)—CH3. Up to two heteroatoms may be consecutive, such as, for example, —CH2—NH—OCH3, or —CH2—CH2—S—S—CH3. As used herein, the term “alkoxy” employed alone or in combination with other terms means, unless otherwise stated, an alkyl group having the designated number of carbon atoms, as defined above, connected to the rest of the molecule via an oxygen atom, such as, for example, methoxy, ethoxy, 1-propoxy, 2-propoxy(isopropoxy) and the higher homologs and isomers. As used herein, the term “halo” or “halogen” alone or as part of another substituent means, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. As used herein, the term “cycloalkyl” refers to a mono cyclic or polycyclic non-aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. In one embodiment, the cycloalkyl group is saturated or partially unsaturated. In another embodiment, the cycloalkyl group is fused with an aromatic ring. Cycloalkyl groups include groups having from 3 to 10 ring atoms. Illustrative examples of cycloalkyl groups include, but are not limited to, the following moieties: Monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Dicyclic cycloalkyls include, but are not limited to, tetrahydronaphthyl, indanyl, and tetrahydropentalene, Polycyclic cycloalkyls include adamantine and norbornane. The term cycloalkyl includes “unsaturated nonaromatic carbocyclyl” or “nonaromatic unsaturated carbocyclyl” groups, both of which refer to a nonaromatic carbocycle as defined herein, which contains at least one carbon double bond or one carbon triple bond. As used herein, the term “heterocycloalkyl” or “heterocyclyl.” refers to a heteroalicyclic group containing one to four ring heteroatoms each selected from O, S and N. In one embodiment, each heterocycloalkyl group has from 4 to 10 atoms in its ring system, with the proviso that the ring of said group does not contain two adjacent O or S atoms. In another embodiment, the heterocycloalkyl group is fused with an aromatic ring. In one embodiment, the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen atom may be optionally quaternized. The heterocyclic system may be attached, unless otherwise stated, at any heteroatom or carbon atom that affords a stable structure, A heterocycle may be aromatic or non-aromatic in nature. In one embodiment, the heterocycle is a heteroaryl. An example of a 3-membered heterocycloalkyl group includes, and is not limited to, aziridine. Examples of 4-membered heterocycloalkyl groups include, and are not limited to, azetidine and a beta lactam. Examples of 5-membered heterocycloalkyl groups include, and are not limited to, pyrrolidine, oxazolidine and thiazolidinedione. Examples of 6-membered heterocycloalkyl groups include, and are not limited to, piperidine, morpholine and piperazine. Other non-limiting examples of heterocycloalkyl groups are: Examples of non-aromatic heterocycles include monocyclic groups such as aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, pyrroline, pyrazolidine, imidazoline, dioxolane, sulfolane, 2,3-dihydrofuran, 2,5-dihydrofuran, tetrahydrofuran, thiophane, piperidine, 1,2,3,6-tetrahydropyridine, 1,4-dihydropyridine, piperazine, morpholine, thiomorpholine, pyran, 2,3-dihydropyran, tetrahydropyran, 1,4-dioxane, 1,3-dioxane, homopiperazine, homopiperidine, 1,3-dioxepane, 4,7-dihydro-1,3-dioxepin, and hexamethyleneoxide. As used herein, the term “aromatic” refers to a carbocycle or heterocycle with one or more polyunsaturated rings and having aromatic character, i.e. having (4n+2) delocatized π (pi) electrons, where n is an integer. As used herein, the term “aryl,” employed alone or in combination with other terms, means, unless otherwise stated, a carbocyclic aromatic system containing one or more rings (typically one, two or three rings), wherein such rings may be attached together in a pendent manner, such as a biphenyl, or may be fused, such as naphthalene. Examples of aryl groups include phenyl, anthracyl, and naphthyl. As used herein, the term “aryl-(C1-C3)alkyl” means a functional group wherein a one- to three-carbon alkylene chain is attached to an aryl group, e.g., —CH2CH2-phenyl. In one embodiment, aryl-(C1-C3)alkyl is aryl-CH2— or aryl-(CH(CH3)—. The term “substituted aryl-(C1-C3)alkyl” means an aryl-(C1-C3)alkyl functional group in which the aryl group is substituted. Similarly, the term “heteroaryl-(C1-C3)alkyl” means a functional group wherein a one to three carbon alkylene chain is attached to a heteroaryl group, e.g., —CH2CH2-pyridyl. The term “substituted heteroaryl-(C1-C3)alkyl” means a heteroaryl-(C1-C3)alkyl functional group in which the heteroaryl group is substituted. As used herein, the term “heteroaryl” or “heteroaromatic” refers to a heterocycle having aromatic character, A polycyclic heteroaryl may include one or more rings that are partially saturated. Examples include the following moieties: Examples of heteroaryl groups also include pyridyl, pyrazinyl, pyrimidinyl (particularly 2- and 4-pyrimidinyl), pyridazinyl, thienyl, furyl, pyrrolyl (particularly pyrrolyl), imidazolyl, oxazolyl, pyrazolyl (particularly 3- and 5-pyrazolyl), isothiazoyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,3,4-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,3,4-thiadiazolyl and 1,3,4-oxadiazolyl. Examples of polycyclic heterocycles and heteroaryls include indolyl (particularly 3-, 4-, 5-, 6- and 7-indolyl), indolinyl, quinolyl, tetrahydroquinolyl, isoquinolyl (particularly 1- and 5-isoquinolyl), 1,2,3,4-tetrahydroisoquinolyl, cinnolinyl quinoxalinyl (particularly 2- and 5-quinoxalinyl), quinazolinyl, phthalazinyl, 1,4-benzodioxanyl, coumarin, dihydrocoumarin, 1,5-naphthyridinyl, benzofuryl (particularly 3-, 4-, 5-, 6- and 7-benzofuryl), 2,3-dihydrobenzofuryl, 1,2-benzisoxazolyl, benzothienyl (particularly 3-, 4-, 5-, 6-, and 7-benzothienyl), benzoxazolyl, benzothiazolyl (particularly 2-benzothiazolyl and 5-benzothiazolyl), purinyl, benzimidazolyl (particularly 2-benzimidazolyl), benzotriazolyl, thioxanthenyl, carbazolyl, carbolinyl, acridinyl, pyrrolizidinyl, and quinolizidine. As used herein, the term “substituted” means that an atom or group of atoms has replaced hydrogen as the substituent attached to another group. The term “substituted” further refers to any level of substitution, namely mono-, di-, tri-, tetra-, or Penta-substitution, where such substitution is permitted. The substituents are independently selected, and substitution may be at any chemically accessible position. In one embodiment, the substituents vary in number between one and four. In another embodiment, the substituents vary in number between one and three. In yet another embodiment, the substituents vary in number between one and two. As used herein, the term “optionally substituted” means that the referenced group may be substituted or unsubstituted. In one embodiment, the referenced group is optionally substituted with zero substituents, i.e., the referenced group is unsubstituted. In another embodiment, the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from groups described herein. In one embodiment, the substituents are independently selected from the group consisting of oxo, halogen, —CN, —NH2, —OH, —NH(CH3), —N(CH3)2, alkyl (including straight chain, branched and/or unsaturated alkyl), substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, fluoro alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted alkoxy, fluoroalkoxy, —S-alkyl, S(═O)2alkyl, —C(═O)NH[substituted or unsubstituted alkyl, or substituted or unsubstituted phenyl], —C(═O)N[H or alkyl]2, —OC(═O)N[substituted or unsubstituted alkyl]2, —NHC(═O)NH[substituted or unsubstituted alkyl, or substituted or unsubstituted phenyl], —NHC(═O)alkyl, —N[substituted or unsubstituted alkyl]C(═O)[substituted or unsubstituted alkyl], —NHC(O)[substituted or unsubstituted alkyl], —C(OH)[substituted or unsubstituted alkyl]2, and —C(NH2)[substituted or unsubstituted alkyl]2. In another embodiment, by way of example, an optional substituent is selected from oxo, fluorine, chlorine, bromine, iodine, —CN, —NH2, —OH, —NH(CH3), —N(CH3)2, —CH3, —CH2CH3, —CH(CH3)2, —CH2CF3, —OCH3, —OCH2CH3, —OCH(CH3)2, —OCF3, —OCH2CF3, —S(═O)2—CH3, —C(═O)NH2, —C(═O)—NHCH3, —NHC(═O)NHCH3, —C(═O)CH3, —ON(O)2, and —C(═O)OH. In yet one embodiment, the substituents are independently selected from the group consisting of C1-6alkyl, —OH, C1-6alkoxy, halo, amino, acetamido, oxo and nitro. In yet another embodiment, the substituents are independently selected from the group consisting of C1-6alkyl, C1-6alkoxy, halo, acetamido, and nitro. As used herein, where a substituent is an alkyl or alkoxy group, the carbon chain may be branched, straight or cyclic. Ranges: throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range. Description In one embodiment, the invention provides organic molecules comprising at least one phosphate moiety, and methods of use for the synthesis of phosphorylated molecules. The phosphorylated molecule generated using the compositions and methods of the invention may be a phosphorylated protein, a phosphorylated sugar, a phosphorylated lipid, or a phosphorylated nucleoside molecule. Phosphorylated nucleosides that can be synthesized using the methods of the invention include, but are not limited to, natural or modified nucleoside hexaphosphates (e.g., 5′ nucleoside hexaphosphate (Np6) and 2′-deoxynucleoside-5′-hexaphosphates (dNp6)), nucleoside hexaphosphate analogs, natural or modified nucleoside pentaphosphates (e.g., pppGpp, dinucleoside pentaphosphate (e.g., diadenosine pentaphosphate), 5′ nucleoside pentaphosphate (Np5) and 2′-deoxynucleoside-5′-pentaphosphates (dNp5)), nucleoside pentaphosphate analogs, natural or modified nucleoside tetraphosphates (e.g., ppGpp, dinucleoside tetraphosphate (e.g., diadenosine tetraphosphate), 5′ nucleoside tetraphosphate (Np4) and 2′-deoxynucleoside-5′-tetraphosphates (dNp4)), nucleoside tetraphosphate analogs, natural or modified nucleoside triphosphates 5′ nucleoside triphosphate (NTPs), 5′ nucleoside triphosphate (NTPs) and 2′-deoxynucleoside-5′-triphosphates (dNTPs)), nucleoside triphosphate analogs, natural or modified nucleoside diphosphates (e.g., 5′ nucleoside triphosphate. (NDPs) and 2′-deoxynucleoside-5′-diphosphates (dNDPs)), nucleoside diphosphate analogs, natural or modified nucleoside monophosphates (e.g., 5′ nucleoside monophosphate (NMPs) and 2′-deoxynucleoside-5′-monophosphates (dNMPs)), and nucleoside monophosphate analogs. Compounds of the Invention The compounds of the present invention may be synthesized using techniques well-known in the art of organic synthesis. The starting materials and intermediates required for the synthesis may be obtained from commercial sources or synthesized according to methods known to those skilled in the art. In one embodiment, the invention relates to a compound represented by formula (1): wherein, m is an integer from 0 to 5;n is an integer from 0 to 5;X is O or C1′12;R11is an aryl, a heteroaryl, an alkyl, a cycloakyl, an alkenyl, an alkynyl, an alkyl-aryl-alkyl, or a silyl group;R12is hydrogen, null, a substituted tetahydrofuranyl group, or an alkyl-substituted tetahydrofuranyl group; andeach occurrence of R13is independently hydrogen or null. In one embodiment, R11is substituted with a group selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, halogen, —CN, —OR14, and —N(R14)2, wherein each occurrence of R14is independently selected from the group consisting hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and halogen. In one embodiment, R11is selected from the group consisting of In one embodiment, R12is hydrogen. In one embodiment, R12is null. In one embodiment, when R12is null there is an anion. In one embodiment, R12is a methyl-tetrahydrofuranyl group wherein the tetrahydrofuranyl group is substituted. In one embodiment, R12is tetrahydrofuranyl group wherein the tetrahydrofuranyl group is substituted. In one embodiment, the tetrahydrofuranyl group is substituted with a oxy-phenyl. In one embodiment, the tetrahydrofuranyl group is substituted with a nitrogenous base. In one embodiment, each occurrence of R13is hydrogen. In one embodiment occurrence of R13is null. In one embodiment, when R13is null there is an anion. In one embodiment, m is 0. In one embodiment m is 1. In one embodiment, m is 2. In one embodiment, m is 3. In one embodiment, n is 0. In one embodiment, n is 1. In one embodiment, n is 2. In one embodiment, n is 3. In one embodiment, n is 4. In one embodiment, n is 5. In one embodiment X is O. In one embodiment, X is CH2. In one embodiment, the compound of formula (1) is a compound of formula (1a): wherein R11is selected from the group consisting of an aryl, a heteroaryl, an alkyl, a cycloalkyl, an alkenyl, an alkynyl, an alkyl-aryl, and an aryl-alkyl, wherein R1ais optionally substituted. In one embodiment, R1ais substituted with a group selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, cycloalkyl; heteroaryl, halogen, —CN, and —N(R11a)2, wherein each occurrence of Riva is independently selected from the group consisting hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and halogen. In one embodiment, R1ais selected from the group consisting of In one embodiment, the compound of formula (1a) is selected from the group consisting of In one embodiment, the compound of formula (1) is a compound of formula (1b): wherein R1bis a hydroxy protecting group. In one embodiment, R1bis selected from the group consisting of an aryl, a heteroaryl, an alkyl, a cycloalkyl, an alkenyl, an alkynyl, an alkyl-aryl, and an aryl-alkyl, wherein R1bis optionally substituted. In one embodiment, R1bis substituted with a group selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, halogen, —CN, —OR11b, and —N(R11b)2, wherein each occurrence of Rub is independently selected from the group consisting hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and halogen. In one embodiment, R1bis selected from the group consisting of In one embodiment, the compound of formula (1b) is selected from the group consisting of In one embodiment, the compound of formula (1) is a compound of formula (1c): wherein:m is an integer from 0 to 5;n is an integer from 0 to 5;X is O or CH2;R11cis an aryl, a heteroaryl, an alkyl, a cycloakyl, an alkenyl, an alkynyl, an alkyl-aryl, an aryl-alkyl, or a silyl group;each occurrence of R13cis independently hydrogen or null;R15cis hydrogen, aryl, or heteroaryl; andR16cis a nitrogenous base, wherein the nitrogenous base is a natural nitrogenous base or artificial nitrogenous base. In one embodiment, R11cis In one embodiment, R15cis phenyl. In one embodiment the compound of formula (1c) is: In one embodiment, the compound of formula (1) is a compound of formula (1d): wherein:m is an integer from 0 to 5;n is an integer from 0 to 5;X is O or CH2;R11dis an aryl, a heteroaryl, an alkyl, a cycloalkyl, an alkenyl, an alkynyl, an alkyl-aryl, an aryl-alkyl, or a silyl group;each occurrence of R13dis independently hydrogen or null;R15dis hydrogen, aryl, or heteroaryl; andR16dis a nitrogenous base, wherein the nitrogenous base is a natural nitrogenous base or artificial nitrogenous base. In one embodiment, R11dis In one embodiment, R15dis phenyl. In one embodiment, the compound of formula (1d) is: In one embodiment, the compound of formula (1) is a compound of formula (13): wherein:m is an integer from 0 to 5;n is an integer from 0 to 5;X is O or CH2;R11eis an aryl, a heteroaryl, an alkyl, a cycloalkyl, an alkenyl, an alkynyl, an alkyl-aryl, an aryl-alkyl, or a silyl group;each occurrence of R13eis independently hydrogen or null;R15eis hydrogen, aryl, acetyl, or heteroaryl; andR16eis a nitrogenous base, wherein the nitrogenous base is a natural nitrogenous base or artificial nitrogenous base. In one embodiment, R11eis In one embodiment, R15eis phenyl. In one embodiment, the compound of formula (1e) is: In one embodiment, the invention relates to a compound represented by formula (2): wherein n is an integer from 1 to 10, andR2is a support. In one embodiment, the invention relates to a compound represented by formula (3): wherein p is an integer from 0 to 10, andR3is a support. In one embodiment, the invention relates to a compound represented by formula (4): wherein q is an integer from 0 to 10, andr is an integer from 0 to 10. In one embodiment q is 1. In one embodiment r is 1. In one embodiment, the compound of formula (4) is Supports The attachment of useful materials such as catalysts, reagents, chelating or complexing agents, and proteins to insoluble supports is well-known. With the attending advantages of ease of removal and recovery from the system, e.g., by simple filtration, regeneration (if necessary), and recycling coupled with the increased utilization of continuous flow systems in both general chemical processing and diagnostic monitoring procedures, supported materials and methods of generating polymer supported reaction reagents are well known in the art. In one embodiment, the invention relates to a compound represented by formula (2) or formula (3) attached to an inorganic polymer support. Inorganic polymer supports include, but are not limited to, silica gel and alumina. In one embodiment, the invention relates to a compound represented by formula (2) or formula (3) attached to an organic polymer support. Organic polymer supports include, but are not limited to, polystyrene. In one embodiment, the invention relates to a compound represented by formula (2) or formula (3) attached to solid support (e.g., controlled pore glass). Nucleoside Triphosphates and Nucleic Acids In one embodiment, the compounds and methods of the invention are used to synthesize a naturally occurring nucleoside triphosphate. Naturally occurring nucleoside triphosphates include, but are not limited to, adenosine triphosphate (ATP), guanosine triphosphate (GTP), cytidine triphosphate (CTP), 5-methyluridine triphosphate (m5UTP), and uridine triphosphate (UTP), The terms ATP, GTP, CTP, and UTP refer to those nucleoside triphosphates that contain ribose. The nucleoside triphosphates containing deoxyribose are called dNTPs, and include deoxyadenosine triphosphate (dATP), deoxyguanosine triphosphate (dGTP), deoxycytidine triphosphate (dCTP), deoxythymidine triphosphate (dTTP) and deoxyuridine triphosphate (dUTP). In one embodiment, the compounds and methods of the invention are used to synthesize isomers or analogs of nucleoside triphosphates. Exemplary nucleoside triphosphate analogs or isomers include, but are not limited to, 2′-deoxythymidine-3′-triphosphate (3′-TTP), 1-(α-L-threofuranosyl)thymidine-3′-triphosphate (tTTP), 1-(α-L-threofuranosyl)cytidine-3′-triphosphate (tCTP), 9-(1-L-threofuranosyl)adenosine-3′-triphosphate (tATP), 9-(α-L-threofuranosyl)guanosine-3′-triphosphate (tGTP), and L-2′-deoxythymidine-5′-triphosphate (L-dTTP). In some embodiments, the nucleoside triphosphate can be labeled. Examples of possible labels include, but are not limited to a radioisotope, an enzyme, an enzyme cofactor, an enzyme substrate, an enzyme inhibitor, a dye, a hapten, chemiluminescent molecule, a fluorescent molecule, a phosphorescent molecule, an electrochemiluminescent molecule, a chromophore, a magnetic particle, an affinity label, a chromogenic agent, an azide group or other groups used for click chemistry, and other moieties known in the art. In one aspect, the nucleoside triphosphates synthesized according to the methods of the invention can be used for synthesizing nucleic acid molecules. Nucleic acid in the context of the present invention includes but is not limited to deoxyribonucleic acid (DNA), ribonucleic acid (RNA) and peptide nucleic acid (PNA). A nucleic acid of the invention also includes artificial genetic polymers, commonly referred to as XNAs or ‘xeno-nucleic acids’ where the backbone structure contains a sugar other than ribose or deoxyribose. While some of these molecules can be considered natural derivatives of RNA, like arabino nucleic acid (ANA), threose nucleic acid (TNA), and glycerol nucleic acid (GNA), others are completely unnatural, like locked nucleic acid (INA), cyclohexene nucleic acid (CeNA), and hexitol nucleic acid (HNA). Therefore in one embodiment, the invention provides artificial or synthetic nucleic acid molecules which incorporate one or more natural or modified nucleoside triphosphate of the invention. The length of the nucleic acids may vary. The nucleic acids may be modified, e.g. may comprise one or more modified nucleobases or modified sugar moieties (e.g., comprising methoxy groups). The backbone of the nucleic acid may comprise one or more peptide bonds as in peptide nucleic acid (PNA). The nucleic acid may comprise a base analog such as non-purine or non-pyrimidine analog or nucleotide analog. It may also comprise additional attachments such as proteins, peptides and/or or amino acids. Activated Nucleoside Monophosphates In one embodiment, the invention relates to the synthesis of activated nucleoside monophosphates for use in synthesizing a nucleoside triphosphate of the invention. An activated nucleoside monophosphates for use in the methods of the invention can be synthesized from any nucleoside monophosphate. In various embodiments, a nucleoside monophosphate is a naturally occurring nucleoside monophosphate, an unnatural nucleoside monophosphate or a modified nucleoside monophosphate. In one embodiment, the activated nucleoside monophosphate comprises 2-methylimidazole linked to a phosphate group. In one embodiment, the phosphate group is linked to a sugar moiety which is attached to a nitrogenous base. In one embodiment, the sugar of the activated nucleoside monophosphate comprises ribose. In one embodiment, the sugar of the activated nucleoside monophosphate comprises deoxyribose. In one embodiment, the activated nucleoside monophosphate is represented by formula (5) wherein R5is selected from the group of hydrogen, aryl, and heteroaryl; and R6is a nitrogenous base. In one embodiment, the nitrogenous base is a natural nitrogenous base or artificial nitrogenous base. In one embodiment, R5is phenyl. A variety of purities, purine analogs, pyrimidines, pyrimidine analogs, and other heterocycles as nitrogenous bases have been well documented (Chemistry of Nucleosides and Nucleotides Vol 1, 2, 3, edited by Townsend, Plenum Press, 1988, 1991, 1994). The condensations of sugars with nitrogenous bases to yield nucleosides are frequently used reactions in nucleoside chemistry. Well-established procedures and methodologies can be found in the literature (Vorbruggen et al., Chem. Ber. 1981, 114, 1234-1268, 1279-1286; Wilson et al., Synthesis, 1995, 1465-1479). A large number of known nucleosides are prepared from the modifications of purine and pyrimidine nucleosides. The modifications can be done on the sugars and/or nucleoside bases. A simple, widely-used reaction is the nucleophilic substitution of halopurine or halopyrimidine base by a variety of nucleophiles such as hydroxide, ammonia, hydrogen sulfide, alkoxides, amities, alkylthiol, hydrazine, hydroxyamines, azide, cyanide, and hydride. This type of reactions can be very useful for preparation of 2-substituted purine nucleoside, 6-substituted purine nucleosides, 8-substituted purine nucleosides, 2,6-disubstituted purine nucleosides, 2,8-disubstituted purine nucleosides, 6,8-disubstituted purine nucleosides, 2,6,8-trisubstituted purine nucleosides (Halbfinger et al., J. Med. Chem. 1999, 42, 5323-5337, Lin et al., J. Med. Chem. 1985, 28, 1481-1485; Bressi et al., J. Med. Chem. 2000, 43, 4135-4150). These substitution reactions are readily extended to purine nucleoside analogs such as 7-deazapurine nucleosides, 7-deaza-8-azapurine nucleosides, 8-azapurine nucleosides, 3-deazapurine nucleosides, 3-deaza-8-azapurine nucleosides, and 3,8-dideazapurine nucleosides. For instance, a number of 7-deaza-7-substituted purine nucleoside have been prepared through such substitutions (Ugarkar et al., J. Med. Chem. 2000, 43, 2894-2905), The same methodologies can be used for the preparation of 4-substituted pyrimidine nucleosides, 5-substituted pyrimidine nucleosides, 4,5-disubstituted pyrimidine nucleosides, 5-substituted 6-azapyrimidine nucleosides, 5-substituted 6-azapyrimidine nucleosides, and 4,5-disubstituted 6-azapyrimidine nucleosides. The sugar moieties of synthesized nucleosides can be further modified. There are a variety of reactions which can be used to modify the sugar moiety of nucleosides. The reactions frequently used include deoxygenation, oxidation/addition, substitution, and halogenation. The deoxygenations are useful for the preparation of 2′-deoxy-, 3′-deoxy-, and 2′,3′-dideoxy-nucleosides. A widely-used reagent is phenyl chlorothionoformate, which reacts with the hydroxy of nucleosides to yield a thionocarbonate. The treatment of the thionocarbonate with tributyltin hydride and AIBN yields deoxygenated nucleosides. The oxidation/addition includes the conversion of a hydroxy group to a carbonyl group, followed by a nucleophilic addition, resulting in C-alkylated nucleosides and C-substituted nucleosides. The substitution may be just a simple displacement of a hydroxyl proton by alkyl, or may be a conversion of a hydroxyl to a leaving group, followed by a nucleophilic substitution. The leaving group is usually a halogen, mesylate, tosylate, nisylate, or a triflate. A variety of nucleophiles can be used, resulting in 2-, or 3-substituted nucleosides. Halogenation can be used to prepare 2′-halo 3′-halo-, or 4′-halonucleosides, Chlorination and fluorination are commonly used and result in important fluoro-sugar and chloro-sugar nucleosides. Nucleosides that can be included in an activated nucleoside monophosphate according to the methods of the invention include, but are not limited to, adenosine, cytidine, guanosine, uridine, 2′-deoxyadenosine, 2′-deoxycytidine, 2′-deoxyguanosine, thymidine, inosine, 9-(β-D-arabinofuranosyl)adenine, 10-D-arabinofuranosyl)cytosine, 9-(β-D-arabinofuranosyl)guanine, 1-(β-D-arabinofuranosyl)uracil, 9-(β-D-arabinofuranosyl)hypoxanthine, 1-(β-D-arabinofuranosyl)thymine, 3′-azido-3′-deoxythymidine, 3′-azido-2′, 3′-dideoxyuridine, 3′-azido-2′, 3′-dideoxycytidine, 3′-azido-2′, 3′-dideoxyadenosine, 3′-azido-2′, 3′-dideoxyguanosine, 3′-azido-2′, 3′-dideoxyinosine, 3′-deoxythymidine, 2′, 3′-dideoxyuridine, 2′, 3′-dideoxyinosine, 2′, 3′-dideoxyadenosine, 2′, 3′-dideoxycytidine, 2′, 3′-dideoxyguanosine, 9-(2, 3-dideoxy-1-β-D-ribofuranosyl)-2, 6-diaminopurine, 3′-deoxy-2′, 3′-didehydrothymidine, 2′, 3′-didehydro-2′, 3′-dideoxyuridine, 2′, 3′-didehydro-2′, 3′-dideoxycytidine, 2′, 3′-didehydro-2′, 3′-dideoxyadenosine, 2′, 3′-didehydro-2′, 3′-dideoxyguanosine, 2′, 3′-didehydro-2′, 3′-dideoxyinosine, 3-deazaadenosine, 3-deazaguanosine, 3-deazainosine, 7-deazaadenosine, 7-deazaguanosine, 7-deazainosine, 6-azauridine, 6-azathymidine, 6-azacytidine, 5-azacytidine, 9-(β-D-ribofuranosyl)-6-thiopurine, 6-methylthio-9-(β-D-ribofuranosyl)purine, 2-amino-9-(β-D-ribofuranosyl)-6-thiopurine, 2-amino-6-methylthio-9-(β-D-ribofuranosyl)purine, 5-fluorocytidine, 5-iodocytidine, 5-bromocytidine, 5-chlorocytidine, 5-fluorouridine, 5-iodouridine, 5-bromouridine, 5-chlorouridine, 2′-C-methyladenosine, 2′-C-methylcytidine, 2′-C-methylguanosine, 2′-C-methylinosine, 2′-C-methyluridine, 2′-C-methylthymidine, 2′-deoxy-2′-fluoroadenosine, 2′-deoxy-2′-fluorocytidine, 2′-deoxy-2′-fluoroguanosine, 2′-deoxy-2′-fluorouridine, 2′-deoxy-2′-fluoroinosine, 2′-α-fluorothymidine, 2′-deoxy-2′-fluoroarabinoadenosine, 2′-deoxy-2′-fluoroarabinocytidine, 2′-deoxy-2′-fluoroarabinoguanosine, 2′-deoxy-2′-fluoroarabinouridine, 2′-deoxy-2′-fluoroarabinoinosine, 2′-β-fluorothymidine, 2′-O-methyladenosine, 2′-O-methylcytidine, 2′-O-methylguanosine, 2′-O-methylinosine, 2′-O-5-dimethyluridine, 2′-C-ethynylcytidine, 2′-C-ethynylguanosine, 2′-C-ethynyluridine, 2′-C-ethynylinosine, 2′-C-ethynyl-5-methyluridine, 3′-C-ethynyladenosine, 3′-C-ethynylcytidine, 3′-C-ethynylguanosine, 3′-C-ethynyluridine, 3′-C-ethynylinosine, 3′-C-ethynyl-5-methyluridine, 3′-deoxyadenosine, 3′-deoxycytidine, 3′-deoxyguanosine, 3′-deoxyuridine, 3′-deoxyinosine, 4′-C-ethynyladenosine, 4′-C-ethynylpyridine, 4′-C-ethynylguanosine, 4′-C-ethynyluridine, 4′-C-ethynylinosine, 4′-C-ethynylthymidine, 4′-C-methyladenosine, 4′-C-methylcytidine, 4′-C-methylguanosine, 4′-C-methyluridine, 4′-C-methylthymidine, 2′-C-methyl-7-deazaadenosine, 2′-C-methyl-7-deazaguanosine, 2′-C-methyl-3-deazaadenosine, 2′-C-methyl-3-deazaguanosine, deazaadenosine, 2′-O-methyl-7-deazaguanosine, 2′-O-methyl-3-deazaadenosine, 2′-O-methyl-3-deazaguanosine, 2′-C-methyl-6-azauridine, 2′-C-methyl-5-fluorouridine, 2′-C-methyl-5-fluorocytidine, 2′-C-methyl-2-chloroadenosine, 2′-deoxy-7-deazaadenosine, 2′-deoxy-3-deazaadenosine, 2′-deoxy-7-deazaguanosine, 2′-deoxy-3-deazaguanosine, 2′-deoxy-6-azauridine, 2′-deoxy-5-fluorouridine, 2′-deoxy-5-fluorocytidine, 2′-deoxy-5-iodouridine, 2′-deoxy-5-iodocytidine, 2′-deoxy-2-chloroadenosine, 2′-deoxy-2-fluoroadenosine, 3′-deoxy-7-deazaadenosine, 3′-deoxy-7-deazaguanosine, 3′-deoxy-3-deazaadenosine, 3′-deoxy-3-deazaguanosine, 3′-deoxy-6-azauridine, 3′-deoxy-5-fluorouridine, 3′-deoxy-5-iodouridine, 3′-deoxy-5-fluorocytidine, 3′-deoxy-2-chloroadenosine, 2′, 3′-dideoxy-7-deazaadenosine, 2′, 3′-dideoxy-7-deazaguanosine, 2′, 3′-dideoxy-3-deazaadenosine, 2′, 3′-dideoxy-3-deazaguanosine, 2′, 3′-dideoxy-6-azauridine, 2′, 3′-dideoxy-5-fluorouridine, 2′, 3′-dideoxy-5-fluorouridine, 2′, 3′-dideoxy-5-iodocytidine, 2′, 3′-dideoxy-2-chloroadenosine, 3′-dideoxy-β-L-cytidine, 2′, 3′-dideoxy-β-L-adenosine, 2′, 3′-dideoxy-β-L-guanosine, 3′-deoxy-L-thymidine, 2′, 3′-dideoxy-5-fluoro-β-L-cytidine, β-L-thymidine, 2′-deoxy-β-L-cytidine, 2′-deoxy-β-L adenosine, 2′-deoxy-β-L-guanosine, 2′-deoxy-β-L-inosine, 13-L-cytidine, β-L-adenosine, β-L-guano sine, β-L-uridine, β-L-inosine, 2′, 3′-didehydro-2′, 3′-dideoxy-β-L-cytidine, 2′, 3′-didehydro-3′-dideoxy-β-L-thymidine, 2′, 3′-didehydro-2′, 3′-dideoxy-β-L-adenosine, 2′, 3′-didehydro-2′, 3′-dideoxy-β-L-guanosine, 2′, 3′-didehydro-2′, 3′-dideoxy-β-L-5-fluorocytidine, 2′-deoxy-2′, 2′-di fluorocytidine, 9-(β-D-arabinofuranosyl)-2-fluoroadenine, 2′-deoxy-2′(E)-fluoromethylenecytidine, 2′-deoxy-2′ (Z)-fluoromethylenecytidine, (−)-2′, 3′-dideoxy-3′-thiacytidine, (+)-2′, 3′-dideoxy-3′-thiacytidine, 1-β-D-ribofuranosy-1, 2, 4-triazole-3-carboxamide, 1-β-L-ribofuranosyl-1, 2, 4-triazole-3-carboxamide, 1-β-D-ribofuranosyl-1, 3-imidazolium-5-olate, 1-β-L-ribofuranosyl-1, 1-β-D-ribofuranosyl-5-ethynylimidazole-4-carboxamide, 1-β-L-ribofuranosyl-5-ethynylimidazole-4-carboxamide, 1-(2-deoxy-2-fluoro-3-D-arabinofuranosyl)-5-iodouracil, 1-(2-deoxy-2-fluoro-β-D-arabinofuranosyl)-5-iodocytosine, 1-(2-deoxy-2-fluoro-β-L-arabinofuranosyl)-5-methyluracil, arabinofuranosyl)-5-(2-bromovinyl)uracil, E-5-(2-bromovinyl)-2′-deoxyuridine, 5-trifluoromethylthymidine, 1-β-D-arabinofuranosyl-5-propynyluracil, 1-(2-deoxy-2-fluoro-β-D-arabinofuranosyl)-5-ethyluracil, 2′, 3′-dideoxy-3′-fluoroguanosine, 3′-deoxy-3′-fluorothymidine, (±)-(1α, 2β, 3α)-9-[2, 3-bis(hydroxymethyl)-1-cyclobutyl]adenine, (±)-(1α, 2β, 3α)-9-[2, 3-bis(hydroxymethyl)-1-cyclobutyl]guanine, (±)-(1β, 2α, 3β)-9-[2, 3-bis(hydroxymethyl)-1-cyclobutyl]guanine, (±)-(1β, 2α, 3β)-9-[2, 3-bis(hydroxymethyl)-1-cyclobutyl]adenine, (1R, 3S, 4R)-9-(3-hydroxy-4-hydroxymethylcyclopent-1-yl)guanine, (1S, 2R, 4R)-9-(1-hydroxy-2-hydroxymethylcyclopent-4-yl)guanine, (2R, 4R)-9-(2-hydroxymethyl-1, 3-dioxolan-4-yl)-2, 6-diaminopurine, (2R, 4R)-1-(2-hydroxymethyl-1, 3-dioxolan-4-yl)cytosine, (2R, 4R)-9-(2-hydroxymethyl-1, 3-dioxolan-4-yl)guanine, (2R, 4R)-1-(2-hydroxymethyl-1, 3-dioxolan-4-yl)-5-fluorocytosine, (1R, 2S, 4S)-9-(4-hydroxy-3-hydroxymethyl-2-methylenecyclopent-4-yl]guanine, and (1S, 3R, 4S)-9-(3-hydroxy-4-hydroxymethyl-5-methylenecyclopent-1-yl]guanine, Methods In one aspect, the invention provides methods of synthesizing phosphorylated molecules. In one aspect, the method comprises forming a mixture comprising an activated precursor molecule and an organic molecule comprising at least one phosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a phosphorylated molecule. In various embodiments, the compositions and methods of the invention can be used to synthesize molecules with mono-, tri-, tetra-, penta- or hexa-peptide moieties. The methods disclosed herein can be used to synthesize phosphorylated sugar molecules, phosphorylated proteins or peptides, phosphorylated lipids, or phosphorylated nucleic acid molecules. In one aspect, the invention provides methods of synthesizing natural or modified nucleoside monophosphates including, but not limited to, 5′ nucleoside monophosphate (NMP), 2′-deoxynucleoside-5′-monophosphate (dNMP), and analogs thereof. In one aspect, the method comprises forming a mixture comprising an activated nucleoside molecule and an organic molecule comprising a monophosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside monophosphate. In one aspect, the invention provides methods of synthesizing natural or modified nucleoside diphosphates including, but not limited to, 5′ nucleoside diphosphate (NDP) and 2′-deoxynucleoside-5′-diphosphates (dNDP), and analogs thereof. In one aspect, the method comprises forming a mixture comprising an activated nucleoside monophosphate and an organic molecule comprising a monophosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside diphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside molecule and an organic molecule comprising a pyrophosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside diphosphate. In one aspect, the invention provides methods of synthesizing natural or modified nucleoside triphosphates including, but not limited to, 5′ nucleoside triphosphate (NTP) and 2′-deoxynucleoside-5′-triphosphates (dNTP), and analogs thereof. In one aspect, the method comprises forming a mixture comprising an activated nucleoside monophosphate and an organic molecule comprising a pyrophosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside triphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside molecule and an organic molecule comprising a triphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside triphosphate. In one aspect, the invention provides methods of synthesizing natural or modified nucleoside tetraphosphates including, but not limited to, 5′ nucleoside tetraphosphate (Np4) and 2′-deoxynucleoside-5′-tetraphosphates (dNp4), and analogs thereof. In one aspect, the method comprises forming a mixture comprising an activated nucleoside monophosphate and an organic molecule comprising a triphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside tetraphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside molecule and an organic molecule comprising a tetraphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside tetraphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside diphosphate molecule and an organic molecule comprising a pyrophosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside tetraphosphate. In one aspect, the invention provides methods of synthesizing natural or modified nucleoside pentaphosphates including, but not limited to, 5′ nucleoside pentaphosphate (Np5) and 2′-deoxynucleoside-5′-pentaphosphates (dNp5), and analogs thereof. In one aspect, the method comprises forming a mixture comprising an activated nucleoside monophosphate and an organic molecule comprising a pentaphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside pentaphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside molecule and an organic molecule comprising a pentaphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside pentaphosphate, in one aspect, the method comprises forming a mixture comprising an activated nucleoside triphosphate molecule and an organic molecule comprising a pyrophosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside pentaphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside diphosphate molecule and an organic molecule comprising a triphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside pentaphosphate. In one aspect, the invention provides methods of synthesizing natural or modified nucleoside hexaphosphates including, but not limited to, 5′ nucleoside hexaphosphate (Np6) and 2′-deoxynucleoside-5′-hexaphosphates (dNp6), and analogs thereof. In one aspect, the method comprises forming a mixture comprising an activated nucleoside monophosphate and an organic molecule comprising a pentaphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside hexaphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside pentaphosphate and an organic molecule comprising a monophosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside hexaphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside tetraphosphate molecule and an organic molecule comprising a pyrophosphate moiety f the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside hexaphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside diphosphate molecule and an organic molecule comprising a tetraphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside hexaphosphate. In one aspect, the method comprises forming a mixture comprising an activated nucleoside triphosphate molecule and an organic molecule comprising a triphosphate moiety of the invention and incubating the mixture in the presence of a catalyst to catalyze synthesis of a nucleoside hexaphosphate. Similarly, the compositions and methods of the invention can be used to synthesize molecules with heptapeptides, octapeptides, nonapeptides, decapeptides or longer peptide moieties. Although it is not intended that the method of the invention be limited to a particular set of reaction conditions, in one embodiment, the reaction conditions include a first step of contacting an activated nucleoside monophosphate with an organic molecule comprising a phosphate moiety of the invention in the presence of an acid catalyst at room temperature (rt) to form a reaction intermediate, followed by second step of contacting the reaction intermediate with a base at a temperature of 37° C. Although it is not intended that the method of the invention be limited to any particular acid catalyst, in one embodiment, any acidic compound (including the so-called Lewis acids) may be used as catalysts. In one embodiment, the acid catalyst is ZnCl2, H2SO4, HCl, H3PO4or BF3. In another embodiment, the acid catalyst is a sulfonic acid or its salt, comprising ortho-toluenesulfonic acid, meta-toluenesulfonic acid, alkylbenzenesulfonic acid, secondary alkyl-sulfonic acid, sulfonic resin, alkylsulfate, alkylbenzenesulfonate, alkyl-sulfonate and sulfosuccinic acid. In a preferred embodiment, the acid catalyst is ZnCl2. Although it is not intended that the method of the invention be limited to a particular base, in one embodiment, the base is selected from a group of organic or inorganic basic materials comprising the alkali metal bases such as alkali metal hydroxide, carbonates, and bicarbonates. In another embodiment, the base is selected from a group comprising the alkaline earth bases such as calcium oxide and magnesium oxide. In another embodiment, aluminum bases such as aluminum hydroxide or its basic alkali aluminum components are contemplated. In a further embodiment, the base is selected from a group comprising ammonia-based compounds, such as ammonium hydroxide, and amines including, but not limited to, primary, secondary tertiary and heterocyclic amines, Applications The compositions comprising modified nucleotides and methods catalyzing the incorporation of modified nucleotides of the present invention may be used in a wide variety of protocols and technologies. For example, in certain embodiments, the modified nucleotides are used in the fields of molecular biology, genomics, transcriptomics, epigenetics, nucleic acid sequencing, and the like. That is, modified nucleotides may be used in any technology that may require or benefit from specific incorporation of modified nucleotides, Exemplary technologies, include, but are not limited to cDNA library construction; DNA epigenome and RNA methylome assays, high-throughput next generation sequencing technologies including but not limited to Illumina, soLiD, and Ion Torrent sequencing; and single nucleic acid molecule real time sequencing (SMRT) including, but not limited to, technologies from Pacific Bioscience and Oxford Nanopore Technologies such as zero-mode waveguide or nanopore sequencing, respectively. Pharmaceutical Compositions In some embodiments, nucleoside triphosphates can function as pharmaceutical agents, e.g. 3′-azido-3′-deoxythymidine (AZT, an anti-HIV drug) triphosphate and arabinosylcytosine (Cytarabine, an anticancer drug) triphosphate. In addition, nucleotides have been considered as antimetabolite drugs, agents for the treatment of diseases and disorders associated with infection (e.g., U.S. Pat. No. 5,763,447). Nucleoside triphosphates or nucleoside triphosphate analogs have also been described for the treatment of diseases and disorders including, but not limited to, sinusitis (e.g., U.S. Pat. No. 5,789,391), ostitis media (e.g., U.S. Pat. No. 6,423,694), inflammatory conditions (e.g., U.S. Patent Application No. 2005/261239), and cancer (e.g., U.S. Pat. No. 5,049,372). Therefore, in one embodiment, the invention also relates to a pharmaceutical composition comprising a therapeutically effective amount of a synthesized nucleotide of the invention, a pharmaceutically acceptable salt thereof, optionally in combination with one or more other active ingredients and/or with a pharmaceutically acceptable carrier. Moreover, the above any of the compounds may be used in a method for the treatment of a disease or disorder including, but not limited to, a microbial infection or proliferative disorder, comprising administering a therapeutically effective amount of any of the above compounds to a subject in need thereof. The pharmaceutical composition of the present invention comprises at least one nucleotide triphosphate synthesized according to the methods of the invention, or pharmaceutically acceptable salts, esters or prodrugs thereof as active ingredients. The compositions include those suitable for oral, topical, intravenous, subcutaneous, nasal, ocular, pulmonary, and rectal administration. The compounds of the invention can be administered to mammalian individuals, including humans, as therapeutic agents. For example, the compounds of the invention are useful as antiviral agents. The present invention provides a method for the treatment of a patient afflicted with a viral infection comprising administering to the patient a therapeutically effective antiviral amount of a compound of the invention. The term “viral infection” as used herein refers to an abnormal state or condition characterized by viral transformation of cells, viral replication and proliferation. Viral infections for which treatment with a compound of the invention will be particularly useful include the viruses mentioned above. A “therapeutically effective amount” of a compound of the invention refers to an amount which is effective, upon single or multiple dose administration to the patient, in controlling the growth of e.g., the microbe or tumor or in prolonging the survivability of the patient beyond that expected in the absence of such treatment. As used herein, “controlling the growth” refers to slowing, interrupting, arresting or stopping the microbial or proliferative transformation of cells or the replication and proliferation of the microbe and does not necessarily indicate a total elimination of e.g., the microbe or tumor. Accordingly, the present invention includes pharmaceutical compositions comprising, as an active ingredient, at least one of the compounds of the invention in association with a pharmaceutical carrier. The compounds of this invention can be administered by oral, parenteral (intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection), topical, transdermal (either passively or using iontophoresis or electroporation), transmucosal (e.g., nasal, vaginal, rectal, or sublingual) or pulmonary (e.g., via dry powder inhalation) routes of administration or using bioerodible inserts and can be formulated in dosage forms appropriate for each route of administration. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert pharmaceutically acceptable carrier such as sucrose, lactose, or starch. Such dosage forms can also comprise, as is normal practice, additional substances other than inert diluents, e.g., lubricating, agents such as magnesium stearate. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, with the elixirs containing inert diluents commonly used in the art, such as water. Besides such inert diluents, compositions can also include adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents. Preparations according to this invention for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, or emulsions. Examples of non-aqueous solvents or vehicles are propylene glycol polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate. Such dosage forms may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They may be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured using sterile water, or some other sterile injectable medium, immediately before use. Compositions for rectal or vaginal administration are preferably suppositories Which may contain, in addition to the active substance, excipients such as cocoa butter or a suppository wax. Compositions for nasal or sublingual administration are also prepared with standard excipients well known in the art. Topical formulations will generally comprise ointments, creams, lotions, gels or solutions. Ointments will contain a conventional ointment base selected from the four recognized classes: oleaginous bases; emulsifiable bases; emulsion bases; and water-soluble bases. Lotions are preparations to be applied to the skin or mucosal surface without friction, and are typically liquid or semiliquid preparations in which solid particles, including the active agent, are present in a water or alcohol base. Lotions are usually suspensions of solids, and preferably, for the present purpose, comprise a liquid oily emulsion of the oil-in-water type. Creams, as known in the art, are viscous liquid or semisolid emulsions, either oil-in-water or water-in-oil. Topical formulations may also be in the form of a gel, i.e., a semisolid, suspension-type system, or in the form of a solution. Formulations of these drugs in dry powder form for delivery by a dry powder inhaler offers yet another means of administration. This overcomes many of the disadvantages of the oral and intravenous routes. The dosage of active ingredient in the compositions of this invention may be varied; however, it is necessary that the amount of the active ingredient shall be such that a suitable dosage form is obtained. The selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment desired. Generally, dosage levels of between 0.001 to 10 mg/kg of body weight daily are administered to mammals. Kits The present invention also relates to a kit for performing any of the above described methods, wherein the kit comprises a synthesized nucleoside triphosphate or a nucleic acid molecule comprising a synthesized nucleoside triphosphate. In one embodiment, the kit may comprise a mixture of synthesized nucleotides. In some embodiments, one or more of the components are premixed in the same reaction container. In particular embodiments, the kit additionally comprises instructional material. EXPERIMENTAL EXAMPLES The invention is further described in detail by reference to the following experimental examples. These examples are provided for purposes of illustration only, and are not intended to be limiting unless so specified. Thus, the invention should in no way be construed as being limited to the following examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein. Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the following illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. The following working examples therefore, specifically point out the preferred embodiments of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure Example 1: Synthesis of Natural and Modified Nucleoside Triphosphates Examples for the synthesis of the nucleotides of the present invention are given in this section. Synthesis of Activated Thymidine Monophosphate Activated thymidine monophosphate was synthesized according to the scheme shown inFIG.10. Synthesis of p-Toluene Pyrophosphate A p-toluene pyrophosphate reagent was synthesized according to the scheme shown inFIG.8. Synthesis of Pyrene Pyrophosphate A pyrene pyrophosphate reagent was synthesized according to the scheme shown inFIG.9. Synthesis of Nucleoside Triphosphates 5′-thymidine triphosphate was synthesized according to the scheme shown inFIG.2B. The activated nucleoside monophosphate is reacted with an organic pyrophosphate reagent (FIG.1,FIG.2A,FIG.5) to form an intermediate product (FIG.2C) which is then reduced to form a nucleoside triphosphate. The reaction shown inFIG.2Bis catalyzed by ZnCl2. The reaction ofFIG.2Bresults in the formation of dinucleoside diphosphate, a minor contaminant (FIG.4). 5′-thymidine triphosphate (5′-TTP), 3′-thymidine triphosphate (3′-TTP), and a-L-threofuranosyl 3′-thymidine triphosphate (tTTP)) as well as four naturally occurring nucleoside triphosphates have all been synthesized using the methods of the invention (FIG.6andFIG.7). Example 2: P(V) Reagents for the Scalable Synthesis of Natural and Modified Nucleoside Triphosphates Historically, the synthesis of chemically modified nucleoside triphosphates has been limited by such factors as scalability, low yields, difficult reaction conditions, and tedious purifications protocols (Burgess and Cook, 2000, Chem. Rev. 100, 2047-2060; Kore and Srinivasan, 2013, Curr. Org, Syn. 10, 903-934; Hollenstein, 2012, Molecules 17, 13569-13591). Efforts to overcome these problems have resulted in an astonishing number of publications, nearly all of which require HPLC purification (Burgess and Cook, 2000, Chem. Rev, 100, 2047-2060; Kore and Srinivasan, 2013, Curr. Org. Syn. 10, 903-934; Hollenstein, 2012, Molecules 17, 13569-13591). Attempts to synthesize α-L-threofuranosyl adenosine triphosphates using the classic Yoshikawa or Ludwig-Eckstein methods (Yoshikawa et at, 1967, Tetrahedron Lett. 8, 5065-5068; Ludwig and Eckstein, 1989, J. Org. Chem. 54, 631-635) produced complex mixtures of phosphorylated compounds with the desired compound present as a minor product (FIG.11). Therefore, a fundamentally different approach was developed for the synthesis of natural and modified nucleoside triphosphates that required invention of a novel P(V)-based organic pyrophosphate reagent to mediate P—O bond formation between the α- and β-phosphate positions, Here, a synthetic route to nucleic acid building blocks is described that (1) is not constrained to natural substrates; (2) is scalable to gram quantities of material; (3) eliminates the requirement for HPLC purification; and (4) avoids the need for freeze drying. The pyrophosphate reagent and activated nucleoside monophosphates described in this study are readily produced on scales of 5-10 grams using synthetic methodology that is inexpensive, straightforward and high yielding. The availability of these reagents makes it possible to generate 1.0 gram of nucleoside triphosphate, which is enough material to perform 5 liters of PCR or 100,000 PCR reactions with a standard 50 μL volume. Unlike conventional tributyl ammonium pyrophosphate, pyrene pyrophosphate is a solid at room temperature and stable to atmospheric conditions, allowing it to be used without specialized equipment and stored for long periods of time without loss of activity. Last, the pyrene pyrophosphate method was found to be superior for synthesizing chemically modified nucleotides that were challenging to synthesize using the classic Yoshikawa or Ludwig-Eckstein methods (14, 15), Based on these considerations, the current approach provides an alternative paradigm for synthesizing large quantities of nucleoside triphosphates for emerging applications in biotechnology and molecular medicine. The invention establishes an Hoard-Ott-like procedure (FIG.12) using nucleoside and pyrophosphate reagents that were suitably hydrophobic so as to generate nucleoside triphosphate derivatives that were amenable to purification by silica gel chromatography but could be readily converted to the desired compound using standard deprotection and precipitation conditions (Hoard and Ott, 1965, J. Am. Chem. Soc. 87, 1785-1788). This type of convergent synthesis strategy provides a direct and scalable route to natural and chemically modified nucleoside triphosphates using operationally simple protocols that process and discovery chemists would find appealing. Critical to this effort was the need to establish a new class of organic pyrophosphate reagents that contain a large hydrophobic moiety attached to the pyrophosphate group via a cleavable linker and to demonstrate that these reagents could mediate the synthesis of nucleoside triphosphates. Reducing this concept to practice involved a systematic evaluation of three key components: (1) hydrophobic groups necessary to construct a stable organic pyrophosphate reagent that is a solid compound at room temperature and exhibits minimal hygroscopic properties; (2) a strong leaving group to activate the nucleoside monophosphate for nucleophilic attack by the pyrophosphate reagent; and (3) a suitable Lewis acid to accelerate phosphodiester bond formation (FIG.12). The goals of this study were ultimately met through a systematic exploration and subsequent optimization of reaction conditions which revealed that pyrene, 2-methylimidazole, and zinc chloride provided the optimal hydrophobic moiety, leaving group, and Lewis acid catalyst, respectively, necessary to mediate the synthesis of nucleoside triphosphates in their fully protected form. The crystal structure of the pyretic pyrophosphate reagent verified correct synthesis of the organic pyrophosphate reagent (FIG.13). The materials and methods used are now described. Analytical Reverse-Phase HPLC Analysis. 2 μL of reaction crude (compounds: 10-11a-d, and 16-17, and 22-23a-d, and 28-29) in DMF or 1 μL of purified pyrene substituted nucleoside triphosphates (compounds: 12a-d, and 18, and 24a-d, and 30) in methanol or 3 of nucleoside triphosphates (compounds: 13a-d, and 19, and 25a-d, and 31) in H2O was added to 50 μL of 0.1 M triethylammonium acetate (TEAA) buffer pH 7.0. The solution was centrifuged by 4000 rpm for 2 minutes at room temperature and 30 μL of supernatant was injected for HPLC analysis, Reaction progress of the synthesis of nucleoside monophosphates (compounds: 10a-d, and 16, and 22a-d, and 28), nucleoside phosphor-2-methylimidazolides (compounds: 11a-d, and 17, and 23a-d, and 29), pyrene substituted nucleoside triphosphates (compounds: 12a-d, and 18, and 24a-d, and 30) and nucleoside triphosphates (compounds: 13a-d, and 19, and 25a-d, and 31) inFIG.32,FIG.44,FIG.56,FIG.68,FIG.80,FIG.92,FIG.101, FIG.110,FIG.119, andFIG.128was monitored by analytical HPLC chromatography with the gradient from 0% to 50% of acetonitrile in 0.1 M triethylammonium acetate buffer pH 7.0 over forty minutes. Purity determination of synthesized 5′-dNTPs (compounds: 13a-d) was monitored by the coinfection with the commercial 5′-dNTPs purchased from Sigma-Aldrich by analytical HPLC chromatography in with the gradient from 0% to 10% of acetonitrile in 0.1 M triethylammonium acetate buffer pH 7.0 over forty minutes of analytical run. PCR Fidelity Assay, A 100 μl PCR reaction was performed in 1× ThermoPol buffer (20 mM Tris-HCl, 10 mM (NH4)2SO4, 10 mM KCl, 2 mM MgSO4, 0.1% Triton X-100, pH 8.8, NEB), 0.5 of Fwd [CAACCGGTCCCCACGTTGCC] (SEQ ID NO:1) and Rev [AACGGCTGGGAGAACCTGGTTCTCAATGTA] (SEQ ID NO:2) PCR primers, 400 μM dNTPs (Chemically Synthesized vs Life Technologies), 4.4 ng of pGDR11 KOD-RS plasmid [Target sequence: CAACCGGTCCCCACGTTGCCGTTGCCAAGAGGTTGGCCGCGAGAGGAGTC AAAATACGCCCTGGAACGGTGATAAGCTACATCGTGCTCAAGGGCTCTGG GAGGATAGGCGACAGGGCGATACCGTTCGACGAGTTCGACCCGACGAAG CACAAGTACGACGCCGAGTACTACATTGAGAACCAGGTTCTCCCAGCCGT T] (SEQ ID NO:3) with final concentration of 5 units/100 μL Taq polymerase (NEB). The PCR conditions were: 95° C., 2.5 min (melt), 95° C., 30 sec, 62° C., 45 sec, 72° C., 30 sec for 20 cycles and an additional 72° C., 2 mm. The amplified amplicon (200 bp) was agarose purified, ligated into a TOPO-TA vector, and subsequently cloned into NEB DH5α E. colicompetent cells following the manufacturer's instructions, Individual colonies were grown in liquid media and sequenced using the M13F primer by Retrogen, San Diego, CA DNA sequences were aligned and analyzed using MEGA7 software. Five sequences clones were analyzed for each condition to give a total of 750 nucleotide positions. TNA Transcription Assay. Primer-extension reactions were performed in a final volume of 20 μl. Each reaction contained 10 pmol of primer [IR680-5′-GTCCCCTTGGGGATACCACC-3] (SEQ ID NO:4) annealed to 10 pmol of template [5′-ATCGAGTACAGTCAGATCGATATGATCTATATATTAATTAGGTGGTATCC CCAAGGGGAC-3] (SEQ ID NO:5), 1× ThermoPol buffer, 0.5 μM KOD-RS, 100 μM of each tNTP. Reactions were incubated tor 120 min at 55° C., quenched with stop buffer (40% Formamide and 1×TBE buffer, 10 mM EDTA), and analyzed by 10% denaturing urea PAGE. The results of the experiments are now described. The pyrene pyrophosphate method was applied to the synthesis of natural thymidine-5′-triphosphate (dTTP). As shown inFIG.14AFIG.14C, activated thymidine-5′-monophosphate is readily converted to a fully protected thymidine-5′-triphosphate derivative, which was purified by silica gel chromatography, deprotected, and precipitated to obtain the desired product as a highly pure compound in sodium form. In this reaction, 10 molar equivalents of ZnCl2enabled a near quantitative coupling (>95%) of the pyrene pyrophosphate reagent to the activated nucleoside monophosphate after 3 hours of stirring in DMF at 24° C. The phosphorylation reaction also generated trace amounts (1-2%) of unwanted side products, including dinucleotide diphosphate, that were removed by silica gel purification using a 10% H2O/isopropanol mobile phase containing 1% base [diisopropylethylamine, DIPEA] as an organic counterion. Following purification, the triphosphate intermediate was deprotected with concentrated ammonium hydroxide (33% aq.) and precipitated as the sodium salt using standard conditions. Analytical HPLC analysis reveals that chemically synthesized dTTP was equivalent in purity to a commercial standard (FIG.14C), demonstrating that the process of the nucleotide triphosphate synthesis is capable of generating material that is identical in purity to commercial compounds obtained by conventional enzymatic synthesis. Because enzymatic approaches are not compatible with most nucleic acid analogs, it was decided to broaden the scope of the reaction by applying the pyrene pyrophosphate method to a select panel of thymidine analogs (FIG.15). The list of target molecules included thymidine-3′-triphosphate (a regioisomer of natural dTTP), L-threofuranosyl thymidine 3′-triphosphate (tTTP, a building block for threose nucleic acid, TNA) (Schoning et al., 2000, Science 290, 1347-1351), and L-thymidine-5′-triphosphate (the mirror image form of natural D-dTTP). The last two examples have become valuable compounds for biomedical research due to recent advances in polymerase engineering that have enabled the synthesis of artificial genetic polymers with non-natural sugar-phosphate backbones (Larsen et at, 2016, Nat. Common. 11235; Wang et al., 2016, Nat. Chem. 8, 698-704), some of which have been used to evolve biologically stable aptamers and catalysts (Pinheiro et al., 2012, Science 336, 341-344; Yu et al., 2012, Nat. Chem. 4, 183-187; Taylor et al., 2015, Nature 518, 427-430; Wang et al., 2018, Nat. Commun., in press). As with natural dTTP, each unnatural thymidine triphosphate was obtained in highly pure form (>95%) as the desired sodium salt. Occasionally, a small amount of nucleoside diphosphate (dNDP) (˜1-2%) was observed after deprotection of the protected nucleoside triphosphates. dNDPs are common contaminants of commercial nucleotide triphosphates due to the slow hydrolysis of ATP to ADP (Hulett, 1970, Nature 225, 1248-1249). However, these molecules do not interfere with normal DNA synthesis as natural DNA polymerases are highly specific for dNTP substrates (Steitz al., 1994, Science 266, 2022-2025). Next, a complete set of DNA nucleoside triphosphates (dNTPs) was prepared using the pyrene pyrophosphate method. Each activated nucleoside monophosphate was converted to the desired nucleoside triphosphate following the standard procedure of pyrene pyrophosphate coupling, purification by silica gel chromatography, deprotection with concentrated ammonium hydroxide, and precipitation as the sodium salt (FIG.16A). Concurrently, all four TNA nucleoside triphosphates (tNTPs) were synthesized, which are the substrates for engineered TNA polymerases developed by directed evolution (FIG.16B) (Larsen et al., 2016, Nat. Commun. 7, 11235). In all cases, analytical HPLC chromatograms reveal high conversion (>95%) of activated nucleoside triphosphate into the fully protected nucleoside triphosphate. Following deprotection and precipitation, the desired DNA and TNA nucleoside triphosphates were produced in highly pure form on scales (>500 milligrams) and timeframes (2-3 days) vastly exceeding those of traditional protocols. Since nucleoside triphosphates would ultimately be used as substrates for oligonucleotide synthesis, the use of these reagents was investigated in a conventional DNA synthesis assay. One critical question that this study aimed to address is whether chemically synthesized DNA triphosphates function with same level of efficiency as enzymatically produced dNTPs obtained from commercial venders. To address this question, the polymerase chain reaction (PCR) was used to amplify an arbitrary DNA sample in reaction mixtures that contained either chemically synthesized or commercial dNTPs. A 200 base pair segment was amplified that defines the finger subdomain of an archaeal DNA polymerase isolated from the thermophilic speciesThermococcus kodakarensis(Kod) (Chico et al., 2017, Nat Common 8, 1810). Analysis of the resulting PCR amplicons by agarose gel electrophoresis confirmed that chemically synthesized dNTP substrates function identically to commercial dNTPs, as both reactions produce equivalent amounts of DNA at each cycle of PCR amplification (FIG.16A). Moreover, DNA sequencing of the amplified product failed to identify any instances of insertions, deletions, or mutations, indicating that the reactions proceed with high template-sequence fidelity. Next, the set of chemically synthesized TNA triphosphates was evaluated as substrates for TNA synthesis using an engineered TNA polymerase (Chim et al., 2017, Nat Common 8, 1810). In this assay, a DNA primer-template complex was extended with TNA using either newly synthesized tNTPs or tNTPs obtained by a traditional Hoard-Ott-like approach that requires HPLC purification (Bala et al, 2017, J. Org. Chem. 82, 5910-5916). Analysis of the resulting primer-extension reactions by denaturing polyacrylamide gel electrophoresis reveals that the two sets of TNA substrates generate equivalent amounts of full-length TNA product after 3 hours of incubation at 55° C. (FIG.16B), This result, along with the PCR assay, confirms that the pyrene pyrophosphate method produces high quality nucleoside triphosphates that function as substrates for natural and engineered polymerases. Example 3: Synthesis of 1-(2-(pyrenesulfonyl)ethyl)pyrophosphate FIG.17shows the synthesis scheme for 1-(2-(pyrenesulfonyl)ethyl)pyrophosphate (compound 7),FIG.18throughFIG.31provide spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. 2-(Pyrenethio)ethanol (Compound 2) To a solution containing 9.98 g (177.8 mmol) of potassium hydroxide in 250 mL of anhydrous DMF was added 12.5 mL (177.8 mmol) of 2-mercaptoethanol. The reaction mixture was heated to 80° C. with stirring until potassium hydroxide dissolved. A premade solution containing 25 g (88.92 mmol) of 1-bromopyrene (compound 1) in 200 mL of anhydrous DMF was dropwise added to the reaction and stirred at 110° C. for 3 hours. Then, the reaction was cooled to room temperature, condensed to 100 mL of solution under diminished pressure and diluted into 500 mil, of CH2Cl2. The organic layer was washed with 200 mL of H2O four times. And the combined aqueous layer was back extracted with 200 mL of CH7Cl2two times. The organic layers were combined and evaporated to the dryness under diminished pressure. The crude product was purified by silica column chromatography with eluents (CH2Cl2/Hexane, 25% to 50%, then EtOAc/CH2Cl2, from 0% to 2%) to afford the compound 2 as a yellowish solid; yield: 21.3 g (85.8%); silica gel TLC (EtOAc/Hexane, 1:2) Rf=0.35; NMR (400 MHz, CDCl3) δ 8.61 (d, 1H, J=9.2 Hz), 8.07-8.00 (m, 4H), 7.93-7.81 (m, 4H), 3.71 (t, 2H, J=6.0 Hz), 3.18 (t 2H, J=6.0 Hz); HRMS (ESI-TOF) calcd. for C18H14OSNa [M+Na]+301.0663. found 301.0658. Compound 2 has been reported in Kathayat et al., 2013, J. Am. Chem. Soc. 135, 12612-12614. 2-(Pyrenesulfonyl)ethanol (Compound 3 To a solution containing 71 g (114.9 mmol, 80% technical grade) of magnesium bis(monoperoxyphthalate) hexahydrate in 0.450 mL of anhydrous DMF was slowly added a pre-made solution of 21.3 g (76.6 mmol) of compound (2) in 150 of anhydrous DMF at 0° C. The reaction was stirring at room temperature for 12 hours. At which the TLC showed the reaction was finished, the reaction was dropwise added to a solution containing 2 L of satd. NaHCO3(aq)while stirring. The precipitate was washed with 300 mL of H2O twice, and dried under high vacuum to afford the compound 3 as a yellowish solid; yield: 22.8 g (96%); silica gel TLC (EtOAc/CH2Cl2. 1:20) Rf=0.22;1H NMR (400 MHz, CDCl3) δ 9.00 (d, 1H, J=9.2 Hz), 8.71 (d, 1H, J=8.4 Hz), 8.34-8.31 (m, 3H), 8.27-8.23 (m, 2H), 8.14-8.09 (in, 214), 4.00 (t, 21H, J=5.2 Hz), 3.64 (t, 2H, J=5.2 Hz);13C NMR (125.8 MHz, CDCl3-10% DMSO-d6) δ 135.5, 130.8, 130.7, 130.6, 130.3, 129.9, 128.9, 127.5, 127.2, 127.1, 127.0, 126.9, 125.0, 124.1, 123.9, 122.4, 58.6, 56.1; HRMS (ESI-TOF) calcd. for C18H14O3SNa [M+Na]+333.0561. found 333.0571. Dibenzyl-1-(2-(pyrenesulfonyl)ethyl) Monophosphate (Compound 4) To a mixture containing 5 g (16.13 mmol) of 2-(pyrenesulfonyl)ethanol (compound 3), 2.03 g (29.03 mmol) of tetrazole in 106 mL of anhydrous solution (MeCN/CH2Cl2, 1:1) was slowly added 6.64 mL (20.96 mmol) of (BnO)2PN(i-Pr)2at room temperature. The reaction was sonicated until the solid disappeared (otherwise add additional 20 mL of anhydrous DMF) and the mixture was stirring at room temperature for 3 hours. Then reaction was cooled to −40° C. followed by adding 15 mL of H2O (33% in 1-120) for additional 1 h stirring at room temperature. The solution was condensed to 20 mL under diminished pressure and poured to 150 mL EtOAc. The organic layer was washed with 100 mL brine, 200 mL satd. NaHCO3, 200 mL H2O, dried over MgSO4and evaporated under diminished pressure. The crude product was purified by silica column chromatography with eluent (CH2Cl2/Hexane, from 25% to 100%, then MeOH/CH2Cl2, from 1% to 3%) to afford the yellowish solid product, compound 4; yield: 6.73 g (73.2%); TLC (EtOAc/CH2Cl21:20) Rf=0.34;1H NMR (400 MHz, CDCl3) δ 8.89 (d, 1H, J=9.2 Hz), 8.60 (d, 1H, J=8.0 Hz), 8.15-8.11 (m, 3H), 8.01-7.94 (m, 3H) 7.82 (d, J 8.8 Hz), 7.20-7.16 (m, 611), 7.06-7.04 (m, 4H), 4.67 (d, 4H, J=8.4 Hz), 4.36 (dd. 2H, J=14, 6.0 Hz), 3.70 (t, 2H, J=6.0 Hz);13C NMR (125.8 MHz, CDCl3) δ135.2, 135.2, 135.1, 130.5, 130.4, 129.9, 129.5, 128.7, 128.3, 128.3, 127.6, 127.4, 127.1, 127.0, 126.7, 126.6, 124.5, 123.9, 123.4, 122.0, 69.2 (d, JC, P=5.8 Hz), 60.7 (d, JC, P=5.0 Hz), 56.3 (d, JC, P=7.2 Hz);31P NMR (162 MHz, CDCl3) δ −0.41; HRMS (ESI-TOF) calcd. for C32H27O6PSNa [M+Na]+593.1164. found 593.1158. 1-(2-(Pyrenesulfonyl)ethyl) Monophosphate (Compound 5) To a solution containing 6.73 g (11.8 mmol) of dibenzyl-1-(2-(pyrenesulfonyl)ethyl) monophosphate (compound 4) in 50 mL of MeOH was purged with nitrogen gas. The solution was added to 0.1-0.2 mass equivalent of 10% Pd/C, and repurged with nitrogen gas followed by adding hydrogen gas. The mixture was stirred at room temperature for 3 hours with monitoring by TLC (MeOH/CH2Cl2, 1:10 with 1% Et3N), The resulting suspension was filtered over a pad of celite, washed with 100 mL of MeOH four times, evaporated, and then coevaporated with dry C2H1Cl2under reduced pressure to afford the product, compound 5, as a yellowish solid; yield 4.42 g (96%); TLC (MeOH/CH2Cl2, 1:10 with 1% triethylamine) Rf=0;1H NMR (400 MHz, CD3OD) δ 8.72 (d, 1H, J=9.6 Hz), 8.44 (d, 1H, J=8.4 Hz), 8.00-7.90 (m, 4H), 7.79-7.75 (m, 2H), 7.66 (d, 1H, J=8.8 Hz), 4.34 (dd, 2H, J=12.4, 6.0 Hz), 3.82 (t, 214, J===6.0 Hz);13C NMR (125.8 MHz, CD3OD+10% DMSO-d6) δ 136.6, 131.7, 131.5, 131.5, 131.4, 130.8, 129.8, 128.4, 128.2, 128.0, 127.8, 127.7, 125.5, 125.2, 124.4, 123.2, 60.9 (d, JC, P=4.2 Hz), 57.6 (d, JC, P=7.2 Hz);31P NMR (162 MHz, CD3OD) δ −0.56. HRMS (ESI-TOF) calcd. for C18H14O6PS [M−H]−389.0249. found 389.0234. 1-(2-(Pyrenesulfonyl)ethyl)-(β-dibenzyl)pyrophosphate (Compound 6) To the 4.42 g (11.3 mmol) of 1-(2-(pyrenesulfonyl)ethyl) monophosphate (compound 5) in 39.3 mL of anhydrous dichloromethane was added 5.33 mL (15.82 mmol) of (BnO)2PN(i-Pr)2at room temperature under a nitrogen atmosphere. After two hours stirring, the reaction was slowly added to 6.16 mL (33.9 mmol) of tert-butyl hydrogen peroxide (5.5 Min decane) at −40° C. and stirred for an additional 30 minutes at room temperature. The reaction was evaporated under diminished pressure and the crude product was purified by silica column chromatography with the eluents (MeCN then MeOH/CH2Cl2from 1% to 3% containing 1% triethylamine) to afford the solid product, compound 6; yield: 6.5 g (76.6%); TLC (2:100 MeOH—CH2Cl2with 1% triethylamine) 0.26;1H NMR (400 MHz, CD3OD) 8.90 (d, 1H, J=9.6 Hz), 8.56 (d, 1H, J=8.0 Hz), 8.15-8.11 (m, 3H), 8.06-8.00 (m, 211), 7.94 (t, 1H, J=8.0 Hz), 7.87 (d, 1H, J=8.8 Hz), 7.19 (m, 10H), 4.94 (d, 4H, J=7.6 Hz), 4.32 (dd, 2H, J=15.2, 6.8 Hz), 3.75 (t, 2H, J=6.8 Hz);13C NMR (125.8 MHz, CDCl3)<136.0, 135.9, 135.6, 130.9, 130.7, 130.7, 130.6, 130.0, 129.1, 128.5, 128.3, 127.8, 127.7, 127.3, 127.2, 127.1, 127.0, 125.1, 124.3, 124.0, 122.6, 69.3 (d, JC, P=5.5 Hz), 59.7 (d, JC, P=5.5 Hz), 57.0 (d, JC, P=6.9 Hz);31P NMR (162 MHz, CDCl3) δ −11.2 (d, 18.5 Hz), −11.6 (d, J=18.5 Hz); HRMS (ESI-TOF) calcd. for C32H27O9P2SNa2[M−H+2Na]+695.0646. found 695.0660. 1-2-(Pyrenesulfonyl)ethyl)pyrophosphate (Compound 7) To a solution containing 6.5 g of 1-(2-(pyrenesulfonyl)ethyl)-β-dibenzyl)pyrophosphate (compound 6) in 30 mL of MeOH was purged with nitrogen gas. The solution was added to 0.1-0.2 mass equivalent of 10% Pd/C and purged with nitrogen gas followed by adding hydrogen gas. The mixture was stirred at room temperature for 3 h with monitoring by TLC (MeOH/CH2Cl2, 1:10 with 1% Et3N). The resulting suspension was filtered over a pad of celite, washed with 100 mL of MeOH three times, evaporated, and coevaporated with dry C2H4Cl2under reduced pressure to afford the product as a yellowish solid (compound 7); yield: 4.27 g (90.9%); TLC (MeOH/CH2Cl2, 1:10 with 1% triethylamine) Rf=0;1H NMR (400 MHz, CD3OD) 8.93 (d, 1H, J=9.2 Hz), 8.61 (d, 1H, J=8.0 Hz), 8.29-8.22 (m, 4H), 8.13 (d, 8.8 Hz), 8.05-8.00 (m, 2H), 4.35 (m, 2H), 3.91 (t, 2H, J=10.4 Hz);13C NMR (125.8 MHz, D2O) δ 135.0, 130.7, 130.1, 129.7, 129.2, 129.0, 128.3, 127.3, 127.1, 127.0, 126.6, 126.3, 124.4, 123.6, 122.4, 121.8, 60.6, 57.3 (d, JC, P=6.0 Hz);31P NMR (162 MHz, D2O) δ −10.0 (d, J=16.8 Hz), −11.5 (d, 16.8 Hz); HRMS (ESI-TOF) calcd. for C18H14O9P2SNa3[M−2H+3Na]+536.9527. found 536.9521. Example 4: Synthesis of Nucleoside Triphosphates FIG.32throughFIG.141depict schematic diagrams for the synthesis of nucleoside triphosphates and NMR spectra of the synthesized intermediates and products. Methods used for the synthesis are now described. General Procedure A: Synthesis of Protected Nucleoside Monophosphates To a mixture containing suitably protected free nucleoside (compounds: 8a-d, 14, 20a-d, and 1.8 equivalents of tetrazole in an anhydrous solvent (MeCN/CH2Cl2, 1:1) was added to 1.3 equivalents of (BnO)2PN(i-Pr)2under a nitrogen atmosphere. The mixture was stirred at room temperature for 1-3 hours and the reaction was monitored by TLC. At which the starting material was consumed, the reaction was slowly added excess H2O2(33% in H2O) at −40° C. and the resulting mixture was stirred at room temperature for 1 hour. The solution was then diluted with 15-20 times volume of CH2Cl2and the organic layer was sequentially washed with saturated NaHCO3(aq), brine, and water. The organic extracts were combined, dried with MgSO4, and evaporated under diminished pressure. The crude residue was purified by silica gel chromatography and the fractions containing the product were collected and evaporated to afford the product, compounds: 9a-d, 15, 21a-d, or 27. General Procedure B: Synthesis of Nucleoside Monophosphates To a solution containing the protected nucleoside monophosphate (compounds 9a-d, 15, 21a-d, and in MeOH was purged with nitrogen gas. The solution was added 0.1-0.2 mass equivalent of 10% Pd/C, and re-purged with nitrogen gas followed by adding hydrogen gas. The mixture was stirred at room temperature for 3-5 hours with monitoring by TLC (MeOH/CH2Cl2, 1:10 with 1% triethylamine). Then the solution was purged with nitrogen, filtered over a pad of celite, and washed with MeOH or MeOH containing 2% triethylamine. The filtrate was collected, evaporated and coevaporated with dry 1, 2-dichloroethane under reduced pressure to afford the product, compounds: 10a-d, 16, 22a-d, or 28. General Procedure C: Synthesis of Activated Nucleoside Monophosphates To a solution containing the nucleoside monophosphate (compounds 10a-d, 16, 22a-d and 28) in anhydrous DMF under a nitrogen atmosphere was slowly added 5 equivalents of anhydrous triethylamine at 0° C. After 5 minutes with stirring, 2 equivalents of 2-methylimidazole was added followed by 2 equivalents of triphenylphosphine. After 10 minutes of stirring at room temperature, 2 equivalents of 2, 2′-dipyridyl disulfide was added and stirring was continued for an additional 3 hours at room temperature with monitoring by analytical HPLC (mobile phase: MeCN/0.1 M TEAA buffer, from 0% to 50% over 40 minutes). After consumption of the starting material, the product was precipitated by adding the reaction dropwise with stirring to 300 mL of diethyl ether. The precipitate was collected by centrifuging at 4400 rpm for 15 minutes at room temperature. The supernatant was discarded, and the pellet was resuspended with minimal amount of CH2Cl2or anhydrous DMF (DMF is used when the nucleoside monophosphate is not soluble in CH2Cl2). The solution was added dropwise to a premade solution of ether/ethyl acetate/triethylamine (5:10:1) containing 8 equivalents of sodium perchlorate for a second precipitation. The suspended solid was centrifuged at 4400 rpm for 15 minutes at room temperature, the supernatant was discarded, and the pellet was washed twice with 40 mL of mixed solvent (ether/ethyl acetate, 1:2), and dried under high vacuum to afford the product, compounds: 11a-d, 17, 23a-d or 29. General Procedure 1): Synthesis of Fully Protected Nucleoside Triphosphates To a mixture containing the activated nucleoside monophosphate (compounds 11a-d, 17, 23a-d, and 29) and 1.2 equivalents of 1-(2-(pyrenesulfonyl)ethyl)pyrophosphate (compound 7) was added along with 8-10 equivalents of a premade solution of ZnCl2(1.0 M in anhydrous DMF) under a nitrogen atmosphere. The mixture was stirred at room temperature for 3-5 hours and the reaction progress was monitored by HPLC (MeCN/0.1 M TEAA buffer, from 0% to 50% over 40 minutes). At which the starting material was consumed, the reaction was dropwise added to ethyl acetate or ether for precipitation. The precipitate was centrifuged and collected at 4400 rpm for 10 minutes at room temperature, and the supernatant was discarded. The pellet was resuspended by 20% H2O in MeCN with 2% Hunig's base and the solid was filtered by pyrex glass funnel with filter paper. The filtrate was collected and evaporated under diminished pressure and dry-packing loaded to silica gel for normal phase silica column chromatography. The fractions containing the product were collected and evaporated under diminished pressure at 30-40° C. The product was resuspended with CH2Cl2and insoluble silica gel was removed by filtration. The filtrate was collected and evaporated to dryness to afford the product, compounds: 12a-d, 18, 24a-d, or 30. General Procedure Synthesis of Free Nucleoside Triphosphates To a solution of fully protected nucleoside triphosphate (compounds: 12a, 18, 24a, and 301 in 50 mL of 33% NH4OH(aq)was stirred for 18 hours at room temperature or (compounds: 12b-d, and 24b-d) was stirred for 3 hours at 37° C. and 15 hours at room temperature in a sealed tube. After the reaction, the solvent was evaporated under diminished pressure. The solid was resuspended with water and the aqueous solution was washed with CH2Cl2and ethyl acetate. The organic portion was discarded and the aqueous extract was collected, and evaporated under diminished pressure. The crude solid was resuspended with minimal amount of RNAse free water, filtrated by 0.22 um syringe filter and dropwise added to the forty times volume of acetone at room temperature containing 15 equivalents of sodium perchlorate. The resulting suspension was centrifuged at 4400 rpm for 15 minutes at room temperature. The supernatant was discarded and the pellet was washed with organic solution acetone/CH2Cl2, 10:1) twice to afford the product, compounds: 13a-d, 19, 25a-d, or 31. Synthesis of thymidine-5′-triphosphate FIG.32shows the synthesis scheme for 1-(2-(pyrenesulfonyl)ethyl)pyrophosphate (dTTP, compound 13a).FIG.33throughFIG.43provide spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. 3′-O-Benzoyl-2′-deoxythymidine-5′-dibenzylmonophosphate (Compound 9a) General procedure A with 1 g (2.89 mmol) of 3′-O-benzoyl-2′-deoxythymidine compound 8a, 364.4 mg (5.2 mmol) of tetrazole, 28.9 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 1.3 mL (3.76 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 3 hours reaction at room temperature. Then, 6 mL of 30% H2O2(aq)for 1 hour oxidation reaction at room temperature. Column chromatography with eluents (MeOH/CH2Cl2, from 1% to 1.4%) to afford the product compound 9a as a white solid; yield: 1.2 g (68.5%); TLC (MeOH/CH2Cl2, 1:40) Rf=0.23;1H NMR (400 MHz, CD3OD) δ 8.03-8.00 (m, 2H), 7.62-7.58 (m, 1H), 7.48-7.45 (m, 3H), 7.36-7.29 (m, 10H), 6.32 (dd, 1H, J=8.4, 6.0 Hz), 5.42-5.40 (m, 1H), 5.12-5.07 (m, 4H), 4.34-4.28 (m, 3H), 2.47 (ddd, 1H, J=14.4, 6.0, 2.0 Hz), 2.26-2.18 (m, 1H), 1.77 (d, 3H, J=1.2 Hz); HRMS (ESI-TOF) calcd. for C31H31N2O9PNa [M+Na]+629.1665. found 629.1664. Compound 9a has been reported in De et al., 2014, Eur. J. Org. Chem. 2322-2348. 3′-O-Benzoyl-2′-deoxythymidine-5′-monophosphate (Compound 10a) General procedure B with 1.2 g (1.98 mmol) of compound 9a, 50 mL of MeOH, and 200 mg of 10% Pd/C for 3 hours stirring at room temperature. The suspension was filtered over a pad of celite, and washed with 100 mL of MeOH containing 2% triethylamine four times to afford the product compound 10a as a white foam of triethylammonium salt; yield: 1.11 g (88.7%); TLC (MeOH/CH2Cl2, 1:10 with 1% triethylamine) Rf=0;1H NMR (400 MHz, DMSO-d6) δ 11.41 (s, 1H), 8.09 (d, 2H, J=8.0 Hz), 7.77-7.74 (m, 2H), 7.62 (t, 2H, J=7.6 Hz), 6.41 (dd, 8.0, 6.8 Hz), 5.57 (s, 1H), 4.41 (s, 1H), 4.18 (m, 2H), 2.58-2.57 (m, 2H), 1.88 (s, 3H);13C NMR (125.8 MHz, DMSO-d6) δ 168.6, 167.3, 152.7, 138.6, 135.6, 131.0, 130.4, 130.3, 113.0, 86.2, 85.1 (d, JC, P=8.7 Hz), 78.0, 66.2, 37.9, 31.9, 13.2;31P NMR (162 MHz, DMSO-d6) δ 0.24; HRMS (ESI-TOF) calcd. for C17H18N2O9PNa2[M−H+2Na]+471.0545. found 471.0539. 3′-O-Benzoyl-2′-deoxythymidine-5′-phosphor-2-methylimidazole (Compound 11a) General procedure C with 1.11 g (1.76 mmol) of compound 10a, 7.8 mL of anhydrous DMF, 1.63 mL (11.72 mmol) of triethylamine, 385 mg (4.69 mmol) of 2-methylimidazole, 1.23 g (4.69 mmol) of triphenylphosphine, 1.04 g (4.72 mmol) of dipyridyl disulfide for 2 hours reaction at room temperature. First precipitation was achieved with 250 mL of diethyl ether. The product was resuspended with 10 mL of DMF and dropwise added to the solution containing 2.01 g of sodium perchlorate, 15 mL of triethylamine in 300 mL of ethyl acetate for second precipitation. The product was afforded as a white solid compound 11a; yield: 0.89 g (98.9%);31P NMR (162 MHz, D2O) δ −6.83; HRMS (ESI-TOF) calcd. for C21H23N4O8PNa [M+Na]+513.1151. found 513.1141. 3′-O-Benzoyl-2′-deoxythymidine-5′-(γ-(2-(pyrenesulfonyl)ethyl))triphosphate (Compound 12a) General procedure 1) with 0.89 g (1.74 mmol) of compound 11a, 1.09 g (1.91 mmol) of 1-2-(pyrenesulfonyl)ethyl) pyrophosphate (compound 7) and 13932 mL (13.92 mmol) of ZnCl2solution (1.0 M in anhydrous DMF) for 3 hours stirring at room temperature. Then crude was precipitated by dropwise adding the solution to the 300 mL of ethyl acetate with stirring, After centrifugation, the pellet was resuspended by 20% H2O/MeCN containing 2% Hunig's base and filtered by pyrex glass funnel. The filtrate was evaporated and the crude material was purified by silica column chromatography with eluents (H2O/(isopropanol-MeCN 1:1) from 2% to 7% containing 1% diisopropylethylamine (DIPEA) to obtain pure compound 12a; yield: 1.43 g (64.9%); TLC (H2O/acetone 1:10 with 2% diisopropylethylamine) Rf=0.29;1H NMR (400 MHz, D2O) δ 8.25 (d, 1H, J=8.8 Hz), 8.10 (d, 1H, J=7.6 Hz), 7.41 (d, 1H, J=9.2 Hz) 7.33-7.27 (m, 3H), 7.19-7.13 (m, 4H), 6.94-6.89 (m, 3H), 6.68 (t, 2H, J=6.4 Hz), 5.61 (s, 1H), 5.16 (s, 1H), 4.49 (s, 2H), 4.15-4.07 (m, 2H), 3.78 (m, 3H) 7.04-1.94 (m, 2H), 1070 (s, 3H);31P NMR (162 MHz, D2O) δ −10.22 (brs, 2P), −20.50 (brs, LP); HRMS (ESI-TOF) calcd. for C35H31N2O17P3SNa3[M−2H+3Na]+945.0250; found 945.0261. 2′-deoxythymidine-5′-triphosphate (Compound 13a) General procedure E with 1.43 g (1.13 mmol) of compound 12a, 50 mL of 33% NH4OH(aq)for 18 h stirring at room temperature. The product compound 13a was afforded as a white solid; yield: 453 mg (83.2%, T, ε267: 9600 M−1cm−1);1H NMR (400 MHz, D2O) δ 7.74 (s, 1H), 6.37 (t, 1H, J=6.0 Hz), 4.69 (s, 1H), 4.28-4.24 (m, 3H), 2.42 (s, 2H), 1.96 (s, 3H);31P NMR (162 MHz, D2O) 6-4.06 (d, 16.2 Hz), −9.30 (d, J=16.2 Hz), −17095 (brs). Synthesis of 2′-deoxycytidine-5′-triphosphate FIG.44shows the synthesis scheme for 2′-deoxycytidine-5′-triphosphate (dCTP, compound 13b).FIG.45throughFIG.55provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. 3′-O, N4-Dibenzoyl-2′-deoxycytidine-5′-dibenzylmonophosphate (Compound 9b) General procedure A with 1.04 g (2.40 mmol) of 3′-O, N4-dibenzoyl-2′-deoxythymidine compound 8b, 306 mg (4.31 mmol) of tetrazole, 16 mL of anhydrous solution (MeCN; CH2Cl2, 1:1), 0.99 mL (3.12 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 1 hour stirring at room temperature followed by 6 mL of H2O2for 1 hour oxidation reaction. Column chromatography with eluents (MeOH/CH2Cl2from 0% to 12.5%) to afford the product compound 9b as a white solid; yield: 1.26 g (75.5%); TLC (MeOH/CH2Cl2, 1:60) Rf=0.32;1H NMR (400 MHz, CDCl3) δ 8.12 (d, 1H, J=7.6 Hz), 8.02 (d, 2H, J=7.6 Hz), 7.96 (d, 2H, J=7.6 Hz), 7.59-7.52 (m, 2H), 7.48-7.41 (m, 5H), 7.36-7.28 (m, 10H), 6.37 (dd, 1H, J=8.4, 5.6 Hz), 5.41 (d, 1H, J=6.4 Hz), 5.14-5.02 (m, 4H), 4.42-4.32 (m, 3H), 2.82 (dd, 1H, J=14.0, 5.2 Hz), 2.05-1.97 (m, 1H);13C NMR (125.8 MHz, CDCl3) δ 165.7, 162.4, 154.6, 143.8, 135.4, 135.3, 133.4, 133.1, 132.9, 129.6, 128.9, 128.7, 128.6, 128.6, 128.4, 127.9, 127.9, 127.7, 96.9, 87.0, 83.6 (d, JC, P=7.8 Hz), 75.0, 69.7 (d, JC, P=5.3 Hz), 66.8 (d, JC, P=5.2 Hz), 38.7;31P NMR (162 MHz, CDCl3) δ 0.76; HRMS (ESI-TOF) calcd. for C37H34N3O9PNa [M+Na]+718.1931. found 718.1949. 3′-O, N4-Dibenzoyl-2′-deoxycytidine-5′-monophosphate (Compound 10b) General procedure B with 1.26 g (1.81 mmol) of compound 9b, 100 mL of MeOH, and 250 mg of 10% Pd/C for 3 h stirring. The suspension was filtered over a pad of celite, washed with 100 mL of MeOH containing 2% triethylamine four times to afford the product compound 10a as a white foam of triethylammonium salt; yield: 1.05 g (84.1%); TLC (MeOH/CH2Cl2, 1:10) Rf=0;1H NMR (400 MHz, CD3OD) δ 8.68 (d, 1H, J=6.8 Hz), 8.08 (d, 2H, J=7.6 Hz), 7.98 (d, 2H, J=7.6 Hz), 7.71 (d, 1H, J=7.2 Hz), 7.65-7.62 (m, 2H), 7.55-7.49 (m, 4H), 6.45 (t, 1H, J=6.0 Hz), 5.69 (d, 1H, J=4.0 Hz), 4.51 (s, 1H), 4.22 (s, 1H), 2.78 (dd, 1H, J=13.6, 4.0 Hz), 2.59-2.52 (m, 1H);13CNMR (125.8 MHz, CD3OD) δ 168.9, 167.2, 164.9, 157.9, 146.9, 134.8, 134.5, 134.0, 131.0, 130.6, 129.8, 129.7, 129.1, 99.1, 88.7, 86.5 (d, JC, P=8.4 Hz), 77.8, 65.8, 40.1;31P NMR (162 MHz, CD3OD) δ 1.22; HRMS (ESI-TOF) calcd. for C23H22N3O9PNa [M+Na]+538.0991. found 538.0984. 3′-O, N4-Dibenzoyl-2′-deoxycytidine-5′ phosphor-2-methylimidazolide (Compound 11b) General procedure C with 1.05 g (1.46 mmol) of compound 10b, 7.3 mL of anhydrous DMF, 1.02 mL (7.30 mmol) of triethylamine, 252 mg (3.07 mmol) of 2-methylimidazole, 918 mg (3.5 mmol) of triphenylphosphine, 804 mg (3.65 mmol) of dipyridyl disulfide for 2 hours reaction at room temperature. First precipitation was achieved with 250 mL of diethyl ether. The product was resuspended with 10 mL of DMF and dropwise added to the solution containing 1.79 g of sodium perchlorate, 15 mL of triethylamine in 300 mL of mixing solution (ethyl acetate/ether 1.5:1) for second precipitation. The product was afforded as a white solid compound 11b; yield: 0.82 g (93.4%);31P NMR (162 MHz, D2O) δ−6.63; HRMS (ESI-TOF) calcd. for C27H25N5O8PNa2[M−H+2Na]+624.1236. found 624.1256. 3′-O,N4-Dibenzoyl-2′-deoxycytidine-5′-(γ-(2-(pyrenesulfonyl)ethyl))triphosphate (Compound 12b) General procedure D with 0.82 g (1.36 mmol) of compound 11b, 0.77 g (1.63 mmol) of 1-(2-(pyrenesulfonyl)ethyl) pyrophosphate (compound 7) and 13.6 mL (13.6 mmol) of ZnCl2solution (1.0 M in anhydrous DMF) for 3 hours with stirring. Then crude was precipitated by dropwise adding the solution to the 300 mL of ethyl acetate with stirring for precipitation. After centrifugation, the pellet was resuspended by 20% H2O/MeCN containing 2% Hunig's base and filtered by pyrex glass funnel. The filtrate was evaporated and crude product was purified by silica column chromatography with eluents 7% H2O/isopropanol containing 1% diisopropylethylamine (DIPEA) and then H2O in (isopropanol/MeCN 1:1) from 5% to 8% containing 1% diisopropylethylamine (DIPEA) to afford the white solid compound 12b; yield: 1.07 g (58.1%); TLC (H2O/acetone 1:10 containing 2% DIPEA): Rf=0.32; 1H NMR (500 MHz, D2O/DMSO-d61:1) δ 11.15 (s, 1H), 8.91 (s, 1H), 8.60-7.95 (m, 7H), 7.62-7.33 (m, 4H), 6.25 (s, 1H), 5.47 (s, 1H), 4.33-3.90 (m, 4H), 2.34 (s, 1H);31P NMR (162 MHz, D2O/DMSO-d61:1) δ −12.13 (d, 1P, J=17.2 Hz), −12.38 (d, 1P, J=14.9 Hz), −22.59 (brs, 1P); HRMS (ESI-TOF) calcd. for C41H34N3O17P3SNa3[M−2H+3Na]+1034.0515. found 1034.0485. 2′-deoxythymidine-5′-triphosphate (Compound 13b) General procedure E with 1.04 g (0.79 mmol) of compound 12b in 55 mL of 33% NH4OH(aq)for 3 hours deprotection at 37° C. and then 15 hours deprotection at room temperature. The product compound 13b was afforded as a white solid; yield: 302 mg (82.5%, C, ε280: 13100 M−1cm−1);1H NMR (400 MHz, D2O) δ 7.96 (d, 1H, J=7.2 Hz), 6.36 (t, 1H, J=6.4 Hz), 6.18 (t, 1H, J=6.8 Hz), 4.64 (s, 1H), 4.26 (s, 3H), 2.47-2.32 (m, 2H);31P NMR (162 MHz, D2O) δ −5.04 (brs), −9.23 (brs), −18.71 (brs). Synthesis of 2′-deoxyadenosine-5′-triphosphate FIG.56shows the synthesis scheme for 2′-deoxyadenosine-5′-triphosphate (dATP, compound 13c).FIG.57throughFIG.67provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. 3′-O, N6, N6-Tribenzoyl-2′-deoxyadenosine-5′-dibenzylmonophosphate (Compound 9c) General procedure A with 1.03 g (1.83 mmol) of 3′-O,N6,N6-tribenzoyl-2′-deoxyadenosine compound 8c, 233 mg (3.29 mmol) of tetrazole, 12 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 0.75 mL (2.38 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 1 hour stirring at room temperature. Then, the reaction was added to 6 mL of H2O2for 1 hour oxidation reaction. Column chromatography with eluents (EtOAc/hexane, from 33% to 66%) to afford the product compound 9c as a white foam; yield: 1.33 g (88.3%); TLC (EtOAc/hexane. 1:60) Rf=0.32;1H NMR (400 MHz, CDCl3) δ 8.63 (s, 1H), 8.38 (s, 1H), 8.06 (d, 2H, J=8.0 Hz), 7.87 (d, 3H, J=8.0 Hz), 7.64-7.61 (m, 1H), 7.50-7.46 (m, 4H), 7.37-7.28 (m, 15H), 6.60-6.56 (m, 1H), 5.55 (d, 1H, J=5.6 Hz), 5.07-5.01 (m, 4H), 4.40 (s, 1H), 4.34-4.24 (m, 2H), 2.72-2.67 (m, 2H);13CNMR (125.8 MHz, CDCl3) δ 172.7, 166.2, 153.2, 152.6, 152.3, 143.5, 134.5, 134.1, 133.4, 129.9, 129.5, 129.1, 129.1, 129.1, 129.1, 129.1, 129.0, 129.0, 128.6, 128.6, 128.1, 84.9, 83.9 (d, JC, P=10.4 Hz), 75.0, 69.7 (d, JC, P=5.3 Hz), 66.8 (d, JC, P=5.2 Hz), 38.7;31P NMR (162 MHz, CDCl3) (50.70; HRMS (ESI-TOF) calcd. for C45H38N5O9PNa [M+Na]+846.2305; found 846.2291. 3′-O, N6. N6-Tribenzoyl-2′-deoxyadenosine-5′-monophosphate (Compound 10c) General procedure B with 1.33 g (1.62 mmol) of compound 9c, 100 mL of MeOH, and 230 mg of 10% Pd/C for 3 hours stirring. The suspension was filtered over a pad of celite, washed with 100 mL of MeOH containing 2% triethylamine four times to afford the product compound 10c as a white foam of triethylammonium salt; yield: 0.88 g (84.5%); TLC (MeOH/CH2Cl21:10 with 1% triethylamine) Rf=0;1H NMR (400 MHz, CD3OD) δ 8.93 (s, 1H), 8.69 (s, 1H), 8.06 (d, 2H, J=7.6 Hz), 7.80-7.78 (m, 3H), 7.62-7.58 (m, 1H), 7.49-7.47 (m, 4H), 7.37-7.34 (m, 3H), 6.71 (t, 1H, J=1.6 Hz), 5.77 (d, 1H, J=5.6 Hz), 4.47 (5, 1H), 4.17-4.13 (m, 2H), 3.14-3.12 (m, 1H), 2.74 (dd, 1H, J=13.6, 5.6 Hz);13C NMR (125.8 MHz, CD3OD) 173.7, 167.2, 154.4, 153.1, 152.6, 146.3, 135.4, 134.5, 134.5, 134.2, 131.0, 130.6, 130.4, 129.8, 129.7, 129.6, 129.4, 128.9, 86.0 (d, JC, P=10.4 Hz), 77.9, 66.2, 55.6, 47.5, 39.1;31P NMR (162 MHz, CD3OD) δ 2.30; HRMS (ESI-TOF) calcd. for C31H25N5O9PNa2[M−H+2Na]+688.1185. found 688.1204. 3′-O, N6, N6-Tribenzoyl-2′-deoxyadenosine-5′-phosphor-2-methylimidazolide (Compound 11c) General procedure C with 0.88 g (1.37 mmol) of compound 10c, 7.15 mL of anhydrous DMF, 1.0 mL (7.19 mmol) of triethylamine, 239 mg (2.85 mmol) of 2-methylimidazole, 760 mg (2.90 mmol) of triphenylphosphine, 642 mg (2.92 mmol) of dipyridyl disulfide for 2 hours reaction at room temperature. The crude was dropwise added to 300 mL of ether for precipitation and the solid was collected by centrifugation at 4400 rpm for 10 minutes at room temperature. The crude product was resuspended with minimal volume of CH2Cl2and dropwise added to 300 mL of ether containing 1.4 g of LiClO4. The precipitate was collected by centrifugation at 4400 rpm for 10 minutes at room temperature to afford the white solid compound 11c; yield: 1.08 g (97.5%);31P NMR (162 MHz, D2O) δ 7.59; HRMS (ESI-TOF) calcd. for C35H30N7O8PNa [M+Na]+730.1791. found 730.1799. 3′-O, N6, N6-Tribenzoyl-2′-deoxyadenosine-5′-γ-(2-(pyrenesulfonyl)ethyl]-triphosphate (Compound 12c) General procedure D with 1.08 g (1.34 mmol) of compound 11c, 0.77 g (1.63 mmol) of 1-(2-(pyrenesulfonyl)ethyl)-pyrophosphate (compound 7) and 13.6 mL (13.6 mmol) of ZnCl2solution (1.0 M in anhydrous DMF) for 3 hours with stirring. After the reaction, the solution was precipitated by 300 mL of ether. After centrifugation, the pellet was resuspended by 20% H2O/MeCN containing 2% Hunig's base and filtered by pyrex glass funnel. The filtrate was evaporated and the crude product was purified by silica column chromatography with eluents [(8% H2O/isopropanol+1% diisopropylethylamine), then (H2O/(isopropanol-MeCN 1:1) from 2% to 7% containing 1% diisopropylethylamine (DIPEA)] to afford the white solid compound 12c; yield: 928 mg (46.7%); TLC (H2O/acetone 1:10 containing 2% DIPEA): Rf=0.28;1H NMR (600 MHz, D2O/DMSO-d61:1) δ 8.58-8.57 (m, 1H), 8.45 (s, 1H), 8.37-8.33 (m, 2H), 8.06-7.70 (m, 11H), 7.57-7.32 (m, 7H), 5.94 (t, 1H, J=7.2 Hz), 5.31 (s, 1H), 4.20 (d, 2H, J=6.6 Hz), 3.99-3.97 (m, 2H), 3.86-3.84 (m, 1H), 3.75-3.74 (m, 1H), 2.55 (s, 1H), 2.10 (dd, 1H, J=13.8, 4.8 Hz);31P NMR (162 MHz, D2O/DMSO-d61:1) δ −10.24 (d, J=14.9 Hz), −10.45 (d, J=14.7 Hz), −20.67 (brs, 1P); HRMS (ESI-TOF) calcd. for C42H34N5O16P3SNa [M−2H+Na]+1012.0832. found 1012.0827. During HRMS, analysis one of the two benzoyl group is removed from the exocyclic amino group. 2′-deoxyadenosine-5′-triphosphate (Compound 13c) General procedure E with 928 mg (0.63 mmol) of compound 12c in 45 mL of 33% NH4OH(aq)in deprotection step for 3 hours at 37° C. and then 15 hours at room temperature. The product compound 13c was afforded as a white solid; yield: 284 mg (92.6%, A, ε259: 15200 M−1cm−1);1H NMR (400 MHz, D2O) δ 8.37 (s, 1H), 8.08 (s, 1H), 6.39 (t, 1H, J=6.4 Hz), 4.33 (s, 1H), 4.26 (s, 2H), 2.81-2.74 (m, 1H), 2.66-2.60 (m, 1H);31P NMR (162 MHz, D2O) δ −3.97 (d, J=15.7 Hz), −9.22 (d, J=16.0 Hz), −17.85 (t, J=13.3 Hz). Synthesis of 2′-deoxyguanosine-5′-triphosphate FIG.68shows the synthesis scheme for 2′-deoxyguanosine-5′-triphosphate (dGTP, compound 13d).FIG.69through andFIG.79provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. 3′-O, N2-Dibenzoyl-2′-deoxyguanosine-5′-dibenzylmonophosphate (Compound 9d) General procedure A with 1.0 g (2.10 mmol) of 3′-O, N2-dibenzoyl-2′-deoxyguanosine. 0.29 g (4.21 mmol) of tetrazole, 15 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 0.85 mL (2.52 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 1 hour stirring at room temperature. Then, the reaction was added to 6 mL of H2O2for 1 hour oxidation reaction. Column chromatography with eluents (EtOAc/hexane, from 33% to 66%) to afford the product compound 9d as a white foam; yield: 1.35 g (85.9%); TLC (EtOAc/hexane, 1:60) Rf=0.32;1H NMR (400 MHz, CDCl3) δ 12.51 (s, 1H), 11.03 (s, 1H), 8.14 (d, 2H, J=6.0 Hz), 8.06 (d, 2H, J=16.0 Hz), 7.73 (s, 1H), 7.62-7.60 (m, 2H), 7.50-7.45 (m, 4H), 7.33 (m, 5H), 7.26-7.22 (m, 1H), 7.19-7.16 (m, 1H), 7.00 (d, 2H, J=16.0 Hz), 6.25 (dd, 1H, J=12.0, 2.8 Hz), 5.70 (d, 11-1, J=4.4 Hz), 5.08 (d, 2H, J=4.4 Hz), 4.80 (m, 2H), 4.56 (m, 1H), 4.43 (s, 1H), 4.35 (m, 1H), 3.60 (m, 1H), 2.51 (s, 1H);13C NMR (125.8 MHz, CDCl3) δ 169.2, 166.0, 155.7, 148.2, 148.1, 139.5, 135.4, 135.1, 135.0, 133.8, 133.2, 132.1, 129.8, 129.3, 128.9, 128.8, 128.7, 128.6, 128.5, 128.3, 128.2, 128.2, 127.3, 127.3, 123.5, 86.8, 83.2, 75.8, 69.9 (d, JC, P=5.3 Hz), 69.4 (d, JC, P=5.2 Hz), 66.8, 35.5;31P NMR (162 MHz, CDCl3) δ −0.55; HRMS (ESI-TOF) calcd. for C38H34N5O9PNa [M+Na]+758.1992; found 758.2000. 3′-O, N2-Dibenzoyl-2′-deoxyguanosine-5′-monophosphate (Compound 10d) General procedure B with 0.88 g (1.19 mmol) of compound 9d, 10 mL of MeOH, and 250 mg of 10% Pd/C for 3 hours with stirring. The suspension was filtered over a pad of celite, washed with 100 mL of MeOH containing 2% triethylamine four times to afford the product compound 10d as a white foam of triethylammonium salt; yield: 0.72 g (84.5%); TLC (MeOH/CH2Cl21:10 with 1% triethylamine);1H NMR (400 MHz, DMSO-d6) δ 8.28 (s, 1H), 8.18 (d, 2H, J=6.4 Hz), 8.03 (m, 2H), 7.70-7.62 (m, 1H), 7.61-7.59 (m, 1H), 7.57-7.54 (m, 2H), 7.51 (m, 2H), 6.44 (m, 1H), 5.71 (d, 1H, J=3.6 Hz), 4.38 (s, 1H), 4.12 (m, 1H), 4.02 (m, 1H), 3.35 (m, 1H), 2.56 (dd, 1H, J=15.6, 6.8 Hz).13C NMR (125.8 MHz, DMSO-d6) δ 169.8, 165.1, 155.2, 148.8, 139.2, 133.6, 132.8, 132.7, 129.4, 129.3, 128.8, 128.7, 128.2, 128.0, 126.8, 121.1, 84.6, 83.7, 76.8, 63.9, 30.5;31P NMR (162 MHz, CD3OD) δ −0.34; HRMS (ESI-TOF) calcd. for C24H21N5O9PNa2[M−H+2Na]+600.0872. found 600.0865. 3′-O, N2-Dibenzoyl-2′-deoxyguanosine-5′-phosphor-2-methylimidazolide (Compound 11d) General procedure C with 0.65 g (1.17 mmol) of compound 10d, 10 mL of anhydrous DMF, 0.82 mL (7.19 mmol) of triethylamine, 240 mg (5.85 mmol) of 2-methylimidazole, 768.3 mg (2.93 mmol) of triphenylphosphine, 645.5 mg (2.93 mmol) of dipyridyl disulfide for 2 hours reaction at room temperature. First precipitation was achieved with 300 mL of diethyl ether. The product was resuspended with 15 mL of CH2Cl2and dropwise added to the solution containing 1.17 g of sodium perchlorate, 15 mL of triethylamine in 300 mL of ethyl acetate for second precipitation. The product was afforded as a white solid compound 11d; yield: 0.68 g (93.4%);31P NMR (162 MHz, D2O) 6-8.70; HRMS (ESI-TOF) calcd. for C28H25N7O8PNa2[M−H+2Na]+664.1298. found 664.1276. 3′-O, N2-Dibenzoyl-2′-deoxyadenosine-5′-(γ-(2-(pyrenesulfonyl)ethyl))-triphosphate (Compound 12d) General procedure D with 0.5 g (0.81 mmol) of compound 11d, 0.495 g (1.05 mmol) of 1-(2-(pyrenesulfonyl)ethyl)-pyrophosphate (compound 7) and 5.38 mL (8.07 mmol) of ZnCl2solution (1.5 M in anhydrous DMF) for 3 h with stirring. After the reaction, the solution was precipitated by 300 mL of ether. Silica column chromatography with eluents [(H2O in isopropanol/MeCN) 1:1 from 3% to 8% containing 1% diisopropylethylamine (DIPEA)] to afford the white solid compound 12d; yield: 1.07 g (58.1%); TLC (H2O/acetonitrile 1:10 containing 2% DIPEA) Rf=0.34;1H NMR (400 MHz, DMSO-d6) δ 7.90-7.60 (m, 1H), 7.31 (s, 6H), 7.03-6.98 (m, 5H), 6.72 (m, 3H), 6.52 (s, 4H), 5.32 (s, 1H), 4.62 (s, 1H), 3.36 (brs, 2H), 3.16 (brs, 2H), 2.87 (s, 1H), 2.09 (s, 2H);31P NMR (162 MHz, DMSO-d6) δ −11.34, −12.12, −20.44; HRMS (ESI-TOF) calcd. for C42H34N5O17P3SNa [M−2H+Na]−1028.0781. found 1028.0796. 2′-deoxyguanosine-5′-triphosphate (Compound 13d) General procedure E with 800 mg (0.81 mmol) of compound 12d in 45 mL of 33% NH4OH(aq)for 3 hours at 37° C. and then 15 hours deprotection reaction at room temperature. The product compound 13d was afforded as a white solid; yield: 372 mg (91.1%, G, ε253: 13700 M−1cm−1);1H NMR (400 MHz, D2O) δ 8.15 (m, 1H), 6.32 (s, 1H), 4.27 (s, 1H), 4.19 (m, 2H), 4.15 (s, 1H), 2.79 (brs, 1H), 2.52 (s, 1H);31P NMR (162 MHz, D2O) 6-5.80 (d, J=15.7 Hz), −10.86 (d, J=16.0 Hz), −20.59 (t, J=13.3 Hz). Synthesis of 2′-deoxythymidine-3′-triphosphate FIG.80shows the synthesis scheme for 2′-deoxythymidine-3′-triphosphate (3′-TTP, compound 19).FIG.81throughFIG.91provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. 5′-Benzoyl-2′-deoxythymidine-3′-dibenzylmonophosphate (Compound 15) General procedure A with 1.04 g (3.0 mmol) of 5′-benzoyl-2′-deoxythymidine compound 14, 383 mg (5.4 mmol) of tetrazole, 30 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 1.23 mL (3.91 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 3 hours reaction at room temperature. Then 8 mL of 30% H2O2(aq)for 1 hour reaction at room temperature. Column chromatography with eluents (MeOH/CH2Cl2, from 1% to 1.4%) to afford the product compound 15 as a white solid; yield: 1.28 g (70.3%); TLC (MeOH/CH2Cl2, 1:40) Rf=0.22;1H NMR (400 MHz, CDCl3) δ 9.46 (s, 1H), 7.97 (d, 2H, J=8.0 Hz), 7.61-7.57 (m, 1H), 7.45 (t, 2H, J=7.2 Hz), 7.35-7.31 (m, 10H), 7.08 (5, 1H), 6.25 (dd, 1H, J=7.6, 6.4 Hz), 5.11-4.97 (m, 5H), 4.56 (dd, 1H, J=13.2, 3.6 Hz), 4.33-4.29 (m, 2H), 2.48 (dd, 1H, J=14.4, 5.6 Hz), 2.11-2.06 (m, 1H), 1.57 (s, 3H);13C NMR (125.8 MHz, CDCl3) δ 165.7, 163.8, 150.4, 135.3, 135.2, 134.3, 133.5, 129.3, 129.2, 128.7, 128.7, 128.6, 128.6, 128.1, 128.1, 111.4, 84.5, 82.6 (d, JC, P=5.4 Hz), 69.8, 69.8, 69.8, 63.6, 38.6 (d, JC, P=4.7 Hz), 12.0; 31P NMR (162 MHz, CDCl3) δ −0.31; HRMS (ESI-TOF) calcd. for C31H31N2O9PNa [M+Na]+629.1665. found 629.1666. 5′-Benzoyl-2′-deoxythymidine-3′-monophosphate (Compound 16) General procedure B with 1.28 g (2.11 mmol) of compound 15, 50 mL of MeOH, and 200 mg of 10% Pd/C for 3 hours with stirring at room temperature. The suspension was filtered over a pad of celite, washed with 100 mL of MeOH four times to afford the product, compound 16, as a white foam; yield: 0.86 g (96.2%); TLC (MeOH/CH2Cl2, 1:10) Rf=0;1H NMR (400 MHz, CD3OD) δ 8.06 (d, 2H, J=7.2 Hz), 7.64 (t, 1H, J=7.6 Hz), 7.51 (t, 2H, J=7.6 Hz), 7.39 (s, 1H), 6.29 (m, 1H), 5.10 (t, 1H, J=6.8 Hz), 4.70 (dd, 1H, J=12.0, 3.2 Hz), 4.57-4.47 (m, 2H), 2.63 (ddd, 1H, J=14.0, 6.0, 2.4 Hz), 2.43 (m, 1H), 1.62 (s, 3H);13C NMR (125.8 MHz, CD3OD) δ 167.4, 166.1, 152.1, 137.0, 134.6, 130.8, 130.5, 129.8, 111.8, 86.4, 84.6, 77.2, 65.1, 39.5, 12.2;31P NMR (162 MHz, CD3OD) δ 0.42; HRMS (ESI-TOF) calcd. for C17H18N2O9PNa2[M−H+2Na]+471.0545. found 471.0551. 5′-Benzoyl-2′-deoxythymidine-3′-phosphor-2-methylimidazolide (Compound 17) General procedure C with 0.86 g (2.0 mmol) of compound 16, 10 mL of anhydrous DMF, 1.39 mL (10.0 mmol) of triethylamine, 328 mg (4.1 mmol) of 2-methylimidazole, 1.05 g (4.0 mmol) of triphenylphosphine, 903 g (4.0 mmol) of dipyridyl disulfide. First precipitation was achieved in 250 mL of diethyl ether. The product was resuspended with 10 mL of DMF and dropwise added to the solution containing 2.0 g of sodium perchlorate, 20 mL of triethylamine in 400 mL of ethyl acetate for the second precipitation. The product was afforded as a white solid compound 17; yield: 0.89 g (86.9%);31P NMR (162 MHz, D2O) δ −7.57; HRMS (ESI-TOF) calcd. for C21H23N4O8PNa [M+Na]+513.1151. found 513.1158. 5′-Benzoyl-2′-deoxythymidine-3′-γ-(2-(pyrenesulfonyl)ethyl)triphosphate (Compound 18) General procedure D with 0.89 g (1.74 mmol) of compound 16, 0.89 g (1.90 mmol) of 1-(2-(pyrenesulfonyl)ethyl)) pyrophosphate (compound 7) and 11.5 mL (17.30 mmol) of ZnCl2solution (1.5 M in anhydrous DMF) for 3 hours with stirring at room temperature. The product was purified by 2.5 cm silica gel flash column with eluents [H2O-isopropanol from 5% to 10% containing 1% diisopropylethylamine (DIPEA)] to afford the yellowish solid compound 18; yield: 1.23 g (55.8%); TLC (1:10 H2O-acetone/2%-DIPEA): Rf=30.8;1H NMR (400 MHz, D2O) δ 8.25 (d, 1H, J=8.8 Hz), 8.11 (d, 1H, J=6.4 Hz), 7.47 (d, 1H, J=8.0 Hz), 7.36-7.34 (m, 4H), 7.15-7.07 (m, 2H), 6.94-6.79 (m, 5H), 6.26 (s, 1H), 5.12 (s, 1H), 4.85 (s, 1H), 4.48-4.25 (m, 5H), 3.80 (s, 2H), 2.35 (s, 1H), 1.84 (s, 1H), 0.66 (s, 3H);31P NMR (162 MHz, D2O) δ −10.56 (d, 1P, J=12.5 Hz), −11.07 (d, 1P, J=14.1 Hz), −20.85 (brs, 1P); HRMS (ESI-TOF) calcd. for C35H30N2O17P3SNa4[M−3H+4Na]+967.0069. found 967.0080. 2′-deoxythymidine-3′-triphosphate (Compound 19) General procedure E with 1.23 g of compound 18, 5 mL of CH2Cl2and 50 mL of 33% NH4OH(aq)for 18 hours deprotection at room temperature. The product, compound 19, was afforded as a white solid; yield: 404 mg (86%, ε267=9600 M−1cm−1);1H NMR (400 MHz, D2O) δ 7.74 (s, 1H), 6.40 (t, 1H, J=7.6 Hz), 5.05 (5, 1H), 4.34 (d, 1H, J=2.8 Hz), 3.92 (d, 2H, J=3.6 Hz), 2.70-2.65 (m, 1H), 2.54-2.47 (m, 1H), 1.96 (s, 3H);31P NMR (162 MHz, D2O) δ −4.03 (d, 1P, J=17.0 Hz), −9.99 (d, 1P, J=16.7 Hz), −18.47 (brs, 1P). Synthesis of 1-(α-L-threofuranosyl)thymidine-3′-triphosphate FIG.92shows the synthesis scheme for 1-(α-L-threofuranosyl)thymidine-3′-triphosphate (tTTP, compound 25a).FIG.93throughFIG.100provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. 1-(2′-O-benzoyl-α-L-threofuranosyl)thymidine-3′-dibenzylmonophosphate (Compound 21a) General procedure A with 1 g (3.01 mmol) of 1-(2′-O-benzoyl-α-L-threofuranosyl)thymine, 379.7 mg (5.42 mmol) of tetrazole, 24 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 1.24 mL (3.91 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 3 hours stirring at room temperature and 6 mL of H2O2for 1 hour oxidation reaction. Silica gel column chromatography with eluents (MeOH/CH2Cl2, from 0 to 2%). The product, compound 21a, was acquired as a white solid; yield: 1.25 g (70.1%). Compound 21a has been reported in Sau et al., 2017, Org. Lett. 19, 4379-4382. 1-(2′-O-benzoyl-α-L-threofuranosyl)thymidine-3′-monophosphate (Compound 22a) General procedure B with 1.25 g (2.11 mmol) of compound 21a, 50 mL of MeOH, and 200 mg of 10% Pd/C for 3 hours stirring. The suspension was filtered over a pad of celite, washed with 100 mL of MeOH four times. The product compound 22a was afforded as a white foam; yield: 0.83 g (92.4%); TLC (MeOH/CH2Cl2, 1:10) Rf=0:1H NMR (400 MHz, CD3OD) δ 8.05 (d, 1H, J=7.6 Hz), 7.64 (t, 1H, J=7.2 Hz), 7.57 (s, 1H), 7.50 (t, 2H, J=7.2 Hz), 6.05 (s, 1H), 5.60 (s, 1H), 5.01 (s, 1H), 4.52 (d, 1H, J=10.4 Hz), 4.29 (d, 1H, J=9.2 Hz), 1.92 (s, 3H);13C NMR (125.8 MHz, CD3OD) δ 166.3, 152.2, 138.0, 134.9, 130.9, 130.1, 129.7, 111.7, 91.2, 82.1 (d, JC, P=5.9 Hz), 79.0, 74.7, 12.5;31P NMR (162 MHz, CDCl3) δ −0.23; HRMS (ESI-TOF) calcd. for C16H17N2O9PNa [M+Na]+435.0569. found 435.0557. 1-(2′-O-benzoyl-α-L-threofuranosyl)thymidine-3′-monophosphor-2-methylimidazolide (Compound 23a) General procedure C with 0.83 g (2 mmol) of compound 22a, 7.0 mL of anhydrous DMF, 1.40 mL (10.1 mmol) of triethylamine, 328 mg (4 mmol) of 2 methylimidazole, 1.05 g (4 mmol) of triphenylphosphine, 0.88 g (4 mmol) of dipyridyl disulfide. First precipitation was achieved with 250 mL of diethyl ether. The product was resuspended with 15 mL of CH2Cl2and dropwise added to the solution containing 2.01 g of sodium perchlorate, 10 mL of triethylamine in 300 mL of ethyl acetate for the second precipitation. The product, compound 23a, was afforded as a white solid; yield: 0.98 g (98.3%);31P NMR (162 MHz, D2O) δ −8.17; HRMS (ESI-TOF) calcd. for C20H21N4O8PNa [M+Na]+499.0995. found 499.0992. 1-(2′-O-benzoyl-α-L-threofuranosyl)thymidine-3′-(γ-(2-(pyrenesulfonyl)ethyl)) Triphosphate (Compound 24a) General procedure D with 0.98 g (1.97 mmol) of compound 23a, 1.11 g (2.36 mmol) of compound 7 and 13.13 mL (19.7 mmol, 1.5 M in anhydrous DMF) of ZnCl2solution for 3 hours stirring at room temperature. The product was purified by 2 cm silica gel column chromatography with eluents [H2O-isopropanol from 5% to 12.5% containing 1% diisopropylethylamine (DIPEA)] to afford the white solid compound 24a; yield: 1.53 g (62.1%); TLC (1:10 H2O-acetone with 2%-DIPEA): Rf=0.25;1H NMR (400 MHz, D2O) δ 8.22 (d, 1H, J=9.2 Hz), 8.09 (d, 1H, J=8.0 Hz), 7.48-7.30 (m, 7H), 7.09-6.98 (m, 4H), 6.84 (t, 2H, J=6.8 Hz), 5.32 (s, 1H), 5.20 (s, 1H), 4.89 (s, 1H), 4.48-4.40 (m, 3H), 3.97 (d, 1H, J=8.0 Hz), 3.76 (m, 2H), 1.50 (s, 3H);31P NMR (162 MHz, D2O) δ −10.23 (d, J=13.0 Hz), −11.65 (d, J=13.9 Hz), 20.59 (brs); HRMS (ESI-TOF) calcd. for C34H29N2O17P3SNa3[M−2H+3Na]+931.0093. found 931.0049. 1-(α-L-threofuranosyl)thymidine-3′-triphosphate (Compound 25a) General procedure E with 1.53 g (1.22 mmol) of compound 24a in 50 mL of 33% NH4OH(aq)for 18 hours deprotection at room temperature. The product, compound 25a, was afforded as a white solid; yield: 445 mg (78.9%, ε267=9600 M−1cm−1);1H NMR (400 MHz, D2O) δ 7.62 (s, 1H), 5.85 (s, 1H), 4.90 (s, 1H), 4.61 (s, 1H), 4.51 (d, 1H, J=10.4 Hz), 4.40 (d, 1H, J=9.2 Hz), 1.95 (s, 1H);31P NMR (162 MHz, D2O) δ −4.89 (d, J=14.7 Hz), −11.02 (d, J=18.63 Hz), −19.80 (brs). Synthesis of 1-(α-L-threofuranosyl)cytidine-3′-triphosphate FIG.101shows the synthesis scheme for 1-(α-L-threofuranosyl)cytidine-3′-triphosphate (tCTP, compound 25b).FIG.102throughFIG.109provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. N4-Benzoyl-1-(2′-O-benzoyl-α-L-threofuranosyl)cytidine-3′-dibenzylmonophosphate (Compound 21 b) Modified general procedure A with 1 g (2.37 mmol) of N4-benzoyl-1-(2′-O-benzoyl-α-L-threofuranosyl)cytosine (compound 20b), 300 mg (4.27 mmol) of tetrazole, 15.8 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 0.98 mL (3.09 mmol) of dibenzyl-N N-diisopropylphosphoramidite for 1 hour reaction at room temperature and 5 mL of H2O2for 1 hour oxidation reaction. After the reaction, the product, compound 21b, was afforded as a white solid; yield: 1.29 g (79.3%). N4-Benzoyl-1-(2′-O-benzoyl-α-L-threofuranosyl)cytidine-3′-monophosphate (Compound 22b) General procedure B with 1.29 g (1.89 mmol) of compound 21b, 50 mL of MeOH, and 250 mg of 10% Pd/C for 5 hours stirring. The suspension was filtered over a pad of celite, washed with 100 mL of MeOH containing 2% triethylamine four times. The product, compound 22b, was afforded as a white foam of a triethylammonium salt; yield 1.23 g (92.8%);1H NMR (400 MHz, CDCl3) δ 8.26 (d, 1H, J=7.6 Hz), 7.99 (d, 2H, J=7.2 Hz), 7.90 (d, 2H, J=7.6 Hz), 7.57-7.53 (m, 2H), 7.47-7.39 (m, 5H), 6.18 (s, 1H), 5.66 (s, 1H), 4.91-4.85 (m, 2H), 4.28 (dd, 1H, J=10.4, 3.2 Hz);13C NMR (125.8 MHz, CDCl3) δ 164.6, 162.7, 154.9, 146.2, 133.5, 132.9, 129.9, 129.3, 128.9, 128.9, 128.5, 128.0, 128.0, 96.6, 91.4, 80.9 (d, JC, P=6.5 Hz), 76.4 (d, JC, P=4.7 Hz), 57.9;31P NMR (162 MHz, CDCl3) δ −1.15; HRMS (ESI-TOF) calcd. for C20H20N3O9PNa3[M−2H+3Na]+546.0630. found 546.0640. N4Benzoyl-1-(2′-O-benzoyl-α-L-threofuranosyl)cytidine-3′-monophosphor-2-methylimidazolide (Compound 23b) General procedure C with 1.02 g (1.75 mmol) of compound 22b, 4.3 mL of anhydrous DMF, 0.87 mL (6.4 mmol) of triethylamine, 287 mg (3.5 mmol) of 2-methylimidazole, 0.94 g (3.6 mmol) of triphenylphosphine, 0.79 g (3.6 mmol) of dipyridyl disulfide for 2 hours stirring at room temperature. First precipitation was achieved with 200 mL of diethyl ether. The product was resuspended with 15 mL of CH2Cl2and dropwise added to the solution containing 2.04 g of sodium perchlorate, 10 mL of triethylamine in 300 mL of ethyl acetate for the second precipitation. The product, compound 23b, was afforded as a white solid; yield: 0.94 g (91.5%);31P NMR (162 MHz, D2O) δ −7.56; HRMS (ESI-TOF) calcd. for C26H24N5O8PNa [M+Na]+588.1260. found 588.1246. N4-Benzoyl-1-(2′-O-benzoyl-α-L-threofuranosyl)cytidine-3′-(γ-(2-(pyrenesulfonyl)ethyl) triphosphate (Compound 24b) General procedure D with 0.94 g (1.6 mmol) of compound 23b, 0.9 g (1.92 mmol) of compound 7 and 10.67 mL (16 mmol, 1.5 M in anhydrous DMF) of ZnCl2solution for 5 hours stirring at room temperature. The product was purified by silica gel chromatography with eluents [(6% H2O/isopropanol containing 1% DIPEA) and then (5% to 10% of H2O/(isopropanol-MeCN 1:1)) containing 1% diisopropylethylamine (DIPEA) to afford the white solid compound 24b; yield: 1.43 g (66.7%); TLC (1:10 H2O-acetone with 2%-DIPEA) Rf=0.25;1H NMR (400 MHz, D2O) (S 8.18 (s, 1H), 8.04 (s, 1H), 7.63-7.25 (m, 13H), 7.14 (s, 3H), 6.95-6.91 (m, 4H), 6.64 (s, 1H), 5.57 (s, 1H), 5.41 (s, 1H), 4.94 (s, 1H), 4.63 (s, 1H), 4.44 (s, 2H), 4.23 (s, 1H), 3.74-3.63 (m, 2H);31P NMR (162 MHz, D2O) δ −10.23 (d, J=13.0 Hz), −11.65 (d, J=13.9 Hz), −20.59 (brs); HRMS (ESI-TOF) calcd. for C40H32N3O17P3SNa [M+Na]+974.0563. found 974.0620. 1-(α-L-threofuranosyl)cytidine-3′-triphosphate (Compound 25b) General procedure E with 1.43 g (1.07 mmol) of compound 24b in 50 mL of 33% NH4OH(aq)for 3 hours at 37° C. and then 15 hours deprotection reaction at room temperature. The product, compound 25b, was afforded as a white solid; yield: 412 mg (85.8%, C280=13100 M−1cm−1);1H NMR (400 MHz, D2O) δ 7.82 (d, 1H, J=7.6 Hz), 6.09 (d, 1H, J=7.6 Hz), 5.90 (s, 1H), 4.91 (d, 1H, J=4.8 Hz), 4.58-4.53 (m, 2H), 4.42 (d, 1H, J=6.8 Hz);31P NMR (162 MHz, D2O) δ −4.26 (brs), −10.93 (d, J=17.3 Hz), −19.15 (brs). Synthesis of 9-(α-L-threofuranosyl)adenosine-3′-triphosphate FIG.110shows the synthesis scheme for 9-(α-L-threofuranosyl)adenosine-3′-triphosphate (tATP, compound 25c).FIG.111throughFIG.118provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. N6-Benzoyl-9-(2′-O-benzoyl-α-L-threofuranosyl)adenosine-3′-dibenzyl Monophosphate (Compound 210 General procedure A with 1.007 g (2.26 mmol) of N6-benzoyl-9-(2′-O-benzoyl-α-L-threofuranosyl)adenine (compound 20c), 316 mg (4.52 mmol) of tetrazole, 15 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 0.91 mL (2.71 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 3 hours reaction at room temperature and 5 mL of H2O2for 1 hour oxidation reaction. The product, compound 21c, was afford as a white solid; yield: 1.41 g (88.4%). N6-Benzoyl-9-(2′-O-benzoyl-α-L-threofuranosyl)adenosine-3′-monophosphate (Compound 22c) General procedure B with 1.3 g (1.84 mmol) of compound 21c, 10 mL of MeOH, and 400 mg of 10% Pd/C for 12 hours with stirring. The suspension was filtered over a pad of celite, washed with 100 mL of MeOH four times. The product, compound 22c, was afford as a white solid; yield: 943 mg (97.4%);1H NMR (400 MHz, CD3OD): δ 8.98 (s, 1H), 8.91 (s, 1H), 8.26-8.17 (m, 4H), 7.83-7.80 (m, 2H), 7.74-7.65 (m, 4H), 6.68 (s, 1H), 6.18 (s, 1H), 5.33 (s, 1H), 4.88-4.85 (m, 1H), 4.66 (dd, 1H, J=14, 6.4 Hz).13C NMR (125.8 MHz, DMSO-d6): δ 164.7, 151.9, 150.4, 142.8, 133.9, 133.3, 132.4, 129.6, 129.0, 128.7, 128.4, 128.4, 125.3, 109.4, 87.0, 80.3, 76.2, 72.6;31P NMR (162 MHz, DMSO-d6): 0.30; HRMS (ESI-TOF) calcd. for C23H21N5O8P [M+H]+526.1128. found 526.1118. N6—Benzoyl-9-(2′-O-benzoyl-α-L-threofuranosyl)adenosine-3′-monophosphor-2-methylimidazolide (Compound 23c) General procedure C with 0.93 g (1.58 mmol) of compound 22c, 12 mL of anhydrous DMF, 1.1 mL (7.89 mmol) of triethylamine, 284 mg (3.94 mmol) of 2 methylimidazole, 1.03 g (3.94 mmol) of triphenylphosphine, 0.87 g (3.95 mmol) of dipyridyl disulfide. First precipitation was achieved with 150 mL of diethyl ether. The product was resuspended with 5 mL of CH2Cl2and dropwise added to the solution containing 1.5 g of sodium perchlorate, 6 mL of triethylamine in 150 mL of ethyl acetate for the second precipitation. The product afford as a white solid compound 23c; yield: 0.89 g (93.02%);31P NMR (162 MHz, DMSO-d6): −9.80; HRMS (ESI-TOF) calcd. for C27H24N7O7PNa [M+Na]+612.1373. found 612.1345. N6-Benzoyl-9-(2′-O-benzoyl-α-L-threofuranosyl)adenosine-3′-γ-[2-(pyrenesulphonyl)ethyl]triphosphate (Compound 2401 General procedure D with 0.6 g (1.02 mmol) of compound 23c, 0.52 g (mmol) of compound 7 and 7 mL (10.17 mmol, 1.5 M in anhydrous DMF) of ZnCl2solution for 3 hours stirring at room temperature. The product was purified by silica gel chromatography with eluents [MeOH—CHCl3from 5% to 12% containing 1% diisopropylethylamine (DIPEA)] to afford the white solid compound 24c; yield 0.64 g (63.1%); TLC (1:10 H2O-acetone with 2%-DIPEA) Rf=0.31;1H NMR (400 MHz, D2O) δ 9.50 (s, 1H), 9.18-7.56 (m, 7H), 7.51-7.46 (m, 6H), 7.35-7.09 (m, 9H), 6.19 (m, 1H), 5.72 (m, 1H), 4.72-4.52 (m, 1H), 3.48 (s, 2H), 2.99 (d, 2H, J=12 Hz);31P NMR (162 MHz, D2O) δ −9.21 (d, J=12.9 Hz), −10.51 (d, J=13.0 Hz), −19.24 (brs); HRMS (ESI-TOF) calcd. for C41H32N5O16P3SNa [M−2H+Na]+998.0676. found 998.0667. 9-(α-L-threofuranosyl)adenosine-3′-triphosphate (Compound 25c) General procedure E with 0.5 g (0.51 mmol) of compound 24c, 35 mL of 33% NH4OH(aq)for 3 hours deprotection at 37° C., and then 15 hours deprotection reaction at room temperature. The product compound 25c was afforded as a white solid; yield: 0.21 g (86.2%, ε280=15200 M−1cm−1);1H NMR (400 MHz, D2O) δ 8.36 (s, 1H), 8.27 (s, 1H), 6.16 (s, 1H), 5.09 (d, 1H, J=8.0 Hz), 4.96 (s, 1H), 4.50 (s, 2H);31P NMR (162 MHz, D2O) δ −4.02 (d, J=16.6 Hz), −10.51 (d, J=16.2 Hz), −17.77 (brs). Synthesis of 9-(α-L-threofuranosyl)guanosine-3′-triphosphate FIG.119shows the synthesis scheme for 9-(α-L-threofuranosyl)guanosine-3′-triphosphate (tGTP, compound 25d).FIG.120throughFIG.127provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. N2-Acetyl-9-(2′-O-benzoyl-α-L-threofuranosyl)guanosine-3′-dibenzylmonophosphate (Compound 21d) General procedure A with 1 g (1.68 mmol) of N2-acetyl-9-(2′-O-benzoyl-α-L-threofuranosyl)guanine (compound 20d), 212 mg (3.03 mmol) of tetrazole, 12 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 735 mL (2.19 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 3 hours reaction at room temperature and 6 mL of H2O, for 1 hour oxidation. Column chromatography with eluents (EtOAc/Hexane, 50%; then EtOAc/CH2Cl2, from 16% to 25%) to afford the product compound 21d as a white solid; yield: 1.14 g (79.4%). TLC (EtOAc/CH2Cl2, 1:4) Rf=0.44; Compound 20d has been reported in Sau et al., 2016, J. Org. Chem. 81, 2302-2307. Compound 21d has been reported in Sau et al., 2017, Org. Lett. 19, 4379-4382. N2-Acetyl-9-(2′-O-benzoyl-α-L-threofuranosyl)guanosine-3′-monophosphate (Compound 22d) General procedure B with 1.63 g (2.47 mmol) of compound 21d, 15 mL of MeOH, and 600 mg of 10% Pd/C for 3 hours stirring. The suspension was filtered over a pad of celite, washed with 100 mL of MeOH four times. The product compound 22d was afford as a white solid; yield; 1.04 g (87.7%).1H NMR (400 MHz, DMSO-d6): δ 12.1 (s, 1H), 11.68 (s, 1H), 8.13 (s, 1H), 8.04-8.02 (m, 2H), 7.74-7.70 (m, 1H), 7.59-7.55 (m, 2H), 6.11 (d, 1H, J=4 Hz), 5.86 (s, 1H), 5.09 (d, 1H, J=2.4 Hz) 4.47-4.45 (m, 1H), 4.38-4.34 (m, 1H), 2.15 (s, 3H);13C NMR (125.8 MHz, DMSO-d6): 173.5, 164.5, 154.8, 148.4, 148.1, 137.7, 134.1, 129.6, 128.9, 128.3, 119.9, 86.9, 80.6, 76.5, 72.8, 48.6, 23.8;31P NMR (162 MHz, DMSO-d6): −0.89; HRMS (ESI-TOF) calcd. for C18H18N5O9PNa [M+Na]+502.0740. found 502.0727. N2-Acetyl-9-(2′-O-benzoyl-α-L-threofuranosyl)guanosine-3′-monophosphor-2-methylimidazolide (Compound 23d) General procedure C with 0.94 g (1.97 mmol) of compound 22d, 12 mL of anhydrous DMF, 1.3 mL (9.86 mmol) of triethylamine, 355 mg (4.93 mmol) of 2-methylimidazole, 1.29 g (4.93 mmol) of triphenylphosphine, 1.086 g (4.93 mmol) of dipyridyl disulfide. First precipitation was achieved with 150 mL of diethyl ether. The product was resuspended with 5 mL of CH2Cl2and dropwise added to the solution containing 1.5 g of sodium perchlorate, 6 mL of trimethylamine in 150 mL of ethyl acetate for the second precipitation. The product afford as a white solid compound 23d; yield: 0.88 g (82.1%);31P NMR (162 MHz, DMSO-d6): −9.77; HRMS (ESI-TOF) calcd. for C22H22N7O8PNa [M+Na]+566.1165. found 566.1163. N2-Acetyl-9-(2′-O-benzoyl-α-L-threofuranosyl)guanosine-3′-(γ-(2-(pyrenesulfonyl)ethyl))triphosphate (Compound 24d) General procedure D with 0.5 g (0.92 mmol) of compound 23d, 0.56 g (1.20 mmol) of 7 and 10 mL (9.2 mmol, 1.5 M in anhydrous DMF) of ZnCl2solution for 3 hours stirring at room temperature. The product was purified by silica gel chromatography with eluents [(H2O isopropanol from 5% to 10% then H2O/(isopropanol-MeCN 1:1) containing 1% diisopropylethylamine (DIPEA)] to afford the white solid compound 24d; yield: 0.74 g (80.9%); TLC (1:10 H2O-acetonitrile with 2% DIPEA) Rf=0.31;1H NMR (400 MHz, D2O) δ 12.00 (m, 1H), 8.96 (d, 1H, J=8.4 Hz), 8.65 (d, 1H, J=8.2 Hz), 8.48-8.42 (m, 3H), 8.39-8.30 (m, 1H), 8.28 (s, 1H), 8.19-8.11 (m, 2H), 7.99-7.97 (m, 1H), 7.51 (brs, 2H), 7.25 (s, 2H), 7.13 (s, 2H), 7.01 (s, 1H), 6.01 (s, 1H), 5.85 (s, 1H), 5.09 (s, 1H), 4.63 (s, 1H), 4.19 (s, 2H), 3.90 (d, 2H, J=5.6 Hz), 3.07 (s, 2H), 1.93 (s, 3H);31P NMR (162 MHz, D2O) δ −12.87 (brs, 2P), −20.71 (brs, 1P); HRMS (ESI-TOF) calcd. for C36H30N5O17P3SNa [M−2H+Na]+952.0473. found 952.0466. 9-(α-L-threofuranosyl)guanosine-3′-triphosphate (Compound 25d) General procedure E with 0.55 g of compound 24c, 50 mL of 33% NH4OH(aq)for 3 hours deprotection at 37° C. and then 15 hours at room temperature. The product was afforded as a white solid; yield: 0.24 g (87.2%, ε253=13700 M−1cm−1);1H NMR (400 MHz, D2O) δ 7.84 (m, 1H), 6.06 (d, 1H, J=7.2 Hz), 5.84 (1H), 4.53 (d, 1H, J=8 Hz), 4.41 (m, 1H), 4.31 (s, 2H), 3.35 (m, 2H), 2.96 (m, 1H);31P NMR (162 MHz, D2O) δ −9.75 (d, J=20.4 Hz), −11.27 (d, J=20.4 Hz), −22.16 (d, J=16.2 Hz). Synthesis of L-2′-deoxythymidine-5′-triphosphate FIG.128shows the synthesis scheme for L-2′-deoxythymidine-5′-triphosphate (L-dTTP, compound 31).FIG.129throughFIG.139provide NMR spectra of the synthesized intermediates and product. Methods used for the synthesis are now described. 3′-Benzoyl-2′-deoxy-L-thymidine-5′-dibenzylmonophosphate (Compound 27) General procedure A with 200 mg (0.58 mmol) of compound 26, 72.9 mg (1.18 mmol) of tetrazole, 12 mL of anhydrous solution (MeCN/CH2Cl2, 1:1), 260 μL (0.75 mmol) of dibenzyl-N, N-diisopropylphosphoramidite for 3 hours reaction at room temperature. Then 2 mL of 30% H2O2(aq)for 1 hour oxidation reaction at room temperature. Column chromatography with eluents (MeOH/CH2Cl2, from 1% to 1.4%) to afford the product compound 27 as a white solid; yield: 0.32 g (68.2%); TLC (MeOH/CH2Cl2, 1:40) Rf=0.23;1H NMR (400 MHz, CD3OD) δ 7.96 (d, 2H, J=7.6 Hz), 7.55-7.52 (m, 1H), 7.41-7.38 (m, 3H), 7.31-7.25 (m, 10H), 6.30 (dd, 1H, J=14.4, 2.4 Hz), 5.35 (d, 1H, J=6.8 Hz), 5.06-5.03 (m, 4H), 4.31-4.28 (m, 2H), 4.24 (brs, 1H), 2.42 (dd, J=19.2, 8.4 Hz), 2.21-2.14 (m, 1H), 1.72 (s, 3H);13CNMR (125.8 MHz, CD3OD) δ 167.9, 166.7, 152.9, 137.8, 137.7, 137.7, 137.7, 137.6, 135.4, 131.5, 131.3, 130.6, 130.5, 130.5, 130.0, 130.0, 112.9, 87.0, 84.7 (d, JC, P=6.5 Hz), 76.9, 71.8 (t, JC, P=4.5 Hz), 69.4 (d, JC, P=4.6 Hz), 38.6, 13.4 (d, JC, P=2.7 Hz);31P NMR (162 MHz, CD3OD) 0.19; HRMS (ESI-TOF) calcd. for C31H31N2O9PNa [M+Na]+629.1665. found 629.1685. 3′-Benzoyl-2′-deoxy-L-thymidine-5′-monophosphate (Compound 28) General procedure B with 300 mg (0.49 mmol) of compound 27, 15 mL of MeOH, and 80 mg of 10% Pd/C for 3 hours stirring at room temperature. The suspension was filtered over a pad of celite, and washed with 60 mL of MeOH containing 2% triethylamine four times to afford the product compound 28 as a white foam of triethylammonium salt; yield: 190 mg (91.3%); TLC (MeOH/CH2Cl2, 1:10 with 1% triethylamine) Rf=0;1H NMR (400 MHz, CD3OD) δ 8.05-7.89 (m, 3H), 7.64-7.50 (m, 3H), 6.47 (s, 1H), 5.66 (s, 1H), 4.40 (brs, 2H), 3.33-3.26 (m, 2H), 2.53 (5, 2H), 1.96 (s, 3H);13C NMR (125.8 MHz, CD3OD) S 168.1, 167.3, 153.4, 138.9, 135.3, 131.8, 131.4, 130.5, 113.2, 86.9, 86.3, 78.8, 67.0, 48.2, 39.3, 13.4;31P NMR (162 MHz, CD3OD) δ 1.02; HRMS (ESI-TOF) calcd. for C10H14N2O8P [M−H]−321.0488. found 321.0493. 3′-Benzoyl-2′-deoxy-L-thymidine-5′-phosphor-2-methylimidazolide (Compound 29) General procedure C with 180 mg (0.42 mmol) of compound 28, 8 mL of anhydrous DMF, 300 μL (2.1 mmol) of triethylamine, 86 mg (1.05 mmol) of 2-methylimidazole, 275 mg (1.05 mmol) of triphenylphosphine, 242 mg (1.05 mmol) of dipyridyl disulfide for 2 hours reaction at room temperature. First precipitation was achieved with 50 mL of diethyl ether. The product was resuspended with 10 mL of DMF and dropwise added to the solution containing 700 mg of sodium perchlorate, 15 mL of triethylamine in 100 mL of ethyl acetate for the second precipitation. The product was afforded as a white solid compound 29; yield: 145 mg (96.9%);31P NMR (162 MHz, CD3OD) δ −7.85; HRMS (ESI-TOF) C21H23N4O8PNa [M+Na]+513.1151. found 513.1146. 3′-Benzoyl-2′-deoxy-L-thymidine-5′-(γ-(2-(pyrenesulfonyl)ethyl))triphosphate (Compound 30) General procedure D with 170 mg (0.44 mmol) of compound 29, 248 mg (0.53 mmol) of 2-(pyrenesulfonyl)ethyl]-pyrophosphate (compound 7) and 3 mL (4.40 mmol) of ZnCl2solution (1.5 M in anhydrous DMF) for 3 hours stirring. After the reaction, the solution was precipitated by 150 mL of ether. Silica column chromatography with eluents (H2O/(isopropanol-MeCN 1:1) from 2% to 7% containing 1% diisopropylethylamine (DIPEA) to afford the white solid compound 30; yield: 150 mg (46.7%); TLC (H2O/isopropanol 1:10 containing 2% DIPEA): Rf=0.35;1H NMR (400 MHz, DMSO-d6+D2O (1:2)) δ: 8.99 (brs, 1H), 8.65 (d, 1H, J=8 Hz), 8.32-8.21 (m, 5H), 8.15-8.09 (m, 2H), 7.98-7.87 (m, 2H), 7.70 (s, 1H), 7.56-7.55 (m, 1H), 7.41 (s, 1H), 6.22 (t, 1H, J=4 Hz), 5.10 (s, 1H), 4.35 (s, 2H), 4.22 (s, 1H), 4.11 (s, 1H), 3.92 (t, 2H, J=4 Hz), 2.33 (s, 1H), 1.88 (s, 1H);31P NMR (162 MHz, DMSO-d6+D2O (1:2)) δ−10.99 (brs, 1P), −11.26 (brs, 1P) −20.94 (brs, 1P); HRMS (ESI-TOF) calcd. for C35H31N2O17P3SNa [M−2H+Na]−899.0459. found 899.0454. “L-2”-deoxythymidine-5′-triphosphate (Compound 31) General procedure E with 20 mg (0.04 mmol) of compound 30.10 mL of 33% NH4OH(aq)for 15 hours stirring at room temperature. The product, compound 31, was afforded as a white solid; yield: 8 mg (83.2%);1H NMR (400 MHz, D2O) δ 7.84 (s, 1H), 6.28 (t, 1H, J=6.0 Hz), 5.17 (s, 1H), 4.54 (s, 1H), 4.15 (d, 1H, J=10.4 Hz), 2.83-2.77 (m, 1H), 2.49 (s, 1H), 2.45 (s, 2H), 2.00 (s, 3H);31P NMR (162 MHz, D2O) δ −4.38 (d, J=16.2 Hz), −10.65 (d, J=16.2 Hz), −19.74 (brs). The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety. While this invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such embodiments and equivalent variations.
149,633
11858954
DETAILED DESCRIPTION OF THE INVENTION The present invention relates to new solid forms of the compound (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid with the formula (Id) below and salts thereof The compound of formula (Id) is a prodrug of (4aR,10aR)-1-Propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol [compound (I)] which is a dual D1/D2 agonist with in vitro data listed in Table 7 of Example 8. The inventors have observed that compound (I) is conjugated in rat and human hepatocytes to sulfate and glucuronide derivatives including compound (Id). The conjugates have shown to be converted to compound (I) by conjugation and de-conjugation in the body. Glucuronide and sulfate derivatives are commonly known to be unstable in the intestine. The derivatives are formed as highly polar and soluble metabolites to facilitate the elimination of compounds from the body and are consequently easily excreted. For example, in bile duct cannulated rats, glucuronide and sulfate conjugates are often found in bile while their de-conjugate (i.e. the parent compound) is found in faeces. The back-conversion of glucuronide and sulfate conjugates in the intestine to the parent compound which is then sometimes subsequently reabsorbed, is known as part of the enterohepatic re-circulation process. As mentioned earlier, oral dosing of phenethyl catecholamines, such as apomorphine, has generally proven unsuccessful due to low bioavailability. Likewise, compound (I) suffers from low oral bioavailability (Liu et al., Bioorganic Med. Chem. (2008), 16: 3438-3444). With this in mind and considering the instability of glucuronide and sulfate conjugates in the gastrointestinal tract, it would not be expected that oral dosing of compounds of the invention can be used to achieve sufficient plasma exposure of the compound. The principle of applying glucuronide derivatives as prodrugs for oral delivery has been explored for retinoic acid (Goswami et al., J. Nutritional Biochem. (2003) 14: 703-709) and for morphine (Stain-Texier et al., Drug Metab. and Disposition (1998) 26 (5): 383-387). Both studies showed very low exposure levels of the parent compounds after oral dosing of the derivatives. Another study suggests the use of budenoside-ß-D-glucuronide as a prodrug for local delivery of budenoside to the large intestine for treatment of Ulcerative Colitis based on poor absorption of the prodrug itself from the intestinal system (Nolen et al., J. Pharm Sci. (1995), 84 (6): 677-681). Nevertheless, surprisingly, it has been observed that oral dosing of compound (Id) which has been identified as a metabolite of compound (I) in rats and minipigs provides a systemic exposure of compound (I) in plasma, suggesting the usefulness of said compound as an orally active prodrug of compound (I). The plasma profile of compound (I) resulting from oral dosing of compounds (Ia) and (Ib) and compound (Id) to Wistar rats according to Example 9 are shown inFIG.1. For all the compounds, the doses were corrected by molecular weight to equal a dose of 300 μg/kg of compound (Ib) corresponding to 287 μg/kg of compound (I). The inventors have found that oral dosing of compounds (Ia) and (Ib) to Wistar rats results in early and high peak concentrations of compound (I). Such high peak concentrations are in humans likely to be associated with dopaminergic side effects such as for example nausea, vomiting and light headedness. In contrast, dosing of the compound (Id), results in a slower absorption rate avoiding rapid peak concentrations accompanied by a sustained exposure of compound (I) in plasma. Additionally, the plasma exposure of compound (I) in Wistar rats is maintained throughout 24 hours although the obtained AUC of compound (I) is generally lower than the AUC obtained after dosing of compound (Ib). However, since the peak concentrations of compound (I) which are expected to drive the side effects are lower, higher doses might be administered of the compound (Id) to potentially achieve higher overall plasma concentrations of compound (I) compared to what is achievable from dosing compounds (Ia) and (Ib). When investigating PK properties of compound (Ic), the inventors found that the plasma concentrations of compound (I) were extremely low, leaving compound (Ic) unsuitable as a prodrug of compound (I) for oral administration and confirming that the oral bioavailability demonstrated for the compound of formula (Id) was highly unpredictable. PK parameters for the PK studies in Wistar rats are listed in Table 8 of Example 9. In vivo conversion of compound (Id) to compound (I) has also been observed by after oral dosing of compound (Id) in minipigs. Bioconversion of compound (Id) in human is supported by the Experiments of Example 6 indicating conversion to the compound of formula (I) in rat and human hepatocytes and in rat and human blood (FIGS.6and7). Thus, in conclusion, the compound of formula (Id) is useful as an orally active prodrug of compound (I) and has been observed in rats to provide a PK profile avoiding the peak Cmaxobserved for the known prodrugs (Ia) and (Ib) and providing a significantly higher AUC of compound (I) than compound (Ic). Compound (Id) has further been explored in the rat locomotor activity assay according to Example 10. The assay demonstrated a dopaminergic effect obtained after oral administration of compound (Id) c.f.FIGS.2,3and4. The fact that the compound of formula (Id) possesses no in vitro dopaminergic activity c.f. Example 7 and Table 3, further indicates that the effect of compound (Id) in the rat locomotor activity assay is obtained by conversion of compound (Id) to compound (I). Finally, an important issue associated with the prior art compound (Ib) is that this compound is an agonist of the 5-HT2B receptor. Since 5-HT2B receptor agonists have been linked to pathogenesis of valvular heart disease (VHD) after long term exposure, such compounds are not suitable for use in the treatment of chronical diseases (Rothman et al., Circulation (2000), 102: 2836-2841; and Cavero and Guillon, J. Pharmacol. Toxicol. Methods (2014), 69: 150-161). Thus, a further advantage of the compounds of the invention is that these are not 5-HT2B agonists c.f. Example 8 and Table 7. The compound of formula (Id) is useful in the treatment of neurodegenerative diseases and disorders such as Parkinson's disease and/or other conditions for which treatment with a dopamine agonist is therapeutically beneficial. The compound, being suitable for oral administration has the potential of providing a new treatment paradigm in Parkinson's Disease. WO2019101917 discloses compound (Id), methods for producing the compound (Id) and uses of the compound (Id). The present invention provides new solid forms of compound (Id). The compound of formula (Id) has three pKa values which may be leading to different major species of ionization as depicted in the Table 1 below. TABLE 1Various major species of ionization of compound (Id) At physiological pH, the compound exists mainly on zwitterion form. The current invention comprises seven solid forms of the zwitterion which have been identified and characterized. At low pH, acid addition salts can be formed with inorganic and/or organic acids on the nitrogen atom of the compound (Id). The present invention comprises two acid addition salts which have been identified and characterized. These are the hydrochloride salt and the hydrobromide salt. At high pH, base addition salts can be formed with inorganic and/or organic bases on the acidic groups of the compound of formula (Id). The present invention comprises two base addition salts which have been identified and characterized. These are the sodium salt and the potassium salt. The scope of the invention encompasses solid forms of compound (Id) selected from solid forms of the zwitterion of compound (Id); alkali metal salts of the compound of formula (Id); and halogen salts of the compound of formula (Id). The solid forms of the invention comprise hydrate and anhydrate forms and various polymorphic forms. Exemplified solid forms encompassed by the invention and method for obtaining said forms are described in brief below. Dihydrate (DH1) of the zwitterion of compound (Id) formed by crystallization at room temperature from a Water:EtOH mixture containing 10-30% vol. water, preferably 15-20%. Anhydrate (AH1) of the zwitterion of compound (Id) obtained by crystallization at room temperature from Water:EtOH mixtures containing 1-5% vol. water, or by crystallization at 37° C. or higher temperature from Water:EtOH mixtures containing 10% vol. water. Heptahydrate (HH) of the zwitterion of compound (Id) formed by crystallization of compound (Id) from water. Forms A, B and C of the zwitterion of compound (Id), which forms are all non-stoichiometric hydrates. Form A was obtained by storage of HH at room temperature at ˜5% RH. Form B was obtained by storage of HH at room temperature at ˜10% RH. Form C was obtained by storage of HH at room temperature at ˜15% RH. Monohydrate (MH1) of the zwitterion of compound (Id). MH1 was obtained by heating of DH1 to 105° C. and subsequent water sorption at ambient conditions. MH1 can also obtained by drying DH1 at room temperature to 0% RH, and subsequent water sorption at ambient conditions. The potassium salt of compound (Id), sodium salt form 1 and sodium salt form 2 of compound (Id) were prepared according to the experimental section herein. Hydrochloride and hydrobromide salts of compound (Id) were prepared according to the experimental section herein. In one specific embodiment, the solid forms as provided by the invention are crystalline forms. In one embodiment, the invention provides solid forms that when analysed with XRPD shows at least one XRPD peak, as shown inFIGS.8-19, or included in Table 2. In one specific embodiment, each of said solid form when analysed with XRPD shows respectively at least 5 or more of the peaks of the 2θ-angles included in Table 2 for each specific form ±0.2° 2θ, such as at least 5 to 10 peaks, e.g. 6, 7, 8, or 9 peaks of the 2θ-angles included in Table 2 for each specific form ±0.2° 2θ, or such as at least 10 to 15 peaks, e.g. such as 11, 12, 13, or 14 peaks of the 2θ-angles included in Table 2±0.2° 2θ for each specific form. In an additional specific embodiment, each of said solid form is characterized respectively by at least 5 or more of the peaks of the 2θ-angles included in Table 2 group (a) for each specific form ±0.2° 2θ, such as at least 5 to 10 peaks, e.g. 6, 7, 8, or 9 peaks of the 2θ-angles included in Table 2 for each specific form ±0.2° 2θ, or such as at least 10 to 15 peaks, e.g. such as 11, 12, 13, or 14 peaks of the 2θ-angles included in Table 2±0.2° 20 for each specific form. In another more specific embodiment, each of said solid form when analysed with XRPD shows respectively at least 5 or more of the peaks of the 2θ-angles included in Table 2 for each specific form ±0.1° 2θ, such as at least 5 to 10 peaks, e.g. 6, 7, 8, or 9 peaks of the 2θ-angles included in Table 2 for each specific form ±0.1° 2θ, or such as at least 10 to 15 peaks, e.g. such as 11, 12, 13, or 14 peaks of the 2θ-angles included in Table 2±0.1° 20 for each specific form. In a further specific embodiment, each of said solid form is characterized respectively by at least 5 or more of the peaks of the 2θ-angles included in Table 2 group (a) for each specific form ±0.2° 2θ, such as at least 5 to 10 peaks, e.g. 6, 7, 8, or 9 peaks of the 2θ-angles included in Table 2 for each specific form ±0.1° 2θ, or such as at least 10 to 15 peaks, e.g. such as 11, 12, 13, or 14 peaks of the 2θ-angles included in Table 2±0.1° 20 for each specific form. In one embodiment, the invention provides solid forms of the present invention with XRPD as shown inFIGS.8-19. In one embodiment, the solid forms of the present invention are in a purified form. The term “purified form” is intended to indicate that the solid form is essentially free of other compounds or other forms of the same compound, as the case may be. In one specific embodiment the solid form of the invention is a purified form of the heptahydrate of the zwitterion of compound (Id), the dihydrate of the zwitterion of compound (Id), or an alkali metal salt of the of compound of formula (Id), preferably a potassium salt of the compound of formula (Id). In an even more specific embodiment of the present invention, the solid form is a purified form of the dihydrate of the zwitterion of compound (Id). Methods for preparation of the exemplified solid forms are given in the Experimental section. EMBODIMENTS OF THE INVENTION In the following, embodiments of the invention are disclosed. The first embodiment is denoted E1, the second embodiment is denoted E2 and so forth: E1. A solid form of the compound of formula (Id) wherein said solid form is selected from:a) a solid form of the zwitterion of compound (Id);b) an alkali metal salt of the compound of formula (Id); andc) a halogen salt of the compound of formula (Id). E2. The solid form according to embodiment 1, wherein said solid form is a) a solid form of the zwitterion of compound (Id). E3. The solid form according to any of embodiments 1-2, wherein said solid form is a dihydrate (DH1) of the zwitterion of compound (Id). E4. The solid form according to embodiment 3, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 10.4, 11.6, 12.3, 13.1, 13.6, 14.3, 15.6, 16.0, 16.8 and 18.5°. E5. The solid form according to any of embodiments 3-4, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 12.3, 13.1, 13.6, 16.0, 16.8, 18.5, 18.9, 19.4, 20.5, 21.4, 23.5, 24.7, 25.4, 26.9 and 28.7°. E6. The solid form according to any of embodiments 3-5, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.8a. E7. The solid form according to any of embodiments 1-2, wherein said solid form is an anhydrate of the zwitterion of compound (Id). E8. The solid form according to any of embodiments 1-2 and 7, wherein said solid form is an anhydrate (AH1) of the zwitterion of compound (Id). E9. The solid form according to embodiment 8, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 8.5, 11.1, 12.4, 12.9, 15.6, 16.7, 18.9, 19.3, 20.0 and 21.2°. E10. The solid form according to any of embodiments 8-9, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 8.5, 12.4, 12.9, 15.6, 16.7, 18.9, 19.3, 20.0, 21.2, 21.5, 22.2, 23.0, 24.2, 27.3 and 28.3°. E11. The solid form according to any of embodiments 8-10, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.9a. E12. The solid form according to any of embodiments 1-2, wherein said solid form is a heptahydrate (HH) of the zwitterion of compound (Id). E13. The solid form according to embodiment 12, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 7.0, 8.6, 10.2, 11.1, 11.9, 13.4, 14.0, 14.5, 17.0 and 17.4°. E14. The solid form according to any of embodiments 12-13, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 7.0, 8.6, 10.2, 11.1, 11.9, 14.0, 17.0, 22.2, 25.9, 27.3, 28.3, 30.8, 34.0, 34.8 and 35.2°. E15. The solid form according to any of embodiments 12-14, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.10a. E16. The solid form according to any of embodiments 1-2, wherein said solid form is form A of the zwitterion of compound (Id). E17. The solid form according to embodiment 16, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 7.6, 9.5, 10.0, 11.2, 12.0, 14.3, 14.6, 15.3, 15.5 and 19.3°. E18. The solid form according to any of embodiments 16-17, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 7.6, 9.5, 10.0, 11.2, 12.0, 14.3, 14.6, 15.3, 15.5, 18.7, 19.3, 23.9, 28.8, 33.7 and 38.7°. E19. The solid form according to any of embodiments 16-18, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.11. E20. The solid form according to any of embodiments 1-2, wherein said solid form is form B of the zwitterion of compound (Id). E21. The solid form according to embodiment 20, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 7.6, 9.0, 10.9, 12.3, 14.3, 15.0, 21.5, 22.1, 22.6 and 23.7°. E22. The solid form according to any of embodiments 20-21, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.12. E23. The solid form according to any of embodiments 1-2, wherein said solid form is form C of the zwitterion of compound (Id). E24. The solid form according to embodiment 23, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5 and 20.6°. E25. The solid form according to any of embodiments 23-24, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5, 20.6, 21.6, 22.9, 23.1, 24.0 and 25.4°. E26. The solid form according to any of embodiments 23-25, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.13. E27. The solid form according to any of embodiments 1-2, wherein said solid form is a monohydrate (MH1) of the zwitterion of compound (Id). E29. The solid form according to embodiment 27, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 9.2, 10.2, 11.8, 12.6, 13.6, 15.7, 16.0, 16.5, 17.5 and 18.1°. E30. The solid form according to any of embodiments 27-28, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 9.2, 10.2, 11.8, 12.6, 13.6, 16.0, 16.5, 17.5, 18.1, 18.7, 19.6, 22.9, 24.7, 25.4 and 26.0°. E31. The solid form according to any of embodiments 27-29, wherein said solid form has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.14. E32. The solid form according to embodiment 1, wherein said solid form is b) an alkali metal salt of the compound of formula (Id). E33. The solid form according to any of embodiments 1 and 32, wherein said salt is a potassium salt of the compound of formula (Id). E34. The solid form according to embodiment 33, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 3.0, 9.0, 12.6, 13.6, 15.0, 17.1, 18.0, 18.4, 18.8 and 19.4°. E35. The solid form according to any of embodiments 33-34, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 3.0, 9.0, 12.6, 13.6, 15.0, 18.0, 19.4, 21.8, 24.7, 27.1, 29.8, 33.3, 35.6, 38.6 and 39.6°. E36. The solid form according to any of embodiments 33-35, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.15. E37. The solid form according to any of embodiments 1 and 36, wherein said salt is a sodium salt of the compound of formula (Id). E38. The solid form according to embodiment 37, wherein said sodium salt is sodium salt form 1 of the compound of formula (Id). E39. The solid form according to any of embodiments 37-38, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 5.9, 8.9, 11.9, 12.8, 13.8, 14.9, 17.7, 18.6, 19.0 and 19.5°. E40. The solid form according to any of embodiments 38-39, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 8.9, 12.8, 13.8, 14.9, 17.7, 18.6, 19.0, 19.5, 21.5, 21.8, 22.2, 22.6, 22.9, 23.4 and 25.1°. E41. The solid form according to any of embodiments 38-40, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.16. E42. The solid form according to embodiment 37, wherein said sodium salt is sodium salt form 2 of the compound of formula (Id). E43. The solid form according to any of embodiments 37 and 42, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 5.6, 8.5, 12.6, 13.6, 14.1, 15.0, 16.7, 17.0, 18.8 and 19.8°. E44. The solid form according to any of embodiments 37 and 42-43, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 5.6, 8.5, 12.6, 13.6, 14.1, 15.0, 17.0, 18.8, 19.8, 21.0, 23.4, 28.5, 34.3, 37.3 and 38.5°. E45. The solid form according to any of embodiments 37 and 42-44, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.17. E46. The solid form according to embodiment 1, wherein said solid form is a halogen salt of the compound of formula (Id). E47. The solid form according to any of embodiments 1 and 46, wherein said salt is a hydrochloride salt of the compound of formula (Id). E48. The solid form according to embodiment 47, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 5.7, 7.3, 10.6, 13.3, 15.3, 15.4, 16.2, 20.1, 22.5 and 23.0°. E49. The solid form according to any of embodiments 47-48, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 5.1, 5.7, 7.3, 10.6, 13.3, 15.3, 15.4, 16.2, 16.7, 18.1, 20.1, 22.5, 23.0, 23.6 and 23.8°. E50. The solid form according to any of embodiments 47-49, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.18. E51. The solid form according to any of embodiments 1 and 46, wherein said salt is a hydrobromide salt of the compound of formula (Id). E52. The solid form according to embodiment 51, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 12.5, 13.9, 14.5, 15.6, 18.6, 18.9, 19.8, 21.3, 22.0 and 22.4°. E53. The solid form according to any of embodiments 51-52, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) showing peaks at the following 2θ-angles: 12.5, 13.9, 14.5, 15.6, 18.6, 18.9, 19.8, 21.3, 22.0, 22.4, 23.3, 24.4, 25.5, 28.2 and 28.9°. E54. The solid form according to any of embodiments 51-53, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) as depicted inFIG.19. E55. The solid form of the compound of formula (Id) according to any of embodiments 1-54, for use in therapy. E56. The solid form of the compound of formula (Id) according to any of embodiments 1-54, for use as a medicament. E57. The solid form of the compound of formula (Id) according to embodiment 56, wherein said medicament is an oral medicament such as a tablet or a capsule for oral administration. E58. A pharmaceutical composition comprising a therapeutically effective amount of the solid form of the compound of formula (Id) according to any of embodiments 1-54, and one or more pharmaceutically acceptable excipients. E59. The pharmaceutical composition according to embodiment 58, wherein said pharmaceutical composition is for oral administration. E60. The pharmaceutical composition according to any of embodiments 58-59, wherein said pharmaceutical composition is an oral pharmaceutical composition. E61. The pharmaceutical composition according to any of embodiments 58-60, wherein said pharmaceutical composition is a solid oral dosage form. E62. The pharmaceutical composition according to any of embodiments 58-61, wherein said pharmaceutical composition is a tablet or a capsule for oral administration. E63. The pharmaceutical composition according to any of embodiments 58-62, wherein said pharmaceutical composition further comprises another agent which is useful in the treatment of a neurodegenerative disease or disorder such as Parkinson's disease. E64. The pharmaceutical composition according to any of embodiments 58-63, wherein said pharmaceutical composition further comprises a compound selected from the group consisting of L-DOPA, droxidopa, foliglurax, a MAO-B inhibitor such as selegiline or rasagiline, a COMT inhibitor such as entacapone or tolcapone, an adenosine 2a antagonist such as istradefylline, an antiglutamatergic agent such as amantadine or memantine, an acetylcholinesterase inhibitor such as rivastigmine, donepezil or galantamine, an antipsychotic agent such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or brexpiprazole; or an antibody targeting alpha-synuclein, Tau or A-beta protein. E65. A solid form of the compound of formula (Id) according to any of embodiments 1-54, for use in the treatment of a neurodegenerative disease or disorder such as Parkinson's Disease, Huntington's disease, Restless leg syndrome or Alzheimer's disease; or a neuropsychiatric disease or disorder such as schizophrenia, attention deficit hyperactivity disorder or drug addiction. E66. The solid form of the compound of formula (Id) according to any of embodiments 1-54, for use in the treatment according to embodiment 65, wherein said neurodegenerative disease or disorder is Parkinson's Disease. E67. The solid form of the compound of formula (Id) according to any of embodiments 1-54, for use in the treatment according to any of embodiments 65-66, wherein said compound is to be used in combination with another agent which is useful in the treatment of a neurodegenerative disease or disorder such as Parkinson's disease. E68. The solid form of the compound of formula (Id) according to any of embodiments 1-54, for use in the treatment according to any of embodiments 66-67, wherein said compound is to be used in combination with a compound selected from the group consisting of L-DOPA, droxidopa, foliglurax, a MAO-B inhibitor such as selegiline or rasagiline, a COMT inhibitor such as entacapone or tolcapone, an adenosine 2a antagonist such as istradefylline, an antiglutamatergic agent such as amantadine or memantine, an acetylcholinesterase inhibitor such as rivastigmine, donepezil or galantamine, an antipsychotic agent such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or brexpiprazole; or in combination with an antibody targeting alpha-synuclein, Tau or A-beta protein. E69. The solid form of the compound of formula (Id) according to any of embodiments 1-54, for use in the treatment according to any of embodiments 66-68, wherein said treatment is performed by oral administration of said compound. E70. The solid form of the compound of formula (Id) according to any of embodiments 1-54, for use in the treatment according to any of embodiments 66-69, wherein said compound is comprised in an oral pharmaceutical composition such as a tablet or a capsule for oral administration. E71. A method for the treatment of a neurodegenerative disease or disorder such as Parkinson's Disease, Huntington's disease, Restless leg syndrome or Alzheimer's disease; or a neuropsychiatric disease or disorder such as schizophrenia, attention deficit hyperactivity disorder or drug addiction; which method comprises the administration of a therapeutically effective amount of solid form of the compound of formula (Id) according to any of embodiments 1-54, to a patient in need thereof. E72. The method according to embodiment 71, wherein said neurodegenerative disease or disorder is Parkinson's Disease. E73. The method according to any of embodiments 71-72, wherein said compound or pharmaceutically acceptable salt thereof according to any of embodiments 1-54, is used in combination with another agent which is useful in the treatment of a neurodegenerative disease or disorder such as Parkinson's disease. E74. The method according to any of embodiments 72-73, wherein said compound or pharmaceutically acceptable salt thereof according to any of embodiments 1-23, is used in combination with a compound selected from the group consisting of L-DOPA, droxidopa, foliglurax, a MAO-B inhibitor such as selegiline or rasagiline, a COMT inhibitor such as entacapone or tolcapone, an adenosine 2a antagonist such as istradefylline, an antiglutamatergic agent such as amantadine or memantine, an acetylcholinesterase inhibitor such as rivastigmine, donepezil or galantamine, an antipsychotic agent such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or brexpiprazole; or in combination with an antibody targeting alpha-synuclein, Tau or A-beta protein. E75. The method according to any of embodiments 71-74, wherein said administration is performed by the oral route. E76. The method according to any of embodiments 71-75, wherein said compound or pharmaceutically acceptable salt thereof according to any of embodiments 1-23 is comprised in an oral pharmaceutical composition such as a tablet or a capsule for oral administration. E77. Use of solid form of the compound of formula (Id) according to any of embodiments 1-54, in the manufacture of a medicament for the treatment of a neurodegenerative disease or disorder such as Parkinson's Disease, Huntington's disease, Restless leg syndrome or Alzheimer's disease; or for the treatment of a neuropsychiatric disease or disorder such as schizophrenia, attention deficit hyperactivity disorder or drug addiction. E78. The use according to embodiment 77, wherein said neurodegenerative disease or disorder is Parkinson's Disease. E79. The use according to any of embodiments 77-78, wherein said medicament is used in combination with another agent which is useful in the treatment of a neurodegenerative disease or disorder such as Parkinson's disease. E80. The use according to any of embodiments 78-79, wherein said medicament is used in combination with a compound selected from the group consisting of L-DOPA, droxidopa, foliglurax a MAO-B inhibitor such as selegiline or rasagiline, a COMT inhibitor such as entacapone or tolcapone, an adenosine 2a antagonist such as istradefylline, an antiglutamatergic agent such as amantadine or memantine, an acetylcholinesterase inhibitor such as rivastigmine, donepezil or galantamine, an antipsychotic agent such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or brexpiprazole; or in combination with an antibody targeting alpha-synuclein, Tau or A-beta protein. E81. The use according to any of embodiments 77-80, wherein said medicament is an oral medicament such as a tablet or a capsule for oral administration. Items The following items serve to further define the invention.Item 1. A solid form of the compound of formula (Id) wherein said solid form is selected from:a) a form of the zwitterion of compound (Id);b) an alkali metal salt of the compound of formula (Id); andc) a halogen salt of the compound of formula (Id).Item 2. The solid form according to item 1, wherein said solid form is a crystalline form.Item 3. The solid form according to any one of items 1-2, wherein said solid form is a solid form of the zwitterion of compound (Id).Item 4. The solid form according to any one of items 1-3, wherein said solid form is a hydrate of the zwitterion of compound (Id).Item 5. The solid form according to any one of items 1-4, wherein said solid form is a hydrate solid form of the zwitterion of compound (Id) selected from the group consisting of a monohydrate form, a dihydrate form and a heptahydrate form.Item 6. The solid form according to any one of items 1-5, wherein said solid form is a hydrate of the zwitterion of compound (Id) selected from the group consisting of the dihydrate form and the heptahydrate form.Item 7. The solid form according to any one of items 1-6, wherein said solid form is the dihydrate (DH1) of the zwitterion of compound (Id).Item 8. The solid form according to item 7, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 10.4, 11.6, 12.3 and 13.1 and 13.6°.Item 9. The solid form according to any one of items 7-8, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 10.4, 11.6, 12.3, 13.1 and 13.6°.Item 10. The solid form according to any one of items 8-9, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 14.3, 15.6, 16.0, 16.8 and 18.5°.Item 11. The solid form according to any one of items 8 and 10, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ:10.4, 11.6, 12.3, 13.1, 13.6, 14.3, 15.6, 16.0, 16.8 and 18.5°.Item 12. The solid form according to any one of items 9-10, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.1° 2θ:10.4, 11.6, 12.3, 13.1, 13.6, 14.3, 15.6, 16.0, 16.8 and 18.5°.Item 13. The solid form according to any one of items 7, 8, 10 and 11, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 10.4, 11.6, 12.3, 13.1, 13.6, 14.3, 15.6, 16.0, 16.8 and 18.5°.Item 14. The solid form according to any one of items 7, 9 and 12, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 10.4, 11.6, 12.3, 13.1, 13.6, 14.3, 15.6, 16.0, 16.8 and 18.5°.Item 15. The solid form according to any one of items 7, 8, 10, 11 and 13, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 12.3, 13.1, 13.6, 16.0, 16.8, 18.5, 18.9, 19.4, 20.5, 21.4, 23.5, 24.7, 25.4, 26.9 and 28.7°.Item 16. The solid form according to any one of items 7,9 and 14, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 12.3, 13.1, 13.6, 16.0, 16.8, 18.5, 18.9, 19.4, 20.5, 21.4, 23.5, 24.7, 25.4, 26.9 and 28.7°Item 17. The solid form according to any one of items 7-16, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.8a.Item 18. The solid form according to any one of items 7-17, exhibiting a weight loss of about 7.6% w/w compared to the initial weight when heated from about 30° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 19. The solid form according to any one of items 7-18, wherein said solid form is a crystal form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.8b.Item 20. The solid form according to any one of items 1-3, wherein said solid form is an anhydrate of the zwitterion of compound (Id).Item 21. The solid form according to any one of items 1-3 and 20, wherein said solid form is the anhydrate (AH1) of the zwitterion of compound (Id).Item 22. The solid form according to any one of items 20-22, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 8.5, 11.1, 12.4, 12.9, and 15.6°.Item 23. The solid form according to any one of items 20-22, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 8.5, 11.1, 12.4, 12.9, and 15.6°.Item 24. The solid form according to any one of items 22-23, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 16.7, 18.9, 19.3, 20.0 and 21.2°.Item 25. The solid form according to any one of items 20-22 and 24, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 8.5, 11.1, 12.4, 12.9, 15.6, 16.7, 18.9, 19.3, 20.0 and 21.2°.Item 26. The solid form according to any one of items 20-25, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 8.5, 11.1, 12.4, 12.9, 15.6, 16.7, 18.9, 19.3, 20.0 and 21.2°.Item 27. The solid form according to any one of items 20-25, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 8.5, 12.4, 12.9, 15.6, 16.7, 18.9, 19.3, 20.0, 21.2, 21.5, 22.2, 23.0, 24.2, 27.3 and 28.3°.Item 28. The solid form according to any one of items 20-27, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 8.5, 12.4, 12.9, 15.6, 16.7, 18.9, 19.3, 20.0, 21.2, 21.5, 22.2, 23.0, 24.2, 27.3 and 28.3°.Item 29. The solid form according to any one of items 20-28, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.9a.Item 30. The solid form according to any one of items 20-29, exhibiting a weight loss of less than 1% w/w compared to the initial weight when heated from about 30° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 31. The solid form according to any one of items 20-30, wherein said solid form is a crystal form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.9b.Item 32. The solid form according to any one of items 1-6, wherein said solid form is a heptahydrate (HH) of the zwitterion of compound (Id).Item 33. The solid form according to item 32, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.0, 8.6, 10.2, 11.1 and 11.9°.Item 34. The solid form according to any one of items 32-33, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.0, 8.6, 10.2, 11.1 and 11.9°.Item 35. The solid form according to any one of items 33-34, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 13.4, 14.0, 14.5, 17.0 and 17.4°.Item 36. The solid form according to any one of items 32-33 and 35, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.0, 8.6, 10.2, 11.1, 11.9, 13.4, 14.0, 14.5, 17.0 and 17.4°.Item 37. The solid form according to any one of items 32-36, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.0, 8.6, 10.2, 11.1, 11.9, 13.4, 14.0, 14.5, 17.0 and 17.4°.Item 38. The solid form according to any one of items 32-33 and 35-36, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.0, 8.6, 10.2, 11.1, 11.9, 14.0, 17.0, 22.2, 25.9, 27.3, 28.3, 30.8, 34.0, 34.8 and 35.2°.Item 39. The solid form according to any one of items 32-38, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.0, 8.6, 10.2, 11.1, 11.9, 14.0, 17.0, 22.2, 25.9, 27.3, 28.3, 30.8, 34.0, 34.8 and 35.2°.Item 40. The solid form according to any one of items 32-39, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.10a.Item 41. The solid form according to any one of items 32-40, exhibiting a weight loss of about 21% w/w compared to the initial weight when heated from about 20° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 42. The solid form according to any one of items 32-41, wherein said solid form is a crystal form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.10b.Item 43. The solid form according to any one of items 1-4, wherein said solid form is form A of the zwitterion of compound (Id).Item 44. The solid form according to item 43, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.6, 9.5, 10.0, 11.2, and 12.0°.Item 45. The solid form according to any one of items 43-44, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.6, 9.5, 10.0, 11.2, and 12.0°.Item 46. The solid form according to any one of items 44-45, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 14.3, 14.6, 15.3, 15.5 and 19.3°.Item 47. The solid form according to any one of items 43-44 and 46, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.6, 9.5, 10.0, 11.2, 12.0, 14.3, 14.6, 15.3, 15.5 and 19.3°.Item 48. The solid form according to any one of items 43-47, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.6, 9.5, 10.0, 11.2, 12.0, 14.3, 14.6, 15.3, 15.5 and 19.3°.Item 49. The solid form according to any one of items 43-44, and 46-47, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.6, 9.5, 10.0, 11.2, 12.0, 14.3, 14.6, 15.3, 15.5, 18.7, 19.3, 23.9, 28.8, 33.7 and 38.7°.Item 50. The solid form according to any one of items 43-49, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.6, 9.5, 10.0, 11.2, 12.0, 14.3, 14.6, 15.3, 15.5, 18.7, 19.3, 23.9, 28.8, 33.7 and 38.7°.Item 51. The solid form according to any one of items 43-50, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.11.Item 52. The solid form according to any one of items 1-4, wherein said solid form is form B of the zwitterion of compound (Id).Item 53. The solid form according to item 52, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.6, 9.0, 10.9, 12.3 and 14.3°.Item 54. The solid form according to any one of items 52-53, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.6, 9.0, 10.9, 12.3 and 14.3°.Item 55. The solid form according to any one of items 53-54, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 15.0, 21.5, 22.1, 22.6 and 23.7°.Item 56. The solid form according to any one of items 52-53 and 55, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.6, 9.0, 10.9, 12.3, 14.3, 15.0, 21.5, 22.1, 22.6 and 23.7°.Item 57. The solid form according to any one of items 52-57, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.12.Item 58. The solid form according to any one of items 1-4, wherein said solid form is form C of the zwitterion of compound (Id).Item 59. The solid form according to item 58, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.5, 8.1, 10.3, 12.6, and 13.5°.Item 60. The solid form according to any one of items 58-59, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.5, 8.1, 10.3, 12.6, and 13.5°.Item 61. The solid form according to any one of items 59-60, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 13.8, 14.9, 17.5, 18.5 and 20.6°.Item 62. The solid form according to any one of items 58-59 and 61, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5 and 20.6°.Item 63. The solid form according to any one of items 58-59 and 61-62, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5 and 20.6°.Item 64. The solid form according to any one of items 58-63, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5 and 20.6°.Item 65. The solid form according to any one of items 58-59 and 61-63, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5, 20.6, 21.6, 22.9, 23.1, 24.0 and 25.4°.Item 66. The solid form according to any one of items 58-65, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5, 20.6, 21.6, 22.9, 23.1, 24.0 and 25.4°.Item 67. The solid form according to any one of items 58-66, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.13.Item 68. The solid form according to any one of items 1-6, wherein said solid form is a monohydrate (MH1) of the zwitterion of compound (Id).Item 69. The solid form according to item 68, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 9.2, 10.2, 11.8, 12.6, 13.6°.Item 70. The solid form according to any one of items 68-69, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 9.2, 10.2, 11.8, 12.6, 13.6°.Item 71. The solid form according to any one of items 69-70, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 15.7, 16.0, 16.5, 17.5 and 18.1°.Item 72. The solid form according to any one of items 68-69 and 71, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 9.2, 10.2, 11.8, 12.6, 13.6, 15.7, 16.0, 16.5, 17.5 and 18.1°.Item 73. The solid form according to any one of items 68-72, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 9.2, 10.2, 11.8, 12.6, 13.6, 15.7, 16.0, 16.5, 17.5 and 18.1°.Item 74. The solid form according to any one of items 68-69 and 71-72, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 9.2, 10.2, 11.8, 12.6, 13.6, 16.0, 16.5, 17.5, 18.1, 18.7, 19.6, 22.9, 24.7, 25.4 and 26.0°.Item 75. The solid form according to any one of items 68-74, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 9.2, 10.2, 11.8, 12.6, 13.6, 16.0, 16.5, 17.5, 18.1, 18.7, 19.6, 22.9, 24.7, 25.4 and 26.0°.Item 76. The solid form according to any one of items 68-75, wherein said solid form is a crystal form characterized by an x-ray powder diffraction pattern as obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.14a.Item 77. The solid form according to any one of items 68-76, exhibiting a weight loss of about 4% w/w compared to the initial weight when heated from about 20° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 78. The solid form according to any one of items 68-77, wherein said solid form is a crystal form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.14b.Item 79. The solid form according to any one of items 1 and 2, wherein said solid form is an alkali metal salt of the compound of formula (Id).Item 80. The solid form according to item 79, wherein said solid form is an alkali metal salt of the compound of formula (Id) selected from the group consisting of a potassium salt and a sodium salt.Item 81. The solid form according to any of items 79-80, wherein said salt is a potassium salt of the compound of formula (Id).Item 82. The solid form according to item 81, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 3.0, 9.0, 12.6, 13.6, and 15.0°.Item 83. The solid form according to any one of items 81-82, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 3.0, 9.0, 12.6, 13.6, and 15.0°.Item 84. The solid form according to any one of items 81-83, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 17.1, 18.0, 18.4, 18.8 and 19.4°.Item 85. The solid form according to any one of items 81-82 and 84, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 3.0, 9.0, 12.6, 13.6, 15.0, 17.1, 18.0, 18.4, 18.8 and 19.4°.Item 86. The solid form according to any one of items 81-85, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 3.0, 9.0, 12.6, 13.6, 15.0, 17.1, 18.0, 18.4, 18.8 and 19.4°.Item 87. The solid form according to any one of items 81-82 and 84-85, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 3.0, 9.0, 12.6, 13.6, 15.0, 18.0, 19.4, 21.8, 24.7, 27.1, 29.8, 33.3, 35.6, 38.6 and 39.6°.Item 88. The solid form according to any one of items 81-87, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 3.0, 9.0, 12.6, 13.6, 15.0, 18.0, 19.4, 21.8, 24.7, 27.1, 29.8, 33.3, 35.6, 38.6 and 39.6°.Item 89. The solid form according to any one of items 81-88, wherein said potassium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.15a.Item 90. The solid form according to any one of items 81-89, exhibiting a weight loss of less than about 1% w/w compared to the initial weight when heated from about 20° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 91. The solid form according to any one of items 81-90, wherein said solid form is a crystal form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.15b.Item 92. The solid form according to any one of items 1, 2, and 79-80, wherein said salt is a sodium salt of the compound of formula (Id).Item 93. The solid form according to item 92, wherein said sodium salt is the sodium salt form 1 of the compound of formula (Id).Item 94. The solid form according to item 93, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 5.9, 8.9, 11.9, 12.8, 13.8°.Item 95. The solid form according to any one of items 93-94, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 5.9, 8.9, 11.9, 12.8, 13.8°.Item 96. The solid form according to any one of items 94-95, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 14.9, 17.7, 18.6, 19.0 and 19.5°.Item 97. The solid form according to any one of items 93-94 and 95-96, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 5.9, 8.9, 11.9, 12.8, 13.8, 14.9, 17.7, 18.6, 19.0 and 19.5°.Item 98. The solid form according to any one of items 93-97, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 5.9, 8.9, 11.9, 12.8, 13.8, 14.9, 17.7, 18.6, 19.0 and 19.5°.Item 99. The solid form according to any one of items 93-94 and 96-97, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 8.9, 12.8, 13.8, 14.9, 17.7, 18.6, 19.0, 19.5, 21.5, 21.8, 22.2, 22.6, 22.9, 23.4 and 25.1°.Item 100. The solid form according to any one of items 93-99, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 8.9, 12.8, 13.8, 14.9, 17.7, 18.6, 19.0, 19.5, 21.5, 21.8, 22.2, 22.6, 22.9, 23.4 and 25.1°.Item 101. The solid form according to any one of items 93-100, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.16a.Item 102. The solid form according to any one of items 93-101, exhibiting a weight loss of about 2% w/w compared to the initial weight when heated from about 20° C. to about 175° C. (heating rate 10° C./min), such as when measured using thermogravimetric analysis.Item 103. The solid form according to any one of items 93-102, wherein said solid form is a crystal form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.16b.Item 104. The solid form according to item 92, wherein said sodium salt is the sodium salt form 2 of the compound of formula (Id).Item 105. The solid form according to item 104, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 5.6, 8.5, 12.6, 13.6, 14.1°.Item 106. The solid form according to any one of items 104-105, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 5.6, 8.5, 12.6, 13.6, 14.1°.Item 107. The solid form according to any one of items 105-106, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 15.0, 16.7, 17.0, 18.8 and 19.8°.Item 108. The solid form according to any one of items 104-105 and 107, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 5.6, 8.5, 12.6, 13.6, 14.1, 15.0, 16.7, 17.0, 18.8 and 19.8°.Item 109. The solid form according to any one of items 104-108, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 5.6, 8.5, 12.6, 13.6, 14.1, 15.0, 16.7, 17.0, 18.8 and 19.8°.Item 110. The solid form according to any one of items 104-105 and 107-108, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 5.6, 8.5, 12.6, 13.6, 14.1, 15.0, 17.0, 18.8, 19.8, 21.0, 23.4, 28.5, 34.3, 37.3 and 38.5°.Item 111. The solid form according to any one of items 104-110, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.1° 2θ: 5.6, 8.5, 12.6, 13.6, 14.1, 15.0, 17.0, 18.8, 19.8, 21.0, 23.4, 28.5, 34.3, 37.3 and 38.5°.Item 112. The solid form according to any one of items 104-111, wherein said sodium salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.17a.Item 113. The solid form according to any one of items 104-112, exhibiting a weight loss of about 5% w/w compared to the initial weight when heated from about 20° C. to about 175° C. (heating rate 10° C./min), such as when measured using thermogravimetric analysis.Item 114. The solid form according to any one of items 104-113, wherein said solid form is a crystal form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.17b.Item 115. The solid form according to any one of items 1-2, wherein said solid form is a halogenide salt of the compound of formula (Id).Item 116. The solid form according to items 1 and 115, wherein said solid form is a halogenide salt of the compound of formula (Id) selected from the group consisting of a hydrochloride salt and a hydrobromide salt of the compound of formula (Id).Item 117. The solid form according to any of items 1 and 115-116, wherein said salt is a hydrochloride salt of the compound of formula (Id).Item 118. The solid form according to any one of items 115-117, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.2° 2θ: 5.7, 7.3, 10.6, 13.3, 15.3°.Item 119. The solid form according to any one of items 115-118, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.1° 2θ: 5.7, 7.3, 10.6, 13.3, 15.3°.Item 120. The solid form according to any one of items 118-119, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 15.4, 16.2, 20.1, 22.5 and 23.0°.Item 121. The solid form according to any one of items 115-118 and 120, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.2° 2θ: 5.7, 7.3, 10.6, 13.3, 15.3, 15.4, 16.2, 20.1, 22.5 and 23.0°.Item 122. The solid form according to any one of items 115-121, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.1° 2θ: 5.7, 7.3, 10.6, 13.3, 15.3, 15.4, 16.2, 20.1, 22.5 and 23.0°.Item 123. The solid form according to any one of items 115-118 and 121, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.2° 2θ: 5.1, 5.7, 7.3, 10.6, 13.3, 15.3, 15.4, 16.2, 16.7, 18.1, 20.1, 22.5, 23.0, 23.6 and 23.8°.Item 124. The solid form according to any one of items 115-123, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.1° 2θ: 5.1, 5.7, 7.3, 10.6, 13.3, 15.3, 15.4, 16.2, 16.7, 18.1, 20.1, 22.5, 23.0, 23.6 and 23.8°.Item 125. The solid form according to any one of items 115-124, wherein said hydrochloride salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.18.Item 126. The solid form according to any one of items 1 and 115-116, wherein said salt is a hydrobromide salt of the compound of formula (Id).Item 127. The solid form according to item 126, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 2θ-angles±0.2° 2θ: 12.5, 13.9, 14.5, 15.6, 18.6°.Item 128. The solid form according to any one of items 126-127, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.1° 2θ: 12.5, 13.9, 14.5, 15.6, 18.6°.Item 129. The solid form according to any one of items 127-128, wherein said x-ray powder diffraction pattern further comprises one or more peaks selected from the group consisting of peaks at the following 2θ-angles±0.2° 2θ: 18.9, 19.8, 21.3, 22.0 and 22.4°.Item 130. The solid form according to any one of items 126-127 and 129, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.2° 2θ: 12.5, 13.9, 14.5, 15.6, 18.6, 18.9, 19.8, 21.3, 22.0 and 22.4°.Item 131. The solid form according to any one of items 126-130, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.1° 2θ: 12.5, 13.9, 14.5, 15.6, 18.6, 18.9, 19.8, 21.3, 22.0 and 22.4°.Item 132. The solid form according to any one of items 126-127 and 129-131, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.2° 2θ: 12.5, 13.9, 14.5, 15.6, 18.6, 18.9, 19.8, 21.3, 22.0, 22.4, 23.3, 24.4, 25.5, 28.2 and 28.9°.Item 133. The solid form according to any one of items 126-132, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) comprising peaks at the following 20-angles±0.1° 2θ: 12.5, 13.9, 14.5, 15.6, 18.6, 18.9, 19.8, 21.3, 22.0, 22.4, 23.3, 24.4, 25.5, 28.2 and 28.9°.Item 134. The solid form according to any one of items 126-133, wherein said hydrobromide salt has a crystal form characterized by an XRPD obtained using CuKα1 radiation (λ=1.5406 Å) essentially as depicted inFIG.19.Item 135. The solid form of the compound of formula (Id) selected from the group consisting of the DH1 as defined in items 7-19, the HH as defined in items 32-42, and the potassium salt as defined in items 80-91.Item 136. The solid form of the compound of formula (Id) selected from the group consisting of the DH1 as defined in items 7-19 and the potassium salt as defined in items 80-91.Item 137. A solid form of the zwitterion of compound (Id), said solid form exhibiting a weight loss of about 7.6% w/w compared to the initial weight when heated from about 30° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 138. A solid form of the zwitterion of compound (Id), said solid form wherein said solid form is a crystal form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.8b.Item 139. A solid form of the zwitterion of compound (Id), said solid form is exhibiting a weight loss of less than 1% w/w compared to the initial weight when heated from about 30° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 140. A solid form of the zwitterion of compound (Id), wherein said solid form is characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.9b.Item 141. A solid form of the zwitterion of compound (Id), wherein said solid form is exhibiting a weight loss of about 21% w/w compared to the initial weight when heated from about 20° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 142. A solid form of the zwitterion of compound (Id), wherein said solid form is characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.10b.Item 143. A solid form of the zwitterion of compound (Id), wherein said solid form is exhibiting a weight loss of about 4% w/w compared to the initial weight when heated from about 20° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 144. A solid form of the zwitterion of compound (Id), wherein said solid form is characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.14b.Item 145. A solid form of compound (Id), wherein said solid form is the potassium salt exhibiting a weight loss of less than about 1% w/w compared to the initial weight when heated from about 20° C. to about 150° C. (heating rate 10° C./min), such as measured using thermogravimetric analysis.Item 146. A solid form of compound (Id), wherein said solid form is the potassium salt characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.15b.Item 147. A solid form of compound (Id), wherein said solid form is the sodium salt form exhibiting a weight loss of about 2% w/w compared to the initial weight when heated from about 20° C. to about 175° C. (heating rate 10° C./min), such as when measured using thermogravimetric analysis.Item 148. A solid form of the zwitterion of compound (Id), wherein said solid form is the sodium salt form characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.16b.Item 149. A solid form of compound (Id), wherein said solid form is the sodium salt form exhibiting a weight loss of about 5% w/w compared to the initial weight when heated from about 20° C. to about 175° C. (heating rate 10° C./min), such as when measured using thermogravimetric analysis.Item 150. A solid form of compound (Id), wherein said solid form is the sodium salt characterized by thermogravimetric analysis (using a heating rate 10° C./min) essentially as depicted inFIG.17b.Item 151. A solid form of the compound of formula (Id) according to any one of items 1-150, for use in therapy.Item 152. The DH1 form according to any one of items 7-19 for use in therapy.Item 153. The potassium salt form according to any one of items 80-91 for use in therapy.Item 154. The solid form of the compound of formula (Id) according to any of items 1-150, for use as a medicament.Item 155. The solid DH1 form according to any one of items 7-19 for use as a medicament.Item 156. The solid potassium salt form according to any one of items 80-91 for use as a medicament.Item 157. The solid form of the compound of formula (Id) according to any one of items 151-156, wherein said medicament is an oral medicament such as a tablet or a capsule for oral administration.Item 158. A pharmaceutical composition comprising a therapeutically effective amount of the solid form of the compound of formula (Id) according to any of items 1-150, and one or more pharmaceutically acceptable excipients.Item 159. The pharmaceutical composition according to item 158, wherein said solid form is the dihydrate of the zwitterion of the compound of formula (Id) DH1 according to any one of items 7-19.Item 160. The pharmaceutical composition according to item 158, wherein said solid form is the potassium salt of the compound of formula (Id) according to any one of items 80-91.Item 161. The pharmaceutical composition according to any one of items 158-160, wherein said pharmaceutical composition is for oral administration.Item 162. The pharmaceutical composition according to any one of items 158-161, wherein said pharmaceutical composition is an oral pharmaceutical composition.Item 163. The pharmaceutical composition according to any one of items 158-162, wherein said pharmaceutical composition is a solid oral dosage form.Item 164. The pharmaceutical composition according to any one of items 158-163, wherein said pharmaceutical composition is a tablet or a capsule for oral administration.Item 165. The pharmaceutical composition according to any one of items 158-164, wherein said pharmaceutical composition further comprises another agent which is useful in the treatment of a neurodegenerative disease or disorder such as Parkinson's disease.Item 166. The pharmaceutical composition according to any one of items 158-165, wherein said pharmaceutical composition further comprises a compound selected from the group consisting of L-DOPA, droxidopa, foliglurax, a MAO-B inhibitor such as selegiline or rasagiline, a COMT inhibitor such as entacapone or tolcapone, an adenosine 2a antagonist such as istradefylline, an antiglutamatergic agent such as amantadine or memantine, an acetylcholinesterase inhibitor such as rivastigmine, donepezil or galantamine, an antipsychotic agent such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or brexpiprazole; or an antibody targeting alpha-synuclein, Tau or A-beta protein.Item 167. A solid form of the compound of formula (Id) according to any of items 1-150, for use in the treatment of a neurodegenerative disease or disorder such as Parkinson's Disease, Huntington's disease, Restless leg syndrome or Alzheimer's disease; or a neuropsychiatric disease or disorder such as schizophrenia, attention deficit hyperactivity disorder or drug addiction.Item 168. The solid form for use according to item 167, wherein said solid form is the dihydrate of the zwitterion of the compound of formula (Id) DH1 according to any one of items 7-19.Item 169. The solid form for use according to item 167, wherein said solid form is the potassium salt form of the compound of formula (Id) as defined by any one of items 80-91.Item 170. The solid form of the compound of formula (Id) for use according to any of items 167-169, wherein said neurodegenerative disease or disorder is Parkinson's Disease.Item 171. The solid form for use according to item 170, wherein said solid form is the dihydrate of the zwitterion of the compound of formula (Id) DH1 according to any one of items 7-19, and wherein said neurodegenerative disease or disorder is Parkinson's Disease.Item 172. The solid form for use according to item 170, wherein said solid form is the potassium salt form of the compound of formula (Id) as defined by any one of items 80-91, and wherein said neurodegenerative disease or disorder is Parkinson's Disease.Item 173. The solid form for use according to any one of items 167-172, wherein said solid form is to be used in combination with another agent which is useful in the treatment of a neurodegenerative disease or disorder such as Parkinson's disease.Item 174. The solid form for use according to any of items 167-173, wherein said solid form is to be used in combination with a compound selected from the group consisting of L-DOPA, droxidopa, foliglurax, a MAO-B inhibitor such as selegiline or rasagiline, a COMT inhibitor such as entacapone or tolcapone, an adenosine 2a antagonist such as istradefylline, an antiglutamatergic agent such as amantadine or memantine, an acetylcholinesterase inhibitor such as rivastigmine, donepezil or galantamine, an antipsychotic agent such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or brexpiprazole; or in combination with an antibody targeting alpha-synuclein, Tau or A-beta protein.Item 175. The solid form for use according to any one of items 167-174, wherein said treatment is performed by oral administration of said compound.Item 176. The solid form for use according to any one of items 167-175, wherein said compound is comprised in an oral pharmaceutical composition such as a tablet or a capsule for oral administration.Item 177. A method for the treatment of a neurodegenerative disease or disorder such as Parkinson's Disease, Huntington's disease, Restless leg syndrome or Alzheimer's disease; or a neuropsychiatric disease or disorder such as schizophrenia, attention deficit hyperactivity disorder or drug addiction; which method comprises the administration of a therapeutically effective amount of solid form of the compound of formula (Id) according to any of items 1-150, to a patient in need thereof.Item 178. The method according to item 177, which method comprises the administration of a therapeutically effective amount of the dihydrate of the zwitterion of the compound of formula (Id) according to any one of items 7-19, to a patient in need thereof.Item 179. The method according to item 177, which method comprises the administration of a therapeutically effective amount of solid form of the potassium salt form of the compound of formula (Id) as defined by any one of items 80-91, to a patient in need thereof.Item 180. The method according to any one of items 177-179, wherein said neurodegenerative disease or disorder is Parkinson's Disease.Item 181. The method according to any one of items 177 and 180, which method comprises the administration of a therapeutically effective amount of the dihydrate of the zwitterion of the compound of formula (Id) according to any one of items 7-19, to a patient in need thereof, and wherein said neurodegenerative disease or disorder is Parkinson's Disease.Item 182. The method according to item 177, 179 and 180, which method comprises the administration of a therapeutically effective amount of solid form of the potassium salt form of the compound of formula (Id) as defined by any one of items 80-91, to a patient in need thereof, and wherein said neurodegenerative disease or disorder is Parkinson's Disease.Item 183. The method according to any one of items 177-182, wherein said solid form of compound (Id) is used in combination with another agent which is useful in the treatment of a neurodegenerative disease or disorder such as Parkinson's disease.Item 184. The method according to any one of items 177-183, wherein said solid form of compound (Id), is used in combination with a compound selected from the group consisting of L-DOPA, droxidopa, foliglurax, a MAO-B inhibitor such as selegiline or rasagiline, a COMT inhibitor such as entacapone or tolcapone, an adenosine 2a antagonist such as istradefylline, an antiglutamatergic agent such as amantadine or memantine, an acetylcholinesterase inhibitor such as rivastigmine, donepezil or galantamine, an antipsychotic agent such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or brexpiprazole; or in combination with an antibody targeting alpha-synuclein, Tau or A-beta protein.Item 185. The method according to any one of items 177-184, wherein said administration is performed by the oral route.Item 186. The method according to any one of items 177-185, wherein said solid form is comprised in an oral pharmaceutical composition such as a tablet or a capsule for oral administration.Item 187. Use of solid form of the compound of formula (Id) according to any one of items 1-150, in the manufacture of a medicament for the treatment of a neurodegenerative disease or disorder such as Parkinson's Disease, Huntington's disease, Restless leg syndrome or Alzheimer's disease; or for the treatment of a neuropsychiatric disease or disorder such as schizophrenia, attention deficit hyperactivity disorder or drug addiction.Item 188. The use according to item 187, for wherein said solid form is the zwitterion of the compound of formula (Id) DH1 according to any one of items 7-19.Item 189. The use according to item 187, for wherein said solid form is the potassium salt form of the compound of formula (Id) as defined by any one of items 80-91,Item 190. The use according to any one of items 172-174, wherein said neurodegenerative disease or disorder is Parkinson's Disease.Item 191. The use according to any one of items 187-190, wherein said medicament is used in combination with another agent which is useful in the treatment of a neurodegenerative disease or disorder such as Parkinson's disease.Item 192. The use according to any one of items 187-191, wherein said medicament is used in combination with a compound selected from the group consisting of L-DOPA, droxidopa, foliglurax a MAO-B inhibitor such as selegiline or rasagiline, a COMT inhibitor such as entacapone or tolcapone, an adenosine 2a antagonist such as istradefylline, an antiglutamatergic agent such as amantadine or memantine, an acetylcholinesterase inhibitor such as rivastigmine, donepezil or galantamine, an antipsychotic agent such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or brexpiprazole; or in combination with an antibody targeting alpha-synuclein, Tau or A-beta protein.Item 193. The use according to any one of items 187-192, wherein said medicament is an oral medicament such as a tablet or a capsule for oral administration. All references, including publications, patent applications and patents, cited herein are hereby incorporated by reference in their entirety and to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety (to the maximum extent permitted by law). Headings and sub-headings are used herein for convenience only and should not be construed as limiting the invention in any way. The description herein of any aspect or aspect of the invention using terms such as “comprising”, “having,” “including” or “containing” with reference to an element or elements is intended to provide support for a similar aspect or aspect of the invention that “consists of”, “consists essentially of” or “substantially comprises” that particular element or elements, unless otherwise stated or clearly contradicted by context (e.g., a composition described herein as comprising a particular element should be understood as also describing a composition consisting of that element, unless otherwise stated or clearly contradicted by context). The use of any and all examples, or exemplary language (including “for instance”, “for example”, “e.g.”, and “as such”) in the present specification is intended merely to better illuminate the invention and does not pose a limitation on the scope of invention unless otherwise indicated. It should be understood that the various aspects, embodiments, items, implementations and features of the invention mentioned herein may be claimed separately, or in any combination. The present invention includes all modifications and equivalents of the subject-matter recited in the claims appended hereto, as permitted by applicable law. EXPERIMENTAL SECTION Example 1: Preparation of Compound (Id) The compound of formula (Id) may be prepared by method described below, together with synthetic methods known in the art of organic chemistry, or modifications that are familiar to those of ordinary skill in the art. The starting materials used herein are available commercially or may be prepared by routine methods known in the art, such as those methods described in standard reference books such as “Compendium of Organic Synthetic Methods, Vol. I-XII” (published with Wiley-Interscience). Preferred methods include, but are not limited to, those described below. The schemes are representative of methods useful in synthesizing the compounds of the present invention. They are not intended to constrain the scope of the invention in any way. Compound (I) which can for example be prepared as disclosed in WO 2009/026934 was used as an intermediate in the synthesis of compounds of the invention. WO2019101917 further discloses methods for preparing compound (Id). LC-MS Methods Analytical LC-MS data were obtained using the methods identified below. Method 550: LC-MS were run on Waters Aquity UPLC-MS consisting of Waters Aquity including column manager, binary solvent manager, sample organizer, PDA detector (operating at 254 nM), ELS detector, and TQ-MS equipped with APPI-source operating in positive ion mode. LC-conditions: The column was Acquity UPLC BEH C18 1.7 μm; 2.1×50 mm operating at 60° C. with 1.2 ml/min of a binary gradient consisting of water+0.05% trifluoroacetic acid (A) and acetonitrile/water (95:5)+0.05% trifluoroacetic acid. Gradient (linear): 0.00 min10% B1.00 min100% B1.01 min10% B1.15 min10% BTotal run time:1.15 minutes. Method 551: LC-MS were run on Waters Aquity UPLC-MS consisting of Waters Aquity including column manager, binary solvent manager, sample organizer, PDA detector (operating at 254 nM), ELS detector, and TQ-MS equipped with APPI-source operating in positive ion mode. LC-conditions: The column was Acquity UPLC HSS T3 1.8 μm; 2.1×50 mm operating at 60° C. with 1.2 ml/min of a binary gradient consisting of water+0.05% trifluoroacetic acid (A) and acetonitrile/water (95:5)+0.05% trifluoroacetic acid. Gradient (linear): 0.00 min2% B1.00 min100% B1.15 min2% BTotal run time:1.15 minutes. Method 555: LC-MS were run on Waters Aquity UPLC-MS consisting of Waters Aquity including column manager, binary solvent manager, sample organizer, PDA detector (operating at 254 nM), ELS detector, and TQ-MS equipped with APPI-source operating in positive ion mode. LC-conditions: The column was Acquity UPLC BEH C18 1.7 μm; 2.1×150 mm operating at 60° C. with 0.6 ml/min of a binary gradient consisting of water+0.05% trifluoroacetic acid (A) and acetonitrile/water (95:5)+0.05% trifluoroacetic acid. Gradient (linear): 0.00 min10% B3.00 min100% B3.60 min10% BTotal run time:3.6 minutes. Preparative LCMS was performed using the method identified below. Waters AutoPurification system using combined mass/UV detection. Column: Sunfire 30×100 mm, 5 um particles. Operating at 40° C. with 90 ml/min of a binary gradient consisting of water+0.05% trifluoroacetic acid (A) and acetonitrile/water (3:5)+0.05% trifluoroacetic acid. Gradient (linear): 0.00 min98% A5.00 min50% A5.50 min98% A6.00 min98% A HighRes MS was run on a Bruker Compact qTOF equipped with electrospray operating in positive or negative mode. Direct infusion was used and calibration was done with sodium formate. Compound (Id) was prepared together with compound (Id′) depicted below and the two compounds were isolated from each other in the last step. Example 2: Preparation of Intermediates for Preparation of Compound (Id) and (Id′) Intermediates (4aR,10aR)-1-propyl-7-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-01, and (4aR,10aR)-1-propyl-6-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-ol (4aR,10aR)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinoline-6,7-diol, hydrochloride (2.21 g, 7.43 mmol) was suspended in dichloromethane (80 ml) under nitrogen atmosphere at room temperature, N,N-diisopropylethylamine (4.44 g, 6.0 ml, 34.4 mmol) was added followed by triisopropylsilyl chloride (2.73 g, 3.0 ml, 14.16 mmol) and the mixture was stirred at room temperature for 92 hours. 10 mL MeOH was added, and the crude mixture was evaporated, co-evaporated twice with dichloromethane/heptane, re-dissolved in dichloromethane, and evaporated directly on filter aid and purified by column chromatography (eluent: n-heptane/ethyl acetate/triethylamine, 100:0:0-35:60:5) affording 3.14 g as a mixture of (4aR,10aR)-1-propyl-7-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-01 (3.14 g) and (4aR,10aR)-1-propyl-6-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-ol as an oil. NMR (CDCl3) showed >30:1 mixture of silylated isomers. Intermediates tert-butyl ((4aR,10aR)-1-propyl-7-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl) carbonate [A], and tert-butyl ((4aR,10aR)-1-propyl-6-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl) carbonate [B] The mixture from the previous step (4aR,10aR)-1-propyl-7-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-ol and (4aR,10aR)-1-propyl-6-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-ol (2.94 g, 7.04 mmol) was dissolved in dichloromethane (30 ml) under a nitrogen atmosphere and cooled to 0° C. Pyridine (6.00 ml) followed by di-tert-butyl dicarbonate (6.30 g) were added and the reaction mixture was allowed to warm to room temperature over 3-4 hours and then stirred at room temperature overnight. 10 mL MeOH was added and the reaction mixture was evaporated, coevaporated with dichloromethane/n-heptane twice, dissolved in dichloromethane, and evaporated on filter aid. Purification by column chromatography (eluent: n-heptane/ethyl acetate/triethylamine, 100:0:0-75:20:5) gave a mixture of tert-butyl ((4aR,10aR)-1-propyl-7-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl) carbonate [A] and tert-butyl ((4aR,10aR)-1-propyl-6-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl) carbonate [B] (3.6 g) as an oil. NMR (CDCl3) after drying showed a mixture of regioisomers. Intermediates (4aR,10aR)-6-((tert-butoxycarbonyl)oxy)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl acetate, and (4aR,10aR)-7-((tert-butoxycarbonyl)oxy)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl acetate tert-Butyl-((4aR,10aR)-1-propyl-7-((triisopropylsilyl)oxy)-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl) carbonate (3.600 g, 6.95 mmol) (mixture of [A]:[B] from the previous step) was dissolved in THF (150 ml) under nitrogen atmosphere at 0° C., triethylamine trihydrofluoride (2.97 g, 3.00 ml, 18.42 mmol) was added and the mixture was stirred at 0° C. After 3 hours at 0° C., pyridine (10.0 ml, 124 mmol) and acetic anhydride (4.33 g, 4.00 ml, 42.4 mmol) were added directly to the reaction mixture at 0° C., and the reaction mixture was allowed to warm to room temperature. After 16 hours, 20 mL MeOH was added, and the reaction mixture was evaporated, redissolved in dichloromethane/n-heptane, and evaporated on filter aid followed by purification by dry column vacuum chromatography affording (4aR,10aR)-6-((tert-butoxycarbonyl)oxy)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl acetate and (4aR,10aR)-7-((tert-butoxycarbonyl)oxy)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl acetate as an oil/foam. LCMS (method 550) rt=0.56 minutes, [M+H]+=404 m/z. Intermediates (2S,3R,4S,5S,6S)-2-(((4aR,10aR)-7-acetoxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate, and (2S,3R,4S,5S,6S)-2-(((4aR,10aR)-6-acetoxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl)oxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (4aR,10aR)-6-((tert-butoxycarbonyl)oxy)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl acetate (2.489 g, 6.17 mmol) (mixture of (4aR,10aR)-6-((tert-butoxycarbonyl)oxy)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl acetate and (4aR,10aR)-7-((tert-butoxycarbonyl)oxy)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl acetate assumed) was dissolved in dichloromethane (60 ml) under nitrogen atmosphere at room temperature, (2S,3R,4S,5S,6S)-6-(Methoxycarbonyl)tetrahydro-2H-pyran-2,3,4,5-tetrayl tetraacetate (7.529 g, 20.01 mmol) was added followed by the addition of boron trifluoride diethyl etherate (6.72 g, 6.0 ml, 47.3 mmol) and the mixture was stirred at room temperature for 5 days. The mixture was diluted with dichloromethane and MeOH and evaporated on filter aid. Purification by dry column vacuum chromatography to give a mixture of (2S,3R,4S,5S,6S)-2-(((4aR,10aR)-7-acetoxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate and (2S,3R,4S,5S,6S)-2-(((4aR,10aR)-6-acetoxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl)oxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyltriacetate (4.37 g) as a foam/solid. LC-MS (method 555) rt=1.94 minutes, [M+H]+=620 m/z. (Id) (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid, and (Id′): (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-6-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid, and A mixture of (2S,3R,4S,5S,6S)-2-(((4aR,10aR)-7-acetoxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate and (2S,3R,4S,5S,6S)-2-(((4aR,10aR)-6-acetoxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl)oxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (3.82 g, 6.17 mmol) was dissolved in MeOH (100 ml) and water (20 ml), cooled to 0° C., potassium cyanide (7.295 g, 112 mmol) was added and the suspension was allowed to slowly warm to room temperature for 17.5 hours. The crude mixture was evaporated on filter aid and dried. The crude mixture was purified by silica gel column chromatography (eluent: ethyl acetate/MeOH/water 100:0:0-0:50:50), affording a 5-6:1 ratio of (Id′) and (Id). The mixture was separated by preparative LCMS. Collected Peak 1 fractions containing (Id′) were pooled, evaporated, and combined with another batch of 186 mg (Id′)-TFA, which had been prepared in a similar manner, using MeOH, evaporated, and dried to give a solid. (Id′) was re-suspended in 10 mL EtOH, and 100 mL MTBE was added, and the resulting suspension was stirred at room temperature for 8 hours, the suspension was filtered and the precipitate washed with 2×10 mL MTBE and dried in a vacuum oven overnight to afford (Id′) 1.601 g, as a solid corresponding to (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-6-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-7-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid. Collected Peak 2 fractions containing (Id) were pooled, evaporated, transferred to smaller flask with MeOH, evaporated, redissolved in ca. 12 mL MeOH, and repurified by preparative LCMS, and evaporated to give a foam/solid. Appropriate fractions were pooled, evaporated, transferred with MeOH to a smaller flask, and evaporated and combined with another batch of 40.7 mg (Id), which had been prepared in a similar manner. The combined batch was dissolved in 2.5 mL EtOH, 25 mL MTBE was added, and the suspension was stirred at room temperature. After 8 hours, the suspension was filtered and the precipitate washed with 2×2.5 mL MTBE and dried in the vacuum oven overnight to give 362.2 mg of (Id) as a solid. (Id) was suspended in ca. 10 mL EtOH, 50 mL MTBE was added, and the suspension was stirred at room temperature and filtered after 19 hours and the precipitate was washed with 2×10 mL MTBE, and dried in the vacuum oven at 40° C. to give (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid (Id) 0.279 g as a solid. (Id′) LCMS (method 551) rt=0.37 minutes, [M+H]+=438.1 m/z. 1H NMR (600 MHz, Methanol-d4) δ 7.02 (d, J=8.4 Hz, 1H), 6.65 (d, J=8.4 Hz, 1H), 4.73 (d, J=7.7 Hz, 1H), 3.89 (d, J=9.7 Hz, 1H), 3.68-3.58 (m, 2H), 3.54 (dd, J=9.3, 7.7 Hz, 1H), 3.49 (t, J=9.1 Hz, 1H), 3.47-3.36 (m, 2H), 3.30 (dt, J=11.2, 5.6 Hz, 1H), 3.21-3.11 (m, 3H), 2.85 (dd, J=15.4, 11.3 Hz, 1H), 2.35 (dd, J=17.6, 11.5 Hz, 1H), 2.12-2.02 (m, 2H), 2.02-1.84 (m, 3H), 1.81-1.71 (m, 1H), 1.49 (qd, J=13.0, 3.7 Hz, 1H), 1.09 (t, J=7.3 Hz, 3H). (Id) LCMS (method 551) rt=0.39 minutes, [M+H]+=438.1 m/z. 1H NMR (600 MHz, Methanol-d4) δ 6.87 (d, J=8.3 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 4.62 (d, J=7.9 Hz, 1H), 3.75 (dd, J=17.7, 4.9 Hz, 1H), 3.66-3.62 (m, 2H), 3.61-3.51 (m, 2H), 3.50-3.35 (m, 3H), 3.31-3.22 (m, 1H), 3.14 (qd, J=12.7, 4.0 Hz, 2H), 2.83 (dd, J=15.2, 11.3 Hz, 1H), 2.37 (dd, J=17.7, 11.7 Hz, 1H), 2.12 (d, J=13.4 Hz, 1H), 2.08-2.00 (m, 1H), 1.98-1.83 (m, 3H), 1.81-1.71 (m, 1H), 1.44 (qd, J=13.2, 3.9 Hz, 1H), 1.09 (t, J=7.3 Hz, 3H). Example 3: Preparation of Exemplified Solid Forms of the Invention The present example describes preparation methods for the solid forms of the invention and a characterization of the solid forms with respect to X-Ray powder diffractograms (XRPD) and Thermo gravimetric analysis (TGA). The characterization was performed using the methods as described below. XRPD: X-Ray powder diffractograms were measured on a PANalytical X'Pert PRO X-Ray Diffractometer using CuKα1radiation (λ=1.5406 Å). The samples were measured in reflection mode in the 2θ-range 2-40° or 3-40 using an X'celerator detector. Selected Peaks: The peaks were found by peak-search on the diffractogram using the program “HighScore Plus” from panalytical. 10 peaks selected as characteristic for the compounds are listed in Table 2 below (a), as well as the 15 peaks with highest intensity for each compound (b). Diffraction data are indicated ±0.1°. It is well known that relative intensities between characterization of batches of the same solid form may vary considerable due to preferred orientation effects. TABLE 2Overview of solid forms and XRPD peaksSolid formpeaksPeaks expressed in degree of diffraction angle 2θDihydrate(a)10.4, 11.6, 12.3, 13.1, 13.6, 14.3, 15.6, 16.0, 16.8, 18.5°(DH1)(b)12.3, 13.1, 13.6, 16.0, 16.8, 18.5, 18.9, 19.4, 20.5, 21.4, 23.5, 24.7,25.4, 26.9, 28.7°Anhydrate(a)8.5, 11.1, 12.4, 12.9, 15.6, 16.7, 18.9, 19.3, 20.0, 21.2°(AH1)(b)8.5, 12.4, 12.9, 15.6, 16.7, 18.9, 19.3, 20.0, 21.2, 21.5, 22.2, 23.0,24.2, 27.3, 28.3°Heptahydrate(a)7.0, 8.6, 10.2, 11.1, 11.9, 13.4, 14.0, 14.5, 17.0, 17.4°(HH)(b)7.0, 8.6, 10.2, 11.1, 11.9, 14.0, 17.0, 22.2, 25.9, 27.3, 28.3, 30.8, 34.0,34.8, 35.2°.Form A(a)7.6, 9.5, 10.0, 11.2, 12.0, 14.3, 14.6, 15.3, 15.5, 19.3°(b)7.6, 9.5, 10.0, 11.2, 12.0, 14.3, 14.6, 15.3, 15.5, 18.7, 19.3, 23.9, 28.8,33.7, 38.7°Form B(a)7.6, 9.0, 10.9, 12.3, 14.3, 15.0, 21.5, 22.1, 22.6, 23.7°(b)Only ten peaks identifiedForm C(a)7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5, 20.6°(b)7.5, 8.1, 10.3, 12.6, 13.5, 13.8, 14.9, 17.5, 18.5, 20.6, 21.6, 22.9, 23.1,24.0, 25.4°Monohydrate(a)9.2, 10.2, 11.8, 12.6, 13.6, 15.7, 16.0, 16.5, 17.5, 18.1°(MH1)(b)9.2, 10.2, 11.8, 12.6, 13.6, 16.0, 16.5, 17.5, 18.1, 18.7, 19.6, 22.9,24.7, 25.4, 26.0°Potassium(a)3.0, 9.0, 12.6, 13.6, 15.0, 17.1, 18.0, 18.4, 18.8, 19.4°salt(b)3.0, 9.0, 12.6, 13.6, 15.0, 18.0, 19.4, 21.8, 24.7, 27.1, 29.8, 33.3, 35.6,(K+salt)38.6, 39.6°Na+salt(a)5.9, 8.9, 11.9, 12.8, 13.8, 14.9, 17.7, 18.6, 19.0 and 19.5°.form 1(b)8.9, 12.8, 13.8, 14.9, 17.7, 18.6, 19.0, 19.5, 21.5, 21.8, 22.2, 22.6,22.9, 23.4, 25.1°Na+salt(a)5.6, 8.5, 12.6, 13.6, 14.1, 15.0, 16.7, 17.0, 18.8 and 19.8°.form 2(b)5.6, 8.5, 12.6, 13.6, 14.1, 15.0, 17.0, 18.8, 19.8, 21.0, 23.4, 28.5, 34.3,37.3, 38.5°HCl salt(a)5.7, 7.3, 10.6, 13.3, 15.3, 15.4, 16.2, 20.1, 22.5, 23.0°.(b)5.1, 5.7, 7.3, 10.6, 13.3, 15.3, 15.4, 16.2, 16.7, 18.1, 20.1, 22.5, 23.0,23.6, 23.8°.HBr salt(a)12.5, 13.9, 14.5, 15.6, 18.6, 18.9, 19.8, 21.3, 22.0, 22.4°.(b)12.5, 13.9, 14.5, 15.6, 18.6, 18.9, 19.8, 21.3, 22.0, 22.4, 23.3, 24.4,25.5, 28.2, 28.9°. TGA: Thermo gravimetric analysis (TGA) was measured using a TA-instruments Discovery TGA. 1-10 mg sample is heated 10°/min in an open pan under nitrogen flow. Sample sizes of about 2-6.4 mg. Preparation of dihydrate (DH1) of compound (Id) Example A To a 250 mL 1-necked round-bottomed flask were charged compound (Id) (4.0 g including some water of hydration), water (12 mL) and ethanol (12 mL). The white suspension was heated to 75° C., where a clear solution was obtained. The solution was cooled to 55° C. At 50-55° C. ethanol (56 mL) was added over 10 minutes. The suspension was stirred overnight at 50° C. The suspension was cooled over 6 hours to 23° C. and filtered. The filter cake was washed twice with ethanol (2×10 mL). The white filter cake was transferred to a drying tray and airdried in the fume hood for 1 day to constant weight. Yield 3.8 grams DH1. Example B To a 5 L 3-necked round-bottomed flask were charged compound (Id) (197 g including some water of hydration), water (0.60 L) and ethanol (0.60 L). The white suspension was heated to reflux, where a clear solution was obtained. The solution was kept at reflux for 30 minutes and then cooled over 35 minutes to 54° C. At 54° C. a slurry of the dihydrate (DH1) of compound (Id) (6.9 g) in ethanol (0.10 L) was added in one portion followed by additional ethanol (0.10 L). The temperature of the resulting suspension was increased from 52-54° C. in 5 minutes followed by addition of ethanol (1.60 L) over 17 minutes holding the temperature of the suspension at 53-55° C. during the addition. The suspension was stirred 1 hour at 53° C. and then cooled slowly overnight to 23° C. The suspension was filtered, and the resulting filter cake was washed twice with ethanol (2×0.40 L). The white filter cake was transferred to a drying tray and airdried in the fume hood for 2 days to constant weight. Yield 188 g DH1. The prepared DH1 was characterized by XRPD (see Table 2 andFIG.8) and TGA (seeFIG.8). Preparation of Anhydrate (AH1) of Compound (Id) Example A In a 500 mL round bottom flask equipped with a stir bar 8.8 g compound (Id) (evaporated mother liquor from other batches) was suspended in 9:1 EtOH/H2O (90 mL) and warmed to 95° C. The suspension was stirred (320 rpm) for 1 h 20 minutes at 95° C. The heat bath was then switched off and the mixture was stirred (320 rpm) for 2 h until the bath had reached RT. The precipitate was collected by vacuum filtration and the flask/filter cake was washed with EtOH (2×50 mL). The resulting solid was dried on the filter pad (with vacuum running) for 1 hour, then scraped into a crystallization dish and air dried for 48 hours. Yield: 7.4 g AH1. Example B To a 250 mL 1-necked round-bottomed flask were charged with compound (Id) (3.0 g), water (9 mL) and ethanol (9 mL). The suspension was heated to 75° C., where a clear solution was obtained. The solution was cooled to 55° C. At 50-55° C. ethanol (162 mL) was added over 15 minutes. Precipitation was observed during the addition of ethanol. The suspension was stirred overnight at 50° C. The suspension was cooled over 6 hours to 23° C. and filtered. The filter cake was washed twice with ethanol (2×10 mL). The filter cake was transferred to a drying tray and airdried in the fume hood for 1 day to constant weight. Yield 2.7 g AH1. The prepared AH1 was characterized by XRPD (see Table 2 andFIG.9A) and TGA (seeFIG.9B). Preparation of Heptahydrate (HH) of Compound (Id) The heptahydrate (HH) of compound (Id) was prepared by precipitation from water. 45.5 mg DH1 of compound (Id) as prepared in example b above was added 0.5 mL of water and shaked for ˜2 minutes. The wet crystals were removed from the solution and analysed by XRPD showing that HH was formed (see Table 2 andFIG.10A). The HH was further analysed by TGA (seeFIG.10b). Preparation of Form a of Compound (Id): Form A is obtained by storage of the heptahydrate of compound (Id) (HH) at room temperature at ˜5% RH. The prepared form A of compound (Id) was characterized by XRPD (see Table 2 andFIG.11). Preparation of Form B of Compound (Id): Form B was obtained by storage of the heptahydrate of compound (Id) (HH) at room temperature at ˜10% RH. The prepared form B of compound (Id) was characterized by XRPD (see Table 2 andFIG.12). Preparation of Form C of Compound (Id): Form C was obtained by storage of the heptahydrate of compound (Id) (HH) at room temperature at ˜15% RH. The prepared form C of compound (Id) was characterized by XRPD (see Table 2 andFIG.13). Preparation of Monohydrate (MH1) of Compound (Id): (MH1) was formed by heating of (DH1) to 105° C. and subsequent water sorption at ambient conditions to give a monohydrate. It is also obtained by drying (DH1) at room temperature to 0% RH, and subsequent water sorption at ambient conditions. The prepared MH1 was characterized by XRPD (see Table 2 andFIG.14a) and TGA (seeFIG.14b). Preparation of the Potassium Salt of Compound (Id): A 25 mL round bottomed flask with a magnetic stir bar was charged with compound (Id) heptahydrate (0.50 g). Then, water (0.5 mL) and aqueous potassium hydroxide (0.11 g, 0.075 mL, 0.90 mmol, 46% (w/w)) was added, and the mixture became a slurry. The mixture was heated to 80° C., then cooled to 50-60° C. Additional water (0.2 mL) was added resulting in an almost clear solution. i-PrOH (1.5 mL) was added dropwise, first a clear solution was obtained, then a white solid precipitated out. The temperature was raised to 80° C. and a clear solution was obtained. i-PrOH (2.5 mL) was added dropwise, then the mixture was warmed to reflux and 1-2 mL was distilled off, and i-PrOH (1-2 mL) was added and the distillation/addition was repeated once. The mixture was cooled slowly to 5° C. and filtered affording 0.41 g potassium salt of compound (Id). The prepared potassium salt was characterized by XRPD (see Table 2 andFIG.15a) and TGA (seeFIG.15b). Preparation of Sodium Salt Form 1 of Compound (Id): A 25 mL round bottomed flask with a magnetic stir bar was charged with (Id) heptahydrate (0.5 g). Then, water (0.500 ml) and NaOH (0.083 ml, 10.8 molar) was added, and the mixture became a slurry). The mixture was heated to 50° C., then additional water (0.500 ml) was added resulting in a clear solution. The temperature was raised to 80° C. and i-PrOH (3.50 ml) was added dropwise and gel-like solid precipitated out. The mixture was stirred for 30 minutes and then allowed to cool slowly to room temperature and then to 5° C. Then, the precipitate was isolated by very slow filtration (filtration for a period of at least 6 hours) and the precipitate was washed with 2×0.5 mL iPrOH. The solid was dried in the vacuum oven at 40° C. overnight. This afforded a sodium salt of compound (Id) (0.35 g) as a solid. The prepared sodium salt form 1 was characterized by XRPD (see Table 2 andFIG.16a) and TGA (seeFIG.16b). Preparation of Sodium Salt Form 2 of Compound (Id): 51.73 mg was added 70 μl water and the mixture was heated to 60° C. to dissolution where after 150 μl iPrOH was added. Then the mixture was heated to 60° C., 250 μl iPrOH was added and the mixture was heated to 60° C. After leaving at room temperature, a precipitation occurred. The liquid was sucked of and the solid part was placed at 90° C. which led to partial dissolution, so it was removed from the heat again and sodium salt form 2 was obtained. The prepared sodium salt form 2 was characterized by XRPD (see Table 2 andFIG.17a) and TGA (seeFIG.17b). Preparation of Hydrochloride Salt of Compound (Id): Ca. 500 mg of compound (Id) Heptahydrate was weighed and then slurried in 3.75 mL of IPA and 1.05 equivalents of HCl was added in 2.5 mL of IPA. The mixture of API/counterion/solvent was temperature cycled between ambient and 40° C. in 4-hour cycles. After ca. 1 day the preparation was observed to have dissolved at 40° C. and to contain a small amount of gum-like material at ambient. The experiment was allowed to evaporate at ambient temperature. The material appeared gum-like after incomplete evaporation and re-slurrying using IPA 500 μL IPA. Scale-up was re-prepared using less IPA, ca. 500 mg of compound (Id) Heptahydrate was weighed and then slurried in 0.9 mL of IPA and 1.05 equivalents of HCl was added in 2.5 mL of IPA. The mixture of API/counterion/solvent was temperature cycled between ambient and 40° C. in 4-hour cycles. After ca. 1 day the preparation was allowed to evaporate at ambient, due to limited solid present. After incomplete evaporation the material was scraped with a spatula, isolated by centrifuge filtration and dried under vacuum at ambient temperature for ca. 20 hours. The prepared hydrochloride salt was characterized by XRPD (see Table 2 andFIG.18). Preparation of Hydrobromide Salt of Compound (Id): Ca. 500 mg of compound (Id) Heptahydrate was weighed and slurried in 3.75 mL of IPA and 1.05 equivalents of HBr was added in 2.5 mL of IPA. The mixture of API/counterion/solvent was temperature cycled between ambient and 40° C. in 4-hour cycles for ca. 3 days. The prepared hydrobromide salt was characterized by XRPD (see Table 2 andFIG.19). Example 4: Stability Studies of Selected Solid Forms Stability studies were performed on the heptahydrate (HH), the dihydrate (DH1) and the potassium salt of compound (Id) (K+salt). The substances were packed individually in sealed polyethylene bags, with a carton box as a secondary packaging material. During stability the different batches were tested for visual appearance, assay (anhydrous i.e. calculated as water free compound), impurities and water content. In addition, XRPD was been performed as described in Example 3 at selected time points. In addition, stress stability studies were performed for the heptahydrate (HH), the dihydrate (DH1) and the potassium salt of compound (Id) (K+salt). For the stress stability studies, the substances were stored in open dishes in the dark at 40° C./75% RH, 60° C. and 60° C./80% RH. The following methods were used for the characterization: LC-UV Methods (Assay and Impurities) LC-UV was run on Agilent HPLC consisting of Agilent 1200 HPLC or equivalent including autosampler and DAD detector (operating at 278 nm). LC-conditions: The column was Synergi Polar-RP 4 μm; 4.6×150 mm operating at 40° C. with 1.0 ml/min of a binary gradient consisting of water/acetonitrile+2 ml TFA/ml (90:10) (A) and water/acetonitrile+2 ml TFA/ml (35:65) (B). Gradient: 0.0 min0% B2.0 min10% B12.0 min100% B14.0 min100% B14.1 min0% B19.0 min0% BTotal run time:19 minutes The amount of impurities was determined as % area of the peak of impurity relative to the area of the main peak. Karl Fischer Determination (Water Determination) The water content was determined by coulometric Karl Fischer titration according to the European Pharmacopoeia, chapter 2.5.32 (Metrohm 874 Oven Sample Processor and Metrohm 851 KF Coulometer). The water content was evaporated by heating samples to 150° C. and the water vapor was transferred by nitrogen to the titration chamber where it was titrated to endpoint using Hydranal Coulomat AG Oven (article number 34739) titration reagent. Results Stability Less than 0.1% degradation as determined by LC-UV were seen for both the heptahydrate (HH), the potassium salt (K+salt) and the dihydrate (DH1) of compound (Id). Changes in visual appearance, water content and physical form (XRPD) were seen for the heptahydrate (HH). After 3 months at 40° C./75% RH the heptahydrate changed appearance to a slightly grey colour and changed into the dihydrate. At 25° C./60% RH a slight change in colour is seen over time in addition to a clear change in physical form. Results are presented in the Table 3 below. TABLE 3Stability of HH of the zwitterion of compound (Id)HH25° C./60% RH40° C./75% RHStabilityWaterWatertimecontent,PhysicalAppear-content,PhysicalpointAppearance%formance%formInitialWhite21.6HHWhite21.6HHpowderpowderwithwithlumpslumps3White22.3HH +Slightly7.6DH1monthspowdersomegreyishwithDH1withlumpslumps6White20.0NPSlightly7.6NPmonthspowdergreyishwithwithlumpslumps9White21.6Form CNPNPNPmonthspowderwithlumps12Off-white15.0DH1 +NPNPNPmonthspowderform C18Off-white20.9NPNPNPNPmonthspowderNP: Not performed For the potassium salt, the results showed that that the samples remained in the same physical form as determined by XRPD. However, some changes in water content were observed, and formation of brown lumps was also observed seen during stability testing. A summary of results for the potassium salt of compound (Id) is shown in Table 4 below. TABLE 4Stability of the potassium salt of compound (Id)K+salt25° C./60% RH40° C./75% RHStabilityWaterWatertimecontent,Physicalcontent,PhysicalpointAppearance%formAppearance%formInitialVery slightly0.11Alfa formVery slightly0.11Alfa formbeige powderbeige powderwith lumpswith lumps3Very slightly0.22NPSlightly beige0.38NPmonthsbeige powderpowder withwith lumps andlumps andbrown spotsbrown spots6Very slightly0.17Alfa formSlightly beige0.25Alfa formmonthsbeige powderpowder withwith lumps andlumps andbrown spotsbrown spots9Very slightly<0.10Alfa formNPNPNPmonthsbeige powderwith lumps andbrown spots13Very slightly<0.10Alfa formNPNPNPmonthsbeige powderwith white andbrown lumps18Very slightly0.23Alfa formNPNPNPmonthsbeige powderwith white andbrown lumpsNP: Not performed, Alpha form: The K+salt form shown in Table 2. For the dihydrate, no changes in appearance, water content or physical form (XRPD) were seen for 6 months stability testing at 40° C./75% RH and at 25° C./60% RH. Even after 10 months at 25° C./60% RH, no changes were observed in the physical appearance and the water content. A summary of results for dihydrate of compound (Id) is shown in Table 5 below. TABLE 5Stability of the dihydrate of the zwitterion of compound (Id)DH125° C./60% RH40° C./75% RHStabilitytimeAppear-WaterPhysicalAppear-WaterPhysicalpointancecontent, %formancecontent, %formInitialWhite7.6DH1White7.6DH1powderpowder4White7.6DH1White7.4DH1monthspowderpowder6White7.6DH1White7.5DH1monthspowderpowder10White7.4NPNPNPNPmonthspowderNP: Not performed The results of the stability studies indicated that the dihydrate of the zwitterion of the compound (Id) (DH1) had best stability with respect to physical appearance, water content at physical form as measured at 40° C./75% RH and at 25° C./60% RH for 6 months. Stress Stability The stress stability testing was performed as described above, and the amount of degradation products were determined based on the LC-UV method described above. The total sum of impurities after 6 months storage is shown in the Table 6 below. TABLE 6Stress stabilityTotal sum ofimpurities after6 months storage40° C./75% RH60° C.60° C./80% RHHeptahydrate*<0.05%4.3%<0.05%Dihydrate0.05%1.0%0.05%Potassium salt0.10%0.10%1.0%*The heptahydrate has turned into the dihydrate at 40° C./75% RH and 60° C./80% RH. At 60° C. the heptahydrate has turned into form A. It can be seen from the results of the stress stability test in Table 6 that the tested solid forms were relatively stable with respect to chemical degradation, particularly the dihydrate and the potassium salt. Example 5: Further Stability Studies of Selected Solid Forms The following example describes further characterization of the heptahydrate (HH), the dihydrate (DH1) of compound (Id), and the potassium salt of compound (Id). DVS Dynamic vapor sorption (DVS) was further used to evaluate selected solid forms. Hygroscopicity and dehydration behavior can be investigated by DVS analysis. DVS experiments were performed using a DVS Advantage 01 instrument from Surface Measurement Systems. Samples of 4-10 mg of the solid forms were used for the analysis. The water absorption/desorption of the target solid was monitored while changing the relative humidity between about 0% to about 90% in steps of about 5-10% RH. FIGS.20and21show the resulting curves for the DH1 and the K+salt. For the DH1, DVS analysis showed that the water content of DH1 is very stable in the humidity range 5-90% RH. Less than 0.1% of water is absorbed or desorbed. For the potassium salt form of compound (Id) (K+salt), the DVS showed a gradual weight change up to 0.6% at 80% RH, and further 1% at 90% RH. Only at 95% RH a steady increase in the weight was observed, however from the curve it can be seen that the water desorbed as soon as the humidity was lowered again. The curve of the second cycle showed the same behavior as the first cycle, thus no change in the crystal lattice occurred and the salt is stable towards change in humidity. The DVS analysis of the heptahydrate (HH) indicated that the heptahydrate absorbs and desorbs water at humidity between 20% RH and 95% RH without changing the crystal form. At humidity below 20% the heptahydrate (HH) changes into other less hydrated forms and does not change back to the heptahydrate unless the material is exposed to high humidity. Thus, from the DVS analysis, it can be concluded that the DH1 is non-hygroscopic in the humidity range between 5-80% RH. Grinding and Pressure HH of Compound (Id) A sample of the heptahydrate was grinded by hand using mortar and pestle for 2 minutes, and subsequently analyzed by XRPD. The XRPD was compared to the XRPD prior to grinding. Upon grinding the reflections became a bit broader and an amorphous halo became visible. The grinded sample was stored at 95% RH for 1 week, and a following XRPD analysis showed that the reflections corresponded to the initial sample prior to grinding. Thus, the heptahydrate regained crystallinity after storage. DH1 of Compound (Id) A sample of the DH1 of compound (Id) was grinded by hand using mortar and pestle for 2 minutes, and a sample was pressured with 300PSI for 5 minutes. Subsequently, the sample was analysed by XRPD. The XRPD after the treatments did not show sign of decreased crystallinity compared to the XRPD of the initial sample prior to grinding and pressure testing. Further, another sample of the DH1 of compound (Id) was milled. XRPD of the milled sample compared with un-milled material did not show any difference in the XRPD pattern. Thus comparison of the XRPDs showed that there was no apparent change in crystallinity of the DH1 sample. It is therefore concluded that the DH1 of compound (Id) is very stable towards physical stress. Potassium Salt of Compound (Id) Samples of the K+salt form as described in Table 2 of Example 3 were either grinded with mortar and pestle or pressed in an IR-press for 5 minutes. The samples were analysed by XRPD after the treatment. Subsequently, the samples were placed at 95% RH for 1 week and reanalysed by XRPD. Results: Grinding lead to severe broadening of the XRPD reflections, but the subsequent exposure to high humidity lead to the sharp reflections again. Exposure to high pressure does also lead to some broadening of the XRPD reflections although to a less extent than grinding. The sharp reflections are regained by exposure to humidity. Thus, the K+salt form regained crystallinity after storage at high humidity. In conclusion, the DVS and grinding studies of the present example showed that the dihydrate of the zwitterion of compound (Id) was the most stable solid form, since it is non-hygroscopic and also was found to be stable when tested after grinding and pressure. Examples 6 to 10: In Vitro and In Vivo Characterization of Compound (Id) Example 6a: Conversion of the Compound of Formula (Id) in Rat and Human Hepatocytes Compound (Id) was incubated at 1 μg/mL with hepatocytes from human or rat suspended in DMEM (Dulbecco's Modified Eagle Medium) with HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid) at pH 7.4. The cell concentration at incubation was 1×106viable cells/mL. The incubations were performed in glass tubes at 37° C. with a total incubation volume of 3.5 mL and with duplicate incubations for each test item. The 3.5 mL of hepatocyte suspension was equilibrated for 10 minutes in a water bath set to 37° C. where after the incubations were initiated by adding 3.5 μL of a stock solution of the test item in DMSO (Dimethyl sulfoxide) and gently inverting the tubes. The final solvent concentration in the incubations was 0.1% DMSO. Samples of 600 μL were withdrawn from the incubations at the pre-determined time points of 0.25, 5, 15, 30 and 60 minutes after ensuring homogeneity of hepatocyte suspensions. The withdrawn volume was added to 1 mL Nunc cryo tubes on wet ice containing 60 μL of ice-cold ascorbic acid (100 mg/mL) and 30 μL of ice cold 100 mM saccharic acid 1.4-lactone in 0.5 M citric acid. The tubes were mixed and 35 μL of a solution of ice cold 20% formic acid was added. The tubes were mixed thoroughly and stored at −80° C. awaiting analysis. Analysis method and Instrumentation used for analysis of (I) from dosing compound (Id) was the one described in Examples 9 and 10 below in the section “Instrumentation used for analysis of compound (I) from dosing of compound (Ic) and (Id).” FIG.7indicates a time dependent conversion to compound (I) from (Id) in both rat and human hepatocytes. Example 6b: Conversion of the Compound of Formula (Id) in Fresh Rat and Human Blood Conversion of (Id) in human blood (average of 3 donors) and rat blood (average of 45 donors) to (I) was shown in fresh blood at 37° C. spiked with 1 μg/mL of (Id). (I) was measured at 0, 5, 15, 30 and 60 minutes in isolated plasma. Analysis method and Instrumentation as described in Examples 9 and 10 below in the section “Instrumentation used for analysis of compound (I) from dosing of compounds (Ic) and (Id).” FIG.8indicates a time dependent conversion to compound (I) from (Id), in both rat and human blood. Example 7: Dopamine Agonist Activity Dopamine D1 Receptor Agonism Dopamine D1 receptor agonism was measured using a HTRF cAMP from CisBio using the protocol developed by HD Biosciences (China). Briefly, the assay is a homogeneous time resolved-fluorescence resonance energy transfer (HTRF) assay that measures production of cAMP by cells in a competitive immunoassay between native cAMP produced by cells and cAMP-labeled with XL-665. A cryptate-labeled anti-cAMP antibody visualizes the tracer. The assay was performed in accordance with instructions from manufacturer. Test compounds were added to wells of microplates (384 format). HEK-293 cells expressing the human D1 receptor were plated at 1000 cells/well and incubated 30 minutes at room temperature. cAMP-d2 tracer was added to wells and followed by addition of Anti-cAMP antibody-cryptate preparation and incubated for 1 hour at room temperature in dark. HTRF cAMP was measured by excitation of the donor with 337 nm laser (the “TRF light unit”) and subsequent (delay time 100 microseconds) measurement of cryptate and d2 emission at 615 nm and 665 nm over a time window of 200 microseconds with a 2000 microseconds time window between repeats/100 flashes). HTRF measurements were performed on an Envision microplate reader (PerkinElmer). The HTRF signal was calculated as the emission-ratio at 665 nm over 615 nm. The HTRF ratio readout for test compounds was normalized to 0% and 100% stimulation using control wells with DMSO-solvent or 30 μM dopamine. Test compound potency (EC50) was estimated by nonlinear regression using the sigmoidal dose-response (variable slope) using Xlfit 4 (IDBS, Guildford, Surrey, UK, model 205). y=(A+((B−A)/(1+((C/x){circumflex over ( )}D)))) where y is the normalized HTRF ratio measurement for a given concentration of test compound, x is the concentration of test compound, A is the estimated efficacy at infinite compound dilution, and B is the maximal efficacy. C is the EC50value and D is the Hill slope coefficient. EC50estimates were obtained from an independent experiment and the logarithmic average was calculated. Dopamine D2 Receptor Agonism Dopamine D2 receptor agonism was measured using a calcium mobilization assay protocol developed by HD Biosciences (China). Briefly, HEK293/GI 5 cells expressing human D2 receptor were plated at a density of 15000 cells/well in clear-bottomed, Matrigel-coated 384-well plates and grown for 24 hours at 37° C. in the presence of 5% CO2. The cells were incubated with calcium-sensitive fluorescent dye, Fluo8, for 60-90 minutes at 37° C. in the dark. Test compounds were prepared at 3-fold concentrated solution in 1×HBSS buffer with Ca2+and Mg2+. Calcium Flux signal was immediately recorded after compounds were added from compound plate to cell plate at FLIPR (Molecular Devices). The fluorescence data were normalized to yield responses for no stimulation (buffer) and full stimulation (1 μM of dopamine) of 0% and 100% stimulation, respectively. Test compound potency (EC50) was estimated by nonlinear regression using the sigmoidal dose-response (variable slope) using Xlfit 4 (IDBS, Guildford, Surrey, UK, model 205). y=(A+((B−A)/(1+((C/x){circumflex over ( )}D)))) where y is the normalized ratio measurement for a given concentration of test compound, x is the concentration of test compound, A is the estimated efficacy at infinite compound dilution, and B is the maximal efficacy. C is the EC50value and D is the Hill slope coefficient. EC50estimates were obtained from independent experiment and the logarithmic average was calculated. Example 8: 5-HT2B Agonist Activity and Binding Assay 5-HT2B Agonist Activity Assay Evaluation of the agonist activity of compounds (I), (Ia) and (Ib) at the human 5-HT2B receptor was performed by Eurofins/Cerep (France) measuring the compound effects on inositol monophosphate (IP1) production using the HTRF detection method. Briefly, the human 5-HT2B receptor was expressed in transfected CHO cells. The cells were suspended in a buffer containing 10 mM Hepes/NaOH (pH 7.4), 4.2 mM KCl, 146 mM NaCl, 1 mM CaCl2), 0.5 mM MgCl2, 5.5 mM glucose and 50 mM LiCl, then distributed in microplates at a density of 4100 cells/well and incubated for 30 minutes at 37° C. in the presence of buffer (basal control), test compound or reference agonist. For stimulated control measurement, separate assay wells contained 1 μM 5-HT. Following incubation, the cells were lysed and the fluorescence acceptor (fluorophen D2-labeled IP1) and fluorescence donor (anti-IP1 antibody labeled with europium cryptate) were added. After 60 minutes at room temperature, the fluorescence transfer was measured at lambda(Ex) 337 nm and lambda(Em) 620 and 665 nm using a microplate reader (Rubystar, BMG). The IP1 concentration was determined by dividing the signal measured at 665 nm by that measured at 620 nm (ratio). The results were expressed as a percent of the control response to 1 μM 5-HT. The standard reference agonist was 5-HT, which was tested in each experiment at several concentrations to generate a concentration-response curve from which its EC50 value is calculated as described above for dopamine functional assays. 5-HT2B Binding Assay Evaluation of the affinity of compound (Id) for the human 5-HT2B receptor was determined in a radioligand binding assay at Eurofins/Cerep (France). Membrane homogenates prepared from CHO cells expressing the human 5HT2B receptor were incubated for 60 minutes at room temperature with 0.2 nM [1251](±)DOI (1-(4-iodo-2, 5-dimethoxyphenyl)propan-2-amine) in the absence or presence of the test compound in a buffer containing 50 mM Tris-HCl (pH 7.4), 5 mM MgCl2, 10 μM pargyline and 0.1% ascorbic acid. Nonspecific binding is determined in the presence of 1 μM (±)DOI. Following incubation, the samples were filtered rapidly under vacuum through glass fiber filters (GF/B, Packard) presoaked with 0.3% polyethyleneimine (PEI) and rinsed several times with ice-cold 50 mM Tris-HCl using a 96-sample cell harvester (Unifilter, Packard). The filters were dried and counted for radioactivity in a scintillation counter (Topcount, Packard) using a scintillation cocktail (Microscint 0, Packard). The results are expressed as a percent inhibition of the control radioligand specific binding. The standard reference compound was (±)DOI, which was tested in each experiment at several concentrations to obtain a competition curve from which its IC50is calculated. TABLE 7In vitro activities for the compounds of formula (I), (Ia), (Ib), (Ic)and (Id) obtained according to Examples 7 and 8.D1 EC50D2 EC505-HT2B EC50Compound(nM)/Emax(nM)/Emax(nM)/EmaxParent(I)3.3/99%1.3/91%2900 nM/50%compoundPrior art(Ia)>1000>1000>6000 nM,prodrugs58% @ 30 μM(Ib)>100046 nM/100%3.8 nM/79%(Ic)ndnd−5% @ 10 μM(Id)2700/98%1100/92%−25% @ 10 μM**indicate binding affinity (% inhibition of control, specific binding at concentration indicated)nd: not determined Example 9: PK Experiments in Rats For all the experiments, blood samples of approximately 0.68 mL were drawn from the tail or sublingual vein and put into K3EDTA tubes that had been pre-cooled and prepared with stabilizing solution consisting of 80 μL ascorbic acid and 40 μL 100 mM D-saccharic acid 1,4 lactone in water. The tubes were inverted gently 6-8 times to ensure thorough mixing and then placed in wet ice. The collecting tube was placed in wet ice for up to 30 minutes until centrifugation. Once removed from the wet ice the centrifugation was initiated immediately. Immediately after end of centrifugation the samples were returned to wet ice. Three sub-samples of 130 μL plasma were transferred to each of three appropriately labelled cryo tubes containing 6.5 μL pre-cooled formic acid (20%) (the tubes were pre-spiked and stored refrigerated prior to use). The tube lid was immediately replaced, and the plasma solution was thoroughly mixed by inverting gently 6-8 times. The samples were stored frozen at nominally −70° C. within 60 minutes after sampling. Centrifugation conditions at 3000 G for 10 minutes at 4° C. Plasma was placed on water-ice following collection. Final storage at approximately −70° C. Plasma samples were analyzed by solid phase extraction or direct protein precipitation followed by UPLC-MS/MS. MS detection using electrospray in the positive ion mode with monitoring of specific mass-to-charge transitions for compound (I) using internal standards for correcting the response. The concentration-time data was analyzed, using standard software using appropriate noncompartmental techniques to obtain estimates of the derived PK parameters. Instrumentation Used for Analysis of Compound (I) from Dosing Compound (La): Mass spectrometer (LC-MS/MS) Waters Acquity-Sciex API 5000. Analytical column Waters BEH UPLC Phenyl 100×2.1 mm column, 1.7 μm particle size. Mobile phase A: 20 mM ammonium formate (aq)+0.5% formic acid. Mobile phase B: Acetonitrile. Gradient run from 95/5% to 2/98 in 6.1 minutes. Flow rate 0.5 mL/min. MRM monitoring (multiple reaction monitoring) of test item and the added analytical standards Dosing and blood sampling: Han Wistar rats were supplied by Charles River Laboratories, Sulzfeld, Germany. An artificial, automatically controlled, light and dark cycle of 12 hours was maintained. The rats received a standard laboratory diet from Brogaarden (Altromin 1324 pellets). The rats had unrestricted access to the diet. During the study (a 4-week toxicity study) the rats received once daily doses of (Ia) orally by gavage. From rats given 300 μg/kg (Ia), blood samples) from 3 male satellite animals were collected on the following time points at Day 29: 0.5, 1, 2, 4, 6, 8, 12 and 24 hours after dosing. Instrumentation Used for Analysis of Compound (I) from Dosing of Compound (Ib): Mass spectrometer (LC-MS/MS) Waters Acquity-Sciex API 5000. Analytical column Waters BEH UPLC Phenyl 100×2.1 mm column, 1.7 μm particle size. Mobile phase A: 20 mM ammonium formate (aq)+0.5% formic acid. Mobile phase B: Acetonitrile. Gradient run from 95/5% to 2/98 in 6.1 minutes. Flow rate 0.5 mL/min. MRM monitoring of test item and the added analytical standards. Dosing and blood sampling: Han Wistar rats were supplied by Charles River Laboratories, UK. An artificial, automatically controlled, light and dark cycle of 12 hours was maintained. The rats received a standard laboratory diet (Teklad 2014C Diet.). The rats had unrestricted access to the diet. During the study (a 26-week toxicity study) the rats received once daily doses of (Ib) orally by gavage. From rats given 300 μg/kg (Ib), blood samples from 3 male satellite animals were collected on the following time points at day 182: 0.5, 1, 2, 4, 8 and 24 hours after dosing. Instrumentation Used for Analysis of Compound (I) from Dosing of Compounds (Ic) and (Id). Mass spectrometer (LC-MS/MS) Waters Acquity-Waters Xevo TQ-S. Analytical column Acquity BEH C18 100×2.1 mm, 1.7 μm. Mobile phase A: 20 mM NH4-Formate+0.2% formic acid. Mobile phase B: Acetonitrile+0.2% formic acid. Gradient run from 95/5% to 5/95% in 11.0 minutes. Flow rate 0.3 mL/min. MRM monitoring of test item and the added analytical standards. Dosing and blood sampling for compound (Id): Han Wistar rats were supplied by Charles River Laboratories, Wiga GmbH, Germany. An artificial, automatically controlled, light and dark cycle of 12 hours was maintained. The rats received a standard laboratory diet from Brogaarden (Altromin 1324 pellets). The rats had unrestricted access to the diet. Male Han Wistar rats were dosed a single oral gavage administration of compound (Id) orally by gavage. Rats were given 633 μg/kg of compound (Id), blood samples from 3 male animals were collected on the following time points at Day 1: 1, 2, 4, 6, 8, and 24 hours after dosing. Dosing and blood sampling for compound (Ic): Han Wistar rats were supplied by Envigo, UK. An artificial, automatically controlled, light and dark cycle of 12 hours was maintained. The rats received a standard laboratory diet Teklad 2014C. The rats had unrestricted access to the diet. Male Han Wistar rats were dosed a single oral gavage administration of (Ic), 494 μg/kg. Blood samples from 3 male animals were collected on the following time points at Day 1: 1, 2, 4, 6, 8, and 24 hours after dosing. Instrumentation Used for Analysis of Apomorphine: Mass spectrometer (UPCLC-MS/MS) Waters Acquity 1-Class-Waters Xevo TQ-S. Analytical column Acquity HSS T3 C18 50×2.1 mm, 1.8 μm. Mobile phase A: 10 mM NH4-Formate 0.2% formic acid:Acetonitril (95:5). Mobile phase B: 10 mM NH4-Formate 0.2% formic acid:Acetonitril (5:95). Gradient run from 95/5% to 5/95% in 2.40 minutes. Flow rate 0.3 mL/min. MRM detection of test items and the added analytical standards. Dosing and Blood Sampling for Apomorphine: Animals for the study were as described in Example 10. Additionally, rats were administered a single dose of apomorphine subcutaneously. From rats administered 3000 μg/kg (apomorphine), blood samples from 3 male animals were collected on the following time points at Day 1: 0.25, 0.5, 1, 1½, 2, 3, 5 and 7 hours SC administration after dosing. TABLE 8PK parameters for (4aR,10aR)-1-Propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol (compound (I)) after oral dosing of 0.300 mg/kg (Ia), 0.300 mg/kg(Ib), 0.633 mg/kg of (Id) and 494 μg/kg (Ic) to Wistar rats according to Example 9.Exposureat 24TmaxCmaxAUC0-24t1/2hourscompound(h)(pg/mL)(pg*h/mL)(h)(pg/mL)Prior art(Ia)1.03160136004.0948 ± 26prodrugs(Ib)1.0499031000N/A147 ± 28(Ic)1.014104N/AN/ACompound(Id)4.01350155006.8208 ± 89(Id) Example 10: PK/PD of compound (1d)/compound (I) in rat hyperactivity assay Animals In total, 206 male CD rats (Charles River, Germany) weighing 200-250 grams (165-190 grams upon arrival) were used in the study. Animals were housed at a standard temperature (22±1° C.) and in a light-controlled environment (lights on from 7 am to 8 μm) with ad libitum access to food and water. The experiment described below was performed in accordance with the standard operating procedures of Charles River Discovery Research Services Finland Ltd. and in accordance with the national Animal Experiment Board of Finland (Eläinkoelautakunta, ELLA) authority on animal testing. Locomotor Activity Testing, Open Field The test device is a square Plexiglass-arena (measuring 40×40×40 cm), in which the movement paths of the rats are recorded by an activity monitor (Med. Associates Inc.). Before the test period is initiated, rats are habituated to their test cage for 60 minutes. Upon completion of habituation, animals were treated with either compound or vehicle and placed back into the open field apparatus. The main test parameter measured is ambulatory distance (recorded in 5-minute segments). Overall time of measurement after receiving initial treatment was 360 minutes. Total follow up period in the study was 420 min, including 60 min of habituation. Results Oral administration of compound (Id) was assessed in the rat locomotor activity assay, and this functional readout was then correlated to plasma concentrations of compound (I). Apomorphine and pramipexole were also concomitantly tested in this assay as comparators (i.e. known standard-of-care (SoC) in the Parkinson's Disease field), and plasma concentration was analyzed for apomorphine. As shown inFIG.2, compound (Id) (10 to 300 μg/kg, p.o.) increases locomotor activity with an effect starting approximatively 2 hours' post-administration (around the 180-minute time point) and lasting until the end of recording (at the 415-minute time point). In contrary, the increased locomotor activity (hyperactivity) induced by apomorphine (3 mg/kg, s.c.) is immediate but short-lasting as the effect is gone 1.5 hours post administration (at the 150-minute time point). Pramipexole (0.3 mg/kg, s.c.) also induces an increase in activity, but it's effect appears about 1 hour post administration and is gone 2.5 hours later (at the 270-minute time point). The total distance travelled as seen inFIG.3demonstrates a significantly increased activity for both compound (Id) and the two comparators tested, and this effect is the one that is to be expected from dopamine agonists. In parallel with the locomotor activity assessment, plasma samples were taken from satellite animals at 6 different time points (1.5, 2, 3, 4, 5 & 7 hours) post-dose for animals treated with compound (Id)). Pharmacokinetic analysis demonstrates that the behavioral effects of compound (Id) (100 μg/kg, p.o.) correlate with the plasma concentrations of compound (I) (seeFIG.4), demonstrating that the behavioral effect of compound (Id) is driven by Compound (I) rather than by Compound (Id) itself. The corresponding exposure analysis of apomorphine administered subcutaneously (at 1.25, 1.5, 2, 3, 5 & 7 hours post-dose) resulted in a correlation between plasma concentrations of apomorphine and hyperactive behavior (seeFIG.5). REFERENCE LIST U.S. Pat. No. 4,543,256WO2001/078713WO 02/100377WO2009/026934WO2009/026935WO2010/097092WO2019101917Alexander et Crutcher, (1990) Trends in Neuroscience 13: 266-71;Bibbiani et al., Chase Experimental Neurology (2005), 192: 73-78;Campbell et al., Neuropharmacology (1982); 21(10): 953-961;Cannon et al., J. Heterocyclic Chem. (1980); 17: 1633-1636;Cavero and Guillon, J. Pharmacol. Toxicol. Methods (2014), 69: 150-161;Delong, (1990) Trends in Neuroscience 13: 281-5;Gerfen et al, Science (1990) 250: 1429-32;Giardina and Williams; CNS Drug Reviews (2001), Vol. 7 (3): 305-316;Goswami et al., J. Nutritional Biochem. (2003) 14: 703-709;Grosset et al., Acta Neurol Scand. (2013), 128:166-171;Hauser et al., Movement Disorders (2016), Vol. 32 (9): 1367-1372;Liu et al., J. Med. Chem. (2006), 49: 1494-1498;Liu et al., Bioorganic Med. Chem. (2008), 16: 3438-3444;Nolen et al., J. Pharm Sci. (1995), 84 (6): 677-681;Poewe et al., Nature Review, (2017) vol 3 article 17013: 1-21;Remington, “The Science and Practice of Pharmacy”, 22th revised edition (2013), Edited byAllen, Loyd V., Jr.Rothman et al., Circulation (2000), 102: 2836-2841;Sprenger and Poewe, CNS Drugs (2013), 27: 259-272;Sozio et al., Exp. Opin. Drug Disc. (2012); 7(5): 385-406;Stain-Texier et al., Drug Metab. and Disposition (1998) 26 (5): 383-387;Wiley-Interscience (publisher): Compendium of Organic Synthetic Methods, Vol. I-XII
140,689
11858955
DETAILED DESCRIPTION To date, there remains a need for a flavor modifier that can provide a desired level of mouth watering and/or lubricating and/or slippery and/or astringent mouthfeel in various edible compositions. The present application relates to flavor compositions that include at least one HMG glucoside compound. In certain non-limiting embodiments, the HMG glucoside comprises a compound of Formula I. The flavor compositions can be used to enhance or modify the taste and/or flavor and/or texture of various edible compositions such as sweet goods and savory goods. The flavor compositions can include combinations of compounds, and can be added to edible compositions in various delivery system formats. 1. Definitions The terms used in this specification generally have their ordinary meanings in the art, within the context of this invention and in the specific context where each term is used. Certain terms are discussed below, or elsewhere in the specification, to provide additional guidance to the practitioner in describing the compositions and methods of the invention and how to make and use them. As used herein, the use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.” Still further, the terms “having,” “including,” “containing” and “comprising” are interchangeable and one of skill in the art is cognizant that these terms are open ended terms. The term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 3 or more than 3 standard deviations, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. As used herein, “taste” refers to a sensation caused by activation or inhibition of receptor cells in a subject's taste buds. In certain embodiments, taste can be selected from the group consisting of sweet, sour, salt, bitter, kokumi and umami. In certain embodiments, a taste is elicited in a subject by a “tastant.” In certain embodiments, a tastant is a synthetic tastant. In certain embodiments, the tastant is prepared from a natural source. As used herein, “taste profile” refers to a combination of tastes, such as, for example, one or more of a sweet, sour, salt, bitter, kokumi and/or umami taste. In certain embodiments, a taste profile is produced by one or more tastant that is present in a composition at the same or different concentrations. In certain embodiments, a taste profile refers to the intensity of a taste or combination of tastes, for example, a sweet, sour, salt, bitter, kokumi and/or umami taste, as detected by a subject or any assay known in the art. In certain embodiments, modifying, changing or varying the combination of tastants in a taste profile can change the sensory experience of a subject. As used herein, “flavor” refers to one or more sensory stimuli, such as, for example, one or more of taste (gustatory), smell (olfactory), touch (tactile) and temperature (thermal) stimuli. The terms “flavor” and “aroma” are synonymous and are used interchangeably. In certain non-limiting embodiments, the sensory experience of a subject exposed to a flavor can be classified as a characteristic experience for the particular flavor. For example, a flavor can be identified by the subject as being, but not limited to, a floral, citrus, berry, nutty, caramel, chocolate, peppery, smoky, cheesy, meaty, etc. flavor As used herein, a flavor composition can be selected from a liquid, dry powder, spray, paste, suspension and any combination thereof. The flavor can be a natural composition, an artificial composition, a nature identical, or any combination thereof. As used herein, “flavor profile” refers to a combination of sensory stimuli, for example, tastes, such as sweet, sour, bitter, salty, kokumi and/or umami tastes, and/or olfactory, tactile and/or thermal stimuli. In certain embodiments, the flavor profile comprises one or more flavors which contribute to the sensory experience of a subject. In certain embodiments, modifying, changing or varying the combination of stimuli in a flavor profile can change the sensory experience of a subject. As used herein, “texture profile” or “mouthfeel” refers to a composition's physical and chemical interaction in the mouth. The texture profile of a composition can include one or more texture, such as, for example, but not limited to, mouth watering, lubricating, slippery, astringency, hardness, cohesiveness, viscosity, elasticity, adhesiveness, brittleness, chewiness, gumminess, moisture content, grittiness, smoothness, oiliness and greasiness. In certain embodiments, the texture profile can comprise one or more texture characteristic in the same or different intensities. In certain embodiments, the texture profile can remain constant or change during a sensory experience, for example, from initial perception of a composition on the palate, to first bite, through mastication and finally, the act of swallowing. As used herein, “sensory experience” refers to a subject's sensory perception of a taste, taste profile, flavor, flavor profile or texture profile. As used herein, “ppb” means parts-per-billion and is a weight relative parameter. A part-per-billion is a nanogram per gram, such that a component that is present at 10 ppb is present at 10 nanograms of the specific component per 1 gram of the aggregate mixture. As used herein, “ppm” means parts-per-million and is a weight relative parameter. A part-per-million is a microgram per gram, such that a component that is present at 10 ppm is present at 10 micrograms of the specific component per 1 gram of the aggregate mixture. As used herein “admixing,” for example, “admixing the HMG glucoside flavor composition, or combinations thereof, of the present application with a food product,” refers to the process where the flavor composition is mixed with or added to the completed product or mixed with some or all of the components of the product during product formation or some combination of these steps. When used in the context of admixing the term “product” refers to the product or any of its components. This admixing step can include a process selected from the step of adding the flavor composition to the product, spraying the flavor composition on the product, coating the flavor composition on the product, suspending the product in the flavor composition, painting the flavor composition on the product, pasting the flavor composition on the product, encapsulating the product with the flavor composition, mixing the flavor composition with the product and any combination thereof. The flavor composition can be a liquid, dry powder, spray, paste, suspension and any combination thereof. As used herein “food product” refers to an ingestible product, such as, but not limited to, human food, animal (pet) foods, and pharmaceutical compositions. As used herein “flavor composition” refers to at least one compound or biologically acceptable salt thereof that modulates, including enhancing, multiplying, potentiating, decreasing, suppressing, or inducing, the tastes, smells, flavors and/or textures of a natural or synthetic tastant, flavoring agent, taste profile, flavor profile and/or texture profile in an animal or a human. In certain embodiments, the flavor composition comprises a combination of compounds or biologically acceptable salts thereof. In certain embodiments, the flavor composition includes one or more excipients. As used herein “savory flavor” refers to a savory, “mouth-watering,” sensation. In certain embodiments, a savory flavor is induced by one or more combination of umami tastants, for example, MSG (monosodium glutamate) in an animal or a human. In certain embodiments, “wet soup category” means wet/liquid soups regardless of concentration or container, including frozen soups. For the purpose of this definition “soup(s)” means a food prepared from meat, poultry, fish, vegetables, grains, fruit and/or other ingredients, cooked in a liquid which may include visible pieces of some or all of these ingredients. It may be clear (as a broth) or thick (as a chowder), smooth, pureed or chunky, ready-to-serve, semi-condensed or condensed and may be served hot or cold, as a first course or as the main course of a meal or as a between meal snack (sipped like a beverage). Soup may be used as an ingredient for preparing other meal components and may range from broths (consomme) to sauces (cream or cheese-based soups). As used herein, “dehydrated and culinary food category” means: (i) Cooking aid products such as: powders, granules, pastes, concentrated liquid products, including concentrated bouillon, bouillon and bouillon like products in pressed cubes, tablets or powder or granulated form, which are sold separately as a finished product or as an ingredient within a product, sauces and recipe mixes (regardless of technology); (ii) Meal solution products such as: dehydrated and freeze dried soups, including dehydrated soup mixes, dehydrated instant soups, dehydrated ready-to-cook soups, dehydrated or ambient preparations of ready-made dishes, meals and single serve entrees including pasta, potato and rice dishes; and (iii) Meal embellishment products such as: condiments, marinades, salad dressings, salad toppings, dips, breading, batter mixes, shelf stable spreads, barbecue sauces, liquid recipe mixes, concentrates, sauces or sauce mixes, including recipe mixes for salad, sold as a finished product or as an ingredient within a product, whether dehydrated, liquid or frozen. As used herein, “beverage category” means beverages, beverage mixes and concentrates, including but not limited to, alcoholic and non-alcoholic ready to drink and dry powdered beverages. Other examples of foods and beverages wherein compounds according to the application may be incorporated included by way of example carbonated and non-carbonated beverages, e.g., sodas, fruit or vegetable juices, alcoholic and non-alcoholic beverages, confectionary products, e.g., salad dressings, and other condiments, cereal, and other breakfast foods, canned fruits and fruit sauces and the like. As used herein, “frozen food category” means chilled or frozen food products. Non-limiting examples of food products of the frozen food category include ice cream, impulse ice cream, single portion dairy ice cream, single portion water ice cream, multi-pack dairy ice cream, multi-pack water ice cream, take-home ice cream, take-home dairy ice cream, ice cream desserts, bulk ice cream, take-home water ice cream, frozen yoghurt, artisanal ice cream, frozen ready meals, frozen pizza, chilled pizza, frozen soup, frozen pasta, frozen processed red meat, frozen processed poultry, frozen processed fish/seafood, frozen vegetables, frozen processed vegetables, frozen meat substitutes, frozen potatoes, frozen bakery products and frozen desserts. As used herein, “snack food category” generally refers to any food that can be a light informal meal including, but not limited to sweet and savory snacks and snack bars. Examples of snack foods include, but are not limited to, fruit snacks, chips/crisps, extruded snacks, tortilla/corn chips, popcorn, pretzels, nuts and other sweet and savory snacks. Examples of snack bars include, but are not limited to granola/muesli bars, breakfast bars, energy bars, fruit bars and other snack bars. As used herein, “meat food product” refers generally to a food product made by processing the edible remains of any dead animal, including birds, fish, crustaceans, shellfish and mammals. Meat food products include, without limitation, for example, prepared beef, lamb, pork, poultry or seafood products. For example, meat food products include bologna, frankfurters, sausage, luncheon, deli slices, loaves, bacon, meatballs, fish sticks, chicken fingers, and ground meats, e.g., meatloaf, meatballs and hamburgers. As used herein, “simulated meat food product” includes, without limitation, for example, a meat alternative, meat analog, soy burger, soy bologna, soy frankfurter, soy sausage, soy luncheon loaves, soy bacon and soy meatball. As used herein, “food product source” refers generally to the raw products from which a food product is made. In certain embodiments, the food product source is a vegetable, fruit or any other plant material. In certain embodiments, the plant material is cacao, cocoa beans, or cocoa liquor. In other embodiments, the food product source comprises the remains of any dead animal, including birds, fish, crustaceans, shellfish and mammals. The term “alkyl” refers to a straight or branched C1-C20(preferably C1-C6) hydrocarbon group consisting solely of carbon and hydrogen atoms, containing no unsaturation, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl). The term “alkenyl” refers to a C2-C20(preferably C1-C4) aliphatic hydrocarbon group containing at least one carbon-carbon double bond and which may be a straight or branched chain, e.g., ethenyl, 1-propenyl, 2-propenyl (allyl), iso-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl. The term “cycloalkyl” denotes an unsaturated, non-aromatic mono- or multicyclic hydrocarbon ring system (containing, for example, C3-C6) such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl. Examples of multicyclic cycloalkyl groups (containing, for example, C6-C15) include perhydronapththyl, adamantyl and norbornyl groups bridged cyclic group or sprirobicyclic groups, e.g., spiro (4,4) non-2-yl. The term “cycloalkalkyl” refers to a cycloalkyl as defined above directly attached to an alkyl group as defined above, that results in the creation of a stable structure such as cyclopropylmethyl, cyclobutylethyl, cyclopentylethyl. The term “alkyl ether” refers to an alkyl group or cycloalkyl group as defined above having at least one oxygen incorporated into the alkyl chain, e.g., methyl ethyl ether, diethyl ether, tetrahydrofuran. The term “alkyl amine” refers to an alkyl group or a cycloalkyl group as defined above having at least one nitrogen atom, e.g., n-butyl amine and tetrahydrooxazine. The term “aryl” refers to aromatic radicals having in the range of about 6 to about 14 carbon atoms such as phenyl, naphthyl, tetrahydronapthyl, indanyl, biphenyl. The term “arylalkyl” refers to an aryl group as defined above directly bonded to an alkyl group as defined above, e.g., —CH2C6H5, and —C2H4C6H5. The term “heterocyclic” refers to a stable 3- to 15-membered ring radical which consists of carbon atoms and one or more, for example, from one to five, heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. For purposes of this application, the heterocyclic ring radical may be a monocyclic or bicyclic ring system, which may include fused or bridged ring systems, and the nitrogen, carbon, oxygen or sulfur atoms in the heterocyclic ring radical may be optionally oxidized to various oxidation states. In addition, the nitrogen atom may be optionally quaternized; and the ring radical may be partially or fully saturated (i.e., heteroaromatic or heteroaryl aromatic). The heterocyclic ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure. The term “heteroaryl” refers to a heterocyclic ring wherein the ring is aromatic. The term “heteroarylalkyl” refers to heteroaryl ring radical as defined above directly bonded to alkyl group. The heteroarylalkyl radical may be attached to the main structure at any carbon atom from alkyl group that results in the creation of a stable structure. The term “heterocyclyl” refers to a heterocylic ring radical as defined above. The heterocyclyl ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure. 2. HMG Glucoside Compounds The present application relates to flavor compositions that include at least one 3-hydroxy-3-methylglutaric acid (HMG) glucoside compound (HMG glucoside). The flavor compositions can be used to enhance or modify the taste and/or flavor and/or texture of various edible compositions such as sweet goods and savory goods. The flavor compositions can include combinations of compounds, and can be added to edible compositions in various delivery system formats. In certain non-limiting embodiments, the HMG glucoside comprises a compound of Formula I having the following structure. wherein R is selected from the group consisting of substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted cycloalkalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroarylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocyclic, substituted or unsubstituted alkoxy, substituted or unsubstituted aryloxy, hydroxyl, hydrogen, substituted or unsubstituted ether, substituted or unsubstituted benzothiazol, substituted or unsubstituted pyridyl, substituted or unsubstituted naphthyl, substituted or unsubstituted phenyl, substituted or unsubstituted thienyl, substituted or unsubstituted benzothienyl, substituted or unsubstituted indol, substituted or unsubstituted isoquinolyl, substituted or unsubstituted quinolyl, —C(O)R1and —S(O)2R1, wherein R1is defined as above for R. The substituents in the substituted groups described herein, for example, “substituted ether”, “substituted alkyl”, “substituted alkenyl”, “substituted cycloalkyl”, “substituted cycloalkalkyl”, “substituted arylalkyl”, “substituted aryl”, “substituted heterocyclic”, “substituted heteroarylalkyl,” “substituted heteroaryl”, “substituted naphthyl”, “substituted phenyl”, “substituted thienyl”, “substituted benzothienyl”, “substituted pyridyl”, “substituted indol”, “substituted isoquinolyl”, “substituted quinolyl”, or “substituted benzothiazol” may be the same or different with one or more selected from the groups described in the present application and hydrogen, halogen, amide, acetyl, nitro, oxo (═O), thio (═S), —NO2, —CF3, —OCH3, -Boc or optionally substituted groups selected from alkyl, alkoxy, aryl, aryloxy, arylalkyl, ether, ester, hydroxyl, heteroaryl, and heterocyclic ring. A “substituted” functionality may have one or more than one substituent. In one non-limiting embodiment, R is isopentyl, or an isopentyl derivative. In certain embodiments, the compounds of the application comprise the following structure (Formula II): In certain embodiments, the compounds of the application comprise the following structure (Formula III): In certain embodiments, the compounds of the application comprise the following structure (Formula IV): In certain embodiments, the compounds of the application comprise the following structure (Formula V): In certain embodiments, the compounds of the application comprise stereoisomers of Formula I. In certain embodiments, the compounds of the application comprise a stereoisomer of Formula II, comprising a structure selected from the group consisting of: and combinations thereof. In certain embodiments, the HMG glucoside compounds of the present application comprise a salt of the HMG glucoside, for example, but not limited to, an acetate salt, a TFA salt, or a formate salt. In certain embodiments, the HMG glucoside salt comprises an anion (−) (for example, but not limited to, Cl−, F−, Br, O2−, CO32−, HCO3−, OH−, NO3−, PO43−, SO42−, CH3COO−, HCOO−, C2O42−and CN) bonded via an ionic bond with a cation (+) (for example, but not limited to, Al3+, Ca2+, Na+, K+, Cu2+, H+, Fe3+, Mg2+, Ag+, NH4+, H3O+, Hg22+). In other embodiments, the HMG glucoside salt comprises a cation (+) bonded via an ionic bond with an anion (−). In certain embodiments, the ionic species of the HMG glucoside salt act in conjunction with other ionic tastants to modify a sensory impression of said tastants. In certain embodiments, the HMG glucoside compound can be combined with a salt or salt mixture. The salt or salt mixture can comprise inorganic, organic, monoatomic as well as polyatomic ions. In certain embodiments, the salts are nontoxic and edible. In certain embodiments, the salt or salt mixtures are inorganic salts, for example, inorganic salts comprising halogen anions or phosphate ions, alkali or earth alkali metal salts. In certain embodiments, the salts are cationic salts such as, but not limited to, NaCl, KCI and Na3PO4. In certain embodiments, the salts are anionic salts such as, but not limited to acetate salt, TFA salt, and formate salt. 3. Flavor Compositions The flavor compositions of the present application can be used to enhance or modify the sensory experience of various edible compositions such as sweet goods and savory goods. The flavor compositions can include combinations of compounds, and can be added to edible compositions in various delivery system formats. In certain embodiments, the application relates to methods for modulating the texture of an edible product comprising: a) providing at least one comestible food product, or a precursor thereof, and b) combining the comestible food product or precursor thereof with at least a mouth watering, lubricating, slippery and/or astringent modulating amount of at least one flavor composition, for example, one or more HMG glucoside compound of Formula I, or a comestibly acceptable salt thereof, so as to form a modified edible food product. In certain embodiments, the flavor compositions of the present application can enhance the mouth watering, lubricating, slippery and/or astringent texture of a food product, such as, for example, an edible composition including pet foods, pharmaceutical compositions and human foods, such as soup, a confection, and/or a snack food. In certain embodiments, the flavor compositions of the present application can be used to modify, enhance or decrease the mouth watering, lubricating, slippery and/or astringent texture of one or more of the following subgenuses of comestible compositions: confectioneries, bakery products, ice creams, dairy products, savory snacks, snack bars, meal replacement products, ready meals, soups, pastas, noodles, canned foods, frozen foods, dried foods, chilled foods, oils and fats, baby foods, or spreads, or a mixture thereof. In certain embodiments of the application, an edible composition can be produced that contains a sufficient amount of at least one flavor composition, or its various subgenuses described herein, for example an HMG glucoside compound, for example, a compound of Formula I, to produce a composition having the desired flavor, taste and/or mouthfeel characteristics such as “mouth watering” and/or “lubricating” and/or “slippery” and/or “astringent” characteristic. In certain embodiments of the application, an edible composition can be produced that contains a sufficient amount of at least one flavor composition, or its various subgenuses described herein, for example an HMG glucoside compound, for example a compound of Formula I, to produce a composition having the desired texture characteristics such as a “mouth watering” texture. In certain embodiments, at least a mouth watering texture modulating amount of one or more of the flavor compositions of the present application can be added to the edible food product, so that the mouth watering texture modified edible food product has an increased or decreased mouth watering texture as compared to the edible food product prepared without the flavor composition, as determined by human beings or animals in general, or in the case of formulation testing, as determined by a taste panel of at least five human taste testers, via procedures known in the art. In certain embodiments of the present application, the flavor composition is added to a food product in an amount effective to provide a mouth watering texture. In certain embodiments of the application, an edible composition can be produced that contains a sufficient amount of at least one flavor composition, or its various subgenuses described herein, for example an HMG glucoside compound, for example a compound of Formula I, to produce a composition having the desired texture characteristics such as a “lubricating” texture. In certain embodiments, at least a lubricating texture modulating amount of one or more of the flavor compositions of the present application can be added to the edible food product, so that the lubricating texture modified edible food product has an increased or decreased lubrication texture as compared to the edible food product prepared without the flavor composition, as determined by human beings or animals in general, or in the case of formulation testing, as determined by a taste panel of at least five human taste testers, via procedures known in the art. In certain embodiments of the present application, the flavor composition is added to a food product in an amount effective to provide a lubricating texture. In certain embodiments of the application, an edible composition can be produced that contains a sufficient amount of at least one flavor composition, or its various subgenuses described herein, for example an HMG glucoside compound, for example a compound of Formula I, to produce a composition having the desired texture characteristics such as a “slippery” texture. In certain embodiments, at least a slippery texture modulating amount of one or more of the flavor compositions of the present application can be added to the edible food product, so that the slippery texture modified edible food product has an increased or decreased slippery texture as compared to the edible food product prepared without the flavor composition, as determined by human beings or animals in general, or in the case of formulation testing, as determined by a taste panel of at least five human taste testers, via procedures known in the art. In certain embodiments of the present application, the flavor composition is added to a food product in an amount effective to provide a slippery texture. In certain embodiments of the application, an edible composition can be produced that contains a sufficient amount of at least one flavor composition, or its various subgenuses described herein, for example an HMG glucoside compound, for example a compound of Formula I, to produce a composition having the desired texture characteristics such as a “astringent” texture. In certain embodiments, at least an astringent texture modulating amount of one or more of the flavor compositions of the present application can be added to the edible food product, so that the astringent texture modified edible food product has an increased or decreased astringent texture as compared to the edible food product prepared without the flavor composition, as determined by human beings or animals in general, or in the case of formulation testing, as determined by a taste panel of at least five human taste testers, via procedures known in the art. In certain embodiments of the present application, the flavor composition is added to a food product in an amount effective to provide a astringent texture. In certain embodiments, the flavor composition, or any of its subgenuses, for example, an HMG glucoside, for example, a compound of Formula I, or a comestibly acceptable salt thereof, of the present application, can be combined with an edible composition in an amount effective to modify, enhance or otherwise alter a taste or taste profile of the edible composition. The modification can include, for example, an increase or decrease in one or more of a sweet, sour, salty, bitter, kokumi and/or umami taste of the composition. In certain embodiments, the flavor composition, or any of its subgenuses, for example, an HMG glucoside, for example, a compound of Formula I, or a comestibly acceptable salt thereof, of the present application, can be combined with an edible composition in an amount effective to modify, enhance or otherwise alter a flavor or flavor profile of the edible composition. The modification can include, for example, an increase or decrease in the perception of one or more sensory stimuli, such as, for example, one or more of taste (gustatory), smell (olfactory), touch (tactile) and temperature (thermal). In certain embodiments, the flavor composition, or any of its subgenuses, for example, an HMG glucoside, for example, a compound of Formula I, or a comestibly acceptable salt thereof, of the present application, can be combined with an edible composition in an amount effective to modify, enhance or otherwise alter a texture profile of the edible composition. The concentration of flavor composition admixed with an edible food product to modulate or improve the flavor of the edible food product or composition can vary dependent on variables, such as, for example, the specific type of edible composition, what mouth watering, lubricating, slippery and/or astringent compounds are already present in the edible food product and the concentrations thereof, and the enhancer effect of the particular flavor composition on such mouth watering, lubricating, slippery and/or astringent compounds. In certain embodiments, admixing the flavor compositions of the present application with an edible food product modulates, for example, induces, enhances or inhibits, the mouth watering, lubricating, slippery and/or astringent (or other taste or flavor properties) of other natural or synthetic mouth watering, lubricating, slippery and/or astringent flavorants. A broad range of concentrations of the flavor compositions can be employed to provide such mouth watering, lubricating, slippery and/or astringent texture modification. In certain embodiments of the present application, the flavor composition is admixed with a food product wherein the flavor composition is present in an amount of from about 0.001 to about 500 ppb, or from about 0.005 to about 250 ppb, or from about 0.01 to about 200 ppb, or from about 0.05 to about 150 ppb, or from about 0.1 to about 100 ppb, or from about 0.5 to about 50 ppb. In certain embodiments of the present application, the flavor composition is admixed with a food product wherein the flavor composition is present in an amount of from between about 0.1 to about 100 ppb. In certain embodiments of the present application, the flavor composition is admixed with a food product wherein the flavor composition is present in an amount of from about 0.01 ppm to 5000 ppm, or narrower alternative ranges from about 0.1 ppm to about 1000 ppm, from about 0.5 ppm to about 500 ppm, from about 1 ppm to about 250 ppm, from about 5 ppm to about 200 ppm, from about 10 ppm to about 150 ppm, from about 10 ppm to about 100 ppm, or from about 20 ppm to about 50 ppm. In certain embodiments of the present application, the flavor composition is admixed with a food product wherein the flavor composition is present in an amount of from about 0.1 ppm to about 200 ppm, or from about 1 ppm and about 150 ppm. In certain embodiments of the present application, the flavor composition is admixed with a food product wherein the flavor composition is present in an amount of 100 ppm. In certain embodiments of the present application, the flavor composition is admixed with a food product wherein the flavor composition is present in an amount of from about 0.0000001 to about 99.999% weight/weight (w/w), or from about 0.00005 to about 75% w/w, or from about 0.0001 to about 50% w/w, or from about 0.0005 to about 25% w/w, or from about 0.001 to about 10% w/w, or from about 0.005 to about 5% w/w of the food product. In certain embodiments, the HMG glucoside compounds of the present application are blended together in various ratios or are blended together with other compounds to form various flavor compositions. In certain embodiments, the HMG glucoside compounds that are blended are cone or more compounds of Formula I. In certain embodiments, the HMG glucoside compounds and other compounds are blended together, wherein each of the HMG glucoside compounds and other compounds are present in an amount of from about 0.0000001 to about 99.999% weight/weight (w/w), or from about 0.00005 to about 75% w/w, or from about 0.0001 to about 50% w/w, or from about 0.0005 to about 25% w/w, or from about 0.001 to about 10% w/w, or from about 0.005 to about 5% w/w of the flavor composition. In certain embodiments, the HMG glucoside compounds that are blended together in various ratios or are blended together with other compounds to form various flavor compositions, are, for example, compounds of Formula I of the present application. In certain embodiments, the flavor composition comprises one or more HMG glucoside compound in combination with one or more additional compound with similar solubilities as the HMG glucoside compounds. Table 1 below provides non-limiting examples of flavor compositions comprising HMG glucosides, such as a compound of Formula I, in combination with other additional compounds. TABLE 1Flavor CompositionsFl. 1Fl. 2Fl. 3Fl. 4Fl. 5Fl. 6Fl. 7Fl. 8Ingredient% w/w% w/w% w/w% w/w% w/w% w/w% w/w% w/wFormula I0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Formula II0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Formula IIA0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Formula II-B0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Formula II-C0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Formula II-D0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Formula III0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Formula IV0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Formula V0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Hydrolyzed cocoa0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999powderHydrolyzed wheat0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999proteinHydrolyzed soy0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999proteinVanilla Extract0-99.9990-99.9990-99.9990-99.9990-99.99910-180-99.9990-99.999Ethyl vanillin0-99.9990-99.9990-99.9990-99.99912-160-99.9990-99.9990-99.999Ethyl maltol0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Isoamyl acetate0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9992-30-99.999Ethyl acetate0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Furaneol0-99.9990-99.9990-99.9990-99.9995-80-99.9990-99.9990-99.999Myrcene0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9991-20-99.999Linalool0-99.9990-99.9990-99.9990-99.9990-99.9991-30-99.9990-99.999Citral0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Geraniol0-99.9990-99.9990-99.9990-99.9990-99.9991-30-99.9990-99.999NaCl99.5-99.99950-99.9992.5-50-99.9990-99.9990-99.9990-99.9990-99.999KCl0-99.99999.5-99.99950-99.9992.5-50-99.9990-99.9990-99.9990-99.999Garlic flavor0-99.9990-99.99914-180-99.9990-99.9990-99.9990-99.9990-99.999Onion flavor0-99.9990-99.9990-99.99912-150-99.9990-99.9990-99.9990-99.999Beef Flavor0-99.9990-99.99970-800-99.9990-99.9990-99.9990-99.9990-99.999Chicken flavor0-99.9990-99.9990-99.99965-750-99.9990-99.9990-99.9990-99.999Acetic acid0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999Butyric acid0-99.9990-99.9990-99.9990-99.9990-99.9996-80-99.9990-99.999Citric acid0-99.9990-99.9990-99.9990-99.9990-99.9990-99.99995-9842-48Lactic acid0-99.9990-99.9990-99.9990-99.99950-6570-800-99.9990-99.999Malic acid0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.99928-32Tartaric acid0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.99920-25Other base flavor0-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.9990-99.999compounds 4. Delivery Systems In certain embodiments, the flavor compositions of the present application can be incorporated into a delivery system for use in edible compositions. In certain embodiments, the composition will comprise another flavor, taste or texture modifier such as a mouth watering, lubricating, slippery and/or astringent flavorant. Delivery systems can be liquid or solid, aqueous or non-aqueous. Delivery systems are generally adapted to suit the needs of the flavor composition and/or the edible composition into which the flavor composition will be incorporated. The flavoring compositions can be employed in liquid form, dried form, and/or solid form. When used in dried form, suitable drying means such as spray drying can be used. Alternatively, a flavoring composition can be encapsulated or absorbed onto water soluble materials, including but not limited to materials such as cellulose, starch, sugar, maltodextrin, gum arabic and so forth. The actual techniques for preparing such dried forms are well-known in the art, and can be applied to the presently disclosed subject matter. The flavoring compositions of the presently disclosed subject matter can be used in many distinct physical forms well known in the art to provide an initial burst of taste, flavor and/or texture; and/or a prolonged sensation of taste, flavor and/or texture. Without being limited thereto, such physical forms include free forms, such as spray dried, powdered, and beaded forms, and encapsulated forms, and mixtures thereof. In specific embodiments, as noted above, encapsulation techniques can be used to modify the flavor systems. In certain embodiments, flavor compounds, flavor components, or the entire flavor system can be fully or partially encapsulated. Encapsulating materials and/or techniques can be selected to determine the type of modification of the flavor system. In specific embodiments, the encapsulating materials and/or techniques are selected to improve the stability of the flavor compounds, flavor components, or flavor systems; while in other embodiments the encapsulating materials and/or techniques are selected to modify the release profile of the flavor compounds, flavor components, or flavor systems. Suitable encapsulating materials can include, but are not limited to, hydrocolloids such as alginates, pectins, agars, guar gums, celluloses, and the like, proteins, polyvinyl acetate, polyethylene, crosslinked polyvinyl pyrrolidone, polymethylmethacrylate, polylactidacid, polyhydroxyalkanoates, ethylcellulose, polyvinyl acetatephthalate, polyethylene glycol esters, methacrylicacid-co-methylmethacrylate, ethylene-vinylacetate (EVA) copolymer, and the like, and combinations thereof. Suitable encapsulating techniques can include, but are not limited to, spray coating, spray drying, spray chilling, absorption, adsorption, inclusion complexing (e.g., creating a flavor/cyclodextrin complex), coacervation, fluidized bed coating, or other process can be used to encapsulate an ingredient with an encapsulating material. Encapsulated delivery systems for flavoring agents or sweetening agents contain a hydrophobic matrix of fat or wax surrounding a sweetening agent or flavoring agent core. The fats can be selected from any number of conventional materials such as fatty acids, glycerides or poly glycerol esters, sorbitol esters, and mixtures thereof. Examples of fatty acids include but are not limited to hydrogenated and partially hydrogenated vegetable oils such as palm oil, palm kernel oil, peanut oil, rapeseed oil, rice bran oil, soybean oil, cottonseed oil, sunflower oil, safflower oil, and mixtures thereof. Examples of glycerides include but are not limited to monoglycerides, diglycerides, and triglycerides. Waxes useful can be chosen from the group consisting of natural and synthetic waxes, and mixtures thereof. Non-limiting examples include paraffin wax, petrolatum, carbowax, microcrystalline wax, beeswax, carnauba wax, candelilla wax, lanolin, bayberry wax, sugarcane wax, spermaceti wax, rice bran wax, and mixtures thereof. The fats and waxes can be use individually or in combination in amounts varying from about 10 to about 70%, and alternatively in amounts from about 30 to about 60%, by weight of the encapsulated system. When used in combination, the fat and wax are preferably present in a ratio from about 70:10 to 85:15, respectively. Typical encapsulated flavor compositions, flavoring agent or sweetening agent delivery systems are disclosed in U.S. Pat. Nos. 4,597,970 and 4,722,845, the disclosures of which are incorporated herein by reference in their entireties. Liquid delivery systems can include, but are not limited to, systems with a dispersion of HMG glucoside compound(s) or the flavor compositions of the present application, such as in carbohydrate syrups and/or emulsions. Liquid delivery systems can also include extracts where the HMG glucoside compound(s) and/or the flavor compositions are solubilized in a solvent. Solid delivery systems can be created by spray drying, spray coating, spray chilling, fluidized bed drying, absorption, adsorption, coacervation, complexation, or any other standard technique. In some embodiments, the delivery system can be selected to be compatible with or to function in the edible composition. In some embodiments, the delivery system will include an oleaginous material such as a fat or oil. In some embodiments, the delivery system will include a confectionery fat such as cocoa butter, a cocoa butter replacer, a cocoa butter substitute, or a cocoa butter equivalent. When used in dried form, suitable drying means such as spray drying may be used. Alternatively, a flavoring composition may be adsorbed or absorbed onto substrates such as water soluble materials, such as cellulose, starch, sugar, maltodextrin, gum arabic and so forth or may be encapsulated. The actual techniques for preparing such dried forms are well known in the art. 5. End Product Systems The flavoring compositions of the present disclosed subject matter can be used in a wide variety of ingestible vehicles. Non-limiting examples of suitable ingestible vehicles include chewing gum compositions, hard and soft confections, dairy products, beverage products including juice products and soft drinks, pharmaceuticals, bakery goods, frozen foods, food products and food categories described herein. The combination of the flavoring composition of the presently disclosed subject matter together with an ingestible vehicle and optional ingredients, when desired, provides a flavoring agent that possesses unexpected taste, flavor and/or texture value and imparts, for example, a mouth watering, lubricating, slippery and/or astringent sensory experience. In the method for flavoring an ingestible composition of the presently disclosed subject matter, the ingestible composition is prepared by admixing the flavoring agent in an ingestible vehicle, together with any optional ingredients, to form a uniform mixture. The final compositions are readily prepared using standard methods and apparatus generally known by those skilled in the corresponding arts, such as confectionary arts. The apparatus useful in accordance with the presently disclosed subject matter comprises mixing apparatus well known in the art, and therefore the selection of the specific apparatus will be apparent to the artisan. In certain embodiments, the present application relates to the modified edible food products produced by the methods disclosed herein. In certain embodiments, the food products can be produced by processes for producing comestible products well known to those of ordinary skill in the art, wherein the flavor composition of the present application is employed as a mouth watering, lubricating, slippery and/or astringent texture enhancer for the food product. The flavor composition and its various subgenuses can be combined with or applied to a comestible or medicinal products or precursor thereof in any of innumerable ways known to cooks the world over, or producers of comestible or medicinal products. For example, the flavor compositions can be dissolved in or dispersed in one of many known comestibly acceptable liquids, solids, or other carriers, such as water at neutral, acidic, or basic pH, fruit or vegetable juices, vinegar, marinades, beer, wine, natural water/fat emulsions such as milk or condensed milk, whey or whey products, edible oils and shortenings, fatty acids, certain low molecular weight oligomers of propylene glycol, glyceryl esters of fatty acids, and dispersions or emulsions of such hydrophobic substances in aqueous media, salts such as sodium chloride, vegetable flours, solvents such as ethanol, solid edible diluents such as vegetable powders or flours, and the like, and then combined with precursors of the comestible or medicinal products, or applied directly to the comestible or medicinal products. In certain embodiments, the flavor compositions of the present application can be admixed with foods, beverages and other comestible compositions wherein savory compounds, especially NaCl, MSG, inosine monophosphate (IMP), or guanosine monophosphate (GMP) are conventionally utilized. These compositions include compositions for human and animal consumption, for example, food or drinks (liquids) for consumption by agricultural animals, pets and zoo animals. Those of ordinary skill in the art of preparing and selling comestible compositions (i.e., edible foods or beverages, or precursors or flavor modifiers thereof) are well aware of a large variety of classes, subclasses and species of the comestible compositions, and utilize well-known and recognized terms of art to refer to those comestible compositions while endeavoring to prepare and sell various of those comestible compositions. Such a list of terms of art is enumerated below, and it is specifically contemplated hereby that the flavor compositions of the present application can be used to modify or enhance the mouth watering, lubricating, slippery and/or astringent mouthfeel of the following list edible compositions, either singly or in all reasonable combinations or mixtures thereof. In certain embodiments, the food products to which the flavor compositions of the present application are admixed with comprise, by way of example, the wet soup category, the dehydrated and culinary food category, the beverage category, the frozen food category, the snack food category, and seasonings or seasoning blends, described herein. In other embodiments, the flavor compositions of the present application are admixed with one or more confectioneries, chocolate confectionery, tablets, countlines, bagged selfmies/softlines, boxed assortments, standard boxed assortments, twist wrapped miniatures, seasonal chocolate, chocolate with toys, allsorts, other chocolate confectionery, mints, standard mints, power mints, boiled sweets, pastilles, gums, jellies and chews, toffees, caramels and nougat, medicated confectionery, lollipops, liquorice, other sugar confectionery, gum, chewing gum, sugarised gum, sugar-free gum, functional gum, bubble gum, bread, packaged/industrial bread, unpackaged/artisanal bread, pastries, cakes, packaged/industrial cakes, unpackaged/artisanal cakes, cookies, chocolate coated biscuits, sandwich biscuits, filled biscuits, savory biscuits and crackers, bread substitutes, breakfast cereals, rte cereals, family breakfast cereals, flakes, muesli, other rte cereals, children's breakfast cereals, hot cereals, ice cream, impulse ice cream, single portion dairy ice cream, single portion water ice cream, multi-pack dairy ice cream, multi-pack water ice cream, take-home ice cream, take-home dairy ice cream, ice cream desserts, bulk ice cream, take-home water ice cream, frozen yoghurt, artisanal ice cream, dairy products, milk, fresh/pasteurized milk, full fat fresh/pasteurized milk, semi skimmed fresh/pasteurized milk, long-life/uht milk, full fat long life/uht milk, semi skimmed long life/uht milk, fat-free long life/uht milk, goat milk, condensed/evaporated milk, plain condensed/evaporated milk, flavored, functional and other condensed milk, flavored milk drinks, dairy only flavored milk drinks, flavored milk drinks with fruit juice, soy milk, sour milk drinks, fermented dairy drinks, coffee whiteners, powder milk, flavored powder milk drinks, cream, cheese, processed cheese, spreadable processed cheese, unspreadable processed cheese, unprocessed cheese, spreadable unprocessed cheese, hard cheese, packaged hard cheese, unpackaged hard cheese, yoghurt, plain/natural yoghurt, flavored yoghurt, fruited yoghurt, probiotic yoghurt, drinking yoghurt, regular drinking yoghurt, probiotic drinking yoghurt, chilled and shelf-stable desserts, dairy-based desserts, soy-based desserts, chilled snacks, fromage frais and quark, plain fromage frais and quark, flavored fromage frais and quark, savory fromage frais and quark, sweet and savory snacks, fruit snacks, chips/crisps, extruded snacks, tortilla/corn chips, popcorn, pretzels, nuts, other sweet and savory snacks, snack bars, granola bars, breakfast bars, energy bars, fruit bars, other snack bars, meal replacement products, slimming products, convalescence drinks, ready meals, canned ready meals, frozen ready meals, dried ready meals, chilled ready meals, dinner mixes, frozen pizza, chilled pizza, soup, canned soup, dehydrated soup, instant soup, chilled soup, uht soup, frozen soup, pasta, canned pasta, dried pasta, chilled/fresh pasta, noodles, plain noodles, instant noodles, cups/bowl instant noodles, pouch instant noodles, chilled noodles, snack noodles, canned food, canned meat and meat products, canned fish/seafood, canned vegetables, canned tomatoes, canned beans, canned fruit, canned ready meals, canned soup, canned pasta, other canned foods, frozen food, frozen processed red meat, frozen processed poultry, frozen processed fish/seafood, frozen processed vegetables, frozen meat substitutes, frozen potatoes, oven baked potato chips, other oven baked potato products, non-oven frozen potatoes, frozen bakery products, frozen desserts, frozen ready meals, frozen pizza, frozen soup, frozen noodles, other frozen food, dried food, dessert mixes, dried ready meals, dehydrated soup, instant soup, dried pasta, plain noodles, instant noodles, cups/bowl instant noodles, pouch instant noodles, chilled food, chilled processed meats, chilled fish/seafood products, chilled processed fish, chilled coated fish, chilled smoked fish, chilled lunch kit, chilled ready meals, chilled pizza, chilled soup, chilled/fresh pasta, chilled noodles, oils and fats, olive oil, vegetable and Seed oil, cooking fats, butter, margarine, spreadable oils and fats, functional spreadable oils and fats, sauces, dressings and condiments, tomato pastes and purees, bouillon/stock cubes, stock cubes, gravy granules, liquid stocks and fonds, herbs and spices, fermented sauces, soy based sauces, pasta sauces, wet sauces, dry sauces/powder mixes, ketchup, mayonnaise, regular mayonnaise, mustard, salad dressings, regular salad dressings, low fat salad dressings, vinaigrettes, dips, pickled products, other sauces, dressings and condiments, baby food, milk formula, standard milk formula, follow-on milk formula, toddler milk formula, hypoallergenic milk formula, prepared baby food, dried baby food, other baby food, spreads, jams and preserves, honey, chocolate spreads, nut-based spreads, and yeast-based spreads. 5.1 Sweet Goods 5.1.1 Chewing Gum The flavor systems can be used in sugarless gum formulations and can also be used in a sugar chewing gum. The flavor systems can be used in either regular chewing gum or bubble gum. Various specifics of chewing gum compositions are disclosed in U.S. Pat. No. 6,899,911, the disclosure of which is incorporated herein by reference in its entirety. The chewing gum composition of the presently disclosed subject matter follows the general pattern outlined below. In general, a chewing gum composition typically contain a chewable gum base portion which is essentially free of water and is water-insoluble, a water-soluble bulk portion and flavors which are typically water insoluble. The water-soluble portion dissipates with a portion of the flavor over a period of time during chewing. The gum base portion is retained in the mouth throughout the chew. The insoluble gum base generally comprises elastomers, elastomer solvents, plasticizers, waxes, emulsifiers and inorganic fillers. Plastic polymers, such as polyvinyl acetate, which behave somewhat as plasticizers, are also often included. Other plastic polymers that can be used include polyvinyl laureate, polyvinyl alcohol and polyvinyl pyrrolidone. Elastomers can include polyisobutylene, butyl rubber, (isobutylene-isoprene copolymer) and styrene butadiene rubber, as well as natural latexes such as chicle. Elastomer solvents are often resins such as terpene resins. Plasticizers, sometimes called softeners, are typically fats and oils, including tallow, hydrogenated and partially hydrogenated vegetable oils, and cocoa butter. Commonly employed waxes include paraffin, microcrystalline and natural waxes such as beeswax and carnauba. Microcrystalline waxes, especially those with a high degree of crystallinity, can be considered bodying agents or textural modifiers. According to the preferred embodiment of the presently disclosed subject matter, the insoluble gum base constitutes between about 5% to about 95% by weight of the gum. More preferably the insoluble gum base comprises between 10% and 50% by weight of the gum and most preferably about 20% to 35% by weight of the gum. The gum base typically also includes a filler component. The filler component can be calcium carbonate, magnesium carbonate, talc, dicalcium phosphate or the like. The filler can constitute between about 5% and about 60% by weight of the gum base. Preferably the filler comprises about 5% to 50% by weight of the gum base. Gum bases typically also contain softeners including glycerol monostearate and glycerol triacetate. Gum bases can also contain optional ingredients such as antioxidants, colors, and emulsifiers. The presently disclosed subject matter contemplates employing any commercially acceptable gum base. The water-soluble portion of the chewing gum can further comprise softeners, sweeteners, flavors, physiological cooling agents and combinations thereof. The sweeteners often fulfill the role of bulking agents in the gum. The bulking agents typically comprise about 5% to about 95% of the gum composition. Softeners are added to the chewing gum in order to optimize the chewability and mouth feel of the gum. Softeners, also known in the art as plasticizers or plasticizing agents, generally constitute between about 0.5% to about 15% of the chewing gum. Softeners contemplated by the presently disclosed subject matter include glycerin, lecithin and combinations thereof. Further, aqueous sweetener solutions such as those containing sorbitol, hydrogenated starch hydrolysate, corn syrup and combinations thereof can be used as softeners and binding agents in gum. As mentioned above, the flavor systems of the presently disclosed subject matter can be used in sugarless gum formulations. However, formulations containing sugar are also within the scope of the invention. Sugar sweeteners generally include saccharide-containing components commonly known in the chewing gum art which comprise, but are not limited to, sucrose, dextrose, maltose, dextrin, dried invert sugar, fructose, galactose, corn syrup solids and the like, alone or in any combination. The flavor systems of the presently disclosed subject matter can also be used in combination with sugarless sweeteners. Generally sugarless sweeteners include components with sweetening characteristics but which are devoid of the commonly known sugars and comprise, but are not limited to, sugar alcohols such as sorbitol, hydrogenated isomaltulose, mannitol, xylitol, lactitol, erythritol, hydrogenated starch hydrolysate, maltitol and the like alone or in any combination Depending on the particular sweetness release profile and shelf-stability needed, coated or uncoated high-intensity sweeteners can be used in the chewing gum composition, or can be used in a coating applied to centers made from those gum compositions. High-intensity sweeteners, preferably aspartame, can be used at levels from about 0.01% to about 3.0%. Encapsulated aspartame is a high intensity sweetener with improved stability and release characteristics, as compared to free aspartame. Free aspartame can also be added, and a combination of some free and encapsulated aspartame is preferred when aspartame is used. Other high intensity sweeteners that can be used in the gum center are: saccharin, Thaumatin, alitame, saccharin salts, sucralose, Stevia, and acesulfame K. Overall, the chewing gum composition will preferable comprise about 0.5% to about 90% sweetening agents. Most typically the sweetening agents will comprises at least one bulk sweetener and at least one high-intensity sweetener. Optional ingredients such as colors, emulsifiers and pharmaceutical agents can also be added as separate components of the chewing gum composition, or added as part of the gum base. Aqueous syrups, such as corn syrup and hydrogenated corn syrup can be used, particularly if their moisture content is reduced. This can preferably be done by coevaporating the aqueous syrup with a plasticizer, such as glycerin or propylene glycol, to a moisture content of less than 10%. Preferred compositions include hydrogenated starch hydrolysate solids and glycerin. Such syrups and their methods of preparation are discussed in detail in U.S. Pat. No. 4,671,967. A preferred method of manufacturing chewing gum according to the presently disclosed subject matter is by sequentially adding the various chewing gum ingredients to any commercially available mixer known in the art. After the ingredients have been thoroughly mixed, the gum is discharged from the mixer and shaped into the desired form such as by rolling into sheets and cutting into sticks, extruding into chunks, or casting into pellets. Generally, the ingredients are mixed by first melting the gum base and adding it to the running mixer. The base can also be melted in the mixer itself. Color or emulsifiers can also be added at this time, along with syrup and a portion of the bulking agent. Further portions of the bulking agent can then be added to the mixer. Flavor systems are typically added with the final portion of the bulking agent. If the flavor system is coated or otherwise modified as when incorporated into a delivery system to modify its release rate, it will preferably be added after the final portion of bulking agent has been added. The entire mixing procedure typically takes from five to twenty minutes, but longer mixing times can sometime be required. Those skilled in the art will recognize that many variations of the above described procedures can be followed. If formed into pellets or balls, the chewing gum composition can be coated. The coating is initially present as a liquid syrup which contains from about 30% to about 80% or 85% sugars or sugar alcohols, and from about 15% or 20% to about 70% of a solvent such as water. In general, the coating process is carried out in conventional panning equipment. Gum center tablets to be coated are placed into the panning equipment to form a moving mass. The material or syrup which will eventually form the coating is applied or distributed over the gum center tablets. The flavor systems of the presently disclosed subject matter can be added before, during and after applying the syrup to the gum centers. Once the coating has dried to form a hard surface, additional syrup additions can be made to produce a plurality of coatings or multiple layers of coating. The flavor systems can be added to any or none of the coatings and/or layers. In the panning procedure, syrup is added to the gum center tablets at a temperature range of from about 100° F. to about 240° F. Preferably, the syrup temperature is from about 140° F. to about 200° F. Most preferably, the syrup temperature should be kept constant throughout the process in order to prevent the polyol in the syrup from crystallizing. The syrup can be mixed with, sprayed upon, poured over, or added to the gum center tablets in any way known to those skilled in the art. In another embodiment, a soft coating is formed by adding a powder coating after a liquid coating. The powder coating can include natural carbohydrate gum hydrolysates, maltodextrin, gelatin, cellulose derivatives, starches, modified starches, sugars, sugar alcohols, natural carbohydrate gums and fillers like talc and calcium carbonate. Each component of the coating on the gum center can be applied in a single layer or in a plurality of layers. In general, a plurality of layers is obtained by applying single coats, allowing the layers to dry, and then repeating the process. The amount of solids added by each coating step depends chiefly on the concentration of the coating syrup. Any number of coats can be applied to the gum center tablet. Preferably, no more than about 75 coats are applied to the gum center. More preferably, less than about 60 coats are applied and most preferably, about 30 to about 60 coats are applied. In any event, the presently disclosed subject matter contemplates applying an amount of syrup sufficient to yield a coated chewing gum product containing about 10% to about 65% coating. Preferably, the final product will contain from about 20% to about 50% coating. Those skilled in the art will recognize that in order to obtain a plurality of coated layers, a plurality of premeasured aliquots of coating syrup can be applied to the gum center. It is contemplated, however, that the volume of aliquots of syrup applied to the gum center can vary throughout the coating procedure. Once a coating of syrup is applied to the gum center, the syrup is dried in an inert medium. A preferred drying medium comprises air. Preferably, forced drying air contacts the wet syrup coating in a temperature range of from about 70° F. to about 110° F. More preferably, the drying air is in the temperature range of from about 80° F. to about 100° F. The invention also contemplates that the drying air possesses a relative humidity of less than about 15 percent. Preferably, the relative humidity of the drying air is less than about 8%. The drying air can be passed over and admixed with the syrup coated gum centers in any way commonly known in the art. Preferably, the drying air is blown over and around the syrup coated gum center at a flow rate, for large scale operations, of about 2800 cubic feet per minute. If lower quantities of material are being processed, or if smaller equipment is used, lower flow rates would be used. If a flavor is applied after a syrup coating has been dried, the presently disclosed subject matter contemplates drying the flavor with or without the use of a drying medium. The amount of flavoring agent employed herein is normally a matter of preference subject to such factors as the type of final chewing gum composition, the individual flavor, the gum base employed, and the strength of flavor desired. Thus, the amount of flavoring can be varied in order to obtain the result desired in the final product and such variations are within the capabilities of those skilled in the art without the need for undue experimentation. In gum compositions, the flavoring agent is generally present in amounts from about 0.02% to about 5%, and preferably from about 0.1% to about 2%, and more preferably, from about 0.8% to about 1.8%, by weight of the chewing gum composition. 5.1.2 Sugar Confectionary Another important aspect of the presently disclosed subject matter includes a confectionery composition incorporating the inventive flavoring agent and a method for preparing the confectionery compositions. The preparation of confectionery formulations is well-known in the art. Confectionery items have been classified as either “hard” confectionery or “soft” confectionery. The flavoring agents of the presently disclosed subject matter can be incorporated into the confections by admixing the compositions of the presently disclosed subject matter into the conventional hard and soft confections. Hard confectionery can be processed and formulated by conventional means. In general, a hard confectionery has a base composed of a mixture of sugar and other carbohydrate bulking agents kept in an amorphous or glassy condition. The hard confectionery can also be sugarless. This form is considered a solid syrup of sugars generally having from about 0.5% to about 1.5% moisture. Such materials normally contain up to about 92% sugar, up to about 55% corn syrup and from about 0.1% to about 5% water, by weight of the final composition. The syrup component is generally prepared from sucrose and corn syrups, but can include other materials. Further ingredients such as flavorings, sweetening agents, acidulants, colorants and so forth can also be added. Such confectionery can be routinely prepared by conventional methods, including but not limited to methods involving fire cookers, vacuum cookers, and scraped-surface cookers also referred to as high speed atmospheric cookers. The apparatus useful in accordance with the presently disclosed subject matter comprises cooking and mixing apparatus well known in the confectionery manufacturing arts, and therefore the selection of the specific apparatus will be apparent to the artisan. Fire cookers involve the traditional method of making a candy base. In this method, the desired quantity of carbohydrate bulking agent is dissolved in water by heating the agent in a kettle until the bulking agent dissolves. Additional bulking agent can then be added and cooked until a final temperature of 145° C. to 156° C. is achieved. The batch is then cooled and worked as a plastic-like mass to incorporate additives such as flavoring agent, colorants and the like. A high-speed atmospheric cooker uses a heat-exchanger surface, which involves spreading a film of candy on a heat exchange surface, the candy is heated to 165° C. to 170° C. within a few seconds. The candy is then rapidly cooled to 100° C. to 120° C. and worked as a plastic-like mass enabling incorporation of the additives, such as flavoring agent, colorants and the like. In vacuum cookers, the carbohydrate bulking agent is boiled to 125° C. to 132° C., vacuum is applied and additional water is boiled off without extra heating. When cooking is complete, the mass is a semi-solid and has a plastic-like consistency. At this point, flavoring agent, colorants, and other additives are admixed in the mass by routine mechanical mixing operations. The optimum mixing required to uniformly mix the flavoring agent, colorants and other additives during conventional manufacturing of hard confectionery is determined by the time needed to obtain a uniform distribution of the materials. Generally, mixing times of from 2 to 10 minutes have been found to be acceptable. Once the candy mass has been properly tempered, it can be cut into workable portions or formed into desired shapes. A variety of forming techniques can be utilized depending upon the shape and size of the final product desired. A general discussion of the composition and preparation of hard confections can be found in H. A. Lieberman, Pharmaceutical Dosage Forms: Tablets, Volume 1 (1989), Marcel Dekker, Inc., New York, N.Y. at pages 419 to 582, which disclosure is incorporated herein by reference. Compressed tablet confections contain particular materials and are formed into structures under pressure. These confections generally contain sugars in amounts up to about 95%, by weight of the composition, and typical tablet excipients such as binders and lubricants as well as flavoring agent, colorants and so forth. These confections can also be sugarless. Similar to hard confectionery, soft confectionery can be utilized in the embodiments of the disclosed subject matter. The preparation of soft confections, such as nougat, involves conventional methods, such as the combination of two primary components, namely (1) a high boiling syrup such as a corn syrup, or the like, and (2) a relatively light textured frappe, generally prepared from egg albumin, gum arabic, gelatin, vegetable proteins, such as soy derived compounds, sugarless milk derived compounds such as milk proteins, and mixtures thereof. The frappe is generally relatively light, and can, for example, range in density from about 0.5 to about 0.7 grams/cc. The high boiling syrup, or “bob syrup” of the soft confectionery is relatively viscous and has a higher density than the frappe component, and frequently contains a substantial amount of carbohydrate bulking agent. Conventionally, the final nougat composition is prepared by the addition of the “bob syrup” to the frappe under agitation, to form the basic nougat mixture. Further ingredients such as flavoring, additional carbohydrate bulking agent, colorants, preservatives, medicaments, mixtures thereof and the like can be added thereafter also under agitation. Soft confectioneries can also be prepared sugarless. A general discussion of the composition and preparation of nougat confections can be found in B. W. Minifie, Chocolate, Cocoa and Confectionery: Science and Technology, 2nd edition, AVI Publishing Co., Inc., Westport, Conn. (1983), at pages 576-580, which disclosure is incorporated herein by reference. In general, the frappe component is prepared first and thereafter the syrup component is slowly added under agitation at a temperature of at least about 65° C., and preferably at least about 100° C. The mixture of components is continued to be mixed to form a uniform mixture, after which the mixture is cooled to a temperature below 80° C., at which point, the flavor can be added. The mixture is further mixed for an additional period until it is ready to be removed and formed into suitable confectionery shapes. In accordance with this invention, effective amounts of the flavoring agents of the presently disclosed subject matter can be admixed into the hard and soft confections. The exact amount of flavoring agent employed is normally a matter of preference subject to such factors as the particular type of confection being prepared, the type of bulking agent or carrier employed, the type of flavor employed and the intensity of breath freshening perception desired. Thus, the amount of flavoring agent can be varied in order to obtain the result desired in the final product and such variations are within the capabilities of those skilled in the art without the need for undue experimentation. In general, the amount of flavoring agent normally present in a hard or soft confection will be from about 0.001% to about 20%, preferably from about 0.01% to about 15%, more preferably from about 0.01% to about 10%, and more preferably from about 0.01% to about 5%, and more preferably 0.01% to about 0.5% by weight of the confection. The presently disclosed subject matter extends to methods for making the improved confections. The flavoring agents can be incorporated into an otherwise conventional hard or soft confection composition using standard techniques and equipment known to those skilled in the art. The apparatus useful in accordance with the presently disclosed subject matter comprises mixing and heating apparatus well known in the confectionery manufacturing arts, and therefore the selection of the specific apparatus will be apparent to the artisan. In such a method, a composition is made by admixing the inventive flavoring agent into the confectionery composition along with the other ingredients of the final desired composition. Other ingredients will usually be incorporated into the composition as dictated by the nature of the desired composition as well known by those having ordinary skill in the art. The ultimate confectionery compositions are readily prepared using methods generally known in the food technology and pharmaceutical arts. Thereafter the confectionery mixture can be formed into desirable confectionery shapes. The flavoring agents can be formulated with conventional ingredients which offer a variety of textures to suit particular applications. Such ingredients can be in the form of hard and soft confections, tablets, toffee, nougat, chewy candy, chewing gum and so forth, center filled candies, both sugar and sugarless. The acceptable ingredients can be selected from a wide range of materials. Without being limited thereto, such materials include diluents, binders and adhesives, lubricants, disintegrants, bulking agents, humectants and buffers and adsorbents. The preparation of such confections and chewing gum products is well known. 5.1.3. Chocolates and Fillings The presently disclosed subject matter is also used with and/or in chocolate products, chocolate-flavored confections, and chocolate flavored compositions. Chocolates also include those containing crumb solids or solids fully or partially made by a crumb process. Various chocolates are disclosed, for example, in U.S. Pat. Nos. 7,968,140 and 8,263,168, the disclosures of which are incorporated herein by reference in their entireties. A general discussion of the composition and preparation of chocolate confections can be found in B. W. Minifie, Chocolate, Cocoa and Confectionery: Science and Technology, 2nd edition, AVI Publishing Co., Inc., Westport, Conn. (1982), which disclosure is incorporated herein by reference. The term “chocolate” as used herein refers to a solid or semi-plastic food and is intended to refer to all chocolate or chocolate-like compositions containing a fat-based component phase or fat-like composition. The term is intended to include standardized or nonstandardized compositions conforming to the U.S. Standards Of Identity (SOI), CODEX Alimentarius and/or other international standards and compositions not conforming to the U.S. Standards Of Identity or other international standards. The term includes dark chocolate, baking chocolate, sweet chocolate, bittersweet or semisweet chocolate, milk chocolate, buttermilk chocolate, skim milk chocolate, mixed dairy product chocolate, white chocolate, sweet cocoa and vegetable fat coating, sweet chocolate and vegetable fat coating, milk chocolate and vegetable fat coating, vegetable fat based coating, pastels including white chocolate or coating made with cocoa butter or vegetable fat or a combination of these, nutritionally modified chocolate-like compositions (chocolates or coatings made with reduced calorie ingredients) and low fat chocolates, aerated chocolates, compound coatings, non-standardized chocolates and chocolate-like compositions, unless specifically identified otherwise. Nonstandardized chocolates result when, for example, the nutritive carbohydrate sweetener is replaced partially or completely; or when the cocoa butter, cocoa butter alternative, cocoa butter equivalent, cocoa butter extender, cocoa butter replacer, cocoa butter substitute or milkfat are replaced partially or completely; or when components that have flavors that imitate milk, butter or chocolate are added or other additions or deletions in formula are made outside the FDA standards of identify of chocolate or combinations thereof. Chocolate-like compositions are those fat-based compositions that can be used as substitutes for chocolate in applications such as panning, molding, or enrobing; for example, carob. In the United States, chocolate is subject to a standard of identity established by the U.S. Food and Drug Administration (FDA) under the Federal Food, Drug and Cosmetic Act. Definitions and standards for the various types of chocolate are well established in the U.S. Nonstandardized chocolates are those chocolates which have compositions that fall outside the specified ranges of the standardized chocolates. The chocolate can contain a sugar syrup/solids, invert sugar, hydrolyzed lactose, maple sugar, brown sugar, molasses, honey, sugar substitute and the like. The term “sugar substitute” includes bulking agents, sugar alcohols (polyols such as glycerol), or high potency sweeteners or combinations thereof. Nutritive carbohydrate sweeteners with varying degrees of sweetness intensity can be any of those typically used in the art and include, but are not limited to, sucrose, e.g. from cane or beet, dextrose, fructose, lactose, maltose, glucose syrup solids, corn syrup solids, invert sugar, hydrolyzed lactose, honey, maple sugar, brown sugar, molasses and the like. Sugar substitutes can partially replace the nutritive carbohydrate sweetener. High potency sweeteners include aspartame, cyclamates, saccharin, acesulfame-K, neohesperidin dihydrochalcone, sucralose, alitame, stevia sweeteners, glycyrrhizin, thaumatin and the like and mixtures thereof. The preferred high potency sweeteners are aspartame, cyclamates, saccharin, and acesulfame-K. Examples of sugar alcohols can be any of those typically used in the art and include sorbitol, mannitol, xylitol, maltitol, isomalt, lactitol and the like. The chocolates can also contain bulking agents. The term “bulking agents” as defined herein can be any of those typically used in the art and include polydextrose, cellulose and its derivatives, maltodextrin, gum arabic, and the like. The chocolate products can contain emulsifiers. Examples of safe and suitable emulsifiers can be any of those typically used in the art and include lecithin derived from vegetable sources such as soybean, safflower, corn, etc., fractionated lecithins enriched in either phosphatidyl choline or phosphatidyl ethanolamine, or both, mono- and digylcerides, diacetyl tartaric acid esters of mono- and diglycerides (also referred to as DATEM), monosodium phosphate derivatives of mono- and diglycerides of edible fats or oils, sorbitan monostearate, hydroxylated lecithin, lactylated fatty acid esters of glycerol and propylene glycol, polyglycerol esters of fatty acids, propylene glycol mono- and di-esters of fats and fatty acids, or emulsifiers that can become approved for the US FDA-defined soft candy category. In addition, other emulsifiers that can be used include polyglycerol polyricinoleate (PGPR), ammonium salts of phosphatidic acid, (e.g. YN) sucrose esters, oat extract, etc., any emulsifier found to be suitable in chocolate or similar fat/solid system or any blend. The term “chocolate-flavored confection” refers to food products, excluding “chocolate”, having a chocolate flavor/aroma and comprising a cocoa fraction. These products are stable at ambient temperatures for extended periods of time (e.g., greater than 1 week) and are characterized as microbiologically shelf-stable at 18-30° C. under normal atmospheric conditions. Examples include chocolate-flavored hard candies, chewables, chewing gums, etc. The term “chocolate-flavored compositions” refers to chocolate-flavored compositions, excluding “chocolate”, containing a cocoa fraction and having a chocolate flavor/aroma. Examples include chocolate-flavored cake mixes, ice creams, syrups, baking goods, etc. The term includes chocolate-flavored compositions (e.g., cakes, nougats, puddings, etc.), as well as compositions not having a chocolate-flavor (e.g., caramels, etc.). 5.2 Savory Goods and Other Food Products In certain embodiments, the flavor compositions of the present application are incorporated into savory goods to impart, enhance, or modify a mouth watering, lubricating, slippery and/or astringent mouthfeel. In certain embodiments, a savory good is a food product that has savory flavors including, for example, but not limited to, spicy flavor, pepper flavor, dairy flavor, vegetable flavor, tomato flavor, dill flavor, meat flavor, poultry flavor, chicken flavor and reaction flavors that are added or generated during heating of a food product. In certain embodiments, the flavor compositions are incorporated into a wet soup category food product, which comprises wet/liquid soups regardless of concentration or container, including frozen soups. In certain embodiments, the a soup food product means a food prepared from meat, poultry, fish, vegetables, grains, fruit and/or other ingredients, cooked in a liquid which may include visible pieces of some or all of these ingredients. It may be clear (as a broth) or thick (as a chowder), smooth, pureed or chunky, ready-to-serve, semi-condensed or condensed and may be served hot or cold, as a first course or as the main course of a meal or as a between meal snack (sipped like a beverage). Soup may be used as an ingredient for preparing other meal components and may range from broths (consomme) to sauces (cream or cheese-based soups). In certain embodiments, the flavor compositions of the present application are incorporated into a dehydrated and culinary food category of food products, which comprises (i) cooking aid products such as: powders, granules, pastes, concentrated liquid products, including concentrated bouillon, bouillon and bouillon like products in pressed cubes, tablets or powder or granulated form, which are sold separately as a finished product or as an ingredient within a product, sauces and recipe mixes (regardless of technology); (ii) meal solutions products such as: dehydrated and freeze dried soups, including dehydrated soup mixes, dehydrated instant soups, dehydrated ready-to-cook soups, dehydrated or ambient preparations of ready-made dishes, meals and single serve entrees including pasta, potato and rice dishes; and (iii) meal embellishment products such as: condiments, marinades, salad dressings, salad toppings, dips, breading, batter mixes, shelf stable spreads, barbecue sauces, liquid recipe mixes, concentrates, sauces or sauce mixes, including recipe mixes for salad, sold as a finished product or as an ingredient within a product, whether dehydrated, liquid or frozen. In certain embodiments, the flavor compositions of the present application are incorporated into a meat food product. In certain embodiments, meat food products include food product made by processing the edible remains of any dead animal, including birds, fish, crustaceans, shellfish and mammals. Meat food products include, without limitation, for example, prepared beef, lamb, pork, poultry or seafood products. Examples of such meat food products include, for example, bologna, frankfurters, sausage, luncheon, deli slices, loaves, bacon, meatballs, fish sticks, chicken fingers, and ground meats, e.g., meatloaf, meatballs and hamburgers. A meat food product may be combined with a simulated meat food product. Simulated meat food products include, without limitation, for example, a meat alternative, meat analog, soy burger, soy bologna, soy frankfurter, soy sausage, soy luncheon loaves, soy bacon and soy meatball. A simulated meat food product may be combined with a meat food product. In certain embodiments, the flavor compositions of the present application are incorporated into a snack food category food product. In certain embodiments, snack food products include any food that can be a light informal meal including, but not limited to sweet and savory snacks and snack bars. Examples of snack food include, but are not limited to fruit snacks, chips/crisps, extruded snacks, tortilla/corn chips, popcorn, pretzels, nuts and other sweet and savory snacks. Examples of snack bars include, but are not limited to granola/muesli bars, breakfast bars, energy bars, fruit bars and other snack bars In certain embodiments, the flavor compositions of the present application are incorporated into frozen of food products, which comprises chilled or frozen food products, for example, but not limited to, ice cream, impulse ice cream, single portion dairy ice cream, single portion water ice cream, multi-pack dairy ice cream, multi-pack water ice cream, take-home ice cream, take-home dairy ice cream, ice cream desserts, bulk ice cream, take-home water ice cream, frozen yoghurt, artisanal ice cream, frozen ready meals, frozen pizza, chilled pizza, frozen soup, frozen pasta, frozen processed red meat, frozen processed poultry, frozen processed fish/seafood, frozen processed vegetables, frozen meat substitutes, frozen potatoes, frozen bakery products and frozen desserts. In certain embodiments, the flavor compositions of the present application are incorporated into food products for animal consumption. This includes food or drinks (liquids) for consumption by agricultural animals, pets and zoo animals. The presently disclosed subject matter can be used in a variety of food products. The term “food product” includes any food product, for example, those set forth in 21 CFR 101.12. Nonlimiting examples of such food products include frozen desserts, baked goods, fillings, nutritional drinks, beverages, salad dressing or similar dressing, sauces, icings, puddings and custards, batters, and the like. Various baked goods are disclosed in U.S. Pat. No. 6,536,599, the disclosure of which is herein incorporated by reference in its entirety. Non-limiting examples of bakery goods includes cookies, cakes, rolls, pastries, pie dough, brownies, breads, bagels and the like. The flavor compositions are also suitable as a component in frozen foods. 5.3 Pharmaceuticals The flavoring compositions can also be in the form of a pharmaceutical. One nonlimiting example of a pharmaceutical form is a suspension. Pharmaceutical suspensions can be prepared by conventional compounding methods. Suspensions can contain adjunct materials employed in formulating the suspensions of the art. The suspensions of the presently disclosed subject matter can comprise preservatives, buffers, suspending agents, antifoaming agents, sweetening agents, flavoring agents, coloring or decoloring agents, solubilizers, and combinations thereof. Flavoring agents such as those flavors well known to the skilled artisan, such as natural and artificial flavors and mints, such as peppermint, menthol, citrus flavors such as orange and lemon, artificial vanilla, cinnamon, various fruit flavors, both individual and mixed and the like can be utilized in amounts from about 0.01% to about 5%, and more preferably 0.01% to about 0.5% by weight of the suspension. The pharmaceutical suspensions of the presently disclosed subject matter can be prepared as follows.(A) admix the thickener with water heated from about 40° C. to about 95° C., preferably from about 40° C. to about 70° C., to form a dispersion if the thickener is not water soluble or a solution if the thickener is water soluble;(B) admix the sweetening agent with water to form a solution;(C) admix the flavoring agent with the thickener-water admixture to form a uniform thickener-flavoring agent;(D) combine the sweetener solution with the thickener-flavoring agent and mix until uniform; and(E) admix the optional adjunct materials such as coloring agents, flavoring agents, decolorants, solubilizers, antifoaming agents, buffers and additional water with the mixture of step (D) to form the suspension. The flavoring compositions can also be in chewable form. To achieve acceptable stability and quality as well as good taste and mouth feel in a chewable formulation several considerations are important. These considerations include the amount of active substance per tablet, the flavoring agent employed, the degree of compressibility of the tablet and additional properties of the composition. Chewable pharmaceutical candy is prepared by procedures similar to those used to make soft confectionery. A general discussion of the lozenge and chewable tablet forms of confectionery can be found in H. A. Lieberman and L. Lachman, Pharmaceutical Dosage Forms: Tablets Volume 1, Marcel Dekker, InC, New York, N.Y. (1989) at pages 367 to 418, which disclosure is incorporated herein by reference. In a typical procedure, a boiled sugar-corn syrup blend is formed to which is added a frappe mixture. The boiled sugar-corn syrup blend can be prepared from sugar and corn syrup blended in parts by weight ratio of about 90:10 to about 10:90. The sugar-corn syrup blend is heated to temperatures above about 120° C. to remove water and to form a molten mass. The frappe is generally prepared from gelatin, egg albumin, milk proteins such as casein, and vegetable proteins such as soy protein, and the like, which is added to a gelatin solution and rapidly mixed at ambient temperature to form an aerated sponge like mass. The frappe is then added to the molten candy mass and mixed until homogeneous at temperatures between about 65° C. and about 120° C. The flavor composition can then be added to the homogeneous mixture as the temperature is lowered to about 65° C.-95° C. whereupon additional ingredients can then be added such as flavoring agents and coloring agents. The formulation is further cooled and formed into pieces of desired dimensions. In other pharmaceutical embodiments, the flavoring agent is incorporated into an ingestible topical vehicle which can be in the form of a mouthwash, rinse, ingestible spray, suspension, dental gel, and the like. Typical non-toxic ingestible vehicles known in the pharmaceutical arts can be used in the presently disclosed subject matter. The preferred ingestible vehicles are water, ethanol, and water-ethanol mixtures. The water-ethanol mixtures are generally employed in a weight ratio from about 1:1 to about 20:1, preferably from about 3:1 to about 20:1, and most preferably from about 3:1 to about 10:1, respectively. The pH value of the ingestible vehicle is generally from about 4 to about 7, and preferably from about 5 to about 6.5. An ingestible topical vehicle having a pH value below about 4 is generally irritating to the ingestible cavity and an ingestible vehicle having a pH value greater than about 7 generally results in an unpleasant mouth feel. The ingestible topical flavoring agents can also contain conventional additives normally employed in those products. Conventional additives include a fluorine providing compound, a sweetening agent, a flavoring agent, a coloring agent, a humectant, a buffer, and an emulsifier, providing the additives do not interfere with the flavoring properties of the composition. The coloring agents and humectants, and the amounts of these additives to be employed, set out above, can be used in the ingestible topical composition. The flavoring agents (flavors, flavorants) which can be used include those flavors known to the skilled artisan, such as natural and artificial flavors. Suitable flavoring agents include mints, such as peppermint, citrus flavors such as orange and lemon, artificial vanilla, cinnamon, various fruit flavors, both individual and mixed, and the like. The amount of flavoring agent employed in the ingestible topical composition is normally a matter of preference subject to such factors as the type of final ingestible composition, the individual flavor employed, and the strength of flavor desired. Thus, the amount of flavoring can be varied in order to obtain the result desired in the final product and such variations are within the capabilities of those skilled in the art without the need for undue experimentation. The flavoring agents, when used, are generally utilized in amounts that can, for example, range in amounts from about 0.05% to about 6%, by weight of the ingestible topical composition. In accordance with the presently disclosed subject matter, effective amounts of the flavoring agents of the presently disclosed subject matter can be admixed with an ingestible topical vehicle to form a topical composition. These amounts are readily determined by those skilled in the art without the need for undue experimentation. In a preferred embodiment, the ingestible topical flavoring agents will comprise the flavoring agent in an amount from about 0.025% to about 2% and an ingestible topical vehicle in a quantity sufficient to bring the total amount of composition to 100%, by weight of the ingestible topical composition. In a more preferred embodiment, the ingestible topical flavoring agents will comprise the flavoring agent in an amount from about 0.05% to about 1% and an ingestible topical vehicle in a quantity sufficient to bring the total amount of composition to 100%, by weight of the ingestible topical composition. The presently disclosed subject matter extends to methods for preparing the ingestible topical flavoring agents. In such a method, the ingestible topical composition is prepared by admixing an effective amount of the flavoring agent of the presently disclosed subject matter and an ingestible topical vehicle. The final compositions are readily prepared using standard methods and apparatus generally known by those skilled in the pharmaceutical arts. The apparatus useful in accordance with the presently disclosed subject matter comprises mixing apparatus well known in the pharmaceutical arts, and therefore the selection of the specific apparatus will be apparent to the artisan. 6. Methods of Measuring Taste and Texture Attributes In certain embodiments of the present application, the taste and texture attributes of a food product can be modified by admixing a flavor composition with the food product as described herein. In certain embodiments, the attribute(s) can be enhanced or reduced by increasing or decreasing the concentration of the flavor composition admixed with the food product. In certain embodiments, the taste or texture attributes of the modified food product can be evaluated as described herein, and the concentration of flavor composition admixed with the food product can be increased or decreased based on the results of the evaluation. Taste and texture attributes can be reliably and reproducibly measured using sensory analysis methods known as descriptive analysis techniques. The Spectrum™ method of descriptive analysis is described in MORTEN MEILGAARD, D. Sc. ET AL., SENSORY EVALUATION TECHNIQUES (3d ed. 1999). The Spectrum™ method is a custom design approach meaning that the highly trained panelists who generate the data also develop the terminology to measure the attributes of interest. Further, the method uses intensity scales created to capture the intensity differences being investigated. These intensity scales are anchored to a set of well-chosen references. Using these references helps make the data universally understandable and usable over time. This ability to reproduce the results at another time and with another panel makes the data potentially more valuable than analytical techniques which offer similar reproducibility but lack the ability to fully capture the integrated sensory experiences as perceived by humans. When conducting quantitative descriptive analysis for compounds that modify other compounds, the testing methodology can be adapted to capture the change in character and intensity of the modified compound. For example, when testing for compounds that modify the saltiness of other compounds, the panelists may first taste a salt reference of agreed upon saltiness in order to establish a reference point for comparison. After tasting the reference, panelists may taste and score the test sample for saltiness as well as any other basic taste, chemical feeling factor, or aromatic notes. To quantify any increase in salt perception, the panelists may then taste re-taste the reference and again assign scores for saltiness as well as any other basic taste, chemical feeling factor, or aromatic notes. To quantify any lingering aftertaste, panelists may re-taste the salt reference at 1 minute intervals until their saltiness perception returns to the level of the reference. During the aftertaste evaluations, the panelists also note and score any other basic taste, chemical feeling factor, or aromatic notes. 7. Methods of Synthesis In certain embodiments, the HMG glucosides of the present application can be synthesized using standard chemosynthesis processes. In certain embodiments, the chemosynthesis process provides an HMG glucoside having a purity of at least 99.999%, or at least 99%, or at least 95%, or at least 90%, or at least 85%, or at least 80%. In certain embodiments, the HMG glucosides can be prepared using standard hydrolysis processes such as those employing acids, enzymes, or a combination of acids and enzymes. In certain embodiments, the HMG glucoside compositions of the present application comprise one or more compounds of Formula I. Such compounds may, without limitation, be synthesized by any means known in the art. In certain non-limiting embodiments, compounds of Formula I can be synthesized according to the following synthesis scheme: In certain non-limiting embodiments, compounds of Formula I can be synthesized according to the following synthesis scheme: In certain embodiments the HMG glucosides of the present application are prepared from a food product source that is fractionated and/or extracted to form an enriched HMG glucoside composition comprising the HMG glucosides. In certain embodiments, the enriched HMG glucoside composition comprises the flavor composition of the present application and is admixed with a food product according to the methods of the present application. In other embodiments, the enriched HMG glucoside composition is combined with other compositions to form the flavor composition of the present application, which is then admixed with the food product according to the methods of the present application. In certain embodiments the HMG glucosides of the present application are prepared from a food product source that is hydrolyzed to form a hydrolysate comprising the HMG glucosides. In certain embodiments, the hydrolysate comprises the flavor composition of the present application and is admixed with a food product according to the methods of the present application. In other embodiments, the hydrolysate is combined with other compositions to form the flavor composition of the present application, which is then admixed with the food product according to the methods of the present application. In certain embodiments the HMG glucosides of the present application are prepared from a food product source that is hydrolyzed and fractionated and/or extracted to form an enriched HMG glucoside hydrolysate composition comprising the HMG glucosides. In certain embodiments, the enriched HMG glucoside hydrolysate composition comprises the flavor composition of the present application and is admixed with a food product according to the methods of the present application. In other embodiments, the enriched HMG glucoside hydrolysate composition is combined with other compositions to form the flavor composition of the present application, which is then admixed with the food product according to the methods of the present application. EXAMPLES The presently disclosed subject matter will be better understood by reference to the following Examples, which are provided as exemplary of the invention, and not by way of limitation. Example 1—Preparation of an HMG Glucoside Composition by Hydrolysis The present example described the preparation of an HMG glucoside for use in a flavor composition through the hydrolysis of cocoa bean liquor made from West African cocoa beans. Reagents: A solution of 4N HCl was prepared by adding 100 mL 34-37% HCl in a 250 mL volumetric flask and filling it with water. A solution of 4N NaOH was prepared by dissolving 80 g NaOH pellets in 500 mL of water in a volumetric flask. Method: Cocoa liquor was run through a sieve and 30.09 g of fine powder was weighed into a 500 mL 3-neck round-bottom flask. The liquor was dissolved in 4N HCl (200 mL) and a stir bar was added to the flask. The sample was stirred at room temperature until the liquor was fully dispersed and flowed freely. A condenser was affixed to the flask and held at 8° C. A digital thermometer was pierced through a rubber stopper to measure the temperature of the solution. The third neck was plugged with a rubber stopper. The flask was wrapped in aluminum foil and heated to approximately 106° C. using a heating mantle. The sample was refluxed for 4.5 hours and left to cool to room temperature. The sample was transferred to a 1 L beaker and neutralized to pH 7 with 4N NaOH using a digital pH meter (pH 6.98 @29° C.). The sample was divided equally into 4 250 mL centrifuge tubes and centrifuged for 10 minutes @ 4500 rpm. The supernatant was filtered under vacuum through a Buchner funnel. The filtrate was then transferred to 2 32 oz plastic containers and lyophilized (yield 52.50 g). Example 1a—Preparation of an HMG Glucoside Composition by Extraction and Fractionation of a Cocoa Liquor Hydrolysate 1. Hydrolysis of Cocoa PowderPreparation: A solution of 4N HCl was prepared by adding 100 mL 34-37% HCl in a 250 mL volumetric flask and filling it to the line with water. A solution of 4N NaOH was prepared by dissolving 80 g NaOH pellets in 500 mL of water in a volumetric flask.Procedure: Cocoa liquor made fromTheobroma cacaococoa beans was run through a sieve and 30.09 g of fine powder was weighed into a 500 mL 3-neck round-bottom flask. The liquor was dissolved in 4N HCl (200 mL) and a stir bar was added to the flask. The sample was stirred at room temperature until the liquor was fully dispersed and flowed freely. A condenser was affixed to the flask and held at 8° C. A digital thermometer was pierced through a rubber stopper to measure the temperature of the solution. The third neck was plugged with a rubber stopper. The flask was wrapped in aluminum foil and heated to approximately 106° C. using a heating mantle. The sample was refluxed for 4.5 hours and left to cool to room temperature. The sample was transferred to a 1 L beaker and neutralized to pH 7 with 4N NaOH using a digital pH meter (pH 6.98 @ 29° C.). The sample was divided equally into 4 250 mL centrifuge tubes and centrifuged for 10 minutes @ 4500 rpm. The supernatant was filtered under vacuum through a Buchner funnel. The filtrate was then transferred to 2 32 oz plastic containers and lyophilized. 2. Ethanol Extraction of Hydrolyzed Cocoa PowderThe hydrolyzed cocoa powder was extracted with ethanol to remove a bulk of the salts generated during neutralization. Hydrolyzed cocoa powder (50.36 g) was divided equally into 2 500 mL centrifuge tubes. Ethanol (200 mL) was added slowly to each tube as to not disturb the sample. The samples were shaken for 15 minutes on an autoshaker and then centrifuged for 10 minutes @4500 rpm. The supernatant was decanted into a 1000 mL round-bottom flask. The residue was scraped off the bottom of the tubes and redissolved in ethanol (200 mL each). The samples were shaken for 15 minutes on an autoshaker and then centrifuged for 10 minutes @ 4500 rpm. The supernatant was combined with the previous supernatant and evaporated under reduced pressure to remove all organic solvent. The remaining solids were redissolved in approximately 100 mL deionized water and lyophilized. 3. SPE (Solid Phase Extraction) Fractionation of HCP (Hydrolysed Cocoa Powder) Ethanol ExtractThe extract previously obtained was further fractionated to exhaustively remove the salts and hydrophilic molecules. HCP ethanol extract was transferred to 14 glass vials (approximately 0.5 g each, 20 mL volume) and dissolved in DI water (10 mL). The samples were shaken until dissolved (approximately 1 minute). The samples were filtered through a syringe and PTFE filter to remove particulates as necessary. A solid phase extraction (SPE) cartridge (20 g/60 mL, C18 stationary phase) was conditioned sequentially with DI water (100 mL), methanol (100 mL), and DI water (100 mL). The sample (10 mL) was then loaded onto cartridge and washed with DI water (100 mL) and extracted with methanol (100 mL). The cartridge was reconditioned and the remaining 13 samples were washed and extracted as previously described. The organic solutions were combined and rotary evaporated under reduced pressure. The residue was redissolved in DI water and lyophilized using a Labconco freeze dryer. The sample was separated by high-performance liquid chromatography (HPLC) to narrow down the taste-active molecules of interest. Example 1b—Preparation of an HMG Glucoside Composition by Extraction and Fractionation of Cocoa Liquor 1. Liquid/Solid Extraction of LiquorCocoa Liquor made from cocoa beans sourced from Papua New Guinea (PNG liquor) (600 g) was frozen in liquid nitrogen and ground into a fine powder with a laboratory mill. The powder was divided equally into six plastic centrifuge tubes (500 mL volume). Each sample (100 g PNG liquor) was extracted with diethyl ether (200 mL) for 15 minutes using an autoshaker to remove the fat. After centrifugation (10 min, 4500 rpm), the supernatant was discarded. The extraction process was repeated three more times for a total of four times. The remaining defatted liquor was left to air dry in a fume hood overnight. Defatted liquor (200 g) was divided equally between four plastic centrifuge bottles (250 mL volume). To each sample (50 g defatted PNG liquor), 150 mL 70:30 acetone:water was added. The bottles were placed on an autoshaker for 15 minutes. Each sample was centrifuged (5 min, 3500 rpm) and then the supernatant was vacuum filtered using Whatman 540 filter paper and a Buchner funnel. The residue was freed from the bottom of the bottles by hand and additional 70:30 acetone:water (100 mL) was added to each sample. The samples were shaken for 15 minutes using an auto-shaker. After centrifugation (10 min, 4500 rpm), the supernatant was vacuum filtered again using the same procedure described above. The supernatants from each extraction were combined (˜800 mL) and the residue was discarded. The supernatant was rotary evaporated under reduced pressure and the remaining aqueous solution (˜250 mL) was transferred into a separatory funnel (1000 mL volume). The aqueous solution was washed with Dichloromethane (3×300 mL) to remove any xanthines. The dichloromethane layer was discarded, then the aqueous solution was washed sequentially with n-butyl acetate (3×300 mL), ethyl acetate (3×300 mL), and methyl acetate (3×300 mL) to remove procyanidins. The organic layers were discarded and the aqueous solution (F7) was rotary evaporated under reduced pressure to remove any remaining solvent. The remaining water solution was lyophilized using a Labconco freeze dryer (100×103mbar, −40° C.). Sensory analysis was performed and the savory attribute was found to be in F7. 2. Solid Phase Extraction (SPE)For removal of any residual salts, treated PNG liquor powder (F7) was transferred to 14 glass vials (20 mL volume, approximately 0.5 g sample in each vial) and dissolved in DI water (10 mL). The samples were shaken until dissolved (approximately 1 minute). A solid phase extraction (SPE) cartridge (20 g/60 mL, C18 stationary phase) was conditioned sequentially with DI water (100 mL), methanol (100 mL), and DI water (100 mL). The vacuum was broken and the sample (10 mL) was then loaded onto cartridge. The vacuum was resumed and the sample was washed with DI water (100 mL). The receptacle flask was changed and the sample was extracted with methanol (100 mL). The cartridge was reconditioned and the remaining 13 samples were washed and extracted as previously described. The organic solutions were combined and rotary evaporated under reduced pressure. The residue was redissolved in DI water and lyophilized using a Labconco freeze dryer (100×103mbar, −40° C.). Sensory analysis confirmed the presence of the savory attribute in the organic fraction. Example 2—Preparation of an HMG Glucoside Composition by Synthetic Chemosynthesis HMG glucosides described by the present application were prepared through synthetic chemosynthesis methods described in synthesis schemes 1 and 2 below. Although the presently disclosed subject matter and its advantages have been described in detail, it should be understood that various changes, substitutions and alterations can be made herein without departing from the spirit and scope of the invention as defined by the appended claims. Moreover, the scope of the present application is not intended to be limited to the particular embodiments of the process, machine, manufacture, composition of matter, means, methods and steps described in the specification. As one of ordinary skill in the art will readily appreciate from the disclosure of the presently disclosed subject matter, processes, machines, manufacture, compositions of matter, means, methods, or steps, presently existing or later to be developed that perform substantially the same function or achieve substantially the same result as the corresponding embodiments described herein may be utilized according to the presently disclosed subject matter. Accordingly, the appended claims are intended to include within their scope such processes, machines, manufacture, compositions of matter, means, methods, or steps. Patents, patent applications publications product descriptions, and protocols are cited throughout this application the disclosures of which are incorporated herein by reference in their entireties for all purposes.
110,228
11858956
DETAILED DESCRIPTION The present invention can be understood more readily by reference to the following detailed description of the invention and the Examples included therein. Before the present compounds, compositions, articles, systems, devices, and/or methods are disclosed and described, it is to be understood that they are not limited to specific synthetic methods unless otherwise specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and is not intended to be limiting. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, example methods and materials are now described. While aspects of the present invention can be described and claimed in a particular statutory class, such as the system statutory class, this is for convenience only and one of skill in the art will understand that each aspect of the present invention can be described and claimed in any statutory class. Unless otherwise expressly stated, it is in no way intended that any method or aspect set forth herein be construed as requiring that its steps be performed in a specific order. Accordingly, where a method claim does not specifically state in the claims or descriptions that the steps are to be limited to a specific order, it is no way intended that an order be inferred, in any respect. This holds for any possible non-express basis for interpretation, including matters of logic with respect to arrangement of steps or operational flow, plain meaning derived from grammatical organization or punctuation, or the number or type of aspects described in the specification. Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided herein may be different from the actual publication dates, which can require independent confirmation. A. DEFINITIONS As used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a functional group,” “an alkyl,” or “a residue” includes mixtures of two or more such functional groups, alkyls, or residues, and the like. As used in the specification and in the claims, the term “comprising” can include the aspects “consisting of” and “consisting essentially of.” Ranges can be expressed herein as from “about” one particular value, and/or to “about” another particular value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10” is also disclosed. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed. As used herein, the terms “about” and “at or about” mean that the amount or value in question can be the value designated some other value approximately or about the same. It is generally understood, as used herein, that it is the nominal value indicated ±10% variation unless otherwise indicated or inferred. The term is intended to convey that similar values promote equivalent results or effects recited in the claims. That is, it is understood that amounts, sizes, formulations, parameters, and other quantities and characteristics are not and need not be exact, but can be approximate and/or larger or smaller, as desired, reflecting tolerances, conversion factors, rounding off, measurement error and the like, and other factors known to those of skill in the art. In general, an amount, size, formulation, parameter or other quantity or characteristic is “about” or “approximate” whether or not expressly stated to be such. It is understood that where “about” is used before a quantitative value, the parameter also includes the specific quantitative value itself, unless specifically stated otherwise. References in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed. Thus, in a compound containing 2 parts by weight of component X and 5 parts by weight component Y, X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound. A weight percent (wt. %) of a component, unless specifically stated to the contrary, is based on the total weight of the formulation or composition in which the component is included. As used herein, “IC50” is intended to refer to the concentration of a substance (e.g., a compound or a drug) that is required for 50% inhibition of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc. In one aspect, an IC50can refer to the concentration of a substance that is required for 50% inhibition in vivo, as further defined elsewhere herein. In a further aspect, IC50refers to the half maximal (50%) inhibitory concentration (IC) of a substance. As used herein, “EC50” is intended to refer to the concentration of a substance (e.g., a compound or a drug) that is required for 50% agonism of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc. In one aspect, an EC50can refer to the concentration of a substance that is required for 50% agonism in vivo, as further defined elsewhere herein. In a further aspect, EC50refers to the concentration of agonist that provokes a response halfway between the baseline and maximum response. As used herein, “EC90” is intended to refer to the concentration of a substance (e.g., a compound or a drug) that is required for 90% agonism of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc. In one aspect, an EC90can refer to the concentration of a substance that is required for 90% agonism in vivo, as further defined elsewhere herein. As used herein, “CC50” is intended to refer to the concentration of a substance (e.g., a compound or a drug) that is required for 50% reduction of cell viability. In one aspect, an CC50can refer to the concentration of a substance that is required for 50% reduction (i.e., cytotoxicity) in vivo, as further defined elsewhere herein. As used herein, the terms “optional” or “optionally” means that the subsequently described event or circumstance can or cannot occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. As used herein, the term “subject” can be a vertebrate, such as a mammal, a fish, a bird, a reptile, or an amphibian. Thus, the subject of the herein disclosed methods can be a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, cat, guinea pig or rodent. The term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered. In one aspect, the subject is a mammal. A patient refers to a subject afflicted with a disease or disorder. The term “patient” includes human and veterinary subjects. As used herein, the term “treatment” refers to the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder. This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder. In various aspects, the term covers any treatment of a subject, including a mammal (e.g., a human), and includes: (i) preventing the disease from occurring in a subject that can be predisposed to the disease but has not yet been diagnosed as having it; (ii) inhibiting the disease, i.e., arresting its development; or (iii) relieving the disease, i.e., causing regression of the disease. In one aspect, the subject is a mammal such as a primate, and, in a further aspect, the subject is a human. The term “subject” also includes domesticated animals (e.g., cats, dogs, etc.), livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), and laboratory animals (e.g., mouse, rabbit, rat, guinea pig, fruit fly, etc.). As used herein, the term “prevent” or “preventing” refers to precluding, averting, obviating, forestalling, stopping, or hindering something from happening, especially by advance action. It is understood that where reduce, inhibit or prevent are used herein, unless specifically indicated otherwise, the use of the other two words is also expressly disclosed. As used herein, the term “diagnosed” means having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition that can be diagnosed or treated by the compounds, compositions, or methods disclosed herein. As used herein, the terms “administering” and “administration” refer to any method of providing a pharmaceutical preparation to a subject. Such methods are well known to those skilled in the art and include, but are not limited to, oral administration, transdermal administration, administration by inhalation, nasal administration, topical administration, intravaginal administration, ophthalmic administration, intraaural administration, intracerebral administration, rectal administration, sublingual administration, buccal administration, and parenteral administration, including injectable such as intravenous administration, intra-arterial administration, intramuscular administration, and subcutaneous administration. Administration can be continuous or intermittent. In various aspects, a preparation can be administered therapeutically; that is, administered to treat an existing disease or condition. In further various aspects, a preparation can be administered prophylactically; that is, administered for prevention of a disease or condition. As used herein, the terms “effective amount” and “amount effective” refer to an amount that is sufficient to achieve the desired result or to have an effect on an undesired condition. For example, a “therapeutically effective amount” refers to an amount that is sufficient to achieve the desired therapeutic result or to have an effect on undesired symptoms, but is generally insufficient to cause adverse side effects. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration; the route of administration; the rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of a compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. If desired, the effective daily dose can be divided into multiple doses for purposes of administration. Consequently, single dose compositions can contain such amounts or submultiples thereof to make up the daily dose. The dosage can be adjusted by the individual physician in the event of any contraindications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days. Guidance can be found in the literature for appropriate dosages for given classes of pharmaceutical products. In further various aspects, a preparation can be administered in a “prophylactically effective amount”; that is, an amount effective for prevention of a disease or condition. As used herein, “dosage form” means a pharmacologically active material in a medium, carrier, vehicle, or device suitable for administration to a subject. A dosage forms can comprise inventive a disclosed compound, a product of a disclosed method of making, or a salt, solvate, or polymorph thereof, in combination with a pharmaceutically acceptable excipient, such as a preservative, buffer, saline, or phosphate buffered saline. Dosage forms can be made using conventional pharmaceutical manufacturing and compounding techniques. Dosage forms can comprise inorganic or organic buffers (e.g., sodium or potassium salts of phosphate, carbonate, acetate, or citrate) and pH adjustment agents (e.g., hydrochloric acid, sodium or potassium hydroxide, salts of citrate or acetate, amino acids and their salts) antioxidants (e.g., ascorbic acid, alpha-tocopherol), surfactants (e.g., polysorbate 20, polysorbate 80, polyoxyethylene9-10 nonyl phenol, sodium desoxycholate), solution and/or cryo/lyo stabilizers (e.g., sucrose, lactose, mannitol, trehalose), osmotic adjustment agents (e.g., salts or sugars), antibacterial agents (e.g., benzoic acid, phenol, gentamicin), antifoaming agents (e.g., polydimethylsilozone), preservatives (e.g., thimerosal, 2-phenoxyethanol, EDTA), polymeric stabilizers and viscosity-adjustment agents (e.g., polyvinylpyrrolidone, poloxamer 488, carboxymethylcellulose) and co-solvents (e.g., glycerol, polyethylene glycol, ethanol). A dosage form formulated for injectable use can have a disclosed compound, a product of a disclosed method of making, or a salt, solvate, or polymorph thereof, suspended in sterile saline solution for injection together with a preservative. As used herein, “kit” means a collection of at least two components constituting the kit. Together, the components constitute a functional unit for a given purpose. Individual member components may be physically packaged together or separately. For example, a kit comprising an instruction for using the kit may or may not physically include the instruction with other individual member components. Instead, the instruction can be supplied as a separate member component, either in a paper form or an electronic form which may be supplied on computer readable memory device or downloaded from an internet website, or as recorded presentation. As used herein, “instruction(s)” means documents describing relevant materials or methodologies pertaining to a kit. These materials may include any combination of the following: background information, list of components and their availability information (purchase information, etc.), brief or detailed protocols for using the kit, trouble-shooting, references, technical support, and any other related documents. Instructions can be supplied with the kit or as a separate member component, either as a paper form or an electronic form which may be supplied on computer readable memory device or downloaded from an internet website, or as recorded presentation. Instructions can comprise one or multiple documents, and are meant to include future updates. As used herein, the terms “therapeutic agent” include any synthetic or naturally occurring biologically active compound or composition of matter which, when administered to an organism (human or nonhuman animal), induces a desired pharmacologic, immunogenic, and/or physiologic effect by local and/or systemic action. The term therefore encompasses those compounds or chemicals traditionally regarded as drugs, vaccines, and biopharmaceuticals including molecules such as proteins, peptides, hormones, nucleic acids, gene constructs and the like. Examples of therapeutic agents are described in well-known literature references such as the Merck Index (14thedition), the Physicians' Desk Reference (64thedition), and The Pharmacological Basis of Therapeutics (12thedition), and they include, without limitation, medicaments; vitamins; mineral supplements; substances used for the treatment, prevention, diagnosis, cure or mitigation of a disease or illness; substances that affect the structure or function of the body, or pro-drugs, which become biologically active or more active after they have been placed in a physiological environment. For example, the term “therapeutic agent” includes compounds or compositions for use in all of the major therapeutic areas including, but not limited to, adjuvants; anti-infectives such as antibiotics and antiviral agents; analgesics and analgesic combinations, anorexics, anti-inflammatory agents, anti-epileptics, local and general anesthetics, hypnotics, sedatives, antipsychotic agents, neuroleptic agents, antidepressants, anxiolytics, antagonists, neuron blocking agents, anticholinergic and cholinomimetic agents, antimuscarinic and muscarinic agents, antiadrenergics, antiarrhythmics, antihypertensive agents, hormones, and nutrients, antiarthritics, antiasthmatic agents, anticonvulsants, antihistamines, antinauseants, antineoplastics, antipruritics, antipyretics; antispasmodics, cardiovascular preparations (including calcium channel blockers, beta-blockers, beta-agonists and antiarrythmics), antihypertensives, diuretics, vasodilators; central nervous system stimulants; cough and cold preparations; decongestants; diagnostics; hormones; bone growth stimulants and bone resorption inhibitors; immunosuppressives; muscle relaxants; psychostimulants; sedatives; tranquilizers; proteins, peptides, and fragments thereof (whether naturally occurring, chemically synthesized or recombinantly produced); and nucleic acid molecules (polymeric forms of two or more nucleotides, either ribonucleotides (RNA) or deoxyribonucleotides (DNA) including both double- and single-stranded molecules, gene constructs, expression vectors, antisense molecules and the like), small molecules (e.g., doxorubicin) and other biologically active macromolecules such as, for example, proteins and enzymes. The agent may be a biologically active agent used in medical, including veterinary, applications and in agriculture, such as with plants, as well as other areas. The term “therapeutic agent” also includes without limitation, medicaments; vitamins; mineral supplements; substances used for the treatment, prevention, diagnosis, cure or mitigation of disease or illness; or substances which affect the structure or function of the body; or pro-drugs, which become biologically active or more active after they have been placed in a predetermined physiological environment. The term “pharmaceutically acceptable” describes a material that is not biologically or otherwise undesirable, i.e., without causing an unacceptable level of undesirable biological effects or interacting in a deleterious manner. As used herein, the term “derivative” refers to a compound having a structure derived from the structure of a parent compound (e.g., a compound disclosed herein) and whose structure is sufficiently similar to those disclosed herein and based upon that similarity, would be expected by one skilled in the art to exhibit the same or similar activities and utilities as the claimed compounds, or to induce, as a precursor, the same or similar activities and utilities as the claimed compounds. Exemplary derivatives include salts, esters, amides, salts of esters or amides, and N-oxides of a parent compound. As used herein, the term “pharmaceutically acceptable carrier” refers to sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), carboxymethylcellulose and suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. These compositions can also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms can be ensured by the inclusion of various antibacterial and antifungal agents such as paraben, chlorobutanol, phenol, sorbic acid and the like. It can also be desirable to include isotonic agents such as sugars, sodium chloride and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the inclusion of agents, such as aluminum monostearate and gelatin, which delay absorption. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide, poly(orthoesters) and poly(anhydrides). Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues. The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable media just prior to use. Suitable inert carriers can include sugars such as lactose. Desirably, at least 95% by weight of the particles of the active ingredient have an effective particle size in the range of 0.01 to 10 micrometers. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described below. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms, such as nitrogen, can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This disclosure is not intended to be limited in any manner by the permissible substituents of organic compounds. Also, the terms “substitution” or “substituted with” include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. It is also contemplated that, in certain aspects, unless expressly indicated to the contrary, individual substituents can be further optionally substituted (i.e., further substituted or unsubstituted). In defining various terms, “A1,” “A2,” “A3,” and “A4” are used herein as generic symbols to represent various specific substituents. These symbols can be any substituent, not limited to those disclosed herein, and when they are defined to be certain substituents in one instance, they can, in another instance, be defined as some other substituents. The term “aliphatic” or “aliphatic group,” as used herein, denotes a hydrocarbon moiety that may be straight-chain (i.e., unbranched), branched, or cyclic (including fused, bridging, and spirofused polycyclic) and may be completely saturated or may contain one or more units of unsaturation, but which is not aromatic. Unless otherwise specified, aliphatic groups contain 1-20 carbon atoms. Aliphatic groups include, but are not limited to, linear or branched, alkyl, alkenyl, and alkynyl groups, and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl. The term “alkyl” as used herein is a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, s-butyl, t-butyl, n-pentyl, isopentyl, s-pentyl, neopentyl, hexyl, heptyl, octyl, nonyl, decyl, dodecyl, tetradecyl, hexadecyl, eicosyl, tetracosyl, and the like. The alkyl group can be cyclic or acyclic. The alkyl group can be branched or unbranched. The alkyl group can also be substituted or unsubstituted. For example, the alkyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, amino, ether, halide, hydroxy, nitro, silyl, sulfo-oxo, or thiol, as described herein. A “lower alkyl” group is an alkyl group containing from one to six (e.g., from one to four) carbon atoms. The term alkyl group can also be a C1 alkyl, C1-C2 alkyl, C1-C3 alkyl, C1-C4 alkyl, C1-C5 alkyl, C1-C6 alkyl, C1-C7 alkyl, C1-C8 alkyl, C1-C9 alkyl, C1-C10 alkyl, and the like up to and including a C1-C24 alkyl. Throughout the specification “alkyl” is generally used to refer to both unsubstituted alkyl groups and substituted alkyl groups; however, substituted alkyl groups are also specifically referred to herein by identifying the specific substituent(s) on the alkyl group. For example, the term “halogenated alkyl” or “haloalkyl” specifically refers to an alkyl group that is substituted with one or more halide, e.g., fluorine, chlorine, bromine, or iodine. Alternatively, the term “monohaloalkyl” specifically refers to an alkyl group that is substituted with a single halide, e.g. fluorine, chlorine, bromine, or iodine. The term “polyhaloalkyl” specifically refers to an alkyl group that is independently substituted with two or more halides, i.e. each halide substituent need not be the same halide as another halide substituent, nor do the multiple instances of a halide substituent need to be on the same carbon. The term “alkoxyalkyl” specifically refers to an alkyl group that is substituted with one or more alkoxy groups, as described below. The term “aminoalkyl” specifically refers to an alkyl group that is substituted with one or more amino groups. The term “hydroxyalkyl” specifically refers to an alkyl group that is substituted with one or more hydroxy groups. When “alkyl” is used in one instance and a specific term such as “hydroxyalkyl” is used in another, it is not meant to imply that the term “alkyl” does not also refer to specific terms such as “hydroxyalkyl” and the like. This practice is also used for other groups described herein. That is, while a term such as “cycloalkyl” refers to both unsubstituted and substituted cycloalkyl moieties, the substituted moieties can, in addition, be specifically identified herein; for example, a particular substituted cycloalkyl can be referred to as, e.g., an “alkylcycloalkyl.” Similarly, a substituted alkoxy can be specifically referred to as, e.g., a “halogenated alkoxy,” a particular substituted alkenyl can be, e.g., an “alkenylalcohol,” and the like. Again, the practice of using a general term, such as “cycloalkyl,” and a specific term, such as “alkylcycloalkyl,” is not meant to imply that the general term does not also include the specific term. The term “cycloalkyl” as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl, and the like. The term “heterocycloalkyl” is a type of cycloalkyl group as defined above, and is included within the meaning of the term “cycloalkyl,” where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted. The cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, amino, ether, halide, hydroxy, nitro, silyl, sulfo-oxo, or thiol as described herein. The term “polyalkylene group” as used herein is a group having two or more CH2groups linked to one another. The polyalkylene group can be represented by the formula —(CH2)a—, where “a” is an integer of from 2 to 500. The terms “alkoxy” and “alkoxyl” as used herein to refer to an alkyl or cycloalkyl group bonded through an ether linkage; that is, an “alkoxy” group can be defined as —OA1where A1is alkyl or cycloalkyl as defined above. “Alkoxy” also includes polymers of alkoxy groups as just described; that is, an alkoxy can be a polyether such as —OA1-OA2or —OA1-(OA2)a-OA3, where “a” is an integer of from 1 to 200 and A1, A2, and A3are alkyl and/or cycloalkyl groups. The term “alkenyl” as used herein is a hydrocarbon group of from 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon double bond. Asymmetric structures such as (A1A2)C═C(A3A4) are intended to include both the E and Z isomers. This can be presumed in structural formulae herein wherein an asymmetric alkene is present, or it can be explicitly indicated by the bond symbol C═C. The alkenyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol, as described herein. The term “cycloalkenyl” as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms and containing at least one carbon-carbon double bound, i.e., C═C. Examples of cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, norbornenyl, and the like. The term “heterocycloalkenyl” is a type of cycloalkenyl group as defined above, and is included within the meaning of the term “cycloalkenyl,” where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkenyl group and heterocycloalkenyl group can be substituted or unsubstituted. The cycloalkenyl group and heterocycloalkenyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein. The term “alkynyl” as used herein is a hydrocarbon group of 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon triple bond. The alkynyl group can be unsubstituted or substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol, as described herein. The term “cycloalkynyl” as used herein is a non-aromatic carbon-based ring composed of at least seven carbon atoms and containing at least one carbon-carbon triple bound. Examples of cycloalkynyl groups include, but are not limited to, cycloheptynyl, cyclooctynyl, cyclononynyl, and the like. The term “heterocycloalkynyl” is a type of cycloalkenyl group as defined above, and is included within the meaning of the term “cycloalkynyl,” where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkynyl group and heterocycloalkynyl group can be substituted or unsubstituted. The cycloalkynyl group and heterocycloalkynyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein. The term “aromatic group” as used herein refers to a ring structure having cyclic clouds of delocalized n electrons above and below the plane of the molecule, where the a clouds contain (4n+2) π electrons. A further discussion of aromaticity is found in Morrison and Boyd, Organic Chemistry, (5th Ed., 1987), Chapter 13, entitled “Aromaticity,” pages 477-497, incorporated herein by reference. The term “aromatic group” is inclusive of both aryl and heteroaryl groups. The term “aryl” as used herein is a group that contains any carbon-based aromatic group including, but not limited to, benzene, naphthalene, phenyl, biphenyl, anthracene, and the like. The aryl group can be substituted or unsubstituted. The aryl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, —NH2, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein. The term “biaryl” is a specific type of aryl group and is included in the definition of “aryl.” In addition, the aryl group can be a single ring structure or comprise multiple ring structures that are either fused ring structures or attached via one or more bridging groups such as a carbon-carbon bond. For example, biaryl can be two aryl groups that are bound together via a fused ring structure, as in naphthalene, or are attached via one or more carbon-carbon bonds, as in biphenyl. The term “aldehyde” as used herein is represented by the formula —C(O)H. Throughout this specification “C(O)” is a short hand notation for a carbonyl group, i.e., C═O. The terms “amine” or “amino” as used herein are represented by the formula —NA1A2, where A1and A2can be, independently, hydrogen or alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. A specific example of amino is —NH2. The term “alkylamino” as used herein is represented by the formula —NH(-alkyl) where alkyl is a described herein. Representative examples include, but are not limited to, methylamino group, ethylamino group, propylamino group, isopropylamino group, butylamino group, isobutylamino group, (sec-butyl)amino group, (tert-butyl)amino group, pentylamino group, isopentylamino group, (tert-pentyl)amino group, hexylamino group, and the like. The term “dialkylamino” as used herein is represented by the formula —N(-alkyl)2where alkyl is a described herein. Representative examples include, but are not limited to, dimethylamino group, diethylamino group, dipropylamino group, diisopropylamino group, dibutylamino group, diisobutylamino group, di(sec-butyl)amino group, di(tert-butyl)amino group, dipentylamino group, diisopentylamino group, di(tert-pentyl)amino group, dihexylamino group, N-ethyl-N-methylamino group, N-methyl-N-propylamino group, N-ethyl-N-propylamino group and the like. The term “carboxylic acid” as used herein is represented by the formula —C(O)OH. The term “ester” as used herein is represented by the formula —OC(O)A1or —C(O)OA1, where A1can be alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “polyester” as used herein is represented by the formula -(A1O(O)C-A2-C(O)O)a— or -(A1O(O)C-A2-OC(O))a—, where A1and A2can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein and “a” is an integer from 1 to 500. “Polyester” is as the term used to describe a group that is produced by the reaction between a compound having at least two carboxylic acid groups with a compound having at least two hydroxyl groups. The term “ether” as used herein is represented by the formula A1OA2, where A1and A2can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein. The term “polyether” as used herein is represented by the formula -(A1O-A2O)a—, where A1and A2can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein and “a” is an integer of from 1 to 500. Examples of polyether groups include polyethylene oxide, polypropylene oxide, and polybutylene oxide. The terms “halo,” “halogen,” or “halide,” as used herein can be used interchangeably and refer to F, Cl, Br, or I. The terms “pseudohalide,” “pseudohalogen,” or “pseudohalo,” as used herein can be used interchangeably and refer to functional groups that behave substantially similar to halides. Such functional groups include, by way of example, cyano, thiocyanato, azido, trifluoromethyl, trifluoromethoxy, perfluoroalkyl, and perfluoroalkoxy groups. The term “heteroalkyl,” as used herein refers to an alkyl group containing at least one heteroatom. Suitable heteroatoms include, but are not limited to, O, N, Si, P and S, wherein the nitrogen, phosphorous and sulfur atoms are optionally oxidized, and the nitrogen heteroatom is optionally quaternized. Heteroalkyls can be substituted as defined above for alkyl groups. The term “heteroaryl,” as used herein refers to an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorus, where N-oxides, sulfur oxides, and dioxides are permissible heteroatom substitutions. The heteroaryl group can be substituted or unsubstituted. The heteroaryl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, amino, ether, halide, hydroxy, nitro, silyl, sulfo-oxo, or thiol as described herein. Heteroaryl groups can be monocyclic, or alternatively fused ring systems. Heteroaryl groups include, but are not limited to, furyl, imidazolyl, pyrimidinyl, tetrazolyl, thienyl, pyridinyl, pyrrolyl, N-methylpyrrolyl, quinolinyl, isoquinolinyl, pyrazolyl, triazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, isothiazolyl, pyridazinyl, pyrazinyl, benzofuranyl, benzodioxolyl, benzothiophenyl, indolyl, indazolyl, benzimidazolyl, imidazopyridinyl, pyrazolopyridinyl, and pyrazolopyrimidinyl. Further not limiting examples of heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiophenyl, pyrazolyl, imidazolyl, benzo[d]oxazolyl, benzo[d]thiazolyl, quinolinyl, quinazolinyl, indazolyl, imidazo[1,2-b]pyridazinyl, imidazo[1,2-a]pyrazinyl, benzo[c][1,2,5]thiadiazolyl, benzo[c][1,2,5]oxadiazolyl, and pyrido[2,3-b]pyrazinyl. The terms “heterocycle” and “heterocyclyl” as used herein can be used interchangeably and refer to single and multi-cyclic aromatic or non-aromatic ring systems in which at least one of the ring members is other than carbon. Thus, the term is inclusive of, but not limited to, “heterocycloalkyl,” “heteroaryl,” “bicyclic heterocycle,” and “polycyclic heterocycle.” Heterocycle includes pyridine, pyrimidine, furan, thiophene, pyrrole, isoxazole, isothiazole, pyrazole, oxazole, thiazole, imidazole, oxazole, including, 1,2,3-oxadiazole, 1,2,5-oxadiazole and 1,3,4-oxadiazole, thiadiazole, including, 1,2,3-thiadiazole, 1,2,5-thiadiazole, and 1,3,4-thiadiazole, triazole, including, 1,2,3-triazole, 1,3,4-triazole, tetrazole, including 1,2,3,4-tetrazole and 1,2,4,5-tetrazole, pyridazine, pyrazine, triazine, including 1,2,4-triazine and 1,3,5-triazine, tetrazine, including 1,2,4,5-tetrazine, pyrrolidine, piperidine, piperazine, morpholine, azetidine, tetrahydropyran, tetrahydrofuran, dioxane, and the like. The term heterocyclyl group can also be a C2 heterocyclyl, C2-C3 heterocyclyl, C2-C4 heterocyclyl, C2-C5 heterocyclyl, C2-C6 heterocyclyl, C2-C7 heterocyclyl, C2-C8 heterocyclyl, C2-C9 heterocyclyl, C2-C10 heterocyclyl, C2-C11 heterocyclyl, and the like up to and including a C2-C18 heterocyclyl. For example, a C2 heterocyclyl comprises a group which has two carbon atoms and at least one heteroatom, including, but not limited to, aziridinyl, diazetidinyl, dihydrodiazetyl, oxiranyl, thiiranyl, and the like. Alternatively, for example, a C5 heterocyclyl comprises a group which has five carbon atoms and at least one heteroatom, including, but not limited to, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, diazepanyl, pyridinyl, and the like. It is understood that a heterocyclyl group may be bound either through a heteroatom in the ring, where chemically possible, or one of carbons comprising the heterocyclyl ring. The term “bicyclic heterocycle” and “bicyclic heterocyclyl” as used herein refers to a ring system in which at least one of the ring members is other than carbon. Bicyclic heterocyclyl encompasses ring systems wherein an aromatic ring is fused with another aromatic ring, or wherein an aromatic ring is fused with a non-aromatic ring. Bicyclic heterocyclyl encompasses ring systems wherein a benzene ring is fused to a 5- or a 6-membered ring containing 1, 2 or 3 ring heteroatoms or wherein a pyridine ring is fused to a 5- or a 6-membered ring containing 1, 2 or 3 ring heteroatoms. Bicyclic heterocyclic groups include, but are not limited to, indolyl, indazolyl, pyrazolo[1,5-a]pyridinyl, benzofuranyl, quinolinyl, quinoxalinyl, 1,3-benzodioxolyl, 2,3-dihydro-1,4-benzodioxinyl, 3,4-dihydro-2H-chromenyl, 1H-pyrazolo[4,3-c]pyridin-3-yl; 1H-pyrrolo[3,2-b]pyridin-3-yl; and 1H-pyrazolo[3,2-b]pyridin-3-yl. The term “heterocycloalkyl” as used herein refers to an aliphatic, partially unsaturated or fully saturated, 3- to 14-membered ring system, including single rings of 3 to 8 atoms and bi- and tricyclic ring systems. The heterocycloalkyl ring-systems include one to four heteroatoms independently selected from oxygen, nitrogen, and sulfur, wherein a nitrogen and sulfur heteroatom optionally can be oxidized and a nitrogen heteroatom optionally can be substituted. Representative heterocycloalkyl groups include, but are not limited to, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, and tetrahydrofuryl. The term “hydroxyl” or “hydroxyl” as used herein is represented by the formula —OH. The term “ketone” as used herein is represented by the formula A1C(O)A2, where A1and A2can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “azide” or “azido” as used herein is represented by the formula —N3. The term “nitro” as used herein is represented by the formula —NO2. The term “nitrile” or “cyano” as used herein is represented by the formula —CN. The term “silyl” as used herein is represented by the formula —SiA1A2A3, where A1, A2, and A3can be, independently, hydrogen or an alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “sulfo-oxo” as used herein is represented by the formulas —S(O)A1, —S(O)2A1, —OS(O)2A1, or —OS(O)2OA1, where A1can be hydrogen or an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. Throughout this specification “S(O)” is a short hand notation for S═O. The term “sulfonyl” is used herein to refer to the sulfo-oxo group represented by the formula —S(O)2A1, where A1can be hydrogen or an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “sulfone” as used herein is represented by the formula A1S(O)2A2, where A1and A2can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “sulfoxide” as used herein is represented by the formula A1S(O)A2, where A1and A2can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “thiol” as used herein is represented by the formula —SH. “R1,” “R2,” “R3,” “Rn,” where n is an integer, as used herein can, independently, possess one or more of the groups listed above. For example, if R1is a straight chain alkyl group, one of the hydrogen atoms of the alkyl group can optionally be substituted with a hydroxyl group, an alkoxy group, an alkyl group, a halide, and the like. Depending upon the groups that are selected, a first group can be incorporated within second group or, alternatively, the first group can be pendant (i.e., attached) to the second group. For example, with the phrase “an alkyl group comprising an amino group,” the amino group can be incorporated within the backbone of the alkyl group. Alternatively, the amino group can be attached to the backbone of the alkyl group. The nature of the group(s) that is (are) selected will determine if the first group is embedded or attached to the second group. As described herein, compounds of the invention may contain “optionally substituted” moieties. In general, the term “substituted,” whether preceded by the term “optionally” or not, means that one or more hydrogen of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. In is also contemplated that, in certain aspects, unless expressly indicated to the contrary, individual substituents can be further optionally substituted (i.e., further substituted or unsubstituted). The term “stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain aspects, their recovery, purification, and use for one or more of the purposes disclosed herein. Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted” group are independently halogen; —(CH2)0-4R∘; —(CH2)0-4OR∘; —O(CH2)0-4R∘, —O—(CH2)0-4C(O)OR∘; —(CH2)0-4CH(OR∘)2; —(CH2)0-4SR∘; —(CH2)0-4Ph, which may be substituted with R∘; —(CH2)0-4O(CH2)0-1Ph which may be substituted with R∘; —CH═CHPh, which may be substituted with R∘; —(CH2)0-4O(CH2)0-1-pyridyl which may be substituted with R∘; —NO2; —CN; —N3; —(CH2)0-4N(R∘)2; —(CH2)0-4N(R∘)C(O)R∘; —N(R∘)C(S)R∘; —(CH2)0-4N(R∘)C(O)NR∘2; —N(R∘)C(S)NR∘2; —(CH2)0-4N(R∘)C(O)OR∘; —N(R∘)N(R∘)C(O)R∘; —N(R∘)N(R∘)C(O)NR∘2; —N(R∘)N(R∘)C(O)OR∘; —(CH2)0-4C(O)R∘; —C(S)R∘; —(CH2)0-4C(O)OR∘; —(CH2)0-4C(O)SR∘; —(CH2)0-4C(O)OSiR∘3; —(CH2)0-4OC(O)R∘; —OC(O)(CH2)0-4SR—, SC(S)SR∘; —(CH2)0-4SC(O)R∘; —(CH2)0-4C(O)NR∘2; —C(S)NR∘2; —C(S)SR∘; —(CH2)0-4OC(O)NR∘2; —C(O)N(OR∘)R∘; —C(O)C(O)R∘; —C(O)CH2C(O)R∘; —C(NOR∘)R∘; —(CH2)0-4SSR∘; —(CH2)0-4S(O)2R∘; —(CH2)0-4S(O)2OR∘; —(CH2)0-4OS(O)2R∘; —S(O)2NR∘2; —(CH2)0-4S(O)R∘; —N(R∘)S(O)2NR∘2; —N(R∘)S(O)2R∘; —N(OR∘)R∘; —C(NH)NR∘2; —P(O)2R∘; —P(O)R∘2; —OP(O)R∘2; —OP(O)(OR∘)2; SiR∘3; —(C1-4straight or branched alkylene)O—N(R∘)2; or —(C1-4straight or branched alkylene)C(O)O—N(R∘)2, wherein each R∘may be substituted as defined below and is independently hydrogen, C1-6aliphatic, —CH2Ph, —O(CH2)0-1Ph, —CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R∘, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below. Suitable monovalent substituents on R∘(or the ring formed by taking two independent occurrences of R∘together with their intervening atoms), are independently halogen, —(CH2)0-2R●, -(haloR●), —(CH2)0-2OH, —(CH2)0-2OR●, —(CH2)0-2CH(OR●)2; —O(haloR●), —CN, —N3, —(CH2)0-2C(O)R●, —(CH2)0-2C(O)OH, —(CH2)0-2C(O)OR●, —(CH2)0-2SR●, —(CH2)0-2SH, —(CH2)0-2NH2, —(CH2)0-2NHR●, —(CH2)0-2NR●2, —NO2, —SiR●3, —OSiR●3, —C(O)SR●, —(C1-4straight or branched alkylene)C(O)OR●, or —SSR●wherein each R●is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently selected from C1-4aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R∘include ═O and ═S. Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: ═O, ═S, ═NNR*2, ═NNHC(O)R*, ═NNHC(O)OR*, ═NNHS(O)2R*, ═NR*, ═NOR*, —O(C(R*2))2-3O—, or —S(C(R*2))2-3S—, wherein each independent occurrence of R* is selected from hydrogen, C1-6aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: —O(CR*2)2-3O—, wherein each independent occurrence of R* is selected from hydrogen, C1-6aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable substituents on the aliphatic group of R* include halogen, —R●, -(haloR●), —OH, —OR●, —O(haloR●), —CN, —C(O)OH, —C(O)OR●, —NH2, —NHR●, —NR●2, or —NO2, wherein each R●is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include —R†, —NR†2, —C(O)R†, —C(O)OR†, —C(O)C(O)R†, —C(O)CH2C(O)R†, —S(O)2R†, —S(O)2NR†2, —C(S)NR†2, —C(NH)NR†2, or —N(R†)S(O)2R†; wherein each R†is independently hydrogen, C1-6aliphatic which may be substituted as defined below, unsubstituted —OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R†, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable substituents on the aliphatic group of R†are independently halogen, —R●, -(haloR●), —OH, —OR●, —O(haloR●), —CN, —C(O)OH, —C(O)OR●, —NH2, —NHR●, —NR●2, or —NO2, wherein each R●is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. The term “leaving group” refers to an atom (or a group of atoms) with electron withdrawing ability that can be displaced as a stable species, taking with it the bonding electrons. Examples of suitable leaving groups include halides and sulfonate esters, including, but not limited to, triflate, mesylate, tosylate, and brosylate. The terms “hydrolysable group” and “hydrolysable moiety” refer to a functional group capable of undergoing hydrolysis, e.g., under basic or acidic conditions. Examples of hydrolysable residues include, without limitation, acid halides, activated carboxylic acids, and various protecting groups known in the art (see, for example, “Protective Groups in Organic Synthesis,” T. W. Greene, P. G. M. Wuts, Wiley-Interscience, 1999). The term “organic residue” defines a carbon-containing residue, i.e., a residue comprising at least one carbon atom, and includes but is not limited to the carbon-containing groups, residues, or radicals defined hereinabove. Organic residues can contain various heteroatoms, or be bonded to another molecule through a heteroatom, including oxygen, nitrogen, sulfur, phosphorus, or the like. Examples of organic residues include but are not limited alkyl or substituted alkyls, alkoxy or substituted alkoxy, mono or di-substituted amino, amide groups, etc. Organic residues can preferably comprise 1 to 18 carbon atoms, 1 to 15, carbon atoms, 1 to 12 carbon atoms, 1 to 8 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms. In a further aspect, an organic residue can comprise 2 to 18 carbon atoms, 2 to 15, carbon atoms, 2 to 12 carbon atoms, 2 to 8 carbon atoms, 2 to 4 carbon atoms, or 2 to 4 carbon atoms. A very close synonym of the term “residue” is the term “radical,” which as used in the specification and concluding claims, refers to a fragment, group, or substructure of a molecule described herein, regardless of how the molecule is prepared. For example, a 2,4-thiazolidinedione radical in a particular compound has the structure: regardless of whether thiazolidinedione is used to prepare the compound. In some embodiments the radical (for example an alkyl) can be further modified (i.e., substituted alkyl) by having bonded thereto one or more “substituent radicals.” The number of atoms in a given radical is not critical to the present invention unless it is indicated to the contrary elsewhere herein. “Organic radicals,” as the term is defined and used herein, contain one or more carbon atoms. An organic radical can have, for example, 1-26 carbon atoms, 1-18 carbon atoms, 1-12 carbon atoms, 1-8 carbon atoms, 1-6 carbon atoms, or 1-4 carbon atoms. In a further aspect, an organic radical can have 2-26 carbon atoms, 2-18 carbon atoms, 2-12 carbon atoms, 2-8 carbon atoms, 2-6 carbon atoms, or 2-4 carbon atoms. Organic radicals often have hydrogen bound to at least some of the carbon atoms of the organic radical. One example, of an organic radical that comprises no inorganic atoms is a 5, 6, 7, 8-tetrahydro-2-naphthyl radical. In some embodiments, an organic radical can contain 1-10 inorganic heteroatoms bound thereto or therein, including halogens, oxygen, sulfur, nitrogen, phosphorus, and the like. Examples of organic radicals include but are not limited to an alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, mono-substituted amino, di-substituted amino, acyloxy, cyano, carboxy, carboalkoxy, alkylcarboxamide, substituted alkylcarboxamide, dialkylcarboxamide, substituted dialkylcarboxamide, alkylsulfonyl, alkylsulfinyl, thioalkyl, thiohaloalkyl, alkoxy, substituted alkoxy, haloalkyl, haloalkoxy, aryl, substituted aryl, heteroaryl, heterocyclic, or substituted heterocyclic radicals, wherein the terms are defined elsewhere herein. A few non-limiting examples of organic radicals that include heteroatoms include alkoxy radicals, trifluoromethoxy radicals, acetoxy radicals, dimethylamino radicals and the like. Compounds described herein can contain one or more double bonds and, thus, potentially give rise to cis/trans (E/Z) isomers, as well as other conformational isomers. Unless stated to the contrary, the invention includes all such possible isomers, as well as mixtures of such isomers. Unless stated to the contrary, a formula with chemical bonds shown only as solid lines and not as wedges or dashed lines contemplates each possible isomer, e.g., each enantiomer and diastereomer, and a mixture of isomers, such as a racemic or scalemic mixture. Compounds described herein can contain one or more asymmetric centers and, thus, potentially give rise to diastereomers and optical isomers. Unless stated to the contrary, the present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. Mixtures of stereoisomers, as well as isolated specific stereoisomers, are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers. Many organic compounds exist in optically active forms having the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L or R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and 1 or (+) and (−) are employed to designate the sign of rotation of plane-polarized light by the compound, with (−) or meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these compounds, called stereoisomers, are identical except that they are non-superimposable mirror images of one another. A specific stereoisomer can also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture. Many of the compounds described herein can have one or more chiral centers and therefore can exist in different enantiomeric forms. If desired, a chiral carbon can be designated with an asterisk (*). When bonds to the chiral carbon are depicted as straight lines in the disclosed formulas, it is understood that both the (R) and (S) configurations of the chiral carbon, and hence both enantiomers and mixtures thereof, are embraced within the formula. As is used in the art, when it is desired to specify the absolute configuration about a chiral carbon, one of the bonds to the chiral carbon can be depicted as a wedge (bonds to atoms above the plane) and the other can be depicted as a series or wedge of short parallel lines is (bonds to atoms below the plane). The Cahn-Ingold-Prelog system can be used to assign the (R) or (S) configuration to a chiral carbon. When the disclosed compounds contain one chiral center, the compounds exist in two enantiomeric forms. Unless specifically stated to the contrary, a disclosed compound includes both enantiomers and mixtures of enantiomers, such as the specific 50:50 mixture referred to as a racemic mixture. The enantiomers can be resolved by methods known to those skilled in the art, such as formation of diastereoisomeric salts which may be separated, for example, by crystallization (see, CRC Handbook of Optical Resolutions via Diastereomeric Salt Formation by David Kozma (CRC Press, 2001)); formation of diastereoisomeric derivatives or complexes which may be separated, for example, by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic esterification; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support for example silica with a bound chiral ligand or in the presence of a chiral solvent. It will be appreciated that where the desired enantiomer is converted into another chemical entity by one of the separation procedures described above, a further step can liberate the desired enantiomeric form. Alternatively, specific enantiomers can be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation. Designation of a specific absolute configuration at a chiral carbon in a disclosed compound is understood to mean that the designated enantiomeric form of the compounds can be provided in enantiomeric excess (e.e.). Enantiomeric excess, as used herein, is the presence of a particular enantiomer at greater than 50%, for example, greater than 60%, greater than 70%, greater than 75%, greater than 80%, greater than 85%, greater than 90%, greater than 95%, greater than 98%, or greater than 99%. In one aspect, the designated enantiomer is substantially free from the other enantiomer. For example, the “R” forms of the compounds can be substantially free from the “S” forms of the compounds and are, thus, in enantiomeric excess of the “S” forms. Conversely, “S” forms of the compounds can be substantially free of “R” forms of the compounds and are, thus, in enantiomeric excess of the “R” forms. When a disclosed compound has two or more chiral carbons, it can have more than two optical isomers and can exist in diastereoisomeric forms. For example, when there are two chiral carbons, the compound can have up to four optical isomers and two pairs of enantiomers ((S,S)/(R,R) and (R,S)/(S,R)). The pairs of enantiomers (e.g., (S,S)/(R,R)) are mirror image stereoisomers of one another. The stereoisomers that are not mirror-images (e.g., (S,S) and (R,S)) are diastereomers. The diastereoisomeric pairs can be separated by methods known to those skilled in the art, for example chromatography or crystallization and the individual enantiomers within each pair may be separated as described above. Unless otherwise specifically excluded, a disclosed compound includes each diastereoisomer of such compounds and mixtures thereof. The compounds according to this disclosure may form prodrugs at hydroxyl or amino functionalities using alkoxy, amino acids, etc., groups as the prodrug forming moieties. For instance, the hydroxymethyl position may form mono-, di- or triphosphates and again these phosphates can form prodrugs. Preparations of such prodrug derivatives are discussed in various literature sources (examples are: Alexander et al., J. Med. Chem. 1988, 31, 318; Aligas-Martin et al., PCT WO 2000/041531, p. 30). The nitrogen function converted in preparing these derivatives is one (or more) of the nitrogen atoms of a compound of the disclosure. “Derivatives” of the compounds disclosed herein are pharmaceutically acceptable salts, prodrugs, deuterated forms, radio-actively labeled forms, isomers, solvates and combinations thereof. The “combinations” mentioned in this context are refer to derivatives falling within at least two of the groups: pharmaceutically acceptable salts, prodrugs, deuterated forms, radio-actively labeled forms, isomers, and solvates. Examples of radio-actively labeled forms include compounds labeled with tritium, phosphorous-32, iodine-129, carbon-11, fluorine-18, and the like. Compounds described herein comprise atoms in both their natural isotopic abundance and in non-natural abundance. The disclosed compounds can be isotopically-labeled or isotopically-substituted compounds identical to those described, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as2H,3H,13C,14C,15N,18O,17O,35S,18F, and36Cl, respectively. Compounds further comprise prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labeled compounds of the present invention, for example those into which radioactive isotopes such as3H and14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e.,3H, and carbon-14, i.e.,14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i.e.,2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labeled compounds of the present invention and prodrugs thereof can generally be prepared by carrying out the procedures below, by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent. The compounds described in the invention can be present as a solvate. In some cases, the solvent used to prepare the solvate is an aqueous solution, and the solvate is then often referred to as a hydrate. The compounds can be present as a hydrate, which can be obtained, for example, by crystallization from a solvent or from aqueous solution. In this connection, one, two, three or any arbitrary number of solvent or water molecules can combine with the compounds according to the invention to form solvates and hydrates. Unless stated to the contrary, the invention includes all such possible solvates. The term “co-crystal” means a physical association of two or more molecules which owe their stability through non-covalent interaction. One or more components of this molecular complex provide a stable framework in the crystalline lattice. In certain instances, the guest molecules are incorporated in the crystalline lattice as anhydrates or solvates, see e.g. “Crystal Engineering of the Composition of Pharmaceutical Phases. Do Pharmaceutical Co-crystals Represent a New Path to Improved Medicines?” Almarasson, O., et. al., The Royal Society of Chemistry, 1889-1896, 2004. Examples of co-crystals include p-toluenesulfonic acid and benzenesulfonic acid. It is also appreciated that certain compounds described herein can be present as an equilibrium of tautomers. For example, ketones with an α-hydrogen can exist in an equilibrium of the keto form and the enol form. Likewise, amides with an N-hydrogen can exist in an equilibrium of the amide form and the imidic acid form. As another example, pyrazoles can exist in two tautomeric forms, N1-unsubstituted, 3-A3and N1-unsubstituted, 5-A3as shown below. Unless stated to the contrary, the invention includes all such possible tautomers. It is known that chemical substances form solids that are present in different states of order which are termed polymorphic forms or modifications. The different modifications of a polymorphic substance can differ greatly in their physical properties. The compounds according to the invention can be present in different polymorphic forms, with it being possible for particular modifications to be metastable. Unless stated to the contrary, the invention includes all such possible polymorphic forms. In some aspects, a structure of a compound can be represented by a formula: which is understood to be equivalent to a formula: wherein n is typically an integer. That is, Rnis understood to represent five independent substituents, Rn(a), Rn(b), Rn(c), Rn(d), Rn(e). By “independent substituents,” it is meant that each R substituent can be independently defined. For example, if in one instance Rn(a)is halogen, then Rn(b)is not necessarily halogen in that instance. Certain materials, compounds, compositions, and components disclosed herein can be obtained commercially or readily synthesized using techniques generally known to those of skill in the art. For example, the starting materials and reagents used in preparing the disclosed compounds and compositions are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Acros Organics (Morris Plains, N.J.), Strem Chemicals (Newburyport, MA), Fisher Scientific (Pittsburgh, Pa.), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and supplemental volumes (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991); March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition); and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989). Unless otherwise expressly stated, it is in no way intended that any method set forth herein be construed as requiring that its steps be performed in a specific order. Accordingly, where a method claim does not actually recite an order to be followed by its steps or it is not otherwise specifically stated in the claims or descriptions that the steps are to be limited to a specific order, it is no way intended that an order be inferred, in any respect. This holds for any possible non-express basis for interpretation, including: matters of logic with respect to arrangement of steps or operational flow; plain meaning derived from grammatical organization or punctuation; and the number or type of embodiments described in the specification. Disclosed are the components to be used to prepare the compositions of the invention as well as the compositions themselves to be used within the methods disclosed herein. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds cannot be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular compound is disclosed and discussed and a number of modifications that can be made to a number of molecules including the compounds are discussed, specifically contemplated is each and every combination and permutation of the compound and the modifications that are possible unless specifically indicated to the contrary. Thus, if a class of molecules A, B, and C are disclosed as well as a class of molecules D, E, and F and an example of a combination molecule, A-D is disclosed, then even if each is not individually recited each is individually and collectively contemplated meaning combinations, A-E, A-F, B-D, B-E, B-F, C-D, C-E, and C-F are considered disclosed. Likewise, any subset or combination of these is also disclosed. Thus, for example, the sub-group of A-E, B-F, and C-E would be considered disclosed. This concept applies to all aspects of this application including, but not limited to, steps in methods of making and using the compositions of the invention. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the methods of the invention. It is understood that the compositions disclosed herein have certain functions. Disclosed herein are certain structural requirements for performing the disclosed functions, and it is understood that there are a variety of structures that can perform the same function that are related to the disclosed structures, and that these structures will typically achieve the same result. B. COMPOUNDS In one aspect, the invention relates to compounds useful in treating disorders associated with a viral infection due to, in particular, an Alphavirus (e.g., Chikungunya virus (CHIKV), Ross River virus, Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), and Western equine encephalitis (WEEV)), a Flavivirus (e.g., dengue virus (DENV), West Nile virus (WNV), zika virus (ZIKV), tick-borne encephalitis virus, and yellow fever virus), and a Coronavirus (e.g., Middle East Respiratory Syndromes coronavirus (MERS-CoV), Severe Acute Respiratory Syndrome coronavirus (SARS-CoV), and SARS-CoV-2). In one aspect, the disclosed compounds exhibit antiviral activity. In one aspect, the compounds of the invention are useful in inhibiting viral activity in a mammal. In a further aspect, the compounds of the invention are useful in inhibiting viral activity in at least one cell. In one aspect, the compounds of the invention are useful in the treatment of viral infections, as further described herein. It is contemplated that each disclosed derivative can be optionally further substituted. It is also contemplated that any one or more derivative can be optionally omitted from the invention. It is understood that a disclosed compound can be provided by the disclosed methods. It is also understood that the disclosed compounds can be employed in the disclosed methods of using. 1. Structure In one aspect, disclosed are compounds having a structure represented by a formula: wherein R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, and —P(O)(OR14a)(OR14b); wherein R10, when present, is selected from C1-C20 alkyl and C2-C20 alkenyl; wherein R11, when present, is an amino acid derivative side chain; wherein R12, when present, is selected from C1-C6 alkyl and C3-C6 cycloalkyl; wherein R13, when present, is selected from C1-C8 alkyl, C3-C8 cycloalkyl, Ar2, and —CH2Ar2; wherein Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; wherein each of R14aand R14b, when present, is independently selected from hydrogen and C1-C8 alkyl; and wherein Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; and wherein R2is a structure represented by a formula selected from: and wherein R15, when present, is selected from hydrogen, —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), and —C(O)(C2-C20 alkenyl), provided that when R2is then R1is —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R11, or —P(O)(OR14a)(OR14b), and provided that when R1is hydrogen, then R15is —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), or —C(O)(C2-C20 alkenyl), or a pharmaceutically acceptable salt thereof. In one aspect, disclosed are compounds having a structure: or a pharmaceutically acceptable salt thereof. In a further aspect, the compound has a structure represented by a formula selected from: In a further aspect, the compound is selected from: In a further aspect, the compound has a structure represented by a formula selected from: In a further aspect, the compound has a structure represented by a formula selected from: In a further aspect, the compound has a structure represented by a formula: In a further aspect, the compound is selected from: In a further aspect, the compound is: a. R1Groups In one aspect, R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, and —P(O)(OR14a)(OR14b). In a further aspect, R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, and —P(O)(OAr1)NHCH(R12)CO2R13. In a still further aspect, R1is selected from hydrogen, —C(O)R10, and —C(O)CH(R11)NH2. In yet a further aspect, R1is selected from hydrogen and —C(O)R10. In an even further aspect, R1is hydrogen. In various aspects, R1is selected from —C(O)R10and —C(O)CH(R11)NH2. In a further aspect, R1is —C(O)R10. In a still further aspect, R1is —C(O)CH(R11)NH2. In various aspects, R1is selected from —P(O)(OAr1)NHCH(R12)CO2R13and —P(O)(OR14a)(OR14b). In a further aspect, R1is —P(O)(OAr1)NHCH(R12)CO2R13. In a still further aspect, R1is —P(O)(OR14a)(OR14b). b. R2Groups In one aspect, R2is a structure represented by a formula selected from: In a further aspect, R2is a structure represented by a formula: In a further aspect, R2is a structure represented by a formula: c. R10Groups In one aspect, R10, when present, is selected from C1-C20 alkyl and C2-C20 alkenyl. In a further aspect, R10, when present, is selected from C1-C16 alkyl and C2-C16 alkenyl. In a still further aspect, R10, when present, is selected from C1-C12 alkyl and C2-C12 alkenyl. In yet a further aspect, R10, when present, is selected from C1-C8 alkyl and C2-C8 alkenyl. In an even further aspect, R10, when present, is selected from C1-C4 alkyl and C2-C4 alkenyl. In a still further aspect, R10, when present, is selected from methyl, ethyl, n-propyl, isopropyl, ethenyl, n-propenyl, and isopropenyl. In yet a further aspect, R10, when present, is selected from methyl, ethyl, and ethenyl. In various aspects, R10, when present, is C1-C20 alkyl. In a further aspect, R10, when present, is C1-C16 alkyl. In a still further aspect, R10, when present, is C1-C12 alkyl. In yet a further aspect, R10, when present, is C1-C8 alkyl. In an even further aspect, R10, when present, is C1-C4 alkyl. In a still further aspect, R10, when present, is selected from methyl, ethyl, and n-propyl. In yet a further aspect, R10, when present, is selected from methyl and ethyl. In an even further aspect, R10, when present, is ethyl. In a still further aspect, R10, when present, is methyl. In various aspects, R10, when present, is C2-C20 alkenyl. In a further aspect, R10, when present, is C2-C16 alkenyl. In a still further aspect, R10, when present, is C2-C12 alkenyl. In yet a further aspect, R10, when present, is C2-C8 alkenyl. In an even further aspect, R10, when present, is C2-C4 alkenyl. In a still further aspect, R10, when present, is selected from ethenyl, n-propenyl, and isopropyl. In yet a further aspect, R10, when present, is ethenyl. d. R11Groups In one aspect, R11, when present, is an amino acid derivative side chain. Amino acid derivative side chains are well-known by those of skill in the art. See, for example, Tsume, et al. (2014) “The development of orally administrable gemcitabine prodrugs with D-enantiomer amino acid: Enhanced membrane permeability and enzymatic stability,”Eur. J. Pharm. Sci.86: 514-523; Zhang, et al. (2013) “A Carrier-Mediated Prodrug Approach To Improve the Oral Absorption of Antileukemic Drug Decitabine,”Mol. Pharmaceutics10: 3195-3202; Vig, et al. (2013) “Amino acids as promoities in prodrug design and development,”Adv. Drug Deliv. Rev.65: 1370-1385; Hasabelnaby, et al. (2012) “Synthesis, chemical and enzymatic hydrolysis, and aqueous solubility of amino acid ester prodrugs of 3-carboranyl thymidine analogs for boron neutron capture therapy of brain tumors,”Eur. J. Med. Chem.55: 325-334; Song, et al. (2005) “Amino acid ester prodrug of the anticancer agent gemcitabine: Synthesis, bioconversion, metabolic bioevasion and hPEPT1-mediated transport,”Mol. Pharm.2: 157-167; Landowski, et al. (2005) “Targeted delivery to PEPT1-overexpresssing cells: Acidic, basic, and secondary floxuridine amino acid ester prodrugs,”Mol. Cancer Ther.4: 659-667; Landowski, et al. (2005) “Floxuridine amino acid ester prodrugs: Enhancing Caco-2 permeability and resistance to glycosidic bond metabolism,”Pharm. Res.22: 1510-1518; Sugawara, et al. (2000) “Transport of valganciclovir, a ganciclovir prodrug, via peptide transporters PEPT1 and PEPT2,” J. Pharm. Sci.89: 781-789; Guo, et al. (1999) “Interactions of a nonpeptidic drug, valacyclovir, with the human intestinal peptide transporter (hPEPT1) expressed in a mammalian cell line,”J. Pharmacol. Exp. Ther.289: 448-454; Balimane, et al. (1998) “Direct evidence for peptide transporter (PepT1)-mediated uptake of a nonpeptide prodrug, valacyclovir,”Biochem. Biophys. Res. Commun.250: 246-251; de Vrueh, et al. (1998) “Transport of Lvalineacyclovir via the oligopeptide transporter in the human intestinal cell line, Caco-2,” J. Pharmacol. Exp. Ther.286: 1166-1170. In various aspects, the amino acid derivative side chain can be a derivative of a synthetic or naturally-occurring amino acid. Thus, in a further aspect, the amino acid is a synthetic amino acid. In a still further aspect, the amino acid is a naturally-occurring amino acid. Examples of naturally-occurring amino acids include, but are not limited to, arginine, histidine, lysine, aspartic acid, glutamic acid, serine, threonine, asparagine, glutamine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan. In various further aspects, the amino acid derivative side chain is a derivative of alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, or tryptophan. In a still further aspect, the amino acid derivative side chain is a derivative of alanine, valine, isoleucine, or leucine. In yet a further aspect, the amino acid derivative side chain is a derivative of isoleucine or leucine. e. R12Groups In one aspect, R12, when present, is selected from C1-C6 alkyl and C3-C6 cycloalkyl. In a further aspect, R12, when present, is selected from C1-C4 alkyl and C3-C4 cycloalkyl. In a still further aspect, R12, when present, is selected from methyl, ethyl, n-propyl, isopropyl, and cyclopropyl. In various aspects, R12, when present, is C1-C6 alkyl. In a further aspect, R2, when present, is C1-C4 alkyl. In a still further aspect, R12, when present, is selected from methyl, ethyl, n-propyl, and isopropyl. In yet a further aspect, R12, when present, is selected from methyl and ethyl. In an even further aspect, R12, when present, is ethyl. In a still further aspect, R12, when present, is methyl. In various aspects, R12, when present, is selected from cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In a further aspect, R12, when present, is selected from cyclopropyl, cyclobutyl, and cyclopentyl. In a still further aspect, R12, when present, is selected from cyclopropyl and cyclobutyl. In yet a further aspect, R12, when present, is cyclopropyl. f. R13Groups In one aspect, R13, when present, is selected from C1-C8 alkyl, C3-C8 cycloalkyl, Ar2, and —CH2Ar2. In a further aspect, R13, when present, is selected from C1-C4 alkyl, C3-C6 cycloalkyl, Ar2, and —CH2Ar2. In a still further aspect, R13, when present, is selected from methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, Ar2, and —CH2Ar2. In yet a further aspect, R13, when present, is selected from methyl, ethyl, cyclopropyl, Ar2, and —CH2Ar2. In various aspects, R13, when present, is selected from C1-C8 alkyl and C3-C8 cycloalkyl. In a further aspect, R13, when present, is selected from C1-C4 alkyl, C3-C6 cycloalkyl. In a still further aspect, R13, when present, is selected from methyl, ethyl, n-propyl, isopropyl, cyclopropyl, and cyclobutyl. In yet a further aspect, R13, when present, is selected from methyl, ethyl, cyclopropyl. In various aspects, R13, when present, is C1-C8 alkyl. In a further aspect, R13, when present, is C1-C4 alkyl. In a still further aspect, R13, when present, is selected from methyl, ethyl, n-propyl, and isopropyl. In yet a further aspect, R13, when present, is selected from methyl and ethyl. In an even further aspect, R13, when present, is ethyl. In a still further aspect, R13, when present, is methyl. In various aspects, R13, when present, is C3-C8 cycloalkyl. In a further aspect, R13, when present, is selected from cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In a still further aspect, R13, when present, is selected from cyclopropyl, cyclobutyl, and cyclopentyl. In yet a further aspect, R13, when present, is selected from cyclopropyl and cyclobutyl. In an even further aspect, R13, when present, is selected from cyclopropyl. In various aspects, R13, when present, is selected from Ar2and —CH2Ar2. In a further aspect, R13, when present, is Ar2. In a still further aspect, R13, when present, is —CH2Ar2. g. R14aand R14bGroups In one aspect, each of R14aand R14b, when present, is independently selected from hydrogen and C1-C8 alkyl. In a further aspect, each of R14aand R14b, when present, is independently selected from hydrogen and C1-C6 alkyl. In a still further aspect, each of R14aand R14b, when present, is independently selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and tert-butyl. In yet a further aspect, each of R14aand R14b, when present, is independently selected from hydrogen, methyl, ethyl, n-propyl, and isopropyl. In an even further aspect, each of R14aand R14b, when present, is independently selected from hydrogen, methyl, and ethyl. In a still further aspect, each of R14aand R14b, when present, is independently selected from hydrogen and ethyl. In yet a further aspect, each of R14aand R14b, when present, is independently selected from hydrogen and methyl. In various aspects, each of R14aand R14b, when present, is independently C1-C8 alkyl. In a further aspect, each of R14aand R14b, when present, is independently C1-C6 alkyl. In a still further aspect, each of R14aand R14b, when present, is independently selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and tert-butyl. In yet a further aspect, each of R14aand R14b, when present, is independently selected from methyl, ethyl, n-propyl, and isopropyl. In an even further aspect, each of R14aand R14b, when present, is independently selected from methyl and ethyl. In a still further aspect, each of R14aand R14b, when present, is ethyl. In yet a further aspect, each of R14aand R14b, when present, is methyl. In various aspects, each of R14aand R14b, when present, is hydrogen. h. R15Groups In one aspect, R15, when present, is selected from hydrogen, —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), and —C(O)(C2-C20 alkenyl). In a further aspect, R15, when present, is selected from hydrogen, —C(O)(C1-C16 alkyl), and —C(O)(C2-C16 alkenyl). In a still further aspect, R15, when present, is selected from hydrogen, —C(O)(C1-C12 alkyl), and —C(O)(C2-C12 alkenyl). In yet a further aspect, R15, when present, is selected from hydrogen, —C(O)(C1-C8 alkyl), and —C(O)(C2-C8 alkenyl). In an even further aspect, R15, when present, is selected from hydrogen, —C(O)(C1-C4 alkyl), and —C(O)(C2-C4 alkenyl). In a still further aspect, R15, when present, is selected from hydrogen, —C(O)CH3, —C(O)CH2CH3, —C(O)CH2CH2CH3, —C(O)(CH)(CH3)2, —C(O)CH═CH2, —C(O)CH═CHCH3, —C(O)CH2CH═CH2, and —C(O)(CH3)C═CH2. In yet a further aspect, R15, when present, is selected from hydrogen, —C(O)CH3, —C(O)CH2CH3, and —C(O)CH═CH2. In various aspects, R15, when present, is selected from hydrogen and —C(O)(C1-C20 alkyl). In a further aspect, R15, when present, is selected from hydrogen and —C(O)(C1-C16 alkyl). In a still further aspect, R15, when present, is selected from hydrogen and —C(O)(C1-C12 alkyl). In yet a further aspect, R15, when present, is selected from hydrogen and —C(O)(C1-C8 alkyl). In an even further aspect, R15, when present, is selected from hydrogen and —C(O)(C1-C4 alkyl). In a still further aspect, R15, when present, is selected from hydrogen, —C(O)CH3, —C(O)CH2CH3, —C(O)CH2CH2CH3, and —C(O)(CH)(CH3)2. In yet a further aspect, R15, when present, is selected from hydrogen, —C(O)CH3, and —C(O)CH2CH3. In an even further aspect, R15, when present, is selected from hydrogen and —C(O)CH3. In various aspects, R15, when present, is —C(O)(C1-C20 alkyl). In a further aspect, R15, when present, is —C(O)(C1-C16 alkyl). In a still further aspect, R15, when present, is —C(O)(C1-C12 alkyl). In yet a further aspect, R15, when present, is —C(O)(C1-C8 alkyl). In an even further aspect, R15, when present, is —C(O)(C1-C4 alkyl). In a still further aspect, R15, when present, is selected from —C(O)CH3, —C(O)CH2CH3, —C(O)CH2CH2CH3, and —C(O)(CH)(CH3)2. In yet a further aspect, R15, when present, is selected from —C(O)CH3and —C(O)CH2CH3. In an even further aspect, R15, when present, is —C(O)CH3. In various aspects, R15, when present, is selected from hydrogen and —C(O)(C2-C20 alkenyl). In a further aspect, R15, when present, is selected from hydrogen and —C(O)(C2-C16 alkenyl). In a still further aspect, R15, when present, is selected from hydrogen and —C(O)(C2-C12 alkenyl). In yet a further aspect, R15, when present, is selected from hydrogen and —C(O)(C2-C8 alkenyl). In an even further aspect, R15, when present, is selected from hydrogen and —C(O)(C2-C4 alkenyl). In a still further aspect, R15, when present, is selected from hydrogen, —C(O)CH═CH2, —C(O)CH═CHCH3, —C(O)CH2CH═CH2, and —C(O)(CH3)C═CH2. In yet a further aspect, R15, when present, is selected from hydrogen and —C(O)CH═CH2. In various aspects, R13, when present, is —C(O)(C2-C20 alkenyl). In a further aspect, R15, when present, is —C(O)(C2-C16 alkenyl). In a still further aspect, R15, when present, is —C(O)(C2-C12 alkenyl). In yet a further aspect, R15, when present, is —C(O)(C2-C8 alkenyl). In an even further aspect, R15, when present, is —C(O)(C2-C4 alkenyl). In a still further aspect, R15, when present, is selected from —C(O)CH═CH2, —C(O)CH═CHCH3, —C(O)CH2CH═CH2, and —C(O)(CH3)C═CH2. In yet a further aspect, R15, when present, is —C(O)CH═CH2. In various aspects, R13, when present, is —C(O)(C3-C6 cycloalkyl). In a further aspect, R15, when present, is selected from —C(O)(cyclopropyl), —C(O)(cyclobutyl), and —C(O)(cyclopentyl). In a still further aspect, R15, when present, is selected from —C(O)(cyclopropyl) and —C(O)(cyclobutyl). In yet a further aspect, R15, when present, is —C(O)(cyclobutyl). In an even further aspect, R15, when present, is —C(O)(cyclopropyl). In various aspects, R13, when present, is hydrogen. i. Ar1Groups In one aspect, Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In a further aspect, Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, or 2 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In a still further aspect, Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0 or 1 group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In yet a further aspect, Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is monosubstituted with a group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In an even further aspect, Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is unsubstituted. In various aspects, Ar1, when present, is C6-C14 aryl substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. Examples of C6-C14 aryls include, but are not limited to, phenyl, naphthyl, anthracenyl, and phenanthrenyl. In a further aspect, Ar1, when present, is C6-C14 aryl substituted with 0, 1, or 2 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In a still further aspect, Ar1, when present, is C6-C14 aryl substituted with 0 or 1 group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In yet a further aspect, Ar1, when present, is C6-C14 aryl monosubstituted with a group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In an even further aspect, Ar1, when present, is unsubstituted C6-C14 aryl. In various aspects, Ar1, when present, is C2-C10 heteroaryl substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. Examples of C2-C10 heteroaryls include, but are not limited to, furanyl, thiophenyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, thiazolyl, pyridinyl, pyrimidinyl, imidazolyl, purinyl, indolyl, and quinolinyl. In a further aspect, Ar1, when present, is C2-C10 heteroaryl substituted with 0, 1, or 2 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In a still further aspect, Ar1, when present, is C2-C10 heteroaryl substituted with 0 or 1 group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In yet a further aspect, Ar1, when present, is C2-C10 heteroaryl monosubstituted with a group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In an even further aspect, Ar1, when present, is unsubstituted C2-C10 heteroaryl. j. Ar2Groups In one aspect, Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In a further aspect, Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, or 2 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In a still further aspect, Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0 or 1 group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In yet a further aspect, Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is monosubstituted with a group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In an even further aspect, Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is unsubstituted. In various aspects, Ar2, when present, is C6-C14 aryl substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. Examples of C6-C14 aryls include, but are not limited to, phenyl, naphthyl, anthracenyl, and phenanthrenyl. In a further aspect, Ar2, when present, is C6-C14 aryl substituted with 0, 1, or 2 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In a still further aspect, Ar2, when present, is C6-C14 aryl substituted with 0 or 1 group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In yet a further aspect, Ar2, when present, is C6-C14 aryl monosubstituted with a group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In an even further aspect, Ar2, when present, is unsubstituted C6-C14 aryl. In various aspects, Ar2, when present, is C2-C10 heteroaryl substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. Examples of C2-C10 heteroaryls include, but are not limited to, furanyl, thiophenyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, thiazolyl, pyridinyl, pyrimidinyl, imidazolyl, purinyl, indolyl, and quinolinyl. In a further aspect, Ar2, when present, is C2-C10 heteroaryl substituted with 0, 1, or 2 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In a still further aspect, Ar2, when present, is C2-C10 heteroaryl substituted with 0 or 1 group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In yet a further aspect, Ar2, when present, C2-C10 heteroaryl monosubstituted with a group selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl. In an even further aspect, Ar2, when present, is unsubstituted C2-C10 heteroaryl. 2. Example Compounds In one aspect, a compound can be present as one or more of the following structures: or a pharmaceutically acceptable salt thereof. In one aspect, a compound can be present as one or more of the following structures: or a pharmaceutically acceptable salt thereof. In one aspect, a compound can be present as one or more of the following structures: or a pharmaceutically acceptable salt thereof. In one aspect, a compound can be present as the following structure: or a pharmaceutically acceptable salt thereof. 3. Prophetic Compound Examples The following compound examples are prophetic, and can be prepared using the synthesis methods described herein above and other general methods as needed as would be known to one skilled in the art. It is anticipated that the prophetic compounds would be active as inhibitors of a viral infection, and such activity can be determined using the assay methods described herein below. In one aspect, a compound can be selected from: It is contemplated that one or more compounds can optionally be omitted from the disclosed invention. It is understood that the disclosed compounds can be used in connection with the disclosed methods, compositions, kits, and uses. It is understood that pharmaceutical acceptable derivatives of the disclosed compounds can be used also in connection with the disclosed methods, compositions, kits, and uses. The pharmaceutical acceptable derivatives of the compounds can include any suitable derivative, such as pharmaceutically acceptable salts as discussed below, isomers, radiolabeled analogs, tautomers, and the like. C. PHARMACEUTICAL COMPOSITIONS In one aspect, disclosed are pharmaceutical compositions comprising a disclosed compound, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. In one aspect, disclosed are pharmaceutical compositions comprising pharmaceutically acceptable carrier and a therapeutically effective amount of at least one compound having a structure represented by a formula: wherein R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, and —P(O)(OR14a)(OR14b); wherein R10, when present, is selected from C1-C20 alkyl and C2-C20 alkenyl; wherein R1, when present, is an amino acid derivative side chain; wherein R12, when present, is selected from C1-C6 alkyl and C3-C6 cycloalkyl; wherein R13, when present, is selected from C1-C8 alkyl, C3-C8 cycloalkyl, Ar2, and —CH2Ar2; wherein Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; wherein each of R14aand R14b, when present, is independently selected from hydrogen and C1-C8 alkyl; and wherein Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; and wherein R2is a structure represented by a formula selected from: wherein R15, when present, is selected from hydrogen, —C(O)(C1-C20 alkyl), and —C(O)(C2-C20 alkenyl), provided that when R2is then R1is —C(O)CH(R11)NH2— P(O)(OAr1)NHCH(R12)CO2R13, or —P(O)(OR14a)(OR14b), and provided that when R1is hydrogen, then R15is —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl) or —C(O)(C2-C20 alkenyl), or a pharmaceutically acceptable salt thereof. In one aspect, disclosed are pharmaceutical compositions comprising pharmaceutically acceptable carrier and a therapeutically effective amount of at least one compound having a structure: or a pharmaceutically acceptable salt thereof. In various aspects, the compounds and compositions of the invention can be administered in pharmaceutical compositions, which are formulated according to the intended method of administration. The compounds and compositions described herein can be formulated in a conventional manner using one or more physiologically acceptable carriers or excipients. For example, a pharmaceutical composition can be formulated for local or systemic administration, e.g., administration by drops or injection into the ear, insufflation (such as into the ear), intravenous, topical, or oral administration. The nature of the pharmaceutical compositions for administration is dependent on the mode of administration and can readily be determined by one of ordinary skill in the art. In various aspects, the pharmaceutical composition is sterile or sterilizable. The therapeutic compositions featured in the invention can contain carriers or excipients, many of which are known to skilled artisans. Excipients that can be used include buffers (for example, citrate buffer, phosphate buffer, acetate buffer, and bicarbonate buffer), amino acids, urea, alcohols, ascorbic acid, phospholipids, polypeptides (for example, serum albumin), EDTA, sodium chloride, liposomes, mannitol, sorbitol, water, and glycerol. The nucleic acids, polypeptides, small molecules, and other modulatory compounds featured in the invention can be administered by any standard route of administration. For example, administration can be parenteral, intravenous, subcutaneous, or oral. A modulatory compound can be formulated in various ways, according to the corresponding route of administration. For example, liquid solutions can be made for administration by drops into the ear, for injection, or for ingestion; gels or powders can be made for ingestion or topical application. Methods for making such formulations are well known and can be found in, for example, Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, Pa. 1990. In various aspects, the disclosed pharmaceutical compositions comprise the disclosed compounds (including pharmaceutically acceptable salt(s) thereof) as an active ingredient, a pharmaceutically acceptable carrier, and, optionally, other therapeutic ingredients or adjuvants. The instant compositions include those suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy. In various aspects, the pharmaceutical compositions of this invention can include a pharmaceutically acceptable carrier and a compound or a pharmaceutically acceptable salt of the compounds of the invention. The compounds of the invention, or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds. The pharmaceutical carrier employed can be, for example, a solid, liquid, or gas. Examples of solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Examples of liquid carriers are sugar syrup, peanut oil, olive oil, and water. Examples of gaseous carriers include carbon dioxide and nitrogen. In preparing the compositions for oral dosage form, any convenient pharmaceutical media can be employed. For example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like can be used to form oral liquid preparations such as suspensions, elixirs and solutions; while carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like can be used to form oral solid preparations such as powders, capsules and tablets. Because of their ease of administration, tablets and capsules are the preferred oral dosage units whereby solid pharmaceutical carriers are employed. Optionally, tablets can be coated by standard aqueous or nonaqueous techniques. A tablet containing the composition of this invention can be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants. Compressed tablets can be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. The pharmaceutical compositions of the present invention comprise a compound of the invention (or pharmaceutically acceptable salts thereof) as an active ingredient, a pharmaceutically acceptable carrier, and optionally one or more additional therapeutic agents or adjuvants. The instant compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy. Pharmaceutical compositions of the present invention suitable for parenteral administration can be prepared as solutions or suspensions of the active compounds in water. A suitable surfactant can be included such as, for example, hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms. Pharmaceutical compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability. The pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof. Pharmaceutical compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, mouth washes, gargles, and the like. Further, the compositions can be in a form suitable for use in transdermal devices. These formulations can be prepared, utilizing a compound of the invention, or pharmaceutically acceptable salts thereof, via conventional processing methods. As an example, a cream or ointment is prepared by mixing hydrophilic material and water, together with about 5 wt % to about 10 wt % of the compound, to produce a cream or ointment having a desired consistency. Pharmaceutical compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories can be conveniently formed by first admixing the composition with the softened or melted carrier(s) followed by chilling and shaping in molds. In addition to the aforementioned carrier ingredients, the pharmaceutical formulations described above can include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti-oxidants) and the like. Furthermore, other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient. Compositions containing a compound of the invention, and/or pharmaceutically acceptable salts thereof, can also be prepared in powder or liquid concentrate form. In a further aspect, an effective amount is a therapeutically effective amount. In a still further aspect, an effective amount is a prophylactically effective amount. In a further aspect, the pharmaceutical composition is administered to a mammal. In a still further aspect, the mammal is a human. In an even further aspect, the human is a patient. In a further aspect, the pharmaceutical composition is used to treat a viral infection such as, for example, chikungunya, Venezuelan equine encephalitis, dengue, influenza, and zika. It is understood that the disclosed compositions can be prepared from the disclosed compounds. It is also understood that the disclosed compositions can be employed in the disclosed methods of using. D. METHODS OF MAKING A COMPOUND The compounds of this invention can be prepared by employing reactions as shown in the following schemes, in addition to other standard manipulations that are known in the literature, exemplified in the experimental sections or clear to one skilled in the art. For clarity, examples having a single substituent are shown where multiple substituents are allowed under the definitions disclosed herein. Reactions used to generate the compounds of this invention are prepared by employing reactions as shown in the following Reaction Schemes, as described and exemplified below. In certain specific examples, the disclosed compounds can be prepared by Routes I-VII, as described and exemplified below. The following examples are provided so that the invention might be more fully understood, are illustrative only, and should not be construed as limiting. 1. Route I In one aspect, substituted 6-aza nucoleoside prodrugs can be prepared as shown below. Compounds are represented in generic form, where PG is an amine protecting group, LG is a leaving group, and with other substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below. In one aspect, compounds of type 1.20, and similar compounds, can be prepared according to reaction Scheme 1B above. Thus, compounds of type 1.13 can be prepared by coupling an appropriate carboxylic acid, e.g., 1.11 as shown above, and an appropriate alcohol, e.g., 1.12 as shown above. Appropriate carboxylic acids and appropriate alcohols are commercially available or prepared by methods known to one skilled in the art. The coupling reaction is carried out in the presence of an appropriate coupling agent, e.g., N,N′-dicyclohexylcarbodiimide (DCC), and an appropriate activating agent, e.g., 4-dimethylaminopyridine (DMAP), in an appropriate solvent, e.g., dimethylformamide (DMF). Compounds of type 1.14 can be prepared deprotection of an appropriate amine, e.g., 1.13 as shown above. The deprotection is carried out in the presence of an appropriate deprotecting agent, e.g., 4N HCl (5 eq) in dioxange, in an appropriate solvent, e.g., dichloromethane (DCM). Compounds of type 1.16 can be prepared by coupling reaction of an appropriate amine, e.g., 1.14 as shown above, and an appropriate phosphoryl chloride, e.g., 1.15 as shown above. Appropriate phosphoryl chlorides are commercially available or prepared by methods known to one skilled in the art. The coupling reaction is carried out in the presence of an appropriate base, e.g., triethylamine (TEA), in an appropriate solvent, e.g., DCM. Compounds of type 1.18 can be prepared by activation of an appropriate organophosphorous compound, e.g., 1.16 as shown above. Appropriate organophosphorous compounds are commercially available or prepared by methods known to one skilled in the art. The activation is carried out in the presence of an appropriate alcohol, e.g., 1.17 as shown above, in the presence of an appropriate base, e.g., TEA, in an appropriate solvent, e.g., DCM. Appropriate alcohols are commercially available or prepared by methods known to one skilled in the art. Compounds of type 1.20 can be prepared by displacement of a leaving group on an appropriate activated organophosphorous compound, e.g., 1.18 as shown above. The displacement reaction is carried out in the presence of an appropriate primary alcohol, e.g., 1.19 as shown above, an appropriate Lewis acid, e.g., trimethylaluminum, and an appropriate base, e.g., pyridine, in an appropriate solvent, e.g., N,N′-dimethylpropyleneurea (DMPU). Appropriate primary alcohols are commercially available or prepared by methods known to one skilled in the art. As can be appreciated by one skilled in the art, the above reaction provides an example of a generalized approach wherein compounds similar in structure to the specific reactants above (compounds similar to compounds of type 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, and 1.9), can be substituted in the reaction to provide substituted 6-aza nucoleoside prodrug derivatives similar to Formula 1.10. 2. Route II In one aspect, substituted 6-aza nucoleoside prodrugs can be prepared as shown below. Compounds are represented in generic form, with substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below. In one aspect, compounds of type 2.4, and similar compounds, can be prepared according to reaction Scheme 2B above. Thus, compounds of type 2.4 can be prepared by hydrolysis of an appropriate aryl alcohol, e.g., 2.3 as shown above. Appropriate aryl alcohols are commercially available or prepared by methods known to one skilled in the art. The hydrolysis is carried out in the presence of an appropriate hydrolysing agent, e.g., phosphorus oxychloride, and an appropriate base, e.g., TEA, in an appropriate solvent, e.g., DCM. As can be appreciated by one skilled in the art, the above reaction provides an example of a generalized approach wherein compounds similar in structure to the specific reactants above (compounds similar to compounds of type 2.1), can be substituted in the reaction to provide substituted 6-aza nucoleoside prodrug derivatives similar to Formula 2.2. 3. Route III In one aspect, substituted 6-aza nucoleoside prodrugs can be prepared as shown below. Compounds are represented in generic form, wherein PG is an amine protecting group, and with other substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below. In one aspect, compounds of type 3.12, and similar compounds, can be prepared according to reaction Scheme 3B above. Thus, compounds of type 3.8 can be prepared by protection of an appropriate diol, e.g., 3.7 as shown above. Appropriate diols are commercially available or prepared by methods known to one skilled in the art. The protection is carried out in the presence of an appropriate catalyst, e.g., copper sulfate, an appropriate acid, e.g., catalytic sulphuric acid, in an appropriate solvent, e.g., acetone, for an appropriate period of time, e.g., 24 h. Compounds of type 3.10 can be prepared by coupling an appropriate alcohol, e.g., 3.8, and an appropriate carboxylic acid, e.g., 3.9 as shown above. Appropriate carboxylic acids are commercially available or prepared by methods known to one skilled in the art. The coupling reaction is carried out in the presence of an appropriate coupling agent, e.g., 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC), and an appropriate activating agent, e.g., DMAP, in an appropriate solvent, e.g., DCM. Compounds of type 3.11 can be prepared by deprotection of an appropriate acetal, e.g., 3.10 as shown above. The deprotection reaction is carried out in the presence of an appropriate acid, e.g., acetic acid, at an appropriate temperature, e.g., 65° C., for an appropriate period of time, e.g., 1 h. Compounds of type 3.12 can be prepared by deprotection of an appropriate amine, e.g., 3.11 as shown above. The deprotection reaction is carried out in the presence of an appropriate base, e.g., 3% piperidine, in an appropriate solvent, e.g., DMF. As can be appreciated by one skilled in the art, the above reaction provides an example of a generalized approach wherein compounds similar in structure to the specific reactants above (compounds similar to compounds of type 3.1, 3.2, 3.3, 3.4, and 3.5), can be substituted in the reaction to provide substituted 6-aza nucoleoside prodrug derivatives similar to Formula 3.6. 4. Route IV In one aspect, substituted 6-aza nucoleoside prodrugs can be prepared as shown below. Compounds are represented in generic form, where X is a halogen, and with other substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below. In one aspect, compounds of type 4.8, and similar compounds, can be prepared according to reaction Scheme 4B above. Thus, compounds of type 4.7 can be prepared by coupling an appropriate alcohol, e.g., 4.5 as shown above, and an appropriate acyl halide, e.g., 4.6 as shown above. Appropriate alcohols and appropriate acyl halides are commercially available or prepared by methods known to one skilled in the art. The coupling reaction is carried out in the presence of an appropriate base, e.g., pyridine, over an appropriate temperature range, e.g., 0° C. to rt, for an appropriate period of time, e.g., 24 h. Compounds of type 4.8 can be prepared by deprotection of an appropriate acetal, e.g., 4.7 as shown above. The deprotection reaction is carried out in the presence of an appropriate acid, e.g., acetic acid:water (1:1), at an appropriate temperature, e.g., 65° C., for an appropriate period of time, e.g., 1 h. As can be appreciated by one skilled in the art, the above reaction provides an example of a generalized approach wherein compounds similar in structure to the specific reactants above (compounds similar to compounds of type 4.1, 4.2, and 4.3), can be substituted in the reaction to provide substituted 6-aza nucoleoside prodrug derivatives similar to Formula 4.4. 5. Route V In one aspect, substituted 6-aza nucoleoside prodrugs can be prepared as shown below. Compounds are represented in generic form, where PG is a hydroxyl protecting group, X is a halogen, and LG is a leaving group, and with other substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below. In one aspect, compounds of type 5.10, and similar compounds, can be prepared according to reaction Scheme 5B above. Thus, compounds of type 5.7 can be prepared by protection of an appropriate nucleoside, e.g., 5.6 as shown above. Appropriate nucleosides are commercially available or prepared by methods known to one skilled in the art or disclosed elsewhere herein. The protection is carried out in the presence of an appropriate protecting agent, e.g., acetic anhydride, and an appropriate base, e.g., DMAP, in an appropriate solvent, e.g., N,N-diisopropylethylamine. Compounds of type 5.9 can be prepared by activation of an appropriate pyrimidine, e.g., 5.7 as shown above. The activation is carried out in the presence of an appropriate activating agent, e.g., 5.8 as shown above, and an appropriate base, e.g., DMAP and TEA. Compounds of type 5.10 can be prepared by displacement and deprotection of an appropriate activated pyrimidine, e.g., 5.9 as shown above. The displacement/deprotection is carried out in the presence of an appropriate amine base, e.g., ammonium hydroxide as shown above. As can be appreciated by one skilled in the art, the above reaction provides an example of a generalized approach wherein compounds similar in structure to the specific reactants above (compounds similar to compounds of type 5.1, 5.2, 5.3, and 5.4), can be substituted in the reaction to provide substituted 6-aza nucoleoside prodrug derivatives similar to Formula 5.5. 6. Route VI In one aspect, substituted 6-aza nucoleoside prodrugs can be prepared as shown below. Compounds are represented in generic form, where PG is a hydroxyl protecting group, and with other substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below. In one aspect, compounds of type 6.10, and similar compounds, can be prepared according to reaction Scheme 63 above. Thus, compounds of type 6.7 can be prepared by protection of an appropriate nucleoside, e.g., 6.7 as shown above. Appropriate nucleosides are commercially available or prepared by methods known to one skilled in the art. The protection is carried out in the presence of an appropriate protecting agent, e.g., tert-butyldimethylsilyl ether, and an appropriate base, e.g., imidazole, in an appropriate solvent, e.g., DMF. Compounds of type 6.9 can be prepared by acylation of an appropriate amine, e.g., 6.7 as shown above. The acylation is carried out in the presence of an appropriate acyl halide, e.g., 6.8 as shown above, and an appropriate base, e.g., N,N-diisopropylethylamine, in an appropriate base, e.g., DCM. Compounds of type 6.10 can be prepared by deprotection of an appropriate nucleoside, e.g., 6.9 as shown above. The deprotection is carried out in the presence of an appropriate deprotecting agent, e.g., tetra-n-butylammonium fluoride, in an appropriate solvent, e.g., tetrahydrofuran. As can be appreciated by one skilled in the art, the above reaction provides an example of a generalized approach wherein compounds similar in structure to the specific reactants above (compounds similar to compounds of type 6.1, 6.2, 6.3, and 6.4), can be substituted in the reaction to provide substituted 6-aza nucoleoside prodrug derivatives similar to Formula 6.5. 7. Route VII In one aspect, substituted 6-aza nucoleoside prodrugs can be prepared as shown below. Compounds are represented in generic form, where PG is a hydroxyl protecting group, PG′ is an amine protecting group, and with other substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below. In one aspect, compounds of type 7.18, and similar compounds, can be prepared according to reaction Scheme 7B above. Thus, compounds of type 7.12 can be prepared by coupling an appropriate nucleobase, e.g., 7.10 as shown above, and an appropriate sugar, e.g., 7.11 as shown above. Appropriate nucleobases and appropriate sugars are commercially available or prepared by methods known to one skilled in the art. The coupling reaction is carried out in the presence of an appropriate base, e.g., bis(trimethylsilyl)amine (HMDS), and an appropriate inorganic salt, e.g., ammonium sulfate, for an appropriate period of time, e.g., 18 h, followed by addition of the sugar and an appropriate activating agent, e.g., trimethylsilyl trifluoromethanesulfonate, in an appropriate solvent, e.g., acetonitrile, at an appropriate temperature, e.g., 0° C., for an appropriate period of time, e.g., 18 h. Compounds of type 7.13 can be prepared by deprotection of an appropriate nucleobase, e.g., 7.12 as shown above. The deprotection is carried out in the presence of an appropriate deprotecting agent, e.g., 7N NH3, in an appropriate solvent, e.g., methanol. Compounds of type 7.14 can be prepared by protection of an appropriate diol, e.g., 7.13 as shown above, The protection reaction is carried out in the presence of an appropriate catalyst, e.g., copper sulfate, and an appropriate acid, e.g., sulphuric acid, in an appropriate solvent, e.g., acetone. Compounds of type 7.16 can be prepared by coupling an appropriate alcohol, e.g., 7.14 as shown above, and an appropriate carboxylic acid, e.g., 7.15 as shown above. Appropriate carboxylic acids are commercially available or prepared by methods known to one skilled in the art. The coupling reaction is carried out in the presence of an appropriate coupling agent, e.g., DCC, and an appropriate activating agent, e.g., DMAP, in an appropriate solvent, e.g., DCM, at an appropriate temperature, e.g., r. Compounds of type 7.17 can be prepared by deprotection of an appropriate acetal, e.g., 7.16 as shown above. The deprotection is carried out in the presence of an appropriate acid, e.g., acetic acid, at an appropriate temperature, e.g., 65° C., for an appropriate period of time, e.g., 1 h. Compounds of type 7.18 can be prepared by deprotection of an appropriate amine, e.g., 7.17 as shown above. The deprotection reaction is carried out in the presence of an appropriate base, e.g., piperidine. As can be appreciated by one skilled in the art, the above reaction provides an example of a generalized approach wherein compounds similar in structure to the specific reactants above (compounds similar to compounds of type 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, and 7.8), can be substituted in the reaction to provide substituted 6-aza nucoleoside prodrug derivatives similar to Formula 7.9. E. METHODS OF USING THE COMPOUNDS The compounds and pharmaceutical compositions of the invention are useful in treating or controlling disorders associated with a viral infection due to, for example, an Alphavirus (e.g., Chikungunya virus (CHIKV), Ross River virus, Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), and Western equine encephalitis (WEEV)), a Flavivirus (e.g., dengue virus (DENV), West Nile virus (WNV), zika virus (ZIKV), tick-borne encephalitis virus, and yellow fever virus), a Coronavirus (e.g., Middle East Respiratory Syndromes coronavirus (MERS-CoV), Severe Acute Respiratory Syndrome coronavirus (SARS-CoV), and SARS-CoV-2), and a influenza virus (influenza A and influenza B). Examples of viral infections for which the compounds and compositions can be useful in treating, include, but are not limited to, human immunodeficiency virus (HIV), human papillomavirus (HPV), chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, viral pneumonia, yellow fever virus, tick-borne encephalitis virus, Chikungunya virus (CHIKV), Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), Western equine encephalitis (WEEV), dengue (DENV), influenza, West Nile virus (WNV), zika (ZIKV), Middle East Respiratory Syndromes (MERS), Severe Acute Respiratory Syndrome (SARS), and coronavirus disease 2019 (COVID-19). To treat or control the disorder, the compounds and pharmaceutical compositions comprising the compounds are administered to a subject in need thereof, such as a vertebrate, e.g., a mammal, a fish, a bird, a reptile, or an amphibian. The subject can be a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, cat, guinea pig or rodent. The term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered. The subject is preferably a mammal, such as a human. Prior to administering the compounds or compositions, the subject can be diagnosed with a need for treatment of a viral infection, such as human immunodeficiency virus (HIV), human papillomavirus (HPV), chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, viral pneumonia, yellow fever virus, tick-borne encephalitis virus, Chikungunya virus (CHIKV), Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), Western equine encephalitis (WEEV), dengue (DENV), influenza, West Nile virus (WNV), zika (ZIKV), Middle East Respiratory Syndromes (MERS), Severe Acute Respiratory Syndrome (SARS), and coronavirus disease 2019 (COVID-19). The compounds or compositions can be administered to the subject according to any method. Such methods are well known to those skilled in the art and include, but are not limited to, oral administration, transdermal administration, administration by inhalation, nasal administration, topical administration, intravaginal administration, ophthalmic administration, intraaural administration, intracerebral administration, rectal administration, sublingual administration, buccal administration and parenteral administration, including injectable such as intravenous administration, intra-arterial administration, intramuscular administration, and subcutaneous administration. Administration can be continuous or intermittent. A preparation can be administered therapeutically; that is, administered to treat an existing disease or condition. A preparation can also be administered prophylactically; that is, administered for prevention of a viral infection, such as human immunodeficiency virus (HIV), human papillomavirus (HPV), chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, viral pneumonia, yellow fever virus, tick-borne encephalitis virus, Chikungunya virus (CHIKV), Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), Western equine encephalitis (WEEV), dengue (DENV), influenza, West Nile virus (WNV), zika (ZIKV), Middle East Respiratory Syndromes (MERS), Severe Acute Respiratory Syndrome (SARS), and coronavirus disease 2019 (COVID-19). The therapeutically effective amount or dosage of the compound can vary within wide limits. Such a dosage is adjusted to the individual requirements in each particular case including the specific compound(s) being administered, the route of administration, the condition being treated, as well as the patient being treated. In general, in the case of oral or parenteral administration to adult humans weighing approximately 70 Kg or more, a daily dosage of about 10 mg to about 10,000 mg, preferably from about 200 mg to about 1,000 mg, should be appropriate, although the upper limit may be exceeded. The daily dosage can be administered as a single dose or in divided doses, or for parenteral administration, as a continuous infusion. Single dose compositions can contain such amounts or submultiples thereof of the compound or composition to make up the daily dose. The dosage can be adjusted by the individual physician in the event of any contraindications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days. 1. Treatment Methods The compounds disclosed herein are useful for treating or controlling disorders associated with a viral infection due to, for example, an Alphavirus (e.g., Chikungunya virus (CHIKV), Ross River virus, Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), and Western equine encephalitis (WEEV)), a Flavivirus (e.g., dengue virus (DENV), West Nile virus (WNV), zika virus (ZIKV), tick-borne encephalitis virus, and yellow fever virus), a Coronavirus (e.g., Middle East Respiratory Syndromes coronavirus (MERS-CoV), Severe Acute Respiratory Syndrome coronavirus (SARS-CoV), and SARS-CoV-2), and influenza virus (influenza A and influenza B). Thus, provided is a method comprising administering a therapeutically effective amount of a composition comprising a disclosed compound to a subject. In a further aspect, the method can be a method for treating a viral infection. a. Treating a Viral Infection In one aspect, disclosed are methods of treating a viral infection in a subject having the viral infection, the method comprising the step of administering to the subject a therapeutically effective amount of at least one disclosed compound, or a pharmaceutically acceptable salt thereof. In one aspect, disclosed are methods for the treatment of a viral infection in a subject having the viral infection, the method comprising the step of administering to the subject a therapeutically effective amount of at least one compound having a structure represented by a formula: wherein R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, and —P(O)(OR14a)(OR14b); wherein R10, when present, is selected from C1-C20 alkyl and C2-C20 alkenyl; wherein R11, when present, is a an amino acid derivative side chain; wherein R12, when present, is selected from C1-C6 alkyl and C3-C6 cycloalkyl; wherein R13, when present, is selected from C1-C8 alkyl, C3-C8 cycloalkyl, Ar2, and —CH2Ar2; wherein Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; wherein each of R14aand R14b, when present, is independently selected from hydrogen and C1-C8 alkyl; and wherein Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; and wherein R2is a structure represented by a formula selected from: and wherein R15, when present, is selected from hydrogen, —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), and —C(O)(C2-C20 alkenyl), provided that when R1is hydrogen, then R2is and R15is —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), or —C(O)(C2-C20 alkenyl), or a pharmaceutically acceptable salt thereof. Examples of viral infections include, but are not limited to, human immunodeficiency virus (HIV), human papillomavirus (HPV), chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, viral pneumonia, yellow fever virus, tick-borne encephalitis virus, Chikungunya virus (CHIKV), Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), Western equine encephalitis (WEEV), dengue (DENV), influenza, West Nile virus (WNV), zika (ZIKV), Middle East Respiratory Syndromes (MERS), Severe Acute Respiratory Syndrome (SARS), coronavirus disease 2019 (COVID-19). In a further aspect, the subject has been diagnosed with a need for treatment of the disorder prior to the administering step. In a further aspect, the subject is a mammal. In a still further aspect, the mammal is a human. In a further aspect, the method further comprises the step of identifying a subject in need of treatment of the viral infection. In a further aspect, the disorder is associated with a viral infection. In a still further aspect, the viral infection is selected from human immunodeficiency virus (HIV), human papillomavirus (HPV), chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, viral pneumonia, yellow fever virus, tick-borne encephalitis virus, Chikungunya virus (CHIKV), Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), Western equine encephalitis (WEEV), dengue (DENV), influenza, West Nile virus (WNV), zika (ZIKV), Middle East Respiratory Syndromes (MERS), Severe Acute Respiratory Syndrome (SARS), and coronavirus disease 2019 (COVID-19). In a further aspect, the viral infection is due to an Alphavirus. Examples of Alphaviruses include, but are not limited to, Chikungunya virus (CHIKV), Ross River virus, Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), and Western equine encephalitis (WEEV). In a further aspect, the viral infection is due to a Flavivirus. Examples of Flaviviruses include, but are not limited to, dengue virus (DENV), West Nile virus (WNV), zika virus (ZIKV), tick-borne encephalitis virus, and yellow fever virus. In a further aspect, the viral infection is due to a Coronavirus. Examples of Coronaviruses include, but are not limited to, Middle East Respiratory Syndromes coronavirus (MERS-CoV), Severe Acute Respiratory Syndrome coronavirus (SARS-CoV), and SARS-CoV-2. In a further aspect, the viral infection is due to an influenza virus. Examples of influenza viruses include, but are not limited to, influenza A and influenza B viruses. In a further aspect, the effective amount is a therapeutically effective amount. In a still further aspect, the effective amount is a prophylactically effective amount. In a further aspect, the method further comprises the step of administering a therapeutically effective amount of at least one antiviral agent. In a still further aspect, the at least one agent is selected from acemannan, acyclovir, acyclovir sodium, adamantanamine, adefovir, adenine arabinoside, alovudine, alvircept sudotox, amantadine hydrochloride, aranotin, arildone, atevirdine mesylate, avridine, cidofovir, cipamfylline, cytarabine hydrochloride, BMS 806, C31G, carrageenan, cellulose sulfate, cyclodextrins, dapivirine, delavirdine mesylate, desciclovir, dextrin 2-sulfate, didanosine, disoxaril, dolutegravir, edoxudine, enviradene, envirozime, etravirine, famciclovir, famotine hydrochloride, fiacitabine, fialuridine, fosarilate, foscamet sodium, fosfonet sodium, FTC, ganciclovir, ganciclovir sodium, GSK 1265744, 9-2-hydroxy-ethoxy methylguanine, ibalizumab, idoxuridine, interferon, 5-iodo-2′-deoxyuridine, IQP-0528, kethoxal, lamivudine, lobucavir, maraviroc, memotine pirodavir, penciclovir, raltegravir, ribavirin, rimantadine hydrochloride, rilpivirine (TMC-278), saquinavir mesylate, SCH-C, SCH-D, somantadine hydrochloride, sorivudine, statolon, stavudine, T20, tilorone hydrochloride, TMC120, TMC125, trifluridine, trifluorothymidine, tenofovir, tenofovir alefenamide, tenofovir disoproxyl fumarate, prodrugs of tenofovir, UC-781, UK-427, UK-857, valacyclovir, valacyclovir hydrochloride, vidarabine, vidarabine phosphate, vidarabine sodium phosphate, viroxime, zalcitabene, zidovudine, and zinviroxime. In a further aspect, the at least one compound and the at least one agent are administered sequentially. In a still further aspect, the at least one compound and the at least one agent are administered simultaneously. In a further aspect, the at least one compound and the at least one agent are co-formulated. In a still further aspect, the at least one compound and the at least one agent are co-packaged. 2. Methods of Inhibiting a Viral Infection in a Mammal In one aspect, disclosed are methods of inhibiting a viral infection in a mammal, the method comprising the step of administering to the mammal a therapeutically effective amount of at least one disclosed compound, or a pharmaceutically acceptable salt thereof. In one aspect, disclosed are methods of inhibiting a viral infection in a mammal, the method comprising the step of administering to the mammal a therapeutically effective amount of at least one compound having a structure represented by a formula: wherein R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, and —P(O)(OR14a)(OR14b); wherein R10, when present, is selected from C1-C20 alkyl and C2-C20 alkenyl; wherein R1, when present, is a an amino acid derivative side chain; wherein R12, when present, is selected from C1-C6 alkyl and C3-C6 cycloalkyl; wherein R13, when present, is selected from C1-C8 alkyl, C3-C8 cycloalkyl, Ar2, and —CH2Ar2; wherein Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; wherein each of R14aand R14b, when present, is independently selected from hydrogen and C1-C8 alkyl; and wherein Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; and wherein R2is a structure represented by a formula selected from: and wherein R15, when present, is selected from hydrogen, —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), and —C(O)(C2-C20 alkenyl), provided that when R1is hydrogen, then R2is and R15is —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), or —C(O)(C2-C20 alkenyl), or a pharmaceutically acceptable salt thereof. In a further aspect, the compound exhibits inhibition of a viral infection. In a still further aspect, the compound exhibits a decrease in a viral infection. In yet a further aspect, the viral infection is due to an Alphavirus (e.g., Chikungunya virus (CHIKV), Ross River virus, Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), and Western equine encephalitis (WEEV)). In an even further aspect, the viral infection is due to a Flavivirus (e.g., dengue virus (DENV), West Nile virus (WNV), zika virus (ZIKV), tick-borne encephalitis virus, and yellow fever virus). In a still further aspect, the viral infection is due to a Coronavirus (e.g., Middle East Respiratory Syndromes coronavirus (MERS-CoV), Severe Acute Respiratory Syndrome coronavirus (SARS-CoV), and SARS-CoV-2). In yet a further aspect, the viral infection is influenza. In a further aspect, the compound exhibits inhibition of viral activity with an EC50of less than about 30 μM. In a still further aspect, the compound exhibits inhibition of viral activity with an EC50of less than about 25 μM. In yet a further aspect, the compound exhibits inhibition of CHIKV activity with an EC50of less than about 20 μM. In an even further aspect, the compound exhibits inhibition of viral activity with an EC50of less than about 15 μM. In a still further aspect, the compound exhibits inhibition of viral activity with an EC50of less than about 10 μM. In yet a further aspect, the compound exhibits inhibition of viral activity with an EC50of less than about 5 μM. In an even further aspect, the compound exhibits inhibition of viral activity with an EC50of less than about 1 μM. In a still further aspect, the compound exhibits inhibition of viral activity with an EC50of less than about 0.5 μM. In a further aspect, the subject is a mammal. In a still further aspect, the subject is a human. In a further aspect, the subject has been diagnosed with a need for treatment of the disorder prior to the administering step. In a still further aspect, the method further comprises the step of identifying a subject in need of treatment of the disorder. 3. Methods of Inhibiting a Viral Infection in at Least One Cell In one aspect, disclosed are methods for inhibiting a viral infection in at least one cell, the method comprising the step of contacting the at least one cell with an effective amount of at least one disclosed compound, or a pharmaceutically acceptable salt thereof. In one aspect, disclosed are methods for inhibiting a viral infection in at least one cell, the method comprising the step of contacting the at least one cell with an effective amount of at least one compound having a structure represented by a formula: wherein R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, and —P(O)(OR14a)(OR14b); wherein R10, when present, is selected from C1-C20 alkyl and C2-C20 alkenyl; wherein R1, when present, is a an amino acid derivative side chain; wherein R12, when present, is selected from C1-C6 alkyl and C3-C6 cycloalkyl; wherein R13, when present, is selected from C1-C8 alkyl, C3-C8 cycloalkyl, Ar2, and —CH2Ar2; wherein Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; wherein each of R14aand R14b, when present, is independently selected from hydrogen and C1-C8 alkyl; and wherein Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; and wherein R2is a structure represented by a formula selected from: wherein R15, when present, is selected from hydrogen, —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), and —C(O)(C2-C20 alkenyl), provided that when R1is hydrogen, then R2is and R15is —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), or —C(O)(C2-C20 alkenyl), or a pharmaceutically acceptable salt thereof. In a further aspect, the cell is mammalian. In a still further aspect, the cell is human. In yet a further aspect, the cell has been isolated from a mammal prior to the contacting step. In a further aspect, contacting is via administration to a mammal. 4. Use of Compounds In one aspect, the invention relates to the use of a disclosed compound or a product of a disclosed method. In a further aspect, a use relates to the manufacture of a medicament for the treatment of a viral infection in a subject. Also provided are the uses of the disclosed compounds and products. In one aspect, the invention relates to use of at least one disclosed compound; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In a further aspect, the compound used is a product of a disclosed method of making. In a further aspect, the use relates to a process for preparing a pharmaceutical composition comprising a therapeutically effective amount of a disclosed compound or a product of a disclosed method of making, or a pharmaceutically acceptable salt, solvate, or polymorph thereof, for use as a medicament. In a further aspect, the use relates to a process for preparing a pharmaceutical composition comprising a therapeutically effective amount of a disclosed compound or a product of a disclosed method of making, or a pharmaceutically acceptable salt, solvate, or polymorph thereof, wherein a pharmaceutically acceptable carrier is intimately mixed with a therapeutically effective amount of the compound or the product of a disclosed method of making. In various aspects, the use relates to a treatment of a viral infection in a subject. Also disclosed is the use of a compound for antagonism of a viral infection. In one aspect, the use is characterized in that the subject is a human. In one aspect, the use is characterized in that the disorder is a viral infection. In a further aspect, the use relates to the manufacture of a medicament for the treatment of a viral infection in a subject. In a further aspect, the use relates to antagonism of a viral infection in a subject. In a further aspect, the use relates to modulating viral activity in a subject. In a still further aspect, the use relates to modulating viral activity in a cell. In yet a further aspect, the subject is a human. It is understood that the disclosed uses can be employed in connection with the disclosed compounds, products of disclosed methods of making, methods, compositions, and kits. In a further aspect, the invention relates to the use of a disclosed compound or a disclosed product in the manufacture of a medicament for the treatment of a viral infection in a mammal. In a further aspect, the viral infection is selected from human immunodeficiency virus (HIV), human papillomavirus (HPV), chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, viral pneumonia, yellow fever virus, tick-borne encephalitis virus, Chikungunya virus (CHIKV), Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), Western equine encephalitis (WEEV), dengue (DENV), influenza, West Nile virus (WNV), zika (ZIKV), Middle East Respiratory Syndromes (MERS), Severe Acute Respiratory Syndrome (SARS), and coronavirus disease 2019 (COVID-19). 5. Manufacture of a Medicament In one aspect, the invention relates to a method for the manufacture of a medicament for treating a viral infection in a subject having the viral infection, the method comprising combining a therapeutically effective amount of a disclosed compound or product of a disclosed method with a pharmaceutically acceptable carrier or diluent. As regards these applications, the present method includes the administration to an animal, particularly a mammal, and more particularly a human, of a therapeutically effective amount of the compound effective in the inhibition of a viral infection. The dose administered to an animal, particularly a human, in the context of the present invention should be sufficient to affect a therapeutic response in the animal over a reasonable time frame. One skilled in the art will recognize that dosage will depend upon a variety of factors including the condition of the animal and the body weight of the animal. The total amount of the compound of the present disclosure administered in a typical treatment is preferably between about 10 mg/kg and about 1000 mg/kg of body weight for mice, and between about 100 mg/kg and about 500 mg/kg of body weight, and more preferably between 200 mg/kg and about 400 mg/kg of body weight for humans per daily dose. This total amount is typically, but not necessarily, administered as a series of smaller doses over a period of about one time per day to about three times per day for about 24 months, and preferably over a period of twice per day for about 12 months. The size of the dose also will be determined by the route, timing and frequency of administration as well as the existence, nature and extent of any adverse side effects that might accompany the administration of the compound and the desired physiological effect. It will be appreciated by one of skill in the art that various conditions or disease states, in particular chronic conditions or disease states, may require prolonged treatment involving multiple administrations. Thus, in one aspect, the invention relates to the manufacture of a medicament comprising combining a disclosed compound or a product of a disclosed method of making, or a pharmaceutically acceptable salt, solvate, or polymorph thereof, with a pharmaceutically acceptable carrier or diluent. 6. Kits In one aspect, disclosed are kits comprising at least one disclosed compound and one or more of: (a) at least one antiviral agent; (b) a instructions for administering the at least one compound in connection with treating a viral infection; (c) instructions for administering the at least one compound in connection with reducing the risk of viral infection; and (d) instructions for treating a viral infection. In a further aspect, the compound has a structure represented by a formula: wherein R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, and —P(O)(OR14a)(OR14b); wherein R10, when present, is selected from C1-C20 alkyl and C2-C20 alkenyl; wherein R1, when present, is a an amino acid derivative side chain; wherein R12, when present, is selected from C1-C6 alkyl and C3-C6 cycloalkyl; wherein R13, when present, is selected from C1-C8 alkyl, C3-C8 cycloalkyl, Ar2, and —CH2Ar2; wherein Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; wherein each of R14aand R14b, when present, is independently selected from hydrogen and C1-C8 alkyl; and wherein Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; and wherein R2is a structure represented by a formula selected from: and wherein R15, when present, is selected from hydrogen, —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), and —C(O)(C2-C20 alkenyl), provided that when R1is hydrogen, then R2is and R15is —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), or —C(O)(C2-C20 alkenyl), or a pharmaceutically acceptable salt thereof. In a further aspect, the compound has a structure represented by a formula: wherein R1is selected from hydrogen, —C(O)R10, —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, and —P(O)(OR14a)OR14b); wherein R10, when present, is selected from C1-C20 alkyl and C2-C20 alkenyl; wherein R11, when present, is an amino acid derivative side chain; wherein R12, when present, is selected from C1-C6 alkyl and C3-C6 cycloalkyl; wherein R13, when present, is selected from C1-C8 alkyl, C3-C8 cycloalkyl, Ar2, and —CH2Ar2; wherein Ar2, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; wherein each of R14aand R14b, when present, is independently selected from hydrogen and C1-C8 alkyl; and wherein Ar1, when present, is selected from C6-C14 aryl and C2-C10 heteroaryl, and is substituted with 0, 1, 2, or 3 groups independently selected from halogen, —CN, —NH2, —OH, —NO2, C1-C4 alkyl, C2-C4 alkenyl, C1-C4 haloalkyl, C1-C4 cyanoalkyl, C1-C4 hydroxyalkyl, C1-C4 haloalkoxy, C1-C4 alkoxy, C1-C4 alkylamino, (C1-C4)(C1-C4) dialkylamino, and C1-C4 aminoalkyl; and wherein R2is a structure represented by a formula selected from: and wherein R15, when present, is selected from hydrogen, —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), and —C(O)(C2-C20 alkenyl), provided that when R2is then R1is —C(O)CH(R11)NH2, —P(O)(OAr1)NHCH(R12)CO2R13, or —P(O)(OR14a)(OR14b), and provided that when R1is hydrogen, then R15is —C(O)(C1-C20 alkyl), —C(O)(C3-C6 cycloalkyl), or —C(O)(C2-C20 alkenyl), or a pharmaceutically acceptable salt thereof. In a further aspect, the compound has a structure: or a pharmaceutically acceptable salt thereof. In a further aspect, the viral infection is selected from human immunodeficiency virus (HIV), human papillomavirus (HPV), chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, viral pneumonia, yellow fever virus, tick-borne encephalitis virus, Chikungunya virus (CHIKV), Venezuelan equine encephalitis (VEEV), Eastern equine encephalitis (EEEV), Western equine encephalitis (WEEV), dengue (DENV), influenza, West Nile virus (WNV), zika (ZIKV), Middle East Respiratory Syndromes (MERS), Severe Acute Respiratory Syndrome (SARS), and coronavirus disease 2019 (COVID-19). In a still further aspect, the viral infection is selected from CHIKV, DENV, WNV, ZIKA, VEEV, SARS, and COVID-19. In a still further aspect, the antiviral agent is selected from selected from acemannan, acyclovir, acyclovir sodium, adamantanamine, adefovir, adenine arabinoside, alovudine, alvircept sudotox, amantadine hydrochloride, aranotin, arildone, atevirdine mesylate, avridine, cidofovir, cipamfylline, cytarabine hydrochloride, BMS 806, C31G, carrageenan, cellulose sulfate, cyclodextrins, dapivirine, delavirdine mesylate, desciclovir, dextrin 2-sulfate, didanosine, disoxaril, dolutegravir, edoxudine, enviradene, envirozime, etravirine, famciclovir, famotine hydrochloride, fiacitabine, fialuridine, fosarilate, foscamet sodium, fosfonet sodium, FTC, ganciclovir, ganciclovir sodium, GSK 1265744, 9-2-hydroxy-ethoxy methylguanine, ibalizumab, idoxuridine, interferon, 5-iodo-2′-deoxyuridine, IQP-0528, kethoxal, lamivudine, lobucavir, maraviroc, memotine pirodavir, MK-4482 (EIDD-2801), penciclovir, raltegravir, ribavirin, rimantadine hydrochloride, rilpivirine (TMC-278), remdesivir, saquinavir mesylate, SCH-C, SCH-D, somantadine hydrochloride, sorivudine, statolon, stavudine, T20, tilorone hydrochloride, TMC120, TMC125, trifluridine, trifluorothymidine, tenofovir, tenofovir alefenamide, tenofovir disoproxyl fumarate, prodrugs of tenofovir, UC-781, UK-427, UK-857, valacyclovir, valacyclovir hydrochloride, vidarabine, vidarabine phosphate, vidarabine sodium phosphate, viroxime, zalcitabene, zidovudine, and zinviroxime. In a further aspect, the immunity booster is selected from vitamin D, elderberry, Echinacea, a probiotic, vitamin C, vitamin B, green tea, turmeric, zinc, ashwagandha, a prebiotic, and a synbiotic. In a further aspect, the at least one compound and the at least one agent are co-formulated. In a further aspect, the at least one compound and the at least one agent are co-packaged. The kits can also comprise compounds and/or products co-packaged, co-formulated, and/or co-delivered with other components. For example, a drug manufacturer, a drug reseller, a physician, a compounding shop, or a pharmacist can provide a kit comprising a disclosed compound and/or product and another component for delivery to a patient. It is understood that the disclosed kits can be prepared from the disclosed compounds, products, and pharmaceutical compositions. It is also understood that the disclosed kits can be employed in connection with the disclosed methods of using. The foregoing description illustrates and describes the disclosure. Additionally, the disclosure shows and describes only the preferred embodiments but, as mentioned above, it is to be understood that it is capable to use in various other combinations, modifications, and environments and is capable of changes or modifications within the scope of the invention concepts as expressed herein, commensurate with the above teachings and/or the skill or knowledge of the relevant art. The embodiments described herein above are further intended to explain best modes known by applicant and to enable others skilled in the art to utilize the disclosure in such, or other, embodiments and with the various modifications required by the particular applications or uses thereof. Accordingly, the description is not intended to limit the invention to the form disclosed herein. Also, it is intended to the appended claims be construed to include alternative embodiments. All publications and patent applications cited in this specification are herein incorporated by reference, and for any and all purposes, as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. In the event of an inconsistency between the present disclosure and any publications or patent application incorporated herein by reference, the present disclosure controls. F. EXAMPLES The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the compounds, compositions, articles, devices and/or methods claimed herein are made and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in ° C. or is at ambient temperature, and pressure is at or near atmospheric. The Examples are provided herein to illustrate the invention, and should not be construed as limiting the invention in any way. Examples are provided herein to illustrate the invention and should not be construed as limiting the invention in any way. 1. Chemistry Experimentals a. General Experimental The reactions were performed under a dry argon atmosphere and reaction temperatures were measured externally. Anhydrous solvents over molecular sieves were purchased from Aldrich and used as such in reactions. Microwave (MW) reactions were performed in CEM Discover Labmate System with Intelligent Technology for Focused™ Microwave Synthesizer (Explorer 48) or Biotage Initiator+ equipped with Robot Eight microwave system. The reactions were monitored by thin-layer chromatography (TLC) on pre-coated silica gel (60F254) aluminium plates (0.25 mm) from E. Merck and visualized using UV light (254 nm). Purification of compounds was performed on an Isco Teledyne Combiflash Rf200. Universal RediSep solid sample loading pre-packed cartridges (5.0 g silica) were used to absorb crude product and purified on silica RediSep Rf Gold Silica (20-40 μm spherical silica) columns using appropriate solvent gradients. Pure samples were dried overnight under high vacuum before analyses. The high resolution electrospray ionization mass spectral data (HR-ESIMS) were obtained on an Agilent LC-MSTOF.1H,13C,19F, and31P NMR spectra were recorded at 400, 101, 376 and 162 MHz respectively on an Agilent/Varian MR-400 spectrometer. The chemical shifts (δ) are in ppm downfield from standard tetramethylsilane (TMS). HPLC of final compounds were run on an Agilent 1100 LC equipped with a diode array UV detector and were monitored at 254 nm by using one of the following methods Method A: Sunfire C18 column (5 μm, 4.6×150 mm) using H2O—CH3CN (both containing 0.1% formic acid) 5-95% in 20 min with flow rate 1.0 mL/min.; Method B: Phenomenex Kinetex 2.6p Phenyl-hexyl 100 Å 50×4.6 mm column using Solvent A: 95:5 H2O:MeCN with 1% HCO2H, Solvent B: MeCN with 0.1% HCO2H, flow rate 2.0 mL/min; 4 min linear gradient from 5-95% B. b. Synthesis of (5-nitrofuran-2-yl)methyl ((((2R,3S,4R,5R)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-alaninate (1) i. Preparation of (5-nitrofuran-2-yl)methyl (tert-butoxy-carbonyl)-L-alaninate (21) To a 500 mL round bottom flask was added 5-nitrofurfuryl alcohol (10.0 g, 69.9 mmol, 1.0 eq), N-Boc-L-alanine (14.5 g, 76.9 mmol, 1.1 eq), 4-dimethylaminopyridine (0.08 g, 0.69 mmol, 0.01 eq), 130 mL of anhydrous N,N-dimethylformamide, followed by dicyclohexylcarbodiimide (21.6 g, 104.8 mmol, 1.5 eq). The reaction mixture was stirred for 2 days at room temperature. The reaction mixture was diluted with 500 mL of ethyl acetate and then filtered by vacuum filtration to remove byproduct dicyclohexyl urea. The filtrate was washed with water (3×200 mL), saturated sodium bicarbonate (2×200 mL), saturated ammonium chloride (100 mL), followed by brine (100 mL). The combined organic layer was separated, dried over sodium sulfate, filtered, and then the filtrate was evaporated under reduced pressure to afford 34 g of a dark red-brown oil. The crude material was purified in two portions (2×220 g RediSep Rf Gold Silica gel column, 100-90% dichloromethane in methanol, gradient elution) to provide 17.79 g (81%) of 21 as an orange oil.1HNMR (DMSO-d6) δ 7.71 (d, J=3.7 Hz, 1H), 7.40 (d, J=7.2 Hz, 1H), 6.94 (d, J=3.8 Hz, 1H), 5.33-5.09 (m, 2H), 4.06 (dq, J=11.5, 7.2 Hz, 2H), 1.37 (s, 9H), 1.26 (d, J=7.4 Hz, 3H). LCMS m/z 529 (M+H)+. i. Preparation of (5-nitrofuran-2-yl)methyl L-alaninate hydro-chloride (22) To a solution of 21 (5.3 g, 16.86 mmol, 1.0 eq) in 50 mL of anhydrous dichloromethane was added 4N hydrochloric acid (21.1 mL, 84.3 mmol, 5.0 eq) in 1,4-dioxane. The reaction mixture was stirred at 20° C. for 18 h. A precipitate formed, which was filtered by vacuum filtration, rinsed with anhydrous diethyl ether and then dried under reduced pressure to afford 3.89 g of 22 (92%) as an off-white solid.1HNMR (DMSO-d6) δ 8.67 (s, 3H), 7.73 (d, J=3.8 Hz, 1H), 7.15-6.93 (m, 1H), 5.37 (s, 2H), 4.18 (q, J=7.2 Hz, 3H), 1.45 (d, J=7.2 Hz, 3H). ii. Preparation of (5-nitrofuran-2-yl)methyl ((perfluoro-phenoxy)-(phenoxy)-phosphoryl)-L-alaninate (24) To a mixture of 22 (10.0 g, 39.9 mmol, 1.0 eq) in 140 mL of anhydrous dichloromethane was added phenyl phosphorodichloridate (6.54 mL, 43.9 mmol, 1.1 eq). The mixture was cooled to −75° C. and then a solution of triethylamine (11.7 mL, 83.8 mmol, 2.1 eq) in 120 mL of anhydrous dichloromethane was added over 70 min at −70° C. Upon completion of addition, the reaction mixture was stirred at −75° C. for 2 h and then for 18 h as it warmed to 20° C. The reaction mixture was evaporated under reduced pressure to afford a semi-solid, which was triturated in 200 mL of anhydrous t-butyl methylether for 1 h. The mixture was filtered by vacuum filtration to remove triethylamine hydrochloride, which was rinsed with anhydrous t-butyl methylether (2×50 mL). The filtrate was evaporated under vacuum to provide 17.88 g of 23 as an orange oil, which was used further as such. To a cold (−5° C.) solution of 23 (15.51 g, 39.9 mmol, 1.0 eq) in 120 mL of anhydrous dichloromethane was added a solution of pentafluorophenol (8.1 g, 43.9 mmol, 1.1 eq) and triethylamine (6.12 mL, 43.9 mmol, 1.1 eq) in 120 mL of anhydrous dichloromethane over 45 min at −5° C. The reaction mixture was stirred at 0° C. for 2 h and then for 18 h as it warmed to 20° C. The reaction mixture was evaporated under reduced pressure to afford a semi-solid, which was triturated in ethyl acetate (250 mL) and then stirred for 30 min. The mixture was filtered by vacuum filtration to remove triethylamine hydrochloride, which was rinsed with 100 mL of ethyl acetate. The filtrate was washed with water (2×100 mL), 10% aqueous sodium carbonate (2×100 mL), followed by brine (25 mL). The organic layer was separated, dried over sodium sulfate, filtered, and then the filtrate was evaporated under vacuum to give 27.8 g of a crude semi-solid. Purification by flash chromatography (220 g silica column, 100-40% hexane in ethyl acetate, gradient elution) provided 8.25 g (38%) of compound 24 as a white solid (mixture of diastereomers 55:45).1HNMR (DMSO-d6) δ 7.67 (t, J=3.7 Hz, 1H), 7.46-7.36 (m, 2H), 7.29-7.16 (m, 3H), 7.02 (ddd, J=14.0, 9.9, 6.2 Hz, 1H), 6.92 (d, J=3.8 Hz, 1H), 5.36-5.09 (m, 2H), 4.23-3.85 (m, 1H), 1.34 (dd, J=7.2, 1.3 Hz, 3H).19FNMR (DMSO-d6) δF−153.83 (ddt, J=19.1, 15.9, 3.9 Hz, 2F), −160.21 (td, J=23.1, 3.3 Hz, 1F), −163.07 (dtd, J=27.8, 23.7, 4.1 Hz, 2F).31PNMR (DMSO-d6) δp0.13, 0.03. LCMS: m/z 537 (M+H)+. iii. Preparation of Compound 1 To an oven-dried 50 mL round bottom flask was added 6-azauridine (400 mg, 1.63 mmol, 1.0 eq) and co-evaporation twice with anhydrous pyridine (2×3.0 mL) under reduced pressure at 40° C. Further, vacuum dried 6-azauridine was dissolved in anhydrous pyridine (4.89 mL) followed by addition of 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidone (0.933 mL, 7.71 mmol., 5.0 eq). The mixture was stirred for 15 min and then 24 (1.05 g, 1.96 mmol, 1.2 eq) was added. The mixture was further stirred for 5 min and then cooled to 0° C. An 1M solution of dimethylaluminum chloride (0.816 mL, 0.82 mmol, 0.50 eq) in hexanes was slowly added over 5 seconds and them the reaction mixture was stirred for 5 days as it warmed to 20° C. The reaction mixture was evaporated under vacuum to afford crude 1 as a brown oil, which was purified by flash chromatography (40 g silica column, 100-95% dichloromethane in methanol, gradient elution) to provide 118 mg (12%) of pure 1 as a yellow foamy solid (mixture of diastereomers, 53:47).1HNMR (DMSO-d6) δ 12.24 (s, 1H), 7.62 (dd, J=10.5, 3.7 Hz, 1H), 7.48 (dd, J=2.9, 0.6 Hz, 1H), 7.37-7.26 (m, 2H), 7.19-7.05 (m, 3H), 6.88 (ddt, J=6.1, 3.7, 0.6 Hz, 1H), 6.07 (dd, J=13.1, 9.9 Hz, 1H), 5.94-5.84 (m, 1H), 5.40 (dd, J=5.9, 5.1 Hz, 1H), 5.25-5.11 (m, 3H), 4.30-3.64 (m, 7H), 1.21 (ddd, J=13.1, 7.1, 1.0 Hz, 3H).31PNMR (DMSO-d6) δ 3.41, 3.32. LCMS m/z 598 (M+H)+. HR-ESIMS m/z calcd for C22H24N5O13P·H, 598.1181, found 598.1170. HPLC purity by Method B: 97.7% at 254 nm. c. Synthesis of Compound Nos. 2-7 i. Synthesis of Benzyl ((((2R,3S,4R,5R)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-(phenoxy)phosphoryl)-L-alaninate (2) (1) Preparation of Benzyl ((S)-(perfluorophenoxy)(phenoxy)-phosphoryl)-L-alaninate (27a) To a solution of commercial benzyl-L-alaninate hydrochloride 25a (10.0 g, 46.37 mmol, 1.0 eq) in 140 mL of anhydrous dichloromethane was added phenyl phosphorodichloridate (7.60 mL, 51.00 mmol, 1.1 eq). The mixture was cooled to −75° C. and then a solution of triethylamine (13.57 mL, 97.37 mmol, 2.1 eq) in 50 mL of anhydrous dichloromethane was added over 70 min at −70° C. Upon completion of addition, the reaction mixture was stirred at −75° C. for 2 h and then for 18 h as it warmed to 20° C. The reaction mixture was evaporated under reduced pressure to afford a semi-solid, which was triturated in 200 mL of anhydrous t-butyl methylether for 1 h. The mixture was filtered by vacuum filtration to remove triethylamine hydrochloride, which was rinsed with anhydrous t-butyl methylether (2×50 mL). The filtrate was evaporated under vacuum to provide 18.02 g of 26a intermediate as a yellow-green oil. This material was used as such in the next step to prepare intermediate 27a. To a cold (−5° C.) solution of 26a (16.40 g, 46.37 mmoles, 1.0 eq) in 120 mL of anhydrous dichloromethane was added a solution of pentafluorophenol (9.31 g, 51.00 mmoles, 1.1 eq) and triethylamine (7.11 mL, 51.00 mmoles, 1.1 eq) in 50 mL of anhydrous dichloromethane over 45 min at −5° C. The reaction mixture was stirred at 0° C. for 2 hrs and then for 18 hrs as it warmed to 20° C. The reaction mixture was evaporated under reduced pressure to afford a semi-solid, which was triturated in ethyl acetate (250 mL) and then stirred for 30 min. The mixture was filtered by vacuum filtration to remove triethylamine hydrochloride, which was rinsed with 100 mL of ethyl acetate. The filtrate was washed with water (2×100 mL), 10% sodium carbonate (2×100 mL), followed by brine (25 mL). The organic layer was separated, dried over sodium sulfate, filtered, and then the filtrate was evaporated under vacuum to give 28.91 g of a crude semi-solid. The crude material was purified in two portions by flash chromatography (120 g silica column, 100 to 70% hexane in ethyl acetate, gradient elution) to provide a combined mass which was triturated from 95% hexane in ethyl acetate (100 mL) to give 11.24 g (48%) of 27a as a single diastereomer.1HNMR (DMSO-d6) δ 7.44-7.30 (m, 7H), 7.29-7.19 (m, 3H), 6.97 (dd, J=14.1, 9.9 Hz, 1H), 5.12 (s, 2H), 4.17-3.94 (m, 1H), 1.33 (dd, J=7.1, 1.3 Hz, 3H).19FNMR (DMSO-d6) δF−153.30-−154.12 (m, 2F), −160.26 (td, J=23.6, 3.5 Hz, 1F), −163.14 (td, J=23.6, 4.1 Hz, 2F).31PNMR (DMSO-d6) δp0.26. LCMS: m/z 502 (M+H)+. (2) Preparation of Compound 2 The final target 2 was prepared from commercial 6-azauridine (250 mg, 1.02 mmoles, 1.0 eq), 27a (613 mg, 1.22 mmol, 1.2 eq), 1,3-dimethyl-3,4,5,6-tetrahydro2(1H)-pyrimidone (0.62 mL, 5.10 mmol, 5.0 eq), and 1M dimethylaluminum chloride in hexanes (0.51 mL, 0.51 mmol, 0.50 eq) in 3.0 mL of anhydrous pyridine according to the procedure described for the preparation of 1 to afford an oil, which was purified by flash chromatography (40 g silica column, 100-90% dichloromethane in methanol, gradient elution) to provide 50 mg (9%) of compound 2 as a white foamy solid (mixture of diasteromers 81:19).1HNMR (DMSO-d6) δ 12.20 (s, 1H), 7.51 (d, J=0.6 Hz, 1H), 7.41-7.30 (m, 7H), 7.21-7.11 (m, 3H), 6.08-5.99 (m, 1H), 5.96-5.93 (m, 1H), 5.43 (d, J=5.1 Hz, 1H), 5.24 (d, J=6.1 Hz, 1H), 5.15-5.04 (m, 2H), 4.34-3.72 (m, 6H), 1.26 (dd, J=7.1, 1.0 Hz, 3H);31PNMR (DMSO-d6) δP3.49, 3.47. LCMS m/z 563 (M+H)+. HR-ESIMS m/z calcd for C24H27N4O10P·H, 563.1538, found 563.1539. HPLC purity 91.6% at 254 nm. ii. Synthesis of 2-ethylbutyl ((((2R,3S,4R,5R)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-alaninate (3) (1) Preparation of 2-ethylbutyl L-alaninate hydrochloride (25b) To a solution of N-Boc-L-alanine (10.0 g, 52.85 mmol, 1.0 eq.) in 2-ethyl-1-butanol (100 mL, 15.5 eq.) was added trimethylsilyl chloride (33.5 mL, 264 mmol, 5.0 eq). The reaction mixture was stirred at 20° C. for 18 h. The reaction mixture was evaporated under reduced pressure at 60° C. to afford an oil, which was triturated in 100 mL of anhydrous diethyl ether. The mixture was filtered by vacuum filtration to collect a solid which was rinsed with anhydrous diethyl ether (2×40 mL) and then dried under reduced pressure at 50° C. to provide 9.40 g (85%) of 25b as a white solid.1HNMR (DMSO-d6) δ 8.59 (s, 3H), 4.18-4.01 (m, 3H), 1.53 (hept, J=6.1 Hz, 1H), 1.44 (d, J=7.2 Hz, 3H), 1.41-1.29 (m, 4H), 0.88 (t, J=7.4 Hz, 6H). (2) Preparation of 2-ethylbutyl ((S)-(perfluorophenoxy)-(phenoxy)phosphoryl)-L-alaninate (27b) Intermediate 26b was prepared from 25b (10.0 g, 46.37 mmol, 1.0 eq) and phenyl phosphorodichloridate (7.82 mL, 52.45 mmol, 1.1 eq) in 140 mL of anhydrous dichloromethane with triethylamine (13.96 mL, 100 mmol, 2.1 eq) as a base according to the procedure described for the preparation of 26a to afford 18.02 g as a colorless oil. Further, compound 27b was prepared from intermediate 26b (16.6 g, 47.68 mmol, 1.0 eq) and pentafuorophenol (8.44 g, 45.86 mmol, 0.96 eq) in 120 mL of anhydrous dichloromethane with triethylamine (6.39 mL, 45.86 mmol, 0.96 eq) as base according to the procedure described for the preparation of 27a to afford 4.9 g (21%) as white needles and as a single diastereomer.1HNMR (DMSO-d6) δ 7.43 (dd, J=8.5, 7.4 Hz, 2H), 7.32-7.17 (m, 3H), 6.91 (dd, J=14.1, 9.9 Hz, 1H), 3.99 (dd, J=14.2, 6.4 Hz, 3H), 1.45 (h, J=6.1 Hz, 1H), 1.37-1.23 (m, 7H), 0.83 (t, J=7.4 Hz, 6H).19FNMR (DMSO-d6) δF−153.68 to −153.78 (m, 2F), −160.39 (td, J=23.6, 3.3 Hz, 1F), −163.21 (td, J=23.6, 4.1 Hz, 2F).31PNMR (DMSO-d6) δp0.27. LCMS: m/z 496 (M+H)+. (3) Preparation of Compound 3 The final target 3 was prepared from commercial 6-azauridine (100 mg, 0.408 mmol, 1.0 eq), 27b (242 mg, 0.489 mmol, 1.2 eq), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidone (0.247 mL, 2.04 mmol, 5.0 eq), and 1M solution of dimethylaluminum chloride in hexanes (0.204 mL, 0.204 mmol, 0.50 eq) in 3.0 mL of anhydrous pyridine according to the procedure described for the preparation of 1 to afford an oil, which was purified by flash chromatography (40 g silica column, 100-90% dichloromethane in methanol, gradient elution) to provide 26 mg (11%) of compound 3 as a colorless solid (mixture of diastereomers 94:6).1HNMR (DMSO-d6) δ 12.27 (d, J=9.5 Hz, 1H), 7.51 (s, 1H), 7.43-7.31 (m, 2H), 7.27-7.11 (m, 3H), 6.02-5.88 (m, 2H), 5.43 (d, J=5.0 Hz, 1H), 5.24 (d, J=6.1 Hz, 1H), 4.25-4.13 (m, 2H), 4.10-3.89 (m, 5H), 3.88-3.78 (m, 1H), 1.47 (hept, J=6.2 Hz, 1H), 1.37-1.22 (m, 7H), 0.85 (t, J=7.5 Hz, 6H).31PNMR (DMSO-d6) δP3.51, 3.47. LCMS m/z 557 (M+H)+. HR-ESIMS m/z calcd for C23H33N4O10P·H, 557.20071, found 557.20075. HPLC purity by Method B: 97.4% at 254 nm. iii. Synthesis of 2-isobutyl ((((2R,3S,4R,5R)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-alaninate (4) (1) Preparation of Isobutyl L-alaninate hydrochloride (25c) Intermediate 25c was prepared from N-Boc-L-alanine (3.0 g, 15.86 mmoles, 1.0 eq.) and trimethylsilyl chloride (10.0 mL, 79.28 mmoles, 5.0 eq) in 2-methyl-1-propanol (100 mL, 69 eq) according to the procedure described for the preparation of 25b to afford 2.66 g (92%) of a white solid.1HNMR (DMSO-d6) δ 8.61 (s, 3H), 4.09 (q, J=7.2 Hz, 1H), 4.04-3.89 (m, 2H), 1.94 (dh, J=13.4, 6.6 Hz, 1H), 1.45 (d, J=7.2 Hz, 3H), 0.93 (dd, J=6.7, 0.7 Hz, 6H). (2) Preparation of Isobutylbutyl ((S)-(perfluorophenoxy)-(phenoxy)phosphoryl)-L-alaninate (27c) Intermediate 26c was prepared from 25c (2.0 g, 11.01 mmol, 1.0 eq) and phenyl phosphorodichloridate (1.81 mL, 12.11 mmol, 1.1 eq) in 20 mL of anhydrous dichloromethane with triethylamine (3.22 mL, 23.12 mmol, 2.1 eq) as base according to the procedure described for the preparation of 26a to afford 3.83 g of 26c as a colorless oil. Further, 27c, was prepared from 26b (3.83 g, 11.98 mmol, 1.0 eq) and pentafuorophenol (2.43 g, 13.18 mmol, 1.1 eq) in 20 mL of anhydrous dichloromethane with triethylamine (1.84 mL, 13.18 mmol, 1.1 eq) as base according to the procedure described for the preparation of 27a to afford 1.43 g (26%) of pure 27c as a single diastereomer.1HNMR (DMSO-d6) δ 7.48-7.38 (m, 2H), 7.30-7.19 (m, 3H), 6.90 (dd, J=14.1, 9.9 Hz, 1H), 4.11-3.94 (m, 1H), 3.84 (dd, J=6.6, 0.6 Hz, 2H), 1.93-1.79 (m, J=6.7 Hz, 1H), 1.32 (dd, J=7.1, 1.2 Hz, 3H), 0.88 (d, J=6.7 Hz, 6H).31PNMR (DMSO-d6) δp0.28. LCMS: m/z 468 (M+H)+. (3) Preparation of Compound 4 The final target 4 was prepared from commercial 6-azauridine (200 mg, 0.816 mmol, 1.0 eq), 27c (457 mg, 0.979 mmol, 1.2 eq), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidone (0.49 mL, 4.08 mmol, 5.0 eq), and 1M dimethylaluminum chloride solution in hexanes (0.49 mL, 0.494 mmol, 0.60 eq) and 1.50 mL of anhydrous pyridine according to the procedure described for the preparation of 1 to afford a residue, which was purified by flash chromatography (40 g silica column, 100-90% dichloromethane in methanol, gradient elution) to provide 47 mg (11%) as a white solid and as a single diastereomer.1HNMR (DMSO-d6) δ 12.24 (d, J=5.0 Hz, 1H), 7.52 (d, J=0.5 Hz, 1H), 7.40-7.33 (m, 2H), 7.24-7.14 (m, 3H), 6.01-5.91 (m, 2H), 5.42 (d, J=5.0 Hz, 1H), 5.23 (d, J=6.2 Hz, 1H), 4.27-4.12 (m, 2H), 4.10-3.90 (m, 3H), 3.89-3.75 (m, 3H), 1.86 (hept, J=6.7 Hz, 1H), 1.25 (dd, J=7.1, 1.0 Hz, 3H), 0.88 (d, J=6.7 Hz, 6H).31PNMR (DMSO-d6) δP: 3.49; LCMS m/z 551 (M+H)+. HRMS m/z calcd. for C21H29N4O10P·H, 529.1694, found, 529.1690. HPLC purity by Method B: 91% at 254 nm. iv. Synthesis of isopropyl ((((2R,3S,4R,5R)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-alaninate (5) (1) Preparation of Isopropyl ((S)-(perfluorophenoxy)-(phenoxy)phosphoryl)-L-alaninate (27d) Intermediate 26d was prepared from commercial isopropyl-L-alaninate hydrochloride 25d (2.0 g, 11.93 mmol, 1.0 eq.) and phenyl phosphorodichloridate (1.96 mL, 13.12 mmol, 1.1 eq.) in 20 mL of anhydrous dichloromethane with triethylamine (3.49 mL, 25.05 mmol, 2.1 eq.) as base according to the procedure described for the preparation of 26a to afford 3.65 g of a colorless oil. Further, compound 27d was prepared from intermediate 26d (3.65 g, 11.93 mmol, 1.0 eq) and pentafuorophenol (2.41 g, 13.12 mmol, 1.1 eq) in 20 mL of anhydrous dichloromethane with triethylamine (1.83 mL, 13.12 mmol, 1.1 eq) as base according to the procedure described for the preparation of 27a to afford 2.22 g (41%) (27d) of a white solid and as a single diastereomer.1HNMR (DMSO-d6) δ 7.48-7.39 (m, 2H), 7.32-7.20 (m, 3H), 6.99-6.74 (m, 1H), 4.89 (pd, J=6.3, 5.5 Hz, 1H), 4.02-3.82 (M, 1 h), 1.29 (ddd, J=7.1, 4.6, 1.2 Hz, 3H), 1.17 (dd, J=6.3, 1.1 Hz, 6H).19FNMR (DMSO-d6) δF−153.76 (t, J=21.2 Hz, 2F), −159.94 to −160.90 (m, 1F), −162.68 to −163.68 (m, 2F).31PNMR (DMSO-d6) δp0.31. LCMS: m/z 454 (M+H)+. (2) Preparation of Compound 5 The final target 5 was prepared from commercial 6-azauridine (250 mg, 1.02 mmoles, 1.0 eq.), 27d (555 mg, 1.22 mmol, 1.2 eq.), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidone (0.616 mL, 5.10 mmol, 5.0 eq.) and 1M (in hexanes) dimethylaluminum chloride (0.51 mL, 0.510 mmol, 0.50 eq.) in 3.0 mL of anhydrous pyridine according to the procedure described for the preparation of 1 to afford a residue, which was purified by flash chromatography (40 g silica column, 100-90% dichloromethane in methanol, gradient elution) to provide 61 mg (12%) (5) as a white solid and as a mixture of diasteromers (63:37).1HNMR (DMSO-d6) δ 12.04 (s, 1H), 7.51 (dd, J=2.3, 0.5 Hz, 1H), 7.41-7.32 (m, 2H), 7.24-7.12 (m, 3H), 5.99-5.86 (m, 2H), 5.42 (td, J=4.8, 2.9 Hz, 1H), 5.24 (dd, J=6.0, 1.4 Hz, 1H), 4.85 (pd, J=6.3, 3.7 Hz, 1H), 4.25-4.13 (m, 2H), 4.12-3.89 (m, 3H), 3.82-3.67 (m, 1H), 1.28-1.10 (m, 9H);31PNMR (DMSO-d6) δP3.51, 3.48. LCMS m/z 515 (M+H)+. HR-ESIMS m/z calcd. for C20H27N4O10P·H, 515.1538, found 515.1536. HPLC purity by Method B: 95% at 254 nm. v. Synthesis of ethyl ((((2R,3S,4R,5R)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-(phenoxy)phosphoryl)-L-alaninate (6) (1) Preparation of ethyl ((S)-(perfluorophenoxy)-(phenoxy)phosphoryl)-L-alaninate (27e) Intermediate 26e was prepared from commercial ethyl-L-alaninate hydrochloride 25e (6.50 g, 42.34 mmol, 1.0 eq) and phenyl phosphorodichloridate (6.94 mL, 46.58 mmol, 1.1 eq) in 70 mL of anhydrous dichloromethane with triethylamine (12.13 mL, 88.92 mmol, 2.1 eq) as base according to the procedure described for the preparation of 26a to afford 14.01 g (26e) of a colorless oil. Further, compound 27e was prepared from 26e (12.35 g, 42.34 mmol, 1.0 eq) and pentafluorophenol (8.57 g, 46.58 mmol, 1.1 eq) in 100 mL of anhydrous dichloromethane with triethylamine (6.49 mL, 46.58 mmol, 1.1 eq) as base according to the procedure described for the preparation of 27a to afford 8.28 g (45%) of a white solid and as a single diastereomer.1HNMR (DMSO-d6) δ 7.48-7.38 (m, 2H), 7.26 (dddt, J=9.8, 7.7, 2.3, 1.1 Hz, 3H), 6.89 (ddd, J=13.9, 9.9, 6.4 Hz, 1H), 4.12-4.04 (m, 2H), 4.04-3.92 (m, 1H), 1.30 (ddd, J=7.1, 5.0, 1.2 Hz, 3H), 1.17 (t, J=7.1 Hz, 3H).19FNMR (DMSO-d6) δFδ −153.69-−154.18 (m, 2F), −160.26-−160.44 (m, 1F), −163.0426-−163.27 (m, 2F);31P NMR (DMSO-d6) δ 0.33. LCMS m/z 440 (M+H)+. (2) Preparation of Compound 6 The final target 6 was prepared from commercial 6-azauridine (250 mg, 1.02 mmol, 1.0 eq.), 27e (537 mg, 1.22 mmol, 1.2 eq.), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidone (0.62 mL, 5.10 mmol, 5.0 eq.), and 1M (in hexanes) dimethylaluminum chloride (0.51 mL, 0.51 mmol, 0.50 eq.) in 3.0 mL of anhydrous pyridine according to the procedure described for the preparation of 1 to afford an oil, which was purified by flash chromatography (40 g silica column, 100-90% dichloromethane in methanol, gradient elution) to provide 53 mg (10%) of 6 as a white solid and as a mixture of diasteromers (68:32).1HNMR (DMSO-d6) δ 12.28 (s, 1H), 7.52 (dd, J=1.9, 0.5 Hz, 1H), 7.41-7.33 (m, 2H), 7.24-7.13 (m, 3H), 6.10-5.77 (m, 2H), 5.42 (dd, J=5.0, 3.8 Hz, 1H), 5.24 (dd, J=6.2, 1.6 Hz, 1H), 4.26-3.90 (m, 7H), 3.79 (dddt, J=17.0, 13.5, 9.9, 7.1 Hz, 1H), 1.29-1.11 (m, 6H).31PNMR (DMSO-d6) δ 3.48, 3.46. LCMS m/z 501 (M+H)+. HR-ESIMS m/z calcd for C19H25N4O10P·H, 501.1381, found, 501.1378. HPLC purity 96.3% at 254 nm. vi. Synthesis of methyl ((((2R,3S,4R,5R)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-(phenoxy)phosphoryl)-L-alaninate (7) (1) Preparation of methyl ((S)-(perfluorophenoxy)-(phenoxy)phosphoryl)-L-alaninate (27F) Intermediate 26f was prepared from commercial methyl-L-alaninate hydrochloride 25f (10.0 g, 71.64 mmol, 1.0 eq.) and phenyl phosphorodichloridate (11.75 mL, 78.08 mmol, 1.1 eq.) in 140 mL of anhydrous dichloromethane with triethylamine (20.97 mL, 150.45 mmol, 2.1 eq.) as base according to the procedure described for the preparation of 26a to afford 21.34 g of a yellow oil. Further, compound 27f was prepared from 26f (19.89 g, 71.64 mmol, 1.0 eq.) and pentafluorophenol (14.51 g, 78.80 mmol, 1.1 eq.) in 120 mL of anhydrous dichloromethane with triethylamine (10.98 mL, 78.80 mmol, 1.1 eq.) as base according to the procedure described for the preparation of 27a to afford 8.39 g (28%) of a white solid and as a single diastereomer.1HNMR (DMSO-d6) δ 7.48-7.39 (m, 2H), 7.30-7.21 (m, 3H), 6.91 (dd, J=14.1, 9.9 Hz, 1H), 4.01 (ddq, J=10.9, 9.9, 7.1 Hz, 1H), 3.61 (s, 3H), 1.29 (dd, J=7.1, 1.2 Hz, 3H).19F NMR (DMSO-d6) δFδ −153.39-−154.18 (m, 2F), −160.05-−160.77 (m, 1F), −163.19 (td, J=23.2, 3.6 Hz, 2F).31PNMR (DMSO-d6) δP0.35. LCMS m/z 426 (M+H)+. (2) Preparation of Compound 7 The final target 7 was prepared from commercial 6-azauridine (250 mg, 1.02 mmol, 1.0 eq.), 27f (520 mg, 1.22 mmol, 1.2 eq.), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidone (0.616 mL, 5.10 mmol, 5.0 eq.), and 1M (in hexanes) dimethylaluminum chloride (0.51 mL, 0.51 mmol, 0.50 eq.) in 3.0 mL of anhydrous pyridine according to the procedure described for the preparation of 1 to afford a residue, which was purified by flash chromatography (40 g silica column, 100-90% dichloromethane in methanol, gradient elution) to provide 51 mg (10%) (7) as a colorless solid and as a mixture of diasteromers (55:45).1HNMR (DMSO-d6) δ 12.22 (s, 1H), 7.53 (dd, J=2.9, 0.5 Hz, 1H), 7.41-7.33 (m, 2H), 7.23-7.13 (m, 3H), 6.02-5.91 (m, 2H), 5.42 (dd, J=5.1, 3.4 Hz, 1H), 5.24 (dd, J=6.2, 3.1 Hz, 1H), 4.27-3.90 (m, 5H), 3.89-3.73 (m, 1H), 3.59 (d, J=8.2 Hz, 3H), 1.27-1.18 (m, 3H).31PNMR (DMSO-d6) δP3.44. LCMS m/z 487 (M+H)+. HR-ESIMS m/z calcd for C18H23N4O10P·H, 487.1225, found 487.1219. HPLC purity by Method B: 91.7% at 254 nm. d. Synthesis of isobutyl ((((2R,3S,4R,5R)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)-methoxy)(naphthalen-1-yloxy)phosphoryl)-L-alaninate (8) i. Preparation of naphthalen-1-yl phosphorodichloridate (28) To a cold (−75° C.) solution of 1-naphthol (2.00 g, 13.9 mmol, 1.0 eq.) in 35 mL of anhydrous diethyl ether was added phosphorusoxychloride (1.42 mL, 15.3 mmol, 1.1 eq.) followed by a solution of triethylamine (2.13 mL, 15.3 mmol, 1.1 eq.) in 10 mL anhydrous diethyl ether dropwise over the period of 20 min at −70° C. Upon completion of addition, the reaction mixture was stirred at −70° C. under argon for 2 h and then for 18 h as it warmed to 20° C. The reaction mixture was filtered by vacuum filtration to remove triethylamine hydrochloride, which was rinsed with diethyl ether (2×20 mL). The filtrate was concentrated under vacuum to afford 3.74 g of 28 as a light tan oil.1HNMR (CDCl3) δ 8.13-8.06 (m, 1H), 7.94-7.87 (m, 1H), 7.84-7.77 (m, 1H), 7.66-7.51 (m, 3H), 7.47 (ddd, J=8.3, 7.6, 0.6 Hz, 1H), 4.34 (s, 1H).31PNMR (CDCl3) δ 3.81. ii. Preparation of isobutyl ((naphthalen-1-yloxy)(perfluoro-phenoxy)phosphoryl)-L-alaninate (30) Intermediate 29 was prepared from 25c (2.37 g, 13.05 mmol, 1.0 eq) and 28 (3.75, 14.35 mmol, 1.1 eq.) in 30 mL of anhydrous dichloromethane with triethylamine (3.82 mL, 27.4 mmol, 2.1 eq.) as base according to the procedure described for the preparation of 26a to afford 5.64 g of a light tan oil. Further, compound 30 was prepared from 29 (5.64 g, 15.30 mmol, 1.0 eq.) and pentafluorophenol (3.09 g, 16.80 mmol, 1.1 eq.) in 30 mL of anhydrous dichloromethane with triethylamine (2.34 mL, 16.80 mmol, 1.1 eq.) as base according to the procedure described for the preparation of 27a to afford a tan oil. Purification by flash chromatography (120 g silica column, 100-70% hexane in ethyl acetate, gradient elution), followed by trituration in 5-10% ethyl acetate in hexane provided 1.60 g (20%) of compound 30 as a white solid (mixture of diastereomers 55:45).1HNMR (DMSO-d6) δ 8.22-8.07 (m, 1H), 8.05-7.96 (m, 1H), 7.84 (ddd, J=8.1, 2.6, 1.3 Hz, 1H), 7.71-7.46 (m, 4H), 7.11 (ddd, J=15.3, 13.8, 9.9 Hz, 1H), 4.22-4.01 (m, 1H), 3.94-3.68 (m, 2H), 1.82 (dhept, J=11.7, 6.7 Hz, 1H), 1.36 (ddd, J=11.1, 7.1, 1.2 Hz, 3H), 0.94-0.75 (m, 6H).19FNMR (DMSO-d6) δ −153.32-−154.10 (m, 2F), −159.86-−160.61 (m, 1F), −163.07 (dtd, J=33.5, 23.2, 3.9 Hz, 2F).31PNMR (DMSO-d6) δP0.93, 0.48. LCMS m/z 518 (M+H)+. iii. Preparation of Compound 8 The final target 8 was prepared from commercial 6-azauridine (250 mg, 1.02 mmoles, 1.0 eq), 30 (633 mg, 1.22 mmol, 1.2 eq.), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidone (0.62 mL, 5.10 mmol, 5.0 eq.), and 1M (in hexanes) dimethylaluminum chloride (0.51 mL, 0.51 mmol, 0.50 eq.) in 3.0 mL of anhydrous pyridine according to the procedure described for the preparation of 2 to afford a residue, which was purified by flash chromatography (40 g silica column, 100-90% dichloromethane in methanol, gradient elution) to provide 8 (71 mg, 12%) as a white solid and as a mixture of diastereomers (55:45).1HNMR (DMSO-d6) δ 12.22 (s, 1H), 8.16-8.04 (m, 1H), 8.01-7.92 (m, 1H), 7.84-7.69 (m, 1H), 7.64-7.53 (m, 2H), 7.52-7.35 (m, 3H), 6.17 (ddd, J=12.2, 10.1, 4.7 Hz, 1H), 5.96 (d, J=3.1 Hz, 1H), 5.43 (ddd, J=6.5, 5.1, 1.1 Hz, 1H), 5.26 (d, J=6.1 Hz, 1H), 4.24 (dddd, J=15.4, 8.2, 5.5, 2.9 Hz, 2H), 4.16-3.98 (m, 3H), 3.97-3.64 (m, 3H), 1.81 (dhept, J=20.0, 6.7 Hz, 1H), 1.25 (td, J=7.0, 1.0 Hz, 3H), 0.84 (dd, J=16.6, 6.7 Hz, 6H).31PNMR (DMSO-d6) δP3.93, 3.72; LCMS m/z 579 (M+H)+. HR-ESIMS m/z calcd for C25H31N4O10P·H, 579.1851, found 579.1850. HPLC purity by Method B: 86% at 254 nm. e. Synthesis of Compound Nos. 9-13 i. Preparation of 2-((3ar,4r,6r,6ar)-6-(hydroxymethyl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)-1,2,4-triazine-3,5(2H,4H)-dione (31) 6-Azauridine (2.7 g, 11.01 mmol, 1.0 eq.) was added to the flask containing copper sulfate (5 g, 31.33 mmol, 3.0 eq.) (after drying under vacuum at 130° C.) and suspended in 70 mL of anhydrous acetone under argon. To this mixture, 0.13 mL of concentrated sulfuric acid was added and stirred for 3 days at room temperature. The mixture was filtered and the filtrate was neutralized slowly to pH 7 by addition of 1.8 mL cold 7N ammonia in methanol. Solvent was removed by evaporation and purified on RediSep Rf Gold Silica column (0-30% methanol in dichloromethane) to afford 31 (2.14 g, 68% yield) as a white solid.1HNMR (DMSO-d6) δ 12.25 (s, 1H), 7.54 (s, 1H), 6.04 (d, J=1.5 Hz, 1H), 4.98 (dd, J=6.2, 1.5 Hz, 1H), 4.82 (t, J=5.9 Hz, 1H), 4.68 (dd, J=6.2, 2.8 Hz, 1H), 4.01 (td, J=6.8, 2.8 Hz, 1H), 3.39 (t, J=6.2 Hz, 2H), 1.45 (s, 3H), 1.27 (s, 3H). ii. Synthesis of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl) methyl-D-valinate (9) (1) Preparation of ((3ar,4r,6r,6ar)-6-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl (((9H-fluoren-9-yl)methoxy)carbonyl)-D-valinate (33a) To a solution of intermediate 31 (64.0 mg, 0.22 mmol, 1.0 eq.) in anhydrous dichloromethane (5 mL) and DMPU (0.1 mL) was added N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (64.5 mg, 0.34 mmol, 1.5 eq.) and 32a (114 mg, 0.34 mmol, 1.5 eq.). The reaction mixture was stirred for 5 min at room temperature followed by addition of 4-(dimethylamino)pyridine (33 mg, 0.27 mmol, 1.2 eq.) and further the mixture was stirred for 18 h at room temperature. The mixture was concentrated under vacuum and purified by column chromatography (0-40% ethyl acetate in dichlorometane) to afford 33a (88 mg, 64.7% yield) as a white foamy solid.1HNMR (DMSO-d6) δ 12.27 (s, 1H), 7.87 (d, J=7.5 Hz, 2H), 7.78-7.66 (m, 3H), 7.52 (s, 1H), 7.40 (td, J=7.5, 1.1 Hz, 2H), 7.30 (dt, J=7.6, 1.6 Hz, 2H), 6.07 (d, J=1.3 Hz, 1H), 5.03 (dd, J=6.1, 1.4 Hz, 1H), 4.75 (dd, J=6.1, 2.3 Hz, 1H), 4.34-4.08 (m, 5H), 3.91 (dd, J=8.2, 6.4 Hz, 1H), 2.03 (dt, J=13.5, 6.5 Hz, 1H), 1.43 (s, 3H), 1.25 (s, 3H), 0.87 (t, J=6.8 Hz, 6H). (2) Preparation of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)-methyl(((9H-fluoren-9-yl)methoxy)carbonyl)-D-valinate (34a) Formic acid (1 mL, 50% v/v in water) was added to 33a (80 mg, 0.13 mmol) and the mixture was warmed to 65° C. and stirred for 90 min. TLC showed complete consumption of starting material and a new spot with lower Rf was observed. The formic acid was removed by evaporation and the residue was purified by column chromatography (0-30% methanol in dichloromethane) to afford 34a (66 mg, 88.3% yield) as a white solid.1HNMR (DMSO-d6) δ 12.23 (s, 1H), 7.87 (d, J=7.5 Hz, 2H), 7.80-7.64 (m, 3H), 7.53 (s, 1H), 7.40 (t, J=7.4 Hz, 2H), 7.31 (td, J=7.2, 6.8, 4.1 Hz, 2H), 5.90 (d, J=2.8 Hz, 1H), 5.40 (d, J=4.9 Hz, 1H), 5.23 (d, J=6.2 Hz, 1H), 4.32 (t, J=7.5 Hz, 1H), 4.26 (d, J=6.2 Hz, 2H), 4.23-4.16 (m, 2H), 4.06 (q, J=5.9 Hz, 1H), 4.01-3.90 (m, 3H), 2.13-1.88 (m, 1H), 0.85 (dd, J=6.9, 1.9 Hz, 6H). (3) Preparation of Compound 9 A solution of piperidine (0.21 mL, 0.11 mmol, 5% by volume in DMF) was added to a vial containing 34a (60 mg, 0.11 mmol) and stirred for 15 min at room temperature. TLC showed completion of starting material and presence of a new spot. The mixture was then concentrated to remove DMF and residue was purified by column chromatography (0-20% methanol in dichloromethane) to afford 9 (28 mg, 76.8% yield) as a foamy white solid.1HNMR (DMSO-d6) δ 7.52 (s, 1H), 5.89 (d, J=2.9 Hz, 1H), 5.40 (s, 1H), 5.21 (s, 2H), 4.28 (dd, J=9.5, 5.1 Hz, 1H), 4.19 (dd, J=5.1, 2.9 Hz, 1H), 4.05 (t, J=5.4 Hz, 1H), 3.97 (dd, J=9.5, 5.7 Hz, 2H), 3.14 (d, J=5.2 Hz, 1H), 1.81 (dq, J=13.2, 6.8 Hz, 1H), 0.80 (dd, J=23.8, 6.8 Hz, 6H).13C NMR (DMSO-d6) δ 175.2, 157.5, 149.0, 136.8, 136.72, 90.3, 72.9, 70.8, 64.6, 59.6, 32.1, 19.5, 17.7. HR-ESIMS m/z calcd for C13H21N4O7[M+H]+345.1405, found 345.1406. HPLC purity by Method A: 96% at 254 nm. iii. Synthesis of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl L-isoleucinate (10) (1) Preparation of ((3ar,4r,6r,6ar)-6-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl-(((9H-fluoren-9-yl)methoxy)carbonyl)-L-isoleucinate (33b) Intermediate 33b was synthesized from 31 (60 mg, 0.21 mmol, 1.0 eq.), 32b (112 mg, 0.32 mmol, 1.4 eq.) and N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (61 mg, 0.32 mmol, 1.4 eq.) and 4-(dimethylamino)pyridine (31 mg, 0.25 mmol, 1.2 eq.) using similar procedure as for 33a and afforded 33b as a white solid (99 mg, 75.7% yield).1HNMR (DMSO-d6) δ 12.28 (s, 1H), 7.87 (d, J=7.5 Hz, 2H), 7.81-7.64 (m, 3H), 7.54 (s, 1H), 7.43-7.22 (m, 4H), 6.08 (s, 1H), 5.03 (d, J=6.0 Hz, 1H), 4.78-4.68 (m, 1H), 4.23 (q, J=8.4 Hz, 5H), 4.10 (dd, J=9.5, 3.3 Hz, 1H), 4.03-3.87 (m, 1H), 1.75 (d, J=14.0 Hz, 1H), 1.44 (s, 3H), 1.38-1.29 (m, 1H), 1.25 (s, 3H), 1.16 (td, J=7.1, 1.0 Hz, 1H), 0.79 (dd, J=9.1, 6.9 Hz, 6H). (2) Preparation of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl (((9H-fluoren-9-yl)methoxy)carbonyl)-L-isoleucinate (34b) Intermediate 34b was synthesized from 33b (90 mg, 0.15 mmol) and formic acid (2 mL, 50% aqueous solution) using similar procedure as for 34a and afforded 34b as a white solid (74 mg, 88% yield).1HNMR (DMSO-d6) δ 12.23 (s, 1H), 7.87 (d, J=7.5 Hz, 2H), 7.72 (t, J=7.3 Hz, 3H), 7.51 (s, 1H), 7.45-7.36 (m, 2H), 7.33-7.26 (m, 2H), 5.90 (d, J=3.2 Hz, 1H), 5.39 (d, J=5.1 Hz, 1H), 5.22 (d, J=6.0 Hz, 1H), 4.36-4.14 (m, 4H), 4.12-3.92 (m, 4H), 3.37 (s, 2H), 1.79 (d, J=7.8 Hz, 1H), 1.40-1.28 (m, 1H), 1.19 (dt, J=14.1, 7.4 Hz, 1H), 0.87-0.73 (m, 6H). (3) Preparation of Compound 10 Compound 10 was synthesized from 34b (73 mg, 0.126 mmol) and piperidine (1 mL of 5% solution v/v in DMF) using similar procedure for 9 and afforded 10 as a white solid in 37 mg, 82% yield.1HNMR (DMSO-d6) δ 7.49 (s, 1H), 5.89 (d, J=3.2 Hz, 1H), 5.38 (s, 1H), 5.22 (s, 2H), 4.30-4.16 (m, 2H), 4.09-3.92 (m, 3H), 3.17 (d, J=5.4 Hz, 1H), 1.64-1.49 (m, 1H), 1.44-1.27 (m, 1H), 1.16-0.99 (m, 1H), 0.79 (dd, J=8.3, 7.0 Hz, 6H).13C NMR (DMSO-d6) δ 175.25, 157.38, 149.02, 136.84, 136.78, 90.17, 90.09, 81.27, 72.87, 70.95, 64.48, 58.98, 38.97, 24.62, 15.98, 11.85. HR-ESIMS m/z calcd for [M+H]+C14H23N4O7359.1561, found, 359.1568. HPLC purity by Method A: 96% at 254 nm. iv. Synthesis of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl L-phenylalaninate (11) (1) Preparation of ((3ar,4r,6r,6ar)-6-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl-(((9H-fluoren-9-yl)methoxy)carbonyl)-L-phenylalaninate (33c) Intermediate 33c was synthesized from 31 (76 mg, 0.27 mmol, 1.0 eq.), 32c (155 mg, 0.40 mmol, 1.5 eq.), N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (77 mg, 0.40 mmol, 1.5 eq.) and 4-(dimethylamino)pyridine (39 mg, 0.32 mmol, 1.2 eq.) using similar procedure as for 33a to afford 33c as a white solid (119 mg, 68% yield).1H NMR (DMSO-d6) δ 12.29 (s, 1H), 7.90-7.80 (m, 3H), 7.62 (dd, J=7.4, 2.9 Hz, 2H), 7.53 (s, 1H), 7.39 (t, J=7.5 Hz, 2H), 7.34-7.11 (m, 7H), 6.06 (d, J=1.3 Hz, 1H), 4.99 (dd, J=6.2, 1.3 Hz, 1H), 4.69 (dd, J=6.0, 2.4 Hz, 1H), 4.29-4.06 (m, 8H), 3.01 (dd, J=13.8, 5.1 Hz, 1H), 2.92-2.82 (m, 1H), 1.43 (s, 3H), 1.24 (s, 3H). (2) Preparation of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl (((9H-fluoren-9-yl)methoxy)carbonyl)-L-phenylalaninate (34c) Intermediate 34c was synthesized from 33c (100 mg, 0.15 mmol) and formic acid (3 mL of 50% aqueous solution) using similar procedure as for 34a to afford 34c as a white solid (80 mg, 85% yield).1HNMR (DMSO-d6) δ 12.23 (s, 1H), 7.85 (t, J=8.0 Hz, 3H), 7.61 (d, J=7.5 Hz, 2H), 7.48 (s, 1H), 7.38 (t, J=7.5 Hz, 2H), 7.33-7.09 (m, 7H), 5.91 (d, J=3.1 Hz, 1H), 5.40 (d, J=5.1 Hz, 1H), 5.22 (d, J=6.1 Hz, 1H), 4.32 (d, J=9.0 Hz, 1H), 4.20 (dd, J=6.6, 4.3 Hz, 3H), 4.17-4.11 (m, 1H), 4.08-3.92 (m, 3H), 3.05 (dd, J=13.8, 4.7 Hz, 1H), 2.87 (dd, J=13.8, 10.3 Hz, 1H). (3) Preparation of Compound 11 Compound 11 was synthesized from 34c (75 mg, 0.12 mmol) and piperidine (1 mL of 5% v/v solution in DMF) using similar procedure as for 9 to afford 11 as a white solid (34 mg, 64% yield, 9:1 diastereomeric ratio).1HNMR (DMSO-d6) δ 7.53 (s, 1H), 7.50 (s, 1H), 7.22 (dd, J=8.2, 6.3 Hz, 2H), 7.16 (td, J=6.8, 6.3, 1.8 Hz, 3H), 5.90 (d, J=3.1 Hz, 1H), 5.38 (s, 1H), 5.22 (s, 3H), 4.26-4.16 (m, 2H), 4.03 (t, J=5.4 Hz, 1H), 4.00-3.87 (m, 2H), 3.57 (dd, J=7.3, 6.2 Hz, 1H), 2.86 (dd, J=13.4, 6.0 Hz, 1H), 2.75 (dd, J=13.4, 7.1 Hz, 1H). HR-ESIMS m/z calcd. for [M+H]+C17H21N4O7393.1405, found 393.1396. HPLC purity by Method B: >99% at 254 nm. v. Synthesis of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl D-isoleucinate (12) (1) Preparation of ((3ar,4r,6r,6ar)-6-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl (((9H-fluoren-9-yl)methoxy)carbonyl)-D-isoleucinate (33d) Intermediate 33d was synthesized from 31 (84 mg, 0.29 mmol, 1.0 eq.), 32d (156 mg, 0.44 mmol, 1.5 eq.), N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (85 mg, 0.44 mmol, 1.5 eq.) and DMAP (43 mg, 0.35 mmol, 1.2 eq.) using similar procedure as for 33a and afforded 33d as a white solid (86 mg, 47% yield).1HNMR (DMSO-d6) δ 12.27 (s, 1H), 7.92-7.82 (m, 2H), 7.79-7.66 (m, 3H), 7.52 (s, 1H), 7.39 (d, J=7.5 Hz, 2H), 7.30 (t, J=7.4 Hz, 2H), 6.07 (d, J=1.3 Hz, 1H), 5.03 (d, J=6.2 Hz, 1H), 4.74 (dd, J=6.1, 2.5 Hz, 1H), 4.36-4.05 (m, 5H), 4.03-3.87 (m, 1H), 1.77 (s, 1H), 1.43 (s, 3H), 1.38 (d, J=3.4 Hz, 1H), 1.25 (s, 3H), 0.81 (ddd, J=12.2, 8.0, 4.7 Hz, 6H). (2) Preparation of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl (((9H-fluoren-9-yl)methoxy)carbonyl)-D-isoleucinate (34d) Intermediate 34d was synthesized from 33d (80 mg, 0.13 mmol) and formic acid (2 mL of 50% aqueous solution) using similar procedure as for 34a and afforded 34d as a white solid (47 mg, 63% yield).1HNMR (DMSO-d6) δ 12.20 (s, 1H), 7.87 (d, J=7.6 Hz, 2H), 7.82-7.66 (m, 3H), 7.53 (s, 1H), 7.40 (t, J=7.5 Hz, 2H), 7.31 (td, J=7.4, 2.6 Hz, 2H), 5.90 (d, J=2.7 Hz, 1H), 5.41 (d, J=4.9 Hz, 1H), 5.23 (d, J=6.1 Hz, 1H), 4.45-4.15 (m, 5H), 4.10-3.86 (m, 4H), 1.76 (s, 1H), 1.34 (dd, J=12.7, 6.1 Hz, 1H), 1.19 (dd, J=14.5, 7.3 Hz, 1H), 0.79 (q, J=7.0 Hz, 6H). (3) Preparation of Compound 12 Compound 10 was synthesized from 34d (45 mg, 0.07 mmol) and piperidine (1 mL of 5% v/v solution in DMF), using similar procedure as for 9 to afford 12 as a white solid (23 mg, 82% yield).1HNMR (DMSO-d6) δ 7.51 (s, 1H), 5.89 (d, J=2.8 Hz, 1H), 5.40 (d, J=5.0 Hz, 1H), 5.21 (s, 1H), 4.30-4.20 (m, 1H), 4.20-4.14 (m, 1H), 4.05 (t, J=5.4 Hz, 1H), 3.99-3.93 (m, 2H), 3.17 (d, J=5.4 Hz, 1H), 1.60-1.48 (m, 1H), 1.36 (ddd, J=13.5, 7.5, 4.4 Hz, 1H), 1.08 (ddd, J=13.5, 8.9, 7.3 Hz, 1H), 0.84-0.71 (m, 6H). HR-ESIMS m/z calcd. for C14H23N4O7[M+H]+359.1561, found 359.1563. HPLC purity by Method A: 98% at 254 nm. vi. Synthesis of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl D-alloisoleucinate (13) (1) Preparation of ((3ar,4r,6r,6ar)-6-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl (((9H-fluoren-9-yl)methoxy)carbonyl)-D-allisoleucinate (33e) Intermediate 33e was synthesized from 31 (84. mg, 0.29 mmol, 1.0 eq.), N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (85 mg, 0.44 mmol, 1.5 eq.) and 32e (156 mg, 0.44 mmol, 1.5 eq.) and 4-(dimethylamino)pyridine (43 mg, 0.35 mmol, 1.2 eq.) using similar procedure as for 33a to afford 33e as a white solid (123 mg, 67% yield).1HNMR (DMSO-d6) δ 12.27 (s, 1H), 7.87 (d, J=7.5 Hz, 2H), 7.72 (t, J=6.9 Hz, 3H), 7.51 (d, J=1.3 Hz, 1H), 7.44-7.36 (m, 2H), 7.30 (tdd, J=7.4, 3.0, 1.2 Hz, 2H), 6.07 (d, J=1.3 Hz, 1H), 5.02 (dd, J=6.1, 1.3 Hz, 1H), 4.74 (dd, J=6.1, 2.5 Hz, 1H), 4.32-3.94 (m, 6H), 1.85 (dq, J=13.4, 6.5 Hz, 1H), 1.43 (s, 3H), 1.25 (s, 3H), 1.18-0.99 (m, 1H), 0.88-0.77 (m, 6H). (2) Preparation of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl (((9H-fluoren-9-yl)methoxy)carbonyl)-D-alloisoleucinate (34e) Intermediate 34e was synthesized from 33e (119 mg, 0.19 mmol) and formic acid (2 mL of 50% aqueous solution) using similar procedure as for 34a to afford 34e as a white solid (84 mg, 75% yield).1HNMR (DMSO-d6) δ 12.24 (s, 1H), 7.88 (d, J=7.5 Hz, 2H), 7.80-7.66 (m, 3H), 7.53 (s, 1H), 7.44-7.20 (m, 4H), 5.90 (d, J=2.7 Hz, 1H), 5.41 (d, J=4.9 Hz, 1H), 5.23 (d, J=6.2 Hz, 1H), 4.41-4.09 (m, 5H), 4.05-3.93 (m, 2H), 1.84 (dt, J=13.1, 6.8 Hz, 1H), 1.41-1.19 (m, 2H), 1.12 (dt, J=13.9, 7.2 Hz, 1H), 0.91-0.68 (m, 6H). (3) Preparation of Final Target 13 Compound 13 was synthesized from 34e (80 mg, 0.14 mmol) and piperidine (1 mL of 5% v/v in DMF) using similar procedure as for 9 to afford 13 as a white solid (34 mg, 69% yield).1HNMR (DMSO-d6) δ 7.50 (d, J=0.5 Hz, 1H), 5.89 (d, J=2.7 Hz, 1H), 5.40 (d, J=4.9 Hz, 1H), 5.21 (s, 1H), 4.34-4.23 (m, 1H), 4.18 (s, 1H), 4.04 (s, 1H), 4.01-3.90 (m, 2H), 3.28 (d, J=4.4 Hz, 1H), 1.60 (dtd, J=7.7, 6.5, 4.4 Hz, 1H), 1.36 (ddd, J=13.7, 7.5, 6.3 Hz, 1H), 1.11 (dt, J=13.3, 7.4 Hz, 1H), 0.82 (t, J=7.4 Hz, 3H), 0.71 (d, J=6.9 Hz, 3H). HR-ESIMS m/z calcd. for C14H23N4O7[M+H]+359.1561 found 359.1562. HPLC purity by Method B: >99% at 254 nm. f. Synthesis of Compound Nos. 14-16 i. Synthesis of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)-methyl isobutyrate (14) (1) Preparation of ((3ar,4r,6r,6ar)-6-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl isobutyrate (35a) 2-Methyl propanoyl chloride (0.11 mL, 1.05 mmol, 1.5 eq.) was added as a solution in anhydrous dichloromethane (1 mL) to a pre-stirred solution of 31 (200 mg, 0.7 mmol, 1.0 eq.) in anhydrous pyridine (5 mL, 0.70 mmol, 1.0 eq.) at 0° C. and the reaction mixture was allowed to warm to room temperature overnight. The reaction mixture was concentrated under vacuum to dryness. The residue was chromatographed (0-30% methanol in dichloromethane) to afford 35a (185 mg, 74% yield) as a white viscous solid.1HNMR (DMSO-d6) δ 12.26 (s, 1H), 7.53 (s, 1H), 6.06 (d, J=1.3 Hz, 1H), 5.03 (dd, J=6.1, 1.3 Hz, 1H), 4.75 (dd, J=6.1, 2.9 Hz, 1H), 4.19 (dd, J=6.5, 2.9 Hz, 1H), 4.09 (dd, J=6.4, 1.0 Hz, 2H), 2.60-2.50 (m, 1H), 1.45 (s, 3H), 1.28 (s, 3H), 1.04 (dd, J=6.9, 0.9 Hz, 6H). (2) Preparation of Compound 14 Intermediate 35a (180 mg, 0.51 mmol) was taken in a 20 mL vial and added formic acid (2 mL, 0.51 mmol, 50% by v/v in water). The mixture was warmed to 65° C. stirred for 30 min and concentrated to dryness under vacuum at 50° C. The residue obtained was co-evaporated with methanol (2×5 mL) to remove residual formic acid. The residue was purified by flash column chromatography (0-30% methanol in dichloromethane) to afford 14 (103 mg, 64% yield) as a sticky white solid.1H NMR (DMSO-d6) δ 12.23 (s, 1H), 7.53 (s, 1H), 5.88 (d, J=3.0 Hz, 1H), 5.36 (d, J=5.0 Hz, 1H), 5.18 (d, J=6.2 Hz, 1H), 4.32-4.14 (m, 2H), 4.04 (q, J=5.5 Hz, 1H), 3.95 (dd, J=8.3, 5.8 Hz, 2H), 2.52 (d, J=7.0 Hz, 1H), 1.05 (dd, J=7.0, 3.4 Hz, 6H).13C NMR (DMSO-d6) δ 176.37, 156.94, 148.64, 136.87, 136.83, 90.14, 90.09, 81.23, 72.91, 70.87, 64.39, 33.58, 19.16. HR-ESIMS calcd. for [M+H]+C12H18N3O7316.1139, found 316.1138. HPLC purity by Method A: 99% at 254 nm. ii. Synthesis of ((2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl stearate (15) (1) Preparation of ((3ar,4r,6r,6ar)-6-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl stearate (35b) Stearoyl chloride (164 mg, 0.54 mmol) was added to a solution of 31 in anhydrous pyridine (3 mL, 0.36 mmol) at 0° C. and the mixture was warmed to room temperature and stirred for 18 h. The reaction was quenched with water (1 mL) and diluted with ethyl acetate (10 mL). The organic layer was washed with water (5 mL), dried over sodium sulfate, filtered, and then the filtrate was evaporated under vacuum. The residual pyridine was removed by co-evaporation with toluene (2×5 mL). The crude residue was purified by column chromatography (0-100% ethyl acetate in dichloromethane) to afford 35b (160 mg, 80% yield) as a white solid.1HNMR (DMSO-d6) δ 12.27 (s, 1H), 7.52 (s, 1H), 6.06 (s, 1H), 5.01 (d, J=6.1 Hz, 1H), 4.74 (dd, J=6.1, 2.9 Hz, 1H), 4.19 (td, J=6.7, 6.0, 3.0 Hz, 1H), 4.08 (d, J=7.3 Hz, 2H), 2.25 (t, J=7.2 Hz, 2H), 1.45 (m, 5H), 1.24 (d, J=26.2 Hz, 33H), 0.83 (t, J=5.6 Hz, 3H). (2) Preparation of Compound 15 Formic acid (1 mL, 50% v/v in water) was added to 35b (128 mg, 0.23 mmol) and mixture was heated at 65° C. with stirring for 1 h. The mixture was concentrated at 50° C. under vacuum to remove formic acid. The residue was re-dissolved in methanol (2 mL) and concentrated down with silica gel (2 g). The slurry was purified by column chromatography to yield 15 (87 mg, 72% yield) as a white solid.1HNMR (DMSO-d6) δ 12.23 (s, 1H), 7.53 (s, 1H), 5.88 (d, J=3.0 Hz, 1H), 5.38 (d, J=5.0 Hz, 1H), 5.19 (d, J=6.2 Hz, 1H), 4.25 (dd, J=10.5, 4.0 Hz, 1H), 4.17 (td, J=4.8, 2.9 Hz, 1H), 4.02 (t, J=5.5 Hz, 1H), 3.98-3.86 (m, 2H), 2.25 (t, J=7.4 Hz, 2H), 1.51-1.42 (m, 3H), 1.21 (d, J=2.4 Hz, 31H), 0.89-0.77 (m, 3H). HR-ESIMS m/z calcd. for C26H46N3O7512.3330, found 512.3319. HPLC purity by Method A: 98% at 254 nm. iii. Synthesis of (2r,3s,4r,5r)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl) methyl diethyl phosphate (16) (1) Preparation of ((3ar,4r,6r,6ar)-6-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl (diethoxyphosphoryl) formate (35c) Compound 31 (100 mg, 0.35 mmol, 1.0 eq.) was co-evaporated with anhydrous pyridine (2×2 mL) and the residue was dried under vacuum overnight. The dried 31 was dissolved in anhydrous pyridine (5 mL), cooled to 0° C. and added 1-[chloro(ethoxy)phosphoryl]oxyethane (0.07 mL, 0.53 mmol, 1.5 eq.) dropwise under argon atmosphere. The reaction mixture was further stirred for 18 h at room temperature. On completion of the reaction, the mixture was concentrated to remove pyridine by co-evaporation with toluene (2×5 mL). The residue was purified by column chromatography (0-100% ethyl acetate in dichloromethane) to afford 35c (31 mg, 21% yield) as a colorless oil.1HNMR (DMSO-d6) δ 12.28 (s, 1H), 7.52 (d, J=2.7 Hz, 1H), 6.08 (d, J=2.4 Hz, 1H), 5.02 (dd, J=6.2, 2.4 Hz, 1H), 4.75 (dt, J=5.9, 2.8 Hz, 1H), 4.22 (tt, J=5.5, 2.7 Hz, 1H), 3.98 (dtt, J=21.5, 8.5, 4.5 Hz, 6H), 1.46 (d, J=2.5 Hz, 3H), 1.28 (d, J=2.5 Hz, 3H), 1.19 (ddd, J=9.0, 6.0, 1.9 Hz, 6H). (2) Preparation of Compound 16 Compound 35c (25 mg, 0.06 mmol) dissolved in formic acid (1 mL of 50% v/v solution in water) and the mixture was heated to 65° C. for 1 h. The mixture was concentrated to remove excess formic acid with co-evaporation with methanol (2×5 mL). The residue was purified by column chromatography to yield 16 (20 mg, 88% yield) as a viscous oil.1H NMR (DMSO-d6) δ 12.25 (s, 1H), 7.52 (d, J=0.6 Hz, 1H), 5.90 (d, J=3.0 Hz, 1H), 5.40 (d, J=5.0 Hz, 1H), 5.23 (d, J=6.3 Hz, 1H), 4.18 (td, J=5.0, 3.1 Hz, 1H), 4.09-3.84 (m, 8H), 1.20 (tdd, J=7.0, 2.4, 0.9 Hz, 6H).31P NMR (DMSO-d6) δ −1.23. HR-ESIMS m/z calcd. for C12H20N3O9P [M+H]+382.1010, found 382.1015. HPLC purity by Method A: 99% at 254 nm. g. Synthesis of 2-ethylbutyl ((((2r,9,4r,5r)-5-(5-amino-3-oxo-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-(phenoxy)phosphoryl)-L-alaninate (17) i. Preparation of (2r,3r,4r,5r)-2-(acetoxymethyl)-5-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3)-yl)tetrahydrofuran-3,4-diyl diacetate (36) To a solution of 6-azauridine (2.0 g, 8.16 mmol, 1.0 eq.) in 120 mL of anhydrous acetonitrile was added 4-dimethylaminopyridine (120 mg, 0.98 mmol, 0.12 eq), N,N′-diisopropylethylamine (5.83 mL, 33.4 mmol, 4.1 eq), followed by acetic anhydride (3.08 mL, 32.6 mmol, 4.0 eq). The reaction mixture was stirred at 20° C. for 18 h and then was evaporated under reduced pressure to afford an oil, which was dissolved in 200 mL of ethyl acetate. The organic layer was washed with saturated sodium bicarbonate (2×50 mL), sat ammonium chloride (2×50 mL), followed by brine (50 mL). The organic layer was separated, dried in sodium sulfate, filtered, and then the filtrate was evaporated in vacuum to afford 2.20 g (73%) of 36 as a white foamy solid.1HNMR (DMSO-d6) δ 12.35 (s, 1H), 7.68 (d, J=0.6 Hz, 1H), 6.27-6.02 (m, 1H), 5.62-5.49 (m, 1H), 5.35 (ddd, J=6.0, 5.6, 0.4 Hz, 1H), 4.40-4.23 (m, 2H), 4.14-3.97 (m, 1H), 2.10 (s, 3H), 2.07 (s, 3H), 2.03 (s, 3H). HR-ESIMS m/z calcd. for C14H17N3O9·H, 372.1038, found 372.1036. ii. Preparation of 5-amino-2-((2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl)-1,2,4-triazin-3(2H)-one (38) To a mixture of 36 (2.00 g, 5.39 mmoles, 1.0 eq), triisopropylbenzenesulfonyl chloride (4.80 g, 15.8 mmoles, 2.94 eq) and 4-dimethylaminopyridine (2.04 g, 16.7 mmoles, 3.1 eq) was added 30 mL of anhydrous acetonitrile, followed by triethylamine (1.58 mL, 11.3 mmoles, 2.1 eq). The reaction mixture was stirred at 20° C. for 24 h to afford 37, which was reacted further without isolation. To the reaction mixture containing intermediate 37 was added 28% aqueous ammonium hydroxide (103 mL, 747 mmol, 135 eq.) and then the reaction mixture was stirred at 20° C. for 3 days. The reaction mixture was evaporated under reduced pressure to afford a solid, which was purified by flash chromatography (80 g silica column, 100-80% dichloromethane in methanol, gradient elution) to provide 1.34 g of crude product as a yellow solid. Recrystallization from a mixture of boiling methanol (18 mL) and water (2 mL) gave 534 mg (40%) of 38 as a yellow crystalline solid.1HNMR (DMSO-d6) δ 7.95 (d, J=47.0 Hz, 2H), 7.51 (d, J=0.6 Hz, 1H), 5.98 (d, J=4.1 Hz, 1H), 5.19 (d, J=5.6 Hz, 1H), 4.98 (d, J=5.9 Hz, 1H), 4.67 (t, J=5.8 Hz, 1H), 4.21 (td, J=5.4, 4.1 Hz, 1H), 3.99 (q, J=5.5 Hz, 1H), 3.78 (td, J=5.6, 4.4 Hz, 1H), 3.56-3.36 (m, 2H). iii. Preparation of Compound 17 The final target 17 was prepared from 38 (200 mg, 0.819 mmol, 1.0 eq), 27b (Scheme-2) (487 mg, 0.983 mmol, 1.2 eq.), and 1M (in hexanes) dimethylaluminum chloride (0.409 mL, 0.409 mmol, 0.50 eq.) in a mixture of anhydrous pyridine (2.46 mL) and 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidone (DMPU) (0.495 mL, 4.09 mmol, 5.0 eq) by the procedure described for the preparation of 2 to afford a semi-solid. Two purifications by flash chromatography (40 g and 24 g silica columns, 100-90% dichloromethane in methanol, gradient elution) provided 35 mg of a yellow gummy solid. The solid was dissolved in a mixture of 2 drops of methanol and 1.0 mL of dichloromethane. Hexane was added until a precipitate formed which was filtered by vacuum filtration to give 14 mg (3%) of a yellow solid as a single diastereomer containing residual DMPU.1HNMR (DMSO-d6) δ 8.05 (s, 1H), 7.95 (s, 1H), 7.51 (d, J=1.7 Hz, 1H), 7.37 (dd, J=8.6, 7.3 Hz, 1H), 7.26-7.11 (m, 3H), 6.05 (t, J=3.6 Hz, 1H), 6.02-5.92 (m, 1H), 5.35 (dd, J=5.2, 1.8 Hz, 1H), 5.18 (dd, J=6.2, 1.6 Hz, 1H), 4.25-3.73 (m, 9H), 1.60-1.16 (m, 15H), 0.94-0.78 (m, 11H);31P NMR (DMSO-d6) δP3.54. LCMS m/z 556 (M+H)+. HR-ESIMS m/z calcd for C23H34N5O9P·H, 556.2167, found 556.2162. HPLC purity by Method B: 94% at 254 nm. h. Synthesis of Compound Nos. 18-24 i. Preparation of 5-amino-2-((2r,3r,4r,5r)-3,4-bis((tert-butyl-dimethylsilyl)oxy)-5-(((tert-butyldimethylsilyl)oxy)-methyl)tetrahydrofuran-2-yl)-1,2,4-triazin-3(2H)-one (39) To a solution of 38 (Scheme-6) (225 mg, 0.92 mmol, 1.0 eq.) in 4.0 mL of anhydrous N,N′-dimethylformamide was added imidazole (376 mg, 5.53 mmol, 6.0 eq.), followed by tetrabutyldimethylchlorosilane (555 mg, 3.69 mmol, 4.0 eq.). The reaction mixture was stirred at 20° C. for 18 h. The reaction mixture was diluted with 100 mL of dichloromethane, washed with water (2×50 mL), saturated sodium bicarbonate (2×50 mL), saturated ammonium chloride (2×50 mL), followed by brine (50 mL). The organic layer was separated, dried over sodium sulfate, filtered, and then the filtrate was evaporated under reduced pressure to afford an oil. Purification by flash chromatography (40 g silica column, 100-0% hexane in ethyl acetate, gradient elution) provided 91 mg (17%) of 39 as a yellow solid.1HNMR (DMSO-d6) δ 8.04 (s, 1H), 7.89 (s, 1H), 7.53 (d, J=0.6 Hz, 1H), 6.03 (d, J=5.3 Hz, 1H), 4.49 (dd, J=5.4, 4.5 Hz, 1H), 4.27 (dd, J=4.6, 3.4 Hz, 1H), 3.89-3.81 (m, 1H), 3.71-3.58 (m, 2H), 0.92 (s, 9H), 0.88 (s, 9H), 0.84 (s, 9H), 0.11 (d, J=5.0 Hz, 6H), 0.05-−0.00 (m, 9H), −0.05 (s, 3H). LCMS m/z 587 (M+H)+. ii. Synthesis of N-(2-((2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)-tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)-2-propylpentanamide (18) (1) Preparation of N-(2-((2r,3r,4r,5r)-3,4-bis((tert-butyl-dimethylsilyl)oxy)-5-(((tert-butyldimethylsilyl)oxy)-methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)-2-propylpentanamide (40a) To a solution of 39 (205 mg, 0.35 mmol, 1.0 eq) in 5.0 mL of anhydrous dichloromethane was added a solution of 2,2-di-n-propylacetyl chloride (62 mg, 0.38 mmol, 1.1 eq.) in 1.0 mL of anhydrous dichloromethane, followed by N,N′-diisopropylethylamine (0.067 mL, 0.38 mmol, 1.1 eq). The reaction mixture was irradiated with microwaves at 120° C. for 1 h and then evaporated under reduced pressure to afford an oil, which was purified by flash chromatography (40 g, silica column, 100-70% hexane in ethyl acetate, gradient elution) to provide 223 mg (89%) of 40a as a colorless solid.1HNMR (CDCl3) δ 8.94 (s, 1H), 8.19 (s, 1H), 6.24 (d, J=4.1 Hz, 1H), 4.51 (t, J=4.3 Hz, 1H), 4.33 (t, J=4.5 Hz, 1H), 4.04 (q, J=4.4 Hz, 1H), 3.81-3.59 (m, 2H), 2.44-2.32 (m, 1H), 1.72-1.60 (m, 2H), 1.55-1.45 (m, 2H), 1.42-1.28 (m, 4H), 0.98-0.82 (m, 33H), 0.13-−0.05 (m, 18H). LCMS m/z 728 (M+H)+. (2) Preparation of Compound 18 To a cold (0° C.) solution of 40a (210 mg, 0.30 mmol, 1.0 eq.) in 10.0 mL of anhydrous tetrahydrofuran was added a 1M solution of tetrabutylammonium fluoride (0.931 mL, 0.93 mmol, 3.1 eq.) in tetrahydrofuran. The reaction mixture was stirred for 18 h as it warmed to 20° C. The reaction mixture was evaporated under reduced pressure to afford a residue, which was purified by flash chromatography (40 g silica column, 100-95% dichloromethane in methanol, gradient elution) to provide 60 mg (54%) of 18 as a white solid.1HNMR (DMSO-d6) δ 11.46 (s, 1H), 8.80 (d, J=0.5 Hz, 1H), 6.05 (d, J=3.6 Hz, 1H), 5.31 (d, J=5.3 Hz, 1H), 5.05 (d, J=6.0 Hz, 1H), 4.65 (dd, J=6.1, 5.5 Hz, 1H), 4.28 (td, J=5.2, 3.6 Hz, 1H), 4.08 (q, J=5.6 Hz, 1H), 3.86 (td, J=5.7, 4.2 Hz, 1H), 3.60-3.37 (m, 2H), 2.67 (tt, J=9.1, 5.0 Hz, 1H), 1.57 (dtd, J=13.0, 8.8, 7.1 Hz, 2H), 1.46-1.34 (m, 2H), 1.33-1.20 (m, 4H), 0.88 (t, J=7.3 Hz, 6H). LCMS m/z 371 (M+H)+. HR-ESIMS m/z calcd for C16H26N4O6·H, 371.19251, found 371.19248. HPLC purity by Method B: 99% at 254 nm. iii. Synthesis of N-(2-((2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxy-methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)dodecanamide (19) (1) Preparation of N-(2-((2r,3r,4r,5r)-3,4-bis((tert-butyl-dimethylsilyl)oxy)-5-(((tert-butyldimethylsilyl)oxy)-methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)dodecanamide (40b) Intermediate 40b was prepared from 39 (85 mg, 0.145 mmol, 1.0 eq.) and dodecyl chloride (32 mg, 0.145 mmol, 1.0 eq.) in 2.0 mL of anhydrous dichloromethane with N,N′-diisopropylethylamine (0.025 mL, 0.145 mmol, 1.0 eq.) as base according to the procedure described for the preparation of 40a to afford a residue. Purification by flash chromatography (40 g silica column, 100-79% hexane in ethyl acetate, gradient elution) provided 81 mg (73%) of a colorless tacky solid.1HNMR (CDCl3) δ 8.94 (s, 1H), 8.57 (s, 1H), 6.25 (d, J=4.4 Hz, 1H), 4.53 (t, J=4.5 Hz, 1H), 4.32 (t, J=4.4 Hz, 1H), 4.05 (q, J=4.4 Hz, 1H), 3.83-3.61 (m, 2H), 2.51 (t, J=7.4 Hz, 2H), 1.70 (q, J=7.3 Hz, 2H), 1.28 (d, J=3.5 Hz, 16H), 0.93 (s, 9H), 0.91-0.85 (m, 21H), 0.11 (d, J=2.6 Hz, 6H), 0.07-0.00 (m, 9H), −0.02 (s, 3H). LCMS m/z 769 (M+H)+. (2) Preparation of Compound 19 The final target 19 was prepared from 40b (73 mg, 0.095 mmol, 1.0 eq.) and 1M (in tetrahydrofuran) tetrabutylammonium fluoride (0.294 mL, 0.294 mmol., 3.1 eq.) in 5.0 mL of anhydrous tetrahydrofuran according to the procedure described for the preparation of 18 to afford an oil. Purification by flash chromatography (24 g silica column, 100-95% dichloromethane in methanol, gradient elution) provided 26 mg (64%) of a white solid.1HNMR (DMSO-d6) δ 11.38 (s, 1H), 8.74 (s, 1H), 6.04 (d, J=3.7 Hz, 1H), 5.32 (dd, J=5.4, 0.8 Hz, 1H), 5.06 (d, J=5.9 Hz, 1H), 4.67 (t, J=5.8 Hz, 1H), 4.27 (td, J=5.3, 3.7 Hz, 1H), 4.06 (q, J=5.6 Hz, 1H), 3.85 (td, J=5.7, 4.2 Hz, 1H), 3.55 (ddd, J=11.7, 5.6, 4.2 Hz, 1H), 3.42 (dt, J=11.9, 6.0 Hz, 1H), 2.47 (t, J=7.3 Hz, 2H), 1.57 (p, J=7.4 Hz, 2H), 1.27 (d, J=3.0 Hz, 16H), 0.95-0.82 (m, 3H). LCMS m/z 427 (M+H)+. HR-ESIMS m/z calcd for C20H34N4O6·H, 427.25511, found 427.25519. HPLC purity by Method A: 99% at 254 nm. iv. N-(2-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)tetrahydro-furan-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)butyramide (20) (1)N-(2-((2R,3R,4R,5R)-3,4-bis((tert-butyldimethylsilyl)oxy)-5-(((tert-butyldimethylsilyl)oxy)methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)butyramide (40c) Intermediate 40c was prepared from 39 (200 mg, 0.341 mmoles, 1.0 eq) and butanoyl chloride (32 39.9, 0.375 mmoles, 1.1 eq) in 3.0 mL of anhydrous dichloromethane with N, N′-diisopropylethylamine (0.065 mL, 0.375 mmoles, 1.0 eq) as base according to the procedure described for the preparation of 40a to afford a residue. Purification by flash chromatography (40 g silica column, 100-0% hexane in ethyl acetate, gradient elution) provided 152 mg (68%) of a white foamy solid; 1H NMR (400 MHz, cdcl3) δ 8.93 (s, 1H), 8.48 (s, 1H), 6.24 (d, J=4.5 Hz, 1H), 4.52 (t, J=4.5 Hz, 1H), 4.31 (t, J=4.4 Hz, 1H), 4.04 (q, J=4.4 Hz, 1H), 3.80-3.60 (m, 2H), 2.48 (t, J=7.3 Hz, 2H), 1.75 (h, J=7.4 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H), 0.95-0.77 (m, 28H), 0.10 (d, J=2.6 Hz, 18H); LCMS m/z 657 (M+H)+. (2) Preparation of 20 The final target 20 was prepared from 40c (145 mg, 0.221 mmoles, 1.0 eq) and tetraethylammonium fluoride hydrate (221 mg, 1.32 mmoles, 6 eq) in 7.0 mL of anhydrous tetrahydrofuran for 3 hrs at 60° C. to afford an oil. Purification by flash chromatography (24 g silica column, 100-90% dichloromethane in methanol, gradient elution) provided 41 mg (59%) of a white crystalline solid. 1H NMR (400 MHz, dmso) δ 11.39 (s, 1H), 8.74 (s, 1H), 6.04 (d, J=3.7 Hz, 1H), 5.32 (d, J=5.5 Hz, 1H), 5.07 (d, J=6.0 Hz, 1H), 4.67 (t, J=5.8 Hz, 1H), 4.27 (td, J=5.2, 3.7 Hz, 1H), 4.06 (q, J=5.6 Hz, 1H), 3.85 (td, J=5.6, 4.1 Hz, 1H), 3.60-3.38 (m, 2H), 2.46 (t, J=7.3 Hz, 2H), 1.60 (h, J=7.4 Hz, 2H), 0.92 (t, J=7.4 Hz, 3H); HRMS: calc for C12H18N4O6·H, 315.12991, found, 315.1296; HPLC 98.6% at 254 nm. v. N-(2-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)tetrahydro-furan-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)cyclopropane-carboxamide (21) (1) N-(2-((2R,3R,4R,5R)-3,4-bis((tert-butyldimethylsilyl)oxy)-5-(((tert-butyldimethylsilyl)oxy)methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)cyclopropane-carboxamide (40d) Intermediate 40d was prepared from 39 (200 mg, 0.341 mmoles, 1.0 eq) and cyclopropanecarbonyl chloride (39 mg, 0.375 mmoles, 1.1 eq) in 3.0 mL of anhydrous dichloromethane with N, N′-diisopropylethylamine (0.065 mL, 0.375 mmoles, 1.1 eq) as base according to the procedure described for the preparation of 40a to afford a residue. Purification by flash chromatography (40 g silica column, 100-0% hexane in ethyl acetate, gradient elution) provided 142 mg (64%) of a white foamy solid; LCMS m/z 655 (M+H)+. (2) Preparation of 21 The final target 21 was prepared from 40d (131 mg, 0.20 mmoles, 1.0 eq) and tetraethylammonium fluoride hydrate (207 mg, 1.24 mmoles, 6.2 eq) in 7.0 mL of anhydrous tetrahydrofuran for 3 hrs at 65° C. to afford an oil. Purification by flash chromatography (24 g silica column, 100-90% dichloromethane in methanol, gradient elution) provided 39 mg (62%) of a white solid.1H NMR (400 MHz, dmso) δ 11.76 (s, 1H), 8.75 (s, 1H), 6.04 (d, J=3.6 Hz, 1H), 5.31 (d, J=5.4 Hz, 1H), 5.06 (d, J=6.0 Hz, 1H), 4.67 (t, J=5.8 Hz, 1H), 4.27 (td, J=5.2, 3.6 Hz, 1H), 4.08 (dq, J=18.3, 5.4 Hz, 1H), 3.85 (td, J=5.6, 4.1 Hz, 1H), 3.59-3.37 (m, 2H), 2.16-1.95 (m, 1H), 1.08-0.82 (m, 4H); LCMS m/z 313 (M+H)+; HRMS: calc for C12H16N4O6·H, 313.11426, found, 313.11397; HPLC 100% at 254 nm. vi. Synthesis of N-(2-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)-tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)-cyclobutanecarboxamide (22) (1) N-(2-((2R,3R,4R,5R)-3,4-bis((tert-butyldimethylsilyl)oxy)-5-(((tert-butyldimethylsilyl)oxy)methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)cyclobutane-carboxamide (40e) Intermediate 40e was prepared from 39 (200 mg, 0.341 mmoles, 1.0 eq) and cyclobutanecarbonyl chloride (44 mg, 0.375 mmoles, 1.1 eq) in 3.0 mL of anhydrous dichloromethane with N, N′-diisopropylethylamine (0.065 mL, 0.375 mmoles, 1.1 eq) as base according to the procedure described for the preparation of 40a to afford a residue. Purification by flash chromatography (40 g silica column, 100-0% hexane in ethyl acetate, gradient elution) provided 176 mg (77%) of an off-white foamy solid;1H NMR (400 MHz, cdcl3) δ 8.96 (s, 1H), 8.54 (s, 1H), 6.24 (d, J=4.5 Hz, 1H), 4.53 (t, J=4.5 Hz, 1H), 4.31 (t, J=4.4 Hz, 1H), 4.04 (q, J=4.4 Hz, 1H), 3.71 (qd, J=11.2, 4.5 Hz, 2H), 3.43-3.28 (m, 1H), 2.47-2.19 (m, 4H), 2.16-1.84 (m, 2H), 1.05-0.75 (m, 28H), 0.10 (d, J=2.6 Hz, 18H); LCMS m/z 669 (M+H)+. (2) Preparation of 22 The final target 22 was prepared from 40e (155 mg, 0.23 mmoles, 1.0 eq) and tetraethylammonium fluoride hydrate (232 mg, 1.39 mmoles, 6 eq) in 7.0 mL of anhydrous tetrahydrofuran for 3 hrs at 65° C. to afford a residue. Purification by flash chromatography (24 g silica column, 100-90% dichloromethane in methanol, gradient elution) provided 31 mg (41%) of a white solid.1H NMR (400 MHz, dmso) δ 11.27 (s, 1H), 8.77 (s, 1H), 6.04 (d, J=3.7 Hz, 1H), 5.32 (d, J=5.3 Hz, 1H), 5.06 (d, J=6.0 Hz, 1H), 4.68 (t, J=5.8 Hz, 1H), 4.27 (td, J=5.2, 3.6 Hz, 1H), 4.06 (q, J=5.5 Hz, 1H), 3.85 (td, J=5.6, 4.1 Hz, 1H), 3.60-3.37 (m, 3H), 2.32-2.07 (m, 4H), 1.95 (dp, J=11.8, 8.6 Hz, 1H), 1.87-1.74 (m, 1H); LCMS m/z 327 (M+H)+; HRMS: calc for C13H18N4O6·H, 327.12991, found, 327.1294; HPLC 92.8% at 254 nm. vii. Synthesis of ethyl (2-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxy-methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)carbamate (23) (1) ethyl (2-((2R,3R,4R,5R)-3,4-bis((tert-butyldimethylsilyl)oxy)-5-(((tert-butyldimethylsilyl)oxy)methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)carbamate (40f) Intermediate 40f was prepared from 39 (200 mg, 0.341 mmoles, 1.0 eq) and ethyl chloroformate (41 mg, 0.375 mmoles, 1.1 eq) in 3.0 mL of anhydrous dichloromethane with N, N′-diisopropylethylamine (0.059 mL, 0.341 mmoles, 1 eq) as base according to the procedure described for the preparation of 40a to afford a residue. Purification by flash chromatography (40 g silica column, 100-0% hexane in ethyl acetate, gradient elution) provided 117 mg (52%) of white solid;1H NMR (400 MHz, cdcl3) δ 8.79 (s, 1H), 7.59 (s, 1H), 6.24 (d, J=4.6 Hz, 1H), 4.53 (t, J=4.6 Hz, 1H), 4.37-4.25 (m, 3H), 4.04 (q, J=4.4 Hz, 1H), 3.71 (qd, J=11.2, 4.5 Hz, 2H), 1.36 (t, J=7.1 Hz, 3H), 1.02-0.77 (m, 27H), 0.10 (d, J=2.4 Hz, 18H); LCMS m/z 659 (M+H)+. (2) Preparation of 23 The final target 23 was prepared from 40f (104 mg, 0.16 mmoles, 1.0 eq) and tetraethylammonium fluoride hydrate (121 mg, 0.73 mmoles, 4.6 eq) in 5.0 mL of anhydrous tetrahydrofuran for 3 hrs at 65° C. to afford a residue. Purification by flash chromatography (24 g silica column, 100-90% dichloromethane in methanol, gradient elution) provided 31 mg (62%) of a white crystalline solid.1H NMR (400 MHz, dmso) δ 11.31 (s, 1H), 8.58 (s, 1H), 6.03 (d, J=3.7 Hz, 1H), 5.31 (d, J=5.4 Hz, 1H), 5.05 (d, J=6.0 Hz, 1H), 4.68 (t, J=5.8 Hz, 1H), 4.34-4.15 (m, 3H), 4.05 (q, J=5.5 Hz, 1H), 3.85 (td, J=5.7, 4.2 Hz, 1H), 3.60-3.37 (m, 2H), 1.27 (t, J=7.1 Hz, 3H); LCMS m/z 339 (M+Na)+; HRMS: calc for C11H16N4O7·H, 317.10981, found, 317.10918; HPLC 96.6% at 254 nm. viii. Synthesis of pentyl (2-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxy-methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)carbamate (24) (1) pentyl (2-((2R,3R,4R,5R)-3,4-bis((tert-butyldimethylsilyl)oxy)-5-(((tert-butyldimethylsilyl)oxy)methyl)tetrahydrofuran-2-yl)-3-oxo-2,3-dihydro-1,2,4-triazin-5-yl)carbamate (40 g) Intermediate 40 g was prepared from 39 (200 mg, 0.341 mmoles, 1.0 eq) and pentyl chloridocarbonate (56 mg, 0.375 mmoles, 1.1 eq) in 3.0 mL of anhydrous dichloromethane with N, N′-diisopropylethylamine (0.065 mL, 0.375 mmoles, 1 eq) as base according to the procedure described for the preparation of 40a to afford a residue. Purification by flash chromatography (40 g silica column, 100-0% hexane in ethyl acetate, gradient elution) provided 129 mg (54%) of white crystalline solid;1H NMR (400 MHz, cdcl3) δ 8.79 (s, 1H), 7.57 (s, 1H), 6.24 (d, J=4.5 Hz, 1H), 4.53 (t, J=4.6 Hz, 1H), 4.30 (t, J=4.3 Hz, 1H), 4.24 (t, J=6.7 Hz, 2H), 4.04 (q, J=4.5 Hz, 1H), 3.80-3.58 (m, 2H), 1.77-1.66 (m, 2H), 1.37 (h, J=3.9 Hz, 4H), 1.00-0.79 (m, 27H), 0.10 (d, J=2.3 Hz, 18H); LCMS m/z 701 (M+H)+. (2) Preparation of 24 The final target 24 was prepared from 40 g (115 mg, 0.164 mmoles, 1.0 eq) and 1M (in tetrahydrofuran) tetrabutylammonium fluoride (0.509 mL, 0.509 mmoles, 3.1 eq) in 7.0 mL of anhydrous tetrahydrofuran according to the procedure described for the preparation of 18 to afford residue. Purification by flash chromatography (24 g silica column, 100-95% dichloromethane in methanol, gradient elution) provided 39 mg (66%) of a white solid.1H NMR (400 MHz, dmso) δ 11.28 (brs, 1H), 8.57 (s, 1H), 6.03 (d, J=3.7 Hz, 1H), 5.31 (d, J=5.4 Hz, 1H), 5.05 (d, J=6.0 Hz, 1H), 4.67 (t, J=5.8 Hz, 1H), 4.27 (td, J=5.2, 3.7 Hz, 1H), 4.17 (t, J=6.7 Hz, 2H), 4.05 (q, J=5.6 Hz, 1H), 3.85 (td, J=5.6, 4.1 Hz, 1H), 3.59-3.38 (m, 2H), 1.65 (dd, J=9.5, 4.5 Hz, 2H), 1.41-1.27 (m, 4H), 0.97-0.82 (m, 3H); HRMS: calc for C14H22N4O7·H, 359.15613, found, 359.15647; HPLC 99.4% at 254 nm. i. Synthesis of ((2r,3s,4r,5r)-5-(5-amino-3-oxo-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl L-isoleucinate (25) i. Preparation of (2r,3r,4r,5r)-2-(acetoxymethyl)-5-(5-amino-3-oxo-1,2,4-triazin-2(3H)-yl)tetrahydrofuran-3,4-diyl diacetate (41) 6-Azacytosine (1.5 g, 13.38 mmol, 1.0 eq.) was charged in a dry flask and HMDS (100 mL) was added under argon atmosphere. After adding catalytic amount of ammonium sulfate (300 mg, 2.63 mmol, 0.2 eq.), the mixture was refluxed for 6 h under argon. The mixture turned clear and the HMDS was then evaporated carefully on the rotavap under vacuum. The system was vented to argon which reduced introduction of any moisture and resulted in a brown solid mass. The resulted silylated 6-azacytosine and commercial [(3R,4R)-3,4,5-triacetoxytetrahydrofuran-2-yl]methyl acetate (5.1 g, 16.06 mmol, 1.2 eq.) were dissolved in anhydrous acetonitrile (100 mL) under argon and cooled to 0° C. Tin(IV) chloride (1.05 g, 4.7 mL, 40.15 mmol) was carefully added and cooled mixture and reaction mixture was further stirred overnight at room temperature. The mixture was carefully quenched into saturated aqueous NaHCO3(500 mL) and saturated aqueous Na2CO3(200 mL) at 0° C. The mixture was extracted with dichloromethane (4×250 mL) and organic extracts were combined. The combined organic layer was washed with water (200 mL) and brine (100 mL). The organic layer was dried over sodium sulfate and concentrated under vacuum. The crude product was purified by column chromatography (0-30% methanol in dichloromethane) to afford desired isomer 41 (1.2 g, 24.2% yield) as a brown solid.1HNMR (DMSO-d6) δ 8.17 (s, 1H), 8.02 (s, 1H), 7.54 (d, J=1.3 Hz, 1H), 6.14 (dd, J=3.7, 1.3 Hz, 1H), 5.50 (ddd, J=5.1, 3.6, 1.3 Hz, 1H), 5.32 (td, J=5.9, 1.4 Hz, 1H), 4.30 (ddd, J=12.0, 3.6, 1.4 Hz, 1H), 4.23 (td, J=5.9, 4.5 Hz, 1H), 4.02 (ddd, J=11.9, 5.2, 1.4 Hz, 1H), 2.11-1.91 (m, 9H). ii. Preparation of 5-amino-2-((2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl)-1,2,4-triazin-3(2H)-one (6-azacytidine) (42) Intermediate 41 (1.15 g, 3.11 mmol, 1.0 eq.) was taken in 7N ammonia solution in methanol (15 mL, 105 mmol, 33 eq.) and heated at 80° C. in a steel pressure vessel for 18 h. The TLC showed all starting material being consumed. The mixture was concentrated to yield the crude residue. Recrystallization of the residue from ethanol afforded 42 (510 mg, 2.08 mmol, 67% yield) as a light orange solid.1H NMR (DMSO-d6) δ 7.97 (s, 1H), 7.86 (s, 1H), 7.48 (s, 1H), 5.95 (d, J=4.0 Hz, 1H), 5.15 (d, J=5.3 Hz, 1H), 4.94 (d, J=5.8 Hz, 1H), 4.64 (t, J=5.8 Hz, 1H), 4.18 (q, J=4.8 Hz, 1H), 3.95 (q, J=5.3 Hz, 1H), 3.74 (td, J=5.6, 4.3 Hz, 1H), 3.48 (ddd, J=11.9, 5.5, 4.3 Hz, 1H), 3.35 (dt, J=11.4, 5.9 Hz, 1H). iii. Preparation of 5-amino-2-((3ar,4r,6r,6ar)-6-(hydroxymethyl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)-1,2,4-triazin-3(2H)-one (43) To a solution of 42 (150 mg, 0.61 mmol, 1.0 eq.) and copper sulfate (500 mg, 3.13 mmol, 5.0 eq.) in 125 mL of anhydrous acetone was added 0.13 mL of concentrated sulfuric acid, and the mixture was stirred under argon at room temperature for 3 days. The mixture was filtered and the filtrate was neutralized slowly to pH 7 by addition of 1.8 mL cold 7N ammonia in methanol. Solvent was removed by evaporation under vacuum. Crude residue was purified on silica gel (0-30% methanol in dichloromethane) to afford 43 (65 mg, 37% yield) as a white solid.1H NMR (DMSO-d6) δ 8.07 (s, 1H), 7.93 (s, 1H), 7.47 (d, J=0.5 Hz, 1H), 6.09 (d, J=1.6 Hz, 1H), 4.97 (dd, J=6.2, 1.7 Hz, 1H), 4.81 (t, J=5.8 Hz, 1H), 4.74-4.65 (m, 1H), 3.98 (td, J=6.7, 2.9 Hz, 1H), 3.39 (ddd, J=6.5, 5.9, 2.8 Hz, 2H), 1.51-1.39 (m, 3H), 1.27 (d, J=0.7 Hz, 3H). iv. Preparation of ((3ar,4r,6r,6ar)-6-(5-amino-3-oxo-1,2,4-triazin-2(3H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl-(((9H-fluoren-9-yl)methoxy)carbonyl)-L-isoleucinate (44) Intermediate 43 (90 mg, 0.32 mmol, 1.0 eq.) was dissolved in anhydrous dichloromethane (5 mL) and added (2S,3S)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-3-methyl-pentanoic acid 32b (168 mg, 0.47 mmol, 1.5 eq.) followed by N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (103 mg, 0.54 mmol, 1.7 eq.) at room temperature. The reaction mixture was stirred at room temperature for 18 h. The mixture was concentrated, re-dissolved in ethyl acetate (50 mL) and washed with saturated aqueous sodium bicarbonate (10 mL) and brine (10 mL). The combined aqueous layer was again extracted with ethyl acetate (2×25 mL). The combined organic layer was dried over sodium sulfate and concentrated under vacuum. The crude product was purified by column chromatography (0-100% ethyl acetate in dichloromethane) to afford 44 (70 mg, 35.7% yield) as an off-white solid.1H NMR (DMSO-d6) δ 8.10 (s, 1H), 7.95 (s, 1H), 7.87 (d, J=7.5 Hz, 2H), 7.78-7.66 (m, 3H), 7.47 (s, 1H), 7.40 (t, J=7.5 Hz, 2H), 7.34-7.26 (m, 2H), 6.11 (s, 1H), 5.02 (d, J=6.1 Hz, 1H), 4.76 (dd, J=6.1, 2.5 Hz, 1H), 4.29-4.09 (m, 6H), 4.06-3.92 (m, 1H), 1.76 (s, 1H), 1.44 (s, 3H), 1.25 (s, 3H), 1.16 (td, J=7.1, 0.8 Hz, 1H), 0.78 (t, J=7.7 Hz, 6H). v. Preparation of ((2r,3s,4r,5r)-5-(5-amino-3-oxo-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl (((9H-fluoren-9-yl)methoxy)carbonyl)-L-isoleucinate (45) Intermediate 45 was synthesized from 44 (60 mg, 0.1 mmol) and formic acid (2 mL, 50% solution v/v in water) using the same procedure as for 34a to afford. 45 (24 mg, 42% yield) as a white solid.1H NMR (DMSO-d6) δ 8.00 (s, 1H), 7.94-7.82 (m, 3H), 7.72 (t, J=6.6 Hz, 3H), 7.47 (s, 1H), 7.39 (d, J=7.5 Hz, 2H), 7.31 (dt, J=8.8, 4.5 Hz, 2H), 6.00 (d, J=3.4 Hz, 1H), 4.36-4.11 (m, 6H), 4.07-3.89 (m, 4H), 1.79 (s, 1H), 1.36 (d, J=11.8 Hz, 1H), 1.20 (d, J=12.9 Hz, 2H), 0.89-0.73 (m, 6H). vi. Preparation of ((2r,3s,4r,5r)-5-(5-amino-3-oxo-1,2,4-triazin-2(3H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl L-isoleucinate (25) Final target 25 was prepared from 45 (16 mg, 0.03 mmol) and piperidine (1 mL of 3% v/v solution in DMF) using a similar procedure as for 9, to afford 25 (7.2 mg, 72% yield) as hygroscopic solid.1HNMR (DMSO-d6) δ 8.00 (s, 1H), 7.89 (s, 1H), 7.46 (s, 1H), 5.99 (d, J=3.4 Hz, 1H), 5.30 (d, J=5.1 Hz, 1H), 5.12 (d, J=6.0 Hz, 1H), 4.26-4.14 (m, 2H), 4.09-3.81 (m, 3H), 3.12 (d, J=5.4 Hz, 1H), 1.56 (ddt, J=9.6, 6.9, 5.0 Hz, 1H), 1.35 (ddt, J=14.9, 7.5, 3.8 Hz, 1H), 1.07 (ddt, J=14.2, 8.8, 7.3 Hz, 1H), 0.79 (dd, J=8.4, 7.0 Hz, 6H).11C NMR (DMSO-d6) δ 175.60, 159.11, 153.53, 128.09, 90.68, 81.09, 79.74, 79.61, 79.41, 79.08, 73.07, 71.02, 64.44, 59.12, 40.59, 40.39, 40.18, 39.97, 39.76, 39.55, 39.34, 24.58, 16.10, 11.85. HRMS m/z calcd. for [M+H]+C14H24N5O6358.1721, found 358.1719. HPLC purity by Method A: 92% at 254 nm 2. Characterization of Antiviral Agents A list of compounds evaluated for antiviral activity is shown in Table 1 below. TABLE 1No.Structure12345678910111213141516171819202122232425 The antiviral activity against Alpharviruses evaluated in HEK293 cells (4 hours pre-incubation with compound before infected with virus) is shown in Table 2 below. VTR=Virus titer reduction in log at 10 μM concentration. NT: not tested, ND: not determined TABLE 2WNVDENVZIKA(HEK293 cells)(HEK293 cells)(HEK 293 cells)EC90CC50EC90EC90No.(μM)(μM)VTR(μM)VTR(μM)VTR10.6403.81.92.43.22.221.3>403.06.91.316ND32.9>403.22.53.53.32.441.2403.12.02.24.82.05>30NDND>30ND>30ND610>40ND>15ND12ND74.3402.653.27.61.382.9>402.83.92.361.690.22293.51.33.21.42100.2293.90.33.40.53.111>30NDND>30ND>30ND12NT6.2NT0.53.9NTNT13NT6.7NT0.54NTNT14NT>40NT52.1NTNT150.330.974.50.163.60.4316>10>40ND>30ND>10ND17NTNTNT>30NDNTNT180.4030.82.80.204.00.102.7190.7>4040.602.11.12.620NT22.9NT1.230.22.8210.418.94.50.343.40.2422NT6.46NT13.20.42.623NT22.4NT1.33.30.3324NT20.4NT1.23.30.33.125NT>40NT5.12NTNT The antiviral activity against Alphaviruses evaluated in NHDF cells (4 hours pre-incubation with compound) is shown in Table 3 below. VTR=Virus titer reduction in log at 10 μM concentration. NT: not tested, ND: not determined. TABLE 3CHIKV (NHDF cells)VEEV (NHDF cells)EC90CC50EC90(μM)(μM)VTR(μM)VTR10.95>404.3>100.8520.4>400>1003>10>400>1004>10>4008.91.185>10>400.15>1006>10>400>1007>10>400>1008>10>400>100.2191>401.343.043.110>10>400.390.623.5111>10>400.05NTNT120.89>404.55NTNT130.51>404.3NTNT14NTNTNTNTNT150.317.962.6>10016NTNTNT10.11.0517NTNTNT51183.2>401.42>100.1319NTNTNTNTNT209ND1.5NTNT214.9ND1.7NTNT228ND0.89NTNT23>10ND0NTNT24>10ND0NTNT250.85>402.581.52.17 The antiviral activity against Influenza viruses (H1N1 and H3N2 strains) evaluated in MDCK cells (4-6 hours pre-incubation with compound) shown in Table 5 below. VTR=Virus titer reduction in log at 20 μM concentration. NT: not tested, ND: not determined. TABLE 4H1N1 strain (MDCK cells)H3N2 strain (MDCK cells)EC90CC50EC90(μM)(μM)VTR(μM)VTR1ND>1001.6ND0.62ND>1002.3ND0.93NTNTNTNTNT4ND>1003.4ND1.35ND>1000.2ND−0.16ND>1001.6ND0.27ND>1003.4ND1.68ND>1003.4ND1.690.7719.63.42.413.4103.6819.33.43.333.4110.14>203.40.783.412NTNTNTNTNT13NDND3.6ND2.8143.6>203.316.443.3150.75>203.33.293.016>100>100NDNTNT17NTNTNTNTNT180.6619.63.32.783.4193.16>203.312.13.3202.8>202.93.52.7212.8>203.63.93220.68192.93.93233.78>203.66.3224NTNT2.9NT1.625NTNTNTNTNT It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.
244,922
11858957
DETAILED DESCRIPTION Definitions Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art of the present disclosure. The following references provide one of skill with a general definition of many of the terms used in this disclosure: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, the following terms have the meanings ascribed to them below, unless specified otherwise. In some embodiments, chemical structures are disclosed with a corresponding chemical name. In case of conflict, the chemical structure controls the meaning, rather than the name. In this disclosure, “comprises,” “comprising,” “containing” and “having” and the like can have the meaning ascribed to them in U.S. Patent law and can mean “includes,” “including,” and the like; “consisting essentially of” or “consists essentially” likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited are not substantially changed by the presence of more than that which is recited, but excludes prior art embodiments. Unless specifically stated or obvious from context, as used herein, the term “or” is understood to be inclusive. Unless specifically stated or obvious from context otherwise, as used herein, the terms “a”, “an”, and “the” are understood to be singular or plural. The term “acyl” is art-recognized and refers to a group represented by the general formula hydrocarbylC(O)—, preferably alkylC(O)—. The term “acylamino” is art-recognized and refers to an amino group substituted with an acyl group and may be represented, for example, by the formula hydrocarbylC(O)NH—. The term “acyloxy” is art-recognized and refers to a group represented by the general formula hydrocarbylC(O)O—, preferably alkylC(O)O—. The term “alkoxy” refers to an alkyl group, preferably a lower alkyl group, having an oxygen attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like. The term “alkoxyalkyl” refers to an alkyl group substituted with an alkoxy group and may be represented by the general formula alkyl-O-alkyl. The term “alkenyl”, as used herein, refers to an aliphatic group containing at least one double bond and is intended to include both “unsubstituted alkenyls” and “substituted alkenyls”, the latter of which refers to alkenyl moieties having substituents replacing a hydrogen on one or more carbons of the alkenyl group. Such substituents may occur on one or more carbons that are included or not included in one or more double bonds. Moreover, such substituents include all those contemplated for alkyl groups, as discussed below, except where stability is prohibitive. For example, substitution of alkenyl groups by one or more alkyl, carbocyclyl, aryl, heterocyclyl, or heteroaryl groups is contemplated. An “alkyl” group or “alkane” is a straight chained or branched non-aromatic hydrocarbon which is completely saturated. Typically, a straight chained or branched alkyl group has from 1 to about 20 carbon atoms, preferably from 1 to about 10 unless otherwise defined. Examples of straight chained and branched alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, pentyl, hexyl, pentyl and octyl. A C1-C6straight chained or branched alkyl group is also referred to as a “lower alkyl” group. Moreover, the term “alkyl” (or “lower alkyl”) as used throughout the specification, examples, and claims is intended to include both “unsubstituted alkyls” and “substituted alkyls”, the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents, if not otherwise specified, can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxy, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. For instance, the substituents of a substituted alkyl may include substituted and unsubstituted forms of amino, azido, imino, amido, phosphoryl (including phosphonate and phosphinate), sulfonyl (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alkylthios, carbonyls (including ketones, aldehydes, carboxylates, and esters), —CF3, —CN and the like. Exemplary substituted alkyls are described below. Cycloalkyls can be further substituted with alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-substituted alkyls, —CF3, —CN, and the like. The term “Cx-y” when used in conjunction with a chemical moiety, such as, acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy is meant to include groups that contain from x to y carbons in the chain. For example, the term “Cx-yalkyl” refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from x to y carbons in the chain, including haloalkyl groups such as trifluoromethyl and 2,2,2-tirfluoroethyl, etc. C0alkyl indicates a hydrogen where the group is in a terminal position, a bond if internal. The terms “C2-yalkenyl” and “C2-yalkynyl” refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively. The term “alkylamino”, as used herein, refers to an amino group substituted with at least one alkyl group. The term “alkylthio”, as used herein, refers to a thiol group substituted with an alkyl group and may be represented by the general formula alkylS—. The term “alkynyl”, as used herein, refers to an aliphatic group containing at least one triple bond and is intended to include both “unsubstituted alkynyls” and “substituted alkynyls”, the latter of which refers to alkynyl moieties having substituents replacing a hydrogen on one or more carbons of the alkynyl group. Such substituents may occur on one or more carbons that are included or not included in one or more triple bonds. Moreover, such substituents include all those contemplated for alkyl groups, as discussed above, except where stability is prohibitive. For example, substitution of alkynyl groups by one or more alkyl, carbocyclyl, aryl, heterocyclyl, or heteroaryl groups is contemplated. The term “amide”, as used herein, refers to a group wherein each R30independently represents a hydrogen or hydrocarbyl group, or two R30are taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure. The terms “amine” and “amino” are art-recognized and refer to both unsubstituted and substituted amines and salts thereof, e.g., a moiety that can be represented by wherein each R31independently represents a hydrogen or a hydrocarbyl group, or two R31are taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure. The term “aminoalkyl”, as used herein, refers to an alkyl group substituted with an amino group. The term “aralkyl”, as used herein, refers to an alkyl group substituted with an aryl group. The term “aryl” as used herein include substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon. Preferably, the ring is a 5- to 7-membered ring, more preferably a 6-membered ring. The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like. The term “carbamate” is art-recognized and refers to a group wherein R32and R33independently represent hydrogen or a hydrocarbyl group, such as an alkyl group, or R32and R33taken together with the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure. The terms “carbocycle”, and “carbocyclic”, as used herein, refers to a saturated or unsaturated ring in which each atom of the ring is carbon. The term carbocycle includes both aromatic carbocycles and non-aromatic carbocycles. Non-aromatic carbocycles include both cycloalkane rings, in which all carbon atoms are saturated, and cycloalkene rings, which contain at least one double bond. The term “carbocycle” includes 5-7 membered monocyclic and 8-12 membered bicyclic rings. Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated and aromatic rings. Carbocycle includes bicyclic molecules in which one, two or three or more atoms are shared between the two rings. The term “fused carbocycle” refers to a bicyclic carbocycle in which each of the rings shares two adjacent atoms with the other ring. Each ring of a fused carbocycle may be selected from saturated, unsaturated and aromatic rings. In an exemplary embodiment, an aromatic ring, e.g., phenyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene. Any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits, is included in the definition of carbocyclic. Exemplary “carbocycles” include cyclopentane, cyclohexane, bicyclo[2.2.1]heptane, 1,5-cyclooctadiene, 1,2,3,4-tetrahydronaphthalene, bicyclo[4.2.0]oct-3-ene, naphthalene and adamantane. Exemplary fused carbocycles include decalin, naphthalene, 1,2,3,4-tetrahydronaphthalene, bicyclo[4.2.0]octane, 4,5,6,7-tetrahydro-1H-indene and bicyclo[4.1.0]hept-3-ene. “Carbocycles” may be substituted at any one or more positions capable of bearing a hydrogen atom. A “cycloalkyl” group is a cyclic hydrocarbon which is completely saturated. “Cycloalkyl” includes monocyclic and bicyclic rings. Typically, a monocyclic cycloalkyl group has from 3 to about 10 carbon atoms, more typically 3 to 8 carbon atoms unless otherwise defined. The second ring of a bicyclic cycloalkyl may be selected from saturated, unsaturated and aromatic rings. Cycloalkyl includes bicyclic molecules in which one, two or three or more atoms are shared between the two rings. The term “fused cycloalkyl” refers to a bicyclic cycloalkyl in which each of the rings shares two adjacent atoms with the other ring. The second ring of a fused bicyclic cycloalkyl may be selected from saturated, unsaturated and aromatic rings. A “cycloalkenyl” group is a cyclic hydrocarbon containing one or more double bonds. The term “carbocyclylalkyl”, as used herein, refers to an alkyl group substituted with a carbocycle group. The term “carbonate” is art-recognized and refers to a group —OCO2—R34, wherein R34represents a hydrocarbyl group. The term “carboxy”, as used herein, refers to a group represented by the formula —CO2H. The term “ester”, as used herein, refers to a group —C(O)OR35wherein R35represents a hydrocarbyl group. The term “ether”, as used herein, refers to a hydrocarbyl group linked through an oxygen to another hydrocarbyl group. Accordingly, an ether substituent of a hydrocarbyl group may be hydrocarbyl-O—. Ethers may be either symmetrical or unsymmetrical. Examples of ethers include, but are not limited to, heterocycle-O-heterocycle and aryl-O-heterocycle. Ethers include “alkoxyalkyl” groups, which may be represented by the general formula alkyl-O-alkyl. The terms “halo” and “halogen” as used herein means halogen and includes chloro, fluoro, bromo, and iodo. The terms “hetaralkyl” and “heteroaralkyl”, as used herein, refers to an alkyl group substituted with a hetaryl group. The term “heteroalkyl”, as used herein, refers to a saturated or unsaturated chain of carbon atoms and at least one heteroatom, wherein no two heteroatoms are adjacent. The terms “heteroaryl” and “hetaryl” include substituted or unsubstituted aromatic single ring structures, preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The terms “heteroaryl” and “hetaryl” also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like. The term “heteroatom” as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, and sulfur. The terms “heterocyclyl”, “heterocycle”, and “heterocyclic” refer to substituted or unsubstituted non-aromatic ring structures, preferably 3- to 10-membered rings, more preferably 3- to 7-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The terms “heterocyclyl” and “heterocyclic” also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Heterocyclyl groups include, for example, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, and the like. The term “heterocyclylalkyl”, as used herein, refers to an alkyl group substituted with a heterocycle group. The term “hydrocarbyl”, as used herein, refers to a group that is bonded through a carbon atom that does not have a ═O or ═S substituent, and typically has at least one carbon-hydrogen bond and a primarily carbon backbone, but may optionally include heteroatoms. Thus, groups like methyl, ethoxyethyl, 2-pyridyl, and trifluoromethyl are considered to be hydrocarbyl for the purposes of this application, but substituents such as acetyl (which has a ═O substituent on the linking carbon) and ethoxy (which is linked through oxygen, not carbon) are not. Hydrocarbyl groups include, but are not limited to aryl, heteroaryl, carbocycle, heterocyclyl, alkyl, alkenyl, alkynyl, and combinations thereof. The term “hydroxyalkyl”, as used herein, refers to an alkyl group substituted with a hydroxy group. The term “lower” when used in conjunction with a chemical moiety, such as, acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy is meant to include groups where there are ten or fewer non-hydrogen atoms in the substituent, preferably six or fewer. A “lower alkyl”, for example, refers to an alkyl group that contains ten or fewer carbon atoms, preferably six or fewer. In certain embodiments, acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy substituents defined herein are respectively lower acyl, lower acyloxy, lower alkyl, lower alkenyl, lower alkynyl, or lower alkoxy, whether they appear alone or in combination with other substituents, such as in the recitations hydroxyalkyl and aralkyl (in which case, for example, the atoms within the aryl group are not counted when counting the carbon atoms in the alkyl substituent). The terms “polycyclyl”, “polycycle”, and “polycyclic” refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls) in which two or more atoms are common to two adjoining rings, e.g., the rings are “fused rings”. Each of the rings of the polycycle can be substituted or unsubstituted. In certain embodiments, each ring of the polycycle contains from 3 to 10 atoms in the ring, preferably from 5 to 7. The term “silyl” refers to a silicon moiety with three hydrocarbyl moieties attached thereto. The term “substituted” refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxy, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as “unsubstituted,” references to chemical moieties herein are understood to include substituted variants. For example, reference to an “aryl” group or moiety implicitly includes both substituted and unsubstituted variants. The term “sulfate” is art-recognized and refers to the group —OSO3H, or a pharmaceutically acceptable salt thereof. The term “sulfonamide” is art-recognized and refers to the group represented by the general formulae wherein R36and R37independently represent hydrogen or hydrocarbyl, such as alkyl, or R36and R37taken together with the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure. The term “sulfoxide” is art-recognized and refers to the group —S(O)—R38, wherein R38represents a hydrocarbyl. The term “sulfonate” is art-recognized and refers to the group SO3H, or a pharmaceutically acceptable salt thereof. The term “sulfone” is art-recognized and refers to the group —S(O)2—R39, wherein R39represents a hydrocarbyl. The term “thioalkyl”, as used herein, refers to an alkyl group substituted with a thiol group. The term “thioester”, as used herein, refers to a group —C(O)SR40or —SC(O)R40wherein R10represents a hydrocarbyl. The term “thioether”, as used herein, is equivalent to an ether, wherein the oxygen is replaced with a sulfur. The term “urea” is art-recognized and may be represented by the general formula wherein R41and R42independently represent hydrogen or a hydrocarbyl, such as alkyl, or either occurrence of R41taken together with R42and the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure. The term “protecting group” refers to a group of atoms that, when attached to a reactive functional group in a molecule, mask, reduce or prevent the reactivity of the functional group. Typically, a protecting group may be selectively removed as desired during the course of a synthesis. Examples of protecting groups can be found in Greene and Wuts,Protective Groups in Organic Chemistry,3rdEd., 1999, John Wiley & Sons, NY and Harrison et al.,Compendium of Synthetic Organic Methods, Vols. 1-8, 1971-1996, John Wiley & Sons, NY. Representative nitrogen protecting groups include, but are not limited to, formyl, acetyl, trifluoroacetyl, benzyl, benzyloxycarbonyl (“CBZ”), tert-butoxycarbonyl (“Boc”), trimethylsilyl (“TMS”), 2-trimethylsilyl-ethanesulfonyl (“TES”), trityl and substituted trityl groups, allyloxycarbonyl, 9-fluorenylmethyloxycarbonyl (“FMOC”), nitro-veratryloxycarbonyl (“NVOC”) and the like. Representative hydroxyl protecting groups include, but are not limited to, those where the hydroxyl group is either acylated (esterified) or alkylated such as benzyl and trityl ethers, as well as alkyl ethers, tetrahydropyranyl ethers, trialkylsilyl ethers (e.g., TMS or TIPS groups), glycol ethers, such as ethylene glycol and propylene glycol derivatives and allyl ethers. In certain embodiments, compounds of the invention may be racemic. In certain embodiments, compounds of the invention may be enriched in one enantiomer. For example, a compound of the invention may have greater than about 30% ee, about 40% ee, about 50% ee, about 60% ee, about 70% ee, about 80% ee, about 90% ee, or even about 95% or greater ee. In certain embodiments, compounds of the invention may have more than one stereocenter. In certain such embodiments, compounds of the invention may be enriched in one or more diastereomer. For example, a compound of the invention may have greater than about 30% de, about 40% de, about 50% de, about 60% de, about 70% de, about 80% de, about 90% de, or even about 95% or greater de. In certain embodiments, the therapeutic preparation may be enriched to provide predominantly one enantiomer of a compound (e.g., of Formula (I)). An enantiomerically enriched mixture may comprise, for example, at least about 60 mol percent of one enantiomer, or more preferably at least about 75, about 90, about 95, or even about 99 mol percent. In certain embodiments, the compound enriched in one enantiomer is substantially free of the other enantiomer, wherein substantially free means that the substance in question makes up less than about 10%, or less than about 5%, or less than about 4%, or less than about 3%, or less than about 2%, or less than about 1% as compared to the amount of the other enantiomer, e.g., in the composition or compound mixture. For example, if a composition or compound mixture contains about 98 grams of a first enantiomer and about 2 grams of a second enantiomer, it would be said to contain about 98 mol percent of the first enantiomer and only about 2% of the second enantiomer. In certain embodiments, the therapeutic preparation may be enriched to provide predominantly one diastereomer of a compound (e.g., of Formula (I)). A diastereomerically enriched mixture may comprise, for example, at least about 60 mol percent of one diastereomer, or more preferably at least about 75, about 90, about 95, or even about 99 mol percent. The term “subject” to which administration is contemplated includes, but is not limited to, humans (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or other primates (e.g., cynomolgus monkeys, rhesus monkeys); mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, goats, cats, and/or dogs; and/or birds, including commercially relevant birds such as chickens, ducks, geese, quail, and/or turkeys. Preferred subjects are humans. As used herein, a therapeutic that “prevents” a disorder or condition refers to a compound that, in a statistical sample, reduces the occurrence of the disorder or condition in the treated sample relative to an untreated control sample, or delays the onset or reduces the severity of one or more symptoms of the disorder or condition relative to the untreated control sample. The term “treating” includes prophylactic and/or therapeutic treatments. The term “prophylactic or therapeutic” treatment is art-recognized and includes administration to the subject of one or more of the disclosed compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the subject) then the treatment is prophylactic (i.e., it protects the subject against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof). The term “prodrug” is intended to encompass compounds which, under physiologic conditions, are converted into the therapeutically active agents of the present invention (e.g., a compound of Formula (I)). A common method for making a prodrug is to include one or more selected moieties which are hydrolyzed under physiologic conditions to reveal the desired molecule. In other embodiments, the prodrug is converted by an enzymatic activity of the subject. For example, esters or carbonates (e.g., esters or carbonates of alcohols or carboxylic acids) are preferred prodrugs of the present invention. In certain embodiments, some or all of the compounds of Formula (I) in a formulation represented above can be replaced with the corresponding suitable prodrug, e.g., wherein a hydroxyl in the parent compound is presented as an ester or a carbonate or carboxylic acid. An “effective amount”, as used herein, refers to an amount that is sufficient to achieve a desired biological effect. A “therapeutically effective amount”, as used herein, refers to an amount that is sufficient to achieve a desired therapeutic effect. For example, a therapeutically effective amount can refer to an amount that is sufficient to improve at least one sign or symptom of cancer. A “response” to a method of treatment can include a decrease in or amelioration of negative symptoms, a decrease in the progression of a disease or symptoms thereof, an increase in beneficial symptoms or clinical outcomes, a lessening of side effects, stabilization of disease, partial or complete remedy of disease, among others. In some embodiments, the invention provides a compound of formula (I): or a pharmaceutically acceptable salt and/or prodrug thereof, whereinY is Het is heterocyclyl or heteroaryl;R1ais selected from H, halo, hydroxy, cyano, azido, amino, C1-6alkyl, hydroxyC1-6alkyl, amino-C1-6alkyl, —O—C(O)—O—C1-6alkyl, C1-6acyloxy, C1-6alkoxy, C2-6alkenyl, and C2-6alkynyl;R1bis selected from H, halo, C1-6alkyl, hydroxy-C1-6alkyl, amino-C1-6alkyl, C2-6alkenyl, and C2-6alkynyl;R2ais selected from halo, hydroxy, cyano, azido, amino, C1-6alkyl, hydroxy-C1-6alkyl, amino-C1-6alkyl, C1-6acyloxy, —O—C(O)—O—C1-6alkyl, C1-6alkoxy, C2-6alkenyl, and C2-6alkynyl;R2bis selected from halo, C1-6alkyl, C2-6alkenyl, and C2-6alkynyl, preferably substituted or unsubstituted C2alkynyl, most preferably unsubstituted C2alkynyl;R3is selected from H and alkyl;R4is selected from H, alkyl, CN, aryl, heteroaryl, —C(O)OR9, —C(O)NR11R12, —S(O)2R10, —P(O)(OR11)(OR12), and —P(O)(OR11)(NR13R14);R5is selected from H, cyano, alkyl, cycloalkylalkyl, heterocyclylalkyl, aralkyl, heteroaralkyl, and —C(O)OR9;R6is selected from —C(O)OR9, —C(O)NR16R17, and —P(O)(OR11)(OR12);R9is independently selected from H, alkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, aryl, aralkyl, heteroaryl, and heteroaralkyl;R10is independently selected from alkyl, alkenyl, alkynyl, amino, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, aryl, aralkyl, heteroaryl, and heteroaralkyl; andeach R11and R12is independently selected from H, alkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl; orR11and R12, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclyl;R13is H or alkyl;R14is alkyl or aralkyl;each R15is independently selected from hydroxy, alkoxy acyloxy and NR13R14;each R16and R17is independently selected from H, hydroxy, alkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl; orR16and R17, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclyl. In certain preferred embodiments of Formula I, the included compounds meet the terms of a) and b); or a) and c);wherein:a) the compound is not b) if R4and R6are each —C(O)OH and R5is benzyl substituted on the phenyl ring with a heterocyclyl or heteroaryl substituent, then the phenyl ring substituent is selected from unsubstituted or substituted pyrrolidinyl, piperazinonyl, piperidonyl, tetrahydropyrimidonyl, pyridonyl, and pyridyl; andc) if R4is —C(O)OH or tetrazolyl, R6is —C(O)OH, and R5is benzyl substituted on the phenyl ring with a second phenyl ring, then either the benzyl phenyl ring or the second phenyl ring is substituted with —C(O)OR9where R9is H or alkyl. In some embodiments, the invention provides a compound of formula (II): or a pharmaceutically acceptable salt and/or prodrug thereof, whereinHet is heterocyclyl or heteroaryl;R1ais selected from H, halo, hydroxy, cyano, azido, amino, C1-6alkyl, hydroxyC1-6alkyl, amino-C1-6alkyl, —O—C(O)—O—C1-6alkyl, C1-6acyloxy, C1-6alkoxy, C2-6alkenyl, and C2-6alkynyl;R1bis selected from H, halo, C1-6alkyl, hydroxy-C1-6alkyl, amino-C1-6alkyl, C2-6alkenyl, and C2-6alkynyl;R2ais selected from halo, hydroxy, cyano, azido, amino, C1-6alkyl, hydroxy-C1-6alkyl, amino-C1-6alkyl, C1-6acyloxy, —O—C(O)—O—C1-6alkyl, C1-6alkoxy, C2-6alkenyl, and C2-6alkynyl;R2bis selected from H, halo, C1-6alkyl, C2-6alkenyl, and C2-6alkynyl;R3is selected from H and alkyl;R4is selected from alkyl, aryl, heteroaryl, —C(O)OR9, —C(O)NR11R12, —S(O)2R10, —P(O)(OR11)(OR12), and —P(O)(OR11)(NR13R14);R5is selected from H, cyano, alkyl, cycloalkylalkyl, heterocyclylalkyl, aralkyl, heteroaralkyl, and —C(O)OR9;R6is selected from —C(O)OR9, —C(O)NR11R12and —P(O)(OR11)(OR12);R9is independently selected from H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, aryl, aralkyl, heteroaryl, and heteroaralkyl;R10is independently selected from alkyl, alkenyl, alkynyl, amino, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, aryl, aralkyl, heteroaryl, and heteroaralkyl; andeach R11and R12is independently selected from H, alkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl; orR11and R12, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclyl;R13is H or alkyl; andR14is alkyl or aralkyl; provided that a), b) and c); or a), b) and d);a) the compound is not andb) R2bis selected from halo, C2-6alkyl, C2-6alkenyl, and C2-6alkynyl, preferably substituted or unsubstituted C2alkynyl, most preferably unsubstituted C2alkynyl,c) if R4and R6are each —C(O)OH and R1is benzyl substituted on the phenyl ring with a heterocyclyl or heteroaryl substituent, then the phenyl ring substituent is selected from unsubstituted or substituted piperidonyl, tetrahydropyrimidonyl, pyridonyl, and pyridyl; andd) if R4is —C(O)OH or tetrazolyl, R6is —C(O)OH, and R5is benzyl substituted on the phenyl ring with a second phenyl ring, then either the benzyl phenyl ring or the second phenyl ring is substituted with —C(O)OR9where R9is H or alkyl. In certain preferred embodiments:a) the compound is not andb) R2bis selected from halo, C2-6alkyl, C2-6alkenyl, and C2-6alkynyl, preferably substituted or unsubstituted C2alkynyl, most preferably unsubstituted C2alkynyl; and eitherc) R5is benzyl substituted on the phenyl ring with a substituent selected from unsubstituted or substituted piperidonyl, tetrahydropyrimidonyl, pyridonyl, and pyridyl, ord) R5is benzyl substituted on the phenyl ring with a second phenyl ring substituted with —C(O)OR9where R9is H or alkyl. The following paragraphs describe various embodiments of compounds of Formula I or II, which may be combined in any combination as consistent with the formulas as defined above. In certain embodiments, R1ais H or hydroxy. In certain embodiments, R1bis H or hydroxyl. In other embodiments, R1ais hydroxy and R1bis H. In some embodiments, R2ais hydroxy or C1-6alkyl. In certain embodiments, R2bis C2-6alkyl, C2-6alkenyl or C2-6alkynyl, preferably substituted or unsubstituted C2alkynyl, such as ethynyl. In certain preferred embodiments, R2ais Me and R2bis ethynyl. In some embodiments, R2ais hydroxy and R2bis ethyl or vinyl. In other preferred embodiments, R2ais hydroxy and R2bis ethynyl. In some embodiments, R2bis propynyl, butynyl, or unsubstituted or substituted In certain preferred embodiments, R3is H. In certain embodiments, the compound of Formula (I) has the following structure: In certain embodiments, the compound of Formula (II) has the following structure: In certain such embodiments, R1ais in the α-configuration. For example, the compound of Formula (I) may have the structure (IA): Further, the compound of Formula (II) may have the structure (IIAa): In alternative embodiments, R1ais in the β-configuration. In some such embodiments, the compound of Formula (I) has the structure (IB): In some such embodiments, the compound of Formula (II) has the structure (IIBa): In further embodiments of compounds of Formula (I), e.g., as described above, R2ais in the α-configuration. For example, the compound of Formula (I) may have the structure (IC): In further preferred embodiments, the compound of Formula (II) has the structure (IICa): In alternative embodiments, R2ais in the β-configuration. In some such embodiments, the compound of Formula (I) has the structure (ID): In further preferred embodiments, the compound of Formula (II) has the structure (IIDa): In certain preferred embodiments, the compound of Formula (I) has the structure (IE): In further preferred embodiments, the compound of Formula (II) has the structure (IIEa): In particularly preferred such embodiments, R1ais hydroxy and R2ais hydroxy and R2bis selected from methyl, ethyl, vinyl, and ethynyl, most preferably ethynyl. In most preferred embodiments of the compound of Formula (IE), R1ais hydroxy, R2ais hydroxy, and R2bis ethynyl. In some preferred embodiments of the compound of Formula (IIEa), R1ais hydroxy, R2ais hydroxy, and R2bis ethynyl. In certain embodiments, Y is In certain embodiments, R4is selected from —C(O)OR9, —C(O)NR11R12, —S(O)2R10, and —P(O)(OR11)(OR12). In some embodiments, R4is —C(O)OR9and R9is H or alkyl. In other embodiments, R4is —C(O)NR11R12. In certain embodiments, each R11and R12is independently selected from H and alkyl; or R11and R12, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclyl. In other embodiments, R4is —S(O)2R10and R10is alkyl or aryl. In some embodiments, R6is —C(O)OR9and R9is H or alkyl, e.g., H or C1-6alkyl. In other embodiments, R6is —C(O)NR11R12. In certain such embodiments, each R11and R12is independently selected from H and alkyl; or R11and R12, together with the nitrogen atom to which they are attached, form a 5- to 7-membered heterocyclyl. In certain preferred embodiments, R4and R6are each —C(O)OH, most preferably wherein R5is benzyl, e.g., as discussed in greater detail below. In certain embodiments, R5is selected from H, alkyl, aralkyl and heteroaralkyl. In certain such embodiments, each alkyl, aralkyl and heteroaralkyl at R5is unsubstituted or substituted with one or more substituents selected from halo, alkyl, alkoxy, carbonyl, amino, amido, cycloalkyl, heterocyclyl, and heteroaryl. In other embodiments, the substituents on the alkyl, aralkyl and heteroaralkyl at R5are selected from halo, haloalkyl, alkoxy, carbonyl, aryl, heterocyclyl, and heteroaryl. In certain embodiments, R5is aralkyl, e.g., substituted on the aryl ring with a 5- to 7-membered heterocyclyl or a 5- to 7-membered heteroaryl. In certain particular embodiments, R5is selected from H, methyl, ethyl, —CH2-ethynyl, and —CH2-vinyl. In other embodiments, R5is selected from benzyl, —CH2-pyridyl, —CH2-pyridazinyl, —CH2-oxazolyl, —CH2-thiophenyl, —CH2-furanyl, —CH2-thiazolyl, and —CH2-benzothiazolyl, preferably benzyl and —CH2-thiophenyl. In certain preferred embodiments, R5is benzyl substituted on the phenyl ring (e.g., at a para position) with a heterocyclyl or heteroaryl substituent, preferably wherein the phenyl ring substituent is selected from substituted piperidonyl, tetrahydropyrimidonyl, pyridonyl, and pyridyl. In some embodiments, the phenyl ring substituent is piperazinonyl. In certain such embodiments, the piperidonyl, tetrahydropyrimidonyl, pyridonyl, or pyridyl is substituted with one or more of alkyl, hydroxyalkyl or alkoxyalkyl. In certain embodiments, R5is aralkyl or heteroaralkyl with a para substituent on the aryl or heteroaryl ring selected from heterocyclyl, heteroaryl, and aryl; and R2bis methyl, ethyl, or C2-6alkynyl. In certain preferred embodiments, R5is benzyl substituted on the phenyl ring (e.g., at the 4-position) with In certain embodiments, certain preferred embodiments, R5is benzyl substituted on the phenyl ring (e.g., at the 4-position) with In some embodiments, represents In some embodiments, represents In some embodiments, Y is In certain embodiments, each R15is hydroxy. In certain embodiments, Het is selected from a 6- to 10-membered aryl, a 5- to 8-membered heterocyclyl, a 5- to 8-membered monocyclic or 5- to 10-membered bicyclic heteroaryl, and may be unsubstituted or substituted with one or more substituents selected from halo, alkyl, haloalkyl, alkoxy, carbonyl, amino, amido, alkylthio, alkoxycarbonyl, cycloalkyl, aryl, heterocyclyl and heteroaryl. In some embodiments, the Het substituents are selected from halo, haloalkyl, amino, and heterocyclyl. In certain embodiments, Het is a nitrogen-containing heterocyclyl or heteroaryl, preferably attached to the core ring via a nitrogen atom of the heterocyclyl or heteroaryl ring. In some embodiments, Het is In other embodiments, Het is In other embodiments, Het is wherein Z is CH or N;Rais selected from H, halo, hydroxy, alkyl, thiophenyl, —NR7R8, aralkyl, aryl, and heteroaryl, preferably from H, Cl, —NR7R8, and phenyl;Rbis selected from halo, alkyl, haloalkyl, hydroxyalkyl, alkylthio, amido, carbonyl, amido, and heteroaryl;R7is selected from H, hydroxy, alkyl, aralkyl, heteroaralkyl, cycloalkyl, and heterocyclyl; andR8is H or alkyl; orR7and R8, together with the nitrogen atom to which they are attached, form a 4- to 7-membered heterocyclyl ring. In some embodiments, Het is In certain embodiments, Z is CH. In other embodiments, Z is N. In certain embodiments, Rais selected from H, halo, alkyl, thienyl, —NR7R8, aryl, and heteroaryl, preferably from H, Cl, —NR7R8, and phenyl. In some embodiments, Rais —NR7R8. In certain embodiments, Rbis selected from halo, alkyl, hydroxyalkyl, haloalkyl, amido, carbonyl, amido, and heteroaryl. In some embodiments, Rbis selected from Cl, —CF3, carbonyl and —CONH2. In some embodiments, Het is In some embodiments, R7is selected from H, alkyl, aralkyl, heteroaralkyl, cycloalkyl, and heterocyclyl. In certain embodiments, R7is alkyl or cycloalkyl, e.g., where the alkyl or cycloalkyl is unsubstituted or substituted with one or more substituents selected from hydroxy, alkoxy, aryl, amino, and cycloalkyl. In other embodiments, R7is aralkyl or heteroaralkyl, e.g., where the aralkyl or heteroaralkyl is unsubstituted or substituted with halo or alkyl. In some embodiments, R8is selected from H, methyl, and ethyl. In other embodiments, R7and R8, together with the nitrogen atom to which they are attached, form a heterocyclyl ring, e.g., selected from azetidinyl, morpholino, pyrrolidinyl, and azepanyl. Methods of Use Provided herein are methods of inhibiting CD73 in a cell, comprising contacting the cell with a compound of the invention, such as a compound of formula (I), or a pharmaceutically acceptable salt thereof. In certain embodiments, contacting the cell occurs in a subject in need thereof, thereby treating a disease or disorder mediated by adenosine. Also, disclosed herein are methods of treating a disease or a disorder mediated by adenosine comprising administering a compound the invention, such as a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, disclosed herein are methods of treating cancer comprising administering a compound the invention, such as a compound of Formula (I), or a pharmaceutically acceptable salt thereof. Adenosine acts on a variety of immune cells to induce immunosuppression, and the immunosuppressive effects of ectonucleotidases that enhance adenosine levels are also associated with enhanced infections of mammalian cells by parasites, fungi, bacteria, and viruses. Apart from immunosuppressive effects, adenosine also has a role in modulating the cardiovascular system (as a vasodilator and cardiac depressor), the central nervous system (CNS) (inducing sedative, anxiolytic and antiepileptic effects), the respiratory system (inducing bronchoconstriction), the kidney (having biphasic action; inducing vasoconstriction at low concentrations and vasodilation at high doses), fat cells (inhibiting lipolysis), and platelets (as an anti-aggregant). Furthermore, adenosine also promotes fibrosis (excess matrix production) in a variety of tissues. Therefore, improved treatments targeting CD73 would provide therapies for treating a wide range of conditions in addition to cancer, including cerebral and cardiac ischemic disease, fibrosis, immune and inflammatory disorders (e.g., inflammatory gut motility disorder), neurological, neurodegenerative and CNS disorders and diseases (e.g., depression, Parkinson's disease), and sleep disorders. In some embodiments, the disease or the disorder mediated by adenosine is selected from cerebral ischemic disease, cancer, cardiac ischemic disease, depression, fibrosis, an immune disorder, an inflammatory disorder (e.g., inflammatory gut motility disorder), neurological disorder or disease, neurodegenerative disorder or disease (e.g., Parkinson's disease), CNS disorders and diseases, and sleep disorders. The methods described herein are useful for the treatment of a wide variety of cancers, including bladder cancer, bone cancer, brain cancer (including glioblastoma), breast cancer, cardiac cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, fibrosarcoma, gastric cancer, gastrointestinal cancer, head & neck cancer, Kaposi's sarcoma, kidney cancer (including renal cell adenocarcinoma), leukemia, liver cancer, lung cancer (including non-small cell lung cancer, small cell lung cancer, and mucoepidermoid pulmonary carcinoma), lymphoma, melanoma, myeloma, ovarian cancer (including ovarian adenocarcinoma), pancreatic cancer, penile cancer, prostate cancer, testicular germcell cancer, thymoma and thymic carcinoma. In some embodiments, the subject has a cancer selected from breast cancer, brain cancer, colon cancer, fibrosarcoma, kidney cancer, lung cancer, melanoma, ovarian cancer, and prostate cancer. In certain embodiments, the subject has a cancer selected from breast cancer, colon cancer, fibrosarcoma, melanoma, ovarian cancer, and prostate cancer. In other embodiments, the subject has a cancer selected from brain cancer, breast cancer, kidney cancer, lung cancer, melanoma, and ovarian cancer. In some embodiments, the subject has head and neck squamous cell carcinoma, ovarian cancer, breast cancer or esophageal cancer. In other embodiments, the subject has pancreatic cancer, esophageal cancer, stomach cancer, head and neck cancer, colon cancer, lung cancer or kidney cancer. In yet other embodiments, the subject has breast cancer. In some embodiments, the breast cancer is breast adenocarcinoma. In certain embodiments, the breast cancer is triple-negative breast cancer. In certain embodiments, the methods for treating or preventing cancer can be demonstrated by one or more responses such as increased apoptosis, inhibition of tumor growth, reduction of tumor metastasis, inhibition of tumor metastasis, reduction of microvessel density, decreased neovascularization, inhibition of tumor migration, tumor regression, and increased survival of the subject. In certain embodiments, the disease or the disorder mediated by adenosine is a disease or disorder mediated by CD73 activity. In some embodiments, the compounds of the invention, such as compounds of Formula (I), are useful as inhibitors of CD73. In some embodiments, the methods described herein treat or prevent cardiovascular disease using inhibitors of CD73. Mutant genes encoding CD73 lead to extensive calcification of lower-extremity arteries and small joint capsules, which is associated with increased risk of cardiovascular disease (Hilaire et al.,N. Engl. J. Med.,364(5): 432-442, 2011). In some embodiments, the methods disclosed herein treat or prevent cancer using inhibitors of CD73. A CD73 small interfering RNA and anti-CD73 monoclonal antibodies showed a significant effect in treating or preventing cancer (Antonioli et al.,Nat. Rev. Cancer,13: 842-857, 2013). A tight correlation exists between CD73 expression and the ability of cancer cells to migrate, invade, and adhere to the extracellular matrix (ECM) (Antonioli 2013; Antonioli et al.,Trends Cancer,2(2): 95-109, 2016). In some embodiments, the treatment or prevention of cancer by inhibitors of CD73 can be demonstrated by one or more responses selected from activation, clonal expansion, and homing of tumor-specific T cells (Antonioli 2016). In other embodiments, the methods disclosed herein increase the number of effector T lymphocytes (e.g., cytolytic effector T lymphocytes). Combination Treatments In some embodiments, the method of treating or preventing cancer may comprise administering a CD39 inhibitor conjointly with one or more other chemotherapeutic agent(s). In one embodiment, the CD73 inhibitor is a compound of the invention, such as a compound of Formula (I). Other chemotherapeutic agents can include CD73-specific monoclonal antibodies which enhance the effects of other antibodies and therapies because of increased overall immune system activity (lower T-regulatory function and higher T-effector function, etc.) (Antonioli 2016). In certain embodiments, the method of treating or preventing cancer may comprise administering a compound of the invention conjointly with one or more other chemotherapeutic agent(s). Chemotherapeutic agents that may be conjointly administered with compounds of the invention include: 1-amino-4-phenylamino-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate (acid blue 25), 1-amino-4-[4-hydroxyphenyl-amino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-aminophenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[1-naphthylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-fluoro-2-carboxyphenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[2-anthracenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, ABT-263, afatinib dimaleate, axitinib, aminoglutethimide, amsacrine, anastrozole, APCP, asparaginase, AZD5363, BacillusCalmette-Guérin vaccine (beg), bicalutamide, bleomycin, bortezomib, β-methylene-ADP (AOPCP), buserelin, busulfan, cabazitaxel, cabozantinib, campothecin, capecitabine, carboplatin, carfilzomib, carmustine, ceritinib, chlorambucil, chloroquine, cisplatin, cladribine, clodronate, cobimetinib, colchicine, crizotinib, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, demethoxyviridin, dexamethasone, dichloroacetate, dienestrol, diethylstilbestrol, docetaxel, doxorubicin, epirubicin, eribulin, erlotinib, estradiol, estramustine, etoposide, everolimus, exemestane, filgrastim, fludarabine, fludrocortisone, fluorouracil, fluoxymesterone, flutamide, gefitinib, gemcitabine, genistein, goserelin, GSK1120212, hydroxyurea, idarubicin, ifosfamide, imatinib, interferon, irinotecan, ixabepilone, lenalidomide, letrozole, leucovorin, leuprolide, levamisole, lomustine, lonidamine, mechlorethamine, medroxyprogesterone, megestrol, melphalan, mercaptopurine, mesna, metformin, methotrexate, miltefosine, mitomycin, mitotane, mitoxantrone, MK-2206, mutamycin, N-(4-sulfamoylphenylcarbamothioyl) pivalamide, NF279, NF449, nilutamide, nocodazole, octreotide, olaparib, oxaliplatin, paclitaxel, pamidronate, pazopanib, pemexetred, pentostatin, perifosine, PF-04691502, plicamycin, pomalidomide, porfimer, PPADS, procarbazine, quercetin, raltitrexed, ramucirumab, reactive blue 2, rituximab, rolofylline, romidepsin, rucaparib, selumetinib, sirolimus, sodium 2,4-dinitrobenzenesulfonate, sorafenib, streptozocin, sunitinib, suramin, talazoparib, tamoxifen, temozolomide, temsirolimus, teniposide, testosterone, thalidomide, thioguanine, thiotepa, titanocene dichloride, tonapofylline, topotecan, trametinib, trastuzumab, tretinoin, veliparib, vinblastine, vincristine, vindesine, vinorelbine, and vorinostat (SAHA). In other embodiments, chemotherapeutic agents that may be conjointly administered with compounds of the invention include: ABT-263, dexamethasone, 5-fluorouracil, PF-04691502, romidepsin, and vorinostat (SAHA). In other embodiments, chemotherapeutic agents that may be conjointly administered with compounds of the invention include: 1-amino-4-phenylamino-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate (acid blue 25), 1-amino-4-[4-hydroxyphenyl-amino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-aminophenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[1-naphthylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-fluoro-2-carboxyphenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[2-anthracenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, APCP, β-methylene-ADP (AOPCP), capecitabine, cladribine, cytarabine, fludarabine, doxorubicin, gemcitabine, N-(4-sulfamoylphenylcarbamothioyl) pivalamide, NF279, NF449, PPADS, quercetin, reactive blue 2, rolofylline sodium 2,4-dinitrobenzenesulfonate, sumarin, and tonapofylline. Many combination therapies have been developed for the treatment of cancer. In certain embodiments, compounds of the invention (e.g., compounds of Formula (I)) may be conjointly administered with a combination therapy. Examples of combination therapies with which compounds of the invention may be conjointly administered are included in Table 1. TABLE 1Exemplary combinatorial therapies for the treatment of cancerNameTherapeutic agentsABVDoxorubicin, Bleomycin, VinblastineABVDDoxorubicin, Bleomycin, Vinblastine, DacarbazineAC (Breast)Doxorubicin, CyclophosphamideACDoxorubicin, Cisplatin(Sarcoma)AC (Neuro-Cyclophosphamide, Doxorubicinblastoma)ACECyclophosphamide, Doxorubicin, EtoposideACeCyclophosphamide, DoxorubicinADDoxorubicin, DacarbazineAPDoxorubicin, CisplatinARAC-DNRCytarabine, DaunorubicinB-CAVeBleomycin, Lomustine, Doxorubicin, VinblastineBCVPPCarmustine, Cyclophosphamide, Vinblastine,Procarbazine, PrednisoneBEACOPPBleomycin, Etoposide, Doxorubicin, Cyclophosphamide,Vincristine, Procarbazine, Prednisone, FilgrastimBEPBleomycin, Etoposide, CisplatinBIPBleomycin, Cisplatin, Ifosfamide, MesnaBOMPBleomycin, Vincristine, Cisplatin, MitomycinCACytarabine, AsparaginaseCABOCisplatin, Methotrexate, Bleomycin, VincristineCAFCyclophosphamide, Doxorubicin, FluorouracilCAL-GCyclophosphamide, Daunorubicin, Vincristine,Prednisone, AsparaginaseCAMPCyclophosphamide, Doxorubicin, Methotrexate,ProcarbazineCAPCyclophosphamide, Doxorubicin, CisplatinCAVCyclophosphamide, Doxorubicin, VincristineCAVE ADDCAV and EtoposideCA-VP16Cyclophosphamide, Doxorubicin, EtoposideCCCyclophosphamide, CarboplatinCDDP/Cisplatin, EtoposideVP-16CEFCyclophosphamide, Epirubicin, FluorouracilCEPP(B)Cyclophosphamide, Etoposide, Prednisone, with orwithout/BleomycinCEVCyclophosphamide, Etoposide, VincristineCFCisplatin, Fluorouracil or Carboplatin FluorouracilCHAPCyclophosphamide or Cyclophosphamide, Altretamine,Doxorubicin, CisplatinChlVPPChlorambucil, Vinblastine, Procarbazine, PrednisoneCHOPCyclophosphamide, Doxorubicin, Vincristine, PrednisoneCHOP-Add Bleomycin to CHOPBLEOCISCACyclophosphamide, Doxorubicin, CisplatinCLD-BOMPBleomycin, Cisplatin, Vincristine, MitomycinCMFMethotrexate, Fluorouracil, CyclophosphamideCMFPCyclophosphamide, Methotrexate, Fluorouracil,PrednisoneCMFVPCyclophosphamide, Methotrexate, Fluorouracil,Vincristine, PrednisoneCMVCisplatin, Methotrexate, VinblastineCNFCyclophosphamide, Mitoxantrone, FluorouracilCNOPCyclophosphamide, Mitoxantrone, Vincristine,PrednisoneCOBCisplatin, Vincristine, BleomycinCODECisplatin, Vincristine, Doxorubicin, EtoposideCOMLACyclophosphamide, Vincristine, Methotrexate,Leucovorin, CytarabineCOMPCyclophosphamide, Vincristine, Methotrexate,PrednisoneCooperCyclophosphamide, Methotrexate, Fluorouracil,RegimenVincristine, PrednisoneCOPCyclophosphamide, Vincristine, PrednisoneCOPECyclophosphamide, Vincristine, Cisplatin, EtoposideCOPPCyclophosphamide, Vincristine, Procarbazine, PrednisoneCP(ChronicChlorambucil, Prednisonelymphocyticleukemia)CP (OvarianCyclophosphamide, CisplatinCancer)CVDCisplatin, Vinblastine, DacarbazineCVICarboplatin, Etoposide, Ifosfamide, MesnaCVPCyclophosphamide, Vincristine, PrednisomeCVPPLomustine, Procarbazine, PrednisoneCYVADICCyclophosphamide, Vincristine, Doxorubicin,DacarbazineDADaunorubicin, CytarabineDATDaunorubicin, Cytarabine, ThioguanineDAVDaunorubicin, Cytarabine, EtoposideDCTDaunorubicin, Cytarabine, ThioguanineDHAPCisplatin, Cytarabine, DexamethasoneDIDoxorubicin, IfosfamideDTIC/Dacarbazine, TamoxifenTamoxifenDVPDaunorubicin, Vincristine, PrednisoneEAPEtoposide, Doxorubicin, CisplatinECEtoposide, CarboplatinEFPEtoposie, Fluorouracil, CisplatinELFEtoposide, Leucovorin, FluorouracilEMA 86Mitoxantrone, Etoposide, CytarabineEPEtoposide, CisplatinEVAEtoposide, VinblastineFACFluorouracil, Doxorubicin, CyclophosphamideFAMFluorouracil, Doxorubicin, MitomycinFAMTXMethotrexate, Leucovorin, DoxorubicinFAPFluorouracil, Doxorubicin, CisplatinF-CLFluorouracil, LeucovorinFECFluorouracil, Cyclophosphamide, EpirubicinFEDFluorouracil, Etoposide, CisplatinFLFlutamide, LeuprolideFZFlutamide, Goserelin acetate implantHDMTXMethotrexate, LeucovorinHexa-CAFAltretamine, Cyclophosphamide, Methotrexate,FluorouracilIDMTX/Methotrexate, Mercaptopurine, Leucovorin6-MPIEIfosfamide, Etoposie, MesnaIfoVPIfosfamide, Etoposide, MesnaIPAIfosfamide, Cisplatin, DoxorubicinM-2Vincristine, Carmustine, Cyclophosphamide, Prednisone,MelphalanMAC-IIIMethotrexate, Leucovorin, Dactinomycin,CyclophosphamideMACCMethotrexate, Doxorubicin, Cyclophosphamide,LomustineMACOP-BMethotrexate, Leucovorin, Doxorubicin,Cyclophosphamide, Vincristine, Bleomycin, PrednisoneMAIDMesna, Doxorubicin, Ifosfamide, Dacarbazinem-BACODBleomycin, Doxorubicin, Cyclophosphamide, Vincristine,Dexamethasone, Methotrexate, LeucovorinMBCMethotrexate, Bleomycin, CisplatinMCMitoxantrone, CytarabineMFMethotrexate, Fluorouracil, LeucovorinMICEIfosfamide, Carboplatin, Etoposide, MesnaMINEMesna, Ifosfamide, Mitoxantrone, Etoposidemini-BEAMCarmustine, Etoposide, Cytarabine, MelphalanMOBPBleomycin, Vincristine, Cisplatin, MitomycinMOPMechlorethamine, Vincristine, ProcarbazineMOPPMechlorethamine, Vincristine, Procarbazine, PrednisoneMOPP/ABVMechlorethamine, Vincristine, Procarbazine, Prednisone,Doxorubicin, Bleomycin, VinblastineMP (multipleMelphalan, Prednisonemyeloma)MP (prostateMitoxantrone, Prednisonecancer)MTX/6-MOMethotrexate, MercaptopurineMTX/6-Methotrexate, Mercaptopurine, Vincristine, PrednisoneMP/VPMTX-Methotrexate, Leucovorin, Cisplatin, DoxorubicinCDDPAdrMV (breastMitomycin, Vinblastinecancer)MV (acuteMitoxantrone, Etoposidemyelocyticleukemia)M-VACVinblastine, Doxorubicin, CisplatinMethotrexateMVPVinblastine, CisplatinMitomycinMVPPMechlorethamine, Vinblastine, Procarbazine, PrednisoneNFLMitoxantrone, Fluorouracil, LeucovorinNOVPMitoxantrone, Vinblastine, VincristineOPAVincristine, Prednisone, DoxorubicinOPPAAdd Procarbazine to OPA.PACCisplatin, DoxorubicinPAC-ICisplatin, Doxorubicin, CyclophosphamidePA-CICisplatin, DoxorubicinPCVLomustine, Procarbazine, VincristinePFLCisplatin, Fluorouracil, LeucovorinPOCPrednisone, Vincristine, LomustineProMACEPrednisone, Methotrexate, Leucovorin, Doxorubicin,Cyclophosphamide, EtoposideProMACE/Prednisone, Doxorubicin, Cyclophosphamide, Etoposide,cytaBOMCytarabine, Bleomycin, Vincristine, Methotrexate,Leucovorin, CotrimoxazolePRoMACE/Prednisone, Doxorubicin, Cyclophosphamide, Etoposide,MOPPMechlorethamine, Vincristine, Procarbazine, Methotrexate,LeucovorinPt/VMCisplatin, TeniposidePVAPrednisone, Vincristine, AsparaginasePVBCisplatin, Vinblastine, BleomycinPVDAPrednisone, Vincristine, Daunorubicin, AsparaginaseSMFStreptozocin, Mitomycin, FluorouracilTADMechlorethamine, Doxorubicin, Vinblastine, Vincristine,Bleomycin, Etoposide, PrednisoneTTTMethotrexate, Cytarabine, HydrocortisoneTopo/CTXCyclophosphamide, Topotecan, MesnaVAB-6Cyclophosphamide, Dactinomycin, Vinblastine, Cisplatin,BleomycinVACVincristine, Dactinomycin, CyclophosphamideVACAdrVincristine, Cyclophosphamide, Doxorubicin,Dactinomycin, VincristineVADVincristine, Doxorubicin, DexamethasoneVATHVinblastine, Doxorubicin, Thiotepa, FlouxymesteroneVBAPVincristine, Carmustine, Doxorubicin, PrednisoneVBCMPVincristine, Carmustine, Melphalan, Cyclophosphamide,PrednisoneVCVinorelbine, CisplatinVCAPVincristine, Cyclophosphamide, Doxorubicin, PrednisoneVDVinorelbine, DoxorubicinVelPVinblastine, Cisplatin, Ifosfamide, MesnaVIPEtoposide, Cisplatin, Ifosfamide, MesnaVMMitomycin, VinblastineVMCPVincristine, Melphalan, Cyclophosphamide, PrednisoneVPEtoposide, CisplatinV-TADEtoposide, Thioguanine, Daunorubicin, Cytarabine5 + 2Cytarabine, Daunorubicin, Mitoxantrone7 + 3Cytarabine with/, Daunorubicin or Idarubicin orMitoxantrone“8 in 1”Methylprednisolone, Vincristine, Lomustine,Procarbazine, Hydroxyurea, Cisplatin, Cytarabine,Dacarbazine In some embodiments, the chemotherapeutic agents that may be conjointly administered with compounds of the invention, such as a compound of Formula (I), include a CD39 inhibitor. CD39 or ecto-nucleoside triphosphate diphosphohydrolase 1 (E-NTPDase1 or ENTPD 1) is a membrane-bound enzyme that catalyzes the conversion of extracellular adenosine triphosphate (ATP) and/or ADP (adenosine diphosphate) to adenosine monophosphate (AMP). In one embodiment, the CD39 inhibitor is poly oxometalate-1 (POM-1). In other embodiments, the chemotherapeutic agents that may be conjointly administered with compounds of the invention, such as a compound of Formula (I), include known CD73 inhibitors. In some embodiments, the CD73 inhibitor is an anthraquinone derivative (Baqi et al.,J. Med. Chem.,53(5): 2076-2086, 2010, herein incorporated by reference). In other embodiments, the CD73 inhibitor is an sulfonic acid derivative (Raza et al.,Med. Chem.,8: 1133-1139, 2012, herein incorporated by reference). In yet other embodiments, the CD73 inhibitor is selected from 1-amino-4-phenylamino-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate (acid blue 25), 1-amino-4-[4-hydroxyphenyl-amino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-aminophenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[1-naphthylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-fluoro-2-carboxyphenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[2-anthracenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, sodium 2,4-dinitrobenzenesulfonate, N-(4-sulfamoylphenylcarbamothioyl) pivalamide, APCP, β-methylene-ADP (AOPCP), PPADS, NF279, NF449, quercetin, reactive blue 2, and sumarin (Baqi 2010; Raza 2012). In certain embodiments, the combination of a compound of the invention, such as a compound of Formula (I), with a second CD73 inhibitor or a CD39 inhibitor may have a synergistic effect in the treatment of cancer and other diseases or disorders mediated by adenosine. Without wishing to be bound by any theory, this synergy may be observed because CD39 and CD73 are often on different cell types. The hypoxic tumor microenvironment also induces greater levels of CD39 and CD73. In some embodiments, the chemotherapeutic agents that may be conjointly administered with compounds of the invention, such as a compound of Formula (I), include an adenosine receptor inhibitor. In other embodiments, the adenosine receptor inhibitor is selected from rolofylline, tonapofylline, ATL-444, istradefylline, MSX-3, preladenant, SCH-58,261, SCH-412,348, SCH-442,416, ST-1535, VER-6623, VER-6947, VER-7835, vipadenant, and ZM-241,385. In some embodiments, the adenosine receptor inhibitor targets the A2Areceptor as this subtype is predominantly expressed in most immune cells. In other embodiments, the chemotherapeutic agents that may be conjointly administered with compounds of the invention, such as a compound of Formula (I), include a nucleoside-based drug. In certain embodiments, the nucleoside-based drug is selected from gemcitabine, capecitabine, cytarabine, fludarabine and cladribine. In further embodiments, the combination therapy comprises a compound of the invention, such as a compound of Formula (I), conjointly administered with an anthracycline. In other embodiments, the combination therapy comprises a compound of the invention, such as a compound of Formula (I), conjointly administered with doxorubicin. Combination treatment with an anti-CD73 antibody and doxorubicin has demonstrated a significant chemotherapeutic effect (Young et al.,Cancer Discov.,4(8): 1-10, 2014, herein incorporated by reference). In certain embodiments, the combination therapy comprises a compound of the invention, such as a compound of Formula (I), conjointly administered with an A2Areceptor inhibitor and an anthracycline. In some embodiments, the anthracycline is doxorubicin. Combination treatment with an anti-CD73 antibody, an A2Areceptor inhibitor, and doxorubicin has demonstrated an increased chemotherapeutic effect (Antonioli 2013). In certain embodiments, the conjoint therapies of the invention comprise conjoint administration with other types of chemotherapeutic agents, such as immuno-oncology agents. Cancer cells often have specific cell surface antigens that can be recognized by the immune system. Thus, immuno-oncology agents, such as monoclonal antibodies, can selectively bind to cancer cell antigens and effect cell death. Other immuno-oncology agents can suppress tumor-mediated inhibition of the native immune response or otherwise activate the immune response and thus facilitate recognition of the tumor by the immune system. Exemplary antibody immuno-oncology agents, include, but are not limited to, abagovomab, adecatumumab, afutuzumab, alemtuzumab, anatumomab mafenatox, apolizumab, blinatumomab, BMS-936559, catumaxomab, durvalumab, epacadostat, epratuzumab, indoximod, inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab, MED14736, MPDL3280A, nivolumab, obinutuzumab, ocaratuzumab, ofatumumab, olatatumab, pembrolizumab, pidilizumab, rituximab, ticilimumab, samalizumab, and tremelimumab. In some embodiments, the antibody immune-oncology agents are selected from anti-CD73 monoclonal antibody (mAb), anti-CD39 mAb, anti-PD-1 mAb, and anti-CTLA4 mAb. Thus, in some embodiments, the methods of the invention comprise conjoint administration of one or more immuno-oncology agents, such as the agents mentioned above. In some embodiments, the combination therapy comprises a compound of the invention, such as a compound of Formula (I), conjointly administered with anti-PD-1 therapy and anti-CTLA4 therapy. Combination treatment with an anti-CD73 monoclonal antibody (mAb), anti-PD-1 mAb, and anti-CTLA4 mAb showed a significant chemotherapeutic effect (Young 2014; Antonioli 2013). In some embodiments, the combination therapy comprises conjoint administration of a compound of the invention, such as a compound of Formula (I), with anti-PD-1 therapy. In certain embodiments, the combination therapy comprises conjoint administration of a compound of the invention, such as a compound of Formula (I), with oxaliplatin. In other embodiments, the combination therapy comprises conjoint administration of a compound of the invention, such as a compound of Formula (I), with doxorubicin. In certain embodiments, a compound of the invention may be conjointly administered with non-chemical methods of cancer treatment. In certain embodiments, a compound of the invention may be conjointly administered with radiation therapy. In certain embodiments, a compound of the invention may be conjointly administered with surgery, with thermoablation, with focused ultrasound therapy, with cryotherapy, or with any combination of these. In certain embodiments, compounds of the invention may be conjointly administered with one or more other compounds of the invention. Moreover, such combinations may be conjointly administered with other therapeutic agents, such as other agents suitable for the treatment of cancer, immunological or neurological diseases, such as the agents identified above. In certain embodiments, conjointly administering one or more additional chemotherapeutic agents with a compound of the invention provides a synergistic effect. In certain embodiments, conjointly administering one or more additional chemotherapeutic agents provides an additive effect. Pharmaceutical Compositions In certain embodiments, the present invention provides a pharmaceutical preparation suitable for use in a human patient, comprising any of the compounds shown above (e.g., a compound of the invention, such as a compound of formula (I), and one or more pharmaceutically acceptable excipients. In certain embodiments, the pharmaceutical preparations may be for use in treating or preventing a condition or disease as described herein. Any of the disclosed compounds may be used in the manufacture of medicaments for the treatment of any diseases or conditions disclosed herein. The compositions and methods of the present invention may be utilized to treat a subject in need thereof. In certain embodiments, the subject is a mammal such as a human, or a non-human mammal. When administered to subject, such as a human, the composition or the compound is preferably administered as a pharmaceutical composition comprising, for example, a compound of the invention and a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are well known in the art and include, for example, aqueous solutions such as water or physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, oils such as olive oil, or injectable organic esters. In a preferred embodiment, when such pharmaceutical compositions are for human administration, particularly for invasive routes of administration (i.e., routes, such as injection or implantation, that circumvent transport or diffusion through an epithelial barrier), the aqueous solution is pyrogen-free, or substantially pyrogen-free. The excipients can be chosen, for example, to effect delayed release of an agent or to selectively target one or more cells, tissues or organs. The pharmaceutical composition can be in dosage unit form such as tablet, capsule (including sprinkle capsule and gelatin capsule), granule, lyophile for reconstitution, powder, solution, syrup, suppository, injection or the like. The composition can also be present in a transdermal delivery system, e.g., a skin patch. The composition can also be present in a solution suitable for topical administration, such as an eye drop. A pharmaceutically acceptable carrier can contain physiologically acceptable agents that act, for example, to stabilize, increase solubility or to increase the absorption of a compound such as a compound of the invention. Such physiologically acceptable agents include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients. The choice of a pharmaceutically acceptable carrier, including a physiologically acceptable agent, depends, for example, on the route of administration of the composition. The preparation or pharmaceutical composition can be a self-emulsifying drug delivery system or a self-microemulsifying drug delivery system. The pharmaceutical composition (preparation) also can be a liposome or other polymer matrix, which can have incorporated therein, for example, a compound of the invention. Liposomes, for example, which comprise phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer. The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. The phrase “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations. A pharmaceutical composition (preparation) can be administered to a subject by any of a number of routes of administration including, for example, orally (for example, drenches as in aqueous or non-aqueous solutions or suspensions, tablets, capsules (including sprinkle capsules and gelatin capsules), boluses, powders, granules, pastes for application to the tongue); absorption through the oral mucosa (e.g., sublingually); anally, rectally or vaginally (for example, as a pessary, cream or foam); parenterally (including intramuscularly, intravenously, subcutaneously or intrathecally as, for example, a sterile solution or suspension); nasally; intraperitoneally; subcutaneously; transdermally (for example as a patch applied to the skin); and topically (for example, as a cream, ointment or spray applied to the skin, or as an eye drop). The compound may also be formulated for inhalation. In certain embodiments, a compound may be simply dissolved or suspended in sterile water. Details of appropriate routes of administration and compositions suitable for same can be found in, for example, U.S. Pat. Nos. 6,110,973, 5,763,493, 5,731,000, 5,541,231, 5,427,798, 5,358,970 and 4,172,896, as well as in patents cited therein. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, the particular mode of administration. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent. Methods of preparing these formulations or compositions include the step of bringing into association an active compound, such as a compound of the invention, with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product. Formulations of the invention suitable for oral administration may be in the form of capsules (including sprinkle capsules and gelatin capsules), cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), lyophile, powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. Compositions or compounds may also be administered as a bolus, electuary or paste. To prepare solid dosage forms for oral administration (capsules (including sprinkle capsules and gelatin capsules), tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; (10) complexing agents, such as, modified and unmodified cyclodextrins; and (11) coloring agents. In the case of capsules (including sprinkle capsules and gelatin capsules), tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like. A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets, and other solid dosage forms of the pharmaceutical compositions, such as dragees, capsules (including sprinkle capsules and gelatin capsules), pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. The active ingredient can also be in microencapsulated form, if appropriate, with one or more of the above-described excipients. Liquid dosage forms useful for oral administration include pharmaceutically acceptable emulsions, lyophiles for reconstitution, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, cyclodextrins and derivatives thereof, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents. Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof. Formulations of the pharmaceutical compositions for rectal, vaginal, or urethral administration may be presented as a suppository, which may be prepared by mixing one or more active compounds with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound. Formulations of the pharmaceutical compositions for administration to the mouth may be presented as a mouthwash, or an oral spray, or an oral ointment. Alternatively or additionally, compositions can be formulated for delivery via a catheter, stent, wire, or other intraluminal device. Delivery via such devices may be especially useful for delivery to the bladder, urethra, ureter, rectum, or intestine. Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate. Dosage forms for the topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required. The ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Powders and sprays can contain, in addition to an active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the active compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel. Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. Exemplary ophthalmic formulations are described in U.S. Publication Nos. 2005/0080056, 2005/0059744, 2005/0031697 and 2005/004074 and U.S. Pat. No. 6,583,124, the contents of which are incorporated herein by reference. If desired, liquid ophthalmic formulations have properties similar to that of lacrimal fluids, aqueous humor or vitreous humor or are compatible with such fluids. A preferred route of administration is local administration (e.g., topical administration, such as eye drops, or administration via an implant). The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion. Pharmaceutical compositions suitable for parenteral administration comprise one or more active compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents. Examples of suitable aqueous and nonaqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin. In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsulated matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue. For use in the methods of this invention, active compounds can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier. Methods of introduction may also be provided by rechargeable or biodegradable devices. Various slow release polymeric devices have been developed and tested in vivo in recent years for the controlled delivery of drugs, including proteinacious biopharmaceuticals. A variety of biocompatible polymers (including hydrogels), including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of a compound at a particular target site. Actual dosage levels of the active ingredients in the pharmaceutical compositions may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of factors including the activity of the particular compound or combination of compounds employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound(s) being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound(s) employed, the age, sex, weight, condition, general health and prior medical history of the subject being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the therapeutically effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the pharmaceutical composition or compound at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. By “therapeutically effective amount” is meant the concentration of a compound that is sufficient to elicit the desired therapeutic effect. It is generally understood that the effective amount of the compound will vary according to the weight, sex, age, and medical history of the subject. Other factors which influence the effective amount may include, but are not limited to, the severity of the subject's condition, the disorder being treated, the stability of the compound, and, if desired, another type of therapeutic agent being administered with the compound of the invention. A larger total dose can be delivered by multiple administrations of the agent. Methods to determine efficacy and dosage are known to those skilled in the art (Isselbacher et al. (1996) Harrison's Principles of Internal Medicine 13 ed., 1814-1882, herein incorporated by reference). In general, a suitable daily dose of an active compound used in the compositions and methods of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. If desired, the effective daily dose of the active compound may be administered as one, two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In certain embodiments of the present invention, the active compound may be administered two or three times daily. In preferred embodiments, the active compound will be administered once daily. In certain embodiments, the dosing follows a 3+3 design. The traditional 3+3 design requires no modeling of the dose-toxicity curve beyond the classical assumption for cytotoxic drugs that toxicity increases with dose. This rule-based design proceeds with cohorts of three patients; the first cohort is treated at a starting dose that is considered to be safe based on extrapolation from animal toxicological data, and the subsequent cohorts are treated at increasing dose levels that have been fixed in advance. In some embodiments, the three doses of a compound of formula (I) range from about 100 mg to about 1000 mg orally, such as about 200 mg to about 800 mg, such as about 400 mg to about 700 mg, such as about 100 mg to about 400 mg, such as about 500 mg to about 1000 mg, and further such as about 500 mg to about 600 mg. Dosing can be three times a day when taken with without food, or twice a day when taken with food. In certain embodiments, the three doses of a compound of formula (I) range from about 400 mg to about 800 mg, such as about 400 mg to about 700 mg, such as about 500 mg to about 800 mg, and further such as about 500 mg to about 600 mg twice a day. In certain preferred embodiments, a dose of greater than about 600 mg is dosed twice a day. If none of the three patients in a cohort experiences a dose-limiting toxicity, another three patients will be treated at the next higher dose level. However, if one of the first three patients experiences a dose-limiting toxicity, three more patients will be treated at the same dose level. The dose escalation continues until at least two patients among a cohort of three to six patients experience dose-limiting toxicities (i.e., ≥about 33% of patients with a dose-limiting toxicity at that dose level). The recommended dose for phase II trials is conventionally defined as the dose level just below this toxic dose level. In certain embodiments, the dosing schedule can be about 40 mg/m2to about 100 mg/m2, such as about 50 mg/m2to about 80 mg/m2, and further such as about 70 mg/m2to about 90 mg/m2by IV for 3 weeks of a 4 week cycle. In certain embodiments, compounds of the invention may be used alone or conjointly administered with another type of therapeutic agent. As used herein, the phrase “conjoint administration” refers to any form of administration of two or more different therapeutic compounds such that the second compound is administered while the previously administered therapeutic compound is still effective in the body (e.g., the two compounds are simultaneously effective in the subject, which may include synergistic effects of the two compounds). For example, the different therapeutic compounds can be administered either in the same formulation or in a separate formulation, either concomitantly or sequentially. In certain embodiments, the different therapeutic compounds can be administered within one hour, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, or a week of one another. Thus, a subject who receives such treatment can benefit from a combined effect of different therapeutic compounds. In certain embodiments, conjoint administration of compounds of the invention with one or more additional therapeutic agent(s) (e.g., one or more additional chemotherapeutic agent(s)) provides improved efficacy relative to each individual administration of the compound of the invention (e.g., compound of formula I or Ia) or the one or more additional therapeutic agent(s). In certain such embodiments, the conjoint administration provides an additive effect, wherein an additive effect refers to the sum of each of the effects of individual administration of the compound of the invention and the one or more additional therapeutic agent(s). This invention includes the use of pharmaceutically acceptable salts of compounds of the invention in the compositions and methods of the present invention. In certain embodiments, contemplated salts of the invention include, but are not limited to, alkyl, dialkyl, trialkyl or tetra-alkyl ammonium salts. In certain embodiments, contemplated salts of the invention include, but are not limited to, L-arginine, benenthamine, benzathine, betaine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)ethanol, ethanolamine, ethylenediamine, N-methylglucamine, hydrabamine, 1H-imidazole, lithium, L-lysine, magnesium, 4-(2-hydroxyethyl)morpholine, piperazine, potassium, 1-(2-hydroxyethyl)pyrrolidine, sodium, triethanolamine, tromethamine, and zinc salts. In certain embodiments, contemplated salts of the invention include, but are not limited to, Na, Ca, K, Mg, Zn or other metal salts. The pharmaceutically acceptable acid addition salts can also exist as various solvates, such as with water, methanol, ethanol, dimethylformamide, and the like. Mixtures of such solvates can also be prepared. The source of such solvate can be from the solvent of crystallization, inherent in the solvent of preparation or crystallization, or adventitious to such solvent. Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions. Examples of pharmaceutically acceptable antioxidants include: (1) water-soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal-chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention. General Synthetic Procedures Compound numbers 1-129 as used in the general synthesis section below refer only to genus structures in this section and do not apply to compounds disclosed elsewhere in this application. Compounds disclosed herein can be made by methods depicted in the reaction schemes below. The starting materials and reagents used in preparing these compounds are either available from commercial supplier such as Aldrich Chemical Co., Bachem, etc., or can be made by methods well known in the art. The schemes are merely illustrative of some methods by which the compounds disclosed herein can be synthesized and various modifications to these schemes can be made and will be suggested to POSITA having referred to this disclosure. The starting materials and the intermediates and the final products of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography, and the like and may be characterized using conventional means, including physical constants and spectral data. Unless specified otherwise, the reactions described herein take place at atmospheric pressure over a temperature range from about −78° C. to about 150° C. General Schemes Compounds of Formula (I) having the structure: where Z, Ru, Rv, Rw, Rx, R5, and R9are analogous to variables Z, Ra, Rb, Rw, R5, R9and Rxis —CH2P(O)(OR15)2or —OP(O)(OH)CH2P(O)(OR15)2as defined in the Summary, can be synthesized as illustrated and described in Scheme 1: Ketone A-3 is prepared from commercially available diol A-1 via selective protecting the primary alcohol with a suitable group such as TBDPS, TBDMS, Ac and Bz, and followed by oxidizing the secondary alcohol in A-2 where R″ is H, alkyl, TMS, or heterocycles. Stereoselective addition of the corresponding ethynyl nucleophile A-4, such as Grignard reagents or Li reagents, to ketone A-3 to provide propargylic alcohol A-5. Removal of the acetonide protecting group is accomplished with diluted aq. acid, such as TFA, HCl, H2SO4, HClO4, PPTS, CSA or other Lewis acids. Acylation of triol A-6 with reagents, such as Ac2O, acetyl chloride, and BzCl, in the presence of a base, such as pyridine, and catalytic 4-DMAP to provide tri-ester such as ti-acetate A-7. Glycosylation under conditions (N,O-bis(trimethylsilyl)-acetamide and TMSOTf) or (TfOH and DBU) in solvent (MeCN, dichloroethane or toluene), between donor A-7 and acceptor A-8, such as 2-chloroadenine, 6-amino-2-chloroadenine, 2,6-dichloroadenine, 5,7-dichloro-1H-imidazo[4,5-b]pyridine, 5-chloro-3H-imidazo[4,5-b]pyridine, uracil, thymine, cytosine or guanine, to provide the nucleoside product A-9. In the case when Ruis NH2, it is protected as N(Boc)2with Boc2O in the presence of TEA and catalytic 4-DMAP. Removal of the protecting group in A-9, in the case of P is TBDMS or TBDPS group, treatment with TBAF to give the primary alcohol A-10 which then is undergone an insertion reaction with diazo reagent A-11 in the presence of catalyst such as Rh2(OAc)4or Cu(OAc)2to provide A-12. Alkylation with an electrophile A-13 such as alkyl halide, triflate, tosylate or mesylate in the presence of base such as Cs2CO3, K2CO3, LiHMDS, DBU or NaH, to provide A-14. The ester groups in A-14 is finally removed by base such as LiOH, NaOH, and KOH in water to provide A-15 in formula (Ia). Alternatively, the alkynyl group at the 3′-position in intermediate A-5 can be substituted with either alkyl or vinyl groups by using the corresponding alkyl or vinyl lithium and Grignard reagents at Step 3 in Scheme 1. Compounds in formula (Ia) can also be prepared according to Scheme 2. The suitable protecting group such as P is a silyl group (TBDPS or TBDMS) in precursor B-1 can be selectively removed by reagent such as TBAF or HF in THF while the P1protecting group such as Ac, Bz and MOM group remains. The resulting primary alcohol B-2 can react with diazo reagent A-11 in solvent such as benzene, toluene, DCM and dichloroethane in the presence of metal catalyst such as Rh2(OAc)4to give intermediate B-3. Alkylation of B-3 with electrophile A-13 such as halide, triflate, mesylate or sulfonate is accomplished in the presence of a base such as K2CO3, Cs2CO3, LiHMDS, NaH and DBU to give intermediate B-4. Removing the acetonide protecting group in B-4 is done by an acid treatment such as aq. TFA, HCl, H2SO4or HClO4in solvent such as DCM, acetone, THF or dioxane to provide diol B-5. Acylation of B-5 with reagent such as Ac2O or acetyl chloride in the presence of pyridine, TEA or DIPEA and catalytic 4-DMAP to give ti-acetate B-6 (for P1═OAc) as a glycosylation donor. This intermediate B-6 is reacted with a glycosylation acceptor heterocycle B-7 such as 2-chloroadenine, 6-amino-2-chloroadenine, 2,6-dichloroadenine, 5,7-dichloro-1H-imidazo[4,5-b]pyridine, 5-chloro-3H-imidazo[4,5-b]pyridine, uracil, thymine, cytosine and guanine under the influence of conditions such as [N,O-bis(trimethylsilyl)-acetamide and TMSOTf] or (TfOH and DBU) in solvent (MeCN, dichloroethane or DME) to provide nucleoside intermediate B-8. Finally removal of the ester protecting groups in B-8 with the treatment of aq. LiOH, NaOH, and KOH in solvent such as THF, dioxane, MeOH or EtOH to provide the desired final product in the formula (Ia). Compounds in formula (Ia) can also be prepared according to Scheme 3. 2,6-Dichloroadenine C-1 prepared according to Scheme 1 can proceed into several synthetic transformations. Selective nucleophilic displacement of the 6-chloro group in precursor 1 with nucleophile Ru—H (C-2) such as amines, alkoxides or thiolates in solvent such as DMF, THF, dioxane, alcohols or NMP to provide intermediate C-3. Precursor C-1 also can undergo a coupling reaction such as Suzuki, Stille or Negishi reaction with the corresponding reagent such as boronic acids (C-4), boronic esters, Tin reagents (C-5) or Zinc reagents (C-6) to provide intermediate C-7, respectively. Treatment of both intermediates C-3 and C-7 with aq. LiOH, NaOH, KOH, NaOMe, NaOEt or KOEt in solvent such as THF, dioxane, MeOH or EtOH to provide the desired final products in the formula (Ia). Compounds in formula (Ia) can also be prepared via C-1 according to Scheme 4. In this method, treatment of A-7 with 2,6-dichloropurine, TMSOTf and N,O-bis(trimethylsilyl)acetamide via the Vorbrüggen reaction gives protected nucleoside D-2. Selective removal of the tert-butyldiphenylsilyl moiety from the 5′-hydroxyl group gives alcohol D-3. Coupling with a desired substituted acyldiazo-reagent D-4 gives substituted nucleoside D-5. A wide variety of diazo reagents can be used in this reaction. Some examples include those where Rwis CO2R9, SOR9, SO2R9, P(O)(OR9)2, and CN and R9is defined as in the Summary. If an alkyl substituent R5is desired, it can be conveniently introduced using an alkylation reaction where a nucleophile such as R5—X (X=halide, OTf, OMs or OTs) is used with a base like cesium carbonate in a polar aprotic solvent like THF or DMF to give key intermediate C-1. A substituent such as an amine can be added to the purine base by displacing the chlorine at the 6-position to give intermediate D-7 with variety of amines D-6 in a solvent such as EtOH, THF or dioxane. Final deprotection of D-7 by using an aqueous hydrolysis with a base such as lithium hydroxide gives target compound in the structure of formula (1). Compounds in formula (Ia) can also be prepared according to Scheme 5. Alkylation of precursor B-3 from Scheme 2 above with an electrophile E-1 such as 4-iodobenzyl halide (Br, Cl, or I) or the corresponding OTf, OMs or OTs with base such as K2CO3, Cs2CO3, NaH or LiHMDS in solvent like DMF or THF leads to intermediate E-2 which can couple with various boronic acids such as (2-oxo-1,2-dihydropyridin-3-yl)boronic acid (E-3) illustrated here. The resulting pyridone product E-4 is alkylated with various alkyl halides (A-13) in the presence of base such as K2CO3, Cs2CO3, NaH or LiHMDS in solvent like DMF and THF to give E-5. The acetonide protecting group in E-5 is removed with the treatment of aq. TFA, HCl, H2SO4or AcOH in a solvent such as DCM, acetone, dioxane or THF to give diol E-6 which is acetylated with Ac2O or acetyl chloride with a catalytic amount of 4-DMAP and a base such as pyridine, TEA or DIPEA in solvent like DCM to provide an anomeric mixture E-7 as a glycosylation donor. Intermediate E-7 can either react with heterocyclic acceptor 2,6-dichloroadenine (D-1) or N-substituted 6-amino-2-chloroadenine (E-9) which is formed from displacing the 6-chloro group in D-1 with the suitable amines (D-7). Both glycosylation can be done under the activation conditions such as [(N,O-bis(trimethylsilyl)-acetamide and TMSOTf] or (TfOH and DBU) in solvent (MeCN, dichloroethane or toluene), between donor E-7 and acceptors D-1 or E-9 to provide the corresponding nucleoside products, E-8 or E-10 respectively. Nucleoside E-8 is converted into E-10 via a nucleophilic displacement with various amines (D-7). Finally, desired compounds in formula (Ia) is produced from E-10 via the deprotection of all its ester groups with treatment of aq. LiOH, NaOH, and KOH in a solvent such as THF, dioxane, MeOH or EtOH. Compounds in formula (Ia) can also be prepared according to Scheme 6. Alkylation with benzyl halide F-4 with various alkyl side chains (R5) and precursor B-3 in the presence of base such as K2CO3, Cs2CO3, LiHMDS or NaH in suitable solvent like DMF or THF leads to intermediate F-5. Triacetate F-7 is formed via a two-step transformation from F-5 via a deprotection (aq. TFA in DCM) and acylation (Ac2O or acetyl chloride in pyridine) as described aforementioned schemes. Glycosylation between F-7 and various acceptors such as 2,6-dichloroadenine (D-1) under an activation conditions [(N,O-bis(trimethylsilyl)-acetamide and TMSOTf] or (TfOH and DBU) in solvent (MeCN, dichloroethane or toluene) to provide nucleoside F-8 which is converted to amino analogs F-10 with various amines (F-9) in the presence of base such as pyridine, TEA or DIPEA in appropriate solvent like dioxane, DMF, or THF. Finally, desired molecules in formula (1) is obtained from F-10 with the treatment of aq. LiOH, NaOH, and KOH in a solvent such as THF, dioxane, MeOH or EtOH. Alternatively, intermediate F-10 can also directly produced from glycosylation between donor F-7 and other acceptors such as N-substituted 6-amino-2-chloroadenines (F-11). The required benzyl halides F-4 is prepared from 4-(((tert-butyldimethylsilyl)oxy)-methyl)aniline (F-1) via a five-step transformation. The cyclic urea ring is initially formed from aniline F-1 reacting with 3-chloropropyl isocyanate and followed by cyclization under the influence of a base such as NaH, NaOH or LiHMDS in solvent like DMF or THF to generate F-2. Intermediate F-2 is then led to F-3 by removal of the TBDMS group with TBAF and proceeds to the final product by converting the primary alcohol to the bromide with CBr4and PPh3in solvent such as DCM or THF. Alternatively, key intermediate F-5 is also prepared from precursor B-3 according to Scheme 7. Alkylation of precursor B-3 with halides such as 4-nitrobenzyl bromide in the presence of base such K2CO3or Cs2CO3in DMF to provide the nitro intermediate which is then reduced to the aniline with Fe in aq. NH4Cl. Cyclic urea formation is carried out from aniline with 3-chloropropyl isocyanate in the presence of base such as TEA in THF and followed by intramolecular ring closure with the treatment of base such as NaH or LiHMDS. Introduction of the N-alkyl side chains is accomplished with electrophile such as A-13 to provide key intermediate F-5. Formula (Ib) can be prepared according to Scheme 8. The required diazo reagent G-2 which Rwis an aryl or heteroaryl group, can be prepared from ester G-1 with the suitable sulfonyl azide reagent, such as 4-acetmidobenzenesulfonyl azide in the presence of a base, such as Et3N and DBU, in MeCN or dioxane. Coupling of diazo reagent G-2 and alcohol A-9 from Scheme 1 via the insertion reaction catalyzed by Rh or Cu catalyst such as Rh2(OAc)4, in a solvent such as toluene, DCM or dichloroethane to give product G-3. Alkylation of G-3 with an electrophile A-13 such as alkyl halide, triflate, tosylate or mesylate in the presence of base such as Cs2CO3, K2CO3, LiHMDS, DBU or NaH, to provide G-4. The ester groups in G-4 is finally removed by an aq. base such as LiOH, NaOH, and KOH to provide the desired product in formula (Ib). Compounds in formula (Ib) can also be prepared according to Scheme 9. The primary alcohol B-2 can react with diazo reagent G-2 from Scheme 8 in the presence of a metal catalyst such as Rh2(OAc)4in a solvent such as benzene, toluene, DCM or dichloroethane to give intermediate H-1. Alkylation of H-1 with an electrophile A-13 such as halide, triflate, mesylate or sulfonate is accomplished in the presence of base such as K2CO3, Cs2CO3, LiHMDS, NaH and DBU to give intermediate H-2. Removing the acetonide protecting group in H-2 is done by acid treatment such as aq. TFA, HCl, H2SO4, HClO4or CSA in solvent such as DCM, acetone, THF or dioxane to provide diol H-3. Acylation of H-3 with a reagent such as Ac2O or acetyl chloride in the presence of pyridine, TEA or DIPEA and catalytic 4-DMAP to give ti-acetate H-4 as a glycosylation donor. This intermediate H-4 is reacted with a glycosylation acceptor heterocycle B-7 such as 2-chloroadenine, 6-amino-2-chloroadenine, 2,6-dichloroadenine, 5,7-dichloro-1H-imidazo[4,5-b]pyridine, 5-chloro-3H-imidazo[4,5-b]pyridine, uracil, thymine, cytosine and guanine under the conditions such as [N,O-bis(trimethylsilyl)-acetamide and TMSOTf] or (TfOH and DBU) in a solvent (MeCN, dichloroethane or DME) to provide nucleoside intermediate H-5. Finally removal of the ester protecting groups in H-5 with the treatment of aq. LiOH, NaOH, and KOH in a solvent such as THF, dioxane, MeOH or EtOH to provide the desired final product in the formula (Ib). Compounds in formula (Ic) can also be prepared according to Scheme 10. Tertiary alcohol I-2 where R1is methyl (prepared according to the procedure reported by Franchetti, P. et al.J. Med. Chem.2005, 48, 4983-4989) or other alkyl groups; ethynyl (prepared according to the procedure by Hulpia, F. et al.Bioorg. Med. Chem. Lett.2016, 26, 1970-1972) or other alkynyl groups; and vinyl groups, was converted into I-4 either directly by treatment of acetylation reagent such as Ac2O and catalytic amount of H2SO4in AcOH or via a 2-step process involving deprotection of the acetonide group with treatment of aq. TFA or other acid in DCM first and followed by acetylation of the resulting diol I-3 with a reagent such as Ac2O or acetyl chloride. Glycosylation of I-4 with a heteroaromatic glycosyl acceptor such as heterocycle B-7 described in Scheme 2 where Ruis H, Cl, NH2, N-alkyl group such as 2-chloroadenine, 6-amino-2-chloroadenine, 2,6-dichloroadenine, 5,7-dichloro-1H-imidazo[4,5-b]pyridine, 5-chloro-3H-imidazo[4,5-b]pyridine, uracil, thymine, cytosine and guanine under the conditions such as [N,O-bis(trimethylsilyl)-acetamide and TMSOTf] or (TfOH and DBU) in a solvent (MeCN, dichloroethane or DME) to provide nucleoside intermediate I-5. Removal of the silyl protecting group in I-5 with a source of fluoride such as TBAF in THF to give primary alcohol I-6 which was further converted into triol I-7 with aq. LiOH or NaOH in solvent such as THF, MeOH or EtOH. Finally, treatment of I-7 with methylenebis(phosphonic dichloride) and trimethylphosphate before it is followed by triethylammonium carbonate to provide the desired final product in the formula (Ic). Compounds in formula (Ic) can be prepared according to Scheme 11. Primary alcohol I-6 (where Ru═C1) is alkylated with electrophile J-1 such as (diethoxyphosphoryl)methyl trifluoromethanesulfonate or diethyl (iodomethyl)phosphonate in the presence of a base, such as TEA, DIPEA, NaH and Cs2CO3in a solvent such as THF, DMF, dioxane or NMP to give intermediate J-2. Installation of the amino group in J-4 via nucleophilic displacement of the chloro group in J-2 with R7R8NH (J-3) where R7and R8are H or alkyl groups, in the presence of a base such as TEA or DIPEA in solvent such as dioxane, THF, or DMF. A two-step deprotection sequence (TMSBr and aq. LiOH or NaOH) is required to convert J-4 to the desired product in formula (Ic). Those having skill in the art will recognize that the starting materials and reaction conditions may be varied, the sequence of the reactions altered, and additional steps employed to produce compounds encompassed by the present invention, as demonstrated by the following examples. In some cases, protection of certain reactive functionalities may be necessary to achieve some of the above transformations. In general, the need for such protecting groups as well as the conditions necessary to attach and remove such groups will be apparent to experienced organic chemists. The disclosures of all articles and references mentioned in this application, including patents, are incorporated herein by reference. The preparation of the compounds of the present invention is illustrated further by the following examples, which are not to be construed as limiting the invention in scope or spirit to the specific procedures and compounds described in them. SYNTHETIC EXAMPLES Example 1 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: To a mixture of (3aR,5R,6aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2,2-dimethyldihydrofuro[2,3-d][1,3]dioxol-6 (3aH)-one (10 g, 23.44 mmol, 1 eq) in THF (100 mL) was added ethynylmagnesium bromide (0.5 M, 328.19 mL, 7 eq) at 15° C. under N2atmosphere. The mixture was stirred for 16 h before additional ethynylmagnesium bromide (0.5 M, 125 mL, 3 eq) was added. The mixture was stirred further for 3 h before it was diluted with saturated aq. NH4Cl (250 mL) and extracted with EtOAc (3×250 mL). The combined organic layer was washed with brine (250 mL), dried over Na2SO4, filtered and concentrated to dryness. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-4:1) to provide (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-6-ethynyl-2,2-dimethyl-tetrahydrofuro[2,3-d][1,3]dioxol-6-ol (19.47 g, 92% yield) as a yellow solid. Step 2: To a solution of (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-ol (9.47 g, 20.92 mmol, 1 eq) in DCM (100 mL) was added H2O (10 mL) and TFA (100 mL) at 0° C. The mixture was stirred at 25° C. for 1 h before it was quenched with saturated aq. NaHCO3to pH 7 and then extracted with DCM (2×300 mL). The combined organic layer was washed with brine (200 mL), dried over Na2SO4, filtered and concentrated to dryness. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-0:1) to provide (3R,4S,5R)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyltetrahydro-furan-2,3,4-triol (5.17 g, 60% yield) as a yellow gum. Step 3: To a solution of (3R,4S,5R)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyltetra-hydrofuran-2,3,4-triol (5.17 g, 12.53 mmol, 1 eq) in pyridine (50 mL) at 15° C. was added 4-DMAP (4.59 g, 37.60 mmol, 3 eq) and Ac2O (11.74 mL, 125.32 mmol, 10 eq). The mixture was stirred at 15° C. for 16 h before H2O (500 mL) was added to the mixture. The reaction mixture was extracted with EtOAc (3×200 mL). The combined organic layer was washed with brine (200 mL), dried over Na2SO4, filtered and concentrated to dryness. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-1:1) to provide (3R,4R,5R)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyltetra-hydrofuran-2,3,4-triyl triacetate (7.19 g, 79% yield) as a yellow gum. Step 4: To a solution of (3R,4R,5R)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyltetra-hydrofuran-2,3,4-triyl triacetate (6.89 g, 12.79 mmol, 1 eq) in MeCN (5 mL) at 0° C. was added 2-chloroadenine (2.39 g, 14.07 mmol, 1.1 eq), DBU (5.78 mL, 38.37 mmol, 3 eq) and TMSOTf (11.56 mL, 63.96 mmol, 5 eq). The mixture was stirred at 0° C. for 0.5 h and then stirred at 65° C. for 1 h before it was diluted with saturated aq. NaHCO3solution (500 mL). The aqueous phase was extracted with EtOAc (2×350 mL). The combined organic layer was washed with brine (350 mL), dried over Na2SO4, filtered and concentrated to dryness. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-0:1) to provide (2R,3R,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (4.52 g, 44% yield) as a yellow solid. Step 5: To a solution of (2R,3R,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (4.5 g, 6.94 mmol, 1 eq) in DMF (50 mL) at 20° C. was added TEA (4.83 mL, 34.71 mmol, 5 eq), 4-DMAP (254 mg, 2.08 mmol, 0.3 eq) and Boc2O (7.58 g, 34.71 mmol, 5 eq). The mixture was stirred at 20° C. for 1 h before H2O (250 mL) was added to the mixture. The reaction mixture was extracted with EtOAc (3×230 mL). The combined organic layer was washed with brine (250 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-1:1) to provide (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (3.26 g, 46% yield) as a yellow foam. Step 6: To a solution of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (3.24 g, 3.82 mmol, 1 eq) in THF (35 mL) at 0° C. was added TBAF (1 M, 5.73 mL, 1.5 eq). The reaction mixture was stirred at 0° C. for 1 h before it was diluted with H2O (150 mL). The reaction mixture was extracted with EtOAc (3×130 mL). The combined organic layer was washed with brine (150 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-1:2) to provide (2R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (1.51 g, 54% yield) as a yellow foam. Step 7: To a solution of (2R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (1.48 g, 2.43 mmol, 1 eq) in toluene (10 mL) at 20° C. under N2atmosphere was added Rh2(OAc)4(214 mg, 485.24 umol, 0.2 eq) and diethyl diazomalonate (903 mg, 4.85 mmol, 2 eq) in toluene (3 mL). The mixture was stirred at 95° C. for 2 h to give a green suspension before it was cooled to room temperature and concentrated to dryness. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-3:1) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (517 mg, 20% yield) as a yellow foam. Step 8: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (497.00 mg, 647.00 umol, 1 eq) in DMF (5 mL) at 25° C. was added K2CO3(178.84 mg, 1.29 mmol, 2 eq). The reaction mixture was stirred for 30 min and followed by addition of benzyl bromide (221.32 mg, 1.29 mmol, 153.69 uL, 2 eq). The mixture was stirred at 25° C. for 15.5 h before additional K2CO3(100 mg) and BnBr (100 uL) were added to the mixture. The resulting mixture was stirred at 25° C. for 24 h before H2O (50 mL) was added to the reaction. The reaction mixture was extracted with EtOAc (2×50 mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-3:1) to provide diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (266 mg, 37% yield) as a yellow foam. Step 9: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (266 mg, 309.92 umol, 1 eq) in DCM (3 mL) was added TFA (0.45 mL) at 0° C. The mixture was stirred at 25° C. for 16 h before it was treated with saturated aq. NaHCO3solution to pH 7. The reaction mixture was extracted with DCM (2×50 mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated to provide crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (195 mg) as a yellow foam. Step 10: The mixture of crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (195 mg, 296.33 umol, 1 eq) in saturated NH3in MeOH (3 mL) was stirred at 10° C. for 16 h before it was concentrated to dryness directly. The crude product was purified by preparative TLC (EtOAc) to provide diethyl 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonate (91.7 mg, 49% yield) as a yellow foam. Step 11: To a solution of diethyl 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonate (81 mg, 141.12 umol, 1 eq) in EtOH (2 mL) was added LiOH·H2O (30 mg, 705.60 umol, 5 eq) in H2O (0.2 mL) at 10° C. The mixture was stirred at 50° C. for 4 h before it was concentrated to dryness. The residue was dissolved in H2O (50 ml) and extracted with EtOAc (2×50 mL). The organic layers were discarded and the aqueous phase was acidified to pH˜2.5 with 1N aq. HCl solution. The aqueous phase was extracted with EtOAc (3×50 mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated to provide the title compound (55.4 mg, 74% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.39 (s, 1H), 7.79 (br s, 2H), 7.20 (br d, J=7.03 Hz, 2H), 7.01-7.12 (m, 3H), 5.82 (d, J=7.53 Hz, 1H), 4.87 (d, J=7.78 Hz, 1H), 4.16 (dd, J=5.27, 2.51 Hz, 1H), 3.99-4.07 (m, 2H), 3.83 (br d, J=8.03 Hz, 1H), 3.56 (s, 1H), 3.25 (dd, J=6.78 Hz, 2H); LC/MS [M+H]=518.0. Example 2 Synthesis of 2-(((2S,3R,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-4-hydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: To a solution of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (5 g, 5.89 mmol, 1 eq) was added 2M NH3in MeOH (50 mL) at 0° C. The reaction mixture was stirred at 25° C. for 16 h before it was concentrated. The crude was purified by flash silica gel column chromatography (0-50% EtOAc in petroleum ether) to provide tert-butyl (9-((2R,3R,4S,5R)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyl-3,4-dihydroxy-tetrahydrofuran-2-yl)-2-chloro-9H-purin-6-yl)carbamate (3.78 g, 90% yield) as a yellow foam. Step 2: To a solution of tert-butyl (9-((2R,3R,4S,5R)-5-(((tert-butyldiphenylsilyl)oxy)-methyl)-4-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)-2-chloro-9H-purin-6-yl)carbamate (3.78 g, 5.29 mmol, 1 eq) in DCM (40 mL) at 0° C. under N2atmosphere was added 4-DMAP (258.63 mg, 2.12 mmol, 0.4 eq) and TCDI (4.72 g, 26.46 mmol, 5 eq). The reaction mixture was stirred at 25° C. for 16 h before it was concentrated. The crude was purified by flash silica gel column chromatography (0-33% EtOAc in petroleum ether) to provide tert-butyl (9-((3aR,4R,6R,6aR)-6-(((tert-butyldiphenylsilyl)oxy)methyl)-6a-ethynyl-2-thioxotetrahydrofuro[3,4-d][1,3]dioxol-4-yl)-2-chloro-9H-purin-6-yl)carbamate (1.4 g, 37% yield) as a yellow foam. Step 3: To a solution of tert-butyl (9-((3aR,4R,6R,6aR)-6-(((tert-butyldiphenylsilyl)oxy)-methyl)-6a-ethynyl-2-thioxotetrahydrofuro[3,4-d][1,3]dioxol-4-yl)-2-chloro-9H-purin-6-yl)carbamate (500 mg, 707.93 umol, 1 eq) in toluene (5 mL) was added AIBN (11.62 mg, 70.79 umol, 0.1 eq) at 20-25° C. The reaction mixture was then heated to 60° C. and followed by addition of (n-Bu)3SnH (561.96 uL, 2.12 mmol, 3 eq). The reaction mixture was stirred at 60° C. for 1.5 h before it was concentrated. The crude was purified by flash silica gel column chromatography (0-33% EtOAc in petroleum ether) to provide tert-butyl (9-((2R,3R,4R,5S)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyl-3-hydroxytetrahydro-furan-2-yl)-2-chloro-9H-purin-6-yl)carbamate (220 mg, 47% yield) as a white foam. Step 4: To a solution of tert-butyl (9-((2R,3R,4R,5S)-5-(((tert-butyldiphenylsilyl)oxy)-methyl)-4-ethynyl-3-hydroxytetrahydrofuran-2-yl)-2-chloro-9H-purin-6-yl)carbamate (220 mg, 339.39 umol, 1 eq) in DCM (1.4 mL) at 0° C. was added TFA (0.7 mL, 9.45 mmol, 28 eq). The reaction mixture was stirred at 25° C. for 1 h before it was diluted with saturated aq. NaHCO3solution (15 mL) and extracted with DCM (3×5 mL). The combined organic layer was washed with brine (10 mL), dried over Na2SO4, filtered and concentrated to provide crude (2R,3R,4R,5S)-2-(6-amino-2-chloro-9H-purin-9-yl)-5-(((tert-butyldiphenyl-silyl)oxy)methyl)-4-ethynyltetrahydrofuran-3-ol (260 mg) as a yellow foam. Step 5: To a solution of (2R,3R,4R,5S)-2-(6-amino-2-chloro-9H-purin-9-yl)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyltetrahydrofuran-3-ol (260 mg, 474.36 umol, 1 eq, crude) in DMF (2.5 mL) was added Boc2O (931.75 mg, 4.27 mmol, 9 eq), TEA (660.25 uL, 4.74 mmol, 10 eq) and 4-DMAP (5.80 mg, 47.44 umol, 0.1 eq) at 0° C. The reaction mixture was stirred at 25° C. for 1.5 h before it was diluted with H2O (20 mL) and extracted with EtOAc (3×5 mL). The combined organic layer was washed with brine (10 mL), dried over Na2SO4, filtered and concentrated to provide crude (2R,3R,4S,5S)-2-(6-(N,N′-bis-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyltetrahydrofuran-3-yl tert-butyl carbonate (560 mg) as an orange gum. Step 6: To a solution of crude (2R,3R,4S,5S)-2-(6-(N,N′-bis-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyltetrahydrofuran-3-yl tert-butyl carbonate (560 mg, 660.02 umol, 1 eq, crude) in THF (6 mL) at 0° C. was added TBAF in THF (1 M, 1 mL, 1.52 eq). The reaction mixture was stirred at 0° C. for 1 h. The reaction mixture was diluted with H2O (10 mL) and extracted with EtOAc (3×5 mL). The combined organic layer was washed with brine (10 mL), dried over Na2SO4, filtered and concentrated. The crude was purified by preparative TLC (petroleum ether:EtOAc=2:1) to provide (2R,3R,4S,5S)-2-(6-(N,N′-bis-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-4-ethynyl-5-(hydroxymethyl)tetrahydrofuran-3-yl tert-butyl carbonate (93 mg, 45% yield over 3 steps) as a yellow foam. Step 7: To a solution of (2R,3R,4S,5S)-2-(6-(N,N′-bis-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-4-ethynyl-5-(hydroxymethyl)tetrahydrofuran-3-yl tert-butyl carbonate (93 mg, 152.45 umol, 1 eq) in toluene (1 mL) at 25° C. under N2atmosphere was added Rh2(OAc)4(6.74 mg, 15.24 umol, 0.1 eq). The reaction mixture was heated to 90° C. and followed by addition of diethyl 2-diazomalonate (85.14 mg, 457.34 umol, 3 eq) in toluene (1 mL). The reaction mixture was stirred at 90° C. for 3 h before it was concentrated. The crude residue was purified by flash column chromatography on silica gel to provide diethyl 2-(((2S,3S,4R,5R)-5-(6-(N,N′-bis-((tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyltetrahydro-furan-2-yl)methoxy)malonate (180 mg) as a gum. Step 8: To a solution of crude diethyl 2-(((2S,3S,4R,5R)-5-(6-(N,N′-bis-((tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyltetrahydro-furan-2-yl)methoxy)malonate (117.11 mg, 152.45 umol, 1 eq) in DMF (3 mL) was added K2CO3(421.39 mg, 3.05 mmol, 20 eq) at 20-25° C. The reaction mixture was stirred for 0.5 h and followed by addition of benzyl bromide (271.61 uL, 2.29 mmol, 15 eq). The reaction was then stirred further for 16 h before it was diluted with H2O (20 mL) and extracted with EtOAc (3×10 mL). The combined organic layer was washed with brine (15 mL), dried over Na2SO4, filtered and concentrated. The crude was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-2:1 gradient) to provide diethyl 2-(((2S,3S,4R,5R)-5-(6-(N,N′-bis-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-benzylmalonate (90 mg) as an off-white gum. Step 9: To a solution of diethyl 2-(((2S,3S,4R,5R)-5-(6-(N,N′-bis-((tert-butoxycarbonyl)-amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-benzylmalonate (90 mg, 104.86 umol, 1 eq) in DCM (0.6 mL) at 0° C. was added TFA (0.3 mL, 4.05 mmol, 39 eq). The reaction mixture was stirred at 25° C. for 1 h before it was diluted with saturated aq. NaHCO3solution (5 mL) and extracted with DCM (3×3 mL). The combined organic layer was washed with brine (5 mL), dried over Na2SO4, filtered and concentrated. The crude residue was purified by preparative TLC (petroleum ether:EtOAc=1:1) to provide diethyl 2-(((2S,3R,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-4-hydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonate (19 mg, 13% yield for 3 steps) as a yellow gum. Step 10: To a solution of diethyl 2-(((2S,3R,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-4-hydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonate (19 mg, 34.05 umol, 1 eq) in THF (0.2 mL) was added LiOH·H2O (7.14 mg, 170.26 umol, 5 eq) in H2O (70 uL) at 25° C. The reaction mixture was stirred for 5.5 h before it was diluted with H2O (5 mL) and then acidified to pH 2-3 with 1N aq. HCl. The mixture was extracted with EtOAc (3×5 mL). The combined organic layer was washed with brine (15 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The crude residue was dissolved in a mixture of H2O (3 mL) and MeCN (2 mL) and then lyophilized to provide the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.30 (s, 1H) 7.27 (br d, J=5.27 Hz, 2H) 7.17-7.24 (m, 1H) 7.15 (br d, J=6.78 Hz, 2H) 5.87-5.96 (m, 1H) 4.94 (br s, 1H) 4.68 (br s, 1H) 3.96-4.11 (m, 2H) 3.34-3.40 (m, 2H) 2.57 (d, J=2.51 Hz, 1H) 2.32 (s, 1H); LC/MS [M+H]=502.0. Example 3 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-(prop-1-yn-1-yl)tetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: To a solution of (3aR,5R,6aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2,2-dimethyldihydrofuro[2,3-d][1,3]dioxol-6 (3aH)-one (10 g, 23.44 mmol, 1 eq) in THF (100 mL) at 20° C. under N2atmosphere was added (prop-1-ynyl)magnesium bromide (0.5 M, 93.77 mL, 2 eq). The mixture was stirred at 40° C. for 2 h before it was diluted with saturated aq. NH4Cl solution (250 mL). The aqueous phase was extracted with EtOAc (3×200 mL). The combined organic layer was washed with brine (200 mL), dried over Na2SO4, filtered and concentrated to give crude (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-6-ol (11.81 g) as a yellow gum. Step 2: To a solution of crude (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)-methyl)-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-6-ol (12.2 g, 26.14 mmol, 1 eq) in pyridine (120 mL) at 20° C. was added 4-DMAP (3.51 g, 28.76 mmol, 1.1 eq) and Ac2O (4.90 mL, 52.29 mmol, 2 eq). The mixture was stirred at 20° C. for 16 h before it was diluted with H2O (200 mL) and extracted with EtOAc (3×100 mL). The combined organic layer was washed with brine (250 mL), dried over Na2SO4and filtered. The filtrate was concentrated to give crude (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (15 g) as a yellow gum. Step 3: To a solution of crude (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)-methyl)-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (15 g, 29.49 mmol, 1 eq) in THF (300 mL) at 0° C. under N2atmosphere was added a mixture of TBAF (1 M, 44.23 mL, 1.5 eq) and AcOH (1.26 mL, 22.12 mmol, 0.75 eq). The mixture was stirred at 20° C. for 7 h before it was diluted with saturated aq. NH4Cl solution (300 mL) and extracted with EtOAc (3×200 mL). The combined organic layer was washed with brine (300 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-1:1) to provide (3aR,5R,6R,6aR)-5-(hydroxymethyl)-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (5.78 g, 72.5% yield) as a white solid. Step 4: To a solution of (3aR,5R,6R,6aR)-5-(hydroxymethyl)-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (5.78 g, 21.39 mmol, 1 eq) in dichloroethane (60 mL) at 15° C. under N2atmosphere was added Rh2(OAc)4(945.21 mg, 2.14 mmol, 0.1 eq) and diethyl diazomalonate (7.96 g, 42.77 mmol, 2 eq). The mixture was stirred at 40° C. for 7 h before it was concentrated to dryness. The crude product was purified by flash silica gel column chromatography (0-25% EtOAc in petroleum ether) to provide diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]-dioxol-5-yl)methoxy)malonate as a yellow gum. Step 5: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (7.28 g, 16.99 mmol, 1 eq) in DMF (70 mL) at 20° C. was added Cs2CO3(11.07 g, 33.98 mmol, 2 eq) and BnBr (3.03 mL, 25.49 mmol, 1.5 eq). The mixture was stirred at 20° C. for 2 h before it was diluted with H2O (300 mL) and extracted with EtOAc (3×100 mL). The combined organic layer was washed with brine (200 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel gel column chromatography (petroleum ether:EtOAc=1:0-3:1) to provide diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-benzylmalonate (7.67 g, 87% yield) as a yellow gum. Step 6: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-benzylmalonate (7.67 g, 14.79 mmol, 1 eq) in TFA (80 mL) at 20° C. was added H2O (6.97 mL, 387.05 mmol, 26 eq). The mixture was stirred at 20° C. for 8 h before it was quenched with saturated aq. NaHCO3solution to pH 7 and partitioned with EtOAc (3×100 mL). The combined organic layer was washed with brine (200 mL), dried over Na2SO4and filtered. The filtrate was concentrated to dryness to provide crude diethyl 2-benzyl-2-(((2R,3S,4R)-3,4,5-trihydroxy-3-(prop-1-yn-1-yl)tetra-hydrofuran-2-yl)methoxy)malonate (5.95 g) as a yellow gum. Step 7: To a solution of crude diethyl 2-benzyl-2-(((2R,3S,4R)-3,4,5-trihydroxy-3-(prop-1-yn-1-yl)tetrahydrofuran-2-yl)methoxy)malonate (5.95 g, 13.63 mmol, 1 eq) in pyridine (60 mL) at 20° C. was added 4-DMAP (5.00 g, 40.90 mmol, 3 eq) and Ac2O (6.38 mL, 68.16 mmol, 5 eq). The mixture was stirred at 20° C. for 16 h before it was diluted with H2O (300 mL) and extracted with EtOAc (3×150 mL). The combined organic layer was washed with brine (300 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-3:1) to provide diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-(prop-1-yn-1-yl)tetrahydrofuran-2-yl)methoxy)malonate (5.76 g, 66% yield) as a yellow gum. Step 8: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-(prop-1-yn-1-yl)tetrahydrofuran-2-yl)methoxy)malonate (1 g, 1.78 mmol, 1 eq) in MeCN (10 mL) at 20° C. was added N,O-bis(trimethylsilyl)acetamide (BSA) (1.32 mL, 5.33 mmol, 3 eq) and 2-chloroadenine (301.43 mg, 1.78 mmol, 1 eq). The mixture was stirred at 65° C. for 30 min before it was cooled to 0° C. and followed by addition of TMSOTf (642 uL, 3.56 mmol, 2 eq) dropwise. The mixture was stirred at 0° C. for 10 min and then at 65° C. for 2 h before it was quenched with saturated aq. NaHCO3(100 mL) and extracted with EtOAc (2×60 mL). The combined organic layer was washed with brine (100 mL) and dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (0-33% EtOAc in petroleum ether) to provide diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-(prop-1-yn-1-yl)tetrahydrofuran-2-yl)methoxy)malonate (218 mg, 18% yield) as a yellow foam. Step 9: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-(prop-1-yn-1-yl)tetrahydrofuran-2-yl)methoxy)malonate (218 mg, 324.37 umol, 1 eq) in THF (2 mL) was added LiOH·H2O (136.12 mg, 3.24 mmol, 10 eq) in H2O (2 mL) at 20° C. The mixture was heated at 45° C. for 2 h before it was diluted with H2O (10 mL) and extracted with EtOAc (10 mL). The organic layer was discarded and the aqueous phase was acidified with 2 N aq. HCl to pH 2-3. Then the aqueous phase was extracted with EtOAc (3×20 mL). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by preparative HPLC to provide the title compound (39.8 mg, 23% yield) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ ppm 12.70-14.12 (m, 2H), 8.37 (s, 1H), 7.80 (br s, 2H), 7.19 (br d, J=7.03 Hz, 2H), 7.00-7.11 (m, 3H), 5.88-6.03 (m, 2H), 5.81 (d, J=7.53 Hz, 1H), 4.78 (br s, 1H), 4.12 (dd, J=4.52, 3.01 Hz, 1H), 3.95 (br dd, J=9.91, 4.89 Hz, 1H), 3.82 (br d, J=8.53 Hz, 1H), 3.25 (s, 2H), 1.81 (s, 3H); LC/MS [M+H]=532.0. Example 4 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-(cyclopropylethynyl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: To a solution of ethynylcyclopropane (4.96 g, 75.02 mmol, 6.22 mL, 2 eq) in THF (80 mL) at −78° C. under N2atmosphere was added n-BuLi (2.5 M, 30.01 mL, 2 eq) dropwise. The solution was stirred at −78° C. for 0.5 h and followed by addition of a solution of (3aR,5R,6aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2,2-dimethyldihydrofuro[2,3-d][1,3]-dioxol-6 (3aH)-one (16.0 g, 37.51 mmol, 1 eq) in THF (60 mL) dropwise. Then the solution was allowed to warm to 20° C. and stirred for 1 h before it was then cooled to 0° C. and quenched with water (120 mL). The mixture was extracted with EtOAc (2×120 mL). The combined organic layer was washed with brine (200 mL), and dried by Na2SO4, filtered and concentrated. The crude was purified by Combi-flash on silica gel (0-15% ethyl acetate in petroleum ether) to give (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)-methyl)-6-(cyclopropylethynyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-ol (15.8 g, 86% yield) as a syrup. Step 2: To a solution of (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-6-(cyclo-propylethynyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-ol (15.8 g, 32.07 mmol, 1 eq) in pyridine (160 mL) at 20° C. was added 4-DMAP (4.70 g, 38.48 mmol, 1.2 eq) and Ac2O (9.01 mL, 96.21 mmol, 3 eq). The solution was stirred for 3 h before it was diluted with water (200 mL) and extracted with ethyl acetate (2×200 mL). The combined organic layer was washed with brine (400 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by Combi-flash on silica gel (0-15% ethyl acetate in petroleum ether) to give (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-6-(cyclopropylethynyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (14.7 g, 86% yield) as a clear syrup. Step 3: To a solution of (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-6-(cyclopropylethynyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (14.7 g, 27.49 mmol, 1 eq) in THF (150 mL) at 0° C. was added a solution of TBAF (1 M, 41.24 mL, 1.5 eq) and AcOH (1.18 mL, 20.62 mmol, 0.75 eq). The solution was stirred at 20° C. for 16 h before it was diluted with water (300 mL) and extracted with ethyl acetate (2×200 mL). The combined organic layer was washed with water (400 mL), brine (400 mL), dried by Na2SO4, filtered and concentrated. The crude was purified by Combi-flash on silica gel (20-60% ethyl acetate in petroleum ether) to give (3aR,5R,6R,6aR)-6-(cyclopropylethynyl)-5-(hydroxymethyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (8.15 g, 100% yield) as a white solid. Step 4: To a solution of (3aR,5R,6R,6aR)-6-(cyclopropylethynyl)-5-(hydroxymethyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (8.15 g, 27.50 mmol, 1 eq) in dichloroethane (80 mL) at 20° C. under N2atmosphere was added Rh2(OAc)4(1.00 g, 2.26 mmol, 0.08 eq) and a solution of diethyl diazomalonate (10.24 g, 55.01 mmol, 2 eq) in dichloroethane (20 mL). The green solution was stirred for 16 h before it was concentrated. The crude was purified by Combi-flash on silica gel (15-50% ethyl acetate in petroleum ether) to give diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-(cyclopropylethynyl)-2,2-dimethyl-tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (9.52 g, 76% yield) as a yellow oil. Step 5: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-(cyclopropylethynyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (4.50 g, 9.90 mmol, 1 eq) in DMF (50 mL) at 20° C. was added Cs2CO3(9.68 g, 29.71 mmol, 3 eq) and benzyl bromide (1.76 mL, 14.85 mmol, 1.5 eq). The suspension was stirred for 16 h before it was diluted with water (80 mL) and extracted with ethyl acetate (3×80 mL). The combined organic layer was washed with water (200 mL), brine (200 mL), dried by Na2SO4, filtered and concentrated. The crude was purified by Combi-flash on silica gel (15-50% ethyl acetate in petroleum ether) to give diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-(cyclo-propylethynyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-benzyl-malonate (4.10 g, 76% yield) as a colorless syrup. Step 6: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-(cyclopropylethynyl)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-benzylmalonate (4.10 g, 7.53 mmol, 1 eq) in DCM (50 mL) at 0° C. was added H2O (10 mL) and TFA (50 mL). The solution was stirred at 20° C. for 2 h before it was quenched with saturated aq. NaHCO3(80 mL) to pH˜7. The reaction mixture was exacted with DCM (100 mL). The organic layer was washed with brine (10 mL), dried by Na2SO4, filtered and concentrated to give crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-(cyclopropylethynyl)-4,5-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonate (3.80 g) as a yellow gum. Step 7: To a solution of crude diethyl diethyl 2-(((2R,3S,4R)-3-acetoxy-3-(cyclopropyl-ethynyl)-4,5-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonate (3.80 g, 7.53 mmol, 1 eq) in pyridine (40 mL) at 20° C. was added 4-DMAP (2.76 g, 22.60 mmol, 3 eq) and Ac2O (5.64 mL, 60.25 mmol, 8 eq). The solution was stirred for 16 h before it was diluted with water (80 mL) and extracted with ethyl acetate (2×80 mL). The combined organic layer was washed with water (150 mL), brine (150 mL), dried by Na2SO4, filtered and concentrated. The crude was purified by Combi-flash on silica gel (10-50% ethyl acetate in petroleum ether) to give diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-(cyclopropylethynyl)-tetrahydro-furan-2-yl)methoxy)malonate (2.91 g, 66% yield) as a yellow gum. Step 8: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-(cyclopropylethynyl)tetrahydrofuran-2-yl)methoxy)malonate (980 mg, 1.66 mmol, 1 eq) in MeCN (24 mL) at 25° C. was added 2-chloroadenine (338.80 mg, 2.00 mmol, 1.2 eq) and BSA (987.71 uL, 4.00 mmol, 2.4 eq). The suspension was stirred at 65° C. for 0.5 h as it turned clear. The resulting solution was cooled down to 0° C. and followed by addition of TMSOTf (444.06 mg, 2.00 mmol, 361.03 uL, 1.2 eq) dropwise. The reaction mixture was stirred at 40° C. for 4 h before it was allowed to cool to room temperature. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (3×20 mL). The combined organic layer was washed with brine (50 mL), dried by Na2SO4, filtered and concentrated. The crude was purified by Combi-flash on silica gel (30-80% ethyl acetate in petroleum ether) to give diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-(cyclopropylethynyl)tetrahydrofuran-2-yl)methoxy)malonate (270 mg, 23% yield) as a yellow gum. Step 9: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-(cyclopropylethynyl)tetrahydrofuran-2-yl)-methoxy)malonate (270 mg, 386.75 umol, 1 eq) in THF (8 mL) was added aq. LiOH solution (1 M, 5.80 mL, 15 eq). The mixture was stirred at 20° C. for 16 h before it was treated with 1N HCl to adjust the pH to 5. The mixture was concentrated. The crude residue was purified by preparative HPLC to give the title compound (23 mg, 11% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.04 (s, 1H) 7.14-7.27 (m, 2H) 7.01-7.08 (m, 3H) 5.92 (d, J=6.63 Hz, 1H) 4.70-4.83 (m, 1H) 4.24 (t, J=3.50 Hz, 1H) 4.02 (t, J=3.31 Hz, 2H) 3.31-3.45 (m, 2H) 1.25-1.33 (m, 1H) 0.72-0.79 (m, 2H) 0.63-0.71 (m, 2H); LC/MS [M+H]=559.0. Example 5 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-((2-chloropyridin-4-yl)methyl)malonic acid Proceeding as described in Example 1 above by substituting BnBr with 2-chloro-4-(chloromethyl)pyridine provided the title compound as a white solid. 1H NMR (CD3OD, 400 MHz) δ ppm 8.40 (s, 1H), 8.00 (d, J=5.13 Hz, 1H), 7.36 (s, 1H), 7.23 (d, J=5.13 Hz, 1H), 6.01 (d, J=7.63 Hz, 1H), 5.08 (d, J=7.63 Hz, 1H), 4.39 (dd, J=4.88, 2.75 Hz, 1H), 4.16 (dd, J=10.07, 5.19 Hz, 1H), 4.05 (dd, J=10.01, 2.63 Hz, 1H), 3.47 (s, 2H), 3.05 (s, 1H); LC/MS [M+H]=553.1. Example 6 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2,4-dioxo-3,4-dihydropyrimidin-1 (2H)-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a mixture of (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-yl acetate (27.4 g, 55.39 mmol, 1 eq) in THF (250 mL) at 0° C. was added AcOH (2.38 mL, 41.54 mmol, 0.75 eq) in TBAF (1 M, 83.09 mL, 1.5 eq). The mixture was stirred at 15° C. for 15 h before it was partitioned between water (800 mL) and EtOAc (300 mL). The aqueous phase was further extracted with EtOAc (3×200 mL). The combined organic layer was washed with brine (300 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography on SiO2(11-33% EtOAc in petroleum ether) to give (3aR,5R,6R,6aR)-6-ethynyl-5-(hydroxymethyl)-2,2-dimethyltetrahydrofuro-[2,3-d][1,3]dioxol-6-yl acetate (15.2 g, 91% yield) as alight yellow solid. Step 2: To a mixture of (3aR,5R,6R,6aR)-6-ethynyl-5-(hydroxymethyl)-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-6-yl acetate (15.2 g, 59.32 mmol, 1 eq) in dichloroethane (150 mL) at 0° C. was added Rh2(OAc)4(1.31 g, 2.97 mmol, 0.05 eq) and diethyl diazomalonate (13.25 g, 71.18 mmol, 1.2 eq) in dichloroethane (30 mL). The mixture was stirred at 15° C. under N2atmosphere for 15 h before additional amount of diethyl diazomalonate (6 g) in dichloroethane (15 mL) was added. The mixture was stirred further at 15° C. for 2 h before it was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (11-33% EtOAc in petroleum ether) to give diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro-[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (15 g, 61% yield) as a white solid. Step 3: To a mixture of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (14 g, 33.78 mmol, 1 eq) in DMF (140 mL) at 25° C. was added Cs2CO3(22.01 g, 67.57 mmol, 2 eq) and BnBr (6.02 mL, 50.68 mmol, 1.5 eq). The mixture was stirred at 25° C. for 3 h before it was filtered and the filter cake was washed with EtOAc (50 mL). The filtrate was diluted with water (400 mL) and extracted with EtOAc (3×150 mL). The combined organic layer was washed with brine (200 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel column chromatography (0-33% EtOAc in petroleum ether) to give diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-benzylmalonate (13.3 g, 78% yield) as a colorless oil. Step 4: To a mixture of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-benzylmalonate (13.20 g, 26.16 mmol, 1 eq) in DCM (100 mL) and H2O (20 mL, 1.11 mol, 42 eq) was added TFA (100 mL, 1.35 mol, 52 eq). The mixture was stirred at 15° C. for 12 h before water (200 mL) was added. The aqueous phase was extracted with DCM (2×100 mL). The combined organic extract was washed with saturated aq. NaHCO3(2×100 mL), brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography (SiO2, petroleum ether:Ethyl acetate=10:1 to 1:3) to give diethyl 2-benzyl-2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxytetrahydrofuran-2-yl)methoxy)malonate (7.8 g, 71% yield) as a colorless oil. Step 5: To a mixture of diethyl 2-benzyl-2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxytetra-hydrofuran-2-yl)methoxy)malonate (7.8 g, 18.46 mmol, 1 eq) in pyridine (70 mL) at 15° C. was added 4-DMAP (6.77 g, 55.39 mmol, 3 eq) and Ac2O (17.29 mL, 184.65 mmol, 10 eq). The mixture was stirred for 15 h before water (400 mL) was added. The mixture was extracted with EtOAc (3×200 mL). The combined organic layer was washed with 1N aq. HCl (2×200 mL), saturated aq. NaHCO3(300 mL), brine (300 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel column chromatography (petroleum ether:Ethyl acetate=1:0 to 2:1) to give the desired product (7 g). This product was triturated with EtOH (10 mL) and filtered to give diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (3.79 g, 37% yield) as a white solid. The filtrate was concentrated under reduced pressure to give slightly impure additional product (3 g). Step 6: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetra-hydrofuran-2-yl)methoxy)malonate (116 mg, 0.21 mmol, 1.0 eq) in MeCN (3 mL) at 25° C. was added uracil (28 mg, 0.25 mmol, 1.2 eq) and followed by N,O-bis(trimethylsilyl)-acetamide (BSA) (124 uL, 0.51 mmol, 2.4 eq). The resulting suspension was heated at 65° C. for 30 min as it became clear. The reaction mixture was cooled to 0° C. and followed by dropwise addition of TMSOTf (46 uL, 0.25 mmol, 1.2 eq). The reaction mixture was allowed to warm up and heated at 65° C. for 3 h as all of the starting material was consumed. The reaction was quenched with cold saturated aq. NaHCO3solution (3 mL) and diluted with EtOAc (15 mL). The organic layer was separated, washed with H2O (2×10 mL), brine, dried (MgSO4), filtered and concentrated. The crude residue was purified by flash silica gel column chromatography (0-75% EtOAc in hexanes) to provide the product diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,4-dioxo-3,4-dihydro-pyrimidin-1 (2H)-yl)-3-ethynyl-tetrahydrofuran-2-yl)methoxy)malonate (104 mg, 82% yield). Step 7: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,4-dioxo-3,4-dihydropyrimidin-1 (2H)-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (100 mg, 0.166 umol, 1 eq) in a mixture of THF (1 mL) and MeOH (2 mL) was added aq. LiOH solution (1 M, 3 mL). The mixture was stirred at 40° C. for 24 h before the organic volatile was removed under reduced pressure. The residue was diluted with water (2 mL) and treated with TN HCl to adjust the pH to 4. The mixture was extracted with EtOAc (3×10 mL). The combined organic layer was washed with brine, dried (MgSO4), filtered and concentrated to provide the title compound (67 mg) as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.09 (bs, 1H), 7.86 (d, J=8 Hz, 1H), 7.33-7.16 (m, 5H), 6.01 (d, J=7 Hz, 1H), 5.07 (d, J=8 Hz, 1H), 4.45 (d, J=7 Hz, 1H), 4.22 (bs, 1H), 4.11-3.94 (m, 2H), 3.55-3.31 (m, 2H), 2.95 (s, 1H), LC/MS [M+H]=461.0. Example 7 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(pyridin-4-ylmethyl)malonic acid Step 1: To a mixture of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (1.2 g, 2.90 mmol, 1 eq) in DMF (20 mL) at 20° C. was added Cs2CO3(6.60 g, 20.27 mmol, 7 eq) and 4-(chloromethyl) pyridine hydrochloride (1.90 g, 11.58 mmol, 4 eq). The mixture was stirred for 2 h before it was filtered and the filter cake was washed with EtOAc (20 mL). The filtrate was diluted with water (60 mL) and extracted with EtOAc (3×50 mL). The combined extract was washed with water (2×50 mL), saturated aq. NH4Cl (50 mL), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on SiO2(14-33% EtOAc in petroleum ether) to give diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]-dioxol-5-yl)methoxy)-2-(pyridin-4-ylmethyl)malonate (900 mg, 61% yield) as a yellow oil. Step 2: To a mixture of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(pyridin-4-ylmethyl)-malonate (900 mg, 1.78 mmol, 1 eq) in DCM (5 mL) and H2O (1 mL, 55.51 mmol, 31.18 eq) was added TFA (5 mL, 67.53 mmol, 37.93 eq). The mixture was stirred at 20° C. for 12 h before it was concentrated under reduced pressure. The crude residue was azeotroped with DCM (3×10 mL) under reduced pressure to provide crude diethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxytetra-hydrofuran-2-yl)methoxy)-2-(pyridin-4-ylmethyl)malonate (1.1 g) as a brown oil. Step 3: To a mixture of crude diethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxy-tetrahydro-furan-2-yl)methoxy)-2-(pyridin-4-ylmethyl)malonate (1.1 g, 2.60 mmol, 1 eq) in pyridine (8 mL) at 20° C. was added 4-DMAP (952.17 mg, 7.79 mmol, 3 eq) and Ac2O (2.43 mL, 25.98 mmol, 10 eq). The mixture was stirred for 12 h before it was partitioned between water (30 mL) and EtOAc (20 mL). The aqueous phase was further extracted with EtOAc (2×20 mL). The combined extract was washed with water (20 mL), 0.5 N aq. HCl (2×10 mL), and brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on SiO2(25-50% EtOAc in petroleum ether) to give diethyl 2-(pyridin-4-ylmethyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyl-tetrahydrofuran-2-yl)methoxy)malonate (640 mg, 45% yield) as a brown syrup. Step 4: To a mixture of diethyl 2-(pyridin-4-ylmethyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (40 mg, 72.79 umol, 1 eq) and 2-chloroadenine (13.58 mg, 80.07 umol, 1.1 eq) in MeCN (1.5 mL) was added BSA (44.98 uL, 181.98 umol, 2.5 eq) at 25° C. under N2atmosphere. The mixture was stirred at 65° C. for 0.5 h before it was cooled to 0° C. and followed by dropwise addition of TMSOTf (26.31 uL, 145.58 umol, 2 eq). The mixture was stirred at 0° C. for 0.5 h and then at 65° C. for 2 h. The reaction mixture was cooled to room temperature and quenched with saturated aq. NaHCO3(6 mL). The mixture was extracted with EtOAc (3×8 mL). The combined organic layer was washed with brine (5 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (EtOAc) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(pyridin-4-ylmethyl)-malonate (13 mg, 26% yield) was obtained as a yellow gum. Step 5: To a mixture of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(pyridin-4-ylmethyl)-malonate (50 mg, 75.87 umol, 1 eq) in THF (1 mL) was added 1N aq. LiOH (1 mL). The mixture was stirred at 50° C. for 1 h before it was cooled to room temperature and adjusted the pH 6-7 with 2N aq. HCl solution. The mixture was concentrated under reduced pressure and the residue was purified by preparative reverse-phase HPLC to provide the title compound (34 mg) as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.50 (bs, 1H), 8.32 (d, J=4 Hz, 2H), 7.50 (d, J=5 Hz, 2H), 6.01 (d, J=7 Hz, 1H), 4.80 (d, J=6 Hz, 1H), 4.38 (q, J=3 Hz, 1H), 4.10-3.95 (m, 2H), 3.45 (bs, 2H), 3.06 (s, 1H); LC/MS [M+H]=519.0. Example 8 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(furan-3-ylmethyl)malonic acid Step 1: To a mixture of PPh3(4.28 g, 16.31 mmol, 1.6 eq) and CBr4(4.06 g, 12.23 mmol, 1.2 eq) in DCM (20 mL) at 0° C. under N2atmosphere was added furan-3-ylmethanol (1 g, 10.19 mmol, 1 eq) in DCM (5 mL) dropwise. The mixture was stirred at 20° C. for 2 h before it was quenched with saturated aq. NaHCO3(30 mL) and then extracted with EtOAc (2×15 mL). The combined organic layer was washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to provide crude 3-(bromomethyl)furan (2.9 g) as yellow gum which was used in the next step directly without further purification. Step 2: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (1 g, 2.41 mmol, 1 eq) in DMF (5 mL) at 20° C. was added Cs2CO3(2.36 g, 7.24 mmol, 3 eq) and crude 3-(bromomethyl)furan (2.9 g) in DMF (6 mL). The mixture was stirred for 2 h before it was partitioned between water (30 mL) and EtOAc (30 mL). The aqueous phase was extracted with EtOAc (3×15 mL). The combined organic extract was washed with water (20 mL), saturated aq. NH4Cl (2×20 mL), brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on SiO2(10-25% EtOAc in petroleum ether) to give diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(furan-3-ylmethyl)malonate (375 mg, 31% yield) as a light yellow oil. Step 3: To a mixture of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(furan-3-ylmethyl)-malonate (375 mg, 758.36 umol, 1 eq) in DCM (2 mL) and H2O (0.4 mL, 22.20 mmol, 29 eq) was added TFA (2 mL, 27.01 mmol, 36 eq). The mixture was stirred at 20° C. for 12 h before it was concentrated under reduced pressure to provide crude diethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxy-tetrahydrofuran-2-yl)methoxy)-2-(furan-3-ylmethyl)malonate (420 mg) as an oil which was used in next step without further purification. Step 4: To a mixture of crude diethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxytetrahydro-furan-2-yl)methoxy)-2-(furan-3-ylmethyl)malonate (420 mg, 1.02 mmol, 1 eq) in pyridine (4 mL) at 20° C. was added Ac2O (954 uL, 10.18 mmol, 10 eq) and 4-DMAP (373 mg, 3.06 mmol, 3 eq). The mixture was stirred for 12 h before it was partitioned between water (15 mL) and EtOAc (10 mL). The aqueous phase was extracted with EtOAc (2×20 mL). The combined organic extract was washed with water (10 mL), 0.5N aq. HCl (2×5 mL), brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by preparative TLC (petroleum ether:EtOAc=3:1) to give diethyl 2-(furan-3-ylmethyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (95 mg, 17% yield) as a yellow oil. Step 5: To a mixture of diethyl 2-(furan-3-ylmethyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (50 mg, 92.85 umol, 1 eq) and 2-chloro-addenine (17.32 mg, 102.14 umol, 1.1 eq) in MeCN (1.2 mL) was added BSA (57.38 uL, 232.13 umol, 2.5 eq) at 25° C. under N2atmosphere. The mixture was stirred at 65° C. for 0.5 h before it was cooled to 0° C. and followed by dropwise addition of TMSOTf (33.56 uL, 185.70 umol, 2 eq). The mixture was stirred at 0° C. for 0.5 h and then at 65° C. for 2 h before it was cooled to room temperature and quenched with saturated aq. NaHCO3solution (2 mL). The mixture was extracted with EtOAc (3×2 mL). The combined organic layer was washed with brine (2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc=1:1) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(furan-3-ylmethyl)malonate (17 mg, 29% yield) as a yellow gum. Step 6: To a mixture of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(furan-3-ylmethyl)-malonate (33 mg, 50.92 umol, 1 eq) in THF (1 mL) was added 1N aq. LiOH (1 mL). The mixture was stirred at 20° C. for 3 h before it was extracted with EtOAc (2 mL). The organic layer was discarded. The aqueous phase was adjusted the pH 2-3 with 2N aq. HCl before it was extracted with EtOAc (4×5 mL). The combined organic extract was washed with brine (3 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was dissolved in a mixture of MeCN (1 mL) and H2O (1 mL) and then dried by lyophilization to provide the title compound (10.0 mg, 37% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.48 (s, 1H) 7.81 (br s, 2H) 7.35 (s, 2H) 6.29 (s, 1H) 6.22 (br s, 1H) 6.02 (br d, J=6.50 Hz, 1H) 5.83 (d, J=7.50 Hz, 1H) 4.75-4.90 (m, 1H) 4.16 (dd, J=4.75, 2.75 Hz, 1H) 3.92 (br dd, J=10.07, 5.07 Hz, 1H) 3.77 (br d, J=8.00 Hz, 1H) 3.48 (s, 1H) 3.08 (s, 2H); LC/MS [M+H]=507.9. Example 9 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Step 1: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy-)malonate (7.26 g, 4.92 mmol, 1 eq) in DMF (80 mL) at 25° C. was added K2CO3(13.60 g, 98.40 mmol, 20 eq). The reaction mixture was stirred for 0.5 h and followed by addition of 1-(bromomethyl)-4-nitro-benzene (15.94 g, 73.80 mmol, 15 eq). The reaction mixture was stirred for 24 h before it was diluted with H2O (300 mL) and extracted with EtOAc (3×60 mL). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, filtered and concentrated. The crude residue was purified by flash silica gel column chromatography (petroleum ether:EtOAc=10:1-2:1) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetra-hydrofuran-2-yl)methoxy)-2-(4-nitrobenzyl)malonate (2.36 g) was obtained as a brown gum. Step 2: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)-amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetra-hydrofuran-2-yl)methoxy)-2-(4-nitrobenzyl)-malonate (2.26 g, 2.81 mmol, 1 eq) in EtOH (23 mL) at 0° C. was added Fe (786 mg, 14.07 mmol, 5 eq) and NH4Cl (151 mg, 2.81 mmol, 1 eq) in H2O (8.5 mL). The reaction mixture was stirred at 50° C. for 4 h before it was filtered and the filtrate was concentrated. The crude residue was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0-1:1) to provide diethyl 2-(4-aminobenzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)-amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetra-hydrofuran-2-yl)methoxy)-malonate (280 mg) as a yellow foam. Step 3: To a solution of diethyl 2-(4-aminobenzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)malonate (280 mg, 362.14 umol, 1 eq) in DCM (3 mL) at 0° C. was added 1-chloro-3-isocyanatopropane (86.59 mg, 724.28 umol, 2 eq). The reaction mixture was stirred at 25° C. for 16 h before it was concentrated. The crude residue was purified by preparative TLC (petroleum ether:EtOAc=1:0-1:1) to provide diethyl 2-(4-(3-(3-chloropropyl)ureido)-benzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (120 mg, 33% yield) as a foam. Step 4: To a solution of diethyl 2-(4-(3-(3-chloropropyl)ureido)benzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetra-hydrofuran-2-yl)methoxy)malonate (120 mg, 134.42 umol, 1 eq) in THF (1.2 mL) at 0° C. was added NaH (11 mg, 268.84 umol, 60% in mineral oil, 2 eq). The reaction mixture was stirred at 25° C. for 2 h before it was quenched with H2O (0.2 mL) at 0° C. The reaction mixture was then stirred at 25° C. for 16 h. The reaction mixture was acidified to pH 3-4 with 1N aq. HCl solution and then extracted with EtOAc (3×5 mL). The combined organic layer was washed with brine (15 mL), dried over anhydrous Na2SO4, filtered and concentrated to provide crude 2-(((2R,3S,4R,5R)-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-tetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid (77 mg, 73% yield) as a white solid. Step 5: To a solution of crude 2-(((2R,3S,4R,5R)-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetra-hydropyrimidin-1 (2H)-yl)benzyl)malonic acid (76 mg, 106.13 umol, 1 eq) in DCM (0.5 mL)) at 0° C. was added TFA (0.25 mL, 3.38 mmol, 32 eq). The reaction mixture was stirred at 25° C. for 2 h before it was concentrated. The residue was re-dissolved with saturated aq. NaHCO3solution (5 mL) and extracted EtOAc (3×5 mL). The combined organic layer was washed with brine (15 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by preparative reversed-phase HPLC to provide the title compound (8.6 mg, 12% yield) as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.28 (s, 1H), 7.11 (d, J=8.52 Hz, 2H), 7.02 (d, J=8.52 Hz, 2H), 5.99 (d, J=7.44 Hz, 1H), 4.79 (d, J=7.41 Hz, 1H), 4.29 (t, J=2.76 Hz, 1H), 4.01-3.91 (m, 2H), 3.54-3.41 (m, 4H), 3.36-3.32 (m, 2H), 3.05 (s, 1H), 2.03-1.94 (m, 2H); LC/MS [M+H]=616.2. Example 10 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxopiperidin-1-yl)benzyl)malonic acid Step 1: To a solution of crude diethyl 2-(4-aminobenzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)malonate (140 mg, 160 umol, 1 eq) in DCM (2 mL) at 25° C. was added TEA (107 mg, 1.06 mmol, 147 uL, 6.59 eq) and followed by 5-chloropentanoyl chloride (24.9 uL, 192 umol, 1.2 eq). The mixture was stirred for 1 h before it was partitioned between DCM (20 mL) and H2O (20 mL). The organic phase was washed with H2O (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether:EtOAc=1:1) to give diethyl 2-(4-(5-chloropentanamido)-benzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (130 mg, 69% yield) as a yellow gum. Step 2: To a solution of diethyl 2-(4-(5-chloropentanamido)-benzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydro-furan-2-yl)methoxy)malonate (104 mg, 105 umol, 1 eq) in THF (2 mL) at 25° C. was added NaH (25.2 mg, 630 umol, 60% in mineral oil, 6 eq). The mixture was stirred for 4 h before it was quenched with H2O (1 mL). The reaction mixture was stirred at 20° C. for 14 before it was partitioned between EtOAc (10 mL) and water (20 mL). The aqueous phase was acidified to pH 5-6 with 2N aq. HCl solution before it was partitioned between EtOAc (20 mL) and brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude 2-(((2R,3S,4R,5R)-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxopiperidin-1-yl)benzyl)malonic acid (58 mg) as a colorless gum. Step 3: To a mixture of crude 2-(((2R,3S,4R,5R)-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxopiperidin-1-yl)benzyl)malonic acid (58 mg, 81 umol, 1 eq) in DCM (500 uL) was added TFA (400 uL, 5.40 mmol, 67 eq). The mixture was stirred at 20° C. for 2 h before it was quenched with 2N aq. LiOH (500 uL). The mixture was partitioned between EtOAc (10 mL) and water (10 mL). The aqueous phase was adjusted to pH 5-6 with 2M aq. HCl solution. The aqueous phase was partitioned between EtOAc (2×20 mL) and brine (5 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by preparative HPLC and lyophilized to give the title compound (6.9 mg, 14% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.32 (s, 1H) 7.33 (d, J=8.53 Hz, 2H) 6.98 (d, J=8.28 Hz, 2H) 5.98 (d, J=7.53 Hz, 1H) 4.79 (m, 1H) 4.28 (t, J=2.76 Hz, 1H) 4.04 (br s, 2H) 3.39-3.54 (m, 4H) 3.05 (s, 1H) 2.43 (m, 2H) 1.88 (br t, J=2.89 Hz, 4H); LC/MS [M+H]=615.3. Example 11 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(1-(methoxymethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Step 1: To a solution of 3-bromopyridin-2 (1H)-one (2.25 g, 12.93 mmol, 1 eq) in acetone (40 mL) at 25° C. was added K2CO3(4.47 g, 32.33 mmol, 2.5 eq). The suspension was stirred for 0.5 h and followed by addition of MOM-Cl (2.79 mL, 36.76 mmol, 2.84 eq) dropwise. The mixture was stirred at 25° C. for 15 h before it was diluted with water (40 mL) and extracted with ethyl acetate (2×30 mL). The combined organic layer was washed with brine (50 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by Combi-flash on silica gel (20-60% ethyl acetate in petroleum ether) to provide 3-bromo-1-(methoxy-methyl)-pyridin-2 (1H)-one (1.22 g, 43% yield) as a clear oil. Step 2: To a solution of 3-bromo-1-(methoxymethyl)pyridin-2 (1H)-one (1.38 g, 6.33 mmol, 1 eq) and (4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)methanol (1.06 g, 6.96 mmol, 1.1 eq) in dioxane (12 mL) was added K2CO3(2.62 g, 18.99 mmol, 3 eq), Pd(dppf)Cl2(463 mg, 632.89 umol, 0.1 eq) and H2O (4 mL). The mixture was de-gassed with N2for 10 min before it was then heated at 80° C. for 16 h under N2atmosphere. The reaction was diluted with water (10 mL) and extracted with ethyl acetate (3×10 mL). The combined organic layer was washed with brine (25 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by Combi-flash on silica gel (50-100% ethyl acetate in petroleum ether) to provide 3-(4-(hydroxymethyl)phenyl)-1-(methoxymethyl)pyridin-2 (1H)-one (1.30 g, 84% yield) as a yellow gum. Step 3: To a solution of PPh3(8.34 g, 31.80 mmol, 6 eq) in DCM (50 mL) at −25° C. was added CBr4(10.55 g, 31.80 mmol, 6 eq). The yellow suspension was stirred at −25° C. for 1 h and followed by addition of a solution of 3-(4-(hydroxymethyl)phenyl)-1-(methoxymethyl)-pyridin-2 (1H)-one (1.30 g, 5.30 mmol, 1 eq) in DCM (10 mL) dropwise. The yellow suspension was stirred at −25° C. for 0.5 h before it was diluted with MTBE (180 mL). The precipitate was filtered off and the filtrate was concentrated to give crude (2.8 g) as a yellow gum. The crude residue was purified by Combi-flash on silica gel (30-100% ethyl acetate in petroleum ether) to provide 3-(4-(bromomethyl)phenyl)-1-(methoxymethyl)-pyridin-2 (1H)-one (760 mg, 46% yield) as a white solid. Step 4: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-malonate (100 mg, 130.18 umol, 1 eq) in DMF (1.5 mL) at 20° C. was added K2CO3(53.97 mg, 390.54 umol, 3 eq). The suspension was stirred for 0.5 h and followed by addition of 3-(4-(bromomethyl)phenyl)-1-(methoxymethyl)pyridin-2 (1H)-one (44.13 mg, 143.20 umol, 1.1 eq). The suspension was stirred at 20° C. for 16 h before it was diluted with water (2 mL) and extracted with ethyl acetate (3×2 mL). The combined organic layer was dried by Na2SO4, filtered and concentrated. The crude residue was purified by preparative TLC (petroleum ether:ethyl acetate=1:1) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(N,N′-bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydro-furan-2-yl)methoxy)-2-(4-(1-(methoxymethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)-malonate (71 mg, 55% yield) as a clear syrup. Step 5: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(N,N′-bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-2-(4-(1-(methoxymethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)-malonate (68 mg, 68.31 umol, 1 eq) in DCM (1.7 mL) at 0° C. was added TFA (0.3 mL, 4.05 mmol, 59 eq). The mixture was stirred at 20° C. for 2 h before it was quenched with saturated aq. NaHCO3solution to adjust the pH to 9. The mixture was extracted with ethyl acetate (3×8 mL). The combined organic layer was concentrated to give crude (98 mg) as a yellow gum. The crude residue was purified by preparative TLC (ethyl acetate) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-2-(4-(1-(methoxymethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)-malonate (21 mg, 38% yield) as a colorless syrup. Step 6: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(1-(methoxy-methyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (20 mg, 25.15 umol, 1 eq) in THF (1 mL) was added 1M aq. LiOH (503 uL, 20 eq). The mixture was stirred at 18° C. for 22 h before it was acidified to pH 2 with 1N aq. HCl and concentrated. The crude residue was purified by preparative HPLC and the fraction was dried by lyophilization to provide the title compound (2.1 mg, 13% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.45 (br s, 1H), 7.81 (br s, 2H), 7.67 (dd, J=6.75, 1.75 Hz, 1H), 7.51 (br d, J=6.75 Hz, 1H), 7.36 (br d, J=6.88 Hz, 2H), 7.20 (br d, J=7.50 Hz, 2H), 6.33 (t, J=6.75 Hz, 1H), 6.20 (br s, 1H), 6.01 (br d, J=6.88 Hz, 1H), 5.82 (d, J=7.50 Hz, 1H), 5.28 (s, 2H), 4.65-4.89 (m, 1H), 4.06-4.23 (m, 1H) 3.88-4.06 (m, 1H), 3.67-3.86 (m, 1H), 3.40-3.52 (m, 3H), 3.18-3.30 (m, 2H); LC/MS [M+H]=655.1. Example 12 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Step 1: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-malonate (12.59 g, 16.39 mmol, 1 eq) and 1-(bromomethyl)-4-iodo-benzene (48.67 g, 163.90 mmol, 10 eq) in DMF (120 mL) at 20° C. was added K2CO3(33.98 g, 245.85 mmol, 15 eq). The solution was stirred for 16 h before it was diluted with water (200 mL) and extracted with ethyl acetate (3×200 mL). The combined organic layer was washed with water (400 mL), brine (400 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by Combi-flash on silica gel (15-40% ethyl acetate in petroleum ether) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)-amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetra-hydrofuran-2-yl)methoxy)-2-(4-iodobenzyl)-malonate (2.94 g, 18% yield) as a yellow solid. Step 2: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-2-(4-iodobenzyl)malonate (1.10 g, 1.12 mmol, 1 eq) and (2-oxo-1,2-dihydropyridin-3-yl)boronic acid (310.53 mg, 2.24 mmol, 2 eq) in dioxane (12 mL) was added K2CO3(463.41 mg, 3.35 mmol, 3 eq), Pd(dppf)Cl2(81.78 mg, 111.77 umol, 0.1 eq) and H2O (4 mL). The mixture was degassed with N2for 10 min and then stirred at 80° C. for 1 h under N2atmosphere. The dark mixture was diluted with water (10 mL) and extracted with ethyl acetate (3×10 mL). The combined organic layer was washed with brine (30 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by Combi-flash on silica gel (40-100% ethyl acetate in petroleum ether) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (220 mg, 21% yield) as a yellow gum. Step 3: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (180 mg, 189.20 umol, 1 eq) in DCM (2.4 mL) was added TFA (0.6 mL, 8.10 mmol, 43 eq). The yellow solution was stirred at 20° C. for 2.5 h before it was quenched with saturated aq. NaHCO3(5 mL) and extracted with ethyl acetate (3×4 mL). The combined organic layer was concentrated to give crude (108 mg) as a yellow gum. The crude residue was purified by preparative TLC (ethyl acetate) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetra-hydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)-malonate (23 mg, 16% yield) as a yellow solid. Step 4: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (23 mg, 30.62 umol, 1 eq) in THF (2.5 mL) was added 1M aq. LiOH (0.6 mL, 20 eq). The reaction mixture was stirred at 20° C. for 4 h before it was acidified to pH 6 with 1N aq. HCl and concentrated to give crude (32 mg) as a yellow gum. The crude residue was purified by preparative HPLC (column: YMC-Triart Prep C18 150*40 mm*7 um; Mobile phase: [water (0.225% FA)-CAN]; B %: 15%-35%, 10 min). The product was dried by lyophilization to give the title compound (2.1 mg, 11% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.24 (s, 1H), 7.46 (dd, J=6.88, 1.63 Hz, 1H), 7.28-7.34 (m, 5H), 6.39 (t, J=6.75 Hz, 1H), 5.96 (d, J=7.38 Hz, 1H), 4.77-4.84 (m, 1H), 4.29 (t, J=2.88 Hz, 1H), 4.03 (d, J=2.75 Hz, 2H), 3.38-3.51 (m, 2H), 3.04 (s, 1H); LC/MS [M+H]=611.0. Example 13 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-((2-carboxy-[1,1′-biphenyl]-4-yl)methyl)malonic acid Step 1: To a mixture of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (99.87 mg, 130.01 umol, 1 eq) in DMF (0.5 mL) was added K2CO3(53.90 mg, 390.03 umol, 3 eq). The mixture was stirred at 40° C. for 0.5 h and followed by addition of methyl 4-(bromomethyl)-[1,1′-biphenyl]-2-carboxylate (79.35 mg, 260.02 umol, 2 eq) which was prepared according to the reported procedure by D. Stoermer et al (J. of Med. Chem.2012, 55, 5922-5932). The mixture was stirred at 40° C. for 15.5 h before it was diluted with water (4 mL) and extracted with EtOAc (3×5 mL). The combined organic layer was washed with water (2×5 mL) and brine (5 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by preparative TLC (petroleum ether:EtOAc=1:1) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)-amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-((2-(methoxy-carbonyl)-[1,1′-biphenyl]-4-yl)methyl)malonate (70 mg, 53% yield) as a colorless syrup. Step 2: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-((2-(methoxycarbonyl)-[1,1′-biphenyl]-4-yl)methyl)malonate (70 mg, 70.53 umol, 1 eq) in DCM (2 mL) was added TFA (0.5 mL, 6.75 mmol, 96 eq). The mixture was stirred at 20° C. for 2 h before it was quenched with saturated aq. NaHCO3to pH 7-8 and extracted with EtOAc (3×10 mL). The combined organic layer was washed with water (2×8 mL) and brine (8 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by preparative TLC (petroleum ether:EtOAc=1:1) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydro-furan-2-yl)-methoxy)-2-((2-(methoxycarbonyl)-[1,1′-biphenyl]-4-yl)methyl)malonate (40 mg, 71% yield) as a colorless syrup. Step 3: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-((2-(methoxy-carbonyl)-[,1′-biphenyl]-4-yl)methyl)malonate (30 mg, 37.87 umol, 1 eq) in THF (1 mL) was added 1M aq. LiOH (568 uL, 15 eq). The mixture was stirred at 25° C. for 20 h before it was diluted with water (1 mL) and extracted with EtOAc (3×2 mL). The organic layer was discarded. The pH of the water phase was adjusted to 2 with 2N aq. HCl to produce a precipitate. The precipitate was collected by filtration to give desired product (28 mg) as the first crop. The aq. phase was further extracted with EtOAc (4×2 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to provide the second crop (10 mg) as a white solid. These two crops were combined and purified by preparative HPLC and the fraction was lyophilized to give the title compound (8.0 mg, 33% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.21 (s, 1H), 7.66 (s, 1H), 7.46 (br d, J=7.6 Hz, 1H), 7.24-7.32 (m, 3H), 7.12 (dd, J=7.1, 2.3 Hz, 2H), 7.07 (d, J=7.9 Hz, 1H), 5.99 (d, J=7.4 Hz, 1H), 4.84 (br s, 1H), 4.33 (br s, 1H), 4.02-4.16 (m, 2H), 3.43-3.60 (m, 2H), 3.03 (s, 1H); LC/MS [M+H]=638.2. Example 14 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-phenyl-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a solution of 2,6-dichloroadenine (0.8 g, 4.23 mmol, 1 eq) in H2O (10 mL) and MeCN (5 mL) was added phenylboronic acid (464.49 mg, 3.81 mmol, 0.9 eq), Cs2CO3(3.45 g, 10.58 mmol, 2.5 eq), Pd(OAc)2(47.51 mg, 211.64 umol, 0.05 eq) and trisodium; 3-bis(3-sulfonatophenyl)phosphanylbenzenesulfonate (601.50 mg, 1.06 mmol, 0.25 eq) at 20° C. under N2atmosphere. The mixture was stirred at 110° C. for 2 h before it was allowed to cool and diluted with H2O (50 mL). The reaction mixture was extracted with EtOAc (5×50 mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The crude product was triturated with a mixture of petroleum ether (9 mL) and EtOAc (3 mL) to provide 2-chloro-6-phenyl-9H-purine (140 mg, 14% yield) as a yellow solid. Step 2: To a solution of 2-chloro-6-phenyl-9H-purine (321.05 mg, 585.30 umol, 1 eq) in MeCN (0.5 mL) was added diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetra-hydrofuran-2-yl)methoxy)malonate (135 mg, 585.30 umol, 1 eq) and BSA (347 uL, 1.40 mmol, 2.4 eq) at 15° C. The mixture was stirred at 65° C. for 30 min as it became clear. The mixture was cooled to 0° C. and followed by dropwise addition of TMSOTf (126.91 uL, 702.35 umol, 1.2 eq). The mixture was stirred for 0° C. 10 min and then at 65° C. for 3 h before it was cooled and quenched with saturated aq. NaHCO3(40 mL). The reaction mixture was extracted with EtOAc (2×30 mL). The combined organic layer was washed with brine (40 mL) and dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (petroleum:EtOAc=1:0-1:1) first and then by preparative TLC (petroleum:EtOAc=1:1) to provide diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-phenyl-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (135 mg) as a yellow gum. Step 3: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-phenyl-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (135 mg, 187.73 umol, 1 eq) in THF (2 mL) was added LiOH·H2O (78.77 mg, 1.88 mmol, 10 eq) in H2O (0.2 mL) at 20° C. The mixture was stirred at 45° C. for 2 h before it was diluted with H2O (10 mL) and with EtOAc (2 mL). The organic layer was discarded and the aqueous phase was acidified with 2N aq. HCl to pH 2-3. Then the aqueous phase was extracted with EtOAc (3×20 mL). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by preparative HPLC to provide the title compound (27.6 mg, 24% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 9.23 (s, 1H), 8.75-8.81 (m, 2H), 7.61-7.68 (m, 3H), 7.07-7.22 (m, 5H), 6.03 (d, J=6.53 Hz, 1H), 4.62 (d, J=6.53 Hz, 1H), 4.20 (dd, J=6.90, 2.64 Hz, 1H), 3.72 (br dd, J=9.91, 7.15 Hz, 2H), 3.55 (s, 1H), 3.03 (br d, J=1.51 Hz, 2H); LC/MS [M+H]=579.1. Example 15 Synthesis of 2-allyl-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (600 mg, 1.45 mmol, 1 eq) in DMF (1 mL) was added allyl bromide (263 mg, 2.17 mmol, 1.5 eq) and Cs2CO3(943 mg, 2.90 mmol, 2 eq). The mixture was stirred at 20° C. for 2 h before it was diluted with water (15 mL) and extracted with EtOAc (4×10 mL). The combined organic phase was washed with water (10 mL), brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated to provide crude diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydro-furo[2,3-d][1,3]-dioxol-5-yl)methoxy)-2-allylmalonate (685 mg) as a colorless gum. Step 2: To a solution of crude diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-allylmalonate (685 mg, 1.51 mmol, 1 eq) in DCM (5 mL) was added TFA (5 mL, 67.53 mmol, 45 eq) and H2O (1 mL, 55.51 mmol, 37 eq). The mixture was stirred at 20° C. for 16 h before it was diluted with water (15 mL) and adjusted the pH to 7-8 with solid NaHCO3. The reaction mixture was extracted with a mixture of DCM and MeOH (4×12 mL; 10:1/v:v). The combined extract was washed with saturated aq. NaHCO3(8 mL), brine (8 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxytetrahydrofuran-2-yl)methoxy)-2-allylmalonate (580 mg) as a yellow gum. Step 3: To a solution of crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-allylmalonate (580 mg, 1.40 mmol, 1 eq) in pyridine (5 mL) was added Ac2O (1.31 mL, 14.00 mmol, 10 eq) and 4-DMAP (513 mg, 4.20 mmol, 3 eq). The mixture was stirred at 20° C. for 15 before it was diluted with water (15 mL) and extracted with EtOAc (3×10 mL). The combined extract was washed with 0.5 N aq. HCl (2×8 mL), NaHCO3(2×8 mL), brine (8 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether:EtOAc=5:1-2:1) to give (420 mg, 60% yield) as a colorless gum. Step 4: To a mixture of diethyl 2-allyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetra-hydrofuran-2-yl)methoxy)malonate (360 mg, 722.20 umol, 1 eq) and 2-chloro-9H-purin-6-amine (135 mg, 794.42 umol, 1.1 eq) in MeCN (5 mL) was added BSA (446.28 uL, 1.81 mmol, 2.5 eq) at 25° C. under N2atmosphere. The mixture was stirred at 65° C. for 0.5 h. The reaction mixture was cooled to 0° C. and followed by dropwise addition of TMSOTf (261.00 uL, 1.44 mmol, 2 eq) in MeCN (1 mL). The mixture was stirred at 65° C. for 3 h before it was allowed to cooled and quenched with saturated aq. NaHCO3solution (15 mL). Then the mixture was extracted with EtOAc (4×10 mL), washed with saturated brine (8 mL), dried over anhydrous Na2SO4, filtrated and concentrated. The crude product was purified by Combi-flash on silica gel (20-40% EtOAc in petroleum ether) to give diethyl 2-allyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetra-hydrofuran-2-yl)methoxy)malonate (190 mg) as a colorless gum. Step 5: To a solution of diethyl 2-allyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (100 mg, 164.47 umol, 1 eq) in THF (0.5 mL) was added LiOH·H2O (6.90 mg, 164.47 umol, 1 eq). The mixture was stirred at 50° C. for 1 h before it was diluted with water (6 mL) and extracted with EtOAc (3×4 mL). The organic layer was discarded. The pH of the aq. phase was adjusted to 2 with 2N aq. HCl solution. The aq. phase was then extracted with EtOAc (4×6 mL). The combined organic phases was washed with brine (6 mL), dried over anhydrous Na2SO4, filtered and concentrated to provide a solid. The solid was dissolved in a mixture of MeCN (1 mL) and water (1 mL) and then lyophilizied directly to give the title compound (65.2 mg, 83% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.83 (s, 1H) 6.05 (d, J=7.5 Hz, 1H) 5.84 (br dd, J=17.2, 10.2 Hz, 1H) 5.15 (dd, J=17.2, 1.6 Hz, 1H) 4.99-5.05 (m, 2H) 4.27 (t, J=2.5 Hz, 1H) 4.00 (dd, J=10.2, 2.6 Hz, 1H) 3.79 (dd, J=10.3, 2.8 Hz, 1H) 3.06 (s, 1H) 2.88 (d, J=7.3 Hz, 2H); LC/MS [M+H]=467.9. Example 16 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-ethylmalonic acid Proceeding as described in Example 15 above but substituting allyl bromide with EtBr provided the title compound as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.53-8.83 (m, 1H) 7.81 (br s, 2H) 5.91-6.40 (m, 2H) 5.83 (d, J=7.88 Hz, 1H) 4.91 (d, J=7.75 Hz, 1H) 4.14 (t, J=2.88 Hz, 1H) 3.75 (dd, J=10.26, 3.25 Hz, 1H) 3.56 (s, 1H) 3.47-3.51 (m, 1H) 1.93-2.04 (m, 2H) 0.76 (t, J=7.38 Hz, 3H); LC/MS [M+H]=455.9. Example 17 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-methylmalonic acid Proceeding as described in Example 15 above but substituting allyl bromide with methyl bromide provided the title compound as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 13.38 (br s, 2H) 8.69 (s, 1H) 7.82 (br s, 2H) 6.17 (br s, 1H) 5.98 (br d, J=7.25 Hz, 1H) 5.82 (d, J=7.63 Hz, 1H) 4.85 (br t, J=6.94 Hz, 1H) 4.15 (t, J=2.75 Hz, 1H) 3.83 (dd, J=10.13, 3.25 Hz, 1H) 3.53-3.65 (m, 2H) 1.52 (s, 3H); LC/MS [M+H]=441.8. Example 18 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(thiophen-2-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a mixture of 2-chloroadenine (800 mg, 4.23 mmol, 1 eq) in MeCN (5 mL) and H2O (10 mL) at 20° C. under N2atmosphere was added 4,4,5,5-tetramethyl-2-(thiophen-2-yl)-1,3,2-dioxaborolane (800.37 mg, 3.81 mmol, 0.9 eq), Pd(OAc)2(47.51 mg, 211.64 umol, 0.05 eq), Cs2CO3(3.45 g, 10.58 mmol, 2.5 eq) and triphenylphosphine-3,3′-3″-trisulfonic acid trisodium salt (601.50 mg, 1.06 mmol, 0.25 eq). The mixture was stirred at 110° C. for 3 h before it was allowed to cool and partitioned between EtOAc (3×20 mL) and H2O (10 mL). The combined organic phase was washed with H2O (2×10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with (petroleum ether:EtOAc=3:1) and left standing for for 14 h. The precipitate was collected by suction filtration and dried to provide 2-chloro-6-(thiophen-2-yl)-9H-purine (190 mg, 19% yield) as a yellow solid. Step 2: To a mixture of 2-chloro-6-(thiophen-2-yl)-9H-purine (140 mg, 591.51 umol, 1 eq) and diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (324.47 mg, 591.51 umol, 1 eq) in MeCN (3 mL) at 0° C. was added DBU (267 uL, 1.77 mmol, 3 eq). The mixture was stirred at 0° C. for 10 min and followed by dropwise addition of TMSOTf (481 uL, 2.66 mmol, 4.5 eq). The mixture was stirred at 0° C. for 30 min and then at 65° C. under N2atmosphere for 14 h. The reaction mixture was allowed to cool and partitioned between EtOAc (3×20 mL) and saturated aq. NaHCO3(2×10 mL). The combined organic phase was washed with brine (2×20 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by preparative TLC (petroleum ether:EtOAc=2:1) to provide diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(thiophen-2-yl)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (150 mg, 31% yield) as a white solid. Step 3: To a mixture of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(thiophen-2-yl)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (150 mg, 206.85 umol, 1 eq) in THF (2 mL) was added LiOH·H2O (2 M, 2 mL, 19.34 eq). The mixture was stirred at 25° C. for 2 h before it was partitioned between EtOAc (10 mL) and water (10 mL). The aqueous phase was adjusted to pH˜2 with 2M aq. HCl solution. The aqueous phase was partitioned between EtOAc (40 mL) and brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by preparative HPLC (column: YMC-Triart Prep C18 150*40 mm*7 um; mobile phase: [water (0.225% FA)-ACN]; B %: 43%-63%, 10 min) and lyophilized to provide the title compound (31.6 mg, 26% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.87 (s, 1H) 8.60 (dd, J=3.76, 1.25 Hz, 1H) 8.02 (dd, J=5.02, 1.00 Hz, 1H) 7.36 (dd, J=4.89, 3.89 Hz, 1H) 7.16-7.28 (m, 2H) 6.93-7.10 (m, 3H) 6.31 (br s, 1H) 6.11 (br d, J=6.02 Hz, 1H) 5.99 (d, J=7.53 Hz, 1H) 4.88-4.97 (m, 1H) 4.23 (dd, J=4.27, 2.76 Hz, 1H) 3.99 (br dd, J=10.42, 4.39 Hz, 1H) 3.84 (br d, J=8.53 Hz, 1H) 3.56 (s, 1H) 3.26 (s, 2H); LC/MS [M+H]=585.0. Example 19 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1-propyl-1,2-dihydropyridin-3-yl)benzyl)malonic acid Step 1: To a solution of 3-bromopyridin-2 (1H)-one (3 g, 17.24 mmol, 1 eq) in acetone (100 mL) was added K2CO3(11.91 g, 86.21 mmol, 5 eq). The suspension was stirred at 20° C. for 0.5 h and followed by addition of 1-bromopropane (4.71 mL, 51.73 mmol, 3 eq) and KI (859 mg, 5.17 mmol, 0.3 eq). The mixture was stirred at 20° C. for 16 h. Additional amount of 1-bromopropane (1.0 g) was added to the reaction mixture and the mixture was stirred further at 20° C. for 3 h. The reaction was diluted with water (50 mL) and extracted with EtOAc (3×40 mL). The combined organic layer was washed with brine (100 mL), and dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by Combi-Flash (silica gel, 20-60% EtOAc in petroleum ether) to provide 3-bromo-1-propylpyridin-2 (1H)-one (1.23 g, 33% yield) as a clear oil. Step 2: To a solution of 3-bromo-1-propylpyridin-2 (1H)-one (1.79 g, 8.28 mmol, 1 eq) and (4-(hydroxymethyl)phenyl)boronic acid (1.38 g, 9.11 mmol, 1.1 eq) in dioxane (18 mL) was added K2CO3(3.43 g, 24.84 mmol, 3 eq), Pd(dppf)Cl2(606 mg, 828.00 umol, 0.1 eq) and H2O (6 mL). The mixture was degassed with N2for 10 min and then stirred at 80° C. for 16 h under N2atmosphere. The reaction mixture was cooled and filtered. The filtrate was concentrated. The residue was purified by Combi Flash (silica gel, 30-50% of EtOAc in petroleum ether) to provide 3-(4-(hydroxymethyl)phenyl)-1-propylpyridin-2 (1H)-one (1.84 g, 910% yield) as a brown solid. Step 3: To a solution of PPh3(1.94 g, 7.40 mmol, 6 eq) in DCM (15 mL) was added CBr4(2.45 g, 7.40 mmol, 6 eq) at −25° C. The yellow solution was stirred at −25° C. for 1 h and followed by addition of 3-(4-(hydroxymethyl)phenyl)-1-propylpyridin-2 (1H)-one (300 mg, 1.23 mmol, 1 eq) in DCM (3 mL) dropwise. The yellow suspension was stirred at −25° C. for 0.5 h to produce a yellow suspension. The reaction mixture was diluted with MTBE (50 mL) to produce more precipitate. The precipitate was filtered off and the filtrate was concentrated. The residue was purified by CombiFlash (silica gel column, 10-100% of EtOAc in petroleum ether) to provide 3-(4-(bromomethyl)phenyl)-1-propylpyridin-2 (1H)-one (243 mg, 64% yield) as a clear oil. Step 4: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (554 mg, 721.20 umol, 1 eq) in DMF (5 mL) was added K2CO3(299.03 mg, 2.16 mmol, 3 eq). The mixture was stirred at 20° C. for 0.5 h and followed by addition of 3-(4-(bromomethyl)phenyl)-1-propylpyridin-2 (1H)-one (242.91 mg, 793.32 umol, 1.1 eq). The mixture was stirred at 20° C. for 16 h before it was diluted with water (30 mL) and extracted by EtOAc (4×20 mL). The combined organic layer was washed with water (100 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by Combi Flash (silica gel, 20-30% of EtOAc in petroleum ether to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-tetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1-propyl-1,2-dihydropyridin-3-yl)benzyl)-malonate (209 mg) as a colorless gum. Step 5: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1-propyl-1,2-dihydropyridin-3-yl)benzyl)malonate (209 mg, 210.38 umol, 1 eq) in DCM (2 mL) at 0° C. was added TFA (0.7 mL, 9.45 mmol, 44.94 eq). The solution was stirred at 20° C. for 2 h before it was quenched by saturated aq. NaHCO3(5 mL) and extracted with EtOAc (4×10 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by Combi Flash (silica gel, 30-70% of EtOAc in petroleum ether) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1-propyl-1,2-dihydro-pyridin-3-yl)benzyl)malonate (91 mg, 55% yield) as a white solid. Step 6: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1-propyl-1,2-dihydro-pyridin-3-yl)benzyl)malonate (91 mg, 114.72 umol, 1 eq) in THF (1 mL) was added 1N aq. LiOH (1 mL). The mixture was stirred at 20° C. for 2.5 h before it was diluted with water (5 mL) and the resulting solution was washed with EtOAc (2×10 mL). The organic extract was discarded. The aqueous layer acidified to pH 2 with 2N aq. HCl and then extracted with EtOAc (4×8 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was dissolved in a mixture of MeOH (5 mL) and water (20 mL) and was dried by lyophilization to provide the title compound (51 mg, 67% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.08 (s, 1H), 7.53 (dd, J=6.80, 2.0 Hz, 1H), 7.28-7.39 (m, 5H), 6.34 (t, J=6.9 Hz, 1H), 5.96 (d, J=7.6 Hz, 1H), 4.77 (d, J=7.6 Hz, 1H), 4.28 (s, 1H), 4.07-4.15 (m, 1H), 4.01 (dd, J=10.3, 2.8 Hz, 1H), 3.92 (t, J=7.4 Hz, 2H), 3.39-3.58 (m, 2H), 3.06 (s, 1H), 1.65-1.80 (m, 2H), 0.94 (t, J=7.4 Hz, 3H); LC/MS [M+H]=653.1. Example 20 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(1-ethyl-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Proceeding as described in Example 19 above but substituting propyl bromide with ethyl bromide provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.15 (s, 1H), 7.53 (dd, J=6.6, 1.3 Hz, 1H), 7.25-7.38 (m, 5H), 6.32 (t, J=6.9 Hz, 1H), 5.96 (d, J=7.6 Hz, 1H), 4.78 (d, J=7.5 Hz, 1H), 4.30 (s, 1H), 4.09-4.15 (m, 1H), 3.95-4.04 (m, 3H), 3.39-3.58 (m, 2H), 3.06 (s, 1H), 1.28-1.32 (m, 3H); LC/MS [M+H]=639.1. Example 21 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(1-(2-hydroxyethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Step 1: To a mixture of 3-bromopyridin-2 (1H)-one (3 g, 17.24 mmol, 1 eq) in acetone (100 mL) at 20° C. was added KI (859 mg, 5.17 mmol, 0.3 eq), K2CO3(5.96 g, 43.10 mmol, 2.5 eq) and 2-bromoethanol (4.90 mL, 68.97 mmol, 4 eq). The mixture was stirred for 4 before it was filtered and the filter cake was washed with EtOAc (100 mL). The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel column chromatography (petroleum ether:EtOAc=5:1 to 0:1) to provide 3-bromo-1-(2-hydroxyethyl)pyridin-2 (1H)-one (2.2 g, 59% yield) as a yellow gum. Step 2: To a mixture of 3-bromo-1-(2-hydroxyethyl)pyridin-2 (1H)-one (2.2 g, 10.09 mmol, 1 eq) in DMF (15 mL) at 20° C. was added imidazole (1.72 g, 25.22 mmol, 2.5 eq) and TBDPSCl (5.18 mL, 20.18 mmol, 2 eq). The mixture was stirred for 2 h before it was diluted with H2O (60 mL) and extracted with EtOAc (3×30 mL). The combined extract was washed with saturated aq. NH4Cl (2×30 mL), brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel column chromatography (petroleum ether:EtOAc=1:0 to 6:1) to provide 3-bromo-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)-pyridin-2 (1H)-one (3.75 g, 79% yield) as a light yellow oil. Steps 3-8: Proceeding as described in Example 19 above but substituting 3-(4-(bromomethyl)-phenyl)-1-propylpyridin-2 (1H)-one with 3-bromo-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)-pyridin-2 (1H)-one provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.33 (s, 1H), 7.50-7.57 (m, 1H), 7.41-7.47 (m, 1H), 7.39 (d, J=8.13 Hz, 2H), 7.29 (br d, J=8.13 Hz, 2H), 6.35 (t, J=6.88 Hz, 1H), 5.98 (d, J=7.13 Hz, 1H), 4.71 (d, J=7.00 Hz, 1H), 4.30 (br s, 1H), 3.93-4.16 (m, 4H), 3.83 (m, 2H), 3.36-3.50 (m, 2H), 3.05 (s, 1H); LC/MS [M+H]=655.1. Example 22 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-methoxypyridin-3-yl)benzyl)malonic acid Step 1: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-2-(4-iodobenzyl)malonate (900 mg, 914.47 umol, 1 eq) and (2-methoxy-3-pyridyl)boronic acid (168 mg, 1.10 mmol, 1.2 eq) in dioxane (9 mL) 25° C. was added K2CO3(379 mg, 2.74 mmol, 3 eq), Pd(dppf)Cl2(67 mg, 91.45 umol, 0.1 eq) and H2O (3 mL). The mixture was degassed with N2for a while and then heated to 80° C. for 16 h before it was diluted with water (10 mL), and extracted with ethyl acetate (2×10 mL). The combined organic layer was dried by Na2SO4, filtered and concentrated. The crude product was purified by Combi-flash (silica gel, 10-50% of EtOAc in petroleum ether) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydro-furan-2-yl)-methoxy)-2-(4-(2-methoxypyridin-3-yl)benzyl)malonate (117 mg, 13% yield) as a yellow gum. Step 2: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)-2-(4-(2-methoxypyridin-3-yl)benzyl)malonate (90 mg, 93.23 umol, 1 eq) in DCM (3 mL) was added TFA (0.4 mL, 5.40 mmol, 58 eq). The solution was stirred at 20° C. for 2 h before it was quenched with saturated aq. NaHCO3(4 mL) and extracted with EtOAc (3×4 mL). The combined organic layer was concentrated to give crude diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-methoxypyridin-3-yl)benzyl)-malonate (42 mg) as a yellow gum. Step 3: To a solution of crude diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-methoxy-pyridin-3-yl)benzyl)malonate (42 mg, 54.89 umol, 1 eq) in THF (2.5 mL) was added 1M aq. LiOH (0.8 mL, 15 eq). The reaction mixture was stirred at 20° C. for 4 h before it was acidified to pH 6 with 1N aq. HCl and concentrated. The crude product was purified by preparative HPLC and the fraction was dried by lyophilization to give the title compound (6 mg, 17% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.10 (s, 1H), 8.01 (dd, J=4.94, 1.69 Hz, 1H), 7.48 (dd, J=7.25, 1.63 Hz, 1H), 7.24-7.35 (m, 4H), 6.93 (dd, J=7.32, 5.07 Hz, 1H), 5.97 (d, J=7.50 Hz, 1H), 4.89-4.96 (m, 1H), 4.29 (br s, 1H), 4.05 (br d, J=5.00 Hz, 2H), 3.76 (s, 3H), 3.51 (br d, J=14.76 Hz, 1H), 3.42 (br d, J=14.51 Hz, 1H), 3.01 (s, 1H); LC/MS [M+H]=625.1. Example 23 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(3-(trifluoromethoxy)benzyl)malonic acid Proceeding as described in Example 1 above but substituting benzyl bromide with 1-(bromomethyl)-4-(trifluoromethoxy)benzene provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.43 (s, 1H), 7.09-7.25 (m, 3H), 6.95-6.98 (d, J=8.1 Hz, 1H), 6.01-6.04 (d, J=7.32 Hz, 1H), 5.00-5.03 (d, J=7.41 Hz, 1H), 4.35-4.37 (t, J=3.33 Hz, 1H), 4.05-4.15 (m, 2H), 3.38-3.53 (q, J=15 Hz, 2H), 2.99 (s, 1H); LC/MS [M+H]=602.0. Example 24 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiophen-3-ylmethyl)malonic acid Proceeding as described in Example 1 above but substituting benzyl bromide with 3-(bromomethyl)thiophene provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.39 (s, 1H), 7.09-7.17 (m, 2H), 6.98-7.00 (d, J=5.04.0 Hz, 1H), 6.01-6.04 (d, J=7.47 Hz, 1H), 5.00-5.02 (d, J=7.29 Hz, 1H), 4.32-4.34 (t, J=2.76 Hz, 1H), 4.01-4.11 (m, 2H), 3.41-3.54 (q, J=15 Hz, J=9.03 Hz, 2H), 2.98 (s, 1H); LC/MS [M+H]=524.0. Example 25 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(prop-2-yn-1-yl)malonic acid Proceeding as described in Example 1 above but substituting benzyl bromide with propargyl bromide provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.96 (s, 1H), 6.07-6.09 (d, J=7.53 Hz, 1H), 5.01-5.04 (d, J=7.53 Hz, 1H), 4.29-4.30 (m, 1H), 3.92-4.05 (m, 2H), 3.01-3.15 (m, 2H), 2.99 (s, 1H), 2.28-2.30 (t, J=2.58 Hz, 1H); LC/MS [M+H]=467. Example 26 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: A solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (200 mg, 0.26 mmol) in CH2Cl2(1 mL) under argon atmosphere at 0° C. was added TFA (0.5 mL). The mixture was stirred for 5 minutes and allowed to warm up and stirred for 1 h. Additional amount of TFA (0.4 mL) was added to the reaction mixture and it was stirred further 1.5 h before it was concentrated. The residue was azeotroped with DCM (5×5 mL) under reduced pressure to provide crude diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydro-furan-2-yl)methoxy)malonate which was used in the next step without further purification. Step 2: To a solution of crude diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (0.26 mmol) from the previous step in a mixture of MeOH (8.5 mL) and water (1.5 mL) was added powdered lithium hydroxide monohydrate (86 mg, 2.08 mmol). The resulting mixture was stirred for 16 h before the organic volatile was removed under reduced pressure. The residue was diluted with additional water (11 mL) and extracted with EtOAc (12 mL). The organic layer was discarded. The aqueous phase was acidified to pH˜2.5 with 1N aq. HCl solution and extracted with EtOAc (3×12 mL). The combined organic layer was dried (Na2SO4), filtered and concentrated to provide the title compound (45.5 mg) as a light brown solid. 1H NMR (CD3OD, 300 MHz): δ 8.94 (s, 1H), 6.08 (d, J=7.52 Hz, 1H), 5.05 (d, J=7.52 Hz, 1H), 4.65 (s, 1H), 4.29 (t, J=2.40 Hz, 1H), 4.06 (dd, J=10.7, 2.5 Hz, 1H), 3.93 (dd, J=10.64, 2.50 Hz, 1H), 3.12 (s, 1H); LC/MS [M+H]=428. Example 27 Synthesis of 2-(((2R,3S,4R,5R)-3-((1H-pyrazol-3-yl)ethynyl)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a mixture of 3-iodo-1H-pyrazole (407 mg, 2.1 mmol), PdCl2(PPh3)2(82 mg, 0.12 mmol), CuI (22 mg, 0.12 mmol), and Et3N (10 mL) in THF (10 mL) under argon atmosphere was added diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)-amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (1 g, 1.2 mmol). The resulting mixture was stirred at 60° C. overnight before it was allowed to cool to room temperature and the organic volatile was removed under reduced pressure. The resulting crude residue was purified by flash silica gel column chromatography (60-100% EtOAc in hexanes) to provide diethyl 2-(((2R,3R,4R,5R)-3-((1H-pyrazol-3-yl)ethynyl)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydro-furan-2-yl)methoxy)malonate as a solid. Step 2: To a solution of diethyl 2-(((2R,3R,4R,5R)-3-((1H-pyrazol-3-yl)ethynyl)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydro-furan-2-yl)methoxy)malonate (100 mg, 0.12 mmol) in a DCM (3 mL) was added TFA (1 mL). The resulting mixture was stirred at 25° C. for 4 h before it was concentrated to provide crude diethyl 2-(((2R,3R,4R,5R)-3-((1H-pyrazol-3-yl)ethynyl)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)malonate which was used in the next step without further purification. Step 3: To a solution of crude diethyl 2-(((2R,3R,4R,5R)-3-((1H-pyrazol-3-yl)ethynyl)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)malonate in a mixture of THF (5 mL) and H2O (2 mL) was added LiOH·H2O (50 mg, 1.2 mmol). The resulting mixture was stirred at 25° C. for 24 h before it was cooled to 0° C. and acidified to pH 6.5 with 1N aq. HCl. The reaction mixture was concentrated. The crude residue was purified by preparative reversed-phase HPLC and dried by lyophilization to provide the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.96 (s, 1H), 7.56-7.76 (m, 1H), 6.50-6.57 (m, 1H), 6.12-6.14 (d, J=7.14 Hz, 1H), 5.13-5.15 (d, J=6.78 Hz, 1H), 4.63-4.70 (m, 1H), 4.38 (s, 1H), 3.99-4.13 (m, 2H); LC/MS [M+H]=495. Examples 28 & 29 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-((1-benzyl-1H-pyrazol-3-yl)ethynyl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid & 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-((1-benzyl-1H-pyrazol-5-yl)ethynyl)-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a solution of diethyl 2-(((2R,3R,4R,5R)-3-((1H-pyrazol-3-yl)ethynyl)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydro-furan-2-yl)methoxy)malonate (100 mg, 0.12 mmol), in anhydrous DMF (2 mL) under argon atmosphere at 0° C. was added oven dried Cs2CO3(78 mg, 0.24 mmole). The mixture was stirred at room temperature for 20 minutes followed by addition of benzyl bromide (29 ul, 0.24 mmole). The resulting mixture was stirred at room temperature for 2 h before it was diluted with EtOAc (15 mL) and H2O (5 mL). The organic layer was separated, washed with H2O (20 mL), brine, dried over Na2SO4and concentrated. The crude residue was purified by flash silica gel column chromatography (0-50% EtOAc in hexanes) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-3-((1-benzyl-1H-pyrazol-3-yl)ethynyl)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)-malonate and diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-3-((1-benzyl-1H-pyrazol-5-yl)ethynyl)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydro-furan-2-yl)methoxy)malonate. Steps 2-3: Proceeding as described in Example 27 above but substituting diethyl 2-(((2R,3R,4R,5R)-3-((1H-pyrazol-3-yl)ethynyl)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)malonate with diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-3-((1-benzyl-1H-pyrazol-3-yl)ethynyl)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)-malonate provided the title compound (Example 28) acid as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.95 (s, 1H), 7.685 (s, 1H), 7.22-7.38 (m, 5H), 6.50 (s, 1H), 6.10-6.13 (d, J=7.05 Hz, 1H), 5.35 (s, 2H), 5.16-5.18 (d, J=7.35 Hz, 1H), 4.68-4.76 (m, 1H), 4.38 (s, 1H), 3.96-4.16 (dd, J=9.84 Hz, J=17 Hz, 2H); LC/MS [M+H]=585. Steps 4-5: Proceeding as described in Example 27 above but substituting diethyl 2-(((2R,3R,4R,5R)-3-((1H-pyrazol-3-yl)ethynyl)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)malonate with diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-3-((1-benzyl-1H-pyrazol-5-yl)ethynyl)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)-malonate provided the title compound (Example 29) as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.88 (s, 1H), 7.53 (s, 1H), 7.23-7.35 (m, 5H), 6.59 (s, 1H), 6.10-6.13 (d, J=6.09 Hz, 1H), 5.46 (s, 2H), 5.15-5.17 (d, J=6.9 Hz, 1H), 4.35-4.41 (m, 2H), 3.71-4.01 (dd, J=10.71, J=33, 2H); LC/MS [M+H]=585. Examples 30 & 31 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-7-(isopropylamino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid & 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(dimethylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetra-hydrofuran-2-yl)methoxy)malonate (500 mg, 0.91 mmol) in MeCN (6 mL) at 25° C. was added 5,7-dichloro-1H-imidazo[4,5-b]pyridine (223 mg, 1.18 mmol) and followed by N,O-bis(trimethylsilyl)acetamide (BSA) (535 uL, 2.19 mmol). The resulting suspension was heated at 85° C. for 15 min as it became clear. The reaction mixture was allowed to cool to room temperature followed by addition of TMSOTf (262 mg, 1.18 mmol) dropwise. The reaction mixture was then refluxed at 85° C. for 3 h as all of the starting material was consumed. The reaction was quenched with cold saturated aq. NaHCO3solution and diluted with EtOAc (15 mL). The organic layer was separated, washed with H2O (20 mL), brine, dried over Na2SO4and concentrated. The crude residue was purified by flash silica gel column chromatography (0-50% EtOAc in hexanes) to provide diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(5,7-dichloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-tetrahydrofuran-2-yl)methoxy)malonate as a foam. Step 2: To a sealed tube containing diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(5,7-dichloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (80 mg, 0.12 mmol) in anhydrous DMF (1 mL) was added isopropyl amine (0.5 mL, 5.9 mmol) and Et3N (1 mL, 7.1 mmol). The reaction mixture was heated at 75° C. for 72 h before it was allowed to cool and diluted with EtOAc (15 mL) and H2O (5 mL). The organic layer was separated, washed with H2O (20 mL), brine, dried over Na2SO4and concentrated. The residue was purified by flash silica gel column chromatography (0-50% EtOAc in hexanes) to provide diethyl 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-7-(isopropylamino)-3H-imidazo-[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonate and diethyl 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-7-(dimethylamino)-3H-imidazo[4,5-b]-pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonate as foam. Step 3: To a solution of diethyl 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-7-(isopropylamino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-malonate (10 mg, 0.016 mmol) in a mixture of THF (3 mL) and H2O (1 mL) was added LiOH·H2O (10 mg, 0.24 mmol). The resulting mixture was stirred at 25° C. for 24 h before it was cooled to 0° C. and acidified to pH 6.5 with 1N aq. HCl. The crude residue was purified by preparative reversed-phase HPLC and dried by lyophilization to provide 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-7-(isopropylamino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.25 (s, 1H), 7.25-7.28 (m, 2H), 7.05 (m, 3H), 6.43 (s, 1H), 6.06-6.08 (d, J=7.17 Hz, 1H), 4.95-4.98 (d, J=7.05 Hz, 1H), 4.32 (s, 1H), 4.05-4.11 (m, 2H), 3.89-3.93 (m, 1H), 3.31-3.39 (m, 2H), 2.99 (s, 1H), 1.30-1.33 (m, 6H); LC/MS [M+H]=560. Step 4: To a solution of diethyl 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-7-(dimethyl-amino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-malonate (21 mg, 0.035 mmol) in a mixture of THF (3 mL) and H2O (1 mL) was added LiOH·H2O (30 mg, 0.71 mmol). The resulting mixture was stirred at 25° C. for 24 h before it was cooled to 0° C. and acidified to pH 6.5 with 1N aq. HCl. The reaction mixture was concentrated. The crude residue was purified by preparative reversed-phase HPLC and dried by lyophilization to provide the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.68 (s, 1H), 7.21-7.24 (m, 2H), 6.99-7.03 (m, 3H), 6.49 (s, 1H), 6.15-6.17 (d, J=7.08 Hz, 1H), 4.99-5.02 (d, J=7.17 Hz, 1H), 4.35-4.37 (t, J=3.12 Hz, 1H), 4.07-4.08 (m, 2H), 3.36-3.50 (m, 8H), 2.99 (s, 1H); LC/MS [M+H]=546. Example 32 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-hydroxyethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiophen-3-ylmethyl)malonic acid Steps 1-3: Proceeding as described in Example 15 above but substituting allyl bromide with 3-(bromomethyl)thiophene provided diethyl 2-(thiophen-3-ylmethyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate as a solid. Step 4: To a solution of 5,7-dichloro-1H-imidazo[4,5-b]pyridine (238 mg, 1.26 mmol) in MeCN (6 mL) at 25° C. was added N,O-bis(trimethylsilyl)acetamide (BSA) (571 uL, 2.34 mmol). The resulting suspension was heated at 85° C. for 15 min as it became clear. The reaction mixture was allowed to cool to room temperature followed by addition of diethyl 2-(thiophen-3-ylmethyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)-methoxy)malonate (540 mg, 0.97 mmol) and TMSOTf (228 ul, 1.26 mmol) dropwise. The reaction mixture was then refluxed at 85° C. for 2.5 h as all of the starting material was consumed. The reaction was quenched with cold saturated aq. NaHCO3solution and diluted with EtOAc (15 mL). The organic layer was separated, washed with H2O (20 mL), brine, dried over Na2SO4and concentrated. The crude residue was purified by flash silica gel column chromatography (0-50% EtOAc in hexanes) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(thiophen-3-ylmethyl)malonate as a foam. Step 5: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(thiophen-3-ylmethyl)malonate (100 mg, 0.146 mmol) in 1,4-dioxane (2 mL) was added TEA (20 uL, 0.146 mmol) followed by ethanolamine (13 ul, 0.219 mmole). The resulting mixture was stirred at 25° C. for 2 h before it was diluted with EtOAc (15 mL) and H2O (5 mL). The organic layer was separated, washed with H2O (20 mL), brine, dried over Na2SO4and concentrated to provide crude diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-((2-hydroxyethyl)amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(thiophen-3-ylmethyl)malonate which was used in the next step without further purification. Step 6: To a solution of crude diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-((2-hydroxyethyl)amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(thiophen-3-ylmethyl)malonate in a mixture of THF (4 mL) and H2O (1 mL) was added LiOH·H2O (80 mg, 1.91 mmol). The resulting mixture was stirred at 25° C. for 24 h before it was cooled to 0° C. and acidified to pH 6.5 with 1N aq. HCl. The reaction mixture was concentrated. The crude residue was purified by preparative reversed-phase HPLC and dried by lyophilization to provide the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.32 (s, 1H), 7.09-7.16 (m, 2H), 6.97-6.99 (m, 1H), 6.00-6.03 (d, J=7.41 Hz, 1H), 4.99-5.02 (d, J=7.38 Hz, 1H), 4.32-4.34 (t, J=3.03 Hz, 1H), 4.00-4.11 (m, 2H), 3.65-3.79 (m, 4H), 3.40-3.53 (q, J=15.42 Hz, J=5.79 Hz, 2H), 2.98 (s, 1H); LC/MS [M+H]=568.0. Example 33 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiophen-3-ylmethyl)malonic acid Proceeding as described in Example 32 above but substituting ethanolamine with i-PrNH2provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.28 (s, 1H), 7.08-7.16 (m, 2H), 6.97-6.99 (m, 1H), 5.99-6.02 (d, J=7.38 Hz, 1H), 5.00-5.02 (d, J=7.35 Hz, 1H), 4.31-4.43 (m, 2H), 4.00-4.12 (m, 2H), 3.40-3.53 (q, J=15.63 Hz, J=5.4 Hz, 2H), 2.98 (s, 1H), 1.27-1.32 (m, 6H); LC/MS [M+H]=566.0. Example 34 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((3-hydroxypropyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a solution of 2,6-dichloro-9H-purine_(690 mg, 3.65 mmol) in dry CH3CN (15 mL) was added N,O-bis(trimethylsilyl)acetamide (0.28 mL, 1.12 mmol) via syringe. The mixture was heated to 95° C. under argon atmosphere for 5 minutes and then cooled to ambient. To this mixture was added diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetra-hydrofuran-2-yl)methoxy)malonate (2 g, 3.65 mmol) and followed by TMSOTf (0.09 mL, 0.494 mmol). The resulting mixture was heated at 95° C. for 2.5 h before it was cooled to ambient temperature and diluted with water (60 mL) and EtOAc (60 mL). The organic phase was washed successively with equal volumes of saturated NaHCO3solution and brine. The aqueous phase was further extracted with EtOAc (2×30 mL). The combined organic phase was dried (MgSO4), filtered and concentrated. The crude residue was purified by flash silica gel column chromatography (5-60% EtOAc in hexane) to provide diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)tetrahydrofuran-2-yl)-methoxy)malonate (1.61 g) as an off-white solid. Step 2: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)malonate (102 mg, 0.15 mmol) in dry dioxane (1 mL) was added triethylamine (0.02 mL, 0.15 mmol) and 3-aminopropanol (16 mg, 0.212 mmol). The resulting mixture was stirred for 1.5 h before it was diluted with water (15 mL) and DCM (15 mL). and the organic phase was collected. The organic layer was washed with brine (15 mL). The aqueous phase were further extracted with EtOAc (2×10 mL). The combined organic layer was dried over MgSO4, filtered and concentrated to provide crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-((3-hydroxypropyl)amino)-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)malonate as a clear viscous oil. Step 3: To a solution of crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-((3-hydroxypropyl)amino)-9H-purin-9-yl)tetrahydrofuran-2-yl)methoxy)malonate (0.15 mmol) in H2O (0.2 mL), MeOH (1 mL) and THF (0.28 mL) was added powdered LiOH mono-hydrate (43 mg, 1.05 mmol)). The mixture was stirred for 4 h and then sonicated for 30 minutes. Additional LiOH mono-hydrate (7 mg) was added and sonication continued for 1 h before the organic volatile was removed under reduced pressure and the residue was diluted with water (10 mL) and EtOAc (10 mL). The mixture was cooled at 0° C. and acidified to pH˜3 with 1N aq. HCl. The organic phase was collected and the aqueous phase was further extracted with EtOAc (2×10 mL). The combined EtOAc phases were dried over MgSO4, filtered and concentrated. The crude residue was purified by preparative reversed-phase HPLC to provide the title compound as a off-white solid. 1H NMR (CD3OD, 300 MHz) δ 8.19 (bs, 1H), 7.21-7.3 (m, 2H), 7.00-7.10 (m, 3H), 6.00 (d, J=7.36 Hz, 1H), 4.98 (d, J=7.36 Hz, 1H), 4.33 (t, J=3.18 Hz, 1H), 4.02-4.15 (m, 2H), 3.68 (t, J=6.18 Hz, 2H), 3.59-3.72 (m, 2H), 3.47 (d, J=14.95 Hz, 1H), 3.38 (d, J=14.95 Hz, 1H), 2.99 (s, 1H), 1.84-1.95 (m, 2H); LC/MS [M+H]=576.0. Example 35 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((R)-2-hydroxypropyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with (R)-1-aminopropan-2-ol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.19 (bs, 1H), 7.22-7.30 (m, 2H), 7.02-7.10 (m, 3H), 6.01 (d, J=7.38 Hz, 1H), 4.98 (d, J=7.38 Hz, 1H), 4.33 (t, J=3.17 Hz, 1H), 3.99-4.13 (m, 3H), 3.58-3.69 (m, 1H), 3.43 (qt, J=14.64 Hz, 2H), 3.41-3.55 (m, 1H), 2.99 (s, 1H), 1.24 (d, J=6.30 Hz, 3H); LC/MS [M+H]=576.0. Example 36 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((S)-2-hydroxypropyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with (S)-1-aminopropan-2-ol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.19 (bs, 1H), 7.23-7.29 (m, 2H), 7.03-7.10 (m, 3H), 6.00 (d, J=7.35 Hz, 1H), 4.96 (d, J=7.35 Hz, 1H), 4.32 (t, J=3.30 Hz, 1H), 3.98-4.13 (m, 3H), 3.59-3.68 (m, 1H), 3.35-3.55 (m, 3H), 2.99 (s, 1H), 1.25 (d, J=6.30 Hz, 3H); LC/MS [M+H]=576.0. Example 37 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(6-(bis(2-hydroxyethyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with diethanolamine and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.16 (s, 1H), 7.22-7.29 (m, 2H), 6.99-7.09 (m, 3H), 6.02 (d, J=7.33 Hz, 1H), 4.97 (d, J=7.33 Hz, 1H), 4.00-4.37 (m, 2H), 4.29-4.34 (m, 2H), 4.03-4.13 (m, 3H), 3.84-3.91 (m, 4H), 3.46 (d, J=14.92 Hz, 1H), 3.37 (d, J=14.92 Hz, 1H), 2.98 (s, 1H); LC/MS [M+H]=606.0. Example 38 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-methoxyethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with 2-methoxyethylamine and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.17 (bs, 1H), 7.22-7.30 (m, 2H), 7.01-7.09 (m, 3H), 6.01 (d, J=7.33 Hz, 1H), 4.98 (d, J=7.33 Hz, 1H), 4.33 (t, J=3.20 Hz, 1H), 4.03-4.13 (m, 2H), 3.69-3.79 (m, 2H), 3.62 (t, J=5.13 Hz, 2H), 3.47 (d, J=14.89 Hz, 1H), 3.40 (s, 3H), 3.38 (d, J=14.89 Hz, 1H), 2.99 (s, 1H); LC/MS [M+H]=576.0. Example 39 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-methoxyethyl)-(methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with (2-methoxyethyl)methylamine and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.18 (bs, 1H), 7.22-7.29 (m, 2H), 6.99-7.09 (m, 3H), 6.02 (d, J=7.21 Hz, 1H), 4.98 (d, J=7.21 Hz, 1H), 4.32 (t, J=3.41 Hz, 1H), 4.03-4.14 (m, 2H), 3.68 (t, J=5.42 Hz, 2H), 3.34-3.49 (m, 7H), 3.36 (s, 3H), 2.99 (s, 1H); LC/MS [M+H]=590.0. Example 40 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((1-hydroxycyclobutyl)-methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with 1-(aminomethyl)cyclobutanol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.18 (bs, 1H), 7.23-7.29 (m, 2H), 7.01-7.09 (m, 3H), 6.01 (d, J=7.36 Hz, 1H), 4.98 (d, J=7.36 Hz, 1H), 4.32 (t, J=3.21 Hz, 1H), 4.03-4.11 (m, 2H), 3.73-3.79 (m, 2H), 3.36-3.50 (m, 2H), 2.99 (s, 1H), 2.03-2.21 (m, 4H), 1.59-1.85 (m, 2H); LC/MS [M+H]=602.0. Example 41 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(3-hydroxyazetidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with azetidin-3-ol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.29 (bs, 1H), 7.21-7.29 (m, 2H), 6.99-7.11 (m, 3H), 6.01 (d, J=7.33 Hz, 1H), 5.01 (d, J=7.33 Hz, 1H), 4.57-4.82 (m, 3H), 4.33 (t, J=3.39 Hz, 1H), 4.14-4.27 (m, 2H), 4.07 (qd, J=4.04, 2.92 Hz, 2H), 3.30-3.50 (m, 2H), 2.98 (s, 1H); LC/MS [M+H]=574.0. Example 42 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(2-(hydroxymethyl)azetidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with azetidin-2-ylmethanol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.17-8.29 (m, 1H), 7.22-7.28 (m, 2H), 7.01-7.12 (m, 3H), 5.98-6.03 (m, 1H), 4.98 (d, J=7.30 Hz, 1H), 4.27-4.45 (m, 3H), 4.00-4.13 (m, 3H), 3.83-3.92 (m, 1H), 3.33-3.50 (m, 3H), 2.99 (s, 0.5H), 2.97 (s, 0.5H), 2.49-2.63 (m, 1H), 2.31-2.64 (m, 1H); LC/MS [M+H]=588.0. Example 43 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((1-(hydroxymethyl)-cyclopropyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with (1-aminocyclopropyl)methanol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.18 (s, 1H), 7.22-7.32 (m, 2H), 7.02-7.11 (m, 3H), 6.01 (d, J=7.36 Hz, 1H), 4.96 (d, J=7.36 Hz, 1H), 4.32 (t, J=3.24 Hz, 1H), 4.02-4.14 (m, 2H), 3.76 (bs, 2H), 3.46 (d, J=15.04 Hz, 1H), 3.38 (d, J=15.04 Hz, 1H), 2.99 (s, 1H), 0.88-1.03 (m, 4H); LC/MS [M+H]=588.0. Example 44 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((1-hydroxycyclopropyl)-methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with 1-(aminomethyl)cyclopropanol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.18 (s, 1H), 7.08-7.25 (m, 5H), 5.98-6.02 (d, J=7 Hz, 1H), 4.93-4.97 (m, 2H), 4.30 (bs, 1H), 3.98-4.10 (m, 2H), 3.71 (bs, 2H), 3.39-3.51 (m, 2H), 3.00-3.13 (s 1H), 0.71-0.80 (m, 4H); LC/MS [M+H]=588.2. Example 45 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclobutylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with cyclobutylmethanamine and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.19 (s, 1H), 7.24-7.27 (m, 2H), 7.05-7.07 (m, 3H), 5.98-6.01 (d, J 8 Hz, 1H), 4.93-4.95 (d, J 7 Hz, 1 Hz), 4.31-4.33 (bs, 1H), 4.01-4.10 (m, 2H), 3.58 (s, 2H), 3.39-3.48 (m, 2H), 3.00 (s, 1H), 2.66-2.71 (m, 1H), 2.12-2.15 (m, 2H) 1.84-1.97 (m, 4H); LC/MS [M+H]=586.2. Example 46 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(3-(hydroxymethyl)azetidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with azetidin-3-ylmethanol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.09 (s, 1H), 7.05-7.25 (m, 5H), 6.00-6.02 (d, J 7 Hz, 1H), 4.93-4.97 (m, 1H), 4.33 (bs, 1H), 3.96-4.11 (m, 2H), 3.78 (bs, 2H), 3.36-3.40 (m, 6H), 2.99 (bs, 2H); LC/MS [M+H]=588.2. Example 47 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(3-hydroxy-3-methylazetidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with 3-methylazetidin-3-ol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.30 (s, 1H), 7.08-7.25 (m, 5H), 6.00-6.03 (d, J 7 Hz, 1H), 4.98-5.01 (m, 1H), 4.33 (bs, 4H), 4.01-4.09 (m, 2H), 3.73 (bs, 1H), 3.35-3.47 (m, 2H), 2.98 (s, 1H), 1.57 (s, 3H); LC/MS [M+H]=588.2. Example 48 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((3-hydroxycyclobutyl)-(methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 34 above but substituting propanolamine with 3-(methylamino)cyclobutanol and followed by ester hydrolysis with LiOH provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.26 (s, 1H), 6.96-7.28 (m, 5H), 6.02-6.04 (d, J 7 Hz, 1H), 5.27 (bs, 1H), 4.98-5.02 (m, 1H), 4.33 (bs, 2H), 3.95-4.15 (m, 3H), 3.36-3.51 (m, 4H), 2.99 (s, 1H), 2.67-2.69 (m, 2H), 2.22-2.25 (m, 2H); LC/MS [M+H]=602.2. Examples 49 & 50 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-ethoxy-3-oxo-2-(3-(trifluoromethyl)-benzyl)propanoic acid & 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(3-(trifluoromethyl)benzyl)malonic acid Proceeding as described in Example 8 above but substituting furan with (3-trifluoro-methyl)benzene provided the title compounds both as white solid by preparative reversed-phase HPLC purification. 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydro-furan-2-yl)methoxy)-3-ethoxy-3-oxo-2-(3-(trifluoromethyl)-benzyl)propanoic acid:1H NMR (CD3OD, 300 MHz) δ 8.33 (bs, 1H), 7.51-7.54 (d, J=8 Hz, 2H), 7.38-7.40 (d, J=6 Hz, 1H), 7.21-7.26 (t, J=7 Hz, 1H), 6.00-6.04 (m, 1H), 4.98-5.02 (dt, J=4.7, 52 Hz, 1H), 4.36 (bs, 1H), 4.02-4.22 (m, 4H), 3.45-3.58 (m, 2H), 3.02-3.12 (d, J=29 Hz, 1H), 1.18-1.24 (m, 3H); LC/MS [M+H]=614.2. 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydro-furan-2-yl)methoxy)-2-(3-(trifluoromethyl)benzyl)malonic acid:1H NMR (CD3OD, 300 MHz) δ 8.30 (s, 1H), 7.52-7.54 (d, J=9 Hz, 2H), 7.37-7.39 (d, J=7 Hz, 1H), 7.23-7.25 (t, J=7 Hz, 1H), 6.01-6.03 (d, J=7 Hz, 1H), 4.97-5.00 (d, J=7 Hz, 1H), 4.37 (bs, 1H), 4.12-4.14 (m, 2H), 3.44-3.57 (m, 2H), 3.01 (s, 1H); LC/MS [M+H]=586.2. Example 51 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(3-chlorobenzyl)malonic acid Proceeding as described in Example 8 above but substituting furan with 3-chloro-benzene provided the title compound as a white solid by preparative reversed-phase HPLC purification. 1H NMR (CD3OD, 300 MHz) δ 8.42 (s, 1H), 7.27 (bs, 1H), 7.14-7.15 (d, J=6 Hz, 1H), 7.02-7.06 (m, 2H), 6.02-6.05 (d, J=8 Hz, 1H), 5.03-5.06 (d, J=7 Hz, 1H), 4.35-4.39 (m, 2H), 3.39-3.49 (m, 2H), 3.01 (s, 1H), 2.48-2.54 (t, J 8 Hz, 1H) 2.22-2.32 (m, 1H); LC/MS [M+H]=552.1. Example 52 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(3-methoxybenzyl)malonic acid Proceeding as described in Example 8 above but substituting furan with 3-methoxy-benzene provided the title compound as a white solid by preparative reversed-phase HPLC purification. 1H NMR (CD3OD, 300 MHz) δ 8.40 (s, 1H), 6.96-7.02 (t, J 8 Hz, 1H), 6.82 (bs, 2H), 6.59-6.62 (m, 1H), 6.02-6.04 (d, J=7 Hz, 1H), 5.01-5.04 (d, J=8 Hz, 1H), 4.35-4.39 (m, 2H), 3.54 (s, 3H), 3.45-3.50 (m, 1H), 2.97 (s, 1H), 2.48-2.54 (t, J 8 Hz, 1H) 2.22-2.32 (m, 1H); LC/MS [M+H]=548.1. Example 53 Synthesis of 2-([1,1′-biphenyl]-4-ylmethyl)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 8 above but substituting furan with 3-biphenyl provided the title compound as a white solid by preparative reversed-phase HPLC purification. 1H NMR (CD3OD, 300 MHz) δ 8.23 (s, 1H), 7.28-7.43 (m, 9H), 5.99-6.02 (d, J=7 Hz, 1H), 4.96-4.98 (d, J=7 Hz, 1H), 4.35 (s, 1H), 4.08-4.15 (m, 2H), 3.41-3.57 (m, 2H), 3.04 (s, 1H): LC/MS [M+H]=594.2. Example 54 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-((2′-carboxy-[1,1′-biphenyl]-4-yl)methyl)malonic acid Proceeding as described in Example 8 above but substituting furan with 3-methyl [1,1′-biphenyl]-2-carboxylate provided the title compound as a white solid by preparative reversed-phase HPLC purification. 1H NMR (CD3OD, 300 MHz) δ 8.27 (s, 1H), 7.71-7.73 (d, J 7 Hz, 1H), 7.29-7.48 (m, 4H), 7.14-7.16 (d, J=8 Hz, 1H), 7.08-7.10 (d, J=8 Hz, 2H), 5.99-6.01 (d, J=8 Hz, 1H), 4.88-4.91 (m, 1H), 4.30 (bs, 1H), 4.03-4.12 (m, 2H), 3.39-3.57 (m, 2H), 2.99 (s, 1H); LC/MS [M+H]=638.2. Example 55 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Diethyl 2-(((2R,3R,4S,5R)-5-(N6,N6-bis-Boc-2-chloro-9H-purin-9-yl)-3-((tert-butoxycarbonyl)oxy)-4-fluorotetrahydrofuran-2-yl)methoxy)malonate (100 mg, 0.13 mmol) was dissolved in EtOAc (2 mL). The solution was purged three times with Argon gas and followed by careful addition of palladium on carbon (20 mg, 10 wt %). The resulting slurry was purged three times with Argon gas and then placed under H2(1 atm in a balloon). The reaction was held for 70 h at ambient temperature. The suspension was filtered through diatomaceous earth, washed with EtOAc (3×2 mL). The filtrate was concentrated to an oil which was then dissolved in DCM (1 mL) and followed by addition of TFA (100 μL). The resulting solution was held overnight before it was concentrated. The pale yellow oil residue was dissolved in THF (1 mL) and cooled at 0° C. To this reaction mixture was added 4M NaOH (100 μL) and the reaction was allowed to warm to ambient temperature over 14 h before it was concentrated. The crude residue was purified by preparative reversed-phase HPLC to provide the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.90 (s, 1H), 6.09-6.11 (d, J=7 Hz, 1H), 4.69-4.72 (d, J=8 Hz, 1H), 4.64 (bs, 1H), 4.19 (s, 1H), 3.83 (bs, 3H), 1.88-1.95 (m, 3H), 1.06-1.11 (t, J 7 Hz, 3H); LC/MS [M+H]=432.2. Example 56 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: A mixture of diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydro-furan-2-yl)methoxy)malonate (1.0 mmol, 549 mg) and palladium on carbon (100 mg, 10 wt %) in EtOH (5 mL) and EtOAc (5 mL) under an atmosphere of H2was stirred for 24 h before it was filtered through diatomaceous earth, rinsed with EtOAc (3×5 mL). The filtrate was concentrated and purified via flash silica gel column chromatography to provide diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethyltetrahydrofuran-2-yl)methoxy)malonate. Steps 2-3: Proceeding as described in Example 7 above but substituting diethyl 2-(pyridin-4-ylmethyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate with diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethyltetrahydrofuran-2-yl)methoxy)-malonate and followed by ester hydrolysis provided the title compound as a white solid via preparative reversed-phase HPLC purification. 1H NMR (CD3OD, 300 MHz) δ 8.46 (s, 1H), 7.18-7.20 (m, 2H), 7.04-7.09 (m, 3H), 6.01-6.04 (d, J 8 Hz, 1H), 4.63-4.66 (d, J=8 Hz, 1H), 4.21 (bs, 1H), 3.76-3.96 (m, 2H), 3.39-3.52 (m, 2H), 1.76-1.83 (m, 2H), 0.98-1.03 (t, J=7 Hz, 3H); LC/MS [M+H]=522.2 Example 57 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-vinyltetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: A mixture of diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetra-hydrofuran-2-yl)methoxy)malonate (525 mg, 0.96 mmol) and Lindlar catalyst (105 mg, 5 wt %) in EtOH (5 mL) and EtOAc (5 mL) under an atmosphere of H2was stirred for 24 h before it was filtered through diatomaceous earth, rinsed with EtOAc (3×5 mL). The filtrate was concentrated and purified via flash silica gel column chromatography to provide diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-vinyltetrahydrofuran-2-yl)methoxy)malonate. Steps 2-3: Proceeding as described in Example 7 above but substituting diethyl 2-(pyridin-4-ylmethyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate with diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-vinyltetrahydrofuran-2-yl)methoxy)-malonate and followed by ester hydrolysis provided the title compound as a white solid via preparative reversed-phase HPLC purification. 1H NMR (CD3OD, 300 MHz) δ 8.44 (s, 1H), 7.21-7.22 (m, 2H), 7.06-7.11 (m, 3H), 6.14-6.23 (m, 1H), 6.08-6.10 (d, J=8 Hz, 1H), 5.55-5.61 (m, 1H), 5.25-5.29 (m, 1H), 4.81 (s, 1H), 4.14 (bs, 1H), 3.91-3.94 (m, 1H), 3.62-3.65 (d, J 9 Hz, 1H), 3.50-3.55 (d, J 15 Hz, 1H), 3.39 (s, 1H); LC/MS [M+H]=520.1. Example 58 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a solution of an anomeric mixture of diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (175 mg, 0.319 mmol) in dry dichloroethane (3.5 mL) was added 5-chloro-3H-imidazo[4,5-b]pyridine (65 mg, 0.422 mmol) and followed by addition of N,O-bis(trimethylsilyl)acetamide (BSA) (0.28 mL, 1.12 mmol) via syringe. The mixture was heated at 95° C. under argon atmosphere for 1 h before it was cooled to ambient temperature and followed by addition of TMSOTf (0.09 mL, 0.494 mmol) via syringe. The resulting mixture was heated at 95° C. for 5 h before it was allowed to cool and diluted with water (30 mL) and extracted with EtOAc (30 mL). The organic phase was washed successively with equal volumes of saturated NaHCO3solution and brine. The aqueous phases was further extracted with EtOAc (2×30 mL). The combined organic phase was dried (MgSO4), filtered and concentrated. The crude residue was purification by preparative TLC (55% EtOAc in hexanes) to provide less polar diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(5-chloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-((trimethyl-silyl)ethynyl)tetrahydrofuran-2-yl)methoxy)malonate (44 mg) as a viscous oil and the desired diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(5-chloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (22 mg) as a viscous oil. Step 2: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(5-chloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (22 mg, 0.034 mmol) in THF (0.6 mL) was added a solution of 1N aq. LiOH (0.24 mL). Additional 1N aq. LiOH (0.58 mL) was applied over a period of 2 days with a combination of periodically sonication and stirring. The reaction mixture was concentrated and diluted with water (10 mL) and EtOAc (10 mL). The reaction mixture was cooled at 0° C. and acidified to pH˜3 with 1N aq. HCl. The layers were separated and the aq. layer was further extracted with EtOAc (2×10 mL). The combined organic layer was dried over MgSO4, filtered and concentrated. The crude residue was purified by preparative reversed-phase HPLC to provide the title compound as a off-white solid. 1H NMR (CD3OD, 300 MHz): δ 8.63 (bs, 1H), 8.01 (d, J=8.14 Hz, 1H), 7.34 (d, J=8.14 Hz, 1H), 7.29-7.23 (m, 2H), 7.07-6.99 (m, 3H), 6.21 (d, J=7.33 Hz, 1H), 5.04 (d, J=7.33 Hz, 1H), 4.34 (t, J=3.02 Hz, 1H), 4.14-4.03 (m, 2H), 3.48 (d, J=12.53 Hz, 1H), 3.37 (d, J=12.53 Hz, 1H), 2.99 (s, 1H); LC/MS [M+H]=502.0. Example 59 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: A suspension of sodium hydride (60%, mineral dispersion; 1.91 g, 47.7 mmoL) in anhydrous THF (150 mL) was cooled to 0° C. and treated with a second solution of (3aR,5R,6R,6aR)-5-(((tert-butyldimethylsilyl)oxy)methyl)-6-ethynyl-2,2-dimethyltetrahydro-furo[2,3-d][1,3]dioxol-6-ol (10 g, 30.4 mmoL) in THF (50 mL) over 15 minutes. After stirring 15 mins at 0° C., the mixture was warmed to room temperature and stirred for an additional 1.5 h. The mixture was then cooled back to 0° C. and treated with MOMCl (6.81 mL, 80.7 mmoL, 2.6 eq) was added dropwise. Once the addition was complete, the cooling bath was removed and stirring was continued for 2 h at room temperature. The reaction was quenched by the slow addition of saturated aqueous ammonium chloride (50 mL), washed with water and extracted with ethyl acetate (100 mL). The organic phase was dried over sodium sulfate, filtered, and concentrated. The crude product was purified via silica gel chromatography (30% ethyl acetate in hexanes) to afford tert-butyl-(((3aR,5R,6R,6aR)-6-ethynyl-6-(methoxy-methoxy)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)dimethylsilane (9.95 g, 88% yield) as a white solid. Step 2: A solution of the above alcohol (9.8 g, 26.3 mmoL) in anhydrous THF (100 mL) was cooled to 0° C. and treated with 1N solution of tetrabutylammonium fluoride in THF (37 mL, 36.8 mmoL, 1.4 eq) over 15 minutes. After the addition is complete, the reaction was warmed to room temperature and stirred for 3 h. When the reaction was complete (3 h), the volatiles were concentrated affording a viscous residual oil which was dissolved in dichloromethane (5 mL), loaded directly onto a silica gel column (˜300 cc) and purified via silica gel chromatography, eluting with hexanes to 50% Ethyl acetate in hexanes to afford ((3aR,5R,6R,6aR)-6-ethynyl-6-(methoxymethoxy)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]-dioxol-5-yl)methanol (6.18 g, 91% yield) as a white solid. Step 3: A solution of the above alcohol (175 mg, 0.678 mmoL) in anhydrous benzene (8 mL) and 1-ethyl 3-(prop-1-en-1-yl) 2-diazomalonate (188 mg, 0.949 mmoL) was treated with rhodium(II) acetate (5.8 mg, 0.013 mmoL, 0.02 eq) and warmed to 60-65° C. for 2 h. Once complete, the solution is concentrated, dissolved in dichloromethane (1.5 mL) and loaded directly onto a silica gel column (˜100 cc) and purified via silica gel chromatography, eluting with (0-50% ethyl acetate in hexanes) to afford 1-ethyl 3-prop-1-en-1-yl 2-(((3aR,5R,6R,6aR)-6-ethynyl-6-(methoxymethoxy)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]-dioxol-5-yl)methoxy)malonate (252 mg, 87%) (mixture of isomers) as a pale-yellow oil. Step 4: While under nitrogen, a solution of malonate from the previous step (250 mg, 0.583 mmol) and benzyl bromide (0.42 mL, 3.5 mmol, 6 eq) in anhydrous DMF (8 mL) was treated with and cesium carbonate (760 mg, 2.33 mmol) and stirred at room temperature for 4 h. Once complete, the reaction was filtered through a celite pad, washed with water and extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate, filtered, and concentrated. The residual oil was dissolved in dichloromethane (2 mL), loaded onto a silica gel column (˜100 cc) eluting with 30% ethyl acetate in hexanes to afford 1-ethyl 3-prop-1-en-1-yl 2-benzyl-2-(((3aR,5R,6R,6aR)-6-ethynyl-6-(methoxymethoxy)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (261 mg, 86%) (mixture of isomers) as a pale yellow oil. Step 5: While under nitrogen, a water cooled (14-17° C.) solution of the acetonide from the last step (500 mg, 0.964 mmoL) in acetic acid (3.9 mL) was treated with acetic anhydride (0.965 mL, 10.3 mmoL, 10.7 eq) and concentrated sulfuric acid (410 uL, 0.326 mmoL, 0.34 eq). The resulting solution was stirred for 4 h, diluted with water and extracted with ethyl acetate. The combined organic solution washed with sodium bicarbonate (aqueous, saturated; 100 mL), dried (Na2SO4), filtered, and concentrated in vacuo. The residual oil was dissolved in dichloromethane (2 mL), and purified on a Biotage flash chromatography system, eluted with hexanes to 50% ethyl acetate in hexanes. A diastereomeric mixture of 1-ethyl 3-prop-1-en-1-yl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (420 mg, 8%) (separable anomers via silica gel chromatography, 40 G silica gel column) was isolated as a clear oil. Step 6: A suspension of 2,6-dichloroadenine (143 mg, 0.76 mmol, 1.01 eq) and N,O-bis(trimethylsilyl)acetamide (0.24 mL, 0.97 mmol, 1.29 eq) in anhydrous acetonitrile (5 mL) was treated with a second solution of 1-ethyl 3-((E)-prop-1-en-1-yl) 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)-methoxy)malonate (143 mg, 0.75 mmol) in anhydrous acetonitrile (15 mL), followed by dropwise addition of trimethylsilyl trifluoromethanesulfonate (0.18 mL, 1.0 mmol, 1.33 eq). After the addition was complete, the reaction was warmed to 50° C. for 18 h, then cooled to room temperature. (Reaction begins a pale-yellow color and after 4 h turns to a transparent amber). Once complete, saturated aqueous sodium bicarbonate was added, and the mixture was stirred for 10 minutes. The crude product was then extracted with ethyl acetate (3×30 mL) and the combined organic layer is dried (Na2SO4), filtered, and concentrated. The residue was dissolved in dichloromethane and purified on a Biotage flash chromatography system, eluted with hexanes to 50% ethyl acetate in hexanes to give 1-ethyl 3-prop-1-en-1-yl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (as a mixture of isomers) as a white solid (400 mg, 77% yield). Step 7: A solution of 1-ethyl 3-prop-1-en-1-yl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (80 mg, 0.116 mmoL) in anhydrous dioxane (2 mL) was cooled to 0° C. was treated with DIPEA (30 μL, 0.174 mmoL, 1.5 eq) and benzylamine (13 μL, 0.116 mmoL, 1 eq). Once the addition was complete, the solution was warmed to room temperature with continued stirring overnight (18H). The mixture was diluted with ethyl acetate (50 mL) and washed with water (20 mL). The organic layer was dried (Na2SO4), filtered, and concentrated. The crude product was purified via Biotage flash chromatography, eluting with 50% ethyl acetate in hexanes to give 1-ethyl 3-prop-1-en-1-yl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (as a mixture of isomers, ˜1:1) as a white solid (75 mg, 85% yield). Step 8: A solution of 1-ethyl 3-prop-1-en-1-yl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate from the last step (70 mg, 0.092 mmoL) in THF (1 mL) was treated with a LiOH solution (31 mg, 1.35 mmoL, 15 eq; in 1 mL water) and stirred overnight. The resulting solution was acidified with 2N HCl to pH 3 and the resulting suspension was stirred for 10 min., then filtered, washed with cold water and dried. The title compound was isolated as a white solid (50 mg, 89% yield). 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 7.98 (s. 1H), 7.25-6.84 (m, 10H), 5.85 (d, J=6.4 Hz, 1H), 4.63 (s, 2H), 4.54 (d, J=6.4 Hz, 1H), 4.17 (t, J=3.2 Hz, 1H), 3.88 (qd, J=10.3, 3.3 Hz, 2H), 3.36-3.16 (m, 2H), 2.49 (s, 1H). HPLC: 9.97 min, 97.0%. ESI-MS (m/z): [M]+calcd for C29H26ClN5O8, 608.15; found 608.1. Example 60 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((tetrahydro-2H-pyran-4-yl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except tetrahydro-2H-pyran-4-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.09 (s, 1H), 7.18-7.12 (m, 2H), 7.01 (dd, J=12.1, 7.2 Hz, 3H), 5.90 (d, J=6.3 Hz, 1H), 4.57 (d, J=6.3 Hz, 1H), 4.25 (t, J=3.1 Hz, 2H), 4.00-3.89 (m, 4H), 3.51 (td, J=11.6, 2.2 Hz, 2H), 3.42-3.330 (m, 2H), 2.52 (s, 1H), 1.94 (d, J=13.0 Hz, 2H), 1.57 (td, J=11.2, 3.5 Hz, 2H). ESI-MS (m/z): [M]+calcd for C27H28ClN5O9, 602.16; found 602.5. Example 61 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-(diethylamino)ethyl)-amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except N1,N1-diethylethane-1,2-diamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, DMSO-66): 9.01 (s, 1H), 8.75 (s, 1H), 8.49 (s, 1H), 7.23-7.03 (m, 5H), 6.16 (s, 1H), 5.96 (d, J=7.0 Hz, 1H), 5.84 (d, J=6.9 Hz, 1H), 4.59 (t, J=7.1 Hz, 1H), 4.13 (dd, J=6.7, 2.7 Hz, 1H), 3.83-3.55 (m, 4H), 3.53 (s, 1H), 3.30-3.15 (m, 6H), 3.06-2.97 (m, 2H), 1.26-1.12 (m, 6H). ESI-MS (m/z): [M]+calcd for C28H33ClN6O8, 617.20; found 617.5. Example 62 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((R)-1-phenylethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (R)-1-phenylethan-1-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.25 (s, 1H), 7.44-7.27 (m, 4H), 7.23-6.94 (m, 6H), 5.91 (dd, J=6.2, 1.7 Hz, 1H), 5.46 (d, J=8.0 Hz, 1H), 4.58 (d, J=6.2 Hz, 1H), 4.25 (t, J=2.4 Hz, 1H), 3.98-3.83 (m, 2H), 3.32 (dd, J=6.3, 1.7 Hz, 2H), 2.48 (d, J=1.7 Hz, 1H), 1.59 (dd, J=6.9, 1.7 Hz, 3H). ESI-MS (m/z): [M]+calcd for C30H28ClN5O8, 622.16; found 622.1. Example 63 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((S)-1-phenylethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (S)-1-phenylethan-1-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.11 (s, 1H), 7.40-7.28 (m, 4H), 7.23-6.93 (m, 6H), 5.90 (d, J=5.8 Hz, 1H), 5.45 (s, 1H), 4.52 (d, J=5.8 Hz, 1H), 4.35-4.22 (m, 1H), 3.99 (t, J=2.4 Hz, 2H), 3.42-3.25 (m, 2H), 2.50 (s, 1H), 1.58 (d, J=6.9 Hz, 3H). ESI-MS (m/z): [M]+calcd for C30H28ClN5O8, 622.16; found 622.2. Example 64 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((tetrahydrofuran-3-yl)-amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except tetrahydrofuran-3-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.10 (s, 1H), 7.19-6.92 (m, 5H), 5.90 (d, J=6.3 Hz, 1H), 4.72 (s, 1H), 4.58 (d, J=6.3 Hz, 1H) 4.25 (t, J=3.1 Hz, 1H), 3.94 (tp, J=7.0, 4.0, 3.3 Hz, 3H), 3.70 (dt, J=9.4, 3.6 Hz, 2H), 3.43-3.24 (m, 2H), 2.52 (s, 1H), 2.29 (dq, J=13.2, 7.6 Hz, 1H), 1.93 (s, 1H). ESI-MS (m/z): [M]+calcd for C26H26ClN5O9, 588.14; found 588.3. Example 65 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((S)-3-hydroxypyrrolidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (S)-pyrrolidin-3-ol was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.10 (s, 1H), 7.22-6.92 (m, 5H), 5.92 (d, J=5.4 Hz, 1H), 4.55-4.48 (m, 2H), 4.29 (t, J=3.7 Hz, 1H), 4.25-3.85 (m, 4H), 3.80-3.65 (m, 2H), 3.35-3.24 (m, 2H), 2.51 (s, 1H), 2.08-1.98 (m, 2H). ESI-MS (m/z): [M]+calcd for C26H26ClN5O9, 588.14; found 588.2. Example 66 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(ethyl(methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except N-methylethanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.05 (s, 1H), 7.24-7.02 (m, 5H), 5.92 (d, J=4.8 Hz, 1H), 4.46 (dd, J=4.8, 1.3 Hz, 1H), 4.32 (dd, J=4.9, 3.2 Hz, 1H), 4.05 (dd, J=8.5, 4.0 Hz, 2H), 3.44-3.30 (m, 2H), 3.10-2.85 (m, 7H), 2.53 (s, 1H), 1.22 (t, J=7.1 Hz, 3H). ESI-MS (m/z): [M]+calcd for C25H26ClN5O8, 560.15; found 560.5. Example 67 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-fluorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (2-fluorophenyl)methanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.29 (s, 1H), 7.47-7.36 (m, 1H), 7.26-6.94 (m, 8H), 5.94 (d, J=6.2 Hz, 1H), 4.82-4.76 (m, 2H), 4.60 (d, J=6.2 Hz, 1H), 4.35-4.23 (m, 1H), 3.98-3.84 (m, 2H), 3.44-3.30 (m, 2H), 2.46 (s, 1H). ESI-MS (m/z): [M]+calcd for C29H25ClFN5O8, 626.14; found 626.7. Example 68 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((4-fluorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (4-fluorophenyl)methanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.49 (s, 1H), 7.37 (dd, J=8.4, 5.3 Hz, 2H), 7.24-6.89 (m, 7H), 5.97 (d, J=6.3 Hz, 1H), 4.72 (s, 2H), 4.63 (d, J=6.3 Hz, 1H), 4.7-4.28 (m, 1H), 4.06-3.87 (m, 2H), 3.46-3.26 (m, 2H), 2.54 (s, 1H). ESI-MS (m/z): [M]+calcd for C29H25ClFN5O8, 626.14; found 626.4. Example 69 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((R)-2,3-dihydro-1H-inden-1-yl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (R)-2,3-dihydro-1H-inden-1-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.30 (s, 1H), 7.29-6.97 (m, 9H), 5.95 (d, J=6.2 Hz, 1H), 5.81 (t, J=7.1 Hz, 1H), 4.59 (d, J=6.3 Hz, 1H), 4.25 (d, J=3.1 Hz, 1H), 3.94-3.73 (m, 2H), 3.40-3.31 (m, 2H), 3.07 (ddd, J=14.0, 8.7, 4.8 Hz, 1H), 2.89 (dt, J=15.8, 7.7 Hz, 1H), 2.69-2.58 (m, 1H), 2.45 (s, 1H), 2.00 (dd, J=13.6, 7.1 Hz, 1H). ESI-MS (m/z): [M]+calcd for C31H28ClN5O8, 634.16; found 634.8. Example 70 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(phenethylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 2-phenylethan-1-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.14 (s, 1H), 7.26-6.91 (m, 10H), 5.91 (d, J=6.2 Hz, 1H), 4.57 (d, J=6.2 Hz, 1H), 4.27 (t, J=3.2 Hz, 1H), 3.96 (t, J=4.0 Hz, 2H), 3.78 (m, 2H), 3.45-3.29 (m, 2H), 2.93 (t, J=7.3 Hz, 2H), 2.52 (s, 1H). ESI-MS (m/z): [M]+calcd for C30H28ClN5O8, 622.16; found 622.5. Example 71 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except methylamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.18 (s, 1H), 7.29-7.20 (m, 2H), 7.04 (dd, J=5.1, 1.9 Hz, 3H), 5.99 (d, J=7.4 Hz, 1H), 4.99 (d, J=7 Hz, 1H), 4.30 (t, J=3.3 Hz, 1H), 4.11-4.01 (m, 2H), 3.49-3.34 (m, 2H), 3.06 (s, 3H), 2.98 (s, 1H). ESI-MS (m/z): [M]+calcd for C23H22ClN5O8, 532.12; found 532.1. Example 72 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((S)-2,3-dihydro-1H-inden-1-yl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (S)-2,3-dihydro-1H-inden-1-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.14 (s, 1H), 7.41-7.13 (m, 6H), 7.04 (t, J=3.5 Hz, 3H), 6.01 (d, J=7.4 Hz, 1H), 5.88-5.76 (m, 1H), 4.99 (d, J=7.4 Hz, 1H), 4.32 (t, J=3.3 Hz, 1H), 4.06 (dd, J=5.5, 3.3 Hz, 2H), 3.53-3.34 (m, 2H), 3.13-3.05 (m, 1H), 3.00 (s, 1H), 2.99-2.87 (m, 1H), 2.67 (dd, J=12.5, 4.2 Hz, 1H), 2.02 (dd, J=12.8, 7.7 Hz, 1H). ESI-MS (m/z): [M]+ calcd for C31H28ClN5O8, 634.16; found 634.5. Example 73 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(ethylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except ethylamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.16 (s, 1H), 7.40-7.19 (m, 2H), 7.04 (dd, J=5.1, 1.9 Hz, 3H), 5.98 (d, J=7.4 Hz, 1H), 4.97 (d, J=7.4 Hz, 1H), 4.30 (T, J=3.3 Hz, 1H), 4.14-3.98 (m, 2H), 3.57 (d, J=7.8 Hz, 2H), 3.49-3.33 (m, 2H), 2.98 (s, 1H), 1.28 (t, J=7.2 Hz, 3H). ESI-MS (m/z): [M]+calcd for C24H24ClN5O8, 546.13; found 546.1. Example 74 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(6-((S)-sec-butylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (S)-butan-2-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.13 (s, 1H), 7.32-7.15 (m, 2H), 7.14-6.98 (m, 3H), 5.98 (d, J=7.4 Hz, 1H), 4.97 (d, J=7.4 Hz, 1H), 4.31 (t, J=3.2 Hz, 1H), 4.24 (s, 1H), 4.14-3.97 (m, 2H), 3.53-3.33 (m, 2H), 2.99 (s, 1H), 1.63 (q, J=7.2 Hz, 2H), 1.27 (d, J=6.5 Hz, 3H), 0.97 (t, J=7.4 Hz, 3H). ESI-MS (m/z): [M]+calcd for C26H28ClN5O8, 574.16; found 574.1. Example 75 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)-(methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 1-cyclopropyl-N-methylmethanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.15 (s, 1H), 7.36-7.18 (m, 2H), 7.03 (dd, J=5.2, 2.0 Hz, 3H), 6.00 (d, J=7.3 Hz, 1H), 4.97 (d, J=7.3 Hz, 1H), 4.30 (dd, J=4.1, 2.8 Hz, 1H), 4.07 (qd, J=10.2, 3.5 Hz, 2H), 3.52-3.31 (m, 7H), 2.98 (s, 1H), 1.20-1.12 (m, 1H), 0.54 (dd, J=8.2, 1.8 Hz, 2H), 0.45-0.30 (m, 2H). ESI-MS (m/z): [M]+calcd for C27H28ClN5O8, 586.16; found 586.9. Example 76 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(6-((carboxymethyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 2-amino-N,N-dimethylacetamide was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.21 (s, 1H), 7.38-7.14 (m, 2H), 7.12-6.98 (m, 3H), 6.11 (d, J=7.4 Hz, 1H), 4.96 (d, J=7.3 Hz, 1H), 4.30 (q, J=4.7, 4.0 Hz, 2H), 4.09-4.02 (m, 2H), 3.54-3.33 (m, 2H), 2.97 (s, 1H). ESI-MS (m/z): [M]−calcd for C24H22ClN5O10, 574.11; found 574.1. Example 77 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-chlorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (2-chlorophenyl)-methanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.17 (s, 1H), 7.43 (m, 2H), 7.28 (m, 3H), 7.22 (m, 1H), 7.00 (m, 3H), 5.99 (d, J=7.4 Hz, 1H), 4.98 (d, J=7.4 Hz, 1H), 4.82 (m, 2H), 4.30 (s, 1H), 4.05 (s, 2H), 3.42 (m, 2H), 2.98 (s, 1H). LC-MS: m/z=597 (M-CO2H); m/z=292 (M-ribose fragment). Example 78 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((pyridin-4-ylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except pyridin-4-ylmethanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 9.08 (bs, 1H), 8.73 (d, J=5.7 Hz, 2H), 8.51 (s, 1H), 7.68 (d, J=5.6 Hz, 2H), 7.22 (m, 2H), 7.06 (m, 3H), 5.82 (d, J=7.7 Hz, 1H), 4.93 (d, J=6.9 Hz, 1H), 4.87 (d, J=7.6 Hz, 1H), 4.20 (m, 1H), 4.03 (m, 2H), 3.86 (m, 2H), 3.62 (s, 1H), 3.27 (m, 2H). HPLC: Room temperature=5.74 min, 97.7%. LC-MS: m/z=610 (M+); m/z=261 (M-ribose fragment). Example 79 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-morpholino-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except morpholine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.20 (s, 1H), 7.22 (m, 2H), 7.01 (m, 3H), 6.00 (d, J=7.6 Hz, 1H), 4.99 (d, J=7.6 Hz, 1H), 4.30 (m, 1H), 4.22 (m, 4H), 4.04 (m, 2H), 3.78 (m, 4H), 3.47 (m, 2H), 2.99 (s, 1H). HPLC: 8.16 min, 98.2%. LC-MS: m/z=588 (M+), 544 (M-CO2H), m/z=240 (M-ribose fragment). Example 80 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-(azepan-1-yl)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except azepane was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.16 (s, 1H), 7.22 (m, 2H), 7.00 (m, 3H), 5.99 (d, J=7.6 Hz, 1H), 4.98 (d, J=7.6 Hz, 1H), 4.29 (m, 3H), 4.07 (m, 2H), 3.85 (m, 2H), 3.40 (m, 2H), 2.97 (s, 1H), 1.85 (m, 4H), 1.59 (m, 4H). HPLC: 9.67 min, 98.1%. LC-MS: m/z=600 (M+), m/z=556 (M-CO2H), m/z=252 (M-ribose fragment). Example 81 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclobutyl(methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except N-methylcyclobutanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.21 (s, 1H), 7.21 (dd, J=7.0, 2.6 Hz, 2H), 6.99 (m, 3H), 5.99 (d, J=7.3 Hz, 1H), 5.74 (br s, 1H), 4.96 (d, J=7.2 Hz, 1H), 4.28 (dd, J=4.1, 2.8 Hz, 1H), 4.06 (dd, J=10.2, 4.2 Hz, 1H), 4.01 (dd, J=10.2, 2.9 Hz, 1H), 3.45-3.31 (m, 5H), 2.96 (s, 1H), 2.35 (q, J=10.0 Hz, 2H), 2.23 (m, 2H), 1.87-1.63 (m, 2H); HPLC: 9.45 min, 98.9%. LC-MS: m/z=587 (M+H), 543 (M-CO2H), m/z=238 (M-ribose fragment). Example 82 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropyl(methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except N-methylpropan-2-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.17 (s, 1H), 7.21 (dd, J=7.3, 2.2 Hz, 2H), 7.03-6.94 (m, 3H), 5.98 (d, J=7.3 Hz, 1H), 4.97 (d, J=7.3 Hz, 1H), 4.28 (dd, J=4.0, 2.8 Hz, 1H), 4.04 (qd, J=10.2, 3.5 Hz, 2H), 3.45-3.31 (m, 6H), 2.96 (s, 1H), 1.25 (d, J=6.8 Hz, 6H); HPLC: 9.08 min, 99.9%. LC-MS: m/z=575 (M+H), 531 (M-CO2H), m/z=226 (M-ribose fragment). Example 83 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except cyclopropanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.14 (s, 1H), 7.23 (dd, J=6.7, 2.8 Hz, 2H), 7.01 (m, 3H), 5.97 (d, J=7.4 Hz, 1H), 4.95 (d, J=7.4 Hz, 1H), 4.28 (t, J=3.3 Hz, 1H), 4.10-3.97 (m, 2H), 3.49-3.30 (m, 2H), 2.99 (m, 1H), 2.96 (s, 1H), 0.92-0.78 (m, 2H), 0.66-0.54 (m, 2H); HPLC: 7.83 min, 99.1%. LC-MS: m/z=559 (M+H). Example 84 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((pyridin-3-ylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except pyridin-3-ylmethanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.87 (s, 1H), 8.70 (d, J=5.6 Hz, 1H), 8.56 (d, J=8.1 Hz, 1H), 8.22 (s, 1H), 7.97 (dd, J=8.1, 5.7 Hz, 1H), 7.21 (dd, J=6.5, 3.0 Hz, 2H), 7.07-6.90 (m, 3H), 5.97 (d, J=7.4 Hz, 1H), 4.96 (d, J=7.4 Hz, 1H), 4.92 (m, 2H), 4.29 (t, J=3.4 Hz, 1H), 4.04 (qd, J=10.2, 3.4 Hz, 2H), 3.38 (m, 2H), 2.97 (s, 1H); HPLC: 5.82 min, 99.9%. LC-MS: m/z=261 (M-ribose fragment). Example 85 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((3-fluorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (3-fluorophenyl)methanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.19 (s, 1H), 7.32 (td, J=8.0, 5.9 Hz, 1H), 7.24-7.16 (m, 3H), 7.12 (dd, J=9.8, 2.3 Hz, 1H), 7.06-6.78 (m, 4H), 5.98 (d, J=7.3 Hz, 1H), 4.94 (d, J=7.3 Hz, 1H), 4.74 (m, 2H), 4.29 (t, J=3.3 Hz, 1H), 4.03 (qd, J=10.3, 3.5 Hz, 2H), 3.47-3.26 (m, 2H), 2.97 (s, 1H); HPLC: 9.04 min, 99.5%. LC-MS: m/z=627 (M+H), 583 (M-CO2H), m/z=278 (M-ribose fragment). Example 86 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((3-chlorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (3-chlorophenyl)methanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.18 (s, 1H), 7.40 (s, 1H), 7.34-7.05 (m, 5H), 6.98 (m, 3H), 5.97 (d, J=7.4 Hz, 1H), 4.95 (d, J=7.4 Hz, 1H), 4.72 (m, 2H), 4.29 (t, J=3.3 Hz, 1H), 4.09-3.87 (m, 2H), 3.46-3.31 (m, 2H), 2.97 (s, 1H); HPLC: 9.45 min, 98.3%. LC-MS: m/z=643 (M+H), 598 (M-CO2H), m/z=294 (M-ribose fragment). Example 87 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((4-chlorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (4-chlorophenyl)methanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.17 (s, 1H), 7.36 (d, J=8.6 Hz, 2H), 7.31 (d, J=8.5 Hz, 1H), 7.21 (dd, J=6.8, 2.8 Hz, 2H), 7.02-6.96 (m, 3H), 5.97 (d, J=7.3 Hz, 1H), 4.94 (d, J=7.3 Hz, 1H), 4.71 (m, 2H), 4.29 (t, J=3.3 Hz, 1H), 4.03 (qd, J=10.2, 3.4 Hz, 2H), 3.46-3.30 (m, 2H), 2.96 (s, 1H); HPLC: 9.50 min, 98.7%. LC-MS: m/z=643 (M+H), 598 (M-CO2H), m/z=294 (M-ribose fragment). Example 88 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-(azetidin-1-yl)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except azetidine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.25 (s, 1H), 7.24-7.08 (m, 2H), 7.10-6.85 (m, 3H), 5.97 (d, J=7.4 Hz, 1H), 4.98 (d, J=7.3 Hz, 1H), 4.43 (m, 4H), 4.29 (dd, J=4.1, 2.8 Hz, 1H), 4.07 (dd, J=10.2, 4.2 Hz, 1H), 4.00 (dd, J=10.2, 2.9 Hz, 1H), 3.42 (d, J=15.0 Hz, 1H), 3.34 (d, J=12.3 Hz, 2H), 2.95 (s, 1H), 2.51 (q, J=7.7 Hz, 2H); HPLC: 7.58 min, 99.5%. LC-MS: m/z=558 (M+H), 554 (M-CO2H), m/z=210 (M-ribose fragment). Example 89 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(dimethylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except dimethylamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.17 (s, 1H), 7.22 (dd, J=7.5, 2.0 Hz, 2H), 7.05-6.89 (m, 3H), 5.98 (d, J=7.2 Hz, 1H), 4.96 (d, J=7.3 Hz, 1H), 4.28 (dd, J=4.0, 2.9 Hz, 1H), 4.04 (qd, J=10.2, 3.5 Hz, 2H), 3.55-3.31 (m, 8H), 2.95 (s, 1H); HPLC: 8.16 min, 99.9%. LC-MS: m/z=546 (M+H), 502 (M-CO2H), m/z=198 (M-ribose fragment). Example 90 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(pyrrolidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except pyrrolidine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.21 (s, 1H), 7.21 (dd, J=7.3, 2.3 Hz, 2H), 7.03-6.76 (m, 3H), 5.98 (d, J=7.3 Hz, 1H), 4.98 (d, J=7.3 Hz, 1H), 4.29 (dd, J=4.2, 2.8 Hz, 1H), 4.15-3.86 (m, 4H), 3.66 (m, 2H), 3.41 (d, J=15.0 Hz, 1H), 3.34 (d, J=15.0 Hz, 1H), 2.95 (s, 1H), 2.02 (m, 4H); HPLC: 8.42 min, 95.6%. LC-MS: m/z=573 (M+H), 529 (M-CO2H), m/z=224 (M-ribose fragment). Example 91 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopentylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except cyclopentanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.09 (s, 1H), 7.23 (dd, J=6.7, 2.9 Hz, 2H), 7.01 (m, 3H), 5.96 (d, J=7.4 Hz, 1H), 4.94 (d, J=7.4 Hz, 1H), 4.47 (m, 1H), 4.28 (t, J=3.3 Hz, 1H), 4.04 (m, 2H), 3.43 (d, J=14.9 Hz, 1H), 3.34 (d, J=14.9 Hz, 1H), 2.96 (s, 1H), 2.19-1.98 (m, 2H), 1.78 (m, 2H), 1.72-1.64 (m, 2H), 1.62-1.52 (m, 2H); HPLC: 9.06 min, 98.9%. LC-MS: m/z=587 (M+H), 543 (M-CO2H), m/z=238 (M-ribose fragment). Example 92 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except cyclopropylmethanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.14 (s, 1H), 7.18 (d, J=6.9 Hz, 2H), 7.12-6.68 (m, 3H), 5.92 (d, J=6.1 Hz, 1H), 4.58 (d, J=6.1 Hz, 1H), 4.28 (t, J=3.2 Hz, 1H), 3.98 (qd, J=10.3, 3.2 Hz, 2H), 3.50-3.04 (m, 4H), 2.55 (s, 1H), 1.08 (m, 1H), 0.52 (m, 2H), 0.26 (m, 2H); HPLC: 8.52 min, 97.7%. LC-MS: m/z=572 (M+H), 528 (M-CO2H), m/z=224 (M-ribose fragment). Example 93 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except propan-2-amine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.05 (s, 1H), 7.23-7.12 (m, 2H), 7.09-6.78 (m, 3H), 5.91 (d, J=6.2 Hz, 1H), 4.59 (d, J=6.2 Hz, 1H), 4.37 (m, 1H), 4.28 (m, 1H), 3.42 (d, J=14.8 Hz, 1H), 3.35-3.22 (m, 3H), 2.56 (s, 1H), 1.24 (d, J=6.5 Hz, 6H); HPLC: 8.39 min, 98.9%. LC-MS: m/z=560 (M+H), 516 (M-CO2H), m/z=212 (M-ribose fragment). Example 94 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-hydroxy-2-methylpropyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 1-amino-2-methylpropan-2-ol was used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.16 (s, 1H) 7.24-7.22 (m, 2H) 7.03-7.01 (m, 3H) 5.97 (d, J=7.4 Hz, 1H) 4.95 (d, J=7.4 Hz, 1H) 4.28 (t, J=3.2 Hz, 1H) 4.05-4.03 (m, 2H) 3.72-3.69 (m, 2H) 3.48-3.31 (m, 2H) 2.97 (s, 1H) 1.24 (s, 6H). ESI-MS (m/z): [M]−calcd for C26H28ClN5O9589.98; found 588.4. Example 95 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((S)-1-hydroxypropan-2-yl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (S)-2-aminopropan-1-ol was used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.18 (s, 1H) 7.24-7.22 (m, 2H) 7.03-7.01 (m, 3H) 5.97 (d, J=7.4 Hz, 1H) 4.93 (d, J=7.4 Hz, 1H) 4.42-4.35 (m, 1H) 4.32-4.26 (m, 1H) 4.04 (d, J=3.2 Hz, 2H) 3.62-3.60 (m, 2H) 3.48-3.32 (m, 2H) 2.96 (s, 1H) 1.29 (d, J=6.7 Hz, 3H). ESI-MS (m/z): [M]−calcd for C25H26ClN5O9575.96; found 574.2. Example 96 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(diethylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except diethylamine was used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.15 (s, 1H) 7.23-7.20 (m, 2H) 7.00-6.98 (m, 3H) 5.98 (d, J=7.3 Hz, 1H) 4.97 (d, J=7.3 Hz, 1H) 4.29 (m, 1H) 4.15-3.83 (m, 6H) 3.43-3.33 (m, 2H) 2.96 (s, 1H) 1.24 (t, J 7.0 Hz, 6H). ESI-MS (m/z): [M]−calcd for C26H28ClN589573.98; found 572.3. Example 97 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-hydroxyethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 2-aminoethan-1-ol was used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.20 (s, 1H) 7.24-7.22 (m, 2H) 7.04-7.02 (m, 3H) 5.97 (d, J=7.4 Hz, 1H) 4.91 (d, J=7.3 Hz, 1H) 4.29 (t, J=3.4 Hz, 1H)) 4.06-3.98 (m, 2H) 3.76-3.73 (m, 2H) 3.66-3.63 (m, 2H) 3.43-3.33 (m, 2H) 2.96 (s, 1H). ESI-MS (m/z): [M]−calcd for C24H24ClN5O9561.93; found 560.2. Example 98 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(((R)-1-hydroxypropan-2-yl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (R)-2-aminopropan-1-ol was used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.12 (s, 1H) 7.24-7.22 (m, 2H) 7.03-7.01 (m, 3H) 5.96 (d, J=7.4 Hz, 1H) 4.95 (d, J=7.4 Hz, 1H) 4.42-4.32 (m, 1H) 4.28 (t, J=3.3 Hz, 1H) 4.08-4.00 (m, 2H) 3.66-3.58 (m, 2H) 3.45-3.33 (m, 2H) 2.97 (s, 1H) 1.27 (d, J=6.7 Hz, 3H). ESI-MS (m/z): [M]−calcd for C25H26ClN5O9575.96; found 574.2. Example 99 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopropyl(2-hydroxyethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 2-(cyclopropylamino)ethan-1-ol was used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.23 (s, 1H) 7.24-7.22 (m, 2H) 7.04-7.01 (m, 3H) 6.02 (d, J=7.4 Hz, 1H) 4.96 (d, J=7.3 Hz, 1H) 4.30-4.28 (m, 1H) 4.10-4.04 (m, 4H) 3.78 (t, J=5.9 Hz, 2H) 3.44-3.32 (m, 2H) 3.23-3.19 (m, 1H) 2.96 (s, 1H) 1.01-0.97 (m, 2H) 0.78-0.74 (m, 2H). Example 100 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((R)-3-hydroxypyrrolidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (R)-pyrrolidin-3-ol was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.24 (s, 1H), 7.23 (d, J=7.2 Hz, 2H), 7.06-6.96 (m, 3H), 6.01 (d, J=7.3 Hz, 1H), 5.01 (s, 1H), 4.56 (d, J=26.1 Hz, 1H), 4.33-4.31 (m, 1H), 4.20 (d, J=12.5 Hz, 1H), 4.10-4.00 (m, 3H), 3.78 (m, 2H), 3.50-3.35 (m, 2H), 2.97 (s, 1H), 2.14-2.05 (m, 2H). Example 101 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((S)-3-(hydroxymethyl)-pyrrolidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (S)-pyrrolidin-3-ylmethanol was used in place of benzylamine in step 7. 1H NMR (400 MHz, DMSO-d6) δ 8.37 (s, 1H), 8.34 (s, 1H), 7.15 (m, 2H), 7.01 (m, 3H), 6.16 (br s, 1H), 5.96 (br s, 1H), 5.81 (d, J=7.5 Hz, 1H), 4.82 (d, J=7.4 Hz, 1H), 4.18-4.08 (m, 1H), 3.94 (m, 2H), 3.71 (m, 2H), 3.50 (m, 2H), 3.20 (s, 4H), 3.13 (s, 1H), 2.33 (m, 1H), 2.04 (m, 1H), 1.98-1.89 (m, 1H), 1.77 (m, 1H), 1.65 (m, 1H); HPLC: 7.22 min, 97.7%. Example 102 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((R)-3-(hydroxymethyl)-pyrrolidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (R)-pyrrolidin-3-ylmethanol was used in place of benzylamine in step 7. 1H NMR (400 MHz, DMSO-d6) δ 8.37 (s, 1H), 8.34 (s, 1H), 7.21-7.11 (m, 2H), 7.01 (m, 3H), 6.16 (br s, 1H), 5.96 (br s, 1H), 5.81 (d, J=7.5 Hz, 1H), 4.82 (d, J=7.5 Hz, 1H), 4.44 (m, 1H), 4.25-4.07 (m, 3H), 3.95 (m, 2H), 3.87-3.64 (m, 3H), 3.51 (m, 2H), 3.20 (s, 1H), 2.37-2.27 (m, 1H), 2.04 (m, 1H), 1.98-1.86 (m, 1H), 1.76 (m, 1H), 1.66 (m, 1H); HPLC: Rt=7.20 min, 97.0%. Example 103 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((R)-2-(hydroxymethyl)-pyrrolidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (R)-pyrrolidin-2-ylmethanol was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.21 (s, 1H), 7.28-7.16 (m, 2H), 7.08-6.87 (m, 3H), 5.99 (d, J=7.3 Hz, 1H), 4.97 (d, J=7.3 Hz, 1H), 4.41 (m, 1H), 4.28 (m, 1H), 4.12-3.93 (m, 4H), 3.72 (m, 2H), 3.42 (d, J=15.0 Hz, 1H), 3.33 (d, J=15.0 Hz, 2H), 2.96 (s, 1H), 2.05 (m, 4H); HPLC: 7.73 min, 98.2%; ESI-MS: m/z=254 (M-ribose fragment). Example 104 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((S)-2-(hydroxymethyl)-pyrrolidin-1-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except (S)-pyrrolidin-2-ylmethanol was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.22 (s, 1H), 7.22 (m, 2H), 7.07-6.88 (m, 3H), 5.99 (d, J=7.3 Hz, 1H), 4.98 (d, J=7.3 Hz, 1H), 4.44 (m, 1H), 4.29 (dd, J=4.0, 2.9 Hz, 1H), 4.07 (m, 2H), 4.05 (qd, J=10.2, 3.5 Hz, 2H), 3.77 (dd, J=11.0, 4.2 Hz, 1H), 3.66 (dd, J=11.1, 6.5 Hz, 1H), 3.42 (d, J=15.1 Hz, 1H), 3.33 (d, J=15.1 Hz, 1H), 2.95 (s, 1H), 2.07 (m, 4H); HPLC: 7.77 min, 99.3%; ESI-MS: m/z=642 (M+ACN). Example 105 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopropyl(methyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except N-methylcyclopropanamine was used in place of benzylamine in step 7. 1H NMR (400 MHz, CD3OD) δ 8.36 (s, 1H), 7.26-7.12 (m, 2H), 7.02 (m, 3H), 6.03 (d, J=7.3 Hz, 1H), 4.97 (d, J=7.3 Hz, 1H), 4.30 (dd, J=3.9, 2.8 Hz, 1H), 4.19-3.97 (m, 2H), 3.57-3.29 (m, 5H), 3.21 (m, 1H), 2.96 (s, 1H), 1.05-0.85 (m, 2H), 0.83-0.62 (m, 2H); HPLC: Rt=8.41 min, 98.6%; ESI-MS: m/z=224 (M-ribose fragment). Example 106 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(methylamino)-3-oxopropanoic acid Step 1: A stirred solution of [(3aR,5R,6R,6aR)-6-ethynyl-6-(methoxymethoxy)-2,2-dimethyl-tetrahydro-2H-furo[2,3-d][1,3]dioxol-5-yl]methanol (480 mg, 1.87 mmol) and methyl 2-diazo-2-(methylcarbamoyl)acetate (440 mg, 2.8 mmol, prepared according to literature:European Journal of Organic Chemistry,2014 (24), 5302-5311) in anhydrous benzene (20 mL) under nitrogen was treated with rhodium tetraacetate (16 mg, 0.04 mmol) and heated to 60° C. for 4 h. The resulting mixture was cooled to room temperature and concentrated. The resulting oil was dissolved in dichloromethane, loaded onto a silica gel column eluting with 10-100% ethyl acetate in hexane to afford methyl 2-{[(3aR,5R,6R,6aR)-6-ethynyl-6-(methoxymethoxy)-2,2-dimethyl-tetrahydro-2H-furo[2,3-d][1,3]dioxol-5-yl]methoxy}-2-(methylcarbamoyl)acetate (455 mg, 63% yield) as a diastereomeric pair. Step 2: A solution of methyl 2-{[(3aR,5R,6R,6aR)-6-ethynyl-6-(methoxymethoxy)-2,2-dimethyl-tetrahydro-2H-furo[2,3-d][1,3]dioxol-5-yl]methoxy}-2-(methylcarbamoyl)acetate (450 mg, 116 mmol) and benzyl bromide (0.97 mL, 8.13 mmol) in anhydrous DMF (4 mL) was treated with cesium carbonate (757 mg, 2.32 mmol). The reaction mixture was stirred at room temperature overnight. The reaction was quenched with water (10 mL), diluted with diethyl ether (70 mL) and washed with saturated aqueous sodium chloride (50 mL) twice. The organic layer was dried with NaSO4and concentrated to give a yellow oil. The residue was purified by silica gel column and eluted with 10-70% ethyl acetate in hexane to afford a diastereomeric mixture methyl 2-benzyl-2-(((3aR,5R,6R,6aR)-6-ethynyl-6-(methoxy-methoxy)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-3-(methylamino)-3-oxopropanoate (500 mg, 90% yield) as a clear oil as. Step 3: While under nitrogen, an ice cooled stirred solution of methyl 2-benzyl-2-(((3aR,5R,6R,6aR)-6-ethynyl-6-(methoxymethoxy)-2,2-dimethyltetrahydrofuro[2,3-d][1,3]-dioxol-5-yl)methoxy)-3-(methylamino)-3-oxopropanoate (500 mg, 1.05 mmol) in acetic acid (4 mL) was treated with acetic anhydride (1 mL, 11.15 mmol) and concentrated sulfuric acid (0.02, 0.35 mmol). The reaction solution was slowly warmed to room temperature. After 4 hours, reaction was diluted with water and extracted with ethyl acetate (80 mL each). The organic solution was washed with saturated sodium bicarbonate (100 mL), dried over Na2SO4and concentrated. The residual oil was dissolved in dichloromethane and purified by flash chromatography to provide (3R,4R,5R)-5-(((2-benzyl-1-methoxy-3-(methylamino)-1,3-dioxopropan-2-yl)oxy)methyl)-4-ethynyltetrahydrofuran-2,3,4-triyl triacetate as an anomer/diastereomeric mixture. Step 4: A suspension of 2-6-dichloroadenine (48 mg, 0.25 mmol) and N,O-bis(trimethylsilyl)-acetamide (0.08 mL, 0.32 mmol) in anhydrous acetonitrile (7 mL) was treated with a second solution of (3R,4R,5R)-5-(((2-benzyl-1-methoxy-3-(methylamino)-1,3-dioxopropan-2-yl)oxy)methyl)-4-ethynyltetrahydrofuran-2,3,4-triyl triacetate (130 mg, 0.25 mmol) in anhydrous acetonitrile (10 mL), followed by dropwise addition of trimethylsilyl trifluoromethanesulfonate (0.06 mL, 0.33 mmol). Once the addition was complete, the reaction was heated to 50° C. for 18 h, cooled to room temperature and quenched with saturated sodium bicarbonate solution (80 mL). After stirring for a 5 minutes the solution was extracted with ethyl acetate (3×80 mL), dried over Na2SO4and concentrated. The residue was dissolved in dichloromethane and purified by flash chromatography to provide (2R,3R,4R,5R)-2-(((2-benzyl-1-methoxy-3-(methylamino)-1,3-dioxopropan-2-yl)oxy)methyl)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (135 mg, 83% yield) as a foamy solid (diastereomer pair). Step 5: While under nitrogen, a solution of (2R,3R,4R,5R)-2-(((2-benzyl-1-methoxy-3-(methylamino)-1,3-dioxopropan-2-yl)oxy)methyl)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyl-tetrahydrofuran-3,4-diyl diacetate (70 mg, 0.11 mmol) in dioxane (2 mL) was cooled to 0° C. and treated with diisopropylethylamine (0.03 mL, 0.16 mmol) and cyclopropylamine (0.01 mL, 0.13 mmol), and warmed to room temperature with stirring overnight. The reaction mixture was diluted with ethyl acetate (80 mL), washed with water (50 mL) and saturated aqueous sodium chloride (50 mL). The organic layer was dried over Na2SO4then filtered and concentrated. The residue was dissolved in dichloromethane and purified by flash column chromatography to afford (2R,3R,4R,5R)-2-(((2-benzyl-1-methoxy-3-(methylamino)-1,3-dioxopropan-2-yl)oxy)methyl)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (70 mg, 95% yield) as an off-white solid (diastereomeric pair). Step 6: To solution of (2R,3R,4R,5R)-2-(((2-benzyl-1-methoxy-3-(methylamino)-1,3-dioxo-propan-2-yl)oxy)methyl)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (70 mg, 0.10 mmol) in THF (1 mL) was treated with LiOH (24 mg, 1.02 mmol) in water (1 mL) and stirred at room temperature overnight. Reaction pH was adjusted to 4-5 using cold 2 N HCl. Upon precipitation, the suspension was stirred for another 10 min. The solid was collected, washed with cold water and dried to provide 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydro-furan-2-yl)methoxy)-3-(methylamino)-3-oxopropanoic acid (45 mg, 75% yield) as an off-white solid (diastereomeric pair). 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.01 (s, 0.45H), 7.48 (s, 0.55H), 7.22-7.03 (m, 5H), 5.86 (dd, J=4.7, 3.0 Hz, 1H), 4.43 (dd, J=8.5, 4.6 Hz, 1H), 4.31-4.06 (m, 2H), 3.84-3.72 (m, 1H), 3.45-3.21 (m, 4H), 2.62 (d, J=40.9 Hz, 1H), 2.46 (d, J=8.1 Hz, 3H), 1.05 (dq, J=8.0, 3.7 Hz, 1H), 0.51 (ddd, J=8.1, 4.0, 1.6 Hz, 2H), 0.31-0.18 (m, 2H). ESI-MS (m/z): [M]+calcd for C27H29ClN6O7, 585.18; found 585.9. Example 107 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(methylamino)-3-oxopropanoic acid The title compound was prepared in a manner analogous to that set forth in Example 106, except benzylamine was used in place of cyclopropylamine in step 5. 1H NMR (400 MHz, CDCl3/CD3OD=5:1) δ 8.03 (s, 0.46H), 7.52 (d, J=2.7 Hz, 0.54H), 7.42-7.04 (m, 10H), 5.89 (t, J=4.3 Hz, 1H), 4.73 (m, 2H), 4.44 (dd, J=6.0, 4.5 Hz, 1H), 4.33-4.15 (m, 2H), 3.79 (dd, J=10.6, 4.5 Hz, 1H), 3.45-3.27 (m, 2H), 2.62 (d, J=36.6 Hz, 1H), 2.48 (d, J=13.2 Hz, 3H). ESI-MS (m/z): [M]+calcd for C30H29ClN6O7, 621.18; found 621.4. Example 108 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((2-chlorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-ethoxy-3-oxopropanoic acid The title compounds is prepared in a manner analogous to that set forth in Example 59, except only 5 equivalents of lithium hydroxide are used in step 8. 1H NMR (400 MHz, CD3OD) δ 8.16 (s, 1H), 7.42 (m, 2H), 7.26 (m, 2H), 7.21 (m, 2H), 6.99 (m, 3H), 5.97 (d, J=7.6 Hz, 1H), 4.96 (d, J=7.6 Hz, 1H), 4.82 (m, 2H), 4.28 (m, 1H), 4.03 (m, 2H), 3.75 (m, 2H), 3.38 (m, 2H), 3.22 (m, 2H), 2.97 (s, 1H) 1.29 (t, J=7.6 Hz, 3H). HPLC: 9.43 min, 96.2%. LC-MS: m/z=642, 597 (M-CO2H); m/z=292 (M-ribose fragment). Example 109 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((pyridin-4-ylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-ethoxy-3-oxopropanoic acid The title compounds is prepared in a manner analogous to that set forth in Example 59, except only 5 equivalents of lithium hydroxide are used in step 8. Compound was isolated as ˜2:1 mixture of diastereomers. 1H NMR (400 MHz, DMSO-d6) of major isomer: δ 8.73 (d, J=5.9 Hz, 2H), 8.15 (s, 0.61H), 7.98 (d, J=5.9 Hz, 2H), 7.23 (m, 2H), 7.06 (m, 3H), 6.00 (m, 1H), 5.00 (m, 2H), 4.10 (m, 4H), 3.39 (m, 2H), 3.12 (s, 0.80H), 1.17 (m, 3H). 1H NMR (DMSO-d6) of minor isomer: δ 8.73 (d, J=5.9 Hz, 2H), 8.31 (s, 0.32H), 7.98 (d, J=5.9 Hz, 2H), 7.23 (m, 2H), 7.06 (m, 3H), 6.00 (m, 1H), 5.00 (m, 2H), 4.10 (m, 4H), 3.39 (m, 2H), 3.01 (s, 0.41H), 1.17 (m, 3H). HPLC: 6.25 min, 94.8%. LC-MS: m/z=638 (M+), 594 (M-CO2H), m/z=259 (M-ribose fragment). Example 110 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-((5-methylisoxazol-3-yl)methyl)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 3-(bromomethyl)-5-methylisoxazole was used in place of benzyl bromide in step 4 and propan-2-amine is used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.46 (s, 1H) 5.99 (d, J 7.6 Hz, 1H) 5.95 (s, 1H) 5.07 (d, J=7.6 Hz, 1H) 4.41-4.38 (m, 1H) 4.31-4.29 (m, 1H) 4.07-4.05 (m, 1H) 3.99-3.88 (m, 1H) 3.50-3.38 (m, 2H) 2.95 (s, 1H) 2.05 (s, 3H) 1.27 (d, J=6.5 Hz, 6H). ESI-MS (m/z): [M]+calcd for C23H25ClN6O9564.93; found 566.1. Example 111 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-((4-fluorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-((5-methylisoxazol-3-yl)methyl)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 3-(bromomethyl)-5-methylisoxazole was used in place of benzyl bromide in step 4 and (4-fluorophenyl)methanamine is used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.62-8.58 (br s, 1H) 7.41-7.38 (m, 2H) 7.07-7.01 (m, 2H) 6.01 (d, J=7.4 Hz, 1H) 5.96 (s, 1H) 5.09 (d, J=7.3 Hz, 1H) 4.82-4.71 (m, 2H) 4.31-4.29 (m, 1H) 4.06 (dd, J=10.0, 3.8 Hz, 1H) 3.97 (dd, J=10.1, 3.1 Hz, 1H) 3.44-3.32 (m, 2H) 3.18 (s, 3H) 2.96 (s, 1H). Example 112 Synthesis of 2-(benzo[d]thiazol-2-ylmethyl)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((4-fluorobenzyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except 2-(bromomethyl)benzo[d]thiazole is used in place of benzyl bromide in step 4 and (4-fluorophenyl)methanamine was used in place of benzylamine in step 7. 1H-NMR (400 MHz, CD3OD) δ 8.57-8.54 (br s, 1H) 7.74-7.70 (m, 2H) 7.45-7.41 (m, 2H) 7.23-7.20 (m, 1H) 7.12-7.16 (m, 1H) 7.09-7.04 (m, 2H) 5.94 (d, J=7.3 Hz, 1H) 5.15 (d, J=7.1 Hz, 1H) 4.77-4.72 (m, 2H) 4.35-4.32 (m, 1H) 4.20 (dd, J=10.1, 2.7 Hz, 1H) 4.11 (dd, J=10.1, 3.1 Hz, 1H) 3.99-3.86 (m, 2H) 2.77 (s, 1H). Example 113 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-oxo-3-(pyrrolidin-1-yl)propanoic acid The title compound was prepared in an analogous manner to Example 59 except ethyl 2-diazo-3-oxo-3-(pyrrolidin-1-yl)propanoate was used instead of 1-ethyl 3-(prop-1-en-1-yl) 2-diazomalonate in Step 3 and ammonia is used in place of benzylamine in Step 7. The product is isolated as a 55/45 mixture of diastereomers. Major diastereomer1H-NMR (400 MHz, CD3OD) δ 8.80 (s, 1H) 5.99 (d, J=7.4 Hz, 1H) 5.05 (d, J=7.4 Hz, 1H) 4.88 (s, 1H) 4.28-4.26 (m, 1H) 4.13 (dd, J=10.7, 2.5 Hz, 1H) 4.00 (dd, J=10.7, 3.9 Hz, 1H) 3.66-3.39 (m, 4H) 3.17 (s, 1H) 1.89-1.79 (m, 4H). Minor diasteromer 1H-NMR (400 MHz, CD3OD) δ 8.89 (s, 1H) 6.03 (d, J=7.4 Hz, 1H) 5.05 (d, J=7.4 Hz, 1H) 4.86 (s, 1H) 4.28-4.26 (m, 1H) 3.90-3.86 (m, 2H) 3.66-3.39 (m, 4H) 3.16 (s, 1H) 1.89-1.79 (m, 4H). ESI-MS (m/z): [M]−calcd for C19H21ClN6O7480.86; found 479.1. Example 114 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopentylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-oxo-3-(pyrrolidin-1-yl)propanoic acid The title compound was prepared in an analogous manner to Example 59 except ethyl 2-diazo-3-oxo-3-(pyrrolidin-1-yl)propanoate was used instead of 1-ethyl 3-(prop-1-en-1-yl) 2-diazomalonate in Step 3 and cyclopentanamine is used in place of benzylamine in Step 7. The product is isolated as a 55/45 mixture of diastereomers. Major diastereomer.1H NMR (400 MHz, CD3OD) δ 8.72 (s, 1H) 5.96 (d, J=7.4 Hz, 1H) 5.04 (d, J=7.4 Hz, 1H) 4.87 (s, 1H) 4.52-4.48 (m, 1H) 4.28-4.25 (m, 1H) 4.14 (d, J=10.5 Hz, 1H) 4.01 (dd, J=10.6, 5.2 Hz, 1H) 3.60-3.36 (m, 4H) 3.17 (s, 1H) 2.10-2.04 (m, 2H) 1.84-1.77 (m, 6H) 1.69-1.58 (m, 4H) Minor diastereomer1H NMR (400 MHz, CD3OD) δ 8.64 (s, 1H) 6.00 (d, J=7.4 Hz, 1H) 5.01 (d, J=7.4 Hz, 1H) 4.85 (s, 1H) 4.52-4.48 (m, 1H) 4.28-4.25 (m, 1H) 3.92-3.86 (m, 2H) 3.60-3.36 (m, 4H) 3.16 (s, 1H) 2.10-2.04 (m, 2H) 1.84-1.77 (m, 6H) 1.69-1.58 (m, 4H) (ESI-MS (m/z): [M]−calcd for C24H29ClN6O7548.98; found 547.3. Example 115 Synthesis of 3-amino-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopentylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-oxopropanoic acid Step 1: While under nitrogen, a suspension of 2,6-dichloroadenine (2.91 g, 15.4 mmoL, 1.01 eq) and N,O-bis(trimethylsilyl)acetamide (4.87 mL, 19.6 mmoL, 1.29 eq) in anhydrous acetonitrile (90 mL). Next, a solution of (2R,3R,4R,5R)-2,4-bis(acetyloxy)-5-{[(tert-butyldiphenylsilyl)oxy]methyl}-4-ethynyloxolan-3-yl acetate (8.2 g, 15.22 mmoL) in anhydrous acetonitrile (10 mL) was added, followed by dropwise addition of trimethylsilyl trifluoromethanesulfonate (3.67 mL, 20.3 mmoL, 1.33 eq). The reaction was warmed to 50° C. for 18 h, then cooled to room temperature. (Reaction begins a pale-yellow color and after 4 h turns to a transparent amber). Saturated aqueous sodium bicarbonate (10 mL), was added and the mixture was stirred for ten minutes. The resulting mixture was extracted with ethyl acetate (3×100 mL) and the combined organic layer was dried (Na2SO4), filtered, and concentrated. The residue was dissolved in dichloromethane/ethyl acetate (˜3 mL, 1:1), loaded onto a silica gel column (˜300 cc), and eluted with 0-30% ethyl acetate in hexanes to provide (2R,3R,4R,5R)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (8.2 g, 81%) as a white solid. Step 2: A solution of (2R,3R,4R,5R)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.6 g, 2.4 mmoL) in anhydrous THF (25 mL) was cooled to 0° C. and treated with acetic acid (0.192 mL, 3.36 mmoL, 1.4 eq) a 1 N solution of tetrabutylammonium fluoride in THF (3.36 mL, 3.36 mmoL, 1.4 eq). After the addition was complete, the reaction was warmed to room temperature with stirring for 3 h. The reaction mixture was concentrated, a purified via flash column chromatography (0 to 50% ethyl acetate in hexanes to afford (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (0.88 g, 86%) as a white foam. Step 3: While under nitrogen, a solution of ((2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (172 mg, 0.40 mmol) and ethyl 3-(aminooxy)-2-diazo-3-oxopropanoate (160 mg, 1.02 mmoL, 2.5 eq) in anhydrous toluene (3 mL) was treated with rhodium (II) acetate dimer (5 mg, 0.011 mmoL, 2.8 mol %) and warmed to 80° C. for 5 h. The reaction was concentrated, and purified via flash column chromatography (25-75% ethyl acetate in dichloromethane) to afford (2R,3R,4R,5R)-2-(((1-amino-3-ethoxy-1,3-dioxopropan-2-yl)oxy)methyl)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (101 mg, 45% in ˜85:15 mixture of diastereomers) as a colorless glass. Step 4: While under nitrogen, a solution of (2R,3R,4R,5R)-2-(((1-amino-3-ethoxy-1,3-dioxo-propan-2-yl)oxy)methyl)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (157 mg, 0.28 mmol) in dioxane (2.5 mL) was treated with diisopropylethyl-amine (0.100 mL, 0.61 mmoL, 2.2 eq) and cyclopentylamine (0.065 mL, 0.066 mmoL, 2.33 eq). After stirring at room temperature for 18 h, the mixture was diluted with ethyl acetate (20 mL), washed with water (10 mL), dried (Na2SO4), filtered, and concentrated to provide crude (2R,3R,4R,5R)-2-(((1-amino-3-ethoxy-1,3-dioxopropan-2-yl)oxy)methyl)-5-(2-chloro-6-(cyclopentylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate. Step 5: The resulting residue from the previous step was stirred in ammonium hydroxide and ethanol (1:1/v:v, 10 mL) at room temperature overnight. The mixture was concentrated, dissolved in THF (2.5 mL) and treated with lithium hydroxide (23 mg, 0.96 mmoL, 3.4 eq) dissolved in water (2.5 mL). The mixture was stirred at room temperature with occasional gentle heating for ˜3 h, then neutralized with TN HCl to pH-6 and concentrated in vacuo. The crude product was dissolved in water and purified by reverse phase HPLC and dried by lyophilization to provide the title compound (52 mg, 37% for 3 steps) as a voluminous white solid. 1H NMR (400 MHz, D2O) of major isomer: δ 8.30 (s, 1H), 5.81 (m, 1H), 4.88 (d, J=6.6 Hz, 1H), 4.80 (d, J=6.8 Hz, 1H), 4.24 (m, 2H), 4.20 (bs, 1H), 3.83 (m, 2H), 3.01 (s, 1H), 1.85 (m, 2H), 1.49 (m, 6H). 1H NMR (400 MHz, D2O) of minor isomer: δ 8.39 (s, 1H), 5.81 (m, 1H), 4.88 (d, J=6.6 Hz, 1H), 4.80 (d, J=6.8 Hz, 1H), 4.24 (m, 2H), 4.20 (bs, 1H), 3.83 (m, 2H), 2.99 (s, 1H), 1.85 (m, 2H), 1.49 (m, 6H). HPLC: Rt=7.08 min, 93.0%. ESI-MS for C20H23ClN6O7calcd. 494.13, found 493.2 (M−); ESI-MS for C10H11ClN5calcd. 236.07, found 236.0 (M-ribose fragment). Example 116 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopentylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonamide The title compound was prepared as a second product from step 5 in the synthesis of example 115. It was isolated as a voluminous white solid (8.5 mg, 6%). 1H NMR (400 MHz, DMSO-d6): 8.57 (s, 0.5H), 8.31 (s, 1H), 7.73 (s, 1H), 7.38 (m, 1.5H), 7.21 (m, 1.5H), 7.05 (m, 0.5H), 6.87 (m, 0.5H), 6.20 (m, 1H), 6.02 (d, J=7.0 Hz, 1H), 5.79 (d, J=7.8 Hz, 1H), 5.02 (s, 1H), 4.73 (s, 1H), 4.39 (m, 1H), 4.18 (m, 1H), 3.79 (m, 1H), 3.68 (s, 1H), 1.85 (m, 2H), 1.49 (m, 6H). HPLC: Rt=6.64 min, 97.1%. ESI-MS for C20H24ClN7O6calcd. 493.15, found 492.3 (M−); ESI-MS for C10H11ClN5calcd. 236.1, found 236.1 (M-ribose fragment). Example 117 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except methylamine was used in place of benzylamine in step 7 and step 4 is eliminated. 1H NMR (400 MHz, CD3OD) δ 8.88 (s, 1H), 6.05 (d, J=7.6 Hz, 1H), 5.03 (d, J=7.6 Hz, 1H), 4.62 (s, 1H), 4.26 (m, 1H), 4.04 (m, 1H), 3.90 (m, 1H), 3.11 (s, 1H), 3.07 (s, 3H). HPLC: Rt=5.92 min, 97.9%. ESI-MS for C16H16ClN5O8calcd. 441.07, found 442.5 (M+); ESI-MS for C6H5ClN5calcd. 182.02, found 184.2 (M-ribose fragment). Example 118 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid The title compound was prepared in a manner analogous to that set forth in Example 59, except step 4 was eliminated. 1H NMR (400 MHz, CD3OD) δ 8.87 (s, 1H), 7.39 (m, 2H), 7.33 (m, 2H), 7.26 (m, 1H), 6.06 (d, J=7.6 Hz, 1H), 5.02 (d, J=7.6 Hz, 1H), 4.76 (m, 2H), 4.62 (s, 1H), 4.26 (s, 1H), 4.03 (m, 1H), 3.90 (m, 1H), 3.10 (s, 1H). HPLC: Rt=7.83 min, 98.2%. ESI-MS for C22H20ClN5O8calcd. 517.10 found 516.7 (M−); ESI-MS for C12H9ClN5calcd. 258.05, found 258 (M-ribose fragment). Example 119 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-morpholino-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(phenylsulfonyl)acetic acid The title compound was prepared in an analogous manner to Example 59 except ethyl 2-diazo-2-(phenylsulfonyl)acetate was used instead of 1-ethyl 3-(prop-1-en-1-yl) 2-diazomalonate in Step 3, morpholine was used in place of benzylamine in Step 7 and Step 4 is eliminated. 1H NMR (400 MHz, DMSO-d6) of major isomer: δ 8.40 (s, 1H), 7.83 (m, 2H), 7.66 (m, 2H), 7.53 (m, 1H), 5.83 (m, 1H), 5.50 (d, J=7.8 Hz, 1H), 4.80 (d, J=7.8 Hz, 1H), 4.16 (m, 4H), 3.98 (m, 1H), 3.76 (m, 4H), 3.63 (s, 1H).1H NMR (400 MHz, DMSO-d6) of minor isomer: δ 8.41 (s, 1H), 7.83 (m, 2H), 7.66 (m, 2H), 7.53 (m, 1H), 5.83 (m, 1H), 5.50 (d, J=7.8 Hz, 1H), 4.66 (d, J=7.8 Hz, 1H), 4.16 (m, 4H), 3.98 (m, 1H), 3.76 (m, 4H), 3.61 (s, 1H). HPLC: Rt=7.83 (minor), 8.18 min (major), 99.6% (40:60). ESI-MS for C26H26ClN5O9calcd. 587.14, found 588 (M+); ESI-MS for C24H24ClN5O9S calcd. 593.10, found 592 (M−); ESI-MS for C23H23ClN5O7S calcd. 548.10, found 548 (M-CO2H); ESI-MS for C9H9ClN5O calcd. 238.05, found 240 (M-ribose fragment). Example 120 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopentylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(methylsulfonyl)acetic acid The title compound was prepared in an analogous manner to Example 59 except ethyl 2-diazo-2-(methylsulfonyl)acetate was used instead of 1-ethyl 3-(prop-1-en-1-yl) 2-diazomalonate in Step 3, cyclopentanamine is used in place of benzylamine in Step 7 and Step 4 is eliminated. 1H NMR (400 MHz, DMSO-d6) of major isomer: δ 8.47 (s, 1H), 5.85 (m, 1H), 5.36 (m, 1H), 5.01 (m, 1H), 4.86 (d, J=8.0 Hz, 1H), 4.42 (m, 2H), 4.33 (m, 1H), 4.25 (m, 2H), 4.14 (m, 1H), 4.00 (m, 1H), 3.37 (s, 1H), 3.05 (s, 3H), 1.97 (m, 2H), 1.72 (m, 2H), 1.57 (m, 4H).1H NMR (400 MHz, DMSO-d6) of minor isomer: δ 8.39 (s, 1H), 5.83 (m, 1H), 5.36 (m, 1H), 4.68 (d, J=8.0 Hz, 1H), 4.42 (m, 2H), 4.33 (m, 1H), 4.25 (m, 2H), 4.14 (m, 1H), 4.00 (m, 1H), 3.37 (s, 1H), 3.05 (s, 3H), 1.97 (m, 2H), 1.72 (m, 2H), 1.57 (m, 4H). HPLC: Rt=8.02 min, 98.4%. ESI-MS for C20H24ClN5O8S calcd. 529.10, found 530 (M+); ESI-MS for C10H11ClN5calcd. 236.07, found 238 (M-ribose fragment). Examples 121 & 122 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-carbamoyl-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzyl-3-ethoxy-3-oxopropanoic acid and 2-(((2R,3S,4R,5R)-5-(6-amino-2-carbamoyl-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(N,N′-bis(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (1.00 g, 1.17 mmol, 1 eq) in DMSO (10 mL) and H2O (2 mL) was added 1,4-diazabicyclo[2.2.2]octane (128 uL, 1.17 mmol, 1 eq) and NaCN (114.20 mg, 2.33 mmol, 2 eq). The solution was stirred at 60° C. for 3 h before it was diluted with water (15 mL) and extracted with ethyl acetate (3×15 mL). The combined organic layer was washed with water (50 mL), brine (50 mL), dried by Na2SO4, and filtered and concentrated. The crude residue was purified by Combi-flash (silica gel, 30-70% EtOAc in petroleum ether) to give diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(N,N′-bis(tert-butoxycarbonyl)-amino)-2-cyano-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (312 mg, 40% yield) as a yellow gum. Step 2: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(N,N′-bis(tert-butoxycarbonyl)amino)-2-cyano-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (50 mg, 75.23 umol, 1 eq) in DCM (1.7 mL) was added TFA (0.3 mL) at 0° C. The solution was stirred at 20° C. for 1 h before it was diluted with saturated aq. NaHCO3to adjust the pH to 9. The mixture was extracted with ethyl acetate (3×3 mL). The organic was concentrated to give crude diethyl 2-(((2R,3S,4R,5R)-5-(6-amino-2-cyano-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-benzylmalonate (45 mg) as a yellow gum. Step 3: To a solution of crude diethyl 2-(((2R,3S,4R,5R)-5-(6-amino-2-cyano-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-benzyl-malonate (65 mg, 115.14 umol, 1 eq) in MeCN (2 mL) was added 3,4,6,7,8,9-hexahydro-2H-pyrimido[1,2-a]-pyrimidine (TBD) (1 M aq., 461 uL, 4 eq). The reaction mixture was stirred at 20° C. for 18 h before it was concentrated. The crude residue was purified by preparative HPLC and the fractions were dried by lyophilization to give 2-(((2R,3S,4R,5R)-5-(6-amino-2-carbamoyl-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzyl-3-ethoxy-3-oxopropanoic acid (Example 121) (4.1 mg, 5% yield) as a white solid and 2-(((2R,3S,4R,5R)-5-(6-amino-2-carbamoyl-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid (Example 122) (2.2 mg, 3% yield) as a white solid. Example 121:1H NMR (400 MHz, CD3OD) δ ppm 8.19-8.39 (m, 1H) 7.26 (br d, J=5.88 Hz, 2H) 7.03-7.12 (m, 3H) 6.20 (dd, J=9.69, 7.19 Hz, 1H) 4.91-4.97 (m, 1H) 4.29 (br s, 1H) 3.98-4.24 (m, 4H) 3.41-3.53 (m, 1H) 3.31-3.40 (m, 1H) 2.97-3.08 (m, 1H) 1.18 (q, J=7.25 Hz, 3H); LC/MS [M+H]=555.1. Example 122:1H NMR (400 MHz, CD3OD) δ ppm 8.504 (s, 1H) 7.16-7.28 (m, 2H) 7.07 (br s, 3H) 6.20 (d, J=5.88 Hz, 1H) 4.92-4.99 (m, 1H) 4.37 (br s, 1H) 3.97 (br d, J=3.63 Hz, 2H) 3.32-3.48 (m, 2H) 3.01 (s, 1H); LC/MS [M+H]=527.0. Example 123 Synthesis of (1-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-methoxy-2-oxoethyl)phosphonic acid Step 1: A mixture of (2R,3R,4R,5R)-5-(6-(N,N′-bis(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (565 mg, 0.926 mmol, 1.0 eq) and methyl 2-diazo-2-(dimethoxyphosphoryl)acetate (247 mg, 1.20 mmol, 1.3 eq) was azeotroped twice with toluene and the resulting oil was re-dissolved in toluene (5.7 mL). The reaction solution was stirred at ambient temperature under argon atmosphere and fitted with a jacketed reflux condenser. Rhodium(II) acetate (0.185 mmol, 82 mg, 0.2 eq) was added and the reaction heated at 75° C. for 9 h before it was cooled to room temperature. The reaction mixture was concentrated and the resulting oil was purified by flash silica gel column chromatography to provide (2R,3R,4R,5R)-5-(6-(N,N′-bis(tert-butoxy-carbonyl)acetamido)-2-chloro-9H-purin-9-yl)-2-((1-(dimethoxyphosphoryl)-2-methoxy-2-oxoethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate. Steps 2-3: Deprotection of the product from the previous step was performed according to the procedure described for step 4 in Example 121. Aq. NaOH solution was used instead of KOEt that lead to the carboxylic acid. The title compound was isolated as a white solid from preparative reversed-phase HPLC. 1H NMR (CD3OD, 300 MHz) δ 8.08 (s, 1H), 6.06 (bs, 1H), 5.06-5.08 (d, J=5 Hz, 1H), 4.28 (s, 1H), 3.90-4.10 (m, 2H), 3.79 (s, 3H), 3.98 (bs, 2H), 3.13 (s, 1H); LC/MS [M+H]=478.2. Example 124 Synthesis of (1-(((2R,3S,4R,5R)-5-(2-chloro-6-morpholino-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-ethoxy-2-oxoethyl)phosphonic acid (1-(((2R,3S,4R,5R)-5-(2-chloro-6-morpholino-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-ethoxy-2-oxoethyl)phosphonic acid is prepared in a manner analogous to that set forth in Example 59, except ethyl 2-diazo-2-(diethoxyphos-phoryl)acetate in place of 2-diazomalonate in step 3, morpholine is used in place of benzylamine in step 7 and step 4 is eliminated. Compound was isolated as a 1:1 mixture of diastereomers. 1H NMR (400 MHz, DMSO-d6) of major isomer: δ 8.59 (s, 1H), 5.90 (m, 1H) 5.00 (d, J=7.0 Hz, 1H), 4.25 (m, 2H), 4.05 (m, 4H), 3.99 (m, 1H), 3.83 (m, 1H), 3.75 (m, 4H), 3.01 (s, 1H) 1.05 (t, J=7.1 Hz, 3H). 1H NMR (400 MHz, DMSO-d6) of minor isomer: δ 8.48 (s, 1H), 5.88 (m, 1H), 4.94 (d, J=6.9 Hz, 1H), 4.25 (m, 2H), 4.05 (m, 4H), 3.99 (m, 1H), 3.83 (m, 1H), 3.75 (m, 4H), 3.01 (s, 1H), 0.99 (t, J=7.1 Hz, 3H). HPLC: minor isomer=6.63 min; major isomer=6.65 min, 98.6%. LC-MS: m/z=562 (M+); m/z=240 (M-ribose fragment). Example 125 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-3-ethynyl-3,4-dihydroxy-5-(5-(trifluoromethyl)-3H-imidazo[4,5-b]pyridin-3-yl)tetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 15 above but substituting 6-amino-2-chloroadenine with 5-(trifluoromethyl)-3H-imidazo[4,5-b]pyridine provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.82 (bs, 1H), 8.23 (d, J=8 Hz, 1H), 7.78 (d, J=8 Hz, 1H), 7.29-7.26 (m, 2H), 7.03-7.00 (m, 3H), 6.32 (d, J=7 Hz, 1H), 5.10 (d, J=7 Hz, 1H), 4.37-4.36 (m, 1H), 4.08 (d, J=3 Hz, 2H), 3.42 (dd, J=15, 28 Hz, 2H), 2.98 (s, 1H); LC/MS [M+H]=535.2. Example 126 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-ylmethyl)malonic acid Proceeding as described in Example 15 above but substituting allyl bromide with 4-(chloromethyl)thiazole provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.76 (bs, 1H), 8.57 (bs, 1H), 7.36 (bs, 1H), 6.00 (d, J=7 Hz, 1H), 5.00-4.95 (m, 1H), 4.35 (bs, 1H), 4.08-4.04 (m, 2H), 3.66-3.64 (m, 2H), 2.98 (s, 1H); LC/MS [M+H]=524.9. Example 127 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-acetyl-6-amino-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: To a suspension of 6-amino-2-chloroadenine (3.0 g, 17.69 mmol, 1 eq) in DCM (60 mL) was added 4-DMAP (2.16 g, 17.69 mmol, 1 eq), TEA (21.48 g, 212.30 mmol, 29.55 mL, 12 eq) and (Boc)2O (30.89 g, 141.53 mmol, 8 eq). The suspension was stirred at 20° C. for 18 h before it was diluted with saturated aq. NH4Cl (100 mL), and extracted with ethyl acetate (2×100 mL). The combined organic layer was washed with brine (200 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by Combi-flash (silica gel, 0-20% EtOAc in petroleum ether) to give tert-butyl 6-(N,N′-bis(tert-butoxycarbonyl)amino)-2-chloro-9H-purine-9-carboxylate (904 mg, 11% yield) as a yellow gum. Step 2: To a solution of tert-butyl 6-(N,N′-bis(tert-butoxycarbonyl)amino)-2-chloro-9H-purine-9-carboxylate (900 mg, 1.92 mmol, 1 eq) in DMF (12 mL) was added Pd(PPh3)2Cl2(134.43 mg, 191.52 umol, 0.1 eq) and tributyl(1-ethoxyvinyl)stannane (832 uL, 2.46 mmol, 1.29 eq) under N2atmosphere. The suspension was stirred at 95° C. for 3 h before it was diluted with saturated aq. KF solution (8 mL) and stirred at 20° C. for 1 h. The mixture was extracted with ethyl acetate (2×8 mL). The combined organic layer was washed with water (20 mL), brine (15 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by Combi-flash (silica gel, 30-70% EtOAc in petroleum ether) to give tert-butyl N-(tert-butoxycarbonyl)amino-(2-(1-ethoxyvinyl)-9H-purin-6-yl)carbamate (256 mg, 33% yield) as a white solid. Step 3: To a solution of the product from the last step (60 mg, 109.38 umol, 1 eq) and diethyl 2-benzyl-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (53.22 mg, 131.26 umol, 1.2 eq) in MeCN (1 mL) was added BSA (65 uL, 262.52 umol, 2.4 eq). The solution was stirred at 65° C. for 0.5 h before it was cooled to 25° C. and followed by addition of TMSOTf (24 uL, 131.26 umol, 1.2 eq). The resulting solution was stirred at 65° C. for 1 h before it was diluted with saturated aq. NaHCO3(5 mL) and extracted with EtOAc (2×5 mL). The combined organic layer was concentrated. The crude residue was purified by preparative TLC (EtOAc) to give diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-(1-ethoxyvinyl)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)-methoxy)malonate (42 mg, 55% yield) as a colorless gum. Step 4: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-(1-ethoxyvinyl)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (40 mg, 57.66 umol, 1 eq) in THF (1.5 mL) was added 1M aq. HCl aq. (0.5 mL, 8.67 eq). The mixture was stirred at 20° C. for 21 before it was diluted with saturated aq. NaHCO3(5 mL) and the mixture was extracted with ethyl acetate (3×5 mL). The combined organic layer was dried by Na2SO4, filtered and concentrated to provide crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-acetyl-6-amino-9H-purin-9-yl)-3-ethynyltetrahydro-furan-2-yl)methoxy)malonate (40 mg) as a light yellow gum. Step 5: To a solution of crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-acetyl-6-amino-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (30 mg, 45.07 umol, 1 eq) in THF (4 mL) was added 1M aq. LiOH aq. (901 uL, 20 eq). The mixture was stirred at 20° C. for 6 before it was acidified with 1N aq. HCl to pH 6 and concentrated. The crude residue was purified by preparative HPLC and the fraction was dried by lyophilization to give the title compound (1.4 mg, 6% yield) as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.43 (bs, 1H), 7.14-6.98 (m, 5H), 6.06 (d, J=6.4 Hz, 1H), 4.89 (d, J=6.8 Hz, 1H), 4.26 (m, 1H), 3.94-3.91 (m, 2H), 3.30-3.21 (m, 2H), 2.91 (s, 1H), 2.61 (s, 3H); LC/MS [M+H]=526.0. Example 128 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiophen-2-ylmethyl)malonic acid Proceeding as described in Example 15 above but substituting allyl bromide with 2-(bromomethyl)thiophene provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.37 (bs, 1H), 7.10 (d, J=5 Hz, 1H), 6.93-6.72 (m, 2H), 6.00 (d, J=7.2 Hz, 1H), 4.98 (d, J=7.6 Hz, 1H), 4.23 (bs, 1H), 4.06 (bs, 2H), 3.62-3.58 (m, 2H), 2.94 (s, 1H); LC/MS [M+H]=523.9. Example 129 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(cyclopentylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(phenylsulfonyl)acetic acid The title compound was prepared in an analogous manner to Example 59 except ethyl 2-diazo-2-(phenylsulfonyl)acetate was used instead of 1-ethyl 3-(prop-1-en-1-yl) 2-diazo malonate in Step 3, cyclopentanamine is used in place of benzylamine in Step 7 and Step 4 is eliminated. LC/MS [M+H]=592.0. Example 130 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(1-ethyl-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Proceeding as described in Example 20 above but substituting diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate with diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)(methyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-tetrahydrofuran-2-yl)methoxy)malonate provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.04 (s, 1H), 7.58 (dd, J=6.7, 1.9 Hz, 1H), 7.27-7.38 (m, 5H), 6.35 (t, J=6.9 Hz, 1H), 5.95 (d, J=7.8 Hz, 1H), 4.77 (d, J=7.8 Hz, 1H), 4.28 (t, J=2.6 Hz, 1H), 3.96-4.13 (m, 4H), 3.38-3.57 (m, 2H), 3.05 (s, 1H) 2.99 (m, 3H), 1.32 (t, J=7.3 Hz, 3H); LC/MS [M+H]=653.1. Example 131 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-chloro-3-methyl-1H-pyrazolo[3,4-b]pyridin-1-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Step 1: To a solution of (4-aminophenyl)methanol (27.65 g, 224.44 mmol, 1 eq) in a mixture of anhydrous DCM (100 mL) and anhydrous THF (50 mL) maintained at 25° C. was added 1-chloro-3-isocyanatopropane (26.83 g, 224.44 mmol, 1 eq) dropwise. The reaction mixture became slightly exothermic and turned yellow as a precipitate was formed within 15 minutes. The mixture was stirred for 1.5 h before hexanes (50 mL) was added. The mixture was stirred for additional 15 min before the solid product was collected by filtration, rinsing with a mixture of DCM and hexanes (5:1=v:v). Upon drying provided 1-(3-chloropropyl)-3-(4-(hydroxymethyl)phenyl)urea (38.45 g) as a light yellow solid. Step 2: To a solution of 1-(3-chloropropyl)-3-(4-(hydroxymethyl)phenyl)urea (30.00 g, 123.6 mmol, 1.0 eq) in THF (300 mL) at 25° C. was added a solution of 1M t-BuOK in THF (247.2 mL, 247.2 mmol, 2.0 eq) dropwise while stirring vigorously with a mechanical stirrer. The resulting heterogeneous mixture was stirred at 25° C. for 6 h before it was cooled to 0° C. and acidified to pH 5-6 with 2N aq. HCl. The organic volatile was then removed under reduced pressure. The crude solid was taken up in MeOH (75 mL) and concentrated. The resulting solid mixture was rinsed with a solution of 7% MeOH in DCM (220 mL) under gentle heating and the solid was filtered off. The solid was rinsed again with 7% MeOH in DCM (150 mL) and filtered. The combined rinse was concentrated to give the desired crude 1-(4-(hydroxymethyl)phenyl)tetrahydropyrimidin-2 (1H)-one as a yellowish solid (27.68 g). Step 3: To a suspension of crude 1-(4-(hydroxymethyl)phenyl)tetrahydropyrimidin-2 (1H)-one (15.00 g, 72.74 mmol, 1 eq) in DCM (250 mL) was added a solution of thionyl chloride (10.61 mL, 145.48 mmol, 2 eq) at 25° C. under a N2atmosphere. The mixture was stirred at 25° C. for 8 h before it was diluted with EtOAc (250 mL) and stirred for 30 min. The solid was collected by filtration, rinsed with EtOAc and dried to provide crude 1-(4-(chloro-methyl)phenyl)tetrahydropyrimidin-2 (1H)-one (15.00 g). Step 4: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (7.04 g, 16.99 mmol, 1 eq) in DMF (70 mL) was added Cs2CO3(11.07 g, 33.98 mmol, 2 eq) and crude 1-(4-(chloro-methyl)phenyl)tetrahydropyrimidin-2 (1H)-one (5.09 g, 25.49 mmol, 1.5 eq) at 20° C. The mixture was stirred at 20° C. for 5 h before it was diluted with H2O (300 mL) and extracted with EtOAc (3×100 mL). The combined organic layer was washed with brine (200 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (10-40% acetone in DCM) to provide diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (9.62 g, 94% yield) as a solid. Step 5: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyl-tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (17.31 g, 28.72 mmol, 1 eq) in DCM (90 mL) was added H2O (18 mL) and TFA (90 mL, 1.22 mol, 42 eq) at 0° C. The reaction mixture was stirred at 20-25° C. for 16 h before it was concentrated under reduced pressure. The residue was azeotroped with DCM (2×50 mL) under reduced pressure to provide the crude product diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate which was used in the next step without further purification. Step 6: To a solution of crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxy-tetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (17.23 g, crude) in DCM (170 mL) was added 4-DMAP (374 mg, 3.06 mmol, 0.1 eq), Ac2O (17.21 mL, 183.77 mmol, 6 eq) and pyridine (19.78 mL, 245.02 mmol, 8 eq) at 0° C. The reaction mixture was stirred at 20-25° C. for 16 h before it was concentrated under reduced pressure. The residue was re-dissolved in EtOAc (200 mL), washed with 1N aq. HCl (150 mL), 10% aq. Cu2SO4(150 mL), saturated aq. NaHCO3(150 mL) and brine (150 mL), dried over Na2SO4, filtered and concentrated to provide crude diethyl 2-(4-(2-oxotetrahydro-pyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (19.24 g) as a foam which was carried onto the next step without further purification. Step 7: To a suspension of crude diethyl 2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (450 mg, 0.70 mmol, 1 eq) and 6-chloro-3-methyl-1H-pyrazolo[3,4-b]pyridine (128 mg, 0.77 mmol, 1.1 eq) in MeCN (4 mL) was added DBU (315 uL, 2.09 mmol, 3.0 eq) at 0° C. The solution was stirred at 0° C. for 5 min and followed by addition of a solution of TMSOTf (566 uL, 3.13 mmol, 4.5 eq) in MeCN (2 mL) dropwise. The solution was stirred at 0° C. for 0.5 h and then stirred at 70° C. for 16 h before it was allowed to cool to 25° C. and adjusted the pH to 9 with saturated aq. NaHCO3. The mixture was extracted with ethyl acetate (2×40 mL). The combined organic layer was dried with Na2SO4, filtered and concentrated. The crude residue which was purified by Combi-flash on silica gel (0-10% MeOH in DCM) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-chloro-3-methyl-1H-pyrazolo[3,4-b]pyridin-1-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-malonate (255 mg, 49% yield) as a yellow gum. Step 8: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-chloro-3-methyl-1H-pyrazolo[3,4-b]pyridin-1-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydro-pyrimidin-1 (2H)-yl)benzyl)malonate (296 mg, 392 umol, 1 eq) in THF (6 mL) was added aq. LiOH solution (2 M, 1.96 mL, 10 eq). The solution was stirred at 50° C. for 2 h before the organic volatile was removed under reduced pressure. To the water layer was added 1N HCl to adjust the pH to 5-6. The mixture was concentrated to give crude product which was purified by preparative HPLC (Column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)-ACN]; B %: 25%-45%, 10 min). The product was isolated by lyophilization to give 2-(((2R,3S,4R,5R)-5-(6-chloro-3-methyl-1H-pyrazolo[3,4-b]pyridin-1-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid (32 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.27 (d, J=8.28 Hz, 1H), 7.31 (d, J=8.28 Hz, 1H), 6.97 (d, J=8.53 Hz, 2H), 6.81 (d, J=8.53 Hz, 2H), 6.49 (s, 1H), 6.20 (s, 1H), 6.13 (d, J=7.53 Hz, 1H), 5.97 (s, 1H), 4.98 (d, J=7.53 Hz, 1H), 4.13 (dd, J=8.41, 2.64 Hz, 1H), 3.93-3.99 (m, 1H), 3.84-3.91 (m, 1H), 3.64 (s, 1H), 3.42-3.52 (t, J=5.60 Hz, 2H), 3.17-3.23 (m, 2H), 3.05-3.16 (m, 2H), 2.45 (s, 3H), 1.90 (m, 2H); LC/MS [M+H]=614.3. Example 132 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-oxo-1H-purin-9 (6H)-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Step 1: To a solution of 2,6-dichloro-9H-purine (379.97 mg, 2.01 mmol) in MeCN (5 mL) was added BSA (956 uL, 3.87 mmol) at 25° C. The reaction mixture was stirred at 65° C. for 0.5 h and then cooled back to 25° C. To this mixture was added diethyl 2-(4-(2-oxotetra-hydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (1 g) in MeCN (5 mL) and TMSOTf (419 uL, 2.32 mmol) at 25° C. and further stirred at 65° C. for 5 h. The reaction mixture was allowed to cool to 25° C. before it was quenched with saturated aq. NaHCO3(10 mL) and extracted with EtOAc (3×5 mL). The combined organic layer was washed brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by flash silica gel column chromatography (0-10% MeOH in DCM) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-di-acetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (365 mg) as a foam. Step 2: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (180 mg) in THF (2 mL) was added LiOH·H2O (97.39 mg, 2.32 mmol, 10 eq) in H2O (1 mL) at 25° C. The reaction mixture was stirred at 40° C. for 2 h before the organic volatile was removed under reduced pressure. The aqueous phase was acidified to pH 5-6 with 1N aq. HCl and concentrated under reduced pressure. The crude residue was purified by preparative HPLC (column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)-ACN]; B %: 13%-33%, 10 min) and dried by lyophilization to provide 2-(((2R,3S,4R,5R)-5-(2-chloro-6-oxo-1H-purin-9 (6H)-yl)-3-ethynyl-3,4-dihydroxy-tetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid (15 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.62 (s, 1H), 7.24 (d, J=8.31 Hz, 2H), 7.03 (d, J=8.31 Hz, 2H), 6.32 (d, J=6.48 Hz, 1H), 4.61 (d, J=6.48 Hz, 1H), 4.28-4.34 (m, 1H), 3.92-4.04 (m, 2H), 3.55-3.66 (m, 2H), 3.33-3.40 (m, 4H), 3.02 (s, 1H), 1.99-2.06 (m, 2H); LC/MS [M+H]=617.2. Example 133 Synthesis of 2-(((2R,3S,4R,5R)-3-ethynyl-3,4-dihydroxy-5-(5-methyl-2,4-dioxo-3,4-dihydropyrimidin-1 (2H)-yl)tetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Step 1: To the mixture of 5-methylpyrimidine-2,4(1H,3H)-dione (100 mg, 792.94 umol, 1 eq) in MeCN (2 mL) was added BSA (490 uL, 1.98 mmol, 2.5 eq). The mixture was stirred at 85° C. for 0.5 h. The mixture was cooled to 0° C. and followed by addition of a solution of diethyl 2-(4-(2-oxotetra-hydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triace-toxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (385 mg) in MeCN (2 mL) and TMSOTf (430 uL, 2.38 mmol, 3.0 eq) was added dropwise. The mixture was stirred at 65° C. under N2atmosphere for 5 h before it was allowed to cool to 25° C. and quenched with saturated aq. NaHCO3(10 mL). The mixture was extracted with EtOAc (3×5 mL). The organic layers were combined, dried over Na2SO4, filtered and concentrated in vacuo. The crude compound was purified by silica gel column chromatography (0-5% MeOH in DCM) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-3-ethynyl-5-(5-methyl-2,4-dioxo-3,4-dihydropyrimidin-1 (2H)-yl)tetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (208 mg, 37% yield) as a solid. Step 2: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-3-ethynyl-5-(5-methyl-2,4-dioxo-3,4-dihydropyrimidin-1 (2H)-yl)tetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydro-pyrimidin-1 (2H)-yl)benzyl)malonate (200 mg, 280.62 umol, 1 eq) in THF (2 mL) was added LiOH·H2O (58.88 mg, 1.40 mmol, 5 eq) in H2O (1 mL) at 20-25° C. The reaction mixture was stirred at 40° C. for 1 h before the organic volatile was removed under reduced pressure. The aqueous phase was acidified to pH is 5-6 with 1N aq. HCl and concentrated under reduced pressure. The crude residue was purified by preparative HPLC (column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)-ACN]; B %: 13%-33%, 10 min) and then followed by lyophilization to provide 2-(((2R,3S,4R,5R)-3-ethynyl-3,4-dihydroxy-5-(5-methyl-2,4-dioxo-3,4-dihydropyrimidin-1 (2H)-yl)tetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid (49 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 7.83 (d, J=0.75 Hz, 1H), 7.33 (d, J=8.28 Hz, 2H), 7.11 (d, J=8.28 Hz, 2H), 6.06 (d, J=7.78 Hz, 1H), 4.44 (d, J=7.78 Hz, 1H), 4.16 (t, J=2.26 Hz, 1H), 3.92-4.05 (m, 2H), 3.47-3.65 (m, 3H), 3.33-3.41 (m, 3H), 2.98 (s, 1H), 2.04 (m, 2H), 1.62 (s, 3H); LC/MS [M+H]=573.1. Example 134 Synthesis of 2-(((2R,3S,4R,5R)-5-(2,4-dioxo-3,4-dihydropyrimidin-1 (2H)-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 133 above but substituting thymine with uracil provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 7.91 (d, J=8.0 Hz, 1H), 7.31 (d, J=8.4 Hz, 2H), 7.15 (d, J=8.4 Hz, 2H), 6.01 (d, J=7.5 Hz, 1H), 5.20 (d, J=8.0 Hz, 1H), 4.36 (d, J=7.5 Hz, 1H), 4.17 (t, J=2.4 Hz, 1H), 3.99 (dd, J=18.8, 2.5 Hz, 2H), 3.55-3.66 (m, 2H), 3.44-3.52 (m, 1H), 3.33-3.38 (m, 3H), 3.01 (s, 1H), 1.99-2.10 (m, 2H); LC/MS [M+H]=559.1. Example 135 Synthesis of 2-(((2R,3S,4R,5R)-5-(4-amino-2-oxopyrimidin-1 (2H)-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 133 above but substituting thymine with cytosine provided the title compound as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.92-8.05 (m, 1H), 7.76-7.92 (br s, 1H), 7.45-7.62 (s, 1H), 7.03-7.19 (m, 4H), 6.55 (s, 1H), 5.91-5.99 (m, 1H), 5.87 (d, J=6.80 Hz, 1H), 5.80 (d, J=6.00 Hz, 1H), 5.57 (d, J=7.20 Hz, 1H), 4.13 (t, J=6.80 Hz, 1H), 4.02-4.08 (m, 1H), 3.68-3.82 (m, 2H), 3.50-3.57 (m, 2H), 3.22 (s, 1H), 3.19-3.22 (m, 3H), 1.82-1.99 (m, 2H); LC/MS [M+H]=558.3. Example 136 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 133 above but substituting thymine with 2-chloro-N-(cyclopropylmethyl)-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.22 (s, 1H), 7.28 (br d, J=8.03 Hz, 2H), 7.03 (d, J=8.28 Hz, 2H), 5.97 (d, J=7.28 Hz, 1H), 4.72-4.77 (m, 1H), 4.28 (s, 1H), 3.95-4.05 (m, 2H), 3.34-3.52 (m, 8H), 3.05 (s, 1H), 1.92-2.02 (m, 2H), 1.11-1.20 (m, 1H), 0.51-0.59 (m, 2H), 0.34 (q, J=4.85 Hz, 2H); LC/MS [M+H]=670.1. Example 137 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-(2-hydroxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Step 1: To a solution of compound 1-(3-chloropropyl)-3-(4-(hydroxymethyl)phenyl)urea (5 g, 20.60 mmol, 1 eq) in THF (100 mL) at 0° C. was added NaH (9.89 g, 247.22 mmol, 60% in mineral oil, 12 eq). The reaction mixture was stirred at 25° C. for 1.5 h before it was then added TBDPSCl (6.80 g, 24.72 mmol, 1.2 eq) and stirred further for additional 1.5 h. To the reaction mixture was then added allyl bromide (9.97 g, 82.41 mmol, 4 eq) and stirred further for 16 h. To the reaction mixture was added H2O (50 mL) and the resulting mixture was extracted with EtOAc (3×100 mL). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, filtered and concentrated. The crude residue was purified by silica gel column chromatography and eluted with EtOAc in petroleum ether (0-30%) to provide 1-allyl-3-(4-(((tert-butyldiphenylsilyl)oxy)methyl)phenyl)tetrahydropyrimidin-2 (1H)-one (6 g, 60% yield) as an oil. Step 2: To a solution of compound 1-allyl-3-(4-(((tert-butyldiphenylsilyl)oxy)methyl)-phenyl)tetrahydropyrimidin-2 (1H)-one (6 g, 12.38 mmol, 1 eq) in a mixture of MeOH (60 mL) and DCM (30 mL) at −78° C., ozone (15 psi) was introduced until the blue color of the solution persisted for 20 minutes. The excess of ozone was removed by bubbling nitrogen gas for 10 minutes. To this reaction mixture was added NaBH4(937 mg, 24.76 mmol, 2 eq) and the mixture was allowed to reach 0° C. and stirred for 15 h at 25° C. The mixture was poured into 1N aq. HCl (50 mL) and extracted with EtOAc (2×100 mL). The combined organic layer was washed with water (100 mL) and brine (100 mL), dried (Na2SO4), filtered and concentrated to give crude 1-(4-(((tert-butyldiphenylsilyl)oxy)methyl)phenyl)-3-(2-hydroxyethyl)tetrahydropyrimidin-2 (1H)-one (6.6 g) as an oil which was used directly in the next step. Step 3: To a solution of crude 1-(4-(((tert-butyldiphenylsilyl)oxy)methyl)phenyl)-3-(2-hydroxyethyl)tetrahydropyrimidin-2 (1H)-one (12.38 mmol, 1 eq) in DCM (40 mL) and pyridine (3.00 mL, 37.14 mmol, 3 eq) at 25° C. was added Ac2O (2.32 mL, 24.76 mmol, 2 eq) and 4-DMAP (151 mg, 1.24 mmol, 0.1 eq). The mixture was stirred for 2 h before it was quenched with H2O (60 mL). The mixture was extracted with EtOAc (3×50 mL). The combined organic layer was washed with water (2×30 mL), brine (30 mL), dried over Na2SO4, filtered and concentrated to provide the crude 2-(3-(4-(((tert-butyldiphenylsilyl)-oxy)methyl)phenyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)ethyl acetate which was used in the next step directly. Step 4: To a solution of crude 2-(3-(4-(((tert-butyldiphenylsilyl)oxy)methyl)phenyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)ethyl acetate (6.57 g, 12.38 mmol, 1 eq) in THF (40 mL) was added 1 M TBAF solution in THF (18.57 mL, 1.5 eq) at 0° C. The mixture was stirred at 25° C. for 1 h before H2O (100 mL) was added. The reaction mixture was extracted with EtOAc (4×100 mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The water phase was further extracted with CH2Cl2(4×100 mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated to provide more product. Combining both batches of crude product and further purified on silica gel column chromatography (40-100% EtOAc in petroleum ether) to provide 2-(3-(4-(hydroxymethyl)phenyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)ethyl acetate (3.14 g) as as a white solid. Step 5: To a mixture of 2-(3-(4-(hydroxymethyl)phenyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)ethyl acetate (1.28 g, 4.38 mmol, 1 eq) and DMF (3.37 uL, 43.79 umol, 0.01 eq) in DCM (25 mL) at 0° C. was added SOCl2(5 mL, 68.92 mmol, 15.74 eq). The mixture was stirred at 50° C. for 2 h before it was concentrated to give crude 2-(3-(4-(chloromethyl)phenyl)-2-oxo-tetrahydropyrimidin-1 (2H)-yl)ethyl acetate (1.51 g) which was used in the next step without further purification. Step 6: To a solution of crude 2-(3-(4-(chloromethyl)phenyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)ethyl acetate (1.51 g, 4.38 mmol, 1 eq) and of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-2,2-dimethyl-6-(prop-1-yn-1-yl)tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-malonate (1.91 g, 4.60 mmol, 1.05 eq) in DMF (8 mL) was added Cs2CO3(4.28 g, 13.14 mmol, 3 eq). The mixture was stirred at 25° C. for 16 h before it was diluted with H2O (40 mL) and extracted with EtOAc (3×30 mL). The combined organic layer was washed with saturated aq. NH4Cl (2×15 mL), water (2×15 mL), brine (15 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (25-70% EtOAc in petroleum ether) to provide diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-(3-(2-acetoxy-ethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (2.20 g, 68% yield) as a yellow gum. Step 7: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyl-tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-(3-(2-acetoxyethyl)-2-oxotetrahydro-pyrimidin-1 (2H)-yl)benzyl)malonate (2.20 g, 3.19 mmol, 1 eq) in DCM (7.5 mL) was added TFA (7.5 mL, 101.30 mmol, 32 eq) and H2O (1.5 mL). The mixture was stirred at 25° C. for 16 h before it was diluted with water (20 mL), Then the pH of the mixture was adjusted to 7-8 with NaHCO3solid. Then the mixture was extracted with CH2Cl2(4×20 mL). Then the organic phase was washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated to provide crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxy-tetrahydrofuran-2-yl)methoxy)-2-(4-(3-(2-acetoxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (2.1 g) as a yellow gum. To a solution of the above crude product (2.1 g, 3.19 mmol, 1 eq) in DCM (15 mL) was added Ac2O (1.79 mL, 19.14 mmol, 6 eq), pyridine (2.06 mL, 25.52 mmol, 8 eq) and 4-DMAP (38.97 mg, 319.00 umol, 0.1 eq). The mixture was stirred at 25° C. for 2 h before it was diluted with EtOAc (100 mL), sequentially washed with 1N aq. HCl (2×30 mL). The organic layer was washed with water (20 mL), saturated aq. NaHCO3solution (2×20 mL), water (20 mL), and brine (10 mL). The organic layer was dried over MgSO4and concentrated to provide crude diethyl 2-(4-(3-(2-acetoxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (2.3 g) as a yellow foam. Step 8: To a solution of 2-chloro-N-methyl-9H-purin-6-amine (151.59 mg, 825.68 umol, 1.1 eq) in MeCN (3 mL) was added BSA (408.19 uL, 1.65 mmol, 2.2 eq). The mixture was stirred at 65° C. for 0.5 h before it was cooled to 0° C. and followed by addition of diethyl 2-(4-(3-(2-acetoxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (550 mg, crude) in MeCN (3 mL) and TMSOTf (406.91 uL, 2.25 mmol, 3 eq). The mixture was stirred at 65° C. for 3 h before it was allowed to cool to 25° C. and quenched with saturated aq. NaHCO3(20 mL). The reaction mixture was extracted with EtOAc (4×20 mL). The combined organic layer was washed with water (10 mL), brine (10 mL), dried over Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (30-70% EtOAc in petroleum ether) first and then further purified by preparative TLC (7% MeOH in DCM) to provide diethyl 2-(4-(3-(2-acetoxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (180 mg, 22% yield) as a foam. Step 9: To a solution of diethyl 2-(4-(3-(2-acetoxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (180 mg, 210.21 umol, 1 eq) in THF (1 mL) was added saturated aq. LiOH solution (1.5 mL). The mixture was stirred at 50° C. for 2 h before the organic volatile was removed under reduced pressure. The pH of the mixture was adjusted to 2-3 with 6N aq. HCl solution and then concentrated. The crude residue was purified by preparative HPLC (column: YMC-Actus ODS-AQ 150*30 5 u; mobile phase: [water (0.225% FA)-ACN]; B %: 15%-35%, 15 min) to provide 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-(2-hydroxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid (71.8 mg, 50% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.14 (s, 1H), 7.27 (d, J=8.0 Hz, 2H), 6.98 (d, J=8.0 Hz, 2H), 5.96 (d, J=7.5 Hz, 1H), 4.76 (d, J=7.4 Hz, 1H), 4.26 (br s, 1H), 4.04 (s, 2H), 3.66 (t, J=5.6 Hz, 2H), 3.36-3.55 (m, 8H), 3.05 (m, 4H), 1.96-2.04 (m, 2H); LC/MS [M+H]=674.1. Example 138 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-(2-hydroxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 137 above but substituting 2-chloro-N-methyl-9H-purin-6-amine with 2-chloro-N-(cyclopropylmethyl)-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.09 (s, 1H), 7.28 (d, J=8.3 Hz, 2H), 7.01 (d, J=8.3 Hz, 2H), 5.96 (d, J=7.5 Hz, 1H), 4.74 (d, J=7.3 Hz, 1H), 4.26 (t, J=2.8 Hz, 1H), 4.04 (d, J=2.3 Hz, 2H), 3.62-3.69 (m, 2H), 3.45-3.54 (m, 4H), 3.34-3.42 (m, 6H), 3.06 (s, 1H) 1.97-2.05 (m, 2H), 1.09-1.21 (m, 1H), 0.52-0.61 (m, 2H), 0.34 (q, J=4.8 Hz, 2H); LC/MS [M+H]=714.1 Example 139 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-(2-hydroxyethyl)-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 137 above but substituting 2-chloro-N-methyl-9H-purin-6-amine with 2-chloro-N-isopropyl-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.08 (s, 1H), 7.29 (d, J=8.3 Hz, 2H), 7.02 (d, J=8.3 Hz, 2H), 5.95 (d, J=7.5 Hz, 1H), 4.73 (d, J=7.5 Hz, 1H), 4.39 (br s, 1H), 4.26 (t, J=2.5 Hz, 1H), 4.04 (d, J=2.0 Hz, 2H), 3.66 (t, J=5.8 Hz, 2H), 3.37-3.52 (m, 9H), 1.97-2.04 (m, 2H), 1.30 (d, J=6.3 Hz, 6H); LC/MS [M+H]=702.1. Example 140 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 133 above but substituting thymine with 2-chloro-N-isopropyl-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.16 (s, 1H) 7.32 (d, J=8.44 Hz, 2H) 7.08 (br d, J=8.31 Hz, 2H) 5.99 (d, J=7.46 Hz, 1H) 4.81 (d, J=7.46 Hz, 1H) 4.41 (br s, 1H) 4.26-4.31 (m, 1H) 3.97-4.12 (m, 2H) 3.40-3.57 (m, 4H), 3.33-3.37 (m, 2H), 3.03 (s, 1H), 1.99 (m, 2H), 1.31 (m, 6H); LC/MS [M+H]=658.3. Example 141 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 133 above but substituting thymine with 2-chloro-N-methyl-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.20 (s, 1H), 7.27 (d, J=8.13 Hz, 2H), 7.00 (d, J=8.00 Hz, 2H), 5.97 (d, J=7.38 Hz, 1H), 4.80 (d, J=7.38 Hz, 1H), 4.27 (s, 1H), 4.04 (m, 2H), 3.37-3.50 (m, 4H), 3.31 (d, J=1.13 Hz, 3H), 3.06 (s, 2H) 3.04 (s, 1H), 1.90-2.00 (m, 2H); LC/MS [M+H]=630.2. Example 142 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-2-ylmethyl)malonic acid Proceeding as described in Example 15 above but substituting allyl bromide with 2-(bromomethyl)thiazole provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.56 (s, 1H), 7.56 (d, J=3.4 Hz, 1H), 7.33 (d, J=3.4 Hz, 1H), 6.00 (d, J=5.6 Hz, 1H), 4.68-4.73 (m, 1H), 4.38 (dd, J=6.3, 3.4 Hz, 1H), 3.98-4.07 (m, 2H), 3.85 (s, 2H), 3.00 (s, 1H); LC/MS [M+H]=524.9. Example 143 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-5-ylmethyl)malonic acid Proceeding as described in Example 15 above but substituting allyl bromide with 5-(chloromethyl)thiazole provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.75 (s, 1H), 8.47 (s, 1H), 7.72 (s, 1H), 6.03 (d, J=7.3 Hz, 1H), 4.97 (d, J=7.3 Hz, 1H), 4.36 (dd, J=4.3, 3.0 Hz, 1H), 4.12-4.18 (m, 1H), 4.04-4.10 (m, 1H), 3.63-3.79 (m, 2H), 3.00 (s, 1H); LC/MS [M+H]=524.9. Example 144 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(pyrrolidin-1-yl)benzyl)malonic acid Step 1: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (10.0 g, 24.13 mmol, 1 eq) in DMF (100 mL) was added Cs2CO3(23.59 g, 72.39 mmol, 3 eq) and 1-(bromomethyl)-4-iodo-benzene (10.75 g, 36.20 mmol, 1.5 eq). The suspension was stirred at 20° C. for 1 h before it was diluted with water (200 mL). The resulting mixture was extracted with EtOAc (4×50 mL). The combined organic layer was washed with water (2×200 mL), brine (200 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (5-20% of EtOAc in petroleum ether) to provide diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]-dioxol-5-yl)methoxy)-2-(4-iodobenzyl)malonate (11.83 g, 74% yield) as a white solid as a white solid. Step 2: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-iodobenzyl)malonate (2.00 g, 3.17 mmol, 1 eq) in DMA (22 mL) was added K2CO3(1.32 g, 9.52 mmol, 3 eq), CuI (120.84 mg, 634.50 umol, 0.2 eq) and proline (438.30 mg, 3.81 mmol, 1.2 eq). The green suspension was stirred at 80° C. under N2atmosphere for 16 h before it was allowed to cool and poured into water (40 mL) and 2N aq. LiOH (1 mL). The mixture was extracted with ethyl acetate (2×30 mL). The resulting aq. layer was acidified to pH 5 with 1N aq. HCl solution and then extracted with ethyl acetate (2×40 mL). The combined organic layer was washed with water (80 mL), brine (80 mL), dried over Na2SO4, filtered and concentrated to give crude (S)-1-(4-(2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-3-ethoxy-2-(ethoxycarbonyl)-3-oxopropyl)phenyl)pyrrolidine-2-carboxylic acid (390 mg) as a yellow gum. Step 3: To a solution of crude (S)-1-(4-(2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-3-ethoxy-2-(ethoxycarbonyl)-3-oxopropyl)phenyl)pyrrolidine-2-carboxylic acid (420 mg, 680.01 umol, 1 eq) in DMF (5 mL) was added K2CO3(282 mg, 2.04 mmol, 3 eq) and EtI (81.58 uL, 1.02 mmol, 1.5 eq). The mixture was stirred at 20° C. for 0.5 h before it was diluted with water (10 mL), and extracted with ethyl acetate (2×10 mL). The combined organic layer was washed with water (20 mL), brine (20 mL), dried over Na2SO4, and filtered and concentrated to give crude diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-((S)-2-(ethoxycarbonyl)pyrrolidin-1-yl)benzyl)malonate (330 mg) as a yellow foam. Step 4: To a solution of crude diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-((S)-2-(ethoxycarbonyl)pyro-lidin-1-yl)benzyl)malonate (330 mg, crude) in DCM (4 mL) was added H2O (0.8 mL) and TFA (4 mL) at 0° C. The solution was stirred at 20° C. for 4.5 h before it was quenched with saturated aq. NaHCO3to adjust the pH to 9. The mixture was extracted with ethyl acetate (2×8 mL). The combined organic layer was washed with brine (20 mL), dried over Na2SO4, filtered and concentrated to crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxy-tetrahydrofuran-2-yl)methoxy)-2-(4-((S)-2-(ethoxycarbonyl)pyrrolidin-1-yl)benzyl)malonate (285 mg) as a yellow foam. Step 5: To a solution of crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-(4-((S)-2-(ethoxycarbonyl)pyrrolidin-1-yl)benzyl)malonate (285 mg) in pyridine (4 mL) was added 4-DMAP (172 mg, 1.41 mmol, 3 eq) and Ac2O (352.60 uL, 3.76 mmol, 8 eq). The solution was stirred at 20° C. for 16 h before it was diluted with water (10 mL), and extracted with ethyl acetate (3×10 mL). The combined organic layer was washed with brine (20 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (20-50% of ethyl acetate in petrol ether) to give diethyl 2-(4-((S)-2-(ethoxycarbonyl)pyro-lidin-1-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (220 mg, 51% yield for four steps) as a yellow gum. Step 6: To a suspension of diethyl 2-(4-((S)-2-(ethoxycarbonyl)pyrrolidin-1-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (50 mg, 72.50 umol, 1 eq) and 6-chloropurine (15 mg, 86.99 umol, 1.2 eq) in MeCN (1 mL) was added BSA (44.80 uL, 181.24 umol, 2.5 eq). The suspension was stirred at 65° C. for 0.5 h before it was cooled down to 0° C. and followed by addition of TMSOTf (32.75 uL, 181.24 umol, 2.5 eq). The mixture was stirred at 65° C. for 1 h before it was poured into saturated aq. NaHCO3(3 mL). The reaction mixture was extracted with ethyl acetate (3×3 mL). The combined organic layer was concentrated. The crude residue was purified by preparative TLC (petroleum ether:EtOAc=2:1) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-((S)-2-(ethoxy-carbonyl)pyrrolidin-1-yl)benzyl)malonate (33 mg, 57% yield) as a yellow gum. Step 7: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-((S)-2-(ethoxycarbonyl)pyrrolidin-1-yl)benzyl)malonate (28 mg, 35.03 umol, 1 eq) in THF (2 mL) was added 1M aq. LiOH (701 uL, 20 eq). The mixture was stirred at 20° C. for 4.5 h before the organic volatile was removed under reduced pressure. The aq. layer was acidified to pH 6 with 1N aq. HCl solution before it was concentrated. The crude residue was purified by preparative HPLC (Column: YMC-Triart Prep C18 150*40 mm*7 um; mobile phase: [water (0.225% FA)-ACN]; B %: 15%-35%, 10 min.) and dried by lyophilization to provide 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(pyrrolidin-1-yl)benzyl)malonic acid (2.6 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.21 (s, 1H), 7.05 (d, J=8.53 Hz, 2H), 6.26 (d, J=8.53 Hz, 2H), 5.98 (d, J=7.53 Hz, 1H), 4.92-5.02 (m, 1H), 4.29-4.33 (m, 1H), 4.09 (dd, J=10.16, 2.38 Hz, 1H), 3.97 (dd, J=10.04, 3.01 Hz, 1H), 3.38 (d, J=14.56 Hz, 1H), 3.23 (d, J=14.56 Hz, 1H), 3.02-3.09 (m, 4H), 3.00 (s, 1H), 1.89-1.97 (m, 4H); LC/MS [M+H]=587.1. Example 145 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-(ethylthio)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: To a mixture of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (300 mg, 349.53 umol, 1 eq) in DMF (3 mL) was added NaSEt (88.20 mg, 1.05 mmol, 3 eq). The mixture was stirred at 20° C. for 20 h before it was partitioned between water (15 mL) and EtOAc (15 mL). The aqueous phase was extracted with EtOAc (2×10 mL). The combined organic layer was washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered and concentrated under reduced pressure to to provide crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-(ethylthio)-9H-purin-9-yl)-3-ethynyl-tetrahydrofuran-2-yl)methoxy)malonate (310 mg) as an oil which was used for next step without further purification. Step 2: To a mixture of crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-(ethylthio)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (310 mg) in DCM (3 mL) was added TFA (1.5 mL, 20.26 mmol). The mixture was stirred at 20° C. for 2 h before it was neutralized to pH 7-8 with saturated aq. NaHCO3. The reaction mixture was extracted with EtOAc (3×20 mL). The combined extract was washed with brine (15 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to provide crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-(ethylthio)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate as a foam. Step 3: To a mixture of crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-(ethylthio)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (280 mg, crude) in THF (3 mL) was added saturated aq. LiOH (3 mL). The mixture was stirred at 55° C. for 1 h before it was cooled to room temperature. The reaction mixture was extracted with EtOAc (3×8 mL). The aqueous phase was adjusted to pH 2-3 with 2M aq. HCl before it was extracted with EtOAc (4×10 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude product was purified by preparative HPLC (column: YMC-Triart Prep C18 150*40 mm*7 um; mobile phase: [water (0.225% FA)-ACN]; B %: 23%-43%, 11 min) and dried by lyophilization to give 2-(((2R,3S,4R,5R)-5-(6-amino-2-(ethylthio)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid (4.4 mg) as a white powder. 1H NMR (400 MHz, DMSO-d6) δ ppm 12.59-14.32 (m, 2H), 8.30 (s, 1H), 7.33 (s, 2H), 7.00-7.21 (m, 5H), 6.15 (s, 1H), 6.00 (d, J=6.78 Hz, 1H), 5.82 (d, J=7.53 Hz, 1H), 5.01 (s, 1H), 4.14 (dd, J=6.40, 2.64 Hz, 1H), 3.99 (d, J=13.05 Hz, 1H), 3.83 (s, 1H), 3.58 (s, 1H), 3.17-3.18 (m, 2H), 2.99-3.13 (m, 2H), 1.31 (t, J=7.28 Hz, 3H); LC/MS [M+H]=544.0. Example 146 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(1-(2-methoxyethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Proceeding as described in Example 11 above but substituting chloro(methoxy)-methane with 1-chloro-2-methoxyethane provided the title compound as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.41 (s, 1H), 7.80 (s, 2H), 7.60 (dd, J=6.78, 2.01 Hz, 1H), 7.46 (dd, J=7.03, 2.01 Hz, 1H), 7.35 (d, J=8.28 Hz, 2H), 7.21 (d, J=8.28 Hz, 2H), 7.19-7.24 (m, 1H), 6.26-6.31 (m, 1H), 5.82 (d, J=7.78 Hz, 1H) 4.86 (d, J=7.78 Hz, 1H), 4.17 (d, J=1.76 Hz, 1H), 4.10 (t, J=5.14 Hz, 2H), 4.02 (dd, J=10.29, 4.77 Hz, 1H), 3.78-3.85 (m, 1H), 3.54-3.64 (m, 3H), 3.29 (d, J=2.01 Hz, 2H), 3.24 (s, 3H); LC/MS [M+H]=669.0. Example 147 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Step 1: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-iodobenzyl)malonate (2.0 g, 3.17 mmol, 1 eq) in DCM (15 mL) was added H2O (3 mL) and TFA (15 mL) at 0° C. The solution was stirred at 25° C. for 16 h before it was quenched with saturated aq. NaHCO3(150 mL). The mixture was extracted with ethyl acetate (2×100 mL). The combined organic layer was washed with brine (200 mL), dried over Na2SO4, filtered and concentrated to give crude diethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-iodobenzyl)malonate (1.74 g) as a yellow foam. Step 2: To a solution of diethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-iodobenzyl)malonate (1.74 g, 3.17 mmol, 1 eq) in pyridine (20 mL) was added 4-DMAP (1.16 g, 9.52 mmol, 3 eq) and Ac2O (2.38 mL, 25.39 mmol, 8 eq). The solution was stirred at 25° C. for 3 h before it was diluted with water (60 mL) and extracted with ethyl acetate (2×60 mL). The combined organic layer was washed with water (100 mL), brine (100 mL), dried over Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (10-50% of ethyl acetate in petroleum ether) to give diethyl 2-(4-iodobenzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyl-tetrahydrofuran-2-yl)methoxy)malonate (1.85 g, 86% yield for two steps) as a yellow foam. Step 3: To a solution of diethyl 2-(4-iodobenzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyl-tetrahydrofuran-2-yl)methoxy)malonate (1.00 g, 1.48 mmol, 1 eq) in MeCN (12 mL) was added 2-chloro-N-methyl-9H-purin-6-amine (327 mg, 1.78 mmol, 1.2 eq) and BSA (916 uL, 3.71 mmol, 2.5 eq). The suspension was stirred at 65° C. for 0.5 h before it was cooled down to 0° C. and followed by addition of TMSOTf (804 uL, 4.45 mmol, 3 eq). The resulting mixture was stirred at 65° C. for 1.5 h before it was diluted with saturated aq. NaHCO3(10 mL) and extracted with ethyl acetate (2×10 mL). The combined organic layer was washed with brine (25 mL), dried over Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (10-50% ethyl acetate in petroleum ether) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-iodobenzyl)malonate (595 mg, 50% yield) as a yellow solid. Step 4: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(methyl-amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-iodobenzyl)malonate (592 mg, 741.88 umol, 1 eq) in DCM (8 mL) was added 4-DMAP (18 mg, 148.38 umol, 0.2 eq), TEA (413 uL, 2.97 mmol, 4 eq) and (Boc)2O (324 mg, 1.48 mmol, 2 eq). The solution was stirred at 20° C. for 2 h before it was diluted with saturated aq. NH4Cl (20 mL) and extracted with ethyl acetate (2×20 mL). The combined organic layer was washed with brine (40 mL), dried over Na2SO4, filtered and concentrated. The crude was purified by flash column chromatography on silica gel (15-50% of ethyl acetate in petroleum ether) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)(methyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-iodobenzyl)malonate (560 mg, 84% yield) as a foam. Step 5: To a mixture of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)-(methyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-iodobenzyl)malonate (660 mg, 734.89 umol, 1 eq) and (2-hydroxypyridin-3-yl)boronic acid (204.18 mg, 1.47 mmol, 2 eq) in dioxane (6 mL) and H2O (2 mL) was added Pd(dppf)Cl2(53.77 mg, 73.49 umol, 0.1 eq) and K2CO3(304.70 mg, 2.20 mmol, 3 eq). The yellow mixture was degassed with N2gas for 10 min before the mixture was stirred at 70° C. for 2.5 h The mixture was diluted with water (5 mL) and extracted with ethyl acetate (3×5 mL). The combined organic layer was dried over Na2SO4, filtered and concentrated. The crude was purified by flash column chromatography on silica gel (50-100% of ethyl acetate in petroleum ether) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxy-carbonyl)(methyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (146 mg) as a foam. Step 6: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)-(methyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (145 mg, 167.58 umol, 1 eq) in DCM (6 mL) was added TFA (1.5 mL, 20.26 mmol, 121 eq). The solution was stirred at 25° C. for 1 h before it was neutralized with saturated aq. NaHCO3solution. The mixture was extracted with ethyl acetate (3×12 mL). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, filtered and concentrated. The crude was purified by preparative TLC (ethyl acetate) to give diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (110 mg) as a solid. Step 7: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(methyl-amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1,2-dihydro-pyridin-3-yl)benzyl)malonate (105 mg, 137.23 umol, 1 eq) in THF (4.5 mL) was added aq. LiOH solution (1 M, 1.5 mL, 11 eq). The mixture was stirred at 25° C. for 4 h before the organic volatile was removed under reduced pressure. The aq. layer was acidified to pH 6 with 1N aq. HCl solution and concentrated. The crude residue was purified by preparative HPLC (Column: YMC-Triart Prep C18 150*40 mm*7 um; mobile phase: [water (0.225% FA)-ACN]; B %: 20%-40%, 10 min) and dried by lyophilization to give 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)-methoxy)-2-(4-(2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid (6.6 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.44 (s, 1H), 8.23 (d, J=3.75 Hz, 1H), 7.48 (d, J=6.63 Hz, 1H), 7.39 (d, J=7.00 Hz, 2H), 7.34 (d, J=5.63 Hz, 1H), 7.13-7.22 (m, 2H), 6.15-6.29 (m, 2H), 6.00 (d, J=6.75 Hz, 1H), 5.82 (d, J=7.38 Hz, 1H), 4.71-4.89 (m, 1H), 4.16 (dd, J=4.94, 2.56 Hz, 1H), 3.86-4.05 (m, 1H), 3.53-3.83 (m, 1H), 3.51 (s, 1H), 3.48-3.30 (m, 5H overlapped under water peak), 2.90 (d, J=4.38 Hz, 3H); LC/MS [M+H]=624.9. Example 148 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Step 1: To a solution of 1-(4-(((tert-butyldiphenylsilyl)oxy)methyl)phenyl)tetrahydro-pyrimidin-2 (1H)-one (7.33 g, 16.48 mmol, 1 eq) in DMF at 0° C. was added NaH (725 mg, 60% in mineral oil, 18.13 mmol, 1.1 eq). The mixture was stirred for 15 min and followed by addition of CH3I (4.10 mL, 65.92 mmol, 4 eq). The reaction mixture was stirred from 0-25° C. over 16 h before it was diluted with H2O (100 mL) and extracted with EtOAc (3×50 mL). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, filtered and concentrated. The crude was purified by flash column chromatography on silica gel (0-50% EtOAc in petroleum ether) to provide 1-(4-(((tert-butyldiphenylsilyl)oxy)-methyl)phenyl)-3-methyltetrahydropyrimidin-2 (1H)-one (3.68 g, 48% yield) as a colourless gum. Step 2: To a solution of 1-(4-(((tert-butyldiphenylsilyl)oxy)methyl)phenyl)-3-methyltetra-hydropyrimidin-2 (1H)-one (3.68 g, 8.02 mmol, 1 eq) in THF (35 mL) was added TBAF in THF (1.5 M, 10.70 mL, 2 eq) at 0° C. The reaction mixture was stirred at 25° C. for 1.5 h before it was diluted with H2O (20 mL) and extracted with EtOAc (3×50 mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The crude was purified by flash column chromatography on silica gel (0-5% MeOH in DCM) to provide 1-(4-(hydroxymethyl)phenyl)-3-methyltetrahydropyrimidin-2 (1H)-one (1.06 g, 60% yield) as a yellow solid. Step 3: To a solution of 1-(4-(hydroxymethyl)phenyl)-3-methyltetrahydropyrimidin-2 (1H)-one (1.06 g, 4.81 mmol, 1 eq) in DCM (10 mL) and DMF (0.1 mL) was added SOCl2(698 uL, 9.62 mmol, 2 eq) at 20-25° C. The reaction mixture was stirred for 0.5 h and additional amount of SOCl2(419 uL, 5.77 mmol, 1.2 eq) was added. The resulting mixture was stirred at 40° C. for 1 h before it was concentrated. The residue was azeotroped with DCM (3×10 mL) under reduced pressure to provide crude 1-(4-(chloromethyl)phenyl)-3-methyltetra-hydropyrimidin-2 (1H)-one which was used in the next step without further purification. Step 4: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (1.78 g, 4.30 mmol, 1 eq) in DMF (20 mL) was added Cs2CO3(4.21 g, 12.91 mmol, 3 eq) at 25° C. The reaction mixture was stirred for 0.5 h and followed by addition of crude 1-(4-(chloromethyl)phenyl)-3-methyltetrahydro-pyrimidin-2 (1H)-one (1.13 g). The reaction mixture was stirred at 25° C. for 16 h before it was diluted with H2O (50 mL) and extracted with EtOAc (3×20 mL). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, filtered and concentrated. The crude was purified by column chromatography on silica gel (0-10% of MeOH in DCM) to provide diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyl-tetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (2.63 g, 78% yield) as a brown foam. Step 5: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (2.62 g, 4.25 mmol, 1 eq) in DCM (25 mL) at 0° C. was added TFA (25 mL, 337.65 mmol, 79 eq) and H2O (2.5 mL, 138.77 mmol, 33 eq). The reaction mixture was stirred at 20-25° C. for 16 h before it was concentrated under reduced pressure. The residue was azeotroped with DCM (3×20 mL) under reduced pressure to provide crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (2.57 g) as a syrup which was used in the next step without further purification. Step 6: To a solution of crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-malonate (2.57 g) in DCM (25 mL) at 20-25° C. was added Ac2O (2.39 mL, 25.50 mmol, 6 eq), 4-DMAP (51.92 mg, 425.00 umol, 0.1 eq) and pyridine (2.74 mL, 34.00 mmol, 8 eq). The reaction mixture was stirred at 25° C. for 16 h before it was concentrated under reduced pressure. The residue was re-dissolved in EtOAc (50 mL), washed with 1N aq. HCl (40 mL), 10% aq. Cu2SO4(40 mL), saturated aq. NaHCO3(40 mL) and brine (40 mL), dried over Na2SO4, filtered and concentrated to provide crude diethyl 2-(4-(3-methyl-2-oxotetrahydro-pyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (2.62 g, 53% yield for two steps) which was used in the next step without further purification. Step 7: To a solution of 2-chloro-N-methyl-9H-purin-6-amine (181 mg, 983.86 umol, 1.3 eq) in MeCN (2.5 mL) under N2atmosphere was added BSA (468 uL, 1.89 mmol, 2.5 eq) at 20-25° C. The reaction mixture was stirred at 65° C. for 0.5 h before it was cooled to 25° C. To this mixture was added crude diethyl 2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (500 mg, 756.81 umol, 1 eq) in MeCN (2.5 mL) and TMSOTf (205 uL, 1.14 mmol, 1.5 eq) and stirred at 65° C. for 5 h before it was quenched with saturated aq. NaHCO3(10 mL). The mixture was then extracted with EtOAc (3×10 mL). The combined organic layer was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The crude was purified by flash column chromatography on silica gel column (0-10% MeOH in DCM) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydro-pyrimidin-1 (2H)-yl)benzyl)malonate (530 mg, 60% yield) as a foam. Step 8: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(methyl-amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetra-hydropyrimidin-1 (2H)-yl)benzyl)malonate (540 mg, 688.59 umol, 1 eq) in THF (6 mL) was added LiOH·H2O (288.96 mg, 6.89 mmol, 10 eq) in H2O (3 mL) at 25° C. The reaction mixture was stirred at 40° C. for 2 h before the organic volatile was removed under reduced pressure. The aq. phase was acidified to pH 2-3 with 1N aq. HCl solution and then concentrated. The crude was purified by preparative HPLC (column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)-ACN]; B %: 23%-43%, 10 min) and dried by lyophilization to provide 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetra-hydropyrimidin-1 (2H)-yl)benzyl)malonic acid (77.6 mg, 17% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 13.00-13.93 (s, 2H), 8.37 (s, 1H), 8.23 (d, J=5.01 Hz, 1H), 7.11 (d, J=8.31 Hz, 2H), 6.90 (d, J=8.19 Hz, 2H), 6.15 (s, 1H), 5.98 (s, 1H), 5.81 (d, J=7.58 Hz, 1H), 4.82 (s, 1H), 4.15 (dd, J=4.34, 3.00 Hz, 1H), 3.96 (m, 1H), 3.78 (m, 1H), 3.53 (s, 1H), 3.43-3.52 (m, 2H), 3.23 (m, 2H), 3.20-3.10 (m, 4H overlapped with solvent water peak), 2.91 (m, 2H), 2.81 (s, 3H), 1.95 (m, 2H); LC/MS [M+H]=644.1. Example 149 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 148 above but substituting 2-chloro-N-methyl-9H-purin-6-amine with 2-chloro-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.21 (s, 1H), 7.26 (m, J=8.28 Hz, 2H), 6.96 (m, J=8.28 Hz, 2H), 5.97 (d, J=7.53 Hz, 1H), 4.70-4.83 (m, 1H), 4.27 (t, J=2.76 Hz, 1H), 3.99-4.07 (m, 2H), 3.32-3.52 (m, 6H), 3.05 (s, 1H), 2.87 (s, 3H), 2.00 (quin, J=5.83 Hz, 2H); LC/MS [M+H]=630.2. Example 150 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 148 above but substituting 2-chloro-N-methyl-9H-purin-6-amine with 2-chloro-N-(cyclopropylmethyl)-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.12 (s, 1H), 7.27 (d, J=8.28 Hz, 2H), 7.00 (d, J=7.68 Hz, 2H), 5.96 (d, J=7.53 Hz, 1H), 4.70 (d, J=7.53 Hz, 1H), 4.26 (t, J=2.89 Hz, 1H), 4.03 (br s, 2H), 3.33-3.54 (m, 8H), 3.05 (s, 1H), 2.88 (s, 3H), 1.97-2.04 (m, 2H), 1.11-1.20 (m, 1H), 0.53-0.59 (m, 2H), 0.34 (m, 2H); LC/MS [M+H]=684.3. Example 151 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(3-methyl-2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 148 above but substituting 2-chloro-N-methyl-9H-purin-6-amine with 2-chloro-N-isopropyl-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.08 (s, 1H), 7.28 (d, J=8.28 Hz, 2H), 7.00 (d, J=8.53 Hz, 2H), 5.95 (d, J=7.53 Hz, 1H), 4.66-4.80 (m, 1H), 4.32-4.48 (m, 1H), 4.25 (t, J=2.89 Hz, 1H), 4.00-4.08 (m, 2H), 3.34-3.53 (m, 6H), 3.05 (s, 1H), 2.88 (s, 3H), 1.94-2.06 (m, 2H), 1.24-1.35 (m, 6H); LC/MS [M+H]=672.1. Example 152 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-3-propyltetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Proceeding as described in Example 148 above but substituting Mel and 2-chloro-N-methyl-9H-purin-6-amine with 1-bromopropane and 2-chloro-9H-purin-6-amine provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.15 (s, 1H), 7.28 (d, J=8.4 Hz, 2H), 6.99 (d, J=8.4 Hz, 2H), 5.97 (d, J=7.5 Hz, 1H), 4.75 (d, J=7.6 Hz, 1H), 4.27 (s, 1H), 3.99-4.11 (m, 2H), 3.46-3.54 (m, 2H), 3.35-3.44 (m, 4H), 3.16-3.27 (m, 2H), 3.05 (s, 1H), 1.97-2.03 (m, 2H), 1.50-1.58 (m, 2H), 0.88 (t, J=7.4 Hz, 3H); LC/MS [M+H]=658.1. Example 153 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(1-(2-hydroxyethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Step 1: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (4.13 g, 9.97 mmol, 1 eq) in DCM (40 mL) at 20° C. was added TFA (40 mL, 540.24 mmol, 54 eq) and H2O (4 mL, 222.03 mmol, 22 eq). The mixture was stirred at 20° C. for 15 h before it was quenched by saturated aq. NaHCO3(200 mL) and extracted with EtOAc (5×50 mL). The combined organic layer was washed brine (200 mL), dried over anhydrous Na2SO4, filtered and concentrated to give crude diethyl 2-(((2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxytetrahydrofuran-2-yl)-methoxy)malonate (4.14 g) as a light yellow syrup which was used in the next step directly. To a solution of the above crude product (4.14 g, 12.46 mmol, 1 eq) in pyridine (40 mL) was added Ac2O (9.33 mL, 99.67 mmol, 8 eq) and 4-DMAP (3.81 g, 31.15 mmol, 2.5 eq). The mixture was stirred at 20° C. for 16 h before it was quenched by water (150 mL) and the resulting solution was extracted with EtOAc (4×50 mL). The combined organic layer was washed with 0.5 N aq. HCl (120 mL) and water (2×100 mL), brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (30-50% EtOAc in petroleum ether) to provide diethyl 2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (2.60 g) as a syrup. Step 2: To a solution of diethyl 2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (1.2 g, 2.62 mmol, 1 eq) and 2-chloro-N-isopropyl-9H-purin-6-amine (665 mg, 3.14 mmol, 1.2 eq) in MeCN (15 mL) was added BSA (1.62 mL, 6.54 mmol, 2.5 eq). The suspension was stirred at 65° C. for 0.5 h before it was cooled down to 0° C. To this solution was added TMSOTf (1.45 g, 6.54 mmol, 1.18 mL, 2.5 eq). Then the mixture was stirred at 65° C. for 2.5 h before it was quenched by saturated aq. NaHCO3(50 mL) and the resulting solution was extracted with EtOAc (4×30 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by flash column chromatography on silica gel (30-50% of EtOAc in petroleum ether) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (814 mg, 51% yield). Step 3: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(isopropyl-amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (120 mg, 197 umol, 1 eq) in DMF (1 mL) was added K2CO3(81.56 mg, 590.15 umol, 3 eq) and 3-(4-(bromo-methyl)phenyl)-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)pyridin-2 (1H)-one (161 mg, 295 umol, 1.5 eq). The mixture was stirred at 20° C. for 1.5 h before it was diluted with water (10 mL) and extracted with EtOAc (4×5 mL). The combined organic layer was washed with water (2×30 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by flash column chromatography on silica gel (30-60% of EtOAc in petroleum ether) to provide diethyl 2-(4-(1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)-2-oxo-1,2-dihydro-pyridin-3-yl)benzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(isopropyl-amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (154 mg) as a colorless oil. Step 4: To a solution of diethyl 2-(4-(1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(isopropyl-amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (150 mg, 139 umol, 1 eq) in THF (1 mL) was added TBAF (1 M, 209 uL, 1.5 eq) and AcOH (5.98 uL, 104.59 umol, 0.75 eq) at 0° C. The mixture was stirred at 20° C. for 16 h before it was diluted with water (5 mL) and the resulting solution was extracted with EtOAc (3×5 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by preparative TLC (petroleum ether:EtOAc=1:4) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-tetrahydrofuran-2-yl)methoxy)-2-(4-(1-(2-hydroxyethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (51 mg, 32% yield) as a white solid. Step 5: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(isopropyl-amino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(1-(2-hydroxyethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonate (50 mg, 60 umol, 1 eq) in THF (0.5 mL) was added sat·LiOH·aq (2.51 mg, 60 umol, 0.5 mL, 1 eq). The mixture was stirred at 20° C. for 2.5 h before the organic volatile was removed under reduced pressure. The resulting aq. solution was acidified to pH 2 with 2 N aq. HCl solution and concentrated. The residue was further purification by preparative HPLC (column: YMC-Triart Prep C18 150*40 mm*7 um; mobile phase: [water (0.225% FA)-ACN]; B %: 28%-48%, 10 min) to provide 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxy-tetrahydrofuran-2-yl)methoxy)-2-(4-(1-(2-hydroxyethyl)-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid (10.8 mg, 26% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.08 (s, 1H), 7.55 (dd, J=6.7, 1.9 Hz, 1H), 7.21-7.42 (m, 5H), 6.34 (t, J=6.8 Hz, 1H), 5.95 (d, J=7.3 Hz, 1H), 4.74 (d, J=7.3 Hz, 1H), 4.24-4.39 (m, 2H), 4.00-4.14 (m, 4H), 3.76-3.87 (m, 2H), 3.40-3.54 (m, 2H), 3.06 (s, 1H), 1.24 (dd, J=6.4, 2.9 Hz, 6H); LC/MS [M+H]=697.0. Examples 154 & 155 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)malonic acid and 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-((2-((carboxymethyl)(methyl)amino)ethyl)amino)benzyl)malonic acid Step 1: To a mixture of tert-butyl 3-oxopiperazine-1-carboxylate (8.7 g, 43.45 mmol, 1.2 eq) and (4-iodophenyl) methanol (8.5 g, 36.32 mmol, 1 eq) in DMF (10 mL) and dioxane (90 mL) was added CuI (1.03 g, 5.43 mmol, 0.15 eq), 2-(hydroxymethyl)-2-methyl-propane-1,3-diol (653 mg, 5.43 mmol, 0.15 eq) and Cs2CO3(35.39 g, 108.62 mmol, 3 eq). The mixture was stirred at 110° C. under N2atmosphere for 14 hours before it was cooled. The inorganic solid was filtered off and the filtrate was concentrated in vacuo. The residue was diluted with water (50 mL) and extracted with ethyl acetate (3×50 mL). The combined organic layer was washed with brine (50 mL), dried by Na2SO4, filtered and concentrated. The crude residue was purified by flash silica gel chromatography (0-10% of EtOAc in petroleum ether) to provide tert-butyl 4-(4-(hydroxymethyl)phenyl)-3-oxopiperazine-1-carboxylate (6.1 g, 55% yield) as a white solid. Step 2: To a solution of tert-butyl 4-(4-(hydroxymethyl)phenyl)-3-oxopiperazine-1-carboxylate (2 g, 6.53 mmol, 1 eq) in DCM (10 mL) was added TFA (5.00 mL, 67.53 mmol, 10.34 eq) at 0° C. The mixture was stirred at 25° C. for 3 h before it was concentrated. The residue was diluted with water (20 mL) and extracted with a mixture of DCM and MeOH (50:1=v:v, 2×20 mL). The combined organic layer was concentrated to provide crude 1-(4-(hydroxymethyl)phenyl)piperazin-2-one (2.45 g) as a colorless oil. Step 3: To a solution of crude 1-(4-(hydroxymethyl)phenyl)piperazin-2-one (2.45 g, 11.88 mmol, 1 eq) in MeOH (15 mL) was added HCHO (720 uL, 26.12 mmol, 2.2 eq), AcOH (5 mL, 87.42 mmol, 7.4 eq). The mixture was stirred at 25° C. under N2atmosphere for 15 h before NaBH3CN (2.05 g, 32.65 mmol, 2.75 eq) was added and the resulting mixture was stirred for 2 h. The reaction mixture was concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (10-40% of MeOH in EtOAc) to give 1-(4-(hydroxymethyl)phenyl)-4-methylpiperazin-2-one (1.23 g) as a colorless oil. Step 4: To a solution of 1-(4-(hydroxymethyl)phenyl)-4-methylpiperazin-2-one (1.23 g, 5.58 mmol, 1 eq) in DCM (2 mL) was added DMF (0.2 mL) and SOCl2(810 uL, 11.17 mmol, 2 eq). The mixture was stirred at 25° C. for 30 min to give white suspension before it was concentrated under reduced pressure to give crude 1-(4-(chloromethyl)phenyl)-4-methyl-piperazin-2-one (1.27 g, 95% yield) as a white solid which was used in the next step directly. Step 5: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)malonate (1.4 g, 3.38 mmol, 1 eq) in DMF (15 mL) was added Cs2CO3(3.30 g, 10.14 mmol, 3 eq) and crude 1-(4-(chloromethyl)-phenyl)-4-methylpiperazin-2-one (1.21 g, 5.07 mmol, 1.5 eq). The mixture was stirred at 25° C. for 1 h before it was filtered and the filtrate was concentrated. The crude residue was purified by flash column chromatography on silica gel (0-50% MeOH in EtOAc) to give diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)malonate (1.02 g). Step 6: To a solution of diethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)-malonate (1.02 g, 1.65 mmol, 1 eq) in DCM (5 mL) and water (1 mL, 55.51 mmol, 34 eq) was added TFA (4.99 mL, 67.36 mmol, 41 eq). The mixture was stirred at 25° C. for 20 h before it was concentrated to give crude diethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxy-tetrahydrofuran-2-yl)methoxy)-2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)malonate (1.13 g) as an oil. To a solution of diethyl 2-[[(2R,3S,4R)-3-ethynyl-3,4,5-trihydroxy-tetrahydrofuran-2-yl]methoxy]-2-[[4-(4-methyl-2-oxo-piperazin-1-yl)phenyl]methyl]propanedioate (1.13 g, 2.11 mmol, 1 eq) in DCM (10 mL) was added 4-DMAP (25.78 mg, 211.00 umol, 0.1 eq), pyridine (1.07 mL, 13.2 mmol, 6.3 eq) and Ac2O (927 uL, 9.9 mmol, 4.7 eq). The mixture was stirred at 25° C. for 16 h before it was concentrated. The residue was diluted with EtOAc (20 mL) and 1N aq. HCl (10 mL). The organic layer was separated and the aqueous layer was extracted with EtOAc (4×20 mL). The combined organic layer was washed with water (50 mL), brine (50 mL) and dried over anhydrous Na2SO4, filtered and concentrated to provide crude diethyl 2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (674 mg) as an syrup. Step 7: To a solution of 2-chloro-9H-purin-6-amine (76 mg, 450.46 umol, 1.2 eq) in dichloroethane (3 mL) was added BSA (204 uL, 825.84 umol, 2.2 eq). The mixture was stirred at 65° C. for 0.5 h before it was cooled to 0° C. and crude diethyl 2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (248 mg, 375.38 umol, 1 eq) in dichloroethane (1 mL) and TMSOTf (102 uL, 563.07 umol, 1.5 eq) was added. The mixture was stirred at 65° C. for 2 h under N2atmosphere before it was quenched with saturated aq. NaHCO3(15 mL) and extracted with EtOAc (4×20 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by flash column chromatography on silica gel (0-30% of MeOH in EtOAc) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(4-methyl-2-oxo-piperazin-1-yl)benzyl)malonate (62 mg) as a white solid. Step 8: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)malonate (171 mg, 222.02 umol, 1 eq) in THF (2 mL) was added aq. LiOH (5.32 mg, 222.02 umol, 2 mL, 1 eq). The mixture was stirred at 25° C. for 2.5 h before it was acidified to pH 2-3 with 2N aq. HCl. The mixture was concentrated under reduced pressure. The crude residue was further purified by preparative HPLC to provide 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxy-tetrahydro-furan-2-yl)methoxy)-2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)-malonic acid (3.3 mg) as an off-white solid and 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-((2-((carboxymethyl)(methyl)amino)ethyl)-amino)benzyl)malonic acid (3.7 mg) as a white solid. 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydro-furan-2-yl)methoxy)-2-(4-(4-methyl-2-oxopiperazin-1-yl)benzyl)malonic acid:1H NMR (400 MHz, CD3OD) δ ppm 8.35 (s, 1H), 7.35 (d, J=8.3 Hz, 2H), 6.97 (d, J=8.5 Hz, 2H), 5.95 (d, J=5.5 Hz, 1H), 4.70-4.79 (m, 1H), 4.33 (dd, J=9.0, 3.3 Hz, 1H), 3.96-4.14 (m, 2H), 3.54-3.66 (m, 2H), 3.49 (s, 2H), 3.35 (s, 1H), 3.21 (s, 2H), 2.83 (t, J=5.4 Hz, 2H), 2.32-2.48 (m, 3H), 1.89 (s, 3H); LC/MS [M+H]=630.2. 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydro-furan-2-yl)methoxy)-2-(4-((2-((carboxymethyl)(methyl)amino)ethyl)amino)benzyl)malonic acid:1H NMR (400 MHz, CD3OD) δ ppm 8.56 (s, 1H), 7.03 (d, J=8.5 Hz, 2H), 6.49 (d, J=8.3 Hz, 2H), 6.01 (d, J=6.0 Hz, 1H), 4.70 (d, J=6.0 Hz, 1H), 4.34 (br d, J=4.3 Hz, 1H), 3.94 (dd, J=9.7, 6.9 Hz, 1H), 3.83 (dd, J=9.9, 2.4 Hz, 1H), 3.59 (s, 2H), 3.34-3.42 (m, 2H), 3.22-3.27 (m, 2H), 3.20 (s, 2H), 3.04 (s, 1H), 2.83 (s, 3H); LC/MS [M+H]=649.3. Example 156 Synthesis of 2-(((2R,3S,4R,5R)-5-(5-chloro-7-((2,4-dimethoxybenzyl)amino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid Step 1: To a mixture of 5,7-dichloro-3H-imidazo[4,5-b]pyridine (317 mg, 1.69 mmol, 1 eq) in MeCN (6 mL) was added BSA (1.04 mL, 4.22 mmol, 2.5 eq). The mixture was stirred at 65° C. under N2atmosphere for 0.5 h before it was cooled to 0° C. To the mixture was added diethyl 2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (1.20 g, 1.85 mmol, 1.1 eq) and TMSOTf (913.99 uL, 5.06 mmol, 3 eq). The mixture was stirred at 65° C. under N2atmosphere for 6 h before it was quenched with NaHCO3(15 mL). The reaction mixture was extracted with EtOAc (3×15 mL). The combined organic layer was washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude residue was purified by flash column chromatography on silica gel (0-100% of EtOAc in petroleum ether) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(5,7-dichloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (752 mg) as a foam. Step 2: To a mixture of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(5,7-dichloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (752 mg, 970.82 umol, 1 eq) and 2,4-dimethoxybenzylamine (292 uL, 1.94 mmol, 2 eq) and DIEA (507 uL, 2.91 mmol, 3 eq) were taken up into a microwave tube in NMP (4 mL). The sealed tube was irradiated in a microwave reactor at 130° C. for 2 h before it was diluted with H2O (10 mL) and extracted with EtOAc (3×15 mL). The combined organic layer was washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude residue was purified by flash column chromatography on silica gel (0-100% of EtOAc in petroleum) to provide diethyl 2-(((2R,3S,4R,5R)-5-(5-chloro-7-((2,4-dimethoxybenzyl)amino)-3H-imidazo-[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetra-hydropyrimidin-1 (2H)-yl)benzyl)malonate (249 mg, 26% yield) as a foam. Step 3: To a mixture of diethyl 2-(((2R,3S,4R,5R)-5-(5-chloro-7-((2,4-dimethoxybenzyl)-amino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)-methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (239 mg, 291.01 umol, 1 eq) in THF (4 mL) and H2O (3 mL) was added LiOH (69.69 mg, 2.91 mmol, 10 eq). The mixture was stirred at 25° C. for 20 h before it was acidified to pH 6-7 with 2N aqueous HCl and concentrated under reduced pressure. The crude residue was purified by preparative HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.1% TFA)-ACN]; B %: 25%-65%, 10 min) and dried by lyophilization to provide 2-(((2R,3S,4R,5R)-5-(5-chloro-7-((2,4-dimethoxybenzyl)amino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid (9.5 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.23 (s, 1H), 7.27 (d, J=8.44 Hz, 2H), 7.21 (d, J=8.31 Hz, 1H), 7.00 (d, J=8.44 Hz, 2H), 6.58 (d, J=2.32 Hz, 1H), 6.46-6.51 (m, 2H), 6.04 (d, J=7.34 Hz, 1H), 4.73 (d, J=7.34 Hz, 1H), 4.43 (s, 2H), 4.27 (t, J=3.06 Hz, 1H), 4.01 (d, J=2.93 Hz, 2H), 3.87 (s, 3H), 3.78 (s, 3H), 3.37-3.47 (m, 2H), 3.31-3.37 (m, 2H), 3.22 (t, J=5.87 Hz, 2H), 3.03 (s, 1H), 1.78-1.85 (m, 2H); LC/MS [M+H]=765.1. Example 157 Synthesis of 2-(((2R,3S,4R,5R)-5-(7-amino-5-chloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid To a mixture of 2-(((2R,3S,4R,5R)-5-(5-chloro-7-((2,4-dimethoxybenzyl)amino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid (160 mg, 209.11 umol, 1 eq) in DCM (3 mL) was added TFA (1 mL, 13.51 mmol, 64.59 eq). The mixture was stirred at 25° C. for 2 h before it was concentrated. The residue was purified by preparative HPLC (column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)-ACN]; B %: 20%-40%, 10 min) and dried by lyophilization to provide 2-(((2R,3S,4R,5R)-5-(7-amino-5-chloro-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)-methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonic acid (5.4 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.34 (s, 1H), 7.28 (d, J=8.31 Hz, 2H), 7.02 (d, J=8.44 Hz, 2H), 6.49 (s, 1H), 6.07 (d, J=7.21 Hz, 1H), 4.78 (d, J=7.21 Hz, 1H), 4.30 (t, J=3.18 Hz, 1H), 3.93-4.08 (m, 2H), 3.45-3.54 (m, 2H), 3.35-3.49 (m, 4H), 3.04 (s, 1H), 1.94-2.00 (m, 2H); LC/MS [M+H]=615.1. Example 158 Synthesis of 2-((1H-pyrazol-5-yl)methyl)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Proceeding as described in Example 1 above but substituting benzyl bromide with tert-butyl 5-(bromomethyl)-1H-pyrazole-1-carboxylate provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.46 (s, 1H), 7.32 (s, 1H), 6.17 (s, 1H), 6.02 (d, J=7.13 Hz, 1H), 4.83 (s, 1H), 4.34 (s, 1H), 3.98-4.11 (m, 2H), 3.43-3.54 (m, 2H), 2.95 (s, 1H); LC/MS [M+H]=508.1. Example 159 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(1-tosyl-1H-pyrazol-4-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a mixture of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (1.3 g, 1.92 mmol, 1 eq) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-tosyl-1H-pyrazole (669 mg, 1.92 mmol, 1 eq) in dioxane (10 mL) and H2O (3 mL) under a N2atmosphere was added Pd(PPh3)4(222 mg, 192 umol, 0.1 eq) and Cs2CO3(1.88 g, 5.76 mmol, 3 eq). The mixture was stirred at 100° C. for 3 h before it was filtered and the filtrate was concentrated under reduced pressure. The crude residue was purified by flash silica gel column chromatography (0-50% of EtOAc in petroleum ether) to provide diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(1-tosyl-1H-pyrazol-4-yl)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (170 mg) as a foam. Step 2: To a solution diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(1-tosyl-1H-pyrazol-4-yl)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (50 mg, 58 umol, 1 eq) in THF (1 mL) was added 1M aq. LiOH (1 mL, 18 eq). The mixture was stirred at 25° C. for 14 h before it was diluted with EtOAc (10 mL) and water (10 mL). The aqueous phase was adjusted to pH 2-3 with 2M aq. HCl solution and extracted with EtOAc (2×40 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude residue was purified by preparative HPLC (column: YMC-Triart Prep C18 150*40 mm*7 um; mobile phase: [water (0.225% FA)-AC]; B %: 40%-60%, 10 min) to provide 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(1-tosyl-1H-pyrazol-4-yl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid (2.0 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.98 (s, 1H), 8.69 (s, 2H) 7.47 (d, J=8.25 Hz, 2H), 7.30 (d, J=6.88 Hz, 2H), 7.07-7.19 (m, 4H), 6.92 (d, J=8.00 Hz, 2H), 6.24 (d, J=7.63 Hz, 1H), 5.85 (d, J=7.63 Hz, 1H), 4.35 (t, J=2.56 Hz, 1H), 4.03-4.09 (m, 1H), 3.90 (d, J=10.63 Hz, 1H), 3.40-3.50 (m, 1H), 2.92 (s, 1H), 2.05 (s, 3H); LC/MS [M+H]=723.2. Example 160 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxo-1-propyl-1,2-dihydropyridin-3-yl)benzyl)malonic acid Proceeding as described in Example 19 above but substituting diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate with diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-((tert-butoxycarbonyl)(methyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-tetrahydrofuran-2-yl)methoxy)malonate provided the title compound as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.44 (s, 1H), 8.25 (d, J=4.3 Hz, 1H), 7.68 (dd, J=6.7, 1.8 Hz, 1H), 7.29-7.51 (m, 3H), 7.18 (d, J=7.5 Hz, 2H), 6.28 (t, J=6.8 Hz, 1H), 6.21 (s, 1H), 6.01 (d, J=6.8 Hz, 1H), 5.83 (d, J=7.3 Hz, 1H), 4.82 (s, 1H), 4.17 (dd, J=5.1, 2.6 Hz, 1H), 3.96 (s, 1H), 3.89 (t, J=7.3 Hz, 2H), 3.79 (s, 1H), 3.58 (s, 1H), 3.25 (s, 1H), 2.90 (d, J=4.5 Hz, 3H), 1.67 (sxt, J=7.3 Hz, 2H), 0.88 (t, J=7.4 Hz, 3H); LC/MS [M+H]=667.1. Example 161 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-(1-hydroxyethyl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid Step 1: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-acetyl-6-amino-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (48 mg, 72.11 umol, 1 eq) in MeOH (2 mL) at 0° C. was added NaBH4(4.09 mg, 108.17 umol, 1.5 eq). The solution was stirred at 0° C. for 1 h. Additional NaBH4(4.1 mg) was added to the reaction mixture and it was stirred at 0° C. for 0.5 h before it was diluted with water (6 mL) and extracted with ethyl acetate (3×6 mL). The combined organic layer was dried by Na2SO4, filtered and concentrated. The crude residue was purified by preparative TLC (ethyl acetate) to give diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-(1-hydroxyethyl)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (20 mg) as a syrup. Step 2: To a solution of diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-(1-hydroxyethyl)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (16 mg, 23.96 umol, 1 eq) in THF (2 mL) was added 1M aq. LiOH (0.5 mL, 21 eq). The mixture was stirred at 20° C. for 3 h before it was acidified to pH 5 with 1N aq. HCl solution. The mixture was extracted with ethyl acetate (5×3 mL). The combined organic layer was concentrated. The crude residue was purified by preparative HPLC (Column: YMC-Triart Prep C18 150*40 mm*7 um; mobile phase: [water (0.225% FA)-ACN]; B %: 13%-33%, 10 min) and dried by lyophilization to give 2-(((2R,3S,4R,5R)-5-(6-amino-2-(1-hydroxyethyl)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-benzylmalonic acid (4.0 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.49 (d, J=8.88 Hz, 1H), 7.18-7.25 (m, 2H), 7.07-7.14 (m, 3H), 6.09 (dd, J=9.82, 6.94 Hz, 1H), 4.76-4.84 (m, 2H), 4.29-4.36 (m, 1H), 3.89-4.03 (m, 2H), 3.32-3.45 (m, 1H), 3.24-3.28 (m, 1H), 3.02 (d, J=10.13 Hz, 1H), 1.52 (d, J=6.50 Hz, 3H); LC/MS [M+H]=528.1. Example 162 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(1-methyl-2-oxo-1,2-dihydropyridin-3-yl)benzyl)malonic acid Proceeding as described in Example 19 above but substituting 3-(4-(bromomethyl)-phenyl)-1-propylpyridin-2 (1H)-one with 3-(4-(bromomethyl)phenyl)-1-methylpyridin-2 (1H)-one provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.12 (s, 1H), 7.55-7.53 (m, 1H), 7.38-7.28 (m, 5H), 6.33 (t, J=6.8 Hz, 1H), 5.94 (d, J=7.6 Hz, 1H), 4.75 (d, J=7.6 Hz, 1H), 4.27-4.26 (m, 1H), 4.09-4.01 (m, 2H), 3.55 (s, 3H), 3.53-3.44 (m, 2H), 3.05 (s, 1H); LC/MS [M+H]=625.0. Example 163 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(6-(3-carboxypropyl)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a solution of 2,6-dichloro-9-(tetrahydro-2H-pyran-2-yl)-9H-purine (4.00 g, 14.65 mmol, 1 eq) and Pd(Ph3P)4 (1.69 g, 1.46 mmol, 0.1 eq) in THF (30 mL) under N2atmosphere at 0° C. was added a solution of 0.5 M (4-ethoxy-4-oxobutyl)zinc(II) bromide (73.23 mL, 36.61 mmol, 2.5 eq) dropwise. The mixture was stirred from 0-25° C. over 16 h before it was cooled to 0° C. and quenched with 0.5N aq. HCl solution. The reaction mixture was extracted with EtOAc (3×100 mL). The combined organic extract was washed with H2O (100 mL), brine (50 mL), dried over Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (10-75% EtOAc in petroleum ether) to provide ethyl 4-(2-chloro-9-(tetrahydro-2H-pyran-2-yl)-9H-purin-6-yl)butanoate (2.83 g). Step 2: To a solution of ethyl 4-(2-chloro-9-(tetrahydro-2H-pyran-2-yl)-9H-purin-6-yl)-butanoate (1.50 g, 4.25 mmol) in DCM (15 mL) was added TFA (10 mL). The mixture was stirred at 25° C. for 7 h before it was concentrated under reduced pressure. The residue was re-taken up in H2O (50 mL) and neutralized to pH 7 with saturated aq. NaHCO3. The resulting mixture was extracted with EtOAc (3×75 mL). The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated to provide ethyl 4-(2-chloro-9H-purin-6-yl)butanoate (1.05 g). Steps 3-4: Proceeding as described in Example 5 above but substituting uracil with ethyl 4-(2-chloro-9H-purin-6-yl)butanoate provided the title compound as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.74 (s, 1H), 7.27-7.24 (m, 2H), 7.09-7.08 (m, 3H), 6.09 (d, J=7.6 Hz, 1H), 5.05 (d, J=7.6 Hz, 1H), 4.31-4.14 (m, 1H), 4.10-4.06 (m, 2H), 3.18-3.16 (m, 3H), 2.41 (t, J=7.2 Hz, 2H), 2.17-2.15 (m, 2H), 1.22 (t, J=7.6 Hz, 2H); LC/MS [M+H]=589.1. Examples 164 and 165 Synthesis of (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-ethoxy-3-oxo-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)propanoic acid and (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-ethoxy-3-oxo-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)propanoic acid Step 1: To a suspension of 2-chloro-9H-purin-6-amine (1.04 g, 6.15 mmol, 1.7 eq) in MeCN (10 mL) at 25° C. was added N,O-bis(trimethylsilyl)acetamide (BSA) (3.1 mL, 0.0127 mol, 3.5 eq). The resulting suspension was heated at 85° C. for 30 min as it became clear. The reaction mixture was allowed to cool to 25° C. followed by addition of a solution of diethyl 2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-2-(((2R,3R,4R)-3,4,5-triacetoxy-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (2.34 g, 0.0036 mol, 1.0 eq) in MeCN (10 mL) and TMSOTf (1.12 mL, 0.00615 mol, 1.7 eq) dropwise. The reaction mixture was then heated at 70-80° C. overnight as all of the starting material was consumed. The reaction was allowed to cool to 25° C. before it was diluted with MeCN (100 mL) and quenched with saturated aq. NaHCO3solution (150 mL). The insoluble was removed by filtration. The organic layer of the filtrate was separated, washed with H2O (50 mL), brine (50 mL), dried over Na2SO4and concentrated. The crude residue was purified by flash silica gel column chromatography (0-5% MeOH in CH2Cl2) to provide diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (1.1 g, 40% yield) as a white solid. Step 2: To a solution of diethyl 2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)malonate (400 mg, 0.53 mmol, 1 eq) in THF (4 mL) and H2O (4 mL) at 0° C. was added LiOH monohydrate (89 mg, 2.12 mmol, 4 eq). The resulting mixture was stirred at room temperature overnight before the organic volatile was removed under reduced pressure. The mixture was cooled to 0° C. and acidified to pH 6 with 1N aq. HCl solution and concentrated under reduced pressure. The crude residue was purified by preparative reversed-phase HPLC to provide a pair of diastereomers as a white solid: (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydro-furan-2-yl)methoxy)-3-ethoxy-3-oxo-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)-propanoic acid and (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-ethoxy-3-oxo-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)propanoic acid which the stereo configuration was arbitrarily assigned. In addition, Example 9 was also isolated as a white solid. (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-ethoxy-3-oxo-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)propanoic acid:1H NMR (300 MHz, CD3OD) δ 8.32 (s, 1H), 7.28 (d, J=8.1 Hz, 2H), 7.01 (d, J=8.1 Hz, 2H), 6.00 (d, J=7.5 Hz, 1H), 4.84 (d, J=7.5, 1H), 4.28-4.06 (m, 3H), 3.99-3.95 (m, 2H), 3.52-3.35 (m, 6H), 3.08 (s, 1H), 2.02-1.97 (m, 2H), 1.21 (t, J=7.1, 3H); LC/MS [M+H]=644.05. (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-ethoxy-3-oxo-2-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)benzyl)propanoic acid:1H NMR (300 MHz, CD3OD) δ 8.08 (s, 1H), 7.26-7.29 (d, J=6.8 Hz, 2H), 7.03-7.01 (d, J=7.23 Hz, 2H), 5.96-5.99 (d, J=7.14 Hz, 1H), 4.75-4.77 (d, J=7.5, 1H), 4.02-4.24 (m, 5H), 3.32-3.66 (m, 6H), 3.15 (s, 1H), 1.95-2.19 (m, 2H), 1.22-1.27 (m, 3H); LC/MS [M+H]=644.05. Examples 166 and 167 Synthesis of (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)propanoic acid and (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)propanoic acid The crude product of Example 9 from the work up was dried in the vacuum oven at 60° C. for 2 days before it was purified by preparative HPLC to provide a pair of diastereomers: (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)-propanoic acid and (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)-propanoic acid which the stereo configuration was arbitrarily assigned. Both were isolated as white solids. (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)propanoic acid: 1H NMR (300 MHz, CD3OD) δ 8.38 (s, 1H), 7.22 (d, J=8.3 Hz, 2H), 7.04 (d, J=8.3 Hz, 2H), 5.92 (d, J=7.3 Hz, 1H), 4.36-4.32 (m, 1H), 4.30 (d, J=7.3 Hz, 1H), 4.17 (t, J=2.3 Hz, 1H), 4.07-4.03 (m, 1H), 3.80-3.75 (m, 1H), 3.54-3.49 (m, 2H), 3.33-3.31 (m, 2H), 3.25-3.19 (m, 1H), 3.12 (s, 1H), 3.09-3.02 (m, 1H), 2.02-1.97 (m, 2H); LC/MS [M+H]=572.0. (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)propanoic acid: 1H NMR (300 MHz, CD3OD) δ 8.47 (s, 1H), 7.26 (d, J=8.2 Hz, 2H), 7.15 (d, J=8.3 Hz, 2H), 5.97 (d, J=7.0 Hz, 1H), 4.97 (d, J=7.0 Hz, 1H), 4.31 (t, J=6.4 Hz, 1H), 4.20 (t, J=3.4 Hz, 1H), 3.91 (d, J=3.4 Hz, 1H), 3.65 (t, J=6.0 Hz, 2H), 3.39-3.33 (m, 2H), 3.23-3.17 (m, 2H), 3.08 (s, 1H), 3.06-3.01 (m, 1H), 2.10-1.95 (m, 2H); LC/MS [M+H]=572.0. Example 168 Synthesis of 2-benzyl-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(hydroxyamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid Step 1: To a solution of diethyl 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-7-(hydroxyamino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-malonate (98 mg, 0.144 mmol) in dioxane (2 mL) was added an aqueous solution of hydroxylamine (0.1 mL, 1.6 mmol, 16 M) and Et3N (35 uL, 0.16 mmol). The reaction mixture was stirred for 2.5 h and then it was diluted with EtOAc (15 mL) and H2O (5 mL). The organic layer was separated, washed with H2O (20 mL), brine (20 mL), dried over Na2SO4and concentrated to provide crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(hydroxyamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)-malonate (88 mg) as an off-solid which was used in the next step without further purification. Step 2: To a solution of crude diethyl 2-benzyl-2-(((2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-(hydroxyamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)malonate (88 mg, 0.14 mmol) in a mixture of THF (0.2 mL), MeOH (0.62 mL) and H2O (0.15 mL) was added LiOH·H2O (31 mg, 0.75 mmol). The resulting mixture was stirred at 25° C. for 5.5 h before the organic volatile was removed under reduced pressure. The aq. layer was cooled to 0° C. and acidified to pH 6.5 with 1N aq. HCl solution before it was concentrated. The crude residue was purified by preparative reversed-phase HPLC to provide 2-benzyl-2-(((2R,3S,4R,5R)-5-(5-chloro-7-(hydroxyamino)-3H-imidazo[4,5-b]pyridin-3-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)malonic acid (17 mg) as a reddish solid. 1H NMR (CD3OD, 300 MHz) δ 8.25 (s, 1H), 7.25-7.28 (m, 2H), 7.05 (m, 3H), 6.43 (s, 1H), 6.06-6.08 (d, J=7.17 Hz, 1H), 4.95-4.98 (d, J=7.05 Hz, 1H), 4.32 (s, 1H), 4.05-4.11 (m, 2H), 3.89-3.93 (m, 1H), 3.31-3.39 (m, 2H), 2.99 (s, 1H), 1.30-1.33 (m, 6H); LC/MS [M+H]=533.1. Example 169 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)-3-(thiophen-3-yl)propanoic acid Step 1: To a solution of ethyl 2-(thiazol-4-yl)acetate (2 g, 11.7 mmole) in CH3CN (15 mL) at 0° C. was added DBU (2.62 ml, 17.6 mmole) and 4-acetamidibenzene sulfonylazide (3.4 g, 14.1 mmole) in CH3CN (10 mL). The reaction mixture was stirred at room temperature for 1.5 h before it was concentrated under reduced pressure to dryness. The resulting crude was purified by silica gel column chromatography (0-40% EtOAc in hexanes) to provide ethyl 2-diazo-2-(thiazol-4-yl)acetate (2.0 g). Step 2: To a mixture of (3aR,5R,6R,6aR)-6-ethynyl-5-(hydroxymethyl)-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-6-yl acetate (7 g, 27.32 mmol, 1 eq) in DCE (15 mL) was added Rh(OAc)2(603.69 mg, 2.73 mmol, 0.1 eq) and ethyl 2-diazo-2-(thiazol-4-yl)acetate (6.46 g, 32.78 mmol, 1.2 eq) in DCE (15 mL) dropwise at 0° C. The mixture was stirred at 25° C. under N2atmosphere for 14 h before the insoluble was filtered and the filtrate was concentrated under reduced pressure. The crude residue was purified by flash silica gel column chromatography (0-50% of EtOAc in petroleum ether) to provide ethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(thiazol-4-yl)acetate (10.81 g, 93% yield) as an oil. Step 3: To a mixture of ethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(thiazol-4-yl)acetate (2.69 g, 6.33 mmol, 1 eq) in DMF (5 mL) was added Cs2CO3(6.18 g, 18.98 mmol, 3 eq). The mixture was stirred at 25° C. under N2atmosphere for 0.5 h before 3-(bromomethyl)thiophene (2.8 g, 15.81 mmol, 2.5 eq) was added. The resulting mixture was stirred at 25° C. for 14 h before the insoluble was filtered and the filtrate was diluted with H2O (15 mL) and extracted with EtOAc (3×15 mL). The combined organic layer was washed with saturated aq. NH4Cl (3×15 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude residue was purified by flash silica gel column chromatography (0-50% of EtOAc in petroleum) to provide ethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(thiazol-4-yl)-3-(thiophen-3-yl)propanoate (2.27 g, 69% yield) as an oil. Step 4: To a mixture of ethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(thiazol-4-yl)-3-(thiophen-3-yl)propanoate (2.27 g, 4.35 mmol, 1 eq) in DCM (5 mL) and H2O (0.5 mL) was added TFA (5 mL, 67.53 mmol, 15.5 eq). The mixture was stirred at 15° C. under N2atmosphere for 14 h before it was adjusted to 7-8 pH with saturated aq. NaHCO3(50 mL) and concentrated under reduced pressure. The residue was diluted with H2O (5 mL) and extracted with EtOAc (4×15 mL). The combined organic layer was washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to provide crude ethyl 2-[[(2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxy-tetrahydrofuran-2-yl]methoxy]-2-thiazol-4-yl-3-(3-thienyl)propanoate (1.82 g) as a syrup. To a solution of ethyl 2-[[(2R,3S,4R)-3-acetoxy-3-ethynyl-4,5-dihydroxy-tetrahydro-furan-2-yl]methoxy]-2-thiazol-4-yl-3-(3-thienyl)propanoate (1.82 g, 3.78 mmol, 1 eq) in pyridine (8 mL) under a N2atmosphere at 0° C. was added 4-DMAP (1.39 g, 11.34 mmol, 3 eq) and Ac2O (2.83 mL, 30.24 mmol, 8 eq). The mixture was stirred at 15° C. for 15 before it was diluted with H2O (20 mL) and extracted with EtOAc (3×15 mL). The combined organic layer was washed with 10% CuSO4solution (2×15 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give crude (3R,4R,5R)-5-(((1-ethoxy-1-oxo-2-(thiazol-4-yl)-3-(thiophen-3-yl)propan-2-yl)oxy)methyl)-4-ethynyltetrahydrofuran-2,3,4-triyl triacetate (2.42 g) as a syrup. Step 5: To a mixture of 2-chloro-N-isopropyl-9H-purin-6-amine (606.20 mg, 2.86 mmol, 1 eq) in DCE (20 mL) was added BSA (1.77 mL, 7.16 mmol, 2.5 eq). The mixture was stirred at 85° C. under a N2atmosphere for 0.5 h before it was allowed to cool to 0° C. and followed by addition of crude (3R,4R,5R)-5-(((1-ethoxy-1-oxo-2-(thiazol-4-yl)-3-(thiophen-3-yl)-propan-2-yl)oxy)methyl)-4-ethynyltetrahydrofuran-2,3,4-triyl triacetate (1.62 g, 2.86 mmol, 1 eq) and TMSOTf (1.55 mL, 8.59 mmol, 3 eq). The resulting mixture was stirred at 65° C. under N2for 14 h before it was quenched with saturated aq. NaHCO3(20 mL). The reaction mixture was diluted with H2O (10 mL) and extracted with DCM (3×20 mL). The combined organic layer was washed with brine (3×15 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude residue was purified by flash silica gel column chromatography (0-50% of EtOAc in petroleum ether) to provide (2R,3R,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-2-(((1-ethoxy-1-oxo-2-(thiazol-4-yl)-3-(thio-phen-3-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (402 mg, crude) as a syrup. Step 6: To a mixture of (2R,3R,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-2-(((1-ethoxy-1-oxo-2-(thiazol-4-yl)-3-(thiophen-3-yl)propan-2-yl)oxy)methyl)-3-ethynyltetra-hydrofuran-3,4-diyl diacetate (384 mg, crude) in THF (2 mL) and H2O (1 mL) was added LiOH (128 mg, 5.35 mmol). The mixture was stirred at 50° C. for 6 h before it was diluted with H2O (40 mL) and extracted with EtOAc (10 mL). The aqueous phase was acidified to pH 2-3 with 2 N aqueous HCl until pH-2-3 and then concentrated under reduced pressure. The crude residue was purified by preparative HPLC (column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)-ACN]; B %: 40%-60%, 10 min) and dried by lyophilization to provide a diastereomeric mixture (ca. 1:1) of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)-3-(thiophen-3-yl)propanoic acid (17.5 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.92-9.01 (m, 1H), 8.04-8.28 (m, 1H), 7.60-7.71 (m, 1H), 7.10-7.15 (m, 1H), 6.98-7.06 (m, 1H), 6.77-6.94 (m, 1H), 5.90-6.02 (m, 1H), 4.91-5.06 (m, 2H), 4.40 (br s, 1H), 4.19-4.32 (m, 1H), 3.89-3.99 (m, 1H), 3.65-3.87 (m, 3H), 2.89-3.02 (m, 1H), 1.25-1.35 (m, 6H); LC/MS [M+H]=605.2. Examples 170 & 171 Synthesis of (S)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid and (R)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid Proceeding as described in Example 169 above but substituting 3-(bromomethyl)thiophene with benzyl bromide provided a pair of diastereomeric products which the stereo configuration was assigned arbitrarily. Both products were purified by preparative HPLC and isolated as white solids. (S)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic:1H NMR (CD3OD, 300 MHz) δ 8.99 (s, 1H), 7.91 (s, 1H), 7.70 (s, 1H), 7.07-7.23 (m, 5H), 5.91-5.94 (d, J=6.9 Hz, 1H), 4.87-4.90 (d, J=7.0 Hz, 1H), 4.21-4.45 (m, 2H), 3.59-3.94 (m, 4H), 3.02 (s, 1H), 1.29-1.31 (d, J=6.48 Hz, 6H); LC/MS [M+H]=599.0. (R)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic:1H NMR (CD3OD, 300 MHz) δ 8.97-8.98 (d, J=1.83 Hz, 1H), 8.07 (s, 1H), 7.573-7.579 (d, J=1.86 Hz, 1H), 6.94-7.09 (m, 5H), 5.97-5.99 (d, J=7.17 Hz, 1H), 4.98-5.00 (d, J=7.23 Hz, 1H), 4.40-4.42 (m, 1H), 4.27-4.29 (t, J=3.84 Hz, 1H), 3.93-3.97 (m, 2H), 3.59-3.81 (q, J=14.31, 37.47 Hz, 2H), 2.95 (s, 1H), 1.29-1.33 (d, J=6.39 Hz, 6H); LC/MS [M+H]=599.0. Examples 172 & 173 Synthesis of (S)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid and (R)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid Proceeding as described in Examples 170 and 171 above but substituting 2-chloro-N-isopropyl-9H-purin-6-amine with 2-chloro-N-(cyclopropylmethyl)-9H-purin-6-amine provided a pair of diastereomeric products which the stereo configuration was assigned arbitrarily. Both products were purified by preparative HPLC and isolated as white solids. (S)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid: 1H NMR (CD3OD, 300 MHz) δ 8.99-8.99 (d, J=1.95 Hz, 1H), 7.93 (s, 1H), 7.69-7.70 (d, J=1.86 Hz, 1H), 7.03-7.23 (m, 5H), 5.92-5.94 (d, J=6.96 Hz, 1H), 4.87-4.89 (d, J=7.11 Hz, 1H), 4.19-4.22 (m, 1H), 3.59-3.94 (m, 4H), 3.42-3.43 (m, 2H), 3.02 (s, 1H), 1.12-1.22 (m, 1H), 0.54-0.61 (m, 2H), 0.32-0.37 (m, 2H); LC/MS [M+H]=611.0. (R)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-((cyclopropylmethyl)amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid: 1H NMR (CD3OD, 300 MHz) δ 8.96-8.97 (d, J=1.89 Hz, 1H), 8.07 (s, 1H), 7.55-7.56 (d, J=2.07 Hz, 1H), 6.94-7.11 (m, 5H), 5.97-5.99 (d, J=7.17 Hz, 1H), 4.98-5.00 (d, J=7.29 Hz, 1H), 4.27-4.29 (t, J=3.66 Hz, 1H), 3.94-3.95 (m, 2H), 3.59-3.80 (q, J=14.64, 32.46 Hz, 2H), 3.39-3.50 (m, 2H), 2.96 (s, 1H), 1.13-1.23 (m, 1H), 0.55-0.62 (m, 2H), 0.32-0.38 (m, 2H); LC/MS [M+H]=611.0. Example 174 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)-3-(4-(trifluoromethoxy)phenyl)propanoic acid Proceeding as described in Example 169 above but substituting 3-(bromomethyl)thiophene with 1-(bromomethyl)-4-(trifluoromethoxy)benzene provided the title compound as a mixture of diastereomers (ca. 1:1) and isolated as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.95-9.02 (m, 1H), 8.05-8.27 (m, 1H), 7.59-7.74 (m, 1H), 7.15-7.32 (m, 2H), 6.78-6.98 (m, 2H), 5.89-6.00 (m, 1H), 4.92-5.10 (m, 1H), 4.32-4.46 (m, 1H), 4.22-4.32 (m, 1H), 3.76-3.98 (m, 2H), 3.57-3.71 (m, 2H), 2.98-3.04 (m, 1H), 1.26-1.32 (m, 6H); LC/MS [M+H]=682.8. Example 175 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)-2-(thiazol-4-yl)propanoic acid Step 1: A mixture of 1-(bromomethyl)-4-nitro-benzene (7.62 g, 35.26 mmol, 3 eq) and NaI (352.31 mg, 2.35 mmol, 0.2 eq) in DMF (50 mL) was stirred at 15° C. for 30 min. Then this mixture was added to a solution of ethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-2-(thiazol-4-yl)acetate (5 g, 11.75 mmol, 1 eq) and Cs2CO3(19.15 g, 58.76 mmol, 5 eq) in DMF (50 mL) at 15° C. was stirred for 30 min. The resulting mixture was stirred for 8 h before it was quenched by water (200 mL). The mixture was extracted with EtOAc (4×30 mL). The combined organic layer was washed with water (3×100 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by flash column chromatography on silica gel (0-40% of EtOAc in petroleum ether) to provide ethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyl-tetrahydrofuro [2,3-d][1,3]dioxol-5-yl)methoxy)-3-(4-nitrophenyl)-2-(thiazol-4-yl)propanoate (4.89 g) as a syrup. Step 2: To a solution of ethyl 2-(((3aR,5R,6R,6aR)-6-acetoxy-6-ethynyl-2,2-dimethyltetra-hydrofuro[2,3-d][1,3]dioxol-5-yl)methoxy)-3-(4-nitrophenyl)-2-(thiazol-4-yl)propanoate (4.89 g, impure) in DCM (25 mL) and H2O (2.5 mL) was added TFA (25 mL 337.65 mmol). The mixture was stirred at 30° C. for 23 h before it was diluted with water (100 mL) and the resulting mixture was extracted with DCM (6×30 mL). The combined organic layer was washed with saturated aq. NaHCO3(2×100 mL), dried over anhydrous Na2SO4, filtered and concentrated to provide crude ethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxytetrahydro-furan-2-yl)methoxy)-3-(4-nitrophenyl)-2-(thiazol-4-yl)propanoate (4.56 g) as a brown oil. To a solution of crude ethyl 2-(((2R,3S,4R)-3-ethynyl-3,4,5-trihydroxytetrahydro-furan-2-yl)methoxy)-3-(4-nitrophenyl)-2-(thiazol-4-yl)propanoate (4.56 g) in DCM (50 mL) was added 4-DMAP (232.86 mg, 1.91 mmol), pyridine (6.15 mL, 76.24 mmol) and Ac2O (8.93 mL, 95.30 mmol) dropwise. The mixture was stirred at 15° C. for 19 h before it was quenched with water (100 mL) and the resulting mixture was extracted with DCM (4×30 mL). The combined organic layer was washed with water (3×100 mL), and dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (10-55% EtOAc in petroleum ether) to provide (3R,4R,5R)-5-(((1-ethoxy-3-(4-nitrophenyl)-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-4-ethynyltetrahydrofuran-2,3,4-triyl triacetate (1.02 g) as a yellow oil. Step 3: To a solution of 2,6-dichloro-9H-purine (382.64 mg, 2.02 mmol) in MeCN (5 mL) was added BSA (1.04 mL, 4.22 mmol). The suspension was stirred at 65° C. for 0.5 h as it became clear. The resulting solution was cooled down to 0° C. and followed by addition of a solution of (3R,4R,5R)-5-(((1-ethoxy-3-(4-nitrophenyl)-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-4-ethynyltetrahydrofuran-2,3,4-triyl triacetate (1.02 g) in MeCN (5 mL) and TMSOTf (4.22 mmol, 762.15 uL). Then the mixture was stirred at 65° C. for 1 h before it was quenched with saturated aq. NaHCO3(40 mL) and the resulting mixture was extracted EtOAc (4×20 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to provide crude (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-2-(((1-ethoxy-3-(4-nitrophenyl)-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.71 g) as a yellow solid. Step 4: To a solution of crude (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-2-(((1-ethoxy-3-(4-nitrophenyl)-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.0 g) in MeOH (20 mL) in a seal tube was added NH4·OH (28.04 mmol, 4.00 mL, 27% concentration). The mixture was sealed and stirred at 100° C. for 1.5 h before it was allowed to cool and diluted with water (20 mL) and the resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give crude ethyl 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-nitrophenyl)-2-(thiazol-4-yl)propanoate (752 mg) as a yellow solid. Step 5: To a solution of crude ethyl 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-nitrophenyl)-2-(thiazol-4-yl)-propanoate (752 mg), 4-DMAP (58.33 mg, 477.44 umol) and Et3N (7.16 mmol, 996.81 uL) in DMF (8 mL) at 0° C. was added Boc2O (1.04 g, 4.77 mmol). The mixture was stirred at 20° C. for 2 h before it was diluted with water (40 mL) and the resulting mixture was extracted with EtOAc (4×20 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give crude ethyl 2-(((2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyl-3-hydroxytetrahydrofuran-2-yl)methoxy)-3-(4-nitrophenyl)-2-(thiazol-4-yl)propanoate (911 mg) as a brown solid. Step 6: To a solution of crude ethyl 2-(((2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)-amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyl-3-hydroxytetra-hydrofuran-2-yl)methoxy)-3-(4-nitrophenyl)-2-(thiazol-4-yl)propanoate (711 mg) in EtOH (7 mL) was added saturated aq. NH4Cl (764.21 umol, 7 mL) and iron (426.77 mg, 7.64 mmol). The mixture was stirred at 50° C. for 2 h before it was filtered through a pad of Celite and the filtrate was concentrated. Then the crude residue was taken up in water (20 mL) and the resulting mixture was extracted with EtOAc (3×15 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give crude ethyl 3-(4-amino-phenyl)-2-(((2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyl-3-hydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)propanoate (552 mg) as a brown solid. Step 7: To a solution of crude ethyl 3-(4-aminophenyl)-2-(((2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyl-3-hydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)propanoate (552 mg) in DCM (5 mL) was added 1-chloro-3-isocyanato-propane (109.94 mg, 919.60 umol). The mixture was stirred at 15° C. for 16 h before it was concentrated under reduce pressure. The crude residue was purified by flash column chromatography on silica gel (20-100% EtOAc in petroleum ether) to provide ethyl 2-(((2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyl-3-hydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(3-(3-chloropropyl)ureido)phenyl)-2-(thiazol-4-yl)propanoate (283 mg) as an off-white solid. Step 8: To a solution of ethyl 2-(((2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-4-((tert-butoxycarbonyl)oxy)-3-ethynyl-3-hydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(3-(3-chloropropyl)ureido)phenyl)-2-(thiazol-4-yl)propanoate (283 mg, 277.47 umol, 1 eq) in THF (3 mL) was added NaH (55.49 mg, 1.39 mmol, 60% in mineral oil, 5 eq). The mixture was stirred at 15° C. for 5 h before it was quenched with H2O (1.5 mL). To this mixture was added NaOH (166.48 mg, 4.16 mmol, 15 eq) and the resulting mixture was stirred at 40° C. for 48 h before the organic volatile was removed under reduced pressure. The aq. layer was acidified with 2N aq. HCl (1 mL) and concentrated under reduced pressure to give crude 2-(((2R,3S,4R,5R)-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)-2-(thiazol-4-yl)propanoic acid (238 mg) as a yellow solid. Step 9: A mixture of crude 2-(((2R,3S,4R,5R)-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetra-hydropyrimidin-1 (2H)-yl)phenyl)-2-(thiazol-4-yl)propanoic acid (238 mg) in DCM (2 mL) was added TFA (9.45 mmol, 0.7 mL). The mixture was stirred at 15° C. for 2 h before it was concentrated under reduced pressure. The crude residue was purified by Preparative HPLC ([water (0.225% FA)-ACN]; B %: 20%-40%, 10 min) to provide a diastereomeric mixture (ca. 1:1) of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)-2-(thiazol-4-yl)propanoic acid (22.9 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 9.01 (m, 1H), 7.98-8.32 (m, 1H), 7.59-7.83 (m, 1H), 6.91-7.34 (m, 4H), 5.88-6.07 (m, 1H), 4.72-4.96 (m, 1H), 4.13-4.32 (m, 1H), 3.60-4.00 (m, 4H), 3.43-3.56 (m, 2H), 3.35-3.42 (m, 2H), 2.96-3.14 (m, 1H), 1.89-2.08 (m, 2H); LC/MS [M+H]=655.3. Example 176 Synthesis of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)-2-(thiazol-4-yl)propanoic acid Step 1: A solution of crude (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-2-(((1-ethoxy-3-(4-nitrophenyl)-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.17 g) in MeCN (10 mL) was added propan-2-amine (1.0 mL, 11.64 mmol) and DIEA (0.9 mL). The mixture was stirred at 15° C. for 16 h before it was diluted with water (30 mL) and the resulting mixture was extracted EtOAc (4×20 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give crude (2R,3R,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-2-(((1-ethoxy-3-(4-nitrophenyl)-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.16 g) as a yellow solid. Step 2: To a solution of crude (2R,3R,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-2-(((1-ethoxy-3-(4-nitrophenyl)-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.16 g) in EtOH (5 mL) was added Fe powder (856.75 mg, 15.34 mmol) and saturated aq. NH4Cl (1.53 mmol, 5 mL). The mixture was stirred at 50° C. for 2 h before it was filtered through a pad of Celite and the filtrate was concentrated to give crude (2R,3R,4R,5R)-2-(((3-(4-aminophenyl)-1-ethoxy-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.05 g) as a yellow solid. Step 3: To a solution of crude (2R,3R,4R,5R)-2-(((3-(4-aminophenyl)-1-ethoxy-1-oxo-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.05 g) in DCM (10 mL) was added 1-chloro-3-isocyanato-propane (172.85 mg, 1.45 mmol). The mixture was stirred at 15° C. for 16 h before it was quenched with water (20 mL) and the resulting mixture was extracted with DCM (3×10 mL). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give crude ethyl 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(3-(3-chloropropyl)-ureido)phenyl)-2-(thiazol-4-yl)propanoate (1.33 g) as a yellow solid. Step 4: To a solution of crude ethyl 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(3-(3-chloropropyl)-ureido)phenyl)-2-(thiazol-4-yl)propanoate (1.23 g) in THF (12 mL) was added NaH (290.84 mg, 7.27 mmol, 60% in mineral oil). The mixture was stirred at 15° C. for 5 h before it was quenched with H2O (6 mL) and followed by addition of NaOH (290.85 mg, 7.27 mmol). The mixture was stirred at 15° C. for 16 h and then at 40° C. for 8 h. Additional NaOH (600 mg) was added to mixture and the mixture was stirred at 40° C. for 4 h before it was quenched with water (20 mL). The resulting solution was extracted with EtOAc (15 mL). The aq. layer was acidified with 2N aq. HCl (15 mL) to produce a precipitate. The solid was collected by filtration and purified by preparative HPLC (column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)-ACN]; B %: 30%-50%, 10 min) to provide a diastereomeric mixture (ca. 1:1) of 2-(((2R,3S,4R,5R)-5-(2-chloro-6-(isopropyl-amino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-(4-(2-oxotetrahydropyrimidin-1 (2H)-yl)phenyl)-2-(thiazol-4-yl)propanoic acid (245 mg) as a white solid 1H NMR (400 MHz, CD3OD) δ ppm 8.99 (d, J=1.6 Hz, 1H), 7.88-8.18 (m, 1H), 7.53-7.79 (m, 1H), 7.30 (d, J=8.3 Hz, 1H), 7.03-7.13 (m, 2H), 6.95 (d, J=8.4 Hz, 1H), 5.83-6.03 (m, 1H), 4.63-4.74 (m, 1H), 4.33-4.43 (m, 1H), 4.09-4.29 (m, 1H), 3.72-4.01 (m, 3H), 3.59-3.70 (m, 1H), 3.41-3.53 (m, 2H), 3.33-3.38 (m, 2H), 2.91-3.14 (m, 1H), 1.83-2.05 (m, 2H), 1.25-1.35 (m, 6H); LC/MS [M+H]=697.4. Example 177 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-chloro-4-(isopropylamino)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid Step 1: To a solution of 4,6-dichloro-1H-pyrazolo[3,4-d]pyrimidine (620 mg, 1.11 mmol, 1 eq) and (3R,4R,5R)-5-(((1-ethoxy-1-oxo-3-phenyl-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-4-ethynyltetrahydrofuran-2,3,4-triyl triacetate (230.35 mg, 1.22 mmol, 1.1 eq) in MeCN (6.5 mL) under a N2atmosphere at 0° C. was added DBU (501 uL, 3.32 mmol, 3 eq). The mixture was stirred at 0° C. for 5 min and followed by addition of TMSOTf (900.93 uL, 4.99 mmol, 4.5 eq) dropwise. The mixture was stirred at 0° C. for 30 min and then stirred at 65° C. for 16 h before it was quenched with saturated aq. NaHCO3(10 mL) and extracted with EtOAc (3×6 mL). The combined organic phase was dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel (10-40% of EtOAc in petroleum ether) to provide (2R,3R,4R,5R)-5-(4,6-dichloro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (220 mg) as a foam. Step 2: To (2R,3R,4R,5R)-5-(4,6-dichloro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (260 mg, 377.61 umol, 1 eq) in EtOH (2 mL) was added propan-2-amine (64.89 uL, 755.23 umol, 2 eq) and DIEA (131.55 uL, 755.23 umol, 2 eq). The mixture was stirred at 15° C. for 4 h before it was diluted with EtOAc (30 mL), washed with water (8 mL), brine (8 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude residue was purified by preparative TLC (EtOAc:petroleum ether=2:1) to give (2R,3R,4R,5R)-5-(6-chloro-4-(isopropylamino)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (70 mg) as a foam. Step 3: To a solution of (2R,3R,4R,5R)-5-(6-chloro-4-(isopropylamino)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (70 mg, 98.43 umol, 1 eq) in THF (1 mL) was added LiOH·H2O (4.13 mg). The mixture was stirred at 50° C. for 14 h before it was concentrated to dryness. The crude residue was purified by preparative HPLC (column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)−ACN]; B %: 43%-63%, 10 min) and dried by lyophilization to provide a diastereomeric mixture (ca. 1:1) of 2-(((2R,3S,4R,5R)-5-(6-chloro-4-(isopropylamino)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-3-ethynyl-3,4-dihydroxytetrahydro-furan-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid (15.7 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.76-9.10 (m, 1H), 7.93-8.08 (m, 1H), 7.40-7.65 (m, 1H), 6.89-7.08 (m, 4H), 6.80-6.88 (m, 1H), 6.11-6.20 (m, 1H), 5.14-5.28 (m, 1H), 4.38-4.50 (m, 1H), 4.29-4.37 (m, 1H), 3.93-4.11 (m, 1H), 3.78-3.86 (m, 1H), 3.47-3.63 (m, 2H), 2.97-3.09 (m, 1H), 1.25-1.31 (m, 6H); LC/MS [M+H]=598.7. Example 178 Synthesis of 2-(((2R,3S,4R,5R)-5-(4-amino-6-chloro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid Step 1: To a solution of (2R,3R,4R,5R)-5-(4,6-dichloro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydro-furan-3,4-diyl diacetate (100 mg, 145.24 umol, 1 eq) in THF (1 mL) was added NH4OH (199.76 uL, 1.45 mmol, 10 eq). The mixture was stirred at 15° C. for 14 h before it was concentrated to dryness to provide crude (2R,3R,4R,5R)-5-(4-amino-6-chloro-1H-pyrazolo-[3,4-d]pyrimidin-1-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(thiazol-4-yl)propan-2-yl)oxy)-methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (120 mg) as a white solid. Step 2: To a solution of crude (2R,3R,4R,5R)-5-(4-amino-6-chloro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(thiazol-4-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (145.24 umol, 1 eq) in THF (4 mL) and H2O (2 mL) was added LiOH·H2O (60.94 mg, 1.45 mmol, 10 eq). The mixture was stirred at 50° C. for 16 h before it was concentrated to dryness. The crude residue was purified by preparative HPLC (column: YMC-Actus Triart C18 150*30 mm*5 um; mobile phase: [water (0.225% FA)-ACN]; B %: 28%-48%, 10 min) and dried by lyophilization to provide a diastereomeric mixture (ca. 1:1) of 2-(((2R,3S,4R,5R)-5-(4-amino-6-chloro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)-propanoic acid (32.8 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ ppm 8.87-9.01 (m, 1H), 7.91-8.06 (m, 1H), 7.47-7.67 (m, 1H), 7.01-7.06 (m, 2H), 6.93-7.00 (m, 2H), 6.84-6.90 (m, 1H), 6.12-6.20 (m, 1H), 5.14-5.27 (m, 1H), 4.26-4.33 (m, 1H), 3.76-4.03 (m, 2H), 3.48-3.70 (m, 2H), 2.96-3.05 (m, 1H); LC/MS [M+H]=557.0. Example 179 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)acetic acid Step 1: To a solution of (2R,3R,4R,5R)-5-((6-N,N′-bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (2 g, 3.28 mmol) in toluene (10 mL) was added ethyl 2-diazo-2-(thiazol-4-yl)acetate (841 mg, 4.26 mmol) and Rh2(OAc)4(145 mg, 0.328 mmol) under an argon atmosphere. The resulting mixture was stirred at 70° C. for 2 h before it was allowed to cool to room temperature. The organic volatile was removed under reduced pressure. The resulting crude was purified by silica gel column chromatography (0-40% EtOAc in hexanes) to provide (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-((2-ethoxy-2-oxo-1-(thiazol-4-yl)ethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.78 g) as a syrup. Step 2: To a solution of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-((2-ethoxy-2-oxo-1-(thiazol-4-yl)ethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (310 mg) in DCM (3 mL) at 25° C. was added TFA (2 mL). The mixture was stirred for 2 h before it was concentrated under reduced pressure to provide crude (2R,3R,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-2-((2-ethoxy-2-oxo-1-(thiazol-4-yl)ethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate. To a solution of crude (2R,3R,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-2-((2-ethoxy-2-oxo-1-(thiazol-4-yl)ethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate in THF (1 mL) and H2O (1 mL) at 0° C. was added LiOH monohydrate (100 mg). The resulting mixture was stirred at 25° C. overnight before the organic volatile was removed under reduced pressure. The mixture was cooled to 0° C. before it was acidified to pH˜6 with 1N aq. HCl solution and concentrated under reduced pressure. The crude residue was purified by preparative reversed-phase HPLC to provide a diastereomeric mixture (ca. 1:1) of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydro-furan-2-yl)methoxy)-2-(thiazol-4-yl)acetic acid as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.84-9.00 (m, 2H), 7.67-7.68 (m, 1H), 6.02-6.06 (m, 1H), 5.28-5.32 (d, J=12.27 Hz, 1.5H), 5.14-5.16 (d, J=7.56 Hz, 0.5H), 4.24-4.28 (m, 1H), 3.69-4.09 (m, 2H), 3.16 (s, 0.5H), 2.95 (s, 0.5H); LC/MS [M+H]=467.0. Examples 180 and 181 Synthesis of (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid and (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid Proceeding as described in Example 169 above but substituting 3-(bromomethyl)thiophene and 2-chloro-N-isopropyl-9purin-6-amine with benzyl bromide and 2-chloroadenine provided a pair of diastereomeric products which the stereo configuration was assigned arbitrarily. Both products were purified by preparative HPLC and isolated as white solids. (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid:1H NMR (CD3OD, 300 MHz) δ 8.95-8.96 (d, J=2.01 Hz, 1H), 8.34 (s, 1H), 7.54-7.55 (d, J=2.01 Hz, 1H), 6.97-7.12 (m, 5H), 5.97-5.99 (d, J=6.99 Hz, 1H), 4.97-4.99 (d, J=7.08 Hz, 1H), 4.27-4.29 (t, J=4.23, 3.18 Hz, 1H), 3.88-3.99 (m, 2H), 3.62-3.79 (q, J=14.82, 39.24 Hz, 2H), 2.97 (s, 1H); LC/MS [M+H]=557.0. (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid:1H NMR (CD3OD, 300 MHz) δ 8.95-8.96 (m, 1H), 7.99 (s, 1H), 7.70-7.71 (d, J=1.98 Hz, 1H), 7.05-7.25 (m, 5H), 5.92-5.94 (d, J=7.02 Hz, 1H), 4.85-4.87 (d, J=7.29 Hz, 1H), 4.20-4.22 (q, J=2.64 Hz, 1H), 3.58-3.90 (m, 4H), 3.02 (s, 1H); LC/MS [M+H]=557.0. Example 182 Synthesis of 3-(2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-carboxy-2-(thiazol-4-yl)ethyl)benzoic acid Proceeding as described in Example 179 above but substituting BnBr with methyl 3-(bromomethyl)benzoate provided the title compound as a mixture of diastereomers (ca. 1:1) and isolated as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.99-9.01 (m, 1H), 8.33 (s, 0.5H), 8.14 (s, 0.5H), 7.66-7.88 (m, 3H), 7.45-7.48 (d, J=7.29 Hz, 0.5H), 7.36-7.39 (d, J=7.86 Hz, 0.5H), 7.17-7.22 (t, J=7.56 Hz, 0.5H), 7.00-7.05 (d, J=7.47 Hz, 0.5H), 5.99-6.01 (d, J=7.29 Hz, 0.5H), 5.93-5.95 (d, J=6.87 Hz, 0.5H), 5.00-5.03 (d, J=7.38 Hz, 0.5H), 4.90-4.95 (d, J=6.80 Hz, 0.5H), 4.23-4.31 (m, 1H), 3.80-4.01 (m, 2H), 3.63-3.69 (m, 2H), 3.01 (s, 0.5H), 2.92 (s, 0.5H); LC/MS [M+H]=601.0. Example 183 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)-3-(3-(trifluoromethoxy)phenyl)propanoic acid Proceeding as described in Example 179 above but substituting BnBr with 1-(bromomethyl)-3-(trifluoromethoxy)benzene provided the title compound as a mixture of diastereomers (ca. 1:1) and isolated as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.97-9.00 (m, 1H), 8.41 (s, 0.5H), 8.26 (s, 0.5H), 7.68-7.69 (d, J=1.92 Hz, 0.5H), 7.62-7.63 (d, J=1.83 Hz, 0.5H), 6.88-7.21 (m, 4H), 6.00-6.02 (d, J=7.14 Hz, 0.5H), 5.94-5.96 (d, J=6.78 Hz, 0.5H), 5.04-5.07 (d, J=7.44 Hz, 0.5H), 4.91-4.94 (d, J=6.87 Hz, 0.5H), 4.28-4.33 (m, 1H), 3.62-3.96 (m, 4H), 2.98 (s, 0.5H), 2.96 (s, 0.5H); LC/MS [M+H]=641.0. Example 184 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)pent-4-ynoic acid Proceeding as described in Example 179 above but substituting BnBr with propargyl bromide provided the title compound as a mixture of diastereomers (ca. 1:1) and isolated as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.95 (s, 1H), 8.79-8.83 (d, J=13.95 Hz, 1H), 7.73 (s, 1H), 6.04-6.06 (d, J=7.05 Hz, 1H), 4.97-5.05 (dd, J=7.29, 17.91 Hz, 1H), 4.24-4.30 (m, 1H), 3.69-3.94 (m, 2H), 3.34-3.38 (m, 2H), 3.04 (s, 0.5H), 2.93 (s, 0.5H), 2.22-2.30 (dt, J=1.74, 19.62 Hz, 1H); LC/MS [M+H]=505.0. Examples 185 and 186 Synthesis of (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-N-hydroxy-2-(thiazol-4-yl)acetamide and (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-N-hydroxy-2-(thiazol-4-yl)acetamide To a solution of (2R,3R,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-2-((2-ethoxy-2-oxo-1-(thiazol-4-yl)ethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (2 g, 3.45 mmole) in MeOH (20 mL) was added 50% NH2OH in H2O (30 mL). The reaction mixture was stirred at 40° C. for 1 h before it was concentrated under reduced pressure. The crude residue was purified by preparative reversed-phase HPLC to provide a pair of diastereomeric title products which the stereo configuration was assigned arbitrarily. Both products were isolated as white solids. (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-N-hydroxy-2-(thiazol-4-yl)acetamide:1H NMR (CD3OD, 300 MHz) δ 9.02-9.03 (d, J=1.77 Hz, 1H), 8.45 (s, 1H), 7.68-7.69 (d, J=1.8 Hz, 1H), 5.98-6.01 (d, J=6.96 Hz, 1H), 4.92-4.95 (d, J=6.93 Hz, 1H), 4.29-4.32 (q, J=2.82, 2.28 Hz, 1H), 3.91-4.10 (m, 2H), 3.18 (s, 1H); LC/MS [M+H]=482.0. (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-N-hydroxy-2-(thiazol-4-yl)acetamide:1H NMR (CD3OD, 300 MHz) δ 8.97 (s, 1H), 8.46 (s, 1H), 7.68 (s, 1H), 5.97-6.00 (d, J=6.96 Hz, 1H), 4.88-4.90 (d, J=6.93 Hz, 1H), 4.33-4.35 (m, 1H), 3.96-4.08 (m, 2H), 3.12 (s, 1H); LC/MS [M+H]=482.0. Examples 187 and 188 Synthesis of (S)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid and (R)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid Example 187 Example 188 Proceeding as described in Examples 170 and 171 above but substituting 2-chloro-N-isopropyl-9H-purin-6-amine with 2-chloro-N-methyl-9H-purin-6-amine provided the title compounds as a pair of diastereomers (ca. 1:1) and isolated as white solids. (S)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxy-tetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid:1H NMR (CD3OD, 300 MHz) δ 8.99 (s, 1H), 7.97 (s, 1H), 7.69 (s, 1H), 7.08-7.22 (m, 5H), 5.93-5.95 (d, J=6.9 Hz, 1H), 4.96-4.98 (d, J=6.0 Hz, 1H), 3.59-4.22 (m, 5H), 3.01-3.06 (m, 4H); LC/MS [M+H]=571.0. (R)-2-(((2R,3S,4R,5R)-5-(2-chloro-6-(methylamino)-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(thiazol-4-yl)propanoic acid:1H NMR (CD3OD, 300 MHz) δ 8.97-8.98 (d, J=1.83 Hz, 1H), 8.15 (s, 1H), 7.58-7.59 (d, J=1.77 Hz, 1H), 6.94-7.09 (m, 5H), 5.98-6.00 (d, J=7.17 Hz, 1H), 4.98-5.01 (d, J=7.26 Hz, 1H), 4.27-4.29 (t, J=3.48 Hz, 1H), 3.93 (m, 2H), 3.58-3.80 (q, J=14.46, 38.7 Hz, 2H), 3.07 (s, 3H), 2.94 (s, 1H); LC/MS [M+H]=571.0. Examples 189 and 190 Synthesis of (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-cyano-3-phenylpropanoic acid and (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-cyano-3-phenylpropanoic acid Step 1: To a solution of R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (510 mg, 0.836 mmol) in toluene (5 mL) was added ethyl 2-cyano-2-diazoacetate (134 mg, 0.961 mmol). The mixture was concentrated in vacuo. The mixture was taken up in dry toluene (2 mL) and followed by addition of Rh2(OAc)4(8 mg, 17 umol) under argon atmosphere. The material was stirred and heated at 80° C. for 30 minutes before additional ethyl 2-cyano-2-diazoacetate (254 mg, 1.82 mmol) was added over 60 min at 80° C. The reaction was further heated at 80° C. for 80 minutes before it was cooled to room temperature and concentrated. The crude product was purified by CombiFlash silica gel chromatography (5-65% of EtOAc in hexanes) to provide a diastereomeric mixture of (2R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-((1-cyano-2-ethoxy-2-oxoethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (230 mg, 38% yield) as an off-white foam. Step 2: An oven dried flask was charged with ((2R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-((1-cyano-2-ethoxy-2-oxoethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (230 mg, 0.319 mmol) and taken up in dry DMF (4 mL). To this mixture was added Cs2CO3(208 mg, 0.639 mmol) and followed by the addition of benzyl bromide (109 mg, 0.639 mmol). The mixture was stirred at 25° C. for 30 minutes before it was diluted with cold saturated aqueous NH4Cl (40 mL) and extracted with EtOAc (40 mL). The aqueous phases were extracted with EtOAc (2×40 mL). The combined organic layer was dried over Na2SO4, filtered and concentrated. The crude product was purified by CombiFlash silica gel column chromatography (10-70% EtOAc in hexanes) to provide (2R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(((2-cyano-1-ethoxy-1-oxo-3-phenylpropan-2-yl)oxy)methyl)-3-ethynyltetrahydro-furan-3,4-diyl diacetate as a pair of diastereomers (215 mg, 83% yield) as an off-white solid. Step 3: A solution (2R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(((2-cyano-1-ethoxy-1-oxo-3-phenylpropan-2-yl)oxy)methyl)-3-ethynyltetra-hydrofuran-3,4-diyl diacetate (215 mg, 0.265 mmol) in a solution of TFA (1 mL) in DCM (1 mL) was stirred for 2 h before it was concentrated under reduced pressure. The residue was azetroped with DCM (8×8 mL) under reduced pressure. The residue was taken up in a mixture of MeOH in H2O (2.2 mL, 5:1=v:v) and followed by addition of LiOH·H2O (77 mg, 1.86 mmol, 7 eq) and THF (0.5 mL). The mixture was stirred at ambient temperature for 40 minutes before it was concentrated to dryness. The residue was dissolved in H2O (15 mL). The aqueous phase was extracted with EtOAc (2×10 mL). The aqueous phase was acidified to pH 2.5 with 1N aq. HCl solution. The aqueous phase was extracted with EtOAc (3×50 mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The crude residue was purified by preparative reversed-phase HPLC to provide the title compounds as a pair of diastereomers: (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-cyano-3-phenylpropanoic acid (33.7 mg, 26% yield) and (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-cyano-3-phenylpropanoic acid (30 mg, 23% yield) which the stereo configuration was assigned arbitrarily. Both were isolated as off-white solids. (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-cyano-3-phenylpropanoic acid:1H NMR (CD3OD, 300 MHz) δ 8.32 (bs, 1H), 7.20-7.36 (m, 5H), 6.02 (d, J=7.00 Hz, 1H), 4.68 (d, J=7.02 Hz, 1H), 4.33-4.37 (m, 1H), 4.18 (dd, J=9.91, 3.96 Hz, 1H), 3.99 (dd, J=9.94, 2.19 Hz, 1H), 3.41 (bs, 2H), 3.03 (s, 1H); LC/MS [M+H]=499.1. (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-cyano-3-phenylpropanoic acid:1H NMR (CD3OD, 300 MHz) δ 8.04 (bs, 1H), 7.21-7.39 (m, 5H), 5.99 (d, J=6.90 Hz, 1H), 4.79 (d, J=6.93 Hz, 1H), 4.31-4.36 (m, 1H), 4.19 (dd, J=10.07, 4.34 Hz, 1H), 4.12 (dd, J=10.10, 3.30 Hz, 1H), 3.38-3.44 (m, 2H), 3.09 (s, 1H); LC/MS [M+H]=499.1. Examples 191 and 192 Synthesis of (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(1H-tetrazol-5-yl)propanoic acid and (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(1H-tetrazol-5-yl)propanoic acid Step 1: To a solution of ethyl 1H-tetrazole-5-acetate (3 g, 19.21 mmol) in DMF (40 mL) under argon atmosphere at 25° C. was added 2-(trimethylsilyl)ethoxymethyl chloride (4.1 mL, 23.05 mmol) and powdered potassium carbonate (5.31 g, 38.42 mmol). The reaction mixture was stirred overnight before it was diluted with brine (70 mL) and EtOAc (70 mL). The aqueous phase was extracted with EtOAc (2×70 mL). The combined organic layer was washed with brine (70 mL) and water (70 mL), dried over Na2SO4and concentrated. The residue was purified by silica gel column chromatography (15-48% EtOAc in hexanes) to provide ethyl 2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)acetate (2.379 g) as a light yellow oil. Step 2: To a solution of ethyl 2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)acetate (2.379 g, 8.31 mmol) in dry acetonitrile (25 mL) under argon atmosphere was added DBU (1.87 mL, 12.47 mmol). To this mixture was added 4-acetamidobenzenesulfonyl azide (2.395 g, 9.96 mmol) in 3 equal portions over 5 minutes. The reaction mixture was stirred for 3.5 h the organic volatile was removed under reduced pressure. The residue was purified by silica gel column chromatography (20% EtOAc in hexanes) to provide ethyl 2-diazo-2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)acetate (2.316 g) as an oil. Step 3: To a solution of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (2 g, 3.28 mmol) in toluene (8 mL) at 20° C. under N2atmosphere was added Rh2(OAc)4(29 mg, 0.066 mmol, 0.066 eq) and ethyl 2-diazo-2-(2-((2-(trimethylsilyl)ethoxy)-methyl)-2H-tetrazol-5-yl)acetate (1.08 g, 3.44 mmol, 1.05 eq). The mixture was stirred at 75° C. for 1 h before additional ethyl 2-diazo-2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)acetate (720 mg) was added over 80 min. The reaction mixture was cooled to ambient temperature and concentrated. The crude material was purified by Combi-Flash silica gel column (5-80% EtOAc in hexanes) to provide a diastereomeric mixture of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-((2-ethoxy-2-oxo-1-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)ethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.608 g) as a gum. Step 4: To a solution of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-((2-ethoxy-2-oxo-1-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)ethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.555 g, 1.739 mmol) in dry toluene (10 mL). The mixture was concentrated under reduced pressure. The residue was taken up in dry DMF (10 mL) and followed by addition of benzyl bromide (1.189 g, 6.96 mmol) and dried Cs2CO3(1.133 g, 3.478 mmol). The mixture was stirred at 25° C. for 5.5 h before it was diluted with saturated aq. NH4Cl solution (60 mL). The aqueous phase was extracted with EtOAc (3×60 mL). The combined organic layer was washed with brine (60 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by flash silica gel column chromatography (5-65% EtOAc in hexanes) to provide a diastereomeric mixture of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.041 g) as a foam. Step 5: To a solution of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(((1-ethoxy-1-oxo-3-phenyl-2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)propan-2-yl)oxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.041 g, 1.057 mmol) in a mixture of MeOH and H2O (12 mL, 6:1=v:v) was added powdered LiOH·H2O (349 mg, 8.5 mmol). The mixture was stirred at 23° C. for 16 h before it was concentrated to dryness. The residue was dissolved in H2O (40 mL) and it was extracted with EtOAc (40 mL). The aqueous phase was acidified to pH 2.5 with 1N aq. HCl solution and extracted with EtOAc (3×40 mL). The combined organic layer was washed with brine (40 mL), dried over Na2SO4, filtered and concentrated to provide a diastereomeric mixture of 2-(((2R,3S,4R,5R)-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)propanoic acid (784 mg) as an oil. Step 6: To a solution of 2-(((2R,3S,4R,5R)-5-(6-((tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)propanoic acid (138 mg, 0.179 mmol) in DCM (0.9 mL) under argon atmosphere at 0° C. was added TFA (0.9 mL). The mixture was stirred at 0° C. for 5 h and then stirred to ambient for 15 min before the organic volatile was removed under the reduced pressure. The residue was azetroped with DCM (3×15 mL) under reduced pressure. The crude residue was purified by preparative reversed-phase HPLC to provide the two title products as a pair of diastereomers: (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(1H-tetrazol-5-yl)propanoic from the first fraction and (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-3-phenyl-2-(1H-tetrazol-5-yl)propanoic acid from the later fraction. Both isolated as off-white solids. (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-phenyl-2-(1H-tetrazol-5-yl)propanoic acid:1H NMR (CD3OD, 300 MHz); δ 8.37 (s, 1H), 7.11-7.23 (m, 5H), 5.96 (d, J=6.57 Hz, 1H), 4.81 (d, J=6.57 Hz, 1H), 4.25-4.30 (m, 1H), 4.01 (dd, J=10.19, 2.29 Hz, 1H), 3.78 (d, J=13.90 Hz, 1H), 3.67 (d, J=13.90 Hz 1H), 3.72-3.79 (m, 1H), 3.06 (s, 1H); LC/MS [M+H]=542.2. (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-3-phenyl-2-(1H-tetrazol-5-yl)propanoic acid:1H NMR (CD3OD, 300 MHz); δ 8.37 (s, 1H), 6.92-7.11 (m, 5H), 6.01 (d, J=7.11 Hz, 1H), 5.06 (d, J=7.11 Hz, 1H), 4.35-4.39 (m, 1H), 4.11 (dd, J=10.06, 2.52 Hz, 1H), 4.01 (dd, J=10.06, 5.49 Hz, 1H), 3.80 (d, J=14.75 Hz 1H), 3.67 (d, J=14.75 Hz 1H), 2.96 (s, 1H); LC/MS [M+H]=542.2. Examples 193 and 194 Synthesis of (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(2-phenylthiazol-4-yl)acetic acid and (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(2-phenylthiazol-4-yl)acetic acid Proceeding as described in Example 179 but substituting ethyl 2-diazo-2-(thiazol-4-yl)acetate with ethyl (2-phenylthiazol-4-yl)diazoacetate which was prepared via the procedure described by Lefebvre, Quentin, et al., (Chemical Communications2014, 50, 6617-6619) provided the title compounds as a pair of diastereomers (ca. 1:1). The stereo configuration was assigned arbitrarily. Both were isolated as off-white solids. (S)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-(2-phenylthiazol-4-yl)acetic acid:1H NMR (CD3OD, 300 MHz); δ 9.03 (bs, 1H), 7.91-7.97 (m, 2H), 7.60 (s, 1H), 7.39-7.45 (m, 3H), 6.06 (d, J=7.45 Hz, 1H), 5.28 (s, 1H), 4.92 (d, J=7.45 Hz, 1H), 4.29-4.33 (m, 1H), 4.12 (dd, J=10.45, 2.51 Hz, 1H), 4.01 (dd, J=10.46, 2.56 Hz, 1H), 2.92 (s, 1H); LC/MS [M+H]=543.1. (R)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)-2-(2-phenylthiazol-4-yl)acetic acid:1H NMR (CD3OD, 300 MHz); δ 9.15 (s, 1H), 7.90-7.96 (m, 2H), 7.61 (s, 1H), 7.38-7.44 (m, 3H), 6.09 (d, J=7.48 Hz, 1H), 5.23 (d, J=7.48 Hz, 1H), 5.30 (s, 1H), 4.26-4.29 (m, 1H), 3.93 (dd, J=10.67, 2.18 Hz, 1H), 3.76 (dd, J=10.64, 2.48 Hz, 1H), 3.17 (s, 1H); LC/MS [M+H]=543.2. Example 195 Synthesis of 4-((((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(carboxy)methyl)thiazole-2-carboxylic acid Step 1: To a microwave vial was charged with ethyl thiooxamate (1.91 g, 14.36 mmol) and ethyl 4-bromoacetoacetate (3 g, 14.36 mmol) in dry toluene (27 mL). The mixture was irradiated in a microwave reactor at 90° C. for 1 hour. The reaction mixture was cooled to ambient and the solvent decanted and then was concentrated. The crude residue was purified by CombiFlash silica gel chromatography (2-56% EtOAc in hexanes) to provide ethyl 4-(2-ethoxy-2-oxoethyl)thiazole-2-carboxylate (890 mg, 26% yield) as a thick oil. Step 2: To a solution of ethyl 4-(2-ethoxy-2-oxoethyl)thiazole-2-carboxylate (890 mg, 3.66 mmol) in dry acetonitrile (12 mL) under argon atmosphere was added DBU (0.82 mL, 5.49 mmol) and 4-acetamidobenzenesulfonyl azide (1.055 g, 4.39 mmol). The reaction mixture was stirred for 1.5 hours before it was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (20% EtOAc in hexanes) to provide ethyl 4-(1-diazo-2-ethoxy-2-oxoethyl)thiazole-2-carboxylate (894 mg, 90% yield) as a yellowish solid. Step 3: Proceeding as described in Example 179 but substituting ethyl 2-diazo-2-(thiazol-4-yl)acetate with ethyl 4-(1-diazo-2-ethoxy-2-oxoethyl)thiazole-2-carboxylate provided the title compound as a mixture of diastereomers (ca. 1:1) and isolated as off-white solids. Isomer 1:1H NMR (CD3OD, 300 MHz): δ 8.93 (s, 1H), 7.91 (s, 1H), 6.06 (d, J=7.42 Hz, 1H), 5.39 (s, 1H), 5.13 (d, J=7.42 Hz, 1H), 4.24-4.31 (m, 1H), 4.03-4.09 (m, 1H), 3.75-3.83 (m, 1H), 2.95 (s, 1H); LC/MS [M+H]=511.1. Isomer 2:1H NMR (CD3OD, 300 MHz): δ 8.87 (s, 1H), 7.93 (s, 1H), 6.04 (d, J=7.42 Hz, 1H), 5.33 (s, 1H), 4.91 (d, J=7.45 Hz, 1H), 4.24-4.31 (m, 1H), 3.92-4.01 (m, 2H), 3.19 (s, 1H); LC/MS [M+H]=511.1. Example 196 Synthesis of 4-(1-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-1-carboxy-2-phenylethyl)thiazole-2-carboxylic acid Proceeding as described in Example 179 above but substituting (2R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxyl-methyl)tetrahydrofuran-3,4-diyl diacetate with (2R,3R,4R,5R)-5-(6-N,N′-(bis-(tert-butoxy-carbonyl)amino))-2-chloro-9H-purin-9-yl)-2-((2-ethoxy-1-(2-(ethoxycarbonyl)thiazol-4-yl)-2-oxoethoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate provided the title compound as a mixture of diastereomers (ca. 1:1) and isolated as an off-white solid. Isomer 1:1H NMR (CD3OD, 300 MHz): δ 8.17 (bs, 1H), 7.81 (s, 1H), 6.92-7.25 (m, 5H), 5.95 (d, J=7.02 Hz, 1H), 4.89 (d, J=7.02 Hz, 1H), 4.29-4.34 (m, 1H), 3.59-4.05 (m, 4H), 3.01 (s, 1H); LC/MS [M+H]=601.1. Isomer 2:1H NMR (CD3OD, 300 MHz): δ 8.05 (bs, 1H), 7.90 (s, 1H), 6.92-7.25 (m, 5H), 6.00 (d, J=7.41 Hz, 1H), 5.04 (d, J=7.42 Hz, 1H), 4.21-4.26 (m, 1H), 3.59-4.05 (m, 4H), 3.06 (s, 1H); LC/MS [M+H]=601.1. Examples 197 Synthesis of 2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)-2-(4-(2-aminopyridin-3-yl)benzyl)malonic acid Proceeding as described in Example 22 above but substituting (2-oxo-1,2-dihydropyridin-3-yl)boronic acid with 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-amine provided the title compound as a white solid. 1H NMR (CD3OD, 300 MHz) δ 8.43 (s, 1H), 7.85-7.87 (dd, J=1.5, 6.42 Hz, 1H), 7.65-7.68 (dd, J=1.53, 7.38 Hz, 1H), 7.37-7.40 (d, J=8.13 Hz, 2H), 7.09-7.12 (d, J=8.07 Hz, 2H), 6.88-6.93 (t, J=6.9 Hz, 1H), 5.99-6.01 (d, J=6.72 Hz, 1H), 4.77-4.79 (d, J=7.0, 1H), 4.37-4.40 (m, 1H), 3.98-4.12 (m, 2H), 3.34-3.42 (m, 2H), 3.09 (s, 1H); LC/MS [M+H]=610.1. Examples 198 & 199 Synthesis of (S)-3-([1,1′-biphenyl]-4-yl)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-methyltetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)propanoic acid and (R)-3-([1,1′-biphenyl]-4-yl)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-methyltetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)propanoic acid Proceeding as described in Example 1 above but substituting (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(((tert-butyldiphenylsilyl)oxy)-methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate and diethyl 2-diazomalonate with (2R,3R,4R,5R)-5-(6-(N,N′-bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-(hydroxylmethyl)-3-methyltetrahydrofuran-3,4-diyl diacetate and methyl 2-diazo-2-(thiazol-4-yl)acetate provided a pair of diastereomeric title products (ca. 1:1) which the stereo configuration was assigned arbitrarily. Both products were purified by preparative HPLC and isolated as white solids. (S)-3-([1,1′-biphenyl]-4-yl)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-methyltetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)propanoic acid:1H NMR (CD3OD, 300 MHz) δ 9.04 (s, 1H), 8.42 (s, 1H), 7.74 (s, 1H), 7.21-7.45 (m, 9H), 6.00-6.03 (d, J=8 Hz, 1H), 4.67-4.70 (d, J=7 Hz, 1H), 4.13 (s, 1H), 3.88-3.85 (m, 2H), 3.62-3.66 (d, J=14 Hz, 1H), 3.44-3.47 (d, J=11 Hz, 1H), 1.37 (s, 3H); LC/MS [M+H]=623.2. (R)-3-([1,1′-biphenyl]-4-yl)-2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-methyltetrahydrofuran-2-yl)methoxy)-2-(thiazol-4-yl)propanoic acid:1H NMR (CD3OD, 300 MHz) δ 9.04 (s, 1H), 8.21 (s, 1H), 7.27-7.69 (m, 10H), 5.93-5.95 (d, J=7 Hz, 1H), 4.52-4.55 (d, J=8 Hz, 1H), 4.04 (s, 1H), 3.79-3.85 (m, 3H), 1.36 (s, 3H); LC/MS [M+H]=623.2. Example 200 Synthesis of 4′-(2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-methyltetrahydrofuran-2-yl)methoxy)-2-carboxy-2-(thiazol-4-yl)ethyl)-[1,1′-biphenyl]-2-carboxylic acid Proceeding as described in Example 179 above but substituting (2R,3R,4R,5R)-5-((6-N,N′-bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxyl-methyl)tetrahydrofuran-3,4-diyl diacetate and BnBr with (2R,3R,4R,5R)-5-(6-(N,N′-bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-2-((2-methoxy-2-oxo-1-(thiazol-4-yl)ethoxy)methyl)-3-methyltetrahydrofuran-3,4-diyl diacetate and 4-(bromomethyl)-1,1′-biphenyl provided the title compound as a mixture of diastereomers (ca. 1:1) and isolated as an off-white solid. LC/MS [M+H]=667.2. Example 201 Synthesis of 4′-(2-(((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-methyltetrahydrofuran-2-yl)methoxy)-2-carboxy-2-(1H-tetrazol-5-yl)ethyl)-[1,1′-biphenyl]-2-carboxylic acid Step 1: To a solution of ethyl 2-(1H-tetrazol-5-yl)acetate (500 mg, 3.24 mmol) and trimethylsilyl)ethoxymethyl chloride (0.69 mL, 3.89 mmol) in dry DMF (7 mL) under argon atmosphere at 25° C. was added powdered potassium carbonate (896 mg, 6.48 mmol). The reaction mixture was stirred overnight before it was diluted with H2O (30 mL) and extracted with EtOAc (3×30 mL). The combined organic layer was washed with brine (30 mL) and water (30 mL) and then dried over Na2SO4and concentrated. The residue was purified by CombiFlash silica gel column chromatography (8-58% EtOAc in hexanes) to provide ethyl 2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)acetate (200 mg) as an oil. Steps 2-6: Proceeding as described in Example 1 above but substituting methyl 2-(thiazol-4-yl)acetate with ethyl 2-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)acetate provided a pair of diastereomeric title products (ca. 1:1) which the stereo configuration was assigned arbitrarily. Both products were purified by preparative HPLC and isolated as off-white solids. 1H NMR (CD3OD, 300 MHz): Isomer 1: δ 8.50 (s, 1H), 7.73-7.79 (m, 2H), 7.31-7.56 (m, 3H), 7.01-7.24 (m, 4H), 6.04 (d, J=7.87 Hz, 1H), 4.64 (d, J=7.88 Hz, 1H), 4.10-4.14 (m, 1H), 3.46-4.00 (m, 4H), 1.35 (s, 3H); Isomer 2: δ 8.31 (s, 1H), 7.73-7.79 (m, 2H), 7.31-7.56 (m, 3H), 7.01-7.24 (m, 4H), 5.99 (d, J=7.75 Hz, 1H), 4.48 (d, J=7.72 Hz, 1H), 4.19-4.23 (m, 1H), 3.46-4.00 (m, 4H), 1.42 (s, 3H); LC/MS [M+H]=652.2. Example 202 Synthesis of (((((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid Step 1: To a solution of (2R,3R,4R,5R)-5-(6-(bis-(tert-butoxycarbonyl)amino)-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (329 mg, 0.539 mmol) in i-PrOH (1.6 mL), MeOH (1.1 mL) and H2O (0.8 mL) was added powdered LiOH (111 mg, 2.69 mmol). The mixture was stirred for 30 minutes before the organic volatile was removed under reduced pressure and the residue was diluted with H2O (12 mL). The pH of the aq. layer was adjusted to ˜ 3 with 1N aq. HCl and extracted with EtOAc (3×12 mL). The combined organic layer was dried over MgSO4, filtered and concentrated to provide tert-butyl (2-chloro-9-((2R,3R,4S,5R)-4-ethynyl-3,4-dihydroxy-5-(hydroxymethyl)tetrahydro-furan-2-yl)-9H-purin-6-yl)carbamate which was used in the next step directly without further purification. Step 2: Tert-butyl (2-chloro-9-((2R,3R,4S,5R)-4-ethynyl-3,4-dihydroxy-5-(hydroxymethyl)-tetrahydrofuran-2-yl)-9H-purin-6-yl)carbamate (0.539 mmol) was taken up in a mixture of DCM (1 mL) and TFA (0.5 mL). The reaction mixture was stirred for 3 h before it was concentrated. The residue was taken up in DCM (10 mL) and concentrated again (repeated 5 cycles). The residue was dried further in the vacuum oven for 18 h to provide crude (2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydro-furan-3,4-diol as an off-white solid. Step 3: To an oven dried flask was charged with crude (2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diol and dry trimethyl phosphate (2.5 mL) under argon atmosphere. The mixture was cooled at 0° C. and followed by dropwise addition of a solution of methylenebis(phosphonic dichloride) (673 mg, 2.7 mmol) in dry trimethyl phosphate (1.1 mL) over 10 minutes. The reaction mixture was stirred at 0° C. for 3 h before a solution of triethylammonium carbonate (1 M, 1.9 mL) was added dropwise. The mixture was stirred for 15 minutes at 0° C. and then stirred for 2 h at ambient temperature. The crude mixture was purified by preparative reversed-phase HPLC to provide a impure product. This impure product was further purified by reserved-phase HPLC twice to provide the desired (((((2R,3S,4R,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(hydroxy)phosphoryl)methyl)-phosphonic acid (34 mg) as a light brown solid. 1H NMR (CD3OD, 300 MHz): δ 8.75 (bs, 1H), 6.07 (br, 1H), 4.86 (bs, 1H), 4.31-4.61 (m, 3H), 3.20 (s, 1H), 2.54 (br, 1H); LC/MS [M+H]=484.0. Example 203 Synthesis of (((((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid Step 1: While under nitrogen, a solution of (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)-oxy)methyl)-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-ol (14.48 g, 32 mmoL) (prepared using the methods described in Hulpia, F. et al. Bioorg. Med. Chem. Lett. 2016, 26, 1970-1972) in acetic acid (130 mL) was cooled to 14-17° C. and treated with acetic anhydride (32.01 mL, 341 mmoL, 10.7 eq) and concentrated sulfuric acid (576 uL, 10.8 mmoL, 0.34 eq). After stirring at for 2.5 h. The mixture was diluted with ethyl acetate (200 mL each) and washed with water. The aqueous phase was extracted with ethyl acetate (25 mL), and the combined organic solution washed with sodium bicarbonate (aqueous, saturated, 200 mL), dried over Na2SO4, filtered, and concentrated. The residual oil was purified by flash column chromatography on silica gel column (0-3% ethyl acetate in dichloromethane) to provide (3R,4R,5R)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-ethynyltetrahydrofuran-2,3,4-triyl triacetate as a mixture of anomers and isolated as a white solid in good yield (9.5 g, 55%). Step 2: While under nitrogen, 2,6-dichloroadenine (2.91 g, 15.4 mmoL, 1.01 eq) and N,O-bis(trimethylsilyl)acetamide (4.87 mL, 19.6 mmoL, 1.29 eq) in anhydrous acetonitrile (90 mL) was stirred at room temperature. Next, a solution of (2R,3R,4R,5R)-2,4-bis(acetyloxy)-5-{[(tert-butyldiphenylsilyl)oxy]methyl}-4-ethynyloxolan-3-yl acetate (8.2 g, 15.22 mmoL) in anhydrous acetonitrile (10 mL) was added, followed by dropwise addition of trimethylsilyl trifluoromethanesulfonate (3.67 mL, 20.3 mmoL, 1.33 eq). The reaction was warmed to 50° C. for 18 h, then cooled to room temperature. Saturated aqueous sodium bicarbonate (10 mL), was added and the mixture was stirred for ten minutes. The resulting mixture was extracted with ethyl acetate (3×100 mL) and the combined organic layer was dried (Na2SO4), filtered, and concentrated. The residue was purified by flash column chromatography on silica gel column (0-30% ethyl acetate in hexanes) to provide (2R,3R,4R,5R)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate as a white solid (8.2 g, 81%). Step 3: A solution of (2R,3R,4R,5R)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (1.6 g, 2.4 mmoL) in anhydrous THF (25 mL) was cooled to 0° C. and treated with acetic acid (0.192 mL, 3.36 mmoL) and tetrabutylammonium fluoride in THF (1N, 3.36 mL, 3.36 mmoL). After the addition was complete, the reaction was warmed to room temperature with continued stirring for 3 h. The reaction mixture was concentrated. The crude residue was purified via flash column chromatography on silica gel (0-50% ethyl acetate in hexanes) to afford (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxymethyl)tetrahydrofuran-3,4-diyl diacetate (0.88 g, 86%) as a white foam. Step 4: A solution of (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxyl-methyl)tetrahydrofuran-3,4-diyl diacetate (100 mg, 0.233 mmoL) in trimethylphosphate (4 mL) was cooled to 0° C. and treated with a second solution of methylenebis(phosphonic dichloride) (116 mg, 0.467 mmoL, 2 eq) in trimethylphosphate (4 mL). After the addition was complete, stirring was continued for 2 h then the cooling bath was removed and stirring was continued for 18 h. Ammonium bicarbonate (0.7 M aqueous TEAB, pH 8.5) was added slowly with vigorous stirring until no more gas evolution was observed. Once quenched, NaHCO3(satd., aqueous; 5 mL) was added and mixture stirred for 1 h at room temperature. The reaction mixture was washes with dichloromethane, acidified with 2N HCl to pH-1 and extracted with ethyl acetate (10×50 mL). The combined organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The residual oil was azeotroped with toluene (3×10 mL) to give (((((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)(hydroxy)phosphoryl)-methyl)phosphonic acid as an off-white solid that was used in the next step without further purification. Step 5: A solution of (((((2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid from Step 4 (˜90 mg) was dissolved in anhydrous dioxane (8 mL), cooled to 0° C., then treated with diisopropylethylamine (0.085 mL, 0.513 mmoL, 2.2 eq) and benzylamine (0.036 mL, 0.33 mmoL, 1.4 eq). After the addition was complete, the reaction was stirred at room temperature for 18 h and concentrated to provide (((((2R,3R,4R,5R)-3,4-diacetoxy-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyltetrahydrofuran-2-yl)methoxy)(hydroxy)-phosphoryl)methyl)phosphonic acid. The crude product was used directly in the subsequent hydrolysis without further purification. The crude product from Step 5 was dissolved in 1:1 MeOH/THF (2 mL) and treated with LiOH (84 mg, 3.5 mmoL, 15 eq) in water (1 mL). After the addition was complete, the reaction was stirred at room temperature for 18 h before it was acidified to pH-1 with 2N HCl and concentrated. The resulting reaction mixture was diluted in 1:1 acetonitrile in water with 0.1% TFA (4 mL) and purified via reverse phase HPLC to give (((((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid as a white solid (9.2 mg, 7%) after lyophilization. 1H NMR (D2O) δ 8.63 (s, 1H), 7.41 (m, 5H), 6.07 (d, J=6.9 Hz, 1H), 4.96 (d, J=7.0 Hz, 1H), 4.46 (s, 1H), 4.37 (d, J=12.0 Hz, 1H), 4.25 (d, J=11.5 Hz, 1H), 3.21 (s, 1H), 2.38 (t, J=20.0 Hz, 2H). HPLC: Rt=17.2 min, 97.9%. ESI-MS for C21H24ClN5O9P2calcd. 587.07, found 586.8 (M−); ESI-MS for C12H9ClN5calcd. 258.05, found 258.4 (M-ribose fragment). Example 204 Synthesis of ((((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)methyl)phosphonic acid Step 1: A flamed dried round bottom flask was charged with diethyl hydroxymethylphos-phonate (780 mg, 4.64 mmol) and triethylamine (0.838 mL, 6.031 mmoL, 1.3 eq) in anhydrous dichloromethane (20 mL) was cooled to −78° C. and treated trifluoromethane-sulfonic anhydride (0.847 mL, 5.10 mmoL, 1.3 eq) dropwise. The reaction was stirred for 10 min as the reaction was warmed to 0° C. After 30 min, the reaction mixture was poured into ether (precooled to 0° C.) and the crystalline precipitate filtered. The filtrate was then washed sequentially with water (1×100 mL), 1 M HCl (1×100 mL), and saturated aqueous sodium chloride (1×125 mL). Organic layer was dried (MgSO4), filtered, and concentrated to provide crude (diethoxyphosphoryl)methyl trifluoromethanesulfonate obtained as a yellow oil, was dissolved in anhydrous THF and this solution used directly in the next step without further purification. Step 2: A solution of (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-3-ethynyl-2-(hydroxyl-methyl)tetrahydrofuran-3,4-diyl diacetate (350 mg, 0.815 mmoL) and (diethoxyphosphoryl)-methyl trifluoromethanesulfonate (294 mg, 0.978 mmoL, 1.2 eq) in THF (20 mL) was cooled to −78° C. and treated with LiHMDS (1M in THF; 0.980 mL, 0.978 mmoL, 1.2 eq) in a dropwise. After stirring for 1.5 h, reaction was quenched with solid NH4Cl, diluted with water and extracted with ethyl acetate. The organic layer was dried (MgSO4), filtered, and concentrated. Purification by flash column chromatography on silica gel (0-100% ethyl acetate in hexanes) afforded (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-2-(((diethoxy-phosphoryl)methoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (140 mg, 30%) as a pale yellow oil. Step 3: A solution of (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-2-(((diethoxyphos-phoryl)methoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (85 mg, 0.147 mmoL) and diisopropylethylamine (40 μL, 0.235 mmoL, 1.6 eq) in anhydrous dioxane (8 mL) was cooled to 0° C. was treated with benzylamine (19 μL, 0.176 mmoL, 1.2 eq). After the addition was complete, the reaction was warmed to room temperature and stirred for 18 h. The mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried (Na2SO4), filtered, and concentrated to provide crude (2R,3R,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-2-(((diethoxyphosphoryl)methoxy)methyl)-3-ethynyltetrahydrofuran-3,4-diyl diacetate (93 mg, 96%) as a white solid which was used directly in the subsequent step without further purification. Step 4: The crude product from the previous step was dissolved in anhydrous acetonitrile (10 mL) and treated with bromotrimethylsilane (0.24 mL, 1.8 mmoL, 12 eq) dropwise. After the addition was complete, the solution was stirred at room temperature for 22 h and quenched with water (5 mL). After stirring an additional 2-3 min, the solution was extracted with ethyl acetate (4×100 mL). The organic layer was, dried (Na2SO4), filtered, and concentrated to afford ((((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetra-hydrofuran-2-yl)methoxy)methyl)phosphonic acid as an off-white solid. This crude solid was dissolved in absolute EtOH at 0° C. and treated with KOEt (51 mg, 0.61 mmoL, 4 eq) in one portion. The reaction was stirred at room temperature for 20 min before it was acidified with AcOH (0.52 mL, 0.91 mmoL, 6 eq) and stirred an additional 10 min. The crude product was purified via reverse-phase HPLC and dried by lyophilization to give ((((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3-ethynyl-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)methyl)phosphonic acid (6 mg, 7%) as a white solid. 1H NMR (D2O) δ 8.41 (s, 1H), 7.19 (m, 5H), 5.84 (d, J=7.2 Hz, 1H), 4.82 (d, J=7.1 Hz, 1H), 4.21 (m, 1H), 4.19 (m, 2H), 3.79 (d, J=3.8 Hz, 2H), 3.57 (m, 2H), 2.98 (s, 1H). HPLC: Rt=7.19 min, 97.5%. ESI-MS for C20H21ClN5O7P calcd, 509.09, found 509 (M+); ESI-MS for C11H8ClN6calcd. 258.05, found 259 (M-ribose fragment). Example 205 Synthesis of (((((2R,3S,4R,5R)-5-(6-(benzylamino)-2-chloro-9H-purin-9-yl)-3,4-dihydroxy-3-methyltetrahydrofuran-2-yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid Step 1: While under nitrogen an ice-cooled solution of (3aR,5R,6aS)-5-(((tert-butyldiphenyl-silyl)oxy)methyl)-2,2-dimethyldihydrofuro[2,3-d][1,3]dioxol-6 (5H)-one (6 g, 14.1 mmoL) in anhydrous THF (100 mL) was treated with 3M methyl magnesium chloride in THF (5.9 mL, 1.4 eq) dropwise. After the addition was complete, the cooling bath was removed, and stirring was continuing for 1 h. The mixture was cooled back to 0° C. and quenched with a saturated aqueous ammonium chloride (10 mL), diluted with ethyl acetate (100 mL) and washed with water (80 mL). The aqueous was re-extracted with ethyl acetate (1×50 mL) and the combined organic layer was dried over sodium sulfate, filtered, and concentrated. The residual oil was purified by flash column chromatography on silica (0-30% ethyl acetate in hexanes) to provide (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2,2,6-trimethyltetrahydrofuro[2,3-d][1,3]dioxol-6-ol as a pale viscous oil (4.2 g, 67%). Step 2: While under nitrogen, a solution of (3aR,5R,6aS)-5-(((tert-butyldiphenylsilyl)oxy)-methyl)-2,2-dimethyldihydrofuro[2,3-d][1,3]dioxol-6 (5H)-one (2.25 g, 5.08 mmoL) in dichloromethane (35 mL) and water (3.5 mL) cooled to 0° C. and treated with trifluoroacetic acid (15 mL). After 2.5 h, saturated aqueous NaHCO3was added until the solution was pH-8 and mixture was extracted with dichloromethane (2×150 mL). The combined organic layer was dried over sodium sulfate, filtered, and concentrated. The crude oil was azeotroped with toluene (3×5 mL), diluted with dichloromethane (45 mL) was treated with pyridine (12 mL), acetic anhydride (4.77 mL, 50.83 mmoL) and catalytic 4-DMAP (142 mg, 1.17 mmoL). After stirring 18 h, the reaction was diluted with ethyl acetate (200 mL each) and washed sequentially with saturated aqueous NH4Cl (3×100 mL), 0.5 N HCl (2×100 mL), and saturated aqueous sodium chloride (1×120 mL). The combined organic layer was dried over sodium sulfate, filtered, and concentrated. The residual oil was purified by flash column chromatography on silica gel (0-30% ethyl acetate in hexanes) to provide (3R,4R,5R)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-4-methyltetrahydrofuran-2,3,4-triyl triacetate (2.1 g, 78%) as colorless solid. Steps 3-6: Proceeding as described in Example 203 above but substituting (3aR,5R,6R,6aR)-5-(((tert-butyldiphenylsilyl)-oxy)methyl)-6-ethynyl-2,2-dimethyltetrahydrofuro[2,3-d][1,3]-dioxol-6-ol with (3aR,5R,6aS)-5-(((tert-butyldiphenyl-silyl)oxy)methyl)-2,2-dimethyldihydrofuro[2,3-d][1,3]dioxol-6 (5H)-one provided the title compound (8 mg, 3%) as as a white solid. 1H NMR (D2O:DMSO-d6, 6:1) 8.46 (bs, 1H), 7.23 (m, 5H), 5.84 (d, J=6.9 Hz, 1H), 4.45 (d, J=7.0 Hz, 1H), 4.12 (s, 1H), 3.95 (m, 3H), 3.54 (m, 1H), 2.18 (bs, 2H), 1.31 (s, 3H). ESI-MS for C19H24ClN5O9P2calcd. 563.1, found 562.1 (M−). Example 206 Assay 1: Inhibition of the CD73 Enzyme In Vitro For measurements of soluble CD73 enzyme activity, recombinant CD73 was obtained from R&D Systems, Cat. No. 5795-EN-010. Serial dilutions of test compounds were incubated with recombinant CD73 and AMP in reaction buffer (25 mM Tris HCl pH7.5, 5 mM MgCl2, 50 mM NaCl, 0.25 mM DTT, 0.005% Triton X-100). The final reaction volume was 25 μL and the final concentrations of recombinant CD73 and AMP were 0.5 nM and 50 μM, respectively. Reactions were allowed to proceed for 30 minutes at room temperature before the addition of 100 μL Malachite Green (Cell Signaling Technology, Cat. No. 12776). After 5 minutes at room temperature, absorbance at 630 nm was determined on a microplate spectrophotometer. The concentration of inorganic phosphate was determined using a phosphate standard curve. Assay 2: Inhibition of the CD73 Enzyme In Vitro For measurements of soluble CD73 enzyme activity, recombinant CD73 was obtained from R&D Systems, Cat. No. 5795-EN-010. Serial dilutions of test compounds were incubated with recombinant CD73 and AMP in reaction buffer (25 mM Tris HCl pH7.5, 5 mM MgCl2, 50 mM NaCl, 0.25 mM DTT, 0.005% Triton X-100). The final reaction volume was 25 μL and the final concentrations of recombinant CD73 and AMP were 0.05 nM and 50 μM, respectively. Reactions were allowed to proceed for 1 hour at 37° C. before the addition of 100 μL Malachite Green (Cell Signaling Technology, Cat. No. 12776). After 5 minutes at room temperature, absorbance at 630 nm was determined on a microplate spectrophotometer. The concentration of inorganic phosphate was determined using a phosphate standard curve. The IC50data for both assays is given below in Table 2. ND indicates not determined. TABLE 2Assay 1Assay 2CD73CD73ExampleIC50IC50#Compound(nM)(nM)1ND1.72ND1473ND14ND0.55ND4652ND7ND28ND29ND0.1710ND0.2611ND0.0612ND0.113ND0.314ND0.615ND2016ND2417ND34718ND0.319ND0.220ND0.221ND0.322ND0.3235ND241.8ND257ND2658ND2718ND283ND292ND30ND531ND232ND333ND234ND235ND436ND437ND638ND339ND340ND341ND0.642ND143ND244ND6245ND146ND147ND248ND0.549273ND503ND513ND524ND53ND0.354ND0.25535ND56ND>20057ND10358ND1.6591ND603ND611.5ND62ND263ND164ND365ND166ND0.367ND0.868ND0.869ND370ND171ND672ND373ND174ND275ND0.676ND677ND278ND279ND480ND281ND0.482ND283ND184ND385ND286ND187ND0.688ND1.289ND1.1900.9ND910.3ND920.8ND931.7ND94ND595ND796ND397ND298ND299ND6100ND2101ND1102ND1103ND3104ND2105ND2106ND296107ND506108ND3109ND284110ND3111ND2,666112ND1,357113331ND114177ND115135ND1168,049ND117ND58118ND48119ND4,6121204,628ND121ND1,711122ND4012371ND12427ND125ND235126ND9127ND170128ND61291,580ND130ND0.36131ND>100132NDND133ND7134ND16135ND91136ND0.24137ND0.31138ND0.15139ND0.13140ND0.29141ND0.23142ND5143ND13144ND1145ND108146ND0.41147ND0.58148ND0.08149ND0.11150ND0.16151ND0.25152ND0.05153ND0.48154ND2155ND2156ND0.14157ND0.37158ND8159ND476160ND0.31161ND561162ND0.42163ND>100164ND261165ND45166ND145167ND139168ND14169ND162170ND8171ND96172ND6173ND112174ND17175ND1176ND5177NDND178NDND179320ND1809ND18121ND18224ND18356ND184102ND18511748ND1868833ND18710ND188134ND1891851ND1909316ND1917ND19225ND193263ND194336ND195236ND19682ND197ND0.731982268ND1995757ND2006091ND201204ND202148ND2034.2ND204100ND20512ND Example 207 Activation of Tumor-Directed Immune Response with CD73 Inhibitors EG7 cells were implanted subcutaneously into C57BL/6 mice. Compound 9 (50 mg/kg) or vehicle was orally administered BID starting day one post implant (N=10 per group). Tumors were excised on day 14 and analyzed by flow cytometry. Compound 9 increased the % CD8+ cells of CD45+ cells as shown inFIG.1A(* indicates p<0.05). EG7 cells were implanted subcutaneously into C57BL/6 mice. Anti-CD8 antibody was dosed i.p. on days −1, 0, 5, and 10. Compound 9 (50 mg/kg) or vehicle was orally dosed BID starting on day 1.FIG.1Bshows that depletion of CD8+ T cells reverses efficacy (**** indicates p<0.0001 vs Compound 9+anti-CD8). Compound 9 alone showed more of a tumor volume reduction that the combination of Compound 9 than the anti-CD8 antibody. Example 208 Reversal of AMP-Mediated Suppression of CD8+ T Cells Using CD73 Inhibitors Human CD8+ T cells were labeled with CellTrace CFSE and then pre-incubated with an adenosine deaminase inhibitor and Compound 9 or vehicle for 20 minutes. 20 μM AMP was added for assessing T cell proliferation and CD25 expression. 10 μM AMP was added for assessing cytokine production. T cells were activated with α-CD3, α-CD28, and hIL2. After 4 days, proliferation and CD25 expression were assessed by flow cytometry and cytokine levels in the supernatant were measured by ELISA. EC50s were determined using a four-parameter dose-response curve equation.FIG.2Adepicts the EC50=11.6 nM for CD8+ T cell proliferation.FIG.2Bdepicts the EC50=9.6 nM for CD8+ T cell activation.FIG.2Cdepicts the EC50=4.5 nM for IFNy production.FIG.2Ddepicts the EC50=5.6 nM for Granzyme B production. Example 209 Selectivity of CD73 Inhibitors Compounds of the invention are selective for CD73 and do not exhibit proliferative effects. Using Compound 9, the activity of cell surface CD39 was assessed using K562 cells expressing human CD39 and Kinase-Glo. Activity of recombinant human ENTPD2 and ENTPD3 was assessed using a malachite green assay. Each of the enzymes CD39, ENTPD2 AND ENTPD3 all showed an IC50of >10,000 nM. Compound 9 was screened in the Eurofins Safety Screen Panel and the Eurofins Express Diversity Kinase Profile Panel. In the Safety Panel, 1/87 targets were inhibited at >50% at 10 μM of Compound 9. The PDE3 enzyme was inhibited at 59%. In the Kinase Panel, none of the 45 targets were inhibited at >50%. Further, Compound 9 did not show anti-proliferative effects against three cell lines. Viability of EG7 and A375 cells treated with 100 μM Compound 9 was measured using CellTiter-Glo after 3 days. Proliferation of human CD8+ T cells was measured by flow cytometry after 4 days of treatment with 100 μM Compound 9 using CellTrace CFSE Cell Proliferation Kit.FIG.3Ashows the comparable % cell survival of EG7 cells, a mouse T cell lymphoma cell line.FIG.3Bshows the comparable % cell survival of A375 cells, a human melanoma cell line.FIG.3Cshows the comparable % divided cells of human CD8+ T cells. Example 210 CD73 Inhibition The potency of Compound 9 was evaluated against recombinant CD73 and CD73-expressing SK-MEL-28 cells using a malachite green assay. Inhibition of CD73 in plasma was measured using LC/MS to assess conversion of15N5-AMP into15N5-ADO.FIG.4Aindicates the nanomolar inhibition of CD73 cells from both human and mouse sources.FIG.4Bdepicts the IC50=0.17 nM for human recombinant CD73 cells.FIG.4Cdepicts the IC50=0.38 nM for human plasma CD73 cells. Inhibition of CD73 in plasma was measured using LC/MS to assess conversion of15N5-AMP into15N5-ADO.FIG.4Ddepicts the IC50=0.21 nM for human CD73 cell surface. Example 211 CD73 Inhibitor Oral Dosing Phamacodynamics Single dose Compound 9 (50 mg/kg) was administered orally to mice and plasma was collected at indicated time points. Compound 9 levels were measured by LC/MS. The IC50in mouse plasma was 1 nM as shown inFIG.5A. Plasma was harvested from mice 2 hours post dose and spiked with15N5-AMP and a TNAP inhibitor.15N5-ADO levels were measured by LC/MS.FIG.5Bdepicts the 92% inhibition of mouse plasma CD73 cells. Example 212 Single-Agent Efficacy of Orally Dosed CD73 Inhibitors Compounds of the invention show potent anti-tumor effects, evidenced in reducing tumor volume in a mouse model. In one model, EG7 cells were implanted subcutaneously into C57BL/6 mice. Compound 9 or vehicle was orally administered BID starting day one post implant (N=10 per group).FIG.6Adepicts the further decrease in tumor volume with increasing doses of Compound 9. In another model, EG7 cells were implanted subcutaneously into C57BL/6 mice. Compound 9 was orally administered BID (100 mg/kg) starting day one post implant (N=10 per group). Vehicle was orally administered BID starting day one post implant (N=20) until day five post implant, at which time, mice were randomized by tumor volume into two groups. Compound 9 (100 mg/kg) or vehicle was orally administered BID to N=10 per group starting day six post implant.FIG.13Adepicts the decrease in tumor volume with administration of Compound 9 to mice harboring established tumors.FIGS.13B-Dshow individual replications of this measurement for each dosing.FIG.13Bis vehicle.FIG.13Cis dosing of Compound 9 started on day 1.FIG.13Dis Compound 9 started on day 6. In another model, CT26 cells were implanted subcutaneously into Balb/c mice. 100 mg/kg Compound 9 or vehicle was orally administered BID starting day one post implant (N=10 per group).FIG.6Bdepicts the decrease in tumor volume compared to vehicle. **** indicates p<0.0001 vs vehicle; NS indicates not significant (two-way ANOVA). Example 213 CD73 Inhibitor Efficacy in Combination with Immunooncology and Chemotherapeutic Agents EG7 cells were implanted subcutaneously into C57BL/6 mice for each experiment. Anti-PD-L1 antibody (5 mg/kg) was dosed i.p. on Study Days 3, 5, 7, 9, 11, 13. Compound 9 (100 mg/kg) or vehicle was orally administered BID starting one day post implant.FIG.7Adepicts the reduction in tumor volume with single agent and combination therapy. ** indicates p<0.01; **** indicates p<0.0001 (two-way ANOVA).FIGS.7B-7Eshow the individual replications of this measurement for each dosing.FIG.7Bis vehicle,FIG.7Cis anti-PD-L1 antibody,FIG.7Dis Compound 9, andFIG.7Eis Compound 9+Anti-PD-L1. Oxaliplatin was dosed i.p. 6 mg/kg on Study Days 7 and 14. Compound 9 (100 mg/kg) or vehicle was orally administered BID starting one day post implant.FIG.8Adepicts the reduction in tumor volume with single agent and combination therapy. **** indicates p<0.0001 (two-way ANOVA).FIGS.8B-8Eshow the individual replications of this measurement for each dosing.FIG.8Bis vehicle,FIG.8Cis oxaliplatin,FIG.8Dis Compound 9, andFIG.8Eis Compound 9+oxaliplatin. Doxorubicin was dosed i.v. 2.5 mg/kg on Study Days 7 and 14. Compound 9 (50 mg/kg) or vehicle was orally administered BID starting one day post implant.FIG.9Adepicts the reduction in tumor volume with single agent and combination therapy. * indicates p<0.05; *** indicates p<0.001 (two-way ANOVA).FIGS.9B-9Eshow the individual replications of this measurement for each dosing.FIG.9Bis vehicle,FIG.9Cis doxorubicin,FIG.9Dis Compound 9, andFIG.9Eis Compound 9+doxorubicin. Docetaxel was dosed i.p. 5 mg/kg on Study Days 5, 12, and 19. Compound 9 (100 mg/kg) or vehicle was orally administered BID starting one day post implant.FIG.12Adepicts the reduction in tumor volume with single agent and combination therapy. * indicates p<0.05; **** indicates p<0.0001 (two-way ANOVA).FIGS.12B-10Eshow the individual replications of this measurement for each dosing.FIG.12Bis vehicle,FIG.12Cis docetaxel,FIG.12Dis Compound 9, andFIG.12Eis Compound 9+docetaxel. Example 214 CD73 Inhibitor Efficacy in Multiple Tumors Serum was procured from Discovery Life Sciences. Serum from head and neck squamous cell carcinoma (HNSCC), ovarian cancer, triple-negative breast cancer and esophageal cancer patients were incubated with a serial dilution of Compound 9 in the presence of a TNAP inhibitor. Conversion of15N5-AMP to15N5-ADO was measured by LC/MS.FIG.10Adepicts the sub-nanomolar inhibition of HNSCC serum.FIG.10Bdepicts the sub-nanomolar inhibition of ovarian cancer serum.FIG.10Cdepicts the sub-nanomolar inhibition of TNBC serum.FIG.10Ddepicts the sub-nanomolar inhibition of esophageal cancer serum. Example 215 Expression of CD73 in Multiple Human Tumors FIG.11depicts normalized mRNA expression levels of CD73 in tumor and normal tissues. Expression levels of CD73 (NT5E) were obtained from the TCGA (tumor) or GTEX (normal) databases using the UCSC Xena platform and analyzed using an unpaired t-test. The expression of CD73 as measured by a Log2(Normalized Count+1) was greater than vehicle for pancreatic, esophageal, stomach, head and neck, colon, lung and kidney clear cell tumors. INCORPORATION BY REFERENCE All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control. EQUIVALENTS While specific embodiments of the subject invention have been discussed, the above specification is illustrative and not restrictive. Many variations of the invention will become apparent to those skilled in the art upon review of this specification and the claims below. The full scope of the invention should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.
496,448
11858958
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT The following examples further illustrate the present invention, but the present invention is not limited thereto. Below presents preferred embodiments of the present invention based on the drawings in order to illustrate the technical schemes of the present invention in detail. 1. Experimental drugs: 20(S)-ginsenoside Rg3, 20(R)-ginsenoside Rg3, 20(S)-ginsenoside Rh2, 20(R)-ginsenoside Rh2 are commercially available in this field, such as Shanghai Ginposome PharmaTech Co., Ltd., Suzhou Star Ocean Ginseng Bio-pharmaceutical Co., Ltd., and/or Shanghai Yuanye Bio-Technology Co., Ltd. 2. Experimental Instruments: The instruments used in the following embodiments are self-owned by Shanghai Ginposome PharmaTech Co., Ltd., the model and supply information of the instruments are listed as follows: Ultra-Micro Pulverizer (ZD-10S, Shanghai Lvyi Machinery Manufacturing Co., Ltd.) High performance liquid chromatography (Agilent 1100), Alltech 3300ELSD detector, Anjielun Technology China Co., Ltd. Rotary evaporator (ZX98-1 5L), Shanghai Looyesh Instrument Co., Ltd.; 20 L Rotary evaporator (R5002K), Shanghai Xiafeng Instrument Factory; Lyophilizer (FD-1D-80), Shanghai Bilang Instrument Manufacturing Co. Ltd.; Lyophilizer (PDFD GLZ-1B), Shanghai Pudong Freeze dryer Equipment Co., Ltd.) Precision weighing balance (CPA2250 0.00001 g Readability), Sartorius (Shanghai) Trade Co., Ltd.; Electronic balance (JY3003 0.001 g Readability), Shanghai Shunyu Hengping Science Instrument Co. Ltd.). 3. The present invention is further explained by the following embodiments, but not limited to the following embodiments. The experimental methods without giving specific conditions, are carried out by conventional methods and conditions used in this field, or according to commodity specifications. The temperature and pressure preferably refer to room temperature of 10 to 30° C. and standard atmosphere pressure if not specified. Reflux temperature, if not specified, is defined by the solvent used. Ultrafine Powder Process To get the ginsenoside Rg3 ultrafine powder, 500 g ginsenoside Rg3 is dried to water content less than 1% and crushed by Ultra-Micro Pulverizer ZD-10S for 30 min. During the process, the inside temperature of pulverizer chamber is maintained at 20-30° C. with a cooled circulating water. The average size of more than 90% particles is less than 10 μm measured by electron microscope. The Preparation of the Liposomes Embodiment 1 The Preparation of a Conventional Rg3 Liposome A mixture of Egg lecithin 1 g, cholesterol 0.1 g and ginsenoside 20(S)-Rg3 (without ultra-micro pulverization) 0.1 g were added to 20 mL anhydrous ethanol and stirred at room temperature to form a clear solution. Then the organic solvent was removed by a rotary evaporator in a thermostatic water bath at 40 to 50° C. The formed thin film was hydrated with 20 mL 5% trehalose aqueous solution (the percentage refers to the ratio of the mass of the trehalose to the total mass of the trehalose aqueous solution). The suspension was then sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. After sonication, the liposome suspension was passed through a 0.22-micron microporous membrane to obtain an aqueous solution of ginsenoside Rg3 liposome. Then the aqueous solution was aliquoted into vials and placed in a freeze-dryer to lyophilization for 72 hours. The conventional Rg3 liposome was obtained and sealed in the vial by a protective gas (argon or nitrogen). By calculation, D10 of the liposome was 75 nm, D50 was 118 nm, D90 was 131 nm. As hereinafter, D10, D50, and D90 describe diameter, where 10%, 50%, and 90% of particle size distribution were under the reported particle size. Embodiment 2 The Preparation of Rg3 Blank Liposome Egg lecithin 1 g and ginsenoside 20(S)-Rg3 ultrafine powder 0.1 g were added to 200 mL chloroform and stirred to form a clear solution at room temperature. The organic solvent was removed by a rotary evaporator in a thermostatic water bath at 40 to 50° C. to form a film. The formed thin film was hydrated with 20 mL 5% trehalose aqueous solution (the percentage refers to the ratio of the mass of the trehalose to the total mass of the trehalose aqueous solution). The liposome suspension was sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. Then the suspension was passed through a 0.22-micron microporous membrane to obtain an aqueous solution of ginsenoside Rg3 liposome. Then the aqueous solution was aliquoted into vials and placed in a freeze-dryer to for 72 hours. After lyophilization, the obtained Rg3 blank liposome was sealed in the vial by a protective gas (argon or nitrogen). By calculation, the D10 of the liposome was 66 nm, D50 was 90 nm, D90 was 105 nm. Embodiment 3 The Preparation of Rg5 Blank Liposome In accordance with the method in embodiment 2, the Rg5 Blank liposome was prepared by replacing Rg3 with Rg5. After evaluation, the D10 of the liposome was 70 nm, D50 was 96 nm and D90 was 111 nm. Embodiment 4 The Preparation of Rg3 Blank Liposome Egg lecithin 0.5 g, ginsenoside 20(R)-Rg3 ultrafine powder 0.1 g and Vitamin E 0.1 g were added into 200 mL dichloromethane and stirred to form a clear solution at room temperature. The organic solvent was removed by a rotary evaporator in a thermostatic water bath at 40 to 50° C. to form a film. The formed thin film was hydrated with 20 mL 5% glucose aqueous solution (the percentage refers to the ratio of the mass of the glucose to the total mass of the glucose aqueous solution). The liposome suspension was sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. Then the suspension was passed through a 0.22-micron microporous membrane to obtain an aqueous solution containing ginsenoside Rg3 liposome. Then the aqueous solution was aliquoted into vials and placed in a freeze-dryer to for 72 hours. After lypholization, the obtained Rg3 blank liposome was sealed in the vial by a protective gas (argon or nitrogen). By calculation, the D10 diameter of the liposome was 88 nm, D50 was 116 nm, D90 was 153 nm. Embodiment 5 The Preparation of Rg3 Blank Liposome Soybean lecithin 0.6 g and ginsenoside 20(S)-Rg3 ultrafine powder 0.2 g were added into 200 mL chloroform/methanol (1:1, v/v) and stirred to form a clear solution at room temperature. The organic solvent was removed by a rotary evaporator in a thermostatic water bath at 50 to 60° C. to form a film. The formed thin film was then hydrated with 20 mL 5% sucrose aqueous solution (the percentage refers to the ratio of the mass of the sucrose to the total mass of the sucrose aqueous solution) and then sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. The liposome suspension was passed through a 0.22-micron microporous membrane to obtain an aqueous solution containing ginsenoside Rg3 blank liposome. Then the aqueous solution was aliquoted into vials and placed in a freeze-dryer for 72 hours. After lyophilization, the obtained Rg3 blank liposome was then sealed by a protective gas (argon or nitrogen). By calculation, the D10 of the liposome was 60 nm, D50 was 84 nm, D90 was 102 nm. Embodiment 6 The Preparation of Rh2 Blank Liposome Hydrogenated soybean lecithin (HSPC) 0.7 g, ginsenoside 20(S)-Rh2 ultrafine powder 0.1 g and cholesterol 0.2 g were added into 200 mL chloroform and stirred to form a clear solution at room temperature. The organic solvent was removed by rotary evaporation in a thermostatic water bath at 60° C. to 65° C. to form a film. The formed film was hydrated using 20 mL 5% mannitol aqueous solution (the percentage refers to the ratio of the mass of the mannitol to the total mass of the mannitol aqueous solution) and then sonicated until the particle size of the liposome was between 0.1 and 0.3 micron to obtain an aqueous solution of ginsenoside Rh2 blank liposome. Then the obtained aqueous solution was aliquoted into vials and placed in a freeze-dryer to lyophilization for 72 hours. Then the obtained Rh2 blank liposome was sealed in the vial by a protective gas (argon or nitrogen). By calculation, the D10 of the liposome was 94 nm, D50 was 120 nm, D90 was 133 nm. Embodiment 7 The Preparation of Rh2 Blank Liposome Egg lecithin 0.4 g, ginsenoside 20(R)-Rh2 ultra-fine powder 0.1 g, soybean oil 0.2 g and vitamin C 0.1 g were added into 200 mL chloroform/isopropyl alcohol(9:1 v/v) and stirred to form a clear solution at room temperature. The organic solvent was removed by rotary evaporation in a thermostatic water bath at 60° C. to 65° C. to form a film. The formed film was hydrated with 20 mL 5% propanediol aqueous solution (the percentage refers to the ratio of the mass of the propanediol to the total mass of the propanediol aqueous solution) and sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. After sonication, the liposome suspension was passed through a 0.22-micron microporousmembrane to obtain an aqueous solution of ginsenoside Rh2 blank liposome. Then the aqueous solution was aliquoted into vials and placed in a freeze-dryer to lyophilization for 72 hours. Then the obtained Rh2 blank liposome was sealed in the vial by a protective gas (argon or nitrogen). By evaluation, the D10 diameter of the liposome was 124 nm, D50 was 157 nm, D90 was 189 nm. Embodiment 8 The Preparation of Rg3 Blank Liposome Egg lecithin 0.9 g, ginsenoside 20(S)-Rg3 ultrafine powder 0.2 g and PEG2000-DSPE 0.05 g were mixed with 200 mL chloroform/methanol (1:1, v/v) and stirred to form a clear solution at room temperature. The organic solvent was removed by rotary evaporation in a thermostatic water bath at 55 to 65° C. to form a film. The formed film was hydrated with 20 mL 5% glycerol aqueous solution (the percentage refers to the ratio of the mass of the glycerol to the total mass of the glycerol aqueous solution) and sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. After sonication, the liposome suspension was passed through a 0.45-micron microporous membrane filter to obtain an aqueous solution of ginsenoside Rg3 blank liposome. Then the aqueous solution was aliquoted into vials and placed in a freeze-dryer to lyophilization for 72 hours. Then the obtained Rg3 blank liposome was sealed in the vial by protective gas (argon or nitrogen). By calculation, the D10 diameter of the liposome was 62 nm, D50 was 71 nm, D90 was 85 nm. Embodiment 9 The Preparation of Rg3 Blank Liposomes Soybean lecithin S100 0.9 g, ginsenoside 20(S)-Rg3 ultrafine powder 0.2 g, Vitamin E 0.01 g, cholesterol 0.1 g and mPEG2000-DSPE 0.05 g were mixed with 20 mL chloroform/acetone (1:1 v/v) and stirred to form a clear solution at room temperature. The organic solvent was removed by rotary evaporation in a thermostatic water bath at 45° C. to 55° C. to form a film. The formed film was hydrated with 20 mL 5% galactose aqueous solution (the percentage refers to the ratio of the mass of the galactose to the total mass of the galactose aqueous solution) and sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. After sonication, the liposome suspension was passed through a 1-micron microporous membrane to obtain an aqueous solution of ginsenoside Rg3 blank liposome. Then the aqueous solution was aliquoted into vials and placed in a freeze-dryer for 72 hours. After lyophilization, Then the obtained Rg3 blank liposome was sealed in the vial and protected by argon gas or nitrogen gas. By calculation the D10 diameter of the liposome was 65 nm, D50 was 130 nm, D90 was 143 nm. Embodiment 10 The Preparation of Paclitaxel Rg3 Liposome Egg lecithin 0.8 g, ginsenoside 20(S)-Rg3 ultrafine powder 0.2 g and Paclitaxel 0.1 g were mixed with 200 mL chloroform and stirred to form a clear solution at room temperature. The organic solvent was removed by rotary evaporation in a water bath thermostatically controlled at 40° C. to 50° C. to form a film. The formed film was hydrated with 20 mL 5% trehalose aqueous solution (the percentage refers to the ratio of the mass of the trehalose to the total mass of the trehalose aqueous solution) and sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. Thus, an aqueous solution of Paclitaxel Rg3 liposome was obtained. Then the aqueous solution was aliquoted into vials making 30 mg Paclitaxel in each vial. The aqueous solution was placed in a freeze-dryer for 72 hours. After lyophilization, the obtained Paclitaxel Rg3 liposome was sealed in the vial and protected by argon gas or nitrogen gas. By evaluation, the D10 diameter of the liposome was 76 nm, D50 was 90 nm, D90 was 105 nm, the encapsulation efficiency was more than 95%. Embodiment 11 The Preparation of Paclitaxel Rg5 Liposome In accordance with the method in embodiment 10, the Paclitaxel Rg5 liposome were prepared by replacing Rg3 with Rg5. By evaluation, the D10 of the liposome was 92 nm, D50 was 128 nm, D90 was 158 nm, the encapsulation efficiency was more than 95%. Embodiment 12 The Preparation of Paclitaxel Rh2 Liposome Soybean lecithin 0.7 g, ginsenoside 20(S)-Rh2 ultrafine powder 0.2 g, Paclitaxel 0.1 g, cholesterol 0.1 g, soybean oil 0.1 g and vitamin C 0.1 g were mixed with 200 mL chloroform/acetonitrile (1:1, v/v) and stirred to form a clear solution at room temperature. The organic solvent was removed by rotary evaporation in a water bath thermostatically controlled at 50-60° C. to form a film. The formed film was hydrated with 20 mL 10% treassose aqueous solution (the percentage refers to the ratio of the mass of the trehalose to the total mass of the trehalose aqueous solution) and sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. After sonication, an aqueous solution of paclitaxel Rh2 liposome was obtained. Then the aqueous solution was aliquoted into vials making 30 mg paclitaxel in each vial. The aqueous solution was placed in a freeze-dryer for 72 hours. After lyophilization, the obtained paclitaxel Rh2 liposome was sealed in the vial and protected by argon gas or nitrogen gas. By evaluation, the D10 diameter of the liposome was 79 nm, D50 was 118 nm, D90 was 130 nm, the encapsulation efficiency is more than 95%. Embodiment 13 The Preparation of Docetaxel Rg3 Liposome Egg lecithin 0.9 g, ginsenoside 20(S)-Rg3 ultrafine powder 0.18 g, Docetaxel 0.1 g and cholesterol 0.225 g were mixed with 200 mL chloroform/methanol (1:1, v/v) and stirred in a water bath thermostatically controlled at 40-50° C. to form a clear solution. The organic solvent was removed by a membrane evaporator at 50° C. to 60° C. to form a film. The formed film was hydrated with 20 mL 5% sucrose aqueous solution (the percentage refers to the ratio of the mass of the sucrose to the total mass of the sucrose aqueous solution) and homogenized by a high-pressure homogenizer until the particle size of the liposome was between 0.1 and 0.3 micron. After homogenization, the liposome suspension was passed through a 0.22-micron microporous membrane to obtain an aqueous solution of docetaxel Rg3 liposome. Then the aqueous solution was aliquoted into vials making 20 mg docetaxel in each vial. The aqueous solution was placed in a freeze-dryer for 72 hours. After lyophilization, the obtained docetaxel Rg3 liposome was sealed in the vial and protected by argon gas or nitrogen gas. By evaluation, the D10 diameter of the liposome was 70 nm, D50 was 109 nm, D90 was 122 nm, the encapsulation efficiency was more than 95%. Embodiment 14 The Preparation of Docetaxel Rg5 Liposome Egg lecithin 0.9 g, ginsenoside Rg5 ultra-fine powder 0.18 g, Docetaxel 0.1 g and cholesterol 0.225 g were mixed with 20 mL chloroform/methanol (1:1, v/v) and stirred in a water bath thermostatically controlled at 40-50° C. to form a clear solution. The organic solvent was removed by a membrane evaporator at 50° C. to 60° C. to form a film. The formed film was hydrated with 20 mL 5% sucrose aqueous solution (the percentage refers to the ratio of the mass of the sucrose to the total mass of the sucrose aqueous solution) and homogenized with a high-pressure homogenizer until the particle size of the liposome was between 0.1 and 0.3 micron. After homogenization, the liposome suspension is filtered by a 0.22-micron microporousmembrane to give an aqueous solution of docetaxel Rg5 liposome. Then the aqueous solution is aliquoted into vials making that each vial contains docetaxel 20 mg, then placed in a freeze-dryer to freeze dry for 72 hours. After lyophilization, the obtained docetaxel Rg5 liposome was sealed in the vial and protected by argon gas or nitrogen gas. By calculation, the D10 of the liposome was 73 nm, D50 was 101 nm, D90 was 118 nm, the encapsulation efficiency was more than 95%. Embodiment 15 The Preparation of Docetaxel Rh2 Liposome Soybean lecithin 300 mg, ginsenoside 20(S)-Rh2 ultrafine powder 60 mg, Docetaxel 30 mg, cholesterol 75 mg and mPEG-DSPE 10 mg were mixed with 200 mL chloroform/methanol (1:1, v/v) and stirred to form a clear solution in a water bath thermostatically controlled at 40-50° C. The organic solvent was removed by a membrane evaporator at 50° C. to 60° C. to form a film. The formed film was hydrated with 20 mL 5% sucrose aqueous solution (the percentage refers to the ratio of the mass of the sucrose to the total mass of the sucrose aqueous solution) and homogenized with a high-pressure homogenizer until the particle size of the liposome was between 0.1 and 0.3 micron. After homogenization, the liposome suspension was passed through a 0.22-micron microporous membrane to give an aqueous solution of docetaxel Rh2 liposome. Then the aqueous solution was aliquoted into vials making that each vial contains docetaxel 20 mg, then placed in a freeze-dryer to freeze dry for 72 hours. After lyophilization, the obtained docetaxel Rh2 liposome was sealed in the vial and protected by argon gas or nitrogen gas. By calculation, the D10 diameter of the liposome was 81 nm, D50 was 129 nm, D90 was 148 nm, and the encapsulation efficiency was 95%. Embodiment 16 The Preparation of Rg3 Irinotecan Liposomes Egg lecithin 0.9 g, ginsenoside 20(S)-Rg3 ultrafine powder 0.3 g and cholesterol 0.1 g were mixed with 200 mL dichloromethane/ethanol (1:1, V/V) and stirred to form a clear solution at room temperature. The organic solvent was removed by a rotary evaporator in a water bath thermostatically controlled at 50° C. to 60° C. to form a film. The formed film was hydrated with 20 mL 6.6% ammonium sulfate aqueous solution (the percentage refers to the ratio of mass of the ammonium sulfate to the total mass of the ammonium sulfate aqueous solution) and sonicated until the particle size of the blank liposome was between 0.1 and 0.3 micron to give an aqueous solution of Rg3 blank liposome. The solution of the blank liposome was dialyzed against 0.15 mol/L trehalose solution for 12 hours. After dialyzation, a certain amount of trehalose was added according to the volume of the dialyzed blank liposome solution to make the mass percentage of trehalose in the blank liposome solution reach 10% (the mass percentage refers to the mass of the trehalose relative to the total mass of the blank liposome solution). Then, 1 mL irinotecan hydrochloride aqueous solution (containing irinotecan hydrochloride 0.2 g with a mass percentage of 20%) was added and kept for 30 minutes in a water bath at 37° C. to give an aqueous solution of ginsenoside Rg3 irinotecan hydrochloride liposome. The aqueous solution was aliquoted into vials making that each vial contains 40 mg irinotecan hydrochloride, and then placed in a freeze-dryer to freeze dry for 72 hours. The obtained ginsenoside Rg3 irinotecan hydrochloride liposome was sealed in the vial filled with protective gas (argon or nitrogen). By calculation, the D10 diameter of the liposome was 92 nm, D50 was 139 nm, D90 was 165 nm. The encapsulation efficiency was more than 95%. Embodiment 17 The Preparation of Rg3 Cisplatin Liposome Egg lecithin 0.8 g, ginsenoside 20(S)-Rg3 ultra-fine powder 0.2 g, cisplatin 0.1 g and soybean oil 0.1 g were mixed with 200 mL chloroform/methanol (1:1, v/v) and stirred to form a clear solution at room temperature. The organic solvent was removed by a rotary evaporator in a water bath thermostatically controlled at 40° C. to 50° C. to form a film. The formed film was hydrated with 20 mL 5% lactose aqueous solution (the percentage refers to the ratio of the mass of the lactose to the total mass of the lactose aqueous solution) and sonicated until the particle size of the liposome was between 0.1 and 0.3 micron. After sonication, the liposome suspension was passed through 1-micron microporous membrane to give an aqueous solution of cisplatin Rg3 liposome. Then the aqueous solution was aliquoted into vials making that each vial contains cisplatin 10 mg, and then placed in a freeze-dryer to freeze dry for 72 hours. After lyophilization, the obtained cisplatin Rg3 liposome was sealed in the vial filled with protective gas (argon or nitrogen). By calculation, the D10 of the liposome was 69 nm, D50 was 109 nm, D90 was 126 nm, and the encapsulation efficiency was more than 95%. Embodiment 18 The Preparation of Rg3 Doxorubicin Liposome Soybean lecithin S100 0.9 g, ginsenoside 20(S)-Rg3 ultrafine powder 0.3 g and vitamin E 0.1 g were mixed with 200 mL chloroform/methanol (9:1, v/v) and stirred to form a clear solution in a water bath thermostatically controlled at 40° C.-50° C. The organic solvent was removed by a membrane evaporator at 50° C.-55° C. to form a film. The formed film was hydrated with 20 mL phosphoric acid buffer salt (PBS), stirred to form a clear solution. The clear solution is homogenized by a high-pressure homogenizer until the particle size of the liposome was between 0.1 and 0.3 micron to give an aqueous solution of Rg3 blank liposome. Then the aqueous solution was mixed with 1 mL doxorubicin hydrochloride aqueous solution with a mass percentage of 20% (doxorubicin hydrochloride 0.2 g) and 6 mL disodium hydrogen phosphate aqueous solution with a mass percentage of 7.1%, and purified water was added to adjust pH to 7.30. The mixture was kept in a water bath at 60° C. for 30 minutes to give an aqueous solution of ginsenoside Rg3 doxorubicin hydrochloride liposome. Then the aqueous solution was aliquoted into vials making that each vial contains 20 mg doxorubicin hydrochloride, and placed in a freeze-dryer for 72 hours. After lyophilzation, the obtained ginsenoside Rg3 doxorubicin hydrochloride liposome was sealed in a vial filled with protective gas (argon or nitrogen). By calculation, the D10 diameter of the liposome was 76 nm, D50 was 101 nm, D90 was 125 nm. The encapsulation efficiency was more than 95%. Application Embodiments 1. Experimental Drugs Ginsenoside 20(S)-Rg3 (Rg3), paclitaxel, docetaxel, irrinotecan hydrochloride, doxorubicin and cisplatin are commercially available in this field. If without giving specific instructions, the conventional Rg3 liposomes were carried out according to embodiment 1, the Rg3 or Rh2 blank liposomes were carried out according to embodiment 2, Rg5 blank liposomes were carried out according to embodiment 3, Paclitaxel Rg3 liposomes were carried out according to embodiment 10, Paclitaxel Rg5 liposomes were carried out according to embodiment 11, Docetaxel Rg3 liposomes were carried out according to embodiment 13, Docetaxel Rg5 liposomes were carried out according to embodiment 14. Each ginsenoside blank liposome was either prepared according to the above-mentioned method in the present invention, or according to embodiment 1 and making corresponding changes according to the needs. 2. Instruments The instruments used in the following embodiments and the application embodiments are self-owned by the School of Pharmacy, Fudan University, and the model and other information of the instruments are listed as follows:High performance liquid chromatography (HPLC), (Agilent 1100),Electronic balance (TB-215, Denver Instrument, USA);Ultrasonic cleaning machine (SB3200DT, Ningbo Xinzhi Biotechnology Co., Ltd.);Terbovap Sample Concentrator (HGC-12A, Tianjin Hengao Technology Development Co., Ltd.)Rotary evaporator (RE-2000A, Shanghai Yarong Biochemical Instrument Factory);Ultrapure water system (ULUP-IV-10T, Sichuan U & P Ultra Technology Co., Ltd.)Thermostatic oscillator (SHA-C, Changzhou Aohua Instrument Co., Ltd.)Ultrasonic cell crusher (JY92-II, Ningbo Xinzhi Biotechnology Co., Ltd.);High pressure homogenizer (EmulsiFlex™-B15, AVESTIN Inc., Canada);Laser particle size analyzer (Zetasizer Nano ZS, Malvern Panalytical Ltd. UK);Mini-extruder Equipment (Avanti Polar Lipids Inc);Photoelectric Microscope (XDS-1B, Chongqing Optical Instrument Co., Ltd.);Clean bench (SW-CJ-1FD, Suzhou Antai air Technology Co., Ltd.);Cell incubator (CCL-170B-8, ESCO, Singapore);Fluorescence inverted microscope (IX-73, Olympus, Japan);Laser granulometer (Mastersizer 2000, Malvern Panalytical Ltd., UK);In-vivo Small animal imaging system (In-vivo Multispectral FX PRO, Bruker Corporation, US). 3. Experimental Cell Lines:4T1 human breast cancer cell line (Nanjing KeyGEN Biotech Co., Ltd)A549 human lung cancer cell line (Nanjing KeyGEN Biotech Co., Ltd)BGC-823 human gastric adenocarcinoma cancer cell line (Nanjing KeyGEN Biotech Co., Ltd)In-situ glioma model in C6 cells (Nanjing KeyGEN Biotech Co., Ltd)Rat C6 glioma cell line (Nanjing KeyGEN Biotech Co., Ltd) 4. In Vitro Hemolysis Test Preparation of 2% red blood cell suspension: The blood from a healthy rabbit was collected into a conical flask containing glass beads and shook for 10 minutes, or the blood was agitated using a glass rod to remove the fibrinogen from blood and make defibrinated blood. Then, about 10 times volum of 0.9% sodium chloride solution was added to wash the cell. After centrifugation for 15 minutes at 1000-1500 RPM, the supernatant was discarded and and red blood cells were collected in the precipitation. Then, the red blood cell was obtained after washing the precipitation using 0.9% sodium chloride solution for 2-3 times according to the method above until the supernatant was clear. To obtain a 2% cell suspension, the obtained red blood cells were suspended in 0.9% sodium chloride solution. Hemolysis Test: 5 clean glass tubes were labelled with numbers. Tube number 1, 2 were used for test samples, tube number 3 was used for negative control, tube number 4 was used for positive control, tube number 5 was used for the contrast sample. As shown in table 5, 2% red blood cell suspension, 0.9% Sodium Chloride Solution, and purified water were added to the tube. After mixing, the tubes were incubated at 37±0.5° C. for 3 h. Results of hemolysis and aggregation were observed and recorded as shown in Table 5. TABLE 5Test tube No.123452% red cell suspensions/mL2.52.52.52.5/0.9% sodium chloride solution/mL2.22.22.5/4.7Purified water/mL///2.5/The test solution/mL0.30.3//0.3 If it gave a clear and red solution in the tube, and no cells were settled at the bottom of the tube, it suggested hemolysis occurred. If it gave a colorless or clear solution and red blood cells were all settled at the bottom of the tube, or the supernatant was lightly colored, but no significant differences were observed between tube 1 or 2 and tube 5, it suggested no hemolysis occurred. If there was red/brown cloudy precipitate in the solution, thoroughly mixed the sample by gently inverting the tube 3 times. If the precipitate was still there, it indicated red blood cell aggregation. The sample should be further observed under microscope to confirm if red blood cell aggregation occurred. Results Analysis: If no hemolysis or aggregation occurs in the tube of negative control, but hemolysis occurred in the tube of positive control, and no hemolysis and aggregation occurs in the two tubes of test samples within 3 hours, the test sample meet the regulations. If hemolysis and aggregation occurs in one of the tubes with test sample within 3 hours, four more sample tests should be performed to confirm. Only when no hemolysis and aggregation occurs within 3 hours in all the four sample tubes, the test sample can be conformed that it meets the requirements, otherwise the test sample does not meet the requirements. In a specific experiment, concentration of the test sample (ginsenoside) can be adjusted according to the needs. 5. Experimental Animals Experimental animals: Kunming mice (or normal mice) are purchased from the Animal Center of the Third Military Medical University, BALB/C-nu/nu mice (or nude mice) are purchased from Shanghai SlACK Laboratory Animal Co., Ltd. 6. Cell Culture Method Cell lines were incubated at 37° C. in a humidified incubator with 5% CO2, and cultured in DMEM or RPMI1640 complete culture-medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 μg/mL streptomycin. A solution of 0.25 trypsin-EDTA was used for sub-culturing cells, which was performed 2 to 3 times per week. 7. Drug Administration A negative control group (e.g. PBS group), a positive control group and a sample group (ginsenoside liposome loaded with a drug) were set up for each experiment. A total of 3-6 concentration gradients were set up, including half dilution or 5 times dilution. Each concentration repeated 3 times. 8. Determination of the Half-Maximal Inhibitory Concentration (IC50) of Tumor Cell Tumor cells in logarithmic growth phase were digested with trypsin and centrifuged, collected the cell pellet and resuspended it in a buffer. Then cells in the suspension solution were counted and seeded into a 96-well culture plate with 5000 cells per well by placing 100 μl cell suspension solution in each well. On the next day, 100 μl fresh culture medium containing different concentrations of samples or solvent as control were added to each well respectively (with a final concentration of DMSO<0.5%). For each sample, 10 different dose groups were set up, and each group repeated 3 times parallelly. After 72-hour incubation at 37° C., the supernatant was discarded and 100 μl PBS and 10 μl CCK-8 were added to each well. Then the plate was well shaked using a micro oscillator for uniform and continually cultured for 3 h. Absorbance is determined by a microplate reader at a reference wavelength of 630 nm and a detection wavelength of 450 nm. Tumor cells treated with a solvent were used as a control, IC50is computed from the median-effect equation. 9. Determination of Cell Viability In Vitro Logarithmically growing tumor cells were collected and resuspended in DMEM complete medium supplemented with 10% fetal bovine serum, 100 U/ml penicillin and 100 μg/ml streptomycin to a final cell density of 4×104cells/ml. Then, 200 μl cell suspension solution was seeded into each well of a 96-well plate (with a concentration of 8×103cells/well) and the plate was cultured in a CO2cell culture incubator at 37° C. After 48 h, DMEM complete medium was removed and respectively replaced with 200 μL different concentrations of anti-cancer drug, at least 6 different concentration groups. The group without replacing DMEM complete medium by anti-cancer drug solution was used as negative control. For each concentration group, 4 replicates were set up. The whole experiment was independently repeated 3 times. The cells were continuously cultured in a CO2cell culture incubator at 37° C. After 72 h, 20 μl 5 mg/mL MTT solution was added into each well and the plate was continuously cultured for 4 h. Then discarded the supernatant, added 150 μl DMSO into each well, and shaked the plate for 10 min. The absorbance was measured at 490 nm using a microplate reader (Tecan infinite M 200 TECAN, Switzerland). The cell survival rate is calculated according to the following formula: Cell⁢⁢Survival⁢⁢Rate⁢⁢(%)=Abs490⁢(sample)Abs490⁢(control)×100 Wherein Abs490(sample)is the absorbance of the experimental sample, Abs490(control)is the absorbance of the negative control. Small Animal Imaging In Vivo As shown in the embodiments. 11. In-Vivo Drug efficacy Test 100 uL logarithmically growing tumor cells with a density of 1×107to 10×107cells/mL was injected subcutaneously into the right armpit of an 18 to 20 g nude mouse slowly using a 1 mL syringe. The growth of the tumor was observed. When the tumor volume was about 100 mm3, animals were randomized to groups and administered with different drugs. All mice were weighed, and the longest diameter and the shortest diameter of the tumor was measured with vernier calipers every two days. At the end of the experiment, the nude mice were sacrificed and the volumes of tumors were calculated. Then, the relative tumor volume (RTV), T/C ratio (the ratio of tumor volume in control versus treated mice) and the percent tumor growth inhibition (TGI) were calculated and statistically analyzed. Tumor volume was calculated according to the following formula: V=(L×W×H)/2, wherein V is tumor volume, L is tumor length, W is tumor width, H is tumor height. Relative tumor volume was calculated according to the following formula: RTV=TVn/TV0, wherein TVnis the tumor volume at day n, TV0is the tumor volume at day zero (the administration day). The T/C ratio was determined by calculating RTV: T/C(%)=TRTV/CRTV×100%, wherein TRTV is the RTV of the treatment group, CRTV is the RTV of the control group. The percent tumor growth inhibition (TGI) was calculated according to the following formula: TGI(%)=((MTVcontrol−MTVtreated/MTVcontrol))×100, wherein MTVcontrol is the median tumor volume of control group, MTVtreated is the median tumor volume of the drug treatment group. Curative effect was evaluated based on the T/C ratio: T/C (%)>60 means the treatment has no effect; T/C (%)≤60 and the differences between the treatment group and the control group are statistically significant (P<0.05) means the treatment is effective. In the following application embodiments, C(μM) means concentration, wherein the concentration of Taxol+Rg3 refers to the concentration of paclitaxel and ginsenoside Rg3 in the ginsenoside Rg3 paclitaxel liposome, for example, 5+30 means that in ginsenoside Rg3 paclitaxel liposome, the concentration of the paclitaxel is 5 μM and the concentration of the ginsenoside Rg3 is 30 μM. Time (d) is calculated by days. 12. Analysis Method of Paclitaxel Analysis of paclitaxel is according to the Paclitaxel analysis method in the United States Pharmacopeia (USP 34). Application Embodiments Embodiment 1 Hemolysis Test Experimental results are listed in table 1. HD50 is 50% of the maximum haemolysis. TABLE 1AbbreviationEmbodimentof LiposomeLiposomeHemolysisNo.NameFull Name(HD50)Embodiment 1Rg3-Cho-Lipoconventional20-50μg/mLRg3 cholesterolliposomeEmbodiment 2Rg3-blankRg3 blank650-700μg/mLliposomeEmbodiment 3Rg5-blankRg5 blank450-500μg/mLliposomeEmbodiment 7Rh2-blankRh2 blank400-500μg/mLliposomeEmbodiment 10PTX-Rg3-GipoPaclitaxel Rg3650-700μg/mLLiposomeEmbodiment 11PTX-Rg5-GipoPaclitaxel Rg5450-500μg/mLLiposomeEmbodiment 12PTX-Rh2-GipoPaclitaxel Rh2400-500μg/mLLiposomeEmbodiment 13DTX-Rg3-GipoDocetaxel Rg3650-700μg/mLLiposomeEmbodiment 14DTX-Rg5-GipoDocetaxell Rg5450-500μg/mLLiposomeEmbodiment 15DTX-Rh2-GipoDocetaxel Rh2400-500μg/mLLiposome As shown in Table 1, Rg3-Cho-Lipo showed severe hemolytic effect, whereas the hemolytic effects of Rg3-Blank, Rh2-Blank, PTX-Rg3-Gipo, PTX-Rh2-Gipo, DTX-Rg3-Gipo and DTX-Rh2-Gipo were similar to those of Rg5-Blank, PTX-Rg5-Gipo and DTX-Rg5-Gipo with HD50 value in the range of 400-700 μg/mL, which can meet the safety standards of medicinal products. In addition, conventional Rg3-Cho-Lipo did not show hemolysis up to a concentration of 20-50 μg/mL, mainly because that the encapsulation efficiency of the conventional Rg3 cholesterol liposome was low and Rg3 may leak more or less, thereby affecting the drug efficacy. Whereas, the encapsulation efficiency of the ginsenoside liposomes obtained from embodiment 2, embodiment 3, embodiment 7, embodiment 10, embodiment 12-13 and embodiment 15 in the present invention were high, similar to the encapsulation efficiency of Rg5-blank, PTX-Rg5-Gipo and DTX-Rg5-Gipo, thus, these drugs were all very efficient. Besides Rg3 and Rh2, it can further encapsulate drugs, such as Paclitaxel, indicating that Rg3 is used as membrane material in these liposomes. Application Embodiment 2 Studies on the Effect of Mass Percentage of Ginsenoside in the Liposome on the Average Particle Size of the Liposome Sample test before lyophilization: 20 mL sample solution was diluted into 900 mL purified water at room temperature. The mixture was stirred for 1 min at 1700 rpm/min. Then, the sample was tested and the results were recordered. Sample test after lyophilization: A vial of lyophilized sample was hydrated with 20 mL purified water. Then, shaked the vial until the sample was fully dissolved. The sample solution was diluted into 900 mL purified water at room temperature and stirred for 1 min at 1700 rpm/min. Then, the sample was tested and the results were recorded. The experimental results are shown in Table 2. TABLE 2Effects of mass percentage of ginsenoside in the liposome on theaverage particle size of the liposomeMass percentageAverageEncapsu-Preparationof 20(S)-Rg3particlelationNamemethodin liposomessizeefficiencyRg3-AccordingEgg lecithin:Rg3 =147nm≥95%cholesterolto10:0.1liposomeembodimentEgg lecithin:Rg3 =438nm≥85%110:1Egg lecithin:Rg3 =≥1μm≤80%10:2Egg lecithin:Rg3 =≥1μm≤80%10:5Rg3-Blankccording toEgg lecithin:Rg3 =116nm≥95%liposomeembodiment10:0.12Egg lecithin:Rg3 =92nm≥95%10:1Egg lecithin:Rg3 =126nm≥95%10:2Egg lecithin:Rg3 =185nm≥95%10:5Paclitaxel-AccordingEgg103nm≥95%Rg3tolecithin:Rg3:Paclitaxel =Liposomeembodiment10:0.1:0.0510Egg85nm≥95%lecithin:Rg3:paclitaxel =10:1:0.5Egg157nm≥95%lecithin:Rg3:Paclitaxel =10:2:1Egg243nm≥95%lecithin:Rg3:paclitaxel =10:5:2.5 As showed in Table 2, the particle size increased and encapsulation efficiency decreased while increasing the mass percentage of Rg3 in Rg3-cholesterol liposome. Huan Yu, et al disclosed a Rg3-cholesterol liposome, which, in fact, is a conventional blank liposome loaded with Rg3 (See: International Journal of Pharmaceutics 450(2013)250-258). In the conventional Rg3-cholesterol liposomes, Rg3 is an active substance. With the increasing mass percentage of the Rg3, the encapsulation efficiency decreases and the particle size increases. Whereas, in the present invention, Rg3 is used as membrane material. With the increasing mass percentage of Rg3, particle size of the liposome becomes smaller and all the encapsulation efficiency are more than 95%. Therefore, Rg3 is used as membrane material in the present invention. Properties of the liposome also changes with the changes of the membrane material. Application Embodiment 3 The Determination of Particle Size Distribution, Dispersion Coefficient and Electron Microscope Imaging of Paclitaxel Cholesterol Liposome and Paclitaxel Rg3 Liposome The determination of particle size distribution and dispersion coefficient: samples of PTX-Cho-Lipo and PTX-Rg3-Gipo were diluted 10 times. Then 1 mL diluted solution was added into the sample pool of Malvern laser particle size analyzer. Test results were recorded and analyzed. Morphology test of liposomes: 150 μL PTX-Cho-Lipo solution and PTX-Rg3-Gipo solution were each diluted into 5 mL purified water. After dilution, a drop was placed on a carbon-coated copper grid and air dried for 10 minutes, then the sample was stained with 2% sodium acetate for 30 minutes. After removing the excess staining solution using a filter paper, the morphology of liposomes was observed and imaged using transmission electron microscope (TEM). The experimental results are listed in Table 3. TABLE 3The particle sizes of PTX-Cho-Lipo and PTX-Rg3-GipoMeanDistributionParticle sizecoefficientZetaName±SD (nm)(PDI)PotentialPaclitaxel cholesterol114.4 ± 5.180.27 ± 0.004−8.7 ± 2.128liposome (PTX-Cho-Lipo)Paclitaxel Rg3 liposome77.8 ± 6.410.17 ± 0.015−4.2 ± 0.777(PTX-Rg3-Gipo) As shown inFIGS.1and2,FIG.2represents a normal distribution. As shown in Table 3, the distribution coefficient of PTX-Rg3-Gipo in the present invention is more optimal than that of PTX-Cho-Lipo, and the particle size of PTX-Rg3-Gipo is also smaller. The results suggest that PTX-Rg3-Gipo is better than PTX-Cho-Lipo in quality. Application Embodiment 4 The Leakage Experiment of PTX-Cho-Lipo and PTX-Rg3-Gipo Freshly prepared PTX-Cho-Lipo and PTX-Rg3-Gipo were filtered using 0.22 micron membrane, and their encapsulation efficiency was determined and considered as a 100%. 3 mL each of the PTX-Cho-Lipo and PTX-Rg3-Gipo solutions were taken out and stored at 4° C. and their encapsulation efficiency were measured daily for 7 days. Plot a graph between the encapsulation efficiency and the time (days). The experimental results are shown inFIG.3and table 4. TABLE 4The encapsulation efficency of paclitaxel in PTX-Cho-Lipoand PTX-Rg3-GipoEncapsulation efficiency, %Time(days)PTX-Cho-LipoPTX-Rg3-Gipo1.00100.00100.002.0062.9094.323.0044.9589.674.0040.6185.855.0036.5783.376.0033.2581.687.0035.8580.32 As shown inFIG.3, there is a sharp drop in the encapsulation efficiency of PTX-Cho-Lipo from the beginning to the third day, however, few changes are observed in the encapsulation efficiency of PTX-Rg3-Gipo within 7 days. As shown in Table 4, under the same conditions, encapsulation efficiency of PTX-Rg3-Gipo in the present invention is higher than that of PTX-Cho-Lipo, which indicates that PTX-Rg3-Gipo is more stable in solution with less leakage. Thus, the quality of PTX-Rg3-Gipo is better than PTX-Cho-Lipo, and Rg3 is better than cholesterol as a liposome membrane material. Application Embodiment 5 Effects of Liposome on Prolonged Circulation Time 30 nude mice (18-22 g) were randomly divided into 5 various groups (6 in each group), administered via mouse tail vein respectively with 0.3 mg/kg Cholesterol-blank liposome loaded with a fluorescent dye DID (DID-Cho-blank), mPEG-DSPE-Cholesterol blank liposome loaded with a fluorescent dye DID (DID-PEG-blank), Rg5-blank liposome loaded with loaded with a fluorescent dye DID (DID-Rg5-blank), Rg3-blank liposome loaded with a fluorescent dye DID (DID-Rg3-blank) and Rh2-blank liposome loaded with a fluorescent dye DID (DID-Rh2-blank). 0.2 mL blood samples were collected into heparinized centrifugal tubes via mice facial vein respectively after 2 min, 5 min, 15 min, 30 min, 1 hour, 3 hour, 6 hour, 12 hour and 24 hour. The DID fluorescence intensity of the collected blood sample was measured by a microplate reader. The fluorescence intensity of the first sample collected after 2 min was considered as 100% and other fluorescence intensity were calculated based on this value. Data Process and Analysis: The pharmacokinetic parameters of each liposome were calculated using pharmaceutical kinetics software 3p97, including Area under the Concentration-time Curve (AUC), half life of distribution (t1/2α, t1/2β) and half-life of elimination(t1/2γ), etc. The experimental results are listed inFIG.4and Table 5. TABLE 5Characterization of liposome on prolonged ciruculation timeParameterDID-Cho-blankDID-PEG-blankDID-Rg5-blankDID-Rg3-blankDID-Rh2-blankt1/2α/h0.030.0160.0170.670.245t1/2β/h0.7980.9170.471.6031.892t1/2γ/h9.04924.64712.99927.24324.844AUC(0-t)/mg · L · h401.352808.472450.461753.111760.584AUC(0-∞)/mg · L · h455.2271163.13613.035827.905916.252 As shown in Table 5, the values of AUC, half life of distribution (t1/2α, t1/2β) and half-life of elimination (t1/2γ) of DID-Rg3-blank and DID-Rh2-blank liposomes in the present invention are similar to the values of DID-PEG-blank, suggesting that they all have similar prolonged circulation time and similar therapeutic effect. Whereas, the circulation time and therapeutic effects of DID-Rg5-blank is shorter and weaker than DID-PEG-blank, only longer and stronger than the conventional DID-Cho-Blank. Application Enbodiment 6 In Vivo Target Specificity Assay BALB/C-nu/nu mice bearing tumors in uniform size of 100 mm3at right forelimbs without hemorrhagic necrosis, were intravenously injected via tail vein with liposomes in the present invention carrying 10% of near-infrared fluorescent probe (IR783) respectively (hereinafter named as the experimental group), which was obtained by encapsulating near-infrared fluorescent probe (IR783) into the present ginsenoside blank liposome, see embodiment 10 for details. A conventional blank lipsome caning near-infrared fluorescent probe (IR783) was hereinafter named as the control group which was obtained by encapsulating near-infrared fluorescent probe (IR783) into the blank liposome. The in vivo distributions of IR783 fluorescence were were recorded by in-vivo animal imaging system at the following time points, 2 h, 4 h, 8 h, 12 h and 24 h hour after administration, seeFIG.5. FIG.5-A1-A5are respectively in vivo distribution of IR783 fluorescence in the control group recorded at 2nd, 4th, 8th, 12thand 24thhour by in-vivo animal imaging system.FIG.5-S is a fluorescence ruler, wherein the color is red, yellow, green and blue in sequence, indicating the fluorescence intensity, from the strongest to the weakest.FIG.5-B1-B5,FIG.5-C1-C5andFIG.5-D1-D5are respectively the in vivo fluorescence distribution in the experimental group recorded at 2nd, 4th, 8th, 12thand 24thhour by in-vivo animal imaging system.FIG.5-B1-B5are respectively the fluorescence distribution of the Rg5-blank group;FIG.5-C1-C5are respectively the fluorescence distribution of the Rh2-blank group;FIG.5-D1-D5are respectively the fluorescence distribution of the Rg3-blank group. As shown inFIG.5, the right forelimbs of the mice in the control group had no fluorescence, while the right forelimbs of the mice in the experimental groups have intensive fluorescence, indicating that ginsenoside blank liposomes can target tumor cells specifically. FIG.6is the in-vitro fluorescence distribution of IR783 after tumor removal imaged by in-vivo animal imaging system.FIG.6-A is control group, andFIGS.6-B,6-C and6-D are the experimental groups. After the in-vivo imaging, the tumors in the experimental group and control group are taken out and imaged in vitro.FIG.6-S is a fluorescence ruler, wherein the color shows the relative fluorescence intensity, from strongest to weakest in a sequence of red, yellow, green and blue.FIG.6-B,FIG.6-C andFIG.6-D respectively show the fluorescence intensity of Rg5-Gipo, Rg3-Gipo and Rh2-Gipo groups, suggesting that ginsenoside blank liposomes have very high specificity toward tumor cells. FIG.7is the comparison results between the fluorescence intensity of the control group and the experimental groups. It shows that the fluorescence intensity of Rg5-Gipo, Rg3-Gipo and Rh2-Gipo are significantly higher than that of the control group. Rg3-Gipo and Rh2-Gipo exhibit a significantly higher specificity to target than Rg5-Gipo group in BGC-823 human gastric cancer. In summary, the results suggest that Rg5-blank, Rg3-blank, and Rh2-blank have significantly higher specificity to target than the Cho-blank liposome. Moreover, Rg3-blank and Rh2-blank show a higher targeting specificity than Rg5-blank. Application Embodiment 7 In Vivo and In Vitro Pharmacological Efficacy Assay 1. In Vitro Drug Efficacy Assay To test the drug efficacy in vitro, a total of 8 various concentrations were set up as shown in Table 6 andFIG.8.FIG.8shows the cell survival rate of human breast cancer cell line (4T1) with addition of Rh2 group, Rh2-blank group, PTX group, PTX-Cho-Lipo group and PTX-Rh2-Gipo group respectively. TABLE 6Concentration and viability of human breast cancer cells (4T1) with addition of Rh2group, Rh2-blank group, PTX group, PTX-Cho-lipo group and PTX-Rh2-Gipo groupC(μM)Cell ViabilityPTX-PTX-PTX-PTX-Rh2-Cho-Rh2-Rh2-Cho-Rh2-Rh2blankPTXLipoGipoRh2blankPTXLipoGipo8.000008.000002.000002.000002.00000100.73100.2552.0036.5223.072.666672.666670.666670.666670.66667104.2593.0848.5436.5024.470.888890.888890.222220.222220.2222296.0495.0348.4839.0026.360.296300.296300.074070.074070.0740798.0797.7249.2552.9238.400.098770.098770.024690.024690.0246999.9095.4355.8371.9544.870.032920.032920.008230.008230.0082394.4791.7759.19101.0757.240.010970.010970.002740.002740.0027492.8593.5594.7096.1271.480.003660.003660.000910.000910.00091104.0497.43106.72101.0377.96 As shown in Table 6 andFIG.8, free Rh2 and Rh2-blank groups show low activity in vitro against human breast cancer cells (4T1). With low concentration, the cell viability of PTX-Cho-lipo group is lower than PTX group. While no matter the concentration is high or low, the cell viability of PTX-Rh2-Gipo group is much higher than the PTX group. 2. In Vivo Drug Efficacy Assay To evaluate the drug efficacy in vivo, 45 subcutaneous tumor-bearing nude mice were randomized into 5 treatment groups (9 in each group) and intravenously injected with PBS solution (control group), ginsenoside Rh2 (Rh2 group), ginsenoside Rh2 blank liposome (Rh2-Blank group), conventional paclitaxel cholesterol liposome (PTX-Cho-Lipo group) and ginsenoside Rh2 paclitaxel liposome (PTX-Rh2-Gipo group) via tail vein at a dose of 30 mg/kg. The changes of body weights of mice in each group were recorded every 2 days, and the longest diameter and the shortest diameter of tumors were measured with vernier calipers. The tumor volume was calculated by the following formula: V=(dmax×dmin2)/2, wherein dmin and dmax are respectively the shortest diameter and the longest diameter (mm) of the tumor; a relative tumor volume (RTV) was calculated according to the measurement results, by the formula: RTV=TVn/TV0, wherein TVn is the volume of the tumor measured every 2 days, TV0is the volume of the tumor measured at day zero (the administration day). TABLE 7Antitumor effects of control group, Rh2 group, Rh2-blank group,PTX-Cho-lipo group and PTX-Rh2-Gipo group in human breastcancer cell 4T14T1Relative tumor volumetime(d)ControlRh2Rh2-blankPTX-Cho-LipoPTX-Rh2-Gipo0100.00100.00100.00100.00100.003273.99200.94214.73199.0195.236249.60316.69193.95229.01166.899290.21276.04273.64289.80162.8512555.41400.20310.41317.20168.0215507.64473.53403.28435.89167.8818700.78510.20400.30449.06178.5521965.30898.52603.59511.90245.27 As shown in Table 7 andFIG.9, after the same period of time, the volume of tumor in control group and Rh2 group are the maximum while in the PTX-Rh2-Gipo group is the minimum, followed by PTX-Cho-lipo group and Rh2-blank group. Results suggest that PTX-Rh2-Gipo group has better antitumor effects. 3. In Vitro Cytotoxicity Studies The in vitro cytotoxicity was evaluated using human breast cancer cell line (4T1). The cell survival rate of human breast cancer cell line (4T1) with addition of DTX group, DTX-Cho-Lipo group, DTX-Rg3-Gipo group and Nanoxel-PM group at various concentration were shown in Table 8 andFIG.10. TABLE 8The viability of human breast cancer cells (4T1) with addition of DTXgroup, DTX-Cho-Lipo group, DTX-Rg3-Gipo group and Nanoxel-PMgroup at various concentrationConcentrationCell viability(%)(μg/ml)DTXDTX-Cho-LipoDTX-Rg3-GipoNanoxel-PM351.8539.8540.6740.540.648.8940.9643.9844.780.1246.2645.5642.6037.620.02449.8655.4548.5244.520.004850.9454.0848.9451.030.0009661.6559.3550.3361.960.0001972.4869.8152.1176.163.84E−0583.5576.6265.0084.217.68E−0686.6679.1581.5988.55 As shown in Table 8 andFIG.10, after the same period of time, the overall viability of human breast cancer cells 4T1 with addition of DTX-Rg3-Gipo group is significantly higher than DTX-Cho-Lipo group, especially in lower concentrations. 4. In Vivo Drug Efficacy Assay To evaluate the drug efficacy in vivo, 45 subcutaneous tumor-bearing nude mice were were randomerized into 5 groups (9 in each group), and intravenously injected with PBS solution (Control group,), Taxotere, Nanoxel-PM, DTX-Rg5-Gipo and DTX-Rg3-Gipovia tail vein at a dose of 10 mg·kg−1. The changes in mice body weights in each group were recorded every 2 days, and the longest diameter and the shortest diameter of tumors were measured with vernier calipers. The tumor volume is calculated by the following formula: V=(dmax×dmin2)/2, wherein dmin and dmax are respectively the shortest diameter and the longest diameter (mm) of the tumor; a relative tumor volume (RTV) is calculated according to the measurement results by the formula: RTV=TVn/TV0, wherein TVn is the volume of the tumor measured every 2 days, TV0is the volume of the tumor measured at day zero (the administration day). TABLE 9Antitumor effect of Control group, Taxotere group, Nanoxel-PM group,DTX-Rg5-Gipo group and DTX-Rg3-Gipo group in human breastcancer cell 4T1Relative tumor volume4T1Nanoxel-DTX-Rg5-DTX-Rg3-time(d)ControlTaxoterePMGipoGipo0100.00100.00100.00100.00100.003273.99206.85192.51168.40115.346249.60254.61140.99141.6784.929290.21198.66172.59203.55125.3312555.41224.67134.94155.1189.0915507.64231.03181.05150.3786.6518700.78361.50175.55197.9765.9921764.79322.15184.46151.5686.11 As shown in Table 9 andFIG.11, after the same period of time, the volume of tumor in the PBS group is the maximum while in the DTX-Rg3-Gipo group is the minimum, followed by the DTX-Rg5-Gipo group and Nanoxel-PM group that are basically equivalent. The results suggest that DTX-Rg3-Gipo group has better anti-tumor activity. Application Embodiment 8 In Vivo and In Vitra Pharmacological Efficacy Assay 8.1. In Vitro Drug Efficiency Assay A total of 10 different concentrations of each sample were set up as shown in Table 10. The survival rate of rat glioma C6 cells with addition of Rg3 group, Rg3-blank group, PTX group, PTX+Rg3 group, PTX-Cho-Lipo group and PTX-Rg3-Gipo group at various concentrations respectively are listed in Table 11 andFIG.12. TABLE 10Concentrations of Rg3 group, Rg3-blank group, PTX group, PTX +Rg3 group, PTX-Cho-Lipo group and PTX-Rg3-Gipo group used toagainst rat glioma cells (C6)C (μg/ml)Rg3Rg3-blankPTXPTX + Rg3PTX-Cho-LipoPTX-Rg3-Gipo2020101010106.676.673.3333.3333.3333.3332.222.221.1111.1111.1111.1110.740.740.3700.3700.3700.3700.250.250.12350.12350.12350.12350.080.080.04120.04120.04120.04120.030.030.01370.01370.01370.01370.010.010.00460.00460.00460.00460.0030.0030.00150.00150.00150.00150.0010.0010.00050.00050.00050.0005 TABLE 11The viability of rat C6 glioma cells with addition of Rg3, Rg3-blank,PTX, PTX + Rg3, PTX-Cho-Lipo and PTX-Rg3-GipoCell ViabilityPTX-Cho-PTX-Rg3-Rg3Rg3-blankPTXPTX + Rg3LipoGipo89.0217146.400932939.5882931.7168130.9853222.9108795.0217165.1012443.2100633.9180432.7774523.962296.1601578.8614443.8946434.3598137.7099224.976792.82284.0347745.9781235.9669438.4850325.5958995.4269286.1549949.9069937.2770245.1321230.1010394.2305888.3388160.4881345.998450.0880833.7673192.0308788.3388165.3640255.3269159.1544337.2054792.4167989.6109473.286762.6087372.8009450.5132898.8229691.3283279.4925266.4475685.554958.3184100100100100100100 As shown in Table 11 andFIG.12, PTX-Rg3-Gipo group show better cell activity than PTX-Cho-Lipo group and PTX+Rg3 group. The results suggest that the cell activity of PTX-Rg3-Gipo group has been greatly improved. 8.2. Survival Curve and Median Survival Day A total of 63 subcutaneous tumor-bearing nude mice were randomized into 7 groups (9 in each group), and intravenously injected with PBS solution (Control group,), Rg3, Rg3-blank, PTX, PTX+Rg3, PTX-Cho-Lipo and PTX-Rg3-Gipo via tail vein at a dose of 10 mg·kg−1. From the 12thday after injection, the numbers of survived nude mice were recorded daily until all nude mice die. Survival curves of nude mice in each group were plotted by GraphPad Prism-5 software, and median survival time was calculated. TABLE 12The number of survived mice in each group at corresponding time against in-situgliomaTime(d)PBSPTXRg3PTX + Rg3PTX-Cho-LipoRg3-blankPTX-Rg3-Gipo12910101010101014910101010101016899910101018689910101020578799102246768910243676899263565799282453678300443558320443558340331447360131337380120337400010327420000327440000326460000326480000226500000225520000215540000115560000115580000013600000001620000000 TABLE 13The median survival days in each group at corresponding time against in-situ gliomaPTX-PTX-PTX +Cho-Rg3-GroupsPBSPTXRg3Rg3LipRg3GipoMedian21272927353254survial(day) As shown in Table 12, Table 13 andFIG.13, the median survival time of PTX-Rg3-Gipo group is significantly longer than those of PTX-Cho-Lipo group and PTX+Rg3 group. Application Embodiment 9 In Vivo and In Vivo Pharmacological Efficacy Assay 1. In Vitro Cell Viability Assay A total of 9 different concentrations were set up as shown in Table 12. The survival rate of human gastric cancer cells (BGC-823) with addition of Rg5 group, Rg3 group, Rh2 group, Rg5-blank group, Rg3-blank group, Rh2-blank group, PTX group, PTX-Cho-Lipo group, PTX-Rg5-Gipo group, PTX-Rg3-Gipo group and PTX-Rh2-Gipo group at various concentrations are shown inFIG.12and Table 14 respectively. TABLE 14Concentrations of Rg5 group, Rg3 group, Rh2 group, Rg5-blank group, Rg3-blankgroup, Rh2-blank group, PTX group, PTX-Cho-Lipo group, PTX-Rg5-Gipo group, PTX-Rg3-Gipo group and PTX-Rh2-Gipo group used in human gastric cancer cells (BGC-823)C(μg/mL)PTXPTX-PTX-PTX-Rg5-Rg3-Rh2-Cho-Rg5-Rg3-Rh2-Rg5Rg3Rh2blankblankblankPTXLipoGipoGipoGipo0.01090.01090.01090.01090.01090.01090.00270.00270.00270.00270.00270.03290.03290.03290.03290.03290.03290.00820.00820.00820.00820.00820.09870.09870.09870.09870.09870.09870.02460.02460.02460.02460.02460.29620.29620.29620.29620.29620.29620.07400.07400.07400.07400.07400.88880.88880.88880.88880.88880.88880.22220.22220.22220.22220.22222.66662.66662.66662.66662.66662.66660.66660.66660.66660.66660.66668888882222224242424242466666 TABLE 15Cell viability of human gastric cancer cells (BGC-823) with addition of Rg5 group,Rg3 group, Rh2 group, Rg5-blank group, Rg3-blank group, Rh2-group, PTX group,PTX-Cho-Lipo group, PTX-Rg5-Gipo group, PTX-Rg3-Gipo group and PTX-Rh2-Gipo groupCell ViabilityRg5blankRg3blankRh2blankPTXPTX-Cho-LipoPTX-Rg5-GipoPTX-Rg3-GipoPTX-Rh2-Gipo102.7100.6101.291.9100.582.694.885.4101.2101.4103.780.585.552.687.668.6103.398.0100.760.953.938.869.836.4100.087.6102.050.343.635.335.532.1100.477.9100.841.638.434.729.329.2104.472.789.342.736.034.028.928.799.864.675.638.034.933.325.528.394.858.865.633.233.123.524.224.392.753.557.732.232.820.76.817.6 As shown in Table 15 andFIG.14, the cell viability in PTX-Rg5-Gipo group, PTX-Rh2-Gipo group and PTX-Rg3-Gipo group are the best, followed by PTX-Cho-Lipo group and PTX group. 2. In vivo pharmacological efficacy assay: A total of 72 subcutaneous tumor-bearing nude mice were randomerized into 8 groups (9 in each group), and intravenously injected with PBS solution (Control group,), Rg3, Rg3-blank, PTX-Cho-Lipo, Abraxane, PTX-Rg5-Gipo, PTX-Rg3-Gipo and PTX-Rh2-Gipo, via tail vein at a dose of 10 mg·kg1. The changes of mice body weights in each group were recorded every 2 days, and the longest diameter and the shortest diameter of tumors were measured with vernier calipers. The tumor volume is calculated by the following formula: V=(dmax×dmin2)/2, wherein dmin and dmax are respectively the shortest diameter and the longest diameter (mm) of the tumor; a relative tumor volume (RTV) is calculated according to the measurement results by the formula: RTV=TVn/TV0, wherein TVn is the volume of the tumor measured every 2 days, TV0is the volume of the tumor measured at day zero (the administration day). Experimental results are listed in Table 16 andFIG.15. TABLE 16Drug efficacy of Control group, Rg3 group, Rg3-blank group,PTX-Cho-Lipo group,Abraxane group, PTX-Rg5-Gipo group, PTX-Rg3-Gipo group andPTX-Rh2-Gipo groupagainst human gastric cancer cell (BGC-823)Relative tumor volumeBGC-PTX-PTX-PTX-PTX-823Cho-Rg5-Rg3-Rh2-time(d)PBSRg3Rg3-blankLipoAbraxaneGipoGipoGipo1164.67166.88168.24165.27164.67165.77163.63163.503215.94267.38275.58275.58225.78187.97239.71197.105322.94317.96319.38381.27316.77165.73161.94134.837469.00417.96472.85553.90362.70134.84119.2979.999777.24717.96657.85782.11401.47115.0178.5653.36111273.611340.69817.791044.41541.84119.7231.5022.67131747.541636.89901.411313.14595.41171.0430.0423.87152039.452034.431107.421410.32736.65186.4715.406.83172039.452034.431118.431515.96840.69193.3218.894.05192039.452034.431207.421826.551187.35439.5219.961.71212039.452034.431321.361982.041215.19535.774.142.00232039.452034.431525.742061.151351.30560.6913.963.24 As shown in Table 16 andFIG.15, after the same period of time, the tumor volume in Control group is the maximum while in the PTX-Rg3-Gipo group and the PTX-Rh2-Gipo group are the minimum, followed by the PTX-Rg5-Gipo group, the Abraxane group and PTX-Cho-Lipo group. The data suggest that PTX-Rg3-Gipo group and PTX-Rh2-Gipo group have significant better anti-tumor activity. It is to be understood that the foregoing description of two preferred embodiments is intended to be purely illustrative of the principles of the invention, rather than exhaustive thereof, and that changes and variations will be apparent to those skilled in the art, and that the present invention is not intended to be limited other than expressly set forth in the following claims.
61,439
11858959
DETAILED DESCRIPTION OF THE INVENTION Glycyrrhizin (or glycyrrhizic acid or glycyrrhizinic acid) is extract of the plant called Glycyrrhiza which is derived from the ancient Greek term ‘glykos’, meaning sweet, and ‘rhiza’, meaning root. Glycyrrhiza was indulged upon by many prophets and pharaohs. Licorice extract has been utilized in the battlefields and the desert where soldiers and travelers drank it to suppress their thirst sensation on long marches. Glycyrrhetic acid, the active metabolite in licorice, inhibits HSD2 with a resultant cortisol-induced mineralocorticoid effect and the tendency towards the reduction of potassium levels. While glycyrrhetic acid lowers potassium levels, it is associated with abnormal heart rhythms, hypertension, edema, lethargy, congestive heart failure, hypokalemia and rhabdomyolysis. Accordingly, it would be desirable to provide a compound that promotes potassium excretion in patients suffering from hyperkalemia like glycyrrhetinic acid without the undesirable side effects. The present invention provides a compound of formula I or a salt thereof: wherein,X is a bond, —O—, —C(O)—, —N(Rx)—, —C(O)N(Rx)—, —N(Rx)—C(O)—, —S(O)n—N(Rx)— or —N(Rx)—S(O)n—;L is a bond, alkylene wherein one or more non-adjacent methylene groups of said alkylene are replaced with —O—; divalent aryl or divalent heteroaryl; or L is alkylene-Y-alkylene wherein Y is O, NRx, S, SO, SO2or a divalent heterocycle; wherein said alkylene groups are optionally substituted with OH, —C(O)O—R1, alkyl or alkyl substituted with OH or —C(O)O—R1; and wherein a carbon of said alkylene groups and Rxoptionally together form a heterocycle; provided that when X is other than a bond, then L is other than a bond;W is O or S;Q is a bond or alkylene;R1is H, alkyl, a carbocycle or a heterocycle wherein said alkyl, carbocycle and heterocycle are each optionally substituted with halogen, OH, amino, oxo, carboxy, acyloxy, alkoxycarbonyl, alkoxyacyloxy, alkoxycarbonyloxy, aminocarbonyl, a carbocycle optionally substituted with alkyl, haloalkyl, oxo, amino and halogen and a heterocycle optionally substituted with alkyl, oxo, amino and halogen; and a carbocycle or heterocycle optionally substituted with alkyl, haloalkyl, oxo, amino and halogen;V is —C(O)O—, —C(O)O—(CHR5)—O—C(O)—, —C(O)O—(CHR5)—O—C(O)—O—, —C(O)N(R5)—, —C(O)N(R5)O—, —NH—C(O)—N(R5)— or NH—S(O)n—;R2is H or R1;R3is absent or alkyl;R4is absent, H, OH, ═O, —R6, —O—R6, —C(O)O—R6, —O—C(O)—R6, —O—C(O)—O—R6, —O—C(O)—NR5R6, —NR5R6, —NR5—C(O)—R6, —NR5—C(O)—O—R6, —NR5—SO2—R6, ═N—O—R5;R5is H or alkyl;R6is H, alkyl, a carbocycle, a heterocycle wherein said alkyl, carbocycle and heterocycle are optionally substituted with halogen, OH, SH, alkylthio, —S(O)-alkyl, —SO2-alkyl, amino, —NHC(O)-alkyl, oxo, alkyl, carboxyl, acyl, acyloxy, alkoxy, alkoxycarbonyl, a carbocycle optionally substituted with halogen, OH, amino or alkyl, or a heterocycle optionally substituted with halogen, OH, amino or alkyl; and wherein one or more non-adjacent methylene groups in each of said alkyl groups of R6are optionally replaced with —O— or —S—;Rxis H, —C(O)O—R1, or alkyl optionally substituted with —C(O)O—R1; and n is 1 or 2. The dashed lines between the 2- and 3-positions of the fused ring system indicate alternatively a single or double bond. The dashed lines converging inside the fused ring system indicate that R4, when present, and the group comprising —X-L-C(O)-Q- (and a dioxalone ring) may be attached alternatively at the 3-position (e.g. as in Formula Ie) or 4-position (e.g. as in Formula Ic) of the fused ring system. Fused ring numbering convention is shown inFIG.3. In a particular embodiment, compounds of the invention have the group comprising the dioxolone ring pending from the 4-position of the fused ring system. In an embodiment, following administration to a subject of a compound of the invention, the ester moiety is metabolized in plasma or liver to a less active acid form. In another embodiment, the compound of the invention has equal or greater HSD2 inhibitory activity than glycyrrhetinic acid. In another embodiment, the compound of the invention has greater HSD2 inhibitory activity than glycyrrhetinic acid. “Acyl” means a carbonyl containing substituent represented by the formula —C(O)—R in which R is H, alkyl, a carbocycle, a heterocycle, carbocycle-substituted alkyl or heterocycle-substituted alkyl, wherein the alkyl, alkoxy, carbocycle and heterocycle are as defined herein. Acyl groups include alkanoyl (e.g., acetyl), aroyl (e.g., benzoyl), and heteroaroyl. “Alkyl” means a branched or unbranched, saturated or unsaturated (i.e. alkenyl, alkynyl) aliphatic hydrocarbon group, having up to 12 carbon atoms unless otherwise specified. When used as part of another term, for example, “alkylamino”, “cycloalkyl”, “alkylene” etc., the alkyl portion may be a saturated hydrocarbon chain, however also includes unsaturated hydrocarbon carbon chains such as “alkenylamino” and “alkynylamino. Examples of particular alkyl groups are methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2,2-dimethylpropyl, n-hexyl, 2-methylpentyl, 2,2-dimethylbutyl, n-heptyl, 3-heptyl, 2-methylhexyl, and the like. The terms “lower alkyl” “C1-C4alkyl” and “alkyl of 1 to 4 carbon atoms” are synonymous and used interchangeably to mean methyl, ethyl, 1-propyl, isopropyl, cyclopropyl, 1-butyl, sec-butyl or t-butyl. Unless specified, substituted, alkyl groups may contain, for example, one, two, three or four substituents, which may be the same or different. Examples of substituents are, unless otherwise defined, halogen, amino, hydroxyl, protected hydroxyl, mercapto, carboxy, alkoxy, nitro, cyano, amidino, guanidino, urea, sulfonyl, sulfinyl, aminosulfonyl, alkylsulfonylamino, arylsulfonylamino, aminocarbonyl, acylamino, alkoxy, acyl, acyloxy, a carbocycle, and a heterocycle. Examples of the above substituted alkyl groups include, but are not limited to; cyanomethyl, nitromethyl, hydroxymethyl, trityloxymethyl, propionyloxymethyl, aminomethyl, carboxymethyl, carboxyethyl, carboxypropyl, alkyloxycarbonylmethyl, allyloxycarbonylaminomethyl, carbamoyloxymethyl, methoxymethyl, ethoxymethyl, t-butoxymethyl, acetoxymethyl, chloromethyl, bromomethyl, iodomethyl, trifluoromethyl, 6-hydroxyhexyl, 2,4-dichloro(n-butyl), 2-amino(isopropyl), 2-carbamoyloxyethyl and the like. The alkyl group may also be substituted with a carbocycle group. Examples include cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, and cyclohexylmethyl groups, as well as the corresponding -ethyl, -propyl, -butyl, -pentyl, -hexyl groups, etc. Substituted alkyls include substituted methyls, e.g., a methyl group substituted by the same substituents as the “substituted Cn-Cmalkyl” group. Examples of the substituted methyl group include groups such as hydroxymethyl, protected hydroxymethyl (e.g., tetrahydropyranyloxymethyl), acetoxymethyl, carbamoyloxymethyl, trifluoromethyl, chloromethyl, carboxymethyl, bromomethyl and iodomethyl. In an embodiment, alkyl is saturated. In an embodiment, alkyl is unsaturated. In an embodiment, alkyl is partially unsaturated. “Amidine” means the group —C(NH)—NHR in which R is H, alkyl, a carbocycle, a heterocycle, carbocycle-substituted alkyl or heterocycle-substituted alkyl wherein the alkyl, alkoxy, carbocycle and heterocycle are as defined herein. A particular amidine is the group —NH—C(NH)—NH2. “Amino” means primary (i.e. —NH2), secondary (i.e. —NRH) and tertiary (i.e. —NRR) amines in which R is H, alkyl, a carbocycle, a heterocycle, carbocycle-substituted alkyl or heterocycle-substituted alkyl wherein the alkyl, alkoxy, carbocycle and heterocycle are as defined herein. Particular secondary and tertiary amines are alkylamine, dialkylamine, arylamine, diarylamine, aralkylamine and diaralkylamine, wherein the alkyl is as herein defined and optionally substituted. Particular secondary and tertiary amines are methylamine, ethylamine, propylamine, isopropylamine, phenylamine, benzylamine dimethylamine, diethylamine, dipropylamine and diisopropylamine. “Amino-protecting group” as used herein refers to a derivative of the groups commonly employed to block or protect an amino group while reactions are carried out on other functional groups on the compound. Examples of such protecting groups include carbamates, amides, alkyl and aryl groups, imines, as well as many N-heteroatom derivatives which can be removed to regenerate the desired amine group. Suitable amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, t-butyloxy carbonyl (“Boc”), benzyloxy carbonyl (“CBz”) and 9-fluorenylmethyleneoxycarbonyl (“Fmoc”). Further examples of these groups are found in Wuts.Greene's Protective Groups in Organic Synthesis.5th ed. New York: John Wiley & Sons, Inc., 2014. The term “protected amino” refers to an amino group substituted with one of the above amino-protecting groups. “Aryl” when used alone or as part of another term means a carbocyclic aromatic group whether or not fused having the number of carbon atoms designated or if no number is designated, up to 14 carbon atoms. Particular aryl groups are phenyl, naphthyl, biphenyl, phenanthrenyl, naphthacenyl, and the like (see e.g., Dean, J. A., ed.Lange's Handbook of Chemistry,13th ed. New York: McGraw-Hill, 1985, Table 7-2). A particular aryl is phenyl. Substituted phenyl or substituted aryl means a phenyl group or aryl group substituted with one, two, three, four or five substituents, for example 1-2, 1-3 or 1-4 substituents chosen, unless otherwise specified, from halogen (F, Cl, Br, I), hydroxy, protected hydroxy, cyano, nitro, alkyl (for example C1-C6alkyl), alkoxy (for example C1-C6alkoxy), benzyloxy, carboxy, protected carboxy, carboxymethyl, protected carboxymethyl, hydroxymethyl, protected hydroxymethyl, aminomethyl, protected aminomethyl, trifluoromethyl, alkylsulfonylamino, alkylsulfonylaminoalkyl, arylsulfonylamino, arylsulonylaminoalkyl, heterocyclylsulfonylamino, heterocyclylsulfonylaminoalkyl, heterocyclyl, aryl, or other groups specified. One or more methyne (CH) and/or methylene (CH2) groups in these substituents may in turn be substituted with a similar group as those denoted above. Examples of the term “substituted phenyl” includes but is not limited to a mono- or di(halo)phenyl group, such as 2-chlorophenyl, 2-bromophenyl, 4-chlorophenyl, 2,6-dichlorophenyl, 2,5-dichlorophenyl, 3,4-dichlorophenyl, 3-chlorophenyl, 3-bromophenyl, 4-bromophenyl, 3,4-dibromophenyl, 3-chloro-4-fluorophenyl, 2-fluorophenyl and the like; a mono- or di(hydroxy)phenyl group such as 4-hydroxyphenyl, 3-hydroxyphenyl, 2,4-dihydroxyphenyl, the protected-hydroxy derivatives thereof and the like; a nitrophenyl group such as 3- or 4-nitrophenyl; a cyanophenyl group, for example, 4-cyanophenyl; a mono- or di(lower alkyl)phenyl group such as 4-methylphenyl, 2,4-dimethylphenyl, 2-methylphenyl, 4-(isopropyl)phenyl, 4-ethylphenyl, 3-(n-propyl)phenyl and the like; a mono or di(alkoxy)phenyl group, for example, 3,4-dimethoxyphenyl, 3-methoxy-4-benzyloxyphenyl, 3-methoxy-4-(1-chloromethyl)benzyloxy-phenyl, 3-ethoxyphenyl, 4-(isopropoxy)phenyl, 4-(t-butoxy)phenyl, 3-ethoxy-4-methoxyphenyl and the like; 3- or 4-trifluoromethylphenyl; a mono- or dicarboxyphenyl or (protected carboxy)phenyl group such as 4-carboxyphenyl; a mono- or di(hydroxymethyl)phenyl or (protected hydroxymethyl)phenyl such as 3-(protected hydroxymethyl)phenyl or 3,4-di(hydroxymethyl)phenyl; a mono- or di(aminomethyl)phenyl or (protected aminomethyl)phenyl such as 2-(aminomethyl)phenyl or 2,4-(protected aminomethyl)phenyl; a mono- or di(N-(methylsulfonylamino))phenyl such as 3-(N-methylsulfonylamino))phenyl; disubstituted phenyl groups such as 3-methyl-4-hydroxyphenyl, 3-chloro-4-hydroxyphenyl, 2-methoxy-4-bromophenyl, 4-ethyl-2-hydroxyphenyl, 3-hydroxy-4-nitrophenyl, 2-hydroxy-4-chlorophenyl; trisubstituted phenyl groups such as 3-methoxy-4-benzyloxy-6-methyl sulfonylamino, 3-m ethoxy-4-benzyl oxy-6-phenyl sulfonylamino; and tetrasubstituted phenyl groups such as 3-methoxy-4-benzyloxy-5-methyl-6-phenyl sulfonylamino. Particular substituted phenyl groups include the 2-chlorophenyl, 2-aminophenyl, 2-bromophenyl, 3-methoxyphenyl, 3-ethoxy-phenyl, 4-benzyloxyphenyl, 4-methoxyphenyl, 3-ethoxy-4-benzyloxyphenyl, 3,4-diethoxyphenyl, 3-methoxy-4-benzyloxyphenyl, 3-methoxy-4-(1-chloromethyl)benzyloxy-phenyl, 3-methoxy-4-(1-chloromethyl)benzyloxy-6-methyl sulfonyl aminophenyl groups. Fused aryl rings may also be substituted with any, for example 1, 2 or 3, of the substituents specified herein in the same manner as substituted alkyl groups. “Carbocyclyl”, “carbocyclic”, “carbocycle” and “carbocyclo” alone and when used as a moiety in a complex group such as a carbocycloalkyl group, refer to a mono-, bi-, or tricyclic aliphatic ring having 3 to 14 carbon atoms, for example 3 to 7 carbon atoms or 3 to 6 carbon atoms, which may be saturated or unsaturated, aromatic or non-aromatic. Particular saturated carbocyclic groups are cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl groups. A particular saturated carbocycle is cyclopropyl. Another particular saturated carbocycle is cyclohexyl. Particular unsaturated carbocycles are aromatic e.g. aryl groups as previously defined, for example phenyl. The terms “substituted carbocyclyl”, “carbocycle” and “carbocyclo” mean these groups substituted by the same substituents as the “substituted alkyl” group. “Carboxy-protecting group” as used herein refers to one of the ester derivatives of the carboxylic acid group commonly employed to block or protect the carboxylic acid group while reactions are carried out on other functional groups on the compound. Examples of such carboxylic acid protecting groups include 4-nitrobenzyl, 4-methoxybenzyl, 3,4-dimethoxybenzyl, 2,4-dimethoxybenzyl, 2,4,6-trimethoxybenzyl, 2,4,6-trimethylbenzyl, pentamethylbenzyl, 3,4-methylenedioxybenzyl, benzhydryl, 4,4′-dimethoxybenzhydryl, 2,2′,4,4′-tetramethoxybenzhydryl, alkyl such as t-butyl or t-amyl, trityl, 4-methoxytrityl, 4,4′-dim ethoxy trityl, 4,4′,4″-trimethoxytrityl, 2-phenylprop-2-yl, trimethylsilyl, t-butyl dimethylsilyl, phenacyl, 2,2,2-trichloroethyl, beta-(trimethylsilyl)ethyl, beta-(di(n-butyl)methylsilyl)ethyl, p-toluenesulfonylethyl, 4-nitrobenzylsulfonylethyl, allyl, cinnamyl, 1-(trimethylsilylmethyl)prop-1-en-3-yl, and like moieties. The species of carboxy-protecting group employed is not critical so long as the derivatized carboxylic acid is stable to the condition of subsequent reaction(s) on other positions of the molecule and can be removed at the appropriate point without disrupting the remainder of the molecule. In particular, it is important not to subject a carboxy-protected molecule to strong nucleophilic bases, such as lithium hydroxide or NaOH, or reductive conditions employing highly activated metal hydrides such as LiAlH4. Such harsh removal conditions are also to be avoided when removing amino-protecting groups and hydroxy-protecting groups, discussed below. Particular carboxylic acid protecting groups are the alkyl (e.g., methyl, ethyl, t-butyl), allyl, benzyl and p-nitrobenzyl groups. Similar carboxy-protecting groups used in the cephalosporin, penicillin and peptide arts can also be used to protect carboxy group substituents. Further examples of these groups are found in Greene, T. W., and P. G. M. Wuts.Protective Groups in Organic Synthesis.2nd ed. New York: John Wiley & Sons, Inc. 1991, Chapter 5; Haslam, E.Protective Groups in Organic Chemistry. New York: Plenum Press 1973, Chapter 5; and Greene, T. W.Protective Groups in Organic Synthesis. New York: John Wiley & Sons, Inc. 1981, Chapter 5. The term “protected carboxy” refers to a carboxy group substituted with one of the above carboxy-protecting groups. “Alkoxycarbonyl” means the group —C(═O)OR in which R is alkyl. A particular group is C1-C6alkoxycarbonyl, wherein the R group is C1-C6alkyl. “Guanidine” means the group —NH—C(NH)—NHR in which R is hydrogen, alkyl, a carbocycle, a heterocycle, carbocycle-substituted alkyl or heterocycle-substituted alkyl, wherein the alkyl, alkoxy, carbocycle and heterocycle are as defined herein. A particular guanidine is the group —NH—C(NH)—NH2. “Hydroxy-protecting group” as used herein refers to a derivative of the hydroxy group commonly employed to block or protect the hydroxy group while reactions are carried out on other functional groups on the compound. Examples of such protecting groups include tetrahydropyranyloxy, benzoyl, acetoxy, carbamoyloxy, benzyl, and silylethers (e.g., TBS, TBDPS) groups. Further examples of these groups are found in Greene, T. W., and P. G. M. Wuts.Protective Groups in Organic Synthesis.2nd ed. New York: John Wiley & Sons, Inc. 1991, Chapters 2-3; Haslam, E.Protective Groups in Organic Chemistry, New York: Plenum Press 1973, Chapter 5; and Greene, T. W.Protective Groups in Organic Synthesis. New York: John Wiley & Sons, Inc. 1981. The term “protected hydroxy” refers to a hydroxy group substituted with one of the above hydroxy-protecting groups. “Heterocyclic group”, “heterocyclic”, “heterocycle”, “heterocyclyl”, or “heterocyclo” alone and when used as a moiety in a complex group such as a heterocycloalkyl group, are used interchangeably and refer to any mono-, bi-, or tricyclic, saturated or unsaturated, aromatic (heteroaryl) or non-aromatic ring having the number of atoms designated, generally from 5 to about 14 ring atoms, where the ring atoms are carbon and at least one heteroatom (nitrogen, sulfur or oxygen), for example 1 to 4 heteroatoms. Heterocyclic groups include four to seven membered cyclic groups containing one, two or three heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Typically, a 5-membered ring has 0 to 2 double bonds and 6- or 7-membered ring has 0 to 3 double bonds. The nitrogen or sulfur heteroatoms may optionally be oxidized (e.g., SO, SO2), and any nitrogen heteroatom may optionally be quaternized. Particular non-aromatic heterocycles are morpholinyl (morpholino), pyrrolidinyl, oxiranyl, oxetanyl, tetrahydrofuranyl, 2,3-dihydrofuranyl, 2H-pyranyl, tetrahydropyranyl, thiiranyl, thietanyl, tetrahydrothietanyl, aziridinyl, azetidinyl, 1-methyl-2-pyrrolyl, piperazinyl and piperidinyl. A “heterocycloalkyl” group is a heterocycle group as defined above covalently bonded to an alkyl group as defined above. Particular 5-membered heterocycles containing a sulfur or oxygen atom and one to three nitrogen atoms are thiazolyl, in particular thiazol-2-yl and thiazol-2-yl N-oxide, thiadiazolyl, in particular 1,3,4-thiadiazol-5-yl and 1,2,4-thiadiazol-5-yl, oxazolyl, for example oxazol-2-yl, and oxadiazolyl, such as 1,3,4-oxadiazol-5-yl, and 1,2,4-oxadiazol-5-yl. Particular 5-membered ring heterocycles containing 2 to 4 nitrogen atoms include imidazolyl, such as imidazol-2-yl; triazolyl, such as 1,3,4-triazol-5-yl; 1,2,3-triazol-5-yl, 1,2,4-triazol-5-yl, and tetrazolyl, such as 1H-tetrazol-5-yl. Particular benzo-fused 5-membered heterocycles are benzoxazol-2-yl, benzthiazol-2-yl and benzimidazol-2-yl. Particular 6-membered heterocycles contain one to three nitrogen atoms and optionally a sulfur or oxygen atom, for example pyridyl, such as pyrid-2-yl, pyrid-3-yl, and pyrid-4-yl; pyrimidyl, such as pyrimid-2-yl and pyrimid-4-yl; triazinyl, such as 1,3,4-triazin-2-yl and 1,3,5-triazin-4-yl; pyridazinyl, in particular pyridazin-3-yl, and pyrazinyl. The pyridine N-oxides and pyridazine N-oxides and the pyridyl, pyrimid-2-yl, pyrimid-4-yl, pyridazinyl and the 1,3,4-triazin-2-yl groups, are a particular group. Substituents for “optionally substituted heterocycles”, and further examples of the 5- and 6-membered ring systems discussed above can be found in W. Druckheimer et al., U.S. Pat. No. 4,278,793. In a particular embodiment, such optionally substituted heterocycle groups are substituted with hydroxyl, alkyl, alkoxy, acyl, halogen, mercapto, oxo, carboxyl, acyl, halo-substituted alkyl, amino, cyano, nitro, amidino and guanidino. “Heteroaryl” alone and when used as a moiety in a complex group such as a heteroaralkyl group, refers to any mono-, bi-, or tricyclic aromatic ring system having the number of atoms designated where at least one ring is a 5-, 6- or 7-membered ring containing from one to four heteroatoms selected from the group nitrogen, oxygen, and sulfur, and in a particular embodiment at least one heteroatom is nitrogen (Lange's Handbook of Chemistry, supra). In one example, the heteroaryl is a five to six membered aromatic ring containing one, two or three heteroatoms selected from nitrogen, oxygen and sulfur. Included in the definition are any bicyclic groups where any of the above heteroaryl rings are fused to a benzene ring. Particular heteroaryls incorporate a nitrogen or oxygen heteroatom. The following ring systems are examples of the heteroaryl (whether substituted or unsubstituted) groups denoted by the term “heteroaryl”: thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, thiatriazolyl, oxatriazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, thiazinyl, oxazinyl, triazinyl, thiadiazinyl, oxadiazinyl, dithiazinyl, dioxazinyl, oxathiazinyl, tetrazinyl, thiatriazinyl, oxatriazinyl, dithiadiazinyl, imidazolinyl, dihydropyrimidyl, tetrahydropyrimidyl, tetrazolo[1,5-b]pyridazinyl and purinyl, as well as benzo-fused derivatives, for example benzoxazolyl, benzofuryl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl, benzoimidazolyl and indolyl. A particular “heteroaryl” may be selected from: 1,3-thiazol-2-yl, 4-(carboxymethyl)-5-methyl-1,3-thiazol-2-yl, 4-(carboxymethyl)-5-methyl-1,3-thiazol-2-yl, 1,2,4-thiadiazol-5-yl, 3-methyl-1,2,4-thiadiazol-5-yl, 1,3,4-triazol-5-yl, 2-methyl-1,3,4-triazol-5-yl, 2-hydroxy-1,3,4-triazol-5-yl, 2-carboxy-4-methyl-1,3,4-triazol-5-yl, 2-carboxy-4-methyl-1,3,4-triazol-5-yl, 1,3-oxazol-2-yl, 1,3,4-oxadiazol-5-yl, 2-methyl-1,3,4-oxadiazol-5-yl, 2-(hydroxymethyl)-1,3,4-oxadiazol-5-yl, 1,2,4-oxadiazol-5-yl, 1,3,4-thiadiazol-5-yl, 2-thiol-1,3,4-thiadiazol-5-yl, 2-(methylthio)-1,3,4-thiadiazol-5-yl, 2-amino-1,3,4-thiadiazol-5-yl, 1H-tetrazol-5-yl, 1-methyl-1H-tetrazol-5-yl, 1-(1-(dimethylamino)eth-2-yl)-1H-tetrazol-5-yl, 1-(carboxymethyl)-1H-tetrazol-5-yl, 1-(carboxymethyl)-1H-tetrazol-5-yl, 1-(methylsulfonic acid)-1H-tetrazol-5-yl, 1-(methylsulfonic acid)-1H-tetrazol-5-yl, 2-methyl-1H-tetrazol-5-yl, 1,2,3-triazol-5-yl, 1-methyl-1,2,3-triazol-5-yl, 2-methyl-1,2,3-triazol-5-yl, 4-methyl-1,2,3-triazol-5-yl, pyrid-2-yl N-oxide, 6-methoxy-2-(n-oxide)-pyridaz-3-yl, 6-hydroxypyridaz-3-yl, 1-methylpyrid-2-yl, 1-methylpyrid-4-yl, 2-hydroxypyrimid-4-yl, 1,4,5,6-tetrahydro-5,6-dioxo-4-methyl-as-triazin-3-yl, 1,4,5,6-tetrahydro-4-(formylmethyl)-5,6-dioxo-as-triazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy-astriazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy-as-triazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy-2-methyl-astriazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy-2-methyl-as-triazin-3-yl, 2,5-dihydro-5-oxo-6-methoxy-2-methyl-as-triazin-3-yl, 2,5-dihydro-5-oxo-as-triazin-3-yl, 2,5-dihydro-5-oxo-2-methyl-as-triazin-3-yl, 2,5-dihydro-5-oxo-2,6-dimethyl-as-triazin-3-yl, tetrazolo[1,5-b]pyridazin-6-yl and 8-aminotetrazolo[1,5-b]-pyridazin-6-yl. An alternative group of “heteroaryl” includes; 4-(carboxymethyl)-5-methyl-1,3-thiazol-2-yl, 4-(carboxymethyl)-5-methyl-1,3-thiazol-2-yl, 1,3,4-triazol-5-yl, 2-methyl-1,3,4-triazol-5-yl, 1H-tetrazol-5-yl, 1-methyl-1H-tetrazol-5-yl, 1-(1-(dimethylamino)eth-2-yl)-1H-tetrazol-5-yl, 1-(carboxymethyl)-1H-tetrazol-5-yl, 1-(carboxymethyl)-1H-tetrazol-5-yl, 1-(methylsulfonic acid)-1H-tetrazol-5-yl, 1-(methylsulfonic acid)-1H-tetrazol-5-yl, 1,2,3-triazol-5-yl, 1,4,5,6-tetrahydro-5,6-dioxo-4-methyl-as-triazin-3-yl, 1,4,5,6-tetrahydro-4-(2-formylmethyl)-5,6-dioxo-as-triazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy-2-methyl-as-triazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy-2-methyl-as-triazin-3-yl, tetrazolo[1,5-b]pyridazin-6-yl, and 8-aminotetrazolo[1,5-b]pyridazin-6-yl. Heteroaryl groups are optionally substituted as described for heterocycles. “Inhibitor” means a compound which reduces or prevents the enzymatic conversion of cortisol to cortisone by HSD2. “Optionally substituted” unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 0, 1, 2, 3 and/or 4) of the substituents listed for that group, as valency allows, in which said substituents may be the same or different. In one embodiment, an optionally substituted group has 1 substituent. In another embodiment, an optionally substituted group has 2 substituents. In another embodiment, an optionally substituted group has 3 substituents. “Pharmaceutically acceptable salts” include both acid and base addition salts. “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid and the like, and organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicyclic acid and the like. “Pharmaceutically acceptable base addition salts” include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Particularly base addition salts are the ammonium, potassium, sodium, calcium and magnesium salts. Salts derived from pharmaceutically acceptable organic nontoxic bases includes salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, trimethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particularly organic non-toxic bases are isopropylamine, diethylamine, ethanolamine, trimethamine, dicyclohexylamine, choline, and caffeine. “Sulfanyl” means —S—R group in which R is alkyl, a carbocycle, a heterocycle, carbocycle-substituted alkyl or heterocycle-substituted alkyl, wherein the alkyl, alkoxy, carbocycle and heterocycle are as defined herein. Particular sulfanyl groups are alkylsulfanyl (i.e., —SO2-alkyl), for example methyl sulfanyl; arylsulfanyl, for example phenylsulfanyl; aralkylsulfanyl, for example benzylsulfanyl. “Sulfinyl” means —SO—R group in which R is hydrogen, alkyl, a carbocycle, a heterocycle, carbocycle-substituted alkyl or heterocycle-substituted alkyl, wherein the alkyl, alkoxy, carbocycle and heterocycle are as defined herein. Particular sulfonyl groups are alkylsulfinyl (i.e., —SO-alkyl), for example methyl sulfinyl; arylsulfinyl, for example phenyl sulfinyl; aralkylsulfinyl, for example benzylsulfinyl. “Sulfonyl” means a —SO2—R group in which R is hydrogen, alkyl, a carbocycle, a heterocycle, carbocycle-substituted alkyl or heterocycle-substituted alkyl, wherein the alkyl, alkoxy, carbocycle and heterocycle are as defined herein. Particular sulfonyl groups are alkylsulfonyl (i.e. —SO2-alkyl), for example methylsulfonyl; arylsulfonyl, for example phenyl sulfonyl; aralkylsulfonyl, for example benzylsulfonyl. The phrase “and salts and solvates thereof” as used herein means that compounds of the inventions may exist in one or a mixture of salts and solvate forms. For example a compound of the invention may be substantially pure in one particular salt or solvate form or else may be mixtures of two or more salt or solvate forms. In particular embodiments of the invention, compounds of Formula I have the structures defined by Formula Ia-Ip: wherein X, L, Q, V R1, R2, R3and R4, are as defined herein. In a particular embodiment, the compounds have the structure according to Formula Ia. In a particular embodiment, the compounds have the structure according to Formula Ib. In a particular embodiment, the compounds have the structure according to Formula Ic. In a particular embodiment, the compounds have the structure according to Formula Id. In a particular embodiment, the compounds have a structure according to Formula Ie. In a particular embodiment, the compounds have a structure according to Formula If. In a particular embodiment, the compounds have a structure according to Formula Ig. In a particular embodiment, the compounds have a structure according to Formula Ih. In a particular embodiment, the compounds have a structure according to Formula Ii. In a particular embodiment, the compounds have a structure according to Formula Ij. In a particular embodiment, the compounds have a structure according to Formula Ik. In a particular embodiment, the compounds have a structure according to Formula II. In a particular embodiment, the compounds have a structure according to Formula Im. In a particular embodiment, the compounds have a structure according to Formula In. In a particular embodiment, the compounds have a structure according to Formula Io. In a particular embodiment, the compounds have a structure according to Formula Ip. In particular embodiments of the invention, compounds of Formula I have the structures defined by Formula Ia′-If: wherein X, L, V, R1, R2, R3, R4and R5are as defined herein. In a particular embodiment, the compounds have the structure according to Formula Ib′. In a particular embodiment, the compounds have the structure according to Formula Ic′. In a particular embodiment, the compounds have the structure according to Formula Id′. In a particular embodiment, the compounds have the structure according to Formula If. In an embodiment, the compound of the invention has a formula of any one of formula I In an embodiment, X is a bond, —O—, —N(Rx)—, —C(O)N(Rx)—, —N(Rx)—C(O)—, —S(O)n—N(Rx)— or —N(Rx)—S(O)n—; wherein Rxis H, —C(O)O—R1, or alkyl optionally substituted with —C(O)O—R1; In an embodiment, X is a bond. In an embodiment, X is —O—. In an embodiment, X is —N(Rx)—. In an embodiment, X is —NH—. In an embodiment, X is —C(O)N(Rx)—. In an embodiment, X is —C(O)NH—. In an embodiment, X is —N(Rx)—C(O)—. In an embodiment, X is —NH—C(O)—. In an embodiment, X is —S(O)n—N(Rx)—. In an embodiment, X is —S(O)—NH—. In an embodiment, X is —S(O)2—NH—. In an embodiment, X is —N(Rx)—S(O)n—. In an embodiment, X is —NH—S(O)—. In an embodiment, X is —NH—S(O)2—. W is O or S. In an embodiment, W is O. In another embodiment, W is S. Q is a bond or alkylene. In an embodiment, Q is a bond. In an embodiment, Q is methylene. In an embodiment, Q is ethylene. V is —C(O)O—, —C(O)O—(CHR5)—O—C(O)—, —C(O)O—(CHR5)—O—C(O)—O—, —C(O)N(R5)—, —C(O)N(R5)O—, —NH—C(O)—N(R5)— or NH—S(O)n—. In an embodiment, V is —C(O)O—. In an embodiment, V is —C(O)O— and R2is H. In an embodiment, V is —C(O)O— and R2is a prodrug group. In an embodiment, V is —C(O)O— and R2is alkyl. In an embodiment, V is —C(O)O— and R2is methyl. In another embodiment V is —C(O)O— and R2is alkyl optionally substituted with oxo, acyloxy, alkoxycarbonyl, alkoxyacyloxy, alkoxycarbonyloxy, a carbocycle optionally substituted with alkyl and oxo, and a heterocycle optionally substituted with alkyl and oxo. In an embodiment, V is —C(O)N(R5)—. In an embodiment, V is —C(O)N(R5)— and both R2and R5are H. In an embodiment, V is —C(O)N(R5)— and R2and R5are independently H and alkyl optionally substituted with OH. In an embodiment, V is —C(O)N(R5)— and R5is H and R2is hydroxy ethyl. In an embodiment, V is —C(O)N(R5)O—. In an embodiment, V is —C(O)N(R5)O— and R2and R5are independently H or alkyl. In an embodiment, V is —C(O)N(R5)O— and R2is methyl and R5is H. In an embodiment, V is —NH—C(O)—N(R5)— and R2and R5are independently H or alkyl. In an embodiment, V is —NH—C(O)—N(R2)— and R2is methyl and R5is H. In an embodiment, V is —NH—C(O)—N(R5)— and both R2and R5are H. In an embodiment, V is NH—S(O)n—. In an embodiment, V is NH—S(O)2—. In an embodiment, V is NH—S(O)2— and R2is alkyl. In an embodiment, V is NH—S(O)2— and R2is methyl. L is a bond, alkylene wherein one or more non-adjacent methylene groups of said alkylene are replaced with —O—; divalent aryl or divalent heteroaryl; or L is alkyl ene-Y-alkyl ene wherein Y is O, NRx, S, SO, SO2or a divalent heterocycle; wherein said alkylene groups are optionally substituted with OH, —C(O)O—R1, alkyl or alkyl substituted with OH or —C(O)O—R1; and wherein a carbon of said alkylene groups and Rxoptionally together form a heterocycle; provided that when X is other than a bond, then L is other than a bond; In an embodiment, L is a bond or alkylene wherein one or more non-adjacent methylene groups of said alkylene are replaced with —O—. In an embodiment, L is a bond. In an embodiment, L is alkylene. In an embodiment, L is alkylene. In an embodiment, L is alkylene in which one or more non-adjacent methylene groups of said alkylene are replaced with —O—. In an embodiment, L is —[(CH2)2—O]1-5—. In an embodiment, L is —(CH2)2—O—. In an embodiment, L is —[(CH2)2—O]2—. In an embodiment, L is —[(CH2)2—O]3—. In an embodiment, L is —[(CH2)2—O]4—. In an embodiment, L is —[(CH2)2—O]5—. In an embodiment, L is alkylene-Y-alkylene wherein Y is O, NRx, S, SO, SO2or a divalent heterocycle; wherein said alkylene groups are optionally substituted with OH, —C(O)O—R1, alkyl or alkyl substituted with OH or —C(O)O—R1; and wherein a carbon of said alkylene groups and Rxoptionally together form a heterocycle; provided that when X is other than a bond, then L is other than a bond. In an embodiment, L is alkylene-Y-alkylene wherein Y is O. In an embodiment, L is alkylene-Y-alkylene wherein Y is NRx. In an embodiment, L is alkylene-Y-alkylene wherein Y is NRxwherein a carbon of said alkylene groups and Rxtogether form a heterocycle. In an embodiment, L is alkylene-Y-alkylene wherein Y is S. In an embodiment, L is alkylene-Y-alkylene wherein Y is SO. In an embodiment, L is alkylene-Y-alkylene wherein Y is SO2. In an embodiment, L is alkylene-Y-alkylene wherein Y is divalent heterocycle. In an embodiment, L is a aryl. In an embodiment, L is phenyl. In an embodiment, L is 1,4-phenylene. In an embodiment, L is heteroaryl. In an embodiment, L is triazole. In an embodiment, L is isoxazole. R1is H, alkyl, a carbocycle or a heterocycle wherein said alkyl, carbocycle and heterocycle are each optionally substituted with halogen, OH, amino, oxo, carboxy, acyloxy, alkoxycarbonyl, alkoxyacyloxy, alkoxycarbonyloxy, aminocarbonyl, a carbocycle optionally substituted with alkyl, haloalkyl, oxo, amino and halogen and a heterocycle optionally substituted with alkyl, oxo, amino and halogen; and a carbocycle or heterocycle optionally substituted with alkyl, haloalkyl, oxo, amino and halogen. In an embodiment, R1is H. In an embodiment, R1is alkyl. In an embodiment, R1is methyl. In an embodiment, R1is ethyl. In an embodiment, R1is n-propyl. In an embodiment, R1is i-propyl. In an embodiment, R1is cyclohexyl. In an embodiment, R1is alkyl substituted with OH. In an embodiment, R1is alkyl substituted with oxo. In an embodiment, R1is alkyl substituted with carboxy. In an embodiment, R1is alkyl substituted with acyloxy. In an embodiment, R1is alkyl substituted with alkoxycarbonyl. In an embodiment, R1is alkyl substituted with alkoxyacyloxy. In an embodiment, R1is alkyl substituted with alkoxycarbonyloxy. In an embodiment, R1is alkyl substituted with aminocarbonyl. In an embodiment, R1is methyl. In an embodiment, R1is propyl. In an embodiment, R1is hydroxyethyl. In an embodiment, R2is H or R5. In an embodiment, R2is H. In an embodiment, R2is R5. In an embodiment, R2is methyl. In an embodiment, R2is t-butyl. In an embodiment, R2is benzhydryl. In an embodiment, R2is benzyl. R3is absent or alkyl. In an embodiment, R3is methyl. In an embodiment, R3is absent. R4is absent, H, OH, ═O, —R6, —O—R6, —C(O)O—R6, —O—C(O)—R6, —O—C(O)—O—R6, —O—C(O)—NR5R6, —NR5R6, —NR5—C(O)—R6, —NR5—C(O)—O—R6, —NR5—SO2—R6, ═N—O—R5. In an embodiment, R4is H. In an embodiment, R4is OH. In an embodiment, R4is ═O. In an embodiment, R4is —O—R6. In an embodiment, R4is —C(O)O—R6. In an embodiment, R4is —O—C(O)—R6. In an embodiment, R4is —O—C(O)—O—R6. In an embodiment, R4is —O—C(O)—NR5R6. In an embodiment, R4is —NR5R6. In an embodiment, R4is —NR5—SO2—R6. In an embodiment, R4is ═N—O—R5. In an embodiment, R4is as defined and the carbon from which it depends is part of a double bond. In an embodiment, R4is H and the carbon from which it depends is not part of a double bond. In an embodiment, R4is —R6. R5is H or alkyl optionally substituted with a carbocycle or heterocycle wherein said carbocycle and heterocycle are optionally substituted with halogen, OH, oxo and alkyl. In an embodiment, R5is H. In an embodiment, R5is alkyl. In an embodiment, R5is methyl. R6is H, alkyl, a carbocycle, a heterocycle wherein said alkyl, carbocycle and heterocycle are optionally substituted with halogen, OH, SH, alkylthio, —S(O)-alkyl, —SO2-alkyl, amino, —NHC(O)-alkyl, oxo, alkyl, carboxyl, acyl, acyloxy, alkoxy, alkoxycarbonyl, a carbocycle optionally substituted with halogen, OH, amino or alkyl, or a heterocycle optionally substituted with halogen, OH, amino or alkyl; and wherein one or more non-adjacent methylene groups in each of said alkyl groups of R6are optionally replaced with —O— or —S—. In an embodiment, R6is H. In an embodiment, R6is alkyl. In an embodiment, R6is methyl. In an embodiment, R6is ethyl. In an embodiment, R6is cyclopropyl. In an embodiment, R6is allyl. In an embodiment, R6is vinyl. In an embodiment, R6is OH. In an embodiment, R6is alkoxycarbonyl. In an embodiment, R6is methyloxycarbonyl. In an embodiment, R6is ethyloxycarbonyl. In an embodiment, R6is amino. In an embodiment, R6is NH2. In an embodiment, R6is alkoxy. In an embodiment, R6is polyalkoxyalkyl. In an embodiment, R6is oxo. In an embodiment, R6is alkylthio. In an embodiment, R6is —S-Me. In an embodiment, R6is —S-Et. In an embodiment, Rxis H. In an embodiment, Rxis —C(O)O—R1. In an embodiment, Rxis alkyl. In an embodiment, Rxis alkyl optionally substituted with —C(O)O—R1. In an embodiment ‘n’ is 1. In another embodiment, ‘n’ is 2. In a further aspect of the invention, there is provided a compound of formula II wherein R1, R2, R3, R4, L, X, Q, V and W are as defined for compounds of formula I. Furthermore, particular embodiments of formula II are analogous to those embodiments specified herein for formula I. For example, particular embodiments of formula II include compounds according to formula Ia-Ip, Ia′-If except that the dioxolone ring is saturated. In an embodiment, the compound of the invention is selected from the group consisting of:(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid (122-3); (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-Heptamethyl-10-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (176-2);(2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-Heptamethyl-10-(((5-methyl-2-oxo-1,3-dioxolan-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (178-1); (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-Hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methylthio)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (194-10);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-Hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methylsulfonyl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (195-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((L-Valyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (196-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(Benzoyloxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (197-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((Cyclopropanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (198-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((R)-2-Methoxypropanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (203-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((S)-2-Methoxypropanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (204-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(Methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (205-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((Ethylcarbamoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (206-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((butylcarbamoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (207-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-((pyrrolidine-1-carbonyl)oxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (208-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (209-3);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (211-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopentanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (212-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-((3-(piperidin-1-yl)propanoyl)oxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (215-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((isopropoxycarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (216-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-Difluoroacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (223-1);(2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-Hydroxy-2,4a,6a,6b,9,12a-hexamethyl-9-((2-((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-oxoethoxy)methyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (240-8);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Acetoxy-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (243-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-Hydroxyacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (244-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-Methoxyacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (245-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-Tetraoxadodecanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (246-3); (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Methoxy-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (249-5);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (252-2);(2S,4aS,6aS,6bR,8aR,9R,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-Hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (253-4);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Acetoxy-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (254-3);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Acetoxy-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (255-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-Difluoroacetoxy)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (256-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (258-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (264-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxolan-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (265-2)(2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-10-(2-((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-oxoethoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (279-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (280-7); (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((methoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (281-3);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(pentanoyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (282-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(4-methylpiperazin-1-yl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (283-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((3-morpholinopropanoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (284-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(methyl sulfonamido)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (285-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(((2-morpholinoethyl)carbamoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (286-4);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methylsulfinyl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (289-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((dimethylglycyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (290-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((acetylglycyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (291-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetamido-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid;(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(allyloxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (297-5);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(3-methoxy-3-oxopropanamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (298-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-methoxy-4-oxobutanamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (299-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (300-1); (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((methoxycarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (301-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (302-3);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid (307-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-methoxyacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (308-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-Difluoroacetamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (309-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (314-4);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (315-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (316-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (317-6); (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (318-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (319-1);(3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-4-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-4,6a,6b,8a,11,14b-hexamethyl-11-(methylcarbamoyl)-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicen-3-yl propionate (320-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (321-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (322-6);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (323-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (324-3);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-10-m ethoxy-2,4a, 6a,6b, 9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (325-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (326-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (327-8);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (326-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (327-8);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(1H-pyrrol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (328-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid;(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (330-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(5-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (331-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(3-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (332-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (333-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (334-8);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (335-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(5-(ethoxycarbonyl)-3-methyl-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (336-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(3-(ethoxycarbonyl)-5-methyl-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (337-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(4-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (338-4);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-10-(4-methyl-1H-pyrazol-1-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (339-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (341-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-amino-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (342-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (343-3);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(allyl(2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (344-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((S)-1-methoxy-1-oxopropan-2-yl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (345-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(1-methyl cyclopropane-1-carboxamido)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (346-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(2-oxopyrrolidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (347-4);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (348-11);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (349-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (350-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((R)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (351-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-2-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (352-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-2-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (353-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(piperidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (356-2);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (357-7);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-3-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (358-7);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-3-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (359-1)(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-3-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (360-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)piperidin-1-yl)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (361-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (362-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-acetamido-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (363-5);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-acetamido-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (363-5);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((R)-5-methyl-2-oxooxazolidin-3-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (364-5);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,5-dioxoimidazolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (365-8);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((R)-4-methyl-2,5-dioxoimidazolidin-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (366-1);2-(3-((1-PEG5K-1H-1,2,3-triazol-4-yl)methoxy)-4-nitrobenzyl) 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a, 6a, 6b, 9,12a-h exam ethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (605-2);(2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-10-{[(2-oxo-1,3-dioxolan-4-yl)methoxy]carbonyl}-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 700-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-{[2-(methylsulfanyl)acetyl]oxy}-13-oxo-9-({[2-oxo-5-(propan-2-yl)-2H-1,3-dioxol-4-yl]methoxy}carbonyl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 701-1);(2S,4aS,6aS,6bR,8aR,9S,10R,12aS,12bR,14bR)-10-(acetyloxy)-2,4a,6a,6b,9,10,12a-heptamethyl-13-oxo-9-({[2-oxo-5-(propan-2-yl)-2H-1,3-dioxol-4-yl]methoxy}carbonyl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 702-1);(2S,4aS,6aS,6bR,8aR,9S,10R,12aS,12bR,14bR)-10-(acetyloxy)-9-{[(5-ethyl-2-oxo-2H-1,3-dioxol-4-yl)methoxy]carbonyl}-2,4a, 6a,6b, 9,10,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 703-1);(2S,4aS,6aS,6bR,8aR,9S,10R,12aS,12bR,14bR)-10-(acetyloxy)-2,4a,6a,6b,9,10,12a-heptamethyl-9-{[(5-methyl-2-oxo-2H-1,3-dioxol-4-yl)methoxy]carbonyl}-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 704-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(acetyloxy)-9-{[(5-tert-butyl-2-oxo-2H-1,3-dioxol-4-yl)methoxy]carbonyl}-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 705-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(acetyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-9-{[(2-oxo-5-propyl-2H-1,3-dioxol-4-yl)methoxy]carbonyl}-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 706-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(acetyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-9-{[(2-oxo-5-phenyl-2H-1,3-dioxol-4-yl)methoxy]carbonyl}-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 707-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-[(2-methoxyacetyl)oxy]-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-9-({[2-oxo-5-(propan-2-yl)-2H-1,3-dioxol-4-yl]methoxy}carbonyl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 708-1);(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-[(2-methoxyacetyl)oxy]-2,4a,6a,6b,9,12a-hexamethyl-9-{[(5-methyl-2-oxo-2H-1,3-dioxol-4-yl)methoxy]carbonyl}-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 709-1);(2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-10-(2-oxo-2-{[2-oxo-5-(2,5,8-trioxa-11-thiadodecan-12-yl)-2H-1,3-dioxol-4-yl]methoxy}ethoxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 710-1);(2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-10-(2-{[5-(hydroxymethyl)-2-oxo-2H-1,3-dioxol-4-yl]methoxy}-2-oxoethoxy)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 711-1);(2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-10-{2-[(5-methyl-2-oxo-2H-1,3-dioxol-4-yl)methoxy]-2-oxoethoxy}-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 712-1); and(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-[4-(ethoxycarbonyl)-5-methoxy-1H-pyrazol-1-yl]-2,4a,6a,6b,9,12a-hexamethyl-9-{[(5-methyl-2-oxo-2H-1,3-dioxol-4-yl)methoxy]carbonyl}-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (cmpd 713-1). Compounds of the invention are “soft drugs” which in parent form are active inhibitors of 11b-HSD2 in the gastrointestinal tract of a patient but upon uptake into plasma are enzymatically converted to inactive, or less active, metabolites. This effect provides a desired preferential inhibition of HSD2 in the GI tract relative to HSD2 in kidney. Compounds of the invention disclosed herein were tested in the assays described in examples 112 and were found to inhibit HSD2 by measuring the amount of cortisol before and after contacting cell lysate human or a colon monolayer organoid derived from human colon tissue. Furthermore, each of the compounds tested were found to be more potent HSD2 inhibitors than their corresponding metabolites. Compounds of the invention may contain one or more asymmetric or chiral centers. Accordingly, the compounds may exist as diastereomers, enantiomers or mixtures thereof. The syntheses of the compounds may employ racemates, diastereomers or enantiomers as starting materials or as intermediates. Diastereomeric compounds may be separated by chromatographic or crystallization methods. Similarly, enantiomeric mixtures may be separated using the same techniques or others known in the art. Unless specified, each of the asymmetric centers may be in the R or S configuration and both of these configurations are within the scope of the invention. It is intended that all stereoisomeric forms of the compounds described herein, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof, such as racemic mixtures, form part of the present compounds. It will also be appreciated that certain compounds of Formula I may be used as intermediates for further compounds of Formula I. It will be further appreciated that the compounds described herein may exist in unsolvated, as well as solvated forms with pharmaceutically acceptable solvents, such as water, ethanol, and the like, and it is intended that the compounds embrace both solvated and unsolvated forms. Compounds of the invention are prepared using standard organic synthetic techniques from commercially available starting materials and reagents. It will be appreciated that synthetic procedures employed in the preparation of compounds of the invention will depend on the particular substituents present in a compound and that various protection and deprotection steps that are standard in organic synthesis may be required but may not be illustrated in the following general schemes. The starting materials are generally available from commercial sources or are readily prepared using methods well known to those skilled in the art. For example, compounds of the invention may be prepared from glycyrrhetinic acid shown inFIG.3. For illustrative purposes, schemes herein show general methods for preparing the compounds of the invention, as well as key intermediates. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art. As illustrated in Scheme I, the compounds of the invention in which -Q-W—C(O)-L-X— form an ester linkage —CH2—O—C(O)— to the dioxalone may be prepared starting with a carboxylic acid derivative of glycyrrhetinic acid (either at the 3- or 4-position) and reacting with, for example, a halogenated dioxalone. Compounds of the invention in which -Q-W—C(O)-L-X— form a linkage —CH2—O—C(O)—O— to the dioxalone may be according to the general Scheme 2. Compounds of the invention in which -Q-W—C(O)-L-X— form an amide linkage —CH2—O—C(O)—NH— to the dioxalone may be prepared starting by reacting an isocyanate at the 3- or 4-position of the fused ring system with a hydroxylated dioxalone as illustrated in Scheme 3. In preparing compounds of the invention, protection of remote functionalities (e.g., primary or secondary amines, etc.) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see Greene, T. W., and P. G. M. Wuts.Greene's Protective Groups in Organic Synthesis.4th ed. New York: Wiley-Interscience, 2006. It may be advantageous to separate reaction products from one another and/or from starting materials. The desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (“SMB”) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography. One skilled in the art will apply techniques most likely to achieve the desired separation. Diastereomeric and enantiomeric mixtures can be separated into their individual stereoisomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Enantiomers can also be separated by use of a chiral HPLC column. The invention also includes pharmaceutical compositions or medicaments containing the compounds of the invention and a therapeutically inert carrier, diluent or excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments. Typically, the compounds of Formula I used in the methods of the invention are formulated by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed. The pH of the formulation depends mainly on the particular use and the concentration of compound, but may range anywhere from about 3 to about 8. Formulation in an acetate buffer at pH 5 is a suitable embodiment. In one embodiment, formulations comprising compounds of the invention are sterile. The compounds ordinarily will be stored as a solid composition, although lyophilized formulations or aqueous solutions are acceptable. Compositions comprising compounds of the invention will be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of administration, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The “effective amount” of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to inhibit conversion of cortisol to cortisone by HSD2. Such amount may be below the amount that is toxic to normal cells, or the mammal as a whole. The compound of the invention may be administered by any suitable means. In a particular embodiment, the compounds are administered orally. In a particular embodiment, the compounds are administered rectally. Generally, the initial pharmaceutically effective amount of the compound of the invention administered parenterally per dose will be in the range of about 0.01-1,000 mg/kg/day, for example about 0.1 to 100 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.5 to 50 mg/kg/day. Oral unit dosage forms, such as tablets and capsules, may contain from about 25 to about 1000 mg of the compound of the invention. In a particular embodiment, an effective amount is the amount of the compound of the invention sufficient to enhance colonic potassium secretion by about 15 mmol/day. In a particular embodiment, an effective amount is the amount of the compound of the invention sufficient to enhance colonic potassium secretion by about 1 mmol/day. In a particular embodiment, an effective amount is the amount of the compound of the invention sufficient to enhance colonic potassium secretion by about 5 mmol/day. In a particular embodiment, an effective amount is the amount of the compound of the invention sufficient to enhance colonic potassium secretion by about 10 mmol/day. In a particular embodiment, an effective amount is the amount of the compound of the invention sufficient to enhance colonic potassium secretion by about 15 mmol/day. In a particular embodiment, an effective amount is the amount of the compound of the invention sufficient to enhance colonic potassium secretion by about 20 mmol/day. The compounds may be administered in any convenient administrative form, e.g., tablets, capsules, solutions, dispersions, suspensions, syrups, suppositories, gels, emulsions etc. An example of a suitable oral dosage form is a tablet containing about 25 mg, 50 mg, 100 mg, 250 mg, or 500 mg of the compound of the invention compounded with about 90-30 mg anhydrous lactose, about 5-40 mg sodium croscarmellose, about 5-30 mg polyvinylpyrrolidone (“PVP”) K30, and about 1-10 mg magnesium stearate. The powdered ingredients are first mixed together and then mixed with a solution of the PVP. The resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment. Another formulation may be prepared by mixing a compound described herein and a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C., et al.,Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al.Remington; The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C.Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound described herein or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament). In an embodiment, the formulation releases the compound in response to contact with colonic enzyme, for example, enzymes created by enterobacteria. Certain starch-based capsule coatings may be used that are resistant to digestion in the stomach and small intestine but are degraded by microbial (normal gut flora) enzymes once the dosage form reaches the colon. In an embodiment, the compound of the invention is administered orally. In another embodiment, the compound is formulated for colonic delivery. Colonic delivery may be effected in response to pH time, microbes, and pressure. In an embodiment, the formulation releases the compound in response to colonic pH. Release of the compound is triggered by the pH increase as the formulation travels through the GI tract. Formulations are based on polymers that are insoluble at the lower pH in the stomach and upper small intestine and soluble in the higher pH found in the distal small intestine, for example, polymers that are derivatives of acrylic acid and cellulose which withstand an environment as low as pH ˜1.2. Suitable enteric polymers include, polyvinyl acetate phthalate (PVAP) e.g. Coateric®, cellulose acetate trimellitate (CAT), hydroxypropyl methylcellulose phthalate (HPMCP) e.g. HP-50, HP-55, HP-55S, hydroxypropylmethylcellulose acetate succinate (HPMCAS) e.g. LF grade, MF grade or HF grade, methacrylic acid copolymer e.g. Eudragit® L100-55, L30D-55, L-1000, L12.5, S-100, S12.5, FS30D, cellulose acetate phthalate (CAP) e.g. Aquateric®, and shellac e.g. MarCoat® 125 or 125N. In an aspect of the invention, there is provided a method of inhibiting conversion of cortisol to cortisone by HSD2 comprising contacting HSD2 with a compound of Formula I. In another aspect of the invention, there is provided a method for promoting activation MR in a mammal, comprising administering to said mammal an effective amount of a compound of Formula I. In another aspect of the invention, there is provided a method of reducing potassium levels in plasma of a mammal, comprising administering to said mammal an effective amount of a compound of Formula I. In another aspect of the invention, there is provided a method for promoting potassium ion secretion into the colonic lumen of a mammal, comprising administering to said mammal an effective amount of a compound of Formula I. In an aspect of the invention, there is provided a method for treating and/or preventing hyperkalemia in a mammal, comprising administering to said mammal an effective amount of a compound of Formula I. Hyperkalemia occurs especially frequently in patients with chronic kidney disease (CKD), hypertension, heart failure and diabetes. Accordingly, in an embodiment of the invention the methods of treating and/or preventing hyperkalemia is in a patient having CKD hypertension, heart failure and diabetes. Patients suffering for these conditions are often treated with certain classes of medications, such as angiotensin-converting-enzyme (ACE) inhibitors, angiotensin-receptor blockers (ARBs) or other inhibitors of the renin-angiotensin-aldosterone system (RAAS) in order to regulate blood pressure. However such medications promote potassium retention. Accordingly, there is provided a method of treating and/or preventing hyperkalemia in a mammal comprising administering a compound of formula I in combination with an inhibitor of the RAAS system. In an embodiment, the RAAS inhibitor is an ACE inhibitor. The compounds described herein and stereoisomers, diastereomers, enantiomers, tautomers and pharmaceutically acceptable salts thereof may be employed alone or in combination with other anti-hyperkalemia agents that works by a different mechanism of action. The compound of the invention may be administered together with the other anti-hyperkalemia agent in a unitary pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order. Such sequential administration may be close in time or remote in time. In an embodiment, the other anti-hyperkalemic compound is a potassium ion binder such as a cross-lined polystyrene sulfonate (PSS) polymer resins. In an embodiment, the PSS resin is crosslinked with divinylbenzene (DVB) co-polymer. DVB-crosslinked PSS is the most common agent used in the management of hyperkalemia in hospitalized patients. PSS is typically provided as a sodium or calcium salt, and in the lumen of the intestine it exchanges sodium or calcium ions for secreted potassium ions. Most of this takes place in the colon, the site of most potassium secretion in the gut. In an embodiment, the anti-hyperkalemic PSS resin is described in WO2016111855 (incorporated herein by reference). In an embodiment, the PSS resin is a calcium salt of a PSS polymer resins crosslinked with DVB co-polymer. In an embodiment, the PSS resin is cross-linked with from 1.0 to 1.9 percent of DVB. In an embodiment, the PSS resin is cross-linked with from 1.6 to 1.9 percent of DVB. In an embodiment, the PSS resin is cross-linked with about 1.8 percent of DVB. In an embodiment, the other anti-hyperkalemia agent is Kayexalate®, Argamate®, Kionex®, Resonium® or RDX7675. In another embodiment, the other anti-hyperkalemia agent is a fluoroacrylate polymer incorporating a potassium-binding carboxylate group e.g. patiromer (Veltassa®). In an embodiment, the other anti-hyperkalemia agent is an insoluble, non-absorbed zirconium-sodium silicate that traps potassium ions within its crystalline lattice structure e.g. ZS-9 (Lokelma®). In an embodiment, the other anti-hyperkalemia agent is a crosslinked polyacrylic acid e.g. CLP-1001. In another aspect of the invention, it has been found unexpectedly that HSD2 inhibition in combination with inhibition of sodium-hydrogen exchanger (NHE) synergistically increase excretion of potassium into feces. NHE is found in the tubulus proximal of the nephron of the kidney and in the apical membrane of enterocytes of the intestine. The isoform known as NHE3 is primarily responsible for maintaining the balance of sodium and also indirectly linked to buffering of blood pH. The NHE3 antiporter imports one sodium ion into the cytosol of a cell as it ejects one hydrogen ion from the cell into the intestinal lumen and proximal tubule lumen. As shown inFIG.1, it has been demonstrated that there is a synergistic effect on fecal potassium excretion when inhibiting HSD2 and NHE. Accordingly, there is provided a method for removing potassium from plasma and/or tissue of a mammal comprising administering to said mammal an effective amount of an HSD2 inhibitor or an MR agonist in combination with a compound that increases fluid volume in the colon. There is also provided a method for removing potassium from plasma and/or tissue of a mammal comprising administering to said mammal an effective amount of an HSD2 inhibitor or an MR agonist in combination with a compound that removes sodium from plasma and/or tissue. There is also provided a method for removing potassium from plasma and/or tissue of a mammal comprising administering to said mammal an effective amount of an HSD2 inhibitor or an MR agonist in combination with a compound that promotes excretions of sodium into the gastrointestinal tract. In an embodiment, the compound is a laxative that increases fluid in the colon. In an embodiment, the laxative is bisacodyl. In an embodiment, the laxative is picosulfate. In an embodiment, the laxative is MgOH. In an embodiment, the laxative is MiraLAX® (PEG 3350). In an embodiment, the laxative is lactulose. In an embodiment, the compound is an activator of intestinal guanylate cyclase. In an embodiment the guanylate cyclase agonist is linaclotide. In an embodiment, the guanylate cyclase agonist is plecanatide. In an embodiment, the compound is an activator of intestinal C1C-2 chloride channel. In an embodiment, the C1C-2 chloride channels activator is lubiprostone. There is also provided a method for removing potassium from plasma and/or tissue of a mammal comprising administering to said mammal an effective amount of an HSD2 inhibitor or an MR agonist in combination with a an NHE inhibitor. In an embodiment, the HSD2 inhibitor or MR agonist and the NHE inhibitor compounds are administered concurrently. In an embodiment, the HSD2 or MR agonist and the NHE inhibitor compounds are administered sequentially. In an embodiment, the HSD2 inhibitor or MR agonist is administered prior to the NHE inhibitor or MR agonist. In an embodiment, the NHE inhibitor or MR agonist compound is administered prior to the HSD2 inhibitor or MR agonist. In an embodiment, the NHE inhibitor is an NHE3 inhibitor. In another aspect of the invention, there is provided a pharmaceutical composition comprising an HSD2 inhibitor and an NHE inhibitor. In another aspect, there is provided a pharmaceutical composition comprising an MR agonist and an NHE inhibitor. In another aspect, there is provided a method for treating hyperkalemia in a mammal comprising administering to said mammal an effective amount of an HSD2 inhibitor or an MR agonist in combination with an NHE inhibitor. In an embodiment, the NHE inhibitor is an NHE3 inhibitor. In an embodiment, the MR agonist is fludrocortisone. In another aspect, there is provided a method for treating hyperkalemia in a mammal comprising administering to said mammal an effective amount of an HSD2 inhibitor in combination with an NHE inhibitor. In an embodiment, the NHE inhibitor is an NHE3 inhibitor. In another aspect, there is provided a composition comprising an HSD2 inhibitor and an NHE inhibitor. In an embodiment, the composition is a pharmaceutical composition. In an embodiment there is an effective amount of HSD2 inhibitor compound and the NHE inhibitor compound. In an embodiment, the composition further comprises a pharmaceutically acceptable carrier, excipient and/or diluent. In an embodiment, the HSD2 inhibitor is glycyrrhetinic acid or an analogue thereof. In an embodiment, the HSD2 inhibitor is glycyrrhetinic acid. In an embodiment, the HSD2 inhibitor is glycyrrhizin. In an embodiment, the HSD2 inhibitor is a compound according to formula I herein. In an embodiment, the NHE inhibitor is an NHE3 inhibitor. In an embodiment the NHE3 inhibitor is a compound described in: U.S. Pat. Nos. 5,866,610; 6,399,824; 6,911,453; 6,703,405; 6,005,010; 6,736,705; 6,887,870; 6,737,423; 7,326,705; 5,824,691 (WO94/026709); 6,399,824 (WO02/024637); U.S. Pat. Pub. Nos. 2004/0039001 (WO02/020496); 2005/0020612 (WO03/055490); 2004/0113396 (WO03/051866); 2005/0020612; 2005/0054705; 2008/0194621; 2007/0225323; 2004/0039001; 2004/0224965; 2005/0113396; 2007/0135383; 2007/0135385; 2005/0244367; 2007/0270414; International Publication Nos. WO 01/072742; WO 01/021582 (CA2387529); WO97/024113 (CA02241531) WO2010078449; WO2014029983; WO2014029984; and European Pat. No. EP0744397 (CA2177007); each of which is incorporated herein by reference in their entirety. In an embodiment, the NHE inhibitor is a compound that is minimally systemic, i.e., it inhibits NHE in the intestine and is substantially non-bioavailable. In an embodiment, the NHE inhibitor is a compound Formula (I) or (IX): wherein:NHE is a NHE-binding small molecule that comprises (i) a hetero-atom containing moiety, and (ii) a cyclic or heterocyclic scaffold or support moiety bound directly or indirectly thereto, the heteroatom-containing moiety being selected from a substituted guanidinyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the scaffold or support moiety to form a fused bicyclic structure; and,Z is a moiety having at least one site thereon for attachment to the NHE-binding small molecule, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable; and,E is an integer having a value of 1 or more. In certain embodiments, the total number of freely rotatable bonds in the NHE-Z molecule is at least about 10. In certain embodiments, the total number hydrogen bond donors in the NHE-Z molecule is at least about 5. In some embodiments, the total number of hydrogen bond acceptors in the NHE-Z molecule is at least about 10. In certain embodiments, the total number of hydrogen bond donors and hydrogen bond acceptors in the NHE-Z molecule is at least about 10. In some embodiments, the Log P of the NHE-Z binding compound is at least about 5. In certain embodiments, the log P of the NHE-Z binding compound is less than about 1, or less than about 0. In certain embodiments, the scaffold is a 5-member or 6-member cyclic or heterocyclic moiety. In certain embodiments, the scaffold is aromatic. In some embodiments, the scaffold of the NHE-binding small molecule is bound to the moiety, Z, the compound having the structure of Formula (II): wherein:Z is a Core having one or more sites thereon for attachment to one or more NHE-binding small molecules, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable;B is the heteroatom-containing moiety of the NHE-binding small molecule, and is selected from a substituted guanidinyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the Scaffold moiety to form a fused, bicyclic structure;Scaffold is the cyclic or heterocyclic scaffold or support moiety of the NHE-binding small molecule, which is bound directly or indirectly to heteroatom-containing moiety, B, and which is optionally substituted with one or more additionally hydrocarbyl or heterohydrocarbyl moieties;X is a bond or a spacer moiety selected from a group consisting of substituted or unsubstituted hydrocarbyl or heterohydrocarbyl moieties, and in particular substituted or unsubstituted C1-7hydrocarbyl or heterohydrocarbyl, and substituted or unsubstituted, saturated or unsaturated, cyclic or heterocyclic moieties, which links B and the Scaffold; andD and E are integers, each independently having a value of 1 or more.In some embodiments, the compound is an oligomer, dendrimer or polymer, and further wherein Z is a Core moiety having two or more sites thereon for attachment to multiple NHE-binding small molecules, either directly or indirectly through a linking moiety, L, the compound having the structure of Formula (X): wherein L is a bond or linker connecting the Core to the NHE-binding small molecule, and n is an integer of 2 or more, and further wherein each NHE-binding small molecule may be the same or differ from the others. In some embodiments, the NHE-binding small molecule has the structure of Formula (IV): or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein:each R1, R2, R3, R5and R9are independently selected from H, halogen, —NR7(CO)R8, —(CO)NR7R8, —SO2—NR7R8, —NR7SO2R8, —NR7R8, —OR7, —SR7, —O(CO)NR7R8, —NR7(CO)OR8, and —NR7SO2NR8, where R7and R8are independently selected from H or a bond linking the NHE-binding small molecule to L, provided at least one is a bond linking the NHE-binding small molecule to L;R4is selected from H, C1-C7alkyl, or a bond linking the NHE-binding small molecule to L;R6is absent or selected from H and C1-C7alkyl; andAr1 and Ar2 independently represent an aromatic ring or a heteroaromatic ring. In certain embodiments, the NHE-binding small molecule has the following structure: or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein:each R1, R2and R3are independently selected from H, halogen, —NR7(CO)R8, —(CO)NR7R8, —SO2—NR7R8, —NR7SO2R8, —NR7R8, —OR7, —SR7, —O(CO)NR7R8, —NR7(CO)OR8, and —NR7SO2NR8, where R7and R8are independently selected from H or a bond linking the NHE-binding small molecule to L, provided at least one is a bond linking the NHE-binding small molecule to L. In some embodiments, the NHE-binding small molecule has one of the following structures: or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof. In certain embodiments, L is a polyalkylene glycol linker. In certain embodiments, L is a polyethylene glycol linker. In some embodiments, n is 2. In certain embodiments, the Core has the following structure: wherein:X is selected from the group consisting of a bond, —O—, —NH—, —S—, C1-6alkylene, —NHC(═O)—, —C(═O)NH—, —NHC(═O)NH—, —SO2NH—, and —NHSO2—;Y is selected from the group consisting of a bond, optionally substituted C1-8alkylene, optionally substituted aryl, optionally substituted heteroaryl, a polyethylene glycol linker, —(CH2)1-6O(CH2)1-6— and —(CH2)1-6NY1(CH2)1-6—; andY1is selected from the group consisting of hydrogen, optionally substituted C1-8alkyl, optionally substituted aryl or optionally substituted heteroaryl. In some embodiments, the Core is selected from the group consisting of: wherein: Lisa bond or a linking moiety; NHE is a NHE-binding small molecule; and n is a non-zero integer. In an embodiment, the NHE inhibitor is:N,N′,N″-(2,2′,2″-nitrilotris(ethane-2,1-diyl))tris(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(2,2′-(ethane-1,2-diylbis(oxy))bis(ethane-2,1-diyl))bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(1,4-phenylenebis(methylene))bis(3-(6,8-di chloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(1,4-phenylenebis(methylene))bis(3-(6,8-di chloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(butane-1,4-diyl)bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(dodecane-1,12-diyl)bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′,N″,N′″-(3,3′,3″,3′″-(butane-1,4-diylbis(azanetriyl))tetrakis(propane-3,1-diyl))tetrakis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(butane-1,4-diyl)bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(dodecane-1,12-diyl)bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′,N″-(2,2′,2″-nitrilotris(ethane-2,1-diyl))tris(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′,N″,N′″-(3,3′,3″,3′″-(butane-1,4-diylbis(azanetriyl))tetrakis(propane-3,1-diyl))tetrakis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(1,4-phenylenebis(methylene))bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(2,2′-(ethane-1,2-diylbis(oxy))bis(ethane-2,1-diyl))bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N1,N8-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)octanediamide;2-(N-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl)sulfamoylamino)ethylphosphonic acid;2-(N-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl)sulfamoylamino)ethylphosphonic acid;N,N′-(butane-1,4-diyl)bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide];N,N′-(1,4-phenylenebis(methyl ene))bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide];N,N′-(2,2′-(ethane-1,2-diylbis(oxy))bis(ethane-2,l-diyl))bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide];N,N′-(2,2′-(2,2′-oxybis(ethane-2,l-diyl)bis(oxy))bis(ethane-2,l-diyl))bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide](E)-3-(4-(4-(N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-N-(diaminomethylene)-2-methylacrylamide;N,N′-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-1,25-diyl)bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide];N,N′-(13,20 dioxo-3, 6, 9, 24, 27, 30-hexaoxa-12, 21-diazadotricontane-1,32-diyl)bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide];N,N′-(2,2′-(2,2′-(2,2′-(2,2′-(4,4′-oxybis(methylene)bis(1H-1,2,3-triazole-4,1-diyl))bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide];N,N′-(2,2′-(2,2′-(2,2′-(2,2′-(4,4′-oxybis(methylene)bis(1H-1,2,3-triazole-4,1-diyl))bis(ethane-2,1-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide)1-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-1H-1,2,3-triazole-4,5-dicarboxylic acid;N1,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N31-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-4,7,10,13,16,19,22,25,28-nonaoxahentriacontane-1,31-diamide;N1,N31-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-4,7,10,13,16,19,22,25,28-nonaoxahentriacontane-1,31-diamide;N,N′-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-1,25-diyl)bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N1,N31-bis(2-(2-(2-(2-(4-(4-((E)-3-(diaminomethyleneamino)-2-methyl-3-oxoprop-1-enyl)-2,6-difluorophenoxy)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-4,7,10,13,16,19,22,25,28-nonaoxahentriacontane-1,31-diamide;N,N′-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-1,25-diyl)bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N1,N4-bis(20-(4-(4-((E)-3-(diaminomethyleneamino)-2-methyl-3-oxoprop-1-enyl)-2,6-difluorophenoxy)phenylsulfonamido)-3,6,9,12,15,18-hexaoxaicosyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroiso-quinolin-4-yl)phenylsulfonamido)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-ethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;3,3′-(2,2′-(2,2′-(2,2′-oxybis(ethane-2,l-diyl)bis(oxy))bis(ethane-2,l-diyl))bis(6,8-dichloro-1,2,3,4-tetrahydroisoquinoline-4,2-diyl))dianiline;N1,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylamino)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(1-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylamino)-1-oxo-5,8,1 l-trioxa-2-azatridecan-13-yl)-2,3-dihydroxysuccinamide;N1,N2-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)oxalamide;N1,N4-bis(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide;2,2′-oxybis(N-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide);(2R,3R)—N1,N4-bis(2-(2-(2-(3-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylamino)-3-oxopropoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N2-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)oxal amide;N1,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide;N1,N3-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,2-dimethylmalonamide;N1,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,2-dimethylmalonamide;N,N′-(2,2′-(2,2′-(2,2′-(2,2′-(pyridine-2,6-diylbis(oxy))bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,1-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);2,2′-(methylazanediyl)bis(N-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide) tris(2,2,2-trifluoroacetate);5-amino-N1,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)isophthalamide tris(2,2,2-trifluoroacetate);2,2′-oxybis(N-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide);5-bromo-N1,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)isophthalamide bis(2,2,2-trifluoroacetate);N1,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2-hydroxymalonamide bis(2,2,2-trifluoro acetate);N1,N2-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)oxal amide;N1,N4-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide;3,5-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethylcarbamoyl)benzenesulfonic acid;N1,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-5-hydroxyisophthalamide;(2R,3R)—N1,N4-bis(3-((3-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)propyl)(methyl)amino)propyl)-2,3-dihydroxysuccinamide;2,2′-oxybis(N-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide);N1,N3-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,2-dimethylmalonamide;N1,N2-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)oxalamide;2,2′-oxybis(N-(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide);N1,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide;N1,N4-bis(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide;2,2′-oxybis(N-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide);(S or R)—N,N′-(10,17-dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis(3-((S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);(S or R)—N,N′-(2,2′-(2,2′-(2,2′-(1,4-phenylenebis(azanediyl))bis(oxomethylene)bis (azanediyl)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(butane-1,4-diyl)bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)acetamido)acetamido)acetamide);N1,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N,N′-(2,2′-(2,2′-(2,2′-(1,4-phenyl enebis(methylene))bis(azanediyl)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);(2R,3R)—N1,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N,N′-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21-tetraazadotriacontane-1,32-diyl)bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(1,1′-(1,4-phenylenebis(azanediyl))bis(1-oxo-5,8,11-trioxa-2-azatridecane-13,l-diyl))bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);(2R,3R)—N1,N4-bis(20-(4-(4-((E)-3-(diaminomethyleneamino)-2-methyl-3-oxoprop-1-enyl)-2,6-difluorophenoxy)phenylsulfonamido)-3,6,9,12,15,18-hexaoxaicosyl)-2,3-dihydroxysuccinamide;(E)-3-(4-(4-(N-(20-amino-3,6,9,12,15,18-hexaoxaicosyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-N-(diaminomethylene)-2-methylacrylamide;(2R,3R)—N1,N4-bis(2-(2-(2-(2-(4-(4-((E)-3-(diaminomethyleneamino)-2-methyl-3-ox oprop-1-enyl)-2,6-difluorophenoxy)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;2,2′,2″-nitrilotris(N-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide);N-(32-amino-3,6,9,12,15,18,21,24,27,30-decaoxadotriacontyl)-3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide;N1,N3,N5-tris(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)benzene-1,3,5-tricarboxamide;N1,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)terephthalamide;N1,N31-bis(32-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)-3,6,9,12,15,18,21,24,27,30-decaoxadotriacontyl)-4,7,10,13,16,19,22,25,28-nonaoxahentriacontane-1,31-diamide;2R,3R)—N1,N4-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N3-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)benzene-1,3-di sulfonamide;N4,N4′-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)biphenyl-4,4′-disulfonamide;(14R,15R)-1-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)-14,15-dihydroxy-13-oxo-3,6,9-trioxa-12-azahexadecan-16-oic acid;(2S,3S)—N1,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(2-(3-((R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(2-(3-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(2-(4-((R or S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N3-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)isophthalamide;(2R,3S)—N1,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;N1,N2-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)phthalamide;N1,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)terephthalamide;N,N′-(10-oxo-3,6,14,17-tetraoxa-9,11-diazanonadecane-1,19-diyl)bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N1,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)terephthalamide;N1,N4-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)terephthalamide;N,N′-(10-oxo-3,6,14,17-tetraoxa-9,11-diazanonadecane-1,19-diyl)bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(10,17-di oxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(2,2′-(2,2′-(2,2′-(1,4-phenylenebis(azanediyl))bis(oxomethylene)bis(azanediyl)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(10,17-di oxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);phenylenebis(azanediyl))bis(oxomethylene)bis(azanediyl)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide;(2S,3S)—N1,N4-bis(2-(2-(2-(2-(3-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;(2R,3R)—N1,N4-bis(2-(2-(2-(2-(3-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;(2S,3S)—N1,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;(2R,3R)—N1,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide;(S or R)—N,N′-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21-tetraazadotriacontane-1,32-diyl)bis(3-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);(S or R)—N,N′-(1,1′-(1,4-phenylenebis(azanediyl))bis(1-oxo-5,8,11-trioxa-2-azatridecane-13,l-diyl))bis(3-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N1,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-terephthalamide;N1-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide;N,N′-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21-tetraazadotriacontane-1,32-diyl)bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);N,N′-(1,1′-(1,4-phenylenebis(azanediyl))bis(1-oxo-5,8,11-trioxa-2-azatridecane-13,l-diyl))bis(4-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);(S or R)—N,N′-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-1,25-diyl)bis(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide);(S or R)—N,N′-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21-tetraazadotriacontane-1,32-diyl)bis(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide); or(S or R)—N,N′-(1,1′-(1,4-phenylenebis(azanediyl))bis(1-oxo-5,8,11-trioxa-2-azatridecane-13,1-diyl))bis(4-((S or R)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide). In an embodiment, the NHE inhibitor is: In some embodiments, the compound has the following structure of Formula (I-H): or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof,wherein:(a) n is an integer of 2 or more;(b) Core is a Core moiety having two or more sites thereon for attachment to two or more NHE-binding small molecule moieties;(c) L is a bond or linker connecting the Core moiety to the two or more NHE-binding small molecule moieties; and(d) NHE is a NHE-binding small molecule moiety having the following structure of Formula (XI-H): wherein:B is selected from the group consisting of aryl and heterocyclyl;each R5is independently selected from the group consisting of hydrogen, halogen, optionally substituted C1-4alkyl, optionally substituted C1-4alkoxy, optionally substituted C1-4thioalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, hydroxyl, oxo, cyano, nitro, —NR7R8, —NR7C(═O)R8, —NR7C(═O)OR8, —NR7C(═O)NR8R9, —NR7SO2R8, —NR7S(O)2NR8R9, —C(═O)OR7, —C(═O)R7, —C(═O)NR7R8, —S(O)1-2R7, and —SO2NR7R8, wherein R7, R8, and R9are independently selected from the group consisting of hydrogen, C1-4alkyl, or a bond linking the NHE-binding small molecule moiety to L, provided at least one is a bond linking the NHE-binding small molecule moiety to L;R3and R4are independently selected from the group consisting of hydrogen, optionally substituted C1-4alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; orR3and R4form together with the nitrogen to which they are bonded an optionally substituted 4-8 membered heterocyclyl; andeach R1is independently selected from the group consisting of hydrogen, halogen, optionally substituted C1-6alkyl and optionally substituted C1-6alkoxy. In some embodiments, n is 2. In certain embodiments, L is a polyalkylene glycol linker. In certain embodiments, L is a polyethylene glycol linker. In certain embodiments, the Core has the following structure: wherein:X is selected from the group consisting of a bond, —O—, —NH—, —S—, C1-6alkylene, —NHC(═O)—, —C(═O)NH—, —NHC(═O)NH—, —SO2NH—, and —NHSO2—;Y is selected from the group consisting of a bond, optionally substituted C1-8alkylene, optionally substituted aryl, optionally substituted heteroaryl, a polyethylene glycol linker, —(CH2)1-6O(CH2)1-6— and —(CH2)1-6NY1(CH2)1-6—; andY1is selected from the group consisting of hydrogen, optionally substituted C1-8alkyl, optionally substituted aryl or optionally substituted heteroaryl. In some embodiments, the Core is selected from the group consisting of In certain embodiments, the NHE-binding small molecule moiety has the following structure of Formula (XII-H): wherein:each R3and R4are independently selected from the group consisting of hydrogen and optionally substituted C1-4alkyl, or R3and R4, taken together with the nitrogen to which they are bonded, form an optionally substituted 4-8 membered heterocyclyl;each R1is independently selected from the group consisting of hydrogen, halogen, C1-6alkyl, and C1-6haloalkyl; andR5is selected from the group consisting of —SO2—NR7— and —NHC(═O)NH—, wherein R7is hydrogen or C1-4alkyl. In some embodiments, R3and R4, taken together with the nitrogen to which they are bonded, form an optionally substituted 5 or 6 membered heterocyclyl. In certain embodiments, the optionally substituted 5 or 6 membered heterocyclyl is pyrrolidinyl or piperidinyl. In certain embodiments, the optionally substituted 5 or 6 membered heterocyclyl is pyrrolidinyl or piperidinyl, each substituted with at least one amino or hydroxyl. In some embodiments, R3and R4are independently C1-4alkyl. In certain embodiments, R3and R4are methyl. In some embodiments, each R1is independently selected from the group consisting of hydrogen or halogen. In certain embodiments, each R1is independently selected from the group consisting of hydrogen, F and Cl. In certain embodiments, the compound has the following structure of Formula (I-I): or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein:(a) NHE is a NHE-binding small molecule moiety having the following structure of Formula (A-I): wherein:each R1, R2, R3, R5and R9are independently selected from H, halogen, —NR7(CO)R8, —(CO)NR7R8, —SO2—NR7R8, —NR7SO2R8, —NR7R8, —OR7, —SR7, —O(CO)NR7R8, —NR7(CO)OR8, and —NR7SO2NR8, where R7and Rx are independently selected from H, C1-6alkyl, —C1-6alkyl-OH or a bond linking the NHE-binding small molecule to L, provided at least one is a bond linking the NHE-binding small molecule to L;R4is selected from H, C1-C7alkyl, or a bond linking the NHE-binding small molecule to L;R6is absent or selected from H and C1-C7alkyl; andAr1 and Ar2 independently represent an aromatic ring or a heteroaromatic ring;(b) Core is a Core moiety having the following structure of Formula (B-I): wherein:X is selected from C(Xi), N and N(C1-6alkyl);X1is selected from hydrogen, optionally substituted alkyl, —NXaXb, —NO2, —NXc—C(═O)—NXc—Xa, —C(═O)NXc—Xa, —NXc—C(═O)—Xa, —NXc—SO2—Xa, —C(═O)—Xaand —OXa,each Xaand Xbare independently selected from hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heteroaryl and optionally substituted heteroarylalkyl;Y is C1-6alkylene;Z is selected from —NZa—C(═O)—NZa—, —C(═O)NZa—, —NZa—C(═O)— and heteroaryl when X is CX1;Z is selected from —NZa—C(═O)—NZa—, —NZa—C(═O)— and heteroaryl when X is N or N(C1-6alkyl); andeach Xcand Zais independently selected from hydrogen and C1-6alkyl; and(c) Lisa bond or linker connecting the Core moiety to the NHE-binding small molecule moieties. In some embodiments, the NHE-binding small molecule moiety has the following structure: wherein:each R1, R2and R3are independently selected from H, halogen, —NR7(CO)R8, —(CO)NR7R8, —SO2—NR7R8, —NR7SO2R8, —NR7R8, —OR7, —SR7, —O(CO)NR7R8, —NR7(CO)OR8, and —NR7SO2NR8, where R7and R8are independently selected from H, C1-6alkyl, —C1-6alkyl-OH or a bond linking the NHE-binding small molecule to L, provided at least one is a bond linking the NHE-binding small molecule to L. In some embodiments, the NHE-binding small molecule moiety has one of the following structures: In some embodiments, L is a polyalkylene glycol linker. In certain embodiments, L is a polyethylene glycol linker. In some embodiments, X is C(X1). In some embodiments, each Xcis hydrogen. In certain embodiments, X is N. In certain embodiments, each Zais hydrogen. In some embodiments, the compound has the structure of Formula (II): or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein:(a) NHE is a NHE-binding small molecule moiety having the structure of Formula (A-I): wherein:each R1, R2, R3, R5and R9are independently selected from H, halogen, —NR7(CO)R8, —(CO)NR7R8, —SO2—NR7R8, —NR7SO2R8, —NR7R8, —OR7, —SR7, —O(CO)NR7R8, —NR7(CO)OR8, and —NR7SO2NR8, where R7and R8are independently selected from H, C1-6alkyl, —C1-6alkyl-OH or a bond linking the NHE-binding small molecule to L, provided at least one is a bond linking the NHE-binding small molecule to L;R4is selected from H, C1-C7alkyl, or a bond linking the NHE-binding small molecule to L;R6is absent or selected from H and C1-C7alkyl; andAr1 and Ar2 independently represent an aromatic ring or a heteroaromatic ring;(b) Core is a Core moiety having the following structure of Formula (C-I): wherein:W is selected from alkylene, polyalkylene glycol, —C(═O)—NH-(alkylene)-NH—C(═O)—, —C(═O)—NH-(polyalkylene glycol)-NH—C(═O)—, —C(═O)-(alkylene)-C(═O)—, —C(═O)-(polyalkylene glycol)-C(═O)— and cycloalkyl,X is N;Y is C1-6alkylene;Z is selected from —NZa—C(═O)—NZa—, —C(═O)NZa—, —NZa—C(═O)— and heteroaryl;each Zais independently selected from hydrogen and C1-6alkyl; and(c) Lisa bond or linker connecting the Core moiety to the NHE-binding small molecules. In certain embodiments, the NHE-binding small molecule moiety has the following structure: wherein:each R1, R2and R3are independently selected from H, halogen, —NR7(CO)R8, —(CO)NR7R8, —SO2—NR7R8, —NR7SO2R8, —NR7R8, —OR7, —SR7, —O(CO)NR7R8, —NR7(CO)OR8, and —NR7SO2NR8, where R7and R8are independently selected from H, C1-6alkyl, —C1-6alkyl-OH or a bond linking the NHE-binding small molecule to L, provided at least one is a bond linking the NHE-binding small molecule to L. In certain embodiments, the NHE-binding small molecule moiety has one of the following structures: In another embodiment, the NHE inhibitor is: and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, isomers, and tautomers thereof,wherein:Linker is —(CHR13)p—[Y—(CH2)r]5—Z—R13—(CH2)t—Z—;W is independently, at each occurrence, S(O)2, C(O), or —(CH2)m—;Z is independently, at each occurrence, a bond, C(O), or —C(O)NH—;Y is independently, at each occurrence, O, S, NH, N(C1-C3alkyl), or —C(O)NH—;Q is a bond, NH, —C(O)NH—, —NHC(O)NH—, —NHC(O)N(CH3)—, or —NHC(O)NH—(CHR13); m is an integer from 1 to 2; n is an integer from 1 to 4;r and p are independently, at each occurrence, integers from 0 to 8;s is an integer from 0 to 4;t is an integer from 0 to 4;u is an integer from 0 to 2;R1and R2are independently H, C1-C6alkyl, C2-C6alkenyl, C4-C8cycloalkenyl, C2-C6alkynyl, C3-C8cycloalkyl, heterocyclyl, aryl, heteroaryl containing 1-5 heteroatoms selected from the group consisting of N, S, P and O, wherein each alkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl is optionally substituted with one or more halogen, OH, CN, —NO2, OXO, —SR9, —OR9, —NHR9, —NR9R10, —S(O)2N(R9)2—, —S(O)2R9, —C(O)R9, —C(O)OR9, —C(O)NR9R10, —NR9S(O)2R10, —S(O)R9, —S(O)NR9R10, —NR8S(O)R9, C1-C6alkyl, C2-C6alkenyl, C4-C8cycloalkenyl, C2-C6alkynyl, C3-C8cycloalkyl, heterocyclyl, heterocycle, aryl, or heteroaryl; orR1and R2together with the nitrogen to which they are attached can form a heterocyclyl or heteroaryl containing 1-5 heteroatoms selected from the group consisting of N, S, P and O, wherein the heterocyclyl or heteroaryl group is optionally substituted with one or more halogen, OH, CN, —NO2, OXO, —SR9, —OR9, —NHR9, —NR9R10, —S(O)2N(R9)2—, —S(O)2R9, —C(O)R9, —C(O)OR9, —C(O)NR9R10, —NR9S(O)2R10, —S(O)R9, —S(O)NR9R10, —NR9S(O)R10, C1-C6alkyl, C2-C6alkenyl, C4-C8cycloalkenyl, C2-C6alkynyl, C3-C8cycloalkyl, heterocyclyl, heterocycle, aryl, or heteroaryl;R3and R4are independently halogen, OH, CN, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, C1-C6haloalkoxy, or —C(O)NR9R10;R5, R6, R7, and R8are independently H, halogen, OH. CN, —NO2, C1-C6alkyl, C2-C6alkenyl, C4-C8cycloalkenyl, C2-C6alkynyl, C3-C8cycloalkyl, heterocyclyl, aryl, heteroaryl containing 1-5 heteroatoms selected from the group consisting of N, S, P and O, —SR9, —OR9, —NHR9, —NR9R10, —S(O)2N(R9)2—, —S(O)2R9, —C(O)R9, —C(O)OR9, —NR9S(O)2R10, —S(O)R9, —S(O)NR9R10, —NR8S(O)R9;R9and R10are independently H, C1-C6alkyl, C2-C6alkenyl, C4-C8cycloalkenyl, C2-C6alkynyl, C3-C8cycloalkyl, heterocyclyl, aryl, or heteroaryl containing 1-5 heteroatoms selected from the group consisting of N, S, P and OX is a bond, H, N, O, CR11R12, CR11, C, —NHC(O)NH—, or C3-C6cyclolakyl;R11and R12are independently H, C1-C6alkyl, OH, NH2, CN, or NO2;R13is independently, at each occurrence, a bond, H, C1-C6alkyl, C4-C8cycloalkenyl, C3-C8cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each cycloalkenyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl is optionally substituted with one or more R19;R14is independently, at each occurrence, H, C1-C6alkyl, or C1-C6haloalkyl; orR6and R14together with the atoms to which they are attached may combine to form, independently, at each occurrence, 5- to-6 membered heterocyclyl, wherein each C3-C5cycloalkyl, or heterocyclyl is optionally substituted with one or more R19; orR13and R14together with the atoms to which they are attached may combine to form independently, at each occurrence, C3-C5cycloalkyl, heterocyclyl, aryl, or heteroaryl containing 1-5 heteroatoms selected from the group consisting of N, S, P and O, wherein each heterocyclyl or heteroaryl is optionally substituted with one or more R19;R15, R16, R17, and R18are independently, at each occurrence, H, OH, NH2, or C1-C3alkyl, wherein the alkyl is optionally substituted with one or more R19; andR19are independently, at each occurrence, H, OH, NH2, oxo, C1-C6alkyl, C1-C6Hhaloalkyl, C1-C6alkoxy. In an embodiment, the NHE3 inhibitor is a compound according to the foregoing formula provided that:(1) when X is H, n is 1;(2) when X is a bond, O, or CR11R12, n is 2;(3) when n is 3, X is CR11or N;(4) when n is 4 X is C;(5) only one of Q or X is —NHC(O)NH— at the time,(6) R1and R2together with the nitrogen to which they are attached, cannot form a pyrrolidinyl;(7) when R1and R2are methyl, R3and R4are halogen, and R5and R8are H, Linker is not (8) when R1and R2together with the nitrogen to which they are attached form a piperidinyl, R3and R4are halogen, and R5and R8are H, Linker is not or(9) when R1and R2, together with the nitrogen to which they are attached, form 3-aminopiperidin-1-yl, R3and R4are halogen, and R5, R6, R7, and R8are H, Linker is not In an embodiment, the NHE3 compound has a structure according to the following formula: and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, isomers, and tautomers thereof, wherein:Linker is —(CHR8)p—[Y—(CH2)r]s—Z—R8—(CH2)t—Z—;Q is a bond or —NHC(O)NH—;Z is independently, at each occurrence, a bond, C(O), or —C(O)NH—;Y is independently, at each occurrence, O, S, NH, N(C1-C3alkyl), or —C(O)NH—;X is a bond, N, O, CR11R12, CR11, C, or —NHC(O)NH—;n is an integer from 2 to 4;r and p are independently, at each occurrence, integers from 0 to 8;s is an integer from 0 to 4;t is an integer from 0 to 4;u is an integer from 0 to 2;R1and R2are independently halogen, OH, CN, C1-C6alkyl, C1-C6alkoxy, C1-C6haloalkyl, C1-C6haloalkoxy, or —C(O)NR9R10;R3, R4, R5, and R6are independently H, halogen, OH. CN, —NO2, C1-C6alkyl, C2-C6alkenyl, C4-C8cycloalkenyl, C2-C6alkynyl, C3-C8cycloalkyl, heterocyclyl, aryl, heteroaryl containing 1-5 heteroatoms selected from the group consisting of N, S, P and O, —SR9, —OR9, —NHR9, —NR9R10, —S(O)2N(R9)2—, —S(O)2R9, —C(O)R9, —C(O)OR9, —NR9S(O)2R10, —S(O)R9, —S(O)NR9R10, —NR8S(O)R9;R7is independently, at each occurrence, H, C1-C6alkyl, or C1-C6haloalkyl;R8is independently, at each occurrence, a bond, H, C1-C6alkyl, C4-C8cycloalkenyl, C3-C8cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each cycloalkenyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl is optionally substituted with one or more R17; orR7and R8together with the atoms to which they are attached may combine to form independently, at each occurrence, heterocyclyl or heteroaryl containing 1-5 heteroatoms selected from the group consisting of N, S, P and O, wherein each heterocyclyl or heteroaryl is optionally substituted with one or more R17;R9and R10are independently H, C1-C6alkyl, C2-C6alkenyl, C4-C8cycloalkenyl, C2-C6alkynyl, C3-C8cycloalkyl, heterocyclyl, aryl, or heteroaryl containing 1-5 heteroatoms selected from the group consisting of N, S, P and O;R11and R12are independently H, C1-C6alkyl, OH, NH2, CN, or NO2;R13, R14, R15, and R16are independently, at each occurrence, H, OH, NH2, or C1-C3alkyl, wherein the alkyl is optionally substituted with one or more R17; andR17is independently, at each occurrence, H, OH, NH2, oxo, C1-C6alkyl, C1-C6haloalkyl, or C1-C6alkoxy. In an embodiment, the NHE3 inhibitor compound has a structure according to the foregoing formula provided that:(1) when X is a bond, O, or CR11R12, n is 2;(2) when n is 3, X is CR11or N;(3) when n is 4 X is C;(4) only one of Q or X is —NHC(O)NH— at the time;(5) when R1and R2are chloro, Q is —NHC(O)NH—, and R3, R4, R5, and R6are H, Linker is not (6) when R1and R2are chloro, Q is —NHC(O)NH—, and R3, R4, R5, and R6are H, Linker is not In an embodiment, the NHE3 inhibitor compound has a structure according to the following formula:N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[5-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[5-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis(5-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide);N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-(10,17-Dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosane-1,26-diyl)bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[5-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[5-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[5-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-[(3S,3′S)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-methylbenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[5-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[5-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[5-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-2-methylbenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)-4-fluorobenzenesulfonamide];N,N′-[(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(piperidine-1,4-diyl)]bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(piperidine-1,4-diyl)]bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];1,1′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[N-([3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl]sulfonyl)formamide];1,1′-[(3R,3′R)-(7,14-Dioxo-3,18-dioxa-6,8,13,15-tetraazaicosane-1,20-diyl)bis(pyrrolidine-1,3-diyl)]bis[N-([3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl]sulfonyl)formamide];1,1′-(5,12-Dioxo-4,6,11,13-tetraazahexadecane-1,16-diyl)bis[N-([3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl]sulfonyl)piperidine-4-carboxamide];1,1′-(5,12-Dioxo-4,6,11,13-tetraazahexadecane-1,16-diyl)bis[N-([3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl]sulfonyl)piperidine-3-carboxamide];N1,N18-Bis([3-(6,8-Dichloro-2-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl]sulfonyl)-6,13-dioxo-5,7,12,14-tetraazaoctadecanediamide;N,N′-[(3S,3′S)-(6,13-Dioxo-5,7,12,14-tetraazaoctadecanedioyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-2,8-dimethyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(3S,3′S)-(6,13-Di oxo-5,7,12,14-tetraazaoctadecanedioyl)bis(pyrrolidine-1,3-diyl)]bis[3-(6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];1-[2-(2-[(1-[(3-[(S)-6,8-Dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl]phenyl)sulfonyl]piperidin-4-yl)oxy]ethoxy)ethyl]-3-[4-(3-[2-(2-[(1-[(3-[(S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl]phenyl)sulfonyl]piperidin-4-yl)oxy]ethoxy)ethyl]ureido)butyl]urea;1-(2-(2-(((R)-1-((3-((S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl)sulfonyl)pyrrolidin-3-yl)oxy)ethoxy)ethyl)-3-(4-(3-(2-(2-(((S)-1-((3-((S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)phenyl)sulfonyl)pyrrolidin-3-yl)oxy)ethoxy)ethyl)ureido)butyl)urea;1-(2-[2-([(S)-1-[(3-[(S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl]phenyl)sulfonyl]pyrrolidin-3-yl]oxy)ethoxy]ethyl)-3-(4-[3-(2-[2-([(S)-1-[(3-[(S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl]phenyl)sulfonyl]pyrrolidin-3-yl]oxy)ethoxy]ethyl)ureido]butyl)urea;3-[(S)-6,8-Dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl]-N-[(3R,2R)-28-[(3-[(S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl]phenyl)sulfonamido]-2,29-dimethyl-12,19-dioxo-5,8,23,26-tetraoxa-11,13,18,20-tetraazatriacontan-3-yl]benzenesulfonamide;N,N′-(10-Oxo-3,6,14,17-tetraoxa-9,11-diazanonadecane-1,19-diyl)bis[3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N,N′-[(3S,3′S)-(7-Oxo-3,11-dioxa-6,8-diazatridecane-1,13-diyl]bis[pyrrolidine-1,3-diyl))bis(3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide];N1,N18-Bis(1-[(3-[(S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinol in-4-yl]phenyl)sulfonyl]piperidin-4-yl)-6,13-dioxo-5,7,12,14-tetraazaoctadecanediamid;N1,N18-Bis(1-[(3-[(S)-6-chloro-8-cyano-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl]phenyl)sulfonyl]piperidin-4-yl)-6,13-dioxo-5,7,12,14-tetraazaoctadecanediamide; orN1,N18-Bis(1-[(3-[(S)-6-chloro-2,8-dim ethyl-1,2,3,4-tetrahydroisoquinolin-4-yl]phenyl)sulfonyl]piperidin-4-yl)-6,13-dioxo-5,7,12,14-tetraazaoctadecanediamide. In one embodiment of the invention, the NHE3 inhibitor is a compound according to the formula: In one embodiment of the invention, the NHE3 inhibitor is a compound according to the formula: In an embodiment the NHE3 inhibitor is one of the following compounds:1-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-cyano-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-cyano-2,3-dihydro-1H-inden-1-yl]oxy]benzene) sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-m ethoxy-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-methoxy-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;1-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-fluoro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1 S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-fluoro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-cyano-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1 S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-cyano-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene) sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-methoxy-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-methoxy-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino) butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluoro benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy) ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-(trifluoromethyl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-(trifluoromethyl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;1-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-(trifluoromethoxy)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-(trifluoromethoxy)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-methoxy-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-l -[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-m ethoxy-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-fluoro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-fluoro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino) butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-methyl benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy) ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-(trifluoromethyl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-(trifluoromethyl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-carbamoyl]amino)butyl]urea;1-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-(trifluoromethoxy)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-(trifluoromethoxy)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-methoxy-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-methoxy-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methyl benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea dihydrochloride;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-(dimethylamino)-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-(dimethylamino)-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-methyl-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-methyl-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-methyl-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-methyl-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-methyl-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-methyl-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-6-chloro-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-6-chloro-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-cyano-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-cyano-2-[(3R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfon amido]ethoxy]ethoxy) ethyl]carb amoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1 S,2 S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1 S,2 S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[[(3S)-1-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]ethoxy)ethyl]-1-[4-([[2-(2-[[(3S)-1-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro -1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]ethoxy)ethyl]carbamoyl]amino) butyl]urea;3-[2-(2-[[(3R)-1-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]ethoxy)ethyl]-1-[4-([[2-(2-[[(3R)-1-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene) sulfonyl]pyrrolidin-3-yl]methoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[[(3S)-1-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]ethoxy)ethyl]-l -[4-([[2-(2-[[(3 S)-1-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[[(3R)-1-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]ethoxy)ethyl]-1-[4-([[2-(2-[[(3R)-1-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[(4-[[(3S)-1-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]pyridin-2-yl)methyl]-1-[4-([[(4-[[(3 S)-1-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]pyridin-2-yl)methyl]carbamoyl]amino)butyl]urea;3-[(4-[[(3R)-1-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]pyridin-2-yl)methyl]-1-[4-([[(4-[[(3R)-1-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]pyridin-2-yl)methyl]carbamoyl]amino)butyl]urea;3-[(4-[[(3S)-1-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]pyridin-2-yl)methyl]-1-[4-([[(4-[[(3 S)-1-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]pyridin-2-yl)methyl]carbamoyl]amino)butyl]urea;3-[(4-[[(3R)-1-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]pyridin-2-yl)methyl]-1-[4-([[(4-[[(3R)-1-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonyl]pyrrolidin-3-yl]methoxy]pyridin-2-yl)methyl]carbamoyl]amino)butyl]urea;3-(2-[2-[(3S)-3-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene) sulfonamido]pyrrolidin-1-yl]ethoxy]ethyl)-1-(4-[[(2-[2-[(3S)-3-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]pyrrolidin-1-yl]ethoxy]ethyl) carbamoyl]amino]butyl)urea;3-(2-[2-[(3R)-3-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]pyrrolidin-1-yl]ethoxy]ethyl)-1-(4-[[(2-[2-[(3R)-3-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]pyrrolidin-1-yl]ethoxy]ethyl)carbamoyl]amino]butyl)urea;3-(2-[2-[(3 S)-3-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]pyrrolidin-1-yl]ethoxy]ethyl)-1-(4-[[(2-[2-[(3S)-3-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]pyrrolidin-1-yl]ethoxy]ethyl)carbamoyl]amino]butyl)urea;3-(2-[2-[(3R)-3-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]pyrrolidin-1-yl]ethoxy]ethyl)-1-(4-[[(2-[2-[(3R)-3-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]pyrrolidin-1-yl]ethoxy]ethyl)carbamoyl]amino]butyl)urea;1-([1-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene) sulfonamido]ethoxy]ethoxy)ethyl]-1H-1,2,3-triazol-4-yl]methyl)-3-(4-[[([1-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy) ethyl]-1H-1,2,3-triazol-4-yl]methyl)carbamoyl]amino]butyl)urea;(2R,3S,4R,5S)—N1,N6-Bis([1-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1H-1,2,3-triazol-4-yl]methyl)-2,3,4,5-tetrahydroxyhexanediamide;3-[(1-[4-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]butyl]-1H-1,2,3-triazol-4-yl)methyl]-1-[4-([[(1-[4-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]butyl]-1H-1,2,3-triazol-4-yl)methyl]carbamoyl]amino)butyl]urea;3-[(1-[6-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]hexyl]-1H-1,2,3-triazol-4-yl)methyl]-1-[4-([[(1-[6-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]hexyl]-1H-1,2,3-triazol-4-yl)methyl]carbamoyl]amino)butyl]urea;(4R,4aS,8S,8aR)—N4,N8-Bis([1-(4-[4-((1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yloxy)phenyl sulfonamide]butyl)-1H-1,2,3-triazol-4-yl]methyl)-2,2,6,6-tetramethyl-tetrahydro-[1,3]dioxino[5,4-d][1,3]dioxine-4,8-dicarboxamide;(4R,4aS,8S,8aR)—N4,N8-Bis([1-(6-[4-((1S,2S)-2-[(3R)-3-amino piperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yloxy)phenylsulfonamido]hexyl)-1H-1,2,3-triazol-4-yl]methyl)-2,2,6,6-tetramethyl-tetrahydro-[1,3]dioxino[5,4-d][1,3]dioxine-4,8-dicarboxamide;3-[8-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]octyl]-1-[4-[([8-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene) sulfonamido]octyl]carbamoyl)amino]butyl]urea;3-[8-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene) sulfonamido]octyl]-1-[4-[([8-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]octyl]carbamoyl)amino]butyl]urea;3-[8-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]octyl]-1-[4-[([8-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]benzene) sulfonamido]octyl]carbamoyl)amino]butyl]urea;3-[8-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]octyl]-1-[4-[([8-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethyl amino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene) sulfonamido]octyl]carbamoyl)amino]butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-[(2R)-2-methylpiperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-[(2R)-2-methylpiperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-[(2S)-2-methylpiperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-[(2S)-2-methylpiperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[2-Azabicyclo[2.2.1]heptan-2-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[2-azabicyclo[2.2.1]heptan-2-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea; l-[2-(2-[2-[(4-[[(1S,2S)-2-[2-Azabicyclo[2.2.2]octan-2-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[2-azabicyclo[2.2.2]octan-2-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[8-azabicyclo[3.2.1]octan-8-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[8-azabicyclo[3.2.1]octan-8-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;1-[2-(2-[2-[(4-[[(1S,2S)-2-[9-Azabicyclo[3.3.1]nonan-9-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[9-azabicyclo[3.3.1]nonan-9-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(4-methylpiperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(4-methylpiperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(4-methylpiperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-l -[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(4-methylpiperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-(4-Acetylpiperazin-1-yl)-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-(4-acetylpiperazin-1-yl)-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-(4-Acetylpiperazin-1-yl)-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-l -[4-([[2-(2-[2-[(4-[[(1S,2S)-2-(4-acetylpiperazin-1-yl)-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;4-[(1S,2S)-4,6-dichloro-1-[4-[(2-[2-[2-([[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-[4-(dimethylcarbamoyl)piperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene) sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]carbamoyl]amino)ethoxy]ethoxy]ethyl)sulfamoyl]phenoxy]-2,3-dihydro-1H-inden-2-yl]-N,N-dimethylpiperazine-1-carboxamide;4-[(1S,2S)-4,6-dichloro-1-[4-[(2-[2-[2-([[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-[4-(dimethylcarbamoyl)piperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]carbamoyl]amino)ethoxy]ethoxy]ethyl)sulfamoyl]-2-methylphenoxy]-2,3-dihydro-1H-inden-2-yl]-N,N-dimethylpiperazine-1-carboxamide;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-[(3R)-3-[methyl(propan-2-yl)amino]piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene) sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-[(3R)-3-[methyl(propan-2-yl)amino]piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3,5-dimethyl benzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3,5-dimethylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea; hydrochloride;l-[2-(2-[2-[(3-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-2,4-dimethylbenzene)sulfonamido]ethoxy]ethoxy) ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3,5-dimethylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2,5-dimethylbenzene) sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2,5-dimethylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-2,5-dimethylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-2,5-dimethylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;1-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluoro-5-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluoro-5-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-carbamoyl]amino)butyl]urea; hydrochloride;1-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluoro-5-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-fluoro-5-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3,5-difluorobenzene) sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-3,5-difluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;4-([(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[26-([4-([(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy)-3,5-difluorophenyl]sulfonamido)-10,17-dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosyl]-3,5-difluorobenzenesulfonamide;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-5-fluoro-2-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1 S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-5-fluoro-2-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-5-fluoro-2-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-5-fluoro-2-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2-fluoro-5-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1 S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2-fluoro-5-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-2-fluoro-5-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-2-fluoro-5-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;1-(2-[2-[(3S)-3-[(4-[[(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]-2-oxopyrrolidin-1-yl]ethoxy]ethyl)-3-(4-[[(2-[2-[(3 S)-3-[(4-[[(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]-2-oxopyrrolidin-1-yl]ethoxy]ethyl)carbamoyl]amino]butyl)urea;1-(2-[2-[(3S)-3-[(4-[[(1S,2S)-6-Chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]-2-oxopyrrolidin-1-yl]ethoxy]ethyl)-3-(4-[[(2-[2-[(3 S)-3-[(4-[[(1S,2S)-6-chloro-4-cyano-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy]-3-methylbenzene)sulfonamido]-2-oxopyrrolidin-1-yl]ethoxy]ethyl)carbamoyl]amino]butyl)urea;3-[2-(2-[[(3R)-1-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonyl]pyrrolidin-3-yl]oxy]ethoxy)ethyl]-1-[4-([[2-(2-[[(3R)-1-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonyl]pyrrolidin-3-yl]oxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[[(3 S)-1-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonyl]pyrrolidin-3-yl]oxy]ethoxy)ethyl]-1-[4-([[2-(2-[[(3S)-1-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonyl]pyrrolidin-3-yl]oxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-[2-([1-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonyl]piperidin-4-yl]oxy)ethoxy]ethyl]-1-[4-[([2-[2-([1-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonyl]piperidin-4-yl]oxy)ethoxy]ethyl]carbamoyl)amino]butyl]urea;1-(2-[2-[(2S)-2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]propoxy]ethoxy]ethyl)-3-(4-[[(2-[2-[(2S)-2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]propoxy]ethoxy]ethyl)carbamoyl]amino]butyl)urea; hydrochloride;3-(2-[2-[(2R)-2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]propoxy]ethoxy]ethyl)-1-(4-[[(2-[2-[(2R)-2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]propoxy]ethoxy]ethyl)carbamoyl]amino]butyl)urea;3-(2-[2-[(2S)-2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]-3-methylbutoxy]ethoxy]ethyl)-1-(4-[[(2-[2-[(2S)-2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene) sulfonamido]-3-methylbutoxy]ethoxy]ethyl)carbamoyl]amino]butyl)urea dihydrochloride;3-(2-[2-[(2R)-2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]-3-methylbutoxy]ethoxy]ethyl)-1-(4-[[(2-[2-[(2R)-2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]-3-methylbutoxy]ethoxy]ethyl)carbamoyl]amino]butyl)urea;1-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]-2-methylpropoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]benzene)sulfonamido]-2-methylpropoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea; hydrochloride;1-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2-methoxybenzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2-methoxybenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;3-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]-1-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2-methylbenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea; l-[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]-3-[4-([[2-(2-[2-[(4-[[(1S,2S)-2-[(3R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy]-2-fluorobenzene)sulfonamido]ethoxy]ethoxy)ethyl]carbamoyl]amino)butyl]urea;4-([(1S,2S)-2-[(R)-3-Aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy)-N-[26-([4-([(1S,2S)-2-[(R)-3-aminopiperidin-1-yl]-4,6-dichloro-2,3-dihydro-1H-inden-1-yl]oxy)-2-chlorophenyl]sulfonamido)-10,17-dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosyl]-2-chlorobenzenesulfonamide;4-([(1S,2S)-4,6-Dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1S,2S)-4,6-dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-3-fluorophenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]-3-fluorobenzenesulfonamide; tetra(trifluoroacetate);4-([(1S,2S)-4,6-Dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-4,6-dichloro-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-3-fluorophenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]-3-fluorobenzenesulfonamide; tetra(trifluoroacetate);4-([(1 S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide; tetra(trifluoroacetate);4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1 S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide; tetra(trifluoroacetate);4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-[(4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-3-fluorophenyl)sulfonamide)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]-3-fluorobenzenesulfonamide; tetra(trifluoroacetate);4-([(1 S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-[(4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-3-fluorophenyl)sulfonamide)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]-3-fluorobenzenesulfonamide; tetra(trifluoroacetate);4-([(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy)-3-fluorophenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl)-3-fluorobenzenesulfonamide; tetra(trifluoroacetate);4-([(1S,2S)-4,6-Dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1S,2S)-4,6-dichloro-2-(dimethylamino)-2,3-dihydro-1H-inden-1-yl]oxy)-3-fluorophenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl)-3-fluorobenzenesulfonamide; tetra(trifluoroacetate);4-([(1S,2S)-6-Chloro-2-[(R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1 S,2 S)-6-chloro-2-[(R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy)-3-methylphenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]-3-methylbenzenesulfonamide; tetra(trifluoroacetate);4-([(1S,2S)-6-Chloro-2-[(R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-6-chloro-2-[(R)-3-(dimethylamino)piperidin-1-yl]-4-methyl-2,3-dihydro-1H-inden-1-yl]oxy)-3-methylphenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]-3-methylbenzenesulfonamide; tetra(trifluoroacetate);4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[1-(18-[4-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)piperidin-1-yl]-6,13,18-trioxo-5,7,12,14-tetraazaoctadecanoyl)piperidin-4-yl]benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(14-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-4,11,14-trioxo-3,5,10,12-tetraazatetradecanoyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1 S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-[(2S,13S)-14-[(S)-3-([4-([(1S,2 S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-2,13-dimethyl-4,11,14-trioxo-3,5,10,12-tetraazatetradecanoyl]pyrrolidin-3-yl]benzenesulfonamide;N1,N14-bis(2-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-2-oxoethyl)-4,11-dioxo-3,5,10,12-tetraazatetradecanedi ami de;N1,N14-bis(2-[(R)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-2-oxoethyl)-4,11-dioxo-3,5,10,12-tetraazatetradecanedi ami de;N1,N18-Bis(1-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonyl)piperidin-4-yl)-6,13-dioxo-5,7,12,14-tetraazaoctadecanediamide;4-([(1S,2S)-6-Chloro-4-cyano-2-[(R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N-[26-([4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)-10,17-dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosyl]benzenesulfonamide;4-([(1 S,2S)-6-Chloro-4-cyano-2-[(S)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N-[26-([4-([(1S,2S)-6-chloro-4-cyano-2-[(S)-3-(dimethylamino)piperidin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)-10,17-dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosyl]benzenesulfonamide;4-([(1 S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[1-(20-[4-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamide]piperidin-1-yl)-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl]piperidin-4-yl)benzenesulfonamide;N1,N18-Bis([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonyl)-6,13-dioxo-5,7,12,14-tetraazaoctadecanediamide;N-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonyl)-1-[16-(4-[([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonyl)carbamoyl]piperidin-1-yl)-5,12-dioxo-4,6,11,13-tetraazahexadecyl]piperidine-4-carboxamide;4-([(1S,2S)-6-chloro-4-cyano-2-(1,4-diazepan-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-(1,4-diazepan-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-chloro-4-cyano-2-(1,4-diazepan-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1 S,2S)-6-chloro-4-cyano-2-(1,4-diazepan-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1 S,2 S)-6-chloro-4-cyano-2-(4-methyl -1,4-diazepan-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-(4-methyl-1,4-diazepan-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1 S,2 S)-6-chloro-4-cyano-2-(4-methyl -1,4-diazepan-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1 S,2S)-6-chloro-4-cyano-2-(4-methyl-1,4-diazepan-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-2-[(1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl]-6-chloro-4-cyano-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-2-[(1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl]-6-chloro-4-cyano-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-2-[(1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl]-6-chloro-4-cyano-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1S,2S)-2-[(1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl]-6-chloro-4-cyano-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-chloro-4-cyano-2-[(S)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1 S,2S)-6-chloro-4-cyano-2-[(S)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-chloro-4-cyano-2-[(S)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-[(S)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-chloro-4-cyano-2-[(3S,5R)-3,5-dimethylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-[(3S,5R)-3,5-dimethylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-chloro-4-cyano-2-[(3S,5R)-3,5-dimethylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(20-[(R)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-[(3S,5R)-3,5-dimethylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1 S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)-2-oxopiperidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)-2-oxopiperidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[2-(2-[2-(3-[(lr,4r)-4-(3-[2-(2-[2-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)ethoxy]ethoxy)ethyl]ureido)cyclohexyl]ureido)ethoxy]ethoxy)ethyl]benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(18-[(R)-3-([4-([(1 S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-6,13,18-trioxo-5,7,12,14-tetraazaoctadecanoyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)benzenesulfonamide;N-(2-[2-(2-Aminoethoxy)ethoxy]ethyl)-4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)benzenesulfonamide;N-[1-(4-Aminobutanoyl)piperidin-4-yl]-4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-(3-oxo-7,10-dioxa-2,4-diazadodecan-12-yl)benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-(1-[4-(3-methylureido)butanoyl]piperidin-4-yl)benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[(2S,3R,4S,5R)-1,3,4,5,6-pentahydroxyhexan-2-yl]benzenesulfonamide;4-([4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)-N-[(2S,3R,4S,5R)-1,3,4,5,6-pentahydroxyhexan-2-yl]piperidine-1-carboxamide;4-(3-[4-([4-([(1S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)-4-oxobutyl]ureido)-N-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonyl)butanamide;4-([(1 S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[1-(4-[3-(4-[4-([4-([(1 S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)piperidin-1-yl]-4-oxobutyl)ureido]butanoyl)piperidin-4-yl]benzenesulfonamide;4-([(1 S,2S)-6-Chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[19-([4-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)-10-oxo-3,6,14,17-tetraoxa-9,11-diazanonadecyl]benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-amido-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[26-([4-([(1S,2S)-6-chloro-4-amido-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)-10,17-dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosyl]benzenesulfonamide;4-([(1S,2S)-4-Cyano-6-methyl-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-N-[26-([4-([(1S,2S)-4-cyano-6-methyl-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)-10,17-dioxo-3,6,21,24-tetraoxa-9,11,16,18-tetraazahexacosyl]benzenesulfonamide;1,1′-(Butane-1,4-diyl)bis[3-(4-[6-([(1 S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-3,4-dihydroisoquinolin-2(1H)-yl]-4-oxobutyl)urea];1,1′-(Butane-1,4-diyl)bis[3-(4-[7-([(1 S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-3,4-dihydroisoquinolin-2(1H)-yl]-4-oxobutyl)urea];N,N′-(6,14-Dioxo-10-oxa-5,7,13,15-tetraazanonadecane-1,19-diyl)bis[6-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-3,4-dihydroisoquinoline-2(1H)-carboxamide];N,N′-(6,14-Dioxo-10-oxa-5,7,13,15-tetraazanonadecane-1,19-diyl)bis[7-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-3,4-dihydroisoquinoline-2(1H)-carboxamide];4-([(1S,2S)-6-Chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(18-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-6,13,18-trioxo-5,7,12,14-tetraazaoctadecanoyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(R)-1-(18-[(R)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-6,13,18-trioxo-5,7,12,14-tetraazaoctadecanoyl)pyrrolidin-3-yl]benzenesulfonamide;4-([(1S,2S)-6-Chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N-[1-(18-[4-([4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)piperidin-1-yl]-6,13,18-trioxo-5,7,12,14-tetraazaoctadecanoyl)piperidin-4-yl]benzenesulfonamide;N1,N14-Bis(2-[(S)-3-([4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-2-oxoethyl)-4,11-dioxo-3,5,10,12-tetraazatetradecanediamide;4-([(1S,2S)-6-Chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N-[l-(20-[4-([4-([(1S,2S)-6-chloro-4-cyano-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)piperidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)piperidin-4-yl]benzenesulfonamide;4-([(1S,2S)-4,6-Dichloro-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)-N—[(S)-1-(20-[(S)-3-([4-([(1S,2S)-4,6-dichloro-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-7,14-dioxo-3,18-dioxa-6,8,13,15-tetraazaicosyl)pyrrolidin-3-yl]benzenesulfonamide;N1,N14-Bis(2-[(S)-3-([4-([(1S,2S)-4,6-dichloro-2-[(R)-3-methylpiperazin-1-yl]-2,3-dihydro-1H-inden-1-yl]oxy)phenyl]sulfonamido)pyrrolidin-1-yl]-2-oxoethyl)-4,11-dioxo-3,5,10,12-tetraazatetradecanediamide;1,1′-(Butane-1,4-diyl)bis(3-[2-(2-[6-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-1-oxoisoindolin-2-yl]ethoxy)ethyl]urea); and1,1′-(Butane-1,4-diyl)bis(3-[2-(2-[5-([(1S,2S)-6-chloro-4-cyano-2-(piperazin-1-yl)-2,3-dihydro-1H-inden-1-yl]oxy)-1-oxoisoindolin-2-yl]ethoxy)ethyl]urea). EXAMPLES The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention. For example, the synthesis of non-exemplified compounds may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds described herein. Example 1 (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-10-(Methoxycarbonyl)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (1-9) Synthesis of (4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((Benzyloxy)carbonyl)-4,4,6a,6b,8a,11,14b-heptamethyl-14-oxo-1,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-octadecahydropicene-3-carboxylic Acid (1-2) Into a 1-L pressure tank reactor (10 atm) purged and maintained with an inert atmosphere of CO, was placed benzyl (2S,4aS,6aS,6bR,8aR,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-10-(((trifluoromethyl)sulfonyl)oxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,12,12a,12b,13,14b-octadecahydropicene-2-carboxylate 1-1 (prepared according to the method described in US Patent application publication no. 20160151387) (11 g, 15.92 mmol, 1.00 equiv), tetrakis(triphenylphosphine)palladium(0) (4 g, 3.46 mmol, 0.20 equiv), THF (250 mL), and water (150 mL). The resulting solution was stirred for 2 days at 50° C. The resulting solution was extracted with CH2Cl2(3×150 mL) and the organic layers combined. The resulting mixture was washed with brine (3×150 mL). The mixture was dried over anhydrous Na2SO4and concentrated under vacuum. The residue was applied onto a silica gel column with EtOAc/petroleum ether (1:10) (20 mL). The crude product was purified by flash-prep-HPLC with the following conditions (CombiFlash-1)—Column: C18silica gel; mobile phase: MeCN:water=100:0; detector: UV 254 nm. 1 L product was obtained. This resulted in 6.5 g (69.6%) of 1-2 as a light yellow solid. Synthesis of 2-Benzyl 10-((2-(trimethylsilyl)ethoxy)methyl) (2S,4aS,6aS,6bR,8aR,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,12,12a,12b,13,14b-octadecahydropicene-2,10-dicarboxylate (1-3) TEA (3 mL, 5.80 equiv) was added dropwise with stirring at 0° C. to 1-2 (3 g, 5.11 mmol, 1 equiv) and DMAP (102.4 mg, 0.84 mmol, 0.10 equiv) in DMF (30 mL). This was followed by the addition of 2-(trimethylsilyl)ethoxymethyl chloride (4.2 mL, 4.8 equiv) dropwise with stirring at 0° C. The reaction was stirred for 1.5 h at rt and then quenched by the addition of aq. K2CO3(50 mL). The mixture was diluted with CH2Cl2(250 mL), washed with brine (3×150 mL), dried (Na2SO4) and concentrated under vacuum. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:10). This resulted in 3.5 g (95.5%) of 1-3 as a light-yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-Heptamethyl-13-oxo-10-(((2-(trimethylsilyl)ethoxy)methoxy)carbonyl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (1-4) Into a 300-mL pressure tank reactor (40 atm) purged and maintained with an inert atmosphere of hydrogen, was placed 1-3 (6.6 g, 9.20 mmol, 1.00 equiv), Pd/C (1.32 g, 0.20 equiv), acetone (150 mL). The reaction was stirred overnight at 50° C. and then concentrated under vacuum. This resulted in 4.2 g (73%) of 1-4 as a white solid. Synthesis of 2-Benzyl 10-((2-(trimethylsilyl)ethoxy)methyl) (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,10-dicarboxylate (1-5) 1-4 (5.25 g, 8.35 mmol, 1 equiv), cesium carbonate (4.1 g, 12.58 mmol, 1.5 equiv) and benzyl bromide (2.86 g, 16.72 mmol, 2 equiv) in DMF (70 mL) were stirred for 2 h at 60° C. The reaction was diluted with water (250 mL) and extracted with CH2Cl2(2×100 mL). The organic layers were combined, dried (Na2SO4) and concentrated. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:10). This resulted in 6 g (99%) of 1-5 as an off-white solid. Synthesis of (3S,4aS,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((Benzyloxy)carbonyl)-4,4,6a,6b,8a,11,14b-heptamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-3-carboxylic Acid (1-6) Hydrogen chloride (4M in dioxane, 30 mL) was added to 1-5 (5.6 g, 7.79 mmol, 1 equiv) in THF (40 mL) and the reaction stirred for 2 h at 60° C. The solution was adjusted to pH=3 with aqueous sodium bicarbonate (sat.). The mixture was extracted with CH2Cl2(3×100 mL). The organic layers were combined, washed with brine (2×150 mL), dried (Na2SO4) and concentrated. The residue was purified by Flash-prep-HPLC with the following conditions (CombiFlash-1) -Column: C18silica gel; mobile phase: MeCN:water=100:0; detector: UV 254 nm. This resulted in 3.1 g (68%, 97% purity) of 1-6 as a white solid. MS (ES, m/z): [M+H]+=589.4;1H-NMR (400 MHz, Chloroform-d): δ 0.76 (s, 4H), 0.91 (s, 5H), 1.01-1.21 (m, 12H), 1.24-1.49 (m, 9H), 1.56-1.75 (m, 4H), 1.83 (td, J=13.6, 4.6 Hz, 1H), 1.91-2.10 (m, 5H), 2.23 (d, J=8.2 Hz, 1H), 2.37 (s, 1H), 2.86 (d, J=13.2 Hz, 1H), 5.11 (d, J=12.0 Hz, 1H), 5.23 (d, J=12.4 Hz, 1H), 5.57 (s, 1H), 7.30-7.45 (m, 5H). Synthesis of Benzyl (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-10-(chlorocarbonyl)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (1-7) Oxalyl chloride (0.144 mL, 1.70 mmol) was added dropwise to 1-6 (0.50 g, 0.85 mmol) and DMF (1 drop) in CH2Cl2(50 mL) at rt. The mixture was stirred at rt for 1 hour and then evaporated to dryness. The material was used in the following steps without purification. Synthesis of 2-Benzyl 10-methyl (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,10-dicarboxylate (1-8) 1-7 (200 mg, 0.33 mmol, 1 equiv) and TEA (0.274 mL, 6 equiv) in MeOH (20 mL) were stirred overnight at rt. The reaction was concentrated under vacuum. This resulted in 198 mg (100%) of 1-8 as a light-yellow crude solid. Synthesis of (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-10-(Methoxycarbonyl)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (1-9) 1-8 (198 mg, 0.33 mmol, 1.00 equiv) and Pd/C (20 mg) in MeOH (40 mL) were placed under a hydrogen atmosphere and stirred for 1 h at rt. The reaction was filtered and concentrated under vacuum. The crude product (200 mg) was purified by prep-HPLC with the following conditions—Column: XBridge Prep C18 OBD, 190*150 mm, 5 μm; mobile phase: water (10 mM NH4HCO3+0.1% NH4OH) and CH3CN (50.0% CH3CN up to 62.0% in 7 min); detector: UV 254/220 nm. This resulted in 111.8 mg (66%) of 1-9 as a light yellow solid. MS (ES, m/z): [M+H]+=513.60;1H NMR (400 MHz, Chloroform-d) δ 0.76 (d, J=11.2 Hz, 1H), 0.84 (s, 3H), 0.88-0.96 (m, 4H), 0.99-1.08 (m, 4H), 1.18 (s, 3H), 1.19-1.28 (m, 7H), 1.31-1.39 (m, 4H), 1.40-1.48 (m, 4H), 1.50-1.59 (m, 1H), 1.60-1.72 (m, 3H), 1.79-1.89 (m, 1H), 1.91-2.09 (m, 4H), 2.15-2.25 (m, 2H), 2.37 (s, 1H), 2.82 (dt, J=10.4, 3.2 Hz, 1H), 3.65 (s, 3H), 5.70 (s, 1H), 9.89 (s, 1H). Example 2 (2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-10-(Carboxymethoxy)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (78-2) Synthesis of Benzhydryl (2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-10-(prop-2-yn-1-yloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (76-2) 76-1 (prepared as described inBioorg. Med Chem.2010, 18, 433-454) (1.674 g, 2.63 mmol, 1 equiv), NaHMDS (2.63 mL, 2 equiv), 3-bromoprop-1-yne (0.45 mL, 2 equiv), and tetrabutylammonium iodide (486 mg, 1.32 mmol, 0.5 equiv) in THF (2 mL) were stirred overnight at rt. The reaction was diluted with water (30 mL), extracted with DCM (2×50 mL). The extract was dried (Na2SO4) and concentrated under vacuum. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:10). This resulted in 1.395 g (79%) of 76-2 as a light yellow solid. Synthesis of 2-(((3S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((Benzhydryloxy)carbonyl)-4,4,6a,6b,8a,11,14b-heptamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicen-3-yl)oxy)acetic Acid (78-1) Ruthenium(III) chloride hydrate (20 mg, 0.10 mmol, 6.8 equiv), 76-2 (100 mg, 0.15 mmol, 1 equiv), sodium periodate (150 mg, 0.70 mmol, 5 equiv) in THF (4 mL) and water (1 mL) were stirred for 2h at rt. The reaction was diluted with water (20 mL) and extracted with DCM (2×50 mL). The extract was dried (Na2SO4) and concentrated under vacuum. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:5). This resulted in 80 mg (78%) of 78-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-10-(Carboxymethoxy)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (78-2) 78-1 (148 mg, 0.21 mmol, 1 equiv) and Pd/C (100 mg) in EtOAc (15 mL) were placed under a hydrogen atmosphere (1 atm) and stirred overnight at rt. The reaction was filtered and concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% NH4OH) and CH3CN (10.0% CH3CN up to 40.0% in 8 min); detector: UV 254 nm. This resulted in 31.1 mg (28%) of 78-2 as a white solid. MS (ES, m/z) [M+H]+=529.40;1H NMR (300 MHz, MeOH-d4, ppm) δ 0.78-0.88 (m, 7H), 0.94-1.08 (m, 5H), 1.16-1.18 (m, 9H), 1.27 (d, J=13.5 Hz, 1H), 1.39-1.51 (m, 8H), 1.56-2.12 (m, 8H), 2.13-2.26 (m, 2H), 2.47 (s, 1H), 2.76 (dt, J=13.5, 3.5 Hz, 1H), 2.94 (dd, J=11.7, 4.2 Hz, 1H), 3.93 (d, J=15.6 Hz, 1H), 4.05 (d, J=15.6 Hz, 1H), 5.61 (s, 1H). Example 3 (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-Hydroxy-9-(hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (121-2) Synthesis of Benzyl (2S,4aS,6aS,6bR,8aR,9S,12aS,12bR,14bR,E)-10-(acetoxyimino)-9-(acetoxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (120-2) 120-1, prepared from (2S,4aS,6aS,6bR,8aR,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-10,13-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (as described inBioorg. Med Chem.2010, 18, 433-454) (1.0 g, 1.7 mmol, 1 equiv), sodium tetrachloropalladate(II) (0.76 g, 2.04 mmol, 1.2 equiv) and sodium acetate (0.21 g, 1.36 mmol, 0.8 equiv) in acetic acid (100 mL) were stirred at rt for 72h. The reaction was poured onto ice and the resulting precipitate collected by filtration and dried under vacuum. DCM (120 mL), acetic anhydride (0.435 g, 3.24 mmol, 1.8 equiv), TEA (0.364 g, 2.72 mmol, 1.6 equiv) and DMAP (6 mg, 0.02 equiv) were added and the mixture stirred for 1 h at rt. The reaction was washed with water (1×300 mL), dried (Na2SO4) and concentrated under vacuum. Pyridine (0.6 mL) and THF (100 mL) were added and the mixture stirred at rt for 15 min. The reaction was cooled to −78° C. in a dry ice/acetone bath and lead tetraacetate (4.9 g, 8.5 mmol, 5 equiv) in acetic acid (100 mL) were added slowly. The mixture was allowed to warm to rt and stirred at rt for 16h. A solution of sodium borohydride (60 mg) in 1 N aqueous NaOH solution (50 mL) was added and stirring was continued for 10 min. The reaction was filtered through celite and extracted with DCM (300 mL). The extract was washed with aqueous sat. NaHCO3(3×300 mL) and brine (2×300 mL), dried (Na2SO4) and concentrated under vacuum. This resulted in 1.03 g (88%, crude) of 120-2 as a light yellow solid. Synthesis of Benzyl (2S,4aS,6aS,6bR,8aR,9S,12aS,12bR,14bR,E)-10-(hydroxyimino)-9-(hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (120-3) 120-2 (1.03 g, 1.53 mmol, 1 equiv) and sodium carbonate (820 mg, 7.74 mmol, 5 equiv) in MeOH (120 mL) were stirred for 16h at rt. The reaction was concentrated under vacuum and the residue dissolved in DCM (200 mL). The mixture was washed with aq. sat. sodium bicarbonate (2×200 mL) and brine (1×200 mL) and concentrated under vacuum. This resulted in 1.1 g (crude) of 120-3 as a yellow solid. Synthesis of Benzyl (2S,4aS,6aS,6bR,8aR,9R,12aS,12bR,14bR)-9-(hydroxymethyl)-2,4a,6a, 6b,9,12a-hexamethyl-10,13-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (120-4) 120-3 (1.1 g, 1.87 mmol, 1 equiv) in THF (70 mL) was added dropwise to ammonium acetate (3.88 g, 27 equiv) and titanium(III) chloride (4 mL) in water (80 mL). The reaction was stirred overnight at rt and then partially concentrated under vacuum. The remaining solution was extracted with DCM (200 mL), washed with aq. sat. sodium bicarbonate (1×200 mL) and brine (1×200 mL), dried (Na2SO4) and concentrated under vacuum. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:1). This resulted in 190 mg (18%) of 120-4 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9R,12aS,12bR,14bR)-9-(Hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-10,13-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (120-5) 120-4 (100 mg, 0.17 mmol) and Pd/C (20 mg) in EtOAc (15 mL) were placed under a hydrogen atmosphere (1 atm) and stirred for 1 h at rt. The reaction was filtered and concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 mm, 19*150 mm; mobile phase: water (0.05% TFA) and MeCN (20.0% MeCN up to 30.0% in 10 min); detector: UV 254 nm. This resulted in 5.3 mg (6%) of 120-5 as a white solid. MS (ES, m/z): [M+H]+=485.30;1H NMR (400 MHz, MeOH-d4, ppm): δ 0.86 (s, 3H), 0.96 (s, 3H), 1.01-1.09 (m, 1H), 1.13 (s, 3H), 1.19-1.42 (m, 9H), 1.46-1.73 (m, 8H), 1.80-2.08 (m, 5H), 2.12-2.29 (m, 1H), 2.19-2.48 (m, 2H), 2.48-2.64 (m, 1H), 2.66 (s, 1H), 2.77-2.98 (m, 1H), 3.35-3.42 (m, 2H), 3.65 (d, J=10.8 Hz, 1H), 5.75 (s, 1H). Synthesis of Benzyl (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-hydroxy-9-(hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (121-1) 120-4 (150 mg, 0.26 mmol, 1 equiv) and sodium borohydride (40 mg, 1.06 mmol, 4 equiv) in MeOH (20 mL) were stirred for 1 h at rt. The reaction was quenched by the addition of water (5 mL) and the mixture concentrated under vacuum. The residue was a diluted with DCM, washed with water and brine, dried (Na2SO4) and concentrated. This resulted in 140 mg (93%) of 121-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-Hydroxy-9-(hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (121-2) 121-1 (180 mg, 0.31 mmol) and Pd/C (36 mg) in EtOAc (15 mL) were placed under a hydrogen atmosphere (1 atm) and stirred for 1 h at rt. The reaction was filtered and concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and MeCN (42.0% MeCN up to 57.0% in 8 min); detector: UV 254 nm. This resulted in 37.9 mg (25%) of 121-2 as a white solid. MS (ES, m/z): [M+H]+=487.25;1H NMR (400 MHz, MeOH-d4, ppm) δ 0.72 (s, 3H), 0.86 (s, 3H), 0.97-1.12 (m, 2H), 1.14-1.33 (m, 11H), 1.37-1.65 (m, 10H), 1.67-1.79 (m, 2H), 1.80-2.03 (m, 4H), 2.10-2.27 (m, 2H), 2.51 (s, 1H), 2.73 (dt, J=13.5, 3.6 Hz, 1H), 3.29-3.32 (m, 1H), 3.56 (d, J=11.0 Hz, 1H), 3.64 (dd, J=11.8, 4.7 Hz, 1H), 5.60 (s, 1H). Example 4 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic Acid (122-3) Synthesis of Benzyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-formyl-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (122-1) 121-1 (300 mg, 0.52 mmol, 1 equiv), pH=8.6 phosphate buffer (5 mL), TEMPO (240 mg, 1.54 mmol, 3 equiv), tetrabutylammonium chloride (0.36 g, 2.5 equiv), N-chlorosuccinimide (280 mg, 2.10 mmol, 4 equiv) in DCM (25 mL) were stirred overnight at 40° C. The reaction was extracted with DCM (200 mL). The extract was washed with water (1×200 mL) and brine (1×200 mL), dried (Na2SO4) and concentrated. This resulted in 0.4 g (134%, crude) of 122-1 as a yellow semi-solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((Benzyl oxy)carbonyl)-3-hydroxy-4,6a, 6b, 8a, 11,14b-hexamethyl -14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (122-2) 122-1 (400 mg, 0.70 mmol, 1 equiv), 2-methylbut-2-ene (2 mL), sodium phosphate monobasic (0.5 g, 6.00 equiv) and sodium chlorite (0.38 g, 6.00 equiv) in water (6 mL) and t-butanol (12 mL) were stirred for 30 min at −2° C. The reaction was partially concentrated under vacuum and the residue extracted with DCM (200 mL). The extract was washed with brine (1×200 mL), dried (Na2SO4) and concentrated under vacuum. This resulted in 0.758 g (184%, crud) of 122-2 as a yellow semi-solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S, 12aS, 12bR,14bR)-10-Hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic Acid (122-3) 122-2 (225 mg, 0.38 mmol) and Pd/C (45 mg) in EtOAc (16 mL) were placed under a hydrogen atmosphere (1 atm) and stirred overnight at rt. The reaction was filtered and concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and MeCN (38.0% MeCN up to 55.0% in 8 min); detector: UV 254 nm. This resulted in 18.5 mg (10%) of 122-3 as a white solid. MS (ES, m/z): [M+H]+=501.20;1H NMR (400 MHz, MeOH-d4, ppm) δ 0.85 (s, 3H), 1.01-1.31 (m, 16H), 1.44 (d, J=13.4 Hz, 7H), 1.54 (d, J=10.8 Hz, 1H), 1.59-1.81 (m, 5H), 1.81-2.01 (m, 3H), 2.10-2.28 (m, 2H), 2.54 (s, 1H), 2.79 (dt, J=13.7, 3.6 Hz, 1H), 3.99 (dd, J=11.8, 4.7 Hz, 1H), 5.62 (s, 1H). Example 5 (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-Heptamethyl-10-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (176-2) Synthesis of 2-Benzyl 10-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,10-dicarboxylate (176-1) 1-6 (400 mg, 0.68 mmol, 1 equiv), potassium iodide (56 mg, 0.5 equiv), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (182 mg, 1.23 mmol, 1.8 equiv), and potassium carbonate (282 mg, 2.04 mmol, 3 equiv) in DMF (2.5 mL) were stirred for 2 h at 60° C. The reaction was diluted with EtOAc, washed with water and brine, dried (Na2SO4) and concentrated under vacuum. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:3). This resulted in 460 mg (97%) of 176-1 as a light-yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-Heptamethyl-10-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (176-2) 176-1 (70 mg) and Pd(OH)2/C (5.6 mg) in THF (5 mL) were placed under a hydrogen atmosphere (1 atm) and stirred for 13h at rt. The reaction was filtered and concentrated. The residue was purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and MeCN (68% Phase B up to 77% in 10 min); detector: UV. This resulted in 13.2 mg (21.64%) of 176-2 as a white solid. MS (ES, m/z): [M+H]+=611.45;1H NMR (400 MHz, MeOH-d4) δ 0.88 (d, J=13.9 Hz, 6H), 0.90-0.93 (m, 1H), 0.95-1.09 (m, 5H), 1.10-1.24 (m, 9H), 1.25-1.30 (m, 1H), 1.31-1.55 (m, 9H), 1.63-1.80 (m, 3H), 1.81-2.06 (m, 4H), 2.09-2.22 (m, 5H), 2.29 (dd, J=13.2, 3.2 Hz, 1H), 2.50 (s, 1H), 2.76 (d, J=13.6 Hz, 1H), 4.89-4.99 (m, 2H), 5.57 (s, 1H). Example 6 (2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-Heptamethyl-10-(((5-methyl-2-oxo-1,3-dioxolan-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (178-1) 176-1 (110 mg) and Pd(OH)2/C (11 mg) in THF (10 mL) and EtOH (10 mL) were stirred for 1 h at rt. The reaction was filtered and concentrated. The residue was purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and MeCN (5% Phase B up to 84% in 1 min, up to 93% in 7 min); detector: UV. This resulted in 26.4 mg 178-1 as a white solid. MS (ES, m/z): [M+H]+=613.40; NMR (400 MHz, MeOH-d4) δ 0.83 (s, 3H), 0.85-0.91 (m, 4H), 0.97-1.09 (m, 5H), 1.13-1.16 (m, 3H), 1.18-1.19 (m, 5H), 1.21-1.31 (m, 2H), 1.38-1.58 (m, 12H), 1.63-1.78 (m, 3H), 1.79-2.08 (m, 4H), 2.16 (qd, J=12.8, 4.4 Hz, 2H), 2.28 (dt, J=13.2, 4.4 Hz, 1H), 2.51 (s, 1H), 2.77 (d, J=13.2 Hz, 1H), 4.19-4.32 (m, 1H), 4.36-4.50 (m, 1H), 4.91-4.99 (m, 1H), 5.00-5.09 (m, 1H), 5.57 (s, 1H). Example 7 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic Acid (190-3) Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzyloxy)carbonyl)-4-formyl-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicen-3-yl 2,5,8,11-tetraoxatetradecan-14-oate (190-1) EDCI (250 mg, 1.30 mmol, 2.5 equiv) was added to 122-1 (300 mg, 0.52 mmol, 1 equiv), 2,5,8,11-tetraoxatetradecan-14-oic acid (370 mg, 1.57 mmol, 3 equiv) and DMAP (130 mg, 1.06 mmol, 2 equiv) in DCM (20 mL). The reaction was stirred for 3 h at rt and then concentrated under vacuum. The residue was purified by silica gel column eluting with EtOAc/hexane (2:1). This resulted in 0.2 g (48%) of 190-1 as a yellow semi-solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-3-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-11-((benzyloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (190-2) 190-1 (200 mg, 0.25 mmol, 1 equiv), 2-methylprop-1-ene (2 mL), sodium dihydrogen phosphate (0.18 g, 6.00 equiv) and sodium chlorite (0.14 g, 6.00 equiv) in t-butanol (9 mL) and water (3 mL) were stirred for 2 h at rt. The reaction was concentrated under vacuum, diluted with DCM, washed with brine, dried over anhydrous sodium sulfate and concentrated under vacuum. This resulted in 0.2 g (98%) of 190-2 as a yellow solid. Example 8 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-Hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methylthio)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (194-10) Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-10,13-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (194-1) Into a 2-L round-bottom flask was placed 76-1 (prepared as described in Bioorg. Med Chem. 2010, 18, 433-454) (105 g, 165 mmol, 1 equiv), CH2Cl2(800 mL), and Dess-Martin periodinane (139.8 g, 330 mmol, 2 equiv). The resulting mixture was stirred overnight at room temperature. The reaction was quenched by the addition of 300 mL of sodium bicarbonate. The reaction mixture was washed with 3×1 L of H2O. The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The crude product was re-crystallized from petroleum ether and CH2Cl2to provide 194-1 (100 g, 96%) as a white solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,12aS,12bR,14bR,E)-10-(hydroxyimino)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (194-2) Into a 2-L round-bottom flask was placed 194-1 (59.5 g, 93.7 mmol, 1 equiv), pyridine (1 L), and NH2OH·HCl (23.2 g, 335 mmol, 3.6 equiv). The reaction slurry was stirred for 1 h at 70° C. The reaction mixture was concentrated under vacuum, diluted with 2 L of CH2Cl2, and washed with 4×1 L of 3 N HCl and 1 L of brine. The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The crude product was re-crystallized from CH2Cl2/petroleum ether to afford 194-2 (55.1 g, 90%) as a white solid. MS (ES, m/z): [M+1]+=650.15;1H NMR (300 MHz, Chloroform-d) δ 7.58-7.21 (m, 10H), 6.96 (s, 1H), 5.55 (s, 1H), 5.32 (s, 1H), 3.09 (ddd, J=15.5, 5.1, 3.6 Hz, 1H), 2.90 (ddd, J=13.3, 5.7, 3.6 Hz, 1H), 2.41-2.21 (m, 2H), 2.12-1.96 (m, 4H), 1.92-1.76 (m, 1H), 1.65 (q, J=18.6, 16.7 Hz, 3H), 1.56-1.25 (m, 11H), 1.24-0.92 (m, 16H), 0.70 (s, 3H). Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,12aS,12bR,14bR,E)-10-(acetoxyimino)-9-(acetoxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (194-3) Into a 2-L round-bottom flask was placed 194-2 (53.4 g, 82.2 mmol, 1 equiv), AcOH (400 mL), AC2O (400 mL), Pd(OAc)2(3.3 g, 14.8 mmol, 0.18 equiv), and PhI(OAc)2(31.8 g, 98.6 mmol, 1.2 equiv). The reaction slurry was stirred overnight at 60° C. The reaction mixture was concentrated, diluted in CH2Cl2, washed with saturated NaHCO3(aq)and brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-40% EtOAc in petroleum ether to afford 194-3 (31.3 g, 51%) as a light yellow solid (mixture of isomers, ˜6:1 C-23:C-24 acetates). Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9R,12aS,12bR,14bR)-9-(hydroxymethyl)-2,4a,6a, 6b,9,12a-hexamethyl-10,13-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (194-4) Into a 1000-mL round-bottom flask was placed diphenylmethyl 194-3 (15 g, 20 mmol), THF (90 mL), MeOH (90 mL), acetone (90 mL), and 2 N HCl (90 mL). The reaction slurry was stirred overnight at 50° C. The reaction mixture was concentrated, diluted with CH2Cl2, and washed with 2×300 mL of saturated NaHCO3and 2×300 mL of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The solid was dried in an oven under reduced pressure. The residue was applied onto a silica gel column with CH2Cl2/ethyl acetate (4:1) to provide 9 g (69%) of 194-4 as a yellow solid (single C-23 OH isomer). Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-hydroxy-9-(hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (194-5) To a stirred slurry of 194-4 (10 g, 15.4 mmol) in methanol (200 mL) was added NaBH4(0.6 g, 17 mmol, 1.1 equiv) in portions as −10° C. The reaction slurry was stirred for 1 h at room temperature. Upon completion the reaction was quenched with ice water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate (0% to 60% in 30 min) in petroleum ether to provide 8.4 g (84%) of 194-5 as a white foam (single C-3-beta isomer). Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-formyl-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (194-6) Into a 250-mL round-bottom flask was placed 194-5 (4 g, 6.13 mmol), CH2Cl2(40 mL), pH 8.6 buffer (20 mL), TEMPO (2.87 g, 18.4 mmol, 3 equiv), TBACl (4.26 g), and NCS (3.2 g, 24 mmol, 3.9 equiv). The resulting solution was stirred for 1.5 h at 40° C. The reaction mixture was cooled and extracted with 3×50 mL of CH2Cl2. The combined organic layers were washed with 2×100 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate (0-60%) in petroleum ether to provide 3.8 g (95%) of 194-6 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-hydroxy-4,6a, 6b, 8a, 11,14b-hexamethyl -14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (194-7) To a stirred solution of 194-6 (4 g, 6.2 mmol) and 2-methylbut-2-ene (6.6 mL) in water (8.9 mL) and t-BuOH (26.6 mL) was added NaH2PO4(4.4 g, 36.87 mmol, 6 equiv) at 0° C. To the above mixture was added NaClO2(3.3 g, 37 mmol, 6 equiv) in portions at 0° C. The reaction slurry was stirred for 2 h at room temperature. The reaction mixture was extracted with CH2Cl2and the combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 4 g of crude 194-7 as light yellow solid, which was used in the next step directly without further purification. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-hydroxy-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (194-8) A solution of 194-7 (6.0 g, 9 mmol), K2CO3(3.7 g, 27 mmol, 3 equiv) and KI (0.75 g, 4.5 mmol, 0.5 equiv) in DMF was stirred for 2 h at 60° C. The reaction mixture was extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, elutin with 2:1 petroleum ether/EtOAc to afford 194-8 (6.0 g, 86%) as a light yellow solid. MS (ES, m/z): [M+1]+=779.00;1H NMR (400 MHz, Chloroform-d) δ 7.50-7.29 (m, 10H), 6.95 (s, 1H), 5.54 (s, 1H), 5.04 (d, J=13.8 Hz, 1H), 4.80 (d, J=13.9 Hz, 1H), 4.44 (s, 2H), 4.03 (dd, J=9.9, 6.5 Hz, 1H), 2.98 (s, 1H), 2.88 (d, J=19.4 Hz, 2H), 2.79 (s, 4H), 2.40 (s, 1H), 2.22 (d, J=6.6 Hz, 6H), 2.14-1.93 (m, 4H), 1.70 (dddd, J=37.5, 31.0, 16.9, 7.4 Hz, 8H), 1.48 (d, J=10.1 Hz, 1H), 1.41-1.32 (m, 6H), 1.27 (dd,7=14.6, 3.4 Hz, 1H), 1.24-1.10 (m, 11H), 1.08 (s, 3H), 1.04-0.94 (m, 1H), 0.88 (d, J=8.8 Hz, 1H), 0.67 (s, 3H). Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(2-(methylthio)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (194-9) EDCI (250 mg, 1.30 mmol, 5 equiv) was added to 194-8 (200 mg, 0.26 mmol, 1 equiv), 2-(methylsulfanyl)acetic acid (270 mg, 2.54 mmol, 10 equiv) and DMAP (120 mg, 0.98 mmol, 4 equiv) in CH2Cl2(4 mL). The reaction was stirred overnight at rt and concentrated under vacuum. The residue was purified by silica gel column with 1:1 EtOAc:petroleum ether to provide 240 mg (quant) of 194-9 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-Hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methylthio)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (194-10) 194-9 (130 mg, 0.15 mmol, 1 equiv) and TFA (0.2 mL) in CH2Cl2(2 mL) were stirred for 1 h at rt. The reaction was concentrated under vacuum and the residue purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and MeCN (65% Phase B up to 75% in 8 min); detector: UV. This resulted in 24.8 mg (24%) of 194-10 as an off-white solid. MS (ES, m/z) [M+H]+=701.10;1H-NMR (300 MHz, MeOH-d4) δ 5.62 (s, 1H), 5.22 (dd, J=11.3, 5.3 Hz, 1H), 5.04 (d, J=13.9 Hz, 1H), 4.91 (s, 1H), 3.15 (s, 2H), 2.85 (d, J=13.7 Hz, 1H), 2.59 (s, 1H), 2.19 (d, J=8.2 Hz, 8H), 2.04-1.62 (m, 9H), 1.45 (d, J=13.6 Hz, 7H), 1.35-1.12 (m, 14H), 1.02 (dd,7=29.5, 10.7 Hz, 2H), 0.85 (s, 3H). Example 9 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-Hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methylsulfonyl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (195-2) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(2-(methylsulfonyl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (195-1) EDCI (175 mg, 0.91 mmol, 5 equiv) was added to 194-8 (140 mg, 0.18 mmol, 1 equiv), 2-methanesulfonylacetic acid (248 mg, 1.80 mmol, 10 equiv) and DMAP (84 mg, 0.69 mmol, 4 equiv) in CH2Cl2(2.5 mL) and the reaction stirred overnight at rt. The resulting mixture was concentrated under vacuum and the residue purified by silica gel column with EtOAc/Petroleum ether (1/1). This resulted in 160 mg (99%) of 195-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-Hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methyl sulfonyl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (195-2) A mixture of 195-1 (100 mg, 0.11 mmol, 1 equiv) and TFA (0.1 mL, 0.01 equiv) in DCM was stirred for 1 h at rt. The resulting mixture was concentrated under vacuum. The crude product was purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and MeCN (50% Phase B up to 63% in 13 min); detector: UV. This resulted in 195-2 (35.1 mg, 43.06%) as a light yellow solid. MS (ES, m/z): [M+H]+=732.95;1H NMR (300 MHz, MeOH-d4) δ 5.63 (s, 1H), 5.39-5.26 (m, 1H), 5.05 (d, J=13.9 Hz, 1H), 4.21 (s, 2H), 3.14 (d, J=1.0 Hz, 3H), 2.87 (d, J=14.0 Hz, 1H), 2.60 (s, 1H), 2.21 (s, 5H), 1.85 (s, 6H), 1.77 (d, J=13.2 Hz, 1H), 1.70 (d, J=16.6 Hz, 2H), 1.46 (d, J=13.0 Hz, 7H), 1.30 (s, 4H), 1.25-1.13 (m, 10H), 1.00 (s, 1H), 0.86 (s, 3H). Example 10 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((L-Valyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (196-2) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((tert-butoxycarbonyl)-L-valyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (196-1) EDCI (172.3 mg, 0.90 mmol, 5 equiv) was added to 194-8 (140 mg, 0.18 mmol, 1 equiv), (2S)-2-[[(tert-butoxy)carbonyl]amino]-3-methylbutanoic acid (195.2 mg, 0.90 mmol, 5 equiv) and DMAP (87.8 mg, 0.72 mmol, 4 equiv) in DCM. The reaction was stirred overnight at rt and concentrated under reduced pressure. The residue was purified by prep-TLC (Petroleum ether/EtOAc 1:1) to afford 196-1 (170 mg, 96.70%) as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((L-Valyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (196-2) 196-1 (170 mg, 0.17 mmol, 1 equiv) and TFA (0.15 mL, 2.02 mmol, 12 equiv) in DCM was stirred for 1 h at rt and then concentrated under vacuum. The crude product was purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase A: water (0.05% TFA), mobile phase B: MeCN; Flow rate: 60 mL/min; Gradient: 37% B to 65% B in 8 min; detector UV 254 nm to afford 196-2 (51.5 mg, 41.63%) as an off-white solid. MS (ES, m/z): [M+H]+=712.45;1H NMR (400 MHz, MeOH-d4) δ 5.63 (s, 1H), 5.43-5.34 (m, 1H), 5.11 (d, J=14.0 Hz, 1H), 4.84 (d, J=14.0 Hz, 1H), 3.92 (d, J=4.2 Hz, 1H), 2.87 (d, J=13.6 Hz, 1H), 2.59 (s, 1H), 2.27-2.12 (m, 2H), 2.21 (s, 4H), 1.98 (d, J=10.0 Hz, 1H), 1.87 (d, J=9.7 Hz, 4H), 1.80-1.60 (m, 4H), 1.44 (d, J=16.3 Hz, 6H), 1.25 (d, J=31.4 Hz, 7H), 1.20 (s, 3H), 1.16 (s, 3H), 1.09 (s, 1H), 1.03 (dd, J=7.0, 2.9 Hz, 5H), 0.99-0.92 (m, 1H), 0.85 (s, 3H). Example 12 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(Benzoyloxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (197-2) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(benzoyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (197-1) EDCI (175 mg, 0.91 mmol, 5 equiv) was added to 194-8 (140 mg, 0.18 mmol, 1 equiv), benzoic acid (110 mg, 0.90 mmol, 5 equiv) and DMAP (84 mg, 0.69 mmol, 4 equiv) in DCM (2.5 mL). The reaction was stirred overnight at rt and then concentrated under vacuum. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1/1). This resulted in 160 mg (101%) of 197-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(Benzoyloxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (197-2) 197-1 (160 mg, 0.18 mmol, 1 equiv) and TFA (0.2 mL, 2.69 mmol, 15 equiv) in DCM was stirred for 1 h at rt. The resulting mixture was concentrated under vacuum and the residue purified by prep-TLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase A: water (0.05% TFA), mobile phase B: MeCN; Flow rate: 60 mL/min; Gradient: 37% B to 65% B in 8 min; detector: UV 254 nm to afford 197-2 (27.8 mg, 21.40%) as an off-white solid. MS (ES, m/z): [M+H]+=717.00;1H NMR (300 MHz, MeOH-d4) δ 8.02-7.88 (m, 2H), 7.63 (t, J=7.4 Hz, 1H), 7.49 (t, J=7.6 Hz, 2H), 5.65 (s, 1H), 5.41 (dd, J=11.5, 5.1 Hz, 1H), 4.95 (s, 2H), 2.91 (d, J=13.9 Hz, 1H), 2.65 (s, 1H), 2.25 (d, J=14.8 Hz, 2H), 1.97 (s, 8H), 1.85-1.68 (m, 4H), 1.51 (s, 3H), 1.43 (d, J=7.6 Hz, 7H), 1.34 (d, J=12.3 Hz, 2H), 1.27 (s, 3H), 1.20 (d, J=6.8 Hz, 6H), 1.06 (t, J=13.9 Hz, 2H), 0.87 (s, 3H). Example 13 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((Cyclopropanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (198-2) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (198-1) EDCI (250 mg, 1.30 mmol, 5 equiv) was added to 194-8 (200 mg, 0.26 mmol, 1 equiv), cyclopropanecarboxylic acid (220 mg, 2.56 mmol, 10 equiv), and DMAP (120 mg, 0.98 mmol, 4 equiv) in DCM (4 mL) and the reaction stirred overnight at rt. The mixture was concentrated under vacuum and the residue purified by silica gel column eluting with EtOAc/petroleum ether (1/1). This resulted in 230 mg (106%) of 198-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((Cyclopropanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (198-2) 198-1 (115 mg, 0.14 mmol, 1 equiv) and TFA (0.15 mL) in DCM (1.5 mL) were stirred for 1 h at rt. The reaction was concentrated under vacuum and the residue (115 mg) purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase A: water (0.05% TFA), mobile phase B: MeCN; Flow rate: 60 mL/min; Gradient: 30% B to 60% B in 8 min; detector: 254 nm. This resulted in 26.9 mg (29%) of 198-2 as an off-white solid. MS (ES, m/z): [M+H]+=681.20;1H NMR (300 MHz, MeOH-d4) δ 5.62 (s, 1H), 5.14 (dd, J=11.5, 5.1 Hz, 1H), 5.02 (d, J=13.9 Hz, 1H), 4.93 (s, 1H), 2.83 (d, J=13.6 Hz, 1H), 2.58 (s, 1H), 2.20 (s, 5H), 2.01-1.58 (m, 8H), 1.57-1.33 (m, 10H), 1.33-1.13 (m, 10H), 1.07 (d, J=12.8 Hz, 3H), 0.95 (s, 2H), 0.87 (d, J=13.2 Hz, 6H). Example 14 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((R)-2-Methoxypropanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (203-2) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((R)-2-methoxypropanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (203-1) EDCI (240 mg, 1 mmol, 5 equiv) was added to 194-8 (200 mg, 0.2 mmol, 1 equiv), (2R)-2-methoxypropanoic acid (132 mg, 1 mmol, 5 equiv) and DMAP (132 mg, 0.8 mmol, 4 equiv) in DCM (2 mL) and the reaction stirred for 2 hr at rt. The mixture was concentrated under vacuum and the residue purified by silica gel column with EtOAc/petroleum ether (1/1). This resulted in 150 mg of 203-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((R)-2-Methoxypropanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (203-2) 203-1 (150 mg, 0.17 mmol, 1 equiv) and TFA (0.2 mL) in DCM (2 mL) were stirred for 2 hr at rt. The resulting mixture was concentrated and the residue purified by Flash-prep-HPLC resulting in 47.5 mg (39.20%) of 203-1 as a white solid. MS (ES, m/z): [M+H]+=699;1H NMR (300 MHz, Chloroform-7) δ 5.72 (s, 1H), 5.25 (dd, J=11.6, 5.0 Hz, 1H), 4.99 (d, J=13.7 Hz, 1H), 4.69 (d, J=13.8 Hz, 1H), 3.79 (q, J=6.8 Hz, 1H), 3.34 (s, 3H), 2.89 (d, J=13.7 Hz, 1H), 2.44 (s, 1H), 2.19 (s, 4H), 1.99 (s, 7H), 1.79 (s, 10H), 1.42-1.30 (m, 14H), 1.27-1.15 (m, 1H), 0.82 (s, 4H). Example 15 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((S)-2-Methoxypropanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (204-2) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((S)-2-methoxypropanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (204-1) EDCI (240 mg, 1.2 mmol, 5 equiv) was added to 194-8 (200 mg, 0.2 mmol, 1 equiv), (2S)-2-methoxypropanoic acid (132 mg, 1.2 mmol, 5 equiv) and DMAP (120 mg, 0.8 mmol, 4 equiv) in DCM (2 mL) and the reaction stirred for 3 hr at 25° C. The mixture was concentrated under vacuum and the residue purified by silica gel column with EtOAc/petroleum ether (1/1). This resulted in 150 mg of 204-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((S)-2-Methoxypropanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (204-2) 204-1 (150 mg, 0.17 mmol, 1 equiv) and TFA (0.2 mL, 0.01 equiv) in DCM (2 mL) were stirred for 2 hr at 25° C. The reaction was concentrated and the crude product purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and MeCN (57% Phase B up to 85% in 8 min); detector: UV. This resulted in 204-2 as a white solid. MS (ES, m/z): [M+H]+=699;1H NMR (300 MHz, Chloroform-7) δ 5.72 (s, 1H), 5.26 (dd, J=11.6, 4.9 Hz, 1H), 4.98 (d, J=13.7 Hz, 1H), 4.68 (d, J=13.8 Hz, 1H), 3.79 (t, 7=6.9 Hz, 1H), 3.36 (s, 3H), 2.89 (d, J=13.8 Hz, 1H), 2.45 (s, 1H), 2.19 (s, 4H), 2.00 (s, 3H), 1.87-1.71 (m, 7H), 1.39 (s, 6H), 1.33 (d, J=6.9 Hz, 4H), 1.27-1.15 (m, 12H), 1.11 (s, 3H), 1.04 (d, J=12.6 Hz, 1H), 0.82 (s, 4H). Example 16 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(Methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (205-2) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (205-1) Bromo(methoxy)methane (0.063 mL, 4.0 equiv) was a added dropwise at 0° C. to 194-8 (150 mg, 1 equiv) and DIEA (0.318 mL, 10.0 equiv) in DCM (10 mL) and then heated at 60° C. for 1 hr. The reaction was concentrated and the residue purified by silica gel column with EtOAc/petroleum ether (1:1). This resulted in 178.1 mg (112.38%) of 205-1 as a white crude solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(Methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (205-2) 205-1 (178.1 mg) and 10% TFA/DCM (10 mL) were stirred for 5 hr at rt. The reaction mixture was washed with brine (3×50 mL), dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (58% Phase B up to 76% in 8 min); detector: UV. This resulted in 40.0 mg (28%) of 205-2 as a white solid. MS (ES, m/z): [M+H]+=657.25;1H NMR (400 MHz, Chloroform-7) δ 0.85 (s, 4H), 1.01-1.15 (m, 5H), 1.20 (d, J=12.0 Hz, 6H), 1.26 (s, 4H), 1.33-1.58 (m, 8H), 1.59-1.61 (m, 1H), 1.65-1.70 (m, 2H), 1.72-1.91 (m, 2H), 1.92-2.17 (m, 4H), 2.18-2.20 (m, 1H), 2.22 (s, 3H), 2.43 (s, 1H), 2.87 (d, J=14.0 Hz, 1H), 3.29 (s, 3H), 3.97 (dd, J=11.6, 4.4 Hz, 1H), 4.52 (d, 7=6.8 Hz, 1H), 4.66 (d, J=7.2 Hz, 1H), 4.75 (d, J=13.6 Hz, 1H), 5.06 (d, J=13.6 Hz, 1H), 5.74 (s, 1H). Example 17 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((Ethylcarbamoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (206-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((ethylcarbamoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (206-1) Isocyanatoethane (0.061 mL, 3 equiv), chlorotrimethylsilane (0.111 mL, 5 equiv), and 194-8 (200 mg, 0.26 mmol) in CH2Cl2(10 mL) were stirred overnight at rt. The reaction was concentrated under vacuum and the residue purified by silica gel column with EtOAc/petroleum ether (1:1). This resulted in 239.9 mg (quant) of 206-1 as a white crude solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((Ethylcarbamoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (206-2) 206-1 (239.9 mg) and 10% TFA/CH2Cl2(10 mL) were stirred for 1 hr at rt. The reaction was concentrated and the residue purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and MeCN (57% Phase B up to 77% in 8 min); detector: UV. This resulted in 65.7 mg (34%) of 206-2 as a white solid. MS (ES, m/z): [M+H]+=684.05;1H NMR (400 MHz, MeOH-d4) δ 0.82 (s, 3H), 0.89-0.99 (m, 1H), 1.01-1.04 (m, 1H), 1.09 (t, J=7.2 Hz, 3H), 1.13 (s, 3H), 1.17-1.28 (m, 11H), 1.33-1.48 (m, 7H), 1.61-1.79 (m, 6H), 1.81-1.90 (m, 2H), 1.95 (d, J=10.0 Hz, 1H), 2.11-2.28 (m, 5H), 2.55 (s, 1H), 2.79 (d, J=14.0 Hz, 1H), 2.98-3.17 (m, 2H), 4.89 (d, J=14.0 Hz, 1H), 4.95-5.05 (m, 2H), 5.59 (s, 1H). Example 18 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((butylcarbamoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (207-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((butylcarbamoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (207-1) 1-Isocyanatobutane (0.0868 mL, 3 equiv), TMSCl (0.111 mL, 5 equiv), and 194-8 (200 mg, 0.26 mmol, 1 equiv) in CH2Cl2(10 mL) were stirred overnight at rt. The reaction was concentrated under vacuum and the residue purified by silica gel column with EtOAc/petroleum ether (1:1). This resulted in 256.9 mg (114%) of 207-1 as a white crude solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((butylcarbamoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (207-2) 207-1 (256.9 mg) and 10% TFA/CH2Cl2(10 mL) were stirred for 1 hr at rt. The reaction was concentrated under vacuum and the crude product purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and MeCN (71% Phase B up to 72% in 8 min); detector: UV. This resulted in 28.8 mg (14%) of 207-2 as an off-white solid. MS (ES, m/z): [M+H]+=712.05;1H NMR (400 MHz, MeOH-d4) δ 0.85 (s, 3H), 0.94 (t, J=7.4 Hz, 4H), 1.07 (d, J=13.6 Hz, 1H), 1.12-1.30 (m, 14H), 1.31-1.59 (m, 11H), 1.63-1.82 (m, 6H), 1.83-1.93 (m, 2H), 1.96-2.08 (m, 1H), 2.09-2.32 (m, 5H), 2.57 (s, 1H), 2.81 (d, J=14.0 Hz, 1H), 2.97-3.14 (m, 2H), 4.90 (d, J=13.6 Hz, 1H), 4.97-5.11 (m, 2H), 5.62 (s, 1H). Example 19 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-((pyrrolidine-1-carbonyl)oxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (208-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-((pyrrolidine-1-carbonyl)oxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (208-1) 194-8 (400 mg), pyrrolidine-1-carbonyl chloride (0.17 mL, 3 equiv), and DMAP (62.7 mg, 1 equiv) in pyridine (10 mL) were stirred for 9 days at 90° C. The reaction mixture was concentrated and the residue dissolved in EtOAc. The solution was washed with 1 M HCl (3 x 50 mL) and brine (1×50 mL), dried over anhydrous Na2SO4, and concentrated to provide 170 mg (38%) of 208-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-((pyrrolidine-1-carbonyl)oxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (208-2) 208-1 (170 mg, 1 equiv) and 10% TFA/CH2Cl2(10 mL) were stirred for 1 hr at rt. The reaction mixture was concentrated under vacuum and the residue purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (68% Phase B up to 80% in 8 min); detector: UV. This resulted in 9.5 mg (6.5%) of 208-2 as a white solid. MS (ES, m/z): [M+H]+=710.20;1H NMR (400 MHz, MeOH-d4) δ 0.83 (s, 3H), 0.94-0.99 (m, 1H), 1.04 (d, J=13.6 Hz, 1H), 1.15 (s, 3H), 1.18 (s, 6H), 1.21-1.27 (m, 6H), 1.29-1.36 (m, 2H), 1.38-1.41 (m, 3H), 1.44 (s, 3H), 1.57-1.61 (m, 1H), 1.63-1.79 (m, 6H), 1.87 (s, 6H), 1.95 (d, J=10.0 Hz, 1H), 2.10-2.15 (m, 5H), 2.56 (s, 1H), 2.80 (d, J=14.0 Hz, 1H), 3.13-3.24 (m, 1H), 3.29 (s, 3H), 4.90-5.04 (m, 3H), 5.60 (s, 1H). Example 20 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (209-3) Synthesis of 2-benzhydryl 9-((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (209-1) 4-(Bromomethyl)-5-isopropyl-1,3-dioxol-2-one was prepared according to literature procedures (Sun et al,Tetrahedron Letters,2002, 43, 1161-1164). A mixture of 4-(bromomethyl)-5-isopropyl-1,3-dioxol-2-one (2.7 g, 1.5 equiv), K2CO3(3.4 g, 3 equiv), KI (0.68 g, 0.5 equiv), and 194-7 (5.5 g, 1 equiv) in DMF (80 mL) was stirred for 1 h at 60° C. The reaction mixture was cooled to room temperature and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column chromatography, eluting with 3:1 petroleum ether:EtOAc to afford 209-1 (5.5 g, 83%) as a light yellow solid. MS (ES, m/z) [M+1]+=806.95;1H NMR (400 MHz, Chloroform-d) δ 7.48-7.28 (m, 10H), 6.95 (s, 1H), 5.54 (s, 1H), 5.07 (d, J=13.8 Hz, 1H), 4.83 (d, J=13.8 Hz, 1H), 4.10-3.93 (m, 1H), 3.11-2.94 (m, 2H), 2.94-2.82 (m, 2H), 2.40 (s, 1H), 2.17-1.93 (m, 5H), 1.91-1.53 (m, 7H), 1.48 (d, J=10.6 Hz, 2H), 1.43-1.31 (m, 6H), 1.31-1.22 (m, 8H), 1.22-1.11 (m, 12H), 1.08 (s, 3H), 1.00 (d, J=13.7 Hz, 1H), 0.90 (t, J=10.1 Hz, 1H), 0.67 (s, 3H). Synthesis of 2-benzhydryl 9-((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (209-2) Into an 8-mL round-bottom flask was placed 209-1 (100 mg, 0.12 mmol), CH2Cl2(2 mL), DMAP (29 mg, 0.24 mmol, 1.9 equiv), AcOH (21.2 mg, 0.35 mmol, 2.85 equiv), and then EDCI (57 mg, 0.3 mmol, 2.4 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was concentrated and the residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 60 mg (57%) of 209-2 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (209-3) Into a 25-mL round-bottom flask was placed 209-2 (60 mg, 0.07 mmol), CH2Cl2(5 mL), and TFA (0.5 mL, 6.7 mmol, 95 equiv). The reaction slurry was stirred for 1 hr at room temperature. The resulting mixture was concentrated and the crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH Prep C18 OBD, 5 μm, 19*150 mm; mobile phase, water (0.05% TFA) and CH3CN (70% Phase B up to 90% in 8 min); detector, uv. This resulted in 29.6 mg (61%) of 209-3 as a white solid. The product was tested in the assay in described in example 112 demonstrating a pIC50 of 7.2 compared to the corresponding acid metabolite (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(acetyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid having a pIC50of 5.9. MS (ES, m/z): [M+1]+=683;1H NMR (300 MHz, methanol-d4) δ 0.85 (s, 3H), 0.95 (s, 1H), 1.07 (d, J=13.8 Hz, 1H), 1.18 (d, J=13.4 Hz, 8H), 1.22-1.34 (m, 9H), 1.45 (d, J=12.8 Hz, 6H), 1.65-1.83 (m, 5H), 1.88 (dd, J=12.1, 4.9 Hz, 1H), 1.96 (s, 3H), 2.09-2.29 (m, 2H), 2.58 (s, 1H), 2.83 (d, J=13.7 Hz, 1H), 3.07 (h, J=6.9 Hz, 1H), 4.89 (d, J=13.9 Hz, 1H), 5.07 (d, J=13.9 Hz, 1H), 5.16 (dd, J=11.4, 5.2 Hz, 1H), 5.62 (s, 1H). Example 21 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (211-1) The title compound was prepared according to the methods for compound 194-10, beginning with 194-8 and propanoic acid. The crude product was purified by prep-HPLC with the following conditions: Column, X Select CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (60% Phase B up to 85% in 8 min); detector, UV. 69.7 mg of 211-1 was obtained as white solid. MS (ES, m/z): [M+1]+=669;1H NMR (300 MHz, Chloroform-d) δ 5.72 (s, 1H), 5.22-5.11 (m, 1H), 4.96 (d, J=13.8 Hz, 1H), 4.72 (d, J=13.8 Hz, 1H), 2.86 (d, J=13.9 Hz, 1H), 2.45 (s, 1H), 2.32-2.16 (m, 6H), 1.99 (s, 3H), 1.79 (s, 5H), 1.70 (d, J=9.9 Hz, 2H), 1.39 (s, 4H), 1.24 (s, 3H), 1.19 (d, J=9.0 Hz, 12H), 1.14-1.03 (m, 6H), 0.83 (s, 4H). Example 22 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopentanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (212-1) The title compound was prepared according to the methods for compound 194-10, beginning with 194-8 and cyclopentanoic acid. The crude product was purified by prep-HPLC with the following conditions: Column, Kinetex EVO C18, 21.2*150; 5 μm; mobile phase, water (0.05% TFA) and CH3CN (70% PhaseB up to 88% in 8 min); detector, UV. This resulted in 66.3 mg of 212-1 as a white solid. MS (ES, m/z): [M+1]+=709;1H NMR (300 MHz, Chloroform-d) δ 5.72 (s, 1H), 5.14 (dd, J=10.5, 6.3 Hz, 1H), 4.94 (d, J=13.8 Hz, 1H), 4.71 (d, J=13.8 Hz, 1H), 2.86 (d, J=14.0 Hz, 1H), 2.64 (q, J=8.0, 7.5 Hz, 1H), 2.44 (s, 1H), 2.18 (s, 4H), 1.99 (s, 3H), 1.82 (d, J=8.5 Hz, 14H), 1.39 (s, 7H), 1.27-1.14 (m, 13H), 1.11 (s, 1H), 1.03 (d, J=12.7 Hz, 1H), 0.83 (s, 4H). Example 23 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-((3-(piperidin-1-yl)propanoyl)oxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (215-1) The title compound was prepared according to the methods for compound 194-10, beginning with 194-8 and 3-(piperidin-1-yl)propanoic acid. The crude product was purified by prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30*150 mm 5 um, n; mobile phase, water (0.05% TFA) and ACN (40% Phase B up to 53% in 8 min); detector, UV 254 nm. This resulted in 35.2 mg (33%) of 215-1 as a white solid. MS (ES, m/z): [M+1]+=752.43;1H NMR (300 MHz, Chloroform-d) δ 11.9 (s, 1H), 5.71 (s, 1H), 5.16 (s, 1H), 4.86 (d, J=4.5 Hz, 2H), 3.56 (s, 3H), 3.27 (s, 6H), 2.88 (d, J=17.7 Hz, 3H), 2.66 (s, 2H), 2.20 (s, 5H), 1.98 (s, 4H), 1.88 (d, J=14.0 Hz, 5H), 1.61 (d, J=12.0 Hz, 7H), 1.39 (d, J=6.4 Hz, 9H), 1.26-1.14 (m, 16H), 1.12 (s, 2H), 0.82 (s, 3H). Example 24 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((isopropoxycarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (216-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((isopropoxycarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (216-1) Into a 8 mL sealed tube were added 194-8 (200 mg, 0.26 mmol), pyridine (3 mL), DMAP (94.1 mg, 0.77 mmol, 3 equiv), and isopropyl chloroformate (315 mg, 2.6 mmol, 10 equiv) at room temperature. The reaction slurry was stirred for 6 h at 60° C. under nitrogen atmosphere. To the above mixture was added an additional portion of isopropyl chloroformate (315 mg, 2.6 mmol, 10 equiv) at 60° C. The reaction slurry was stirred for additional 6 h at 60° C. To the reaction mixture was added a third portion of isopropyl chloroformate (315 mg, 2.6 mmol, 10 equiv) at 60° C. The reaction slurry was stirred for an additional 6 h at 60° C. The reaction mixture was concentrated under vacuum. The residue was purified by prep-TLC (PE/EtOAc 1:1) to afford 216-1 (130 mg, 59%) as a pale yellow foam. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((isopropoxycarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (216-2) Into a 25-mL round-bottom flask was 216-1 (130 mg, 0.15 mmol, 1 equiv), CH2Cl2(2 mL), and TFA (0.2 mL). The resulting solution was stirred for 30 min at room temperature. The resulting mixture was concentrated. The crude product was purified by prep-HPLC with the following condition: Column, Xselect CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (64% Phase B up to 87% in 8 min); detector, UV. This resulted in 40.9 mg (39%) of 216-2 as a white solid. MS (ES, m/z): [M+1]+=698.37;1H NMR (400 MHz, Chloroform-d) δ 7.38 (dd, J=17.4, 5.5 Hz, 1H), 5.75 (s, 1H), 5.14-4.93 (m, 2H), 4.87-4.75 (m, 2H), 2.90 (d, J=13.8 Hz, 1H), 2.45 (s, 1H), 2.21 (s, 4H), 2.11-1.90 (m, 4H), 1.87-1.76 (m, 3H), 1.65 (dd, J=12.4, 6.5 Hz, 5H), 1.57-1.32 (m, 9H), 1.28 (dd, J=6.3, 3.4 Hz, 7H), 1.25 (d, J=6.9 Hz, 7H), 1.19 (d, J=5.5 Hz, 5H), 1.13 (s, 4H), 1.10-1.00 (m, 2H), 0.85 (s, 5H), 0.09 (s, 3H). Example 25 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-Difluoroacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (223-1) The title compound was prepared according to the methods for compound 194-10, beginning with 194-8 and 2,2-difluoroacetic acid. The crude product was purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (68% Phase B up to 80% in 8 min); detector: UV. This resulted in 82.1 mg (46%) of 223-1 as a white solid. MS (ES, m/z): [M+H]+=691.15;1H NMR (400 MHz, MeOH-d4) δ 0.83 (s, 3H), 0.97 (d, J=7.6 Hz, 1H), 1.05 (d, J=13.6 Hz, 1H), 1.14 (s, 3H), 1.18 (d, J=8.4 Hz, 6H), 1.22-1.31 (m, 5H), 1.40 (s, 4H), 1.45 (s, 3H), 1.64-1.80 (m, 5H), 1.82-1.90 (m, 3H), 1.92-2.00 (m, 1H), 2.09-2.29 (m, 5H), 2.57 (s, 1H), 2.88 (d, J=10.4 Hz, 1H), 4.88 (d, J=14.0 Hz, 1H), 5.02 (d, J=14.0 Hz, 1H), 5.33 (dd, J=14.0, 5.2 Hz, 1H), 5.61 (s, 1H), 6.05 (t, J=53.2 Hz, 1H). Example 26 (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-Hydroxy-2,4a,6a,6b,9,12a-hexamethyl-9-((2-((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-oxoethoxy)methyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (240-8) Synthesis of Benzyl (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-9-((2-(tert-butoxy)-2-oxoethoxy)methyl)-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (240-1) Sodium bis(trimethylsilyl)amide (0.5 mL, 2 equiv) was added to 121-1 (355 mg, 0.62 mmol, 1 equiv) and tert-butyl 2-bromoacetate (240.6 mg, 1.24 mmol, 2 equiv) in DMF (5 mL). The reaction slurry was stirred overnight at rt, diluted with CH2Cl2(100 mL), washed with brine (2×100 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:1) to provide 300 mg (71%) of 240-1 as a white solid. Synthesis of 2-(((3S,4R,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((Benzyloxy)carbonyl)-3-hydroxy-4,6a, 6b, 8a, 11,14b-hexamethyl -14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicen-4-yl)methoxy)acetic Acid (240-2) 240-1 (300 mg, 0.43 mmol, 1 equiv) and TFA (1 mL) were combined in CH2Cl2(10 mL) for 1 h at rt. The reaction mixture was diluted with CH2Cl2(100 mL), washed with brine (3×50 mL), dried over anhydrous Na2SO4, filtered, and concentrated. This resulted in 270 mg (98%) of 240-2 as a yellow solid. Synthesis of Benzyl (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-hydroxy-9-((2-methoxy-2-oxoethoxy)methyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (240-3) (Trimethylsilyl)diazomethane (1 mL) was added dropwise to 240-2 (270 mg, 0.43 mmol, 1 equiv) in MeOH (5 mL) and CH2Cl2(10 mL). The reaction slurry was stirred for 2 h at rt. The reaction mixture was concentrated to provide 260 mg (94%) of 240-3 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-Hydroxy-9-((2-methoxy-2-oxoethoxy)methyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (240-4) 240-3 (270 mg, 0.42 mmol) and Pd/C (20 mg, 10% wt) in EtOAc (30 mL) were stirred for 2 h under H2(1 atm). The reaction mixture was filtered and concentrated to provide 216 mg (93%) of 240-4 as a white solid. Synthesis of Benzhydryl (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-hydroxy-9-((2-methoxy-2-oxoethoxy)methyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (240-5) (Diazomethylene)dibenzene (180 mg) in MeOH (20 mL) was added to 240-4 (216 mg, 1 equiv) in ether (10 mL). The reaction slurry was stirred for 3 h at rt. The reaction mixture was concentrated and the residue purified by silica gel column eluting with EtOAc/petroleum ether (1:10) to provide 220 mg (79%) of 240-5 as a white solid. Synthesis of 2-(((3S,4R,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((Benzhydryloxy)carbonyl)-3-hydroxy-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicen-4-yl)methoxy)acetic Acid (240-6) 240-5 (220 mg, 0.30 mmol, 1 equiv) and lithium hydroxide (73 mg, 3 mmol, 10 equiv) were combined in THF (10 mL), H2O (1 mL), and MeOH (1 mL), stirring for 2 h at rt. The reaction mixture was adjusted to pH=4 with 1 M HCl and extracted with CH2Cl2(2×100 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated to provide 200 mg (93%) of 240-6 as a white solid. Synthesis of Benzhydryl (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-9-((2-((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-oxoethoxy)methyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (240-7) 240-6 (200 mg, 0.28 mmol, 1 equiv), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (84 mg, 0.57 mmol, 2 equiv), K2CO3(194 mg, 1.40 mmol, 5 equiv), and potassium iodide (70 mg, 0.42 mmol, 1.5 equiv) were combined in DMF (10 mL) and stirred for 1 h at 50° C. The reaction mixture was diluted with CH2Cl2(100 mL), washed with brine (3×100 mL), dried over anhydrous Na2SO4, filtered, and concentrated to provide 200 mg (86%) of 240-7 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9R,10S,12aS,12bR,14bR)-10-Hydroxy-2,4a,6a,6b,9,12a-hexamethyl-9-((2-((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-oxoethoxy)methyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (240-8) 240-7 (200 mg, 0.24 mmol, 1 equiv) and TFA (1 mL) were combined in CH2Cl2(10 mL) and stirred for 1 h. The solids were collected by filtration. The resulting mixture was concentrated and the residue purified by prep-HPLC with the following conditions—mobile phase: water (0.05% TFA) and CH3CN (hold 5% Phase B in 0 min, up to 63% in 1 min, up to 68% in 8 min); detector: UV. This resulted in 14.5 mg (9%) of 240-8 as a white solid. MS (ES, m/z): [M+H]+=657.25;1H NMR (400 MHz, chloroform-rf) δ 0.72 (s, 3H), 0.86 (s, 3H), 1.03 (t, J=15.2 Hz, 2H), 1.13-1.22 (m, 9H), 1.22-1.37 (m, 2H), 1.43 (d, J=5.6 Hz, 8H), 1.66-1.97 (m, 8H), 2.21 (s, 5H), 2.51 (s, 1H), 2.73 (d, J=13.6 Hz, 1H), 3.39 (s, 2H), 3.72 (dd, J=11.8, 4.8 Hz, 1H), 4.09 (d, J=16.8 Hz, 1H), 4.26 (d, J=16.8 Hz, 1H), 4.92 (s, 1H), 5.03 (s, 2H), 5.60 (s, 1H). Example 27 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Acetoxy-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (243-1) The title compound was prepared according to the methods for compound 194-10, beginning with 194-8 and acetic acid. The crude product was purified by prep-HPLC with the following conditions—Column: XSelect CSHPrep C18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and CH3CN (hold 5% Phase B in 0 min, up to 65% in 1 min, up to 85% in 8 min); detector: UV. This resulted in 24.7 mg (20%) of 243-1 as a white solid. MS (ES, m/z): [M+H]+=655.15; 1HNMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.96 (d, J=5.6 Hz, 1H), 1.06 (d, J=14.1 Hz, 1H), 1.16-1.29 (m, 14H), 1.36-1.49 (m, 7H), 1.63-1.80 (m, 6H), 1.80-1.92 (m, 2H), 1.92-2.01 (m, 4H), 2.08-2.08 (m, 5H), 2.55 (s, 1H), 2.78-2.86 (m, 1H), 4.89 (d, J=13.9 Hz, 1H), 5.03 (d, J=14.0 Hz, 1H), 5.15 (dd, J=11.7, 4.9 Hz, 1H), 5.62 (s, 1H). Example 28 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-Hydroxyacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (244-1) The title compound was prepared according to the methods for compound 194-10, beginning with 194-8 and 2-[(4-methoxyphenyl)methoxy]acetic acid. The crude product was purified by prep-HPLC with the following conditions—Column: XBridge Prep Cl8 OBD, 19*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (35% Phase B up to 90% in 7 min); detector: UV. This resulted in 25.8 mg (28%) of 244-1 as a white solid. MS (ES, m/z): [M+H]+=671.35;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.95 (s, 1H), 1.06 (d, J=14.0 Hz, 1H), 1.15 (s, 3H), 1.17-1.27 (m, 10H), 1.27 (d, J=8.0 Hz, 1H), 1.33-1.524 (m, 7H), 1.61-2.00 (m, 9H), 2.20 (s, 5H), 2.58 (s, 1H), 2.80-2.88 (m, 1H), 3.96-4.11 (m, 2H), 4.87 (d, J=13.9 Hz, 1H), 5.06 (d, J=14.0 Hz, 1H), 5.26 (dd, J=11.7, 5.0 Hz, 1H), 5.62 (s, 1H). Example 29 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-Methoxyacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (245-1) The title compound was prepared according to the methods for compound 194-10, beginning with 194-8 and 2-methoxyacetic acid. The crude product was purified by prep-HPLC with the following conditions—Column: XSelect CSHPrep C18 OBD, 5 μm, 19*150 mm: mobile phase: water (0.05% TFA) and CH3CN (hold 5% Phase B in 0 min, up to 62% in 1 min, up to 82% in 8 min); detector: UV. This resulted in 25.9 mg (19%) of 245-1 as a white solid. MS (ES, m/z): [M+H]+=685.15;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 1.06 (d, J=14.1 Hz, 1H), 1.00-1.10 (m, 1H), 1.10-1.30 (m, 14H), 1.35-1.51 (m, 7H), 1.61-2.08 (m, 9H), 2.09-2.28 (m, 5H), 2.59 (s, 1H), 2.80-2.88 (m, 1H), 3.39 (s, 3H), 3.97 (d, J=1.3 Hz, 2H), 4.88 (d, J=14.0 Hz, 1H), 5.05 (d, J=13.9 Hz, 1H), 5.27 (dd, J=11.8, 5.0 Hz, 1H), 5.62 (s, 1H). Example 30 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-Tetraoxadodecanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (246-3) Synthesis of 2-(2-(2-Methoxyethoxy)ethoxy)ethyl (4-nitrophenyl) carbonate (246-1) 2-[2-(2-Methoxyethoxy)ethoxy]ethan-1-ol (3.0 g, 18.3 mmol, 1 equiv), p-nitrophenyl chloroformate (4.4 g, 20.4 mmol, 1.1 equiv) and Et3N (5 mL, 36 mmol, 2 equiv) in CH2Cl2(20 mL) were stirred for 1 h at rt. The reaction was concentrated and the residue purified by silica gel column eluting with EtOAc/petroleum ether (1:1) to provide 4.2 g (67%) of 246-1 as a clear liquid. Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxadodecanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (246-2) 194-8 (150 mg, 0.19 mmol, 1 equiv), 246-1 (640 mg, 1.9 mmol, 10 equiv), and DMAP (94 mg, 0.77 mmol, 4 equiv) in THF (8 mL) were stirred for 24 h at 50° C. The reaction mixture was diluted with CH2Cl2(300 mL), washed with brine (3×100 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:1) to provide 156 mg (84%) of 246-2 as a yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-Tetraoxadodecanoyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (246-3) 246-2 (156 mg, 0.16 mmol, 1 equiv), and TFA (1 mL) in CH2Cl2(10 mL) were stirred for 1 h at rt. The reaction mixture was concentrated and the residue purified by prep-HPLC with the following conditions—Column, XBridge Prep C18 OBD, 19*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (35% Phase B up to 90% in 7 min); detector: UV. This resulted in 21.5 mg (16%) of 246-3 as a white solid. MS (ES, m/z): [M+H]+=803.20;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.98 (d, J=9.2 Hz, 1H), 1.06 (d, J=14.1 Hz, 1H), 1.13-1.25 (m, 14H), 1.44 (d, J=18.2 Hz, 7H), 1.61-1.93 (m, 8H), 1.97 (d, J=9.6 Hz, 1H), 2.11-2.37 (m, 5H), 2.58 (s, 1H), 2.81-2.89 (m, 1H), 3.38 (s, 3H), 3.52-3.61 (m, 2H), 3.61-3.73 (m, 8H), 4.12-4.30 (m, 2H), 4.91-5.07 (m, 3H), 5.62 (s, 1H). Example 31 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Methoxy-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (249-5) Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (249-1) (Trimethylsilyl)diazomethane (1 mL) was added to 194-7 (200 mg, 0.30 mmol, 1 equiv) in CH2Cl2(5 mL) and MeOH (2.5 mL), The resulting solution was stirred for 1 h at rt. The reaction mixture was concentrated to provide 200 mg (98%) of 249-1 as a white solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (249-2) Sodium hydride (100 mg, 4.17 mmol, 14 equiv) was added portionwise at rt to 249-1 (200 mg, 0.29 mmol, 1 equiv) and iodomethane (0.5 mL, 8.03 mmol) in THF (10 mL). The reaction slurry was stirred overnight at 50° C. and then quenched by the addition of water. The reaction mixture was extracted with EtOAc (3×50 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:1) to provide 180 mg (88%) of 249-2 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-methoxy-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (249-3) A slurry of 249-2 (180 mg, 0.26 mmol, 1 equiv) and lithium iodide (100 mg, 0.75 mmol, 2.9 equiv) in pyridine (5 mL) was stirred for 2 days at 125° C. The reaction mixture was diluted with CH2Cl2, washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:1) to provide 110 mg (62%) of 249-3 as a yellow solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (249-4) 249-3 (110 mg, 0.16 mmol, 1 equiv), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (150 mg, 1.0 mmol, 6.3 equiv), K2CO3(150 mg, 1.1 mmol, 6.7 equiv), and potassium iodide (50 mg, 0.30 mmol, 1.9 equiv) in DMF (10 mL) were stirred for 1 h at 50° C. The reaction mixture was diluted with CH2Cl2(100 mL), washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:1) to provide 100 mg (78%) of 249-4 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (249-5) 249-4 (150 mg, 0.19 mmol, 1 equiv) and TFA (1 mL) in CH2Cl2(10 mL) were stirred for 1 h at rt. The reaction mixture was concentrated and the residue purified by prep-HPLC with the following conditions—mobile phase: water (0.05% TFA) and CH3CN (66% Phase B up to 74% in 8 min); detector: UV. This resulted in 38.3 mg (32%) of 249-5 as a white solid. MS (ES, m/z) [M+H]+=627.55;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.90 (d, J=9.8 Hz, 1H), 0.96-1.29 (m, 15H), 1.44 (d, J=8.1 Hz, 5H), 1.54 (t, J=13.0 Hz, 2H), 1.60-1.99 (m, 6H), 2.22 (s, 5H), 2.54 (s, 1H), 2.83 (d, J=13.8 Hz, 1H), 3.26 (s, 3H), 3.64 (dd, J=11.8, 4.2 Hz, 1H), 4.92 (s, 1H), 5.13 (d, J=14.0 Hz, 1H), 5.61 (s, 1H). Example 32 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (252-2) Synthesis of 2-Benzhydryl 9-((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (252-1) 249-3 (300 mg, 0.44 mmol, 1 equiv), 4-(bromomethyl)-5-(propan-2-yl)-2H-1,3-dioxol-2-one (100 mg, 0.45 mmol, 1.03 equiv), K2CO3(200 mg, 1.45 mmol, 3.3 equiv), and potassium iodide (50 mg, 0.30 mmol, 0.68 equiv) in DMF (30 mg, 0.41 mmol, 0.93 equiv) were stirred for 1 h at 50° C. The reaction mixture was diluted with CH2Cl2(100 mL), washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column eluting with EtOAc/petroleum ether (1:1) to provide 190 mg (52%) of 252-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (252-2) 252-1 (190 mg, 0.23 mmol, 1 equiv) and TFA (1 mL) in CH2Cl2(10 mL) were stirred for 1 h at rt. The reaction was concentrated and purified by prep-TLC (PE/EtOAc; 1:1). This resulted in 68.6 mg (45%) of 252-2 as a white solid. MS (ES, m/z): [M+H]+=657.25;1H NMR (400 MHz, chloroform-tf) δ 0.85 (s, 4H), 1.00 (d, J=12.8 Hz, 1H), 1.07 (s, 1H), 1.10-1.41 (m, 20H), 1.36-1.57 (m, 8H), 1.65-1.92 (m, 6H), 2.01 (s, 3H), 2.21 (d, J=10.9 Hz, 1H), 2.43 (s, 1H), 2.90 (d, J=13.8 Hz, 1H), 3.03 (p, J=7.2 Hz, 1H), 3.27 (s, 3H), 3.60 (dd, J=11.8, 4.4 Hz, 1H), 4.88 (d, J=13.8 Hz, 1H), 5.02 (d, J=13.8 Hz, 1H), 5.74 (s, 1H). Example 33 (2S,4aS,6aS,6bR,8aR,9R,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-Hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (253-4) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-((phenoxycarbonothioyl)oxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (253-1) 194-8 (120 mg, 0.15 mmol, 1 equiv), phenyl chloromethanethioate (133.0 mg, 0.77 mmol, 5 equiv), and DMAP (37.6 mg, 0.31 mmol, 2 equiv) in CH2Cl2(5 mL) were stirred for 2 days at 40° C. The reaction mixture was concentrated and the residue purified by prep-TLC (petroleum ether/EtOAc; 5:1) to afford 253-1 (100 mg, 71%) as a light-yellow solid. Synthesis of (4R,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((Benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (253-2) 253-1 (100 mg, 0.11 mmol, 1 equiv), azobisisobutyronitrile (43.1 mg, 0.26 mmol, 2.4 equiv), and tributyltin hydride (139.4 mg, 0.48 mmol, 4.4 equiv) in toluene (3 mL) were stirred overnight at 110° C. The reaction mixture was concentrated, diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, and concentrated. The crude 253-2 was used in the next step directly without further purification. Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9R,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (253-3) 253-2 (100 mg, 0.15 mmol, 1 equiv), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (45.6 mg, 0.31 mmol, 2 equiv), K2CO3(63.7 mg, 0.46 mmol, 3 equiv), and potassium iodide (12.8 mg, 0.08 mmol, 0.5 equiv) in DMF (3 mL) were stirred for 2 h at 60° C. The reaction was diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, and concentrated. The crude 253-3 was used in the next step directly without further purification. Synthesis of (2S,4aS,6aS,6bR,8aR,9R,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (253-4) 253-3 (100 mg, 0.13 mmol, 1 equiv) and TFA (0.1 mL, 1.35 mmol, 10 equiv) in CH2Cl2(5 mL) were stirred for 1 h at rt. The reaction mixture was concentrated and the residue purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (68% Phase B up to 80% in 8 min); detector: UV. This resulted in 253-4 (8.1 mg, 10%) as a colorless oil. MS (ES, m/z) [M+H]+=597.15;1H NMR (400 MHz, chloroform-7) δ 6.89 (d, J=102.7 Hz, 2H), 5.74 (s, 1H), 5.00 (d, J=13.8 Hz, 1H), 4.75 (d, J=13.8 Hz, 1H), 2.78 (d, J=13.1 Hz, 1H), 2.52 (s, 1H), 2.22 (s, 4H), 2.11-1.89 (m, 3H), 1.88-1.47 (m, 9H), 1.47-1.31 (m, 8H), 1.26 (s, 4H), 1.20 (d, J=7.8 Hz, 6H), 1.14 (s, 3H), 1.10-0.88 (m, 4H), 0.85 (s, 3H). Example 34 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Acetoxy-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (254-3) Synthesis of Benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-isocyanato-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (254-1) Triethylamine (0.135 mL, 0.97 mmol, 1.5 equiv) was added at rt to 210-1 (450 mg, 0.63 mmol, 1 equiv) and DPPA (0.225 mL, 1.04 mmol, 1.6 equiv) in anisole (5 mL). The reaction was stirred for 1.5 h at 90° C. The reaction mixture was concentrated under vacuum and the residue purified by prep-TLC (CH2Cl2/MeOH; 5:1) to afford 254-1 (400 mg, 89%) as a light-yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (254-2) 4-(Hydroxymethyl)-5-methyl-2H-1,3-dioxol-2-one (66.3 mg, 0.51 mmol, 3 equiv) in CH2Cl2was added dropwise at rt to 254-1 (120 mg, 0.17 mmol, 1 equiv) and chlorotrimethylsilane (92.3 mg, 0.85 mmol, 5 equiv) in CH2Cl2(5 mL). The reaction slurry was stirred overnight at rt and then concentrated. The residue was purified by prep-TLC (petroleum ether/EtOAc 2:1) to afford 254-2 (120 mg, 84%) as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S, 12aS, 12bR,14bR)-10-acetoxy-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (254-3) 254-2 (120 mg, 0.14 mmol, 1 equiv) and TFA (0.1 mL, 1.35 mmol, 9.4 equiv) in CH2Cl2were stirred for 1 h at rt. The reaction mixture was concentrated under vacuum and the residue purified by prep-TLC to afford 254-3 (21.2 mg, 22%) as an off-white solid. MS (ES, m z): [M+H]+=670.25;1H NMR (400 MHz, chloroform-d) δ 6.94 (d, J=67.6 Hz, 2H), 5.75 (s, 1H), 5.47 (t, J=5.7 Hz, 1H), 4.77 (s, 2H), 4.44 (s, 1H), 2.81 (d, J=13.5 Hz, 1H), 2.50 (s, 1H), 2.21 (s, 2H), 2.05 (s, 3H), 1.91-1.53 (m, 8H), 1.42 (d, J=19.1 Hz, 8H), 1.26 (s, 5H), 1.19-0.97 (m, 11H), 0.86 (s, 3H). Example 35 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Acetoxy-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (255-2) Synthesis of Benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (255-1) 4-(Hydroxymethyl)-5-(propan-2-yl)-2H-1,3-dioxol-2-one (67.2 mg, 0.42 mmol, 3 equiv) in CH2Cl2was added dropwise at rt to 254-1 (100 mg, 0.14 mmol, 1 equiv) and chlorotrimethylsilane (76.9 mg, 0.71 mmol, 5 equiv) in CH2Cl2. The reaction slurry was stirred overnight at rt and concentrated. The residue was purified by prep-TLC (petroleum ether/EtOAc 2:1) to afford 255-1 (100 mg, 82%) as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-Acetoxy-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (255-2) 255-1 (100 mg, 0.12 mmol, 1 equiv) and TFA (0.1 mL) in CH2Cl2(10 mL) were stirred for 1 h at rt. The mixture was concentrated under vacuum and the residue purified by prep-HPLC with the following conditions—Column: XSelect CSHPrep C18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and CH3CN (70% Phase B up to 85% in 7 min); detector: UV. This resulted in 255-2 (11.7 mg, 14%) as an off-white solid. MS (ES, m/z): [M+H]+=698.25;1H NMR (400 MHz, chloroform-7) δ 7.20-6.72 (m, 2H), 5.75 (s, 1H), 5.46 (dd, J=11.1, 5.0 Hz, 1H), 4.87 (d, J=13.8 Hz, 1H), 4.72 (d, J=14.0 Hz, 1H), 4.43 (s, 1H), 3.03 (p, J=6.9 Hz, 1H), 2.82 (d, J=13.5 Hz, 1H), 2.50 (s, 1H), 2.22 (d, J=12.2 Hz, 2H), 2.13-1.91 (m, 6H), 1.90-1.53 (m, 6H), 1.42 (d, J=24.7 Hz, 8H), 1.31-1.21 (m, 10H), 1.21-0.99 (m, 12H), 0.86 (s, 3H). Example 36 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-Difluoroacetoxy)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (256-2) Synthesis of 2-Benzhydryl 9-((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-difluoroacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (256-1) EDCI (178.1 mg, 5 equiv) was added to 209-1 (150 mg, 1 equiv), 2,2-difluoroacetic acid (0.0234 mL, 2 equiv), and DMAP (11.4 mg, 0.5 equiv) in CH2Cl2(10 mL) and the reaction slurry stirred for 1 h at rt. The reaction mixture was concentrated and the residue purified by silica gel column with EtOAc/petroleum ether (1:3) to provide 234.2 mg (100%) of 256-1 as an off-white semi-solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-Difluoroacetoxy)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (256-2) 256-1 (164 mg, 1 equiv) and TFA (1 mL) in CH2Cl2(10 mL) were stirred for 1 h at rt. The reaction mixture was concentrated and the residue purified by prep-HPLC with the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (65% Phase B up to 85% in 8 min); detector: UV. This resulted in 74.8 mg (56%) of 256-2 as a white solid. MS (ES, m/z): [M+H]+=719.20;1H NMR (400 MHz, methanol-d4) δ 0.83 (s, 3H), 0.96 (d, J=9.6 Hz, 1H), 1.05 (d, J=14.4 Hz, 1H), 1.17 (s, 3H), 1.18-1.32 (m, 17H), 1.38-1.49 (m, 7H), 1.63-1.91 (m, 8H), 1.94 (d, J=9.5 Hz, 1H), 2.09-2.29 (m, 2H), 2.57 (s, 1H), 2.86 (d, J=14.0 Hz, 1H), 3.05 (h, 7=13.6 Hz, 1H), 4.88 (d, J=14.0 Hz, 1H), 5.06 (d, J=14.0 Hz, 1H), 5.34 (dd, J=12.0, 5.2 Hz, 1H), 5.60 (s, 1H), 6.04 (t, 7=53.0 Hz, 1H). Example 37 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (258-2) Synthesis of 2-Benzhydryl 9-((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (258-1) Bromomethyl methyl ether (0.0687 mL, 4.0 equiv) was added dropwise with stirring at 0° C. to 209-1 (150 mg, 0.19 mmol, 1 equiv) and iPr2EtN (0.307 mL, 10 equiv) in CH2Cl2(10 mL) and then stirred for 2 h at 60° C. The reaction mixture was concentrated and the residue purified by silica gel column eluting with EtOAc/petroleum ether (1:2) to provide 158 mg (100%) of 258-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (258-2) 258-1 (158 mg, 1 equiv) and TFA (0.2068 mL, 15.0 equiv) in CH2Cl2(15 mL) were stirred for 6 h at rt. The reaction mixture was washed with brine (3×20 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by prep-HPLC with the following conditions—Column: XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and CH3CN (65-85% Phase B in 7 min); detector: UV. This resulted in 55.8 mg (44%) of 258-2 as a white solid. MS (ES, m/z): [M+H]+=685.15;1H NMR (400 MHz, methanol-d4) δ 0.82 (s, 3H), 0.87 (d, J=10.0 Hz, 1H), 1.04 (d, J=13.6 Hz, 1H), 1.10-1.20 (m, 13H), 1.24 (d, J=6.8 Hz, 7H), 1.34 (d, J=8.4 Hz, 2H), 1.38 (d, J=14.8 Hz, 6H), 1.54 (d, J=10.4 Hz, 1H), 1.61-1.71 (m, 3H), 1.73-1.90 (m, 3H), 1.95 (d, J=10.0 Hz, 1H), 2.16 (ddd, J=30.3, 16.8, 4.8 Hz, 2H), 2.51 (s, 1H), 2.78 (d, J=13.6 Hz, 1H), 3.10 (p, 7=6.9 Hz, 1H), 3.25 (s, 3H), 3.95 (dd, J=12.0, 4.4 Hz, 1H), 4.48 (d, J=7.2 Hz, 1H), 4.63 (d, J=6.8 Hz, 1H), 4.83 (d, J=14.0 Hz, 1H), 5.15 (d, J=14.0 Hz, 1H), 5.59 (s, 1H). Example 38 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (264-1) The title compound was prepared according to the methods for compound 194-10, beginning with 194-8 and 2,5,8,1 l-tetraoxatetradecan-14-oic acid. The crude product was purified by prep-HPLC with the following conditions: Column, XBridge Prep OBD Cl8, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (55.0% CH3CN up to 69.0% in 8 min); detector, UV 220 nm. This resulted in 103.0 mg (55%) of 264-1 as a white solid. MS (ES, m/z): [M+H]+=848.25.1H NMR (400 MHz, methanol-d4) δ 0.86 (s, 3H), 0.96 (s, 1H), 1.07 (d, J=13.9 Hz, 1H), 1.10-1.29 (m, 14H), 1.35-1.50 (m, 7H), 1.65-1.80 (m, 6H), 1.81-1.91 (m, 2H), 1.95 (d, J=9.6 Hz, 1H), 2.22 (s, 5H), 2.49 (t, J=10.8 Hz, 2H), 2.56 (s, 1H), 2.81 (d, J=13.6 Hz, 1H), 3.35 (s, 3H), 3.51-3.71 (m, 14H), 4.88 (s, 1H), 5.01 (d, J=14.0 Hz, 1H), 5.18 (dd, J=11.6, 4.8 Hz, 1H), 5.60 (s, 1H). Example 39 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxolan-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (265-2) Synthesis of 2-benzyl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (265-1) Into a 50-mL round-bottom flask was placed 190-2 (220 mg, 1 equiv), KI (22.5 mg, 0.5 equiv), DMF (1 mL), K2CO3(112.7 mg, 3 equiv), and 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (72.7 mg, 7.2 equiv). The reaction slurry was stirred for 2 days at 60° C. The reaction mixture was extracted with ethyl acetate and the organic layer washed with 3×100 ml of H2O and 1×100 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with CH2Cl2/methanol (30:1) to provide 248.6 mg (99%) of 265-1 as pale yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxolan-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (265-2) Into a 25-mL round-bottom flask purged and maintained with an inert atmosphere of H2(1 atm) was placed 265-1 (124 mg, 1 equiv), THF (5 mL), and Pd(OH)2/C (49.6 mg). The reaction slurry was stirred for 1.5 hr at room temperature. The solids were filtered off and the filtrate concentrated. The residue was purified by prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD, 5 μm, 19*150 mm; mobile phase, water (0.05% TFA) and CH3CN (hold 5% Phase B in 0 min, up to 55% in 1 min,up to 71% in 8 min); detector, UV. This resulted in 19.9 mg (18%) of 265-2 as an off-white semi-solid. MS (ES, m/z): [M+H]+=833.25;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 1.06 (d, J=10.4 Hz, 2H), 1.16 (s, 3H), 1.20 (s, 6H), 1.24-1.30 (m, 5H), 1.39-1.46 (m, 4H), 1.47-1.53 (m, 6H), 1.62-1.82 (m, 5H), 1.83-2.01 (m, 4H), 2.13-2.28 (m, 2H), 2.54 (q, J=6.4 Hz, 2H), 2.60 (s, 1H), 2.79-2.90 (m, 1H), 3.38 (s, 3H), 3.55-3.59 (m, 2H), 3.59-3.61 (m, 2H), 3.62-3.69 (m, 8H), 3.69-3.73 (m, 2H), 4.18-4.30 (m, 1H), 4.38-4.50 (m, 1H), 4.95-5.11 (m, 2H), 5.15-5.25 (m, 1H), 5.62 (s, 1H). Example 40 (2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-10-(2-((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-oxoethoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (279-2) Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-10-(2-((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-oxoethoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (279-1) Into a 50-mL round-bottom flask was placed 78-1 (100 mg, 1 equiv), DMF (1 mL), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (38.5 mg, 1.8 equiv), KI (11.9 mg, 0.5 equiv), and K2CO3(59.6 mg, 3 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate, washed with 3×20 ml of H2O and 1×20 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:3) to provide 107.5 mg (93%) of 279-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-heptamethyl-10-(2-((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-oxoethoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (279-2) Into a 50-mL round-bottom flask was placed 279-1 (107.5 mg, 1 equiv), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD, 5 μm, 19*150 mm; mobile phase, water (0.05% TFA) and CH3CN (hold 5% Phase B in 0 min, up to 68% in 1 min, up to 85% in 8 min); detector, UV. This resulted in 34.9 mg (41%) of 279-2 as a white solid. MS (ES, m/z) [M+H]+=641.25;1H NMR (400 MHz, chloroform-7) δ 0.70 (d, J=11.2 Hz, 1H), 0.84 (s, 7H), 1.01 (s, 1H), 1.05 (s, 3H), 1.14 (d, J=12 Hz, 6H), 1.22 (s, 4H), 1.36 (s, 4H), 1.39-1.49 (m, 4H), 1.57-1.76 (m, 5H), 1.79-2.09 (m, 4H), 2.19 (s, 4H), 2.33 (s, 1H), 2.83 (d, J=13.2 Hz, 1H), 2.94 (dd,7=11.6, 4.4 Hz, 1H), 4.16 (t, 7=18.8 Hz, 2H), 4.89 (d, J=10.6 Hz, 2H), 5.71 (s, 1H). Example 41 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (280-7) Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,12aS,12bR,14bR,E)-10-(hydroxyimino)-9-(hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (280-1) Into a 250-mL round-bottom flask was placed 194-3 (3.0 g, 4.45 mmol, 1 equiv), MeOH (50 mL), and Na2CO3(2.4 g, 22.6 mmol, 5.1 equiv). The reaction slurry was stirred for 48 hr at room temperature. The reaction mixture was concentrated, diluted in 500 mL of CH2Cl2, and the pH adjusted to 4 with 2 M HCl(aq). The organic layer was washed with 2×500 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 2.46 g (94%) of 280-1 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-9-(hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (280-2) Into a 500-mL round-bottom flask was placed 280-1 (2.48 g, 4.20 mmol, 1 equiv), MeOH (250 mL), NH4OAC (4.9 g, 63.6 mmol, 15 equiv), NaBH3CN (3.2 g, 51 mmol, 12 equiv), TiCl3(18 mL) at 0° C. The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with 500 mL of ethyl acetate and the pH adjusted to 12 with 4 M NaOH(aq). The organic layer was washed with 3×500 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 1.77 g (73%) of crude 280-2 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((tert-butoxycarbonyl)amino)-9-(hydroxymethyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (280-3) Into a 3000-mL round-bottom flask was placed 280-2 (34.6 g, 53 mmol), CH2Cl2(100 mL), BoC2O (23.2 g, 106 mmol, 2 equiv), and Et3N (36.9 mL, 265 mmol, 5 equiv). The reaction slurry was stirred for 1 hr at room temperature. The reaction mixture was extracted with 1000 mL of CH2Cl2and washed with 3×1000 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:5) to provide 18.3 g (46%) of 280-3 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((tert-butoxycarbonyl)amino)-9-formyl-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (280-4) Into a 1000-mL round-bottom flask was placed 280-3 (27.0 g, 35.9 mmol), CH2Cl2(150 mL), pH 8.6 buffer (150 mL), TEMPO (28.0 g, 179 mmol, 5 equiv), TBACl (39.9 g, 144 mmol, 4 equiv), and NCS (33.6 g, 251 mmol, 7 equiv). The reaction slurry was stirred for 2 hr at 40° C. The reaction mixture was washed with 3×500 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:3) to provide 19.3 g (72%) of 280-4 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-((tert-butoxycarbonyl)amino)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (280-5) Into a 100-mL round-bottom flask was placed 280-4 (1.57 g, 2.1 mmol), t-BuOH (10 mL), H2O (5 mL), and 2-methylbut-2-ene (1 mL). To this slurry was added NaH2PO4(2.01 g, 16.8 mmol, 8 equiv) at 0° C. followed by NaClO2(1.51 g, 16.7 mmol, 8 equiv) at 0° C. The reaction slurry was stirred for 1 hr at room temperature. The reaction mixture was extracted with 500 mL of CH2Cl2. The solution pH was adjusted to 4 with 2 M HCl(aq)and then washed with 3×500 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 1.67 g (quant) of crude 280-5 as a yellow solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((tert-butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (280-6) Into a 100-mL round-bottom flask was placed 280-5 (1.67 g, 2.18 mmol), DMF (10 mL), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (1.62 g, 11 mmol, 5 equiv), KI (0.36 g, 2.2 mmol, 1 equiv), and K2CO3(1.50 g, 11 mmol, 5 equiv). The reaction slurry was stirred for 1 hr at 60° C. The reaction mixture was diluted with 500 mL of CH2Cl2, washed with 5×500 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:4) to provide 1.07 g (56%) of 280-6 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (280-7) Into a 100-mL round-bottom flask was placed 280-6 (100 mg, 0.11 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature. The reaction mixture was concentrated, and the crude product was purified by prep-HPLC with the following conditions: Column, XBridge Prep C18 OBD, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (40% Phase B up to 73% in 7 min); detector, UV. This resulted in 8.5 mg (12%) of 280-7 as a white solid. MS (ES, m/z): [M+H]+=612.25;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.99-1.11 (m, 2H), 1.13-1.32 (m, 14H), 1.44 (d, J=18.2 Hz, 7H), 1.49-1.58 (m, 1H), 1.60-1.92 (m, 7H), 1.97 (d, J=9.6 Hz, 1H), 2.11-2.37 (m, 5H), 2.58 (s, 1H), 2.81-2.99 (m, 1H), 3.65-3.80 (m, 1H), 4.90-4.99 (m, 1H), 5.12-5.28 (m, 1H), 5.62 (s, 1H). Example 42 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((methoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (281-3) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((tert-butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (281-1) Into a 500-mL round-bottom flask, was placed PH-RDX-013-869-8 (10.1 g, 11.50 mmol, 1 equiv), DCM (150 mL), 2,6-dimethylpyridine (6.6 mL, 5.0 equiv). This was followed by the addition of (10.2 g, 46.01 mmol, 4.0 equiv) at 0° C. The resulting solution was stirred for 1.5 hr at room temperature. The resulting solution was extracted with 3×200 mL of dichioromethane. The resulting mixture was washed with 3 x500 ml of 2M HCl. The mixture was dried over anhydrous sodium sulfate. The solids were filtered out. The resulting mixture was concentrated. This resulted in 10.0728 g (112.56%) of 281-1 as a yellow solid. MS (ES, m/z): [M+H]+=778.30;1H NMR (400 MHz, DMSO-d6) δ 0.62 (s, 3H), 0.83-0.90 (m, 1H), 1.00 (d, J=18.4 Hz, 7H), 1.12 (s, 4H), 1.13-1.18 (m, 4H), 1.20-1.58 (m, 10H), 1.60-1.73 (m, 3H), 1.75-1.96 (m, 4H), 2.00-2.20 (m, 5H), 2.42 (s, 1H), 2.65 (d, J=13.7 Hz, 1H), 3.39 (dd, J=11.4, 5.1 Hz, 1H), 4.93 (d, J=14.0 Hz, 1H), 5.02-5.14 (m, 1H), 5.28 (s, 1H), 6.86 (s, 1H), 7.24-7.44 (m, 10H), 8.31 (d, J=9.2 Hz, 1H). Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((methoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (281-2) Into a 100-mL round-bottom flask was placed 281-1 (230 mg, 0.30 mmol), CH2Cl2(5 mL), methyl chloroformate (280 mg, 3 mmol, 10 equiv), and Et3N (0.33 mL, 2.4 mmol, 8 equiv). The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was diluted with 300 mL of CH2Cl2and washed with 3×300 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 130 mg (53%) of crude 281-2 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((methoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (281-3) Into a 100-mL round-bottom flask was placed 281-2 (100 mg, 0.12 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XBridge Prep C18 OBD, 5 μm, 19*150 mm; mobile phase, water (0.05% TFA) and CH3CN (43% Phase B up to 73% in 8 min); detector, UV. This resulted in 24.8 mg (29%) of 281-3 as a white solid. MS (ES, m/z): [M+H]+=670.15;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.91 (d, J=12.7 Hz, 1H), 1.05-1.34 (m, 16H), 1.33-1.57 (m, 8H), 1.59-1.82 (m, 5H), 1.87 (dd, J=12.6, 6.2 Hz, 2H), 1.97 (d, J=9.7 Hz, 1H), 2.11-2.29 (m, 5H), 2.58 (s, 1H), 2.75-2.84 (m, 1H), 3.58 (s, 3H), 3.98 (dd, J=12.5, 4.3 Hz, 1H), 4.87 (s, 1H), 5.01 (d, J=14.0 Hz, 1H), 5.61 (s, 1H). Example 43 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(pentanoyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (282-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(pentanoyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (282-1) Into a 100-mL round-bottom flask was placed 194-8 (120 mg, 0.15 mmol), CH2Cl2(8 mL), pentanoic acid (0.17 mL), DMAP (75 mg, 0.61 mmol, 4 equiv), and EDCI (150 mg, 0.78 mmol, 5.2 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with 300 mL of CH2Cl2and the pH of the solution adjusted to 4 with 2 M HCl(aq). The resulting mixture was washed with 3×500 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 115 mg (86%) of crude 282-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(pentanoyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (282-2) Into a 100-mL round-bottom flask was placed 282-1 (120 mg, 0.14 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature and concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XBridge Prep C18 OBD, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (65% Phase B up to 95% in 7 min); detector, UV. This resulted in 54.1 mg (53%) of 282-2 as a white solid. MS (ES, m/z): [M+H]+=697.20;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.93 (t, J=7.3 Hz, 4H), 1.06 (d, J=14.5 Hz, 1H), 1.19 (dd, J=16.9, 15.8 Hz, 14H), 1.27-1.39 (m, 2H), 1.35-1.49 (m, 6H), 1.48-1.60 (m, 2H), 1.61-1.81 (m, 6H), 1.81-2.01 (m, 3H), 2.05 (s, 1H), 2.11-2.29 (m, 7H), 2.58 (s, 1H), 2.77-2.87 (m, 1H), 4.89 (s, 1H), 5.02 (d, J=13.9 Hz, 1H), 5.17 (dd, J=11.7, 5.0 Hz, 1H), 5.62 (s, 1H). Example 44 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(4-methylpiperazin-1-yl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (283-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(2-(4-methylpiperazin-1-yl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (283-1) Into a 100-mL round-bottom flask was placed 194-8 (120 mg, 0.15 mmol, 1 equiv), CH2Cl2(6 mL), 2-(4-methylpiperazin-1-yl)acetic acid (240 mg, 1.52 mmol, 10 equiv), DMAP (94 mg, 0.77 mmol, 5.1 equiv), and EDCI (300 mg, 1.56 mmol, 10 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with 300 mL of CH2Cl2and the pH of the solution adjusted to 4 with 2 M HCl(aq). The resulting mixture was washed with 3 x 300 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 125 mg (88%) of 283-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(4-methylpiperazin-1-yl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (283-2) Into a 100-mL round-bottom flask was placed 283-1 (125 mg, 0.14 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XBridge Prep C18 OBD, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (30% Phase B up to 68% in 7 min); detector, UV. This resulted in 59.5 mg (58%) of 283-2 as a white solid. MS (ES, m/z): [M+H]+=753.25;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.98-1.30 (m, 17H), 1.30-1.53 (m, 8H), 1.54-2.09 (m, 10H), 2.12-2.28 (m, 5H), 2.58 (s, 1H), 2.63-3.03 (m, 8H), 3.34 (s, 3H), 4.94 (d, J=14.0 Hz, 1H), 5.01 (d, J=13.9 Hz, 1H), 5.24 (dd, J=11.7, 4.9 Hz, 1H), 5.62 (s, 1H). Example 45 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((3-morpholinopropanoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (284-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((3-morpholinopropanoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (284-1) Into a 100-mL round-bottom flask was placed 194-8 (150 mg, 0.19 mmol), CH2Cl2(8 mL), 3-(morpholin-4-yl)propanoic acid (310 mg, 1.95 mmol, 10 equiv), DMAP (75 mg, 0.61 mmol, 3.2 equiv), and EDCI (200 mg, 1.04 mmol, 5.4 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with 300 mL of CH2Cl2and the pH of the solution was adjusted to 4 with 2 M HCl(aq). The resulting mixture was washed with 3×300 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 143 mg (81%) of crude 284-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((3-morpholinopropanoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (284-2) Into a 100-mL round-bottom flask was placed 284-1 (143 mg, 0.16 mmol), CH2Cl2(139 mL), TFA (13.9 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XBridge Prep Cl8 OBD, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (35% Phase B up to 75% in 7 min); detector, UV. This resulted in 10.3 mg (8%) of 284-2 as a white solid. MS (ES, m/z): [M+H]+=754.45;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.94-1.30 (m, 16H), 1.44 (d, J=18.7 Hz, 7H), 1.67-2.08 (m, 9H), 2.14-2.30 (m, 5H), 2.58 (s, 1H), 2.72-2.98 (m, 3H), 3.15-2.30 (m, 2H), 3.45 (s, 1H), 3.40-3.50 (m, 3H), 3.65-4.28 (m, 4H), 4.99 (d, J=1.9 Hz, 2H), 5.23 (dd, J=11.6, 5.1 Hz, 1H), 5.62 (s, 1H). Example 46 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(methylsulfonamido)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (285-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(methylsulfonamido)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (285-1) Into a 100-mL round-bottom flask was placed 281-1 (230 mg, 0.30 mmol) and CH2Cl2(6 mL) followed by methanesulfonyl chloride (0.5 mL) then Et3N (0.3 mL, 2.2 mmol, 7.3 equiv) at 0° C. The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was diluted with 300 mL of CH2Cl2and washed with 3×300 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 201 mg (79%) of crude 285-1 as a yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(methylsulfonamido)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (285-2) Into a 100-mL round-bottom flask was placed 285-1 (200 mg, 0.23 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XBridge Prep C18 OBD, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (5% Phase B up to 45% in 5 min, up to 61% in 10 min); detector, UV. This resulted in 16.2 mg (10%) of 285-2 as a white solid. MS (ES, m/z): [M+H]+=690.05;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.92 (d, J=11.7 Hz, 1H), 1.00-1.31 (m, 16H), 1.31-1.49 (m, 7H), 1.56-1.81 (m, 6H), 1.81-2.01 (m, 3H), 2.08-2.30 (m, 5H), 2.58 (s, 1H), 2.82 (dt, J=13.6, 3.5 Hz, 1H), 2.91 (s, 3H), 3.76 (dd, J=11.9, 4.9 Hz, 1H), 4.82 (d, J=13.9 Hz, 1H), 5.09 (d, J=14.0 Hz, 1H), 5.62 (s, 1H). Example 47 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(((2-morpholinoethyl)carbamoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (286-4) Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-formyl-2,4a,6a,6b,9,12a-hexamethyl-10-(((2-morpholinoethyl)carbamoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (286-1) Into a 100-mL round-bottom flask was placed 194-6 (190 mg, 0.29 mmol) and CH2Cl2(5 mL) followed by triphosgene (69 mg, 0.23 mmol, 0.8 equiv) at 0° C. To this slurry was added Et3N (0.081 mL, 0.58 mmol, 2 equiv) dropwise. After 1 hour, 2-(morpholin-4-yl)ethan-1-amine (380 mg, 2.9 mmol, 10 equiv) was added at 0° C. The reaction slurry was stirred for 1 hr at 0° C. The reaction mixture was diluted with 300 mL of CH2Cl2and washed with 3×300 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 170 mg (72%) of 286-1 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-3-(((2-morpholinoethyl)carbamoyl)oxy)-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (286-2) Into a 100-mL round-bottom flask was placed 286-1 (170 mg, 0.21 mmol), t-BuOH (6 mL), H2O (2 mL), and 2-methylbut-2-ene (0.5 mL) followed by the addition of NaH2PO4(250 mg, 2.1 mmol, 10 equiv) at 0° C. To this slurry was added NaClO2(190 mg, 2.1 mmol, 10 equiv) at 0° C. The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was diluted with 300 mL of CH2Cl2and washed with 3×300 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 168 mg (97%) of 286-2 as a white solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(((2-morpholinoethyl)carbamoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (286-3) Into a 100-mL round-bottom flask was placed 286-2 (210 mg, 0.26 mmol), DMF (8 mL), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (200 mg, 1.35 mmol, 5.3 equiv), KI (43 mg, 0.26 mmol, 1 equiv), and K2CO3(180 mg, 1.3 mmol, 5.1 equiv). The reaction slurry was stirred for 1 hr at 60° C. The reaction slurry was cooled to room temperature, diluted with 300 mL of CH2Cl2, and washed with 5×300 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 108 mg (45%) of 286-3 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(((2-morpholinoethyl)carbamoyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (286-4) Into a 100-mL round-bottom flask was placed 286-3 (108 mg, 0.12 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD, 5 μm, 19*150 mm; mobile phase, water (0.05% TFA) and CH3CN (32% Phase B up to 49% in 8 min); detector, UV. This resulted in 4.0 mg (4%) of 286-4 as a white solid. MS (ES, m/z): [M+H]+=769.25;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.99 (s, 1H), 1.07 (d, J=13.7 Hz, 1H), 1.11-1.37 (m, 15H), 1.38-1.40 (m, 7H), 1.62-1.83 (m, 6H), 1.83-1.94 (m, 3H), 2.12-2.28 (m, 5H), 2.57 (s, 1H), 2.79-2.90 (m, 1H), 3.20-3.30 (m, 3H), 3.35-1.42 (m, 2H), 3.51-3.61 (m, 2H), 3.69-3.84 (m, 2H), 3.97-4.21 (m, 2H), 4.92-5.10 (m, 3H), 5.50 (s, 1H), 5.62 (s, 1H). Example 48 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methylsulfinyl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (289-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(2-(methylsulfinyl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (289-1) A mixture of 194-9 (100 mg, 0.12 mmol) and NaIO4(27.1 mg, 0.13 mmol, 1.1 equiv) in MeOH and H2O (0.11 mL) was stirred overnight at room temperature. The reaction mixture was extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (petroleum ether/EtOAc 5:1) to afford 289-1 (110 mg, quant) as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-(methyl sulfinyl)acetoxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (289-2) A mixture of 289-1 (110 mg, 0.12 mmol) and TFA (0.1 mL, 1.35 mmol, 11 equiv) in CH2Cl2was stirred for 1 h at room temperature. The resulting mixture was concentrated under vacuum. The crude product was purified by prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD, 19*250 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (51% Phase B up to 62% in 8 min); detector, UV. This resulted in 289-2 (27.7 mg, 31%) as an off-white solid. MS (ES, m/z): [M+H]+=717;1H NMR (400 MHz, methanol-d4) δ 5.62 (s, 1H), 5.31 (dd, J=11.4, 5.4 Hz, 1H), 5.06 (d, J=14.0 Hz, 1H), 4.91 (d, J=4.2 Hz, 1H), 3.93 (d, 7=14.3 Hz, 1H), 3.73 (dd, J=14.3, 5.3 Hz, 1H), 2.85 (d, J=13.5 Hz, 1H), 2.75 (d, J=2.8 Hz, 3H), 2.59 (s, 1H), 2.30-2.10 (m, 5H), 1.97 (d, J=9.8 Hz, 1H), 1.94-1.62 (m, 8H), 1.53-1.36 (m, 7H), 1.34-1.12 (m, 14H), 1.07 (d, J=14.0 Hz, 1H), 0.99 (d, J=9.8 Hz, 1H), 0.85 (s, 3H). Example 49 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((dimethylglycyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (290-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((dimethylglycyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (290-1) Into a 25-mL round-bottom flask was placed 194-8 (140 mg, 0.18 mmol), 2-(dimethylamino)acetic acid (185 mg, 1.8 mmol, 10 equiv), 4-dimethylaminopyridine (84 mg, 0.69 mmol, 4 equiv), CH2Cl2(2.5 mL), EDCI (175 mg, 0.91 mmol, 5 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was concentrated under vacuum. The residue was applied onto a silica gel column with CH2Cl2/methanol (12/1) to provide 100 mg (64%) of 290-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((dimethylglycyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (290-2) A mixture 290-1 (100 mg, 0.12 mmol) and TFA (0.1 mL, 1.35 mmol, 12 equiv) in CH2Cl2was stirred for 1 h at room temperature. The reaction mixture was concentrated under reduced pressure. The crude product was purified by prep-HPLC with the following conditions; column: Xselect CSH OBD 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (60 mL/min; gradient: 30% B to 60% B in 8 min; 254 nm; Rt: 6.27 min). The residue was repurified by prep-TLC to afford 290-2 (26.6 mg, 33%) as an off-white solid. MS (ES, m/z) [M+H]+=698.05;1H NMR (300 MHz, methanol-d4) δ 5.64 (s, 1H), 5.45-5.31 (m, 1H), 5.10 (d, J=13.9 Hz, 1H), 4.95 (d, J=14.1 Hz, 1H), 4.16 (d, J=2.4 Hz, 2H), 2.98 (s, 6H), 2.89 (d, J=14.1 Hz, 1H), 2.60 (s, 1H), 2.22 (s, 5H), 1.92 (d, J=32.7 Hz, 5H), 1.82-1.66 (m, 4H), 1.46 (d, J=12.1 Hz, 7H), 1.28 (s, 5H), 1.25-1.14 (m, 9H), 1.14-0.95 (m, 2H), 0.86 (s, 3H). Example 50 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((acetylglycyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (291-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((acetylglycyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (291-1) Into a 25-mL round-bottom flask was placed 194-8 (140 mg, 0.18 mmol), 2-acetamidoacetic acid (110 mg, 0.94 mmol, 5 equiv), 4-dimethylaminopyridine (84 mg, 0.69 mmol, 4 equiv), CH2Cl2(2.5 mL), and EDCI (175 mg, 0.91 mmol, 5 equiv). The reaction mixture was stirred overnight at room temperature and concentrated under vacuum. The residue was applied onto a silica gel column with CThCk/methanol (13/1) to provide 170 mg (quant) of 291-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((acetylglycyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (291-2) A mixture of 291-1 (170 mg, 0.19 mmol) and TFA (0.15 mL, 2 mmol, 10 equiv) in CH2Cl2was stirred for 1 h at room temperature. The reaction mixture was concentrated under vacuum. The crude product was purified by prep-HPLC with the following conditions: column, Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase A: water (0.05% TFA), mobile phase B: CH3CN; Flow rate: 60 mL/min; Gradient: 30% B to 65% B in 8 min; 254 nm; Rt: 5.54 min. The residue was repurified by prep-TLC to afford 291-2 (27.1 mg, 20%) as an off-white solid. MS (ES, m z): [M+H]+=712;1H NMR (300 MHz, methanol-d4) δ 5.63 (s, 1H), 5.23 (dd, J=10.9, 5.7 Hz, 1H), 5.05 (d, J=14.0 Hz, 1H), 4.92 (s, 1H), 3.86 (d, J=2.4 Hz, 2H), 2.85 (d, J=13.8 Hz, 1H), 2.59 (s, 1H), 2.21 (s, 5H), 2.04 (d, J=16.2 Hz, 5H), 1.88 (d, J=10.3 Hz, 2H), 1.74 (dd, J=21.5, 11.7 Hz, 6H), 1.45 (d, J=12.5 Hz, 7H), 1.21 (t, J=13.0 Hz, 15H), 1.08 (d, J=13.9 Hz, 1H), 0.96 (s, 1H), 0.86 (s, 3H). Example 51 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetamido-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetamido-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (294-1) Into a 100-mL round-bottom flask was placed the HCl salt of 281-1 (200 mg, 0.26 mmol), CH2Cl2(5 mL), Et3N (0.29 mL, 2.1 mmol, 8 equiv), and acetyl chloride (0.18 mL, 10 equiv). The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was diluted with 300 mL of CH2Cl2and was washed with 3×300 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 150 mg (71%) of 294-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetamido-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (294-2) Into a 100-mL round-bottom flask was placed 294-1 (100 mg, 0.12 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XBridge Prep C18 OBD, 19-150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (40% Phase B up to 73% in 7 min); detector, uv. This resulted in 14.1 mg (17%) of 294-2 as a white solid. MS (ES, m/z): [M+1]+=654.15;1H NMR (400 MHz, methanol-d4) δ 5.61 (s, 1H), 5.04 (d, J=13.9 Hz, 1H), 4.74 (d, J=13.9 Hz, 1H), 4.24 (dd, J=12.6, 4.3 Hz, 1H), 2.84-2.75 (m, 1H), 2.58 (s, 1H), 2.27-2.11 (m, 5H), 2.05 (s, 1H), 1.97 (d, J=9.9 Hz, 1H), 1.89 (s, 4H), 1.87-1.72 (m, 5H), 1.72-1.60 (m, 2H), 1.47 (s, 4H), 1.42 (d, J=3.0 Hz, 3H), 1.40-1.36 (m, 1H), 1.30-1.21 (m, 2H), 1.21-1.13 (m, 12H), 1.06 (d, J=13.9 Hz, 1H), 0.90 (d, J=13.1 Hz, 1H), 0.85 (s, 3H). Example 52 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(allyloxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (297-5) Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-hydroxy-4,6a, 6b, 8a, 11,14b-hexamethyl -14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (297-1) Into a 100-mL round-bottom flask was placed 194-7 (200 mg, 0.30 mmol), CH2Cl2(5 mL, 0.06 mmol, 0.2 equiv), MeOH (2.5 mL, 0.08 mmol, 0.26 equiv), and TMSCHN2(1 mL, 0.01 mmol, 0.03 equiv). The reaction slurry was stirred for 1 hr at room temperature. The reaction mixture was concentrated to provide 200 mg (98%) of 297-1 as a colorless solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(allyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (297-2) Into a 100-mL round-bottom flask was placed 297-1 (292 mg, 0.43 mmol), allyl bromide (103 mg, 0.85 mmol, 2 equiv), TBAI (79.3 mg, 0.21 mmol, 0.5 equiv), THF (0.4 mL), and NaHMDS (0.429 mL, 0.85 mmol, 2 M in THF, 2 equiv). The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 2×150 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 190 mg (61%) of 297-2 as a colorless solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-3-(allyloxy)-11-((benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (297-3) Into a 100-mL round-bottom flask was placed 297-2 (220 mg, 0.31 mmol), lithium iodide (426 mg, 3.18 mmol, 10.4 equiv), and pyridine (5 mL). The reaction slurry was stirred for 2 days at 125° C. The reaction mixture was cooled to room temperature and the pH of the solution was adjusted to 5 with 1 M HCl(aq). The mixture was diluted with 100 mL of CH2Cl2and washed with 3×50 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with CH2Cl2/methanol (10:1) to provide 110 mg (51%) of 297-3 as a white solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(allyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (297-4) Into a 100-mL round-bottom flask was placed 297-3 (110 mg, 0.16 mmol), 4-(bromomethyl)-5-methyl-2H-1,3-dioxol-2-one (68.8 mg, 0.36 mmol, 2.3 equiv), K2CO3(64.5 mg, 0.47 mmol, 3 equiv), KI (12.9 mg, 0.08 mmol, 0.5 equiv), and DMF (5 mL). The reaction slurry was stirred for 1 hr at 50° C. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 2 x 50 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 80 mg (63%) of 297-4 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(allyloxy)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (297-5) Into a 100-mL round-bottom flask was placed 297-4 (190 mg), TFA (1 mL), and CH2Cl2(10 mL). The resulting solution was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, mobile phase, water (0.05% TFA) and CH3CN (70% Phase B up to 84% in 8 min); detector, UV. This resulted in 33.6 mg (22%) of 297-5 as a white solid. MS (ES, m/z): [M+H]+=653;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 4H), 1.08-1.28 (m, 15H), 1.28-1.51 (m, 7H), 1.51-1.80 (m, 5H), 1.85-2.02 (m, 4H), 2.05 (s, 1H), 2.09-2.38 (m, 4H), 2.54 (s, 1H), 2.81 (dd, J=13.6, 3.6 Hz, 1H), 3.72-3.88 (m, 2H), 4.05 (ddt, J=13.2, 5.4, 1.6 Hz, 1H), 4.88 (d, J=14.0 Hz, 1H), 5.02-5.26 (m, 3H), 5.61 (s, 1H), 5.70-5.90 (m, 1H). Example 53 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(3-methoxy-3-oxopropanamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (298-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(3-methoxy-3-oxopropanamido)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (298-1) Into a 100-mL round-bottom flask was placed 281-1 (200 mg, 0.26 mmol), DMF (5 mL), 3-methoxy-3-oxopropanoic acid (150 mg, 1.27 mmol, 5 equiv), iPr2EtN (0.17 mL, 1.03 mmol, 4 equiv), and HATU (390 mg, 1.03 mmol, 4 equiv). The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 4×50 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 230 mg (quant) of crude 298-1 as a yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(3-methoxy-3-oxopropanamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (298-2) Into a 100-mL round-bottom flask was placed 298-1 (230 mg, 0.26 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature and concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (52% Phase B up to 63% in 8 min); detector, UV. This resulted in 40.3 mg (20.53%) of 298-2 as a white solid. MS (ES, m/z): [M+H]+=712.25;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.91 (d, J=12.8 Hz, 1H), 1.06 (d, J=13.9 Hz, 1H), 1.14-1.22 (m, 12H), 1.22-1.30 (m, 2H), 1.35-1.46 (m, 7H), 1.48-1.58 (m, 1H), 1.59-2.03 (m, 8H), 2.09-2.30 (m, 5H), 2.59 (s, 1H), 2.81 (d, J=13.6 Hz, 1H), 3.25 (d, J=1.4 Hz, 2H), 3.72 (s, 3H), 4.29 (dd, J=12.5, 4.4 Hz, 1H), 4.77 (d, J=13.9 Hz, 1H), 5.02 (d, J=14.0 Hz, 1H), 5.62 (s, 1H). Example 54 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-methoxy-4-oxobutanamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (299-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-methoxy-4-oxobutanamido)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (299-1) Into a 100-mL round-bottom flask was placed 281-1 (200 mg, 0.26 mmol), DMF (5 mL), 4-methoxy-4-oxobutanoic acid (170 mg, 1.3 mmol, 5 equiv), iPr2EtN (0.17 mL, 1.03 mmol, 4 equiv), and HATU (390 mg, 1.03 mmol, 4 equiv). The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was diluted with 100 mL of CH2Cl2, washed with 3×50 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 238 mg (quant) of crude 299-1 as a yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-methoxy-4-oxobutanamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (299-2) Into a 100-mL round-bottom flask was placed 299-1 (238 mg, 0.27 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (55% Phase B up to 66% in 8 min); detector, UV. This resulted in 52.5 mg (26%) of 299-2 as a white solid. MS (ES, m/z): [M+H]+=726.25;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 4H), 1.06 (d, J=14.0 Hz, 1H), 1.12-1.32 (m, 14H), 1.33-1.56 (m, 8H), 1.59-1.99 (m, 8H), 2.11-2.27 (m, 5H), 2.34-2.49 (m, 2H), 2.48-2.66 (m, 3H), 2.76-2.84 (m, 1H), 3.68 (s, 3H), 4.25 (dd, J=12.6, 4.3 Hz, 1H), 4.76 (d, J=14.0 Hz, 1H), 5.01 (d, J=13.9 Hz, 1H), 5.61 (s, 1H). Example 55 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (300-1) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (300-1) Into a 100-mL round-bottom flask was placed 281-1 (200 mg, 0.26 mmol), CH2Cl2(5 mL), n-butyl chloroformate (180 mg, 1.32 mmol, 5 equiv), and Et3N (0.054 mL, 0.39 mmol, 1.5 equiv). The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 3×50 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 215 mg (90%) of 300-1 as a yellow semi-solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (300-2) Into a 100-mL round-bottom flask was placed 300-1 (215 mg, 0.24 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (73% Phase B up to 85% in 8 min); detector, UV. This resulted in 41.0 mg (22%) of 300-2 as a white solid. MS (ES, m/z): [M+H]+=712.25;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.86-1.02 (m, 4H), 1.03-1.09 (m, 1H), 1.10-1.18 (m, 9H), 1.19-1.31 (m, 5H), 1.33-1.50 (m, 9H), 1.52-1.82 (m, 8H), 1.83-1.94 (m, 2H), 1.94-2.00 (m, 1H), 2.05-2.27 (m, 5H), 2.58 (s, 1H), 2.75-2.84 (m, 1H), 3.91-4.05 (m, 3H), 4.86 (s, 1H), 5.01 (d, J=13.9 Hz, 1H), 5.61 (s, 1H). Example 56 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((methoxycarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (301-2) Synthesis of 2-benzhydryl 9-((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((methoxycarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (300-1) Into a 100-mL round-bottom flask was placed 209-1 (190 mg, 0.24 mmol), pyridine (5 mL), and methyl chloroformate (0.109 g, 1.2 mmol, 5 equiv). The reaction slurry was stirred for 3 days at 50° C. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 3×100 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 95 mg (48%) of 301-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((methoxycarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (301-2) Into a 100-mL round-bottom flask was placed 301-1 (180 mg, 0.22 mmol), TFA (1 mL), and CH2Cl2(10 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: mobile phase, water (0.05% TFA) and CH3CN (70% Phase B up to 90% in 8 min); This resulted in 49.8 mg (35%) of 301-2 as a white solid. MS (ES, m/z): [M+H]+=671;1H NMR (400 MHz, methanol-df) δ 0.85 (s, 3H), 0.92 (d, J=7.2 Hz, 1H), 1.06 (d, J=13.1 Hz, 1H), 1.13-1.30 (m, 21H), 1.41-1.57 (m, 7H), 1.58-1.73 (m, 4H), 1.78-1.88 (m, 3H), 2.00 (dd, J=21.8, 13.0 Hz, 3H), 2.22 (d, J=12.4 Hz, 1H), 2.46 (s, 1H), 2.91 (d, J=14.0 Hz, 1H), 3.02 (p, J=7.0 Hz, 1H), 3.75 (s, 3H), 4.85 (d, J=13.6 Hz, 1H), 4.95-5.10 (m, 2H), 5.75 (s, 1H). Example 57 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (302-3) Synthesis of (5-methyl-2-oxo-1,3-dioxol-4-yl)methyl (4-nitrophenyl) carbonate (302-1) Into a 100-mL round-bottom flask was placed 4-(hydroxymethyl)-5-methyl-2H-1,3-dioxol-2-one (3 g, 23 mmol), 4-nitrophenyl chloroformate (5 g, 24.8 mmol, 1.08 equiv), chloroform (100 mL), and pyridine (2 g, 25.3 mmol, 1.1 equiv). The reaction slurry was stirred for 12 hr at room temperature. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 2×100 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 1.8 g (26%) of 302-1 as a yellow solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (302-2) Into a 100-mL round-bottom flask was placed 194-8 (150 mg, 0.19 mmol), 302-1 (113 mg, 0.38 mmol, 2 equiv), pyridine (5 mL), and DMAP (164 mg, 0.38 mmol, 2 equiv). The reaction slurry was stirred for 12 hr at room temperature. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 3×100 of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 90 mg (50%) of 302-2 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (302-3) Into a 100-mL round-bottom flask was placed 302-2 (180 mg, 0.19 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: mobile phase, water (0.05% TFA) and CH3CN (73% Phase B up to 77% in 5 min); detector, UV. This resulted in 31.8 mg (21%) of 302-3 as a white solid. MS (ES, m/z): [M+H]+=770;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 1.01-1.33 (m, 16H), 1.41 (d, J=5.6 Hz, 7H), 1.56-1.73 (m, 4H), 1.75-1.89 (m, 3H), 2.03 (d, J=10.4 Hz, 3H), 2.21 (d, J=4.8 Hz, 7H), 2.45 (s, 1H), 2.93 (d, J=13.9 HZ, 1H), 4.78-5.11 (m, 5H), 5.75 (s, 3H). Example 58 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic Acid (307-2) Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-3-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (307-1) Into a 100-mL round-bottom flask was placed 302-1 (66 mg, 0.22 mmol), 194-7 (60 mg, 0.09 mmol, 0.4 equiv), pyridine (5 mL), and DMAP (76 mg, 0.44 mmol, 2 equiv). The reaction slurry was stirred for 12 min at 60° C. The reaction mixture was diluted with 50 mL of CH2Cl2, washed with 2×100 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1) to provide 30 mg (17%) of 307-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic Acid (307-2) Into a 100-mL round-bottom flask was placed 307-1 (170 mg, 0.21 mmol), TFA (1 mL), and CH2Cl2(10 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: mobile phase, water (0.05% TFA) and CH3CN (50% Phase B up to 55% in 8 min); detector, UV. This resulted in 6.7 mg (4.9%) of 307-2 as a white solid. MS (ES, m/z): [M+H]+=657.06;1H NMR (400 MHz, methanol-d4) δ 0.86 (s, 3H), 1.07-1.29 (m, 17H), 1.45 (d, J=11.6 Hz, 8H), 1.59-2.02 (m, 10H), 2.01-2.29 (m, 6H), 2.57 (s, 1H), 2.87 (d, J=13.6 Hz, 1H), 4.92 (m, 2H), 5.07 (dd, J=11.2, 5.6 Hz, 1H), 5.63 (s, 1H). Example 59 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-methoxyacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (308-2) Synthesis of 2-Benzhydryl 9-((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-methoxyacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (308-1) 2-Methoxyacetic acid (667 mg, 7.4 mmol, 20 equiv), DMAP (908.3 mg, 7.4 mmol, 20 equiv), and EDCI (1.07 mg, 5.6 mmol, 15 equiv) in CH2Cl2were stirred for 1 h at rt. 209-1 (300 mg, 0.37 mmol, 1 equiv) in CH2Cl2was added and the reaction stirred for additional 1 h. The mixture was diluted with CH2Cl2, washed with 1 M HCl(aq), and concentrated under vacuum. The residue was purified by prep-TLC (7:1 CH2Cl2:MeOH) to afford 308-1 (200 mg, 61%) as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-Isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(2-methoxyacetoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (308-2) 308-1 (300 mg, 0.34 mmol) and TFA (0.3 mL, 4.04 mmol, 12 equiv) in CH2Cl2(3 mL) were stirred for 1 h at rt. The reaction was concentrated and the residue purified by prep-HPLC under the following conditions—Column: Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (70% Phase B up to 90% in 8 min); detector: UV. This resulted in 308-2 (104.9 mg, 43%) as an off-white solid. MS (ES, m/z): [M+H]+=713.05;1H NMR (400 MHz, methanol-d4) δ 5.62 (s, 1H), 5.28 (dd, J=11.6, 5.1 Hz, 1H), 5.10 (d, J=13.9 Hz, 1H), 4.87 (s, 1H), 3.97 (d, J=3.1 Hz, 2H), 3.39 (s, 3H), 3.09 (p, J=7.0 Hz, 1H), 2.85 (d, J=13.8 Hz, 1H), 2.59 (s, 1H), 2.27-2.10 (m, 2H), 1.97 (d, J=10.2 Hz, 1H), 1.85 (t, 7=14.9 Hz, 3H), 1.80-1.66 (m, 5H), 1.47 (s, 3H), 1.41 (d, J=14.2 Hz, 4H), 1.32-1.22 (m, 6H), 1.20 (d, J=2.3 Hz, 6H), 1.16 (s, 3H), 1.07 (d, J=14.0 Hz, 1H), 0.96 (s, 1H), 0.85 (s, 3H). Example 60 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-Difluoroacetamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (309-2) Synthesis of 2-Benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-difluoroacetamido)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (309-1) EDCI (246.4 mg, 10 equiv) was added to 280-7 (100 mg, 1 equiv), 2,2-difluoroacetic acid (0.0323 mL, 4 equiv) and DMAP (15.6 mg, 1 equiv) in CH2Cl2(10 mL) and the reaction slurry stirred overnight at rt. The reaction mixture was concentrated under vacuum and the residue purified by silica gel column eluting with EtOAc/petroleum ether (1:1). This resulted in 113 mg (100%) of 309-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,2-Difluoroacetamido)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (309-2) 309-1 (186 mg, 1 equiv) and TFA (1 mL) in CH2Cl2(10 mL) were stirred for 1 h at rt. The reaction was concentrated and the residue purified by prep-HPLC with the following conditions—column, Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase: water (0.05% TFA) and CH3CN (58% Phase B up to 62% in 8 min); detector: UV. This resulted in 63.8 mg (43%) of 309-2 as a white solid. MS (ES, m/z): [M+H]+=690.15;1H NMR (400 MHz, methanol-d4) δ 0.83 (s, 3H), 0.90 (d, J=14.0 Hz, 1H), 1.05 (d, J=13.6 Hz, 1H), 1.13-1.20 (m, 12H), 1.25 (d, J=13.6 Hz, 2H), 1.38-1.57 (m, 8H), 1.59-2.00 (m, 8H), 2.10-2.27 (m, 5H), 2.58 (s, 1H), 2.82 (d, J=13.6 Hz, 1H), 4.29 (dd, J=12.4, 4.4 Hz, 1H), 4.77 (d, J=14.0 Hz, 1H), 4.99 (d, J=13.6 Hz, 1H), 5.60 (s, 1H), 5.93 (t, J=27.0 Hz, 1H). Example 61 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (314-4) Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-((cyclopropanecarbonyl)oxy)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (314-1) Into a 25-mL round-bottom flask was placed 194-7 (600 mg, 0.9 mmol), CH2Cl2(6 mL), DMAP (220 mg, 1.8 mmol, 2 equiv), EDCI (432 mg, 2.25 mmol, 2.50 equiv), and cyclopropanecarboxylic acid (232 mg, 2.7 mmol, 3 equiv). The reaction slurry was stirred for 2 hr at room temperature before quenching by the addition of HCl. The reaction mixture was extracted with 3×50 mL of ethyl acetate. The organic layers were combined, washed with 3 x50 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with CH2Cl2:methanol (10:1) to provide 170 mg (26%) of 314-1 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-9-isocyanato-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (314-2) Into a 8-mL round-bottom flask was placed 314-1 (170 mg, 0.23 mmol), anisole (1.9 mL), Et3N (0.05 mL, 0.35 mmol, 1.5 equiv), and DPPA (0.075 mL, 0.35 mmol, 1.5 equiv). The reaction slurry was stirred for 1.5 hr at 90° C. in an oil bath. The reaction mixture was concentrated and applied onto a silica gel column with ethyl acetate:petroleum ether (5:1) to provide 140 mg (83%) of 314-2 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (314-3) Into a 8-mL round-bottom flask was placed 314-2 (120 mg, 0.16 mmol), CH2Cl2(3 mL, 0.04 mmol, 0.22 equiv), TMSCl (0.071 mL), and 4-(hydroxymethyl)-5-methyl-1,3-dioxolan-2-one (63.6 mg, 0.48 mmol, 3 equiv). The reaction slurry was stirred overnight at room temperature then concentrated to provide 100 mg (71%) of crude 314-3 as a yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (314-4) Into a 8-mL round-bottom flask was placed 314-3 (30 mg, 0.04 mmol), CH2Cl2(1 mL), and TFA (0.1 mL). The resulting solution was stirred for 2 hr at room temperature. The crude product was purified by prep-HPLC with the following conditions: column, SunFire C18 OBD column, 5 μm, 19 mm*250 mm; flow rate: 20 mL/min; mobile phase, water (0.05% TFA) and CH3CN (60% Phase B to 69% 16 min); detector, 254 nm. This resulted in 6.5 mg (22%) of 314-4 as a white solid. MS (ES, m/z): [M+1]+=696;1H NMR (400 MHz, chloroform-tf) δ 0.87 (d, J=8.8 Hz, 5H), 0.96 (dt, J=8.1, 4.3 Hz, 2H), 1.06 (d, J=13.8 Hz, 1H), 1.12-1.22 (m, 10H), 1.25 (d, J=6.3 Hz, 4H), 1.31-1.53 (m, 8H), 1.59 (tt, J=8.1, 3.6 Hz, 2H), 1.64-1.79 (m, 4H), 1.80-2.12 (m, 4H), 2.20 (s, 5H), 2.50 (s, 1H), 2.82 (d, J=13.9 Hz, 1H), 4.43 (s, 1H), 4.70 (d, J=13.9 Hz, 1H), 4.84 (d, J=13.9 Hz, 1H), 5.46 (dd, J=11.2, 5.5 Hz, 1H), 5.74 (s, 1H). Example 62 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (315-2) Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (315-1) Into a 100-mL round-bottom flask was placed 314-2 (0.76 g, 1 mmol), CH2Cl2(12 mL), TMSCl (0.68 mg, 0.006 mmol, 6 equiv), and 4-ethyl-5-(hydroxymethyl)-2H-1,3-dioxol-2-one (0.60 mg, 0.004 mmol, 4 equiv). The reaction slurry was stirred for 2 hr at room temperature then concentrated to provide 0.6 g of crude 315-1 as a tan oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (315-2) Into a 250-mL round-bottom flask was placed 315-1 (0.6 g, 0.685 mmol), CH2Cl2(15 mL), and TFA (2 mL). The reaction slurry was stirred for 30 min at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH Prep C18 OBD, 5 μm, 19*150 mm; mobile phase, water (0.05% TFA) and CH3CN (65% Phase B up to 70% in 10 min); detector, UV. This resulted in 306 mg of 315-2 as a white solid. MS (ES, m/z): [M+1]+=710;1H NMR (300 MHz, methanol-d4) δ 0.86 (d, J=9.7 Hz, 7H), 1.04-1.31 (m, 17H), 1.43 (d, J=9.1 Hz, 8H), 1.57 (s, 3H), 1.65-1.81 (m, 4H), 1.81-1.98 (m, 3H), 2.20 (s, 2H), 2.30 (d, J=10.3 Hz, 1H), 2.59 (q, J=7.3 Hz, 3H), 2.75 (d, J=13.3 Hz, 1H), 4.72 (d, J=14.2 Hz, 2H), 5.59 (d, J=14.6 Hz, 2H). Example 63 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (316-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (316-1) The title compound was prepared with 4-(bromomethyl)-5-ethyl-1,3-dioxol-2-one (prepared according to literature procedures from Sun et al, Tetrahedron Letters, 2002, 43, 1161-1164) according to the methods to synthesize 209-3. The crude product was purified by Prep-HPLC with the following conditions: column, XBridge Prep OBD C18 Column, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (65% phase B up to 70% in 8 min); detector, UV. This resulted in 99.9 mg (62%) of 316-1 as a white solid. MS (ES, m/z) [M+1]+=669;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.96 (d, J=5.9 Hz, 1H), 1.07 (d, J=13.6 Hz, 1H), 1.16 (s, 3H), 1.18-1.25 (m, 12H), 1.27 (d, J=3.8 Hz, 1H), 1.33-1.49 (m, 6H), 1.66-1.83 (m, 6H), 1.86 (s, 1H), 1.96 (s, 3H), 1.98 (s, 1H), 2.12-2.28 (m, 2H), 2.56-2.66 (m, 3H), 2.79-2.88 (m, 1H), 4.89 (d, J=14.0 Hz, 1H), 5.05 (d, J=13.9 Hz, 1H), 5.16 (dd, J=11.7, 5.0 Hz, 1H), 5.62 (s, 1H). Example 64 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic (317-6) Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (317-1) (Trimethylsilyl)diazomethane (1 mL) was added to 194-7 (200 mg, 0.30 mmol, 1 equiv) in CH2Cl2(5 mL) and MeOH (2.5 mL), The resulting solution was stirred for 1 h at rt. The reaction mixture was concentrated to provide 200 mg (98%) of 317-1 as a white solid, used directly in the next step. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (317-2) Into a 100-mL round-bottom flask was placed 317-1 (500 mg, 1 equiv), CH2Cl2(4 mL), propanoic acid (0.164 mL, 3 equiv), DMAP (180 mg, 2 equiv), and EDCI (211 mg, 1.5 equiv). The resulting solution was stirred for 2 hr at room temperature. The residue was applied onto a silica gel column with 1:5 ethyl acetate:petroleum ether to provide 480 mg of 317-2 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-3-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (317-3) Into a 100-mL round-bottom flask was placed 317-2 (540 mg, 1 equiv), pyridine (7 mL), and LiI (98 mg, 10 equiv). The reaction mixture was stirred for 2 days at 120° C. The reaction slurry was concentrated, washed with 3×100 ml of 0.5 M HCl and 3×100 mL of brine. The organic layers were combined, dried over anhydrous Na2SO4, filtered, and concentrated to provide 480 mg of crude 317-3 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-isocyanato-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (317-4) Into a 100-mL round-bottom flask was placed 317-3 (340 mg, 1 equiv), anisole (4 mL), Et3N (0.1 mL, 1.5 equiv), and DPPA (0.152 mL, 1.5 equiv). The reaction mixture was stirred at 90° C. for 1.5 hr. The reaction slurry was concentrated and the residue applied onto a silica gel column with 1:7 ethyl acetate:petroleum ether to provide 300 mg of 317-4 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (317-5) Into a 25-mL round-bottom flask was placed 317-4 (130 mg, 1 equiv), CH2Cl2(2 mL), chlorotrimethylsilane (0.094 mL, 6 equiv), and 4-(hydroxymethyl)-5-(propan-2-yl)-2H-1,3-dioxol-2-one (115 mg, 4 equiv). The reaction mixture was stirred overnight at room temperature. The reaction slurry was concentrated to provide 160 mg of crude 317-5 as a yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (317-6) Into a 100-mL round-bottom flask was placed 317-5 (120 mg, 1 equiv), CH2Cl2(5 mL), and TFA (0.5 mL). The reaction mixture was stirred for 1 hr at room temperature. The reaction slurry was concentrated and purified by Prep-HPLC with the following conditions: column, Xselect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (74% Phase B up to 78% in 8 min); detector, UV. This resulted in 66.1 mg of 317-6 as a white solid. MS (ES, m/z): [M+1]+=712;1H NMR (300 MHz, methanol-d4) δ 5.60 (d, J=11.9 Hz, 2H), 4.80 (t, J=13.5 Hz, 1H), 3.08 (p, J=7.0 Hz, 1H), 2.76 (d, J=13.5 Hz, 1H), 2.59 (s, 1H), 2.38-2.27 (m, 2H), 2.31-2.19 (m, 1H), 1.99-1.82 (m, 3H), 1.81-1.67 (m, 4H), 1.58 (s, 1H), 1.44 (d, J=9.6 Hz, 7H), 1.31-1.17 (m, 9H), 1.21-1.06 (m, 12H), 1.05 (s, 1H), 0.85 (s, 3H). Example 65 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (318-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (318-1) The title compound was prepared with 317-4 and 4-ethyl-5-(hydroxymethyl)-1,3-dioxol-2-one (prepared according to literature procedures from Sun et al, Tetrahedron Letters, 2002, 43, 1161-1164) according to the methods to synthesize 317-6. The crude product was purified by Prep-HPLC with the following conditions: XSelect CSH Prep C18 OBD column, 5 μm, 19*150 mm; mobile phase, water (0.05% TFA) and CH3CN (60% phase B up to 79% in 8 min); detector, UV to provide 37.2 mg of 318-1 as a white solid. The product was tested in the assay in described in example 112 demonstrating a pIC50of 7.45 compared to the corresponding acid metabolite (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-amino-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propanoyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid having a pIC50of 6.4 and the subsequent amine metabolite (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-amino-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid having a pIC50less than 5. MS (ES, m/z): [M+1]+=698;1H NMR (400 MHz, methanol-d4) δ 5.62 (s, 2H), 4.86 (slH), 4.74 (d, J=14.1 Hz, 1H), 2.76 (d, J=13.1 Hz, 2H), 2.61 (q, J=7.4 Hz, 2H), 2.31 (q, J=7.9 Hz, 3H), 2.22 (s, 2H), 1.93 (d, J=16.9 Hz, 3H), 1.77 (d, J=13.3 Hz, 2H), 1.70 (s, 2H), 1.57 (s, 2H), 1.44 (d, J=10.7 Hz, 6H), 1.29-1.05 (m, 22H), 0.86 (s, 5H). Example 66 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (319-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (319-1) The title compound was prepared with 317-4 and 4-methyl-5-(hydroxymethyl)-1,3-dioxol-2-one (prepared according to literature procedures from Sun et al, Tetrahedron Letters, 2002, 43, 1161-1164) according to the methods to synthesize 317-6. The crude product was purified by Prep-HPLC with the following conditions: XSelect CSH Prep C18 OBD column, 5 μm, 30*150 mm; mobile phase, water (0.05% TFA) and CH3CN (68% phase B up to 71% in 8 min); detector, UV to provide 56.4 mg of 319-1 as a white solid. MS (ES, m/z): [M+1]+=684;1H NMR (400 MHz, methanol-d4) δ 5.61 (d, J=11.2 Hz, 2H), 4.89 (s, 1H), 4.70 (d, J=14.1 Hz, 1H), 2.76 (d, J=13.6 Hz, 1H), 2.60 (s, 1H), 2.36-2.23 (m, 3H), 2.20 (s, 3H), 1.96 (s, 1H), 1.88 (d, J=13.4 Hz, 1H), 1.74 (dd, J=19.2, 12.8 Hz, 4H), 1.59 (d, J=13.4 Hz, 1H), 1.45 (d, J=14.4 Hz, 7H), 1.31 (d, J=31.1 Hz, 1H), 1.27-1.03 (m, 17H), 0.86 (s, 3H). Example 67 (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-4-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-4,6a,6b,8a,11,14b-hexamethyl-11-(methylcarbamoyl)-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicen-3-yl propionate (320-1) Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-4-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-4,6a,6b,8a,11,14b-hexamethyl-11-(methylcarbamoyl)-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicen-3-yl propionate (320-1) Into a 25-mL round-bottom flask was placed 318-1 (60 mg, 0.086 mmol), DMF (2 mL), methylamine hydrochloride (5.8 mg, 0.086 mmol, 1 equiv), iPr2EtN (0.07 mL, 0.58 mmol, 5 equiv), and HATU (49.0 mg, 0.13 mmol, 1.5 equiv). The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was extracted with 3×50 mL of ethyl acetate. The combined organic layers were washed with 50 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XBridge Prep OBD Cl8, mobile phase, water (0.05% TFA) and CH3CN (55% phase B up to 70% in 8 min); detector, UV. This resulted in 38.9 mg of 320-1 as a white solid. MS (ES, m/z): [M+1]+=711;1H NMR (300 MHz, methanol-d4) δ 5.68 (s, 1H), 5.64-5.54 (m, 1H), 4.74 (d, J=14.1 Hz, 1H), 2.76 (s, 4H), 2.68-2.54 (m, 3H), 2.31 (q, J=7.7 Hz, 3H), 2.17 (d, J=13.8 Hz, 2H), 1.93 (t, J=12.4 Hz, 3H), 1.74 (dd, J=17.5, 11.0 Hz, 4H), 1.43 (d, J=16.4 Hz, 9H), 1.29-1.01 (m, 22H), 0.84 (s, 3H). Example 68 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (321-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((cyclopropanecarbonyl)oxy)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (321-1) The title compound was prepared with 314-2 and 4-(hydroxymethyl)-5-(propan-2-yl)-2H-1,3-dioxol-2-one (prepared according to literature procedures from Sun et al, Tetrahedron Letters, 2002, 43, 1161-1164) according to the methods to synthesize 315-2. The crude product was purified by prep-HPLC with the following conditions: XSelect CSH Prep C18 OBD column, 5 μm, 19*150 mm; mobile phase, water (0.05% TFA) and CH3CN (75% phase B up to 86% in 7 min); detector, 254 nm, to provide 42.3 mg of 321-1 as a white solid. MS (ES, m/z) [M+1]+=724;1H NMR (400 MHz, chloroform-d) δ 5.74 (s, 1H), 5.43 (dd, J=11.8, 5.3 Hz, 1H), 4.83 (d, J=13.9 Hz, 1H), 4.75 (d, J=14.1 Hz, 1H), 4.44 (s, 1H), 3.02 (p, J=7.0 Hz, 1H), 2.82 (d, J=13.4 Hz, 1H), 2.49 (s, 1H), 2.21 (d, J=12.6 Hz, 2H), 2.04-1.91 (m, 1H), 1.73 (d, J=15.5 Hz, 1H), 1.70-1.53 (m, 3H), 1.44 (s, 4H), 1.39 (s, 3H), 1.29-1.23 (m, 8H), 1.22 (s, 1H), 1.20-1.12 (m, 9H), 1.06 (d, J=14.2 Hz, 1H), 0.97 (s, 2H), 0.88 (d, J=3.5 Hz, 1H), 0.86 (s, 4H). Example 69 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (322-6) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(methoxycarbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (322-1) Into a 100-mL round-bottom flask was placed methanol (5 mg), 297-2 (315 mg, 0.44 mmol, 1 equiv), Pd/C (10%, 152 mg, 1.4 mmol, 3.3 equiv), and acetone (5 mL). The vessel was charged with H2(g) and the reaction slurry stirred for 4 hr at room temperature. The solids were filtered off and the filtrate was concentrated under vacuum to provide 287 mg of crude 322-1 as a white solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (322-2) Into a 100-mL round-bottom flask, was placed methanol (4 mL), 322-1 (287 mg, 0.52 mmol, 1 equiv), ether (2 mL), and Ph2CN2(300 mg, 1.5 mmol, 3 equiv). The reaction slurry was stirred for 2 hr at 45° C. The reaction mixture was concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (10:1) to provide 284 mg (76%) of 322-2 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-3-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (322-3) Into a 100-mL round-bottom flask was placed pyridine (5 mL), 322-2 (284 mg, 0.39 mmol, 1 equiv), and LiI (526 mg, 3.9 mmol, 10 equiv). The reaction slurry was stirred for 2 days at 125° C. The reaction mixture was concentrated under vacuum. The residue was diluted with 90 mL of ethyl acetate and washed with 30 mL of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 280 mg of 322-3 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-isocyanato-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (322-4) Into a 100-mL round-bottom flask was placed anisole (5 mL), 322-3 (287 mg, 0.4 mmol, 1 equiv), Et3N (102.4 mg, 1 mmol, 2.5 equiv), and DPPA (189.4 mg, 0.69 mmol, 1.7 equiv). The reaction slurry was stirred for 1 hr at 90° C. then concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 158 mg (55%) of 322-4 as a white solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (322-5) Into a 50-mL round-bottom flask was placed CH2Cl2(3 mL), 322-4 (135 mg, 0.19 mmol, 1 equiv), TMSCl (104 mg, 1 mmol, 5 equiv), and 4-(hydroxymethyl)-5-methyl-2H-1,3-dioxol-2-one (74.6 mg, 0.57 mmol, 3 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with brine and extracted with 3×30 mL of ethyl acetate. The combined organic layers were washed with 2×20 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 100 mg (64%) of 322-5 as a yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (322-6) Into a 100-mL round-bottom flask was placed 322-5 (80 mg, 1 equiv), CH2Cl2(10 mL), and TFA (1 mL). The resulting solution was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, Xselect CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (70% Phase B up to 75% in 8 min); detector, UV. This resulted in 13.9 mg of 322-6 as a white solid. MS (ES, m/z): [M+1]+=670.4;1H NMR (400 MHz, methanol-d4) δ 5.61 (s, 1H), 4.91 (d, J=14.2 Hz, 1H), 4.77 (d, J=14.1 Hz, 1H), 4.02 (d, J=10.0 Hz, 1H), 3.52 (dt, J=9.5, 6.4 Hz, 1H), 2.72 (d, J=13.2 Hz, 1H), 2.55 (s, 1H), 2.24 (s, 1H), 2.19 (s, 3H), 2.21-2.10 (m, 2H), 1.97 (d, J=9.0 Hz, 1H), 1.87 (d, J=12.9 Hz, 1H), 1.83-1.64 (m, 2H), 1.56 (dd, J=18.8, 10.8 Hz, 2H), 1.49 (t, J=7.0 Hz, 2H), 1.43 (d, J=10.4 Hz, 7H), 1.36-1.19 (m, 2H), 1.19 (s, 3H), 1.14 (d, J=2.0 Hz, 6H), 1.04 (s, 6H), 0.93-0.83 (m, 6H). Example 70 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (323-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-propoxy-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (323-1) The title compound was prepared with 322-4 and 4-(hydroxymethyl)-5-(propan-2-yl)-2H-1,3-dioxol-2-one (prepared according to literature procedures from Sun et al, Tetrahedron Letters, 2002, 43, 1161-1164) according to the methods to synthesize 322-6. The crude product was purified by prep-HPLC with the following conditions: XSelect CSH Prep C18 OBD column, 5 μm, 30*150 mm; mobile phase, water (0.05% TFA) and CH3CN (78% phase B up to 84% in 8 min); detector, 254 nm, to provide 49.9 mg (41%) of 321-1 as a white solid. MS (ES, m/z) [M+1]+=698;1H NMR (400 MHz, methanol-d4) δ 0.88 (dd, J=13.5, 6.0 Hz, 6H), 1.02 (d, J=15.2 Hz, 3H), 1.11-1.31 (m, 17H), 1.48 (dt, J=20.1, 6.7 Hz, 8H), 1.58 (s, 3H), 1.76 (d, J=13.2 Hz, 1H), 1.82-1.99 (m, 1H), 2.13 (s, 1H), 2.55 (s, 1H), 2.73 (d, J=13.8 Hz, 1H), 3.03-3.14 (m, 1H), 3.35 (d, J=2.8 Hz, 3H), 3.52 (dt, J=12.4, 6.2 Hz, 1H), 4.02 (d, J=10.9 Hz, 1H), 4.92 (d, J=14.9 Hz, 2H), 5.62 (d, J=5.1 Hz, 1H). Example 71 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (324-3) Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-isocyanato-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (324-1) Into a 8-mL round-bottom flask was placed 249-3 (160 mg, 0.24 mmol), anisole (1.6 mL), Et3N (50 uL, 0.35 mmol, 1.5 equiv), and DPPA (76 uL, 0.35 mmol, 1.5 equiv). The reaction slurry was stirred for 1 hr at 90° C. in an oil bath. The reaction mixture was concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 140 mg (88%) of 324-1 as a white solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (324-2) Into an 8-mL round-bottom flask was placed 324-1 (140 mg, 0.21 mmol), CH2Cl2(2 mL), TMSCl (89 uL, 5 equiv), amd 4-(hydroxym ethyl)-5-methyl-2H-1,3-dioxol-2-one (81 mg, 0.62 mmol, 3 equiv). The reaction slurry was stirred overnight at room temperature. The resulting mixture was concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:3) to provide 150 mg (90%) of 324-2 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-9-((((5-methyl-2-oxo-1,3-di oxol-4-yl)methoxy)carbonyl)amino)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (324-3) Into a 50-mL round-bottom flask was placed 324-2 (150 mg, 0.19 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 30 minutes at room temperature. The reaction mixture was concentrated. The crude product was purified by prep-HPLC with the following conditions: column, Xselect CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (61% Phase B up to 66% in 8 min); detector, UV. This resulted in 47.8 mg (40%) of 324-3 as a white solid. MS (ES, m/z): [M+1]+=642;1H NMR (300 MHz, methanol-d4) δ 0.85 (s, 3H), 1.03 (s, 3H), 1.09 (s, 1H), 1.12-1.23 (m, 9H), 1.23-1.37 (m, 1H), 1.44 (d, J=7.4 Hz, 13H), 1.53-1.81 (m, 2H), 1.85 (s, 2H), 1.89 (s, OH), 1.96 (s, 1H), 2.12 (d, J=11.1 Hz, 1H), 2.20 (s, 3H), 2.24 (s, 1H), 2.55 (s, 1H), 2.74 (d, J=13.5 Hz, 1H), 3.36 (s, 3H), 3.97 (d, J=9.1 Hz, 1H), 5.61 (s, 1H), 7.34 (dt, J=14.9, 7.5 Hz, 1H). Example 72 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (325-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-10-methoxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (325-1) The title compound was prepared with 324-1 and 4-(hydroxymethyl)-5-(propan-2-yl)-2H-1,3-dioxol-2-one (prepared according to literature procedures from Sun et al, Tetrahedron Letters, 2002, 43, 1161-1164) according to the methods to synthesize 324-3. The crude product was purified by prep-HPLC with the following conditions: column, Xselect CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (70% Phase B up to 75% in 8 min); detector, UV. This resulted in 66.5 mg (52%) of 325-1 as a white solid. MS (ES, m/z): [M+1]+=670;1H NMR (300 MHz, methanol-d4) δ 0.85 (s, 3H), 1.03 (s, 4H), 1.09 (s, 1H), 1.12-1.22 (m, 9H), 1.22-1.37 (m, 8H), 1.43 (d, J=5.2 Hz, 8H), 1.57 (s, 3H), 1.69 (s, 1H), 1.76 (d, J=13.3 Hz, 1H), 1.87 (d, J=13.1 Hz, 3H), 1.97 (d, J=9.7 Hz, 1H), 2.14 (dd, J=18.4, 7.5 Hz, 2H), 2.23 (d, J=10.7 Hz, 1H), 2.55 (s, 1H), 2.74 (d, J=13.7 Hz, 1H), 3.08 (p, J=6.9 Hz, 1H), 3.36 (s, 3H), 3.96 (d, J=12.0 Hz, 1H), 4.89 (s, 1H), 5.61 (s, 1H). Example 73 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (326-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-acetoxy-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (326-1) The title compound was prepared with 254-1 and 4-(hydroxymethyl)-5-ethyl-2H-1,3-dioxol-2-one (prepared according to literature procedures from Sun et al, Tetrahedron Letters, 2002, 43, 1161-1164) according to the methods to synthesize 255-2. The mixture was concentrated under vacuum and the residue purified by prep-HPLC with the following conditions—Column: XSelect CSH Prep C18 OBD, 5 μm, 19*150 mm; mobile phase: water (0.05% TFA) and CH3CN (63% Phase B up to 65% in 8 min); detector: UV. This resulted in 39.3 mg of 326-1 as a white solid. MS (ES, m/z): [M+H]+=684;1H NMR (300 MHz, methanol-d4) δ 0.83-0.89 (s, 3H), 1.08-1.34 (m, 18H), 1.40-1.60 (d, J=10.0 Hz, 7H), 1.67-1.82 (dd, J=11.3, 15.7 Hz, 2H), 1.83-1.98 (m, 4H), 1.98-2.04 (s, 6H), 2.18-2.24 (s, 2H), 2.29-2.39 (d, J=10.8 Hz, 1H), 2.54-2.67 (q, 7=7.5 Hz, 3H), 2.71-2.81 (d, J=12.9 Hz, 1H), 4.70-4.86 (t, 7=14.8 Hz, 2H), 5.51-5.65 (m, 2H). Example 74 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (327-8) Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((tert-butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (327-1) Into a 50-mL round-bottom flask was placed 280-5 (600 mg, 0.78 mmol), DMF (6 mL), 3-bromoprop-1-ene (0.27 mL, 4 equiv), KI (66 mg, 0.4 mmol, 0.5 equiv), and K2CO3(540 mg, 4 mmol, 5 equiv). The reaction slurry was stirred for 1 hr at room temperature. The reaction mixture was extracted with 300 mL of CH2Cl2, washed with 3×300 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 616 mg (98%) of 327-1 as a white solid. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (327-2) Into a 100-mL round-bottom flask was placed 327-1 (616 mg, 0.76 mmol), CH2Cl2(50 mL), and 2,6-lutidine (0.44 mL, 5 equiv). To this slurry was added TMSOTf (679 mg, 3.1 mmol, 4 equiv) at 0° C. The reaction slurry was stirred for 1 hr at room temperature. The reaction mixture was washed with 3×300 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 600 mg (quant) of 327-2 as a yellow solid. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(4-oxopiperidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (327-3) Into a 100-mL round-bottom flask was placed 327-2 (3 g), 1-ethyl-1-methyl-4-oxopiperidin-1-ium iodide (2.18 g, 2 equiv), EtOH (3 mL), H2O (6 mL), and NaHCO3(850 mg, 2.5 equiv). The reaction slurry was stirred for 1 hr at 80° C. then concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 2.5 g of 327-3 as a light yellow solid. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (327-4) Into a 25-mL round-bottom plastic flask was placed 327-3 (300 mg), BAST (170 mg, 2 equiv), EtOH (5 mg, 0.3 equiv), and CH2Cl2(3 mL). The reaction slurry was stirred for 12 hr at room temperature and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:2) to provide 210 mg (68%) of 327-4 as a yellow solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-(4,4-difluoropiperidin-1-yl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (327-5) Into a 100-mL round-bottom flask was placed 327-4 (810 mg), Pd(PPh3)4(700 mg, 0.6 equiv), pTolSO2Na (360 mg, 1.4 equiv), THF (10 mL), and MeOH (30 mL). The reaction slurry was stirred for 1 hr at room temperature at N2atmosphere then concentrated. The residue was applied onto a silica gel column with CH2Cl2:methanol (10:1) to provide 660 mg (86%) of 327-5 as a yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-9-isocyanato-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (327-6) Into a 100-mL round-bottom flask was placed 327-5 (200 mg), DPPA (107 mg, 1.5 equiv), Et3N (40 mg, 1.5 equiv), and anisole (2 mL). The reaction slurry was stirred for 1 hr at 90° C. The reaction mixture was concentrated and the residue applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 130 mg (65%) of 327-6 as a white solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (327-7) Into a 100-mL round-bottom flask was placed 327-6 (70 mg), CH2Cl2(3 mL), TMSCl (50 mg, 5 equiv), and 4-(hydroxymethyl)-5-isopropyl-1,3-dioxol-2-one (50 mg, 4.2 equiv). The reaction slurry was stirred for 12 hr at room temperature then concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 100 mg of 327-7 as colorless oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-9-((((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (327-8) Into a 25-mL round-bottom flask, was placed 327-7 (90 mg), TFA (1 ml), and CH2Cl2(5 ml). The resulting solution was stirred for 1 hr at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: Column: XSelect CSH Prep C18; mobile phase, water (0.05% TFA) and CH3CN (38% Phase B up to 45% in 8 min); detector: UV. This resulted in 4.6 mg (3%) of 327-8 as a colorless oil. MS (ES, m/z) [M+H]+=759.5;1H NMR (300 MHz, methanol-d4) δ 0.85 (s, 3H), 1.07 (d, J=14.8 Hz, 2H), 1.14-1.22 (m, 11H), 1.20-1.33 (m, 11H), 1.43 (s, 7H), 1.48 (d, J=12.8 Hz, 2H), 1.58 (s, 2H), 1.71 (q,7=12.8, 12.4 Hz, 3H), 1.87 (d, J=14.6 Hz, 3H), 2.12 (s, 2H), 2.36 (d, J=11.2 Hz, 3H), 2.99 (s, 1H),3.10 (s, 1H), 3.30 (s, 1H), 3.50 (d, J=12.0 Hz, 2H), 3.55-3.60 (m, 1H), 4.95 (d, J=14.2 Hz, 1H), 5.05 (d, J=14.2 Hz, 1H), 5.63 (s, 1H). Example 75 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(1H-pyrrol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (328-2) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid trifluoroacetate (328-1) Into a 100-mL round-bottom flask was placed 281-1 (300 mg, 0.37 mmol), CH2Cl2(10 mL), and TFA (1 mL). The resulting solution was stirred for 1 hr at room temperature. The resulting mixture was concentrated to provide 270 mg of 328-1 as a yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(1H-pyrrol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (328-2) Into a 25-mL round-bottom flask was placed NaOAc (20 mg, 0.3 mmol, 1 equiv), H2O (2 mL), 328-1 (200 mg, 0.31 mmol, 1 equiv), AcOH (0.5 mL), and 2,5-dimethoxyoxolane (0.032 mL, 0.31 mmol, 1 equiv). The reaction slurry was stirred for 2 hr at 75° C. The reaction mixture was extracted with 3×100 mL of ethyl acetate, washed with 2×150 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (50% Phase B up to 55% in 8 min); detector, UV. This resulted in 44.2 mg of 328-2 as a white solid. MS (ES, m/z): [M+1]+=662;1H NMR (300 MHz, methanol-d4) δ 6.61 (t, J=2.1 Hz, 2H), 5.98 (t, J=2.1 Hz, 2H), 5.64 (s, 1H), 5.15 (d, J=14.0 Hz, 1H), 4.84 (d, J=13.8 Hz, 1H), 4.52 (dd, J=13.0, 3.7 Hz, 1H), 2.97 (d, J=13.6 Hz, 1H), 2.63 (s, 1H), 2.39 (d, J=13.5 Hz, 1H), 2.25 (d, J=11.8 Hz, 1H), 2.20 (s, 3H), 1.97 (s, 1H), 1.89 (d, J=8.8 Hz, 1H), 1.83-1.70 (m, 2H), 1.52-1.24 (m, 12H), 1.19 (d, J=9.6 Hz, 6H), 1.06 (s, 4H), 0.93 (d, J=11.4 Hz, 1H), 0.86 (s, 3H). Example 76 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,12aS,12bR,14bR)-2,4a,6a, 6b,9,12a-hexamethyl-10,13-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (329-1) Into a 25-mL round-bottom flask was 249-1 (3.06 g, 4.5 mmol), CH2Cl2(30 mL), and DMP (3.8 g, 9 mmol, 2 equiv). The reaction slurry was stirred for 3 hr at room temperature, adjusting the pH value of the solution to 8 with saturated NaHCO3(aq). The reaction mixture was extracted with 2×20 mL of CH2Cl2. The organic layer was washed with 3×100 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 2.5 g (82%) of 329-1 as a light yellow solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-(tert-butoxycarbonyl)hydrazinyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (329-2) Into a 25-mL round-bottom flask was placed 329-1 (1.0 g, 1.5 mmol), (tert-butoxy)carbohydrazide (211 mg, 1.6 mmol, 1.1 equiv), CH2Cl2(2 ml), and AcOH (2 ml). The reaction slurry was stirred for 2 h at room temperature, then treated with NaBH3CN (111 mg, 1.76 mmol, 1.2 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with CH2Cl2(100 ml) and the reaction quenched by the addition of 100 mL of water. The pH of the solution was adjusted to 9 with saturated NaHCO3(aq) and extracted with 2×100 ml of CH2Cl2. The combined organic layers were washed with 2×200 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with 1:5 ethyl acetate:petroleum ether to provide 880 mg (78%) of 329-2 as a white solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-hydrazinyl-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate dihydrochloride (329-3) Into a 100-mL round-bottom flask was placed 329-2 (850 mg, 1.1 mmol), CH2Cl2(50 ml), and 2,6-lutidine (0.63 mL, 6.4 mmol, 5 equiv), followed by the addition of TMSOTf (0.77 mL, 4.3 mmol, 4 equiv) dropwise with stirring at rt. The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was diluted with CH2Cl2(450 ml) and washed with 200 ml of 0.07 M HCl in brine then 1×300 mL of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 650 mg (83%) of crude 329-3 as a light yellow solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (329-4 Into a 25-mL round-bottom flask was placed 329-3 (800 mg, 1.04 mmol) and t-BuOH (3 ml), 1,1,3,3-tetramethoxypropane (171 mg, 1.04 mmol, 1.0 equiv) followed by the addition of cone HCl (0.33 mL, v/v: 1/0.11) dropwise with stirring at rt. The reaction slurry was stirred for 1 h at 95° C. The reaction mixture was cooled and diluted with 100 mL of CH2Cl2, washed with 2×100 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The solid was dried in an oven under reduced pressure and applied onto a silica gel column with 1:5 ethyl acetate:petroleum ether to provide 631 mg (79%) of 329-4 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-3-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (329-5) Into a 25-mL sealed tube was placed 329-4 (400 mg, 0.55 mmol), pyridine (4 ml), and LiI (1.11 g, 1.62 mmol, 3 equiv). The reaction slurry was stirred overnight at 125° C. The reaction mixture was cooled and diluted with 100 mL of CH2Cl2and washed with 2×100 ml of 0.5 M HCl. The organic layer was washed with 2×100 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 350 mg (89%) of 329-5 as a yellow solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (329-6) Into a 8-mL vial was 329-5 (140 mg, 0.195 mmol), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (44 mg, 0.293 mmol, 1.5 equiv), DMF (3 ml), KI (16 mg, 0.098 mmol, 0.5 equiv), and K2CO3(80 mg, 0.59 mmol, 3 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with 30 mL of ethyl acetate and the reaction quenched by the addition of 20 ml of water. The layers were separated and the aqueous was extracted with 2×30 ml of ethyl acetate. The combined organic layers were washed with 3×80 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with 1:1 ethyl acetate:petroleum ether to provide 131 mg (81%) of 329-6 as a light yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (329-7) Into a 8-mL vial was placed 329-6 (131 mg, 0.16 mmol), CH2Cl2(3 ml), and TFA (0.3 ml). The reaction slurry was stirred for 30 min at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, X-Select CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (68% Phase B up to 70% in 8 min); detector, UV. This resulted in 53.3 mg (51%) of 329-7 as a white solid. MS (ES, m/z): [M+1]+=663;1H NMR (400 MHz, methanol-d4) δ 0.86 (s, 3H), 0.93 (s, 3H), 1.08 (d, J=14.0 Hz, 1H), 1.19 (d, J=13.4 Hz, 6H), 1.26 (s, 3H), 1.28-1.37 (m, 1H), 1.46 (d, J=24.2 Hz, 6H), 1.60-1.90 (m, 5H), 1.89 (s, 1H), 1.98 (d, J=9.9 Hz, 1H), 2.20 (s, 3H), 2.26 (s, 1H), 2.39-2.53 (m, 1H), 2.64 (s, 1H), 4.82 (dd, J=13.1, 3.9 Hz, 1H), 4.89 (d, J=14.0 Hz, 1H), 5.15 (d, J=13.9 Hz, 1H), 5.64 (s, 1H), 6.25 (t, J=2.1 Hz, 1H), 7.40 (d, J=1.9 Hz, 1H), 7.62 (d, J=2.4 Hz, 1H). Example 77 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic (330-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(1H-pyrazol-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (329-7) The title compound was prepared with 249-1 and 4-(bromomethyl)-5-isopropyl-1,3-dioxol-2-one according to the methods to synthesize 329-7. The crude product was purified by prep-HPLC with the following conditions: column, X-Select CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (73% Phase B up to 77% in 8 min); detector, UV. This resulted in 46.3 mg of 330-1 as a white solid. MS (ES, m/z): [M+1]+=692;1H NMR (400 MHz, methanol-d4) δ 0.86 (s, 3H), 0.93 (s, 3H), 1.08 (d, J=13.7 Hz, 1H), 1.17 (s, 3H), 1.19-1.33 (m, 12H), 1.31-1.49 (m, 5H), 1.49 (s, 3H), 1.70 (dd, J=29.2, 13.2 Hz, 2H), 1.77-1.88 (m, 1H), 1.85-1.92 (m, 1H), 1.98 (d, J=10.1 Hz, 1H), 2.21 (ddd, J=27.9, 13.4, 4.2 Hz, 2H), 2.39-2.54 (m, 1H), 2.64 (s, 1H), 2.96-3.04 (m, 1H), 3.09 (p, J=7.0 Hz, 1H), 4.83 (dd, J=13.1, 3.9 Hz, 1H), 4.93 (d, J=13.9 Hz, 1H), 5.16 (d, J=13.9 Hz, 1H), 5.64 (s, 1H), 6.25 (t, J=2.2 Hz, 1H), 7.40 (d, J=1.9 Hz, 1H), 7.62 (d, J=2.4 Hz, 1H). Example 78 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(5-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (331-2) Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(5-methyl-1H-pyrazol-1-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (331-1) Into a 25-mL round-bottom flask was placed 329-3 (800 mg, 1.15 mmol), MeOH (6.8 mL), and (3H)-4-(di methyl amino) but-3-en-2-one (156 mg, 1.4 mmol, 1.2 equiv). The reaction slurry was stirred overnight at 70° C. The reaction mixture was cooled, diluted with 100 mL of CH2Cl2, and washed with 2×100 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 640 mg of 331-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(5-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (331-2) The title compound was prepared with 331-1 according to the methods to synthesize 329-7. The crude product was purified by prep-HPLC with the following conditions: column, X-Select CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (hold 68% Phase B for 8 min); detector, UV. This resulted in 49.0 mg of 331-2 as a white solid. MS (ES, m/z) [M+1]+=677;1H NMR (400 MHz, DMSO-d6) δ 0.77 (s, 3H), 0.98 (d, J=13.8 Hz, 1H), 1.06 (s, 3H), 1.12 (s, 3H), 1.17 (d, J=12.5 Hz, 5H), 1.18 (s, 2H), 1.27 (d, J=13.5 Hz, 2H), 1.36 (s, 2H), 1.40 (s, 3H), 1.46 (d, J=14.1 Hz, 1H), 1.62 (d, J=9.1 Hz, 1H), 1.72 (d, J=6.5 Hz, 3H), 1.74-1.84 (m, 1H), 2.06-2.16 (m, 7H), 2.34-2.45 (m, 1H), 2.76 (d, J=13.6 Hz, 1H), 4.54 (d, J=11.8 Hz, 1H), 4.84 (d, J=14.0 Hz, 1H), 5.15 (d, J=14.0 Hz, 1H), 5.45 (s, 1H), 5.90 (s, 1H), 7.28 (d, J=1.7 Hz, 1H). Example 79 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(3-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (332-2) Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(3-methyl-1H-pyrazol-1-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (332-1) Into a 100-mL round-bottom flask was placed 329-3 (800 mg, 1.15 mmol), 4,4-dimethoxybutan-2-one (0.25 mL, 1.4 mmol, 1.2 equiv), and EtOH (25 mL). The reaction slurry was stirred for 2 h at reflux. Concentrated HCl (0.025 mL) was added dropwise and the slurry stirred for 2 h at reflux. The reaction mixture was cooled, diluted with 200 mL of CH2Cl2, and washed with 2×200 mL of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 430 mg of 332-1 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(3-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (331-2) The title compound was prepared with 332-1 according to the methods to synthesize 329-7. The crude product was purified by prep-HPLC with the following conditions: column, X-Select CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (57% Phase B up to 77% over 9 min); detector, UV. This resulted in 46.7 mg of 332-2 as a white solid. MS (ES, m/z): [M+1]+=677;1H NMR (300 MHz, DMSO-76) δ 0.77 (d, J=3.6 Hz, 7H), 0.87-1.02 (m, 1H), 1.05 (s, 3H), 1.09-1.15 (m, 6H), 1.35 (d, J=24.6 Hz, 7H), 1.54-1.66 (m, 2H), 1.71 (s, 1H), 1.81 (d, J=13.3 Hz, 1H), 2.12 (d, J=31.2 Hz, 6H), 2.33 (s, 1H), 2.81 (d, J=12.7 Hz, 1H), 4.57 (d, J=11.9 Hz, 1H), 4.96 (d, J=14.0 Hz, 1H), 5.07 (d, J=14.0 Hz, 1H), 5.45 (s, 1H), 5.93 (d, J=2.2 Hz, 1H), 7.52 (d, J=2.3 Hz, 1H). Example 80 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(propionyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (333-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-10-(3-methyl-1H-pyrazol-1-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (333-1) The title compound was prepared from 329-3 according to the methods to synthesize 332-2. The crude product was purified by Prep-HPLC with the following conditions: Column, X select CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (68% phase B up to 70% in 8 min); detector, UV to provide 41.4 mg (34%) of 333-1 as a white solid. MS (ES, m/z): [M+1]+=691;1H NMR (400 MHz, methanol-d4) δ 7.45 (d, J=2.3 Hz, 1H), 6.01 (d, J=2.3 Hz, 1H), 5.63 (s, 1H), 5.14 (d, J=13.9 Hz, 1H), 4.88 (d, J=14.0 Hz, 1H), 4.71 (dd, J=13.1, 3.9 Hz, 1H), 2.98 (d, J=13.4 Hz, 1H), 2.66-2.54 (m, 3H), 2.50-2.36 (m, 1H), 2.23 (d, J=12.7 Hz, 1H), 2.19 (s, 3H), 1.97 (d, J=9.9 Hz, 1H), 1.91-1.73 (m, 2H), 1.73-1.58 (m, 1H), 1.48 (s, 3H), 1.42 (s, 2H), 1.40-1.33 (m, 1H), 1.33-1.14 (m, 12H), 1.07 (d, J=14.1 Hz, 1H), 1.04-0.94 (m, 1H), 0.95 (s, 3H), 0.94-0.87 (m, 1H), 0.85 (s, 3H). Example 81 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (334-8) Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (334-1) Into a 250-mL round-bottom flask was placed 194-7 (3.0 g), DMF (20 mL), KI (0.37 g, 0.5 equiv), 3-bromoprop-1-ene (1.557 mL, 4 equiv), and K2CO3(3.1 g, 5 equiv). The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The combined organic layers were washed with 2×150 ml of H2O and 2×150 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (3:1) to provide 2.69 g (85%) of 334-1 as a light yellow solid. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10,13-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (334-2) Into a 250-mL round-bottom flask was placed 334-1 (2.70 g), CH2Cl2(40 mL), then DMP (4.86 g, 3 equiv) in several batches at 0° C. The reaction slurry was stirred for 3 h then quenched by the addition of saturated NaHCO3(aq)and extracted with CH2Cl2. The organic layer was washed with 2×150 ml of H2O and 2×150 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (15:85) to provide 2.61 g (97%) of 334-2 as a white solid. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-(tert-butoxycarbonyl)hydrazinyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (334-3) Into a 250-mL round-bottom flask was placed 334-2 (2.61 g), CH2Cl2(10 mL), AcOH (10 mL), and (tert-butoxy)carbohydrazide (538 mg, 1.1 equiv). The reaction slurry was stirred for 2 h at room temperature. The reaction slurry was cooled to 0° C. before portionwise addition of NaBH3CN (279 mg, 1.2 equiv). After 30 min, the pH of the solution was adjusted to 7 with saturated NaHCO3(aq)and the slurry was extracted with CH2Cl2. The organic layer was washed with 2×200 of H2O and 2×200 mL of brine. The mixture was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 2.25 g (74%) of 334-3 as a white solid. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-hydrazinyl-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate dihydrochloride (334-4) Into a 100-mL round-bottom flask was placed 334-3 (700 mg, 0.85 mmol), CH2Cl2(6 mL), 2,6-lutidine (0.5 mL, 4 equiv) followed by TMSOTf (756 mg, 3.4 mmol, 4 equiv) at 0° C. The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was washed with 3×200 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 683 mg (quant) of crude 334-4 as a yellow solid. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (334-4) Into a 50-mL round-bottom flask was placed 334-4 (684 mg, 0.95 mmol), EtOH (6 mL), and ethyl 2-formyl-3-oxopropanoate (144 mg, 1 mmol, 1.05 equiv). The reaction slurry was stirred for 2 h at room temperature then concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (3:7) to provide 567 mg (72%) of 334-5 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (334-6) Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed 334-5 (573 mg, 0.69 mmol), THF (2 mL), MeOH (6 mL), p-TolSO2Na (500 mg, 2.8 mmol, 4 equiv), and Pd(PPh3)4(400 mg, 0.35 mmol, 0.5 equiv). The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was extracted with 200 mL of CH2Cl2and the solution pH adjusted to 3 with 1 M HCl(aq). The mixture was concentrated and residue was applied onto a silica gel column with CH2Cl2:methanol (10:1) to provide 508 mg (93%) of 334-6 as a yellow solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (334-7) Into a 100-mL round-bottom flask was placed 334-6 (288 mg, 0.37 mmol), DMF (5 mL), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (220 mg, 1.5 mmol, 4 equiv), KI (30 mg, 0.18 mmol, 0.5 equiv), and K2CO3(250 mg, 1.8 mmol, 5 equiv). The reaction slurry was stirred for 1 h at 60° C. The reaction mixture was cooled and extracted with 200 mL of CH2Cl2. The combined organic layers were washed with 3×200 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 248 mg (75%) of 334-7 as a yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (334-8) Into a 100-mL round-bottom flask was placed 334-7 (248 mg, 0.28 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 h at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XBridge Shield RP18 OBD Column, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (60% Phase B up to 75% in 8 min); detector, UV. This resulted in 67.0 mg (33%) of 334-8 as a white solid. MS (ES, m/z): [M+1]+=735.3;1H NMR (400 MHz, chloroform-d) δ 0.87 (d, J=10.7 Hz, 6H), 0.96 (d, J=12.3 Hz, 1H), 1.06 (d, J=13.9 Hz, 1H), 1.15 (s, 3H), 1.24 (d, J=11.2 Hz, 7H), 1.27-1.40 (m, 9H), 1.40-1.50 (m, 3H), 1.52-1.73 (m, 4H), 1.73-90 (m, 1H), 1.90-2.12 (m, 4H), 2.12-2.31 (m, 5H), 2.51 (s, 1H), 3.09 (d, J=14.0 Hz, 1H), 4.30 (q, J=7.1 Hz, 2H), 4.79 (dd, J=13.0, 3.9 Hz, 1H), 4.91 (d, J=13.8 Hz, 1H), 4.99 (d, J=13.8 Hz, 1H), 5.76 (s, 1H), 7.80 (s, 1H), 7.89 (s, 1H). Example 82 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (335-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)-1H-pyrazol-1-yl)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (335-1) The title compound was prepared from 194-7 according to the methods to synthesize 334-8. The crude product was purified by prep-HPLC with the following conditions: column, XBridge Shield RP18 OBD Column, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (65% Phase B up to 80% in 8 min); detector, UV. This resulted in 86.7 mg (37%) of 335-1 as a white solid. MS (ES, m/z): [M+1]+=763.3;1H NMR (400 MHz, chloroform-d) δ 0.87 (d, J=11.0 Hz, 6H), 0.95 (d, J=11.9 Hz, 1H), 1.06 (d, J=14.4 Hz, 2H), 1.15 (s, 3H), 1.19-1.28 (m, 13H), 1.28-1.49 (m, 11H), 1.56-1.77 (m, 4H), 1.77-1.88 (m, 1H), 1.92-2.12 (m, 4H), 2.19-2.33 (m, 2H), 2.51 (s, 1H), 3.01 (h, J=6.9 Hz, 1H), 3.09 (d, J=13.7 Hz, 1H), 4.30 (q, J=7.1 Hz, 3H), 4.40 (m, 2H), 4.80 (dd, J=13.0, 4.0 Hz, 1H), 4.93 (d, J=13.7 Hz, 1H), 5.00 (d, J=13.8 Hz, 1H), 5.76 (s, 1H), 7.81 (s, 1H), 7.90 (s, 1H). Example 83 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(5-(ethoxycarbonyl)-3-methyl-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (336-2) Synthesis of ethyl (E)-2-(methoxyimino)-4-oxopentanoate Into a 100-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed ethyl 2,4-dioxopentanoate (2 g), ethanol (13.3 mL), and H-methyl hydroxyl amine hydrochloride (1.11 g, 1.05 equiv). The reaction slurry was stirred for 2 days at room temperature. The reaction mixture was concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:10) to provide 1.24 g (52%) of ethyl (E)-2-(methoxyimino)-4-oxopentanoate as a light yellow oil. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(5-(ethoxycarbonyl)-3-methyl-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (336-1) Into a 50-mL round-bottom flask, was placed 334-4 (375 mg), ethanol (10 mL), and ethyl (E)-2-(methoxyimino)-4-oxopentanoate (195 mg, 2 equiv). The reaction slurry was stirred for 2 h at 80° C. The reaction mixture was concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:3) to provide 318 mg (72%) of 336-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(5-(ethoxycarbonyl)-3-methyl-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (336-2) The title compound was prepared with 336-1 according to the methods to synthesize 334-8. The crude product was purified by prep-HPLC with the following conditions: column, XBridge Prep OBD C18 Column, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (67% phase B up to 69% in 8 min); detector, UV. This resulted in 55.6 mg (51%) of 336-2 as a white solid. MS (ES, m/z): [M+1]+=749.35;1H NMR (400 MHz, methanol-d4) δ 0.86 (s, 3H), 0.94 (d, J=13.6 Hz, 1H), 1.08 (d, J=13.6 Hz, 1H), 1.20 (d, J=10.4 Hz, 6H), 1.28 (d, J=10.4 Hz, 7H), 1.33-1.48 (m, 8H), 1.51 (s, 3H), 1.62-1.82 (m, 4H), 1.85-2.09 (m, 4H), 2.15 (s, 3H), 2.19-2.30 (m, 5H), 2.52-2.68 (m, 2H), 2.95 (d, J=13.6 Hz, 1H), 4.25-4.44 (m, 2H), 4.61 (d, J=14.0 Hz, 1H), 4.96 (d, J=14.0 Hz, 1H), 5.64 (s, 1H), 5.67 (d, J=3.6 Hz, 1H), 6.57 (s, 1H). Example 84 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(3-(ethoxycarbonyl)-5-methyl-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (337-2) Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(3-(ethoxycarbonyl)-5-methyl-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (337-1) Into a 100-mL round-bottom flask was placed 334-4 (392 mg), acetic acid (4 mL), and ethyl 2,4-dioxopentanoate (0.076 mL, 1 equiv). The reaction slurry was stirred for 1 h at room temperature. The solution pH was adjusted to 8 with saturated NaHCO3(aq)and the mixture was extracted with ethyl acetate. The organic layer was washed with 2×100 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:3) to provide 352 mg (77%) of 337-1 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(3-(ethoxycarbonyl)-5-methyl-1H-pyrazol-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (337-2) The title compound was prepared with 337-1 according to the methods to synthesize 334-8. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH Prep OBD C18 column, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (65% phase B up to 85% in 8 min); detector, UV. This resulted in 80.6 mg (52%) of 337-2 as a white solid. MS (ES, m/z): [M+1]+=749.30;1H NMR (300 MHz, methanol-d4) δ 0.84 (s, 3H), 0.90 (d, J=10.5 Hz, 1H), 1.06 (d, J=12.0 Hz, 1H), 1.12-1.50 (m, 24H), 1.52-2.00 (m, 8H), 2.03 (s, 1H), 2.08-2.38 (m, 8H), 2.50-2.78 (m, 2H), 2.94 (d, J=13.5 Hz, 1H), 4.32 (q, 7=7.1 Hz, 2H), 4.62-4.79 (m, 2H), 5.10 (d, J=13.8 Hz, 1H), 5.62 (s, 1H), 6.46 (s, 1H). Example 85 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(4-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (338-4) Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(4-methyl-1H-pyrazol-1-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (338-1) Into a 50-mL round-bottom flask was placed 334-4 (378 mg, 0.53 mmol), acetic acid (5 mL), and (2Z)-3-(dimethylamino)-2-methylprop-2-enal (0.12 mL, 2 equiv). The reaction slurry was stirred for 1.5 h at room temperature. The solution pH was adjusted to 8 with saturated NaHCO3(aq) and the mixture was extracted with ethyl acetate. The organic layer was washed with 2×100 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:3) to provide 284 mg (70%) of 338-1 as a light yellow solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-4,6a,6b, 8a, 11,14b-h exam ethyl-3-(4-methyl-1H-pyrazol-1-yl)-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (338-2) Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed 338-1 (284 mg), THF (2 mL), MeOH (6 mL), and sodium toluene-4-sulphinate (263 mg, 4 equiv), and Pd(PPh3)4(213 mg, 0.5 equiv). The reaction slurry was stirred for 1 h at room temperature then concentrated. The residue was applied onto a silica gel column with CH2Cl2:methanol (10:1) to provide 420 mg (quant) of 338-2 as a yellow solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(4-methyl-1H-pyrazol-1-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (338-3) Into a 50-mL round-bottom flask was placed 338-2 (170 mg), DMF (5 mL), KI (19.3 mg, 0.5 equiv), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (69 mg, 2 equiv), and K2CO3(96.4 mg, 3 equiv). The reaction slurry was stirred for 30 min at 60° C. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The organic layer was washed with 2×100 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:3) to provide 112 mg (57%) of 338-3 as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(4-methyl-1H-pyrazol-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (338-4) Into a 50-mL round-bottom flask was placed 338-3 (112 mg), CH2Cl2(5 mL), and TFA (0.5 mL). The resulting solution was stirred for 30 min at room temperature. The resulting mixture was concentrated under vacuum. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSHPrep OBD C18 column, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (65% phase B up to 72% in 8 min); detector, UV. This resulted in 80.5 mg (89%) of 338-4 as a white solid. MS (ES, m/z): [M+1]+=677.35;1H NMR (400 MHz, methanol-d4) δ 0.83 (s, 3H), 0.89 (s, 1H), 0.92 (s, 3H), 1.05 (d, J=13.6 Hz, 1H), 1.16 (d, J=14.8 Hz, 6H), 1.23 (s, 3H), 1.25-1.50 (m, 9H), 1.59-1.99 (m, 8H), 2.03 (s, 3H), 2.10-2.17 (m, 1H), 2.18 (s, 3H), 2.20-2.29 (m, 1H), 2.32-2.50 (m, 1H), 2.61 (s, 1H), 2.96 (d, J=14.0 Hz, 1H), 4.71 (dd,7=12.8, 3.6 Hz, 1H), 4.84 (s, 1H), 5.12 (d, J=14.0 Hz, 1H), 5.61 (s, 1H), 7.19 (s, 1H), 7.37 (s, 1H). Example 86 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-10-(4-methyl-1H-pyrazol-1-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (339-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-10-(4-methyl-1H-pyrazol-1-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (339-1) The title compound was prepared from 338-2 according to the methods to synthesize 338-4. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH C18 OBD column, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (65% Phase B up to 79% in 8 min); detector, UV. This resulted in 29.4 mg (40%) of 339-1 as a white solid. MS (ES, m/z): [M+1]+=691.30;1H NMR (400 MHz, methanol-d4) δ 0.83 (s, 3H), 0.92 (s, 3H), 1.05 (d, J=14.8 Hz, 1H), 1.14 (s, 3H), 1.18-1.20 (m, 6H), 1.23 (s, 3H), 1.26-1.44 (m, 7H), 1.46 (s, 3H), 1.59-1.81 (m, 5H), 1.82-1.91 (m, 2H), 1.95 (d, J=10.4 Hz, 1H), 2.03 (s, 3H), 2.12-2.28 (m, 2H), 2.30-2.45 (m, 1H), 2.52-2.64 (m, 3H), 2.96 (d, J=13.6 Hz, 1H), 4.72 (dd, J=13.1, 3.9 Hz, 1H), 4.89 (s, 1H), 5.12 (d, J=13.6 Hz, 1H), 5.61 (s, 1H), 7.17 (s, 1H), 7.35 (s, 1H). Example 87 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (341-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (341-1) Methyl bromoacetate (33 μL, 0.36 mmol) was added to a slurry of 281-1 (150 mg, 0.18 mmol) and K2CO3(124 mg, 0.90 mmol) in DMF (1.5 mL) and stirred at RT. After 90 minutes, EtOAc (50 mL) was added. The mixture was washed with water (3×10 mL), dried (Na2SO4) and concentrated onto SiO2(3 g). Purification by flash chromatography (4 g SiO2, 30-70% EtOAc/DCM) gave 341-1 (69 mg). Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (341-2) Trifluoroacetic acid (0.30 mL) was added to a solution of 341-1 (19 mg, 0.022 mmol) in CH2Cl2(0.30 mL). After stirring for 30 minutes, the solution was concentrated under vacuum and purified by preparative HPLC to give a TFA salt of the title compound (14 mg). MS (ES, m/z): [M+H]+=684.3;1H NMR (400 MHz, methanol-d4) δ 5.60 (s, 1H), 5.61 (s, 1H), 5.20 (d, J=14.1, 1H), 4.98 (d, J=14.0 Hz, 1H), 4.02 (dd, J=17.1, J=27.4 Hz, 2H), 3.84 (s, 3H), 3.69 (dd, J=4.3 Hz, J=12.5 Hz, 1H), 2.90 (dt, J=14.1 Hz, J=2.5 Hz, 1H), 2.54 (s, 1H), 2.22 (s, 3H), 1.42 (s, 3H), 1.28 (s, 3H), 1.22 (s, 3H), 1.17 (s, 3H), 1.14 (s, 3H), 0.83 (s, 3H). Example 88 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-amino-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (342-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (342-1) The title compound was prepared through the same route as for 341-2 but using 2-bromoacetamide. The crude product was purified by preparative HPLC to give a TFA salt of 342-1 (13 mg). MS (ES, m/z): [M+H]+=724.3;1H NMR (400 MHz, methanol-d4) δ 5.61 (s, 1H), 5.22 (d, J=14.1 Hz, 1H), 4.94 (d, J=14.1 Hz, 1H), 3.80 (dd, J=27.2 Hz, J=16.0 Hz, 2H), 3.59 (dd, J=12.6 Hz, J=4.5 Hz, 1H), 2.90 (dt, J=13.5 Hz, J=3.0 Hz, 1H), 2.55 (s, 1H), 2.22 (s, 3H), 1.43 (s, 3H), 1.29 (s, 3H), 1.19 (s, 3H), 1.17 (s, 3H), 1.14 (s, 3H), 0.83 (s, 3H). Example 89 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (343-3) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-ethoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (343-1) Ethyl bromoacetate (68 μL, 0.61 mmol) was added to a slurry 281-1 (250 mg, 0.31 mmol) and K2CO3(212 mg, 1.53 mmol) in DMF (2.5 mL) and stirred at RT. After 75 minutes, EtOAc (65 mL) was added. The mixture was washed with water (3×20 mL), dried (Na2SO4) and concentrated onto SiO2(3 g). Purification by flash chromatography (12 g SiO2, 30-70% EtOAc/DCM) gave 343-1 (117 mg). Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-ethoxy-2-oxoethyl)(methyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (343-2) Iodomethane (59 μL, 0.95 mmol) was added to a slurry of 343-1 (117 mg, 0.14 mmol) and K2CO3(131 mg, 0.95 mmol) in DMF (1.3 mL) and heated at 45° C. After 75 minutes, EtOAc (50 mL) was added. The mixture was washed with water (3×20 mL), dried (Na2SO4) and concentrated. Purification by flash chromatography (12 g SiO2, 10-60% EtOAc/hexane) gave 343-2 (80 mg). Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2-ethoxy-2-oxoethyl)(methyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (343-3) Trifluoroacetic acid (0.30 mL) was added to a solution of 343-2 (80 mg, 0.091 mmol) in CH2Cl2(0.30 mL). After stirring for 30 minutes, the solution was concentrated under vacuum and purified by preparative HPLC to give a TFA salt of 343-3 (58 mg). MS (ES, m/z): [M+H]+=712.3;1H NMR (400 MHz, methanol-d4) δ 5.60 (s, 1H), 5.22 (d, J=13.8 Hz, 1H), 4.93 (d, J=13.7 Hz, 1H), 4.27 (quar, J=7.2 Hz, 2H), 4.00 (m, 1H), 3.88 (m, 1H), 3.72 (m, 1H), 3.92 (dt, J=13.3 Hz, J=3.0 Hz, 1H), 2.80 (s, 3H), 2.53 (s, 1H), 2.22 (s, 3H), 1.42 (s, 3H), 1.33 (s, 3H), 1.31 (t, J=7.2 Hz), 1.19 (s, 3H), 1.17 (s, 3H), 1.13 (s, 3H), 0.83 (s, 3H). Example 90 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(allyl(2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (344-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(allyl(2-methoxy-2-oxoethyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (344-1) The title compound was prepared from 341-1 through the same route as for 343-3 but using allylbromide. The crude product was purified by preparative HPLC to give a TFA salt of 344-1 (13 mg). MS (ES, m/z): [M+H]+=724.3;1H NMR (400 MHz, methanol-d4) δ 5.78 (m, 1H), 5.59 (s, 1H), 5.25 (m, 2H), 5.18 (d, J=13.8 Hz), 4.87 (d, 1H), 3.71 (s, 3H), 3.59-3.36 (m, 5H), 2.86 (dt, J=13.7 Hz, J=2.5 Hz, 1H), 2.53 (s, 1H), 2.21 (s, 3H), 1.43 (s, 3H), 1.23 (s, 3H), 1.17 (s, 3H), 1.16 (s, 3H), 1.12 (s, 3H), 0.83 (s, 3H). Example 91 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((S)-1-methoxy-1-oxopropan-2-yl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (345-2) Synthesis of methyl (2,V)-2-(trifluoromethyl sulfonyloxy)propanoate Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed methyl (2S)-2-hydroxypropanoate (1.00 g, 9.6 mmol) and CH2Cl2(40 mL) followed by Tf2O (1.77 mL, 6.3 mmol, 1.1 equiv) dropwise with stirring at 0° C. To this was added 2,6-lutidine (1.53 mL, 14.3 mmol, 1.4 equiv) at 0° C. The reaction slurry was stirred for 2 hr at room temperature. The reaction mixture was concentrated and the residue applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 1.2 g (53%) of methyl (2S)-2-(trifluoromethanesulfonyloxy)propanoate as a yellow oil. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((S)-1-methoxy-1-oxopropan-2-yl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (345-1) Into a 8-mL vial was placed methyl (2S)-2-(trifluoromethanesulfonyloxy)propanoate (16 mg, 2.5 equiv) and CH2Cl2(0.8 mL). The slurry was cooled to 0° C. and 1,8-bis(dimethylamino)naphthalene (14 mg, 2.5 equiv) in CH2Cl2(0.2 mL) was added dropwise followed by a solution of 281-1 (20 mg, 1.0 equiv) in CH2Cl2(0.2 mL) dropwise with stirring at 0° C. The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was concentrated and the residue applied onto a silica gel column with ethyl acetate:petroleum ether (1:2) to provide 15 mg (68%) of 345-1 as a yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((S)-1-methoxy-1-oxopropan-2-yl)amino)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (345-2) Into a 25-mL round-bottom flask was placed 345-1 (200 mg), CH2Cl2(5 mL), and TFA (0.5 mL). The reaction slurry was stirred for 30 min at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.1% TFA) and CH3CN (30% phase B up to 55% in 9 min); detector, UV. This resulted in 119 mg (72%) of 345-2 as a white solid MS (ES, m/z): [M+H]+=698;1H NMR (300 MHz, methanol-d4) δ 0.82-0.88 (s, 3H), 0.92-0.98 (s, 1H), 1.02-1.13 (d, J=13.1 Hz, 1H), 1.13-1.24 (m, 9H), 1.28-1.34 (s, 3H), 1.41-1.47 (s, 6H), 1.56-1.65 (d, J=7.2 Hz, 3H), 1.65-1.71 (s, 4H), 1.71-1.80 (d, J=13.2 Hz, 1H), 1.81-1.91 (d, J=12.0 Hz, 2H), 1.95-2.01 (s, 2H), 2.19-2.23 (s, 2H), 2.23-2.28 (s, 3H), 2.53-2.59 (s, 1H), 2.85-2.95 (d, J=13.7 Hz, 1H), 3.33-3.39 (s, 2H), 3.54-3.63 (d, J=8.4 Hz, 1H), 3.85-3.91 (s, 3H), 4.30-4.38 (d, J=7.3 Hz, 1H), 5.24-5.35 (d, J=14.0 Hz, 1H), 5.60-5.66 (s, 1H), 7.26-7.42 (dt, J=7.5, 15.1 Hz, 1H). Example 92 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(1-methylcyclopropane-1-carboxamido)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (346-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-(1-methyl cyclopropane-1-carboxamido)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (346-1) To a mixture of 281-1·HCl (50.2 mg, 0.0616 mmol) and 1-methylcyclopropane-1-carboxylic acid (9 mg, 0.09 mmol, 1.5 equiv) in DMF (0.4 mL) were added iPr2EtN (32.2 μL, 0.19 mmol, 3 equiv) and HATU (28 mg, 0.074 mmol, 1.2 equiv). The reaction slurry was stirred at rt overnight. Upon completion, the reaction mixture was diluted with water, washed with 2×H2O and 1×brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (0-20% EtOAc in CH2Cl2) to provide 346-1 as a white solid (32 mg, 61%). Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-(l -methyl cyclopropane-1-carboxamido)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (346-2) To a mixture of 346-1 (32 mg, 0.038 mmol) in CH2Cl2(0.5 mL) was added TFA (0.5 mL). The mixture was stirred at room temperature for 1 hour, concentrated, and purified by column (0-30% EtOAc/CH2Cl2) to give 346-2 as a white solid (20 mg, 76%). LCMS (ES, m/z) [M+H]+=694.1;1H-NMR (400 MHz, chloroform-7) δ 5.70 (s, 1H), 5.53 (d, J=9.4 Hz, 1H), 4.92 (d, J=13.8 Hz, 1H), 4.68 (d, J=13.9 Hz, 1H), 4.36-4.23 (m, 1H), 2.83 (d, J=13.7 Hz, 1H), 2.45 (s, 1H), 2.25-2.19 (m, 1H), 2.18 (s, 3H), 2.12-1.49 (m, 12H), 1.46-1.41 (m, 1H), 1.39 (s, 3H), 1.34 (d, J=15.4 Hz, 2H), 1.26 (s, 3H), 1.23 (s, 3H), 1.20-1.16 (m, 1H), 1.14 (d, J=1.8 Hz, 6H), 1.09 (s, 3H), 1.07 (dd, J=6.3, 3.6 Hz, 2H), 1.02 (d, J=13.6 Hz, 1H), 0.86 (d, J=12.8 Hz, 1H), 0.81 (s, 3H), 0.53 (dd, J=6.4, 3.7 Hz, 2H). Example 93 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(2-oxopyrrolidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic (347-4) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-chlorobutanamido)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (347-1) Into a 100-mL round-bottom flask was placed 281-1 (150 mg), CH2Cl2(2 mL), and Et3N (0.077 mL, 3 equiv). To this slurry was added 4-chlorobutanoyl chloride (0.022 mL, 1.05 equiv) dropwise at room temperature. The reaction slurry was stirred for 2 h at room temperature. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 100 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 140 mg of crude 347-1 as a yellow solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-3-(2-oxopyrrolidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (347-2) Into a 100-mL round-bottom flask was placed 347-1 (140 mg), THF (3 mL), and t-BuOK (54 mg, 3 equiv). The reaction slurry was stirred for 48 h at room temperature. The reaction mixture was diluted with neutralized with 0.1 M HCl(aq) and extracted with 3×100 mL of CH2Cl2. The combined organic layers were washed with 3×100 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 120 mg of crude 347-2 as a yellow solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(2-oxopyrrolidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (347-3) Into a 100-mL round-bottom flask was placed 347-2 (102 mg), DMF (0.5 mL), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (41 mg, 2 equiv), K2CO3(58 mg, 3 equiv), and KI (12 mg, 0.5 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with water and extracted with 3×100 mL of ethyl acetate. The combined organic layers were washed with 3×150 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:5) to provide 100 mg of 347-3 as a yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(2-oxopyrrolidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (347-4) Into a 50-mL round-bottom flask was placed 347-3 (100 mg), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was dried over anhydrous Na2SO4, filtered, and concentrated. The crude product was purified by Prep-HPLC with the following conditions: column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (50% phase B up to 55% in 8 min); detector, UV. This resulted in 28.2 mg of 347-4 as a white solid. MS (ES, m/z): [M+1]+=670;1H NMR (300 MHz, methanol-d4) δ 5.63 (s, 1H), 5.06 (d, J=13.9 Hz, 1H), 4.71 (d, J=13.9 Hz, 1H), 4.30 (dd, J=12.8, 3.9 Hz, 1H), 3.55 (q, J=8.3, 7.4 Hz, 2H), 2.89 (d, J=13.5 Hz, 1H), 2.61 (s, 1H), 2.33 (d, J=7.8 Hz, 1H), 2.27 (d, J=13.4 Hz, 1H), 2.21 (s, 3H), 2.11-1.96 (m, 2H), 1.92 (d, J=11.4 Hz, 1H), 1.86 (s, 1H), 1.71 (dt, J=24.0, 12.9 Hz, 1H), 1.48 (s, 3H), 1.41 (d, J=13.5 Hz, 4H), 1.30 (s, 1H), 1.27-1.14 (m, 13H), 1.08 (d, J=13.1 Hz, 1H), 0.86 (s, 4H). Example 94 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (348-11) Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((tert-butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (348-1) Into a 50-mL round-bottom flask was placed 280-5 (600 mg, 0.78 mmol, 1 equiv), DMF (6 mL), allyl bromide (0.27 mL, 4 equiv), KI (66 mg, 0.4 mmol, 0.5 equiv), and K2CO3(540 mg, 3.9 mmol, 5 equiv). The reaction mixture was stirred for 1 hr at room temperature then diluted with 300 mL of CH2Cl2. The resulting mixture was washed with 3×300 ml of brine and the organic layer dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with 1:5 ethyl acetate:petroleum ether to provide 616 mg (98%) of 348-1 as a white solid. Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (348-2) Into a 100-mL round-bottom flask was placed 348-1 (616 mg, 1 equiv), CH2Cl2(50 mL), and 2,6-lutidine (0.44 mL, 5 equiv) followed by the addition of TMSOTf (679 mg, 4 equiv) at 0° C. The reaction mixture was stirred for 1 hr at room temperature. The reaction slurry was washed with 2×2 M HCl and 2 x brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 600 mg (quant) of crude 348-2 as a yellow solid (HCl salt). Synthesis of 9-allyl 2-benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (348-3) Into a 25-mL sealed tube was placed 348-2 (600 mg, 0.85 mmol, 1 equiv), CH3CN (1 mL), methyl 2,5-dibromopentanoate (0.133 mL, 0.85 mmol, 1 equiv), and iPr2EtN (0.70 mL, 4.0 mmol, 4.7 equiv). The reaction mixture was stirred for 48 hr at 82° C. Upon completion, the reaction slurry was concentrated. The residue was applied onto a silica gel column with 1:5 ethyl acetate:petroleum ether to provide 363 mg (52%) of 348-3 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-(2-(methoxycarbonyl)pyrrolidin-1-yl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (348-4) Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed 348-3 (363 mg, 0.44 mmol, 1 equiv), THF (1 mL), MeOH (3 mL), TolSO2Na (316 mg, 1.78 mmol, 4 equiv), and Pd(PPh3)4(256 mg, 0.22 mmol, 0.5 equiv). The reaction mixture was stirred for 1 hr at room temperature. Upon completion the reaction slurry was concentrated and the residue applied onto a silica gel column with 10:1 CH2Cl2:methanol to provide 300 mg (87%) of 348-4 as a yellow solid. Synthesis of 2-benzhydryl 9-((5-ethyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (348-5) Into a 100-mL round-bottom flask was placed 348-4 (940 mg, 1.2 mmol, 1 equiv), DMF (10 mL), 4-(bromomethyl)-5-ethyl-1,3-dioxol-2-one (800 mg, 3.9 mmol, 3.2 equiv), KI (100 mg, 0.6 mmol, 0.5 equiv), and K2CO3(830 mg, 6 mmol, 5 equiv). The reaction slurry was stirred for 1 hr at 60° C. The reaction mixture was extracted with 300 mL of CH2Cl2and the organic layer washed with 3×300 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 780 mg (71%) of 348-5 as a brown solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (348-6) Into a 50-mL round-bottom flask was placed 348-5 (350 mg, 0.38 mmol, 1 equiv), CH2Cl2(5 mL), and TFA (0.5 mL). The reaction mixture was stirred for 1 hr at room temperature then concentrated. The crude product was purified by Prep-HPLC with the following conditions: column, XBridge Prep OBD C18, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (30% phase B up to 60% in 8 min); detector, UV to provide 40.2 mg (14%) of 348-6 as a white solid. MS (ES, m/z): [M+1]+=738.3;1H NMR (400 MHz, methanol-d4) δ 0.84 (s, 3H), 0.92 (d, J=9.7 Hz, 1H), 1.07 (d, J=14.5 Hz, 1H), 1.15 (s, 3H), 1.17 (d, J=6.9 Hz, 6H), 1.20-1.28 (m, 5H), 1.33 (s, 3H), 1.35-1.46 (m, 7H), 1.51-1.78 (m, 4H), 1.81-2.01 (m, 6H), 2.01-2.26 (m, 4H), 2.38 (s, 1H), 2.53 (s, 1H), 2.64 (q, J=7.5 Hz, 2H), 2.91 (d, J=13.8 Hz, 1H), 3.20 (d, J=8.8 Hz, 1H), 3.60 (s, 1H), 3.88 (s, 4H), 4.50 (s, 1H), 4.96 (d, J=14.0 Hz, 1H), 5.29 (d, J=13.9 Hz, 1H), 5.62 (s, 1H). Example 95 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (349-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (349-1) The title compound was prepared from 348-2 through the same route as for 348-6. The crude product was purified by prep-HPLC with the following conditions: column, XBridge Prep C18 OBD, 5 μm, 19*150 mm; mobile phase, water (0.1% TFA) and CH3CN (45% phase B up to 75% in 11 min); detector, UV. This resulted in 58.0 mg of 349-1 as a white solid. MS (ES, m/z): [M+H]+=724.3;1H NMR (400 MHz, methanol-d4) δ 0.84 (s, 3H), 0.93 (d, J=11.2 Hz, 1H), 1.07 (d, J=13.9 Hz, 1H), 1.11-1.21 (m, 9H), 1.21-1.31 (m, 2H), 1.32 (s, 3H), 1.36-1.51 (m, 7H), 1.57 (d, J=10.9 Hz, 1H), 1.60-1.78 (m, 3H), 1.78-2.02 (m, 6H), 2.02-2.30 (m, 7H), 2.40-2.47 (m, 1H), 2.53 (s, 1H), 2.80-3.02 (m, 1H), 3.24 (q, J=9.3 Hz, 1H), 3.65 (s, 1H), 3.90 (s, 3H), 3.96 (d, J=11.1 Hz, 1H), 4.61 (s, 1H), 4.96 (d, J=14.1 Hz, 1H), 5.31 (d, J=14.0 Hz, 1H), 5.62 (s, 1H). Example 96 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (350-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((S)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (350-1) The title compound was prepared from 348-2 through the same route as for 348-6. The crude product was purified by prep-HPLC with the following conditions: column, Sunfire Prep C18 OBD, 10 μm, 19*250 mm; mobile phase, water (0.05% TFA) and CH3CN (50% phase B up to 56% in 8 min); detector, UV. This resulted in 23 mg of 350-1 as a white solid. MS (ES, m/z) [M+H]+=752.35;1H NMR (400 MHz, methanol-d4) δ 0.84 (s, 3H), 0.90 (d, J=5.7 Hz, 1H), 1.00-1.10 (m, 1H), 1.12-1.21 (m, 9H), 1.21-1.40 (m, 15H), 1.40-1.50 (m, 6H), 1.60-1.78 (m, 4H), 1.78-2.01 (m, 7H), 2.10-2.27 (m, 3H), 2.40 (s, 1H), 2.55 (s, 1H), 2.91-3.00 (m, 1H), 3.04-3.16 (m, 2H), 3.54 (s, 1H), 3.87 (s, 3H), 4.96 (d, J=13.9 Hz, 1H), 5.04 (d, J=0.8 Hz, 1H), 5.27 (d, J=13.9 Hz, 1H), 5.62 (s, 1H). Example 97 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((R)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (351-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-(((5-isopropyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((R)-2-(methoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (351-1) The title compound was prepared from 348-2 through the same route as for 348-6. The crude product was purified by prep-HPLC with the following conditions: column, Sunfire Prep C18 OBD, 10 μm, 19*250 mm; mobile phase, water (0.05% TFA) and CH3CN (50% phase B up to 56% in 8 min); detector, UV. This resulted in 29 mg of 351-1 as a white solid. MS (ES, m/z) [M+H]+=752.35;1H NMR (400 MHz, methanol-d4) δ 0.84 (s, 3H), 0.90 (d, J=5.7 Hz, 1H), 1.00-1.10 (m, 1H), 1.10-1.16 (m, 4H), 1.16-1.22 (m, 6H), 1.22-1.33 (m, 11H), 1.36-1.50 (m, 6H), 1.60-1.78 (m, 4H), 1.79-1.91 (m, 3H), 1.91-2.10 (m, 5H), 2.10-2.27 (m, 2H), 2.40 (s, 1H), 2.55 (s, 1H), 2.91-3.00 (m, 1H), 3.04-3.16 (m, 2H), 3.45 (s, 1H), 3.54 (s, 1H), 3.87 (s, 4H), 4.44 (s, 1H), 5.36 (d, J=14.0 Hz, 1H), 5.62 (s, 1H). Example 98 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-2-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (352-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-2-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (352-1) The title compound was prepared from 348-2 through the same route as for 348-6. The crude product was purified by prep-HPLC with the following conditions: column, Sunfire Prep C18 OBD, 10 μm, 19*250 mm; mobile phase, water (0.1% TFA) and CH3CN (37% phase B up to 40% in 8 min); detector, UV. This resulted in 32.9 mg of 352-1 as a white solid. MS (ES, m/z) [M+H]+=738.35;1H NMR (400 MHz, methanol-d4) δ 0.84 (s, 3H), 0.92 (d, J=7.7 Hz, 1H), 1.06 (d, J=14.0 Hz, 1H), 1.10-1.20 (m, 11H), 1.20-1.36 (m, 9H), 1.36-1.47 (m, 8H), 1.50-1.61 (m, 1H), 1.61-2.00 (m, 11H), 2.10-2.26 (m, 7H), 2.53 (s, 1H), 2.90 (d, J=13.7 Hz, 1H), 4.33 (d, J=14.6 Hz, 2H), 5.26 (s, 1H), 5.62 (s, 1H). Example 99 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-2-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (353-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-2-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (353-1) The title compound was prepared from 348-2 through the same route as for 348-6. The crude product was purified by prep-HPLC with the following conditions: column, Sunfire Prep C18 OBD, 10 μm, 19*250 mm; mobile phase, water (0.1% TFA) and CH3CN (37% phase B up to 40% in 8 min); detector, UV. This resulted in 36.1 mg of 353-1 as a white solid. MS (ES, m/z) [M+H]+=738.35;1H NMR (400 MHz, methanol-d4) δ 0.84 (s, 3H), 0.91 (s, 1H), 1.07 (d, J=13.7 Hz, 1H), 1.12-1.23 (m, 9H), 1.25-1.37 (m, 8H), 1.38-1.51 (m, 6H), 1.60-1.77 (m, 4H), 1.81-1.89 (m, 3H), 2.07 (d, J=12.6 Hz, 5H), 2.10-2.26 (m, 5H), 2.40 (s, 1H), 2.55 (s, 1H), 2.95 (d, J=13.8 Hz, 1H), 3.46 (s, 1H), 3.55 (s, 1H), 3.88 (s, 1H), 4.33 (qq, J=7.1, 3.5 Hz, 2H), 4.44 (s, 1H), 5.31 (d, J=14.0 Hz, 1H), 5.62 (s, 1H). Example 100 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(piperidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (356-2) Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(piperidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (356-1) Into a 100-mL round-bottom flask, was placed 281-1 (300 mg, 0.39 mmol), DMF (5 mL), K2CO3(267 mg, 1.9 mmol, 5 equiv), and 1,5-dibromopentane (176 mg, 0.77 mmol, 2 equiv). The reaction slurry was stirred for 2 d at 40° C. The reaction mixture was diluted with water and extracted with 300 mL of CH2Cl2. The organic layer was washed with 3×300 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 167 mg (51%) of 356-1 as a yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-10-(piperidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (356-2) Into a 100-mL round-bottom flask, was placed 356-1 (167 mg, 0.2 mmol), CH2Cl2(10 mL), and TFA (1 mL). The reaction slurry was stirred for 1 h at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (35% phase B up to 41% in 8 min); detector, UV. This resulted in 27.8 mg (21%) of 356-2 as a white solid. MS (ES, m/z): [M+1]+=680.30;1H NMR (400 MHz, methanol-d4) δ 0.84 (s, 3H), 1.07 (d, J=14.0 Hz, 1H), 1.13-1.33 (m, 13H), 1.35-1.47 (m, 11H), 1.51-1.78 (m, 4H), 1.79-1.88 (m, 4H), 1.90-2.00 (m, 4H), 2.01-2.16 (m, 2H), 2.17-1.31 (m, 4H), 2.54 (s, 1H), 2.96-3.09 (m, 1H), 2.90-3.17 (m, 3H), 3.36-3.78 (m, 1H), 3.63 (d, J=12.3 Hz, 1H), 3.87 (dd, J=12.5, 4.0 Hz, 1H), 5.07 (d, J=13.9 Hz, 1H), 5.20 (d, J=14.0 Hz, 1H), 5.62 (s, 1H). Example 101 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (357-7) Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((tert-butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (357-1) Into a 250-mL round-bottom flask was placed 280-5 (6.0 g, 7.8 mmol), CH2Cl2(30 mL), MeOH (30 mL), and TMSCHN2(4.47 g, 39 mmol, 5 equiv). The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was diluted with water and extracted with 300 mL of CH2Cl2. The organic layer was washed with 3×300 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 6.12 g (100%) of crude 357-1 as a yellow solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (357-2) Into a 250-mL round-bottom flask was placed 357-1 (6.12 g, 7.85 mmol), CH2Cl2(66 mL), and 2, 6-lutidine (4.15 g, 39 mmol, 5 equiv) followed by TMSOTf (6.99 g, 31.5 mmol, 4 equiv) at 0° C. The reaction slurry was stirred overnight at room temperature. The reaction was quenched with water and the mixture was extracted with 500 mL of CH2Cl2. The organic layer was washed with 3×500 ml of 1 M HCl(aq), dried over anhydrous Na2SO4, filtered, and concentrated to provide 5.05 g (95%) of 357-2 as a white solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(4-oxopiperidin-1-yl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (357-3) Into a 100-mL round-bottom flask was placed 357-2 hydrochloride (500 mg), 1-ethyl-1-methyl-4-oxop-iperidin-1-ium iodide (396 mg), NaHCO3(154 mg), EtOH (10 mL), and H2O (2 mL). The reaction slurry was stirred for 1 hr at 80° C. The reaction mixture was diluted with 100 mL of CH2Cl2and washed with 3×150 ml of brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated to provide 420 mg of 357-3 as a light yellow solid. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (357-4) Into a 100-mL round-bottom flask was placed XtalFluor-M (CAS: 63517-33-9, 151 mg), Et3N (1 mg), CH2Cl2(10 mL), 3HF.Et3N (149 mg), and 357-3 (300 mg). The reaction slurry was stirred for 2 seconds at room temperature. The reaction mixture was applied directly onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 270 mg of 357-4 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-(4,4-difluoropiperidin-1-yl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (357-5) Into a 100-mL round-bottom flask was placed 357-4 (210 mg, 0.27 mmol) followed by n-PrSLi (5 mL, 1 M in HMPA) at 0° C. The reaction slurry was stirred for 3 h at 40° C. The reaction mixture was cooled and diluted with 100 mL of CH2Cl2. The resulting mixture was washed with 3×150 ml of 1 M HCl(aq), dried over anhydrous Na2SO4, filtered, and concentrated to provide 180 mg of crude 357-5 as a white solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (357-6) Into a 100-mL round-bottom flask was placed 357-5 (660 mg, 0.86 mmol), 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (690 mg, 4.6 mmol, 5.4 equiv), DMF (45 mL), KI (390 mg, 2.3 mmol, 2.7 equiv), and K2CO3(1.09 g, 0.008 mmol, 0.01 equiv). The reaction slurry was stirred for 1 h at 60° C. The reaction mixture was extracted with 100 mL of CH2Cl2and the organic layer washed with 3×100 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:10) to provide 660 mg (87%) of 357-6 as a yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4,4-difluoropiperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (357-7) Into a 100-mL round-bottom flask was placed 357-6 (110 mg), TFA (1 mL), and CH2Cl2(10 mL). The reaction slurry was stirred for 1 hr at room temperature then concentrated. The crude product was purified by Chiral-Prep-HPLC with the following conditions: mobile phase, hexane (0.1% formic acid) and EtOH (hold 50% EtOH in 10 min); detector, UV 254 nm. This resulted in 15.1 mg of 357-7 as a light yellow solid. MS (ES, m/z): [M+1]+=716.39;1HNMR (300 MHz, methanol-d4) δ 5.61 (s, 1H), 5.10 (d, J=14.0 Hz, 1H), 4.92 (s, 1H), 3.12 (dd, J=12.5, 3.9 Hz, 1H), 2.97-2.72 (m, 3H), 2.56 (d, J=9.3 Hz, 3H), 2.23 (s, 5H), 2.00-1.49 (m, 14H), 1.47-1.28 (m, 8H), 1.28-1.01 (m, 16H), 0.84 (s, 4H). Example 102 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-3-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (358-7) Synthesis of 2-benzhydryl 9-benzyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (358-1) Into a 100-mL round-bottom flask was placed 194-7 (1 g, 1.24 mmol), DMF (10 mL), K2CO3(1.0 g, 7.2 mmol, 5 equiv), and BnBr (636 mg, 3.7 mmol, 3 equiv). The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was diluted with 100 mL of ethyl acetate and quenched by the addition of 100 mL of water. The layers were separated, and the aqueous layer was extracted with 2×100 mL of ethyl acetate. The combined organic layers were washed with 3×200 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (eluting 0-20%) to provide 1.0 g of 358-1 as a white solid. Synthesis of 2-benzhydryl 9-benzyl (2S,4aS,6aS,6bR,8aR,9S,12aS,12bR,14bR)-2,4a,6a, 6b,9,12a-hexamethyl-10,13-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (358-2) Into a 100-mL round-bottom flask was placed 358-1 (1.0 g, 1.32 mmol), CH2Cl2(20 mL), and Dess-Martin periodinane (1.12 g, 2.64 mmol, 2 equiv). The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was diluted with 150 mL of CH2Cl2. The pH of the solution was adjusted to 8-9 with saturated NaHCO3(aq)and extracted with 2×100 mL of CH2Cl2. The combined organic layers were washed with 1×200 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (eluting 0-20%) to provide 900 mg (90%) of 358-2 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-((benzyloxy)carbonyl)-10-((R)-3-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (358-3) To a slurry of 358-2 (160 mg, 0.21 mmol) and ethyl (3R)-pyrrolidine-3-carboxylate (60.7 mg, 0.42 mmol, 2 equiv) in CH2Cl2at 0° C. was added TiCl4(161 mg, 0.85 mmol, 4 equiv) dropwise. The reaction slurry was stirred for 3 h at room temperature under nitrogen atmosphere. To the above mixture was added sodium triacetoxyborohydride (449 mg, 2.1 mmol, 10 equiv) dropwise in portions over 1 min at room temperature. The reaction slurry was stirred for additional 1 min at room temperature. The reaction was quenched with water at room temperature and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by prep-TLC (CH2Ch:MeOH 12:1) to afford 358-3 (90 mg, 59%) Synthesis of 9-benzyl 2-(2-(trimethylsilyl)ethyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-3-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (358-4) A solution of 358-3 (65 mg, 0.091 mmol) and TMSE (53.6 mg, 0.45 mmol, 5 equiv), and DMAP (44 mg, 0.36 mmol, 4 equiv) in CH2Cl2was stirred for 3 h at room temperature. To the above mixture was added EDCI (87.0 mg, 0.45 mmol, 5 equiv) at room temperature. The reaction slurry was stirred overnight at room temperature. The reaction was quenched with water at room temperature and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by prep-TLC (petroleum ether:EtOAc 3:1) to afford 358-4 (45 mg, 61%). Synthesis of (3 S,4S,4aR,6aR,6bS,8aS,11S, 12aR, 14aR,14bS)-3-((R)-3-(ethoxycarbonyl)pyrrolidin-1-yl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-11-((2-(trimethylsilyl)ethoxy)carbonyl)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (358-5) A mixture of 358-4 (45 mg, 0.055 mmol) and Pd/C (10%, 45 mg, 0.42 mmol, 7.7 equiv) in EtOAc was stirred for 1 h at room temperature under hydrogen atmosphere. The reaction mixture was filtered and the filter cake washed with EtOAc. The filtrate was concentrated under reduced pressure. The crude product 358-5 was used in the next step directly without further purification. Synthesis of 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) 2-(2-(trimethylsilyl)ethyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-3-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (358-6) A solution of 358-5 (40 mg, 0.055 mmol),4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (32.7 mg, 0.22 mmol, 4 equiv), K2CO3(22.8 mg, 0.17 mmol, 3 equiv), and KI (4.6 mg, 0.028 mmol, 0.5 equiv) in DMF was stirred for 1 h at 60° C. The reaction mixture was cooled and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by prep-TLC (petroleum ether:EtOAc 5:1) to afford 358-6 (30 mg, 65%) as a yellow oil. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-3-(ethoxycarbonyl)pyrrolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (358-7) A solution of 358-6 (30 mg, 0.036 mmol) and TFA (0.5 mL) in CH2Cl2was stirred for 3 h at room temperature. The reaction mixture was concentrated. The crude product was purified by prep-HPLC with the following conditions: Column, XBridge CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (25% to 50% over 8 min); detector, UV 254 nM. This resulted in 358-7 (9.9 mg, 37%) as an off-white solid. MS (ES, m/z) [M+1]+=738.30;1H NMR (400 MHz, methanol-d4) δ 5.63 (s, 1H), 5.35 (s, 1H), 4.22 (m, 2H), 3.84 (s, 2H), 2.94 (m, 2H), 2.50 (s, 2H), 2.21 (m, 9H), 1.87 (m, 10H), 1.61-1.33 (m, 22H), 1.19-0.97 (m, 16H), 0.86 (s, 4H). Example 103 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-3-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (359-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((R)-3-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (359-1) The title compound 359-1 was prepared through the same synthetic route as 358-7, beginning with 358-2 and ethyl (3R)-nipecotate. The crude product was purified by prep-HPLC with the following conditions: column, XBridge CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (28% to 45% over 8 min); detector, UV 254 nm. This resulted in 359-1 (9.4 mg, 36%) as an off-white solid. MS (ES, m/z): [M+1]+=752.50;1H NMR (400 MHz, methanol-d4) δ 5.64 (s, 1H), 5.75 (s, 1H), 5.11 (s, 1H), 4.22 (s, 2H), 3.94 (s, 1H), 3.01 (m, 3H), 2.50 (s, 1H), 2.01 (m, 14H), 1.67 (m, 3H), 1.61-1.33 (m, 11H), 1.22 (m, 5H), 1.19-0.97 (m, 12H), 0.86 (s, 3H). Example 104 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-3-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (360-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((S)-3-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (360-1) The title compound 360-1 was prepared through the same synthetic route as 358-7, beginning with 358-2 and ethyl (35′)-nipecotate. The crude product was purified by prep-HPLC with the following conditions: column, XBridge CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (25% to 45% over 10 min); detector, UV 254 nm. This resulted in 360-1 (7.0 mg, 26%) as an off-white solid. MS (ES, m/z): [M+1]+=752.50;1H NMR (400 MHz, methanol-d4) δ 5.64 (s, 1H), 5.75 (s, 1H), 5.11 (s, 1H), 4.22 (s, 2H), 3.94 (s, 1H), 3.01 (m, 3H), 2.50 (s, 1H), 2.01 (m, 14H), 1.67 (m, 3H), 1.61-1.33 (m, 11H), 1.22 (m, 5H), 1.19-0.97 (m, 12H), 0.86 (s, 3H). Example 105 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)piperidin-1-yl)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (361-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)piperidin-1-yl)-9-(((5-ethyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (361-1) The title compound 361-1 was prepared through the same synthetic route as 358-7, beginning with 358-2 and ethyl isonipecotate and 4-(bromomethyl)-5-ethyl-2H-1,3-dioxol-2-one. The crude product was purified by prep-HPLC with the following conditions: column, XBridge CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (40% to 57% over 8 min); detector, UV 254 nm. This resulted in 361-1 (3.7 mg, 14%) as an off-white solid. MS (ES, m/z): [M+1]+=752.35;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 0.92 (t, J=6.8 Hz, 1H), 1.00-1.50 (m, 29H), 1.51-1.78 (m, 3H), 1.79-2.10 (m, 6H), 2.11-2.31 (m, 4H), 2.55 (s, 1H), 2.65 (q, 7=7.5 Hz, 3H), 3.00 (d, J=13.6 Hz, 1H), 3.08-3.29 (m, 2H), 3.73 (s, 3H), 3.89 (s, 1H), 5.09 (d, J=14.0 Hz, 1H), 5.19 (d, J=14.0 Hz, 1H), 5.63 (s, 1H). Example 106 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (362-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(4-(ethoxycarbonyl)piperidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (362-1) The title compound 362-1 was prepared through the same synthetic route as 358-7, beginning with 358-2 and ethyl isonipecotate. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH OBD, 19*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (41% to 47% over 8 min); detector, UV 254 nm. This resulted in 362-1 (2.0 mg, 9%) as an off-white solid. MS (ES, m/z): [M+1]+=752.30;1H NMR (400 MHz, methanol-d4) δ 0.85 (s, 3H), 1.01-1.57 (m, 29H), 1.60-2.38 (m, 14H), 2.55 (s, 1H), 2.65-2.88 (m, 1H), 3.00 (d, J=13.6 Hz, 1H), 3.18-3.30 (m, 3H), 3.38-3.99 (m, 3H), 4.19 (d, J=6.4 Hz, 2H), 5.08 (d, J=14.0 Hz, 1H), 5.20 (d, J=14.0 Hz, 1H), 5.63 (s, 1H). Example 107 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-acetamido-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (363-5) Synthesis of benzhydryl (3aS,3bR,5aR,5bS,7aS,10S,11aR,13aR,13bS,15aS)-3a,5a,5b,7a,10,13b-hexamethyl-2,13-dioxo-2,3,3a,3b,4,5,5a,5b,6,7,7a,8,9,10,11,11a,13,13a,13b,14,15,15a-docosahydropiceno[4,3-d]oxazole-10-carboxylate (363-1) A mixture of 194-7 (1 g, 1.5 mmol), DPPA (0.7 g, 2.6 mmol, 1.7 equiv), and Et3N (0.4 g, 3.75 mmol, 2.5 equiv) in anisole (10 mL) was stirred for 1.5 h at 90° C. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with petroleum ether:ethyl acetate (1:1) to afford 363-1 (1 g, 100%) as a light yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-amino-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (363-2) A mixture of 363-1 (600 mg, 0.90 mmol) and KOH (12 mL, 1 M in 2:1 EtOH:H2O) was stirred for 1 h at 90° C. The reaction mixture was cooled and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by prep-TLC (5:1 CH2Cl2:MeOH) to afford 363-2 (400 mg, 69%) as a light yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-acetamido-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (363-3) To a stirred mixture of 363-2 (400 mg, 0.63 mmol), HATU (358 mg, 0.94 mmol, 1.5 equiv), and iPr2EtN (324 mg, 2.5 mmol, 4 equiv) in DMF was added AcOH (75.3 mg, 1.25 mmol, 2 equiv) dropwise in portions at room temperature. The reaction mixture was stirred for 3 h at room temperature. The residue was purified by prep-TLC (1:1 petroleum ether:ethyl acetate) to afford 363-3 (300 mg, 70%) as a light yellow solid. Synthesis of benzhydryl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-9-acetamido-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylate (363-4) A mixture of 363-3 (200 mg, 0.29 mmol), (5-methyl-2-oxo-2H-1,3-dioxol-4-yl) methyl 4-nitrophenyl carbonate (434 mg, 1.5 mmol, 5 equiv), and DMAP (90 mg, 0.74 mmol, 2.5 equiv) in pyridine was stirred overnight at 60° C. The reaction mixture was concentrated under reduced pressure. The residue was purified by prep-TLC (3:1 petroleum ether:ethyl acetate) to afford 363-4 (120 mg, 49%) as a light yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S, 12aS, 12bR, 14bR)-9-acetamido-2,4a,6a,6b,9,12a-hexamethyl-10-((((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid (363-5) A mixture of 363-4 (100 mg, 0.12 mmol) and TFA (0.1 mL, 1.35 mmol) in CFhCh was stirred for 1 h at room temperature. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH OBD 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (52% phase B up to 63% in 8 min); detector, UV. This resulted in 363-5 (27.7 mg, 35%) as a light yellow solid. MS (ES, m/z): [M+1]+=670.30;1H NMR (300 MHz, methanol-d4) δ 5.76-5.59 (m, 2H), 5.07-4.89 (m, 2H), 2.78 (d, J=13.6 Hz, 1H), 2.60 (d, J=10.3 Hz, 2H), 2.20 (s, 5H), 2.05-1.63 (m, 9H), 1.55 (d, J=7.9 Hz, 2H), 1.44 (d, J=8.2 Hz, 7H), 1.17 (dd, J=11.0, 6.7 Hz, 15H), 0.85 (s, 3H). Example 108 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((R)-5-methyl-2-oxooxazolidin-3-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (364-5) Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(((R)-2-hydroxypropyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (364-1) Into a 25-mL round-bottom flask was placed 357-2 (300 mg), EtOH (2.8 mL), (2R)-2-methyloxirane (31 mg, 1.3 equiv). The reaction slurry was stirred for overnight at 40° C. The reaction mixture was cooled and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1). This resulted in 140 mg of 364-1 as a yellow oil. Synthesis of 2-benzhydryl 9-methyl (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((R)-5-methyl-2-oxooxazolidin-3-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (364-2) Into a 50-mL round-bottom flask was placed 364-1 (340 mg), toluene (5 mL), CDI (187 mg, 2 equiv), and DMAP (111 mg, 2.5 equiv). The reaction slurry was stirred for 1.5 h at room temperature. The reaction mixture was diluted with water and extracted with 3×30 mL of ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 140 mg of 364-2 as a brown oil. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-4,6a,6b,8a,11,14b-hexamethyl-3-((R)-5-methyl-2-oxooxazolidin-3-yl)-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (364-3) Into a 25-mL round-bottom flask was placed 364-2 (134 mg), pyridine (1.3 mL), and LiI (235 mg, 10 equiv). The reaction slurry was stirred for overnight at 130° C. in an oil bath. The reaction mixture was cooled and extracted with 3×30 mL of CH2Cl2. The combined organic layers were washed with 2×50 ml of 1 M HCl(aq)and 3×50 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with CH2Cl2/methanol (10:1) to provide 110 mg of 364-3 as a white solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((R)-5-methyl-2-oxooxazolidin-3-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (364-4) Into a 25-mL round-bottom flask was 364-3 (60 mg), DMF (0.6 mL), KI (7 mg, 0.5 equiv), K2CO3(24 mg, 3 equiv), and 4-(chloromethyl)-5-methyl-2H-1,3-dioxol-2-one (18 mg, 1.5 equiv). The reaction slurry was stirred overnight at room temperature. The reaction mixture was diluted with 50 mL of ethyl acetate and water. The layers were separated and the aqueous was extracted with 3×30 mL of ethyl acetate. The combined organic layers were washed with 3 x 100 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 50 mg of 364-4 as a yellow solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-10-((R)-5-methyl-2-oxooxazolidin-3-yl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (364-5) Into a 25-mL round-bottom flask was placed 364-4 (60 mg), CH2Cl2(1 mL), and TFA (0.2 mL). The reaction slurry was stirred for 40 min at room temperature then concentrated. The crude product (60 mg) was purified by prep-HPLC with the following conditions: column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (65% phase B up to 70% in 8 min); Detector, UV. 10.9 mg product was obtained and concentrated. This resulted in 10.9 mg of 364-5 as a white solid. MS (ES, m/z): [M+1]+=696;1H NMR (400 MHz, methanol-d4) δ 0.83-0.87 (s, 3H), 0.87-0.94 (d, J=13.6 Hz, 1H), 1.03-1.11 (d, J=14.2 Hz, 1H), 1.14-1.23 (m, 8H), 1.24-1.27 (s, 3H), 1.27-1.37 (d, J=11.5 Hz, 3H), 1.37-1.45 (dd, J=3.9, 15.5 Hz, 6H), 1.46-1.50 (s, 3H), 1.55-1.70 (m, 1H), 1.70-1.82 (m, 1H), 1.82-1.91 (dd, J=6.5, 12.5 Hz, 2H), 1.94-2.10 (m, 1H), 2.17-2.21 (s, 3H), 2.21-2.27 (d, J=13.5 Hz, 1H), 2.59-2.63 (s, 1H), 2.86-2.94 (d, J=13.6 Hz, 1H), 3.21-3.30 (t, J=8.9 Hz, 1H), 3.74-3.83 (t, J=8.3 Hz, 1H), 4.07-4.16 (dd, J=4.1, 12.9 Hz, 1H), 4.58-4.68 (q, J=7.7 Hz, 1H), 4.69-4.77 (d, J=13.9 Hz, 1H), 5.05-5.13 (d, J=14.0 Hz, 1H), 5.61-5.65 (s, 1H). Example 109 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,5-dioxoimidazolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (365-8) Synthesis of 2-benzhydryl 9-(2-(trimethylsilyl)ethyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((tert-butoxycarbonyl)amino)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (365-1) Into a 8-mL vial was placed 280-5 (450 mg, 0.59 mmol), THF (1.5 mL), PPh3(240 mg, 0.92 mmol, 1.6 equiv), and 2-(trimethylsilyl)ethan-1-ol (0.45 mL, 3 mmol, 5 equiv) followed by DIAD (0.18 mL, 0.92 mmol, 1.6 equiv) dropwise with stirring at 40° C. The reaction slurry was stirred for 2 h at 40° C. The resulting mixture was concentrated and the residue applied onto a silica gel column with ethyl acetate:petroleum ether (1:2) to provide 400 mg of 365-1 as a white solid. Synthesis of 2-benzhydryl 9-(2-(trimethylsilyl)ethyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-amino-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (365-2) Into a 250-mL round-bottom flask was placed 365-1 (400 mg, 0.46 mmol), CH2Cl2(24 mL), and 2,6-lutidine (0.27 mL, 2.3 mmol, 5 equiv) followed by the addition of TMSOTf (0.34 mL, 1.85 mmol, 4 equiv) at 0° C. The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was washed with 2×30 ml of 0.5 M HCl(aq). The organic layer was washed with 2×30 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 300 mg of crude 365-2 as a light yellow solid. Synthesis of 2-benzhydryl 9-(2-(trimethylsilyl)ethyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-((tert-butoxycarbonyl)amino)acetamido)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (365-3) Into a 50-mL round-bottom flask was placed 365-2 (300 mg, 0.39 mmol), CH2Cl2(6 mL), Boc-glycine (205 mg, 1.17 mmol, 3 equiv), EDCI (225 mg, 1.17 mmol, 3.00 equiv), and DMAP (143 mg, 1.17 mmol, 3.00 equiv). The reaction slurry was stirred for 1 h at room temperature then concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:2) to provide 320 mg of 365-3 as a light yellow solid. Synthesis of 2-benzhydryl 9-(2-(trimethylsilyl)ethyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2-aminoacetamido)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (365-4) Into a 100-mL round-bottom flask was placed 365-3 (260 mg, 0.28 mmol), CH2Cl2(13 mL), and 2,6-lutidine (0.16 mg, 1.4 mmol, 5 equiv) followed by the addition of TMSOTf (0.20 mL, 1.1 mmol, 4 equiv) at 0° C. The reaction slurry was stirred for 1 h at room temperature. The reaction mixture was washed with 2×50 ml of 0.5 M HCl(aq)and 2×50 mL of brine, dried over anhydrous Na2SO4, filtered, and concentrated to provide 240 mg of 365-4 as a light yellow solid. Synthesis of 2-benzhydryl 9-(2-(trimethylsilyl)ethyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,5-dioxoimidazolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (365-5) Into a 100-mL round-bottom flask was placed 365-4 (288 mg, 0.35 mmol), toluene (12 mL), DMAP (86 mg, 0.7 mmol, 2 equiv), and CDI (141 mg, 0.87 mmol, 2.5 equiv). The reaction slurry was stirred overnight at 100° C. The reaction mixture was concentrated and the residue applied onto a silica gel column with ethyl acetate:petroleum ether (1:2) to provide 210 mg of 365-5 as a white solid. Synthesis of (3S,4S,4aR,6aR,6bS,8aS,11S,12aR,14aR,14bS)-11-((benzhydryloxy)carbonyl)-3-(2,5-dioxoimidazolidin-1-yl)-4,6a,6b,8a,11,14b-hexamethyl-14-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-icosahydropicene-4-carboxylic Acid (365-6) Into a 50-mL round-bottom flask was placed 365-5 (170 mg, 0.20 mmol), THF (5.5 mL), and TBAF (1.8 mL, 1 mmol, 5 equiv). The resulting solution was stirred overnight at room temperature then concentrated. The residue was applied onto a silica gel column with CH2Cl2:methanol (10:1) to provide 150 mg of 365-6 as a white solid. Synthesis of 2-benzhydryl 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,5-dioxoimidazolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (365-7) Into a 25-mL round-bottom flask, was placed 365-6 (130 mg, 0.17 mmol), DMF (3 mL), 4-(bromomethyl)-5-methyl-2/7-1,3-dioxol-2-one (50 mg, 0.26 mmol, 1.5 equiv), K2CO3(72 mg, 0.52 mmol, 3 equiv), and KI (14 mg, 0.09 mmol, 0.5 equiv). The reaction slurry was stirred for 3 h at room temperature. The resulting solution was diluted with 50 mL of ethyl acetate and 50 mL of water. The mixture was extracted with 2×50 mL of ethyl acetate. The combined organic layers were washed with 3×50 ml of brine, dried over anhydrous Na2SO4, filtered, and concentrated. The residue was applied onto a silica gel column with ethyl acetate:petroleum ether (1:1) to provide 135 mg of 365-7 as a white solid. Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-(2,5-dioxoimidazolidin-1-yl)-2,4a,6a,6b,9,12a-hexamethyl-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (365-8) Into a 8-mL vial was placed 365-7 (100 mg, 0.12 mmol), CH2Cl2(2 mL), and TFA (0.2 mL). The reaction slurry was stirred for 30 min at room temperature then concentrated. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (40% phase B up to 45% in 8 min); Detector, UV. This resulted in 54.3 mg of 365-8 as a white solid. MS (ES, m/z) [M+1]+=695;1H NMR (300 MHz, methanol-d4) δ 0.86 (s, 3H), 0.93 (d, J=12.6 Hz, 1H), 1.08 (d, J=14.2 Hz, 1H), 1.19 (d, J=8.3 Hz, 6H), 1.25 (d, J=2.3 Hz, 7H), 1.46 (d, J=17.5 Hz, 6H), 1.77 (d, J=13.2 Hz, 1H), 1.93 (q, J=14.7, 13.4 Hz, 4H), 2.21 (s, 3H), 2.25 (s, 1H), 2.62 (s, 1H), 2.90 (d, J=13.4 Hz, 1H), 3.07 (d, J=13.3 Hz, 1H), 3.87 (s, 2H), 4.28 (dd, J=13.0, 4.0 Hz, 1H), 4.75 (d, J=13.9 Hz, 1H), 5.00 (d, J=13.9 Hz, 1H), 5.63 (s, 1H). Example 110 (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((R)-4-methyl-2,5-dioxoimidazolidin-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,1142,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (366-1) Synthesis of (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,12a-hexamethyl-10-((R)-4-methyl-2,5-dioxoimidazolidin-1-yl)-9-(((5-methyl-2-oxo-1,3-dioxol-4-yl)methoxy)carbonyl)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic Acid (366-1) The title compound was prepared using the methods from the synthesis of 365-8, beginning with 365-2 and Boc-(R)-alanine. The crude product was purified by prep-HPLC with the following conditions: column, XSelect CSH OBD, 30*150 mm, 5 μm; mobile phase, water (0.05% TFA) and CH3CN (45% phase B up to 55% in 8 min); detector, UV 254 nM. This resulted in 14.1 mg (35%) of 366-1 as a white solid. MS (ES, m/z): [M+1]+=709;1H NMR (300 MHz, methanol-d4) δ 0.86 (s, 3H), 0.93 (s, 2H), 1.08 (d, J=15.2 Hz, 1H), 1.14-1.28 (m, 14H), 1.28-1.38 (m, 3H), 1.46 (d, J=17.6 Hz, 8H), 1.67 (d, J=12.9 Hz, 1H), 1.77 (d, J=13.2 Hz, 1H), 1.90 (t, J=14.4 Hz, 5H), 2.21 (d, J=1.5 Hz, 4H), 2.62 (s, 1H), 2.90 (d, J=13.0 Hz, 1H), 3.05 (d, J=14.7 Hz, 2H), 3.26 (s, 1H), 3.97 (dd, J=6.9, 4.1 Hz, 1H), 4.25 (s, 1H), 4.75 (dd, J=13.9, 6.1 Hz, 1H), 4.99 (dd, J=13.9, 9.5 Hz, 1H), 5.63 (s, 1H). Example 111 2-(3-((1-PEGSK-1H-1,2,3-triazol-4-yl)methoxy)-4-nitrobenzyl) 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (605-2) Synthesis of 4-Nitro-3-(prop-2-yn-1-yloxy)benzaldehyde (600-1) 3-Hydroxy-4-nitrobenzaldehyde (5 g, 0.0299 mol), propargyl bromide (1.05 equiv., 3.50 mL of 80% solution in toluene, 0.0314 mol) and potassium carbonate (1.3 equiv., 5.37 g, 0.0389 mol) in DMF (50 mL) were heated at 65° C. for 2 hours. The reaction was diluted with water (200 mL) and extracted with EtOAc (3×50 mL). The extract was washed with brine, dried (MgSO4) and evaporated. The residue was purified by flash column chromatography (CH2Cl2/Hexane) to give product as an off-white solid (5.83 g, 95%). Synthesis of (4-nitro-3-(prop-2-yn-1-yloxy)phenyl)methanol (600-2) Sodium borohydride (1.3 equiv., 1.72 g, 0.0454 mol) was added portionwise to 600-1 (7.17 g, 0.0349 mol) in 10% MeOH/CH2Cl2(50 mL) at 0° C. and then allowed to warm to rt. The reaction mixture was carefully quenched with 2N HCl. The organic layer was separated, washed with brine, dried (MgS04) and evaporated to give product suitable for use directly in the next step (6.51 g, 90%). Synthesis of 4-(Bromomethyl)-1-nitro-2-(prop-2-yn-1-yloxy)benzene (600-3) N-Bromosuccinimide (1.20 equiv., 4.29 g, 0.0241 mol) was added to 600-2 (4.17 g, 0.0201 mol) and triphenylphosphine (1.5 equiv., 7.91 g, 0.0302 mol) in CH2Cl2(60 mL) at 0° C. The reaction was allowed to warm to rt and stirred for 30 minutes. The mixture was evaporated and the residue purified by flash column chromatography (EtOAc/Hexane) to give product as a light yellow solid (4.23 g, 78%). Synthesis of 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) 2-(4-nitro-3-(prop-2-yn-1-yloxy)benzyl) (2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,l 1-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (605-1) 605-0 (0.075 g, 0.0903 mmol), 600-3 (0.027 g, 0.0993 mmol) and potassium carbonate (0.019 g, 0.135 mmol, 1.5 eqiuv.) in DMF (3 mL) were heated at 65° C. for 2 hours. The reaction was diluted with water (20 mL) and extracted with EtOAc. The extract was washed with brine, dried (MgSO4) and evaporated. The residue was purified by flash column chromatography (EtOAc) to give 605-1 as a white solid (90 mg, 98%). MS (ES, m/z) [M+H]+=1020. Synthesis of 2-(3-((1-PEG5K-1H-1,2,3-triazol-4-yl)methoxy)-4-nitrobenzyl) 9-((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl)(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-((2,5,8,11-tetraoxatetradecan-14-oyl)oxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylate (605-2) The addition of copper sulfate (5 mg) in water (0.6 mL) to 305-1 (0.090 g, 0.0882 mmol, 1.3 equiv), PEG5K-Azide (Average MW=5000)(0.339 g, 0.0679 mmol) and ascorbic acid (10 mg) in DMF (3 mL), followed by addition of fresh ascorbic acid (10 mg) after 1 hour and again (5 mg) at 2 hours gave 0.37 g (90%) product as a white solid.1H NMR (400 MHz, relaxation time=10 sec, DMSO-d6) δ 8.20 (s, 1H), 7.89 (d, J=8 Hz, 1H), 7.60 (s, 1H), 7.12 (d, J=8 Hz, 1H), 5.18-5.38 (m, 5H), 4.54 (t, J=8 Hz, 2H), 4.38-4.50 (m, 1H), 4.18-4.30 (m, 1H), 3.82 (t, J=8 Hz, 2H), 3.60 (m, 2H); 3.46 (s, 444H), 3.38 (s, 3H), 3.24 (s, 3H), 2.79-2.90 (m, 1H), 2.60 (s, 1H), 2.54 (q, J=6.4 Hz, 2H), 2.13-2.28 (m, 2H), 1.83-2.01 (m, 4H), 1.62-1.82 (m, 5H), 1.47-1.53 (m, 6H), 1.39-1.46 (m, 4H), 1.24-1.30 (m, 5H), 1.20 (s, 6H), 1.16 (s, 3H), 1.06 (d, j+10.4 Hz, 2H), 0.85 (s, 3H). Example 112 HSD2 Activity Human descending colon epithelial stem cells were cultured as 3D organoids in accordance with Sato et al Gastroenterology. 2011 November; 141(5): 1762-72. Organoids were dissociated using TrypLE Express (life technologies) and plated on 96-well transwells (corning) in supplemented basal media (SBM—advanced DMEM/F12 containing 10 mM HEPES, 1:100 Glutamax, 1:100 penicillin/streptomycin, 1:100N2, 1:50 B27, 1 mM N-acetylcysteine, 10 nM [Leu15]-gastrin I) containing 100 ng/mL Wnt3A (W), 50 ng/mL EGF (E), 100 ng/mL Noggin (N), 500 ng/mL RSpondinl (R), 500 nM A83-01 (S) and 2.5 uM thiazovivin (T). Cultures were differentiated using SBM containing ENRA and 30 nM aldosterone on day 3, and cultures were used for assay on day 6 or 7. Compounds were diluted in DMSO and serial dilutions prepared by titrating in DMSO. Compounds were then diluted into DMEM/F12. Transwell plates containing descending colon cultures were washed twice with DMEM/F12 and compound was added to the apical compartment. Cells were incubated with test compound for 30 minutes at 37° C., 5% CO2to equilibrate across the cell membrane. A second compound plate was prepared in which the serially diluted compounds in DMSO were diluted into DMEM/F12 containing 40 nM cortisol. Following the 30 minute pre-incubation step, the apical media was aspirated and compounds diluted in DMEM/F12 with 40 nM cortisol were added to the apical side of the transwell. The plate was then incubated for four hours at 37° C., 5% CO2. Cortisol levels were measured using a cortisol HTRF assay kit as described by the manufacturer (Cisbio). Concentration-response curves were then plotted and IC50(and pIC50) values were determined using least squares non-linear regression. Glycyrrhetinic acid had a pIC50of 6.6. Inhibition of HSD2 activity as measured by the inhibition of the conversion of cortisone to cortisol. Compounds of the invention demonstrated greater HSD2 inhibition compared to the corresponding acid metabolite (chemically named). CompoundpIC50(2S,4aS,6aS,6bR,8aS,10S,12aS,12bR,14bR)-2,4a,6a,6b,9,9,12a-5.5heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,10-dicarboxylic acid178-16.3176-26.3700-16.1 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-5.42,4a,6a,6b,9,12a-hexamethyl-10-{[2-(methylsulfanyl)acetyl]oxy}-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid194-108195-26.3701-17 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-[(2-<5methanesulfonylacetyl)oxy]-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid195-26.3 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-6.6(benzoyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid197-27.9 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-5.8(cyclopropanecarbonyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid198-27.7 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-5.1(methoxymethoxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid205-26.4258-26.9 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-5.9(acetyloxy)-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid209-3702-16.4703-16.1704-16.3316-17.5705-16.6706-16.7707-17.1209-37.2243-17.5 CompoundpIC50(2S,4aS,6aS,6bR,9R,10S,12aS,12bR,14bR)-9-<5[(carboxymethoxy)methyl]-10-hydroxy-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid240-85 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-[(2-5.9hydroxyacetyl)oxy]-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid244-16.8 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-[(2-<5methoxyacetyl)oxy]-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid708-16.3709-17.2 CompoundpIC50(2S,4aS,6aS,6bR,8aR,10S,12aS,12bR,14bR)-10-5.4(carboxymethoxy)-2,4a,6a,6b,9,9,12a-heptamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2-carboxylic acid710-16711-15.8712-15 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-10-<5[(methoxycarbonyl)amino]-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid281-36.7 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-<52,4a,6a,6b,9,12a-hexamethyl-10-({[2-(morpholin-4-yl)ethyl]carbamoyl}oxy)-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid286-46.9 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-<510-{[2-(dimethylamino)acetyl]oxy}-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylicacid290-26.6 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-5.610-[4-(ethoxycarbonyl)-5-methoxy-1H-pyrazol-1-yl]-2,4a,6a,6b,9,12a-hexamethyl-13-oxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid713-16.9 CompoundpIC50(2S,4aS,6aS,6bR,8aR,9S,10S,12aS,12bR,14bR)-<52,4a,6a,6b,9,12a-hexamethyl-13-oxo-10-(2,5,8,11-tetraoxatetradecanoyloxy)-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,12b,13,14b-icosahydropicene-2,9-dicarboxylic acid605-26.9 Example 113 Stability Assays Sample were analyzed on an Agilent 6410 triple-quadrapole LC-MS system consisting of an Agilent 1260 LC with a Phenomenex Gemini 5 μm column (NX-C18, 110A, 30×2 mm) and the mass spectrometer with an electrospray interface running under a positive ionization mode. Mobile phases were 0.1% formic acid in water and 0.1% formic acid in acetonitrile. Plasma Stability—Plasma from pooled male rat or human (purchased from BioreclamationIVT, LLC) were pre-warmed to 37° C. Compounds were then added to the plasma samples to make a final concentration of 1 μM and vortexed. Duplicate samples of 100 μL each were taken out at Time 0, 10, 20, 30 and 60 min for extraction and analysis. Extraction and analysis of parent drug were by addition of 300 μL of acetonitrile containing 500 ng/mL of internal standard (labetalol), vortexing, and centrifugation. 150 μL of the supernatant was added to 100 μL of deionized water and 10 μL injected onto LC/MS. Liver S9 Homogenate Stability—Liver S9 homogenate from pooled male rat or human (purchased from Xenotech, LLC, 20 mg protein/mL) was diluted with 0.05M KH2PO4, pH 7.4 buffer to make 0.8 mg protein/mL and pre-warmed to 37° C. Compounds were then added to the homogenate samples to make a final concentration of 1 μM and vortexed. Duplicate samples of 100 μL each were taken out at Time 0, 5, 15, 30 and 120 min for extraction and analysis. Extraction and analysis of parent drug were by addition of 300 μL of acetonitrile containing 100 ng/mL of internal standard (labetalol), vortexing, and centrifugation. 150 μL of the supernatant was added to 100 μL of deionized water and 10 μL injected onto LC/MS. Liver Microsomal Stability—Liver microsome from pooled male rat or human (purchased from Xenotech, LLC, 20 mg protein/mL) was diluted with 0.05M KH2PO4, pH 7.4 buffer containing 5 mM MgCh to make 0.5 mg protein/mL and pre-warmed to 37° C. Compounds were then added to the homogenate samples to make a final concentration of 1 μM and vortexed. NADPH in 0.05M KH2PO4, pH 7.4 buffer was then added to make the final concentration of 2 mM to start the reaction. Duplicate samples of 100 μL each were taken out at Time 0, 3, 6, 10, 15, 20 and 30 min for extraction and analysis. Extraction and analysis of parent drug were by addition of 100 μL of acetonitrile containing 100 ng/mL of internal standard (labetalol), vortexing, and centrifugation. 10 μL of the supernatant was injected onto LC/MS. An incubation without the NADPH addition was used as a control for the experiment. Cecal-Colonic Extract Stability—Female rats (non-fasted) were euthanized and the cecum and colon taken out and weighed. The intestinal contents in the cecum and colon were flushed out with 20 mL deionized water and the tissues re-weighed. Deionized water was added to the cecal-colonic content mixture to make a 10 X w/v dilution. The mixture was then homogenized by a Polytron homogenizer for 2 minutes and centrifuged at 5000 rpm in a Beckman Allegra 25r centrifuge for 10 minutes. The supernatant was taken out and warmed to 37° C. in a shaking water. 1.5 mL Aliquots were then added compounds to make a final concentration of 1 μM and vortexed. Duplicate samples of 100 μL each were taken out at Time 0, 10, 20, 40, 60 and 180 min for extraction and analysis. Extraction and analysis of parent drug were by addition of 300 μL of acetonitrile containing 500 ng/mL of internal standard (labetalol), vortexing, and centrifugation. 150 μL of the supernatant was added to 100 μL of deionized water and 10 μL injected onto LC/MS.
491,725
11858960
DETAILED DESCRIPTION TO THE INVENTION For purposes of diagnostic or molecular imaging in vivo as well as for therapeutic purposes, the imaging agent or therapeutic agent must be able to arrive at its target with high efficiency. This requires a combination of small-enough size in order to be able to achieve sufficient tissue penetration, selective binding to the target in order to achieve a high signal/noise ratio at the target site (applies especially to imaging agent but likewise contributes to the specificity of a therapeutic agent), and low overall body retention or accumulation (as a consequence of elimination from the body; typically in liver or kidneys; and all the more problematic with small-sized imaging agent) to avoid sites of high background signal which negatively influence signals at the target site (imaging agents) or to avoid potential unwanted side effects (therapeutic agents). In work leading to the current invention, first of all a number of immunoglobulin single variable domain (ISVD) molecules, herein also referred to a single domain antibodies (sdAb), binding with high specificity to human PD-L1 (huPDL1) were identified. Surprisingly, and in contrast to similar ISVD molecules binding to murine (but not human) PD-L1, in contrast to other control ISVD molecules, and in contrast to other small-molecule PD-L1 imaging agents, the huPDL1-binding ISVDs were characterized by an extremely low renal retention whilst providing an excellent target signal/background ratio in vivo. Anti-PD-L1 sdAbs were identified after screening of alpaca immune libraries and were evaluated for binding and affinity using enzyme-linked immunosorbent assay (ELISA), flow cytometry and Surface Plasmon Resonance (SPR). Single photon emission computed tomography imaging in mice following intravenous injection of Technetium-99m (99mTc)-labelled anti-PD-L1 sdAb revealed that this sdAb has several properties to make it an interesting diagnostic, including (i) high signal to noise ratio's; (ii) strong ability to specifically detect human PD-L1 in melanoma and breast tumours, and (iii) relatively low kidney retention, which is unique as typically radiometal-labelled sdAbs show high retention in the proximal tubuli. Moreover, we showed using SPR that the anti-PD-L1 sdAb binds to the same epitope on PD-L1 as the FDA-approved mAb avelumab, and that the anti-PD-L1 sdAb efficiently antagonizes the PD-1:PD-L1 interaction. Different in vitro human cell-based assays corroborated the PD-1:PD-L1 blocking activity, showing enhanced antigen-specific T-cell receptor signalling and tumour cell killing ability of PD-1-expressing T cells interacting with PD-L1-positive tumour cells; as well as showing enhancement of the capacity of dendritic cells (DCs) to stimulate T-cell activation and cytokine production, opening an avenue to include these anti-PD-L1 ISVDs in DC-vaccination protocols. These combined characteristics render the identified huPDL1-binding ISVDs extremely well-suited as diagnostic agent, e.g. for molecular imaging, besides being useful in therapies implying immune checkpoint inhibitors. Based hereon, the invention is defined in the following aspects and embodiments, and described in more detail hereafter. As the invention relates to polypeptides comprising complementarity determining regions (CDRs), some explanation is first provided on how such CDRs are determined. The determination of the CDR regions in an antibody/immunoglobulin sequence generally depends on the algorithm/methodology applied (Kabat-, Chothia-, Martin (enhanced Chothia), IMGT (ImMunoGeneTics information system)-numbering schemes; see, e.g. http://www.bioinf.org.Uk/abs/index.html#kabatnum and http://www.imgt.org/IMGTScientificChart/Numbering/IMGTnumbering.html). Applying different methods to the same antibody/immunoglobulin sequence may give rise to different CDR amino acid sequences wherein the differences may reside in CDR sequence length and/or—delineation within the antibody/immunoglobulin/IVD sequence. The CDRs of the huPDL1-binding polypeptides of the invention can therefore be described as the CDR sequences as present in the single variable domain anti-human PD-L1 antibodies characterized herein, or alternatively as determined or delineated according to a well-known methodology such as according to the Kabat-, Chothia-, Martin (enhanced Chothia), or IMGT-numbering scheme or -method. The CDR sequences defined in SEQ ID NOs: 2-4, 6-7, 9-10, and 12, for instance, have, been delineated from the anti-human PD-L1 single domain antibodies defined by SEQ ID NOs: 1, 5, 8 and 11 by means of the IMGT-method (seeFIG.1). Applying another method may result in CDR sequences (slightly) different from those defined in SEQ ID NOs: 2-4, 6-7, 9-10, and 12. In a first aspect, the invention relates to polypeptides specifically binding to human PD-L1, wherein the amino acid sequence of the polypeptide is comprising a CDR1 region, a CDR2 region, and a CDR3 region, wherein the CDR1, CDR2 and CDR3 regions are selected from those CDR1, CDR2 and CDR3 regions, respectively, as present in any of huPDL1-binding single domain antibodies defined by SEQ ID Nos:1, 5, 8 or 11. In particular, the polypeptides specifically binding to human PD-L1 comprise an immunoglobulin variable domain (IVD) conveying specificity of the polypeptide for binding to human PD-L1 wherein the IVD is comprising a CDR1 region, a CDR2 region, and a CDR3 region, wherein the CDR1, CDR2 and CDR3 regions are selected from those CDR1, CDR2 and CDR3 regions, respectively, as present in any of huPDL1-binding single domain antibodies defined by SEQ ID Nos:1, 5, 8 or 11 (seeFIG.1). In an embodiment thereto, the CDR regions are determined by applying the Kabat, Chothia, Martin, or IMTG method to SEQ ID Nos:1, 5, 8 or 11. In a more specific embodiment, the CDR regions are determined by the IMTG method and further defined as a CDR1 region chosen from SEQ ID Nos: 2 and 6, a CDR2 region chosen from SEQ ID Nos: 3, 7, 9, or 22, and a CDR3 region chosen from SEQ ID Nos:4, 10, or 12. Given the high degree of similarity between individual CDR1 amino acid sequences, between individual CDR2 amino acid sequences, and between individual CDR3 amino acid sequences, any huPDL1-binding polypeptide comprising any possible combination of CDR1-CDR2-CDR3 amino acid sequences (determined with any of the above-described methods) is herewith envisaged (e.g. for the IMTG-delineated CDRs: CDR1-CDR2-CDR3 with respectively SEQ ID Nos:2-3-4, or 6-7-4, or 2-9-10, or 6-3-12, or 2-7-4, or 2-9-12, or 6-9-10, and so on, to list only a few). In one embodiment, the CDR regions are the CDR regions as present in SEQ ID NO:5, or, alternatively, as defined by IMTG as SEQ ID Nos:6, 7, and 4 for CDR1, CDR2, and CDR3, respectively. In any of the above, the CDR regions and/or the IVD may be humanized. Humanized CDRs and/or IVDs can be obtained in any suitable manner known and thus are not strictly limited to polypeptides that have been obtained using a polypeptide that comprises a naturally occurring VHH domain as starting material. Humanized immunoglobulin single variable domains, may have several advantages, such as a reduced immunogenicity, compared to the corresponding naturally occurring VHH domains. Such humanization generally involves replacing one or more amino acid residues in the sequence of a naturally occurring CDR and/or framework region (FR) with the amino acid residues that occur at the same position in a human VH domain, such as a human VH3 domain. The humanizing substitutions should be chosen such that the resulting humanized immunoglobulin domains still retain the favourable properties of the originator immunoglobulin (or further improved by e.g. affinity maturation). The skilled person will be able to select humanizing substitutions or suitable combinations of humanizing substitutions, which optimize or achieve a suitable balance between the favourable properties provided by the humanizing substitutions on the one hand and the favourable properties of naturally occurring VHH domains on the other hand. In general, the specificity of binding to the target is not significantly (negatively) affected in a humanized antibody/immunoglobulin/IVD (or polypeptide comprising such antibody/immunoglobulin/IVD) and, in general, the affinity and/or avidity of binding to the target is not significantly (negatively) affected in a humanized antibody/immunoglobulin/IVD (or polypeptide comprising such antibody/immunoglobulin/IVD). The huPDL1-binding polypeptides of the invention may comprise (in a fusion, conjugated therewith, or complexed therewith), one or more non-(poly)peptidic constituents (such as detectable moieties—see further; or such as pegylation—see e.g. WO2017/059397), one or more further polypeptide(s) or polypeptide domain(s) (such as e.g. a His-tag, or sortag motif, i.e., sortase amino acid substrate motif LPXTG (SEQ ID NO:17), e.g. LPETG (SEQ ID NO:18)), referred to herein as “functional moiety”. In one instance, the huPDL1-binding polypeptide itself may be duplicated or multiplicated (wherein the monomers are e.g. connected through a flexible linker such as a linker based on Gly-Pro repeats, Pro-Ala repeats, Gly-Ser repeats, or combinations thereof) to form a multivalent (though monospecific) binding molecule. In another instance, the further polypeptide or polypeptide domain (which may be connected through a flexible linker such as a linker based on Gly-Pro repeats, Pro-Ala repeats, Gly-Ser repeats, or combinations thereof, to the huPDL1-binding polypeptide) may confer binding to an entity different from huPDL1, may exert an enzymatic function (such as for, but not limited to, ADEPT (antibody-directed enzyme prodrug therapy)), may exert a toxic function (such as for, but not limited to, ADC (antibody-drug conjugates)), may confer a fluorescent signalling function to the combined polypeptide (e.g. fluorescent protein), may confer increased serum half-life (e.g. a serum albumin binding protein or peptide; less desired for imaging purposes but desired for therapeutic purposes), or may confer an additional therapeutic function. Clearly, any of these can be combined in any way in a huPDL1-binding polypeptide of the invention. Thus, in any of the above, the huPDL-1 binding polypeptide may further comprise a functional moiety. In one embodiment, the functional moiety is a detectable moiety. HuPDL1-binding polypeptides as defined herein and carrying a detectable moiety therewith may be immunotracers; in case the detectable moiety is a radiolabel, the huPDL-1 binding polypeptides may be radioimmunotracers. A “detectable moiety” in general refers to a moiety that emits a signal or is capable of emitting a signal upon adequate stimulation, and is detectable by any means, preferably by a non-invasive means, once inside the human body. Furthermore, the detectable moiety may allow for computerized composition of an image, as such the detectable moiety may be called an imaging agent. Detectable moieties include fluorescence emitters, positron emitters, radioemitters, etc. Measuring the amount of detectable moiety/imaging agent (comprised in, carried by, coupled to, chelated on a huPDL1-binding polypeptide) is typically done with a device counting radioactivity or determining radiation (which can be of photonic nature) density or radiation concentration. The counted or determined radioactivity can be transformed into an image. Depending on the nature of the emission by the detectable moiety, it may be detectable by techniques such as PET (positron emission tomography), SPECT (single-photon emission computed tomography), fluorescence imaging, fluorescence tomography, near infrared imaging, near infrared tomography, optical tomography, etc. Examples of radioemitters/radiolabels include68Ga,110mIn,18F,45Ti,44Sc,47Sc,61Cu,60Cu,62Cu,66Ga,64Cu,55Ca,72As,86Y,90Y,89Zr,125I,74Br,75Br,76Br,77Br,78Br,111In,114mIn,114In,99mTc,11C,32Cl,34Cl,123I,124I,131I,186Re,188Re,177Lu,99Tc,212Bi,213Bi,212Pb,225Ac,153Sm, and67Ga. Fluorescence emitters include cyanine dyes (e.g. Cy5, Cy5.5, Cy7, Cy7.5), indolenine-based dyes, benzoindolenine-based dyes, phenoxazines, BODIPY dyes, rhodamines, Si-rhodamines, Alexa dyes, and derivatives of any thereof. Many of the radionuclides have a metallic nature and are typically incapable of forming stable covalent bonds with proteins or peptides. One solution is to label proteins or peptides with radioactive metals by means of chelators, i.e. multidentate ligands, which form non-covalent compounds, called chelates, with the metal ions. A huPDL1 binding polypeptide may thus be coupled in any way to such chelator, which enables incorporation of a radionuclide; this allows a radionuclide to be coordinated, chelated or complexed to the huPDL1-binding polypeptide. Chelators include polyaminopolycarboxylate-type chelators which can be macrocyclic or acyclic. A polyaminopolycarboxylate chelator can be conjugated to a huPDL1-binding polypeptide e.g. via a thiol group of a cysteine residue or via an epsilon amine group of a lysine residue. Macrocyclic chelators for radioisotopes such as indium, gallium, yttrium, bismuth, radioactinides and radiolanthanides include DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) and derivatives thereof such as maleimidomonoamide-DOTA (1,4,7,10-tetraazacyclododecane-1,4,7-tris-acetic acid-10-maleimidoethylacetamide), DOTAGA (2,2′,2″-(10-(2,6-dioxotetrahydro-2H-pyran-3-yl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl)triacetic acid) with said polypeptide. Other chelators include NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid), and derivatives thereof such as NODAGA (2,2′-(7-(1-carboxy-4-((2,5-dioxopyrrolidin-1-yl)oxy)-4-oxobutyl)-1,4,7-triazonane-1,4-diyl)diacetic acid). Acyclic polyaminopolycarboxylate chelators include different derivatives of DTPA (diethylenetriamine-pentaacetic acid). Further chelating agents include DFO, CB-DO2A, 3p-C-DEPA, TCMC, Oxo-DO3A, TE2A, CB-TE2A, CB-TE1A1P, CB-TE2P, MM-TE2A, DM-TE2A, diamsar, NODASA, NETA, TACN-TM, 1B4M-DTPA, CHX-A″-DTPA, TRAP, NOPO, AAZTA, DATA, H2dedpa, H4octapa, H2azapa, H5decapa, H6phospa, HBED, SHBED, BPCA, CP256, PCTA, HEHA, PEPA, EDTA, TETA, and TRITA. The detectable moiety in a huPDL1-binding polypeptide, may itself be comprised in a prosthetic group and the prosthetic group may be linked to the polypeptide through a chelator or conjugating moiety such as a cyclooctyne comprising a reactive group that forms a covalent bond with an amine, carboxyl, carbonyl or thiol functional group on the huPDL1-binding polypeptide. Cyclooctynes include dibenzocyclooctyne (DIBO), biarylazacyclooctynone (BARAC), dimethoxyazacyclooctyne (DIMAC) and dibenzocyclooctyne (DBCO), DBCO-PEG4-NHS-Ester, DBCO-Sulfo-NHS-Ester, DBCO-PEG4-Acid, DBCO-PEG4-Amine or DBCO-PEG4-Maleimide. An example of an18F-labelled prosthetic group is18F-3-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethoxy)-2-fluoropyridine (18F-FFPEGA). Other18F-labelled prosthetic groups include N-Succinimidyl-4-[18F]fluorobenzoate ([18F]SFB) (e.g. Li et al. 2014, Applied Radiation and Isotopes 94:113-117); I-labelled prosthetic groups include N-succinimidyl 4-guanidinomethyl-3-[(*)I]iodobenzoate ([(*)I]SGMIB) and N-succinimidyl 3-guanidinomethyl-5-[(*)I]iodobenzoate (iso-[(*)I]SGMIB) wherein (*)I is for instance 131I (see e.g. Choi et al. 2014, Nucl Med Biol 41:802-812). Conjugation methods as described above may result in heterogeneous tracer populations. Site-specific conjugation strategies try to overcome this shortcoming and include chemoenzymatic methods to couple polypeptides such as antibodies/immunoglobulins/IVDs with a chelator or detectable moiety such as via sortase-mediated transpeptidation (Antos et al. 2009, Curr Protoc Protein Sci, Chapter 15:unti-15.3) (reviewed by e.g. Massa et al. 2016, Exp Opin Drug Deliv 13:1149-1163). Other aspects relate to isolated nucleic acids encoding a huPDL1-binding polypeptide as described hereinabove; to vectors comprising such nucleic acid; and to host cells comprising such nucleic acid or vector, and/or expressing huPDL1-binding polypeptide as described hereinabove. A further aspect relates to pharmaceutical compositions comprising a huPDL1-binding polypeptide as described hereinabove (huPDL1-binding polypeptides without/not comprising a functional moiety, huPDL1-binding polypeptides with/comprising a functional moiety, or huPDL1-binding polypeptides with/comprising a detectable moiety). Yet a further aspect relates to huPDL-1 binding polypeptide as described hereinabove, or to a pharmaceutical composition comprising it, for use as a medicament, for use in diagnosis, for use in surgery, for use in treatment, for use in therapy monitoring, for use in dendritic cell vaccination, and in particular for use as an imaging agent. Diagnosis In general “diagnosis” herein refers to detection of human PD-L1. This can be ex vivo or in vitro such as in a sample from a human subject (and such as by for instance ELISA, immunocytochemistry (ICH), western blot, or surface Plasmon resonance). This can also be in vivo diagnosis, in particular non-invasive in vivo diagnosis such as by medical imaging or molecular imaging as described hereinabove. Diagnosis, whether on a sample from a human subject or by in vivo (imaging) methods allows to identify patients eligible to treatment with a PD1- or PDL-1-based immune checkpoint inhibitor (such as with huPDL1-binding polypeptides of the current invention without/not comprising a detectable moiety), therewith avoiding non-effective treatment and saving on payer's budgets, and/or to monitor the effect of such immune checkpoint therapy and to monitor whether, at any time, such immune checkpoint therapy is expected to still be effective. Diagnosis, and especially imaging, may also assist in defining e.g. a tumour in need of surgical resection, thus in assisting surgery. Treatment “Treatment”/“treating” refers to any rate of reduction, delaying or retardation of the progress of the disease or disorder, or a single symptom thereof, compared to the progress or expected progress of the disease or disorder, or single symptom thereof, when left untreated. More desirable, the treatment results in no/zero progress of the disease or disorder, or single symptom thereof (i.e. “inhibition” or “inhibition of progression”), or even in any rate of regression of the already developed disease or disorder, or single symptom thereof. “Suppression/suppressing” can in this context be used as alternative for “treatment/treating”. Treatment/treating also refers to achieving a significant amelioration of one or more clinical symptoms associated with a disease or disorder, or of any single symptom thereof. Depending on the situation, the significant amelioration may be scored quantitatively or qualitatively. Qualitative criteria may e.g. by patient well-being or quality of life. In the case of quantitative evaluation, the significant amelioration is typically a 10% or more, a 20% or more, a 25% or more, a 30% or more, a 40% or more, a 50% or more, a 60% or more, a 70% or more, a 75% or more, a 80% or more, a 95% or more, or a 100% improvement over the situation prior to treatment. The time-frame over which the improvement is evaluated will depend on the type of criteria/disease observed and can be determined by the person skilled in the art. Treatment also refers to prevention of disease relapse. Relapse in this context refers to the return of a disease or the signs and symptoms of a disease after a period of improvement. In particular herein, treatment is meant to be a treatment including a PD1- or PDL1-based immune checkpoint inhibitor (such as with huPDL1-binding polypeptides of the current invention without/not comprising a detectable moiety). Such treatment including a PD1- or PDL-1-based immune checkpoint inhibitor may also be combined with other complementary forms of treatment, such as surgery, chemotherapy, radiotherapy, oncolytic viruses, blocking of immune checkpoints other than PD1 or PDL-1, adoptive transfer of natural or engineered immune cells (such as the so-called chimeric antigen receptor T cells or CAR-T cells) and/or vaccination using proteins, nucleic acids or cells (such as dendritic cells). Alternatively, the anti-PD-L1 IVDS as described herein can be combined in any way (in the same or in separate (pharmaceutical) compositions; concurrent or sequentially in any order; in one or more or multiple doses or administrations) with one or more other immunotherapeutic or immunogenic agents. Therapy Monitoring The FDA has approval anti-PD-1 mAbs pembrolizumab and nivolumab, and anti-PD-L1 mAbs durvalumab, atezolizumab and avelumab, which have since become available as standard-of-care for several cancer types. The downside of this success story is the high cost of such treatments, easily surpassing $100,000 per patient (Aguiar et al. 2017, Ann Oncol 28:2256-2263), and the observation that these immune checkpoint blockers are only of benefit for a subset of patients (Alsaab et al. 2017, Front Pharmacol 8:561). The failure rate, combined with the high cost for society, drives the search for predictive biomarkers that can help select the right treatment for the right patient. Currently the most commonly used predictive biomarker is PD-L1 expression assessed via IHC on tumor biopsies, although limitations are obviously present. Limitations such as heterogeneous expression, the role of expression outside of the tumor, and its dynamic expression during the disease process could be overcome by noninvasive molecular imaging using radiolabeled tracers that allow deep tumor penetration and repeated quantification of PD-1 and/or PD-L1 expression, which should enable mapping of primary tumors and metastatic lesions both before and during the treatment. Data generated by England et al. 2018 (Eur J Nucl Med Mol Imaging 45:110-120) show that PD-1-targeted tumor imaging in vivo can assist in disease diagnostics, patient stratification (determining which patients are more likely to respond to immunotherapy), disease monitoring (changes in the tumor images obtained during therapy reflect response or non-response to immunotherapy) and the design and development of new immunotherapies (throughout pre-clinical or clinical development). In particular, imaging (such as immunoPET imaging) of cancer and immune cells based on labeled anti-PL1 moieties of the current invention can likewise assist in monitoring the efficacy of immunotherapy or immunogenic therapy, while also assisting in patient stratification and providing valuable information when designing and/or developing new immunotherapies or immunogenic therapies. Dendritic Cells (DC) and DC Vaccination Dendritic cell [DC] vaccines can induce durable clinical responses, at least in a fraction of previously treated, late-stage cancer patients. Several preclinical studies suggest that shielding programmed death-ligand 1 [PD-L1] on the DC surface may be an attractive strategy to extend such clinical benefits to a larger patient population. Dendritic cell [DC] vaccination is therefore extensively studied as a strategy to activate cancer-specific cytotoxic T lymphocytes [CTLs]. To induce potent antitumour CTLs three requirements need to be fulfilled: first, the peptide/MHC-I complex on the surface of DCs must be correctly recognized by the T-cell receptor [TCR] expressed on CD8posT cells. Second, co-stimulatory molecules, like CD80 and CD86, expressed on DCs, need to bind with co-stimulatory receptors, like CD28, expressed on CD8posT cells. Finally, a third signal is provided by DCs under the form of cytokine secretion. Only, when those requirements are fulfilled, activated and effective T cells will be able to attack tumour cells (Santos & Butterfield 2018, J Immunol 200:443-449). DCs also express inhibitory molecules, like programmed death-ligand 1 [PD-L1], which binds to its receptor programmed death-1 [PD-1] on activated CTLs, and acts as a brake on T-cell activation (Liechtenstein et al. 2012, J Clin Cell Immunol S12). Interaction of PD-L1 with PD-1 during antigen presentation results in TCR down-modulation (Karwacz et al. 2011, EMBO Mol Med 3:581-592; Yokosuka et al. 2012, J Exp Med 209:1201-1217). As a consequence TCR-signalling is down-regulated as well, preventing T-cell hyper activation (Boding et al. 2009, J Immunol 183:4994-5005). However, in the case of vaccination in the context of cancer as wells as of infectious diseases (see, e.g., Qu et al. 2014, Int J Infect Dis 19:1-5 “Monocyte-derived dendritic cells: targets as potent antigen-presenting cells for the design of vaccines against infectious diseases”), hyperactivation of T-cells is warranted. Several strategies have been successfully employed to interfere with PD-L1:PD-1 interactions during antigen presentation by DCs to CD8posT cells. These include silencing of PD-L1 (Karwacz et al. 2011, EMBO Mol Med 3:581-592; Hobo et al. 2010, Blood 116:4501-4511), use of soluble PD-1 or PD-L1 (He et al. 2005, Anticancer Res 25:3309-3313; Pen et al. 2014, Gene Ther 21:3309-3313) and use of antibodies (Karwacz et al. 2011, EMBO Mol Med 3:581-592; Ge et al. 2013, Cancer Lett 336:253-259; Lichtenegger et al. 2018, Front Immunol 9:385). Reported in the Examples herein is the development of a single domain antibody [sdAb] that binds human PD-L1 with high affinity on the same epitope as the monoclonal antibody [mAb] avelumab. This sdAb was demonstrated in the Examples hereinto have high potential for imaging of PD-L1 expressed on tumour cells. It was further established that the sdAb blocks the interaction between PD-1 and PD-L1 on the protein level, and that this blocking ability facilitates killing of tumour cells by cytolytic immune cells present in peripheral blood mononuclear cells [PBMCs]. As sdAbs are versatile antigen binding moieties, further studies pointed to the applicability of the sdAb to enhance the activation of tumour antigen-specific CD8posT cells by monocyte-derived DCs [moDCs]. In particular, a high affinity, antagonistic, PD-L1-specific sdAb (single domain antibody) was evaluated for its ability to enhance DC-mediated T-cell activation, and benchmarked against the use of the monoclonal antibodies [mAbs], MIH1, 29E.2A3 and avelumab. Similar to mAbs, the sdAb enhanced antigen-specific T-cell receptor signaling in PD-1posreporter cells activated by DCs. It was further shown that the activation and function of antigen-specific CD8posT cells, activated by DCs, was enhanced by inclusion of an sdAb, but not mAbs. This was most pronounced when less mature DCs were used for T-cell activation. The failure of mAbs to enhance T-cell activation might be explained by their low efficacy to bind PD-L1 on DCs when compared to binding of PD-L1 on non-immune cells and binding of PD-L1 by an sdAb. These data provide a rationale for the inclusion of anti-PD-L1 sdAb in DC-based immunotherapy strategies (such as for treating or inhibiting cancer or infectious diseases). Immunotherapy and Immunogenic Therapy Immunotherapy in general is defined as a treatment that uses the body's own immune system to help fight a disease, more specifically cancer in the context of the current invention. Immunotherapeutic treatment as used herein refers to the reactivation and/or stimulation and/or reconstitution of the immune response of a mammal towards a condition such as a tumour, cancer or neoplasm evading and/or escaping and/or suppressing normal immune surveillance. The reactivation and/or stimulation and/or reconstitution of the immune response of a mammal in turn in part results in an increase in elimination of tumorous, cancerous or neoplastic cells by the mammal's immune system (anticancer, antitumour or anti-neoplasm immune response; adaptive immune response to the tumour, cancer or neoplasm). Immunotherapeutic agents of particular interest include immune checkpoint inhibitors (such as anti-PD-1, anti-PD-L1 or anti-CTLA-4 antibodies), bispecific antibodies bridging a cancer cell and an immune cell, dendritic cell vaccines, Immunotherapy is a promising new area of cancer therapeutics and several immunotherapies are being evaluated pre-clinically as well as in clinical trials and have demonstrated promising activity (Callahan et al. 2013, J Leukoc Biol 94:41-53; Page et al. 2014, Annu Rev Med 65:185-202). However, not all the patients are sensitive to immune checkpoint blockade and sometimes PD-1 or PD-L1 blocking antibodies accelerate tumour progression. An overview of clinical developments in the field of immune checkpoint therapy is given by Fan et al. 2019 (Oncology Reports 41:3-14). Monoclonal antibodies targeting and inhibiting PD-1 include pembrolizumab, nivolumab, and cemiplimab. Monoclonal antibodies targeting and inhibiting PD-L1 include atezolizumab, avelumab, and durvalumab. Monoclonal antibodies targeting and inhibiting CTLA-4 include ipilimumab. Combinatorial cancer treatments that include chemotherapies can achieve higher rates of disease control by impinging on distinct elements of tumour biology to obtain synergistic antitumour effects. It is now accepted that certain chemotherapies can increase tumour immunity by inducing immunogenic cell death and by promoting escape in cancer immunoediting, such therapies are therefore called immunogenic therapies as they provoke an immunogenic response. Drug moieties known to induce immunogenic cell death include bleomycin, bortezomib, cyclophosphamide, doxorubicin, epirubicin, idarubicin, mafosfamide, mitoxantrone, oxaliplatin, and patupilone (Bezu et al. 2015, Front Immunol 6:187). Other forms of immunotherapy include chimeric antigen receptor (CAR) T-cell therapy in which allogeneic T-cells are adapted to recognize a tumour neo-antigen and oncolytic viruses preferentially infecting and killing cancer cells. Treatment with RNA, e.g. encoding MLKL, is a further means of provoking an immunogenic response (Van Hoecke et al. 2018, Nat Commun 9:3417), as well as vaccination with neo-epitopes (Brennick et al. 2017, Immunotherapy 9:361-371). In a final aspect, the invention relates to methods for producing a huPDL1-binding polypeptide according to the invention, such methods comprising the steps of:expressing the huPDL1-binding polypeptide in a suitable host cell (such as comprising a nucleic acid or vector as described herein; andpurifying the expressed huPDL1-binding polypeptide. Such methods may further comprise a step of coupling, incorporating, binding, ligating, bonding, complexing, chelating, conjugating (e.g. site-specifically conjugating) or otherwise linking, covalently or non-covalently, a detectable moiety to the purified huPDL1-binding polypeptide. Other Definitions The present invention is described with respect to particular embodiments and with reference to certain drawings but the invention is not limited thereto but only by the claims. Any reference signs in the claims shall not be construed as limiting the scope. The drawings described are only schematic and are non-limiting. In the drawings, the size of some of the elements may be exaggerated and not drawn on scale for illustrative purposes. Where the term “comprising” is used in the present description and claims, it does not exclude other elements or steps. Where an indefinite or definite article is used when referring to a singular noun e.g. “a” or “an”, “the”, this includes a plural of that noun unless something else is specifically stated. Furthermore, the terms first, second, third and the like in the description and in the claims, are used for distinguishing between similar elements and not necessarily for describing a sequential or chronological order. It is to be understood that the terms so used are interchangeable under appropriate circumstances and that the embodiments of the invention described herein are capable of operation in other sequences than described or illustrated herein. Unless specifically defined herein, all terms used herein have the same meaning as they would to one skilled in the art of the present invention. Practitioners are particularly directed to Sambrook et al., Molecular Cloning: A Laboratory Manual, 4th ed., Cold Spring Harbor Press, Plainsview, N.Y. (2012); and Ausubel et al., current Protocols in Molecular Biology (Supplement 100), John Wiley & Sons, New York (2012), for definitions and terms of the art. The definitions provided herein should not be construed to have a scope less than understood by a person of ordinary skill in the art. The term “defined by SEQ ID NO:X” as used herein refers to a biological sequence consisting of the sequence of amino acids or nucleotides given in the SEQ ID NO:X. For instance, a CDR defined in/by SEQ ID NO:X consists of the amino acid sequence given in SEQ ID NO:X. A further example is an amino acid sequence comprising SEQ ID NO:X, which refers to an amino acid sequence longer than the amino acid sequence given in SEQ ID NO:X but entirely comprising the amino acid sequence given in SEQ ID NO:X (wherein the amino acid sequence given in SEQ ID NO:X can be located N-terminally or C-terminally in the longer amino acid sequence, or can be embedded in the longer amino acid sequence), or to an amino acid sequence consisting of the amino acid sequence given in SEQ ID NO:X. The term “antibody” as used herein, refers to an immunoglobulin (Ig) molecule, which specifically binds with an antigen. Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources and can be immunoreactive portions of intact immunoglobulins. Antibodies are typically tetramers of immunoglobulin molecules. The term “immunoglobulin domain” as used herein refers to a globular region of an antibody chain (such as e.g., a chain of a conventional 4-chain antibody or a chain of a heavy chain antibody), or to a polypeptide that essentially consists of such a globular region. Immunoglobulin domains are characterized in that they retain the immunoglobulin fold characteristic of antibody molecules, which consists of a two-layer sandwich of about seven antiparallel β-strands arranged in two (3-sheets, optionally stabilized by a conserved disulphide bond. The specificity of an antibody/immunoglobulin/IVD for an antigen is defined by the composition of the antigen-binding domains in the antibody/immunoglobulin/IVD (usually one or more of the CDRs, the particular amino acids of the antibody/immunoglobulin/IVD interacting with the antigen forming the paratope) and the composition of the antigen (the parts of the antigen interacting with the antibody/immunoglobulin/IVD forming the epitope). Specificity of binding is understood to refer to a binding between an antibody/immunoglobulin/IVD with a single target molecule or with a limited number of target molecules that (happen to) share an epitope recognized by the antibody/immunoglobulin/IVD. Affinity of an antibody/immunoglobulin/IVD for its target is a measure for the strength of interaction between an epitope on the target (antigen) and an epitope/antigen binding site in the antibody/immunoglobulin/IVD. It can be defined as: KA=[Ab-Ag][Ab]⁡[Ag] Wherein KA is the affinity constant, [Ab] is the molar concentration of unoccupied binding sites on the antibody/immunoglobulin/IVD, [Ag] is the molar concentration of unoccupied binding sites on the antigen, and [Ab−Ag] is the molar concentration of the antibody-antigen complex. Avidity provides information on the overall strength of an antibody/immunoglobulin/IVD-antigen complex, and generally depends on the above-described affinity, the valency of antibody/immunoglobulin/IVD and of antigen, and the structural interaction of the binding partners. The term “immunoglobulin variable domain” (abbreviated as “IVD”) as used herein means an immunoglobulin domain essentially consisting of four “framework regions” which are referred to in the art and herein below as “framework region 1” or “FR1”; as “framework region 2” or “FR2”; as “framework region 3” or “FR3”; and as “framework region 4” or “FR4”, respectively; which framework regions are interrupted by three “complementarity determining regions” or “CDRs”, which are referred to in the art and herein below as “complementarity determining region 1” or “CDR1”; as “complementarity determining region 2” or “CDR2”; and as “complementarity determining region 3” or “CDR3”, respectively. Thus, the general structure or sequence of an immunoglobulin variable domain can be indicated as follows: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4. It is the immunoglobulin variable domain(s) (IVDs) that confer specificity to an antibody for the antigen by carrying the antigen-binding site. Methods for delineating/confining a CDR in an antibody/immunoglobulin/IVD have been described hereinabove. The term “immunoglobulin single variable domain” (abbreviated as “ISVD”), equivalent to the term “single variable domain”, defines molecules wherein the antigen binding site is present on, and formed by, a single immunoglobulin domain. This sets immunoglobulin single variable domains apart from “conventional” immunoglobulins or their fragments, wherein two immunoglobulin domains, in particular two variable domains, interact to form an antigen binding site. Typically, in conventional immunoglobulins, a heavy chain variable domain (VH) and a light chain variable domain (VL) interact to form an antigen binding site. In this case, the complementarity determining regions (CDRs) of both VH and VL will contribute to the antigen binding site, i.e. a total of 6 CDRs will be involved in antigen binding site formation. In view of the above definition, the antigen-binding domain of a conventional 4-chain antibody (such as an IgG, IgM, IgA, IgD or IgE molecule; known in the art) or of a Fab fragment, a F(ab′)2 fragment, an Fv fragment such as a disulphide linked Fv or a scFv fragment, or a diabody (all known in the art) derived from such conventional 4-chain antibody, would normally not be regarded as an immunoglobulin single variable domain, as, in these cases, binding to the respective epitope of an antigen would normally not occur by one (single) immunoglobulin domain but by a pair of (associated) immunoglobulin domains such as light and heavy chain variable domains, i.e., by a VH-VL pair of immunoglobulin domains, which jointly bind to an epitope of the respective antigen. In contrast, immunoglobulin single variable domains are capable of specifically binding to an epitope of the antigen without pairing with an additional immunoglobulin variable domain. The binding site of an immunoglobulin single variable domain is formed by a single VH/VHH or VL domain. Hence, the antigen binding site of an immunoglobulin single variable domain is formed by no more than three CDRs. As such, the single variable domain may be a light chain variable domain sequence (e.g., a VL-sequence) or a suitable fragment thereof; or a heavy chain variable domain sequence (e.g., a VH-sequence or VHH sequence) or a suitable fragment thereof; as long as it is capable of forming a single antigen binding unit (i.e., a functional antigen binding unit that essentially consists of the single variable domain, such that the single antigen binding domain does not need to interact with another variable domain to form a functional antigen binding unit). In one embodiment of the invention, the immunoglobulin single variable domains are heavy chain variable domain sequences (e.g., a VH-sequence); more specifically, the immunoglobulin single variable domains can be heavy chain variable domain sequences that are derived from a conventional four-chain antibody or heavy chain variable domain sequences that are derived from a heavy chain antibody. For example, the immunoglobulin single variable domain may be a (single) domain antibody (or an amino acid sequence that is suitable for use as a (single) domain antibody), a “dAb” or dAb (or an amino acid sequence that is suitable for use as a dAb) or a Nanobody® (as defined herein, and including but not limited to a VHH); other single variable domains, or any suitable fragment of any one thereof. In particular, the immunoglobulin single variable domain may be a Nanobody® (as defined herein) or a suitable fragment thereof. Note: Nanobody®, Nanobodies® and Nanoclone® are registered trademarks of Ablynx N.V. For a general description of Nanobodies®, reference is made to the further description below, as well as to the prior art cited herein, such as e.g. described in WO2008/020079. “VHH domains”, also known as VHHs, VHH domains, VHH antibody fragments, and VHH antibodies, have originally been described as the antigen binding immunoglobulin (variable) domain of “heavy chain antibodies” (i.e., of “antibodies devoid of light chains”; Hamers-Casterman et al (1993) Nature 363: 446-448). The term “VHH domain” has been chosen to distinguish these variable domains from the heavy chain variable domains that are present in conventional 4-chain antibodies (which are referred to herein as “VH domains”) and from the light chain variable domains that are present in conventional 4-chain antibodies (which are referred to herein as “VL domains”). For a further description of VHHs and Nanobody®, reference is made to the review article by Muyldermans (Reviews in Molecular Biotechnology 74: 277-302, 2001), as well as to the following patent applications, which are mentioned as general background art: WO 94/04678, WO 95/04079 and WO 96/34103 of the Vrije Universiteit Brussel; WO 94/25591, WO 99/37681, WO 00/40968, WO 00/43507, WO 00/65057, WO 01/40310, WO 01/44301, EP 1134231 and WO 02/48193 of Unilever; WO 97/49805, WO 01/21817, WO 03/035694, WO 03/054016 and WO 03/055527 of the Vlaams Instituut voor Biotechnologie (VIB); WO 03/050531 of Algonomics N.V. and Ablynx N.V.; WO 01/90190 by the National Research Council of Canada; WO 03/025020 (=EP 1433793) by the Institute of Antibodies; as well as WO 04/041867, WO 04/041862, WO 04/041865, WO 04/041863, WO 04/062551, WO 05/044858, WO 06/40153, WO 06/079372, WO 06/122786, WO 06/122787 and WO 06/122825, by Ablynx N.V. and the further published patent applications by Ablynx N.V. As described in these references, Nanobody® (in particular VHH sequences and partially humanized Nanobody®) can in particular be characterized by the presence of one or more “Hallmark residues” in one or more of the framework sequences. A further description of the Nanobody®, including humanization and/or camelization of Nanobody®, as well as other modifications, parts or fragments, derivatives or “Nanobody® fusions”, multivalent constructs (including some non-limiting examples of linker sequences) and different modifications to increase the half-life of the Nanobody® and their preparations can be found e.g. in WO 08/101985 and WO 08/142164. “Domain antibodies”, also known as “Dabs” (the terms “Domain Antibodies” and “dAbs” being used as trademarks by the GlaxoSmithKline group of companies) have been described in e.g., EP 0368684, Ward et al. (Nature 341: 544-546, 1989), Holt et al. (Tends in Biotechnology 21: 484-490, 2003) and WO 03/002609 as well as for example WO 04/068820, WO 06/030220, WO 06/003388 and other published patent applications of Domantis Ltd. Domain antibodies essentially correspond to the VH or VL domains of non-camelid mammalians, in particular human 4-chain antibodies. In order to bind an epitope as a single antigen binding domain, i.e., without being paired with a VL or VH domain, respectively, specific selection for such antigen binding properties is required, e.g. by using libraries of human single VH or VL domain sequences. Domain antibodies have, like VHHs, a molecular weight of approximately 13 to approximately 16 kDa and, if derived from fully human sequences, do not require humanization for e.g. therapeutic use in humans. It should also be noted that single variable domains can be derived from certain species of shark (for example, the so-called “IgNAR domains”, see for example WO 05/18629). Immunoglobulin single variable domains such as Domain antibodies and Nanobody® (including VHH domains and humanized VHH domains), can be subjected to affinity maturation by introducing one or more alterations in the amino acid sequence of one or more CDRs, which alterations result in an improved affinity of the resulting immunoglobulin single variable domain for its respective antigen, as compared to the respective parent molecule. Affinity-matured immunoglobulin single variable domain molecules of the invention may be prepared by methods known in the art, for example, as described by Marks et al. (Biotechnology 10:779-783, 1992), Barbas, et al. (Proc. Nat. Acad. Sci, USA 91: 3809-3813, 1994), Shier et al. (Gene 169: 147-155, 1995), Yelton et al. (Immunol. 155: 1994-2004, 1995), Jackson et al. (J. Immunol. 154: 3310-9, 1995), Hawkins et al. (J. MoI. Biol. 226: 889 896, 1992), Johnson and Hawkins (Affinity maturation of antibodies using phage display, Oxford University Press, 1996). The process of designing/selecting and/or preparing a polypeptide, starting from an immunoglobulin single variable domain such as a Domain antibody or a Nanobody®, is also referred to herein as “formatting” said immunoglobulin single variable domain; and an immunoglobulin single variable domain that is made part of a polypeptide is said to be “formatted” or to be “in the format of” said polypeptide. Examples of ways in which an immunoglobulin single variable domain can be formatted and examples of such formats for instance to avoid glycosylation will be clear to the skilled person based on the disclosure herein. Immunoglobulin single variable domains such as Domain antibodies and Nanobody® (including VHH domains) can be subjected to humanization, i.e. increase the degree of sequence identity with the closest human germline sequence. In particular, humanized immunoglobulin single variable domains, such as Nanobody® (including VHH domains) may be immunoglobulin single variable domains that are as generally defined for in the previous paragraphs, but in which at least one amino acid residue is present (and in particular, at least one framework residue) that is and/or that corresponds to a humanizing substitution (as defined herein). Potentially useful humanizing substitutions can be ascertained by comparing the sequence of the framework regions of a naturally occurring VHH sequence with the corresponding framework sequence of one or more closely related human VH sequences, after which one or more of the potentially useful humanizing substitutions (or combinations thereof) thus determined can be introduced into said VHH sequence (in any manner known per se, as further described herein) and the resulting humanized VHH sequences can be tested for affinity for the target, for stability, for ease and level of expression, and/or for other desired properties. In this way, by means of a limited degree of trial and error, other suitable humanizing substitutions (or suitable combinations thereof) can be determined by the skilled person. Also, based on what is described before, (the framework regions of) an immunoglobulin single variable domain, such as a Nanobody® (including VHH domains) may be partially humanized or fully humanized. A “serum albumin binding agent”, or “serum albumin binding polypeptide”, as used herein, is a protein-based agent capable of specific binding to serum albumin. In various embodiments, the serum albumin binding agent may bind to the full-length and/or mature forms and/or isoforms and/or splice variants and/or fragments and/or any other naturally occurring or synthetic analogues, variants or mutants of serum albumin. In various embodiments, the serum albumin binding agent of the invention may bind to any forms of serum albumin, including monomeric, dimeric, trimeric, tetrameric, heterodimeric, multimeric and associated forms. In an embodiment, the serum albumin binding agent binds to the monomeric form of serum albumin. In an embodiment, the present serum albumin binding polypeptide comprises immunoglobulin variable domain with an antigen binding site that comprises three complementarity determining regions (CDR1, CDR2 and CDR3). In an embodiment said antigen binding site recognizes one or more epitopes present on serum albumin. In various embodiments, the serum albumin binding agent comprises a full length antibody or fragments thereof. In an embodiment, the serum albumin binding agent comprises a single domain antibody or an immunoglobulin single variable domain (ISVD). In a specific embodiment, the serum albumin binding agent binds to serum albumin of rat (Uniprot P02770). In a specific embodiment, the serum albumin binding agent binds to serum albumin of mouse (Uniprot P07724). In a specific embodiment, the serum albumin binding agent binds to human serum albumin (Uniprot P02768). The aspects and embodiments described above in general may comprise the administration of a huPDL1-binding polypeptide or pharmaceutical composition comprising it to a mammal in need thereof, i.e., harbouring a tumour, cancer or neoplasm in need of (non-invasive) medical imaging, diagnosis, treatment, surgery, therapy monitoring, or dendritic cell vaccination. In general a (therapeutically) effective amount of the huPDL1-binding polypeptide or pharmaceutical composition comprising it is administered to the mammal in need thereof in order to meet the desired effect. The (therapeutically) effective amount will depend on many factors such as route of administration and will need to be determined on a case-by-case basis by the physician. In general the maximum dose of (therapeutically) effective amount of huPDL1-binding polypeptide or pharmaceutical composition comprising it that may be administered to a mammal is determined by the possible toxicity and is reflected in the maximum tolerated dose (MTD), i.e. the highest dose that does not cause unacceptable side effects. “Administering” means any mode of contacting that results in interaction between an agent (e.g. a huPDL1-binding polypeptide as described herein) or composition comprising the agent (such as a medicament or pharmaceutical composition) and an object (e.g. cell, tissue, organ, body lumen) with which said agent or composition is contacted. The interaction between the agent or composition and the object can occur starting immediately or nearly immediately with the administration of the agent or composition, can occur over an extended time period (starting immediately or nearly immediately with the administration of the agent or composition), or can be delayed relative to the time of administration of the agent or composition. More specifically the “contacting” results in delivering an effective amount of the agent or composition comprising the agent to the object. The term “effective amount” refers to the dosing regimen of the agent (e.g. huPDL1-binding polypeptide as described herein) or composition comprising the agent (e.g. medicament or pharmaceutical composition). The effective amount will generally depend on and/or will need adjustment to the mode of contacting or administration. To obtain or maintain the effective amount, the agent or composition comprising the agent may be administered as a single dose or in multiple doses. The effective amount may further vary depending on the severity of the condition that needs to be diagnosed, imaged, or treated; this may depend on the overall health and physical condition of the mammal or patient and usually a doctor's or physician's assessment will be required to establish what is the effective amount. The effective amount may further be obtained by a combination of different types of contacting or administration. It is to be understood that although particular embodiments, specific configurations as well as materials and/or molecules, have been discussed herein for cells and methods according to the present invention, various changes or modifications in form and detail may be made without departing from the scope and spirit of this invention. The following examples are provided to better illustrate particular embodiments, and they should not be considered limiting the application. The application is limited only by the claims. The content of the documents cited herein are incorporated by reference. EXAMPLES 1. Materials and Methods 1.1. Reagents All Biacore consumables were from GE Healthcare. A recombinant His-tagged human PD-L1 protein (SINO Biologicals, 10084-H08H) was used to determine the affinity of purified single domain antibodies (sdAbs) in Surface Plasmon Resonance (SPR). Recombinant Fc-tagged human PD-L1 (R&D Systems, 156-B7) or PD-1 (R&D Systems, 1086-PD) proteins were used to evaluate the IC50in SPR. Avelumab (Bavencio) was provided by Merck KGaA [EMD Serono] and Pfizer. A sdAb specific for a multiple myeloma paraprotein, designated R3B23 (Lemaire et al. 2014, Leukemia 28:444-447), and trastuzumab (Herceptin®, Roche) served as negative controls. The following blocking anti-PD-L1 mAbs were used in the functional assays; the IgG1 mAbs, MIH1 [eBioscience] and avelumab [Bavencio®, Merck KGaA], and the IgG2b mAb 29E.2A3 [Bioxcell]. The isotype-matched control mAbs, P3.6.2.8.1 [IgG1, eBiosciences] and MOPC-21 [IgG2b, Bioxcell], were used as controls. The human PD-L1-specific sdAb K2 is described herein. An sdAb specific for the 5T2MM paraprotein, sdAb R3B23, was used as a control (Lemaire et al. 2014, Leukemia 28:444-447). An anti-His monoclonal antibody (mAb) (AbD Serotec, AD1.1.10) and phycoerythrin (PE) conjugated anti-mouse IgG antibody (BD biosciences, A85-1) was used to detect binding of purified His-tagged sdAbs to PD-L1 expressed on cells in flow cytometry. An allophycocyanin (APC) conjugated antibody specific for human PD-L1 (eBioscience, MIH5) was used in flow cytometry to evaluate PD-L1 expression on cells. A PE conjugated anti-HLA-A2 antibody (BD Biosciences, BB7.2) and conjugated anti-CD45 antibody were used to discriminate tumour cells from immune cells. An anti-human PE-labelled IgG1 antibody (Miltenyi Biotec, IS11-12E4.23.20) was used to detect binding of avelumab to PD-L1POS 293T cells. Expression of PD-L1 on cells was evaluated with anti-PD-L1 antibodies coupled to allophycocyanin (APC, eBioscience, MIH1) or PE-CF594 (Biolegend, MIH1), HLA-A2 using a PE-conjugated anti-HLA-A2 antibody (BD biosciences, BB7.2), PD-1 using a PE-conjugated anti-PD-1 antibody (Biolegends, EH12.2H7). 2D3 cells were discriminated from tumour cells in the 2D3 functional assay using an APC-H7-labelled anti-CD8 antibody (BD biosciences, SK1). Expression of the T-cell receptor (TCR) on electroporated 2D3 cells was evaluated with a PE-labelled anti-TCRα/β antibody (Biolegend, IP26). Isotype-matched antibodies served as controls (BD biosciences). The following antibodies were used to phenotype the cells used in functional assays: a PECF594 conjugated anti-CD3 (Biolegend, UCHT1) and anti-CD70 (BD Biosciences, Ki-24), a PerCP-Cy5.5 conjugated anti-CD4 (BD Biosciences, RPA-T4), an APC-H7 conjugated anti-CD8 (BD Biosciences, SK1), a PE conjugated anti-PD-1 (BD Biosciences, MIH4) and anti-HLA-A2 (BD Biosciences, BB7.2), a PE-Cy7 conjugated anti-HLA-DR (BD Biosciences, G46-6), a fluorescein isothiocyanate conjugated anti-CD86 (BD Biosciences, FUN-1), an APC conjugated anti-PD-L2 (BD Biosciences, MIH18), a PerCPEF710 conjugated anti-CD80 (eBiosciences, 2D10.4), an anti-PD-L1-APC [eBioscience, MIH5], an anti-PD-1-PE [Biolegend, EH12.2H7], an anti-CD11c-AF700 [BD biosciences, clone B-ly6], an anti-PD-L1-PE-CF594 [BD Biosciences, clone MIH1], an anti-CD86-BV421 [BD biosciences, clone HB15e], an anti-CD83-PE [BD Biosciences, clone HB15e], an anti-CD40-APC [Biolegend, clone 5C3], an anti-CD80-PerCP-EF710 [eBioscience, clone 2D10.4], an anti-HLA-ABC-FITC [BD biosciences, clone G46-2.6]. Isotype matched control (IC) antibodies were purchased from BD Biosciences. A Melan-A/MART-1 HLA-A2 dextramer conjugated to PE (ELAGIGILTV, SEQ ID NO:19; Immudex) was used to detect Melan-A specific T cells in flow cytometry. A gp100 HLA-A2 dextramer conjugated to PE (YLEPGPVTV, SEQ ID NO:20; Immudex) was used as a control. The gp100280-288peptide (YLEPGPVTA, SEQ ID NO:21; Eurogentec) was used to pulse antigen presenting cells in the 2D3 assay. A blocking anti-PD-L1 antibody (eBioscience, MIH1) and an isotype matched control antibody (eBioscience, P3.6.2.8.1) were used in the 2D3 assay. The anti-PD-L1 antibody (29E.2A3) and its isotype matched control antibody (MPC-11) purchased from Bioxcell were used in the other functional assays. Avelumab (Bavencio®, provided by Merck KGaA [EMD Serono] and Pfizer), an isotype-matched control antibody (Bioxcell, MOPC-21) and R3B23 were used in the 2D3 and 3D spheroid assays as controls. 1.2. Generation and Selection of PD-L1 Specific sdAbs Human PD-L1 specific sdAbs were generated in alpacas. Briefly, alpacas were immunized subcutaneously for 5 to 6 times at a weekly to biweekly interval with either 10×10E6 RAW264.7 cells or with 100 μg recombinant human PD-L1-Fc protein (R&D Systems, 156-B7). Peripheral blood lymphocytes were purified and used as a source to create a sdAb phage display library. PD-L1 reactive sdAbs were identified by biopanning of this library and ELISA screening of periplasmatic extracts of individual sdAb clones on recombinant mouse or human PD-L1 protein. Sequence analysis was performed on sdAb clones that specifically bound PD-L1. Anti-PD-L1 sdAbs and the control sdAb R3B23 were produced and purified as described (Broos et al. 2017, Oncotarget 8:41932-41946). Therefore, the sdAb cDNA was cloned in the vector pHEN6 to incorporate a C-terminal HIS-tag. 1.3. Large-Scale Selection, Production and Purification of sdAbs The selected sdAbs and sdAb R3B23, specific for the 5T2MM paraprotein (Lemaire et al. 2014, Leukemia 28:444-447), were produced and purified including cloning of the sdAb encoding cDNAs into the vector pHEN6 as to incorporate a C-terminal HIS-tag. 1.4. Surface Plasmon Resonance All measurements were performed on a Biacore T200 device (GE Healtcare) at 25° C. and using Hepes-buffered saline (0.01M HEPES, pH 7.4; 0.15M NaCl, 3 mM EDTA, 0.005% Tween20) as running buffer. All recombinant proteins were dissolved to 10 μg/mL in 10 mM Na-acetate (pH 5.0) for immobilization on a CM5 sensor chip using linkage chemistry with 1-(3-(dimethylamino)propyl)-3-ethylcarbodiimide (EDC) and N-hydroxy-succinimide (NETS). Unreacted EDC-NHS linkers were blocked with 1M ethanolamine-HCl. For all measurements, SPR signals in the flow cell with immobilized protein were subtracted with those in a flow cell that underwent the same manipulations but where recombinant protein was omitted, to obtain specific binding signals (response units, RU). Affinity for human PD-L1 of the purified sdAbs was evaluated on immobilized PD-L1 protein. To evaluate the sdAb's IC50, the sdAb concentration at which the relative response of the interaction between PD-1 and PD-L1 is inhibited by half, Fc-PD-1 protein was immobilized on a CM5 chip. Different concentrations of the sdAb (400 to 0.78 nM using a 2-fold dilution series or an excess amount of 1000 nM) were mixed with recombinant human Fc-PD-L1 protein using the KD-value concentration of the PD-L1:PD-1 interaction (25 nM), and run over the chip. The maximum relative response values were plotted in function of competing sdAb concentration and analyzed with a “Log inhibitor versus response (variable lope)” model in Prism to calculate IC50values. To evaluate competition between sdAbs and avelumab for binding to PD-L1, competition studies were performed as described (Vaneycken et al. 2011, FASEB J 25:2433-2446). 1.5. Mice and Cell Lines Female C57BL/6 mice and athymic nude mice (Crl:NU(NCr)-Foxn1nu) were supplied by Charles River Laboratories (France) at 6 weeks of age. All experiments were performed in accordance to the European guidelines for animal experimentation under licenses LA1230214 and LA1230272. Experiments were approved by the Ethical Committee for the use of laboratory animals of the Vrije Universiteit Brussel (ECD 15-214-1 and 17-272-6). Human embryonal kidney (HEK) 293T cells and HLA-A*0201+breast carcinoma cells (MCF7) were purchased from the American Type Culture Collection (ATCC). HLA-A*0201+ 624-MEL or 938-MEL cells were provided by S.L. Topalian (National Cancer Institute, USA). 624-MEL and 938-MEL cells were cultured in RPMI1640 medium supplemented with 10% Fetal clone I serum (Thermoscientific), 2 mM L-Glutamine, 100 U/ml penicillin, 100 μg/ml streptomycin, 1 mM sodium pyruvate and nonessential amino acids (Sigma-Aldrich). HEK293T cells were cultured in Dulbecco's modified Eagle's medium (Sigma-Aldrich) supplemented with 10% foetal bovine serum (FBS, Harlan), 2 mM L-Glutamine (L-Glu, Sigma Aldrich) and 100 U/ml penicillin, 100 μg/ml streptomycin (PS, Sigma-Aldrich). MCF7 cells were cultured in Roswell Park Memorial Institute (RPMI) 1640 medium supplemented with FBS, L-Glu, PS, 1 mM sodium pyruvate and nonessential amino acids (Sigma-Aldrich). 2D3 cells were generated as described in Versteven et al. 2018 (Oncotarget 9:27797-27808) and maintained in Iscove's Modified Dulbecco's Medium (IMDM, Invitrogen) supplemented with 10% FBS. Experiments were performed using blood samples from healthy HLA-A*0201+donors provided by the Blood Transfusion Center of the University Hospital Brussel (Brussels, Belgium). Isolation of peripheral blood mononuclear cells (PBMCs), CD14+monocytes and their differentiation to monocyte derived dendritic cells (moDCs) as well as isolation of CD8+T cells from the remaining PBMCs was performed as described in Tuyaerts et al. 2002 (J Immunol Methods 264:135-151). This study was approved by the Ethics Committees of the Brussels University Hospital (2013/198). 1.6. Lentiviral Production, Characterization and Transduction The plasmids pCMVΔR8.9 and pMD.G were a gift from D. Trono (Ecole Polytechnique Federal de Lausanne, Swiss). The transfer plasmids encoding eGFP, human PD-L1 and PD-1 were described (Pen et al. 2014, Gene Ther 21:262-271; Breckpot et al. 2003, Gene Med 5:654-667). The production and characterization of lentiviral vectors was described in Goyvaerts et al. 2013 (Gene Ther 19:1133-1140). Transduction of HEK293T, MCF7 and 624-MEL cells with PD-L1 or eGFP encoding lentiviral vectors was carried out at a MOI of 10, while transduction of 2D3 cells with PD-1 encoding lentiviral vectors was carried out at a MOI of 5 using the protocol described to transduce human moDCs (Breckpot et al. 2003, J Gene Med 5:654-667). 1.7. Tumour Challenge Athymic nude mice were injected subcutaneously with 5×10E6 MCF7, 624-MEL, 938-MEL, or PD-L1 modified MCF7 or 624-MEL cells. One day before transplanting MCF7 cells, mice were implanted with oestrogen pellets (Innovative research of America; 0.36 mg/mice). When mice developed a palpable tumour, tumour volume was followed using an electronic calliper. The tumour length and width were measured using an electronic calliper, and used to calculate the tumour volume using the formula: (length×width2)/2. One day prior to imaging, 938-MEL tumour bearing mice were injected intratumourally with 50 μl phosphate buffered saline (PBS; Sigma-Aldrich) or IFN-gamma (2×106IUs/ml, ImmunoTools). Tumour tissue was reduced to single cells using the GentleMACS tumour dissociation protocol (Miltenyi Biotec) (Maenhout et al. 2014, Oncotarget 30:6801-6815). 1.8.99mTc-sdAb Labelling, Pinhole SPECT-Micro-CT Imaging and Image Analysis The sdAbs were labelled as described by Xavier et al. 2012 (Methods Mol Biol 911:485-490). Briefly, the sdAb's C-terminal HIS-tag was coupled to99mTc-tricarbonyl intermediate [99mTc(H2O)3(CO)3]99m, which was synthesized using the Isolink® labelling kit (Mallinckrodt Medical BV). The99mTc-sdAb solution was purified on a NAP-5 column (GE Healthcare) pre-equilibrated with PBS to remove unbound (99mTc(H20)3(CO)3)+and finally filtered through a 0.22 μm filter (Millipore) to remove aggregates. The labelling efficiency was determined both directly after labelling and after purification by instant thin-layer chromatography (iTLC) with 100% acetone as the mobile phase. Mice were injected intravenously with 100-2004, of 45-155MBq of99mTc-labelled sdAbs (10 μg), one hour prior to pinhole SPECT-micro-CT imaging. Imaging was performed as described (Put et al. 2013, J Nucl Med 54:807-814). Micro-CT was performed using a dual-source CT scanner (Skyscan 1178; Skyscan) with 60 kV and 615 mA at a resolution of 83 μm. CT images were reconstructed using filtered back projection (NRecon; Skyscan). Pinhole SPECT micro-CT imaging and image analysis in naive C57BL6 mice, the MCF7 and 624-MEL tumour model were performed as described (Broos et al. 2017, Oncotarget 8:41932-41946). For the 938-MEL model, SPECT/CT was performed on a MILabs VECTor/CT camera. The CT-scan was set to 60 kV and 615 mA. CT scan time was 139 seconds. SPECT-images were obtained using a rat SPECT-collimator (1.5-mm pinholes) in spiral mode, 6 positions for whole-body imaging, with 150 seconds per position, total body SPECT scan was 15 minutes. Images were reconstructed with 0.4 mm voxels with 2 subsets and 4 iterations, without post-reconstruction filter. SPECT images were reconstructed using an iterative reconstruction algorithm (ordered-subset expectation maximization) modified for the 3-pinhole geometry and automatically reoriented for fusion with CT images based on six57Co landmarks (Vanhove et al. 2009, Eur J Nucl Med Mol Imaging 36:1049-1063). Images were further visually analyzed and quantified where appropriate using AMIDE (Medical Image Data Examiner software) (Loening & Gambhir 2003, Mol Imaging 2:131-137). Maximum intensity projections (MIP) were generated using OsiriX Lite software. After imaging, mice were sacrificed and selected organs were isolated to measure radioactivity using a γ-counter (Cobra Inspector 5003, Packard). The amount of radioactivity in organs is expressed as percent injected activity per gram (% IA/g). 1.9. mRNA Production, Electroporation The human gp100 TCRα and TCRβ pGEM-vectors were kindly provided by Prof. N. Schaft (Universitätsklinikum Erlangen, Germany) (Schaft et al. 2003, J Immunol 170:2186-2194). The peTheRNA plasmids encoding CD40 Ligand, CD70 and a constitutively active form of TLR4 were described in De Keersmaecker et al (in press). The pGEM-sig-Melan-A-DCLamp plasmid encoding the full-length Melan-A/MART-1 antigen containing the optimized immunodominant Melan-A:HLA-A2 epitope linked to the HLA-II targeting sequence of DC-Lamp was described in Bonehill et al. 2008 (Mol Ther 16:1170-1180). The production, purification, quantification and quality control of mRNA was performed as described (Tuyaerts et al. 2002, J Immunol Meth 264:135-151). Human gp100 TCRα and β mRNA (2.5 μg each/10E6 cells) was electroporated into 2D3 cells in 2004, OptiMEM medium (Life Technologies) in a 4 mm electroporation cuvette (Cell Projects) using a time constant protocol (300V, 7 ms) and the Gene Pulser Xcell™ device (BIORAD). Electroporation of moDCs with mRNA was performed as described (Tuyaerts et al. 2002, J Immunol Meth 264:135-151). 1.10. 2D3 Assay The 2D3 assay is detailed elsewhere (Versteven et al., submitted for publication). Briefly, 2D3 cells electroporated to express the TCR recognizing the gp100280-288peptide (YLEPGPVTA, SEQ ID NO:21) restricted to HLA-A2 and modified (or not) to express PD-1 were plated in a 96-well round-bottom plate at 10E5 cells in 200 μL IMDM containing 10% FBS (triplicate). moDCs, MCF7, 624-MEL, PD-L1 engineered MCF7 or 624-MEL cells were pulsed with 50 μg/mL gp100280-288peptide and added to the cultures at effector-stimulator ratios of 10:1 in 100 μL medium. Co-cultures were performed for 24 hours at 37° C., 5% CO2in the presence of 1 μg/200 μL neutralizing anti-PD-L1 antibody, avelumab (360 nM), or anti-PD-L1 sdAb. Isotype matched control antibodies or sdAb R3B23 were used as controls. The activation of 2D3 cells was measured in flow cytometry as percentage eGFP+cells within CD8+2D3 cells. 1.11. Stimulation of CD8+Melan-A Specific T Cells by Dendritic Cells CD8+T cells were plated at 10E5 cells in triplicate in a 96-well round-bottom plate in 100 μL IMDM containing 1% heat-inactivated human AB serum (Innovative Research), PS, L-Glu and non-essential amino acids. moDCs were electroporated with CD40 Ligand, CD70 and constitutively active TLR4 (10 μg each per 4×10E6 cells) and 10 μg Melan-A mRNA (referred to as TriMixDC-MEL) or solely 10 μg Melan-A mRNA (referred to as DC-MEL). Electroporated DCs were added to the T cells at an effector: stimulator ratio of 10:1 in 100 μL medium. Co-cultures with TriMixDC-MEL were performed for 7 days at 37° C., 5% CO2in the presence of 1 μg/200 μL neutralizing anti-PD-L1 antibody or anti-PD-L1 sdAb. Isotype matched control antibodies or sdAb R3B23 were used as controls. Stimulation of T cells with DC-MEL was performed in analogy to the stimulation with TriMixDC-MEL, however, T cells were in this case restimulated on day 7 and analysis of T cell activation through dextramer staining (flow cytometry) and evaluation of the production of cytokines IFN-γ (ELISA, Thermo Scientific) was performed on day 14. 1.12. Proliferation Assay PBMCs depleted from CD14+cells from healthy donor were labelled with 0.5 μM CellTrace Violet (Invitrogen). These cells (10E5) were co-cultured for 6 days with or without TriMixDC-MEL (4.8 μg each per 4×10E6 cells) or DC-MEL (4.8 μg Melan-A mRNA) at a effector:stimulator ratio of 10:1 in 200 μL IMDM containing 1% heat-inactivated human AB serum, PS, L-Glu and non-essential amino acids. T-cell proliferation was measured in flow cytometry as the dilution of the CellTrace Violet dye in the CD8+ T-cell population. Proliferation observed in cultures without TriMixDC-MEL or DC-MEL was considered as background. 1.13. Quantitative Reverse Transcriptase Polymerase Chain Reaction Isolation of total RNA from CD8+T cells and its reverse transcription to cDNA was performed as described in Van der Jeught et al. 2014 (Oncotarget 5:10100-10113). To evaluate PD-1 mRNA levels, samples were subjected to a SYBRgreen (Thermofisher) based real-time PCR-analysis on a BIORAD device. Primers for amplification of PD-1 were as follow: reverse: 5′-CTTCTCTCGCCACTGGAAAT-3′ (SEQ ID NO:13) and forward: 5′-CCGCACGAGGGACAATAG-3′ (SEQ ID NO:14) (Integrated DNA Technologies). Primers for the amplification of peptidylprolyl isomerase A (Ppia) were as follow: 5′-TTCACCTTCCCAAAGACCAC-3′ (SEQ ID NO:15) and 5′CAAACACAAACGGTTCCCAG-3′ (SEQ ID NO:16) (Integrated DNA Technologies). 1.14. Preparation of Single Cell Suspensions from In Vivo Grown Tumours Single cell suspensions were prepared after isolation of tumours from mice using the GentleMACS single cell isolation protocol (Miltenyi Biotec) in order to perform flow cytometry to analyze expression of PD-L1 on tumour cells. 1.15. Flow Cytometry The procedure for staining of cellular surface markers was previously described (Breckpot et al. 2003, J Gene Med 5:654-667). All cells were acquired on the LSRFortessa flow cytometer (BD Biosciences) and data were analyzed with FACSDiva (BD Biosciences) or FlowJo (Tristar Inc.) software. 1.16. Statistical Analysis Results are expressed as mean±standard error of the mean. A non-parametric Mann-Whitney U test was carried out to compare data sets. Sample sizes and number of times experiments were repeated are indicated in the figure legends. The number of asterisks in the figures indicates the statistical significance as follows: *P<0.05; **P<0.01; ***P<0.001. 1.17. 3D Spheroid Cytotoxicity Assay 624-MEL cells engineered to express eGFP and PD-L1, were plated at 200 cells in an ultra-low attachment 96-well plate (Costar®, ref 7007) and kept in culture for 1 day to form 3D spheroids. Subsequently, PBMCs stimulated for 24 hours with 10 ng/mL interleukin-2 (IL-2) (Peprotech) and 10 ng/mL anti-CD3 mAb (BioLegend, ref 317302) were added to the cells at a ratio of 1:50 in the presence of 3.6 μM avelumab, isotype-matched mAbs, K2, R3B23, or the combination of mAbs and sdAbs. In a separate assay, K2 or R3B23 were added every 24 hours to the co-culture after centrifuging the plate at 1200 rpm for 10 minutes and removing 50 μl of the co-culture. The reduction of total amount of green object area within each well containing eGFPPOS and PD-L1POS cells was evaluated every hour for seven consecutive days in an IncuCyte Zoom® live cell imaging system (EssenBio). 1.18. Evaluation of DC Maturation in Response to Endotoxins Present in sdAb Preparations To evaluate the effect of any endotoxins in the sdAb solutions, we incubated moDCs for 24 hours with 10 μg sdAb K2 or sdAb R3B23 at 37° C. and 5% CO2. Untreated moDCs and moDCs treated with 1 ng/ml lipopolysaccharide [LPS] served as negative and positive controls, respectively. Up-regulation of maturation markers was evaluated in flow cytometry. 2. Results 2.1. Generation of High Affinity Single Domain Antibodies Specific for PD-L1 sdAbs were raised against PD-L1 through immunization of alpacas with recombinant human PD-L1 protein or with RAW624.7 macrophages that expressed mouse PD-L1. Peripheral blood lymphocytes from these alpacas were used to create a sdAb phage display library. Biopanning and screening on the immunogen was performed, resulting in a total of 42 sdAbs that were selected for binding to human and/or mouse PD-L1, both on recombinant proteins and on cells. Based on the amino acid sequence of the CDR1, 2 and 3 regions, these sdAbs were divided into 13 sequence families of which the mouse PD-L1 binding sdAbs were reported in Broos et al. 2017 (Oncotarget 8:41932). Several sdAbs bound to human PD-L1. Of these, the sequence family K, represented by sdAbs K2, K3 and K4, showed high affinity binding to human PD-L1 (FIG.1,FIG.2A-B). We showed that sdAbs K2, K3 and K4 bind with similar nanomolar affinity (KD=5.2, 3.5 and 4.5 nM respectively) to human PD-L1 (FIG.2B-D). Furthermore, we showed that sdAbs K2, K3 and K4 and avelumab were able to bind with human PD-L1 expressed on HEK293T cells (FIG.2C). The affinity of avelumab for human PD-L1 was determined as KD=1.6 nM. 2.2. The PD-L1 Specific sdAb K2 Generates Strong Positive Contrast in SPECT/CT Imaging We radiolabelled sdAb K2, K3 and K4 with99mTc through complexation of the99mTc-tricarbonyl with its HIS-tag followed by purification by filtration and NAP-5 column. This resulted in a radiochemical purity of >98% for sdAb K2 and K3, however, yielded low radiochemical purity (93%) for sdAb K4 (Table 1). Therefore, sdAb K4 was excluded for further analysis. The biodistribution of sdAbs K2 and K3 was evaluated in healthy C57BL/6 mice using SPECT/CT imaging, showing low signals in the liver, kidneys and bladder (FIG.3A). These signals are a consequence of the metabolization and renal excretion typically observed in sdAb mediated imaging. Quantification of tracer uptake by ex vivo biodistribution analysis, revealed that uptake of sdAb K2 and K3 in liver (1.292±0.063 and 3.191±0.060% IA/g, respectively), left kidney (33.99±1.014 and 55.39±4.27% IA/g) and right kidney (34.06±1.530 and 57.42±4.219% IA/g) were extremely low when compared to values typically obtained with sdAbs used for imaging of other marker (Broisat et al. 2012, Circ Res 110:927-937) (FIG.3B). We selected sdAb K2 for further analysis, because this sdAb showed the lowest uptake in liver and kidneys. TABLE 1Nap5EluateFilterfiltrateRadiochemical puritysdAb(mCi)(mCi)(mCi)mCiafter purificationK23.4923.543.6119.8698%K412.064.452.032.3093%K35.2322.515.8216.2498% We transplanted MCF7 breast cancer and 624-MEL melanoma cells, or their PD-L1 engineered counterparts subcutaneously in athymic nude mice (FIG.7A-B). SPECT/CT imaging was performed, generating strong positive contrast images in mice bearing PD-L1+tumours (FIG.4A&FIG.8A). Ex vivo analysis (gamma-counting) confirmed the accumulation of sdAb K2 in PD-L1+MCF7 and 624-MEL tumours (3.07±0.24 and 4.86±0.73% IA/g, respectively) when compared to PD-L1−MCF7 and 624-MEL tumours (0.73±0.15 and 0.85±0.24% IA/g, respectively) (FIG.3B&FIG.8B). Using flow cytometry we confirmed the expression of PD-L1 on the tumour cells (FIG.3C&FIG.8C). See alsoFIGS.13and14. 2.3. sdAb K2 Facilitates Activation of 2D3 Cells by Dendritic Cells and Tumour Cells Because sdAb K2 showed high capacity to penetrate tumours, we decided to evaluate whether it has blocking activity and therefore might be used as a therapeutic agent. We showed in SPR that sdAb K2 is able to reduce the interaction between PD-1:PD-L1 with an IC50value of 9.5 nM (FIG.5A). We previously optimized a functional cell based assay to evaluate the blocking capacity of mAbs targeting PD-1 or PD-L1, using 2D3 cells engineered to express a specific TCR and PD-1 (Versteven et al, Oncotarget, under review). Therefore, we decided to use this platform to evaluate the blocking ability of sdAb K2 using HLA-A2+moDCs or MCF7 cells pulsed with the gp100280-288peptide as cells to activate PD-F or PD-1+2D3 cells expressing the TCR recognizing gp100280-288in the context of HLA-A2 (FIG.5B). We observed that the activation of 2D3 cells as measured by their expression of eGFP in flow cytometry was inhibited upon interaction of PD-1:PD-L1, and that this inhibition could be alleviated through addition of blocking anti-PD-L1 mAbs [MIH, IgG1) or sdAb K2 but not isotype matched mAbs or sdAb R3B23 (recognizing the 5T2MM paraprotein) (FIG.5C-D). Comparable results as with mAb MIH1 and sdAb K2 were obtained with the IgG1 mAb avelumab (results not shown). To exclude that the activation of 2D3 cells in the context of moDC stimulation was due to maturation of the moDCs through endotoxins present in the sdAb preparations (3.96EU/mL), we compared the phenotype of moDCs that were untreated with moDCs treated with sdAb K2 or sdAb R3B23. Upregulation of activation associated phenotypic markers like PD-L1, CD83, CD40, CD80 and MHC-I was only observed when moDCs were treated with LPS ((FIG.5E). These results indicate that the increase in TCR-signalling in PD-1pos2D3 cells during antigen-presentation by PD-L1posmoDCs in the presence of sdAb K2 is due to inhibition of the PD-1/PD-L1 interaction and not due to an increase in HLA-I expression, therefore antigen presentation. In conclusion sdAb K2 potently inhibits the interaction between PD-1/PD-L1 during antigen presentation by moDCs and as such enhances TCR-signalling, as shown by the NFAT-mediated up-regulation of eGFP in PD-1pos2D3 cells. The above platform was further relied on to evaluate the blocking ability of sdAbK2, as schematized inFIG.16C,D. HLA-A2POS PD-L1POS or PD-L1NEG 624-MEL melanoma cells or MCF7 breast cancer cells (FIG.16C) were pulsed with the gp100280-288peptide and used to activate PD-1POS 2D3 cells expressing the TCRαβ recognizing gp100280-288in the context of HLA-A2 (FIG.16D). In the absence of PD-L1 blocking agents (FIG.16E-F, condition ‘no’), co-culture of PD-1POS 2D3 cells with PD-L1POS 624-MEL tumour cells reduced the percentage of eGFPPOS cells as compared to co-culture with PD-L1NEG tumour cells. This reflects the interaction of PD-1 on 2D3 cells with PD-L1 on melanoma cells, which inhibits TCR signalling (Karwacz et al. 2011, EMBO Mol Med 3:581-592). The inhibition could be alleviated through addition of avelumab or sdAb K2 but not of isotype matched mAbs or R3B23 (FIG.16E). A similar experiment was performed using HLA-A2POS PD-L1POS or PD-L1NEG MCF7 breast cancer cells as stimulator cells, confirming in another tumour model that sdAb K2 is able to block the interaction between PD-1 and PD-L1 and as such enhances TCR signalling (FIG.16F). 2.4. sdAb K2 Facilitates Activation of T Cells by Dendritic Cells We previously developed a DC-manufacturing protocol in which moDCs were electroporated with 4 different mRNA molecules. More-specifically mRNA encoding for a melanoma antigen fused to an HLA-II targeting signal, and 3 mRNA molecules encoding for proteins that enhance the immunogenicity of the moDCs; CD40 ligand, CD70 and a constitutively active toll-like receptor 4, together referred to as TriMix. These moDCs are referred to as TriMixDCs, and were shown to be potent activators of antigen-specific CD8posT cells (Bonehill et al. 2009, Clin Cancer Res 15:3366-3375). Moreover, these TriMixDCs induce durable objective responses in 4 out of 15 [26.7%] pre-treated advanced-stage melanoma patients, thereby being among the most potent DC-vaccines described in literature (Wilgenhof et al. 2013, Ann Oncol 24:2686-2693). Vaccination with moDCs in particular TriMixDC-MEL was shown to be a promising strategy to treat patients with melanoma (Wilgenhof et al. 2013, Annals Oncol 24:2686-2693; Van Lint et al. 2014, Cancer Immunol Immunother 63:959-967; Wilgenhof et al. 2015, Cancer Immunol Immunother 64:381-388). Since TriMixDC-MEL represent mature, PD-L1 expressing moDCs, we assessed whether we could improve this vaccination strategy by supplementing the TriMixDC-MEL vaccine with sdAb K2 during the antigen presentation process (FIG.9A). We stimulated CD8+T cells from HLA-A2-positive healthy donors with TriMixDC-MEL (TriMixDC modified to present Melan-A,FIG.6G) in the presence of blocking anti-PD-L1 mAbs (mAb 29E.2A3, IgG2b), sdAb K2, isotype matched mAbs or sdAb R3B23. In these cultures, CD8+T cells showed no expression of PD-1, PD-L1 or CD80 at the start of culture as assessed by flow cytometry (FIGS.6H and9B). Quantitative real time PCR confirmed the lack of PD-1 at the start of culture, however, showed upregulation of PD-1 during stimulation (FIG.9C). We showed that the presence of sdAb K2 or the blocking anti-PD-L1 mAb 29E.2A3 during antigen presentation by TriMixDC-MEL to CD8+ T-cells did not significantly increase the number of Melan-A specific T cells. Corroborating these data, no significant increase in secretion of IFN-γ by Melan-A-specific T cells was observed in the presence of mAb 29E.2A3 compared to isotype-matched control mAb, or in the presence of sdAb K2 compared to sdAb R3B23 (FIG.6A-C). These results suggest that the co-inhibitory signal provided by PD-L1 is a lesser determinant in the degree of T-cell activation when co-stimulatory signals such as CD70, CD86, . . . , are abundantly provided by the antigen-presenting cells, in this case TriMixDC-MEL. We hypothesized that the lack of a statistical significant increase in T-cell activation could be due to the fact that TriMixDC-MEL, which express the strong co-stimulatory molecules CD70, CD80 and CD86, are already optimally equipped to activate T cells, while this might not be the case for DC vaccines that are less mature, and even lack CD70, CD80 and CD86 expression. Therefore, we repeated the T-cell stimulation experiment using DC-MEL, moDCs electroporated with Melan-A mRNA as antigen presenting cells (FIGS.6I and9A). Two rounds of stimulation were performed with these less mature, Melan-A presenting moDCs to obtain sufficient Melan-A specific CD8+T cells for analysis both in the presence of blocking anti-PD-L1 mAbs (29E.2A3), sdAb K2, isotype matched mAbs or sdAb R3B23. We showed that the presence of sdAb K2 but not the mAb resulted in significantly higher amounts of Melan-A specific T cells, which showed higher proliferation and secretion of IFN-γ (FIGS.6D-F, and6J). It was previously shown that an increase in antigen-specific CD8posT cells in cultures with PBMCs was more pronounced when using mAbs with an IgG1 isotype [avelumab, MIH1] when compared to mAbs with an IgG2b isotype [29E.2A3] (Grenga et al. 2016, Clin Transl immunol 5:e83). Therefore, we repeated these experiments using avelumab as an IgG1 blocking mAb. Similar to the findings with the IgG2b mAb 29E.2A3, we did not observe enhanced CD8posT-cell activation by DC-MEL in the presence of avelumab [data not shown]. It was unexpected that the anti-PD-L1 mAbs used in the DC-MEL study, both 29E.2A3 and avelumab, were unable to enhance the activation of Melan-A-specific CD8posT cells by DC-MEL, as both have been described as a blocking mAbs, and have been previously used to enhance activation of antigen-specific CD8posT cells by PBMCs (Grenga et al. 2016, Clin Transl immunol 5:e83). In search for an explanation for this lack of effect, we studied binding of mAbs 29E.2A3 and avelumab to moDCs. Staining of moDCs with mAb MIH1 and sdAb K2 were performed for comparison. We observed that mAb MIH1 was the most efficient in detecting PD-L1 on moDCs followed by sdAb K2, avelumab and mAb 29E.2A3 [FIG.11a]. In fact, in flow cytometry mAb 29E.2A3 was not proficient in staining PD-L1. In contrast, efficient staining of PD-L1 on PD-L1pos293T cells was observed [FIG.11b], suggesting a different sensitivity of mAb 29E.2A3 for binding to PD-L1 on immune cells versus non immune cells. Such differences in immune cell versus tumour cell sensitivity of mAbs used for detection of PD-L1 in immunohistochemistry (IHC) was previously described (Schats et al. 2018, Arch Pathol Lab Med 142:982-991). In conclusion, these results suggest that the inability and low efficacy to bind PD-L1 on moDCs when compared to binding of PD-L1 on non-immune cells explains the lack of effect with mAb 29E.2A3 and avelumab, respectively. Furthermore, the results generated with sdAb K2 in the context of TriMixDC-MEL and DC-MEL-mediated CD8posT-cell activation suggest that in the absence of strong co-stimulatory signals, PD-L1 is a major determinant of T-cell activation. Finally, the data generated with DC-MEL and sdAb K2 provide a rationale for the inclusion of sdAb K2 in DC-based immunotherapy strategies. The inhibitory function of PD-1/PD-L1 interaction during antigen presentation by DCs to T cells is generally recognized, pinpointing this inhibitory pathway as an attractive therapeutic target to enhance the potency of DC-vaccines. Several strategies have been successfully employed in preclinical studies to interfere with PD-1/PD-L1 interactions during antigen presentation by DCs to CD8posT cells. In particular the use of mAbs in combination with DC-vaccination has found its way to the clinic, as evidenced by a number of clinical trials in a range of malignancies (Versteven et al. 2018, Front Immunol 9:394). However, the immune synapse clears and even excludes molecules above a certain size, including mAbs (Cartwright et al. 2014, Nat Commun 5:5479). Therefore, the use of small-sized, blocking PD-1/PD-L1 agents might be more advantageous. Described herein are human PD-L1-specific sdAbs, more specifically sdAb K2, as a PD-1/PD-L1 neutralizing moiety with high target specificity and affinity. Its small size (≈15 kDa) and therefore predicted high potential to penetrate within cell-cell interfaces like immune synapses (Cartwright et al. 2014, Nat Commun 5:5479), make it an interesting candidate for implementation in combination therapy with DCs. We showed that sdAb K2 could shift the balance between stimulatory and inhibitory signals during the early stage of T-cell activation when using DCs with a low stimulatory profile. Although DC-activation, here achieved through delivery of TriMix mRNA, results in enhanced expression of PD-L1, we could not observe a significant increase in CD8posT-cell activation when sdAb K2 was added during the antigen presentation by mature DCs. This might be explained by the fact that activation of moDCs coincides with up-regulation of various co-stimulatory molecules, creating an environment in which co-stimulatory signals supersede co-inhibitory signals. This finding corroborates previous studies in which moDCs of different potency were used to activate allogeneic CD4posT cells in the presence of PD-L1 blocking mAbs (Brown et al. 2003, J Immunol 170:1257-1266), and suggests that blockade of the PD-1/PD-L1 pathway has most impact in conditions of ‘weak’ stimulation. We were unable to show any benefit of adding mAbs to the moDC-CD8posT-cell co-cultures. This is in contrast to other studies reporting on the use of avelumab and/or mAb 29E.2A3 to enhance the activation of human T-cell populations of healthy donors (Grenga et al. 2016, Clin Transl immunol 5:e83; Brown et al. 2003, J Immunol 170:1257-1266). Several reasons can explain this discrepancy. Grenga et al. 2016 (Clin Transl immunol 5:e83) studied interaction between PBMCs and CD8posT cells, rather than moDCs and CD8posT cells, showing that in this setting, activation of virus-specific CD8posT cells was most pronounced in the presence of avelumab when compared to mAb 29E.2A3. The use of viral peptides as antigens is a major difference, as in this case most likely memory CD8posT cells are activated rather than naïve T cells. Brown et al. 2003 (J Immunol 170:1257-1266) used moDCs as stimulator cells, however, performed allogeneic mixed lymphocyte reactions, evaluating CD4posT-cell activation in the presence of the mAb 29E.2A3. In this setting the presence of allogeneic HLA-antigens may serve as a danger signal, also inducing overall T-cell activation, including memory T-cell activation (Merrick et al. 2008, Cancer Immunol immunother 57:897-906). Re-activation of antigen-experienced effector memory T cells was suggested to be the driver of the efficacy of PD-L1/PD1 blockade in human cancer therapy, while activation of CD8poseffector T cells was not reported (Ribas et al. 2016, Cancer Immunol Res 4:194-203). The source of stimulator cells might also contribute to the difference in experimental outcome. We observed that mAb 29E.2A3 was unable to detect PD-L1 in flow cytometry on the moDCs we generated, and that detection of PD-L1 with avelumab was less evident as with mAb MH1 and sdAb K2. Staining op PD-L1 expressed on 293T cells precludes that this observation is a technical artefact, as avelumab, mAbs 29E.2A3 and MIH1 as well as sdAb K2, were able to stain PD-L1 on these cells. The reason for this different sensitivity to PD-L1 expressed on moDCs versus 293T cells is at present unclear, however, provides an explanation as to why de novo activation of antigen-specific CD8posT cells was not observed in the presence of these mAbs in our study. It is conceivable that our moDCs differ from the moDCs used by Brown et al. 2003 (J Immunol 170:1257-1266), as different culture conditions were used [e.g. culture medium]. For sure, the moDCs used in this study are different from the PBMCs used by Grenga et al. 2016 (Clin Transl immunol 5:e83). Further studies are required to assess binding of different mAbs to different PD-L1posimmune and non-immune cell populations, including DCs. Such studies have already been performed with other antibodies in the context of immunohistochemical detection of PD-L1 in tumour tissue and lymph nodes, showing that different antibodies indeed have different propensities to bind PD-L1 on tumour cells versus immune cells, and sometimes even discriminate between lymphocyte-like cells versus DCs (Schats et al. 2018, Arch Pathol Lab Med 142:982-991). In our study, sdAb K2, similar to mAb MIH1, did not make the distinction between PD-L1 expressed on moDCs versus 293T cells. We previously showed that sdAb K2 competes with avelumab for binding to PD-L1. Studies showed that avelumab binds mainly with its VH domain on the strands of the front β-sheet face of the IgV domain of PD-L1, which is different from the epitopes bound by other PD-L1 targeting mAbs, such as durvalumab, atezolizumab and BMS-936559 (Liu et al. 2017, Cell Res 27:151-153; Tan et al. 2018, Protein Cell 9:135-139). As such sdAb K2 is a unique small-sized, biological inhibitor, when compared to other small molecule inhibitors, such as the anti-PD-L1 sdAb KN035 and the non-peptide anti-PD-L1 inhibitors, BMS-202 and BMS-8, which show similar binding to PD-L1 as durvalumab (Tan et al. 2018, Protein Cell 9:135-139; Zhang et al. 2017, Cell Discov 3:17004). We showed that sdAb K2 blocks PD-1/PD-L1 interactions on the protein level and tumour cell-T cell level. We now show that sdAb K2 can also block PD-1/PD-L1 interactions at the immunological synapse created when DCs interact with CD8posT cells. The single domain nature of sdAbs offers interesting perspectives in view of DC-vaccine development. Many protocols are available to deliver tumour antigens and activation stimuli to DCs. Many of these are based on genetic engineering using viral and non-viral vectors (Breckpot et al. 2004, J Gene Med 6:1175-1188; Benteyn et al. 2015, Expert Rev Vaccines 14:161-176). While cloning of classical mAbs or mAb-fragments offers serious challenges, cloning of sdAbs is straightforward, therefore can be easily incorporated into existing DC-engineering protocols. Several of these ex vivo DC-engineering strategies have also been used to specifically engineer DCs in situ, even in the tumour environment (Van Lint et al. 2012, Cancer Res 72:1661-1671; Van Lint et al. 2016, Cancer Immunol Res 4:146-156; Goyvaerts et al. 2013, J Virol 87:11304-11308; Van der Jeught et al. 2018, ACS Nano 12:9815-9829; Verbeke et al. 2019, ACS Nano). The targeted delivery of sdAb K2, and its release in the immunological synapse offers attractive safety considerations compared to systemic mAb or sdAb-administration. It will tip the balance from immune inhibitory to stimulatory signals only between antigen and sdAb K2-engineered DCs and cognate T cells, thereby ensuring increased on-target T-cell responses with little to no off-target T-cell activation. In conclusion, we report on the use of sdAb K2, a versatile PD-L1/PD-1 blocking moiety, to enhance the capacity of DCs to stimulate T-cell activation and cytokine production. Inclusion of sdAb K2 in DC-vaccination protocols may have therapeutic potential in the clinical setting where several technologies to modify DCs for T-cell activation are investigated in the setting of cancer as well as infectious disease. 2.5. sdAb K2 Maintains T-Cell Activation During Interaction with Tumour Cells The PD-1:PD-L1 immune checkpoint axis is a major culprit in the tumour microenvironment. Therefore, we evaluated whether T cells electroporated with mRNA encoding PD-1 and the TCR recognizing gp100280-288in the context of HLA-A2 were hampered in their ability to proliferate and produce IFN-gamma upon interaction with gp100280-288presenting HLA-A2+tumour cells that are PD-L1−or PD-L1+. 2.6. sdAb K2 is a Promising Theranostic The first patient studies with blocking antibodies were correlated to the PD-L1 status of tumours using immunohistochemical staining (IHC) of tumour biopsies, confirming that responses were significantly higher in PD-L1 expressing tumours. Nonetheless, in subsequent studies, responses were also observed in PD-L1 negative cancers, although to a lesser extent (Topalian et al. 2012, N Engl J Med 366:2443-2454). These observations highlight the need for tools that allow assessment of PD-L1 expression, and that can target PD-L1 within the tumour microenvironment to maintain the function of T cells. As sdAb K2 is suited for imaging of PD-L1 and as our in vitro assays suggest it has the potential to activate and maintain the function of CD8+T cells, we studied whether PD-L1 upregulation on 624-MEL cells upon interaction with tumour specific T cells could be visualized in SPECT/CT imaging with99m-Tc sdAb K2, and whether the signal predicted the outcome of therapy with sdAb K2. 2.7 Kidney Uptake of Various huPDL1-Binding Agents. Table 2 lists kidney uptake values for the huPDL1-binding nanobodies of the current invention, and for other huPDL1-binding agents and murine PDL1-binding nanobodies:murine PDL1-binding nanobody C3: Broos et al. 2017 (Oncotarget 8:41932)affibody: Gonzalez et al. 2017 (J Nucl Med 58:1852)adnectins: Donnely et al. 2017 (J Nucl Med doi:10.2967/jnumed.117.199596)macrocyclic peptide: Chatterjee et al. 2017 (Biochem Biophys Res Comm 483:258)ectodomain huPD-1: Maute et al. 2015 (Proc Natl Acad Sci 112:E6506). % IA/g (as also referred to in Legends toFIGS.3,4, and8, and in Examples 1.8 and 2): uptake of the radiolabel in different tissues is expressed by % IA/g or % of injected activity per gram of tissue. If it refers to organs (such as kidneys), it means that at the indicated time point post injection, the mouse was killed, dissected and organs (such as kidneys) were removed. All organs are weighed and radioactivity was counted in a gamma counter. This results in a number of counts per minute (or second). This is arbitrary as it will go down over time with the decay of the radionuclide. Therefore, a standard is also measured in the gamma counter and the counts in the organs (e.g. kidneys) is then correlated to the counts in the standard by which it is extrapolated to the amount of radioactivity that was injected (IA). The uptake in the organs is then expressed as the relative uptake of the injected activity (so % of total amount of injected activity or % IA). This is then divided by the weight of the organs, resulting in a value in % IA/g. This can be higher than 100%, because the kidneys do not weigh a full gram in mice (typically around 100 mg). timepointuptakepostkidneysagentlabellinganimal modelinjection(% IA/g)K2 (huPDL199m-Tcnaive C57BL/6 mice80 min34.02nanobody)K3 (huPDL199m-Tcnaive C57BL/6 mice80 min56.41nanobody)K2 (huPDL199m-Tcathymic nude mice bearing80 min14.40nanobody)MCF7 PD-L1 negative tumorK2 (huPDL199m-Tcathymic nude mice bearing80 min15.23nanobody)MCF7 PD-L1 positive tumorK2 (huPDL199m-Tcathymic nude mice bearing80 min17.22nanobody)MEL624 PD-L1 negative tumorK2 (huPDL199m-Tcathymic nude mice bearing80 min17.10nanobody)MEL624 PD-L1 positive tumorK2 (huPDL168-Ga (sitenaive C57BL/6 mice80 min10.06nanobody)specific coupling)K2 (huPDL168-Ga (randomnaive C57BL/6 mice80 min19.18nanobody)coupling)C3 (muPDL199m-Tcnaive C57BL/6 mice80 min212.00nanobody)C3 (muPDL199m-TcPD-L1 KO mice80 min316.10nanobody)C3 (muPDL199m-TcC57BL/6 mice bearing PD-L180 min114.30nanobody)positive tumor (TC-1 WT)C3 (muPDL199m-TcPD-L1 KO mice bearing PD-L180 min178.00nanobody)knock-out tumor (TC-1 KO)C3 (muPDL199m-TcC57BL/6 mice bearing PD-L180 min197.30nanobody)overexpressing tumor (TC-1 KI)C3 (muPDL199m-TcC57BL/6 mice bearing TC-1 WT80 min205.40nanobody)PD-L1 knock down tumorhuPD-1ectodomain64-CuNSG mice with CT261 hbetweentumor models100 and 125huPDL118-FFemale SCID Beige mice bearing90 min312.69affibodyLOX tumorhuPDL118-FFemale SCID Beige mice bearing90 min254.59affibodySUDHL6 tumorhuPDL118-Fmice implanted with L2987 and90 mincpm/ID/adrectinHT-29 xenograftstissue weighthuPDL164CuNSG mice bearing hPD-L11 hbetweenmacrocyclicpositive and negative tumors30 en 40peptideWL-12 3. Site-Specific Radiolabelling Immune checkpoints such as Programmed death-ligand 1 (PD-L1) limit the T-cell function, and tumour cells have developed this receptor to escape the anti-tumour immune response. Monoclonal antibody-based treatments have shown long-lasting responses, but only in a subset of patients. Therefore, there is a need to predict response to treatments. In support of this, an IVD-based probe to assess human PD-L1 (hPD-L1) expression using PET imaging was developed by site-specific coupling of the anti-PD-L1 sdAb to the NOTA-chelator for68Ga labelling, or to the RESCA-chelator for [18F]AlF labelling. As a comparison study, anti-PD-L1 IVD was also coupled to the NOTA-chelator by the random coupling strategy, since this strategy is already implemented in the production of other sdAbs in clinical trials. The anti-hPD-L1 sdAb K2 with a sortag-motif (sortase A amino acid substrate motif) at its C-terminal was site-specifically coupled to a bifunctional chelator (BFC) via the Sortase A enzyme coupling reaction. BFCs were synthesized by attaching p-SCN-Bn-NOTA or RESCA-(tBu)—COOH to a GGGYK peptide. Site-specifically modified anti-hPD-L1 sdAb K2 were purified by incubation with 150 M EDTA solution, by IMAC and by size exclusion chromatography (SEC). For random functionalization, 6×Histidine-tagged anti-hPD-L1 sdAb K2 in 0.05 M sodium carbonate buffer, pH 8.7 was added to a twenty-fold excess of the p-SCN-Bn-NOTA BFC, pH adjusted to 8.5-8.7 with 0.2 M Na2CO3. After 2 h incubation at room temperature (RT), the pH of the reaction mixture was lowered to pH 7.4 by adding HCl 1N. The NOTA-(anti-hPD-L1 sdAb K2) protein solution was loaded on a SEC column. The collected fractions containing monomeric NOTA-anti-hPD-L1 sdAb K2 protein were pooled. Modified anti-hPD-L1 sdAb K2 were characterized by Mass Spectrometry (ESI-Q-TOF), SDS-PAGE and Western Blot. NOTA-(anti-hPD-L1 sdAb K2), site-specific and random, were labelled with 68Ga and RESCA-(anti-hPD-L1 sdAb K2) with [18F]AlF. Radiochemical purity (RCP) was assayed by SEC and iTLC. Stability of site-specifically radiolabelled probes was evaluated in vitro. Binding capacity of [68Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2) was evaluated in vitro on PD-L1 positive mel624 cells and compared with the randomly68Ga-labelled anti-hPD-L1 sdAb K2, while affinity and specificity were tested on PD-L1 negative cells and on PD-L1 positive cells in presence of a 100 fold excess of unlabelled sdAb K2. In vivo stability (Blood curve and metabolization study) was performed with both random and site-specific [68Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2), as well as with site-specific [67Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2) by analyzing blood and urine samples from different time points. In vivo biodistribution in C57BL/6 mice was performed with both random and site-specific [68Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2), as well as with site-specific [67Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2) and [18F]AlF-RESCA-(anti-hPD-L1 sdAb K2). In vivo tumour targeting studies were performed with both random and site-specific [68Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2) in xenografted-athymic nude mice bearing PD-L1 positive cells, or PD-L1 negative cells as a control. In vivo tumour targeting was also performed with site-specific [67Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2) in xenografted-athymic nude mice bearing PD-L1 positive cells. PET/CT and SPECT/CT imaging of tumour-bearing mice was performed with site-specific [68Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2) and [67Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2) respectively. Site-specifically functionalized anti-hPD-L1 sdAb K2 with NOTA and RESCA were obtained with high purity (≥99%) in 56% and 59% yield respectively. Functionalization did not affect affinity nor specificity. Randomly functionalized anti-hPD-L1 sdAb K2 was obtained in 52% yield. Radiolabelling of both random and site-site-specific NOTA-(anti-hPD-L1 sdAb K2) with68Ga was performed at RT for 10 min at pH 4.4-4.7 in a 80% decay corrected radiochemical yield (DC-RCY) and ≥99% RCP. Apparent molar specific activity of 85 GBq/μmol was obtained for the site-specifically radiolabelled sdAb K2, and 63 GBq/μmol for the randomly radiolabelled sdAb K2. Over 4 hours, the site-specifically radiolabelled probe and metal complex were stable in injection buffer and in presence of DTPA excess (≥99% RCP). RCP after 1 hour at 37° C. in human serum was ≥94%. Site-specific radiolabelling with67Ga was performed at rt for 10 min at pH 4.4-4.7 in a 86% DC-RCY, ≥95% RCP with an apparent molar specific activity up to 40 GBq/μmol. The radiolabelled probe was stable in injection buffer over 17 h (≥98% RCP) and in human serum at 37° C. over 4 h (≥0.95% RCP). In vivo metabolization studies with site-specific [67Ga]Ga-NOTA-(hPD-L1) showed that the probe was ≥95% intact in the blood and ≥90% intact in the urines after 2 hours. In vivo tumour targeting and biodistribution studies with both random and site-specific [68Ga]Ga-NOTA-(anti-hPD-L1sdAb K2) revealed high tumour uptake of (3.664±0.764) % IA/g organ for the site-specific compared to (1.551±0.467) % IA/g organ for the random (seeFIG.10B). No unspecific organ targeting was observed, except in the kidneys (seeFIG.10B) and excretion to the bladder (expected route of excretion). Compared to random68Ga-labelled sdAb K2, kidney accumulation of site-specific68Ga-labelled sdAb K2 is lower whereas tumour uptake of site-specific68Ga-labelled sdAb K2 is higher, therewith increasing the signal/noise ratio and increasing tumour-selective labelling. Similar selectivity of site-specific68Ga-labelled sdAb K2 compared to random68Ga-labelled sdAb K2 was observed on PD-L1+ cells (seeFIG.10A). In vivo tumour targeting and biodistribution studies profiles obtained with site-specific [67Ga]Ga-NOTA-(anti-hPD-L1 sdAb K2) were similar as for68Ga labelled probe (seeFIG.10C). Tumour uptake was slightly lower as previous experiments, which can be explained by the different position of the tumour (neck region instead of leg) and a longer tumour growing period in vivo, leading to necrotic tumours. Quantification of SPECT/CT scans with67Ga-labeled allowed to quantify tumour uptake, giving similar results as the ex-vivo measurement: 0.328% of total injected activity quantified from scan compared with 0.350% of total injected activity calculated dissected tumour. Radiolabelling of RESCA-(anti-hPD-L1 sdAb K2) with [18F]AlF was performed at rt for 12 min at pH 4.4-4.7 in a 29% DC-RCY and with a RCP≥99%. The radiolabelled probe was stable over 2.5 hours in injection buffer (RCP≥98%). Biodistribution in healthy animals was similar as for [68Ga]Ga-NOTA-(hPD-L1), except for slightly higher bone uptake. The Sortase enzyme-mediated labelling approach thus allowed to obtain a site-specifically functionalized anti-hPD-L1 sdAbs, which could be easily radiolabelled with67Ga,67Ga or [18F]AlF. [68Ga]Ga-NOTA-(anti-hPD-L1 sdAbs) proved to specifically target the hPD-L1 receptor in vivo and the targeting experiment will be repeated with [18F]AlF-RESCA-(anti-hPD-L1 sdAbs). SPECT/CT images obtained with 67Ga-labelled probe proved to be quantifiable. 4. Detection of Human PD-L1 Induced by IFN-γ in Xenograft Tumour Models by sdAb K2 Following validation in two PDL1-engineered tumour cell mouse models, we evaluated whether sdAb K2 can be used to detect PD-L1 expression in response to IFN-γ. The 938-MEL model was used as we observed in flow cytometry that in vitro treatment of 938-MEL cells with 100 IU/mL IFN-γ leads to upregulation of PD-L1 (FIG.15A). We next injected recombinant IFN-γ in 938-MEL tumours grown in athymic nude mice and used 99m Tc-sdAb K2 and SPECT/CT imaging to evaluate PD-L1 expression (FIG.15B). Tumours of on average 150 mm3 were injected with PBS (negative control) or 104 IUs IFN-γ. One day later, we performed SPECT/CT imaging, showing detection of PD-L1 in the tumour of IFN-γ but not of PBS-treated mice (FIG.15C). Furthermore, ex vivo γ-counting showed higher uptake of 99m Tc-sdAb K2 in mice treated with IFN-γ (0.55±0.08% IA/g) compared to mice treated with PBS (0.28±0.02% IA/g) (FIG.15D). Evaluation of PD-L1 expression on tumour cells using flow cytometry confirmed higher PD-L1 expression on IFN-γ-treated tumours compared to PBS-treated tumours, although PD-L1 expression levels were low (FIG.15E). 5. sdAb K2 Competes with Avelumab for Binding to PD-L1 and has PD-1:PD-L1 Blocking Capacity We showed that sdAb K2 serves as a potential diagnostic tool to detect PD-L1 expression levels in vivo on tumour cells and as such might select patients for anti-PD-L1 treatment. We next wondered whether sdAb K2 also has therapeutic potential and evaluated whether sdAb K2 is able to inhibit the PD-1:PD-L1 interaction leading to enhanced T-cell activity. We showed by SPR that sdAb K2 recognizes the same epitope on PD-L1 as avelumab (FIG.16A). Moreover, sdAb K2 is able to inhibit the interaction between PD-1:PD-L1 with an IC50 of 8.5 nM. In the same assay, the IC50 value of avelumab was 4 nM, whereas both controls, R3B23 and trastuzumab, did not influence the PD-1:PD-L1 interaction (FIG.16B). 6. sdAb K2 Restores the Tumour Cell Killing Ability of Activated PBMCs We explored the effect of adding sdAb K2 to co-cultures of activated PBMCs and tumour cells. First, we evaluated the expression of PD-1 and PD-L1 on CD8POS T cells, present within the pool of PBMCs stimulated with a cocktail of anti-CD3 antibodies and IL-2, as these cells are critical to mediate tumour cell killing. We showed using flow cytometry that both PD-1 and PD-L1 were upregulated on CD8POS T cells, thereby confirming activation of these cells (FIG.17A). The activated cells were added to PD-L1pos 624-MEL cells that were lentivirally engineered to express eGFP (FIG.17B) and that were grown in a 3D spheroid. In the absence of PD-1:PD-L1 blocking moieties, we observed that the amount of eGFPPOS tumour cells, measured as green objective area, increased in time, or in other words were not destroyed sufficiently by activated T cells, confirming the inhibitory role of PD-L1 on T cell-mediated tumour cell killing (FIG.17C). Addition of avelumab or sdAb K2 to PD-L1POS tumour cells and stimulated PBMCs enhanced tumour cell killing when compared to addition of a control mAbs or R3B23, as measured as reduction in green objective area (FIG.17D-E). The effect of adding avelumab on tumour cell killing could be observed at 80 hours (FIG.17D), while the effect of sdAb K2 was observed in the first hours of culture (FIG.17E). Hence, sdAb K2 showed early and short therapeutic activity, while avelumab showed a more durable blocking activity. To enhance the blocking activity, we evaluated329the effect of combined sdAb K2 and avelumab treatment, showing more efficient tumour cell killing (after 70 hours) compared to addition of avelumab or sdAb K2 separately (FIG.17F). Remedying the short action of sdAb K2, we tested the effect of repeated administrations of sdAb K2 with 24-hour intervals. This resulted in efficient tumour cell killing by activated PBMCs after 50 hours (FIG.17G), supporting the therapeutic potential of sdAb K2. 7. Discussion In this study we showed that the sdAb, designated sdAb K2, is able to detect human PD-L1 expression levels in the tumour microenvironment and to block PD-L1 on tumour cells resulting in enhanced T-cell activity. sdAb K2 binds with nanomolar affinity to human PD-L1 and can be used as a diagnostic to detect PD-L1 expression in the tumour as fast as one hour after injection. PD-L1 expression could even be detected after intratumoural administration of IFN-γ, which led to in situ upregulation of PD-L1, although expression levels remained low. Moreover, we showed that sdAb K2 has therapeutic potential as it exhibits an IC50 of 8.5 nM to block PD-1:PD-L1 interactions and releases the break on antigen-specific TCR signalling and on tumour killing activity in vitro. Nowadays, in clinical trials, PD-L1 expression is mainly evaluated by IHC, which has some limitations. Staining of fixed selected tissue samples does not allow assessment of heterogenic expression of tumour markers, or the dynamic PD-L1 expression during treatment. Molecular imaging is a good alternative to assess PD-L1 expression, as this non-invasive method can show regional differences within the tumour environment and can assess PD-L1 expression in metastatic lesions. Here, we showed that sdAb K2 has several properties to make it an interesting diagnostic. Ideal radiotracers combine fast renal clearance and efficient tumour penetration with good affinity for their target, resulting in high tumour-to-background ratios shortly after tracer administration. We showed that 99m Tc-sdAb K2 fulfils these requirement since administration in healthy C57BL/6 mice revealed little to no signals in all organs except in the kidneys and urinary bladder, which is due to the renal uptake and elimination because of their small size (Chakravarty et al. 2014, Theranostics 4:386-398). Noteworthy, the uptake of 99m Tc-sdAb K2 in the kidneys was much lower compared to 99m Tc-R3B23, the sdAb used as a negative control, and to our knowledge any other sdAb that was labelled in a similar fashion. This low kidney retention makes sdAb K2 particularly suited as a radiotracer, since such important decrease in kidney retention not only lowers the irradiation burden for the patient but also improves the assessment of lesions in the vicinity of the kidneys. This can be useful to assess patients with renal cell carcinoma for expression of PD-L1, as these patients can derive benefit from such treatments (Alsaab et al. 2017, Front Pharmacol 8:561). 99m Tc-sdAb K2 showed intense and specific uptake in two human PD-L1-expressing tumour models, melanoma and breast cancer, with tumour-to-blood ratios of 20.2 and 8.9, respectively. Moreover, PD-L1 expression could be detected after intratumoural injection of IFN-γ, leading to elevated, albeit still low, PD-L1 expression levels on tumour cells, as confirmed with flow cytometry. In all imaging studies, high tumour-to-background uptake levels could be obtained as fast as one hour after injection. When translated to patients, this would allow short, same-day imaging procedures, very similar to the current daily practice with 18F-FDG (Vaneycken et al. 2011, FASEB J 25:2433-2446). The absolute tumour uptake we observed with sdAb K2 is at the same level compared to other studies using sdAbs that target tumours (Xavier et al. 2013, J Nucl Med 54:776-784; Xavier et al. 2019, Mol Imaging Biol). Although absolute tumour uptake for sdAbs is generally lower than what can be obtained with mAb, the contrast that can be obtained at early time points is much higher, due to the very fast clearance of the unbound tracer. For future clinical translation, the here proposed SPECT tracer will be further engineered into a clinical PET-tracer, similar to what was done for other sdAb translations (Keyaerts et al. 2016, J Nucl Med 57:27-33; Xavier et al. 2013, J Nucl Med 54:776-784; Xavier et al. 2019, Mol Imaging Biol). Other research groups have as well developed radiotracers for PD-L1 imaging using both mAbs (Bensch et al. 2018, Nat Med 12:1852-1858; Lesniak et al. 2016, Bioconjug Chem 27:2103-2110) or smaller proteins (Chatterjee et al. 2017, Biochem Biophys Res Commun 483:258-263; Donnelly et al. 2017, J Nucl Med 59:529-535; Niemeijer et al. 2018, Nat Commun 9:4664) of which some have entered clinical testing. Bensch et al used 89Zr-labelled atezolizumab, a clinically approved therapeutic mAb, for molecular imaging in cancer patients (Bensch et al. 2018, Nat Med 12:1852-1858). A better correlation between PET images and clinical responses compared to IHC was reported. However, optimal tumour-to-blood ratios were only obtained on day 7 after injection (Bensch et al. 2018, Nat Med 12:1852-1858). This time point could be tangibly reduced to 5 days using a 89Zr-labelled-heavy chain-only antibody KN035 (i.e. an anti-PD-L1 sdAb fused to an Fc domain), which is smaller (80 kDa) than a full antibody (150 kDa) but still is substantially larger than sdAbs such as sdAb K2 (15 kDa). However, the tumour-to-blood ratios reported were low, i.e. 1.1 (Li et al. 2018, Mol Pharm 15:1674-1681). The 18F-labelled adnectin 18F-BMS-986192 has a size of about 10 kDa, and is therefore at least in terms of size closer to sdAbs. This compound could visualize PD-L1POS tumours with a 3.5-fold higher uptake in PD-L1POS versus PD-L1NEG tumours using PET imaging in mice. However, kidney uptake of 18F-BMS-986192 was relatively high (Donnelly et al. 2017, J Nucl Med 59:529-535). This compound was as well recently evaluated in cancer patients (non-small-cell lung cancer). Tracer uptake in the tumour correlated with PD-L1 expression levels on tumour cells evaluated with IHC. However, a subset of tumours showed low PD-L1 expression by IHC but relatively high uptake with 18F-labelled adnectin, which could be explained by the heterogeneity of PD-L1 in the lesion. Furthermore, response rates correlated with tracer uptake, with responders showing higher tracer uptake compared to non-responders (Niemeijer et al. 2018, Nat Commun 9:4664). These observations make us believe that small imaging agents, such as the here presented sdAb K2, can be used as a diagnostic tool in cancer patients. Indeed, sdAb K2 is able to image PD-L1 with high contrast levels as fast as one hour after injection, which is much faster than imaging with 89Zr-labelled atezolizumab. Secondly, because of its small size sdAb K2 is able to efficiently penetrate tumours resulting in higher tumour-to-blood ratios compared to compound KN035 (8.9 and 20.2 for sdAb K2 compared to 1.1 for KN035). Finally, sdAb K2 is able to detect PD-L1 expression levels with higher contrast compared to similar-sized 395 18F-labelled adnectin (tumour-to-blood ratios of 8.9 and 20.2 for sdAb K2 versus <3 for the adnectin at the same time point) and with lower kidney retention. Besides its diagnostic value, we furthermore evaluated the therapeutic value of sdAb K2. The use of sdAbs for therapy exhibits some advantages compared to mAbs. sdAbs are 10 times smaller than mAbs and are therefore better suited for fast and homogenous tumour penetration. As the PD-(L)1 immune checkpoint is mainly relevant in the tumour microenvironment rather than in other immune organs (Zou et al. 2016, Sci Transl Med 8:328rv4), this could be a key characteristic for optimal therapeutic effect in larger, difficult-to-penetrate tumours, which was observed for the HAC-I variant (10 kDa; KD=100 pM; IC50=210 pM) for example. This smaller blocking moiety induced equal tumour reduction compared to mAbs targeting human PD-L1 when treating smaller tumours, but when larger tumours were treated it appeared that the mAb lost its therapeutic efficiency whereas the HAC-I variant did not (Maute et al. 2015, PNAS USA 112:E6506-E6514). Also the previously described sdAb-Fc compound KN035 enhanced tumour cell killing in a xenograft model (Zhang et al. 2017, Cell Discov 3:17004). However, sdAbs that target human PD-L1 have not yet been studied in their monovalent format in a therapy setting. We were able to show that sdAb K2 binds to the same epitope on PD-L1 as the FDA-approved antibody avelumab and is able to block the PD-1:PD-L1 interaction in a similar magnitude as avelumab in a human antigen-specific T cell assay, even though the IC50 value of sdAb K2 was slightly higher. This difference in IC50 can be explained by the bivalent format of avelumab, which renders two binding places for avelumab compared to one for sdAb K2. When evaluating both compounds in the 3D tumour cell killing assay, we observed that tumour cell killing started rapidly after sdAb K2 addition, whereas for avelumab the effect was only observed after 80 hours. This may be explained by differences in valency, IC50 as well as diffusion between both agents. Whereas sdAb K2 is small and should be able to rapidly bind to its target, it likely also rapidly detaches from its target. In contrast, avelumab is larger and probably reaches its target later but the higher avidity due to the bivalent format results in better off-rates and longer retention times. Hence, as a therapeutic, repeated administration of sdAb K2 could be necessary to obtain the same effect as avelumab. Alternatively, sdAb K2 could be modified to a bivalent format to optimize its effect. We could already confirm in vitro that adding sdAb K2 every 24 hours had the same effect on activated PBMC-mediated tumour cell killing compared to adding one dose of avelumab. However, it remains to be shown if this could also improve clinical outcome. Exploiting their differences in pharmacokinetics and avidity we moreover demonstrate that combinatorial treatment with sdAb K2 and avelumab results in a superior antitumour killing effect. Further research to determine the exact value of such a combination approach in an in vivo tumour setting is warranted. Taken together, these data show that sdAb K2 holds promise as a small antagonistic therapeutic compound targeting human PD-L1.
120,116
11858961
DETAILED DESCRIPTION Which carbohydrates, or glycans, are attached to a glycoprotein is important to understanding the pharmacokinetics, immunogenicity, and potential therapeutic effectiveness of the glycoprotein. Accordingly, removing the glycans and analyzing them to determine which glycans are attached to a given glycoprotein has become an important aspect of quality control for glycoproteins, such as antibodies and other biologics, intended for therapeutic use. Further, analyzing the aglycosylated glycoprotein after its glycans have been removed has also become important for confirming the composition of the aglycosylated protein. Unfortunately, some glycoproteins, especially some antibody and antibody-derived therapeutics, are resistant to enzymatic deglycosylation by standard protocols, especially those involving release of N-glycans by the exemplar enzyme PNGase F. Since there is no way to determine in advance whether any particular glycoprotein of interest is easy or hard to deglycosylate by enzymatic release of the glycans, it is generally determined empirically by subjecting the glycoprotein to an enzymatic deglycosylation protocol intended to remove N-glycans, O-glycans, or both, known to be present on the glycoprotein and analyzing the molecular weight of the protein following the procedure to determine whether the molecular weight indicates that the N-glycans, O-glycans, or both, present on the glycoprotein have been removed or that some remain attached. Glycoproteins are typically denatured prior to enzymatic deglycosylation so that the enzyme has access to glycosylation sites which otherwise might be hidden or blocked by the protein's secondary or tertiary structure. Often, a detergent is used to solubilize the glycoprotein as part of the denaturation step. The detergent may also participate in denaturation of the glycoprotein, and may be referred to as a denaturant in addition to being referred to as a detergent. It would be convenient to have additional detergents that can work across a range of glycoproteins from those relatively easy-to-deglycosylate to those relatively hard-to-deglycosylate in an attempt to reduce the number of glycoproteins for which protocols have to be adopted. Experiments were performed in attempts to find detergents that would be effective in enzymatic deglycosylation protocols over a range of glycoproteins. As antibodies and antibody-derived glycoproteins are increasingly important as therapeutic agents, some of our studies focused on examining the deglycosylation of a selection of some of the currently-available antibody and anti-derived agents. In initial studies underlying the present invention, we found that fatty acid salts with alkali metal cations, such as sodium laurate, were good detergents for deglycosylation of many glycoproteins. We further found that substituting an exemplar quaternary ammonium cation, tetramethyl ammonium, in place of the alkali metal, sodium, in sodium laurate, resulted in deglycosylation about as good as that when using sodium laurate for many glycoproteins, but in markedly better deglycosylation of some glycoproteins. It was found, for example, that in the presence of the exemplar quaternary ammonium cation fatty acid salt, a deglycosylation protocol resulted in the release of 21% more glycan from IgM than did the same protocol using the fatty acid salt, but with an alkali metal cation rather than the quaternary ammonium cation. Thus, we found that quaternary ammonium fatty acid salts are superior to fatty acid salt detergents for denaturing glycoproteins for enzymatic deglycosylation protocols, and particularly for deglycosylation protocols for antibodies and antibody-derived glycoproteins. References herein to “quaternary ammonium carboxyl salts” or “quaternary ammonium fatty acid salts” or “quaternary ammonium fatty acid detergents” refer to compounds with a quaternary ammonium cation, and an aliphatic chain attached to a carboxylate anion. Surprisingly, when we tested an exemplar quaternary ammonium cation-sulfate detergent, tetramethylammonium dodecyl sulfate, with a sulfate anion replacing the laurate of the corresponding quaternary ammonium cation fatty acid detergent, we got markedly better results than were obtained with the exemplar quaternary ammonium fatty acid salt in deglycosylating glycoproteins. The detergents were tested on a recombinant fusion protein Ziv-aflibercept, a cancer therapeutic sold under the name ZALTRAP®. To permit ready comparison, the glycans released by the exemplar quaternary ammonium-sulfate detergent, tetramethylammonium dodecyl sulfate, were treated as representing 100% release of glycans from Ziv-aflibercept, and that amount was compared to the amount of glycans released from Ziv-aflibercept using the same protocol and the corresponding exemplar quaternary ammonium fatty acid salt detergent, tetramethylammonium dodecyl laurate. Use of the quaternary ammonium fatty acid salt detergent resulted in the release of 80-90% of the amount of glycans released from ZALTRAP® by the quaternary ammonium-sulfate detergent. Tests with other hard-to-deglycosylate glycoproteins confirmed that the exemplar quaternary ammonium—fatty acid detergent tested was not as successful at deglycosylating such glycoproteins as the exemplar quaternary ammonium-sulfate detergent. Accordingly, we concluded that quaternary ammonium-sulfate detergents are surprisingly better detergents for denaturing glycoproteins for enzymatic deglycosylation protocols than are either quaternary ammonium—fatty acid detergents or fatty acid detergents. Sodium dodecyl sulfate (“SDS,” also known as sodium lauryl sulfate) is a detergent widely used to denature and solubilize proteins, particularly before subjecting them to electrophoresis, and is commonly used to denature glycoproteins in deglycosylation protocols. As a denaturant for deglycosylation applications, SDS is often used in combination with a non-ionic surfactant, typically NP-40, after the denaturing step but prior to adding the enzyme to prevent denaturation of the enzyme. A series of studies was conducted to determine how the exemplar quaternary ammonium-sulfate detergent, tetramethyl ammonium dodecyl sulfate, compared to SDS. In the studies reported herein, nine exemplar glycoproteins or mixtures of glycoproteins were subjected to enzymatic deglycosylation following denaturing using either an exemplar quaternary ammonium-sulfate detergent, or SDS. To allow ready comparison, the amount of glycans released from each glycoprotein or mixture by digestion following denaturing with tetramethyl ammonium lauryl sulfate was considered to be a 100% release, and that amount was then compared to the amount released from the same glycoprotein or glycoprotein mixture following denaturion with SDS. Surprisingly, for four out of nine of the glycoproteins or glycoprotein mixtures, denaturing with SDS resulted in the release of significantly fewer glycans than did the use of the exemplar quarternary ammonium sulfate detergent, and for a mixture of human IgGs, denaturing with SDS resulted in the release of almost 25% less glycan than did denaturing with the quarternary ammonium sulfate detergent. For a mixture of hIgM antibodies, denaturing with SDS resulted in the release of 18% less glycan than did denaturing with the quarternary ammonium sulfate detergent. For the therapeutic biologic drug ORENCIA®, denaturing with SDS resulted in the release of almost 15% less glycan than did denaturing with the quarternary ammonium sulfate detergent. For three of the nine glycoproteins or mixtures, using SDS to denature the glycoprotein resulted in about the same release of glycan compared with the amount released following denaturation of the same glycoprotein with the quarternary ammonium sulfate detergent. Only in the case of one glycoprotein did denaturing with SDS result in the release of more glycan than did the use of quarternary ammonium sulfate detergent, and in that case the difference was less than 10%. The results show that the exemplar quarternary ammonium sulfate detergent provided generally better or equal results to using SDS in the deglycosylation protocol, and in many cases, provided surprisingly better results compared SDS, both for some hard-to-denature glycoproteins and for mixtures of glycoproteins. As noted above, enzymatic deglycosylation protocols in which SDS is used as a detergent typically recite the addition of a non-ionic detergent prior to introducing the enzyme to avoid denaturing it. Studies with an exemplar quaternary ammonium-sulfate detergent revealed that no non-ionic detergent was needed. Indeed, the studies showed that the presence of a non-ionic detergent is preferably not used. Thus, embodiments of the inventive methods allow eliminating a reagent used in typical protocols. Given the results with an exemplar quarternary ammonium sulfate detergent, it is believed that other detergents which provide a quaternary ammonium cation, an aliphatic chain, and a sulfate anion will also be surprisingly useful detergents for denaturing glycoproteins compared to detergents with an aliphatic chain and a sulfate anion, but a counterion different from a quaternary ammonium cation or a tertiary ammonium cation, as further discussed below. It is further believed that, given the structural and functional similarity of a sulfonate anion to a sulfate anion, that detergents with a quarternary ammonium cation, and an aliphatic chain covalently attached to a sulfonate anion will also be surprisingly useful detergents for the same purposes. Given the difference in charge between the sulfate and the sulfonate anions, we believe that, between detergents differing only between a sulfate versus a sulfonate anion, the one with the sulfate anion will be the better detergent. Further, given the structural and functional similarity of a tertiary ammonium cation to a quaternary ammonium cation, we expect tertiary ammonium cations to form effective detergents with sulfate or sulfonate anions. In some embodiments, the detergents have a quarternary ammonium cation, an aliphatic chain, and a sulfate anion. In some embodiments, the detergents have a quarternary ammonium cation, an aliphatic chain, and a sulfonate anion. In some embodiments, the detergents have a tertiary ammonium cation, an aliphatic chain, and a sulfate anion. In some embodiments, the detergents have a tertiary ammonium cation, an aliphatic chain, and a sulfonate anion. As noted in the Background, once the glycoproteins or glycopeptides are denatured, they are frequently subjected to enzymatic digestion to release N-linked carbohydrates as glycosylamines, which are then typically labeled with an amine-reactive dye. Primary and secondary ammonium salts are less preferred for use in the inventive methods because they could compete with glycans released as glycosylamines following enzymatic digestion and reduce the availability of the glycosylamines for labeling. Tertiary and quaternary ammonium sulfate and sulfonate detergents are expected to be useful for denaturing the glycoprotein but not to react with glycosylamines released from the glycoproteins or glycopeptides of interest by enzymatic digestion. To avoid constant repetition of “glycoprotein or glycopeptide,” references to deglycosylation or analysis of a “glycoprotein” herein encompass deglycosylation or analysis of a “glycopeptide” unless otherwise required by context. Ammonium Cation Sulfate and Sulfonate Detergents As reported above, an exemplar quaternary ammonium sulfate detergent, was surprisingly more effective in denaturing glycoproteins than a like quaternary ammonium detergent based on a fatty acid, and then the commonly used detergent SOS. These results indicate that effective detergents can be made following the formula of the following formula: R—Y−-⋅C+, in which:  Formula 1R is a saturated or unsaturated aliphatic group with 8-22 carbon atoms;Y−is sulfate or sulfonate;C+is a quaternary ammonium cation or a tertiary ammonium cation; and,the dot indicates that the cation is ionically, not covalently associated, with the Y−. In preferred embodiments, the aliphatic group is saturated. In some embodiments, Y−is sulfate. In some embodiments. Y−is sulfonate. In some embodiments in which Y−is a sulfate or sulfonate, the aliphatic group is 8-20, 9-19, 9-18, 9-17, 9-16, 10-16, 10-15, 10-15, 11-14, 11-13, or 12 carbon atoms in length, with each succeeding range or number stated being more preferred than the one preceding it. An aliphatic group of twelve carbons is particularly preferred. The aliphatic group is preferably straight. In some embodiments, it can be in a ring configuration. The aliphatic group is covalently attached to the Y−. As used herein, the phrase “ammonium sulfate detergent” or “ammonium sulfonate detergent” refers to a detergent of Formula 1. As used herein, the phrases “quaternary ammonium sulfate detergent” and “quaternary ammonium sulfonate detergent” refer to a detergent of Formula 1 in which C+is a quaternary ammonium cation. As used herein, the phrases “tertiary ammonium sulfate detergent” and “tertiary ammonium sultanate detergent” refer to a detergent of Formula 1 in which C+is a tertiary ammonium cation. Quaternary ammonium cations have the advantage of maintaining their positive charge independent of the pH of their environment, whereas the charge of primary, secondary and tertiary ammonium cations can vary according to the surrounding pH. It is anticipated that most deglycosylation procedures and other denaturing procedures will be conducted at pHs at which tertiary ammonium cations will maintain their positive charge and can be useful as cations of compounds of Formula 1. For example, the ammonium cation can be a tertiary ammonium cation, such as trimethyl ammonium, triethyl ammonium, tripropyl ammonium or tributyl ammonium. Primary and secondary ammonium cations are expected to be less useful as cations for detergents, as the nitrogen may be accessible to react with amine-reactive dyes or other reagents. In some embodiments, the cation is a quaternary ammonium cation. As used herein, “quaternary ammonium cation” refers to a moiety having the formula N+1R1R2R3R4, wherein each R can be the same or different and are chosen from aryl or alkyl, can be saturated or unsaturated, can be unsubstituted or substituted, may contain atoms other than carbon or hydrogen in the chain or ring or attached to the chain or ring, including carbon-bonding substituents such as sulfur, oxygen, nitrogen, boron, or a halogen, and functional groups containing any of these, and can be another quaternary group. In some preferred embodiments, the quaternary ammonium cation is selected from the group consisting of: Cations with a smaller number of atoms are generally preferred over cations with a larger number of atoms. In some embodiments, the cation can instead be a tertiary ammonium cation. As used herein, a “tertiary ammonium cation” refers to a moiety having the formula HN+1R1R2R3, wherein R1, R2, and R3, can be the same or different and are chosen from aryl or alkyl, can be saturated or unsaturated. can be unsubstituted or substituted, may contain atoms other than carbon or hydrogen in the chain or ring or attached to the chain or ring, including carbon-bonding substituents such as sulfur, oxygen, nitrogen, boron, or a halogen, and functional groups containing these, and can be another tertiary group. Fatty acid salts of ammonium cations which are not commercially available can be synthesized by reacting the fatty acid of choice with the hydroxide conjugate base. For example, tetramethylammonium laurate can be synthesized by reacting lauric acid with tetramethyl ammonium hydroxide and we made tetraethyl ammonium laurate, tetrapropyl ammonium laurate, and tetrabutyl ammonium laurate by the same process. The hydroxide conjugate bases of some, if not all, of the quaternary ammonium cations listed are commercially available. Any that are not can be synthesized by methods well known in the art, such as reacting a salt of the quaternary ammonium cation with a strong base. For example, tetramethyl ammonium hydroxide is typically made by mixing tetramethylammonium chloride and potassium hydroxide in dry methanol. Ammonium cation sulfate and sulfonate detergents can be made by any convenient method known in the art, such as by acid based neutralization, as discussed above, or by ion exchange. A procedure for preparing an exemplar quaternary ammonium sulfate cation detergent by ion exchange is set forth in the Examples. Combinations of ammonium salts and periodic table group 1 detergents It is believed that sulfate or sulfonate detergents having a cation of an element of periodic table group 1 can be used in combination with quaternary ammonium cations and tertiary ammonium cations to form unexpectedly powerful detergents for denaturing glycoproteins for deglycosylation. The quaternary ammonium cations or tertiary ammonium cations can be contributed by, for example, a quaternary ammonium fatty acid salt. These detergents comprising elements in periodic table group 1 have the following formula: R—Y-⋅M+, in which:  Formula 2R is a saturated or unsaturated straight chain aliphatic group with 8-13 carbon atoms;Y− is a sulfate or sulfonate anion;M+ is a cation of an element of periodic table group 1; and,the dot indicates that the cation is ionically, rather than covalently, associated with the Y− anion. It is believed compounds of this formula will be good detergents to be mixed with one or more quaternary or tertiary ammonium cation salts, or quaternary or tertiary ammonium cation cations, for denaturing glycoproteins or glycopeptides in deglycosylation protocols. In preferred embodiments, the aliphatic group is saturated. In preferred embodiments, the aliphatic group is 10-12 carbons in length. In some embodiments, Y− is a sulfate anion. In some embodiments, Y− is a sulfonate anion. For convenience of reference, compounds of Formula 2 may also sometimes be referred to herein as “M+ detergents.” Given the structural similarity of tertiary ammonium salts to quaternary ammonium salts, it is also expected that tertiary ammonium salts can be used with a M+ detergent of Formula 2 to achieve similar results. An excess of quaternary or tertiary ammonium cations can be provided by any convenient means known in the art. Primary and secondary ammonium salts are less preferred because they could compete with glycans released as glycosylamines during a subsequent enzymatic digestion and thus would require an additional cleanup step in a deglycosylation protocol to remove them after the denaturing step but before the enzymatic digestion step. In some embodiments, the solution contains quaternary ammonium cations. In some embodiments, the solution comprises a molar excess of quaternary ammonium cations compared to the compound of Formula 2. In some embodiments, the solution contains tertiary ammonium cations. In some embodiments, the solution contains a molar excess of tertiary ammonium cations to the compound of Formula 2. Use of the mixtures and detergents described above allows methods of releasing glycans from glycoproteins that reduce the time needed for workflows to release glycans from glycoproteins and then to analyze the released glycans, the protein from which the glycans have been released, or both. Mixtures of M+detergents and Ammonium Cation Salts for Use In Preparing Glycoproteins for Deglycosylation Protocols It is believed a mixture of a compound of Formula 2 and a quaternary ammonium salt in a deglycosylation protocol allows rapid and effective enzymatic deglycosylation of a range of antibody and antibody-derived glycoproteins. The effective deglycosylation is expected to be due at least in part to the detergent effect of the mixture in allowing access of PNGase F to sites at which N-glycans are attached to the protein. It is further believed that this is due in part to the interaction of the quaternary ammonium cation with the sulfated aliphatic chain contributed by the compound of Formula 2 to the reaction mixture at the pH of the solution. Salts of other quaternary ammonium cations can contribute quaternary ammonium cations to the mixture and result in effective deglycosylation of a range of glycoproteins. Such salts are suitable so long as they are capable of dissociating under the pH and temperature conditions to be utilized in the deglycosylation procedure. Whether any particular salt can be used in the inventive methods can be readily determined by simply running a deglycosylation protocol in parallel on two aliquots of the same glycoprotein, preferably an antibody or an antibody-derived glycoprotein, with the protocol differing only in the detergent mix used, and comparing the amounts and types of glycans released from each aliquot. Test salts that are suitable for use in the inventive methods will result in the release of at least all of the same glycans as are released by the reference salt, in amounts equal to, greater than, or not more than 5% less than the amounts released by the reference mix. Due to their structural and functional similarity to quaternary ammonium cations, it is believed that salts of tertiary ammonium cations can also contribute ammonium cations to the mixture and result in effective deglycosylation of a range of glycoproteins. Secondary and primary ammonium cations are believed likely to work for effective deglycosylation, but are less preferred because they could interfere with later labeling of released glycans unless additional cleanup steps are introduced into the protocol. Mixtures of Detergents of Formula 1 and of Formula 2 It is believed that combining a detergent of Formula 2 with an ammonium cation sulfate or sulfonate detergent of Formula 1 will be useful in unfolding glycoproteins while keeping them in solution, rendering sites on the glycoprotein accessible to the deglycosylation enzyme that might not be made accessible by either detergent used on its own. Accordingly, in some embodiments, mixtures of detergents of Formula 1 and of Formula 2 are used in the enzymatic deglycosylation of glycoproteins, and particularly of antibodies and antibody-derivatives. Ammonium Fatty Acid Salts As noted above, initial studies using a fatty acid salt bearing an exemplar quaternary ammonium cation, proved to be good detergent for denaturing and deglycosylating glycoproteins. The exemplar ammonium fatty acid salt tested, tetramethyl ammonium laurate, caused the release of as much glycan as did an exemplar fatty acid salt, sodium laurate, when used in deglycosylation protocols on a number of glycoproteins, but resulted in releasing significantly more glycan from some hard-to-deglycosylate proteins. A comparison was made of the relative ability of tetramethyl ammonium laurate and an exemplar alkali metal fatty acid salt, sodium laurate, to serve as a detergent in denaturing glycans in an exemplar deglycosylation protocol. The detergents were tested on 18 different glycoproteins, including a number of FDA-approved therapeutic antibodies, and the resulting released glycans were analyzed. Previous work with similar data sets indicated that greater than a 5% difference in glycan release is significant. The quaternary ammonium salt tetramethyl ammonium laurate and the alkali metal salt sodium laurate were also tested as detergents on complex mixtures of glycoproteins. First, the detergents were tested on cell lysates of Chinese hamster ovary (“CHO”) cells. Cell lysates contain, among other glycoproteins, membrane proteins. As noted by Waas et al., Anal. Chem., 2014, 86(3):1551-1559, membrane proteins are hard to subject to enzymatic digestion due to their hydrophobic properties. Second, the detergents were tested on mammalian blood serum. In both cases, the exemplar alkali metal fatty acid salt and glycoprotein mixture precipitated after the denaturation, interfering with the release of glycans, while the quaternary ammonium fatty acid salt and glycoprotein mixture remained in solution and allowed enzymatic release of glycans present in the sample. Based on the results with two fusion proteins and two complex glycoprotein mixtures, ammonium cation fatty acid detergents were better in deglycosylation protocols for fusion proteins, particularly those containing Fc portions of antibodies, and for complex glycoprotein mixtures (including those containing membrane proteins) than are alkali metal or alkali earth metal fatty acid detergents, while working as well as alkali metal or alkali earth metal fatty acid detergents in deglycosylation protocols for glycoproteins such as antibodies The glycosylamines were released by enzymatic digestion and were then labeled with an amine-reactive dye. Embodiments in which the denaturant is present during the deglycosylation step is advantageous because it helps prevent the glycoprotein from refolding and perhaps rendering some sites once again inaccessible to the enzyme before deglycosylation can occur. Ammonium Cation Detergents As noted, our initial studies involved detergents having a quaternary ammonium cation and a fatty acid chain terminating in a carboxylate (an “ammonium carboxylate detergent”). We then discovered that an exemplar detergent with a quaternary ammonium cation and a sulfate anion was surprisingly better than the like detergent with a carboxylate anion. Accordingly, in some embodiments, the inventive compositions, methods, and kits employ a tertiary or quaternary ammonium cation and a sulfate or sulfonate anion. In other embodiments, the inventive compositions, methods, and kits employ a compound of Formula 2 and a tertiary or quaternary ammonium cation detergent, such as a quaternary ammonium sulfate detergent or a quaternary ammonium carboxylate detergent. The initial studies conducted with an exemplar quaternary ammonium carboxylate detergent showed it was useful in denaturing complex mixtures of glycoproteins and other proteins found in cell lysates and mammalian blood serum. Given the surprisingly better results in deglycosylating glycoproteins and mixtures of glycoproteins that we obtained using an exemplar quaternary ammonium sulfate detergent compared to the exemplar quaternary ammonium carboxylate detergent, it is believed that tertiary ammonium sulfate detergents and quaternary ammonium sulfate detergents, and tertiary ammonium sultanate and quaternary ammonium sulfonate detergents, will likewise prove surprisingly more useful in denaturing complex mixtures of glycoproteins and other proteins found in cell lysates and mammalian blood serum than are tertiary or quaternary ammonium carboxylate detergents. It is expected that tertiary ammonium sulfate and sulfonate detergents and quaternary ammonium sulfate and sulfonate detergents, and particularly tertiary or quaternary ammonium sulfate detergents, will remain soluble with a somewhat longer aliphatic chain than fatty acid-based detergents, and thus in some embodiments may be 8-22 carbons long. In some embodiments, the aliphatic chain may be 8-21 carbons long, in some embodiments may be 8-20 carbons long, in some embodiments may be 8-19 carbons long, in some embodiments may be 8-18 carbons long, in some embodiments may be 8-17 carbons long and in some embodiments may be 8-16 carbons long, while in still others, the chain may be 8-15 carbons long, in some embodiments may be 8-14 carbons long, and in some other embodiments may be 8-13 carbons long. In some preferred embodiments, the aliphatic chain is saturated. In some embodiments, the aliphatic chain is unsaturated. Lauric acid, which has 12 carbons, is one preferred for making ammonium cation sulfate and sulfonate detergents, with fatty acids with 11 carbons being next preferred. In some embodiments, the fatty acid used to form the salt has 12 carbons and is saturated. In some embodiments, the fatty acid used to form the salt has 11 carbons and is saturated. It is noted that “lauric acid” and the other trivial names for fatty acids refer to acids bearing the carboxylic acid end. Ammonium cation sulfate and sulfonate detergents derived from such fatty acids are often referred to by the cation, the length of the aliphatic chain, and the anion, such as “triethanol dodecanoic sulfate,” “trimethanol decanoic sulfonate”, or tributyl tetradecanoic sulfate.” This practice is not universally followed as, for example, “ammonium lauryl sulfate” is an anionic surfactant commonly found as an ingredient of shampoos and body washes, while the IUPAC name can state the chain length first, then the ammonium cation, then the anion, as in “octadecyl trimethyl ammonium sulfate” or “hexadecyl-trimethylammonium sulfonate.” The particular nomenclature chosen is not important. Branched fatty acid chains do not seem to be useful. In some embodiments, however, the fatty acid can have 8-13 carbons and be in a ring configuration. Use of Formula 1 and Formula 2 Detergents as Reagents to Denature other Proteins As reported in the Examples, an exemplar quaternary ammonium sulfate detergent proved surprisingly useful in denaturing glycoproteins and glycoprotein mixtures as part of a deglycosylation protocol. These results indicate that tertiary and quaternary ammonium sulfate and sulfonate detergents, will also be useful reagents for other proteolytic, analytic, and diagnostic protocols, such as in denaturing proteins prior to conducting a Western blot, and in denaturing a fusion protein or glycoproteins or proteins in a complex mixture, such as a cell lysate or blood serum or plasma. Tertiary and quaternary ammonium sulfate and sulfonate detergents are expected to be particularly useful for denaturing cell membrane proteins. Concentrations Typically, mixtures in which a compound of Formula 2 is mixed with a quaternary or tertiary ammonium cation salt, such as an ammonium cation fatty acid salt, will be 0.50% to 3.0%, 0.75-2.50%, 0.75.-2.25%, 1.0-2.0%, or 1.0-1.50% Formula 2 detergent, with each successive range being more preferred to the one before it. Good results can be obtained using a concentration of 125%, which concentration is the most preferred. The quaternary or tertiary ammonium cation salt is preferably present at 10 mM-150 mM, 10-125 mM, 20-100 mM, 25-100 mM, 30-100 mM. 30-90 mM. 30-80 mM. 30-75 mM, 40-70 mM, 40-65 mM, 40-60 mM, or 45-55 mM. Good results were obtained using 50 mM of exemplar quaternary ammonium detergents. The mixtures were in an aqueous base. The pH can be between 6 and 9.5 and may be a pH of 7-9, or of 8-9, Some of the underlying studies were performed in solutions without significant buffering capacity, at a pH of 8.5, which is a preferred pH for some embodiments. In embodiments in which a quaternary or tertiary ammonium sulfate or sulfonate (Formula 1) detergent is used, the detergent will typically be used in a concentration of 0.01 to 2.5%, more preferably 0.05 to about 2.0%, still more preferably 0.1 to about 1.5%, and in still more preferred embodiments, 0.01 to about 1%, where the term “about” means plus or minus 0.05% and the concentration is measured as volume/volume. Persons of skill are aware that glycoproteins differ in how hard they are to denature and that how hard any particular glycoprotein is to denature is usually determined empirically. If a particular glycoprotein proves to be hard to denature, the concentration of the detergent can be at the higher end of the stated amounts. As glycoproteins that are hard to denature are typically denatured at a higher temperature, a hard-to-denature glycoprotein will usually also be subjected to a higher temperature. Use of Multiple Detergents to Provide More Complete Solubilization Glycoproteins are usually denatured in a buffer which already contains one or more salts. For example, phosphate buffered saline is a common buffer which, as its name states, is a saline solution. Some combinations of ammonium cation sulfate or sulfonate detergent in combination with the salt already in the buffer, or of a mixture of a Formula 2 detergent and a quaternary or tertiary ammonium cation sulfate or sulfonate salt, in combination with salt already in the buffer, may result in a concentration of salt as to cause the glycoprotein to precipitate. Where the practitioner intends to perform an assay using a new combination of a particular ammonium cation detergent and a particular buffer with a particular glycoprotein, or of a mixture of a Formula 2 detergent and an quaternary or tertiary ammonium cation sulfate or sulfonate detergent, it is good practice to combine a small amount of these components to verify that the glycoprotein remains in solution. If the glycoprotein precipitates, which is easily observed visually, that indicates that the particular combination is too salty for use with that glycoprotein and that the practitioner should select a buffer with a lower salt concentration or a different ammonium cation detergent. As persons of skill are aware, these kinds of preliminary tests to find combinations of reagents suitable for use with a particular glycoprotein are usual in this art. Persons of skill will further appreciate that the ease of denaturing glycoproteins depends on a range of factors, including their secondary and tertiary structure, and some glycoproteins are resistant to denaturing even under conditions that would denature most other glycoproteins. In some embodiments, particularly with regard to denaturing a glycoprotein known to be hard to denature or one proving in practice to be hard to denature completely using an ammonium cation sulfate or sulfonate detergent, the practitioner may wish to also add one or more additional detergents or additional organic solvent denaturants, such as acetonitrile or tetrahydrofuran, or a chaotrope, such as urea or guanidinium chloride. As noted in the Background, current protocols often combine the anionic detergent SDS with a non-ionic detergent. We have found that a nonionic detergent is not needed with the exemplar quaternary ammonium detergents. Accordingly, in preferred embodiments, a non-ionic detergent is not present during deglycosylation of target glycoproteins or glycopeptides. A detergent of Formula 2 may be mixed with more than one ammonium cation salt at a time in an effort to improve solubilization of a glycoprotein or glycoprotein mixture for deglycosylation. Similarly, more than one Formula 2 detergent may be used with an ammonium cation salt or two such salts. In some embodiments, two or three different ammonium cation detergents may be combined in solution with the glycoprotein or mixture of glycoproteins to be denatured. In some of these embodiments, the ammonium cation of the two or the three salts is the same, but the anion is different. In other embodiments, the anion of two detergents is the same, but one of the detergents has a tertiary ammonium cation and the other a quaternary ammonium cation, or one has one tertiary cation and the other a different tertiary cation, or one has a first quaternary cation and the second has a second quaternary ammonium cation. In practice, there are diminishing returns as the number of detergents increases and while it might not be unusual to use two or three, it is unusual to see protocols calling for five or more. The use of multiple detergents and salts can complicate cleanup and removal for downstream analytic steps. In some embodiments, the ammonium cation detergents are all quaternary ammonium sulfate detergents. In some embodiments, the ammonium cation detergents are all quaternary sulfonate detergents. In some embodiments, the detergents are of the same or of different types, but can be conveniently be removed by use of a suitable cleanup column. Persons of skill are knowledgeable about the use of these reagents and their removal after they have served their purpose. Other Agents that May be Used During Denaturation of the Glycoprotein The solution containing the glycoprotein and the ammonium cation detergent can contain further agents commonly used in protocols for deglycosylating glycoproteins. In particular, the solution can contain reductants, such as tris(2-carboxyethyl)phosphine, dithiothreitol (DTT), beta-mercaptoethanol (BME), alkylants, such as iodoacetamide, or a combination of reductants and alkylants. The solution can contain an organic solvent denaturant, such as acetonitrile, tetrahydrofuran, trifluoroethanol, or hexafluoroisopropanol and may contain a chaotrope, such as urea or guanidinium chloride. It is expected that persons of skill in denaturing and deglycosylating glycoproteins are familiar with the use of each of these types of reagents and the compounds usually used for these purposes. Heating the Glycoprotein-Detergent Mixture to Speed Denaturation To denature the glycoprotein, a solution containing the ammonium cation detergent of choice is added to the glycoprotein and the resulting mixture is incubated. In some embodiments in which the practitioner want to complete the denaturation more quickly, the mixture can be heated. In some embodiments, the mixture is heated to a high temperature (typically, 90° C., although for glycoproteins known to be hard to denature, it may be higher). In some embodiments, the mixture is heated as high as 100° C. In most embodiments, the mixture will not be heated higher than 100° C., although in some embodiments, the mixture may be heated as high as 120° C. Typically, the solution will be heated for a time between 1 minute and about 10 minutes, more preferably 2-7 minutes, still more preferably about 2-5 minutes, even more preferably about 3 to about 5 minutes and most preferably about 3 minutes. It is not expected that heating the mixture for more than about 5 minutes will improve the denaturation of the glycoprotein. As used herein in connection with a statement of a time, the term “about” means plus or minus 30 seconds. Cooling The denatured glycoprotein or glycopeptide will preferably be cooled before being deglycosylated by a deglycosylation enzyme, such as the exemplar deglycosylation enzyme PNGase F, both to avoid denaturing the deglycosylation enzyme once it is added and to permit the deglycosylation to occur at a temperature in a range at which the enzyme is most active. The solution containing the glycoprotein is preferably cooled to about 22-60° C., and more preferably about 35-55° C. In some preferred embodiments, the glycoprotein or glycopeptide is cooled to about 45-50° C. and most preferably about 50° C. Persons of skill will appreciate that for some equipment, the heat transfer from the apparatus to the reactants is not complete and that a temperature setting of the heating apparatus will result in the reactants being at a temperature several degrees cooler than the temperature setting, and will adjust accordingly. As used herein in connection with a temperature, the term “about” means plus or minus 1 degree C. Deglycosylation In some embodiments, the glycoprotein is deglycosylated by a deglycosylation enzyme. In some embodiments, the deglycosylation enzyme is an amidase. In some embodiments, the deglycosylation enzyme is the amidase PNGase F. In some embodiments, the deglycosylation enzyme is an endoglycosidase such as Endoglycosidase F1, Endoglycosidase F2, Endoglycosidase F3, or Endoglycosidase H. In some embodiments, the practitioner wishes to distinguish between any N-glycans that may be present on the glycoprotein from any O-glycans that may be present. In some embodiments, the enzymes mentioned above are used in connection with an ammonium cation detergent denaturation to provide a fast method of removing the N-glycans from the glycoprotein so that any O-glycans or glycosaminoglycans (GAGS) that may be on the glycoprotein can be analyzed. For example, the first digestion may be made using an enzyme to remove N-glycans, followed by a second enzymatic digestion with endo-alpha-N-acetyl-galactosaminidase to remove O-glycans. It is expected that persons of skill are familiar with the various enzymes used for enzymatic release of carbohydrates from glycoconjugates in general and from glycoproteins in particular. Labeling PNGase F, a widely used deglycosylation enzyme, releases glycans from glycoproteins as glycosylamines. Various methods of labeling glycosylamines are known in the art, as exemplified by co-owned U.S. Pat. Nos. 8,124,792 and 8,445,292. If the glycosylamines are to be labeled with an amine-reactive dye, the dye labeling can be conducted without removal of the ammonium cation detergent. If labeling is to be performed using reductive amination of the reducing end of a glycan, rather than by releasing them from a glycoprotein as glycosylamines, the ammonium cation detergent is preferably first removed. Regardless of the method of labeling, the ammonium cation detergent may be removed before subjecting the labeled glycans or unlabeled glycoprotein to analytical methods, such as mass spectrometry, in which the presence of the ammonium cation detergent might be incompatible or would be a confounding factor. Removal of the Detergents Detergents used in deglycosylation protocols are preferably removed after labeling but before analysis of the labeled glycans, of the labeled glycosylamines, or of the glycoprotein or glycopeptide (which, following the deglycosylation step, is deglycosylated or aglycosylated), as detergents can be incompatible with some analytical instruments. Quaternary ammonium fatty acid detergents can be removed by precipitation with an acid, leaving behind in solution the now-deglycosylated or aglycosylated protein and the glycans released from the glycoprotein behind in the supernatant. The supernatant can then be removed by, for example, pipetting the supernatant away from the precipitate. Detergents of Formula 1 or Formula 2 can be removed by a solid phase extraction called hydrophilic interaction liquid chromatography, or “HILIC.” In studies conducted with an exemplar quaternary ammonium sulfate detergent, we found that HILIC was very effective at removing detergent from the deglycosylation solution. HILIC is a preferred embodiment for removal of ammonium sulfate or ammonium sulfonate detergents of Formula 1 used in some embodiments of the invention. In some embodiments, the detergent or detergents may be removed by using other solid- or liquid-phase techniques. It is expected that persons of skill are familiar with various types of liquid-liquid techniques and solid phase extraction devices which are used in the art to remove detergents from a solution. The solid phase extraction devices usually comprise resins on a solid support, and the resins are conveniently disposed in a cartridge or column (for convenience of reference, reference to a “column” in the following discussion refers to either a column or a cartridge, unless otherwise required by context). Common solid phase extraction devices include reverse phase columns, normal phase cartridges or columns, ion exchange columns, and size exclusion columns. Typically, the cleanup columns bind the glycans, allowing the detergent to flow through and be discarded, after which the glycans are eluted from the column. The use of a solid- or liquid-phase extraction technique is particularly preferred when the ammonium cation detergent is not susceptible to acid precipitation or to ensure removal of any detergent that does not precipitate out of solution. If one or more denaturants are used in addition to an ammonium cation detergent, they are typically also removed by using a cleanup column, such as a solid phase extraction column, and do not have to be LC or MS compatible. Analysis of Glycans, Glycoproteins, or Both Glycans can typically be eluted from a solid phase extraction device with water, after which they can be put in an analytical column or subjected to mass spectrometry (“MS”). Typical analytical means for analyzing labeled glycans or glycosylamines include high-pressure liquid chromatography, capillary electrophoresis, fluorescence analysis, mass spectrometry, or a combination of two or more of any of these. In some embodiments, the combination is of fluorescence analysis and mass spectrometry. Glycoproteins or glycopeptides deglycosylated during the course of releasing the glycans can themselves be analyzed, by any convenient means, for example, high-performance liquid chromatography, hydrophilic interaction chromatography, nuclear magnetic resonance, Western blotting, gel electrophoresis, fluorescence analysis, capillary electrophoresis, microfluidic separation, mass spectrometry, or a combination of two or more of any of these. EXAMPLES Example 1 This Example sets forth the method used to prepare an exemplar quaternary ammonium sulfate detergent, tetramethyl ammonium dodecyl sulfate. Twenty mL of AMBERLYST® 36 (Sigma-Aldrich, St. Louis, MO), a cation-exchange resin, was loaded into a glass chromatography column with a coarse frit and rinsed with ten column volumes of water. Tetramethylammonium hydroxide was passed through the column as an aqueous solution, and the eluent was monitored with pH paper to determine the transition from neutral to basic pH, Excess tetramethylammonium hydroxide was rinsed from the column with water, and an aqueous solution of sodium dodecyl sulfate was loaded onto the column. The molar amount of SDS loaded was approximately tenfold less than the molar amount of tetramethylammonium ions estimated to be present on the resin. The column was flushed with water. Finally, the water was removed by rotary evaporation, yielding a white solid composed of tetramethylammonium dodecyl sulfate. Example 2 This Example sets forth the protocol used for comparisons of the amount of glycan released from glycoproteins by enzymatic digestion following denaturing with either (a) sodium dodecyl sulfate or (b) an exemplar quaternary ammonium sulfate detergent, tetramethyl ammonium dodecyl sulfate. Denaturation Step A series of glycoproteins were selected for testing ranging from having one glycosylation to multiple sites. Aliquots of 40 to 100 μg solution of each glycoprotein to be tested were prepared in 20 μl of pH 7.5 in a compatible reaction buffer of choice, forming a solution containing glycoprotein and water. Two μl of a 50 mM solution of either sodium dodecyl sulfate (detergent 1) or tetramethyl ammonium dodecyl sulfate (detergent 2) were added to the glycoprotein/buffer solution for a final concentration of 4.5 mM of the detergent. The mixtures were then incubated for 3 minutes at 90° C. Enzymatic Digestion Step Two μl of 1 mg/ml of PNGase F in tetramethyl ammonium HEPES buffer, pH8, was added and the resulting mixture was incubated for at 37° C. for one hour. Labeling Step Following this incubation, glycans released by the PNGase F were labeled with InstantPC® dye (ProZyme, Inc., Hayward, CA) by adding 5 μl of the dye solution to each sample and incubating the samples for one minute at 50° C. Cleanup Step The labeled glycans were resuspended in 600 μl of 95:5 acetonitrile: formic acid and the solutions were loaded onto cleanup cartridges. The cartridges were then washed three times with 600 μl of a 95:5 acetonitrile: formic acid solution. The labeled glycans were then eluted with 100 μl of a solution of 200 mM ammonium formate, pH 7, containing 10% acetonitrile. Analysis Step One μl of each eluted sample was injected into high performance liquid chromatography (HPLC) equipment for analysis. The amount of glycans released from each glycoprotein when tetramethyl ammonium dodecyl sulfate was used as the detergent was considered to be “100%” release, which allowed a ready comparison to the amount of glycan released using the same assay on the same glycoprotein, but using sodium dodecyl sulfate rather than tetramethyl ammonium dodecyl sulfate as the detergent to denature the glycoprotein. Example 3 This Example sets forth the results of the study reported in Example 2. FIG.1is a table setting forth the results of studies comparing the use of tetramethyl ammonium dodecyl sulfate and sodium dodecyl sulfate (SDS) as detergents in a deglycosylation protocol on a series of nine glycoproteins and mixtures of glycoproteins. To permit ready comparison of how well each detergent served in the deglycosylation protocol, the amount of glycans released from each glycoprotein or mixture of glycoproteins in the assay in which tetramethyl ammonium dodecyl sulfate was used as the detergent was normalized to 100%, and the amount released then compared to the amount of glycan released when SDS was used as the detergent in the same deglycosylation protocol on the same glycoprotein. The studies included three mixtures of human antibodies of different antibody classes: hIgG, hIgA, and hIgM; an antibody, designated RM 8671, provided by the National Institute of Standards and Technology (“NIST”) as a reference material for testing, and five glycoproteins currently approved by the Food and Drug Administration for therapeutic use.FIG.2presents the results of the comparative studies reported inFIG.1in the form of a graph, with error bars representing the results of three replicates. To allow ready comparison, the amount of glycans released from each glycoprotein or mixture by digestion following denaturing with tetramethyl ammonium lauryl sulfate was considered to be a 100% release, and that amount was then compared to the amount released from the same glycoprotein or glycoprotein mixture following denaturing with SDS. For four out of nine of the glycoproteins or glycoprotein mixtures, denaturing with SDS resulted in the release of significantly fewer glycans than did the use of the exemplar quarternary ammonium sulfate detergent, and for the mixture of human IgGs, denaturing with SDS resulted in the release of almost 25% less glycan than did denaturing with the quarternary ammonium sulfate detergent. For a mixture of hIgM antibodies, denaturing with SDS resulted in the release of 18% less glycan than did denaturing with the quarternary ammonium sulfate detergent. For the therapeutic biologic agent ORENCIA®, denaturing with SDS resulted in the release of almost 15% less glycan than did denaturing with the quarternary ammonium sulfate detergent. For three of the nine glycoproteins or mixtures, using SDS to denature the glycoprotein resulted in roughly the same release of glycan compared with the amount released following denaturation of the same glycoprotein with the quarternary ammonium sulfate detergent. In the case of one glycoprotein, the chimeric mouse/human monoclonal antibody cetuximab, denaturing with SDS resulted in the release of more glycan than did the use of quarternary ammonium sulfate detergent, and in that case the difference was less than 10%. The results indicate that quarternary ammonium sulfate detergents provide generally better or equal results, and in many cases, surprisingly better results in deglycosylation protocols compared to SDS. It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.
51,713
11858962
EXAMPLES The following examples are only intended to illustrate to the present invention. They shall not limit the scope of the claims in any way. Example 1 Sf rhabdovirus glycoprotein (SFRVG) Baculoviral Expression Constructs An SFRVG ectodomain was initially generated, by removing the transmembrane helix and cytoplasmic domains of SFRVG. A further modification was to fuse wild type and mutant SFRV ecto to an immunoglobulin G fragment crystallizable (IgG Fc) protein. SfRV glycoprotein ectodomain (SFRVGecto) has been cloned, inserted into a baculoviral transfer plasmid, and recombinant baculovirus generated to express SFRVGecto. Successive iterations of recombinant baculoviruses were made resulting in a recombinant baculovirus that could express full length SFRVGecto which could subsequently be recovered in high molar quantity. Discovery of Furin Cleavage Sites in SFRVG Secondary structure prediction programs Jpred (http://www.compbio.dundee.ac.uk/jpred/) and PSIPRED (http://bioinf.cs.ucl.ac.uk/psipred/) were used to locate coiled regions in SFRVG (SEQ ID NO:16) while Globplot (http://globplot.embl.de/) was used to find predicted regions of disorder as such regions are anticipated to be more sensitive to proteolytic cleavage. These programs predicted a large coiled region roughly comprising amino acids 285-335 (numbering according to the immature protein) that overlapped with a disordered region roughly comprising amino acids 285-305. Review of this region identified a highly charged region with sequence RERR from amino acid 303 to 306, which is a furin protease site with presumed cleavage site following the final arginine. The proprotein convertase recognition site prediction program ProP (http://www.cbs.dtu.dk/services/ProP/; Duckert, 2004) was then used to confirm the furin site. Not only was this position predicted with high confidence to be a furin site, a second site within the largely coiled region from amino acid 330 to 333 with sequence RHKR was also predicted to be cleavable by furin. Removal of SFRVG Furin Sites Site-directed mutagenesis was performed to remove the furin sites from SFRVG. Three different mutations to SFRVGecto were then made: R306Q, R333Q, and R306Q/R333Q, which were then assessed for their ability to express SFRVGecto. The SFRVGecto sequence used for this purpose contained the first 550 amino acids of SFRVG, including the native signaling peptide, fused (C-term) to a TEV protease site followed by a 6×His tag. Point mutants were generated as follows: the first 550 amino acids that comprise the SFRVG ectodomain were PCR amplified, gel purified, and TOPO cloned. Following verification of the insert by colony screen by PCR and DNA sequencing, R306Q and R333Q point mutations were individually made using a QuikChange Lightning Site Directed Mutagenesis kit (Agilent, cat #000628596). Inserts were verified by DNA sequencing, and using TOPO-SFRVGecto-R306Q as the template the R306Q/R333Q double mutant was made and subsequently verified by DNA sequencing. All three TOPO-SFRVGecto mutants were EcoRI/PstI digested, and gel purified while pVL1393 was EcoRI/PstI digested, dephosphorylated, and gel purified. Ligations were done using T4 DNA ligase. Inserts into pVL1393 were verified by colony screen by PCR and DNA sequencing. Baculoviruses were generated by co-transfecting with FlashBAC ULTRA (FBU) into Sf9 cells. IFAs were performed using anti-baculovirus envelope gp64 purified clone AcV1 (eBiosciences, cat #14-6991-83) or anti-His (C-term; Invitrogen, cat #46-0693) primary antibodies at 1:100 and FITC-conjugated goat anti-mouse (JIR, cat #115-095-003) secondary antibody, also at 1:100 dilution. Sf9 cells transfected with pVL1393-SFRVGecto-R306Q, -R333Q, or -R306Q/R333Q plasmids were positive for both 6×His and baculovirus gp64 protein. FBU/pVL1393-SFRVGecto-R306Q, -R333Q, and -R306Q/R333Q baculoviruses were expanded on T25 flasks of Sf9 cells for six days and P2 baculoviruses titered. P3 expansion and protein expression trials were done by inoculating 100 mL of Sf+ cells in 500 mL spinner flasks at 0.1 MOI. Samples of spent media and cells were harvested 3-5 DPI (Days Post Infection) with the remaining culture harvested 5 DPI. Cell pellets were lysed in buffer containing 1% Triton X-100, and samples centrifuged 20 minutes at 20,000 g. Resulting samples were run out on SDS-PAGE, transferred to nitrocellulose, and western blot probed with 1:500 dilution of anti-His (C-term; Invitrogen, cat #46-0693) primary antibody and 1:1000 dilution of HRP-conjugated goat anti-mouse (JIR, cat #115-035-146) secondary antibody Fusion of SFRVG Ectodomain to Swine Immunoglobulin G 2a Fragment Crystallizable (IgG Fc) To aid in expression of SFRVG and provide a means of protein purification SFRVGecto-R306Q/R333Q was fused to a swine IgG 2a Fc domain (said IgG Fc domain having the sequence of SEQ ID NO:6). Simultaneously SFRVGecto with the furin sites intact (SFRVGecto-WT) was also fused to an IgG Fc to determine if in the context of a fusion protein removal of the two furin sites was required. Assembly of the two protein coding sequences and insertion into pVL1393 baculovirus transfer plasmid is briefly as follows: primers for amplifying SFRVGecto and IgG Fc were received and SFRVGecto-WT, SFRVG-R306Q/R333Q, and IgG Fc were amplified by PCR, gel purified, and OEPCR done to generate the fusion protein inserts. OEPCR products were gel purified and TOPO cloned, with inserts verified by colony screen by PCR and DNA sequencing. TOPO clones containing the SFRVGecto-WT-IgG2a and SFRVGecto-R306Q/R333Q-IgG2a inserts were EcoRI/PstI digested and gel purified while pVL1393 was EcoRI/PstI digested, dephosphorylated, and gel purified. Ligations were done using T4 DNA ligase with inserts verified by colony screen by PCR and DNA sequencing. Baculoviruses were generated by co-transfecting pVL1393-SFRVGecto-R306Q/R333Q-IgG2a or pVL1393-SFRVGecto-WT-IgG2a with FlashBAC ULTRA (FBU) into Sf9 cells. IFAs were performed using anti-baculovirus envelope gp64 purified clone AcV1 (eBiosciences, cat #14-6991-83) primary antibody at 1:100 and FITC-conjugated goat anti-mouse (JIR, cat #115-095-003) secondary antibody, also at 1:00 dilution. Sf9 cells transfected with either pVL1393 plasmid were positive for baculovirus gp64 protein. Both FBU/pVL1393-SFRVGecto-R306Q/R333Q-IgG2a and FBU/pVL1393-SFRVGecto-WT-IgG2a baculoviruses were expanded on T25 flasks of Sf9 cells for six days and P2 baculoviruses titered. P3 expansion and protein expression trials were done by inoculating 100 mL of Sf+ cells in 500 mL spinner flasks with either baculovirus at 0.1 MOI. Samples of spent media and cells were harvested 3 and 4 DPI with the remaining culture harvested 4 DPI. Cell pellets were lysed in buffer containing 1% Triton X-100, and samples centrifuged 20 minutes at 20,000 g. Resulting samples were run out on SDS-PAGE, transferred to nitrocellulose, and western blot probed with 1:1000 dilution of HRP-conjugated goat anti-swine (JIR, cat #115-035-003) antibody. From the experiments including the above described SDS Page and Western Blot analyses it was seen that (i) the fusion of the SFRVG ectodomain to Swine Immunoglobulin G 2a Fragment Crystallizable (IgG Fc), or (ii) the SFRVG ectodomain having one of the substitutions (R306Q or R333Q), respectively, resulted in a significantly higher molar yield in the expression system as compared to the expression of the respective unmodified SFRVG ectodomain. Further, it was found that the combination of both substitutions R306Q and R333Q within the SFRVG ectodomain resulted in a significantly higher molar yield as compared to the expression of SFRVG ectodomain having only one of these substitutions (R306Q or R333Q). Finally, it was then surprisingly seen that the combination of the above modifications resulted in a synergistic effect, as it was found that the expression of the SFRV ectodomain with both substitutions R306Q/R333Q fused to IgG2a (pVL1393-SFRVGecto-R306Q/R333Q-IgG2a), revealed a much higher yield (by at least a factor 8) as compared to the expression ofthe respective SFRVG wild type ectodomain fused to IgG2a (pVL1393-SFRVGecto-WT-IgG2a) orthe SFRVG ectodomains having both substitutions R306Q and R333Q (pVL1393-SFRVGecto-R306Q/R333Q). A respective synergistic effect, resulting in a much higher yield, was also observed for a corresponding combination including a guinea pig IgG Fc domain, namely for a fusion protein comprising the sequence of SEQ ID NO:1. Furthermore, said sequence of SEQ ID NO:1 can be linked via a linker to the guinea pig IgG Fc domain, e.g. to the sequence of SEQ ID NO:5. Therefore, in particular, a respective synergistic effect, resulting in a much higher yield, was observed for a fusion protein having the sequence of SEQ ID NO:12, which comprises the sequence of SEQ ID NO:1 and the sequence of SEQ ID NO:5 linked to said sequence of SEQ ID NO:1 via a peptide linker. Example 2 An ELISA is employed to evaluate the presence of anti-rhabdovirus antibodies in different liquid samples. For this purpose, a fusion protein of the above-mentioned formula x-y-z is immobilized as the antigen to an ELISA plate (with x being the ectodomain of a glycoprotein of the rhabdovirus for which the antibodies to be detected are specific, y being a peptide linker and z being an IgG Fc domain), wherein for example the fusion protein comprising the sequence of SEQ ID NO:12 is immobilized. The ELISA method used in this context is described in the following protocol:1. Coat plates or strips with 5-500 ng/well of antigen (include plates with different binding capacities, material (polystyrene etc), formats (strips/96 well plates) etc). Incubate overnight at 2-8° C. for binding.2. Wash plates and block wells with blocking buffer containing 2-10% non fat milk in PBS and 0.5-10% additional protein including BSA/non-relevant serum.3. After the blocking step, wash plates in a plate washer and tap plate on a wad of paper towels to get rid of remaining wash fluids.4. Dilute test serum 1:100 and add 100 μL diluted test serum per well. Add 100 μL negative control serum (and, where necessary, positive control serum), diluted 1:100, to control wells.5. Tap side of plates to shake and mix. Seal the plate/strip and incubate at 37° C. (98.6° F.) for 1 hour.6. Wash plates in a plate washer and tap plate on a wad of paper towels to get rid of remaining wash fluids.7. Add 100 μL of pre-diluted (dilution 1:1000-1:100000) HRP Conjugate (e.g. anti-pig IgG (whole molecule), HRP conjugated) to each well. Seal the plate and incubate at 37° C. for 1 hour.8. Wash plates in a plate washer and tap plate on a wad of paper towels to get rid of remaining wash fluids.9. Add 100 μL of Substrate Solution to each well. Incubate for 10 minutes at room temperature. Start timer when the first well is filled.10. Stop the reaction by adding 50 μL of Stop Solution to each well and mix gently by tapping sides.11. Measure the OD at 450 nm within 15 minutes after the addition of Stop Solution to prevent fluctuation in OD values. The results of the ELISA show a clear difference between the samples containing the anti-rhabdovirus antibodies to be detected (said samples showing e.g. a S/P ratio of above 0.5) and the negative controls (i.e. corresponding samples not containing such antibodies, which show e.g. a S/P ratio of approx. 0). In conclusion, the use of the polypeptide of the present invention for detecting anti-rhabdovirus antibodies allows to readily discern samples including anti-rhabdovirus antibodies from samples not including such antibodies. In the Sequence Listing: SEQ ID NO:1 corresponds to the sequence of an ectodomain (without N-terminal signaling peptide) of the glycoprotein set forth in SEQ ID NO:16 having a substitution (i.e., a glutamine residue instead of an arginine residue) at each of the amino acid positions 306 and 333, and wherein said amino acid positions of SEQ ID NO:16 correspond to the sequence positions 285 and 312 of SEQ ID NO:1, SEQ ID NO:2 corresponds to the sequence of SEQ ID NO:1 N-terminally extended by a serine residue (corresponding to the serine residue at amino acid position 21 of SEQ ID NO:16), SEQ ID NO:3 corresponds to the sequence of SEQ ID NO:1 N-terminally extended by the N-terminal 21 amino acid residues (i.e., including the N-terminal signaling peptide) of SEQ ID NO:16, SEQ ID NO:4 corresponds to the sequence of the glycoprotein set forth in SEQ ID NO:16 having a substitution (i.e., a glutamine residue instead of an arginine residue) at each of the amino acid positions 306 and 333, SEQ ID NO:5 corresponds to the sequence of a guinea pig IgG Fc domain, SEQ ID NO:6 corresponds to the sequence of a swine IgG Fc domain, SEQ ID NO:7 corresponds to the sequence of a GCN4 leucine zipper domain, SEQ ID NO:8 corresponds to anEscherichiavirus T4 fibritin sequence, SEQ ID NO:9 corresponds to the sequence of a linker moiety, SEQ ID NO:10 corresponds to the sequence of a linker moiety, SEQ ID NO:11 corresponds to the sequence of a linker moiety, SEQ ID NO:12 corresponds to the sequence of a polypeptide of the present invention, SEQ ID NO:13 corresponds to the sequence of a polypeptide of the present invention, SEQ ID NO:14 corresponds to the sequence of a polypeptide of the present invention, SEQ ID NO:15 corresponds to the sequence of an ectodomain of the wild type glycoprotein set forth in SEQ ID NO:16, SEQ ID NO:16 corresponds to the sequence of a wild type Sf-rhabdovirus glycoprotein, SEQ ID NO:17 corresponds to the sequence of a polynucleotide encoding a polypeptide of the present invention, SEQ ID NO:18 corresponds to the sequence of a polynucleotide encoding a polypeptide of the present invention, SEQ ID NO:19 corresponds to the sequence of a polynucleotide encoding a polypeptide of the present invention, SEQ ID NO:20 corresponds to the sequence of a furin cleavage site, SEQ ID NO:21 corresponds to the sequence of a furin cleavage site, SEQ ID NO:22 corresponds to the sequence of a furin cleavage site, SEQ ID NO:23 corresponds to the sequence of a furin cleavage site, SEQ ID NO:24 corresponds to the sequence of a furin cleavage site, SEQ ID NO:25 corresponds to the sequence of a furin cleavage site. The following clauses are also disclosed herein:1. A polypeptide comprisingan ectodomain of a rhabdovirus glycoprotein, anda heterologous multimerization domain linked to said ectodomain.2. The polypeptide of clause 1,wherein said heterologous multimerization domain is linked to said ectodomain via a linker moiety,or wherein said heterologous multimerization domain is linked to said ectodomain via a peptide bond between the N-terminal amino acid residue of said heterologous multimerization domain and the C-terminal amino acid residue of said ectodomain.3. A polypeptide, in particular the polypeptide of clause 1 or 2, wherein said polypeptide is a fusion protein of the formula x-y-z, whereinx consists of or comprises an ectodomain of a rhabdovirus glycoprotein;y is a linker moiety; andz is a heterologous multimerization domain.4. The polypeptide of any one of clauses 1 to 3, wherein said ectodomain is free of a furin cleavage site.5. The polypeptide of clause 4, wherein said furin cleavage site is an amino acid sequence selected from the group consisting of the following (a), (b), and (c):(a) amino acid sequence selected from the group consisting of RXKR (SEQ ID NO:20) and RXRR (SEQ ID NO:21);(b) amino acid sequence selected from the group consisting of RX1KRX2(SEQ ID NO:22) and RX1RRX2(SEQ ID NO:23), whereinX1can be any amino acid residue, andX2can be any amino acid residue other thana lysine residue oran amino acid residue selected from the group consisting of valine residue, leucine residue, isoleucine residue and tryptophane residue;(c) amino acid sequence selected from the group consisting of RX1KRX2X3(SEQ ID NO:24) and RX1RRX2X3(SEQ ID NO:25), whereinX1can be any amino acid residue,X2can be any amino acid residue other thana lysine residue oran amino acid residue selected from the group consisting of valine residue, leucine residue, isoleucine residue and tryptophane residue,and X3can be any amino residue other than a lysine residue.6. The polypeptide of any one of clauses 1 to 5, wherein said rhabdovirus glycoprotein is aS. frugiperdarhabdovirus (SF-rhabdovirus) glycoprotein.7. The polypeptide of any one of clauses 1 to 6, wherein said ectodomain is an ectodomain of a SF-rhabdovirus glycoprotein having(i) one or more mutations selected from the group consisting of substitution at amino acid position 306, substitution at amino acid position 303, substitution at amino acid position 305, substitution at amino acid position 307, and substitution at amino acid position 308,and(ii) one or more mutations selected from the group consisting of substitution at amino acid position 333, substitution at amino acid position 330, substitution at amino acid position 332, substitution at amino acid position 334, and substitution at amino acid position 335,wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type SF-rhabdovirus glycoprotein.8. The polypeptide of any one of clauses 1 to 7, wherein said ectodomain is an ectodomain of a SF-rhabdovirus glycoprotein comprising or consisting of an amino acid sequence having at least 70%, preferably at least 80%, more preferably at least 90% or in particular at least 95% sequence identity with the sequence of SEQ ID NO:4.9. The polypeptide of any one of clauses 1 to 8, wherein said ectodomain is an ectodomain of a SF-rhabdovirus glycoprotein havingat amino acid position 306 an amino acid residue other than an arginine residue, and/orat amino acid position 303 an amino acid residue other than an arginine residue, and/orat amino acid position 305 an amino acid residue other than a basic amino acid residue, and/orat amino acid position 307 an amino acid residue selected from the group consisting of lysine residue, leucine residue, isoleucine residue, valine residue and tryptophane residue, and/orat amino acid position 308 a lysine residue,and havingat amino acid position 333 an amino acid residue other than an arginine residue, and/orat amino acid position 330 an amino acid residue other than an arginine residue, and/orat amino acid position 332 an amino acid residue other than a basic amino acid residue, and/orat amino acid position 334 an amino acid residue selected from the group consisting of lysine residue, leucine residue, isoleucine residue, valine residue and tryptophane residue, and/orat amino acid position 335 a lysine residue,and wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type SF-rhabdovirus glycoprotein.10. The polypeptide of any one of clauses 1 to 9, wherein said ectodomain is an ectodomain of a SF-rhabdovirus glycoprotein havingat amino acid position 306 an amino acid residue other than an arginine residue,andat amino acid position 333 an amino acid residue other than an arginine residue,and wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type SF-rhabdovirus glycoprotein.11. The polypeptide of any one of clauses 1 to 10, wherein said ectodomain is an ectodomain of a SF-rhabdovirus glycoprotein comprising or consisting of an amino acid sequence having at least 70%, preferably at least 80%, more preferably at least 90% or in particular at least 95% sequence identity with the sequence of SEQ ID NO:4 and havingat amino acid position 306 an amino acid residue other than an arginine residue, andat amino acid position 333 an amino acid residue other than an arginine residue,and wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type SF-rhabdovirus glycoprotein.12. The polypeptide of any one of clauses 9 to 11, wherein said amino acid residue other than an arginine residue is a naturally occurring, preferably a genetically encoded, amino acid residue.13. The polypeptide of any one of clauses 6 to 12, wherein the N-terminal amino acid residue of said ectodomain corresponds to any one of the amino acid positions 1-22 of the amino acid sequence of wild type SF-rhabdovirus glycoprotein.14. The polypeptide of any one of clauses 6 to 13, wherein the N-terminal amino acid residue of said ectodomain corresponds to any one of the amino acid positions 22, 21 or 1 of the amino acid sequence of wild type SF-rhabdovirus glycoprotein.15. The polypeptide of any one of clauses 1 to 14, wherein said ectodomain is an ectodomain of a SF-rhabdovirus glycoprotein havingat amino acid position 306 an amino acid residue selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residueand/or at amino acid position 303 an amino acid residue selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residue;andat amino acid position 333 an amino acid residue selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residueand/or at amino acid position 330 an amino acid residue selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residue;and wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type SF-rhabdovirus glycoprotein.16. The polypeptide of clause 15, wherein said amino acid residue with a polar but uncharged side chain is selected from the group consisting of serine residue, threonine residue, tyrosine residue, asparagine residue, and glutamine residue, and/or wherein said amino acid residue with a hydrophobic side chain is selected from the group consisting of alanine residue, valine residue, leucine residue, methionine residue, isoleucine residue, phenylalanine residue, and tryptophan residue.17. The polypeptide of any one of clauses 1 to 16, wherein said ectodomain is an ectodomain of a SF-rhabdovirus glycoprotein havingat amino acid position 306 an amino acid residue selected from the group consisting of glutamine residue and asparagine residue and/or at amino acid position 303 an amino acid residue selected from the group consisting of glutamine residue and asparagine residue;andat amino acid position 333 an amino acid residue selected from the group consisting of glutamine residue and asparagine residue and/or at amino acid position 330 an amino acid residue selected from the group consisting of glutamine residue and asparagine residue;and wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type SF-rhabdovirus glycoprotein.18. The polypeptide of any one of clauses 7 to 17, wherein said amino acid sequence of wild type SF-rhabdovirus glycoprotein consists of or is the amino acid sequence of SEQ ID NO:16.19. The polypeptide of any one of clauses 1 to 18, wherein said ectodomain comprises or consists of an amino acid sequence having at least 70%, preferably at least 80%, more preferably at least 90% or still more preferably at least 95% sequence identity with a sequence selected from the group consisting of SEQ ID NO:1, SEQ ID NO:2 and SEQ ID NO:3.20. The polypeptide of any one of clauses 1 to 19, wherein said ectodomain comprises or consists of an amino acid sequence having at least 70%, preferably at least 80%, more preferably at least 90% or still more preferably at least 95% sequence identity with the amino acid sequence of SEQ ID NO:1,and wherein said ectodomain hasat amino acid position 285 an amino acid residue other than an arginine residue, and/orat amino acid position 282 an amino acid residue other than an arginine residue, and/orat amino acid position 286 an amino acid residue selected from the group consisting of lysine residue, leucine residue, isoleucine residue, valine residue and tryptophane residue, and/orat amino acid position 287 a lysine residue,and hasat amino acid position 312 an amino acid residue other than an arginine residue, and/orat amino acid position 309 an amino acid residue other than an arginine residue, and/orat amino acid position 313 an amino acid residue selected from the group consisting of lysine residue, leucine residue, isoleucine residue, valine residue and tryptophane residue, and/orat amino acid position 314 a lysine residue,wherein the numbering of the amino acid positions refers to the amino acid sequence of SEQ ID NO:1.21. The polypeptide of clause 20, wherein said ectodomain hasat amino acid position 285 an amino acid residue other than an arginine residue,andat amino acid position 312 an amino acid residue other than an arginine residue,and wherein the numbering of the amino acid positions refers to the amino acid sequence of SEQ ID NO:1.22. The polypeptide of any one of clauses 9 to 21, wherein said amino acid residue other than an arginine residue is a naturally occurring, preferably a genetically encoded, amino acid residue,and/or wherein said amino acid residue other than a basic amino acid residue is a naturally occurring, preferably a genetically encoded, amino acid residue.23. The polypeptide of any one of clauses 20 to 22, wherein said ectodomain hasat amino acid position 285 an amino acid residue selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residueand/or at amino acid position 282 an amino acid residue selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residue;andat amino acid position 312 an amino acid residue selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residueand/or at amino acid position 309 an amino acid residue selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residue;and wherein the numbering of the amino acid positions refers to the amino acid sequence of SEQ ID NO:1.24. The polypeptide of clause 23, wherein said amino acid residue with a polar but uncharged side chain is selected from the group consisting of serine residue, threonine residue, tyrosine residue, asparagine residue, and glutamine residue, and/or wherein said amino acid residue with a hydrophobic side chain is selected from the group consisting of alanine residue, valine residue, leucine residue, methionine residue, isoleucine residue, phenylalanine residue, and tryptophan residue.25. The polypeptide of any one of clauses 20 to 24, wherein said ectodomain hasat amino acid position 285 an amino acid residue selected from the group consisting of glutamine residue and asparagine residue and/or at amino acid position 282 an amino acid residue selected from the group consisting of glutamine residue and asparagine residue;andat amino acid position 312 an amino acid residue selected from the group consisting of glutamine residue and asparagine residue and/or at amino acid position 309 an amino acid residue selected from the group consisting of glutamine residue and asparagine residue;and wherein the numbering of the amino acid positions refers to the amino acid sequence of SEQ ID NO:1.26. The polypeptide of any one of clauses 1 to 25, wherein said ectodomain comprises or consists of an amino acid sequence being 529-550 amino acid residues in length.27. The polypeptide of any one of clauses 1 to 26, wherein said ectodomain comprises or consists of an amino acid sequence being 529, 530 or 550 amino acid residues in length.28. The polypeptide of any one of clauses 1 to 27, wherein said ectodomain has the sequence of any one of SEQ ID NO:1 to SEQ ID NO:3.29. The polypeptide of any one of clauses 1 to 28, wherein the heterologous multimerization domain is selected from the group consisting of immunoglobulin sequence, coiled coil sequence, streptavidin sequence, fibritin sequence, and avidin sequence.30. The polypeptide of any one of clauses 1 to 29, wherein the heterologous multimerization domain is selected from the group consisting of immunoglobulin constant region domain, leucine zipper domain andEscherichiavirus T4 fibritin sequence.31. The polypeptide of any one of clauses 1 to 30, wherein the heterologous multimerization domain is a dimerization domain, preferably selected from the group consisting of IgG Fc domain and leucine zipper domain.32. The polypeptide of any one of clauses 1 to 31, wherein the heterologous multimerization domain comprises or consists of a IgG Fc domain.33. The polypeptide of any one of clauses 1 to 32, wherein the heterologous multimerization domain comprises or consists of a guinea pig IgG Fc domain.34. The polypeptide of any one of clauses 1 to 33, wherein the heterologous multimerization domain comprises or consists of an amino acid sequence having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a sequence selected from the group consisting of SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7 and SEQ ID NO:8.35. The polypeptide of any one of clauses 2 to 34, wherein said linker moiety is an amino acid sequence being 1 to 50 amino acid residues in length.36. The polypeptide of any one of clauses 2 to 35, wherein said linker moiety is an amino acid sequence being 3 to 20 amino acid residues in length.37. The polypeptide of any one of clauses 2 to 36, wherein said linker moiety comprises or consists of an amino acid sequence having at least 66%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a sequence selected from the group consisting of SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11.38. The polypeptide of any one of clauses 1 to 37, wherein said polypeptide is a protein comprising or consisting of an amino acid sequence having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% sequence identity with a sequence selected from the group consisting of SEQ ID NO:12, SEQ ID NO:13 and SEQ ID NO:14.39. The polypeptide of any one of clauses 1 to 38, wherein said polypeptide is a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NO:12, SEQ ID NO:13 and SEQ ID NO:14.40. The polypeptide of any one of clauses 1 to 39, wherein in a baculovirus expression system the yield of said polypeptide is higher, preferably by at least a factor 2, more preferably by at least a factor 3, still more preferably by at least a factor 5, yet more preferably by at least a factor 8, compared to the yield of the polypeptide of SEQ ID NO:15.41. The polypeptide of any one of clauses 1 to 40, wherein said polypeptide is a recombinant protein, preferably a recombinant baculovirus expressed protein.42. A polynucleotide which encodes the polypeptide of any one of clauses 1 to 41.43. The polynucleotide of clause 42, wherein said polynucleotide comprises a nucleotide sequence having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a sequence selected from the group consisting of SEQ ID NO:17, SEQ ID NO:18 and SEQ ID NO:19.44. A plasmid, preferably an expression vector, comprising a polynucleotide which encodes the polypeptide of any one of clauses 1 to 41.45. A cell comprising a plasmid, preferably an expression vector, which comprises a polynucleotide encoding the polypeptide of any one of clauses 1 to 41.46. A baculovirus containing a polynucleotide which encodes the polypeptide of any one of clauses 1 to 41.47. A cell, preferably an insect cell, infected with a baculovirus which contains a polynucleotide encoding the polypeptide of any one of clauses 1 to 41.48. The plasmid of clause 44, the cell of clause 45, the baculovirus of clause 46 or the cell of clause 47, wherein said polynucleotide comprises a nucleotide sequence having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a sequence selected from the group consisting of SEQ ID NO:17, SEQ ID NO:18 and SEQ ID NO:19.49. A kit containing the polypeptide of any one of clauses 1 to 41 immobilized to a solid support.50. A method of producing the polypeptide of any one of clauses 1 to 41, wherein the method comprisestransfecting a cell with a plasmid, preferably an expression vector, which comprises a polynucleotide comprising a sequence which encodes said polypeptide,orinfecting a cell, preferably an insect cell, with a baculovirus containing a polynucleotide comprising a sequence which encodes said polypeptide.51. The method of clause 50, wherein said plasmid is the plasmid of clause 44 or 48.52. The method of clause 50, wherein said baculovirus is the baculovirus of clause 46 or 48.53. A method of determining in a biological sample obtained from an individual the presence or absence of antibodies specific for a rhabdovirus, comprising the steps of:a. contacting the biological sample with a capture reagent immobilized to a solid support, wherein the capture reagent is the polypeptide of any of clauses 1 to 41; andb. determining the presence or absence of said antibodies bound to said capture reagent.54. The method of clause 53, further comprising the steps of:c. separating the biological sample from the immobilized capture reagent;d. contacting the immobilized capture reagent-antibody complex with a detectable agent that binds to the antibody of the reagent-antibody complex; ande. measuring the level of antibody bound to the capture reagent using a detection means for the detectable agent, and wherein the measuring step (e) preferably further comprises a comparison with a standard curve to determine the level of antibody bound to the capture reagent.55. Use of the polypeptide of any one of clauses 1 to 41 in a method for determining in a biological sample obtained from an individual the presence or absence of antibodies specific for a rhabdovirus, wherein said method is preferably the method of clause 53 or 54.
34,641
11858963
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT The invention is based on the surprising finding that a single mutation in the amino acid sequence of PCV2 subtype b (PCV2b) ORF2 protein is sufficient to increase the VLP production levels dramatically, thereby enabling the fast production of an effective PCV2 subunit vaccine. In the work underlying the invention positions of major amino acid differences between PCV2a and PCV2b ORF2 sequences were identified as potential positions for mutation. Within this context, six amino acid positions typical for the PCV2b ORF2 protein were identified, namelyat amino acid position 59 an arginine residue or a lysine residue,at amino acid position 63 an arginine residue or a lysine residue,at amino acid position 88 a proline residue,at amino acid position 151 a threonine residue,at amino acid position 206 an isoleucine residue, andat amino acid position 232 an asparagine residue. As described herein, the numbering of amino acid positions refers to the amino acid sequence of full length wild type PCV2 ORF2 protein (SEQ ID NO:2 or SEQ ID NO:5). Hence, the numbering of the amino positions as mentioned herein is with reference to a wild type PCV2 ORF2 protein sequence having 234 or 233 amino acid residues, including a methionine residue at the (N-terminal) amino acid position 1. Thus, the phrase “wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type PCV2 ORF2 protein”, as used in the context of the present invention, relates to the sequence of a naturally occurring PCV2 ORF2 protein, as exemplarily set forth in SEQ ID NO:2 or SEQ ID NO:5. Mutations of the six amino acid positions typical for PCV2b ORF2 protein unexpectedly showed that one mutation of the position within the amino acid sequence of the BC loop of the PCV2 ORF2 protein, namely a substitution of the arginine residue or lysine residue at position 63, was sufficient to increase the expression of a PCV2 ORF2 protein significantly in comparison to a PCV2 ORF2 protein that does not contain such mutation. In one aspect, the invention thus relates to a polypeptide selected from the group consisting of the following (a), (b), and (c): (a) a PCV2 ORF2 protein having: at amino acid position 59 an arginine residue or a lysine residue, and/or at amino acid position 88 a proline residue, and/or at amino acid position 151 a threonine residue, and/or at amino acid position 206 an isoleucine residue, and/or at amino acid position 232 an asparagine residue, and having at amino acid position 63 an amino acid residue other than an arginine residue or a lysine residue, wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type PCV2 ORF2 protein; (b) a PCV2 ORF2 protein characterized in that it (i) contains at least one mutation in the BC loop and (ii) is preferably expressed in a significantly higher amount compared to a PCV2 ORF2 protein that does not contain such mutation; and (c) a combination of (a) and (b). Preferably, said polypeptide, which is also termed “polypeptide of the present invention” hereinafter, is an isolated polypeptide. In particular, the polypeptide of the present invention is a non-naturally-occurring polypeptide. According to the first aspect (a), the polypeptide of the invention is thus a PCV2 ORF2 protein having one, two, three, four, or five amino acid residues (single letter code in brackets) selected from the group consisting of an arginine residue (R) or a lysine residue (K) at amino acid position 59, a proline residue (P) at amino acid position 88, a threonine residue (T) at amino acid position 151, an isoleucine residue (I) at amino acid position 206, and an asparagine residue (N) at amino acid position 232, and having at amino acid position 63 an amino acid residue other than an arginine residue or a lysine residue. In particular, the amino acid residue other than an arginine residue or a lysine residue at position 63 is a naturally occurring, preferably a genetically encoded, amino acid residue other than an arginine residue or a lysine residue. Subsequently, also the following abbreviations are used:“R59” as abbreviation for “an arginine residue at amino acid position 59”,“K59” as abbreviation for “a lysine residue at amino acid position 59”,“P88” as abbreviation for “a proline residue at amino acid position 88”,“T151” as abbreviation for “a threonine residue at amino acid position 151”,“I206” as abbreviation for “an isoleucine residue at amino acid position 206”,“N232” as abbreviation for “an asparagine residue at amino acid position 232”. Preferably, the polypeptide according to aspect (a) is thus a PCV2 ORF2 protein having P88,or having T151,or having I206,or having N232,or having R59 or K59,or having P88 and T151,or having P88 and I206,or having P88 and N232,or having P88 and R59 or K59,or having T151 and I206,or having T151 and N232,or having T151 and R59 or K59,or having I206 and N232,or having I206 and R59 or K59,or having N232 and R59 or K59,or having P88 and T151 and I206,or having P88 and T151 and N232,or having P88 and T151 and R59 or K59,or having P88 and I206 and N232,or having P88 and I206 and R59 or K59,or having P88 and N232 and R59 or K59,or having T151 and I206 and N232,or having T151 and I206 and R59 or K59,or having T151 and N232 and R59 or K59,or having I206 and N232 and R59 or K59,or having P88 and T151 and I206 and N232,or having P88 and T151 and I206 and R59 or K59,or having P88 and T151 and N232 and R59 or K59,or having P88 and I206 and N232 and R59 or K59,or having T151 and I206 and N232 and R59 or K59,or having P88 and T151 and I206 and N232 and R59 or K59. More preferably, the polypeptide according to aspect (a) is hence selected from the group consisting ofPCV2 ORF 2 protein having P88,PCV2 ORF 2 protein having T151,PCV2 ORF 2 protein having I206,PCV2 ORF 2 protein having N232,PCV2 ORF 2 protein having R59PCV2 ORF 2 protein having K59,PCV2 ORF 2 protein having P88 and T151,PCV2 ORF 2 protein having P88 and I206,PCV2 ORF 2 protein having P88 and N232,PCV2 ORF 2 protein having P88 and R59,PCV2 ORF 2 protein having P88 and K59,PCV2 ORF 2 protein having T151 and I206,PCV2 ORF 2 protein having T151 and N232,PCV2 ORF 2 protein having T151 and R59,PCV2 ORF 2 protein having T151 and K59,PCV2 ORF 2 protein having I206 and N232,PCV2 ORF 2 protein having I206 and R59,PCV2 ORF 2 protein having I206 and K59,PCV2 ORF 2 protein having N232 and R59,PCV2 ORF 2 protein having N232 and K59,PCV2 ORF 2 protein having P88 and T151 and I206,PCV2 ORF 2 protein having P88 and T151 and N232,PCV2 ORF 2 protein having P88 and T151 and R59,PCV2 ORF 2 protein having P88 and T151 and K59PCV2 ORF 2 protein having P88 and I206 and N232,PCV2 ORF 2 protein having P88 and I206 and R59,PCV2 ORF 2 protein having P88 and I206 and K59,PCV2 ORF 2 protein having P88 and N232 and R59,PCV2 ORF 2 protein having P88 and N232 and K59,PCV2 ORF 2 protein having T151 and I206 and N232,PCV2 ORF 2 protein having T151 and I206 and R59,PCV2 ORF 2 protein having T151 and I206 and K59,PCV2 ORF 2 protein having T151 and N232 and R59,PCV2 ORF 2 protein having T151 and N232 and K59,PCV2 ORF 2 protein having I206 and N232 and R59,PCV2 ORF 2 protein having I206 and N232 and K59,PCV2 ORF 2 protein having P88 and T151 and I206 and N232,PCV2 ORF 2 protein having P88 and T151 and I206 and R59,PCV2 ORF 2 protein having P88 and T151 and I206 and K59,PCV2 ORF 2 protein having P88 and T151 and N232 and R59,PCV2 ORF 2 protein having P88 and T151 and N232 and K59,PCV2 ORF 2 protein having P88 and I206 and N232 and R59,PCV2 ORF 2 protein having P88 and I206 and N232 and K59,PCV2 ORF 2 protein having T151 and I206 and N232 and R59,PCV2 ORF 2 protein having T151 and I206 and N232 and K59,PCV2 ORF 2 protein having P88 and T151 and I206 and N232 and R59, andPCV2 ORF 2 protein having P88 and T151 and I206 and N232 and K59. According to the second aspect (b), the polypeptide of the invention is in particular a PCV2 ORF2 protein characterized in that it (i) contains at least one mutation in the BC loop and (ii) is expressed, in particular in a baculovirus expression system, in a significantly higher amount, preferably in a higher amount by at least a factor 2, more preferably in a higher amount by at least a factor 3, still more preferably in a higher amount by at least a factor 5, yet more preferably in a higher amount by at least a factor 8, compared to a PCV2 ORF2 protein that does not contain such mutation, wherein the PCV2 ORF2 protein that does not contain such mutation preferably has an amino acid sequence identical to the polypeptide of the invention except the at least one mutation in the BC loop. It is thus in particular understood, that the amino acid sequences of both PCV2 ORF2 proteins the expression of which is compared according to this aspect of the invention are identical except said at least one mutation in the BC loop. The term “BC loop”, within the context of the invention, in particular refers to the part of the PCV2 ORF2 amino acid sequence located between the first two N-terminal amino acid stretches folding into B sheet secondary structures, as can be seen in the crystal structure of PCV2 ORF2 protein as published by Khayat et al. J Virol 85:7856-62 (2011), which is incorporated herein by reference. In particular Khayat et al. describes loops connecting β strands BC, DE, FG, and HI as four to nine amino acid residues long, and loops BC and HI as defining knob-like protrusions extending furthest from the PCV capsid surface and decorating the 5-fold axes. To determine if the PCV2 ORF2 protein containing at least one mutation in the BC loop is expressed in a higher amount compared to the PCV2 ORF2 protein that does not contain such mutation, preferably a method as described hereinafter in Example 1 is used. Thus, in one example, to determine if the PCV2 ORF2 protein containing at least one mutation in the BC loop is expressed in a higher amount compared to the PCV2 ORF2 protein that does not contain such mutation, a baculovirus expression system is used in a method comprising the steps of: infecting Sf+ cells with baculovirus at a target MOI of 0.1, allowing the infection to progress for 5-7 days, harvesting by centrifugation at 20,000 g for 20 min to remove cellular debris and insoluble protein, 0.2 μm filtering of the harvest supernatants, and evaluating directly for PCV2 ORF2 expression by western blot using α-PCV2 antibodies. Preferably, said method further comprises the preparation of baculovirus to be used for the step of infecting Sf+ cells at a target MOI of 0.1, and in particular further comprises one or more of the following steps: cloning a coding sequence which encodes the PCV2 ORF2 protein containing at least one mutation in the BC loop into a baculovirus transfer vector, cloning a coding sequence which encodes the PCV2 ORF2 protein that does not contain such mutation into a baculovirus transfer vector, co-transfecting said baculovirus transfer vector including the coding sequence which encodes the PCV2 ORF2 protein containing at least one mutation in the BC loop with baculovirus DNA in Sf9 cells, co-transfecting said baculovirus transfer vector including the coding sequence which encodes the PCV2 ORF2 protein that does not contain such mutation with baculovirus DNA in Sf9 cells. More preferably, said method additionally further comprises one or more of the following steps: checking the resulting recombinant baculovirus for expression of PCV2 ORF2 protein by IFA, preparing an amplified stock of each recombinant baculovirus on Sf+ cells, titrating said amplified stock via the TCID50method to determine the baculoviral titer. In particular, the polypeptide of the invention being a PCV2 ORF2 protein containing at least one mutation in the BC loop is expressed in a higher amount compared to the PCV2 ORF2 protein that does not contain such mutation under the same and/or comparable ambient conditions, preferably in a baculovirus expression system. More particular, said PCV2 ORF2 protein that does not contain such mutation is a wild type PCV2 ORF2 protein. Preferably, the at least one mutation in the BC loop according to the invention is at least one mutation in the region of the amino acid positions 58 to 66 and in particular comprises or consists of a deletion, substitution, and/or an addition of one to 7 amino acid residues in the region of the amino acid positions 60 to 66. More preferably, the at least one mutation in the BC loop is a deletion, substitution, and/or an addition of one amino acid residue at amino acid position 63, wherein a substitution of the amino acid residue at amino acid position 63 by an amino acid residue other than an arginine residue or a lysine residue is most preferred. Still more preferably, the substitution of the amino acid residue at amino acid position 63 by an amino acid residue other than an arginine residue or a lysine residue is a substitution by a naturally occurring, preferably a genetically encoded, amino acid residue other than an arginine residue or a lysine residue. Preferred sequences of the BC loop according to the invention including a substitution of the amino acid residue at amino acid position 63 by an amino acid residue other than an arginine residue or a lysine residue are set forth in SEQ ID NOs: 10-45. Thus, in particular, the at least one mutation in the BC loop in accordance with the invention comprises or is a substitution of an arginine residue or a lysine residue at amino acid position 63 by an amino acid residue other than an arginine residue or a lysine residue. Thus, the PCV2 ORF2 protein that does not contain such mutation, as described herein, preferably has an arginine residue or a lysine residue at amino acid position 63, which is then substituted according to this preferred embodiment of the invention, thereby resulting in a polypeptide according to the invention. Most preferably, the polypeptide of the present invention comprises a sequence selected from the group consisting of SEQ ID NOs: 10-45, wherein said sequence is in particular located at amino acid positions 58 to 66 of the sequence of the polypeptide of the present invention. According to the third aspect (c), the polypeptide of the invention is any combination of the PCV2 ORF2 protein according to aspect (a) and aspect (b), as described herein, and is thus any PCV2 ORF2 protein having:at amino acid position 59 an arginine residue or a lysine residue, and/orat amino acid position 88 a proline residue, and/orat amino acid position 151 a threonine residue, and/orat amino acid position 206 an isoleucine residue, and/orat amino acid position 232 an asparagine residue, and having at amino acid position 63 an amino acid residue other than an arginine residue or a lysine residue, wherein the numbering of the amino acid positions refers to the amino acid sequence of wild type PCV2 ORF2 protein; and being characterized in that it (i) contains at least one mutation in the BC loop and (ii) is preferably expressed in a significantly higher amount compared to a PCV2 ORF2 protein that does not contain such mutation. The term “genetically encoded amino acid residue other than an arginine residue or a lysine residue”, as described in the context of the present invention, in particular refers to an amino acid residue (single letter code in brackets) selected from the group consisting of alanine residue (A), aspartate residue (D), asparagine residue (N), cysteine residue (C), glutamine residue (Q), glutamate residue (E), phenylalanine residue (F), glycine residue (G), histidine residue (H), isoleucine residue (I), leucine residue (L), methionine residue (M), proline residue (P), serine residue (S), threonine residue (T), valine residue (V), tryptophan residue (W), and tyrosine residue (Y). More particular, said amino acid residue other than an arginine residue or a lysine residue amino is selected from the group consisting of amino acid residue with a polar but uncharged side chain, amino acid residue with a hydrophobic side chain, and glycine residue, wherein preferably the amino acid residue with a polar but uncharged side chain is selected from the group consisting of serine residue, threonine residue, tyrosine residue, asparagine residue, and glutamine residue, and/or wherein said amino acid residue with a hydrophobic side chain is preferably selected from the group consisting of alanine residue, valine residue, leucine residue, isoleucine residue, phenylalanine residue, and tryptophan residue. Most preferably, the amino acid residue other than an arginine residue or a lysine residue, as mentioned in the context of the present invention, is selected from the group consisting of serine residue and threonine residue. In a further preferred aspect, the polypeptide of the present invention is a recombinant PCV2 ORF2 protein, such as a recombinant baculovirus expressed PCV2 ORF2 protein. The term “recombinant PCV2 ORF2 protein”, as used herein, in particular refers to a protein molecule which is expressed from a recombinant DNA molecule, such as a polypeptide which is produced by recombinant DNA techniques. An example of such techniques includes the case when DNA encoding the expressed protein is inserted into a suitable expression vector, preferably a baculovirus expression vector, which is in turn used to transfect, or in case of a baculovirus expression vector to infect, a host cell to produce the protein or polypeptide encoded by the DNA. The term “recombinant PCV2 ORF2 protein”, as used herein, thus in particular refers to a protein molecule which is expressed from a recombinant DNA molecule. According to a particular example, the recombinant PCV2 ORF2 protein is produced by a method with the following steps: The gene for PCV2 ORF2 is cloned into a baculovirus transfer vector; the transfer vector is used to prepare recombinant baculovirus containing said gene by homologous recombination in insect cells; and the PCV2 ORF2 protein is then expressed in insect cells during infection with the recombinant baculovirus. According to an alternative example, the recombinant PCV2 ORF2 protein is expressed in insect cells from a recombinant expression plasmid. In the case of this alternative example baculovirus is not needed. It is further understood that the term “recombinant PCV2 protein consisting of a sequence” in particular also concerns any cotranslational and/or posttranslational modification or modifications of the sequence affected by the cell in which the polypeptide is expressed. Thus, the term “recombinant PCV2 ORF2 protein consisting of a sequence”, as described herein, is also directed to the sequence having one or more modifications effected by the cell in which the polypeptide is expressed, in particular modifications of amino acid residues effected in the protein biosynthesis and/or protein processing, preferably selected from the group consisting of glycosylations, phosphorylations, and acetylations. Preferably, the recombinant PCV2 ORF2 protein according to the invention is produced or obtainable by a baculovirus expression system, in particular in cultured insect cells. In another preferred aspect, the polypeptide of the present invention is a PCV2 subtype b (PCV2b) ORF2 protein. In yet a further preferred aspect, the polypeptide of the present invention is a PCV2 ORF2 protein comprising or consisting of an amino acid sequence having at least 90%, preferably at least 92%, more preferably at least 94%, even more preferably at least 96%, still more preferably at least 98%, or in particular 100% sequence identity with the amino acid sequence of SEQ ID NO: 1. Most preferably, the polypeptide of the present invention is selected from the group consisting of the sequences of SEQ ID NOs: 6-9, which are also shown inFIG.8. Thus, the polypeptide of the present invention is preferably selected from the following sequences (i)-(iv): (i)(SEQ ID NO: 6)MTYPRRRXRRRRHRPRSHLGQILRRRPWLVHPRHRYRWRRKNGIFNTRLSRTXGYTXKRTTVXTPSWXVDMMRFNINDFLPPGGGSNPXXVPFEYYRIRKVKVEFWPCSPITQGDRGVGSXAVILDDNFVTKAXALTYDPYVNYSSRHTITQPFSYHSRYFTPKPVLDXTIDYFQPNNKRNQLWLRLQTXGNVDHVGLGTAFENSIYDQYNIRXTMYVQFREFNLKDPPLNP,(ii)(SEQ ID NO: 7)MTYPRRRXRRRRHRPRSHLGQILRRRPWLVHPRHRYRWRRKNGIFNTRLSRTXGYTXKKTTVXTPSWXVDMMRFNINDFLPPGGGSNPXXVPFEYYRIRKVKVEFWPCSPITQGDRGVGSXAVILDDNFVTKAXALTYDPYVNYSSRHTITQPFSYHSRYFTPKPVLDXTIDYFQPNNKRNQLWLRLQTXGNVDHVGLGTAFENSIYDQYNIRXTMYVQFREFNLKDPPLNP,(iii)(SEQ ID NO: 8)MTYPRRRXRRRRHRPRSHLGQILRRRPWLVHPRHRYRWRRKNGIFNTRLSRTXGYTXKRTTVXTPSWXVDMMRFNINDFLPPGGGSNPXXVPFEYYRIRKVKVEFWPCSPITQGDRGVGSXAVILDDNFVTKAXALTYDPYVNYSSRHTITQPFSYHSRYFTPKPVLDXTIDYFQPNNKRNQLWLRLQTXGNVDHVGLGTAFENSIYDQYNIRXTMYVQFREFNLKDPPLNPX,(iv)(SEQ ID NO: 9)MTYPRRRXRRRRHRPRSHLGQILRRRPWLVHPRHRYRWRRKNGIFNTRLSRTXGYTXKKTTVXTPSWXVDMMRFNINDFLPPGGGSNPXXVPFEYYRIRKVKVEFWPCSPITQGDRGVGSXAVILDDNFVTKAXALTYDPYVNYSSRHTITQPFSYHSRYFTPKPVLDXTIDYFQPNNKRNQLWLRLQTXGNVDHVGLGTAFENSIYDQYNIRXTMYVQFREFNLKDPPLNPX, wherein in said sequences (i)-(iv):“X” is any amino acid residue selected from the group consisting of A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, and Y;“X” is any amino acid residue selected from the group consisting of A, C, D, E, F, G, H, I, L, M, N, P, Q, S, T, V, W, and Y; and“x” is any amino acid residue selected from the group consisting of D and E. For explanatory purposes and in a non-limiting example, the polypeptide according to the invention is a polypeptide consisting of the sequence: (SEQ ID NO: 46)MTYPRRRFRRRRHRPRSHLGQILRRRPWLVHPRHRYRWRRKNGIFNTRLSRTIGYTVKKTTVXTPSWNVDMMRFNINDFLPPGGGSNPLTVPFEYYRIRKVKVEFWPCSPITQGDRGVGSTAVILDDNFVTKANALTYDPYVNYSSRHTITQPFSYHSRYFTPKPVLDRTIDYFQPNNKRNQLWLRLQTTGNVDHVGLGTAFENSIYDQDYNIRITMYVQFREFNLKDPPLNPK,wherein “X” is any amino acid residue selected from the group consisting of A, C, D, E, F, G, H, I, L, M, N, P, Q, S, T, V, W, and Y. In still another preferred aspect of the present invention, the wild type PCV2 ORF2 protein, as described herein, is the protein set forth in SEQ ID NO: 2. According to another aspect, the invention further provides an immunogenic composition containing the polypeptide of the present invention. According to another preferred aspect, the invention further provides an immunogenic composition containing the polypeptide of the present invention, and a PCV2a ORF-2 polypeptide, wherein said PCV2a ORF-2 polypeptide is preferably a polypeptide that is at least 94% or preferably at least 95% identical to the sequence of SEQ ID NO: 3. According to a further aspect, the invention also provides a polynucleotide comprising a sequence which encodes the polypeptide of the present invention, wherein said polynucleotide according to the invention is preferably an isolated polynucleotide. For explanatory purposes and in a non-limiting example, the polynucleotide according to the invention is a polynucleotide comprising the sequence set forth in SEQ ID NO: 4. Production of the polynucleotides described herein is within the skill in the art and can be carried out according to recombinant techniques described, among other places, in Sam brook et al., 2001, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Amusable, et al., 2003, Current Protocols In Molecular Biology, Greene Publishing Associates & Wiley Interscience, NY; Innis et al. (eds), 1995, PCR Strategies, Academic Press, Inc., San Diego; and Erlich (ed), 1994, PCR Technology, Oxford University Press, New York, all of which are incorporated herein by reference. Also, the invention in particular provides a baculovirus which contains a polynucleotide comprising a sequence which encodes the polypeptide of the present invention, wherein said baculovirus according to the invention is preferably an isolated baculovirus. Further, the invention also provides a plasmid, preferably an expression vector, which comprises a polynucleotide comprising a sequence which encodes the polypeptide of the present invention, wherein said plasmid according to the invention is in particular an isolated plasmid. The invention also provides a cell comprising a baculovirus which contains a polynucleotide comprising a sequence which encodes the polypeptide of the present invention, or a plasmid, preferably an expression vector, which comprises a polynucleotide comprising a sequence which encodes the polypeptide of the present invention, wherein said cell according to the invention is preferably an isolated cell. In still another aspect, the invention also relates to the use of the polypeptide of the present invention; the baculovirus according to the invention; the immunogenic composition according to the invention; the polynucleotide according to the invention; the plasmid according to the invention; and/or the cell according to the invention for the preparation of a medicament, preferably of a vaccine. In this context, the invention also provides a method of producing the polypeptide of the present invention of, wherein said method comprises the step of infecting a cell, preferably an insect cell, with the baculovirus of the invention. Further, the invention also provides a method of producing the polypeptide of the present invention, wherein said method comprises the step of transfecting a cell with the plasmid according to the invention. The polypeptide of the present invention is preferably expressed in high amounts sufficient for the stable self-assembly of virus like particles, which may then be used for a single shot vaccination, in particular if they are contained in an immunogenic composition, thereby allowing the reduction and prevention of clinical signs caused by an infection with PCV2, such as an infection with PCV2b and/or PCV2a. The invention is thus in particular further based on the polypeptide of the present invention or on the immunogenic composition according to the invention, respectively, wherein said polypeptide of the present invention or said immunogenic composition comprising the polypeptide of the present invention may be used for particular purposes. In one aspect, the invention thus relates to the polypeptide of the present invention or an immunogenic composition comprising the polypeptide of the present invention for use in a method for the treatment or prevention of an infection with PCV2, the reduction, prevention or treatment of clinical signs caused by an infection with PCV2, or the prevention or treatment of a disease caused by an infection with PCV2. The invention also provides a method for the treatment or prevention of an infection with PCV2, the reduction, prevention or treatment of clinical signs caused by an infection with PCV2, or the prevention or treatment of a disease caused by an infection with PCV2, comprising administering the polypeptide of the present invention or an immunogenic composition comprising the polypeptide of the present invention to an animal, in particular to an animal in need thereof. Also, the invention provides the use of the polypeptide of the present invention or of an immunogenic composition comprising the polypeptide of the present invention for the preparation of a medicament for the treatment or prevention of an infection with PCV2, the reduction, prevention or treatment of clinical signs caused by an infection with PCV2, or the treatment or prevention of a disease caused by an infection with PCV2. In a preferred aspect, the infection with PCV2, as described herein, is an infection with PCV2 subtype b (PCV2b) and/or an infection with PCV2 of a subtype other than subtype 2b. As used herein, the term “infection with PCV2” is equivalent to the term “PCV2 infection”. In particular, the infection with PCV2 of a subtype other than subtype 2b, as mentioned herein, is an infection with PCV2 subtype a (PCV2a) and/or PCV2 subtype c (PCV2c), and is preferably an infection with PCV2a. The term “PCV2 subtype b (PCV2b) ORF2 protein”, as described herein, relates to the protein encoded by the ORF2 gene of a PCV-2b as defined by the standardized nomenclature for PCV2 genotype definition (Segales, J. et al., 2008, PCV-2 genotype definition and nomenclature, Vet Rec 162:867-8) which is incorporated herein by reference). According to another preferred aspect, the infection with PCV2 of a subtype other than subtype 2b, as described herein, is a concurrent infection with (i) PCV2 of a subtype other than subtype 2b and (ii) PCV2b, in particular a concurrent infection with PCV2a and PCV2b. The terms “PCV2a”, “PCV2b” and “PCV2c”, respectively, as described herein, relate to PCV-2a, PCV-2b and PCV-2c, respectively, according to the standardized nomenclature for PCV2 genotype definition (Segales, J. et al., 2008, PCV-2 genotype definition and nomenclature, Vet Rec 162:867-8, which is incorporated herein by reference). In particular, the infection with PCV2b, as mentioned herein, is an infection with (i) a PCV2 comprising a polypeptide that is at least 94%, preferably at least 95%, more preferably at least 96%, still more preferably at least 97%, yet more preferably at least 98%, and most preferably at least 99% identical to the sequence of SEQ ID NO: 2 or (ii) a PCV2 comprising a polynucleotide which comprises a sequence encoding a polypeptide that is at least 94%, preferably at least 95%, more preferably at least 96%, still more preferably at least 97%, yet more preferably at least 98%, and most preferably at least 99% identical to the sequence of SEQ ID NO:2. As used herein, it is in particular understood that the term “identical to the sequence of SEQ ID NO: X” is equivalent to the term “identical to the sequence of SEQ ID NO: X over the length of SEQ ID NO: X” or to the term “identical to the sequence of SEQ ID NO: X over the whole length of SEQ ID NO: X, respectively. In this context, X” is any integer selected from 1 to 3 so that “SEQ ID NO: X” represents any of the SEQ ID NOs mentioned herein. Preferably, the infection with PCV2a, as described herein, is an infection with (i) a PCV2 comprising a polypeptide that is at least 94%, preferably at least 95%, more preferably at least 96%, still more preferably at least 97%, yet more preferably at least 98%, and most preferably at least 99% identical to the sequence of SEQ ID NO:3 or (ii) a PCV2 comprising a polynucleotide which comprises a sequence encoding a polypeptide that is at least 94%, preferably at least 95%, more preferably at least 96%, still more preferably at least 97%, yet more preferably at least 98%, and most preferably at least 99% identical to the sequence of SEQ ID NO:3. Preferably, in the context of the present invention, the treatment or prevention of an infection with PCV2 is based on or comprises or consists of the induction of an immune response against said PCV2, the clinical signs, as mentioned herein, are selected from the group consisting of lymphoid depletion, lymphoid inflammation, positive IHC for PCV2 antigen of lymphoid tissue, viremia, nasal shedding, pyrexia, reduced average daily weight gain, lung inflammation, positive IHC for PCV2 antigen of lung tissue, and/or the disease, as mentioned herein, is PMWS. In particular, in the context of the present invention, the treatment or prevention of an infection with PCV2 of a subtype other than 2b is based on or comprises or consists of the induction of an immune response against said PCV2 of a subtype other than 2b or the concurrent induction of an immune response against said PCV2 of a subtype other than 2b and PCV2b. The term “prevention” or “reduction” or “preventing” or “reducing”, respectively, as used herein, means, but is not limited to a process which includes the administration of a PCV2 antigen, namely of the polypeptide of the present invention, which is included in the composition of the invention, to an animal, wherein said PCV2 antigen, when administered to said animal elicits or is able to elicit an immune response in said animal against PCV2. Altogether, such treatment results in reduction of the clinical signs of a disease caused by PCV2 or of clinical signs associated with PCV2 infection, respectively. More specifically, the term “prevention” or “preventing”, as used herein, means generally a process of prophylaxis in which an animal is exposed to the immunogenic composition of the present invention prior to the induction or onset of the disease process caused by PCV2. Herein, “reduction of clinical signs associated with PCV2 infection” means, but is not limited to, reducing the number of infected subjects in a group, reducing or eliminating the number of subjects exhibiting clinical signs of infection, or reducing the severity of any clinical signs that are present in the subjects, in comparison to wild-type infection. For example, it should refer to any reduction of pathogen load, pathogen shedding, reduction in pathogen transmission, or reduction of any clinical sign symptomatic of PCV2 infection. Preferably these clinical signs are reduced in subjects receiving the composition of the present invention by at least 10% in comparison to subjects not receiving the composition and may become infected. More preferably, clinical signs are reduced in subjects receiving the composition of the present invention by at least 20%, preferably by at least 30%, more preferably by at least 40%, and even more preferably by at least 50%. The term “reduction of viremia” means, but is not limited to, the reduction of PCV2 virus entering the bloodstream of an animal, wherein the viremia level, i.e., the number of PCV2 RNA copies per mL of blood serum or the number of plaque forming colonies per deciliter of blood serum, is reduced in the blood serum of subjects receiving the composition of the present invention by at least 50% in comparison to subjects not receiving the composition and may become infected. More preferably, the viremia level is reduced in subjects receiving the composition of the present invention by at least 90%, preferably by at least 99.9%, more preferably by at least 99.99%, and even more preferably by at least 99.999%. As used herein, the term “viremia” is particularly understood as a condition in which PCV2 particles reproduce and circulate in the bloodstream of an animal. The term “animal”, as used herein, in particular relates to a mammal, preferably to swine, more preferably to a pig, most preferably to a piglet. According to a particular preferred aspect of the invention, the polypeptide of the present invention or the immunogenic composition according to the invention is administered only once. Preferably, in the context of the present invention, the polypeptide of the present invention or the immunogenic composition according to the invention is to be administered or is administered, respectively, in particular only once, to an animal, preferably to a swine, more preferably to a pig, in particular preferably to a piglet. The present invention overcomes the problems inherent in the prior art and provides a distinct advance in the state of the art. According to another aspect, the present invention also provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in animals, preferably animals having anti-PCV2 antibodies, comprising the step of administering an effective amount of the polypeptide of the present invention or the immunogenic composition according to the invention to that animal in need of such treatment. The terms “vaccine” or “immunogenic composition” (both terms are used synonymously) as used herein refers to any pharmaceutical composition containing the polypeptide of the present invention, which composition can be used to prevent or treat a PCV2 infection-associated disease or condition in a subject. A preferred immunogenic composition can induce, stimulate or enhance the immune response against PCV2. The term thus encompasses both subunit immunogenic compositions, as described below, as well as compositions containing whole killed, or attenuated and/or inactivated PCV2b mutant. It is in particular understood that the term “PCV2b mutant”, as described herein, relates to a PCV2b mutant comprising the polypeptide of the present invention and/or the polynucleotide according to the invention. According to another aspect, the present invention also provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in animals, preferably animals having anti-PCV2 antibodies, in particular maternal derived anti-PCV2 antibodies, comprising the step of administering an effective amount of the polypeptide of the present invention or an immunogenic composition comprising the polypeptide of the present invention to that animal in need of such treatment, wherein the immunogenic composition is subunit immunogenic composition, a compositions containing whole killed, or attenuated and/or inactivated PCV2b. The term “subunit immunogenic composition” as used herein refers to a composition containing at least one immunogenic polypeptide or antigen, but not all antigens, derived from or homologous to an antigen from a PCV2b mutant. Such a composition is substantially free of intact PCV2b mutant. Thus, a “subunit immunogenic composition” is prepared from at least partially purified or fractionated (preferably substantially purified) immunogenic polypeptides from a PCV2b mutant, or recombinant analogs thereof. A subunit immunogenic composition can comprise the subunit antigen or antigens of interest substantially free of other antigens or polypeptides from a PCV2b mutant, or in fractionated from. A preferred immunogenic subunit composition comprises the polypeptide of the present invention as described herein. An “immune response” means but is not limited to the development in a host of a cellular and/or antibody-mediated immune response to the composition or vaccine of interest. Usually, an “immune response” includes but is not limited to one or more of the following effects: the production or activation of antibodies, B cells, helper T cells, suppressor T cells, and/or cytotoxic T cells, directed specifically to an antigen or antigens included in the composition or vaccine of interest. Preferably, the host will display either a therapeutic or a protective immunological (memory) response such that resistance to new infection will be enhanced and/or the clinical severity of the disease reduced. Such protection will be demonstrated by either a reduction in number or severity of, or lack of one or more of the signs associated with PCV2 infections, in particular an infection with PCV2 subtype b (PCV2b) and/or an infection with PCV2 of a subtype other than subtype 2b, in delay of onset of viremia, in a reduced viral persistence, in a reduction of the overall viral load and/or a reduction of viral excretion. The term “antigen” as used herein refer to an amino acid sequence which elicits an immunological response as described above. According to a further aspect, the immunogenic composition as used herein most preferably comprises the polypeptide of the present invention, or a fragment thereof, expressed by the polypeptide according to the invention. A preferred polypeptide of the present invention is that of SEQ ID NO: 1. However, it is understood by those of skill in the art that this sequence could vary by as much as 1-5% in sequence homology and still retain the antigenic characteristics that render it useful in immunogenic compositions according to invention. “Sequence identity” as it is known in the art refers to a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, namely a reference sequence and a given sequence to be compared with the reference sequence. Sequence identity is determined by comparing the given sequence to the reference sequence after the sequences have been optimally aligned to produce the highest degree of sequence similarity, as determined by the match between strings of such sequences. Upon such alignment, sequence identity is ascertained on a position-by-position basis, e.g., the sequences are “identical” at a particular position if at that position, the nucleotides or amino acid residues are identical. The total number of such position identities is then divided by the total number of nucleotides or residues in the reference sequence to give % sequence identity. Sequence identity can be readily calculated by known methods, including but not limited to, those described in Computational Molecular Biology, Lesk, A. N., ed., Oxford University Press, New York (1988), Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York (1993); Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey (1994); Sequence Analysis in Molecular Biology, von Heinge, G., Academic Press (1987); Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M. Stockton Press, New York (1991); and Carillo, H., and Lipman, D., SIAM J. Applied Math., 48: 1073 (1988), the teachings of which are incorporated herein by reference. Preferred methods to determine the sequence identity are designed to give the largest match between the sequences tested. Methods to determine sequence identity are codified in publicly available computer programs which determine sequence identity between given sequences. Examples of such programs include, but are not limited to, the GCG program package (Devereux, J., et al., Nucleic Acids Research, 12(1): 387 (1984)), BLASTP, BLASTN and FASTA (Altschul, S. F. et al., J. Molec. Biol., 215:403-410 (1990). The BLASTX program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S. et al., NCVI NLM NIH Bethesda, Md. 20894, Altschul, S. F. et al., J. Molec. Biol., 215:403-410 (1990), the teachings of which are incorporated herein by reference). These programs optimally align sequences using default gap weights in order to produce the highest level of sequence identity between the given and reference sequences. As an illustration, by a polynucleotide having a nucleotide sequence having at least, for example, 85%, preferably 90%, even more preferably 95% “sequence identity” to a reference nucleotide sequence, it is intended that the nucleotide sequence of the given polynucleotide is identical to the reference sequence except that the given polynucleotide sequence may include up to 15, preferably up to 10, even more preferably up to 5 point mutations per each 100 nucleotides of the reference nucleotide sequence. In other words, in a polynucleotide having a nucleotide sequence having at least 85%, preferably 90%, even more preferably 95% identity relative to the reference nucleotide sequence, up to 15%, preferably 10%, even more preferably 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 15%, preferably 10%, even more preferably 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. These mutations of the reference sequence may occur at the 5′ or 3′ terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence. Analogously, by a polypeptide having a given amino acid sequence having at least, for example, 85%, preferably 90%, even more preferably 95% sequence identity to a reference amino acid sequence, it is intended that the given amino acid sequence of the polypeptide is identical to the reference sequence except that the given polypeptide sequence may include up to 15, preferably up to 10, even more preferably up to 5 amino acid alterations per each 100 amino acids of the reference amino acid sequence. In other words, to obtain a given polypeptide sequence having at least 85%, preferably 90%, even more preferably 95% sequence identity with a reference amino acid sequence, up to 15%, preferably up to 10%, even more preferably up to 5% of the amino acid residues in the reference sequence may be deleted or substituted with another amino acid, or a number of amino acids up to 15%, preferably up to 10%, even more preferably up to 5% of the total number of amino acid residues in the reference sequence may be inserted into the reference sequence. These alterations of the reference sequence may occur at the amino or the carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in the one or more contiguous groups within the reference sequence. Preferably, residue positions which are not identical differ by conservative amino acid substitutions. However, conservative substitutions are not included as a match when determining sequence identity. “Sequence homology”, as used herein, refers to a method of determining the relatedness of two sequences. To determine sequence homology, two or more sequences are optimally aligned, and gaps are introduced if necessary. However, in contrast to “sequence identity”, conservative amino acid substitutions are counted as a match when determining sequence homology. In other words, to obtain a polypeptide or polynucleotide having 95% sequence homology with a reference sequence, 85%, preferably 90%, even more preferably 95% of the amino acid residues or nucleotides in the reference sequence must match or comprise a conservative substitution with another amino acid or nucleotide, or a number of amino acids or nucleotides up to 15%, preferably up to 10%, even more preferably up to 5% of the total amino acid residues or nucleotides, not including conservative substitutions, in the reference sequence may be inserted into the reference sequence. Preferably the homolog sequence comprises at least a stretch of 50, even more preferably at least 100, even more preferably at least 250, and even more preferably at least 500 nucleotides. A “conservative substitution” refers to the substitution of an amino acid residue or nucleotide with another amino acid residue or nucleotide having similar characteristics or properties including size, hydrophobicity, etc., such that the overall functionality does not change significantly. “Isolated” means altered “by the hand of man” from its natural state, i.e., if it occurs in nature, it has been changed or removed from its original environment, or both. For example, a polynucleotide or polypeptide naturally present in a living organism is not “isolated,” but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is “isolated”, as the term is employed herein. Thus, according to a further aspect, the present invention also provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in animals, preferably animals having anti-PCV2 antibodies, in particular maternal derived anti-PCV2 antibodies, comprising the step of administering an effective amount of the polypeptide of the present invention or an immunogenic composition comprising the polypeptide of the present invention to that animal in need of such treatment, wherein said the polypeptide of the present invention is anyone of those, described herein. Preferably, the polypeptide of the present invention protein is: (i) a polypeptide comprising or consisting of the sequence of SEQ ID NO: 1; or (ii) any polypeptide that is at least 95% homologous to the polypeptide of (i). According to a further aspect, the polypeptide of the present invention is provided in the immunogenic composition at a protein inclusion level effective for inducing the desired immune response, namely reducing the incidence of, lessening the severity of, or preventing or reducing one or more clinical signs resulting from or associated with a PCV2 infection. Preferably, the inclusion level of the polypeptide of the present invention is at least 0.2 μg protein/ml of the final immunogenic composition (μg/ml), more preferably from about 0.2 to about 400 μg/ml, still more preferably from about 0.3 to about 200 μg/ml, even more preferably from about 0.35 to about 100 μg/ml, still more preferably from about 0.4 to about 50 μg/ml, still more preferably from about 0.45 to about 30 μg/ml, still more preferably from about 0.5 to about 18 μg/ml, even more preferably from about 0.6 to about 15 μg/ml even more preferably from about 0.75 to about 8 μg/ml, even more preferably from about 1.0 to about 6 μg/ml, still more preferably from about 1.3 to about 3.0 μg/ml, even more preferably from about 1.4 to about 2.5 μg/ml, even more preferably from about 1.5 to about 2.0 μg/ml, and most preferably about 1.6 μg/ml. According to a further aspect, the protein inclusion level is at least 0.2 μg/PCV2b ORF-2 protein as described above per dose of the final immunogenic composition (μg/dose), more preferably from about 0.2 to about 400 μg/dose, still more preferably from about 0.3 to about 200 μg/dose, even more preferably from about 0.35 to about 100 μg/dose, still more preferably from about 0.4 to about 50 μg/dose, still more preferably from about 0.45 to about 30 μg/dose, still more preferably from about 0.5 to about 18 μg/dose, even more preferably from about 0.6 to about 15 μg/ml, even more preferably from about 0.75 to about 8 μg/dose, even more preferably from about 1.0 to about 6 μg/dose, still more preferably from about 1.3 to about 3.0 μg/dose, even more preferably from about 1.4 to about 2.5 μg/dose, even more preferably from about 1.5 to about 2.0 μg/dose, and most preferably about 1.6 μg/dose. Also, an inclusion level of the polypeptide of the present invention (antigen content) of less than 20 μg/dose, preferably of about 0.5 to 18 μg/dose is suitable to confer immunity in young animals and/or in animals which are positive for PCV2 antibodies, in particular which are positive for anti-PCV2 maternal derived antibodies. Thus, according to a further aspect, the present invention also provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in animals, preferably animals having anti-PCV2 antibodies, in particular maternal derived anti-PCV2 antibodies, comprising the step of administering less than 20 μg/dose, preferably of about 0.5 to 18 μg/dose of the polypeptide of the present invention or an immunogenic composition comprising the polypeptide of the present invention to that animal in need of such treatment. Said polypeptide of the present invention is anyone described in this patent application. The polypeptide of the present invention used in the immunogenic composition in accordance with the present invention can be derived in any fashion including isolation and purification of the polypeptide of the present invention, standard protein synthesis, and recombinant methodology. Preferred methods for obtaining the polypeptide of the present invention are provided in WO06/072065, the teachings and content of which are hereby incorporated by reference in its entirety, since surprisingly it has been found that the methods described therein for obtaining PCV2a ORF-2 polypeptide can be used accordingly for obtaining the polypeptide of the present invention. Briefly, susceptible cells are infected with a recombinant viral vector containing DNA coding sequences encoding the polypeptide of the present invention, the polypeptide of the present invention protein is expressed by the recombinant virus, and the expressed polypeptide of the present invention is recovered from the supernatant by filtration and inactivated by any conventional method, preferably using binary ethylenimine, which is then neutralized to stop the inactivation process. The immunogenic composition as used herein also refers to a composition that comprises i) any of the polypeptides of the present invention described above, preferably in concentrations described above, and ii) at least a portion of the viral vector expressing said polypeptide of the present invention, preferably of a recombinant baculovirus. Moreover, the immunogenic composition can comprise i) any of the polypeptides of the present invention described above, preferably in concentrations described above, ii) at least a portion of the viral vector expressing said polypeptide of the present invention, preferably of a recombinant baculovirus, and iii) a portion of the cell culture supernatant. Thus, according to a further aspect, the present invention also provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in animals, preferably animals having anti-PCV2 antibodies, in particular maternal derived anti-PCV2 antibodies, comprising the step of administering an effective amount of the polypeptide of the present invention or an immunogenic composition comprising the polypeptide of the present invention to that animal in need of such treatment, wherein the polypeptide of the present invention is a recombinant, preferably a baculovirus expressed, polypeptide of the present invention. Preferably those recombinant or baculovirus expressed polypeptides of the present invention having the sequence as described above. The immunogenic composition as used herein also refers to a composition that comprises i) any of the polypeptides of the present invention described above, preferably in concentrations described above, ii) at least a portion of the viral vector expressing said polypeptide of the present invention, preferably of a recombinant baculovirus, and iii) a portion of the cell culture; wherein about 90% of the components have a size smaller than 1 μm. The immunogenic composition as used herein also refers to a composition that comprises i) any of the polypeptides of the present invention described above, preferably in concentrations described above, ii) at least a portion of the viral vector expressing said polypeptide of the present invention, iii) a portion of the cell culture, iv) and inactivating agent to inactivate the recombinant viral vector preferably BEI, wherein about 90% of the components i) to iii) have a size smaller than 1 μm. Preferably, BEI is present in concentrations effective to inactivate the baculovirus, preferably in an amount of 2 to about 8 mM BEI, preferably of about 5 mM BEI. The immunogenic composition as used herein also refers to a composition that comprises i) any of the polypeptides of the present invention described above, preferably in concentrations described above, ii) at least a portion of the viral vector expressing said polypeptide of the present invention, iii) a portion of the cell culture, iv) an inactivating agent to inactivate the recombinant viral vector preferably BEI, and v) an neutralization agent to stop the inactivation mediated by the inactivating agent, wherein about 90% of the components i) to iii) have a size smaller than 1 μm. Preferably, if the inactivating agent is BEI, said composition comprises sodium thiosulfate in equivalent amounts to BEI. The protein is incorporated into a composition that can be administered to an animal susceptible to PCV2 infection. In preferred forms, the composition may also include additional components known to those of skill in the art (see also Remington's Pharmaceutical Sciences. (1990). 18th ed. Mack Publ., Easton). Additionally, the composition may include one or more veterinary-acceptable carriers. As used herein, “a veterinary-acceptable carrier” includes any and all solvents, dispersion media, coatings, adjuvants, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, adsorption delaying agents, and the like. In a preferred embodiment, the immunogenic composition comprises the polypeptide of the present invention as provided herewith, preferably in concentrations described above, which is mixed with an adjuvant, preferably Carbopol, and physiological saline. Those of skill in the art will understand that the composition used herein may incorporate known injectable, physiologically acceptable sterile solutions. For preparing a ready-to-use solution for parenteral injection or infusion, aqueous isotonic solutions, such as e.g. saline or corresponding plasma protein solutions are readily available. In addition, the immunogenic and vaccine compositions of the present invention can include diluents, isotonic agents, stabilizers, or adjuvants. Diluents can include water, saline, dextrose, ethanol, glycerol, and the like. Isotonic agents can include sodium chloride, dextrose, mannitol, sorbitol, and lactose, among others. Stabilizers include albumin and alkali salts of ethylendiamintetracetic acid, among others. “Adjuvants” as used herein, can include aluminum hydroxide and aluminum phosphate, saponins e.g., Quil A, QS-21 (Cambridge Biotech Inc., Cambridge Mass.), GPI-0100 (Galenica Pharmaceuticals, Inc., Birmingham, Ala.), water-in-oil emulsion, oil-in-water emulsion, water-in-oil-in-water emulsion. The emulsion can be based in particular on light liquid paraffin oil (European Pharmacopea type); isoprenoid oil such as squalane or squalene oil resulting from theoligomerization of alkenes, in particular of isobutene or decene; esters of acids or of alcohols containing a linear alkyl group, more particularly plant oils, ethyl oleate, propylene glycol di-(caprylate/caprate), glyceryl tri-(caprylate/caprate) or propylene glycol dioleate; esters of branched fatty acids or alcohols, in particular isostearic acid esters. The oil is used in combination with emulsifiers to form the emulsion. The emulsifiers are preferably nonionic surfactants, in particular esters of sorbitan, of mannide (e.g., anhydromannitol oleate), of glycol, of polyglycerol, of propylene glycol and of oleic, isostearic, ricinoleic or hydroxystearic acid, which are optionally ethoxylated, and polyoxypropylene-polyoxyethylene copolymer blocks, in particular the Pluronic products, especially L121. See Hunter et al., The Theory and Practical Application of Adjuvants (Ed. Stewart-Tull, D. E. S.). John Wiley and Sons, NY, pp 51-94 (1995) and Todd et al., Vaccine 15:564-570 (1997). For example, it is possible to use the SPT emulsion described on page 147 of “Vaccine Design, The Subunit and Adjuvant Approach” edited by M. Powell and M. Newman, Plenum Press, 1995, and the emulsion MF59 described on page 183 of this same book. A further instance of an adjuvant is a compound chosen from the polymers of acrylic or methacrylic acid and the copolymers of maleic anhydride and alkenyl derivative. Advantageous adjuvant compounds are the polymers of acrylic or methacrylic acid which are cross-linked, especially with polyalkenyl ethers of sugars or polyalcohols. These compounds are known by the term carbomer (Pharmeuropa Vol. 8, No. 2, June 1996). Persons skilled in the art can also refer to U.S. Pat. No. 2,909,462 which describes such acrylic polymers cross-linked with a polyhydroxylated compound having at least 3 hydroxyl groups, preferably not more than 8, the hydrogen atoms of at least three hydroxyls being replaced by unsaturated aliphatic radicals having at least 2 carbon atoms. The preferred radicals are those containing from 2 to 4 carbon atoms, e.g., vinyls, allyls and other ethylenically unsaturated groups. The unsaturated radicals may themselves contain other substituents, such as methyl. The products sold under the name Carbopol; (BF Goodrich, Ohio, USA) are particularly appropriate. They are cross-linked with an allyl sucrose or with allyl pentaerythritol. Among them, there may be mentioned Carbopol 974P, 934P and 971P. Most preferred is the use of Carbopol, in particular the use of Carbopol 971P, preferably in amounts of about 500 μg to about 5 mg per dose, even more preferred in an amount of about 750 μg to about 2.5 mg per dose and most preferred in an amount of about 1 mg per dose. Further suitable adjuvants include, but are not limited to, the RIBI adjuvant system (Ribi Inc.), Block co-polymer (CytRx, Atlanta Ga.), SAF-M (Chiron, Emeryville Calif.), monophosphoryl lipid A, Avridine lipid-amine adjuvant, heat-labile enterotoxin fromE. coli(recombinant or otherwise), cholera toxin, IMS 1314, or muramyl dipeptide among many others. Preferably, the adjuvant is added in an amount of about 100 μg to about 10 mg per dose. Even more preferably, the adjuvant is added in an amount of about 100 μg to about 10 mg per dose. Even more preferably, the adjuvant is added in an amount of about 500 μg to about 5 mg per dose. Even more preferably, the adjuvant is added in an amount of about 750 μg to about 2.5 mg per dose. Most preferably, the adjuvant is added in an amount of about 1 mg per dose. Additionally, the composition can include one or more pharmaceutical-acceptable carriers. As used herein, “a pharmaceutical-acceptable carrier” includes any and all solvents, dispersion media, coatings, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, adsorption delaying agents, and the like. Most preferably, the composition provided herewith, contains polypeptide of the present invention recovered from the supernatant of in vitro cultured cells, wherein said cells were infected with a recombinant viral vector containing DNA encoding the polypeptide of the present invention and expressing the polypeptide of the present invention, and wherein said cell culture was treated with about 2 to about 8 mM BEI, preferably with about 5 mM BEI to inactivate the viral vector, and an equivalent concentration of a neutralization agent, preferably sodium thiosulfate solution to a final concentration of about 2 to about 8 mM, preferably of about 5 mM. The present invention also relates to an immunogenic composition that comprises i) any of the polypeptides of the present invention described above, preferably in concentrations described above, ii) at least a portion of the viral vector expressing said polypeptide of the present invention, iii) a portion of the cell culture, iv) an inactivating agent to inactivate the recombinant viral vector preferably BEI, and v) an neutralization agent to stop the inactivation mediated by the inactivating agent, preferably sodium thiosulfate in equivalent amounts to BEI; and vi) a suitable adjuvant, preferably Carbopol 971 in amounts described above; wherein about 90% of the components i) to iii) have a size smaller than 1 μm. According to a further aspect, this immunogenic composition further comprises a pharmaceutical acceptable salt, preferably a phosphate salt in physiologically acceptable concentrations. Preferably, the pH of said immunogenic composition is adjusted to a physiological pH, meaning between about 6.5 and 7.5. The immunogenic composition as used herein also refers to a composition that comprises per one ml (i) at least 1.6 μg of the polypeptide of the present invention described above, preferably less than 20 μg (ii) at least a portion of baculovirus expressing said polypeptide of the present invention (iii) a portion of the cell culture, (iv) about 2 to 8 mM BEI, (v) sodium thiosulfate in equivalent amounts to BEI; and (vi) about 1 mg Carbopol 971, and (vii) phosphate salt in a physiologically acceptable concentration; wherein about 90% of the components (i) to (iii) have a size smaller than 1 μm and the pH of said immunogenic composition is adjusted to about 6.5 to 7.5. The immunogenic compositions can further include one or more other immuno-modulatory agents such as, e.g., interleukins, interferons, or other cytokines. The immunogenic compositions can also include Gentamicin and Merthiolate. While the amounts and concentrations of adjuvants and additives useful in the context of the present invention can readily be determined by the skilled artisan, the present invention contemplates compositions comprising from about 50 μg to about 2000 μg of adjuvant and preferably about 250 μg/ml dose of the vaccine composition. Thus, the immunogenic composition as used herein also refers to a composition that comprises from about 1 ug/ml to about 60 μg/ml of antibiotics, and more preferably less than about 30 μg/ml of antibiotics. The immunogenic composition as used herein also refers to a composition that comprises (i) any of the polypeptides of the present invention described above, preferably in concentrations described above; (ii) at least a portion of the viral vector expressing said polypeptide of the present invention; (iii) a portion of the cell culture; (iv) an inactivating agent to inactivate the recombinant viral vector preferably BEI; and (v) an neutralization agent to stop the inactivation mediated by the inactivating agent, preferably sodium thiosulfate in equivalent amounts to BEI; (vi) a suitable adjuvant, preferably Carbopol 971 in amounts described above; (vii) a pharmaceutical acceptable concentration of a saline buffer, preferably of a phosphate salt; and (viii) an anti-microbiological active agent; wherein about 90% of the components (i) to (iii) have a size smaller than 1 μm. For investigation of a possible interference of the polypeptide of the present invention with the maternal antibody a study may be conducted in which the antibody titers of study animals are determined at the time of vaccination which are then grouped into a low, moderate and high antibody class: Geometric mean titers of <1:100 are considered as low antibody titers, titers of 1:100 to 1:1000 are considered as moderate antibody titers and titers of >1:1000 are considered as high antibody titers. This grouping pattern is comparable to that done in a Canadian field study where antibody titers of 1:80 were considered as low, antibody titers of 1:640 as moderate and antibody titers of >1:1280 as high (Larochelle et al., 2003, Can. J. Vet. Res.; 67: 114-120). In order to analyze the impact of low, medium and high antibody titers at the time of vaccination on viremia, vaccinated and placebo-treated animals are compared with regard to the onset, end, duration of viremia, the number of positive sampling days and the virus load. The presence of anti-PCV2 antibodies, in particular of maternal derived antibodies, preferably has no significant impact of any of those parameters. In other words, the efficacy of the polypeptide of the present invention in prevention and treatment of a PCV2 infection or in reduction of clinical signs caused by or associated with a PCV2 infection in animals is preferably not affected at the day of vaccination by the presence of anti-PCV2 antibodies, preferably by anti-PCV2 antibody titers of up to 1:100, preferably of more than 1:100, even more preferably of more than 1:250, even more preferably of more than 1:500, even more preferably of 1:640; even more preferably of more than 1:750, most preferably of more than 1:1000. This effect can be shown in a one shot vaccination experiment, which means that the polypeptide of the present invention is administered only once and without any subsequent administration of the polypeptide of the present invention. Methods for detection and quantification of anti-PCV2 antibodies are well known in the art. For example detection and quantification of PCV2 antibodies can be performed by indirect immunofluorescence as described in Magar et al., 2000, Can. J. Vet Res.; 64: 184-186 or Magar et al., 2000, J. Comp. Pathol.; 123: 258-269. Further assays for quantification of anti-PCV2 antibodies are described in Opriessnig et al., 2006, 37th Annual Meeting of the American Association of Swine Veterinarians. Moreover, an indirect immunofluorescence assay, that can be used by a person skilled in the art comprises the steps of: seeding about 20.000 to 60.000 PK15 or VIDO R1 cells per well onto a 96 well plate; infecting cells with a PCV2 isolate, when monolayers are approximately 65 to 85% confluent; incubating infected cells for 48 hours; removing medium and washing cells 2 times with PBS; discarding the wash buffer and treating cells with cold 50/50 methanol/acetone fixative (−100 μl/well) for about 15 min at about −20° C.; discarding the fixative and air drying of the plates; preparing serial dilutions of porcine serum samples in PBS and serial dilutions of an anti-PCV2 positive and negative control sample (Positive Control and Negative Control Samples); adding the serial dilutions to the plates and incubating to allow antibodies to bind if present in the serum samples for about 1 hr. at 36.5±1° C.; washing the plates three times with PBS an discarding the PBS; staining the plates with a commercial Goat anti-Swine FITC conjugate diluted 1:100 in PBS and incubated for about 1 hr. at 36.5±1° C.; removing microplates are removed from incubator, the conjugate is discarded and the plates are washed 2 times with PBS; reading the plates using UV microscopy and reporting individual wells as positive or negative, wherein the Positive Control and Negative Control samples are used to monitor the test system; and calculating the serum antibody titers using the highest dilution showing specific IFA reactivity and the number of wells positive per dilution, or a 50% endpoint is calculated using the appropriate Reed-Muench formula. Such an assay is described in Example 2 of WO 2008/076915 A2. In cases of controversial results and in any question of doubt, anti-PCV2 titers as mentioned herein, refer to those which are/can be estimated by this assay. Thus according to a further aspect, the present invention provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in animals, preferably animals having anti-PCV2 antibodies, in particular maternal antibodies, comprising the step of administering an effective amount of a polypeptide of the present invention to that animal in need of such treatment, preferably of less than 20 μg/dose wherein said animal have a detectable anti-PCV2 antibody titer of up to 1:100, preferably of more than 1:100, even more preferably of more than 1:250, even more preferably of more than 1:500, even more preferably of 1:640, even more preferably of more than 1:750, most preferably of more than 1:1000. Preferably, those anti-PCV2 antibody titer is detectable and quantifiable in a specific anti-PCV2 immune assay, preferably in the assay as described above, as exemplarily described in Example 2 of WO 2008/076915 A2. More preferably, those anti-PCV-2 antibodies are maternal derived antibodies. Most preferably, the polypeptide of the present invention is only administered once, preferably with a dose of less than 20 μg/dose. Piglets with only low titers (<1:100) or moderate titers (<1:1000) of maternal derived anti-PCV2 antibodies are not sufficient protected against PCV2 infections which occur prior to week 3 of age. Therefore, vaccination at a very early stage of life is desirable. Within the context of the invention, vaccination/treatment of animals at or before 3 weeks of age is preferred. Moreover, anti-PCV2 antibody titers of more than 1:1000 preferably have no influence on the efficacy of the PCV2 vaccine regardless of the level of the existing initial antibody titer. For example, vaccination of high-titer animals (anti-PCV2 antibody titer>1:1000) preferably result in a shorter duration of viremia, an earlier end of viremia, less viremic sampling days and a reduction of the sum of genomic equivalents/ml as compared to non-vaccinated control animals. Upon comparison of vaccinated “high”, “moderate” and “low titer animals” no significant differences are preferably observed with regard to the different parameters of PCV2 viraemia. Also in the presence of high anti-PCV2 antibody titers the polypeptide of the present invention used for vaccination preferably still significantly reduces viremia in blood (end of viremia, duration of viremia, virus load). Preferably, no differences are found with regard to the live body weight when comparing low and high titer animals of the vaccinated group. Furthermore, vaccinated animals with a high anti-PCV2 antibody titer at the time of vaccination/treatment (>1:1000) also preferably show a significantly higher body weight after the onset of viremia compared to placebo-treated animals with initial high antibody titers. Consequently, according to a preferred aspect, vaccination/treatment of animals of 1 day of age or older with the polypeptide of the present invention is possible. However, vaccination should be done within the first 8, preferably within the first 7 weeks of age. Thus according to a further aspect, the present invention provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in animals, comprising the step of administering to that animal in need of such treatment at day 1 of age or later, preferably but not later than at week 8 of age an effective amount of the polypeptide of the present invention. According to a preferred embodiment, less than 20 μg/dose polypeptide of the present invention are required to confer immunity in such animal. According to a more preferred embodiment, the polypeptide of the present invention, preferably less than a 20 μg/dose thereof is only administered once to the animal in need of such treatment. According to a further, more general aspect, the present invention provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in young animals, comprising the step of administering an effective amount of the polypeptide of the present invention to that animal in need of such treatment. The term “young animal” as used herein refers to an animal of 1 to 22 days of age. Preferably, by the term young animal, an animal of 1 to 20 days of age is meant. More preferably, the term young animal refers to an animal of 1 to 15 days of age, even more preferably of 1 day of age to 14 days of age, even more preferably of 1 to 12 days of age, even more preferably of 1 to 10 days of age, even more preferably of 1 to 8 days of age, even more preferably of 1 to 7 days of age, even more preferably of 1 to 6 days of age, even more preferably of 1 to 5 days of age, even more preferably of 1 to 4 days of age, even more preferably of 1 to 3 days of age, even more preferably of 1 or 2 day(s) of age, most preferably to an animal of 1 day of age. Thus according to a further aspect, the present invention provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in young animals, comprising the step of administering an effective amount of the polypeptide of the present invention to an animal of 1 to 22 days of age, preferably of 1 to 20 days of age, more preferably of 1 to 15 days of age, even more preferably of 1 to 14 days of age, even more preferably of 1 to 12 days of age, even more preferably of 1 to 10 days of age, even more preferably of 1 to 8 days of age, even more preferably of 1 to 7 days of age, even more preferably of 1 to 6 days of age, even more preferably of 1 to 5 days of age, even more preferably of 1 to 4 days of age, even more preferably of 1 to 3 days of age, even more preferably of 1 or 2 day(s) of age, most preferably at 1 day of age in need of such treatment. For example, the vaccination/treatment on 19 to 22 days of age preferably shows high efficacy of vaccination. Moreover, vaccination/treatment at 12 to 18, preferably 12 to 14 days of age is preferably very effective in reduction of clinical signs associated with PCV2 infections, reduction of overall viral load, reduction of duration of viremia, delay in onset of viremia, weight gain. Moreover, vaccination at 1 week of age is preferably very effective in reduction of clinical signs associated with PCV2 infections, reduction of overall viral load, reduction of duration of viremia, delay in onset of viremia, weight gain. Preferably less than 20 μg/dose of the polypeptide of the present invention is required to confer immunity in those young animals. According to a more preferred embodiment, the polypeptide of the present invention, preferably less than 20 μg is only administered once to that young animal in need of such treatment. Due to the ubiquity of PCV2 in the field most of the young piglets are seropositive in respect to PCV2. Thus according to a further aspect, the present invention provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in young animals, preferably animals having anti-PCV2 antibodies at the day of vaccination, comprising the step of administering an effective amount of the polypeptide of the present invention to an animal of 1 to 22 days of age, preferably of 1 to 20 days of age, more preferably of 1 to 15 days of age, even more preferably of 1 to 14 days of age, even more preferably of 1 to 12 days of age, even more preferably of 1 to 10 days of age, even more preferably of 1 to 8 days of age, even more preferably of 1 to 7 days of age, even more preferably of 1 to 6 days of age, even more preferably of 1 to 5 days of age, even more preferably of 1 to 4 days of age, even more preferably of 1 to 3 days of age, even more preferably at 1 or 2 day(s) of age, most preferably at 1 day of age in need of such treatment. Preferably, said young animals, at the day of vaccination/treatment, have a detectable anti-PCV2 antibody titer of up to 1:100, preferably of more than 1:100, even more preferably of more than 1:250, even more preferably of more than 1:500, even more preferably of 1:640, even more preferably of more than 1:750, most preferably of more than 1:1000 at the day of vaccination/treatment. Preferably less than 20 μg/dose of the polypeptide of the present invention are required to confer a sufficient immunity in those young animals. According to more preferred embodiment, the polypeptide of the present invention, preferably less than 20 μg is only administered once to that young animal in need of such treatment. As described above, vaccination/treatment of young animals with the polypeptide of the present invention preferably results in shortening of viremic phase as compared to non-vaccinated control animals. The average shortening time may preferably, for instance, be 9.5 days as compared to non-vaccinated control animals of the same species. Therefore, according to a further aspect, the present invention also provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in young animals, comprising the step of administering an effective amount of the polypeptide of the present invention to that animal in need of such treatment, wherein the treatment or prevention results in shortening of the viremia phase of 5 or more days, preferably 6 or more days, even more preferably of 7 or more days, even more preferably of 8 or more days, even more preferably of 9, even more preferably of 10, even more preferably of 12, even more preferably of 14, most preferably of more than 16 days as compared to animals of a non-treated control group of the same species. In some cases, the viremic phase is preferably shortening for more than 20 days. In general, the vaccination of young piglets preferably results in a reduction in the loss of weight gain, a shorter duration of viremia, an earlier end to viremia, and a lower virus load. Therefore, according to a further aspect, the present invention provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in young animals, comprising the step of administering an effective amount of the polypeptide of the present invention to that animal in need of such treatment, wherein said treatment or prevention of PCV2 infection results in an improvement in comparison to animals of a non-treated control group of the same species in a vaccine efficacy parameter selected from the group consisting of a reduction in the loss of weight gain, a shorter duration of viremia, an earlier end to viremia, a lower virus load, or combinations thereof. Preferably less than 20 μg/dose polypeptide of the present invention are required to cause any of the improved vaccine efficacy parameter mentioned above. Moreover such improved vaccine efficacy parameter are achieved by a singly administration of only one dose. The term “an effective amount” as used herein means but is not limited to an amount of the polypeptide of the present invention, that elicits or is able to elicit an immune response in an animal, to which said effective dose of the polypeptide of the present invention is administered. Preferably, an effective amount is defined as an amount of the polypeptide of the present invention that confers at least a 10 weeks duration of immunity (DOI), preferably at least a 12 weeks (DOI), more preferably at least a 15 weeks (DOI), most preferably at least a 20 weeks (DOI). The amount that is effective depends on the ingredients of the vaccine and the schedule of administration. Typically, when an inactivated virus or a modified live virus preparation is used in the combination vaccine, an amount of the vaccine containing about 102.0to about 109.0TCID50per dose, preferably about 103.0to about 108.0TCID50per dose, more preferably, about 104.0to about 108.0TCID50per dose. In particular, when modified live PCV2 is used in the vaccines, the recommended dose to be administered to the susceptible animal is preferably about 103.0TCID50(tissue culture infective dose 50% end point)/dose to about 106.0TCID50/dose and more preferably about 104.0TCID50/dose to about 105.0TCID50/dose. In general, the quantity of antigen will be between 0.2 and 5000 micrograms, and between 102.0and 109.0TCID50, preferably between 103.0and 106.0TCID50, more preferably between 104.0and 105.0TCID50, when purified antigen is used. Sub-unit vaccines are normally administered with an protein inclusion level of at least 0.2 μg protein per dose, preferably with about 0.2 to about 400 μg/dose, still more preferably with about 0.3 to about 200 μg/dose, even more preferably with about 0.35 to about 100 μg/dose, still more preferably with about 0.4 to about 50 μg/dose, still more preferably with about 0.45 to about 30 μg/dose, still more preferably with about 0.5 to about 18 μg/dose, still more preferably with about 0.6 to about 16 μg/dose, even more preferably with about 0.75 to about 8 μg/dose, even more preferably with about 1.0 to about 6 μg/dose, still more preferably with about 1.3 to about 3.0 μg/dose. Preferably, the prophylactic use of the immunogenic compositions described supra, is effective for reduction of clinical signs caused by or associated with PCV2 infections, preferably in young animals and/or in animals having passive immunity against PCV2 at the day of treatment. In particular, the prophylactic use of the immunogenic compositions as described herein, and specifically of compositions comprising the polypeptide of the present invention, is preferably effective for reducing lymphadenopathy, lymphoid depletion and/or multinucleated/giant histiocytes in animals infected with PCV2 and having maternal anti-PCV-2 antibodies at the day of treatment/vaccination. For example it was discovered that the prophylactic use of the immunogenic compositions as described herein is effective for reducing lymphoid depletion, lymphoid inflammation, positive IHC for PCV2 antigen of lymphoid tissue, viremia, nasal shedding, pyrexia, reduced average daily weight gain, lung inflammation, positive IHC for PCV2 antigen of lung tissue. Furthermore, the prophylactic use of the immunogenic compositions as described herein is preferably effective for reducing (1) interstitial pneumonia with interlobular edema, (2) cutaneous pallor or icterus, (3) mottled atrophic livers, (4) gastric ulcers, (5) nephritis and (6) reproductive disorders, e.g. abortion, stillbirths, mummies, etc., (7) Pia like lesions, normally known to be associated with Lawsonia intracellularis infections (leitis), (8) lymphadenopathy, (9) lymphoid depletion and/or (10) multinucleated/giant histiocytes (11) Porcine Dermatitis and Nephropathy Syndrome (PDNS), (12) PCVAD associated mortality, (13) PCVAD associated weight loss, (14), reduced growth variability (15), reduced frequency of ‘runts’ (16), reduced co-infections with Porcine Reproductive and Respiratory Disease Complex (PRRSV). Such immunogenic composition is also effective in improving economically important growth parameters such as time to slaughter, carcass weight, and lean meat ratio. Thus the term “clinical signs” as used herein, means, but is not limited to (1) interstitial pneumonia with interlobular edema, (2) cutaneous pallor or icterus, (3) mottled atrophic livers, (4) gastric ulcers, (5) nephritis and (6) reproductive disorders, e.g. abortion, stillbirths, mummies, etc., (7) Pia-like lesions, normally known to be associated with Lawsonia intracellularis infections (Ileitis), (8) lymphadenopathy, (9) lymphoid depletion and/or (10) multinucleated/giant histiocytes (11) Porcine Dermatitis and Nephropathy Syndrome (PDNS), (12) PCVAD associated mortality, (13) PCVAD associated weight loss, (14) reduced growth variability (15) reduced frequency of ‘runts’ (16) reduced co-infections with Porcine Reproductive and Respiratory Disease Complex (PRRSV), (17) lymphoid inflammation, (18) positive IHC for PCV2 antigen of lymphoid tissue, (19) viremia, (20) nasal shedding, (21) pyrexia, (22) reduced average daily weight gain, (23) lung inflammation, (24) positive IHC for PCV2 antigen of lung tissue. Moreover, the immunogenic composition described herein reduces the overall circovirus load including a later onset, a shorter duration, an earlier end of viremia, and a reduced viral load and its immunosuppressive impact in young animals, in particular in those having anti-PCV2 antibodies at the day of vaccination, thereby resulting in a higher level of general disease resistance and a reduced incidence of PCV2 associated diseases and clinical signs. Thus, according to a further aspect, the present invention provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in young animals and/or in animals, preferably animals having anti-PCV2 antibodies, comprising the step of administering an effective amount of the polypeptide of the present invention or an immunogenic composition comprising the polypeptide of the present invention to that animal in need of such treatment, wherein those clinical signs are selected from the group consisting of: (1) interstitial pneumonia with interlobular edema, (2) cutaneous pallor or icterus, (3) mottled atrophic livers, (4) gastric ulcers, (5) nephritis and (6) reproductive disorders, e.g. abortion, stillbirths, mummies, etc., (7) Pia-like lesions, normally known to be associated with Lawsonia intracellularis infections (leitis), (8) lymphadenopathy, (9) lymphoid depletion and/or (10) multinucleated/giant histiocytes (11), Porcine Dermatitis and Nephropathy Syndrome (PDNS), (12) PCVAD associated mortality, (13) PCVAD associated weight loss, (14) reduced growth variability (15) reduced frequency of ‘runts’, (16) reduced co-infections with Porcine Reproductive and Respiratory Disease Complex (PRRSV), (17) lymphoid inflammation, (18) positive IHC for PCV2 antigen of lymphoid tissue, (19) viremia, (20) nasal shedding, (21) pyrexia, (22) reduced average daily weight gain, (23) lung inflammation, (24) positive IHC for PCV2 antigen of lung tissue. According to a further aspect, the present invention provides a method for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in young animals, comprising the step of administering an effective amount of the polypeptide of the present invention to that animal in need of such treatment, wherein those clinical signs are selected from the group consisting of: (1) interstitial pneumonia with interlobular edema, (2) cutaneous pallor or icterus, (3) mottled atrophic livers, (4) gastric ulcers, (5) nephritis and (6) reproductive disorders, e.g. abortion, stillbirths, mummies, etc., (7) Pia-like lesions, normally known to be associated with Lawsonia intracellularis infections (Ileitis), (8) lymphadenopathy, (9) lymphoid depletion and/or (10) multinucleated/giant histiocytes (11) Porcine Dermatitis and Nephropathy Syndrome (PDNS), (12) PCVAD associated mortality, (13) PCVAD associated weight loss, (14) reduced growth variability (15) reduced frequency of ‘runts’ (16) reduced co-infections with Porcine Reproductive and Respiratory Disease Complex (PRRSV), (17) lymphoid inflammation, (18) positive IHC for PCV2 antigen of lymphoid tissue, (19) viremia, (20) nasal shedding, (21) pyrexia, (22) reduced average daily weight gain, (23) lung inflammation, (24) positive IHC for PCV2 antigen of lung tissue. The composition according to the invention may be applied, orally, intradermally, intratracheally, or intravaginally. The composition preferably may be applied intramuscularly or intranasally, most preferably intramuscularly. In an animal body, it can prove advantageous to apply the pharmaceutical compositions as described above via an intravenous or by direct injection into target tissues. For systemic application, the intravenous, intravascular, intramuscular, intranasal, intraarterial, intraperitoneal, oral, or intrathecal routes are preferred. A more local application can be effected subcutaneously, intradermally, intracutaneously, intracardially, intralobally, intramedullarly, intrapulmonarily or directly in or near the tissue to be treated (connective-, bone-, muscle-, nerve-, epithelial tissue). Depending on the desired duration and effectiveness of the treatment, the compositions according to the invention may be administered once or several times, also intermittently, for instance on a daily basis for several days, weeks or months and in different dosages. Preferably, one dose of the immunogenic composition as described above is intramuscularly administered to the subject in need thereof. According to a further aspect, the polypeptide of the present invention or the immunogenic composition comprising any such polypeptide of the present invention as described herein is bottled in and administered at one (1) mL per dose. Thus, according to a further aspect, the present invention also provides a 1 ml immunogenic composition, comprising the polypeptide of the present invention as described herein, for the treatment or prevention of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in young animals, comprising the step of administering an effective amount of the polypeptide of the present invention protein to that animal in need of such treatment. According to a further aspect, the present invention also provides a 1 ml immunogenic composition, comprising the polypeptide of the present invention as described herein, for the treatment or prophylaxis of a PCV2 infection or for reduction of clinical signs caused by or associated with a PCV2 infection in animals, preferably animals having anti-PCV2 antibodies, comprising the step of administering an effective amount of the polypeptide of the present invention or an immunogenic composition comprising the polypeptide of the present invention to that animal in need of such treatment. According to a further aspect, at least one further administration of at least one dose of the immunogenic composition as described above is given to a subject in need thereof, wherein the second or any further administration is given at least 14 days beyond the initial or any former administrations. Preferably, the immunogenic composition is administered with an immune stimulant. Preferably, said immune stimulant is given at least twice. Preferably, at least 3 days, more preferably at least 5 days, even more preferably at least 7 days are in between the first and the second or any further administration of the immune stimulant. Preferably, the immune stimulant is given at least 10 days, preferably 15 days, even more preferably 20, even more preferably at least 22 days beyond the initial administration of the immunogenic composition provided herein. A preferred immune stimulant is, for example, keyhole limpet hemocyanin (KLH), preferably emulsified with incomplete Freund's adjuvant (KLH/ICFA). However, it is herewith understood, that any other immune stimulant known to a person skilled in the art can also be used. The term “immune stimulant” as used herein, means any agent or composition that can trigger the immune response, preferably without initiating or increasing a specific immune response, for example the immune response against a specific pathogen. It is further instructed to administer the immune stimulant in a suitable dose. DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS The following examples set forth preferred materials and procedures in accordance with the present invention. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods, devices, and materials are now described. It is to be understood, however, that these examples are provided by way of illustration only, and nothing therein should be deemed a limitation upon the overall scope of the invention. Example 1 Materials & Procedure/Design of Mutants The PCV2a ORF2 amino acid sequence of the PCV2 ORF2 protein included in the product CIRCOFLEX® was aligned with the PCV2b ORF2 BDH amino acid sequence and a number of other published PCV2a and PCV2b ORF2 amino acid sequences from Genbank using the Clustal W method. Positions of major amino acid differences between PCV2a and PCV2b ORF2 sequences were identified as potential positions for mutation (seeFIG.1). Using the identified major amino acid changes, seven PCV2b ORF2 coding sequences were prepared exchanging the amino acid from PCV2b ORF2 BDH for the corresponding amino acid from PCV2a ORF2. PCV2a ORF2 (CIRCOFLEX) codons were used to code for the mutant amino acids. The seven PCV2b ORF2 mutant constructs are detailed below:1. PCV2b ORF2 BDH K59A2. PCV2b ORF2 BDH R63T3. PCV2b ORF2 BDH R63K4. PCV2b ORF2 BDH SFCO P88K T151P**5. PCV2b ORF2 BDH G191R6. PCV2b ORF2b BDH I206K7. PCV2b ORF2 BDH N232E All coding sequences were synthesized at Integrated DNA Technologies except #4 which was created by site-directed mutagenesis of a synthesized PCV2b ORF2b BDH SFCO coding sequence. ** SFCO=codon optimized forSpodoptera frugiperda. This construct was created prior to the alignment described above through a preliminary sequence assessment. The two mutations were also identified in this sequence assessment. Preparation of Mutant PCV2b ORF2 Baculovirus Each of the seven PCV2b ORF2 mutant coding sequences, as well as the unmutated PCV2b ORF2 BDH coding sequence, were cloned into baculovirus transfer vector pVL1393 and co-transfected with baculovirus DNA in Sf9 cells. Each resulting recombinant baculovirus was checked for PCV2b ORF2 expression by IFA. Amplified stocks of each recombinant baculovirus were prepared on Sf+ cells and titrated via the TCID50method to determine the baculoviral titer. Expression Evaluation of Mutant PCV2b ORF2 Baculovirus Each recombinant baculovirus was evaluated for expression of its PCV2b ORF2 coding sequence by infecting Sf+ cells at a target MOI of 0.1. The infections were allowed to progress for 5-7 days then were harvested by centrifugation at 20,000 g for 20 min to remove cellular debris and insoluble protein. The harvest supernatants were 0.2 μm filtered and evaluated directly for PCV2b ORF2 expression by western blot using α-PCV2 antibodies (e.g.FIG.2). The harvest supernatants were also evaluated for the presence of macromolecular structures. Briefly, a sample of each harvest supernatant was centrifuged at 100,000 g for two hours. The resulting pellets were resuspended in a small volume of TBS and separated by SDS-PAGE. PCV2b ORF2 bands were detected in stained gels by size comparison to PCV2a ORF2 (e.g.FIG.3). Resuspended pellets were also separated on a 10%-60% discontinuous sucrose gradient by centrifugation at 100,000 g for two hours to partially purify the PCV2b ORF2 proteins for quantitation and VLP confirmation by electron microscopy (EM) (e.g.,FIG.4). After sucrose gradient separation, the PCV2b ORF2 containing fractions were pooled and the PCV2b ORF2 concentration was determined by SDS-PAGE gel densitometry compared to a BSA standard curve. In addition, a sample of the sucrose gradient-purified material was further concentrated and submitted for VLP confirmation by EM using phosphotungstic acid as a negative stain (e.g.,FIG.5). A table of the results from the evaluation of the PCV2b ORF2 BDH mutant constructs is shown inFIG.6. The results demonstrated that a single amino acid mutation from arginine to threonine at position 63 increased expression of PCV2b ORF2 BDH in Sf+ cells nearly ten-fold. The single R63T mutation increased PCV2b ORF2 BDH expression in Sf+ cells to levels similar to PCV2a ORF2. An analysis of the amino acid sequence of PCV2b ORF2 BDH suggests that the BC loop may be susceptible to cleavage by trypsin-like proteases. Structural data published by Khayat et al. in 2011 suggests that arginine 63 is on the BC loop that projects out furthest from the PCV2 viral capsid formed by the ORF2 protein, leaving it susceptible to proteases released after the lysis of Sf+ cells during baculovirus replication. In addition to threonine substitution at position 63, in another embodiment of the invention the arginine is substituted by other uncharged polar amino acids including serine, tyrosine, asparagine and glutamine to obtain the same stabilizing effect. In addition, nonpolar amino acids including glycine, alanine, valine, leucine, isoleucine, phenylalanine and tryptophan may achieve the same effect as well. Example 2 This study demonstrates the efficacy of one embodiment of the Porcine Circovirus Type 2 ORF2b Vaccine against a PCV2a and/or PCV2b challenge. Cesarean-derived colostrum-deprived (CDCD) piglets are used in this study and separated into 2 groups; 1) pigs vaccinated with an experimental Porcine Circovirus Vaccine including the PCV2b ORF2 R63T variant of Example 1 (Killed Baculovirus Vector) that are challenged with virulent PCV2b and, 2) non-vaccinated challenged control pigs that are challenged with virulent PCV2b. On Day 0, Group 1 is administered 1 mL of vaccine intramuscularly (IM) whereas Group 2, non-vaccinated challenge control pigs do not receive any treatment. On Day 28, all pigs in groups 1 and 2 are challenged with virulent PCV2b 1 mL intranasally (IN) and 1 mL IM with an approximate dosage of 3.0 Log10TCID5/mL of live virus. All pigs receive 2.0 mL Keyhole Limpet Hemocyanin emulsified in Incomplete Freunds Adjuvant (KLH/ICFA) IM on Days 25 and 31. Pigs are monitored daily for clinical signs, and blood is drawn for serologic testing periodically. On Day 56 all pigs are necropsied and select tissues are collected and gross pathology observations are made. As a whole, vaccinated animals exhibit reduction when compared to their respective challenge control group in all parameters tested. Example 3 Several other substitutions at amino acid site 63 were produced to compare to the PCV2b ORF BDH native strain. The results from the evaluation of the PCV2b ORF2 BDH mutant constructs are shown inFIGS.7A and7B. The results demonstrate that in addition to the amino acid mutation from arginine (R) to threonine (T) at position 63, arginine (R) 63 to glycine (G), arginine (R) 63 to glutamine (Q), and arginine (R) 63 to aspartate (D) increased the expression of PCV2b ORF2 BDH in Sf+ cells at least Four-fold as compared to the wild type. In particular the single mutations R63G and R63Q increased PCV2b ORF2 BDH expression in Sf+ cells to levels similar to PCV2a ORF2. Generation of Recombinant Baculovirus Encoding PCV2b ORF2 R63 Mutants Point mutations in the coding sequence of PCV2b ORF2 at amino acid position 63 were generated by site-directed mutagenesis. Briefly, baculovirus transfer plasmid pVL1393-PCV2b ORF2 was subjected to site-directed mutagenesis using primers in Table 1. The resulting baculovirus transfer vectors were sequenced to confirm proper mutation of the coding sequence and then co-transfected with linearized baculovirus DNA into Sf9 cells. Co-transfections were harvested after 5 days and evaluated for PCV2b ORF2 expression by IFA using PCV2-specific antibodies. Amplified stocks of each baculovirus were generated on Sf9 cells and titered via an IFA-based TCID50method using an α-baculovirus gp64 monoclonal antibody. TABLE 1Primers for site-directed mutagenesis.PrimerSequenceForward5′-CTGTCAAGAAAACCACAGTCX1X2X3ACGCCCTCCTGGAATGTG-3′ReverseReverse complement of ForwardMutationX1X2X3R63DGACR63QCAGR63GGGAR63LTTGR63TACA Expression and Quantitation of PCV2b ORF2 VLPs SF+ cells in spinner flasks were infected with recombinant baculovirus at an MOI of 0.1 and incubated at 27° C. with constant agitation at approximately 100 rpm. Infected cultures were harvested once SF+ cell viability dropped below 30% or at 7 days post infection. Raw baculovirus harvests were centrifuged at 20,000 g for 20 minutes at 4° C. to pellet cells and insoluble debris and then 0.2 μm filtered. Clarified baculovirus harvest fluids (35 mL) were subjected to centrifugation at 100,000 g RCF for 2 hours at 4° C. to pellet PCV2b ORF2 VLPs. The resulting pellets were resuspended in TBS and further separated on a 10%-60% discontinuous sucrose gradient at 100,000 g RCF for 2 hrs at 4° C. The fractions containing the majority of the PCV2b ORF2, as determined by SDS-PAGE and Western blot utilizing 0-PCV2 antibodies, were pooled and evaluated by densitometry. Briefly, pooled PCV2b ORF2-containing fractions were separated by SDS-PAGE and stained with SIMPLYBLUE™ Safe Stain. Gel images were captured and analyzed using an Alpha View camera and software. The mass of PCV2b ORF2 bands were calculated using a BSA standard curve included on each gel. The PCV2b ORF2 concentration of the pool was calculated by dividing the mass of the PCV2b ORF2 band(s) by the total volume of sample loaded on the gel. PCV2b ORF2 concentrations in harvest material were calculated by multiplying the PCV2b ORF2 concentration in the pool by the volume of the pool and then dividing the result by the starting volume of harvest fluids used for centrifugation. Example 4 This study evaluates the efficacy of Porcine Circovirus Type 2 ORF2b Prototype Vaccine (including recombinant baculovirus expressed PCV2b ORF2 protein of SEQ ID NO: 1) against a PCV2b challenge when given at three weeks of age. Forty two healthy CDCD pigs (X pigs from each of X litters and X pigs from each of X litters) were blocked and housed amongst six pens. Pigs within a pen were equally randomized to 1 of 5 treatment groups: Group 1 (Strict Negative Controls) consisted of X pigs and received no treatment, Group 2 (Challenge Controls, n=X) received no treatment, Group 3 (Experimental PCV2b comprising SEQ ID NO: 1+ carbopol vaccine, n=X), Group 4 (Experimental PCV2b comprising SEQ ID NO: 1+ISA207VG vaccine, n=X). An overview of the treatment groups is provided in Table 2. TABLE 2No.ofDay 11DayGroupPigsTreatmentDay 0and Day 17Day 14421≥5Strict Negn/an/aNecropsyn/aCont2≥20Challengen/aKLH/ICFAPCV2bNec-ControlTreatmentchallengeropsy3≥20PCV2b ORF2VaccinateKLH/ICFAPCV2bNec-protein +TreatmentchallengeropsyCarbopol4>20PCV2b ORF2VaccinateKLH/ICFAPCV2bNec-protein +TreatmentchallengeropsyISA207VG5≥20PCV2a/VaccinateKLH/ICFAPCV2bNec-PCV2b ORF2Treatmentchallengeropsyprotein +Carbopol On D0 pigs were 24 days of age and Group 3 pigs are administered a 1 mL dose of vaccine intramuscularly (IM). On D11 and D17, all pigs receive a 2.0 mL dose of KLH/ICFA, intramuscularly (IM). On D14 all pigs are challenged with approximately 5.0 log10TCID50/mL of live virulent PCV2b 1.0 mL IM in the right neck and 1.0 mL intranasally. Pigs are examined daily for overall health. Blood samples are collected on D-4, D14, D21, D28, D33 and D42, and sera were tested for PCV2 viremia by quantitative real time polymerase chain reaction on all days with the exception of Day −4. Animals vaccinated show significantly lower viremia and reduced to no clinical symptoms compared to non-vaccinated animals after the PCV2b challenge. Within the context of the invention made and the experimental data provided herewith, in particular the following was considered:with respect to lymphoid depletion: to support evidence of “aid in the prevention of lymphoid depletion”, a pig was considered positive if one or more of the 4 lymphoid tissue samples (tonsil, TBLN, MLN or ILN) was histologically positive for lymphoid depletion;with respect to lymphoid inflammation: to support evidence of “aid in the prevention of lymphoid inflammation”, a pig was considered positive if one or more of the 4 lymphoid tissue samples (tonsil, TBLN, MLN or ILN) was histologically positive for lymphoid inflammation;with respect to lymphoid colonization: to support evidence that pigs cleared infection by 4 weeks post-virus exposure, a pig was considered positive if one or more of the 4 lymphoid tissue samples (tonsil, TBLN, MLN or ILN) was positive for PCV2 lymphoid colonization by IHC;with respect to viremia: to support evidence of “aid in the prevention of viremia”, a pig was considered positive on the day of sampling if the serum rt-PCR test was ≥1.0×104 PCV2 genomic equivalents (the linear lower level); andwith respect to mortality: to support evidence of “aid in the prevention of mortality”, a pig was considered positive for mortality if it succumbed to challenge (died or required euthanasia for humane reasons with attributable clinical signs, gross lesions and/or histological lesions consistent with PCV2). In the Sequence Listing:SEQ ID NO: 1 corresponds to SEQ ID NO: 2 including the substitution R63T.SEQ ID NO: 2 corresponds to the sequence of a wild type PCV2b ORF2 protein.SEQ ID NO: 3 corresponds to the sequence of a wild type PCV2a ORF2 protein.SEQ ID NO: 4 corresponds to a polynucleotide sequence encoding SEQ ID NO: 1.SEQ ID NO: 5 corresponds to the sequence of a wild type PCV2b ORF2 protein.SEQ ID NO: 6 corresponds to the sequence of a polypeptide of the present invention being 233 amino acid residues in length and having at amino acid position 59 an arginine residue.SEQ ID NO:7 corresponds to the sequence of a polypeptide of the present invention being 233 amino acid residues in length and having at amino acid position 59 a lysine residue.SEQ ID NO:8 corresponds to the sequence of a polypeptide of the present invention being 234 amino acid residues in length and having at amino acid position 59 an arginine residue.SEQ ID NO:9 corresponds to the sequence of a polypeptide of the present invention being 234 amino acid residues in length and having at amino acid position 59 a lysine residue.SEQ ID NO:10 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 an alanine residue.SEQ ID NO:11 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a cysteine residue.SEQ ID NO:12 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 an aspartate residue.SEQ ID NO:13 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a glutamate residue.SEQ ID NO:14 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a phenylalanine residue.SEQ ID NO:15 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a glycine residue.SEQ ID NO:16 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a histidine residue.SEQ ID NO:17 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 an isoleucine residue.SEQ ID NO:18 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a leucine residue.SEQ ID NO:19 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a methionine residue.SEQ ID NO:20 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 an asparagine residue.SEQ ID NO:21 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a proline residue.SEQ ID NO:22 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a glutamine residue.SEQ ID NO:23 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a serine residue.SEQ ID NO:24 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a threonine residue.SEQ ID NO:25 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a valine residue.SEQ ID NO:26 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a tryptophan residue.SEQ ID NO:27 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 a tyrosine residue.SEQ ID NO:28 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 an arginine residue and having at amino acid position 63 an alanine residue.SEQ ID NO:29 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a cysteine residue.SEQ ID NO:30 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 an aspartate residue.SEQ ID NO:31 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a glutamate residue.SEQ ID NO:32 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a phenylalanine residue.SEQ ID NO:33 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a glycine residue.SEQ ID NO:34 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a histidine residue.SEQ ID NO:35 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 an isoleucine residue.SEQ ID NO:36 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a leucine residue.SEQ ID NO:37 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a methionine residue.SEQ ID NO:38 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 an asparagine residue.SEQ ID NO:39 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a proline residue.SEQ ID NO:40 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a glutamine residue.SEQ ID NO:41 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a serine residue.SEQ ID NO:42 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a threonine residue.SEQ ID NO:43 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a valine residue.SEQ ID NO:44 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a tryptophan residue.SEQ ID NO:45 corresponds to the sequence of amino acid positions 58-66 (also referred to as “BC-loop” herein) of a polypeptide of the present invention having at amino acid position 59 a lysine residue and having at amino acid position 63 a tyrosine residue.SEQ ID NO:46 corresponds to the sequence of a polypeptide of the present invention being 234 amino acid residues in length and having at amino acid position 63 a threonine residue.
117,611
11858964
DETAILED DESCRIPTION Definitions This specification describes exemplary embodiments and applications of the disclosure. This disclosure, however, is not limited to these exemplary embodiments and applications or to the manner in which the exemplary embodiments and applications operate or are described herein. Various embodiments, features, objects, and advantages of the present teachings will be apparent from the description and accompanying drawings, and from the claims. As used herein, the terms “comprise,” “comprises,” “comprising,” “contain,” “contains,” “containing,” “have,” “having,” “include,” “includes,” and “including,” and their variants, are not intended to be limiting, are inclusive or open-ended, and do not exclude additional, unrecited additives, components, integers, elements, or method steps. For example, a process, method, system, composition, kit, or apparatus that comprises a list of features is not necessarily limited only to those features but may include other features not expressly listed or inherent to such process, method, system, composition, kit, or apparatus. “About” is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value. “Immune redirector capsid” or “IRC” as used herein is a capsid backbone that also comprises a peptide bound, attached, or conjugated, to the capsid backbone. “Cleavage sequence” as used herein includes, for example, specific peptide sequences, or more often, peptide motifs at which site-specific proteases cleave or cut the protein. Cleavage sites are used, for example, to cleave off an affinity tag, thereby restoring the natural protein sequence, or to inactivate a protein, or to activate proteins. In the present disclosure “cleavage” refers to proteolytic cleavage. In various embodiments, proteolytic cleavage is catalyzed by peptidases, proteases, or proteolytic cleavage enzymes before the final maturation of the protein. Proteins are also known to be cleaved as a result of intracellular processing of, for example, misfolded proteins. Another example of proteolytic processing of proteins is secretory proteins or proteins targeted to organelles, which have their signal peptide removed by specific signal peptidases before release to the extracellular environment or specific organelle. In one embodiment of the present disclosure, the cleavage sequence is specifically recognized by furin which cleaves and releases the peptides from the IRC, making the peptide available for loading onto or binding by the tumor cell surface receptors. In various embodiments, the cleavage sequence is comprised of cysteine, lysine, and/or arginine residues, that not only allow the peptide to be cleaved from the capsid backbone, but also serve as anchors to conjugate the peptide to the capsid protein until release by the cleavage protein, such as furin, which are in some instances enriched in, or selectively present at, the site of the tumor, i.e., in the tumor microenvironment. “Epitope” or “antigen” or “antigenic epitope” is a set of amino acid residues that create recognition by or are recognized by a particular immunoglobulin or, in the context of T cells, those residues necessary for recognition by T cell receptor proteins and/or major histocompatibility (MHC) receptors. The amino acid residues of an epitope need not be contiguous/consecutive. In an immune system setting, in vivo or in vitro, an epitope are in some instances a composite of the collective features of a molecule, such as primary, secondary, and tertiary peptide structure, and charge, that together form a three-dimensional structure recognized by an immunoglobulin, T cell receptor, and/or human leukocyte (HLA) molecule. “HPV” and “human papillomavirus” refer to the members of the family Papillomaviridae that are capable of infecting humans. There are two major groups of HPVs defined by their tropism (genital/mucosal and cutaneous groups), each of which contains multiple virus “types” or “strains/genotypes,” e.g., HPV 16, HPV 18, HPV 31, HPV 32, etc. “MusPV,” “MMuPV1,” “MPV,” and “mouse papillomavirus,” all alternatively and interchangeably refer to the known members of the family Papillomaviridae that are capable of infecting mice (Mus musculus). “Human vaccine” as used herein means a biological preparation that improves immunity to a particular disease in a human. A vaccine typically contains an antigenic agent(s) that resembles a disease-causing agent (pathogen), and is often made from weakened or killed forms of the microbe, its toxins, or one or multiple immunogenic surface proteins of the disease-causing agent. The antigenic agent stimulates the body's immune system to recognize the disease-causing agent as foreign, destroy it, and “remember” it, so that the immune system can more easily recognize and destroy any of these pathogens should an actual future infection/exposure occur. Human vaccines include vaccines against viral diseases and bacterial diseases. In various embodiments, vaccines against viral diseases include hepatitis A, B, E virus, human papillomavirus, influenza virus, Japanese encephalitis virus, measles virus, mumps virus, polio virus, rabies virus, rotavirus, rubella virus, tick-borne encephalitis virus, varicella zoster virus, variola virus, and yellow fever virus. Human vaccines against viral diseases that are under development include, for example, dengue vaccine, eastern equine encephalitis virus, HTLV-1 T lymphocyte leukemia vaccine, and respiratory syncytial virus vaccine. Such a vaccine includes, in some embodiments, current vaccines in development or currently United States Food and Drug Administration (FDA)-approved vaccinations. A non-limiting list of examples of vaccines that are compatible with the compositions and methods described herein is provided in Table 2. The embodiments described herein, however, are not limited to these listed vaccines, and are contemplated to apply to any vaccine developed to provide immunity in a human subject. “Inhibiting,” “reducing,” “prevention,” or “reducing the occurrence of,” and similar terms, when used herein, includes any measurable decrease or complete inhibition/reduction or elimination to achieve a desired result, such as inhibiting, reducing, or preventing, or reducing the occurrence of, or reducing tumor mass, progression, and/or metastasis. “MHC” or “major histocompatibility complex” is a group of genes that encode proteins found on the surfaces of cells that help the immune system recognize foreign substances. MHC proteins (receptors, or molecules) are expressed by all higher vertebrates. There are two main types of MHC molecules, MHC class I and MHC class II. In humans there are three different genetic loci that encode MHC class I molecules (the MHC-molecules of the human are also designated human leukocyte antigens (HLA)): HLA-A, HLA-B, and HLA-C. HLA-A*01, HLA-A *02, and HLA-A*11 are examples of different MHC class I alleles that can be expressed from these loci. “Papillomavirus” (PV) refers to all members of the papillomavirus family (Papillomaviridae). An extensive list of papillomavirus types and the ability to make the respective capsid backbones can be referenced using this publication: “Classification of papillomaviruses (PVs) based on 189 PV types and proposal of taxonomic amendments,” de Villers et al., 401(1):70-79, 2010, PMID: 20206957 (all the tables specifically incorporated herein by reference for all purposes). “Preferentially cleaved protein” as used herein means that the peptide is preferentially cleaved from the capsid or capsomere or L1 protein at the site of a tumor or tumor microenvironment. Without wishing to be bound by any particular theory, the preferential tumor-site cleavage is in some instances due to: (1) the unique cleavage sequence on the peptide, and/or (2) the unique tumor microenvironment. For example, in one embodiment, the peptide comprises a cleavage sequence that is preferentially cleaved by the enzyme furin, which is known to be expressed in relatively higher concentrations around tumor cells as compared with elsewhere in an organism. “Protein,” “polypeptide,” and “peptide,” as used herein, are not restricted to any particular number of amino acids; these terms are sometimes used interchangeably herein. The properties and amino acid sequences of the proteins described herein, and of the nucleic acids encoding them, are well-known and are determined routinely, as well as downloaded from various known databases. (See, e.g., the NCBI GenBank databases). Some peptide sequences are provided herein. However, some peptide sequence information is routinely updated, e.g., to correct mistakes in the previous entries, so updated (corrected) information about the proteins and nucleic acids encoding them is included in this application. Information provided in the sequence databases discussed herein is incorporated by reference. An immune “response” is a humoral and/or cellular response of the subject's immune system in which, in a cellular response, an antigen-primed cytotoxic T cell, Th1 T cell, Th2 T cell, and/or B cells primed by a vaccine or other pathogen present in the subject, or that the subject was previously exposed to, binds the epitope or antigen. The term “preexisting immune response” as used herein means an immune response that is present in an individual prior to initiation of the inventive cancer treatment methods described herein. Thus, an individual having a preexisting immune response has an immune response capacity stored within their memory T cells or other immune system components against an antigen, prior to the initiation of a method of treatment as described herein with the antigen to treat cancer. A preexisting immune response is in some instances a naturally-occurring immune response. In other instances, the preexisting immune response is an induced immune response. As used herein, a naturally-occurring preexisting immune response is an immune response in an individual that was elicited in response to an antigen, such as a bacterial, fungal, parasitic, or viral antigen, with which the individual unintentionally contacted or contracted. That is, an individual having a preexisting immune response was, in some instances, not exposed to an antigen with the intent to generate an immune response to the antigen. An induced preexisting immune response is an immune response resulting from an intentional exposure to an antigen, such as when receiving a vaccine. The preexisting immune response is in some instances a naturally-occurring immune response, or in other instances the preexisting immune response is an induced immune response. A “subject,” or “subject in need thereof,” as used herein, includes any animal that has a tumor/cancer or has had a tumor/cancer or has a precancerous medical condition or cell or has a genetic or other susceptibility, predisposition, or occupational risk of developing cancer or a tumor. Suitable subjects (patients) include laboratory animals, such as mouse, rat, rabbit, guinea pig, or pig, farm animals, such as cattle, sporting animals, such as dogs or horses, domesticated animals or pets, such as a horse, dog, or cat, nonhuman primates, and humans. “T cell response” as used herein refers to the immune response elicited by T cells as they encounter antigens. Naïve mature T cells are activated upon encountering antigen presented by B cells, macrophages, and dendritic cells, and then thereby produce armed effector T cells. Effector T cells are, in some instances, either CD8+ T cells that differentiate into cytotoxic T cells, or CD4+ T cells that primarily induce the humoral immune response. The T cell immune response further generates immunological memory that gives protection from the subsequent challenge of the subject by the same or a similar pathogen comprising the same or similar epitopes. In various embodiments, the T cell response is at a threshold of at least 2-fold above the baseline of total CD8+ T cells. In various embodiments, the CD8+ T cells are CD69+ as well. “Therapeutic compositions” are compositions that are designed and administered to patients for the use of treatment of a disease, such as cancer. Therapeutic compositions, e.g., therapeutic IRC-containing compositions, are used to treat benign or malignant tumors or patients/subjects at risk for such tumors, as well as non-solid cancers. In some embodiments, the IRCs are administered to a subject who previously had a tumor and is currently apparently tumor/cancer free, in an effort to enhance the inhibition or the recurrence of the tumor/cancer. “Capsid backbone” refers to a multi-protein structure comprised of viral structural proteins, such as envelop or capsid proteins, such as an L1 protein, that in some instances self-assemble into a capsomere that resembles a virus but lack viral genetic material. Capsid backbones are non-infectious and non-replicating, yet morphologically similar to viruses. The capsid backbones disclosed herein bind to, or possess an inherent tropism for, tumor cells. Capsid Structure Viruses exist in many different morphologies and are generally smaller in size than bacteria, with a diameter between 20 nm and 300 nm, although some filoviruses possess filament lengths of up to 1400 nm. Visualization of viruses or virus capsid backbones requires transmission electron microscopes (TEM) that are more powerful than optical microscopes. Viruses are particle in shape and exist as virions having a nucleic acid surrounded by a protective coat of proteins called the capsid. These capsids are also in turn in some instances surrounded by a protective lipid bilayer that may include surface proteins, receptors, and the like. Capsids are formed from a plurality of identical capsomeres. Capsids generally fall into helical or icosahedral structures, with the exception of bacteriophages that possess more complex structures. The most common icosahedral shape is composed of 20 equilateral triangular faces and resembles a three-dimensional sphere in overall shape. Helical capsids resemble a common spring shape in the form of a three-dimensional cylinder. Each face of the capsid is comprised of anywhere from one to three different proteins or monomer units (protomers). Capsids, when not surrounding papillomavirus genomes, are commonly referred to in the art as virus-like particles, or herein referred to as capsid backbones. That is, an empty capsid with no viral genomic material is referred to herein at times as a capsid backbone. Capsid backbones are excellent delivery molecules for treatment and/or prevention of various diseases, especially in the human body, because they are non-infectious and are optionally re-engineered to specifically target or bind to tumor cells, although most capsid backbone, as described above, possess an inherent tissue tropism without further engineering. Capsomeres are formed from individual subunits or protomers. Native L1 protomers self-assemble through intermolecular disulfide bonds to form pentamers (capsomeres). As noted above, the capsid is comprised of many capsomeres. As used herein, the term “capsomere” is intended to mean a pentameric assembly of papillomavirus L1 polypeptides, including full-length L1 protein, or fragments and mutants thereof. A standard icosahedral capsid is comprised of twenty faces and is a polyhedron including twelve vertices. The vertices are comprised of pentagonal capsomeres and the faces of the capsid are comprised of hexagonal capsomeres. There are always twelve pentagons (pentons) and a varying number of hexagons (hexons) in any given capsid depending on the virus type. Capsids that do not have an exogenous peptide attached thereto are termed “capsid backbones” herein. The icosahedral structure found in most viruses is very common and consists of twenty triangular faces and twelve fivefold vertexes as noted above. The number of capsomeres included in a capsid follows well-known mathematical principles, such as found in the Goldberg polyhedron first described by Michael Goldberg in 1937. The structures can be indexed by two integers h and k, with h being greater than or equal to one and k being greater than or equal to zero, the structure is visualized by taking h steps from the edge of a pentamer, turning 60 degrees counter-clockwise, then taking k steps to get to the next pentamer. The triangulation number “T” for this type of capsid is therefore defined as T=h2+h·k+k2. In this scheme, icosahedral capsids contain twelve pentamers plus 10(T−1) hexamers. (See, Carrillo-Tripp, et al.,Nuc. Acids Res.,37(Database issue):D436-D442, 2009). Thus, it can be seen that the “T” number, or triangulation number, is representative of the size and complexity of a given capsid. However, there are many known exceptions to this general “rule of thumb” found in, for instance, the Papillomaviridae family of viruses that can at times possess pentamers instead of hexamers in hexavalent positions, for instance in a quasi T=7 lattice. Outside of the canonical T=7 capsid structure, other structures such as T=1, T=2, and T=3, are known. A T=1 triangulation value indicates that the capsid is either only an icosahedron or a dodecahedron. Some viruses are enveloped and further comprise a lipid membrane coating surrounding the capsid structure. The envelope is acquired from the host intracellular membrane. The nucleic acid material is either DNA or RNA and can be either single stranded or double stranded. The Papillomaviridae family of viruses is a non-enveloped double-stranded DNA virus. There are several hundred family members within the Papillomaviridae family, each of which is referred to as a “type” that infect most known mammals and other vertebrates such as birds, snakes, turtles, and fish. The Papillomaviridae family members are considered to be relatively highly host- and tissue-tropic, meaning that its members usually possess a specific tissue tropism (preference for infection target) and a preference for host type, and are rarely transmitted between species. For example, it is known that the Papillomaviridae family member human papillomavirus (HPV) type 1 exhibits tropism for the soles of the feet, whereas HPV type 2 prefers tissues in the palms of the hands. Papillomaviruses replicate exclusively in keratinocytes. There are over 170 known human papillomavirus types that have been sequenced and are divided into five genera, including: Alphapapillomavirus, Betapapillomavirus, Gammapapillomavirus, Mupapillomavirus, and Nupapillomavirus. Many more human papillomaviruses have been identified but not yet sequenced. The papillomavirus has but a single protomer called L1 protein, or major capsid protein L1, that is both necessary and sufficient to form its capsid which is comprised of 72 star-shaped capsomers. The papillomavirus family member capsids are non-enveloped and icosahedral. The papillomavirus genome also includes a second structural protein called L2 that is less abundantly expressed than L1. The presence of L2 in the capsid is optional and not necessary for virus function or for formation of the capsid. All of the capsomeres of the Papillomaviridae family are made of pentamer interactions between proteins. As described herein, when describing mutant L1 proteins and the like, such mutants, and capsomers, and capsids made therefrom, are meant to include all Papillomaviridae family members and not just human or mouse family members. Thus, mutant L1 proteins as described herein are meant to encompass all L1 proteins in general, and in some instances specifically Papillomaviridae family L1 proteins in particular. The amino acid domains and sequences of the human papillomavirus L1 protein and its mouse counterpart are presented inFIGS.1and2. A fairly high level of sequence conservation is generally observed across all such L1 proteins of the Papillomaviridae family and is also reflected in this alignment. Further shown inFIG.2are sites of possible mutation of the L1 sequence. Some of these mutations are known historically, such as the deletion of ten amino acids from the amino- or N-terminus of the L1 protein. (See, for instance, Conway et al.,J. Dent. Res.,88(4):307-317, 2009). Other structural mutations of the peptide sequence of the L1 protein in Papillomaviridae family members are known, such as the removal of the carboxy- or C-terminal residues in a truncation mutation. The study of an N-terminal truncation mutant of L1 was begun partly in order to obtain stable crystal structures of the protein for high resolution structural analysis of the capsid. Thus, it was found that full length HPV16 L1 were unable to be crystallized under most tested conditions, but upon removal of the ten N-terminal residues, a crystal was able to be formed for further studies. (Conway et al., 2009). Surprisingly, it was found that upon removal of these ten N-terminal residues, the capsomers formed a T=1 capsid structure comprising icosahedral lattices made from twelve L1 pentamers (for a total of 60 protomers). As noted above, the natural structure of the Papillomaviridae family member capsid is that of 72 L1 pentamers to form a T=7 structure. The T=1 structure of the N-terminal truncation mutant of HPV16 lacks certain disulfide bonds normally formed during capsid formation in wild type HPV16 capsids. Studies have shown that serine to cysteine mutation of C428 or deletion of the helix 4 region on human papillomavirus L1 capsid protein results in disrupting both the T=1 or the T=7 capsid backbone formation. (See, Varsani et al.,Virus Res.,122(1-2):154-163, 2006, and Schadlich et al., ibid.). The overall structure of the papillomavirus L1 protein is presented inFIG.1AandFIG.1Band has a tertiary structure consisting generally of various secondary structures including a core of β-strands that form a classic “jelly roll” β-sandwich and five C-terminal α-helices that support five surface-exposed loop regions generally designated in the art as loops BC, DE, EF, FG, and HI. (See, Chen et al.,Mol. Cell,5:557-567, 2000, and Bissett et al.,Scientific Reports,6:39730, 2016). Three of the α-helices, commonly referred to as h2, h3, and h4, form the surface of contact with other monomers and pentamers (FIG.1B). (See, Chen et al.,FIG.4, page 561). The five α-helices generally reside at the carboxy-terminus of the L1 sequence. Design and Production of Mutant L1 Proteins Deletions of the MPV L1 sequence were made to facilitating the formation of 10 nm to 15 nm capsomeres made from five L1 proteins. It was previously shown that truncation of the amino-, helix-four, and carboxy-terminus residues of the HPV16 L1 protein results in capsomere formation. (See, Bishop et al.,Virol. J.,4:3, 2007, and Schadlich et al.,J. Virol.,83(15):7690-7705, 2009). On the other hand, it was shown in HPV11 and HPV16 that truncation of the amino-terminal ten residues of L1, alone, would yield T=1 icosahedral capsid backbones. These T=1 icosahedral capsid backbones are approximately 20 nm to 30 nm in diameter and consist of 60 L1 proteins (or 12 capsomers). Deletion of up to 34 amino acids at the carboxy-terminus did not inhibit T=1 formation. However, if deletions in the helix-four region of L1 occurred (amino acids 411 to 436), the formation of T=1 would be ablated, even in the presence of N-terminal or C-terminal truncations. In all permutations, capsomers would be observed. (Chen et al.,Mol. Cell,5(3):557-567, 2000, and WO 2000054730). These results were consistent with papillomavirus type 16 L1 produced inE. colior in insect cells. (See, Schadlich et al.,J. Virol.,83(15):7690-7705, 2009). Taken together, the authors of this study concluded that the helix-4 structure was needed for the assembly of capsomers into both higher ordered T=1 and T=7 icosahedral structures. (See, Bishop et al.,Virol.,4:3, 2007). Various deletions of the MPV L1 sequence were generated, resulting in the construct called “MPV.10.34.d” shown schematically inFIG.1B. This construct was designed to create an MPV capsomere as a therapeutic platform. MPV L1 proteins were selected as the carrier vehicle construct instead of HPV L1 because humans have for the most part not been exposed to MPV and therefore it was postulated that virion-derived capsids from MPV would not be sensitive to innate immune response as is seen with HPV L1 proteins. Recently a ΔN10 deletion of HPV16 L1, in which the amino-terminal ten residues of the HPV L1 sequence are removed, was crystallized and found to conform to the shape of a T=1 capsid backbone. (See, Chen et al., 2000). The structure revealed that the carboxy terminal segment from residue 384 to 446 of L1 folds into three helices with connecting loops and turns. These helices are the primary inter-pentamer bonding contacts in the assembled T=1 capsid backbone. To test whether these helices also affect capsid backbone assembly, L1 proteins comprising the ΔN10 deletion were generated with a specific deletion of helix 4 for both HPV16 (residues 408 to 431) and HPV11 (residues 409 to 429). The pentamers were purified by FPLC and were shown to possess a “donut” shape as observed by electron microscopy (EM). No assembly of capsid backbones from these pentamers was found under any condition tested, suggesting that this carboxy-terminal helical domain is essential for T=1 or T=7 capsid backbone assembly. Crystallographic analysis of the T=1 capsid backbone revealed that inter-pentameric contacts are established by hydrophobic interactions between the α-helices 2 and 3 of one capsomere and α-helix 4 of a neighboring capsomere. (Chen et al., 2000). Consequently, a mutant L1 with helix 4 deleted formed homogenous capsomeres but failed to form T=1 and T=7 capsid backbones. (See, Bishop et al., 2006). The constructs with helix 4 deleted did not exhibit any ability to self-assemble, consistent with previous reports. (See, Schadlich et al.,J. Virol.,83(15):7690-7705, 2009). For the purposes of this description, the term mutant L1 protein means an L1 protein or protomer comprising one or more non-wild type sequences. Such non-wild type sequences include truncations or deletions (internal or at the ends of the sequences), single residue substitutions, and the like. For instance, a mutant L1 protein includes an L1 protein in which any of the following are true: 1) a certain number of the N-terminal residues are deleted, a certain number of the C-terminal residues are deleted, and/or 3) a certain number of internal residues are deleted, in some instances in more than one location internally within the sequence. The mutant L1 protein is in some embodiments derived from a wild type papillomavirus L1 protein. Any papillomavirus L1 protein is useful in the presently described compositions. L1 protein sequences are relatively conserved. Thus, description of mouse papillomavirus mutant L1 proteins, below, are exemplary and it is contemplated that the same mutations made in other L1 proteins of the papillomavirus family is expected to yield similar results. In various embodiments, a capsid backbone is provided comprising a papillomavirus L1 protein and/or a papillomavirus L2 protein. Thus, the capsid backbone in some embodiments comprises both papilloma L1 and L2 proteins. In other embodiments, the capsid backbone is comprised of only L1 proteins. In some embodiments the L1 protein is a hybrid or chimeric protein comprised of L1 sequences from more than one source merged together into a single L1 sequence. The L1 protein sequences are known for substantially all papillomavirus genotypes identified to date, and any of these L1 sequences or fragments are contemplated as being included in the present compositions. Examples of L1 polypeptides include, without limitation, full-length L1 polypeptides, e.g., HPV16 L1 polypeptide, SEQ ID NO: 128, L1 truncations that lack any one or more residues of the native C-terminus, L1 truncations that lack any one or more residues of the native N-terminus, and L1 truncations that lack any one or more internal domain residues in any one or more internal locations. The L1 protein is in some instances exemplified as a modified L1 protein, e.g., a modified HPV16 or MPV16 L1 protein, wherein the HPV16 L2 amino acids 17 to 36 (the RG1 epitope) are inserted within the DE-surface loop of HPV16 L1. (See, Schellenbacher et al.,J. Invest. Dermatol.,133(12):2706-2713, 2013; Slupetzky et al.,Vaccine,25:2001-2010, 2007; Kondo et al.,J. Med. Virol.,80:841-6, 2008; Schellenbacher et al.,J. Virol.,83:10085-10095, 2009; and Caldeira et al.,Vaccine,28:4384-93, 2010). The L2 polypeptide is in some embodiments full-length L2 protein or an L2 polypeptide fragment. The L2 sequences are known for substantially all papillomavirus genotypes identified to date, and any of these L2 sequences or fragments can be employed in the present disclosure. Examples of L2 polypeptides include, without limitation, full-length L2 polypeptides, e.g., HPV16 L2 polypeptide (SEQ ID NO: 1), or mouse papillomavirus L2 (SEQ ID NO: 2), L2 truncations that lack any one or more of the native C-terminus, L2 truncations that lack any one or more of the native N-terminus, and L2 truncations that lack any one or more internal domain residues in any one or more locations. The papillomavirus capsid backbone is in some embodiments formed using the L1 and optionally L2 polypeptides from any animal papillomavirus, or derivatives or fragments thereof. Thus, any known (or hereafter identified) L1 and optionally L2 sequences of human, bovine, equine, ovine, porcine, deer, canine, feline, rodent, rabbit, etc., papillomaviruses are employed to prepare the capsid backbones described herein. (See, de Villiers et al.,Virology,324:17-27, 2004, for a current description of papillomavirus genotypes and their relatedness, incorporated herein by reference for all purposes). In certain embodiments, the L1 and optionally L2 polypeptides that are used to form the capsid backbones are from a non-human papillomavirus or a human papillomavirus genotype other than HPV6, HPV11, HPV16, and HPV18. For example, the L1 and/or L2 proteins are in some embodiments from HPV 1, 2, 3, 4, 5, 6, 8, 9, 15, 17, 23, 27, 31, 33, 35, 38, 39, 45, 51, 52, 58, 66, 68, 70, 76, or 92. As described above, in human papillomavirus HPV16, several different mutations of L1 protein have been characterized. (See, for instance, Chen et al., 2000). Some of these mutations include the following in Table 1. (Chen et al., 2000, Table 1, page 558): TrypsinApparent Diameter ofDeletionSensitivityAssembled Particle (Å)aΔN = 0No600ΔN = 8No600ΔN = 9No600ΔN = 10No300ΔN = 15YesbNAΔN = 20YesNAΔC = 16No600ΔC = 30No600ΔC = 46YesNAΔC = 86YesNA In Table 1, the delta symbol (A) designates deletion and the “N” or “C” designated whether the deletion is located at the N-terminus or C-terminus, respectively. The number following these two symbols indicates the number of residues of the L1 sequence that were deleted. It is noted that Chen et al. does not report any double, triple, or higher number of mutations within a single L1 protein. Thus, the L1 mutant proteins described herein include N-terminal truncation L1 mutant proteins. The N terminus is truncated by at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids. In some embodiments the N-terminal truncation is 5 amino acids. In some embodiments the N-truncation is 10 amino acids. In some embodiments the N-terminal truncation is 37, 38, 39, or even 40 amino acids. The L1 mutant proteins described herein further include C-terminal truncation L1 mutant proteins. The C terminus is truncated by at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids. In some embodiments the C-terminal truncation is 5 amino acids. In some embodiments the C-truncation is 10 amino acids. The L1 mutant proteins described herein further include L1 mutants in which any number of internal residues are deleted. Surprisingly, the retention of the helix-4 region is in some embodiments needed for the formation of capsid backbones having a T=1 geometry, whereas in the literature it is reported, as discussed above, that its deletion is not supposed to yield any capsid backbone assembly. Generally, the internal residues deleted in the described mutant L1 proteins are those shown inFIGS.1and2. Contemplated also are deletions of 34 residues in the helix 4 (H4) region. In some instances, the truncation of internal residues of L1 proteins is 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or even 40 residues in length. Also described herein are L1 mutant proteins in which any one or more of the C-terminus and/or N-terminus and/or internal residues are deleted simultaneously. For instance, in some embodiments the mutant L1 protein has both C- and N-terminal truncation mutations of similar or varying length. In other embodiments, the mutant L1 protein has a C-terminal truncation and an internal residue truncation. In some embodiments, the mutant L1 protein has an N-terminal truncation and an internal truncation. In certain embodiments, the mutant L1 protein has truncations simultaneously in all three locations, C-terminus, N-terminals, and internal truncations. The mutant L1 proteins described herein are generally produced recombinantly but are also produced by any known protein expression methodology. For instance, the mutant L1 proteins are generated by first designing DNA primers complimentary to the wild type L1 sequence and then performing PCR amplification of the sequence in the presence of the primers designed to truncate or otherwise mutate the wild type L1 sequence, as further explained in detail below, in the Examples section. (See also, Touze et al.,J. Clin. Microbiol.,36(7):2046-2051, 1998). The design and implementation of proper primer sequences and PCR protocols are known and such methods are used to ultimately generate the desired mutant L1 protein nucleic acid sequence, from which the mutant L1 proteins are expressed. The mutant L1 protein nucleic acid sequence is then in some embodiments codon-optimized for better protein expression and production depending on the organism in which the expression is conducted. Utilization of different codon optimization methods for certain expression vectors and host expression systems are known in the art. (See, Mauro and Chappell,Trends in Molecular Medicine,20(11):604-613, 2014, for instance). The mutant L1 protein nucleic acid sequence is then ligated into an acceptably prepared and commercially-available expression vector designed for protein expression. Expression vectors of various types possessing functionality for certain expression hosts are widely commercially available. Recombinant mutant L1 proteins described herein are expressed in bacterial as well as eukaryotic cells and in certain embodiments are expressible in vitro. Often expression of recombinant proteins in bacterial hosts results in the formation of inclusion bodies (IBs). Thus, in some instances, recombinant mutant L1 protein expressed as IBs are solubilized using known procedures. In a particular embodiment, the solubilization of IBs of expressed mutant L1 proteins described herein includes the steps set forth, for instance, inFIG.3, and Example 3. The processes include various steps, such as: (a) transforming a prokaryotic cell with an expression vector encoding the L1 protein; (b) culturing the transformed prokaryotic cell under conditions that promote expression of the L1 protein; (c) lysing the transformed prokaryotic cells to release expressed L1 protein; (d) separating cell debris from the expressed L1 protein and recovering the L1 protein in the form of IBs; (e) optionally washing the L1 protein IBs; (f) solubilizing the L1 protein IBs; (g) refolding the L1 protein optionally in the presence of one or more denaturants, reducing agents, and the like; and (h) forming the icosahedron or dodecahedron capsid having a triangulation number T equal to 1 by incubating the refolded L1 protein in assembly buffer. Such processes, in some embodiments, further include conjugating in a conjugation buffer the one or more peptides to the assembled L1 protein by incubating the assembled L1 protein under reducing conditions in the presence of one or more peptides and/or removing denaturant from the conjugation buffer but maintaining reducing agent when forming the icosahedron or dodecahedron capsid having a triangulation number T equal to 1. The described methods and processes for creating and purifying the described mutant L1 proteins is different in many aspects from such processes described in the art for assembly of papillomavirus capsids. Indeed, it is known in the art that assembly into higher ordered papillomavirus capsids requires that the L1 protein must first be subjected to a disassembly buffer that includes a reducing agent. This step is then often followed by subjecting the L1 protein to an assembly buffer that then removes the reducing agent. This legacy methodology results in stable capsids with improved properties. (See, McCarthy et al., 10.1128/JVI.72.1.32-41, 1998, Zhao et al.,Virol. J.,9:52, 2012, Mach et al.,J. Pharm. Sci.,95:2195-2206, 2006, and U.S. Pat. No. 6,436,402). Remarkable Properties of Capsid Backbones Formed From Mutant L1 Proteins It was serendipitously discovered during the studies described herein that certain mutant L1 proteins possess beneficial and unexpected properties. For instance, certain mutant L1 proteins led primarily to the formation of a T=1 capsid backbone possessing helpful and unexpected conjugation properties. The formation of a T=1 capsid backbone instead of a T=7 capsid backbone leads to higher stability under reducing conditions and therefore higher conjugation efficiency as compared with wild type sequences that form T=7 capsid backbone. For instance, the efficiency with which the mutant capsid backbone, e.g., the MPV.10.34.d backbone, is able to be conjugated with peptide is from 25 to 85% (w/w). In some embodiments, the conjugation efficiency is about 25%. In other embodiments, the conjugation efficiency is about 25, about 35, about 45, about 55, about 65, about 75, or even about 85% (w/w). In contrast, wild type T=7 capsid backbones have generally a lower efficiency of conjugation that is less than about 25%. See, for instance, WO 2020/139978. The ability to achieve a higher amount of peptide conjugated to the T=1 capsid backbone compared to T=7 capsid backbone allows for delivery of a higher number of peptides to the target tumor or cancer at an overall lower IRC dose amount compared with IRC forms from T=7 capsid backbones. Additionally, T=1 capsid backbones having a smaller geometric shape or size as compared to T=7 capsid backbones allows for less stearic hindrance with the IRC made from T=1 capsid backbones is injected into a subject and the IRC infiltrate tumor microenvironments. This beneficial and unexpected effect then leads to a lower IRC dose needed to achieve the same effect as an equivalent T=7, or higher order, capsid backbone-based IRC. These and other additional beneficial features of the T=1 capsid backbone geometry are described in further detail hereinbelow. Mutant L1 Protein IRC and Mechanism of Action In some embodiments, the mutant L1 protein is conjugated to another peptide. To add further beneficial functionality to capsids or capsid backbones comprised of the mutant L1 proteins, additional peptides are conjugated to the surface of such capsids. These peptides add beneficial functionality to the capsid and result in added functionality such as treatment of cancer in subjects in need thereof. In an embodiment, the conjugated papillomavirus capsid backbone comprises an L1 capsid protein and a peptide. In other embodiments, the IRC comprises an (at least one) L1 capsid protein, an (at least one) L2 capsid protein, and at least one peptide. The L1 polypeptide is in some embodiments a full length L1 protein or in other embodiments is an L1 polypeptide fragment. In specific embodiments, the full-length L1 protein or L1 polypeptide fragment is capsid backbone assembly-competent; that is, the L1 polypeptide will self-assemble to form capsomeres under proper conditions that are competent for self-assembly into higher-order structural geometries, thereby forming a capsid backbone. In more specific embodiments, the capsid backbones comprise a T=1 particle, a structure of about 20 nm to 30 nm in diameter, and composed of 12 capsomeres or 60 copies of L1 protein. In other embodiments, the capsid backbones comprise a fully assembled papillomavirus capsid, a structure of about 50 nm and composed of 72 capsomeres or 360 copies of L1 protein. In various embodiments, the IRC presented herein bind to, specifically or non-specifically, or otherwise contact, one or more cancer cells. This is in part due to the capsid backbone's selectivity (tropism) for proteins and/or molecules that are in some instances specific to, or expressed in higher abundance by, tumor cells. In various embodiments, the IRC binds to a certain sub-family type of heparin sulfate proteoglycan (HSPG), which is preferentially expressed on tumor cells. As used herein, “binding to a cancer cell” refers to the formation of non-covalent interactions between the capsid protein of the IRC and the tumor cell such that the IRC comes into close proximity to the tumor cell and the peptide is cleaved from the capsid backbone, and then the peptide binds to, or is bound by, or otherwise interacts with, the MHC receptor present on the tumor cell surface. In various embodiments, the peptide is an epitope that is recognized by a T cell or T cell population that already exists in the subject. In various embodiments, this existing T cell or T cell population exists because of a prior infection or vaccination. In various embodiments, the peptide is an epitope that is capable of being bound by a T cell. In various embodiments, the peptide is an epitope capable of being bound by a T cell already present in a subject. In this context, “capable of being bound” means that an “epitope” is presented on the surface of a cell, where it is bound to MHC molecules. T cell epitopes presentable by MHC class I receptors are bound by the T cell receptor of cytotoxic CD8 T lymphocytes (CD8 T cells or CTLs). T cell epitopes presentable by MHC class I molecules are typically peptides of about 9 to about 12 amino acids in length. In various embodiments, an IRC is provided that releases a T cell response-eliciting peptide that upon release is directly bound by and consequently appropriately presentable by one or more MHC molecules expressed on the surface of one or more cancer or tumor cells. As the released peptide does not require processing by the antigen processing machinery in the cytosol, the T cell response-eliciting peptides are presented on the surface of the target cell in a short amount of time. The process of release of such peptides from the IRC and subsequent binding of the peptides by the MHC molecules of target cells is akin to labelling, tagging, or otherwise “marking” these tumor or cancer cells. This tagging or marking leads to ready identification by other components of the subject's immune system, thereby recruiting these components of the subject's immune system to remove the cancer or tumor cells via the various known cell destruction pathways. Hence, in one embodiment of the described methods, uses, and compositions described herein, in less than about 8.5 hours after administration of the IRC dose to the subject, the IRC will naturally migrate to the target cell after which the T cell response-eliciting peptide released from the IRC, is bound by the MHC molecule on the cancer cell, and then the peptide is presented on the surface of the target cell via an MHC class I molecule to other components of the subject's immune system for recognition thereby. In another embodiment of the invention, in less than 23.5 hours after introduction of the IRC to the target cell the T cell response eliciting peptide is presented on the surface of the target cell via an MHC class I molecule. In another embodiment of the invention, the IRC is capable of mediating T cell cytotoxicity against the target cell within less than 6 hours after administration of the IRC to the target cell. In various embodiments, the peptide comprises one epitope or comprises at least two epitopes. The peptide epitopes are in some instances derived from different proteins, or in other embodiments they are epitopes from the same protein (or antigen). In various embodiments, the pathogen is a virus, a bacterium, a fungus, a parasite, or a combination thereof. In various embodiments, the subject's preexisting T cells are specific to a vaccine epitope. In various embodiments the epitope is derived from a childhood, early childhood, adolescent, or elderly (geriatric), vaccine. In various embodiments the subject's preexisting immunity is the result of prior administration of a human vaccine. Antigens described herein that comprise epitopes incorporated into the peptides described herein are found in any of the known infectious agents, such as viruses, bacteria, parasites, fungi, and the like. In various embodiments, the peptide is selected from the list provided by Table 2. For instance, non-limiting examples of a viruses from which antigens bearing epitopes that are incorporated in some embodiments into the described peptides include, for instance, a vaccinia virus, a varicella zoster virus, a herpesvirus, e.g., herpes zoster virus or cytomegalovirus or Epstein-Barr virus, rubella, a hepatitis virus, e.g., hepatitis A virus or hepatitis B virus or hepatitis C virus, influenza, e.g., type A or type B, a measles virus, a mumps virus, a polio virus, a variola (smallpox) virus, a rabies virus, a coronavirus, Dengue virus, an Ebola virus, a West Nile virus, a yellow fever virus, or a zika virus. For instance, non-limiting examples of a bacteria from which antigens bearing epitopes that are incorporated in some embodiments into the described peptides include, for example, aBordetella pertussis, chlamydia, trachomatis, Clostridium tetani, diphtheria, Hemophilus influenza, Meningococcus, pneumococcus, Vibrio cholera, Mycobacterium tuberculosis, BCG, typhoid,E. coli, salmonella, Legionella pneumophila, rickettsia, Treponema pallidum pallidum, Streptococcusgroup A or group B,Streptococcus pneumonia, Bacillus anthracis, Clostridium botulinum, or aYersiniasp bacteria. For instance, non-limiting examples of a parasite from which antigens bearing epitopes that are incorporated in some embodiments into the described peptides include,Entamoeba histolytica, Toxoplasma gondii, aTrichinellasp., e.g.,Trichinella spiralis, aTrichomonassp., e.g.,Trichomonas vaginalis, aTrypanosomasp., e.g.,Trypanosoma brucei gambiense, Trypanosoma brucei rhodesiense, or aTrypanosoma cruzi, or aplasmodium, e.g.,Plasmodium falciparum, Plasmodium vivax, orPlasmodium malariae. TABLE 2Epitope Peptide SequencesSEQIDEpitope SequenceNOVirus TypeMHC alleleViral ProteinSLPRSRTPI4Chicken PoxA*02:01IE62(VZV)SAPLPSNRV5Chicken PoxA*02:01IE62(VZV)GSAPLPSNRV6Chicken PoxA*02:01IE62(VZV)ALWALPHAA7Chicken PoxA*02:01IE62(VZV)SLSGLYVFV8ShinglesA*02:01Glycoprotein EvaccinesYLGVYIWNM9ShinglesA*02:01Glycoprotein EvaccinesKIHEAPFDL10ShinglesA*02:01Glycoprotein EvaccinesLLCLVIFLI11ShinglesA*02:01Glycoprotein EvaccinesDLLLEWLYV12ShinglesA*02:01Glycoprotein EvaccinesSMYYAGLPV13ShinglesA*02:01Glycoprotein EvaccinesILHDGGTTL14ShinglesA*02:01Glycoprotein EvaccinesWLYVPIDPT15ShinglesA*02:01Glycoprotein EvaccinesVLMGFGIIT16ShinglesA*02:01Glycoprotein EvaccinesCLVIFLICT17ShinglesA*02:01Glycoprotein EvaccinesKEADQPWIV18ShinglesA*02:01Glycoprotein EvaccinesVVSTVDHFV19ShinglesA*02:01Glycoprotein EvaccinesFLICTAKRM20ShinglesA*02:01Glycoprotein EvaccinesVLRTEKQYL21ShinglesA*02:01Glycoprotein EvaccinesHMWNYHSHV22ShinglesA*02:01Glycoprotein EvaccinesTVNKPVVGV23ShinglesA*02:01Glycoprotein EvaccinesFVVYFNGHV24ShinglesA*02:01Glycoprotein EvaccinesWIVVNTSTL25ShinglesA*02:01Glycoprotein EvaccinesVAYTVVSTV26ShinglesA*02:01Glycoprotein EvaccinesFMYMSLLGV27measlesA*02:01m50SLWGSLLML28measlesA*02:01C proteinLLAVIFVMFL29measlesA*02:01H38SMYRVFEVGV30measlesA*02:01H250-259ILPGQDLQYV31measlesA*02:01H516-525KLWCRHFCV32measlesA*02:01H576KLWCRHFCVL33measlesA*02:01H576RLSDNGYYTV34measlesA*02:01M164KLLRYYTEI35measlesA*02:01F205KLWESPQEI36measlesA*02:01C 84RLLDRLVRL37measlesA*02:01N50KLMPNITLL38measlesA*02:01F57TLLNNCTRV39measlesA*02:01F64EMLTLATWV40Hep BA*02:01C64-72FLPSDFFPSV41Hep BA*02:01Core 18FLPADFFPSV42Hep BA*02:01Core 19FLPSDFFPSI43Hep BA*02:01Core 20WLSLLVPF44Hep BA*02:01ENV335FLLTRILTI or FLLTRILTL45Hep BA*02:01ENV183or46GLSPTVWLSV47Hep BA*02:01ENV348LLDYQGMLPV48Hep BA*02:01ENV260LLCLIFLLV49Hep BA*02:01ENV251SIVSPFIPLL50Hep BA*02:01ENV370FLLTKILTI51Hep BA*02:01ENV183ILSPFLPLL52Hep BA*02:01ENV371FLLSLGIHL53Hep BA*02:01POL 575GLSRYVARL54Hep BA*02:01POL 455SLYADSPSV55Hep BA*02:01POL 816YMDDVVLGA56Hep BA*02:01POL 551ALMPLYACI57Hep BA*02:01POL 655VLHKRTLGL58Hep BA*02:01HBx 92CLFKDWEEL59Hep BA*02:01Hbx115STLPETTVVRR60Hep BA*03, A*11Core 141EYLVSFGVW61Hep BA*31, A*68core 117FFPSIRDLL62Hep BA*24Core 23SWLSLLVPF63Hep BA*24Env 334KYTSFPWLL64Hep BA*24Pol 756HLSLRGLFV65Hep BA*02:01HBx 52-60CLFKDWEEL66Hep BA*02:01HBx 115-123LPSDFFPSV67Hep BB*51Core 19GILGFVFTL68InfluenzaHLA-A2M1ILGFVFTLTVPSERGLQRRRF69InfluenzaLIRHENRMVLASTTAKA70InfluenzaLQAYQKRMGVQMQR71InfluenzaYVYDHSGEAVK72MeaslesWLSLLVPFV73Hep B(K)GILGFVFTL(T)(V)74InfluenzaKLSTRGVQIASNEN75InfluenzaRGLQRRRFVQNALNGNG76InfluenzaFMYSDFHFI77InfluenzaNLVPMVATV3CytomegalovirusHLA-A2VAIIEVDNEQPTTRAQKL78PoliovirusAny 9-mer sequence79Poliovirusof GACVAIIEVDNEQPTTRAQKLFAMWRITYKDTVQLRRKLSVRDRLARL80EBVLLDRVRFMGV81EBVCLGGLLTMV82EBVGLCTLVAML83EBVSVLGPISGHVLK84CytomegalovirusHLA-A11RPHERNGFTVL85CytomegalovirusHLA-B7FTSQYRIQGKL86CytomegalovirusHLA-A24YSEHPTFTSQY87CytomegalovirusHLA-A1EFFWDANDIY88CytomegalovirusHLA-B44TTVYPPSSTAK90CytomegalovirusHLA-A3FVFPTKDVALR91CytomegalovirusHLA-A68QTVTSTPVQGR92CytomegalovirusHLA-A68PTFTSQYRIQGKL93CytomegalovirusHLA-B38FPTKDVAL94CytomegalovirusHLA-B35SIINFEKL95RAHYNIVTF96SSPPMFRV97KLWAQCVQL98SARS-CoV-2A*02:01KLPDDFTGCV99SARS-CoV-2A*02:01YLQPRTFLL100SARS-CoV-2A*02:01LLYDANYFL101SARS-CoV-2A*02:01ALWEIQQVV102SARS-CoV-2A*02:01LLLDRLNQL103SARS-CoV-2A*02:01YLFDESGEFKL104SARS-CoV-2A*02:01FTSDYYQLY105SARS-CoV-2A*01:01PTDNYITTY106SARS-CoV-2A*01:01ATSRTLSYY107SARS-CoV-2A*01:01CTDDNALAYY108SARS-CoV-2A*01:01NTCDGTTFTY109SARS-CoV-2A*01:01DTDFVNEFY110SARS-CoV-2A*01:01GTDLEGNFY111SARS-CoV-2A*01:01KTFPPTEPK112SARS-CoV-2A*03:01KCYGVSPTK113SARS-CoV-2A*03:01VTNNTFTLK114SARS-CoV-2A*03:01KTIQPRVEK115SARS-CoV-2A*03:01KTFPPTEPK116SARS-CoV-2A*11:01VTDTPKGPK117SARS-CoV-2A*11:01ATEGALNTPK118SARS-CoV-2A*11:01SARS-CoV-2ASAFFGMSR119SARS-CoV-2A*11:01ATSRTLSYYK120SARS-CoV-2A*11:01QYIKWPWYI121SARS-CoV-2A*24:02VYFLQSINF122SARS-CoV-2A*24:02VYIGDPAQL123SARS-CoV-2A*24:02SPRWYFYYL124SARS-CoV-2B*07:02RPDTRYVL125SARS-CoV-2B*07:02IPRRNVATL126SARS-CoV-2B*07:02 In various embodiments the epitope is found in one or more known human vaccines, such as a childhood vaccine, early childhood, adolescent, or elderly (geriatric), vaccine. In various embodiments the vaccine is an early childhood vaccine. Certain non-limiting examples of suitable vaccines from which such epitopes are found that are compatible with the described peptides are listed in Table 3. TABLE 3Human Vaccines Containing Peptide-Compatible EpitopesCommercialResponsible NationalTypeNameFormSourceRegulatory AuthorityDiphtheria-Tetanus-QuinvaxemLiquid:JanssenMinistry of FoodPertussisready to useVaccinesand Drug Safety(whole cell)-Corp.Hepatitis B-Hemophilusinfluenzae type bDiphtheria-TetanusAdsorbedLiquid:PT BioNational Agency ofDT Vaccineready to useFarmaDrug and Food(Persero)Control IndonesiaDiphtheria-Tetanus-DTPLiquid:PT BioNational Agency ofPertussisVaccineready to useFarmaDrug and Food(whole cell)(Persero)Control IndonesiaHepatitis BHepatitis BLiquid:PT BioNational Agency ofVaccineready to useFarmaDrug and FoodRecombinant(Persero)Control IndonesiaPolio Vaccine - OralOral polioLiquid:PT BioNational Agency of(OPV) Trivalentready to useFarmaDrug and Food(Persero)Control IndonesiaPolio Vaccine - OralOral polioLiquid:PT BioNational Agency of(OPV) Trivalentready to useFarmaDrug and Food(Persero)Control IndonesiaTetanus ToxoidTT vaccineLiquid:PT BioNational Agency ofready to useFarmaDrug and Food(Persero)Control IndonesiaTetanus ToxoidTT vaccineLiquid:PT BioNational Agency ofready to useFarmaDrug and Food(Persero)Control IndonesiaTetanus ToxoidTT vaccineLiquid:PT BioNational Agency ofready to useFarmaDrug and Food(Persero)Control IndonesiaMeaslesMeaslesLyophilizedPT BioNational Agency ofvaccineLyophilizedFarmaDrug and Foodactive(Persero)Control Indonesiacomponentto bereconstitutedwithexcipientdiluentbefore useYellow FeverYellow FeverLyophilizedBio-Agencia NacionalLyophilizedManguinhos/da VigilanciaactiveFiocruzSanitariacomponentto bereconstitutedwithexcipientdiluentbefore useYellow FeverYellow FeverLyophilizedBio-Agencia NacionalLyophilizedManguinhos/da VigilanciaactiveFiocruzSanitariacomponentto bereconstitutedwithexcipientdiluentbefore useYellow FeverYellow FeverLyophilizedBio-Agencia NacionalLyophilizedManguinhos/da VigilanciaactiveFiocruzSanitariacomponentto bereconstitutedwithexcipientdiluentbefore useHepatitis BHeberbiovacLiquid:Centro deCentro para elHBready to useIngenieriaControl Estatal deGenetica yla Calidad de losBiotecnologiaMedicamentosHepatitis BHeberbiovacLiquid:Centro deCentro para elHBready to useIngenieriaControl Estatal deGenetica yla Calidad de losBiotecnologiaMedicamentosRabiesRabipurLyophilizedChironCentral DrugsactiveBehringStandard ControlcomponentVaccinesOrganizationto bePrivate Ltd.reconstitutedwithexcipientdiluentbefore useRabiesRabipurLyophilizedGlaxoSmithKlinePaul-Ehrlich-activeVaccinesInstitutcomponentGmbHto bereconstitutedwithexcipientdiluentbefore useHaemophilusVaxem HIBLiquid:NovartisAgenzia Italianainfluenzae type bready to useVaccinesdel FarmacoandDiagnosticsS.r.lHepatitis BEngerixLiquid:GlaxoSmithKlineFederal Agency forready to useBiologicalsMedicines andSAHealth ProductsHepatitis BEngerixLiquid:GlaxoSmithKlineFederal Agency forready to useBiologicalsMedicines andSAHealth ProductsHepatitis BEngerixLiquid:GlaxoSmithKlineFederal Agency forready to useBiologicalsMedicines andSAHealth ProductsPolio Vaccine - OralPolio sabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) Trivalentready to useBiologicalsMedicines andSAHealth ProductsPolio Vaccine - OralPolio sabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) Trivalentready to useBiologicalsMedicines andSAHealth ProductsMeasles,PriorixLyophilizedGlaxoSmithKlineFederal Agency forMumps andactiveBiologicalsMedicines andRubellacomponentSAHealth Productsto bereconstitutedwithexcipientdiluentbefore useRotavirusRotarixLiquid:GlaxoSmithKlineFederal Agency forready to useBiologicalsMedicines andSAHealth ProductsPolio Vaccine - OralPolioviralLiquid:HaffkineCentral Drugs(OPV) Trivalentvaccineready to useBioStandard ControlPharmaceuticalOrganizationCorporationLtdYellow FeverStabilizedLyophilizedInstitutMinistère de laYellow FeveractivePasteur deSanté publiqueVaccinecomponentDakarto bereconstitutedwithexcipientdiluentbefore useYellow FeverStabilizedLyophilizedInstitutMinistère de laYellow FeveractivePasteur deSanté publiqueVaccinecomponentDakarto bereconstitutedwithexcipientdiluentbefore useYellow FeverStabilizedLyophilizedInstitutMinistère de laYellow FeveractivePasteur deSanté publiqueVaccinecomponentDakarto bereconstitutedwithexcipientdiluentbefore useBCGBCG FreezeLyophilizedJapan BCGChiba LocalDriedactiveLaboratoryGovernmentGlutamatecomponentvaccineto bereconstitutedwithexcipientdiluentbefore useHepatitis BEuvax BLiquid:LG ChemMinistry of Foodready to useLtdand Drug SafetyHepatitis BEuvax BLiquid:LG ChemMinistry of Foodready to useLtdand Drug SafetyBCGBCGLyophilizedBul Bio -Bulgarian DrugVaccineactiveNationalAgencycomponentCenter ofto beInfectiousreconstitutedandwithParasiticexcipientDiseasesdiluentLtd.before useBCGBCGLyophilizedBul Bio -Bulgarian DrugVaccineactiveNationalAgencycomponentCenter ofto beInfectiousreconstitutedandwithParasiticexcipientDiseasesdiluentLtd.before useTetanus ToxoidTetatoxLiquid:Bul Bio -Bulgarian Drugready to useNationalAgencyCenter ofInfectiousandParasiticDiseasesLtd.Tetanus ToxoidTetatoxLiquid:Bul Bio -Bulgarian Drugready to useNationalAgencyCenter ofInfectiousandParasiticDiseasesLtd.Diphtheria-TetanusDiftetLiquid:Bul Bio -Bulgarian Drugready to useNationalAgencyCenter ofInfectiousandParasiticDiseasesLtd.Diphtheria-TetanusDiftetLiquid:Bul Bio -Bulgarian Drugready to useNationalAgencyCenter ofInfectiousandParasiticDiseasesLtd.Diphtheria-TetanusTetadifLiquid:Bul Bio -Bulgarian Drug(reduced antigenready to useNationalAgencycontent)Center ofInfectiousandParasiticDiseasesLtd.Diphtheria-TetanusTetadifLiquid:Bul Bio -Bulgarian Drug(reduced antigenready to useNationalAgencycontent)Center ofInfectiousandParasiticDiseasesLtd.Diphtheria-Tetanus-Easyfive-TTLiquid:PanaceaCentral DrugsPertussisready to useBiotec Ltd.Standard Control(whole cell)-OrganizationHepatitis B-Haemophilusinfluenzae type bDiphtheria-TetanusIMOVAXLiquid:SanofiAgence nationale(reduced antigendT adultready to usePasteur SAde sécurite ducontent)médicament et desproduits de santéPolio Vaccine -IMOVAXLiquid:SanofiAgence nationaleInactivated (IPV)POLIOready to usePasteur SAde sécurité dumédicament et desproduits de santéPolio Vaccine - OralOPVEROLiquid:SanofiAgence nationale(OPV) Trivalentready to usePasteur SAde sécurité dumédicament et desproduits de santéPolio Vaccine - OralOPVEROLiquid:SanofiAgence nationale(OPV) Trivalentready to usePasteur SAde sécurité dumédicament et desproduits de santéPolio Vaccine - OralOPVEROLiquid:SanofiAgence nationale(OPV) Trivalentready to usePasteur SAde sécurité dumédicament et desproduits de santéTetanus ToxoidTETAVAXLiquid:SanofiAgence nationaleready to usePasteur SAde sécurité dumédicament et desproduits de santéTetanus ToxoidTETAVAXLiquid:SanofiAgence nationaleready to usePasteur SAde sécurité dumédicament et desproduits de santéHaemophilusAct-HIBLyophilizedSanofiAgence nationaleinfluenzae type bactivePasteur SAde sécurité ducomponentmédicament et desto beproduits de santéreconstitutedwithexcipientdiluentbefore useRabiesVERORABLyophilizedSanofiAgence nationaleactivePasteur SAde sécurité ducomponentmédicament et desto beproduits de santéreconstitutedwithexcipientdiluentbefore useYellow FeverSTAMARILLyophilizedSanofiAgence nationaleactivePasteur SAde sécurité ducomponentmédicament et desto beproduits de santéreconstitutedwithexcipientdiluentbefore useMeningococcalPOLYSACCHARIDELyophilizedSanofiAgence nationaleA + CMENINGOCOCCALactivePasteur SAde sécurité duA + C VACCINEcomponentmédicament et desto beproduits de santéreconstitutedwithexcipientdiluentbefore usePolio Vaccine - OralORALLiquid:SanofiAgence nationale(OPV) MonovalentMONOVALENTready to usePasteur SAde sécurité duType 1TYPE 1médicament et desPOLIOMYELITISproduits de santéVACCINEcholera: inactivatedDukoralLiquid:ValnevaMedical Productsoralready to useSwedenAgencyABBCGBCGLyophilizedSerumCentral DrugsVaccineactiveInstitute ofStandard ControlcomponentIndia Pvt.Organizationto beLtd.reconstitutedwithexcipientdiluentbefore useDiphtheria-TetanusDiphtheriaLiquid:SerumCentral Drugsand Tetanusready to useInstitute ofStandard ControlVaccineIndia Pvt.OrganizationAdsorbedLtd.(Paediatric)Diphtheria-TetanusDiphtheriaLiquid:SerumCentral Drugsand Tetanusready to useInstitute ofStandard ControlVaccineIndia Pvt.OrganizationAdsorbedLtd.(Pediatric)Diphtheria-TetanusDiphtheriaLiquid:SerumCentral Drugsand Tetanusready to useInstitute ofStandard ControlVaccineIndia Pvt.OrganizationAdsorbedLtd.(Pediatric)Diphtheria-TetanusDiphtheriaLiquid:SerumCentral Drugs(reduced antigenand Tetanusready to useInstitute ofStandard Controlcontent)VaccineIndia Pvt.OrganizationAdsorbed forLtd.Adults andAdolescentsDiphtheria-TetanusDiphtheriaLiquid:SerumCentral Drugs(reduced antigenand Tetanusready to useInstitute ofStandard Controlcontent)VaccineIndia Pvt.OrganizationAdsorbed forLtd.Adults andAdolescentsDiphtheria-TetanusDiphtheriaLiquid:SerumCentral Drugs(reduced antigenand Tetanusready to useInstitute ofStandard Controlcontent)VaccineIndia Pvt.OrganizationAdsorbed forLtd.Adults andAdolescentsDiphtheria-Tetanus-Diphtheria-Tetanus-Liquid:SerumCentral DrugsPertussisPertussisready to useInstitute ofStandard Control(whole cell)VaccineIndia Pvt.OrganizationAdsorbedLtd.Diphtheria-Tetanus-Diphtheria-Tetanus-Liquid:SerumCentral DrugsPertussisPertussisready to useInstitute ofStandard Control(whole cell)VaccineIndia Pvt.OrganizationAdsorbedLtd.Diphtheria-Tetanus-Diphtheria-Tetanus-Liquid:SerumCentral DrugsPertussisPertussisready to useInstitute ofStandard Control(whole cell)VaccineIndia Pvt.OrganizationAdsorbedLtd.Diphtheria-Tetanus-Diphtheria,Liquid:SerumCentral DrugsPertussisTetanus,ready to useInstitute ofStandard Control(whole cell)-Pertussis andIndia Pvt.OrganizationHepatitis BHepatitis BLtd.VaccineAdsorbedDiphtheria-Tetanus-Diphtheria,Liquid:SerumCentral DrugsPertussisTetanus,ready to useInstitute ofStandard Control(whole cell)-Pertussis andIndia Pvt.OrganizationHepatitis BHepatitis BLtd.VaccineAdsorbedDiphtheria-Tetanus-Diphtheria,Liquid:SerumCentral DrugsPertussisTetanus,ready to useInstitute ofStandard Control(whole cell)-Pertussis andIndia Pvt.OrganizationHepatitis BHepatitis BLtd.VaccineAdsorbedHepatitis BHepatitis BLiquid:SerumCentral DrugsVaccineready to useInstitute ofStandard Control(rDNA)India Pvt.Organization(Adult)Ltd.Hepatitis BHepatitis BLiquid:SerumCentral DrugsVaccineready to useInstitute ofStandard Control(rDNA)India Pvt.Organization(Adult)Ltd.Hepatitis BHepatitis BLiquid:SerumCentral DrugsVaccineready to useInstitute ofStandard Control(rDNA)India Pvt.Organization(Paediatric)Ltd.Hepatitis BHepatitis BLiquid:SerumCentral DrugsVaccineready to useInstitute ofStandard Control(rDNA)India Pvt.Organization(Paedriatic)Ltd.Tetanus ToxoidTetanus ToxoidLiquid:SerumCentral DrugsVaccineready to useInstitute ofStandard ControlAdsorbedIndia Pvt.OrganizationLtd.Tetanus ToxoidTetanus ToxoidLiquid:SerumCentral DrugsVaccineready to useInstitute ofStandard ControlAdsorbedIndia Pvt.OrganizationLtd.Tetanus ToxoidTetanus ToxoidLiquid:SerumCentral DrugsVaccineready to useInstitute ofStandard ControlAdsorbedIndia Pvt.OrganizationLtd.Measles andMeasles andLyophilizedSerumCentral DrugsRubellaRubellaactiveInstitute ofStandard ControlVaccine,componentIndia Pvt.OrganizationLive,to beLtd.Attenuatedreconstitutedwithexcipientdiluentbefore useMeasles andMeasles andLyophilizedSerumCentral DrugsRubellaRubellaactiveInstitute ofStandard ControlVaccine,componentIndia Pvt.OrganizationLive,to beLtd.Attenuatedreconstitutedwithexcipientdiluentbefore useMeasles andMeasles andLyophilizedSerumCentral DrugsRubellaRubellaactiveInstitute ofStandard ControlVaccine,componentIndia Pvt.OrganizationLive,to beLtd.Attenuatedreconstitutedwithexcipientdiluentbefore useMeasles andMeasles andLyophilizedSerumCentral DrugsRubellaRubellaactiveInstitute ofStandard ControlVaccine,componentIndia Pvt.OrganizationLive,to beLtd.Attenuatedreconstitutedwithexcipientdiluentbefore useMeasles,Measles,LyophilizedSerumCentral DrugsMumps andMumps andactiveInstitute ofStandard ControlRubellaRubellacomponentIndia Pvt.OrganizationVaccine,to beLtd.Live,reconstitutedAttenuatedwithexcipientdiluentbefore useMeasles,Measles,LyophilizedSerumCentral DrugsMumps andMumps andactiveInstitute ofStandard ControlRubellaRubellacomponentIndia Pvt.OrganizationVaccine,to beLtd.Live,reconstitutedAttenuatedwithexcipientdiluentbefore useMeasles,Measles,LyophilizedSerumCentral DrugsMumps andMumps andactiveInstitute ofStandard ControlRubellaRubellacomponentIndia Pvt.OrganizationVaccine,to beLtd.Live,reconstitutedAttenuatedwithexcipientdiluentbefore useMeasles,Measles,LyophilizedSerumCentral DrugsMumps andMumps andactiveInstitute ofStandard ControlRubellaRubellacomponentIndia Pvt.OrganizationVaccine,to beLtd.Live,reconstitutedAttenuatedwithexcipientdiluentbefore useMeaslesMeaslesLyophilizedSerumCentral DrugsVaccine,activeInstitute ofStandard ControlLive,componentIndia Pvt.OrganizationAttenuatedto beLtd.reconstitutedwithexcipientdiluentbefore useMeaslesMeaslesLyophilizedSerumCentral DrugsVaccine,activeInstitute ofStandard ControlLive,componentIndia Pvt.OrganizationAttenuatedto beLtd.reconstitutedwithexcipientdiluentbefore useMeaslesMeaslesLyophilizedSerumCentral DrugsVaccine,activeInstitute ofStandard ControlLive,componentIndia Pvt.OrganizationAttenuatedto beLtd.reconstitutedwithexcipientdiluentbefore useMeaslesMeaslesLyophilizedSerumCentral DrugsVaccine,activeInstitute ofStandard ControlLive,componentIndia Pvt.OrganizationAttenuatedto beLtd.reconstitutedwithexcipientdiluentbefore useRubellaRubellaLyophilizedSerumCentral DrugsVaccine,activeInstitute ofStandard ControlLive,componentIndia Pvt.OrganizationAttenuatedto beLtd.reconstitutedwithexcipientdiluentbefore useRubellaRubellaLyophilizedSerumCentral DrugsVaccine,activeInstitute ofStandard ControlLive,componentIndia Pvt.OrganizationAttenuatedto beLtd.reconstitutedwithexcipientdiluentbefore useRubellaRubellaLyophilizedSerumCentral DrugsVaccine,activeInstitute ofStandard ControlLive,componentIndia Pvt.OrganizationAttenuatedto beLtd.reconstitutedwithexcipientdiluentbefore useRubellaRubellaLyophilizedSerumCentral DrugsVaccine,activeInstitute ofStandard ControlLive,componentIndia Pvt.OrganizationAttenuatedto beLtd.reconstitutedwithexcipientdiluentbefore useTetanus ToxoidShanTTLiquid:ShanthaCentral Drugsready to useBiotechnicsStandard ControlPrivateOrganizationLimited (ASanofiCompany)Tetanus ToxoidShanTTLiquid:ShanthaCentral Drugsready to useBiotechnicsStandard ControlPrivateOrganizationLimited (ASanofiCompany)Diphtheria-Tetanus-Shan-5Liquid:ShanthaCentral DrugsPertussisready to useBiotechnicsStandard Control(whole cell)-PrivateOrganizationHepatitis B-Limited (AHaemophilusSanofiinfluenzae type bCompany)Diphtheria-Tetanus-Shan-5Liquid:ShanthaCentral DrugsPertussisready to useBiotechnicsStandard Control(whole cell)-PrivateOrganizationHepatitis B-Limited (AHaemophilusSanofiinfluenzae type bCompany)BCGBCGLyophilizedAJDanish MedicinesVaccine SSIactiveVaccinesAgencycomponentA/Sto bereconstitutedwithexcipientdiluentbefore useRotavirusRotateqLiquid:MerckCBER/FDAready to useVaccinesMeasles,rHA M-M-RLyophilizedMerckEuropeanMumps andIIactiveVaccinesMedicines AgencyRubellacomponentto bereconstitutedwithexcipientdiluentbefore useRotavirusRotarixLiquid:GlaxoSmithKlineFederal Agency forready to useBiologicalsMedicines andSAHealth ProductsYellow Fever—LyophilizedFederalFederal Service onactiveStateSurveillance incomponentBudgetaryHealthcareto beScientific(ROSZDRAVNADZOR)reconstitutedInstitutionof the Russian Federationwith«ChumakovexcipientFederaldiluentScientificbefore useCenter forReserch &Developmentof Immune-AndBiologicalProducts»,RussianAcademyof SciencesYellow Fever—LyophilizedFederalFederal Service onactiveStateSurveillance incomponentBudgetaryHealthcareto beScientific(ROSZDRAVNADZOR)reconstitutedInstitutionof the Russian Federationwith«ChumakovexcipientFederaldiluentScientificbefore useCenter forReserch &Developmentof Immune-AndBiologicalProducts»,RussianAcademyof SciencesYellow Fever—LyophilizedFederalFederal Service onactiveStateSurveillance incomponentBudgetaryHealthcareto beScientific(ROSZDRAVNADZOR)reconstitutedInstitutionof the Russian Federationwith«ChumakovexcipientFederaldiluentScientificbefore useCenter forReserch &Developmentof Immune-AndBiologicalProducts»,RussianAcademyof SciencesHumanGardasilLiquid:MerckEuropeanPapillomavirusready to useVaccinesMedicines Agency(Quadrivalent)HumanCervarixLiquid:GlaxoSmithKlineFederal Agency forPapillomavirusready to useBiologicalsMedicines and(Bivalent)SAHealth ProductsHumanCervarixLiquid:GlaxoSmithKlineFederal Agency forPapillomavirusready to useBiologicalsMedicines and(Bivalent)SAHealth ProductsPolio Vaccine - OralPolio SabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) MonovalentMono T1ready to useBiologicalsMedicines andType 1SAHealth ProductsPolio Vaccine - OralPolio SabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) MonovalentMono T1ready to useBiologicalsMedicines andType 1SAHealth ProductsPolio Vaccine - OralPolio SabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) Bivalent TypesOne andready to useBiologicalsMedicines and1 and 3ThreeSAHealth ProductsPolio Vaccine - OralPolio SabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) Bivalent TypesOne andready to useBiologicalsMedicines and1 and 3ThreeSAHealth ProductsHaemophilusHaemophilusLyophilizedSerumCentral Drugsinfluenzae type binfluenzae type bactiveInstitute ofStandard ControlConjugatecomponentIndia Pvt.OrganizationVaccineto beLtd.reconstitutedwithexcipientdiluentbefore usePolio Vaccine - OralMonovalentLiquid:HaffkineCentral Drugs(OPV) Monovalenttype 1 Oralready to useBioStandard ControlType 1PoliomyelitisPharmaceuticalOrganizationvaccine, IPCorporation(mOPV1)LtdPolio Vaccine - OralMonovalentLiquid:PT BioNational Agency of(OPV) MonovalentOralready to useFarmaDrug and FoodType 1Poliomyelitis(Persero)Control IndonesiaVaccineType 1(mOPV1)Tetanus ToxoidNone usedLiquid:BiologicalCentral Drugson labellingready to useE. LimitedStandard Controlfor supplyOrganizationthrough UNagencies.Alsomarketedwith labelledcommercialname BEtt.PneumococcalSynflorixLiquid:GlaxoSmithKlineEuropean(conjugate)ready to useBiologicalsMedicines AgencySADiphtheria-Tetanus-Diphtheria,LyophilizedSerumCentral DrugsPertussisTetanus,activeInstitute ofStandard Control(whole cell)-Pertussis,componentIndia Pvt.OrganizationHepatitis B-Hepatitis B andto beLtd.HaemophilusHaemophilusreconstitutedinfluenzae type binfluenzae type bwith liquidConjugateactiveVaccinecomponentbefore usePolio Vaccine - OralBivalentLiquid:PT BioNational Agency of(OPV) Bivalent TypesOralready to useFarmaDrug and Food1 and 3Poliomyelitis(Persero)Control IndonesiaVaccineType 1&3(bOPV 1&3)Meningococcal AMeningococcal ALyophilizedSerumCentral DrugsConjugate 10 μgConjugateactiveInstitute ofStandard ControlMenAfriVaccomponentIndia Pvt.Organizationto beLtd.reconstitutedwithexcipientdiluentbefore useHaemophilusQuimi-HibLiquid:Centro deCentro para elinfluenzae type bready to useIngenieriaControl Estatal deGenetica yla Calidad de losBiotecnologiaMedicamentosPneumococcalSynflorixLiquid:GlaxoSmithKlineEuropean(conjugate)ready to useBiologicalsMedicines AgencySAInfluenza, seasonalFluvirinLiquid:SeqirusCBER/FDAready to useVaccinesLimitedPolio Vaccine - OralBivalent typeLiquid:HaffkineCentral Drugs(OPV) Bivalent Types1&3 Oralready to useBioStandard Control1 and 3PoliomyelitisPharmaceuticalOrganizationvaccine, IPCorporation(bOPV1&3)LtdInfluenza, seasonalFluzoneLiquid:SanofiCBER/FDAready to usePasteur-USAInfluenza, seasonalFluzoneLiquid:SanofiCBER/FDAready to usePasteur-USADiphtheria-Tetanus-Diphtheria,LyophilizedSerumCentral DrugsPertussisTetanus,activeInstitute ofStandard Control(whole cell)-Pertussis,componentIndia Pvt.OrganizationHepatitis B-Hepatitis B andto beLtd.HaemophilusHaemophilusreconstitutedinfluenzae type binfluenzae type bwith liquidConjugateactiveVaccinecomponentbefore useInfluenza, seasonalGC FLULiquid:GreenMinistry of FoodMulti inj.ready to useCrossand Drug SafetyCorporationDiphtheria-Tetanus-Diphtheria,LyophilizedSerumCentral DrugsPertussisTetanus,activeInstitute ofStandard Control(whole cell)-Pertussis,componentIndia Pvt.OrganizationHepatitis B-Hepatitis B andto beLtd.HaemophilusHaemophilusreconstitutedinfluenzae type binfluenzae type bwith liquidConjugateactiveVaccinecomponentbefore useInfluenza, pandemicPanvaxLiquid:SeqirusTherapeutic GoodsH1N1ready to useLimitedAdministrationInfluenza, pandemicGreen Flu-SLiquid:GreenMinistry of FoodH1N1ready to useCrossand Drug SafetyCorporationInfluenza, pandemicInfluenza ALiquid:MedImmuneCBER/FDAH1N1(H1N1) 2009ready to usemonovalentvaccineInfluenza, pandemicCelturaLiquid:SeqirusPaul-Ehrlich-H1N1ready to useGmbHInstitutInfluenza, pandemicFocetriaLiquid:SeqirusH1N1ready to useVaccinesLimitedInfluenza, pandemicFluvirin-Liquid:SeqirusCBER/FDAH1N1H1N1ready to useVaccinesLimitedInfluenza, pandemicPanenzaLiquid:SanofiAgence nationaleH1N1ready to usePasteur SAde sécurité dumédicament et desproduits de santéInfluenza, pandemicInfluenza ALiquid:SanofiCBER/FDAH1N1(H1N1) 2009ready to usePasteur-monovalentUSAvaccineInfluenza, pandemicInfluenza ALiquid:SanofiCBER/FDAH1N1(H1N1) 2009ready to usePasteur-monovalentUSAvaccineDiphtheria-Tetanus-Diphtheria,LyophilizedSerumCentral DrugsPertussisTetanus,activeInstitute ofStandard Control(whole cell)-Pertussis andcomponentIndia Pvt.OrganizationHaemophilusHaemophilusto beLtd.influenzae type binfluenzae type breconstitutedConjugatewith liquidVaccineactivecomponentbefore usePolio Vaccine -PoliorixLiquid:GlaxoSmithKlineFederal Agency forInactivated (IPV)ready to useBiologicalsMedicines andSAHealth ProductsPolio Vaccine -PoliorixLiquid:GlaxoSmithKlineFederal Agency forInactivated (IPV)ready to useBiologicalsMedicines andSAHealth ProductsPneumococcalPrevenar 13Liquid:PfizerEuropean(conjugate)ready to useMedicines AgencyDiphtheria-Tetanus-Diphtheria,Liquid:SerumCentral DrugsPertussisTetanus,ready to useInstitute ofStandard Control(whole cell)-Pertussis,India Pvt.OrganizationHepatitis B-Hepatitis B andLtd.HaemophilusHaemophilusinfluenzae type binfluenzae type bConjugateVaccineAdsorbedDiphtheria-Tetanus-Diphtheria,Liquid:SerumCentral DrugsPertussisTetanus,ready to useInstitute ofStandard Control(whole cell)-Pertussis,India Pvt.OrganizationHepatitis B-Hepatitis B andLtd.HaemophilusHaemophilusinfluenzae type binfluenzae type bConjugateVaccineAdsorbedDiphtheria-Tetanus-Diphtheria,Liquid:SerumCentral DrugsPertussisTetanus,ready to useInstitute ofStandard Control(whole cell)-Pertussis,India Pvt.OrganizationHepatitis B-Hepatitis B andLtd.HaemophilusHaemophilusinfluenzae type binfluenzae type bConjugateVaccineAdsorbedPolio Vaccine - OralPolio SabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) MonovalentMono Threeready to useBiologicalsMedicines andType 3(oral)SAHealth ProductsPolio Vaccine - OralPolio SabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) MonovalentMono Threeready to useBiologicalsMedicines andType 3(oral)SAHealth ProductsPolio Vaccine -PoliomyelitisLiquid:BilthovenMedicinesInactivated (IPV)vaccineready to useBiologicalsEvaluation Board(MEB)Polio Vaccine -IPV VaccineLiquid:AJDanish MedicinesInactivated (IPV)SSIready to useVaccinesAgencyA/SInfluenza, seasonalGC FLU injLiquid:GreenMinistry of Foodready to useCrossand Drug SafetyCorporationPolio Vaccine - OralPolio SabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) MonovalentMono Twoready to useBiologicalsMedicines andType 2(oral)SAHealth ProductsPolio Vaccine - OralPolio SabinLiquid:GlaxoSmithKlineFederal Agency for(OPV) MonovalentMono Twoready to useBiologicalsMedicines andType 2(oral)SAHealth ProductsTyphoidTyphim-ViLiquid:SanofiAgence nationale(Polysaccharide)ready to usePasteur SAde sécurité dumédicament et desproduits de santéInfluenza, seasonalVaxigripLiquid:SanofiAgence nationaleready to usePasteur SAde sécurité dumédicament et desproduits de santéPolio Vaccine - OralBIOPOLIOLiquid:BharatCentral Drugs(OPV) Bivalent TypesB1/3ready to useBiotechStandard Control1 and 3InternationalOrganizationLimitedDiphtheria-TetanusnoneLiquid:PT BioNational Agency of(reduced antigenready to useFarmaDrug and Foodcontent)(Persero)Control IndonesiaPolio Vaccine - OralnoneLiquid:SanofiAgence nationale(OPV) Bivalent Typesready to usePasteur SAde sécurité du1 and 3médicament et desproduits de santéDiphtheria-Tetanus-None usedLyophilizedBiologicalCentral DrugsPertussison labellingactiveE. LimitedStandard Control(whole cell)-for supplycomponentOrganizationHepatitis B-through UNto beHaemophilusagencies.reconstitutedinfluenzae type bAlsowith liquidmarketedactivewith labelledcomponentcommercialbefore usenameComBE Five(Reconstituted).Diphtheria-Tetanus-None usedLyophilizedBiologicalCentral DrugsPertussison labellingactiveE. LimitedStandard Control(whole cell)-for supplycomponentOrganizationHepatitis B-through UNto beHaemophilusagencies.reconstitutedinfluenzae type bAlsowith liquidmarketedactivewith labelledcomponentcommercialbefore usenameComBE Five(Reconstituted).cholera: inactivatedShancholLiquid:ShanthaCentral Drugsoralready to useBiotechnicsStandard ControlPrivateOrganizationLimited (ASanofiCompany)Measles,PriorixLyophilizedGlaxoSmithKlineFederal Agency forMumps andactiveBiologicalsMedicines andRubellacomponentSAHealth Productsto bereconstitutedwithexcipientdiluentbefore useMeaslesMeaslesLyophilizedPT BioNational Agency ofvaccineactiveFarmaDrug and Foodcomponent(Persero)Control Indonesiato bereconstitutedwithexcipientdiluentbefore usePolio Vaccine - OralPoliomyelitisLiquid:SerumCentral Drugs(OPV) Bivalent TypesVaccineready to useInstitute ofStandard Control1 and 3(Oral),India Pvt.OrganizationBivalentLtd.types 1 and 3Influenza, pandemicNASOVACLyophilizedSerumCentral DrugsH1N1InfluenzaactiveInstitute ofStandard ControlVaccine,componentIndia Pvt.OrganizationLiveto beLtd.Attenuatedreconstituted(Human)withFreeze-Driedexcipientdiluentbefore useInfluenza, pandemicNASOVACLyophilizedSerumCentral DrugsH1N1InfluenzaactiveInstitute ofStandard ControlVaccine,componentIndia Pvt.OrganizationLiveto beLtd.Attenuatedreconstituted(Human)withFreeze-Driedexcipientdiluentbefore useTetanus ToxoidNone usedLiquid:BiologicalCentral Drugson labellingready to useE. LimitedStandard Controlfor supplyOrganizationthrough UNagencies.Alsomarketedwith labelledcommercialname BEtt.Tetanus ToxoidNone usedLiquid:BiologicalCentral Drugson labellingready to useE. LimitedStandard Controlfor supplyOrganizationthrough UNagencies.Alsomarketedwith labelledcommercialname BEtt.JapaneseJEEV ®Liquid:BiologicalCentral DrugsEncephalitisready to useE. LimitedStandard ControlVaccineOrganization(Inactivated)6 μgHepatitis A (HumanHavrix 1440Liquid:GlaxoSmithKlineFederal Agency forDiploid Cell),Adultready to useBiologicalsMedicines andInactivated (Adult)SAHealth ProductsHepatitis A (HumanHavrix 720Liquid:GlaxoSmithKlineFederal Agency forDiploid Cell),Juniorready to useBiologicalsMedicines andInactivatedSAHealth Products(Paediatric)Diphtheria-Tetanus-BoostrixLiquid:GlaxoSmithKlineFederal Agency forPertussisready to useBiologicalsMedicines and(acellular)SAHealth ProductsMeningococcalMenveoLyophilizedGlaxoSmithKlineEuropeanACYW-135activeVaccinesMedicines Agency(conjugate vaccine)componentS.r.l.to bereconstitutedwith liquidactivecomponentbefore useMeningococcalMenactraLiquid:SanofiCBER/FDAACYW-135ready to usePasteur-(conjugate vaccine)USADiphtheria-Tetanus-Easyfive-TTLiquid:PanaceaCentral DrugsPertussisready to useBiotec Ltd.Standard Control(whole cell)-OrganizationHepatitis B-Haemophilusinfluenzae type bJapaneseJapaneseLyophilizedChengduNational MedicalEncephalitisEncephalitisactiveInstitute ofProductsVaccine (live,Vaccine LivecomponentBiologicalAdministrationattenuated)(SA14-14-2)to beProductsreconstitutedCo., Ltdwithexcipientdiluentbefore useJapaneseJapaneseLyophilizedChengduNational MedicalEncephalitisEncephalitisactiveInstitute ofProductsVaccine (live,Vaccine LivecomponentBiologicalAdministrationattenuated)(SA14-14-2)to beProductsreconstitutedCo., Ltdwithexcipientdiluentbefore useDiphtheria-Tetanus-None usedLiquid:BiologicalCentral DrugsPertussison labellingready to useE. LimitedStandard Control(whole cell)-for supplyOrganizationHepatitis B-through UNHaemophilusagencies.influenzae type bAlsomarketedwith labelledcommercialnameComBE Five(Liquid).Diphtheria-Tetanus-None usedLiquid:BiologicalCentral DrugsPertussison labellingready to useE. LimitedStandard Control(whole cell)-for supplyOrganizationHepatitis B-through UNHaemophilusagencies.influenzae type bAlsomarketedwith labelledcommercialnameComBE Five(Liquid).JapaneseIMOJEVLyophilizedGPO-MBPThai Food andEncephalitisMDactiveCo., Ltd.DrugVaccine (live,componentAdministrationattenuated)to bereconstitutedwithexcipientdiluentbefore useDiphtheria-Tetanus-None usedLiquid:BiologicalCentral DrugsPertussison labellingready to useE. LimitedStandard Control(whole cell)for supplyOrganizationthrough UNagencies.Alsomarketedwith labelledcommercialnameTRIPVACDiphtheria-Tetanus-None usedLiquid:BiologicalCentral DrugsPertussison labellingready to useE. LimitedStandard Control(whole cell)for supplyOrganizationthrough UNagencies.Alsomarketedwith labelledcommercialnameTRIPVACDiphtheria-TetanusNone usedLiquid:BiologicalCentral Drugs(reduced antigenon labellingready to useE. LimitedStandard Controlcontent)for supplyOrganizationthrough UNagencies.Alsomarketedwith labelledcommercialname BE TdDiphtheria-TetanusNone usedLiquid:BiologicalCentral Drugs(reduced antigenon labellingready to useE. LimitedStandard Controlcontent)for supplyOrganizationthrough UNagencies.Alsomarketedwith labelledcommercialname BE TdPolio Vaccine - OralPoliomyelitisLiquid:SerumCentral Drugs(OPV) Bivalent TypesVaccineready to useInstitute ofStandard Control1 and 3(Oral),India Pvt.OrganizationBivalentLtd.types 1 and 3Polio Vaccine -PoliomyelitisLiquid:BilthovenMedicinesInactivated (IPV)vaccineready to useBiologicalsEvaluation Boardmultidose,(MEB)suspensionfor injection2.5 mLInfluenza, seasonalNasovac-SLyophilizedSerumCentral DrugsInfluenzaactiveInstitute ofStandard ControlVaccine,componentIndia Pvt.OrganizationLive,to beLtd.Attenuatedreconstituted(Human)withexcipientdiluentbefore useDiphtheria-Tetanus-HexaximLiquid:SanofiEuropeanPertussisready to usePasteur SAMedicines Agency(acellular)-Hepatitis B-Haemophilusinfluenzae type b-Polio (Inactivated)Meningococcal AMeningococcal ALyophilizedSerumCentral DrugsConjugate 5 μgConjugate 5activeInstitute ofStandard ControlmicrogramscomponentIndia Pvt.OrganizationMenAfriVacto beLtd.5 μgreconstitutedwithexcipientdiluentbefore useDiphtheria-Tetanus-None usedLiquid:BiologicalCentral DrugsPertussison labellingready to useE. LimitedStandard Control(whole cell)-for supplyOrganizationHepatitis B-through UNHaemophilusagencies.influenzae type bAlsomarketedwith labelledcommercialnameComBE Five(Liquid).Diphtheria-Tetanus-None usedLiquid:BiologicalCentral DrugsPertussison labellingready to useE. LimitedStandard Control(whole cell)-for supplyOrganizationHepatitis B-through UNHaemophilusagencies.influenzae type bAlsomarketedwith labelledcommercialnameComBE Five(Liquid).Polio Vaccine -PoliomyelitisLiquid:SerumCentral DrugsInactivated (IPV)Vaccineready to useInstitute ofStandard Control(Inactivated)India Pvt.OrganizationLtd.Polio Vaccine - OralBIOPOLIOLiquid:BharatCentral Drugs(OPV) Trivalentready to useBiotechStandard ControlInternationalOrganizationLimitedPolio Vaccine - OralBIOPOLIOLiquid:BharatCentral Drugs(OPV) Trivalentready to useBiotechStandard ControlInternationalOrganizationLimitedInfluenza, seasonalInfluenzaLiquid:HualanNational MedicalVaccineready to useBiologicalProducts(Split virion,BacterinAdministrationinactivated)Co., LtdInfluenza, seasonalIL-YANGLiquid:IL-YANGMinistry of FoodFLUready to usePHARMACEUTICALand Drug SafetyVaccine INJ.CO., LTD.BCGBCG vaccineLyophilizedGreenSignalCentral Drugs(FreezeactiveBioStandard ControlDried) -componentPharmaOrganizationIntradermalto beLimitedreconstitutedwithexcipientdiluentbefore useInfluenza, seasonalFluzoneLiquid:SanofiCBER/FDAQuadrivalentQuadrivalentready to usePasteur-USAInfluenza, seasonalFluzoneLiquid:SanofiCBER/FDAQuadrivalentQuadrivalentready to usePasteur-USAPolio Vaccine - OralBivalentLiquid:PT BioNational Agency of(OPV) Bivalent TypesOralready to useFarmaDrug and Food1 and 3Poliomyelitis(Persero)Control IndonesiaVaccineType 1&3(bOPV 1&3)cholera: inactivatedEuvicholLiquid:EuBiologicsMinistry of Foodoralready to useCo., Ltd.and Drug SafetyPolio Vaccine - OralORALLiquid:SanofiAgence nationale(OPV) MonovalentMONOVALENTready to usePasteur SAde sécurité duType 2TYPE 2médicament et desPOLIOMYELITISproduits de santéVACCINE(mOPV2)Polio Vaccine - OralORALLiquid:SanofiAgence nationale(OPV) MonovalentMONOVALENTready to usePasteur SAde sécurité duType 3TYPE 3médicament et desPOLIOMYELITISproduits de santéVACCINEMeningococcalNimenrixLyophilizedPfizerEuropeanACYW-135activeMedicines Agency(conjugate vaccine)componentto bereconstitutedwithexcipientdiluentbefore useDiphtheria-Tetanus-EupentaLiquid:LG ChemMinistry of FoodPertussisready to useLtdand Drug Safety(whole cell)-Hepatitis B-Haemophilusinfluenzae type bDiphtheria-Tetanus-EupentaLiquid:LG ChemMinistry of FoodPertussisready to useLtdand Drug Safety(whole cell)-Hepatitis B-Haemophilusinfluenzae type bHumanGardasil 9Liquid:MerckEuropeanPapillomavirusready to useVaccinesMedicines Agency(Ninevalent)Influenza, seasonalGCFLULiquid:GreenMinistry of FoodQuadrivalentQuadrivalentready to useCrossand Drug Safetyinj.CorporationDiphtheria-Tetanus-PentabioLiquid:PT BioNational Agency ofPertussisready to useFarmaDrug and Food(whole cell)-(Persero)Control IndonesiaHepatitis B-Haemophilusinfluenzae type bDiphtheria-Tetanus-PentabioLiquid:PT BioNational Agency ofPertussisready to useFarmaDrug and Food(whole cell)-(Persero)Control IndonesiaHepatitis B-Haemophilusinfluenzae type bHepatitis A (HumanHEALIVELiquid:SinovacNational MedicalDiploid Cell),ready to useBiotechProductsInactivated (Adult)Co. LtdAdministrationVaricellaVarivaxLyophilizedMerckCBER/FDAactiveVaccinescomponentto bereconstitutedwithexcipientdiluentbefore useRotavirus (live,RotavacLiquid:BharatCentral Drugsattenuated)ready to useBiotechStandard ControlInternationalOrganizationLimitedDiphtheria-Tetanus-AdacelLiquid:SanofiHealth Canada -Pertussisready to usePasteurSanté Canada(acellular)LimitedInfluenza, seasonalAGRIFLULiquid:SeqirusHealth Canada -ready to useVaccinesSanté CanadaLimitedPneumococcalPrevenar 13Liquid:PfizerEuropean(conjugate)Multidoseready to useMedicines AgencyVialTyphoidTypbar-TVCLiquid:BharatCentral Drugs(Conjugate)ready to useBiotechStandard ControlInternationalOrganizationLimitedPolio Vaccine - OralPoliomyelitisLiquid:BeijingNational Medical(OPV) Bivalent TypesVaccineready to useBio-Products1 and 3(live, oralInstituteAdministrationattenuated,BiologicalhumanProductsDiploidCo., LtdCell), type 1and 3JapaneseJEEV ®Liquid:BiologicalCentral DrugsEncephalitisready to useE. LimitedStandard ControlVaccineOrganization(Inactivated)(3 μg Pediatric)Rotavirus (live,ROTASIILLyophilizedSerumCentral Drugsattenuated)activeInstitute ofStandard ControlcomponentIndia Pvt.Organizationto beLtd.reconstitutedwithexcipientdiluentbefore usePolio Vaccine -PoliomyelitisLiquid:SerumCentral DrugsInactivated (IPV)Vaccineready to useInstitute ofStandard Control(Inactivated)India Pvt.OrganizationLtd.Polio Vaccine -PoliomyelitisLiquid:SerumCentral DrugsInactivated (IPV)Vaccineready to useInstitute ofStandard Control(Inactivated)India Pvt.OrganizationLtd.Influenza, seasonalGCFLULiquid:GreenMinistry of FoodQuadrivalentQuadrivalentready to useCrossand Drug SafetyMulti inj.CorporationInfluenza, seasonalSerinfluLiquid:AbbottMedicinesready to useBiologicalsEvaluation BoardBV(MEB)Polio Vaccine -ShanIPVLiquid:ShanthaCentral DrugsInactivated (IPV)ready to useBiotechnicsStandard ControlPrivateOrganizationLimited (ASanofiCompany)Polio Vaccine - OralBivalentLiquid:PanaceaCentral Drugs(OPV) Bivalent TypesOPV Type 1ready to useBiotec Ltd.Standard Control1 and 3and 3OrganizationPoliomyelitisVaccine,Live (Oral)cholera: inactivatedEuvichol-Liquid:EuBiologicsMinistry of FoodoralPlusready to useCo., Ltd.and Drug SafetyPolio Vaccine - OralBIOPOLIOLiquid:BharatCentral Drugs(OPV) Bivalent TypesB1/3ready to useBiotechStandard Control1 and 3InternationalOrganizationLimitedBCGBCG FreezeLyophilizedJapan BCGChiba LocalDriedactiveLaboratoryGovernmentGlutamatecomponentvaccineto bereconstitutedwithexcipientdiluentbefore usePneumococcalSynflorixLiquid:GlaxoSmithKlineEuropean(conjugate)ready to useBiologicalsMedicines AgencySARotavirus (live,RotavacLiquid:BharatCentral Drugsattenuated)ready to useBiotechStandard ControlInternationalOrganizationLimitedHepatitis A (HumanHEALIVELiquid:SinovacNational MedicalDiploid Cell),ready to useBiotechProductsInactivatedCo. LtdAdministration(Paediatric)TyphoidTypbar-TVCLiquid:BharatCentral Drugs(Conjugate)ready to useBiotechStandard ControlInternationalOrganizationLimitedRotavirus (live,ROTASIILLyophilizedSerumCentral Drugsattenuated)activeInstitute ofStandard ControlcomponentIndia Pvt.Organizationto beLtd.reconstitutedwithexcipientdiluentbefore useJapaneseJEEV ®Liquid:BiologicalCentral DrugsEncephalitisready to useE. LimitedStandard ControlVaccineOrganization(Inactivated)6 μgJapaneseJEEV ®Liquid:BiologicalCentral DrugsEncephalitisready to useE. LimitedStandard ControlVaccineOrganization(Inactivated)(3 μg Pediatric)SARS-CoV-2PFIZER-Liquid:Pfizer-CEBR/FDABIONTECHready to useBIONTECHCOVID-19VACCINE-bnt162b2SARS-CoV-2ModernaLiquid:ModernaCEBR/FDACOVID-19ready to useIncSARS-CoV-2COVID-19Liquid:AstraEuropeanVaccineready to useZenecaMedicines Agency(ChAdOx1-S[recombinant])SARS-CoV-2JanssenLiquid:Johnson &CEBR/FDACOVID-19ready to useJohnsonVaccineSARS-CoV-2CoronaVac,Liquid:SinovacNational MedicalCOVID-19ready to useProductsVaccineAdministration(Vero Cell),Inactivated In various embodiments, the epitope is released following proteolytic cleavage of the peptide from the IRC. After proteolytic cleavage of the peptide from the IRC, the epitope binds to an MHC, optionally an MHC class I, molecule. The MHC molecule is in some embodiments from the HLA-A, B, and/or HLA C families. The specific epitope that binds to the MHC class I molecule is any of those recited in Table 2 or Table 3 or found elsewhere in the art. The MHC class I molecule itself is, in some embodiments, one or more of the following non-limiting examples: HLA-A*02:01, HLA-A*03:01, HLA-A*11:01, HLA-A*201, HLA-A*020101, HLA-A*0203, HLA-A*0206, HLA-A2, HLA-A2.1, or HLA-A*02. In an aspect the described methods, uses, and compositions, the epitope is about 8 amino acid to about 50 amino acids in length, or about 8 amino acid to about 45 amino acids in length, or about 8 amino acid to about 40 amino acids in length, about 8 amino acid to about 35 amino acids in length, or about 8 amino acid to about 30 amino acids in length, about 8 amino acid to about 25 amino acids in length, about 8 amino acid to about 20 amino acids in length, or is about 8 amino acid to about 15 amino acids in length. In an aspect of the invention the peptide is about 13 amino acid to about 50 amino acids in length, or about 13 amino acid to about 45 amino acids in length, or about 13 amino acid to about 40 amino acids in length, about 13 amino acid to about 35 amino acids in length, or about 13 amino acid to about 30 amino acids in length, about 13 amino acid to about 25 amino acids in length, about 13 amino acid to about 20 amino acids in length, or is about 13 amino acid to about 15 amino acids in length. In some embodiments, the CD8+ T cell epitope is, e.g., about 8, 9, 10, 11, 12, 13, 14, 15, 16, or about 17 amino acids in length. Cleavage Sequence. In various embodiments, one or more protease cleavage sequences are incorporated into the IRC that, upon cleavage, allows the peptide to be released from the IRC so that the peptide then is free to bind to the MHC on the tumor or cancer cell surface. In various embodiments, the IRC must escape the endosome, disassemble, and release their therapeutic cargo to the cytosol in a functional form. In various embodiments the IRC and/or peptide of the IRC is susceptible to cleavage by a proteolytic enzyme within the tumor microenvironment, i.e., in the nearby interstitial space surrounding tumors or tumor cells, and the position of the target cleavage sequence in the IRC or peptide is such that the cleavage of the target site releases all or a portion of the peptide comprising the CD8+ T cell epitope from the IRC, which then is free to bind to, and/or form a complex with, an MHC molecule expressed on the surface of the tumor cell in the subject. Pharmaceutically effective, or therapeutic amounts of IRC required to achieve this end goal are determined by the skilled artisan by known clinical methods utilizing in vitro cell culture techniques, animal model studies, and small scale to large scale human clinical trials. It will be appreciated that the amount of IRC administered to the subject in need thereof in the described methods and uses herein will depend on, e.g., the characteristics of the subject, e.g., age, weight, gender, and/or medical condition/history, genetic makeup, and other factors pertinent to the subject or class of subjects, and that the characteristics of the tumor, e.g., type, volume, and developmental status will also be taken into account when designing the dosage range finding clinical studies. The proteolytic cleavage sequence is in some embodiments recognized by any protease present in, on, around, or nearby a tumor cell. At least about 569 known proteases have been described. (See, Lopez-Otin, et al.,Nature Reviews Cancer,7(10):800-808, 2007). All human proteolytic enzymes identified to date are classifiable into five catalytic classes: metalloproteinases, serine, threonine, cysteine, and aspartic proteases. A non-limiting list of potential proteases is demonstrated in Table 4, which is a table summarizing exemplars of the most well-studied proteases distributed into the five noted classes. (See Choi, Ki Young et al., “Protease-activated drug development,”Theranostics, (2)2:156-78, 2012). Several of these proteases have been found to be over-expressed in cancer cells relative to healthy cells. In various embodiments, the proteolytic cleavage sequence is recognized by the protease furin, a matrix metalloproteinase (MMP), of which several different members are identified, e.g., MMP, 1, 2, 3, 7, 8, 9, 11, 13, 14, or 19, an ADAM (a disintegrin and metalloproteinase), e.g., ADAMS 8, 9, 10, 15, 17, or 28, a cathepsin, e.g., cathepsin D, G, H, or N. Also contemplated herein are the proteases elastase, proteinase-3, azurocidin, and ADAMTS-1. In various embodiments, the cleavage sequence is recognized by any one or more of the aforementioned proteases, and in a certain embodiment the sequence is recognized by a human furin protease. In various embodiments, the cleavage sequence comprises at least about 4 amino acid residues, at least about three of which are arginine residues. In various embodiments, the cleavage sequence comprises at least 4 amino acid residues, at least three of which are arginine residues and one of which is either a lysine residue or an arginine residue. In various embodiments, the cleavage sequence is R—X—R/K—R (SEQ ID NO: 89). In various embodiments, the cleavage sequence comprises additional residues. In various embodiments, the cleavage sequence further comprises about 1, 2, 3, 4, 5, 6, 7, 8, or about 9 additional arginine residues. It is known that arginines are positively charged and it has been discovered that a longer chain of positive charged arginine residues will bring the peptides closer to the surface of the capsid backbone which is more negatively charged. TABLE 4Proteases and cancers associated with overexpressed proteasesFamilyProteaseLocationCancerRef.Other DiseasesRef.CysteineGeneralIntracellular,MostTable inCathepsinslysosomes[121]Cathepsin KExtracellular,Breast[178]Artherosclerosis,[179-182]boneosteoporosisCathepsin BExtracellularBreast, cervix, colon,[31, 38, 81,andcolorectal, gastric, head and183-196]pericellularneck, liver, lung, melanoma,underovarian, pancreatic, prostate,pathologicalthyroidconditionsAsparticCathepsin LBreast, colorectal[28]AD[197]CathepsinsCathepsin EEndosomalCervical, gastric, lung,[51-55]structures, ER,pancreas adenocarcinomasGolgiCathepsin DLysosomeBreast, colorectal, ovarian[47-49,Atherosclerosis[121]198-200]GeneralIntracellular,MostTable insecreted[15, 58]KallikreinshK1Hypertension,[24](hK)inflammationPSA (hK 3)Prostate, ovarian[201-202]hK10Colon, ovarian, pancreatic,[203-206]head and neckhK15Ovarian, prostate[207-208]SerineuPA, uPARMembrane,Cervical, colorectal, gastric,[86, 116,ProteasesPericellularprostate209-210]CaspasesIntracellularNeurodegenerative[82]disordersGeneralExtracellularMostTable in[211]MMPsMMP-1, -8, -13Breast[85, 102-104,Artherosclerosis, RA[213-214]211-212]MMP-2, -9Breast, colorectal, lung,[91-94]Bronchiectasis, chronic[87, 113-malignant gliomas, ovarian[95-98]asthma, COPD, cystic117]fibrosis, HIVassociated dementia,hypertension, strokeMMP-14MembraneBreast[212]ADAMExtracellularAD[105, 107,112]*Abbreviations: AD: Alzheimer's disease; ADAM: a disintegrin and metalloproteinase domain protease; COPD: chronic obstructive pulmonary disease; ER: endoplasmic reticulum; RA: rheumatoid arthritis In various embodiments, the peptide is bound to the capsid backbone, as described in more detail below. There are multiple known means by which the peptide is able to be associated with, or bound to, the capsid backbone. In various embodiments of the present disclosure the cleavage sequence is chemically conjugated by way of a maleimide linkage or an amide linkage (discussed below). The peptide is generally linked to any residue on the capsid backbone; however, disulfide linkages, maleimide linkages, and amide linkages are formed by conjugating the peptide to cysteine, lysine, or arginine residues of the mutant L1 proteins that comprise the capsid backbones. In various embodiments the peptide comprises at least one protease cleavage sequence. In some embodiments, the protease cleavage sequence is any sequence capable of being preferentially cleaved by or near a tumor cell. The insertion of this cleavage sequence into the peptide allows the protein to remain attached to the capsid backbone carrier until the IRC enters the tumor microenvironment. By taking advantage of the elevated activities of particular proteases in cancer tissues or tumor microenvironments, the peptide is to a large extent not released from the capsid backbone and able to actively coat MHC receptors until the peptide enters the tumor microenvironment. Several proteases are known in the art to be active in the tumor microenvironment. For example, several metallo-, cysteine and serine proteases are known. From the standpoint of cancer therapy, an additional attraction is that because the proteases responsible for prodrug cleavage may come not just from cancer cells but also from the stromal components of tumors, release of the active drug direction into the tumor microenvironment does not depend on a target expressed only by the cancer cells. Instead, it is the entire tumor ecosystem that represents the target. Methods of Attaching Peptides to the L1 Protein The capsid backbones described herein are in some embodiments first functionalized to deliver an epitope containing on one or more peptides associated with the capsid backbone to the target cells, thereby labeling the tumor or cancer cells for destruction. In various embodiments, peptides are conjugated to the capsid backbone through cysteine residues on the capsid protein. Such cysteine molecule are presented naturally, or by mutation, on the surface of the capsid backbone. In various embodiments, the capsid backbone is subjected to reducing conditions sufficient to reduce the sulfhydryl groups of cysteine residues on the surface of the capsid backbone while maintaining the capsid-like icosahedron structures of the capsid backbone. Because of its free sulfhydryl group, cysteine will readily and spontaneously form disulfide bonds with other sulfhydryl-containing ligands under oxidative conditions. Alternatively, a series of compounds are known to add a maleimide moiety to receptive substrates that readily and irreversibly form thioester linkages with cysteine residues at a pH between about 6.5 and about 7.5. Thus, in one embodiment, the peptide is associated with the capsid backbone via a maleimide linkage. In various embodiments, the peptide is conjugated to a lysine residue on the capsid backbone. Lysine residues are easily modified because of their primary amine moiety. Using reactions termed n-hydroxysuccinimide (NHS) ester reactions (because NHS is released as prat of the reaction), amide bonds are formed at surface-exposed lysine residues on the capsid backbone. The NHS reaction occurs spontaneously between about pH 7.2 and about pH 9. In various embodiments, the peptide is conjugated to an aspartate or glutamate residue. Unlike chemical coupling strategies involving cysteine and lysine groups, chemically coupling to aspartate or glutamate residues requires multiple steps. First, the carboxylic acid of the aspartate or glutamate is activated using 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC), or similar chemical cross-linking reagent. Once activated, this adduct will react with NHS to form an NHS ester. The NHS ester is then reacted with a ligand with an exposed primary amine to from a stable amide bond. In various embodiments, the capsid backbone comprises a region of negatively charged amino acids on a surface-exposed area that is capable of binding to the peptide comprising a region of positively charged amino acids. In various embodiments, the region of negatively charged amino acids is flanked, on one or on both sides, by one or more cysteine residues, referred to as polyanionic: cysteine or more specifically, polyglutamic acid:cysteine or polyaspartic acid:cysteine. In such cases, the conjugation of the capsid backbone and the peptide would result from non-covalent binding between the complementary amino acid charges of the capsid backbone and the peptide and a disulfide bond between the cysteines. In various embodiments, the cysteine(s) are one or more amino acids away from the region of charged amino acids such that any secondary/tertiary structure would bring the charged amino acid region in close proximity to the cysteine(s). In various embodiments, the peptide comprises at least one peptide and a polyionic:cysteine for attaching the peptide to the capsid backbone comprising a complementary polyionic:cysteine sequence and an enzyme cleavage site positioned between the terminal cysteine and the CD8+ T cell epitope. In various embodiments, the peptide comprises, a terminal cysteine, at least one peptide, and an enzyme cleavage sequence positioned between the terminal cysteine and the peptide(s). Negatively charged amino acids that are useful in producing the described IRC include, e.g., glutamic acid and aspartic acid. These amino acids are used singly in some embodiments, e.g., polyglutamic acid, or in combination. In a specific embodiment, the negatively charged region comprises glutamic acid. The number of negatively charged amino acids can vary and can include about 4 to about 20 amino acids, about 6 to about 18 amino acids, or about 8 to about 16 amino acids, and the like. In a specific embodiment, the negatively charged region comprises about 8 negatively charged amino acids. In a more specific embodiment, the negatively charged region comprises EEEEEEEEC (E8C, SEQ ID NO: 130). In another embodiment, the negatively charged region comprises CEEEEEEEEC (SEQ ID NO: 131). Methods for conjugating peptides to a capsid backbones via disulfide bonding are known. For instance, the presence of a polyarginine-cysteine moiety on the peptide allows docking of the peptide to the polyanionic site (EEEEEEEEC, E8C, SEQ ID NO: 130) present in the various loops of the capsid backbone. Covalent cross-linking between the two cysteine residues should render this association irreversible under oxidizing conditions. For the conjugation reactions, purified capsid backbones are dialyzed in conjugation buffer (20 mM Tris/HCl, pH 7.5, 150 mM NaCl, 5% glycerol, 0.5 mM CaCl2) and then the peptide and the oxidizing reagents are added, allowing the reaction to proceed for 16 hrs at 4° C. At the end of the incubation, the reaction mixtures are applied to a size-exclusion column (such as SEPHADEX® G-100, Pharmacia, New Jersey, US, volume 20 ml, flow rate 1 ml/min, 10 mM Tris/HCl (pH 7.4), 150 mM NaCl, 0.5 mM CaCl2)) to remove unconjugated peptide and exchange buffer. IRCs that elute in the void volume are identified by the presence of the L1 protein on SDS-PAGE. The conjugated capsid backbones (IRC) are than optionally analyzed by electron microscopy. In various embodiments, the peptide is genetically fused to the L1 protein. In various embodiments, the peptide is either covalently or non-covalently linked to the capsid backbone. Rather than attaching the peptide to the capsid backbone via, e.g., binding of negatively and positively charged amino acids, or via maleimide based conjugation, a nucleic acid sequence encoding the peptide is inserted in some embodiments into the nucleic acid encoding the L1 protein such that upon expression a peptide is produced that is inserted into a loop of the capsid protein and displayed on the surface of the capsid backbone. In various embodiments, non-natural amino acids are used to conjugate the peptide to the capsid backbone. Beyond the 20 natural amino acids, many non-natural amino acids have been used for site-specific protein conjugation reactions. For example, an azidohomoalanine (AHA) or a p-amino-phenylalanine (pAF) may be incorporated into the capsid backbone coat protein for conjugation. These amino acids are incorporated into proteins in two ways: global methionine replacement and amber stop codon suppression. Because AHA is very similar to methionine, AHA will be incorporated at each AUG codon if the methionine supply is rate limiting, this is termed global methionine replacement. Bacteria auxotrophic for methionine or cell-free protein synthesis can be used to limit-methionine availability. Amber stop codon suppression will incorporate pAF. Amber stop codon suppression uses nonnative synthetases and tRNAs that do not react with the natural amino acids to incorporate the non-natural amino acid at the amber stop codon UAG. AHA, displaying an azide, will participate in in copper(I)-catalyzed azide-alkyne cycloaddition (“click” reaction) and form covalent triazole rings with alkyne-containing ligands. In various embodiments, the IRC comprises, at least one-tenth of the L1 proteins display a peptide. In various embodiments, at least one-fifth of the L1 proteins display a peptide. In various embodiments, about half of the L1 proteins display a peptide. In various embodiments, about two-thirds of the L1 proteins display a recall peptide. In various embodiments, nearly all of the L1 proteins display a peptide. IRCs and Uses Thereof in Clinical Therapies In various embodiments, the capsid backbone binds preferentially to tumor cells. The capsid backbones' tumor preference originates, in some embodiments, from several sources such as the capsid backbone's charge (positive or negative), shape and size (different aspect ratio filaments and diameter spheres), shielding (self-proteins/peptides and polymers of various sizes and densities), and targeting (ligands for receptors or environmental factors displayed on different linkers at various densities). In terms of charge, in various embodiments, the capsid backbone contains a positive surface charge. Positively charged capsid backbones have been shown in some studies to remain longer in circulation when injected into a subject. Due to the abundant presence of proteoglycan in cell membranes that confer a negative charge to cell membranes, and collagen within the tumor interstitial space conferring a positive charge, positively charged IRCs are more likely to possess enhanced binding to mammalian cells as compared with non-charged or negatively charged IRCs, and therefor are better able to avoid aggregation and as a result, are able to better penetrate tumor tissue. Some examples demonstrating these charge-based effects include polyarginine-decorated cowpea mosaic virus (CPMV) found to be taken up eight times more efficiently than native CPMV in a human cervical cancer. (Wen et al.Chem. Soc. Rev.,45(15):4074-4126, 2016). With regards to shape, the shape and flexibility of the capsid backbone in some instances plays an additional functional role in the ability of capsid backbones to diffuse throughout a tumor. A comparison between the diffusion profiles of a spherical and rod-shaped particle was performed with CPMV and TMV using a spheroid model. It was shown in this study that the CPMV (spherical) experienced a steady diffusion profile, but the TMV (rod shaped) exhibited a two-phase diffusion behavior that entailed an extremely rapid early loading phase that could be attributed to its movement axially, like a needle. (Wen et al.,Chem. Soc. Rev.,45(15):4074-41:26, 2016). Some other advantageous properties that are conferred by elongated particles include better margination toward the vessel wall and stronger adherence due to greater surface area for interaction, which not only have implications for tumor homing but also for enhanced targeting of cardiovascular disease. Besides passive tumor homing properties, natural interactions of viruses with certain cells can also be exploited. CPMV in particular exhibits unique specificity in interacting with surface vimentin, which is found on endothelial, cancer, and inflammatory cells. (Wen et al.,Chem. Soc. Rev.,45(15):4074-4126, 2016). The native affinity of CPMV for surface vimentin allows for high-resolution imaging of microvasculature up to 500 μm in depth, which cannot be achieved through the use of other nanoparticles, as they tend to aggregate and block the vasculature. This interaction can be utilized for a range of applications, such as delivery to a panel of cancer cells including cervical, breast, and colon cancer cell lines, delineation of atherosclerotic lesions, and intravital imaging of tumor vasculature and angiogenesis. Another example of an existing endogenous association is canine parvovirus (CPV) with transferrin receptor (TfR), an important receptor for iron transport into cells and highly upregulated by numerous cancer cell lines. Even after dye labelling, CPV retains its specificity for TfR and was shown to bind to receptors found on HeLa cervical cancer cells, HT-29 colon cancer cells, and MDA-MB-231 breast cancer cells. (Wen et al.,Chem. Soc. Rev.,45(15):4074-4126, 2016). In various embodiments, the capsid backbone targets a protein expressed preferentially on the tumor cell surface in the subject. Such proteins are typically overexpressed on the surface of tumor cells, but some if not all, are also found in the blood, i.e., serum. Non-limiting examples of such surface markers include: CEA (carcinoembryonic antigen), E-cadherin, EMA (epithelial membrane antigen; aka MUC-1), vimentin, fibronectin, Her2/neu (human epidermal growth factor receptor type 2, also called Erb b2), αvβ3 integrin, EpCAM (epithelial cell adhesion molecule), FR-α (folate receptor-alpha), PAR (urokinase-type plasminogen activator receptor), and transferrin receptor (over expressed in tumor cells). Peptides are often used to label cancerous cells based on recognition of their transmembrane proteins. The most commonly used peptide is arginylglycylaspartic acid (RGD), which is composed of L-arginine, glycine, and L-aspartic acid. RGD was first isolated from the cell-binding domain of fibronectin, a glycoprotein that binds to integrins, and is involved in cell-cell and cell-extracellular matrix (ECM) attachment and signaling by binding collagen, fibrin, and proteoglycans. RGD peptides have the highest affinity for a type of cell surface integrins, αvβ which are highly expressed in tumoral endothelial cells, but not in normal endothelial cells. In various embodiments such a peptide sequence is incorporated into the IRC. Methods of treating cancers in a subject in need thereof by administering an IRC to patient in need thereof, and related uses of the described IRC compositions, are described herein. The methods described herein comprise, for instance, administering the IRCs described herein to a subject in need thereof in an amount sufficient to inhibit tumor growth, progression or metastasis, i.e., a therapeutic amount or dose. In various embodiments, the IRC is administered to a subject in need thereof in amount sufficient to stimulate cytokine production and/or cellular immunity, particularly innate immunity, including stimulation of the cytotoxic activity of macrophages and natural killer cells. In various embodiments described herein, a subject in need thereof is a subject who has been previously treated for a tumor and is currently deemed cancer-free or disease free in accordance with medical standards. Briefly, various understood aspects of what is believed to be the mechanism of action of the described IRCs are described and supported by the examples, below. The IRC first bind to a tumor cell, in some embodiments the binding is specific. (See, Example 9,FIGS.18A and18B). The peptide epitope on the IRC is then proteolytically cleaved by furin, in some embodiments, or by any other resident protease nearby the tumor cell, which is over-expressed in the tumor microenvironment. This in turn leads to release of the peptide from the IRC and the loading, or binding, of the peptide by an MHC molecule expressed on the surface of the tumor cell (“epitope coating”). (See, Examples 10 and 16, andFIGS.19,21,22, and34). The epitope-coated tumor cell is then recognized as a pathogen-infected cell by one or more T-cells responsive to the specific peptide bound in the MHC molecules, and pre-existing CD8 T cells, yielding a triggered immune redirection response. (See, Examples 11 and 12). That is, this recognition event leads to triggering or activation of the subject's preexisting immune memory against pathogens and childhood vaccines against the tumor, leading to the attacking and destroying of the subject's tumor cells. Destruction of tumor cells can result in components of the preexisting immune response being exposed to cancer cell antigens. Thus, antigens released from the killed tumor cells will initiate a further immune response to recruit additional tumor-specific CD8 T cells, or a “second wave” of T cells that then proceed to attack additional tumor cells in the area. This can result in elicitation of an endogenous immune response against the cancer cell antigens (referred in some instances to “epitope spreading”) and leads to anti-tumor immune memory. Thus, the methods and uses disclosed herein are methods of treating cancer in an subject in need thereof that occurs through utilizing, or the re-orienting of, the subject's own preexisting adaptive memory immune system to attack cancer cells. The methods and uses described herein make use of the fact that subjects, in some instances, possess preexisting immune responses that were not originally elicited in response to a cancer, but that were elicited instead by routine vaccination or via natural infection by a parasite or pathogen. Because the cancer cells would not normally express such epitopes that elicit preexisting immune responses, it would not be expected that such an immune response would not normally, without exogeneous intervention, be capable of attacking any cancer cell. However, by way of the present methods and uses described herein, such preexisting immune responses are readily recruited to attack, kill, and clear a cancer in a subject. This recruitment or repurposing effect is therefore achieved by way of the present IRC compositions since these IRC, upon injection or other means of delivery into the subject, introduce into or onto the surface of the cancer one or more epitopes known to be recognized by the preexisting immune response in the subject, resulting in cells of the immune response attacking antigen-displaying cancer cells. Thus, without wishing to be bound by any specific theory, the methods, uses, and compositions described herein act by recruiting a preexisting immune response in a subject to the site of a cancer, such that the preexisting immune response attacks and kills the cancer cells. Thus, there are generally four or five steps involved in the described methods, including: 1) binding IRC to the tumor cells, 2) cleavage of the epitope from the IRC, 3) MHC binding of the epitopes for display on the tumor cell surface, 4) recognition of the loaded MHC by the subject's pre-existing recalled immunity against the epitope, and optionally 5) triggering of a second wave and longer-term anti-tumoral immunity thereafter. Data obtained from cell culture assays and animal studies are often used in formulating a range of dosages for use in humans. The dosages of such compositions lie preferably within a range of circulating concentrations that include the ED50with little or no toxicity. The dosage varies within this range depending on the dosage form employed and the route of administration utilized. For any composition used in the methods described herein, the therapeutically effective dose is capable of being estimated initially from cell culture assays. A dose is formulated in animal models to achieve a circulating plasma concentration range that includes the IC50(the concentration of the test composition that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information is then used to accurately determine useful doses in humans. Levels in plasma are measured, for example, by high performance liquid chromatography. In many instances, it will be desirable to have multiple administrations of the IRC-containing compositions, usually at most, at least, or not exceeding 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more doses including all ranges therebetween. The administrations will normally be at 1, 2, 3, 4, 5, 6, to 5, 6, 7, 8, 9, 10, 11, to 12 week/month/year intervals, including all values and ranges there between, more usually from three- to five-week intervals. In various embodiments, a method is provided for stimulating the cytotoxic activity of macrophages and natural killer (NK) cells by administering to a subject in need thereof an effective amount of an IRC described herein. The macrophages and natural killer cells are in some instances those that are present in the tumor microenvironment. In one aspect, the IRCs are administered to the subject in an amount effective to stimulate the cytotoxic activity of macrophages and natural killer cells already present in the tumor microenvironment. In various other embodiments, the IRCs are administered to the subject in an amount effective to attract macrophages and natural killer cells to the tumor microenvironment. In various embodiments, the IRCs are administered to the subject in an amount effective to bind sufficient numbers of antibodies to the peptide or IRC capsid itself to attract and stimulate macrophages, neutrophils and natural killer cells. In various embodiments, methods and uses are provided for redirecting the cytotoxic activity of an existing memory CD8+ T cell to a tumor cell or tumor microenvironment by administering to a subject in need thereof an effective amount of the IRC described herein. Preferably, the T cell epitope of the peptide of the IRC is from a pathogen for which the subject has been vaccinated or from a pathogen that has previously infected the subject and the subject has memory CD8+ T cells that recognize the T cell epitope in complex with an MHC class I molecule on the tumor cells. In an aspect described herein, the effective or therapeutic amount of the IRC compositions described herein is an amount sufficient to attract the memory CD8+ T cell to the tumor microenvironment. In another alternative aspect, the effective amount of the IRC is an amount sufficient to stimulate the memory CD8+ T cell present in the tumor microenvironment. In various embodiments, the tumor is a small lung cell cancer, hepatocellular carcinoma, liver cancer, hepatocellular carcinoma, melanoma, metastatic melanoma, adrenal cancer, anal cancer, aplastic anemia, bile duct cancer, bladder cancer, bone cancer, brain/CNS cancer, breast cancer, cancer of unknown primary origin, Castleman disease, cervical cancer, colon/rectum cancer, endometrial cancer, esophagus cancer, Ewing family of tumors, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumor (gist), gestational trophoblastic disease, Hodgkin disease, Kaposi sarcoma, kidney cancer, laryngeal and hypopharyngeal cancer, leukemia, liver cancer, lung cancer, lymphoma, malignant mesothelioma, multiple myeloma, myelodysplastic syndrome, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer. neuroblastoma, oral cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumors, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma, skin cancer, stomach cancer, testicular cancer, thymus cancer, thyroid cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Waldenstrom macroglobulinemia, Wilms tumor, non-Hodgkin lymphoma, Hodgkin lymphoma, Burkitt's lymphoma, lymphoblastic lymphomas, mantle cell lymphoma (MCL), multiple myeloma (MM), small lymphocytic lymphoma (SLL), splenic marginal zone lymphoma, marginal zone lymphoma (extra-nodal or nodal), mixed cell type diffuse aggressive lymphomas of adults, large cell type diffuse aggressive lymphomas of adults, large cell immunoblastic diffuse aggressive lymphomas of adults, small non-cleaved cell diffuse aggressive lymphomas of adults, or follicular lymphoma, head and neck cancer, endometrial or uterine carcinoma, non-small cell lung cancer, osteosarcoma, glioblastoma, or metastatic cancer. In a preferred embodiment, the cancer is a breast cancer, a cervical cancer, an ovarian cancer, a pancreatic cancer or melanoma. The term “cancer” as used herein refers to proliferative diseases, such as lymphomas, lymphocytic leukemias, lung cancer, non-small cell lung (NSCL) cancer, bronchioloalveolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, mesothelioma, hepatocellular cancer, biliary cancer, neoplasms of the central nervous system (CNS), spinal axis tumors, brain stem glioma, glio-blastoma multiforme, astrocytomas, schwanomas, ependymonas, medulloblastomas, meningiomas, squamous cell carcinomas, pituitary adenoma and Ewing's sarcoma, including refractory versions of any of the above cancers, or a combination of one or more of the above cancers. An aspect described herein is a method for treating a cancer in a subject in need thereof by administering an IRC described herein to the subject wherein the CD8+ epitope of the peptide is of a failed therapeutic cancer vaccine against a viral-induced cancer, e.g., HPV cervical cancer, HPV+ oral cancer, EBV nasopharyngeal cancer (the “therapeutic vaccine”). The methods and uses described herein therefore comprise determining whether the subject has been actively vaccinated but did not respond with an anti-tumor effect to the treatment. The IRC composition is then administering to the subject an effective amount of an IRC of this invention wherein the CD8+ epitope of the peptide is of the antigenic determinant in the vaccine previously administered to the subject that infected the subject. Capsid backbones have inherent adjuvant properties. In some embodiments, the immunogenicity of the IRC compositions described herein are further enhanced by the combination with additional nonspecific stimulators of the immune response, known as adjuvants. Suitable adjuvants include all acceptable immunostimulatory compounds, such as, but not limited to, cytokines, toxins, or synthetic compositions such as alum. Adjuvants include, but are not limited to, oil-in-water emulsions, water-in-oil emulsions, mineral salts, polynucleotides, and natural substances. Specific adjuvants that may be used include IL-1, IL-2, IL-4, IL-7, IL-12, y-interferon, GM-CSF, BCG, aluminum salts, such as aluminum hydroxide or other aluminum compound, methylenedioxyphenyl (MDP) compounds, such as thur-MDP and nor-MOP, CGP (MTP-PE), lipid A, and monophosphoryl lipid A (MPL), or inactivated microbial agents. RIBI, which contains three components extracted from bacteria, MPL, trehalose dimycolate (TOM), and cell wall skeleton (CWS) in a 2% squalene/Tween 80 emulsion. MHC antigens may even be used. Various methods of achieving adjuvant affect for the IRC compositions includes use of agents such as aluminum hydroxide or phosphate (alum), commonly used as about 0.05 to about 0.1% solution in phosphate buffered saline, admixture with synthetic polymers of sugars (CARBOPOL®) used as an about 0.25% solution, aggregation of a protein in the composition by heat treatment with temperatures ranging between about 70° C. to about 101° C. for a 30-second to 2-minute period, respectively. Aggregation by reactivating with pepsin-treated (Fab) antibodies to albumin; mixture with bacterial cells, e.g.,C. parvum, endotoxins or lipopolysaccharide components of Gram-negative bacteria; emulsion in physiologically acceptable oil vehicles, e.g., mannide monooleate (Aracel ATM), or emulsion with a 20% solution of a perfluorocarbon (FLUOSOL-DA®) used as a block substitute may also be employed to produce an adjuvant effect. A typical adjuvant is complete Freund's adjuvant (containing killedMycobacterium tuberculosis), incomplete Freund's adjuvants, and aluminum hydroxide. For administration to humans, a variety of suitable adjuvants will be evident to a skilled worker. These include, e.g., Alum-MPL as adjuvant, or the comparable formulation, ASO4, which is used in the approved HPV vaccine CERVARIX®, AS03, AS02, MF59, montanide, saponin-based adjuvants such as GPI-0100, CpG-based adjuvants, or imiquimod. In embodiments of the invention, an adjuvant is physically coupled to the capsid backbone, or encapsulated by the capsid backbone, rather than simply mixed with them. In addition to adjuvants, it may be desirable to co-administer biologic response modifiers (BRM) to enhance immune responses. BRMs have been shown to upregulate T cell immunity or downregulate suppresser cell activity. Such BRMs include, but are not limited to, Cimetidine (CIM; 1200 mg/d) (Smith/Kline, PA, US); or low-dose Cyclophosphamide (CYP; 300 mg/ml) (Johnson/Mead, NJ, US) and cytokines such as γ-interferon, IL-2, or IL-12 or genes encoding proteins involved in immune helper functions, such as B-7. In embodiments described herein, these genes are encapsulated by the capsid backbone to facilitate their delivery into a subject. The preparation of compositions that contain polypeptide or peptide sequence(s) as active ingredients is generally well understood in the art. Typically, such compositions are prepared as injectables either as liquid solutions or suspensions: solid forms suitable for solution in or suspension in liquid prior to injection may also be prepared. The preparation is in some instances emulsified. The active immunogenic ingredient is in some embodiments mixed with excipients that are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof. In addition, if desired, the compositions may contain amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or adjuvants that enhance the effectiveness of the vaccines. In specific embodiments, vaccines are formulated with a combination of substances. The compositions comprising the IRCs of the present disclosure are intended to be in a biologically-compatible form that is suitable for administration in vivo to subjects. The pharmaceutical compositions described herein further comprise one or more optional pharmaceutically acceptable carriers. The term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government, e.g., the FDA, or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly, in humans. The term “carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the capsid backbone is administered. Such pharmaceutical carriers include, for example, sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, including but not limited to peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a carrier in some instances when the pharmaceutical composition described herein is administered orally. Saline and aqueous dextrose are carriers, for example, when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions are employed, for instance, as liquid carriers for injectable solutions. Suitable pharmaceutical excipients include, without limitation, starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried slim milk, glycerol, propylene, glycol, water, ethanol and the like. The pharmaceutical composition in some embodiments optionally contains minor amounts of wetting or emulsifying agents, or pH buffering agents. The pharmaceutical compositions comprising the IRCs of the present disclosure take the form of, for example, solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained-release formulations, and the like. Oral formulation includes in some embodiments standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. In a specific embodiment, a pharmaceutical composition comprises an effective amount of an IRC of the present disclosure together with a suitable amount of a pharmaceutically acceptable carrier so as to provide the form for proper administration to the subject. The formulation should suit the mode of administration. The pharmaceutical compositions of the present disclosure are administered by any particular route of administration including, but not limited to, intravenous, intramuscular, intraarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intraos seous, intrapelvic, intrapericardial, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, oral, parenteral, subcutaneous, vaginal, rectal, buccal, sublingual, intranasal, iontophoretic means, or transdermal means. Most suitable routes are intravenous injection or oral administration. In particular embodiments, the compositions are administered at or near the target area, e.g., intratumoral injection. For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, intratumoral, subcutaneous, and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage could be dissolved in isotonic NaCl solution and either added to hypodermoclysis fluid or injected at the proposed site of infusion. (See, for example, Remington's Pharmaceutical Sciences, 1990). Some variation in dosage necessarily occurs depending on the condition of the subject. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. The IRC-containing compositions described herein, in some embodiments, are administered by inhalation. In certain embodiments a composition is administered as an aerosol. As used herein the term “aerosol” or “aerosolized composition” refers to a suspension of solid or liquid particles in a gas. These terms are used generally to refer to a composition that has been vaporized, nebulized, or otherwise converted from a solid or liquid form to an inhalable form including suspended solid or liquid drug particles. Such aerosols can be used to deliver a composition via the respiratory system. As used herein, “respiratory system” refers to the system of organs in the body responsible for the intake of oxygen and the expiration of carbon dioxide. The system generally includes all the air passages from the nose to the pulmonary alveoli. In mammals it is generally considered to include the lungs, bronchi, bronchioles, trachea, nasal passages, and diaphragm. For purposes of the present disclosure, delivery of a composition to the respiratory system indicates that a drug is delivered to one or more of the air passages of the respiratory system, in particular to the lungs. Additional formulations that are suitable for other modes of administration include suppositories (for anal or vaginal application) and, in some cases, oral formulations. For suppositories, traditional binders and carriers may include, for example, polyalkalene glycols or triglycerides: such suppositories may be formed from mixtures containing the active ingredient in the range of about 0.5% to about 10%, preferably about 1% to about 2%. Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations, or powders and contain about 10% to about 95% of active ingredient, preferably about 25% to about 70%. The IRC compositions described herein are, in some instances, formulated into a vaccine as neutral or salt forms. Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the peptide) and those that are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like. The pharmaceutical compositions of the present disclosure also include, in certain embodiments, an effective amount of an additional adjuvant. As noted herein, papillomavirus capsid backbones have adjuvant properties. Suitable additional adjuvants include, but are not limited to, Freund's complete or incomplete, mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinitrophenol, and potentially useful human adjuvants such as Bacille Calmette-Guerin (BCG),Corynebacterium parvum, and non-toxic cholera toxin. Under ordinary conditions of storage and use, the described IRC compositions in some embodiments also contain a preservative to prevent the growth of microorganisms. In all cases the pharmaceutical form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier is in some embodiments a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity is maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms is brought about in some instances by incorporation of various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions is achieved by the addition to the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the IRCs in the required amount in the appropriate solvent with various ingredients enumerated above, as required may be followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques, which yield a powder of the active ingredient, plus any additional desired ingredient from a previously sterile-filtered solution thereof. Different aspects of the present disclosure involve administering an effective amount of a composition comprising the IRCs to a subject in need thereof. In some embodiments of the present disclosure, an IRC comprising a target peptide comprising a CD8+ T cell epitope is administered to the patient to treat a tumor or prevent the recurrence of such tumor. Such compositions will generally be dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. Design of IRC for Specific Uses In various embodiments, a method for providing an IRC to a subject in need thereof is provided comprising: (1) measuring the preexisting immunity in a subject, and (2) selecting the appropriate IRC for administration of a subject in need. The appropriate IRC to administer to the subject will depend upon the patients T cell profile. The appropriate IRC will be one that is capable of eliciting a T cell response that is at least twice the baseline total of CD8+ cells. In various embodiments, the appropriate IRC will be one that is capable of eliciting a T cell response that is twice the baseline total of CD8+ or total CD8+CD69+ T cells. The goal is to choose the appropriate IRC based on the subject's vaccination history or prior exposure to a pathogen. Determining which IRC is appropriate is, for example, achieved through: (1) subject interviews; (2) review of a subject's medical records; and/or (3) assessing the subject's T cell profile. In various embodiments, more than one peptide is suitable for eliciting an immune response directed at a tumor. In various embodiments, an IRC carrying either peptide or a mixture of both peptides will be appropriate. In various embodiments, more than one peptide is expressed and bound to the capsid backbone. In various embodiments, a single peptide will comprise more than one peptide. In various embodiments, multiple peptides comprising different peptides will be conjugated to the capsid backbone. In various embodiments, the invention comprises a population of IRCs as described herein and a pharmaceutically acceptable excipient. In various embodiments, the IRCs administered to the subject are identical. In various embodiments, IRCs carrying different peptide(s) are administered to a subject. Selection Based on Prior Vaccination. In various embodiments of the methods and uses described herein, contemplated is also a method of selecting an appropriate IRC to administer to a subject in need thereof. In various embodiments this involves ascertaining if the subject has been actively vaccinated against a given pathogen, e.g., a parasite, a bacterium, or virus, e.g., measles or polio, and then selecting and administering to the subject an IRC as disclosed herein wherein the CD8+ T cell epitope of the peptide is from the pathogen against which the subject has been immunized in the past. In various embodiments, a subject's vaccination history is obtained by reviewing the subject's medical record. In various embodiments, a subject's vaccination history is obtained by interviewing the subject. Selection Based on Prior Infection. In various embodiments, the method of selecting an appropriate IRC for administration to a subject in need thereof involves ascertaining if a subject has been previously infected with a given pathogen, e.g., a parasite, a bacterium, or virus, e.g., measles or polio, and resolved the infection. In various embodiments, the subject is then administered an IRC comprising a peptide which comprises said pathogen for which the subject has been previously infected. One may ascertain if a subject has been infected with a particular pathogen by reviewing the subject's medical records or interviewing the subject. Non-limiting examples of CD8+ T cell epitopes that bind to particular MHC class I molecules are set forth in Table 1. The method also comprises, in certain embodiments, determining which MHC class I determinant(s) the subject's cells express and then administering an IRC described herein wherein the CD8+ T cell epitope of the peptide is a CD8+ T cell epitope of the antigenic component of the pathogen in the vaccine or of the pathogen that previously infected the subject that forms a complex with the subject's MHC class I determinant(s). Measuring T cell Responses. In various embodiments, a subject's T cell profile is also assessed in order to select an appropriate IRC using various techniques known in the art. This profile is then used to guide selection of the appropriate IRC to administer to the subject. Such techniques include, for example, measuring interferon-γ levels, using flow cytometry to isolate Ag-specific CD8+ T cells, and/or cytotoxicity assays. To measure interferon-γ (a marker of T cell activation), intracellular staining of isolated T cells. Alternatively, an enzyme-linked immunosorbent spot (ELISPOT) assay for interferon-γ may be conducted. This technique allows for a high throughput assessment of a patient's T cell profile. This method can potentially detect one in 100,000-300,000 cells. Briefly, a monoclonal antibody for a specific cytokine is pre-coated onto a polyvinylidene difluoride (PVDF)-backed microplate. CD8+ T cells are pipetted into the wells along with dendritic cells and individual peptides and the microplate is placed into a humidified 37° C. CO2incubator for a period ranging from 24 to 48 h. During incubation, the immobilized antibody binds the cytokine secreted from the cells. After washing a detection antibody specific for the chosen analyte is added to the wells. Following the washes, enzyme conjugated to streptavidin is added and a substrate is added. A colored precipitate forms, according to the substrate utilized and appears as spot at the sites of cytokine secretion, with each individual spot representing a single producing cell. In various embodiments, provided are methods of determining the appropriate IRC to administer to a subject in need thereof, by assessing the subject's T cell profile, comprising: (1) collecting PBMCs from subject (pre-vaccination sample), (2) preparing enzyme-linked immune absorbent spot (ELISPot) plates by coating with anti-IFN-γ antibody (incubate overnight), (3) incubating PBMCs with one of the pool of peptides of interest, i.e., the peptides expected to elicit a T cell response (incubate for 1-2 days), (4) washing the plates, adding a biotinylated secondary antibody (incubating for a few hours), (5) washing the plates, adding avidin conjugated horseradish peroxidase and incubating, (6) washing plates, adding aminoethyl carbazole (AEC) for a few minutes, (7) stopping the reaction (by adding water), and (8) visualizing on an ELISPot reader. The disclosed methods detect up to one in 100,000 to 300,000 cells. A two-fold increase in the frequency of antigen-specific T cells should be considered as a signal. In various embodiments T cell proliferation is measured by 3H (tritiated)-thymidine. Such methods are sensitive and can be used for high throughput assays. Such techniques include, for instance, carboxyfluorescein succinimidyl ester (CFSE) and Ki64 intracellular staining. Selecting Peptides based on Tropism. It is known in the art that some viruses display a tropism for particular type of tissue. For example: viruses that display a tropism for brain tissue include without limitation, JC virus, measles, LCM virus, arbovirus and rabies; viruses that display a tropism for eye tissue include without limitation herpes simplex virus, adenovirus, and cytomegalovirus; viruses that display a tropism for nasal tissue include without limitation, rhinoviruses, parainfluenza viruses, and respiratory syncytial virus; viruses that display a tropism for oral tissue, e.g., oral mucosa, gingiva, salivary glands, pharynx, include without limitation, herpes simplex virus type I and type II, mumps virus, Epstein Barr virus, and cytomegalovirus; viruses that display a tropism for lung tissue include without limitation, influenza virus type A and type B, parainfluenza virus, respiratory syncytial virus, adenovirus, and SARS coronavirus; viruses that display a tropism for nerve tissue, e.g., the spinal cord, include without limitation poliovirus and HTLV-1; viruses that display a tropism for heart tissue, include without limitation, Coxsackie B virus; viruses that display a tropism for liver tissue, include without limitation, hepatitis viruses types A, B, and C; viruses that display a tropism for gastrointestinal tissue, e.g., stomach, and large and small intestine, include without limitation, adenovirus, rotavirus, norovirus, astrovirus, and coronavirus; viruses that display a tropism for pancreatic tissue, include without limitation, coxsackie B virus; viruses that display a tropism for skin tissue, include without limitation, varicella zoster virus, herpes simplex virus 6, smallpox virus, molluscum contagiosum, papilloma viruses, parvovirus B19, rubella, measles and coxsackie A virus; and viruses that display a tropism for genital tissue, include without limitation, herpes simplex type 2, papillomaviruses, human immunodeficiency virus (HIV). In various embodiments, a method for treating a cancer in a subject in need thereof is provided by administering an IRC described herein to the subject wherein the peptide is a CD8+ epitope of a pathogen that has a tropism for the tissue that is the source of the cancer (the “source tissue”). In various embodiments, the appropriate IRC is selected by first determining the source tissue of the tumor cell and then selecting a peptide: (1) to which the patient already has existing CD8+ T cells, and (2) that has a tropism for the source tissue of the tumor. The selected IRC(s) are then administered to the subject in need thereof. In various embodiments, provided are methods for treating a lung cancer comprising determining if a subject has been actively vaccinated against a pathogen that infects lung cells, e.g., an influenza virus, e.g., influenza virus type A or type B, then administering an effective amount of an IRC composition described herein, wherein the CD8+ T cell epitope of the peptide is of the antigenic determinants of the pathogen contained in the vaccine and which T cell epitope forms a complex with an MHC molecule class I of the subject. The methods and uses described herein for treating a lung cancer includes, in some embodiments, determining if a subject has been infected with pathogen that infects lung cells, e.g., an influenza virus, e.g., influenza virus type A or type B, then administering an effective amount of an IRC composition described herein wherein the CD8+ T cell epitope of the peptide is of that pathogen and which T cell epitope forms a complex with an MHC class I molecule of the subject. Provided also are methods for treating an oral cancer, which are part of the group of cancers commonly referred to as head and neck cancers, by administering an IRC compositions described herein, wherein the CD8+ epitope of the peptide is of a pathogen that has a tropism for oral tissue, e.g., a mumps virus, Epstein Barr virus, cytomegalovirus, or a herpes simplex virus type 1. The method comprises determining if a subject in need thereof has been actively vaccinated against, or infected with, e.g., a mumps virus, Epstein Barr virus, cytomegalovirus, or a herpes simplex virus type 1, and if the subject has been vaccinated or infected previously then administering to the subject an IRC composition described herein wherein the CD8+ epitope of the peptide is of a mumps virus or a measles virus or of the antigenic component of the vaccine the subject had received, or of the pathogen, i.e., mumps, measles, Epstein Barr virus, cytomegalovirus, or a herpes simplex virus type 1, that had previously infected the subject. Combination Therapy In various embodiments, the IRC compositions described herein are co-administered with other cancer therapeutics. Furthermore, in some embodiments, the IRCs described herein are administered in conjunction with other cancer treatment therapies, e.g., radiotherapy, chemotherapy, surgery, and/or immunotherapy. In some aspects of methods and uses described herein, the IRC compositions described herein are administered in conjunction with checkpoint inhibitors. In various embodiments the capsid backbone is administered in conjunction with an immune agonist. In various embodiments, the IRC is administered in conjunction with treatment with a therapeutic vaccine. In various embodiments, the IRC is administered in conjunction with treatment with a conjugated antigen receptor expressing T cell (CAR-T cell). In various embodiments, the IRC is administered in conjunction with treatment with another immuno-oncology product. The IRCs of the present disclosure and other therapies or therapeutic agents are, in some embodiments, administered simultaneously or sequentially by the same or different routes of administration. The determination of the identity and amount of therapeutic agent(s) for use in the methods of the present disclosure is readily made by ordinarily skilled medical practitioners using standard techniques known in the art. All of the references cited above, as well as all references cited herein, are incorporated herein by reference in their entireties for all purposes. While the methods, uses, and compositions described herein have been illustrated and described in detail in above, such illustration and description are to be considered illustrative or exemplary and not restrictive. It will be understood that changes and modifications may be made by those of ordinary skill within the scope and spirit of the following claims. In particular, the present disclosure covers further embodiments with any combination of features from different embodiments described above and below. The present disclosure is additionally described by way of the following illustrative non-limiting examples that provide a better understanding of the present disclosure and of its many advantages. The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques used in the present disclosure to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the present disclosure. EXAMPLES Example 1 Production of Truncated Mouse Papillomavirus (MPV1) L1 Protein The truncated mouse papillomavirus L1 DNA sequence of 1138 base pairs was codon-optimized forE. coliexpression and synthesized (SEQ ID NOS:135 and 136, two varieties of codon optimization) (GeneScript Biotech, Piscataway, NJ) and subsequently cloned into the T7 expression vector Pet-24a(+) (MilliporeSigma, Burlington, MA). The sequence was based on the wild type mouse (Mus musculus) papillomavirus L1 protein sequence except that it contains three deletion mutations at three specific regions: one deletion at the amino-terminus (10 amino acids removed), one at deletion the carboxy-terminus (34 amino acids deleted), and a third deletion in the helix four (H4) region close to the carboxy-terminal region (deletion of amino acids 411 to 436 of the MPV L1 sequence). This mutant MPV L1 protein is hereinafter referred to as “MPV.10.34.d.” (See,FIG.1B). The wild type mouse (Mus musculus) L1 wild type protein sequence is depicted inFIG.1Aand has the following protein sequence (SEQ ID NO: 132, NCBI Reference Sequence: YP_003778198.1, DNA: 9434943): Met Ala Met Trp Thr Pro Gln Thr Gly Lys Leu Tyr Leu Pro ProThr Thr Pro Val Ala Lys Val Gln Ser Thr Asp Glu Tyr Val TyrPro Thr Ser Leu Phe Cys His Ala His Thr Asp Arg Leu Leu ThrVal Gly His Pro Phe Phe Ser Val Ile Asp Asn Asp Lys Val ThrVal Pro Lys Val Ser Gly Asn Gln Tyr Arg Val Phe Arg Leu LysPhe Pro Asp Pro Asn Lys Phe Ala Leu Pro Gln Lys Asp Phe TyrAsp Pro Glu Lys Glu Arg Leu Val Trp Arg Leu Arg Gly Leu GluIle Gly Arg Gly Gly Pro Leu Gly Ile Gly Thr Thr Gly His ProLeu Phe Asn Lys Leu Gly Asp Thr Glu Asn Pro Asn Lys Tyr GlnGln Gly Ser Lys Asp Asn Arg Gln Asn Thr Ser Met Asp Pro LysGln Thr Gln Leu Phe Ile Val Gly Cys Glu Pro Pro Thr Gly GluHis Trp Asp Val Ala Lys Pro Cys Gly Ala Leu Glu Lys Gly AspCys Pro Pro Ile Gln Leu Val Asn Ser Val Ile Glu Asp Gly AspMet Cys Asp Ile Gly Phe Gly Asn Met Asn Phe Lys Glu Leu GlnGln Asp Arg Ser Gly Val Pro Leu Asp Ile Val Ser Thr Arg CysLys Trp Pro Asp Phe Leu Lys Met Thr Asn Glu Ala Tyr Gly AspLys Met Phe Phe Phe Gly Arg Arg Glu Gln Val Tyr Ala Arg HisPhe Phe Thr Arg Asn Gly Ser Val Gly Glu Pro Ile Pro Asn SerVal Ser Pro Ser Asp Phe Tyr Tyr Ala Pro Asp Ser Thr Gln AspGln Lys Thr Leu Ala Pro Ser Val Tyr Phe Gly Thr Pro Ser GlySer Leu Val Ser Ser Asp Gly Gln Leu Phe Asn Arg Pro Phe TrpLeu Gln Arg Ala Gln Gly Asn Asn Asn Gly Val Cys Trp His AsnGlu Leu Phe Val Thr Val Val Asp Asn ThrArg Asn Thr Asn Phe Thr Ile Ser Gln Gln Thr Asn Thr Pro AsnPro Asp Thr Tyr Asp Ser Thr Asn Phe Lys Asn Tyr Leu Arg HisVal Glu Gln Phe Glu Leu Ser Leu Ile Ala Gln Leu Cys Lys ValPro Leu Asp Pro Gly Val Leu Ala His Ile Asn Thr Met Asn ProThr Ile Leu Glu Asn Trp Asn Leu Gly Phe Val Pro Pro Pro GlnGln Ser Ile Ser Asp Asp Tyr Arg Tyr Ile Thr Ser Ser Ala ThrArg Cys Pro Asp Gln Asn Pro Pro Lys Likewise, the wild type nucleic acid sequence for MPV1 L1 protein (SEQ ID NO: 133) is as follows: ATGGCAATGTGGACACCCCAGACCGGGAAGCTTTACCTCCCACCTACAACTCCAGTGGCAAAAGTGCAGAGCACAGACGAATATGTGTACCCTACGTCTCTCTTCTGTCATGCACACACGGACCGTTTGCTAACAGTGGGCCACCCTTTTTTTTCTGTCATTGACAATGACAAGGTCACTGTGCCTAAAGTGTCTGGCAACCAATATAGGGTTTTCAGACTTAAATTCCCAGATCCAAATAAATTTGCATTGCCCCAAAAGGATTTCTATGATCCTGAGAAAGAACGGTTAGTGTGGAGGTTAAGGGGTCTGGAAATTGGAAGAGGTGGCCCATTAGGGATTGGCACTACCGGGCACCCCCTTTTTAACAAGCTTGGAGACACGGAAAATCCAAATAAATATCAGCAAGGCTCTAAGGATAATAGGCAGAACACTTCCATGGACCCCAAACAAACACAGCTGTTTATTGTTGGCTGTGAACCCCCTACAGGGGAACACTGGGATGTAGCTAAGCCCTGTGGAGCTCTGGAGAAGGGTGACTGCCCTCCTATCCAACTTGTAAATAGTGTAATTGAGGATGGGGATATGTGTGACATTGGCTTTGGGAATATGAACTTCAAAGAGCTGCAGCAGGATAGGAGTGGTGTGCCTCTTGATATTGTATCTACCCGGTGCAAATGGCCCGACTTTCTGAAAATGACCAATGAGGCATATGGGGATAAGATGTTCTTCTTTGGAAGGAGAGAGCAAGTGTATGCAAGACACTTTTTCACCAGGAATGGCTCTGTGGGGGAGCCCATACCAAACTCTGTGAGTCCCAGTGACTTTTACTACGCACCCGACAGCACACAGGACCAGAAGACACTCGCACCCTCCGTGTACTTTGGAACTCCTAGTGGGTCACTTGTGTCGAGTGATGGTCAGCTGTTTAACAGGCCATTTTGGCTTCAAAGGGCTCAGGGAAACAATAATGGTGTGTGCTGGCACAATGAGCTCTTTGTTACTGTTGTCGACAACACAAGGAATACAAACTTTACTATCTCCCAGCAAACCAACACACCAAACCCAGATACATATGACTCTACTAATTTTAAAAACTATTTAAGACATGTGGAACAATTTGAGCTGTCCCTTATTGCTCAACTGTGTAAGGTTCCACTTGACCCGGGTGTGCTTGCCCATATAAACACTATGAACCCAACCATCTTGGAGAACTGGAACTTGGGTTTTGTACCTCCCCCACAGCAGTCCATCTCTGATGACTATAGGTATATAACATCATCGGCAACTCGCTGTCCAGATCAGAATCCGCCCAAGGAAAGAGAGGATCCTTACAAGGGTCTTATATTTTGGGAAGTTGATCTTACTGAGAGGTTTTCTCAGGACCTTGATCAGTTTGCTCTGGGACGAAAGTTTCTGTATCAAGCTGGTATACGTACTGCTGTTACGGGCCGCGGGGTCAAAAGGGCAGCGTCTACAACCTCTGCGTCTTCTAGACGAGTTGTAAAACGGAAGAGGGGAAGCAAATAA In contrast, the mutant MPV sequence selected for the following studies is depicted inFIG.1Band has the following amino acid sequence (SEQ ID NO:134): Met Leu Tyr Leu Pro Pro Thr Thr Pro Val Ala Lys Val Gln SerThr Asp Glu Tyr Val Tyr Pro Thr Ser Leu Phe Cys His Ala HisThr Asp Arg Leu Leu Thr Val Gly His Pro Phe Phe Ser Val IleAsp Asn Asp Lys Val Thr Val Pro Lys Val Ser Gly Asn Gln TyrArg Val Phe Arg Leu Lys Phe Pro Asp Pro Asn Lys Phe Ala LeuPro Gln Lys Asp PheTyr Asp Pro Glu Lys Glu Arg Leu Val Trp Arg Leu Arg Gly LeuGlu Ile Gly Arg Gly Gly Pro Leu Gly Ile Gly Thr Thr Gly HisPro Leu Phe Asn Lys Leu Gly Asp Thr Glu Asn Pro Asn Lys TyrGln Gln Gly Ser Lys Asp Asn Arg Gln Asn Thr Ser Met Asp ProLys Gln Thr Gln Leu Phe Ile Val Gly Cys Glu Pro Pro Thr GlyGlu His Trp Asp Val Ala Lys Pro Cys Gly Ala Leu Glu Lys GlyAsp Cys Pro Pro Ile Gln Leu Val Asn Ser Val Ile Glu Asp GlyAsp Met Cys Asp Ile Gly Phe Gly Asn Met Asn Phe Lys Glu LeuGln Gln Asp Arg Ser Gly Val Pro Leu Asp Ile Val Ser Thr ArgCys Lys Trp Pro Asp Phe Leu Lys Met Thr Asn Glu Ala Tyr GlyAsp Lys Met Phe Phe Phe Gly Arg Arg Glu Gln Val Tyr Ala ArgHis Phe Phe Thr Arg Asn Gly Ser Val Gly Glu Pro Ile Pro AsnSer Val Ser Pro Ser Asp Phe Tyr Tyr Ala Pro Asp Ser Thr GlnAsp Gln Lys Thr Leu Ala Pro Ser Val Tyr Phe Gly Thr Pro SerGly Ser Leu Val Ser Ser Asp Gly Gln Leu Phe Asn Arg Pro PheTrp Leu Gln Arg Ala Gln Gly Asn Asn Asn Gly Val Cys Trp HisAsn Glu Leu Phe Val Thr Val Val Asp Asn Thr Arg Asn Thr AsnPhe Thr Ile Ser Gln Gln Thr Asn Thr Pro Asn Pro Asp Thr TyrAsp Ser Thr Asn Phe Lys Asn Tyr Leu Arg His Val Glu Gln PheGlu Leu Ser Leu Ile Ala Gln Leu Cys Lys Val Pro Leu Asp ProGly Val Leu Ala His Ile Asn Thr Met Asn Pro Thr Ile Leu GluAsn Trp Asn Leu Gly Phe Val Pro Pro Lys Glu Arg Glu Asp ProTyr Lys Gly Leu Ile Phe Trp Glu Val Asp Leu Thr Glu Arg PheSer Gln Asp Leu Asp Gln Phe Ala Leu Gly Arg Lys Phe Leu TyrGln Alignment of the wild type sequence with the triple truncation MPV.10.34.d sequence is shown inFIG.2. Additionally, the nucleic acid sequence (below) of MPV.10.34.d was optimized for expression. The sequence was optimized for codon usage within the target host as well as for expression level to maximize expression efficiency within the host. Below are provided two alternative optimized nucleic acid sequences for MPV.10.34.d used herein (SEQ ID NO: 135): ATGCTGTACCTGCCGCCGACCACCCCGGTGGCGAAAGTTCAGAGCACCGACGAATACGTTTATCCGACCAGCCTGTTCTGCCACGCGCACACCGATCGTCTGCTGACCGTGGGTCACCCGTTCTTTAGCGTTATCGACAACGATAAGGTGACCGTTCCGAAAGTGAGCGGCAACCAGTACCGTGTTTTTCGTCTGAAGTTCCCGGACCCGAACAAATTTGCGCTGCCGCAAAAGGACTTCTATGATCCGGAGAAGGAACGTCTGGTGTGGCGTCTGCGTGGTCTGGAAATTGGTCGTGGTGGCCCGCTGGGTATTGGTACCACCGGTCACCCGCTGTTCAACAAACTGGGCGATACCGAGAACCCGAACAAATATCAGCAAGGTAGCAAGGACAACCGTCAGAACACCAGCATGGACCCGAAGCAGACCCAACTGTTTATTGTTGGTTGCGAGCCGCCGACCGGTGAACACTGGGATGTTGCGAAACCGTGCGGTGCGCTGGAAAAGGGCGATTGCCCGCCGATCCAACTGGTGAACAGCGTTATTGAGGACGGTGATATGTGCGACATCGGTTTTGGCAACATGAACTTCAAAGAACTGCAGCAAGACCGTAGCGGCGTGCCGCTGGATATTGTTAGCACCCGTTGCAAATGGCCGGACTTCCTGAAGATGACCAACGAAGCGTACGGTGATAAGATGTTCTTTTTCGGCCGTCGTGAGCAGGTTTATGCGCGTCACTTTTTCACCCGTAACGGTAGCGTGGGCGAGCCGATCCCGAACAGCGTTAGCCCGAGCGACTTCTACTATGCGCCGGACAGCACCCAGGATCAAAAAACCCTGGCGCCGAGCGTGTACTTTGGTACCCCGAGCGGCAGCCTGGTTAGCAGCGATGGTCAACTGTTTAACCGTCCGTTCTGGCTGCAGCGTGCGCAGGGTAACAACAACGGCGTGTGCTGGCACAACGAACTGTTTGTTACCGTGGTTGACAACACCCGTAACACCAACTTCACCATCAGCCAGCAAACCAACACCCCGAACCCGGACACCTACGATAGCACCAACTTTAAAAACTATCTGCGTCACGTGGAGCAGTTCGAACTGAGCCTGATTGCGCAACTGTGCAAAGTGCCGCTGGACCCGGGTGTGCTGGCGCACATCAACACCATGAACCCGACCATTCTGGAGAACTGGAACCTGGGTTTCGTTCCGCCGAAAGAGCGTGAAGACCCGTACAAGGGCCTGATCTTCTGGGAAGTGGATCTGACCGAACGTTTCAGCCAGGACCTGGATCAATTTGCGCTGGGCCGTAAATTCCTG TATCAGTAAAnd (SEQ ID NO: 136):GAATTGGCGGAAGGCCGTCAAGGCCACGTGTCTTGTCCGCGGTACCCATATGCTGTATCTGCCTCCAACTACACCGGTTGCAAAAGTTCAGAGCACCGATGAATATGTTTATCCGACCAGCCTGTTTTGTCATGCACATACCGATCGTCTGCTGACCGTTGGTCATCCGTTTTTTAGCGTTATTGATAACGATAAAGTGACCGTTCCGAAAGTTAGCGGTAATCAGTATCGTGTTTTTCGCCTGAAATTTCCGGATCCGAACAAATTTGCACTGCCGCAGAAAGATTTTTACGACCCGGAAAAAGAACGTCTGGTTTGGCGTCTGCGTGGTCTGGAAATTGGTCGTGGTGGTCCGTTAGGTATTGGCACCACCGGTCATCCGCTGTTTAACAAACTGGGTGATACCGAAAATCCGAATAAATACCAGCAGGGCAGCAAAGATAATCGTCAGAATACCAGTATGGATCCGAAACAGACCCAGCTGTTTATTGTTGGTTGTGAACCGCCTACCGGTGAACATTGGGATGTTGCAAAACCGTGTGGTGCACTGGAAAAAGGTGATTGTCCGCCTATTCAGCTGGTTAATAGCGTGATTGAAGATGGTGATATGTGCGATATTGGCTTTGGCAACATGAACTTTAAAGAACTGCAGCAGGATCGTAGCGGTGTTCCGCTGGATATTGTTAGCACCCGTTGTAAATGGCCTGATTTTCTGAAAATGACCAATGAAGCCTATGGCGACAAAATGTTTTTTTTCGGTCGTCGTGAACAGGTTTATGCCCGTCACTTTTTTACCCGTAATGGTAGCGTTGGTGAACCGATTCCGAATAGCGTTAGCCCGAGCGATTTCTATTATGCACCGGATAGCACCCAGGATCAGAAAACCCTGGCACCGAGCGTTTATTTTGGCACCCCGAGCGGTAGCCTGGTTAGCAGTGATGGTCAGCTGTTCAATCGTCCGTTTTGGCTGCAGCGTGCACAGGGTAATAACAATGGTGTTTGTTGGCATAACGAACTGTTTGTTACCGTTGTTGATAATACCCGCAATACCAACTTTACCATTAGCCAGCAGACCAATACACCGAATCCGGATACCTATGATAGCACCAACTTCAAAAACTATCTGCGTCATGTGGAACAGTTTGAACTGAGCCTGATTGCCCAGCTGTGTAAAGTGCCGCTGGATCCGGGTGTTCTGGCACATATTAACACCATGAATCCGACCATTCTGGAAAATTGGAATCTGGGTTTTGTTCCGCCTAAAGAACGTGAAGATCCGTATAAAGGTCTGATTTTTTGGGAAGTTGATCTGACCGAACGTTTTAGCCAGGATCTGGATCAGTTTGCACTGGGTCGCAAATTTCTGTATCAGTAACTCGAGGAGCTCGGAGCACAAGACTGGCCTCATGGGCCTTCCGCTCACTGCC The general protocol for recombinant expression and purification of the mutant MPV.10.34.d is schematically depicted inFIG.3. The MPV.10.34.d nucleic acid sequence was generated from wild type mouse papillomavirus sequence via site-mutagenesis (Genscript Biotech, Piscataway, N.J.) using the following primer sequence (SEQ ID NO: 137): AAGCTTGTCGACGGAGCTCGAATTCGGATCCTTATTACTGATACAGGAATTTACGGCCCAGC The MPV.10.34.d nucleic acid sequence was then cloned into the multicloning site of expression vector pet24a(+) (MilliporeSigma, Burlington, MA) using restriction endonucleases Ndel and BamH1 according to standard protocols. The correct cloning into the multiple cloning site and construct sequence was confirmed by both restriction endonuclease enzyme digestion using MIu1 and BamH1 as well as Sanger sequencing using both T7 forward and reverse primers. Expression was achieved by transforming the pet24a(+) plasmid containing MPV.10.34.d into T7 expression competentEscherichia coli2566 cells (New England Biolabs, Ipswich, MA, US), and colony selection on solid media. A single colony was grown according to standard protocols in Lurea broth (LB) media. Briefly, 5 mL sterile LB including 50 μg/mL kanamycin (Quality Biological, Gaithersburg, MD, US) was seeded with a single colony selected from the solid media and grown overnight at 37° C. with shaking. The seed culture was then diluted 1:25 and growth was continued at 37° C. until OD600 reached about 0.6 to 0.8. Then about 1 mM final concentration of isopropyl β-d-1-thiogalactopyranoside (IPTG, Invitrogen, Carlsbad, CA, US) was added to the culture to induce expression from the plasmid. Induction was continued under these conditions for an additional four hours after which cell pellets are collected by centrifugation at 4000×g for 15 minutes at 4° C. The supernatant was discarded and the cell pellets were stored at −20° C. unless immediately used. MPV.10.34.d was expressed as inclusion bodies (IBs). To recover IB MPV.10.34.d, pellets were first thawed (if frozen) and then resuspended in 20 mL per 1 L pellet lysis buffer (50 mM Tris, pH 8.0, 500 mM NaCl, 1 mM EDTA, 1 mM protease inhibitor phenylmethylsulfonyl fluoride (PMSF). Resuspended material was then homogenized using a high pressured homogenizer (Avestin Emulsiflex C3™, ATA Scientific, Taren Point, Australia) and cells were passed through the homogenizer and lysed 4 times at about 15,000 to 20,000 PSI. The lysed bacterial cells were then centrifuged at 25,000×g at 4° C. for 20 min. Supernatant was then discarded and the inclusion body pellet was stored at −20° C. Next the IB were solubilized by resuspending the pellet (50 mL per 1 L pellet) in of 6 M urea buffer (8 M Urea, 50 mM Tris, pH 8.0, 500 mM NaCl, 1 mM EDTA, 1 mM PMSF, and 1 mM DTT). Resuspended contents were once more passaged three to four times through the homogenizer (Avestin Emulsiflex C3™, ATA Scientific, Taren Point, Australia) at about 15,000 to 20,000 PSI. The resolubilized samples were centrifuged at 25,000×g at 4° C. for 20 min. The supernatant was collected into a container that is sufficiently large enough to hold the volume of a sample. The pellet was discarded. The supernatant was stored at 4° C. or −20° C. Following solubilization, the MPV.10.34.d was refolded by removal of the denaturant (6M Urea) in a step-gradient manner. The solubilized samples were inserted into dialysis tubing (snakeskin dialysis tubing, 10,000 Da molecular weight cut off, 35 mm. (ThermoFisher Scientific, Waltham, MA, US). In general, about 100 to about 150 mL of resolubilized sample solution was dispensed into a single dialysis tube. The samples were first dialyzed (sample to buffer ratio 1:12.5) against 4 M urea buffer (50 mM Tris, pH 8.0, 500 mM NaCl, 1 mM EDTA, 1 mM PMSF, 1 mM DTT, and 0.05% Tween®-80) for 3±1 hour in a cold room at about 4° C. on a stir plate. Then, the samples were again dialyzed against a fresh 1 M urea buffer (50 mM Tris, pH 8.0, 500 mM NaCl, 1 mM EDTA, 1 mM PMSF, 1 mM DTT, and 0.05% Tween-80) for 3±1 hour in a cold room on a stir plate. Subsequently, the samples were dialyzed against 0 M urea buffer (50 mM Tris, pH 8.0, 500 mM NaCl, 1 mM EDTA, 1 mM PMSF, 1 mM DTT, and 0.05% Tween-80) overnight (about 16 to 18 hours) in a cold room at about 4° C. on a stir plate. The dialyzed/refolded sample solutions were aliquoted into 50 mL conical tubes and stored in a −20° C. freezer. To obtain a MPV.10.34.d of greater than 95% purity for subsequent medicinal use, samples were subjected to a two-step chromatography purification which involves a capture step utilizing cation exchange chromatography (CEX) followed by a polishing step using a hydrophobic interaction column (HIC). For the capture step, the refolded MPV.10.34.d samples were removed from the −20 and thawed on ice. Next, the sample was dialyzed into capture buffer A (25 mM NaPO4, 25 mM NaCl, pH 6.0). Following dialysis, samples were centrifuged 4000×g, for about 10 min, at 4° C. and then filtered through a 0.22 μm polyethersulfone (PES) membrane. The refolded MPV.10.34.d protein was then captured by CEX (Fractogel® EMD S03-M, EMD Millipore, Burlington, MA, US) and then step eluted with 30% 25 mM NaPO4, 1.5 M NaCl, pH 6.0. This resulted in purified refolded MPV.10.34.d of purity of at least 80%. To further remove contaminants and increase purity of the MPV.10.34.d to above 95%, the CEX eluate was diluted with high-salt buffer to achieve loading conditions of 25 mM NaPO4, 3 M NaCl, pH 6.0, and applied to HIC resin (butyl-S-Sepharose® Fast Flow, GE Healthcare Life Sciences/Fisher Scientific, Waltham, MA, US). The bound refolded MPV.10.34.d product was subjected to a pre-elution wash with 30% 25 mM NaPO4, 25 mM NaCl, pH 6.0, and then eluted with a single step gradient of 70% 25 mM NaPO4, 25 mM NaCl, pH 6.0. Greater than 95% purity MPV.10.34.d was stored in a −20° C. freezer in the elution buffer. Greater than 95% purity MPV.10.34.d was confirmed via SDS-PAGE followed by Coomassie blue gel and silver staining. For Coomassie staining, gels were incubated in water to remove SDS-PAGE running buffer, then incubated for 5 minutes in SimplyBlue SafeStain (Novex, Carlsbad, CA). Gels were de-stained in water. (See photographs of gels inFIG.4A). Silver staining was performed using a Pierce Silver stain kit (ThermoFisher Scientific, Rockford, IL) according to manufacturer's instructions. (See,FIG.4B). To estimate purity, the images of the gels were taken using the Bio-Rad Image Lab 6.01 software. Gel images were then uploaded into the software and the entire vertical lane containing the band of interest (“lane profile”) was analyzed using the image analysis software. The specific total density of the band of the protein of interest was calculated by drawing a box or freehand shape around the band. Subsequently, the total density of the entire lane was measured in the same manner. After obtaining the measurements, the background density of a suitably matched area on the gel in each case was subtracted. This background-corrected density of the protein band by the background-corrected density of the whole lane was then multiplied by 100 to obtain the percent purity. From this analysis and as seen in bothFIGS.4A and4B, the main process steps described above provided incremental purification of the ˜50 kDA MPV.10.34.d protein. Non-specific proteins above and below the 50 kDA band were significantly reduced across each of the purification steps, where lane 1 was a post-cell harvest sample, wash, and homogenization; lane 2 was a post-IB solubilization sample, lane 3 was a post-refolding sample, lane 4 was a post-capture chromatography via CEX sample, and lane 5 was a post-polishing step sample using HIC. Example 2 Determination of MPV.10.34.d Structure and Size DLS (dynamic light scattering) and TEM revealed that upon refolding MPV.10.34.d unexpectedly forms capsid backbones that are about 20 nm to 30 nm in diameter. (See,FIG.5). To analyze the refolded MPV.10.34.d samples, the purified samples were first analyzed by DLS to obtain determine whether refolding of MPV.10.34.d occurred. 60 μl of sample was placed in a 40 μL solvent-resistant micro-cuvette (ZEN0040, Malvern Panalytical, Waltham, MA) and the cell was subsequently placed into a Zetasizer Nano ZS Dynamic Light scattering instrument (Malvern Panalytical, Waltham, MA). This was a research-grade dynamic light scattering system for measurement of protein size, electrophoretic mobility of proteins, zeta potential of colloids and nanoparticles, and optionally the measurement of protein mobility, and microrheology of protein and polymer solutions. The high performance of the Zetasizer Nano ZS also enables the measurement of the molecular weight and second virial coefficient, A2, of macromolecules and kD, the DLS interaction parameter. The system can also be used in a flow configuration to operate as a size detector for SEC or FFF. Once in the machine, the sample was processed with the companion software (Zetasizer Nano Software, Malvern Panalytical). The program was set to read the sample for a total of 5 runs to generate two plots. The two plots generated were determine capsid backbone size and structure, intensity (FIG.6A), and volume (FIG.6B). The intensity distribution provides the amount of light scattered by the particles in the different sized bins. The volume distribution demonstrates the total volume of particles in various sized bins. In other words, the intensity plot (FIG.6A) provides an assessment of the overall population sizes of MPV.10.34.d particles including host cell contaminants within the sample, whereas the volume plot (FIG.6B) determines the relative proportion of refolded protein with respect to other contaminants, i.e., host bacterial cell proteins. Note that each individual plot line in the graphs ofFIGS.6A and6Brepresent individual samples (five samples for each of A and B). If the volume curve was between about 10 to 15 nm (X-axis), the shell was determined to be a capsomer made up of 5 MPV.10.34.d units. For T=1 icosahedral capsid backbones made up of 60 MPV.10.34.d, the volume curve plot is about 20 to 30 nm. For T=7 icosahedral capsid backbones made up of 360 MPV.10.34.d, the volume curve plot was about 50 to 60 nm. As is readily apparent from the intensity graph (FIG.6A), there are two peaks, one lower at approximately 20 to 30 nm and one at approximately >100 nm, whereas the volume graph (FIG.6B) shows a single size of about 20 nm to 30 nm. It was postulated that the two peaks on the intensity figure were attributable to the presence of distinct populations of capsid backbones that arose after refolding of the MPV.10.34.d into the capsid backbone since the samples at this stage had not undergone purification. However, the larger of the two peaks shown in the intensity plot constitute only a small portion of the sample, and a majority of the sample falls within the smaller peak. Hence, the DLS results show that a majority of the MPV.10.34.d are structures that are about 20 nm to 30 nm in size. In DLS, information regarding the motion (diffusion) of submicron particles in a solution is extracted from the rate of scattering intensity fluctuations using a statistical technique called intensity autocorrelation. The mean particle size and distribution are calculated from the distribution of diffusion coefficients using the Stokes Einstein equation. Because the magnitude of the scattering intensity varies roughly with the 6thpower of the particle size, DLS is highly sensitive to the presence of small amounts of aggregates in a mixture of capsid backbones which is believed to be reflected in the intensity plot shown inFIG.6A. TEM analysis was also employed to obtain further visual confirmation of the structure and size of the refolded proteins. Samples (10 μL) were adsorbed to glow discharged (EMS GloQube) carbon coated 400 mesh copper grids (EMS), by floatation for 2 min. Grids were quickly blotted and then rinsed in 3 drops (1 min each) of TBS. Grids were negatively stained in 2 consecutive drops of 1% uranyl acetate with tylose (1% UAT, double filtered, 0.22 μm filter), blotted then quickly aspirated to obtain a thin layer of stain covering the sample. Grids were imaged on a Phillips CM-120 TEM operating at 80 kV with an AMT XR80 CCD (8 megapixel). TEM results revealed that MPV.10.34.d formed capsid backbones that had a markedly grooved appearance, with pentagonal/capsomer “towers.” (See,FIG.5). Measurements showed that these capsid backbones were approximate 30 nm is size (compare with the 500 nm scale on TEM micrograph,FIG.5). These results were unexpected because deletion of residues in the helix four H4 region of L1 has been reported to not lead to T=1 geometry capsid backbone formation. Further, it has been shown that the same deletions result in capsomers of T=7 in HPV11 and HPV16 L1 proteins. (See, Chen et al.,Mol. Cell,5:557-567, 2000, WO 2000054730, Bishop et al.,Virol. J.,4:3, 2007, and Schädlich et al.,J. Virol.,83(15):7690-7705, 2009). In summary, these findings support the conclusion that the MPV.10.34.d constructs form icosahedral capsid backbones of T=1 lattice geometry comprised of 60 monomers, or 12 capsomers. Example 3 MPV.10.34.d Capsid Backbone Assembly Requires Reductant HPV particles form T=7 geometry particles that are 50 to 60 nm is diameter. The manufacture and production of such capsid backbones in either eukaryotic or prokaryotic host cell systems involves the expression in suitable host cell system followed by a series of purification step to yield highly purified capsid backbones. These capsid backbones are then subjected to a disassembly and re-assembly (DARA) step. Briefly, this involves the addition of DTT or a similar reducing agent to disassemble the capsid backbones into capsomers/pentamers (made from five L1 monomers) and then removal of the reducing agent for assembly back to T=7 capsid backbones that have been documented as more symmetric and stable. To further delineate how MPV.10.34.d refolds into a T=1 capsid backbone, the refolding steps in Example 1 were repeated. Briefly, following solubilization of the IB, MPV.10.34.d was subjected to refolding via removal of the denaturant (6M Urea) in a step-gradient manner. The solubilized samples were inserted into dialysis tubing (snakeskin dialysis tubing, 10,000 Da molecular weight cut off 35 mm. (ThermoFisher Scientific, Waltham, MA, US). In general, about 100 mL to about 150 mL of resolubilized sample solution was dispensed into a four dialysis tubes. The samples were then dialyzed against 4 M urea buffer with the general buffer recipe 50 mM Tris, pH 8.0, 500 mM NaCl and 0.05% Tween-80 for 3±1 hour in a cold room at about 4° C. on a stir plate. Then the samples were again dialyzed against a fresh 1 M urea buffer for 3±1 hour in a cold room on a stir plate. Subsequently, the samples were dialyzed against 0 M urea buffer overnight (about 16 to 18 hours) in a cold room at about 4° C. on a stir plate. The main difference between the four buffer conditions in this experiment were the presence of: (i) 1 mM EDTA, 1 mM PMSF, and 1 mM DTT; (ii) 1 mM EDTA, (iii) 1 mM DTT, and (iv) no added ETDA, PMSF or DTT. The dialyzed/refolded sample solutions under all four conditions were aliquoted into 50 mL conical tubes and analyzed via DLS as described in Example 2 before being stored in a −20° C. freezer. As shown inFIG.7using volume plots (and as explained in Example 2), aggregates as marked by the arrow were observed in sample (ii) and sample (iv). (See,FIG.7CandFIG.7D, respectively). Little to no aggregates were observed on the DLS volume plots in sample (i) or sample (iii). (See,FIG.7AandFIG.7B, respectively). To further confirm whether MPV.10.34.d successfully refolded into T=1 capsid backbones, all samples were dialyzed into capture buffer A (25 mM NaPO4, 25 mM NaCl, pH 6.0) overnight at 4° C. Following dialysis, samples were centrifuged at 4000×g for 10 min at 4° C. and then filtered through a 0.22 μm PES membrane. The T=1 capsid backbone platform was then captured by CEX (EMD Fractogel S03 (M), EMD Millipore, Darmstadt, Germany) and then step eluted with 30% 25 mM NaPO4, 1.5M NaCl, pH 6.0. Results are shown inFIG.8.FIG.8shows successful capture and elution of correctly refolded T=1 capsid backbones in samples that were either refolded in DTT/ETDA/PMSF (FIG.8A) or DTT alone (FIG.8B). In contrast, there was little to no capture and elution of correctly refolded T=1 capsid backbones under refolding conditions that included ETDA alone (FIG.8C) or no additives (FIG.8D) as demarcated by the arrows inFIG.8. Taken together, the data indicate that the process of producing T=1 capsid backbones favors reducing conditions. This contrasts with the known process of producing HPV T=7 capsid backbones that require the eventual removal of any reducing agents for successful capsid backbone assembly. Example 4 Production of Soluble MPV.10.34.d Bacterial expression of MPV.10.34.d at 37° C. for 4 hours in the presence of 1 mM IPTG results in the expression and formation of IBs that must be treated in a series of process steps as described in Example 1 to obtain highly purified 20 nm to 30 nm T=1 capsid backbone structures. To investigate whether soluble MPV.10.34d can be expressed intracellularly inE. coliand whether it can assemble into a T=1 capsid backbone as opposed to an IB, a strategy was adopted to lower the induction temperature and IPTG concentrations to slow down expression, and thereby to prevent formation of IBs. From Example 1, induction temperature was lowered from 37° C. to 16° C. at a concentration of IPTG of 100 μM and 1 mM. Briefly, freshly pickedE. coliC2566 colonies transformed with the MPV.10.34d DNA were inoculated into LB with 50 μg/mL of kanamycin (henceforth LB+KAN) starter cultures, grown overnight at 37° C. and 250 rpm. The next day the starter culture was used to inoculate a fresh 250 mL LB+KAN culture at 1:25 dilution and grown at 37° C., 250 rpm, for about 1.5 to 2 hours, such that the culture reached an OD600 of about 0.5 to 0.7. At this point, the culture was induced with 1 mM and other tested IPTG concentration(s) and shaken at 16° C. induction temperature overnight. The next day, post-induction samples (1 mL each) were pelleted at 1500×g for 10 min. The supernatant was removed and the remaining bulk samples were pelleted at 4000×g for 10 min at 4° C. These samples were stored at −20° C. until analysis. To determine soluble material, cell were lysed using a homogenizer (15,000-20,000 psi, 3 cycles). Samples of uninduced, induced, post-lysis soluble, and post-lysis insoluble material were analyzed by SDS-PAGE and Western blot. Results show that lowering the induction temperature to 16° C. resulted in undetectable levels of protein (Data not shown). Since expression of MPV.10.34.d was low to undetectable at 16° C., induction was attempted at a higher temperature, 25° C. At an induction temperature of 25° C., MPV.10.34d was successfully expressed. A majority of MPV.10.34.d was expressed in the form of IBs (data not shown). A third attempt at 30° C. was attempted under the induction conditions described above. It was discovered that soluble expression improved tremendously at this temperature. There appeared to be a critical transition between 25° C. and 30° C. that allows for MPV.10.34.d to be successfully translated with approximately 50% expressed in a soluble form. In contrast, the concentration of IPTG appeared to have a minor effect in the amount of translated MPV.10.34.d. To purify these soluble MPV.10.34.d, a single step elution chromatography purification was developed and subsequently employed. Briefly, soluble lysate containing MPV.10.34d was prepared in 50 mM NaPO4, 50 mM NaCl, pH 7.0. A 1 mL prepacked Fractogel SO3 (M) column was equilibrated to 50 mM NaPO4, 50 mM NaCl, pH 7.0 and 500 μL of MPV.10.34d lysate was injected onto the column. A series of step gradient elutions were performed to identify a conductivity window for elution of MPV.10.34d, including 5, 10, 15, 20, 25, and 100% B steps (B=50 mM NaPO4, 1.5M NaCl, pH 7.0; which translates to a theoretical NaCl concentration of 0.075M, 0.15M, 0.225M, 0.3M, 0.375M, and 1.5M). Based on the elution of MPV.10.34d, a single step method was developed and scaled up to a 5 mL prepacked Fractogel SO3 (M) column with injection volumes of up to 5 mL. This single step method used 15% B, where B was 50 mM NaPO4, 500 mM NaCl, pH 7.0. (The lower concentration of NaCl allowed for more consistent control of the conductivity, as relying on small percentage changes of a 1.5M NaCl resulted in conductivity irregularities when using 1 mL columns, likely due to pre-column mixing volumes). Based on these efforts, a single step gradient elution method was developed to generate milligram-scale amounts of CEX-captured MPV.10.34d. Elution material was subsequently collected and analyzed as described in Example 2. Results revealed that DLS (FIGS.9A and9B) and TEM data (FIG.9C) from eluted MPV.10.34d yielded T=1 capsid backbones of 20 to 30 nm diameter. Example 5 Soluble and IB MPV.10.34.D Form T=1 Capsid Backbones The MPV.A4 antibody is a conformational antibody that specifically binds to MPV L1 in the form of T=1 or T=7 capsid backbone structure. This antibody will not bind to denatured or monomeric MPV L1. (Hafenstein et al., 2020, “Atomic Resolution CRYOEM structure of Mouse Papillomavirus,” International Papillomavirus Conference, July 20-24, 2020). To determine whether MPV.10.34.d undergoing the steps in Example 1 or Example 4 yields a T=1 capsid backbone, ELISA was performed on these samples with the MPV.A4 monoclonal antibody. Samples from Example 1 and Example 4 of equal concentrations (starting concentration of 1000 ng/well) were subjected to ELISA. To ensure that both soluble and refolded MPV.10.34.d were equally bound to the ELISA plate (Nunc Maxisorp, ThermoFisher Scientific, Waltham, MA, US), both samples were first buffer exchanged into either 50 mM NaPO4, 450 mM NaCl at pH 6 or pH 7. This resulted in two different pH conditions for both samples. Based on this, a total of four sample conditions were two-fold serially diluted and subjected to ELISA with the MPV.A4 monoclonal antibody. Briefly, eight different amounts of protein (7.8 ng to 1μg) for each sample under both pH conditions (into either 50 mM NaPO4, 450 mM NaCl at pH 6 or pH 7) were first added to the ELISA plate and the plate was stored at 4° C. Two days later, ELISA was performed by incubating each plate for one hour at room temperature on an orbital shaker (300 rpm) with MPV.A4 mAb diluted 1:1000 using blocking buffer (4% dry milk, 0.2% Tween-20) and the plates incubated for one hour at 4° C. A wash step was then employed using wash buffer (0.35 M NaCl, 1.5 mM KH2PO4, 6.5 mM Na2HPO4, 0.05% Tween-20) at room temperature for a total of three washes (200 μL per sample per wash). Following the wash step, a goat anti-mouse IgG-HRP antibody (Millipore Sigma, St. Louis, MO, US) was added at 1:7000 dilution in blocking buffer (4% dry milk, 0.2% Tween-20) to a final concentration of 82.9 ng/mL and the plates incubated for one hour at room temperature on an orbital shaker (300 rpm). After the incubation, the plate was washed and incubated with a peroxidase substrate (3, 3′, 5, 5′ tetramethyl benzidine, SeraCare Life Sciences, Inc., Milford, MA, US) for 30 minutes, followed by the addition and incubation of stop solution (0.36 N H2504) (J. T. Baker/Avantor, Allentown, PA, US) for 20 minutes. The absorbance of the sample plates were read at 450 nm and 620 nm with a plate reader (BioTek, Winooski, VT, US). Results (FIG.10) showed that the undialyzed soluble MPV10.34.d capsid backbone, which are captured using buffer at pH 7 (solid circles), as well as the soluble form dialyzed against buffer at pH 7 (solid squares) and pH 6 (solid triangles) were recognized by the MPV.A4 monoclonal antibody. In summary, both MPV.10.34.d capsid backbones refolded from IBs (Example 1) and soluble MPV.10.34.d capsid backbones (Example 4) are both recognized by the MPV.A4 conformational monoclonal antibody. Example 6 IRC Formation: MPV.10.34.d Capsid Backbone Conjugation To functionalize the MPV.10.34.d capsid backbones such that they are effective in recruiting preexisting immune system to attack cancer cells in the subject, the MPV.10.34.d capsid backbones were conjugated to various peptide epitopes including ovalbumin peptide SIINFEKL (OVA, SEQ ID NO: 95), HPV16 E7 protein (SEQ ID NO: 96), and CMV peptide pp65 (SEQ ID NO: 129) to form IRCs. Design of Peptides: The peptides are epitopes having a general length of about 8 to 10 amino acids that are preceded upstream by a protease recognition site. (See,FIG.11). The following experiments incorporate an exemplary protease recognition site, the furin protease cleavage sequence R X R/K R (SEQ ID NO:89) which is designed to be located upstream of the epitope peptide. In addition, the epitope peptide is chemically modified at the N-terminus to contain maleimide. The incorporation of maleimide, a sulfhydryl reactive reagent, to the N-terminus of the peptide antigen allows for conjugation of the protease/peptide to the reduced sulfhydryl groups, i.e., cysteines, on the MPV.10.34.d capsid backbones. The end production of this reaction is a conjugated MPV.10.34.d capsid backbone. To conjugate purified MPV.10.34.d capsid backbones of about >95% purity, the MPV.10.34.d were further dialyzed in conjugation reaction buffer (50 mM NaPO4, pH 6.5, 500 mM NaCl, 2 mM EDTA, and 0.05% Tween® 80), exchanging the buffer three times (3±1 hours, 3±1 hours, and overnight 16±3 hours, at 2° C. to 8° C.). The next day, the MPV.10.34.d was adjusted to a final concentration of at least 0.6 μg/μL. The MPV.10.34.d were then treated with a mild reducing agent, tris(2-carboxyethyl)phosphine (TCEP), for 1 hour without shaking at room temperature (21° C.) at a TCEP:MPV.10.34.d ratio of 10:1. Subsequently, the peptide was then added to the reaction at a molar ratio of X10 the amount of MPV.10.34.d. The reaction was shaken at room temperature (21° C.), 200 rpm, for 1 hour. Following conjugation, to remove excess free peptide, contents from the reaction were subjected to 10 rounds of Amicon spin filtration (molecular cut-off 100 kDa) at 1000 rcf for 10 mins each round. Following this purification step, samples were analyzed by SDS-PAGE stained with Coomassie Brilliant Blue R-250 dye (Bio-Rad, Hercules, CA, US) to determine percent conjugation (4-20% CRITERION™ TGX Stain-Free™ Precast Gels, 10 Well Comb, 30 μL, 1.0 mm, Bio-Rad, Hercules, CA, US). As seen inFIG.12, the upper band at about 50 kDa was determined to correspond to IRC. (See,FIG.12, lane 4). The lower bands represent the MPV.10.34.d lacking epitope peptide. These band identities were further confirmed via the conjugated control. (See,FIG.12, lane 3). Importantly, the conjugation of MPV.10.34.d yielded a conjugation efficiency of about 50% as determined by densitometry. (See,FIG.12, lane 4). Percent conjugation was calculated by densitometry. The gel images were scanned into the computer using manufacturer-recommended software. Subsequently, the total density of the upper band and lower band was calculated by drawing a box or freehand shape around each band. The total density of these regions corresponds to the total amount of protein in each band. Next, only the upper lane was measured in the same way. Background density of a suitably matched area on each gel was collected and subtracted from the band signals. The background-corrected density of the upper protein band was then divided by the entire background-corrected density of the region of interest and then multiplied by 100 to obtain the percent conjugation. Example 7 Conjugation Efficiency of MPV.10.34.d and HPV16 Capsid Backbones Conjugation of HPV L1 particles using the same conjugation reaction steps as described in Example 6 has been previously described. (See, for instance, WO 2018/237115 and WO 2020/139978). Conjugation experiments were conducted on HPV16 capsid backbones and MPV.10.34.d capsid backbones in the manner described in Example 6. The peptide epitope conjugated to the capsid backbones was the HLA-A*0201 restricted epitope NLVPMVATV (NLV, SEQ ID NO: 138) from the HLA-A2 supertype derived from the CMV pp65 antigen. As seen inFIG.13, under the same conjugation reaction conditions of 10:1 (TCEP:L1) and 10:1 (Peptide:L1) ratios, the MPV.10.34.d capsid backbones exhibited a higher conjugation percentage (50%,FIG.13, lane 5) as compared to HPV16 (approximately 20%,FIG.13, lane 3). Percent conjugation was determined by densitometry as described above. Control samples included unconjugated HPV16 (FIG.13, lane 2) and unconjugated MPV.10.34.d (FIG.13, lane 4). The percent peptide conjugation is believed to depend on at least two factors: (1) the ratio of reducing agent to L1 protein, and (2) the amount of free peptide added to the conjugation reaction. For the results shown inFIG.13, it was determined that a ratio of reducing agent to L1 of 10:1 and peptide to L1 ratio of 10:1 results in at least 50% conjugation for MPV.10.34.d capsid backbone (FIG.13, lane 5) and at least 20% for WT HPV16 T=7 capsid backbone (FIG.13, lane 3). To further assess impact of reducing agent concentration, the conjugation reaction was performed at 10:1 reducing agent to L1 protein (FIG.14, lane 3 and lane 10 for HPV16 IRC or MPV.10.34.d IRC, respectively), a 100-fold ratio (FIG.14, lane 4 and lane 11, for HPV16 IRC or MPV.10.34.d IRC, respectively), and a 1000-fold ratio (FIG.14, lane 5 and lane 12, for HPV16 IRC or MPV.10.34.d IRC, respectively). Surprisingly, a ratio of reducing agent to L1 of 100:1 and 1000:1 yielded a lower percent conjugation, and in some instances even no detectable conjugation, as compared with the standard 10:1 reducing agent to L1 protein ratio. Without wishing to be bound by any specific theory, it is possible that excess reducing agent reacts with the peptides to form an -ylene by-product with approximately the same rate as conjugation of peptides to L1 surface thiols. Such a phenomenon could result in depletion of peptides available for conjugation to L1. The relative stability of IRC was also assessed. Following conjugation, the IRC samples were filtered with an Amicon 10 kDa filter spin column to remove excess free peptide. Following filtration, the protein concentration of the samples were checked to ensure no protein was lost during filtration. (See,FIG.14, lanes 6 to 8 and lanes 13 to 15, for HPV16 IRC or MPV.10.34.d IRC, respectively). The samples were then analyzed by TEM as described in Example 2.FIGS.15A and15Bare exemplary TEM micrographs at 150,000× and 200,000× magnification, respectively, showing the breakdown of the HPV16 IRC after Amicon purification. In contrast, MPV.10.34.d IRC did not exhibit any breakdown, as shown inFIGS.15C and15Dat 180,000× and 200,000× magnification, respectively. Without wishing to be bound by any specific theory, it is postulated that perhaps the additional stability of the MPV.10.34.d IRC may be due to the inherent structural stability of the capsid backbone itself, being held together by hydrophobic bonds, as compared to T=7 particles that are held together instead by disulfide bonds. Indeed, the reducing step during the conjugation reaction may in fact destabilize the T=7 structures held together by disulfide bonds via reduction of the necessary thiol groups. As a consequence, the MPV.10.34.d capsid backbone may be comparatively more stable after being treated with up to a ratio of 100:1 or 1000:1 of TCEP to L1. As a result of these findings it was concluded that MPV.10.34.d capsid backbone would serve well as a stable conjugation platform to recruit preexisting immune system components in a subject for the purpose of treating cancer in subjects. Although there is no improvement to conjugation (50% as seen by densitometry on SDS-PAGE gel) of MPV.10.34.d capsid backbone at reducing agent ratios above 1:100, reducing agent ratios higher than 1:10 but lower than 1:100 were investigated to determine whether such ranges might increase conjugation efficiency. Thus, the conjugation reactions were repeated as previously described with varying amounts of reducing agent ratios under 1:100, specifically ratios of 5:1, 10:1, and 25:1. Peptide to MPV.10.34.d ratios (5:1, 10:1, and 25:1) were also evaluated. As seen inFIG.16, a dose-dependent peptide conjugation on MPV.10.34.d capsid backbone using 5:1, 10:1 and 25:1 peptide:L1 ratio when the reducing agent: L1 protein ratio is between 5:1 (lanes 1, 4, and 7) was observed. No peptide dose dependence was seen with reducing agent: L1 protein ratio at 10:1 (lanes 2, 5, and 8) and 20:1 (lanes 3, 6, and 9). Lane 6 is a reference point in which 10:1 reducing agent to L1 ratio and 10:1 peptide to L1 ratio were used, conditions under which an approximately 50% level of peptide conjugation is routinely observed as determined by densitometry. The lane labelled “CEX-FB035” is a control containing only MPV.10.34.d capsid backbones. It was determined that a 5:1 ratio of reducing agent to L1 along with a 10:1 ratio of peptide to L1 yielded a conjugation efficiency above 50%. (See,FIG.16, lanes 4 and 7). Conjugation conditions with ratios of 1:1, 2.5:1, and 5:1 reducing agent to L1 protein were also tested. The results obtained at these ratios are reported inFIG.17. It was determined that lowering the amount of the reducing agent from 5:1 to 1:1 did not improve conjugation efficiency, regardless of the amount of peptide included in the reactions. InFIG.17, lane R is a reference point sample in which a peptide to L1 ratio of 10:1 was included as well as reducing agent to L1 ratio of 10:1, which typically yields an approximately 50% level of peptide conjugation. The lane labelled “CEX-FB035” is a control containing only MPV.10.34.d capsid backbones. In summary, it was determined that a 5:1 reducing agent to L1 ratio and a 10:1 peptide to L1 ratio achieves conjugation rates above 50%. Example 8 Binding of IRC to Tumor Cells via HSPG To assess whether IRC bind to tumor cells, an in vitro cell binding assay was conducted. Specifically, both MPV.10.34.d capsid backbones (unconjugated) as well as different conjugated IRC (human CMV pp65, murine E7, and murine OVA peptide) were examined. Briefly, 2×105MC38 cells (murine colon adenocarcinoma, #ENH204-FP Kerafast, Inc., Boston MA) or pgsA-745 cells (Chinese hamster ovary cell mutant deficient in xylosyltransferase (UDP-D-xylose:serine-1,3-D-xylosyltransferase, ATCC CRL-2242) in which heparin sulfate proteoglycan (HSPG) expression is knocked out, were seeded overnight. The next day, the cells were treated with human CMV pp65, murine HPV16 E7, and murine OVA peptide, as well as the MPV.10.34.d capsid backbone for one hour at 37° C. Cells were then washed twice with 2 to 3 mL of a fluorescence activated cell sorting (FACS) buffer (1% bovine serum albumin in PBS) and then stained with 1 mL of rabbit anti-musPsV serum antibodies for 30 minutes at 4° C. Following this, samples were washed once with 3 mL FACS buffer and stained with 0.5 mL of donkey anti-rabbit IgG-PE antibody (Biolegend, San Diego, CA) for 30 min at 4° C. in the dark. Finally, samples were washed once more with 3 mL FACS buffer and then resuspended in 250 mL of FACS buffer before being analyzed by a CytoFLEX flow cytometer (Beckman Coulter Life Sciences, Brea, CA, US). As shown inFIG.18AandFIG.18B, all of the constructs, MPV.10.34.d-CMV pp65 IRC (solid line), MPV.10.34.d-E7 IRC (thick solid line), MPV.10.34.d-OVA IRC (thick dashed line), and MPV.10.34.d capsid backbones (dashed-line), exhibited specificity for tumor cells as evidences by the peak shifts to the right. The positive control in these experiments was MPV capsid backbone (wild type, dotted line). The negative control included no IRC or L1 (long-dashed line). These experiments further show that the IRC exhibited HSPG-specific binding since no binding of MPV.10.34.d capsid backbones was observed in the cell line lacking HSPG expression (pgsA-745 cells, indicated by no shift in the peaks inFIG.17B). In summary, these results show that binding specificity of MPV.10.34.d capsid backbones for tumor cells is HSPG specific, and importantly, conjugation of epitope peptides to MPV.10.34.d capsid backbone does not reduce or otherwise negatively impact binding of MPV.10.34.d IRC to tumor cells in vitro. Example 9 Loading of Peptide onto Tumor Cells by MPV.10.34.d IRC The MPV.10.34.d IRC are designed such that upon entering the tumor microenvironment, the peptide will be cleaved from the IRC, thereby releasing the peptide in the near vicinity of a tumor cell surface. The cleavage event occurs, in some embodiments, upon contact with a tumor-specific protease, i.e., a protease present, in some embodiments at relatively higher concentrations than elsewhere in the subject's system, on or nearby a tumor cell. This cleavage event then is designed to result in the loading, or binding, of the peptide by MHC molecules expressed on the surface of tumor cells. The following experiments are designed to test this mode of operation and whether the designed IRC operate in the manner expected. For this purpose, an MHC class I molecule loading assay was developed that directly detects peptide loading from IRC onto MHC class I molecules expressed on the surface of tumor cells. This assay involves the use of an antibody that specifically recognizes an OVA peptide (SIINFEKL, SEQ ID NO:95)—MHC class I alloantigen H-2Kbmolecule complex but not free peptide, empty MHC class 1 molecules, or peptides conjugated to the IRC. (See, Zhang et al.,Proc. Nat'l. Acad. Sci. USA,89:8403-84-7, 1992). In this experiment, the OVA conjugated MPV.10.34.d IRC from Example 6 were examined side-by-side with OVA conjugated HPV16 IRC at equivalent molarity based on concentration of conjugated peptide. Briefly, 0.1 to 0.2×106MC38 tumor cells (C57BL6 murine colon adenocarcinoma-derived cells, #ENH204-FP, Kerafast, Inc., Boston MA) were incubated with the IRC for one hour at 37° C. A positive control including just free peptide and a negative control including no peptide or IRC were also tested. Cells were then washed twice with 2 to 3 mL FACS buffer and then stained with PE-conjugated-mouse anti-mouse MHC I bounded with OVA (SIINFEKL, SEQ ID NO:95) monoclonal antibody (Biolegend, San Diego, CA) for 30 minutes at 4° C. Following this, samples were washed once with 3 mL FACS buffer then the cells were resuspended in 250 μL of FACS buffer before being analyzed by a CytoFLEX flow cytometer (Beckman Coulter Life Sciences, Brea, CA, US). Results of these assays are provided inFIG.19. The results show that OVA-conjugated MPV.10.34.d IRC (1.4 μg/mL, thick solid line) and OVA-conjugated HPV16 IRC (2.5 μg/mL, solid line) demonstrated loading of epitopes on the surface of MC38 murine tumor cells, with the OVA-conjugated MPV.10.34.d IRC out-performing the OVA-conjugated HPV16 IRC. (See,FIG.18, negative control—long-dashed line, positive control—thin-dashed line). These results suggest that the MPV.10.34.d IRC is superior to the HPV16 IRC because a smaller amount of MPV.10.34.d IRC achieved the same, or better, “loading” potential of a larger amount of HPV16 IRC. As OVA is a model antigen utilized for murine MHCs, this experiment was repeated substituting the CMV pp65 peptide for the OVA peptide. The HLA-A*0201 restricted epitope NLVPMVATV (NLV, SEQ ID NO: 138) from the CMV pp65 was used for these studies and the pp65-conjugated MPV.10.34.d IRC were produced as described in Example 6. As there was no commercially available monoclonal antibody that recognizes an MHC class I—NLV complex, a soluble T-cell receptor antibody (2S16) was employed that recognizes this HLA-A2 complex. (See, Wagner et al.,J. Biol. Chem.,295(15):5790-5804, 2019). The IRC constructs were analyzed in the same manner as above except that the cell lines HCT116 (human colorectal carcinoma cell line, HCT 116, ATCC, CCL-247) and MCF7 (human breast cancer cell line, MCF7, ATCC, HTB-22) were utilized in this study. These cell lines are HLA-A*0201 restricted and thus are able to present the HLA-A*0201 restricted epitope NLVPMVATV (NLV, SEQ ID NO: 138) from the CMV pp65 peptide. Consistent with the OVA MHC class I loading results, loading of the NLV peptide onto human tumor cells was observed. Results are presented inFIG.19A(HCT116 cells) andFIG.19B(MCF7 cells). InFIG.20AandFIG.20B, an unrelated hepatitis B peptide was used as a negative control (thin dashed line, 10 μg/mL), unconjugated MPV.10.34.d capsid backbones were used as a further negative control (thin solid line, 100 μg/mL), CMV conjugated MPV.10.34.d IRC is represented as a thick solid line (100 μg/mL, at about 1.7 μg/mL of hCMV peptide conjugated), hCMV free peptide is represented as a thick dashed line (1 μg/mL). FIGS.19and20show that incubation of the MPV.10.34.d IRC in vitro with the indicated cell lines leads to release of the peptide and binding to the tumor cell surface MHC Class 1 molecules. To further demonstrate that the mechanism of the IRC first involves the MPV.10.34.d IRC binding to the tumor cell followed by furin cleavage, competitive inhibition experiments were performed that either inhibited tumor cell binding or furin cleavage to show that ablation of either step results in an absence of peptide loading onto the tumor cells. These studies were performed with the OVA-conjugated MPV.10.34.d IRC and conducted under the same conditions as the binding assays described above. To block tumor binding, soluble heparin (Sigma Aldrich, St. Louis, MO) at 1 mg/mL, 5 mg/mL, or 10 mg/mL was incubated with 2.5 μg/mL of OVA-conjugated MPV.10.34.d IRC for 1 hour at 37° C., 5% CO2in the presence of 2×105MC38 cells in a FACs tube. The final volume of the cells with the sample was 200 μL. A positive control sample was included which contained no soluble heparin as well as a negative control that contained no IRC or heparin. Cells were then washed twice with 2 to 3 mL FACS buffer and then stained with PE-conjugated-mouse anti-mouse MHC I bound to the OVA peptide monoclonal antibody (this monoclonal antibody is able to specifically detect OVA peptide, SIINFEKL, SEQ ID NO: 95, in complex with MHC-I Kb) for 30 minutes at 4° C. Following this, samples were washed once with 3 mL FACS buffer, then the cells were resuspended in 250 μL of FACS buffer before being analyzed by a CytoFLEX flow cytometer (Beckman Coulter Life Sciences, Brea, CA, US). As seen inFIGS.21A,21B, and21C, no OVA peptide loading was observed in the negative control (thin black line inFIGS.21A,21B, and21C). No OVA peptide loading was observed in samples including 10 mg/ml (dashed line,FIG.21A), 5 mg/ml (dashed line,FIG.20B), or 1 mg/ml (dashed line,FIG.21C) soluble heparin (these curves overlapped with the negative control data). Loading of OVA peptide was only detected in the samples containing OVA-conjugated MPV.10.34.d IRC with no heparin (thick black line to the right,FIGS.21A,21B, and21C). These results show that OVA-conjugated MPV.10.34.d IRC is HSPG-specific. To show that loading of peptide from IRC onto tumor cells is dependent on protease cleavage of the epitope peptide form the MPV.10.34.d IRC, the experiments of Example 10 were repeated in the presence of a furin inhibitor, furin inhibitor I—Calbiochem, decanoyl-RVKR-CMKa, peptidyl chloromethylketone. (Millipore-Sigma, St. Louis, MO). This furin inhibitor binds irreversibly to the catalytic site of furin, blocking all furin protease activity. Briefly, 2×105MC38 cells (murine colon adenocarcinoma, #ENH204-FP, Kerafast, Inc., Boston MA) were seeded in a FACs tube and then incubated with either 0.5 μM, 5 μM, or 50 μM furin inhibitor dissolved in DMSO at total final sample volume of 200 μL. Control samples containing no inhibitor were prepared the same way with the same volume equivalent of DMSO. The samples were incubated for fifteen minutes in a tissue culture incubator at 37° C., 5% CO2. Then, 2.5 μg/mL of OVA-conjugated MPV.10.34.d IRC was added to all samples and the samples were incubated in a tissue culture incubator at 37° C., 5% CO2. Samples were then washed twice with 2 to 3 mL FACS buffer and then stained with PE-conjugated-mouse anti-mouse MHC I bounded with OVA (SIINFEKL, SEQ ID NO:95) monoclonal antibody (Biolegend, Cat #141604, San Diego, CA) for 30 minutes at 4° C. Following this, samples were washed once with 3 mL FACS buffer then the cells were resuspended in 250 μL of FACS buffer before being analyzed by a CytoFLEX flow cytometer. (Beckman Coulter Life Sciences, Brea, CA, US). As seen inFIGS.22A,22B, and22C, OVA-conjugated MPV.10.34.d IRC loaded OVA peptide onto tumor cells in the samples that had no inhibitor added (thin black line,FIGS.22A,22B, and22C), and samples treated only with DMSO and no furin inhibitor (dashed black line,FIGS.22A,22B, and22C). In contrast, no OVA peptide loading was observed in the negative control (thin grey line inFIGS.22A,22B, and22C, the control curves overlapped with the experimental data) as well as samples treated with furin inhibitor at 50 μM (arrow pointing to dark thin line,FIG.22A), 5 μM (arrow pointing to dark thin line,FIG.22B), and 0.5 μM (arrow pointing to dark thin line,FIG.22C). Therefore, inhibition of furin cleavage of the epitope peptide from the IRC prevented binding of OVA to the MHC molecules of the target cancer cell, thereby confirming the mechanism of action of the IRC. This mechanism is further confirmed by, and consistent with, the results shown inFIGS.18to22. Example 10 In Vitro Cytotoxic Killing Assays with MPV.10.34.d IRC Since it was shown in Example 9, that in vitro MPV.10.34.d IRC were able to deposit peptide epitopes onto murine and human MHC Class I molecules and that this mechanism was dependent on furin activity, additional experiments were designed to determine whether labelling these cancer cells would trigger activation and redirection of cellular immune system components against target tumor cells. Upon activation and redirection, the goal is delivery of a cytotoxic signal to the tumor cells and tumor cell death. For this purpose, three different in vitro cytotoxic T-cell-dependent tumor cell killing assays were designed involving the co-culture of tumor cells and viral antigen-specific CD8+ T cells in the presence or absence of MPV.10.34.d IRC. The three CD8 T-cells were tested, including: (1) murine OVA-specific preclinical CD8+ T cells, (2) murine HPV16 E7-specific CD8+ T cells, and (3) human HLA-A*0201-restricted CMV-specific T cells (Astarte Biologicals, cat #1049-4367JY19). (See, Example 12, for (3)). Murine B16 (melanoma/skin) (B16-F10 (ATCC® CRL-6475™), and murine ID8 (ovarian) tumor cells (Hung et al.,Gene Ther.,14(12):921-020, 2007) overexpressing luciferase gene (B16-luc and ID8-luc) were grown in culture. Under normal circumstances, murine tumor cell lines B16 and ID8 will not be killed by murine OVA-specific CD8+ T cells since these cell lines do not express the murine OVA (SIINFIKEL, SEQ ID NO: 95) antigen. Approximately 0.01×106B16-luciferase mutant (B16-luc) or 0.005×106ID8-luciferase mutant (ID8-luc) tumor cells were seeded in 100 μL per well on a 96-well assay plate overnight. The cells were then treated with 100 μL of 2.5 μg/mL of MPV.10.34.d capsid backbones, OVA-conjugated MPV.10.34.d IRC, OVA-conjugated HPV16 IRC, and positive control containing 1 μg/mL of free OVA peptide (SIINFIKEL, SEQ ID NO: 95), for one hour at 37° C. in a final volume of 200 μL per well. Cells not receiving any antigen were included as negative control (No Ag). The cells were then washed twice with 200 μL of Roswell Park Memorial Institute (RPMI) media and co-incubated with OVA-specific CD8+ T-cells (Jackson, stock no. 003831) at an effector (CD8+ T-cell) to target cell (tumor cell) ratio (“E:T Ratio”) of 10:1 (B16-luc) or 20:1 (ID8-luc) for 16 hours in a final volume of 200 μL per well in a cell incubator at 37° C., 5% CO2. An E:T ratio of 10:1 means that for every 1 tumor cell, ten CD8+OT-1 T cells will be co-incubated with the tumor cell. These co-incubated cells were then washed with 200 μL of PBS and lysed with 35 μL of 1× cell lysis buffer (Promega, Madison, WI, US) for 15 to 20 minutes before adding 50 μL of luciferase assay substrate and detected on a Promega GloMax Explorer Microplate Reader (Promega, Madison, WI, US). The number of viable tumor cells after co-incubation with T cells were measured by quantification of concentration of luciferase released from lysed cells. This acts as a surrogate marker for cell viability since the target cells were incubated under conditions in which they over-express luciferase. Reduced luciferase activity indicated more cell death suggesting greater immune redirection and hence greater cytotoxicity. As shown inFIG.23AandFIG.23B, the OVA-conjugated MPV.10.34.d IRC, OVA-conjugated HPV16 IRC, and the peptide positive control showed much higher tumor cell cytotoxicity (>70%) than the negative control samples in both B16-luc (FIG.23A) and ID8-luc (FIG.23B) tumor killing assays. At the same concentration (2.5 μg/mL), OVA-conjugated MPV.10.34.d IRC also showed similar high cytotoxicity on tumor cells as the OVA-conjugated HPV16 IRC. Similar to the above experiment, a second experiment was performed in like manner, except with the substitution of the E7 peptide (RAHYNIVTF, SEQ ID NO:96) for the OVA peptide. The same tumor cells and samples were investigated in this experiment using the same protocol. As shown inFIG.24AandFIG.24B, the E7-conjugated MPV.10.34.d IRC, E7-conjugated HPV16 IRC, and the positive control showed much higher tumor cell cytotoxicity (>70%) than the negative control groups in both B16-luc (FIG.24A) and ID8-luc (FIG.24B) tumor killing assays. At the same concentration (2.5 μg/mL), OVA-conjugated MPV.10.34.d IRC also showed similar high cytotoxicity on tumor cells as the OVA-conjugated HPV16 IRC. Example 11 MPV.10.34.d IRC Effectiveness in Human Assays While the in vitro functional test results of the above experiments were promising, the next desired step in the analysis was to perform similar experiments in human-based assays. To this end, the response of mock human cellular immune system components to tumor cells exposed to MPV.10.34.d IRC was examined in vitro. Human CMV (HCMV) was selected for this study since human CMV is highly prevalent (infecting 50-90% of the human population) and mostly asymptomatic in healthy individuals. (See, Longmate et al.,Immunogenetics,52(3-4):165-73, 2001; Pardieck et al., F1000Res, 7, 2018; and van den Berg et al.,Med. Microbiol. Immunol.,208(3-4):365-373, 2019). Importantly, HCMV establishes a life-long persistent infection that requires long-lived cellular immunity to prevent disease. Hence, it is rational to hypothesize that a complex adaptive cell-mediated anti-viral immunity developed over many years to strongly control a viral infection in an aging person can be repurposed and harnessed to treat cancer. In these experiments, CD8+ T cell responses to CMV peptides were tested in three different human tumor cell lines, including HCT116, OVCAR3, and MCF7. All three of these human tumor cell lines are HLA-A*0201 positive. In vitro cytotoxicity assays. HTC112, human colon cancer cells, MCF7, human breast cancer cells, and OVCAR3, human ovarian cancer cells (all from ATCC, Manassas, VA, US) were seeded overnight at 0.01 to 0.2×106per well per 100 μL per 96 well plate. The next day (about 20 to 22 hrs later), each cell line was incubated for one hour at 37° C. under the following conditions: (1) CMV peptide at a final concentration of 1 μg/mL (positive control), (2) MPV.10.34.d at a final concentration of 2.5 μg/mL (negative control), (3) CMV-conjugated MPV.10.34.d IRC at a final concentration of 2.5 μg/mL, (4) CMV-conjugated HPV16 IRC at a final concentration of 2.5 μg/mL, and (5) no antigen (negative control). After 1 hour, the cells were washed vigorously with 200 μL of media for three times to remove non-specific binding. Human patient donor CMV T cells (ASTARTE Biologics, Seattle, WA, US) were added at the E:T (effector cell:target cell) ratio of 10:1 and incubated in a tissue culture incubator for 24 hrs at 37 C, 5% CO2. The total final volume of each sample after co-culture was 200 μL. Cell viability was measured after co-culturing. Cell viability was measured with CELLTITER-GLO® (Promega, Madison, WI, US). This assay provides a luciferase-expressing chemical probe that detects and binds to ATP, a marker of cell viability. The amount of ATP generated from tumor cells was quantified according to manufacturer protocols. In these assays, reduced luciferase activity indicates cell death and suggests greater immune redirection and greater cytotoxicity. The results are provided inFIG.25. CMV-conjugated MPV.10.34.d IRC (“VERI-101” inFIGS.25A,25B, and25C) was equally effective as CMV-conjugated HPV16 IRC (“CMV AIR-VLP” inFIGS.25A,25B, and25C) in redirecting human healthy donor CMV pp65-specific CD8+ T-cells (Astarte Biologics, Inc., Bothell, WA, US) to kill immortalized HLA.A2 positive human colon cancer cells (HCT116), human ovarian cancer cells (OVCAR3), and human breast cancer cells (MCF7). The control samples (“No Ag” or “VERI-000” inFIGS.25A,25B, and25C) showed no background tumor killing. Together, these data demonstrate that MPV.10.34.d IRC redirects mouse and human immune responses against tumor cells in vitro. Example 12 Sequential Mechanism of MPV.10.34.d IRC Binding and Peptide Cleavage Example 9 demonstrates that MPV.10.34.d IRC binding must occur prior to furin-dependent cleavage of the peptide and peptide loading onto target tumor cells. A dose-response curve using different concentrations of OVA-conjugated MPV.10.34.d IRC to detect binding and loading in separate assays was generated. These assays were performed as described in Examples 7 and 8. Based on the geometric MFIs from both assays, a correlation analysis was conducted. The results shown inFIG.26indicate that there is a highly statistically significant correlation between the number of OVA-conjugated MPV.10.34.d IRC binding to tumor cells with the level of the OVA peptide/Kbcomplex on the tumor cells (Spearman r=0.92, P=0.0003; Pearson r=0.98, p<0.001). This statistical analysis further demonstrates the requirement for the sequential steps of OVA-conjugated MPV.10.34.d IRC to first bind or contact the tumor cell, followed by furin-dependent cleavage of the peptide from the IRC, and MHC loading of the peptide. Example 13 Sequential Mechanism of MPV.10.34.d IRC Binding and Tumor Cell Death Example 9 shows that inhibition of OVA-conjugated MPV.10.34.d IRC binding results in inhibition of furin-dependent cleavage of the peptide from the IRC and OVA peptide loading onto tumor cell surfaces. To further show that inhibition of this binding step also inhibits redirection of CD8+ T-cells and tumor cell death, cytotoxicity studies conducted as in Example 10 were performed in the presence and absence of soluble heparin, a competitor of HSPG binding. A range of OVA-conjugated MPV.10.34.d IRC concentrations (0.156 μg/mL to 0.625 μg/mL) as well as E:T ratios (1:4.5, 1:9 and 1:18) were investigated in the presence and absence of 10 mg/mL of soluble heparin in the assays described in Example 10. This concentration of soluble heparin was previously shown to cause complete inhibition of OVA-conjugated MPV.10.34.d IRC binding, as well as inhibition of peptide loading onto tumor cells. In these assays, 15,000 TC-1 cells overexpressing luciferase were first seeded in a flat-bottom 96-well plate overnight in a cell culture incubator at 37° C., 5% CO2. The next day, cells were washed 3 times with PBS before being incubated with AIM-V media (serum free) with 2% BSA for 1.5 hours in a cell culture incubator at 37° C., 5% CO2. In parallel, OVA-conjugated MPV.10.34.d IRC was diluted in the same AIM-V media+2% BSA into 0.625, 0.3125, 0.156 μg/mL. (See,FIGS.27A,27B, and27C). Each sample was incubated with (thick dash lines) or without (solid line) 10 mg/mL of soluble heparin for 1 hour at 2° C. to 8° C. MPV.10.34.d capsid backbones (thin dash line, “ViP” only) was included as a negative control. After 1 hour, the samples were added to the TC-1 cells and co-incubated for a further 30 minutes in a cell culture incubator at 37° C., 5% CO2. Following this, treated cells were washed 3 times with just AIM-V media before being incubated with OT-1 T-cells at an effector to target (E:T) ratio of 18:1, 9:1, or 4.5:1. This co-culture was then incubated for a further 3 hours at 37° C., 5% CO2. After 3 hours, target cells were analyzed for cytotoxicity using the Promega Luciferase Assay system as per the manufacturer's protocol (Promega, Madison, WI, US). Cytotoxicity was determined by detection of loss of luciferase signal which is used as a surrogate marker of cell viability in this assay. All studies were performed in triplicate. Results are shown inFIG.27. The presence of soluble heparin exhibited no OVA-conjugated MPV.10.34.d IRC-mediated cytotoxicity under all concentrations and E:T ratio conditions tested. These results further substantiate the sequential nature of the MPV.10.34.d IRC mechanism of action. A correlation analysis was conducted on the binding and cytotoxicity activities of OVA-conjugated MPV.10.34.d IRC. Briefly, a dose-response curve using different concentrations of OVA-conjugated MPV.10.34.d IRC to detect binding and cytotoxicity in separate assays was generated. Cytotoxicity assays were conducted as previously described in Example 10 with the following changes: a range of 6.25×10−5μg/mL to 2.5 μg/mL of OVA-conjugated MPV.10.34.d IRC was tested at 3 different E:T ratios (18:1, 9:1, and 4.5:1). Under all 3 E:T ratio conditions tested, a dose dependent killing was observed with OVA-conjugated MPV.10.34.d IRC concentrations below 0.04 μg/mL and higher, whereas concentrations of OVA-conjugated MPV.10.34.d IRC between 0.156 μg/mL to 2.5 μg/mL lead to a maximal level of cytotoxicity. Binding assays were conducted according to the protocols described in Example 7 with the following changes: a concentration range of 6.24×10−4to 2.5 μg/mL of OVA-conjugated MPV.10.34.d IRC was investigated. Results show that a dose-dependent binding was observed and that the limit of binding detection was reached at 2.5×10−4μg/mL. Both assays were repeated twice (with at least 3 replicates). The mean values of geometric mean fluorescent intensity (MFI) was reported from the two experiments and is summarized inFIG.28. Based on the MFIs from both assays (FIG.28), a graphical and correlation analysis was conducted using Spearman correlational analysis (FIG.29). Briefly, the mean of the percentages of two independent OVA-conjugated MPV.10.34.d IRC cytotoxicity assays performed on two different days and the mean of MFIs of OVA-conjugated MPV.10.34.d IRC binding experiments from two different days were calculated (FIG.28) and plotted (FIG.29). Spearman correlational analysis was performed on these results reveals a significant relationship (r=0.83-0.9) between these two variables at all three E:T ratios. These results show that there is a highly statistically significant correlation (r value between 0.83 to 0.9, depending on E:T ratio) between the OVA-conjugated MPV.10.34.d IRC binding to tumor cells and the level of cytotoxicity that followed. Example 14 GARDASIL®9-Generated Antibodies do Not Inhibit MPV.10.34.d IRC Effects Vaccination with GARDASIL®9 results in long term (>10 years) of sustained HPV L1 capsid-specific antibodies that are able to prevent HPV infection and subsequently, prevent HPV-associated cervical cancers. Although GARDASIL®9 has been reported to be only effective against nine types of HPVs, some cross-neutralization against other types of papillomavirus capsids may be expected. As MPV.10.34.d IRC is derived from murine papillomavirus capsids, it was desirable to determine whether vaccine sera elicited from GARDASIL®9 vaccination could inhibit MPV.10.34.d IRC tumor cell killing. GARDASIL®9 sera was generated as follows: New Zealand white rabbits (n=10) were administered three intra-muscular vaccinations of a human dose of GARDASIL®9 (270 μg of VLPs per dose). Rabbits were vaccinated at months 0, 1, and 2. After two weeks post final vaccination, rabbits were bled to obtain the GARDASIL®9 sera. 100 μL aliquots of sera from each rabbit were pooled. As a control, GARDASIL®9 sera were also tested for neutralizing activity against HPV types 6, 11, 16, 18, 31, 45, 52, and 58 and results showed no neutralization activity (data not shown). OVA-conjugated MPV.10.34.d IRCs were tested with GARDASIL®9 sera using the protocol described in Examples 11 and 13. Briefly, 15,000 TC-1 cells overexpressing luciferase were first seeded in a flat-bottom 96-well plate overnight in a cell culture incubator at 37° C., 5% CO2. The next day, cells were washed 3 times with PBS before being incubated with AIM-V media (serum free)+2% BSA for 1.5 hours in a cell culture incubator at 37° C., 5% CO2. In parallel, OVA-conjugated MPV.10.34.d IRC was diluted in the same AIM-V media+2% BSA into 0.625 μg/mL, 0.3125 μg/mL, and 0.156 μg/mL, and each sample was incubated with a 1:200 dilution of GARDASIL®9 serum (thick-dashed lines) or without (solid line) for 1 hour at 2° C. to 8° C. MPV.10.34.d alone (thin dashed line) was also included as a negative control. (See,FIGS.30A,30B, and30C). After 1 hour, the samples were added to the TC-1 cells and co-incubated for 30 minutes in a cell culture incubator at 37° C., 5% CO2. Following this, treated cells were washed 3 times with just AIM-V media before being incubated with OT-1 T-cells at an effector to target (E:T) ratio of 18:1, 9:1, or 4.5:1. This co-culture was then incubated for 3 hours at 37° C., 5% CO2. After 3 hours, target cells were analyzed for cytotoxicity using the Promega Luciferase Assay system as per the manufacturer's protocol (Promega, Madison, WI, US). Cytotoxicity was determined by quantitation of the loss of luciferase signal which is used as a surrogate marker of cell viability. All studies were performed in triplicate. No inhibition of cytotoxicity was observed in the presence of GARDASIL®9 sera. The results inFIG.30suggests that MPV.10.34.d IRC would not be negatively impacted in subjects who might possess preexisting HPV vaccine-generated antibodies. Example 15 Anti-MPV.10.34.d IRC Antibodies do Not Inhibit Tumor Cell Cytotoxicity Since MPV.10.34.d IRC sequences are based on MPV L1 capsids, it was desirable to test whether antibodies generated against wild type MPV or MPV.10.34.d IRC affect the mechanism of action of MPV.10.34.d IRCs against tumors. Antibodies against wild type MPV were generated as follows: New Zealand white rabbits (n=3) were administered three intra-muscular vaccinations of 50 μg of wild type mouse papillomavirus particles per dose. Rabbits were vaccinated at months 0, 1, and 2. After two weeks post final vaccination, rabbits were bled to obtain the anti-MPV sera. Antibodies against MPV.10.34.d IRC were obtained as follows: naïve 6 to 8 week old C57/BL6 mice (n=10) were injected systemically with two doses of 150 μg of E7-conjugated MPV.10.34.d over a period of 48 hours. After 48 hours, the mice were bled to obtain the anti-MPV.10.34.d IRC sera. Specificity of anti-MPV.10.34.d IRC sera and anti-MPV sera to both MPV.10.34.d capsid backbone and MPV.10.34.d IRC was examined by ELISA. The assays were performed as described in Example 5 with the following differences: serum samples were tested at a 1:100 dilution factor and two-fold serial dilutions. Goat anti-mouse IgG-HRP secondary antibody was used in the ELISA (1:7000). Binding of anti-MPV.10.34.d IRC sera (FIG.31) and anti-MPV serum (data not shown) to both MPV.10.34.d capsid backbone (FIG.31A) and MPV.10.34.d IRC (FIG.31B) was observed. To determine whether binding of either antibody serum to MPV.10.34.d IRCs would affect the subsequent tumor cytotoxicity, binding and cytotoxicity assays were conducted with OVA-conjugated MPV.10.34.d IRC in the presence or absence of either sera. Cytotoxicity assays were conducted as in Example 14 in the presence of anti-MPV serum (FIGS.32A,32B, and32C) or anti-MPV.10.34.d IRC serum (FIGS.32D,32E, and32F). Results reveal that no difference in cytotoxicity when OVA-conjugated MPV.10.34.d IRC was pre-incubated with either anti-MPV sera (FIGS.32A,32B, and32C) or anti-MPV.10.34.d IRC sera (FIGS.32D,32E, and32F) for all concentrations and E:T ratios tested. The inability of antibodies to MPV.10.34.d IRC or MPV to inhibit cytotoxicity despite these antibodies showing specificity for binding to both MPV.10.34.d capsid backbones and MPV.10.34.d IRCs via ELISA was further investigated by conducting binding assays in the presence of these sera. Samples were pre-incubated with different OVA-conjugated MPV.10.34.d IRC concentrations (0.0025 μg/mL to 2.5 μg/mL). These samples were tested in the same binding assays described in Example 5. Briefly, 20,000 TC-1 cells overexpressing luciferase were seeded into FACs tubes and incubated in a cell culture incubator at 37° C., 5% CO2, until needed. In parallel, OVA-conjugated MPV.10.34.d IRC was diluted in the same AIM-V media +2% BSA into a range of concentrations 0.0025 to 2.5 μg/mL and each sample was incubated with 1:200 dilution of either anti-MPV serum (A) or anti-MPV.10.34.d IRC serum (B) for 1 hour at 2° C. to 8° C. After 1 hour, the samples were added to the TC-1 cells seeded in the FACs tubes and co-incubated for 1 hour in a cell culture incubator at 37° C., 5% CO2. Samples were then washed twice with FACS buffer (DBPS, pH 7, 0.1% BSA). The samples were then stained with AF647-conjugated donkey anti-rabbit Ig antibody or PE-conjugated goat anti-mouse IgG antibody for 30 minutes in the dark. Samples were then washed with FACS buffer (DBPS, pH 7, 0.1% BSA). After this, samples were resuspending in 250 μL of FACs buffer and binding was detected by flow cytometry at different concentrations of OVA-conjugated MPV.10.34.d IRC pre-treated with MPV sera (FIG.33A) or anti-MPV.10.34.d IRC sera (FIG.33B). All studies were performed in triplicate. Results of these tests reveals that incubation of samples with anti-MPV rabbit IgG serum (FIG.33A) or anti-MPV.10.34.d IRC mouse IgG serum (FIG.33B) are still able to bind to target tumor cells. In summary, the results from Example 15 (FIGS.31,32, and33) collectively show that antibodies specific for MPV.10.34.d IRCs elicited in vivo bind specifically to the MPV.10.34.d IRCs. However, these antibodies do not block MPV.10.34.d IRCs from binding to tumor cells (FIG.33) and thus, also do not inhibit the overall cytotoxic mechanism of the MPV.10.34.d IRCs (FIG.32). In contrast, these same antibodies were able to inhibit MPV infection as seen in the pseudo-virus neutralization assays (data not shown). As MPV infection requires cell internalization, one possible explanation is that these anti-MPV.10.34.d IRC serum antibodies block cell internalization of MPV.10.34.d IRC but do not inhibit target cell binding of MPV.10.34.d IRC. Since the mechanism of MPV.10.34.d IRCs is extracellular, it is possible that this is why they are not affected by these antibodies. Example 16 Peptide Loading on Cells Deficient in MHC Class 1 Intracellular Pathway Components Example 9 demonstrates loading of peptide from IRCs onto the tumor cell surface. It was desirable to determine whether the released peptides from the IRC are bound by the tumor cell and then phagocytosed to be processed through the MHC Class 1 antigen presentation pathway. To test this possibility, peptide loading assays were performed with a cell line (RMA-S cells) that is genetically deficient (TAP-deficient) in intracellular MHC Class 1 processing proteins. If peptide loading onto the tumor cell surface still occurs in this context, it must be through extracellular mechanisms. A range of OVA-conjugated MPV.10.34.d IRC concentrations (0.625 μg/mL to 10 μg/mL) was tested for their ability to load RMA-S cells with peptide. These binding assays were conducted as previously described. Briefly, RMA-S cells at 2×106cells/mL were resuspended into a single cell suspension and 100 μL (0.2×106of RMA-S cells) was dispensed into FACS tubes. Varying amounts of OVA-conjugated MPV.10.34.d IRC was added (0.625 μg/mL to 10 μg/mL) to the samples and the samples were then incubated at 37° C., 5% CO2for one hour. Afterwards, the cells were washed twice in 2 mL of FACS buffer (PBS, pH7.0, 1% BSA). The cells were stained with 1 μL of PE-conjugated anti-SIINFEKL (SEQ ID NO:95)/Kb antibody. The cells were washed twice in 2 mL of FACS buffer (PBS, pH7.0, 1% BSA). The samples were then resuspended in about 250 μL of FACS buffer and peptide binding was analyzed via MFI. The data reveal that increasing levels of OVA peptide-MHC complex in these cells as higher concentrations of OVA-conjugated MPV.10.34.d IRCs were added. These data appear to establish that the MPV.10.34.d IRCs label target tumor cells by an extracellular mechanism.
232,284
11858965
DETAILED DESCRIPTION OF THE INVENTION The present disclosure can be more readily understood by reference to the following detailed description of various embodiments of the disclosure, the examples, and the drawings and tables with their relevant descriptions. It is to be understood that unless otherwise specifically indicated by the claims, the disclosure is not limited to specific preparation methods, carriers or formulations, or to particular modes of formulating the extract of the disclosure into products or compositions intended for topical, oral or parenteral administration, because as one of ordinary skill in the relevant arts is well aware, such things can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. As utilized in accordance with the present disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meaning: Often, ranges are expressed herein as from “about” one particular value and/or to “about” another particular value. When such a range is expressed, an embodiment includes the range from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the word “about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to and independently of the other endpoint. “Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, the phrase “optionally comprising an agent” means that the agent may or may not exist. It must be noted that, as used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Thus, unless otherwise required by context, singular terms shall include the plural and plural terms shall include the singular. The term “cellulosome” as used herein refers to a large bacterial extracellular multienzyme complex able to degrade crystalline cellulosic substrates. The term “complex” as used herein means a coordination or association of components linked by chemical or biological interaction. The term “recombinant polypeptide or “recombinant protein” can be used interchangable and refers to a polypeptide or protein produced by a host organism through the expression of a recombinant nucleic acid molecule, which has been introduced into said host organism or an ancestor thereof, and which comprises a sequence encoding said polypeptide or protein. The term “genetic engineering” as used herein refers to a process by which genetic materials, including DNA and/or RNA, are manipulated in a cell or introduced into a cell to affect expression of certain proteins in said cell. Manipulation may include introduction of a foreign (or “exogenous”) gene into the cell or inactivation or modification of an endogenous gene. Such a modified cell may be called a “genetically engineered cell” or a “genetically modified cell.” It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements. A cellulosome is a highly efficient cellulolytic enzyme complex. Naturally, it is produced byC thermocellumand other anaerobic cellulolytic bacteria. The entire scaffold and all enzymatic subunits of the cellulosome are attached to a cell surface through a surface anchoring protein with cohesin-dockerin interaction. However, the efficiency and enzyme synergism of the cellulosome highly depends on the number of cohesin-dockerin interaction. Accordingly, the present disclosure provides a recombinant protein comprising a plurality of type II cohesin repeats. In some embodiments of the disclosure, the recombinant protein comprises multiple tandem type II cohesin repeats that effectively and stably integrated in chromosome integration of a host cell, and is able to highly expressed and secreted. All references cited herein are incorporated in their entirety. Specifically, exemplary cohesins, dockerins and GPI useful in accordance with various aspects and embodiments of the present disclosure are described in the references incorporated herein. In some embodiments of the disclosure, the recombinant protein comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 type II cohesin repeats. In some embodiments of the disclosure, the recombinant protein is obtained by modifying surface anchoring protein comprising the type II cohesin repeats. Examples of the surface anchoring protein include but are limited to SdbA, Orf2p, or “outer layer protein B” (OlpB). Among the anchoring proteins, the OlpB typically includes seven Type II cohesins and can accommodate up to 63 cellulases in a cellulosome complex to form one of the largest known cellulolytic enzyme complexes. In some embodiments of the disclosure, the recombinant protein is derived fromClostridium thermocellum. In some embodiments of the disclosure, the recombinant protein further comprises an anchoring domain. Examples of the anchoring domain include but are not limited to a recombinant SLH anchoring domain or a recombinant glycosylphosphatidylinositol (GPI). For example, the SLH anchor domain is derived fromC. thermocellumOlpB; the GPI is a cell surface protein fromS. cerevisiae. The anchoring domain such as SLH anchoring domain or GPI in the recombinant protein is linked to a cell membrane-anchored protein. In some embodiments of the disclosure, the codon of the recombinant protein is optimized according to a codon system utilized by the host. In some embodiments of the disclosure, the recombinant protein is co-expressed with a recombinant cellulosome complex integrating protein A (CipA) comprising a plurality of type I cohesin repeats, a plurality of cellulose-binding modules (CBMs) and a type II dockerin. Accordingly, the present disclosure provides a recombinant cellulosome complex comprising:the recombinant protein mentioned above;a recombinant cellulosome complex integrating protein A comprising a plurality of type I cohesin repeats, a plurality of cellulose-binding modules and a type II dockerin; anda plurality of recombinant enzymes each comprising a type I dockerin. The CBM as used herein binds cellulosic substrates. A cellulosomal enzyme contains a type I dockerin, which interacts with the type I cohesin of CipA. The type II dockerin binds to the type II cohesin repeats in the recombinant protein. In some embodiments of the disclosure, the recombinant cellulosome complex is derived fromClostridium thermocellum. A typicalC. thermocellumcellulosome is comprised of a central non-enzymatic scaffold subunit known as “cellulosome integrating protein A” (CipA) with nine Type I cohesins (Kruus et al.Proceedings of the National Academy of Sciences92, 9254-9258 (1995)). In some embodiments of the disclosure, the recombinant cellulosome complex integrating protein A comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 type I cohesin repeats. In some embodiments of the disclosure, the recombinant cellulosome complex integrating protein A comprises 3, 6, or 9 type I cohesin repeats. In some embodiments of the disclosure, the recombinant cellulosome complex integrating protein A comprises 2, 3, 4, or 5 cellulose-binding modules. In some embodiments of the disclosure, the recombinant cellulosome complex integrating protein A comprises two cellulose-binding modules. It is believed, though not intended to be restricted by any theoretical, that the cohesin-dockerin interaction of a cellulosome facilitates spatial proximity among various cellulases (proximity effect) and the cellulose-binding capacity of CBM enhances cellulose utilization (targeting effect) (Blumer-Schuette et al.FEMS Microbiology Reviews38, 393-448 (2014)). These advantages provide clues for developing an efficient way to elevate the saccharification of cellulosic substrates (Schwarz,Applied Microbiology and Biotechnology56 (2001)). In some embodiments of the disclosure, the recombinant protein is co-expressed with a plurality of recombinant enzymes each comprising a type I dockerin. In some embodiments of the disclosure, the recombinant enzymes are cellulolytic enzymes. In some embodiments of the disclosure, the cellulolytic enzymes are endoglucanases, exoglucanases or β-glucosidases. In one some embodiment of the disclosure, the recombinant protein is co-expressed with a recombinant oxidative enzyme. In some embodiments of the disclosure, the recombinant oxidative enzyme is a lytic polysaccharide monooxygenase (LPMO). In some embodiments of the disclosure, the recombinant protein is co-expressed with a recombinant cellobiose dehydrogenase (CDH). It is believed, though not intended to be restricted by any theory, that the conversion of cellulose into simple sugars requires at least three types of enzymes: endoglucanases (EGs), exoglucanases (CBHs) and beta-glucosidases (BGSs) (Lambertz et al.Biotechnol Biofuels7, 135 (2014)). Apart from these hydrolases, recent studies have reported a new class of oxidative enzymes, called lytic polysaccharide monooxygenases (LPMO), which can efficiently degrade the crystalline cellulose and increase the soluble sugar released by 2.6-fold (Arfi et al.Proc Natl Acad Sci USA111, 9109-9114 (2014)). Hence, LPMO was named “cellulase booster”. It requires an enzyme-copartner, called cellobiose dehydrogenase (CDH), which donates the electron required by LPMO for its activity. Therefore, three types of fungal cellulases were selected—endoglucanase (TrEgIII), exoglucanase (CBHII) and β-glucosidase (NpaBGS), and also selected were the cellulase booster (TaLPMO) and its enzyme-copartner (MtCDH), which were then fused with Type I dockerin ofC. thermocellumto facilitate the cellulosomal integration. In one embodiment of the disclosure, adding LPMO and CDH to the cellulase mixture greatly increased the soluble sugar release. Yeast cells generally utilize the glucose released from hydrolysis, and the addition of methylglyoxal inhibits the host's glucose metabolism. With this method, the amount of glucose released during hydrolysis could be quantified readily. The glucose amount is low without the addition of methylglyoxal, due to the immediate uptake of glucose by the cells. In contrast, the co-culture of the enzyme host and the scaffold host accumulate a higher amount of glucose in the presence of methylglyoxal. These results confirm that the cellulosome has stronger cellulolytic activity on higher order celluloses, i.e., avicel. In some embodiments of the disclosure, to achieve efficient secretion and cellulosome assembly, the cellulosomal genes (i.e., CipA and OlpB) and dockerin-fused enzymes are engineered into two different hosts. The cell consortium approach was adopted and the ratio of each host was controlled according to the complexity of the cellulosic substrates to achieve a higher ethanol titer. In one embodiment of the disclosure, the recombinant protein is expressed by a cell. In some embodiments of the disclosure, the recombinant protein is expressed on the surface of the cell. In one embodiment, the cell is a yeast cell; in a further embodiment, the yeast cell isKluyveromyces marxianus. K. marxianushas several advantages overS. cerevisiaeand other yeast strains, including faster growth, higher thermotolerance, pentose sugar fermentation and higher secretion capacity (Chang et al.Biotechnology for Biofuels5, 53 (2012); Chang et al.Biotechnology for Biofuels6, 19 (2013); Chang et al.Applied Energy132, 465-474, (2014)). It is Crabtree-negative, thermotolerant (up to 46° C.) and capable of fermenting various sugars, including pentose sugars (i.e., xylose, arabinose, and inulin) (Fonseca et al.Applied Microbiology and Biotechnology79, 339-354 (2008); Lertwattanasakul et al.Biotechnology for Biofuels8, 47, (2015)).K. marxianusgrows faster thanS. cerevisiaeand other yeasts (Lee et al.Sci Rep8, 7305 (2018); Banat et al.World J Microbiol Biotechnol8, 259-263 (1992); Ho et al.Applied Energy100, 27-32, (2012)). Moreover, the secretory capacity ofK. marxianusis higher than that ofS. cerevisiaedue to its efficient signal peptides (Fonseca et al.Applied Microbiology and Biotechnology79, 339-354 (2008)). Finally,K. marxianusis a GRAS (generally regarded as safe) and QPS (qualified presumption of safety) certified organism (Spencer et al.Appl Microbiol Biotechnol58, 147-156 (2002)). These attributes makeK. marxianusan excellent host for industrial applications (Chang et al.Applied Energy132, 465-474 (2014)). In one embodiment of the disclosure, the recombinant protein, recombinant cellulosome complex, recombinant enzymes or recombinant oxidative enzyme are encoded by the genome of a cell. In one embodiment of the disclosure, the CipA gene (with 9 Type I cohesin repeats) and the largest OlpB gene (with 7 Type II cohesin repeats) are synthesized. The multiple repeats in the cohesins of CipA and OlpB genes make it extremely difficult for their cDNA cloning or even DNA synthesis. This problem was overcome by randomizing the codons in the repeats and synthesizing CipA and OlpB genes. In one embodiment of the disclosure, these two genes were then integrated into theK. marxianusgenome. In one embodiment, the recombinant cellulosome complex is expressed by a cell. In one embodiment, the recombinant cellulosome complex is expressed on a surface of the cell. In one embodiment of the disclosure, the cell is a yeast cell. In one embodiment, the yeast isKluyveromyces marxianus. The present disclosure provides a cell expressing the recombinant protein or the recombinant cellulosome complex described herein. In one embodiment of the disclosure, the cell expresses the recombinant protein or the recombinant cellulosome complex on the surface of the cell. In one embodiment, the cell is a yeast cell. In one embodiment, the yeast isKluyveromyces marxianus. The present disclosure provides a cell-culture comprising the cell described herein. The present disclosure provides a method for digesting a cellulose, comprising the cell, the recombinant protein, or the recombinant cellulosome complex described herein. In one embodiment of the disclosure, the cellulose is a microcrystalline cellulose. In one embodiment, the method is for converting the cellulose into reducing sugars and/or ethanol. The present disclosure also provides a method for producing ethanol, comprising the method described herein. The present disclosure provide exemplary methods and composition embodiments that overcame these difficulties by designing and synthesizing the scaffold gene (CipA) and the anchoring protein gene (OlpB) using synthetic biology techniques. The engineeredKluyveromyces marxianus, a probiotic yeast, secreted a cocktail of cellulolytic enzymes, including cellulases, a cellulase booster and its co-enzyme. It was shown that both the number of enzyme-binding sites and the number of cellulose-binding modules in the cellulosome improve its ethanol production rate. The largest cellulosome of the present disclosure can accommodate many enzymes, whereas previously the largest engineered cellulosome could accommodate only few enzymes and was in a plasmid, not in the yeast genome. The present engineered host released higher quantities of ethanol from cellulosic substrates than any previously constructed yeast cellulosome. The engineered cellulosomal yeast strains efficiently converted the microcrystalline cellulose into reducing sugars and/or ethanol and are suitable for consolidated bioprocessing (CBP). The performance of the present construct is not dramatically higher than that of conventional construct, but it used genome integration, instead of using an episomal plasmid, which has the advantage of increasing the gene copy number in the yeast cell, but it is not stable in the absence of selection markers. The following examples are provided to aid those skilled in the art in practicing the present disclosure. Examples Methods Strains and Media Escherichia colistrain DH5a was used as the host for plasmid construction and propagation.E. colistrain JM110 was used to demethylate plasmid and to ligate large DNA fragments.E. colistrains were cultured in Luria-Bertani (LB) medium (10 g/L tryptone, 5 g/L yeast extract, and 10 g/L NaCl) supplemented with 50 μg/mL ampicillin. YeastKluyveromyces marxianus4G5 strain was used to develop a CBP host, which was maintained in YPG medium (1% yeast extract, 2% peptone, 2% galactose) and cultured at 30° C. at 300 rpm. Yeast transformants were cultured in YPG media supplemented with either G418 or hygromycin B at a concentration of 200 μg/mL. Cellulosomal Construct Design and Synthesis Scaffolds (CipA) were designed in a biomimetic manner using theC. thermocellumCipA gene as a backbone, which has nine Type I cohesins with a CBM. The original CipA was named CipA 1B9C since it has one CBM (B) and nine cohesins (C). Similarly, synthetic scaffolds CipA 1B6C and CipA 1B3C were designed by removing the cohesins 6-9 and 4-9, respectively. The synthetic scaffold CipA 2B9C was designed by adding one more CBM between cohesin 7 and 8, while CBM was removed to form CipA 0B9C. The anchoring protein (OlpB) mimics theC. thermocellumOlpB, which contains seven Type II cohesins with a SLH anchoring domain and a long tandem repeat (TR). The SLH domain was replaced with the ScGPI domain and the TR domain was removed to avoid the synthesis constraints. All the constructs were codon optimized and difficult sequences were randomized using the Build Optimization Software Tools (BOOST) developed by DOE-Joint Genome Institute (JGI), USA (Oberortner et al.ACS Synthetic Biology6, 485-496 (2017)). All of the constructs were synthesized and inserted into the yeast integration plasmid pKLac2-α-Kan using AvrII and NotI. EgIII and NpaBGS genes were obtained from previous studies (Chang et al.Biotechnology for Biofuels5, 53 (2012); Chen et al.Biotechnology for Biofuels5, 24 (2012)). The CBHII (KC311337), LPMO and CDH genes were chemically synthesized by GenScript Inc. (Piscataway, NJ, USA) and codon-optimized forK. marxianusexpression (Chang et al.Biotechnology for Biofuels6, 19 (2013)). To fuse the C-terminal dockerin each cellulase gene was amplified with gene-specific primers using High-Fidelity DNA polymerases (New England Biolabs), digested with AvrII and SpeI and inserted into the yeast integration plasmid pKLac2-α-Kan-DocT-kit, which carries a Type I dockerin. Yeast Transformation and Clone Screening K. marxianuscells were cultured in 5 mL YPG medium at 30° C. at 300 rpm for 16 h. Yeast competent cells were prepared as described in Chang, et al. (Chang et al.Biotechnology for Biofuels5, 53 (2012)). SacII digested gene cassettes (1 μg) were electroporated (1.0 kV, 400Ω, and 25 μF capacitance) with 40 μl of competent cells, in a BioRad system (Gene Pulser Xcell™ Bio-Rad, Hercules, CA) with an aluminum cuvette (2 mm). Then, electroporated cells were recovered at 30° C. for 3 h and plated onto YPG plates supplemented with G418 or hygromycin B (200 μg/mL). Transformed yeast colonies were mixed with QuickExtract™ DNA Extraction Solution (EPICENTRE, Madison, Wisconsin) to remove the cell wall, and were used as a template for PCR verification. Chromosomal integration of each gene was confirmed by the PCR reaction using gene-specific primers and the stability was verified up to five generations. Cellulosome Assembly The complex formation of dockerin-fused cellulases and scaffolds were confirmed by native polyacrylamide gel electrophoresis (Native-PAGE). Briefly, culture supernatants of cellulases, scaffold and anchoring protein hosts were collected after 48 h cultivation at 37° C. at 300 rpm. Then, supernatants were condensed 50-fold using Vivaspin 20 (10-kDa cutoff, Merck Millipore, USA) and complex formation was performed by mixing each supernatant at different ratios, incubated for 2 h at 37° C. without shaking. After incubation, the enzyme mixture was loaded onto native-PAGE (stacking gel-5%; separating gel-8%) with 2×sample loading dye (without SDS). The same sample was heated at 100° C. and loaded onto 8% SDS-PAGE (Arfi et al.Proc Natl Acad Sci USA111, 9109-9114 (2014); Morais et al. Deconstruction of Lignocellulose into Soluble Sugars by Native and Designer Cellulosomes. mBio 3, (2012)). Immunofluorescence microscopy and FACS. Immunostaining was performed using the method described in Chang, et al. (Chang et al.Biotechnol Biofuels11, 157 (2018)). Yeast cells expressing anchoring protein was collected (A600of 1.0) and washed three times with PBS (pH 7.4). Cells were resuspended in 4% paraformaldehyde (PFA) and incubated for 30-60 min at 4° C. Then, cells were centrifuged and washed with PBS buffer to remove the excess PFA and resuspended in 500 μL of bovine serum albumin (BSA, 1 mg/mL in PBS) containing 0.5 μg of anti-FLAG primary antibody (Biotools, Taiwan) and incubated for 3 h. The cells were harvested by centrifugation and resuspended in 500 of PBS containing secondary antibody conjugated with DyLight™405 fluorophores (Jackson Immunoresearch, USA) and incubated for 1 h. After incubation, the cells were washed three times and 2 μL of cell suspension were fixed on a slide and observed under Leica TCS SP5 II confocal microscopy (Wetzlar, Germany). Similarly, immunostained yeast cells were subjected to FACS analysis to further confirm the anchoring efficiency. Approximately 50,000 cells were used for the analysis using MoFlo™ XDP Flow Cytometer (Beckman Coulter, Inc., CA, USA). Real-Time Quantitative PCR Total RNA was isolated from the transformants using AccuPure Yeast RNA mini Kit in the iColumn automated system (iColumn12, AccuBioMed, Taiwan). cDNA synthesis was conducted using a reverse transcription kit (SuperScript™ II kit, Invitrogen, USA). The relative quantification of each gene was carried out using gene-specific Universal Probe Library probes (LightCycler W480 Probes Master, Roche) with a specific primer pair (amplicon size around 100 to 150 bp) on a LightCycler (LightCycler 480, Roche). The actin gene was used as the reference gene and each analysis was repeated three times. The relative expression level was calculated using the 2−ΔΔCtmethod (Livak & Schmittgen.Methods25, 402-408 (2001)). Enzyme Assays Enzyme activity was assayed as described in Chang, et al. (Chang et al.Biotechnology and Bioengineering115, 751-761 (2018)). The activity of dockerin-fused cellulases was determined by various assays using specific substrates. Endo-glucanase (EgIII-t) activity was measured using dye-CMC (Megazyme, Wicklow, Ireland) as a substrate. The 100 μL assay reaction contains 40 μL of condensed supernatants with 60 μL of buffer solution (0.4% (w/v) dye-CMC, 50 mM sodium acetate, pH 5), incubated at 40° C. for 6 h. After incubation, 250 μL precipitation solution was added, centrifuged at 8000 rpm for 10 min and absorption was measured at 590 nm. Similarly, exo-glucanase (CBHII-t) activity was estimated using PASC. The assay was performed by adding 100 μL of supernatants with 900 μL buffer solution containing 50 mM sodium acetate (pH 5), 0.4% PASC and incubated in Thermal Shaker at 40° C. for 12 h. The released reducing sugars were measured by the dinitrosalicylic acid (DNS) method and sugar concentration was calculated using glucose standard (Ghose, T. K. inPure and Applied ChemistryVol. 59, 257 (1987)). β-glucosidase (NpaBGS-t) activity was determined by the formation of p-Nitrophenol from the hydrolysis of substrate p-Nitrophenyl-3-D-glucuronide (pNPG) and absorption was measured at 410 nm. Total cellulase activity was determined using filter paper as the substrate and the reducing sugar release was estimated by the DNS method. Fermentation and Ethanol Production The yeast cells expressing cellulases, scaffold and anchoring proteins were cultured and washed twice with YP medium (1% yeast extract, 2% peptone, 10 mM CaCl2)). Then, 20 O.D. cells (A600) were resuspended in YP media, supplemented with 0.042% Tween 80, 1% Avicel or PASC. In sugar release experiment, 100 mM methylglyoxal was added to the media to inhibit the glucose metabolism of the host. Fermentation was carried out in an anaerobic 50 mL serum tube with 5% volumetric headspace and incubated for several days at 37° C. with agitation at 300 rpm (Chang et al.Biotechnology for Biofuels6, 19 (2013)). The ethanol and glucose concentrations were analyzed using High-performance liquid chromatography (HPLC) (Jasco PU-2089 Quaternary HPLC PUMP, JASCO International Co., Ltd., Japan) coupled with the ICSep ICE-COREGEL 87H3 Column (Transgenomic, USA) and Shodex RI-101 Refractive Index Detector (RID) (ECOM, Czech Republic). Homology Modeling of Dockerin-Fused Enzymes To predict the three-dimensional structures of dockerin-fused enzymes, the protein sequence was blasted against the protein data bank (PDB) database. The online software Protein Homology/analogY Recognition Engine V 2.04 (Phyre2) (Kelley et al.Nature Protocols10, 845 (2015)) and RaptorX (Milberg et al.Nature Protocols7, 1511 (2012)) were used to predict the homology model of each enzyme. Bumps present in the predicted model were removed and missing side chains were added using the “WHAT IF Web Interface”. The predicted protein structure was validated using a protein structure validation (PSVS) tool. A Ramachandran plot was used to analyze the structure quality. The modeled protein structures were visualized and images were rendered using UCSF-Chimera software (Pettersen et al.J Comput Chem25, 1605-1612 (2004)). Statistical Analysis All the experiments were performed in triplicate and results were expressed as means±standard deviations (SD). Statistical significance was analysed using two-tailed student t-tests (with unequal variance) in Microsoft Excel 2016. P<0.05 was considered statistically significant throughout the study. Results Design and Synthesis of Cellulosomal Scaffolds Several components were designed and synthesized to engineer an entire cellulosome system into the yeast genome (see Methods). A synthetic scaffold was designed according to the sequence of the CipA gene (5.6 kb) inC. thermocellumATCC 27405 (CP000568). To evaluate the effect of cohesin number, three synthetic scaffolds containing 3, 6 and 9 cohesins with a single CBM (denoted 1B3C, 1B6C, and 1B9C) were designed. Similarly, to examine the effect of CBMs on cellulose-binding, two more synthetic scaffolds were designed with no or two CBMs (0B9C and 2B9C) (FIG.1a). An anchoring protein plays a critical role in the attachment of the cellulosome complex to the yeast cell surface. However, the anchoring domain ofC. thermocellumis not suitable for a eukaryotic host. Therefore, the anchoring efficiency of glycosylphosphatidylinositol (ScGPI), a cell surface protein fromS. cerevisiae, onK. marxianus(Kondo & Ueda.Applied Microbiology and Biotechnology64, 28-40 (2004)) was tested. The pKlac2 plasmid containing ScGPI fused with a green fluorescent protein (GFP) and a 12×His was expressed inK. marxianusand its anchoring efficiency was confirmed by fluorescence microscopic analysis. The imaging data showed a successful display of ScGPI on theK. marxianuscell surface. Then the largest cell surface scaffold was designed using OlpB as the backbone (see Methods). The seven Type II cohesins of OlpB were selected and the original anchoring domain (SLH) was replaced with the ScGPI domain to facilitate surface display. OlpB contains long tandem repeats (TRs) with highly repetitive TPSDEP (SEQ ID NO: 1) amino acid sequences (Lemaire et al.Journal of Bacteriology177, 2451-2459 (1995)). In view of the repetitiveness of the OlpB DNA sequences, the TR length was reduced and 7 cohesins were directly fused with the trimmed TRs and with the ScGPI domain (FIG.1c). For efficient translation inK. marxianus, both CipA and OlpB gene sequences were codon optimized forK. marxianusand the repeats in the cohesins were randomized to avoid DNA synthesis constraints. Conversion of Free Cellulases into Cellulosomal Mode As fungal cellulases have no dockerin, two kinds of dockerin fusion kits were designed according to the Type I dockerin ofC. thermocellum(DocT). Based on the domain organization of each enzyme, the dockerin module was fused at either the N- or C-terminus. A 36 bp glycine-rich linker was used between the dockerin module and the catalytic domain to avoid the conformational changes due to dockerin fusion (Arfi et al.Proc Natl Acad Sci USA111, 9109-9114 (2014)) and an 8×His tag was fused for purification and detection purposes (see Methods). For efficient secretion of each cellulase into the medium, the native secretory signal peptide (α-mating type) ofKluyveromyces lactiswas fused at the N-terminus. In the previous studies, several fungal cellulases inK. marxianus, including TrEgIII, CBHII and NpaBGS (Chang et al.Biotechnology for Biofuels6, 19 (2013); Chang et al.Biotechnology and Bioengineering115, 751-761 (2018)) were successfully expressed. In this study, along with cellulases, cellulase boosters LPMO (fromThermoascus aurantiacus, TaLPMO) and CDH (fromMyceliophthora thermophila, MtCDH) were also included. To convert the free fungal cellulases into the cellulosomal mode, DocT was fused at either the N- or C-terminus of TrEgIII, CBHII and NpaBGS. Surprisingly, CBHII and TrEgIII retained their activity after C-terminus dockerin fusion but not after N-terminus dockerin fusion. However, the enzyme activity was recovered after adding a ScafT. On the other hand, NpaBGS displayed similar amounts of enzyme activity in either N- or C-terminal dockerin fusion. Based on a previous study (Arfi et al.Proc Natl Acad Sci USA111, 9109-9114 (2014)), a dockerin was fused at the C-termini of TaLPMO and MtCDH, since the N-terminal histidine residue has been evolutionarily conserved in LPMO and is essential for its function. Three-dimensional protein structures of dockerin-fused chimeric enzymes were predicted using homology modeling, and the results showed that the dockerin fusion did not alter their substrate-binding pockets. Genome Integration of Cellulosomal Genes The cellulosomal genes were integrated into the Lac4 region of theK. marxianus4G5 genome using yeast integration plasmid pKLac2. To study the effect of cohesin numbers and CBMs on crystalline cellulose, five different CipA variants-CipA1B3C, 1B6C, 1B9C, 2B9C, and 0B9C-were transformed individually intoK. marxianus4G5 (see Online Methods). The chromosomal integration was confirmed by PCR verification, and their expression was confirmed by Western blot (WB) analysis using anti-His antibody. The WB results of CipA1B3C showed a distinctive band at ˜89 kDa. Similarly, transformants expressing 1B6C, 1B9C, 0B9C and 2B9C displayed bands at ˜142 kDa, ˜194 kDa, ˜172 kDa and ˜217 kDa, respectively. Finally, the anchoring host (AH) was developed by transforming OlpB-ScGPI into theK. marxianus4G5 genome, and their expression was confirmed by WB analysis and a band was observed at −211 kDa. Demonstration of OlpB Cell-Surface Display Using Immunofluorescence Analysis Anchoring of the entire OlpB-ScGPI on the cell surface was confirmed by immunofluorescence microscopy and Fluorescence-Activated Cell Sorting (FACS) analysis (see Methods). The yeast cells expressing OlpB protein exhibited a strong green fluorescence signal throughout the cell, making it hard to distinguish the protein anchored on the cell surface from the internal OlpB protein. Since the monoclonal antibodies did not penetrate the yeast cell wall, the yeast cells expressing OlpB protein were immuno-stained with DyeLight-405 secondary antibody conjugated with cyan fluorescence protein (CFP). A clear blue fluorescence was observed around the anchoring host, thus confirming that the OlpB protein was successfully displayed on theK. marxianuscell surface, whereas no fluorescence was observed on wild-type strains (FIG.2a). Similarly, FACS analysis was performed using 50,000 cells expressing the OlpB protein, among which 81.47% showed a fluorescence signal, further confirming the anchoring of OlpB on the cell surface (FIG.2b). Cellulosome Complex Formation The cellulosome complex assembly was demonstrated using the supernatants of CipA1B9C, BGS-t and cell extract of OlpB-ScGPI. They were allowed to form complexes at 37° C. and were analyzed using native- or SDS-PAGE followed by Western blot analysis. In native-PAGE WB, a shift in the band was observed and thus confirmed that the cellulosome complex had been formed (FIG.3a). A smear of the band in the BGS-t lane was observed, which might be due to the N-glycosylation of the DocT domain. As expected, the Type I dockerin of BGS-t bound randomly to the available nine Type I cohesins of CipA scaffold and shifted the band upward. Similarly, the Type II dockerins of CipA interacted with the Type II cohesins of OlpB-ScGPI and formed a complex. When mixing the three components together, a clear band was observed at the top of the lane, thus confirming the assembly of all three components to form cellulosomes. The cellulosome complex was dissociated by heating to 100° C., and separated bands were observed on SDS-PAGE (FIG.3b). These data confirm that the N-glycosylation of CipA and enzymes did not affect their ability to form cellulosomes. Constructing Enzyme Hosts A cellulase host (CH) was constructed by electroporating equimolar ratios of CBHII-t, TrEgIII-t, and NpaBGS-t genes intoK. marxianus4G5. The transformants were selected on YPG plates supplemented with hygromycin. The transformants integrated with all of the cellulase genes were selected by PCR screening, and their activity was confirmed by enzyme assays. Transformant CH-17 showed the highest enzyme activity and released 2.84 mM and 2.91 mM reducing sugars from avicel and filter paper, respectively. Similarly, to demonstrate the effects of the cellulase booster, TaLPMO-t and MtCDH-t genes were transformed intoK. marxianus4G5 (booster host: BH) and the booster functionality was confirmed by measuring the H2O2production using the Amplex red/HRP assay kit. The production of H2O2is directly proportional to the LPMO activity, which requires enzymatic (CDH together with cellobiose) or non-enzymatic (ascorbate) electron donors. Hence, the enzyme activity was assayed using two kinds of electron donors: cellobiose and ascorbate. The transformant BH-20 released 1.60 μM and 12.13 μM of H2O2using the substrate cellobiose and ascorbate, respectively. The lowest H2O2production was observed while using cellobiose as the substrate, and its concentration was increased in a time-dependent manner. In this case, CDH needs to oxidize the cellobiose and transfer an electron through its heme domain to LPMO (Phillips et al.ACS Chem Biol6, 1399-1406 (2011)). In contrast, ascorbate can directly donate the electron to LMPO, thereby producing higher quantities of H2O2in a short time. Boosting effects of cellulase boosters The selected booster host BH-20, which expresses TaLPMO-t and MtCDH-t, and the selected cellulase host CH-17, which expresses CBHII-t, TrEgIII-t, and NpaBGS-t, were cultured separately. Then the condensed supernatants of CH-17 and BH-20 were mixed at three different ratios (10:1, 10:2 and 10:3) and incubated along with avicel. The addition of CH-17 supernatant to the BH-20 supernatant at the ratio of 10:2 increased the soluble sugar release by 31%, confirming that LPMO has a proper function inK. marxianus. Similarly, the effects of cellulase boosters on the cellulosome complex were examined by mixing the supernatants of CH-17, BH-20 and CipA variants (including 1B3C, 1B6C, 1B9C, 0B9C, or 2B9C) in the molar ratio of 10:2:2 (CH:BH:CipA) and the enzyme mix was allowed to form the cellulosome complex at 37° C. Avicel was added to the cellulosome complex and reducing sugar release was measured. The results showed that the activity of the cellulase host improved by 32% (6.07 mM) with cellulase boosters, and the addition of CipA 1B6C or CipA 1B9C along with CH-17 increased the sugar release by 30% and 36%, respectively (FIG.4). Importantly, the addition of CH-17 and BH-20 supernatants together with either CipA 1B6C or 1B9C increased the sugar release by 51% and 70%, respectively. These results corroborate the effects of cellulase boosters on the cellulosome complex. Therefore, a new host was constructed, called Enzyme Host (EH), with three cellulases (TrEgIII-t, CBHII-t, and NpaBGS-t) and two cellulase boosters (TaLPMO-t and MtCDH-t). Transformants with all the integrated genes were selected, and their expression was confirmed by qPCR analysis using actin as a reference gene. Transformant EH-P1-44 demonstrated the highest enzyme activity and released 4.55 mM and 4.97 mM of reducing sugar from avicel and filter paper, respectively. Effects of cohesin and CBM numbers on cellulosome efficiency The effects of cohesin and CBM numbers on sugar release were assayed using avicel or phosphoric acid-swollen cellulose (PASC) as the substrate. The condensed supernatant of EH-P1-44 (expressing CBHII-t, TrEgIII-t, NpaBGS-t, TaLPMO-t and MtCDH-t) and the hosts expressing CipA variants were mixed at different ratios (10:1, 10:2 and 10:3). After the complex formation at 37° C., substrates were added to the mixture and sugar release was measured following 48 h incubation at 37° C. The data exhibited a tremendous increase in enzyme activity when the EH-P1-44 supernatant was mixed with CipA variants at the ratio of 10:2. CipA 2B9C exhibited the highest increase in sugar release, 118%, followed by 97%, 85% and 70% increases for CipA 1B9C, CipA 1B6C, and CipA 1B3C, respectively (FIG.5a). These data indicate that the cohesin number has a strong influence on sugar release, especially when using avicel as the substrate. However, when PASC was used as the substrate, the influence of the cohesin number was less significant (FIG.5b). The addition of CipA 1B9C, CipA 1B6C, or CipA 1B3C only increased 32%, 24%, or 24% in the sugar release compared to the activity of EH-P1-44 alone (FIG.5b). These results substantiate the vital role of the cohesin number in enzyme synergism, which is important for digesting complex substrates (i.e., microcrystalline cellulose or plant biomass). The number of CBMs also plays an important role in the efficiency of the cellulosome complex because they bring the entire complex towards the substrate. The data for sugar release from avicel showed that the construct with two CBMs (CipA 2B9C) remarkably increased the sugar release by 118%, followed by 97% for CipA with one CBM. Although CipA without any CBM increased the sugar release by 52%, it was the lowest among all the tested CipA variants (FIG.5a). However, the number of CBMs did not significantly improve the sugar release from PASC. The CipA 2B9C exhibited 38% increased sugar release, whereas CipA 1B9C and CipA 0B9C increased the sugar release by 32% and 28%, respectively (FIG.5b). These results confirm the significance of the CBM on a synthetic scaffold for microcrystalline cellulose degradation. Demonstration of consolidated bioprocessing Consolidated bioprocessing (CBP) is a single-process approach that includes the production of cellulase enzymes, biomass saccharification and fermentation, and could be an ideal approach to cellulosic ethanol production. The hydrolytic activity of cellulosome on cellulosic substrates was demonstrated with and without methylglyoxal, a glucose metabolism inhibitor. Cells were cultured in YP containing 1% avicel or PASC supplemented with or without 100 mM methylglyoxal for 6 h. The lowest amount of glucose was observed in all conditions in the absence of methylglyoxal (FIG.6a). The concentration of glucose increased tremendously in the presence of 100 mM methylglyoxal. Notably, EH-P1-44 along with Scaffold Host (SH) released a higher amount of glucose using avicel than PASC as the substrate. This confirms the above observation that the scaffold increased the cellulolytic activity on microcrystalline cellulose. For efficient secretion and assembly of the cellulosomal protein complex, the EH-P1-44 host and the SH-1B9C host expressing CipA 1B9C and OlpB were co-cultured in different ratios, and their ethanol-producing ability was analyzed using avicel. The direct ethanol fermentation was conducted under a microaerobic condition to provide the oxygen required for LPMO activity. The microaerobic condition was achieved by culturing the cells with 5% headspace in 50 ml serum bottles. The maximal ethanol production (2.68 g/L) was achieved on avicel when the EH-P1-44 and SH-1B9C hosts were co-cultured at the ratio of 10:1. The cellulosic ethanol production rates of SH-CipA variants (1B3C, 1B6C, 0B9C, 1B9C and 2B9C) along with EH-P1-44 were investigated to show the influences of CBM and cohesin numbers on enzyme-substrate-microbe complex synergism. As shown inFIG.6b, when using avicel as the substrate, the maximum ethanol production (3.09 g/L) was achieved in the consortium that contained EH-P1-44 and SH-2B9C. In comparison, EH-P1-44+SH-1B9C and EH-P1-44+SH-0B9C produced 2.86 g/L and 1.37 g/L of ethanol, respectively. Cellulosome hosts expressing 1B3C and 1B6C produced 1.93 and 2.29 g/L of ethanol, which are higher than the full scaffold without the CBM (0B9C). These observations indicate that the number of CBMs had a strong influence on ethanol production using microcrystalline cellulose. In contrast, while using PASC as the substrate, EH-P1-44+SH-1B9C and EH-P1-44+SH-2B9C produced similar quantities of ethanol, i.e., 8.25 g/L and 8.49 g/L, respectively (FIG.6c). Interestingly, when PASC was used as the substrate, the ethanol titer of the consortium containing SH-0B9C cells (lacking CBM) was higher (6.95 g/L) than the cellulosomes containing SH-1B3C and SH-1B6C (5.81 g/L and 6.91 g/L, respectively). In terms of ethanol production, the mini-cellulosome engineered by Fan et al. (Natl Acad Sci USA109, 13260-13265 (2012)), which could display up to 12 enzymes, produced 1.41 g/L and 1.09 g/L of ethanol from avicel and PASC, respectively. By including the LPMO and CDH genes in the cellulosome, Liang et al. (Appl Environ Microbiol80, 6677-6684 (2014)) improved the ethanol titer to 1.5 g/L and 2.7 g/L for avicel and PASC, respectively. Compared to Liang et al.“s study, the present disclosure achieved two-fold higher ethanol production (3.09 g/L) using avicel and three-fold higher ethanol production (8.61 g/L) using PASC as the sole carbon source (FIGS.6band6c). While the present disclosure has been described in conjunction with the specific embodiments set forth above, many alternatives thereto and modifications and variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are regarded as falling within the scope of the present disclosure.
43,782
11858966
DETAILED DESCRIPTION Provided herein are mutantMycobacterium smegmatisporins (Msp). A mutant Msp can be a multimer complex comprised of two or more Msp monomers, wherein at least one of the monomers is a mutant Msp monomer. An Msp monomer is encoded by a gene inMycobacterium smegmatis. Mycobacterium smegmatishas four identified Msp genes, denoted MspA, MspB, MspC, and MspD. An alignment of the wild-type polypeptide sequences for the MspA, MspB, MspC and MspD monomers ofMycobacterium smegmatisis shown inFIG.1. The numbering of each protein starts with the first amino acid of the mature portion of the sequence, as indicated by the number “1” above the first amino acid of the mature amino acid sequence. The amino acid sequences for a MspA, MspB, MspC and a MspD monomer without a signal sequence, i.e., the mature portion of the sequence, are provided as SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3 and SEQ ID NO: 4, respectively. The amino acid sequences for a MspA, MspB, MspC and a MspD monomer with a signal/leader sequence are provided as SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8 and SEQ ID NO: 9, respectively. Further, sequences of wild-type Msp monomers that can be modified are disclosed in GenBank, and these sequences and others are herein incorporated by reference in their entireties as are individual subsequences or fragments contained therein. For example, the nucleotide and amino acid sequences of a wild-type MspA monomer can be found at GenBank Accession Nos. AJ001442 and CAB56052, respectively. The nucleotide and amino acid sequences of a wild-type MspB monomer can be found, for example, at GenBank Accession Nos. NC_008596.1 (from nucleotide 600086 to 600730) and YP 884932.1, respectively. The nucleotide and amino acid sequences of a wild-type MspC monomer can be found, for example, at GenBank Accession Nos. AJ299735 and CAC82509, respectively. The nucleotide and amino acid sequences of a wild-type MspD monomer can be found, for example, at GenBank Accession Nos. AJ300774 and CAC83628, respectively. A mutant Msp monomer can be a full-length monomer or a functional fragment thereof encoded by a MspA, MspB, MspC or MspD-encoding nucleic acid, for example, an mRNA or a genomic sequence encoding MspA, MspB, MspC or MspD, wherein the monomer comprises one or more modifications. Optionally, a mutant Msp is a mutant single-chain Msp or is a multimer of several single-chain Msps, wherein the multimer comprises at least one mutant single-chain Msp. A mutant Msp can also be a multimer of several Msp monomers wherein at least one Msp monomer is a mutant Msp monomer. A single-chain Msp can, for example, comprise a multimer formed by two or more Msp monomers (e.g., eight monomers) connected by one or more amino acid linker peptides. A partial single-chain Msp refers to a single-chain multimer complex that dimerizes, trimerizes, or the like to form a porin. A full single-chain Msp porin refers to a single-chain multimer complex that forms a porin without the need to dimerize, trimerize or the like to form a porin. Stated differently, the single-chain folds to form a porin, but all components are in one amino acid chain, as compared to a porin that must associate with other partial single-chain Msp(s) or monomeric Msp monomers to form a porin. Mutant Single-Chain Msps and the Nucleic Acids Encoding them Provided herein are nucleic acid sequences encoding mutant single-chain Msps. For example, the nucleic acid sequence encoding a mutant single-chain Msp comprises: (a) a first and second nucleotide sequence, wherein the first nucleotide sequence encodes a first Msp monomer sequence and the second nucleotide sequence encodes a second Msp monomer sequence; and (b) a third nucleotide sequence encoding an amino acid linker sequence, wherein at least one of the first and second Msp monomer sequences is a mutant Msp monomer sequence has a mutation at position P97. Optionally, the mutant Msp monomer sequence can comprise a mutation at P97, wherein the mutation is not a P97S mutation or a P97C mutation. Optionally, the mutant Msp monomer sequence can comprise a P97F mutation. As shown in the Examples, additional hydrophobic residues, for example, phenylalanine, located in loop 6 of scMspA (amino acids 91-103) promote faster and more efficient insertion of the pores into lipid bilayers. For a description of loop 6 of MspA and residues contained therein, see Huffe et al.,J. Biol. Chem.284: 10223-10231 (2009), which is hereby incorporated in its entirety by this reference. Therefore, provided herein is a single chain Msp comprising one or more hydrophobic substitutions in loop 6 (amino acids 91-103) of Msp. For example, provided herein is a nucleic acid sequence encoding a mutant single-chainMycobacterium smegmatisporin (Msp), wherein the nucleic acid sequence comprises (a) a first and second nucleotide sequence, wherein the first nucleotide sequence encodes a first Msp monomer sequence and the second nucleotide sequence encodes a second Msp monomer sequence and (b) a third nucleotide sequence encoding an amino acid linker sequence, wherein at least one of the first and second Msp monomer sequences is a mutant Msp monomer sequence comprising one or more mutations at any of amino acid positions D91, G92, D93, 194, T95, A96, P97, P98, F99, G100, L101, N102 or S103, wherein one or more of D91, G92, D93, 194, T95, A96, P97, P98, F99, G100, L101, N102 or S103 is substituted with a hydrophobic amino acid. For example, hydrophobic amino acids can be selected from the group consisting of alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tyrosine, tryptophan, proline and glycine. By way of example, and not to be limiting, a mutant Msp monomer sequence can comprise (i) a D90N, a D91N and a D93N mutation; and one or more of (ii) a G92F, T95F, A96F, P97F, P98F, G100F, L101F, N102F or S103F mutation. As set forth above, substitutions at position G92, T95, A96, P97, P98, G100, L101, N102 or S103 are not limited to phenylalanine, as one or more of these amino acids can be replaced with another hydrophobic residue, for example, alanine, valine, leucine, isoleucine, proline, methionine, tyrosine, tryptophan, proline and glycine. One or more of G92, T95, A96, P97, P98, G100, L101, N102 or S103 can be substituted with the same hydrophobic amino acid or different hydrophobic amino acids. As used throughout, a mutation at a specific amino acid is indicated by the single letter code for the amino acid at a position, followed by the number of the amino acid position in an Msp polypeptide sequence (for example, an amino acid position in SEQ ID NO: 1), and the single letter code for the amino acid substitution at this position. Therefore, it is understood that a P97 mutation is a proline to phenylalanine substitution at amino acid 97 of SEQ ID NO: 1. Similarly, a D90N mutation is an aspartic acid to arginine substitution at amino acid 90 of SEQ ID NO: 1, a D91N mutation is an aspartic to arginine substitution at amino acid 91 of SEQ ID NO: 1, etc. It is also understood that amino acids corresponding to positions in SEQ ID NO: 1 are also provided herein (SeeFIG.1). For example, and not to be limiting, one of skill in the art would understand that, the corresponding amino acid for E139 of SEQ ID NO: 1 in MspB (SEQ ID NO:2), MspC (SEQ ID NO: 3) and MspD (SEQ ID NO: 4) is A139, A139 and K138, respectively. Optionally, any mutant Msp monomer sequence described herein can further comprise a mutation at amino acid position D118, a mutation at position D134 or a mutation at position E139. Optionally, a mutation at position E139 can be an E to R (arginine) or an E to K (lysine) substitution. Optionally, a mutation at position D118 can be a D to R substitution or a D to K substitution. Optionally, a mutation at position D134 can be a D to R substitution or a D to K substitution. For example, any mutant Msp monomer sequence described herein can comprise one or more mutations selected from the group consisting of: a D118R mutation, a D134R mutation and a E139K mutation. Optionally, any mutant Msp monomer sequence described herein can further comprise at least one of (i) a mutation at position 93 and (ii) a mutation at position D90, position D91 or both positions D90 and D91. Optionally, the amino acid at position 90, 91 or 93 is substituted with arginine, lysine, histidine, glutamine, methionine, threonine, phenylalanine, tyrosine or tryptophan. Optionally, any mutant Msp monomer sequence described herein can further comprise a D90N, a D91N and a D93N mutation. For example, a mutant Msp monomer sequence comprising a mutation at position 97 can further comprise (i) a mutation at amino acid position D118, D134 and/or E139 (ii) a mutation at position D93, and/or (iii) a mutation at position D90, position D91 or both positions D90 and D91. For example, a mutant MspA monomer sequence can comprise a D90N mutation, a D91N mutation, a D93N mutation, a P97F mutation, a D118R mutation, a D134R mutation and a E139K mutation. The mutant MspA monomer sequence can also comprise a D90N mutation, a D91N mutation, a D93N mutation, a P97F mutation, a D118R mutation, a D134R mutation and a E139K mutation. Also provided herein is a nucleic acid sequence encoding a mutant single-chain Msp which comprises (a) a first and second nucleotide sequence, wherein the first nucleotide sequence encodes a first Msp monomer sequence and the second nucleotide sequence encodes a second Msp monomer sequence, and(b) a third nucleotide sequence encoding an amino acid linker sequence, wherein at least one of the first and second Msp monomer sequences is a mutant Msp monomer sequence comprising one or more mutations at any of the following amino acid positions: I68, S73, S116, P123 or V128. Provided herein is a nucleic acid encoding a mutant single-chain Msp, wherein the mutant Msp monomer sequence comprises one or more mutations at any of I68, S73, S116, P123 or V128, wherein the mutation is not a I68V mutation or a S73C mutation. Provided herein is a mutant Msp monomer sequence comprising one or more mutations at I68, S73, S116, P123 and/or V128 further comprising (i) a mutation at amino acid position D118, (ii) a mutation at position D93, and/or (iii) a mutation at position D90, position D91 or both positions D90 and D91. For example, a mutant MspA monomer sequence can comprise one or more mutations at amino acid positions I68, S73, S116, P123 or V128, a mutation at D93, a mutation at D118, a mutation at D134 and a mutation at E139 or any subset thereof. In another example, a mutant MspA monomer sequence can comprise one or more mutations at amino acid positions I68, S73, S116, P123 or V128, a mutation at amino acid position D118, a mutation at D134, a D90N mutation and/or a D91N mutation. In yet another example, a mutant MspA monomer sequence can comprise one or more mutations at amino acid positions I68, S73, S116, P123 or V128, a mutation at amino acid position D118, a mutation at D134 and a mutation at E139, a D90N mutation, a D91N mutation and a D93N mutation. Provided herein is a nucleic acid encoding a mutant single-chain Msp, wherein the mutant Msp monomer sequence comprises one or more mutations at any of I68, S73, S116, P123 or V128, wherein the mutation is not a I68V mutation or a S73C mutation. In any of the mutant single-chain Msps provided herein, the mutant Msp monomer sequence can comprise one or more mutations at any of I68, S73, S116, P123 or V128, wherein the mutation is not a I68V mutation or a S73C mutation. Optionally, any of the mutant Msp monomer sequences described herein can further comprise one or more mutations at any of the following amino acid positions: D13, A55, D56, E57, F58, E63, S136, G137 or D172. Optionally, one or more of D13, A55, D56, E57, F58, E63, S136, G137 or D172 in a mutant Msp monomer sequence provided herein can be substituted with lysine or arginine. Optionally, any mutant Msp monomer sequence described herein comprising one or more mutations at D13, A55, D56, E57, F58, E63, S136, G137 or D172 can further comprise one or more mutations at the following positions: D118, D134 or E139. Optionally, any mutant Msp monomer sequence described herein comprising one or more mutations at D13, A55, D56, E57, F58, E63, S136, G137 or D172 can further comprise a mutation at position 93, and/or a mutation at position 90, position 91 or both positions 90 and 91. Therefore, provided herein is a nucleic acid sequence encoding a mutant single-chainMycobacterium smegmatisporin (Msp), wherein the nucleic acid sequence comprises (a) a first and second nucleotide sequence, wherein the first nucleotide sequence encodes a first Msp monomer sequence and the second nucleotide sequence encodes a second Msp monomer sequence, and (b) a third nucleotide sequence encoding an amino acid linker sequence, wherein at least one of the first and second Msp monomer sequences is a mutant Msp monomer sequence comprising one or more mutations at any of the following amino acid positions: D13, A55, D56, E57, F58, E63, S136, G137 or D172. Optionally, the third nucleotide sequence encoding the linker is located between the first and second nucleotide sequence. Also provided is a nucleic acid sequence encoding a mutant single-chainMycobacterium smegmatisporin (Msp), wherein the nucleic acid sequence comprises: (a) a first and second nucleotide sequence, wherein the first nucleotide sequence encodes a first Msp monomer sequence and the second nucleotide sequence encodes a second Msp monomer sequence, and (b) a third nucleotide sequence encoding an amino acid linker sequence, wherein at least one of the first and second Msp monomer sequences is a mutant Msp monomer sequence comprising (i) a mutation at position 93, and/or (ii) a mutation at position 90, position 91 or both positions 90 and 91 and (iii) one or more mutations at any of the following amino acid positions: D13, A55, D56, E57, F58, E63, S136, G137 or D172. Further provided is a nucleic acid encoding a mutant Msp monomer, wherein the Msp monomer comprises a mutation at one or more of the following positions: D13, A55, D56, E57, F58, E63, S136, G137 or D172. Optionally, a mutant Msp monomer sequence comprising one or more mutations at D13, A55, D56, E57, F58, E63, S136, G137 or D172 can further comprise a mutation at position 93 and/or a mutation at position 90, position 91 or both positions 90 and 91. Optionally, a mutant Msp monomer sequence comprising one or more mutations at positions D13, A55, D56, E57, F58, E63, S136, G137 or D172 can further comprise a D90N, a D91N and a D93N mutation. In the mutant single-chain mutant Msps provided herein, the first monomer sequence can be any mutant monomer sequence described herein. For example, the mutant monomer sequence can be a mutant MspA sequence. The second monomer can be selected from the group consisting of a wildtype Msp monomer, a second mutant Msp monomer, a wild-type Msp paralog or homolog monomer, and a mutant Msp paralog or homolog monomer. It is understood that the second mutant Msp monomer can be the same or different than the first mutant Msp monomer. These include, but are not limited to, MspA/Msmeg0965, MspB/Msmeg0520, MspC/Msmeg5483, MspD/Msmeg6057, MppA, PorM1, PorM2, PorM1, Mmcs4296, Mmcs4297, Mmcs3857, Mmcs4382, Mmcs4383, Mjls3843, Mj1s3857, Mjls3931 Mjls4674, Mjls4675, Mjls4677, Map3123c, Mav3943, Mvan1836, Mvan4117, Mvan4839, Mvan4840, Mvan5016, Mvan5017, Mvan5768, MUL 2391, Mflv1734, Mflv1735, Mflv2295, Mflv1891, MCH4691c, MCH4689c, MCH4690c, MAB1080, MAB1081, MAB2800, RHA1 ro08561, RHA1 ro04074, and RHA1 ro03127. A wild-type MspA paralog or homolog monomer may be a wild-type MspB monomer. Wild-type MspA paralog and homolog monomers are well-known in the art. Table 1 provides a non-limiting list of such paralogs and homologs. TABLE 1Wild-type MspA and Wild-type MspA paralogs and homolog monomersIdentity/Similarityto MspALengthProtein#Organism(%)(aa)ReferenceMspA/Msmeg0965M. smegmatis100/100211gb|ABK74363.1|, (Stahlet al., 2001)*MspB/Msmeg0520M. smegmatis94/95215gb|ABK73437.1|, (Stahlet al., 2001)*MspC/Msmeg5483M. smegmatis93/95215gb|ABK74976.1|, (Stahlet al., 2001)*MspD/Msmeg6057M. smegmatis82/89207gb|ABK72453.1|, (Stahlet al., 2001)*MppAM. phlei100/100211AJ812030, (Dorner et at.,2004)**PorM1M. fortuitum95/96211emb|CAI54228.1|PorM2M. fortuitum91/93215emb|CAL29811.1|PorM1M. peregrinum94/96211emb|CAI54230.1|Mmcs4296Mycobacteriumsp. MCS85/91216gb|ABG10401.1|Mmcs4297Mycobacteriumsp. MCS85/91216gb|ABG10402.1|Mmcs3857Mycobacteriumsp. MCS30/44235gb|ABG09962.1|Mmcs4382Mycobacteriumsp. MCS85/91216gb|ABL93573.1|Mmcs4383Mycobacteriumsp. MCS85/91216gb|ABL93574.1|Mjls3843Mycobacteriumsp. JLS26/40235gb|ABN99619.1|Mjls3857Mycobacteriumsp. JLS26/40235gb|ABG09962.1|Mjls3931Mycobacteriumsp. JLS26/40235gb|ABL93123.1|Mjls4674Mycobacteriumsp. JLS85/89216gb|ABO00440.1|Mjls4675Mycobacteriumsp. JLS83/89216gb|ABO00441.1|Mjls4677Mycobacteriumsp. JLS84/89216gb|ABO00443.1|Map3123cM. avium24/39220gb|AAS05671.1|paratuberculosisMav3943M. avium24/39227gb|ABK66660.1|Mvan1836M. vanbaaleniiPYR-182/88209gb|ABM12657.1|Mvan4117M. vanbaaleniiPYR-132/43239gb|ABM14894.1|Mvan4839M. vanbaaleniiPYR-183/88209gb|ABM15612.1|Mvan4840M. vanbaaleniiPYR-183/89209gb|ABM15613.1|Mvan5016M. vanbaaleniiPYR-130/41238gb|ABM15788.1|Mvan5017M. vanbaaleniiPYR-125/35227gb|ABM15789.1|Mvan5768M. vanbaaleniiPYR-121/32216gb|ABM16533.1|MUL_2391M. ulceransAgy9921/34233gb|ABL04749.1|Mflv1734M. gilvumPYR-GCK21/32225gb|ABP44214.1|Mflv1735M. gilvumPYR-GCK32/41226gb|ABP44215.1|Mflv2295M. gilvumPYR-GCK25/40250gb|ABP44773.1|Mflv1891M. gilvumPYR-GCK84/90217gb|ABP44371.1|MCH4691cM. chelonae70/80223gb|ACV04474.1|MCH4689cM. chelonae66/78223gb|ACV04472.1|MCH4690cM. chelonae72/81217gb|ACV04473.1|MAB1080M. abscessus69/79223emb|CAM61170.1|MAB1081M. abscessus68/78222emb|CAM61171.1|MAB2800M. abscessus27/44246emb|CAM62879.1|RHAl ro08561Rhodococcus jostii34/51233gb|ABG99605.1|RHA1n.d.Rhodococcus opacusB434/51233gbj|BAH52196.1|RHA1 ro04074Rhodococcussp. RHA134/50233gb|ABG95871.1|RHA1 ro03127Rhodococcussp. RHA134/50233gb|ABG94930.1|n.d.Rhodococcus35/50229gbj|BAH30938.1|erythropolisPR4Only proteins with significant amino acid similarities over the full length of the protein were included. Data were obtained by PSI-Blast algorithm (BLOSUM62 matrix) using the NIH GenBank database on the world wide web at ncbi.nlm.nih.gov/blast/Blast.cgi.n.d.: “not determined”*Stahl etal., Mol. Microbial. 40:451 (2001)**Domer et al., Biochim. Biophys. Acta. 1667:47-55 (2004) As used herein, a mutant single-chain Msp is a polypeptide comprising at least two Msp monomers, or functional fragments thereof, connected by one or more amino acid linker peptides wherein at least one of the Msp monomers is a mutant Msp monomer. For example, the mutant single-chain Msp can comprise two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, or more Msp monomers connected by one or more amino acid linker peptides, wherein at least one of the Msp monomers is a mutant Msp monomer. As set forth above, a single-chain mutant Msp can form a porin, for example, by folding, without the need to dimerize, trimerize or the like to form a porin. Alternatively, a mutant single-chain Msp can be a partial single-chain mutant Msp comprising at least two Msp monomers or fragments thereof connected by one or more amino acid linked peptides, that can dimerize, trimerize or the like to form a porin. Optionally, a Msp porin comprising a mutant single-chain Msp can, for example, comprise two or more single-chain Msp porin dimers, two or more single-chain Msp porin trimers, two or more single-chain Msp porin quadrimers, two or more single-chain Msp porin pentamers, one or more single-chain Msp porin hexamers, one or more single-chain Msp porin septamers, one or more single-chain Msp porin octamers, or combinations thereof. For example, a Msp porin can comprise a single-chain Msp porin dimer and two single-chain Msp porin trimers. By way of another example, a Msp porin can comprise a single-chain Msp porin quadrimer and two single-chain Msp porin dimers. Amino acid linker sequences are described herein. In any single-chain Msp described herein, a linker sequence can, for example, comprise 10 to 20 amino acids. For example, an amino acid linker sequence comprises 15 amino acids. Optionally, the amino acid linker sequence comprises a (GGGGS)3(SEQ ID NO: 5) peptide sequence. The same or different nucleic acid encoding linker sequence can be provided between nucleic acid sequences encoding more than two Msp monomers. Optionally, a linker sequence can be provided between all or some of the nucleic acid sequences encoding Msp monomers in the single chain Msps provided herein. Further provided is a nucleic acid sequence encoding a mutant single-chain Msp, wherein the nucleic acid sequence comprises (a) a first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequence or any subset thereof, wherein the first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequences encode a first, second, third, fourth, fifth, sixth, seventh, and eighth Msp monomer sequence, respectively; and (b) a ninth nucleotide sequence encoding an amino acid linker sequence, wherein the first Msp monomer sequence is a mutant Msp monomer sequence that comprises a mutation at position P97. The mutant Msp monomer sequence can comprise a mutation at P97, wherein the mutation is not a P97S mutation or a P97C mutation. The mutant Msp monomer sequence can comprise a P97F mutation. As set forth above, any mutant Msp monomer sequence described herein can further comprise a mutation at amino acid position D118, a mutation at position D134 or a mutation at position E139. For example, any mutant Msp monomer sequence described herein can comprise a D118R mutation, a D134R mutation and/or a E139K mutation. Any mutant Msp monomer sequence described herein can further comprise (i) a mutation at position 93 and/or (ii) a mutation at position D90, position D91 or both positions D90 and D91. Optionally, the amino acid at position 90, 91 or 93 is substituted with arginine, lysine, histidine, glutamine, methionine, threonine, phenylalanine, tyrosine or tryptophan. Any mutant Msp monomer sequence described herein can further comprise a D90N, a D91N and a D93N mutation. For example, provided herein is a nucleic acid sequence encoding a mutant single-chain Msp, wherein the nucleic acid sequence comprises (a) a first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequence or any subset thereof, wherein the first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequences encode a first, second, third, fourth, fifth, sixth, seventh, and eighth Msp monomer sequence, respectively; and (b) a ninth nucleotide sequence encoding an amino acid linker sequence, wherein the first Msp monomer sequence is a mutant Msp monomer sequence that comprises a mutation at position P97 can further comprise (i) a mutation at amino acid position D118, D134 and/or E139 (ii) a mutation at position D93, and/or (iii) a mutation at position D90, position D91 or both positions D90 and D91. For example, the first Msp monomer sequence can be a mutant Msp monomer sequence that comprises a D90N mutation, a D91N mutation, a D93N mutation, a P97F mutation, a D118R mutation, a D134R mutation and a E139K mutation. Further provided is a nucleic acid sequence encoding a mutant single-chain Msp, wherein the nucleic acid sequence comprises (a) a first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequence or any subset thereof, wherein the first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequences encode a first, second, third, fourth, fifth, sixth, seventh, and eighth Msp monomer sequence, respectively, and (b) a ninth nucleotide sequence encoding an amino acid linker sequence, wherein the first Msp monomer sequence is a mutant Msp monomer sequence that comprises one or more mutations at any of the following amino acid positions: I68, S73, S116, P123 or V128. The first Msp monomer sequence can also be a mutant Msp monomer sequence that comprises one or more mutations at any of the following amino acid positions: I68, S73, S116, P123 or V128 and further comprises a mutation at amino acid position D118, optionally with (i) a mutation at position 93, and/or (ii) a mutation at position D90, position D91 or both positions D90 and D91. In any of the mutant Msp monomer sequences described herein, the amino acid at position 91 or the amino acid at position 90 can be substituted with arginine, lysine, histidine, glutamine, methionine, threonine, phenylalanine, tyrosine or tryptophan. The mutant Msp monomer sequence can further comprise a D90N, a D91N and a D93N mutation. The mutant Msp monomer comprising one or more mutations at amino acids I68, S73, S116, P123 or V128 can further comprise a mutation in one or more of the amino acids at positions D13, A55, D56, E57, F58, E63, S136, D134, G137, E139 or D172. In the mutant Msp monomer sequences described herein, D13, A55, D56, E57, F58, E63, S136, D134, G137, E139 or D172 can be substituted with lysine or arginine. Therefore, a mutant Msp monomer comprising one or more mutations at amino acids I68, S73, S116, P123 or V128, for example, can further comprise (i) a mutation at amino acid position D118, (ii) a mutation at position D93, (iii) a mutation at position D90, position D91 or both positions D90 and D91, (iv) a D90N, a D91N and a D93N mutation and/or (v) a mutation in one or more of the amino acids at positions D13, A55, D56, E57, F58, E63, S136, D134, G137, E139 or D172. For example, and not to be limiting, a first mutant Msp monomer can be a mutant Msp monomer comprising a mutation at positions D56, I68, S73, D118, D134 and E139. Optionally, the mutant Msp monomer can further comprise a D90N, a D91N and D93N mutation. Optionally, one or more of the amino acids selected from the group consisting of D56, I68, S73, D118, D134 and E139 can be substituted with lysine or arginine. Further provided is a nucleic acid sequence encoding a mutant single-chain Msp, wherein the nucleic acid sequence comprises: (a) a first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequence or any subset thereof, wherein the first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequences encode a first, second, third, fourth, fifth, sixth, seventh, and eighth Msp monomer sequence, respectively; and (b) a ninth nucleotide sequence encoding an amino acid linker sequence, wherein the first Msp monomer sequence is a mutant Msp monomer sequence that comprises one or more mutations at any of the following amino acid positions: I68, S73, S116, P123 or V128; and wherein one or more of the first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequence encodes a mutant Msp monomer sequence comprising a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. For example, and not to be limiting, the seventh nucleotide can encode a mutant Msp monomer sequence comprising a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. It is understood that the first nucleotide sequence and the seventh nucleotide sequence can be arranged, but are not necessarily arranged as the first nucleotide sequence and the seventh nucleotide sequence in the nucleic acid sequence that comprises a first, second, third, fourth, fifth, sixth, seventh, and eighth nucleotide sequence in that order. In this context, the first nucleotide sequence is a nucleotide sequence encoding the first or starting monomer of a single-chain Msp, and can be the first, second, third, fourth, fifth, sixth, seventh, or eighth nucleotide sequence of the single-chain Msp. The starting nucleotide sequence is referred to as the first nucleotide sequence no matter where it occurs in the single-chain Msp. For example, if the starting subunit of the single-chain Msp is the first Msp monomer (first nucleotide sequence), then the seventh Msp monomer (seventh nucleotide sequence) comprises a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. In another example, if the starting subunit of the single-chain Msp is the second Msp monomer (first nucleotide sequence), then the eighth Msp monomer (seventh nucleotide sequence) comprises a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. In another example, if the starting subunit of the single-chain Msp is the third Msp monomer (first nucleotide sequence), then the first Msp monomer (seventh nucleotide sequence) comprises a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. In another example, if the starting subunit of the single-chain Msp is the fourth Msp monomer (first nucleotide sequence), then the second Msp monomer (seventh nucleotide sequence) comprises a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. In another example, if the starting subunit of the single-chain Msp is the fifth Msp monomer (first nucleotide sequence), then the third Msp monomer (seventh nucleotide sequence) comprises a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. In another example, if the starting subunit of the single-chain Msp is the sixth Msp monomer (first nucleotide sequence), then the fourth Msp monomer (seventh nucleotide sequence) comprises a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. In another example, if the starting subunit of the single-chain Msp is the seventh Msp monomer (first nucleotide sequence), then the fifth Msp monomer (seventh nucleotide sequence) comprises a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. In another example, if the starting subunit of the single-chain Msp is the eighth Msp monomer (first nucleotide sequence), then the sixth Msp monomer (seventh nucleotide sequence) comprises a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. For example, and not to be limiting, the first Msp monomer can be a mutant Msp monomer that comprises a mutation at positions D56, I68, S73, D118, D134 and E139 and the seventh monomer can be a mutant Msp monomer that comprises a mutation at positions L88 and I105. Optionally, each of the amino acid positions at positions D56, I68, S73, D118, D134 and E139 of the first mutant Msp monomer can be substituted with lysine or arginine. Optionally, each of the amino acid positions at positions D56, I68, S73, D118, D134 and E139 of the first mutant Msp monomer can be substituted with phenylalanine, tryptophan, histidine or tyrosine. Optionally, each of the amino acid positions at positions L88 and I105 of the seventh mutant Msp monomer can be substituted with lysine or arginine. Optionally, each of the amino acid positions at positions L88 and I105 of the seventh mutant Msp monomer can be substituted with phenylalanine, tryptophan, histidine or tyrosine. Substitution of D56, I68, S73, D118, D134, E139, L88 and/or I105 with aromatic amino acids, such as, phenylalanine, tryptophan, histidine or tyrosine can promote p-stacking interactions with an analyte, for example, nucleotides, to decrease translocation velocity. Optionally, the first, second, third, fourth, fifth, sixth, seventh, and eighth Msp monomer sequence, or a subset thereof can comprise a D90N, a D91N and a D93N mutation. FIGS.10A and10Bshow a non-limiting example of a positive ramp created in a single-chain Msp comprising a first mutant Msp monomer that comprises a mutation at positions D56, I68, S73, D118, D134 and E139 and a seventh mutant Msp monomer that comprises a mutation at positions L88 and I105. This positively charged ramp inside the vestibule of the MspA guides single-stranded nucleic acids, for example DNA, through the Msp. The electrostatic interactions between the nucleic acid and the ramp enable controlled translocation of DNA through the pore. This reduces Brownian motion of the nucleic acid and the translocation rate. This also increases the precision and the interaction between the nucleic acid bases and the amino acids in the constriction zone.FIG.10Cis a schematic of a single-chain Msp. Numbers under subunits #1 and #7 represent locations of the positive ramp. In any of the mutant single-chain Msps set forth herein, the constriction zone can be modified to increase the nucleobase, protein or analyte recognition properties of MspA. Modifications to the constriction zone can create a reading head that increases, for example, base-specific interactions. A reading head can be created by introducing an amino acid with a longer side chain that protrudes into the path of DNA or another analyte. For example, and not to be limiting, in order to create one or more reading heads, the amino acid at position 90 and/or 91 in any of the mutant Msp monomers of the single-chain Msps described herein can be substituted with arginine, lysine, histidine, glutamine, methionine, threonine, phenylalanine, tyrosine, tryptophan or an unnatural amino acid. Positioning heads can also be created to increase the efficiency of one or more reading heads. For example, amino acids with longer side chains, preferably hydrophobic or negatively charged, can be introduced, opposite to the reading head, in order to reduce escape motions of DNA or another analyte in the constriction zone. Amino acids that are suitable, include but are not limited to, aspartate, glutamate, valine, leucine, isoleucine, phenylalanine, tyrosine, tryptophan and unnatural amino acids. In order to further slow down translocation rates, a stacking slide can be created by mutating one or more of the amino acids at positions 83, 86, 88 and 105. For example, and not to be limiting, one or more of the amino acids at positions 83, 86, 88 and 105 can be substituted with tryptophan, tyrosine or phenylalanine. Optionally, the stacking slide is positioned such that it is located in proximity to a positive ramp. One or more of the second, third, fourth, fifth, sixth, seventh, and eighth Msp monomer sequence or any subset thereof, can be independently selected from the group consisting of a wildtype MspA monomer, a mutant MspA monomer, a wild-type MspA paralog or homolog monomer, and a mutant MspA paralog or homolog monomer. It is understood that, when the second, third, fourth, fifth, sixth, seventh and/or eight Msp monomer sequence is a mutant MspA monomer sequence, the mutant MspA monomer sequence can be the same or different than the first mutant MspA monomer sequence. Optionally, the second, third, fourth, fifth, sixth, seventh, and eighth Msp monomer sequence, or any subset thereof, is a wild-type MspA paralog or homolog monomer. These include, but are not limited to, MspA/Msmeg0965, MspB/Msmeg0520, MspC/Msmeg5483, MspD/Msmeg6057, MppA, PorM1, PorM2, PorM1, Mmcs4296, Mmcs4297, Mmcs3857, Mmcs4382, Mmcs4383, Mjls3843, Mjls3857, Mjls3931 Mjls4674, Mjls4675, Mjls4677, Map3123c, Mav3943, Mvan1836, Mvan4117, Mvan4839, Mvan4840, Mvan5016, Mvan5017, Mvan5768, MUL 2391, Mflv1734, Mflv1735, Mflv2295, Mflv1891, MCH4691c, MCH4689c, MCH4690c, MAB1080, MAB1081, MAB2800, RHA1 ro08561, RHA1 ro04074, and RHA1 ro03127. A wild-type MspA paralog or homolog monomer may be a wild-type MspB monomer. Mutant Msp Monomers and the Nucleic Acids Encoding them Further provided is a nucleic acid encoding a mutant Msp monomer, wherein the Msp monomer comprises a mutation at position 97. Optionally, the mutant Msp monomer can comprise a mutation at P97, wherein the mutation is not a P97S mutation or a P97C mutation. Optionally, the mutant Msp monomer can comprise a P97F mutation. Optionally the mutant Msp monomer can further comprise a mutation at amino acid position D118, a mutation at position D134 or a mutation at position E139. For example, the mutant Msp monomer comprising a mutation at position 97 can further comprise a D118R mutation, a D134R mutation and/or a E139K mutation. Optionally, the mutant Msp monomer comprising a mutation at position 97 can further comprise (i) a mutation at position 93 and/or (ii) a mutation at position D90, position D91 or both positions D90 and D91. Optionally, the amino acid at position 90 or 91 is substituted with arginine, lysine, histidine, glutamine, methionine, threonine, phenylalanine, tyrosine or tryptophan. Optionally, the mutant Msp monomer can further comprise a D90N, a D91N and a D93N mutation. For example, and not to be limiting, a mutant MspA monomer sequence can comprise a D90N mutation, a D91N mutation, a D93N mutation, a P97F mutation, a D118R mutation, a D134R mutation and a E139K mutation. Further provided is a nucleic acid encoding a mutant Msp monomer, wherein the Msp monomer comprises a mutation at one or more of the following positions: I68, S73, S116, P123 or V128. Optionally, the mutant monomer further comprises a mutation at amino acid position D118. Optionally, the Msp monomer further comprises a mutation at position D90, position D91 or both positions D90 and D91. Optionally, the amino acid at position 91 or the amino acid at position 90 can be substituted with arginine, lysine, histidine, glutamine, methionine, threonine, phenylalanine, tyrosine or tryptophan. Optionally, the mutant Msp monomer sequence can further comprise a D90N, a D91N and a D93N mutation. Optionally, the mutant Msp monomer sequence can further comprise a mutation in one or more of the amino acids at positions D13, A55, D56, E57, F58, E63, S136, D134, G137, E139 or D172. Optionally, one or more of D13, A55, D56, E57, F58, E63, S136, D134, G137, E139 or D172 can be substituted with lysine or arginine. Further provided is a nucleic acid encoding a mutant Msp monomer, wherein the Msp monomer comprises a mutation at one or more of the following positions: T83, N86, G92 or A96. Optionally, the mutant Msp monomer sequence further comprises a mutation at position L88 or 1105. Optionally, the mutant monomer further comprises a mutation at amino acid positions D118. Optionally, the Msp monomer further comprises a mutation at position D90, position D91 or both positions D90 and D91. Optionally, the amino acid at position 91 or the amino acid at position 90 can be substituted with arginine, lysine, histidine, glutamine, methionine, threonine, phenylalanine, tyrosine or tryptophan. Optionally, the mutant Msp monomer sequence can further comprise a D90N, a D91N and a D93N mutation. As used herein, a mutant Msp monomer refers to an Msp monomer that has at least or at most 70, 75, 80, 85, 90, 95, 98, or 99 percent or more identity, or any range derivable therein, but less than 100% identity, as compared to a wild-type Msp monomer, and retains tunnel-forming capability when associated with one or more other Msp monomers (wild-type or mutant). Therefore, in addition to the mutations described herein, any mutant Msp provided herein can further comprise additional modifications such as substitutions, insertions, deletions, and/or additions, as long as the mutant Msp monomer has at least or at most 70, 75, 80, 85, 90, 95, 98, or 99 percent or more identity, or any range derivable therein, but less than 100%, to a wild-type Msp monomer, and retains tunnel-forming capability when associated with one or more other Msp monomers. Any mutant Msp described herein can comprise 2-15 Msp monomers that are the same or different, wherein at least one of the Msp monomers is a mutant Msp monomer. Optionally, a mutant Msp comprises 7-9 Msp monomers that are the same or different. Optionally, at least a second monomer is selected from the group consisting of a wildtype Msp monomer, a second mutant Msp monomer, a wild-type Msp paralog or homolog monomer, and a mutant Msp paralog or homolog monomer, wherein the second mutant Msp monomer may be the same or different than the first mutant Msp monomer. For example, any mutant Msp described herein can comprise 2-15 Msp monomers wherein at least one of the Msp monomers is a mutant MspA monomer. Optionally, at least a second monomer is selected from the group consisting of a wildtype MspA monomer, a second mutant MspA monomer, a wild-type MspA paralog or homolog monomer, and a mutant MspA paralog or homolog monomer, wherein the second mutant MspA monomer can be the same or different than the first mutant MspA monomer. Optionally, the second monomer is a wild-type MspA paralog or homolog monomer. For example, a mutant Msp can comprise one or more Msp monomers comprising a mutation at position 97. In another example, a mutant Msp can comprise one or more Msp monomers comprising a mutation at one or more of I68, S73, S116, P123 or V128 and one or more Msp monomers comprising a mutation at one or more of the following positions: T83, N86, L88, I105, D90, D91, G92, D93 or A96. In another example, a mutant Msp can comprise one or more Msp monomers with mutations at positions D56, I68, S73, D118, D134 and E139 and one or more Msp monomers with mutations at positions L88 and I105. Modifications in amino acid sequence may arise as allelic variations (e.g., due to genetic polymorphism), may arise due to environmental influence (e.g., due to exposure to ultraviolet radiation), or other human intervention (e.g., by mutagenesis of cloned DNA sequences), such as induced point, deletion, insertion, and substitution mutants. These modifications can result in changes in the amino acid sequence, provide silent mutations, modify a restriction site, or provide other specific mutations. Amino acid sequence modifications typically fall into one or more of three classes: substitutional, insertional, or deletional modifications. Insertions include amino and/or terminal fusions as well as intrasequence insertions of single or multiple amino acid residues. Insertions ordinarily will be smaller insertions than those of amino or carboxyl terminal fusions, for example, on the order of one to four residues. Deletions are characterized by the removal of one or more amino acid residues from the protein sequence. Typically, no more than about from 2 to about 6 residues are deleted at any one site within the protein molecule. Amino acid substitutions are typically of single residues, but can occur at a number of different locations at once; insertions usually will be on the order of about from 1 to about 10 amino acid residues; and deletions will range from about 1 to about 30 residues. Deletions or insertions preferably are made in adjacent pairs, i.e., a deletion of 2 residues or insertion of 2 residues. Substitutions, deletions, insertions or any combination thereof can be combined to arrive at a final construct. The mutations may or may not place the sequence out of reading frame and may or may not create complementary regions that could produce secondary mRNA structure. Substitutional modifications are those in which at least one residue has been removed and a different residue inserted in its place. Modifications, including the specific amino acid substitutions disclosed herein, are made by known methods. By way of example, modifications are made by site specific mutagenesis of nucleotides in the DNA encoding the protein, thereby producing a DNA encoding the modification, and thereafter expressing the DNA in recombinant cell culture to produce the Msp monomers or single chain multimers. Techniques for making substitution mutations at predetermined sites in DNA having a known sequence are well known, for example M13 primer mutagenesis and PCR mutagenesis. The amino acids in the Msp proteins described herein can be any of the 20 naturally occurring amino acids, D-stereoisomers of the naturally occurring amino acids, unnatural amino acids and chemically modified amino acids. Unnatural amino acids (that is, those that are not naturally found in proteins) are also known in the art, as set forth in, for example, Williams et al., Mol. Cell. Biol. 9:2574 (1989); Evans et al., J. Amer. Chem. Soc. 112:4011-4030 (1990); Pu et al., J. Amer. Chem. Soc. 56:1280-1283 (1991); Williams et al., J. Amer. Chem. Soc. 113:9276-9286 (1991); and all references cited therein. B and γ amino acids are known in the art and are also contemplated herein as unnatural amino acids. As used herein, a chemically modified amino acid refers to an amino acid whose side chain has been chemically modified. For example, a side chain can be modified to comprise a signaling moiety, such as a fluorophore or a radiolabel. A side chain can also be modified to comprise a new functional group, such as a thiol, carboxylic acid, or amino group. Post-translationally modified amino acids are also included in the definition of chemically modified amino acids. Also contemplated are conservative amino acid substitutions. By way of example, conservative amino acid substitutions can be made in one or more of the amino acid residues of any Msp monomer provided herein. One of skill in the art would know that a conservative substitution is the replacement of one amino acid residue with another that is biologically and/or chemically similar. The following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) Nonconservative substitutions, for example, substituting a proline with glycine are also contemplated. Those of skill in the art readily understand how to determine the identity of two polypeptides or nucleic acids. For example, the identity can be calculated after aligning the two sequences so that the identity is at its highest level. Another way of calculating identity can be performed by published algorithms. Optimal alignment of sequences for comparison can be conducted using the algorithm of Smith and Waterman,Adv. Appl. Math.2: 482 (1981); by the alignment algorithm of Needleman and Wunsch,J. Mol. Biol.48: 443 (1970); by the search for similarity method of Pearson and Lipman,Proc. Natl. Acad. Sci. U.S.A.85: 2444 (1988); by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.; the BLAST algorithm of Tatusova and Madden FEMS Microbiol. Lett. 174: 247-250 (1999) available from the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/blast/b12seq/b12.html); or by inspection. The same types of identity can be obtained for nucleic acids by, for example, the algorithms disclosed in Zuker,Science244:48-52, 1989; Jaeger et al.Proc. Natl. Acad. Sci. USA86:7706-7710, 1989; Jaeger et al.Methods Enzymol.183:281-306, 1989 that are herein incorporated by this reference for at least material related to nucleic acid alignment. It is understood that any of the methods typically can be used and that, in certain instances, the results of these various methods may differ, but the skilled artisan understands if identity is found with at least one of these methods, the sequences would be said to have the stated identity. For example, as used herein, a sequence recited as having a particular percent identity to another sequence refers to sequences that have the recited identity as calculated by any one or more of the calculation methods described above. For example, a first sequence has 80 percent identity, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent identity to the second sequence using the Zuker calculation method even if the first sequence does not have 80 percent identity to the second sequence as calculated by any of the other calculation methods. As yet another example, a first sequence has 80 percent identity, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent identity to the second sequence using each of calculation methods (although, in practice, the different calculation methods will often result in different calculated identity percentages). Further, any Msp or Msp monomer can also be chemically or biologically modified. For example, one can modify an Msp or Msp monomer with chemicals to produce disulfide bridges, as is known by those of skill in the art. An Msp can comprise a nucleotide binding site. As used herein, a nucleotide binding site refers to a site in an Msp where a nucleotide stays in contact with, or resides at, an amino acid for a period of time that is longer than attributable to diffusion movement, such as greater than one picosecond or one nanosecond. Molecular dynamics calculations can be employed to assess these temporary resting times. Polypeptides encoded by nucleic acids described herein are also provided. Therefore polypeptides comprising a mutant Msp monomer or functional fragment thereof, are provided. Non-limiting examples of mutant Msp monomers include but are not limited to, polypeptides comprising SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3 and SEQ ID NO: 4 comprising any of the mutations described herein. Further provided is a Msp monomer comprising an amino acid sequence that has least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% identity or any percentage in between to a polypeptide comprising SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3 and SEQ ID NO: 4, wherein the polypeptide comprises any of the mutations described herein. Also provided are polypeptides comprising a mutant single-chain Msp or functional fragment thereof. Also provided are polypeptides comprising a mutant single-chain Msp comprising any of the mutant Msp monomers described herein, or a fragment thereof. Non-limiting examples of mutant Msp monomers comprising mutations set forth herein are provided in Table 2. Each exemplary mutant Msp monomer comprises all of the mutations listed for each monomer. For example, D90N/D91N/D93N/P97F indicates that all four mutations are present. Single chain Msps comprising any of the exemplary mutant Msp monomer sequences provided in Table 2 are also provided herein. It is understood that amino acids listed in parentheses are listed as alternatives for substitutions at that position. For example, P97 (A/V/L/IF/M/Y/W or G) means that P97 can be substituted with A, V, L, I, F, M, Y W or G. TABLE 2D90N/D91N/D93N/P97(A/V/L/I/F/M/Y/W or G)D90N/D91N/D93N/P97F/D118R/D134R/E139KD90N/D91N/D93N/P97FD90N/D91N/D93N/G92(A/V/L/I/P/F/M/Y or W)D90N/D91N/D93N/I94(A/V/L/P/F/M/Y/W or G)D90N/D91N/D93N/T95(A/V/L/P/F/M/Y/W or G)D90N/D91N/D93N/A96(V/L/P/F/I/M/Y/W or G)D90N/D91N/D93N/P98(A/V/L/F/I/M/Y/W or G)D90N/D91N/D93N/F99(A/V/L/P/I/M/Y/W or G)D90N/D91N/D93N/G100(A/V/L/P/F/I/M/Y or W)D90N/D91N/D93N/L101(A/V/I/F/M/Y/W or G)D90N/D91N/D93N/N102((AN/L/I/P/F/M/Y/W or G)D90N/D91N/D93N/S103((A/V/L/I/P/F/M/Y/W or G)D90N/G92F/D91N/D93N/P97FD90N/T95F/D91N/D93N/P97FD90N/A96F/D91N/D93N/P97FD90N/A96F/D91N/D93N/P97F/P98FD90N/G92F/D91N/D93N/P97F/D118R/D134R/E139KI68(R/K/F/W/Y or H)/D90N/D91N/D93NI68(R/K/F/W/Y or H)/D90N/D91N/D93N/D118R/D134R/E139KI68(K/R/F/W/Y or H)/D90N/D91N/D93NI68(K/R/F/W/Y or H)/D90N/D91N/D93N/D118R/D134R/E139KS73 (K/R/F/W/Y or H)/D90N/D91N/D93NS73 (K/R/F/W/Y or H)/D90N/D91N/D93N/D118R/D134R/E139KS73 (K/R/F/W/Y or H)/D90N/D91N/D93NS73 (K/R/F/W/Y or H)/D90N/D91N/D93N/D118R/D134R/E139KD90N/D91N/D93N/S116RD90N/D91N/D93N/S116R/D118R/D134R/E139KD90N/D91N/D93N/S116KD90N/D91N/D93N/S116K/D118R/D134R/E139KD90N/D91N/D93N/P123RD90N/D91N/D93N/P123R/D118R/D134R/E139KD90N/D91N/D93N/P123KD90N/D91N/D93N/P123K/D118R/D134R/E139KD90N/D91N/D93N/L88(K/R/F/W/H or Y)D90N/D91N/D93N/1105(K/R/F/W/H or Y)D90N/D91N/D93N/L88(K/R/F/W/H or Y)/D118R/D134R/E139KD90N/D91N/D93N/I105(K/R/F/W/H or Y)/D118R/D134R/E139KD90N/D91N/D93N/L88(K/R/F/W/H or Y)/I105(K/R/F/W/H or Y)D90N/D91N/D93N/L88(K/R/F/W/H or Y)/I105(K/R/F/W/H orY)/D118R/D134R/E139KD90N/D91N/D93N/L88(K/R/F/W/H or Y)D90N/D91N/D93N/T83(K/R/F/W/H or Y)/D118R/D134R/E139KD90N/D91N/D93N/T83(K/R/F/W/H or Y)D90N/D91N/D93N/N86(K/R/F/W/H or Y)D90N/D91N/D93N/N86(K/R/F/W/H or Y)/D118R/D134R/E139K Tunnel-Forming Proteins Methods of determining whether a protein is a tunnel-forming protein are well known in the art. One can determine if an Msp forms a tunnel by determining whether the protein inserts into a bilayer, such as described in Example 2 of U.S. Patent Publication No. 20120055792, incorporated herein in its entirety by this reference. All of the methods of making and using porins described in U.S. Patent Publication No. 20120055792 can be employed to make and use the Msp porins described herein. If the protein inserts into the bilayer, then the porin is a tunnel-forming protein. Typically, tunnel formation is detected by observing a discrete change in conductivity. See, U.S. Patent Publication No. 20120055792, and Niederweis et al.,Mol. Microbiol.33:933 (1999), both of which are incorporated herein by reference. Bilayers are described herein. An Msp will typically be able to be inserted in a lipid bilayer or other thin film, which are each well-known in the art. An example of inserting a mutant MspA into a lipid bilayer is provided in U.S. Patent Publication No. 20120055792; this technique can be applied to other Msp proteins as well. In addition, U.S. Pat. No. 6,746,594, incorporated herein by reference, describes a variety of lipid bilayers and thin films, including inorganic materials, that can be employed with respect to the Msps discussed herein. Methods, apparatuses, and techniques described in U.S. Pat. No. 6,267,872, incorporated herein by reference in its entirety, are also employable with respect to Msps discussed herein. Moreover, more than one Msp can be comprised in a lipid bilayer. For example, 2 3, 4, 5, 10, 20, 200, 2000, or more can be comprised in a lipid bilayer. Optionally, anywhere from 2 to 1010Msps can be employed in methods described herein. Such a plurality of Msps can be in the form of clusters of Msps. Clusters can be randomly assembled or can adopt a pattern. As used herein, a cluster refers to molecules that are grouped together and move as a unit, but are not covalently bound to one another. Optionally, Msps do not gate spontaneously. As used herein, to gate or gating refers to the spontaneous change of electrical conductance through the tunnel of the protein that is usually temporary (e.g., lasting for as few as 1-10 milliseconds to up to a second). Long lasting gating events can often be reversed by changing the polarity. Under most circumstances, the probability of gating increases with the application of higher voltages. Gating and the degree of conductance through the tunnel change are highly variable among Msps, depending on, for example, the make-up of the vestibule and constriction zone as well as the properties of the liquid medium in which the protein is submerged. Typically, the protein becomes less conductive during gating, and conductance can permanently stop (i.e., the tunnel may permanently shut) as a result, such that the process is irreversible. Optionally, gating refers to the conductance through the tunnel of a protein spontaneously changing to less than 75% of its open state current. Various conditions such as light and liquid medium, including its pH, buffer composition, detergent composition, and temperature, can affect the behavior of an Msp, particularly with respect to its conductance through the tunnel as well as the movement of an analyte with respect to the tunnel, either temporarily or permanently. As used throughout, a tunnel refers to the central, empty portion of an Msp that is defined by the vestibule and the constriction zone, through which a gas, liquid, ion, or analyte can pass. As used herein, “cis” refers to the side of an Msp tunnel through which an analyte enters the tunnel or across the face of which the analyte moves. As used herein, “trans” refers to the side of an Msp tunnel through which an analyte (or fragments thereof) exits the tunnel or across the face of which the analyte does not move. Any mutant Msp described herein, for example a mutant MspA, can comprise a vestibule and a constriction zone that define a tunnel. Further, the diameter of a mutant Msp or mutant Msp paralog or homolog can be less than the diameter of the constriction zone of a corresponding wild-type Msp or wild-type Msp paralog or homolog. A mutant Msp or mutant Msp paralog or homolog can have a mutation in the vestibule or the constriction zone that permits an analyte to translocate, electrophoretically or otherwise, through the tunnel of the mutant Msp or mutant Msp paralog or homolog with a translocation velocity or an average translocation velocity that is less than the translocation velocity or average translocation velocity at which the analyte translocates through the tunnel of a wild-type Msp or wild-type Msp paralog or homolog. Also, any mutant Msp described herein can comprise a vestibule having a length from about 2 to about 6 nm and a diameter from about 2 to about 6 nm, and a constriction zone 5 having a length from about 0.3 to about 3 nm and a diameter from about 0.3 to about 3 nm, wherein the vestibule and constriction zone together define a tunnel. It is understood that, one or more mutations, can be made in the vestibule or the constriction zone of any of the Msp described herein in order to increase or decrease conductance through the tunnel of an Msp. For example, any of the mutant Msps described herein can further comprise a deletion, substitution or insertion of an amino acid in the vestibule and/or the constriction zone in order to modify conductance. As used throughout, a vestibule refers to the cone-shaped portion of the interior of an Msp whose diameter generally decreases from one end to the other along a central axis, where the narrowest portion of the vestibule is connected to the constriction zone. A vestibule can also be referred to as a goblet. The vestibule and the constriction zone together define the tunnel of an Msp. When referring to a diameter of the vestibule, it is understood that because the vestibule is cone-like in shape, the diameter changes along the path of a central axis, where the diameter is larger at one end than the opposite end. The diameter can range from about 2 nm to about 6 nm. Optionally, the diameter is about, at least about, or at most about 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, or 6.0 nm, or any range derivable therein. The length of the central axis can range from about 2 nm to about 6 nm. Optionally, the length is about, at least about, or at most about 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, or 6.0 nm, or any range derivable therein. When referring to diameter herein, one can determine a diameter by measuring center-to-center distances or atomic surface-to-surface distances. As used throughout, a constriction zone refers to the narrowest portion of the tunnel of an Msp, in terms of diameter, that is connected to the vestibule. The length of the constriction zone can range from about 0.3 nm to about 2 nm. Optionally, the length is about, at most about, or at least about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, or 3 nm, or any range derivable therein. The diameter of the constriction zone can range from about 0.3 nm to about 2 nm. Optionally, the diameter is about, at most about, or at least about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, or 3 nm, or any range derivable therein. Any Msp discussed herein can be comprised in a lipid bilayer. Optionally, an analyte electrophoretically or otherwise translocates from the cis side through a tunnel to the trans side. Optionally, an analyte electrophoretically or otherwise translocates from the trans side through a tunnel to the cis side. Optionally, an analyte is electrophoretically or otherwise driven from the cis side or the trans side into a tunnel and stays in the tunnel or then retracts to the cis side or the trans side, respectively. It is understood that analytes can translocate through the tunnel in the presence or absence of an electric field. Single-chain Msps function at a wide range of electrolyte concentration, for example from about 0.3-1M KCl (seeFIG.22). To optimize channel activity, lipid association can be performed prior to insertion of Msp in a membrane or lipid bilayer. In a non-limiting example,FIG.22shows that no channel activity was observed in a buffer containing only 0.3 M KCl at pH 8.0. However, breaking the membrane and subsequent repainting of the membrane leads to increased channel activity of scMspA PN1 in the electrolyte containing 0.3 M KCl at pH 8.0. Therefore, in any of the methods set forth herein, an Msp can be contacted or preincubated with one or more lipids to optimize channel activity. Vectors and Cells A vector comprising a nucleic acid encoding a polypeptide described herein is also provided. The vector can further comprise a promoter sequence, for example, a constitutive promoter or an inducible promoter. Examples of constitutive promoter include, but are not limited to, the psmycpromoter and Phsp60. Examples of inducible promoters include, but are not limited to, an acetamide-inducible promoter and a tetracycline inducible promoter. Cultured cells transfected with any vector described herein, or progeny thereof, wherein the cell is capable of expressing a Msp (either as a single-chain Msp, an Msp comprising Msp monomers or an Msp monomer, are also provided). AMycobacterium smegmatisstrain comprising any vector described herein is also provided. AMycobacterium smegmatisstrain free of endogenous porins is also contemplated and can further comprise any vector described herein. By “free” is meant that an endogenous porin cannot be detected in an immunoblot when using an appropriate Msp-specific antiserum, or comprising less than 1% endogenous porins. Any of the Msp monomers or single-chain Msps disclosed herein can be produced by transforming a mutant bacterial strain comprising a deletion of a wild-type MspA, a wild-type MspB, a wild-type MspC, a wildtype MspD, with a vector comprising an inducible promoter operably linked to a nucleic acid sequence encoding the Msp monomer or single-chain Msp porin; and purifying the Msp monomer or single-chain Msp porin (See, for example, U.S. Pat. No. 6,746,594 incorporated herein by reference). Optionally, the mutant bacterial strain comprises a deletion of a recA gene. Optionally, the vector comprises any of the nucleic acids encoding an Msp monomer or single-chain Msp described herein. The bacterial strain can further compriseM. smegmatisstrain ML16, ML714 or ML712. Systems and Methods of Use Also provided is a system comprising a mutant Msp described herein having a vestibule and a constriction zone that define a tunnel, wherein the tunnel is positioned between a first liquid medium and a second liquid medium, wherein at least one liquid medium comprises an analyte, and wherein the system is operative to detect a property of the analyte. A system can be operative to detect a property of any analyte comprising subjecting an Msp to an electric field such that the analyte interacts with the Msp. A system can be operative to detect a property of the analyte comprising subjecting the Msp to an electric field such that the analyte electrophoretically translocates through the tunnel of the Msp. Also provided is a system comprising an Msp having a vestibule and a constriction zone that define a tunnel, wherein the tunnel is positioned in a lipid bilayer between a first liquid medium and a second liquid medium, and wherein the only point of liquid communication between the first and second liquid media occurs in the tunnel. Moreover, any system described herein can comprise any mutant Msp described herein. The first and second liquid media can be the same or different, and either one or both can comprise one or more salts, detergents, or buffers. In fact, any liquid media described herein can comprise one or more of a salt, a detergent, or a buffer. Optionally, at least one liquid medium is conductive. Optionally, at least one liquid medium is not conductive. Any liquid medium described herein can comprise a viscosity-altering substance or a velocity-altering substance. The liquid medium can comprise any analyte described herein. A property of an analyte can be an electrical, chemical, or physical property. An Msp can be comprised in a lipid bilayer in a system or any other embodiment described herein. A system can comprise a plurality of Msps. A system can comprise any Msp described herein, such as a single-chain mutant MspA or a mutant Msp comprising at least 2-15 monomers, wherein at least one of the monomers is a mutant MspA monomer. A mutant Msp comprised in a system can comprise a vestibule having a length from about 2 to about 6 nm and a diameter from about 2 to about 6 nm, and a constriction zone having a length from about 0.3 to about 3 nm and a diameter from about 0.3 to about 3 nm, wherein the vestibule and constriction zone together define a tunnel. Any Msp described herein, including an Msp comprised in a system, can further comprise a molecular motor. The molecular motor in a system is capable of moving an analyte into or through a tunnel with a translocation velocity or an average translocation velocity that is less than the translocation velocity or average translocation velocity at which the analyte translocates into or through the tunnel in the absence of the molecular motor. The molecular motor can be, for example, an enzyme, such as a polymerase, an exonuclease, or a helicase such as DnaB or a phage nucleic acid packing motors (see, for example, SerwerViruses3(7): 1249-80 (2011)). Any system described herein can further comprise a patch-clamp amplifier or a data acquisition device. A system can further comprise one or more temperature regulating devices in communication with the first liquid medium, the second liquid medium, or both. Any system described herein can be operative to translocate an analyte through an Msp tunnel either electrophoretically or otherwise. The mutant MspA can have a mutation in the vestibule or the constriction zone that permits an analyte to translocate, e.g., electrophoretically, through the tunnel with an average translocation velocity of less than 0.5 nm/μs or less than 0.05 nm/μs. The analyte can be selected from the group consisting of a nucleotide(s), a nucleic acid, amino acid(s), a peptide, a protein, a polymer, a drug, an ion, a biological warfare agent, a pollutant, a nanoscopic object, or a combination or cluster thereof. Optionally, the analyte is further defined as a nucleic acid. The nucleic acid can translocate, electrophoretically or otherwise, through the tunnel with an average translocation velocity of less than 1 nucleotide/μs, or less than 0.1 nucleotide/μs. A nucleic acid can be further defined as ssDNA, dsDNA, RNA, or a combination thereof. As used herein, electrophoretically translocating an analyte, refers to applying an electric field to an Msp porin that is in contact with one or more solutions (e.g., immersed in a solution), such that current flows through the Msp tunnel. The electric field moves an analyte such that it interacts with the tunnel. As used herein, “interacts” means that the analyte moves into and, optionally, through the tunnel, where “through the Msp tunnel” (or “translocates”) means to enter one side of the tunnel and move to and out of the other side of the tunnel. It is specifically contemplated that any analyte discussed herein can translocate through an Msp tunnel, either electrophoretically or otherwise, in any embodiment discussed herein. In this regard, it is specifically contemplated that any embodiment herein comprising translocation can refer to electrophoretic translocation or nonelectrophoretic translocation, unless specifically noted. Optionally, methods that do not employ electrophoretic translocation are contemplated. As used throughout, a liquid medium includes aqueous, organic-aqueous, and organic-only liquid media. Organic media include, e.g., methanol, ethanol, dimethylsulfoxide, and mixtures thereof. Liquids employable in methods described herein are well-known in the art. Descriptions and examples of such media, including conductive liquid media, are provided in U.S. Pat. No. 7,189,503, for example, which is incorporated herein by reference in its entirety. Salts, detergents, or buffers may be added to such media. Such agents can be employed to alter pH or ionic strength of the liquid medium. Viscosity-altering substances, such as glycerol or various polymers (e.g., polyvinylpyrrolidone, polyethylene glycol, polyvinyl alcohol, cellulose polymers), and mixtures thereof, can be included in liquid media. Methods of measuring viscosity are well-known in the art. Any agent that can be added to a liquid medium can also alter the velocity of an analyte that is being studied. As such, a velocity-altering agent may be a salt, a detergent, a buffer, a viscosity-altering substance, or any other agent added to a liquid medium that increases or decreases the velocity of an analyte. Typically, an analyte employed herein is soluble or partially soluble in at least one liquid medium that is in contact with an Msp described herein. As used herein, nucleic acid refers to a deoxyribonucleotide or ribonucleotide polymer in either single- or double-stranded form, and unless otherwise limited, encompasses known analogs of natural nucleotides that hybridize to nucleic acids in a manner similar to naturally occurring nucleotides, such as peptide nucleic acids (PNAs) and phosphorothioate DNA. Unless otherwise indicated, a particular nucleic acid sequence includes the complementary sequence thereof. Nucleotides include, but are not limited to, ATP, dATP, CTP, dCTP, GTP, dGTP, UTP, TTP, dUTP, 5-methyl-CTP, 5-methyldCTP, ITP, diTP, 2-amino-adenosine-TP, 2-amino-deoxyadenosine-TP, 2-thiothymidine triphosphate, pyrrolo-pyrimidine triphosphate, and 2-thiocytidine, as well as the alphathiotriphosphates for all of the above, and 2′-O-methyl-ribonucleotide triphosphates for all the above bases. Modified bases include, but are not limited to, 5-Br-UTP, 5-BrdUTP, 5-F-UTP, 5-F-dUTP, 5-propynyl dCTP, and 5-propynyl-dUTP. As used herein, a drug refers to any substance that may alter a biological process of a subject. Drugs can be designed or used for or in the diagnosis, treatment, or prevention of a disease, disorder, syndrome, or other health affliction of a subject. Drugs can be recreational in nature, that is, used simply to alter a biological process and not used for or in the diagnosis, treatment, or prevention of a disease, disorder, syndrome, or other health affliction of a subject. Biologics, which refer to substances produced by biological mechanisms involving recombinant DNA technology, are also encompassed by the term drug. Drugs include, for example, antibacterials, anti-inflammatories, anticoagulants, antivirals, antihypertensives, antidepressants, antimicrobials, analgesics, anesthetics, beta-blockers, bisphosphonates, chemotherapeutics, contrast agents, fertility medications, hallucinogens, hormones, narcotics, opiates, sedatives, statins, steroids, and vasodilators. Non-limiting examples of drugs can also be found in theMerck Index: an Encyclopedia of Chemicals, Drugs, and Biologicals,15thed. New Jersey: Merck, 2013. Antibacterial drugs used in the treatment of tuberculosis, for example, include isoniazid, rifampicin, pyrazinamide, and ethambutol. Methods employing a drug as an analyte can further comprise drug screening. For example, uptake of a drug into a cell or an organism can be investigated using an Msp by observing ion current blockades. Specific Msp porin constriction zones and/or vestibules with various sizes, electrostatic properties, and chemical properties can be constructed to closely emulate the desired pathway for drugs to enter or exit a cell or organism. These methods could greatly accelerate screening for drugs as well as drug design (see, for example, Pagel et al.,J. Bacteriology189:8593 (2007)). As used herein, a biological warfare agent refers to any organism or any naturally occurring, bioengineered, or synthesized component of any such microorganism capable of causing death or disease in plants or animals (including humans) or degradation of food or water supplies, or degradation of the environment. Non-limiting examples include Ebola viruses, Marburg virus,Bacillus anthracisandClostridium botulinum, Variola major, Variola minor, anthrax, and ricin. As used herein, a pollutant refers to a material that pollutes air, water, or soil. Non-limiting examples of pollutants include fertilizers, pesticides, insecticides, detergents, petroleum hydrocarbons, smoke, and heavy metal-containing substances, such as those containing zinc, copper, or mercury (e.g., methylmercury). Any analyte can be used herein, including, for example, a nucleotide(s), a nucleic acid, an amino acid(s), a peptide, a protein, a polymer, a drug, an ion, a biological warfare agent, a pollutant, a nanoscopic object, or any other molecule comprising one of these analytes or a combination of thereof. An analyte can be a cluster of molecules (e.g. 2-10 nucleotides or amino acids), in that the cluster as a whole is considered an analyte. Typically, an analyte's size will not be so great such that it cannot enter a tunnel of an Msp. In other words, a typical analyte will be smaller in size than the opening of a tunnel of an Msp. However, an analyte having a size larger than the opening of a tunnel can be employed, and it can be determined that the analyte's size is too large to enter the tunnel. Optionally, the molecular weight of the analyte is less than one million Da. Optionally, the molecular weight of the analyte is about, at most about, or at least about 1,000,000, 950,000, 900,000, 850,000, 800,000, 750,000, 700,000, 650,000, 600,000, 550,000, 500,000, 450,000, 400,000, 350,000, 300,000, 250,000, 200,000, 150,000, 100,000, 75,000, 50,000, 25,000, 20,000, 15,000, 10,000, 7,500, 5,000, 2,500, 2,000, 1,500, 1,000, or 500 Da or less, or any range derivable therein. An analyte can also be a nanoscopic object, which is an object that is smaller than 100 nm in two of its dimensions. As used herein, an analyte can further comprise a magnetic bead. A magnetic bead can be further defined as a streptavidin-coated magnetic bead. An analyte can further comprise an optical bead. Any analyte described herein can be an ion or can be neutral. An analyte can comprise biotin. Beads that can be employed include magnetic beads and optical beads. For example, one can use streptavidin-coated magnetic beads to apply an opposing force to the electrostatic forces that pull DNA through the tunnel of an Msp. In this latter technique a magnetic bead is attached to biotinylated DNA, and a force comparable to the electrostatic driving force (−10 pN) would be applied using a strong magnetic field gradient. See Gosse and Croquette,Biophys. J.82:3314 (2002). In this way, the blockade-current readout would be unaffected, but the forces on the DNA could be independently controlled. Tens or hundreds of complete, independent reads of each DNA could then be correlated and assembled to reconstruct an accurate DNA sequence. Optical beads manipulated by “optical tweezers” are also known in the art, and such methods can be applied to the Msps described herein. Optical tweezers are a common tool used to exert a force on a nanoscopic object. An analyte is attached on one end of the bead, while the other end can be inserted into the tunnel of the porin. The position and force of the bead is controlled and measured with the optical tweezers. Such methods control the passage of the analyte into the tunnel and allow for more control of the reading of the analyte, such as the reading of the units of a polymer. See, e.g., Trepagnier et al., Nano Lett. 7:2824 (2007) for a description of such methods in the context of artificial nanopores. U.S. Pat. No. 5,795,782, incorporated herein by reference, also discusses the use of optical tweezers. Fluorescence resonance energy transfer (FRET), a well-known technique, can be employed in analytical methods described herein. For example, a fluorescent FRET acceptor or FRET-donor molecule can be incorporated into an Msp. The analyte is then labeled with a matching FRET-donor or FRET-acceptor. When the matching FRET donor is within the Forster distance to the FRET acceptor, energy transfer will likely occur. The resulting signal could be used for analytical purposes instead of or in addition to methods using ion current as described herein. Accordingly, methods of detection, identification, or sequencing can comprise FRET technology. Other optical methods that can be employed include introducing optically active molecules into the interior of an Msp (such as the vestibule or the constriction zone). External light would be applied to affect the interior of the protein. Such methods could be used to affect the translocation velocity of an analyte or could allow the analyte's entry or exit from the tunnel, offering controlled passage of the analyte. Alternatively, optical pulses focused onto the pore could be used to heat the pore to affect how it interacts with the analyte. Such control could be very fast as the heat from a small volume of a focal point would dissipate rapidly. Methods of controlling the translocation velocity of an analyte can therefore employ such optically active molecules or optical pulses. Manipulation of translocation velocity can also be accomplished by attaching an object to one end of an analyte, and the other end of the analyte then interacts with the Msp. The object can be a bead (e.g., a polystyrene bead), a cell, a large molecule such as streptavidin, neutravidin, DNA, etc., or a nanoscopic object. The object could then be subjected to a fluid flow or could be subject to passive viscous drag. Molecular motors are well-known in the art and refer to a molecule (e.g., an enzyme) that physically interacts with an analyte, such as a polymer (e.g., a 15 polynucleotide), and is capable of physically moving the analyte with respect to a fixed location, such as the vestibule, constriction zone, or tunnel of an Msp. Although not intending to be bound by theory, molecular motors utilize chemical energy to generate mechanical force. A molecular motor can interact with each unit (or “mer”) of a polymer in a sequential manner. Non-limiting examples of molecular motors include DNA polymerases, RNA polymerases, helicases, ribosomes, and exonucleases. Nonenzymatic motors are also known, such as virus motors that pack DNA. See Smith et al., Nature 413:748 (2001). A variety of molecular motors and desirable properties of such motors are described in U.S. Pat. No. 7,238,485, which is incorporated herein by reference in its entirety. A molecular motor can be disposed on the cis side or the trans side of an Msp porin and can optionally be immobilized, such as described by the '485 patent. Methods of incorporating a molecular motor into an Msp can be performed using methods described in the '485 patent. Systems and apparatuses described in the '485 patent can be employed with respect to an Msp described herein as well. Indeed, any embodiment discussed in the '485 patent can be employed using an Msp, as described herein. Molecular motors are also discussed in, e.g., Cockroft et al., J. Amer. Chem. Soc. 130:818 (2008); Benner et al., Nature Nanotech. 2:718 (2007); and Gyarfas et al., ACS Nano 3:1457 (2009). A molecular motor is typically employed to regulate the rate or translocation velocity at which an analyte interacts with an Msp. Any Msp described herein can comprise a molecular motor. Optionally, a molecular motor is employed to decrease the rate at which an analyte enters an Msp porin tunnel or to decrease the translocation velocity at which an analyte translocates through an Msp tunnel. Optionally, the translocation velocity or average translocation velocity is less than 0.5 nm/μs. Optionally, the translocation velocity or average translocation velocity is less than 0.05 nm/μs. Optionally, the translocation velocity or average translocation velocity is less than 1 nucleotide/μs. Optionally, the translocation velocity or average translocation velocity is less than 0.1 nucleotide/μs. Optionally, the rate of movement of an analyte ranges from greater than 0 Hz to 2000 Hz. Here, rate refers to the number of subunits (or “mers”) of a regular polymer advancing in one second (Hz). Optionally, the range is between about 50-1500 Hz, 100-1500 Hz, or 350-1500 Hz. Optionally, the rate of movement is about, at most about, or at least about 25, 75, 100, 150, 200, 250, 300, 15 350,400,450,500,550,600,650,700,750,800,850,900,950,1000,1050,1100,1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550, 1600, 1650, 1700, 1750, 1800, 1850, 1900, 1950, or 2000 Hz, or any range derivable therein. The rate can be controlled by the use of a molecular motor that moves an analyte at a substantially constant rate, at least for a portion of time during a characterization. In addition, the range of rate of movement can depend on the molecular motor. For example, for an RNA polymerase, a range can be 350-1500 Hz; for a DNA polymerase, a range can be 75-1500 Hz; and for ribosomes, helicases, and exonucleases, a range can be 50-1500 Hz. Recording and detection techniques can be employed in the methods described herein. In addition, U.S. Pat. Nos. 5,795,782 and 7,189,503, incorporated herein by reference in its entirety, also describes recording methods and instrumentation that can be employed with respect to Msps, as well as methods for optimizing conductance readings. U.S. Pat. No. 6,746,594, incorporated herein by reference in its entirety, describes a support for thin films containing nanopores and methods for using such supports that can be employed with respect to the Msps described herein. Method of Making a Single Chain Msp The Msp pore, for example, MspA, is currently the best available channel protein for nanopore sequencing of nucleic acids. However, its composition of eight subunits makes it impossible to introduce asymmetric changes in the pore that could optimize the properties of the Msp for nucleic acid sequencing. In order to overcome this difficulty, methods of making a single chain Msp are provided herein. These methods can be used to produce a full or partial single-chain Msp. Generally, the method comprises transforming a mutant bacterial strain. The mutant strain comprises a deletion of a wild-type MspA, a wild-type MspB, a wild-type MspC, a wild-type MspD, and optionally a deletion of the recA gene. The mutant strain is transformed with a vector comprising a nucleic acid sequence encoding a single-chain Msp porin. The single-chain Msp porin is then purified from the bacteria. Optionally, the single-chain Msp porin comprises a single-chain MspA porin. Optionally, the vector comprises any of the nucleic acids described herein. As described in the Examples, in order to combine the superior sequencing capabilities of MspA with an increased ability to adapt vestibule and constriction properties to DNA sequencing, a single-chain MspA porin octamer was constructed that allows for the optimal properties of the vestibule and the constriction zone for DNA sequencing. The MspA chain termini are close together in the MspA porin and are connected by a short peptide linker. The (GGGGS)3(SEQ ID NO:3) peptide, for example, is used to connect the carboxy-terminus of the preceding MspA monomer (or multimer) to the amino-terminus of the following MspA monomer (or multimer), which lacks signal peptide. To create a vector comprising the MspA porin sequence, each MspA monomer sequence is flanked by a unique restriction site, which allows the capability to mutate any individual monomer. To create an MspA porin sequence, each MspA sequence can be assembled stepwise to form a dimeric, tetrameric, and octameric single-chain MspA utilizing the unique restriction sites. To avoid problems of recombination in creating the single-chain MspA multimer, seven MspA genes are synthesized with different codon usages i.e., the genes encode the exact same amino acid sequence, however, the DNA sequence has been altered from the native MspA nucleotide sequence (SEQ ID NO: 10). To create the MspA porin sequence, the nucleotide sequence encoding the first Msp monomer can optionally contain a nucleic acid sequence encoding a leader sequence (e.g., amino acids 1 to 27 of SEQ ID NO: 6). Each of the seven Msp monomer sequences following the first Msp monomer sequence can comprise SEQ ID NO: 1 or SEQ ID NO: 1 with one or more mutations described herein. The vector comprising the MspA porin sequence is transformed into the quadruple porin mutant bacterial strain, as described in the Examples. Optionally, single chain Msps can be purified and subjected to a refolding procedure. For example, anion exchange chromatography in the presence of 8M urea can be used to obtain a pure fraction of a single chain Msp which is dialyzed against a buffer to remove urea. After dialysis, a refolding buffer comprising a refolding agent, for example, L-arginine and detergent, are added to the sample and purified, refolded single chain Msp is obtained. Refolding agents are known to those of skill in the art. These include, but are not limited to, arginine, arginine hydrochloride, arginineamide, glycineamide, proline, glycerol, and cyclodextrains (see, for example, Yamaguchi et al.Biomolecules4: 235-251 (2014); and expression levels and oligomeric status of the MspA porin can be checked by Western blot or other immunohistochemical techniques known to those of skill in the art. The tunnel activity of the MspA porin can be determined by lipid bilayer experiments, as described in the Examples and as known to those of skill in the art. Single chain M18-MspA pores insert much more frequently into lipid bilayers than a similar amount of octameric M1-MspA. Insertion of octameric MspA is a tedious procedure. Thus, single-chain Msps, such as those described herein facilitate setup of systems and methods of using Msp for detecting and identifying analytes, for example, for nucleic acid sequencing. Method of Increasing Msp Insertion in a Lipid Bilayer Provided herein is a method of increasing the number of Msp insertions in a lipid bilayer, comprising contacting any Msp described herein with a lipid to form a lipid-associated Msp and inserting the lipid-associated Msp of step into a lipid bilayer. Optionally, the contacting step comprises inserting the Msp in a lipid bilayer and disrupting the lipid bilayer to form a lipid-associated Msp. For example, an Msp can be inserted in a lipid bilayer that is subsequently disrupted. The disrupted lipid bilayer comprises Msp(s). Therefore, the Msp(s) are lipid associated. The lipid-associated Msp can then be contacted with other lipids to form another lipid bilayer that comprises the lipid-associated Msps. As used herein, a lipid bilayer is a thin membrane comprising lipid molecules, for example, phosopholipids, that can be used to insert any Msp provided herein. Therefore, in the methods provided herein, the Msp can be contacted with phospholipids, either as part of a lipid bilayer or not, in order to form lipid-associated Msp. As set forth above, one of skill in the art can determine if an Msp inserts into a bilayer, by using techniques such as those described in Example 2 of U.S. Patent Publication No. 20120055792, incorporated herein in its entirety by this reference. All of the methods of making and using porins described in U.S. Patent Publication No. 20120055792 can be employed to make and use the Msp porins described herein. If the protein inserts into the bilayer, then the porin is a tunnel-forming protein. Typically, tunnel formation is detected by observing a discrete change in conductivity. See, U.S. Patent Publication No. 20120055792, and Niederweis et al.,Mol. Microbiol.33:933 (1999), both of which are incorporated herein by reference. The increase in Msp insertions can be an increase of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 300%, 400% or greater. Detection Methods Further provided is a method for detecting the presence of an analyte, comprising: (a) applying an electric field sufficient to translocate an analyte from a first conductive medium to a second conductive medium in liquid communication through any mutant Msp described herein; and (b) measuring an ion current, wherein a reduction in the ion current indicates the presence of the analyte in the first medium. Optionally, the first and second liquid conductive media are the same. Optionally, the first and second liquid conductive media are different. The mutant Msp porin can be any mutant Msp porin discussed herein. For example, the Msp porin can be a mutant single-chain Msp, a mutant Msp comprising 2-15 monomers or combinations thereof. As set forth above, a plurality of Msps can also be used in the methods described herein. In the methods disclosed herein, an Msp can further comprise a molecular motor. The molecular motor can be capable of moving an analyte into or through a tunnel with a translocation velocity or an average translocation velocity that is less than the translocation velocity or average translocation velocity at which the analyte electrophoretically translocates into or through the tunnel in the absence of the molecular motor. Accordingly, in any embodiment herein comprising application of an electric field, the electric field can be sufficient to cause the analyte to electrophoretically translocate through the tunnel. Any liquid medium discussed herein, such as a conductive liquid medium, can comprise an analyte. In the methods comprising measuring an ion current, the analyte interacts with an Msp porin tunnel to provide a current pattern, wherein the appearance of a blockade in the current pattern indicates the presence of the analyte. The methods disclosed herein can further comprise identifying the analyte. For example, such methods can comprise comparing the current pattern obtained with respect to an unknown analyte to that of a known current pattern obtained using a known analyte under the same conditions. In another example, and not to be limiting, identifying the analyte can comprise (a) measuring the ion current to provide a current pattern, wherein a reduction in the current defines a blockade in the current pattern, and (b) comparing one or more blockades in the current pattern to (i) one or more blockades in the current pattern, or (ii) one or more blockades in a known current pattern obtained using a known analyte. The analyte can be any analyte described herein. For example, the analyte can be a nucleotide(s), a nucleic acid, an amino acid(s), a peptide, a protein, a polymer, a drug, an ion, a pollutant, a nanoscopic object, or a biological warfare agent. In the methods provided herein, optionally, at least one of the first or second conductive liquid media comprises a plurality of different analytes. In methods where the analyte is a polymer, for example, a protein, a peptide or a nucleic acid, the method can further comprise identifying one or more units of the polymer. For example, identifying one or more units of the polymer can comprise measuring the ion current to provide a current pattern comprising a blockade for each polymer unit, and comparing one or more blockades in the current pattern to (i) one or more other blockades in the current pattern or (ii) one or more blockades in a current pattern obtained using a polymer having known units. These methods can comprise identifying sequential units of the polymer, for example, and not to be limiting, sequential or consecutive nucleotides in a nucleic acid. In another example, sequential or consecutive amino acids in a polypeptide can be identified using the methods described herein. The methods provided herein can comprise distinguishing at least a first unit within a polymer from at least a second unit within the polymer. Distinguishing can comprise measuring the ion current produced as the first and second units separately translocate through a tunnel to produce a first and a second current pattern, respectively, where the first and second current patterns differ from each other. The methods provided herein can further comprise sequencing a polymer. Sequencing can comprise measuring the ion current or optical signals as each unit of the polymer is separately translocated through the tunnel to provide a current pattern that is associated with each unit, and comparing each current pattern to the current pattern of a known unit obtained under the same conditions, such that the polymer is sequenced. Further provided is a method of sequencing nucleic acids or polypeptides using any of the mutant Msps provided herein. The method comprises creating a lipid bilayer comprising a first and second side, adding a purified Msp to the first side of the lipid bilayer, applying positive voltage to the second side of the lipid bilayer, translocating an experimental nucleic acid or polypeptide sequence through the Msp porin, comparing the experimental blockade current with a blockade current standard, and determining the experimental sequence. Any of the detection methods provided herein can further comprise determining the concentration, size, molecular weight, shape, or orientation of the analyte, or any combination thereof. As used herein, a polymer refers to a molecule that comprises two or more linear units (also known as a “mers”), where each unit may be the same or different. Non-limiting examples of polymers include nucleic acids, peptides, and proteins, as well as a variety of hydrocarbon polymers (e.g., polyethylene, polystyrene) and functionalized hydrocarbon polymers, wherein the backbone of the polymer comprises a carbon chain (e.g., polyvinyl chloride, polymethacrylates). Polymers include copolymers, block copolymers, and branched polymers such as star polymers and dendrimers. Methods of sequencing polymers using Msp are described herein. In addition, sequencing methods can be performed in methods analogous to those described in U.S. Pat. No. 7,189,503, incorporated herein by reference in its entirety. See also U.S. Pat. No. 6,015,714, incorporated herein by reference in its entirety. More than one read can be performed in such sequencing methods to improve accuracy. Methods of analyzing characteristics of polymers (e.g., size, length, concentration, identity) and identifying discrete units (or “mers”) of polymers are discussed in the '503 patent as well, and can be employed with respect to the present Msps. Indeed, an Msp can be employed with respect to any method discussed in the '503 patent. At present, several types of observable signals can be used as readout mechanisms in nanopore sequencing and analyte detection. An exemplary readout method relies on an ionic blockade current or copassing current, uniquely determined by the identity of a nucleotide or other analyte occupying the narrowest constriction in the pore. This method is referred to as blockade current nanopore sequencing or BCNS. Blockade current detection and characterization of nucleic acids has been demonstrated in both the protein pore ahemolysin (aHL) and solid-state nanopores. Blockade current detection and characterization has been shown to provide a host of information about the structure of DNA passing through, or held in, a nanopore in various contexts. In general, a blockade is evidenced by a change in ion current that is clearly distinguishable from noise fluctuations and is usually associated with the presence of an analyte molecule at the pore's central opening. The strength of the blockade will depend on the type of analyte that is present. More particularly, a blockade refers to an interval where the ionic current drops below a threshold of about 5-100% of the unblocked current level, remains there for at least 1.0 μs, and returns spontaneously to the unblocked level. For example, the ionic current may drop below a threshold of about, at least about, or at most about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%, or any range derivable therein. Blockades are rejected if the unblocked signal directly preceding or following it has an average current that deviates from the typical unblocked level by more than twice the rms noise of the unblocked signal. Deep blockades are identified as intervals where the ionic current drops <50% of the unblocked level. Intervals where the current remains between 80% and 50% of the unblocked level are identified as partial blockades. Disclosed are materials, compositions, and components that can be used for, can be used in conjunction with, can be used in preparation for, or are products of the disclosed methods and compositions. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutations may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a method is disclosed and discussed and a number of modifications that can be made to a number of compositions included in the method are discussed, each and every combination and permutation of the method, and the modifications that are possible are specifically contemplated unless specifically indicated to the contrary. Likewise, any subset or combination of these is also specifically contemplated and disclosed. This concept applies to all aspects of this disclosure including, but not limited to, steps in methods. Thus, if there are a variety of additional steps that can be performed, it is understood that each of these additional steps can be performed with any specific method steps or combination of method steps of the disclosed methods, and that each such combination or subset of combinations is specifically contemplated and should be considered disclosed. It is also contemplated that any embodiment discussed in this specification can be implemented with respect to any method, compound, protein, porin, peptide, polypeptide, multimer, monomer, nucleic acid, vector, strain, cultured cell, system, or composition, etc., described herein, and vice versa. Publications cited herein and the material for which they are cited are hereby specifically incorporated by reference in their entireties. A number of embodiments have been described. Nevertheless, it will be understood that various modifications can be made. Accordingly, other embodiments are within the scope of the following claims. Examples A single-chain MspA was constructed consisting of eight covalently connected monomers. As shown herein, an expression plasmid encoding single-chain M1-MspA is not stable in wild-typeM. smegmatisbut can be propagated in anM. smegmatislacking the recA gene, which is required for homologous recombination. The recA gene was deleted in the quadruple porin mutantM. smegmatisML712, which lacks the four known Msp porins. This strain enabled expression of single-chain M1-MspA. Tunnels made by single-chain M1-MspA had a similar conductance as octameric M1-MspA tunnels, but had drastically improved resistance to voltage gating. This unforeseen advantage of single-chain MspA is of great importance for nanopore sequencing of nucleic acids, for example, DNA. Construction of an Msp Quadruple Deletion Mutant ofM. smegmatis For isolation of mutant MspA porins a triple porin deletion mutantMycobacterium smegmatisML16 strain (ΔmspA::FRT, ΔmspC::FRT, ΔmspD::FRT) was used (see Stephan et al., Mol. Microbiol. 58: 714-730 (2005)). However, low levels of MspB could still be detected in this strain in immunoblots with MspA-specific rabbit antiserum. The presence of MspB can contribute to the heterogeneity observed in single-channel experiments and complicates data analysis. In order to overcome this problem and improve MspA preparations anM. smegmatisstrain lacking all four msp genes was constructed. Since the deletion of all four msp genes ofM. smegmatisis lethal, the first step was to integrate an expression cassette for the N-terminal channel-forming domain of CpnT (1) into theM. smegmatisporin triple mutant ML16. To this end, the plasmid pML2622 was constructed, which carries the N-terminal domain of CpnT tagged with His6 and HA under the control of a nitrile-inducible promoter (FIG.2). The N-terminal domain of CpnT formed channels in planar bilayer experiments and complemented the porin mutantM. smegmatisML16 strain in glycerol uptake experiments. The rescue plasmid pML2622 was integrated into the mycobacteriophage L5-site in the chromosome of the porin triple mutantM. smegmatisML16. Integration was confirmed by PCR with the sets of primers complimentary to L5 genomic region. This strain was namedM. smegmatisML709. After integration of pML2622, the plasmid backbone was excised from the chromosome by Flp recombinase as described in Stephan et al. (Gene343: 181-190 (2004)) to remove the genes encoding hygromycin phosphotransferase and L5 integrase. This strain was namedM. smegmatisML709-234. To delete the remaining mspB gene inM. smegmatisML711 the mspB deletion vector pML1611 containing the two reporter genes gfp and xylE as markers for integration and allelic replacement was constructed (FIG.2). The mspB deletion vector pML1611 carries 863 bp and 946 bp-long upstream and downstream regions of mspB and was used to delete the mspB gene inM. smegmatisML709-234 by allelic exchange. This Msp porin quadruple mutant was namedM. smegmatisML711. The plasmid pCreSacB containing Cre recombinase was used to excise gfp and hyg genes flanked by loxP sites from the chromosome. The deletion of all four msp genes was confirmed by PCR with chromosomal DNA using specific sets of primers and by Southern blot with chromosomal DNA using specific probe (FIG.3). This unmarked Msp porin quadruple mutant was namedM. smegmatisML712 (relevant genotype: ΔmspA::FRT, ΔmspB::loxP ΔmspC::FRT, ΔmspD::FRT, attB L5::FRT-pNIT-cpnTd1-FRT). The growth of the strain ML712 on Middlebrook 7H10 agar plates was impaired in comparison to wt and ML16 strains. Next, the expression of msp genes in the ML712 strain was assessed by extraction ofM. smegmatiscells using the detergent octylpolyethyleneoxide as described in Heinz et al. (Anal. Biochem. 285: 113-120 (2000)). The Msp quadruple porin deletion mutantM. smegmatisML712 grown in Middlebrook 7H9 medium does not produce any Msp protein in contrast to the porin triple mutant ML16 (FIG.4). This demonstrated that deletion of all msp genes was achieved inM. smegmatisML712. The expression levels of the MspA were similar to that of wtM. smegmatis, when wt mspA or M1 mspA were expressed in ML712 using the plasmids pMN016 and pML904, respectively (FIG.4). Construction of Single-Chain M1-MspA Previously a M12-MspA subunit dimer was constructed (Pavlenok et al.PLoS One7(6): e38726). As the next step towards single-chain MspA, four mspA M1 genes were fused to encode a M14-MspA subunit tetramer. The resulting plasmid pML2647 was transformed into the quadruple porin deletion strain ofM. smegmatisML712 for protein production and purification. However, the tetrameric mspA plasmid was unstable. In order to avoid recombination, the recA gene was deleted in the quadruple porin mutant ML712 and the strainM. smegmatisML714 was created. Then, a gene encoding single-chain MspA, in which eight M1-MspA subunits are linked (M18-MspA), was cloned intoE. coli. Each of the subunits has a D90N mutation, a D91N mutation and a D93N mutation. This was achieved by fusing two genes encoding tetrameric M1 MspA together (M14-MspA) using pML2647 as a template. The individual MspA subunits are separated by (GGGGS)3linkers. In the resulting plasmid pML3213, the two tetrameric M1-mspA constructs are flanked by unique restriction sites (tetrad A: PacI, MluI; tetrad B: EcoRV, HindIII) (FIG.5). Genes within the tetramers are flanked by the same restriction sites with the exception of the first and last genes of the tetrads. The resulting m18-mspA gene was placed under the control of the constitutive psmycpromoter (FIG.5). The plasmid pML3213 was transformed intoM. smegmatisML714 (quadruple porin mutant with recA deletion) for protein production and purification. Western blot experiments showed that the expression level of single-chain M18-MspA inM. smegmatisML714 is lower compared to M12-MspA and is reduced to approximately 7% of wt MspA levels (FIG.6). Stability of Single-Chain M1-MspA The MspA pore is very resistant against thermal and chemical denaturation (Heinz et al.J. Biol. Chem.278: 8678-8685 (2003)). To test the thermal stability of single-chain MspA, the M18-MspA protein was subjected to increasing temperatures for 15 min in the presence of 2% SDS. (FIG.7) A significant amount of M18-MspA is stable even after heating the protein sample to 100° C. (FIG.7). This result shows that M18-MspA is at least as stable against thermal denaturation as the wt MspA protein. Channel Properties of Single-Chain M1-MspA To examine whether M18-MspA forms functional channels, in vitro lipid bilayer experiments were performed. No insertions were recorded when only n-octyl-POE buffer was added to the lipid bilayer. Addition of approximately 70 ng of M18-MspA protein resulted in the step-wise increase in the current across the lipid bilayer indicating the insertion of M18-MspA channels into the membrane (FIG.8A). Analysis of the current recordings of M18-MspA showed a major peak of 1.1 nS (FIG.8B). This channel conductance is similar to those of the pores made from the M1-MspA dimer (1.3 nS) and M1 MspA monomers (1.4 nS) (see Pavlenok et al.). Voltage Gating Voltage gating is defined as a spontaneous channel closure at a certain voltage threshold and is an intrinsic property of bacterial β-barrel channel proteins (Bainbridge et al.FEBS Lett431(3): 305-308 (1998)). Resistance to voltage gating is very important for nanopore sequencing experiments since voltages as high as +180 mV are used to translocate ssDNA through MspA pore (Manrao et al.Nat. Biotechnol.30(4): 349-353 (2012); Derrington et al.Proc. Natl. Acad. Sci. USA107(37): 16060-16065 (2010); Butler et al.Proc. Natl. Acad. Sci. USA105(52): 20647-20652 (2008)). Therefore, the voltage gating of M18-MspA in lipid bilayer experiments was analyzed. After insertion of approximately 220 M18-MspA pores, the voltage across the lipid bilayer was sequentially increased in 10 mV increments, and the ion current passing through the pores was measured for three minutes. The critical voltage Vc is defined as the voltage at which pores start to close, and is measured in these experiments as decrease of ion current. The M18-MspA channels started to close at +90 mV and were completely stable at all applied negative voltages (FIG.9). In a second experiment with gel-purified M18-MspA protein no voltage gating, up to voltages of ±100 mV, was observed. Thus, the critical voltage Vcof M18-MspA is two-fold higher than that of M1-MspA or M12-MspA (Vcrit+40 mV, −50 mV for both proteins). These results show that linking all eight subunits into a single polypeptide drastically increased the resistance of single-chain MspA to voltage gating. This unforeseen advantage of single-chain MspA is of great importance for nanopore sequencing of nucleic acids, for example, DNA. Construction of a Mutant Single Chain MspA (scMspA M2) As described herein, mutations in MspA are useful for improving its interactions with DNA, its base recognition properties and its interactions with membranes and accessory proteins such, for example, Phi29 DNA polymerase. Using the approach described above for single-chain M1 MspA, a mutant single-chain MspA (MspA M2), in which eight mutant MspA monomers are linked together was constructed. Expression of both single-chain M1 MspA and single-chain MspA M2 constructs inM. smegmatisML712 was shown by Western blots using an MspA antibody demonstrating that production of scMspA inM. smegmatisis feasible. As shown herein, single chain Msps can be expressed inE. coli. The single chain M2 MspA (scMspA M2) protein is made in mg amounts, but is not folded. A folding protocol has been developed that allows isolation of active scMspA M2. A single-chain m2-mspA (scm2-mspA) where eight m2-mspA genes (containing the mutations D90N/D91N/D93N/D118R/D134R/E139K as described in Butler et al. (PNAS 105: 20647-20652 (2008)) were connected by DNA fragments encoding (GGGGS)3polypeptide linkers. In addition, each gene was flanked by unique restriction sites to enable specific modifications of each MspA subunit. The genes in the sequence are named m2-1 through m2-8 beginning from the ATG start codon (FIG.11and Table 3). For protein production and purification of the single-chain MspA M2 protein inE. colicells the signal peptide of MspA was removed. The scm2-mspA sequence was codon optimized for optimal expression inE. coliand was synthesized by GenScript. The resulting scm2-mspA gene was flanked by EcoRI and HindIII and was obtained in a pUC57 plasmid from GenScript. Next, the whole sc m2-mspA was excised and cloned into the pET-21(a)+ vector. The scm2-mspA gene is under the control of the T7 promoter in the resulting plasmid pML3216 (FIG.11). For scMspA M2 protein production and purification, the plasmid pML3216 was transformed intoE. coliBL21(DE3)Omp8 strain which lacks 3 major porins (See Prilipov et al.FEMS Microbiol. Lett163: 65-72 (1998)). The BL21(DE3) Omp8 strain was chosen to avoid contamination of scMspA M2 with endogenous porins ofE. coli. After induction of scm2-mspA expression with 1.5 mM IPTG cells were grown at 37° C. in LB medium supplemented with ampicillin. Maximal expression of the target protein was observed two hours after induction accounting for approximately 4% of the total protein in the cell lysate (FIG.12). A protein band corresponding to scMspA M2 had an apparent mass of 170 kDa which is consistent with its predicted molecular mass of 165.6 kDa (FIG.12). Next, scMspA M2 from inclusion bodies was isolated and purified as described in Sambrook et al. (CSH Protocols2006) Inclusion bodies containing predominantly scMspA M2 protein were solubilized in 8 M urea. This sample was later a subject to anion exchange chromatography using HiTrap QFF column (GE HealthCare, United Kingdom) in the presence of 8 M urea. The elution profile of scMspA M2 protein was very similar to that of wt MspA published previously (Heinz et al., 2003). This protein is probably not folded and has no channel activity. Then, scMspA M2 was purified and subjected to a refolding procedure. After anion exchange chromatography a pure fraction of scMspA M2 with a concentration of 50 μg/ml was diluted by a factor of 10 in a buffer containing 10 mM NaCl, 25 mM HEPES, 0.6 M L-Arginine, 0.1% (v/v) LDAO, pH 8.0 to give final volume of 1 ml. The mixture was incubated overnight at room temperature (approximately 21° C.) on a rotating mixer. Then, the sample was transferred into a dialysis tube with 3.5 kDa MWCO and dialyzed against 2 L of a buffer containing 10 mM NaCl, 25 mM HEPES (pH 8.0), 0.023% (v/v) LDAO overnight at room temperature. The dialyzed protein was transferred into a microtube and incubated at a room temperature for an additional day. Next, the refolding efficiency was assessed by Western blot analysis using MspA-specific rabbit antiserum. After the refolding procedure, the band which reacts with MspA polyclonal antibodies migrated from 170 kDa to approximately 130 kDa indicating that folding of scMspA M2 to a more compact form with an increased electrophoretic mobility had occurred (FIG.13). Such an electrophoretic mobility shift upon folding has been observed for outer membrane proteins ofE. colipreviously. However, it was not clear whether MspA would show a similar phenomenon. In order to examine if scMspA M2 forms functional channels in vitro after the refolding procedure lipid bilayer experiments were performed. No channel activity was observed when only 0.023% LDAO-buffer was added to the planar bilayer. In contrast, addition of scMspA M2 protein after the refolding step resulted in a step-wise current increase indicative of channel insertions into lipid bilayer (FIG.14). Analysis of the current traces showed an average conductance of 2.3 nS (FIG.14). Of interest, analysis of MspA M2 made from monomers showed two peaks at 1.2 nS and 2.4 nS suggesting two different protein conformations. In addition, a multi-channel experiment with scMspA M2 showed improved voltage-gating resistance with a critical voltage of +80 mV/−70 mV (FIG.15). The increased voltage resistance is beneficial for example, for ssDNA experiments performed at relatively high voltages. Restriction sites of scMspA M2# ofAminoGeneRSRSSequenceEndsacidsflanked1EcoRIGAATTCcohesiveEF2KpnIGGTACCcohesiveGTm2-13NsiIATGCATcohesiveMH4ScaIAGTACTbluntSTm2-25NheIGCTAGCcohesiveAS6HpaIGTTAACbluntVNm2-37XbaITCTAGAcohesiveSR8NdeICATATGcohesiveHMm2-49EcorVGATATCbluntDI10PstICTGCAGcohesiveLQm2-511BstBITTCGAAcohesiveFE12BamHIGGATCCcohesiveGSm2-613MluIACGCGTcohesiveTR14PvuIICAGCTGbluntQLm2-715Afl IICTTAAGcohesiveLK16HindIIIAAGCTTcohesiveKLm2-8 Construction of Mutant Single Chain MspA (MspA PN1) A single-chain mspA pn1 (scmspA PN1) gene where eight mspA genes (containing a P97F mutation and mutations D90N/D91N/D93N/D118R/D134R/E139K as described in Butler et al. (PNAS105: 20647-20652 (2008)) were connected by DNA fragments encoding (GGGGS)3polypeptide linkers. In addition, each gene was flanked by unique restriction sites to enable specific modifications of each MspA subunit. The genes in the sequence are named m2-97-1 through m2-97-8 beginning from the ATG start codon (FIG.16and Table 3). For protein production and purification of the single-chain MspA PN1 protein inE. colicells the signal peptide of MspA was removed. The scmspA PN1 sequence was codon optimized for optimal expression inE. coliand was synthesized by GenScript. The resulting scmspA PN1 gene was flanked by EcoRI and HindIII and was obtained in a pUC57 plasmid from GenScript. Next, the entire scmspA PN1 was excised and cloned into the pET-21(a)+ vector. The scmspA PN1 gene is under the control of the T7 promoter in the resulting plasmid pML3216 (FIG.16). For scMspA PN1 protein production and purification the plasmid pML3216 was transformed intoE. coliBL21(DE3)Omp8 strain which lacks 3 major porins (See Prilipov et al.FEMS Microbiol. Lett163: 65-72 (1998)). The BL21(DE3) Omp8 strain was chosen to avoid contamination of scMspA PN1 with endogenous porins ofE. coli. After induction of scmspA PN1 expression with 1.5 mM IPTG, cells were grown at 37° C. in LB medium supplemented with ampicillin. Maximal expression of the target protein was observed two hours after induction accounting for approximately 5% of the total protein in the cell lysate (FIG.17). A protein band corresponding to scMspA PN1 had an apparent mass of 170 kDa which is consistent with its predicted molecular mass of 165.6 kDa (FIG.17). Next, scMspA PN1 from inclusion bodies was isolated and purified as described in Sambrook et al. (CSH Protocols2006) Inclusion bodies containing predominantly scMspA PN1 protein were solubilized in 8 M urea. This sample was later a subject to anion exchange chromatography using HiTrap QFF column (GE HealthCare, United Kingdom) in the presence of 8 M urea. The elution profile of scMspA PN1 protein was very similar to that of wt MspA published previously (Heinz et al., 2003). This protein is probably not folded and has no channel activity. Then, scMspA PN1 was purified and subjected to a refolding procedure. After anion exchange chromatography a pure fraction of scMspA PN1 was dialyzed against 2 L of buffer containing 140 mM NaCl, 10 mM K2HPO4/KH2PO4, 2 mM KCl (pH 7.5) to remove urea. The mixture was incubated overnight at room temperature (approximately 21° C.). After dialysis, L-arginine and LDAO were added to the sample to give a final concentration of 0.6M and 0.1% (v/v), respectively. The protein sample in the refolding buffer (140 mM NaCl, 10 mM K2HPO4/KH2PO4, 2 mM KCl, 0.6 M L-Arginine, 0.1% (v/v) LDAO, pH 7.5) was incubated overnight on an orbital shaker (FIG.18). The concentration of the purified sample was calculated to be 1.37 mg/ml as determined by absorbance at 280 nm. The protein yield was 0.45 mg per 1 liter of bacterial culture. To test the effect of phenylalanine at position 97 on incorporation of the single-chain MspA into artificial lipid membranes the insertion activity of different MspA constructs was measured by monitoring the release of fluorescent carboxyfluorescein dye from the liposomes as described (See Schwarz et al., Biophys. J. 58(3):577-83 (1990); Schwarz et al., Biochim. Biophys. Acta 1239(1): 51-7 (1995)). Briefly, DPhPC liposomes were prepared by extrusion in the presence of 30 mM carboxyfluorescein. Carboxyfluorescein is self-quenched when it is enclosed into lipid vesicles. After insertion of MspA pore into the dye-loaded liposome, diffusion-mediated efflux of the dye results in the increase of fluorescence in the reaction mixture.FIG.19shows the results of these carboxyfluorescein release experiments. Addition of buffers containing either LDAO (0.1% v/v) or OPOE (0.5% v/v) resulted in only minimal dye release from the liposomes, in contrast to Triton X-100 (1% v/v) buffer that was used as a positive control. Importantly, addition of scMspA PN1 (60 ng/ml, final) lead to faster and larger release of carboxyfluorscein than addition of scMspA M2(120 ng/ml, final). Interestingly, wt MspA (60 ng/ml, final) resulted in slower dye diffusion from the liposomes than scMspA PN1 (FIG.19). These data indicate that additional phenylalanines located in the loop 6 of scMspA promote faster and more efficient insertion of the pores into lipid bilayers. Next, the time of the first pore insertion into DPhPC membrane was measured in a bilayer set up. It was hypothesized that pores with enhanced insertion abilities would require less time to insert into lipid membrane. To examine the effect of phenylalanines in loop 6 on the time of membrane insertion of scMspA, scMspA PN1 was compared with scMspA M2. Briefly, the bilayer cuvette was filled with electrolyte, −10 mV potential was applied, and the data were acquired and recorded using TestPoint software. The same cuvette was always used in these experiments. The protein was added to both sides at a final concentration of 100 ng/ml. Importantly, successful insertion events were observed in 89% of the experiments for scMspA PN1, but only in 40% of the experiments for scMspA M2. This is consistent with the results of the carboxyfluorescein release experiments. Although the median insertion time for scMspA PN1 was 399 seconds as opposed to 695 seconds for scMspA M2, this difference was not significant. Surprisingly, the rate of insertion decreased when scMspA PN1 was analyzed in 0.3M KCl solution (median time: 859 seconds, 50%). However, half of the experiments resulted in successful insertions with scMspA PN1, while only one successful insertion was observed with scMspA M2 in 0.3M KCl with a time of 1270 seconds (8 membranes analyzed, 12% successful insertions) (FIG.20). This result shows the beneficial effect of phenylalanines in loop 6 for membrane insertion by single-chain MspA. In order to examine whether scMspA PN1 forms functional channels in vitro after the refolding procedure, lipid bilayer experiments were performed. No channel activity was observed when only 0.1% LDAO-buffer was added to the planar bilayer. In contrast, addition of scMspA PN1 protein after the refolding step resulted in a step-wise current increase indicative of channel insertions into the lipid bilayer (FIG.21). Analysis of the current traces showed an average conductance of 2.0 nS (FIG.21). This could translate into larger residual currents for each nucleotide and better signals in DNA sequencing experiments. Effect of Lipids on Channel Activity of Single-Chain MspA PN1 scMspA PN1 was stored for more than a month at room temperature in 1 μg/mg and 0.2 μg/ml amounts. The scMspA PN1 was diluted in 0.1% LDAO, 140 mM NaCl, 10 mM K2HPO4/KH2PO4 (pH 7.5), 2 mM KCL. Methods for making horizontal bilayers for channel experiments are known in the art. See, for example, Butler et al. (2008) and Akeson et al. Biophysical Journal; 77: 3227-3233 (1999), both of which are incorporated herein in their entireties. For the channel experiments, 2% diphtanoyl-phosphatidylcholine (DiphPC) in chloroform was used to form membrane bilayers for insertion of MspA essentially as described in Butler et al. and Akeson et al. After insertion of the MspA into the bilayers, the membrane was broken and the membrane was reapplied using 1% DiphPC in n-decane. The electrolyte used in these experiments was 0.3 or 1M KCl, 10 mM Hepes, pH 8.0 or pH 7.0, respectively. As shown inFIG.22, single-chain MspAs function at a wide range of electrolyte concentration, for example from about 0.3-1M KCl. To optimize channel activity, lipid association can be performed prior to insertion of MspA in a membrane or lipid bilayer. Therefore, in any of the methods set forth herein, an MspA can be contacted or preincubated with one or more lipids to optimize channel activity. In a non-limiting example,FIG.22shows that no channel activity was observed in a buffer containing only 0.3 M KCl at pH 8.0. However, breaking the membrane and subsequent repainting of the membrane leads to increased channel activity of scMspA PN1 in the electrolyte containing 0.3 M KCl at pH 8.0.
123,116
11858967
SEQUENCE LISTING The nucleic and amino acid sequences listed herein and in the accompanying sequence listing are shown using standard letter abbreviations for nucleotide bases, and three letter code for amino acids, as defined in 37 C.F.R. 1.822. Only one strand of the nucleic acid sequence is shown, but the complementary strand is understood as included by any reference to the displayed strand. In the accompanying sequence listing: SEQ ID NOs: 1 and 2 are oligonucleotide primers used to generate the insert of plasmid pXY152-endorf24. SEQ ID NO: 3 is the nucleic acid sequence of plasmid pXY152-endorf24. SEQ ID NOs: 4-7 are oligonucleotide primers used to generate the insert of plasmid pXY300-orf18ifd. SEQ ID NO: 8 is the nucleic acid sequence of plasmid pXY300-orf18ifd. SEQ ID NOs: 9 and 10 are oligonucleotide primers used to generate the oriT fragment of plasmid pKS-T-orf18pfrd. SEQ ID NO: 11 is the nucleic acid sequence of plasmid pKS-T-orf18pfrd. SEQ ID NOs: 12 and 13 are oligonucleotide primers used to generate the amR fragment of plasmid pKS-T-orf18pfrd-AmR. SEQ ID NO: 14 is the nucleic acid sequence of plasmid pKS-T-orf18pfrd-AmR. SEQ ID NOs: 15-18 are oligonucleotide primers used to generate the oriT and amR fragments of plasmid pKS-orf18ifd-T-AmR(NS). SEQ ID NO: 19 is the nucleic acid sequence of plasmid pKS-orf18ifd-T-AmR(NS). SEQ ID NO: 20 is the nucleic acid sequence of plasmid pXY152-endorf24-camtsr. SEQ ID NOs: 21 and 22 are oligonucleotide primers used to generate the bla fragment of plasmid pXY152-endorf24-blatsr. SEQ ID NO: 23 is the nucleic acid sequence of plasmid pXY152-endorf24-blatsr. SEQ ID NO: 24 is an oligonucleotide primer which corresponds to a region of a apramycin resistance gene. SEQ ID NO: 25 is the amino acid sequence of streptomycin activator StrR protein. SEQ ID NO: 26 is the amino acid sequence encoded by ORF24. SEQ ID NO: 27 is the nucleic acid sequence illustrating an in-frame deletion in orf18. SEQ ID NO: 28 is the amino acid sequence of Bbr insert. SEQ ID NO: 29 is the amino acid sequence of KasT insert. SEQ ID NO: 30 is the amino acid sequence of NovG insert. SEQ ID NO: 31 is the amino acid sequence of SgcR1 insert. SEQ ID NO: 32 is the amino acid sequence of Teil5* insert. SEQ ID NO: 33 is the amino acid sequence of response regulator ortholog SCO1745/AbrA2 fromS. coelicolorA3(2) (GenBank Accession No. CAB50960). SEQ ID NO: 34 is the amino acid sequence of response regulator ortholog SCO/3226/AbsA2 fromS. coelicolorA3(2) (GenBank Accession No. AAB08053). SEQ ID NO: 35 is the amino acid sequence of response regulator ortholog SC03818 fromS. coelicolorA3(2) (GenBank Accession No. CAB46941). SEQ ID NO: 36 is the amino acid sequence encoded by ORF18. SEQ ID NO: 37 is the nucleic acid sequence of orf18. SEQ ID NO: 38 is the nucleic acid sequence of orf24. SEQ ID NO: 39 is the nucleic acid sequence of the fosmid pXYF148 with the orf24 located at nucleotide position 23109 through 24044). SEQ ID NO: 40 is the nucleic acid sequence of fosmid pXYF24 with the orf18 located at nucleotide position 31091-31753). DETAILED DESCRIPTION OF SEVERAL EMBODIMENTS I. Introduction Enduracidin (FIG.1), also called enramycin, is a 17 amino acid lipodepsipeptide antibiotic produced by the soil bacteriumS. fungicidicusB-5477 (ATCC 21013). The peptide is isolated from the fermentation broth and mycelia primarily as a mixture of enduracidins A and B, which differ by one carbon in the length of the attached lipid chain. Structurally, the enduracidins are distinguished by a C12or C132Z,4E branched fatty acid moiety attached by an amide linkage to an aspartic acid residue, and the presence of numerous nonproteinogenic amino acid residues such as enduracididine (End), 4-hydroxyphenylglycine (Hpg), 3,5-dichloro-4-hydroxyphenylglycine (Dpg), citrulline (Cit) and ornithine (Orn) (cf.FIG.1). Seven of the 17 amino acids have the D configuration and six of the residues are Hpg or the chlorinated derivative Dpg. Enduracidin (for simplicity, the peptides will be referred to singularly) exhibits potent in vitro and in vivo antibacterial activity against a wide spectrum of Gram-positive organisms, including methicillin-resistantStaphylococcus aureus(MRSA) and vancomycin-resistantEnterococcus(VRE). Minimum inhibitory concentrations (MICs) are as low as 0.05 μg/mL and the effect is bactericidal. A study with 100 strains ofS. aureuscollected from various pathological products, and including 40% MRSA, established MICs ranging from 0.09 to 0.56 μg/mL with no strain able to survive exposure to 1 μg/mL. For comparison, typical MICs for vancomycin toward sensitive strains ofS. aureusrange from 0.5 to 2 μg/mL. In addition, enduracidin has an excellent toxicological profile. In a study in mice, rabbits, dogs and monkeys the acute LD50s were: intravenous, 30-125 mg/kg; intraperitoneal, 750-910 mg/kg; subcutaneous, intramuscular (i.m.) or oral, >5-10 g/kg. In the same study, monkeys receiving enduracidin i.m. for 6 months and rats that were similarly dosed for 12 months were found to only have localized inflammation at the injection site. In humans, enduracidin was administered i.m. (100 mg every 12 hours) to 20 hospitalized adult patients infected with MRSA. The peptide was reported to be free of side effects and also highly effective for treating urinary tract and skin infections caused by MRSA, but not chronic bone infections (Peromet et al.,Chemotherapy19:53-61, 1973). Enduracidin inhibits bacterial peptidoglycan cell wall biosynthesis by complexing with extracellular Lipid II, a precursor to the bacterial cell wall structure. The site of Lipid II complexation is distinct from that recognized by vancomycin and accounts for the action of enduracidin against vancomycin-resistant organisms. To date, there is no documented cross-resistance of enduracidin with any clinically-used antibiotic and no evidence of developed, acquired or transferrable resistance. The absence of any known form of transferrable resistance mechanism, the lack of oral bioavailability, its low toxicity, and excellent activity towardsClostridiumspp. have made enduracidin a key commercial peptide antibiotic used as a poultry feed additive for controlling clostridial enteritis. To derive a strain of the producing organism that could supply the quantities of the peptides required for commercial uses, Japan Takeda Animal Health (now part of Intervet/Merck Animal Health) subjectedS. fungicidicusB-5477 to various traditional strain improvement methods and selected for mutants that produced higher yields of enduracidin. An increasing worldwide market for enduracidin has driven efforts to further improve the yield of this antibiotic in BM38-2 (ATCC PTA-122342). With the genetic sequence of the enduracidin biosynthesis gene cluster available (GenBank accession no. DQ403252 which is hereby incorporated by reference as available on the world wide web on Oct. 3, 2006, BM38-2 (ATCC PTA-122342) served as the starting strain for the targeted genetic manipulation of regulatory genes associated with the gene cluster and constituted the basis for this disclosure. Herein, it is disclosed that the product of orf18 has a negative effect on enduracidin production and the orf24 gene product has a positive effect on enduracidin production and that recombinant strains derived from both theS. fungicidicuswild-type and BM38-2 (ATCC PTA-122342) organisms that exploit these regulatory effects produce elevated yields of enduracidin. In addition, disclosed herein are new gene replacement and integrative expression vectors based on pBluescript II KS and pSET152, respectively. II. Abbreviations and Terms a. AbbreviationsAA: amino acidAm: apramycinAmR: apramycin resistance markeramRp: native apramycin resistance promoterATCC: American Type Culture Collectionbla: ampicillin resistance geneBLAST: Basic Local Alignment Search Toolcam: chloramphenicol resistance geneCFU colony forming unitsCTAB: Cetyl Trimethyl Ammonium BromideCit: L-citrullineDpg: 3,5-dichloro-L-4-hydroxyphenylglycineEDTA: disodium EthyleneDiamineTetra-AcetateEnd: enduracididineEnradin: Enduracidin, EnramycinEPM: Enduracidin Production MediumHpg: D- and L-4-hydroxyphenylglycineHPLC: High Performance Liquid ChromatographyHTH: Helix-Turn-HelixIM: IntramuscularISP2: InternationalStreptomycesProject Medium 2ISP4: InternationalStreptomycesProject Medium 4LB: Luria-Bertani BrothLD50: Lethal Dosage, an LD50 represents the individual dose required to kill 50 percent of a population of test animalsMAH: Intervet/Merck Animal HealthMeOH: MethanolMICs: Minimum Inhibitory Concentrations,MRSA: methicillin-resistantStaphylococcus aureusnm: NanometerNRPS: non-ribosomal peptide synthetaseORF: open reading frameOrn: D-ornithinePCP: peptidyl carrier proteinPCR: Polymerase Chain ReactionPfrd: Plus Flanking Region DeletionSDS: Sodium Dodecyl SulfateSNP: single nucleotide polymorphismSPD: SpectrophotodiodeTFA: TriFluoroacetic AcidTSB: Tryptic Soy Brothtsr Thiostrepton resistance geneUV: ultravioletVRE: vancomycin-resistant enterococci b. Terms Unless otherwise noted, technical terms are used according to conventional usage. Definitions of common terms in molecular biology may be found in Benjamin Lewin Genes V published by Oxford University Press, 1994 (ISBN 0-19-854287-9); Kendrew et al. (eds.)The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182-9); and Robert A. Meyers (ed.)Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1-56081-569-8). In order to facilitate review of the various embodiments of this disclosure, the following explanations of specific terms are provided: Administering: Administration by any route to the animal. As used herein, administration typically refers to oral administration. Allelic variant: An alternate form of a polypeptide that is characterized as having a substitution, deletion, or addition of one or more amino acids. In one example, the variant does not alter the biological function of the polypeptide. Amplification: When used in reference to nucleic acids, techniques that increase the number of copies of a nucleic acid molecule in a sample or specimen. An example of amplification is the polymerase chain reaction, in which a biological sample collected from a subject is contacted with a pair of oligonucleotide primers, under conditions that allow for the hybridization of the primers to nucleic acid template in the sample. The primers are extended under suitable conditions, dissociated from the template, and then re-annealed, extended, and dissociated to amplify the number of copies of the nucleic acid. The product of in vitro amplification can be characterized by electrophoresis, restriction endonuclease cleavage patterns, oligonucleotide hybridization or ligation, and/or nucleic acid sequencing, using standard techniques. Other examples of in vitro amplification techniques include strand displacement amplification (see U.S. Pat. No. 5,744,311); transcription-free isothermal amplification (see U.S. Pat. No. 6,033,881); repair chain reaction amplification (see WO 90/01069); ligase chain reaction amplification (see EP-A-320 308); gap filling ligase chain reaction amplification (see U.S. Pat. No. 5,427,930); coupled ligase detection and PCR (see U.S. Pat. No. 6,027,889); and NASBA™ RNA transcription-free amplification (see U.S. Pat. No. 6,025,134). Analog, derivative or mimetic: An analog is a molecule that differs in chemical structure from a parent compound, for example a homolog (differing by an increment in the chemical structure, such as a difference in the length of an alkyl chain), a molecular fragment, a structure that differs by one or more functional groups, and/or a change in ionization. Structural analogs are often found using quantitative structure activity relationships (QSAR), with techniques such as those disclosed in Remington (The Science and Practice of Pharmacology,19th Edition (1995), chapter 28). When the changes to the original compound are substantial, or many incremental changes are combined, the compound is no longer an analog. For example, ramoplanin is not considered herein to be an analog of enduracidin: ramoplanin does not have either enduracididine amino acid, includes different amino acids, and though it has a lipid side chain, the chain length is substantially shorter. Analogs of enduracidin may be prepared by addition or deletion of functional groups on the amino acids that constitute the lipodepsipeptides, by substitution of one amino acid for another (excepting the enduracididine amino acids) or a combination of functional group modification and amino acid substitution. Exemplary enduracidin analogs include tetrahydorenduracidin A, tetrahydroenduracidin B, deschloroenduracidin A, and deschloroenduracidin B. A derivative is a biologically active molecule derived from the base structure. A mimetic is a molecule that mimics the activity of another molecule by mimicking the structure of such a molecule, such as a biologically active molecule. Thus, the term “mimetic” indicates a definite structure related to activity. Antibiotic: A substance, for example, enduracidin, penicillin or streptomycin, often produced by or derived from certain fungi, bacteria, and other organisms, that can destroy or inhibit the growth of other microorganisms. Antisense, Sense, and Antigene: Double-stranded DNA (dsDNA) has two strands, a 5′→3′ strand, referred to as the plus strand, and a 3′→5′ strand (the reverse compliment), referred to as the minus strand. Because RNA polymerase adds nucleic acids in a 5′→3′ direction, the minus strand of the DNA serves as the template for the RNA during transcription. Thus, the RNA formed will have a sequence complementary to the minus strand and identical to the plus strand (except that U is substituted for T). Antisense molecules are molecules that are specifically hybridizable or specifically complementary to either RNA or plus strand DNA. Sense molecules are molecules that are specifically hybridizable or specifically complementary to the minus strand of DNA. Antigene molecules are either antisense or sense molecules complimentary to a dsDNA target. In one embodiment, an antisense molecule specifically hybridizes to a target mRNA and inhibits transcription of the target mRNA. Binding or stable binding: A molecule, such as an oligonucleotide or protein, binds or stably binds to a target molecule, such as a target nucleic acid or protein, if binding is detectable. In one example, an oligonucleotide binds or stably binds to a target nucleic acid if a sufficient amount of the oligonucleotide forms base pairs or is hybridized to its target nucleic acid, to permit detection of that binding. Binding can be detected by either physical or functional properties of the target: oligonucleotide complex. Binding between a target and an oligonucleotide can be detected by any procedure known to one of ordinary skill in the art, including both functional and physical binding assays. Binding can be detected functionally by determining whether binding has an observable effect upon a biosynthetic process such as expression of a gene, DNA replication, transcription, translation and the like. Physical methods of detecting the binding of complementary strands of DNA or RNA are well known in the art, and include such methods as DNase I or chemical footprinting, gel shift and affinity cleavage assays, Northern blotting, dot blotting and light absorption detection procedures. For example, one method that is widely used, because it is so simple and reliable, involves observing a change in light absorption of a solution containing an oligonucleotide (or an analog) and a target nucleic acid at 220 to 300 nm as the temperature is slowly increased. If the oligonucleotide or analog has bound to its target, there is a sudden increase in absorption at a characteristic temperature as the oligonucleotide (or analog) and the target disassociate from each other, or melt. The binding between an oligomer and its target nucleic acid is frequently characterized by the temperature (Tm) at which 50% of the oligomer is melted from its target. A higher Tmmeans a stronger or more stable complex relative to a complex with a lower Tm. The binding between a protein and its target protein, such as an antibody for an antigen, is frequently characterized by determining the binding affinity. In one embodiment, affinity is calculated by a modification of the Scatchard method described by Frankel et al.,Mol. Immunol.,16:101-106, 1979. In another embodiment, binding affinity is measured by a specific binding agent receptor dissociation rate. In yet another embodiment, a high binding affinity is measured by a competition radioimmunoassay. In several examples, a high binding affinity is at least about 1×10−8M. In other embodiments, a high binding affinity is at least about 1.5×10−8, at least about 2.0×10−8, at least about 2.5×10−8, at least about 3.0×10−8, at least about 3.5×10−8, at least about 4.0×10−8, at least about 4.5×10−8, or at least about 5.0×10−8M. Biological function: The function(s) of a polypeptide in the cells in which it naturally occurs. A polypeptide can have more than one biological function. cDNA (complementary DNA): A piece of DNA lacking internal, non-coding segments (introns) and transcriptional regulatory sequences. cDNA can also contain untranslated regions (UTRs) that are responsible for translational control in the corresponding RNA molecule. cDNA is synthesized in the laboratory by reverse transcription from messenger RNA extracted from cells. Conservative substitution: Amino acid substitutions that do not substantially alter the activity (specificity or binding affinity) of the molecule. Typically conservative amino acid substitutions involve substitutions of one amino acid for another amino acid with similar chemical properties (e.g., charge or hydrophobicity). The following table shows exemplar conservative amino acid substitutions: OriginalConservativeResidueSubstitutionsAlaSerArgLysAsnGln; HisAspGluCysSerGlnAsnGluAspGlyProHisAsn; GlnIleLeu; ValLeuIle; ValLysArg; Gln; GluMetLeu; IlePheMet; Leu; TyrSerThrThrSerTrpTyrTyrTrp; PheValIle; Leu ControlStreptomyces fungicidicusstrain: The naturally-occurring wild-type strain,Streptomyces fungicidicusATCC21013. DNA (deoxyribonucleic acid): A long chain polymer which comprises the genetic material of most living organisms (some viruses have genes comprising ribonucleic acid (RNA)). The repeating units in DNA polymers are four different nucleotides, each of which comprises one of the four bases, adenine, guanine, cytosine and thymine, bound to a deoxyribose sugar to which a phosphate group is attached. Triplets of nucleotides (referred to as codons) code for each amino acid in a polypeptide. The term codon is also used for the corresponding (and complementary) sequences of three nucleotides in the mRNA into which the DNA sequence is transcribed. Unless otherwise specified, any reference to a DNA molecule is intended to include the reverse complement of that DNA molecule. Except where single-strandedness is required by the text herein, DNA molecules, though written to depict only a single strand, encompass both strands of a double-stranded DNA molecule. Thus, a reference to the nucleic acid molecule that encodes a specific protein, or a fragment thereof, encompasses both the sense strand and its reverse complement. Thus, for instance, it is appropriate to generate probes or primers from the reverse complement sequence of the disclosed nucleic acid molecules. Domain: A portion of a molecule such as proteins or nucleic acids that is structurally and/or functionally distinct from another portion of the molecule. Encode: A polynucleotide is said to “encode” a polypeptide if, in its native state or when manipulated by methods well known to those skilled in the art, it can be transcribed and/or translated to produce the mRNA for and/or the polypeptide or a fragment thereof. The anti-sense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced therefrom. Enduracidin: Enduracidins A and B are 17 amino acid lipodepsipeptides discovered in the late 1960s from fermentations of the soil bacteriumStreptomyces fungicidicusB-5477 (ATCC 21013). The A and B peptides are homologs that differ by one carbon in the length of the attached lipid chain. Structurally, the enduracidins are distinguished by C12or C132Z,4E branched fatty acid moiety and the presence of numerous nonproteinogenic amino acid residues such as enduracididine (End), 4-hydroxyphenylglycine (Hpg), 3,5-dichloro-4-hydroxyphenylglycine (Dpg), citrulline (Cit) and ornithine (Orn). Seven of the 17 amino acids have the D configuration and six of the residues are Hpg or the chlorinated analog Dpg. Functional fragments and variants of a polypeptide: Included are those fragments and variants that maintain one or more functions of the parent polypeptide. It is recognized that the gene or cDNA encoding a polypeptide can be considerably mutated without materially altering one or more of the polypeptide's functions. First, the genetic code degenerates, and thus different codons encode the same amino acids. Second, even where an amino acid substitution is introduced, the mutation can be conservative and have no material impact on the essential function(s) of a protein. See StryerBiochemistry3rd Ed., (c) 1988. Third, part of a polypeptide chain can be deleted without impairing or eliminating all of its functions. Fourth, insertions or additions can be made in the polypeptide chain for example, adding epitope tags, without impairing or eliminating its functions (Ausubel et al.J. Immunol.159(5): 2502-12, 1997). Other modifications that can be made without materially impairing one or more functions of a polypeptide include, for example, in vivo or in vitro chemical and biochemical modifications or the incorporation of unusual amino acids. Such modifications include, for example, acetylation, carboxylation, phosphorylation, glycosylation, ubiquination, labeling, e.g., with radionucleides, and various enzymatic modifications, as will be readily appreciated by those well skilled in the art. A variety of methods for labeling polypeptides, and labels useful for such purposes, include radioactive isotopes such as32P, ligands which bind to or are bound by labeled specific binding partners (e.g., antibodies), fluorophores, chemiluminescent agents, enzymes, and antiligands. Functional fragments and variants can be of varying length. For example, some fragments have at least 10, 25, 50, 75, 100, 200, or even more amino acid residues. Effective amount: A quantity or concentration of a specified compound or composition sufficient to achieve a desired effect in a subject. The effective amount may depend at least in part on the species of animal being treated, the size of the animal, and/or the nature of the desired effect. Gene Cluster: A set of genetic elements grouped together on the chromosome, the protein products of which have a related function, such as forming a natural product biosynthetic pathway. Heterologous: As it relates to nucleic acid sequences such as coding sequences and control sequences, “heterologous” denotes sequences that are not normally associated with a region of a recombinant construct, and/or are not normally associated with a particular cell. Thus, a “heterologous” region of a nucleic acid construct is an identifiable segment of nucleic acid within or attached to another nucleic acid molecule that is not found in association with the other molecule in nature. For example, a heterologous region of a construct could include a coding sequence flanked by sequences not found in association with the coding sequence in nature. Another example of a heterologous coding sequence is a construct where the coding sequence itself is not found in nature (e.g., synthetic sequences having codons different than the native gene). Similarly, a host cell transformed with a construct which is not normally present in the host cell would be considered heterologous for purposes of this disclosure. Homologous amino acid sequence: Any polypeptide which is encoded, in whole or in part, by a nucleic acid sequence that hybridizes to any portion of the coding region nucleic acid sequences. A homologous amino acid sequence is one that differs from an amino acid sequence shown in the sequence listing by one or more conservative amino acid substitutions. Such a sequence also encompasses allelic variants (defined above) as well as sequences containing deletions or insertions which retain the functional characteristics of the polypeptide. Preferably, such a sequence is at least 75%, more preferably 80%, more preferably 85%, more preferably 90%, more preferably 95%, and most preferably 98% identical to any one of the amino acid sequences. Homologous amino acid sequences include sequences that are identical or substantially identical to the amino acid sequences of the sequence listing. By “amino acid sequence substantially identical” it is meant a sequence that is at least 90%, preferably 95%, more preferably 97%, and most preferably 99% identical to an amino acid sequence of reference and that preferably differs from the sequence of reference by a majority of conservative amino acid substitutions. Consistent with this aspect of the invention, polypeptides having a sequence homologous to any one of the amino acid sequences of the sequence listing include naturally-occurring allelic variants, as well as mutants or any other non-naturally occurring variants that retain the inherent characteristics of any polypeptide of the sequences disclosed herein. Homology can be measured using sequence analysis software such as Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, WI 53705. Amino acid sequences can be aligned to maximize identity. Gaps can also be artificially introduced into the sequence to attain optimal alignment. Once the optimal alignment has been set up, the degree of homology is established by recording all of the positions in which the amino acids of both sequences are identical, relative to the total number of positions. Homologous polynucleotide sequences are defined in a similar way. Preferably, a homologous sequence is one that is at least 45%, 50%, 60%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or identical to any one of the coding sequences. Hybridization: Oligonucleotides and other nucleic acids hybridize by hydrogen bonding, which includes Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary bases. Generally, nucleic acid consists of nitrogenous bases that are either pyrimidines (cytosine (C), uracil (U), and thymine (T)) or purines (adenine (A) and guanine (G)). These nitrogenous bases form hydrogen bonds between a pyrimidine and a purine, and the bonding of the pyrimidine to the purine is referred to as base pairing. More specifically, A will hydrogen bond to T or U, and G will bond to C. Complementary refers to the base pairing that occurs between two distinct nucleic acid sequences or two distinct regions of the same nucleic acid sequence. Specifically hybridizable and specifically complementary are terms that indicate a sufficient degree of complementarity such that stable and specific binding occurs between a first nucleic acid (such as, an oligonucleotide) and a DNA or RNA target. The first nucleic acid (such as, an oligonucleotide) need not be 100% complementary to its target sequence to be specifically hybridizable. A first nucleic acid (such as, an oligonucleotide) is specifically hybridizable when there is a sufficient degree of complementarity to avoid non-specific binding of the first nucleic acid (such as, an oligonucleotide) to non-target sequences under conditions where specific binding is desired. Such binding is referred to as specific hybridization. Hybridization conditions resulting in particular degrees of stringency will vary depending upon the nature of the hybridization method of choice and the composition and length of the hybridizing nucleic acid sequences. Generally, the temperature of hybridization and the ionic strength (especially the Na+concentration) of the hybridization buffer will determine the stringency of hybridization, though wash times also influence stringency. Calculations regarding hybridization conditions required for attaining particular degrees of stringency are discussed by Sambrook et al. (ed.)Molecular Cloning: A Laboratory Manual,2nd ed., vol. 1-3, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989, chapters 9 and 11. The following are exemplary sets of hybridization conditions and are not meant to be limiting. Very High Stringency (detects sequences that share 90% sequence identity) Hybridization: 5×SSC at 65EC for 16 hours Wash twice: 2×SSC at room temperature (RT) for 15 minutes each Wash twice: 0.5×SSC at 65EC for 20 minutes each High Stringency (detects sequences that share 80% sequence identity or qreater) Hybridization: 5×-6×SSC at 65EC-70EC for 16-20 hours Wash twice: 2×SSC at RT for 5-20 minutes each Wash twice: 1×SSC at 55EC-70EC for 30 minutes each Low Stringency (detects sequences that share greater than 50% sequence identity) Hybridization: 6×SSC at RT to 55EC for 16-20 hours Wash at least twice: 2×-3×SSC at RT to 55EC for 20-30 minutes each. Isolated: An isolated biological component (such as a nucleic acid molecule or protein) is one that has been substantially separated or purified away from other biological components in the cell of the organism in which the component naturally occurs, such as other chromosomal and extra-chromosomal DNA and RNA, proteins and organelles. With respect to nucleic acids and/or polypeptides, the term can refer to nucleic acids or polypeptides that are no longer flanked by the sequences typically flanking them in nature. Nucleic acids and proteins that have been isolated include nucleic acids and proteins purified by standard purification methods. The term also embraces nucleic acids and proteins prepared by recombinant expression in a host cell as well as chemically synthesized nucleic acids. Mutate: The process of causing a change in the sequence of a genetic material (usually DNA or RNA) of a cell or organism. Mutations can be intentionally introduced into genetic material using molecular techniques well known in the art (e.g., site-directed mutagenesis, PCR mutagenesis and others). Nonribosomal peptide (NRP): A class of secondary metabolites, usually produced by microorganisms, such as bacteria and fungi. Unlike polypeptides synthesized on the ribosome, these peptides are synthesized by nonribosomal peptide synthetases (NRPS) from amino acids. Nonribosomal peptide backbone assembly: The second step in nonribosomal peptide biosynthesis, which includes amide bond formation (condensation) of the peptide sequence. Nonribosomal peptide synthetase (NRPS): A large multi-functional protein that synthesizes polypeptides by a nonribosomal mechanism, often known as thiotemplate synthesis (Kleinkauf and von Doehren Ann. Rev.Microbiol.41: 259-289, 1987). Such nonribosomal polypeptides can have a linear, cyclic, or branched cyclic structure and often contain amino acids not present in proteins or amino acids modified through methylation or epimerization. In particular examples, NRPS produce dipeptides. Nonribosomal peptide tailoring: The third step in nonribosomal peptide biosynthesis. There are numerous novel precursor amino acids found in nonribosomal peptides and many of these building blocks are formed or modified while attached to PCP domains of specialized proteins or the NRPS. This post-synthetic modification can occur after amide bond formation of the peptide backbone. Exemplary modifications include a-carbon epimerization, N-methylation, heterocyclization of Cys or Ser/Thr residues to thiazolines and oxazolines, and side chain halogenation or hydroxylation. Other modifications such as oxidation, alkylation, acylation and glycosylation can occur after release of the nascent peptide from the NRPS complex and are often needed for full biological activity. Nonribosomal precursor amino acid biosynthesis: The first step in nonribosomal peptide biosynthesis. Nonribosomal peptides often possess amino acids not found in peptides and proteins that are assembled on ribosomes. These nonproteinogenic amino acids contribute to the diversity of these peptides and often have roles in their biological activity. Biosynthesis of these amino acids can occur via protein-bound intermediates or as free, soluble species. Nucleic Acid: A deoxyribonucleotide or ribonucleotide polymer in either single or double stranded form, and unless otherwise limited, encompasses known analogues of natural nucleotides that hybridize to nucleic acids in a manner similar to naturally occurring nucleotides. Nucleotide: This term includes, but is not limited to, a monomer that includes a base linked to a sugar, such as a pyrimidine, purine or synthetic analogs thereof, or a base linked to an amino acid, as in a peptide nucleic acid. A nucleotide is one monomer in a polynucleotide. A nucleotide sequence refers to the sequence of bases in a polynucleotide. Oligonucleotide: A plurality of joined nucleotides joined by native phosphodiester bonds, between about 6 and about 300 nucleotides in length. An oligonucleotide analog refers to moieties that function similarly to oligonucleotides but have non-naturally occurring portions. For example, oligonucleotide analogs can contain non-naturally occurring portions, such as altered sugar moieties or inter-sugar linkages, such as a phosphorothioate oligodeoxynucleotide. Functional analogs of naturally occurring polynucleotides can bind to RNA or DNA, and include peptide nucleic acid molecules. Particular oligonucleotides and oligonucleotide analogs can include linear sequences up to about 200 nucleotides in length, for example a sequence (such as DNA or RNA) that is at least 6 bases, for example at least 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100 or even 200 bases long, or from about 6 to about 50 bases, for example about 10-25 bases, such as 12, 15, or 20 bases. Open reading frame (ORF): A series of nucleotide triplets (codons) coding for amino acids without any internal termination codons. These sequences are usually translatable into a peptide. For example, ORF, open reading frame, and enduracidin ORF refer to an open reading frame in the enduracidin biosynthetic gene cluster as isolated fromStreptomyces fungicidicus. The term also embraces the same ORFs as present in other enduracidin-synthesizing organisms. The term encompasses allelic variants and single nucleotide polymorphisms (SNPs). In certain instances the term enduracidin ORF is used synonymously with the polypeptide encoded by the enduracidin ORF and may include conservative substitutions in that polypeptide. The particular usage will be clear from context. An open Reading Frame that has been nulled is an open reading frame that has been rendered non-functional through the deletion, insertion or mutation of one of more nucleotides in the coding sequence. AStreptomyces fungicidicuscomprising a diminished open reading frame-18 (orf 18) is an organism that has a decrease in, such as a 2-fold decrease, or even complete loss of the biological function of the gene product of orf18, relative to a wild typeStreptomyces fungicidicuse.g., through genetic modification of orf18, including the orf18 being nulled as exemplified below, and/or through regulatory manipulation, e.g., modifying, inserting into, removing, and/or replacing non-coding regions of the gene encoding ORF18 that result in a decrease in the expression of the orf18 gene product. For example, the wild type promoter of orf18 could be modified so as to substantially decrease the transcription of orf18. AStreptomyces fungicidicuscomprising an augmented open reading frame-24 (orf24) is an organism that has an increase, such as a 2-fold increase or more, in the biological function of the gene product of orf24, relative to a wild typeStreptomyces fungicidicus, e.g., through genetic modification of or/24 to enhance biological function of the gene product of orf24 and/or by regulatory manipulation, e.g., modifying, inserting into, removing, and/or replacing non-coding regions of the gene encoding ORF24 that result in an increase in the expression of the orf24 gene product. For example, the wild type promoter for orf24 was replaced with a strong constitutive promoter which enhanced the transcription of orf24, as exemplified below. Modified gene: A gene sequence which contains a modification as compared to that found in the naturally occurring (wild-type) gene. Operably linked: A first nucleic acid sequence is operably linked with a second nucleic acid sequence when the first nucleic acid sequence is placed in a functional relationship with the second nucleic acid sequence. For instance, a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence. Generally, operably linked DNA sequences are contiguous and, where necessary to join two protein-coding regions, in the same reading frame. Ortholog: Two nucleic acid or amino acid sequences are orthologs of each other if they share a common ancestral sequence and diverged when a species carrying that ancestral sequence split into two species. Orthologous sequences are also homologous sequences. Polypeptide: A polymer in which the monomers are amino acid residues which are joined together through amide bonds. When the amino acids are alpha-amino acids, either the L-optical isomer or the D-optical isomer can be used, the L-isomers being preferred in some instances. The term polypeptide or protein as used herein encompasses any amino acid sequence and includes modified sequences such as glycoproteins. The term polypeptide is specifically intended to cover naturally occurring proteins (whether produced by ribosomal or nonribosomal mechanisms), as well as those that are recombinantly or synthetically produced. The term polypeptide fragment refers to a portion of a polypeptide that exhibits at least one useful epitope. The phrase functional fragment of a polypeptide refers to all fragments of a polypeptide that retain an activity (such as a biological activity), or a measurable portion of an activity, of the polypeptide from which the fragment is derived. Fragments, for example, can vary in size from a polypeptide fragment as small as an epitope capable of binding an antibody molecule to a large polypeptide capable of participating in the characteristic induction or programming of phenotypic changes within a cell. The term substantially purified polypeptide as used herein refers to a polypeptide that is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. In one embodiment, the polypeptide is at least 50%, for example at least 80% free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. In another embodiment, the polypeptide is at least 90% free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. In yet another embodiment, the polypeptide is at least 95% free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. Probes and primers: Nucleic acid probes and primers can be readily prepared based on the nucleic acid molecules provided in this disclosure. A probe comprises an isolated nucleic acid attached to a detectable label or reporter molecule. Typical labels include radioactive isotopes, enzyme substrates, co-factors, ligands, chemiluminescent or fluorescent agents, haptens, and enzymes. Methods for labeling and guidance in the choice of labels appropriate for various purposes are discussed, e.g., in Sambrook et al. (InMolecular Cloning: A Laboratory Manual, CSHL, New York, 1989) and Ausubel et al. (InCurrent Protocols in Molecular Biology, Greene Publ. Assoc. and Wiley-Intersciences, 1992). Primers are short nucleic acid molecules, preferably DNA oligonucleotides, 10 nucleotides or more in length. More preferably, longer DNA oligonucleotides can be about 15, 17, 20, or 23 nucleotides or more in length. Primers can be annealed to a complementary target DNA strand by nucleic acid hybridization to form a hybrid between the primer and the target DNA strand, and then the primer extended along the target DNA strand by a DNA polymerase enzyme. Primer pairs can be used for amplification of a nucleic acid sequence, e.g., by the polymerase chain reaction (PCR) or other nucleic-acid amplification methods known in the art. Methods for preparing and using probes and primers are described, for example, in Sambrook et al. (InMolecular Cloning: A Laboratory Manual, CSHL, New York, 1989), Ausubel et al. (InCurrent Protocols in Molecular Biology, Greene Publ. Assoc. and Wiley-Intersciences, 1998), and Innis et al. (PCR Protocols, A Guide to Methods and Applications, Academic Press, Inc., San Diego, CA, 1990). PCR primer pairs can be derived from a known sequence, for example, by using computer programs intended for that purpose such as Primer (Version 0.5, © 1991, Whitehead Institute for Biomedical Research, Cambridge, MA). The specificity of a particular probe or primer increases with its length. Thus, in order to obtain greater specificity, probes and primers can be selected that comprise at least 17, 20, 23, 25, 30, 35, 40, 45, 50 or more consecutive nucleotides of desired nucleotide sequence. Protein: A biological molecule expressed by a gene and comprised of amino acids. Purified: The term purified does not require absolute purity; rather, it is intended as a relative term. Thus, for example, a purified protein preparation is one in which the protein referred to is more pure than the protein in its natural environment within a cell. Recombinant: A nucleic acid that has a sequence that is not naturally occurring or has a sequence that is made by an artificial combination of two otherwise separated segments of sequence. This artificial combination can be accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques. “Recombinant” also is used to describe nucleic acid molecules that have been artificially manipulated, but contain the same control sequences and coding regions that are found in the organism from which the gene was isolated. Regulating antibiotic production: To cause an alteration, such as an increase or decrease, in the amount, type or quality of antibiotic production. Disclosed herein are recombinant strains ofStreptomyces fungicidicuswith enhanced enduracidin production. Sequence identity: The similarity between two nucleic acid sequences or between two amino acid sequences is expressed in terms of the level of sequence identity shared between the sequences. Sequence identity is typically expressed in terms of percentage identity; the higher the percentage, the more similar the two sequences. Methods for aligning sequences for comparison are well known in the art. Various programs and alignment algorithms are described in: Smith and Waterman,Adv. Appl. Math.2:482, 1981; Needleman and Wunsch,J. Mol. Biol.48:443, 1970; Pearson and Lipman,Proc. Natl. Acad. Sci. USA85:2444, 1988; Higgins and Sharp,Gene73:237-244, 1988; Higgins and Sharp,CAB/OS5:151-153, 1989; Corpet et al.,Nucleic Acids Research16:10881-10890, 1988; Huang, et al.,Computer Applications in the Biosciences8:155-165, 1992; Pearson et al.,Methods in Molecular Biology24:307-331, 1994; Tatiana et al., (1999),FEMS Microbiol. Lett.,174:247-250, 1999. Altschul et al. present a detailed consideration of sequence-alignment methods and homology calculations (J. Mol. Biol.215:403-410, 1990). The National Center for Biotechnology Information (NCBI) Basic Local Alignment Search Tool (BLAST™, Altschul et al.,J. Mol. Biol.215:403-410, 1990) is available from several sources, including the National Center for Biotechnology Information (NCBI, Bethesda, MD) and on the Internet, for use in connection with the sequence-analysis programs blastp, blastn, blastx, tblastn and tblastx. A description of how to determine sequence identity using this program is available on the internet under the help section for BLAST™. For comparisons of amino acid sequences of greater than about 30 amino acids, the “Blast 2 sequences” function of the BLAST™ (Blastp) program is employed using the default BLOSUM62 matrix set to default parameters (cost to open a gap [default=5]; cost to extend a gap [default=2]; penalty for a mismatch [default=−3]; reward for a match [default=1]; expectation value (E) [default=10.0]; word size [default=3]; number of one-line descriptions (V) [default=100]; number of alignments to show (B) [default=100]). When aligning short peptides (fewer than around 30 amino acids), the alignment should be performed using the Blast 2 sequences function, employing the PAM30 matrix set to default parameters (open gap 9, extension gap 1 penalties). Proteins (or nucleic acids) with even greater similarity to the reference sequences will show increasing percentage identities when assessed by this method, such as at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, or at least 95% sequence identity. For comparisons of nucleic acid sequences, the “Blast 2 sequences” function of the BLAST™ (Blastn) program is employed using the default BLOSUM62 matrix set to default parameters (cost to open a gap [default=11]; cost to extend a gap [default=1]; expectation value (E) [default=10.0]; word size [default=11]; number of one-line descriptions (V) [default=100]; number of alignments to show (B) [default=100]). Nucleic acid sequences with even greater similarity to the reference sequences will show increasing percentage identities when assessed by this method, such as at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% sequence identity. An alternative indication that two nucleic acid molecules are closely related is that the two molecules hybridize to each other under stringent conditions (see “Hybridization” above). Nucleic acid sequences that do not show a high degree of identity can nevertheless encode similar amino acid sequences, due to the degeneracy of the genetic code. It is understood that changes in nucleic acid sequence can be made using this degeneracy to produce multiple nucleic acid molecules that all encode substantially the same protein. Transfected: A process by which a nucleic acid molecule is introduced into cell, for instance by molecular biology techniques, resulting in a transfected (or transformed) cell. As used herein, the term transfection encompasses all techniques by which a nucleic acid molecule might be introduced into such a cell, including transduction with viral vectors, transfection with plasmid vectors, and introduction of DNA by electroporation, lipofection, and particle gun acceleration. Transformed: A transformed cell is a cell into which has been introduced a nucleic acid molecule by molecular biology techniques. The term encompasses all techniques by which a nucleic acid molecule might be introduced into such a cell, including transfection with viral vectors, transformation with plasmid vectors, and introduction of naked DNA by electroporation, lipofection, and particle gun acceleration. Transposon: A mobile genetic element having nearly identical repeating sequences at either end, and containing at least a gene encoding a transposase (the enzyme needed to insert the transposon in the DNA sequence). Transposons can be integrated into different positions in the genome of a cell, or over an isolated plasmid, cosmid, or fosmid DNA template in vitro. Transposons may also contain genes other than those needed for insertion. Vector: A nucleic acid molecule as introduced into a host cell, thereby producing a transfected host cell. Recombinant DNA vectors are vectors having recombinant DNA. A vector can include nucleic acid sequences that permit it to replicate in a host cell, such as an origin of replication. A vector can also include one or more selectable marker genes and other genetic elements known in the art. Viral vectors are recombinant DNA vectors having at least some nucleic acid sequences derived from one or more viruses. A plasmid is a vector. Unless otherwise explained, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The singular terms “a,” “an,” and “the” include plural referents unless context clearly indicates otherwise. Similarly, the word “or” is intended to include “and” unless the context clearly indicates otherwise. It is further to be understood that all base sizes or amino acid sizes, and all molecular weight or molecular mass values, given for nucleic acids or polypeptides are approximate, and are provided for description. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The term “comprises” means “includes.” In case of conflict, the present specification, including explanations of terms, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. Suitable methods and materials for the practice of the disclosed embodiments are described below. In addition, any appropriate method or technique well known to the ordinarily skilled artisan can be used in the performance of the disclosed embodiments. Some conventional methods and techniques applicable to the present disclosure are described, for example, in Sambrook et al.,Molecular Cloning: A Laboratory Manual,2d ed., Cold Spring Harbor Laboratory Press, 1989; Sambrook et al.,Molecular Cloning: A Laboratory Manual,3d ed., Cold Spring Harbor Press, 2001; Ausubel et al.,Current Protocols in Molecular Biology, Greene Publishing Associates, 1992 (and Supplements to 2000); Ausubel et al.,Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology,4th ed., Wiley & Sons, 1999; Harlow and Lane,Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1990; Harlow and Lane,Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1999; and Kieser, T., Bibb, M. J., Buttner, M. J., Chater, K. F., and Hopwood, D. A.: PracticalStreptomycesgenetics, John Innes Centre, Norwich Research Park, Colney, Norwich NR4 &UH, England, 2000. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including explanations of terms, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. III. Engineered Recombinant Expression Vectors ofStreptomyces fungicidicus Disclosed herein are engineered recombinantStreptomyces fungicidicusexpression plasmid vectors. In some embodiments, an engineered recombinantStreptomyces fungicidicusvector comprises at least one selected open reading frame ofStreptomyces fungicidicus. In some embodiments, an engineered recombinantStreptomyces fungicidicusvector comprises at least one selected open reading frame ofStreptomyces fungicidicusexpressed under the control of a promoter. In some examples, the promoter is a strong constitutiveStreptomycespromoter that results in the enhanced production of enduracidin when the vector is expressed in a strain ofStreptomyces fungicidicus. In some embodiments, the open reading frame is operatively linked to a heterologous promoter instead of its own native promoter. For example, it may be operatively linked to a constitutive promoter, such as a strong constitutive expression promoter or an inducible promoter. In some examples, the strong constitutive promoter is ermE*p from the erythromycin producer. In some examples, the inducible promoter is tipA. In some examples, the P(nitA)-NitR system (Herai S, Hashimoto Y, Higashibata H, Maseda H, Ikeda H, Omura S, Kobayashi M, Proc Natl Acad Sci USA. 2004. 101(39):14031-5) or the streptomycete promoter SF14 is employed. In some examples, a native promoter of the apramycin resistant gene (amRp) is employed. In some examples, PhrdB, Ptcp830, PSF14, PermE*and/or Pneos are employed. In some embodiments, the engineered recombinant vector comprises an open reading frame orf 24 (SEQ ID NO: 38) and/or open reading frame orf18 (SEQ ID NO: 37) which has been nulled. In some examples, the open reading frame orf18 (SEQ ID NO: 37) is nulled by an in-frame-deletion, frame-shifting and/or point mutation. In some embodiments, the engineered recombinant vector comprises an open reading frame orf24 from the enduracidin gene cluster ofStreptomyces fungicidicus. In some examples, the open reading frame orf24 (SEQ ID NO: 38) is operatively linked to a heterologous promoter. For example, it is linked to a strong constitutive promoter such as ermE*p. In other examples, the open reading frame orf24 is operatively linked to promoter tipA, SF14, amRp, PhrdB, Ptcp830, PSF14, PermE*and/or Pneos. In another embodiment, an engineered recombinant vector comprises an open reading frame orf18 that resides in the upstream region of the enduracidin gene cluster. The open reading frame orf18 (SEQ ID NO: 37) is nulled by insertional disruption, in-frame deletion, frame-shifting and/or point mutation. In some examples, the open reading frame orf18 is nulled by an in-frame deletion, such as an in-frame deletion as illustrated inFIG.9B. In one example, the open reading frame orf18 (SEQ ID NO: 37) is nulled by an in-frame deletion. For example, the open reading frame orf18 (SEQ ID NO: 37) is nulled by an in-frame deletion of nucleic acids 5 through 660 of orf18 (SEQ ID NO: 37). In general, any internal in-frame deletion over orf18 results in a nulled function of Orf18 due to its incompleteness. In some examples, the in-frame deletion includes deletion of at least 3 nucleic acids in orf18 (SEQ ID NO: 37), such as at least 3 nucleic acids, including 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81, 84, 87, 90, 93, 96, 99, 102, 105, 108, 111, 114, 117, 120, 123, 126, 129, 132, 135, 138, 141, 144, 147, 150, 153, 156, 159, 162, 165, 168, 171, 174, 177, 180, 183, 186, 189, 192, 195, 198, 201, 204, 207, 210, 213, 216, 219, 221, 224, 227, 230, 233, 236, 239, 242, 245, 248, 251, 254, 257, 260, 263, 266, 269, 272, 275, 278, 281, 284, 287, 290, 293, 296, 299, 302, 305, 308, 311, 314, 317, 320, 323, 326, 329, 332, 335, 338, 341, 344, 347, 350, 353, 356, 359, 362, 365, 368, 371, 374, 377, 380, 383, 386, 389, 392, 395, 398, 401, 404, 407, 410, 413, 416, 419, 421, 424, 427, 430, 433, 436, 439, 442, 445, 448, 451, 454, 457, 460, 463, 466, 469, 472, 475, 478, 481, 484, 487, 490, 493, 496, 499, 502, 505, 508, 511, 514, 517, 520, 523, 526, 529, 532, 535, 538, 541, 544, 547, 550, 553, 556, 559, 562, 565, 568, 571, 574, 577, 580, 583, 586, 589, 592, 595, 598, 601, 604, 607, 610, 613, 616, 619, 621, 624, 627, 630, 633, 636, 639, 642, 645, 648, 651, or 654 nucleic acids between nucleic acids 5 through 660 of orf18 (SEQ ID NO: 37). In related embodiments, an engineered recombinant plasmid vector involves two or more open reading frames from the enduracidin gene cluster and/or the regions flanking the gene cluster or from other actinomycete strains. The two or more open reading frames may be linked to a single promoter. Alternatively, they may be operatively linked to two different promoters. The two promoters may be the same type of promoter. Alternatively, they may be two different types of promoters. In further embodiments, additional or alternative open reading frames that may enhance enduracidin production may be introduced, or inactivated, in the engineered strain ofStreptomyces fungicidicus. In some examples, the recombinant plasmid is pXY152-endorf24 (SEQ ID NO:3). In some examples, the recombinant plasmid is pXY300-orf18ifd (SEQ ID NO: 8). In some examples, the recombinant plasmid is pKS-T-orf18ifd (SEQ ID NO: 11). In some examples, the recombinant plasmid is pKS-T-orf18pfrd-AmR (SEQ ID NO: 14). In some examples, the recombinant plasmid is pKS-orf18ifd-T-AmR(NS)(SEQ ID NO: 19). In some examples, the recombinant plasmid is pXY152-endorf24-camtsr (SEQ ID NO: 20). In some examples, the recombinant plasmid is pXY152-endorf24-blatsr (SEQ ID NO: 23). IV. Engineered Recombinant Strains ofStreptomyces fungicidicus Disclosed herein are engineered recombinantStreptomyces fungicidicusstrains capable of producing enhanced enduracidin as compared to a control strain (such as a wild-typeStreptomyces fungicidicusstrain or industrial parent strain). In some embodiments, an engineered recombinantStreptomyces fungicidicusstrain comprises at least one selected open reading frame fromStreptomyces fungicidicusintroduced onto the chromosome and expressed under the control of a promoter, such as a strong constitutiveStreptomycespromoter, that results in the enhanced production of enduracidin in the engineered strain. In some embodiments, the expression of the introduced open reading frame in theStreptomyces fungicidicusis driven by a heterologous promoter instead of its own native promoter. For example, it may be operatively linked to a constitutive promoter, such as a strong constitutive expression promoter or an inducible promoter. In some examples, the strong constitutive promoter is ermE*p from the erythromycin producer. In some examples, the inducible promoter is tipA. In some examples, the P(nitA)-NitR system (see Herai S, Hashimoto Y, Higashibata H, Maseda H, Ikeda H, Omura S, Kobayashi M, Proc Natl Acad Sci USA., 2004. 101(39):14031-5) or the streptomycete promoter SF14 is employed. In some examples, the constitutive expression promoter is amRp. In some examples, PhrdB, Ptcp830, PSF14, PermE*and/or Pneos promoters are employed. In some embodiments, the engineered strain comprises an open reading frame orf24 from the enduracidin gene cluster ofStreptomyces fungicidicus. In some examples, the open reading frame orf24 is operatively linked to a heterologous promoter. For example, it is linked to a strong constitutive promoter such as ermE*p. In other examples, the open reading frame orf24 is operatively linked to promoter tipA, SF14, amRp, PhrdB, Ptcp830, PSF14, PermE*and/or Pneos. In another embodiment, the engineered strain is related to an open reading frame orf18 that resides in the upstream region of the enduracidin gene cluster. The open reading frame orf18 is nulled by insertional disruption, in-frame deletion, frame-shifting and/or point mutation. In some examples, the open reading frame orf18 is nulled by an in-frame deletion, such as an in-frame deletion as illustrated inFIG.9B. In one example, the open reading frame orf18 (SEQ ID NO: 37) is nulled by an in-frame deletion. For example, the open reading frame orf18 (SEQ ID NO: 37) is nulled by an in-frame deletion of nucleic acids 5 through 660 of (SEQ ID NO: 37). In general, any internal in-frame deletion over orf18 should result in a nulled function of Orf18 due to its incompleteness. In related embodiments, the engineered strain involves two or more open reading frames from the enduracidin gene cluster and/or the regions flanking the gene cluster or from other actinomycete strains. The two or more open reading frames may be linked to a single promoter. Alternatively, they may be operatively linked to two different promoters. The two promoters may be the same type of promoter. Alternatively, they may be two different types of promoters. In further embodiments, additional or alternative open reading frames that may enhance enduracidin production may be introduced, or inactivated, in the engineered strain ofStreptomyces fungicidicus. In some embodiments, the engineered strain ofStreptomyces fungicidicusis derived from a wild type parent strain, such as, but not limited to,Streptomyces fungicidicusAmerican Tissue Culture Company (ATCC) 21013. In other embodiments, the engineered strain ofStreptomyces fungicidicusis derived from an industrial parent strain, such as, but not limited to BM38-2 (ATCC PTA-122342). In other embodiments, the engineered strain ofStreptomyces fungicidicusis derived from the conventional mutant strains, such as, but not limited toStreptomyces fungicidicusATCC 31729,Streptomyces fungicidicusATCC 31730 andStreptomyces fungicidicusATCC 31731. In some embodiments, enhanced production of enduracidin is an at least 1.2 fold increase, such as at least 1.5 fold, at least 2 fold, at least 2.5 fold, at least a 3 fold, at least a 3.5 fold, at least a 4 fold, at least a 4.5 fold increase, including, but not limited to a 1.2 to 10 fold increase, a 1.2 to 4.6 fold increase, a 2 to 5 fold increase, such as 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5 and 10 fold increase in enduracidin production as compared to the controlStreptomyces fungicidicusstrain. In some embodiments, the controlStreptomyces fungicidicusstrain is a wild-typeStreptomyces fungicidicusstrain, including, but not limited to,Streptomyces fungicidicusAmerican Tissue Culture Company (ATCC) 21013 or an industrial parent strain, such as, but not limited to, BM38-2 (ATCC PTA-122342), or the conventional mutant strain, such as, but not limited toStreptomyces fungicidicusATCC 31729,Streptomyces fungicidicusATCC 31730 andStreptomyces fungicidicusATCC 31731. In one example, the control isStreptomyces fungicidicusATCC 21013 and the increase in enhanced enduracidin production is at least a 1.2 fold increase, such as a 1.2 to 4.6 fold increase. In one example, the control isStreptomyces fungicidicusBM38-2 (ATCC PTA-122342) and the increase in enhanced enduracidin productions is at least a 1.2 fold increase, such as a 1.2 to 4.6 fold increase. V. Construction of Engineered Recombinant Strains ofStreptomyces fungicidicus In embodiments, recombinant strains ofStreptomyces fungicidicusmay be constructed by integration of a recombinant plasmid comprising at least one enduracidin production enhancing open reading frame into the chromosome of a parent strain ofStreptomyces fungicidicus. The integrative conjugal vector may have, or may be engineered to have, a strong constitutiveStreptomycespromoter. In some embodiments, the plasmid may lack a streptomycete replicon and may be integrated into the chromosome by site-specific single crossover homologous recombination. In other embodiments, the plasmid may be present as a free plasmid. In some embodiments, an conjugal vector may be engineered in which the plasmid insert carries a partially or completely deleted gene of interest, and its flanking regions, that may be integrated into the chromosome after double crossover homologous recombination to generate an in-frame deletion mutant. VI. Production of Enduracidin from Engineered Recombinant Strains ofStreptomyces fungicidicus The engineered recombinant strains ofStreptomyces fungicidicusprovided by the present disclosure provide for methods of producing enhanced levels of enduracidin. This technical advance in the art allows for significant cost savings associated with the production of enduracidin. In some examples, methods of producing enduracidin comprises culturing a disclosed recombinant strain ofStreptomyces fungicidicusunder conditions sufficient for producing enduracidin. In some examples, the method further comprises isolating the enduracidin from the culture medium following culturing. In some examples, the method further comprising determining the antibacterial activity of the produced enduracidin, such as by HPLC analysis or bioassay using theS. aureusATCC 29213 orBacillis subtilisATCC 6633 as indicating microorganisms. In some examples, enduracidin is produced by a disclosedStreptomyces fungicidicusstrain by utilizing fermentation conditions as previously described for the production of enduracidin (Higashide et al.J. Antibiot.21: 126-137, 1968). After production, the compounds can be purified and/or analyzed including HPLC analysis as described in Example 1. Methods of producing enduracidin and harvesting this compound from growth medium can be found in U.S. Pat. No. 4,465,771, which is hereby incorporated by reference in its entirety. In some examples, a disclosedStreptomyces fungicidicusstrain is cultured in tryptic soy broth (TSB) on a shaker (such as at 225 rpm and 30° C. for 48 hours) and then transferred to a enduracidin production medium (EPM, Table 1 below) for a period of time for continuous fermentation, such as for at least five days and up to eleven days, including 5, 6, 7, 8, 9, 10 or 11 days of continuous fermentation. In some examples, production of enduracidin by the wild-type and derivative strains is conducted in automatic fermenters. TABLE 1Enduracidin Production Medium (EPM) Composition (pH 6.7)IngredientConcentration (%)Soluble starch1.5Glucose1.0Corn flour2.5Corn gluten meal2.0Corn steep liquor0.25Sodium chloride0.25NaH2PO41.3KH2PO40.05(NH4)2SO40.15CaCO30.5Lactose0.5ZnCl20.005Chicken oil0.7 In some examples,Streptomyces fungicidicusbiomass is produced by a fermentation process in deep tank sanitary design industrial fermenters with systems to monitor and control pH, temperature, oxygen, aeration, agitation. For example, each fermented batch ofS. fungicidicusis initiated from a characterized and controlled working seed stock of the production seed stored in a secure location and held in low temperature environment. In some examples, the fermentation process occurs in one or more stages, such as following three stages and can optionally be followed by further processing downstream: Stage I: Characterized established working seed cultures are used to start a fermentation batch. One-to-five vials of frozen seed vials are retrieved from low temperature storage and thawed either naturally or placed in a water bath at 28° C.-32° C. until the contents are thawed. The thawed culture(s) are aseptically transferred into sterile water held at room temperature and gently mixed to re-suspend the culture. Stage II: The re-suspended culture is aseptically transferred into 0.005 m3-0.05 m3 seed medium. The seed medium is composed of glucose (0.1-1.0 g/L), Dextrin (0.1-3 g/L), corn steep liquor (0-5.0 mL/L), soybean powder (1-5.0 g/L), ammonium sulfate (0.1-0-0.5 g/L), mono-potassium phosphate (0.13-0.54 g/L), ferrous sulfate (0.00-0.5 g/L), potassium hydroxide (0.13 mL/L), calcium carbonate (1-2 g/L), silicone-based de-foaming agent (0.1 mL/L), water, q. s. The medium is sterilized at 125° C.-128° C. for 30-45 minutes and then cooled to 28° C.-32° C. The volume of medium is adjusted using sterile water to the desired working volume. The pH is adjusted to 6.5-7.0. The operating parameters of the seed scale up cycle include: Incubation temperature of 28° C.±2° C., an internal pressure of 1.0±0.5 kg/cm2, an aeration rate of 3±2 Nm3/min, and agitation rate of approximately 80 rpm, depending upon size and configuration of the vessel. The pH, oxygen consumption and viscosity is monitored but not controlled. The culture is grown for 40-80 hours before transfer into the main production fermenter. The viscosity at the time of transfer should range from 200-600 cps, and the pH should be <6.0, and there should be an increase in oxygen consumption. The seed culture is aseptically transferred into the main fermentation medium to complete the fermentation cycle. Stage III: Production Fermenter medium (10 m3-250 m3) composition includes natural and chemical components such as corn flour (13.0-15.0 w/v %), corn gluten meal (3.0-6.0 w/v %), cotton seed flour (0.1-0.3 w/v %), corn steep liquor (0.1-0.6 v/v %), sodium chloride (0.3 w/v %), ammonium sulfate (0.25-0.6 w/v %), lactic acid (0-0.5 v/v %), zinc chloride (0.01 w/v %), ferrous sulfate (0.0-0.02 w/v %), potassium hydroxide (0.20-0.5 v/v %), calcium sulfate (0.0-0.5 w/v %), calcium carbonate (0.5 w/v %), amylase (0.02-0.06 w/v %), potassium hydroxide (0.05 v/v %), vegetable oil (0.5-2.0 v/v %), de-foaming agent, and water, q. s. The ingredients ae added according to the order listed. Add water to the ingredients then heat to 70-90° C. to allow the enzyme to break down the complex carbohydrates for 15 minutes at temperature. Add remaining ingredients, adjust pH to 6.6-6.8, and add water q. s., sterilize at 125° C.-128° C. for 25-50 minutes to sterilize the media. Cool the media to 25° C.-32° C., and add water to q. s., working volume. Transfer the contents from the seed fermenter into the main fermentation medium and set the fermenter to the following conditions: Temperature 28° C.±3° C., aeration rate 20-60 Nm3/min, internal pressure 0.1-1.0 kg/cm2, agitation rate equivalent to about 1.85 kW/m3. The aeration rate, internal pressure and agitation rates are adjusted a needed to ensure that the dissolved oxygen is not a rate limiting determinate. Carefully control foaming throughout the cycle to prevent contamination or outflow. Start controlling pH after oxygen demand increases. The following parameters are controlled and/or monitored throughout the fermentation cycle: pH, aeration, dissolved oxygen, CO2, viscosity, purity, agitation speed, internal pressure, and residual sugar. Maintain pH at 6.8 until the bacteria growth ceases, then allow pH to change naturally until harvest. The typical fermentation cycle is 210-300 hours. The culture is ready to be harvested when potency is greater than 5,000 μl/L, pH rises to 7.5 or higher, viscosity decreases, and oxygen demand ceases. The fermentation is harvested by heating the culture to 70° C. for 30 minutes to inactivate the bacteria, and then cool the harvest fluids to 25° C.−32° C. In some examples, downstream processing includes removing water from the biomass, drying the biomass and formulating the dried biomass into a premix. Deposits of Biological Material The following biological materials have been deposited under the terms of the Budapest Treaty with The American Type Culture Collection, and given the following accession numbers: DepositAccession NumberDate of DepositBM38-2-18pfrdPTA-124007Mar. 2, 2017BM38-2-24/16PTA-124006Mar. 2, 2017 The above strains have been deposited under conditions that assure that access to the culture will be available during the pendency of this patent application to one determined by the Commissioner of Patents and Trademarks to be entitled thereto under 37 C.F.R. § 1.14 and 35 U.S.C. § 122. The deposits represent substantially pure culture of the deposited strains. The deposits are available as required by foreign patent laws in countries wherein counterparts of the subject application, or its progeny are filed. However, it should be understood that the availability of a deposit does not constitute a license to practice the subject invention in derogation of patent rights granted by governmental action. The following non-liming examples are provided to illustrate certain particular features and/or embodiments. These examples should not be construed to limit the disclosure to the particular features or embodiments described. EXAMPLES Example 1 Materials and Methods for Enhanced Enduracidin Production This example provides representative methods for enhanced enduracidin production. Bacterial Strains, Plasmids, Fosmids and Culture Conditions. Streptomyces fungicidicusB-5477 (ATCC 21013) andEscherichia coliS17-1 (ATCC 47055) were purchased from ATCC. TheS. fungicidicusstrain BM38-2 (ATCC PTA-122342) and standards of enduracidins A and B were provided by Intervet/Merck Animal Health (MAH).E. colistrains DH5a (Life Technologies, Inc.), EP1300 (Epicentre) and XL10-Gold (Stratagene) were used as hosts forE. coliplasmids, fosmids andE. coli-Streptomycesshuttle vectors. Plasmids pSET152 (Bierman et al.,Gene116: 43-49, 1992, which is hereby incorporated by reference in its entirety) and plJ773 were provided by Professor Keith Chater (JIC, Norwich, UK). Plasmid pWHM860 harboring ermE*p promoter was provided by Professor Bradley Moore (UCSD, San Diego). ISP2 (Difco™ ISP Medium 2), ISP4 and TSB (Bacto™ Tryptic Soy Broth) were purchased from VWR. Primers used for PCR and DNA sequencing were synthesized from Fisher and Sigma-Aldrich. Media and culture conditions for growingS. fungicidicuswere described by Higashide et al. (Journal of Antibiotics,21:126-137, 1968). AllE. coliprocedures were performed according to standard protocols. DNA Isolation and Manipulations. To prepare genomic DNA fromS. fungicidicusB-5477, BM38-2 (ATCC PTA-122342) and derivative recombinant and mutant strains for sequencing, fosmid library construction, subcloning and PCR, freshly harvested spores from the individual strains were inoculated and grown in 100 mL TSB liquid medium supplemented with 5 mM MgCl2and 0.5% glycine. The representative culture was conducted in 500 mL Erlenmeyer flasks on a rotary shaking incubator at 225 rpm and 30° C. for 48 to 72 hours. Mycelial cells were harvested by centrifugation at 4000 rpm and 4° C. for 15 minutes. The supernatant was discarded and the pellet was successively washed once with 10.3% sucrose and twice with 10 mM Tris-HCl and 1 mM disodium ethylenediaminetetra-acetate (EDTA), pH 8.0 (TE buffer). The wet cells, equivalent to the volume of 80 μL water were distributed into 1.5 mL sterile micro-centrifuge tubes. After adding 300 μL of the lysis solution containing 200 μL of 10 mM Tris-HCl and 1 mM EDTA, pH 8.0 and 0.3 M sucrose (TES buffer), 50 μL of 0.5 M EDTA, 50 μL of lysozyme (50 mg/mL), the tubes were incubated at 37° C. for 30 to 60 minutes until the solution became viscous. Next, 5 μL of proteinase K (20 mg/mL) and 180 μL of 10% sodium dodecyl sulfate (SDS) were added to each tube. After gentle but thorough mixing, the solutions were incubated at 37° C. for 90 minutes. Then, 80 μL of 10% Cetyl Trimethyl Ammonium Bromide (CTAB) was added. After thorough mixing, the tubes were incubated at 65° C. for 10 minutes. The solutions were extracted twice with 600 μL of phenol/chloroform/isoamyl alcohol (25/24/1). The genomic DNA in the upper aqueous phases were recovered and precipitated with 0.6 volume of isopropanol. The harvested genomic DNA was washed twice with 70% ethanol. After drying at room temperature for 10 minutes, the genomic DNA was dissolved in 50 to 100 μL of sterile water. The high quality of the genomic DNA preparation was confirmed by digestion with HindIII and Sau3AI which showed complete digestion and no degradation of undigested genomic DNA by 0.8% agarose gel electrophoresis. Pooled genomic DNA was further digested with RNase to remove RNA contamination. The purity and quantity of the genomic DNA were determined with a Nanodrop spectrophotometer. General streptomycete DNA manipulations including agarose gel electrophoresis were performed and QIAprep Spin Miniprep kits (Qiagen) were used to prepare plasmids and fosmids fromE. colistrains. Restriction endonucleases, DNA ligase, DNA polymerase, transposase, Klenow enzyme, alkaline phosphatase and ligase were purchased from Biolabs, Invitrogen, Epicentre and Roche, and used according to the manufacturers' recommendations. DNA fragments were purified using QIAquick Gel Extraction kits. PCR. The colony PCR was conducted as follows: spores from independent mutant candidate colonies were inoculated in TSB liquid culture. After growing for 48 to 72 hours, mycelia were harvested by centrifugation and washed twice with TE buffer (10 mM Tris, 1 mM EDTA), pH 8.0. Mycelia were re-suspended in sterile H2O and used as template in PCR reaction mixture in a final volume of 100 μL containing 60 μL of mycelia, 150 μmol of each primer, 20 μL of 5× AccuPrime GC-rich buffer A (Invitrogen), and 1 μL of Polymix (added at 80° C.) from the Expand long template PCR system (Roche). PCR was performed as follows: 1 cycle at 95° C. for 3 minutes, 30 cycles at 95° C. for 1 minute, at 55° C. for 1 minute, and at 72° C. for 2 minutes. The reaction was terminated with one extension cycle at 72° C. for 10 minutes. PCR products were gel-purified and sequenced. General PCR was similarly conducted as described above except that the isolated genomic DNA, plasmid/fosmid DNA was used as template instead of the direct use of DNA released from mycelial colonies without prior purification. Construction of the Integrative Expression Plasmid pXY152-Endorf24 In order to ectopically express the putative regulatory gene orf24 from the enduracidin gene cluster inS. fungicidicuswild-type and BM38-2 (ATCC PTA-122342) strains, orf24 was cloned into the integrative plasmid pXY152 derived from pXY152aR20 (Yin et al.,J. Natural Products,73: 583-589, 2010 which is hereby incorporated by reference in its entirety) orf24 was PCR-amplified fromS. fungicidicusgenomic DNA using the forward primer (End24Ndpf: 5′-CCACCACATATGGAAATAAGTTCGCTCTCCA-3′ (SEQ ID NO:1, Ndel site is in bold) and the reverse primer (End24ERpr:5′-GTGTGTGAATTCCTCGTTCACCCGGCCAGATG-3′ (SEQ ID NO: 2, EcoRI site is in bold). The PCR product was digested with Ndel and EcoRI. The gel-purified orf24 fragment was then ligated with the similarly restricted vector pXY152. The resulting plasmid was designated pXY152-endorf24 (FIG.2; SEQ ID NO: 3). The orf24 insert was confirmed to be error free by sequencing. Construction of Plasmid pXY300-orf18ifd for In-Frame Deletion of Orf18 pXY300-orf18ifd was constructed by cloning two fragments that flank orf18 and are destined for deletion into pXY300, anE. coli-Streptomycesshuttle conjugal temperature-sensitive vector containing the thiostrepton resistance gene (tsr) for selection inS. fungicidicus. An “upstream” 2 kb and a “downstream” 2 kb flanking sequence, designated orf18ifdNP and orf18ifdPH, respectively, that flank orf18 were generated by PCR usingS. fungicidicusgenomic DNA as the template and two sets of primers. Fragment orf18ifdPH was amplified by using the forward and reverse primers (Ifdenorf18pf1, 5′-TTATTGAAGCTTGCCGGGGCCGACGCGGCGGGCGGCCT-3′ (SEQ ID NO: 4), Ifdendorf18pr1, 5′-GTTGTTTTAATTAAACACCAGGCCTCCTGGGGTG-3′ (SEQ ID NO: 5), HindIII and PacI sites are in bold). Fragment orf18ifdNP was amplified by using the forward and reverse primers (Ifdendorf18pf2, 5′-TTTATATTAATTAATGACCCTTCCGTCCCGCCCCCGAT-3′ (SEQ ID NO: 6), Ifdendorf18pr2, 5′ TTTGGTGCTAGCTGGTCGTGGCGCTGTTCC-3′ (SEQ ID NO: 7), PacI and NheI sites are in bold). These two PCR fragments were appropriately restricted and simultaneously ligated with the pXY300 vector prepared by digestion with NheI and HindIII, to yield plasmid pXY300-orf18ifd (FIG.3; SEQ ID NO: 8). The error-free in-frame deletion insert of pXY300-orf18ifd was confirmed by sequencing. Construction of Plasmid pKS-T-orf18pfrd-AmR for Deletion of Orf18 and its Flanking Regions. The oriTfragment was amplified by PCR from plasmid plJ773 using the forward primer (Oritnhexbahd3f, 5′-AGCACAGCTAGCTTCTAGAAGCTTCATTCAAAGGCCGGCA-3′ (SEQ ID NO: 9) HindIII site is in bold) and the reverse primer (Oriter1 pstxhor, 5′-GCCAGTGAATTCTGCAGCTCGAGCAGAGCAGGATTCCCGTTGA-3′ (SEQ ID NO: 10), XhoI site is in bold). The oriTfragment was digested with HindIII and XhoI, gel-purified and then ligated into the similarly restricted vector pBluescript II KS derivative to yield plasmid pKS-T (Alting-Mees and Short, Nucleic acids Research, 17: 9494, 1989). The insert of plasmid pXY300-orf18ifd was excised by digestion with NheI and HindIII, gel-purified and then ligated with NheI and HindIII linearized plasmid pKS-T to afford the plasmid pKS-T-orf18ifd (FIG.4; SEQ ID NO: 11). A 1 kb fragment carrying aac(3)IV, the apramycin resistance gene (amR), was amplified from pIJ773 using forward primer (ApraNcolpf, 5′-GAATGGCCATGGTTCATGTGCAGCTCCAT-3′ (SEQ ID NO: 12), NcoI site is in bold) and reverse primer (ApraBamHlpr, 5′-TCTCGAGGATCCGAATAGGAACTTCGGAAT-3′ (SEQ ID NO: 13), BamHI site is in bold). Digestion of the fragment AmR and plasmid pKS-T-orf18ifd with NcoI and BamHI prepared both the insert and vector for ligation. The resulting plasmid was designated pKS-T-orf18pfrd-AmR (FIG.5; SEQ ID NO: 14). Construction of Plasmid pKS-T-orf18ifd-AmR(NS) for In-Frame-Deletion of orf18. The insert of pXY300-orf18ifd was excised by digestion with NheI and HindIII, gel-purified and then ligated with SpeI and HindIII linearized vector pBluescript II KS to produce a plasmid pKS-orf18ifd. The oriTfragment was amplified by PCR using the forward primer (Oritnhexbahd3f, 5′-AGCACAGCTAGCTTCTAGAAGCTTCATTCAAAGGCCGGCA-3′ (SEQ ID NO: 15), HindIII site is in bold) and the reverse primer (oriTXhNdSpr, 5′-AGGCAGCTCGAGCATATGACTAGTCAGAGCAGGATTCCCGTTGA-3′(SEQ ID NO: 16), XhoI, Ndel and SpeI sites are in bold). The oriTfragment was digested with XhoI and HindIII, gel-purified and then ligated with the similarly restricted plasmid pKS-orf18ifd to obtain a plasmid pKS-orf18ifd-T. A 1 kb fragment carrying aac(3)IV gene conferring apramycin resistance (AmR) was amplified from pIJ773 by PCR using the forward primer (ApraNdepf, 5′-GAATGGCATATGGTTCATGTGCAGCTCCAT-3′ (SEQ ID NO: 17), Ndel site is in bold) and the reverse primer (ApraSpelpr, 5′-TCTAGAACTAGTGAATAGGAACTTCGGAAT-3′ (SEQ ID NO: 18), SpeI site is in bold). Plasmid pKS-orf18ifd-T was linearized by digestion with Ndel and SpeI and then ligated with the similarly restricted fragment AmR to generate the plasmid pKS-orf18ifd-T-AmR(NS) (FIG.6; SEQ ID NO: 19). Intergeneric Conjugation, pXY300-Based and pKS-Based Gene Disruption Procedures. The gene disruption plasmids were individually introduced intoE. coliS17-1 by transformation and then transferred toS. fungicidicusor its derivatives via conjugation. Briefly, freshly harvestedS. fungicidicusspores were pre-germinated andE. coliS17-1 cells were grown overnight at 37° C. in Terrific broth. Serial dilutions of the germinated spore suspension were made and 100 mL of each dilution was mixed with an equal volume ofE. coliS17-1 harboring the pXY300-based disruption plasmids. The solutions were plated onto ISP4 agar plates with addition of 10 mM MgCl2and incubated for 22 hours at either 30 or 37° C. Each plate was overlaid with 3 mL soft nutrient agar containing sodium nalidixate and apramycin (0.5 mg/mL) and further incubated at 30° C. for about one week. Isolated exconjugants that survived antibiotic selection were purified by streaking onto ISP4 agar plates supplemented with sodium nalidixate and apramycin (50 μg/mL each). To conduct the gene disruption studies with the pXY300-based plasmids, exconjugants were first cultured in TSB liquid medium containing apramycin (5 μg/mL) at 30° C. for 24 hours at which time the mycelia were harvested, homogenized and used to inoculate TSB liquid media supplemented with apramycin (5 μg/mL). After 3-6 days incubation at 40° C., the mycelia were homogenized and plated onto ISP4 agar plates containing apramycin (50 μg mL) and incubated at 30° C. for one week. Genomic DNA was isolated from randomly selected individual surviving colonies and analyzed by either PCR or Southern blot to confirm that single- or double crossover disruption had occurred. For pKS-based gene disruption and in-frame-deletion plasmids, exconjugants were passed through three successive rounds of incubations on ISP4 agar plates for sporulation without addition of any antibiotic selection in order to stimulate the conversion to double crossover recombinants. The pKS-based exconjugants were not passed through the 40° C. temperature selection. The correct construction of all mutants was confirmed by PCR and/or Southern blot analysis. Construction of the Integrative Expression Plasmids pXY152-endorf24-camtsr and pXY152-endorf24-Blatsr. To ectopically express orf24 in the apramycin resistant mutant carrying the deletion of orf18 and its flanking regions, the integrative expression plasmid pXY152-endorf24-blatsr was designed. To construct this plasmid, a cassette (camtsr) harboring the chloramphenicol resistance gene and thiostrepton resistance gene (tsr) was excised from a plasmid pUC57 derivative by digestion with SacI and NheI. The camtsr cassette was then ligated with SacI and NheI linearized plasmid pXY152-endorf24 to yield a new construct pXY152-endorf24-camtsr (FIG.7; SEQ ID NO: 20). An ampicillin resistance gene (bla) was PCR-amplified from pBluescript KS using the forward primer (amp2956SwaIpf, 5′-GTGGCAATTTAAATGGAAATGTGCGCGGAA-3′ (SEQ ID NO: 21), SwaI site is in bold) and reverse primer (amp1973SacIpr, 5′-TATATAGAGCTCAACTTGGTCTGACAGTTAC-3′ (SEQ ID NO: 22), SacI site is in bold). bla was then cloned into the SacI and SwaI sites of pXY152-endorf24-camtsr to replace the cassette camtsr with blatsr. The resulting conjugal expression plasmid was designated pXY152-endorf24-blatsr (FIG.8; SEQ ID NO: 23). Construction of the Tn5AT Cassette for In Vitro Transposon Mutation The Tn5AT cassette was designed to combine three genetic elements: the transposon Tn5, oriT and aac3 (IV). Tn5 is specifically and uniquely recognized by Tn5 transposase (Epicentre) and readily inserts into high G+CStreptomycesDNA cloned intoE. coliplasmids and fosmids (also referred to in U.S. Pat. No. 8,188,245 which his hereby incorporated by reference). oriT is required for the conjugal transfer of DNA fromE. coliS17-1 toStreptomycesand aac(3)IV is anE. coli-Streptomycesbifunctional selection marker conferring apramycin resistance. Both oriT and aac3 (IV) were excised from plasmid pIJ773 as a XbaI fragment and then cloned into the transposon donor plasmid pMOD™-2(MCS) (Epicentre), previously linearized with XbaI. The resulting plasmids pXYTn5ATa and pXYTn5ATb only differ by the orientation of XbaI fragment and were used to prepare the Tn5AT cassette by digestion with PvuII according to the manufacturer's specification. In Vitro Transposon Mutation and Selection of Mutagenized Fosmid pXYF24D3 and pXYF148D12 To generate a library of random mutagenized fosmids carrying segments of the enduracidin biosynthesis cluster for gene replacement studies, in vitro transposon insertional mutation studies of fosmids pXYF24 and pXYF148 were performed. Two putative enduracidin biosynthesis regulatory genes, orf18 and orf24, reside on the inserts of fosmids pXYF24 and pXYF148, respectively (GenBank accession no. DQ403252). The in vitro transposon reaction was performed at 37° C. for 2 hours after mixing 10 μL (0.5 μg) fosmid template DNA, 2 μL (20 ng) Tn5AT cassette DNA, 2 μL 10× reaction buffer, 1 μL Tn5 transposase and 5 μL sterile water. Transformation ofE. colicompetent cells EP1300™-T1R(Epicentre) with the transposon reaction mixture was performed by electroporation. Mutagenized fosmids were selected on LB agar plates supplemented with 100 μg/mL apramycin. Plates were incubated overnight at 37° C. and surviving colonies were randomly picked and grown in LB liquid culture with addition of 100 μg/mL apramycin. The mutagenized fosmid DNA from these colonies and control fosmid pXYF24 or pXYF148 were digested with HindIII and analyzed by electrophoresis on 1% agarose gels. The Tn5AT cassette contains a single HindIlI site that is useful when screening for single versus multiple disruption events over the fosmid insert. No HindIII sites are present in the fosmid inserts of pXYF24 or pXYF148, and only one HindIII site is present in the fosmid vector. Hence, digestion with HindIII readily identifies fosmids with a single insertion of Tn5AT by the presence of two bands in the gel. Colonies carrying mutagenized fosmids with a single transposon insertion were randomly selected and grown in LB liquid culture to permit fosmid isolation and identification of the disrupted gene. Screening was conducted by sequence analysis using the primer 5′-AAGGAGAAGAGCCTTCAGAAGGAA-3′ (SEQ ID NO: 24), which corresponds to a region of the apramycin resistance gene. In this manner, fosmid pXYF24D3 and pXYF148D12 were found to have Tn5AT inserted into orf18 at the nucleotide position 26386 and orf24 at the nucleotide position 34333 (GenBank accession no. DQ403252), respectively. Insertional Disruption of Orf18 and Orf24 in the Wild-TypeS. fungicidicusATCC 21013. The gene replacement fosmids pXYF24D3 and pXYF148D12 were separately transformed intoE. coliS17-1 by electroporation and then introduced intoS. fungicidicusby intergeneric conjugation (Mazodier et al.,J. Bacteriology171: 3583-3585, 1989 which is hereby incorporated by reference in its entirety). Exconjugant colonies surviving apramycin selection were passed through three successive rounds of sporulation without antibiotic selection on ISP2 agar plates to create the stable mutant strain via double crossover homologous recombination. The resulting spores were pooled, diluted and plated on ISP2 agar plates supplemented with 50 μg/mL apramycin for confirmation of the apramycin resistance and for use in seed culture and enduracidin production fermentation. The mutant strain with the insertional disruption of orf18 inS. fungicidicuswild-type was designated SfpXYF24D3 and the mutant strain with the insertional disruption of orf24 inS. fungicidicuswild-type was designated SfpXYF148D12. Production of Enduracidin in Laboratory Scale and in 10-Liter Fermenter. Laboratory shake flask fermentation conditions for the production of enduracidin inS. fungicidicuswild-type, BM38-2 (ATCC PTA-122342) and derivative strains were as described by Higashide et al. (J. Antibiotics,21: 126-137, 1968) except for the enduracidin production media which was disclosed in a patent (U.S. Pat. No. 4,465,771). For laboratory scale fermentation, 5 mL TSB was used for inoculation of the seed culture with freshly harvested streptomycete spores. Typically 5 to 10 mL of the seed culture incubated on a rotary shaker at 225 rpm and 30° C. for 48 hours and was then transferred to a 50 mL enduracidin production medium for 10 days continuous fermentation. Production of enduracidin by the wild-type and derivative strains under closely controlled conditions was also conducted in 10-liter automatic fermenters. TABLE 2Comparison of enduracidin (enramycin) yields in wild-type, mutantand genetically engineered strains ofStreptomyces fungicidicusFermentationS. fungicidicusStrainConditionsYield (HPLC)Wild-type (ATCC21013)Shake flask5-30mg/LBM38-2Shake flask60-90mg/LSfpXY52endorf24Shake flask60mg/LSfpXYF24D3Shake flask40mg/LBM38-2.orf18pfrd-Shake flask67mg/LAmRBM38-2.24/16Shake flask30-130mg/LBM38-210 L fermentor80-145mg/LBM38-2.24/1610 L fermentor375mg/L Extraction of Enduracidin from Fermentation Products for HPLC Analysis. To extract the metabolites for HPLC analysis of enduracidin production, the fresh mycelia was harvested by centrifugation and washed with deionized water and re-suspended in 5× volume (ratio of the aqueous methanol (mL) to the wet mycelial weight (g)) 70% aqueous methanol (pH was adjusted to 3.5 with 1 N HCl). The suspension was shaken at 200 rpm at room temperature overnight and then centrifuged at 4000 rpm and 4° C. for 20 minutes. Then 1.4 mL of supernatant from each sample was transferred to individual 1.5 mL microcentrifuge tubes and centrifuged at 13,000 rpm at room temperature for 10 minutes. The filtrate was passed through a 0.22 μm syringe filter and then analyzed by HPLC. Metabolite extraction from mycelia produced in 10 L fermenters was conducted on a small scale equivalent to laboratory fermentations. HPLC Analysis and Enduracidin Yield Determination. A 50 μL HPLC sample prepared as describe above was injected onto a Gemini C18column (5 μm, 4.6×150 mm, Phenomenex, Torrance, CA) attached to a Shimadzu HPLC. Separation was achieved using an 18 min stepwise linear gradients with solvent A: water+0.1% TFA and solvent B: acetonitrile. The flow rate was 1 mL/minute starting with 10% B, increasing to 40% B over 10 min, and then further increasing to 95% B over 8 minutes. The UV region from 200 to 300 nm was scanned with a SPD M20A photodiode array detector. Yields of enduracidins were calculated by comparison with a standard curve constructed from a stock solution of enduracidin standards in 70% methanol. A series of injections including 2, 4, 6, 8, 10 and 12 μg of enduracidin was used to construct the standard curve using the sum of the absorbance areas for enduracidins A and B at 230 nm. A regression equation was generated from the standard curve and used to calculate enduracidin yields. Evaluation of Antibacterial Activity. Staphylococcus aureus(ATCC 29213) was used as an indicating microorganism in the bioassay. Cells were used to inoculate LB broth, grown at 37° C. overnight, and then 100 μL of the culture was mixed with 5 mL of the top agar (mixture of equal volumes of nutrient agar and nutrient broth). The top agar was overlaid onto a nutrient agar plate in which appropriately spaced wells were made by cutting out the agar plugs. Enduracidin standards and aliquots of culture extractions were dissolved or diluted in 50% MeOH at a concentration of 20 μg/mL, and 100 μL of each solution was loaded into the wells. After incubating the plates at 37° C. for 16 hours, the zones of inhibition were observed and compared, and the plates photographed or stored at 4° C. Example 2 Disruption of Orf18 and Orf24 in Wild-TypeS. fungicidicusand Effect on Enduracidin Production This example describes the disruption of orf18 and orf24 in wild-typeS. fungicidicusand the effect on enduracidin production. A 116,000 bp DNA sequence from the wild-typeS. fungicidicusATCC 21013 that harbors the enduracidin biosynthetic gene cluster and its flanking regions (U.S. Pat. No. 8,188,245 which is hereby incorporated by reference in its entirety) was previously identified and is available in GenBank (accession No. DQ403252). Among the 48 annotated orfs are eight putative regulatory genes: orf5, orf12, orf18, orf22, orf24, orf41, orf42 and orf43. To decipher the role of each of the gene products in enduracidin production, fosmid inserts carrying segments of the enduracidin cluster harboring these putative regulatory genes were randomly mutated using a transposon-mediated insertion of an apramycin resistance marker as described in Example 1. The subsequent screening for apramycin resistance and insert location amongE. colicolonies carrying mutagenized fosmids identified pXYF24D3 to carry the disrupted orf18 and pXYF148D12 to harbor the disrupted orf24. A single insertional mutation in each of these fosmids and the site of the insertion was confirmed by sequencing. These two mutagenized fosmids were then individually introduced by conjugation into theS. fungicidicuswild-type strain. Exconjugants showing apramycin resistance were then passed through three rounds of sporulation on ISP2 agar without addition of any antibiotic selection to promote conversion of the single crossover homologous recombination to double crossover mutation. The resulting stable mutant strains SfpXYF24D3 and SfpXYF148D12 were fermented in enduracidin production medium (EPM) on laboratory scale in shake flasks. HPLC analysis of the 70% methanol extraction of the mycelia from 10 days fermentation revealed an increase of 1.3-fold in enduracidin yield by the orf18-disrupted strain SfpXYF24D3 and the complete loss of enduracidin production by strain SfpXYF148D12 having a disrupted orf24. The mycelia extracts were also evaluated for activity towardsS. aureus. The orf18 disruptant SfpXYF24D3 retained activity whereas the orf24 disruptant SfpXYF148D12 lost activity towardsS. aureus. Example 3 Construction of the Recombinant Strain SfpXY152-Endorf24 and Effect on Enduracidin Production This example describes the construction of the recombinant strain SfpXY152-endorf24 and the ability of this strain to produce enduracidin. The loss of enduracidin production in the mutant strain SfpXYF148D12 indicated a possible regulatory role for orf24. A BLAST search of the GenBank database using the Orf24 protein sequence revealed high sequence similarity with a pathway-specific regulatory protein, StrR, involved in streptomycin biosynthesis. A sequence alignment between Orf24 and StrR showed the proteins share a significant similarity (54% aa identity,FIG.9). The loss of enduracidin production upon orf24 disruption and the similarity with StrR indicate that Orf24 may act as a pathway-specific activator in enduracidin production. To explore the role of orf24 as a positive regulatory target for strain improvement, the integrative expression plasmid pXY152-endorf24 (FIG.2) was constructed (Example 1). Plasmid pXY152-endorf24 was introduced into wild-typeS. fungicidicusby conjugation and exconjugants were screened for the apramycin resistance phenotype, leading to the identification of the new recombinant strain SfpXY152-endorf24. At least ten independent exconjugant colonies from this strain were randomly selected and purified. These colony strains carry the pXY152-endorf24 plasmid integrated into an attB site on theS. fungicidicuschromosome by single crossover homologous recombination with the attP site on the plasmid. To investigate the metabolites produced by the recombinant strains, spores from two colony strains were inoculated into TSB seed culture and then transferred to enduracidin production medium for laboratory scale fermentation. HPLC analysis of the 70% methanol extracts of the harvested mycelia revealed a 2-fold increase (60 mg/L) in the enduracidin production by both recombinant strains compared to the wild-type strain (30 mg/L). The elevated yields of enduracidin observed in these colony strains that are capable of overexpressing orf24 is further evidence of the positive regulatory role this gene has in enduracidin production and the results are consistent with those obtained from the disruption of orf24 that led to the loss of enduracidin production. Example 4 Construction of the Strain BM38-2.24/16 Overexpressing Orf24 inS. fungicidicusBM38-2 (ATCC PTA-122342) and Effect on Enduracidin Production This example describes construction of the strain BM38-2.24/16 (ATCC Deposit No. PTA-124006), overexpressing orf24 inS. fungicidicusBM38-2 (ATCC PTA-122342) and effect on enduracidin production. To further explore the positive regulatory role of Orf24, plasmid pXY152-endorf24 was incorporated into the chromosome of the commercial production strainS. fungicidicusBM38-2 (ATCC PTA-122342), as described above for the wild-type organism. Selection of exconjugants exhibiting the apramycin resistance phenotype yielded a number of recombinant colony strains, includingS. fungicidicusBM38-2.24/16, capable of producing elevated enduracidin levels up to 200 mg/L (for a 3.3-fold increase over BM38-2 (ATCC PTA-122342)) in laboratory shake flask cultures.S. fungicidicusBM38-2-24/16 was selected for further evaluation of enduracidin production capacity based on yields during the preliminary screening. Enduracidin production by recombinant strainS. fungicidicusBM38-2.24/16 in laboratory shake flask cultures showed clear potential for significant improvement over BM38-2 (ATCC PTA-122342) and yields were also observed to vary greatly. To more closely control culture conditions over the 10 day growth period, including pH and dissolved oxygen that are not easily managed in shake flasks, production was evaluated through multiple runs in 10 L fermenters. Under these more closely controlled conditions, the yields were more consistent and triplicate 10 L fermentations averaged 375 mg/mL (4.6-fold of BM38-2 (ATCC PTA-122342)). The increased enduracidin yields in the recombinant strain S.fungicidicusBM38-2.24/16 (ATCC Deposit No. PTA-124006) further support a positive upregulation role of Orf24 in enduracidin production. Example 5 Construction of the Deletion Mutant Strain BM38-1.18Pfrd-AmR and the Effect on Enduracidin Production This example describes construction of the deletion mutant strain BM38-2.18pfrd-AmR (ATCC Deposit No. PTA-124007) and the effect on enduracidin production. orf18 is located in the upstream region of the enduracidin biosynthetic gene cluster (GenBank accession no. DQ403252). Orf18 appears to have a negative role in enduracidin production inasmuch as insertional disruption of the gene in the mutant strain SfpXYF24D3 elevated the yield of enduracidin. Based on this observation, constructs were designed for the deletion of orf18 alone and orf18 and portions of its flanking regions. For this purpose, plasmid pKS-T-orf18pfrd-AmR was constructed (FIG.5). This pKS vector-derived plasmid possesses neither a streptomycete replicon nor an element for integration into the streptomycete chromosome. It can only exchange its insert with a defined segment of DNA in the host chromosome via double crossover homologous recombination. The insert map of this plasmid is shown inFIG.10. orf18 and its flanking regions containing the entire orf19 and the region coding for the N-terminal portion of orf17 is deleted in plasmid pKS-T-orf18pfrd-AmR. The 1-kb left arm contains the region coding for the C-terminal portion of orf17 and its downstream region and the 1-kb right arm contains a partial segment of orf20 coding for the N-terminal region. Therefore the deletion after double crossover homologous recombination resulted in a recombinant strain where the entire orf18 plus orf19 and the region coding for the N-terminal portion of orf17 are deleted and replaced with the apramycin resistant gene. Plasmid pKS-T-orf18pfrd-AmR was conjugally introduced intoS. fungicidicusBM38-2 (ATCC PTA-122342) and single and double crossover homologous recombination was promoted on ISP4 agar plates without apramycin supplementation. Exconjugants that were able to survive subsequent apramycin selection were purified and this new recombinant strain was designated BM38-2.18pfrd-AmR (ATCC Deposit No. PTA-124007). Spores from this strain were inoculated into TSB medium for seed culture and then transferred into enduracidin production medium. After 10 days fermentation the mycelia were harvested, processed and analyzed by HPLC. Relative to the parent strain BM38-2 (ATCC PTA-122342), an increase of 1.2-fold in enduracidin production was observed from these laboratory scale fermentations. The relative increase in yield is similar to that observed with the wild-type derived strain SfpXYF24D3 and the results imply that orf19 and orf17, which flank orf18 and were affected in the construction of BM38-2.18pfrd-AmR, have little or no effect on enduracidin production. Therefore, the increased enduracidin production in the recombinant strain BM38-2.18pfrd-AmR is due to elimination of the negative regulatory role of Orf18. Regarding the deletion of orf18 alone with the plasmid pXY300-orf18ifd in BM38-2 (ATCC PTA-122342), difficulties were encountered with positively selecting the exconjugants and single/double mutants with thiostrepton resistance marker. Therefore, alternative vector pBluescript KS II was used to construct the markerless gene replacement delivery plasmids such as pKS-T-orf18ifd (FIG.4) or pKS-orf18ifd-T, pKS-orf18ifd-T-AmR(NS) (apramycin resistance gene is carried on the vector instead of insertion into orf18, seeFIG.6). Example 6 Development of the pKS-Derived Gene Inactivation Vector pKS-T-orf18pfrd-AmR Series This example describes development of pKS-derived gene inactivation vector pKS-T-orf18pfrd-AmR series. A series of pKS-derived gene inactivation vectors were developed (FIGS.4,5and6) that possess the conjugative function and do not require passing transformants through a high temperature selection to eliminate the plasmid as some other gene disruption vectors require. These pKS-derived vectors carry a non-streptomycete replicon allowing replication inE. coliand can maintain and be selected with the apramcyin resistance marker inStreptomycesandE. colior ampicillin inE. coli. They produced copious stable copies of recombinant plasmids inE. colifor conjugation and they have been designed with several rare and unique restriction sites found in streptomycete DNA, such as PacI, HindIII, NheI, and XbaI, that can be conveniently used to assembly the target DNA into the plasmid for insertional gene disruption and in-frame-deletion studies. Example 7 Development of pSET152-Derived Integrative Gene Expression Vectors pXY152-endorf24-camtsr and pXY152-endorf24-blatsr This example describes development of pSET152-derived integrative gene expression vectors pXY152-endorf24-camtsr (SEQ ID NO: 20) and pXY152-endorf24-blatsr (SEQ ID NO: 23). Two new vectors, pXY152-endorf24-camtsr (FIG.7) and pXY152-endorf24-blatsr (FIG.8) were developed. They possess conjugative and integrative functions like vector pSET152, the most widely used integrative vector for streptomycete gene expression and complementation. Both these vectors carry several restriction sites that are rare inStreptomycesDNA for convenient cloning and assembly of the expression construct. Vector pXY152-endorf24-camtsr can be maintained and selected inE. coliwith chloramphenicol at 12.5 μg/mL and inStreptomyceswith thiostrepton at 50 μg/mL. Vector pXY152-endorf24-blatsr can be maintained and selected inE. coliwith ampicillin and inStreptomyceswith thiostrepton. Summary of Examples 1-7 Genetic Manipulation ofStreptomycesRegulatory and Biosynthesis Genes for Strain Improvement Among the numerous microbial producers of natural products, approximately 75% of the known microbial antibiotics are produced by actinomycetes.Streptomyces, Gram-positive filamentous soil bacteria, are members of the actinomycete family and are known for their unrivaled ability to produce a versatile array of structurally diverse, pharmacologically and biologically active secondary metabolites. Polyketides produced by polyketide synthases (PKS) and peptide natural products made by nonribosomal peptide synthetases (NRPS) are representatives. Research on natural product antibiotic biosynthesis has some common challenges: first, how to overcome the typical low production of the parent or structurally modified compounds produced by the wild-type or genetically engineered strains; second, how to activate the many cryptic or orphan secondary metabolite biosynthetic pathways identified from genome sequences so the biological function of the products can be studied. Advances in the study of natural product antibiotic biosynthesis over the past decades have indicated that production of secondary metabolites is regulated by many pathways. For example, the precursor and structural assembly biosynthetic genes (such as PKS and NRPS), regulatory genes and self-resistance genes can be clustered on the bacterial chromosome. Antibiotic production may be regulated by pathway specific regulatory genes, including activators and/or repressors, pleiotropic ectopic regulatory genes, and two-component regulatory systems. Mutations occurring in any of these regulatory genes or systems may increase, decrease or completely abolish antibiotic production. Cryptic biosynthetic pathway can be activated by an unpredicted mutation leading to the production of a previously unknown product. Strain improvement may play an important role in the cost effective industrial scale production of antibiotics or other microbial secondary metabolites. Mutant strains able to produce increased yields of particular metabolites can be generated through random mutations or by targeted disruption of specific genes or by the introduction of gene(s) that eliminate bottlenecks in a biosynthesis pathway. Genetic manipulation of positive and negative regulatory genes, as well as biosynthetic genes, to generate hyper-production of a targeted secondary metabolites has been proven to be a powerful and highly successful strategy of actinomycete strain improvement. In the current disclosure, the positive regulatory role of orf24 and the negative regulatory role of orf18 on enduracidin production was demonstrated. Targeted insertional inactivation of orf24 resulted in a complete loss of enduracidin production in the recombinant strain SfpXYF148D12. Subsequent overexpression of orf24 under the control of the strong constitutive promoter ermE*p in the recombinant strains SfpXY152-endorf24 and BM38-2.24/16 led to increases in enduracidin yields of approximately 2 to 4.6-fold. The deletion of orf18 and its flanking regions, including the entire orf19 and a portion of orf17, increased enduracidin yields by 1.2-fold. These results provided strong genetic evidence in support of the roles of orf24 and orf18 as positive activator and negative repressor, respectively, in enduracidin biosynthesis. Orf24 Orthologs have been Functionally Confirmed from Other Antibiotic Biosynthesis Pathways A BLAST query with Orf24 protein sequence against GenBank database revealed hundreds of hits (GenBank accession no. DQ403252). Many show very high amino acid similarity (from 60% to 99% identities) and are annotated as transcriptional regulators in the biosynthesis of the aminoglycoside antibiotic streptomycin. However, none of this group of genes has had the function verified experimentally. Analysis of the BLAST results identified several related proteins that share a lower similarity (over 40% but below 60% aa identity) to Orf24 that were functionally characterized. These include the well-characterized protein StrR which shares a lower but significant similarity (54% aa identities in 311 aa overlap) with Orf24. StrR has been genetically and biochemically demonstrated to function as a pathway specific positive activator of the expression of the streptomycin biosynthesis genes inStreptomyces griseus. StrR represents a family of pathway-specific activators, a handful of which have been characterized by either genetic manipulation or biochemical studies.FIG.11shows the alignment of Orf24 with six functionally confirmed actinomycete StrR-like proteins. A typical and highly conserved helix-turn-helix (HTH) DNA-binding domain is present in all seven proteins as underlined inFIG.11. Orf24 also shares a significant sequence similarity (54% aa identities) to Teil15*, a pathway specific activator governing biosynthesis of the nonribosomally generated glycopeptide antibiotic teicoplanin. Teil5* positively regulates the transcription of at least 17 genes in the teicoplanin cluster. The wild-typeActinoplanes teichomyceticusproduces about 100 mg/L of teicoplanin whereas the genetic recombinant strains, derived from the parentA. teichomyceticusand carrying tei15*expressed under the control of different promoters, increased teicoplanin yield to 1 g/L in the case of ermE*p promoter and to 4 g/L in the case of the native apramycin resistance gene promoter. As illustrated inFIG.11, Orf24 also shares a significant sequence similarity (54% aa identities) to Bbr, from the balhimycin glycopeptide antibiotic biosynthesis cluster; to KasT (50% aa identities) governing the expression of aminoglycoside antibiotic kasugamycin biosynthesis genes; and NovG (45% aa identities) the pathway specific activator involved in novobiocin biosynthesis. The ΔnovG mutant produced only 2% as much novobiocin as wild-type and overexpression of novG from a multi-copy plasmid in the recombinant strain led to a three-fold increase in the novobiocin production. Orf24 also shares 42% aa identities with SgcR1, one of four regulator genes (sgcR1, sgcR2, sgcR3 and sgcR) experimentally confirmed to be involved in production of the antitumor antibiotic C-1027 inS. globisporus. Overexpression of sgcR1 inS. globisporusSB1022 increased the C-1027 yield approximately seven-fold compared to the wild-type strain. Overexpression of the positive regulator sgcR3 in a recombinant strain resulted in a 30-40% increase in C-1027 production. In contrast, inactivation of the negative regulator sgcR led to increases both C-1027 and heptaene production. Moreover, overexpression of sgcR1 in the ΔsgcR mutant strain led to about a seven-fold increase of C-1027 production. sgcR3 occupies a higher level regulation by control of sgcR1 and sgcR2 in the hierarchy regulation of C-1027 production. In conclusion, the disruption and expression effects of orf24 and the comparison of Orf24 with other functionally characterized orthologs indicate Orf24 acts as a pathway specific positive regulator/activator in enduracidin production. Orf18 is a Putative Atypical Orphan Response Regulator and Aligns with Functionally Confirmed Orthologs Production of antibiotics inStreptomycesspecies is tightly regulated by complex genetic networks that limit the ability of many wild-type antibiotic producers from generating yields necessary for large-scale, cost-effective industrial production. One important regulatory mechanism is the two-component signal transduction systems. Two-component systems include a sensor kinase and a cognate response regulator. The sensor kinase responds to specific external environmental stimuli/signals such as stress, nutrition and chemicals, etc., and then relays the signal to a cytoplasmic response regulator that triggers and activates the transcription of target genes. A response regulator that is unpaired with a sensor kinase is designated an orphan response regulator. Two-component systems and orphan response regulators are present in streptomycete genomes and can function to repress secondary metabolite production. In the enduracidin gene cluster fromS. fungicidicus, orf18 encodes a putative orphan response regulator that shares a low to moderate sequence similarity to three other characterizedStreptomycesresponse regulators including one orphan response regulator, SC03818, fromS. coelicolor(FIG.12). Orf18 has a longer N-terminal sequence compared to the other aligned proteins and appears to be an atypical orphan response regulator because a highly conserved lysine at position 118 (relative to the common position 105) is absent in Orf18 and replaced with a threonine. The lysine is proposed to be required for forming the phosphorylation pocket. Only a few streptomycete response regulators have been functionally characterized. TheS. coelicolorgenome contains a total of five atypical and seven typical orphan response regulators. Orf18 shares 26% aa identities in 191 aa overlap with AbsA2. The deletion of AbsA2 inS. coelicolorresulted in increased production of two antibiotics, actinorhodin and undecylprodigiosin. Orf18 shows 32% aa identities in 176 aa overlap with SC03818. Deletion of sco3818 led to enhanced production of actinorhodin. Orf18 shares 29% aa identities in 166 overlap aa with SCO1745 (AbrA2). Deletion of the AbrA2-containing-response regulator operon resulted in 100% increase of the antitumor antibiotic oviedomycin in the recombinant strainS. coelicolorM145 compared to the wild-type producer. The observed negative regulatory role of Orf18 in enduracidin production is consistent with the demonstrated activities of the related negative regulators (FIG.12). In addition, it is noticed that Orf18 shares the highest protein sequence similarity with the members of the LuxR family of transcriptional regulators in the BLAST search. Absence of Polar Effects in the Mutant BM38-1.orf18pfrd-AmR The deleted region in the mutant BM38-1.18pfrd-AmR strain involves three genes, orf18, the region coding for the N-terminal portion of or17 located downstream of orf18, and the entire orf19 located upstream of orf18 (FIGS.5and10). orf17 is predicted to encode a ribonuclease apparently having no function related to the biosynthesis or regulation of enduracidin. Also, the apramycin resistance gene replacing orf18 and its flanking region is transcribed divergently with orf17 and should not create any read-through events from the apramycin resistance gene promoter. Therefore, there should be no polar effects resulting from the partial deletion of orf17. orf19 is transcribed and translated in the same direction as orf18. This gene is annotated to encode a protein of unknown function. The mutant strain SfpXYF24D3 carrying the disruption of orf18 alone and the mutant BM38-1.18pfrd-AmR carrying the deletion of orf18 and orf19 together have similarly enhanced effects on enduracidin production which implies orf19 has no role or a negligible role in enduracidin production. The gene orf20 is located upstream of orf19 and transcribed and translated in the same direction as the inserted apramycin resistance marker (FIG.10) orf20 is still intact in BM38-1.18pfrd-AmR and the product apparently does not have a role in enduracidin production. Therefore any polar effects on the expression of orf20 are not believed to be responsible for the enhanced enduracidin production in BM38-1.18pfrd-AmR. Example 8 Further Applications and Manipulations of Orf24 and/or Orf18 for Enhanced Enduracidin Producing Strains In addition to the examples provided above, there are other possible ways to utilize the regulatory roles of orf24 and orf18 to improve the enduracidin production. i. Expression of Orf24 Under an Alternative, Constitutive or Inducible Overexpression Promoter pXY152-endorf24 (shown inFIG.2) was constructed for the integrative ectopic expression of orf24 under the control of ermE*p, a widely used streptomycete strong constitutive expression promoter. The overexpression of orf24 may also be driven by other constitutive or inducible promoters. The tipA promoter is a thiostrepton inducible overexpression streptomycete promoter. A multicopy tipA promoter-containingE. coli-Streptomycesshuttle plasmid, pXY200, was developed that has been successfully used for overexpression of streptomycete genes. For applications relevant to this disclosure, the tipA promoter can be excised from pXY200 and cloned into pXY152 to replace ermE*p and drive the expression of orf24. Likewise, orf24 can be easily transferred from pXY152-endorf24 to pXY200 for plasmid-based expression. Other promoter options include, but are not limited to, the P(nitA)-NitR system and the streptomycete promoter SF14. Recently, the integrative plasmid pKC1139 and the native promoter of the apramycin resistant gene were successfully used to express regulatory genes for hyperproduction of the peptide antibiotic teicoplanin. The regulatory gene sanG encodes a pathway specific activator for nikkomycin production. The expression of an extra copy of sanG under the control of five different promoters (PhrdB, Ptcp830, PSF14, PermE*and Pneos) led to increases in nikkomycin yields by 69%, 51%, 26%, 22%, and 13%, respectively (see Du et al., Applied Microbiology and Biotechnology 97: 6383-6396, 2013). ii. Double Mutant Strains ofS. fungicidicuswith Deletion of Orf18 and Overexpression of Orf24 With both the orf18 deletion mutant and the orf24 overexpression strains exhibiting increased enduracidin production, a double mutant containing both can be generated and whether an additive effect on the yield of this peptide antibiotic is observed. The double mutant can be created by introducing the overexpression plasmid pXY152-endorf24-blatsr (FIG.8) into the mutant BM38-2.18pfrd-AmR. pXY152-endorf24-blatsr is a conjugal integrative plasmid carrying a thiostrepton resistance gene (tsr) for selection in streptomycetes and ampicillin resistance gene (bla) for selection inE. coli. Because theE. colistrain S17-1 used for conjugation is naturally resistant to chloramphenicol (cam), the chloramphenicol resistance marker in pXY152-endorf24-camtsr (see above) has been replaced with ampicillin resistance (bla) in order to select S17-1 transformants. Alternatively, pXY152-endorf24-camtsr and derivatives can be introduced into streptomycetes by using a different conjugalE. colistrain, ET12567/pUZ8002. Using either plasmid pXY152-endorf24-blatsr or pXY152-endorf24-camtsr to introduce the second copy of orf24 into the orf18 deficient mutant, it is possible to select for the double mutant by thiostrepton resistance. To generate a null orf18 in-frame-deletion mutant in BM38-2 (ATCC PTA-122342), plasmids pXY300-orf18ifd (FIG.3) and pKS-orf18ifd-T-AmR(NS) (FIG.6) were constructed for this purpose. pXY300-orf18ifd allows for selection of the orf18 in-frame deletion mutant with thiostrepton while pKS-orf18ifd-T-AmR(NS) uses apramycin to select in-frame deletion mutants. Although mutant strains of wild-typeS. fungicidicusare readily selected using the thiostrepton resistance marker, difficulties have been encountered using this resistance marker in the BM38-2 (ATCC PTA-122342) strain. Thus, two plasmids, pXY300-orf18ifd and pKS-orf18ifd-T-AmR(NS), were constructed for the same purpose. In view of the many possible embodiments to which the principles of the disclosed invention may be applied, it should be recognized that the illustrated embodiments are only preferred examples of the invention and should not be taken as limiting the scope of the invention. Rather, the scope of the invention is defined by the following claims. We therefore claim as our invention all that comes within the scope and spirit of these claims.
122,087
11858968
DETAILED DESCRIPTION Unless defined otherwise herein, all technical and scientific terms used in this disclosure have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure pertains. Every numerical range given throughout this specification includes its upper and lower values, as well as every narrower numerical range that falls within it, as if such narrower numerical ranges were all expressly written herein. The disclosure includes all steps and compositions of matter described herein in the text and figures of this disclosure, including all such steps individually and in all combinations thereof, and includes all compositions of matter including but not necessarily limited to all yeast strains, progeny of the yeast, fusions of yeast cells and non-yeast cells, progeny of yeast cell cultures, and the medium in which the modified yeast are grown and/or preserved. Because they serve as a central interface for hundreds of other proteins, histones are among the most conserved genes in eukaryotes (Talbert and Henikoff, 2010). They serve central cellular roles by regulating genome access, DNA compaction, transcription, replication, and repair (Talbert and Henikoff, 2017). Whereas higher eukaryotes evolved myriad histone variants with specialized functions,Saccharomyces cerevisiae(budding yeast) encodes but a few, a simplicity that has facilitated many fundamental discoveries in chromatin biology (Rando and Winston, 2012). In this disclosure we investigated why budding yeast have streamlined chromatin compared to humans, and analyzed whether differences in histone sequences reflect functional divergence (FIG.1A). Without intending to be constrained by any particular theory, it is considered that the disclosure demonstrates use of yeast serve as a “chassis” for understanding how histone variants exert control over cellular transcription. Thus, in embodiments, the disclosure relates to providing modifiedS. cerevisiaethat utilizes core histones from humans. It is believed that, prior to the present disclosure, only single yeast genes have been individually humanized (Hamza et al., 2015; Kachroo et al., 2015; Laurent et al., 2016; Osborn and Miller, 2007), but never a whole protein complex, nor one of such central importance as histones. In this regard, the present disclosures could have revealed intrinsically different properties between yeast nucleosomes and those from humans. We initially suspected two possible outcomes: i) that human histones could be used in place of yeast histones because of their high conservation, suggesting that histone sequence divergence provides only minor functional differences; or ii) that human histones in yeast would very poorly complement or even fail entirely, suggesting that the divergent residues are highly optimized for each species and serve specialized or novel functions. Our results are consistent with the latter. In particular, the humanized yeast as described further below had pronounced delays in adapting to new environmental conditions, likely due to slowed remodeling of human core nucleosomes. In addition, our results suggest that human core nucleosomes may have evolved to occupy DNA more tenaciously, as we observed reduced RNA content and greater DNA occupancy by MNase-seq. Results presented herein suggest that yeast may maintain human chromatin even when given access to native yeast histones. This may represent a type of chromatin “memory”, whereby cells partition and reproduce parental chromatin to new daughter cells. Thus, while the species-specific coevolution of histones and their associated protein networks is extensive, the present disclosure demonstrates that it is nonetheless possible to reprogram the epigenome of at least one organism to accept histones of a very distant relative. In view of these results, the disclosure provides in certain aspects compositions and methods for modifying yeast chromatin, and yeast comprising modified chromatin. While these aspects are illustrated by fully and partially replacing histones ofSaccharomyces cerevisiaewith human histones, given the benefit of this disclosure, those skilled in the art can adapt these representative embodiments to generate modified yeast having endogenous histones fully or partially replaced, or optimized for any particular phenotype or other characteristics, with histones from a variety of other eukaryotic organisms/cell types. Such other eukaryotic organisms and cell types can include but are not necessarily limited to yeast other thanS. cerevisiae, including for example, pathogenic fungi, or any other fungi of interest, any single-celled eukaryote such as amoeba, or any animal, including but not necessarily limited to mammals, and any plant. In embodiments, the disclosure includes producing human artificial chromosomes (HACs) in yeast using the compositions and methods of this disclosure. Yeast comprising the HACs are also included. The HACs comprise yeast chromosomes that are modified such that at least one of yeast histones H3, H4, H2A or H2B are fully or partially replaced by their human histone counterparts, i.e., human H3, H4, H2A or H2B, respectively. Such yeast histone replacements can be adapted so that histones from any other cell eukaryotic cell type can be incorporated into yeast chromosomes. Thus, in embodiments, the disclosure relates to replacing all or some histones or some portions of histones in budding yeast with some or all non-endogenous histone counterparts from a distinct organisms(s). As discussed above, in embodiments, the histone counterparts are human histones, which may comprise certain amino acid substitutions, as described further below. In embodiments, the human histone sequences in partially replaced yeast histones comprise one or more yeast histone amino acids from the yeast homologous histone (i.e., a swap-back mutation). Thus, a partially replaced yeast histone comprises a histone that is a non-yeast histone, but retains at least some yeast histone amino acids, when taken in context of the wild type yeast histone amino acid sequence. In embodiments, a partially replaced yeast histone comprises only, or at least, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more yeast amino acids, provided that the partially replaced yeast histone has an amino acid sequence that is distinct from an unmodified yeast histone amino acid sequence, and contains histone amino acids from a non-yeast source, such as human histones. In embodiments, the human or other non-yeast chromosomes comprise histones that have one or more amino acid modifications. The modifications can comprise, for example, amino acid deletions, insertions, and substitutions. In embodiments, the human or other non-S. cerevisiaehistone comprises a swapped-back amino acid, thus providing a partially humanized (or other non-S. cerevisiae) histone comprisingS. cerevisiaeand non-S. cerevisiaehistone amino acids. Suitable swap-back mutations can be identified using approaches described herein, and all specific amino acid residue modifications identified herein, and all combinations of such modifications, are included in the invention. Any individual or combination of mutations can also be excluded from the invention. Human and yeast histone amino acid sequences are known in the art. The disclosure includes all human and yeast histone amino acid sequences. Representative and non-limiting examples of human histone amino acid sequences are available via GenBank accessions numbers, as follows: H3.1—NP_003522.1; H3.3—NP_002098.1; H4—NP_003539.1; H2A.1—NP_003501.1; H2B.1—NP_066406.1. Representative and non-limiting examples of yeast histone amino acid sequences are available via GenBank accessions numbers, as follows: H3—NP_014367.1; H4—NP_014368.1; H2A—NP_010511.3; H2B—ONH73823.1. In embodiments, yeast H2 histone is fully or partially replaced with human H2A.1 or human H2B.1 histone. In embodiments, yeast H3 histone is fully or partially replaced with human H3.1 or H3.3 histone. In embodiments, yeast H4 histone is partially or fully replaced with human H4 histone. All of the amino acid sequences associated with the GenBank accession numbers are incorporated herein by reference as they exist on the filing date of this application or patent. The disclosure includes amino acid sequences that are from 80%-99% similar to those amino acid sequences, and includes amino acid sequences that include insertions and deletions, provided yeast comprising such modified histones are viable. The disclosure includes all polynucleotide sequences encoding the histone proteins, and all sequences complementary to those sequences. As is known in the art, by convention, human histone amino acid residue numbers generally do not include the Met in the residue numbering. In embodiments, for human H3, H4, and H2B, this convention is followed in this disclosure, with the exception of human H2A, for which the first Met is counted in the residue numbering, such as inFIGS.3C and11A. In embodiments, the disclosure includes modified yeast comprising a mutation in the yeast DAD1 gene, such as a mutation that changes DAD1 at residue 50, such as an E to D amino acid substitution (referred to herein as DAD1-E50D). As a consequence of this change, the yeast DAD1 protein comprises the following amino acid sequence: MMASTSNDEEKLISTTDKYFIEQRNIVLQEINETMNSILNGLNGLNISLSSIAVGREF QSVSDLWKTLYDGLESLSDEAPIDEQPTLSQSKTK (SEQ ID NO:20) wherein the D at position 50 is in enlarged and bold font. In embodiments, the mutation in the DAD1 gene comprises a missese mutation in the DAD1 gene, as set forth in Table 1 below. Thus, in embodiments, yeast of this disclosure comprise a mutated DAD1 protein, wherein the protein comprises an E50D mutation. In an embodiment, the disclosure comprise a modified yeast with a human H3 histone amino acid sequence, except for swapping back only two yeast amino acids. In an embodiment, such a sequence comprises an H3kk mutation comprising the sequence: MARTKQTARKSTGGKAPRKQLATKAARKSAPATGGVKKPHRYRPGTVALREIRRY QKSTELLIRKLPFQRLVREIAQDFKTDLRFQSSAVMALQEACEAYLVGLFEDTNLCAI HAKRVTIMKDILARRIRGERA (SEQ ID NO:17), wherein the two yeast K amino acids swapped back to the human histone sequence are in enlarged, bold font. In this sequence, the yeast K amino acid replaces the human P and Q amino acids, respectively in the N to C direction. Per convention for this H3, the first Met is not counted, thus the positions in this amino acid sequence are K121 and K125. In an embodiment, the disclosure comprises a modified yeast with a human H2 histone (H2Ac) sequence comprising: MSGRGKQGGKARAKAKTRSSRAGLQFPVGRVHRLLRKGNYAERVGAGAPVYLAA VLEYLTAEILELAGNAARDNKKTRIIPRHLQLAIRNDEELNKLLGKVTIAQGGVLPNILLPKKTESHHKAKGK (SEQ ID NO:18). In this sequence, the human QAV amino acids at positions 113, 114 and 115 (QAV) counting the first Met are replaced with HQN. The swapped back yeast amino acids are shown in enlarged, bold font. In an embodiment, the disclosure provides a single epigenetic element, such as a YAC or other plasmid, encoding human histones H3.1, H3.3, H4, H2A, H2B, or histones comprising yeast swap back mutations or other amino acid changes as described herein, wherein the single epigenetic element is present in a yeast comprising the DAD1-E50D mutation. In embodiments, modified yeast described herein comprise human histones with mutations selected from the following: in human H3, a replacement of human H3 amino acids selected from: human T22 with S; human R42 with K; human Y45 with F; human A87 with S; human A95 with S; human S96 with V; human G102 with S; human 110C with A; human 120M with Q, human 121P with K; human 125Q with K; human 130I with L; human 135A with S; and combinations thereof, and wherein optionally only human→yeast P121K and Q125K are changed; and/or in human H4, a replacement of human H4 amino acids selected from: human T54 with V; human G56 with A; human V60 with S; human N64 with S; human A69 with S; human A82 with S; human M84 with L; and combinations thereof; and/or in human H2A a deletion of at least one of R, Q and R in positions 3, 5 and 12, respectively, of the human H2A histone sequence; or a replacement of human H2A amino acids selected from: human K16 with Q; human T17 with S; human S19 with A; human R20 with K; human K36 with R; human E41 with Q; human V43 with I; human A45 with S; human A52 with T; human T59 with A; human E91 with D; human K99 with N; human Q112 with H; human A113 with Q; human V114 with N; human T115 with S; human S122 with K; human H123 with K; human H124 with T; human K126 with S; human G127 with S; human K128 with E; human V114 with N; and combinations thereof, and/or, in human H2B: a deletion of one or both human H2B amino acids GF in the contiguous sequence ISGFKK (SEQ ID NO:19); and/or a replacement of human H2B amino acids selected from: human P5 with E; human E6 with K; human V7 with K; human S8 with P; human S9 with A human K10 with S; human G11 with K; human T13 with P; human I14 with A; human S15 with E; human K18 with P; human K19 with A; human V21 with K; human V22 with K; human K23 with T; human T24 with S; human Q25 with T; human K26 with S; human K27 with T; human E28 with D; human K33 with S; human R34 with K; human T35 with V; human S39 with T; human 142 with S; human V51 with T; human S59 with Q; human A61 with S; human M65 with L; human T70 with N; human S78 with T; human R83 with K; human H85 with A; human S87 with N; human R89 with K; human S94 with A; human L104 with I; human K119 with R; human T126 with S; human K128 with T; and/or any mutation listed in Table 1 or 2. The disclosure also includes discovering, generating, and/or engineering mutations in other yeast genes (i.e., non-histone genes) that may suppress negative effects caused by or correlated with the presence of the non-endogenous histone(s). In embodiments, such suppressor mutations are present in genes directly or indirectly related to cell cycle regulation. In embodiments, the disclosure comprises fusing a modified yeast cell comprising fully or partially modified histones with a distinct cell type, thereby facilitating “uploading” the modified chromosomes into the distinct cells. In embodiments, the distinct cell types are non-S. cerevisiaeeukaryotic cells. In embodiments, the distinct cell types are non-S. cerevisiaefungi, including but not limited to pathogenic fungi. In embodiments, the distinct cell types to which modified yeasts of this disclosure are fused are animal cells. In embodiments, the cells are human, non-human primate, or porcine cells. In embodiments, the cells are stem cells. In embodiments, the stem cells are totipotent, pluripotent, multipotent, or oligopotent stem cells when the modified histones are introduced. In embodiments, the cells are pluripotent, and the pluripotency of the cells is induced, such as by using exogenous genes or compounds. In embodiments, the cells are neural stem cells. In embodiments, the cells are hematopoietic stem cells. In embodiments, the cells are leukocytes. In embodiments, the leukocytes are of a myeloid or lymphoid lineage. In embodiments, the cells are embryonic stem cells, or adult stem cells. In embodiments, the cells are epidermal stem cells or epithelial stem cells. In embodiments, the modified cells of this disclosure are allowed to differentiate, and/or are coaxed into differentiation, such as into an organism, organ, or tissue. In embodiments, the cells are differentiated cells when the modified histone(s) is/are introduced. In embodiments, the cells are cancer cells, or cancer stem cells. In certain approaches, cells produced according to this disclosure are maintained as cell lines. In embodiments, cells of this disclosure comprise one or more histone mutations, and may be at least for a period of time heterozygous or homozygous for such mutated histones. In certain embodiments, cells modified to comprise non-endogenous histones according to this disclosure are engineered to produce a protein or other compound, such as an antibody, and the cells themselves or the protein or compound they produce is used for prophylactic or therapeutic applications, or industrial applications, including but not necessarily limited to food and beverage technologies. In embodiments, one or more histone mutations or other genes as described herein can be made by direct modification of an endogenous histone-encoding or other gene, such as by CRISPR-mediated gene editing. The histone mutants, or non-endogenous histone can also be introduced on a plasmid or “neochromosome”, which comprises a yeast artificial chromosomes containing essential components for replication. In certain approaches, cells modified according to this disclosure are such that they comprise fully or partially non-endogenous histones and are used for screening any of a wide variety of test compounds. In embodiments, the cells are modified such that the histones comprise, in addition to non-endogenous histone amino acid residues, at least one mutation or other genetic or metabolic feature that is causative of or is correlated with a particular disease, disorder, or condition. In embodiments, a plurality of cells modified to comprise non-endogenous histones as described herein is contacted with distinct test agents, wherein a change in any characteristic of the cells as a result of being contacted with the test agent indicates the test agent is a candidate for eliciting a similar response in cells that are the source of the histones used to modify yeast of this disclosure. By way of non-limiting example,S. cerevisiaecan be modified to comprise, for instance, full or partial histones that are endogenous to pathogenic fungi such asCandidaspp.,Pneumocystisspp., and/orCryptococcusspp. Such modified budding yeast can be used to test candidate agents for use as anti-fungal agents, wherein the anti-fungal agent's cytostatic activity is at least partially attributable to the presence of the modified histones. It will be clear to one skilled in the art that such screens are readily adaptable to high-throughput approaches, which furthermore can be fully or partially automated. In embodiments, the test agents are contacted with cultures of cells comprising histones modified as described herein, wherein the cell cultures comprise a liquid culture which is separated into a plurality of reaction chambers, such as in a high-throughput configuration. In an embodiment, the plurality of reaction chambers comprises up to or at least 1536 reaction chambers. Into each reaction chamber one or more test agents may be added, and a change in the cells in the cell culture due to the presence of the test agent can be observed, thereby identifying the test agent as a candidate for use in eliciting a similar change in non-modified cells that are the source of the modified histones in the cell culture. In an embodiment, a test agent is tested for killing and/or inhibiting the growth of, for example, yeast cells comprising histones from cancer cells. The following Examples are intended to illustrate but not limit the disclosure. Example 1 Saccharomyces cerevisiaecan Subsist on Fully Human Core Histones S. cerevisiaecells possess a relatively simple repertoire of histones. The core nucleosome, the four histones H3, H4, H2A, and H2B, comprise duplicate copies in the genome—each with divergent promoters and terminators—for a total of eight histone copies (Eriksson et al., 2012) (FIG.8). Additionally, there are three histone variants, H2A.Z, CENPA, and H1 (HTZ1, CSE4, and HHO1, respectively), which were not altered here. To humanize the core nucleosome of yeast, we constructed three strains in which the target histones of interest (e.g., all 8 core histones) were deleted from the genome. We then used a plasmid shuffle approach (i.e., yeast vs. human histones on plasmids) for quickly eliminating the native yeast histones in favor of their human counterparts, by 5-FOA counter-selection (Boeke et al., 1987) (FIGS.1B and8; see Methods). The dual plasmids, containing either human or yeast histone genes, are expressed from different sets of endogenous histone promoters and terminators to eliminate recombination. Any designed humanized histone strain will carry only half the target histone copies (e.g., 4 instead of 8 histones). First, we determined the relative “humanization frequencies” (i.e., humanized colonies per cell plated) for individual or pairs of human histones. Human histone H4 (hH4) had the highest humanization frequency (fast growth, 20%), followed by hH2B (slow growth, 20%), hH2A (slow growth, 10-2), and finally hH3.1 and hH3.3 (very slow growth, 10-4 and 10-5 respectively) (FIG.1C). Combining hH4 with either hH3.1 or hH3.3 also produced very slow growth (frequency of 10-4 and 10-5, respectively), whereas combining hH2A with hH2B led to slow growth, but a modest humanization frequency of ˜10-3. For both human histone combinations, we confirmed the loss of yeast histones by PCRtag analysis (Mitchell et al., 2015), which uses PCR to discriminate between sequence differences of yeast and human histones, and a lack-of histone mutations by Sanger sequencing of recovered plasmids (FIG.8D, E). We then attempted to exchange all four histone genes simultaneously (FIG.1C). An “isogenic-WT” strain (yDT67) that replaces one native yeast histone plasmid with a plasmid containing the other set of native histones “shuffled” readily (FIGS.1C and8). Yet neither plasmid encoding fully human core nucleosomes containing either hH3.1 or hH3.3 produced colonies within nine days. However, upon plating at least 107 cells and waiting 20 days we did see colonies representing humanized yeast, but only for the hH3.1 plasmid (FIG.1D). These humanized yeast colonies were confirmed via PCRtag analysis and sequencing of extracted plasmids (FIG.1E), which showed no mutations in the human histones. These humanized colonies are unlikely to contain residual yeast histones, as “old”-histones turn-over by at least two-fold per cell division (Annunziato, 2005; Radman-Livaj a et al., 2011). Since each haploid cell contains about 67,000 nucleosomes (Brogaard et al., 2012), and a small yeast colony contains at least 107 cells, those underwent at least 23 cell divisions. Therefore, the cells contain on average 0.01 original yeast nucleosomes, assuming an infinite nucleosome/histone half-life. To date, we have only identified 8 such confirmed direct humanization events (“yHs”-series;FIG.8F). Increased (high-copy plasmid) or decreased (genomic integration) human nucleosome gene copy number did not enhance humanization frequency (Table 3). The “yHs” cells grow on both synthetic complete (SC) and yeast complete (YPD) medium at 30° C. and 25° C., but not at higher temperatures (e.g., 37° C.), can mate, and grow to various degrees on media that enhance defects in DNA replication, DNA repair, and vacuole formation (FIG.9A, B). Finally, each of the humanized strains possessed differing rates of substantially slower than normal growth, and frequently produced larger and faster growing colonies over time (FIG.1DandFIG.8H). These observations are consistent with the accumulation of suppressors. A second factor reducing the humanization frequency is that a substantial proportion of the humanized cells in a population are unable to form a living colony. Example 2 Bypass of Cell-Division Genes Promotes Growth with Human Nucleosomes We performed evolution experiments on seven “yHs” lineages to determine how and to what extent yeast cells adapt to “live with” core human nucleosomes. The seven lineages were selected by serially diluting and sub-culturing liquid stationary phase cultures for 5 cycles (FIG.2A). The evolved pools and isolates outperformed the pre-evolved strains on solid media, and doubled 33% more rapidly in liquid culture (FIGS.2B and9C). We then performed WGS on 32 of these humanized yeasts. Pulsed-field gel electrophoresis of whole chromosomes from each yHs-lineage showed normal chromosome size (FIG.9D). However, WGS revealed recurrent aneuploidy of specific chromosomes (FIGS.2D,9E, and Table 2). The human-histone plasmid copy number was no more than 2-fold higher than that in the parental strain yDT51. The majority of aneuploidies may be a detrimental consequence of human nucleosomes—as aneuploidies typically reduce fitness (Sheltzer et al., 2011)—but our frequently recurring aneuploidies (FIG.2D) were consistent with other studies (Pavelka et al., 2010), which consider them possible reservoirs for positive selection. Chromosome number was often unstable during lineage evolution, as fractional differences (e.g., 1.5-fold chr1) were not due to insertions/deletions or diploidy, indicating potentially variable levels of aneuploidy at the population level. Only the evolved isolates yHs4C5i1 and the yHs7C5 lineage had a normal chromosomal sequence coverage, the latter perhaps due to a mutation in the gene DAD1, which controls microtubule force at the centromere (Sanchez-Perez et al., 2005). By contrast, yHs5 and its progeny had higher levels of aneuploidy, perhaps due to a subtle mutation in the gene SCC4, a cohesin loader (Lopez-Serra et al., 2014). All humanized strains were either missing segments of mitochondrial DNA (mtDNA) (ρ−) or showed complete loss of mtDNA (ρ0), except for the lineages from yHs5 (ρ+). We investigated whether mtDNA loss alone might explain the slow growth rates (Veatch et al., 2009), but found that isogenic-WT ρ0 cells grow better than all humanized lines, and moreover the ρ+yHs5C5i1 isolate was not the fastest growing isolate (Table 2). We identified 36 mutations in or near genes among the 8 isolates and their derivatives (FIG.1C, Tables 1 and 2), and 22 unique mutations appeared likely to affect gene function based on alterations to protein sequences. We constructed an interaction network from these 22 mutations using the String algorithm (Szklarczyk et al., 2015) (FIG.2E). The enrichment of GO terms in this network was non-random, as the genes clustered in 4 processes: chromosome segregation, cytoskeleton, cell-cycle progression, and genes affecting RNA metabolism. These first 3 processes collectively affect mitotic cell-division (Janke et al., 2001; Lew and Reed, 1995). Therefore, mutations in genes that affect cell-division may suppress defects arising from human histones, possibly by circumventing cellular checkpoints triggered by aberrant chromatin properties. These results illustrate how much easier it is to evolve the surrounding the gene network to accommodate new functions rather than the gene itself. Example 3 Specific Residues in Termini of Human Histones H3 and H2A Limit Yeast Growth We were surprised to not identify any mutations within the human histone genes themselves. Converting the C-terminal residues of human histone H3 back to the yeast sequence enhances complementation (McBurney et al., 2016), and some species-specific residues cause lethality when mutated to alanine (Dai et al., 2008; Nakanishi et al., 2008). We systematically swapped residues from human to yeast across histones H3, H4, and H2A, in order to identify species-specific regions (FIGS.10and11), but did not perform such studies on H2B as it complemented relatively well. Swapping-back three residues in the C-terminus of hH3 enhances the humanization frequency (FIG.10A), consistent with a recent study (McBurney et al., 2016), whereas swapping-back the lethal residues provided no benefit (Dai et al., 2008). Although hH4 already worked well, we identified two residues in its C-terminus that enhanced humanization (FIG.10B). Only two swapped-back residues in hH3 (hH3KK; human→yeast P121K and Q125K) were required for complementation when combined with completely human H4 (FIG.10C), although there appear to be differences between hH3.1 versus hH3.3 in this regard. A possible explanation for the two hH3 swap-back residues may be that in yeast H3, the two lysine swap-back residues are ubiquitylated by Rtt101Mms1, and mutations in H3 of K121R/K125R reduced H3/H4 dimer release from Asf1, restricting transfer to other histone chaperones (Han et al., 2013). Swapping-back three broad regions in hH2A enhanced complementation in combination with fully human hH2B, the N-terminus, the C-terminus, and a region from residues 19 to 42 (FIG.11). Further analyses narrowed the essential residues to three residues each in the N-terminus and C-terminus. Combining all six of these residues significantly enhanced the humanization frequency and growth rate of the yeast (FIG.11D). Intriguingly, the mammalian lineage-specific N-terminal arginine residues, when inserted into yeast H2A, have been shown to increase chromosome compaction (Macadangdang et al., 2014). The C-terminal portion, which is exposed on the nucleosome face (White et al., 2001), may interact with histone chaperones (e.g., NAP1) analogous to the H3/H4 interaction with Asf1. We combined the 3 terminal-regions (hH3.1KK, hH2AN, and hH2AC) into human nucleosomes as various “Swapback strains” (FIG.3). As expected, combining all 3 swapped-back regions enhanced humanization (8-residue swayback strain yDT98) to 10-2 in only 3 days (FIG.3B). However, the swapback strain with only the two C-terminal regions (hH3.1KK and hH2AC; 5-residue swap strain yDT97) grew as fast as the 8-residue swapback version (yDT98), and both of these strains grew nearly as fast as our isogenic-WT strain (yDT67) in 3 days. The 5-residue swapback strain (yDT97) was used for further studies. Example 4 Human nucleosomes delay adaptation to new transcriptional programs in yeast Intriguingly, we often observed that the humanized cells had difficulty adapting to new environments (e.g., colony to liquid culture), which suggested slowed chromatin remodeling to new transcriptional programs. Consistent with this hypothesis, using a GAL1-promoter driven eGFP as a proxy for switching to the galactose utilization transcriptome using the RSC complex (Floer et al., 2010) we showed that cells with human nucleosomes had a pronounced delay in transcriptional response to galactose as the sole carbon source, as well as decreased maximal expression on induction (FIG.4A). We then assessed how readily the cells adjust to new phases of the cell-cycle, a process that also requires extensive chromatin remodeling. Using both bud-counting and flow cytometry of log-phase cells, we observed reduced cell-division, as only 40-60% of humanized cells reach the G2/M phase compared to ˜90% in isogenic-WT (FIG.12B). More importantly, the lag-phase cultures of the yDT97 “swap” strain display a prolonged S phase, indicative of a delay in adjusting to log-phase growth (FIGS.4B and12D). This could result from an inability to accumulate new histones onto nascent DNA or an inability to remodel and remove chromatin-bound factors (Ma et al., 2015). The humanized cells were also larger in size on average and produced a greater range in cell sizes (FIGS.4C and12), which could indicate an inability to regulate cell-size control due to less permissive chromatin. By micro-manipulating single cells onto YPD plates, we found no growth difference between large and small cells (FIG.12E). However, unbudded cells (G1) were less likely to continue to grow than budding cells, although they all mostly remained intact after several days of monitoring. Surprisingly, a high fraction of single cells grew for a number of cell-divisions before arresting as a population (i.e., arrested before reaching the size of a visible colony). Together, these results are consistent with the hypothesis that human nucleosomes delay adaptation to other phases of the cell-cycle. Nucleosome Organization is Specified by the Chromatin Remodeling Network To evaluate the organization of human nucleosomes on the yeast genome we performed MNase digestion titrations and MNase-seq on evolved and swapback strains using ‘high’ and ‘low’ enzyme concentrations (FIGS.5and13), to reveal possible differences in nucleosome accessibility (Kubik et al., 2015). Unexpectedly, the nucleosome repeat length (NRL) of yeast chromatin built using human nucleosomes was identical to the NRL in isogenic-WT yeast, and is substantially shorter than that for human HeLa cells (FIG.5A). The di-nucleosome length (˜300 bp) from low concentration MNase-seq confirms a short mean nucleosome repeat length (FIG.13C). These data indicate that the NRL in humans is not an intrinsic property of human core nucleosomes, but is likely specified by nucleosome remodelers, by the genomic sequence itself (Segal and Widom, 2009) or by some combination of these factors. To our surprise, the numbers of nucleosomes with altered positioning or fuzziness (movement) was no different than that of isogenic-WT “noise” (Chen et al., 2013). However, there are substantial occupancy differences, which are distinct even amongst the humanized lines (FIG.5B). Nevertheless, this suggests that nucleosome positioning is determined less so by the type of nucleosome, and much more so by the underlying DNA sequences and the network of chromatin-remodelers for a given species. As suggested by the chromosome segregation suppressor mutations identified earlier (FIG.2E), we find that human nucleosomes lead to depletion of centromeric nucleosomes as well as relative to the surrounding nucleosomes, perhaps due to conflict with the yeast centromeric H3 variant CSE4 (FIGS.5C and13D). Relative to the neighboring nucleosomes, depletion was greatest for centromeres on aneuploid chromosomes observed earlier by WGS (FIGS.2D and5C). Strain yHs5C5i1, which had the highest levels of aneuploidy, had greater depletion at these nucleosomes, whereas strain yHs7C5i1, which has normal chromosome numbers and carries a relatively subtle missense mutation (E50D) in the essential gene DAD1, has slightly better positioning at these nucleosomes (FIG.5C). Finally, all 275 tRNA genes had depleted sequence coverage in their gene-bodies compared to WT (FIG.5D). Unlike RNAP2 genes, tRNAs possess an ‘internal control region’, thus, the depleted regions could represent a loss of RNAP3 and accessory factors (Acker et al., 2013), or nucleosome depletion coupled to RNAP3 transcription elongation. In fact, substantially elevated tRNA levels were observed in RNA from yHs cells (FIG.14A), perhaps suggesting human nucleosomes are less stably bound to tRNA sequences. However, as yeast tRNAs are already highly expressed and mostly devoid of nucleosomes, this could instead indicate that tRNA levels are normal, and that it is mRNAs that are highly repressed by human nucleosomes, thus altering the tRNA/mRNA ratio. Example 5 Human Nucleosomes Produce Chromatin More Generally Repressive for RNAP2 As predicted, total RNA content—predominantly mRNA and rRNA—is reduced by 6-8 fold in all the pre-evolved humanized yeast, and only slightly increased in the evolved and swapback strains (FIG.6A). The mRNA to rRNA ratios remain similar to our isogenic-WT strain (FIG.14A). However, tRNA sized molecule(s) are elevated relative to total RNA, and this may alter the balance of RNA types in the cell. However, we found the reduced total RNA is not explained by substantially altered cell numbers per A600 or reduced cell viability as determined by sytox green or trypan blue staining of dead cells (Kwolek-Mirek and Zadrag-Tecza, 2014) (FIG.14B, C). Humanized whole-cell extracts had similar bulk protein yields to isogenic-WT, but the SDS-page gel stained with Coomassie shows numerous proteins with reduced levels, consistent with reduced RNA (FIG.14D), whereas other presumably highly stable proteins are relatively unaffected. Immunoblots using antibodies more specific for human H3 and H4 show greater signal for humanized strains (FIG.6B). Finally, both H3K4 trimethylation and H3K36 trimethylation signals were similar to the isogenic-WT strain, as these modifications are in regions conserved between yeast and humans. This suggests that low mRNA levels are not due to changes in these histone modifications. The reduced RNA content and slowed growth might reflect differences in nucleosome dynamics (Chen et al., 2013), and could indicate a fundamental property of human histones or their relative inability to interact with yeast chromatin remodelers. To understand this effect, we mapped the MNase-seq reads across the transcription start sites (TSS) of the top 1500 genes by expression, and the bottom 1500 genes by expression. Genome-wide, the MNase-seq reveals a less “open” nucleosome depleted region (NDR) upstream of the TSS for humanized yeast than that found in isogenic-WT yeast (FIGS.6C and14E). The amplitude (difference between the NDR and gene bodies) is smaller for fully humanized yeast compared to isogenic-WT, suggesting reduced RNAP2 transcription that is consistent with the decreased total mRNA/rRNA content (FIG.6A). The amplitude of highly expressed genes in humanized yeast looks more similar to the amplitude of the lowest expression genes in isogenic-WT. Even amongst themselves, the humanized strains show distinct nucleosome profiles, suggesting chromatin heterogeneity at the population-level. Furthermore, nucleosome fragment lengths at high concentration MNase of isogenic-WT yeast show a greater fraction of sub-nucleosomal particles (90-125 bp) compared to humanized yeast (˜147 bp), suggesting that human chromatin is less accessible to MNase (FIG.13C). The above results combined with the poor environmental adaptability and cell cycle delays suggest that human nucleosomes are more generally repressive to RNAP2 transcription than yeast nucleosomes, possibly because they have a higher intrinsic affinity for DNA (the model we favor), thus making them more static, or are less easily removed by the yeast chromatin-remodelers, which did not coevolve with these histone sequences. Such a finding is consistent with the relatively more unstable nature of yeast chromatin in vitro compared to human chromatin (Leung et al., 2016), as well as their biology—yeast genes are predominantly expressed (Rando and Winston, 2012)—while humans repress the majority of the genome in virtually all cell types (Djebali et al., 2012; Talbert and Henikoff, 2017; Thurman et al., 2012) (Buschbeck and Hake, 2017). Thus, specialized histone variants with higher DNA affinity and stronger gene repression, might enable multicellular organisms to generate a larger variety of transcriptional landscapes. Example 6 Suppressor Mutations and Human Chromatin “Memory” Enhance Humanization Frequency The numerous suppressor mutations identified earlier (FIG.2) may counteract the various defects observed in yeast with human nucleosomes. If the suppressors make human histones more tolerable, they would be predicted to enhance the “humanization frequency”. To determine this, we re-introduced the native yeast histone plasmid into 13 humanized suppressor strains (FIG.7A; black dots), and allowed mitotic loss of the human histone plasmid, thus reverting these cells to native yeast chromatin, whereupon their growth properties improve. These lines were used for dual-plasmid histone shuffling as before, by re-introducing the human histone plasmid. The humanization frequencies, 10-100 fold greater than for non-suppressor ρ0 or ρ+ strains, confirms that the identified suppressors enhance tolerance of human nucleosomes. Furthermore, humanized colonies appeared as early as 12 days instead of the 20 originally required. These frequencies are high enough to support histone shuffling using any histone variant or from potentially any species. While the suppressor mutations improved the humanization frequency going from native yeast chromatin to fully human chromatin, we also contemplated how readily human chromatin resists “invasion” by native yeast histones. If the humanization frequency improves in this scenario relative to the above, this might suggest maintenance of human chromatin, and a preference for re-incorporation of nucleosomes of their own type. To test this hypothesis, we re-introduced the native yeast histones into the fully humanized suppressor strains, and allowed for single colonies containing both types of chromatin to grow for approximately 26 cell division generations (FIG.7A; red dots). This is a suitable time frame for native yeast chromatin to outcompete human chromatin, if not for “parental” nucleosome maintenance. Indeed, upon performing 5-FOA plasmid shuffling to remove the native yeast histones, the humanization frequencies reached 10-5-10-3, and cells appeared as early as 7 days. We interpret this result to suggest that pre-existing human chromatin might help maintain chromatin of its own kind, at least regionally—a type of chromatin “memory” and transgenerational inheritance—thus pre-disposing some small fraction of cells to resist native yeast chromatin even after many cellular divisions. These results are surprising, as with few exceptions yeast do not have protein machinery dedicated to maintenance of different histone variants, let alone for human histones. In our model, (FIG.7B) nucleosomes prefer their own type, thus seeding and maintaining similar chromatin domains. Therefore, different histone-types or nucleosome compositions are less likely to invade and outcompete this “parental” chromatin during the partitioning of chromatin during cellular division. This may occur in all cells or perhaps a smaller fraction of “older” cells that retain more of the earlier human chromatin. Finally, these results are consistent with our earlier observation demonstrating the relatively static nature of human core nucleosomes, thus making them more likely to maintain a certain epigenetic state. From the foregoing Examples, the following will be apparent; Because histones are some of the most conserved genes amongst eukaryotes, it was surprising that fully human nucleosomes so rarely led to bona fide humanized yeast. This speaks to the centrality of nucleosomes in regulating diverse cellular processes, including transcription and chromosome structure and movement. Cumulatively, our data suggest that human histones in yeast are deposited less efficiently, possibly due in part to sequence incompatibilities mapping to only 5 residues in the C-termini of H3 and H2A. When they do get deposited, the human histones lead to greater gene repression via less accessible NDRs, variations in chromatin, delayed environmental adaptation resulting from slowed chromatin remodeling, and depleted centromeres that possibly limit kinetochore assembly. The sum of these effect leads to partial cell-cycle arrest in G1 and a slower S-phase, which suppressor mutations in these same pathways alleviate. The human core nucleosome, consisting of H3.1, H4, H2A.1 and H2B.1, may bind DNA more tightly, as it is predominantly deposited during DNA replication, (Campos et al., 2015) and then can remain in place for years, if not for decades, in a terminally differentiated state (Toyama et al., 2013). Earlier studies on in vitro reconstitution of yeast and mammalian nucleosomes suggested that mammalian nucleosomes bind DNA more readily, and that yeast nucleosomes are comparatively unstable (Lee et al., 1982). Given that yeast genes are generally euchromatic, and human genes are heterochromatic, this might indicate an evolutionary basis for histone sequence divergence. Yeast, which must readily adapt to new environments, evolved highly dynamic histones, that retain bifunctional characteristics of histone variants found in humans (Rando and Winston, 2012). For instance, yeast H3 acts as both as an H3.1 (replication-dependent) and H3.3 (replication-independent) variant, while yeast H2A acts as both an H2A.1 and H2A.X (DNA-damage) variant (Eriksson et al., 2012). In contrast, human cells must retain transcriptional states corresponding to cellular type. Thus, specialized histone variants with higher DNA affinity and stronger gene repression, enables multicellular organisms to generate more diverse transcriptional landscapes (Buschbeck and Hake, 2017). Indeed, these yeast with human nucleosomes had great difficulty adapting to new environmental conditions, perhaps due to the fundamentally more static biophysical properties of human core nucleosomes (Leung et al., 2016). Furthermore, our data suggests that hi stone sequences do not contribute to the nucleosome repeat length (NRL), as the NRL of yeast with human histones remained yeast-like. In higher eukaryotes, longer linker length is partially attributed to linker histone H1 (Fan et al., 2005; Woodcock et al., 2006), but in yeast, it has been shown that expressing the human H1.2 linker had no effect on the NRL (Panday and Grove, 2016). In humans, the NRL ranges from ˜178-205 bp, depending on histone modification state (Valouev et al., 2011), with activation marks having the shortest NRL. Thus, the underlying DNA sequence (Segal and Widom, 2009) and the proteins that interact with histones, such as Isw1a (Krietenstein et al., 2016), are more likely to specify this property. Converting only five residues, 2 in H3, and 3 in H2A promoted relatively robust utilization of human nucleosomes as “Swapback” strains. In the case of human histone H3, the incompatibility with yeast may be attributed to an absence of two lysines required for ubiquitilation in yeast (Han et al., 2013). Human H2A may also poorly interact with yeast histone-interacting genes. However, this finding is still somewhat surprising as numerous other residues differ from yeast to humans that presumably should have larger roles—including many modified residues. As just one example, histone H3 position 42 is a lysine in yeast, but an arginine in humans (Hyland et al., 2011). Numerous other positions differ (FIG.1A), thus, the relative inability to interact and modify histones at these different sites poses a serious question about the cumulative role of histone modifications during cell growth. Results presented herein suggests a type of chromatin partitioning “memory”, as yeast with pre-existing human chromatin more readily resisted native yeast histones (FIG.7). Histones are displaced from DNA during transcription, replication, and repair, and then reassembled onto DNA strands (Campos et al., 2014). How cells determine which histone sub-type and modification state must be deposited on the parent and daughter DNA strands in the replication fork remains a continuing question (Lai and Pugh, 2017). Based on our results, the restoration of nucleosomes to the parental strand and inheritance to the daughter strand may occur as a type of “semi-conservative replication” of chromatin, whereby both parent and daughter strands retain a portion of the ancestral nucleosome (human), and then may simply attract similarly composed or modified new nucleosomes. This may be a simple way to retain an epigenetic state. Our results argue against a “conservative” model, wherein daughter strands acquire only fresh nucleosomes. This model predicts that humanized yeast transformed with yeast histones, would react similarly to native yeast transformed with human histones (FIG.7B). However, the results leave open the possibility of a “dispersive” model, which is a mixture of the two models. Nevertheless, humanized yeast may permit more systematic study of this process, coupled with future advances in single cell chromatin-profiling methods. More generally, humanizing the chromatin of budding yeast provides new avenues to study fundamental properties of nucleosomes. We have explored some of these longstanding questions about histone variants: how they alter the dynamics of the genome at the structural and transcriptional level (Talbert and Henikoff, 2017); how they associate into different compositions of nucleosomes (Bernstein and Hake, 2006); and how they are partitioned and repositioned from cell-to-cell across generations (Budhavarapu et al., 2013; Campos et al., 2014). These questions remain fundamental, as many human cells are reprogrammed and differentiate using histone variants during development and disease (Santenard and Torres-Padilla, 2009; Wen et al., 2014). The number of histone variants found in humans is large, and includes many variants that accumulate during aging and disease. Thus, introducing foreign histones from a distant species into the simple yeast genome as described herein will help address these many questions. Example 7 The following materials and methods were used to produce the results described above, and in the figures and tables of this disclosure. Strains, plasmids, and media. All yeast strains used in this disclosure were haploid MATα, except as indicated in Table 5. Yeast to human complementation studies of histones H3 or H4 alone or in combination, were performed in strain yDT17. Strain yDT17 was generated by replacing the HHT1-HHF1 locus with NatMX4 by one-step PCR recombination, reintroducing HHT1-HHF1 on a URA3 containing pRS416 plasmid, and then replacing the HHT2-HHF2 locus with HygMX4. Experiments involving H2A or H2B alone or in combination used strain yDT30. Strain yDT30 was generated by replacing the HTA2-HTB2 locus with HygMX4, transformation with pRS416-HTA2HTB2, and then deleting the HTA1-HTB1 locus as above with KanMX4. Analysis of all four histones was performed in strain yDT51. yDT51 was generated similarly to the above, but contains plasmid yDT83 (pRS416-HTA2-HTAB2-HHT1-HHF1). The antibiotic markers (KanMX4, NatMX4, HygMX4) and the His3MX4 cassette used in replacing the four loci were reclaimed by deleting these open reading frames using the CRISPR/Cas9 system (DiCarlo et al., 2013) at the positions indicated with red arrows inFIG.8A. Human histone genes were codon-optimized for yeast and synthesized by Epoch Biolabs. All cloning was performed by Gibson Assembly (Gibson et al., 2009). Swapback residue (human to yeast) histone variants were generated either by gene synthesis or site-directed mutagenesis. A complete list of available strains and plasmids are in the Supplemental Data. Dual-plasmid histone shuffle assay. Shuffle strains (yDT17, yDT30, or yDT51), which already contains a set of yeast histones on a URA3 plasmid, were transformed by a standard Lithium Acetate protocol with a TRP1 human histone plasmid, which uses the endogenous promoters/terminators from the other yeast histone set. Colonies were selected for 3 days at 30° C. on SC-Ura-Trp plates. Single colonies were picked and grown up overnight at 30° C. in 2 ml of SC-Trp. Spot assays (as indicated) were diluted 10-fold from overnight cultures (A600of ˜10) and spotted on both SC-Trp and SC-Trp+5FOA plates. Shuffle assays for fully human nucleosomes using strain yDT51, were done as above, except 400 μl of overnight culture were spread onto a 10-cm SC-Trp+5FOA plate (25 ml) and incubated at 30° C. for up to 20 days in a sealed Tupper-ware container. Plasmid isolation from yeast cells. Cells were harvested from 5 ml SC-Trp overnight culture and re-suspended in 600 μl of water and glass beads. Cells were vortexed for 10 minutes to disrupt cells. Plasmids were then isolated by alkaline lysis using the Zymo Zyppy miniprep kit and eluted in 20 μl of water. 5 μl was used to transformE. coli, and isolate pure plasmid. PCRtag analysis. Crude genomic DNA was generated using a SDS/Lithium Acetate method (Looke et al., 2011). Comparative PCRtag analysis was performed using 0.5 μl of crude gDNA in a 20 μl GoTaqGreen Hot Start Polymerase reaction (Promega) containing 400 mM of each primer (Table 7). Reactions were run as follows: 95° C./5 min, followed by 35 cycles of (95° C./30 s, 62° C./30 s, 72° C./30 s) followed by a 72° C./2 min extension. A 10 μl aliquot was run on a 1% agarose/TTE gel. Pulsed-field gel electrophoresis. Intact chromosomal DNA plugs were prepared as described elsewhere (Hage and Houseley, 2013). Chromosome identity was inferred from the known molecular karyotype of parental cells (yDT51) itself derived from S288C that was run on the same gel. Samples were run on a 1.0% agarose gel in 0.5×TBE for 24 h at 14° C. on a CHEF apparatus. The voltage was 6 V/cm, at an angle of 120° and 60-120 s switch time ramped over 24 h. Mating and sporulation tests. Mating tester lawns (his1 strains 17/17 MATα or 17/14 MATα) were replica plated to YPD plates. A large amount of humanized strains (HIS1) were then smeared onto the replica plate to form rectangles, and then incubated overnight at 30° C. Plates were then replica plated onto synthetic defined (SD) plates and incubated overnight at 30° C. The diploids were sporulated for 7 days as previously described (Dai et al., 2008). Microscopy. All yeast were grown to an A600of 0.5-0.9 in SC-Trp liquid media, and imaged under phase-contrast conditions at 100× magnification using a Nikon Eclipse Ti microscope. Cellular diameters were measure from 4 images each, comprising a total of 50 single cells. Violin plots and boxplots were generated using the R-package ggplot2. Cell counting and viability. Cells were manually counted using a hemacytometer with Trypan blue vital dye under a microscope. Cell viability was also measured by incubating cells in 1 μM Sytox Green solution in PBS, and counting number of fluorescent cells (dead) by flow cytometry on a BD Accuri C6 flow cytometer. Coulter counting was performed using a Millipore Scepter by diluting log phase cultures 1:100 in PBS, and then taking up cells according to manufacturer's recommendations. Micromanipulation of single cells was performed using a Singer MSM 400 onto YPD plates. Cell-cycle analysis using sytox green. Cell-cycle analysis by DNA content was adapted from (Rosebrock, 2017). Yeast were grown to log phase unless otherwise indicated in SC-Trp. Lag-phase for yDT67 took 45 min, whereas yDT97 took 2 h. Briefly, 107cells were fixed overnight in 70% EtOH overnight. Cells were incubated in 500 μl 2 mg/ml RNaseA solution for 2 h at 37° C. Then, 25 μl of 20 mg/ml Proteinase K solution was added, and cells incubated for 45 min at 37° C. Cells were washed and then stored in 1 ml 50 mM Tris pH 7.5. 50 μl of cells were re-suspended in 1 ml of 1 μM solution of Sytox Green (Thermofisher), and then 10,000 events were analyzed by flow cytometry on a BD Accuri C6 flow cytometer. Flow cytometry of GAL1-eGFP induction. Strains as indicated were transformed by standard lithium acetate with plasmid pAV115-GAL-GFP, and selected on SC-Leu+2% glucose plates at 30° C. Single colonies were grown overnight at 30° C. in SC-Leu+glucose (2%). Cells were washed once in PBS, and then sub-cultured into SC-Leu+galactose (2%)+raffinose (1%) media and incubated at 30° C. For the times indicated, 25 μl of cells were diluted into 0.2 ml PBS and 10,000 events were analyzed by flow cytometry on a BD Accuri C6 flow cytometer. “Re-humanization” of suppressor mutants starting with human or yeast chromatin. Humanized lineages were re-transformed with native yeast histone plasmid pDT83 (URA3) using standard Lithium Acetate transformation, and selected on SC-Ura-Trp plates for 4 days at 30° C. To determine the “human histone memory”, single colonies were grown overnight in 2 ml SC-Trp and directly used in dual-plasmid histone shuffle as described above. To determine the “rehumanization” rate of suppressor mutations, single colonies from the above re-transformed strains were grown in SC-Ura for multiple sub-cultures to allow mitotic loss of the TRP1 human histone plasmid pDT109. Cells were replica plated onto SC-Ura and SC-Trp to identify those containing only the native yeast histones. These strains were then used for another round of dual-histone plasmid shuffle as described above. Protein Analysis and Western Blotting. Whole-cell extracts were generated using a modified protocol from (Zhang et al., 2011). Briefly, 108log-phase yeast cells were re-suspended in 400 μl 0.15 M NaOH and 0.5 mM dithiothreitol (DTT), and incubated for 10 min on ice. Cells were pelleted at top speed for 10 min at 4° C., and re-suspended in 65 μl lysis buffer (20 mM HEPES pH 7.4, 0.1% Tween20, 2 mM MgCl2, 300 mM NaCl, 0.5 mM DTT, and 1 mM Roche Complete protease inhibitor) and an equal volume of 0.5 mm glass beads. Mixture was vortexed at top speed for 10 min in the cold room. Subsequently, 25 μl of NuPAGE (4×) LDS Sample buffer and 10 μl beta-mercaptoethanol was added, and the mixture was heated at >95° C. for 10 min. The debris was pelleted and the supernatant was run on a 12% Bis-Tris SDS acrylamide gel and stained with Coomassie blue. Acid extracted histones were generated by first re-suspending 5×108log-phase yeast in spheroblasting buffer (1.2M Sorbitol, 100 mM potassium phosphate pH 7.5, 1 mM CaCl2), and 0.5 mM β-mercaptoethanol) containing Zymolase 40T (40 units/ml) and incubating for 20 min at 37° C. Spheroblasts were gently spun down at 3000 rpm for 3 min and then re-suspended in 1 ml of 0.5 M HCl/10% glycerol with glass beads on ice for 30 min. Cells were vortexed at top speed for 1 min every 5 min and kept on ice. Mixture was spun at 16,000×g for 10 min and the supernatant was added to 8 volumes of acetone and left at −20° C. overnight. The following day, mixture was pelleted for 5 min at 16,000×g, the solution poured off and the pellet was air-dryed. Pellet was resuspended in 130 μl water, and then 50 μl NuPAGE (4×) LDS Sample buffer and 20 μl beta-mercaptoethanol was added, and the mixture was heated at >95° C. for 10 min. Supernatant was run on a 12% Bis-Tris SDS acrylamide gel and stained with Coomassie blue, or directly used for Western blotting. Protein samples run on 12% Bis-Tris SDS acrylamide gel were transferred to membrane (Millipore, Immobilon-FL) using the BioRad Trans-Blot Turbo system according to manufacturer's recommendations. Membranes were blocked for 1.5 h at room temperature in 1:1 Tris-buffered saline (TB S)/Odyssey blocking buffer (LiCor). Blocking buffer was removed and membrane re-suspended in primary buffer overnight at 4° C. containing 1:1 TBS+0.05% Tween-20 (TBST)/Odyssey and the following antibodies used at 1:2,000 dilution: human H3 (abcam ab24834), H3K4me3 (abcam ab1012), H3K36me3 (abcam ab9050), human H4 (abcam ab10158). The following day, membrane was washed 5 times for 5 min each in TBST/Odyssey, re-suspended in secondary antibody buffer TBST/Odyssey/0.01% SDS with 1:20,000 dilution of both IRDye 800 goat anti-mouse and IRDye 680 goat anti-rabbit (LiCor) for 1.5 h at room temperature. Secondary was washed 5 times for 5 min each in TBST/Odyssey and then imaged using dual channels on a LiCor Odyssey CLx machine. Growth assay on various types of solid media. Cultures were normalized to an A600of 10 and serially diluted in 10-fold increments in water and plated onto each type of medium. The following drugs and conditions were mixed into YPD+2% dextrose+2% agar: benomyl (15 μg/ml; microtubule inhibitor), camptothecin (0.5 μg/ml; topoisomerase inhibitor), hydroxyurea (0.2 M; defective DNA replication), NaOH (pH 9.0; vacuole formation defects), HCl (pH 4.0; vacuole formation defects), and methyl methanosulfate (MMS 0.05%; defective DNA repair). Galactose plates were prepared in Synthetic Complete media+1% raffinose and 2% galactose. Whole genome sequencing and data analysis. Genomic DNA was isolated using Norgen Biotek's Fungal/Yeast Genomic DNA isolation kit, which included a spheroblasting step and bead-beating step. At least 1 μg of genomic DNA was used for Illumina library preparation using the Kapa Truseq library prep, and we routinely multiplexed 30 yeast genomes on a single HiSeq 4000 or 2500 lane. Paired-end FASTQ files were aligned with the following pipeline. First, adapters, reads shorter than 50 bp, and poor quality reads near ends, were removed using Trimmomatic (Bolger et al., 2014). Data quality was assessed using FastQC. Processed reads were aligned to a custom genome reference (yDT51H.fa) using Burrows Wheeler aligner (BWA) mem algorithm (Li and Durbin, 2010), and Sam files were converted to sorted Bam files using Samtools (Li, 2011). Variants were called using the GATK “best practices” pipeline for Haplotype caller, custom scripts, and manually verified on the IGV viewer. Variants were identical using Samtools “mpileup”. Read counts for each chromosome were determined from WGS Bam files using Bedtools “genome coverage” (Quinlan, 2014). Chromosome copy number was then calculated by generating boxplots in R using ggplot2. Networks for suppressor mutants were generated by uploading genes into the String online server (Szklarczyk et al., 2015). GO-terms were identified using the Panther database (Mi et al., 2016). Of the 37 mutations identified (Tables 1 and 2), 6 synonymous mutations were considered “innocuous” based on their similar codon usage. MNase-digestions and MNase-sequencing. Experiments were adapted from (Kubik et al., 2015). Biological triplicate yeast colonies were each grown at 30° C. to an A600of ˜0.9 in 100 ml of SC-Trp media. Cultures were crosslinked with 1% formaldehyde for 15 min at 25° C., and then quenched with 125 mM glycine. Cultures were washed twice in water, and pellets were then stored at −80° C. To perform MNase digestions, cells were first spheroplasted by suspending pellets in 4 ml spheroplasting buffer (1.2M Sorbitol, 100 mM potassium phosphate pH 7.5, 1 mM CaCl2), and 0.5 mM β-mercaptoethanol) containing Zymolase 40T (40 units/ml) and incubated for 20 min at 37° C. Spheroblasts were gently washed twice with spheroblasting buffer, and then re-suspended in 1 ml digestion buffer (1M Sorbitol, 50 mM NaCl, 10 mM Tris-HCL (pH 7.4), 5 mM MgCl2, 1 mM CaCl2), 0.5 mM spermidine, 0.075% NP-40, and 1 mM β-mercaptoethanol). Samples were split into 500 μl aliquots equivalent to 50 ml culture each. To each sample, micrococcal nuclease (Sigma: N5386) was added to a final concentration for high digestion (2 units/ml) or low digestion (0.2 units/ml) or as specified inFIG.13. Digestions proceeded at 37° C. for 45 minutes. Reactions were quenched with 16.6 μl of 0.5 M EDTA. Crosslinks were reversed in 0.5% SDS and 0.5 mg/ml proteinase K, by incubating at 37° C. for 1 h, followed by 65° C. for 2 h. Nucleic acid was extracted with phenol/chloroform twice, followed by chloroform. Nucleic acid was precipitated by adding 50 μl Sodium Acetate (3M, pH 5.2), an equal volume of isopropanol, and spinning for 20 min at 16,0000×g. Pellets were washed once with 70% EtOH, and then resuspended in 50 μl TE buffer containing 6 kUnitz of RNase A, and incubated for 30 min at 37° C. DNA was then purified using a Zymo DNA clean and concentrator, and eluted in 20 μl. MNase digested fragment DNA was measured by Qubit, and assessed on a 1.5% agarose TTE gel. At least 200 ng of DNA (PCR-free or minimal PCR of 2-3 cycles) for each replicate was used to generate a library for paired-end sequencing on an Illumina Hiseq 4000. Nucleosome positioning analysis. MNase-seq FASTQ reads were processed using Trimmomatic (Bolger et al., 2014), FastQC, and then aligned to the sacCer3 reference genome using BWA-mem (Li and Durbin, 2010), and then converted to a sorted Bam file using samtools (Li, 2011). Custom bed files corresponding to the top and bottom 1500 genes, centromere regions, and tRNA regions were used to align MNase reads using Ngs.plot (Shen et al., 2014) to regions as specified. Fragment lengths were obtained from Sam files and plotted using ggplot2. Nucleosome dynamics were analyzed using DANPOS2 (Chen et al., 2013). Custom scripts were used to process the data to reduce erroneously called and altered nucleosomes as based on comparing MNase-seq data from WT experiment 1 against WT experiment 2 (“noise”). Nucleosome shifts passed the threshold when both nucleosome comparisons had aligned reads >300 and when shifts were greater than 70 bp. Nucleosome occupancies required that at least one nucleosome comparison have an aligned read count >300, and the False Discovery Rate (FDR) was lower than 0.05 with a p<10−85. Fuzzy nucleosomes required that both nucleosome comparisons have read counts >300 and an FDR of <0.05. TABLE 1Whole genome sequencing mutations, Related to FIG. 2.Chr-Amino acidPositionGeneMutation; posΔMut EffectFunction1-14,018TDA8A −> C; −275n/anoncodingPutative protein of unknown function3-142,928MAK32G −> T; 805A269SmissenseStability of L-A dsRNA-containing particles3-130,008PGK1CTT −> C; −60n/anoncoding3-phosphoglycerate kinase4-54,617WHI4G −> C; 1729H577DmissenseRegulates Start and commitment to cell division4-177,251CLB3C −> G; 480Y160stopnonsenseActivates Cdc28 to promote the G2/M transition4-478,608DAD1G −>C; 150E50DmissenseAids in chromosome segregation; DASH complex4-527,742ENA1T −>C; 2955S985SsynonymousP-type ATPase sodium pump-527,796T −>C; 2901S967Ssynonymous-527,850T −>C; 2847T949Tsynonymous5-463,552SCC4C −> A; 193D65YmissenseCohesin loader; chromosome segregation5-478,709BEM2T −> G; 3042P1014P*synonymousCytoskeleton organization; cellular morphogenesis5-528,878BRR2G −> T; 6046V2016FmissenseRNA helicase; spliceosome6-51,324YFL040WT −> G; −26n/anoncodingPutative sugar transporter6-235,653SAP155T −> G; 1424V475GmissenseEssential for Sit4 (G1/S transition); DNA stress7-336,635SPC105G −> C; 1749R583SmissenseBridges centromeric heterochromatin and kinetochore7-669,081DBF2A −> T; 8895297TmissenseActivated by Cdc15 during mitotic exit; regulates Clb28-461,050STB5A −> T; 1752A584AsynonymousTranscription factor; regulates oxidative stress response9-79,776TID3A −> C; 1703E568AmissenseKinetochore component; chromosome segregation9-254,845YIL054WG −> C; 301D101HmissenseProtein of unknown function10-172,891URA2C −> G; −524n/anoncodingDe novo biosynthesis of pyrimidines11-257,541MTC2T −> C; 768Y256YsynonymousProtein of unknown function; sick with cdc13-111-511,612SPC34T −> A; 188L63QmissenseAids in chromosome segregation; DASH complex11-660,938-GEX2Deletionn/adeletionProton:glutathione antiporter; expressed at low level-662,78511-666,600TY5Deletionn/adeletionTy5 retrotransposon-666,83811-662,164GEX2A −> C; 721K241QmissenseProton:glutathione antiporter; expressed at low level12-5,742YLL066W-BT −> C; 137F465missenseOverexpression causes cell-cycle delay or arrest12-573,283CPR6A −> G; -59n/anoncodingBinds Hsp82p and contributes to chaperone activity12-802,533VRP1C −> T; 176G59DmissenseCytoskeleton organization and cytokinesis12-1,029,289FPR4G −> C; 858L286LsynonymousIsomerization of proline residues in histones H3 and H413-477,824ARS1317A −> G; 240n/anoncodingPutative replication origin13-812,924ZDS1G −> T; 1036A346SmissenseMitotic exit; maintains Cdc55; regulates Cdc14, Swe114-42,487PHA2C −> G; 592Q594EmissensePhenylalanine biosynthesis pathway14-67,507TRF5C −> T; 988R329stopnonsenseNuclear RNA degradation; TRAMP complex; poly(A) pol14-689,433SOL1GT −> G; 882FV294LstopnonsenseDNA replication stress; tRNA export15-143,923TRm10C −> T; +227n/anoncodingDNA replication stress; tRNA methyltransferase15-630,433NFI1C −> G; 397V133LmissenseSumoylates Cse4, Sir4, Yku70/80; regulates telomeraseMutation; pos refers to the nucleotide mutation and position relative to the sense strand ORF. Negative values (−X) indicate promoter mutations and positive values (+X) refer to terminator mutations. Synonymous mutations were considered innocuous if codon usage difference was <2-fold.*Synonymous mutation codon usage was different: BEM2 (CCT (0.31) to CCG (0.12), all other cases codon usage was similar. TABLE 2Whole genome sequencing mutation genotypes by lineage, Related to Figure 2.Presumed InnocuousmtDNADoublingIsolateAliasMutationsMutations(ρ+/−/0)Chromosome aneuploidy(h:min)parent: yHs11 original poolyHs1—tda8(−275)ρ−XII (2x)n/t1 maintenanceyHs1mn/tn/tn/tn/t16:021 cycle 5 poolyHs1C5—tda8(−275)trm10(+227)ρ−I, II, III, V, IX, XI, XVI (2X)10:331 cycle 5 i11C5i1spa105(R583S)vrp1(G59D)nfi1(V133L)tda8(−275)trm10(+227)ρ−XII (2x)10:18mtc2(Y256Y)1 cycle 5 i21C5i2spa105(R583S)vrp1(G59D)nfi1(V133L)tda8(−275)trm10(+227)ρ−XII (2x)n/tmtc2(Y256Y)1 plate 5 i31PC5i3brr2(V2016F)tid3(E568A)pha2(Q594E)tda8(−275)pgk1(−60)ρ−II, III, XII (2x)13:10sol1(FV294Lstop)1 plate 5 i51PC5i5brr2(V2016F)tid3(E568A)pha2(Q594E)tda8(−275)pgk1(−60)ρ−II, III, XII (2x)n/tsol1(FV294Lstop)parent: yHs22 original poolyHs2zds1(A346S)tda8(−275)ρ−I (2x); II, III, IX, XVI (1.5x)n/t2 maintenanceyHs2mn/tn/tn/tn/t13:482 cycle 5 poolyHs2C5zds1(A346S)tda8(−275)ρ−I, XVI (2x); II, III, V, IX, XI (1.7x)10:392 cycle 5 il2C5i1zds1(A346S)stb5(A584A)tda8(−275)ars1317(240)ρ0I (3x); III, XVI (2x); II, V, VIII, IX,n/tXI (1.5x)2 plate 6 il2PC6i1zds1(A346S)stb5(A584A)tda8(−275)ρ0I (2x)11:542 phe AEphe2AEzds1(A346S)whi4(H577D)ttda8(−275)fpr4(L286L)ρ0I (2x)8:562 phe AFphe2AFzds1(A346S)whi4(H577D)gex2(K241Q)tda8(−275)ρ0—12:132 phe AHphe2AHzds1(A346S)whi4(H577D)tda8(−275)ρ0I (2x); III (1.7x)9:162 phe BCphe2BCzds1(A346S)whi4(H577D)tda8(−275)ρ0I (2x); IX, XVI (1.5x)13:022 phe BDphe2BDzds1(A346S)whi4(H577D)tda8(−275)ρ0I (1.8x); XVI (1.5x)n/a2 phe 3Cphe23Czds1(A346S)whi4(H577D)dbf2(S297T)tda8(−275)yfl040w(−26)ρ0I (1.5x)10:18parent: yHs33 poolyHs3yll066w-b(F46S)tda8(−275)ρ−XII (2x); II (1.5x)n/t3 maintenanceyHs3mn/tn/tn/tn/t13:253 cycle SpoolyHs3C5yll066w-b(F46S)clb3(Y160stop)tda8(−275)ρ−I (2x)10:183 cycle 5 il3C5i1yll066w-b(F46S)clb3(Y160stop)tda8(−275)ρ−I, II, III, IX, X, XVI (1.5x)12:00sap155(V475G)parent: yHs44 original poolyHs4——ρ−XII (2x)n/t4 maintenanceyHs4mn/tn/tn/tn/t15:374 cycle 5 poolyHs4C5trf5(R329stop)—ρ−I, III, XVI (1.5x)9:444 cycle 5 il4C5i1bem2(P1014P)gex2-ty5(deletion)cpr6(−59)—ρ−9:24parent: yHs55 original poolyHs5scc4(D65Y)—ρ+I, II, III, V, IX, XI, XII, XVI (2x)n/t5 maintenanceyHs5mn/tn/tn/tn/t15:045 cycle SpoolyHs5C5scc4(D65Y)—ρ+I, II, III, V, IX, XI, XII, XVI (1.5x)10:195 cycle 5 il5C5i1scc4(D65Y)—ρ+I, II, III, V, IX, XI, XII, XVI (1.5x)9:44parent: yHs66 original poolyHs6——ρ0II, XII (2x)n/t6 maintenanceyHs6mn/tn/tn/tn/t17:546 cycle 5 pool6C5yil054W(D101H)ena1(T949T, S9676, S985S)ρ0I, III, XVI (2x)9:536 cycle 5 il6C5i1yil054W(D101H)ena1(T949T, S9676, S985S)ρ0I, II, III, XI, XVI (1.5x)10:23parent: yHs77 original poolyHs7—ura2(−524)ρ−XII (2x), II (1.5x)n/t7 maintenanceyHs7mn/tn/tn/tn/t17:187 cycle SpoolyHs7C5dad1(E50D)ura2(−524)ρ−—9:157 cycle 5 il7C5i1dad1(E50D)ura2(−524)ρ−—9:20parent: yHs88 maintenanceyHs8mmak32(A269S)ρ−II, III, XII (1.5x)12:07parent: yDT51yDT988-swap——ρ+III (2x)~4strainyDT975-swap——ρ+VI (0.5x)~4strainyDT67WTn/tn/tn/tn/t1:50yDT51parental——ρ+—n/tAll mutations are in 65% or greater of reads. Spaces showing a “—” indicate an absence of mutations or abnormalities, while “n/t” refers to not tested. TABLE 3Initial humanization trials.Human histonesPlates (107cellsHumanStrainlocationper platecoloniesDateNotesyDT51Low-copy plasmid33Apr. 02, 201615 cm 20 ml platesyDT51Low-copy plasmid44May 31, 201615 cm 20 ml platesyDT51Low-copy plasmid201Jun. 10, 201615 cm 30 ml platesyDT51Genomic integration100Jun. 10, 201615 cm 30 ml platesat HO-locusyDT512-micron High-copy200Jun. 10, 201615 cm 30 ml platesplasmidyDT51Low-copy plasmid31Nov. 10, 201615 cm 20 ml plates TABLE 4Evolution cycles from Figure 2A (A600starts at 0.01).cycle 1cycle 2cycle 3cycle 4cycle 5A600daysA600daysA600daysA600daysA600daysyHs14.0155.1106.186.662.34yHs25.3116.264.247.255.44yHs36.9116.265.156.552.54yHs45.286.346.244.945.94yHs56.874.047.146.046.64yHs65.3106.344.036.436.34yHs76.0104.875.045.346.24 TABLE 5Strains used in this disclosureStrainnameOther nameMATGenotypeBY4741aleu2Δ0 met15Δ40 ura3Δ0 his3Δ1BY4742αleu2Δ0 lys2Δ0 ura3Δ0 his3ΔA1yAS03417/17αhis1yAS03317/14ahis1yDT17H3/H4ahis3Δ200 leu2Δ1 lys2Δ202 trp1Δ63 ura3-52 hht1-hhf1::NatMX4 hht2-hhf2::HygMX-4[pDMI9 (HHT1-HHF1/URA3/CEN-ARS/Amp)]yDT30H2A/H2Bαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2::HygMIX-4 hta1-htb1::KanMX [pJD78 (HTA2-HTB2/URA3/CEN-ARS/Amp)]yDT51parentalαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)]yDT67isogenic WTαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT105 (pRS414-HHT2-HHF2-HTA1-HTB1/TRP1/CEN-ARS/Amp)]yDT64yHs1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)ρ−yDT65yHs2αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(4346S)ρ−yDT66yHs3αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)yll066w-b(F46S)ρ−yDT71yHs4αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] ρ−yDT72yHs5αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] scc4(D65Y)ρ+yDT73yHs6αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] ρ0yDT74yHs7αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] ura2(−524)ρ−yDT79yHs8αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] mak32(A269S)ρ−yDT75yHs1C5αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)trm10(+227)ρ−yDT76yHs2C5αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(A346S) ρ−yDT77yHs3C5αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)yll066w-b(F46S)clb3(Y160stop)ρ−yDT81yHs4C5αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] trf5(R329stop)ρ−yDT82yHs5C5αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] scc4(D65Y)p+yDT83yHs6C5αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] ena1(T949T, S967S, S985S)yil054W(D101H)ρ0yDT84yHs7C5αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] dad1(E50D)ura2(−524)ρ−yDT86yHs1C5i2αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)trm10(+227)mtc2(Y256Y)spc105(R583S)vrp1(G59D)nfi1(V133L)ρ−yDT87yHs2C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(A346S)ars1317(240)stb5(A584A)ρ0yDT88yHs3C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)yll066w-b(F46S)clb3(Y160stop)sap155(V475G)ρ−yDT94yHs4C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] bem2(P1014P)gex2-ty5(deletion)cpr6(−59)ρ−yDT92yHs5C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] scc4(D65Y)ρ+yDT93yHs6C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] ena1(T949T, S967S, S985S)yil054W(D101H)ρ0yDT95yHs7C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] dad1(D50D)ura2(−524)ρ−yDT852PC6i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(A346S)stb5(A584A)ρ−yDT105phe2AEαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(A346S)wh4(H577D)fpr4(L286L)ρ0yDT106phe2AFαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)gex2(K24IQ)ρ0yDT107pheAHαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)ρ0yDT109pheBCαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.1-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)ρ0yDT108phe23Cαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-hhf1Δ0 hht2-hhf2Δ0 [pDT109 (pRS414-hH3.+-hH4-hH2A-hH2B/TRP1/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)dbf2(S297T)yfl040w(−26)ρ0yDT975-residueαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-swaphhf1Δ0 hht2-hhf2Δ0 pDT128 (pRS414-hH3.1KK-hH4-hH2AC-hH2B/TRP1/CEN-ARS/Amp)]yDT988-residueαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-swaphhf1Δ0 hht2-hhf2Δ0 pDT130 (pRS414-hH3.1KK-hH4-hH2ANC-hH2B/TRP1/CEN-ARS/Amp)]yDT1241B 105i2αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] tda8(−275)trm10(+227)mtc2(Y256Y)spc105(R583S)vrp1(G59D)nfi1(V133L)ρ−yDT1252A pl C5i3αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] tda8(−275)pgk1(−60)brr2(V2016F)tid3(E568A)pha2(Q594E)soL1(FV294Lstop)ρ−yDT1284A 2pC6i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] tda8(−275)zds1(A346S)stb5(A584A)ρ−yDT1305A phe2AEαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [[pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)fpr4(L286L)ρ0yDT1326A phe2AFαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht 1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)gex2(K241Q)ρ0yDT1347A phe2AHαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)ρ0yDT1368A phe2BCαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)ρ0yDT1389A phe23Cαhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] tda8(−275)zds1(A346S)whi4(H577D)dbf2(S297T)yfl040w(−26)ρ0yDT14011A 4C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] bem2(P1014P)gex2-ty5(deletion)cpr6(−59)ρ−yDT14214A 7C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] dad1(E50D)ura2(−524)ρ−yDT14812A 5C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] scc4(D65Y)ρ+yDT15213A 6C5i1αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht 1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] ena1(T949T, S967S, S985S)yil054W(D101H)ρ0yDT15415A yHs8αhis3Δ200 leu2Δ0 lys2Δ0 trp1Δ63 ura3Δ0 met15Δ0 hta2-htb2Δ0 hta1-htb1Δ0 hht1-shufflehhf1Δ0 hht2-hhf2Δ0 [pDT83 (pRS416-HTA2-HTB2-HHT1-HHF1/URA3/CEN-ARS/Amp)] mak32(A269S)ρ− TABLE 6PlasmidsPlasmidOtherPlasmidnamenamemarkersDescriptionpDT076pRS414-amp/TrppRS414 with HTA1, HTA1/HTB1 promoter, HTB1, each gene flankedHTA1HTB1by restriction sites. CYC1 and ADH1 terminators.pDT077pRS414-amp/TrppRS414 with human H2A, HTA1/HTB1 promoter, human H2B, eachhH2AhH2Bgene flanked by restriction sites. CYC1 and ADH1 terminators.pDT083yHistonesamp/UrapRS416-HTA2-HTB2-HHT1-HHF1. Shuffle plasmid, parental strain.pDT100amp/TrppRS414-hH3.1QKK-hH4pDT101amp/TrppRS414-hH3.1KK-hH4pDT102amp/TrppRS414-hH3.3QKK-hH4pDT103amp/TrppRS414-hH3.3KK-hH4pDT105yHistones2amp/TrppRS414-HHT2-HHF2-HTA-HTB1. Isogenic-WT plasmid.pDT107amp/TrppRS414-hH3.3-hH4-hH2A-hH2B (HHT2F2HTA1B1 PROs/TERs)pDT108amp/TrppRS414-hH3.1QKK-hH4-hH2A-hH2B (HHT2F2HTA1B1PROs/TERs)pDT109hHistonesamp/TrppRS414-hH3.1-hH4-hH2A-hH2B (HHT2F2HTA1B1 PROs/TERs).yHs strains.pDT110amp/TrppRS414-hH3.1KK-hH4-hH2A-hH2B (HHT2F2HTA1B 1 PROs/TERs)pDT111amp/TrppRS414-hH3.1QKK-hH4-hH2A2+4-hH2B (HHT2F2HTA1B1PROs/TERs)pDT112amp/TrppRS414-hH3.1QKK-hH4-hH2A4-hH2B (HHT2F2HTA1B1PROs/TERs)pDT113amp/TrppRS414-hH3.1KK-hH4-hH2A2+4-hH2B (HHT2F2HTA1B1PROs/TERs)pDT114amp/TrppRS414-hH3.1KK-hH4-hH2A4-hH2B (HHT2F2HTA1B1PROs/TERs)pDT116amp/LeuE99 with hH3.1hH4hH2A4hH2B(HHT2F2HTA1B1PROs/TERs)cloned in place of RFP. HO-integration vector with Leu tag.pDT121amp/TrppRS414-hH2ANc-hH2BpDT125amp/TrppRS414-hH3.1-hH4-hH2Ac-hH2B (HHT2F2HTA1B1 PROs/TERs)pDT126amp/TrppRS414-hH3.1-hH4-hH2AN-hH2B (HHT2F2HTA1B1 PROs/TERs)pDT127amp/TrppRS414-hH3.1-hH4-hH2ANc-hH2B (HHT2F2HTA1B1 PROs/TERs)pDT128amp/TrppRS414-hH3.1KK-hH4-hH2Ac-hH2B (HHT2F2HTA1B1PROs/TERs)pDT130amp/TrppRS414-hH3.1KK-hH4-hH2ANc-hH2B (HHT2F2HTA1B1PROs/TERs)pDT130amp/TrppRS424-hH3.1-hH4-hH2A-hH2B (HHT2F2HTA1B1 PROs/TERs) 2micron version TABLE 7PCRtag primers.Primer nameTargetSpeciesOrientationSequenceJB00133_DTHTB1/2YeastForwardGGTAACAGCTCTAGTACCTTCAGAG(SEQ ID NO: 1)JB00134_DTHTB1/2YeastReverseGCCGAAAAGAAACCAGC(SEQ ID NO: 2)JB00137_DTHTA1/2YeastForwardAGGTGGTAAAGCTGGTTCAG(SEQ ID NO: 3)JB00138_DTHTA1/2YeastReverseTTCTTGAGAAGCCTTGGTAGC(SEQ ID NO: 4)DT484HHT1/2YeastForwardGCTGCCAGAAAATCCGCC(SEQ ID NO: 5)DT557HHT1/2YeastReverseGCCAACTTGATATCCTTCTTTTGGATAGT(SEQ ID NO: 6)DT488HHF1/2YeastForwardAGAGGTAAAGGTGGTAAAGGTCTA(SEQ ID NO: 7)DT567HHF1/2YeastReverseGGATTTCAAGACNGCTCTGAC(SEQ ID NO: 8)JB00131_DThH2B.JHumanForwardGTAACAGCCTTGGTACCTTCAG(SEQ ID NO: 9)JB00132_DThH2B.JHumanReverseAACCAGCTAAGTCTGCTCCAG(SEQ ID NO: 10)JB00135_DThH2A.IHumanForwardAGAGGTAAGCAAGGTGGTAAGG(SEQ ID NO: 11)JB00136_DThH2A.IHumanReverseCTTACCCTTAGCCTTGTGGTG(SEQ ID NO: 12)DT482hH3HumanForwardAGGCTGCTAGAAAGTCTGCT(SEQ ID NO: 13)DT483hH3HumanReverseTCTCTTAGCGTGGATAGCACA(SEQ ID NO: 14)DT565hH4HumanForwardGGTGGTAAGGGTTTGGGTAAG(SEQ ID NO: 15)DT566hH4HumanReverseGAAAACCTTCAAAACACCTCTGGT(SEQ ID NO: 16) TABLE 8Illumina high-throughput whole-genome sequencing filesSRAFilesizeaccessionTypeFile name 1File name 2Description(Gb)DateSRR5359525PE150yDT51_R1_001.fastq.gzyDT51_R2_001.fastq.gzWGS of parental strain1.46; 1.6707-19-16SRR5359524PE150yHs1_R1_001.fastq.gzyHs1_R2_001.fastq.gzWGS of yHs1 original1.27; 1.4707-19-16SRR5359523PE150yHs2_R1_001.fastq.gzyHs2_R2_001.fastq.gzWGS of yHs2 original1.32; 1.5207-19-16SRR5359522PE150yHs3_R1_001.fastq.gzyHs3_R2_001.fastq.gzWGS of yHs3 original1.41; 1.6107-19-16SRR5359521PE150yHs1m_R1_001.fastq.gzyHs lm_R2_001.fastq.gzWGS of yHs1 maintenance1.30; 1.5007-19-16SRR5359520PE150yHs2m_R1_001.fastq.gzyHs2m_R2_001.fastq.gzWGS of yHs2 maintenance1.41; 1.6207-19-16SRR5359519PE150yHs3m_R1_001.fastq.gzyHs3m_R2_001.fastq.gzWGS of yHs3 maintenance1.41; 1.6107-19-16SRR5359518PE150yHs4_R1_001.fastq.gzyHs4_R2_001.fastq.gzWGS of yHs4 original1.38; 1.6007-19-16SRR5359517PE150yHs5_R1_001.fastq.gzyHs5_R2_001.fastq.gzWGS of yHs5 original1.32; 1.5307-19-16SRR5359516PE150yHs7_R1_001.fastq.gzyHs7_R2_001.fastq.gzWGS of yHs7 original1.41; 1.6207-19-16SRR5359515PE150yHs1C5_R1_001.fastq.gzyHs1C5_R2_001.fastq.gzWGS of yHs1 cycle 51.44; 1.6507-19-16SRR5359514PE150yHs2C5_R1_001.fastq.gzyHs2C5_R2_001.fastq.gzWGS of yHs2 cycle 51.28; 1.4507-19-16SRR5359513PE150yHs3C5_R1_001.fastq.gzyHs3C5_R2_001.fastq.gzWGS of yHs3 cycle 51.36; 1.5607-19-16SRR5359512PE100yHs6_R1_001.fastq.gzyHs6_R2_001.fastq.gzWGS of yHs6 original0.53; 0.5209-02-16SRR5359511PE100yHs8_R1_001.fastq.gzyHs8_R2_001.fastq.gzWGS of yHs8 original0.60; 0.5909-02-16SRR5359510PE100yHs4C5_R1_001.fastq.gzyHs4C5_R2_001.fastq.gzWGS of yHs4 cycle 50.96; 0.9409-02-16SRR5359509PE100yHs5C5_R1_001.fastq.gzyHs5C5_R2_001.fastq.gzWGS of yHs5 cycle 50.75; 0.7409-02-16SRR5359508PE100yHs6C5_R1_001.fastq.gzyHs6C5_R2_001.fastq.gzWGS of yHs6 cycle 50.53; 0.5209-02-16SRR5359507PE100yHs7C5_R1_001.fastq.gzyHs7C5_R2_001.fastq.gzWGS of yHs7 cycle 50.52; 0.5209-02-16SRR5359506PE100yHs1C5i2_R1_001.fastq.gzyHs1C5i2_R2_001.fastq.gzWGS of yHs1 cycle 50.56; 0.5509-02-16isolate 2SRR5359505PE100yHs2C5i1_R1_001.fastq.gzyHs2C5i1_R2_001.fastq.gzWGS of yHs2 cycle 51.01; 0.9909-02-16isolate 1SRR5359504PE100yHs3C5i1_R1_001.fastq.gzyHs3C5i1_R2_001.fastq.gzWGS of yHs3 cycle 50.71; 0.9609-02-16isolate 1SRR5359503PE100yHs4C5i1_R1_001.fastq.gzyHs4C5i1_R2_001.fastq.gzWGS of yHs4 cycle 50.68; 0.6809-02-16isolate 1SRR5359502PE100yHs5C5i1_R1_001.fastq.gzyHs5C5i1_R2_001.fastq.gzWGS of yHs5 cycle 50.82; 0.8109-02-16isolate 1SRR5359501PE100yHs6C5i1_R1_001.fastq.gzyHs6C5i1_R2_001.fastq.gzWGS of yHs6 cycle 50.58; 0.5809-02-16isolate 1SRR5359500PE100yHs7C5i1_R1_001.fastq.gzyHs7C5i1_R2_001.fastq.gzWGS of yHs7 cycle 50.69; 0.6809-02-16isolate 1SRR5359499PE100phe2AE_R1_001.fastq.gzphe2AE_R2_001.fastq.gzWGS of phe2AE0.77; 0.7609-02-16SRR5359498PE100phe2AF_R1_001.fastq.gzphe2AF_R2_001.fastq.gzWGS of phe2AF0.45; 0.4509-02-16SRR5359497PE100phe2AH_R1_001.fastq.gzphe2AH_R2_001.fastq.gzWGS of phe2AH0.68; 0.6709-02-16SRR5359496PE100phe2BC_R1_001.fastq.gzphe2BC_R2_001.fastq.gzWGS of phe2BC0.67; 0.6909-02-16SRR5359495PE100phe2BD_R1_001.fastq.gzphe2BD_R2_001.fastq.gzWGS of phe2BD0.57; 0.5609-02-16SRR5359494PE100phe23C_R1_001.fastq.gzphe23C_R2_001.fastq.gzWGS of phe23C0.55; 0.5509-02-16SRR5359493PE1002PC6i1_R1_001.fastq.gz2PC6i1_R2_001.fastq.gzWGS of yHs2 plate cycle 60.54; 0.5309-02-16isolate 1SRR5359492PE100yHs1C5il_R1_001.fastq.gzyHs1C5il_R2_001.fastq.gzWGS of yHs1 cycle 50.61; 0.6009-02-16isolate 1SRR5359491PE1001PC5i5_R1_001.fastq.gz1PC5i5_R2_001.fastq.gzWGS of yHs1 plate cycle 50.58; 0.5809-02-16isolate 5SRR5359490PE1001PC5i3_R1_001.fastq.gz1PC5i3_R2_001.fastq.gzWGS of yHs1 plate cycle 50.37; 0.3609-02-16isolate 3SRR5359489PE100yDT97_R1_001.fastq.gzyDT97_R2_001.fastq.gzWGS of yDT970.78; 0.7309-02-16SRR5359488PE100yDT98_R1_001.fastq.gzyDT98_R2_001.fastq.gzWGS of yDT980.82; 0.8009-02-16 TABLE 9Illumina high-throughput MNase DNA sequencing filesFilesizeSRA accessionTypeFile name 1File name 2Description(Gb)DateSAMN06619428PE150WTH1_R1_001.fastq.gzWTH1_R2_001.fastq.gzyDT67 (WT) high MNase0.76; 0.8912-08-16experiment 1, bio rep 1SAMN06619429PE150WTH2_R1_001.fastq.gzWTH2_R2_001.fastq.gzyDT67 (WT) high MNase0.73; 0.8512-08-16experiment 1, bio rep 2SAMN06619430PE150WTH3_R1_001.fastq.gzWTH3_R2_001.fastq.gzyDT67 (WT) high MNase0.69; 0.8012-08-16experiment 1, bio rep 3SAMN06619431PE150WTL1_R1_001.fastq.gzWTL1_R2_001.fastq.gzyDT67 (WT) low MNase0.87; 0.9912-08-16experiment 1, bio rep 1SAMN06619432PE150WTL2_R1_001.fastq.gzWTL2_R2_001.fastq.gzyDT67 (WT) low MNase0.93; 1.0912-08-16experiment 1, bio rep 2SAMN06619433PE150WTL3_R1_001.fastq.gzWTL3_R2_001.fastq.gzyDT67 (WT) low MNase0.85; 0.9812-08-16experiment 1, bio rep 3SAMN06619434PE1505C5iH1_R1_001.fastq.gz5C5iHl_R2_001.fastq.gzyHs5C5i1 high MNase0.87; 1.0012-08-16experiment 1, bio rep 1SAMN06619435PE1505C5iH2_R1_001.fastq.gz5C5iH2_R2_001.fastq.gzyHs5C5i1 high MNase0.83; 1.0012-08-16experiment 1, bio rep 2SAMN06619436PE1505C5iH3_R1_001.fastq.gz5C5iH3_R2_001.fastq.gzyHs5C5i1 high MNase0.86; 1.0012-08-16experiment 1, bio rep 3SAMN06619437PE1505C5iL1_R1_001.fastq.gz5C5iL1_R2_001.fastq.gzyHs5C5i1 low MNase0.98; 1.2012-08-16experiment 1, bio rep 1SAMN06619438PE1505C5iL2_R1_001.fastq.gz5C5iL2_R2_001.fastq.gzyHs5C5i1 low MNase0.93; 1.1312-08-16experiment 1, bio rep 2SAMN06619439PE1505C5iL3_R1_001.fastq.gz5C5iL3_R2_001.fastq.gzyHs5C5i1 low MNase0.93; 1.1412-08-16experiment 1, bio rep 3SAMN06619440PE1507C5iH1_R1_001.fastq.gz7C5iH1_R2_001.fastq.gzyHs7C5i1 high MNase0.75; 0.8612-08-16experiment 1, bio rep 1SAMN06619441PE1507C5iH2_R1_001.fastq.gz7C5iH2_R2_001.fastq.gzyHs7C5i1 high MNase0.77; 0.9012-08-16experiment 1, bio rep 2SAMN06619442PE1507C5iH3_R1_001.fastq.gz7C5iH3_R2_001.fastq.gzyHs7C5i1 high MNase0.77; 0.8912-08-16experiment 1, bio rep 3SAMN06619443PE1507C5iL1_R1_001.fastq.gz7C5iL1_R2_001.fastq.gzyHs7C5i1 low MNase0.90; 1.0612-08-16experiment 1, bio rep 1SAMN06619444PE1507C5iL2_R1_001.fastq.gz7C5iL2_R2_001.fastq.gzyHs7C5i1 low MNase0.84; 0.9812-08-16experiment 1, bio rep 2SAMN06619445PE1507C5iL3_R1_001.fastq.gz7C5iL3_R2_001.fastq.gzyHs7C5i1 low MNase0.79; 0.8212-08-16experiment 1, bio rep 3SAMN06619446PE15097H1_R1_001.fastq.gz97H1_R2_001.fastq.gzyDT97 high MNase0.81; 0.9312-08-16experiment 1, bio rep 1SAMN06619447PE15097H2_R1_001.fastq.gz97H2_R2_001.fastq.gzyDT97 high MNase0.82; 0.9312-08-16experiment 1, bio rep 2SAMN06619448PE15097H3_R1_001.fastq.gz97H3_R2_001.fastq.gzyDT97 high MNase0.82; 0.9412-08-16experiment 1, bio rep 3SAMN06619449PE15097L1_R1_001.fastq.gz97L l_R2_001.fastq.gzyDT97 low MNase0.94; 1.1012-08-16experiment 1, bio rep 1SAMN06619450PE15097L2_R1_001.fastq.gz97L2_R2_001.fastq.gzyDT97 low MNase0.75; 0.8712-08-16experiment 1, bio rep 2SAMN06619451PE15097L3_R1_001.fastq.gz97L3_R2_001.fastq.gzyDT97low MNase0.95; 1.1312-08-16experiment 1, bio rep 3SAMN06619452PE150WTH4_R1_001.fastq.gzWTH4_R2_001.fastq.gzyDT67 high MNase1.32; 1.5301-30-17experiment 2SAMN06619453PE150WTL4_R1_001.fastq.gzWTL4_R2_001.fastq.gzyDT67 low MNase1.31; 1.5901-30-17experiment 2SAMN06619454PE1505C5iH4_R1_001.fastq.gz5C5iH4_R2_001.fastq.gzyHs5C5i1 high MNase1.31; 1.5201-30-17experiment 2SAMN06619455PE1505C5iL4_R1_001.fastq.gz5C5iL4_R2_001.fastq.gzyHs5C5i1 low MNase1.50; 1.8501-30-17experiment 2SAMN06619456PE1507C5iH4_R1_001.fastq.gz7C5iH4_R2_001.fastq.gzyHs7C5i1 high MNase1.16; 1.3901-30-17experiment 2SAMN06619457PE1507C5iL4_R1_001.fastq.gz7C5iL4_R2_001.fastq.gzyHs7C5i1 low MNase1.30; 1.5701-30-17experiment 2SAMN06619458PE15097H4_R1_001.fastq.gz97H4_R2_001.fastq.gzyDT97 high MNase1.47; 1.6701-30-17experiment 2SAMN06619459PE15097L4_R1_001.fastq.gz97L4_R2_001.fastq.gzyDT97 low MNase1.57; 1.8701-30-17experiment 2 REFERENCES Acker, J., Conesa, C., and Lefebvre, O. (2013). Yeast RNA polymerase III transcription factors and effectors. Biochim Biophys Acta 1829, 283-295.Annunziato, A. T. (2005). Split decision: what happens to nucleosomes during DNA replication? J Biol Chem 280, 12065-12068.Bernstein, E., and Hake, S. B. (2006). The nucleosome: a little variation goes a long way. Biochem Cell Biol 84, 505-517.Boeke, J. D., Trueheart, J., Natsoulis, G., and Fink, G. R. (1987). 5-Fluoroorotic acid as a selective agent in yeast molecular genetics. Methods Enzymol 154, 164-175.Bolger, A. M., Lohse, M., and Usadel, B. (2014). Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114-2120.Brogaard, K., Xi, L., Wang, J. P., and Widom, J. (2012). A map of nucleosome positions in yeast at base-pair resolution. Nature 486, 496-501.Budhavarapu, V. N., Chavez, M., and Tyler, J. K. (2013). How is epigenetic information maintained through DNA replication? Epigenetics Chromatin 6, 32.Buschbeck, M., and Hake, S. B. (2017). Variants of core histones and their roles in cell fate decisions, development and cancer. Nat Rev Mol Cell Biol.Campos, E. I., Smits, A. H., Kang, Y. H., Landry, S., Escobar, T. M., Nayak, S., Ueberheide, B. M., Durocher, D., Vermeulen, M., Hurwitz, J., et al. (2015). Analysis of the Histone H3.1 Interactome: A Suitable Chaperone for the Right Event. Mol Cell 60, 697-709.Campos, E. I., Stafford, J. M., and Reinberg, D. (2014). Epigenetic inheritance: histone bookmarks across generations. Trends Cell Biol 24, 664-674.Chen, K., Xi, Y., Pan, X., Li, Z., Kaestner, K., Tyler, J., Dent, S., He, X., and Li, W. (2013). DANPOS: dynamic analysis of nucleosome position and occupancy by sequencing. Genome Res 23, 341-351.Dai, J., Hyland, E. M., Yuan, D. S., Huang, H., Bader, J. S., and Boeke, J. D. (2008). Probing nucleosome function: a highly versatile library of synthetic histone H3 and H4 mutants. Cell 134, 1066-1078.DiCarlo, J. E., Norville, J. E., Mali, P., Rios, X., Aach, J., and Church, G. M. (2013). Genome engineering inSaccharomyces cerevisiaeusing CRISPR-Cas systems. Nucleic Acids Res 41, 4336-4343.Djebali, S., Davis, C. A., Merkel, A., Dobin, A., Lassmann, T., Mortazavi, A., Tanzer, A., Lagarde, J., Lin, W., Schlesinger, F., et al. (2012). Landscape of transcription in human cells. Nature 489, 101-108.Eriksson, P. R., Ganguli, D., Nagarajavel, V., and Clark, D. J. (2012). Regulation of histone gene expression in budding yeast. Genetics 191, 7-20.Fan, Y., Nikitina, T., Zhao, J., Fleury, T. J., Bhattacharyya, R., Bouhassira, E. E., Stein, A., Woodcock, C. L., and Skoultchi, A. I. (2005). Histone H1 depletion in mammals alters global chromatin structure but causes specific changes in gene regulation. Cell 123, 1199-1212.Floer, M., Wang, X., Prabhu, V., Berrozpe, G., Narayan, S., Spagna, D., Alvarez, D., Kendall, J., Krasnitz, A., Stepansky, A., et al. (2010). A RSC/nucleosome complex determines chromatin architecture and facilitates activator binding. Cell 141, 407-418.Gibson, D. G., Young, L., Chuang, R. Y., Venter, J. C., Hutchison, C. A., 3rd, and Smith, H. O. (2009). Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat Methods 6, 343-345.Hage, A. E., and Houseley, J. (2013). Resolution of budding yeast chromosomes using pulsed-field gel electrophoresis. Methods Mol Biol 1054, 195-207.Hamza, A., Tammpere, E., Kofoed, M., Keong, C., Chiang, J., Giaever, G., Nislow, C., and Hieter, P. (2015). Complementation of Yeast Genes with Human Genes as an Experimental Platform for Functional Testing of Human Genetic Variants. Genetics 201, 1263-1274.Han, J., Zhang, H., Zhang, H., Wang, Z., Zhou, H., and Zhang, Z. (2013). A Cul4 E3 ubiquitin ligase regulates histone hand-off during nucleosome assembly. Cell 155, 817-829.Hyland, E. M., Molina, H., Poorey, K., Jie, C., Xie, Z., Dai, J., Qian, J., Bekiranov, S., Auble, D. T., Pandey, A., et al. (2011). An evolutionarily ‘young’ lysine residue in histone H3 attenuates transcriptional output inSaccharomyces cerevisiae. Genes Dev 25, 1306-1319.Janke, C., Ortiz, J., Lechner, J., Shevchenko, A., Shevchenko, A., Magiera, M. M., Schramm, C., and Schiebel, E. (2001). The budding yeast proteins Spc24p and Spc25p interact with Ndc80p and Nuf2p at the kinetochore and are important for kinetochore clustering and checkpoint control. EMBO J 20, 777-791.Kachroo, A. H., Laurent, J. M., Yellman, C. M., Meyer, A. G., Wilke, C. O., and Marcotte, E. M. (2015). Evolution. Systematic humanization of yeast genes reveals conserved functions and genetic modularity. Science 348, 921-925.Krietenstein, N., Wal, M., Watanabe, S., Park, B., Peterson, C. L., Pugh, B. F., and Korber, P. (2016). Genomic Nucleosome Organization Reconstituted with Pure Proteins. Cell 167, 709-721 e712.Kubik, S., Bruzzone, M. J., Jacquet, P., Falcone, J. L., Rougemont, J., and Shore, D. (2015). Nucleosome Stability Distinguishes Two Different Promoter Types at All Protein-Coding Genes in Yeast. Mol Cell 60, 422-434.Kwolek-Mirek, M., and Zadrag-Tecza, R. (2014). Comparison of methods used for assessing the viability and vitality of yeast cells. FEMS Yeast Res 14, 1068-1079.Lai, W. K. M., and Pugh, B. F. (2017). Understanding nucleosome dynamics and their links to gene expression and DNA replication. Nat Rev Mol Cell Biol.Laurent, J. M., Young, J. H., Kachroo, A. H., and Marcotte, E. M. (2016). Efforts to make and apply humanized yeast. Brief Funct Genomics 15, 155-163.Lee, K. P., Baxter, H. J., Guillemette, J. G., Lawford, H. G., and Lewis, P. N. (1982). Structural studies on yeast nucleosomes. Can J Biochem 60, 379-388.Leung, A., Cheema, M., Gonzalez-Romero, R., Eirin-Lopez, J. M., Ausio, J., and Nelson, C. J. (2016). Unique yeast histone sequences influence octamer and nucleosome stability. FEBS Lett 590, 2629-2638.Lew, D. J., and Reed, S. I. (1995). A cell cycle checkpoint monitors cell morphogenesis in budding yeast. J Cell Biol 129, 739-749.Li, H. (2011). A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data. Bioinformatics 27, 2987-2993.Li, H., and Durbin, R. (2010). Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 26, 589-595.Looke, M., Kristjuhan, K., and Kristjuhan, A. (2011). Extraction of genomic DNA from yeasts for PCR-based applications. Biotechniques 50, 325-328.Lopez-Serra, L., Kelly, G., Patel, H., Stewart, A., and Uhlmann, F. (2014). The Scc2-Scc4 complex acts in sister chromatid cohesion and transcriptional regulation by maintaining nucleosome-free regions. Nat Genet 46, 1147-1151.Ma, Y., Kanakousaki, K., and Buttitta, L. (2015). How the cell cycle impacts chromatin architecture and influences cell fate. Front Genet 6, 19.Macadangdang, B. R., Oberai, A., Spektor, T., Campos, O. A., Sheng, F., Carey, M. F., Vogelauer, M., and Kurdistani, S. K. (2014). Evolution of histone 2A for chromatin compaction in eukaryotes. Elife 3.McBurney, K. L., Leung, A., Choi, J. K., Martin, B. J., Irwin, N. A., Bartke, T., Nelson, C. J., and Howe, L. J. (2016). Divergent Residues Within Histone H3 Dictate a Unique Chromatin Structure inSaccharomyces cerevisiae. Genetics 202, 341-349.Mi, H., Poudel, S., Muruganujan, A., Casagrande, J. T., and Thomas, P. D. (2016). PANTHER version 10: expanded protein families and functions, and analysis tools. Nucleic Acids Res 44, D336-342.Mitchell, L. A., Phillips, N. A., Lafont, A., Martin, J. A., Cutting, R., and Boeke, J. D. (2015). qPCRTag Analysis—A High Throughput, Real Time PCR Assay for Sc2.0 Genotyping. J Vis Exp, e52941.Nakanishi, S., Sanderson, B. W., Delventhal, K. M., Bradford, W. D., Staehling-Hampton, K., and Shilatifard, A. (2008). A comprehensive library of histone mutants identifies nucleosomal residues required for H3K4 methylation. Nat Struct Mol Biol 15, 881-888.Osborn, M. J., and Miller, J. R. (2007). Rescuing yeast mutants with human genes. Brief Funct Genomic Proteomic 6, 104-111.Panday, A., and Grove, A. (2016). The high mobility group protein HMO1 functions as a linker histone in yeast. Epigenetics Chromatin 9, 13.Pavelka, N., Rancati, G., Zhu, J., Bradford, W. D., Saraf, A., Florens, L., Sanderson, B. W., Hattem, G. L., and Li, R. (2010). Aneuploidy confers quantitative proteome changes and phenotypic variation in budding yeast. Nature 468, 321-325.Quinlan, A. R. (2014). BEDTools: The Swiss-Army Tool for Genome Feature Analysis. Curr Protoc Bioinformatics 47, 11 12 11-34.Radman-Livaja, M., Verzijlbergen, K. F., Weiner, A., van Welsem, T., Friedman, N., Rando, O. J., and van Leeuwen, F. (2011). Patterns and mechanisms of ancestral histone protein inheritance in budding yeast. PLoS Biol 9, e1001075.Rando, O. J., and Winston, F. (2012). Chromatin and transcription in yeast. Genetics 190, 351-387.Rosebrock, A. P. (2017). Analysis of the Budding Yeast Cell Cycle by Flow Cytometry. Cold Spring Harb Protoc 2017, pdb prot088740.Sanchez-Perez, I., Renwick, S. J., Crawley, K., Karig, I., Buck, V., Meadows, J. C., Franco-Sanchez, A., Fleig, U., Toda, T., and Millar, J. B. (2005). The DASH complex and Klp5/Klp6 kinesin coordinate bipolar chromosome attachment in fission yeast. EMBO J 24, 2931-2943.Santenard, A., and Torres-Padilla, M. E. (2009). Epigenetic reprogramming in mammalian reproduction: contribution from histone variants. Epigenetics 4, 80-84.Segal, E., and Widom, J. (2009). What controls nucleosome positions? Trends Genet 25, 335-343.Sheltzer, J. M., Blank, H. M., Pfau, S. J., Tange, Y., George, B. M., Humpton, T. J., Brito, I. L., Hiraoka, Y., Niwa, O., and Amon, A. (2011). Aneuploidy drives genomic instability in yeast. Science 333, 1026-1030.Shen, L., Shao, N., Liu, X., and Nestler, E. (2014). ngs.plot: Quick mining and visualization of next-generation sequencing data by integrating genomic databases. BMC Genomics 15, 284.Szklarczyk, D., Franceschini, A., Wyder, S., Forslund, K., Heller, D., Huerta-Cepas, J., Simonovic, M., Roth, A., Santos, A., Tsafou, K. P., et al. (2015). STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43, D447-452.Talbert, P. B., and Henikoff, S. (2010). Histone variants—ancient wrap artists of the epigenome. Nat Rev Mol Cell Biol 11, 264-275.Talbert, P. B., and Henikoff, S. (2017). Histone variants on the move: substrates for chromatin dynamics. Nat Rev Mol Cell Biol 18, 115-126.Thurman, R. E., Rynes, E., Humbert, R., Vierstra, J., Maurano, M. T., Haugen, E., Sheffield, N.C., Stergachis, A. B., Wang, H., Vernot, B., et al. (2012). The accessible chromatin landscape of the human genome. Nature 489, 75-82.Toyama, B. H., Savas, J. N., Park, S. K., Harris, M. S., Ingolia, N. T., Yates, J. R., 3rd, and Hetzer, M. W. (2013). Identification of long-lived proteins reveals exceptional stability of essential cellular structures. Cell 154, 971-982.Valouev, A., Johnson, S. M., Boyd, S. D., Smith, C. L., Fire, A. Z., and Sidow, A. (2011). Determinants of nucleosome organization in primary human cells. Nature 474, 516-520.Veatch, J. R., McMurray, M. A., Nelson, Z. W., and Gottschling, D. E. (2009). Mitochondrial dysfunction leads to nuclear genome instability via an iron-sulfur cluster defect. Cell 137, 1247-1258.Wen, D., Banaszynski, L. A., Liu, Y., Geng, F., Noh, K. M., Xiang, J., Elemento, O., Rosenwaks, Z., Allis, C. D., and Rafii, S. (2014). Histone variant H3.3 is an essential maternal factor for oocyte reprogramming. Proc Natl Acad Sci USA 111, 7325-7330.White, C. L., Suto, R. K., and Luger, K. (2001). Structure of the yeast nucleosome core particle reveals fundamental changes in internucleosome interactions. EMBO J 20, 5207-5218.Woodcock, C. L., Skoultchi, A. I., and Fan, Y. (2006). Role of linker histone in chromatin structure and function: H1 stoichiometry and nucleosome repeat length. Chromosome Res 14, 17-25.Zhang, T., Lei, J., Yang, H., Xu, K., Wang, R., and Zhang, Z. (2011). An improved method for whole protein extraction from yeastSaccharomyces cerevisiae. Yeast 28, 795-798. Although the embodiments have been described in detail for the purposes of illustration, it is understood that such detail is solely for that purpose, and variations can be made therein by those skilled in the art without departing from the spirit and scope of the disclosure, embodiments of which are defined by the following sample claims.
103,543
11858969
DETAILED DESCRIPTION In the following detailed description, reference is made to the accompanying drawings, which form a part hereof. In the drawings, similar symbols typically identify similar components, unless context dictates otherwise. The illustrative embodiments described in the detailed description, drawings, and claims are not meant to be limiting. Other embodiments may be utilized, and other changes may be made, without departing from the spirit or scope of the subject matter presented herein. It will be readily understood that the aspects of the present disclosure, as generally described herein, and illustrated in the Figures, can be arranged, substituted, combined, separated, and designed in a wide variety of different configurations, all of which are explicitly contemplated herein and make part of this disclosure The present application provides recombinant or synthetic light-sensitive proteins with improved properties, such as improvement in light sensitivity, ion conductance, or both. Also provided herein include nucleic acid molecules comprising coding sequences for the light-sensitive proteins; cells comprising the light-sensitive proteins, the nucleic acid molecules comprising the coding sequence for the light-sensitive proteins, or both; and compositions comprises the proteins, the nucleic acid molecules, the cells, or any combination thereof. Methods for expressing a light-sensitive protein in a subject are also provided. The methods can be used, for example, treating or ameliorating ocular disorders and neuronal disorders, restoring or enhancing the visual function of the subject, restoring or enhancing the photosensitivity of the retinal neurons in the subject, restoring or enhancing the photosensitivity of a retina or a portion thereof of the subject; treating or ameliorating blindness or vision loss caused by retinal detachment and/or photoreceptor loss due to trauma or head injury. The method can also be used to effect light-controlled neuronal activation in the subject, or to control light-induced behaviors for the subject. Definitions Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the present disclosure belongs. See, e.g. Singleton et al., Dictionary of Microbiology and Molecular Biology 2nd ed., J. Wiley & Sons (New York, NY 1994); Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Springs Harbor Press (Cold Springs Harbor, NY 1989). For purposes of the present disclosure, the following terms are defined below. As used herein, the term “vector,” can refer to a vehicle for carrying or transferring a nucleic acid. Non-limiting examples of vectors include viral vectors (for example, adenovirus vectors, adeno-associated virus (AAV) vectors, retrovirus vectors, lentiviral vectors, herpes virus vectors, phages, and poxvirus vectors); non-viral vectors such as liposomes, naked DNA, plasmids, cosmids; and the like. As used herein, the term “construct,” refers to a recombinant nucleic acid that has been generated for the purpose of the expression of a specific nucleotide sequence(s), or that is to be used in the construction of other recombinant nucleotide sequences. As used herein, the term “plasmid” refers to a nucleic acid that can be used to replicate recombinant DNA sequences within a host organism. The sequence can be a double stranded DNA. As used herein, the terms “nucleic acid” and “polynucleotide” are interchangeable and refer to any nucleic acid, whether composed of phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphoramidate, bridged phosphoramidate, bridged methylene phosphonate, phosphorothioate, methylphosphonate, phosphorodithioate, bridged phosphorothioate or sultone linkages, and combinations of such linkages. The terms “nucleic acid” and “polynucleotide” also specifically include nucleic acids composed of bases other than the five biologically occurring bases (adenine, guanine, thymine, cytosine and uracil). The term “element” refers to a separate or distinct part of something, for example, a nucleic acid sequence with a separate function within a longer nucleic acid sequence. The terms “transcription regulatory element” and “expression control element” are used to refer to nucleic acid molecules that can influence the expression (including at the transcription and/or translation level) of an operably linked coding sequence in a specific host organism. These terms are used broadly to and cover all elements that promote or regulate transcription, including promoters, core elements required for basic interaction of RNA polymerase and transcription factors, upstream elements, enhancers, and response elements (see, e.g., Lewin, “Genes V” (Oxford University Press, Oxford) pages 847-873). Exemplary regulatory elements in prokaryotes include promoters, operator sequences and ribosome binding sites. Regulatory elements that are used in eukaryotic cells can include, without limitation, transcriptional and translational control sequences, such as promoters, enhancers, splicing signals, polyadenylation signals, terminators, protein degradation signals, internal ribosome-entry element (IRES), 2A sequences, and the like, that provide for and/or regulate expression of a coding sequence and/or production of an encoded polypeptide in a host cell. The promoter can be a specific promoter, e.g., cell type-specific and/or tissue-specific. The promoter can be constituent or inducible (e.g., by chemical agent, biological agent, temperature, and/or pH). As used herein, the term “variant” refers to a polynucleotide or polypeptide having a sequence substantially similar to a reference (e.g., the parent) polynucleotide or polypeptide. In the case of a polynucleotide, a variant can have deletions, substitutions, additions of one or more nucleotides at the 5′ end, 3′ end, and/or one or more internal sites in comparison to the reference polynucleotide. Similarities and/or differences in sequences between a variant and the reference polynucleotide can be detected using conventional techniques known in the art, for example polymerase chain reaction (PCR) and hybridization techniques. Variant polynucleotides also include synthetically derived polynucleotides, such as those generated, for example, by using site-directed mutagenesis. Generally, a variant of a polynucleotide, including, but not limited to, a DNA, can have at least, or at least about, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to the reference polynucleotide as determined by sequence alignment programs known in the art. In the case of a polypeptide, a variant can have deletions, substitutions, additions of one or more amino acids in comparison to the reference polypeptide. Similarities and/or differences in sequences between a variant and the reference polypeptide can be detected using conventional techniques known in the art, for example Western blot. A variant of a polypeptide can have, for example, at least, or at least about, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to the reference polypeptide as determined by sequence alignment programs known in the art. The term “AAV” or “adeno-associated virus” refers to a Dependoparvovirus within the Parvoviridae genus of viruses. Unless specified otherwise, the left-hand end of any single-stranded polynucleotide sequence discussed herein is the 5′ end; the left-hand direction of double-stranded polynucleotide sequences is referred to as the 5′ direction. The term “naturally occurring” as used herein refers to materials which are found in nature or a form of the materials that is found in nature. Standard techniques can be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques can be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures can be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose. Unless specific definitions are provided, the nomenclatures utilized in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those commonly known and used in the art. Standard techniques can be used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. As used herein, a “subject” refers to an animal that is the object of treatment, observation or experiment. “Animal” includes cold- and warm-blooded vertebrates (e.g., mammals) and invertebrates (e.g., fish, shellfish and reptiles). “Mammal,” as used herein, refers to an individual belonging to the class Mammalia and includes, but not limited to, humans, domestic and farm animals, zoo animals, sports and pet animals. Non-limiting examples of mammals include mice; rats; rabbits; guinea pigs; dogs; cats; sheep; goats; cows; horses; primates, such as monkeys, chimpanzees, apes, and humans. In some embodiments, the subject is a human. However, in some embodiments, the subject is not a human. As used herein, the term “treatment” refers to an intervention made in response to a disease, disorder or physiological condition manifested by a patient, particularly a patient suffering from one or more serotonin-related diseases. The aim of treatment may include, but is not limited to, one or more of the alleviation or prevention of symptoms, slowing or stopping the progression or worsening of a disease, disorder, or condition and the remission of the disease, disorder or condition. The term “treat” and “treatment” includes, for example, therapeutic treatments, prophylactic treatments, and applications in which one reduces the risk that a subject will develop a disorder or other risk factor. Treatment does not require the complete curing of a disorder and encompasses embodiments in which one reduces one or more symptoms of the disorder and/or underlying risk factors. In some embodiments, “treatment” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already affected by a disease or disorder or undesired physiological condition as well as those at a risk of developing the disease or disorder, and those in which the disease or disorder or undesired physiological condition is to be prevented. For example, in some embodiments treatment may enhance or reduce the level of serotonin in the subject, thereby to reduce, alleviate, or eradicate the symptom(s) of the disease(s). As used herein, the term “prevention” refers to any activity that reduces the burden of the individual later expressing those serotonin-related disease symptoms. This can take place at primary, secondary and/or tertiary prevention levels, wherein: a) primary prevention avoids the development of symptoms/disorder/condition; b) secondary prevention activities are aimed at early stages of the condition/disorder/symptom treatment, thereby increasing opportunities for interventions to prevent progression of the condition/disorder/symptom and emergence of symptoms; and c) tertiary prevention reduces the negative impact of an already established condition/disorder/symptom by, for example, restoring function and/or reducing any condition/disorder/symptom or related complications. The term “prevent” does not require the 100% elimination of the possibility of an event. Rather, it denotes that the likelihood of the occurrence of the event has been reduced in the presence of the compound or method. As used herein, the term “effective amount” refers to an amount sufficient to effect beneficial or desirable biological and/or clinical results. As used herein, a “therapeutically effective amount” of a compound is an amount sufficient to provide any therapeutic benefit in the treatment or management of a disorder (e.g., a neuron mediated disorder or an ocular disorder), or to delay or minimize one or more symptoms associated with a disorder (e.g., a neuron mediated disorder or an ocular disorder). A therapeutically effective amount of an agent (e.g., a light-sensitive protein) refers to an amount of the agent, alone or in combination with one or more other therapies and/or therapeutic agents that provide any therapeutic benefit in the treatment or management of a disorder (e.g., a neuron mediated disorder or an ocular disorder). The term “therapeutically effective amount” can encompass an amount that alleviates a neuron mediated disorder or ocular disorder, improves or reduces the neuron mediated disorder or the ocular disorder, improves overall therapy, or enhances the therapeutic efficacy of another therapeutic agent. “Pharmaceutically acceptable” carriers are ones which are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed. “Pharmaceutically acceptable” carriers can be, but not limited to, organic or inorganic, solid or liquid excipients which is suitable for the selected mode of application such as oral application or injection, and administered in the form of a conventional pharmaceutical preparation, such as solid such as tablets, granules, powders, capsules, and liquid such as solution, emulsion, suspension and the like. Often the physiologically acceptable carrier is an aqueous pH buffered solution such as phosphate buffer or citrate buffer. The physiologically acceptable carrier may also comprise one or more of the following: antioxidants including ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, such as serum albumin, gelatin, immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone, amino acids, carbohydrates including glucose, mannose, or dextrins, chelating agents such as EDTA, sugar alcohols such as mannitol or sorbitol, salt-forming counterions such as sodium, and nonionic surfactants such as Tween™, polyethylene glycol (PEG), and Pluronics™ Auxiliary, stabilizer, emulsifier, lubricant, binder, pH adjustor controller, isotonic agent and other conventional additives may also be added to the carriers. As used herein, the term “blindness” refers to total or partial loss of vision. The blindness can be caused by, for example, degeneration or non-functioning of photoreceptors caused by any diseases and conditions (e.g., physical injuries). In some embodiments, the blindness is associated with conditions such as glaucoma, late stage diabetic retinopathy, hereditary optic neuropathies, optic nerve injuries, and any combination thereof. As used herein, the term “vision” refers to the ability of a subject to detect light as a stimulus for differentiation or action. Vision is intended to encompass the following: (1) light detection or perception, that is the ability to discern whether or not light is present; (2) light projection, that is the ability to discern the direction from which a light stimulus is coming; (3) resolution, that is the ability to detect differing brightness levels (i.e., contrast) in a grating or letter target; and (4) recognition, that is the ability to recognize the shape of a visual target by reference to the differing contrast levels within the target. Thus, “vision” encompasses the ability to simply detect the presence of light (for example red light), including light having a wavelength between about 365 nm and about 700 nm, between about 530 nm and about 640 nm. In some embodiments, a peak activation occurs upon contact with light having a wavelength of about 590 nm. In some embodiments, transfection of retinal neurons with a nucleic acid molecule (e.g. vector) encoding a light-sensitive protein disclosed herein provides retinal neurons, for example bipolar cells and/or ganglion cells, with photosensitive membrane channels. Thus, it is possible to measure, with a light stimulus, the transmission of a visual stimulus to the animal's visual cortex, the area of the brain responsible for processing visual signals which constitutes a form of vision, as intended herein. As used herein, the term “retinal cell” can refer herein to any of the cell types that comprise the retina, such as retinal ganglion cells; amacrine cells; horizontal cells; bipolar cells; and photoreceptor cells including rods and cones. As used herein, the terms “light sensitivity” and “photosensitivity” are used interchangeably and refer to a notable or increased reactivity to light. Light-Sensitive Proteins Engineered light-sensitive proteins, including channelrhodopsins (ChRs) with desirable current strength and light sensitivity, are provided. ChRs are light-gated ion channels found in photosynthetic algae. Transgenic expression of ChRs in the brain enables light-dependent neuronal activation. These channels are widely applied as tools in neuroscience research. For example, in the field of optogenetics, ChRs are expressed in neurons in different areas of the animal brain and then fiber-optic cables are implanted in the brain to deliver light directly to the areas of the brain of interest. Turning on the light activates the neurons in these areas. However, these channels have broad activation spectra in the visible range and require high-intensity light for activation [˜1 mW mm2]. ChRs are naturally low-conductance channels requiring approximately 105-106functional ChRs expressed in the plasma-membrane of a neuron to produce sufficient light-dependent depolarization to induce neuronal activation. When applied to the mouse brain, ChRs require ˜1-15 mW light delivered ˜100 μm from the target cell population to reliably activate action potentials. This confines light-dependent activation to a small volume of brain tissue [˜1 mm3] in conjunction of the requirement of intracranial surgery for transgene injection and implantation of invasive fiber-optic cables. Therefore, this is a highly invasive method for neuronal control with light. There is a need for enabling optogenetics for large brain volumes without the need to implant invasive optical fibers for light delivery, for example, the light delivery in neuroscience applications. Novel and high-performance ChRs which can, for example, facilitate expansive optogenetics without the need for invasive implants, have been designed and produced, and are disclosed herein. For example, the engineered ChRs can have sufficient photocurrent strength and light sensitivity to enable minimally-invasive neuronal circuit interrogation in live organisms, and to avoid, for example, the need of intracranial surgery for transgene injection and implantation of invasive fiber-optic cables to produce light-dependent activation of brain and eye tissues. The high light sensitivity and ion conductance of the engineered ChRs allow these ChRs to be packaged and delivered non-invasively to desired locations and tissues using engineered viruses (e.g. rAAV-PHP.eB), and to be compatible with low per-cell transgene copy produced by systemic delivery (e.g., viral vector-based gene delivery intravenously). Coupled with non-invasive systemic delivery, the ChRs can be activated with light delivered through the skull of a live animal (by fiber optic-cables placed on the skull surface). In some embodiments, these high-conductance, high-sensitivity ChRs are used for robust light-dependent neuronal activation and light-dependent behavioral control. Recombinant or synthetic light-sensitive proteins are disclosed herein. The light-sensitive protein can, for example, comprise, or consist of, an amino acid sequence having at least 80% sequence identity to an amino acid sequence of any of the ChR proteins disclosed herein (e.g., ChR proteins having an amino acid sequence of any one of SEQ ID NOs: 1-475), including but is not limited to, an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-139, 141-147, and 149-196. In some embodiments, the light-sensitive protein comprises, or consists of, an amino acid sequence having, or having about, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 97.5%, 98%, 98.5%, 99%, 99.5%, 100%, or a range between any two of these values, sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-139, 141-147, and 149-196. In some embodiments, the light-sensitive protein comprises, or consists of, an amino acid sequence having at least, or at least about, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 97.5%, 98%, 98.5%, 99%, 99.5%, sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-139, 141-147, and 149-196. In some embodiments, the light-sensitive protein comprises or consists of an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-139, 141-147, and 149-196. In some embodiments, the amino acid sequence of the light-sensitive protein is selected from the group consisting of SEQ ID NOs: 5-139, 141-147, and 149-196. The light-sensitive protein can, for example, comprise, or consists of, an amino acid sequence having, or having about, one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, or a range between any two of these values, mismatch compared to an amino acid sequence of any of the ChR proteins disclosed herein (e.g., ChR proteins having an amino acid sequence of any one of SEQ ID NOs: 1-475), including but is not limited to, an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-139, 141-147, and 149-196. In some embodiments, the light-sensitive protein comprises, or consists of, an amino acid sequence having at most, or having at most about, one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty mismatches compared to an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-139, 141-147, and 149-196. In some embodiments, the recombinant or synthetic light-sensitive protein comprises, or consists of, an amino acid sequence having at least 95% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 93, 109, 125-130, 132, 133, 136-138, 142, 146, 149, 150, and 155-196. In some embodiments, the recombinant or synthetic light-sensitive protein comprises, or consists of, an amino acid sequence selected from the group consisting of SEQ ID NOs: 93, 109, 125-130, 132, 133, 136-138, 142, 146, 149, 150, and 155-196. In some embodiments, the recombinant or synthetic light-sensitive protein comprises, or consists of, an amino acid sequence having at most, or having at most about, one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty mismatches compared to SEQ ID NOs: 93, 109, 125-130, 132, 133, 136-138, 142, 146, 149, 150, and 155-196. In some embodiments, the recombinant or synthetic light-sensitive protein comprises, or consists of, an amino acid sequence having, or having about, one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, or a range between any two of these values, mismatches compared to SEQ ID NOs: 93, 109, 125-130, 132, 133, 136-138, 142, 146, 149, 150, and 155-196. In some embodiments, the recombinant or synthetic light-sensitive protein is a ChR comprising, or consisting of, an amino acid sequence having at least 95% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 178-196. In some embodiments, the recombinant or synthetic light-sensitive protein is a ChR comprising, or consisting of, an amino acid sequence selected from the group consisting of SEQ ID NOs: 178-196. In some embodiments, the recombinant or synthetic light-sensitive protein comprises, or consists of, an amino acid sequence having, or having about, one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, or a range between any two of these values, mismatches compared to SEQ ID NOs: 178-196. In some embodiments, the light-sensitivity protein does not comprise, or is not consisted of, an amino acid sequence selected from SEQ ID NOs: 140, 148, 170, 173, 191, and 194. In some embodiments, the light-sensitivity protein does not comprise, or is not consisted of, an amino acid sequence having at least, or at least about, 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.5%, or more, sequence identity to SEQ ID NOs: 140, 148, 170, 173, 191, and 194. In some embodiments, the light-sensitivity protein does not comprise, or is not consisted of, an amino acid sequence having 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.5%, or a range of any two of these values, sequence identity to SEQ ID NOs: 140, 148, 170, 173, 191, and 194. The recombinant or synthetic light-sensitive protein can be better in one or more functional properties (e.g., higher light sensitivity, photocurrent strength, ion conductance, light-induced firing, plasma membrane localization, and spectra properties) than a reference ChR. The reference ChR can be, for example, C1C2, CsChrim, CheRiff, and/or any one of the ChRs having the amino acid sequence of SEQ ID NO: 1, 3, 4, 155, 156, 176, or 177. The extent of which the recombinant or synthetic light-sensitive protein is higher in one or more of light sensitivity, ion conductance and photocurrent strength, as compared to the reference ChR can vary. For example, the recombinant or synthetic light-sensitive protein can have at least, or at least about, 1.05, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 4, 5, 6, 7, 8, 9, 10-fold improvement in one or more of light sensitivity, ion conductance and photocurrent strength, as compared to the reference ChR. In some embodiments, the recombinant or synthetic light-sensitive protein can have 1.05, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 4, 5, 6, 7, 8, 9, 10, or a range between any two of these values, -fold improvement in one or more of light sensitivity, ion conductance and photocurrent strength, as compared to the reference ChR. The light sensitivity of the recombinant or synthetic light-sensitive protein can be, for example, 1.05, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 4, 5, 6, 7, 8, 9, 10, or a range between any two of these values, times higher compared to the reference ChR. In some embodiments, the photocurrent strength of the recombinant or synthetic light-sensitive protein is, or is about, 1.05, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 4, 5, 6, 7, 8, 9, 10, or a range between any two of these values, times higher compared to the reference ChR. In some embodiments, the ion conductance of the recombinant or synthetic light-sensitive protein is, or is about, 1.05, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 4, 5, 6, 7, 8, 9, 10, or a range between any two of these values, times higher compared to the reference ChR. The measurement of light sensitivity, ion conductance, and photocurrent strength of the light-sensitive protein can be performed by techniques known in the art, for example patch-clamp electrophysiology. In some embodiments, the light-sensitive protein disclosed herein is a blue-shifted ChR. In some embodiments, the light-sensitive protein disclosed herein is a red-shifted ChR. Functional properties of various non-limiting examples of ChR proteins are provided in Table 1. Amino acid sequences of each of the ChR described in Table 1 are provided in the Sequence Listing submitted herewith. The parent ChRs used in the Examples described herein for designing and generating the engineered ChRs are italicized, and the three engineered ChRs with top light sensitivity are bold in Table 1. TABLE 1Functional properties of non-limiting examples of ChR proteinscyan_green_red_cyan_SEQpeakpeakpeakssgreen_red_Kinetics_green_max_peakmax_ssIDChR name(nA)(nA)(nA)(nA)ss (nA)ss (nA)offblock_IDmnorm(nA)(nA)NO.C1C20.660.160.010.450.14028c111111111100.240.660.451c10.140.010.010.08007c1000000000320.080.140.082CsChrim0.830.980.770.690.770.4251c2222222222010.980.773CheRiff0.660.060.010.460.05016c000000000000.10.660.464c620.010.010.01000c2222222022100.0105c640.480.70.410.420.60.2761c22222222201710.70.66c660.070.080.040.060.070.031528c22122222222510.080.077c670.040.040.020.030.020.0188c2221222222100.80.040.038c71.260.440.021.010.420.01822c1111101111210.351.261.019c700.450.590.530.410.510.3130c22222212221010.590.5110c710.050.180.050.040.140.038c2222222122810.180.1411c730.460.750.360.420.660.3182c2222222221710.750.6612c960.090.130.010.020.10c21021212007710.130.113n10.160.010.010.120020n0000001000400.070.160.1214n101.790.820.011.230.810152n1111111101110.461.791.2315n111.230.260.010.750.25080n111111111080.211.230.7516c110.340.090.010.250.08023c1111111110200.250.340.2517c610.340.560.330.310.480.3184c22222202221710.560.4818c120.410.120.010.290.11027c121111111170.290.410.2919c60.380.060.010.280.05020c111101111150.160.380.2820n830.130.040.010.120.0305719n1100212201840.310.130.1221c391.650.190.011.050.17043c000010000050.111.651.0522c480.20.020.010.170.01015c0002000000100.090.20.1723c370.40.090.010.260.08021c0010000000230.240.40.2624c490.20.030.010.090.020112c0000200000100.150.20.0925c30.070.020.010.060.02042c1011111111100.350.070.0626c50.120.020.010.060.02014c111011111190.190.120.0627n130.010.010.01000n1211111111330.01028c280.020.010.02000c1222121000850.02029c210.230.110.050.190.10.03337c1112221221700.50.230.1930c200.760.160.010.490.15029c111111111270.210.760.4931c20.230.460.130.190.350.0723c12222222222610.460.3532c521.130.30.010.860.280135c0000000200100.271.130.8633c560.490.510.430.430.470.3263c20222222221010.510.4734c160.050.030.030.030.010.01504c1111121111280.560.050.0335c580.070.060.020.060.050.0188c2220222222100.880.070.0636c500.240.150.010.220.14097c0000020000250.640.240.2237c420.680.020.010.450.01013c000000010060.030.680.4538n140.020.020.020.010.010n1121111111180.730.020.0139n170.020.010.01000n1111121111110.02040n520.030.010.010.020.010772n0000000200110.460.030.0241n570.040.020.010.030.010.013038n2202222222150.630.040.0342n580.10.090.060.080.080.0458n222022222240.980.10.0843n590.240.40.240.210.330.1421n22220222222410.40.3344n60.270.040.010.150.03015n111011111130.140.270.1545n600.170.290.170.150.270.1551n22222022221410.290.2746n630.20.150.060.180.140.05316n222222220290.730.20.1847n640.110.250.070.10.190.0337n22222222201010.250.1948n650.280.550.320.260.470.2243n1222222222610.550.4749n680.260.230.180.210.190.0820n222122222220.920.260.2150n690.20.270.210.170.220.1328n22221222221010.270.2251n70.090.020.010.070.0108n1111011111250.190.090.0752n700.20.310.240.180.270.1432n22222122221110.310.2753n720.040.030.010.030.010286n2222222212160.640.040.0354n730.120.210.090.110.20.0449n2222222221610.210.255n510.650.040.010.530.03017n0000002000320.060.650.5356n150.330.060.010.220.05033n111211111120.190.330.2257n50.140.020.010.080.01032n1101111111190.140.140.0858n481.190.170.010.960.16074n000200000040.141.190.9659n180.140.040.010.130.0305125n1111111211100.280.140.1360n190.160.070.020.10.050.0157n1111111121160.420.160.161n20.060.060.020.050.040.0146n2222221222330.970.060.0562n200.670.370.010.60.3501135n111111111260.550.670.663n210.050.040.020.040.020576n2212111212570.730.050.0464n240.010.010.01000n2112001222820.01065n250.180.090.010.170.0801413n1022011211740.490.180.1766n380.260.050.010.170.04012n0010000000190.180.260.1767n360.470.030.010.380.0109n100000000050.050.470.3868n40.960.410.010.720.38053n1011111111190.430.960.7269n401.180.160.010.730.14073n0000100000250.131.180.7370n411.010.250.010.890.240182n0000010000170.241.010.8971n420.610.050.010.20.04031n000000010040.080.610.272n430.090.010.010.050013n0000000010110.150.090.0573n450.80.050.010.50.04016n200000000070.070.80.574n391.030.370.010.930.35071n000100000030.361.030.9375c411.50.230.011.140.220171c0000001000170.151.51.1476n4_20.020.010.010.0100n0211111212540.760.020.0177c4_20.260.330.310.240.30.226c22222212211710.330.378n5_20.030.020.010.020.010520n2211112212570.690.030.0279n8_20.240.70.220.210.480.1618n12221222221610.70.4880c1_20.220.320.180.210.30.142c22222212202710.320.381c3_40.020.020.0200.010c10221201208410.020.0182n7_40.010.010.01000n0221012201590.01083c2_40.020.020.020.010.010.011186c2121021100780.910.020.0184c18_40.010.010.01000c2202121120620.01085c15_40.120.050.010.090.040103c2012000200680.430.120.0986c14_40.330.230.020.230.210.01268c1012021002740.690.330.2387c11_40.020.020.010.010.010c2011221220720.020.0188n20_40.020.010.020.010.0101270n2201022212570.80.020.0189n19_40.320.230.020.290.220.01886n1010012220930.710.320.2990n12_40.010.010.010.0100n0210102212690.010.0191ChR_9_42.422.120.051.931.90.04356c1210001101500.872.421.9392[ChRger1]c8_40.170.190.040.080.160.03c20201212015910.190.1693n34_50.050.030.010.040.020420n1000020220390.70.050.0494n28_50.010.010.01000n1220212201360.01095c38_50.020.010.01000c2122220022340.02096c31_50.020.010.01000c2020222121350.02097c21_50.010.010.01000c0201200100350.01098n4_70.130.10.020.120.080.012400n1211122202450.760.130.1299c2_70.010.020.0100.01014c22222201204210.020.01100n1_70.10.070.030.090.060.023788n1211022222500.740.10.09101c3_70.470.690.560.420.60.3525c11222222223310.690.6102ChR_19_90.290.060.010.230.050230c1202001100750.220.290.23103ChR_1_90.180.130.010.150.110627n1012012202980.710.180.15104ChR_15_90.080.070.040.070.060.032386n1010112201760.890.080.07105ChR_21_90.210.120.010.190.110683n1000012212860.580.210.19106ChR_23_90.090.050.010.070.0401117n1002002221690.560.090.07107ChR_24_90.020.020.010.010.010n2220112200440.020.01108ChR-25-92.540.230.011.750.220145c2000001100480.092.541.75109[ChRger2]ChR_26_90.040.030.020.020.010452n1210112220590.740.040.02110ChR_11_90.560.170.010.420.1602522n1000011002720.310.560.42111ChR_10_90.150.030.010.140.0203440n2002012201790.230.150.14112ChR_17_90.10.080.020.090.060.01116n1020002202730.820.10.09113ChR_5_90.180.140.040.140.120.031412c2112221100830.740.180.14114ChR_28_90.270.160.020.20.150.0196c2012021100960.60.270.2115ChR_32_90.010.010.01000n1202112201570.010116ChR_7_90.890.270.010.570.240294c1012101100690.310.890.57117ChR_38_90.020.020.01000n1220022201490.020118ChR_34_91.230.460.010.920.440269c1112001101440.371.230.92119ChR_30_91.310.660.020.820.620456c1211001100610.511.310.82120ChR_4_90.010.010.01000c2002221120660.010121ChR_3_90.040.030.020.020.01066n1021012220880.780.040.02122ChR_6_90.010.030.01000n12120122007810.030123ChR_29_90.010n1221102221380.010124ChR_12_103.321.80.032.261.610.02179c1111001101340.543.322.26125ChR_13_100.770.30.010.640.250n1111111002210.390.770.64126ChR_18_102.561.440.041.821.260.03390c1211001101410.562.561.82127ChR_14_102.111.390.031.471.240.02607n1111111100190.662.111.47128ChR_10_1021.030.021.330.950.01229n1011111100380.5221.33129ChR_15_101.610.970.021.350.870.013544n1111111102170.61.611.35130ChR_7_100.310.030.010.250.0206723n1002011002760.10.310.25131ChR_5_101.420.150.0110.13038n1001002000400.11.421132ChR_16_102.631.060.021.7310.01323n1112111100210.42.631.73133ChR_17_100.270.50.340.260.460.2529c11222212224310.50.46134ChR_4_100.710.060.010.520.050170n1000012000540.090.710.52135ChR-11-103.471.640.022.191.390.01311c1110001101430.473.472.19136[ChRger3]ChR_9_102.561.280.021.921.170.01940n1011111000420.52.561.92137ChR_3_100.720.030.010.490.02010n1000002000370.040.720.49138ChR_1_100.460.030.010.310.02013c1000000100380.060.460.31139ChR_29_100.030.020.010.010.010216c1210021101570.970.030.01140ChR_20_100.210.410.270.190.380.1730c12222212223610.410.38141ChR_21_100.850.030.010.560.02011c2000000100310.040.850.56142ChR_22_100.40.040.010.280.02014c2002000100410.10.40.28143ChR_23_100.140.240.140.120.220.0956c20222212222010.240.22144ChR_24_100.190.330.170.180.310.1262c20222222202710.330.31145ChR_6_100.460.030.010.350.0208491n1001011002750.060.460.35146ChR_26_100.210.330.220.180.280.1219c21222212221710.330.28147ChR_30_100.020.030.02000c12100011216010.030148ChR_2_102.120.330.011.480.310146c1000001100550.152.121.48149ChR_8_102.41.030.021.590.940.01217c1010001101530.432.41.59150ChR_27_100.410.650.410.320.470.1620c21222222202410.650.47151ChR_28_100.821.220.820.690.960.4228c21222222211411.220.96152ChR_19_100.230.550.290.210.420.2213n12211222221810.550.42153ChR_25_100.290.430.290.260.380.1733c20222222211710.430.38154 The light-sensitive protein disclosed herein can comprise a signal peptide sequence, for example natural ChR signal peptides (e.g., those described in Klapoetke et al., Nature Methods 11:338-346, 2014) or any signal peptide sequences known to be able to target proteins to the plasma cell membrane. Non-limiting examples of natural ChR signal peptides include MSRRPWLLALALAVALAAGSAGA (SEQ ID NO: 197) and MSRLVAASWLLALLLCGITSTTTAS (SEQ ID NO: 198). In some embodiments, the signal peptide in the light-sensitive protein comprises an amino acid sequence having, or having about, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, or a range between any two of these values, sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO: 197 or SEQ ID NO: 198. In some embodiments, the signal peptide in the light-sensitive protein comprises an amino acid sequence having at least, or having at least about, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO: 197 or SEQ ID NO: 198. In some embodiments, the recombinant or synthetic light-sensitive protein is a mature protein. In some embodiments, the recombinant or synthetic light-sensitive protein does not comprise any signal peptide. The recombinant or synthetic light-sensitive protein can comprise one or more insertions (e.g., a synthetic tag at the 5′-terminal region of the protein). In some embodiments, the recombinant or synthetic light-sensitive protein does not comprise any insertions, e.g., tags. The light-sensitive protein (e.g., ChR) can vary in length. For example, the light-sensitive protein can be, or be about, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, or a range between any two of these values, amino acids in length. In some embodiments, the light-sensitive protein is at least, or at least about, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550 amino acids in length. The light-sensitive protein can, for example, comprises, consists of, or consists essentially of, any one or more of the sequences shown in Table 2 which provides non-limiting examples of the ChRs disclosed herein. TABLE 2Sequences of non-limiting examples of ChR proteins (the sequence ofa synthetic tag (SpyTag)is bold and underlined)SEQIDNO.ChR NameAmino acid sequence1C1C2MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHLSNLIGLANDYNKRTMGLLVSDIGTIVWGITAALSKGYVRVIFFLMGLCYGIYIFFNAAKVYIEAYHTVPKGRCRQVVIGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTIKLNIGGTEIEVETLVEDEAEAGAV3CsChrimMSRLVAASWLLALLLCGITSITTASAHIVMVDAYKPTKSAPAASSIDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGTPGEKIGAQVCQWIAFSIAIALLIFYGFSAWKATCGWEEVYVCCVEVLFVTLEIFKEFSSPATVYLSIGNHAYCLRYFEWLLSCPVILIRLSNLSGLKNDYSKRTMGLIVSCVGMIVFGMAAGLATDWLKWLLYIVSCIYGGYMYFQAAKCYVEANHSVPKGHCRMVVKLMAYAYFASWGSYPILWAVGPEGLLKLSPYANSIGHSICDIIAKEFWTFLAHHLRIKIHEHILIHGDIRKTIKMEIGGEEVEVEEFVEEEDEDTV4CheRiffMGGAPAPDAHSAPPGNDSAAHIVMVDAYKPIKGGSEYHAPAGYQVNPPYHPVHGYEEQCSSIYIYYGALWEQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWLMFPGLFIFGPEGMHTLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV92ChR_9_4MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQC[ChRger1]FCLAWLKSNGTPGEKIGAQVCQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVIGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTIKLNIGGTEIEVETLVEDEAEAGAV109ChR_25_9MSRLVAASWLLALLLCGITSITTASAHIVMVDAYKPTKSAPAASSIDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYC[ChRger2]SAWLHSRGALWEQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVIGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV125ChR_12_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVIGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTIKLNIGGTEIEVETLVEDEAEAGAV126ChR_13_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLIGLANDYNKRTMGLLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVEAYHTVPKGRCRQVVIGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTIKLNIGGTEIEVETLVEDEAEAGAV127ChR_18_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGEKIGAQVCQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV128ChR_14_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMALLVSDIGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV129ChR_10_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFAFFLSALFLAFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMALLVSDIGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV130ChR_15_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV132ChR_5_10MSRLVAASWLLALLLCGITSTTTASAHIVMVDAYKPTKSAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPATVYLSGGNHAYWLRYAEWLLTCPVILIHLSNITGLANDYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWLMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV133ChR_16_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLKNDYSKRTMALLVSDIGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV136ChR_11_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQC[ChRger3]FCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV137ChR_9_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFAFFLSALFLAFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV138ChR_3_10MSRLVAASWLLALLLCGITSTTTASAHIVMVDAYKPTKSAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPATVYLSGGNHAYWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWLMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV140ChR_29_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVEHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGEKIGAQVCQWITEALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSCVGMIVEGMAAGLATDWLKWLLYIVSCIYGGYMYFQAAKCYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV142ChR_21_10MSRLVAASWLLALLLCGITSTTTASAHIVMVDAYKPTKSAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV146ChR_6_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVEHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNITGLANDYNKRTMGLLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVESYYIMPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHYLRVLIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV148ChR_30_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVEHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGEKIGAQVCQWITEALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSPYANSIGHSICDIIAKEFWTFLAHHLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV149ChR_2_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVEHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV150ChR_8_10MSRRPWLLALALAVALAAGSAGAAHIVMVDAYKPTKSTGSDATVPVATQDGPDYVEHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFTEALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV155C1C2MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAE(withoutKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFFLMGLCYGIYTFFNAAKVYIEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV156CsChrimMSRLVAASWLLALLLCGITSTTTASAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGTPGEKI(withoutGAQVCQWIAFSIAIALLTFYGFSAWKATCGWEEVYVCCVEVLFVTLEIFKEFSSPATVYLSTGNHAYCLRYFEWLLSCPVILIRLSSpyTag)NLSGLKNDYSKRTMGLIVSCVGMIVFGMAAGLATDWLKWLLYIVSCIYGGYMYFQAAKCYVEANHSVPKGHCRMVVKLMAYAYFASWGSYPILWAVGPEGLLKLSPYANSIGHSICDIIAKEFWTFLAHHLRIKIHEHILIHGDIRKTTKMEIGGEEVEVEEFVEEEDEDTV157ChR_9_4MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGE[ChRger1]KIGAQVCQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIH(withoutLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFSpyTag)VSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV158ChR_25_9MSRLVAASWLLALLLCGITSTTTASAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQE[ChRger2]TARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLS(withoutNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSSpyTag)WGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV159ChR_12_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAE(withoutKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV160ChR_13_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAE(withoutKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNLTGLANDYNKRTMGLLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV161ChR_18_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGE(withoutKIGAQVCQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV162ChR_14_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAE(withoutKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNLTGLANDYNKRTMALLVSDIGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV163ChR_10_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWE(withoutQETARGFQWFAFFLSALFLAFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNLTGLANDYNKRTMALLVSDIGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV164ChR_15_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAE(withoutKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV165ChR_5_10MSRLVAASWLLALLLCGITSTTTASSAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQ(withoutETARGFQWFAVFLSALFLAFYGWHATKASVGWEEVYVCSVELIKVILEIYFEFTSPATVYLSGGNHAYWLRYAEWLLTCPVILIHLSpyTag)SNITGLANDYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWLMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV166ChR_16_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAE(withoutKLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNLTGLKNDYSKRTMALLVSDIGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV167ChR_11_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAE[ChRger3+KLAANILQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIH(withoutLSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFSpyTag)VSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV168ChR_9_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWE(withoutQETARGFQWFAFFLSALFLAFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNLTGLANDYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV169ChR_3_10MSRLVAASWLLALLLCGITSTTTASAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQE(withoutTARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPATVYLSGGNHAYWLRYAEWLLTCPVILIHLSSpyTag)NITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWLMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV170ChR_29_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGE(withoutKIGAQVCQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHSpyTag)LSNITGLSEAYNKRTMALLVSCVGMIVFGMAAGLATDWLKWLLYIVSCIYGGYMYFQAAKCYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV171ChR_21_10MSRLVAASWLLALLLCGITSTTTASAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQE(withoutTARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSSpyTag)NITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV172ChR_6_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWE(withoutQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHSpyTag)LSNITGLANDYNKRTMGLLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVESYYIMPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHYLRVLIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV173ChR_30_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGE(withoutKIGAQVCQWITFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHSpyTag)LSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSPYANSIGHSICDIIAKEFWTFLAHHLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV174ChR_2_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWE(withoutQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHSpyTag)LSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV175ChR_8_10MSRRPWLLALALAVALAAGSAGATGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWE(withoutQETARGFQWFTFALSALCLMFYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHSpyTag)LSNITGLSEAYNKRTMALLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV176matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGC1C2YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDI(withoutGTIVWGTTAALSKGYVRVIFFLMGLCYGIYTFFNAAKVYIEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYSpyTag)GSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV177matureAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGTPGEKIGAQVCQWIAFSIAIALLTFYGFSAWCsChrimKATCGWEEVYVCCVEVLFVTLEIFKEFSSPATVYLSTGNHAYCLRYFEWLLSCPVILIRLSNLSGLKNDYSKRTMGLIVSCVGMIV(withoutFGMAAGLATDWLKWLLYIVSCIYGGYMYFQAAKCYVEANHSVPKGHCRMVVKLMAYAYFASWGSYPILWAVGPEGLLKLSPYANSISpyTag)GHSICDIIAKEFWTFLAHHLRIKIHEHILIHGDIRKTTKMEIGGEEVEVEEFVEEEDEDTV178matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGEKIGAQVCQWITFALSALCLMFYGChR_9_4YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDL[ChRger1]GTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVA(withoutGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVSpyTag)179matureAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQETARGFQWFAVFLSALFLAFYGWHAYChR_25_9KASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTIC[ChRger2]MGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVAGSTI(withoutGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKVSpyTag)180matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGChR_12_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDL(withoutGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVASpyTag)GSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV181matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGChR_13_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDL(withoutGTICMGVTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYSpyTag)GSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV182matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGEKIGAQVCQWITFALSALCLMFYGChR_18_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDL(withoutGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVASpyTag)GSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV183matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGChR_14_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMALLVSDI(withoutGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYSpyTag)GSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV184matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFAFFLSALFLAFYGChR_10_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMALLVSDI(withoutGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVASpyTag)GSTIGHTIADLLSKNIWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV185matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGChR_15_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDI(withoutGTIVWGTTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYSpyTag)GSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV186matureAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQETARGFQWFAVFLSALFLAFYGWHAYChR_5_10KASVGWEEVYVCSVELIKVILEIYFEFTSPATVYLSGGNHAYWLRYAEWLLTCPVILIHLSNITGLANDYNKRTMALLVSDLGTIC(withoutMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWLMFPILFILGPEGFGVLSVAGSTISpyTag)GHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV187matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGChR_16_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLKNDYSKRTMALLVSDI(withoutGTIVWGTTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYSpyTag)GSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV188matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGChR_11_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDL[ChRger3]GTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVA(withoutGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVSpyTag)189matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFAFFLSALFLAFYGChR_9_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMALLVSDL(withoutGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVASpyTag)GSTIGHTIADLLSKNIWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV190matureAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQETARGFQWFAVFLSALFLAFYGWHAYChR_3_10KASVGWEEVYVCSVELIKVILEIYFEFTSPATVYLSGGNHAYWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTIC(withoutMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWLMFPILFILGPEGFGVLSVAGSTISpyTag)GHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV191matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGEKIGAQVCQWITFALSALCLMFYGChR_29_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSCV(withoutGMIVFGMAAGLATDWLKWLLYIVSCIYGGYMYFQAAKCYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVASpyTag)GSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV192matureAPAASSTDGTAAAAVSHYAMNGFDELAKGAVVPEDHFVCGPADKCYCSAWLHSRGALWEQETARGFQWFAVFLSALFLAFYGWHAYChR_21_10KASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDLGTIC(withoutMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGGCKKLVLAMTAVYYSSWGMFPILFILGPEGFGVLSVAGSTISpyTag)GHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV193matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFAVFLSALFLAFYGChR_6_10WHAYKASVGWEEVYVCSVELIKVILEIYFEFTEPAVIYSSGGNKTVWLRYAEWLLTCPVILIHLSNITGLANDYNKRTMGLLVSDL(withoutGTICMGVTAALATGWVKWLFYCIGLVYGGYMYFQAAKCYVESYYIMPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVASpyTag)GSTIGHTIADLLSKNIWGLLGHYLRVLIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV194matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTPGEKIGAQVCQWITFALSALCLMFYGChR_30_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDL(withoutGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSPYSpyTag)ANSIGHSICDIIAKEFWTFLAHHLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV195matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFAVFLSALFLAFYGChR_2_10WHAYKASVGWEEVYVCSVELIKVILEIYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDL(withoutGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVASpyTag)GSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV196matureTGSDATVPVATQDGPDYVFHRAHERMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGALWEQETARGFQWFTFALSALCLMFYGChR_8_10YQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEAYNKRTMALLVSDL(withoutGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVASpyTag)GSTIGHTIADLLSKNIWGLLGHFLRIKIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV Also disclosed herein are nucleic acid molecules comprising the nucleotide sequences that encode one or more of the light-sensitive proteins described herein. The nucleic acid molecule can be a recombinant expression vector, for example, a viral vector. Examples of viral vector include, but are not limited to, adeno-associated viral vectors, lentiviral vectors, herpes simplex virus vectors, and retroviral vectors. In the nucleic acid molecule, the nucleotide sequence encoding the one or more light-sensitive proteins is operably linked to a transcriptional control element. It can be advantageous in some embodiments that the transcriptional control element is functional in a photoreceptor cell. The photoreceptor cell can be a rod cell, a cone cell, a retinal cell, or a combination thereof. The transcriptional control element can be, for example, promoter (e.g., a retinal cell-specific promoter). Non-limiting examples of the promoter include synapsin promoter, a CAG promoter, a cytomegalovirus promoter (CMV) promoter, a grm6 promoter, a Pleiades promoter, a ChAT promoter, a V-glut promoter, a GAD promoter, a PV promoter, a somatostatin (SST) promoter, a neuropeptide Y (NPY) promoter, a VIP promoter, a red cone opsin promoter, rhodopsin promoter, a rhodopsin kinase promoter, vitelliform macular dystrophy 2 (VMD2) gene promoter, an interphotoreceptor retinoid-binding protein (IRBP) gene promoter, elongation factor-1 alpha (EF-1 alpha) promoter, and a combination thereof. Disclosed herein includes methods and compositions for expressing one or more of the light-sensitive proteins (e.g., ChRs) disclosed herein in cells, tissues, organs, and/or subjects, where the ChR(s) can be activated by contact with one or more pulses of light, which results in strong depolarization of the cells or the cells in the tissues, organs and/or subjects. In some embodiments, the expression of the ChR(s) is used to control cells, tissues, organs, and/or subjects in vivo, ex vivo, and/or in vitro in response to pulses of light of a suitable wavelength. A cell, comprising (a) a recombinant or synthetic light-sensitive protein, (b) a nucleic acid molecule comprising a coding sequence of a light-sensitive protein, or both, is provided. The cell can be, for example, a mammalian cell or non-mammalian cell. In some embodiments, the cell is a rod cell, a cone cell, or a retina cell. The cell can be a neuronal cell, an electrically active cell, or both. In some embodiments, the cell is a recombinant host cell, for example, a mammalian cell, a bacterial cell, a yeast cell, an insect cell, a plant cell, or a combination thereof. Some embodiments provided a composition comprising one or more of (a) the cell, (b) the recombinant or synthetic light-sensitive protein, and (c) the nucleic acid molecule comprising the coding sequence of the light-sensitive protein. The composition can be, for example, a pharmaceutical composition comprising one or more pharmaceutically acceptable excipient. Compositions for Delivering Light-Sensitive Proteins to a Subject Various systems and methods are known in the art for delivering nucleic acid molecules into a cell, a tissue, an organ, and/or a subject. The delivery can be, for example, target-specific, tissue-specific, cell type specific, organ specific, nonspecific, and/or systematic. In some embodiments, the nucleic acid molecule comprises a coding sequence for one or more proteins, and the delivery is used for expressing the one or more proteins encoded by the nucleic acid molecule in the target cell, tissue, organ, and/or subject. Disclosed herein include a nucleic acid molecule (e.g., an expression vector) comprising a coding sequence for the light-sensitive protein (e.g., one or more of the ChRs disclosed herein) for use in treating or ameliorating blindness, restoring or enhancing vision and photosensitivity, treating or ameliorating vision loss in a subject. In some embodiments, the method comprises delivering (e.g., injecting) the nucleic acid molecule into the LGN of the subject. The expression of the light-sensitive protein can be controlled by a transcription regulatory element, for example a promoter selected from the group of Human elongation factor-1 alpha (EF-1 alpha), Human cytomegalovirus promoter (CMV) or CAG promoter. Also disclosed include a composition, for example a pharmaceutical composition, comprising the nucleic acid molecule (e.g., a vector) comprising the coding sequence for the light-sensitive protein. The nucleic acid molecule can be any of the nucleic acid molecule encoding the light-sensitive protein and disclosed herein, Many different viral and non-viral vectors and methods of their delivery, for use in gene delivery (including gene therapy), are known, including adenovirus vectors, adeno-associated virus (AAV) vectors, retrovirus vectors, lentiviral vectors, herpes virus vectors, liposomes, poxviruses, naked DNA administration, plasmids, cosmids, phages, encapsulated cell technology, and the like. A detailed review of possible techniques for transforming genes into desired cells of the eye is taught by Wright (Br J Ophthalmol, 1997; 81: 620-622). The vectors can be used to deliver one or more of the light-sensitive proteins (e.g., ChRs) disclosed herein or the coding sequences for the one or more of the proteins to a subject in need thereof. Expression of the light-sensitive proteins disclosed herein can be controlled by, for example, a cell specific promoter to allow expression occurred only in a specific cell type (e.g., retinal cells). Adeno-associated virus (AAV) is a replication-deficient parvovirus, the single-stranded DNA genome of which is about 4.7 kb in length including 145 nucleotides inverted terminal repeat (ITRs). The ITRs play a role in integration of the AAV DNA into the host cell genome. When AAV infects a host cell, the viral genome integrates into the host's chromosome resulting in latent infection of the cell. In a natural system, a helper virus (for example, adenovirus or herpesvirus) provides genes that allow for production of AAV virus in the infected cell. In the case of adenovirus, genes E1A, E1B, E2A, E4 and VA provide helper functions. Upon infection with a helper virus, the AAV provirus is rescued and amplified, and both AAV and adenovirus are produced. In the instances of recombinant AAV vectors having no Rep and/or Cap genes, the AAV can be non-integrating. AAV vectors that comprise coding regions of one or more light-sensitive proteins (e.g., the ChRs disclosed herein) are provided. The AAV vector can include a 5′ inverted terminal repeat (ITR) of AAV, a 3′ AAV ITR, a promoter, and a restriction site downstream of the promoter to allow insertion of a polynucleotide encoding one or more light-sensitive proteins, wherein the promoter and the restriction site are located downstream of the 5′ AAV ITR and upstream of the 3′ AAV ITR. In some embodiments, the AAV vector includes a posttranscriptional regulatory element downstream of the restriction site and upstream of the 3′ AAV ITR. In some embodiments, the AAV vectors disclosed herein can be used as AAV transfer vectors carrying a transgene encoding a light-sensitive protein for producing recombinant AAV viruses that can express the light-sensitive protein in a cell. Generation of the viral vector can be accomplished using any suitable genetic engineering techniques well known in the art, including, without limitation, the standard techniques of restriction endonuclease digestion, ligation, transformation, plasmid purification, and DNA sequencing, for example as described in Sambrook et al. (Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press, N.Y. (1989)). The viral vector can incorporate sequences from the genome of any known organism. The sequences can be incorporated in their native form or can be modified in any way to obtain a desired activity. For example, the sequences can comprise insertions, deletions or substitutions. In some embodiments, the viral vectors can include additional sequences that make the vectors suitable for replication and integration in eukaryotes. In other embodiments, the viral vectors disclosed herein can include a shuttle element that makes the vectors suitable for replication and integration in both prokaryotes and eukaryotes. In some embodiments, the viral vectors can include additional transcription and translation initiation sequences, such as promoters and enhancers; and additional transcription and translation terminators, such as polyadenylation signals. Various regulatory elements that can be included in an AAV vector have been described in detail in US Patent Publication 2012/0232133 which is hereby incorporated by reference in its entirety. Pharmaceutical Compositions and Methods of Administration Light-sensitive proteins (e.g., ChRs) with desirable properties are provided herein. Disclosed herein include cells, tissues, organs, and subjects that comprises one or more of the light-sensitive proteins, one or more of the nucleic acid molecules (e.g., vectors) comprising coding sequence(s) for the light-sensitive protein(s). Also disclosed include pharmaceutical compositions comprising one or more of the light-sensitive proteins, one or more of the nucleic acid molecules (e.g., vectors) comprising coding sequence(s) for the light-sensitive proteins, and/or one or more of the cells comprising the light-sensitive protein(s) disclosed herein, and one or more pharmaceutically acceptable carriers. The compositions can also comprise additional ingredients such as diluents, stabilizers, excipients, and adjuvants. As used herein, “pharmaceutically acceptable” carriers, excipients, diluents, adjuvants, or stabilizers are the ones nontoxic to the cell or subject being exposed thereto (preferably inert) at the dosages and concentrations employed or that have an acceptable level of toxicity as determined by the skilled practitioners. The carriers, diluents and adjuvants can include buffers such as phosphate, citrate, or other organic acids; antioxidants such as ascorbic acid; low molecular weight polypeptides (e.g., less than about 10 residues); proteins such as serum albumin, gelatin or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as Tween™, Pluronics™ or polyethylene glycol (PEG). In some embodiments, the physiologically acceptable carrier is an aqueous pH buffered solution. In some embodiments, the pharmaceutical composition comprises a non-viral vector or a viral (e.g., AAV) vector comprising a coding sequence of any one of the light-sensitive proteins described herein. Titers of the viral vector to be administered will vary depending, for example, on the particular viral vector, the mode of administration, the treatment goal, the individual, and the cell type(s) being targeted, and can be determined by methods standard in the art. As will be readily apparent to one of skill in the art, the useful in vivo dosage of the recombinant virus to be administered and the particular mode of administration will vary depending upon the age, weight, the severity of the affliction, and animal species treated, the particular recombinant virus expressing the protein of interest that is used, and the specific use for which the recombinant virus is employed. The determination of effective dosage levels, that is the dosage levels necessary to achieve the desired result, can be accomplished by one skilled in the art using routine pharmacological methods. Typically, human clinical applications of products are commenced at lower dosage levels, with dosage level being increased until the desired effect is achieved. Alternatively, acceptable in vitro studies can be used to establish useful doses and routes of administration of the compositions identified by the present methods using established pharmacological methods. Although the exact dosage will be determined on a drug-by-drug basis, in most cases, some generalizations regarding the dosage can be made. In some embodiments, the viral vector for delivery a nucleic acid to a subject (e.g., systematic delivery, or delivery to the eye or brain tissue of the subject) can be administered, for example via injection, to a subject at a dose of between 1×1010genome copies (GC) of the recombinant virus per kg of the subject and 2×1014GC per kg, for example between 5×1011GC/kg and 5×1012GC/kg. In some embodiments, the dose of the viral vector (e.g., AAV vectors) administered to the subject is no more than 2×1014GC per kg. In some embodiments, the dose of the viral vector administered to the subject is no more than 5×1012GC per kg. In some embodiments, the dose of the viral vector administered to the subject is no more than 5×1011GC per kg. The nucleic acid molecule, for example, a vector (e.g., a viral vector)) comprising a nucleotide sequence encoding the light-sensitive protein can be administered to a subject (e.g., a human) in need thereof. The route of the administration is not particularly limited. For example, a therapeutically effective amount of the nucleic acid molecule can be administered to the subject by via routes standard in the art. Non-limiting examples of the route include intravitreal, intravenous, intraocular, or subretinal administration (e.g., intravitreal, intravenous, intraocular, or subretinal injection), depending on the retinal layer being targeted. In some embodiments, the nucleic acid molecule is administered to the subject by systematic transduction. In some embodiments, the nucleic acid molecule is administered to the subject by intravenous injection. In some embodiments, the nucleic acid molecule is administered to the subject by subretinal injection. In some embodiments, the administration of the nucleic acid molecule targeting of retinal pigment epithelium—the most distal layer from the vitreal space. In some embodiments, the delivery of the nucleic acid molecule is targeted to retinal ganglion cells, bipolar cells, or both. The ganglion cells are, in some embodiments, accessible to intravitreal injection as disclosed herein. Intravitreal and/or subretinal injection can be used, in some embodiments to target the bipolar cells, for example in circumstances in which the photoreceptor cell layer is absent due to degeneration. Actual administration of the expression vectors for the light-sensitive proteins can be accomplished by using any physical method that will transport the vectors (e.g., viral vectors) into the target tissue(s) (e.g., eye tissue and brain tissue) of the subject. In some embodiments, the vectors can be administered systematically, e.g., by intravenous injection. Pharmaceutical compositions can be prepared, for example, as injectable formulations. The recombinant virus to be used can be utilized in liquid or freeze-dried form (in combination with one or more suitable preservatives and/or protective agents to protect the virus during the freeze-drying process). For gene therapy (e.g., of neuronal and ocular disorders which may be ameliorated by a specific gene product) a therapeutically effective dose of the recombinant virus expressing the therapeutic protein is administered to a host in need of such treatment. The use of the recombinant virus disclosed herein in the manufacture of a medicament for inducing immunity in, or providing gene therapy to, a host is within the scope of the present application. In instances where human dosages for the viral vector (e.g., AAV vector) have been established for at least some condition, those same dosages, or dosages that are between about 0.1% and 500%, more preferably between about 25% and 250% of the established human dosage can be used. Where no human dosage is established, as will be the case for newly-discovered pharmaceutical compositions, a suitable human dosage can be inferred from ED50or ID50values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals. A therapeutically effective amount of the expression vector (e.g., AAV vector) can be administered to a subject at various points of time. For example, the expression vector can be administered to the subject prior to, during, or after the subject has developed a disease or disorder. The expression vector can also be administered to the subject prior to, during, or after the occurrence of a disease or disorder (e.g., neuronal disorders, ocular disorders, or a combination thereof). In some embodiments, the expression vector is administered to the subject during remission of the disease or disorder. In some embodiments, the expression vector is administered prior to the onset of the disease or disorder in the subject. In some embodiments, the expression vector is administered to a subject at a risk of developing the disease or disorder. The dosing frequency of the expression vector (e.g., viral vector) can vary. For example, the viral vector can be administered to the subject about once every week, about once every two weeks, about once every month, about one every six months, about once every year, about once every two years, about once every three years, about once every four years, about once every five years, about once every six years, about once every seven years, about once every eight years, about once every nine years, about once every ten years, or about once every fifteen years. In some embodiments, the viral vector is administered to the subject at most about once every week, at most about once every two weeks, at most about once every month, at most about one every six months, at most about once every year, at most about once every two years, at most about once every three years, at most about once every four years, at most about once every five years, at most about once every six years, at most about once every seven years, at most about once every eight years, at most about once every nine years, at most about once every ten years, or at most about once every fifteen years. Uses of Light-Sensitive Proteins Light-sensitive proteins, for example the engineered ChRs, disclosed herein can be used to treat or prevent neuronal disorders, ocular disorders, or both. In some embodiments, the light-sensitive proteins can be used to restore and/or improve light sensitivity and/or vision of a subject. The visual signal is initially processed in the retina and most of conscious vision is then relayed to the lateral geniculate nucleus (LGN) of the thalamus, which in turn projects to the primary visual cortex. Since the visual signal is processed less in the LGN than in the cortex, and the number of cells dedicated to the same visual angle or retinal area is smaller in the LGN than in the cortex, it is contemplated herein that, in some embodiments, the cells of the LGN can be stimulated to restore vision. For example, LGN cells can be activated optogenetically using a composition (e.g., an expression vector, including a viral vector) comprising a coding sequence for a light-sensitive protein (e.g., one or more of the ChRs disclosed herein) to illuminate with visual patters the axon terminals of LGN cells where they form connections with the visual cortex, in the more accessible surface of the brain. LGN cell stimulation can, in some embodiments, evoke meaningful responses in blind and/or normal-sighted subjects. For example, normal or blind subjects can be caused to express one or more of the engineered ChRs in the LGN cells (e.g., via an AAV vector), as well as with GCAMP in the cortex to stimulate the axon terminals of LGN cell in the cortex and evoke responses in the cortex. A method, which comprises expressing a light-sensitive protein in a subject in need thereof is provided, where the light-sensitive protein is any one of the light-sensitive protein disclosed herein (e.g., the engineered ChRs). The light-sensitive protein can comprise an amino acid sequence having at least 80% sequence identity to SEQ ID NOs: 1-139, 141-147 and 149-196. In some embodiments, the light-sensitive protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 93, 109, 125-130, 132, 133, 136-138, 142, 146, 149, 150, and 155-196. In some embodiments, the light-sensitive protein comprises an amino acid sequence having at least 95% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 178-196. It is advantageous, in some embodiments, to use the light-sensitive protein with increased light sensitivity, ion conductance, photocurrent strength, or any combination thereof, in the method. For example, the light-sensitive protein can have at least two-fold improvement in one or more of light sensitivity, photocurrent strength, and ion conductance, as compared to a ChR consisting of the amino acid sequence of SEQ ID NO: 1, 3, 4, 155, 156, 176, or 177. The methods and composition (e.g., one or more of the ChRs and the expression vectors for the ChRs) disclosed herein can be used to treat a subject suffering from an ocular disorder, whereby the expression of the light-sensitive protein in the subject treats or ameliorates the ocular disorder. Examples of ocular disorders that can be treated or ameliorated include, but are not limited to, developmental abnormalities that affect both anterior and posterior segments of the eye. Anterior segment disorders include, for example, glaucoma, cataracts, corneal dystrophy, keratoconus. Posterior segment disorders include, for example, blinding disorders caused by photoreceptor malfunction and/or death caused by retinal dystrophies and degenerations. Retinal disorders include, for example, congenital stationary night blindness, age-related macular degeneration, congenital cone dystrophies, and a large group of retinitis-pigmentosa (RP)-related disorders. These disorders include genetically pre-disposed death of photoreceptor cells, rods and cones in the retina, occurring at various ages. Among those are severe retinopathies, such as subtypes of RP itself that progresses with age and causes blindness in childhood and early adulthood and RP-associated diseases, such as genetic subtypes of LCA, which frequently results in loss of vision during childhood, as early as the first year of life. The latter disorders are generally characterized by severe reduction, and of ten complete loss of photoreceptor cells, rods and cones. As disclosed herein, expressing the light-sensitive protein can comprise administering to the subject a recombinant expression vector comprising a nucleotide sequence encoding the light-sensitive protein. In some embodiments, the recombinant expression vector is a viral vector. The administering can be, for example, via intraocular injection, intravitreal injection, subretinal injection, intravenous delivery, or any combination thereof. In some embodiments, administering to the subject the recombinant expression vector comprises injecting the vector into the lateral geniculate nucleus of the subject. In some embodiments, injecting the vector into the lateral geniculate nucleus of the subject comprises injecting the vector into two or more locations of the lateral geniculate nucleus of the subject. In some embodiments, the subject is provided with a visual prosthesis before, at the same time as, or after delivery of said vector. In some embodiments, the visual prosthesis is a retinal implant, a cortical implant, a lateral geniculate nucleus implant, or an optic nerve implant. The method, in some embodiments, further comprises exposing the visual cortex of the subject to light signals Methods disclosed herein, in some embodiments, can be used in the treatment and/or restoration of at least partial vision to subjects that have lost vision due to ocular disorders (e.g., RPE-associated retinopathies, which are characterized by a long-term preservation of ocular tissue structure despite loss of function and by the association between function loss and the defect or absence of a normal gene in the ocular cells of the subject). A variety of such ocular disorders are known, including childhood onset blinding diseases, retinitis pigmentosa, macular degeneration, diabetic retinopathy, and ocular blinding diseases. Visual information is processed through the retina through two pathways: an ON pathway which signals the light ON, and an OFF pathway which signals the light OFF. It is generally believed that the existence of the ON and OFF pathway is important for the enhancement of contrast sensitivity. The visual signal in the ON pathway is relay front ON-cone bipolar cells to ON ganglion cells. Both ON-cone bipolar cells and ON-ganglion cells are depolarized in response to light. On the other hand, the visual signal in the OFF pathway is carried from OFF-cone bipolar cells to OFF ganglion cells. Both OFF-cone bipolar cells and OFF-ganglion cells are hypopolarized in response to light. Rod bipolar cells, which are responsible for the ability to see in dim light (scotopic vision), are ON bipolar cells (depolarized in response to light). Rod bipolar cells relay the vision signal through All amacrine cells (an ON type retinal cells) to ON an OFF cone bipolar cells. In some embodiments, the ocular disorders are refractive errors, cataracts, optic nerve disorders (e.g., glaucoma), retinal disorders, macular degeneration, diabetic eye problems, conjunctivitis, amblyopia, strabismus, or any combination thereof. The ocular disorder can be a hereditary ocular disease. Non-limiting examples of hereditary ocular disease include 3-methylglutaconic Aciduria with Cataracts; Neurologic involvement and neurtropenia; 3MC syndromes; Abetalipoproteinemia; Ablepharon-Macrostomia syndrome; Acrofacial Dysostosis; Cincinnati type; Adenomatous Polyposis of the Colon; autosomal Adrenoleukodystrophy; X-linked Adrenoleukodystrophy; Aicardi syndrome; Al Kaissi syndrome, Alagille syndrome, Aland Island eye disease; Albinism, ocular type 1; Albinism, Oculocutaneous, types I, II, III, IV, V, VI, and VII; Alkaptonuria; Alport syndrome (Collagen IV-related Nephropathies); Alström syndrome; Angiopathy, hereditary, with Nephropathy, Aneurysms, and muscle cramps; Aniridia types 1, 2, and 3; Anterior Segment Dysgenesis 6; Anterior Segment Dysgenesis 8; Anterior Segment Mesenchymal Dysgenesis; Anterior segment, brain, and facial anomalies; Apert syndrome; Aphakia, Congenital Primary; Arthrogryposis, Perthes disease, and Upward Gaze Palsy; Asphyxiating Thoracic Dysplasia 1; Ataxia and Polyneuropathy, Adult-Onset; Ataxia with Oculomotor Apraxia types 1, 2, 3, and 4; Ataxia-Telangiectasia; Autoinflammation with Arthritis and Dyskeratosis; Axenfeld-Rieger Anomaly, Plus; Axenfeld-Rieger syndrome types 1, 2, 3, and 4; Ayme-Gripp syndrome; Baker-Gordon syndrome; Baller-Gerold syndrome; Baraitser-Winter syndrome types 1 and 2; Barber-Say syndrome; Bardet-Biedl syndromes; Basal Cell Nevus syndrome; Basel-Vanagaite-S mirin-Yo sef syndrome; Beare-Stevenson syndrome; Behcet-Like Familial autoinflammatory syndrome; Behr Early Onset Optic Atrophy syndromes; Behr syndrome; Biemond syndrome II; Bietti Crystalline Corneoretinal dystrophy; Birk-Landau-Perez syndrome; Blatt Distichiasis; Blepharocheilodontic syndrome types 1 and 2; Blepharoptosis, Myopia, Ectopia Lentis; Blue Cone Monochromacy; Blue Diaper syndrome; Bornholm Eye disease; Bosma Arhinia Microphthalmia syndrome; BPES syndrome; Branchiooculofacial syndrome; Brittle Cornea syndrome 1 and 2; Brown-Vialetto-Van Laere syndrome 2; Canavan disease; Carey-Fineman-Ziter syndrome; Carpenter syndrome; Cataract and Ichthyosis; Cataracts 13, Congenital, in Adult i RBC Phenotype; Cataracts 34, 43 and 45; Cataracts 46, Juvenile-Onset; Cataracts, Anterior Polar 2; Cataracts, Anterior Polar with Guttata; Cataracts, Ataxia, Short Stature, and Mental Retardation; Cataracts, Congenital Cerulean; Cataracts, Congenital Nuclear; Cataracts, Congenital Sutural with Punctate and Cerulean Opacities; Cataracts, Congenital with Sclerocornea and Glaucoma; Cataracts, Congenital Zonular Pulverulent 1; Cataracts, Congenital Zonular Pulverulent 3; Cataracts, Congenital Zonular With Sutural Opacities; Cataracts, Congenital, and Hypomyelinating Leukodystrophy; Cataracts, Congenital, Autosomal Dominant; Cataracts, Congenital, Autosomal Recessive 2; Cataracts, Congenital, Autosomal Recessive types 3, 4 and 5; Cataracts, Congenital, Deafness, Short Stature, Developmental Delay; Cataracts, Congenital, Facial Dysmorphism, and Neuropathy; Cataracts, Congenital, Intellectual Disability, Abnormal Striatum, and ADHD; Cataracts, Congenital, Posterior Polar; Cataracts, Congenital, Volkmann type; Cataracts, Congenital, with Brain Hemorrhage and Subependymal Calcification; Cataracts, Congenital, with Cleft Palate; Cataracts, Congenital, with Intellectual Disability; Cataracts, Congenital, With Short Stature and Minor Skeletal Anomalies; Cataracts, Congenital, X-Linked; Cataracts, Coppock-Like; Cataracts, CRYAA Mutations; Cataracts, Growth Hormone Deficiency, and Skeletal Dysplasia; Cataracts, Hearing Loss, and Neurodegeneration; Cataracts, Lamellar; Cerebellar Atrophy, Visual Impairment, and Psychomotor Retardation; Cerebral Amyloid Angiopathy; Cerebral Atrophy, Autosomal Recessive; Cerebral Cavernous Malformations; Cerebral Palsy, Spastic Quadriplegic, 3; Cerebrooculofacio skeletal syndrome; Cerebrotendinous Xanthomatosis; Charcot-Marie-Tooth disease with Glaucoma; Charcot-Marie-Tooth diseases; CHARGE syndrome; Chédiak-Higashi syndrome; ChondrodysplasiaPunctata2; CHOPS syndrome; Chorioretinal dysplasia, lymphedema, and microcephaly; Chorioretinal dysplasia, microcephaly, and mental retardation; Chorioretinopathy with Microcephaly 1, 2 and 3; Chorioretinopathy, Ataxia, and Hypogonadism; Choroidal dystrophy, Central Areolar 1 and 2; Choroideremia; Cleft Palate, Psychomotor Retardation, and Distinctive Facial Features; Coats Plus syndrome; Cockayne syndrome, types A and B; CODAS syndrome; Cohen syndrome; Cole-Carpenter syndrome 1 and 2; Coloboma of the Optic Nerve; Coloboma, Isolated; Coloboma, Microphthalmia, Albinism, and Deafness; Coloboma, Ptosis, Hypertelorism, and Global Delay; Color Blindness, Red-Green, Partial; Colorblindness-Achromatopsia types 2, 3, 4, and 5; Colorblindness-Tritanopia; Combined Oxidative Phosphorylation Deficiency 32; Cone dystrophy 3; Cone dystrophy, Peripheral; Cone-Rod Dystrophies, AD and AR; Cone-Rod Dystrophies, X-Linked; Cone-Rod dystrophy with Decreased Male Fertility; Cone-Rod dystrophy with Hearing Loss; Congenital disorder of Glycosylation, types Ia, Ij and Iq; Congenital Heart Defects, Dysmorphic Facies, and Intellectual Developmental disorder; Conjunctivitis, Ligneous; Cornea Plana; Cornea, Ring Dermoid; Corneal dystrophy, Avellino type; Corneal dystrophy, Band-Shaped; Corneal dystrophy, Congenital Endothelial types 1 and 2; Corneal dystrophy, Congenital Stromal; Corneal dystrophy, Endothelial X-Linked; Corneal dystrophy, Epithelial Basement Membrane; Corneal dystrophy, Fleck; Corneal dystrophy, Fuchs Endothelial, Early Onset; Corneal dystrophy, Fuchs Endothelial, Late Onset; Corneal dystrophy, Fuchs Endothelial, Late Onset 2; Corneal dystrophy, Gelatinous Drop-like; Corneal dystrophy, Granular; Corneal dystrophy, Lattice types I and II; Corneal dystrophy, Lisch Epithelial; Corneal dystrophy, Macular; Corneal dystrophy, Meesmann; Corneal dystrophy, Posterior Amorphous; Corneal dystrophy, Posterior Polymorphous types 1, 2, 3, and 4; Corneal dystrophy, Recurrent Erosions; Corneal dystrophy, Reis-Bücklers; Corneal dystrophy, Schnyder; Corneal dystrophy, Stocker-Holt; Corneal dystrophy, Subepithelial Mucinous; Corneal dystrophy, Thiel-Behnke; Cornelia de Lange syndrome; Corpus Callosum Agenesis with Facial Anomalies and Cerebellar Ataxia; Cranial Dysinnervation disorders with Strabismus and Arthrogryposis; Craniofacial-Deafness-Hand syndrome; Crouzon syndrome; Cryptophthalmos; Cystinosis; Danon disease; Dermochondrocorneal dystrophy; Developmental Delay with Short Stature, Dysmorphic Features, and Sparse Hair; Donnai-Barrow syndrome; Doyne Honeycomb Macular dystrophy; Duane Retraction syndrome types 1, 2, and 3; Duane-Radial Ray syndrome; Dysautonomia, Familial; Dyskeratosis Congenita; Dyskeratosis, Hereditary Benign Intraepithelial; Dystonia, Childhood Onset, With Optic Atrophy; Ectopia Lentis et Pupillae; Ectopia lentis, Isolated AD and AR; EDICT syndrome; EEM syndrome; Ehlers-Danlos syndrome, type VIA; Elsahy-Waters syndrome; Encephalocraniocutaneous Lipomatosis; Encephalopathy Due To Defective Mitochondrial And Peroxisomal Fission 2; Encephalopathy, Early-Onset, With Brain Atrophy and Thin Corpus Callo sum; Encephalopathy, Progressive, Early-Onset, with Brain Atrophy and Spasticity; Encephalopathy, Progressive, with Amyotrophy and Optic Atrophy; Epileptic Encephalopathy, Early Infantile types 28, 47, 48 and 58; Epileptic Encephalopathy, Infantile or Early Childhood 2; Exfoliation Glaucoma; External Ophthalmoplegia, ANTI and mtDNA Mutations; External Ophthalmoplegia, C10ORF2 and mtDNA Mutations; External Ophthalmoplegia, Facial Weakness, and Malignant Hyperthermia; External Ophthalmoplegia, POLG and mtDNA Mutations; External Ophthalmoplegia, Progressive, with mtDNA Deletions, AR 3 and AR 4; Eye Movement disorders with CACNA1A Mutations; Fabry disease; Facial Palsy, Congenital, with Ptosis and Velopharyngeal Dysfunction; Familial Acorea, Microphthalmia and Cataract syndrome; Familial Exudative Vitreoretinopathy EVR1, EVR2, EVR4, EVR5, EVR6 and EVR7; Familial Internal Retinal Membrane dystrophy; Fanilial Exudative Vitreoretinopathy, EVR3; Feingold syndrome 1; Fibrosis of Extraocular Muscles with Synergistic Divergence; Fibrosis of Extraocular Muscles CFEOM1, CFEOM2, CFEOM3C and CFEOMS; Fibrosis of Extraocular Muscles, Tukel CFEOM syndrome; Filippi syndrome; Fleck Retina of Kandori; Fleck Retina, Benign Familial; Flecked Retina syndromes; Focal Dermal Hypoplasia; Foveal Hypoplasia 1 and 2; Foveal Hypoplasia and Anterior Chamber Dysgenesis; Fraser syndromes 1, 2 and 3; Friedreich Ataxia 1; Fructose Intolerance; Fucosidosis; Fundus Albipunctatus; Gabriele-de Vries syndrome; Galactokinase Deficiency; Galactose Epimerase Deficiency; Galactosemia; Galloway-Mowat syndrome; GAPO syndrome; Gaucher disease; Gaze Palsy, Familial Horizontal, with Progressive Scoliosis 1 and 2; Gillespie syndrome; Glaucoma, Congenital Primary A, B, C, D, and E; Glaucoma, Open Angle, Juvenile and Primary; Glaucoma, pigment dispersion syndrome; GM1 Gangliosidosis; GM3 Synthase Deficiency; Goldenhar syndrome Spectrum; Goldmann-Favre syndrome; Gorlin-Chaudhry-Moss syndrome; Gracile Bone Dysplasia; Gurrieri syndrome; Gyrate Atrophy; Hallermann-Streiff syndrome; Harboyan syndrome; Heart and Brain Malformation syndrome; Heimler syndrome 1 and 2; HELIX syndrome; Hereditary Mucoepithelial Dysplasia; Hermansky-Pudlak syndrome; Histiocytic Dermatoarthritis; Homocystinuria, Beta-Synthase Deficiency; Homocystinuria, MTHFR Deficiency; Hoyeraal-Hreidarsson syndrome; Hunter syndrome (MPS II); Hurler and Scheie syndromes (MPS IH, IS, IH/S); Hyperferritinemia-Cataract syndrome; Hyperoxaluria, Primary, type I; Hyperphosphatasia with Mental Retardation syndrome 6; Hypoparathyroidism, Familial Isolated; Hypotonia, Infantile, with Psychomotor Retardation; Hypotonia, Infantile, with Psychomotor Retardation and Characteristic Facies 1, 2 and 3; Hypotrichosis with Juvenile Macular Degeneration; Hypotrichosis-Lymphedema-Telangiectasia-Renal Defect syndrome; IFAP (BRESHECK) syndrome; Immunodeficiency-Centromeric Instability-Facial Anomalies syndrome 3; Incontinentia Pigmenti; Infantile Cerebellar-Retinal Degeneration; Intellectual Disability with Dysmorphic Facies and Ptosis; Iridogoniodysgenesis and Skeletal Anomalies; Iridogoniodysgenesis, types 1 and 2; Jackson-Weiss syndrome; Jalili syndrome; Joint Laxity, Short Stature, and Myopia; Joubert syndrome and Related disorders; Kabuki syndrome 1 and 2; Kahrizi syndrome; Kaufman Oculocerebrofacial syndrome; Kearns-Sayre syndrome; Kenny-Caffey syndrome, type 2; Keratitis, hereditary; Keratoconus types 1, 2, 3, 4, and 9; Keratoconus Posticus Circumscriptus; Keratoendotheliitis Fugax Hereditaria; Keratosis Follicularis Spinulosa Decalvans, X-Linked; Keritosis Follicular Spinulosa Decalvans, AD; KID syndrome; Kniest Dysplasia; Knobloch syndrome 1, 2 and 3; Krabbe disease; Kufor-Rakeb syndrome; Lacrimal Puncta Agenesis; LCAT Deficiency; Leber Congenital Amaurosis; Leber Congenital Amaurosis with Early-Onset Deafness; Leber Optic Atrophy; LEOPARD syndrome; Leukodystrophy, Hypomyelinating, 13 and 15; Leukoencephalopathy with Vanishing White Matter; Lowe Oculocerebrorenal syndrome; Lymphedema-Distichiasis syndrome; Macrophthalmia, Colobomatous, with Microcornea; Macular Degeneration, Early-Onset; Macular dystrophy with Central Cone Involvement; Macular dystrophy, Fenestrated type; Macular dystrophy, North Carolina; Macular dystrophy, Occult; Macular dystrophy, Patterned 1, 2 and 3; Macular dystrophy, Vitelliform types 1, 2, 3, 4, and 5; Macular Edema, Autosomal Dominant Cystoid; Majewski syndrome; Mandibulofacial Dysostosis with Alopecia; Manitoba Oculotrichoanal syndrome; Mannosidosis, Alpha B; Marfan Lipodystrophy syndrome; Marfan syndrome; Marinesco-Sjogren syndrome; Maroteaux-Lamy syndrome (MPS VI); Marshall syndrome; McCune-Albright syndrome; Meckel syndrome; Meester-Loeys syndrome; Megalocornea; Megalocornea, Ectopia Lentis, and Spherophakia; MELAS syndrome; Mental Retardation AD 31, AD 34, AD 53 and AD 57; Mental Retardation, X-Linked 99, Syndromic, Female-Restricted; Microcephaly 20, Primary, Autosomal Recessive; Microcephaly, Congenital Cataracts, and Psoriasiform Dermatitis; Microcoria, Congenital; Microcornea, Myopia, Telecanthus and Posteriorly-Rotated Ears; Microphthalmia and Anophthalmia, ALDH1A3 Associated; Microphthalmia with Coloboma, AD; Microphthalmia with Coloboma, X-Linked; Microphthalmia with Limb Anomalies; Microphthalmia with Retinitis Pigmentosa; Microphthalmia, AR; Microphthalmia, Isolated, with Cataract; Microphthalmia, Syndromic 1, 10, 2, 3, 4, 5, 6, 7, 8, and 9; Mitochondrial DNA Depletion syndrome 1 and 3; Mitochondrial Short-Chain Enoyl-CoA Hydratase 1 Deficiency; Moebius syndrome; Morquio syndrome (MPS IVA); Morquio syndrome (MPS IVB); Mowat-Wilson syndrome; Multiple Endocrine Neoplasia, type IIBMultiple Mitochondrial; Dysfunctions syndrome 4; Muscular dystrophy, Congenital, with Cataracts and Intellectual Disability; Myasthenic syndromes, Congenital, Including AChR Deficiency; Myopathy, Mitochondrial Anomalies, and Ataxia; Myopia 1, X-linked, Nonsyndromal; Myopia 2, Autosomal Dominant, Nonsyndromal; Myopia 25, Autosomal Dominant, Nonsyndromic; Myopia 26, X-Linked, Female-Limited; Myopia and Deafness; Myopia, AR, with Cataracts and Vitreoretinal Degeneration; Myotonic dystrophy 1 and 2; Nance-Horan syndrome; Nanophthalmos 1, 3, AD, Plus syndrome, with Retinitis Pigmentosa, and with Retinopathy; Nemaline Myopathy 10; Neu-Laxova syndrome 1 and 2; Neuhauser syndrome; Neuraminidase Deficiency; Neurodegeneration with Ataxia, Dystonia, and Gaze Palsy, Childhood-Onset; Neurodegeneration with Brain Iron Accumulation; Neurodevelopmental disorder With or Without Seizures and Gait Abnormalities; Neurodevelopmental disorder with Progressive Microcephaly, Spasticity, and Brain Anomalies; Neurodevelopmental disorder, Mitochondrial, with Abnormal Movements and Lactic Acidosis; Neurofibromatosis types I and II; Neuronal Ceroid Lipofuscinoses; Neuropathy, Ataxia, and Retinitis Pigmentosa; Niemann-Pick disease, types C2, A and B, and C1 (D); Night Blindness, Congenital Stationary (CSNB1A, CSNB1B, CSNB1C, CSNB1E, CSNB1H, CSNB2A, CSNB2B, CSNBAD1, CSNBAD2, and CSNBAD3); Noonan syndrome; Norrie disease; Nystagmus 1, Congenital, X-linked; Nystagmus 2, Congenital, AD; Nystagmus 3, Congenital, AD; Nystagmus 4, AD; Nystagmus 5, Congenital, X-linked; Nystagmus 6, Congenital, X-linked; Nystagmus 7, Congenital, AD; Nystagmus-Split Hand syndrome; Oculoauricular syndrome (including with Hypopigmentation); Oculodentodigital Dysplasia; Oculomotor Apraxia; Oculootofacial Dysplasia; Oculopharyngeal Muscular dystrophy; Oculopharyngodistal Myopathy; Oguchi disease type 1 and 2; Optic Atrophy (1, 10, 11, 2 (X-linked), 3, Cataracts, 4, 5, 6, 7, 9, with Intellectual Disability, with Areflexia, Ataxia, Hearing Loss, with Ophthalmoplegia, Myopathy, and Neuropathy); Optic Nerve Edema, Splenomegaly, Cytopenias; Optic Nerve Hypoplasia, Bilateral; Organoid Nevus syndrome; Orofaciodigital syndrome types TX and VI; Osteogenesis Imperfecta (including type VII); Osteoporosis-Pseudoglioma syndrome; Palmoplantar Keratoderma and Woolly Hair; Pantothenate Kinase-Associated Neurodegeneration; Papillorenal syndrome; Pearson Marrow-Pancreas syndrome; PEHO syndrome; PEHO-Like syndrome; Pelizeaus-Merzbacher disease; Peroxisome Biogenesis disorders (1A (Zellweger), 1B (neonatal adrenoleukodystrophy, 3B (Infantile Refsum disease)); Peroxisomol Fatty Acyl-CoA Reductase 1 disorder; Perrault syndrome; Persistent Hyperplastic Primary Vitreous; Peters Anomaly; Peters-Plus syndrome; Pfeiffer syndrome; Pierson syndrome; Pigmentary Retinopathy with Congenital Sideroblastic Anemia; Pigmented Paravenous Chorioretinal Atrophy; Pontocerebellar Hypoplasia 11, 3 and 7; Potter disease type I; Progeroid Short Stature with Pigmented Nevi; Pseudohypoparathyroidism type 1A; Pseudoxanthoma Elasticum; Pseudoxanthoma Elasticum-Like disease; RAB18 Deficiency; Refsum disease, Adult; Retinal Arteriolar Tortuosity; Retinal Cone dystrophy 3B; Retinal detachment with Lattice Degeneration; Retinal dystrophy and Obesity; Retinal dystrophy (with Inner Retinal Abnormalities, with or without Extraocular Anomalies, with or without Macular Staphyloma, Bothnia type, and Newfoundland type); Retinal dystrophy, Cataracts, and Short Stature; Retinal Nonattachment, Congenital; Retinitis Pigmentosa 1, 2 (X-linked), 25, 3, X-Linked, 38, 42, 47, 71, 72, 75, 76, 77, 78, 79, 80, and 81; Retinitis Pigmentosa and Mental Retardation; Retinitis Pigmentosa with Ataxia; Retinitis Pigmentosa With or Without Skeletal Anomalies; Retinitis Pigmentosa (AD; AR; deafness, Mental Retardation and Hypogonadism; Hearing Loss, Ataxia, Cataract, and Polyneuropathy; RDH11 syndrome); RetinitisPunctata Albescens; Retinoblastoma; Retinopathy with Neutropenia; Retinoschisis, Juvenile; Revesz syndrome; Rhizomelic ChondrodysplasiaPunctata; Roberts syndrome; Rosenthal-Kloepfer syndrome; Rothmund-Thomson syndrome; Rubinstein-Taybi syndrome 1 and 2; Saethre-Chotzen syndrome; Sandhoff disease; Sanfilippo syndrome (MPS IIIA, B, C, D); Schurrs-Hoeijmakers syndrome; Sclerocornea; Sengers syndrome; Senior-Loken syndromes; Septooptic Dysplasia; Setleis syndrome; Short Stature, Hearing Loss, Retinitis Pigmentosa, and Distinctive Facies; SHORT syndrome; Short-Rib Thoracic Dysplasia 9; Sickle Cell Anemia; Singleton-Merten syndrome 1 and 2; Sjogren-Larsson syndrome; Smith-Lemli-Opitz syndrome; Smith-Magenis syndrome; Sorsby Macular Coloboma syndrome; Sorsby Pseudoinflammatory Fundus dystrophy; Spastic Ataxia (2; 4, mtPAP Deficiency; 6, Charlevoix-Saguenay type; 7, with Miosis; 8, Autosomal Recessive, with Hypomyelinating Leukodystrophy; and Optic Atrophy, Mental Retardation); Spastic Paraplegia (including types 11; 15; 2; 46; 5A; 7; 74; 75; 78; with Psychomotor Retardation and Seizures, with Intellectual Disability, Nystagmus, and Obesity; with Optic Atrophy, and Neuropathy); Spherophakia and Metaphyseal Dysplasia; Spherophakia with Inguinal Hernia; Spherophakia, Isolated; Spinocerebellar Ataxia (including types 1, 18, 3, 37, 38, 42, 7, Autosomal Recessive 7, and Infantile-Onset); Spondyloepiphyseal Dysplasia Congenita; Spondylometaphyseal Dysplasia, Axial; Spondyloocular syndrome; Stargardt disease; Stickler syndrome (including types I, II and IV); Strøomme syndrome; Sulfite Oxidase Deficiency; Sweeney-Cox syndrome; Takenouchi-Kosaki syndrome; Tangier disease; Tay-Sachs disease; Temtamy syndrome; Tenorio syndrome; Treacher Collins-Franceschetti syndrome; Trichomegaly Plus syndrome; Tuberous Sclerosis 1 and 2; Tyrosinemia, type II; Usher syndrome types I, II, III and IV; Vici syndrome; Vitreoretinal Degeneration, Snowflake type; Vitreoretinochoroidopathy; Vitreoretinopathy with Epiphyseal Dysplasia; Von Hippel-Lindau syndrome; Waardenburg syndrome types 1, 2, 3 and 4; Wagner syndrome; Walker-Warburg syndrome; Watson syndrome; Weill-Marchesani syndrome 1; Weill-Marchesani syndrome 2; Weill-Marchesani-Like syndrome; Wildervanck syndrome; Williams syndrome; Wilson disease; Wolfram syndrome 1 and 2; and Zhu-Tokita-Takenouchi-Kim syndrome (“ZTTK syndrome”). The methods and compositions disclosed herein can restore and/or enhance visual function in a subject in need thereof. In some embodiments, the restoration and/or the enhancement of visual function provides for patterned vision and image recognition by the subject. The image recognition can be, for example, of a static image or a pattern. The light intensity that can be provided by the restoration and/or enhancement for image recognition can vary, for example, it can be at a light intensity of from about 104W/cm2to about 1 W/cm2. In some embodiments, the image recognition is of a moving image or a pattern. The methods and compositions disclosed herein can restore and/or enhance vision in a subject in need thereof. The method, for example, can comprise measuring vision before and/or after administering a nucleic acid molecule (e.g., a vector) comprising a coding sequence for a light-sensitive protein (e.g., the engineered ChR). Many methods are known in the art to measure vision, including the following visual responses: (1) a light detection response by the subject after exposure to a light stimulus—in which evidence is sought for a reliable response of an indication or movement in the general direction of the light by the subject individual when the light it is turned on is; (2) a light projection response by the subject after exposure to a light stimulus in which evidence is sought for a reliable response of indication or movement in the specific direction of the light by the individual when the light is turned on; (3) a light resolution by the subject of a light vs. dark patterned visual stimulus, which measures the subject's capability of resolving light vs dark patterned visual stimuli as evidenced by: (a) the presence of demonstrable reliable optokinetically produced mystagmoid eye movements and/or related head or body movements that demonstrate tracking of the target, and/or (b) the presence of a reliable ability to discriminate a pattern visual stimulus and to indicate such discrimination by verbal or non-verbal means, including, for example pointing, or pressing a bar or a button; and (4) an electrical recording of a visual cortex response to a light flash stimulus or a pattern visual stimulus, which is an endpoint of electrical transmission from a restored retina to the visual cortex. Measurement may be by electrical recording on the scalp surface at the region of the visual cortex, on the cortical surface, and/or recording within cells of the visual cortex. The methods and compositions disclosed herein can be used in combination with other forms of vision therapy, including the use of visual prostheses. Visual prostheses include, but are not limited to, retinal implants, cortical implants, lateral geniculate nucleus implants, optic nerve implants, and any combination thereof. For example, the subject being treated with the methods and/or compositions disclosed herein can be provided with a visual prosthesis before, at the same time as, or after the treatment. In some embodiments, the methods and/or compositions disclosed herein are used in combination of one or more visual stimulation techniques used in, e.g., low vision rehabilitation. Some embodiments provide a method for treating a subject suffering from a retinal degenerative or neurodegenerative disease. The method comprises, for example, expressing a light-sensitive protein (e.g., the ChRs disclosed herein) in the subject or administering the light-sensitive protein to the subject. Expressing the light-sensitive protein in the subject comprises, in some embodiments, delivering a nucleic acid molecule encoding the light-sensitive protein (e.g., a viral expression vector with the coding sequence of the light-sensitive protein) to the subject, thereby expressing the light-sensitive protein in the subject. In the method disclosed herein, a therapeutically effective amount of the light-sensitive protein and/or the nucleic acid molecule encoding the light-sensitive protein can be administered to the subject. The administration can be conducted, for example, via injection(s). Provided herein are light-sensitive proteins (e.g., ChRs) with improved properties and characteristics that enhance, among other things, optogenetic techniques. For example, some of the light-sensitive protein provide greater unitary conductance, sodium specificity, or the enhancement of the short-wavelength sensitivity, by inducing a blueshift in absorption maxima. Optogenetic techniques involve the introduction of light-activated channels and enzymes that allow manipulation of neural activity and control of neuronal function. In some embodiments, the disclosed methods and compositions can be introduced into cells and facilitate the manipulation of the cells' activity and function. The cells can be retinal neurons, for example, one or more of ON- and OFF-type retinal ganglion cells, retinal rod bipolar cells, amacrine cells, and ON and OFF retinal cone bipolar cells, or any combination thereof. Disclosed methods and/or compositions can be used in, among other things, retinal gene therapy for mammals. For example, a genetically engineered ocular cell is produced by contacting the cell with an exogenous nucleic acid under conditions in which the exogenous nucleic acid is introduced to the cell for expressing one or more of the light-sensitive proteins disclosed herein. In some embodiments, the introduction and/or expression of the light-sensitive protein(s) to the cell, for example an monocular neuronal cell or binocular neuronal cell, result in sensitivity to the retinas and restoring one or more aspects of visual responses and functional vision to a subject, for example a subject suffering from macular degeneration. Without being limited to any particular theory, it is believed that by restoring light sensitivity to a retina lacking this capacity, due to disease, a mechanism for the most basic light-responses that are required for vision is provided. In some embodiments, a blue-shifted ChR is inserted into the retinal neurons that survived after the rods and cones have died in an area or portion of the retina of a subject. In some embodiments, a blue-shifted ChR is inserted into retinal interneurons. These cells then can become light sensitive and send signals via the optic nerve and higher order visual pathways to the visual cortex where visual perception occurs. In some embodiments, expressing the light-sensitive protein in the subject restores or enhances the photosensitivity of the retinal neurons in the subject, and/or the photosensitivity of a retina or a portion thereof of the subject. It is advantageous, in some embodiments, for the light-sensitive protein to be expressed in retinal cells, monocular neuronal cells, binocular neuronal cells, electrically active cells, or any combination thereof in the subject. In some embodiments, the one or more retinal cells comprises retinal ganglion cells, retinal neurons or any combination thereof. In some embodiments, the subject suffers from blindness or vision loss, and optionally the blindness or visional loss is a result of a degenerative diseases. In some embodiments, one or more photoreceptor cells of the subject are degenerating or have degenerated. In some embodiments, the subject suffered and/or is suffering from retinal detachment and/or photoreceptor loss due to trauma or head injury. In some embodiments, the methods and compositions disclosed herein can be used to treat or ameliorate one or more neuronal disorders, such as neuropathic pain. In some embodiments, the neuronal disorder is affected by light sensitivity of the subject. In some embodiments, the neuronal disorder is related to a behavior abnormality controlled or affected by light sensitivity of the subject. In some embodiments, the neuronal disorder is affected by light. In some embodiments, the neuronal disorder is related to a behavior abnormality controlled or affected by light. In some embodiments, the neuronal disorder has one or more symptoms affected by light sensitivity of the subject. In some embodiments, the neuronal disorder has one or more symptoms controlled or affected by light sensitivity of the subject. In some embodiments, at least one or more symptoms of the neuronal disorder are affected by light. The method can further comprise delivering light to the subject, and optionally delivery light comprises placing a plurality of fiber optic-cables on the skull of the subject. In some embodiments, the light activates the light-sensitive protein, thereby activating light-dependent neuronal cells in the subject. In some embodiments, the method comprises effecting light-controlled neuronal activation, light-induced behavioral control, or both in the subject. In some embodiments, the effecting light-controlled neuronal activation, light-induced behavioral control, or both is performed without disruption to any tissues in the subject. In some embodiments, the effecting light-controlled neuronal activation, light-induced behavioral control, or both is performed without disruption to any tissues in the subject. In some embodiments, the effecting light-controlled neuronal activation, light-induced behavioral control, or both is performed without disruption to one or more of the tissues in the subject. The tissues can be, or can comprise, brain tissue, eye tissue, or both. EXAMPLES Some aspects of the embodiments discussed above are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the present disclosure. Experimental Material and Methods The following experimental methods were used for Examples 1-7 described below. Construct Design and Cloning The design, construction, and characterization of the recombination library of chimeras is described in detail in Bedbrook et al. (Proc Natl Acad Sci USA., 2017, 114(13):E2624-E2633). The 10-block contiguous and 10-block noncontiguous recombination libraries were designed and built using SCHEMA recombination as described in Bedbrook et al. Software packages for calculating SCHEMA energies are openly available at cheme.che.caltech.edu/groups/fha/Software.htm. Each chimeric ChR variant in these libraries is composed of blocks of sequence from the parental ChR (CsChrimR, C1C2, and CheRiff), including chimeras with single-block swaps (chimeras consisting of 9 blocks of one parent and a single block from one of the other two parents) and multi-block-swap chimera sequences. Selected ChR variant genes were inserted into a constant vector backbone [pFCK from Addgene plasmid #51693] with a CMV promoter, Golgi export trafficking signal (TS) sequence (KSRITSEGEYIPLDQIDINV (SEQ ID NO: 199)), and fluorescent protein (mKate). All ChR variants contain the SpyTag sequence following the N-terminal signal peptide for the SpyTag/SpyCatcher labeling assays used to characterize ChR membrane localization. The C1C2 parent for the recombination libraries is mammalian codon-optimized. ChR variant sequences used in this study are provided in the Sequence Listing submitted herewith. All selected ChR genes were synthesized and cloned in the pFCK mammalian expression vector by Twist Bioscience (San Francisco, CA). For visualization, sequence alignment between C1C2 and engineered ChRs were created using ClustalΩ and visualized using ENDscript (FIGS.5C and5D). For characterization in neurons, selected ChR variants [ChRger1, ChRger2, ChRger3, CoChR, and hChR2(H134R)] were inserted into a pAAV-hSyn vector backbone [Addgene plasmid #26973], a pAAV-CamKIIa vector backbone [Addgene plasmid #51087], and a pAAV-CAG-DIO vector backbone [Addgene plasmid #104052]. In all backbones, each ChR was inserted with a TS sequence and fluorescent protein (eYFP). HEK293T Cell and Primary Neuronal Cultures The culturing and characterization ChRs in HEK cells is described in Bedbrook et al. Briefly, HEK cells were cultured at 37° C. and 5% CO2in D10 [DMEM supplemented with 10% (vol/vol) FBS, 1% sodium bicarbonate, and 1% sodium pyruvate]. HEK cells were transfected with purified ChR variant DNA using FuGENE®6 reagent according to the manufacturer's (Promega) recommendations. Cells were given 48 hours to express the ChRs before photocurrent measurements. Primary hippocampal neuronal cultures were prepped from C57BL/6N mouse embryos 16-18 days post-fertilization (E16-E18 Charles-River Labs) and cultured at 37° C. in the presence of 5% CO2in Neurobasal media supplemented with glutamine and B27. Cells were transduced 3-4 days after plating with rAAV-PHP.eB packaging ChR2(H134R), CoChR, ChRger1, ChRger2, or ChRger3. Whole-cell recordings were performed 5-10 days after transduction. Patch-Clamp Electrophysiology Whole-cell patch-clamp and cell-attached recordings were performed in transfected HEK cells, transduced cultured neurons, and acute brain slices to measure light-activated inward currents or neuronal firing. For electrophysiological recordings, cultured cells were continuously perfused with extracellular solution at room temperature (in mM: 140 NaCl, 5 KCl, 10 HEPES, 2 MgCl2, 2 CaCl2, 10 glucose; pH 7.35) while mounted on the microscope stage. For slice recordings, 32° C. artificial cerebrospinal fluid (ACSF) was continuously perfused over slices. ACSF contained 127 mM NaCl, 2.5 mM KCl, 25 mM NaHCO3, 1.25 mM NaH2PO4, 12 mM d-glucose, 0.4 mM sodium ascorbate, 2 mM CaCl2, and 1 mM MgCl2and was bubbled continuously with 95% oxygen/5% CO2. Firing and photocurrent measurements were performed in the presence of 3 mM kynurenic acid and 100 μM picrotoxin to block optically evoked ionotropic glutamatergic and GABAergic currents, respectively. Patch pipettes were fabricated from borosilicate capillary glass tubing (1B150-4; World Precision Instruments) using a model P-2000 laser puller (Sutter Instruments) to resistances of 3-6 MΩ. Pipettes were filled with K-gluconate intracellular solution containing the following (in mM): 134 K gluconate, 5 EGTA, 10 HEPES, 2 MgCl2, 0.5 CaCl2, 3 ATP, and 0.2 GTP. Whole-cell patch-clamp and cell-attached recordings were made using a Multiclamp 700B amplifier (Molecular Devices), a Digidata 1440 digitizer (Molecular Devices), and a PC running pClamp (version 10.4) software (Molecular Devices) to generate current injection waveforms and to record voltage and current traces. Photocurrents were recorded from cells in voltage clamp held at −60 mV. Neuronal firing was measured in current clamp mode with current injection for a −60 mV holding potential. Access resistance (Ra) and membrane resistance (Rm) were monitored throughout recording, and cells were discarded if Raor Rmchanged more than 15%. During ChR variant functional screening in HEK cells, photocurrents were recorded from cells that passed our recording criteria: Rm>200 MΩ and holding current >−100 pA. Our measured membrane properties of ChR expressing neurons were consistent with previous literature of opsin-expressing cells and are also consistent with previous reports of properties of cultured hippocampal neurons and PFC neurons in slice (FIG.14A-B). For cell culture experiments, the experimenter was blinded to the identity of the ChR being patched but not to the fluorescence level of the cells. For acute slice recordings, the experimenter was not blinded to the identity of the ChR. Light Delivery and Imaging Patch-clamp recordings were done with short light pulses to measure photocurrents. Light pulse duration, wavelength, and power were varied depending on the experiment as described herein. Light pulses were generated using a Lumencor SPECTRAX light engine. The illumination/output spectra for each color were measured (FIGS.9A-B). To evaluate normalized green photocurrent, photocurrent strength was measured at three wavelengths (peak±half width at half maximum): (red) 640±3 nm, (green) 546±16 nm, and (cyan) 481±3 nm with a 0.5 s light pulse. Light intensity was matched for these measurements, with 481 nm light at 2.3 mW mm2, 546 nm light at 2.8 mW mm2, and 640 nm light at 2.2 mW mm−2. For full spectra measurements depicted inFIG.2E, photocurrents were measured at seven different wavelengths (peak±half width half maximum): (red) 640±3 nm, (yellow) 567±13 nm, (green) 546±16 nm, (teal) 523±6 nm, (cyan) 481±3 nm LED, (blue) 439±8 nm LED, and (violet) 397±3 nm with a 0.5 s light pulse for each color. Light intensity is matched across wavelengths at 1.3 mW mm2. Imaging of ChR variants expression in HEK cells was performed using an Andor Neo 5.5 sCMOS camera and Micro-Manager Open Source Microscopy Software. Imaging of ChR expression in neuronal cultures and in brain slices was performed using a Zeiss LSM 880 confocal microscope and Zen software. Electrophysiology Data Analysis Electrophysiology data were analyzed using Clampfit 10.7 from Molecular Devices, LLC (San Jose, CA) and custom data-processing scripts written using open-source packages in the Python programming language to perform baseline adjustments, find the peak and steady state inward currents, perform monoexponential fits of photocurrent decay for off-kinetic properties, and quantify spike fidelity. Only neurons with an uncompensated series resistance between 5 and 25 MΩ, Rm>90 MΩ, and holding current >−150 pA (holding at −60 mV) were included in data analysis (FIGS.14A-B). The photocurrent amplitude was not adjusted for expression level since both expression and conductance contribute to the in vivo utility of the tool. Comparisons of expression with photocurrent strength for all ChR variants tested are included inFIGS.10A-Land11. As metrics of photocurrent strength, peak and steady-state photocurrent were used (FIG.1A). As a metric for the ChR activation spectrum, the normalized current strength induced by exposure to green light (546 nm) was used (FIG.1A). Two parameters were used to characterize ChR off-kinetics: the time to reach 50% of the light-activated current and the photocurrent decay rate, τoff(FIG.1A). AAV Production and Purification Production of recombinant AAV-PHP.eB packaging pAAV-hSyn-X-TS-eYFP-WPRE, pAAV-CAG-DIO[X-TS-eYFP]-WPRE, and pAAV-CaMKIIa-X-TS-eYFP-WPRE (X=ChR2(H134R), CoChR, ChRger1, ChRger2, and ChRger3) was done following the methods described in Deverman et al. (Nat Biotechnol 2016, 34:204-209) and Challis et al. (Nat Protoc. 2019, 14(2):379-414). Briefly, triple transfection of HEK293T cells (ATCC) was performed using polyethylenimine (PEI). Viral particles were harvested from the media and cells. Virus was then purified over iodixanol (Optiprep, Sigma; D1556) step gradients (15%, 25%, 40% and 60%). Viruses were concentrated and formulated in phosphate buffered saline (PBS). Virus titers were determined by measuring the number of DNase I-resistant viral genomes using qPCR with linearized genome plasmid as a standard. Animals Dat-Cre mice (006660) and C57Bl/6J mice (000664) were purchased from Jackson Laboratory (Bar Harbor, ME). Intravenous Injections, Stereotactic Injections, and Cannula Implantation Intravenous administration of rAAV vectors was performed by injecting the virus into the retro-orbital sinus at viral titers indicated in the text. There were no observed health issues with animals after systemic injection of virus at the titers presented in the paper. Mice remain healthy >6 months after systemic delivery of ChR2 and ChRgers. With slice electrophysiology, there was no observed indication of poor cell health due to viral-mediated expression, which was quantified by measuring the membrane resistance [Rm], leak current [holding at −60 mV], and resting membrane potential (FIGS.14A-B). Local expression in the prefrontal cortex (PFC) was achieved by direct stereotactic injection of 1 μl of purified AAV vectors at 5×1012vg ml−1targeting the following coordinates: anterior-posterior (AP), −1.7; media-lateral (ML), +/−0.5; and dorsal-ventral (DV), −2.2. For stimulation of the VTA, 300 μm outer diameter mono fiber-optic cannulae (Doric Lenses, MFC_300/330-0.37_6mm_ZF1.25_FLT) were stereotaxically implanted 200 μm above the VTA bilaterally targeted to the following coordinates: AP, −3.44 mm; ML, +/−0.48 mm; DV, 4.4 mm. For stimulation of the right secondary motor cortex (M2), 3 mm long, 400 μm mono fiber-optic cannulae (Doric Lenses, MFC_400/430-0.48_3mm_ZF1.25_FLT) were surgically secured to the surface of the skull above M2 (unilaterally) targeted to the following coordinates: AP, 1 mm; ML, 0.5 mm. The skull was thinned ˜40-50% with a standard drill to create a level surface for the fiber-skull interface. Light was delivered from either a 447 nm or 671 nm laser (Changchun New Industries [CNI] Model with PSU-H-LED) via mono fiber-optic patch cable(s) (Doric Lenses, MFP_400/430/1100-0.48_2m_FC-ZF1.25) coupled to the fiber-optic cannula(e). Fiber-optic cannulae were secured to the skull with Metabond (Parkel, SKU 5396) and dental cement. Analysis of behavioral experiments was performed using the open-source MATLAB program OptiMouse43to track mouse nose, body, and tail position while the mouse was running on the treadmill. Optogenetic intracranial self-stimulation was performed using a mouse modular test chamber (Lafayette Instruments, Model 80015NS) outfitted with an IR nose port (Model 80116TM). Gaussian Process Modeling Both the GP regression and classification modeling methods applied in this paper are based on work detailed in ref 8 and 23. For modeling, all sequences were aligned using MUltiple Sequence Comparison by Log-Expectation (MUSCLE) (ebi.ac.uk/Tools/msa/muscle/). For modeling, aligned sequences were truncated to match the length of the C1C2 sequence, eliminating N- and C-terminal fragments with poor alignment quality due to high sequence diversity. Structural encodings (i.e., the contact map) use the C1C2 crystal structure (3UG9.pdb) and assume that ChR chimeras share the contact architecture observed in the C1C2 crystal structure. Models built using structural encodings built from the ChR2 structure (6EID.pdb) and the C1Chrimson structure (5ZIH.pdb) performed as well as models using the C1C2 structure (FIGS.5C-D). The models are robust to differences in contact maps because they use both sequence and structural information, which is somewhat redundant. For a given ChR, the contact map is simply a list of contacting amino acids with their positions. For example, a contact between alanine at position 134 and methionine at position 1 of the amino acid sequence would be encoded by [(‘A134’), (‘M1’)]. Both sequence and structural information were one-hot encoded. Regression models for ChR properties were trained to predict the logarithm of the measured properties. All training data was normalized to have mean zero and standard deviation one. Gaussian process regression and classification models require kernel functions that measure the similarity between protein sequences. Learning involves optimizing the form of the kernel and its hyperparameters (Table 2). The Matérn kernel was found to be optimal for all ChR properties (Table 1). For classification model training, all 102 functionally characterized ChR variants from the recombination libraries (Table 2) were used as well as data from 61 sequence variants published by others (Dataset 1). The model was then updated with data collected from the 22 additional ChR recombination variants with high sequence diversity (˜70 mutations from the closest parent) and predicted to be functional (FIG.1D). For training the regression models, all 102 functionally characterized training sequences (Dataset 2) were initially used and then the models were updated with data collected from the 22 additional ChR variants (FIG.1D). GP Regression In regression, the goal is to infer the value of an unknown function ƒ(x) at a novel point x*given observations y at inputs X. Assuming that the observations are subject to independent and identically distributed Gaussian noise with variance σn2, the posterior distribution of ƒ*=ƒ(x*) for Gaussian process regression is Gaussian with mean ƒ*=k*T(K+σn2I)−1y(1) and variance ν*=k(x*,x*)−k*T(K+σn2I)−1k*(2) Where K is the symmetric, square covariance matrix for the training set: Kij=k(xi, xj) for xiand xjin the training set. k*is the vector of covariances between the novel input and each input in the training set, and k*i=k(x*, xi). The hyperparameters in the kernel functions and the noise hyperparameter σnwere determined by maximizing the log marginal likelihood: log⁢⁢p⁡(y|X)=-12⁢yT⁡(K+σn2⁢I)-1⁢y-12⁢log⁢K+σn2⁢I-n2⁢log⁢⁢2⁢⁢π(3) where n is the dimensionality of the inputs. Regression was implemented using open-source packages in the SciPy ecosystem. GP Classification In binary classification, instead of continuous outputs y, the outputs are class labels yi∈{+1, −1}, and the goal is to use the training data to make probabilistic predictions π(x*)=p(y*=+1|x*). Laplace's method was used to approximate the posterior distribution. Hyperparameters in the kernels are found by maximizing the marginal likelihood. Classification was implemented using open-source packages in the SciPy ecosystem. The binary classification model was trained to predict if a ChR sequence is or is not functional. A ChR sequence was considered to be functional if its photocurrents were >100 pA upon light exposure, a threshold set as an approximate lower bound for current necessary for neuronal activation. GP Kernels for Modeling Proteins Gaussian process regression and classification models require kernel functions that measure the similarity between protein sequences. A protein sequence s of length L is defined by the amino acid present at each location. This can be encoded as a binary feature vector xsethat indicates the presence or absence of each amino acid at each position resulting in a vector of length 20L (for 20 possible amino acids). Likewise, the protein's structure can be represented as a residue-residue contact map. The contact map can be encoded as a binary feature vector xstthat indicates the presence or absence of each possible contacting pair. Both the sequence and structure feature vectors were used by concatenating them to form a sequence-structure feature vector. Three types of kernel functions k(si, sj) were considered: polynomial kernels, squared exponential kernels, and Matérn kernels. These different forms represent possible functions for the protein's fitness landscape. The polynomial kernel is defined as: k(s,s′)=(σ02+σp2xTx′)d(4) where σ0and σpare hyperparameters. Polynomial kernels were considered with d=3. The squared exponential kernel is defined as: k⁡(s,s′)=σp2⁢exp⁡(-x-x′22l)(5) where l and σpare also hyperparameters and |⋅|2is the L2 norm. Finally, the Matérn kernel with ν=5/2 is defined as: k⁢(s,s′)=(1+5⁢x-x′22l+5⁢x-x′223⁢l2)⁢exp⁡(-5⁢x-x′22l)(6) Where l is once again a hyperparameter. L1 Regression Feature Identification and Weighting L1 regression was used to identify residues and contacts in the ChR structure most important for each ChR functional property of interest. First, residues and contacts that covary were identified using the concatenated sequence and structure binary feature vector for each of the training set ChR variants. Each set of covarying residues and contacts was combined into a single feature. L1 linear regression was used to select the features that contribute most to each ChR functional property of interest. The level of regularization was chosen by maximizing the log marginal likelihood of the Gaussian process regression model trained on the features selected at that level of regularization. Bayesian ridge regression was then performed on the selected features using the default settings in scikit-learn. Residues and contacts with the largest absolute Bayesian ridge linear regression weights were plotted onto the C1C2 structure (FIGS.8A-D). For feature identification and weighting, models were trained on both the training set and also the test set of 28 ChR sequences predicted to have useful combinations of diverse properties. Statistical Analysis Plotting and statistical analysis were done in Python 2.7 and 3.6 and GraphPad Prism 7.01. For statistical comparisons, a D'Agostino & Pearson normality test was first performed. If the p-value of a D'Agostino & Pearson normality test was <0.05, the non-parametric Kruskal-Wallis test with Dunn's multiple comparisons post hoc test was used. If the data passed the normality test, a one-way ANOVA was used. Dataset 1 (shown in Tables 3 and 4). ChR sequence and photocurrent data from published sources including 19 natural ChR variants, 14 point-mutant ChR variants, and 28 recombination variants from various recombination libraries. The source of the photocurrent data is included (‘Reference’). When possible, references were used with side-by-side measurements of multiple ChRs. For modeling, all sequences were aligned and truncated to match the length of the C1C2 sequence. The truncated and aligned sequences are included (‘Aligned amino acid sequence’) as well as the full-length sequence (‘Amino acid sequence’). TABLE 3ChR sequence and photocurrent data from published sources including 19 natural ChR variants,14 point-mutant ChR variants, and 28 recombination variants from various recombination libraries.Photo-SEQ IDChRcurrentAccessionNO.name(nA)ReferencecodesAmino acid sequence200PsChR10.1IndependentKF992074MTTISEVCGVWALDNPECIEVSGTNDNVKMAQLCFCMVCVCQILFMASQYPKVGopticalWEAIYLPSCECFLYGLASSGNGFIQLYDGRLIPWARYAAWICTCPSILLQINTIexcitation ofHKCKISHFNLNTFIVQADLIMNIMGVTGALTTNIAFKWIYFAIGCILFIFIVLVdistinct neuralVYDIMTSAAKEWKAKGDSKGNLVSTRLILLRWIFIVSWCVYPLLWILSPQATCApopulationsVSEDVISVAHFICDAFAKNMFGFIMWRTLWRDLDGHWDISRHYPQSSYAKDGKEEEQMTAMSQTDDTEKPHSSQG201PsChR20.12IndependentKF992056MTMLEHLEGTMDGWYAENDLGQGAIIAHWVTFFFHMITTFYLGYVSFHSKGPGGopticalKQPYFAGYHEENNIGIFVNLFAAISYFGKVVSDTHGHNYQNVGPFIIGLGNYRYexcitation ofADYMLTCPLLVMDLLFQLRAPYKITCAMLIFAVLMIGAVTNFYPGDDMKGPAVAdistinct neuralWFCFGCFWYLIAYIFMAHIVSKQYGRLDYLAHGTKAEGALFSLKLAIITFFAIWpopulationsVAFPLVWLLSVGTGVLSNEAAEICHCICDVVAKSVYGFALANFREQYDRELYGLLNSIGLDGEDVVQQLEKEMQTNHHKKKSINSPAVG202MyChR10IndependentJF922293MSPPTSPTPDTGHDTPDTGHDTGGHGAVEICFAPCEEDCVTIRYFVENDFEGCIopticalPGHFDQYSSHGSLHDIVKAALYICMVISILQILFYGFQWWRKTCGWEVWFVACIexcitation ofETSIYIIAITSEADSPFTLYLTNGQISPQLRYMEWLMTCPVILIALSNITGMAEdistinct neuralEYNKRTMTLLTSDVCCIVLGMMSAASKPRLKGILYAVGWAFGAWTYWTALQVYRpopulationsDAHKAVPKPLAWYVRAMGYVFFTSWLTFPGWFLLGPEGLEVVTGTVSTLMHACSDLISKNLWGFMDWHLRVLVARHHRKLFKAEEEHALKKGQTLEPGMPRSTSFVRGLGDDVEI203SdChR1.35IndependentKF992072MGGAPAPDAHSAPPGNDSAGGSEYHAPAGYQVNPPYHPVHGYEEQCSSIYIYYGopticalALWEQETARGFQWFAVFLSALFLAFYGWHAYKASVGWEEVYVCSVELIKVILEIexcitation ofYFEFTSPAMLFLYGGNITPWLRYAEWLLTCPVILIHLSNITGLSEEYNKRTMALdistinct neuralLVSDLGTICMGVTAALATGWVKWLFYCIGLVYGTQTFYNAGIIYVESYYIMPAGpopulationsGCKKLVLAMTAVYYSSWLMFPGLFIFGPEGMHTLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKIHEHIIMYGDIRRPVSSQFLGRKVDVLAFVTEEDKV204TcChR0.24IndependentKF992057MGWKINPLYSDEVAILEICKENEMVFGPLWEQKLARALQWFTVILSAIFLAYYVopticalYSTLRATCGWEELYVCTVEFTKVVVEVYLEYVPPFMIYQMNGQHTPWLRYMEWLexcitation ofLTCPVILIHLSNITGLNDEYSGRTMSLLTSDLGGIAFAVLSALAVGWQKGLYFGdistinct neuralIGCIYGASTFYHAACIYIESYHTMPAGKCKRLVVAMCAVFFTSWFMFPALFLAGpopulationsPECFDGLTWSGSTIAHTVADLLSKNIWGLIGHFLRVGIHEHILVHGDVRRPIEVTIFGKETSLNCFVENDDEEDDV205TsChR0.16IndependentKF992089MFAINPEYMNETVLLDECTPIYLDIGPLWEQVVARVTQWFGVILSLVFLIYYIWopticalNTYKATCGWEELYVCTVEFCKIIIELYFEYTPPAMIFQTNGQVTPWLRYAEWLLexcitation ofTCPVILIHLSNITGLNDDYSGRTMSLITSDLGGICMAVTAALSKGWLKALFFVIdistinct neuralGCGYGASTFYNAACIYIESYYTMPQGICRRLVLWMAGVFFTSWFMFPGLFLAGPpopulationsEGTQALSWAGTTIGHTVADLLSKNAWGMIGHFLRVEIHKHIIIHGDVRRPVTVKALGRQVSVNCFVDKEEEEEDERI206CbChR10IndependentKF992062MAAGLEGLVSSASRGLHASIPENPYHSDGHHLPCGLTPFGCMDDFWCNPEYGMSopticalYAGYTYCFSELAFGKLVMVPEADAGWLHSHGTQAEFVAATACQYTALSLALLLLexcitation ofSFYAYSAWKATCGWEEGYVCCVEVLFVTLEISNEFNSPATLYLSTGNYCYFLRYdistinct neuralGEWLLSCPVILIHLSNLSGLKNDYSMRTMRLLVSCIGMLITGMAGGLGVGWVKWpopulationsTLYFVSCAYSAQTYLQAAKCYVEVYATVPKGYCRTVVKLMAYAFFTAWGAYPILWAIGPEGLKYISGYSNTIAHTFCDILAKEIWTFLGHHLRIKIHEHILIHGDIRKKVQVRVAGELMNVEELMEEEGEDTV207Chrimson0.67IndependentKF992060MAELISSATRSLFAAGGINPWPNPYHHEDMGCGGMTPTGECFSTEWWCDPSYGLopticalSDAGYGYCFVEATGGYLVVGVEKKQAWLHSRGTPGEKIGAQVCQWIAFSIAIALexcitation ofLTFYGFSAWKATCGWEEVYVCCVEVLFVTLEIFKEFSSPATVYLSTGNHAYCLRdistinct neuralYFEWLLSCPVILIKLSNLSGLKNDYSKRTMGLIVSCVGMIVFGMAAGLATDWLKpopulationsWLLYIVSCIYGGYMYFQAAKCYVEANHSVPKGHCRMVVKLMAYAYFASWGSYPILWAVGPEGLLKLSPYANSIGHSICDIIAKEFWTFLAHHLRIKIHEHILIHGDIRKTTKMEIGGEEVEVEEFVEEEDEDTV208Chronos1.22IndependentKF992040METAATMTHAFISAVPSAEATIRGLLSAAAVVTPAADAHGETSNATTAGADHGCopticalFPHINHGTELQHKIAVGLQWFTVIVAIVQLIFYGWHSFKATTGWEEVYVCVIELexcitation ofVKCFIELFHEVDSPATVYQTNGGAVIWLRYSMWLLTCPVILIHLSNLTGLHEEYdistinct neuralSKRTMTILVTDIGNIVWGITAAFTKGPLKILFFMIGLFYGVTCFFQIAKVYIESpopulationsYHTLPKGVCRKICKIMAYVFFCSWLMFPVMFIAGHEGLGLITPYTSGIGHLILDLISKNTWGFLGHHLRVKIHEHILIHGDIRKTTTINVAGENMEIETFVDEEEEGGV209HdChR0.14IndependentKF992059MSVNLSLWEHGEDAGYGHWYQGTPNGTLVCSHEDNIAWLKNKGTDEEMLGANICopticalMWMAFAACLLCLSFYAYSTWRATCGWEEVYVCLVEMVKVMIEVFHENDSPATLYexcitation ofLSTGNFIMWIRYGEWLLSCPVILIHLSNITGLQDQYSKRTMQLLVSDLGTITMGdistinct neuralVTAALCGNYVKWIFFILGLCYGVNTYFHAAKVYIESYHIVPKGVCRVCVRVMAWpopulationsCFFGAWTCYPLLFVFGPEGLGVLSYNASAIGHTIIDIFSKQVWGFVGHYLRIKIHEHIVIHGNLVKPTKVKVAGMEIDAEEMVEKDEEGAI210BsChR20.68IndependentKF992034MEAYAYPELLGSAGRSLFAATVPENISESTWVDAGYQHFWTQRQNETVVCEHYTopticalHASWLISHGTKAEKTAMIACQWFAFGSAVLILLLYAWHTWKATSGWEEVYVCCVexcitation ofELVKVLFEIYHEIHHPCTLYLVTGNFILWLRYGEWLLTCPVILIHLSNITGLKNdistinct neuralDYNKRTMQLLVSDIGCVVWGVTAALCYDYKKWIFFCLGLVYGCNTYFHAAKVYIpopulationsEGYHTVPKGECRIIVKVMAGVFYCSWTLFPLLFLLGPEGTGAFSAYGSTIAHTVADVLSKQLWGLLGHHLRVKIHEHIIIHGNLTVSKKVKVAGVEVETQEMVDSTEEDAV211CnChR20.83IndependentKF992073MEPVLGLASTAVRELTAGGSGNPYESYKPPEDPCALTPFGCLTNFWCDPQFGLAopticalDAKYDYCYVKAAYGELAIVETSRLPWLYSHGSDAEHQGALAMQWMAFALCIICLexcitation ofVFYAYHSWKATTGWEEVYVCVVELVKVLLEIYKEFESPASIYLPTANAALWLRYdistinct neuralGEWLLTCPVILIHLSNITGLKDDYNKRTMQLLVSDIGCVVWGITAAFSVGWLKWpopulationsVFFVLGLLYGSNTYFHAAKVYIESYHTVPKGHCRLIVRLMAYCFYVAWTMYPILFILGPEGLGHMSAYMSTALHGVADMLSKQIWGLLGHHLRVKIFEHILIHGDIRKTTTMQVGGQMVQVEEMVDEEDEDTI212CsChR1.07IndependentKF992078MSRLVAASWLLALLLCGITSTTTASSAPAASSTDGTAAAAVSHYAMNGFDELAKopticalGAVVPEDHFVCGPADKCYCSAWLHSHGSKEEKTAFTVMQWIVFAVCIISLLFYAexcitation ofYQTWRATCGWEEVYVTIIELVHVCFGLWHEVDSPCTLYLSTGNMVLWLRYAEWLdistinct neuralLTCPVILIHLSNLTGMKNDYNKRTMALLVSDVGCIVWGTTAALSTDFVKIIFFFpopulationsLGLLYGFYTFYAAAKIYIEAYHTVPKGICRQLVRLQAYDFFFTWSMFPILFMVGPEGFGKITAYSSGIAHEVCDLLSKNLWGLMGHFIRVKIHEHILVHGNITKKTKVNVAGDMVELDTYVDQDEEHDEG213AgChR0IndependentKF992038MGTPDPLLSSIPGTDIGLGDWTEYSNYYFLNATNSTHKWVAGPEDDCFCKAWTFopticalNRGSDEESVAAFAIAWVVFSLSVLQLLYYAYAQWRSTCGWEEVYVGIIELTHICexcitation ofIAIFREFDSPAMLYLSTGNFVVWARYASWLLSCPVILIHLSNLTGMKGNYSKRTdistinct neuralMALLVSDIGTIVWGSTSAMSPHNHVKIIFFFLGLVFGLFTFYAAAKVYLEAYHTpopulationsVPKGKCRNIVRFMAWTYYVTWALFPILFILGPEGFGHITYYGSSIGHYVLEIFSKNLWSGTGHYLRLKIHEHIILHGNLTKKTKINIAGEPLEVEEYVEADDTDEGV214NsChR0.03IndependentKF992054MADFVWQGAGNGGPSAMVSHYPNGSVLLESSGSCYCEDWYTSRGNHVEHSLSNAopticalCDWFAFAISVIFLVYYAWAAFNSSVGWEEIYVCTVELIKVSIDQFLSSNSPCTLexcitation ofYLSTGNRVLWIRYGEWLLTCPVILIHLSNVTGLKDNYSKRTMALLVSDIGTIVFdistinct neuralGVTSAMCTGYPKVIFFILGCCYGANTFFNAAKVYLEAHHTLPKGSCRTLIRLMApopulationsYTYYASWGMFPILFVLGPESFGHMNMYQSNIAHTVIDLMSKNIWGMLGHFLRHKIREHILIHGDLRTTTTVNVAGEEMQVETMVAAEDADETTV215CoChR3.25IndependentKF992041MLGNGSAIVPIDQCFCLAWTDSLGSDTEQLVANILQWFAFGFSILILMFYAYQTopticalWRATCGWEEVYVCCVELTKVIIEFFHEFDDPSMLYLANGHRVQWLRYAEWLLTCexcitation ofPVILIHLSNLTGLKDDYSKRTMRLLVSDVGTIVWGATSAMSTGYVKVIFFVLGCdistinct neuralIYGANTFFHAAKVYIESYHVVPKGRPRTVVRIMAWLFFLSWGMFPVLFVVGPEGpopulationsFDAISVYGSTIGHTIIDLMSKNCWGLLGHYLRVLIHQHIIIYGDIRKKTKINVAGEEMEVETMVDQEDEETV216V2V1-0Color-tuned—MDHPVARSLIGSSYTNLNNGSIVIPSDACFCMKWLKSKGSPVALKMANALQWAA43ChRs forFALSVIILIYYAYATWRTTCGWEEVYVCCVELTKVVIEFFHEFDEPGMLYLANGmultiwavelengthNRVLWLRYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATSAoptogeneticsMCTGWTKILFFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED217V2V1-0.8Color-tuned—MDHPVARSLIGSSYTNLNNGSIVIPSDACFCMKWLKSKGSPVALKMANALQWAA25ChRs forFALSVIILIYYAYATWRTTCGWEEVYVCCVELTKVVIEFFHEFDEPGMLYLANGmultiwavelengthNRVLWLRYGEWLLTCPVILIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAoptogeneticsMCTGWTKILFFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED218V2V1-0Color-tuned—MDHPVARSLIGSSYTNLNNGSIVIPSDACFCMKWLKSKGSPVALKMANALQWAA52ChRs forFALSVIILIYYAYATWRTTCGWEEVYVCCVELTKVVIEFFHEFDEPGMLYLANGmultiwavelengthNRVLWLRYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAoptogeneticsMCTGWTKILFFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED219V2V1-0.2Color-tuned—MDHPVARSLIGSSYTNLNNGSIVIPSDACFCMKWLKSKGSPVALKMANALQWAA61ChRs forFALSVIILIYYAYATWRTTCGWEEVYVCCVELTKVVIEFFHEFDEPGMLYLANGmultiwavelengthNRVLWLRYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAoptogenetics.MSTGYIKVIFFLLGCMYGANTFFHAAKVYIESYHTVPKGLCRQLVRAMAWLFFVSWGMFPVLFLLGPEGFGHLSVYGSTIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED220V1V2-0.3Color-tuned—MDYPVARSLIVRYPTDLGNGTVCMPRGQCYCEGWLRSRGTSIEKTIAITLQWVV133ChRs forFALSVIILIYYAYATWRTTCGWEEVYVCCVELTKVVIEFFHEFDEPGMLYLANGmultiwavelengthNRVLWLRYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAoptogeneticsMSTGYIKVIFFLLGCMYGANTFFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED221VChR10.23IndependentEU622855MDYPVARSLIVRYPTDLGNGTVCMPRGQCYCEGWLRSRGTSIEKTIAITLQWVVopticalFALSVACLGWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLWLSSGexcitation ofNGVVWMRYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAdistinct neuralMCTGWTKILFFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVpopulationsAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED222V1V2-0.5Color-tuned—MDYPVARSLIVRYPTDLGNGTVCMPRGQCYCEGWLRSRGTSIEKTIAITLQWVV223ChRs forFALSVACLGWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPAMLYLANGmultiwavelengthNRVLWLRYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAoptogeneticsMSTGYIKILFFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED223V1V2-0.3Color-tuned—MDYPVARSLIVRYPTDLGNGTVCMPRGQCYCEGWLRSRGTSIEKTIAITLQWVV421ChRs forFALSVACLGWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLWLSSGmultiwavelengthNGVVWMRYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAoptogeneticsMCTGYIKVIFFLLGCMYGANTFFHAAKVYIESYHTVPKGLCRQLVRAMAWLFFVSWGMFPVLFLLGPEGFGHLSVYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED224V1V2-0Color-tuned—MDYPVARSLIVRYPTDLGNGTVCMPRGQCYCEGWLRSRGTSIEKTIAITLQWVV322ChRs forFALSVACLGWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLWLSSGmultiwavelengthNGVVWMRYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAoptogeneticsMSTGYIKVIFFLLGCMYGANTFFHAAKVYIESFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED225V1V2-0.8Color-tuned—MDYPVARSLIVRYPTDLGNGTVCMPRGQCYCEGWLRSRGTSIEKTIAITLQWVV52ChRs forFALSVACLGWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLWLSSGmultiwavelengthNGVVWMRYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAoptogeneticsMCTGWTKILFFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRAMAWLFFVSWGMFPVLFLLGPEGFGHLSVYGSTIGHTIIDLLSKNCWGLLGHFLRLKIHEHILLYGDIRKVQKIRVAGEELEVETLMTEEAPDTVKKSTA226V1V2-0Color-tuned—MDYPVARSLIVRYPTDLGNGTVCMPRGQCYCEGWLRSRGTSIEKTIAITLQWVV25ChRs forFALSVACLGWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLWLSSGmultiwavelengthNGVVWMRYGEWLLTCPVLLIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAoptogeneticsMSTGYIKVIFFLLGCMYGANTFFHAAKVYIESYHTVPKGLCRQLVRAMAWLFFVSWGMFPVLFLLGPEGFGHLSVYGSTIGHTIIDLLSKNCWGLLGHFLRLKIHEHILLYGDIRKVQKIRVAGEELEVETLMTEEAPDTVKKSTA227SFO_0.06Bi-stable—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVC128Sneural stateLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLswitchesYLATGHRVQWLRYAEWLLTSPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV228CatCh1.28Principles for—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVapplyingLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLoptogeneticYLATGHRVQWLRYAEWLLTCPVICIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWtools derivedGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAfrom directWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLcomparativeIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVanalysis ofmicrobialopsins229SFO_0.07Bi-stable—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVC128Aneural stateLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLswitchesYLATGHRVQWLRYAEWLLTAPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV230SFO_0.18Bi-stable—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVC128Tneural stateLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLswitchesYLATGHRVQWLRYAEWLLTTPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV231ChR20.77IndependentAF461397MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVopticalLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLexcitation ofYLATGHRVQWLRYAEWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWdistinct neuralGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMApopulationsWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV232TC1.43Principles for—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVapplyingLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLoptogeneticYLATGHRVQWLRYAEWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGCIVWtools derivedGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAfrom directWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLcomparativeIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVanalysis ofmicrobialopsins233ChETA_1.26Principles for—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVTCapplyingLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLoptogeneticYLATGHRVQWLRYATWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGCIVWtools derivedGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAfrom directWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLcomparativeIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVanalysis ofmicrobialopsins234ChR2_R1.03Principles for—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVapplyingLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLoptogeneticYLATGHRVQWLRYAEWLLTCPVILIRLSNLTGLSNDYSRRTMGLLVSDIGTIVWtools derivedGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAfrom directWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLcomparativeIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVanalysis ofmicrobialopsins235ChETA_0.41Principles for—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVTRapplyingLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLoptogeneticYLATGHRVQWLRYATWLLTCPVILIRLSNLTGLSNDYSRRTMGLLVSDIGTIVWtools derivedGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAfrom directWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLcomparativeIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVanalysis ofmicrobialopsins236ChETA_0.3Principles for—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVARapplyingLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLoptogeneticYLATGHRVQWLRYAAWLLTCPVILIRLSNLTGLSNDYSRRTMGLLVSDIGTIVWtools derivedGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAfrom directWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLcomparativeIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVanalysis ofmicrobialopsins237ChETA_0.39Principles for—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVAapplyingLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLoptogeneticYLATGHRVQWLRYAAWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWtools derivedGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAfrom directWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLcomparativeIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAVanalysis ofmicrobialopsins238ChETA_0.64Ultrafast—MDYGGALSAVGRELLFVTNPVVVNGSVLVPEDQCYCAGWIESRGTNGAQTASNVToptogeneticLQWLAAGFSILLLMFYAYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLcontrolYLATGHRVQWLRYATWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMATGYVKVIFFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTKLNIGGTEIEVETLVEDEAEAGAV239Abcdefg10.23Molecular—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTdeterminantsSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFdifferentiatingYGYQTWKSTCGWEEIYVCAIEMVKVILEFFFEFKNPSMLYLATGHRVQWLRYAEphotocurrentWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMATGYVKVIFproperties ofFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFItwo ChRs fromLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTchlamydomonas.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK240ChR10.08IndependentAF385748MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTopticalSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFexcitation ofYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEdistinct neuralWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFpopulationsFLMGLCYGIYTFFNAAKVYIEAYHTVPKGICRDLVRYLAWLYFCSWAMFPVLFLLGPEGFGHINQFNSAIAHAILDLASKNAWSMMGHFLRVKIHEHILLYGDIRKKQKVNVAGQEMEVETMVHEEDD241ChR_f0.17Opto-current-—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTclamp actuationSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFof corticalYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEneurons using aWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFstrategicallyFLMGLCYGIYTFFNAAKVYIEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFLdesigned ChR.LGPEGFGHINQFNSAIAHAILDLASKNAWSMMGHFLRVKIHEHILLYGDIRKKQKVNVAGQEMEVETMVHEEDD242GR0.43Principles for—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTapplyingSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFoptogeneticYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEtools derivedWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFfrom directFLMGLCYGIYTFFNAAKVYIEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFLcomparativeLGPEGFGHINQFNSAIAHAILDLASKNCWGLLGHYLRVLIHEHILIHGDIRKTTanalysis ofKLNIGGTEIEVETLVEDEAEAGAVNKGTGKmicrobialopsins'243ABCDEFg60.06Molecular—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTdeterminantsSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFdifferentiatingYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEphotocurrentWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFproperties ofFLMGLCYGIYTFFNAAKVYIEAYHTVPKGICRDLVRYLAWLYFCSWAMFPVLFLtwo ChRs fromLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTchlamydomonas.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK244ChF0.09Characterization—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTof engineeredSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFchannelrhodopsinYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEvariants withWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFimprovedFLMGLCYGIYTFFNAAKVYIEAYHTVPKGICRDLVRYLAWLYFCSWAMFPVLFLproperties andLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTkinetics.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK245ABCDEfg50.41Molecular—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTdeterminantsSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFdifferentiatingYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEphotocurrentWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFproperties ofFLMGLCYGIYTFFNAAKVYIEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFItwo ChRs fromLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTchlamydomonas.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK246ChEF1.06Characterization—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTof engineeredSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFchannelrhodopsinYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEvariants withWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFimprovedFLMGLCYGIYTFFNAAKVYIEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFIproperties andLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTkinetics.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK247ChIEF1.41Principles for—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTapplyingSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFoptogeneticYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEtools derivedWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFfrom directFLMGLCYGIYTFFNAAKVYIEAYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFIcomparativeLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTanalysis ofKLNIGGTEIEVETLVEDEAEAGAVNKGTGKmicrobialopsins'248ABCDefg40.35Molecular—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTdeterminantsSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFdifferentiatingYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEphotocurrentWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVKVIFproperties ofFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFItwo ChRs fromLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTchlamydomonas.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK249ChD1.24Characterization—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTof engineeredSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFchannelrhodopsinYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEvariants withWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGATSAMATGYVKVIFimprovedFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFIproperties andLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTkinetics.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK250ABcdefg20.41Molecular—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTdeterminantsSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFdifferentiatingYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEphotocurrentWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMATGYVKVIFproperties ofFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFItwo ChRs fromLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTchlamydomonas.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK251ABCdefg30.49Molecular—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTdeterminantsSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFdifferentiatingYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEphotocurrentWLLTCPVILIHLSNLTGLANDYSRRTMGLLVSDIGTIVWGATSAMATGYVKVIFproperties ofFCLGLCYGANTFFHAAKAYIEGYHTVPKGRCRQVVTGMAWLFFVSWGMFPILFItwo ChRs fromLGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLIHEHILIHGDIRKTTchlamydomonas.KLNIGGTEIEVETLVEDEAEAGAVNKGTGK252C1V1-0.2Color-tuned—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQT52ChRs forSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFmultiwavelengthYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEoptogenetics.WLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALSKGYVRVIFFLMGLCYGIYTFFNAAKVYIEAYHTVPKGICRDLVRVMAWTFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED253C_VChR10.07ReaChR: a red-—MVSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQshifted variantTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWVVFALSVACLGofWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLWLSSGNGVVWMRYGchannelrhodopsinEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMCTGWTKILenables deepFFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFtranscranialLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKoptogeneticQKITIAGQEMEVETLVAEEEDexcitation254bReaChES1.37ProjectionsAME16506.1MDYGGALSAVGLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANIfrom neocortexLQWVVFALSVACLGWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLmediate top-WLSSGNGVVWMRYGSWLLTCPVILIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWdown control ofGATSAMCTGWTKILFFLISLSYGMYTYFHAAKVYIEAFHTVPKGLCRQLVRAMAmemoryWLFFVSWGMFPVLFLLGPEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKretrievalIHEHILLYGDIRKKQKITIAGQEMEVETLVAEEED255ReaChR0.67ReaChR: a red-KF448069MVSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQShifted variantTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWVVFALSVACLGofWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLWLSSGNGVVWMRYGchannelrhodopsinEWLLTCPVILIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMCTGWTKILenables deepFFLISLSYGMYTYFHAAKVYIEAFHTVPKGLCRQLVRAMAWLFFVSWGMFPVLFtranscranialLLGPEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKoptogeneticQKITIAGQEMEVETLVAEEEDKYESSexcitation256VCOMET0.4ReaChR: a red-KF448070MVSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQshifted variantTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWVVFALSVACLGofWYAYQAWRATCGWEEVYVALIEMMKSIIEAFHEFDSPATLWLSSGNGVVWMRYGchannelrhodopsinEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMCTGWTKILenables deepFFLISLSYGMYTYFHAAKVYIEAFHTVPKGLCRQLVRAMAWLFFVSWGMFPVLFtranscranialLLGPEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKoptogeneticQKITIAGQEMEVETLVAEEEDKYESSexcitation257C1V1_T10.32Neocortical—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTexcitation/SYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFinhibitionYGYQTWKSTCGWETIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEbalanceWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMCTGWTKILFin informationFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLprocessing andLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQsocialKITIAGQEMEVETLVAEEEDdysfunction.258C1V10.6Neocortical—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTexcitation/SYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFinhibitionYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYAEbalanceWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMCTGWTKILFin informationFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLprocessing andLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQsocialKITIAGQEMEVETLVAEEEDdysfunction.259C1V1_TT0.78Principles for—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTapplyingSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFoptogeneticYGYQTWKSTCGWETIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYATtools derivedWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMCTGWTKILFfrom directFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLcomparativeLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQanalysis ofKITIAGQEMEVETLVAEEEDmicrobialopsins'260C1V1_T21.21Neocortical—MSRRPWLLALALAVALAAGSAGASTGSDATVPVATQDGPDYVFHRAHERMLFQTexcitation/SYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFinhibitionYGYQTWKSTCGWEEIYVATIEMIKFIIEYFHEFDEPAVIYSSNGNKTVWLRYATbalanceWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMCTGWTKILFin informationFLISLSYGMYTYFHAAKVYIEAFHTVPKGICRELVRVMAWTFFVAWGMFPVLFLprocessing andLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKIHEHILLYGDIRKKQsocialKITIAGQEMEVETLVAEEEDdysfunction. TABLE 4Alignment of truncated ChR sequencesSEQIDNO.ChR nameAligned amino acid sequence261PsChR1MTTISEVCGVWALDNPECIEVS------------GTNDNVKMAQLC----FCMVCVCQILFMASQYPKV----------GW-EAIYLPSCECFLYGLAS------SGNGFIQLYDGRLIPWA---RYAAWICTCPSILLQINTIHKCKISHFNLNIFIVQADLIMNIMGVTGALTTNIAFK---WIYFAIGCILFIFIVLVVYDIMISAAK--EWKAKGDSKGNLVSTRLILLRWIFIVSWCVYPLLWILSPQATCAVSEDVISVAHFICDAFAKNMFGFI--MWRILW-----------RDLDGHWDISRH-----YPQSSYAKDG-KEEEQMTAMSQTD262PsChR2TMLEHLEGTMD-----------------GWYAENDLG---QGAIIAHWVIFFFHMITTFYLGYVSFHSKGPGGKQPYFAGYHEENNIGIFVNLFAAISYFGKVVSDTHGHNYQNVGPFIIGLGNYRYADYMLTCPLLVMDL--LFQLRAPY-KITCAMLI--FAVLMIGAVINFYPGDDMKGPAVAWFCFGCFWYLIAYIFMAHIVSKQYGRLDYLAHGTKAEG-ALFSLKLAIIIFFAIWVAFPLVWLLSV-GTGVLSNEAAEICHCICDVVAKSVYGFALANFREQY----------------DRELYGL-----L---NSIGLD-GE--DVVQQLEKE263MITChR1EEDCVTIRYFVENDFEGCIPGH-----FDQYSSHGSLHDIVKAAL--YICMVISILQILFYGFQWWRKTC---------GW-EVWFVACIETSIYIIAITSEA--DSPFTLYLINGQISPQL---RYMEWLMTCPVILIALSNITGMAEEYNKRIMILLTSDVCCIVLGMMSAAS-KPRLK---GILYAVGWAFGAWTYWTALQVYRDAH-------KAVPKPL-AW-YVRAMGYVFFTSWLIFPGWFLLGPEGLEVVIGIVSTLMHACSDLISKNLWGFMDWHLRVLVARHHRKLFKAEEE---HALKKGQTLEPGMPRSTSFVRGLGDDVEI-------264SdChREYHAPAGYQVNPPYHPVHGYEE---QCSSIYIYYGALWEQETARGFQWFAVFLSALFLAFYGWHAYKASV---------GW-EEVYVCSVELIKVILEIYFEF--TSPAMLFLYGGNITPWL---RYAEWLLTCPVILIHLSNITGLSEEYNKRTMALLVSDLGTICMGVTAALA-TGWVK---WLFYCIGLVYGTQTFYNAGIIYVESY-------YIMPAGG-CKKLVLAMTAVYYSSWLMFPGLFIFGPEGMHTLSVAGSTIGHTIADLLSKNIWGLLGHFLRIKI-----------HE---HIIMYGD-----IRRPVSSQFL-GRKVDVLAFVTEE265TcChR-----MGWKINPLYSDEVAILE---ICKENEMVFGPLWEQKLARALQWFTVILSAIFLAYYVYSTLRATC---------GW-EELYVCTVEFTKVVVEVYLEY--VPPFMIYQMNGQHTPWL---RYMEWLLTCPVILIHLSNITGLNDEYSGRIMSLLTSDLGGIAFAVLSALA-VGWQK---GLYFGIGCIYGASTFYHAACIYIESY-------HTMPAGK- CKRLVVAMCAVFFTSWFMFPALFLAGPECFDGLIWSGSTIAHTVADLLSKNIWGLIGHFLRVGI-----------HE---HILVHGD-----VRRPIEVTIF-GKETSLNCFVEND266TsChR------MFAINPEYMNETVLLD---ECTPIYLDIGPLWEQVVARVTQWFGVILSLVFLIYYIWNTYKATC---------GW-EELYVCTVEFCKIIIELYFEY--TPPAMIFQINGQVIPWLRYAEWLLTCPVILIHLSNITGLNDDYSGRIMSLITSDLGGICMAVTAALS-KGWLK---ALFFVIGCGYGASTFYNAACIYIESY-------YTMPQGI- CRRLVLWMAGVFFTSWFMFPGLFLAGPEGTQALSWAGITIGHTVADLLSKNAWGMIGHFLRVEI-----------HK---HIIIHGD-----VRRPVTVKAL-GRQVSVNCFVDKE267CbChR1YAGYTYCFSELAFGKLVMVPEA----DAGWLHSHGTQAEFVAATACQYTALSLALLLLSFYAYSAWKATC---------GW-EEGYVCCVEVLFVTLEISNEF--NSPATLYLSIGNYCYFL---RYGEWLLSCPVILIHLSNLSGLKNDYSMRTMRLLVSCIGMLITGMAGGLG-VGWVK---WILYFVSCAYSAQTYLQAAKCYVEVY-------ATVPKGY-CRIVVKLMAYAFFTAWGAYPILWAIGPEGLKYISGYSNTIAHTFCDILAKEIWTFLGHHLRIKI-----------HE---HILIHGD-----IRKKVQVRVA-GELMNVEELMEEE268ChrimsonDAGYGYCFVEATGGYLVVGVEK----KQAWLHSRGTPGEKIGAQVCQWIAFSIAIALLIFYGFSAWKATC---------GW-EEVYVCCVEVLFVTLEIFKEF--SSPATVYLSIGNHAYCL---RYFEWLLSCPVILIKLSNLSGLKNDYSKRTMGLIVSCVGMIVFGMAAGLA-TDWLK---WLLYIVSCIYGGYMYFQAAKCYVEAN-------HSVPKGH-CRMVVKLMAYAYFASWGSYPILWAVGPEGLLKLSPYANSIGHSICDIIAKEFWTFLAHHLRIKI-----------HE---HILIHGD-----IRKTTKMEIG-GEEVEVEEFVEEE269ChronosDAHGETSNATTAGADHGCFPHI----------NHGTELQHKIAVGLQWFTVIVAIVQLIFYGWHSFKATT---------GW-EEVYVCVIELVKCFIELFHEV--DSPATVYQINGGAVIWL---RYSMWLLTCPVILIHLSNLIGLHEEYSKRTMTILVIDIGNIVWGITAAFT-KGPLK---ILFFMIGLFYGVICFFQIAKVYIESY-------HTLPKGV-CRKICKIMAYVFFCSWLMFPVMFIAGHEGLGLITPYTSGIGHLILDLISKNIWGFLGHHLRVKI-----------HE---HILIHGD-----IRKTITINVA-GENMEIETFVDEE270HdChRDAGYGHWYQGTPNGTLVCSHED----NIAWLKNKGTDEEMLGANICMWMAFAACLLCLSFYAYSTWRATC---------GW-EEVYVCLVEMVKVMIEVFHEN--DSPATLYLSIGNFIMWI---RYGEWLLSCPVILIHLSNITGLQDQYSKRTMQLLVSDLGTITMGVTAALC-GNYVK---WIFFILGLCYGVNTYFHAAKVYIESY-------HIVPKGV-CRVCVRVMAWCFFGAWTCYPLLFVFGPEGLGVLSYNASAIGHTIIDIFSKQVWGFVGHYLRIKI-----------HE---HIVIHGN-----LVKPTKVKVA-GMEIDAEEMVEKD271BsChR2DAGYQHFWTQRQNETVVCEHYT----HASWLISHGTKAEKTAMIACQWFAFGSAVLILLLYAWHTWKATS---------GW-EEVYVCCVELVKVLFEIYHEI--HHPCTLYLVIGNFILWL---RYGEWLLTCPVILIHLSNITGLKNDYNKRTMQLLVSDIGCVVWGVTAALC-YDYKK---WIFFCLGLVYGCNTYFHAAKVYIEGY-------HTVPKGE-CRIIVKVMAGVFYCSWILFPLLFLLGPEGTGAFSAYGSTIAHTVADVLSKQLWGLLGHHLRVKI-----------HE---HIIIHGN------LTVSKKVKVA-GVEVETQEMVDST272CnChR2DAKYDYCYVKAAYGELAIVETS----RLPWLYSHGSDAEHQGALAMQWMAFALCIICLVFYAYHSWKATT---------GW-EEVYVCVVELVKVLLEIYKEF--ESPASIYLPTANAALWL---RYGEWLLTCPVILIHLSNITGLKDDYNKRTMQLLVSDIGCVVWGITAAFS-VGWLK---WVFFVLGLLYGSNTYFHAAKVYIESY-------HTVPKGH-CRLIVRLMAYCFYVAWTMYPILFILGPEGLGHMSAYMSTALHGVADMLSKQIWGLLGHHLRVKI-----------FE---HILIHGD-----IRKTITMQVG-GQMVQVEEMVDEE273CsChRGFDELAKGAVVPEDHFVCGPAD-KCYCSAWLHSHGSKEEKTAFTVMQWIVFAVCIISLLFYAYQTWRATC---------GW-EEVYVTIIELVHVCFGLWHEV--DSPCTLYLSIGNMVLWL---RYAEWLLTCPVILIHLSNLIGMKNDYNKRTMALLVSDVGCIVWGITAALS-TDFVK---IIFFFLGLLYGFYIFYAAAKIYIEAY-------HTVPKGI-CRQLVRLQAYDFFFTWSMFPILFMVGPEGFGKITAYSSGIAHEVCDLLSKNLWGLMGHFIRVKI-----------HE---HILVHGN-----ITKKTKVNVA-GDMVELDTYVDQD274AgChRYSNYYFLNATNSTHKWVAGPED-DCFCKAWTENRGSDEESVAAFAIAWVVESLSVLQLLYYAYAQWRSTC---------GW-EEVYVGIIELTHICIAIFREF--DSPAMLYLSIGNEVVWA---RYASWLLSCPVILIHLSNLTGMKGNYSKRTMALLVSDIGTIVWGSTSAMSPHNHVK---IIFFELGLVEGLFTFYAAAKVYLEAY-------HTVPKGK-CRNIVRFMAWTYYVTWALFPILFILGPEGFGHITYYGSSIGHYVLEIFSKNLWSGTGHYLRLKI-----------HE---HIILHGN-----LTKKTKINIA-GEPLEVEEYVEAD275NsChRGNGGPSAMVSHYPNGSVLLESSGSCYCEDWYTSRGNHVEHSLSNACDWFAFAISVIELVYYAWAAENSSV---------GW-EEIYVCTVELIKVSIDQFLSS--NSPCTLYLSTGNRVLWI---RYGEWLLTCPVILIHLSNVTGLKDNYSKRTMALLVSDIGTIVEGVTSAMC-TGYPK---VIFFILGCCYGANTFFNAAKVYLEAH-------HTLPKGS-CRTLIRLMAYTYYASWGMFPILFVLGPESFGHMNMYQSNIAHTVIDLMSKNIWGMLGHFLRHKI-----------RE---HILIHGD-----LRTTTTVNVA-GEEMQVETMVAAE276CoChR----------MLGNGSAIVPID-QCFCLAWTDSLGSDTEQLVANILQWFAFGFSILILMFYAYQTWRATC---------GW-EEVYVCCVELTKVIIEFFHEF--DDPSMLYLANGHRVQWL---RYAEWLLTCPVILIHLSNLTGLKDDYSKRTMRLLVSDVGTIVWGATSAMS-TGYVK---VIFFVLGCIYGANTFFHAAKVYIESY-------HVVPKGR- PRIVVRIMAWLEFLSWGMFPVLEVVGPEGFDAISVYGSTIGHTIIDLMSKNCWGLLGHYLRVLI-----------HQ---HIIIYGD-----IRKKTKINVA-GEEMEVETMVDQE277V2V1-43RSLIGSSYTNLNNGSIV-IPSD-ACFCMKWLKSKGSPVALKMANALQWAAFALSVIILIYYAYATWRTTC---------GW-EEVYVCCVELTKVVIEFFHEF--DEPGMLYLANGNRVLWL---RYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE278V2V1-25RSLIGSSYTNLNNGSIV-IPSD-ACFCMKWLKSKGSPVALKMANALQWAAFALSVIILIYYAYATWRTTC---------GW-EEVYVCCVELTKVVIEFFHEF--DEPGMLYLANGNRVLWL---RYGEWLLTCPVILIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE279V2V1-52RSLIGSSYTNLNNGSIV-IPSD-ACFCMKWLKSKGSPVALKMANALQWAAFALSVIILIYYAYATWRTTC---------GW-EEVYVCCVELTKVVIEFFHEF--DEPGMLYLANGNRVLWL---RYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE280V2V1-61RSLIGSSYTNLNNGSIV-IPSD-ACFCMKWLKSKGSPVALKMANALQWAAFALSVIILIYYAYATWRTTC---------GW-EEVYVCCVELTKVVIEFFHEF--DEPGMLYLANGNRVLWL---RYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAMS-TGYIK---VIFFLLGCMYGANTFFHAAKVYIESY-------HTVPKGL-CRQLVRAMAWLFFVSWGMFPVLELLGPEGFGHLSVYGSTIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE281V1V2-133ARSLIVRYPTDLGNGTVCMPRG-QCYCEGWLRSRGTSIEKTIAITLQWVVFALSVIILIYYAYATWRTTC---------GW-EEVYVCCVELTKVVIEFFHEF--DEPGMLYLANGNRVLWL---RYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAMS-TGYIK---VIFFLLGCMYGANIFFHAAKVYIEAF-HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE282VChR1ARSLIVRYPTDLGNGTVCMPRG-QCYCEGWLRSRGTSIEKTIAITLQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE283V1V2-223ARSLIVRYPTDLGNGTVCMPRG-QCYCEGWLRSRGTSIEKTIAITLQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPAMLYLANGNRVLWL---RYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAMS-TGYIK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE284V1V2-421ARSLIVRYPTDLGNGTVCMPRG-QCYCEGWLRSRGTSIEKTIAITLQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGYIK---VIFFLLGCMYGANTFFHAAKVYIESY-------HTVPKGL-CRQLVRAMAWLFFVSWGMFPVLFLLGPEGFGHLSVYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE285V1V2-322ARSLIVRYPTDLGNGTVCMPRG-QCYCEGWLRSRGTSIEKTIAITLQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGEWLLTCPVILIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAMS-TGYIK---VIFFLLGCMYGANTFFHAAKVYIESF-------HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE286V1V2-52ARSLIVRYPTDLGNGTVCMPRG-QCYCEGWLRSRGTSIEKTIAITLQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRAMAWLFFVSWGMFPVLELLGPEGFGHLSVYGSTIGHTIIDLLSKNCWGLLGHFLRLKI-----------HE---HILLYGD-----IRKVQKIRVA-GEELEVETLMTEE287V1V2-25ARSLIVRYPTDLGNGTVCMPRG-QCYCEGWLRSRGTSIEKTIAITLQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGEWLLTCPVLLIHLSNLTGLKDDYNKRTMRLLVSDVGTIVWGATAAMS-TGYIK---VIFFLLGCMYGANTFFHAAKVYIESY-------HTVPKGL-CRQLVRAMAWLFFVSWGMFPVLELLGPEGFGHLSVYGSTIGHTIIDLLSKNCWGLLGHFLRLKI-----------HE---HILLYGD-----IRKVQKIRVA-GEELEVETLMTEE288SFO_ELLEVINPVVVNGS--VLVPED-QCYCAGWIESRGINGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-C128SEEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAEWLLTSPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE289CatChELLFVTNPVVVNGS--VLVPED-QCYCAGWIESRGTNGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAEWLLTCPVICIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE290SFO_ELLFVTNPVVVNGS--VLVPED-QCYCAGWIESRGTNGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-C128AEEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAEWLLTAPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE291SFO_ELLFVTNPVVVNGS--VLVPED-QCYCAGWIESRGTNGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-C128TEEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAEWLLTTPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE292ChR2ELLFVTNPVVVNGS--VLVPED-QCYCAGWIESRGTNGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAEWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE293TCELLFVTNPVVVNGS--VLVPED-QCYCAGWIESRGTNGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAEWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGCIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE294ChETA_TCELLFVTNPVVVNGS--VLVPED-QCYCAGWIESRGTNGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYATWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGCIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE295ChR2_RELLFVTNPVVVNGS--VLVPED-QCYCAGWIESRGTNGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAEWLLTCPVILIRLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE296ChETA_TRELLFVTNPVVVNGS--VLVPED-QCYCAGWIESRGTNGAQTASNVLQWLAAGFSILLLMFYAYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYATWLLTCPVILIRLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE297ChETA_ARELLEVINPVVVNGS--VLVPED-QCYCAGWIESRGINGAQTASNVLQWLAAGESILLLMFYAYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAAWLLTCPVILIRLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE298ChETA_AELLEVINPVVVNGS--VLVPED-QCYCAGWIESRGINGAQTASNVLQWLAAGESILLLMFYAYQTWKSTC-----GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAAWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE299ChETA_TELLEVINPVVVNGS--VLVPED-QCYCAGWIESRGINGAQTASNVLQWLAAGESILLLMFYAYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYATWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE300Abcdefg1RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITEALSALCLMFYGYQTWKSTC---------GW-EEIYVCAIEMVKVILEFFFEF--KNPSMLYLATGHRVQWL---RYAEWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPIL ILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE301ChR1RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITEALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVR---VIFFLMGLCYGIYIFFNAAKVYIEAY-------HTVPKGI-CRDLVRYLAWLYFCSWAMFPVLFLLGPEGFGHINQFNSAIAHAILDLASKNAWSMMGHFLRVKI-----------HE---HILLYGD-----IRKKQKVNVA-GQEMEVETMVHEE302ChR_fRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITEALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVR---VIFFLMGLCYGIYIFFNAAKVYIEAY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFLLGPEGFGHINQFNSAIAHAILDLASKNAWSMMGHFLRVKI-----------HE---HILLYGD-----IRKKQKVNVA-GQEMEVETMVHEE303GRRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITEALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVR---VIFFLMGLCYGIYIFFNAAKVYIEAY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFLLGPEGFGHINQFNSAIAHAILDLASKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE304ABCDEFg6RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITEALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVR---VIFFLMGLCYGIYTFFNAAKVYIEAY-------HTVPKGI-CRDLVRYLAWLYFCSWAMFPVLELLGPEGEGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE305ChFRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVR---VIFFLMGLCYGIYTFFNAAKVYIEAY-------HTVPKGI-CRDLVRYLAWLYFCSWAMFPVLELLGPEGEGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE306ABCDEfg5RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTCGW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWLRYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVR---VIFFLMGLCYGIYTFFNAAKVYIEAY-HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HEHILIHGDIRKTTKLNIG-GTEIEVETLVEDE307ChEFRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVR---VIFFLMGLCYGIYTFFNAAKVYIEAY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE--HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE308ChIEFRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVR---VIFFLMGLCYGIYTFFNAAKVYIEAY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE309ABCDefg4RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGITAALS-KGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE--HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE310ChDRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE311ABcdefg2RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLSNDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HEHILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE312ABCdefg3RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYSRRTMGLLVSDIGTIVWGATSAMA-TGYVK---VIFFCLGLCYGANTFFHAAKAYIEGY-------HTVPKGR-CRQVVTGMAWLFFVSWGMFPILFILGPEGFGVLSVYGSTVGHTIIDLMSKNCWGLLGHYLRVLI-----------HE---HILIHGD-----IRKTTKLNIG-GTEIEVETLVEDE313C1V1-52RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVILIHLSNLTGLANDYNKRTMGLLVSDIGTIVWGTTAALS-KGYVR---VIFFLMGLCYGIYTFFNAAKVYIEAY-------HTVPKGI-CRDLVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE314C_VChR1RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE315bReaChESVGLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGSWLLTCPVILIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGL-CRQLVRAMAWLFFVSWGMFPVLFLLGPEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE316ReaChRRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGEWLLTCPVILIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGL-CRQLVRAMAWLFFVSWGMFPVLFLLGPEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE317VCOMETRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWVVFALSVACLGWYAYQAWRATC---------GW-EEVYVALIEMMKSIIEAFHEF--DSPATLWLSSGNGVVWM---RYGEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGL-CRQLVRAMAWLFFVSWGMFPVLFLLGPEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HEHILLYGD-----IRKKQKITIA-GQEMEVETLVAEE318C1V1_T1RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-ETIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWTFEVAWGMFPVLELLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE319C1V1RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYAEWLLTCPVLLIHLSNLTGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWTK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWIFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE320C1V1_TTRMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-ETIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYATWLLTCPVLLIHLSNLIGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWIK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWIFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE321C1V1_T2RMLFQTSYTLENNGSVICIPNNGQCFCLAWLKSNGTNAEKLAANILQWITFALSALCLMFYGYQTWKSTC---------GW-EEIYVATIEMIKFIIEYFHEF--DEPAVIYSSNGNKTVWL---RYATWLLTCPVLLIHLSNLIGLKDDYSKRTMGLLVSDVGCIVWGATSAMC-TGWIK---ILFFLISLSYGMYTYFHAAKVYIEAF-------HTVPKGI-CRELVRVMAWIFFVAWGMFPVLFLLGTEGFGHISPYGSAIGHSILDLIAKNMWGVLGNYLRVKI-----------HE---HILLYGD-----IRKKQKITIA-GQEMEVETLVAEE Dataset 2 (shown in Tables 1 and 2). ChR variant sequences and functional properties for designed variants from our recombination libraries. Functional properties were tested in HEK cells. Measurements of peak and steady-state photocurrent (nA) with 481 nm light at 2.3 mW mm−2(“cyan_peak” & “cyan_ss”), 546 nm light at 2.8 mW mm−2(“green_peak” & “green_ss”), and 640 nm light at 2.2 mW mm−2(“red_peak” & “red_ss”) are included. The maximum peak (“max_peak”) and maximum steady-state (“max_ss”) photocurrent (nA) obtained with any wavelength are included. Measurement of the time (ms) to reach 50% of the light-exposed photocurrent after light removal is included (“kinetics_off”). The ratio of peak photocurrent with 546 nm light to maximum photocurrent was calculated per each cell and average for each ChR variant (“norm_green”). Off-kinetics (“kinetics_off”) and spectral properties (“norm_green”) were only included for ChR variants with steady-state photocurrent strength >0.02 nA. Each ChR recombination variant has a chimera identity (“block_ID”) beginning with either ‘c’ or ‘n’ to indicate the contiguous or non-contiguous library followed by 10 digits indicating the parent that contributes each of the 10 blocks (‘0’: CheRiff, ‘1’:C1C2, and ‘2’:CsChrimR). Each ChR variant's number of mutations away from the nearest parent (‘m’) is included. For modeling, all sequences were aligned and truncated to match the length of the C1C2 sequence. The truncated and aligned sequences are included (“Aligned_amino_acid_sequence”) as well as the full-length sequence (“Amino_acid_sequence”). Full sequences of non-limiting examples of ChR proteins listed in Table 1 are provided in SEQ ID NOs: 1-154, and the respective truncated and aligned sequences for those ChR proteins are provided in SEQ ID NOs: 322-475. Dataset 3. ChR variants predicted to localize and function. 1,161 ChR variants from the recombination libraries are above the 0.4 threshold for the product (‘pp’) of the predicted probabilities of localization (‘p_loc’) and function (‘p_func’). For all remaining variants (i.e., variants not yet measured), the regression models' prediction of peak photocurrent in nA (‘mu_peak_nA’), off-kinetics (time [ms] to reach 50% of the light-exposed photocurrent after light removal; ‘mu_kin_ms’), and normalized photocurrent with 546 nm light (‘mu_green’) were included. ChR variants' amino acid and nucleic acid sequences were also included. Dataset 4 (shown in Table 5). Limited set of amino acid residues and structural contacts important for model predictions identified with L1-regularized linear regression. The relative importance (‘weight’) of these sequence and structural features is learned using Bayesian ridge regression. A different limited set of features was found for each of the three functional properties of interest (‘norm_green’, ‘off_kinetics’, and ‘peak_photocurrent’). Features are either amino acid residues (i.e. a sequence feature [‘seq’]) or contacts. The feature position is indicated with numbering according to the aligned and truncated ChR sequence. The parental features were included at each position with numbering according the parental sequence. Highly-weighted features highlighted in color inFIGS.8A-Dare indicated by their corresponding color. Features not highlighted inFIGS.8A-Dare listed as gray. TABLE 5Limited set of amino acid residues and structural contacts important for model predictions identified with L1-regularized linearregressionfeatureweightstypesgroupsfeature_C1C2_adjustfeature_CheRiff_adjustfeature_CsChrim_adjustcolorproperty[‘D161’,−0.047700148contact3[‘D195’, ‘T227’][‘D220’, ‘T252’][‘C193’, ‘M225’]graynorm_green‘T197’][‘T164’,−0.047700148contact3[‘T198’, ‘T227’][‘T223’, ‘T252’][‘M196’, ‘M225’]graynorm_green‘T197’][‘T164’,−0.099968892contact14[‘T198’, ‘G220’][‘T223’, ‘G245’][‘M196’, ‘S218’]skybluenorm_green‘G190][‘T170’,−0.099968892contact14[‘T204’, ‘F216’][‘T229’, ‘F241’][‘A202’, ‘L214’]skybluenorm_green‘F186’][‘A172’,−0.22555191contact16[‘A206’, ‘F269’][‘A231’, ‘F294’][‘G204’, ‘W267’]skybluenorm_green‘F247’][‘L158’,−0.106229081contact2[‘L192’, ‘D195’][‘L217’, ‘D220’][‘I190’, ‘C193’]skybluenorm_green‘D161’]L158−0.014052219seq11L192L217I190graynorm_green[‘P134’,−0.014052219contact11[‘P168’, ‘L192’][‘P193’, ‘L217’][‘P166’, ‘I190’]graynorm_green‘L158’][‘L137’,−0.014052219contact11[‘L171’, ‘L192’][‘L196’, ‘L217’][‘L169’, ‘I190’]graynorm_green‘L158’][‘I138’,−0.014052219contact11[‘I172’, ‘L192’][‘I197’, ‘L217’][‘I170’, ‘I190]graynorm_green‘L158’][‘T154’,−0.014052219contact11[‘T188’, ‘L192’][‘T213’, ‘L217’][‘T186’, ‘I190]graynorm_green‘L158’][‘M155',−0.014052219contact11[‘M189’, ‘L192’][‘M214’, ‘L217’][‘M187’, ‘I190’]graynorm_green‘L158’][‘L157’,−0.014052219contact11[‘L191’, ‘L192’][‘L216’, ‘L217’][‘L189’, ‘I190]graynorm_green‘L158’][‘L158’,−0.014052219contact11[‘L192’, ‘V193’][‘L217’, ‘V218’][‘I190’, ‘V191’]graynorm_green‘V159’][‘L158’,−0.014052219contact11[‘L192’, ‘S194’][‘L217’, ‘S219’][‘I190’, ‘S192’]graynorm_green‘S160]G177−0.001155787seq9G210G235D208graynorm_greenV179−0.001155787seq9V212V237L210graynorm_greenF186−0.001155787seq9F216F241L214graynorm_greenG190−0.001155787seq9G220G245S218graynorm_greenL191−0.001155787seq9L221L246C219graynorm_green[‘A171’,−0.001155787contact9[‘A205’, ‘F216’][‘A230’, ‘F241’][‘A203’, ‘L214’]graynorm_green‘F186’][‘G177’−0.001155787contact9[‘G210’, ‘V212’][‘G235’, ‘V237’][‘D208’, ‘L210’]graynorm_green‘V179’][‘V179’,−0.001155787contact9[‘V212’, ‘F216’][‘V237’, ‘F241’][‘L210’, ‘L214’]graynorm_green‘F186’][‘F186’,−0.001155787contact9[‘F216’, ‘G220’][‘F241’, ‘G245’][‘L214’, ‘S218’]graynorm_green‘G190’][‘G190’,−0.001155787contact9[‘G220’, ‘L221’][‘G245’, ‘L246’][‘S218’, ‘C219’]graynorm_green‘L191’][‘G190’,−0.001155787contact9[‘G220’, ‘Y223’][‘G245’, ‘Y248’][‘S218’, ‘Y221’]graynorm_green‘Y193’][‘G190’,0.001155787contact9[‘G220’, ‘G224’][‘G245’, ‘G249’][‘S218’, ‘G222’]graynorm_green‘G194’][‘L191’,0.001155787contact9[‘L221’, ‘Y223’][‘L246’, ‘Y248’][‘C219’, ‘Y221’]graynorm_green‘Y193’][‘L191’,0.001155787contact9[‘L221’, ‘G224’][‘L246’, ‘G249’][‘C219’, ‘G222’]graynorm_green‘G194’]D1770.001155787seq19G210G235D208graynorm_greenL1790.001155787seq19V212V237L210graynorm_greenL1860.001155787seq19F216F241L214graynorm_greenI1880.001155787seq19L218C243I216graynorm_greenV1890.001155787seq19M219I244V217graynorm_greenS1900.001155787seq19G220G245S218graynorm_greenC1910.001155787seq19L221L246C219graynorm_greenI1920.001155787seq19C222V247I220graynorm_green[‘A171’,0.001155787contact19[‘A205’, ‘F216’][‘A230’, ‘F241’][‘A203’, ‘L214’]graynorm_green‘L186’][‘A174’,0.001155787contact19[‘S208’, ‘F212’][‘A233’, ‘V237’][‘A206’, ‘L210’]graynorm_green‘L179’][‘A174’,0.001155787contact19[‘S208’, ‘F216’][‘A233’, ‘F241’][‘A206’, ‘L214’]graynorm_green‘L186’][‘T176’,0.001155787contact19[‘K209’, ‘G210’][‘T234’, ‘G235’][‘T207’, ‘D208’]graynorm_green‘D177’][‘T176’,0.001155787contact19[‘K209’, ‘V212’][‘T234’, ‘V237’][‘T207’, ‘L210’]graynorm_green‘L179’][‘D177’,0.001155787contact19[‘G210’, ‘Y211’][‘G235’, ‘W236’][‘D208’, ‘W209’]graynorm_green‘W178’][‘D177’,0.001155787contact19[‘G210’, ‘V212’][‘G235’, ‘V237’][‘D208’, ‘L210’]graynorm_green‘L179’][‘D177’,0.001155787contact19[‘G210’, ‘R213’][‘G235’, ‘K238’][‘D208’, ‘K211’]graynorm_green‘K180’][‘D177’,0.001155787contact19[‘G210’, ‘V214’][‘G235’, ‘W239’][‘D208’, ‘W212’]graynorm_green‘W184’][‘W178’,0.001155787contact19[‘Y211’, ‘V212’][‘W236’, ‘V237’][‘W209’, ‘L210’]graynorm_green‘L179’][‘L179’,0.001155787contact19[‘V212’, ‘R213’][‘V237’, ‘K238’][‘L210’, ‘K211’]graynorm_green‘K180’][‘L179’,0.001155787contact19[‘V212’, ‘V214’][‘V237’, ‘W239’][‘L210’, ‘W212’]graynorm_green‘W184’][‘L179’,0.001155787contact19[‘V212’, ‘I215’][‘V237’, ‘L240’][‘L210’, ‘L213’]graynorm_green‘L185’][‘L179’,0.001155787contact19[‘V212’, ‘F216’][‘V237’, ‘F241’][‘L210’, ‘L214’]graynorm_green‘L186’][‘K180’,0.001155787contact19[‘R213’, ‘F216’][‘K238’, ‘F241’][‘K211’, ‘L214’]graynorm_green‘L186’][‘W184’,0.001155787contact19[‘V214’, ‘F216’][‘W239’, ‘F241’][‘W212’, ‘L214’]graynorm_green‘L186’][‘W184’,0.001155787contact19[‘V214’, ‘L218’][‘W239’, ‘C243’][‘W212’, ‘I216’]graynorm_green‘I188’][‘L185’,0.001155787contact19[‘I215’, ‘F216’][‘L240’, ‘F241’][‘L213’, ‘L214’]graynorm_green‘L186’][‘L185’,0.001155787contact19[‘I215’, ‘L218’][‘L240’, ‘C243’][‘L213’, ‘I216’]graynorm_green‘I188’][‘L185’,0.001155787contact19[‘I215’, ‘M219’][‘L240’, ‘I244’][‘L213’, ‘V217’]graynorm_green‘V189’][‘L186’,0.001155787contact19[‘F216’, ‘F217’][‘F241’, ‘Y242’][‘L214’, ‘Y215’]graynorm_green‘Y187’][‘L186’,0.001155787contact19[‘F216’, ‘L218’][‘F241’, ‘C243’][‘L214’, ‘I216’]graynorm_green‘I188’][‘L186’,0.001155787contact19[‘F216’, ‘M219’][‘F241’, ‘I244’][‘L214’, ‘V217’]graynorm_green‘V189’][‘L186’,0.001155787contact19[‘F216’, ‘G220’][‘F241’, ‘G245’][‘L214’, ‘S218’]graynorm_green‘S190’][‘Y187’,0.001155787contact19[‘F217’, ‘L218’][‘Y242’, ‘C243’][‘Y215’, ‘I216’]graynorm_green‘I188’][‘Y187’,0.001155787contact19[‘F217’, ‘M219’][‘Y242’, ‘I244’][‘Y215’, ‘V217’]graynorm_green‘V189’][‘Y187’,0.001155787contact19[‘F217’, ‘G220’][‘Y242’, ‘G245’][‘Y215’, ‘S218’]graynorm_green‘S190’][‘Y187’,0.001155787contact19[‘F217’, ‘L221’][‘Y242’, ‘L246’][‘Y215’, ‘C219’]graynorm_green‘C191’][‘I188’,0.001155787contact19[‘L218’, ‘M219’][‘C243’, ‘I244’][‘I216’, ‘V217’]graynorm_green‘V189’][‘I188’,0.001155787contact19[‘L218’, ‘G220’][‘C243’, ‘G245’][‘I216’, ‘S218’]graynorm_green‘S190’][‘I188’,0.001155787contact19[‘L218’, ‘L221’][‘C243’, ‘L246’][‘I216’, ‘C219’]graynorm_green‘C191’][‘I188’,0.001155787contact19[‘L218’, ‘C222’][‘C243’, ‘V247’][‘I216’, ‘I220’]graynorm_green‘I192’][‘V189’,0.001155787contact19[‘M219’, ‘G220’][‘I244’, ‘G245’][‘V217’, ‘S218’]graynorm_green‘S190’][‘V189’,0.001155787contact19[‘M219’, ‘L221’][‘I244’, ‘L246’][‘V217’, ‘C219’]graynorm_green‘C191’][‘V189’,0.001155787contact19[‘M219’, ‘C222’][‘I244’, ‘V247’][‘V217’, ‘I220’]graynorm_green‘I192’][‘V189’,0.001155787contact19[‘M219’, ‘Y223’][‘I244’, ‘Y248’][‘V217’, ‘Y221’]graynorm_green‘Y193’][‘S190’,0.001155787contact19[‘G220’, ‘L221’][‘G245’, ‘L246’][‘S218’, ‘C219’]graynorm_green‘C191’][‘S190’,0.001155787contact19[‘G220’, ‘C222’][‘G245’, ‘V247’][‘S218’, ‘I220’]graynorm_green‘I192’][‘S190’,0.001155787contact19[‘G220’, ‘Y223’][‘G245’, ‘Y248’][‘S218’, ‘Y221’]graynorm_green‘Y193’][‘S190’,0.001155787contact19[‘G220’, ‘G224’][‘G245’, ‘G249’][‘S218’, ‘G222’]graynorm_green‘G194’][‘C191’,0.001155787contact19[‘L221’, ‘C222’][‘L246’, ‘V247’][‘C219’, ‘I220’]graynorm_green‘I192’][‘C191’,0.001155787contact19[‘L221’, ‘Y223’][‘L246’, ‘Y248’][‘C219’, ‘Y221’]graynorm_green‘Y193’][‘C191’,0.001155787contact19[‘L221’, ‘G224’][‘L246’, ‘G249’][‘C219’, ‘G222’]graynorm_green‘G194’][‘I192’,0.001155787contact19[‘C222’, ‘Y223’][‘V247’, ‘Y248’][‘I220’, ‘Y221’]graynorm_green‘Y193’][‘I192’,0.001155787contact19[‘C222’, ‘G224’][‘V247’, ‘G249’][‘I220’, ‘G222’]graynorm_green‘G194’][‘F167’,0.083751234contact10[‘W201’, ‘F217’][‘M226’, ‘Y242’][‘F199’, ‘Y215’]pinknorm_green‘Y187’][‘A170’,0.083751234contact10[‘T204’, ‘S208’][‘T229’, ‘A233’][‘A202’, ‘A206’]pinknorm_green‘A174’][‘G172’,0.083751234contact10[‘A206’, ‘S208’][‘A231’, ‘A233’][‘G204’, ‘A206’]pinknorm_green‘A174’][‘G172’,0.083751234contact10[‘A206’, ‘R213’][‘A231’, ‘K238’][‘G204’, ‘K211’]pinknorm_green‘K180’][‘G202’,−0.456801278contact17[‘A232’, ‘F259’][‘G257’, ‘Y284’][‘A230’, ‘F257’]skybluenorm_green‘Y237’][‘C192’,−0.339700103contact0[‘C222’, ‘I225’][‘V247’, ‘T250’][‘I220’, ‘G223’]skybluenorm_green‘I195’]P360.002608094seq8N85L110P83graynorm_greenG370.002608094seq8A86W111G84graynorm_greenI400.002608094seq8L89E114I87graynorm_greenG410.002608094seq8A90T115G88graynorm_greenQ430.002608094seq8N92R117Q90graynorm_greenV440.002608094seq8I93G118V91graynorm_greenC450.002608094seq8L94F119C92graynorm_green[‘G34’,0.002608094contact8[‘G83’, ‘N85’][‘G108’, ‘L110’][‘G81’, ‘P83’]graynorm_green‘P36’][‘T35’,0.002608094contact8[‘T84’, ‘N85’][‘A109’, ‘L110’][‘T82’, ‘P83’]graynorm_green‘P36’][‘T35’,0.002608094contact8[‘T84’, ‘A86’][‘A109’, ‘W111’][‘T82’, ‘G84’]graynorm_green‘G37’][‘P36’,0.002608094contact8[‘N85’, ‘A86’][‘L110’, ‘W111’][‘P83’, ‘G84’]graynorm_green‘G37’][‘P36’,0.002608094contact8[‘N85’, ‘E87’][‘L110’, ‘E112’][‘P83’, ‘E85’]graynorm_green'E38’][‘P36’,0.002608094contact8[‘N85’, ‘K88’][‘L110’, ‘Q113’][‘P83’, ‘K86’]graynorm_green‘K39’][‘P36’,0.002608094contact8[‘N85’, ‘L89’][‘L110’, ‘E114’][‘P83’, ‘I87’]graynorm_green‘I40’][‘G37’,0.002608094contact8[‘A86’, ‘E87’][‘W111’, ‘E112’][‘G84’, ‘E85’]graynorm_green‘E38’][‘G37’,0.002608094contact8[‘A86’, ‘K88’][‘W111’, ‘Q113’][‘G84’, ‘K86’]graynorm_green‘K39’][‘G37’,0.002608094contact8[‘A86’, ‘L89’][‘W111’, ‘E114’][‘G84’, ‘I87’]graynorm_green‘I40’][‘G37’,0.002608094contact8[‘A86’, ‘A90’][‘W111’, ‘T115’][‘G84’, ‘G88’]graynorm_green‘G41’][‘E38’,0.002608094contact8[‘E87’, ‘L89’][‘E112’, ‘E114’][‘E85’, ‘I87’]graynorm_green‘I40’][‘E38’,0.002608094contact8[‘E87’, ‘A90’][‘E112’, ‘T115’][‘E85’, ‘G88’]graynorm_green‘G41’][‘K39’,0.002608094contact8[‘K88’, ‘L89’][‘Q113’, ‘E114’][‘K86’, ‘I87’]graynorm_green‘I40’][‘K39’,0.002608094contact8[‘K88’, ‘A90’][‘Q113’, ‘T115’][‘K86’, ‘G88’]graynorm_green‘G41’][‘K39’,0.002608094contact8[‘K88’, ‘N92’][‘Q113’, ‘R117’][‘K86’, ‘Q90’]graynorm_green‘Q43’][‘I40’,0.002608094contact8[‘L89’, ‘A90’][‘E114’, ‘T115’][‘I87’, ‘G88’]graynorm_green‘G41’][‘I40’,0.002608094contact8[‘L89’, ‘A91’][‘E114’, ‘A116’][‘I87’, ‘A89’]graynorm_green‘A42’][‘I40’,0.002608094contact8[‘L89’, ‘N92’][‘E114’, ‘R117’][‘I87’, ‘Q90’]graynorm_green‘Q43’][‘I40’,0.002608094contact8[‘L89’, ‘I93’][‘E114’, ‘G118’][‘I87’, ‘V91’]graynorm_green‘V44’][‘G41’,0.002608094contact8[‘A90’, ‘A91’][‘T115’, ‘A116’][‘G88’, ‘A89’]graynorm_green‘A42’][‘G41’,0.002608094contact8[‘A90’, ‘N92’][‘T115’, ‘R117’][‘G88’, ‘Q90’]graynorm_green‘Q43’][‘G41’,0.002608094contact8[‘A90’, ‘I93’][‘T115’, ‘G118’][‘G88’, ‘V91’]graynorm_green‘V44’][‘G41’,0.002608094contact8[‘A90’, ‘L94’][‘T115’, ‘F119’][‘G88’, ‘C92’]graynorm_green‘C45’][‘A42’,0.002608094contact8[‘A91’, ‘N92’][‘A116’, ‘R117’][‘A89’, ‘Q90’]graynorm_green‘Q43’][‘A42’,0.002608094contact8[‘A91’, ‘I93’][‘A116’, ‘G118’][‘A89’, ‘V91’]graynorm_green‘V44’][‘A42’,0.002608094contact8[‘A91’, ‘L94’][‘A116’, ‘F119’][‘A89’, ‘C92’]graynorm_green‘C45’][‘Q43’,0.002608094contact8[‘N92’, ‘I93’][‘R117’, ‘G118’][‘Q90’, ‘V91’]graynorm_green‘V44’][‘Q43’,0.002608094contact8[‘N92’, ‘L94’][‘R117’, ‘F119’][‘Q90’, ‘C92’]graynorm_green‘C45’][‘Q43’,0.002608094contact8[‘N92’, ‘Q95’][‘R117’, ‘Q120’][‘Q90’, ‘Q93’]graynorm_green‘Q46’][‘Q43’,0.002608094contact8[‘N92’, ‘W96’][‘R117’, ‘W121’][‘Q90’, ‘W94’]graynorm_green‘W47’][‘V44’,0.002608094contact8[‘I93’,‘L94’][‘G118’, ‘F119’][‘V91’, ‘C92’]graynorm_green‘C45’][‘V44’,0.002608094contact8[‘I93’, ‘Q95’][‘G118’, ‘Q120’][‘V91’, ‘Q93’]graynorm_green‘Q46’][‘V44’,0.002608094contact8[‘I93’, ‘W96’][‘G118’, ‘W121’][‘V91’, ‘W94’]graynorm_green‘W47’][‘V44’,0.002608094contact8[‘I93’, ‘I97’][‘G118’, ‘F122’][‘V91’, ‘I95’]graynorm_green‘I48’][‘C45’,0.002608094contact8[‘L94’, ‘Q95’][‘F119’, ‘Q120’][‘C92’, ‘Q93’]graynorm_green‘Q46’][‘C45’,0.002608094contact8[‘L94’, ‘W96’][‘F119’, ‘W121’][‘C92’, ‘W94’]graynorm_green‘W47’][‘C45’,0.002608094contact8[‘L94’, ‘I97’][‘F119’, ‘F122’][‘C92’, ‘I95’]graynorm_green‘I48’][‘C45’,0.002608094contact8[‘L94’, ‘I290’][‘F119’, ‘I315’][‘C92’, ‘I288’]graynorm_green‘I268’][‘M242’,−0.085205182contact1[‘M264’, ‘I267’][‘M289’, ‘G292’][‘S262’, ‘I265’]skybluenorm_green‘I245’][‘F243’,−0.085205182contact1[‘F265’, ‘I267’][‘F290’, ‘G292’][‘Y263’, ‘I265’]skybluenorm_green‘1245’][‘F48’,−0.146294383contact5[‘I97’, ‘F99’][‘F122’, ‘V124’][‘I95’, ‘F97’]skybluenorm_green‘V50’]I1580.014052219seq20L192L217I190graynorm_green[‘P134’,0.014052219contact20[‘P168’, ‘L192’][‘P193’, ‘L217’][‘P166’, ‘I190’]graynorm_green‘I158’][‘L137’,0.014052219contact20[‘L171’, ‘L192’][‘L196’, ‘L217’][‘L169’, ‘I190’]graynorm_green‘I158’][‘I138’,0.014052219contact20[‘I172’, ‘L192’][‘I197’, ‘L217’][‘I170’, ‘I190’]graynorm_green‘I158’][‘T154’,0.014052219contact20[‘T188’, ‘L192’][‘T213’, ‘L217’][‘T186’, ‘I190’]graynorm_green‘I158’][‘M155’,0.014052219contact20[‘M189’, ‘L192’][‘M214’, ‘L217’][‘M187’, ‘I190’]graynorm_green‘I158’][‘L157’,0.014052219contact20[‘L191’, ‘L192’][‘L216’, ‘L217’][‘L189’, ‘I190’]graynorm_green‘I158’][‘I158’,0.014052219contact20[‘L192’, ‘V193’][‘L217’, ‘V218’][‘I190’, ‘V191’]graynorm_green‘V159’][‘I158’,0.014052219contact20[‘L192’, ‘S194’][‘L217’, ‘S219’][‘I190’, ‘S192’]graynorm_green‘S160’][‘V50’,−0.090367708contact18[‘F99’, ‘L101’][‘V124’, ‘L126’][‘F97’, ‘I99’]skybluenorm_green‘L52’][‘V50’,−0.090367708contact18[‘F99’, ‘S102’][‘V124’, ‘S127’][‘F97’, ‘A100’]skybluenorm_green‘S53’][‘V50’,−0.090367708contact18[‘F99’, ‘A103’][‘V124’, ‘A128’][‘F97’, ‘I101’]skybluenorm_green‘A54’][‘S53’,−0.090367708contact18[‘S102’, ‘M130’][‘S127’, ‘L155’][‘A100’, ‘V128’]skybluenorm_green‘L91’][‘A54’,−0.090367708contact18[‘A103’, ‘M130’][‘A128’, ‘L155’][‘I101’, ‘V128’]skybluenorm_green‘L91’]V50−0.032549576seq21F99V124F97graynorm_greenW62−0.032549576seq21Y111W136F109graynorm_greenH63−0.032549576seq21C112H137S110graynorm_greenY65−0.032549576seq21W114Y139W112graynorm_greenS68−0.032549576seq21T117S142T115graynorm_greenV69−0.032549576seq21C118V143C116graynorm_greenS88−0.032549576seq21T127S152C125graynorm_greenL91−0.032549576seq21M130L155V128graynorm_greenY99−0.032549576seq21F138Y163F136graynorm_greenF100−0.032549576seq21H139F164K137graynorm_greenT105−0.032549576seq21D142T167S140graynorm_green[‘Q46’,−0.032549576contact21[‘Q95’, ‘F99’][‘Q120’, ‘V124’][‘Q93’, ‘F97’]graynorm_green‘V50’][‘W47’,−0.032549576contact21[‘W96’, ‘F99’][‘W121’, ‘V124’][‘W94’, ‘F97’]graynorm_green‘V50’][‘V50’,−0.032549576contact21[‘F99’, ‘M130’][‘V124’, ‘L155’][‘F97’, ‘V128’]graynorm_green‘L91’][‘V50’,−0.032549576contact21[‘F99’, ‘F133’][‘V124’, ‘V158’][‘F97’, ‘V131’]graynorm_green‘V94’][‘V50’,−0.032549576contact21[‘F99’, ‘I134’][‘V124’, ‘I159’][‘F97’, ‘T132’]graynorm_green‘195’][‘L57’,−0.032549576contact21[‘L106’, ‘T127’][‘L131’, ‘S152’][‘L104’, ‘C125’]graynorm_green‘S88’][‘L57’,−0.032549576contact21[‘L106’, ‘M130’][‘L131’, ‘L155’][‘L104’, ‘V128’]graynorm_green‘L91’][‘V69’,−0.032549576contact21[‘C118’, ‘G119’][‘V143’, ‘G144’][‘C116’, ‘G117’]graynorm_green‘G79’][‘V69’,−0.032549576contact21[‘C118’, ‘W120’][‘V143’, ‘W145’][‘C116’, W118’]graynorm_green‘W80’][‘V69’,−0.032549576contact21[‘C118’, ‘E121’][‘V143’, ‘E146’][‘C116’, ‘E119’]graynorm_green‘E82’][‘V69’,−0.032549576contact21[‘C118’, ‘R307’][‘V143’, ‘R332’][‘C116’, ‘R305’]graynorm_green‘R285’][‘V84’,−0.032549576contact21[‘I123’, ‘T127’][‘V148’, ‘S152’][‘V121’, ‘C125’]graynorm_green‘S88’][‘Y85’,−0.032549576contact21[‘Y124’, ‘T127’][‘Y149’, ‘S152’][‘Y122’, ‘C125’]graynorm_green‘S88’][‘V86’,−0.032549576contact21[‘V125’, ‘T127’][‘V150’, ‘S152’][‘V123’, ‘C125’]graynorm_green‘S88’][‘C87’,−0.032549576contact21[‘A126’, ‘T127’][‘C151’, ‘S152’][‘C124’, ‘C125’]graynorm_green‘S88’][‘C87’,−0.032549576contact21[‘A126’, ‘M130’][‘C151’, ‘L155’][‘C124’, ‘V128’]graynorm_green‘L91’][‘S88’,−0.032549576contact21[‘T127’, ‘I128’][‘S152’, ‘V153’][‘C125’, ‘V126’]graynorm_green‘V89’][‘S88’,−0.032549576contact21[‘T127’, ‘E129’][‘S152’, ‘E154’][‘C125’, ‘E127’]graynorm_green‘E90’][‘S88’,−0.032549576contact21[‘T127’, ‘M130’][‘S152’, ‘L155’][‘C125’, ‘V128’]graynorm_green‘L91’][‘S88’,−0.032549576contact21[‘T127’, ‘I131’][‘S152’, ‘I156’][‘C125’, ‘L129’]graynorm_green‘I92’][‘V89’,−0.032549576contact21[‘I128’, ‘M130’][‘V153’, ‘L155’][‘V126’, ‘V128’]graynorm_green‘L91’][‘V89’,−0.032549576contact21[‘I128’, ‘I131’][‘V153’, ‘I156’][‘V126’, ‘L129’]graynorm_green‘192’][‘V89’,−0.032549576contact21[‘I128’, ‘K132’][‘V153’, ‘K157’][‘V126’, ‘F130’]graynorm_green‘K93’][‘E90’,−0.032549576contact21[‘E129’, ‘M130’][‘E154’, ‘L155’][‘E127’, ‘V128’]graynorm_green‘L91’][‘L91’,−0.032549576contact21[‘M130’, ‘I131’][‘L155’, ‘I156’][‘V128’, ‘L129’]graynorm_green‘I92’][‘L91’,−0.032549576contact21[‘M130’, ‘K132’][‘L155’, ‘K157’][‘V128’, ‘F130’]graynorm_green‘K93’][‘L91’,−0.032549576contact21[‘M130’, ‘F133’][‘L155’, ‘V158’][‘V128’, ‘V131’]graynorm_green‘V94’][‘L91’,−0.032549576contact21[‘M130’, ‘I134’][‘L155’, ‘I159’][‘V128’, ‘T132’]graynorm_green‘I95’][‘I92’,−0.032549576contact21[‘I131’, ‘F133’][‘I156’, ‘V158’][‘L129’, ‘V131’]graynorm_green‘V94’][‘I92’,−0.032549576contact21[‘I131’, ‘I135’][‘I156’, ‘L160’][‘L129’, ‘L133’]graynorm_green‘L96’][‘K93’,−0.032549576contact21[‘K132’, ‘F133’][‘K157’, ‘V158’][‘F130’, ‘V131’]graynorm_green‘V94’][‘K93’,−0.032549576contact21[‘K132’, ‘I135’][‘K157’, ‘L160’][‘F130’, ‘L133’]graynorm_green‘L96’][‘V94’,−0.032549576contact21[‘F133’, ‘I134’][‘V158’, ‘I159’][‘V131’, ‘T132’]graynorm_green‘I95’][‘I95’,−0.032549576contact21[‘I134’, ‘I135’][‘I159’, ‘L160’][‘T132’, ‘L133’]graynorm_green‘L96’][‘I95’,−0.032549576contact21[‘I134’, ‘Y137’][‘I159’, ‘I162’][‘T132’, ‘I135’]graynorm_green‘I98’][‘I95’,−0.032549576contact21[‘I134’, ‘F138’][‘I159’, ‘Y163’][‘T132’, ‘F136’]graynorm_green‘Y99’][‘L96’,−0.032549576contact21[‘I135’, ‘F138’][‘L160’, ‘Y163’][‘L133’, ‘F136’]graynorm_green‘Y99’][‘L96’,−0.032549576contact21[‘I135’, ‘H139’][‘L160’, ‘F164’][‘L133’, ‘K137’]graynorm_green‘F100’][‘E97’,−0.032549576contact21[‘E136’, ‘F138’][‘E161’, ‘Y163’][‘E134’, ‘F136’]graynorm_green‘Y99’][‘E97’,−0.032549576contact21[‘E136’, ‘H139’][‘E161’, ‘F164’][‘E134’, ‘K137’]graynorm_green‘F100’][‘I98’,−0.032549576contact21[‘Y137’, ‘F138’][‘I162’, ‘Y163’][‘I135’, ‘F136’]graynorm_green‘Y99’][‘I98’,−0.032549576contact21[‘Y137’, ‘H139’][‘I162’, ‘F164’][‘I135’, ‘K137’]graynorm_green‘F100’][‘Y99’,−0.032549576contact21[‘F138’, ‘H139’][‘Y163’, ‘F164’][‘F136’, ‘K137’]graynorm_green‘F100’][‘Y99’,−0.032549576contact21[‘F138’, ‘E140’][‘Y163’, ‘E165’][‘F136’, ‘E138’]graynorm_green‘E101’][‘Y99’,−0.032549576contact21[‘F138’, ‘D142’][‘Y163’, ‘T167’][‘F136’, ‘S140’]graynorm_green‘T105’][‘F100’,−0.032549576contact21[‘H139’, ‘E140’][‘F164’, ‘E165’][‘K137’, ‘E138’]graynorm_green‘E101’][‘F100’,−0.032549576contact21[‘H139’, ‘F141’][‘F164’, ‘F166’][‘K137’, ‘F139’]graynorm_green‘F102’][‘F100’,−0.032549576contact21[‘H139’, ‘D142’][‘F164’, ‘T167’][‘K137’, ‘S140’]graynorm_green‘T105’][‘F100’,−0.032549576contact21[‘H139’, ‘A145’][‘F164’, ‘A170’][‘K137’, ‘A143’]graynorm_green‘A108’][‘E101’,−0.032549576contact21[‘E140’, ‘D142’][‘E165’, ‘T167’][‘E138’, ‘S140’]graynorm_green‘T105’][‘F102’,−0.032549576contact21[‘F141’, ‘D142’][‘F166’, ‘T167’][‘F139’, ‘S140’]graynorm_green‘T105’][‘T105’,−0.032549576contact21[‘D142’, ‘P144’][‘T167’, ‘P169’][‘S140’, ‘P142’]graynorm_green‘P107’][‘T105’,−0.032549576contact21[‘D142’, ‘A145’][‘T167’, ‘A170’][‘S140’, ‘A143’]graynorm_green‘A108’][‘V89’,−0.006329808contact15[‘I128’, ‘T166’][‘V153’, ‘T191’][‘V126’, ‘S164’]graynorm_green‘T132’][‘L112’,−0.1067745contact7[‘S149’, ‘N151’][‘L174’, ‘G176’][‘L147’, ‘T149’]skybluenorm_green‘G114’][‘V228’,0.042364014contact12[‘V250’, ‘L309’][‘L275’, ‘K334’][‘V248’, ‘K307’]graynorm_green‘K287’][‘V206’,0.06519277contact13[‘I236’, ‘A238’][‘V261’, ‘S263’][‘V234’, ‘A236’]pinknorm_green‘A208’][‘V206’,0.06519277contact13[‘I236’, ‘H240’][‘V261’, ‘Y265’][‘V234’, ‘H238’]pinknorm_green‘H217’][‘A49’,−0.010932852contact4[‘T98’, ‘F99’][‘A123’, ‘V124’][‘A96’, ‘F97’]graynorm_green‘F50’]F500.032549576seq6F99V124F97graynorm_greenW650.032549576seq6W114Y139W112graynorm_greenT680.032549576seq6T117S142T115graynorm_greenC690.032549576seq6C118V143C116graynorm_greenF990.032549576seq6F138Y163F136graynorm_green[‘Q46’,0.032549576contact6[‘Q95’, ‘F99’][‘Q120’, ‘V124’][‘Q93’, ‘F97’]graynorm_green‘F50’][‘W47’,0.032549576contact6[‘W96’, ‘F99’][‘W121’, ‘V124’][‘W94’, ‘F97’]graynorm_green‘F50’][‘C69’,0.032549576contact6[‘C118’, ‘G119’][‘V143’, ‘G144’][‘C116’, ‘G117’]graynorm_green‘G79’][‘C69’,0.032549576contact6[‘C118’, ‘W120’][‘V143’, ‘W145’][‘C116’, W118’]graynorm_green‘W80’][‘C69’,0.032549576contact6[‘C118’, ‘E121’][‘V143’, ‘E146’][‘C116’, ‘E119’]graynorm_green‘E82’][‘C69’,0.032549576contact6[‘C118’, ‘R307’][‘V143’, ‘R332’][‘C116’, ‘R305’]graynorm_green‘R285’][‘E97’,0.032549576contact6[‘E136’, ‘F138’][‘E161’, ‘Y163’][‘E134’, ‘F136’]graynorm_green‘F99’][‘F99’,0.032549576contact6[‘F138’, ‘E140’][‘Y163’, ‘E165’][‘F136’, ‘E138’]graynorm_green‘E101’][‘T144’,0.469988727contact14[‘T178’, ‘A181’][‘T203’, ‘S206’][‘S176’, ‘K179’]deepoliveoff_kinetics‘S147’]C1880.139320185seq5L218C243I216grayoff_kineticsI1890.139320185seq5M219I244V217grayoff_kineticsV1920.139320185seq5C222V247I220grayoff_kinetics[‘A174’,0.139320185contact5[‘S208’, ‘V212’][‘A233’, ‘V237’][‘A206’, ‘L210’]grayoff_kinetics‘V179’][‘A174’,0.139320185contact5[‘S208’, ‘F216’][‘A233’, ‘F241’][‘A206’, ‘L214’]grayoff_kinetics‘F186’][‘T176’,0.139320185contact5[‘K209’, ‘G210’][‘T234’, ‘G235’][‘T207’, ‘D208’]grayoff_kinetics‘G177’][‘T176’,0.139320185contact5[‘K209’, ‘V212’][‘T234’, ‘V237’][‘T207’, ‘L210’]grayoff_kinetics‘V179’][‘G177’,0.139320185contact5[‘G210’, ‘Y211’][‘G235’, ‘W236’][‘D208’, ‘W209’]grayoff_kinetics‘W178’][‘G177’,0.139320185contact5[‘G210’, ‘R213’][‘G235’, ‘K238’][‘D208’, ‘K211’]grayoff_kinetics‘K180’][‘G177’,0.139320185contact5[‘G210’, ‘V214’][‘G235’, ‘W239’][‘D208’, ‘W212]grayoff_kinetics‘W184’][‘W178’,0.139320185contact5[‘Y211’, ‘V212’][‘W236’, ‘V237’][‘W209’, ‘L210’]grayoff_kinetics‘V179’][‘V179’,0.139320185contact5[‘V212’, ‘R213’][‘V237’, ‘K238’][‘L210’, ‘K211’]grayoff_kinetics‘K180’][‘V179’,0.139320185contact5[‘V212’, ‘V214’][‘V237’, ‘W239’][‘L210’, ‘W212’]grayoff_kinetics‘W184’][‘V179’,0.139320185contact5[‘V212’, ‘I215’][‘V237’, ‘L240’][‘L210’, ‘L213’]grayoff_kinetics‘L185’][‘K180’,0.139320185contact5[‘R213’, ‘F216’][‘K238’, ‘F241’][‘K211’, ‘L214’]grayoff_kinetics‘F186’][‘W184’,0.139320185contact5[‘V214’, ‘F216’][‘W239’, ‘F241’][‘W212’, ‘L214’]grayoff_kinetics‘F186’][‘W184’,0.139320185contact5[‘V214’, ‘L218’][‘W239’, ‘C243’][‘W212’, ‘I216’]grayoff_kinetics‘C188’][‘L185’,0.139320185contact5[‘I215’, ‘F216’][‘L240’, ‘F241’][‘L213’, ‘L214’]grayoff_kinetics‘F186’][‘L185’,0.139320185contact5[‘I215’, ‘L218’][‘L240’, ‘C243’][‘L213’, ‘I216’]grayoff_kinetics‘C188’][‘L185’,0.139320185contact5[‘I215’, ‘M219’][‘L240’, ‘I244’][‘L213’, ‘V217’]grayoff_kinetics‘I189’][‘F186’,0.139320185contact5[‘F216’, ‘F217’][‘F241’, ‘Y242’][‘L214’, ‘Y215’]grayoff_kinetics‘Y187’][‘F186’,0.139320185contact5[‘F216’, ‘L218’][‘F241’, ‘C243’][‘L214’, ‘I216’]grayoff_kinetics‘C188’][‘F186’,0.139320185contact5[‘F216’, ‘M219’][‘F241’, ‘I244’][‘L214’, ‘V217’]grayoff_kinetics‘I189’][‘Y187’,0.139320185contact5[‘F217’, ‘L218’][‘Y242’, ‘C243’][‘Y215’, ‘I216’]grayoff_kinetics‘C188’][‘Y187’,0.139320185contact5[‘F217’, ‘M219’][‘Y242’, ‘I244’][‘Y215’, ‘V217’]grayoff_kinetics‘I189’][‘Y187’,0.139320185contact5[‘F217’, ‘G220’][‘Y242’, ‘G245’][‘Y215’, ‘S218’]grayoff_kinetics‘G190’][‘Y187’,0.139320185contact5[‘F217’, ‘L221’][‘Y242’, ‘L246’][‘Y215’, ‘C219’]grayoff_kinetics‘L191’][‘C188’,0.139320185contact5[‘L218’, ‘M219’][‘C243’, ‘I244’][‘I216’, ‘V217’]grayoff_kinetics‘I189’][‘C188’,0.139320185contact5[‘L218’, ‘G220’][‘C243’, ‘G245’][‘I216’, ‘S218’]grayoff_kinetics‘G190’][‘C188’,0.139320185contact5[‘L218’, ‘L221’][‘C243’, ‘L246’][‘I216’, ‘C219’]grayoff_kinetics‘L191’][‘C188’,0.139320185contact5[‘L218’, ‘C222’][‘C243’, ‘V247’][‘I216’, ‘I220’]grayoff_kinetics‘V192’][‘I189’,0.139320185contact5[‘M219’, ‘G220’][‘I244’, ‘G245’][‘V217’, ‘S218’]grayoff_kinetics‘G190’][‘I189’,0.139320185contact5[‘M219’, ‘L221’][‘I244’, ‘I246’][‘V217’, ‘C219’]grayoff_kinetics‘L191’][‘I189’,0.139320185contact5[‘M219’, ‘C222’][‘I244’, ‘V247’][‘V217’, ‘I220’]grayoff_kinetics‘V192’][‘I189’,0.139320185contact5[‘M219’, ‘Y223’][‘I244’, ‘Y248’][‘V217’, ‘Y221’]grayoff_kinetics‘Y193’][‘G190’0.139320185contact5[‘G220’, ‘C222’][‘G245’, ‘V247’][‘S218’, ‘I220’]grayoff_kinetics‘V192’][‘L191’,0.139320185contact5[‘L221’, ‘C222’][‘L246’, ‘V247’][‘C219’, ‘I220’]grayoff_kinetics‘V192’][‘V192’,0.139320185contact5[‘C222’, ‘Y223’][‘V247’, ‘Y248’][‘I220’, ‘Y221’]grayoff_kinetics‘Y193’][‘V192’,0.139320185contact5[‘C222’, ‘G224’][‘V247’, ‘G249’][‘I220’, ‘G222’]grayoff_kinetics‘G194’][‘D161’,−1.483821278contact13[‘D195’, ‘T227’][‘D220’, ‘T252’][‘C193’, ‘M225’]deepoliveoff_kinetics‘T197’][‘T164’,−1.483821278contact13[‘T198’, ‘T227’][‘T223’, ‘T252’][‘M196’, ‘M225’]deepoliveivoff_kinetics‘T197’][‘L191’,0.142100117contact9[‘L221’, ‘I225’][‘L246’, ‘T250’][‘C219’, ‘G223’]orangeoff_kinetics‘G195’][‘M164’,0.059247072contact6[‘T198’, ‘G220’][‘T223’, ‘G245’][‘M196’, ‘S218’]grayoff_kinetics‘G190’][‘F167’,0.059247072contact6[‘W201’, ‘F216’][‘M226’, ‘F241’][‘F199’, ‘L214’]grayoff_kinetics‘F186’][‘F167’,0.059247072contact6[‘W201’, ‘G220’][‘M226’, ‘G245’][‘F199’, ‘S218’]grayoff_kinetics‘G190’][‘A170’,0.059247072contact6[‘T204’, ‘F216’][‘T229’, ‘F241’][‘A202’, ‘L214’]grayoff_kinetics‘F186’][‘W167’,−0.248666835contact12[‘W201’, ‘F217’][‘M226’, ‘Y242’][‘F199’, ‘Y215’]grayoff_kinetics‘F187’][‘W167’,−0.248666835contact12[‘W201’, ‘M219’][‘M226’, ‘I244’][‘F199’, ‘V217’]grayoff_kinetics‘M189’][‘A53’,−0.225257299contact2[‘S102’, ‘M130’][‘S127’, ‘L155’][‘A100’, ‘V128’]grayoff_kinetics‘V91’][‘I54’,−0.225257299contact2[‘A103’, ‘M130’][‘A128’, ‘L155’][‘I101’, ‘V128’]grayoff_kinetics‘V91’][‘L235’,0.032623818contact10[‘L257’, ‘M294’][‘V282’, ‘L319’][‘A255’, ‘I292’]grayoff_kinetics‘L272’][‘L235’,0.032623818contact10[‘L257’, ‘C298’][‘V282’, ‘I323’][‘A255’, ‘F296’]grayoff_kinetics‘I276’][‘G202’,0.123927605contact8[‘A232’, ‘F259’][‘G257’, ‘Y284’][‘A230’, ‘F257’]grayoff_kinetics‘F237’][‘V238’,0.225164501contact7[‘V260’, ‘M264’][‘S285’, ‘M289’][‘A258’, ‘S262’]orangeoff_kinetics‘M242’][‘S238’,−0.560359462contact1[‘V260’, ‘M264’][‘S285’, ‘M289’][‘A258’, ‘S262’]deepoliveoff_kinetics‘M242’][‘S105’,−0.46738427contact4[‘D142’, ‘V146’][‘T167’, ‘M171’][‘S140’, ‘T144’]deepoliveoff_kinetics‘T109’][‘S105’,−0.387740793contact11[‘D142’, ‘E143’][‘T167’, ‘S168’][‘S140’, ‘S141’]grayoff_kinetics‘S106’][‘V89’,−0.357155396contact3[‘I128’, ‘T166’][‘V153’, ‘T191’][‘V126’, ‘S164’]grayoff_kinetics‘S132’][‘I204’,0.350950625contact0[‘V234’, ‘A238’][‘I259’, ‘S263’][‘C232’, ‘A236’]orangeoff_kinetics‘A208’][‘L52’,0.136551669contact7[‘L101’, ‘L293’][‘L126’, ‘L318’][‘I99’, ‘I291’]graypeak_photocurrent‘L271’][‘L52’,0.136551669contact7[‘L101’, ‘N297’][‘L126’, ‘N322’][‘I99’, ‘E295’]graypeak_photocurrent‘N275’][‘S53’,0.136551669contact7[‘S102’, ‘N297’][‘S127’, ‘N322’][‘A100’, ‘E295’]graypeak_photocurrent‘N275’][‘S105’,0.487954324contact11[‘D142’, ‘V146’][‘T167’, ‘M171’][‘S140’, ‘T144’]palegreenpeak_photocurrent‘T109’][‘N114’,−0.065994456seq6N151G176T149graypeak_photocurrent[‘N114’,−0.065994456contact6[‘N151’, ‘G152’][‘G176’, ‘G177’][‘T149’, ‘G150’]graypeak_photocurrent‘G115’][‘N114’,−0.065994456contact6[‘N151’, ‘N153’][‘G176’, ‘N178’][‘T149’, ‘N151’]graypeak_photocurrent‘N116’][‘N114’,−0.065994456contact6[‘N151’, ‘L207’][‘G176’, ‘L232’][‘T149’, ‘L205’]graypeak_photocurrent‘L173’][‘H139’,−0.021355167contact4[‘H173’, ‘G300’][‘H198’, ‘G325’][‘R171’, ‘T298’]graypeak_photocurrent‘G278’][‘H139’,−0.021355167contact4[‘H173’, ‘G303’][‘H198’, ‘G328’][‘R171’, ‘A301’]graypeak_photocurrent‘G281’][‘T118’,−0.199600713contact10[‘T155’, ‘T203’][‘T180’, ‘V228’][‘A153’, ‘M201’]graypeak_photocurrent‘M169’][‘T118’,−0.199600713contact10[‘T155’, ‘A206’][‘T180’, ‘A231’][‘A153’, ‘G204’]graypeak_photocurrent‘G172’][‘V206’,0.326253727contact0[‘I236’, ‘A238’][‘V261’, ‘S263’][‘V234’, ‘A236’]palegreenpeak_photocurrent‘A208’][‘V206’,0.326253727contact0[‘I236’, ‘H240’][‘V261’, ‘Y265’][‘V234’, ‘H238’]palegreenpeak_photocurrent‘H217’][‘L235’,−0.191853161contact2[‘L257’, ‘M294’][‘V282’, ‘L319’][‘A255’, ‘I292’]graypeak_photocurrent‘I272’][‘L235’,−0.191853161contact2[‘L257’, ‘C298’][‘V282’, ‘I323’][‘A255’, ‘F296’]graypeak_photocurrent‘F276’][‘L235’,0.112963237contact1[‘L257’, ‘M294’][‘V282’, ‘L319’][‘A255’, ‘I292’]graypeak_photocurrent‘L272’][‘L235’,0.112963237contact1[‘L257’, ‘C298’][‘V282’, ‘I323’][‘A255’, ‘F296’]graypeak_photocurrent‘I276’][‘M242’,−0.756058536contact9[‘M264’, ‘I291’][‘M289’, ‘A316’][‘S262’, ‘C289’]deeptealpeak_photocurrent‘C269’][‘F243’,−0.756058536contact9[‘F265’, ‘I291’][‘F290’, ‘A316’][‘Y263’, ‘C289’]deeptealpeak_photocurrent‘C269’][‘F243’,−0.756058536contact9[‘F265’, ‘S295’][‘F290’, ‘S320’][‘Y263’, ‘A293’]deeptealpeak_photocurrent‘A273’][‘T170’,0.245774599contact3[‘T204’, ‘S208’][‘T229’, ‘A233’][‘A202’, ‘A206’]palegreenpeak_photocurrent‘A174’][‘A172’,0.245774599contact3[‘A206’, ‘S208’][‘A231’, ‘A233’][‘G204’, ‘A206’]palegreenpeak_photocurrent‘A174’][‘A172’,0.245774599contact3[‘A206’, ‘R213’][‘A231’, ‘K238’][‘G204’, ‘K211’]palegreenpeak_photocurrent‘K180’][‘G41’,−0.349941733contact12[‘A90’, ‘V286’][‘T115’, ‘I311’][‘G88’, ‘I284’]deeptealpeak_photocurrent‘I264’][‘C45’,−0.349941733contact12[‘L94’, ‘V286’][‘F119’, ‘I311’][‘C92’, ‘I284’]deeptealpeak_photocurrent‘I264’][‘A156’,0.155622833contact5[‘G190’, ‘L192’][‘A215’, ‘L217’][‘G188’,‘I190’]palegreenpeak_photocurrent‘L158’][‘L158’,0.425036524contact8[‘L192’, ‘D195’][‘L217’, ‘D220’][‘I190’, ‘C193’]palegreenpeak_photocurrent‘D161’][‘G172'−0.377312825contact13[‘A206’, ‘F269’][‘A231’, ‘F294’][‘G204’, ‘W267’]deeptealpeak_photocurrent‘F247’] Example 1 Functional Characterization of ChR Variants for Machine Learning Structure-guided recombination were performed on three highly-functional ChR parents [CsChrimsonR (CsChrimR), C1C2, and CheRiff] by designing two 10-block recombination libraries with a theoretical size of ˜120,000 (i.e. 2×310) chimeric variants with diverse functions. 102 ChR recombinant variants were selected from these recombination libraries and used as the primary dataset for model training. This dataset was supplemented with data from other published sources including 19 ChR variants from nature, 14 single-mutant ChR variants, and 28 recombination variants from other libraries (Dataset 1). Data from other sources were used to train binary classification models for ChR function. Photocurrent strength, wavelength sensitivity and off-kinetics were used as measured properties to train machine-learning models (FIG.1A). Enhancing ChR photocurrent strength can enable reliable neuronal activation even under low-light conditions. Different off-rates can be useful for specific applications, e.g., fast off-kinetics enable high-frequency optical stimulation, slow off-kinetics is correlated with increased light sensitivity, and very slow off-kinetics can be used for constant depolarization (step-function opsins [SFOs]). In addition to opsin functional properties, optimization or maintenance of plasma-membrane localization is also advantageous for ChR function. Example 2 Training Gaussian Process (GP) Classification and Regression Models Gaussian process (GP) classification and regression models were trained using the ChR sequence/structure and functional data as inputs (FIGS.1A-F). GP models successfully predicted thermostability, substrate binding affinity, and kinetics for several soluble enzymes, and ChR membrane localization. Briefly, these models infer predictive values for new sequences from training examples by assuming that similar inputs (ChR sequence variants) will have similar outputs (photocurrent properties). To quantify the relatedness of inputs (ChR sequence variants), both sequence and structure are compared. ChR sequence information is encoded in the amino acid sequence. For structural comparisons, the 3D crystal-structural information was converted into a “contact map” that is convenient for modeling. Two residues are considered to be in contact and potentially important for structural and functional integrity if they have any non-hydrogen atoms within 4.5 Å in the C1C2 crystal structure (3UG9.pdb). The sequence and structural similarity between two variants was defined by aligning them and counting the number of positions and contacts at which they are identical. A binary classification model was trained to predict if a ChR sequence will be functional using all 102 training sequences from the recombination library (Dataset 2) as well as data from 61 variants published by others (Dataset 1). This trained classification model was then used to predict whether uncharacterized ChR sequence variants were functional (FIG.1B). To test prediction accuracy, 20-fold cross validation was performed on the training data set and achieved an area under the receiver operator curve (AUC) of 0.78, indicating good predictive power (Table 6). For Table 6, AUC or Pearson correlation was calculated after 20-fold cross validation on training set data for classification and regression models. The test set for both the classification and regression models was the 28 ChR sequences predicted to have useful combinations of diverse properties. Accuracy of model predictions on the test set is evaluated by AUC (for classification model) or Pearson correlation (for the regression models). The Matérn kernel is with ν=5/2. TABLE 6Evaluation of prediction accuracy for different ChR property models.CrossModel typeChR propertyKernelvalidationTest setGP classificationfunctionMatérnAUC = 0.78AUC = 1.0GP regressioncurrent strengthMatérnR = 0.77R = 0.92GP regressionoff-kineticsMatérnR = 0.78R = 0.97GP regressionwavelengthMatérnR = 0.89R = 0.96sensitivity Next, three regression models were trained, one for each of the ChR photocurrent properties of interest: photocurrent strength, wavelength sensitivity of photocurrents, and off-kinetics (FIG.1C). Once trained, these models were used to predict photocurrent properties of new, untested ChRs sequence variants. To test prediction accuracy, 20-fold cross validation was performed on the training dataset and observed high correlation between predicted and measured properties for all models (Pearson correlation [R] between 0.77-0.9; Tables 6 and 7). Models built using contact maps from either the ChR2 crystal structure or C1Chrimson crystal structure perform as well as models built with a contact map from the C1C2 structure (Table 8,FIGS.5C-D) even though these maps share only 82% and 89% of their contacts with the C1C2 map, respectively (FIGS.5A-B). For Table 3, Pearson correlation was calculated after 20-fold cross validation on training set data for regression models. The test set for the regression models was the 28 ChR sequences predicted to have useful combinations of diverse properties. Accuracy of model predictions on the test set is evaluated by Pearson correlation. All models use the Matérn kernel is with ν=5/2. TABLE 7GP regression model hyperparameters for each ChR propertyof interest for the Matérn kernel.NoiseLengthModel typeChR propertyhyperparameter: σn2hyperparameter: lGP regressioncurrent strength0.0484865219.65389071GP regressionoff-kinetics0.0290259719.72715834GP regressionwavelength0.1092706737.7883682sensitivity TABLE 8Comparison of prediction accuracy for different ChR propertymodels with different contact maps.Contact mapCrossstructure (pdb)ChR propertyvalidationTest setC1C2 (3UG9)current strengthR = 0.77R = 0.93off-kineticsR = 0.79R = 0.96wavelength sensitivityR = 0.90R = 0.96C1Chrimson (5ZIH)current strengthR = 0.77R = 0.94off-kineticsR = 0.79R = 0.96wavelength sensitivityR = 0.91R = 0.96ChR2 (6EID)current strengthR = 0.80R = 0.93off-kineticsR = 0.80R = 0.96wavelength sensitivityR = 0.91R = 0.96 Example 3 Selection of Engineered ChRs Using Trained Models A tiered approach was used to select ChRs predicted to have a useful combination of properties (FIG.1D). First, all ChR sequences predicted to not localize to the plasma membrane or predicted to be non-functional were eliminated. Classification models of ChR localization and function were used to predict the probability of localization and function for each ChR sequence in the 120,000-variant recombination library. Most ChR variants were predicted to not localize and not function. To focus on ChR variants predicted to localize and function, a threshold was set for the product of the predicted probabilities of localization and function (FIG.1B); any ChR sequence above that threshold were considered for the next tier of the process. A threshold of 0.4 was selected. The training data showed that the higher the mutation distance from one of the three parents, the less likely it was that a sequence would be functional; however, more diverse sequences could also offer more diverse functional properties. To explore diverse sequences predicted to function, 22 ChR variants that passed the 0.4 threshold were selected and were multi-block-swap sequences containing on average 70 mutations from the closest parent. These 22 sequences were synthesized, expressed in HEK cells, and their photocurrent properties were measured with patch-clamp electrophysiology. 59% of the tested sequences were functional (FIG.1E), compared to 38% of the multi-block swap sequences randomly selected (i.e., not selected by the model) and having comparable average mutation level. This validates the classification model's ability to make useful predictions about novel functional sequences, even for sequences that are very distant from those previously tested. The models were updated by including data from these 22 sequences for future rounds of predictions. From the 120,000-variant recombination library, 1,161 chimeric sequence variants passed the conservative 0.4 predicted localization and function threshold (FIGS.1A-F). For the second tier of the selection process, the three regression models trained on all functional variants collected up to this point were used to predict the photocurrent strength, wavelength sensitivity of photocurrents, and off-kinetics for each of these 1,161 ChR sequence variants (Dataset 3). 28 engineered ChRs predicted to be highly functional with different combinations of properties including those predicted to have the highest photocurrent strength, most red-shifted or blue-shifted activation wavelengths, and off-kinetics from very fast to very slow were selected (FIGS.6-7). Genes encoding the 28 selected engineered ChR variants were synthesized, expressed in HEK cells, and characterized for their photocurrent properties with patch-clamp electrophysiology. All 28 selected engineered ChRs were functional: 100% of variants selected using the updated classification model above the 0.4 threshold both localize and function. For each of the engineered ChR variants, the measured photocurrent properties correlated well with the model predictions (R>0.9 for all models) (FIG.1F, Table 10). This outstanding performance on a novel set of sequences demonstrated the power of the data-driven predictive method described herein for engineering engineered ChRs. As a negative control, two ChR variant sequences from the recombination library that the model predicted would be non-functional (ChR_29_10 and ChR_30_10) were selected. These sequences resulted from a single-block swap from two of the most highly functional ChR recombination variants were tested and demonstrated to be non-functional (FIG.2B), which shows that ChR functionality can be attenuated by incorporating even minimal diversity at certain positions. Example 4 Sequence and Structural Determinants of ChR Functional Properties L1-regularized linear regression models were used to identify a limited set of residues and structural contacts that strongly influence ChR photocurrent strength, spectral properties, and off-kinetics (FIG.8A). Relative importance of these sequence and structural features were assessed by weighting their contributions using L2-regularized linear regression (Dataset 4 and FIGS.8A-D). For each functional property, a set of important residues and contacts, and their respective weights were identified. A specific residue or contact at a given position was weighted as likely to lead to, e.g., low (negative weight) or high (positive weight) photocurrents. A number of residues and contacts most important for tuning spectral properties are proximal to the retinal-binding pocket, including the blue-shifting contact between A206 and F269 and the blue-shifting contact between F265 and 1267 that are conserved in the blue-shifted parents C1C2 and CheRiff while the red-shifting contact between F201 and Y217 originates from the red-shifted CsChrimR parent (FIGS.8A-D). The most heavily weighted contact contributing to off-kinetics includes the reside D195 (i.e., D156 according to ChR2 numbering) (FIGS.8A-D), a residue that is part of the DC-gate. Mutation of either the aspartic acid or cysteine within the DC-gate has been shown to decrease off-kinetic speed. While the cysteine in the DC-gate is conserved in all three ChR parents, the aspartic acid at position 195 is only conserved in CheRiff and C1C2 but not in CsChrimR, which has a cysteine at that position. Interestingly, D195 is also part of a contact with L192 that contributes strongly to photocurrent strength (FIGS.8A-D). A number of contacts proximal to retinal contribute strongly to photocurrent strength. For example, the most heavily weighted contact includes A295 (from CsChrimR), which is adjacent to the conserved lysine residue that covalently links retinal (FIGS.8A-D). This position is a serine in both C1C2 and CheRiff. Example 5 Machine-Guided Search Identifies ChRs with a Range of Useful Functional Properties Photocurrent amplitude, wavelength sensitivity, and off-kinetics of the engineered ChRs and the three parental ChRs were assessed (FIGS.2A-E). In addition to the 28 regression model-predicted ChRs, the two top-performing ChRs from the classification models' predictions (ChR_9_4 and ChR_25_9), for a total of 30 highly-functional model-predicted ChRs as well as the two negative control ChRs (ChR_29_10, ChR_30_10) were also assessed. Of the 30 model-predicted ChRs, 12 variants were found with >2-times higher blue-light activated photocurrents than the top-performing parent (CsChrimR) (FIG.2B). Three variants exhibit >1.7-times higher green-light activated photocurrents than CsChrimR. Eight variants have larger red-light activated photocurrents when compared with the blue-light activated parents (CheRiff and C1C2), though none out-perform CsChrimR. Both ChR variants predicted to be non-functional by the models produce <30 pA currents. Engineered ChRs' off-kinetics span three orders of magnitude (τoff=10 ms→10 s) (FIG.2C). This range is quite remarkable given that all engineered ChRs were built from sequence blocks of three parents that have similar off-kinetics (τoff=30-50 ms). 5 engineered ChRs were found to have faster off-kinetics than the fastest parent, while 16 have >5-times slower off-kinetics. The two fastest variants, ChR_3_10 and ChR_21_10 exhibit τoff=13±0.9 ms and 12±0.4 ms, respectively (mean±SEM). Four ChRs have particularly slow off-kinetics with τoff>1 s, including ChR_15_10, ChR_6_10, and ChR_13_10 (τoff=4.3±0.1 s, 8.0±0.5 s, and 17±7 s, respectively). Two ChRs with very large photocurrents, ChR_25_9 and ChR_11_10, exhibit τoff=220±10 ms and 330±30 ms, respectively. Three engineered ChRs exhibit interesting spectral properties (FIG.2E,FIGS.9A-B). ChR_28_10's red-shifted spectrum matches that of CsChrimR, demonstrating that incorporating sequence elements from blue-shifted ChRs into CsChrimR can still generate a red-shifted activation spectrum. Two engineered ChRs exhibit novel spectral properties: ChR_11_10 has a broad activation spectrum relative to the parental spectra, with similar steady-state current strength from 400-546 nm light and strong currents (700±100 pA) when activated with 567 nm light. ChR_25_9, on the other hand, exhibits a narrow activation spectrum relative to the parental spectra, with a peak at 481 nm light. Light sensitivity of select engineered ChRs was assessed. Compared with CsChrimR, CheRiff, and C1C2, the engineered ChRs have >9-times larger currents at the lowest intensity of light tested (10−1mW mm−2), larger currents at all intensities of light tested, and minimal decrease in photocurrent magnitude over the range of intensities tested (10−1-101mW mm−2), suggesting that photocurrents were saturated at these intensities and would only attenuate at much lower light intensities (FIG.2D). These selected engineered ChRs are expressed at levels similar to the CsChrimR parent (the highest expressing parent) indicating that the improved photocurrent strength of these ChRs is not solely due to improved expression (FIGS.10A-L,11A-C). Three of the engineered ChRs, i.e., ChR_9_4, ChR_25_9, and ChR_11_10, were compared with ChR2(H134R), an enhanced photocurrent single mutant of ChR2 commonly used for in vivo optogenetics, and CoChR (fromChloromonas oogama), reported to be one of the highest conducting ChRs activated by blue light. The selected engineered ChRs produce 3-6× larger photocurrents than ChR2(H134R) when exposed to high-intensity (2.2 mW mm2) 481 nm light and 10-18× larger photocurrents than ChR2(H134R) when exposed to low-intensity (6.5×10−2mW mm−2) 481 nm light (FIGS.12F-G). Although CoChR produced peak currents of similar magnitude to the engineered ChRs, CoChR decays to a much lower steady-state level (FIGS.12D-E) with the engineered ChRs producing 2-3× larger steady-state photocurrents than CoChR when exposed to high-intensity light and 3-4× larger steady-state photocurrents than CoChR when exposed to low-intensity light (FIGS.12F-G; Table 9). For Table 9, ChR2(H134R), n=11 cells; CoChR, n=7 cells; ChR_9_4, n=9 cells; ChR_25_9, n=12 cells; ChR_11_10, n=16 cells. The increased low-light sensitivity of these select engineered ChRs can be due to their relatively slow off-kinetics leading to the increased accumulation of the open state under low-light conditions. TABLE 9Statistical analysis of peak and steady-state photocurrent presented inFIGS. 12F-G with CoChR used as a control group for Dunn's posthoc test.Light intensity[mW mm−2]P-value peakP-value steady-stateCoChR ×2.20.220.83ChR2(H134R)CoChR × 9_42.20.160.030CoChR × 25_92.20.770.040CoChR × 11_102.20.250.014CoChR ×0.00650.370.63ChR2(H134R)CoChR × 9_40.00650.120.048CoChR × 25_90.00650.100.050CoChR × 11_100.00650.0160.0035 Example 6 Validation of Engineered ChRs for Neuroscience Applications Three of the top high-conductance ChRs, ChR_9_4, ChR_25_9, and ChR_11_10, were selected for further validation, and renamed ChRger1, ChRger2, and ChRger3, respectively, for channelrhodopsin Gaussian process-engineered recombinant opsin (FIGS.13A-B). When expressed in cultured neurons, the ChRgers display robust membrane localization and expression throughout the neuron soma and neurites (FIG.3B). The ChRgers outperformed both CoChR and ChR2(H134R) in photocurrent strength with low-intensity light in neuronal cultures (FIG.3C). The ChRgers require 1-2 orders of magnitude lower light intensity than CoChR and ChR2(H134R) for neuronal activation (FIG.3D,FIG.12H). Next, direct intracranial injections into the mouse prefrontal cortex (PFC) of rAAV-PHP.eB packaging either ChRger1-3, or ChR2(H134R) under the hSyn promoter were performed (Table 10). TABLE 10List of different constructs made for validation of the ChRgers.VectorInsert (X)Virus testedpAAV-hSyn-X-TS-eYFP-WPREhChR2(H134R)YesCoChRChRger1ChRger2ChRger3pAAV-CaMKIIa-X-TS-eYFP-WPREhChR2(H134R)YesChRger1ChRger2ChRger3pAAV-CAG-DIO[X-TS-eYFP]-WPREhChR2(H134R)YesChRger1ChRger2ChRger3 After 3-5 weeks of expression, light sensitivity in ChR-expressing neurons was measured in acute brain slices. Greater light sensitivity for the ChRgers compared with ChR2(H134R) was observed (FIGS.3G-H). The ChRgers exhibit >200 pA photocurrent at 10−3mW mm−2while at the equivalent irradiance ChR2(H134R) exhibits undetectable photocurrents. The ChRgers reach >1000 pA photocurrents with ˜10−2mW mm−2light, a four-fold improvement over ChR2(H134R)'s irradiance-matched photocurrents (FIG.3G). Example 7 Engineered ChRs and Systemic AAVs Enable Minimally-Invasive Optogenetic Excitation Light-sensitive, high-photocurrent ChRs were investigated for optogenetic activation coupled with minimally-invasive gene delivery. Previous reports of “non-invasive optogenetics” relied on invasive intracranial virus delivery, which results in many copies of virus per cell and thus very high expression levels of the injected construct. AAV capsid rAAV-PHP.eB19that produces broad transduction throughout the central nervous system with a single minimally-invasive intravenous injection in the adult mouse were described. Systemic delivery of rAAV-PHP.eB results in brain-wide transgene delivery without invasive intracranial injections. Use of rAAV-PHP.eB for optogenetic applications has been limited, however, by the low multiplicity of infection with systemically delivered viral vectors resulting in insufficient opsin expression and light-evoked currents to control neuronal firing with commonly-used channels (e.g., ChR2). As described herein, ChRgers can allow large-volume optogenetic excitation following systemic transgene delivery. rAAV-PHP.eB packaging either ChRger1, ChRger2, CoChR, or ChR2(H134R) under the hSyn promoter was systemically delivered and observed broad expression throughout the brain (FIG.3I). The fraction of opsin-expressing cells with sufficient opsin-mediated currents for light-induced firing was measured (FIG.3J). Only 4% of ChR2(H134R)-expressing neurons produced light-induced firing, while 77% of CoChR-expressing neurons, 89% of ChRger1-expressing neurons, and 100% of ChRger2- or ChRger3-expressing neurons produced light-induced activity. With systemic delivery, superior light sensitivity of ChRgers was observed compared with CoChR in both photocurrent strength (FIG.3K) and spike fidelity (FIG.3L). ChRger2-expressing neurons exhibit healthy membrane properties similar to CoChR- or ChR2(H134R)-expressing neurons both in culture and in slice (FIGS.14A-B). rAAV-PHP.eB packaging ChRger1-3 under the CaMKIIa promoter were systemically delivered. With systemic delivery of ChRger2, photocurrent strength similar to results observed after direct injection into the PFC was observed (FIG.3G). When expressed in pyramidal neurons in the cortex, ChRger2 and ChRger3 enabled robust optically-induced firing at rates between 2-10 Hz, although spike fidelity was reduced at higher frequency stimulation (FIGS.3M-N). ChRger2 performed best with higher frequency stimulation while ChRger1 performed worst. CoChR has better spike fidelity than the ChRgers at higher frequency stimulation (20-40 Hz) (FIG.3M). Optogenetic efficiency of ChRger2 was evaluated after systemic delivery using optogenetic intracranial self-stimulation (oICSS) of dopaminergic neurons of the ventral tegmental area (VTA)32. rAAV-PHP.eB packaging a double-floxed inverted open reading frame (DIO) containing either ChRger2 or ChR2(H134R) were systemically delivered into Dat-Cre mice (FIG.4A, Table 10). Three weeks after systemic delivery and stereotaxic implantation of fiber-optic cannulas above the VTA, mice were placed in an operant box and were conditioned to trigger a burst of 447 nm laser stimulation via nose poke. Animals expressing ChRger2 displayed robust optogenetic self-stimulation in a frequency-dependent and laser power-dependent manner. Higher frequencies (up to 20 Hz) and higher light power (up to 10 mW) promoted greater maximum operant response rates (FIG.4A). Conversely, laser stimulation failed to reinforce operant responding in ChR2(H134R)-expressing animals (FIG.4A); these results were consistent with results in acute slice where the light-induced currents of ChR2(H134R) were too weak at the low copy number produced by systemic delivery for robust neuronal activation. In order to determine if ChRger2 would enable both minimally-invasive transgene delivery and minimally-invasive optical excitation, directional control of locomotion was assayed in freely moving animals by optogenetic stimulation of the right secondary motor cortex (M2). In this assay, unilateral stimulation of M2 disrupts motor function in the contralateral lower extremities, causing mice to turn away from the stimulation side. rAAV-PHP.eB packaging either ChRger2 or ChR2(H134R) under a CaMKIIa promoter were systemically administered for transgene expression in excitatory pyramidal neurons in the cortex (FIG.4B, Table 10). Broad expression was observed throughout the cortex for both ChRger2 and ChR2(H134R) injected animals (FIG.15). A fiber-optic cannula guide was secured to the surface of the thinned skull above M2 without puncturing the dura and therefore leaving the brain intact (FIG.4B), which is considered to be minimally invasive. Despite the presence of the highly optically scattering calavarial bone, stimulation with 20 mW 447 nm light induced left-turning behavior in animals expressing ChRger2 but not in animals expressing ChR2(H134R) (FIG.4B). It was observed that left-turning behavior terminated upon conclusion of optical stimulation. Behavioral effects were seen at powers as low as 10 mW. To ensure that the turning behavior was not due to visual stimuli or heating caused by the stimulation laser, treadmill experiments were repeated using 671 nm light, which is outside the excitation spectrum of both opsins. 20 mW 671 nm light failed to induce turning in both ChRger2 and ChR2(H124R). Overall, these experiments demonstrated that ChRger2 is compatible with minimally-invasive systemic gene delivery and can enable minimally-invasive optogenetic excitation. Coupling ChRgers with recently reported upconversion nanoparticles may allow for non-invasive optogenetics in deep brain areas with systemic transgene delivery and tissue-penetrating near-infrared (NIR) light for neuronal excitation. As described herein, a data-driven approach was utilized herein to engineering ChR properties that enables efficient discovery of highly functional ChR variants based on data from relatively few variants. In this approach, a set of ˜120,000 chimeric ChRs was approximate and used to efficiently search sequence space and select top-performing variants for a given property. By first eliminating the vast majority of non-functional sequences, local peaks scattered throughout the landscape were focused. Then, using regression models, sequences lying on the fitness peaks were predicted. Machine learning provides a platform for simultaneous optimization of multiple ChR properties that follow engineering specifications. ChR variants with large variations in off-kinetics (10 ms to >10s) and photocurrents that far exceed any of the parental or other commonly used ChRs were generated. The machine-learning models were also used to identify the residues and contacts most important for ChR function. For example, this machine-learning pipeline (data collection from diverse sequences, model training and validation, and prediction and testing of new sequences) can be used to refine other neuroscience tools, e.g., anion-conducting ChRs, calcium sensors, voltage sensors, and AAVs. High-performance ChRs (e.g., ChRger1-3) with unprecedented light sensitivity are described herein for, e.g., in vivo optogenetics. The high-photocurrent properties of these ChRs can overcome the limitation of low per-cell copy number after systemic delivery. For example, as described herein, ChRger2 enabled neuronal excitation with high temporal precision without invasive intracranial surgery for virus delivery or fiber optic implantation for superficial brain areas, extending what is currently possible for optogenetics experiments All references cited herein, including patents, patent applications, papers, text books, and the like, and the references cited therein, to the extent that they are not already, are hereby incorporated herein by reference in their entirety. To the extent that any of the definitions or terms provided in the references incorporated by reference differ from the terms and discussion provided herein, the present terms and definitions control. The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The foregoing description and examples detail certain preferred embodiments of the invention and describe the best mode contemplated by the inventors. It will be appreciated, however, that no matter how detailed the foregoing may appear in text, the invention may be practiced in many ways and the invention should be construed in accordance with the appended claims and any equivalents thereof. In at least some of the previously described embodiments, one or more elements used in an embodiment can interchangeably be used in another embodiment unless such a replacement is not technically feasible. It will be appreciated by those skilled in the art that various other omissions, additions and modifications may be made to the methods and structures described above without departing from the scope of the claimed subject matter. All such modifications and changes are intended to fall within the scope of the subject matter, as defined by the appended claims. With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity. It will be understood by those within the art that, in general, terms used herein, and especially in the appended claims (e.g., bodies of the appended claims) are generally intended as “open” terms (e.g., the term “including” should be interpreted as “including but not limited to,” the term “having” should be interpreted as “having at least,” the term “includes” should be interpreted as “includes but is not limited to,” etc.). It will be further understood by those within the art that if a specific number of an introduced claim recitation is intended, such an intent will be explicitly recited in the claim, and in the absence of such recitation no such intent is present. For example, as an aid to understanding, the following appended claims may contain usage of the introductory phrases “at least one” and “one or more” to introduce claim recitations. However, the use of such phrases should not be construed to imply that the introduction of a claim recitation by the indefinite articles “a” or “an” limits any particular claim containing such introduced claim recitation to embodiments containing only one such recitation, even when the same claim includes the introductory phrases “one or more” or “at least one” and indefinite articles such as “a” or “an” (e.g., “a” and/or “an” should be interpreted to mean “at least one” or “one or more”); the same holds true for the use of definite articles used to introduce claim recitations. In addition, even if a specific number of an introduced claim recitation is explicitly recited, those skilled in the art will recognize that such recitation should be interpreted to mean at least the recited number (e.g., the bare recitation of “two recitations,” without other modifiers, means at least two recitations, or two or more recitations). Furthermore, in those instances where a convention analogous to “at least one of A, B, and C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “a system having at least one of A, B, and C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). In those instances where a convention analogous to “at least one of A, B, or C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “a system having at least one of A, B, or C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). It will be further understood by those within the art that virtually any disjunctive word and/or phrase presenting two or more alternative terms, whether in the description, claims, or drawings, should be understood to contemplate the possibilities of including one of the terms, either of the terms, or both terms. For example, the phrase “A or B” will be understood to include the possibilities of “A” or “B” or “A and B.” In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group. As will be understood by one skilled in the art, for any and all purposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible sub-ranges and combinations of sub-ranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into sub-ranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 articles refers to groups having 1, 2, or 3 articles. Similarly, a group having 1-5 articles refers to groups having 1, 2, 3, 4, or 5 articles, and so forth. While various aspects and embodiments have been disclosed herein, other aspects and embodiments will be apparent to those skilled in the art. The various aspects and embodiments disclosed herein are for purposes of illustration and are not intended to be limiting, with the true scope and spirit being indicated by the following claims.
250,537
11858970
DESCRIPTION OF EMBODIMENTS Solutions of the present disclosure will be explained below in connection with examples. Those skilled in the art will appreciate that the following examples are only illustrative of the present disclosure and are not to be construed as limiting the scope of the present disclosure. If no specific technology or conditions are indicated in the embodiments, the technology or conditions described in the literature in this field or the product specification shall be followed. The reagents or instruments used of which the manufacturer is not noted are conventional products commercially available. Example 1 In this example, the oligopeptide FTLE in chili pepper seeds was extracted as follows: 1) deseeding: fresh chili peppers were taken, and the flesh was separated from the seeds to obtain chili pepper seeds; 2) pulverizing: the chili pepper seeds were pulverized and sieved by an 80 mesh to obtain chili pepper seed powder ; 3) degreasing: the chili pepper seed powder was mixed with n-hexane at a ratio of 1:10 (g/ml); the mixture was stirred and degreased overnight; n-hexane was removed by suction filtration after the degreasing was completed to obtain a chili pepper seed meal; 4) protein extraction: the degreased chili pepper seed meal was dissolved in water at a ratio of 1:10 (w/v, g/mL); the pH value of the solution was adjusted to 9.5 with a NaOH solution to conduct dissolving for 4 h; then the pH value of the solution was adjusted to 4.5 with HCl to conduct precipitating for 2 h; the reaction solution was centrifuged at 8,000 rpm for 20 min, and the precipitate was collected as a crude protein extract; 5) ultra-high pressure assisted enzymolysis: the protein isolated was dissolved in water, and was subjected to an ultra-high pressure treatment at 300 MPa for 30 min; then the product obtained by the ultra-high pressure treatment was subjected to an enzymolysis treatment, in which the enzyme was Bacillus licheniformis, the mass ratio of the enzyme to the substrate was 1:20 (w/w, g/g), the temperature was 40° C., the pH value was adjusted to 8 with 1 mol/L NaOH, and the enzymolysis treatment was performed for 3 h; 6) enzyme inactivation: at the end of the enzymolysis, the enzyme was inactivated at 90° C. for 10 min to obtain a chili pepper seed zymolyte solution; 7) isolation and purification of zymolyte: the chili pepper seed zymolyte solution was passed through a DEAE anion chromatography column, where the mobile phase included deionized water and NaCl; the eluent in a periodfrom 35 min to 45 min was collected; then, isolation and purification were conducted by an ODS-A reverse phase C18 column (hydrophobic column), where the mobile phase included deionized water and 50% methanol, and the eluent in a periodfrom 75 min to 90 min was collected. The peptide fragments in the obtained eluate were subjected to mass spectrometry identification analysis, and information of multiple peptide sequences was obtained. Example 2 Chemical systhesis was conducted in accordance with the peptide sequences obtained by mass spectrometry identification analysis of Example 1 to obtain synthetic peptides. The effect of each peptide on HepG2 cell proliferation was studied, and the specific steps were as follows: 1) HepG2 cell culture: hepG2 cells were obtained from the ATCC cell bank and were cultured in a DMEM medium containing 10% FBS at 37° C. in a 5% CO2cell incubator. Cells were cultured in a 25 cm2flask, passaged when cells were grown to a density of 70% to 90%, and seeded in a 96-well plate. 2) Peptide fragment treatment: after 24 hours of cell culture in the 96-well plate, the original DMEM medium was aspirated from the wells. DMEM containing peptide fragments at concentrations of 0.1, 0.3, and 0.6 mM were added to each well to continue culturing for 24 hours. 3) Cell proliferation rate measured by MTT method: MTT at a concentration of 5 mg/mL was added to a 96-well plate in 20 μL per well. After incubation for 4 hours, the liquid was aspirated from each well. 150 μL DMSO was added to each well. The absorbance was measured after reacting for 20 min. The results are shown in the figure. It can be seen that the oligopeptide FTLE has a better HepG2 cell inhibition rate than other oligopeptides, which is helpful for the prevention or treatment of liver cancer. In the description of this specification, descriptions with reference to the terms “one embodiment”, “some embodiments”, “example”, “specific examples”, or “some examples”, etc. mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the present disclosure. In this description, schematic representations of the terms above do not necessarily refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. The different embodiments or examples and the features of the different embodiments or examples described in this description can be integrated and combined by a person skilled in the art without contradicting each other. While embodiments of the present disclosure have been shown and described, it will be understood that the above-described embodiments are illustrative and not restrictive and that changes, modifications, substitutions, and variations may be made to the embodiments by those skilled in the art without departing from the scope of the present disclosure.
5,616
11858971
The figures depict various embodiments of the present disclosure for purposes of illustration only. One skilled in the art will readily recognize from the following discussion that alternative embodiments of the structures and methods illustrated herein may be employed without departing from the principles described herein. Definitions Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this disclosure pertains. The terms “a” and “an” and “the” and similar referents as used herein refer to both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The term “about,” “approximately,” or “similar to” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which can depend in part on how the value is measured or determined, or on the limitations of the measurement system. It should be understood that all ranges and quantities described below are approximations and are not intended to limit the invention. Where ranges and numbers are used these can be approximate to include statistical ranges or measurement errors or variation. In some embodiments, for instance, measurements could be plus or minus 10%. Amino acids can be referred to by their single-letter codes or by their three-letter codes. The single-letter codes, amino acid names, and three-letter codes are as follows: G—Glycine (Gly), P—Proline (Pro), A—Alanine (Ala), V—Valine (Val), L—Leucine (Leu), I—Isoleucine (Ile), M—Methionine (Met), C—Cysteine (Cys), F—Phenylalanine (Phe), Y—Tyrosine (Tyr), W—Tryptophan (Trp), H—Histidine (His), K—Lysine (Lys), R—Arginine (Arg), Q—Glutamine (Gln), N—Asparagine (Asn), E—Glutamic Acid (Glu), D—Aspartic Acid (Asp), S—Serine (Ser), T—Threonine (Thr). The terms “including,” “includes,” “having,” “has,” “with,” or variants thereof are intended to be inclusive in a manner similar to the term “comprising”. The term “microbe” as used herein refers to a microorganism, and refers to a unicellular organism. As used herein, the term includes all bacteria, all archaea, unicellular protista, unicellular animals, unicellular plants, unicellular fungi, unicellular algae, all protozoa, and all chromista. The term “native” as used herein refers to compositions found in nature in their natural, unmodified state. The terms “optional” or “optionally” mean that the feature or structure may or may not be present, or that an event or circumstance may or may not occur, and that the description includes instances where a particular feature or structure is present and instances where the feature or structure is absent, or instances where the event or circumstance occurs and instances where the event or circumstance does not occur. The term “secreted fraction” as used herein refers to the fraction of recombinant resilins that are secreted from cells compared to the total resilins produced by the cells. The term “secretion signal” as used herein refers to a short peptide that when fused to a polypeptide mediates the secretion of that polypeptide from a cell. The term “secreted resilin coding sequence” as used herein refers to a nucleotide sequence that encodes a resilin as provided herein fused at its N-terminus to a secretion signal and optionally at its C-terminus to a tag peptide or polypeptide. The term “recombinant” as used herein in reference to a polypeptide (e.g., resilin) refers to a polypeptide that is produced in a recombinant host cell, or to a polypeptide that is synthesized from a recombinant nucleic acid. The term “recombinant host cell” as used herein refers to a host cell that comprises a recombinant nucleic acid. The term “recombinant nucleic acid” as used herein refers to a nucleic acid that is removed from its naturally occurring environment, or a nucleic acid that is not associated with all or a portion of a nucleic acid abutting or proximal to the nucleic acid when it is found in nature, or a nucleic acid that is operatively linked to a nucleic acid that it is not linked to in nature, or a nucleic acid that does not occur in nature, or a nucleic acid that contains a modification that is not found in that nucleic acid in nature (e.g., insertion, deletion, or point mutation introduced artificially, e.g., by human intervention), or a nucleic acid that is integrated into a chromosome at a heterologous site. The term includes cloned DNA isolates and nucleic acids that comprise chemically-synthesized nucleotide analog. The term “vector” as used herein refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid,” which generally refers to a circular double stranded DNA loop into which additional DNA segments can be ligated, but also includes linear double-stranded molecules such as those resulting from amplification by the polymerase chain reaction (PCR) or from treatment of a circular plasmid with a restriction enzyme. Other vectors include bacteriophages, cosmids, bacterial artificial chromosomes (BAC), and yeast artificial chromosomes (YAC). Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a cell into which they are introduced (e.g., vectors having an origin of replication that functions in the cell). Other vectors can be integrated into the genome of a cell upon introduction into the cell, and are thereby replicated along with the cell genome. The term “repeat” as used herein, in reference to an amino acid or nucleic acid sequence, refers to a sub-sequence that is present more than once in a polynucleotide or polypeptide (e.g., a concatenated sequence). A polynucleotide or polypeptide can have a direct repetition of the repeat sequence without any intervening sequence, or can have non-consecutive repetition of the repeat sequence with intervening sequences. The term “quasi-repeat” as used herein, in reference to amino acid or nucleic acid sequences, is a sub-sequence that is inexactly repeated (i.e., wherein some portion of the quasi-repeat subsequence is variable between quasi-repeats) across a polynucleotide or polypeptide. Repeating polypeptides and DNA molecules (or portions of polypeptides or DNA molecules) can be made up of either repeat sub-sequences (i.e., exact repeats) or quasi-repeat sub-sequences (i.e., inexact repeats). The term “native resilin” as used herein refers to an elastomeric polypeptide or protein produced by insects. GenBank Accession Nos. of non-limiting examples of native resilin includes the following NCBI sequence numbers: NP 995860 (Drosophila melanogaster), NP 611157 (Drosophila melanogaster), Q9V7U0 (Drosophila melanogaster), AAS64829, AAF57953 (Drosophila melanogaster), XP 001817028 (Tribolium castaneum) and XP001947408 (Acyrthosiphon pisum). The term “modified” as used herein refers to a protein or polypeptide sequence that differs in composition from a native protein or polypeptide sequence, where the functional properties are preserved to within 10% of the native protein or polypeptide properties. In some embodiments, the difference between the modified protein or polypeptide and the native protein or polypeptide can be in primary sequence (e.g., one or more amino acids are removed, inserted or substituted) or post-translation modifications (e.g., glycosylation, phosphorylation). Amino acid deletion refers to removal of one or more amino acids from a protein. Amino acid insertion refers to one or more amino acid residues being introduced in a protein or polypeptide. Amino acid insertions may comprise N-terminal and/or C-terminal fusions as well as intra-sequence insertions of single or multiple amino acids. Amino acid substitution includes non-conservative or conservative substitution, where conservative amino acid substitution tables are well known in the art (see for example Creighton (1984) Proteins. W. H. Freeman and Company (Eds)). In some embodiments, the modified protein or polypeptide and the native protein or polypeptide amino acid or nucleotide sequence identity is at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% of the amino acids or nucleotide bases. The term “truncated” as used herein refers to a protein or polypeptide sequence that is shorter in length than a native protein or polypeptide. In some embodiments, the truncated protein or polypeptide can be greater than 10%, or greater than 20%, or greater than 30%, or greater than 40%, or greater than 50%, or greater than 60%, or greater than 70%, or greater than 80%, or greater than 90% of the length of the native protein or polypeptide. The term “homolog” or “substantial similarity,” as used herein, when referring to a polypeptide, nucleic acid or fragment thereof, indicates that, when optimally aligned with appropriate amino acid or nucleotide insertions or deletions with another amino acid or nucleic acid (or its complementary strand), there is amino acid or nucleotide sequence identity in at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% of the amino acids or nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST or Gap, as discussed above. The term “resilin” as used herein refers to a protein or a polypeptide, capable of cross-linking to form an elastomer, where the protein or polypeptide is a native resilin, or a native resilin that is modified, or a native resilin that is truncated. Resilins of the present invention are preferably recombinant resilins. In some embodiments, recombinant resilins comprise a natural or modified (e.g., truncated or concatenated) nucleotide sequence coding for resilin or resilin fragments (e.g., isolated from insects), heterologously expressed and secreted from a host cell. In preferred embodiments, the secreted recombinant resilin protein is collected from a solution extracellular to the host cell. As used herein, the term “elastomer” refers to a polymer with viscoelasticity and typically weak inter-molecular forces (except for covalent cross-links between molecules, if they are present). Viscoelasticity is a property of materials that exhibit both viscous and elastic characteristics when undergoing deformation, and therefore exhibit time-dependent strain. Elasticity is associated with bond stretching along crystallographic planes in an ordered solid, and viscosity is the result of the diffusion of atoms or molecules inside an amorphous material. Elastomers that are viscoelastic, therefore, generally have low Young's modulus and high failure strain compared with other materials. Due to the viscous component of the material, viscoelastic materials dissipate energy when a load is applied and then removed. This phenomenon is observed as hysteresis in the stress-strain curve of viscoelastic materials. As a load is applied there is a particular stress-strain curve, and as the load is removed the stress-strain curve upon unloading is different than that of the curve during loading. The energy dissipated is the area between the loading and unloading curves. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value inclusively falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. DETAILED DESCRIPTION Provided herein are compositions comprising recombinant resilins, and methods for their production. Resilins have many unique properties compared to petroleum-based elastomers. Most notably, resilin has an extreme elastic efficiency, where very little of the energy input into deformation is lost as heat. Other desirable properties of resilin include, for example, desirable resilience, compressive elastic modulus, tensile elastic modulus, shear modulus, extension to break, maximum tensile strength, hardness, rebound, and compression set. Moreover, resilin is a protein, and therefore can be biodegraded, which makes it more environmentally friendly than petroleum-based polymers. Also, resilin is biocompatible and can therefore be used in applications that involve contact with humans or animals. Lastly, the mechanical properties of recombinant resilins can be tuned through varying protein sequence, protein structure, amount of intermolecular cross-linking and processing variables to produce elastomers designed for a universe of specific applications. In some embodiments, the methods and compositions provided herein provide efficient means for producing large quantities of recombinant resilins. In some embodiments, large quantities of resilin and resilin-like polypeptides are obtained using recombinant host cells that secrete recombinant resilins via their secretory pathways. Such secretion of recombinant resilins a) avoids toxicity from intracellular accumulation of recombinant resilins, b) simplifies purification by eliminating cell disruption or protein refolding processes, and c) provides opportunities for post-translational events (e.g., proteolytic maturation, glycosylation, disulfide bond formation) that can modulate the properties of the recombinant resilins. Compositions Comprising Recombinant Resilins In some embodiments, the compositions provided herein comprise recombinant resilins. FIG.1illustrates an example of a native resilin, which contains an N-terminal A-domain comprising a plurality of repeat units comprising the consensus amino acid sequence YGXP (“A-repeat”), where X is any amino acid; a chitin-binding type RR-2 (C) domain (Pfam reference PF00379; Rebers J E & Willis, J H. A conserved domain in anthropod cuticular proteins binds chitin. Insect Biochem Mol Biol 31:1083-1093); and a C-terminal B-domain comprising a plurality of repeat units comprising the consensus amino acid sequence UYZXZ (“B-repeat”), where U is glycine or serine; Z is serine, glycine, arginine, or proline; and X is any amino acid. Not all naturally occurring resilins have A-, C-, and B-domains. Native resilins produced by various insects typically have inexact repeats (i.e., quasi-repeats) within the A- and/or B-domains with some amino acid variation between the quasi-repeats. In some embodiments, the recombinant resilins provided herein comprise one or more A-repeats. In some embodiments, the recombinant resilins comprise N-terminal A-domains comprising a plurality of blocks of A-repeat and/or quasi-A-repeat amino acid sub-sequences each with the consensus sequence SXXYGXP, where S is serine, X is an amino acid, Y is tyrosine, G is glycine, and P is proline. In some embodiments, the recombinant resilins provided herein comprise one or more B-repeats. In some embodiments, the recombinant resilins comprise a C-terminal B-domain comprising a plurality of blocks of B-repeat and/or quasi-B-repeat amino acid sub-sequences each with the consensus sequence GYZXZZX and/or SYZXZZX, where G is glycine; Y is tyrosine; Z is serine, glycine, proline, or arginine; S is serine; and X is any amino acid. In some embodiments, the recombinant resilins provided herein comprise one or more A-repeats. In some such embodiments, the recombinant resilins comprise between 1 and 100 A-repeats, or from 2 to 50 A-repeats, or from 5 to 50 A-repeats, or from 5 to 20 A-repeats. In some embodiments, the recombinant resilins comprise one or more consensus sequences described by the formula, (X1—X2—X3—X4)n(1) wherein the brackets delineate a repeat or quasi-repeat of the consensus sequence; wherein n describes the number of A-repeats or quasi-A-repeats, and is from 1 to 100, or from 2 to 50, or from 5 to 50, or from 5 to 20; wherein X1is a motif that is 4 amino acids in length, wherein the first amino acid of X1is Y, and wherein the remaining amino acids of X1are GAP, GLP, GPP, GTP, or GVP; wherein X2is a motif that is from 3 to 20 amino acids in length; wherein X2comprises GGG, GGGG, N, NG, NN, NGN, NGNG, GQGG, GQGN, GQGQ, GQGQG, or 3 or more glycine residues, or wherein 50% or more of the residues of X2are either glycine or asparagine, or wherein 60% or more of the residues of X2are either glycine or asparagine, or wherein 70% or more of the residues of X2are either glycine or asparagine, or wherein 80% or more of the residues of X2are either glycine or asparagine; wherein X3is a motif that is from 2 to 6 amino acids in length, wherein X3is GG, LS, APS, GAG, GGG, KPS, RPS, or GGGG; and wherein X4is a motif that is from 1 to 2 amino acids in length, wherein X4is S, D, T, N, L, DS, DT, LS, SS, ST, TN, or TS. In some such embodiments, the recombinant resilins comprise motifs X1, X2, X3, and X4whereas in other embodiments, the recombinant resilins comprise motifs X1, X2, X3, or X4, or combinations thereof. In some embodiments, the recombinant resilins provided herein comprise one or more B-repeats. In some such embodiments, the recombinant resilins comprise between 1 and 100 B-repeats, or from 2 to 50 A-repeats, or from 5 to 50 A-repeats, or from 5 to 20 A-repeats. In some embodiments, the recombinant resilins comprise one or more consensus sequences described by the formula, (X11—X12—X13)m(2) wherein the brackets delineate a repeat or quasi-repeat of the consensus sequence; wherein m describes the number of B-repeats or quasi-B-repeats, and is from 1 to 100; wherein X11is a motif that is from 1 to 5 amino acids in length, the first amino acid is Y, and where the remaining amino acids can comprise GAP, GPP, SSG, or SGG; wherein X12is a motif that is from 2 to 5 amino acids in length and comprises GQ, GN, RPG, RPGGQ, RPGGN, SSS, SKG, or SN; and wherein X13is a motif that is from 4 to 30 amino acids in length and comprises GG, DLG, GFG, GGG, RDG, SGG, SSS, GGSF, GNGG, GGAGG, or 3 or more glycine residues, or 30% or more of the residues are glycine, or 40% or more of the residues are glycine, or 50% or more of the residues are glycine, or 60% or more of the residues are glycine. In some such embodiments, the recombinant resilins comprise motifs X11, X12, and X13whereas in other such embodiments, the recombinant resilins comprise motifs X11, X12, or X13, or combinations thereof. In some embodiments, the recombinant resilins provided herein comprise one or more A-repeats, one or more B-repeats, and/or one or more C-domain. In some embodiments, the recombinant resilins comprise one or more A-repeats or one or more B-repeats but not both. In some embodiments, the recombinant resilins comprise one or more A-repeats but not B-repeats or C-domains. In some embodiments, the recombinant resilins comprise one or more B-repeats but not A-repeats or C-domains. In embodiments in which the recombinant resilins comprise a C-domain, the C-domain can be situated either on the N-terminal or the C-terminal sides of the A-repeats or B-repeats, or between the A-repeats and the B-repeats. In some embodiments, the recombinant resilins further comprise the sequence XXEPPVSYLPPS, where X is any amino acid. In some such embodiments, the sequence is located on the N-terminal side of an A-repeat or B-repeat. In some embodiments, the recombinant resilins are full-length native resilins expressed in a non-native environment. In some embodiments, the recombinant resilins comprise a truncated version of native resilins. In some embodiments, the truncated native resilins comprise at least one A-repeat. In some embodiments, the truncated native resilins comprise at least one B-repeat. Non-limiting examples of full-length and truncated native resilins are provided as SEQ ID NOs: 1 through 44. In some embodiments, the recombinant resilins are full-lengthDrosophila sechelliaresilin (SEQ ID NO: 1). In some embodiments, the recombinant resilins are truncatedAcromyrmex echinatiorresilin (SEQ ID NO: 4). In some embodiments, the recombinant resilins are full-length or truncated native resilins that are cross-linked in a non-native manner (e.g., less or more cross-linking, cross-linking via different amino acid residues). In some embodiments, the recombinant resilins are modified full-length or truncated native resilins. In some embodiments, the recombinant resilins are at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% identical to a full-length or truncated native resilin. In some embodiments, the recombinant resilins are at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% identical to full-lengthDrosophila sechelliaresilin (SEQ ID NO: 1). In some embodiments, the recombinant resilins are at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% identical to truncatedAcromyrmex echinatiorresilin (SEQ ID NO: 4). There are a number of different algorithms known in the art which can be used to measure nucleotide sequence or protein sequence identity. For instance, polynucleotide sequences can be compared using FASTA, Gap, or Bestfit, which are programs in Wisconsin Package Version 10.0, Genetics Computer Group (GCG), Madison, Wis. FASTA provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences. See, e.g., Pearson, Methods Enzymol. 183:63-98, 1990 (hereby incorporated by reference in its entirety). For instance, percent sequence identity between nucleic acid sequences can be determined using FASTA with its default parameters (a word size of 6 and the NOPAM factor for the scoring matrix) or using Gap with its default parameters as provided in GCG Version 6.1, herein incorporated by reference. Alternatively, sequences can be compared using the computer program, BLAST (Altschul et al., J. Mol. Biol. 215:403-410, 1990; Gish and States, Nature Genet. 3:266-272, 1993; Madden et al., Meth. Enzymol. 266:131-141, 1996; Altschul et al., Nucleic Acids Res. 25:3389-3402, 1997; Zhang and Madden, Genome Res. 7:649-656, 1997, especially blastp or tblastn (Altschul et al., Nucleic Acids Res. 25:3389-3402, 1997. In some embodiments, the modified resilins differ from full-length or truncated native resilins in amino acid residues that are post-translationally modified (e.g., glycosylated, phosphorylated) such that the modified resilins have one or more different locations and/or different amounts and/or different types of post-translational modifications than the full-length or truncated native resilins. In some embodiments, the modified resilins differ from full-length or truncated native resilins in amino acid residues that are involved in cross-linking such that the modified resilins have one or more different locations and/or different amounts and/or different types of amino acids that are involved in cross-linking than full-length or truncated native resilins. In some such embodiments, the modified resilins differ from the full-length or truncated native resilin in comprising one or more additional or fewer tyrosine residues, one or more additional or fewer lysine residues, and/or one or more additional or fewer cysteine residues. In some embodiments, the recombinant resilins comprise concatenated native or truncated native resilins or concatenated modified resilins. In some embodiments, the concatenated native or truncated native resilins or concatenated modified resilins comprise at least 2 A-repeats (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more). In some embodiments, the concatenated truncated native resilins or concatenated modified resilins comprise at least 2 B-repeats (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more). The compositions provided herein comprise at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%; between 10% and 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, or 20%; between 20% and 100%, 90%, 80%, 70%, 60%, 50%, 40%, or 30%; between 30% and 100%, 90%, 80%, 70%, 60%, 50%, or 40%; between 40% and 100%, 90%, 80%, 70%, 60%, or 50%; between 50% and 100%, 90%, 80%, 70%, or 60%; between 60% and 100%, 90%, 80%, or 70%; between 70% and 100%, 90%, or 80%; between 80% and 100%, or 90%; or between 90% and 100% by weight of recombinant resilins. The recombinant resilins can be identical recombinant resilins or mixtures of recombinant resilins having at least 2 different amino acid sequences. In some embodiments, the compositions provided herein have similar properties compared to compositions comprising native resilins. In other embodiments, the compositions provided herein have different properties compared to compositions comprising native resilins. Non-limited examples of such properties include resilience, compressive elastic modulus, tensile elastic modulus, shear modulus, extension to break, maximum tensile strength, hardness, rebound, and compression set. Parameters that can be modified to obtain compositions with specific mechanical properties include, for example, the length and/or sequence of the recombinant resilins, the extent and/or type of post-translational modifications of the recombinant resilins, and/or the extent and/or type of cross-linking of the recombinant resilins. In some embodiments, mechanical properties such as maximum tensile strength, compressive elastic modulus, tensile elastic modulus, shear modulus, extension to break and resilience can be measured using many different types of tensile and compression systems that conduct stress-strain measurements on elastomeric samples. The resulting stress-strain curves, including curves with hysteresis, can be measured in tension or compression. In some embodiments, tension and compression test systems can apply a strain to a sample and measure the resulting force using a load cell. In some embodiments, the mechanical properties can be measured at the macroscopic scale (e.g., using macroscopic compression testers), microscopic, or nanoscopic scale (e.g., using atomic-force microscopy (AFM) or nanoindentation measurements). In some embodiments, the compressive mechanical properties of elastomers can be measured according to the standard ASTM D575-91(2012) Standard Test Methods for Rubber Properties in Compression. Mechanical measurements of elastomers in tension can be performed using ASTM D412-15a Standard Test Methods for Vulcanized Rubber and Thermoplastic Elastomers—Tension. In some embodiments, tear strength of elastomers can be performed using ASTM D624-00 Standard Test Method for Tear Strength of Conventional Vulcanized Rubber and Thermoplastic Elastomers. In some embodiments, mechanical properties of slab, bonded, and molded elastomers can be performed using ASTM D3574-11 Standard Test Methods for Flexible Cellular Materials—Slab, Bonded, and Molded Urethane Foams. In some embodiments, the mechanical properties of elastomers can be measured using ASTM D5992-96(2011) Standard Guide for Dynamic Testing of Vulcanized Rubber and Rubber-Like Materials Using Vibratory Methods. In some embodiments, the compositions provided herein have a resilience of greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95%; from 50% to 100%, 90%, 80%, 70%, or 60%; from 60% to 100%, 90%, 80%, or 70%; from 70% to 100%, 90%, or 80%; from 80% to 100%, or 90%; from 90% to 100%; from 95% to 100%, from 90% to 99%, or from 95% to 99%. In some embodiments, the compositions provided herein have a compressive elastic modulus of less than 10 MPa, less than 7 MPa, less than 5 MPa, less than 2 MPa, less than 1 MPa, less than 0.5 MPa, or less than 0.1 MPa; from 0.01 MPa to 10 MPa, 7 MPa, 5 MPa, 2 MPa, 1 MPa, 0.5 MPa, or 0.1 MPa; from 0.1 MPa to 10 MPa, 7 MPa, 5 MPa, 2 MPa, 1 MPa, or 0.5 MPa; from 0.5 MPa to 10 MPa, 7 MPa, 5 MPa, 2 MPa, or 1 MPa; from 1 MPa to 10 MPa, 7 MPa, 5 MPa, or 2 MPa; from 2 MPa to 10 MPa, 7 MPa, or 5 MPa; from 5 MPa to 10 MPa, or 7 MPa; or from 7 MPa to 10 MPa. In some embodiments, the compressive elastic modulus of a composition can be measured as defined by the ASTM D575-91(2012) Standard Test Methods for Rubber Properties in Compression. In some embodiments, the compositions provided herein have a tensile elastic modulus of less than 10 MPa, less than 7 MPa, less than 5 MPa, less than 2 MPa, less than 1 MPa, less than 0.5 MPa, or less than 0.1 MPa; from 0.01 MPa to 10 MPa, 7 MPa, 5 MPa, 2 MPa, 1 MPa, or 0.5 MPa; from 0.5 MPa to 10 MPa, 7 MPa, 5 MPa, 2 MPa, or 1 MPa; from 1 MPa to 10 MPa, 7 MPa, 5 MPa, or 2 MPa; from 2 MPa to 10 MPa, 7 MPa, or 5 MPa; from 5 MPa to 10 MPa, or 7 MPa; or from 7 MPa to 10 MPa. In some embodiments, the compositions provided herein have a shear modulus of less than 1 MPa, less than 100 kPa, less than 50 kPa, less than 20 kPa, less than 10 kPa, or less than 1 kPa; from 0.1 kPa to 1 MPa, 100 kPa, 50 kPa, 20 kPa, 10 kPa, or 1 kPa; from 1 kPa to 1 MPa, 100 kPa, 50 kPa, 20 kPa, or 10 kPa; from 10 kPa to 1 MPa, 100 kPa, 50 kPa, or 20 kPa; from 20 kPa to 1 MPa, 100 kPa, or 50 kPa; from 50 kPa to 1 MPa, or 100 kPa; or from 100 kPa to 1 MPa. In some embodiments, the compositions provided herein have an extension to break of greater than 1%, greater than 10%, greater than 50%, greater than 100%, greater than 300%, or greater than 500%; from 1% to 500%, 300%, 100%, 50%, or 10%; from 10% to 500%, 300%, 100%, or 50%; from 50% to 500%, 300%, or 100%; from 100% to 500%, or 300%; or from 300% to 500%. In some embodiments, the compositions provided herein have a maximum tensile strength of greater than 0.1 kPa, greater than 1 kPa, greater than 2 kPa, greater than 5 kPa, or greater than 10 kPa; from 0.1 kPa to 100 kPa, 10 kPa, 5 kPa, 2 kPa, or 1 kPa; from 1 kPa to 100 kPa, 10 kPa, 5 kPa, or 2 kPa; from 2 kPa to 100 kPa, 10 kPa, or 5 kPa; from 5 kPa to 100 kPa, or 10 kPa; or from 10 kPa to 100 kPa. In some embodiments, mechanical properties such as hardness and compressive elastic modulus can be measured using indentation and nanoindentation measurement systems. In some embodiments, indentation measurements utilizing a tip to indent the sample to a given amount of strain are used to measure the hardness and compressive elastic modulus of resilin, and the resulting force is measured using a load cell. In some embodiments, different tip shapes can be used including Vickers and Berkovich shaped tips. In some embodiments, the hardness measured by indentation techniques is characterized by the relation, Hardness=(Peak Force)/(Contact Area). In some embodiments, the hardness in polymers, elastomers, and rubbers can be measured using a durometer. In some embodiments the hardness of an elastomer can be measured using the standard ASTM D2240, which recognizes twelve different durometer scales using combinations of specific spring forces and indentor configurations. The most common scales are the Shore 00, A and D Hardness Scales. Hardness scales range from 0 to 100, where 0 is softer material and 100 is harder material. In some embodiments, the compositions provided herein have a Shore 00 Hardness of less than 90, less than 80, less than 70, less than 60, less than 50, less than 40, less than 30, or less than 20; from 10 to 90, 80, 70, 60, 50, 40, 30, or 20; from 20 to 90, 80, 70, 60, 50, 40, or 30; from 30 to 90, 80, 70, 60, 50, or 40; from 40 to 90, 80, 70, 60, or 50; from 50 to 90, 80, 70, or 60; from 60 to 90, 80, or 70; from 70 to 90, or 80; or from 80 to 90. In some embodiments, hardness measurements in resilin are performed according to ASTM D2240. As used here, the term “rebound” refers to a particular measure of resilience. In some embodiments, rebound can be measured with a number of different tools including pendulum tools and dropped balls. In the pendulum type measurements, RB, commonly called percentage rebound, is determined from the equation: R⁢B=[1-cos⁡(angle⁢⁢of⁢⁢rebound)][1-cos⁡(originial⁢⁢angle)]×1⁢0⁢0 The rebound resilience can be calculated as: R=hH where h=apex height of the rebound, and H=initial height. The rebound resilience can also be determined by the measurement of the angle of rebound. Some examples of test methods for determining rebound in elastomers are ASTM D2632-15 and ASTM D7121-05(2012). In some embodiments, the compositions provided herein have a rebound greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95%; from 50% to 100%, 90%, 80%, 70%, or 60%; from 60% to 100%, 90%, 80%, or 70%; from 70% to 100%, 90%, or 80%; from 80% to 100%, or 90%; from 90% to 100%; from 95% to 100%, from 90% to 99%, or from 95% to 99%. In some embodiments, rebound measurements in resilin are performed according to ASTM D2632-15, or ASTM D7121-05(2012). As used herein, the term “compression set” refers to a measure of the permanent deformation remaining after an applied force is removed. In some embodiments, compression set can be measured in different ways, including compression set under constant force in air (referred to as Compression Set A), compression set under constant deflection in air (referred to as Compression Set B), and compression set under constant deflection in air considering material hardness (referred to as Compression Set C). Compression Set A (CA) is calculated by the following expression: CA=[(to−ti)/to]×100, where tois the original specimen thickness, and tiis the specimen thickness after testing). Compression set B (CB) is given by CB=[(to−ti)/(to−tn)]100, where tois the original specimen thickness, tiis the specimen thickness after testing, and tois the spacer thickness or the specimen thickness during the test. Some examples of test methods for determining compression set in elastomers are ASTM D3574-11 and ASTM D395-16. In some embodiments, the compositions provided herein have a Compression Set A or a Compression Set B of greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95%; from 50% to 100%, 90%, 80%, 70%, or 60%; from 60% to 100%, 90%, 80%, or 70%; from 70% to 100%, 90%, or 80%; from 80% to 100%, or 90%; from 90% to 100%; from 95% to 100%, from 90% to 99%, or from 95% to 99%. In some embodiments, compression set measurements in resilin are performed according to ASTM D3574-11 and ASTM D395-16. The processing and forming of resilin into products can take many forms for different applications. Accordingly, the compositions provided herein can have any shape and form, including but not limited to gels, porous sponges, films, machinable solids, cast forms, molded forms, and composites. The compositions provided herein have a number of uses, including but not limited to applications in aerospace, automotive, sporting equipment, vibration isolation, footwear, and clothing among others. Some applications from these categories are listed as non-limiting examples. Due to the desirable elastic efficiency, resilin can be used as an energy storage device (e.g., a rubber band) for storing and recovering mechanical energy. Automobile suspension systems can be improved by application of resilin bushings to keep more tire contact on the road when going over bumps and through potholes at speed. Additionally, there are a number of sporting equipment applications for resilin with differently tuned mechanical properties including cores of golf balls, tennis racket grips, golf club grips, and table tennis paddles. An application of particular interest is footwear due to the unique properties of resilin compositions provided herein. As an insole or midsole, resilin can improve the comfort and bioefficiency of shoes by cushioning the foot strike and restoring more of the energy from that footstrike as forward momentum. As a midsole, resilin can make up the entire midsole or be encapsulated within another material to complement its properties (e.g., an abrasion or wear resistant material, or a material tuned for traction). The resilin midsole can also contain a plurality of resilin materials with differently tuned mechanical properties that work in concert to provide enhanced performance (e.g., softer heel strike area and firmer arch support). As used herein, the term “density” refers to the mass of the sample divided by the volume. In some embodiments, the density of an elastomer can be determined using a pycnometer with alcohol in place of water to eliminate air bubbles. In some embodiments, the density of an elastomer can be determined using a hydrostatic method. As used herein, the term “compressed volume density” refers to the ratio of the sample mass to the compressed volume of the sample, where the “compressed volume” is defined as the final equilibrium volume attained by an elastomeric sample when it is subjected to a compressive force sufficient to cause it to flow until it fully conforms to the surrounding shape of the piston-cylinder test chamber enclosure. In some embodiments, the compressed volume density of an elastomer can be determined using a compressed volume densimeter. In some embodiments, the compositions provided herein have a density or a compressed volume density are from 0.5 mg/cm3to 2.0 mg/cm3, or from 1.0 mg/cm3to 1.5 mg/cm3, or from 1.1 mg/cm3to 1.4 mg/cm3, or from 1.2 mg/cm3to 1.35 mg/cm3. In some embodiments, the determination of the density or the compressed volume density of elastomers can be performed using ASTM D297-15 Standard Test Methods for Rubber Products—Chemical Analysis. Recombinant Resilin Vectors, Recombinant Host Cells, and Fermentations Further provided herein are vectors encoding recombinant resilins, recombinant host cells comprising such vectors, and fermentations comprising such recombinant host cells and recombinant resilins. In some embodiments, the vectors provided herein comprise secreted resilin coding sequences, which encode a resilin polypeptide fused at its N-terminus to a secretion signal and optionally at its C-terminus to a tag peptide or polypeptide. In some embodiments, the vectors comprise secreted resilin coding sequences that are codon-optimized for expression in a particular host cell. Suitable secretion signals are secretion signals that mediate secretion of polypeptides in the recombinant host cells provided herein. Non-limiting examples of suitable secretion signals are the secretion signals of the alpha mating factor (α-MF) ofSaccharomyces cerevisiae, acid phosphatase (PHO1) ofPichia pastoris, and phytohemagglutinin (PHA-E) from the common beanPhaseolus vulgaris. Additional secretion signals are known in the art, or can be identified by identification of proteins secreted by a host cell followed by genomic analysis of the secreted proteins and identification of the non-translated N-terminal sequences (see, for example, Huang et al. A proteomic analysis of thePichia pastorissecretome in methanol-induced cultures. Appl Microbiol Biotechnol. 2011 April; 90(1):235-47). The resilins encoded by the secreted resilin coding sequences can be further fused to tag peptides or polypeptides. Non-limiting examples of tag peptides or polypeptides include affinity tags (i.e., peptides or polypeptides that bind to certain agents or matrices), solubilization tags (i.e., peptides or polypeptides that assist in proper folding of proteins and prevent precipitation), chromatography tags (i.e., peptides or polypeptides that alter the chromatographic properties of a protein to afford different resolution across a particular separation techniques), epitope tags (i.e., peptides or polypeptides that are bound by antibodies), fluorescence tags (i.e., peptides or polypeptides that upon excitation with short-wavelength light emit high-wavelength light), chromogenic tags (i.e., peptides or polypeptides that absorb specific segments of the visible light spectrum), enzyme substrate tags (i.e., peptides or polypeptides that are the substrates for specific enzymatic reactions), chemical substrate tags (i.e., peptides or polypeptides that are the substrates for specific chemical modifications), or combinations thereof. Non-limiting examples of suitable affinity tags include maltose binding protein (MBP), glutathione-S-transferase (GST), poly(His) tag, SBP-tag, Strep-tag, and calmodulin-tag. Non-limiting examples of suitable solubility tags include thioredoxin (TRX), poly(NANP), MBP, and GST. Non-limiting examples of chromatography tags include polyanionic amino acids (e.g., FLAG-tag [GDYKDDDDKDYKDDDDKDYKDDDDK (SEQ ID NO: 45)]) and polyglutamate tag. Non-limiting examples of epitope tags include V5-tag, VSV-tag, Myc-tag, HA-tag, E-tag, NE-tag, and FLAG-tag. Non-limiting examples of fluorescence tags include green fluorescent protein (GFP), blue fluorescent protein (BFP), cyan fluorescent protein (CFP), yellow fluorescent protein (YFP), orange fluorescent protein (OFP), red fluorescent protein (RFP), and derivatives thereof. Non-limiting examples of chromogenic tags include non-fluorescent members of the GFP-like family of proteins (e.g., BlitzenBlue, DonnerMagenta; DNA2.0, Neward, CA). Non-limiting examples of enzyme substrate tags include peptides or polypeptides comprising a lysine within a sequence suitable for biotinilation (e.g., AviTag, Biotin Carboxyl Carrier Protein [BCCP]). Non-limiting examples of chemical substrate tags include substrates suitable for reaction with FIAsH-EDT2. The fusion of the C-terminal peptide or polypeptide to the resilin can be cleavable (e.g., by TEV protease, thrombin, factor Xa, or enteropeptidase) on non-cleavable. In some embodiments, the vectors comprise single secreted resilin coding sequences. In other embodiments, the vectors comprise 2 or more (e.g., 3, 4, or 5) secreted resilin coding sequences. In some such embodiments, the secreted resilin coding sequences are identical. In other such embodiments, at least 2 of the secreted resilin coding sequences are not identical. In embodiments in which at least 2 of the secreted resilin coding sequences are not identical, the at least 2 secreted resilin coding sequences can differ from each other in the resilins and/or in the secretion signals and/or the optional tag peptides or polypeptides they encode. In some embodiments, the vectors comprise promoters that are operably linked to the secreted resilin coding sequences such that they drive the expression of the secreted resilin coding sequences. The promotors can be constitutive promoters or inducible promoters. In some embodiments, induction of the inducible promoter occurs via glucose repression, galactose induction, sucrose induction, phosphate repression, thiamine repression, or methanol induction. Suitable promoters include promoters that mediate expression of proteins in the recombinant host cells provided herein. Non-limiting examples of suitable promoters include the AOX1 promoter, GAP promoter, LAC4-PBI promoter, T7 promoter, TAC promoter, GCW14 promoter, GAL1 promoter, λPL promoter, λPR promoter, beta-lactamase promoter, spa promoter, CYC1 promoter, TDH3 promoter, GPD promoter, TEF1 promoter, ENO2 promoter, PGL1 promoter, SUC2 promoter, ADH1 promoter, ADH2 promoter, HXT7 promoter, PHO5 promoter, and CLB1 promoter. Additional promoters that can be used to facilitate expression of the secreted resilin coding sequences are known in the art. In some embodiments, the vectors comprise terminators that are operably linked to the secreted resilin coding sequences such that they effect termination of transcription of the secreted resilin coding sequences. Suitable terminators include terminators that terminate transcription in the recombinant host cells provided herein. Non-limiting examples of suitable terminators include the AOX1 terminator, PGK1 terminator, and TPS1 terminator. Additional terminators that effect termination of transcription of the secreted resilin coding sequences are known in the art. In embodiments in which the vectors comprise 2 or more resilin coding sequences, the 2 or more resilin coding sequences can be operably linked to the same promoters and/or terminators or to 2 or more different promoters and/or terminators. The vectors provided herein can further comprise elements suitable for propagation of the vectors in recombinant host cells. Non-limiting examples of such elements include bacterial origins of replication and selection markers (e.g., antibiotic resistance genes, auxotrophic markers). Bacterial origins of replication and selection markers are known in the art. In some embodiments, the selection marker is a drug resistant marker. A drug resistant maker enables cells to detoxify an exogenously added drug that would otherwise kill the cell. Illustrative examples of drug resistant markers include but are not limited to those for resistance to antibiotics such as ampicillin, tetracycline, kanamycin, bleomycin, streptomycin, hygromycin, neomycin, Zeocin™, and the like. In some embodiments, the selection marker is an auxotrophic marker. An auxotrophic marker allows cells to synthesize an essential component (usually an amino acid) while grown in media that lacks that essential component. Selectable auxotrophic gene sequences include, for example, hisD, which allows growth in histidine-free media in the presence of histidinol. Other selection markers suitable for the vectors of the present invention include a bleomycin-resistance gene, a metallothionein gene, a hygromycin B-phosphotransferase gene, the AURI gene, an adenosine deaminase gene, an aminoglycoside phosphotransferase gene, a dihydrofolate reductase gene, a thymidine kinase gene, and a xanthine-guanine phosphoribosyltransferase gene. The vectors of the present invention can further comprise targeting sequences that direct integration of the secreted resilin coding sequences to specific locations in the genome of host cells. Non-limiting examples of such targeting sequences include nucleotide sequences that are identical to nucleotide sequences present in the genome of a host cell. In some embodiments, the targeting sequences are identical to repetitive elements in the genome of host cells. In some embodiments, the targeting sequences are identical to transposable elements in the genome of host cells. In some embodiments, recombinant host cells are provided herein that comprise the vectors described herein. In some embodiments, the vectors are stably integrated within the genome (e.g., a chromosome) of the recombinant host cells, e.g., via homologous recombination or targeted integration. Non-limiting examples of suitable sites for genomic integration include the Ty1 loci in theSaccharomyces cerevisiaegenome, the rDNA and HSP82 loci in thePichia pastorisgenome, and transposable elements that have copies scattered throughout the genome of the recombinant host cells. In other embodiments, the vectors are not stably integrated within the genome of the recombinant host cells but rather are extrachromosomal. Recombinant host cells can be of mammalian, plant, algae, fungi, or microbe origin. Non-limiting examples of suitable fungi include methylotrophic yeast, filamentous yeast,Arxula adeninivorans, Aspergillus niger, Aspergillus nigervar.awamori, Aspergillus oryzae, Candida etchellsii, Candida guilliermondii, Candida humilis, Candida lipolytica, Candida pseudotropicalis, Candida utilis, Candida versatilis, Debaryomyces hansenii, Endothia parasitica, Eremothecium ashbyii, Fusarium moniliforme, Hansenula polymorpha, Kluyveromyces lactis, Kluyveromyces marxianus, Kluyveromyces thermotolerans, Morteirella vinaceaevar.raffinoseutilizer, Mucor miehei, Mucor mieheivar. Cooney et Emerson,Mucor pusillus Lindt, Penicillium roquefortii, Pichia methanolica, Pichia pastoris(Komagataella phaffii),Pichia(Scheffersomyces)stipitis, Rhizopus niveus, Rhodotorulasp.,Saccharomyces bayanus, Saccharomyces beticus, Saccharomyces cerevisiae, Saccharomyces chevalieri, Saccharomyces diastaticus, Saccharomyces ellipsoideus, Saccharomyces exiguus, Saccharomyces florentinus, Saccharomyces fragilis, Saccharomyces pastorianus, Saccharomyces pombe, Saccharomyces sake, Saccharomyces uvarum, Sporidiobolus johnsonii, Sporidiobolus salmonicolor, Sporobolomyces roseus, Trichoderma reesi, Xanthophyllomyces dendrorhous, Yarrowia lipolytica, Zygosaccharomyces rouxii, and derivatives and crosses thereof. Non-limiting examples of suitable microbes includeAcetobactersuboxydans,Acetobacter xylinum, Actinoplane missouriensis, Arthrospira platensis, Arthrospira maxima, Bacillus cereus, Bacillus coagulans, Bacillus licheniformis, Bacillus stearothermophilus, Bacillus subtilis, Escherichia coli, Lactobacillus acidophilus, Lactobacillus bulgaricus, Lactobacillus reuteri, Lactococcus lactis, Lactococcus lactisLancefield Group N,Leuconostoc citrovorum, Leuconostoc dextranicum, Leuconostoc mesenteroidesstrain NRRL B-512(F),Micrococcus lysodeikticus, Spirulina, Streptococcus cremoris, Streptococcus lactis, Streptococcus lactissubspeciesdiacetylactis, Streptococcus thermophilus, Streptomyces chattanoogensis, Streptomyces griseus, Streptomyces natalensis, Streptomyces olivaceus, Streptomyces olivochromogenes, Streptomyces rubiginosus, Xanthomonas campestris, and derivatives and crosses thereof. Additional strains that can be used as recombinant host cells are known in the art. It should be understood that the term “recombinant host cell” is intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but is still included within the scope of the term “recombinant host cell” as used herein. In some embodiments, the recombinant host cells comprise genetic modifications that improve production of the recombinant resilins provided herein. Non-limiting examples of such genetic modifications include altered promoters, altered kinase activities, altered protein folding activities, altered protein secretion activities, altered gene expression induction pathways, and altered protease activities. The recombinant host cells provided herein are generated by transforming cells of suitable origin with vectors provided herein. For such transformation, the vectors can be circularized or be linear. Recombinant host cell transformants comprising the vectors can be readily identified, e.g., by virtue of expressing drug resistance or auxotrophic markers encoded by the vectors that permit selection for or against growth of cells, or by other means (e.g., detection of light emitting peptide comprised in vectors, molecular analysis of individual recombinant host cell colonies, e.g., by restriction enzyme mapping, PCR amplification, or sequence analysis of isolated extrachromosomal vectors or chromosomal integration sites). In some embodiments, the recombinant host cells provided herein can produce high titers of the recombinant resilins provided herein. In some such embodiments, the recombinant host cells produce the recombinant resilins at a rate of greater than 2 mg resilin/g dry cell weight/hour, 4 mg resilin/g dry cell weight/hour, 6 mg resilin/g dry cell weight/hour, 8 mg resilin/g dry cell weight/hour, 10 mg resilin/g dry cell weight/hour, 12 mg resilin/g dry cell weight/hour, 14 mg resilin/g dry cell weight/hour, 16 mg resilin/g dry cell weight/hour, 18 mg resilin/g dry cell weight/hour, 20 mg resilin/g dry cell weight/hour, 25 mg resilin/g dry cell weight/hour, or 30 mg resilin/g dry cell weight/hour; from 2 to 40, 30, 20, 10, or 5 mg resilin/g dry cell weight/hour; from 5 to 40, 30, 20, or 10 mg resilin/g dry cell weight/hour; from 10 to 40, 30, or 20 mg resilin/g dry cell weight/hour; from 20 to 40, or 30 mg resilin/g dry cell weight/hour; or from 30 to 40 mg resilin/g dry cell weight/hour. In other such embodiments, the recombinant host cells secrete the recombinant resilins at a rate of greater than 2 mg resilin/g dry cell weight/hour, 4 mg resilin/g dry cell weight/hour, 6 mg resilin/g dry cell weight/hour, 8 mg resilin/g dry cell weight/hour, 10 mg resilin/g dry cell weight/hour, 12 mg resilin/g dry cell weight/hour, 14 mg resilin/g dry cell weight/hour, 16 mg resilin/g dry cell weight/hour, 18 mg resilin/g dry cell weight/hour, 20 mg resilin/g dry cell weight/hour, 25 mg resilin/g dry cell weight/hour, or 30 mg resilin/g dry cell weight/hour; from 2 to 40, 30, 20, 10, or 5 mg resilin/g dry cell weight/hour; from 5 to 40, 30, 20, or 10 mg resilin/g dry cell weight/hour; from 10 to 40, 30, or 20 mg resilin/g dry cell weight/hour; from 20 to 40, or 30 mg resilin/g dry cell weight/hour; or from 30 to 40 mg resilin/g dry cell weight/hour. The identities of the recombinant resilins produced can be confirmed by HPLC quantification, Western blot analysis, polyacrylamide gel electrophoresis, and 2-dimensional mass spectroscopy (2D-MS/MS) sequence identification. In some embodiments, the recombinant host cells provided herein have high secreted fractions of the recombinant resilins provided herein. In some such embodiments, the recombinant host cells have secreted fractions of recombinant resilient that is greater than 50%, 60%, 70%, 80%, or 90%; from 50% to 100%, 90%, 80%, 70%, or 60%; from 60% to 100%, 90%, 80%, or 70%; from 70% to 100%, 90%, or 80%; from 90% to 100%, or 90%; or from 90% to 100%. Production and secretion of recombinant resilins can be influenced by the number of copies of the secreted resilin coding sequences comprised in the recombinant host cells and/or the rate of transcription of the secreted resilin coding sequences comprised in the recombinant host cells. In some embodiments, the recombinant host cells comprise a single secreted resilin coding sequence. In other embodiments, the recombinant host cells comprise 2 or more (e.g., 3, 4, 5, or more) secreted resilin coding sequences. In some embodiments, the recombinant host cells comprise secreted resilin coding sequences that are operably linked to strong promoters. Non-limiting examples of strong promoters include the pGCW14 promoter ofPichia pastoris. In some embodiments, the recombinant host cells comprise secreted resilin coding sequences that are operably linked to medium promoters. Non-limiting examples of such medium promoters include the pGAP promoter ofPichia pastoris. In some embodiments, the recombinant host cells comprise coding sequences encoding resilins under the control of weak promoters. The fermentations provided herein comprise recombinant host cells described herein and a culture medium suitable for growing the recombinant host cells. The fermentations are obtained by culturing the recombinant host cells in culture media that provide nutrients needed by the recombinant host cells for cell survival and/or growth, and for secretion of the recombinant resilins. Such culture media typically contain an excess carbon source. Non-limiting examples of suitable carbon sources include monosaccharides, disaccharides, polysaccharides, and combinations thereof. Non-limiting examples of suitable monosaccharides include glucose, galactose, mannose, fructose, ribose, xylose, arabinose, ribose, and combinations thereof. Non-limiting examples of suitable disaccharides include sucrose, lactose, maltose, trehalose, cellobiose, and combinations thereof. Non-limiting examples of suitable polysaccharides include raffinose, starch, glycogen, glycan, cellulose, chitin, and combinations thereof. In some embodiments, the fermentations comprise recombinant resilins in amounts of at least 1%, 5%, 10%, 20%, or 30%; from 1% to 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, or 10%; from 10% to 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, or 20%; from 20% to 100%, 90%, 80%, 70%, 60%, 50%, 40%, or 30%; from 30% to 100%, 90%, 80%, 70%, 60%, 50%, or 40%; from 40% to 100%, 90%, 80%, 70%, 60%, or 50%; from 50% to 100%, 90%, 80%, 70%, or 60%; from 60% to 100%, 90%, 80%, or 70%; from 70% to 100%, 90%, or 80%; from 80% to 100%, or 90%; or from 90% to 100% by weight of the total fermentation. In some embodiments, the fermentations comprise recombinant resilin in an amount of at least 2 g/L, 5 g/L, 10 g/L, 15 g/L, 20 g/L, 25 g/L, or 30 g/L; from 2 g/L to 300 g/L, 200 g/L, 100 g/L, 90 g/L, 80 g/L, 70 g/L, 60 g/L, 50 g/L, 40 g/L, 30 g/L, 20 g/L, or 10 g/L; from 10 g/L to 300 g/L, 200 g/L, 100 g/L, 90 g/L, 80 g/L, 70 g/L, 60 g/L, 50 g/L, 40 g/L, 30 g/L, or 20 g/L; from 20 g/L to 300 g/L, 200 g/L, 100 g/L, 90 g/L, 80 g/L, 70 g/L, 60 g/L, 50 g/L, 40 g/L, or 30 g/L; from 30 g/L to 300 g/L, 200 g/L, 100 g/L, 90 g/L, 80 g/L, 70 g/L, 60 g/L, 50 g/L, or 40 g/L; from 40 g/L to 300 g/L, 200 g/L, 100 g/L, 90 g/L, 80 g/L, 70 g/L, 60 g/L, or 50 g/L; from 50 g/L to 300 g/L, 200 g/L, 100 g/L, 90 g/L, 80 g/L, 70 g/L, or 60 g/L; from 60 g/L to 300 g/L, 200 g/L, 100 g/L, 90 g/L, 80 g/L, or 70 g/L; from 70 g/L to 300 g/L, 200 g/L, 100 g/L, 90 g/L, or 80 g/L; from 80 g/L to 300 g/L, 200 g/L, 100 g/L, or 90 g/L; from 90 g/L to 300 g/L, 200 g/L, or 100 g/L; from 100 g/L to 300 g/L, or 200 g/L; or from 200 g/L to 300 g/L. Methods Further provided herein are methods for the production of the recombinant resilins described herein. The methods are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification unless otherwise indicated. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989; Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates, 1992, and Supplements to 2002); Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1990; Taylor and Drickamer, Introduction to Glycobiology, Oxford Univ. Press, 2003; Worthington Enzyme Manual, Worthington Biochemical Corp., Freehold, N.J.; Handbook of Biochemistry: Section A Proteins, Vol I, CRC Press, 1976; Handbook of Biochemistry: Section A Proteins, Vol II, CRC Press, 1976; Essentials of Glycobiology, Cold Spring Harbor Laboratory Press, 1999. In some embodiments, a novel method is utilized to secrete resilin extracellularly from a host cell. In some embodiments, the method comprises constructing a vector comprising a secreted resilin coding sequence (step1001inFIG.2), transforming the vector into a host cell (step1002inFIG.2), and then culturing the recombinant host cells to secrete resilin extracellularly (step1003inFIG.2). In some embodiments, the method includes secreting the resilin extracellularly at a rate greater than 2 mg resilin/g dry cell weight/hour, 4 mg resilin/g dry cell weight/hour, 6 mg resilin/g dry cell weight/hour, 8 mg resilin/g dry cell weight/hour, 10 mg resilin/g dry cell weight/hour, 12 mg resilin/g dry cell weight/hour, 14 mg resilin/g dry cell weight/hour, 16 mg resilin/g dry cell weight/hour, 18 mg resilin/g dry cell weight/hour, 20 mg resilin/g dry cell weight/hour, 25 mg resilin/g dry cell weight/hour, or 30 mg resilin/g dry cell weight/hour; from 2 to 40, 30, 20, 10, or 5 mg resilin/g dry cell weight/hour; from 5 to 40, 30, 20, or 10 mg resilin/g dry cell weight/hour; from 10 to 40, 30, or 20 mg resilin/g dry cell weight/hour; from 20 to 40, or 30 mg resilin/g dry cell weight/hour; or from 30 to 40 mg resilin/g dry cell weight/hour. In some embodiments, the secreted resilin is then purified (step1004inFIG.2), and the purified resilin is cross-linked to form an elastomer (step1005inFIG.2). In some embodiments, the methods provided herein comprise the step of transforming cells with vectors provided herein to obtain recombinant host cells provided herein (step1002inFIG.2). Methods for transforming cells with vectors are well-known in the art. Non-limiting examples of such methods include calcium phosphate transfection, dendrimer transfection, liposome transfection (e.g., cationic liposome transfection), cationic polymer transfection, electroporation, cell squeezing, sonoporation, optical transfection, protoplast fusion, impalefection, hydrodynamic delivery, gene gun, magnetofection, and viral transduction. One skilled in the art is able to select one or more suitable methods for transforming cells with vectors provided herein based on the knowledge in the art that certain techniques for introducing vectors work better for certain types of cells. In some embodiments, the methods further comprise the step of culturing the recombinant host cells provided herein in culture media under conditions suitable for obtaining the fermentations provided herein (step1003inFIG.2). In some embodiments, the conditions and culture media are suitable to facilitate secretion of the recombinant proteins from the recombinant host cells into the culture media. Suitable culture media for use in these methods are known in the art, as are suitable culture conditions. Exemplary details of culturing yeast host cells are described in Idiris et al., Appl. Microbiol. Biotechnol. 86:403-417, 2010; Zhang et al., Biotechnol. Bioprocess. Eng. 5:275-287, 2000; Zhu, Biotechnol. Adv. 30:1158-1170, 2012; Li et al., MAbs 2:466-477, 2010. In some embodiments, the methods further comprise the step of purifying secreted recombinant resilins from the fermentations provided herein to obtain the recombinant resilins provided herein (step1004inFIG.2). Purification can occur by a variety of methods known in the art for purifying secreted proteins from fermentations. Common steps in such methods include centrifugation (to remove cells) followed by precipitation of the proteins using precipitants or other suitable cosmotropes (e.g., ammonium sulfate). The precipitated protein can then be separated from the supernatant by centrifugation, and resuspended in a solvent (e.g., phosphate buffered saline [PBS]). The suspended protein can be dialyzed to remove the dissolved salts. Additionally, the dialyzed protein can be heated to denature other proteins, and the denatured proteins can be removed by centrifugation. Optionally, the purified recombinant resilins can be coacervated. In various embodiments, methods of purifying the secreted recombinant proteins from the fermentation can include various centrifugation steps in conjunction with solubilizing protein in a whole cell broth or cell pellet with known chaotropes such as urea or guanidine thiocyanate. In some embodiments, the methods provided herein further comprise the step of cross-linking the recombinant resilins to obtain the recombinant resilin compositions provided herein (step1005inFIG.2). Methods for cross-linking proteins are known in the art. In some embodiments, cross-linking is achieved via enzymatic cross-linking (e.g., using horseradish peroxidase). In other embodiments, cross-linking is achieved via photochemical cross-linking (see, for example, Elvin C M, Carr A G, Huson M G, Maxwell J M, Pearson R D, Vuocolo T, Liyou N E, Wong D C C, Merritt D J, Dixon N E. Nature 2005, 437, 999-1002; Whittaker J L, Dutta N K, Elvin C M, Choudhury N R. Journal of Materials Chemistry B 2015, 3, 6576-79; Degtyar E, Mlynarczyk B, Fratzl P, Harrington M J. Polymer 2015, 69, 255-63). In some embodiments, cross-linking is achieved via chemical cross-linking (see, for example, Renner J N, Cherry K M, Su R S C, Liu J C. Biomacromolecules 2012, 13, 3678-85; Charanti, M B, Ifkovits, J L, Burdick, J A, Linhardt J G, Kiick, K L. Soft Matter 2009, 5, 3412-16; Li L Q, Tong Z X, Jia X Q, Kiick K L. Soft Matter 2013, 9, 665-73; Li L, Mahara A, Tong Z, Levenson E A, McGann C L, Jia X, Yamaoka T, Kiick K L. Advanced Healthcare Materials 2016, 5, 266-75). In some embodiments, cross-linking is achieved via tyrosine residues. In other embodiments, cross linking is achieved via lysine residues. In some embodiments, cross linking is achieved via cysteine residues. In some embodiments, cross-linking employs transglutaminase (see, for example, Kim Y, Gill E E, Liu J C. Enzymatic Cross-Linking of Resilin-Based Proteins for Vascular Tissue Engineering Applications. Biomacromolecules. 17(8):2530-9). In some embodiments, cross-linking employs poly(ethylene glycol) (PEG) (McGann C L, Levenson E A, Kiick K L. Macromol. Chem. Phys. 2013, 214, 203-13; McGann C L, Akins R E, Kiick K L. Resilin-PEG Hybrid Hydrogels Yield Degradable Elastomeric Scaffolds with Heterogeneous Microstructure. Biomacromolecules. 2016; 17(1):128-40). In some embodiments, cross-linking occurs in vessels or molds such that the recombinant resilin compositions obtained have specific shapes or forms. EXAMPLES Example 1: Generation ofPichia pastorisRecombinant Host Cells that Secrete Recombinant Resilin Pichia pastorisrecombinant host cells that secrete recombinant resilin were generated by transforming a HIS+ derivative of GS115 (NRRL Y15851)Pichia pastoris(Komagataella phaffii) with vectors comprising secreted resilin coding sequences. The vectors each comprised 3 resilin coding sequences fused in frame to an N-terminal secretion signal (alpha mating factor leader and pro sequence), and in some instances a C-terminal 3×FLAG tag (SEQ ID NO: 45) (seeFIG.3). Each of the secreted resilin coding sequences was flanked by a promoter (pGCW14) and a terminator (tAOX1 pA signal). The vectors further comprised a targeting region that can direct integration of the 3 secreted resilin coding sequences to the HSP82 locus of thePichia pastorisgenome, dominant resistance markers for selection of bacterial and yeast transformants, as well as a bacterial origin of replication. The resilin coding sequences were obtained from scientific literature and from searching public sequence databases. The nucleotide sequences were translated into amino acid sequences and then codon-optimized. Both full length and truncated resilin sequences were chosen. Selected secreted resilin coding sequences are listed in Table 1. TABLE 1Exemplary full-length and truncated resilin amino acid sequences and recombinant host strainsAmino AcidShortSEQ IDWith FLAG tagWithout FLAG tagSpeciesTypeNameNO:PlasmidStrainPlasmidStrainDrosophilaFull lengthDs_ACB1RMp4830RMs1209RMp4842RMs1221sechelliaDrosophilaA repeats +Ds_AC2RMp4831RMs1210RMp4843RMs1222sechelliaChitinbindingdomainDrosophilaA repeats onlyDs_A3RMp4832RMs1211RMp4844RMs1223sechelliaAcromyrmexA repeats onlyAe_A4RMp4833RMs1212RMp4845RMs1224echinatiorAeshnasp.B repeats onlyAs_B5RMp4834RMs1213RMp4846RMs1225Aeshnasp.Full lengthAs_ACB6RMp4835RMs1214RMp4847RMs1226HaematobiaA repeats onlyHi_A7RMp4836RMs1215RMp4848RMs1227irritansHaematobiaFull lengthHi_ACB8RMp4837RMs1216RMp4849RMs1228irritansCtenocephalidesA repeats onlyCf_A9RMp4838RMs1217RMp4850RMs1229felisCtenocephalidesB repeats onlyCf_B10RMp4839RMs1218RMp4851RMs1230felisBombusA repeats onlyBt_A11RMp4840RMs1219RMp4852RMs1231terrestrisTriboliumA repeats onlyTc_A12RMp4841RMs1220RMp4853RMs1232castaneum The vectors were transformed intoPichia pastorisusing electroporation to generate host strains comprising 3 integrated copies of each secreted resilin coding sequence. Transformants were plated on YPD agar plates supplemented with an antibiotic, and incubated for 48 hours at 30° C. Clones from each final transformation were inoculated into 400 μL of Buffered Glycerol-complex Medium (BMGY) in 96-well blocks, and incubated for 24 hours at 30° C. with agitation at 1,000 rpm. A sample was removed, the recombinant host cells were pelleted via centrifugation, and the supernatant was recovered and run on a SDS-PAGE gel for analysis of resilin content via Coomassie gel and Western blot analysis (for polypeptides comprising the 3×FLAG tag). For FLAG-tagged proteins, the remaining cultures were used to inoculate minimal media cultures in duplicate for ELISA measurements. One duplicate was pelleted and the supernatant was measured directly. The second duplicate was extracted with guanidine thiocyanate and both the intra- and extra-cellular fractions were measured. As shown inFIG.4BandFIG.4C, recombinant resilin from numerous species expressed successfully in thePichia pastorisrecombinant host cells. (Note: Some proteins have very few basic residues, and are therefore difficult to detect by Coomassie, though they have a signal on Western.) As shown inFIG.4A, recombinant host cells secreted up to 90% of the recombinant resilin produced. Example 2: Measuring Productivity ofPichia pastorisRecombinant Host Cells Expressing and Secreting Recombinant Resilin To measure productivity, 3 clones of each recombinant host cell were inoculated into 400 μL of BMGY in a 96-well square-well block, and incubated for 48 hours at 30° C. with agitation at 1,000 rpm. Following the 48-hour incubation, 4 μL of each culture was used to inoculate 400 μL of minimal media in a 96-well square-well block, which was then incubated for 48 hours 30° C. with agitation at 1,000 rpm. 400 uL of 5M guanidine thiocyanate was added to the cultures, and the mixtures were pelleted by centrifugation. The supernatants were saved whereas the pellets were resuspended in 800 μL of 2.5M guanidine thiocyanate. The resuspended cells were physically lysed using beads, the lysed cell mixture was pelleted by centrifugation, and the supernatant was saved. The concentration of resilin in each fraction was determined by direct enzyme-linked immunosorbent assay (ELISA) analysis quantifying the 3×FLAG epitope (FIG.5AandFIG.5B). Example 3: Purification of Recombinant Resilin The non-FLAG-tagged Ds_ACB and Ae_A polypeptides were chosen for purification and cross-linking. Strains RMs1221 (expressing Ds_ACB) and RMs1224 (expressing Ae_A) were grown in 500 mL of BMGY in flasks for 48 hours at 30° C. with agitation at 300 rpm. The protocol for purification was adapted from Lyons et al. (2007). Cells were pelleted by centrifugation, and supernatants were collected. Proteins were precipitated by addition of ammonium sulfate. The precipitated proteins were resuspended in a small volume of phosphate buffered saline (PBS), and the resuspended samples were dialyzed against PBS to remove salts. The dialyzed samples were then heated to denature native proteins, and denatured proteins were removed by centrifugation. The retained supernatants contained the purified resilin polypeptides. Optionally, the retained supernatants were chilled, which caused coacervation, resulting in a concentrated lower phase and dilute upper phase. As shown inFIG.6, Ae_A was obtained in relatively pure form whereas Ds_ACB produced 3 bands at 70 kDa, 50 kDa, and 25 kDa. Example 4: Cross-Linking of Purified, Secreted, Recombinant Resilin Concentrated Ds_ACB resilin was cross-linked via one of two methods: photo cross-linking (adapted from Elvin et al. 2005) and enzymatic cross-linking (adapted from Qin et al. 2009). For photo cross-linking, resilin protein was mixed with ammonium persulfate and tris (bipyridine) ruthenium (II) ([Ru(bpy)3]2+). The mixture was exposed to bright white light, after which the mixture formed a rubbery solid. For enzymatic cross-linking, resilin protein was mixed with horseradish peroxidase (HRP) and hydrogen peroxide. The mixture was incubated at 37° C., after which the mixture formed a rubbery solid. Example 5: Production of a Block of Recombinant Resilin Strain RMs1221 (expressing the Ds_ACB resilin) was run in two 2 L fermentation tanks to produce a larger quantity of protein. The strain was grown in a minimal basal salt media with 15 g/L of glucose as a starting feedstock and 1 g/L L81 antifoam, in a stirred fermentation vessel controlled at 30° C., with 1 VVM of air flow and minimum agitation of 700 rpm. The pH of the fermentation was controlled at 5 with on-demand addition of ammonium hydroxide. Once batch glucose was depleted, glucose was added via a programmed feed recipe that was designed to maintain the oxygen uptake rate 120 mmole/L/h, the temperature was decreased to 25° C., and dissolved oxygen was maintained at 20%. The fermentation was harvested after 70 hours, at about 700-800 OD of cell density. The protein was purified as described in Example 3, and combined with reagents for enzymatic cross-linking as described in Example 4. The cross-linking mixture was filled into small cylindrical, rectangular, spherical, and shoe-shaped molds, and finally incubated at 37° C. Resulting recombinant resilin solids are shown inFIG.7. Example 6: Material Testing of Resilin Solids A resilin cylinder produced as described in Example 5 was subjected to a compression test using a rheometer. The recombinant resilin cylinder could be compressed from an initial height of 7.3 mm (avg width 5.4 mm) to less than 0.66 mm without any breakage. As shown inFIG.8, the cylinder returned to a height of 6.7 mm (avg width 5.6 mm) upon release of the compressive load. Example 7: Methods for Recovering Full-Length Recombinant Resilin from Whole Cell Broth Various recovery and separation techniques were used to purify Ds_ACB (SEQ ID NO: 1) that was produced in strains with a 3× FLAG tag (RMs1209) and without a 3× FLAG tag (RMs1221) according to Example 1 above. A first set of samples was prepared by centrifuging a whole cell broth to produce a first pellet of cells and a first supernatant, and extracting the first supernatant to produce a clear cell broth. The first supernatant was then precipitated using ammonium sulfate and centrifuged to produce a second pellet and second supernatant which was discarded. The second pellet was then re-suspended in PBS for dialysis. The dialyzed solution was then subject to high temperature to denature proteins other than Ds_ACB, which is stable at high temperatures. The denatured proteins were removed by centrifuging the dialyzed and denatured solution to produce a third pellet and third supernatant. The third supernatant was retained from the denatured solution, then coacervated by chilling the third supernatant to induce a phase separation into a dense lower layer containing the Ds_ACB and an upper layer. These samples are referred to in Table 2 below and elsewhere herein as the “CCB” samples. In some CCB samples, multiple coacervations were performed by retaining the lower layer and incubating the lower layer at a lower temperature to induce further phase separation. These CCB samples are respectively referred to in Table 2 below and elsewhere herein as the “first coacervation” and “second coacervation” samples. A second set of samples was prepared by centrifuging a whole cell broth to produce a first pellet of cells and protein proximal to the cells (e.g. adherent to the cells, on the surface of the cells) and/or insoluble protein (e.g. protein aggregates) and first supernatant, then discarding the first supernatant to obtain the first pellet. The first pellet was re-suspended in guanidine thiocyanate to solubilize Ds_ACB. The re-suspension was centrifuged again produce a second pellet and a second supernatant. The second supernatant was then dialyzed against PBS and subject to high temperature in order to denature proteins other than Ds_ACB and centrifuged to produce a third pellet and third supernatant. The third supernatant was subject to coacervation by chilling to yield phase separation into a dense lower layer containing Ds_ACB and an upper layer. These samples are referred to in Table 2 below and elsewhere herein as the “gel layer” samples. In some gel layer samples, multiple coacervations were performed by retaining the lower layer and incubating the lower layer at a lower temperature to induce further phase separation. These gel layer samples are referred to in Table 2 below and elsewhere herein as the “first coacervation” and “second coacervation” samples. A third set of samples was prepared by centrifuging a whole cell broth to produce a pellet and supernatant, then discarding the supernatant to obtain a pellet of cells and protein proximal to the cells (e.g. adherent to the cells, on the surface of the cells) and/or insoluble protein (e.g. protein aggregates). The pellet of cells was re-suspended in guanidine thiocyanate to solubilize the protein that was proximal to the cells. The re-suspension was centrifuged again produce a second pellet of cells and a second supernatant. The second supernatant was then precipitated with ammonium sulfate and centrifuged to produce a third pellet and third supernatant. The third pellet was suspended in guanidine thiocyanate, then dialyzed against PBS and subject to high temperature to denature proteins other than Ds_ACB and centrifuged to produce a fourth supernatant and fourth pellet. The fourth supernatant was then subject to coacervation by chilling to yield phase separation. These samples are referred to in Table 2 below and elsewhere herein as the “gel layer precipitated” samples. A single sample was produced by adding urea to a whole cell broth to solubilize the protein, then centrifuging the whole cell broth to produce a first pellet and first supernatant. The first supernatant was then precipitated using ammonium sulfate and centrifuged to produce a second pellet and second supernatant. The second supernatant was discarded and the second pellet was then re-suspended in guanidine thiocyanate and dialyzed against PBS, then subject to high temperature in order to denature proteins other than Ds_ACB and centrifuged again to produce a third pellet and a third supernatant. The third supernatant was then coacervated by chilling the third supernatant to induce a phase separation into a dense lower layer containing Ds_ACB and an upper layer. This sample is referred to in Table 2 below and elsewhere herein as the “Urea WCBE” sample. Another single sample was prepared by centrifuging a whole cell broth to produce a first pellet and first supernatant, then discarding the first supernatant to obtain a first pellet of cells and protein proximal to the cells (e.g. adherent to the cells, on the surface of the cells) and/or insoluble protein (e.g. protein aggregates). The first pellet of cells was re-suspended in guanidine thiocyanate to solubilize the protein. The re-suspension was centrifuged again to produce a second pellet of cells and a second supernatant. The second supernatant was then dialyzed against PBS and then centrifuged to produce a heavy phase of protein, a light phase of supernatant and a film separating the heavy phase from the light phase. The heavy phase of protein was then isolated by discarding the light phase and the film. This sample is referred to in Table 2 below and elsewhere herein as the “Dense layer” sample. Table 2 (below) lists the various combinations of strains and recovery techniques along with the relative amount of degradation seen in the gel pictured atFIG.9. As shown inFIG.9, samples E, F, G, K and L showed bands at approximately 110 kDa and minimal or faint bands at lower molecular weights (labeled in Table 2 as “Minimal”). Samples A, B, C, D, G, I and J had degradation products corresponding to bands at approximately 90, 30, 22, 17 and 12 kDa (labelled in Table 2 as “Substantial”). Among these, samples A and I also showed bands at approximately 110 kDa indicating the presence of full-length resilin. Accordingly, the “Gel layer” samples produced full-length resilin while the CCB samples produced degradation products sometimes in addition to full-length resilin (e.g. sample A) or without full-length resilin (e.g. samples C and D). The Urea WCBE sample only produced degradation products. CCB/gel layer precipitated indicates the combination of isolated material from both the CCB purification and the gel layer purification methods. TABLE 2Samples from recovery methods yielding full-length resilin and degradation productsSampleStrainFLAGDescriptionCoacervation110 kDa band?DegradationARMs1221−CCBFirstYesSubstantialBRMs1221−Urea WCBENoneNoSubstantialCRMs1221−CCBFirstNoSubstantialDRMs1221−CCBSecondNoSubstantialERMs1221−Gel layerFirstYesMinimalFRMs1221−Gel layerSecondYesMinimalGRMs1209+CCBFirstNoSubstantialHRMs1209+Gel layerFirstYesMinimalprecipitatedIRMs1209+CCB/gel layerFirstYesSubstantialprecipitatedJRMs1221−CCBFirstNoSubstantialKRMs1221−Gel layerFirstYesMinimalLRMs1221−Dense layerNoneYesMinimal To verify that the 110 kDa bands shown in samples A, I, E, F, G, K and L corresponded to the full-length resilin (SEQ ID NO: 1), the 110 kDa band in sample H (indicated inFIG.9with an arrow) was excised and sent for N-terminus sequencing by Edman degradation. Edman degradation is a cyclic procedure where amino acid residues are cleaved off one at a time and identified by chromatography. There are 3 steps in the cyclic procedure. In step1the PITC reagent is coupled to the N-terminal amino group under alkaline conditions. In step2the N-terminal residue is cleaved in acidic media. In step3, the PITC coupled residue is transferred to a flask, converted to a PTH-residue and identified by HPLC chromatography. The next cycle is then started for identification of the next N-terminal residue. Edman degradation analysis was performed on a Shimadazu PPSQ-33 sequencer and a PVDF membrane. FIG.10shows the full-lengthDrosophila sechelliaresilin sequence (Ds_ACB) that is expressed along with signal sequences that are later cleaved. The first sequence (italics), is an alpha mating factor precursor protein signal sequence (SEQ ID NO: 46) that is cleaved twice after transcription by a signal peptidase followed by cleavage with Kex2. The second sequence (bold) is an EAEA repeat that is cleaved by Ste13 (SEQ ID NO: 47). The third sequence (lower case) corresponds toDrosophila sechelliafull-length resilin (SEQ ID NO 1). The fourth sequence (bold and italicized) corresponds to a linker sequence (SEQ ID NO: 46). The fifth sequence (underlined) corresponds to the 3× FLAG tag (SEQ ID NO: 45). Edman sequencing confirmed that the N-terminus of the protein sequences at the approximately 110 kDa band corresponded to the full-length lengthDrosophila sechelliaresilin sequence. Specifically, the N-terminus sequencing showed that the N-terminus either corresponded to “EAEA” or “GRPE”, respectively the full-lengthDrosophila sechelliaresilin sequence with or without the EAEA repeat. Example 8: Quantifying the Stability of Crosslinked Resilin Resilin samples generated by the methods described in Example 7 with varying levels of degradation products and full-length resilin were subject to enzymatic cross-linking as described above with respect to Example 4. The stability of the cross-linked samples was assessed over time by determining the duration each cross-linked samples remained a solid through daily observation. Table 3 shows the time as a solid for each cross-linked sample. As shown in Table 3, samples comprising full-length resilin had a longer duration of stability than the samples that did not comprise full-length resilin. TABLE 3Stability of cross-linked resilinSample110 kDa band?DegradationTime as solidAYesSubstantial13 daysBNoSubstantial8 daysCNoSubstantial6 daysDNoSubstantial6 daysEYesMinimalN/AFYesMinimal27 daysGNoSubstantial8 daysHYesMinimal15 daysIYesSubstantial15 daysJNoSubstantial6 daysKYesMinimal13 daysLYesMinimal13 days ADDITIONAL CONSIDERATIONS The foregoing description of the embodiments of the disclosure has been presented for the purpose of illustration; it is not intended to be exhaustive or to limit the claims to the precise forms disclosed. Persons skilled in the relevant art can appreciate that many modifications and variations are possible in light of the above disclosure. The language used in the specification has been principally selected for readability and instructional purposes, and it may not have been selected to delineate or circumscribe the inventive subject matter. It is therefore intended that the scope of the disclosure be limited not by this detailed description, but rather by any claims that issue on an application based hereon. Accordingly, the disclosure of the embodiments is intended to be illustrative, but not limiting, of the scope of the invention, which is set forth in the claims. SEQUENCE LISTING SEQ IDNO: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 melliferaRSEPPVNSYLPPSGNGNGGGGGGSSNVYGPPGFDGQNGIGEGDNGRNGISNSYGVPTGGNGYNGDSSGNGRPGTNGGRNGNGNGRGNGYGGGQPSNSYGPPSNGHGGNGAGRPSSSYGAPGGGNGFAGGSNGKNGFGGGPSSSYGPPENGNGFNGGNGGPSGLYGPPGRNGGNGGNGGNGGRPSGSYGTPERNGGRLGGLYGAPGRNGNNGGNGYPSGGLNGGNGGYPSGGPGNGGANGGYPSGGSNGDNGGYPSGGPNGNGNGNGGYGQDENNEPAKYEFSYEVKDEQSGADYGHTESRDGDRAQGEFNVLLPDGRKQIVEYEADQDGFKPQIRYEGEANSQGYGSGGPGGNGGDNGYPSGGPGGNGYSSGRPNGGSDFSDGGYPSTRPGGENGGYRNGNNGGNGNGGYPSGNGGDAAANGGYQY16Apis 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.GHSNGGGSSFGGSAPSAPSQSYGAPSFGGQSSGGFGGHSSGGFGGHSSGGHPESTGGNGNGNGNGYSSGRPSSQYGPPQQQQQQQSFRPPSTSYGVPAAPSQSYGAPAQQHSNGGNGGYSSGRPSTQYGAPAQSNGNGFGNGRPSSSYGAPARPSTQYGAPSAGNGNGYAGNGNGRSYSNGNGNGHGNGHSNGNGNNGYSRGPARQPSQQYGPPAQAPSSQYGAPAQTPSSQYGAPAQTPSSQYGAPAQTPSSQYGAPAQTPSSQYGAPAPSRPSQQYGAPAPSRPSQQYGAPAQTPSSQYGAPAQTPSSQYGAPAQTPSSQYGAPAQTPSSQYGAPAQQPSSQYGAPAPSRPSQQYGAPAQQPSAQYGAPAQTPSSQYGAPAPSRPSQQYGAPAQAPSSQYGAPAPSSQYGAPAQQPSSQYGAPAQTPSSQYGAPSFGPTGGASFSSGNGNVGGSYQVSSTGNGFSQASFSASSFSPNGRTSLSAGGFSSGAPSAQSAGGYSSGGPSQVPATLPQSYSSNGGYNY21GlossinaRPEPPVNTYLPPSAGGGSGGGSPLAPSDTYGAPGVNGGGGGGGGPSSTYGAmorsitansPGSGGGNGNGGGGFGKPSSTYGAPGLGGGGNGGGRPSETYGAPSGGGGNGFGKPSSTYGAPNGGGGNGGPGRPSSTYGAPGSGGGNGGSGRPSSTYGAPGLGGGNGGSGRPSSMYGAPGLGGGNGGSGRPSSTYGAPGSGGGNGGSGRPSSTYGAPGSGGGNGGSGRPSSTYGAPGNGNGGNGFGRPSSTYGAPGSGGSNGNGKPSSTYGAPGSGGGGGRPSDSYGPPASGNGGRNGNGNGQSQEYLPPGQSGSGGGGGYGGGSGSGGSGGGGGGGYGGDQDNNVVEYEADQEGYRP QIRYEGDGSQGGFGGDGDGYSYEQNGVGGDGGGAGGAGGYSNGQNLGANGYS SGRPNGGNGGGRRGGGGGGGGSGGGQNLGSNGYS SGAPNGFGGGNGQGYSGGRSNGNGGGGGGRNGGRYRNGGGGGGGRNGGGSNGYNYDQPGSNGFGRGGGNGENDGSGYHY22Atta cephalotesRSEPPVNSYLHPGSDTSGTNGGRTDLSTQYGAPDFNNRGNGNSGATSFGGSGAGNGPSKLYDVPIRGNTGGNGLGQFRGNGFESGQPSSSYGAPKGGFGENRGNRGRPSTSYGVPDSNRNNRGGFGNGGSEARPSTSYGVPGANGNQGGFGSGSIGGRPSTSYGVPGANGNNGDSFRNGDIGGRPSTNYGAPGANGNHGGGNGGNGRPSNNYGVPGANGNTNGKGRLNGNSGGGPSNNYGSPNGFGKGLSTSYGSPNRGGNDNHYPSRGSFINGGINGYSSGSPNGNAGNFGHGDESFGRGGGEGENTGEGYNANAQEESTEPAKYEFSYKVKDQQTGSDYSHTETRDGDHAQGEFNVLLPDGRKQIVEYEADQDGFKPQIRYEGEANADGGYGSGLNDNNDGYSSGRPDSESGGFANSGFNGGSSNGGYPNGGPGERKLGGFNNGGSSGYQSGRSAGQSFGRDNAGDLNNDIGGYFSNSPNNIGDSDNANVGSNRQNDGNSGYQY23AnophelesKREAPLPPSGSYLPPSGGGGGGGGYPAAQTPSSSYGAPAGGAGGWGGNGNGdarlingiNGNGNGGRGGYSNGGGHSGSAPSQSYGAPSAPSQSYGAPSQSYGAPAAAPSQSYGAPSFGGNGGGASHGSGGFTGGHGGNGNGNGYSSGRPSSQYGPPQQQQQPQQQSFRPPSTSYGVPAAPSSSYGAPSANGFSNGGRPSSQYGAPAPQSNGNEFGAPRPSSSYGAPSRPSTQYGAPSNGNGNGYAGHGNGNGHGNGNGHSNGNGNGYNRGPARQPSSQYGPPSQGPPSSQYGPPSQYGPPSSGTSFIAYGPPSQGPPSSQYGAPAPSRPSSQYGAPAQTPSSQYGAPAQTPSSQYGPPRQSSPQFGAPAPRPPSSQYGAPAQAPSSQYGAPAQTPSSQYGAPAQAPSSQYGAPAPSRPSSQYGVPAQAPSSQYGAPAQAPSSQYGAPAQTPSSQYGAPSFGSTGGSSFGGNGGVGGSYQTASSGNGFSQASFSASSFSSNGRSSQSAGGYSSGGPSQVPATIPQQYSSGGGSYSSGGHSQVPATLPQQYSSNGGYNY24AcromyrmexRSEPPVNSYLPPGPGTSGANGGQTDLSIQYRASDFNNRGNVNGNSGATSFGechinatiorGPGASNGPSKLYDVPIGGNAGGNGLGQFRGNGFEGGQPSSSYGAPNGGFGENRGNGGKPSTSYGVPDSNGNNRGGFGNGGSEGRPSTSYGLPDASRNNGNGFGNVGNEDKPSTNYGIPANGNKVSGFGNVGSEGRPSTSYGVPGANGNQGFGSGGIGGRPSTSYGVPGVNGNNGGGFENVGRPSTSYGTPDARGNNGGSFRNGDIGGRPSTNYGIPGANGNHGGGNGGNGRPSSNYGVPGGNGNTNGKGRFNGNSGGRPSNSYGSPNGFGKGLSTSYSPSNRDGNGNHYPSGDSNRGSFVNGGINGYPSGSPNGNAGNFRHGDESFGRGGEGGGRSTGEGYNANAQEESTEPAKYEFSYKVKDQQTGSDYSHTETRDGDHAQGEFNVLLPDGRKQIVEYEADQDGFKPQIRYEGEANADGEYDSGGLNDNNDGYSSGRPGSESGGFANNSGFNGGSSNGGYPSGGSGEGKLGFNSGGNSGYQSGRPAGQSFGRDNAGDLSNDIGGFSNSPNNIGGDNANVGSNRQNGGNSGYQY25AcyrthosiphonESPYGGGSSNSNGNGRNGGYGGKGQYGGGNGGGVGSSSASPFFSGANQYGSpisumQSGLSGAANNRYPSFGSKFGGNKGSYGGSSSRNNGRYGSGSASGYGSGSSGGLGSTGRSTGGYGGGSSGSYGSGSSGSLGSSTGSNGIYGAGSSGGFGSGSSGSYGGGSSGGFGSGSSGSYGGGSSGGFGSGSSGSYGGGSSGGFGSGSSGSYGGGSSGGFGSGSSGNYGSGSSGSYGSGGGGLGGASSGNNDGYGAGGSGSYDQLGGANGNGLGGSGNDPLSEPANYEFSYEVNAPESGAIFGHKESRQGEEATGVYHVLLPDGRTQIVEYEADEDGYKPKITYTDPVGGYAGDRQSGNSYGGNGGFGGSGSLGGSGGNLGGLYNGGGSSNNGAGYGGSSSSLGSRYGGSGGSSGSGVGGGYGGSGSSSGGIGSSYGGSGSLSGGLGGGYGGSGSSSGGLGGGYGGSGGSSGGGFGGLGGSGGSSGSGYGGSGSSSGGLGNSYGGSGSSNGGLGGGYSGSGGSSGGLGGGYGASSGSSGSGLGGGYGGSGSSSGGLGSGYGGLGSSSGGLGGGYGGSGSSSGGLGGGYGGSGSSNGGIGGGYGGSSGSSGGLGGGYGGSGSSSGGLGGGYGGSGGSNSGLGSSYGGSGSTNGGLGGGYGGLGSSSGGLGGGYGGSGGSNGGIGGGYGGSSGSGGSQGSAYGGSGSSSGSQGGGYGGSGSSSGGLGGGYGSSSGSSSGLGGSYGSNRNGLGSGSSYS26DrosophilaRPEPPVNSYLPPSPGDSYGAPGQGQGQGQGGFGGKPSDSYGAPGAGNGNGNvirilis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rosophilaRPEPPVNSYLPPSDSYGAPGQSGPGGRPSDSYGAPGGGNGGRPSDSYGAPGerecta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utzomyiaRPEPPANTYLPPSSSYAAPGQQGGSGFGGGGGSGGSGGFGQPGAFGRPSSSlongipalpis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hodnius prolixusKRDDPLRRFLAPLVGGGNGSGGGGGGYNYNKPANGLSLPGGGGALPPATSYGVPDRPAPVPSSPPSSSYGAPQPSPNYGAPSSSYGAPSQQPSRSYGAPSQGPSTSYSQRPSSSYGAPAPQTPSSSYGAPAQQPSGSYGAPSGGGGSSGYTGGAQRPSGSYGAPSQGGPSGNYGPPSQQPSSNYGAPSQTPSSNYGAPAQRPSTSYGAPSQPPSSSYGSPPQRASGYPSSSSGPSNGYSPPAQRPSSSYGPPSQQPASSYGAPSQTPSSNYGPPAPIPSSNYGAPSQPPSKPSAPSSSYGTPSQTPSTSYGAPSQAPSSSYGAPSRPSPPSSSYGAPSQGPSSSYGPPSRPSQPSSPSSGYGAPSQGPSSSYGAPSRPSSPSSSYGAPPSSSYGAPSRPSPPSSSYGAPSQGPSSSYGPPSRPSQPSSPSSGYGAPSQGPSSSYGAPSRPSSPSSSYGAPPSSSYGAPSRPSPPSSSYGAPSQGPSSSYGPPSRPSQPSSPSSSYGAPSQGPSSSYGAPSRPSPPSSSYGAPSQGPSSSYGPPSRPSQPSSTYGVPSGGRPSTPSSSYGAPPQALSSTYGAPSGRPGAPSQKPSSSYGAPSLGGNASRGPKSSPPSSSYGAPSVGTSVSSYAPSQGGAGGFQSSRPSSSYGAPSTGPSSTYGPPSQPPSSSYGVPSQPPSSNYGVPSQGVSGSVGSSSPSSSYGAPSQIPSSSYGAPSQSSIGGFGSSRPSSSYGAPPQAPSSSYSAPLRAPSTSYGAPSGGSGSNFGSKPSTNYGAPSQPPSTNYGPPSQPPSSSYGTPSRAPSPTYSTPQSSGTSFGSRPSSSYGVPSQPTTNYGAPSQTPSSNYGAPPASSAPSSTYGRPSQSPSSSYGAPSPSSSSSSYESPSQPPSSSYGAPSQGPSSSYGAPSRPSSTYGAPSPSSPSTNYGAPAPSSNYGTPAQDLTGSYAAPSQPPSAGYGAPSGQPSSGGKQNFQVKNPFAGQTHQVYPAVSSISFGLPSQSFNTAIQGQEPSQSYGAPTASSPSSSYGAPTGTGSSQPGQSYASNGGYSYS30Rhodnius prolixusQPPFNHYLPAARGSGSNSAQYTAPSSKFGTSTGQYGQPPSEVPRGLQQGSYAEDVHSSRSVNPSSQNGIPSGHFSSLSSNYGAPSSDYSRSFLRYGTLSNKYGVPNSALGSLSSRNNKTPATQLSYQPSSHYDSRSTSEDQFISSRVSDSQYGASSVRRFLPSSQYSTPSSQYGTPSSQYGTPSSQYGTPSSQYGTPSSQYGTPSSQYGTPSSQYGTPSSQYGTPSSQYGTPSSQYGTPSSPPSQYGGPYSMRTSAPNSQYGTPSSFRTSPSSQFGSSSAHSSSLSKFRSVPSSPYGTLSAIRSTHSSQYGTPSSFSDSTSSSHNGLPSHYPGSGFSGSSVNDQKSYTGNVFGQSHSRVANGDQHARSYTLAGGNEISEPAKYDFNYDVSDGEQGVEFGQEESRDGEETNGSYHVLLPDGRRQRVQYTAGQYGYKPTISYENTGTLTTGRQQFSNGFYNVQQSGSESQEHLGRSTGQNSYGGSNGYESGVGYQSGVGRRSRPAGSY31Solenopsis invictaRSEPPINSYLPPRAGSSGANGGRTDLTTQYGAPDFNNGGGATSFSGNGAGDGPSKLYDVPVRGNAGGNGLGRGNGFGGGQPSSSYGAPNGGSNENRGNGGRPSTSYGVPGANGNNGGGFGNGGDKGRPSTSYGVPDASGSSQGSFGNVGNGGRPSTNYGVPGANGNGGGFGNAANEGKPSTSYGVPGANGNSQGGFGNGGRPSTGYGVPGANGNNGGGFGGRPSTSYGAPGANGNHRGGNGGNASPSTNYGVPGGNNGNTNGKGRFNGGNSGGGPSNNYGVPNENAFGGGLSTSYGPPSRGGNGNSGYPSGGSNGGSFVNNGANGYPSGGPNGNAGNFGDGRGGKGGGSSGEGYNDNAQEGSTEPAKYEFSYKVKDQQTGSEYSHTETRDGDRAQGEFNVLLPDGRKQIVEYEADQDGFKPQIRYEGEANAGGGYSSGGSNDNNDGYSSGRPGSEAGGFANNSGFNGSGTNGGRSSGGPGDGNPGGFNSGGGGGYQSGRPAGQSFGRDNDGGLSGDIGGYFANSPSNNIGGSDSANVGSNRQNGGNGGYQY32CulexKREAPLPGGSYLPPSNGGGAGGYPAAGPPSGSYGPPSNGNGNGNGAGGYPSquinquefasciatusAPSQQYGAPAGGAPSQQYGAPSNGNGGAGGYPSAPSQQYGAPNGNGNGGFGGRPQAPSQQYGAPSNGNGGARPSQQYGAPNGGNGNGRPQTPSSQYGAPSGGAPSSQYGAPSGGAPSQQYGAPNGGNGNGRPQTPSSQYGAPSGGAPSQQYGAPNGGNGNGRPQTPSSQYGAPSGGAPSSQYGAPSGGAPSSQYGAPAGGAPSSQYGAPAGGAPSSQYGAPAGGAPSSQYGAPAGGAPSSQYGAPAGGAPSSQYGAPSSQYGAPAGGAPSSQYGAPAGGAPSSQYGAPSGGAPSSQYGAPSGGAPSSQYGAPAGGAPSSQYGAPSGGAPSS33BactroceraRPEPPVNSYLPPSANGNGNGGGRPSSQYGAPGLGSNSNGNGNGNGGGRPSScucurbitaeQYGVPGLGGNGNGNGNGGGGGRPSSSYGAPGLGGNGNGNGNGGGRPSSQYGVPGLGGNGNGNGNGNGGGRPSSTYGAPGLRGNGNGNGNGNGRPSSTYGAPGLGGNGNGNGNGNGRPSSTYGAPGLGGNGNGNGNGNGRPSSTYGAPGLNGNGLGGGQKPSDSYGPPASGNGNGYSNGGNGNGNGGGRPGQEYLPPGRNGNGNGNGGRGNGNGGGANGYDYSQGGSDSGESGIVDYEADQGGYRPQIRYEGEANNGAGGLGGGAGGANGYDYEQNGNGLGGGNGYSNGQDLGSNGYSSGRPNGNGNGNGNGNGNGYSGRNGKGRNGNGGGQGLGRNGYSDGRPSGQDLGDNGYASGRPGGNGNGNGGNGNGYSNGNGYSNGNGNGTGNGGGQYNGNGNGYSDGRPGGQDNLDGQGYSSGRPNGFGPGGQNGDNDGNGYRY34TrichogrammaRPEPPVNSYLPPGQGGQGGFGGSGGRPGGGSPSNQYGPPNFQNGGGQNGGSpretiosumGFGGNGNGNSFGPPSNSYGPPEFGSPGAGSFGGGRPQDTYGPPSNGNGNGNGFGGNGNGGGRPSSRPSDSYGPPSSGNGFGGGNSGRPSESYGPPQNGGGSGNGNQGGGNGFGNGGGRGGQGKPSDSYGPPNSGNRPGSSNGGGQQQNGFGGGNGGRPSNTYGPPGGGNGGGRPGGSSGGFGGQNGGRPSDSYGPPSNGNGNGGRPSNNYGPPNSGGGNGNGFGGSNGKPSNSYGPPSNGNGGGFGGSNGRPSNSYGPPSGGNGGGFGGSSAVGRPGNSGSPSSSGSGFGGNGGASRPSSSYGPPSNGGGFGNGGGSNGRPSSSYGPPNSGSNGGGFGGQNGNGRQNGNNGQGGFGGQPSSSYGPPSNGNGFGGGGGSNGYPQNSQGGNGNGFGQGSGGRPSSSYGPPSNGGGGGDNGYSSGGPGGFGGQPQDSYGPPPSGAVDGNNGFSSGGSSGDNNGYSSGGPGGNGFEDGNDEPAKYEFSYEVKDEQSGSSFGHTEMRDGDRAQGEFNVLLPDGRKQIVEYEADQDGFKPQIRYEGEANTGGAGGYPSGGPGGQGGNGNGGYPSGGPSNGGFGGQNGGGNGGYPSGGPSGGGFGGQNGGSGGYPSGGPSGGGFGGQGGFGGQNSGGNGGYSSGGPASGGFGGQNGGNGGYPSGGPSGGGFGGQGGFGGQNSGGNGGYPSGGPSSGGFGGQNGGGGGNYPAGSGGDAEANGGYQYS35DrosophilaQSGAGGRPSDTYGAPGGGNGGRPSDSYGAPGQGQGQGQGQGGYGGKPSDSYsechelliaGAPGGGNGNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGGRPSDTYGAPGGGGNGNGGRPSSSYGAPGQGQGNGNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGGRPSDTYGAPGGGNNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGNGSGGRPSSSYGAPAQGQGGFGGRPSDSYGAPGQNQKPSDSYGAPGSGNGSAGRPSSSYGAPGSGPGGRPSDSYGP36DrosophilaQSGAGGRPSDTYGAPGGGNGGRPSDSYGAPGQGQGQGQGQGGYGGKPSDSYsechelliaGAPGGGNGNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGGRPSDTYGAPGGGGNGNGGRPSSSYGAPGQGQGNGNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGGRPSDTYGAPGGGNNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGNGSGGRPSSSYGAPAQGQGGFGGRPSDSYGAPGQNQKPSDSYGAPGSGNGSAGRPSSSYGAPGSGPGGRPSDSYGP37DrosophilaQSGAGGRPSDTYGAPGGGNGGRPSDSYGAPGQGQGQGQGQGGYGGKPSDSYsechelliaGAPGGGNGNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGGRPSDTYGAPGGGGNGNGGRPSSSYGAPGQGQGNGNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGGRPSDTYGAPGGGNNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGNGSGGRPSSSYGAPAQGQGGFGGRPSDSYGAPGQNQKPSDSYGAPGSGNGSAGRPSSSYGAPGSGPGGRPSDSYGP38DrosophilaQSGAGGRPSDTYGAPGGGNGGRPSDSYGAPGQGQGQGQGQGGYGGKPSDSYsechelliaGAPGGGNGNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGGRPSDTYGAPGGGGNGNGGRPSSSYGAPGQGQGNGNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGGRPSDTYGAPGGGNNGGRPSSSYGAPGGGNGGRPSDTYGAPGGGNGNGSGGRPSSSYGAPAQGQGGFGGRPSDSYGAPGQNQKPSDSYGAPGSGNGSAGRPSSSYGAPGSGPGGRPSDSYGP39DrosophilaYSSGRPGNGNGNGNGGYSSGRPGGQDLGPSGYSGGRPGGQDLGAGGYSNVKsechelliaPGGQDLGPGGYSGGRPGGQDLGRDGYSGGRPGGQDLGAGAYSNGRPGGNGNGGSDGGRVIIGGRVIGGQDGGDQGYSGGRPGGQDLGRDGYSSGRPGGRPGGNGQDSQDGQ40DrosophilaYSSGRPGNGNGNGNGGYSSGRPGGQDLGPSGYSGGRPGGQDLGAGGYSNVKsechelliaPGGQDLGPGGYSGGRPGGQDLGRDGYSGGRPGGQDLGAGAYSNGRPGGNGNGGSDGGRVIIGGRVIGGQDGGDQGYSGGRPGGQDLGRDGYSSGRPGGRPGGNGQDSQDGQ41DrosophilaYSSGRPGNGNGNGNGGYSSGRPGGQDLGPSGYSGGRPGGQDLGAGGYSNVKsechelliaPGGQDLGPGGYSGGRPGGQDLGRDGYSGGRPGGQDLGAGAYSNGRPGGNGNGGSDGGRVIIGGRVIGGQDGGDQGYSGGRPGGQDLGRDGYSSGRPGGRPGGNGQDSQDGQ42DrosophilaYSSGRPGNGNGNGNGGYSSGRPGGQDLGPSGYSGGRPGGQDLGAGGYSNVKsechelliaPGGQDLGPGGYSGGRPGGQDLGRDGYSGGRPGGQDLGAGAYSNGRPGGNGNGGSDGGRVIIGGRVIGGQDGGDQGYSGGRPGGQDLGRDGYSSGRPGGRPGGNGQDSQDGQ43DrosophilaYSSGRPGNGNGNGNGGYSSGRPGGQDLGPSGYSGGRPGGQDLGAGGYSNVKsechelliaPGGQDLGPGGYSGGRPGGQDLGRDGYSGGRPGGQDLGAGAYSNGRPGGNGNGGSDGGRVIIGGRVIGGQDGGDQGYSGGRPGGQDLGRDGYSSGRPGGRPGGNGQDSQDGQ44DrosophilaYSSGRPGNGNGNGNGGYSSGRPGGQDLGPSGYSGGRPGGQDLGAGGYSNVKsechelliaPGGQDLGPGGYSGGRPGGQDLGRDGYSGGRPGGQDLGAGAYSNGRPGGNGNGGSDGGRVIIGGRVIGGQDGGDQGYSGGRPGGQDLGRDGYSSGRPGGRPGGNGQDSQDGQ453X FLAGGDYKDDDDKDYKDDDDKDYKDDDDK46Alpha matingMRFPSIFTAVLFAASSALAAPVNTTTEDETAQIPAEAVIGYSDLEGDFDVAfactor precursorVLPFSNSTNNGLLFINTTIASIAAKEEGVSLEKRprotein sequence47EA repeatEAEA48LinkerSG
103,724
11858972
DETAILED DESCRIPTION OF THE INVENTION The present invention relates to the provision of new compounds for the treatment of cancer. The Micropeptides of SEQ ID NO: 1, 2 and 3 In a first aspect the invention relates to a micropeptide comprising the amino acid sequence selected from the group consisting of SEQ ID NO: 1, 2 and 3 or a functionally equivalent variant thereof. The term “micropeptide”, as used herein, is used to refer to peptides having from 4 to 100 amino acids, preferably from 20 to 90 amino acids. They can be of natural or synthetic origin. In nature, they are coded by an open reading frame contained in a lncRNA, in a RNA molecule annotated as non-protein coding, or in a RNA molecule annotated as “intergenic region”. The term “open reading frame” corresponds to a nucleotide sequence in a DNA or RNA molecule that may potentially encode a peptide or a protein. Said open reading frame begins with a start codon (the start codon generally encoding a methionine), followed by a series of codons (each codon encoding an amino acid), and ends with a stop codon (the stop codon not being translated). The term “long non-coding RNA” or “lncRNA” as used herein includes a transcript longer than 200 nucleotides and annotated as non-coding. The terms “polypeptide” and “peptide” are used interchangeably herein to refer to polymers of amino acids of any length. The polymer can be linear or branched, it can comprise modified amino acids, and it can be interrupted by non-amino acids. The term “amino acid” refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Furthermore, the term “amino acid” includes both D- and L-amino acids (stereoisomers). The term “natural amino acids” or “naturally occurring amino acid” comprises the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine and phosphothreonine; and other unusual amino acids including, but not limited to, 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine and ornithine. As used herein the term “non-natural amino acid” or “synthetic amino acid” refers to a carboxylic acid, or a derivative thereof, substituted at position “a” with an amine group and being structurally related to a natural amino acid. Illustrative non-limiting examples of modified or uncommon amino acids include 2-aminoadipic acid, 3-aminoadipic acid, beta-alanine, 2-aminobutyric acid, 4-aminobutyric acid, 6-aminocaproic acid, 2-aminoheptanoic acid, 2-aminoisobutyric acid, 3-aminoisobutyric acid, 2-aminopimelic acid, 2,4-diaminobutyric acid, desmosine, 2,2′-diaminopimelic acid, 2,3-diaminopropionic acid, N-ethylglycine, N-ethylasparagine, hydroxy lysine, alio hydroxy lysine, 3-hydroxyproline, 4-hydroxyproline, isodesmosine, alloisoleucine, N-methylglycine, N-methyliso leucine, 6-N-methyl-lysine, N-methylvaline, norvaline, norleucine, ornithine, etc Tables 1 and 2 below list naturally occurring amino acids (Table 1) and nonconventional or modified amino acids (Table 2) which can be used within the present invention. TABLE 1Three-LetterOne-letterAbbreviationSymbolalanineAlaAArginineArgRAsparagineAsnNAspartic acidAspDCysteineCysCGlutamineGlnQGlutamic AcidGluEglycineGlyGHistidineHisHisoleucineIieIleucineLeuLLysineLysKMethionineMetMphenylalaninePheFProlineProPSerineSerSThreonineThrTtryptophanTrpWtyrosineTyrYValineValVAny amino acidXaaXas above TABLE 2Non-conventional amino acidCodeNon-conventional amino acidCodeα-aminobutyric acidAbuL-N-methylalanineNmalaα -amino- α -methylbutyrateMgabuL-N-methylarginineNmargaminocyclopropane-CproL-N-methylasparagineNmasncarboxylateL-N-methylaspartic acidNmaspaminoisobutyric acidAibL-N-methylcysteineNmcysaminonorbornyl-NorbL-N-methylglutamineNmgincarboxylateL-N-methylglutamic acidNmglucyclohexylalanineChexaL-N-methylhistidineNmhiscyclopentylalanineCpenL-N-methylisolleucineNmileD-alanineDalL-N-methylleucineNmleuD-arginineDargL-N-methyllysineNmlysD-aspartic acidDaspL-N-methylmethionineNmmetD-cysteineDcysL-N-methylnorleucineNmnleD-glutamineDglnL-N-methylnorvalineNmnvaD-glutamic acidDgluL-N-methylornithineNmornD-histidineDhisL-N-methylphenylalanineNmpheD-isoleucineDileL-N-methylprolineNmproD-leucineDleuL-N-methylserineNmserD-lysineDlysL-N-methylthreonineNmthrD-methionineDmetL-N-methyltryptophanNmtrpD-ornithineDornL-N-methyltyrosineNmtyrD-phenylalanineDpheL-N-methylvalineNmvalD-prolineDproL-N-methylethylglycineNmetgD-serineDserL-N-methyl-t-butylglycineNmtbugD-threonineDthrL-norleucineNleD-tryptophanDtrpL-norvalineNvaD-tyrosineDtyrα -methyl-aminoisobutyrateMaibD-valineDvalα -methyl-Υ-aminobutyrateMgabuD- α -methylalanineDmalaα ethylcyclohexylalanineMchexaD- α -methylarginineDmargα -methylcyclopentylalanineMcpenD- α -methylasparagineDmasnα -methyl- α -napthylalanineManapD- α -methylaspartateDmaspα - methylpenicillamineMpenD- α -methylcysteineDmcysN-(4-aminobutyl)glycineNgluD- α -methylglutamineDmglnN-(2-aminoethyl)glycineNaegD- α -methylhistidineDmhisN-(3-aminopropyl)glycineNornD- α -methylisoleucineDmileN- amino- α -methylbutyrateNmaabuD- α -methylleucineDmleuα -napthylalanineAnapD- α -methyllysineDmlysN-benzylglycineNpheD- α -methylmethionineDmmetN-(2-carbamylethyl)glycineNglnD- α -methylornithineDmornN-(carbamylmethyl)glycineNasnD- α -methylphenylalanineDmpheN-(2-carboxyethyl)glycineNgluD- α -methylprolineDmproN-(carboxymethyl)glycineNaspD- α -methylserineDmserN-cyclobutylglycineNcbutD- α -methylthreonineDmthrN-cycloheptylglycineNchepD- α -methyltryptophanDmtrpN-cyclohexylglycineNchexD- α -methyltyrosineDmtyN-cyclodecylglycineNcdecD- α -methylvalineDmvalN-cyclododeclglycineNcdodD- α -methylalnineDnmalaN-cyclooctylglycineNcoctD- α -methylarginineDnmargN-cyclopropylglycineNcproD- α -methylasparagineDnmasnN-cycloundecylglycineNcundD- α -methylasparatateDnmaspN-(2,2-diphenylethyl)glycineNbhmD- α -methylcysteineDnmcysN-(3,3-diphenylpropyl)glycineNbheD-N-methylleucineDnmleuN-(3-indolylyethyl) glycineNhtrpD-N-methyllysineDnmlysN-methyl-Υ-aminobutyrateNmgabuN-methylcyclohexylalanineNmchexaD-N-methylmethionineDnmmetD-N-methylornithineDnmornN-methylcyclopentylalanineNmcpenN-methylglycineNalaD-N-methylphenylalanineDnmpheN-methylaminoisobutyrateNmaibD-N-methylprolineDnmproN-(1-methylpropyl)glycineNileD-N-methylserineDnmserN-(2-methylpropyl)glycineNileD-N-methylserineDnmserN-(2-methylpropyl)glycineNleuD-N-methylthreonineDnmthrD-N-methyltryptophanDnmtrpN-(1-methylethyl)glycineNvaD-N-methyltyrosineDnmtyrN-methyla-napthylalanineNmanapD-N-methylvalineDnmvalN-methylpenicillamineNmpenΥ-aminobutyric acidGabuN-(p-hydroxyphenyl)glycineNhtyrL-t-butylglycineTbugN-(thiomethyl)glycineNcysL-ethylglycineEtgpenicillaminePenL-homophenylalanineHpheL- α -methylalanineMalaL- α -methylarginineMargL- α -methylasparagineMasnL- α -methylaspartateMaspL- α -methyl-t-butylglycineMtbugL- α -methylcysteineMcysL-methylethylglycineMetgL- α thylglutamineMglnL- α -methylglutamateMgluL- α -methylhistidineMhisL- α -methylhomo phenylalanineMhpheL- α -methylisoleucineMileN-(2-methylthioethyl)glycineNmetD-N-methylglutamineDnmglnN-(3-guanidinopropyl)glycineNargD-N-methylglutamateDnmgluN-(1-hydroxyethyl)glycineNthrD-N-methylhistidineDnmhisN-(hydroxyethyl)glycineNserD-N-methylisoleucineDnmileN-(imidazolylethyl)glycineNhisD-N-methylleucineDnmleuN-(3-indolylyethyl)glycineNhtrpD-N-methyllysineDnmlysN-methyl-Υ-aminobutyrateNmgabuN-methylcyclohexylalanineNmchexaD-N-methylmethionineDnmmetD-N-methylornithineDnmornN-methylcyclopentylalanineNmcpenN-methylglycineNalaD-N-methylphenylalanineDnmpheN-methylaminoisobutyrateNmaibD-N-methylprolineDnmproN-(1-methylpropyl)glycineNileD-N-methylserineDnmserN-(2-methylpropyl)glycineNleuD-N-methylthreonineDnmthrD-N-methyltryptophanDnmtrpN-(1-methylethyl)glycineNvalD-N-methyltyrosineDnmtyrN-methyla-napthylalanineNmanapD-N-methylvalineDnmvalN-methylpenicillamineNmpenΥ-aminobutyric acidGabuN-(p-hydroxyphenyl)glycineNhtyrL-t-butylglycineTbugN-(thiomethyl)glycineNcysL-ethylglycineEtgpenicillaminePenL-homophenylalanineHpheL- α -methylalanineMalaL- α -methylarginineMargL- α -methylasparagineMasnL- α -methylaspartateMaspL- α -methyl-t-butylglycineMtbugL- α -methylcysteineMcysL-methylethylglycineMetgL- α -methylglutamineMglnL- α -methylglutamateMgluL- α ethylhistidineMhisL- α -methylhomophenylalanineMhpheL- α thylisoleucineMileN-(2-methylthioethyl)glycineNmetL- α -methylleucineMleuL- α -methyllysineMlysL- α -methylmethionineMmetL- α -methylnorleucineMnleL- α -methylnorvalineMnvaL- α -methylornithineMornL- α -methylphenylalanineMpheL- α -methylprolineMproL- α -methylserinemserL- α -methylthreonineMthrL- α ethylvalineMtrpL- α -methyltyrosineMtyrL- α -methylleucineMvalL-N-methylhomophenylalanineNmhpheNnbhmN-(N-(2,2-diphenylethyl)N-(N-(3,3-diphenylpropyl)carbamylmethyl-glycineNnbhmcarbamylmethyl(1)glycineNnbhe1-carboxy-1-(2,2-diphenylNmbcethylamino)cyclopropane “Functionally equivalent variant” is understood to mean all those micropeptides derived from the sequences SEQ ID NO: 1, 2 or 3, by modification, substitution, insertion and/or deletion of one or more amino acids, whenever the function is substantially maintained. Preferably, “variants” of sequences SEQ ID NO:1, 2 or 3 are (i) micropeptides in which one or more amino acid residues are substituted by a conserved or non-conserved amino acid residue (preferably a preserved amino acid residue) and such substituted amino acid may be coded or not by the genetic code, (ii) micropeptides in which there is one or more modified amino acid residues, for example, residues modified by substituent bonding, (iii) micropeptides resulting from alternative processing of a similar mRNA, (iv) micropeptide fragments and/or (v) micropeptides resulting from the fusion of sequences SEQ ID NO: 1, 2 or 3 or of the variants defined above in (i) to (iii) with another polypeptide, such as a secretory leader sequence, a sequence being used for purification (for example, His tag), for detection (for example, Sv5 epitope tag) or for delivery to a specific target cell. The fragments include polypeptides generated through proteolytic cut (including multisite proteolysis) of an original sequence. The functionally equivalent variant may be the result of a post-translationally or chemically modification. Such variants will be apparent to those skilled in the art. The variants of sequences SEQ ID NO: 1, 2 or 3 may be both natural and artificial. The expression “natural variant” relates to all those variants of human SEQ ID NO: 1, 2 or 3 which appear naturally in other species, i.e. the orthologues of SEQ ID NO: 1. 2 or 3. A functionally equivalent variant of sequences SEQ ID NO: 1, 2 or 3 can be an amino acid sequence derived from sequences SEQ ID NO: 1, 2 or 3 comprising the addition, substitution or modification of one or more amino acid residues. By way of illustration, functionally equivalent variants of the sequences SEQ ID NO: 1, 2 or 3 include sequences comprising the addition of 1 amino acid, 2 amino acids, 3 amino acids, 4 amino acids, 5 amino acids, 10 amino acids, 15 amino acids, 20 amino acids, 25 amino acids, 30 amino acids, 35 amino acids, 40 amino acids, 45 amino acids, 50 amino acids, 60 amino acids, 70 amino acids, 80 amino acids, 90 amino acids, 100 amino acids, 150 amino acids, 200 amino acids, at least 500 amino acids, at least 1000 amino acids or more at the amino terminus of the sequence SEQ ID NO: 1, 2 or 3, and/or comprising the addition of 1 amino acid, 2 amino acids, 3 amino acids, 4 amino acids, 5 amino acids, amino acids, 1 1 amino acids, 12 amino acids, 13 amino acids, 14 amino acids, 15 amino acids, 20 amino acids, 25 amino acids, 30 amino acids, 35 amino acids, 40 amino acids, 45 amino acids, 50 amino acids, 60 amino acids, 70 amino acids, 80 amino acids, 90 amino acids, 100 amino acids, 150 amino acids, 200 amino acids, at least 500 amino acids, at least 1000 amino acids or more at the carboxy terminus of the sequences SEQ ID NO: 1, 2 or 3, and maintaining at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 100% of the activity of the sequences SEQ ID NO: 1, 2 or 3. The term “conservative substitution” or “conserved amino acid residue” as used herein, refers to the replacement of an amino acid present in the native sequence in the peptide with a naturally or non-naturally occurring amino acid or a peptidomimetics having similar steric properties. Where the side-chain of the native amino acid to be replaced is either polar or hydrophobic, the conservative substitution should be with a naturally occurring amino acid, a non-naturally occurring amino acid or with a peptidomimetic moiety which is also polar or hydrophobic (in addition to having the same steric properties as the side-chain of the replaced amino acid). As naturally occurring amino acids are typically grouped according to their properties, conservative substitutions by naturally occurring amino acids can be easily determined bearing in mind the fact that in accordance with the invention replacement of charged amino acids by sterically similar non-charged amino acids are considered as conservative substitutions. For producing conservative substitutions by non-naturally occurring amino acids it is also possible to use amino acid analogs (synthetic amino acids) well known in the art. When affecting conservative substitutions the substituting amino acid should have the same or a similar functional group in the side chain as the original amino acid. The phrase “non-conservative substitutions” or “non-conserved amino acid residue” as used herein refers to replacement of the amino acid as present in the parent sequence by another naturally or non-naturally occurring amino acid, having different electrochemical and/or steric properties. Thus, the side chain of the substituting amino acid can be significantly larger (or smaller) than the side chain of the native amino acid being substituted and/or can have functional groups with significantly different electronic properties than the amino acid being substituted. Examples of non-conservative substitutions of this type include the substitution of phenylalanine or cycohexylmethyl glycine for alanine, isoleucine for glycine, or —NH—CH[(—CH2)5-COOH]—CO— for aspartic acid. Those non-conservative substitutions which fall under the scope of the present invention are those which still constitute a peptide having anti-proliferative properties. The peptides of the present invention may also comprise non-amino acid moieties, such as for example, hydrophobic moieties (various linear, branched, cyclic, polycyclic or heterocyclic hydrocarbons and hydrocarbon derivatives) attached to the peptides; various protecting groups, especially where the compound is linear, which are attached to the compound's terminals to decrease degradation. Suitable protecting functional groups are described in Green and Wuts, “Protecting Groups in Organic Synthesis”, John Wiley and Sons, Chapters 5 and 7, 1991. Chemical (non-amino acid) groups present in the compound may be included in order to improve various physiological properties such; decreased degradation or clearance; decreased repulsion by various cellular pumps, improve immunogenic activities, improve various modes of administration (such as attachment of various sequences which allow penetration through various barriers, through the gut, etc.); increased specificity, increased affinity, increased stability, bioavailability, solubility, decreased toxicity and the like. The variants of the invention can encompass native peptides (either degradation products, synthetically synthesized peptides or recombinant peptides), analogs, derivatives, salts, retro-inverso isomers, mimics, mimetics, or peptidomimetics (typically, synthetically synthesized peptides), as well as peptoids and semipeptoids. “Analog”, “derivative” and “mimetic” include molecules which mimic the chemical structure of a peptidic structure and retain the functional properties of the peptidic structure. Approaches to designing peptide analogs, derivatives and mimetics are known in the art. For example, see Farmer, P. S. in Dmg Design (E. J. Ariens, ed.) Academic Press, New York, 1980, vol. 10, pp. 119-143; Ball, J. B. and Alewood, P. F. (1990) J. Mol. Recognition 3:55. Morgan, B. A. and Gainor, J. A. (1989) Ann. Rep. Med. Chem. 24:243; and Freidinger, R. M. (1989) Trends Pharmacol. Sci. 10:270. See also Sawyer, T. K. (1995) Peptidomimetic Design and Chemical Approaches to Peptide Metabolism in Taylor, M. D. and Amidon, G. L. (eds.) Peptide-Based Drug Design: Controlling Transport and Metabolism, Chapter 17; Smith, A. B. 3rd, et al. (1995) J. Am. Chem. Soc. 117: 11113-11123; Smith, A. B. 3rd, et al. (1994) J. Am. Chem. Soc. 116:9947-9962; and Hirschman, R., et al. (1993) J. Am. Chem. Soc. 115: 12550-12568. A “derivative” (e.g., a peptide or amino acid) includes forms in which one or more reaction groups on the compound have been derivatized with a substituent group. Examples of peptide derivatives include peptides in which an amino acid side chain, the peptide backbone, or the amino- or carboxy-terminus has been derivatized (e.g., peptidic compounds with methylated amide linkages). An “analog” of a compound X includes compounds which retain chemical structures necessary for functional activity, yet which also contains certain chemical structures which differ. An example of an analog of a naturally-occurring peptide is a peptide which includes one or more non-naturally-occurring amino acids. A “mimetic” of a compound includes compounds in which chemical structures of the compound necessary for functional activity have been replaced with other chemical structures which mimic the conformation of the compound. Examples of peptidomimetics include peptidic compounds in which the peptide backbone is substituted with one or more benzodiazepine molecules (see e.g., James, G. L. et al. (1993) Science 260: 1937-1942) or oligomers that mimics peptide secondary structure through use of amide bond isosteres and/or modification of the native peptide backbone, including chain extension or heteroatom incorporation; examples of which include azapeptides, oligocarbamates, oligoureas, beta-peptides, gamma-peptides, oligo (phenylene ethynylene)s, vinylogous sulfonopeptides, poly-N-substituted glycines (peptoids) and the like. Methods for preparing peptidomimetic compounds are well known in the art and are specified, for example, in Quantitative Drug Design, C. A. Ramsden Gd., Chapter 17.2, F. Choplin Pergamon Press (1992). In addition to the above, the micropeptides of the present invention may also include one or more modified amino acids or one or more non-amino acid monomers (e.g. fatty acids, complex carbohydrates etc). The present teachings further contemplate cyclic peptides or cyclic structures within the peptides. Methods of cyclization are well known in the art, see for instance in WO2010/041237. The micropeptides of the present invention can be biochemically synthesized such as by using standard solid phase techniques. These methods include exclusive solid phase synthesis, partial solid phase synthesis methods, fragment condensation, classical solution synthesis. Solid phase polypeptide synthesis procedures are well known in the art and further described by John Morrow Stewart and Janis Dillaha Young, Solid Phase Polypeptide Syntheses (2nd Ed., Pierce Chemical Company, 1984). Synthetic peptides can be purified by preparative high performance liquid chromatography (Creighton T. (1983) Proteins, structures and molecular principles. WH Freeman and Co. NY) and the composition of which can be confirmed via amino acid sequencing. Recombinant techniques may also be used to generate the micropeptides of the present invention. To produce a peptide of the present invention using recombinant technology, a polynucleotide encoding the micropeptide of the present invention is ligated into a nucleic acid expression vector, which comprises the polynucleotide sequence under the transcriptional control of a cis-regulatory sequence (e.g., promoter sequence) suitable for directing constitutive, tissue specific or inducible transcription of the monomers of the present invention in the host cells. In addition to being synthesizable in host cells, the peptides of the present invention can also be synthesized using in vitro expression systems. These methods are well known in the art and the components of the system are commercially available. The activity or function of the sequences SEQ ID NO: 1, 2 or 3 and their functionally equivalent variants can be determined, by assaying the anti-proliferative activity of the peptides by a method shown in the examples of the present application. Functional equivalent variants of sequences SEQ ID NO: 1, 2 or 3 also include amino acid sequences with a sequence identity of at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% with the sequences SEQ ID NO: 1, 2 or 3. In a particular embodiment the functionally equivalent variant of the micropeptide of the invention has a sequence which shows at least 80% identity with the sequence of SEQ ID NO:1, 2 or 3. The terms “identity”, “identical” or “percent identity” in the context of two or more amino acid or nucleotide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences. One such non-limiting example of a sequence alignment algorithm is the algorithm described in Karlin et al., 1990, Proc. Natl. Acad. Sci., 87:2264-8, as modified in Karlin et al., 1993, Proc. Natl. Acad. Sci., 90:5873-7, and incorporated into the N BLAST and XBLAST programs (Altschul et al., 1997, Nucleic Acids Res., 25:3389-402). In certain embodiments, Gapped BLAST can be used as described in Altschul et al., 1997, Nucleic Acids Res. 25:3389-402. BLAST-2, WU-BLAST-2 (Altschul et al., 1996, Methods in Enzymology, 266:460-80), ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or Megalign (DNASTAR) are additional publicly available software programs that can be used to align sequences. In certain embodiments, the percent identity between two nucleotide sequences is determined using the GAP program in GCG software (e.g., using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 90 and a length weight of 1, 2, 3, 4, 5, or 6). In certain alternative embodiments, the GAP program in the GCG software package, which incorporates the algorithm of Needleman and Wunsch (J. Mol. Biol. 48:444-53 (1970)) can be used to determine the percent identity between two amino acid sequences (e.g., using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5). Alternatively, in certain embodiments, the percent identity between nucleotide or amino acid sequences is determined using the algorithm of Myers and Miller (CABIOS, 4:1 1-7 (1989)). For example, the percent identity can be determined using the ALIGN program (version 2.0) and using a PAM120 with residue table, a gap length penalty of 12 and a gap penalty of 4. Appropriate parameters for maximal alignment by particular alignment software can be determined by one skilled in the art. In certain embodiments, the default parameters of the alignment software are used. In certain embodiments, the percentage identity “X” of a first amino acid sequence to a second amino acid sequence is calculated as 100×(Y/Z), where Y is the number of amino acid residues scored as identical matches in the alignment of the first and second sequences (as aligned by visual inspection or a particular sequence alignment program) and Z is the total number of residues in the second sequence. If the second sequence is longer than the first sequence, then the global alignment taken the entirety of both sequences into consideration is used, therefore all letters and null in each sequence must be aligned. In this case, the same formula as above can be used but using as Z value the length of the region wherein the first and second sequence overlaps, said region having a length which is substantially the same as the length of the first sequence. As a non-limiting example, whether any particular polynucleotide or polypeptide has a certain percentage sequence identity (e.g., is at least 80% identical, at least 85% identical, at least 90% identical, and in some embodiments, at least 95%, 96%, 97%, 98%, or 99% identical) to a reference sequence can, in certain embodiments, be determined using the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 5371 1). Bestfit uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-9 (1981), to find the best segment of homology between two sequences. When using Bestfit or any other sequence alignment program to determine whether a particular sequence is, for instance, 95% identical to a reference sequence according to the present invention, the parameters are set such that the percentage of identity is calculated over the full length of the reference amino acid sequence and that gaps in homology of up to 5% of the total number of nucleotides in the reference sequence are allowed. In some embodiments, two amino acid sequences are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection. Identity can exist over a region of the sequences that is at least about 10, about 20, about 40-60 residues in length or any integral value therebetween, and can be over a longer region than 60-80 residues, for example, at least about 90-100 residues, and in some embodiments, the sequences are substantially identical over the full length of the sequences being compared. Antibodies Against the Micropeptides of the Invention In another aspect the invention relates to an antibody which is capable of specifically binding to the micropeptides of the invention. As used herein, the term “antibody” relates to a monomeric or multimeric protein which comprises at least one polypeptide having the capacity for binding to a determined antigen, or epitope within the antigen, and comprising all or part of the light or heavy The term antibody also includes any type of known antibody, such as, for example, polyclonal antibodies, monoclonal antibodies and genetically engineered antibodies, such as chimeric antibodies, humanized antibodies, primatized antibodies, human antibodies and bispecific antibodies (including diabodies), multispecific antibodies (e.g. bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity. The invention also comprises the use of fragments of the different types of antibodies mentioned above which substantially preserve the anti-angiogenic activity of the antibody. The term “antibody fragment” includes antibody fragments such as Fab, F(ab′)2, Fab′, single chain Fv fragments (scFv), diabodies and nanobodies. The phrase “specifically binds” when referring to antibodies and antigen binding fragments thereof means that the antibody binds to the micropeptides of the invention of variants thereof with no or insignificant binding to other human proteins. The term however does not exclude the fact that antibodies of the invention may also be cross-reactive with other forms of the micropeptides of the invention. Typically, the antibody binds with an association constant (Kd) of at least about 1×10−6M or 10−7M, or about 10−8M to 10−9M, or about 10−10M to 10−11M or higher, and binds to the predetermined antigen with an affinity that is at least two-fold greater than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen. The phrases “an antibody recognizing an antigen” and “an antibody specific for an antigen” are used interchangeably herein with the term “an antibody which binds specifically to an antigen”. The phrase “specifically bind(s)” or “bind(s) specifically” when referring to a peptide refers to a peptide molecule which has intermediate or high binding affinity, exclusively or predominately, to a target molecule. The phrase “specifically binds to” refers to a binding reaction that is determinative of the presence of a target protein in the presence of a heterogeneous population of proteins and other biologics. Thus, under designated assay conditions, the specified binding moieties bind preferentially to a particular target protein and do not bind in a significant amount to other components present in a test sample. Specific binding to a target protein under such conditions may require a binding moiety that is selected for its specificity for a particular target antigen. A variety of assay formats may be used to select ligands that are specifically reactive with a particular protein. For example, solid-phase ELISA immunoassays, immunoprecipitation, Biacore, and Western blot are used to identify peptides that specifically react with the micropeptides of the invention or their variants thereof. Typically a specific or selective reaction will be at least twice background signal or noise and more typically more than times background. Polynucleotides Encoding the Micropeptides of the Invention, and Vectors and Host Cells Containing Said Polynucleotides In a third aspect the invention relates to a polynucleotide encoding the micropeptide of the invention, with the proviso that said polynucleotide is not a polynucleotide consisting of the sequence having the NCBI accession number NM_203306.2 corresponding to the version of 10 Sep. 2017, NR_027064 corresponding to the version of 29 Aug. 2017 and NR_040248 corresponding to the version of 23 Apr. 2017. As used herein, the term “polynucleotide” refers to a polymer composed of a multiplicity of nucleotide units (deoxyribonucleotides or ribonucleotides, or related structural variants or synthetic analogues thereof) linked via phosphodiester bonds (or related structural variants on synthetic analogues thereof). The term polynucleotide includes double or single stranded genomic and cDNA, RNA, any synthetic and genetically manipulated polynucleotide, and both sense and anti-sense polynucleotide (although only sense stands are being disclosed in the present invention). This includes single- and double-stranded molecules, i.e., DNA-DNA, DNA-RNA and RNA-RNA hybrids. The polynucleotide of the invention can be found isolated as such or forming part of vectors allowing the propagation of said polynucleotides in suitable host cells. Therefore, in another aspect, the invention relates to a vector, hereinafter vector of the invention, encoding the polynucleotide of the invention as described above. Vectors suitable for the insertion of said polynucleotide are vectors derived from expression vectors in prokaryotes such as pUC18, pUC19, Bluescript and the derivatives thereof, mpl8, mp19, pBR322, pMB9, CoIEI, pCRI, RP4, phages and “shuttle” vectors such as pSA3 and pAT28; expression vectors in yeasts such as vectors of the type of 2 micron plasmids, integration plasmids, YEP vectors, centromere plasmids and the like; expression vectors in insect cells such as vectors of the pAC series and of the pVL; expression vectors in plants such as plBI, pEarleyGate, pAVA, pCAMBIA, pGSA, pGWB, pMDC, pMY, pORE series and the like; and expression vectors in eukaryotic cells, including baculovirus suitable for transfecting insect cells using any commercially available baculovirus system. The vectors for eukaryotic cells include preferably viral vectors (adenoviruses, viruses associated to adenoviruses such as retroviruses and, particularly, lentiviruses) as well as non-viral vectors such as pSilencer 4.1-CMV (Ambion), pcDNA3, pcDNA3.1/hyg, pHMCV/Zeo, pCR3.1, pEFI/His, pIND/GS, pRc/HCMV2, pSV40/Zeo2, pTRACER-HCMV, pUB6/V5-His, pVAXI, pZeoSV2, pCl, pSVL and PKSV-10, pBPV-1, pML2d and pTDTI. The vectors may also comprise a reporter or marker gene which allows identifying those cells that have incorporated the vector after having been put in contact with it. Useful reporter genes in the context of the present invention include lacZ, luciferase, thymidine kinase, GFP and on the like. Useful marker genes in the context of this invention include, for example, the neomycin resistance gene, conferring resistance to the aminoglycoside G418; the hygromycin phosphotransferase gene, conferring resistance to hygromycin; the ODC gene, conferring resistance to the inhibitor of the ornithine decarboxylase (2-(difluoromethyl)-DL-ornithine (DFMO); the dihydrofolatereductase gene, conferring resistance to methotrexate; the puromycin-N-acetyl transferase gene, conferring resistance to puromycin; the ble gene, conferring resistance to zeocin; the adenosine deaminase gene, conferring resistance to 9-beta-D-xylofuranose adenine; the cytosine deaminase gene, allowing the cells to grow in the presence of N-(phosphonacetyl)-L-aspartate; thymidine kinase, allowing the cells to grow in the presence of aminopterin; the xanthine-guanine phosphoribosyltransferase gene, allowing the cells to grow in the presence of xanthine and the absence of guanine; the trpB gene ofE. coli, allowing the cells to grow in the presence of indol instead of tryptophan; the hisD gene ofE. coli, allowing the cells to use histidinol instead of histidine. The selection gene is incorporated into a plasmid that can additionally include a promoter suitable for the expression of said gene in eukaryotic cells (for example, the CMV or SV40 promoters), an optimized translation initiation site (for example, a site following the so-called Kozak's rules or an IRES), a polyadenylation site such as, for example, the SV40 polyadenylation or phosphoglycerate kinase site, introns such as, for example, the beta-globulin gene intron. Alternatively, it is possible to use a combination of both the reporter gene and the marker gene simultaneously in the same vector. On the other hand, as the skilled person in the art knows, the choice of the vector will depend on the host cell in which it will subsequently be introduced. By way of example, the vector in which said polynucleotide is introduced can also be a yeast artificial chromosome (YAC), a bacterial artificial chromosome (BAC) or a PI-derived artificial chromosome (PAC). The characteristics of the YAC, BAC and PAC are known by the person skilled in the art. Detailed information on said types of vectors has been provided, for example, by Giraldo and Montoliu (Giraldo, P. & Montoliu L., 2001 Size matters: use of YACs, BACs and PACs in transgenic animals, Transgenic Research 10(2): 83-110). The vector of the invention can be obtained by conventional methods known by persons skilled in the art (Sambrook J. et al., 2000 “Molecular cloning, a Laboratory Manual”, 3rd ed., Cold Spring Harbor Laboratory Press, N.Y. Vol 1-3). The polynucleotide of the invention can be introduced into the host cell in vivo as naked DNA plasmids, but also using vectors by methods known in the art, including but not limited to transfection, electroporation (e.g. transcutaneous electroporation), microinjection, transduction, cell fusion, DEAE dextran, calcium phosphate precipitation, use of a gene gun, or use of a DNA vector transporter. See J M Wilson, et al., J. Biol. Chem. 1992; 267:963-967, Wu C and Wu G, Biol. Chem. 1988; 263: 14621-14624, and Williams R, et al., Proc. Natl. Acad. Sci. USA 1991; 88:2726-2730. Methods for formulating and administering naked DNA to mammalian muscle tissue are also known. See Feigner P, et al., U.S. Pat. Nos. 5,580,859, and 5,589,466. Other molecules are also useful for facilitating transfection of a nucleic acid in vivo, such as cationic oligopeptides, peptides derived from DNA binding proteins, or cationic polymers. See Bazile D, et al., WO 1995021931, and BykG, et al., WO 1996025508. Another well-known method that can be used to introduce polynucleotides into host cells is particle bombardment (aka biolistic transformation). Biolistic transformation is commonly accomplished in one of several ways. One common method involves propelling inert or biologically active particles at cells. See Sanford J, et al., U.S. Pat. Nos. 4,945,050, 5,036,006, and 5,100,792. Alternatively, the vector can be introduced in vivo by lipofection. The use of cationic lipids can promote encapsulation of negatively charged nucleic acids, and also promote fusion with negatively charged cell membranes. See Feigner P, Ringold G, Nature, 1989; 337:387-388. Particularly useful lipid compounds and compositions for transfer of nucleic acids have been described. See Feigner P, et al., U.S. Pat. No. 5,459,127, Behr J, et al., WO1995018863, and Byk G, WO1996017823. Thus, in another aspect, the invention relates to a host cell, hereinafter cell of the invention, comprising the polynucleotide of the invention or a vector of the invention. The cells can be obtained by conventional methods known by persons skilled in the art (see e.g. Sambrook et al., cited ad supra). The term “host cell”, as used herein, refers to a cell into which a nucleic acid of the invention, such as a polynucleotide or a vector according to the invention, has been introduced and is capable of expressing the micropeptides of the invention. The terms “host cell” and “recombinant host cell” are used interchangeably herein. It should be understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein. The term includes any cultivatable cell that can be modified by the introduction of heterologous DNA. Preferably, a host cell is one in which the polynucleotide of the invention can be stably expressed, post-translationally modified, localized to the appropriate subcellular compartment, and made to engage the appropriate transcription machinery. The choice of an appropriate host cell will also be influenced by the choice of detection signal. For example, reporter constructs, as described above, can provide a selectable or screenable trait upon activation or inhibition of gene transcription in response to a transcriptional regulatory protein; in order to achieve optimal selection or screening, the host cell phenotype will be considered. A host cell of the present invention includes prokaryotic cells and eukaryotic cells. Prokaryotes include gram negative or gram positive organisms, for example,E. colior Bacilli. It is to be understood that prokaryotic cells will be used, preferably, for the propagation of the transcription control sequence comprising polynucleotides or the vector of the present invention. Suitable prokaryotic host cells for transformation include, for example,E. coli, Bacillus subtilis, Salmonella typhimurium, and various other species within the generaPseudomonas, Streptomyces, andStaphylococcus. Eukaryotic cells include, but are not limited to, yeast cells, plant cells, fungal cells, insect cells (e.g., baculovirus), mammalian cells, and the cells of parasitic organisms, e.g., trypanosomes. As used herein, yeast includes not only yeast in a strict taxonomic sense, i.e., unicellular organisms, but also yeast-like multicellular fungi of filamentous fungi. Exemplary species includeKluyverei lactis, Schizosaccharomyces pombe, andUstilaqo maydis, withSaccharomyces cerevisiaebeing preferred. Other yeasts which can be used in practicing the present invention areNeurospora crassa, Aspergillus niger, Aspergillus nidulans, Pichia pastoris, Candida tropicalis, andHansenula polymorpha. Mammalian host cell culture systems include established cell lines such as COS cells, L cells, 3T3 cells, Chinese hamster ovary (CHO) cells, embryonic stem cells, with BHK, HeK or HeLa cells being preferred. Eukaryotic cells are, preferably, used to for recombinant gene expression by applying the transcription control sequence or the expression vector of the present invention. Compositions and Pharmaceutical Compositions Comprising the Micropeptides, Polynucleotides, Vectors and Host Cells of the Invention In a further aspect the invention relates to a composition comprising the micropeptide according of the invention, or the polynucleotide, the expression vector or the host cell of the invention. The term “composition”, as used herein, relates to a material composition that comprises the above-mentioned components, as well as any product resulting, directly or indirectly, from the combination of the different components in any quantity thereof. Those skilled in the art will observe that the composition may be formulated as a single formulation or may be presented as separate formulations of each of the components, which may be combined for joint use as a combined preparation. The composition may be a kit-of-parts wherein each of the components is individually formulated and packaged. In another aspect the invention also relates to a pharmaceutical composition comprising the composition of the invention and a pharmaceutically acceptable carrier. The term “pharmaceutical composition”, as used herein, refers to a composition comprising a therapeutically effective amount of the agents according to the present invention and at least one pharmaceutically acceptable excipient. Pharmaceutical compositions according to the invention can be prepared, for instance, as injectables such as liquid solutions, suspensions, and emulsions. The term “therapeutically effective amount”, as used herein in relation to the micropeptides, the polynucleotides, the vectors or the cells of the invention comprised by the pharmaceutical composition of the invention, relates to the sufficient amount of the micropeptides, the polynucleotides, the vectors or the cells according to the present invention to provide the desired effect, i.e. to achieve an appreciable prevention, cure, delay, reduction of severity or amelioration of one or more symptoms derived from a disease, and will generally be determined by, among other causes, the characteristics of the agent itself and the therapeutic effect to be achieved. It will also depend on the subject to be treated, the severity of the disease suffered by said subject, the chosen dosage form, etc. For this reason, the doses mentioned in this invention must be considered only as guides for the person skilled in the art, who must adjust the doses depending on the aforementioned variables. In an embodiment, the effective amount produces the amelioration of one or more symptoms of the disease that is being treated. Even though individual needs vary, determination of optimal ranges for effective amounts of the compound of the invention belongs to the common experience of those experts in the art. In general, the dosage needed to provide an effective amount of such compound, which can be adjusted by one expert in the art will vary depending on age, health, fitness, sex, diet, weight, degree of alteration of the receptor, frequency of treatment and the nature and extent of impairment or illness, medical condition of the patient, route of administration, pharmacological considerations such as activity, efficacy, pharmacokinetic and toxicology profile of the particular compound used, if using a system drug delivery, and if the compound is administered as part of a combination of drugs. Those skilled in the art are familiar with the principles and procedures discussed in widely known and available sources as Remington's Pharmaceutical Science (17th Ed., Mack Publishing Co., Easton, Pa., 1985) and Goodman and Gilman's The terms “pharmaceutically acceptable excipient” or “pharmaceutically acceptable carrier”, refer to any compound or combination of compounds that is essentially non-toxic to the subject at the dosage and concentration employed, and is compatible with the other components of a pharmaceutical composition. Thus, an excipient is an inactive substance formulated alongside the active ingredient of a pharmaceutical composition, for the purpose of bulking-up compositions that contain said active ingredients. Bulking up allows convenient and accurate dispensation of a drug substance when producing a dosage form. Excipients also can serve various therapeutic-enhancing purposes, such as facilitating compound (drug) absorption or solubility, or other pharmacokinetic considerations. Excipients can also be useful in the manufacturing process, to aid in the handling of the active substance concerned such as by facilitating powder flowability or non-stick properties, in addition to aiding in vitro stability such as prevention of denaturation over the expected shelf life. The selection of appropriate excipients depends upon the route of administration and the dosage form, as well as the active ingredient and other factors. An excipient can be a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any conventional type. Illustrative, non-limitative, examples of excipients or carriers include water, salt (saline) solutions, alcohol, dextrose, vegetable oils, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, surfactants, silicic acid, viscous paraffin, perfume oil, monoglycerides and diglycerides of fatty acids, fatty acid esters petroetrals, hydroxymethyl cellulose, polyvinylpyrrolidone and the like. In a particular embodiment, the pharmaceutical composition containing the compound for use in the present invention is a pharmaceutical composition for parenteral administration. Thus, said pharmaceutical composition suitable for parenteral injection, include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or may comprise sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous or non-aqueous excipients or carriers, diluents, solvents or vehicles include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, and the like), suitable mixtures thereof, triglycerides, including vegetable oils such as olive oil, or injectable organic esters such as ethyl oleate. In a particular embodiment, the pharmaceutical composition containing the compound for use in the present invention is a pharmaceutical composition for intravenous, intramuscular or subcutaneous administration. Typically, pharmaceutical compositions for intravenous, intramuscular or subcutaneous administration are solutions in sterile isotonic aqueous buffer. If necessary, the pharmaceutical composition including the compound for use according to the invention also includes a local anesthetic to ameliorate any pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampule or sachette indicating the quantity of active ingredient. Where the pharmaceutical composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the pharmaceutical composition is administered by injection, an ampule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration. In another particular embodiment, the pharmaceutical composition containing the compound for use in the present invention is a pharmaceutical composition for oral administration. Solid dosage forms for oral administration include conventional capsules, sustained release capsules, conventional tablets, sustained-release tablets, chewable tablets, sublingual tablets, effervescent tablets, pills, suspensions, powders, granules and gels. In the solid dosage forms, the active ingredient (i.e., the compound selected from the group consisting of a) a peptide of SEQ ID NO:1, 2 or 3; b) a functionally equivalent variant of the peptide according to a); c) a polynucleotide encoding a) or b); d) a vector comprising a polynucleotide according to c); e) a cell capable of secreting into the medium a peptide according to a) or b); and f) a nanoparticle comprising the peptide according to a) or b)) is admixed with at least one suitable excipient or carrier, such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, such as for example, starches, lactose, sucrose, mannitol, or silicic acid; (b) binders, such as for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, or acacia; (c) humectants, such as for example, glycerol; (d) disintegrating agents, such as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, or sodium carbonate; (e) solution retarding agents, such as for example, paraffin; (f) absorption accelerators, such as for example, quaternary ammonium compounds; (g) wetting agents, such as for example, cetyl alcohol or glycerol monostearate; (h) adsorbents, such as for example, kaolin or bentonite; and/or (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules and tablets, the dosage forms may also comprise buffering agents. Solid formulations of a similar type may also be used as fillers in soft or hard filled gelatin capsules using excipients such as lactose or milk sugar, as well as high molecular weight polyethylene glycols, and the like. Solid dosage forms such as coated tablets, capsules and granules can be prepared with coatings or shells, such as enteric coatings and others known in the art. They may also contain opacifying agents, and can be formulated such that they release the active ingredient or ingredients in a delayed manner. Examples of embedding formulations that can be used are polymeric substances and waxes. The active ingredients can also be in micro-encapsulated form, if appropriate, with one or more of the aforementioned excipients. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs containing suitable excipients or carriers used in the art. In addition to the active ingredient (i.e., the compound selected from the group consisting of a) a peptide of SEQ ID NO:1, 2 or 3; b) a functionally equivalent variant of the peptide according to a); c) a polynucleotide encoding a) or b); d) a vector comprising a polynucleotide according to c); e) a cell capable of secreting into the medium a peptide according to a) or b) and f) a nanoparticle comprising the peptide according to a) or b)) the liquid dosage form may contain one or more excipients or carriers commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, such as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils, particular cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, sesame seed oil, Miglyol®, glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols, fatty acid esters of sorbitan, or mixtures of these substances, and the like. In addition to said inert diluents, the formulation can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfuming agents. Suspensions, in addition to the active ingredient or ingredients, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol or sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, or tragacanth, or mixtures of these substances, and the like. In another particular embodiment, the pharmaceutical composition containing the compound for use in the present invention is a pharmaceutical composition for topical administration. For topical administration, said pharmaceutical composition can be formulated as a cream, gel, lotion, liquid, pomade, spray solution, dispersion, solid bar, emulsion, microemulsion and the like which may be formulated according to conventional methods that use suitable excipients, such as, for example, emulsifiers, surfactants, thickening agents, coloring agents and combinations of two or more thereof. The pharmaceutical composition comprising a compound for use according to the invention may be also administered in the form of transdermal patches or iontophoresis devices. Thus, in a specific embodiment, a compound for use according to the invention is administered as a transdermal patch, for example, in the form of sustained-release transdermal patch. Suitable transdermal patches are described in more detail in, for example, U.S. Pat. Nos. 5,262,165, 5,948,433, 6,010,715 and 6,071,531. Several drug delivery systems are known and can be used to administer the compounds for use according to the invention, including, for example, encapsulation in liposomes, microbubbles, emulsions, microparticles, microcapsules or by means of other nanotechnology systems such as for example polymer therapeutics and the like. The required dosage can be administered as a single unit or in a sustained release form. Sustainable-release forms and appropriate materials and methods for their preparation are described in the art. In a particular embodiment, the orally administrable form of a pharmaceutical composition comprising a compound for use according to the invention is in a sustained release form that further comprises at least one coating or matrix. The coating or sustained release matrix include, without limitation, natural polymers, semisynthetic or synthetic water-insoluble, modified, waxes, fats, fatty alcohols, fatty acids, natural semisynthetic or synthetic plasticizers, or a combination of two or more of them. Enteric coatings may be applied using conventional processes known to experts in the art. In a particular embodiment, when the compound for use according to the invention comprises a nucleic acid, the pharmaceutical composition may be formulated as a composition intended for use in gene therapy; by way of illustration, not limitation, that pharmaceutical composition may contain a viral or non-viral vector, which comprises the suitable polynucleotide or gene construction. By way of illustration and not limitation, said vectors, may be viral, for example, based on retrovirus, adenovirus, etc., or nonviral such as ADN-liposome, ADN-polymer, ADN-polymer-liposome complexes, etc. (see “Nonviral Vectors for Gene Therapy”, edited by Huang, Hung and Wagner, Academic Press (1999)). Said vectors, which contain the corresponding polynucleotide or gene construction, may be administered directly to a subject by conventional methods. Alternatively, said vectors may be used to transform, or transfect or infect cells, for example, mammal cells, including human, ex vivo, which subsequently will be implanted into a human body or an animal to obtain the desired therapeutic effect. For administration to a human body or an animal, said cells will be formulated in a suitable medium that will have no adverse influence on cell viability. In a further aspect the invention relates to the micropeptide, the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention for use in medicine. Therapeutic Uses in Cancer of the Micropeptides, Polynucleotides and Vectors of the Invention As it is shown in the examples of the present application, the authors of the invention have found out that the micropeptides of the invention are able to act as tumor suppressor agents, being therefore interesting tools for treatment of cancer. Thus, in another aspect, the invention relates to the micropeptide, the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention for use in the treatment of cancer. The term “treatment”, as used herein, comprises any type of therapy, which aims at terminating, preventing, ameliorating and/or reducing the susceptibility to a clinical condition as described herein, e.g. cancer. Thus, “treatment,” “treating,” and the like, as used herein, refer to obtaining a desired pharmacologic and/or physiologic effect, covering any treatment of a pathological condition or disorder in a mammal, including a human. The effect may be prophylactic in terms of completely or partially preventing a disorder or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disorder and/or adverse effect attributable to the disorder. That is, “treatment” includes (1) preventing the disorder from occurring or recurring in a subject, (2) inhibiting the disorder, such as arresting its development, (3) stopping or terminating the disorder or at least symptoms associated therewith, so that the host no longer suffers from the disorder or its symptoms, such as causing regression of the disorder or its symptoms, for example, by restoring or repairing a lost, missing or defective function, or stimulating an inefficient process, or (4) relieving, alleviating, or ameliorating the disorder, or symptoms associated therewith, where ameliorating is used in a broad sense to refer to at least a reduction in the magnitude of a parameter. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented. The term “cancer” or “tumour” or “tumour disease”, as used herein, refers to a broad group of diseases involving unregulated cell growth and which are also referred to as malignant neoplasms. The term is usually applied to a disease characterized by uncontrolled cell division (or by an increase of survival or apoptosis resistance) and by the ability of said cells to invade other neighboring tissues (invasion) and spread to other areas of the body where the cells are not normally located (metastasis) through the lymphatic and blood vessels, circulate through the bloodstream, and then invade normal tissues elsewhere in the body. Depending on whether or not they can spread by invasion and metastasis, tumours are classified as being either benign or malignant: benign tumours are tumours that cannot spread by invasion or metastasis, i.e., they only grow locally; whereas malignant tumours are tumours that are capable of spreading by invasion and metastasis. Biological processes known to be related to cancer include angiogenesis, immune cell infiltration, cell migration and metastasis. Cancers usually share some of the following characteristics: sustaining proliferative signalling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and eventually metastasis. Cancers invade nearby parts of the body and may also spread to more distant parts of the body through the lymphatic system or bloodstream. Cancers are classified by the type of cell that the tumour cells resemble, which is therefore presumed to be the origin of the tumour. Examples of cancer or tumor include without limitation, breast, heart, lung, small intestine, colon, spleen, kidney, bladder, head, neck, ovarian, prostate, brain, rectum, pancreas, skin, bone, bone marrow, blood, thymus, uterus, testicles, hepatobiliary and liver tumors. In particular, the tumor/cancer can be selected from the group of adenoma, angiosarcoma, astrocytoma, epithelial carcinoma, germinoma, glioblastoma, glioma, hemangioendothelioma, hepatoblastoma, leukaemia, lymphoma, medulloblastoma, melanoma, neuroblastoma, hepatobiliary cancer, osteosarcoma, retinoblastoma, rhabdomyosarcoma, sarcoma, teratoma, acrallentiginous melanoma, actinic keratosis adenocarcinoma, adenoid cystic carcinoma, adenosarcoma, adenosquamous carcinoma, astrocytictumors, bartholin gland carcinoma, basal cell carcinoma, bronchial gland carcinoma, carcinosarcoma, cholangiocarcinoma, cystadenoma, endodermal sinus tumor, endometrial hyperplasia, endometrial stromal sarcoma, endometrioid adenocarcinoma, ependymal sarcoma, Swing's sarcoma, focal nodular hyperplasia, germ cell tumors, glucagonoma, hemangioblastoma, hemangioma, hepatic adenoma, hepatic adenomatosis, hepatocellular carcinoma, insulinoma, intraepithelial neoplasia, interepithelial squamous cell neoplasia, invasive squamous cell carcinoma, large cell carcinoma, leiomyosarcoma, malignant melanoma, malignant mesothelialtumor, medulloepithelioma, mucoepidermoid carcinoma, neuroepithelial adenocarcinoma, nodular melanoma, papillary serous adenocarcinoma, pituitary tumors, plasmacytoma, pseudosarcoma, pulmonary blastoma, renal cell carcinoma, serous carcinoma, small cell carcinoma, soft tissue carcinoma, somatostatin-secreting tumor, squamous carcinoma, squamous cell carcinoma, undifferentiated carcinoma, uveal melanoma, verrucous carcinoma, vipoma, Wilm's tumor. In a particular embodiment the invention relates to the micropeptide, polynucleotide, expression vector, host cell, composition or pharmaceutical composition of the invention for use in the treatment of cancer, wherein the cancer is a primary tumor or cancer metastasis. The term “primary tumor”, as used herein, refers to a tumor that originated in the location or organ in which it is present and did not metastasize to that location from another location In the context of the present invention, “metastasis” is understood as the propagation of a cancer from the organ where it started to a different organ. It generally occurs through the blood or lymphatic system. When the cancer cells spread and form a new tumor, the latter is called a secondary or metastatic tumor. The cancer cells forming the secondary tumor are like those of the original tumor. If a breast cancer, for example, spreads (metastasizes) to the lung, the secondary tumor is formed of malignant breast cancer cells. The disease in the lung is metastatic breast cancer and not lung cancer. In another embodiment, the micropeptide, polynucleotide, expression vector, host cell, composition or pharmaceutical composition, wherein the micropeptide is the micropeptide of sequence SEQ ID NO: 1 or SEQ ID NO: 3, are used for the treatment of a cancer characterized in that it comprises an inactivating mutation in the p53 gene. Thus, in a particular embodiment, the cancer cells carry an inactivating mutation in at least one allele of the p53 gene. The term “inactivating mutation”, as used herein, refers to mutations that partially or completely abrogate the activity of the polypeptide encoded by the mutated polynucleotide. In the particular case of p53, inactivating mutations are those which result in a partial or total deficiency in its ability to initiate a DNA-repair response. Suitable mutations leading to inactivation of p53 are usually missense mutations such as those known in the art (Michalovitz et al., J. Cell. Biochem., 1991, 45(1):22-9; Vogelstein and Kinzler, Cell, 1992, 70(4):523-6; Donehower and Bradley, Biochim. Biophys. Acta., 1993, 1155(2): 181-205; Levine, Cell, 1997, 88(3):323-31). These mutations affect almost exclusively the core DNA-binding domain of p53 that is responsible for making contacts with p53 DNA-binding sites, although some inactivating mutations have been described in the N-terminal transactivation domain or the C-terminal tetramerization domain (Beroud and Soussi, Nucleic Acids Res., 1998, 26(1):200-4; Cariello et al., Nucleic Acids Res., 1998, 26(1): 198-9; Hainaut et al., P., Nucleic Acids Res. 1998; 26:205-213). In another particular embodiment, the transgenic non-human animal of the invention is homozygous for a totally defective p53 gene (p53−/−). In a particular embodiment the micropeptide, the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention are used in the treatment of a carcinoma cancer. As used herein, the term “carcinoma” refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas. In another particular embodiment the micropeptide, the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention are used in the treatment or carcinoma selected from squamous cell carcinoma, adenocarcinoma, transitional cell carcinoma or basal cell carcinoma. In a further particular embodiment the micropeptide, the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention are used in the treatment of carcinoma selected from lung carcinoma, breast carcinoma, bladder carcinoma, prostate carcinoma, colon and rectum carcinoma, skin carcinoma, pancreas carcinoma, ovarian carcinoma, cervix carcinoma, hepatocellular carcinoma or renal cell carcinoma. Types of carcinoma include without limitation include: Acinic cell carcinoma, Actinic keratosis adenocarcinoma, Adenocarcinoma, Adenoid cystic carcinoma, Adenosquamous carcinoma, Adrenocortical carcinoma, Anaplastic carcinoma, Anaplastic carcinoma of pancreas, Anaplastic carcinoma of thyroid, Bartholin gland carcinoma, Basal cell carcinoma, Basaloid carcinoma, Bronchial gland carcinoma, Carcinoma in situ, CASTLE, Chromophobe cell carcinoma, Clear cell carcinoma, Collecting duct carcinoma, Colloid carcinoma, Ductal carcinoma in situ, Duct cell carcinoma, Embryonal carcinoma, Endometrial carcinoma, Epithelial carcinoma, Epithelial-myoepithelial carcinoma, Fibrolamellar carcinoma, Follicular carcinoma, Giant cell carcinoma, Glassy cell carcinoma, Hepatocellular carcinoma, Hürthle cell carcinoma, Inflammatory carcinoma, In situ carcinoma, Intraductal carcinoma, Intramucosal carcinoma, Juvenile carcinoma, Krebs' carcinoma, Large cell undifferentiated carcinoma of lung, Laryngeal carcinoma, Lobular carcinoma in situ, Medullary carcinoma, Merkel cell carcinoma, Microinvasive carcinoma, Minimal deviation adenocarcinoma of cervix, Mucoepidermoid carcinoma, ‘Murky cell’ carcinoma, Nasopharyngeal carcinoma, Neuroepithelial adenocarcinoma, Non-small cell carcinoma of lung, Oat cell carcinoma, Ovarian small cell carcinoma-hypercalcemic type, Pleomorphic carcinoma, Pleomorphic lobular carcinoma, Renal cell carcinoma, Sarcomatoid carcinoma, Scirrhous carcinoma, Secretory carcinoma, Serous carcinoma, Soft tissue carcinoma, Spindle cell carcinoma, Squamous carcinoma, Squamous cell carcinoma, Stump carcinoma, Superficial spreading carcinoma, Terminal duct carcinoma, Transglottic carcinoma, Transitional cell carcinoma, Tubular carcinoma and Undifferentiated carcinoma. In a particular embodiment, the micropeptide of SEQ ID NO: 1, or the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention encoding or comprising the micropeptide of SEQ ID NO: 1 or comprising the polynucleotide encoding the micropeptide of SEQ ID NO: 1, are used in the treatment of pancreatic, breast, lung, colon or skin cancer. More particular, in the treatment of pancreatic, breast, lung or colon carcinoma, or squamous cell carcinoma. In another embodiment, the micropeptide of SEQ ID NO: 2 or the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention encoding or comprising the micropeptide of SEQ ID NO: 2 or comprising the polynucleotide encoding the micropeptide of SEQ ID NO: 2, are used in the treatment of pancreatic and breast cancer. More particular, the cancers are triple negative breast cancer and pancreatic adenocarcinoma. In another particular embodiment, the micropeptide of SEQ ID NO: 2 or the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention encoding or comprising the micropeptide of SEQ ID NO: 2 or comprising the polynucleotide encoding the micropeptide of SEQ ID NO: 2 targets both tumour and stromal compartment (cancer associated fibroblasts) in the tumour In another particular embodiment, the micropeptide of SEQ ID NO: 3 or the polynucleotide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention encoding or comprising the micropeptide of SEQ ID NO: 3 or comprising the polynucleotide encoding the micropeptide of SEQ ID NO: 3, are used in the treatment of colon and lung cancer. More particular, in the treatment of colon and lung carcinoma. Uses in the Treatment of Fibrosis As shown in the examples of the application, it is proved that micropeptide of sequence SEQ ID NO: 2 induces downregulation of adhesion factors, such as COLLAGEN I, COLLAGEN IV, FIBRONECTIN and CD44. The upregulation of these factors is related to the development of fibrosis, a pathological feature associated with chronic inflammatory diseases, defined by the overgrowth, hardening, and/or scarring of tissues attributed to excess deposition of extracellular matrix components, such as collagen (Wynn T A et al., 2012, Nat. Med. 18:1028-40). In addition CD44, the receptor of hyaluronic acid and other ligands related with cell adhesion such as collagen and metalloproteinases, has been shown to be required for extracellular matrix adhesion and fibrosis (Li Y, et al, 2011; J Exp Med. 208, 1459-71 and Yang L W et al, 2018, doi: 10.1097/SHK.0000000000001132). Thus, in another aspect, the invention relates to the micropeptide of SEQ ID NO:2, the polynucleotide encoding said micropeptide, the expression vector, the host cell, the composition or the pharmaceutical composition of the invention for use in the treatment of fibrosis. As used herein the term “fibrosis” refers to a condition characterized by an excess deposition of fibrous tissue, and it is an underlying manifestation of many disease states. Fibrosis is similar to the process of scarring, in that both involve stimulated cells laying down connective tissue, including collagen and glycosaminoglycans. This can be a reactive, benign, or pathological state. The deposition of connective tissue in the organ and/or tissue can obliterate the architecture and function of the underlying organ or tissue. Fibrosis can occur in a variety of tissues or organs. These fibrotic conditions include dermal fibrosis (e.g., associated with scleroderma). Dermal fibrosis is fibrosis that manifests itself in the skin (or dermis). Fibrotic conditions also include hypertrophic scars, keloids, burns, Peyronie's disease, and Dupuytren's contractures. Fibrotic conditions also include non-dermal fibrosis. Non-dermal fibrosis is fibrosis that manifests itself in an organ other than the skin (or dermis). As important example of non-dermal fibrosis is lung (or pulmonary) fibrosis. Lung fibrosis can be associated with interstitial lung disease and diffuse proliferative lung disease. An example of lung fibrosis is idiopathic pulmonary fibrosis (JPF), including IPF with severe airway restriction (referred to herein as severe IPF). Other examples of non-dermal fibrosis include liver/hepatic fibrosis, ocular fibrosis, fibrosis of the gut, kidney/renal fibrosis, pancreatic fibrosis, vascular fibrosis, cardiac fibrosis, myelofibrosis, and the like. Some forms of fibrosis are referred to as interstitial fibrosis, and they include dermal or non-dermal interstitial fibrosis. Fibrosis may be further categorized by its etiology, to the extent such etiology is known. For example, the fibrosis may be associated with or resulting from an infection (e.g., a viral infection or a parasitic infection), or it may be drug-induced fibrosis (e.g., chemotherapy), or it may be associated with or resulting from substance abuse (e.g., alcohol-induced fibrosis), or it may be associated with or resulting from surgery or other invasive procedure, or it may be associated with or resulting from an underlying condition or event (e.g., a myocardial infarction or diabetes), or it may be associated with or resulting from radiation exposure (e.g., radiation treatment for cancer). Examples include liver/hepatic fibrosis associated with alcohol consumption, viral hepatitis and/or schistosomiasis; post-myocardial infarction cardiac fibrosis; kidney/renal fibrosis associated with diabetes; and post-inflammatory kidney/renal fibrosis. Fibrosis may be transplant-induced, or it may occur independently of transplant (i.e., in a subject that has not undergone a transplant and who is not in need of a transplant) The term “collagen I”, “type I collagen”, “alpha 1” or “alpha-1 type I collagen” are used indistinctly in the present application. They refer to a triple helix protein comprising two alpha1 chains and one alpha2 chain (coded by COL1A1 and COL1A2 genes respectively). The term CD44 as used herein makes reference to a cell-surface glycoprotein involved in cell-cell interactions, cell adhesion and migration. In humans, the CD44 antigen is encoded by the CD44 gene on Chromosome 11. CD44 is also referred to as HCAM (homing cell adhesion molecule), Pgp-1 (phagocytic glycoprotein-1), Hermes antigen, lymphocyte homing receptor, ECM-Ill, and HUTCH-1. In a particular embodiment the invention relates to the micropeptide, polynucleotide, expression vector, host cell, composition or pharmaceutical composition of the invention for use in the treatment of fibrosis, wherein the fibrosis is associated with loss of function in an organ or tissue, and surgical and/or esthetic complications In another particular embodiment the invention relates to the micropeptide, polynucleotide, expression vector, host cell, composition or pharmaceutical composition of the invention for use in the treatment of fibrosis, wherein the fibrosis is selected from among pulmonary fibrosis, hepatic fibrosis (cirrhosis), renal fibrosis, corneal fibrosis, fibrosis associated with skin and peritoneal surgery, fibrosis associated with burns, osteoarticular fibrosis or keloids. The invention will be described by way of the following examples which are to be considered as merely illustrative and not limitative of the scope of the invention. EXAMPLES Methodology Chemical Treatments Doxorubicin, Actinomycin D and Nutlin-3a were purchased from Sigma-Aldrich. Doxorubicin was used at 1 μM, Actinomycin D was used at 5 nM and Nutlin-3a was used at 10 μM in drug treatment experiments. Cloning Procedures SEQ ID NO: 1, SEQ ID NO: 2 and SEQ ID NO:3 coding sequence (ORFs) were synthesized (IDT technologies) fused with a flexible linker of amino acidic sequence GGGGSGGGGSGGGGS and a HA-Tag at the C-terminal, and flanked by EcoRI and XhoI enzyme restrictions sites. After digestion by EcoRI and XhoI, the constructs were ligated into pMSCV-Puro retroviral vector. SEQ ID NO: 1 was cloned in the pINDUCER20 vector (Invitrogen) using the Gateway@ Cloning Technology following manufacturer's instructions. Cell Culture BxPC-3 and A549 cell lines were cultured in RPMI supplemented with 10% of FBS and penicillin-streptomycin. Patient derived-H10 skin squamous cell carcinoma cell line was cultured in DMEM-F12 supplemented with B27 and penicillin-streptomycin. Cancer associated fibroblasts (CAFs) and PDAC cell lines were derived from LSL−KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cremice and cultured in DMEM-F12 supplemented with 10% FBS and penicillin-streptomycin. All the other cell lines were cultured in DMEM supplemented with 10% of FBS and penicillin streptomycin. Cultures were routinely tested formycoplasmaand were always negative. Retroviral and Lentiviral Transduction 5×106HEK293T cells were transfected with retroviral or lentiviral vectors and the packaging vectors (pCL-Ampho for retroviral transduction, pLP1 pLP2 and pLP-VSVG for lentiviral transduction) using Fugene HD (Roche). Viral supernatants were collected twice a day on two consecutive days starting 36 h after transfection and were used to infect IMR90 as well as cancer cell lines, previously plated at a density of 8×105cells per 10 cm plates. Before infection, polybrene was added to the viral supernatants at a concentration of 8 mg/ml. Analysis of mRNA Levels Total RNA was extracted with Trizol® and treated with TURBO™ DNase following provider's recommendations. 1 μg of RNA was retrotranscribed using iScript™ cDNA Synthesis Kit (BioRad). Gene expression was analysed by quantitative real time PCR using PowerUp SYBR Green Master Mix (Thermo Fisher Scientific) in the 7900HT Fast Real-Time PCR System (Applied Biosystems). Cycle threshold (Ct) values were normalized to GAPDH. Western Blot Cells pellets (4-5*106cells) were homogenized in medium-salt lysis buffer (150 mM NaCl, 50 mM Tris pH 8, 1% NP40 and protein inhibitors cocktail). 50 μg of protein was loaded per lane in a 12% acrylamide gel and electrophoresed in Tris-Glycine SDS Running Buffer. The following antibodies were used: Anti-HA-Tag (Abcam, 1:5000), Anti-p53 (Santa Cruz, 1:1000), Anti-p21 (Thermo Fisher, 1:1000), Anti-GAPDH (Thermo Fisher, 1:5000). Immunofluorescence Cells were seeded in glass coverslips coated with fibronectin at a cell density of 5×104cells/ml. Then, cells were fixed in 4% paraformaldehyde 15 min, washed twice with PBS and then permeabilized with 0.2% (v/v) Triton X-100 for 15 min at room temperature. Blocking step was made in 3% BSA for 1 hour. Cells were incubated in the same blocking buffer containing primary antibody HA Tag Monoclonal Antibody 5B1D10 (1:150, Thermo Scientific) and a custom antibody generated by Abyntek (1:150) at 4° C. overnight. Next day, cells were washed three times with PBS and secondary antibody Alexa-Fluor488 goat anti-mouse and Alexa-Fluor 488 goat anti-rabbit was added at 1:500 dilution and incubated for 1 hour at room temperature in the dark. Finally, cells were washed three times with PBS, mounted in VECTASHIELD® Mounting Medium with DAPI (PALEX MEDICAL, S.A.) and observed at 488-500 nm excitation using a Nikon Eclipse Ti-E inverted microscope system (Nikon, Melville, NY). Annexin V Assay After lentiviral infection and puromycin selection for 72 h, infected cells were washed twice with cold PBS and resuspended in Binding Buffer at a cell density of 1×106cells/mL. Following manufacturer's instructions (BD Pharmingen™) 100 μL of cells were treated with 5 μL of Annexin V-FITC. After incubation for 15 min in the dark at room temperature, 400 ul of Binding Buffer was added and each sample was supplemented with 10 μL of Propidium Iodide. Flow cytometry analysis was performed on a LSR Fortessa cytometer. Cell Cycle Analysis and Cell Viability Assays Micropeptide of SEQ ID NO: 1 expression was induced in BxPC3 cells by treatment with doxycycline (2 μg/ml) for 4 days. Cells were fixed using 100% ethanol at 4° C., stained with Propidium Iodide, and cell cycle was analysed by flow cytometry (LSR-Fortessa cytometer). Cell viability in micropeptide of SEQ ID NO: 1-expressing cells was evaluated using CellTiter-Glo 2.0 (Promega) following manufacturer's instructions. For drug sensitivity assays, doxycycline induction was coupled with Doxorubicin (1.5 mM) or Actinomycin D (5 nM) (from days 2 to 4), and cell viability was measured by staining with crystal violet. SA-β-G Staining Assay For SA-β-gal staining, cells were fixed and stained using a commercial senescence β-galactosidase staining kit (Cell Signalling) following manufacture's protocol. Cell Proliferation Assay A549 and H10 cell lines were dissociated by trypsin digestion and seeded into 24-well cell culture plates at density 5×103cells/well. Every two days, cells were dissociated from wells and counted using a Neubauer chamber. Cell growth curves were monitored for 14 days. Wound Healing Assay MIA PaCa-2 and BxPC-3 cells were trypsinized, counted, and 400,000 cells/well were seeded in 6-well plates. Next day, a pipette tip was used to scratch the surface of cell monolayer, forming a wound. An Olympus CeIIR microscope equipped with a Hamamatsu C9100 camera was used to follow the closure of the wound. Invasion Assay BxPC-3, MDA-MB-231, A549 and H10 cell lines were trypsinized, counted, and 20.000 cells/well were seeded in triplicate in a Corning® cell culture insert (Boyden chamber) in which the porous membrane is coated with a layer of Matrigel® to mimic an extra cellular matrix (ECM). Cells were allowed to invade through the coated membrane for 24 hours in the cell incubator at 37° C. After the incubation time, culture inserts were fixed in methanol 5 minutes at RT and stained using Crystal Violet 20 minutes at RT. The excess of Crystal Violet staining was removed washing with PBS and pictures of the chambers were taken using an Olympus CeIIR microscope. Invading cells were counted using ImageJ and fold change was calculated normalizing to the control. In order to test CAFs pro-invasive activity, 40.000 CAFs/well were seeded in 24-well plates. 48 h later, PDAC cells were trypsinized, counted, and 20.000 cells were seeded in the upper compartment of Matrigel® coated inserts that were placed over CAFs containing wells. CAFs and PDAC cells were this way co-cultured for 24 h, and stained as described above. Example 1: Micropeptides Identification The authors of the invention have identified 3 micropeptides corresponding to sequences SEQ ID NO: 1, 2 and 3. The micropeptide of SEQ ID NO 1 is a highly conserved 87 aa micropeptide whose sequence is: (FIG. 1A)MEGLRRGLSRWKRYHIKVHLADEALLLPLTVRPRDTLSDLRAQLVGQGVSSWKRAFYYNARRLDDHQTVRDARLQDGSVLLLVSDPR. In silico analysis of the amino acid sequence predicts a 3D structure resembling the protein UBIQUITIN (FIG.1B). SEQ ID NO 1 micropeptide is coded by the lncRNA TINCR (LINC00036 in humans and Gm20219 in mice). The micropeptide of SEQ ID NO: 2 is a 64-amino acid micropeptide whose sequence is: (FIG. 2A)MVRRKSMKKPRSVGEKKVEAKKQLPEQTVQKPRQECREAGPLFLQSRRETRDPETRATYLCGEG. It is encoded by ZEB2 antisense 1 (ZEB2AS1) long non-coding RNA (lncRNA). ZEB2AS1 is a natural antisense transcript corresponding to the 5′ untranslated region (UTR) of zinc finger E-box binding homeobox 2 (ZEB2). The ORF encoding the micropeptide spams part of the second and third exons of the lncRNA. I-Tasser, a 3D protein structure predictor, has been used in order to build a model of SEQ ID NO: 2 micropeptide 3D structure (FIG.2B). Further in-silico analysis has revealed high amino acidic sequence conservation across the species and a potential cytoplasmatic localization of the micropeptide of SEQ ID NO: 2. The micropeptide of SEQ ID NO: 3 is a 78-amino acid micropeptide encoded by the first exon of LINC0086 lncRNA. Its sequence, highly conserved across evolution is: (FIG. 3A)MAASAALSAAAAAAALSGLAVRLSRSAAARGSYGAFCKGLTRTLLTFFDLAWRLRMNFPYFYIVASVMLNVRLQVRIE. In silico analysis of this sequence predicted a tertiary structure (FIG.3B) with a transmembrane domain at C-terminal of the protein and a signal peptide in the first 25 amino acids. Example 2: Overexpression of the Micropeptide of SEQ ID NO: 1 Induces Cell Cycle Arrest, Cellular Senescence, and Sensitizes Cancer Cells to Stress in Pancreatic and Breast Cancer Cells To determine the biological function of the micropeptide, the ORF encoding for SEQ ID NO: 1 was cloned in frame with the HA epitope tag in both, a retroviral expression vector (pMSCV) and in a doxycycline-inducible lentiviral vector (pINDUCER20). BxPC-3 pancreatic cancer cell line was transduced with the pINDUCER20-SEQ ID NO: 1-HA vector, and infected cells were selected with Neomycin. Upon treatment with doxycycline, SEQ ID NO: 1 expression was detected by qPCR (FIG.4A) and by Western Blot (FIG.4B), demonstrating that the micropeptide is expressed and stable in cells. Immunostaining using a custom-made antibody against SEQ ID NO: 1 revealed a cytoplasmic cellular localization (FIG.4C). The micropeptide of SEQ ID NO: 1 expression significantly reduces cell proliferation (FIGS.5A and5B) and induces cell cycle arrest in G1 phase (FIG.5C), together with the upregulation of the CDK inhibitors p27 and p21 (FIGS.5D and5E), and an increase in the number of senescent cells (SA-β-Gal-positive cells) (FIGS.5F and5G). Remarkably, the micropeptide of SEQ ID NO: 1 expression induces an extensive cell death when coupled with cellular stress, such as the one produced by Puromycin during selection of infected cells (when using pMSCV non-inducible vector), as shown in several pancreatic and breast cancer cell lines (FIG.6C). Annexin V/PI assay revealed a substantial increase in the percentage of early apoptotic (22.4%) and late apoptotic cells (58.6%) in SEQ ID NO: 1-Puromycin conditions compared with the controls (FIG.6D). In agreement with these results, the micropeptide of SEQ ID NO: 1 expression sensitizes to cell death induced by drugs as Doxorubicin and Actinomycin D (FIG.6E). Example 3: Overexpression of the Micropeptide of SEQ ID NO: 1 Decreases Cell Proliferation and Induces Cell Cycle Arrest in Lung Adenocarcinoma Cells and Squamous Cell Carcinoma Cells Lung cancer cell line A549 and squamous cell carcinoma cell line H10 expressing inducible SEQ ID NO: 1-HA vector were established as described previously. SEQ ID NO: 1 expression was detected by qPCR (FIG.7A) and by Western Blot (FIG.7B). Immunostaining using a custom-made antibody against SEQ ID NO: 1 reveals a predominant cytoplasmic localization with a filamentous pattern. This data demonstrates that the micropeptide can also be expressed and detectable in these cell lines. To evaluate the effects of SEQ ID NO: 1 on proliferation, A549 and H10 cells transduced with SEQ ID NO: 1-HA vector or control vector were monitored for 14 days. Growth curves show that cells overexpressing micropeptide SEQ ID NO: 1 have a consistently lower growth rate compared to the control (FIG.8A). This effect in proliferation is also accompanied by an increase in cells arrested in G1 phase (FIG.8B). Collectively with the data shown before in the pancreatic cell line BxPC-3, there is a strong evidence of the role of the micropeptide of SEQ ID NO: 1 in decreasing cell proliferation in several cancer types (pancreas, lung and squamous cell carcinoma). Example 4: The Micropeptide of SEQ ID NO:1 Impairs Cell Invasion Capability and Downregulates the Mesenchymal Program Given the effect of the overexpression of the SEQ ID NO:1, further experiments were performed to study its effect in cell invasion, another key oncogenic trait. Boyden chamber assay was used to determine invasiveness of A549 and H10 cancer cells after 4 days of doxycycline induction of SEQ ID NO:1, showing that the expression of the micropeptide induces a significant decrease in invasion (FIGS.9Aand B). In line with this observation, overexpression of the micropeptide of SEQ ID NO: 1 represses the expression of the EMT regulators VIMENTIN, SLUG, SNAIL, N-CADHERIN, TWIST1, TWIST2, ZEB1 and ZEB2 in H10 SCC cell line (FIG.9C). This downregulation of the mesenchymal program further validates the role of the micropeptide of SEQ ID NO: 1 as a tumor suppressor. Example 5: Transcription and Translation of Micropeptide of SEQ ID NO:1 is Increased Upon Genotoxic Stress in a p53-Dependent Manner The tumor suppressor gene p53 is well known to have a role in controlling apoptosis, cell cycle arrest, senescence and cell migration and invasion upon activation when DNA is damaged. Due to the potential tumor suppressor function of micropeptide of SEQ ID NO:1 further studies were performed to study how both proteins could be connected. Different cell lines (A549, BxPC-3 and HCT116) were treated with different DNA-damaging agents commonly used in cancer treatment (Doxorubicin and Actinomycin D) and one p53 stabilizer (Nutlin-3a) to promote p53 activation. Then, the expression of SEQ ID NO:1 were measured, and it was observed that SEQ ID NO:1 is induced (mRNA and protein levels) upon genotoxic stress (FIGS.10A, B and C). Importantly, only the cells with wild-type p53 (HCT116 and A549) show a transcriptional and translational upregulation of SEQ ID NO:1 (FIG.10A). On the other hand, cells with a p53 status altered, protein knock-out (HCT116 p53KO) or pathogenic mutant p53 (BxPC-3), do not shown the same response (FIG.10B). As a tumor suppressor, p53 is responsible for protecting cells from oncogenic alterations. Activation of p53 can promote apoptosis of tumor cells and is considered a key mechanism of action of some antitumor drugs, as Doxorubicin. Inactivating mutations of p53 are frequently observed in various human cancers and are known to confer tumor resistance. These results suggest that SEQ ID NO:1 is regulated in a p53-dependent manner and thereby, possibly downregulated in all cancers with mutational inactivation of p53. Therefore, its tumor suppressor activity can be related with p53 function, supporting the use of the micropeptide as a therapeutic agent in cancer. Example 6: The Micropeptide of SEQ ID NO: 1 Encodes a Ubiquitin-Like Protein that Changes Cell Ubiquitylation Pattern Structural modelling by in silico tools predicted the micropeptide of sequence SEQ ID NO: 1 as an ubiquitin-like protein (FIG.1B). Ubiquitin is a highly conserved small protein widely present in all eukaryotic cells, which can exist either not anchorage or covalently attached to another protein. When conjugated to its target proteins it orchestrates important biological processes, as protein degradation via the proteasome, DNA repair, cell-cycle regulation, signalling cascades and DNA-damage responses. Recently, it has been demonstrated that ubiquitin-like proteins (ULPs), which have structural similarity to ubiquitin, are also present in cells and have protein-conjugation capability that can resemble ubiquitylation (Hochstrasser, 2009). Given the structural features of the micropeptide of sequence SEQ ID NO: 1, ubiquitin-related functions of this micropeptide were tested. Immunoprecipitation experiments have shown that the micropeptide of sequence SEQ ID NO: 1 binds to ubiquitylated proteins (FIG.11A). Moreover, micropeptide of sequence SEQ ID NO: 1 overexpression alters protein ubiquitylation pattern in BxPC-3 infected cells (FIG.11B). Altogether, our results suggest that micropeptide of SEQ ID NO:1 induces cell cycle arrest, cellular senescence, sensitizes cancer cells to stress and reduces cell invasion and mesenchymal traits, possibly by changing cell ubiquitylation pattern. This opens the possibility of using micropeptide of SEQ ID NO:1 as a therapy to reduce tumour growth and invasiveness and make them more sensitive to chemo/radiotherapy in combined therapies. Example 7: Overexpression of the Micropeptide of SEQ ID NO: 2 Downregulates EMT Factors and Reduces Migration and Invasion in Pancreatic Adenocarcinoma The ORF encoding the micropeptide of SEQ ID NO: 2 was cloned into pMSCV retroviral vector fused to a HA-tag. The construct thus obtained was used to retrovirally transduce human primary fibroblasts (IMR90), pancreatic cancer cell lines (MIA PaCa-2 and BxPC-3) and murine pancreatic cancer associated fibroblasts (mCAFs). The overexpression of the micropeptide of SEQ ID NO: 2 was confirmed in all the cell lines by quantitative PCR and Western Blot analysis using a HA-tag antibody (FIGS.12and13). Immunostaining using a custom polyclonal antibody specific for the micropeptide of SEQ ID NO: 2 showed that the micropeptide of SEQ ID NO: 2 was expressed and located in the cytoplasm (FIG.14). Then, the inventors tested the effect of the overexpression of the SEQ ID NO: 2 micropeptide in epithelial-to-mesenchymal transition (EMT) related genes expression. Of note, the micropeptide of SEQ ID NO: 2 induces the loss of mesenchymal identity in primary fibroblasts and pancreatic cancer cell lines as well as in pancreatic cancer associated fibroblasts, as shown by the downregulation of the EMT regulators SNAIL, SLUG, ZEB1, ZEB2, TWIST1 and VIMENTIN (if expressed), and adhesion factors such as COLLAGEN I, COLLAGEN IV, CD44 and FIBRONECTIN (FIGS.15and16). Remarkably, the decreased expression of adhesion molecules in mCAFs is also accompanied by a downregulation of METALLOPROTEINASE-2 (FIG.16). Adhesion molecules, as well as metalloproteinases, are factors secreted by cancer associated fibroblasts that are required in order to create a pro-metastatic environment (Hwang R F et al., Cancer Res. 2008; 68:918-26 and Vonlaufen A et al., Cancer Res. 2008; 68:2085-93). Consistent with these results, overexpression of the micropeptide of SEQ ID NO: 2 leads to a decreased migration capability in wound healing assays in MIA PaCa-2 and BxPC-3 cell lines (FIGS.17Aand B) and a decreased invasion of BxPC-3 cell line (FIGS.18Aand B). Moreover, the downregulation of extracellular matrix factors such as COLLAGEN I, COLLAGEN IV and FIBRONECTIN togetherwith the secreted METALLOPROTEINASE-2 in cancer associated fibroblasts addresses the possibility of using the micropeptide of SEQ ID NO: 2 as an antifibrotic agent to target the pro-tumoral activity of this particular cell group within tumours, and in general in pathological fibrotic conditions. In order to test CAFs pro-invasive activity, CAFs were co-cultured with PDAC cells that were seeded in the upper compartment of Matrigel® coated inserts and allowed to invade through the matrigel layer.FIGS.19Aand B show that the micropeptide of SEQ ID NO:2 expression in CAFs reduces pancreatic cancer cell invasion in a non-cell autonomous manner, supporting the use of the micropeptide of SEQ ID NO:2 as a therapeutic agent that could potentially target both cancer cells and the stromal counterpart. Example 8: Overexpression of the Micropeptide of SEQ ID NO: 2 Downregulates EMT Factors and Impairs Invasion in Triple Negative Breast Cancer The construct containing the micropeptide of SEQ ID NO: 2 (explained in Example 7) was also used to retrovirally transduce a human triple negative breast cancer cell line (MDA-MB-231). The overexpression of the micropeptide of SEQ ID NO: 2 was confirmed in MDA-MB-231 cells by quantitative PCR and Western Blot analysis using a HA-tag antibody (FIGS.20Aand B). The overexpression of the micropeptide of SEQ ID NO: 2 induces the loss of mesenchymal identity in human triple negative breast cancer cell line. Of note, the downregulation of the EMT regulators SLUG, ZEB1, ZEB2 and VIMENTIN in MDA-MB-231 is followed by the upregulation of epithelial markers such as E-CADHERIN and CYTOKERATIN-5 (FIG.21), further validating the role of the micropeptide of SEQ ID NO: 2 in promoting an epithelial phenotype. Consistent with these results, the overexpression of the micropeptide of SEQ ID NO: 2 leads to a decreased invasion capability in invasion assays of MDA-MB-231 breast cancer cell line (FIG.22), altogether indicating that the micropeptide of SEQ ID NO:2 could act as an EMT negative regulator, contributing to decrease cancer malignant features such as invasion, metastasis and resistance to chemotherapeutic drugs. Example 9: Overexpression of the Micropeptide of SEQ ID NO: 3 Induces Cell Death The ORF encoding the micropeptide of SEQ ID NO: 3 was cloned in frame with the HA epitope tag in the pMSCV retroviral vector. Western blot and qPCR analysis demonstrated that the micropeptide of SEQ ID NO: 3 was successfully expressed after retroviral transduction, and that the protein product was stable (FIG.23Ay23B). Importantly, overexpression of the micropeptide of SEQ ID NO: 3 induces massive cell death in cancer cell lines (A549, human lung cancer and HCT116, human colorectal cancer) (FIG.23C). Example 10: Overexpression of the Micropeptide of SEQ ID NO: 3 Induces p21 and a Pro-Apoptotic Program The impact of the overexpression of the micropeptide of SEQ ID NO: 3 on the expression of the CDK inhibitor (and tumor suppressor) p21 was tested. Interestingly, overexpression of the micropeptide of SEQ ID NO: 3 resulted in a substantial p21 upregulation in A459 and HCT116 cells (FIG.24A). This correlated with increased p21 protein levels upon SEQ ID NO: 3 micropeptide overexpression, as shown inFIG.24B. Given the induction of cell death by the micropeptide of SEQ ID NO: 3 and the role of p21 in apoptosis, the authors of the invention tested the expression of apoptosis-related genes. Overexpression of the micropeptide of SEQ ID NO: 3 induces the upregulation of the pro-apoptotic genes BAX and BAK and the downregulation of the antiapoptotic gene BCL-2 in HCT116 colorectal cancer cell line (FIG.24C). Example 11: SEQ ID NO:3 is Upregulated Upon Genotoxic Stress in a p53-Dependent Manner As previously indicated, p53 is an important tumour suppressor. Given the potential role of micropeptide SEQ ID NO:3 in tumor suppression, the authors of the invention tested the potential regulation of SEQ ID NO:3 by stress and by p53 protein. The authors of the invention treated the isogenic cell lines HCT116 and HCT116 p53 knock-out with the p53 activator Nutlin3a (10 μM) and with the genotoxic chemotherapeutic agent Doxorubicin (1 μM). Interestingly, SEQ ID NO 3 was upregulated with genotoxic stress in HCT116, but the upregulation was impaired in HCT116 p53 KO (FIG.25). These results suggest that SEQ ID NO:3 regulated by stress/damage in a p53-dependent manner and thereby, possibly downregulated in all cancers with mutational inactivation of p53. Therefore, its tumor suppressor activity can be related with p53 function, supporting the use of the micropeptide as a therapeutic agent in cancer.
99,328
11858973
DETAILED DESCRIPTION OF THE INVENTION Examples Example 1 Materials and Methods Cell Cultures Cell lines. The following cell lines of human origin were used:1. Jurkat, T-cell acute lymphoblastic leukemia cell line, clone E6-1, negative for CEA expression and positive for Tn expression.2. HT-29, colon adenocarcinoma cell line, positive for CEA expression.3. HeLa-CEA, cervical cancer cell line transfected with a cDNA construct encoding CEA and thereby expressing high levels of this antigen. The transfection was carried out in the laboratory of Dr. Laura Sanz, Hospital Puerta de Hierro, Madrid.4. MDA-MB-231, breast adenocarcinoma cell line, negative for Tn expression.5. MCF-7, metastatic breast adenocarcinoma cell line, positive for Tn expression.6. Capan-2, pancreatic carcinoma cell line, positive for Tn expression. All the cell lines were originally obtained from the American Type Culture Collection (ATCC, USA). Cell Culture Maintenance The cells were incubated in a temperature-controlled incubator at 37° C. in moist air and with 5% CO2. The filter cap culture flasks, pipettes, and all the material for that purpose were used under sterile conditions in the laminar flow hood of the Biochemistry and Molecular Biology Department of UNIZAR. The Jurkat cells were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS), 100 U/ml of penicillin, 100 ug/ml of streptomycin, and 2 mM GlutaMAX. The culture was maintained with a density of up to one million cells per ml, which corresponds to one passage every 3 or 4 days. The rest of the cell lines were cultured in Dulbecco's Modified Eagle's Medium (DMEM), Catalog No. 30-2002. This medium was supplemented with 10% FBS, 100 U/ml of penicillin, 100 ug/ml of streptomycin, and 2 mM GlutaMAX. For the passages, considerations were given to ATCC recommendations on the concentration of cells in the medium, i.e., about one million cells per ml before trypsinization and the subculture ratios depending on each cell type, in that sense CAPAN-2 required ratios of 1:2 to 1:4, MIA PACA-2 required ratios of 1:3 to 1:8, etc. These considerations were used for expanding the cells for in vivo assays. Likewise, according to ATCC recommendations, cryopreservation of the cell lines was performed in 95% FBS and 5% dimethylsulfoxide (DMSO) at a cell density of 5×106/ml. First, the cells were frozen for 24 to 48 hours in a freezer at −86° C. and then transferred to a liquid nitrogen container. For the process of thawing the cells, a wash was performed with 10 ml of culture medium and the cells were then seeded at a density that was twice the density of that used for the passages. Cell Count and Viability Cell viability was determined by means of a 0.4% trypan blue solution in 0.15 M of NaCl (Sigma, Madrid), this dye being capable of entering the cell cytoplasm with a loss of cell membrane integrity. After mixing the dye with the cell suspension (50 ul of each), the cells were counted in a Neubauer chamber (hemocytometer) and observed under a microscope (Optiphot, Nikon). The formula used for calculating cell density is as follows: Concentration=Total⁢⁢cells⁢⁢counted×10,000Number⁢⁢of⁢⁢squares The percentage of cell viability was calculated from this formula so that assays are always performed with a viability greater than 90%. Anti-Tn Chimera Cloning Starting genetic material: the pPICZα integrative plasmid was used, with this construct having the following characteristics:pUC ori: allows plasmid replication inE. coliAOX1 promoter: induced by methanol and directs plasmid integration in the AOX1 locus ofPichiaby means of homologous recombination.Factor α: allows efficient protein secretionMultiple cloning site: allows DNA insertion in the expression vectorc-myc epitope: allows detection with anti-myc antibodyPolyhistidine tag: facilitates recombinant protein purificationAOX1 transcription terminator: increases mRNA stability by allowing efficient mRNA 3′ end processing, including polyadenylationZeocin resistance gene (zeocin being a broad-spectrum antibiotic): serves as a selection marker. It is preceded by the TEF1 and EM7 promoters and followed by the CYC1 transcription terminator. The cDNA of human GRNLY and the cDNA of human GRNLY-conjugated SM3 and Ar20.5 are directionally integrated between the cleaving sites of restriction enzymes Cla I and Xba I found at the multiple cloning site of pPICZαC and pPICZαA, within the reading frame. The plasmid pPICZαC-GRNLY was synthesized by Dr. Laura Sanz (Hospital Puerta de Hierro, Madrid) from the human GRNLY DNA sequence kindly donated to the group by Dr. Alan Krensky (Northwestern University, Chicago). The plasmids pPICZαA-SM3 13° and pPICZαA-Ar20.5 were kindly donated by Dr. Ramón Hurtado (Institute for Biocomputation and Physics of Complex Systems BIFI of the University of Zaragoza). Competent Cell Preparation DH5α E. coliwas made “competent” for transformation by means of opening the pores in the membrane with CaCl2at a low temperature. The method consisted of seeding p DH5α E. coliin a 50 ml tube with 10 ml of liquid LB medium under stirring (180 rpm) at 37° C. overnight. 200 ul of that product were used for diluting in 20 ml of fresh LB medium and for repeating incubation until reaching an absorbance of about 0.3-0.4 at 600 nm, which is equivalent to a concentration of 5-10×107bacteria/ml). It was then incubated on ice for 30 minutes and centrifuged for 8 minutes at 4° C. and 8000×g. It was washed once with sterile water, incubated in 10 ml of 50 mM cold CaCl2for 15 minutes and centrifuged again for 8 minutes at 3000×g. Finally, the decant was resuspended in 4 ml of a 50 mM CaCl2solution with 15% glycerol and frozen at −80° C. until use. Bacterial Transformation and Expansion The transformation of competent p DH5α E. coliwith pPICZαC-Ar20.5-GRNLY or pPICZαC-SM3-GRNLY was performed by heat shock. Two aliquots of about 200 ul were thawed by incubating them on ice and 50 ng of plasmid DNA were added in each aliquot (these methods were performed by blazing in a flame) for 20 minutes, it was introduced in a 42° C. bath for a minute and a half and again for two minutes on ice. 1 ml of liquid LB medium was added in each aliquot and it was incubated at 37° C. under stirring at 200 rpm for two hours. The transformed bacteria were then seeded in solid LB medium (1.5%) with zeocin (25 μg/ml) at 37° C. overnight, such that the bacteria incorporating the plasmid grew. From the colonies that developed, four colonies were randomly selected for each protein and cultured in 50 ml tubes with 10 ml of liquid LB medium and 25 ug/ml of zeocin (one colony per tube) under stirring at 180 rpm at 37° C. overnight. Selection of Bacteria with a Higher Granulysin Expression Level The plasmid was isolated by means of minipreparation with the “Nucleospin® Plasmid Easypure” kit. To check if plasmids with the suitable molecular weight were isolated, electrophoresis was performed in 1% agarose gel made up of 0.3 g of agarose (Scharlau), 30 ml of 1× TAE buffer (Tris-Acetate-EDTA buffer, Invitrogen, pH 8.3), and for DNA staining 3 μl of SYBR® Safe (Invitrogen). The gels were viewed in a Gel Doc 2000 transilluminator (BioRad). To expand the plasmids to a larger amount, transformedE. Colicolonies were cultured in LB medium with zeocin (25 μg/ml) using the “Quantum Prep™ Plasmid Midiprep” kit. In both cases, the concentration and purity of the obtained plasmids were determined by means of a NanoDrop® (NanoVue) apparatus. Starting Genetic Material: pPICKαC-GRNLY And pPICKαC-Chimera pPICZαC is an commercially available integrative plasmid [pPICZα A, B, and C:Pichlan expression vectors for selection on Zeocin™ and purification of secreted, recombinant proteins, MAN0000035, Invitrogen, Corporate Headquarters, Rev. Date: Jul. 7, 2010] containing, as shown inFIG.2, the following characteristics:pUC ori: allows plasmid replication withinE. coliAOX1 promoter: induced by methanol and directs plasmid integration in the AOX1 locus ofPichiaby means of homologous recombination.Factor α: allows efficient protein secretionMultiple cloning site: allows DNA insertion in the expression vectorc-myc epitope: allows detection with anti-myc antibodyPolyhistidine tag: allows easier recombinant protein purificationAOX1 transcription terminator: increases mRNA stability by allowing efficient mRNA 3′ end processing, including polyadenylationZeocin resistance gene (zeocin being a broad-spectrum antibiotic): serves as a selection marker. It is preceded by the TEF1 and EM7 promoters and followed by the CYC1 transcription terminator. The cDNA of human GRNLY and the cDNA of human GRNLY-bound MFE23 are directionally integrated between the cleaving sites of restriction enzymes Cla I and Xba I found at the multiple cloning site of pPICZαC (framed inFIG.2), within the reading frame. The human granulysin cDNA sequence (SEQ ID No. 1) was kindly donated to the group by Dr. Alan Krensky (Northwestern University, Chicago). Chimera Structure and Parameters In generic terms, the fusion protein that is obtained has the following structure: MFE23-LINKER-GRANULYSIN (9 kDa)-HISTIDINE TAG TABLE 1Chimera parameters obtained with the ProtParam tool (Expasy) based on the amino acids thereofCHIMERA (MFE23-GRNLY)Extinction coefficient at280 nm (M−1cm−1)Extinction coefficient atNumber ofTheoreticalassuming that all Cys280 nm (M−1cm−1)aminoMolecularisoelectric pointpairs form disulphideassuming that all Cysacidsweight (kDa)(pI)bridgesresidues are reduced34737.10699.036284062340 Pichia pastorisTransfection by Means of Electroporation The plasmids must remain linear so that they can be more easily integrated in the genome ofPichia pastoris. To that end, digestion is performed with the enzyme Sacl, which has a single cleaving site within pPICZαC and does not cleave within the inserts to be introduced. Next, to check that the plasmid has been properly digested, electrophoresis is carried out in 1% agarose gel. After checking that the digestion was satisfactory, digestion residues (salts and residual agarose gel) are cleaned away and the plasmid is purified using the AccuPrep® Gel Purification kit. The prior treatments described in the manual [Pichia Expression Kit, Invitrogen, Rev. Date: Oct. 1, 2014], first with 100 mM LiAc solution, 10 mM DTT, 0.6 M sorbitol, and 10 mM Tris-HCL, pH 7.5, and then with another 1 M sorbitol solution, are necessary before proceeding to the electroporation ofPichia pastorisSMD1168 with the plasmids. The electroporator (BIORAD MicroPulser™) is set to the program for electroporatingPichia. Furthermore,Pichia pastorisSMD1168 cells electroporated in the absence and presence of the linearized plasmid are plated on plates containing zeocin (cells which do not contain the plasmid would not grow in plates with zeocin, and this accordingly indicates that the electroporation was satisfactory) and the plates are left in the oven at 30° C. throughout the entire electroporation process by way of control. The transfected colonies are selected, plating them on a YPDS plate (10 g/L of yeast extract, 20 g/L of peptone, 2% dextrose, 182.2 g/L of sorbitol, pH 6) with zeocin (200 μg/ml) and the plates are incubated for 3-10 days at 30° C. until colonies emerge. Finally, some well isolated colonies which have grown in the plate with zeocin are chosen and plated again in a new plate so as to later choose the best strain. Recombinant Protein Production inPichia pastorisCultures The selected colonies are inoculated in BMGY medium (10 g/L of yeast extract, 20 g/L of peptone, 100 mM phosphate buffer, pH 6, 13.4 g/L of nitrogenated yeast base without amino acids or ammonium sulfate, 1 ml/L of glycerol, and 0.4 mg/L of biotin), being cultured at 30° C. for a day for the yeast to grow. A change of medium to BMMY (10 g/L of yeast extract, 20 g/L of peptone, 100 mM sodium acetate buffer, pH 5, 13.4 g/L of nitrogenated yeast base without amino acids or ammonium sulfate, 0.5 ml/L methanol, and 0.4 mg/L of biotin) is then performed, maintaining the culture at 18° C. under stirring for a day for the induction of the expression of the recombinant protein to begin. After the first day of induction and every 24 hours for 2 more days, methanol is added at a final concentration of 1% in the culture medium and it is left under stirring until the next day at 18° C. First, a small-scale production of the selected colonies was performed. Once the colonies producing the most recombinant protein were chosen, different pH and temperature conditions were tested to find the optimum production conditions. Finally, large-scale recombinant protein production was performed. Recombinant Protein Purification The yeast supernatant is filtered by means of a vacuum filtration system first with a 0.45 μm filter and then with a 0.22 μm filter. It is then concentrated in a Pellicon XL Ultracel 5 kDa 0.005 m2concentrator (Millipore) from 1 L to about 50 ml. Next, in the case of the chimera, dialysis is performed using a dialysis bag (Millipore), and in the case of the recombinant GRNLY, dialysis is performed using a “Slide-A-Lyzer™” dialysis cassette (Thermo Scientific Pierce) with a membrane having a pore size of 3.5 kDa due to the small molecular weight thereof. The dialysis bag or membrane is immersed in 5 L of washing buffer (300 mM NaCl, 50 mM Tris-HCl, and 20 mM imidazol, pH 7.4) and left to dialyze overnight. Dialysis is performed to change the medium originating from the yeast supernatant in which GRNLY can be found by the buffer that will be used later in nickel affinity chromatography. Imidazol is found at a low concentration in the buffer such that it competes with molecules that do not specifically bind to the nickel column. Affinity Chromatography Recombinant GRNLY and chimera are expressed with a histidine tag for the purpose of purifying them by means of nickel affinity chromatography since the imidazol rings of histidines have a high affinity for Ni2+cation. To that end, the Ni-NTA agarose resin (Qiagen) is mixed with washing buffer (300 mM NaCl, 50 mM Tris-HCl, and 20 mM imidazol, pH 7.4), centrifuged at 2500 rpm for 2 minutes, and the supernatant is removed. Washing buffer is then added and it is centrifuged again at 2500 rpm for 2 minutes to remove the supernatant. The resin is then resuspended in washing buffer and mixed with the resulting solution after dialysis. It is then placed in a rotating end-over-end shaker at 4° C. for about an hour and a half. It is then centrifuged at 2500 rpm for 5 minutes. The precipitate is washed three times with washing buffer, rotated in an end-over-end shaker for 15 minutes, and centrifuged at 2500 rpm for 5 minutes. The precipitate is placed in a column with washing buffer, and the resin is left to settle. The column is eluted with elution buffer (500 mM imidazol, 300 mM NaCl, and 50 mM Tris-HCl, pH 7,4). The amount of protein in the eluded fractions originating from affinity chromatography is then quantified by means of a NanoDrop® apparatus (NanoVue). The elution fractions containing an acceptable amount of protein are then pooled. Buffer and Concentration Change To change the elution buffer for PBS, the chimera in elution buffer is passed through a column with Sephadex G-25 (Thermo-Fisher) previously equilibrated with PBS and concentrated with an Amicon filter having a membrane pore size of 15 kDa (Millipore), or the buffer is directly changed and concentrated with said Amicon filter. In the case of the recombinant GRNLY, the buffer is changed and the elution is concentrated at the same time by means of an Amicon filter having a membrane pore size of 3 kDa. Finally, with a NanoDrop® apparatus (NanoVue), the protein concentration in the final concentrate is measured and it is sterilized by filtration through a 0.22 μm filter. Coomassie Staining and Immunoblot To perform the expression test, denaturing electrophoresis is performed in 12% acrylamide gel made up of two types of gels having the same composition but in different proportions (stacking gel and resolving gel), loading the supernatant obtained from each colony together with a molecular weight marker. Said gels are then stained with Coomassie Blue to find out which colony produced the highest amount of recombinant protein. Furthermore, in all the purification steps aliquots are kept at 4° C. to enable analyzing them by means of electrophoresis of the different aliquots in 12% polyacrylamide gel, Coomassie Blue staining is performed, and immunoblot is also performed by transferring the proteins separated in the gel to nitrocellulose membranes according to the previously described method [Anel, A., et. al., J Biol Chem, 1993. 268 (23): p. 17578-87] and incubating the membrane with a rabbit polyclonal primary antibody kindly donated by Dr. Carol Clayberger (Northwestern University, Chicago). After washing, a peroxidase-conjugated rabbit anti-IgG secondary antibody (Sigma) is then added. It is later washed with buffer B (PBS with 0.05% of Tween-20, pH 7.4) under stirring to remove excess antibodies. The complexes are detected by means of chemiluminescence (ECL) development. This technique is based on the detection of the light emitted as a result of the oxidation of luminol, a chemiluminescent substrate, by peroxidase. This light is captured by photographic films (high-performance chemiluminescence film, GE HealthCare) in the dark and following membrane incubation with “Pierce® ECL Western Blotting Substrate” (Thermo Scientific). The films are exposed in a radiological developing cassette (Hypercassette™, Amersham Bioscience) and in a dark room with suitable lighting for photographic development. The films are developed after exposure by means of immersion in developer-distilled water-fixer solutions, varying the developer solution time according to the signal that is obtained. Specificity Assay An ELISA was performed using 200 ng of CEA per well, which was incubated overnight at 4° C., PBS (negative control), 500 ng of the recombinant scFv MFE23 (positive control), and theP. pastorissupernatant transformed with pPICZαC-Chimera after inducing expression with methanol were then added. To develop the binding of scFvs to the CEA antigen, an anti-histidine tag antibody and a peroxidase-conjugated goat anti-mouse IgG secondary antibody were used. The peroxidase substrate was OPD, producing a yellow-orange product that can be detected at 492 nm. Flow Cytometry For the purpose of studying the binding of the chimera to the CEA antigen, an experiment was performed with HT29 cells (ATCC) which express the CEA antigen on the surface, and with Jurkat cells (ATCC) which do not express the CEA antigen on the surface as a negative control. These cells were deposited in a round bottom 96-well plate at a concentration of 100,000 cells per well, washed with PBS with 5% FBS, and chimera was added (10 μg/ml). After incubating for 1 hour at 4° C. and washing with PBS with 5% FBS, they were labeled with anti-HIS murine antibody (1:200), and after incubating for another hour at 4° C., an FITC-conjugated anti-mouse IgG antibody was added (1:200). Finally, it was incubated for another hour at 4° C. and fluorescence was analyzed by means of flow cytometry. In this manner, fluorescence is observed if the chimera binds to the surface of the cells. Several negative controls were carried out in the absence of chimera and/or antibodies to assure that the chimera binds specifically to CEA. Fluorescence Microscopy Furthermore, the same process was performed on a slip in a 24-well plate with HT29 cells, the chimera, and anti-His and FITC-conjugated anti-mouse IgG antibodies, after which Hoechst 33342 staining was performed to enable viewing the cell nuclei and performing analysis by means of fluorescence microscopy. In Vitro Cytotoxicity Assay The cytotoxicity of GRNLY or of the chimera was assayed on T-cell acute leukemia Jurkat which constitutes the standard for sensitivity to GRNLY in this laboratory, and on human colon carcinoma HT29. The Jurkat cells grow in suspension in RPMI 1640 culture medium (Gibco®) supplemented with 5% unsupplemented fetal bovine serum, glutamine, and antibiotics, and do not express the CEA antigen. The HT29 cells are adherent cells which grow in DMEM culture medium (Gibco®) supplemented in a similar manner and express the CEA antigen on their surface. Adherent cells must be trypsinized for handling and reseeding. In the control samples, a volume of PBS which is equivalent to the added volume of GRNLY or the chimera is added on the cells. In the case of the Jurkat cell line, cells at a concentration of 30000 cells per well, PBS, or the chimera/GRNLY are added, the cells are seeded in 96-well plates, and incubated at 37° C. with 5% CO2for 24 hours. In the case of the HT29 cell line, first only the cells are seeded at a concentration of 30000 cells per well in DMEM culture medium and incubated for 24 hours at 37° C. in an incubator with 5% CO2 for the cells to adhere, and once adhered, the chimera or PBS is added and they are incubated at 37° C. for another 24 hours. An important characteristic of the apoptotic phenotype is the exposure of phosphatidylserine in the outer layer of the plasma membrane [Martin, S. J., et al., J Exp Med, 1995. 182 (5): p. 1545-56]. To measure this translocation, annexin V, a protein which binds specifically to phosphatidylserine, can be used. In late stages of apoptosis, when the plasma membrane has lost its integrity and the DNA becomes accessible, fluorophores such as 7-AAD, which act as intercalating agents in double-stranded nucleic acids, are used. In these experiments, the cells are incubated with annexin V conjugated with Alexa-46 fluorophore (Immunostep), and 7AAD (Immunostep) in the case of HT29 cells, in ABB buffer, 140 mM NaCl, 2.5 mM CaCl2, 10 mM Hepes (NaOH, pH 7.4, for 15 minutes in the dark. The percentage of apoptotic cells in each of the assay conditions can thereby be quantified by means of flow cytometry. The cytotoxicity of GRNLY had not yet been assayed on HT29 cells, such that the cytotoxicity of both GRNLY and the chimera were assayed by means of trypan blue staining. Trypan blue is a dye derived from toluidine having the capacity to stain only dead tissues and cells as it is not capable of going through the intact membranes of living cells. A microscope and a Neubauer chamber were used for the cell count. After the count, the number of cells of each well was compared with the control well which only contained cells and culture medium. The percentage of growth with respect to the control was thereby obtained. Example 2 Recombinant Protein Production inPichia astorisCultures After transfecting and culturingPichia pastorisas indicated in the materials and methods section, the recombinant protein was successfully expressed and secreted into the medium as a result of factor-α. There can be seen inFIG.3A, in all the chosen and induced colonies, a band corresponding to a molecular weight close to 11 kDa which corresponds with the weight of recombinant GRNLY given that GRNLY, which has a weight of about 9 kDa, experiences a molecular weight increase to about 11 kDa since it is fused to a histidine tag. In reality, at least two bands with very similar molecular weights belonging to different recombinant GRNLY isoforms are seen. This is because GRNLY undergoes post-translational O-glycosylation modifications. Similar observations had previously been made by another research group which wrote a similar paper in which it was determined that the different bands obtained were due to O-glycosylation [Guo, Y., et al., Appl Microbel Biotechnol, 2013. 97 (17): p. 7669-77]. Furthermore, there can be seen inFIG.3B, in all the chosen and induced colonies, a band corresponding to a molecular weight close to 35 kDa which corresponds with the weight of recombinant MFE23-bound GRNLY (FIG.3B). In view of these results (FIG.3), the decision was made to perform large-scale expression with colony 8 transformed with pPICZαC-Chimera and with colony 2 transformed with pPICZαC-GRNLY. Another group has described large-scale recombinant GRNLY production as a result of a fermenter and obtained better expression results when induction was carried out at pH 5 and 30° C. [Guo, Y., et al., Appl Microbel Biotechnol, 2013. 97 (17): p. 7669-77]. Accordingly, an expression test was performed in which induction was carried out at different pH and temperature conditions in order to check under which conditions the proteins of the present invention were best expressed. In both cases, the best results were achieved by carrying out induction at pH 5 and 18° C. (FIG.9). By carrying out induction at 30° C. and pH 5, manyPichia pastorisproteins in addition to GRNLY are expressed, and this is not of interest as it may negatively affect protein purity during purification (FIG.4A). Example 3 Recombinant Protein Purification FIG.5shows the steps of one of the purifications carried out on the chimera. In the case of GRNLY, this process was shown in an earlier paper [Ibáñez, R.,University of Zaragoza.2015]. It can be seen inFIG.5Athat theP. Pastorissupernatant obtained after induction (lane 1) contains rather diluted proteins. After concentrating same with Pellicom, protein bands are not seen in the permeate (lane 3), but proteins that are much more concentrated than in the supernatant are seen in the concentrate (lane 2). After dialysis (lane 4), the band profile remains similar to the concentrate. Furthermore, protein bands are not seen in the buffer in which the dialysis bag (lane 5) was introduced. Upon addition of the nickel resin, the chimera binds to said resin as it has a histidine tag. After adding the resin (lane 6), the intensity of a band corresponding to a protein of about 40 kDa decreases with respect to the concentrate and dialysate. This band may correspond to the chimera. The fact that this band does not altogether disappear may indicate that the nickel resin was saturated. In the washes performed on the resin, particularly in the first wash (lane 7), it can be seen how the residues of other proteins are removed. Finally, after the elution of the nickel column, a major protein with a molecular weight of about 40 kDa corresponding to the molecular weight of the chimera (lane 11) is clearly observed. As shown inFIG.5B, it was confirmed by means of immunoblot that this band of about 40 kDa corresponds to the chimera (lane 11). It is also confirmed that the resin was saturated because a band appears in the post-resin dialysis phase (lane 6). FIG.6shows different elution fractions and the pooling of all of them with the exception of elution fraction1.FIG.6Ashows several bands in the different elution fractions and in the total eluate. The band with the highest intensity has a molecular weight corresponding to the chimera. Furthermore, other bands having intermediate molecular weights are observed, which means that the chimera undergoes partial proteolysis. The band with the second highest intensity has a molecular weight of about 10 kDa, which corresponds to 9-kDa GRNLY, as its molecular weight increases since it is bound to a histidine tag. InFIG.6B, it was confirmed by means of immunoblot that these bands of about 40 and 10 kDa correspond to the chimeric recombinant protein and to recombinant GRNLY, respectively. Once the chimera is generated, its functionality must be assured, that is, on one hand the scFv still recognizes the CEA antigen, and on the other hand GRNLY is still cytotoxic. Example 4 Specificity Assay Elisa Part of the supernatant was used to check, by means of ELISA, that the chimera is capable of binding to the CEA antigen. The result showed that the chimera is capable of binding to the CEA antigen in a manner similar to scFv MFE23 (FIG.7). Flow Cytometry An experiment was carried out by means of flow cytometry and fluorescence detection using HT29 colon carcinoma cells expressing CEA or T-cell leukemia Jurkat cells not expressing CEA as a negative control. To carry out this analysis, the chimera was added under cold conditions to the cells, followed by a mouse anti-histidine tag antibody, and an FITC-bound anti-mouse IgG antibody. Fluorescent labeling will therefore be observed if the chimera binds to CEA expressed on the surface of HT29 cells. Several negative controls were carried out in the absence of chimera and/or antibodies to assure that the chimera binds specifically to CEA. As can be seen inFIG.8, although a certain increase in fluorescence is detected in Jurkat cells when the chimera and the two antibodies are added (FIG.8A), this increase is much lower than that observed when the same experiment is performed on HT29 cells (FIG.8B). The data obtained in the controls seem to indicate that the combination of the anti-His antibody with the anti-mouse IgG antibody leads to unspecific labeling which is more prominent in the case of HT29 than in the case of Jurkat cells (Table 2). However, specific labeling in the presence of the chimera is in any case more prominent in the case of HT29 cells. TABLE 2Mean fluorescence intensity (MFI) obtained after analyzing the bindingof the chimera to the CEA antigen by means of flow cytometryMFIJURKATHT29Cells2.293.4Cells + α-mouse2.463.4Cells + MFE23-GRNLY + α-mouse2.464.22Cells + α-His + α-mouse2.646.04Cells + MFE23-GRNLY + α-His + α-mouse3.928.35 Fluorescence Microscopy For the purpose of illustrating these results, fluorescence microscopy was performed, the results of which are shown inFIG.9which depicts the staining of HT29 cell nuclei with the fluorescent Hoechst 33342 molecule.FIG.9Bshows the cell nuclei surrounded by a green halo corresponding to CEA labeling by the chimera in the plasma membrane of the cells, whereas said halo is not seen inFIG.9A, used as a negative control, in which no chimera was added. Example 5 In Vitro Cytotoxicity Assay Cells from the Jurkat cell line were treated with different doses of chimera. The result was that the chimera is toxic to Jurkat cells in a dose-dependent manner as can be seen inFIG.10. Almost all the cells undergo apoptotic death with a chimera concentration of 6 μM. HT29 cells were treated with different doses of GRNLY and chimera. The result was that both GRNLY and the chimera are toxic to HT29 cells in a dose-dependent manner as can be seen inFIG.11. GRNLY or chimera concentrations of 4 or 5 μM seem to have a similar effect, but the chimera is more cytolytic than GRNLY at a concentration of 6 μM, achieving a percentage of growth of 30% with respect to the control, i.e., 70% cytotoxicity. To match said effect, a GRNLY concentration of about 20 μM must be used. Furthermore, labeling was also performed with Alexa-46-conjugated annexin-V showing phosphatidylserine exposure and with 7AAD showing membrane integrity on HT29 cells treated with different concentrations of chimera for analyzing the type of induced cell death. By increasing the concentration of chimera, an increase in cells labeled with annexin which still have not lost membrane integrity is observed, indicating that cell death is caused by apoptosis (FIG.12). Furthermore, a significant increase in cytotoxicity is observed when incubating the cells with a chimera concentration of 6 μM, as shown inFIG.16. The maximum dose of chimera used was 6 μM, whereas in the case of GRNLY, a concentration of up to 20 μM was reached. Example 5 In Vivo Assay with HELA-CEA Cells Five mice per group (control group, granulysin group, and MFE group (with the chimera) were assayed. Although there was a mouse in the MFE group that died after the sixth injection, the other 4 mice, however, reached the end of the experiment in good conditions state. The tumor was subcutaneously injected with Matrigel at 2 million cells. Treatments began when the tumors reached a size of 150 mm3. The treatments were systemic intraperitoneal treatments performed every two days (injections):Control group, 500 ul of PBS.Granulysin group, 220 ul of a stock at 500 ug/ml (40 uM), i.e., 110 ug per injection, which yields a concentration of about 5 uM in 2 ml of total blood.MFE group, 500 ul of stocks of about 900 ug/ml (25 uM), i.e., 425 ug per injection, which yields a concentration of about 5 uM in 2 ml of total blood. Ten injections were performed and the mice were sacrificed 2 days after the last injection. The results are illustrated inFIGS.13to19.FIG.13shows that if the control group is compared with MFE group (chimera), significant differences can be seen after the 7thinjection, with the difference being very significant in the last injections. It can be seen how tumor growth in treated mice is somehow contained or attenuated.FIG.14shows that if the control group is compared with the (non-chimeric) granulysin group, there are no significant differences, although the granulysin curve is below the control curve for all the points.FIG.15shows all the results shown inFIG.13andFIG.14.FIG.16shows the means±SD of the sizes of the tumors once removed and subjected to different treatments, a smaller tumor size with granulysin treatment, and an even smaller size when the chimera is used, being shown.FIG.16shows the means±SD of the weights of the tumors once removed and subjected to different treatments, a lower tumor weight with granulysin treatment, and an even lower weight when the chimera is used, being shown.
33,529
11858974
SUMMARY OF THE INVENTION It is an object of the present invention to provide short, drug-like peptide macrocycles with the ability to rescue the misfolding, aggregation and associated pathogenic effects of a prominent aggregation-prone and disease-associated protein target. Particularly, the object of the present invention is to identify cyclic oligopeptides that rescue the misfolding and modulate the natural aggregation process of SOD1 and of its variants, which are implicated in pathogenicity of ALS and fALS. Various tetra- and penta- and hexapeptides with these properties are described in the present invention. More particularly the inventors have identified the general formula cyclo-NuX1X2. . . XN, wherein Nu=C, S, or T; X is any of the twenty natural amino acid and N=3-5 as a very rich source of chemical rescuers of SOD1 misfolding and modulators of its aggregation. Another aspect of the present invention is the identification of pentapeptide macrocycles with the general formula cyclo-TXSXW, wherein the first amino acid is Threonine, the third amino acid is a Serine, the last amino acid is Tryptophan and X is any amino acid, as effective and preferred misfolding rescuers and modulators of the natural process of SOD1. More preferred misfolding rescuers and modulators of SOD1 aggregation are cyclic oligopeptide sequences exhibiting the cyclo-TΨ1SΨ2W motif, where Ψ1=any amino acid excluding isoleucine (I), asparagine (N), glutamine (Q), methionine (M), glutamic acid (E), histidine (H), and lysine (K); and Ψ2=any amino acid excluding isoleucine (I), asparagine (N), glutamine (Q), cysteine (C), aspartic acid (D), glutamic acid (E), lysine (K) and proline (P). Even more preferred misfolding rescuers and modulators of SOD1 aggregation are cyclic oligopeptide sequences exhibiting the cyclo-T(Φ1,S)S(Φ2,M,H)W motif, where Φ1is preferably one of the hydrophobic (Φ) amino acids A, W or F, while Φ2is preferably V, W or F. A small group of three cyclic pentapeptide rescuers with this general formula T(Φ1,S)S(Φ2,M,H)W are analyzed further. In the present invention, isolated cyclic oligopeptides are also provided, which comprise the amino acid sequences set forth in SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, up to SEQ ID NO:46. Nucleic acid sequences encoding a polypeptide of the invention are also provided. Vectors containing such nucleic acids, and cells containing such vectors, are also provided. Another object of the present invention is to provide sufficient evidence that the selected peptide macrocycles are successful in inhibiting the misfolding and aggregation of SOD1. More particularly the inventors have studied the SOD1(A4V), a fALS-associated variant, whose misfolding and aggregation causes a very aggressive form of the disease with an average survival time of only 1.2 years after diagnosis. Indeed the effects of three selected oligopeptide macrocycles in inhibiting the misfolding and aggregation of SOD1 are shown using appropriate biochemical and/or biophysical assays. Another object of the present invention is to provide sufficient evidence that the selected peptide macrocycles are successful in inhibiting the aggregation of SOD1* and the neurotoxicity caused by SOD1* aggregation in vitro. Indeed the effects of one selected pentapeptide macrocycle in inhibiting the aggregation and toxicity of SOD1(A4V) are shown in cultured mammalian cells. Another objective of the present invention is to provide hybrid polypeptides that comprise a peptide motif that specifically interacts with the target polypeptide, which is then inserted into an appropriate protein scaffold. The polypeptide motif is inserted into the scaffold such that any desired function of the scaffold is retained and the inserted motif as presented retains its ability to specifically bind to the target and/or modulate the natural aggregation process of the target polypeptide. The scaffold can include, for example, neuroprotective agents to make SOD1 aggregates less toxic, aggregate-destroying molecules to eliminate amyloid SOD1 species, reagents that prevent SOD1 aggregate formation, or reagents useful for specifically imaging SOD1 aggregates in brain tissue. It is also an object of the present invention to provide an integrated bacterial platform for the discovery of chemical/biological modulators of the problematic folding and aggregation of SOD1. In this system, large combinatorial libraries of genetically encoded macrocycles are biosynthesized inE. colicells and are simultaneously screened for their ability to modulate the problematic folding and aggregation of SOD1 using a high-throughput genetic screen, which is based on the detection of enhanced fluorescence of chimeric SOD1-GFP fusions. It is an object of the present invention to provide short, drug-like peptide macrocycles with the ability to rescue the misfolding, aggregation and associated pathogenic effects of a second prominent aggregation-prone and disease-associated protein target. Particularly, the object of the present invention is to identify cyclic oligopeptides that rescue the misfolding and modulate the natural aggregation process of the β-amyloid peptide, which is implicated in pathogenicity of Alzheimer's disease. Various tetra-, penta- and hexapeptides with these properties are described in the present invention. More particularly the inventors have identified the general formula cyclo-NuX1X2. . . XN, wherein Nu=C, S, or T; X is any of the twenty natural amino acid and N=3-5 as a very rich source of chemical rescuers of Aβ misfolding and modulators of its aggregation. Another aspect of the present invention is the identification of pentapeptide macrocycles with the general formula cyclo-TXXXR, wherein the first amino acid is Threonine, the last amino acid is Arginine and X is any amino acid, as effective and preferred modulators of the natural process of Aβ aggregation. More preferred modulators of Aβ aggregation are cyclic oligopeptide sequences exhibiting the cyclo-TΦZΠR motif, where Φ=any amino acid excluding phenylalanine (F), tryptophan (W), tyrosine (Y), glutamine (Q), aspartic acid (D), glutamic acid (E), lysine (K) and proline (P); Z is any amino acid excluding E; and H is any natural amino acid excluding Q, methionine (M) and lysine (K). Even more preferred are cyclic oligopeptide sequences exhibiting the cyclo-T(T,A,V)Φ(A,D,W)R motif, where T is a non-negatively charged amino acid. A small group of six cyclic pentapeptide rescuers with this general formula cyclo-TXXXR are analyzed further. Another aspect of the present invention is the identification of tetrapeptide macrocycles with the general formula cyclo-TXXR, wherein the first amino acid is Threonine, the last amino acid is Arginine and X is any amino acid, as effective and preferred modulators of the natural process of Aβ aggregation. More preferred modulators of Aβ aggregation are cyclic oligopeptide sequences exhibiting the cyclo-TΘΛR motif, where Θ=T, R, D, L, F or A, and Λ=C, R, S, G, Q, I, W, D, or F. A small group of two cyclic tetrapeptide rescuers with this general formula cyclo-TXXR are analyzed further. Another aspect of the present invention is the identification of hexapeptide macrocycles as effective and preferred modulators of the natural process of Aβ aggregation. Preferred modulators of Aβ aggregation are cyclic oligopeptide sequences exhibiting the cyclo-NuX1X2X3X4X5motif for the use in rescuing Aβ misfolding and modulating aggregation, wherein Nu is T; wherein X1is any amino acid selected from I, L, V, C, S, K or P, and is more preferably P, V or L; wherein X2is selected from A, I, L, V, F, W, C, S, T, D, E, H, K, P, or G and is more preferably V or A; wherein X3is selected from I, L, V, F, W, Y, E or R, and is more preferably W; wherein X4is selected from L, V, F, Y, S or R and is more preferably F; wherein X5is selected from W, M, N, D or E and is more preferably D. The cyclic hexapeptide comprising the sequence TPVWFD is analyzed further. In the present invention, isolated cyclic oligopeptides are also provided, which comprise the amino acid sequences set forth in SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:50, up to SEQ ID NO:205. The inventors also identified the pentapeptide macrocycles cyclo-SASPT (SEQ ID NO:206), cyclo-SHSPT (SEQ ID NO:207), cyclo-SICPT (SEQ ID NO:208) and cyclo-SITPT (SEQ ID NO:209) as effective and preferred modulators of the natural process of Aβ aggregation. Furthermore the inventors also identified the tetrapeptide macrocycles cyclo-TTCR (SEQ ID NO:210), cyclo-TTRR (SEQ ID NO:211), cyclo-TTSR (SEQ ID NO:212), cyclo-TTGR (SEQ ID NO:213), cyclo-TRGR (SEQ ID NO:214), cyclo-TRRR (SEQ ID NO:215), cyclo-TDQR (SEQ ID NO:216), cyclo-TLIR (SEQ ID NO:217), cyclo-TLWR (SEQ ID NO:218), cyclo-TLGR (SEQ ID NO:219), cyclo-TFDR (SEQ ID NO:220), and cyclo-TAFR (SEQ ID NO:221) as effective and preferred modulators of the natural process of Aβ aggregation. Finally, the inventors also identified the hexapeptide macrocycles comprising the amino acid sequences set forth in SEQ ID NO:222, SEQ ID NO:223, . . . , up to SEQ ID NO:255 as effective and preferred modulators of the natural process of Aβ aggregation. Nucleic acid sequences encoding a polypeptide of the invention are also provided. Vectors containing such nucleic acids, and cells containing such vectors, are also provided. Another object of the present invention is to provide sufficient evidence that the selected peptide macrocycles are successful in modulating Aβ aggregation. Indeed the effects of ten selected oligopeptide macrocycles in modulating Aβ aggregation are shown using appropriate biochemical and/or biophysical assays. Another object of the present invention is to provide sufficient evidence that the selected peptide macrocycles are successful in inhibiting the neurotoxicity caused by Aβ aggregation in vitro. Indeed the effects of two selected pentapeptide macrocycles in inhibiting neurotoxicity of Aβ aggregation are shown in cultured primary hippocampal neurons. A main objective of the present invention is to show the successful in vivo rescuing of the misfolding and aggregation of β-amyloid peptide by the selected macrocycles produced by the technique described in the present invention. The protective effects of the selected cyclic peptides against Aβ aggregation and toxicity in vivo are demonstrated in established AD animal models in the nematodeCaenorhabditis elegans(C. elegans). Another objective of the present invention is to provide hybrid polypeptides that comprise a peptide motif that specifically interacts with the target polypeptide, which is then inserted into an appropriate protein scaffold. The polypeptide motif is inserted into the scaffold such that any desired function of the scaffold is retained and the inserted motif as presented retains its ability to specifically bind to the target and/or modulate the natural aggregation process of the target polypeptide. The scaffold can include, for example, neuroprotective agents to make amyloid plaques less toxic, amyloid destroying molecules to eliminate plaques, reagents that prevent amyloid plaque formation, or reagents useful for specifically imaging amyloid plaques in brain tissue. It is also an object of the present invention to provide an integrated bacterial platform for the discovery of chemical/biological modulators of the problematic folding and aggregation of Aβ. In this system, large combinatorial libraries of genetically encoded macrocycles are biosynthesized inE. colicells and are simultaneously screened for their ability to modulate the problematic folding and aggregation of Aβ using a high-throughput genetic screen, which is based on the detection of enhanced fluorescence of chimeric Aβ-GFP fusions. It is also an object of the present invention to provide a generalizable integrated bacterial platform for the discovery of chemical/biological modulators of the problematic folding and aggregation of disease-associated, misfolding-prone proteins (MisPs). In this system, large combinatorial libraries of genetically encoded macrocycles are biosynthesized inE. colicells and are simultaneously screened for their ability to modulate the problematic folding and aggregation of the target MisP using a high-throughput genetic screen, which is based on the detection of enhanced fluorescence of chimeric MisP-GFP fusions. In some embodiments, the present disclosure provides a method of identifying modulators of a misfolding-prone protein associated with a protein misfolding disease, comprising: (A) obtaining a population of transformed bacterial cells that co-express: (a) a nucleic acid encoding a library of peptide macrocycles, operably linked to a promoter and (b) a bipartite nucleic acid comprising a sequence for a gene encoding a misfolding-prone protein associated with a protein misfolding disease (MisP) and a sequence encoding a protein reporter; (B) identifying bacterial cells of step (A) that exhibit enhanced levels of protein reporter activity; and (C) identifying the bioactive peptide macrocycles in the library that modulate MisP misfolding. In some aspects, the protein reporter is a fluorescent protein (FP) reporter, and step (B) comprises identifying bacterial cells that exhibit enhanced levels of MisP-FP fluorescence. In some aspects, the protein reporter is an enzyme. In some aspects, the identification in step (B) comprises selection. In some aspects, step (C) comprises sequencing the nucleic acid of step (Aa). In certain aspects, the nucleic acids of (a) and (b) are encoded on the same vector. In particular aspects, the vector is a plasmid. In aspects of the embodiments, said MisP is selected from β-amyloid peptide, SOD1, tau, α-synuclein, polyglutaminated huntingtin, polyglutaminated ataxin-1, polyglutaminated ataxin-2, polyglutaminated ataxin-3, prion protein, islet amyloid polypeptide (amylin), β2-microglbulin, fragments of immunoglobulin light chain, fragments of immunoglobulin heavy chain, serum amyloid A, ABri peptide, ADan peptide, transthyretin, apolipoprotein A1, gelsolin, transthyretin, lysozyme, phenylalanine hydroxylase, apolipoprotein A-I, calcitonin, prolactin, TDP-43, FUS/TLS; insulin, hemoglobin, al-antitrypsin, p53, or variants thereof. In some aspects, said peptide macrocycle can be a ribosomally synthesized as a head-to-tail cyclic peptide, side-chain-to-tail cyclic peptide, bicyclic peptide, lanthipeptide, linaridin, proteusin, cyanobactin, thiopeptide, bottromycin, microcin, lasso peptide, microviridin, amatoxin, phallotoxin, θ-defensin, orbitide, or cyclotide. In some aspects, the disease is selected from amyotrophic lateral sclerosis, Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease, Creutzfeldt-Jakob disease, cancer, phenylketonuria, type 2 diabetes, senile systemic amyloidosis, familial amyloidotic polyneuropathy, familial amyloid cardiomyopathy, leptomeningeal amyloidosis, systemic amyloidosis, familial British dementia, familial Danish dementia, light chain amyloidosis, heavy chain amyloidosis, serum amyloid A amyloidosis, lysozyme amyloidosis, dialysis-related amyloidosis, ApoAI amyloidosis, Finnish type familial amyloidosis, hereditary cerebral hemorrhage with amyloidosis (Icelandic type), medullary carcinoma of the thyroid, pituitary prolactinoma, injection-localized amyloidosis, frontotemporal dementia, spinocerebellar ataxia 1, spinocerebellar ataxia 2, spinocerebellar ataxia 3, al-antitrypsin deficiency, sickle-cell anemia, or transmissible spongiform encephalopathy. In further aspects, the method comprises recombinantly producing or chemically synthesizing the identified bioactive peptide macrocycle. In some aspects, there is provided a method of treatment, prevention or diagnosis of a protein misfolding disease comprising administering to a subject a therapeutically effective amount of the bioactive peptide macrocycle identified by the embodiments and aspects of the invention provided herein. In another aspect, there is provided a pharmaceutical composition comprising the bioactive peptide macrocycle according to the embodiments or aspects of the invention provided herein, and a pharmaceutically acceptable carrier. In some aspects, the pharmaceutical composition may be used for the treatment or prevention of a protein misfolding disease. In some aspects, there is provided a hybrid molecule comprising: a) a peptide macrocycle identified or produced according to the method of any of the embodiments or aspects of the invention provided herein, and b) a scaffold molecule linked to the peptide macrocycle. In some aspects, the scaffold molecule is a diagnostic or a therapeutic reagent. In particular aspects, the therapeutic reagent is a cytoprotective agent that renders the aggregates of the target protein less toxic or inhibits target protein aggregate formation. In some aspects, the scaffold molecule comprises all or a sufficient portion of a protein selected from the group consisting of antibodies, enzymes, chromogenic proteins, and fluorescent proteins. In specific aspects, the diagnostic reagent specifically targets protein aggregates in diseased or healthy tissue. In some aspects, there is provided a method of treating or diagnosing a protein misfolding disease associated with aberrant aggregate formation, the method comprising administering a hybrid molecule according to the embodiments and aspects provided herein, wherein the peptide macrocycle of the hybrid molecule specifically interacts with the amyloid or non-amyloid form of the target MisP. In some embodiments, there is provided a peptide comprising the amino acid sequence NuX1X2. . . XN, wherein: Nu is T; N=4; X1is any amino acid excluding I, N, Q, M, E, H, and K; X2═S; X3is any amino acid excluding I, N, Q, C, D, E, K and P; and X4=W, wherein the specifically interacts with the amyloid or non-amyloid form of SOD1 and/or mutant SOD1. In some aspects, the X1is A, L, V, F, W, Y, C, S, T, D, R, P or G. In particular aspects, the X1is is S, A, F or W. In some aspects, the X3is A, L, V, F, W, Y, M, S, T, R, H or G. In certain aspects, the X3is V, F, W, M, or H. In specific aspects, the X3is W. In some aspects, Nu is T; N=4; X1is A, L, V, F, W, Y, C, S, T, D, R, P or G; X2═S; X3is A, L, V, F, W, Y, M, S, T, R, H or G; and X4=W. In particular aspects, Nu is T; N=4; X1is S, A, F or W; X2═S; X3is V, F, W, M, or H; and X4=W. In some embodiments, there is provided a peptide comprising the amino acid sequence set forth in any one of SEQ ID NO:1-46, wherein the peptide prevents misfolding and aggregation of SOD1 and/or mutant SOD1. In some aspects, the peptide comprises an amino acid sequence selected from TWSVW (SEQ ID NO: 4), TASFW (SEQ ID NO: 2), and TFSMW (SEQ ID NO: 6). In some aspects, said peptide prevents misfolding and aggregation of SOD1 and/or mutant SOD1. In some aspects, at least one position of the peptide is a D amino acid. In certain aspects, the peptide is a cyclic peptide. In other aspects, the peptide is a linear peptide. In some aspects, there is provided a hybrid molecule comprising: a) a peptide set forth in the embodiments and aspects provided herein, and b) a scaffold molecule. In some aspects, the scaffolding molecule comprises a cell penetrating peptide. In specific aspects, the scaffold molecule comprises a diagnostic or therapeutic reagent. In certain aspects, the scaffold molecule comprises a polypeptide, small molecule or compound. In some aspects, the scaffold molecule comprises all or a sufficient portion of a protein selected from the group consisting of antibodies, enzymes, chromogenic proteins, fluorescent proteins and fragments thereof. In some aspects, the therapeutic agent is a neuroprotective agent that renders SOD1 aggregates less toxic or inhibits SOD1 aggregate formation. In some aspects, the diagnostic reagent specifically images SOD1 aggregates in neuronal tissue. In some aspects of the embodiments provided herein, there is provided a peptide or a molecule to inhibit protein misfolding and aggregation wherein the peptide prevents misfolding and aggregation of SOD1 and/or mutant SOD1. In particular, there is provided a method of treatment, prevention or diagnosis of amyotrophic lateral sclerosis comprising administering to a subject a therapeutically effective amount of a peptide or hybrid molecule according to any one of the embodiments or aspects provided herein. In some aspects, there is provided a pharmaceutical composition comprising a peptide or hybrid molecule according to any of the aspects or embodiments provided herein, and a pharmaceutically acceptable carrier. In some aspects, the pharmaceutical composition is used for the treatment or prevention of amyotrophic lateral sclerosis. In some aspects, there is provided an isolated nucleic acid sequence encoding the peptide of the aspects or embodiments provided herein. In some aspects, there is a vector comprising said nucleic acid sequence. In some aspects, the vector is an expression vector. In certain aspects, there is provided a host cell comprising said vector. In certain aspects, the host cell is a prokaryotic or eukaryotic cell. In some embodiments, there is provided a peptide wherein the peptide comprises the amino acid sequence NuX1X2. . . XN, wherein: (A) Nu=T; N=3; X1is selected from T, R, D, L, F or A; X2is selected from C, R, S, G, Q, I, W, D, or F; and X3=R; (B) Nu=T; N=4; X1is any amino acid excluding F, W, Y, Q, D, E, K and P; X2is any amino acid excluding E; X3is any amino acid excluding Q, M and K; and X4is R; or (C) Nu=T; N=5; X1is I, L, V, C, S, K or P; X2is any amino acid excluding Y, N, Q, M, and R; X3is selected from I, L, V, F, W, Y, E, or R; X4is selected from L, V, F, Y, S or R; and X5is selected from W, M, N, D, or E, wherein the peptide specifically interacts with a monomeric, oligomeric and/or amyloid form of the Aβ peptide. In some aspects, Nu=T; N=3; X1is selected from T, R, D, L, F or A; X2is selected from C, R, S, G, Q, I, W, D, or F; and X3=R. In certain aspects, Nu=T; N=4; X1is any amino acid excluding F, W, Y, Q, D, E, K and P; X2is any amino acid excluding E; X3is any amino acid excluding Q, M and K; and X4is R. In specific aspects, X1is selected from A, I, L, V, N, C, M, S, T, R, H, or G. In particular aspects, X1is T, V or A. In some aspects, X2is selected from A, I, L, V, F, W, Y, N, Q, C, M, S, T, D, R, H, K, P or G. In certain aspects, X2is I, L, V, F, Y or T. In specific aspects, X3is A, I, L, V, F, W, Y, N, C, S, T, D, E, R, H, P or G. In particular aspects, X3is A, W, or D. In some aspects, Nu=T; N=4; X1is selected from A, I, L, V, N, C, M, S, T, R, H, or G; X2is selected from A, I, L, V, F, W, Y, N, Q, C, M, S, T, D, R, H, K, P or G; X3is A, I, L, V, F, W, Y, N, C, S, T, D, E, R, H, P or G; and X4is R. In certain aspects, Nu=T; N=4; X1is T, V or A; X2is I, L, V, F, Y or T; X3is A, W, or D; and X4is R. In specific aspects, Nu=T; N=5; X1is I, L, V, C, S, K or P; X2is any amino acid excluding Y, N, Q, M, and R; X3is selected from I, L, V, F, W, Y, E, or R; X4is selected from L, V, F, Y, S or R; and X5is selected from W, M, N, D, or E. In particular aspects, X1is selected from I, L, V, C, S, K or P. In specific aspects, the X1is P, V or L. In some aspects, X2is selected from A, I, L, V, F, W, C, S, T, D, E, H, K, P, or G. In certain aspects, X2is V or A. In some aspects, X3is selected from I, L, V, F, W, Y, E or R. In specific aspects, X3is W. In some aspects, X4is selected from L, V, F, Y, S or R. In specific aspects, X4is F. In some aspects, X5is selected from W, M, N, D, or E. In specific aspects, X5is D. In some aspects, Nu=T; N=5; X1is P, V or L; X2is V or A; X3is selected from I, F, or W; X4is selected from L, F, or R; and X5is selected from W, N, or D. In certain aspects, Nu=T; N=5; X1is P, V or L; X2is V or A; X3is W; X4is F; and X5is D. In some aspects, the peptide comprises the sequence TTCR, TTRR, TTSR, TRGR, TTGR, TRRR, TDQR, TLIR, TLWR, TLGR, TFDR, or TAFR (SEQ ID NOs 210-221). In some embodiments, there is provided a peptide comprising the amino acid sequence set forth in any one of SEQ ID NO:47-209, wherein, the peptide is cyclic and specifically interacts with a monomeric, oligomeric and/or amyloid form of the Aβ peptide. In some aspects, the peptide comprises an amino acid sequence selected from TAFDR (SEQ ID NO: 86), TAWCR (SEQ ID NO: 63), TTWCR (SEQ ID NO: 60), TTVDR (SEQ ID NO: 48), TTYAR (SEQ ID NO: 47), TTTAR (SEQ ID NO: 56) or SASPT (SEQ ID NO: 206). In some aspects, the the peptide comprises an amino acid sequence selected from the amino acid sequences set forth in SEQ ID NO:176-209. In some aspects, the peptide comprises an amino acid sequence of TPVWFD (SEQ ID NO:176) or TPAWFD (SEQ ID NO:177). In some aspects, at least one position of the peptide is a D amino acid. In some aspects, the peptide is a cyclic peptide. In other aspects, the peptide is a linear peptide. In some aspects, there is provided a hybrid molecule comprising: a) a peptide set forth in any one of the embodiments or aspects, that specifically interacts with a monomeric, oligomeric and/or amyloid form of the Aβ peptide; and b) a scaffold molecule. In some aspects, the scaffolding molecule comprises a cell penetrating peptide. In certain aspects, the scaffold molecule comprises a diagnostic or therapeutic reagent. In particular aspects, the scaffold molecule comprises a polypeptide, small molecule or compound. In specific aspects, the polypeptide comprises all or a sufficient portion of a protein selected from the group consisting of antibodies, enzymes, chromogenic proteins, or a fluorescent protein. In some aspects, the therapeutic agent is a neuroprotective agent that renders amyloid plaques less toxic or inhibits plaque formation. In some aspects, the diagnostic reagent specifically images oligomers and/or amyloid aggregates in neuronal tissue. In some aspects of the invention, there is provided methods for the use of a peptide or molecule according to any of the embodiments or aspects provided herein, to inhibit protein misfolding and aggregation. In some aspects, said peptide prevents misfolding and aggregation of the β-amyloid peptide. In some embodiments, there is provided a method of treatment, prevention or diagnosis of a disease related to protein misfolding and aggregation, comprising administering to a subject a therapeutically effective amount of a peptide or molecule according to any of the embodiments or aspects provided herein, wherein the disease is selected from Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, Creutzfeldt-Jakob disease, type 2 diabetes, familial amyloidotic polyneuropathy, systemic amyloidosis, and transmissible spongiform encephalopathy. In some aspects, there is provided a method of treatment, prevention or diagnosis of Alzheimer's disease comprising administering to a subject a therapeutically effective amount of a peptide according to any one of the aspects or embodiments provided herein. In some aspects, there is provided a pharmaceutical composition comprising a peptide according to any one of the aspects or embodiments provided herein and a pharmaceutically acceptable carrier. In some aspects, there is provided a nucleic acid encoding any of said peptides. In some aspects, there is a provided a vector comprising said nucleic acid. In some aspects, the vector is an expression vector. In some aspects, there is provided a host cell comprising said vector. In some cases, the host cell is a prokaryotic or eukaryotic cell. Other objects and advantages of the present invention will become apparent to those skilled in the art in view of the following detailed description. DETAILED DESCRIPTION OF THE INVENTION I. General It is now well established that fALS-linked amino acid substitutions in SOD1 introduce a toxic-gain-of-function property in SOD1 by causing protein misfolding and aggregation, and the formation of oligomeric/aggregated SOD1 species, which are highly toxic for motor neurons. Gradual accumulation of such toxic oligomers/aggregates of mutated SOD1 initiates motor neuron degeneration and the development of fALS. Also, the accumulation of misfolded and aggregated wild-type SOD1 has been implicated in sporadic forms of ALS. This invention pertains to compounds, and pharmaceutical compositions thereof, that can modulate the aggregation of amyloidogenic proteins and peptides, in particular compounds that can rescue the misfolding and inhibit the aggregation of SOD1 or of its fALS-associated variants and inhibit the neurotoxicity of these aggregated SOD1 species. A compound of the invention that modulates aggregation of SOD1, referred to herein interchangeably as a SOD1 modulator compound, a SOD1 modulator or simply a modulator, alters the aggregation of SOD1 or of its fALS-associated variants when the modulator is contacted with SOD1 or of its fALS-associated variants. Thus, a compound of the invention acts to alter the natural aggregation process or rate of SOD1 or of its fALS-associated variants, thereby disrupting the normal course this process. A modulator which inhibits SOD1 and/or mutant SOD1 aggregation (an “inhibitory modulator compound”) can be used to prevent or delay the onset of the deposition of SOD1 and/or mutant SOD1 aggregates. Moreover, inhibitory modulator compounds of the invention inhibit the formation and/or activity of neurotoxic aggregates of SOD1 or of its fALS-associated variants (i.e., the inhibitory compounds can be used to inhibit the neurotoxicity of SOD1 or of its fALS-associated variants). Alternatively, in another embodiment, a modulator compound of the invention promotes the aggregation of SOD1 and/or mutant SOD1. The various forms of the term “promotion” refer to an increase in the amount and/or rate of SOD1 and/or mutant SOD1 aggregation in the presence of the modulator, as compared to the amount and/or rate of SOD1 and/or mutant SOD1 aggregation in the absence of the modulator. Such a compound which promotes SOD1 and/or mutant SOD1 aggregation is referred to as a stimulatory modulator compound. Stimulatory modulator compounds may be useful, for example, in decreasing the amounts of neurotoxic SOD1 and/or mutant SOD1 oligomeric species by driving the natural SOD1 aggregation process towards the (possibly) less neurotoxic higher-order SOD1 and/or mutant SOD1 aggregates. Compounds of the present invention may inhibit SOD1 and/or mutant SOD1 aggregation and/or oligomerization. In particular, preferred modulator compounds of the invention comprise cyclic oligopeptides with the general formula cyclo-NuX1X2. . . XN, where X is any one of the twenty natural amino acids, N=3-5 and Nu=cysteine (Cys or C), serine (Ser or S) or threonine (Thr or T), which is sufficient to alter (and preferably inhibit) the natural aggregation process or rate of SOD1 and/or mutant SOD1. This SOD1 and/or mutant SOD1 modulator can comprise as few as four amino acid residues (or derivative, analogues or mimetics thereof). According to the prevalent amyloid cascade hypothesis, the high tendency of Aβ for misfolding and aggregation results in the formation of neurotoxic oligomers/aggregates, whose accumulation ultimately leads to neuron degeneration and the development of the disease. This invention pertains to compounds, and pharmaceutical compositions thereof, that can modulate the aggregation of amyloidogenic proteins and peptides, in particular compounds that can modulate the aggregation of the β-amyloid peptide (Aβ) and inhibit the neurotoxicity of Aβ. A compound of the invention that modulates aggregation of Aβ, referred to herein interchangeably as a Aβ modulator compound, a Aβ modulator or simply a modulator, alters the aggregation of natural Aβ when the modulator is contacted with natural Aβ. Thus, a compound of the invention acts to alter the natural aggregation process or rate of Aβ, thereby disrupting the normal course this process. A modulator which inhibits Aβ aggregation (an “inhibitory modulator compound”) can be used to prevent or delay the onset of β-amyloid deposition. Moreover, inhibitory modulator compounds of the invention inhibit the formation and/or activity of neurotoxic aggregates of natural Aβ peptide (i.e., the inhibitory compounds can be used to inhibit the neurotoxicity of Aβ. Alternatively, in another embodiment, a modulator compound of the invention promotes the aggregation of natural Aβ peptides. The various forms of the term “promotion” refer to an increase in the amount and/or rate of Aβ aggregation in the presence of the modulator, as compared to the amount and/or rate of Aβ aggregation in the absence of the modulator. Such a compound which promotes Aβ aggregation is referred to as a stimulatory modulator compound. Stimulatory modulator compounds may be useful, for example, in decreasing the amounts of neurotoxic oligomeric species by driving the natural Aβ aggregation process towards the (generally) less neurotoxic higher-order Aβ aggregates. Compounds of the present invention may inhibit Aβ aggregation and/or oligomerization. In particular, preferred modulator compounds of the invention comprise cyclic oligopeptides with the general formula cyclo-NuX1X2. . . XN, where X is any one of the twenty natural amino acids, N=3-5 and Nu=cysteine (Cys or C), serine (Ser or S) or threonine (Thr or T), which is sufficient to alter (and preferably inhibit) the natural aggregation process or rate Aβ. This Aβ modulator can comprise as few as four amino acid residues (or derivative, analogues or mimetics thereof). II. Discovery of Peptide SOD1 and Aβ Modulators The present application describes the invention of a generalizable bacterial platform for the discovery of macrocyclic peptide rescuers of the misfolding of disease-associated, misfolding-prone proteins (MisPs). The inventors demonstrate the generalizability of this integrated bacterial platform by discovering macrocyclic peptides that modulate the problematic folding and aggregation of SOD1, mutant SOD1, or Aβ. This approach offers a number of important advantages. First, it allows the screening of molecular libraries with expanded diversities. Here, a library with diversity>10 million different macrocycles has been investigated. In another embodiment, the present invention can be applied to construct and screen macrocyclic libraries with diversities up to 1010 different molecules. Importantly,E. colican support the in vivo biosynthesis not only of head-to-tail cyclic peptides like the ones investigated here, but also of other macrocyclic structures, such as side-chain-to-tail cyclic peptides, bicyclic peptides, cyclotides, macrolides and other macrocyclic structures, which can accommodate not only naturally occurring amino acids, but a large variety of artificial ones as well. In addition, the analysis of these large libraries is carried out using a very high-throughput genetic screen, which enables the identification of bioactive molecules simply by isolating compounds that enhance the fluorescence ofE. colicells expressing MisP-GFP, such as SOD1*-GFP or Aβ-GFP fusions, by flow cytometric sorting (FACS). Compared to affinity-based approaches for screening DNA-encoded chemical libraries, such as phage and mRNA display, the herein described approach does not detect mere target MisP, such as SOD1* or Aβ, binding, but selects directly the bioactive compounds with the ability to rescue MisP, such as SOD1* or Aβ, misfolding, without requiring the availability of purified MisP, such as SOD1* or Aγ3. Moreover, synthesis of the studied compounds and their screening for bioactivity are carried out in vivo as part of a single-step process, without the need for laborious organic synthesis and product isolation steps. Importantly, screening for bioactivity is carried out in a fully unbiased manner without requiring a priori knowledge of the structures of the MisP, such as SOD1* or Aβ, monomers, oligomers, or higher-order aggregates, specific assumptions about possible binding sites, or prior preparation of specific MisP, such as SOD1* or Aβ, oligomerization states. More particularly, combinatorial libraries of random cyclic tetra-, penta-, and hexapeptides have been created and the most prominent targets have been selected to study as potential rescuers of SOD1 and Aβ misfolding. The technology described in the present invention utilizes a technique termed split intein circular ligation of peptides and proteins (SICLOPPS) for producing peptide libraries inE. coli. SICLOPPS uses split inteins, i.e. self-splicing protein elements for performing N- to C-terminal peptide cyclization and biosynthesize cyclic peptides as short as four amino acids long. The only requirement for the intein splicing reaction and peptide cyclization to occur is the presence of a nucleophilic amino acids cysteine (C), serine (S), or threonine (T) as the first amino acid of the extein following the C-terminus of the intein. According to the present invention, peptides belonging to the general formula NuX1X2. . . XN, can be used for rescuing protein misfolding and modulating protein aggregation; wherein X is any one of the twenty natural amino acids, N=3-5 and Nu is selected from cysteine (Cys or C), serine (Ser or S) or threonine (Thr or T). According to the preferred embodiment of the present invention the peptide is a cyclic peptide. According to the preferred embodiment of the present invention Nu is T. In a preferred embodiment of the present invention the peptide with the general formula cyclo-NuX1X2. . . XN, for the use in protein misfolding and aggregation, has the following specifications wherein N is 3, wherein Nu is T; wherein X1is selected from T, R, D, L, F or A, wherein X2is selected form C, R, S, G, Q, I, W, D, or F; wherein X3is R. According to the above specification the preferred cyclic tetrapeptide is selected from cyclo-TTCR (SEQ ID NO:164), cyclo-TTRR (SEQ ID NO:165), cyclo-TTSR (SEQ ID NO:166), cyclo-TTGR (SEQ ID NO:167), cyclo-TRGR (SEQ ID NO:168), cyclo-TRRR (SEQ ID NO:169), cyclo-TDQR (SEQ ID NO:167), cyclo-TLIR (SEQ ID NO:171), cyclo-TLWR (SEQ ID NO:172), cyclo-TLGR (SEQ ID NO:173), cyclo-TFDR (SEQ ID NO:174), and cyclo-TAFR (SEQ ID NO:175) as effective and preferred modulators of the natural process of Aβ aggregation. In another preferred embodiment of the present invention the peptide with the general formula NuX1X2. . . XN, for the use in rescuing protein misfolding and modulating, has the following specifications wherein N is 4, wherein Nu is preferably T; wherein X1is any amino acid excluding I, N, Q, M, E, H, and K, and more preferably it is S, A, W, or F; wherein X2is preferably S; wherein X3is any amino acid excluding I, N, Q, C, D, E, K and P, and is more preferably selected from V, W, F, M, or H; wherein X4is preferably W. According to the above specification the preferred pentapeptide is selected from the amino acid sequences set forth in SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, . . . , up to SEQ ID NO:46. According to the above specification the more preferred pentapeptide is selected from cyclo-TASFW (SEQ ID NO:2), cyclo-TWSVW (SEQ ID NO:4), and cyclo-TFSMW (SEQ ID NO:6). In another preferred embodiment of the present invention the peptide with the general formula NuX1X2. . . XN, for the use in rescuing protein misfolding and modulating aggregation, has the following specifications wherein N is 4, wherein Nu is T or S; wherein X1is any amino acid excluding F, W, Y, Q, D, E, K and P, preferably it is S, H, T, V or A, and more preferably it is T, V or A; wherein X2is any amino acid excluding E, preferably a non-negatively charged amino acid, and more preferably it is selected from I, L, V, F, W, Y, M, S, T, R, H, or G; wherein X3is any amino acid excluding Q, M and K, and is more preferably selected from A, V, F, W, C, S, T, D, C, R, H, P or G; wherein X4is preferably R or T. According to the above specification the preferred pentapeptide is selected from the amino acid sequences set forth in SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:50, . . . , up to SEQ ID NO: 205. According to the above specification the more preferred pentapeptide is selected from cyclo-TAFDR (SEQ ID NO:86), cyclo-TAWCR (SEQ ID NO:63), cyclo-TTWCR (SEQ ID NO:60), cyclo-TTVDR (SEQ ID NO:48), cyclo-TTYAR (SEQ ID NO:47), cyclo-TTTAR (SEQ ID NO:56), and cyclo-SASPT (SEQ ID NO:206). In another preferred embodiment of the present invention the peptide with the general formula NuX1X2. . . XN, for the use in rescuing protein misfolding and modulating aggregation, has the following specifications wherein N is 5, wherein Nu is T; wherein X1is any amino acid selected from I, L, V, C, S, K or P, and is more preferably P, V or L; wherein X2is selected from A, I, L, V, F, W, C, S, T, D, E, H, K, P, or G and is more preferably V or A; wherein X3is selected from I, L, V, F, W, Y, E or R, and is more preferably W; wherein X4is selected from L, V, F, Y, S or R and is more preferably F; wherein X5is selected from W, M, N, D or E and is more preferably D. The hexapeptide according to the above specifications is selected from the amino acid sequences set forth in SEQ ID NO:222, SEQ ID NO:223, . . . , up to SEQ ID NO:255, and is most preferably TPVWFD (SEQ ID NO:222) or TPAWFD (SEQ ID NO:223). The maximum theoretical diversity of the combined cyclo-NuX1X2X3-X5library investigated here was >10 million different sequences. The libraries of genes encoding this combinatorial library of random cyclic oligopeptides were constructed using degenerate codons. The inventors constructed the high diversity pSICLOPPS-NuX1X2X3-X5vector library which is expected to be encoding the vast majority of the theoretically possible designed cyclic tetra-, penta-, and hexapeptide cyclo-NuX1X2X3-X5sequences using molecular biology techniques already known and used in the art. The invention provided herein can be used as a method of treatment, prevention or diagnosis of all diseases related to protein misfolding and aggregation, including but not limited to amyotrophic lateral sclerosis (ALS), Alzheimer's disease, Parkinson's disease, Huntington's disease, Creutzfeldt-Jakob disease, type 2 diabetes, familial amyloidotic polyneuropathy, systemic amyloidosis, and transmissible spongiform encephalopathy comprising administering to a subject a therapeutically effective amount of a peptide. Preferably the invention presented herein can be used as a method of treatment, prevention or diagnosis of amyotrophic lateral sclerosis or Alzheimer's disease. To identify cyclic oligopeptide sequences with the ability to interfere with the problematic folding of Aβ and modulate its oligomerization/aggregation, a bacterial high-throughput genetic screen was utilized. This system monitors Aβ misfolding and aggregation by measuring the fluorescence ofE. colicells overexpressing a chimeric fusion of the human Aβ42with GFP. It has been demonstrated previously that due to the high aggregation propensity of Aβ,E. colicells overexpressing Aβ-GFP fusions produce misfolded fusion protein that accumulates into insoluble inclusion bodies that lack fluorescence, despite the fact that they express these fusions at high levels. Mutations in the coding sequence of Aβ or the addition of compounds that inhibit Aβ aggregation, however, result in the formation of soluble and fluorescent Aβ-GFP, and bacterial cells expressing Aβ-GFP under these conditions acquire a fluorescent phenotype. The inventors of the present invention adapted this system to perform screening for aggregation-inhibitory macrocycles in a very high-throughput fashion by isolating cyclic oligopeptide-producing bacterial clones that exhibit enhanced levels of Aβ42-GFP fluorescence using fluorescence-activated cell sorting (FACS). Herein, the inventors describe that the integrated bacterial platform for the discovery of macrocyclic rescuers that modulate the problematic folding and aggregation of Aβ as described in the present invention, is also generalizable, i.e., it can be more generally applied for the discovery of macrocyclic peptide rescuers of the misfolding of other disease-associated, misfolding-prone proteins (MisPs) as well. To demonstrate this generalizability, the inventors have used the same system to discover macrocyclic peptides that modulate the problematic folding and aggregation of SOD1 and/or mutant SOD1. It has been demonstrated previously that the fluorescence ofE. colicells expressing a recombinant protein whose C terminus is fused to GFP correlates well with the amount of soluble and folded protein (Waldo G S, Standish B M, Berendzen J, Terwilliger T C. Nat Biotechnol. 1999 July; 17(7):691-5). Based on this, it was reasoned that the fluorescence of MisP-GFP fusions can serve as a reliable reporter for the identification of chemical rescuers of MisP misfolding for a number of disease-associated MisPs, including SOD1. In order to test this hypothesis, the inventors generated fusions of SOD1 variants, whose misfolding and aggregation have been linked with the pathology of familial forms of ALS (fALS), with GFP. Expression of these fusions inE. coli, yielded levels of cellular fluorescence, which were significantly decreased compared to that of the generally non-pathogenic, wild-type SOD1. Western blot analysis indicated that this occurs because the accumulation of soluble SOD1-GFP is decreased in the presence of misfolding-inducing amino acid substitutions, which in turn takes place due to enhanced misfolding/aggregation of fusion-free SOD1. Thus, as in the case of Aβ, the fluorescence ofE. colicells overexpressing SOD1-GFP fusions appears to be a good indicator of SOD1 folding and misfolding. To identify rescuers of disease-associated SOD1 misfolding, the inventors screened for cyclic oligopeptides that inhibit the aggregation of SOD1(A4V), a fALS-associated variant, whose misfolding and aggregation causes a very aggressive form of the disease with an average survival time of only 1.2 years after diagnosis. E. coliBL21(DE3) cells producing the combined cyclo-NuX1X2X3-X5library, while simultaneously overexpressing the SOD1(A4V)-GFP reporter, were subjected to FACS sorting for the isolation of clones exhibiting enhanced SOD1(A4V)-GFP fluorescence. This selection yielded anE. colipopulation with ˜10-fold increased fluorescence after four rounds of sorting. Among twenty individual clones tested, four exhibited the highest levels of SOD1(A4V)-GFP fluorescence compared to cells expressing the same SOD1(A4V)-GFP fusion in the presence of random cyclic peptide sequences picked from the initial (unselected) cyclo-NuX1X2X3-X5library and were selected for further analyses. Furthermore, the observed phenotypic effects were dependent on the ability of the Ssp DnaE intein (utilized for peptide cyclization as part of SICLOPPS) to perform protein splicing, as the double amino acid substitution H24L/F26A in the C-terminal half of the Ssp DnaE intein, which is known to abolish asparagine cyclization at the IC/extein junction, and prevent extein splicing and peptide cyclization, was found to reduce SOD1(A4V)-GFP fluorescence back to wild-type levels. Finally, the observed increases in fluorescence were found to be SOD1-specific, as the isolated pSICLOPPS-NuX1X2X3-X5vectors from these selected clones did not enhance the levels of cellular green fluorescence when the sequence of SOD1(A4V) in the SOD1(A4V)-GFP reporter was replaced with that of the human β-amyloid peptide (Aβ). On the contrary, the selected pSICLOPPS-NuX1X2X3-X5vectors were efficient in enhancing the fluorescence of SOD1-GFP containing wild-type SOD1, as well as three additional SOD1 variants, SOD1(G37R), SOD1(G85R), and SOD1(G93A), all of which are associated with familial forms of ALS. Western blot analysis indicated that this enhanced SOD1(A4V)-GFP fluorescence phenotype occurs due to accumulation of enhanced amounts of soluble SOD1(A4V) in these clones. The inventors further analyzed the selected peptides by DNA sequencing of the peptide-encoding regions of the four selected clones. This revealed three distinct putative SOD1(A4V) misfolding-rescuing and aggregation-inhibitory cyclic peptide sequences, all of which encoded cyclic pentapeptides with sequences TASFW (SEQ ID NO: 2), TWSVW (SEQ ID NO: 4), and TFSMW (SEQ ID NO: 6), thus indicating a dominant TXSXW bioactive motif. Interestingly, the Ser residue at position 3, encountered among all selected pentapeptides, was encoded by two different codons in the selected pSICLOPPS plasmids, thus suggesting that its dominance among the isolated clones was not coincidental. As depicted in Examples 4 and 5, the inventors chose the peptide cyclo-TWSVW (SEQ ID NO: 4) for further analysis. This cyclic pentapeptide is hereafter referred to as SOD1C5-4 and was produced in mg quantities by solid-phase synthesis. Isolated SOD1(A4V) was utilized to assess the effect of the selected cyclic pentapeptide SOD1C5-4 on its aggregation process. CD spectroscopy indicated that SOD1C5-4—but not the Aβ-targeting cyclic peptides AβC5-34 or AβC5-116—interacts with SOD1(A4V), and that the time-dependent conformational transition that is indicative of SOD1(A4V) aggregation is significantly delayed in the presence of SOD1C5-4. Moreover, analysis by dynamic light scattering (DLS) revealed that SOD1C5-4 addition results in the time-dependent formation of oligomeric/aggregated SOD1(A4V) species with markedly smaller sizes. Detection of large, amyloid-like SOD1(A4V) aggregates by ThT staining and a filter retardation assay indicated that the formation of such species was dramatically decreased in the presence of SOD1C5-4. Finally, staining of SOD1(A4V) with the conformation-sensitive dye SYPRO Orange under heat-induced denaturation conditions, suggested that the aggregation-inhibitory action of SOD1C5-4 may be occurring due to its ability to decrease the propensity of SOD1(A4V) to expose hydrophobic surfaces, a feature which has been proposed to be a molecular determinant of the pathogenesis of fALS-associated SOD1 variants. Taken together, these results demonstrate that SOD1C5-4 is an efficient and specific rescuer of SOD1(A4V) misfolding and aggregation. The protective effects of SOD1C5-4 in mammalian cells were evaluated in human embryonic kidney 293 (HEK293) cells transiently expressing SOD1(A4V)-GFP. Cells treated with SOD1C5-4 exhibited higher fluorescence, fewer inclusions comprising aggregated SOD1(A4V)-GFP, and higher viability compared to untreated cells. To identify all bioactive cyclic oligopeptide SOD1 modulators contained in the tested cyclo-NuX1X2X3-X5library and to facilitate structure-activity analyses of the isolated sequences, To determine structure-activity relationships for the identified mutant SOD1-targeting cyclic oligopeptides, the sequences of the peptide-encoding regions from ˜5.3 million clones selected after the fourth round of FACS sorting were determined by deep sequencing. 367 distinct oligopeptide sequences appeared more than 50 times among the selected clones and were selected for subsequent analysis, which revealed the following. First, pentapeptides were the dominant peptide species within the sorted pool, with 197 of the distinct oligopeptide sequences selected corresponding to pentapeptides (54%), 148 to hexapeptides (40%) and 22 corresponding to tetrapeptides (6%). Second, the vast majority of the selected peptides exhibited the cyclo-TXSXW motif of SOD1C5-4 (˜92% of all selected clones and ˜97% of the selected pentapeptide-encoding clones. Third, among the selected cyclo-TXSXW pentapeptides, I, N, Q, M, E, H, and K residues were excluded at position 2, and were preferably S, A, W or F. At position 4, I, N, Q, C, D, E, K and P residues were excluded, and were preferably V, W, F, M, or H. Taken together, these results indicate that the most bioactive macrocyclic structures against SOD1(A4V) misfolding and aggregation in the library are cyclic pentapeptides of the cyclo-T(Φ1,S)S(Φ2,M,H)W motif, where Φ1is preferably one of the hydrophobic (Φ) amino acids A, W or F, while Φ2is preferably V, W or F. E. coliBL21(DE3) cells producing the combined cyclo-NuX1X2X3-X5library, while simultaneously overexpressing the Aβ42-GFP reporter, were subjected to FACS sorting for the isolation of clones exhibiting enhanced Aβ42-GFP fluorescence. Increase in the mean fluorescence was measured and ten random clones were picked from the sorted population. Aβ42-GFP fluorescence of the isolated peptide-expressing clones was found to be dramatically increased compared to cells expressing the same Aβ42-GFP fusion in the presence of random cyclic peptide sequences picked from the initial (unselected) cyclo-NuX1X2X3-X5library. Furthermore, the observed phenotypic effects were dependent on the ability of the Ssp DnaE intein (utilized for peptide cyclization as part of SICLOPPS) to perform protein splicing, as the double amino acid substitution H24L/F26A in the C-terminal half of the Ssp DnaE intein, which is known to abolish asparagine cyclization at the IC/extein junction, and prevent extein splicing and peptide cyclization, was found to reduce Aβ42-GFP fluorescence back to wild-type levels. Finally, the observed increases in fluorescence were found to be Aβ-specific, as the isolated pSICLOPPS-NuX1X2X3-X5vectors from these selected clones did not enhance the levels of cellular green fluorescence when the sequence of Aβ in the Aβ42-GFP reporter was replaced with that of each one of two unrelated disease-associated MisPs, the DNA-binding (core) domain of the human p53 containing a Tyr220Cys substitution (p53C(Y220C)) and an Ala4Val substitution of human Cu/Zn superoxide dismutase 1 (SOD1(A4V)). On the contrary, the selected pSICLOPPS-NuX1X2X3-X5vectors were efficient in enhancing the fluorescence of Aβ-GFP containing two additional Aβ variants, Aβ40and the E22G (arctic) variant of Aβ42, which is associated with familial forms of AD. Analysis of the expressed Aβ42-GFP fusions by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting revealed that the bacterial clones expressing the selected cyclic peptides produce markedly increased levels of soluble Aβ42-GFP compared to random cyclic peptide sequences. Furthermore, when the same cell lysates were analyzed by native PAGE and western blotting, it was observed that co-expression of the selected cyclic peptides reduced the accumulation of higher-order Aβ42-GFP aggregates, which could not enter the gel, and increased the abundance of species with higher electrophoretic mobility. The inventors further analyzed the selected peptides by DNA sequencing of the peptide-encoding regions of ten isolated clones. This revealed eight distinct putative Aβ aggregation-inhibitory cyclic peptide sequences: one corresponded to a hexapeptide (TPVWFD (SEQ ID NO: 222); present twice among the sequenced clones) and seven pentapeptides (TAFDR (SEQ ID NO: 86), TAWCR (SEQ ID NO: 63), TTWCR (SEQ ID NO: 60), TTVDR (SEQ ID NO: 48), TTYAR (SEQ ID NO: 47; present twice), TTTAR (SEQ ID NO: 56), and SASPT (SEQ ID NO: 206)). Interestingly, the Arg residue at position 5, frequently encountered among the selected pentapeptides, was encoded by three different codons in the selected pSICLOPPS plasmids, thus suggesting that its dominance among the isolated clones was not coincidental. As depicted in Examples 7-10, the inventors chose two cyclic oligopeptide sequences for further analysis. These were cyclo-TAFDR (SEQ ID NO: 86) and cyclo-SASPT (SEQ ID NO: 206), hereafter referred to as AβC5-116 and AβC5-34 (Aft-targeting cyclic 5-peptide number 116 and 34), and were produced by solid-phase synthesis in mg quantities. To further analyze the results the inventors of the present invention chose to focus on pentapeptides, as this was the type of peptide most frequently present among the ones selected from the genetic screen. The inventors decided to further study the sequence AβC5-116 since the TXXXR motif was particularly dominant among the selected pentapeptides, while AβC5-34 was chosen because it was the only selected pentapeptide whose sequence appeared to deviate from this motif. Circular dichroism (CD) spectroscopy was first used to assess the effect of the selected pentapeptides on the aggregation process of Aβ40and Aβ42. Addition of AβC5-116 was found to strongly inhibit the aggregation of Aβ40, which remained at a random coil conformation for extended periods of time. The addition of AβC5-34 did not have the same effect and resulted in the appearance of a low-intensity negative peak. When the same solutions were subjected to a ThT dye-binding assay detecting amyloid fibrils, Aβ40fibril formation was reduced in the presence of AβC5-116, while it remained unaffected by AβC5-34. In the case of Aβ42, both selected cyclic pentapeptides affected its normal aggregation pathway strongly and stabilized β-sheet-like structures. ThT staining of the same samples revealed that the extent of amyloid fibril formation was greatly reduced in both cases. When the cyclic peptides were added at a higher ratio, similar CD patterns were observed, however the negative peaks were much more pronounced and fibril formation was completely prevented. The addition of the control cyclic pentapeptide SOD1C5-4 targeting another protein and of randomly selected cyclic control peptides did not have any effect on Aβ40and Aβ42aggregation. Finally, the inventors also performed Transmission electron microscopy (TEM) to verify the above findings. Taken together, these results demonstrate that the selected cyclic oligopeptides interfere with the normal aggregation process of Aβ. The protective effects of AβC5-34 and AβC5-116 on Aβ40- and Aβ42-induced toxicity were evaluated in primary mouse hippocampal neurons and in glioblastoma cell lines. The addition of AβC5-34 and AβC5-116 was found to markedly inhibit the neurotoxicity of both Aβ40and Aβ42in a dose-responsive manner. The inventors also studied the effect of AβC5-34 and AβC5-116 on the morphology of Aβ-exposed neuronal cells by phase-contrast microscopy. In the presence of pre-aggregated Aβ, the population of attached cells was greatly reduced compared to the control, with many detached rounded-up cells floating in the supernatant, while hallmarks of degenerating neurons, such as cell shrinkage, membrane blebbings, fragmented neurites and ill-developed axons were obvious in the preparations. This phenotype was reversed with the addition of the selected cyclic peptides. To further evaluate the protective effects of the selected cyclic peptides against Aβ aggregation and toxicity in vivo, the inventors employed three established models of AD in the nematode wormCaenorhabditis elegans. The conservation of genetic and metabolic pathways betweenC. elegansand mammals, in combination with its completely characterized nervous and muscular system, its easy visualization and simple manipulation, has nominatedC. elegansas an excellent model for neurodegenerative diseases including AD, while chemical screening against Aβ-induced toxicity inC. elegansis increasingly used in AD drug discovery. A paralysis assay was performed in theC. elegansstrain CL4176, where human Aβ42is expressed in the animals' body wall muscle cells under the control of a heat-inducible promoter and Aβ aggregate formation is accompanied by the emergence of a paralysis phenotype. When chemically synthesized AβC5-34 (10 μM) and AβC5-116 (5 μM) were supplied to CL4176 worms, the emergence of the characteristic paralysis phenotype upon temperature up-shift was significantly decelerated compared to the untreated animals. The strain CL2331, which expresses a Aβ(3-42)-GFP fusion again in its body wall muscle cells upon temperature up-shift was also used, and treatment with either one of the selected peptides resulted in a significant reduction of Aβ deposits, which was further shown with biochemical analysis of the accumulation levels of both total and oligomeric Aβ levels in CL4176 animals. To identify the functionally important residues within the isolated peptides, the inventors performed position 1 substitutions with the other two nucleophilic amino acids present in the initial libraries, as well as alanine scanning mutagenesis at positions 3-5 of the AβC5-34 and AβC5-116 pentapeptides. As judged by the ability of the generated variants to enhance the fluorescence ofE. colicells overexpressing Aβ42-GFP, AβC5-116 was found to be much more tolerant to substitutions compared to AβC5-34. All tested sequence alterations within AβC5-34, apart from the S1T substitution, were found to be deleterious for its Aβ aggregation-inhibitory. On the contrary, only the initial Thr and the ultimate Arg were found to be absolutely necessary for the bioactivity of AβC5-116, whereas residues at positions 3 and 4 could be substituted by Ala without significant loss of activity. To identify all bioactive cyclic oligopeptide Aβ modulators contained in the tested cyclo-NuX1X2X3-X5library and to facilitate structure-activity analyses of the isolated sequences, the peptide sequences isolated from the genetic screen were analyzed by next-generation sequencing. This analysis revealed the following. First, pentapeptides were the dominant peptide species within the sorted pool. Second, the most prevalent motif among the selected pentapeptide sequences were cyclo-TXXXR pentapeptides (˜47% of the selected pentapeptide-encoding pSICLOPPS plasmids; ˜42% of the unique selected pentapeptide sequences), in accordance with previous observations. On the contrary, only three pentapeptide sequence was found to have high similarity with AβC5-34. Third, for the selected peptides corresponding to the cyclo-TXXXR motif, residues at positions 3 and 4 were highly variable and included the majority of natural amino acids, with position 3 exhibiting the highest diversity. At position 2, Thr, Ala, and Val were preferred, while aromatic residues (Phe, Trp, Tyr) were completely excluded from the selected cyclo-TXXXR peptide pool, in full agreement with the aforementioned site-directed mutagenesis studies. At the highly variable position 3, the complete absence of the negatively charged amino acids Glu and Asp among the selected sequences was notable. In general, both negatively (Glu and Asp) and positively charged residues (Lys, His, and Arg) were found to be disfavored among the selected cyclo-TXXXR sequences at positions 2 and 3. At position 4, Ala, Asp, and Trp were found to be the preferred residues. It is noteworthy, that Lys and Gln residues were practically absent from all positions, while the β sheet-breaking amino acid Pro that is typically included in designed peptide-based inhibitors of amyloid aggregation appeared with strikingly low frequencies. Thus, preferred Aβ modulators are cyclic oligopeptide sequences exhibiting the cyclo-TXXXR motif, where X is any natural amino acid. More preferred are cyclic oligopeptide sequences exhibiting the cyclo-TΦZΠR motif, where Φ=any amino acid excluding F, W, Y, Q, D, E, K and P; Z is any amino acid excluding E; and Π is any natural amino acid excluding Q, M, K. Even more preferred are cyclic oligopeptide sequences exhibiting the cyclo-T(T,A,V)Ψ(A,D,W)R motif, where Ψ is a non-negatively charged amino acid. The high residue variability observed at position 3 of the selected TXXXR peptides prompted the inventors to investigate whether AβC5-116 could be further minimized. Indeed, production of truncated variants of AβC5-116, from which Ala2 or Asp4 had been deleted, resulted in a respective two- and three-fold enhancement in the fluorescence of bacterially expressed Aβ42-GFP. In accordance with this, a total of ten distinct cyclic tetrapeptide sequences belonging to the TXXR motif were identified among the selected peptide pool. Taken together, these results indicate that the minimal bioactive entity against Aβ aggregation among this peptide family is a TXXR cyclic tetrapeptide. The present invention describes a versatile and generally applicable method for identifying macrocyclic chemical rescuers of the misfolding of misfolding-prone proteins associated with a protein misfolding disease (MisP), wherein said MisP is selected from SOD1, β-amyloid peptide, tau, α-synuclein, polyglutaminated huntingtin, polyglutaminated ataxin-1, polyglutaminated ataxin-2, polyglutaminated ataxin-3, prion protein, islet amyloid polypeptide (amylin), β2-microglbulin, fragments of immunoglobulin light chain, fragments of immunoglobulin heavy chain, serum amyloid A, ABri peptide, ADan peptide, transthyretin, apolipoprotein A1, gelsolin, transthyretin, lysozyme, phenylalanine hydroxylase, apolipoprotein A-I, calcitonin, prolactin, TDP-43, FUS/TLS; insulin, hemoglobin, α1-antitrypsin, p53; or variants thereof. Preferably the invention presented herein can be used as a method for the identification of chemical agents for the treatment, prevention or diagnosis of diseases related to protein misfolding and aggregation, including amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, Huntington's disease, Creutzfeldt-Jakob disease, cancer, phenylketonuria, type II diabetes, senile systemic amyloidosis, familial amyloidotic polyneuropathy, familial amyloid cardiomyopathy, leptomeningeal amyloidosis, systemic amyloidosis, familial British dementia, familial Danish dementia, light chain amyloidosis, heavy chain amyloidosis, serum amyloid A amyloidosis, lysozyme amyloidosis, dialysis-related amyloidosis, ApoAI amyloidosis, Finnish type familial amyloidosis, hereditary cerebral hemorrhage with amyloidosis (Icelandic type), medullary carcinoma of the thyroid, pituitary prolactinoma, injection-localized amyloidosis, frontotemporal dementia, spinocerebellar ataxia 1, spinocerebellar ataxia 2, spinocerebellar ataxia 3, al-antitrypsin deficiency, sickle-cell anemia, and transmissible spongiform encephalopathy. Most preferably the invention presented herein can be used as a method of treatment, prevention or diagnosis of amyotrophic lateral sclerosis or Alzheimer's disease. TABLE 1Sequences and frequency of appearance of the selected cyclo-TXXXR pentapeptidesas determined by high-throughput sequencing of the isolated pSICLOPPS-NuX1X2X3-X5vectors after the second round of bacterial sorting for enhanced Aβ42-GFP fluorescence.Reads/Reads/Reads/TotalTotalTotalSEQNumberTXXXRpentapeptidepeptidePeptide-encodingIDPeptideAminoacidofreadsreadsreadsnucleotideNOnamesequencereads(%)(%)(%)sequence47AβC5-2TTYAR304,75316.0237.5066.727ACCACGTACGCCAGG(SEQ ID NO: 302)48AβC5-3TTVDR214,46111.2765.2824.734ACCACCGTGGACCGG(SEQ ID NO: 303)49AβC5-5TTTWR175,5109.2284.3233.874ACCACGACCTGGAGG(SEQ ID NO: 304)50AβC5-7TTLHR134,0187.0463.3012.958ACCACGCTGCACCGG(SEQ ID NO: 305)51AβC5-8TTFAR96,7005.0842.3822.134ACCACCTTCGCCCGG(SEQ ID NO: 306)52AβC5-9TVLDR89,6694.7152.2091.979ACCGTCTTGGACCGG(SEQ ID NO: 307)53AβC5-12TTWAR65,9293.4661.6241.455ACCACGTGGGCCAGG(SEQ ID NO: 308)54AβC5-13TALDR62,7923.3011.5471.386ACCGCGCTGGACCGG(SEQ ID NO: 309)55AβC5-15TANVR47,8552.5161.1791.056ACCGCGAACGTGAGG(SEQ ID NO: 310)56AβC5-17TTTAR40,1352.1100.9890.886ACCACCACGGCCCGG(SEQ ID NO: 311)57AβC5-18TTIAR37,1501.9530.9150.820ACCACCATCGCCCGG(SEQ ID NO: 312)58AβC5-19TVWDR37,0911.9500.9140.819ACCGTGTGGGACCGG(SEQ ID NO: 313)59AβC5-20TTISR37,0441.9480.9120.818ACCACCATCAGCCGG(SEQ ID NO: 314)60AβC5-21TTWCR36,2951.9080.8940.801ACCACCTGGTGCCGG(SEQ ID NO: 315)61AβC5-22TVLWR35,8201.8830.8820.791ACCGTCCTGTGGAGG(SEQ ID NO: 316)62AβC5-25TTLAR28,9891.5240.7140.640ACCACCTTGGCGAGG(SEQ ID NO: 317)63AβC5-26TAWCR28,3911.4930.6990.627ACCGCGTGGTGCCGC(SEQ ID NO: 318)64AβC5-27TTSAR28,1881.4820.6940.622ACCACGAGCGCCCGC(SEQ ID NO: 319)65AβC5-29TTLER27,5141.4470.6780.607ACCACCCTCGAGAGG(SEQ ID NO: 320)66AβC5-30TSTAR27,4561.4440.6760.606ACCTCGACGGCGCGG(SEQ ID NO: 321)67AβC5-35TVRDR25,4281.3370.6260.561ACCGTCCGGGACCGG(SEQ ID NO: 322)68AβC5-41TGWAR21,7841.1450.5370.481ACCGGCTGGGCGAGG(SEQ ID NO: 323)69AβC5-44TAWAR20,8071.0940.5120.459ACCGCCTGGGCGAGG(SEQ ID NO: 324)70AβC5-45TTWVR20,7981.0940.5120.459ACCACCTGGGTGCGG(SEQ ID NO: 325)71AβC5-46TLLWR19,9571.0490.4920.440ACCCTATTGTGGCGG(SEQ ID NO: 326)72AβC5-47TTIDR19,7351.0380.4860.436ACCACGATCGACAGG(SEQ ID NO: 327)73AβC5-50TALAR19,4331.0220.4790.429ACCGCGCTCGCGCGC(SEQ ID NO: 328)74AβC5-51TSVDR19,2491.0120.4740.425ACCAGCGTGGACAGG(SEQ ID NO: 329)75AβC5-53TTVWR18,6690.9820.4600.412ACCACCGTGTGGCGC(SEQ ID NO: 330)76AβC5-66TTHWR14,3040.7520.3520.316ACCACGCACTGGCGG(SEQ ID NO: 331)77AβC5-67TARDR14,2130.7470.3500.314ACCGCGAGGGACCGG(SEQ ID NO: 332)78AβC5-73TTRDR12,8940.6780.3180.285ACCACGCGGGACCGG(SEQ ID NO: 333)79AβC5-80TSVHR10,1810.5350.2510.225ACCAGCGTGCACCGG(SEQ ID NO: 334)80AβC5-82TAVWR9,7810.5140.2410.216ACCGCCGTCTGGCGG(SEQ ID NO: 335)81AβC5-83TTGCR9,3620.4920.2310.207ACCACGGGGTGCCGG(SEQ ID NO: 336)82AβC5-89TATDR7,9840.4200.1970.176ACCGCCACCGACAGG(SEQ ID NO: 337)83AβC5-94TVLFR7,4420.3910.1830.164ACCGTCTTGTTCCGC(SEQ ID NO: 338)84AβC5-102TTYNR6,0670.3190.1490.134ACCACCTACAACCGC(SEQ ID NO: 339)85AβC5-105TVRWR5,4500.2870.1340.120ACCGTGCGCTGGCGC(SEQ ID NO: 340)86AβC5-116TAFDR4,2430.2230.1050.094ACCGCGTTCGACCGG(SEQ ID NO: 341)87AβC5-117TTRCR4,2370.2230.1040.094ACCACGCGGTGCAGG(SEQ ID NO: 342)88AβC5-118TTFWR4,2160.2220.1040.093ACCACCTTCTGGCGG(SEQ ID NO: 343)89AβC5-121TIKDR3,9700.2090.0980.088ACCATCAAGGACCGG(SEQ ID NO: 344)90AβC5-123TTVHR3,3710.1770.0830.074ACCACCGTCCACCGG(SEQ ID NO: 345)91AβC5-126TTLLR3,0160.1590.0740.067ACCACGCTCCTCAGG(SEQ ID NO: 346)92AβC5-129TTLFR2,6300.1380.0650.058ACCACGCTCTTCCGG(SEQ ID NO: 347)93AβC5-130TAYHR2,5940.1360.0640.057ACCGCGTACCACCGG(SEQ ID NO: 348)94AβC5-136TALHR2,0260.1070.0500.045ACCGCGTTGCACCGG(SEQ ID NO: 349)95AβC5-139TTSPR1,9040.1000.0470.042ACCACCTCGCCCCGG(SEQ ID NO: 350)96AβC5-146TTWSR1,6120.0850.0400.036ACCACCTGGTCGCGG(SEQ ID NO: 351)97AβC5-147TAMHR1,6110.0850.0400.036ACCGCCATGCACAGG(SEQ ID NO: 352)98AβC5-155TSLDR1,2510.0660.0310.028ACCTCGCTCGACAGG(SEQ ID NO: 353)99AβC5-158TTGAR1,1720.0620.0290.026ACCACGGGGGCGCGC(SEQ ID NO: 354)100AβC5-162TSVWR1,0940.0580.0270.024ACCTCGGTGTGGAGG(SEQ ID NO: 355)101AβC5-173TTHAR9530.0500.0230.021ACCACGCACGCCAGG(SEQ ID NO: 356)102AβC5-176TAGWR9450.0500.0230.021ACCGCGGGCTGGAGG(SEQ ID NO: 357)103AβC5-177TATAR9250.0490.0230.020ACCGCCACCGCGAGG(SEQ ID NO: 358)104AβC5-184TVLAR8180.0430.0200.018ACCGTGCTCGCGCGG(SEQ ID NO: 359)105AβC5-185TTFNR8000.0420.0200.018ACCACGTTCAACAGG(SEQ ID NO: 360)106AβC5-188TGMRR7680.0400.0190.017ACCGGGATGAGGCGG(SEQ ID NO: 361)107AβC5-189TTVAR7570.0400.0190.017ACCACCGTCGCCAGG(SEQ ID NO: 362)108AβC5-190TLCLR7390.0390.0180.016TGCTTGCGCACGCTG(SEQ ID NO: 363)109AβC5-192TGLAR7200.0380.0180.016ACCGGGCTGGCGCGG(SEQ ID NO: 364)110AβC5-198TSWCR6790.0360.0170.015ACCAGCTGGTGCAGG(SEQ ID NO: 365)111AβC5-209TTRAR5800.0300.0140.013ACCACCAGGGCGCGG(SEQ ID NO: 366)112AβC5-215TTPWR5240.0280.0130.012ACCACGCCCTGGAGG(SEQ ID NO: 367)113AβC5-218TVLHR4970.0260.0120.011ACCGTCTTGCACAGG(SEQ ID NO: 368)114AβC5-223TGLDR4640.0240.0110.010ACCGGCCTCGACAGG(SEQ ID NO: 369)115AβC5-230TTSDR4420.0230.0110.010ACCACGTCGGACCGG(SEQ ID NO: 370)116AβC5-239TTMHR3840.0200.0090.008ACCACGATGCACCGC(SEQ ID NO: 371)117AβC5-242TTSTR3760.0200.0090.008ACCACCTCGACCCGG(SEQ ID NO: 372)118AβC5-244TTRVR3660.0190.0090.008ACCACGCGCGTGAGG(SEQ ID NO: 373)119AβC5-245TTRFR3640.0190.0090.008ACCACCCGGTTCCGG(SEQ ID NO: 374)120AβC5-248TTTHR3390.0180.0080.007ACCACGACGCACCGG(SEQ ID NO: 375)121AβC5-250THAWR3340.0180.0080.007ACCCACGCCTGGAGG(SEQ ID NO: 376)122AβC5-252TVIWR3310.0170.0080.007ACCGTGATCTGGCGC(SEQ ID NO: 377)123AβC5-253TTWFR3270.0170.0080.007ACCACGTGGTTCCGG(SEQ ID NO: 378)124AβC5-255TTSRR3250.0170.0080.007ACCACCTCGAGACGG(SEQ ID NO: 379)125AβC5-258TTSCR3010.0160.0070.007ACCACGTCGTGCCGG(SEQ ID NO: 380)126AβC5-260TTWTR2950.0160.0070.007ACCACCTGGACCCGG(SEQ ID NO: 381)127AβC5-262TTSSR2860.0150.0070.006ACCACCTCGAGCCGG(SEQ ID NO: 382)128AβC5-263THLAR2840.0150.0070.006ACCCACCTCGCCCGG(SEQ ID NO: 383)129AβC5-264TSGAR2820.0150.0070.006ACCAGCGGGGCCCGG(SEQ ID NO: 384)130AβC5-266TTLRR2740.0140.0070.006ACCACGCTGCGCCGG(SEQ ID NO: 385)131AβC5-270TATWR2660.0140.0070.006ACCGCGACCTGGAGG(SEQ ID NO: 386)132AβC5-272TCMWR2540.0130.0060.006ACCTGCATGTGGCGC(SEQ ID NO: 387)133AβC5-275TAHVR2490.0130.0060.005ACCGCGCACGTGCGC(SEQ ID NO: 388)134AβC5-276TSWAR2490.0130.0060.005ACCTCGTGGGCGCGG(SEQ ID NO: 389)135AβC5-278TTWLR2410.0130.0060.005ACCACGTGGCTCAGG(SEQ ID NO: 390)136AβC5-291TTLDR2130.0110.0050.005ACCACCCTGGACCGG(SEQ ID NO: 391)137AβC5-294TTPHR2070.0110.0050.005ACCACGCCTCACCGG(SEQ ID NO: 392)138AβC5-298TTRGR2010.0110.0050.004ACCACCCGTGGCCGG(SEQ ID NO: 393)139AβC5-299TTVGR2000.0110.0050.004ACCACCGTGGGCCGG(SEQ ID NO: 394)140AβC5-301TTTRR1910.0100.0050.004ACCACGACGCGCCGC(SEQ ID NO: 395)141AβC5-304TSINR1820.0100.0040.004ACCTCGATCAACAGG(SEQ ID NO: 396)142AβC5-305TTADR1810.0100.0040.004ACCACCGCGGACCGG(SEQ ID NO: 397)143AβC5-315TTSER1580.0080.0040.003ACCACCTCCGAGAGG(SEQ ID NO: 398)144AβC5-316TTCAR1570.0080.0040.003ACCACGTGCGCCAGG(SEQ ID NO: 399)145AβC5-317TTAWR1560.0080.0040.003ACCACGGCCTGGAGG(SEQ ID NO: 400)146AβC5-320TTVER1500.0080.0040.003ACCACCGTCGAGCGG(SEQ ID NO: 401)147AβC5-321TTTFR1480.0080.0040.003ACCACGACGTTCAGG(SEQ ID NO: 402)148AβC5-323TAVDR1470.0080.0040.003ACCGCCGTGGACCGG(SEQ ID NO: 403)149AβC5-325TVWIR1440.0080.0040.003ACCGTGTGGATCAGG(SEQ ID NO: 404)150AβC5-329TTVRR1410.0070.0030.003ACCACCGTACGCAGG(SEQ ID NO: 405)151AβC5-333THVRR1370.0070.0030.003ACCCACGTACGCAGG(SEQ ID NO: 406)152AβC5-343TNLDR1250.0070.0030.003ACCAACCTGGACCGG(SEQ ID NO: 407)153AβC5-344TTPGR1250.0070.0030.003ACCACGCCTGGACGG(SEQ ID NO: 408)154AβC5-348TTLTR1190.0060.0030.003ACCACGCTCACCCGG(SEQ ID NO: 409)155AβC5-355TATVR1150.0060.0030.003ACCGCGACGGTGCGC(SEQ ID NO: 410)156AβC5-359TAMWR1100.0060.0030.002ACCGCCATGTGGCGG(SEQ ID NO: 411)157AβC5-361TTKWR1080.0060.0030.002ACCACGAAGTGGAGG(SEQ ID NO: 412)158AβC5-362TTWDR1070.0060.0030.002ACCACCTGGGACCGG(SEQ ID NO: 413)159AβC5-364TTMAR1060.0060.0030.002ACCACCATGGCCCGG(SEQ ID NO: 414)160AβC5-365TTGGR1060.0060.0030.002ACCACCGGTGGCCGG(SEQ ID NO: 415)161AβC5-366TTMVR1050.0060.0030.002ACCACGATGGTGCGG(SEQ ID NO: 416)162AβC5-375TNLAR970.0050.0020.002ACCAACCTCGCCCGG(SEQ ID NO: 417)163AβC5-376TIRDR960.0050.0020.002ACCATCAGGGACCGG(SEQ ID NO: 418)164AβC5-378TTTGR960.0050.0020.002ACCACGACTGGTAGG(SEQ ID NO: 419)165AβC5-379TRLGR950.0050.0020.002ACCCGTCTTGGCAGG(SEQ ID NO: 420)166AβC5-381TTHTR930.0050.0020.002ACCACGCACACCAGG(SEQ ID NO: 421)167AβC5-382TTITR920.0050.0020.002ACCACCATCACCCGG(SEQ ID NO: 422)168AβC5-384TTYTR900.0050.0020.002ACCACGTACACCAGG(SEQ ID NO: 423)169AβC5-385TTLYR900.0050.0020.002ACCACGCTGTACCGG(SEQ ID NO: 424)170AβC5-389THLDR890.0050.0020.002ACCCACCTGGACCGG(SEQ ID NO: 425)171AβC5-391TLLIR880.0050.0020.002ACCTTGTTGATCAGG(SEQ ID NO: 426)172AβC5-392TTCDR870.0050.0020.002ACCACGTGCGACCGG(SEQ ID NO: 427)173AβC5-393TTGRR870.0050.0020.002ACCACGGGTCGCCGG(SEQ ID NO: 428)174AβC5-394TTVSR860.0050.0020.002ACCACCGTGAGCCGG(SEQ ID NO: 429)175AβC5-395TTQHR850.0040.0020.002ACCACGCAGCACCGG(SEQ ID NO: 430)176AβC5-396TTTPR840.0040.0020.002ACCACTACGCCCAGG(SEQ ID NO: 431)177AβC5-399TAFAR820.0040.0020.002ACCGCCTTCGCCCGG(SEQ ID NO: 432)178AβC5-405TTSHR780.0040.0020.002ACCACGTCACACCGG(SEQ ID NO: 433)179AβC5-410TVLGR760.0040.0020.002ACCGTCTTGGGCCGG(SEQ ID NO: 434)180AβC5-411TTQRR750.0040.0020.002ACCACGCAGCGCAGG(SEQ ID NO: 435)181AβC5-413TSHAR740.0040.0020.002ACCAGTCACGCCAGG(SEQ ID NO: 436)182AβC5-415TTTCR740.0040.0020.002ACCACGACGTGCCGG(SEQ ID NO: 437)183AβC5-422TAWRR720.0040.0020.002ACCGCGTGGCGCCGC(SEQ ID NO: 438)184AβC5-428TTCGR690.0040.0020.002ACCACGTGTGGCCGG(SEQ ID NO: 439)185AβC5-434TTSGR650.0030.0020.001ACCACCTCTGGCCGG(SEQ ID NO: 440)186AβC5-438TTTSR620.0030.0020.001ACCACGACGTCGAGG(SEQ ID NO: 441)187AβC5-440TATGR610.0030.0020.001ACCGCGACTGGACGG(SEQ ID NO: 442)188AβC5-441TAWDR610.0030.0020.001ACCGCGTGGGACCGG(SEQ ID NO: 443)189AβC5-443TTHHR600.0030.0010.001ACCACGCATCACCGG(SEQ ID NO: 444)190AβC5-448TAYAR580.0030.0010.001ACCGCGTACGCCAGG(SEQ ID NO: 445)191AβC5-449TANAR580.0030.0010.001ACCGCGAACGCGAGG(SEQ ID NO: 446)192AβC5-450TRDVR580.0030.0010.001ACCCGCGACGTGAGG(SEQ ID NO: 447)193AβC5-452THVDR580.0030.0010.001ACCCACGTCGACAGG(SEQ ID NO: 448)194AβC5-453TLFWR570.0030.0010.001ACCCTATTCTGGCGG(SEQ ID NO: 449)195AβC5-459TTAAR550.0030.0010.001ACCACCGCGGCCCGG(SEQ ID NO: 450)196AβC5-463TVVDR540.0030.0010.001ACCGTCGTGGACCGG(SEQ ID NO: 451)197AβC5-464TTPAR540.0030.0010.001ACCACTCCGGCCCGG(SEQ ID NO: 452)198AβC5-469TTIGR530.0030.0010.001ACCACGATCGGCAGG(SEQ ID NO: 453)199AβC5-472TMYAR510.0030.0010.001ACCATGTACGCCAGG(SEQ ID NO: 454)200AβC5-473THVAR510.0030.0010.001ACCCACGTGGCCAGG(SEQ ID NO: 455)201AβC5-474TTWPR510.0030.0010.001ACCACCTGGCCGCGG(SEQ ID NO: 456)202AβC5-475TTGDR510.0030.0010.001ACCACCGGTGACCGG(SEQ ID NO: 457)203AβC5-479TTTVR500.0030.0010.001ACCACGACCGTGCGG(SEQ ID NO: 458)204AβC5-481TVFGR500.0030.0010.001ACCGTCTTTGGCAGG(SEQ ID NO: 449)205AβC5-483TRVGR500.0030.0010.001ACCCGTGTGGGCCGG(SEQ ID NO: 460)Sum1,901,945100.00046.84741.980 TABLE 2Sequences and frequency of appearance of the selected cyclic pentapeptidesresembling AβC5-34 as determined by high-throughput sequencing of theisolated pSICLOPPS-NuX1X2X3-X5vectors after the second round ofbacterial sorting for enhanced Aβ42-GFP fluorescence.Reads/Reads/Reads/TotalTotalTotalSEQPeptideAmino acidNumber ofSASPT-likepentapeptidepeptideID NOnamesequencereadsreads (%)reads (%)reads (%)206AβC5-34SASPT2567397.3490.6320.567207AβC5-216SICPT5161.9570.0130.011208AβC5-380SITPT940.3560.0020.002209AβC5-387SHSPT890.3370.0020.002Sum26,3721000.6450.578 TABLE 3Sequences and frequency of appearance of the selected cyclo-TXXRtetrapeptides as determined by high-throughput sequencing ofthe isolated pSICLOPPS-Nu X1X2X3-X5vectors for enchancedAβ42-GFP fluorescence.SEQCYCLICNormalizedIDPEPTIDEPEPTIDEPeptide-encodingread numberNO.NAMESEQUENCEnucleotide sequence(%)210AβC4-9TTCRACCACGTGCCGG1.247884(SEQ ID NO: 461)211AβC4-11TTRRACCACTCGCCGG1.199516(SEQ ID NO: 462)212AβC4-31TTSRACCACGTCGCGG0.324063(SEQ ID NO: 463)213AβC4-34TRGRACACGTGGACGG0.304716(SEQ ID NO: 464)214AβC4-35TTGRACCACTGGCCGG0.295042(SEQ ID NO: 465)215AβC4-41TRRRACACGTCGCAGG0.246675(SEQ ID NO: 466)216AβC4B-9TDQRACCGACCAGCGG2.090359(SEQ ID NO: 467)217AβC4B-41TLIRACCCTGATCCGC0.774951(SEQ ID NO: 468)218AβC4B-80TLWRACCCTGTGGCGG0.256828(SEQ ID NO: 469)219AβC4B-86TLGRACCTTGGGCCGG0.16973(SEQ ID NO: 470)220AβC5(ΔA2)TFDRACCTTCGACCGG—(SEQ ID NO: 471)221AβC5(ΔD4)TAFRACCGCGTTCCGG—(SEQ ID NO: 472) TABLE 4Sequences and frequency of appearance of the selected cyclic hexapeptides asdetermined by high-throughput sequencing of the isolated pSICLOPPS-NuX1X2X3-X5vectors after the second round of bacterial sortingfor enhanced Aβ42-GFP fluorescence.Reads/Reads/SEQNumberTotalTotalIDPeptideofhexapeptidepeptidePeptide-encodingNOnameAminoacid sequencereadsreads (%)reads (%)nucleotide sequence222AβC6-1TPVWFD131,93529.1512.912ACCCCGGTCTGGTTCGAC(SEQ ID NO: 473)223AβC6-2TPAWFD111,13224.5552.453ACCCCGGCCTGGTTCGAC(SEQ ID NO: 474)224AβC6-4TLEFFD27,0575.9780.597ACCTTGGAGTTCTTCGAC(SEQ ID NO: 475)225AβC6-6TVTWFD17,1003.7780.377ACCGTCACGTGGTTCGAC(SEQ ID NO: 476)226AβC6-8TLLIRW13,1352.9020.290ACCTTGTTGATCAGGTGG(SEQ ID NO: 477)227AβC6-10TLKWLN11,0162.4340.243ACCCTCAAGTGGCTGAAC(SEQ ID NO: 478)228AβC6-21TKEYFD1,2310.2720.027ACCAAGGAGTACTTCGAC(SEQ ID NO: 479)229AβC6-26TLHWFE6470.1430.014ACCCTCCACTGGTTCGAG(SEQ ID NO: 480)230AβC6-27TCSWFD6230.1380.014ACCTGCTCGTGGTTCGAC(SEQ ID NO: 481)231AβC6-28TLEYFM5560.1230.012ACCCTCGAGTACTTCATG(SEQ ID NO: 482)232AβC6-32TLCWLN4550.1010.010ACCCTGTGCTGGCTCAAC(SEQ ID NO: 483)233AβC6-36TPIVFD3840.0850.008ACCCCGATCGTGTTCGAC(SEQ ID NO: 484)234AβC6-37TLWVFD3550.0780.008ACCCTGTGGGTCTTCGAC(SEQ ID NO: 485)235AβC6-40TPLWFN3160.0700.007ACCCCCTTGTGGTTCAAC(SEQ ID NO: 486)236AβC6-41TSVEYE3070.0680.007ACCTCGGTCGAGTACGAG(SEQ ID NO: 487)237AβC6-42TLGWLD3070.0680.007ACCCTGGGCTGGTTGGAC(SEQ ID NO: 488)238AβC6-44TPPWFD2890.0640.006ACCCCGCCCTGGTTCGAC(SEQ ID NO: 489)239AβC6-46TPCWFD2520.0560.006ACCCCGTGCTGGTTCGAC(SEQ ID NO: 490)240AβC6-47TLSWYD2390.0530.005ACCTTGTCCTGGTACGAC(SEQ ID NO: 491)241AβC6-48TPVLVD2360.0520.005ACCCCGGTCCTGGTCGAC(SEQ ID NO: 492)242AβC6-49TLEYLW2330.0510.005ACCCTCGAGTACTTGTGG(SEQ ID NO: 493)243AβC6-50TIFWFD2270.0500.005ACCATCTTCTGGTTCGAC(SEQ ID NO: 494)244AβC6-53TPALVD2080.0460.005ACCCCGGCCCTGGTCGAC(SEQ ID NO: 495)245AβC6-55TPGWFD1800.0400.004ACCCCCGGCTGGTTCGAC(SEQ ID NO: 496)246AβC6-57TLSVFD1760.0390.004ACCTTGTCCGTCTTCGAC(SEQ ID NO: 497)247AβC6-58TPGLVD1420.0310.003ACCCCCGGTCTGGTCGAC(SEQ ID NO: 498)248AβC6-59TLSWFN1410.0310.003ACCCTCTCCTGGTTCAAC(SEQ ID NO: 499)249AβC6-63TLDFFD1140.0250.003ACCTTGGACTTCTTCGAC(SEQ ID NO: 500)250AβC6-65TPSWFD1050.0230.002ACCCCGTCCTGGTTCGAC(SEQ ID NO: 501)251AβC6-68TPALFD1010.0220.002ACCCCGGCCCTGTTCGAC(SEQ ID NO: 502)252AβC6-69TPAWSD860.0190.002ACCCCGGCCTGGTCCGAC(SEQ ID NO: 503)253AβC6-78TPARFD550.0120.001ACCCCGGCCCGGTTCGAC(SEQ ID NO: 504)254AβC6-79TPAWLD550.0120.001ACCCCGGCCTGGCTCGAC(SEQ ID NO: 505)255AβC6-80TPVWLD550.0120.001ACCCCGGTCTGGCTCGAC(SEQ ID NO: 506)Sum319,55370.6067.053 III. Peptide Modifications Peptides and polypeptides of the invention include those corresponding to linearized versions of the described cyclic oligopeptide SOD1 modulators, i.e., sequences where a break in the amino acid backbone chain of a described cyclic oligopeptide modulator has been introduced and which thereafter contains a free N-terminal NH2amino group and a free C-terminal —COOH carboxyl group. For example, for the cyclic pentapeptide SOD1 modulator SOD1C5-4 with amino acid sequence cyclo-TWSVW (SEQ ID NO: 4), a preferred peptide SOD1 modulator of the present invention is also a linearized version of SOD1C5-4, namely the oligopeptide NH2-TWSVW-COOH (SEQ ID NO: 4). In addition, since the herein described oligopeptide SOD1 modulators are cyclic in nature, they do not possess a “starting point” (e.g. N terminus) or “end point” (e.g. C terminus). Thus, all circular permutants, e.g., linear variants resulting from cleavage of an existing peptide bond to introduce new termini elsewhere in the peptide sequence, of the described cyclic oligopeptide SOD1 modulators are also preferred cyclic oligopeptide SOD1 modulators of the present invention. For example, for the cyclic pentapeptide SOD1 modulator SOD1C5-4 with amino acid sequence cyclo-TWSVW (SEQ ID NO: 4), preferred peptide SOD1 modulators of the present invention are also all equivalent circular permutants of SOD1C5-4, namely the oligopeptides cyclo-WSVWT (SEQ ID NO: 507), cyclo-SVWTW (SEQ ID NO: 508), cyclo-VWTWS (SEQ ID NO: 509), and cyclo-WTWSV (SEQ ID NO: 510). Similarly, preferred peptide SOD1 modulators of the present invention are the linearized versions of all described cyclic oligopeptide SOD1 modulators and of all of their equivalent circular permutants. For example, for SOD1C5-4, apart from the modification mentioned above, preferred peptide SOD1 modulator of the present invention are linearized versions of all circular permutants equivalent SOD1C5-4, namely the oligopeptides NH2-WSVWT-COOH (SEQ ID NO: 507), NH2-SVWTW-COOH (SEQ ID NO: 508), NH2-VWTWS-COOH (SEQ ID NO: 509), and NH2-WTWSV-COOH (SEQ ID NO: 510). Similarly, for the cyclic pentapeptide Aβ modulator AβC5-116 with amino acid sequence cyclo-TAFDR (SEQ ID NO: 86), a preferred peptide Aβ modulator of the present invention is also a linearized version of AβC5-116, namely the oligopeptide NH2-TAFDR-COOH. In addition, since the herein described oligopeptide Aβ modulators are cyclic in nature, they do not possess a “starting point” (e.g. N terminus) or “end point” (e.g. C terminus). Thus, all circular permutants, e.g., linear variants resulting from cleavage of an existing peptide bond to introduce new termini elsewhere in the peptide sequence, of the described cyclic oligopeptide Aβ modulators are also preferred cyclic oligopeptide Aβ modulators of the present invention. For example, for the cyclic pentapeptide Aβ modulator AβC5-116 with amino acid sequence cyclo-TAFDR (SEQ ID NO: 86), preferred peptide Aβ modulators of the present invention are also all equivalent circular permutants of AβC5-116, namely the oligopeptides cyclo-AFDRT (SEQ ID NO: 511), cyclo-FDRTA and NH2-FDRTA-COOH (SEQ ID NO: 512), cyclo-DRTAF (SEQ ID NO: 513), and cyclo-RTAFD (SEQ ID NO: 514). Similarly, preferred peptide Aβ modulators of the present invention are the linearized versions of all described cyclic oligopeptide Aβ modulators and of all of their equivalent circular permutants. For example, for AβC5-116, apart from the modification mentioned above, preferred peptide Aβ modulator of the present invention are linearized versions of all circular permutants equivalent AβC5-116, namely the oligopeptides NH2-AFDRT-COOH (SEQ ID NO: 511), NH2-FDRTA-COOH (SEQ ID NO: 512), NH2-DRTAF-COOH (SEQ ID NO: 513), and NH2-RTAFD-COOH (SEQ ID NO: 514). Peptides and polypeptides of the invention include those containing conservative amino acid substitutions. Such peptides and polypeptides are encompassed by the invention provided the peptide or polypeptide can bind to SOD1 or Aβ. As used herein, suitable conservative substitutions of amino acids are known to those of skill in this art and can be made generally without altering the biological activity of the resulting molecule. Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. Molecular Biology of the Gene, 4th Edition, 1987, The Benjamin/Cummings Pub. co., p. 224). Such substitutions can be made in accordance with those set forth as follows: Ala (A) Gly; Ser Arg (R) Lys Asn (N) Gln; His Cys (C) Ser Gln (Q) Asn Glu (E) Asp Gly (G) Ala; Pro His (H) Asn; Gln Ile (I) Leu; Val Leu (L.) Ile; Val Lys (K) Arg; Gln; Glu Met (N) Leu; Tyr; Ile Phe (F) Met; Leu; Tyr Ser (S) Thr Thr (T) Ser Trp (W) Tyr Tyr (Y) Trp; Phe Val (V) Ile; Leu. Other substitutions also are permissible and can be determined empirically or in accord with known conservative substitutions. The peptidic compounds of the present invention can inhibit protein misfolding and aggregation, wherein said peptidic compounds can be a head-to-tail cyclic peptide, side-chain-to-tail cyclic peptide, bicyclic peptide, lanthipeptide, linaridin, proteusin, cyanobactin, thiopeptide, bottromycin, microcin, lasso peptide, microviridin, amatoxin, phallotoxin, 0-defensin, orbitide, or cyclotide. The peptidic compounds of the present invention also serve as structural models for non-peptidic molecules or “mimetics” with similar biological activity. One can also readily modify peptides by phosphorylation, and other methods for making peptide derivatives of the compounds of the present invention are described in Hruby, et al. Biochm. J. 268(2): 249-262, 1990, incorporated herewith by reference. Thus, the peptide compounds of the invention also serve as structural models for non-peptidic compounds with similar biological activity. Those of skill in the art recognize that a variety of techniques are available for constructing compounds with the same or similar desired biological activity as the lead peptide compound but with more favorable activity than the lead with respect to solubility, stability, and susceptibility to hydrolysis and proteolysis. See Morgan and Gainor, Ann. Rep. Med. Chem. 24:243-252, 1989, incorporated herein by reference. These techniques include replacing the peptide backbone with a backbone composed of phosphonates, amidates, carbamates, sulfonamides, and secondary amines. The term mimetic, and in particular, peptidomimetic, is intended to include isosteres. The term “isostere” as used herein is intended to include a chemical structure that can be substituted for a second chemical structure because the steric conformation of the first structure fits a binding site specific for the second structure. The term specifically includes peptide backbone modifications (i.e., amide bond mimetics) well known to those skilled in the art. Such modifications include modifications of the amide nitrogen, the α-carbon, amide carbonyl, complete replacement of the amide bond, extensions, deletions or backbone crosslinks. Several peptide backbone modifications are known, including ψ[CH2S], ψ[CH2NH], ψ[CSNH2], ψ[NHCOO], ψ[COCH2], and ψ[(E) or (Z) CH═CH]. In the nomenclature used above, w indicates the absence of an amide bond. The structure that replaces the amide group is specified within the brackets. Other examples of isosteres include peptides substituted with one or more benzodiazepine molecules (see e.g., James, G. L. et al. (1993) Science 260:1937-1942). Other possible modifications include an N-alkyl (or aryl) substitution (ψ[CONR]), backbone crosslinking to construct lactams and other cyclic structures, substitution of all D-amino acids for all L-amino acids Within the compound (“inverso” compounds) or retro-inverso amino acid incorporation (ψ[NHCO]). By “inverso” is meant replacing L-amino acids of a sequence With D-amino acids, and by “retro-inverso” or “enantio-retro” is meant reversing the sequence of the amino acids (“retro”) and replacing the L-amino acids With D-amino acids. For example, if the parent peptide is Thr-Ala-Tyr, the retro modified form is Tyr-Ala-Thr, the inverso form is thr-ala-tyr, and the retro inverso form is tyr-ala-thr (lower case letters refer to D-amino acids). Compared to the parent peptide, a retro-inverso peptide has a reversed backbone while retaining substantially the original spatial conformation of the side chains, resulting in a retro-inverso isomer with a topology that closely resembles the parent peptide. See Goodman et al. “Perspectives in Peptide Chemistry” pp. 283-294 (1981). See also U.S. Pat. No. 4,522,752 by Sisto for further description of “retro-inverso” peptides. Preferably, the modulator compound inhibits aggregation of SOD1 or natural β-amyloid peptides when contacted with SOD1 or the natural β-amyloid peptides, and/or inhibits SOD1 or Aβ neurotoxicity. Alternatively, the modulator compound can promote aggregation of SOD1 or natural β-amyloid peptides when contacted with the SOD1 or natural β-amyloid peptides. The type and number of modifying groups coupled to the modulator are selected such that the compound alters (and preferably inhibits) aggregation of SOD1 or natural β-amyloid peptides when contacted with SOD1 or the natural β-amyloid peptides. A single modifying group can be coupled to the modulator or, alternatively, multiple modifying groups can be coupled to the modulator. Within a modulator compound of the invention, a peptidic structure (such as a cyclic oligopeptide SOD1 or Aβ modulator or an amino acid sequence corresponding to a rearranged or modified cyclic oligopeptide SOD1 or Aβ modulator) is coupled directly or indirectly to at least one modifying group. The term “modifying group” is intended to include structures that are directly attached to the peptidic structure (e.g., by covalent coupling), as well as those that are indirectly attached to the peptidic structure (e.g., by a stable non-covalent association or by covalent coupling to additional amino acid residues, or mimetics, analogues or derivatives thereof, which may flank the cyclic oligopeptide SOD1 or Aβ modulator). For example, the modifying group can be coupled to a side chain of at least one amino acid residue of a cyclic oligopeptide SOD1 or Aβ modulator, or to a peptidic or peptidomimetic region flanking the cyclic oligopeptide SOD1 or Aβ modulator (e.g., through the epsilon amino group of a lysyl residue(s), through the carboxyl group of an aspartic acid residue(s) or a glutamic acid residue(s), through a hydroxy group of a tyrosyl residue(s), a serine residue(s) or a threonine residue(s) or other suitable reactive group on an amino acid side chain). Modifying groups covalently coupled to the peptidic structure can be attached by means and using methods well known in the art for linking chemical structures, including, for example, amide, alkylamino, carbamate or urea bonds. The modifying group(s) is selected such that the modulator compound alters, and preferably inhibits, SOD1 or β-amyloid peptides aggregation when contacted with SOD1 or the β-amyloid peptides or inhibits the neurotoxicity of SOD1 or the β-amyloid peptides when contacted by them. Although not intending to be limited by mechanism, the modifying group(s) of the modulator compounds of the invention is thought to function as a key pharmacophore which is important for conferring on the modulator the ability to disrupt SOD1 or Aβ aggregation. In one embodiment, the modifying group is a “biotinyl structure”, which includes biotinyl groups and analogues and derivatives thereof (such as a 2-iminobiotinyl group). In another embodiment, the modifying group can comprise a “fluorescein-containing group”, such as a group derived from reacting a SOD1- or an Aβ-derived peptidic structure with 5-(and 6-)-carboxyfluorescein, succinimidyl ester or fluorescein isothiocyanate. In various other embodiments, the modifying group(s) can comprise an N-acetylneuraminyl group, a trans-4-cotininecarboxyl group, a 2-imino-1-imidazolidineacetyl group, an (S)-(−)-indoline-2-carboxyl group, a (−)-menthoxyacetyl group, a 2-norbornaneacetyl group, a γ-oxo-5-acenaphthenebutyryl, a (−)-2-oxo-4-thiazolidinecarboxyl group, a tetrahydro-3-furoyl group, a 2-iminobiotinyl group, a diethylenetriaminepentaacetyl group, a 4-morpholinecarbonyl group, a 2-thiopheneacetyl group or a 2-thiophenesulfonyl group. Preferred modifying groups include groups comprising cholyl structures, biotinyl structures, fluorescein-containing groups, a diethylene-triaminepentaacetyl group, a (−)-menthoxyacetyl group, and a N-acetylneuraminyl group. More preferred modifying groups those comprising a cholyl structure or an iminiobiotinyl group. Yet another type of modifying group is a compound that contains a non-natural amino acid. SOD1 or β-amyloid modulator compounds of the invention can be further modified to alter the specific properties of the compound while retaining the ability of the compound to alter SOD1 or Aβ aggregation and inhibit SOD1 or Aβ neurotoxicity. For example, in one embodiment, the compound is further modified to alter a pharmacokinetic property of the compound, such as in vivo stability or half-life. In another embodiment, the compound is further modified to label the compound with a detectable substance. In yet another embodiment, the compound is further modified to couple the compound to an additional therapeutic moiety. To further chemically modify the compound, such as to alter the pharmacokinetic properties of the compound, reactive groups can be derivatized. A modulator compound can be further modified to label the compound by reacting the compound with a detectable substance. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include14C,123I,124I,125I,131,99mTc,35S or3H. In a preferred embodiment, a modulator compound is radioactively labeled with1C, either by incorporation of14C into the modifying group or one or more amino acid structures in the modulator compound. Labeled modulator compounds can be used to assess the in vivo pharmacokinetics of the compounds, as well as to detect SOD1 or Aβ aggregation, for example for diagnostic purposes. SOD1 or Aβ aggregation can be detected using a labeled modulator compound either in vivo or in an in vitro sample derived from a subject. Preferably, for use as an in vivo diagnostic agent, a modulator compound of the invention is labeled with radioactive technetium or iodine. Accordingly, in one embodiment, the invention provides a modulator compound labeled with technetium, preferably99mTc. Methods for labeling peptide compounds with technetium are known in the art (see e.g., U.S. Pat. Nos. 5,443,815, 5,225,180 and 5,405,597, all by Dean et al.; Stepniak-Biniakiewicz, D., et al. (1992) J. Med. Chem. 35:274-279; Fritzberg, A. R., et al. (1988) Proc. Natl. Acad. Sci. USA 85:4025-4029; Baidoo, K. E., et al. (1990) Cancer Res. Suppl. 50:799s-803s; and Regan, L. and Smith, C. K. (1995) Science 270:980-982). Furthermore, an additional modification of a modulator compound of the invention can serve to confer an additional therapeutic property on the compound. That is, the additional chemical modification can comprise an additional functional moiety. For example, a functional moiety which serves to break down or dissolve amyloid plaques can be coupled to the modulator compound. In this form, the modified modulator serves to target the compound to SOD1 or Aβ peptides and disrupt their polymerization, whereas the additional functional moiety serves to break down or dissolve SOD1 aggregates or amyloid plaques after the compound has been targeted to these sites. In an alternative chemical modification, a SOD1 or β-amyloid compound of the invention is prepared in a “prodrug” form, wherein the compound itself does not modulate aggregation, but rather is capable of being transformed, upon metabolism in vivo, into a SOD1 or β-amyloid modulator compound as defined herein. For example, in this type of compound, the modulating group can be present in a prodrug form that is capable of being converted upon metabolism into the form of an active modulating group. Such a prodrug form of a modifying group is referred to herein as a “secondary modifying group.” A variety of strategies are known in the art for preparing peptide prodrugs that limit metabolism in order to optimize delivery of the active form of the peptide-based drug (see e.g., Moss, J. (1995) in Peptide-Based Drug Design: Controlling Transport and Metabolism, Taylor, M. D. and Amidon, G. L. (eds), Chapter 18. Additionally strategies have been specifically tailored to achieving CNS delivery based on “sequential metabolism” (see e.g., Bodor, N., et al. (1992) Science 257:1698-1700; Prokai, L., et al. (1994) J. Am. Chem. Soc. 116:2643-2644; Bodor, N. and Prokai, L. (1995) in Peptide-Based Drug Design: Controlling Transport and Metabolism, Taylor, M. D. and Amidon, G. L. (eds), Chapter 14. In one embodiment of a prodrug form of a modulator of the invention, the modifying group comprises an alkyl ester to facilitate blood-brain barrier permeability. Modulator compounds of the invention can be prepared by chemical synthesis using standard techniques known in the art. The peptide component of a modulator composed, at least in part, of a peptide, can be synthesized using standard techniques such as those described in Bodansky, M. Principles of Peptide Synthesis, Springer Verlag, Berlin (1993) and Grant, G. A. (ed.). Synthetic Peptides: A User's Guide, W. H. Freeman and Company, New York (1992). Automated peptide synthesizers are commercially available (e.g., Advanced ChemTech Model 396; Milligen/Biosearch 9600). Additionally, one or more modulating groups can be attached to the SOD1 or Aβ modulator (e.g., a SOD1 or an Aβ aggregation core domain) by standard methods, for example using methods for reaction through an amino group, a carboxyl group, a hydroxyl group (e.g., on a tyrosine, serine or threonine residue) or other suitable reactive group on an amino acid side chain (see e.g., Greene, T. W. and Wuts, P. G. M. Protective Groups in Organic Synthesis, John Wiley and Sons, Inc., New York (1991)). Alternatively, modulator compounds of the invention can be prepared biosynthetically and isolated in pure or enriched form from a recombinant production host, such a bacterial, yeast, plant, or mammalian cell (see, e.g., Scott C P, Abel-Santos E, Jones A D, Benkovic S J, Structural requirements for the biosynthesis of backbone cyclic peptide libraries. Chem Biol. 2001 August; 8(8):801-15, as an example of recombinant production of cyclic oligopeptides in bacterial cells). Alternatively, modulator compounds of the invention can be prepared biosynthetically from a recombinant production host, such a bacterial, yeast, plant, or mammalian cell, but may not be isolated in pure or enriched form, and instead be provided to the diseased organism as part of recombinant production host, such a bacterial, yeast, plant, or mammalian cell producing the specific modulator compound recombinantly in the form of a probiotic. By the term “probiotic”, we mean living microorganisms or other cultured cells that may provide health benefits when administered and consumed in adequate amounts (see, e.g., O'Toole P W, Marchesi J R, Hill C, Next-generation probiotics: the spectrum from probiotics to live biotherapeutics, Nat Microbiol. 2017 Apr. 25; 2:17057). IV. Hybrid Modulators A hybrid molecule of the invention includes a peptide or polypeptide that binds to the amyloid or non-amyloid form of SOD1 or amyloid form of Aβ, and a scaffold molecule. The scaffold molecule can include a diagnostic or therapeutic reagent. The therapeutic or diagnostic reagent can be a polypeptide, small molecule or compound. In particular, provided herein are hybrid molecules, such as hybrid polypeptides, that include a peptide or polypeptide provided herein, and additional amino acid residues (typically, 5, 10, 15, 20, 30, 40, 50, 100 or more) such that the resulting hybrid molecule specifically interacts with SOD1 or Aβ. The motif can be modified, such as by replacing certain amino acids or by directed and random evolution methods, to produce motifs with greater affinity. As used herein, a hybrid polypeptide refers to a polypeptide that includes regions from at least two sources, such as from an antibody or enzyme or other scaffold that can be a recipient, and a binding motif, such as a polypeptide or peptide that binds to an amyloid or non-amyloid form of SOD1 or the amyloid form of the Aβ peptide. Thus, among the hybrid molecules provided herein are hybrid molecules, particularly hybrid polypeptides that are produced by grafting a binding motif (e.g., peptide) from one molecule into a scaffold, such as an antibody or fragment thereof or an enzyme or other reporter molecule. The hybrid polypeptides provided herein, even the hybrid immunoglobulins, are not antibodies per se, but are polypeptides that are hybrid molecules containing a selected motif (e.g., a peptide that binds to the amyloid or non-amyloid form of SOD1 or the amyloid form of the Aβ peptide) inserted into another polypeptide such that the motif retains or obtains the ability to bind to a protein involved in disease of protein aggregation. The hybrid polypeptides can include portions of antibodies or other scaffolds, but they also include a non-immunoglobulin or non-scaffold portion grafted therein. The non-immunoglobulin portion is identified by its ability to specifically bind to a targeted polypeptide isoform. The hybrid polypeptide can specifically bind to the targeted infectious or disease-related or a selected isoform of a polypeptide as monomer with sufficient affinity to detect the resulting complex or to precipitate the targeted polypeptide. The scaffold is selected so that insertion of the motif therein does not substantially alter (i.e., retains) the desired binding specificity of the motif. The scaffold additionally can be selected for its properties, such as its ability to act as a reporter. Methods for production of hybrid molecules that specifically interact with a one form of a conformer of a protein associated with a disease of protein conformation or involving protein aggregation are provided. In these methods a polypeptide motif from the protein is inserted into a scaffold such that the resulting molecule exhibits specific binding to one conformer compared to other conformers. In particular, the hybrid molecule can exhibit specific binding to the amyloid or non-amyloid form of SOD1 or the amyloid form of the Aβ peptide. Peptides of the invention have been shown to bind to SOD1 or Aβ in vitro and in vivo. The peptides can be incorporated into a scaffold that comprises additional amino acid sequences and/or compounds. The hybrid molecule can then be used to label or treat the aggregates associated with SOD1 or plaques associated with Aβ amyloid. The polypeptides, nucleic acids encoding the polypeptides, and methods of using the polypeptides or nucleic acids can be used to identify, diagnose and/or treat disorders associated with plaque formation in brain tissue. Any molecule, such as a polypeptide, into which the selected polypeptide motif is inserted (or linked) such that the resulting hybrid polypeptide has the desired binding specificity, is contemplated for use as part of the hybrid molecules herein. The polypeptides can be inserted into any sequence of amino acids that at least contains a sufficient number (10, 20, 30, 50, 100 or more amino acids) to properly present the motif for binding to the targeted amyloid plaque. The purpose of the scaffold is to present the motif to the targeted polypeptide in a form that binds thereto. The scaffold can be designed or chosen to have additional properties, such as the ability to serve as a detectable marker or label or to have additional binding specificity to permit or aid in its use in assays to detect particular isoforms of a target protein (e.g., the amyloid or non-amyloid form of SOD1 or the amyloid form of the Aβ peptide) or for screening for therapeutics or other assays and methods. The scaffolds include reporter molecules, such as fluorescent proteins and enzymes or fragments thereof, and binding molecules, such as antibodies or fragments thereof. Selected scaffolds include all or portions of antibodies, enzymes, such as luciferases, alkaline phosphatases, β-galactosidases and other signal-generating enzymes, chemiluminescence generators, such as horseradish peroxidase; fluorescent proteins, such as red, green and blue fluorescent proteins, which are well known; and chromogenic proteins. The peptide motif is inserted into the scaffold in a region that does not disturb any desired activity. The scaffolds can include other functional domains, such as an additional binding site, such as one specific for a second moiety for detection. V. Nucleic Acid Molecules Nucleic acid molecules encoding any of the peptides, polypeptides or hybrid polypeptides provided herein are provided in the general experimental procedures. Such molecules can be introduced into plasmids and vectors for expression in suitable host cells. As used herein, the term “nucleic acid” refers to single-stranded and/or double-stranded polynucleotides such as deoxyribonucleic acid (DNA), and ribonucleic acid (RNA) as well as analogs or derivatives of either RNA or DNA. Also included in the term “nucleic acid” are analogs of nucleic acids such as peptide nucleic acid (PNA), phosphorothioate DNA, and other such analogs and derivatives or combinations thereof. The term should be understood to include, as equivalents, derivatives, variants and analogs of either RNA or DNA made from nucleotide analogs, single (sense or antisense) and double-stranded polynucleotides. Deoxyribonucleotides include deoxyadenosine, deoxycytidine, deoxyguanosine and deoxythymidine. For RNA, the uracil base is uridine. Plasmids and vectors containing the nucleic acid molecules also are provided in the general experimental procedures. Cells containing the vectors, including cells that express the encoded proteins are also provided. The cell can be a bacterial cell, a yeast cell, a fungal cell, a plant cell, an insect cell or an animal cell. Methods for producing a cyclic oligopeptide or a hybrid polypeptide, for example, growing the cell under conditions whereby the encoded polypeptide is expressed by the cell, and recovering the expressed protein, are provided herein. The cells are used for expression of the cyclic oligopeptide or the protein, which can be secreted or expressed in the cytoplasm. The hybrid polypeptides also can be chemically synthesized using standard methods of protein synthesis known in the art. VI. Pharmaceutical Compositions It is envisioned that one would use the modulators of the present invention as an Alzheimer's disease or amyotrophic lateral sclerosis therapeutic. If the modulator were peptide in nature, one could use a gene therapy technique to deliver DNA constructs encoding the modulator to the affected sites. For drug formulations, one would expect that the formulations reach and be effective at the affected site. These modulators would more likely be carbohydrate and peptide mixtures, especially mixtures capable of overcoming the blood brain barrier. For examples, see Tamai, et al., Adv. Drug Delivery Review 19:401-424, 1996, hereby incorporated by reference. In these cases, the disrupting element of the modulators would also facilitate transport across the blood-brain barrier. Thus, the present invention encompasses methods for therapeutic treatments of amyotrophic lateral sclerosis and Alzheimer's disease, comprising administering a compound of the invention in amounts sufficient to modulate the natural course of SOD1 or Aβ aggregation. Accordingly, the present invention includes pharmaceutical compositions comprising, as an active ingredient, at least one of the peptides or other compounds of the invention in association with a pharmaceutical carrier or diluent. The compounds of the invention can be administered by oral, parenteral (intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection), transdermal, nasal, vaginal, rectal, or sublingual routes of administration. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert pharmaceutically acceptable carrier such as sucrose, lactose, or starch. Such dosage forms can also comprise, as is normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, with the elixirs containing inert diluents commonly used in the art, such as water. Besides such inert diluents, compositions can also include adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents. Preparations according to this invention for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, or emulsions. Examples of non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate. Such dosage forms may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They may be sterilized by, for example, filtration through bacteria-retaining filters, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured using sterile water, or some other sterile injectable medium, immediately before use. Compositions for rectal or vaginal administration are preferably suppositories which may contain, in addition to the active substance, excipients such as cocoa butter or a suppository wax. Compositions for nasal or sublingual administration area are also prepared with standard excipients well known in the art. The dosage of active ingredient in the compositions of this invention may be varied; however, it is necessary that the amount of the active ingredient shall be such that a suitable dosage form is obtained. The selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment desired. The following examples illustrate aspects of the present invention including the construction and screening of a peptide macrocycle library; the identification of macrocyclic peptide rescuers of the misfolding and aggregation of the prominent PMD-associated protein target SOD1 and fALS-associated variants thereof, as well as as a second prominent PMD-associated protein target, Aβ, and finally their use in rescuing of SOD1 and Aβ aggregation and toxicity in vitro and in vivo. The Examples are not in any way limiting the scope of invention. EXAMPLES Example 1 Combinatorial libraries of random cyclic tetra-, penta-, and hexapeptides have been selected to be studied as potential rescuers of SOD1 and mutant SOD1 misfolding and pathogenic aggregation. A technique named split intein circular ligation of peptides and proteins (SICLOPPS) (U.S. Pat. No. 7,354,756 B1 “Intein-mediated cyclization of peptides”) for producing peptide libraries inE. coliis being used. SICLOPPS uses split inteins, i.e. self-splicing protein elements, for performing N- to C-terminal peptide cyclization and biosynthesize cyclic peptides as short as four amino acids long. The only requirement for the intein splicing reaction and peptide cyclization to occur is the presence of a nucleophilic amino acids cysteine (C), serine (S), or threonine (T) as the first amino acid of the extein following the C-terminus of the intein. In order for the inventors to maximize the diversity of the libraries, they chose to study peptides with the general formula cyclo-NuX1X2. . . XN, where Nu=C, S or T; X is any one of the twenty natural amino acids and N=3-5 (FIG.1A). The maximum theoretical diversity of the combined cyclo-NuX1X2X3-X5library is >10 million different sequences (FIG.1B). The libraries of genes encoding this combinatorial library of random cyclic oligopeptides were constructed using degenerate codons. The inventors constructed the high diversity pSICLOPPS-NuX1X2X3-X5vector library which is expected to be encoding all of the theoretically possible designed cyclic tetra-, penta-, and hexapeptide NuX1X2X3-X5sequences using molecular biology techniques already known and used in the art. It has been demonstrated previously that the fluorescence ofE. colicells expressing a recombinant protein whose C terminus is fused to GFP correlates well with the amount of soluble and folded protein (Waldo G S, Standish B M, Berendzen J, Terwilliger T C. Nat Biotechnol. 1999 July; 17(7):691-5). Based on this, the inventors reasoned that the fluorescence of MisP-GFP fusions can serve as a reliable reporter for the identification of chemical rescuers of MisP misfolding for a number of disease-associated MisPs, including SOD1. In order to test this hypothesis, the inventors generated fusions of SOD1 variants, whose misfolding and aggregation have been linked with the pathology of familial forms of ALS (fALS), with GFP. Expression of these fusions inE. coli, yielded levels of cellular fluorescence, which were significantly decreased compared to that of the generally non-pathogenic, wild-type SOD1 (FIG.2A). Western blot analysis indicated that this occurs because the accumulation of soluble SOD1-GFP is decreased in the presence of misfolding-inducing amino acid substitutions, which in turn takes place due to enhanced misfolding/aggregation of GFP-fused, as well as fusion-free SOD1 (FIG.2B,2C). Thus, the fluorescence ofE. colicells overexpressing SOD1-GFP fusions appears to be a good indicator of SOD1 folding and misfolding. Example 2 To test whether the bacterial platform can be utilized to identify chemical rescuers of disease-associated SOD1, the inventors screened for cyclic oligopeptides that inhibit the aggregation of SOD1(A4V), a fALS-associated variant, whose misfolding and aggregation causes a very aggressive form of the disease with an average survival time of only 1.2 years after diagnosis. FACS screening of the cyclo-NuX1X2X3-X5oligopeptide library for bacterial clones exhibiting enhanced levels of SOD1(A4V)-GFP fluorescence yielded anE. colipopulation with about 10-fold increased fluorescence after four rounds of sorting (FIGS.3A,3B). Twenty randomly selected clones from the isolated population exhibited up to 10-fold enhanced fluorescence compared toE. colicells producing randomly selected cyclic oligopeptides from the initial library. Four of the isolated clones exhibited the highest levels of cellular SOD1(A4V)-GFP fluorescence (FIG.3C), and were selected for further analysis. These clones (i) expressed tetra-partite IC-peptide-IN-CBD fusions, which could undergo splicing (FIG.3D), (ii) exhibited splicing-activity-dependent enhanced SOD1(A4V)-GFP fluorescence (FIG.3C), and (iii) exhibited SOD1-specific enhancement of bacterial fluorescence (FIG.3E). Western blot analysis indicated that this enhanced SOD1(A4V)-GFP fluorescence phenotype occurs due to accumulation of enhanced amounts of soluble SOD1(A4V) in these clones (FIG.3F). Sequencing of the peptide-encoding region of the pSICLOPPS vector contained in these clones revealed that they all encode cyclic pentapeptides with sequences TASFW (SEQ ID NO: 2), TWSVW (SEQ ID NO: 4), and TFSMW (SEQ ID NO: 6) (FIG.3G), thus indicating a dominant cyclo-TXSXW bioactive motif. Example 3 The peptide cyclo-TWSVW (SEQ ID NO: 4), hereafter referred to as SOD1C5-4 (FIG.4A), which was present twice among the four selected clones, was selected for further analysis and was produced in mg quantities by solid-phase synthesis. Isolated SOD1(A4V) was utilized to assess the effect of the selected cyclic pentapeptide SOD1C5-4 on its aggregation process. CD spectroscopy indicated that SOD1C5-4—but not the control Aβ-targeting cyclic pentapeptides AβC5-34 or AβC5-116— interacts with SOD1(A4V), and that the time-dependent conformational transition that is indicative of SOD1(A4V) aggregation is significantly delayed in the presence of SOD1C5-4 (FIG.4B). Moreover, dynamic light scattering (DLS) analysis revealed that SOD1C5-4 addition results in the time-dependent formation of oligomeric/aggregated SOD1(A4V) species with markedly smaller sizes (FIG.4C). Detection of large, amyloid-like SOD1(A4V) aggregates by ThT staining and a filter retardation assay indicated that the formation of such species was dramatically decreased in the presence of SOD1C5-4 (FIGS.4D and4E). Finally, staining of SOD1(A4V) with the conformation-sensitive dye SYPRO Orange under heat-induced denaturation conditions, suggested that the aggregation-inhibitory action of SOD1C5-4 may be occurring due to its ability to decrease the propensity of SOD1(A4V) to expose hydrophobic surfaces (FIG.4F), a feature which has been proposed to be a molecular determinant of the pathogenesis of fALS-associated SOD1 variants (Munch C, Bertolotti A. J Mol Biol. 2010; 399(3):512-25). Taken together, these results demonstrate that SOD1C5-4 is an efficient and specific rescuer of SOD1(A4V) misfolding and aggregation. Example 4 The protective effects of SOD1C5-4 in mammalian cells were evaluated in human embryonic kidney 293 (HEK293) cells transiently expressing SOD1(A4V)-GFP. Cells treated with SOD1C5-4 exhibited higher fluorescence, fewer inclusions comprising aggregated SOD1(A4V)-GFP, and higher viability compared to untreated cells (FIGS.5A-5C). Example 5 To determine structure-activity relationships for the identified mutant SOD1-targeting cyclic oligopeptides, the sequences of the peptide-encoding regions from −5.3 million clones selected after the fourth round of FACS sorting (FIG.3B) were determined by deep sequencing. 367 distinct oligopeptide sequences appeared more than 50 times among the selected clones and were selected for subsequent analysis, which revealed the following. First, pentapeptides were the dominant peptide species within the sorted pool, with 197 of the distinct oligopeptide sequences selected corresponding to pentapeptides (54%), 148 to hexapeptides (40%) and 22 corresponding to tetrapeptides (6%) (FIG.6A). Second, the vast majority of the selected peptides exhibited the cyclo-TXSXW motif of SOD1C5-4 (˜92% of all selected clones and ˜97% of the selected pentapeptide-encoding clones (FIG.7). Third, among the selected cyclo-TXSXW pentapeptides, I, N, Q, M, E, H, and K residues were excluded at position 2, and were preferably S, A, W or F. At position 4, I, N, Q, C, D, E, K and P residues were excluded, and were preferably V, W, F, M, or H (FIGS.6B-6D). Taken together, these results indicate that the most bioactive macrocyclic structures against SOD1(A4V) misfolding and aggregation in the library are cyclic pentapeptides of the cyclo-T(Φ1,S)S(Φ2,M,H)W motif, where Φ1is preferably one of the hydrophobic (Φ) amino acids A, W or F, while Φ2is preferably V, W or F. Interestingly, selected cyclic pentapeptides belonging to this functional motif were found to be efficient in enhancing the fluorescence of SOD1-GFP containing wild-type SOD1, as well as three additional SOD1 variants, SOD1(G37R), SOD1(G85R), and SOD1(G93A), all of which are associated with familial forms of ALS, thus indicating that these peptide macroccycles are effective rescuers of the misfolding of not only SOD1(A4V), but also of other ALS-related SOD1 variants, as well as wild-type SOD1 (FIG.6E). Example 6 Combinatorial libraries of random cyclic tetra-, penta-, and hexapeptides have been selected to be studied as potential rescuers of Aβ misfolding and pathogenic aggregation. To identify cyclic oligopeptide sequences with the ability to interfere with the problematic folding of Aβ and inhibit its oligomerization/aggregation, the inventors utilized a bacterial high-throughput genetic screen. This system monitors Aβ misfolding and aggregation by measuring the fluorescence ofE. colicells overexpressing a chimeric fusion of the human Aβ(1-42) peptide (Aβ42) with GFP (US20070077552A1 “High throughput screen for inhibitors of polypeptide aggregation”). It has been demonstrated previously that due to the high aggregation propensity of Aβ,E. colicells overexpressing Aβ-GFP fusions produce misfolded fusion protein that accumulates into insoluble inclusion bodies that lack fluorescence, despite the fact that they express these fusions at high levels. Mutations in the coding sequence of Aβ or the addition of compounds that inhibit Aβ aggregation, however, result in the formation of soluble and fluorescent Aβ-GFP, and bacterial cells expressing Aβ-GFP under these conditions acquire a fluorescent phenotype. The inventors of the present invention adapted this system to perform screening for aggregation-inhibitory macrocycles in a very high-throughput fashion by isolating cyclic oligopeptide-producing bacterial clones that exhibit enhanced levels of Aβ42-GFP fluorescence using fluorescence-activated cell sorting (FACS) as also performed in a similar manner and demonstrated for SOD1 inFIG.3A. E. coliBL21(DE3) cells producing the combined cyclo-NuX1X2X3-X5library, while simultaneously overexpressing the Aβ42-GFP reporter, were subjected to FACS sorting for the isolation of clones exhibiting enhanced Aβ42-GFP fluorescence. After two rounds of sorting, the mean fluorescence of the bacterial population increased by almost three-fold compared to that of the initial library (FIG.8A). Ten individual clones were randomly picked from the sorted population and their Aβ42-GFP fluorescence was measured using a plate reader. Aβ42-GFP fluorescence of the isolated peptide-expressing clones was found to be dramatically increased compared to cells expressing the same Aβ42-GFP fusion in the presence of random cyclic peptide sequences picked from the initial (unselected) cyclo-NuX1X2X3-X5library (FIG.8B). Furthermore, the observed phenotypic effects were dependent on the ability of the Ssp DnaE intein to perform protein splicing, as the double amino acid substitution H24L/F26A in the C-terminal half of the Ssp DnaE intein, which is known to abolish asparagine cyclization at the IC/extein junction, and prevent extein splicing and peptide cyclization, was found to reduce Aβ42-GFP fluorescence back to wild-type levels (FIG.8B). Finally, the observed increases in fluorescence were found to be Aβ-specific, as the isolated pSICLOPPS-NuX1X2X3-X5vectors did not enhance the levels of cellular green fluorescence when the sequence of Aβ in the Aβ42-GFP reporter was replaced with those of two unrelated disease-associated MisPs, the DNA-binding (core) domain of the human p53 containing a tyrosine to cysteine substitution at position 220 (p53C(Y220C)) and an alanine to valine substitution at position 4 of human Cu/Zn superoxide dismutase 1 (SOD1(A4V)) (FIG.8C). On the contrary, the selected pSICLOPPS-NuX1X2X3-X5vectors were efficient in enhancing the fluorescence of Aβ-GFP containing two additional Aβ variants, Aβ40and the E22G (arctic) variant of Aβ42, which is associated with familial forms of AD (FIG.8D). Analysis of the expressed Aβ42-GFP fusions by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting revealed that the bacterial clones expressing the selected cyclic peptides produce markedly increased levels of soluble Aβ42-GFP compared to random cyclic peptide sequences, despite the fact that accumulation of total Aβ42-GFP protein remained at similar levels in all cases (FIG.8E). Furthermore, when the same cell lysates were analyzed by native PAGE and western blotting, it was observed that co-expression of the selected cyclic peptides reduced the accumulation of higher-order Aβ42-GFP aggregates, which could not enter the gel, and increased the abundance of species with higher electrophoretic mobility (FIG.8F, left). As revealed by in-gel fluorescence analysis, these higher electrophoretic mobility species correspond to the fraction of the total Aβ42-GFP that exhibits fluorescence (FIG.8F, right). Since the solubility and fluorescence of bacterially expressed Aβ-GFP has been found to be inversely proportional to the aggregation propensity of Aβ, these results suggest that Aβ aggregation is significantly decreased in the presence of the selected cyclic peptides. DNA sequencing of the peptide-encoding regions of ten isolated clones from the selected pool revealed eight distinct putative Aβ aggregation-inhibitory cyclic peptide sequences: one corresponded to a hexapeptide (TPVWFD (SEQ ID NO: 222); present twice among the sequenced clones) and seven pentapeptides (TAFDR (SEQ ID NO: 86), TAWCR (SEQ ID NO: 63), TTWCR (SEQ ID NO: 60), TTVDR (SEQ ID NO: 48), TTYAR (SEQ ID NO: 47; present twice), TTTAR (SEQ ID NO: 56), and SASPT (SEQ ID NO: 206)) (FIG.8G). Interestingly, the Arg residue at position 5, frequently encountered among the selected pentapeptides, was encoded by three different codons in the selected pSICLOPPS plasmids, thus suggesting that its dominance among the isolated clones was not coincidental. Example 7 Two of the selected cyclic peptide sequences, cyclo-TAFDR (SEQ ID NO: 86) and cyclo-SASPT (SEQ ID NO: 206), hereafter referred to as AβC5-116 and AβC5-34 (Aβ-targeting cyclic 5-peptide number 116 and 34), respectively, were chosen for subsequent analysis and were produced by solid-phase synthesis in mg quantities (FIG.9A). The inventors of the present invention chose to focus on pentapeptides, as this was the type of peptide most frequently present among the ones selected from the genetic screen. The inventors decided to further study the sequence AβC5-116 since the cyclo-TXXXR motif was particularly dominant among the selected pentapeptides, while AβC5-34 was chosen because it was the only selected pentapeptide whose sequence appeared to deviate from this motif (FIG.8G). Circular dichroism (CD) spectroscopy was first used to assess the effect of the selected pentapeptides on the aggregation process of Aβ40and Aβ42. Addition of AβC5-116 was found to strongly inhibit the aggregation of Aβ40, which remained at a random coil conformation in the presence of this cyclic peptide for extended periods of time (FIG.9B). The addition of AβC5-34 did not have the same effect and resulted in the appearance of a low-intensity negative peak (FIG.9B). When the same CD solutions were subjected to a thioflavin T (ThT) dye-binding assay that detects amyloid fibrils, we observed that Aβ40fibril formation was reduced in the presence of AβC5-116, while it remained almost unaffected by AβC5-34 (FIG.9C). In the case of Aβ42, both selected cyclic pentapeptides affected its normal aggregation pathway strongly and stabilized β-sheet-like structures (FIG.9B). ThT staining of the same samples revealed that the extent of amyloid fibril formation was greatly reduced in both cases (FIG.9C). When the cyclic peptides were added at a higher ratio, similar CD patterns were observed, however the negative peaks were much more pronounced and fibril formation was completely prevented (FIG.9C, bottom;FIG.9D). The addition of two control peptides, a randomly designed cyclic pentapeptide sequence and a cyclic pentapeptide (SODC5-4) targeting a different protein did not have any effect on the aggregation process of Aβ40and Aβ42(FIGS.9B-9C, and data not shown), thus demonstrating that cyclic peptides are not general inhibitors of Aβ aggregation and that the Aβ aggregation-modulating effect of the selected sequences relies on their cyclic nature. Transmission electron microscopy (TEM) images of solutions of Aβ42incubated without/with AβC5-34 and AβC5-116 are presented inFIG.9E. The Aβ42samples were the same as those employed in the CD and ThT studies to allow for direct correlation of findings. Aβ42incubated alone presented the typical dense network of intertwined fibrils. In the presence of the selected cyclic peptides, however, the fibrils were notably fewer, shorter and ill-developed, and the dense fibrillary network observed in their absence was not detected anywhere on the TEM grid, in agreement with the ThT data. Taken together, these results indicate that the selected cyclic oligopeptides modulate the normal aggregation process of Aβ, and their presence likely stabilizes the formation of species, which cannot develop into larger fibril-like structures. Example 8 The effects of AβC5-34 and AβC5-116 on Aβ40- and Aβ42-induced toxicity were evaluated in primary mouse hippocampal neurons. The addition of AβC5-34 and AβC5-116 was found to markedly inhibit the neurotoxicity of both Aβ40and Aβ42in a dose-responsive manner (FIG.10A). Similarly, AβC5-34 and AβC5-116 exhibited toxicity-suppressing effects also in the glioblastoma cell line U87MG (FIG.10B). On their own, AβC5-34 and AβC5-116 did not exhibit general growth-promoting effects or considerable cytotoxicity (FIGS.10A and10B). Control SOD1-targeting cyclic peptides previously found not to interfere with Aβ aggregation (FIGS.9B and9Cand data not shown), were also found ineffective in rescuing Aβ-induced cytotoxicity (FIG.10C). The effect of AβC5-34 and AβC5-116 on the morphology of Aβ-exposed neuronal cells was assessed by phase-contrast microscopy. In the presence of pre-aggregated Aβ, the population of attached cells was greatly reduced compared to the control, with many detached rounded-up cells floating in the supernatant, while hallmarks of degenerating neurons, such as cell shrinkage, membrane blebbings, fragmented neurites and ill-developed axons were obvious in the preparations (FIG.10D). The addition of the selected cyclic peptides, however, mitigated the effects of Aβ toxicity and a marked recovery of the Aβ-induced alterations was recorded (FIG.10D). Example 9 To evaluate the protective effects of the selected cyclic peptides against Aβ aggregation and toxicity in vivo, the inventors employed two established models of AD in the nematode wormCaenorhabditis elegans. The conservation of genetic and metabolic pathways betweenC. elegansand mammals, in combination with its completely characterized nervous and muscular system, its easy visualization and simple manipulation, has nominatedC. elegansas an excellent model for neurodegenerative diseases including AD, while chemical screening against Aβ-induced toxicity inC. elegansis increasingly used in AD drug discovery. The inventors performed initially a paralysis assay in CL2006, a strain where human Aβ42is constitutively expressed in the body wall muscle cells of the animals and Aβ aggregate formation is accompanied by adult-onset paralysis. Animals fed throughout their lifespan withE. coliOP50 cells producing AβC5-34 or AβC5-116 biosynthetically from their corresponding pSICLOPPS vectors, exhibited a significant delay in the appearance of the characteristic paralysis phenotype (FIG.11A). Similar protective effects were observed in a dose-responsive fashion when synthetic AβC5-34 or AβC5-116 were supplied to CL4176, a strain conditionally expressing human Aβ42under the control of a heat-inducible promoter. When chemically synthesized AβC5-34 (10 μM) and AβC5-116 (5 μM) were supplied to CL4176 animals, a significant delay in the appearance of the characteristic paralysis phenotype was recorded indicating protective effects against Aβ aggregation and toxicity (FIG.11B). To evaluate the state of Aβ aggregation in vivo, we utilized the strain CL2331, which expresses an Aβ3-42-GFP fusion in its body wall muscle cells upon temperature up-shift. Treatment with either one of the selected peptides resulted in a significant reduction of Aβ deposits (FIG.11C). Biochemical analysis of the accumulation levels of total and oligomeric Aβ levels in CL4176 worms, revealed a significant reduction of both Aβ species upon treatment with AβC5-34 and AβC5-116 (FIG.11D), an effect coinciding with the observed decelerated paralysis. Taken together, our results demonstrate that AβC5-34 and AβC5-116 exert a protective role against Aβ Example 10 To identify the functionally important residues within the isolated peptides, the inventors performed position 1 substitutions with the other two nucleophilic amino acids present in the initial libraries, as well as alanine scanning mutagenesis at positions 3-5 of the AβC5-34 and AβC5-116 pentapeptides. As judged by the ability of the generated variants to enhance the fluorescence ofE. colicells overexpressing Aβ42-GFP, AβC5-116 was found to be much more tolerant to substitutions compared to AβC5-34. All tested sequence alterations within AβC5-34 were found to be deleterious for its Aβ aggregation-inhibitory effects (FIG.12A). On the contrary, only the initial Thr and the ultimate Arg were found to be absolutely necessary for the bioactivity of AβC5-116, whereas residues at positions 3 and 4 could be substituted by Ala without significant loss of activity (FIG.12B). These observations are in line with the high frequency of initial Thr and ultimate Arg residues in the sequences of the isolated pentapeptides, as well as with the high amino acid variabilities at the corresponding positions 3 and 4, and indicates that all isolated sequences with this pattern may belong to the same consensus motif. In order to investigate this hypothesis, the inventors performed semi-saturation mutagenesis of the Ala residue of AβC5-116 with representative amino acids from all categories. Among the tested amino acids, only Thr and Ser could be tolerated at position 2 (FIG.12C), in agreement with the fact that four out of six identified pentapeptides containing the cyclo-TXXXR motif included a Thr at position 2. The results presented in the invention indicated that there should be a significant number of pentapeptide sequences with the ability to modulate Aβ aggregation that resemble AβC5-116. On the other hand, very few bioactive sequences resembling AβC5-34 should exist. To test this hypothesis and to identify all the additional bioactive cyclic oligopeptide sequences with the ability to modulate Aβ oligomerization/aggregation, the inventors turned back to the selected bacterial population exhibiting high Aβ42-GFP fluorescence (FIG.8A). The peptide-encoding vectors contained in these clones were isolated and the peptide-encoding region of approximately 5.6 million of these plasmids was sequenced using an Ion Torrent high-throughput sequencing platform. 605 distinct oligopeptide sequences appeared more than 50 times within the analyzed population, suggesting that their presence in the isolated pool is not coincidental. Indeed, cloning of four randomly chosen sequences appearing in the sorted pool only with very low frequencies, revealed that they are also efficient in increasing the fluorescence of bacterially expressed Aβ42-GFP (FIG.12F; Table 1). Analysis of the peptide sequences isolated from the genetic screen, and after considering all circular permutants thereof, revealed the following. First, pentapeptides were the dominant peptide species within the sorted pool (FIG.12D), in agreement with previous observations (FIG.8G). Second, the most prevalent motif among the selected pentapeptide sequences were TXXXR pentapeptides (˜47% of the selected pentapeptide-encoding pSICLOPPS plasmids; ˜42% of the unique selected pentapeptide sequences) (Table 1), in accordance with previous observations (FIG.8G). On the contrary, only three pentapeptide sequences were found to have high similarity with AβC5-34 (Table 2). Third, for the selected peptides corresponding to the TXXXR motif, residues at positions 3 and 4 were highly variable and included the majority of natural amino acids, with position 3 exhibiting the highest diversity (FIG.12E). At position 2, Thr, Ala, and Val were preferred, while aromatic residues (Phe, Trp, Tyr) were completely excluded from the selected TXXXR peptide pool, in full agreement with our site-directed mutagenesis studies. At the highly variable position 3, the complete absence of the negatively charged amino acids Glu and Asp among the selected sequences was notable (FIG.12E). In general, both negatively (Glu and Asp) and positively charged residues (Lys, His, and Arg) were found to be strongly disfavored among the selected TXXXR sequences at positions 2 and 3. At position 4, Ala, Asp, and Trp were found to be the preferred residues. It is noteworthy, that Lys and Gln residues were practically absent from all positions, while the β sheet-breaking amino acid Pro that is typically included in designed peptide-based inhibitors of amyloid aggregation appeared with strikingly low frequencies (FIG.12E). The motif cyclo-T(T,A,V)Ψ(A,D,W)R, where Ψ is anyone of the twenty natural amino acids excluding negatively charged ones, was found to be the most bioactive motif against Aβ in the investigated macrocycle library. The high residue variability observed at position 3 of the selected TXXXR peptides prompted the inventors to investigate whether AβC5-116 could be further minimized. Indeed, production of truncated variants of AβC5-116, from which Ala2 or Asp4 had been deleted, resulted in a respective two- and three-fold enhancement in the fluorescence of bacterially expressed Aβ42-GFP (FIG.12G). In accordance with this, a total of ten distinct cyclic tetrapeptide sequences belonging to the TXXR motif were identified among the selected peptide pool (Table 3). Taken together, our results indicate that the minimal bioactive entity against Aβ aggregation among this peptide family is a TXXR cyclic tetrapeptide, albeit with significantly reduced efficiency compared to the more privileged cyclic pentapeptide scaffold. In terms of the selected cyclic hexapeptides, sequences with an initial Threonine (T) and an ultimate Aspartic acid (D) were highly dominant among the selected pool (FIG.12H; Table 4). As in the case of the selected Aβ-targeting pentapeptides, charged amino acids were strongly disfavored among the selected sequences, with the exception of the dominant ultimate D residue. It is striking that aromatic amino acids were completely (or almost completely) absent at positions 2 and 3 of the selected hexapeptides, but highly dominant at positions 4 and 5. This sequence analysis revealed the motif cyclo-T(P,L)(V,A)WFD as the most bioactive hexapeptide motif against Aβ in the investigated macrocycle library. Example 13 Materials Synthetic human amyloid peptides Aβ40and Aβ42were purchased from Eurogentec, Belgium (>95% pure). AβC5-34 and AβC5-116 were synthesized by and purchased from Genscript (USA), while SOD1C5-4 was synthesized and purchased from CPC Scientific (USA). All DNA-processing enzymes were purchased from New England Biolabs (USA) apart from alkaline phosphatase FastAβ, which was purchased from ThermoFisher Scientific (USA). Recombinant plasmids were purified using NucleoSpin Plasmid from Macherey-Nagel (Germany) or Plasmid Midi kits from Qiagen (Germany). PCR products and DNA extracted from agarose gels were purified using Nucleospin Gel and PCR Clean-up kits from Macherey-Nagel (Germany), respectively. All chemicals were purchased from Sigma-Aldrich (USA), unless otherwise stated. Isopropyl-β-D-thiogalactoside (IPTG) was purchased from MP Biomedicals (Germany). Stock solutions of the synthetic cyclic peptides were as follows: 32.5 mM in water for AβC5-34, 10 mM in 40% DMSO for AβC5-116 and 30 mM in 40% DMSO for SOD1C5-4. Cyclic Oligopeptide Library Construction and Initial Characterization Initially, nine distinct combinatorial cyclic peptide sub-libraries were constructed: the cyclo-CysX1X2X3, cyclo-SerX1X2X3, and cyclo-ThrX1X2X3tetrapeptide sub-libraries (pSICLOPPS-CysX1X2X3, pSICLOPPS-SerX1X2X3, and pSICLOPPS-ThrX1X2X3vector sub-libraries), the cyclo-CysX1X2X3X4, cyclo-SerX1X2X3X4, and cyclo-ThrX1X2X3X4cyclic pentapeptide sub-libraries (pSICLOPPS-CysX1X2X3X4, pSICLOPPS-SerX1X2X3X4, and pSICLOPPS-ThrX1X2X3X4vector sub-libraries) and the cyclo-CysX1X2X3X4X5, cyclo-SerX1X2X3X4X5, and cyclo-ThrX1X2X3X4X5cyclic hexapeptide sub-libraries (pSICLOPPS-CysX1X2X3X4X5, pSICLOPPS-SerX1X2X3X4X5, and pSICLOPPS-ThrX1X2X3X4X5vector sub-libraries). These vectors express libraries of fusion proteins comprising four parts: (i) the C-terminal domain of the split Ssp DnaE intein (IC), (ii) a tetra-, penta-, or hexapeptide sequence, (iii) the N-terminal domain of the split Ssp DnaE intein (IN), and (iv) a chitin-binding domain (CBD) under the control of the PBADpromoter and its inducer L(+)-arabinose (FIG.1A). The libraries of genes encoding these combinatorial libraries of random cyclic oligopeptides were constructed using degenerate primers. Cys, Ser, and Thr were encoded in these primers by the codons UGC, AGC, and ACC, respectively, which are the most frequently utilized ones for these amino acids inE. coli, while the randomized amino acids (X) were encoded using random NNS codons, where N=A, T, G, or C and S=G or C. A second PCR reaction was conducted in each case to eliminate mismatches. The resulting PCR products were digested with BglI and HindIII for 5 h and inserted into the similarly digested and dephosphorylated auxiliary vector pSICLOPPSKanR (see below). The ligation reactions were optimised at a 12:1 insert:vector molar ratio and performed for 4 h at 16° C. Approximately 0.35, 0.7 and 3.5 μg of the pSICLOPPSKanR vector were used for each one of the tetra-, penta- and hexapeptide libraries, respectively. The ligated DNA was then purified using spin columns (Macherey-Nagel, Germany), transformed into electro-competent MC1061 cells prepared in-house, plated onto LB agar plates containing 25 μg/mL chloramphenicol and incubated at 37° C. for 14-16 h. This procedure resulted in the construction of the combined pSICLOPPS-NuX1X2X3-X5library with a total diversity of about 31,240,000 independent transformants, as judged by plating experiments after serial dilutions. Colony PCR of 124 randomly selected clones with intein-specific primers revealed that 88 of them (˜71%) contained the correct insert. Overexpression of the tetra-partite fusion in 150 randomly selected clones using 0.002% arabinose and monitoring of the production of this fusion protein by western blotting using a mouse anti-CBD primary antibody (New England Biolabs, USA; 1:100,000 dilution) and a goat anti-mouse HRP-conjugated secondary antibody (Bio-Rad, USA; 1:4,000 dilution), showed that 99 of them (˜66%) produced high yields of the tetra-partite fusion protein. Among these 99 clones that produced precursor fusion protein (molecular mass ˜25 kDa), 82 clones (˜55% of total clones tested) also yielded a lower molecular weight band (molecular mass ˜20 kDa), which corresponds to one of the splicing reaction products, the N-terminal domain of the Ssp DnaE intein fused to CBD (IN-CBD), after intein splicing and cyclic peptide formation takes place. Therefore, according to these results, the generated bacterial libraries encoding for cyclic tetra-, penta- and hexapeptide contain approximately 20,760,000 clones, which express tetra-partite peptide fusions at high levels and which are capable of undergoing splicing and potentially yielding cyclic peptide products. This diversity covers fully the theoretical diversity of our combined cyclo-NuX1X2X3, NuX1X2X3X4and NuX1X2X3X4X5libraries (3×203+3×204+3×205=10,104,000) by more than two-fold (FIG.1B). Expression Vector Construction For the construction of pETSOD1-GFP, the human SOD1 cDNA was generated by PCR-mediated gene assembly. The assembled gene was further amplified by PCR and the resulting product was digested with Ndel and BamHI, and inserted into similarly digested pAβ42-GFP vector GFP (Wurth C, Guimard N K, Hecht M H., J Mol Biol. 2002; 319(5):1279-90), in the place of Aβ42. For pETSOD1(A4V)-GFP, SOD1 was amplified by PCR from the pETSOD1-GFP vector using the mutagenic forward primer GS059 and the reverse primer GS060. The resulting PCR product was then digested with Ndel and BamHI, and inserted into similarly digested pETAβ42-GFP. For pETSOD1(G37R)-GFP, pETSOD1(G85R)-GFP and pETSOD1(G93A)-GFP construction, SOD1 was mutated by overlap extension PCR starting from pETSOD1-GFP as a template. All SOD1 PCR products were then digested with Ndel and BamHI, and inserted into similarly digested pETAβ42-GFP vector. For the construction of pETSOD1, pETSOD1(G37R), pETSOD1(G85R) and pETSOD1(G93A), the corresponding SOD1 genes were amplified by PCR from pETSOD1-GFP, pETSOD1(G37R)-GFP, pETSOD1(G85R)-GFP and pETSOD1(G93A)-GFP, respectively. For the construction of pETSOD1(A4V), SOD1 was amplified from pETSOD1(A4V)-GFP. All SOD1 PCR products were digested with XbaI and BamHI, and cloned into similarly digested pET28a(+) (Novagen). For the construction of the pSICLOPPS vectors encoding for variants of the selected AβC5-34 and AβC5-116 peptides, the auxiliary pSICLOPPSKanR vector was generated initially. pSICLOPPSKanR was constructed by PCR amplification of the gene encoding aminoglycoside 3′-phosphotransferase (KanR—the enzyme conferring resistance to the antibiotic kanamycin) from pET28a(+), digestion with BglI and HindIII and insertion into similarly digested pSICLOPPS. For the construction of the vectors pSICLOPPS-AβC5-34(S1C), pSICLOPPS-AβC5-34(S1T), pSICLOPPS-AβC5-34(S3A), pSICLOPPS-AβC5-34(P4A) and pSICLOPPS-AβC5-34(T5A), mutagenic PCR was carried out starting from pSICLOPPS-AβC5-34, followed by digestion of the generated product with BglI and HindIII and insertion into similarly digested pSICLOPPSKanR. The vectors pSICLOPPS-AβC5-116(T1C), pSICLOPPS-AβC5-116(T1S), pSICLOPPS-AβC5-116(F3A), pSICLOPPS-AβC5-116(D4A), pSICLOPPS-AβC5-116(R5A), pSICLOPPS-AβC5-116(A2F), pSICLOPPS-AβC5-116(A25), pSICLOPPS-AβC5-116(A2P), pSICLOPPS-AβC5-116(A2T), pSICLOPPS-AβC5-116(A2Y), pSICLOPPS-AβC5-116(A2H), pSICLOPPS-AβC5-116(A2K), pSICLOPPS-AβC5-116(A2E), pSICLOPPS-AβC5-116(A2W), pSICLOPPS-AβC5-116(A2R), pSICLOPPS-AβC5-116(A2del), pSICLOPPS-AβC5-116(F3del) and pSICLOPPS-AβC5-116(D4del) were generated in a similar fashion. Cyclic Oligopeptide Library Screening ElectrocompetentE. coliBL21(DE3) cells (Novagen, USA) carrying either the expression vector pETSOD1(A4V)-GFP, which produces SOD1(A4V)-GFP under control of the strong bacteriophage T7 promoter, or pETAβ42-GFP, which produces Aβ42-GFP under control of the T7 promoter, were co-transformed with the combined pSICLOPPS-NuX1X2X3-X5vector library. Approximately 108 transformants carrying both the vector library and either pETSOD1(A4V)-GFP or pETAβ42-GFP vectors were harvested, pooled together, and grown in Luria-Bertani (LB) liquid medium containing either 0.005% (pETSOD1(A4F)-GFP) or 0.002% (pETAβ42-GFP) L-arabinose—the inducer of cyclic peptide production—at 37° C. with shaking. When the optical density at 600 nm (OD600) of the bacterial culture was about 0.5, 0.01 (pETSOD1(A4F)-GFP) or 0.1 (pETAβ42-GFP) mM isopropyl-β-D-thiogalactoside (IPTG) was added to the medium to induce overexpression of the reporter. After about two hours at 37° C., ˜108cells were screened and the population exhibiting the top 1-3% fluorescence was isolated using FACS (BD FACSAria, BD Biosciences, USA). The isolated cells were re-grown and screened for additional rounds in an identical manner until the desired enrichment in high-fluorescence clones was achieved. Protein/Cyclic Peptide Production in Liquid Cultures E. colicells freshly transformed with the appropriate expression vector(s) were used for protein production experiments in all cases. Single bacterial colonies were used to inoculate overnight liquid LB cultures containing the appropriate antibiotics for plasmid maintenance (100 μg/mL ampicillin, 40 μg/mL chloramphenicol (Sigma, USA)) at 37° C. These cultures were used with a 1:100 dilution to inoculate fresh LB cultures in all cases. For SOD1 or SOD1-GFP production, BL21(DE3) (Novagen, USA) or Origami 2(DE3) cells (Novagen, USA) were transformed with the corresponding SOD1- or SOD1-GFP-encoding vector, either with the appropriate pSICLOPPS vector or alone. Cells were grown in 5 mL liquid LB cultures containing 50 μg/mL kanamycin (or 100 μg/mL ampicillin for pASK75-based vectors), 40 μg/mL chloramphenicol (for cell cultures carrying also a pSICLOPPS vector), 200 μM CuCl2, 200 μM ZnCl2and 0.005% arabinose (for cell cultures carrying also a pSICLOPPS vector) at 37° C. to an OD600of ˜0.3-0.5 with shaking, at which point SOD1 or SOD1-GFP production was induced by the addition of 0.01 mM IPTG (0.2 μg/mL anhydrotetracycline (aTc) for pASK-based vectors) for 2-3 h. For Aβ42-GFP production, BL21(DE3) cells were transformed with pETAβ42-GFP and the appropriate pSICLOPPS vector. Cells were grown in 5 mL liquid LB cultures containing 50 μg/mL kanamycin, 40 μg/mL chloramphenicol and 0.02% arabinose at 37° C. to an OD600of ˜0.3-0.5 with shaking, at which point Aβ42-GFP production was induced by the addition of 0.1 mM IPTG for 2-3 h. Bacterial Cell Fluorescence Bacterial cells corresponding to 1 mL culture with OD600=1 were harvested by centrifugation and re-suspended in 100 μL phosphate-buffered saline (PBS), transferred to a 96-well FLUOTRAC 200 plate (Greiner Bio One International, Austria), and their fluorescence was measured using a TECAN Safire II-Basic plate reader (Tecan, Austria). Excitation was set at 488 nm and emission was measured at 510 nm. High-Throughput Sequencing Analysis For the characterization of the initial libraries, a combined pSICLOPPS-NuX1X2X3-X5vector library was prepared containing approximately equal amounts of each one of the tetra-, penta- and hexapeptide sub-libraries. These samples were digested with NcoI and BsrGI and the resulting ˜250 bp product that contained the variable peptide-encoding region was isolated. High-throughput sequencing analysis was performed using an Ion Torrent high-throughput sequencing platform. From the obtained data, all the sequences with mismatches outside of the variable peptide-encoding region were removed, and only the 12-, 15- or 18-bp-long peptide-encoding sequences were subjected to further analysis. The libraries of the selected cyclic peptides that enhance either SOD1(A4V)-GFP or Aβ42-GFP fluorescence were sequenced in a similar manner, with the only exception being that all sequences including stop codons were discarded from subsequent analysis. Protein Electrophoresis and Western Blot Analysis Bacterial cells corresponding to 1 mL culture with OD600=1 were harvested by centrifugation and re-suspended in 200 μL PBS. Samples were lysed by brief sonication for 10 s on ice twice. These lysates (total lysate fraction) were then centrifuged at 13,000×g for 10 min, the supernatant was collected (soluble fraction) and the pellet was re-suspended in 200 μL PBS (insoluble fraction). For analysis by SDS-PAGE, samples were boiled for 5 min and 10 μL of each sample were loaded onto 12% or 15% gels. For analysis by native PAGE, 10-20 μL of each sample were loaded onto SDS-free 10% gels without prior boiling. In-gel fluorescence was analyzed on a ChemiDoc-It2Imaging System equipped with a CCD camera and a GFP filter (UVP, UK), after exposure for 3-5 sec. For western blotting, proteins were transferred to polyvinylidene fluoride (PVDF) membranes (Merck, Germany) for 50 min at 12 V on a semi-dry blotter (Thermo Fisher, USA). Membranes were blocked with 5% non-fat dry milk in Tris-buffered saline containing 0.1% Tween-20 (TBST) for 1 h at room temperature. After washing with TBST three times, membranes were incubated with the appropriate antibody dilution in TBST containing 0.5% non-fat dried milk at room temperature for 1 h. The utilized antibodies are described in SI Materials and Methods. The proteins were visualized using a ChemiDoc-It2Imaging System (UVP, UK). The utilized antibodies were a mouse monoclonal, horseradish peroxidase (HRP)-conjugated anti-polyhistidine antibody (Sigma, USA) at 1:2,500 dilution, a mouse monoclonal anti-FLAG (Sigma, USA) at 1:1,000 dilution, a mouse anti-GFP at 1:20,000 dilution (Clontech, USA), a mouse anti-Aβ (6E10) (Covance, USA) at 1:2,000 dilution, a mouse anti-CBD (New England Biolabs, USA) at 1:25,000 or 1:100,000 dilution, and a HRP-conjugated goat anti-mouse antibody (Bio-Rad, USA) at 1:4,000. Preparation of SOD1 Stocks and Solutions SOD1 or mutants thereof were overexpressed from the appropriate pET-SOD1 or pASK-SOD1 vectors inE. coliOrigami 2(DE3) cells in LB medium containing 50 μg/mL kanamycin (for pET-SOD1) or 100 μg/mL ampicillin (for pASK-SOD1), 200 μM CuCl2, and 200 μM ZnCl2by the addition of 0.01 mM IPTG (for pET-SOD1) or 0.2 μg/mL anhydrotetracycline (aTc) (for pASK-SOD1), either at 37° C. for 2-3 h or at 18° C. for about 16 h. Origami 2(DE3) cells were utilized in order to provide an oxidizing cytoplasmic environment in order to promote correct formation of disulfide bonds, which are required for proper SOD1 folding and function. Under these conditions, bacterially produced SOD1 is produced in dimeric and enzymatically active form, while it simultaneously co-exists with misfolded, soluble and insoluble SOD1 oligomeric/aggregated species (FIG.3C). Thus, the acquired protein is found in a state that resembles the conditions encountered in human cells under stressful or pathogenic conditions. The appearance of misfolded SOD1 oligomers/aggregates is enhanced with increasing incubation temperatures. Thus, for assays that are more appropriate for monitoring the early steps of SOD1 oligomerization/aggregation, such as dynamic light scattering (DLS), we utilized SOD1 produced at 18° C., whereas for assays that are more appropriate for monitoring the later steps of SOD1 aggregation, such as filter retardation, ThT staining and CD spectroscopy, we utilized SOD1 produced at 37° C. Preparation of Aβ Stocks and Solutions Synthetic Aβ40and Aβ42peptides were gently dissolved without vortexing in doubly deionized water to a final concentration of 100 μM. These solutions were then diluted by PBS addition (10 mM, pH 7.33) to achieve a final Aβ concentration of 50 μM. Circular Dichroism Appropriate amounts of synthetic cyclic peptides were added to either 40 μM SOD1(A4V) or 50 μM Aβ solutions at the desired cyclic peptide:target protein molar ratio. SOD1(A4V) structural changes were monitored for 90 d at 25° C., under quiescent conditions. Aβ structural changes were monitored for 30 d at 33° C. under quiescent conditions. CD spectra in the range 190-260 nm were recorded on a JASCO J-715 spectropolarimeter (Jasco Co., Japan) using quartz cuvettes with 1 mm path length. Each reported spectrum is the average of three scans at a rate of 100 nm·min−1and a resolution of 0.5 nm. Dynamic Light Scattering The sizes of the SOD1 particles were measured using a Zetasizer NanoZS90 (Malvern) instrument. After a 2-min temperature-equilibration step at 37° C., eighteen consecutive 10-s measurements, per sample, were averaged to produce the particle size (Z average) distributions. Thioflavin T Staining 40 μM SOD1(A4V) solutions, aged for 90 d at 25° C., with or without the selected synthetic peptides, were diluted to 10 μM with PBS. 5 μL from a stock solution of ThT (Sigma-Aldrich, USA) in PBS (10 mM, pH 7.33) was added to these SOD1(A4V) solutions to achieve a final ThT concentration of 10 μM. The mixture was agitated adequately by pipetting and immediately thereafter, fluorescence was monitored with excitation at 440 nm (EM slit=2.5 nm, PMT Voltage 700 V, response 0.4 s) using a HITACHI F-2500 (Japan) spectrofluorometer. For Aβ ThT staining, 100 μL of the 30-d aged 50 μM CD solutions were diluted in PBS (10 mM, pH 7.33) to form a 25 μM Aβ solution with 200 μL final volume. 2.5 μL from a stock solution of ThT (Sigma-Aldrich, USA) in PBS (10 mM, pH 7.33) was added to the prepared Aβ solutions to achieve a final ThT concentration of 5 μM. The mixture was agitated adequately by pipetting and immediately thereafter, fluorescence was monitored with excitation at 440 nm (EM slit=2.5 nm, PMT Voltage 700 V, response 0.4 s) using a HITACHI F-2500 (Japan) spectrofluorometer. Filter Retardation Assay SOD1(A4V) solutions (10 μM), incubated in the presence or absence of the selected cyclic peptides for 25 d at 37° C., were mixed with a stock solution of SDS to achieve a final SDS concentration of 2% and then boiled for 10 min. These samples were subsequently applied under vacuum on a 0.2 μm-pore size PVDF membrane (Merck), which had been previously equilibrated with transfer buffer containing 0.1% SDS, and then washed twice with 100 μl TB S under vacuum. The membrane was blocked with 5% non-fat dry milk in TBST for 1 h at room temperature and then stained with a HRP-conjugated anti-polyHis antibody at a 1:2,500 dilution (Sigma-Aldrich) overnight at 4° C. SOD1 Aggregation and Viability Measurements in HEK293 Cells Human embryonic kidney (HEK) 293 cells were transfected using a Nucleofector (Amaxa) following the manufacturer's protocol. 6 ug DNA (SOD1 or SOD1(A4V) cloned into the pEGFP-N3 plasmid vector) were used per 2×106 cells and 5 μM synthetic SOD1C5-4 was added, where appropriate, before plating. Transfected cells were sorted 18 h later on a FACSAria to isolate GFP-positive clones. 4′,6-Diamidino-2-phenylindole dihydrochloride (DAPI) dye was used to exclude dead cells. ˜28% of the SOD1 and ˜15% of the SOD1(A4V) total cells were found to be GFP-positive. Collected cells were plated onto a 24-well plate at a density of 50,000 cells/well. Microscopy analysis was performed under an inverted microscope on day 1 and day 5 in culture after sorting. Cell counts are the average number of viable GFP-fluorescing cells of two areas per triplicate of wells of 24-well plates (magnification 20×). Cell counts are presented as percentage of viability of SOD1-overexpressing cell. As aggregate-positive cells are counted the fluorescing inclusion body-positive cells. Again, two areas per triplicate of wells of 24-well plate are averaged (magnification 20×). Aggregate-positive cells are presented as percentage of the total viable GFP-fluorescing cells. Transmission Electron Microscopy (TEM) For TEM analysis, the 30-d aged 50 μM CD solutions of Aβ42(with 100 μM of the selected peptides or without) was mixed well by pipetting. 2 μL of this solution were placed in a carbon-coated film on 200-mesh copper grids (Agar Scientific, UK) for 5 min. After adsorption, grids were washed in deionized water and negatively stained by applying a 2-μl drop of freshly prepared 1% (w/v) uranyl acetate (Sigma-Aldrich, USA) in Milli-Q water for 5 min. Excess fluid was blotted off, and grids were washed in deionized water and dried in air. Images were recorded using a FEI CM20 electron microscope (FEI, USA) with a Gatan GIF200 imaging filter (Gatan, USA), equipped with a Peltier-cooled slow-scan CCD camera. Neuronal Cell Cultures The media/agents for primary neuronal cell cultures were purchased from Thermo Fisher Scientific (USA). Hippocampal neuronal cultures were obtained from postnatal day 1 female pups of C57BL/6 mice. Briefly, after being dissected, the hippocampus was incubated with 0.25% trypsin for 15 min at 37° C. The hippocampi were then rinsed in 10 mL of Hibernate containing 10% (v/v) heat-inactivated fetal bovine serum (FBS). Cultures were maintained in Neurobasal-A medium containing 2% B-27 supplement, 0.5 mM Gluta-MAX and 1% penicillin/streptomycin at 37° C. and 5% CO2. Half of the medium was replaced twice a week. Neuronal hippocampal cells were plated at a density of approximately 2×104per well in 96-well plates and 5×105per well in 24-well plates for MTT and induced cell death assays, respectively. After seven days of incubation in culture well plates, the primary hippocampal neurons were used for the cell viability measurements. The utilized U87MG cells (human glioblastoma-astrocytoma, epithelial-like cell line) were kind a gift from Dr. Maria Paravatou-Petsotas, Radiobiology Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, NC SR “Demokritos”. The utilized media/agents for U87MG cell cultures were obtained from Biochrom AG (Germany) and PAA Laboratories (USA). U87MG cells were grown in Dulbecco's modified Eagle medium (DMEM), supplemented with 10% fetal bovine serum (FBS), 2.5 mM L-glutamine, 1% penicillin/streptomycin at 37° C. and 5% CO2. For MTT cytotoxicity studies, cells were plated at a density of 2×104cells per well in 96-well plates and incubated at 37° C. for 24 h to allow cells to attach. The medium was subsequently removed and cells were rendered quiescent by incubation in serum-free medium for 24 h. For cell viability measurements cells were subsequently treated with the indicated concentrations of Aβ in the presence or absence of synthetic peptides, as described in Materials and Methods. Cell Viability Measurements Solutions of synthetic Aβ40or Aβ42(10 μM) in PBS, preincubated at 37° C. (3 d for Aβ40solutions and 1 d for Aβ42solutions) in the presence or absence of synthetic cyclic peptides (1:1 and 2:1 ratio of peptides:Aβ), were diluted with fresh medium and transferred into wells at a 1 μM final Aβ concentration. Cell viability was determined using the MTT assay. MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) was purchased from Applichem (Germany). After 24 h of exposure to Aβ solutions, 100 μL of a 0.5 mg/mL stock solution of MTT in Neurobasal-A was added to each well of primary hippocampal neurons followed by a 3 h incubation at 37° C., while 100 μL of a 1 mg/mL stock solution of MTT in DMEM complete medium was added to each well of U87MG cells followed by a 4 h incubation at 37° C. The medium was then removed and the cells were diluted in DMSO. The relative formazan concentration was measured by determination of the absorbance at 540 nm using a plate reader (Tecan, Austria). Results were expressed as the percentage of MTT reduction, assuming that the absorbance of control (untreated) cells was 100%, and represent the mean of three independent experiments with six replicate wells for each condition. Induced cell death was also qualitatively examined by phase-contrast microscopy (Carl Zeiss, Axiovert 25 CFL, Germany) using the above solutions. In each run, the effect of solutions of plain synthetic peptides and plain Aβ40or Aβ42was independently checked to serve as internal control. In Vivo Assays inC. elegans Strains We followed standard procedures forC. elegansstrains maintenance at 16° C. The following strains were used: CL2179: dvIs179 [myo-3p::GFP::3′UTR(long)+rol-6(su1006)] (available on the world wide web at: cgc.cbs.umn.edu/strain.php?id=26134); CL2331: dvIs37 [myo-3p::GFP::Aβ(3-42)+rol-6(su1006)] (available on the world wide web at: cgc.cbs.umn.edu/strain.php?id=26135); CL4176: smg-1(cc546) I; dvIs27 [myo-3::Aβ(1-42)-let 3′UTR(pAF29); pRF4 (rol-6(su1006)] (available on the world wide web at: cgc.cbs.umn.edu/strain.php?id=7663). Treatment with Cyclic Peptides For treatments with synthetic cyclic peptides, nematodes were exposed to the indicated AβC5-34 and AβC5-116 concentrations per NGM plate. Stock solutions of the two chemically synthesized pure peptides were obtained after dissolution in DMSO and stored at −20° C. The appropriate amount of compound or DMSO (control cultures) was added onto anE. coliOP50 bacterial lawn. Synchronized offspring were randomly distributed to treatment plates to avoid systematic differences in egg lay batches. Treatment and control plates were handled, scored and assayed in parallel. Paralysis Assay Synchronized CL4176 animals (150-300 animals per condition) were transferred to NGM plates containing synthetic AβC5-34, AβC5-116 or 0.26% DMSO at 16° C. for 48 h before transgene induction via temperature up-shift to 25° C. Synchronized offspring were randomly distributed to treatment plates to avoid systematic differences in egg lay batches. Treatment and control plates were handled, scored and assayed in parallel. Scoring of paralyzed animals was initiated 24 h after temperature up-shift for the CL4176 strain. Nematodes were scored as paralyzed upon failure to move their half end-body upon prodding. Animals that died were excluded. Plates were indexed as 1, 2, 3 etc by an independent person and were given to the observer for scoring in random order. The index was revealed only after scoring. Dot Blot Analysis CL4176 animals were allowed to lay eggs for 3 h on NGM plates containing either synthetic peptides or 0.26% DMSO. Paralysis was induced upon temperature up-shift and the progeny were exposed to either pure peptides or 0.26% DMSO until 50% of the control population was paralyzed. The animals were then collected and boiled in non-reducing Laemmli buffer. For dot blot analysis, 1-5 μg of protein lysates were spotted onto 0.2 μm nitrocellulose membranes (Bio-Rad, USA) after soaking into TBS pre-heated at 80° C. Immunoblotting was performed using the anti-Aβ antibody 6E10 (recognizes total Aβ) and the anti-amyloid protein, oligomer-specific antibody AB9234 (Merck Millipore, Germany). Actin was used as a loading control. Blots were developed with chemiluminescence by using the Clarity™ Western ECL substrate (Bio-Rad, USA). Quantification of the ratio of each detected protein to actin using the anti-actin antibody sc-1615 (Santa Cruz, Germany), and normalization to control appears next to each representative blot. Confocal Microscopy Analysis For Aβ3-42deposit measurements, synchronized (at the L4 larval stage) CL2331 and CL2179 (control strain) animals exposed to solvent (0.26% DMSO), 10 μM AβC5-34 or 5 μM AβC5-116 and grown at 20° C. (to induce aggregation) until day 2 of adulthood were collected. Animals were mounted onto 2% agarose pads on glass slides, anesthetized with 10 mM levamisole and observed at RT using a Leica TCS SPE confocal laser scanning microscope (Leica Lasertechnik GmbH, Germany). The LAS AF software was used for image acquisition. At least twenty animals/condition in three independent experiments were processed. Images of whole worms and focused images in the posterior area of nematodes were acquired with 10×0.45 and 20×0.70 numerical aperture, respectively. While the invention has been described with respect to specific embodiments, it is apparent that modifications are possible without departing from the scope of the invention.
168,889
11858975
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS Explanation of Terms: The term “diabetes” includes Type 1 diabetes, Type 2 diabetes, gestational diabetes, and other symptoms that cause hyperglycemia. This term is used for metabolic disorders, in which the pancreas cannot produce enough insulin, or the body cells fail to respond to insulin properly, which leads to a decrease in glucose absorption efficiency of tissue cells and causes glucose to accumulate in the blood. Type 1 diabetes, also known as insulin-dependent diabetes and juvenile-onset diabetes, is caused by the β cells destruction and usually results in absolute insulin deficiency. Type 2 diabetes, also known as non-insulin-dependent diabetes and adult-onset diabetes, is generally associated with insulin resistance. The term “obesity” refers to excess fatty tissue, caused by excess calories stored in fat when energy intake exceeds energy consumption. In this article, individuals with a body mass index (BMI=body weight (kg) divided by the square of height (m)) exceeding 25 are considered obese. Incretin is a gastrointestinal hormone that regulates blood glucose by enhancing glucose-stimulated insulin secretion (also known as glucose-dependent insulin secretion, GSIS) (Drucker.D J, Nauck, M A, Lancet 368: 1696-705, 2006). Incretin can also slow down the rate of nutrient absorption by delaying gastric emptying, and directly reduce food absorption. At the same time, Incretin can also inhibit intestinal a cells from secreting Glucagon. Hitherto, there are two known types of incretin: glucagon-like peptide-1 (GLP-1) and Glucose-dependent insulinotropic polypeptide (GIP). PreproGlucagon is a precursor polypeptide consisting of 158 amino acids. PreproGlucagon is differentially processed in tissues to form a variety of structurally related proglucagon-derived peptides, including Glucagon, Glucagon-like peptide-1 (GLP-1), Glucagon-like peptide-2 (GLP-2) and Oxyntomodulin (OXM). GIP is a 42-amino acid peptide obtained by the proteolytic processing of 133-amino acid precursor (pre-pro-GIP). These molecules are involved in various biological functions, including glucose homeostasis, insulin secretion, gastric emptying and intestinal growth, and food intake regulation. The sequence of Glucagon-like peptide (GLP-1) is shown in SEQ ID NO: 1; GLP-1 is a 30- or 31-amino acid polypeptide intestinal incretin hormone secreted from intestinal L-cells. GLP-1 includes two active forms of GLP-1 (7-36) and GLP-1 (7-37). GLP-1 is released into the circulation after a meal and exerts its biological activity by activating GLP-1 receptors. GLP-1 has various biological effects, including stimulation of glucose-dependent insulin secretion, inhibition of glucagon production, delay of gastric emptying, and appetite suppression (Tharakan G, Tan T, Bloom S. Emerging therapies in the treatment of ‘diabesity’: beyond GLP-1. Trends Pharmacol Sci 2011; 32 (1): 8-15). Native GLP-1 is readily degraded by dipeptidyl peptidase-4 (DPP-IV), neutral endopeptidase (NEP), plasma kallikrein or plasmin, which limits its therapeutic potential. Since native GLP-1 has a short half-life of about 2 minutes in vivo, chemical modifications and/or formulations design are usually considered to improve therapeutic efficacy of GLP-1 derived medicines for diabetes and obesity (Lorenz M, Evers A, Wagner M. Recent progress and future options in the development of GLP-1 receptor agonists for the treatment of diabesity. Bioorg Med Chem Lett 2013; 23 (14): 4011-8; Tomlinson B, Hu M, Zhang Y, Chan P, Liu ZM. An overview of new GLP-1 receptor agonists for type 2 diabetes. Expert Opin Investig Drugs 2016; 25 (2): 145-58). Oxyntomodulin is a small peptide of 37 amino acids, the sequence is shown in SEQ ID NO: 2; Oxyntomodulin contains the complete 29 amino acid sequence of Glucagon (SEQ ID NO: 42). Oxyntomodulin is a dual-agonist for GLP-1R and GCGR, and is secreted together with GLP-1 through intestinal L-cells after a meal. Similar to Glucagon, Oxyntomodulin induces significant weight loss in humans and rodents. The weight-loss activity of oxyntomodulin has been compared with equimolar doses of GLP-1R mono-agonists in obese mice. Oxyntomodulin showed an anti-hyperglycemic effect compared with GLP-1R mono-agonists, and significantly induced weight loss and lipid decrease (The Glucagon receptor is involved in mediating the body weight-lowering effects of oxyntomodulin, Kosinski J R, etc., Obesity (Silver Spring), 20): 1566-71, 2012). Overweight and obese subjects receiving subcutaneous administration of oxyntomodulin over a 4-week period resulted in an average weight loss of 1.7 kg. Oxyntomodulin is also proved to reduce food intake and increase energy expenditure in humans (Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects: a double-blind, randomized, controlled trial, Wynne K et al., Diabetes, 54: 2390-5, 2005; Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomized controlled trial; Wynne K et al., Int J Obes (Lond), 30: 1729-36, 2006). But likewise, Oxyntomodulin has a short half-life due to low molecular weight and degradation by DPP-IV. Currently, dual GLP-1Rand GCGR agonists are generally derived from Oxyntomodulin. Mutations (Oxyntomodulin analogue) are introduced to improve defect of Oxyntomodulin, including short half-life and incapacity to resisting enzymatic hydrolysis. Most of the mutations substituting the second serine (Ser) to a-aminoisobutyric acid (Aib), a non-natural amino acid contributes to resistance to DPP-IV cleavage. Although Oxyntomodulin analogues have shown initial hypoglycemic and fat-reducing effects, the exact mechanism remains unclear. The Oxyntomodulin receptor has not been found. Currently, only results from GCGR- or GLP-1R-knockout mice or cell bioassaies have provided evidences for Oxyntomodulin's functions through binding to the two receptors. Glucagon is a 29-amino acid peptide, which corresponds to amino acids at position 53-81 of preproGlucagon, the sequence is shown in SEQ ID NO: 42 (CG Fanelli et al., Nutrition, Metabolism & Cardiovascular Diseases (2006) 16, S28-S34). Glucagon receptor activation has been shown to increase energy consumption and reduce food intake in both rodents and humans (Habegger K M et al., The metabolic actions of Glucagon revisited, Nat. Rev. Endocrinol. 2010, 6,689-697), and the effects are stable and persistent in rodents. Glucagon has many physiological effects, such as increasing blood glucose levels in hypoglycemic conditions by stimulating glycogen breakdown and gluconeogenesis, regulating the production of liver ketone, regulating the bile acid metabolism and vagus nerve-through satiety effects. Glucagon is indicated for acute hypoglycemia clinically. Glucagon receptor activation reduces food intake, and promotes fat breakdown and weight loss in animals and humans. The term “receptor agonist” may be defined as a polypeptide, protein, or other small molecules that bind to a receptor and triggers a reasonable response as its native ligand. “GLP-1 receptor (GLP-1R) agonist” may be defined as a polypeptide, protein, or other small molecules that bind to GLP-1R and trigger the same or similar response as native GLP-1. GLP-1R agonists activate GLP-1R completely or partially, and then cause a series of downstream signaling pathway reactions inside the cell to produce corresponding biological activity, such as β cells secreting insulin. Typical GLP-1R agonists include native GLP-1 and mutants and analogues thereof, such as Exenatide, Liraglutide and the like. GLP-1 analogues: as used herein, “GLP-1 analogues” or “GLP-1 mutants” all mean GLP-1R agonists and may be used interchangeably. Glucagon receptor (GCGR) agonist: Glucagon receptor agonist, which may be defined as a polypeptide, protein or other small peptides that bind to GCGR and can initiate the same or similar characteristic response as native Glucagon. GCGR agonists activate GCGR completely or partially, and then cause a series of downstream signaling pathway reactions inside the cell to produce corresponding biological activity, such as glycogenolysis of hepatocytes, gluconeogenesis, fatty acid oxidation and ketogenesis. Glucagon analogues: as used herein, “Glucagon analogues”, “GCG analogues”, “Glucagon mutants” and “GCG mutants” all mean Glucagon receptor agonists and may be used interchangeably. GCGR/GLP-1R dual-agonist active peptide: the GCGR/GLP-1R dual-agonist active peptide of the present disclosure includes proteins or polypeptides that can simultaneously stimulate GLP-1R and GCGR. Such as the Oxyntomodulin-based dual-agonist as reported by Alessandro Pocai et al. (Glucagon-Like Peptide 1/Glucagon Receptor Dual Agonism Reverses Obesity in Mice, Diabetes; 58 (10): 2258-2266, 2009) or the dual-agonist based on Glucagon as reported by Richard D. DiMarchi et al. (US9018164 B2). Herein, “dual-agonist” or “bispecific active protein” or “dual-effective protein” are synonymous. FGF21 (Fibroblast growth factor 21), FGF15/19 and FGF23 belong to the “endocrine” hormone of the FGF family. FGF21 is an important hormone that regulates glucose and lipid metabolism. Unlike insulin, FGF21 promotes glucose uptake in adipocytes by up-regulating the expression of GLUT1. The binding of FGF21 to the receptor requires the assistance of the transmembrane protein β-Klotho, which stimulates signal transduction by binding to the FGFR/β-Klotho receptor complex, to trigger the biological effects of liver, adipose tissue, and pancreas. β-Klotho is exclusively expressed in pancreas, liver, and adipose, which also explains the specificity of FGF21 on these tissues (Kurosu H et al., Tissue-specific expression of beta Klotho and fibroblast growth factor (FGF) receptor isoforms determines the metabolic activity of FGF19 and FGF21. J Biol Chem 282: 26687-26695, 2007; Kharitonenkov A et al., (2008b) FGF-21/FGF-21 receptor interaction and activation is determined by beta Klotho. J Cell Physiol 215: 1-7). In the presence of the co-receptor β-Klotho, FGF21 can bind to and activate three FGFR subtypes (1c, 2c and 3c). Other FGFR subtypes, such as FGFR1b, FGFR2b and FGFR3b are not considered as FGF21 receptors due to inability to form complexes with β-Klotho Evidence suggests that among FGFR receptors binding to FGF21, FGFR1 plays a dominant role in the regulation of FGF21 activity. The N-terminal and C-terminal of FGF21 are very important for functional activity, where the N-terminal binds to FGFR and the C-terminal binds to β-Klotho (Micanovic R, et al (2009) Different roles of N- and C-termini in the functional activity of FGF21. J Cell Physiol 219: 227-234). The mouse FGF21 protein is composed of 210 amino acids with a N-terminal 30-amino acid signal peptide, while human FGF21 protein is 209-amino acid in length with a N-terminal signal peptide of 28 amino acids. Human FGF21 shares about 75% sequence identity with mouse FGF21. FGF21 is mainly expressed in pancreatic β cells, liver, WAT, skeletal muscle, and shows obvious tissue specificity. Human FGF21 is readily degraded by prolinase (FAP, a serine protease) in vivo, with a half-life of 30 minutes in mice and 2 hours in monkeys. Multi-domain: a domain is a region in a biological macromolecule that has a specific structure and independent function, especially in a protein. In a globular protein, a domain has its own specific tertiary structure and functions independently of the rest of the protein molecule. Different domains in the same protein often be connected by short linker sequences without secondary structure. Individual domains in a protein make up a multi-domain. In the present disclosure, multi-domain refers to a fusion protein containing GCG analogues, FGF21 or FGF21 analogues and FC, which have GCGR agonist activity, GLP-1R agonist activity and FGF21 activity. Dimer: A dimer in the present disclosure is formed by the native non-covalent and covalent action of the constant region (FC) of the immunoglobulin. If not otherwise specified, the dimers formed by FCare all homodimers, as described in the dimers provided by the present disclosure. The active protein described in Formula I will form a dimer due to the presence of FC. Tri-agonist active protein: herein, “tri-agonist active protein”, “three-activity agonist active protein”, and “trispecific dimer active protein” are all synonymous and can be used interchangeably. EC50(concentration for 50% of maximal effect) refers to the concentration required for a drug or substance to stimulate 50% of its corresponding biological response. The lower the EC50value, the stronger the stimulation or agonism of the drug or substance. More intuitively, for example, the stronger the intracellular signal caused, the better the ability to induce the production of a hormone. Cell-Based Bioactivity Assay A luciferase reporter assay was used to determine the in vitro GLP-1R and GCGR agonist activities in a cell-based bioactivity assay in present disclosure. The luciferase reporter assay is based on the principle that GLP-1R and GCGR can activate the downstream cAMP pathway after activation. Bioactivity determination of FGF21 and its analogues was obtained by detecting fluorescence signal change in a CHO cell co-transfected with β-klotho and FGF21R genes. According to the report of Joseph R. Chabenne et al. and Richard D. DiMarchi et al., adding a C-terminal small peptide Cex (SEQ ID NO: 3 GPSSGAPPPS) from Exendin-4 to the C-terminal of Glucagon increased the GLP-1R agonist activity by about 2 times (Optimization of the Native Glucagon Sequence for Medicinal Purposes, J Diabetes Sci Technol, 4 (6): 1322-1331, 2010 and patent US9018164 B2), but the ratio of GCGR agonist activity to GLP-1R agonist activity was only about 35: 1. In addition, Evers A et al. reported that (Evers A et al., Design of Novel Exendin-Based Dual Glucagon-like Peptide 1 (GLP-1)/Glucagon Receptor Agonists, J Med Chem.; 60 (10): 4293-4303. 2017) after adding the Cex sequence to the C-terminal of GCG analogue, GLP-1R agonist activity decreased by about 3 times, and GCG activity decreased by about 14 times (Table 2, peptides 7 and 8 in the article). In an embodiment of the present disclosure, when a GCG analogue containing GPSSGAPPPS (SEQ ID NO. 3) or a similar sequence was further fused to a an Fcdomain, GLP-1R agonist activity increased by a staggering 200 times (EC50of about 1.1 nM). While for the GCG analogues disclosed in US9018164 B2 and other patents and literatures by Joseph R. Chabenne et al., the ratio of GLP-1R agonist activity changed only about 2 times before and after adding GPSSGAPPPS (SEQ ID NO. 3) or a similar sequence (For example, GLP-1R agonist activity of native Glucagon relative to native GLP-1 in paper was 0.7%, but increased to 1.6% after adding the GPSSGAPPPS (SEQ ID NO. 3) sequence). That is, addition of GPSSGAPPPS (SEQ ID NO. 3) sequence to the Glucagon polypeptide C-terminally did not necessarily increase GLP-1R agonist activity significantly. Stability of the Multi-Domain Active Protein There are several sensitive cleavage sites in native Glucagon, including position 2 recognized by DPP-IV, and SRR at positions 16-18. Although several reporters speculated that FCimprove the chemical stability and serum stability of some active protein, the role of FCto GLP-1 or Glucagon analogues which N-terminal must be exposed seems to be inconclusive. After fusion of native GLP-1 or Glucagon with FCfragment, N-terminal degradation was still obviously observed under 37 ° C. in serum. To improve stability, the present disclosure introduces a mutation that is resistant to protease hydrolysis on the basis of native Glucagon. After the fusion of the mutant with FC, the stability is further improved. At present, almost all GCGR/GLP-1R dual-agonists derived from Oxyntomodulin and Glucagon have introduced mutations that resist to DPP-IV cleavage at position two. For example, the mutation of L-amino acid to D-amino acid (L-Ser mutated to D-Ser), or the introduction of an unnatural amino acid such as Aib (Matthias H. Tschop, etc., Unimolecular Polypharmacy for Treatment of Diabetes and Obesity, 24:51-62, 2016). However, in the embodiments of the present disclosure, the dual-agonist active protein derived from Formula II and retaining native L-Ser in position two exhibits very high serum stability, without any sign of significant degradation by DPP-IV at 24 hours, while the corresponding peptides without FCfusion were rapidly hydrolyzed by DPP-IV (Table 4). The inventors prepared an active protein COO1L13F8(SEQ ID NO.93) in which native Glucagon was fused with FC, and an active protein C002L13F8(SEQ ID NO.94) in which Glucagon-cex was fused with FCaccording to the report of Joseph R. Chabenne et al. The two active proteins serve as a control to verify whether FCfusion did improve stability. However, neither COO1L13F8(SEQ ID NO.93) nor C002L13F8(SEQ ID NO.94) showed obvious signs of resistance to DPP-IV cleavage. Although some reports have suggested that binding to serum albumin (such as HSA) may help improve protein stability (such as liraglutide), half-life was still shorter than 12 hours if position two remained unchanged. That is, it is impossible to support a dosing frequency of once-a-week. The pharmacokinetic and pharmacodynamic profiles have shown that the GCG analogues provided by the present disclosure are sufficient to support a dosing frequency of once-a-week rather than once a day as generally reported (for example, albumin-binding liraglutide). The retention of native amino acids further reduces immunogenicity risk, avoids chemical crosslinking and makes the preparation process easier and more convenient. Intraperitoneal Glucose Tolerance Test (IPGTT) In an embodiment, an IPGTT experiment was conducted. Mice administered with multi-domain active protein showed extremely stable blood glucose fluctuations after injecting glucose. Weight Loss and Appetite Control in DIO Mice and Pharmacokinetic Studies GCGR agonists have been reported to have a potential effect on weight loss. However, the therapeutic use of native Glucagon is limited by its rapid degradation and low molecular weight. At present, most Glucagon analogues are used for acute hypoglycemia symptoms potentially. Clinical reports of long-acting GCG analogues for weight loss in diabetic patients are also emerging. It is well known that obesity is one of the causes of insulin resistance in diabetic patients, and weight loss is an important indicator to evaluate a glucose-lowering drug. In addition, the multi-domain active protein of the present disclosure induces a significant weight loss after administration to DIO mice. The roles of GCGR/GLP-1R dual-agonist, FGF21 and its analogues in blood glucose control and lipid metabolism is widely known. Stanislaus S et al. (Stanislaus S et al., A novel Fc FGF21 with improved resistance to proteolysis, increased affinity towards β-Klotho and enhanced efficacy in mice and cynomolgus monkeys Endocrinology. 2017 May 1; 158 (5): 1314-1327.) has reported that FC-FGF21 analogues at a dose of 3 mg/kg (about 30 nM/kg) resulted in a weight loss of more than 15% after 4 weeks of continuous administration. The tri-agonist active proteins (dose of 30 nM/kg) In Embodiment 9 of the present disclosure triggered a weight loss of approximately 30% with appetite basically unchanged. Likewise, a dose of 10 nM/kg triggered a weight loss about 15%. In addition, the combination (A-La-F plus F-Lb-B) of the dual-agonist active protein and the long-acting FGF21 analogue in Embodiment 10 also played a synergistic effect: the combination administered reduced body weight by 35% or more. However, the dual-agonist active proteins with equivalent dose only reduced body weight by no more than 13%, while the long-acting FGF21 analogues only reduced body weight by less than 10%. Prospect of Clinical Application Clinically, the multi-domain active proteins of the present disclosure are potentially suitable for once weekly administration or longer according to its pharmacokinetic profiles. The doses selected depend on dosing frequency and route, age, gender, weight, physiological state of the subjects, treatment regimen and pathological conditions any associated diseases and other known to the skilled in the art. At the same time, according to the subjects' physiological state and pathological conditions, the multi-domain active proteins of the present disclosure may be administered or applied in combination with one or more other therapeutically active compounds or substances. For example, the above-mentioned other therapeutically active compounds available include, but are not limited to, anti-diabetic drugs, anti-hyperlipidemia drugs, anti-obesity drugs, anti-hypertensive drugs, and reagents for the treatment of complications arising from or related to diabetes. Metabolic syndrome is associated with an increased risk of coronary heart disease and other conditions related to the accumulation of vascular plaque, such as stroke and peripheral vascular disease, which become an atherosclerotic cardiovascular disease (ASCVD). Patients with metabolic syndrome may progress from an early insulin resistance state to fully mature Type 2 diabetes, accompanied by an increased risk of ASCVD. Not to be limited to any particular theory, the relationship between insulin resistance, metabolic syndrome and vascular disease may involve one or more common pathogenesis, including insulin-stimulated vasodilation disorder, decreased availability of insulin resistance-related correlations due to increased oxidative stress, and abnormalities of adipocyte-derived hormones (such as adiponectin) (Lteif, Mather, Can. J. Cardiol. 20 (Suppl B): 66B-76B, 2004). The active proteins provided by the present disclosure may be used to treat obesity. In some aspects, the active proteins of the present disclosure treats obesity by reducing appetite, decreasing food intake, lowering body fat level, increasing energy consumption and other mechanisms in patients. In some potential embodiments, the active proteins of the present disclosure can be used for the treatment of non-alcoholic fatty liver disease (NAFLD). NAFLD refers to broad-spectrum liver diseases, ranging from simple fatty liver (steatosis) to non-alcoholic steatosis hepatitis (NASH) to liver cirrhosis (irreversible late stage of scarring of the liver). All stages of NAFLD show fat accumulation in liver cells. Simple fatty liver is an abnormal accumulation of certain types of fat and triglycerides in the liver cells, but without signs of inflammation or scar formation. In NASH, fat accumulation is associated with varying degrees of liver inflammation (hepatitis) and scar formation (fibrosis). Inflammatory cells may destroy liver cells (hepatocyte necrosis). In the terms “Steatosis hepatitis” and “Steatosis necrosis”, steatosis refers to fatty infiltration, hepatitis refers to inflammation in the liver, and necrosis refers to destroyed liver cells. NASH may eventually lead to liver scarring (fibrosis) and then result in irreversible advanced scarring (liver cirrhosis). Liver cirrhosis caused by NASH is the final and the most severe stage of disease within the NAFLD spectrum. Before further describing the specific embodiments of the present disclosure, it is understood that the scope of the present disclosure is not limited to the specific embodiments described below. It is also to be understood that the terminology of the disclosure is used to describe the specific embodiments, and not to limit the scope of the disclosure. The test methods without specific conditions noted in the following embodiments are generally based on conventional conditions or the conditions recommended by the manufacturers. When the numerical values are given by the embodiments, it is to be understood that the two endpoints of each numerical range and any one between the two may be selected unless otherwise stated. Unless otherwise defined, all technical and scientific terms used in the present disclosure have the same meaning as commonly understood by one skill in the art. In addition to the specific methods, equipments and materials used in the embodiments, any method, equipments and materials in the existing technologies similar or equivalent to the methods, equipments and materials mentioned in the embodiments of the present disclosure may be used to realize the invention according to the grasp of the existing technologies and the record of the invention by those skilled in the art. Unless otherwise stated, the methods of experiments, assays and sample preparation disclosed in the present invention all employ conventional techniques of molecular biology, biochemistry, chromatin structure and analysis, analytical chemistry, cell culture, recombinant DNA technology in the technical field and related fields. These techniques are well described in the prior literatures. For details, see Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, Second edition, Cold Spring Harbor Laboratory Press, 1989 and Third edition, 2001; Ausubel et al, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, 1987 and periodic updates. The series METHODS IN ENZYMOLOGY, Academic Press, San Diego; Wolffe, CHROMATIN STRUCTURE AND FUNCTION, Third Edition, Academic Press, San Diego, 1998; METHODS IN ENZYMOLOGY, Vol. 304, Chromatin (P. M. Wassarman and A. P. Wolffe, eds.), Academic Press, San Diego, 1999; and METHODS IN MOLECULAR BIOLOGY, Vol. 119, Chromatin Protocols (P. B. Becker, ed.) Humana Press, Totowa, 1999, and the like. Embodiment 1 Screening of GCG Analogues (Screening of Glucagon Analogue) The amino acid sequence of native GLP-1 is shown in SEQ ID NO. 1, specifically: HAEGTFTSDVSSYLEGQAAKEFIAWLVKGRG. The amino acid sequence of native Oxyntomodulin is shown in SEQ ID NO. 2, specifically: HSQGTFTSDYSKYLDSRRAQDFVQWLMNTKRNRNNIA. The GCG analogue of the present disclosure is denoted as A. The A is a GCGR/GLP-1R dual-agonist active peptide, selected from any peptide that process GCGR and GLP-1R dual-agonist activity. The structure of A is shown in Formula II: HSQGTFTSD-X10-S-X12-X13-X14-X15-X16-X17-X18-X19-X20-X21-F-X23-X24-WL-X27-X28-X29-X30-Xz. The X10is selected from V, L or Y; the X12is selected from S, E or K; the X13is selected from Y or Q; the X14is selected from L or M; the X15is selected from D or E; the X16is selected from S, E or G; the X17is selected from R, E or Q; the X18is selected from R, E or A; the X19is selected from A or V; the X20is selected from Q, R or K; the X21is selected from D, L or E. The X23is selected from V or I; the X24is selected from Q, A or E; the X27is selected from M, K or V; the X28is selected from N or K; the X29is selected from G or T; the X30is G or missing. The Xzdoes not exist or is selected from any one of SEQ ID NO. 3-8. The amino acid sequences of the exemplary GCG analogues may be independently selected from SEQ ID NO. 42-92, and the corresponding polypeptide codes are C001, C002, C240, C241, C276, C225, C222, C163, C164, C271, C368, C495, C353, C352, C355, C382, C232, C227, C266, C137, C399, C398, C396, C392, C462, C228, C187, C363, C364, C209, C289, C611, C618, C623, C627, C654, C673, C563, C549, C555, C487, C488, C489, C503, C508, C711, C708, C743, C756, C788 and C731, respectively. Embodiment 2 Preparation of Dual-Agonist Active Protein A-La-F In this embodiment, the dimeric dual-agonist active protein A-La-F is obtained by fusing the GCG analogue with the peptide linker Laand F. The A is the same as the A in Embodiment 1. The F is a long-acting protein unit, which may be selected from a complete FCportion of an immunoglobulin, a fragment of an FCportion of an immunoglobulin, or a mutant of an FCportion of an immunoglobulin. The amino acid sequence of F is shown in SEQ ID NO. 9-18, and the corresponding abbreviations are F1-F10, respectively. Lais a flexible polypeptide with an appropriate length consisting of glycine (G), serine (S) and/or alanine (A), so that adjacent protein domains can move freely relative to each other. A longer peptide linker may be used if two adjacent domains spatially interfering with each other. The exemplary peptide linkers are (GS)n, (GGS)n, (GGSG)n, (GGGS)nA (GGGGS)nA and (GGGGA)nA, and n is an integer of 1-10. The exemplary peptide linkers may be independently selected from SEQ ID NO. 19-41, and the corresponding codes of the sequence are L1-L23, respectively. The amino acid sequences of the dual-agonist active proteins moiety, A-La-F, may be independently selected from SEQ ID NO. 93-133, and the DNA sequences may be independently selected from SEQ ID NO. 222-262, the corresponding codes of the dual-agonist active proteins of A-La-F are COO1L13F8, C002L13F8, CG283L13F8, C240L13F8, CG214L13F8, C382L13F8, CG267L13F8, C276L13F8, C308L13F8, C368L13F8, C224L13F8, C225L13F8, CG308L13F8, C495L13F8, C319L13F8, C364L13F8, C214L13F8, C232L13F8, C303L13F8, C392L13F8, CG303L13F8, C462L13F8, C240 L12F8, C368 L10F4, C364 L10F4, C352 L13F4, C225 L10F10, C228L13F4, C187 L9F4, C618 L12F4, C623 L9F3, C228 L13F4, C498 L13F4, C503 L15F2, C508 L7F4, C289 L1F4, C756 L20F4 C209 L13F8 and C627 L12F10, respectively. The skilled in the art can prepare the A-La-F using the traditional technologies on the basis of knowing the amino acid sequences of the A-La-F: due to the presence of FCsequence, Protein A chromatography with high affinity and high specificity for Fc can be used for protein purification. A feasible preparation method is given here illustratively. The Preparation Process is as Follows: (1) DNA sequences design according to protein sequences and amino acid codons. Polynucleotide DNA fragments corresponding to A, Laand F in the recombinant proteins were prepared respectively. Individual DNA fragment can be synthesized and spliced by conventional solid-phase synthesis technologies. (2) Primer design for nested PCR amplification: DNA splicing for the corresponding fragments of A, La, and F to obtain the target genes and PCR splicing (including primer design, PCR introduced mutation and enzyme digestion, etc.) are known technologies for the skilled in the art. It's known to the skilled in the art that the PCR splicing method in this embodiment is not exclusive, target genes may also be obtained through gene synthesis. Target genes were cloned into mammalian cell expression vector pTT5 (Yves Durocher) and transformed intoE. coliTop10F′. Identified positive clones were inoculated into 500 ml LB medium and incubated overnight, followed by cell collection by centrifugation, and finally plasm ids were exacted by OMEGA E.Z.N.A.® Endo-Free Plasmid Maxi Kit. (3) Transfection of Hek293F cells and expression: 1.0 mg plasmid was diluted to 25 ml Freestyle 293 expression medium (Thermofisher). 3.0 mg PEI (linear, 25KD) was diluted to 25 ml Freestyle 293 medium and mixed well with the plasmid solution, followed by incubation at room temperature for 30 minutes. At the same time, Hek293F cells in log phase (viability>95%) was counted, followed by centrifugation at 1100 rpm for 10 minutes. After discarding the supernatant, resuspending the cells pellet in 450 ml Freestyle 293 expression medium. After incubation of the PEI-plasmid mixture, adding the PEI-plasmid mixture into the cell suspension and shake-culturing at 37 ° C., 5% CO2, and 140 RPM. After 7 hours, the Freestyle 293 expression medium was replaced with 1000 ml 293 SFM II medium (Thermofisher) followed by culture for 7 days. (4) Purification of the recombinant protein: cell culture was centrifuge at 8000 rpm for 10 min to collect the supernatant. The supernatant was then loaded onto a Protein A column (Bestchrom (Shanghai) Biotechnology Co., Ltd.) pre-equilibrated with equilibration buffer (20 mM PB, 0.5M NaCl, pH7), and eluted in 100% elution buffer (0.1M Gly-HCl, pH3.0). Eluted sample was collected in a collection tube prefilled with neutralization buffer (1M Tris-HCl, pH 8.0). The final neutralization buffer was 1/10 of the volume of the eluted samples, and protein concentration was determined by Bradford method. (5) identification of the physicochemical properties of the recombinant proteins: SDS-PAGE electrophoresis or amino acid sequence verification results of the purified recombinant protein were consistent with expectations. Embodiment 3 Preparation of Tri-Agonist Active Protein The tri-agonist active proteins of the present disclosure contains multiple domains and has tri-agonist activity. The structure of the tri-agonist active proteins is shown in Formula I: A-La-F-Lb-B. A is a GCGR/GLP-1R dual-agonist agonist active peptide, F is a long-acting protein unit, B is a native FGF21 or FGF21 analogue, Laand Lbare peptide linkers. In Formula I, A is the same as the A in Embodiment 1. In Formula I, F may be selected from a complete FCportion of an immunoglobulin, a fragment of an FCportion of an immunoglobulin, or a mutant of an FCportion of an immunoglobulin, as shown in SEQ ID NO.9-18. In formula I, B is native FGF21 (SEQ ID NO. 136) or FGF21 analogue. The structure of B is as follows: HPIPDSSPLLQFGGQVRQX19YLYTDDAQQTEX31HLEIX36EDGTVGX43AX45DQSPESLLQLX56ALKPGVIQILGVKTSRFLCQRPDGALYGSLHFDPEACSFREX98LLEDGYNVYQSEAHGLPLHX118PGNX122SPHRDPAPRGPX134RFLPLPGLPPALPEPPGILAPQPPDVGSSDPLX167MVX170X171SQX174RSPSX179X180X181. The N terminal HPIPDSS may be missing or partially missing; X19is selected from R, Y,V,Eor C; X31is selected from A or C; X36is selected from R or K; X43is selected from G or C; X45is selected from A, K, E or V; X56is selected from K, R, V or I; X98is selected from L, R or D; X118is selected from L or C; X122is selected from K or R; X134is selected from A or C; X167is selected from S, A or R; X170is selected from G or E; X171is selected from P or G; X174is selected from G, A or L; X179is selected from Y, A or F; X180is selected from A or E; X181is selected from S, K or is missing. The FGF21 analogue is an active protein that has the same or similar biological function as native FGF21 (SEQ ID NO. 136) and shares a sequence identity above 80% with native FGF21 (SEQ ID NO. 136). Preferably, the FGF21 analogue shares a sequence identity above 85% with native FGF21 (SEQ ID NO. 136); More preferably, the FGF21 analogue shares a sequence identity above 90% with native FGF21 (SEQ ID NO. 136); More preferably, the FGF21 analogue shares a sequence identity above 95% with native FGF21 (SEQ ID NO. 136). Preferably, the FGF21 analogue is shown in SEQ ID NO. 137-148. Ladoes not exist or is a peptide linker, Lbdoes not exist or is a peptide linker. When Laand Lbare peptide linkers, the peptide linkers are the same as the Lain Embodiment 2. Based on the GCGR/GLP-1R dual-agonist active protein, FGF21 or FGF21 analogue is fused at the C-terminal of FCby a peptide linker, to prepare the exemplary tri-agonist active proteins with amino acids as shown in SEQ ID NO.149-208 and nucleotides as shown in SEQ ID NO.263-322. The corresponding codes of the tri-agonist active protein are COO2L13F8L10W, C240 L12F8L12M1, C240 L9F7L13M2, C240 L13F4L9M1, C240 L9F2L13M3, C240 L13F10L9M2, C225 L10F10L14M2, C163 L13F8L13M2, C271 L9F4L8M2, C368 L10F4L10M2, C495 L13F8L13M1, C495 L13F8L10M2, C495 L9F10L9M1, C353 L13F3L10M4, C352 L13F4L9M3, C382 L9F3L9M2, C382 L10F2L13M2, C382 L13F10L9M1, C382 L13F8L10M2, C382 L12F7L9M2, C382 L14F4L9M2, C232 L9F3L10M3, C227 L12F5L14M4, C266 L13F7L13M4, C137 L10F8L9M5, C399 L12F4L19M8, C392 L11F7L10M5, C462 L9F2L9M10, C462 L10F5L10M3, C462 L11F8L13M1, C462 L13F4L10M2, C462 L13F8L10M2, C462 L13F10L9M4, C228 L13F4L13M12, C187 L9F4L12M7, C364 L10F4L12M8, C209 L13F8L13M9, C289 L12F4L8M10, C611 L11F4L11M11, C618 L13F7L13M1, C618 L12F4L12M2, C623 L9F3L10M1, C623 L9F8L9M2, C627 L12F10L9M6, C654 L13F9L6M3, C673 L8F3L8M8, C563 L14F8L9M3, C549 L12F4L9M4, C555 L10F6L12M1, C487 L13F7L13M4, C488 L9F9L9M2, C498 L13F4L13M4, C503 L15F2L9M5, C508 L7F4L9M3, C711 L13F5L10M4, C708 L10F4L14M7, C743 L18F7L8M10, C756 L20F4L10M8, C788 L1F5L5M5 and C731 L5F2L9M9, respectively. A-La-F-Lb-B can be prepared by the skilled in the art using the existing technologies on the basis of knowing the amino acid sequence. Due to the presence of FCsequence, Protein A resin chromatography with high affinity and high specificity can be used for protein purification. The specific method may refer to the preparation method in Embodiment 2. Purified recombinant proteins were performed on SDS-PAGE electrophoresis or amino acid sequence verification, and the results were consistent with expectations.FIG.1indicates SDS-PAGE results of some purified samples. Embodiment 4 Preparation of FGF21 Analogues Fused with FC In this Embodiment, an FC(code: F9) is fused with the native FGF21 and an FGF21 analogue (code: M1-M12) to obtain a long-acting FGF21 analogue. The structure of the long-acting FGF21 analogue is shown as: F-Lb-B. F may be selected from a complete FCportion of an immunoglobulin, a fragment of an FCportion of an immunoglobulin, or a mutant of an FCportion of an immunoglobulin. The amino acid sequence of F may be shown in any one of SEQ ID NO. 9-18. Lbdoes not exist or is a peptide linker. When Lbis a peptide linker , the peptide linker is the same as the Lain Embodiment 2. The peptide linker includes units rich in G, S and/or A, for example, (GS)n, (GGS)n, (GGSG)n, (GGGS)nA, (GGGGS)nA, (GGGGA)nA, and n is an integer of 1-10. In a preferred embodiment, the amino acid length of the peptide linker is 5-26. Further, the amino acid sequence of the peptide linker may be shown in any one of SEQ ID NO. 19-41. The B is native FGF21 (SEQ ID NO. 136) or FGF21 analogue. The B is the same as the B in Embodiment 3. The amino acid sequence of the long-acting FGF21 analogue may be as shown in SEQ ID NO.209-221, respectively, the codes are F9L10W, F9L10M1, F9L10M2, F9L10M3, F9L10M4, F9L10M5, F9L10M6, F9L10M7, F9L10M8, F9L10M9, F9L10M9, F9L10M11 and F9L10M12, respectively. Synthesizing DNAs according to the protein sequences and subcloning the DNAs into a recombinant expression vector is a conventional method in the technical field, and the transfection of Hek293F cells and cell expression are the same as in Embodiment 2. Similarly, due to the presence of the FCsequence, the separation and purification process may also refer to Embodiment 2. Embodiment 5 In Vitro Cell-Based Bioactivity Assay The dual-agonist active proteins obtained in Embodiment 2 were subjected to in vitro cell-based bioactivity assays, including GLP-1R agonist activity assay and GCGR agonist activity assay. GLP-1R Agonist Activity Assay: A luciferase reporter assay was used to determine the in vitro GLP-1R agonist activity (Jonathan W Day et al.: Nat Chem Biol. 2009 October; 5 (10): 749-57). A human GLP-1R gene was cloned into the mammalian cell expression plasmid pCDNA3.1 to construct a recombinant expression plasmid pCDNA3.1-GLP-1R, and the full-length luciferase gene was cloned into pCRE plasmid to obtain a pCRE-Luc recombinant plasmid. CHO cells were transfected with pCDNA3.1-GLP-1R and pCRE-Luc plasmids at a molar ratio of 1:10, and stably transfected strains were selected to obtain recombinant CHO/GLP-1R stably transfected cell strains. Cells were cultured using DMEM/F12 medium containing 10% FBS and 300 μg/ml G418 in a 9-cm cell culture dish. When the cells reached about 90% confluence, the supernatant was discarded. 2m1 Trypsin was added to digestion for 3min was added for digestion for 3min followed by addition of 2 ml DMEM/F12 medium containing 10% FBS and 300 μg/ml G418 for neutralizing. After transferring to a 15 ml centrifuge tube, the cells were centrifuged at 1000 rpm for 5 min and the supernatant was discarded, followed by addition of 2 ml DMEM/F12 medium containing 10% FBS and 300 μg/ml G418 for resuspending, and finally counted. The cells were diluted to 3×105per ml with DMEM/F12 medium containing 10% FBS and aliquots of 100 ul were seeded into each well of a 96-well plate, i.e. 5×104per well. The cells were cultured in DMEM/F12 medium containing 0.2% FBS after adherence. After discarding the supernatant of the cells in the 96-well plate, the purified recombinant proteins (Table 1, Table 2) or native Glucagon (Hangzhou Chinese Peptide Biochemical Co., Ltd, GLUC-004) and native GLP-1 (Hangzhou Chinese Peptide Biochemical Co., Ltd., GLUC-016B) as positive controls were diluted with DMEM/F12 medium containing 0.1% FBS to a series of specified concentrations, followed by addition to cell culture (100111/well). Luciferase signal was recorded after stimulating for 6h. The determination was carried out according to the instructions of Luciferase reporter kit (Ray Biotech, Cat: 68-LuciR-S200). GCGR Agonist Activity Assay: The luciferase reporter assay was also used in determination of GCGR agonist activity. The GCGR gene is cloned into mammalian cell expression plasmid pCDNA3.1 to construct a recombinant expression plasmid pCDNA3.1-GCGR. The recombinant expression plasmid pCDNA3.1-GCGR is co-transfected with the pCRE-Luc recombinant plasmid into HEK 293T cells, and the stably transfected cell strains HEK 293T/GCGR are screened and constructed. FGF21 Activity Assay: The assay for FGF21 activity determination was performed using similar methods as in the literature with some proper modifications (Xu J etc., Polyethylene glycol modified FGF21engineered to maximize potency and minimize vacuole formation, Bioconjug Chem.; 24 (6): 915-25, 2013). The puromycin resistance gene pac was amplified by PCR and cloned into pcDNA3.1(+) to replace the original G418 resistance gene. The GAL4DBD-ELK1, IRES, and KLB (β-klotho) genes were amplified by PCR and cloned into the pcDNA-Puro plasmid in sequence to construct the plasmid pcDNA-GAL4DBD-ELK1-IRES-KLB-Puro for cell transfection and screening. The plasmid was extracted by OMEGA E.Z.N.A.® Endo-Free Plasmid Midi Kit. The process of cell transfection was as follows: Hek293T cells were plated in a 6-well plate, 3×105cells per well, and cultured overnight. The cells were washed twice with Opti-MEM medium, followed by addition of 2 ml Opti-MEM medium. Mixture for cell transfection was prepared according to the following proportion: Lipofectamine 2000 (6 μl): pFR-Luc (4.6 μg): pcDNA-GAL4DBD-ELK1-IRES-KLB-Puro (1 μg). The mixture was slowly added to a 6-well plate and mixed well after incubation for 20 minutes. After culturing for 6 h, the medium was replaced with DMEM medium plus 10% FBS, followed by culturing at 37° C. with 5% CO2. Screening was performed to obtain stably transfected cell strains respond to FGF21 stimulation. When full confluence in the dish was observed, the cells were digested with Trypsin to prepare a cell suspension (1×105cells/ml, DMEM +5% FBS +1 μg/ml puromycin), followed by plating in a 96-well plate (100 μl per well) and culturing overnight. Fluorescence signals were recorded using the Luciferase Reporter Assay Kit (68-LucifR-5200) after samples with gradient concentrations were added for stimulation for 6 h. The results of the activity assays of some dual-agonist active proteins are shown in Table 1 and Table 2: TABLE 1Codes ofGLP-1Rdual-agonistEC50(nM)agonist activityactive proteinsSEQ ID NO.GCGRGLP-1RratioaC001L13F8937.98380.3227C002L13F8948.1613.89CG283L13F8951.24330.41215C240L13F8961.451.54CG214L13F8971.12360.49206C382L13F8981.331.75CG267L13F8991.15360.87214C276L13F81001.231.69C308L13F81011.09409.20244C368L13F81021.511.68C224L13F81031.11335.2211C225L13F81041.361.59CG308L13F81051.16350.22237C495L13F81061.501.48C319L13F81071.44378.12209C364L13F81081.571.81C214L13F81090.97437.83226C232L13F81101.341.94C303L13F81111.03389.12224C392L13F81121.671.74CG303L13F81131.19452.48214C462L13F81141.382.11Glucagon421.4bGlucagon Cex43Glucagon422.3cGlucagon Cex43 Notes: The codes of proteins in table are named in accordance with the following rules: polypeptide code+peptide linker code +FCcode. For example, C240L13F4indicates that the C240 polypeptide is fused with an IgG FCcodenamed F4) via a peptide linker codenamed L13. a is the ratio of GLP-1R agonist activity before and after the insertion of GPSSGAPPPS (SEQ ID NO: 3) or similar sequence (also known as sequence Cex, selected from SEQ ID NO.3-8 in the present disclosure) between the GCG analogue and the Fc. b is the ratio calculated based on the GLP-1R agonist activity of native Glucagon and Glucagon Cex disclosed in Table 2 of U.S. Pat. No. 9,018,164 B2. c is the ratio calculated based on the GLP-1R agonist activity of native Glucagon and Glucagon Cex disclosed in Table 1 in a paper of Joseph r. Chabenne et al. (Joseph R. Chabenne etc., Optimization of the Native Glucagon Sequence for Medicinal Purposes, J Diabetes Sci Technol. 4(6): 1322-1331, 2010). As shown in Table 1 and Table 2, when a sequence containing Cex (selected from SEQ ID NO.3-8 in the present disclosure) is fused with F (SEQ ID NO.16) via (GGGGS)3A (SEQ ID NO.31) to form a dimer, the GLP-1R agonist activity is increased by more than 200 times, while the GCGR agonist activity shows no significant change. TABLE 2Codes of dual-effectEC50(nM)active proteinsSEQ ID NO.GCGRGLP-1RC240L12F81151.331.46C368L10F41161.421.72C364L10F41171.481.65C352L13F41181.231.75C225L10F101191.391.47C228L13F41201.521.81C187L9F41211.201.53C618L12F41221.271.32C623L9F31231.171.26C228L13F41241.511.87C498L13F41251.882.01C503L15F21261.782.13C508L7F41271.451.77C756L20F412821.2313.70C788L1F512926.6618.54C289L12F41302.012.54C611L11F41311.191.32C209L13F81321.632.41C627L13F101331.271.41 Results of Activity Assay of Tri-Agonist Active Proteins The activity results of the tri-agonist active proteins prepared in Embodiment 3 are shown in Table 3: TABLE 3AminoGCGRGLP-1RacidagonistagonistFGF21sequenceactivityactivityactivityCodes of active(SEQ(EC50,(EC50,(EC50,proteinsID NO.)nM)nM)nM)Native Glucagon420.94120.87Native GLP-11>10000.52Native FGF211360.12C002L13F8L10W1491.221.340.55C240 L12F8L12M11501.201.130.79C240 L9F7L13M21511.301.170.83C240 L13F4L9M11521.161.260.77C240 L9F2L13M31531.341.311.36C240 L13F10L9M21541.251.210.89C225 L10F10L14M21551.171.420.85C163 L13F8L13M21561.371.280.93C271 L9F4L8M21571.181.330.87C368 L10F4L10M21581.251.300.74C495 L13F8L13M11591.131.330.82C495 L13F8L10M21601.211.300.71C495 L9F10L9M11611.431.210.84C353 L13F3L10M41621.111.361.67C352 L13F4L9M31631.151.211.45C382 L9F3L9M21641.351.180.81C382 L10F2L13M21651.211.190.87C382 L13F10L9M11661.271.130.76C382 L13F8L10M21671.331.210.91C382 L12F7L9M21681.421.300.88C382 L14F4L9M21691.221.250.76C232 L9F3L10M31701.451.790.65C227 L12F5L14M41711.381.550.71C266 L13F7L13M41721.341.370.69C137 L10F8L9M51731.771.790.78C399 L12F4L19M81741.821.890.96C392 L11F7L10M51751.461.670.85C462 L9F2L9M101761.341.211.54C462 L10F5L10M31771.221.231.37C462 L11F8L13M11781.171.410.78C462 L13F4L10M21791.271.320.88C462 L13F8L10M21801.631.210.58C462 L13F10L9M41811.231.111.33C228 L13F4L13M121821.381.271.37C187 L9F4L12M71831.691.880.79C364 L10F4L12M81841.271.870.73C209 L13F8L13M91851.451.741.13C289 L12F4L8M101861.381.771.36C611 L11F4L11M111871.391.101.73C618 L13F7L13M11881.251.470.68C618 L12F4L12M21891.321.530.78C623 L9F3L10M11901.561.880.74C623 L9F8L9M21911.451.930.88C627 L12F10L9M61921.691.481.11C654 L13F9L6M31931.251.530.71C673 L8F3L8M81941.441.921.01C563 L14F8L9M31951.251.530.77C549 L12F4L9M41961.441.920.80C555 L10F6L12M11971.221.370.60C487 L13F7L13M41981.471.330.89C488 L9F9L9M21991.341.710.73C498 L13F4L13M42002.302.280.83C503 L15F2L9M52013.244.360.91C508 L7F4L9M32023.385.450.67C711 L13F5L10M42039.5510.810.75C708 L10F4L14M72045.619.561.67C743 L18F7L8M102057.346.671.36C756 L20F4L10M82066.498.820.96C788 L1F5L5M52077.6410.610.88C731 L5F2L9M92086.579.651.08F9 L10W2090.48F9 L10M22110.71 The codes of proteins in the table are in accordance with the following rules: polypeptide code+peptide linker code+FCcode+peptide linker code+FGF21 mutant code. For example, C209L13F4L13M9indicates that the C209 polypeptide is fused with an IgG FCcodenamed F4via a peptide linker codenamed L13, and further fused with an FGF21 mutant codenamed M9via a peptide linker codenamed L13. Embodiment 6 Stability Study of the Tri-Agonist Active Proteins in Presence of DPP-IV The purified tri-agonist active proteins were dissolved in 10 mM HEPES buffer (containing 0.05 mg/ml BSA) at a final concentration of 5 uM. Recombinant protease DPP-IV was added (final concentration of 10 nM), and in-vitro GCGR agonist activity was determined after bathing at 37° C. for 24 hours. Percentage of residual activity=(activity after DPP-IV treatment/activity before DPP-IV treatment) ×100%. In this Embodiment, GCG analogues with an unnatural amino acid Aib or D-Ser introduced at the second position were used as controls: GDSerGS:(SEQ ID NO. 134)H-D-Ser-QGTFTSDYSKYLDSQAAQDFVQWLMNGGPSSGAPPPS;GAibGS:(SEQ ID NO. 135)H-Aib-QGTFTSDYSKYLDSQAAQDFVQWLMNGGPSSGAPPPS; C364 (SEQ ID NO.70), C382 (SEQ ID NO.57), C495 (SEQ ID NO.53), C462 (SEQ ID NO.66), C225 (SEQ ID NO.47) and C209 (SEQ ID NO.71) serve as controls for the stability study in this embodiment. The results are shown in Table 4: TABLE 4ActivityActivityCodes of activepreservationCodes of activepreservationproteinsrate (%)proteinsrate (%)C001L13F83.2C228 L13F4L13M1291.4C002L13F82.9C187 L9F4L12M795.7C002L13F8L10W3.8C364 L10F4L12M890.6C240 L12F8L12M198.9C209 L13F8L13M991.1C240 L9F7L13M298.4C289 L12F4L8M1093.3C240 L13F4L9M194.7C611 L11F4L11M1197.5C240 L9F2L13M396.1C618 L13F7L13M192.7C240 L13F10L9M297.7C618 L12F4L12M289.4C225 L10F10L14M289.2C623 L9F3L10M195.6C163 L13F8L13M297.3C623 L9F8L9M297.6C271 L9F4L8M296.0C627 L12F10L9M695.6C368 L10F4L10M297.9C654 L13F9L6M393.3C495 L13F8L13M196.9C673 L8F3L8M896.1C495 L13F8L10M299.3C563 L14F8L9M395.4C495 L9F10L9M195.6C549 L12F4L9M497.5C353 L13F3L10M494.5C555 L10F6L12M188.3C352 L13F4L9M398.7C487 L13F7L13M498.7C382 L9F3L9M296.6C488 L9F9L9M299.1C382 L10F2L13M295.8C498 L13F4L13M487.9C382 L13F10L9M197.8C503 L15F2L9M595.6C382 L13F8L10M299.3C508 L7F4L9M397.2C382 L12F7L9M296.9C711 L13F5L10M495.9C382 L14F4L9M294.9C708 L10F4L14M790.5C232 L9F3L10M398.2C743 L18F7L8M1099.0C227 L12F5L14M491.9C756 L20F4L10M894.9C266 L13F7L13M488.2C788 L1F5L5M596.5C137 L10F8L9M596.8C731 L5F2L9M998.6C399 L12F4L19M887.3GAibGS98.7C392 L11F7L10M594.5GDSerGS99.2C462 L9F2L9M1093.8C3647.5C462 L10F5L10M396.4C3826.8C462 L11F8L13M195.7C4959.2C462 L13F4L10M292.2C4624.3C462 L13F8L10M297.9C2255.8C462 L13F10L9M493.7C2098.9 Embodiment 7 Stability Study of the Tri-Agonist Active Proteins in Serum In-Vitro Cell-Based Bioactivity Assay: (1) Tri-agonist active proteins were concentrated by ultrafiltration and diluted with 2 0mM PB (pH7.4) to 1.6 mg/ml. After sterilization and filtration, the proteins were diluted with serum (FBS, GEMINI 900-108, A97E00G) 10 times, mixed and divided into sterile centrifuge tubes; (2) Glucagon (SEQ ID NO: 42, Hangzhou Chinese Peptide Biochemical Co., Ltd, GLUC-004) was diluted to 0.2 mg/ml. After sterilization and filtration, the diluted Glucagon was further diluted with serum as above 10 times, mixed and divided into sterile centrifuge tubes; (3) One or two tubes of the above samples were stored at −20 ° C. as controls; the other tubes were incubated at a 37° C. At different time points, samples were selected to detect GCGR agonist activity; (4) HEK 293T/GCGR cells were subcultured twice and then plated in a 96-well plate for sample activity determination. The residual activity was obtained by taking the activity at 0 hour as 100%, and comparing the activity measured at subsequent time points with that at 0 hour. Except for C002L13F4L13W, the results of stability study in serum are similar to those in Table 4, there is no significant difference in stability among the tri-agonist active proteins. The relative activity of the exemplary tri-agonist active proteins over time is shown inFIGS.2A-C. Embodiment 8 Intraperitoneal Glucose Tolerance Test (IPGTT) in Normal ICR Mice Normal ICR mice were divided into several groups, 8 mice per group. Mice were fasted overnight followed by blood collection from the tail (sample marked as t=0 min blood glucose) and subcutaneously injection of the tri-agonist active proteins (40 nmol/kg, in acetate buffer), combined proteins (combination group) or saline PBS, respectively. The combination group was pre-mixed before administration (40 nmol/kg each, acetate buffer). Fifteen minutes after administration, glucose was injected intraperitoneally (2 g/kg of body weight) and blood glucose levels were recorded at t=30 min, t=60 min, t=120 min, and t=240 min. The animals kept fasted during the test period to prevent interference from food intake. The result is shown inFIG.3. Embodiment 9 Pharmacodynamic Study of Continuous Administration of Tri-Agonist Active Proteins in Diet-Induced Obese (DIO) Mice 7-Week-old C57BL/6J male mice were fed a high-fat diet (60% kcal from fat) for another 16 weeks (a total of 23 weeks). The study was initiated when body weight of the mice reached approximate 55 g. Feeding conditions were as followed: 12 h light/12 h darkness, free food intake, single cage. Mice were grouped (8 mice per group) according to body weights and body weight growth curves the day before administration and administered subcutaneously at the next day. According to Table 5, the active proteins were administered at a dose of 10 nmol/kg of body weight or 30 nmol/kg of body weight once every 4 days. The negative control group was injected with saline (PBS) at 5 μl/kg of body weight. The positive control group was administered with Liraglutide (30 nmol/kg of body weight) once daily for 28 consecutive days. Body weight and food intake of mice were recorded every day. Mice were sacrificed 5 days after the last administration. Retro-orbital bleeding was performed. Plasma samples were frozen and stored at −80 ° C. Average weight change of animals before administration and sacrifice was calculated. The results of weight change are shown inFIG.4. The change in total food intake is shown inFIG.5. TABLE 5SamplesSEQ ID NO.Dose (nM)C002L13F8L10W14910C495 L13F8L10M216010C382 L13F8L10M216710C462 L13F8L10M218010C495 L13F8L10M216030C382 L13F8L10M216730C462 L13F8L10M218030 Embodiment 10 Pharmacodynamic Study of Combined Administration in Diet-Induced Obese (DIO) Mice The difference between this Embodiment and Embodiment 9 is that the dual-agonist active proteins and long-acting FGF21 analogues were co-administered as combination. 7-Week-old C57BL/6J male mice were fed a high-fat diet (60% kcal from fat) for another 16 weeks (a total of 23 weeks). The study was initiated when the body weight of the mice reached approximate 55g. Feeding conditions were as followed: 12 h light/12 h darkness, free food intake, single-cage; mice were grouped (8 mice per group) according to body weight and body weight growth curves the day before administration and administered subcutaneously at the next day. The dual-agonist active proteins and long-acting FGF21 analogues were mixed according to the dose information shown in Table 6 before administration. The administration was given at a dose of 15 nmol/kg of body weight or 30 nmol/kg of body weight once every 4 days. The negative control group was injected with saline (PBS) at 5 μl/kg of body weight. The positive control group was injected with Liraglutide (30 nmol/kg of body weight) and administered once a day for 28 consecutive days. The body weight and food intake of mice were measured every day. Mice were sacrificed 5 days after the last administration. Retro-orbital bleeding was performed. Plasma samples were frozen and stored at −80 ° C. Average weight change of animals before administration and sacrifice was calculated. The results of weight change are shown inFIG.6. The change in total food intake is shown inFIG.7. TABLE 6SamplesSEQ ID NO.Dose (nM)C495 L13F8+ F9L10M1106, 21030 eachC495 L13F8+ F9L10M2106, 21130 eachC382 L13F8+ F9L10M298, 21130 eachC382 L13F8+ F9L10M398, 21230 eachC462 L13F8+ F9L10M1114, 21030 eachC462 L13F8+ F9L10M2114, 21130 eachC462 L13F8+ F9L10M3114, 21230 eachC462 L13F811430C382 L13F89830C495 L13F810630F9L10M121030F9L10M221130F9L10M321230 Embodiment 11 Assay Method Development and Activity Determination of Tri-Agonist Active Proteins Structures of the tri-agonist active proteins of the present invention are shown in Embodiment 3. The fusion proteins C382L13F3L10M2 and C382L13F9L10M2 were prepared according to amino acids shown in SEQ ID NO.336 and SEQ ID NO.338, respectively, and their corresponding nucleotides are shown in SEQ ID NO.337 and SEQ ID NO.339, respectively. The preparation method used in this Embodiment is the same as that in Embodiment 2. The tri-agonist active proteins obtained in this Embodiment was subjected to in-vitro activity bioassays, including GLP-1R agonist activity bioassay, GCGR agonist activity bioassay and FGF21 activity bioassay. The assay method is the same as in Embodiment 5. The results are shown in Table 7. TABLE 7AminoGCGRGLP-1RacidagonistagonistFGF21sequencesactivityactivityactivityCodes of active(SEQ(EC50,(EC50,(EC50,proteinsID NO.)nM)nM)nM)Native Glucagon420.94120.87Native GLP-11>10000.52Native FGF211360.12C382L13F3L10M23361.021.140.61C382L13F9L10M23381.131.230.83 Embodiment 12 Stability Study of the Tri-Agonist Active Proteins in Serum The tri-agonist active proteins obtained in Embodiment 11 were subjected to stability study in serum, and the assay method is the same as in Embodiment 7. The residual relative activity of the tri-agonist active proteins over time is shown inFIG.8. FIG.8suggests that the tri-agonist active proteins showed improved residual activity after incubation for 7 days in serum, indicating a greatly improved stability compared with native GLP1 or Glucagon peptide. Embodiment 13 Intraperitoneal Glucose Tolerance Ttest (IPGTT) in Normal ICR Mice Normal ICR mice were divided into several groups, 8 mice per group. Mice were fasted overnight followed by blood collection from the tail (marked as t=0 min blood glucose sample), and subcutaneously injection of the tri-agonist active proteins (40 nmol/kg, acetate buffer) obtained in Embodiment 1, COO2L13F8L1OW (40 nmol/kg, acetate buffer) or PBS, respectively. Fifteen minutes later, glucose was injected intraperitoneally (2 g/kg of body weight) and blood glucose levels at t=30 min, t=60 min, t=120 min, and t=240 min were recorded. The animals kept fasted during the test period to prevent interference from food intake. The result is shown inFIG.9. The data inFIG.9shows that C382L13F3L10M2 and C382L13F9L10M2 have a significant hypoglycemic effect compared to C002L13F8L10W. Embodiment 14 Pharmacodynamic Study of Continuous Administration of Tri-Agonist Active Proteins in Diet-Induced Obese (DIO) Mice The tri-agonist active proteins obtained in Embodiment 11 were continuously administered in the diet-induced obese (DIO) mice for pharmacodynamic study, following the same method illustrated in Embodiment 9. The results of weight changes are shown inFIG.10. The change in total food intake is shown inFIG.11. FIG.10shows that the tri-agonist active proteins obtained in Embodiment 11 were significantly different from Liraglutide both at the doses of 10 nmol/kg and 30 nmol/kg. The weight loss was positively correlated with dose, and the weight loss effect at the dose of 30 nmol/kg was very obvious.FIG.11indicated that the tri-agonist active proteins obtained in Embodiment 11 have no significant effect on food intake at doses of 10 nmol/kg and 30 nmol/kg. Embodiment 15 Random Blood Glucose Test after Continuous Administration of Tri-Agonist Active Proteins in db/db Mice Hypoglycemic study in leptin receptor-deficient Type 2 diabetes (db/db) mice. db/db mice were screened and evenly grouped according to body weights, non-fasting blood glucose, and OGTT response before drug administration. Each group consists of 10 mice. Individuals too large or too small were excluded as far as possible. Nonfasting blood glucose of animals selected should be greater than 15 mM. In addition, normal ICR mice were selected as basal blood glucose control. According to Table 8, the active proteins were injected subcutaneously at a dose of 20 nmol/kg of body weight. The administration is given once every 4 days, the first administration on day 0 and the last administration on day 24. Saline (PBS) (5 μl/g of body weight) was given to the negative control group; liraglutide (10 nmol/kg of body weight) was given to the positive control group. The above groups were administrated subcutaneously once a day for 26 consecutive days. Random blood glucose values were measured at 9 am before the first administration and day 2, 6, 10, 14, 18, 22, and 26. The results of random blood glucose changes are shown inFIG.12. FIG.12shows that the hypoglycemic effect of the tri-agonist active proteins in Table 8 were significantly superior to the positive control, liraglutide, in the leptin receptor-deficient Type 2 diabetes (db/db) mice. The blood glucose level of C382L13F3L10M2was comparable to the normal ICR mice control (not shown in the figure). TABLE 8SamplesSEQ ID NO.Dose (nM)C462L13F4L10M217920C382L13F3L10M233620C382L13F9L10M233820C495L13F3L10M234020 Nucleotide sequences corresponding to the active proteins mentioned in this specification are shown in Table 9. TABLE 9CorrespondingSerial numberActive proteinnucleotide sequence1C001L13F8SEQ ID NO. 2222C002L13F8SEQ ID NO. 2233CG283L13F8SEQ ID NO. 2244C240L13F8SEQ ID NO. 2255CG214L13F8SEQ ID NO. 2266C382L13F8SEQ ID NO. 2277CG267L13F8SEQ ID NO. 2288C276L13F8SEQ ID NO. 2299C308L13F8SEQ ID NO. 23010C368L13F8SEQ ID NO. 23111C224L13F8SEQ ID NO. 23212C225L13F8SEQ ID NO. 23313CG308L13F8SEQ ID NO. 23414C495L13F8SEQ ID NO. 23515C319L13F8SEQ ID NO. 23616C364L13F8SEQ ID NO. 23717C214L13F8SEQ ID NO. 23818C232L13F8SEQ ID NO. 23919C303L13F8SEQ ID NO. 24020C392L13F8SEQ ID NO. 24121CG303L13F8SEQ ID NO. 24222C462L13F8SEQ ID NO. 24323C240 L12F8SEQ ID NO. 24424C368 L10F4SEQ ID NO. 24525C364 L10F4SEQ ID NO. 24626C352 L13F4SEQ ID NO. 24727C225 L10F10SEQ ID NO. 24828C228L13F4SEQ ID NO. 24929C187 L9F4SEQ ID NO. 25030C618 L12F4SEQ ID NO. 25131C623 L9F3SEQ ID NO. 25232C228 L13F4SEQ ID NO. 25333C498 L13F4SEQ ID NO. 25434C503 L15F2SEQ ID NO. 25535C508 L7F4SEQ ID NO. 25636C756 L20F4SEQ ID NO. 25737C788 L1F5SEQ ID NO. 25838C289 L12F4SEQ ID NO. 25939C611 L11F4SEQ ID NO. 26040C209 L13F8SEQ ID NO. 26141C627 L12F10SEQ ID NO. 26242C002L13F8L10WSEQ ID NO. 26343C240 L12F8L12M1SEQ ID NO. 26444C240 L9F7L13M2SEQ ID NO. 26545C240 L13F4L9M1SEQ ID NO. 26646C240 L9F2L13M3SEQ ID NO. 26747C240 L13F10L9M2SEQ ID NO. 26848C225 L10F10L14M2SEQ ID NO. 26949C163 L13F8L13M2SEQ ID NO. 27050C271 L9F4L8M2SEQ ID NO. 27151C368 L10F4L40M2SEQ ID NO. 27252C495 L13F8L13M1SEQ ID NO. 27353C495 L13F8L10M2SEQ ID NO. 27454C495 L9F10L9M1SEQ ID NO. 27555C353 L13F3L10M4SEQ ID NO. 27656C352 L13F4L9M3SEQ ID NO. 27757C382 L9F3L9M2SEQ ID NO. 27858C382 L10F2L13M2SEQ ID NO. 27959C382 L13F10L9M1SEQ ID NO. 28060C382 L13F8L10M2SEQ ID NO. 28161C382 L12F7L9M2SEQ ID NO. 28262C382 L14F4L9M2SEQ ID NO. 28363C232 L9F3L10M3SEQ ID NO. 28464C227 L12F5L14M4SEQ ID NO. 28565C266 L13F7L13M4SEQ ID NO. 28666C137 L10F8L9M5SEQ ID NO. 28767C399 L12F4L19M8SEQ ID NO. 28868C392 L11F7L10M5SEQ ID NO. 28969C462 L9F2L9M10SEQ ID NO. 29070C462 L10F5L10M3SEQ ID NO. 29171C462 L11F8L13M1SEQ ID NO. 29272C462 L13F4L10M2SEQ ID NO. 29373C462 L13F8L10M2SEQ ID NO. 29474C462 L13F10L9M4SEQ ID NO. 29575C228 L13F4L13M12SEQ ID NO. 29676C187 L9F4L12M7SEQ ID NO. 29777C364 L10F4L12M8SEQ ID NO. 29878C209 L13F8L13M9SEQ ID NO. 29979C289 L12F4L8M10SEQ ID NO. 30080C611 L11F4L11M11SEQ ID NO. 30181C618 L13F7L13M1SEQ ID NO. 30282C618 L12F4L12M2SEQ ID NO. 30383C623 L9F3L10M1SEQ ID NO. 30484C623 L9F8L9M2SEQ ID NO. 30585C627 L12F10L9M6SEQ ID NO. 30686C654 L13F9L6M3SEQ ID NO. 30787C673 L8F3L8M8SEQ ID NO. 30888C563 L14F8L9M3SEQ ID NO. 30989C549 L12F4L9M4SEQ ID NO. 31090C555 L10F6L12M1SEQ ID NO. 31191C487 L13F7L13M4SEQ ID NO. 31292C488 L9F9L9M2SEQ ID NO. 31393C498 L13F4L13M4SEQ ID NO. 31494C503 L15F2L9M5SEQ ID NO. 31595C508 L7F4L9M3SEQ ID NO. 31696C711 L13F5L10M4SEQ ID NO. 31797C708 L10F4L14M7SEQ ID NO. 31898C743 L18F7L8M10SEQ ID NO. 31999C756 L20F4L10M8SEQ ID NO. 320100C788 L1F5L5M5SEQ ID NO. 321101C731 L5F2L9M9SEQ ID NO. 322102F9L10WSEQ ID NO. 323103F9L10M1SEQ ID NO. 324104F9L10M2SEQ ID NO. 325105F9L10M3SEQ ID NO. 326106F9L10M4SEQ ID NO. 327107F9L10M5SEQ ID NO. 328108F9L10M6SEQ ID NO. 329109F9L10M7SEQ ID NO. 330110F9L10M8SEQ ID NO. 331111F9L10M9SEQ ID NO. 332112F9L10M10SEQ ID NO. 333113F9L10M11SEQ ID NO. 334114F9L10M12SEQ ID NO. 335115C382L13F3L10M2SEQ ID NO. 336116C382L13F3L10M2SEQ ID NO. 337117C382L13F9L10M2SEQ ID NO. 338118C382L13F9L10M2SEQ ID NO. 339119C495L13F3L10M2SEQ ID NO. 340120C495L13F3L10M2SEQ ID NO. 341 The above are only some preferred embodiments of the present disclosure instead of limitations on the present disclosure in any form or substance. It should be noted that, for those skilled in the art, improvements and supplements (including the fusion of the exemplary GCG analogues of the present disclosure with various FCand FGF21 analogue) may be made without departing from the methods of the present disclosure, the improvements and supplements shall also be covered by the protection of the present disclosure. The equivalent changes of alternations, modifications and evolutions can be made by those skilled in the art using the technical contents revealed above and without departing from the spirit and scope of the present disclosure, and those equivalent changes are regarded as equivalent embodiments of the present disclosure. Meanwhile, any alterations, modifications, and evolutions of any equivalent changes made to the above embodiments according to the essential technology of the present disclosure still fall within the scope of the technical solution of the present disclosure.
66,071
11858976
DETAILED DESCRIPTION OF THE INVENTION The present invention embraces a chimeric receptor molecule composed of a natural killer cell receptor and an immune signaling receptor expressed on the surface of a T cell to activate killing of a tumor cell. Nucleic acid sequences encoding the chimeric receptor molecule are introduced into T-cells ex vivo and T-cells that express the chimeric receptor molecule are subsequently injected into a subject in need of treatment. In this manner, the chimeric receptor molecules provide a means for the subject's own immune cells to recognize and activate anti-tumor immunity and establish long-term, specific, anti-tumor responses for treating tumors or preventing regrowth of dormant or residual tumor cells. To prevent potential side effects that may occur from uncontrolled inflammation or response against non-tumor tissue, suicide genes are further introduced into the T-cells expressing the chimeric receptor molecule. The suicide gene is activated by administering an agent, specific for the suicide gene, to the patient thereby eliminating all cells expressing the chimeric receptor molecule. By way of illustration, murine chimeric receptor molecules composed of NKG2D or Dap10 in combination with a N-terminally attached CD3ζ were generated and expressed in murine T-cells. NKG2D is a type II protein, in which the N-terminus is located intracellularly (Raulet (2003)Nat. Rev. Immunol.3:781-790), whereas the CD3ζ chain is type I protein with the C-terminus in the cytoplasm (Weissman, et al. (1988)Proc. Natl. Acad. Sci. USA85:9709-9713). To generate a chimeric NKG2D-CD3 fusion protein, an initiation codon ATG was placed ahead of the coding sequence for the cytoplasmic region of the CD3ζ chain (without a stop codon TAA) followed by a wild-type NKG2D gene. Upon expression, the orientation of the CD3ζ portion is reversed inside the cells. The extracellular and transmembrane domains are derived from NKG2D. A second chimeric gene encoding the Dap10 gene followed by a fragment coding for the CD3ζ cytoplasmic domain was also constructed. The structures of the chimeric and wild-type receptors used are diagrammed inFIG.1. To determine whether murine chimeric NKG2D or murine chimeric Dap10 receptors could be expressed in a similar manner as wild-type murine NKG2D or Dap10, a NKG2D gene with an adaptor protein gene (Dap10/Dap12) were co-transfected into Bosc23 cells and NKG2D expression was determined by flow cytometry. To analyze those cells that were transfected, a bicistronic vector with a green fluorescent protein (GFP) gene controlled by an internal ribosome entry site (IRES) was used. NKG2D surface expression was normalized by gating on the GFP+cell population. Like many NK receptors, such as CD94/NKG2C, Ly49D, and Ly49H, NKG2D needs to be associated with adaptor proteins (i.e., Dap10 and/or Dap12) for surface expression (Raulet (2003)supra; Lanier (2003)Curr. Opin. Immunol.15:308-314) Packaging cell Bosc23 did not express either NKG2D or Dap10/Dap12, and transfection with only one of the two components did not give rise to surface expression of NKG2D. However, co-transfection of a NKG2D gene along with an adaptor protein gene led to significant membrane expression of NKG2D. Compared with Dap12, Dap10 transfection resulted in higher NKG2D surface expression. Surface expression of NKG2D after association with chimeric Dap10 adaptor was higher than that with wild-type DAP10. Higher surface expression of NKG2D was also observed after transfection with chimeric NKG2D than with wild-type NKG2D genes, especially when pairing with the Dap12 gene (>5-fold increase in MFI). Concentrated, high-titer, retroviral vectors (ecotropic) were used to infect C57BL/6 spleen cells, and NKG2D surface expression was determined by flow cytometry seven days after retroviral transduction. Genetic modification of T cells with wild-type Dap10, Dap12 and NKG2D did not significantly increase the surface expression of NKG2D (10-20%) compared to vector alone. In contrast, significantly higher NKG2D expression was observed in T cells modified with either chimeric NKG2D (42%) or chimeric Dap10 (64%). In chimeric Dap10-transduced T cells, the surface-expressed NKG2D molecules were only due to endogenous molecules, whereas both endogenous and exogenous NKG2D molecules were responsible for surface expression in chimeric NKG2D-modified T cells. Taken together, these data indicate that chimeric NKG2D and chimeric Dap10 molecules are expressed in a similar manner as the wild-type molecules and that they increase NKG2D expression on T cells. To assess whether the murine chimeric DAP10 or murine chimeric NKG2D-transduced T cells were capable of recognizing NKG2D ligands, NKG2D ligand-positive tumor cells (RMA/Rae-1p, RMA/H60 and YAC-1) were used as targets for chimeric NKG2D-bearing T cells. Chimeric DAP10 or chimeric NKG2D-transduced T cells produced high amounts of IFN-γ (20-30 ng/mL) after co-culture with RMA/Rae-1β, RMA/H60 or YAC-1 cells (Table 1) but not with RMA cells (no ligands), indicating that these chimeric NKG2D-modified T cells could functionally recognize NKG2D ligand-bearing tumor cells. TABLE 1IFN-γ (ng/mL ± SD)ConstructMediaRMARMA/Rae-1βRMA/H60YAC-1Vector0.03 ± 0.030.09 ± 0.180.02 ± 0.030.11 ± 0.630.84 ± 0.29OnlyWild-type0.01 ± 0.010.10 ± 0.210.04 ± 0.060.05 ± 0.001.08 ± 1.48NKG2D*Chimeric0.07 ± 0.080.37 ± 0.344.70 ± 0.788.40 ± 1.6017.80 ± 4.60NKG2DWild-type0.01 ± 0.100.11 ± 0.110.04 ± 0.030.09 ± 0.031.43 ± 1.72DAP10*Chimeric0.49 ± 0.550.82 ± 0.527.50 ± 4.4018.60 ± 7.6028.70 ± 8.30DAP10Wild-Type0.00 ± 0.010.00 ± 0.010.53 ± 0.670.13 ± 0.100.73 ± 0.09DAP12#*p = 0.74;#p = 0.56.Data are representative of 3 experiments. Similarly, chimeric human NKG2D-bearing CD8+T cells secrete IFN-γ when brought into contact with human tumor cells from breast cancer (MCF-7, T47D), prostate cancer (DU145), pancreatic cancer (Pan-1), and melanoma cancer (A375) (Table 2). T cells were cultured with irradiated tumor cells at a 4:1 ratio for 72 hours and IFN-γ was measured by ELISA. T cells cultured without tumor cells functioned as a media only control which produced no detectable IFN-γ. The specificity of the interaction was evident by comparing chimeric NKG2D transduced T cells to vector only. TABLE 2IFN-γ (pg/mL ± SD)ConstructT47DMCF-7Panc-1DU-145A375Vector28.953.597.261.4262.4Only(±12.5)(±3.6)(±8.0)(±4.2)(±44.2)Wild-type35.243.6115.884.4200.5NKG2D*(±30.0)(±9.2)(±89.8)(±47.1)(±79.5)Chimeric130.32928.35028.14427.92609.2NKG2D(±70.4)(±251.1)(±407.2)(±470.1)(±293.2)Tumor23.417.726.627.411.4Alone(±26.9)(±8.7)(±7.5)(±10.2)(±17.7) In addition, upon NKG2D ligation, chimeric DAP10 or chimeric NKG2D-modified T cells also released significant amounts of proinflammatory chemokines (CCL3 and CCL5), as well as Th1 cytokines, GM-CSF and IL-3, but not Th2 cytokines IL-5 and IL-10. In contrast, wild-type Dap10, Dap12 or NKG2D alone-modified T cells did not show any significant response to the stimulation by RMA/Rae-1p, RMA/H60 or YAC-1 cells. These data demonstrate that the chimeric molecules led to the direct activation of T cells. The cytotoxic activity of murine chimeric NKG2D-modified splenic T cells against tumor cells was also determined. Chimeric Dap10 or chimeric NKG2D-transduced T cells were able to lyse NKG2D ligand-expressing target cells (RMA/Rae-1(3, RMA/H60, EG7 and YAC-1) in vitro (FIG.2, Panels B-E). The specificity of the interaction was apparent from the absence of lysis of YAC-1, EG7, RMA/Rae-1β and RMA/H60 cells by vector only-transduced T cells, and the lack of lysis of RMA cells by chimeric Dap10 or chimeric NKG2D-modified T cells (FIG.2, Panel A). Similar to cytokine production, no significant specific lysis of tumor cells was observed by wild-type Dap10 or wild-type NKG2D-modified T cells. T cells transduced with wild-type Dap12 were able to kill target cells that expressed ligands for NKG2D. Activated murine CD8+ T cells express NKG2D (associated with Dap10), so expression of Dap12 would allow the endogenous NKG2D to associate with Dap12 and provide a primary activation signal. It is noteworthy that T cells transduced with Dap12 were three- to five-fold less efficient than T cells transduced with chimeric NK receptors at killing tumor cells. The killing of YAC-1 and EG7 tumor cells demonstrates that chimeric NK receptors provide the T cells with a means to kill tumor cells that express endogenous NKG2D ligands. These data demonstrated the need for NKG2D ligand expression on the target cells. To investigate the role of the NKG2D receptor, it was determined whether blocking antibodies to NKG2D would diminish cytotoxic activity. Chimeric NKG2D-transduced T cells killed RMA/Rae-1β and EG7 tumor cells and this activity was reduced when anti-NKG2D antibodies were included in the assay. Vector only-transduced T cells were unable to kill the target cells and the activity was not changed with the addition of anti-NKG2D antibodies. While the data indicate that the NKG2D receptor was responsible for the activity in these assays, the chimeric receptors may have, in some way, altered the T cells to kill via their T cell receptor. To address this, the ability of chimeric NKG2D-transduced T cells to kill RMA/Rae-1β tumor cells was examined. RMA-S cells are deficient in TAP genes and express very low levels of MHC class I molecules on the cell surface and no MHC class II molecules (Aldrich, et al. (1992)J. Immunol.149:3773-3777). Chimeric NKG2D-bearing T cells killed RMA/Rae-1p tumor cells but not RMA-S cells. Vector-transduced T cells did not kill either RMA-S cell line. Thus, these data indicate that chimeric NKG2D functions via direct NKG2D recognition of its ligand on target cells. Having shown that chNKG2D-modified T cells could react against NKG2D ligand-positive tumor cells in vitro, the therapeutic potential of chimeric NKG2D-modified T lymphocytes was determined in vivo. Chimeric NKG2D-bearing T cells (106) were co-injected with RMA/Rae-1p tumor cells (105) subcutaneously to C57BL/6 mice. T cells transduced with the chimeric NKG2D construct significantly (P<0.05 at days 5-15) inhibited the growth of RMA/Rae-1 tumors compared with vector-transduced T cells or tumor alone (Table 3). Approximately 36% ( 4/11) of chimeric NKG2D-bearing T cell-treated mice were tumor-free after 30 days. Chimeric NKG2D-bearing T cells did not show any significant inhibition effects on the growth of wild-type RMA cells, indicating that inhibition of RMA/Rae-1β tumor growth by chimeric NKG2D T cells was mediated by chimeric NKG2D-Rae-1β engagement. TABLE 3Tumor Area (Mean mm2± SEM)T Cells transducedT cells transducedwith chimericwith vector only +DayNKG2D + RMA/Rae-1βRMA/Rae-1βRMA/Rae-1β00.00 ± 0.000.00 ± 0.000.00 ± 0.0050.00 ± 0.0016.14 ± 2.866.79 ± 1.4773.10 ± 1.4038.45 ± 3.7928.69 ± 5.4998.11 ± 3.0957.40 ± 6.4342.22 ± 6.381111.84 ± 5.2490.31 ± 11.6460.60 ± 12.101314.73 ± 7.24127.30 ± 16.8582.67 ± 19.441520.60 ± 8.32N. D.110.51 ± 29.07 Results are a summary of three experiments. In a second and more stringent model, transduced T cells (107) were adoptively transferred i.v. into B6 mice one day before s.c. tumor inoculation in the right flank. These chimeric NKG2D-bearing T cells significantly (P<0.05 at days 9-17) suppressed the growth of RMA/Rae-1β tumors (s.c.) compared with control vector-modified T cells (Table 4). As for the toxicity of treatment with chimeric NKG2D-modified T cells, the animals treated with chimeric NKG2D-bearing T cells did not show any overt evidence of inflammatory damage (i.e., ruffled hair, hunchback or diarrhea, etc.) indicating there was no overt toxicity. TABLE 4Tumor Area (Mean mm2± SEM)T Cells transduced withControl T cells withDaychimeric NKG2DVector Only53.06 ± 1.974.41 ± 2.20712.14 ± 3.0617.81 ± 1.75913.94 ± 2.8530.58 ± 3.871125.92 ± 4.7745.13 ± 3.271332.11 ± 5.8464.83 ± 10.451534.39 ± 9.7780.72 ± 13.341737.81 ± 11.6896.30 ± 14.15 Results are a summary of three experiments. Because the immune system can select for tumor variants, the most effective immunotherapies for cancer are likely going to be those that induce immunity against multiple tumor antigens. Thus, it was tested whether treatment with chimeric NKG2D-bearing T cells could induce host immunity against wild-type tumor cells. Mice that were treated with chimeric NKG2D-bearing T cells and RMA/Rae-1β tumor cells, and were tumor-free after 30 days, were challenged with RMA tumor cells. These tumor-free mice were resistant to a subsequent challenge of wild-type RMA cells (104), whereas all control näive mice had aggressive tumors (tumor area: ˜100 mm2) after 2 weeks (Table 5). This observation indicates that adoptive transfer of chimeric NKG2D-bearing T cells allows hosts to generate T cell memory. TABLE 5Tumor Area (Mean mm2± SEM)Mice treated with T Cellstransduced with chimericDayNKG2D + RMA/Rae-1βNäive Mice50.00 ± 0.001.33 ± 2.3170.00 ± 0.0011.65 ± 10.2090.00 ± 0.0038.75 ± 8.84110.00 ± 0.0060.17 ± 6.10130.00 ± 0.0091.10 ± 5.59150.00 ± 0.00102.81 ± 17.94190.00 ± 0.00146.71 ± 45.72 Results are a summary of three experiments. In similar experiments, human chimeric receptor molecules composed of NKG2D or Dap10 in combination with a N-terminally attached CD3ζ were generated and expressed in Bosc23 cells. Surface expression of NKG2D was not observed when either human Dap10 or human chimeric NKG2D were transfected alone. However, co-transfection of a human chimeric NKG2D or human chimeric NKG2D-GFP gene along with a wild-type human DAP10 gene or mouse DAP10-GFP construct led to significant membrane expression of NKG2D. Binding of a human NKG2D fusion protein, composed of NKG2D with an N-terminally attached murine IgG1 Fc portion, to human NKG2D ligand on various tumor cell lines was assessed. Human NKG2D ligand was found to be present on Jurkat (T lymphocyte origin), RPMI8866 (B cell origin), K562 (erythroid origin), Daubi (B cell origin), and U937 (monocyte origin) tumor cell lines. Therefore, like the mouse chimeras, a human chimeric NKG2D construct can functionally recognize NKG2D ligand-bearing tumor cells. The cytotoxic activity of human chimeric NKG2D-modified T cells against tumor cells was also determined. Human chimeric NKG2D-transduced primary human T cells were able to lyse mastocytoma cell line P815 transduced with human MIC-A (P815/MICA-A) in vitro (Table 6). The specificity of the interaction was apparent from the absence of lysis of wild-type P815 tumor cells and the absence of lysis by vector only-transduced T cells. TABLE 6Tumor Cell LineSpecific Lysis (%)P815/MIC-AP815Effector:Target Ratio15251525Vector Only0.00.00.60.0−0.60.0Human chimeric5.417.335.4−2.5−0.31.1NKG2D Further, blocking of human chimeric NKG2D with an anti-NKG2D antibody prevented killing of K562 and RPMI8866 tumor cells by chimeric NKG2D-transduced human T cells (Table 7). These data demonstrate receptor specificity because a control antibody could not prevent killing. TABLE 7Tumor Cell LineSpecific Lysis (%)K562RPMI8866Effector:Target Ratio15251525Vector + control0.00.32.30.01.00.0antibodyVector + anti-0.00.72.10.01.01.3NKG2D antibodyHuman chimeric3.117.653.89.327.441.6NKG2D + controlantibodyHuman chimeric1.610.127.01.02.57.5NKG2D + anti-NKG2D antibody Similar to the mouse studies, human chimeric NKG2D-transduced T cells produced high amounts of IFN-γ (˜150-2250 pg/mL) after a 24 hour co-culture with tumor cells that express ligands for NKG2D (i.e., Jurkat, RPMI8866, K652, ECC-01 and P815/MIC-A tumor cells) compared with tumor cells that do not express NKG2D ligands (P815) or T cells incubated alone (Table 8). Vector only-transduced T cells did not produce IFN-γ, except against RPMI8866, indicating another ligand on this cell type for these activated T cells; however, IFN-γ production was almost 10-times as high with the human chimeric NKG2D-bearing T cells. Tumor cells alone produce no detectable IFN-γ. TABLE 8IFN-γ (Mean pg/mL ± SD)Tumor cellChimericTumor cellTypeNKG2DVector OnlycontrolP81530.32 ± 1.3115.11 ± 0.945.53 ± 1.31P815/MIC-A140.19 ± 5.919.19 ± 3.302.85 ± 1.50Jurkat182.66 ± 18.317.64 ± 1.042.83 ± 1.33RPMI88662239.95 ± 19.59280.41 ± 13.842.47 ± 2.47K6522305.46 ± 75.841.91 ± 0.571.82 ± 1.39ECC-1469.97 ± 18.792.67 ± 2.670.00 ± 0.00T Cells only13.67 ± 2.550.94 ± 0.54N. D. Amounts represent the average of three experiments Chimeric NK cell receptor-bearing T cells are used to provide a means to attack tumor cells by activating host immunity and destroying tumor cells. In addition to tumors expressing NK cell receptor ligand, it is expected that analysis of ligand-deficient tumor cells will also result in tumor cell killing. For example, it has been demonstrated that ectopic expression of NK cell receptor ligands in tumor cell lines results in potent tumor rejection by syngeneic mice (Diefenbach, et al. (2001)Nature413:165-171). However, using these same mice, subsequent challenge with tumor cell lines not expressing the ligands also resulted in tumor rejection. Using the mouse model as described herein, it is expected that NKG2D ligand-negative tumor cell lines, e.g., EL4, a B6 thymoma; RMA, a B6 T lymphoma derived from Rauscher virus-induced RBL-5 cell line (Karre, et al. (1986)Nature319:675-678); and B16-BL6, a B6 melanoma derived from the B16-FO cell line (Hart (1979)Am. J. Pathol.97:587-600), can be eliminated using the chimeric NK cell receptor-bearing T cells of this invention. Although specific immunity may be obtained against tumors, local immunosuppressive mechanisms, such as regulatory T cells, prevents the function of tumor-specific T cells. The ability to remove these cells and enhance local anti-tumor immune function would be of great benefit for the treatment of cancer. In addition to activating host anti-tumor immunity, it has also now been found that adoptive transfer of chimeric NK cell receptor-bearing T cells can eliminate host regulatory T cells. Indeed, it was observed that treatment of mice with advanced tumors with chimeric receptor-bearing T cells results in a rapid loss of FoxP3+T regulatory cells from within the tumor microenvironment (within 3 days) and a destruction of advanced tumor cells. Accordingly, the chimeric NK cell receptors of the invention can also be used to enhance immunotherapy and vaccination approaches via elimination of host regulatory T cells. Having demonstrated the activation of host anti-tumor immunity and tumor elimination using chimeric NK cell receptors expressed in the T cells of an animal model of cancer and likewise demonstrated human tumor cell killing with human chimeric NK cell receptors, the present invention embraces a nucleic acid construct for expressing a chimeric receptor in host T cells to reduce or eliminate a tumor. The nucleic acid construct contains a first nucleic acid sequence encoding a promoter operably linked to a second nucleic acid sequence encoding a chimeric receptor protein containing a C-type lectin-like natural killer cell receptor, or a protein associated therewith, fused to an immune signaling receptor having an immunoreceptor tyrosine-based activation motif of SEQ ID NO:1. In general, the C-type lectin-like NK cell type II receptor (or a protein associated therewith) is located at the C-terminus of the chimeric receptor protein of the present invention whereas the immune signaling receptor is at the N-terminus, thereby facilitating intracellular signal transduction from the C-type lectin-like NK cell type II receptor. A C-type lectin-like NK cell receptor protein particularly suitable for use in the chimeric receptor of the present invention includes a receptor expressed on the surface of natural killer cells. The receptor can work alone or in concert with other molecules. Ligands for these receptors may or may not be expressed on the surface of one or more tumor cell types, e.g., tumors associated with cancers of the colon, lung, breast, kidney, ovary, cervix, and prostate; melanomas; myelomas; leukemias; and lymphomas (Wu, et al. (2004)J. Clin. Invest.114:60-568; Groh, et al. (1999)Proc. Natl. Acad. Sci. USA96:6879-6884; Pende, et al. (2001)Eur. J. Immunol.31:1076-1086) and are not widely expressed on the surface of cells of normal tissues. Examples of such ligands include, but are not limited to, MIC-A, MIC-B, heat shock proteins, ULBP binding proteins (e.g., ULPBs 1-4), and non-classical HLA molecules such as HLA-E and HLA-G, whereas classical MHC molecules such as HLA-A, HLA-B, or HLA-C and alleles thereof are not generally considered strong ligands of the C-type lectin-like NK cell receptor protein of the present invention. C-type lectin-like NK cell receptors which bind these ligands generally have a type II protein structure, wherein the N-terminal end of the protein is intracellular. Exemplary NK cell receptors of this type include, but are not limited to, Dectin-1 (GENBANK accession number AJ312373 or AJ312372), Mast cell function-associated antigen (GENBANK accession number AF097358), HNKR-P1A (GENBANK accession number U11276), LLT1 (GENBANK accession number AF133299), CD69 (GENBANK accession number NM_001781), CD69 homolog, CD72 (GENBANK accession number NM 001782), CD94 (GENBANK accession number NM 002262 or NM_007334), KLRF1 (GENBANK accession number NM 016523), Oxidised LDL receptor (GENBANK accession number NM_002543), CLEC-1, CLEC-2 (GENBANK accession number NM 016509), NKG2D (GENBANK accession number BC039836), NKG2C (GENBANK accession number AJ001684), NKG2A (GENBANK accession number AF461812), NKG2E (GENBANK accession number AF461157), WUGSC:H_DJ0701016.2, or Myeloid DAP12-associating lectin (MDL-1; GENBANK accession number AJ271684). In particular embodiments, the NK cell receptor is human NKG2D (SEQ ID NO:2) or human NKG2C (SEQ ID NO:3). Similar type I receptors which would be useful in the chimeric receptor of the present invention include NKp46 (e.g., GENBANK accession number AJ001383), NKp30 (e.g., GENBANK accession number AB055881), or NKp44 (e.g., GENBANK accession number AJ225109). As an alternative to the C-type lectin-like NK cell receptor protein, a protein associated with a C-type lectin-like NK cell receptor protein can be used in the chimeric receptor protein of the present invention. In general, proteins associated with C-type lectin-like NK cell receptor are defined as proteins that interact with the receptor and transduce signals therefrom. Suitable human proteins which function in this manner include, but are not limited to DAP10 (e.g., GENBANK accession number AF072845; SEQ ID NO:4) and DAP12 (e.g., GENBANK accession number AF019562; SEQ ID NO:5). To the N-terminus of the C-type lectin-like NK cell receptor is fused an immune signaling receptor having an immunoreceptor tyrosine-based activation motif (ITAM), (Asp/Glu)-Xaa-Xaa-Tyr*-Xaa-Xaa-(Ile/Leu)-Xaa6-8-Tyr*-Xaa-Xaa-(Ile/Leu) (SEQ ID NO:1) which is involved in the activation of cellular responses via immune receptors. Similarly, when employing a protein associated with a C-type lectin-like NK cell receptor, an immune signaling receptor can be fused to the C-terminus of said protein (FIG.1). Suitable immune signaling receptors for use in the chimeric receptor of the present invention include, but are not limited to, the zeta chain of the T-cell receptor, the eta chain which differs from the zeta chain only in its most C-terminal exon as a result of alternative splicing of the zeta mRNA, the delta, gamma and epsilon chains of the T-cell receptor (CD3 chains) and the gamma subunit of the FcR1 receptor. In particular embodiments, the immune signaling receptor is CD3-zeta (CD3ζ) (e.g., GENBANK accession number human NM 198053; SEQ ID NO:6), or human Fc epsilon receptor-gamma chain (e.g., GENBANK accession number M33195; SEQ ID NO:7) or the cytoplasmic domain or a splicing variant thereof. In particular embodiments, a chimeric receptor of the present invention is a fusion between NKG2D and CD3ζ or Dap10 and CD3ζ. As used herein, a nucleic acid construct or nucleic acid sequence is intended to mean a DNA molecule which can be transformed or introduced into a T cell and be transcribed and translated to produce a product (e.g., a chimeric receptor or a suicide protein). In the nucleic acid construct of the present invention, the promoter is operably linked to the nucleic acid sequence encoding the chimeric receptor of the present invention, i.e., they are positioned so as to promote transcription of the messenger RNA from the DNA encoding the chimeric receptor. The promoter can be of genomic origin or synthetically generated. A variety of promoters for use in T cells are well-known in the art (e.g., the CD4 promoter disclosed by Marodon, et al. (2003)Blood101(9):3416-23). The promoter can be constitutive or inducible, where induction is associated with the specific cell type or a specific level of maturation. Alternatively, a number of well-known viral promoters are also suitable. Promoters of interest include the B-actin promoter, SV40 early and late promoters, immunoglobulin promoter, human cytomegalovirus promoter, retrovirus promoter, and the Friend spleen focus-forming virus promoter. The promoters may or may not be associated with enhancers, wherein the enhancers may be naturally associated with the particular promoter or associated with a different promoter. The sequence of the open reading frame encoding the chimeric receptor can be obtained from a genomic DNA source, a cDNA source, or can be synthesized (e.g., via PCR), or combinations thereof. Depending upon the size of the genomic DNA and the number of introns, it may be desirable to use cDNA or a combination thereof as it is found that introns stabilize the mRNA or provide T cell-specific expression (Barthel and Goldfeld (2003)J. Immunol.171(7):3612-9). Also, it may be further advantageous to use endogenous or exogenous non-coding regions to stabilize the mRNA. For expression of a chimeric receptor of the present invention, the naturally occurring or endogenous transcriptional initiation region of the nucleic acid sequence encoding N-terminal component of the chimeric receptor can be used to generate the chimeric receptor in the target host. Alternatively, an exogenous transcriptional initiation region can be used which allows for constitutive or inducible expression, wherein expression can be controlled depending upon the target host, the level of expression desired, the nature of the target host, and the like. Likewise, the signal sequence directing the chimeric receptor to the surface membrane can be the endogenous signal sequence of N-terminal component of the chimeric receptor. Optionally, in some instances, it may be desirable to exchange this sequence for a different signal sequence. However, the signal sequence selected should be compatible with the secretory pathway of T cells so that the chimeric receptor is presented on the surface of the T cell. Similarly, a termination region can be provided by the naturally occurring or endogenous transcriptional termination region of the nucleic acid sequence encoding the C-terminal component, of the chimeric receptor. Alternatively, the termination region can be derived from a different source. For the most part, the source of the termination region is generally not considered to be critical to the expression of a recombinant protein and a wide variety of termination regions can be employed without adversely affecting expression. As will be appreciated by one of skill in the art, in some instances, a few amino acids at the ends of the C-type lectin-like natural killer cell receptor (or protein associated therewith) or immune signaling receptor can be deleted, usually not more than 10, more usually not more than 5 residues. Also, it may be desirable to introduce a small number of amino acids at the borders, usually not more than 10, more usually not more than 5 residues. The deletion or insertion of amino acids will usually be as a result of the needs of the construction, providing for convenient restriction sites, ease of manipulation, improvement in levels of expression, or the like. In addition, the substitute of one or more amino acids with a different amino acid can occur for similar reasons, usually not substituting more than about five amino acids in any one domain. The chimeric construct, which encodes the chimeric receptor according to this invention can be prepared in conventional ways. Since, for the most part, natural sequences are employed, the natural genes are isolated and manipulated, as appropriate (e.g., when employing a Type IT receptor, the immune signaling receptor component may have to be inverted), so as to allow for the proper joining of the various components. Thus, the nucleic acid sequences encoding for the N-terminal and C-terminal proteins of the chimeric receptor can be isolated by employing the polymerase chain reaction (PCR), using appropriate primers which result in deletion of the undesired portions of the gene. Alternatively, restriction digests of cloned genes can be used to generate the chimeric construct. In either case, the sequences can be selected to provide for restriction sites which are blunt-ended, or have complementary overlaps. The various manipulations for preparing the chimeric construct can be carried out in vitro and in particular embodiments the chimeric construct is introduced into vectors for cloning and expression in an appropriate host using standard transformation or transfection methods. Thus, after each manipulation, the resulting construct from joining of the DNA sequences is cloned, the vector isolated, and the sequence screened to insure that the sequence encodes the desired chimeric receptor. The sequence can be screened by restriction analysis, sequencing, or the like. The chimeric constructs of the present invention find application in subjects having or suspected of having cancer by decreasing FoxP3+T regulatory cells present in the tumor microenvironment and reducing the size of a tumor thereby preventing the growth or regrowth of a tumor in these subjects. Accordingly, the present invention further embraces chimeric NK receptor-bearing T cells and methods for using the same to decrease FoxP3+T regulatory cells present in the tumor microenvironment and reduce growth or prevent tumor formation in a subject. The methods of this invention involve introducing a chimeric construct of the present invention into an isolated T cell and introducing into a subject the transformed T cell. Suitable T cells which can be used include, cytotoxic lymphocytes (CTL), tumor-infiltrating-lymphocytes (TIL) or other cells which are capable of killing target cells when activated. As is well-known to one of skill in the art, various methods are readily available for isolating these cells from a subject. For example, using cell surface marker expression or using commercially available kits (e.g., ISOCELL™ from Pierce, Rockford, IL). The T cells used in accordance with the invention are, in order of preference, autologous, allogeneic or xenogeneic, and the choice can largely depend on the urgency of the need for treatment. However, in particular embodiments, the T cells are autologous or isolated from the subject being treated. While the present invention relates to the elimination of tumors, the chimeric NK receptors of the present invention can also be used in the treatment of other diseases where these ligands may be present. For example, the immune response can be down-modulated during autoimmune disease or transplantation by expressing these type of chimeric NK receptors in T regulatory or T suppressor cells. Thus, these cells would mediate their regulatory/suppressive function only in the location where the body has upregulated one of the ligands for these receptors. This ligand upregulation may occur during stress or inflammatory responses. It is contemplated that the chimeric construct can be introduced into T cells as naked DNA or in a suitable vector. Methods of stably transfecting T cells by electroporation using naked DNA are known in the art. See, e.g., U.S. Pat. No. 6,410,319. Naked DNA generally refers to the DNA encoding a chimeric receptor of the present invention contained in a plasmid expression vector in proper orientation for expression. Advantageously, the use of naked DNA reduces the time required to produce T cells expressing the chimeric receptor of the present invention. Alternatively, a viral vector (e.g., a retroviral vector, adenoviral vector, adeno-associated viral vector, or lentiviral vector) can be used to introduce the chimeric construct into T cells. Suitable vectors for use in accordance with the method of the present invention are non-replicating in the T cells. A large number of vectors are known which are based on viruses, where the copy number of the virus maintained in the cell is low enough to maintain the viability of the cell. Illustrative vectors include the pFB-neo vectors (STRATAGENE@) disclosed herein as well as vectors based on HIV, SV40, EBV, HSV or BPV. Once it is established that the transfected or transduced T cell is capable of expressing the chimeric receptor as a surface membrane protein with the desired regulation and at a desired level, it can be determined whether the chimeric receptor is functional in the host cell to provide for the desired signal induction (e.g., production of Rantes, Mipl-alpha, GM-CSF upon stimulation with the appropriate ligand). Subsequently, the transduced T cells are introduced or administered to the subject to reduce the regulatory T cell population and activate anti-tumor responses in said subject. To facilitate administration, the transduced T cells according to the invention can be made into a pharmaceutical composition or made implant-appropriate for administration in vivo, with appropriate carriers or diluents, which further can be pharmaceutically acceptable. The means of making such a composition or an implant have been described in the art (see, for instance,Remington's Pharmaceutical Sciences,16th Ed., Mack, ed. (1980)). Where appropriate, the transduced T cells can be formulated into a preparation in semisolid or liquid form, such as a capsule, solution, injection, inhalant, or aerosol, in the usual ways for their respective route of administration. Means known in the art can be utilized to prevent or minimize release and absorption of the composition until it reaches the target tissue or organ, or to ensure timed-release of the composition. Desirably, however, a pharmaceutically acceptable form is employed which does not ineffectuate the cells expressing the chimeric receptor. Thus, desirably the transduced T cells can be made into a pharmaceutical composition containing a balanced salt solution, preferably Hanks' balanced salt solution, or normal saline. A pharmaceutical composition of the present invention can be used alone or in combination with other well-established agents useful for treating cancer. Whether delivered alone or in combination with other agents, the pharmaceutical composition of the present invention can be delivered via various routes and to various sites in a mammalian, particularly human, body to achieve a particular effect. One skilled in the art will recognize that, although more than one route can be used for administration, a particular route can provide a more immediate and more effective reaction than another route. For example, intradermal delivery may be advantageously used over inhalation for the treatment of melanoma. Local or systemic delivery can be accomplished by administration comprising application or instillation of the formulation into body cavities, inhalation or insufflation of an aerosol, or by parenteral introduction, comprising intramuscular, intravenous, intraportal, intrahepatic, peritoneal, subcutaneous, or intradermal administration. A composition of the present invention can be provided in unit dosage form wherein each dosage unit, e.g., an injection, contains a predetermined amount of the composition, alone or in appropriate combination with other active agents. The term unit dosage form as used herein refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the composition of the present invention, alone or in combination with other active agents, calculated in an amount sufficient to produce the desired effect, in association with a pharmaceutically acceptable diluent, carrier, or vehicle, where appropriate. The specifications for the novel unit dosage forms of the present invention depend on the particular pharmacodynamics associated with the pharmaceutical composition in the particular subject. Desirably an effective amount or sufficient number of the isolated transduced T cells is present in the composition and introduced into the subject such that long-term, specific, anti-tumor responses are established to reduce the size of a tumor or eliminate tumor growth or regrowth than would otherwise result in the absence of such treatment. Desirably, the amount of transduced T cells introduced into the subject causes a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 100% decrease in tumor size when compared to otherwise same conditions wherein the transduced T cells are not present. In embodiments drawn to an elimination or decrease in the regulatory T cell population of a subject, it is desired that the transduced T cells introduced into the subject cause a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 100% decrease in the number of FoxP3+T regulatory cells in the subject being treated as compared to a subject not receiving such treatment. In particular embodiments, the transduced T cells are administered to a subject having or suspected of having cancer so that the population of regulatory T cells is decreased at or near a tumor. A decrease in regulatory T cell population can be determined by, e.g., FACS analysis of a cell sample, wherein cells are sorted based upon the presence of cell surface markers. For example, regulatory T cells can be sorted and counted based upon the presence of CD4, CD25, and Fox3P. The amount of transduced T cells administered should take into account the route of administration and should be such that a sufficient number of the transduced T cells will be introduced so as to achieve the desired therapeutic response. Furthermore, the amounts of each active agent included in the compositions described herein (e.g., the amount per each cell to be contacted or the amount per certain body weight) can vary in different applications. In general, the concentration of transduced T cells desirably should be sufficient to provide in the subject being treated at least from about 1×106to about 1×109transduced T cells, even more desirably, from about 1×107to about 5×108transduced T cells, although any suitable amount can be utilized either above, e.g., greater than 5×108cells, or below, e.g., less than 1×107cells. The dosing schedule can be based on well-established cell-based therapies (see, e.g., Topalian and Rosenberg (1987)Acta Haematol.78 Suppl 1:75-6; U.S. Pat. No. 4,690,915) or an alternate continuous infusion strategy can be employed. These values provide general guidance of the range of transduced T cells to be utilized by the practitioner upon optimizing the methods of the present invention for practice of the invention. The recitation herein of such ranges by no means precludes the use of a higher or lower amount of a component, as might be warranted in a particular application. For example, the actual dose and schedule can vary depending on whether the compositions are administered in combination with other pharmaceutical compositions, or depending on interindividual differences in pharmacokinetics, drug disposition, and metabolism. One skilled in the art readily can make any necessary adjustments in accordance with the exigencies of the particular situation. In particular embodiments, the chimeric nucleic acid construct further contains a suicide gene such as thymidine kinase (TK) of the HSV virus (herpesvirus) type I (Bonini, et al. (1997)Science276:1719-1724), a Fas-based “artificial suicide gene” (Thomis, et al. (2001)Blood97:1249-1257), orE. colicytosine deaminase gene which are activated by gancyclovir, AP1903, or 5-fluorocytosine, respectively. The suicide gene is advantageously included in the nucleic acid construct of the present invention to provide for the opportunity to ablate the transduced T cells in case of toxicity and to destroy the chimeric construct once a tumor has been reduced or eliminated. The use of suicide genes for eliminating transformed or transduced cells is well-known in the art. For example, Bonini, et al. ((1997)Science276:1719-1724) teach that donor lymphocytes transduced with the HSV-TK suicide gene provide antitumor activity in patients for up to one year and elimination of the transduced cells is achieved using ganciclovir. Further, Gonzalez, et al. ((2004)J. Gene Med.6:704-711) describe the targeting of neuroblastoma with cytotoxic T lymphocyte clones genetically modified to express a chimeric scFvFc: ζ immunoreceptor specific for an epitope on L1-CAM, wherein the construct further expresses the hygromycin thymidine kinase (HyTK) suicide gene to eliminate the transgenic clones. It is contemplated that the suicide gene can be expressed from the same promoter as the chimeric receptor or from a different promoter. Generally, however, nucleic acid sequences encoding the suicide protein and chimeric receptor reside on the same construct or vector. Expression of the suicide gene from the same promoter as the chimeric receptor can be accomplished using any well-known internal ribosome entry site (IRES). Suitable IRES sequences which can be used in the nucleic acid construct of the present invention include, but are not limited to, IRES from EMCV, c-myc, FGF-2, poliovirus and HTLV-1. By way of illustration, a nucleic acid construct for expressing a chimeric receptor can have the following structure: promoter→chimeric receptor→IRES→suicidal gene. Alternatively, the suicide gene can be expressed from a different promoter than that of the chimeric receptor (e.g., promoter 1→chimeric receptor→promoter 2→suicidal gene). The following non-limiting examples are presented to better illustrate the invention. Example 1: Mice and Cell Lines C57BL/6 mice were purchased from the National Cancer Institute, and all animal work was conducted in accordance with standard guidelines. Cell lines Bosc23, PT67, GP+E86, EG7 (H-2b), and YAC-1 were obtained from the American Type Culture Collection (ATCC, Rockville, MD). RMA cells (H-2b) originated from a Rauscher virus-induced C57BL/6 T-cell lymphoma (Ljunggren and Karre (1985)J. Exp. Med.162:1745-1759). RMAS-S is a sub-line of RMA which lacks MHC class-I surface expression (Karre, et al. (1986)Nature319:675-678). All packaging cells were grown in Dulbecco's modified Eagle medium (DMEM) with a high glucose concentration (4.5 gram/liter) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Hyclone, Logan, UT), 20 U/mL penicillin, 20 μg/mL streptomycin, 1 mM pyruvate, 10 mM HEPES, 0.1 mM non-essential amino acids, 50 μM 2-mercaptoethanol. RMA, EG7, RMA-S and YAC-1 cells were cultured in RPMI plus the same supplements described above. Example 2: Retroviral Vector Construction The full-length murine NKG2D cDNA was purchased from Open Biosystems (Huntsville, AL). Murine CD3ζ chain, Dap10 and Dap12 cDNAs were cloned by RT-PCR using RNAs from ConA- or IL-2 (1000 U/mL)-activated spleen cells as templates. Mouse NKG2D ligands Rae-1p and H60 were cloned from YAC-1 cells by RT-PCR. All PCR reactions were performed using high-fidelity enzyme Pfu or PFUULTRA™ (STRATAGENE@, La Jolla, CA). The oligonucleotides employed in these PCR reactions are listed in Table 9. TABLE 9SEQIDNo.PrimerSequenceNO:15′ wtNKG2DGCGAATTCGCCACCATGGCATTGATTCGTGATCGA823′ wtNKG2DGGCGCTCGAGTTACACCGCCCTTTTCATGCAGAT935′ chNKG2DGGCGAATTCGCATTGATTCGTGATCGAAAGTCT1045′ wtDAP10GCAAGTCGACGCCACCATGGACCCCCCAGGCTACC1153′ wtDAP10GGCGAATTCTCAGCCTCTGCCAGGCATGTTGAT1263′ chDAP10GGCAGAATTCGCCTCTGCCAGGCATGTTGATGTA1375′ wtDAP12GTTAGAATTCGCCACCATGGGGGCTCTGGAGCCCT1483′ wtDAP12GCAACTCGAGTCATCTGTAATATTGCCTCTGTG1595′ ATG-CD3ζGGCGTCGACACCATGAGAGCAAAATTCAGCAGGAG16103′ ATG-CD3ζGCTTGAATTCGCGAGGGGCCAGGGTCTGCATAT17115′ CD3ζ-TAAGCAGAATTCAGAGCAAAATTCAGCAGGAGTGC18123′ CD3ζ-TAAGCTTTCTCGAGTTAGCGAGGGGCCAGGGTCTGCAT19135′ Rae-1GCATGTCGACGCCACCATGGCCAAGGCAGCAGTGA20143′ Rae-1GCGGCTCGAGTCACATCGCAAATGCAAATGC21155′ H60GTTAGAATTCGCCACCATGGCAAAGGGAGCCACC22163′ H60GCGCTCGAGTCATTTTTTCTTCAGCATACACCAAG23 Restriction sites inserted for cloning purposes are underlined. Chimeric NKG2D was created by fusing the murine CD3 chain cytoplasmic region coding sequence (CD3′-CYP) to the full-length gene of murine NKG2D. Briefly, the SalI-EcoRI fragment of CD3′-CYP (with the initiation codon ATG at the 5′ end, primer numbers 9 and 10) and the EcoRI-XhoI fragment of NKG2D (without ATG, primer numbers 2 and 3) were ligated into the SalI/XhoI-digested pFB-neo retroviral vector (STRATAGENE®, La Jolla, CA). Similarly, chimeric Dap10 was generated by fusing the SalI-EcoRI fragment of full-length Dap10 (primer numbers 4 and 6) to the EcoRI-XhoI fragment of CD3ζ-CYP (primer numbers 11 and 12). Wild-type NKG2D (primer numbers 2 and 3), Dap10 (primer numbers 4 and 5) and Dap12 (primer numbers 7 and 8) fragments were inserted between the EcoRI and XhoI sites in pFB-neo. In some cases, a modified vector pFB-IRES-GFP was used to allow co-expression of green fluorescent protein (GFP) with genes of interest. pFB-IRES-GFP was constructed by replacing the 3.9 kb AvrUScaI fragment of pFB-neo with the 3.6kb AvrII/ScaI fragment of a plasmid GFP-RV(Ouyang, et al. (1998)Immunity9:745-755). Rae-1β (primer numbers 13 and 14) and H60 (primer numbers 15 and 16) cDNAs were cloned into pFB-neo. Constructs containing human NKD2D and human CD3ζ or murine Fc were prepared in the same manner using the appropriate cDNAs as templates. Example 3: Retrovirus Production and Transduction Eighteen hours before transfection, Bosc23 cells were plated in 25 cm2flasks at a density of 4×106cells per flask in 6 mL of DMEM-10. Transfection of retroviral constructs into Bosc23 cells was performed using LIPOFECTAMINE™ 2000 (INVITROGEN™, Carlsbad, CA) according to the manufacturer's instruction. Viral supernatants were collected 48 and 72 hours post-transfection and filtered (0.45 μm) before use. For generation of large scale, high-titer ecotropic vectors, the ecotropic viruses produced above were used to transduce the dualtropic packaging cell PT67 in the presence of polybrene (8 μg/mL). After three rounds of transduction, PT67 cells were selected in G418 (1 mg/mL) for 7 days. Dualtropic vectors were then used to transduce ecotropic cell line GP+E86. Through this process, the virus titer from pooled GP+E86 cells generally was over 1×106CFU/mL. Concentration of retroviruses by polyethylene glycol (PEG) was performed according to standard methods (Zhang, et al. (2004)Cancer Gene Ther.11:487-496; Zhang, et al. (2003)J. Hametother. Stem Cell Res.12:123-130). Viral stocks with high titer (1˜2×107CFU/mL) were used for transduction of T cells. Primary T cells from spleens of C57BL/6 (B6) mice were infected 18-24 hours after concanavalin A (ConA, 1 μg/mL) stimulation based on a well-established protocol (Sentman, et al. (1994)J. Immunol.153:5482-5490). Two days after infection, transduced primary T cells (0.5˜1×106/mL) were selected in RPMI-10 media containing G418 (0.5 mg/mL) plus 25 U/mL rHuIL-2 for an additional 3 days. Viable cells were isolated using HISTOPAQUE®-1083 (Sigma, St. Louise, MO) and expanded for 2 days without G418 before functional analyses. NKG2D ligand-expressing RMA (RMA/Rae-1 and RMA/H60) or RMA-S(RMA-S/Rae-1β) cells were established by retroviral transduction with dualtropic vectors from PT67. Example 4: Cytokine Production by Gene-Modified T Cells Gene-modified primary T cells (105) were co-cultured with an equal number of RMA, RMA/Rae-1β, RMA/H60 or YAC-1 cells in 96-well plates in complete media. After twenty-four hours, cell-free supernatants were collected. IFN-γ was assayed by ELISA using a DUOSET® ELISA kits (R&D, Minneapolis, MN). In some cases, T cells were cultured with equal numbers of irradiated (100 Gys) tumor cells for 3 days. Detection of other cytokines in culture was performed using a BIO-PLEX® kit (BIO-RAD®, Hercules, CA) based on the manufacturer's protocol. Example 5: Flow Cytometry For FACS analysis of NKG2D ligand expression, tumor cells were stained with mouse NKG2D-Ig fusion protein (R&D systems) according to manufacturer's instruction. Cell-surface phenotyping of transduced primary T cells was determined by direct staining with APC-anti-CD3e (clone 145-2C11; Pharmingen, San Diego, CA), PE-anti-NKG2D (clone 16-10A1; eBioscience, San Diego, CA) and FITC-anti-CD4 (Clone RM4-5; Caltag, Burlingame, CA) monoclonal antibodies. Cell fluorescence was monitored using a FACSCALIBER™ cytometer. Sorting of NKG2D ligand-expressing cells was performed on a FACSTARM™ cell sorter (Becton Dickinson, San Jose, CA). Example 6: Cytotoxicity Assay Three or four days after G418 selection (0.5 mg/mL), retroviral vector-transduced primary T cells were cultured in complete RPMI media containing 25 U/mL human IL-2 for an additional 2-3 days. Viable lymphocytes were recovered by centrifugation over HISTOPAQUE®-1083 (Sigma, St. Louis, MO) and used as effector cells. Lysis of target cells (RMA, RMA/Rae-1R, RMA/H60, EG7, RMA-S, RMA-S/Rae-1p, and YAC-1) was determined by a 4-hour51Cr release assay (Sentman, et al. (1994)supra). To block NKG2D receptors, anti-NKG2D (clone: CX5, 20 μg/mL) was included in those assays. The percentage of specific lysis was calculated as follows: % Specific lysis=[(Specific51Cr release−spontaneous51Cr release)/(Maximal51Cr release-spontaneous51Cr release)]×100. Example 7: Treatment of Mice with Genetically Modified T Cells For the determination of direct effects of chimeric NKG2D-bearing T cells (106) on the growth of RMA or RMA/Rae-1β tumors, chimeric NKG2D- or vector-transduced T cells were mixed with tumor cells (105) and then injected s.c. into the shaved right flank of recipient mice. Tumors were then measured using a caliper, and tumor areas were calculated. Animals were regarded as tumor-free when no tumor was found four weeks after inoculation. For the rechallenge experiments, mice were inoculated with 104RMA cells on the shaved left flank. In other experiments, transduced T cells were injected intravenously the day before s.c. inoculation of tumor cells. Mice were monitored for tumor size every two days and were sacrificed when tumor burden became excessive. Example 8: Statistical Analysis Differences between groups were analyzed using the student's t-test. p values<0.05 were considered significant.
51,160
11858977
DETAILED DESCRIPTION Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which the disclosure belongs. Although any method and material similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, preferred methods and materials are described. For the purposes of the present disclosure, the following terms are defined below. The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element. By “about” is meant a quantity, level, value, number, frequency, percentage, dimension, size, amount, weight or length that varies by as much as 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1% to a reference quantity, level, value, number, frequency, percentage, dimension, size, amount, weight or length. The term “activation,” as used herein, refers to the state of a cell that has been sufficiently stimulated to induce detectable cellular proliferation. Activation can also be associated with induced cytokine production and detectable effector functions. The term “activated T cells” refers to, among other things, T cells that are undergoing cell division. The term “antibody” is used in the broadest sense and refers to monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multi-specific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity or function. The antibodies in the present disclosure may exist in a variety of forms including, for example, polyclonal antibodies; monoclonal antibodies; Fv, Fab, Fab′, and F(ab′)2fragments; as well as single chain antibodies and humanized antibodies (Harlow et al., 1999, In: Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY; Harlow et al., 1989, In: Antibodies: A Laboratory Manual, Cold Spring Harbor, New York; Houston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; Bird et al., 1988, Science 242:423-426). The term “antibody fragments” refers to a portion of a full-length antibody, for example, the antigen binding or variable region of the antibody. Other examples of antibody fragments include Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multi-specific antibodies formed from antibody fragments. The term “Fv” refers to the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain in tight, non-covalent association. From the folding of these two domains emanates six hypervariable loops (3 loops each from the H and L chain) that contribute amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv including only three complementarity determining regions (CDRs) specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site (the dimer). An “antibody heavy chain,” as used herein, refers to the larger of the two types of polypeptide chains present in all antibody molecules in their naturally occurring conformations. An “antibody light chain,” as used herein, refers to the smaller of the two types of polypeptide chains present in all antibody molecules in their naturally occurring conformations. K and A light chains refer to the two major antibody light chain isotypes. The term “synthetic antibody” refers to an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage. The term also includes an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and the expression of the DNA molecule to obtain the antibody or to obtain an amino acid encoding the antibody. The synthetic DNA is obtained using technology that is available and well known in the art. The term “antigen” refers to a molecule that provokes an immune response, which may involve either antibody production, or the activation of specific immunologically-competent cells, or both. Antigens include any macromolecule, including all proteins or peptides, or molecules derived from recombinant or genomic DNA. For example, DNA including a nucleotide sequence or a partial nucleotide sequence encoding a protein or peptide that elicits an immune response, and therefore, encodes an “antigen” as the term is used herein. An antigen need not be encoded solely by a full-length nucleotide sequence of a gene. An antigen can be generated, synthesized or derived from a biological sample including a tissue sample, a tumor sample, a cell, or a biological fluid. The term “anti-tumor effect” as used herein, refers to a biological effect associated with a decrease in tumor volume, a decrease in the number of tumor cells, a decrease in the number of metastases, decrease in tumor cell proliferation, decrease in tumor cell survival, an increase in life expectancy of a subject having tumor cells, or amelioration of various physiological symptoms associated with the cancerous condition. An “anti-tumor effect” can also be manifested by the ability of the peptides, polynucleotides, cells, and antibodies in the prevention of the occurrence of tumor in the first place. The term “auto-antigen” refers to an endogenous antigen mistakenly recognized by the immune system as being foreign. Auto-antigens include cellular proteins, phosphoproteins, cellular surface proteins, cellular lipids, nucleic acids, glycoproteins, including cell surface receptors. The term “autologous” is used to describe a material derived from a subject which is subsequently re-introduced into the same subject. The term “allogeneic” is used to describe a graft derived from a different subject of the same species. As an example, a donor subject may be a related or unrelated to the recipient subject, but the donor subject has immune system markers which are similar to the recipient subject. The term “xenogeneic” is used to describe a graft derived from a subject of a different species. As an example, the donor subject is from a different species than a recipient subject, and the donor subject and the recipient subject can be genetically and immunologically incompatible. The term “cancer” is used to refer to a disease characterized by the rapid and uncontrolled growth of aberrant cells. Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body. Examples of various cancers include breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, lung cancer, and the like. Throughout this specification, unless the context requires otherwise, the words “comprise,” “includes” and “including” will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements. The phrase “consisting of” is meant to include, and is limited to, whatever follows the phrase “consisting of.” Thus, the phrase “consisting of” indicates that the listed elements are required or mandatory and that no other elements may be present. The phrase “consisting essentially of” is meant to include any element listed after the phrase and can include other elements that do not interfere with or contribute to the activity or action specified in the disclosure for the listed elements. Thus, the phrase “consisting essentially of” indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present depending upon whether or not they affect the activity or action of the listed elements. For example, if the element does not affect the expansion, function, or the phenotype of the cells, then the element is not required and is considered optional. The terms “complementary” and “complementarity” refer to polynucleotides (i.e., a sequence of nucleotides) related by the base-pairing rules. For example, the sequence “A-G-T,” is complementary to the sequence “T-C-A.” Complementarity may be “partial,” in which only some of the nucleic acids' bases are matched according to the base pairing rules, or there may be “complete” or “total” complementarity between the nucleic acids. The degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. The term “corresponds to” or “corresponding to” refers to (a) a polynucleotide having a nucleotide sequence that is substantially identical or complementary to all or a portion of a reference polynucleotide sequence or encoding an amino acid sequence identical to an amino acid sequence in a peptide or protein; or (b) a peptide or polypeptide having an amino acid sequence that is substantially identical to a sequence of amino acids in a reference peptide or protein. The term “co-stimulatory ligand,” refers to a molecule on an antigen presenting cell (e.g., an APC, dendritic cell, B cell, and the like) that specifically binds a cognate co-stimulatory molecule on a T cell, thereby providing a signal which, in addition to the primary signal provided by, for instance, binding of a TCR/CD3 complex with an MHC molecule loaded with peptide, mediates a T cell response, including at least one of proliferation, activation, differentiation, and other cellular responses. A co-stimulatory ligand can include B7-1 (CD80), B7-2 (CD86), PD-L1, PD-L2, 4-1BBL, OX4OL, inducible co-stimulatory ligand (ICOS-L), intercellular adhesion molecule (ICAM), CD3OL, CD40, CD70, CD83, HLA-G, MICA, MICB, HVEM, lymphotoxin beta receptor, 3/TR6, ILT3, ILT4, HVEM, a ligand for CD7, an agonist or antibody that binds the Toll ligand receptor, and a ligand that specifically binds with B7-H3. A co-stimulatory ligand also includes, inter alia, an agonist or an antibody that specifically binds with a co-stimulatory molecule present on a T cell, such as CD27, CD28, 4-1BB, OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, and a ligand that specifically binds CD83. The term “co-stimulatory molecule” refers to the cognate binding partner on a T cell that specifically binds with a co-stimulatory ligand, thereby mediating a co-stimulatory response by the T cell, such as proliferation. Co-stimulatory molecules include an MHC class I molecule, BTLA, and a Toll-like receptor. The term “co-stimulatory signal” refers to a signal, which in combination with a primary signal, such as TCR/CD3 ligation, leads to T cell proliferation and/or upregulation or downregulation of key molecules. The terms “disease” and “condition” may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out), and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians. The term “disease” is a state of health of a subject wherein the subject cannot maintain homeostasis, and wherein if the disease is not ameliorated then the subject's health continues to deteriorate. In contrast, a “disorder” in a subject is a state of health in which the animal is able to maintain homeostasis, but in which the animal's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal's state of health. The term “effective” refers to adequate to accomplish a desired, expected, or intended result. For example, an “effective amount” in the context of treatment may be an amount of a compound sufficient to produce a therapeutic or prophylactic benefit. The term “encoding” refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as a template for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system. Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence (except that a “T” is replaced by a “U”) and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA. The term “exogenous” refers to a molecule that does not naturally occur in a wild-type cell or organism but is typically introduced into the cell by molecular biological techniques. Examples of exogenous polynucleotides include vectors, plasmids, and/or man-made nucleic acid constructs encoding the desired protein. With regard to polynucleotides and proteins, the term “endogenous” or “native” refers to naturally-occurring polynucleotide or amino acid sequences that may be found in a given wild-type cell or organism. Also, a particular polynucleotide sequence that is isolated from a first organism and transferred to a second organism by molecular biological techniques is typically considered an “exogenous” polynucleotide or amino acid sequence with respect to the second organism. In specific embodiments, polynucleotide sequences can be “introduced” by molecular biological techniques into a microorganism that already contains such a polynucleotide sequence, for instance, to create one or more additional copies of an otherwise naturally-occurring polynucleotide sequence, and thereby facilitate overexpression of the encoded polypeptide. The term “expression or overexpression” refers to the transcription and/or translation of a particular nucleotide sequence into a precursor or mature protein, for example, driven by its promoter. “Overexpression” refers to the production of a gene product in transgenic organisms or cells that exceeds levels of production in normal or non-transformed organisms or cells. As defined herein, the term “expression” refers to expression or overexpression. The term “expression vector” refers to a vector including a recombinant polynucleotide including expression control (regulatory) sequences operably linked to a nucleotide sequence to be expressed. An expression vector includes sufficient cis-acting elements for expression; other elements for expression can be supplied by the host cell or in an in vitro expression system. Expression vectors include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentiviruses, retroviruses, adenoviruses, and adeno-associated viruses) that incorporate the recombinant polynucleotide. Viruses can be used to deliver nucleic acids into a cell in vitro and in vivo (in a subject). Examples of viruses useful for delivery of nucleic acids into cells include retrovirus, adenovirus, herpes simplex virus, vaccinia virus, and adeno-associated virus. There also exist non-viral methods for delivering nucleic acids into a cell, for example, electroporation, gene gun, sonoporation, magnetofection, and the use of oligonucleotides, lipoplexes, dendrimers, and inorganic nanoparticles. The term “homologous” refers to sequence similarity or sequence identity between two polypeptides or between two polynucleotides when a position in both of the two compared sequences is occupied by the same base or amino acid monomer subunit, e.g., if a position in each of two DNA molecules is occupied by adenine, then the molecules are homologous at that position. The percent of homology between two sequences is a function of the number of matching or homologous positions shared by the two sequences divided by the number of positions compared ×100. For example, if 6 of 10 of the positions in two sequences are matched or homologous, then the two sequences are 60% homologous. By way of example, the DNA sequences ATTGCC and TATGGC share 50% homology. A comparison is made when two sequences are aligned to give maximum homology. The term “immunoglobulin” or “Ig,” refers to a class of proteins, which function as antibodies. The five members included in this class of proteins are IgA, IgG, IgM, IgD, and IgE. IgA is the primary antibody that is present in body secretions, such as saliva, tears, breast milk, gastrointestinal secretions and mucus secretions of the respiratory and genitourinary tracts. IgG is the most common circulating antibody. IgM is the main immunoglobulin produced in the primary immune response in most subjects. It is the most efficient immunoglobulin in agglutination, complement fixation, and other antibody responses, and is important in defense against bacteria and viruses. IgD is the immunoglobulin that has no known antibody function but may serve as an antigen receptor. IgE is the immunoglobulin that mediates immediate hypersensitivity by causing the release of mediators from mast cells and basophils upon exposure to the allergen. The term “isolated” refers to a material that is substantially or essentially free from components that normally accompany it in its native state. The material can be a cell or a macromolecule such as a protein or nucleic acid. For example, an “isolated polynucleotide,” as used herein, refers to a polynucleotide, which has been purified from the sequences which flank it in a naturally-occurring state, e.g., a DNA fragment which has been removed from the sequences that are normally adjacent to the fragment. Alternatively, an “isolated peptide” or an “isolated polypeptide” and the like, as used herein, refer to in vitro isolation and/or purification of a peptide or polypeptide molecule from its natural cellular environment, and from association with other components of the cell. The term “substantially purified” refers to a material that is substantially free from components that are normally associated with it in its native state. For example, a substantially purified cell refers to a cell that has been separated from other cell types with which it is normally associated in its naturally occurring or native state. In some instances, a population of substantially purified cells refers to a homogenous population of cells. In other instances, this term refers simply to a cell that has been separated from the cells with which they are naturally associated in their natural state. In embodiments, the cells are cultured in vitro. In embodiments, the cells are not cultured in vitro. In the context of the present disclosure, the following abbreviations for the commonly occurring nucleic acid bases are used. “A” refers to adenosine, “C” refers to cytosine, “G” refers to guanosine, “T” refers to thymidine, and “U” refers to uridine. Unless otherwise specified, a “nucleotide sequence encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. The phrase nucleotide sequence that encodes a protein or an RNA may also include introns to the extent that the nucleotide sequence encoding the protein may in some version contain an intron(s). The term “lentivirus” refers to a genus of the Retroviridae family. Lentiviruses are unique among the retroviruses in being able to infect non-dividing cells; they can deliver a significant amount of genetic information into the DNA of the host cell, so they are one of the most efficient methods of a gene delivery vector. Moreover, the use of lentiviruses enables integration of the genetic information into the host chromosome resulting in stably transduced genetic information. HIV, SIV, and FIV are all examples of lentiviruses. Vectors derived from lentiviruses offer the means to achieve significant levels of gene transfer in vivo. The term “modulating,” refers to mediating a detectable increase or decrease in the level of a response in a subject compared with the level of a response in the subject in the absence of a treatment or compound, and/or compared with the level of a response in an otherwise identical but untreated subject. The term encompasses perturbing and/or affecting a native signal or response thereby mediating a beneficial therapeutic response in a subject, preferably, a human. Nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. The term “under transcriptional control” refers to a promoter being operably linked to and in the correct location and orientation in relation to a polynucleotide to control (regulate) the initiation of transcription by RNA polymerase and expression of the polynucleotide. The term “overexpressed” tumor antigen or “overexpression” of the tumor antigen is intended to indicate an abnormal level of expression of the tumor antigen in a cell from a disease area such as a solid tumor within a specific tissue or organ of the patient relative to the level of expression in a normal cell from that tissue or organ. Patients having solid tumor or a hematological malignancy characterized by overexpression of the tumor antigen can be determined by standard assays known in the art. Solid tumors are abnormal masses of tissue that usually do not contain cysts or liquid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors, such as sarcomas and carcinomas, include fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, lymphoid malignancy, pancreatic cancer, breast cancer, lung cancers, ovarian cancer, prostate cancer, hepatocellular carcinoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, pheochromocytomas sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, Wilms' tumor, cervical cancer, testicular tumor, seminoma, bladder carcinoma, Melanoma, and CNS tumors (such as a glioma (such as brainstem glioma and mixed gliomas), glioblastoma (also known as glioblastoma multiforme), astrocytoma, CNS lymphoma, germinoma, medulloblastoma, Schwannoma craniopharyogioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, neuroblastoma, retinoblastoma, and brain metastases). A solid tumor antigen is an antigen expressed on a solid tumor. In embodiments, solid tumor antigens are also expressed at low levels on healthy tissue. Examples of solid tumor antigens and their related disease tumors are provided in Table 1. TABLE 1Solid Tumor antigenDisease tumorPRLRBreast CancerCLCA1colorectal CancerMUC12colorectal CancerGUCY2Ccolorectal cancer and other digestivecancer typesGPR35colorectal CancerCR1LGastric CancerMUC 17Gastric CancerTMPRSS11Besophageal CancerMUC21esophageal CancerTMPRSS11Eesophageal CancerCD207bladder CancerSLC30A8pancreatic CancerCFC1pancreatic CancerSLC12A3Cervical CancerSSTR1Cervical tumorGPR27Ovary tumorFZD10Ovary tumorTSHRThyroid TumorSIGLEC15Urothelial cancerSLC6A3Renal cancerKISS1RRenal cancerQRFPRRenal cancer:GPR119Pancreatic cancerCLDN6Endometrial cancer/ Urothelial cancerUPK2Urothelial cancer (including bladder cancer)ADAM12Breast cancer, pancreatic cancer and the likeSLC45A3Prostate cancerACPPProstate cancerMUC21Esophageal cancerMUC16Ovarian cancerMS4A12Colorectal cancerALPPEndometrial cancerCEAColorectal carcinomaEphA2GliomaFAPMesoteliomaGPC3Lung squamous cell carcinomaIL13-Rα2GliomaMesothelinMetastatic cancerPSMAProstate cancerROR1Breast lung carcinomaVEGFR-IIMetastatic cancerGD2NeuroblastomaFR-αOvarian carcinomaErbB2CarcinomasbEpCAMCarcinomasaEGFRvIIIGlioma-GlioblastomaEGFRGlioma-NSCL cancertMUC1Cholangiocarcinoma, Pancreatic cancer,BreastPSCApancreas, stomach, or prostate cancerFCER2, GPR18, FCRLA,breast cancerCXCR5, FCRL3, FCRL2,HTR3A, and CLEC17ATRPMI, SLC45A2, andLymphomaSLC24A5DPEP3MelanomaKCNK16ovarian, testisLIM2 or KCNV2PancreaticSLC26A4thyroid cancerCD171NeuroblastomaGlypican-3SarcomaIL-13GliomaCD79a/bLymphomaMAGE A4Lung cancer and multiple cancer types The term “parenteral administration” of a composition includes, e.g., subcutaneous (s.c.), intravenous (i.v.), intramuscular (i.m.), intrasternal injection, or infusion techniques. The terms “patient,” “subject,” and “individual,” and the like are used interchangeably herein and refer to any human, or animal, amenable to the methods described herein. In certain non-limiting embodiments, the patient, subject, or individual is a human or animal. In embodiments, the term “subject” is intended to include living organisms in which an immune response can be elicited (e.g., mammals). Examples of subjects include humans, and animals, such as dogs, cats, mice, rats, and transgenic species thereof. A subject in need of treatment or in need thereof includes a subject having a disease, condition, or disorder that needs to be treated. A subject in need thereof also includes a subject that needs treatment for prevention of a disease, condition, or disorder. The term “polynucleotide” or “nucleic acid” refers to mRNA, RNA, cRNA, rRNA, cDNA or DNA. The term typically refers to a polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide. The term includes all forms of nucleic acids including single and double-stranded forms of nucleic acids. The terms “polynucleotide variant” and “variant” and the like refer to polynucleotides displaying substantial sequence identity with a reference polynucleotide sequence or polynucleotides that hybridize with a reference sequence under stringent conditions that are defined hereinafter. These terms also encompass polynucleotides that are distinguished from a reference polynucleotide by the addition, deletion or substitution of at least one nucleotide. Accordingly, the terms “polynucleotide variant” and “variant” include polynucleotides in which one or more nucleotides have been added or deleted or replaced with different nucleotides. In this regard, it is well understood in the art that certain alterations inclusive of mutations, additions, deletions, and substitutions can be made to a reference polynucleotide whereby the altered polynucleotide retains the biological function or activity of the reference polynucleotide or has increased activity in relation to the reference polynucleotide (i.e., optimized). Polynucleotide variants include, for example, polynucleotides having at least 50% (and at least 51% to at least 99% and all integer percentages in between, e.g., 90%, 95%, or 98%) sequence identity with a reference polynucleotide sequence described herein. The terms “polynucleotide variant” and “variant” also include naturally-occurring allelic variants and orthologs. The terms “polypeptide,” “polypeptide fragment,” “peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues and to variants and synthetic analogues of the same. Thus, these terms apply to amino acid polymers in which one or more amino acid residues are synthetic non-naturally occurring amino acids, such as a chemical analogue of a corresponding naturally occurring amino acid, as well as to naturally-occurring amino acid polymers. In certain aspects, polypeptides may include enzymatic polypeptides, or “enzymes,” which typically catalyze (i.e., increase the rate of) various chemical reactions. The term “polypeptide variant” refers to polypeptides that are distinguished from a reference polypeptide sequence by the addition, deletion, or substitution of at least one amino acid residue. In embodiments, a polypeptide variant is distinguished from a reference polypeptide by one or more substitutions, which may be conservative or non-conservative. In embodiments, the polypeptide variant comprises conservative substitutions and, in this regard, it is well understood in the art that some amino acids may be changed to others with broadly similar properties without changing the nature of the activity of the polypeptide. Polypeptide variants also encompass polypeptides in which one or more amino acids have been added or deleted or replaced with different amino acid residues. The term “promoter” refers to a DNA sequence recognized by the synthetic machinery of the cell or introduced synthetic machinery, required to initiate the specific transcription of a polynucleotide sequence. The term “expression control (regulatory) sequences” refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers. The term “bind,” “binds,” or “interacts with” refers to a molecule recognizing and adhering to a second molecule in a sample or organism but does not substantially recognize or adhere to other structurally unrelated molecules in the sample. The term “specifically binds,” as used herein with respect to an antibody, refers to an antibody which recognizes a specific antigen, but does not substantially recognize or bind other molecules in a sample. For example, an antibody that specifically binds an antigen from one species may also bind that antigen from one or more species. But, such cross-species reactivity does not itself alter the classification of an antibody as specific. In another example, an antibody that specifically binds an antigen may also bind different allelic forms of the antigen. However, such cross reactivity does not itself alter the classification of an antibody as specific. In some instances, the terms “specific binding” or “specifically binding,” can be used in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds a specific protein structure rather than to any protein. If an antibody is specific for epitope “A,” the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled “K” and the antibody, will reduce the amount of labeled A bound to the antibody. By “statistically significant,” it is meant that the result was unlikely to have occurred by chance. Statistical significance can be determined by any method known in the art. Commonly used measures of significance include the p-value, which is the frequency or probability with which the observed event would occur if the null hypothesis were true. If the obtained p-value is smaller than the significance level, then the null hypothesis is rejected. In simple cases, the significance level is defined at a p-value of 0.05 or less. A “decreased” or “reduced” or “lesser” amount is typically a “statistically significant” or a physiologically significant amount, and may include a decrease that is about 1.1, 1.2, 1.3, 1.4, 1.5, 1.6 1.7, 1.8, 1.9, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 or more times (e.g., 100, 500, 1000 times) (including all integers and decimal points in between and above 1, e.g., 1.5, 1.6, 1.7. 1.8, etc.) an amount or level described herein. The term “stimulation,” refers to a primary response induced by binding of a stimulatory molecule (e.g., a TCR/CD3 complex) with its cognate ligand thereby mediating a signal transduction event, such as signal transduction via the TCR/CD3 complex. Stimulation can mediate altered expression of certain molecules, such as downregulation of TGF-β, and/or reorganization of cytoskeletal structures. The term “stimulatory molecule” refers to a molecule on a T cell that specifically binds a cognate stimulatory ligand present on an antigen presenting cell. For example, a functional signaling domain derived from a stimulatory molecule is the zeta chain associated with the T cell receptor complex. The stimulatory molecule includes a domain responsible for signal transduction. The term “stimulatory ligand” refers to a ligand that when present on an antigen presenting cell (e.g., an APC, a dendritic cell, a B-cell, and the like.) can specifically bind with a cognate binding partner (referred to herein as a “stimulatory molecule”) on a cell, for example a T cell, thereby mediating a primary response by the T cell, including activation, initiation of an immune response, proliferation, and similar processes. Stimulatory ligands are well-known in the art and encompass, inter alia, an MHC Class I molecule loaded with a peptide, an anti-CD3 antibody, a superagonist anti-CD28 antibody, and a superagonist anti-CD2 antibody. The term “therapeutic” refers to a treatment and/or prophylaxis. A therapeutic effect is obtained by suppression, remission, or eradication of a disease state or alleviating the symptoms of a disease state. The term “therapeutically effective amount” refers to the amount of the subject compound that will elicit the biological or medical response of a tissue, system, or subject that is being sought by the researcher, veterinarian, medical doctor or another clinician. The term “therapeutically effective amount” includes that amount of a compound that, when administered, is sufficient to prevent the development of, or alleviate to some extent, one or more of the signs or symptoms of the disorder or disease being treated. The therapeutically effective amount will vary depending on the compound, the disease and its severity and the age, weight, etc., of the subject to be treated. The term “treat a disease” refers to the reduction of the frequency or severity of at least one sign or symptom of a disease or disorder experienced by a subject. The term “transfected” or “transformed” or “transduced” refers to a process by which an exogenous nucleic acid is transferred or introduced into the host cell. A “transfected” or “transformed” or “transduced” cell is one which has been transfected, transformed, or transduced with exogenous nucleic acid. The cell includes the primary subject cell and its progeny. The term “vector” refers to a polynucleotide that comprises an isolated nucleic acid and which can be used to deliver the isolated nucleic acid to the interior of a cell. Numerous vectors are known in the art including linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses. Thus, the term “vector” includes an autonomously replicating plasmid or a virus. The term also includes non-plasmid and non-viral compounds which facilitate the transfer of nucleic acid into cells, such as, for example, polylysine compounds, liposomes, and the like. Examples of viral vectors include adenoviral vectors, adeno-associated virus vectors, retroviral vectors, and others. For example, lentiviruses are complex retroviruses, which, in addition to the common retroviral genes gag, pol, and env, contain other genes with regulatory or structural function. Lentiviral vectors are well known in the art. Some examples of lentivirus include the Human Immunodeficiency Viruses: HIV-1, HIV-2, and the Simian Immunodeficiency Virus: SIV. Lentiviral vectors have been generated by multiply attenuating the HIV virulence genes, for example, the genes env, vif, vpr, vpu, and nef are deleted making the vector biologically safe. Ranges: throughout this disclosure, various aspects of the disclosure can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range. A “chimeric antigen receptor” (CAR) molecule is a recombinant polypeptide including at least an extracellular domain, a transmembrane domain and a cytoplasmic domain or intracellular domain. In embodiments, the domains of the CAR are on the same polypeptide chain, for example a chimeric fusion protein. In embodiments, the domains are on different polypeptide chains, for example the domains are not contiguous. The extracellular domain of a CAR molecule includes an antigen binding domain. The antigen binding domain is for expanding and/or maintaining the modified cells, such as a CAR T cell or for killing a tumor cell, such as a solid tumor. In embodiments, the antigen binding domain for expanding and/or maintaining modified cells binds an antigen, for example, a cell surface molecule or marker, on the surface of a WBC. In embodiments, the WBC is at least one of GMP (granulocyte macrophage precursor), MDP (monocyte-macrophage/dendritic cell precursors), cMoP (common monocyte precursor), basophil, eosinophil, neutrophil, SatM (Segerate-nucleus-containing atypical monocyte), macrophage, monocyte, CDP (common dendritic cell precursor), cDC (conventional D.C.), pDC (plasmacytoid D.C.), CLP (common lymphocyte precursor), B cell, ILC (Innate Lymphocyte), Natural Killer (NK) cell, megakaryocyte, myeloblast, pro-myelocyte, myelocyte, meta-myelocyte, band cells, lymphoblast, prolymphocyte, monoblast, megakaryoblast, promegakaryocyte, megakaryocyte, platelets, or MSDC (Myeloid-derived suppressor cell). In embodiments, the WBC is a granulocyte, monocyte and or lymphocyte. In embodiments, the WBC is a lymphocyte, for example, a B cell. In embodiments, the WBC is a B cell. In embodiments, the cell surface molecule of a B cell includes CD19, CD22, CD20, BCMA, CD5, CD7, CD2, CD16, CD56, CD30, CD14, CD68, CD11 b, CD18, CD169, CD1c, CD33, CD38, CD138, or CD13. In embodiments, the cell surface molecule of the B cell is CD19, CD20, CD22, or BCMA. In embodiments, the cell surface molecule of the B cell is CD19. The cells described herein, including modified cells such as CAR cells and modified T cells can be derived from stem cells. Stem cells may be adult stem cells, embryonic stem cells, more particularly non-human stem cells, cord blood stem cells, progenitor cells, bone marrow stem cells, induced pluripotent stem cells, totipotent stem cells or hematopoietic stem cells. A modified cell may also be a dendritic cell, a NK-cell, a B-cell or a T cell selected from the group consisting of inflammatory T-lymphocytes, cytotoxic T-lymphocytes, regulatory T lymphocytes or helper T-lymphocytes. In embodiments, Modified cells may be derived from the group consisting of CD4+ T lymphocytes and CD8+ T lymphocytes. Prior to expansion and genetic modification of the cells, a source of cells may be obtained from a subject through a variety of non-limiting methods. T cells may be obtained from a number of non-limiting sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. In embodiments, any number of T cell lines available and known to those skilled in the art, may be used. In embodiments, modified cells may be derived from a healthy donor, from a patient diagnosed with cancer or from a patient diagnosed with an infection. In embodiments, a modified cell is part of a mixed population of cells which present different phenotypic characteristics. A population of cells refers to a group of two or more cells. The cells of the population could be the same, such that the population is a homogenous population of cells. The cells of the population could be different, such that the population is a mixed population or a heterogeneous population of cells. For example, a mixed population of cells could include modified cells comprising a first CAR and cells comprising a second CAR, wherein the first CAR and the second CAR bind different antigens. The term “stem cell” refers to any of certain types of cell which have the capacity for self-renewal and the ability to differentiate into other kind(s) of cell. For example, a stem cell gives rise either to two daughter stem cells (as occurs in vitro with embryonic stem cells in culture) or to one stem cell and a cell that undergoes differentiation (as occurs e.g. in hematopoietic stem cells, which give rise to blood cells). Different categories of stem cells may be distinguished on the basis of their origin and/or on the extent of their capacity for differentiation into other types of cell. For example, stem cells may include embryonic stem (E.S.) cells (i.e., pluripotent stem cells), somatic stem cells, induced pluripotent stem cells, and any other types of stem cells. The pluripotent embryonic stem cells are found in the inner cell mass of a blastocyst and have an innate capacity for differentiation. For example, pluripotent embryonic stem cells have the potential to form any type of cell in the body. When grown in vitro for long periods of time, E.S. cells maintain pluripotency as progeny cells retain the potential for multilineage differentiation. Somatic stem cells can include fetal stem cells (from the fetus) and adult stem cells (found in various tissues, such as bone marrow). These cells have been regarded as having a capacity for differentiation that is lower than that of the pluripotent E.S. cells—with the capacity of fetal stem cells being greater than that of adult stem cells. Somatic stem cells apparently differentiate into only a limited number of types of cells and have been described as multipotent. The “tissue-specific” stem cells normally give rise to only one type of cell. For example, embryonic stem cells may be differentiated into blood stem cells (e.g., Hematopoietic stem cells (HSCs)), which may be further differentiated into various blood cells (e.g., red blood cells, platelets, white blood cells, etc.). Induced pluripotent stem cells (i.e., iPS cells or iPSCs) may include a type of pluripotent stem cell artificially derived from a non-pluripotent cell (e.g., an adult somatic cell) by inducing an expression of specific genes. Induced pluripotent stem cells are similar to natural pluripotent stem cells, such as embryonic stem (E.S.) cells, in many aspects, such as the expression of certain stem cell genes and proteins, chromatin methylation patterns, doubling time, embryoid body formation, teratoma formation, viable chimera formation, and potency and differentiability. Induced pluripotent cells can be obtained from adult stomach, liver, skin, and blood cells. In embodiments, the antigen binding domain for killing a tumor, binds an antigen on the surface of a tumor, for example a tumor antigen or tumor marker. Tumor antigens are proteins that are produced by tumor cells that elicit an immune response, particularly T cell mediated immune responses. Tumor antigens are well known in the art and include, for example, tumor associated MUC1 (tMUC1), a glioma-associated antigen, carcinoembryonic antigen (CEA), β-human chorionic gonadotropin, alphafetoprotein (AFP), lectin-reactive AFP, thyroglobulin, RAGE-1, MN-CA IX, human telomerase reverse transcriptase, RU1, RU2 (AS), intestinal carboxyl esterase, mut hsp70-2, M-CSF, prostase, prostate-specific antigen (PSA), PAP, NY-ESO-1, LAGE-1a, p53, prostein, PSMA, Her2/neu, surviving, telomerase, prostate-carcinoma tumor antigen-1 (PCTA-1), MAGE, ELF2M, neutrophil elastase, ephrinB2, CD22, insulin growth factor (IGF)-I, IGF-II, IGF-I receptor, CD19, and mesothelin. For example, when the tumor antigen is CD19, the CAR thereof can be referred to as CD19 CAR (19CAR, CD19CAR, or CD19-CAR) which is a CAR molecule that includes an antigen binding domain that binds CD19. In embodiments, the extracellular antigen binding domain of a CAR includes at least one scFv or at least a single domain antibody. As an example, there can be two scFvs on a CAR. The scFv includes a light chain variable (V.L.) region and a heavy chain variable (V.H.) region of a target antigen-specific monoclonal antibody joined by a flexible linker. Single chain variable region fragments can be made by linking light and/or heavy chain variable regions by using a short linking peptide (Bird et al., Science 242:423-426, 1988). An example of a linking peptide is the G.S. linker having the amino acid sequence (GGGGS)3(SEQ ID NO: 118), which bridges approximately 3.5 nm between the carboxy terminus of one variable region and the amino terminus of the other variable region. Linkers of other sequences have been designed and used (Bird et al., 1988, supra). In general, linkers can be short, flexible polypeptides and preferably comprised of about 20 or fewer amino acid residues. The single chain variants can be produced either recombinantly or synthetically. For synthetic production of scFv, an automated synthesizer can be used. For recombinant production of scFv, a suitable plasmid containing a polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect, or mammalian cells, or prokaryotic, such asE. coli. Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides. The resultant scFv can be isolated using standard protein purification techniques known in the art. The cytoplasmic domain of the CAR molecules described herein includes one or more co-stimulatory domains and one or more signaling domains. The co-stimulatory and signaling domains function to transmit the signal and activate molecules, such as T cells, in response to antigen binding. The one or more co-stimulatory domains are derived from stimulatory molecules and/or co-stimulatory molecules, and the signaling domain is derived from a primary signaling domain, such as the CD3 zeta domain. In embodiments, the signaling domain further includes one or more functional signaling domains derived from a co-stimulatory molecule. In embodiments, the co-stimulatory molecules are cell surface molecules (other than antigens receptors or their ligands) that are required for activating a cellular response to an antigen. In embodiments, the co-stimulatory domain includes the intracellular domain of CD27, CD28, 4-1BB, OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, or any combination thereof. In embodiments, the signaling domain includes a CD3 zeta domain derived from a T cell receptor. The CAR molecules described herein also include a transmembrane domain. The incorporation of a transmembrane domain in the CAR molecules stabilizes the molecule. In embodiments, the transmembrane domain of the CAR molecules is the transmembrane domain of a CD28 or 4-1BB molecule. Between the extracellular domain and the transmembrane domain of the CAR, there may be incorporated a spacer domain. As used herein, the term “spacer domain” generally means any oligo- or polypeptide that functions to link the transmembrane domain to the extracellular domain and/or the cytoplasmic domain on the polypeptide chain. A spacer domain may include up to 300 amino acids, preferably 10 to 100 amino acids, and most preferably 25 to 50 amino acids. Embodiments relate to a polynucleotide encoding a modified component of the TCR-CD3 complex. Embodiments relate to a vector comprising the polynucleotide. Embodiments relate to a cell modified comprising the polynucleotide. Embodiments relate to a modified cell engineered to express a modified component of the TCR-CD3 complex, wherein the modified cell includes an antigen binding molecule. Embodiments relate to a method or use of polynucleotide, the method comprising providing a viral particle (e.g., AAV, lentivirus or their variants) comprising a vector genome, the vector genome comprising the polynucleotide; and administering an amount of the viral particle to a subject such that the polynucleotide is expressed in the subject. In embodiments, the AAV preparation may include AAV vector particles, empty capsids, and host cell impurities, thereby providing an AAV product substantially free of AAV empty capsids. Embodiments relate to a pharmaceutical composition comprising the population of the cells. Embodiments relate to a method of causing or eliciting T cell response in a subject in need thereof and/or treating a tumor of the subject, the method comprising administering an effective amount of the composition. In embodiments, the polynucleotide comprises at least one of the sequences listed in Table 2. For example, the antigen-specific T cell receptor (TCR) is composed of a disulfide-linked heterodimer, containing two clonally distributed, integral membrane glycoprotein chains, α, and β, or γ and δ non-covalently associated with a complex of low molecular weight invariant proteins, commonly designated as CD3 (i.e., TCR-CD3 complex). The TCR α and β chains determine antigen specificities, and the CD3 structures are thought to represent accessory molecules that may be the transducing elements of activation signals initiated upon binding of the TCR αβ to its ligand. TCR complex interacts with small peptidic antigen presented in the context of major histocompatibility complex (MHC) proteins. The MHC proteins represent another highly polymorphic set of molecules randomly dispersed throughout the species. The modified components of the TCR-CD3 complex can be expressed in TIL or TCR T as a whole and can replace the peptide chain in CD3 so that when CD3 is activated, there will be a signal of a co-stimulatory domain and enhance TIL/TCR-T. Ordinary TCR only activates CD3, the designed TCR-CD3 complex here has been added with a co-stimulation domain, and the signal is stronger. The designed TCR-CD3 complex may be associated with the uses of a CAR and can be a general-purpose component. And when CD3 is activated, there will be a co-stimulus domain signal, which can enhance the killing effect of TCR-T. In embodiments, the TCR-CD3 complex comprises TCRα, TCRβ, CD3γ, ζ-chain, CD3ε, and CD3δ. In embodiments, the TCR-CD3 complex comprises TCRγ, TCRδ, CD3γ, ζ-chain, CD3ε, and CD3δ. In embodiments, the modified component of the TCR-CD3 complex comprises components of TCR-CD3 complex linked to one or more co-stimulatory signaling domains. In embodiments, the one or more co-stimulatory signaling domains comprise one or more functional signaling domains of one or more proteins selected from the group consisting of CD27, CD28, 4-1BB(CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM(LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, vFLIP K13, K13-opt, a NEMO mutant, a NEMO-fusion protein, IKKI-S176E-S180E, IKK2-S177E-S181E, RIP, IKKα, IKKβ, Tcl-I, MyD88-L265,ally NF-KB actuating protein or protein fragment. any inhibitor of an inhibiior of NF-kB, pathway, any gene editing sysiem capable of selectively activating NF-κB. In embodiments, the one or more co-stimulatory signaling domains comprise one or more functional signaling domains of one or more proteins selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D. In embodiments, the modified component of the TCR-CD3 complex comprises CD3γ, ζ-chain, CD3ε, and/or CD3δ. In embodiments, the modified component of the TCR-CD3 complex comprises CD3γ linked to one or more co-stimulatory signaling domains. In embodiments, the modified component of the TCR-CD3 complex comprises CD3ζ linked to one or more co-stimulatory signaling domains. In embodiments, the modified component of the TCR-CD3 complex comprises CD3ε linked to one or more co-stimulatory signaling domains. In embodiments, the modified component of the TCR-CD3 complex comprises CD3δ linked to one or more co-stimulatory signaling domains. In embodiments, the CD3γ, ζ-chain, CD3ε, and/or CD3δ and the one or more co-stimulatory signaling domains are linked by a linker (e.g., G.S. linker). In embodiments, the one or more co-stimulatory signaling domains comprise at least two co-stimulatory signaling domains. In embodiments, at least two co-stimulatory signaling domains are linked by a linker (e.g., G.S. linker). In embodiments, the modified component of the TCR-CD3 complex is the modified CD3 domain. In embodiments, the modified TCR-CD3 complex is overexpressed by the modified cell, and/or the modified CD3 domain is overexpressed by the modified cell. In embodiments, the expression of the modified component of the TCR-CD3 complex may be regulated by an inducible expression system. The inducible expression system allows for a temporal and spatial controlled activation and/or expression of genes. For example, Tetracycline-Controlled Transcriptional Activation is a method of inducible gene expression where transcription is reversibly turned on or off in the presence of the antibiotic tetracycline or one of its derivatives (e.g., doxycycline). For example, an inducible suicide gene expression system allows for a temporal and spatial controlled activation and/or expression of a suicide gene, which causes a cell to kill itself through apoptosis. In embodiments, the modified cells comprise a nucleic acid sequence encoding a reverse tetracycline transactivator (rtTA). In embodiments, the expression of one or more molecules is regulated by the rtTA, such that the modified component of the TCR-CD3 complex is expressed in the presence of tetracycline. In embodiments, a concentration of tetracycline in the cell culture medium is not less than about 2 μg/ml. In embodiments, the tetracycline is selected from the group consisting of tetracycline, demeclocycline, meclocycline, doxycycline, lymecycline, methacycline, minocycline, oxytetracycline, rolitetracycline, and chlortetracycline. In embodiments, the tetracycline is doxycycline. In embodiments, the inducible suicide system is an HSV-TK system or an inducible caspase-9 system. In embodiments, the modified cells comprise a nucleic acid sequence encoding a suicide gene, such that when the modified cells are in the presence of a nucleoside analogue in a manner permitting expression of the suicide gene, to render the nucleoside analogue cytotoxic to the modified cells. In embodiments, the suicide gene is selected from the group consisting of thymidine kinase of herpes simplex virus, thymidine kinase of varicella zoster virus, and bacterial cytosine deaminase. In embodiments, the suicide gene is thymidine kinase of herpes simplex virus. In embodiments, the nucleoside analogue is selected from the group consisting of ganciclovir, acyclovir, buciclovir, famciclovir, penciclovir, valciclovir, trifluorothymidine, 1-[2-deoxy, 2-fluoro, beta-D-arabino furanosyl]-5-iodouracil, ara-A, araT 1-beta-D-arabinofuranoxyl thymine, 5-ethyl-2′-deoxyuridine, 5-iodo-5′-amino-2,5′-dideoxpridine, idoxuridine, AZT, AIU, dideoxycytidine, and AraC. In embodiments, the nucleoside analogue is ganciclovir. In embodiments, the expression of the modified component of the TCR-CD3 complex is regulated by one or more promoters. In embodiments, the polynucleotide comprises a promoter comprising a binding site for a transcription modulator that modulates the expression and/or secretion of the modified component of the TCR-CD3 complex in the cell. For example, the transcription modulator is or includes Hif1a, NFAT, FOXP3, and/or NFkB. For example, the modified component of the TCR-CD3 complex comprises at least one co-stimulatory signaling domain associated with an oxygen-sensitive polypeptide domain, and the oxygen-sensitive polypeptide domain comprises HIF VHL binding domain. In embodiments, the polynucleotide may integrate into the genome of the modified cell, and descendants of the modified cell will also express the polynucleotide, resulting in a stably transfected modified cell. In embodiments, the modified cell may express the polynucleotide encoding the CAR, but the polynucleotide does not integrate into the genome of the modified cell such that the modified cell expresses the transiently transfected polynucleotide for a finite period of time (e.g., several days), after which the polynucleotide is lost through cell division or other factors. For example, the polynucleotide is present in the modified cell in a recombinant DNA construct, in an mRNA, or in a viral vector, and/or the polynucleotide is an mRNA, which is not integrated into the genome of the modified cell. Embodiments related to a method or use of polynucleotide, the method comprising providing a viral particle (e.g., AAV, lentivirus or their variants) comprising a vector genome, the vector genome comprising the polynucleotide encoding the one more molecules and a polynucleotide encoding a binding molecule, the polynucleotide operably linked to an expression control element conferring transcription of the polynucleotides; and administering an amount of the viral particle to a subject such that the polynucleotide is expressed in the subject. In embodiments, the AAV preparation may include AAV vector particles, empty capsids, and host cell impurities, thereby providing an AAV product substantially free of AAV empty capsids. Embodiments relate to a method or use of polynucleotide. The method or use includes: providing a viral particle (e.g., AAV, lentivirus or their variants) comprising a vector genome, the vector genome comprising the polynucleotide, wherein the polynucleotide is operably linked to an expression control element conferring transcription of the polynucleotide, and administering an amount of the viral particle to the subject such that the polynucleotide is expressed in the subject. In embodiments, the AAV preparation may include AAV vector particles, empty capsids, and host cell impurities, thereby providing an AAV product substantially free of AAV empty capsids. More information of the administration and preparation of the viral particle may be found at the U.S. Pat. No. 9,840,719 and Milani et al., Sci. Transl. Med. 11, eaav7325 (2019) May 22, 2019, which are incorporated herein by reference. In embodiments, the bioreactor may be inoculated at a cell density of approximately 0.5×106cells/mL with viability greater than 95%. When the cell density reaches approximately 1.0×106cells/ml, the cells may be transfected with the PEI/DNA complexes (polyplexes) with a PEI to DNA ratio of 2:1. At the time of harvest, AAV from the cell culture in the bioreactor may be released using the Triton X-100 method. All solutions may be added directly to the bioreactor, and the lysate was centrifuged at 4000×g for 20 min. The supernatant may be stored at −80° C. for further processing. AAV may be further purified. For example, AAV samples (12.3 mL) may be purified by overlaying them on top of series of step gradients using 15, 25, 40, and 54% iodixanol concentrations containing 1, 5, 7, and 5 mL, respectively. The 15% iodixanol concentration also contains 1 M NaCl to avoid aggregation of AAV with other cellular proteins and negatively charged nuclear components. After the completion of centrifugation, 5 mL may be withdrawn from 2 mm below the 40/54 interface marked before starting the ultracentrifugation at 385,000×g for 1 h 45 min in Sorvals T-865 rotor in Sorval Ultracentrifuge. The viral vectors may then be quantified. For example, vectors AAV infectivity may be determined by the gene transfer assay (GTA)using GFP as a reporter gene in all cases. AAV infectivity assay where the sample may be diluted before addition to the cells to have the GFP positive cells in the range of 2-20% to assure that only a single virus has entered the cell for GFP expression. The GFP-positive cells may be quantified by FACS using HEK293 cells in suspension. The AAV may be then administrated to a subject. For example, AAV may be diluted in 0.9% sterile NaCl saline solution (supplemented with 0.25% human serum albumin [HSA]) for infusion in patients, and the final volume of infusion will be calculated based on the patient's weight as 3 mL/kg. In embodiments, the modified cell comprises the antigen binding molecule, the antigen binding molecule is chimeric antigen receptor (CAR), which comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain. In embodiments, the antigen-binding domain binds to a tumor antigen is selected from a group consisting of: TSHR, CD19, CD123, CD22, CD30, CD171, CS-1, CLL-1, CD33, EGFRvIII, GD2, GD3, BCMA, Tn Ag, PSMA, ROR1, FLT3, FAP, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, Mesothelin, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-beta, SSEA-4, CD20, Folate receptor alpha, ERBB2 (Her2/neu), MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, IGF-I receptor, CAIX, LMP2, gp100, bcr-abl, tyrosinase, EphA2, Fucosyl GM1, sLe, GM3, TGS5, HMWMAA, o-acetyl-GD2, Folate receptor beta, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, Polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, NY-ESO-1, LACE-1a, MAGE-A1, legumain, HPV E6, E7, MAGE A1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53, p53 mutant, prostein, survivin and telomerase, PCTA-1/Galectin 8, MelanA/MART1, Ras mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, Androgen receptor, Cyclin B1, MYCN, RhoC, TRP-2, CYP1B1, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxyl esterase, mut hsp70-2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1. In embodiments, the intracellular signaling domain comprises a co-stimulatory signaling domain, or a primary signaling domain and a co-stimulatory signaling domain, wherein the co-stimulatory signaling domain comprises a functional signaling domain of a protein selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D In embodiments, the modified cell comprises the antigen binding molecule, the antigen binding molecule is a modified TCR. In embodiments, the TCR is derived from spontaneously occurring tumor-specific T cells in patients. In embodiments, the TCR binds to a tumor antigen. In embodiments, the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1. In embodiments, the TCR comprises TCRγ and TCRδ Chains or TCRα and TCRβ chains, or a combination thereof. In embodiments, the first antigen binding domain is on a CAR, and the second antigen binding domain is on a T Cell Receptor (TCR). In embodiments, the TCR is a modified TCR. In embodiments, the TCR is derived from spontaneously occurring tumor-specific T cells in patients. In embodiments, the TCR binds a tumor antigen. In embodiments, the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1. In embodiments, a T cell clone that expresses a TCR with a high affinity for the target antigen may be isolated. Tumor-infiltrating lymphocytes (TILs) or peripheral blood mononuclear cells (PBMCs) can be cultured in the presence of antigen-presenting cells (APCs) pulsed with a peptide representing an epitope known to elicit a dominant T cell response when presented in the context of a defined HLA allele. High-affinity clones may be then selected on the basis of MHC-peptide tetramer staining and/or the ability to recognize and lyse target cells pulsed with low titrated concentrations of cognate peptide antigen. After the clone has been selected, the TCRα and TCRβ chains or TCRγ and TCRδ chains are identified and isolated by molecular cloning. For example, for TCRα and TCRβ chains, the TCRα and TCRβ gene sequences are then used to generate an expression construct that ideally promotes stable, high-level expression of both TCR chains in human T cells. The transduction vehicle, for example, a gammaretrovirus or lentivirus, can then be generated and tested for functionality (antigen specificity and functional avidity) and used to produce a clinical lot of the vector. An aliquot of the final product can then be used to transduce the target T cell population (generally purified from patient PBMCs), which is expanded before infusion into the patient. Various methods may be implemented to obtain genes encoding tumor-reactive TCR. More information is provided in Kershaw et al., Clin Transl Immunology. 2014 May; 3(5): e16. In embodiments, specific TCR can be derived from spontaneously occurring tumor-specific T cells in patients. Antigens included in this category include the melanocyte differentiation antigens MART-1 and gp100, as well as the MAGE antigens and NY-ESO-1, with expression in a broader range of cancers. TCRs specific for viral-associated malignancies can also be isolated, as long as viral proteins are expressed by transformed cells. Malignancies in this category include liver and cervical cancer, associated with hepatitis and papilloma viruses, and Epstein-Barr virus-associated malignancies. In embodiments, target antigens of the TCR may include CEA (e.g., for colorectal cancer), gp100, MART-1, p53 (e.g., for Melanoma), MAGE-A3 (e.g., Melanoma, esophageal and synovial sarcoma), NY-ESO-1 (e.g., for Melanoma and sarcoma as well as Multiple myelomas). In embodiments, preparation and transfusion of tumor-infiltrating lymphocytes (TIL) may be implemented by the following. For example, tumor tissue comes from surgical or biopsy specimens, may be obtained under aseptic conditions, and transported to the cell culture chamber in an icebox. Necrotic tissue and adipose tissue may be removed. The tumor tissue may be cut into small pieces of about 1-3 cubic millimeters. Collagenase, hyaluronidase, and DNA enzyme may be added and digested overnight at 4° C. Filtering with 0.2 um filter, cells may be separated and collected by lymphocyte separation fluid, 1500 rpm for 5 min. Expanding the cells with a culture medium comprising PHA, 2-mercaptoethanol, and a CD3 monoclonal antibody, a small dose of IL-2 (10-20 IU/ml) may be added to induce activation and proliferation. According to the growth situation, the cell density may be carefully detected and maintained within the range of 0.5-2×106/ml under the condition of 37° C. and 5% CO2 for 7-14 days. TIL positive cells have the ability to kill homologous cancer cell may be screened out by co-culture. The positive cells may be amplified in a serum-free medium containing a high dose of IL2 (5000-6000 IU/ml) until greater than 1×1011TILs may be obtained. To administer TILs, they may be first collected in saline using continuous-flow centrifugation and then filtered through a platelet-administration set into a volume of 200-300 ml containing 5% albumin and 450 000 IU of IL-2. The TILs may be infused into patients through a central venous catheter over a period of 30-60 minutes. In embodiments, TILs may be often infused in two to four separate bags; the infusions may be separated by several hours. If more than approximately 1.5×1011TILs may be administered, they may be generally given on 2 successive days. In embodiments, the cell is an immune effector cell (e.g., a population of immune effector cells). In embodiments, the immune effector cell is a T cell or an NK Cell. In embodiments, the immune effector cell is a T cell. In embodiments, the T cell is a CD4+ T cell, a CD8+ T cell, or a combination thereof. In embodiments, the cell is a human cell. In embodiments, the enhanced expression and/or function of the modified component of TCR-CD3 complex is implemented by introducing a nucleic acid sequence encoding the modified component of TCR-CD3 complex and/or the binding molecule, which is present in the modified cell in a recombinant DNA construct, in an mRNA, or in a viral vector. In embodiments, the nucleic acid sequence is an mRNA, which is not integrated into the genome of the modified cell. In embodiments, the nucleic acid sequence is associated with an oxygen-sensitive polypeptide domain. In embodiments, the oxygen-sensitive polypeptide domain comprises HIF VHL binding domain. In embodiments, the nucleic acid sequence is regulated by a promoter comprising a binding site for a transcription modulator that modulates the expression and/or secretion of the therapeutic agent in the cell. In embodiments, the transcription modulator is or includes Hif1a, NFAT, FOXP3, and/or NFkB. Embodiments relate to compositions and methods for enhancing modified cells (e.g., immune cells) metabolism in the tumor microenvironment. For example, the metabolism of lactic acid (e.g, transporters MCT1, and MCT4) may be enhanced for lactic acid metabolism. Anaerobic and aerobic respiration and mitochondrial function and/or the metabolism of amino acids may be enhanced. The modified cells have enhanced metabolism by enhancing the pathway of metabolizing lactic acid (e.g., uses of transporters MCT1, MCT4 to enhance lactic acid metabolism) (e.g., molecules listed in Table 7), anaerobic and aerobic respiration mitochondrial function (e.g., molecules listed in Table 8), and/or the metabolism of amino acids (e.g., molecules listed in Table 9). Conditions of the tumor microenvironment (such as hypoxia, high acid, etc.) inhibit T cell viability, and enhancing the function of T cell monocarboxylate transporter can effectively help T cells survive in the tumor microenvironment. MCT1 is normally expressed on T cells, regulates the two-way transport of lactic acid inward and outward, and is also expressed in tumor cells. MCT4 is highly expressed in some tumor cells, regulates the outward transport of lactic acid, and does not express on normal T cells. MCT2 is expressed on normal T cells and is expressed on some other cells in the body, regulating the transport of lactic acid into cells. CD147 is an accessory protein of MCT family proteins, which regulates the correct localization of MCT family proteins on the cell membrane. LDHB converts lactic acid to pyruvate. Pyruvate can enter the mitochondria through the pyruvate transporter (MPC) on the mitochondrial membrane and eventually oxidize and decompose through mitochondria. By knockdown/out MCT1/2, overexpression of MCT3 allows lactic acid not to enter T cells. By overexpressing MCT1/2, knockdown/out MCT3, and over-expressing LDHB and MPC, it can enhance the entry of lactic acid into T cells and enhance the metabolism of lactic acid, helping immune cells to enhance their effects in the face of solid tumors. The oxidative function of mitochondria may be implemented by overexpressing mitochondrial proteins such as Frataxin, HBA, HBB, HBD, HBE, and/or HBG, etc., it promotes the synthesis of heme and hemoglobin and enhances the mitochondrial oxygen storage capacity; By over-expressing TOMM20, TOMM22, TOMM40, and/or TOM70 to promote the assembly of mitochondria, the function of mitochondria is finally enhanced, and the immune cells are adapted to the tumor microenvironment, and the tumor treatment ability is increased. In order to make T cells more amino acids can be used: Overexpression of amino acid transporters CD98, SNAT1, SNAT2, and/or ASCT2, etc., transport amino acids from the extracellular to intracellular, or can be converted to glutamine salt into the tricarboxylic acid cycle by GLS glutamine. It is then converted from POA to PEP by PCK enzyme. Embodiments relate to a cell modified to express one or more molecules at a level that is higher or lower than the level of the one or more expressed by a cell that has not been modified to expression the one or more molecules, wherein the one or more molecules are associated with metabolism of the modified cell. Embodiments relate to a modified cell engineered to express an antigen binding molecule, wherein expression and/or function of one or more molecules in the modified cell has been enhanced or reduced (including eliminated), where the one or more molecules are associated with the metabolism of the modified cell. In some embodiments, the modified cell comprises a disruption in an endogenous gene or addition of exogenous gene that is associated with a biosynthesis or transportation pathway of the one or more molecules. Embodiments relate to a pharmaceutical composition comprising the population of the cells. Embodiments relate to a method of causing or eliciting T cell response in a subject in need thereof and/or treating a tumor of the subject, the method comprising administering an effective amount of the composition of claim6to the subject. Embodiments relate to an isolated nucleic acid sequence encoding one or more molecules are associated with metabolism of the modified cell. Embodiments relate to a method or use of polynucleotide, the method comprising providing a viral particle (e.g., AAV, lentivirus or their variants) comprising a vector genome, the vector genome comprising the polynucleotide encoding the one more molecules and a polynucleotide encoding a binding molecule, the polynucleotide operably linked to an expression control element conferring transcription of the polynucleotides; and administering an amount of the viral particle to a subject such that the polynucleotide is expressed in the subject, where the one or more molecules are overexpressed in cancer cells, associated with recruitment of immune cells, and/or associated with autoimmunity. In embodiments, the AAV preparation may include AAV vector particles, empty capsids, and host cell impurities, thereby providing an AAV product substantially free of AAV empty capsids. In embodiments, the one more molecules comprise at least one of MCT1, MCT2, MCT3, LDHB, and MPC, a functional variant of the one or more molecules, or a functional fragment of the one or more molecules; and/or the metabolism comprises metabolism of lactic acid. In embodiments, the metabolism comprises the transportation of lactic acid of the modified cells, which is changed. In embodiments, the modified cells transport less or more lactic acid into the modified cells than that of corresponding wild-type cells. In embodiments, the modified cells overexpress MCT 3 and express less MCT1 and MCT2, and transport less lactic acid into the modified cells than that of corresponding wild-type cells. In embodiments, the modified cells overexpress MCT1, MCT2, LDHB, and MPC, express less MCT3, and transport more lactic acid into the modified cells than that of corresponding wild-type cells. In embodiments, the one more molecules comprise at least one of Frataxin, HBA, HBB, HBD, HBE, HBG, TOMM20, and TOMM22, a functional variant of the one or more molecules, or a functional fragment of the one or more molecules; and/or the metabolism comprises metabolism of lactic acid. In embodiments, the metabolism comprises the oxidative function of mitochondria of the modified cells, which is enhanced. In embodiments, the modified cells overexpress Frataxin, HBA, HBB, HBD, HBE, HBG, such as to enhances the mitochondrial oxygen storage capacity of the modified cells. In embodiments, the modified cells overexpress TOMM20 and TOMM22, such as to enhance functions of mitochondria of the modified cells. In embodiments, the one more molecules comprise at least one of CD98, SNAT1, SNAT2, ASCT2, a functional variant of the one or more molecules, or a functional fragment of the one or more molecules; and/or the metabolism comprises metabolism of lactic acid. In embodiments, the metabolism comprises the metabolism of amino acids of the modified cells, which is enhanced. In embodiments, the modified cells overexpress CD98, SNAT1, SNAT2, ASCT2, such as to enhances the transportation capability for the modified cells to transport the amino acids into the modified cells. Embodiments relate to a method of modifying a target genomic locus in a T cell to downregulate a gene of interest, the method comprising: introducing into the T cell a nuclease agent that makes a single or double-strand break within the target genomic locus; and introducing into the cell a nucleic acid insert such as to knock down or out the gene of interest; and selecting the cell comprising the nucleic acid insert integrated into the target genomic locus. In some embodiments, the nucleic acid insert is flanked by a 5′ homology arm and a 3′ homology arm, and the 3′ homology arm of the nucleic acid insert and the 5′ homology arm of the nucleic acid insert are homologous to corresponding genomic segments within the target genomic locus. In some embodiments, the nuclease agent is a zinc finger nuclease (ZFN), a Transcription Activator-Like Effector Nuclease (TALEN), or a meganuclease. In certain embodiments, the nuclease agent comprises a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-associated (Cas) protein and a guide RNA (gRNA). For example, the Cas protein is Cas9. In embodiments, expression of the polynucleotide is regulated or modulated by a synthetic Notch receptor comprising, from N-terminal to C-terminal and in covalent linkage: a) an extracellular domain comprising an antibody (e.g., a single-chain Fv (scFv) or a nanobody) that specifically binds to an antigen; b) a Notch regulatory region (NRR) and c) an intracellular domain comprising a transcriptional activator comprising a DNA binding domain. In embodiments, the Notch regulatory region comprises a Lin 12-Notch repeat, a heterodimerization domain comprising an S2 proteolytic cleavage site, and a transmembrane domain comprising an S3 proteolytic cleavage site. The intracellular domain is heterologous to the Notch regulatory region. In some embodiments, the transcriptional activator replaces a naturally-occurring intracellular notch domain, and binding of the antibody to the antigen induces cleavage at the S2 and S3 proteolytic cleavage sites, thereby releasing the intracellular domain. The release of the intracellular domain causes the transcriptional activator to induce expression of the polynucleotide encoding one or more target proteins in the modified cell. In embodiments, the modified cell comprises a polynucleotide encoding the synthetic Notch receptor and a polynucleotide encoding a transcriptional control element that is responsive to the transcriptional activator and operably linked to the polynucleotide encoding one or more target proteins (e.g., overexpression of molecules related to metabolism described herein). EXEMPLARY EMBODIMENTS The following are exemplary embodiments:1. A polynucleotide encoding a modified component of the TCR-CD3 complex.2. A vector comprising the polynucleotide of embodiment 1.2. A cell modified comprising the polynucleotide of embodiment 1.3. A modified cell engineered to express a modified component of the TCR-CD3 complex, wherein the modified cell includes an antigen binding molecule.4. A method or use of polynucleotide, the method comprisingproviding a viral particle (e.g., AAV, lentivirus or their variants) comprising a vector genome, the vector genome comprising the polynucleotide of embodiment 1; andadministering an amount of the viral particle to a subject such that the polynucleotide is expressed in the subject.5. The method of embodiment 4, wherein the AAV preparation may include AAV vector particles, empty capsids, and host cell impurities, thereby providing an AAV product substantially free of AAV empty capsids.6. A pharmaceutical composition comprising the population of the cells of any of embodiments 2 and 3.7. A method of causing or eliciting T cell response in a subject in need thereof and/or treating a tumor of the subject, the method comprising administering an effective amount of the composition of embodiment 6 to the subject.8. The polynucleotide of any proceeding embodiments 1-7, wherein the polynucleotide comprises at least one of the sequences listed in Table 2.9. The polynucleotide, vector, modified cell, and method of any of embodiments 1-8, wherein the TCR-CD3 complex comprises TCRα, TCRβ, CD3γ, ζ-chain, CD3ε, and CD3δ.10. The polynucleotide, vector, modified cell, and method of any of embodiments 1-8, wherein the TCR-CD3 complex comprises TCRγ, TCRδ, CD3γ, ζ-chain, CD3ε, and CD3δ.11. The polynucleotide, vector, modified cell, and method of any of embodiments 1-10, wherein the modified component of TCR-CD3 complex comprises components of TCR-CD3 complex linked to one or more co-stimulatory signaling domains.12. The polynucleotide, vector, modified cell, and method of embodiment 11, wherein the one or more co-stimulatory signaling domains comprise one or more functional signaling domains of one or more proteins selected from the group consisting of CD27, CD28, 4-1BB(CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM(LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, vFLIP K13, K13-opt, a NEMO mutant, a NEMO-fusion protein, IKKI-S176E-S180E, IKK2-S177E-S181E, RIP, IKKα, IKKβ, Tcl-I, MyD88-L265,ally NF-KB actuating protein or protein fragment. any inhibitor of an inhibiior of NF-kB, pathway, any gene editing sysiem capable of selectively activating NF-κB. 13. The polynucleotide, vector, modified cell, and method of embodiment 11, wherein the one or more co-stimulatory signaling domains comprise one or more functional signaling domains of one or more proteins selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D.14. The polynucleotide, vector, modified cell, and method of any of embodiments 1-13, wherein the modified component of TCR-CD3 complex comprises CD3γ, ζ-chain, CD3ε, and/or CD3δ.15. The polynucleotide, vector, modified cell, and method of any of embodiments 1-13, wherein the modified component of TCR-CD3 complex comprises CD3γ linked to one or more co-stimulatory signaling domains, and/or the modified component of TCR-CD3 complex comprises CD3ζ linked to one or more co-stimulatory signaling domains.16. The polynucleotide, vector, modified cell, and method of any of embodiments 1-13, wherein the modified component of TCR-CD3 complex comprises CD3ε linked to one or more co-stimulatory signaling domains.17. The polynucleotide, vector, modified cell, and method of any of embodiments 1-13, wherein the modified component of TCR-CD3 complex comprises CD3δ linked to one or more co-stimulatory signaling domains.18. The polynucleotide, vector, modified cell, and method of any of embodiments 14-17, wherein the CD3γ, ζ-chain, CD3ε, and/or CD3δ and the one or more co-stimulatory signaling domains are linked by a linker (e.g., G.S. linker).19. The polynucleotide, vector, modified cell, and method of any of embodiments 14-17, wherein the one or more co-stimulatory signaling domains comprise at least two co-stimulatory signaling domains.20. The polynucleotide, vector, modified cell, and method of embodiment 19, wherein the at least two co-stimulatory signaling domains are linked by a linker (e.g., G.S. linker).21. The modified cell of any proceeding embodiments 1-20, wherein the modified component of the TCR-CD3 complex is modified CD3 domain.22. The modified cell of any proceeding embodiments 1-21, wherein the modified TCR-CD3 complex is overexpressed by the modified cell, and/or the modified CD3 domain is overexpressed by the modified cell.23. The modified cell of any of the preceding embodiments, wherein the modified cell comprises the antigen binding molecule, the antigen binding molecule is chimeric antigen receptor (CAR), which comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain.24. The modified cell of embodiment 23, wherein the antigen-binding domain binds to a tumor antigen is selected from a group consisting of: TSHR, CD19, CD123, CD22, CD30, CD171, CS-1, CLL-1, CD33, EGFRvIII, GD2, GD3, BCMA, Tn Ag, PSMA, ROR1, FLT3, FAP, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, Mesothelin, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-beta, SSEA-4, CD20, Folate receptor alpha, ERBB2 (Her2/neu), MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, IGF-I receptor, CAIX, LMP2, gp100, bcr-abl, tyrosinase, EphA2, Fucosyl GM1, sLe, GM3, TGS5, HMWMAA, o-acetyl-GD2, Folate receptor beta, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, Polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, NY-ESO-1, LACE-1a, MAGE-A1, legumain, HPV E6, E7, MAGE A1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53, p53 mutant, prostein, survivin and telomerase, PCTA-1/Galectin 8, MelanA/MART1, Ras mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, Androgen receptor, Cyclin B1, MYCN, RhoC, TRP-2, CYP1B1, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxyl esterase, mut hsp70-2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1.25. The modified cell of any one of embodiments 23 and 24, wherein the intracellular signaling domain comprises a co-stimulatory signaling domain, or a primary signaling domain and a co-stimulatory signaling domain, wherein the co-stimulatory signaling domain comprises a functional signaling domain of a protein selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D26. The modified cell of any one of embodiments 1-22, wherein the modified cell comprises the antigen binding molecule, the antigen binding molecule is a modified TCR (TCR) or TCR (TIL).27. The modified cell of embodiment 26, wherein the TCR is derived from spontaneously occurring tumor-specific T cells in patients.28. The modified cell of embodiment 27, wherein the TCR binds to a tumor antigen.29. The modified cell of embodiment 28, wherein the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1.30. The modified cell of embodiment 28, wherein the TCR comprises TCRγ and TCRδ Chains or TCRα and TCRβ chains, or a combination thereof.31. The modified cell of any of the preceding embodiments, wherein the cell is an immune effector cell (e.g., a population of immune effector cells).32. The modified cell of embodiment 31, wherein the immune effector cell is a T cell or an NK Cell.33. The modified cell of embodiment 32, wherein the immune effector cell is a T cell.34. The modified cell of embodiment 32 wherein the T cell is a CD4+ T cell, a CD8+ T cell, or a combination thereof.35. The modified cell of any of the preceding embodiments, wherein the cell is a human cell.36. The modified cell of any of the preceding embodiments, wherein the enhanced expression and/or function of the modified component of TCR-CD3 complex is implemented by introducing a polynucleotide encoding the modified component of TCR-CD3 complex and/or the binding molecule, which is present in the modified cell in a recombinant DNA construct, in an mRNA, or in a viral vector.37. The modified cell of embodiment 36, wherein the polynucleotide is an mRNA, which is not integrated into the genome of the modified cell.38. The modified cell of embodiment 36, wherein the polynucleotide is associated with an oxygen-sensitive polypeptide domain.39. The modified cell of embodiment 38, wherein the oxygen-sensitive polypeptide domain comprises HIF VHL binding domain.40. The modified cell of embodiment 36, wherein the polynucleotide is regulated by a promoter comprising a binding site for a transcription modulator that modulates the expression and/or secretion of the therapeutic agent in the cell.41. The modified cell of embodiment 40, wherein the transcription modulator is or includes Hif1a, NFAT, FOXP3, and/or NFkB.42. A cell modified to express one or more molecules at a level that is higher or lower than the level of the one or more expressed by a cell that has not been modified to expression the one or more molecules, wherein the one or more molecules are associated with metabolism of the modified cell.43. A modified cell engineered to express an antigen binding molecule, wherein expression and/or function of one or more molecules in the modified cell has been enhanced or reduced (including eliminated), where the one or more molecules are associated with metabolism of the modified cell.44. The modified cell of any one of embodiments 42 and 43, wherein the modified cell comprises a disruption in an endogenous gene or an addition of exogenous gene that is associated with a biosynthesis or transportation pathway of the one or more molecules.45. A method or use of polynucleotide, the method comprisingproviding a viral particle (e.g., AAV, lentivirus or their variants) comprising a vector genome, the vector genome comprising the polynucleotide encoding the one more molecules and a polynucleotide encoding an antigen binding molecule, the polynucleotide operably linked to an expression control element conferring transcription of the polynucleotides; andadministering an amount of the viral particle to a subject such that the polynucleotide is expressed in the subject, where the one or more molecules are associated with metabolism of the modified cell.46. The method of embodiment 45, wherein the AAV preparation may include AAV vector particles, empty capsids and host cell impurities, thereby providing an AAV product substantially free of AAV empty capsids.47. A pharmaceutical composition comprising the population of the cells of any of embodiments 1-3.48. A method of causing or eliciting T cell response in a subject in need thereof and/or treating a tumor of the subject, the method comprising administering an effective amount of the composition of embodiment 47 to the subject or an isolated nucleic acid sequence encoding one or more molecules associated with metabolism of the modified cell.49. The isolated nucleic acid sequence, modified cell, method, or pharmaceutical composition of any one of embodiments 42-48, wherein the one more molecules comprise at least one of MCT1, MCT2, MCT3, LDHB, and MPC, a functional variant of the one or more molecules, or a functional fragment of the one or more molecules; and/or the metabolism comprises metabolism of lactic acid.50. The isolated nucleic acid sequence, modified cell, method, or pharmaceutical composition of any one of embodiments 42-48, wherein the metabolism comprises transportation of lactic acid of the modified cells, which is changed.51. The modified cell of any proceeding embodiments 42-50, wherein the modified cells transport less or more lactic acid into the modified cells than that of corresponding wild-type cells.52. The modified cell of any proceeding embodiments 42-50, wherein the modified cells overexpress MCT 3 and express less MCT1 and MCT2, and transport less lactic acid into the modified cells than that of corresponding wild-type cells.53. The modified cell of any proceeding embodiments 42-50, wherein the modified cells overexpress MCT1, MCT2, LDHB, and MPC, express less MCT3, and transport more lactic acid into the modified cells than that of corresponding wild-type cells.54. The isolated nucleic acid sequence, modified cell, method, or pharmaceutical composition of any one of embodiments 42-53, wherein the one more molecules comprise at least one of Frataxin, HBA, HBB, HBD, HBE, HBG, TOMM20, and TOMM22, a functional variant of the one or more molecules, or a functional fragment of the one or more molecules; and/or the metabolism comprises metabolism of lactic acid.55. The isolated nucleic acid sequence, modified cell, method, or pharmaceutical composition of any one of embodiments 42-54, wherein the metabolism comprises the oxidative function of mitochondria of the modified cells, which is enhanced.56. The modified cell of any proceeding embodiments 42-55, wherein the modified cells overexpress Frataxin, HBA, HBB, HBD, HBE, HBG such as to enhances the mitochondrial oxygen storage capacity of the modified cells.57. The modified cell of any proceeding embodiments 42-55, wherein the modified cells overexpress TOMM20 and TOMM22 such as to enhance functions of mitochondria of the modified cells.58. The isolated nucleic acid sequence, modified cell, method, or pharmaceutical composition of any one of embodiments 42-47, wherein the one more molecules comprise at least one of CD98, SNAT1, SNAT2, ASCT2, a functional variant of the one or more molecules, or a functional fragment of the one or more molecules; and/or the metabolism comprises metabolism of amino acids.59. The isolated nucleic acid sequence, modified cell, method, or pharmaceutical composition of any one of embodiments 42-48, wherein the metabolism comprises metabolism of lactic acid and/or amino acids of the modified cells, which is enhanced.60. The modified cell of any proceeding embodiments 42-58, wherein the modified cells overexpress CD98, SNAT1, SNAT2, ASCT2 such as to enhances the transportation capability for the modified cells to transport the amino acids into the modified cells.61. The isolated nucleic acid sequence, modified cell, method, or pharmaceutical composition of embodiment 42-60, wherein one or more sequences listed in Table 7, 8, and 9 are overexpressed or downregulated in the modified cell.62. The modified cell of any of the preceding embodiments 42-61, wherein the modified cell comprises an antigen binding molecule.63. The modified cell of embodiment 62, the antigen binding molecule is chimeric antigen receptor (CAR), which comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain.64. The modified cell of embodiment 63, wherein the antigen-binding domain binds to a tumor antigen is selected from a group consisting of: TSHR, CD19, CD123, CD22, CD30, CD171, CS-1, CLL-1, CD33, EGFRvIII, GD2, GD3, BCMA, Tn Ag, PSMA, ROR1, FLT3, FAP, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, Mesothelin, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-beta, SSEA-4, CD20, Folate receptor alpha, ERBB2 (Her2/neu), MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, IGF-I receptor, CAIX, LMP2, gp100, bcr-abl, tyrosinase, EphA2, Fucosyl GM1, sLe, GM3, TGS5, HMWMAA, o-acetyl-GD2, Folate receptor beta, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, Polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, NY-ESO-1, LACE-1a, MAGE-A1, legumain, HPV E6, E7, MAGE A1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53, p53 mutant, prostein, survivin and telomerase, PCTA-1/Galectin 8, MelanA/MART1, Ras mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, Androgen receptor, Cyclin B1, MYCN, RhoC, TRP-2, CYP1B1, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxyl esterase, mut hsp70-2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1.65. The modified cell of any one of embodiments 63 and 64, wherein the intracellular signaling domain comprises a co-stimulatory signaling domain, or a primary signaling domain and a co-stimulatory signaling domain, wherein the co-stimulatory signaling domain comprises a functional signaling domain of a protein selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D66. The modified cell of embodiment 62, wherein the modified cell comprises the antigen binding molecule, the antigen binding molecule is a modified TCR (TCR) or TCR (TIL).67. The modified cell of embodiment 65, wherein the TCR is derived from spontaneously occurring tumor-specific T cells in patients.68. The modified cell of embodiment 67, wherein the TCR binds to a tumor antigen.69. The modified cell of embodiment 68, wherein the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1.70. The modified cell of embodiment 68, wherein the TCR comprises TCRγ and TCRδ Chains or TCRα and TCRβ chains, or a combination thereof.71. The modified cell of embodiment 70, wherein the cell is an immune effector cell (e.g., a population of immune effector cells), and the immune effector cell is a DC, macrophage, T cell or an NK cell.72. The modified cell of embodiment 71, wherein the immune effector cell is a T cell.73. modified cell of embodiment 71 wherein the T cell is a CD4+ T cell, a CD8+ T cell, or a combination thereof.74. The modified cell of any of the preceding embodiments 42-73, wherein the cell is a human cell.75. The modified cell of any of the preceding embodiments 42-74, wherein the enhanced expression and/or function of the one or more molecules is implemented by introducing a nucleic acid sequence encoding the one or more molecules and/or the binding molecule, which is present in the modified cell in a recombinant DNA construct, in an mRNA, or in a viral vector.76. The modified cell of embodiment 75, wherein the nucleic acid sequence is an mRNA, which is not integrated into the genome of the modified cell.77. The modified cell of embodiment 75, wherein the nucleic acid sequence is associated with an oxygen-sensitive polypeptide domain.78. The modified cell of embodiment 77, wherein the oxygen-sensitive polypeptide domain comprises HIF VHL binding domain.79. The modified cell of embodiment 75, wherein the nucleic acid sequence is regulated by a promoter comprising a binding site for a transcription modulator that modulates the expression and/or secretion of the therapeutic agent in the cell.80. The modified cell of embodiment 79, wherein the transcription modulator is or includes Hif1a, NFAT, FOXP3, and/or NFkB. EXAMPLES Lentiviral vectors that encode individual CAR molecules were generated and transfected with T cells, which are elaborated below. Techniques related to cell cultures, construction of cytotoxic T lymphocyte essay may be found in “Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains,” PNAS, Mar. 3, 2009, vol. 106 no. 9, 3360-3365, and “Chimeric Receptors Containing CD137 Signal Transduction Domains Mediate Enhanced Survival of T Cells and Increased Antileukemic Efficacy In Vivo,” Molecular Therapy, August 2009, vol. 17 no. 8, 1453-1464, which are incorporated herein by reference in their entirety. On day 0, peripheral blood was extracted from healthy volunteers. CD3+T cells were sorted by pan T Kit, and 100 ul TransAct per 1×107T cells were added. On day 1, 1×106T cells were transfected with vector 8301. 1×106T cells were transfected with vector 8307, and 2×106T cells were non-transduced T cells (N.T.). On day 2, culture media were changed. The lentivirus and TransAct were removed, and the cells were resuspended in fresh media. On day 7, the flow detection of the TCR ratio was performed.FIG.6shows flow cytometry results of the expression of the various vectors in T cells. Since both vectors encode Vβ13.1 (a variant of the TCR β chain), the anti-TCR Vβ13.1 was used. The expression ratio of Vβ13.1 with vector 8301 is 83.58%. The expression ratio of Vβ13.1 with vector 8307 is 51.38%. The experiment was carried out according to Tables 3 and 4. The samples were co-cultured for 24h, and FCM staining of Vβ 13.1+multicolor was taken, and the amplification was detected by FCM staining with a cell trace marker at 120 h. Sequences can be found in Table 2 below. More information on sequences, compositions, and related clinical trials can be found in WO2020106843 and WO2020146743, which are hereby incorporated by reference in their entirety. TABLE 2SEQDescriptionID NO:CD3 ZetaMKWKALFTAAILQAQLPITEAQSFGLLDPKLCYLLDGILFIYGVILT1ChainALFLRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPQRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRCD3 DeltaMEHSTFLSGLVLATLLSQVSPFKIPIEELEDRVFVNCNTSITWVE2ChainGTVGTLLSDITRLDLGKRILDPRGIYRCNGTDIYKDKESTVQVHYRMCQSCVELDPATVAGIIVTDVIATLLLALGVFCFAGHETGRLSGAADTQALLRNDQVYQPLRDRDDAQYSHLGGNWARNKCD3 EpsilonMQSGTHWRVLGLCLLSVGVWGQDGNEEMGGITQTPYKVSISG3ChainTTVILTCPQYPGSEILWQHNDKNIGGDEDDKNIGSDEDHLSLKEFSELEQSGYYVCYPRGSKPEDANFYLYLRARVCENCMEMDVMSVATIVIVDICITGGLLLLVYYWSKNRKAKAKPVTRGAGAGGRQRGQNKERPPPVPNPDYEPIRKGQRDLYSGLNQRRICD3 gammaMEQGKGLAVLILAIILLQGTLAQSIKGNHLVKVYDYQEDGSVLLT4ChainCDAEAKNITWFKDGKMIGFLTEDKKKWNLGSNAKDPRGMYQCKGSQNKSKPLQVYYRMCQNCIELNAATISGFLFAEIVSIFVLAVGVYFIAGQDGVRQSRASDKQTLLPNDQLYQPLKDREDDQYSHLQGNQLRRNGS-LinkerSGGGGS5NY-ESO-1METLLGLLILWLQLQWVSSKQEVTQIPAALSVPEGENLVLNCSF6VαTDSAIYNLQWFRQDPGKGLTSLLLIQSSQREQTSGRLNASLDKSSGRSTLYIAASQPGDSATYLCAVRPTSGGSYIPTFGRGTSLIVHPYIQNPDPAVYQLRDSKSSDKSVCLFTDFDSQTNVSQSKDSDVYITDKTVLDMRSMDFKSNSAVAWSNKSDFACANAFNNSIIPEDTFFPSPESSCDVKLVEKSFETDTNLNFQNLSVIGFRILLLKVAGFNLLMTLRLWSSP2AGSGATNFSLLKQAGDVEENPGP7NY-ESO-1MSIGLLCCAALSLLWAGPVNAGVTQTPKFQVLKTGQSMTLQC8VβAQDMNHEYMSWYRQDPGMGLRLIHYSVGAGITDQGEVPNGYNVSRSTTEDFPLRLLSAAPSQTSVYFCASSYVGNTGELFFGEGSRLTVLEDLKNVFPPEVAVFEPSEAEISHTQKATLVCLATGFYPDHVELSWWVNGKEVHSGVSTDPQPLKEQPALNDSRYSLSSRLRVSATFWQNPRNHFRCQVQFYGLSENDEWTQDRAKPVTQIVSAEAWGRADCGFTSESYQQGVLSATILYEILLGKATLYAVLVSALVLMAMVKRKDSRGT2AGSGEGRGSLLTCGDVEENPGP9ζ-chain (full-MKWKALFTAAILQAQLPITEAQSFGLLDPKLCYLLDGILFIYGVIL10length)TALFLRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRCD137KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL11(intracellular)ζ-chain-2A-MKWKALFTAAILQAQLPITEAQSFGLLDPKLCYLLDGILFIYGVIL18CD137TALFLRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDK(intracellular)RRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRGSGEGRGSLLTCGDVEENPGPKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELNY-ESO- 1METLLGLLILWLQLQWVSSKQEVTQIPAALSVPEGENLVLNCSF19TCRα + β-TDSAIYNLQWFRQDPGKGLTSLLLIQSSQREQTSGRLNASLDK2A-ζ-chainSSGRSTLYIAASQPGDSATYLCAVRPTSGGSYIPTFGRGTSLIV(full-length)-HPYIQNPDPAVYQLRDSKSSDKSVCLFTDFDSQTNVSQSKDSCD137DVYITDKTVLDMRSMDFKSNSAVAWSNKSDFACANAFNNSIIP(intracellular)EDTFFPSPESSCDVKLVEKSFETDTNLNFQNLSVIGFRILLLKVAGFNLLMTLRLWSSGSGATNFSLLKQAGDVEENPGPMSIGLLCCAALSLLWAGPVNAGVTQTPKFQVLKTGQSMTLQCAQDMNHEYMSWYRQDPGMGLRLIHYSVGAGITDQGEVPNGYNVSRSTTEDFPLRLLSAAPSQTSVYFCASSYVGNTGELFFGEGSRLTVLEDLKNVFPPEVAVFEPSEAEISHTQKATLVCLATGFYPDHVELSWWVNGKEVHSGVSTDPQPLKEQPALNDSRYSLSSRLRVSATFWQNPRNHFRCQVQFYGLSENDEWTQDRAKPVTQIVSAEAWGRADCGFTSESYQQGVLSATILYEILLGKATLYAVLVSALVLMAMVKRKDSRGGSGEGRGSLLTCGDVEENPGPMKWKALFTAAILQAQLPITEAQSFGLLDPKLCYLLDGILFIYGVILTALFLRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL TABLE 3NameConstruction8301NY-ESO-1 TCRα + β8307NY-ESO-1 TCRα + β-2A-ζ-chain (full-length)-CD137(intra) (See Embodiment 102 in FIG. 1) TABLE 4293T8505CK19E:TsystemNT−−−40 w:13.3 wNo IL2 texmacs media,+−−400 ul resuspended−+−−−+8301−−−+−−−+−−−+8307−−−+−−−+−−−+ FIGS.7and8shows the expression of HLA-A2 and NY-ESO-1 in substrate cells and expression of ζ-chain in 8301 and 8307. The TCRT of NY-ESO-1, which recognizes HLA-A2 presentation, was used in the experiment as the experimental control material (8301), so the flow expression of HLA-A2 in the substrate cells was detected, and K19 appeared to be HLA-A2 negative, 293T was weak HLA-A2 positive, and 8505C, and SAOS-2 were HLA-A2 positive. RT-PCR was used to detect the mRNA expression of NY-ESO-1 in the four substrate cells, and the results showed that 8505C and SAOS-2 were positive for NY-ESO-1. The following Table 6 shows the primer information of RT-PCR.FIG.8shows Western blot results confirming the structure of modified TCR. Based on the Western blot results, 8307 cells have a size of about 23.3 kDa stripe, which is ζ-chain after CD137 restructuring. TABLE 5CtnumberSample NameACTINNY-ESO-1-1NY-ESO-1-21K56215.4127.5721.602293T15.4225.5730.5138505C15.5922.5017.884Saos-215.0024.8720.02 TABLE 6PrimerSequence (5′ to 3′)SEQ ID NONY-ESO-1-RTF1CGGCAACATACTGACTATCCG12NY-ESO-1-RTR1CTGGAGACAGGAGCTGATGGA13NY-ESO-1-RTF2TGCAGACCACCGCCAACT14NY-ESO-1-RTR2TCCACATCAACAGGGAAAGCT15β-actin-RTFCGCCCAGCACGATGAAA16β-actin-RTRCCGCCGATCCACACAGAG17 TABLE 7Molecules related to lactic acid metabolismnameFeaturesMethod of operationMCT1 is alsoNormal expression on T cells, mainlyKnockdown/knockoutcalledregulating the inward transport of lactic(or overexpression, combinedSLC16A1acid,with LDHB expression)MCT2Lactic acid transportKnockdown/knockout(or overexpression, combinedwith LDHB expression)MCT4 is alsoMCT4 is the transport of lactic acid out ofOverexpression or inducedcalledcells, contrary to MCT1 function.expressionSLC16A3.However, it may also help T cells from(or knockdown/knockout,lactic acid inhibition in a high lactic acidcombined with LDHBenvironment.expression)BSGCD147 is an accessory protein of MCTKnockdown/knockout(CD147)family proteins, regulating the correct(or overexpression or inductionlocalization of MCT family proteins on theof expression in combinationcell membranewith LDHB expression)LDHBConverting lactic acid to pyruvic acid,Overexpression or inducedexpressionMPC1Pyruvate can enter the mitochondriaOverexpression or inducedthrough the pyruvate transporter (MPC)expressionon the mitochondrial membrane.MPC2Pyruvate can enter the mitochondriaOverexpression or inducedthrough the pyruvate transporter (MPC)expressionon the mitochondrial membrane.PDK1/2/3Pyruvate dehydrogenase kinase (PDK)Knockout/knockdowninhibits PDH activity,Thereby inhibiting pyruvate metabolism TABLE 8Molecules related to mitochondrial functionPGC1aEnhance mitochondrial volume andOverexpressionenhance oxidative phosphorylationor inducedexpressionOPA1Promote mitochondrial fusion, enhance″mitochondrial function, and improve electrontransport chain efficiencyHBE1 (HemoglobinThe epsilon chain is a beta-type chain of″subunit epsilon)early mammalian embryonic hemoglobin,mitochondrial proteinHBZ (HemoglobinThe zeta chain is an alpha-type chain of″subunit zeta)mammalian embryonic hemoglobin.mitochondrial proteinHBD (HemoglobinInvolved in oxygen transport from the lung″subunit delta)to the various peripheral tissues.mitochondrial proteinHBA1 (HemoglobinInvolved in oxygen transport from the lung″subunit alpha)to the various peripheral tissues.mitochondrial proteinHBB (HemoglobinInvolved in oxygen transport from the lung″subunit beta)to the various peripheral tissues.mitochondrial proteinHBG1 (HemoglobinGamma chains make up the fetal″subunit gamma-1)hemoglobin F, in combination with alphachains. mitochondrial proteinHBG2 (HemoglobinGamma chains make up the fetal″subunit gamma-2)hemoglobin F, in combination with alphachains. mitochondrial proteinLYRM4 (LYR motif-Required for nuclear and mitochondrial iron-″containing protein 4)sulfur protein biosynthesis. MitochondrialproteinFXN (Frataxin,Promotes the biosynthesis of heme and″mitochondrial)assembly and repair of iron-sulfur clusters.Mitochondrial proteinTOMM20(MitochondrialThe central component of the receptor″import receptor subunitcomplex responsible for the recognition andTOM20 homolog)translocation of cytosolically synthesizedmitochondrial preproteins. Mitochondrialassembling proteinsTOMM22(MitochondrialThe central receptor component of the″import receptor subunittranslocase of the outer membrane ofTOM22 homolog)mitochondria (TOM complex) responsiblefor the recognition and translocation ofcytosolically synthesized mitochondrialpreproteins. Mitochondrial assemblingproteins<HSPE1 (10 kDa heatCo-chaperonin implicated in mitochondrial″shock protein,protein import and macromolecularmitochondrial)assembly. Together with Hsp60, facilitatesthe correct folding of importedproteins. Mitochondrial assembling proteins<HSPD1 (60 kDa heatChaperonin implicated in mitochondrial″shock protein,protein import and macromolecularmitochondrial)assembly. Together with Hsp10, it facilitatesthe correct folding of imported proteins.Mitochondrial assembling proteins<HSPA9(Stress-70 protein,Chaperone protein which plays an important″mitochondrial)role in the mitochondrial iron-sulfur cluster(ISC) biogenesis. Interacts with andstabilizes ISC cluster assembly proteins.Mitochondrial assembling proteins< TABLE 9Molecules related to amino acid metabolismMethod ofnameFeaturesoperationAsct2Amino acid transporterOverexpressionor inducedexpressionCD98Amino acid transporterOverexpressionor inducedexpressionSNAT1Amino acid transporterOverexpressionor inducedexpressionSNAT2Amino acid transporterOverexpressionor inducedexpressionPCK1Conversion of OAAOverexpression(oxaloacetate) to PEPor induced(phosphoenolpyruvate)expressionAmino acid metabolismPCK2Conversion of OAAOverexpression(oxaloacetate) to PEPor induced(phosphoenolpyruvate)expressionAmino acid metabolismGLSGlutaminase (GLS)Overexpression(GlutaminaseConverts glutamine to glutamateor inducedkidney isoform,to support the tricarboxylic acidexpressionmitochondrial)cycle and redox and epigeneticreactions.Glutamic enzymeGLS2Plays an important role in theOverexpression(Glutaminaseregulation of glutamineor inducedliver isoform,catabolism. Promotesexpressionmitochondrial)mitochondrial respiration andincreases ATP generation in cellsby catalyzing the synthesis ofglutamate and alpha-ketoglutarate. Increases cellularanti-oxidant function via NADHand glutathione production. Mayplay a role in preventing tumorproliferation.Glutamic enzyme The present disclosure is further described by reference to the following exemplary embodiments and examples. These exemplary embodiments and examples are provided for purposes of illustration only and are not intended to be limiting unless otherwise specified. Thus, the present disclosure should in no way be construed as being limited to the following exemplary embodiments and examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.
116,569
11858978
DETAILED DESCRIPTION OF A PREFERRED EMBODIMENT The following tables show the peptides according to the present invention, their respective SEQ ID NOs, and the prospective source (underlying) genes for these peptides. In Table 1, peptides with SEQ ID NO: 1 to SEQ ID NO: 9 bind to HLA-A*02, peptides with SEQ ID NO: 10 to SEQ ID NO: 19 bind to HLA-A*24, peptides with SEQ ID NO: 20 to SEQ ID NO: 30 bind to HLA-A*03, peptide with SEQ ID NO: 31 binds to HLA-A*01, peptides with SEQ ID NO: 32 to SEQ ID NO: 41 bind to HLA-B*07, peptides with SEQ ID NO: 42 to SEQ ID NO: 51 bind to HLA-B*08, peptides with SEQ ID NO: 52 to SEQ ID NO: 59 bind to HLA-B*44. In Table 2, peptides with SEQ ID NO: 60 to SEQ ID NO: 75 bind to HLA-A*02, peptides with SEQ ID NO: 76 to SEQ ID NO: 82 bind to HLA-A*24, peptides with SEQ ID NO: 83 to SEQ ID NO: 111 bind to HLA-A*03, peptides with SEQ ID NO: 112 to SEQ ID NO: 116 bind to HLA-A*01, peptides with SEQ ID NO: 117 to SEQ ID NO: 149 bind to HLA-B*07, peptides with SEQ ID NO: 150 to SEQ ID NO: 172 bind to HLA-B*08, peptides with SEQ ID NO: 173 to SEQ ID NO: 215 bind to HLA-B*44. In Table 3, peptides with SEQ ID NO: 216 to SEQ ID NO: 245 bind to HLA-A*02, peptides with SEQ ID NO: 246 to SEQ ID NO: 255 bind to HLA-A*24, peptides with SEQ ID NO: 256 to SEQ ID NO: 287 bind to HLA-A*03, peptides with SEQ ID NO: 288 to SEQ ID NO: 292 bind to HLA-A*01, peptides with SEQ ID NO: 293 to SEQ ID NO: 392 bind to HLA-B*07, peptides with SEQ ID NO: 393 to SEQ ID NO: 395 bind to HLA-B*08, peptides with SEQ ID NO: 396 to SEQ ID NO: 438 bind to HLA-B*44. In Table 4, peptides with SEQ ID NO: 439 to SEQ ID NO: 551 bind to several HLA class I alleles, peptide with SEQ ID NO: 773 binds to HLA-A*02, peptide with SEQ ID NO: 774 binds to HLA-A*24. In Table 5, peptides with SEQ ID NO: 552 to SEQ ID NO: 772 bind to several HLA class II alleles. TABLE 1Peptides according to the present invention.Seq IDHLANoSequenceGeneUniprot Accessionallotype1MIPTFTALLLILRB4Q8NHJ6A*022TLLKALLEIIDO1P14902A*023ALIYNLVGIATP7A, ATP7B,P35670A*02CTAGE14ALFKAWALIRF4Q15306A*025RLLDFINVLOVGP1Q12889A*026SLGKHTVALOVGP1Q12889A*027ALQAFEFRPCDHB5, PCDHB15,Q9Y5E7A*02VPCDHB11, PCDHB10,PCDHB9, PCDHB8,PCDHB7, PCDHB4,PCDHB3, PCDHB2,PCDHB168YLVTKVVAVPCDHGA12, PCDHB5,O60330, Q96TA0,A*02PCDHB1, PCDHB18,Q9NRJ7, Q9UN66,PCDHGB7, PCDHGB6,Q9UN67, Q9UN71,PCDHGB5, PCDHGB3,Q9Y5E1, Q9Y5E2,PCDHGB2, PCDHGB1,Q9Y5E3, Q9Y5E4,PCDHGA11,Q9Y5E5, Q9Y5E6,PCDHGA10, PCDHGA9,Q9Y5E7, Q9Y5E8,PCDHGA7, PCDHGA6,Q9Y5E9, Q9Y5F0,PCDHGA5, PCDHGA4,Q9Y5F1, Q9Y5F2,PCDHGA3, PCDHGA2,Q9Y5F3, Q9Y5F8,PCDHGA1, PCDHGB8P,Q9Y5F9, Q9Y5G0,PCDHB15, PCDHB14,Q9Y5G1, Q9Y5G2,PCDHB13, PCDHB12,Q9Y5G3, Q9Y5G4,PCDHB11, PCDHB10,Q9Y5G5, Q9Y5G6,PCDHB9, PCDHB8,Q9Y5G7, Q9Y5G8,PCDHB7, PCDHB6,Q9Y5G9, Q9Y5H0,PCDHB4, PCDHB3,Q9Y5H1, Q9Y5H2,PCDHB2, PCDHB16,Q9Y5H3, Q9Y5H4PCDHGB4, PCDHGA89VLLAGFKPPRNF17Q9BXT8A*02L10RYSDSVGRCAPN13Q6MZZ7A*24VSF11SYSDLHYGCAPN13Q6MZZ7A*24F12KYEKIFEMLCT45A3, CT45A4,Q5DJT8A*24CT45A5, CT45A6,CT45A1, CT45A213VYTFLSSTLESR1P03372A*2414FYFPTPTVLFOLR1P15328A*2415VYHDDKQPGXYLT2A0PJZ3A*24TF16IYSPQFSRLMYO3BQ8WXR4A*2417RFTTMLSTFOVGP1Q12889A*2418KYPVHIYRLRAD54BQ9Y620A*2419KYVKVFHQZNF90, ZNF93, ZNF486Q96H40A*24F20RMASPVNVC2orf88Q9BSFOA*03K21AVRKPIVLKCDCA5Q96FF9A*0322SLKERNPLKCDH3P22223A*0323GMMKGGIRESR1P03372A*03K24SMYYPLQLGXYLT2A0PJZ3A*03K25GTSPPSVEMUC16Q8WXI7A*03K26RISEYLLEKMYO3BQ8WXR4A*0327VLYGPAGLNLRP2Q9NX02A*03GK28KTYETNLEINLRP7Q8WX94A*03KK29QQFLTALFYNLRP7, NLRP2Q8WX94A*0330ALEVAHRLKZBTB12Q9Y330A*0331LLDEGAMLLNLRP7Q8WX94A*01Y32SPNKGTLSBCAMP50895B*07V33SPTFHLTLBCAMP50895B*0734LPRGPLASLCDH3P22223B*07L35FPDNQRPAETV4P43268B*07L36APAAWLRSMMP11P24347B*07/B*55A37RPLFQKSSMUC16Q8WXI7B*07M38SPHPVTALLMUC16Q8WXI7B*07TL39RPAPFEVVNXNL2Q5VZ03B*07F40KPGTSYRVSPON1Q9HCB6B*07TL41RVRSRISNLTCEA1P2, TCEA1,TCEA2, TCEA3Q15560B*0742TLKVTSALBCAMP50895B*0843ALKARTVTFCCR2, CCR5P51681B*0844LNKQKVTFCTAGE4, CTAGE5Q15320B*0845VGREKKLACTAGE4, CTAGE5O15320B*08L46DMKKAKEQFUNDC2P2,Q9BWH2B*08LFUNDC2P3, FUNDC247MPNLRSVDLRRTM1Q86UE6B*08L48DVKKKIKEVMFN1Q8IWA4B*0849LPRLKAFMIST6GALNAC5Q9BVH7B*0850DMKYKNRVTCEA1P2, TCEA1,Q15560B*08TCEA251SLRLKNVQLVTCN1Q7Z7D3B*0852AEFLLRIFLCAPN13Q6MZZ7B*4453MEHPGKLLESR1P03372B*44F54AEITITTQTGMUC16Q8WXI7B*44Y55HETETRTTMUC16Q8WXI7B*44W56SEPDTTASMUC16Q8WXI7B*44W57QESDLRLFLNLRP7, NLRP2Q9NX02B*4458GEMEQKQLPNOCQ13519B*4459SENVTMKVVTCN1Q7Z7D3B*44V TABLE 2Additional peptides according to the present invention.Seq IDHLANoSequenceGeneUniprot Accessionallotype60GLLSLTSTLBCAMP50895A*02YL61YMVHIQVTLCD70P32970A*0262KVLGVNVMCRABP2P29373A*02L63MMEEMIFNEYA2O00167A*02L64FLDPDRHFLFAM83HQ6ZRV2A*0265TMFLRETSLGUCY1A2P33402A*0266GLLQELSSIHTR3AP46098A*0267SLLLPSIFLHTR3AP46098A*0268KLFDTQQFLIRF4Q15306A*0269TTYEGSITVMUC16Q8WXI7A*0270VLQGLLRSLMUC16Q8WXI7A*0271YLEDTDRNNFE2L3Q9Y4A8A*02L72YLTDLQVSLNFE2L3Q9Y4A8A*0273FLIEELLFAOVGP1Q12889A*0274SQSPSVSQPRAMEP78395A*02L75KVVSVLYNVTCN1Q7Z7D3A*02V76KYVAELSLLCCNA1P78396A*2477RYGPVFTVCYP2W1Q8TAV3A*2478SFAPRSAVHOXD9P28356A*24F79SYNEHWNYLTBRP36941A*24L80TAYMVSVASDK2Q58EX2A*24AF81VYNHTTRPSPINT1O43278A*24L82SYFRGFTLISPON1Q9HCB6A*2483GTYAHTVNALPI, ALPP, ALPPL2P05187, P09923A*03/A*31R84KLQPAQTAALPP, ALPPL2P05187A*03AK85VLLGSLFSRBCL2L1Q07817A*03K86VVLLGSLFSBCL2L1Q07817A*03/A*31/RKA*6687AVAPPTPACBX2Q14781A*03/A*11SK88VVHAVFALKCCR5P51681A*0389RVAELLLLHCDKN2A, CDKN2BP42771, P42772A*0390KVAGERYVETV1, ETV4, ETV5P41161, P43268,A*03YKP5054991RSLRYYYEETV1, ETV4, ETV5P43268A*03K92SVFPIENIYEYA2O00167A*0393KILEEHTNKFSBP, RAD54BO95073A*0394ATFERVLLRGUCY1A2P33402A*03/A*1195QSMYYPLQGXYLT2A0PJZ3A*03LK96TAFGGFLKLAMA1P25391A*03Y97TMLDVEGLLAMA1P25391A*03FY98LLQPPPLLAMMP11P24347A*03R99KVVDRWNEMRPL51Q4U2R6A*03K100RLFTSPIMTMUC16Q8WXI7A*03K101RVFTSSIKTMUC16Q8WXI7A*03K102SVLTSSLVKMUC16Q8WXI7A*03103TSRSVDEAMUC16Q8WXI7A*03Y104VLADSVTTKMUC16Q8WXI7A*03105RLFSWLVNMYO1BQ8WXR4A*03R106AAFVPLLLKNCAPD2Q15021A*03/A*11107RLQEWKALPDCL2Q8N4E4A*03K108VLYPVPLESPRAMEP78395A*03Y109KTFTIKRFLRPL39LQ96EH5A*03AK110SAAPPSYFSPON1Q9HCB6A*03/A*11/RA*66111TLPQFRELWNT7AO00755A*03GY112TVTGAEQIQCAPN13Q6MZZ7A*01Y113QLDSNRLTLRRTM1Q86UE6A*01Y114VMEQSAGILYPD1Q8N2G4A*01MY115FVDNQYWRMMP12P39900A*01Y116VLLDEGAMNLRP7Q8WX94A*01LLY117APRLLLLAVBCAMP50895B*07L118SPASRSISLCD70P32970B*07119APLPRPGACTAG2O75638B*07VL120RPAMNYDKETV1, ETV4, ETV5,P43268B*07LSPDEF121VPNQSSESEYA2O00167B*07/B*35L122YPGFPQSQEYA2O00167B*07/B*35Y123KPSESIYSAFAM111BQ65J93B*07L124LPSDSHFKIFAM111BQ65J93B*07TF125VPVYILLDEFAM83HQ6ZRV2B*07/B*35M126KPGPEDKLFOLR1, FOLR2P15328B*07127APRAGSQVFUNDC2Q9BWH2B*07V128YPRTITPGMKLK14Q9P0G3B*07129APRPASSLMMP11P24347B*07130FPRLVGPDMMP11P24347B*07F131APTEDLKALMSLNQ13421B*07132IPGPAQSTIMUC16Q8WXI7B*07133MPNLPSTTMUC16Q8WXI7B*07SL134RPIVPGPLLMUC16Q8WXI7B*07135RVRSTISSLMUC16Q8WXI7B*07136SPFSAEEAMUC16Q8WXI7B*07NSL137SPGATSRGMUC16Q8WXI7B*07TL138SPMATTSTLMUC16Q8WXI7B*07139SPQSMSNTMUC16Q8WXI7B*07L140SPRTEASSMUC16Q8WXI7B*07AVL141SPMTSLLTSMUC16Q8WXI7B*07GL142TPGLRETSIMUC16Q8WXI7B*07143SPAMTSTSMUC16Q8WXI7B*07/B*35F144SPSPVSSTLMUC16Q8WXI7B*07/B*35145SPSSPMSTMUC16Q8WXI7B*07/B*35F146IPRPEVQALPLEKHG4Q58EX7B*07147APRWFPQPVTCN1Q7Z7D3B*07TVV148KPYGGSGPZNF217O75362B*07L149GPREALSRZSCAN30, ZNF263,O14978, O43309,LZNF500, ZKSCAN4,O60304, P17029,ZNF323, ZKSCAN1,P49910, Q16670,ZNF165, ZNF187,Q86W11, Q8NF99,ZKSCAN3, ZNF397,Q969J2, Q96LW9,ZSCAN12Q9BRR0B*07150MAAVKQALBCL2L1Q07817B*08151HLLLKVLAFCCNA1P78396B*08152MGSARVAECDKN2A,CDKN2BP42771 B*08L153NAMLRKVACRABP1P29762B*08V154MLRKIAVAACRABP2P29373B*08155NKKMMKRLDPPA2Q7Z7J5B*08M156HVKEKFLLFAM83HQ6ZRV2B*08157EAMKRLSYILAMC2Q13753B*08158LPKLAGLLLINC00176Q6ZNR8B*08/B*07159VLKHKLDELMSLNQ13421B*08160YPKARLAFMSLNQ13421B*08161ALKTTTTALDNAJC22,MUC16Q8WXI7 B*08162QAKTHSTLMUC16Q8WXI7B*08163QGLLRPVFMUC16Q8WXI7B*08164SIKTKSAEMMUC16Q8WXI7B*08165SPRFKTGLMUC16Q8WXI7B*08166TPKLRETSIMUC16Q8WXI7B*08167TSHERLTTLMUC16Q8WXI7B*08168TSHERLTTYMUC16Q8WXI7B*08169TSMPRSSAMUC16Q8WXI7B*08M170YLLEKSRVIMYO3B, MYH15, MYH6,A7E2Y1, B0I1T2,B*08MYH7, MYO1D, MYO3A,O94832, P12883,MYH7BP13533, Q8NEV4,Q8WXR4, Q9Y2K3171FAFRKEALOVGP1Q12889B*08172KLKERNREOVGP1Q12889B*08L173AEAQVGDEBCAMP50895B*44RDY174AEATARLNBCAMP50895B*44VF175AEIEPKADGBCAMP50895B*44176AEIEPKADGBCAMP50895B*44SW177TEVGTMNLBCAT1P54687B*44F178NELFRDGVBCL2L1Q07817B*44NW179REAGDEFEBCL2L1Q07817B*44L180REAGDEFEBCL2L1Q07817B*44LRY181GEGPKTSWCRABP2P29373B*44182KEATEAQSCTAGE4, CTAGE10P,Q96RT6B*44/B*40LCTAGE16P, CTAGE5,CTAGE1183YEKGIMQKETV1, ETV4, ETV5P43268B*44/B*49V184AELEALTDLEYA2O00167B*44W185AERQPGAAFAM83HQ6ZRV2B*44SL186REGPEEPGFAM83HQ6ZRV2B*44L187GEAQTRIAFOLR1P15328B*44W188AEFAKKQPFUNDC2Q9BWH2B*44WW189KEFLFNMYHOXA9, HOXA10,P28356B*44HOXB9, HOXC9,HOXC10, HOXD9,HOXD10190YEVARILNLHOXD9P28356B*44191EEDAALFKAIRF4Q15306B*44W192YEFKFPNRLLGALS1P09382B*44/B*18/B*40193LEAQQEALMAGEA1, MRPL40P43355B*44194KEVDPTSHMAGEA11P43364B*44SY195AEDKRHYSMFN1Q8IWA4B*44V196REMPGGPVMMP12P39900B*44W197AEVLLPRLVMSLNQ13421B*44198QEAARAALMSLNQ13421B*44199REIDESLIFYMSLNQ13421B*44200AESIPTVSFMUC16Q8WXI7B*44201AETILTFHAMUC16Q8WXI7B*44F202HESEATASMUC16Q8WXI7B*44W203IEHSTQAQDMUC16Q8WXI7B*44TL204RETSTSEETMUC16SLQ8WXI7B*44205SEITRIEMMUC16Q8WXI7B*44206SESVTSRTMUC16Q8WXI7B*44SY207TEARATSDMUC16Q8WXI7B*44SW208TEVSRTEAIMUC16Q8WXI7B*44209TEVSRTELMUC16Q8WXI7B*44210VEAADIFQNNXNL2Q5VZ03B*44F211EEKVFPSPLPNOCQ13519B*44W212MEQKQLQKPNOCQ13519B*44RF213KESIPRWYSPINT1O43278B*44Y214VEQTRAGSTDRD5Q8NAT2B*44LL215SEDGLPEGIZNF217O75362B*44HL TABLE 3Additional peptides according to the present invention.Seq IDHLANoSequenceGeneUniprot Accessionallotype216IMFDDAIERALPP, ALPPL2P05187A*02A217VSSSLTLKVBCAMP50895A*02218TIASQRLTPCD70P32970A*02L219PLPRPGAVCTAG2O75638A*02L220RMTTQLLLLFOLR1P15328A*02/B*13221SLLDLYQLFTHL17Q9BXU8A*02/B*35222ALMRLIGCPGPC2Q8N158A*02L223FAHHGRSLIRF4Q15306A*02224SLPRFQVTLIRF4Q15306A*02225SVFAHPRKMAGEA2B, MAGEA2,P43365A*02LMAGEA6, MAGEA12226QVDPKKRISMELKQ14680A*02M227YTFRYPLSLMMP11P24347A*02228RLWDWVPLMRPL51Q4U2R6A*02A229ISVPAKTSLMUC16Q8WXI7A*02230SAFREGTSMUC16Q8WXI7A*02L231SVTESTHHLMUC16Q8WXI7A*02232TISSLTHELMUC16Q8WXI7A*02233GSDTSSKSMUC16Q8WXI7A*02/B*14L234GVATRVDAIMUC16Q8WXI7A*02/B*14235SAIETSAVLMUC16Q8WXI7A*02/B*35236SAIPFSMTLMUC16Q8WXI7A*02/B*35237SAMGTISIMMUC16Q8WXI7A*02/B*35238PLLVLFTIMUC16Q8WXI7A*02/B*51239FAVPTGISMMUC16Q8WXI7A*02/C*03240FSTDTSIVLMUC16Q8WXI7A*02/C*03241RQPNILVHLMUC16Q8WXI7A*02:05242STIPALHEIMUC16Q8WXI7A*02:05243YASEGVKQSPON1Q9HCB6A*02/B*51V244DTDSSVHVTENM4Q6N022A*02QV245LAVEGGQSUBXN8O00124A*02L246RYLAVVHACCR5P51681A*24/A*23VF247ARPPWMWKLK5Q9Y337A*24/B*27VL248SVIQHLGYMSLNQ13421A*24249VYTPTLGTLDNAJC22, MUC16Q8WXI7A*24250HFPEKTTHMUC16Q8WXI7A*24/C*14SF251KQRQVLIFFPCDHB2O9Y5E7A*24/B*15252LYQPRASEPNOCQ13519A*24/A*25M253AYPEIEKFPTTG2, PTTG1O95997A*24/C*04254IIQHLTEQFSTAG3Q9UJ98A*24/C*03255VFVSFSSLFZNF560Q96MR9A*24/B*27256RTEEVLLTFGPR64Q8IZP9A*03K257VTADHSHVALPI, ALPL, ALPP,P05187A*03FALPPL2258GAYAHTVNALPPL2P10696A*03R259KTLELRVAYBCAMP50895A*03/A*32260GTNTVILEYC2orf88Q9BSFOA*03261HTFGLFYQFAM111BQ65J93A*03R262RSRLNPLVFAM83HQ6ZRV2A*03QR263SSSSATISKHOXD3P31249A*03/A*11264AIKVIPTVFKIDO1P14902A*03265QIHDHVNPIDO1P14902A*03/A*11K266ISYSGQFLVIGF2BP1Q9NZI8A*03K267VTDLISPRKLAMA1P25391A*03268GLLGLSLRYLRRTM1Q86UE6A*03/A*11/A*29269RLKGDAWVMELKQ14680A*03YK270AVFNPRFYMMP12P39900A*03/A*11RTY271RMFADDLHMRPL51Q4U2R6A*03NLNK272RQPERTILRMSLNQ13421A*03PR273RVNAIPFTYMSLNQ13421A*03/A*26274KTFPASTVFMUC16Q8WXI7A*03275STTFPTLTKMUC16Q8WXI7A*03276VSKTTGMEMUC16Q8WXI7A*03F277TTALKTTSRDNAJC22, MUC16Q8WXI7A*03/A*66278NLSSITHERMUC16Q8WXI7A*03/A*68279SVSSETTKIMUC16Q8WXI7A*03/A*68KR280SVSGVKTTMUC16Q8WXI7A*03/B*15F281RAKELEATFNLRP7, NLRP2Q9NX02A*03282CLTRTGLFLNLRP7, NLRP2Q9NX02A*03RF283IVQEPTEEKPAGE2, PAGE2BQ7Z2X7A*03/A*11284KSLIKSWKKTCEA1P2, TCEA1,P23193, Q15560A*03/A*11TCEA2285GTVNPTVGTENM4Q6N022A*03/A*11K286TVAPPQGVZBTB12Q9Y330A*03/A*68VK287RRIHTGEKPZNF271, KLF8, ZNF816,Q8IW36A*03/A*11YKZFP28, ZSCAN29,ZNF597, ZNF480,ZNF714, ZNF836,ZNF600, ZNF320,ZNF100, ZNF721,ZNF841, ZNF678,ZNF860, ZNF429,ZNF888, ZNF761,ZNF701, ZNF83,ZNF695, ZNF471,ZNF22, ZNF28,ZNF137P, ZNF665,ZNF606, ZNF430,ZNF34, ZNF616,ZNF468, ZNF160,ZNF765, ZNF845288SPVTSVHGLILRB4Q8NHJ6A*01/B*35GTY289RWEKTDLTMMP11P24347A*01Y290DMDEEIEAEMYO3BQ8WXR4A*01/A*25Y291ETIRSVGYYTENM4Q6N022A*01/A*25292NVTMKVVSVTCN1Q7Z7D3A*01VLY293VPDSGATAALPP, ALPPL2P05187B*07/B*35TAY294YPLRGSSIFALPP, ALPPL2P05187B*07/B*35295YPLRGSSIFALPP, ALPPL2P05187B*07/B*35GL296YPLRGSSIALPP, ALPPL2P05187B*51/B*07297TVREASGLLBCAMP50895B*07298YPTEHVQFBCAMP50895B*07/B*35299HPGSSALHBCAT1P54687B*07/B*35Y300IPMAAVKQABCL2L1Q07817B*07L301SPRRSPRISCDCA5Q96FF9B*07F302RVEEVRALLCDKN2AP42771B*07303LPMWKVTACLDN6P56747B*07F304LPRPGAVLCTAG2Q75638B*07305TPWAESSTDPEP3Q9H4B8B*07/B*35KF306APVIFSHSADPEP2, DPEP3Q9H4B8B*55/B*56/B*07307LPYGPGSEESR1P03372B*07/B*35AAAF308YPEGAAYEESR1P03372B*07/B*35F309FPQSQYPQEYA2O00167B*07/B*35Y310RPNPITIILFBN2P35556B*07311RPLFYVVSLHTR3AP46098B*07312LPYFREFSHTR3AP46098B*07/B*35M313KVKSDRSVHTR3AP46098B*15/B*07F314VPDQPHPEIIRF4Q15306B*07/B*35315SPRENFPDKLK8O60259B*07TL316EPKTATVLLAMA1P25391B*42/B*07317FPFQPGSVLGALS1P09382B*51/B*07318FPNRLNLEALGALS1P09382B*54/B*55/B*07319SPAEPSVYLILRB4Q8NHJ6B*07ATL320FPMSPVTSLILRB4Q8NHJ6B*07/B*51V321SPMDTFLLILILRB4Q8NHJ6B*51/B*07322SPDPSKHLLLRRK1Q385D2B*07/B*35323RPMPNLRSLRRTM1Q86UE6B*55/B*07V324VPYRVVGLMEX3D, MEX3C,A1L020B*51/B*07MEX3B, MEX3A325GPRNAQRVMFN1Q8IWA4B*07L326VPSEIDAAFMMP11P24347B*07/B*35327SPLPVTSLIMUC16Q8WXI7B*07328EPVTSSLPNMUC16Q8WXI7B*07/B*35F329FPAMTESGMUC16Q8WXI7B*07/B*35GMIL330FPFVTGSTEMUC16Q8WXI7B*07/B*35M331FPHPEMTTMUC16Q8WXI7B*07/B*35SM332FPHSEMTTMUC16Q8WXI7B*07/B*35L333FPHSEMTTMUC16Q8WXI7B*07/B*35VM334FPYSEVTTLMUC16Q8WXI7B*07/B*35335HPDPVGPGMUC16Q8WXI7B*07/B*35L336HPKTESATMUC16Q8WXI7B*07/B*35PAAY337HPVETSSALMUC16Q8WXI7B*07/B*35338HVTKTQATMUC16Q8WXI7B*07/B*35F339LPAGTTGSLMUC16Q8WXI7B*07/B*35VF340LPEISTRTMMUC16Q8WXI7B*07/B*35341LPLDTSTTLMUC16Q8WXI7B*07/B*35342LPLGTSMTFMUC16Q8WXI7B*07/B*35343LPSVSGVKMUC16Q8WXI7B*07/B*35TTF344LPTQTTSSLMUC16Q8WXI7B*07/B*35345LPTSESLVSMUC16Q8WXI7B*07/B*35F346LPWDTSTTMUC16Q8WXI7B*07/B*35LF347MPLTTGSQMUC16Q8WXI7B*07/B*35GM348MPNSAIPFSMUC16Q8WXI7B*07/B*35M349MPSLSEAMMUC16Q8WXI7B*07/B*35TSF350NPSSTTTEFMUC16Q8WXI7B*07/B*35351NVLTSTPAFMUC16Q8WXI7B*07/B*35352SPAETSTNMUC16Q8WXI7B*07/B*35M353SPAMTTPSMUC16Q8WXI7B*07/B*35L354SPLPVTSLLMUC16Q8WXI7B*07/B*35355SPLVTSHIMMUC16Q8WXI7B*07/B*35356SPNEFYFTVMUC16Q8WXI7B*07/B*35357SPSPVPTTLMUC16Q8WXI7B*07/B*35358SPSPVTSTLMUC16Q8WXI7B*07/B*35359SPSTIKLTMMUC16Q8WXI7B*07/B*35360SPSVSSNTMUC16Q8WXI7B*07/B*35Y361SPTHVTQSMUC16Q8WXI7B*07/B*35L362SPVPVTSLFMUC16Q8WXI7B*07/B*35363TAKTPDATFMUC16Q8WXI7B*07/B*35364TPLATTQRFMUC16Q8WXI7B*07/B*35365TPLATTQRFMUC16Q8WXI7B*07/B*35TY366TPLTTTGSAMUC16Q8WXI7B*07/B*35EM367TPSVVTEGMUC16Q8WXI7B*07/B*35F368VPTPVFPTMUC16Q8WXI7B*07/B*35M369FPHSEMTTMUC16Q8WXI7B*07/B*35/VB*51370PGGTRQSLMUC16Q8WXI7B*14:02/B*07371LYVDGFTHMUC16Q8WXI7B*35/B*55/WB*07372IPRNPPPTLMYO3BQ8WXR4B*07L373RPRALRDLNLRP7, NLRP2Q9NX02B*07RIL374NPIGDTGVKNLRP7Q8WX94B*07/B*35F375AAASPLLLLNMUP48645B*07376RPRSPAGQNMUP48645B*07/B*55VA377RPRSPAGQNMUP48645B*07/B*55VAAA378RPRSPAGQNMUP48645B*07/B*56VAA379GPFPLVYVLOVGP1Q12889B*07/B*35380IPTYGRTFOVGP1Q12889B*07/B*35381LPEQTPLAFOVGP1Q12889B*07/B*35382SPMHDRWTOVGP1Q12889B*07/B*35F383TPTKETVSLOVGP1Q12889B*07/B*35384YPGLRGSPOVGP1Q12889B*07/B*35M385SPALHIGSVPCDHB5, PCDHB18,Q96TA0, Q9NRJ7,B*07PCDHB17, PCDHB15,Q9UN66, Q9UN67,PCDHB14, PCDHB11,Q9Y5E1, Q9Y5E3,PCDHB10, PCDHB9,Q9Y5E4, Q9Y5E5,PCDHB8, PCDHB6,Q9Y5E6, Q9Y5E7,PCDHB4, PCDHB3,Q9Y5E8, Q9Y5E9,PCDHB2,PCDHB16Q9Y5F2386FPFNPLDFPTTG1O95997B*07/B*35387APLKLSRTPSPON1Q9HCB6B*07/B*55A388SPAPLKLSRSPON1Q9HCB6B*07/B*55/TPAB*56389SPGAQRTFSTAG3, STAG3L3,P0CL83, Q9UJ98B*07FQLSTAG3L2, STAG3L1390NPDLRRNVTCEA2Q15560B*07L391APSTPRITTTCEA2Q15560B*07F392KPIESTLVATMEM158Q8WZ71B*07/B*55393ASKPHVEICRABP1P29762B*08394MYKMKKPIMAGEB3O15480B*08395VLLPRLVSCMSLNQ13421B*08/A*02396REASGLLSLBCAMP50895B*44397REGDTVQLBCAMP50895B*44L398SFEQVVNEBCL2L1Q07817B*44LF399RELLHLVTLCAPN13Q6MZZ7B*44/B*37400GEIEIHLLCCDC146Q8IYE0B*44/B*40401EDLKEELLLCPXCR1Q8N123B*44/B*18402RELANDELICRABP1P29762B*44L403EEAQWVRKFAM111BQ6SJ93B*44Y404NEAIMHQYFAM111BQ6SJ93B*44/B*18405NEIWTHSYFOLR1P15328B*44/B*18406EDGRLVIEFFRAS1Q86XX4B*44/B*18407AEHEGVSVGXYLT2A0PJZ3B*44L408LEKALQVFIDO1P14902B*44409REFVLSKGIDO1P14902B*44DAGL410SEDPSKLEIDO1P14902B*44A411LELPPILVYIDO1P14902B*44/B*18412QEILTQVKQIGF2BP3O00425B*44/B*40413IEALSGKIELIGF2BP3O00425B*44/B*45414EDAALFKAIRF4Q15306B*44W415REEDAALFIRF4Q15306B*44KAW416SEEETRVVMELKQ14680B*44F417AEHFSMIRAMEX3C, MEX3B,A1L020, Q5U5Q3,B*44/B*50MEX3AQ6ZN04418FEDAQGHIMMP11P24347B*44W419HEFGHVLGMMP11P24347B*44/B*40L420FESHSTVSMUC16Q8WXI7B*44A421GEPATTVSLMUC16Q8WXI7B*44422SETTFSLIFMUC16Q8WXI7B*44423SEVPTGTTMUC16Q8WXI7B*44A424TEFPLFSAAMUC16Q8WXI7B*44425SEVPLPMAIMUC16Q8WXI7B*44/B*18426PEKTTHSFMUC16Q8WXI7B*44/C*04:01427HESSSHHDNFE2L3Q9Y4A8B*44L428LDLGLNHINLRP2Q9NX02B*44/B*47429REKFIASVIOVGP1Q12889B*44430DEKILYPEFOVGP1Q12889B*44/B*18431AEQDPDELPOMZP3, ZP3P21754, Q6PJE2B*44/B*41NKA432EEQYIAQFPRAMEP78395B*44/B*18433SDSQVRAFSTAG1, STAG3, STAG2Q8N3U4, Q8WVM7,B*44/B*37Q9UJ98434KEAIREHQTCEA1P2, TCEA1,P23193, Q15560B*44/B*41MTCEA2435REEFVSIDHTMPRSS3P57727B*44L436REPGDIFSEWISP3O95389B*44L437TEAVVTNELXPR1Q9UBH6B*44438SEVDSPNVZNF217O75362B*44L TABLE 4HLA Class I peptides according to the present invention.Seq IDHLANoSequenceGeneUniprot Accessionallotype439EALAKLMSLATP7BP35670B*51440ELFEGLKAFBCAT1P54687A*25441HQITEVGTBCAT1P54687B*15M442ILSKLTDIQYBCAT1P54687B*15443GTFNPVSLBCAT1P54687B*58W444KLSQKGYSBCL2L1Q07817A*32W445LHITPGTAYBCL2L1Q07817B*13446GRIVAFFSFBCL2L1Q07817B*27447MQVLVSRIBCL2L1Q07817B*52/B*13448LSQKGYSWBCL2L1Q07817B*57449RAFSDLTSBCL2L1Q07817C*15QL450KQTFPFPTIC2orf88Q9BSF0B*13451DYLNEWGSCDH3P22223A*23RF452LKVLGVNVCRABP2P29373C*07M453DVKLEKPKDPPA2Q7Z7J5A*68454AQTDPTTGLOXL2, ENTPD4Q9Y4K0B*15Y455AAAANAQVESR1P03372B*35Y456IPLERPLGEESR1P03372B*35VY457NAAAAANAESR1P03372B*35QVY458TDTLIHLMESR1P03372B*37459KVAGERYVETV1, ETV4, ETV5P41161, P43268,A*32/A*31YP50549460RLSSATANFAM83HQ6ZRV2A*26ALY461AQRMTTQLFOLR1P15328B*15L462QRMTTQLLFOLR1P15328B*27/C*07L463VNQSLLDLYFTHL17Q9BXU8A*26464MSALRPLLGPC2Q8N158C*15465DLIESGQLRIDO1P14902A*66466DLIESGQLRIDO1P14902A*66ER467MQMQERDIDO1P14902B*15TL468ALAKLLPLKLK10O43240B*35469QEQSSVVRKLK6Q92876B*45A470QGERLLGALAG3P18627C*03AV471AQRLDPVYLAMC2Q13753B*15F472MRLLVAPLLRRN2O75325B*14473MLNNNALSLRRN2O75325B*35AL474AADGGLRALY6EQ16553C*05SVTL475GRDPTSYPMAGEA11P43364B*39SL476ISYPPLHEWMAGEA3, MAGEA12P43357B*57477RIQQQTNTMEX3AA1L020B*15Y478VVGPKGATIMEX3D, MEX3C,A1L020, Q5U503,C*14MEX3B, MEX3AQ6ZN04, Q86XN8479TEGSHFVEMFN1Q8IWA4B*45A480GRADIMIDFMMP11P24347B*27481GRWEKTDLMMP11P24347B*27TY482GRWEKTDLMMP11P24347B*27TYR483VRFPVHAAMMP11P24347B*27LVW484AWLRSAAAMMP11P24347B*56485VRFPVHAAMMP11P24347C*07L486DRFFWLKVMMP12P39900B*14487GMADILVVFMMP12P39900B*15488RSFSLGVPMRPL51Q4U2R6A*31R489EVSGLSTEMSLNQ13421A*68R490AEVQKLLGMSLNQ13421B*50P491EAYSSTSSMUC16Q8WXI7A*25W492EVTPWISLTMUC16Q8WXI7A*25L493DTNLEPVTMUC16Q8WXI7A*68R494ETTASLVSRMUC16Q8WXI7A*68495EVPSGATTMUC16Q8WXI7A*68EVSR496EVPTGTTAMUC16Q8WXI7A*68EVSR497EVSRTEVISMUC16Q8WXI7A*68SR498EVYPELGTMUC16Q8WXI7A*68QGR499SSETTKIKRMUC16Q8WXI7A*68500AHVLHSTLMUC16Q8WXI7B*14501IQIEPTSSLMUC16Q8WXI7B*14502SGDQGITSLMUC16Q8WXI7B*14503TVFDKAFTAMUC16Q8WXI7B*14A504TVSSVNQGMUC16Q8WXI7B*14L505YVPTGAITQMUC16Q8WXI7B*14A506HQFITSTNTMUC16Q8WXI7B*15F507TSIFSGQSLMUC16Q8WXI7B*15508TVAKTTTTFMUC16Q8WXI7B*15509GRGPGGVSMUC16Q8WXI7B*27W510RRIPTEPTFMUC16Q8WXI7B*27511SRIPQDVSMUC16Q8WXI7B*27W512SRSPENPSMUC16Q8WXI7B*27W513SRTEISSSRMUC16Q8WXI7B*27514SRTEVASSMUC16Q8WXI7B*27R515TRIEMESTFMUC16Q8WXI7B*27516TASTPISTFMUC16Q8WXI7B*35517TAETILTFHMUC16Q8WXI7B*35AF518TSDFPTITVMUC16Q8WXI7B*35519VTSLLTPGMUC16Q8WXI7B*35MV520THSAMTHGMUC16Q8WXI7B*38F521THSTASQGMUC16Q8WXI7B*38F522THSTISQGFMUC16Q8WXI7B*38523APKGIPVKPMUC16Q8WXI7B*55TSA524AVSPTVQGMUC16Q8WXI7C*07L525QRFPHSEMMUC16Q8WXI7C*07526SVPDILSTMUC16Q8WXI7C*07527QSTPYVNSMUC16Q8WXI7C*16V528TRTGLFLRFNLRP7, NLRP2Q9NX02B*27529PFSNPRVLNLRP2Q9NX02C*04530MLPRAALLNLRP7Q8WX94B*51531QGAQLRGANLRP7, NLRP2Q8WX94B*52L532AISFSYKAWOVGP1Q12889A*25533GQHLHLETPRAMEP78395B*15F534CRPGALQIERAD54BQ9Y620C*02L535IKDVRKIKRNF17Q9BXT8B*13536VQDQACVARNF17Q9BXT8B*15KF537IRRLKELKDRPL37A, RPL37AP8A6NKH3, P61513n/aQ538QLEKALKEISAGE1Q9NXZ1C*05539IPIPSTGSVSPINT1O43278B*35/B*42EM540AGIPAVALWSPINT1O43278B*58541RLSPAPLKLSPON1Q9HCB6B*13542QIIDEEETQSPON1Q9HCB6B*15F543MRLSPAPLSPON1Q9HCB6B*27K544LRNPSIQKLSPON1Q9HCB6C*07545RVGPPLLITMEM158Q8WZ71B*15546GRAFFAAATMEM158Q8WZ71B*27F547EVNKPGVYTMPRSS3P57727A*68TR548VSEASLVSSZBTB12Q9Y330C*05I549ARSKLQQGZNF217O75362B*27L550RRFKEPWFZNF217, ZNF516,O15090, O75362,B*27LZNF536Q92618551RLHTGEKPZNF816, ZNF813,A2RRD8, A6NHJ4,A*30YKZNF578, ZNF599,A6NK21, A6NK53,ZNF600, ZNF320,A6NK75, A6NN14,ZNF525, ZNF485,A6NNF4, A6NP11,ZNF860, ZNF429,A8MTY0, A8MUV8,ZNF808, ZNF888,B4DU55, B4DX44,ZNF761, ZNF701,B4DXR9, O14628,ZNF83, ZNF167, ZFP62,O14709, O15090,ZNF28, ZSCAN21,O43309, O43345,ZNF91, ZNF229,O43361, O75346,ZNF702P, ZNF528,O75373, O75437,ZNF468, ZNF765,O75820, O95600,ZNF845O95780, P0CB33,POCJ79, P0DKX0,P10073, P17019,P17026, P17035,P17038, P17040,P17097, P35789,P51522, P51815,P52742, Q02386,Q03923, Q03924,Q03936, Q03938,Q05481, Q08AN1,Q09FC8, Q0VGE8,Q14584, Q14586,Q14590, Q14591,Q14593, Q15928,Q15929, Q15937,Q16587, Q2M3W8,Q2M3X9, Q2VY69,Q3KP31, Q3MI56,Q3SXZ3, Q4V348,Q53GI3, Q5HY98,Q5JNZ3, Q5SXM1,Q5VIY5, Q5VV52,Q68DY1, Q6AZW8,Q6P280, Q6P9G9,Q6PDB4, Q6ZMV8,Q6ZMW2, Q6ZN06,Q6ZN08, Q6ZN19,Q6ZN57, Q6ZNA1,Q6ZNG1, Q6ZR52,Q76KX8, Q7L2R6,Q7L945, Q7Z3V5,Q7Z7L9, Q86TJ5,Q86UE3, Q86V71,Q86XN6, Q86XU0,Q86Y25, Q81W36,Q8IWY8, Q8IYN0,Q8IZ26, Q8N4W9,Q8N782, Q8N703,Q8N823, Q8N859,Q8N8C0, Q8N8J6,Q8N972, Q8N988,Q8N9F8, Q8NB50,Q8NCK3, Q8NDQ6,Q8NEM1, Q8NF99,Q8NHY6, Q8TAQ5,Q8TBZ5, Q8TD23,Q8TF20, Q8TF32,Q8TF39, Q8WV37,Q8WX64, Q96CX3,Q96IR2, Q96JC4,Q96LX8, Q96MR9,Q96N22, Q96N38,Q96N58, Q96NI8,Q96NL3, Q96PE6,Q96RE9, Q965E7,Q99676, Q9BX82,Q9H5H4, Q9H7R5,Q9H8G1, Q9H963,Q9HBT7, Q9HCG1,Q9HCL3, Q9NQX6,Q9NV72, Q9P0L1,Q9P255, Q9P2F9,Q9P2J8, Q9UEG4,Q9UII5, Q9UJW7,Q9UL36, Q9Y2Q1,Q9Y473, Q9Y5A6773ALYGKLLKLVPS13BA*02774VYVDDIYVICASC5A*24 TABLE 5HLA Class II peptides according to the present invention.Seq IDNoSequenceAdditional Sequence variantsGene552GVNAMLRKVAVAAASKPHVECRABP1553VNAMLRKVAVAAASKPHVECRABP1554GVNAMLRKVAVAAASKPHCRABP1555VNAMLRKVAVAAASKPHCRABP1556NAMLRKVAVAAASKPHCRABP1557AMLRKVAVAAASKPHCRABP1558LRKVAVAAASKPHCRABP1559RKVAVAAASKPHCRABP1560PNFSGNWKIIRSENFEELLKCRABP2561PNFSGNWKIIRSENFEELLCRABP2562GNWKIIRSENFEELLKVLCRABP2563PNFSGNWKIIRSENFEELCRABP2564GNWKIIRSENFEELLKVCRABP2565NWKIIRSENFEELLKVCRABP2566NWKIIRSENFEELLKCRABP2567NWKIIRSENFEELLCRABP2568WKIIRSENFEELLKCRABP2569WKIIRSENFEELLCRABP2570GNWKIIRSENFCRABP2571PNFSGNWKIIRCRABP2572INFKVGEEFEEQTVCRABP2573RLLSADTKGWVRLQDPPA2574LPDFYNDWMFIAKHLPDLIDO1575VGDDHLLLLQGEQLRRTKLK10576VGDDHLLLLQGEQLRRKLK10577GDDHLLLLQGEQLRRKLK10578DDHLLLLQGEQLRRKLK10579SGGPLVCDETLQGILSKLK10580GGPLVCDETLQGILSKLK10581GGPLVCDETLQGILKLK10582GSQPWQVSLFNGLSFHKLK10583LTVKLPDGYEFKFPNRLNLEAINYLGALS1584TVKLPDGYEFKFPNRLNLEAINYLGALS1585LTVKLPDGYEFKFPNRLNLLGALS1586TVKLPDGYEFKFPNRLNLLGALS1587DQANLTVKLPDGYEFKFPNRLNLLGALS1588VAPDAKSFVLNLGKDSNNLLGALS1589APDAKSFVLNLGKDSNNLLGALS1590RVRGEVAPDAKSFVLNLGLGALS1591VRGEVAPDAKSFVLNLLGALS1592VRGEVAPDAKSFVLNLGLGALS1593GEVAPDAKSFVLNLGLGALS1594VRGEVAPDAKSFVLNLGALS1595VRGEVAPDAKSFVLLGALS1596MAADGDFKIKCVAFDLGALS1597SPDAESLFREALSNKVDELMAGEA4598AESLFREALSNKVDELMAGEA4599AESLFREALSNKVDEMAGEA4600FREALSNKVDEMAGEA4601LSNKVDELAHFLLRKMAGEA4602KDPVAWEAGMLMHMAGEB1603KARDETRGLNVPQMAGEB2604KLITQDLVKLKYLEYRQMAGEB3605LTVAEVQKLLGPHVEGLKAEERHRPMSLN606LTVAEVQKLLGPHVEGLKAEERMSLN607LTVAEVQKLLGPHVEGLKAEEMSLN608LTVAEVQKLLGPHVEGLKAEMSLN609LTVAEVQKLLGPHVEGLKAMSLN610LTVAEVQKLLGPHVEGLKMSLN611LTVAEVQKLLGPHVEGLMSLN612TVAEVQKLLGPHVEGLKMSLN613LTVAEVQKLLGPHVEGMSLN614TVAEVQKLLGPHVEGLMSLN615VAEVQKLLGPHVEGLKMSLN616TVAEVQKLLGPHVEGMSLN617VAEVQKLLGPHVEGLMSLN618VAEVQKLLGPHVEGMSLN619VAEVQKLLGPHVEMSLN620EVQKLLGPHVEGMSLN621LTVAEVQKLLGMSLN622MDALRGLLPVLGQPIIRSIPQGIVAMSLN623ALRGLLPVLGQPIIRSIPQGIVAMSLN624LRGLLPVLGQPIIRSIPQGIVAMSLN625DALRGLLPVLGQPIIRSIPQGMSLN626RGLLPVLGQPIIRSIPQGIVAMSLN627ALRGLLPVLGQPIIRSIPQGMSLN628DALRGLLPVLGQPIIRSIPQMSLN629GLLPVLGQPIIRSIPQGIVAMSLN630ALRGLLPVLGQPIIRSIPQMSLN631DALRGLLPVLGQPIIRSIPMSLN632LLPVLGQPIIRSIPQGIVAMSLN633LRGLLPVLGQPIIRSIPQMSLN634DALRGLLPVLGQPIIRSMSLN635ALRGLLPVLGQPIIRSMSLN636DALRGLLPVLGQPIIRMSLN637ALRGLLPVLGQPIIRMSLN638LRGLLPVLGQPIIRSMSLN639ALRGLLPVLGQPIIMSLN640ALRGLLPVLGQPIMSLN641RGLLPVLGQPIIRMSLN642GLLPVLGQPIIRMSLN643LRGLLPVLGQPIMSLN644RGLLPVLGQPIMSLN645RGLLPVLGQPIIRSIPQGIVAAWRQMSLN646GLLPVLGQPIIRSIPQGIVAAWRQMSLN647LPVLGQPIIRSIPQGIVAAWRQMSLN648GLLPVLGQPIIRSIPQGIVAAMSLN649LLPVLGQPIIRSIPQGIVAAMSLN650LPVLGQPIIRSIPQGIVAAWMSLN651LPVLGQPIIRSIPQGIVAAMSLN652PVLGQPIIRSIPQGIVAAWMSLN653LPVLGQPIIRSIPQGIVAMSLN654PVLGQPIIRSIPQGIVAMSLN655LGQPIIRSIPQGIVAAMSLN656VLGQPIIRSIPQGIVAMSLN657QPIIRSIPQGIVAMSLN658VSTMDALRGLLPVLGQPIIRSIPQGMSLN659VSTMDALRGLLPVLGQPIIRSIPQMSLN660VSTMDALRGLLPVLGQPIIRMSLN661LRGLLPVLGQPIIRSIPQGMSLN662LRTDAVLPLTVAEVQKLLGPHVEGMSLN663RTDAVLPLTVAEVQKLLGPHVEGMSLN664AVLPLTVAEVQKLLGPHVEGMSLN665VLPLTVAEVQKLLGPHVEGMSLN666LPLTVAEVQKLLGPHVEGMSLN667TDAVLPLTVAEVQMSLN668AVLPLTVAEVQKMSLN669VLPLTVAEVQKLLGPHVEGLKAEEMSLN670VLPLTVAEVQKLLGPHVEGLKMSLN671LPLTVAEVQKLLGPHVEGLKMSLN672LRGLLPVLGQPIIRSIPQGIVAAMSLN673IPFTYEQLDVLKHKLDELYPQMSLN674IPFTYEQLDVLKHKLDEMSLN675IPFTYEQLDVLKHKLDMSLN676VPPSSIWAVRPQDLDTCDPRMSLN677IWAVRPQDLDTCDPRMSLN678AVRPQDLDTCDPRMSLN679WGVRGSLLSEADVRALGGLAMSLN680GVRGSLLSEADVRALGGLAMSLN681WGVRGSLLSEADVRALGGLMSLN682GVRGSLLSEADVRALGGLMSLN683VRGSLLSEADVRALGGLAMSLN684WGVRGSLLSEADVRALGGMSLN685GVRGSLLSEADVRALGGMSLN686VRGSLLSEADVRALGGLMSLN687WGVRGSLLSEADVRALGMSLN688GVRGSLLSEADVRALGMSLN689WGVRGSLLSEADVRALMSLN690GSLLSEADVRALGGLMSLN691GVRGSLLSEADVRALMSLN692RGSLLSEADVRALGGMSLN693WGVRGSLLSEADVRAMSLN694GSLLSEADVRALGGMSLN695RGSLLSEADVRALGMSLN696WGVRGSLLSEADVRMSLN697GSLLSEADVRALGMSLN698VRGSLLSEADVRAMSLN699LLSEADVRALGGMSLN700SLLSEADVRALGMSLN701GSLLSEADVRAMSLN702LLSEADVRALGMSLN703LSEADVRALGGMSLN704SEADVRALGGMSLN705EADVRALGGMSLN706LSTERVRELAVALAQKNVKMSLN707LSTERVRELAVALAQKNMSLN708ERVRELAVALAQKNVKMSLN709LSTERVRELAVALAQKMSLN710LSTERVRELAVALAQMSLN711STERVRELAVALAQKMSLN712VTERVRELAVALAQKNMSLN713VRELAVALAQKNVKMSLN714AIPFTYEQLDVLKHKLDEMSLN715GLSTERVRELAVALAQKNMSLN716GLSTERVRELAVALAQMSLN717IPQGIVAAWRQRSSRDPSMSLN718GIVAAWRQRSSRDPSMSLN719IPQGIVAAWRQRSSRMSLN720ALGGLACDLPGRFVAESMSLN721RELAVALAQKNVKLSTEMSLN722LKALLEVNKGHEMSPQMSLN723TFMKLRTDAVLPLTVAMSLN724FMKLRTDAVLPLTVAMSLN725FMKLRTDAVLPLTMSLN726FMKLRTDAVLPLMSLN727TLGLGLQGGIPNGYLVMSLN728DLPGRFVAESAEVLLMSLN729DLPGRFVAESAEVLMSLN730LPGRFVAESAEVLMSLN731DLPGRFVAESAMSLN732ERHRPVRDWILRQRQMSLN733SPRQLLGFPCAEVSGMSLN734SRTLAGETGQEAAPLMSLN735VTSLETLKALLEVNKMSLN736LGLQGGIPNGYLVLMSLN737LQGGIPNGYLVLMSLN738GGIPNGYLVLMSLN739LQGGIPNGYLVLDLMSLN740APERQRLLPAALAMSLN741FVKIQSFLGGAPTMSLN742FVKIQSFLGGMSLN743FVKIQSFLGMSLN744FLKMSPEDIRKMSLN745WELSQLTNSVTELGPYTLDRDMUC16746EITITTQTGYSLATSQVTLPMUC16747ATTPSWVETHSIVIQGFPHMUC16748GIKELGPYTLDRNSLYVNGMUC16749GIKELGPYTLDRNSLMUC16750GPYTLDRNSLYVNGMUC16751GIKELGPYTLDRNMUC16752LGPYTLDRNSLYVMUC16753LGPYTLDRNSLYMUC16754LGPYTLDRNSLMUC16755IELGPYLLDRGSLYVNGMUC16756LGPYLLDRGSLYVNGMUC16757LGPYLLDRGSLYVNMUC16758LGPYLLDRGSLYVMUC16759EELGPYTLDRNSLYVNGMUC16760LKPLFKSTSVGPLYSGMUC16761LKPLFKSTSVGPLYSMUC16762LKPLFKSTSVGPLYMUC16763LKPLFKSTSVGPLMUC16764FDKAFTAATTEVSRTEMUC16765ELGPYTLDRDSLYVNMUC16766GLLKPLFKSTSVGPLMUC16767LLKPLFKSTSVGPLMUC16768SDPYKATSAVVITSTMUC16769SDPYKATSAVVITSMUC16770SRKFNTMESVLQGLLMUC16771SRKFNTMESVLQGMUC16772LGFYVLDRDSLFINMUC16 The present invention furthermore generally relates to the peptides according to the present invention for use in the treatment of proliferative diseases, such as, for example, hepatocellular carcinoma, colorectal carcinoma, glioblastoma, gastric cancer, esophageal cancer, non-small cell lung cancer, small cell lung cancer, pancreatic cancer, renal cell carcinoma, prostate cancer, melanoma, breast cancer, chronic lymphocytic leukemia, Non-Hodgkin lymphoma, acute myeloid leukemia, gallbladder cancer and cholangiocarcinoma, urinary bladder cancer, uterine cancer, head and neck squamous cell carcinoma, mesothelioma. Particularly preferred are the peptides—alone or in combination—according to the present invention selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 772. More preferred are the peptides—alone or in combination—selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 215 (see Tables 1 and 2), and their uses in the immunotherapy of ovarian cancer, hepatocellular carcinoma, colorectal carcinoma, glioblastoma, gastric cancer, esophageal cancer, non-small cell lung cancer, small cell lung cancer, pancreatic cancer, renal cell carcinoma, prostate cancer, melanoma, breast cancer, chronic lymphocytic leukemia, Non-Hodgkin lymphoma, acute myeloid leukemia, gallbladder cancer and cholangiocarcinoma, urinary bladder cancer, uterine cancer, head and neck squamous cell carcinoma, mesothelioma, and preferably ovarian cancer. Thus, another aspect of the present invention relates to the use of the peptides according to the present invention for the—preferably combined—treatment of a proliferative disease selected from the group of ovarian cancer, hepatocellular carcinoma, colorectal carcinoma, glioblastoma, gastric cancer, esophageal cancer, non-small cell lung cancer, small cell lung cancer, pancreatic cancer, renal cell carcinoma, prostate cancer, melanoma, breast cancer, chronic lymphocytic leukemia, Non-Hodgkin lymphoma, acute myeloid leukemia, gallbladder cancer and cholangiocarcinoma, urinary bladder cancer, uterine cancer, head and neck squamous cell carcinoma, mesothelioma. The present invention furthermore relates to peptides according to the present invention that have the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I or—in an elongated form, such as a length-variant—MHC class-II. The present invention further relates to the peptides according to the present invention wherein said peptides (each) consist or consist essentially of an amino acid sequence according to SEQ ID NO: 1 to SEQ ID NO: 772. The present invention further relates to the peptides according to the present invention, wherein said peptide is modified and/or includes non-peptide bonds. The present invention further relates to the peptides according to the present invention, wherein said peptide is part of a fusion protein, in particular fused to the N-terminal amino acids of the HLA-DR antigen-associated invariant chain (Ii), or fused to (or into the sequence of) an antibody, such as, for example, an antibody that is specific for dendritic cells. The present invention further relates to a nucleic acid, encoding the peptides according to the present invention. The present invention further relates to the nucleic acid according to the present invention that is DNA, cDNA, PNA, RNA or combinations thereof. The present invention further relates to an expression vector capable of expressing and/or expressing a nucleic acid according to the present invention. The present invention further relates to a peptide according to the present invention, a nucleic acid according to the present invention or an expression vector according to the present invention for use in the treatment of diseases and in medicine, in particular in the treatment of cancer. The present invention further relates to antibodies that are specific against the peptides according to the present invention or complexes of said peptides according to the present invention with MHC, and methods of making these. The present invention further relates to T-cell receptors (TCRs), in particular soluble TCR (sTCRs) and cloned TCRs engineered into autologous or allogeneic T cells, and functional fragments thereof, and methods of making these, as well as NK cells or other cells bearing said TCR or cross-reacting with said TCRs. The antibodies and TCRs are additional embodiments of the immunotherapeutic use of the peptides according to the present invention. The present invention further relates to a host cell comprising a nucleic acid according to the present invention or an expression vector as described before. The present invention further relates to the host cell according to the present invention that is an antigen presenting cell, and preferably is a dendritic cell. The present invention further relates to a method for producing a peptide according to the present invention, said method comprising culturing the host cell according to the present invention, and isolating the peptide from said host cell or its culture medium. The present invention further relates to said method according to the present invention, wherein the antigen is loaded onto class I or II MHC molecules expressed on the surface of a suitable antigen-presenting cell or artificial antigen-presenting cell by contacting a sufficient amount of the antigen with an antigen-presenting cell. The present invention further relates to the method according to the present invention, wherein the antigen-presenting cell comprises an expression vector capable of expressing or expressing said peptide containing SEQ ID No. 1 to SEQ ID No.: 772, preferably containing SEQ ID No. 1 to SEQ ID No. 215, or a variant amino acid sequence. The present invention further relates to activated T cells, produced by the method according to the present invention, wherein said T cell selectively recognizes a cell which expresses a polypeptide comprising an amino acid sequence according to the present invention. The present invention further relates to a method of killing target cells in a patient which target cells aberrantly express a polypeptide comprising any amino acid sequence according to the present invention, the method comprising administering to the patient an effective number of T cells as produced according to the present invention. The present invention further relates to the use of any peptide as described, the nucleic acid according to the present invention, the expression vector according to the present invention, the cell according to the present invention, the activated T lymphocyte, the T cell receptor or the antibody or other peptide- and/or peptide-MHC-binding molecules according to the present invention as a medicament or in the manufacture of a medicament. Preferably, said medicament is active against cancer. Preferably, said medicament is a cellular therapy, a vaccine or a protein based on a soluble TCR or antibody. The present invention further relates to a use according to the present invention, wherein said cancer cells are ovarian cancer, hepatocellular carcinoma, colorectal carcinoma, glioblastoma, gastric cancer, esophageal cancer, non-small cell lung cancer, small cell lung cancer, pancreatic cancer, renal cell carcinoma, prostate cancer, melanoma, breast cancer, chronic lymphocytic leukemia, Non-Hodgkin lymphoma, acute myeloid leukemia, gallbladder cancer and cholangiocarcinoma, urinary bladder cancer, uterine cancer, head and neck squamous cell carcinoma, mesothelioma, and preferably ovarian cancer cells. The present invention further relates to biomarkers based on the peptides according to the present invention, herein called “targets” that can be used in the diagnosis of cancer, preferably ovarian cancer. The marker can be over-presentation of the peptide(s) themselves, or over-expression of the corresponding gene(s). The markers may also be used to predict the probability of success of a treatment, preferably an immunotherapy, and most preferred an immunotherapy targeting the same target that is identified by the biomarker. For example, an antibody or soluble TCR can be used to stain sections of the tumor to detect the presence of a peptide of interest in complex with MHC. Optionally the antibody carries a further effector function such as an immune stimulating domain or toxin. The present invention also relates to the use of these novel targets in the context of cancer treatment. Both therapeutic and diagnostic uses against additional cancerous diseases are disclosed in the following more detailed description of the underlying expression products (polypeptides) of the peptides according to the invention. ALPP, also known as ALP, PLAP or PALP, encodes an alkaline phosphatase, a metallo-enzyme that catalyzes the hydrolysis of phosphoric acid monoesters (RefSeq, 2002). ALPP was described to be hyper-expressed in various human tumors and their cell lines, particularly in cancers of the testis and ovary (Millan and Fishman, 1995). ALPP was identified as an independent prognostic factor for the survival of osteosarcoma patients which also correlates with lung metastasis. Furthermore, ALPP was described as an immunohistochemical marker of gastrointestinal smooth muscle neoplasms, germ cell tumor precursors, such as carcinoma in situ and gonadoblastoma, and as a promising ovarian cancer biomarker (Ravenni et al., 2014; Wong et al., 2014b; Faure et al., 2016; Han et al., 2012). ALPPL2, also known as GCAP, encodes a membrane bound glycosylated enzyme, localized to testis, thymus and certain germ cell tumors, which is closely related to both the placental and intestinal forms of alkaline phosphatase (RefSeq, 2002). ALPPL2 was shown to be ectopically expressed in seminoma as well as in many pancreatic cancer cell lines at both mRNA and protein levels and to be involved in cancer cell growth and invasion. Additionally, ALPPL2 was described as a potential diagnostic marker of pancreatic ductal adenocarcinoma (Hofmann and Millan, 1993; Dua et al., 2013; Fishman, 1995). RT-PCR for ALPPL2 was described to be suitable for the sensitive detection of residual germ cell tumor cells in peripheral blood and progenitor cell harvests (Hildebrandt et al., 1998). BCAM encodes the basal cell adhesion molecule (Lutheran blood group), a member of the immunoglobulin superfamily and a receptor for the extracellular matrix protein, laminin (RefSeq, 2002). BCAM is a specific receptor for laminin alpha5 (LAMAS), a subunit of laminin-511 (LM-511) that is a major component of basement membranes in various tissues; the BCAM/LAMAS system plays a functional role in the metastatic spreading of KRAS-mutant colorectal cancer as well as in the migration of hepatocellular carcinoma (Kikkawa et al., 2013; Kikkawa et al., 2014; Bartolini et al., 2016). Serum levels of BCAM were found to be significantly increased in breast cancer patients and its over-expression was found to be associated with skin, ovarian and pancreatic cancers as well as with endometrioid endometrial carcinoma, ovarian endometrioid carcinoma and cutaneous squamous cell carcinoma (Kikkawa et al., 2008; Planaguma et al., 2011; Latini et al., 2013; Kim et al., 2015a; Li et al., 2017). Being able to form a fusion protein with AKT2, BCAM was identified as AKT2 kinase activator in high-grade serous ovarian cancer (Kannan et al., 2015). CBX2 encodes chromobox 2 which is a component of the polycomb multiprotein complex, which is required to maintain the transcriptionally repressive state of many genes throughout development via chromatin remodeling and modification of histones (RefSeq, 2002). CBX2 is involved in cell proliferation and metastasis (Clermont et al., 2016). CBX2 is regulated by SMARCE1 leading to suppressed EGFR transcription. CBX2 is involved in the regulation of three tumor suppressor genes encoded in the INK4A/ARF locus (Papadakis et al., 2015; Agherbi et al., 2009; Miyazaki et al., 2008). CBX2 is over-expressed in cancer including breast cancer, ovarian cancer, lung cancer, metastatic castration-resistant and neuroendocrine prostate cancer and basal-like endometrioid endometrial carcinoma (Parris et al., 2010; Clermont et al., 2016; Clermont et al., 2014; Clermont et al., 2015; Jiang et al., 2015; Xu et al., 2016). CBX2 is associated with lower patient survival and metastatic progression. CBX2 is linked to peritumoral inflammatory infiltration, metastatic spread to the cervical lymph nodes, and tumor size (Parris et al., 2014; Clermont et al., 2014; Xu et al., 2016). CBX2 over-expression results in hematopoietic stem cell differentiation and exhaustion (Klauke et al., 2013). CCNA1 encodes cyclin A1, which belongs to the highly conserved cyclin family involved in the regulation of CDK kinases (RefSeq, 2002). Elevated levels of CCNA1 were detected in epithelial ovarian cancer, lymphoblastic leukemic cell lines as well as in childhood acute lymphoblastic leukemia patients. Others have observed over-expression of CCNA1 protein and mRNA in prostate cancer and in tumor tissues of anaplastic thyroid carcinoma patients (Holm et al., 2006; Wegiel et al., 2008; Marlow et al., 2012; Arsenic et al., 2015). Recent studies have shown that silencing of CCNA1 in highly cyclin A1 expressing ML1 leukemic cells slowed S phase entry, decreased proliferation and inhibited colony formation (Ji et al., 2005). CD70 encodes CD70 molecule which is a cytokine that belongs to the tumor necrosis factor (TNF) ligand family. It induces proliferation of co-stimulated T cells, enhances the generation of cytolytic T cells, and contributes to T cell activation. This cytokine is also reported to play a role in regulating B-cell activation, cytotoxic function of natural killer cells, and immunoglobulin synthesis (RefSeq, 2002). Targeting of CD70 may be used to specifically target and kill cancer cells. It may be a potential target in oral cancer (Bundela et al., 2014; Jacobs et al., 2015b; Wang et al., 2016a). CD70 is expressed in head-and-neck squamous cell carcinoma. It is ectopically expressed in lymphomas, renal cell carcinomas, and glioblastomas. CD70 expression levels decrease during melanoma progression. CD70 is highly expressed on CD4+CD25+ T-cells from patients with acute-type adult T-cell leukemia/lymphoma (Jacobs et al., 2015b; Curran et al., 2015; De et al., 2016; Jacobs et al., 2015a; Masamoto et al., 2016; Pich et al., 2016b; Ruf et al., 2015a). CD70 is involved in immune response, cancer development, and cancer progression (Petrau et al., 2014; Pich et al., 2016a). CD70 up-regulation in clear cell renal cell carcinoma is associated with worse survival (Ruf et al., 2015b). Cisplatin mediates cytotoxicity through APCs expressing relatively higher levels of CD70 (Beyranvand et al., 2016). CD70 expression is almost not affected by ionizing radiation. It is associated with radio sensitivity in lung cancer. Single-dose external beam radiation up-regulates CD70 in PC3 cells (Bernstein et al., 2014; Kumari and Garnett-Benson, 2016; Pu et al., 2014). CDH3 (also known as P-cadherin) encodes cadherin 3 which is a classical cadherin of the cadherin superfamily. This calcium-dependent cell-cell adhesion protein is comprised of five extracellular cadherin repeats, a transmembrane region and a highly conserved cytoplasmic tail. This gene is located in a gene cluster in a region on the long arm of chromosome 16 that is involved in loss of heterozygosity events in breast and prostate cancer. In addition, aberrant expression of this protein is observed in cervical adenocarcinomas (RefSeq, 2002). CDH3 is involved in oncogenic signaling and activates integrins, receptor tyrosine kinases, small molecule GTPases, EMT transcription factors, and other cadherin family members. CDH3 signaling induces invasion and metastasis (Albergaria et al., 2011; Paredes et al., 2012; Bryan, 2015; Vieira and Paredes, 2015). Oncogenic activation of CDH3 is involved in gastric carcinogenesis (Resende et al., 2011). CDH3 over-expression promotes breast cancer, bladder cancer, ovarian cancer, prostate cancer, endometrial cancer, skin cancer, gastric cancer, pancreas cancer, and colon cancer (Albergaria et al., 2011; Paredes et al., 2007; Bryan and Tselepis, 2010; Reyes et al., 2013; Vieira and Paredes, 2015). CDH3 is a basal epithelial marker expressed in basal-like breast cancer. BRCA1 carcinomas are characterized by the expression of basal markers like CDH3 and show a high-grade, highly proliferating, ER-negative, and HER3-negative phenotype (Honrado et al., 2006; Palacios et al., 2008; Rastelli et al., 2010; Dewar et al., 2011). CDH3 is a tumor suppressor in melanoma and oral squamous cell carcinoma (Haass et al., 2005; Vieira and Paredes, 2015). CDH3 may be used as EMT marker. There is a shift from E-cadherin to N-cadherin and CDH3 expression during tumor formation and progression (Piura et al., 2005; Bonitsis et al., 2006; Bryan and Tselepis, 2010; Ribeiro and Paredes, 2014). Competitive interaction between CDH3 and beta-catenin causes impaired intercellular interactions and metastases in gastric cancer (Moskvina and Mal'kov, 2010). CDH3 may be an early marker of cancer formation in colon cancer (Alrawi et al., 2006). Dys-regulation of CDH3 is a marker for poor prognosis and increased malignancy (Knudsen and Wheelock, 2005). CDKN2A (also known as p16 and p16INK4a) encodes cyclin dependent kinase inhibitor 2A which generates several transcript variants which differ in their first exons. At least three alternatively spliced variants encoding distinct proteins have been reported, two of which encode structurally related isoforms known to function as inhibitors of CDK4 kinase. The remaining transcript includes an alternate first exon located 20 Kb upstream of the remainder of the gene; this transcript contains an alternate open reading frame (ARF) that specifies a protein which is structurally unrelated to the products of the other variants. This ARF product functions as a stabilizer of the tumor suppressor protein p53 as it can interact with, and sequester, the E3 ubiquitin-protein ligase MDM2, a protein responsible for the degradation of p53 (RefSeq, 2002). CDKN2A is mutated in pancreatic ductal adenocarcinoma, cutaneous malignant melanoma, vulvar squamous cell carcinoma, and biliary tract cancer. Mutations may be inherited and increase the risk for developing pancreatic cancer. CDKN2A is deleted in malignant pleural mesothelioma. CDKN2A is down-regulated in bladder cancer (Clancy et al., 2016; Fabbri et al., 2017; Gan et al., 2016; Kleeff et al., 2016; Nabeshima et al., 2016; Pacholczyk et al., 2016; Petersen, 2016; Sohal et al., 2016; Tatarian and Winter, 2016). CDKN2A is involved in cancer cell proliferation, tumorigenesis, metastasis, Wnt signaling, senescence, apoptosis, and DNA repair mechanism (Gupta et al., 2016; Ko et al., 2016; Low et al., 2016; Sedgwick and D'Souza-Schorey, 2016; Zhao et al., 2016). CDKN2A is a tumor suppressor gene which is down-regulated upon over-expression of the oncogenic protein UHRF1. CDKN2A interacts with p53 to suppress breast cancer (Alhosin et al., 2016; Fry et al., 2017). CDKN2A promotor hyper-methylation is associated with increased risk for low-grade squamous intra-epithelial lesion, high-grade squamous intra-epithelial lesion, and cervical cancer and with smoking habit. CDKN2A is epigenetically dysregulated during the development of hepatocellular carcinoma and esophageal squamous cell carcinoma (Han et al., 2017; Khan et al., 2017; Ma et al., 2016a). CDKN2A may be used in the diagnosis of cervical cancer and oropharyngeal squamous cell carcinoma (Mahajan, 2016; Savone et al., 2016; Tjalma, 2017). CDKN2A expression is caused by HPV infection, a virus which is known to have oncogenic potential (Hoff et al., 2017; Lorincz, 2016). CDKN2B (also known as p15) encodes cyclin dependent kinase inhibitor 2B which lies adjacent to the tumor suppressor gene CDKN2A in a region that is frequently mutated and deleted in a wide variety of tumors. This gene encodes a cyclin-dependent kinase inhibitor, which forms a complex with CDK4 or CDK6, and prevents the activation of the CDK kinases, thus the encoded protein functions as a cell growth regulator that controls cell cycle G1 progression. The expression of this gene was found to be dramatically induced by TGF beta, which suggested its role in the TGF beta induced growth inhibition (RefSeq, 2002). CDKN2B is involved in the regulation of the cell cycle progression and the inhibition of cell proliferation (Hu and Zuckerman, 2014; Roy and Banerjee, 2015). CDKN2B deletion is associated with schistosomal-associated bladder cancer. Mutations in CDKN2B may be involved in inherited susceptibility to glial tumors. CDKN2B is altered in meningiomas and mutated in non-muscle-invasive urothelial carcinoma (Mawrin and Perry, 2010; Melin, 2011; Pollard et al., 2010; Alentorn et al., 2013; Idbaih, 2011; Koonrungsesomboon et al., 2015). CDKN2B is hyper-methylated in acute myeloid leukemia and pituitary adenomas. CDKN2B is aberrantly regulated in cutaneous malignant melanoma (Bailey et al., 2010; Jiang et al., 2014; Popov and Gil, 2010; van den Hurk et al., 2012; Wolff and Bies, 2013; Zhou et al., 2014). CDKN2B interacts with the tumor suppressor RB and is regulated by Miz-1 and TGF-beta (Zhou et al., 2014; Geyer, 2010; Moroy et al., 2011). CDKN2B is a tumor suppressor gene which is affected by long non-coding RNAs. CDKN2B itself in association with AS1 is part of a long non-coding RNA (ANRIL) which may be involved in cancer development (Popov and Gil, 2010; Aguilo et al., 2016; Shi et al., 2013; Wanli and Ai, 2015). CLDN6, also known as claudin 6, encodes a member of the claudin family which is a component of tight junction strands and an integral membrane protein (RefSeq, 2002). CLDN6 expression was shown to be associated with lymph node metastasis and TNM stage in non-small cell lung cancer (Wang et al., 2015b). Furthermore, low expression of CLDN6 was shown to be associated with significantly lower survival rates in patients with non-small cell lung cancer (Wang et al., 2015b). Thus, low CLDN6 expression is an independent prognostic biomarker that indicates worse prognosis in patients with non-small cell lung cancer (Wang et al., 2015b). CLDN6 was shown to be down-regulated in cervical carcinoma and gastric cancer (Zhang et al., 2015e; Lin et al., 2013b). CLDN6 was shown to be up-regulated in BRCA1-related breast cancer and ovarian papillary serous carcinoma (Wang et al., 2013; Heerma van Voss et al., 2014). CLDN6 was described as a tumor suppressor for breast cancer (Zhang et al., 2015e). Gain of CLDN6 expression in the cervical carcinoma cell lines HeLa and C33A was shown to suppress cell proliferation, colony formation in vitro, and tumor growth in vivo, suggesting that CLDN6 may function as a tumor suppressor in cervical carcinoma cells (Zhang et al., 2015e). CLDN6 may play a positive role in the invasion and metastasis of ovarian cancer (Wang et al., 2013). CLDN6 was shown to be consistently expressed in germ cell tumors and thus is a novel diagnostic marker for primitive germ cell tumors (Ushiku et al., 2012). CLDN6 expression was shown to be positive in most tumors of an assessed set of atypical teratoid/rhabdoid tumors of the central nervous system, with strong CLDN6 positivity being a potential independent prognostic factor for outcome of the disease (Dufour et al., 2012). CT45A1, also known as CT45, encodes the cancer/testis antigen family 45 member A1 protein and is located on chromosome Xq26.3 (RefSeq, 2002). CT45 genes were shown to be potential prognostic biomarkers and therapeutic targets in epithelial ovarian cancer (Zhang et al., 2015d). The CT45A1 protein which is usually only expressed in testicular germ cells was shown to be also expressed in lung cancer, breast cancer and ovarian cancer (Chen et al., 2009). CT45A1 was also shown to be associated with poor prognosis and poor outcomes in multiple myeloma (Andrade et al., 2009). CT45A1 was described as gene up-regulating epithelial-mesenchymal transition (EMT) and metastatic genes, promoting EMT and tumor dissemination. Furthermore, CT45A1 was described as being implicated in the initiation or maintenance of cancer stem-like cells, promoting tumorigenesis and malignant progression (Yang et al., 2015b). CT45A1 over-expression in a breast cancer model was shown to result in the up-regulation of various oncogenic and metastatic genes, constitutively activated ERK and CREB signaling pathways and increased tumorigenesis, invasion and metastasis. Silencing of CT45A1 was shown to reduce cancer cell migration and invasion. Thus, CT45A1 may function as a novel proto-oncogene and may be a target for anticancer drug discovery and therapy (Shang et al., 2014). CT45A2 encodes one of a cluster of several similar genes, which is a member of the cancer/testis family of antigens and is located on chromosome Xq26.3 (RefSeq, 2002). CT45A2 was shown to be a novel spliced MLL fusion partner in a pediatric patient with de novo bi-phenotypic acute leukemia and thus might be relevant for leukemogenesis (Cerveira et al., 2010). The cancer/testis antigen family 45 was shown to be frequently expressed in both cancer cell lines and lung cancer specimens (Chen et al., 2005). CT45 genes were shown to be potential prognostic biomarkers and therapeutic targets in epithelial ovarian cancer (Zhang et al., 2015d). CT45A3 encodes the cancer/testis antigen family 45 member A3 protein and is located on chromosome Xq26.3 (RefSeq, 2002). The cancer/testis antigen family 45 was shown to be frequently expressed in both cancer cell lines and lung cancer specimens (Chen et al., 2005). CT45 genes were shown to be potential prognostic biomarkers and therapeutic targets in epithelial ovarian cancer (Zhang et al., 2015d). CT45A4 encodes the cancer/testis antigen family 45 member A4 protein and is located on chromosome Xq26.3 (RefSeq, 2002). The cancer/testis antigen family 45 was shown to be frequently expressed in both cancer cell lines and lung cancer specimens (Chen et al., 2005). CT45 genes were shown to be potential prognostic biomarkers and therapeutic targets in epithelial ovarian cancer (Zhang et al., 2015d). CT45A5 encodes the cancer/testis antigen family 45 member A5 and is located on chromosome Xq26.3 (RefSeq, 2002). The cancer/testis antigen family 45 was shown to be frequently expressed in both cancer cell lines and lung cancer specimens (Chen et al., 2005). CT45 genes were shown to be potential prognostic biomarkers and therapeutic targets in epithelial ovarian cancer (Zhang et al., 2015d). CT45A6 encodes the cancer/testis antigen family 45 member A6 protein and is located on chromosome Xq26.3 (RefSeq, 2002). The cancer/testis antigen family 45 was shown to be frequently expressed in both cancer cell lines and lung cancer specimens (Chen et al., 2005). CT45 genes were shown to be potential prognostic biomarkers and therapeutic targets in epithelial ovarian cancer (Zhang et al., 2015d). CTAG2 encodes cancer/testis antigen 2 which is an auto immunogenic tumor antigen that belongs to the ESO/LAGE family of cancer-testis antigens. This protein is expressed in a wide array of cancers including melanoma, breast cancer, bladder cancer and prostate cancer. This protein is also expressed in normal testis tissue (RefSeq, 2002). CTAG2 is involved in cancer cell migration and invasiveness (Maine et al., 2016). CTAG2 expression is up-regulated by LSAMP resulting in reduced cell proliferation (Baroy et al., 2014). CTAG2 is expressed in liposarcoma, lung cancer, urothelial cancer, and colorectal cancer. CTAG2 is over-expressed in several entities including esophageal squamous cell carcinoma (Kim et al., 2012; Dyrskjot et al., 2012; Hemminger et al., 2014; Forghanifard et al., 2011; McCormack et al., 2013; Shantha Kumara et al., 2012). Engineered T cells against CTAG2 may be used in multiple myeloma treatment. Autoantibodies against CTAG2 may be used in cancer diagnosis. CTAG2 may be a target in immunotherapy. CTAG2 expression is associated with shorter progression-free survival (van et al., 2011; Dyrskjot et al., 2012; Hudolin et al., 2013; Pollack et al., 2012; Rapoport et al., 2015; Wang et al., 2015a). CYP2W1 encodes a member of the cytochrome P450 superfamily of enzymes which are monooxygenases catalyzing many reactions involved in drug metabolism and in the synthesis of cholesterol, steroids and other lipids (RefSeq, 2002). CYP2W1 is over-expressed in a variety of human cancers including hepatocellular, colorectal and gastric cancer. CYP2W1 over-expression is associated with tumor progression and poor survival (Aung et al., 2006; Gomez et al., 2010; Zhang et al., 2014a). Due to tumor-specific expression, CYP2W1 is an interesting drug target or enzymatic activator of pro-drugs during cancer therapy (Karlgren and Ingelman-Sundberg, 2007; Nishida et al., 2010). DPPA2 encodes developmental pluripotency associated 2 and is located on chromosome 3q13.13 (RefSeq, 2002). DPPA2 is over-expressed in gastric cancer, non-small cell lung cancer, epithelial ovarian cancer, and colorectal cancer. DPPA2 is an oncogene up-regulated in several entities. DPPA2 is reciprocally repressed in teratoma (Tung et al., 2013; Ghodsi et al., 2015; John et al., 2008; Raeisossadati et al., 2014; Shabestarian et al., 2015; Tchabo et al., 2009; Western et al., 2011). DPPA2 expression correlates with tumor invasion depth, stage, lymph node metastasis, and aggressiveness (Ghodsi et al., 2015; Raeisossadati et al., 2014; Shabestarian et al., 2015). DPPA2 is involved in the pathogenesis of non-small cell lung cancer (Watabe, 2012). DPPA2 is differentially methylated in thyroid cancer (Rodriguez-Rodero et al., 2013). ENTPD4 (UDPase) encodes ectonucleoside triphosphate diphosphohydrolase 4, a member of the apyrase protein family and may play a role in salvaging nucleotides from lysosomes (RefSeq, 2002). UDPase activity is increased in patients with ovarian cancer or testicular cancer and decreased after chemotherapy (Papadopoulou-Boutis et al., 1985). ESR1 encodes an estrogen receptor, a ligand-activated transcription factor important for hormone binding, DNA binding and activation of transcription, that is essential for sexual development and reproductive function (RefSeq, 2002). Mutations and single nucleotide polymorphisms of ESR1 are associated with risk for different cancer types including liver, prostate, gallbladder and breast cancer. The up-regulation of ESR1 expression is connected with cell proliferation and tumor growth but the overall survival of patients with ESR1 positive tumors is better due to the successfully therapy with selective estrogen receptor modulators (Sun et al., 2015; Hayashi et al., 2003; Bogush et al., 2009; Miyoshi et al., 2010; Xu et al., 2011; Yakimchuk et al., 2013; Fuqua et al., 2014). ESR1 signaling interferes with different pathways responsible for cell transformation, growth and survival like the EGFR/IGFR, PI3K/Akt/mTOR, p53, HER2, NFkappaB and TGF-beta pathways (Frasor et al., 2015; Band and Laiho, 2011; Berger et al., 2013; Skandalis et al., 2014; Mehta and Tripathy, 2014; Ciruelos Gil, 2014). ETV1 encodes ETS variant 1 which is a member of the ETS (E twenty-six) family of transcription factors. The ETS proteins regulate many target genes that modulate biological processes like cell growth, angiogenesis, migration, proliferation and differentiation (RefSeq, 2002). ETV1 is involved in epithelial-to-mesenchymal transition, DNA damage response, AR and PTEN signaling, cancer cell invasion, and metastasis. ETV1 interacts with JMJD2A to promote prostate carcinoma formation and to increase YAP1 expression affecting the Hippo signaling pathway (Mesquita et al., 2015; Baty et al., 2015; Heeg et al., 2016; Higgins et al., 2015; Kim et al., 2016; Lunardi et al., 2015). ETV1 expression is decreased in prostate cancer. ETV1 is over-expressed in pancreatic cancer, gastrointestinal stromal tumors, oligodendroglial tumors, and renal cell carcinoma. ETV1 may be an oncogene in non-small cell lung cancer (Heeg et al., 2016; Gleize et al., 2015; Ta et al., 2016; Al et al., 2015; Hashimoto et al., 2017; Jang et al., 2015). Increased mRNA levels of ETV1 in microvesicles of prostate cancer cell lines are correlated with prostate cancer progression (Lazaro-lbanez et al., 2017). ETV1 is an oncogene which interacts with the Ewing's sarcoma breakpoint protein EWS. ETV1 interacts with Sparc and Has2 which mediate in part cancer cell metastasis and desmoplastic stromal expansion (Heeg et al., 2016; Kedage et al., 2016). ETV1 gene fusion products as well as ETV1 promotor methylation status are diagnostically useful (Angulo et al., 2016; 2015; Kumar-Sinha et al., 2015; Linn et al., 2015). ETV4 (also called E1AF or PEA3) encodes a member of the Ets oncogene family of transcription factors and is involved in the regulation of metastasis gene expression and in the induction of differentiation-associated genes in embryonic stem cell (Akagi et al., 2015; Coutte et al., 1999; Ishida et al., 2006). ETV4 is over-expressed in different cancer entities including breast, lung, colorectal and gastric cancer and is associated with migration, invasion, metastasis and poor prognosis (Benz et al., 1997; Horiuchi et al., 2003; Yamamoto et al., 2004; Keld et al., 2011; Hiroumi et al., 2001). ETV4 is up-regulated by different pathways like ERK/MAPK, HER2, PI3K and Ras following an induction of several targets including MMPs and IL-8 (Maruta et al., 2009; Keld et al., 2010; Chen et al., 2011b; Aytes et al., 2013). ETV5 encodes the ETS variant 5 protein and is located on chromosome 3q28 (RefSeq, 2002). Pathways including ETV5 were described as being deeply related to the epithelial to mesenchymal process in endometrial cancer (Colas et al., 2012). ETV5 was shown to interact with several signaling pathways such as cell-cycle progression and the TGF-beta signaling pathway in the OV90 ovarian cancer cell line, and ETV5 expression was shown to be associated with the expression of the oncogenic transcription factor FOXM1 in ovarian cancer (Llaurado et al., 2012b). Furthermore, ETV5 was shown to be up-regulated in ovarian cancer. In the spheroid model, the inhibition and up-regulation of ETV5 effected cell proliferation, cell migration, cell adhesion to extracellular matrix components, cell-cell adhesion and cell survival. Thus, ETV5 may play a role in ovarian cancer progression, cell dissemination and metastasis (Llaurado et al., 2012a). Chromosomal rearrangements of ETV5 among other members of the oncogenic PEA3 subfamily, were described to occur in prostate tumors and are thought to be one of the major driving forces in the genesis of prostate cancer. Furthermore, ETV5 was also described as an oncoprotein which is implicated in melanomas, breast and some other types of cancer (Oh et al., 2012). ETV5 was suggested to have a significant role in regulating matrix metalloproteinase 2 expression and therefore resorption in human chondrosarcoma, and thus may be a targetable up-stream effector of the metastatic cascade in this cancer (Power et al., 2013). EYA2 encodes EYA transcriptional coactivator and phosphatase 2, a member of the eyes absent (EYA) family of proteins involved in eye development (RefSeq, 2002). EYA2 over-expression has been observed in several tumor types such as epithelial ovarian tumor, prostate, breast cancer, urinary tract cancers, glioblastoma, lung adenocarcinoma, cervical cancer, colon and hematopoietic cancers (Bierkens et al., 2013; Zhang et al., 2005; Guo et al., 2009; Patrick et al., 2013; Kohrt et al., 2014). Studies have revealed that EYA2 influences transcription of TGF beta pathway members as well as phosphorylation of TGFBR2, implying a dual role of EYA2 in the pancreas (Vincent et al., 2014). FAM111B encodes the family with sequence similarity 111 member B, a protein with a trypsin-like cysteine/serine peptidase domain in the C-terminus which leads, in case of a mutation, to mottled pigmentation, telangiectasia, epidermal atrophy, tendon contractures, and progressive pulmonary fibrosis (RefSeq, 2002). FAM111B was found to be down-regulated during metformin and aspirin induced inhibition of pancreatic cancer development (Yue et al., 2015). FAM83H encodes family with sequence similarity 83 member H which plays an important role in the structural development and calcification of tooth enamel. Defects in this gene are a cause of amelogenesis imperfecta type 3 (A13) (RefSeq, 2002). The long non-coding RNA FAM83H-AS1 is involved in cell proliferation, migration, and invasion and regulates MET/EGFR signaling (Zhang et al., 2017). The long non-coding RNA FAM83H-AS1 is over-expressed in lung cancer and colorectal cancer. FAM83H is an oncogene over-expressed in several entities including breast cancer and colorectal cancer (Zhang et al., 2017; Kuga et al., 2013; Snijders et al., 2017; Yang et al., 2016c; Yang et al., 2016b). Increased expression of long non-coding RNA FAM83H-AS1 is associated with shorter overall survival. FAM83H-AS1 is associated with poor prognosis (Yang et al., 2016c; Yang et al., 2016b). FAM83H may be involved in androgen independent prostate cancer (Nalla et al., 2016). FAM83H interacts with CK1alpha to form keratin filaments and desmosomes (Kuga et al., 2016). FBN2, also known as fibrillin 2, encodes a protein which is a component of the connective tissue and may be involved in elastic fiber assembly (RefSeq, 2002). FBN2 was described as an extracellular matrix regulatory protein of TGF-beta signaling activity (Lilja-Maula et al., 2014). Hyper-methylation of FBN2 was described as an epigenetic biomarker for clear cell renal cell carcinoma and early detection of colorectal cancer and as being associated with poor prognosis by colorectal cancer patients (Ricketts et al., 2014; Rasmussen et al., 2016; Yi et al., 2012). FBN2 was shown to be a candidate cell surface target enriched in medulloblastoma which could be used for the development of tumor-specific probes for guided resection in medulloblastoma (Haeberle et al., 2012). FOLR1 encodes the folate receptor 1, which binds folic acid and its reduced derivatives, and transports 5-methyltetrahydrofolate into cells; FOLR1 is a secreted protein that either anchors to membranes via a glycosyl-phosphatidylinositol linkage or exists in a soluble form (RefSeq, 2002). Being a major part of the FOLR1/cSrc/ERK1/2/NFκB/p53 pathway, which is required for the up-take of folic acid, FOLR1 is able to regulate the proliferation of cancer cells such as breast, lung and colon cancer (Kuo and Lee, 2016; Cheung et al., 2016). FOLR1 was found to be widely expressed in epithelial ovarian cancer, where its expression increases with tumor stage and might represent a potential therapeutic target (Leung et al., 2016; Ponte et al., 2016; Moore et al., 2016; Hou et al., 2017; Notaro et al., 2016; Bergamini et al., 2016). Reducing FOLR1 expression during colorectal cancer therapy was shown to increase the effectiveness of 5-fluorouracil treatment (Tsukihara et al., 2016). FOLR1 represents an ideal tumor-associated marker for immunotherapy for triple-negative breast cancer as well as colon cancer (Liang et al., 2016; Song et al., 2016). GPR64 encodes adhesion G protein-coupled receptor G2, a member of the G protein-coupled receptor family described as an epididymis-specific transmembrane protein (RefSeq, 2002). In breast cancer cell lines, knockdown of GPR64 resulted in a strong reduction in cell adhesion as well as in cell migration (Peeters et al., 2015). HOXA10 encodes homebox A10. This gene is part of the A cluster on chromosome 7 and encodes a DNA-binding transcription factor that may regulate gene expression, morphogenesis, and differentiation. More specifically, it may function in fertility, embryo viability, and regulation of hematopoietic lineage commitment. Read-through transcription exists between this gene and the downstream homeobox A9 (HOXA9) gene (RefSeq, 2002). HOXA10 is a stem cell factor whose expression correlates with CD133 expression in glioma and may be involved in cancer progression. HOXA10 is involved in cancer cell proliferation, migration, invasion, and metastasis. HOXA10 is involved in multidrug resistance by inducing P-gp and MRP1 expression. HOXA10 promotes epithelial-to-mesenchymal transition. HOXA10 may be a downstream target of miR-218/PTEN/AKT/PI3K signaling. HOXA10 promotes expression of the AML-associated transcription factor Prdm16. HOXA10 may mediate G1 cell cycle arrest in a p21-dependent manner. HOXA10 is involved in TGF-beta2/p38 MAPK signaling promoting cancer cell invasion in a MMP-3-dependent manner (Carrera et al., 2015; Cui et al., 2014; Emmrich et al., 2014; Han et al., 2015; Li et al., 2014a; Li et al., 2016a; Sun et al., 2016; Xiao et al., 2014; Yang et al., 2016a; Yi et al., 2016; Yu et al., 2014; Zhang et al., 2014b; Zhang et al., 2015b). HOXA10 is up-regulated in gastric cancer and acute myeloid leukemia. HOXA10 is differentially expressed in oral squamous cell carcinoma. HOXA10 is differentially methylated in non-serous ovarian carcinoma and glioblastoma (Carrera et al., 2015; Han et al., 2015; Kurscheid et al., 2015; Niskakoski et al., 2014; Oue et al., 2015; Shima et al., 2014). HOXA10 methylation status may be used in breast cancer diagnosis. HOXA10 and CD44 co-expression is correlated with tumor size and patient survival in gastric cancer. HOXA10 and miR-196b co-expression is correlated with poor prognosis in gastric cancer (Han et al., 2015; Lim et al., 2013; Uehiro et al., 2016). SGI-110 treatment hypo-methylate HOXA10 which sensitizes ovarian cancer cells for chemotherapy (Fang et al., 2014a). HOXA9 encodes homebox protein A9. This gene is part of the A cluster on chromosome 7 and encodes a DNA-binding transcription factor which may regulate gene expression, morphogenesis, and differentiation. A specific translocation event which causes a fusion between this gene and the NUP98 gene has been associated with myeloid leukemogenesis (RefSeq, 2002). HOXA9 is expressed in acute myeloid leukemia and high expression is associated with adverse prognosis. HOXA9 and MEIS1 co-expression induces AML. HOXA9 is down-regulated in cervical cancer. HOXA9 is frequently methylated in endometrial cancer (Alvarado-Ruiz et al., 2016; Chen et al., 2015; Li et al., 2016b; Li et al., 2016e; Sykes et al., 2016). The gene fusion product NUP98-HOXA9 acts as oncogene (Abe et al., 2016; Sontakke et al., 2016). Response to cisplatin-based chemotherapy is linked to HOXA9 promotor methylation status. HOXA9, MEIS1, and MN1 co-expression in leukemia make the cells sensitive to pharmacologic inhibition of DOT1 L (Li et al., 2016c; Riedel et al., 2016; Xylinas et al., 2016). HOXA9 is a tumor suppressor whose expression may be used to diagnose cancer (Ma et al., 2016b). HOXA9 mediates leukemic stem cell self-renewal and HIF-2alpha deletion accelerates this process (Vukovic et al., 2015; Zhu et al., 2016). HOXB9 encodes homebox B9 which is a member of the Abd-B homeobox family and encodes a protein with a homeobox DNA-binding domain. It is included in a cluster of homeobox B genes located on chromosome 17. The encoded nuclear protein functions as a sequence-specific transcription factor that is involved in cell proliferation and differentiation. Increased expression of this gene is associated with some cases of leukemia, prostate cancer and lung cancer (RefSeq, 2002). HOXB9 is involved in angiogenic pathways which are regulated by miR-192. HOXB9 is a downstream target of Wnt/beta-catenin signaling induced by N-acetylgalactosaminyltransferase resulting in metastasis. HOXB9 may regulate mesenchymal-to-epithelial transition in gastric carcinoma and colon adenocarcinoma and epithelial-to-mesenchymal transition in breast cancer and hepatocellular carcinoma in a TGF-beta1-dependent manner. HOXB9 is involved in cell proliferation, migration, and invasion. TGF-beta1 down-regulates HOXB9 in a Kindlin-2/PDAC-dependent manner (Chang et al., 2015b; Darda et al., 2015; Hoshino et al., 2014; Huang et al., 2014; Kwon et al., 2015; Seki et al., 2012; Sha et al., 2015; Wu et al., 2016; Zhan et al., 2014; Zhan et al., 2015; Zhussupova et al., 2014). HOXB9 is differentially expressed in PBRM1 mutated clear cell renal cell carcinoma. HOXB9 is over-expressed in platinum-resistant high-grade serous ovarian cancer, breast cancer, glioma, colon adenocarcinoma, hepatocellular carcinoma, and head and neck squamous cell carcinoma. HOXB9 expression is decreased in gastric carcinoma. HOXB9 is mutated in leukemia (Menezes et al., 2014; Chang et al., 2015b; Darda et al., 2015; Zhan et al., 2014; Zhussupova et al., 2014; Fang et al., 2014b; Hayashida et al., 2010; Kelly et al., 2016; Sha et al., 2013; Shrestha et al., 2012; Wang et al., 2016b; Yuan et al., 2014). HOXB9 expression is regulated by E2F1 and FAT10 (Zhussupova et al., 2014; Yuan et al., 2014). HOXB9 expression is correlated with tumor size in oral cancer. HOXB9 expression is associated with advanced clinical stage in glioma. HOXB9 down-regulation is associated with decreased patient survival in gastric carcinoma (Fang et al., 2014b; Sha et al., 2013; Oliveira-Costa et al., 2015; Tomioka et al., 2010). HOXB9 regulates bladder cancer progression (Zhang et al., 2016b). Long non-coding RNA nc-HOXB9-205 is down-regulated in urothelial carcinoma of the bladder (Luo et al., 2014). BCAS3-HOXB9 gene fusion product is expressed in breast cancer (Schulte et al., 2012). HOXC10 encodes homeobox C10 which belongs to the homeobox family of genes. The homeobox genes encode a highly conserved family of transcription factors that play an important role in morphogenesis in all multicellular organisms. This gene is one of several homeobox HOXC genes located in a cluster on chromosome 12. The protein level is controlled during cell differentiation and proliferation, which may indicate this protein has a role in origin activation (RefSeq, 2002). HOXC10 is involved in chemo resistance by suppressing apoptosis and up-regulating NF-kappaB and DNA damage repair. HOXC10 induces apoptosis and inhibits cell growth. HOXC10 may be involved in cervical cancer progression and invasion (Pathiraja et al., 2014; Sadik et al., 2016; Zhai et al., 2007). HOXC10 is up-regulated in thyroid cancer, cervical squamous cell carcinoma, and breast cancer (Abba et al., 2007; Zhai et al., 2007; Ansari et al., 2012; Feng et al., 2015). HOXC10 expression correlates with shorter recurrence-free and overall survival in ER-negative breast cancer. HOXC10 expression is associated with advanced stage, poor pathologic stage, poor prognosis, cytokine-cytokine receptor interaction, and chemokine signaling pathways in thyroid cancer (Sadik et al., 2016; Feng et al., 2015). HOXC10 is differentially methylated in oral squamous cell carcinoma and small B cell lymophoma (Marcinkiewicz and Gudas, 2014a; Marcinkiewicz and Gudas, 2014b; Rahmatpanah et al., 2006). HOXC9 encodes homebox C9 which belongs to the homeobox family of genes. The homeobox genes encode a highly conserved family of transcription factors that play an important role in morphogenesis in all multicellular organismsThis gene is one of several homeobox HOXC genes located in a cluster on chromosome 12 (RefSeq, 2002). HOXC9 is involved in cancer cell invasion and proliferation. HOXC9 knock-down results in reduced cell viability, migration, invasion, tumorigenicity, and increased autophagy. HOXC9 is involved in chemo resistance in bladder cancer in a miR-193a-3p-dependent manner. HOXC9 is involved in retinoic acid signaling and is involved in cell growth and differentiation (Hur et al., 2016; Kocak et al., 2013; Lv et al., 2015a; Mao et al., 2011; Simeone et al., 1991; Stornaiuolo et al., 1990; Xuan et al., 2016; Zha et al., 2012). HOXC9 is differentially expressed in breast cancer, lung cancer, and neuroblastoma. HOXC9 is methylated in stage I non-small cell lung cancer. HOXC9 is up-regulated in astrocytoma. HOXC9 is expressed in esophageal cancer and cervical cancer (Hur et al., 2016; Xuan et al., 2016; Gu et al., 2007; Lin et al., 2009; Lopez et al., 2006; Okamoto et al., 2007). HOXC9 may be transcriptionally repressed by Smad4 (Zhou et al., 2008). HOXC9 expression is inversely associated with disease-free survival and distant metastasis-free survival in breast cancer. HOXC9 expression is associated with poor prognosis in glioblastoma (Hur et al., 2016; Xuan et al., 2016). HOXC9 inhibits DAPK1 resulting in disturbed autophagy induced by Beclin-1 (Xuan et al., 2016). HOXD10 encodes homeobox D10 protein, which functions as a sequence-specific transcription factor that is expressed in the developing limb buds and is involved in differentiation and limb development (RefSeq, 2002). HOXD10 was identified as target gene of miR-10b, which is up-regulated in gastric cancer (GC) and may have a key role in GC pathogenesis and development (Ma et al., 2015; Wang et al., 2015c). HOXD10 was found to be up-regulated in neck squamous cell carcinoma and urothelial cancer promoting cell proliferation and invasion and may represent a new biomarker for ductal invasive breast carcinoma (Sharpe et al., 2014; Vardhini et al., 2014; Heubach et al., 2015). However, HOXD10 also showed tumor-suppressive functions in cholangiocellular carcinoma by inactivating the RHOC/AKT/MAPK pathway and inducing G1 phase cell cycle arrest (Yang et al., 2015a). As part of the miR-224/HOXD10/p-PAK4/MMP-9 signaling pathway, HOXD10 is contributed to the regulation of cell migration and invasion and provides a new bio target for hepatocellular carcinoma treatment (Li et al., 2014b). HOXD9 encodes homeobox D9 which belongs to the homeobox family of genes. The homeobox genes encode a highly conserved family of transcription factors that play an important role in morphogenesis in all multicellular organisms. This gene is one of several homeobox HOXD genes located at 2q31-2q37 chromosome regions. Deletions that removed the entire HOXD gene cluster or 5′ end of this cluster have been associated with severe limb and genital abnormalities. The exact role of this gene has not been determined (RefSeq, 2002). HOXD9 is involved in epithelial-to-mesenchymal transition, cancer cell migration, invasion, and metastasis in a ZEB1-dependent manner. Over-expressed HOXD9 increases anchorage-independent growth and reduces contact inhibition. HOXD9 is involved in growth arrest and neuronal differentiation. Depletion of HOXD9 results in decreased cell proliferation, cell cycle arrest, and induction of apoptosis (Zha et al., 2012; Lawrenson et al., 2015b; Lv et al., 2015b; Tabuse et al., 2011). HOXD9 is up-regulated in lung squamous carcinoma and invasive hepatocellular carcinoma. HOXD9 is expressed in esophageal carcinoma, astrocytomas and glioblastomas. HOXD9 is differentially expressed in cervical cancer (Bao et al., 2016; Gu et al., 2007; Lv et al., 2015b; Tabuse et al., 2011; Li et al., 2002; Liu et al., 2005). HOXD9 expression is induced by retinoic acid and Wnt signaling (Ishikawa and Ito, 2009). HOXD9 may be involved in cervical carcinogenesis (Lopez-Romero et al., 2015). HOXD9 hyper-methylation is associated with poorer disease-free and overall survival in lymph node metastasis (Marzese et al., 2014). HOXD9 is hyper-methylated in cholangiocarcinoma and melanoma brain metastasis (Marzese et al., 2014; Sriraksa et al., 2013). HOXD9 may be involved in mucinous ovarian carcinoma susceptibility (Kelemen et al., 2015). HOXD9 may be an oncogene (Wu et al., 2013). HTR3A encodes a 5-hydroxytryptamine (serotonin) receptor belonging to the ligand-gated ion channel receptor superfamily that causes fast, depolarizing responses in neurons after activation (RefSeq, 2002). HTR3A (also called 5-HT3) is de-regulated in several cancer types for example a down-regulation in mantle cell lymphomas, a differential expression in diverse B cell tumors and a decreased expression in breast cancer cell lines (Pai et al., 2009; Rinaldi et al., 2010; Ek et al., 2002). IGF2BP1, also known as CRD-BP, encodes a member of the insulin-like growth factor 2 mRNA-binding protein family which functions by binding to the mRNAs of certain genes and regulating their translation (RefSeq, 2002). Two members of the IGF2 mRNA binding protein family, including IGF2BP1 were described as bona fide oncofetal proteins which are de novo synthesized in various human cancers and which may be powerful post-transcriptional oncogenes enhancing tumor growth, drug-resistance and metastasis (Lederer et al., 2014). Expression of IGF2BP1 was reported to correlate with an overall poor prognosis and metastasis in various human cancers (Lederer et al., 2014). Thus, IGF2BP1 was suggested to be a powerful biomarker and candidate target for cancer therapy (Lederer et al., 2014). IGF2BP family members were described to be highly associated with cancer metastasis and expression of oncogenic factors such as KRAS, MYC and MDR1 (Bell et al., 2013). IGF2BP1 was shown to interact with C-MYC and was found to be expressed in the vast majority of colon and breast tumors and sarcomas as well as in benign tumors such as breast fibroadenomas and meningiomas (Ioannidis et al., 2003). IGF2BP1 was shown to be up-regulated in hepatocellular carcinoma and basal cell carcinoma (Noubissi et al., 2014; Zhang et al., 2015c). Up-regulation of IGF2BP1 and other genes was shown to be significantly associated with poor post-surgery prognosis in hepatocellular carcinoma (Zhang et al., 2015c). IGF2BP1 was shown to be a target of the tumor suppressor miR-9 and miR-372 in hepatocellular carcinoma and in renal cell carcinoma, respectively (Huang et al., 2015; Zhang et al., 2015c). Loss of stromal IGF2BP1 was shown to promote a tumorigenic microenvironment in the colon, indicating that IGF2BP1 plays a tumor-suppressive role in colon stromal cells (Hamilton et al., 2015). IGF2BP1 was shown to be associated with stage 4 tumors, decreased patient survival and MYCN gene amplification in neuroblastoma and may therefore be a potential oncogene and an independent negative prognostic factor in neuroblastoma (Bell et al., 2015). IGF2BP1 was described as a direct target of WNT/B-catenin signaling which regulates GLI1 expression and activities in the development of basal cell carcinoma (Noubissi et al., 2014). IGF2BP3 encodes insulin-like growth factor II mRNA binding protein 3, an oncofetal protein, which represses translation of insulin-like growth factor II (RefSeq, 2002). Several studies have shown that IGF2BP3 acts in various important aspects of cell function, such as cell polarization, migration, morphology, metabolism, proliferation and differentiation. In vitro studies have shown that IGF2BP3 promotes tumor cell proliferation, adhesion, and invasion. Furthermore, IGF2BP3 has been shown to be associated with aggressive and advanced cancers (Bell et al., 2013; Gong et al., 2014). IGF2BP3 over-expression has been described in numerous tumor types and correlated with poor prognosis, advanced tumor stage and metastasis, as for example in neuroblastoma, colorectal carcinoma, intrahepatic cholangiocarcinoma, hepatocellular carcinoma, prostate cancer, and renal cell carcinoma (Bell et al., 2013; Findeis-Hosey and Xu, 2012; Hu et al., 2014; Szarvas et al., 2014; Jeng et al., 2009; Chen et al., 2011a; Chen et al., 2013; Hoffmann et al., 2008; Lin et al., 2013a; Yuan et al., 2009). IRF4 encodes the interferon regulatory factor 4, a transcription factor that negatively regulates Toll-like-receptor (TLR) signaling in lymphocytes, what is central to the activation of innate and adaptive immune system (RefSeq, 2002). IRFA is considered to be a key regulator of several steps in lymphoid, myeloid, and dendritic cell differentiation and maturation and is characterized by varying within the hematopoietic system in a lineage and stage-specific way (Shaffer et al., 2009; Gualco et al., 2010). IRF4 plays a pivotal role in adaptive immunity, cell growth, differentiation and tumorigenesis of chronic myeloid leukemia, primary central nervous system lymphoma, T-cell lymphoma, HTLV-I-induced adult T cell leukemia and intravascular large B-cell lymphoma (Mamane et al., 2002; Orwat and Batalis, 2012; Bisig et al., 2012; Ponzoni et al., 2014; Manzella et al., 2016). IRF4 is a well-known oncogene that is regulated by enhancer of zeste homolog 2 (EZH2) in multiple myeloma (Alzrigat et al., 2016). KLK14 encodes kallikrein related peptidase 14 which is a member of the kallikrein subfamily of serine proteases that have diverse physiological functions such as regulation of blood pressure and desquamation. The altered expression of this gene is implicated in the progression of different cancers including breast and prostate tumors. The encoded protein is a precursor that is proteolytically processed to generate the functional enzyme. This gene is one of the fifteen kallikrein subfamily members located in a cluster on chromosome 19 (RefSeq, 2002). KLK14 is involved in cell proliferation via phosphorylation of ERK1/2/MAP kinase and tumorigenesis. KLK14 induces PAR-2 signaling. KLK14 may be involved in tumor progression, growth, invasion, and angiogenesis (Walker et al., 2014; Borgono et al., 2007; Chung et al., 2012a; Devetzi et al., 2013; Gratio et al., 2011; Sanchez et al., 2012; Zhang et al., 2012a). KLK14 is down-regulated by miR-378/422a and androgen receptor signaling. Androgen receptor signaling up-regulates KLK14 expression in breast cancer (Paliouras and Diamandis, 2008b; Lose et al., 2012; Paliouras and Diamandis, 2007; Paliouras and Diamandis, 2008a; Samaan et al., 2014). KLK14 is over-expressed in chronic lymphocytic leukemia, non-small cell lung cancer, salivary gland tumors, and ovarian cancer. KLK14 is differentially expressed in breast cancer (Planque et al., 2008b; Fritzsche et al., 2006; Hashem et al., 2010; Kontos et al., 2016; Papachristopoulou et al., 2013; Planque et al., 2008a). KLK14 expression is inversely associated with overall survival. KLK14 expression may be used as biomarker and to predict risk of disease recurrence. KLK14 expression correlates with clinical tumor stage and positive nodal status (Devetzi et al., 2013; Lose et al., 2012; Fritzsche et al., 2006; Kontos et al., 2016; Borgono et al., 2003; Obiezu and Diamandis, 2005; Rabien et al., 2008; Rajapakse and Takahashi, 2007; Talieri et al., 2009). KLK8 encodes the kallikrein related peptidase 8, a serine protease that may be involved in proteolytic cascade in the skin and may serve as a biomarker for ovarian cancer (RefSeq, 2002). KLK8 expression was shown to correlate with the progression of breast cancer colorectal cancer (CRC), endometrial carcinoma and ovarian cancer and might represent a potential independent prognostic indicator for colorectal, breast and ovarian cancer (Liu et al., 2017; Jin et al., 2006; Kountourakis et al., 2009; Darling et al., 2008; Michaelidou et al., 2015; Borgono et al., 2006). KLK8 is able to undergo alternative splicing that generates an mRNA transcript missing exon 4; this alternative variant is, in contrast to KLK8, significantly down-regulated in cancer cells (Angelopoulou and Karagiannis, 2010). Nevertheless, the KLK8-T4 alternative splice variant, alone or in combination, may be a new independent marker of unfavorable prognosis in lung cancer (Planque et al., 2010). KLK8 expression confers a favorable clinical outcome in non-small cell lung cancer by suppressing tumor cell invasiveness (Sher et al., 2006). LAMA1 encodes an alpha 1 subunit of laminin an extracellular matrix glycoprotein with heterotrimeric structure, which constitute a major component of the basement membrane (RefSeq, 2002). LAMA1 is de-regulated in different cancer types including up-regulation in glioblastomas, hyper-methylation in colorectal cancer, abnormal methylation in breast cancer and frameshift mutations in gastric cancer (Scrideli et al., 2008; Choi et al., 2015; Simonova et al., 2015; Kim et al., 2011). TGFbeta can induce the expression of LAMA1. LAMA1 in turn promotes collagenase IV production, which leads to an invasive phenotype in benign tumor cells, but is not sufficient to confer metastatic potential (Chakrabarty et al., 2001; Royce et al., 1992). LAMC2 belongs to the family of laminins, a family of extracellular matrix glycoproteins. Laminins are the major non-collagenous constituent of basement membranes. They have been implicated in a wide variety of biological processes including cell adhesion, differentiation, migration, signaling, neurite outgrowth and metastasis. LAMC2 encodes a protein which is expressed in several fetal tissues and is specifically localized to epithelial cells in skin, lung and kidney (RefSeq, 2002). LAMC2 is highly expressed in anaplastic thyroid carcinoma and is associated with tumor progression, migration, and invasion by modulating signaling of EGFR (Garg et al., 2014). LAMC2 expression predicted poorer prognosis in stage II colorectal cancer patients (Kevans et al., 2011). LAMC2 expression together with three other biomarkers was found to be significantly associated with the presence of LN metastasis in oral squamous cell carcinoma patients (Zanaruddin et al., 2013). LILRB4 (also known as ILT-3) encodes leukocyte immunoglobulin like receptor B4 which is a member of the leukocyte immunoglobulin-like receptor (LIR) family, which is found in a gene cluster at chromosomal region 19q13.4. The receptor is expressed on immune cells where it binds to MHC class I molecules on antigen-presenting cells and transduces a negative signal that inhibits stimulation of an immune response. The receptor can also function in antigen capture and presentation. It is thought to control inflammatory responses and cytotoxicity to help focus the immune response and limit autoreactivity (RefSeq, 2002). Over-expression of LILRB4 may be involved in tolerance of dendritic cells during cancer. LILRB4 may be involved in immune suppression. LILRB4 is involved in cancer immune escape (Zhang et al., 2012b; Trojandt et al., 2016; Cortesini, 2007; de Goeje et al., 2015; Suciu-Foca et al., 2007). LILRB4 expression is induced by TNF-alpha. Over-expression of LILRB4 inhibits NF-kappaB activation, transcription of inflammatory cytokines, and co-stimulatory molecules. LILRB4 is over-expressed by cyclosporine resulting in decreased tumor cytotoxicity by natural killer cells (Si et al., 2012; Thorne et al., 2015; Vlad and Suciu-Foca, 2012). LILRB4 is over-expressed on dendritic cells in cancer. LILRB4 is expressed in monocytic acute myeloid leukemia. LILRB4 is over-expressed in ovarian cancer (Dobrowolska et al., 2013; Khan et al., 2012; Orsini et al., 2014). LILRB4 expression is associated with shorter survival in non-small cell lung cancer. LILRB4 expression may be used in chronic lymphocytic leukemia prognosis (Colovai et al., 2007; de Goeje et al., 2015). LOXL2 encodes an extracellular copper-dependent amine oxidase, known as lysyl oxidase like 2. The enzyme is essential to the biogenesis of connective tissue and catalyses the first step in the formation of crosslinks between collagens and elastin (RefSeq, 2002). LOXL2 was shown to be involved in regulation of extracellular and intracellular cell signaling pathways. Extracellularly, LOXL2 remodels the extracellular matrix of the tumor microenvironment. Intracellularly, it regulates the epithelial-to-mesenchymal transition (Cano et al., 2012; Moon et al., 2014). In general, LOXL2 has been associated with tumor progression including the promotion of cancer cell invasion, metastasis, angiogenesis, and the malignant transformation of solid tumors in various tumors. A high expression of LOXL2 is associated with a poor prognosis (Wu and Zhu, 2015). LOXL2 was shown to be overexpressed in colon, esophageal squamous cell, breast ceel, clear cell renal cell, hepatocellular, cholangio-, lung squamous cell and head and neck squamous cell carcinomas. In various cancer types, the high expression of LOXL2 was associated with higher recurrence, progression, or metastasis. In various cancer cell lines, the high expression of LOXL2 was associated with increased cell mobility and invasion and its silencing showed the opposite effects (Xu et al., 2014a; Kim et al., 2014; Wong et al., 2014a; Hase et al., 2014; Lv et al., 2014; Torres et al., 2015). In gastric cancer, fibroblast-derived LOXL2 was shown potentially to stimulate the motility of gastric cancer cells. The expression of LOXL2 in stromal cells could serve as a prognostic marker (Kasashima et al., 2014). A number of micro RNAs family is significantly reduced in cancer tissues. LOXL2 was shown to be a direct regulator of those tumor-suppressive micro-RNAs (Fukumoto et al., 2016; Mizuno et al., 2016). EGF induces LRRK1 translocation as it is an EGF receptor specific interaction partner (Ishikawa et al., 2012; Hanafusa and Matsumoto, 2011; Reyniers et al., 2014). LRRK1 is a component of the Grb2/Gab2/Shc1 complex and interacts with Arap1. It may be a component of the MAPK signaling in response to cellular stress (Titz et al., 2010). Arsenic trioxide which is used for acute promyelocytic leukemia treatment up-regulates LRRK1 in breast cancer cells (Wang et al., 2011). LRRK1 shows extreme allele-specific expression in familial pancreatic cancer (Tan et al., 2008). LRRK1 encodes leucine rich repeat kinase 1 and is located on chromosome 15q26.3. It belongs to the ROCO proteins, a novel subgroup of Ras-like GTPases (RefSeq, 2002; Korr et al., 2006). LYPD1 encodes LY6/PLAUR domain containing 1 and is located on chromosome 2q21.2 (RefSeq, 2002). LYPD1 is over-expressed in brain metastases derived from breast cancer. LYPD1 is over-expressed in metastasis. LYPD1 is differentially expressed in ovarian cancer. LYPD1 is a tumor suppressor which is down-regulated in CD133+ cancer stem cell-like cells derived from uterine carcinosarcoma (Burnett et al., 2015; Choijamts et al., 2011; Dat et al., 2012; Ge et al., 2015b; Lawrenson et al., 2015a). LYPD1 is a negative regulator of cell proliferation (Salazar et al., 2011). MAGEA11 encodes MAGE family member A11 which is a member of the MAGEA gene family. The members of this family encode proteins with 50 to 80% sequence identity to each other. The promoters and first exons of the MAGEA genes show considerable variability, suggesting that the existence of this gene family enables the same function to be expressed under different transcriptional controls. The MAGEA genes are clustered at chromosomal location Xq28 (RefSeq, 2002). MAGEA11 is a cancer germline antigen which is involved in tumor progression and correlates with poor prognosis and survival in silico. MAGEA11 is involved in PR-B signaling and acts as co-regulator for the androgen receptor. MAGEA11 directly interacts with TIF2. MAGEA11 is involved in hypoxic signaling and knock-down leads to decreased HIF-1alpha expression (Aprelikova et al., 2009; Askew et al., 2009; James et al., 2013; Liu et al., 2011; Su et al., 2012; Wilson, 2010; Wilson, 2011). MAGEA11 is up-regulated in oral squamous cell carcinoma, paclitaxel-resistant ovarian cancer, and during prostate cancer progression (Duan et al., 2003; Wilson, 2010; Ge et al., 2015a; Karpf et al., 2009). MAGEA11 expression is associated with hypo-methylation in prostate and epithelial ovarian cancer (James et al., 2013). MAGEA12 encodes MAGE family member A12 and is closely related to several other genes clustered on chromosome X (RefSeq, 2002). MAGEA12 is expressed in 20.5% of multiple myeloma patients (Andrade et al., 2008). The surfacing of systemic immune reactivity toward a cryptic epitope from the MAGEA12, after temporary regression of a single melanoma metastasis, in response to specific vaccination was reported (Lally et al., 2001). MAGEA12 was expressed at the highest frequencies, relative to the other MAGE antigens, in early stage lesions of malignant melanoma (Gibbs et al., 2000). MAGEA3 encodes melanoma-associated antigen family member A3. MAGEA3 is widely known as cancer-testis antigen (RefSeq, 2002; Pineda et al., 2015; De et al., 1994). MAGEA3 has been known long time for being used in therapeutic vaccination trials of metastatic melanoma cancer. The currently performed percutaneous peptide immunization with MAGEA3 and 4 other antigens of patients with advanced malignant melanoma was shown to contribute significantly to longer overall survival by complete responders compared to incomplete responders (Coulie et al., 2002; Fujiyama et al., 2014). In NSCLC, MAGEA3 was shown to be frequently expressed. The expression of MAGEA3 correlated with higher number of tumor necrosis in NSCLC tissue samples and was shown to inhibit the proliferation and invasion and promote the apoptosis in lung cancer cell line. By the patients with adenocarcinomas, the expression of MAGEA3 was associated with better survival. The whole cell anti MAGEA3 vaccine is currently under the investigation in the promising phase III clinical trial for treatment of NSCLC (Perez et al., 2011; Reck, 2012; Hall et al., 2013; Grah et al., 2014; Liu et al., 2015b). MAGEA3 together with 4 other genes was shown to be frequently expressed in HCC. The expression of those genes was correlated with the number of circulating tumor cells, high tumor grade and advanced stage in HCC patients. The frequency of liver metastasis was shown to be significantly higher in cases with tumor samples that expressed MAGE3 than in those that did not express this gene (Bahnassy et al., 2014; Hasegawa et al., 1998). Cancer stem cell-like side populations isolated from a bladder cancer cell line as well as from lung, colon, or breast cancer cell lines showed expression of MAGEA3 among other cancer-testis antigens. In general, cancer stem cells are known for being resistant to current cancer therapy and cause post-therapeutic cancer recurrence and progression. Thus, MAGEA3 may serve as a novel target for immunotherapeutic treatment in particular of bladder cancer (Yamada et al., 2013; Yin et al., 2014). In head and neck squamous cell carcinoma, the expression of MAGEA3 was shown to be associated with better disease-free survival (Zamuner et al., 2015). Furthermore, MAGEA3 can be used as a prognostic marker for ovarian cancer (Szajnik et al., 2013). MAGEA4, also known as MAGE4, encodes a member of the MAGEA gene family and is located on chromosome Xq28 (RefSeq, 2002). MAGEA4 was described as a cancer testis antigen which was found to be expressed in a small fraction of classic seminomas but not in non-seminomatous testicular germ cell tumors, in breast carcinoma, Epstein-Barr Virus-negative cases of Hodgkin's lymphoma, esophageal carcinoma, lung carcinoma, bladder carcinoma, head and neck carcinoma, and colorectal cancer, oral squamous cell carcinoma, and hepatocellular carcinoma (Ries et al., 2005; Bode et al., 2014; Li et al., 2005; Ottaviani et al., 2006; Hennard et al., 2006; Chen et al., 2003). MAGEA4 was shown to be frequently expressed in primary mucosal melanomas of the head and neck and thus may be a potential target for cancer testis antigen-based immunotherapy (Prasad et al., 2004). MAGEA4 was shown to be preferentially expressed in cancer stem-like cells derived from LHK2 lung adenocarcinoma cells, SW480 colon adenocarcinoma cells and MCF7 breast adenocarcinoma cells (Yamada et al., 2013). Over-expression of MAGEA4 in spontaneously transformed normal oral keratinocytes was shown to promote growth by preventing cell cycle arrest and by inhibiting apoptosis mediated by the p53 transcriptional targets BAX and CDKN1A (Bhan et al., 2012). MAGEA4 was shown to be more frequently expressed in hepatitis C virus-infected patients with cirrhosis and late-stage hepatocellular carcinoma compared to patients with early stage hepatocellular carcinoma, thus making the detection of MAGEA4 transcripts potentially helpful to predict prognosis (Hussein et al., 2012). MAGEA4 was shown to be one of several cancer/testis antigens that are expressed in lung cancer and which may function as potential candidates in lung cancer patients for polyvalent immunotherapy (Kim et al., 2012). MAGEA4 was described as being up-regulated in esophageal carcinoma and hepatocellular carcinoma (Zhao et al., 2002; Wu et al., 2011). A MAGEA4-derived native peptide analogue called p286-1Y2L9L was described as a novel candidate epitope suitable to develop peptide vaccines against esophageal cancer (Wu et al., 2011). MAGEA6 encodes melanoma-associated antigen family member A6. MAGEA3 is widely known as cancer-testis antigen (RefSeq, 2002; Pineda et al., 2015; De et al., 1994). MAGEA6 was shown to be frequently expressed in melanoma, advanced myeloma, pediatric rhabdomyosarcoma, sarcoma, lung, bladder, prostate, breast, and colorectal cancers, head and neck squamous cell, esophageal squamous cell, and oral squamous cell carcinomas (Ries et al., 2005; Hasegawa et al., 1998; Gibbs et al., 2000; Dalerba et al., 2001; Otte et al., 2001; van der Bruggen et al., 2002; Lin et al., 2004; Tanaka et al., 1997). MAGEA6 expression has been associated with shorter progression-free survival in multiple myeloma patients. In contrast in head and neck squamous cell carcinoma, the expression of MAGEA6 was shown to be associated with better disease-free survival (van et al., 2011; Zamuner et al., 2015). MAGEA6 was among a set of genes overexpressed in a paclitaxel-resistant ovarian cancer cell line. Moreover, transfection of MAGEA6 also conferred increased drug resistance to paclitaxel-sensitive cells (Duan et al., 2003). MAGEA6 can be used as a prognostic marker for ovarian cancer (Szajnik et al., 2013). Cancer stem cell-like side populations isolated from lung, colon, or breast cancer cell lines showed expression of MAGEA6 among other cancer-testis antigens (Yamada et al., 2013). MAGEB2 is classified as cancer testis antigen, since it is expressed in testis and placenta, and in a significant fraction of tumors of various histological types, amongst others multiple myeloma and head and neck squamous cell carcinoma (Pattani et al., 2012; van et al., 2011). MELK encodes maternal embryonic leucine zipper kinase and is located on chromosome 9p13.2 (RefSeq, 2002). MELK is a member of the SNF1/AMPK family of serine-threonine kinases and is a cell cycle dependent protein kinase. It plays a key role in multiple cellular processes such as the proliferation, cell cycle progression, mitosis and spliceosome assembly and has recently emerged as an oncogene and a biomarker over-expressed in multiple cancer stem cells (Du et al., 2014). MELK is over-expressed in various cancers, including colon, gastric, breast, ovaries, pancreas, prostate and brain cancer and over-expression correlates with poor prognosis (Pickard et al., 2009; Kuner et al., 2013; Gu et al., 2013; Liu et al., 2015a). Inhibition of MELK is under investigation as a therapeutic strategy for a variety of cancers, including breast cancer, lung cancer and prostate cancer. MELK-T1 inhibits catalytic activity and MELK protein stability and might sensitize tumors to DNA-damaging agents or radiation therapy by lowering the DNA-damage threshold. MELK inhibitor OTSSP167 is undergoing phase I clinical trials (Chung et al., 2012b; Ganguly et al., 2014; Beke et al., 2015). MEX3A encodes a member of the mex-3 RNA binding family which consists of evolutionarily conserved RNA-binding proteins recruited to P bodies and potentially involved in post-transcriptional regulatory mechanisms (Buchet-Poyau et al., 2007). MEX3A is over-expressed and the gene is amplified in Wilms tumors associated with a late relapse (Krepischi et al., 2016). MEX3A regulates CDX2 via a post-transcriptional mechanism with impact in intestinal differentiation, polarity and stemness, contributing to intestinal homeostasis and carcinogeneses (Pereira et al., 2013). MMP-11, also named stromelysin-3, is a member of the stromelysin subgroup belonging to MMPs superfamily, which has been detected in cancer cells, stromal cells and adjacent microenvironment. Differently, MMP-11 exerts a dual effect on tumors. On the one hand MMP-11 promotes cancer development by inhibiting apoptosis as well as enhancing migration and invasion of cancer cells, on the other hand MMP-11 plays a negative role against cancer development via suppressing metastasis in animal models. Overexpression of MMP-11 was discovered in sera of cancer patients compared with normal control group as well as in multiple tumor tissue specimens, such as gastric cancer, breast cancer, and pancreatic cancer (Zhang et al., 2016c). MMP-11 was demonstrated to be over-expressed at mRNA level and protein level in CRC tissue than paired normal mucosa. Further MMP-11 expression was correlated with CRC lymph node metastasis, distant metastasis and TNM stage (Tian et al., 2015). MMP-11 overexpression is associated with aggressive tumor phenotype and unfavorable clinical outcome in upper urinary tract urothelial carcinomas (UTUC) and urinary bladder urothelial carcinomas (UBUC), suggesting it may serve as a novel prognostic and therapeutic target (Li et al., 2016d). MMP12 (also called MME) encodes a member of the matrix metalloproteinase family which is involved in the breakdown of extracellular matrix in normal physiological processes, such as embryonic development, reproduction and tissue remodeling as well as in disease processes, such as arthritis and metastasis (RefSeq, 2002). De-regulation of MMP12 is shown for different cancer entities. MMP12 is up-regulated in lung, skin, pancreatic and gastric cancer and related to tumor invasion and metastasis. In contrast, over-expression of MMP12 mRNA was found in gastric and colorectal cancer and correlated with a better prognosis (Zhang et al., 2007; Yang et al., 2001; Balaz et al., 2002; Zheng et al., 2013; Wen and Cai, 2014; Zhang et al., 2015f). MMP12 is up-regulated by TNF-alpha or EGF via the NF-kappaB/MAPK and JNK/AP-1 pathways (Yu et al., 2010; Yang et al., 2012). MYO3B encodes the myosin IIIB, a member of a myosin-class that is characterized by an amino-terminal kinase domain and shown to be present in photoreceptors (RefSeq, 2002). MYO3B was identified as an antagonist to trastuzumab treatment among HER2+ cell lines (Lapin et al., 2014). Nucleotide polymorphisms in the MYOB3 gene were found to be associated with changes in the AUA Symptom Score after radiotherapy for prostate cancer (Kerns et al., 2013). NFE2L3 encodes nuclear factor, erythroid 2 like 3, a member of the cap ‘n’ collar basic-region leucine zipper family of transcription factors (RefSeq, 2002). Recent work has revealed that loss of NFE2L3 predisposes mice to lymphoma development. Others have observed high levels of NFE2L3 in colorectal cancer cells, whereas aberrant expression of NFE2L3 was found in Hodgkin lymphoma. Furthermore, NFE2L3 exhibited hyper-methylation in ER positive tumors (Kuppers et al., 2003; Chevillard et al., 2011; Palma et al., 2012; Rauscher et al., 2015). NLRP2 (also known as NALP2) encodes the NLR family, pyrin domain containing 2 protein and is involved in the activation of caspase-1 and may also form protein complexes activating proinflammatory caspases. NLRP7 is a paralog of NLRP2 (RefSeq, 2002; Wu et al., 2010; Slim et al., 2012). The PYRIN domain of NLRP2 inhibits cell proliferation and tumor growth of glioblastoma (Wu et al., 2010). An ATM/NLRP2/MDC1-dependent pathway may shut down ribosomal gene transcription in response to chromosome breaks (Kruhlak et al., 2007). Mutations in NLRP2 can cause rare human imprinting disorders such as familial hydatidiform mole, Beckwith-Wiedemann syndrome and familial transient neonatal diabetes mellitus (Aghajanova et al., 2015; Dias and Maher, 2013; Ulker et al., 2013). NLRP2 inhibits NF-kappaB activation (Kinoshita et al., 2005; Kinoshita et al., 2006; Fontalba et al., 2007; Bruey et al., 2004). NLRP7 encodes the NLR family pyrin domain containing 7, a member of the NACHT, leucine rich repeat, and PYD containing (NLRP) protein family that may act as a feedback regulator of caspase-1-dependent interleukin 1-beta secretion (RefSeq, 2002). NLRP7 expression correlates significantly with the depth of tumor invasion and poor prognosis in endometrial cancer and was identified as one of the genes highly expressed in embryonal carcinomas (Ohno et al., 2008; Skotheim et al., 2005). NLRP7 might play a crucial role in cell proliferation in testicular tumorigenesis and represents a promising therapeutic target for testicular germ cell tumors (Okada et al., 2004). OVGP1 or oviduct-specific glycoprotein, encodes a large, carbohydrate-rich, epithelial glycoprotein which is secreted from non-ciliated oviductal epithelial cells and associates with ovulated oocytes, blastomeres and spermatozoan acrosomal regions (RefSeq, 2002). Gain of OVGP1 was shown to be associated with the development of endometrial hyperplasia and endometrial cancer (Woo et al., 2004). OVGP1 was described as a molecular marker for invasion in endometrial tumorigenesis and a differentiation-based marker of different ovarian cancers (Maines-Bandiera et al., 2010; Wang et al., 2009). PAGE2 encodes a member of the PAGE protein family, which is predominantly expressed in testis (Brinkmann et al., 1998). The cancer-testis gene PAGE2 is up-regulated by de-methylation during spontaneous differentiation of colorectal cancer cells resulting in mesenchymal-to-epithelial transition (MET). Accordingly, down-regulation of PAGE2 has been shown in EMT (Yilmaz-Ozcan et al., 2014). A genome-wide screening identifies PAGE2 as a possible regulator of telomere signaling in human cells (Lee et al., 2011). PNOC encodes prepronociceptin which is a preproprotein that is proteolytically processed to generate multiple protein products. These products include nociceptin, nocistatin, and orphanin FQ2 (OFQ2). Nociceptin, also known as orphanin FQ, is a 17-amino acid neuropeptide that binds to the nociceptin receptor to induce increased pain sensitivity, and may additionally regulate body temperature, learning and memory, and hunger. Another product of the encoded preproprotein, nocistatin, may inhibit the effects of nociception (RefSeq, 2002). Inhibition of cancer pain also inhibits tumor growth and lung metastasis. PNOC is involved in morphine tolerance development. PNOC is involved in neuronal growth. PNOC is involved in cell damage, viability, inflammation and impaired immune function (Caputi et al., 2013; Chan et al., 2012; Kirkova et al., 2009; Kuraishi, 2014; Stamer et al., 2011). PNOC is up-regulated in ganglioglioma. PNOC expression is down-regulated in end-stage cancer. PNOC is highly expressed in the plasma of hepatocellular carcinoma patients (Chan et al., 2012; Stamer et al., 2011; Horvath et al., 2004; Spadaro et al., 2006; Szalay et al., 2004). Cebranopadol is an analgesic PNOC peptide may be used in bone cancer treatment and buprenorphine in lung cancer treatment (Davis, 2012; Linz et al., 2014). PNOC is involved in c-Fos expression (Gottlieb et al., 2007; Kazi et al., 2007). PRAME encodes an antigen that is preferentially expressed in human melanomas and acts as a repressor of retinoic acid receptor, likely conferring a growth advantage to cancer cell via this function (RefSeq, 2002). PRAME was shown to be up-regulated in multiple myeloma, clear cell renal cell carcinoma, breast cancer, acute myeloid leukemia, melanoma, chronic myeloid leukemia, head and neck squamous cell carcinoma and osteosarcoma cell lines (Dannenmann et al., 2013; Yao et al., 2014; Zou et al., 2012; Szczepanski and Whiteside, 2013; Zhang et al., 2013; Beard et al., 2013; Abdelmalak et al., 2014; Qin et al., 2014). PRAME is associated with myxoid and round-cell liposarcoma (Hemminger et al., 2014). PRAME is associated with shorter progression-free survival and chemotherapeutic response in diffuse large B-cell lymphoma treated with R-CHOP, markers of poor prognosis in head and neck squamous cell carcinoma, poor response to chemotherapy in urothelial carcinoma and poor prognosis and lung metastasis in osteosarcoma (Tan et al., 2012; Dyrskjot et al., 2012; Szczepanski et al., 2013; Mitsuhashi et al., 2014). PRAME is associated with lower relapse, lower mortality and overall survival in acute lymphoblastic leukemia (Abdelmalak et al., 2014). PRAME may be a prognostic marker for diffuse large B-cell lymphoma treated with R-CHOP therapy (Mitsuhashi et al., 2014). RAD54 encodes a protein belonging to the DEAD-like helicase superfamily. It shares similarity withSaccharomyces cerevisiaeRAD54 and RDH54, both of which are involved in homologous recombination and repair of DNA. This protein binds to double-stranded DNA, and displays ATPase activity in the presence of DNA. This gene is highly expressed in testis and spleen, which suggests active roles in meiotic and mitotic recombination (RefSeq, 2002). Homozygous mutations of RAD54B were observed in primary lymphoma and colon cancer (Hiramoto et al., 1999). RAD54B counteracts genome-destabilizing effects of direct binding of RAD51 to dsDNA in human tumor cells (Mason et al., 2015). RNF17 encodes ring finger protein 17 which is similar to a mouse gene that encodes a testis-specific protein containing a RING finger domain. Alternatively spliced transcript variants encoding different isoforms have been found (RefSeq, 2002). RNF17 is involved in cytokine production and apoptosis. RNF17 enhances c-Myc function (Jnawali et al., 2014; Lee et al., 2013; Yin et al., 1999; Yin et al., 2001). RNF17 is up-regulated upon RHOXF1 knock-down (Seifi-Alan et al., 2014). RNF17 is expressed in liver cancer (Yoon et al., 2011). RNF17 is a cancer-associated marker (de Matos et al., 2015). SDK2 encodes the sidekick cell adhesion molecule 2, a member of the immunoglobulin superfamily that contains two immunoglobulin domains and thirteen fibronectin type III domains which represent binding sites for DNA, heparin and the cell surface (RefSeq, 2002). It was shown that SDK2 guides axonal terminals to specific synapses in developing neurons and promotes lamina-specific targeting of retinal dendrites in the inner plexiform layer (Kaufman et al., 2004; Yamagata and Sanes, 2012). SPDEF (also called PDEF) encodes SAM pointed domain containing ETS transcription factor, a member of the E26 transformation-specific (ETS) family of transcription factors. It is highly expressed in prostate epithelial cells where it functions as an androgen-independent transactivator of prostate specific antigen (PSA) promoter (RefSeq, 2002). SPDEF expression is often lost or down-regulated in late-stage of tumor progression which means that it plays a role in tumor cell invasion and metastasis. In earlier stages of tumor progression SPDEF is sometimes up-regulated. De-regulation of SPDEF is described for several cancer entities including breast, prostate and colorectal cancer (Moussa et al., 2009; Schaefer et al., 2010; Steffan and Koul, 2011). SPDEF induces the transcription of E-cadherin and suppresses thereby cell invasion and migration (Pal et al., 2013). SPDEF interacts with beta-catenin and blocks the transcriptional activity resulting in lower protein levels of the oncogenes cyclin D1 and c-Myc (Noah et al., 2013). SPON1 encodes spondin 1 and is located on chromosome 11p15.2 (RefSeq, 2002). SPON1 is involved in cancer cell proliferation, migration, invasion, and metastasis. SPON1 is involved in Fak and Src signaling. SPON1 is involved in IL-6 maintenance via MEKK/p38 MAPK/NF-kappaB signaling and this may support murine neuroblastoma survival (Chang et al., 2015a; Cheng et al., 2009; Dai et al., 2015). SPON1 is down-regulated by miR-506 (Dai et al., 2015). SPON1 is over-expressed in ovarian cancer (Davidson et al., 2011; Jiao et al., 2013; Pyle-Chenault et al., 2005). SPON1 may have diagnostic potential in cancer prognosis (Pagnotta et al., 2013). STAG3 encodes stromal antigen 3, which is expressed in the nucleus and is a subunit of the cohesin complex which regulates the cohesion of sister chromatids during cell division (RefSeq, 2002). Researchers have reported the involvement of a common allele of STAG3 in the development of epithelial ovarian cancer. Another group has identified STAG3 to be capable of effectively discriminating lung cancer, chronic obstructive lung disease and fibrotic interstitial lung diseases. Others have detected expression of the STAG3 gene in p53 mutated lymphoma cells (Notaridou et al., 2011; Wielscher et al., 2015; Kalejs et al., 2006). TDRD5 encodes tudor domain containing 5 and is located on chromosome 1q25.2 (RefSeq, 2002). TDRD5 may be over-expressed in breast cancer (Jiang et al., 2016). TDRD5 methylation is altered upon resveratrol treatment in triple negative breast cancer (Medina-Aguilar et al., 2017). TDRD5 is part of a run of homozygosity associated with thyroid cancer (Thomsen et al., 2016). TENM4 encodes teneurin transmembrane protein 4 which is expressed in the nervous systems and mesenchymal tissues and is a regulator of chondrogenesis (Suzuki et al., 2014). Among the four most frequently mutated genes was TENM4 showing protein-changing mutations in primary CNS lymphomas (Vater et al., 2015). MDA-MB-175 cell line contains a chromosomal translocation that leads to the fusion of TENM4 and receptors of the ErbB family. Chimeric genes were also found in neuroblastomas (Wang et al., 1999; Boeva et al., 2013). TMPRSS3 encodes transmembrane protease, serine 3 which is a protein that belongs to the serine protease family. The encoded protein contains a serine protease domain, a transmembrane domain, an LDL receptor-like domain, and a scavenger receptor cysteine-rich domain. Serine proteases are known to be involved in a variety of biological processes, whose malfunction often leads to human diseases and disorders. This gene was identified by its association with both congenital and childhood onset autosomal recessive deafness. This gene is expressed in fetal cochlea and many other tissues, and is thought to be involved in the development and maintenance of the inner ear or the contents of the perilymph and endolymph. This gene was also identified as a tumor-associated gene that is overexpressed in ovarian tumors (RefSeq, 2002). TMPRSS3 is involved in cell proliferation, invasion, and migration. TMPRSS3 induces ERK1/2 signaling (Zhang et al., 2016a). TMPRSS3 affects E-cadherin, vimentin, and Twist expression. TMPRSS3 is down-regulated by hexamethylene bisacetamide (Zhang et al., 2016a; Zhang et al., 2004). TMPRSS3 is up-regulated in breast cancer, pancreatic cancer, and ovarian cancer. TMPRSS3 is de-regulated in gastric cancer and pancreatic ductal adenocarcinoma (Rui et al., 2015; Zhang et al., 2016a; Zhang et al., 2004; Amsterdam et al., 2014; lacobuzio-Donahue et al., 2003; Luo et al., 2017; Underwood et al., 2000; Wallrapp et al., 2000). TMPRSS3 is associated with TNM stage, lymph node metastasis, distant organ metastasis, shorter survival, shorter disease-free survival, and poor prognosis. TMPRSS3 may be used as biomarker in cancer. TMPRSS3 mutations are associated with cancer risk. TMPRSS3 may be used for early pancreatic ductal adenocarcinoma detection (Rui et al., 2015; Amsterdam et al., 2014; Luo et al., 2017; Dorn et al., 2014; Luostari et al., 2014; Pelkonen et al., 2015; Sawasaki et al., 2004). TMPRSS3 is hypo-methylated in cancer (Guerrero et al., 2012). VTCN1, also known as B7-H4, encodes a member of the B7 costimulatory protein family which is present on the surface of antigen-presenting cells and interacts with ligands bound to receptors on the surface of T cells (RefSeq, 2002). VTCN1 was shown to be up-regulated in lung cancer, colorectal cancer, hepatocellular carcinoma, osteosarcoma, breast cancer, cervical cancer, urothelial cell carcinoma, gastric cancer, endometrial cancer, thyroid cancer and laryngeal carcinoma (Klatka et al., 2013; Zhu et al., 2013; Vanderstraeten et al., 2014; Shi et al., 2014; Fan et al., 2014; Wang et al., 2014; Leong et al., 2015; Dong and Ma, 2015; Zhang et al., 2015a; Peng et al., 2015; Xu et al., 2015a). VTCN1 is associated with poor overall survival and higher recurrence probability in hepatocellular carcinoma and poor overall survival in osteosarcoma, urothelial cell carcinoma, pancreatic cancer, gastric cancer, cervical cancer, melanoma and thyroid cancer (Zhu et al., 2013; Seliger, 2014; Liu et al., 2014b; Chen et al., 2014; Fan et al., 2014; Dong and Ma, 2015; Zhang et al., 2015a). VTCN1 is associated with clear cell renal cell carcinoma (Xu et al., 2014b). VTCN1 expression levels were shown to be inversely correlated with patient survival in ovarian cancer (Smith et al., 2014). VTCN1 may be a potential prognostic indicator of urothelial cell carcinoma and gastric cancer (Shi et al., 2014; Fan et al., 2014). WNT7A encodes Wnt family member 7A which is a member of the WNT gene family. These proteins have been implicated in oncogenesis and in several developmental processes, including regulation of cell fate and patterning during embryogenesis. This gene is involved in the development of the anterior-posterior axis in the female reproductive tract, and also plays a critical role in uterine smooth muscle pattering and maintenance of adult uterine function. Mutations in this gene are associated with Fuhrmann and Al-Awadi/Raas-Rothschild/Schinzel phocomelia syndromes (RefSeq, 2002). WNT7A is induced by STAT4 resulting in the activation of cancer-associated fibroblasts. WNT7A potentiates TGF-beta receptor signaling. WNT7A is involved in cell proliferation and migration. WNT7A is an upstream inducer of senescence. PG545 interacts with WNT7A resulting in inhibited cell proliferation. WNT7A suppresses tumor growth. WNT7A is involved in Wnt/beta-catenin signaling and regulates hsa-miR29b (Avasarala et al., 2013; Avgustinova et al., 2016; Bikkavilli et al., 2015; Borowicz et al., 2014; Jung et al., 2015; King et al., 2015; Ramos-Solano et al., 2015; Zhao et al., 2017). WNT7A is regulated by miR-15b and down-regulated by DNMT1. Endosulfan disrupts WNT7A. WNT7A is a target gene of miR-199a-5p and miR-195/497. WNT7A is down-regulated by chronic ethanol exposure and rescued by PPAR-delta agonist treatment. Dkk-1 affects WNT7A. Bilobalide enhances WNT7A expression (Kim et al., 2015a; Chandra et al., 2014; Ingaramo et al., 2016; Itesako et al., 2014; Liu et al., 2014a; MacLean et al., 2016; Mercer et al., 2014; Mercer et al., 2015; Xu et al., 2015b). WNT7A is down-regulated and hyper-methylated in cervical cancer. WNT7A is lost in lung cancer. WNT7A is over-expressed in endometrial cancer (Ramos-Solano et al., 2015; Kim et al., 2015b; Liu et al., 2013). WNT7A expression correlates with poor prognosis and poor patient outcome. WNT7A promotor methylation correlates with advanced tumor stage, distant metastasis, and loss of E-cadherin. Decreased WNT7A expression correlates with decreased overall survival in malignant pleural mesothelioma and may be used for chemosensitivity prediction (Avgustinova et al., 2016; King et al., 2015; Kim et al., 2015b; Hirata et al., 2015). WNT7A may be a tumor suppressor gene in nasopharyngeal cancer (Nawaz et al., 2015). DETAILED DESCRIPTION OF THE INVENTION Stimulation of an immune response is dependent upon the presence of antigens recognized as foreign by the host immune system. The discovery of the existence of tumor associated antigens has raised the possibility of using a host's immune system to intervene in tumor growth. Various mechanisms of harnessing both the humoral and cellular arms of the immune system are currently being explored for cancer immunotherapy. Specific elements of the cellular immune response are capable of specifically recognizing and destroying tumor cells. The isolation of T-cells from tumor-infiltrating cell populations or from peripheral blood suggests that such cells play an important role in natural immune defense against cancer. CD8-positive T-cells in particular, which recognize class I molecules of the major histocompatibility complex (MHC)-bearing peptides of usually 8 to 10 amino acid residues derived from proteins or defect ribosomal products (DRIPS) located in the cytosol, play an important role in this response. The MHC-molecules of the human are also designated as human leukocyte-antigens (HLA). The term “T-cell response” means the specific proliferation and activation of effector functions induced by a peptide in vitro or in vivo. For MHC class I restricted cytotoxic T cells, effector functions may be lysis of peptide-pulsed, peptide-precursor pulsed or naturally peptide-presenting target cells, secretion of cytokines, preferably Interferon-gamma, TNF-alpha, or IL-2 induced by peptide, secretion of effector molecules, preferably granzymes or perforins induced by peptide, or degranulation. The term “peptide” is used herein to designate a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids. The peptides are preferably 9 amino acids in length, but can be as short as 8 amino acids in length, and as long as 10, 11, or 12 or longer, and in case of MHC class II peptides (elongated variants of the peptides of the invention) they can be as long as 13, 14, 15, 16, 17, 18, 19 or 20 or more amino acids in length. Furthermore, the term “peptide” shall include salts of a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids. Preferably, the salts are pharmaceutical acceptable salts of the peptides, such as, for example, the chloride or acetate (trifluoroacetate) salts. It has to be noted that the salts of the peptides according to the present invention differ substantially from the peptides in their state(s) in vivo, as the peptides are not salts in vivo. The term “peptide” shall also include “oligopeptide”. The term “oligopeptide” is used herein to designate a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids. The length of the oligopeptide is not critical to the invention, as long as the correct epitope or epitopes are maintained therein. The oligopeptides are typically less than about 30 amino acid residues in length, and greater than about 15 amino acids in length. The term “polypeptide” designates a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids. The length of the polypeptide is not critical to the invention as long as the correct epitopes are maintained. In contrast to the terms peptide or oligopeptide, the term polypeptide is meant to refer to molecules containing more than about 30 amino acid residues. A peptide, oligopeptide, protein or polynucleotide coding for such a molecule is “immunogenic” (and thus is an “immunogen” within the present invention), if it is capable of inducing an immune response. In the case of the present invention, immunogenicity is more specifically defined as the ability to induce a T-cell response. Thus, an “immunogen” would be a molecule that is capable of inducing an immune response, and in the case of the present invention, a molecule capable of inducing a T-cell response. In another aspect, the immunogen can be the peptide, the complex of the peptide with MHC, oligopeptide, and/or protein that is used to raise specific antibodies or TCRs against it. A class I T cell “epitope” requires a short peptide that is bound to a class I MHC receptor, forming a ternary complex (MHC class I alpha chain, beta-2-microglobulin, and peptide) that can be recognized by a T cell bearing a matching T-cell receptor binding to the MHC/peptide complex with appropriate affinity. Peptides binding to MHC class I molecules are typically 8-14 amino acids in length, and most typically 9 amino acids in length. In humans, there are three different genetic loci that encode MHC class I molecules (the MHC-molecules of the human are also designated human leukocyte antigens (HLA)): HLA-A, HLA-B, and HLA-C. HLA-A*01, HLA-A*02, and HLA-B*07 are examples of different MHC class I alleles that can be expressed from these loci. TABLE 6Expression frequencies F of HLA-A*02, HLA-A*01, HLA-A*03,HLA-A*24, HLA-B*07, HLA-B*08 and HLA-B*44 serotypes. Haplotypefrequencies Gf are derived from a study which used HLA-typingdata from a registry of more than 6.5 million volunteer donorsin the U.S. (Gragert et al., 2013). The haplotype frequency isthe frequency of a distinct allele on an individual chromosome.Due to the diploid set of chromosomes within mammalian cells,the frequency of genotypic occurrence of this allele ishigher and can be calculated employing the Hardy-Weinbergprinciple (F = 1 − (1 − Gf)2).Calculated phenotype fromAllelePopulationallele frequency (F)A*02African (N = 28557)32.3%European Caucasian49.3%(N = 1242890)Japanese (N = 24582)42.7%Hispanic, S + Cent Amer.46.1%(N = 146714)Southeast Asian (N = 27978)30.4%A*01African (N = 28557)10.2%European Caucasian30.2%(N = 1242890)Japanese (N = 24582)1.8%Hispanic, S + Cent Amer.14.0%(N = 146714)Southeast Asian (N = 27978)21.0%A*03African (N = 28557)14.8%European Caucasian26.4%(N = 1242890)Japanese (N = 24582)1.8%Hispanic, S + Cent Amer.14.4%(N = 146714)Southeast Asian (N = 27978)10.6%A*24African (N = 28557)2.0%European Caucasian8.6%(N = 1242890)Japanese (N = 24582)35.5%Hispanic, S + Cent Amer.13.6%(N = 146714)Southeast Asian (N = 27978)16.9%B*07African (N = 28557)14.7%European Caucasian25.0%(N = 1242890)Japanese (N = 24582)11.4%Hispanic, S + Cent Amer.12.2%(N = 146714)Southeast Asian (N = 27978)10.4%B*08African (N = 28557)6.0%European Caucasian21.6%(N = 1242890)Japanese (N = 24582)1.0%Hispanic, S + Cent Amer.7.6%(N = 146714)Southeast Asian (N = 27978)6.2%B*44African (N = 28557)10.6%European Caucasian26.9%(N = 1242890)Japanese (N = 24582)13.0%Hispanic, S + Cent Amer.18.2%(N = 146714)Southeast Asian (N = 27978)13.1% The peptides of the invention, preferably when included into a vaccine of the invention as described herein bind to A*02, A*01, A*03, A*24, B*07, B*08 or B*44. A vaccine may also include pan-binding MHC class II peptides. Therefore, the vaccine of the invention can be used to treat cancer in patients that are A*02-, A*01-, A*03-, A*24-, B*07-, B*08- or B*44-positive, whereas no selection for MHC class II allotypes is necessary due to the pan-binding nature of these peptides. If A*02 peptides of the invention are combined with peptides binding to another allele, for example A*24, a higher percentage of any patient population can be treated compared with addressing either MHC class I allele alone. While in most populations less than 50% of patients could be addressed by either allele alone, a vaccine comprising HLA-A*24 and HLA-A*02 epitopes can treat at least 60% of patients in any relevant population. Specifically, the following percentages of patients will be positive for at least one of these alleles in various regions: USA 61%, Western Europe 62%, China 75%, South Korea 77%, Japan 86%. TABLE 7HLA alleles coverage in European Caucasian population(calculated from (Gragert et al., 2013)).coveragecombined(at leastwithonecombinedcombinedB*07 andA-allele)with B*07with B*44B*44A*02/A*0170%78%78%84%A*02/A*0368%76%76%83%A*02/A*2461%71%71%80%A*′01/A*0352%64%65%75%A*01/A*2444%58%59%71%A*03/A*2440%55%56%69%A*02/A*01/A*0384%88%88%91%A*02/A*01/A*2479%84%84%89%A*02/A*03/A*2477%82%83%88%A*01/A*03/A*2463%72%73%81%A*02/A*01/A*03/A*2490%92%93%95% In a preferred embodiment, the term “nucleotide sequence” refers to a heteropolymer of deoxyribonucleotides. The nucleotide sequence coding for a particular peptide, oligopeptide, or polypeptide may be naturally occurring or they may be synthetically constructed. Generally, DNA segments encoding the peptides, polypeptides, and proteins of this invention are assembled from cDNA fragments and short oligonucleotide linkers, or from a series of oligonucleotides, to provide a synthetic gene that is capable of being expressed in a recombinant transcriptional unit comprising regulatory elements derived from a microbial or viral operon. As used herein the term “a nucleotide coding for (or encoding) a peptide” refers to a nucleotide sequence coding for the peptide including artificial (man-made) start and stop codons compatible for the biological system the sequence is to be expressed by, for example, a dendritic cell or another cell system useful for the production of TCRs. As used herein, reference to a nucleic acid sequence includes both single stranded and double stranded nucleic acid. Thus, for example for DNA, the specific sequence, unless the context indicates otherwise, refers to the single strand DNA of such sequence, the duplex of such sequence with its complement (double stranded DNA) and the complement of such sequence. The term “coding region” refers to that portion of a gene which either naturally or normally codes for the expression product of that gene in its natural genomic environment, i.e., the region coding in vivo for the native expression product of the gene. The coding region can be derived from a non-mutated (“normal”), mutated or altered gene, or can even be derived from a DNA sequence, or gene, wholly synthesized in the laboratory using methods well known to those of skill in the art of DNA synthesis. The term “expression product” means the polypeptide or protein that is the natural translation product of the gene and any nucleic acid sequence coding equivalents resulting from genetic code degeneracy and thus coding for the same amino acid(s). The term “fragment”, when referring to a coding sequence, means a portion of DNA comprising less than the complete coding region, whose expression product retains essentially the same biological function or activity as the expression product of the complete coding region. The term “DNA segment” refers to a DNA polymer, in the form of a separate fragment or as a component of a larger DNA construct, which has been derived from DNA isolated at least once in substantially pure form, i.e., free of contaminating endogenous materials and in a quantity or concentration enabling identification, manipulation, and recovery of the segment and its component nucleotide sequences by standard biochemical methods, for example, by using a cloning vector. Such segments are provided in the form of an open reading frame uninterrupted by internal non-translated sequences, or introns, which are typically present in eukaryotic genes. Sequences of non-translated DNA may be present downstream from the open reading frame, where the same do not interfere with manipulation or expression of the coding regions. The term “primer” means a short nucleic acid sequence that can be paired with one strand of DNA and provides a free 3′-OH end at which a DNA polymerase starts synthesis of a deoxyribonucleotide chain. The term “promoter” means a region of DNA involved in binding of RNA polymerase to initiate transcription. The term “isolated” means that the material is removed from its original environment (e.g., the natural environment, if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment. The polynucleotides, and recombinant or immunogenic polypeptides, disclosed in accordance with the present invention may also be in “purified” form. The term “purified” does not require absolute purity; rather, it is intended as a relative definition, and can include preparations that are highly purified or preparations that are only partially purified, as those terms are understood by those of skill in the relevant art. For example, individual clones isolated from a cDNA library have been conventionally purified to electrophoretic homogeneity. Purification of starting material or natural material to at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated. Furthermore, a claimed polypeptide which has a purity of preferably 99.999%, or at least 99.99% or 99.9%; and even desirably 99% by weight or greater is expressly encompassed. The nucleic acids and polypeptide expression products disclosed according to the present invention, as well as expression vectors containing such nucleic acids and/or such polypeptides, may be in “enriched form”. As used herein, the term “enriched” means that the concentration of the material is at least about 2, 5, 10, 100, or 1000 times its natural concentration (for example), advantageously 0.01%, by weight, preferably at least about 0.1% by weight. Enriched preparations of about 0.5%, 1%, 5%, 10%, and 20% by weight are also contemplated. The sequences, constructs, vectors, clones, and other materials comprising the present invention can advantageously be in enriched or isolated form. The term “active fragment” means a fragment, usually of a peptide, polypeptide or nucleic acid sequence, that generates an immune response (i.e., has immunogenic activity) when administered, alone or optionally with a suitable adjuvant or in a vector, to an animal, such as a mammal, for example, a rabbit or a mouse, and also including a human, such immune response taking the form of stimulating a T-cell response within the recipient animal, such as a human. Alternatively, the “active fragment” may also be used to induce a T-cell response in vitro. As used herein, the terms “portion”, “segment” and “fragment”, when used in relation to polypeptides, refer to a continuous sequence of residues, such as amino acid residues, which sequence forms a subset of a larger sequence. For example, if a polypeptide were subjected to treatment with any of the common endopeptidases, such as trypsin or chymotrypsin, the oligopeptides resulting from such treatment would represent portions, segments or fragments of the starting polypeptide. When used in relation to polynucleotides, these terms refer to the products produced by treatment of said polynucleotides with any of the endonucleases. In accordance with the present invention, the term “percent identity” or “percent identical”, when referring to a sequence, means that a sequence is compared to a claimed or described sequence after alignment of the sequence to be compared (the “Compared Sequence”) with the described or claimed sequence (the “Reference Sequence”). The percent identity is then determined according to the following formula: percent identity=100[1−(C/R)]wherein C is the number of differences between the Reference Sequence and the Compared Sequence over the length of alignment between the Reference Sequence and the Compared Sequence, wherein(i) each base or amino acid in the Reference Sequence that does not have a corresponding aligned base or amino acid in the Compared Sequence and(ii) each gap in the Reference Sequence and(iii) each aligned base or amino acid in the Reference Sequence that is different from an aligned base or amino acid in the Compared Sequence, constitutes a difference and(iiii) the alignment has to start at position 1 of the aligned sequences; and R is the number of bases or amino acids in the Reference Sequence over the length of the alignment with the Compared Sequence with any gap created in the Reference Sequence also being counted as a base or amino acid. If an alignment exists between the Compared Sequence and the Reference Sequence for which the percent identity as calculated above is about equal to or greater than a specified minimum Percent Identity then the Compared Sequence has the specified minimum percent identity to the Reference Sequence even though alignments may exist in which the herein above calculated percent identity is less than the specified percent identity. As mentioned above, the present invention thus provides a peptide comprising a sequence that is selected from the group of consisting of SEQ ID NO: 1 to SEQ ID NO: 772 or a variant thereof which is 88% homologous to SEQ ID NO: 1 to SEQ ID NO: 772, or a variant thereof that will induce T cells cross-reacting with said peptide. The peptides of the invention have the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I or elongated versions of said peptides to class II. In the present invention, the term “homologous” refers to the degree of identity (see percent identity above) between sequences of two amino acid sequences, i.e. peptide or polypeptide sequences. The aforementioned “homology” is determined by comparing two sequences aligned under optimal conditions over the sequences to be compared. Such a sequence homology can be calculated by creating an alignment using, for example, the ClustalW algorithm. Commonly available sequence analysis software, more specifically, Vector NTI, GENETYX or other tools are provided by public databases. A person skilled in the art will be able to assess, whether T cells induced by a variant of a specific peptide will be able to cross-react with the peptide itself (Appay et al., 2006; Colombetti et al., 2006; Fong et al., 2001; Zaremba et al., 1997). By a “variant” of the given amino acid sequence the inventors mean that the side chains of, for example, one or two of the amino acid residues are altered (for example by replacing them with the side chain of another naturally occurring amino acid residue or some other side chain) such that the peptide is still able to bind to an HLA molecule in substantially the same way as a peptide consisting of the given amino acid sequence in consisting of SEQ ID NO: 1 to SEQ ID NO: 772. For example, a peptide may be modified so that it at least maintains, if not improves, the ability to interact with and bind to the binding groove of a suitable MHC molecule, such as HLA-A*02 or -DR, and in that way it at least maintains, if not improves, the ability to bind to the TCR of activated T cells. These T cells can subsequently cross-react with cells and kill cells that express a polypeptide that contains the natural amino acid sequence of the cognate peptide as defined in the aspects of the invention. As can be derived from the scientific literature and databases (Rammensee et al., 1999; Godkin et al., 1997), certain positions of HLA binding peptides are typically anchor residues forming a core sequence fitting to the binding motif of the HLA receptor, which is defined by polar, electrophysical, hydrophobic and spatial properties of the polypeptide chains constituting the binding groove. Thus, one skilled in the art would be able to modify the amino acid sequences set forth in SEQ ID NO: 1 to SEQ ID NO 772, by maintaining the known anchor residues, and would be able to determine whether such variants maintain the ability to bind MHC class I or II molecules. The variants of the present invention retain the ability to bind to the TCR of activated T cells, which can subsequently cross-react with and kill cells that express a polypeptide containing the natural amino acid sequence of the cognate peptide as defined in the aspects of the invention. The original (unmodified) peptides as disclosed herein can be modified by the substitution of one or more residues at different, possibly selective, sites within the peptide chain, if not otherwise stated. Preferably those substitutions are located at the end of the amino acid chain. Such substitutions may be of a conservative nature, for example, where one amino acid is replaced by an amino acid of similar structure and characteristics, such as where a hydrophobic amino acid is replaced by another hydrophobic amino acid. Even more conservative would be replacement of amino acids of the same or similar size and chemical nature, such as where leucine is replaced by isoleucine. In studies of sequence variations in families of naturally occurring homologous proteins, certain amino acid substitutions are more often tolerated than others, and these are often show correlation with similarities in size, charge, polarity, and hydrophobicity between the original amino acid and its replacement, and such is the basis for defining “conservative substitutions.” Conservative substitutions are herein defined as exchanges within one of the following five groups: Group 1-small aliphatic, nonpolar or slightly polar residues (Ala, Ser, Thr, Pro, Gly); Group 2-polar, negatively charged residues and their amides (Asp, Asn, Glu, Gln); Group 3-polar, positively charged residues (His, Arg, Lys); Group 4-large, aliphatic, nonpolar residues (Met, Leu, Ile, Val, Cys); and Group 5-large, aromatic residues (Phe, Tyr, Trp). Less conservative substitutions might involve the replacement of one amino acid by another that has similar characteristics but is somewhat different in size, such as replacement of an alanine by an isoleucine residue. Highly non-conservative replacements might involve substituting an acidic amino acid for one that is polar, or even for one that is basic in character. Such “radical” substitutions cannot, however, be dismissed as potentially ineffective since chemical effects are not totally predictable and radical substitutions might well give rise to serendipitous effects not otherwise predictable from simple chemical principles. Of course, such substitutions may involve structures other than the common L-amino acids. Thus, D-amino acids might be substituted for the L-amino acids commonly found in the antigenic peptides of the invention and yet still be encompassed by the disclosure herein. In addition, non-standard amino acids (i.e., other than the common naturally occurring proteinogenic amino acids) may also be used for substitution purposes to produce immunogens and immunogenic polypeptides according to the present invention. If substitutions at more than one position are found to result in a peptide with substantially equivalent or greater antigenic activity as defined below, then combinations of those substitutions will be tested to determine if the combined substitutions result in additive or synergistic effects on the antigenicity of the peptide. At most, no more than 4 positions within the peptide would be simultaneously substituted. A peptide consisting essentially of the amino acid sequence as indicated herein can have one or two non-anchor amino acids (see below regarding the anchor motif) exchanged without that the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I or -II is substantially changed or is negatively affected, when compared to the non-modified peptide. In another embodiment, in a peptide consisting essentially of the amino acid sequence as indicated herein, one or two amino acids can be exchanged with their conservative exchange partners (see herein below) without that the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I or -II is substantially changed, or is negatively affected, when compared to the non-modified peptide. The amino acid residues that do not substantially contribute to interactions with the T-cell receptor can be modified by replacement with other amino acid whose incorporation does not substantially affect T-cell reactivity and does not eliminate binding to the relevant MHC. Thus, apart from the proviso given, the peptide of the invention may be any peptide (by which term the inventors include oligopeptide or polypeptide), which includes the amino acid sequences or a portion or variant thereof as given. TABLE 8Variants and motif of the peptides according to SEQ ID NO: 3,225, 13, 17, 84, 108, 113, 114, 147, 36, 51, 172, 54, and 57Position123456789SEQ ID No 3ALIYNLVGIVariantVLAMVMMLMAAVAALAAVVVVLVATVTTLTAQVQQLQASEQ ID No 225SVFAHPRKLVariantLVLILLAMVMIMMAAVAIAAAVIATVTITTAQVQIQQASEQ ID No 13VYTFLSSTLVariantIFFIFFFSEQ ID No 17RFTTMLSTFVariantYIYLYILPosition12345678910SEQ ID No 84KLQPAQTAAKVariantYRFIIYIRIFMMYMRMFVVYVRVFTTYTRTFSEQ ID No 108VLYPVPLESYVariantKRFIKIIRIFMKMMRMFVKVVRVFTKTTRTFPosition123456789SEQ ID No 113QLDSNRLTYVariantSSASESEATTATETEAPosition12345678910SEQ ID No 114VMEQSAGIMYVariantSDSDASSATDTDATTAPosition1234567891011SEQ ID No 147APRWFPQPTVVVariantLFMAIPosition123456789SEQ ID No 36APAAWLRSAVariantLFVMISEQ ID No 51SLRLKNVQLVariantKKVKIKMKFKRKRVKRIKRMKRFKHKHVKHIKHMKHFVIMFRRVRIRMRFHHVHIHMHFLLVLILMLFLRLRVLRILRMLRFLHLHVLHILHMLHFSEQ ID No 172KLKERNRELVariantKKVKIKMKFVIMFHHVHIHMHFRKRKVRKIRKMRKFRRVRIRMRFRHRHVRHIRHMRHFLKLKVLKILKMLKFLLVLILMLFLHLHVLHILHMLHFPosition1234567891011SEQ ID No 54AEITITTQTGYVariantFWLDFDWDDLPosition123456789SEQ ID No 57QESDLRLFLVariantFWYDFDWDYD Longer (elongated) peptides may also be suitable. It is possible that MHC class I epitopes, although usually between 8 and 11 amino acids long, are generated by peptide processing from longer peptides or proteins that include the actual epitope. It is preferred that the residues that flank the actual epitope are residues that do not substantially affect proteolytic cleavage necessary to expose the actual epitope during processing. The peptides of the invention can be elongated by up to four amino acids, that is 1, 2, 3 or 4 amino acids can be added to either end in any combination between 4:0 and 0:4. Combinations of the elongations according to the invention can be found in Table 9. TABLE 9Combinations of the elongations of peptides of the inventionC-terminusN-terminus4030 or 120 or 1 or 210 or 1 or 2 or 300 or 1 or 2 or 3 or 4N-terminusC-terminus4030 or 120 or 1 or 210 or 1 or 2 or 300 or 1 or 2 or 3 or 4 The amino acids for the elongation/extension can be the peptides of the original sequence of the protein or any other amino acid(s). The elongation can be used to enhance the stability or solubility of the peptides. Thus, the epitopes of the present invention may be identical to naturally occurring tumor-associated or tumor-specific epitopes or may include epitopes that differ by no more than four residues from the reference peptide, as long as they have substantially identical antigenic activity. In an alternative embodiment, the peptide is elongated on either or both sides by more than 4 amino acids, preferably to a total length of up to 30 amino acids. This may lead to MHC class II binding peptides. Binding to MHC class II can be tested by methods known in the art. Accordingly, the present invention provides peptides and variants of MHC class I epitopes, wherein the peptide or variant has an overall length of between 8 and 100, preferably between 8 and 30, and most preferred between 8 and 14, namely 8, 9, 10, 11, 12, 13, 14 amino acids, in case of the elongated class II binding peptides the length can also be 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids. Of course, the peptide or variant according to the present invention will have the ability to bind to a molecule of the human major histocompatibility complex (MHC) class I or II. Binding of a peptide or a variant to a MHC complex may be tested by methods known in the art. Preferably, when the T cells specific for a peptide according to the present invention are tested against the substituted peptides, the peptide concentration at which the substituted peptides achieve half the maximal increase in lysis relative to background is no more than about 1 mM, preferably no more than about 1 μM, more preferably no more than about 1 nM, and still more preferably no more than about 100 pM, and most preferably no more than about 10 pM. It is also preferred that the substituted peptide be recognized by T cells from more than one individual, at least two, and more preferably three individuals. In a particularly preferred embodiment of the invention the peptide consists or consists essentially of an amino acid sequence according to SEQ ID NO: 1 to SEQ ID NO: 772. “Consisting essentially of” shall mean that a peptide according to the present invention, in addition to the sequence according to any of SEQ ID NO: 1 to SEQ ID NO 772 or a variant thereof contains additional N- and/or C-terminally located stretches of amino acids that are not necessarily forming part of the peptide that functions as an epitope for MHC molecules epitope. Nevertheless, these stretches can be important to provide an efficient introduction of the peptide according to the present invention into the cells. In one embodiment of the present invention, the peptide is part of a fusion protein which comprises, for example, the 80 N-terminal amino acids of the HLA-DR antigen-associated invariant chain (p33, in the following “Ii”) as derived from the NCBI, GenBank Accession number X00497. In other fusions, the peptides of the present invention can be fused to an antibody as described herein, or a functional part thereof, in particular into a sequence of an antibody, so as to be specifically targeted by said antibody, or, for example, to or into an antibody that is specific for dendritic cells as described herein. In addition, the peptide or variant may be modified further to improve stability and/or binding to MHC molecules in order to elicit a stronger immune response. Methods for such an optimization of a peptide sequence are well known in the art and include, for example, the introduction of reverse peptide bonds or non-peptide bonds. In a reverse peptide bond amino acid residues are not joined by peptide (—CO—NH—) linkages but the peptide bond is reversed. Such retro-inverso peptidomimetics may be made using methods known in the art, for example such as those described in Meziere et al (1997) (Meziere et al., 1997), incorporated herein by reference. This approach involves making pseudopeptides containing changes involving the backbone, and not the orientation of side chains. Meziere et al. (Meziere et al., 1997) show that for MHC binding and T helper cell responses, these pseudopeptides are useful. Retro-inverse peptides, which contain NH—CO bonds instead of CO—NH peptide bonds, are much more resistant to proteolysis. A non-peptide bond is, for example, —CH2—NH, —CH2S—, —CH2CH2—, —CH═CH—, —COCH2—, —CH(OH)CH2—, and —CH2SO—. U.S. Pat. No. 4,897,445 provides a method for the solid phase synthesis of non-peptide bonds (—CH2—NH) in polypeptide chains which involves polypeptides synthesized by standard procedures and the non-peptide bond synthesized by reacting an amino aldehyde and an amino acid in the presence of NaCNBH3. Peptides comprising the sequences described above may be synthesized with additional chemical groups present at their amino and/or carboxy termini, to enhance the stability, bioavailability, and/or affinity of the peptides. For example, hydrophobic groups such as carbobenzoxyl, dansyl, or t-butyloxycarbonyl groups may be added to the peptides' amino termini. Likewise, an acetyl group or a 9-fluorenylmethoxy-carbonyl group may be placed at the peptides' amino termini. Additionally, the hydrophobic group, t-butyloxycarbonyl, or an amido group may be added to the peptides' carboxy termini. Further, the peptides of the invention may be synthesized to alter their steric configuration. For example, the D-isomer of one or more of the amino acid residues of the peptide may be used, rather than the usual L-isomer. Still further, at least one of the amino acid residues of the peptides of the invention may be substituted by one of the well-known non-naturally occurring amino acid residues. Alterations such as these may serve to increase the stability, bioavailability and/or binding action of the peptides of the invention. Similarly, a peptide or variant of the invention may be modified chemically by reacting specific amino acids either before or after synthesis of the peptide. Examples for such modifications are well known in the art and are summarized e.g. in R. Lundblad, Chemical Reagents for Protein Modification, 3rd ed. CRC Press, 2004 (Lundblad, 2004), which is incorporated herein by reference. Chemical modification of amino acids includes but is not limited to, modification by acylation, amidination, pyridoxylation of lysine, reductive alkylation, trinitrobenzylation of amino groups with 2,4,6-trinitrobenzene sulphonic acid (TNBS), amide modification of carboxyl groups and sulphydryl modification by performic acid oxidation of cysteine to cysteic acid, formation of mercurial derivatives, formation of mixed disulphides with other thiol compounds, reaction with maleimide, carboxymethylation with iodoacetic acid or iodoacetamide and carbamoylation with cyanate at alkaline pH, although without limitation thereto. In this regard, the skilled person is referred to Chapter 15 of Current Protocols In Protein Science, Eds. Coligan et al. (John Wiley and Sons NY 1995-2000) (Coligan et al., 1995) for more extensive methodology relating to chemical modification of proteins. Briefly, modification of e.g. arginyl residues in proteins is often based on the reaction of vicinal dicarbonyl compounds such as phenylglyoxal, 2,3-butanedione, and 1,2-cyclohexanedione to form an adduct. Another example is the reaction of methylglyoxal with arginine residues. Cysteine can be modified without concomitant modification of other nucleophilic sites such as lysine and histidine. As a result, a large number of reagents are available for the modification of cysteine. The websites of companies such as Sigma-Aldrich provide information on specific reagents. Selective reduction of disulfide bonds in proteins is also common. Disulfide bonds can be formed and oxidized during the heat treatment of biopharmaceuticals. Woodward's Reagent K may be used to modify specific glutamic acid residues. N-(3-(dimethylamino)propyl)-N′-ethylcarbodiimide can be used to form intra-molecular crosslinks between a lysine residue and a glutamic acid residue. For example, diethylpyrocarbonate is a reagent for the modification of histidyl residues in proteins. Histidine can also be modified using 4-hydroxy-2-nonenal. The reaction of lysine residues and other α-amino groups is, for example, useful in binding of peptides to surfaces or the cross-linking of proteins/peptides. Lysine is the site of attachment of poly(ethylene)glycol and the major site of modification in the glycosylation of proteins. Methionine residues in proteins can be modified with e.g. iodoacetamide, bromoethylamine, and chloramine T. Tetranitromethane and N-acetylimidazole can be used for the modification of tyrosyl residues. Cross-linking via the formation of dityrosine can be accomplished with hydrogen peroxide/copper ions. Recent studies on the modification of tryptophan have used N-bromosuccinimide, 2-hydroxy-5-nitrobenzyl bromide or 3-bromo-3-methyl-2-(2-nitrophenylmercapto)-3H-indole (BPNS-skatole). Successful modification of therapeutic proteins and peptides with PEG is often associated with an extension of circulatory half-life while cross-linking of proteins with glutaraldehyde, polyethylene glycol diacrylate and formaldehyde is used for the preparation of hydrogels. Chemical modification of allergens for immunotherapy is often achieved by carbamylation with potassium cyanate. A peptide or variant, wherein the peptide is modified or includes non-peptide bonds is a preferred embodiment of the invention. Another embodiment of the present invention relates to a non-naturally occurring peptide wherein said peptide consists or consists essentially of an amino acid sequence according to SEQ ID No: 1 to SEQ ID No: 772 and has been synthetically produced (e.g. synthesized) as a pharmaceutically acceptable salt. Methods to synthetically produce peptides are well known in the art. The salts of the peptides according to the present invention differ substantially from the peptides in their state(s) in vivo, as the peptides as generated in vivo are no salts. The non-natural salt form of the peptide mediates the solubility of the peptide, in particular in the context of pharmaceutical compositions comprising the peptides, e.g. the peptide vaccines as disclosed herein. A sufficient and at least substantial solubility of the peptide(s) is required in order to efficiently provide the peptides to the subject to be treated. Preferably, the salts are pharmaceutically acceptable salts of the peptides. These salts according to the invention include alkaline and earth alkaline salts such as salts of the Hofmeister series comprising as anions PO43−, SO42−, CH3COO−, Cl−, Br−, NO3−, ClO4−, I−, SCN−and as cations NH4+, Rb+, K+, Na+, Cs+, Li+, Zn2+, Mg2+, Ca2+, Mn2+, Cu2+and Ba2+. Particularly salts are selected from (NH4)3PO4, (NH4)2HPO4, (NH4)H2PO4, (NH4)2SO4, NH4CH3COO, NH4Cl, NH4Br, NH4NO3, NH4ClO4, NH4I, NH4SCN, Rb3PO4, Rb2HPO4, RbH2PO4, Rb2SO4, Rb4CH3COO, Rb4Cl, Rb4Br, Rb4NO3, Rb4ClO4, Rb4I, Rb4SCN, K3PO4, K2HPO4, KH2PO4, K2SO4, KCH3COO, KCl, KBr, KNOB, KClO4, KI, KSCN, Na3PO4, Na2HPO4, NaH2PO4, Na2SO4, NaCH3COO, NaCl, NaBr, NaNO3, NaClO4, NaI, NaSCN, ZnCl2, Cs3PO4, Cs2HPO4, CsH2PO4, Cs2SO4, CsCH3COO, CsCl, CsBr, CsNO3, CsClO4, CsI, CsSCN, Li3PO4, Li2HPO4, LiH2PO4, Li2SO4, LiCH3COO, LiCl, LiBr, LiNO3, LiClO4, LiI, LiSCN, Cu2SO4, Mg3(PO4)2, Mg2HPO4, Mg(H2PO4)2, Mg2SO4, Mg(CH3COO)2, MgCl2, MgBr2, Mg(NO3)2, Mg(ClO4)2, MgI2, Mg(SCN)2, MnCl2, Ca3(PO4), Ca2HPO4, Ca(H2PO4)2, CaSO4, Ca(CH3COO)2, CaCl2), CaBr2, Ca(NO3)2, Ca(ClO4)2, CaI2, Ca(SCN)2, Ba3(PO4)2, Ba2HPO4, Ba(H2PO4)2, BaSO4, Ba(CH3COO)2, BaCl2, BaBr2, Ba(NO3)2, Ba(ClO4)2, Bale, and Ba(SCN)2. Particularly preferred are NH acetate, MgCl2, KH2PO4, Na2SO4, KCl, NaCl, and CaCl2), such as, for example, the chloride or acetate (trifluoroacetate) salts. Generally, peptides and variants (at least those containing peptide linkages between amino acid residues) may be synthesized by the Fmoc-polyamide mode of solid-phase peptide synthesis as disclosed by Lukas et al. (Lukas et al., 1981) and by references as cited therein. Temporary N-amino group protection is afforded by the 9-fluorenylmethyloxycarbonyl (Fmoc) group. Repetitive cleavage of this highly base-labile protecting group is done using 20% piperidine in N, N-dimethylformamide. Side-chain functionalities may be protected as their butyl ethers (in the case of serine threonine and tyrosine), butyl esters (in the case of glutamic acid and aspartic acid), butyloxycarbonyl derivative (in the case of lysine and histidine), trityl derivative (in the case of cysteine) and 4-methoxy-2,3,6-trimethylbenzenesulphonyl derivative (in the case of arginine). Where glutamine or asparagine are C-terminal residues, use is made of the 4,4′-dimethoxybenzhydryl group for protection of the side chain amido functionalities. The solid-phase support is based on a polydimethyl-acrylamide polymer constituted from the three monomers dimethylacrylamide (backbone-monomer), bisacryloylethylene diamine (cross linker) and acryloylsarcosine methyl ester (functionalizing agent). The peptide-to-resin cleavable linked agent used is the acid-labile 4-hydroxymethyl-phenoxyacetic acid derivative. All amino acid derivatives are added as their preformed symmetrical anhydride derivatives with the exception of asparagine and glutamine, which are added using a reversed N, N-dicyclohexyl-carbodiimide/1hydroxybenzotriazole mediated coupling procedure. All coupling and deprotection reactions are monitored using ninhydrin, trinitrobenzene sulphonic acid or isotin test procedures. Upon completion of synthesis, peptides are cleaved from the resin support with concomitant removal of side-chain protecting groups by treatment with 95% trifluoroacetic acid containing a 50% scavenger mix. Scavengers commonly used include ethanedithiol, phenol, anisole and water, the exact choice depending on the constituent amino acids of the peptide being synthesized. Also a combination of solid phase and solution phase methodologies for the synthesis of peptides is possible (see, for example, (Bruckdorfer et al., 2004), and the references as cited therein). Trifluoroacetic acid is removed by evaporation in vacuo, with subsequent trituration with diethyl ether affording the crude peptide. Any scavengers present are removed by a simple extraction procedure which on lyophilization of the aqueous phase affords the crude peptide free of scavengers. Reagents for peptide synthesis are generally available from e.g. Calbiochem-Novabiochem (Nottingham, UK). Purification may be performed by any one, or a combination of, techniques such as re-crystallization, size exclusion chromatography, ion-exchange chromatography, hydrophobic interaction chromatography and (usually) reverse-phase high performance liquid chromatography using e.g. acetonitrile/water gradient separation. Analysis of peptides may be carried out using thin layer chromatography, electrophoresis, in particular capillary electrophoresis, solid phase extraction (CSPE), reverse-phase high performance liquid chromatography, amino-acid analysis after acid hydrolysis and by fast atom bombardment (FAB) mass spectrometric analysis, as well as MALDI and ESI-Q-TOF mass spectrometric analysis. For the identification of HLA ligands by mass spectrometry, HLA molecules from shock-frozen tissue samples were purified and HLA-associated peptides were isolated. The isolated peptides were separated and sequences were identified by online nano-electrospray-ionization (nanoESI) liquid chromatography-mass spectrometry (LC-MS) experiments. The resulting peptide sequences were verified by comparison of the fragmentation pattern of natural tumor-associated peptides (TUMAPs) recorded from ovarian cancer samples (N≥80 samples) with the fragmentation patterns of corresponding synthetic reference peptides of identical sequences. Since the peptides were directly identified as ligands of HLA molecules of primary tumors, these results provide direct evidence for the natural processing and presentation of the identified peptides on primary cancer tissue obtained from ≥80 ovarian cancer patients (cf. Example 1). The discovery pipeline XPRESIDENT® v2.1 (see, for example, US 2013-0096016, which is hereby incorporated by reference in its entirety) allows the identification and selection of relevant over-presented peptide vaccine candidates based on direct relative quantitation of HLA-restricted peptide levels on cancer tissues in comparison to several different non-cancerous tissues and organs. This was achieved by the development of label-free differential quantitation using the acquired LC-MS data processed by a proprietary data analysis pipeline, combining algorithms for sequence identification, spectral clustering, ion counting, retention time alignment, charge state deconvolution and normalization. HLA-peptide complexes from ovarian cancer tissue samples were purified and HLA-associated peptides were isolated and analyzed by LC-MS (see example 1). All TUMAPs contained in the present application were identified with this approach on primary ovarian cancer samples confirming their presentation on primary ovarian cancer. Besides presentation of the peptide, mRNA expression of the underlying gene was tested. mRNA data were obtained via RNASeq analyses of normal tissues and cancer tissues (cf. Example 2,FIGS.1A-1V). Peptides which are derived from proteins whose coding mRNA is highly expressed in cancer tissue, but very low or absent in vital normal tissues, were preferably included in the present invention. The present invention provides peptides that are useful in treating cancers/tumors, preferably ovarian cancer that over- or exclusively present the peptides of the invention. These peptides were shown by mass spectrometry to be naturally presented by HLA molecules on primary human ovarian cancer samples. Many of the source gene/proteins (also designated “full-length proteins” or “underlying proteins”) from which the peptides are derived were shown to be highly over-expressed in cancer compared with normal tissues—“normal tissues” in relation to this invention shall mean either healthy ovarian cells or other normal tissue cells, demonstrating a high degree of tumor association of the source genes (see Example 2). Moreover, the peptides themselves are presented on tumor tissue—“tumor tissue” in relation to this invention shall mean a sample from a patient suffering from ovarian cancer. HLA-bound peptides can be recognized by the immune system, specifically T lymphocytes. T cells can destroy the cells presenting the recognized HLA/peptide complex, e.g. ovarian cancer cells presenting the derived peptides. The peptides of the present invention have been shown to be capable of stimulating T cell responses and/or are over-presented and thus can be used for the production of antibodies and/or TCRs, such as soluble TCRs, according to the present invention (see Example 3, Example 4). Furthermore, the peptides when complexed with the respective MHC can be used for the production of antibodies and/or TCRs, in particular sTCRs, according to the present invention, as well. Respective methods are well known to the person of skill, and can be found in the respective literature as well (see also below). Thus, the peptides of the present invention are useful for generating an immune response in a patient by which tumor cells can be destroyed. An immune response in a patient can be induced by direct administration of the described peptides or suitable precursor substances (e.g. elongated peptides, proteins, or nucleic acids encoding these peptides) to the patient, ideally in combination with an agent enhancing the immunogenicity (i.e. an adjuvant). The immune response originating from such a therapeutic vaccination can be expected to be highly specific against tumor cells because the target peptides of the present invention are not presented on normal tissues in comparable copy numbers, preventing the risk of undesired autoimmune reactions against normal cells in the patient. The present description further relates to T-cell receptors (TCRs) comprising an alpha chain and a beta chain (“alpha/beta TCRs”). Also provided are peptides according to the invention capable of binding to TCRs and antibodies when presented by an MHC molecule. The present description also relates to fragments of the TCRs according to the invention that are capable of binding to a peptide antigen according to the present invention when presented by an HLA molecule. The term particularly relates to soluble TCR fragments, for example TCRs missing the transmembrane parts and/or constant regions, single chain TCRs, and fusions thereof to, for example, with Ig. The present description also relates to nucleic acids, vectors and host cells for expressing TCRs and peptides of the present description; and methods of using the same. The term “T-cell receptor” (abbreviated TCR) refers to a heterodimeric molecule comprising an alpha polypeptide chain (alpha chain) and a beta polypeptide chain (beta chain), wherein the heterodimeric receptor is capable of binding to a peptide antigen presented by an HLA molecule. The term also includes so-called gamma/delta TCRs. In one embodiment the description provides a method of producing a TCR as described herein, the method comprising culturing a host cell capable of expressing the TCR under conditions suitable to promote expression of the TCR. The description in another aspect relates to methods according to the description, wherein the antigen is loaded onto class I or II MHC molecules expressed on the surface of a suitable antigen-presenting cell or artificial antigen-presenting cell by contacting a sufficient amount of the antigen with an antigen-presenting cell or the antigen is loaded onto class I or II MHC tetramers by tetramerizing the antigen/class I or II MHC complex monomers. The alpha and beta chains of alpha/beta TCR's, and the gamma and delta chains of gamma/delta TCRs, are generally regarded as each having two “domains”, namely variable and constant domains. The variable domain consists of a concatenation of variable region (V), and joining region (J). The variable domain may also include a leader region (L). Beta and delta chains may also include a diversity region (D). The alpha and beta constant domains may also include C-terminal transmembrane (TM) domains that anchor the alpha and beta chains to the cell membrane. With respect to gamma/delta TCRs, the term “TCR gamma variable domain” as used herein refers to the concatenation of the TCR gamma V (TRGV) region without leader region (L), and the TCR gamma J (TRGJ) region, and the term TCR gamma constant domain refers to the extracellular TRGC region, or to a C-terminal truncated TRGC sequence. Likewise the term “TCR delta variable domain” refers to the concatenation of the TCR delta V (TRDV) region without leader region (L) and the TCR delta D/J (TRDD/TRDJ) region, and the term “TCR delta constant domain” refers to the extracellular TRDC region, or to a C-terminal truncated TRDC sequence. TCRs of the present description preferably bind to an peptide-HLA molecule complex with a binding affinity (KD) of about 100 μM or less, about 50 μM or less, about 25 μM or less, or about 10 μM or less. More preferred are high affinity TCRs having binding affinities of about 1 μM or less, about 100 nM or less, about 50 nM or less, about 25 nM or less. Non-limiting examples of preferred binding affinity ranges for TCRs of the present invention include about 1 nM to about 10 nM; about 10 nM to about 20 nM; about 20 nM to about 30 nM; about 30 nM to about 40 nM; about 40 nM to about 50 nM; about 50 nM to about 60 nM; about 60 nM to about 70 nM; about 70 nM to about 80 nM; about 80 nM to about 90 nM; and about 90 nM to about 100 nM. As used herein in connect with TCRs of the present description, “specific binding” and grammatical variants thereof are used to mean a TCR having a binding affinity (KD) for a peptide-HLA molecule complex of 100 μM or less. Alpha/beta heterodimeric TCRs of the present description may have an introduced disulfide bond between their constant domains. Preferred TCRs of this type include those which have a TRAC constant domain sequence and a TRBC1 or TRBC2 constant domain sequence except that Thr 48 of TRAC and Ser 57 of TRBC1 or TRBC2 are replaced by cysteine residues, the said cysteines forming a disulfide bond between the TRAC constant domain sequence and the TRBC1 or TRBC2 constant domain sequence of the TCR. With or without the introduced inter-chain bond mentioned above, alpha/beta heterodimeric TCRs of the present description may have a TRAC constant domain sequence and a TRBC1 or TRBC2 constant domain sequence, and the TRAC constant domain sequence and the TRBC1 or TRBC2 constant domain sequence of the TCR may be linked by the native disulfide bond between Cys4 of exon 2 of TRAC and Cys2 of exon 2 of TRBC1 or TRBC2. TCRs of the present description may comprise a detectable label selected from the group consisting of a radionuclide, a fluorophore and biotin. TCRs of the present description may be conjugated to a therapeutically active agent, such as a radionuclide, a chemotherapeutic agent, or a toxin. In an embodiment, a TCR of the present description having at least one mutation in the alpha chain and/or having at least one mutation in the beta chain has modified glycosylation compared to the unmutated TCR. In an embodiment, a TCR comprising at least one mutation in the TCR alpha chain and/or TCR beta chain has a binding affinity for, and/or a binding half-life for, a peptide-HLA molecule complex, which is at least double that of a TCR comprising the unmutated TCR alpha chain and/or unmutated TCR beta chain. Affinity-enhancement of tumor-specific TCRs, and its exploitation, relies on the existence of a window for optimal TCR affinities. The existence of such a window is based on observations that TCRs specific for HLA-A2-restricted pathogens have KD values that are generally about 10-fold lower when compared to TCRs specific for HLA-A2-restricted tumor-associated self-antigens. It is now known, although tumor antigens have the potential to be immunogenic, because tumors arise from the individual's own cells only mutated proteins or proteins with altered translational processing will be seen as foreign by the immune system. Antigens that are upregulated or overexpressed (so called self-antigens) will not necessarily induce a functional immune response against the tumor: T-cells expressing TCRs that are highly reactive to these antigens will have been negatively selected within the thymus in a process known as central tolerance, meaning that only T-cells with low-affinity TCRs for self-antigens remain. Therefore, affinity of TCRs or variants of the present description to peptides can be enhanced by methods well known in the art. The present description further relates to a method of identifying and isolating a TCR according to the present description, said method comprising incubating PBMCs from HLA-A*02-negative healthy donors with, for example, A2/peptide monomers, incubating the PBMCs with tetramer-phycoerythrin (PE) and isolating the high avidity T-cells by fluorescence activated cell sorting (FACS)-Calibur analysis. The present description further relates to a method of identifying and isolating a TCR according to the present description, said method comprising obtaining a transgenic mouse with the entire human TCRαβ gene loci (1.1 and 0.7 Mb), whose T-cells express a diverse human TCR repertoire that compensates for mouse TCR deficiency, immunizing the mouse with a peptide, incubating PBMCs obtained from the transgenic mice with tetramer-phycoerythrin (PE), and isolating the high avidity T-cells by fluorescence activated cell sorting (FACS)-Calibur analysis. In one aspect, to obtain T-cells expressing TCRs of the present description, nucleic acids encoding TCR-alpha and/or TCR-beta chains of the present description are cloned into expression vectors, such as gamma retrovirus or lentivirus. The recombinant viruses are generated and then tested for functionality, such as antigen specificity and functional avidity. An aliquot of the final product is then used to transduce the target T-cell population (generally purified from patient PBMCs), which is expanded before infusion into the patient. In another aspect, to obtain T-cells expressing TCRs of the present description, TCR RNAs are synthesized by techniques known in the art, e.g., in vitro transcription systems. The in vitro-synthesized TCR RNAs are then introduced into primary CD8+ T-cells obtained from healthy donors by electroporation to re-express tumor specific TCR-alpha and/or TCR-beta chains. To increase the expression, nucleic acids encoding TCRs of the present description may be operably linked to strong promoters, such as retroviral long terminal repeats (LTRs), cytomegalovirus (CMV), murine stem cell virus (MSCV) U3, phosphoglycerate kinase (PGK), β-actin, ubiquitin, and a simian virus 40 (SV40)/CD43 composite promoter, elongation factor (EF)-1a and the spleen focus-forming virus (SFFV) promoter. In a preferred embodiment, the promoter is heterologous to the nucleic acid being expressed. In addition to strong promoters, TCR expression cassettes of the present description may contain additional elements that can enhance transgene expression, including a central polypurine tract (cPPT), which promotes the nuclear translocation of lentiviral constructs (Follenzi et al., 2000), and the woodchuck hepatitis virus posttranscriptional regulatory element (wPRE), which increases the level of transgene expression by increasing RNA stability (Zufferey et al., 1999). The alpha and beta chains of a TCR of the present invention may be encoded by nucleic acids located in separate vectors, or may be encoded by polynucleotides located in the same vector. Achieving high-level TCR surface expression requires that both the TCR-alpha and TCR-beta chains of the introduced TCR be transcribed at high levels. To do so, the TCR-alpha and TCR-beta chains of the present description may be cloned into bi-cistronic constructs in a single vector, which has been shown to be capable of over-coming this obstacle. The use of a viral intraribosomal entry site (IRES) between the TCR-alpha and TCR-beta chains results in the coordinated expression of both chains, because the TCR-alpha and TCR-beta chains are generated from a single transcript that is broken into two proteins during translation, ensuring that an equal molar ratio of TCR-alpha and TCR-beta chains are produced (Schmitt et al., 2009). Nucleic acids encoding TCRs of the present description may be codon optimized to increase expression from a host cell. Redundancy in the genetic code allows some amino acids to be encoded by more than one codon, but certain codons are less “op-timal” than others because of the relative availability of matching tRNAs as well as other factors (Gustafsson et al., 2004). Modifying the TCR-alpha and TCR-beta gene sequences such that each amino acid is encoded by the optimal codon for mammalian gene expression, as well as eliminating mRNA instability motifs or cryptic splice sites, has been shown to significantly enhance TCR-alpha and TCR-beta gene expression (Scholten et al., 2006). Furthermore, mispairing between the introduced and endogenous TCR chains may result in the acquisition of specificities that pose a significant risk for autoimmunity. For example, the formation of mixed TCR dimers may reduce the number of CD3 molecules available to form properly paired TCR complexes, and therefore can significantly decrease the functional avidity of the cells expressing the introduced TCR (Kuball et al., 2007). To reduce mispairing, the C-terminus domain of the introduced TCR chains of the present description may be modified in order to promote interchain affinity, while de-creasing the ability of the introduced chains to pair with the endogenous TCR. These strategies may include replacing the human TCR-alpha and TCR-beta C-terminus domains with their murine counterparts (murinized C-terminus domain); generating a second interchain disulfide bond in the C-terminus domain by introducing a second cysteine residue into both the TCR-alpha and TCR-beta chains of the introduced TCR (cysteine modification); swapping interacting residues in the TCR-alpha and TCR-beta chain C-terminus domains (“knob-in-hole”); and fusing the variable domains of the TCR-alpha and TCR-beta chains directly to CD3ζ (CD3ζ fusion) (Schmitt et al., 2009). In an embodiment, a host cell is engineered to express a TCR of the present description. In preferred embodiments, the host cell is a human T-cell or T-cell progenitor. In some embodiments the T-cell or T-cell progenitor is obtained from a cancer patient. In other embodiments the T-cell or T-cell progenitor is obtained from a healthy donor. Host cells of the present description can be allogeneic or autologous with respect to a patient to be treated. In one embodiment, the host is a gamma/delta T-cell transformed to express an alpha/beta TCR. A “pharmaceutical composition” is a composition suitable for administration to a human being in a medical setting. Preferably, a pharmaceutical composition is sterile and produced according to GMP guidelines. The pharmaceutical compositions comprise the peptides either in the free form or in the form of a pharmaceutically acceptable salt (see also above). As used herein, “a pharmaceutically acceptable salt” refers to a derivative of the disclosed peptides wherein the peptide is modified by making acid or base salts of the agent. For example, acid salts are prepared from the free base (typically wherein the neutral form of the drug has a neutral —NH2 group) involving reaction with a suitable acid. Suitable acids for preparing acid salts include both organic acids, e.g., acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methane sulfonic acid, ethane sulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like, as well as inorganic acids, e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid phosphoric acid and the like. Conversely, preparation of basic salts of acid moieties which may be present on a peptide are prepared using a pharmaceutically acceptable base such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, trimethylamine or the like. In an especially preferred embodiment, the pharmaceutical compositions comprise the peptides as salts of acetic acid (acetates), trifluoro acetates or hydrochloric acid (chlorides). Preferably, the medicament of the present invention is an immunotherapeutic such as a vaccine. It may be administered directly into the patient, into the affected organ or systemically i.d., i.m., s.c., i.p. and i.v., or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation of immune cells derived from the patient, which are then re-administered to the patient. If the nucleic acid is administered to cells in vitro, it may be useful for the cells to be transfected so as to co-express immune-stimulating cytokines, such as interleukin-2. The peptide may be substantially pure, or combined with an immune-stimulating adjuvant (see below) or used in combination with immune-stimulatory cytokines, or be administered with a suitable delivery system, for example liposomes. The peptide may also be conjugated to a suitable carrier such as keyhole limpet haemocyanin (KLH) or mannan (see WO 95/18145 and (Longenecker et al., 1993)). The peptide may also be tagged, may be a fusion protein, or may be a hybrid molecule. The peptides whose sequence is given in the present invention are expected to stimulate CD4 or CD8 T cells. However, stimulation of CD8 T cells is more efficient in the presence of help provided by CD4 T-helper cells. Thus, for MHC Class I epitopes that stimulate CD8 T cells the fusion partner or sections of a hybrid molecule suitably provide epitopes which stimulate CD4-positive T cells. CD4- and CD8-stimulating epitopes are well known in the art and include those identified in the present invention. In one aspect, the vaccine comprises at least one peptide having the amino acid sequence set forth SEQ ID No. 1 to SEQ ID No. 772, and at least one additional peptide, preferably two to 50, more preferably two to 25, even more preferably two to 20 and most preferably two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen or eighteen peptides. The peptide(s) may be derived from one or more specific TAAs and may bind to MHC class I molecules. A further aspect of the invention provides a nucleic acid (for example a polynucleotide) encoding a peptide or peptide variant of the invention. The polynucleotide may be, for example, DNA, cDNA, PNA, RNA or combinations thereof, either single- and/or double-stranded, or native or stabilized forms of polynucleotides, such as, for example, polynucleotides with a phosphorothioate backbone and it may or may not contain introns so long as it codes for the peptide. Of course, only peptides that contain naturally occurring amino acid residues joined by naturally occurring peptide bonds are encodable by a polynucleotide. A still further aspect of the invention provides an expression vector capable of expressing a polypeptide according to the invention. A variety of methods have been developed to link polynucleotides, especially DNA, to vectors for example via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary homopolymeric tails to form recombinant DNA molecules. Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors. Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc. New Haven, CT, USA. A desirable method of modifying the DNA encoding the polypeptide of the invention employs the polymerase chain reaction as disclosed by Saiki R K, et al. (Saiki et al., 1988). This method may be used for introducing the DNA into a suitable vector, for example by engineering in suitable restriction sites, or it may be used to modify the DNA in other useful ways as is known in the art. If viral vectors are used, pox- or adenovirus vectors are preferred. The DNA (or in the case of retroviral vectors, RNA) may then be expressed in a suitable host to produce a polypeptide comprising the peptide or variant of the invention. Thus, the DNA encoding the peptide or variant of the invention may be used in accordance with known techniques, appropriately modified in view of the teachings contained herein, to construct an expression vector, which is then used to transform an appropriate host cell for the expression and production of the polypeptide of the invention. Such techniques include those disclosed, for example, in U.S. Pat. Nos. 4,440,859, 4,530,901, 4,582,800, 4,677,063, 4,678,751, 4,704,362, 4,710,463, 4,757,006, 4,766,075, and 4,810,648. The DNA (or in the case of retroviral vectors, RNA) encoding the polypeptide constituting the compound of the invention may be joined to a wide variety of other DNA sequences for introduction into an appropriate host. The companion DNA will depend upon the nature of the host, the manner of the introduction of the DNA into the host, and whether episomal maintenance or integration is desired. Generally, the DNA is inserted into an expression vector, such as a plasmid, in proper orientation and correct reading frame for expression. If necessary, the DNA may be linked to the appropriate transcriptional and translational regulatory control nucleotide sequences recognized by the desired host, although such controls are generally available in the expression vector. The vector is then introduced into the host through standard techniques. Generally, not all of the hosts will be transformed by the vector. Therefore, it will be necessary to select for transformed host cells. One selection technique involves incorporating into the expression vector a DNA sequence, with any necessary control elements, that codes for a selectable trait in the transformed cell, such as antibiotic resistance. Alternatively, the gene for such selectable trait can be on another vector, which is used to co-transform the desired host cell. Host cells that have been transformed by the recombinant DNA of the invention are then cultured for a sufficient time and under appropriate conditions known to those skilled in the art in view of the teachings disclosed herein to permit the expression of the polypeptide, which can then be recovered. Many expression systems are known, including bacteria (for exampleE. coliandBacillus subtilis), yeasts (for exampleSaccharomyces cerevisiae), filamentous fungi (for exampleAspergillusspec.), plant cells, animal cells and insect cells. Preferably, the system can be mammalian cells such as CHO cells available from the ATCC Cell Biology Collection. A typical mammalian cell vector plasmid for constitutive expression comprises the CMV or SV40 promoter with a suitable poly A tail and a resistance marker, such as neomycin. One example is pSVL available from Pharmacia, Piscataway, NJ, USA. An example of an inducible mammalian expression vector is pMSG, also available from Pharmacia. Useful yeast plasmid vectors are pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA. Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (YIps) and incorporate the yeast selectable markers HIS3, TRP1, LEU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps). CMV promoter-based vectors (for example from Sigma-Aldrich) provide transient or stable expression, cytoplasmic expression or secretion, and N-terminal or C-terminal tagging in various combinations of FLAG, 3×FLAG, c-myc or MAT. These fusion proteins allow for detection, purification and analysis of recombinant protein. Dual-tagged fusions provide flexibility in detection. The strong human cytomegalovirus (CMV) promoter regulatory region drives constitutive protein expression levels as high as 1 mg/L in COS cells. For less potent cell lines, protein levels are typically ˜0.1 mg/L. The presence of the SV40 replication origin will result in high levels of DNA replication in SV40 replication permissive COS cells. CMV vectors, for example, can contain the pMB1 (derivative of pBR322) origin for replication in bacterial cells, the b-lactamase gene for ampicillin resistance selection in bacteria, hGH polyA, and the f1 origin. Vectors containing the pre-pro-trypsin leader (PPT) sequence can direct the secretion of FLAG fusion proteins into the culture medium for purification using ANTI-FLAG antibodies, resins, and plates. Other vectors and expression systems are well known in the art for use with a variety of host cells. In another embodiment two or more peptides or peptide variants of the invention are encoded and thus expressed in a successive order (similar to “beads on a string” constructs). In doing so, the peptides or peptide variants may be linked or fused together by stretches of linker amino acids, such as for example LLLLLL, or may be linked without any additional peptide(s) between them. These constructs can also be used for cancer therapy, and may induce immune responses both involving MHC I and MHC II. The present invention also relates to a host cell transformed with a polynucleotide vector construct of the present invention. The host cell can be either prokaryotic or eukaryotic. Bacterial cells may be preferred prokaryotic host cells in some circumstances and typically are a strain ofE. colisuch as, for example, theE. colistrains DH5 available from Bethesda Research Laboratories Inc., Bethesda, MD, USA, and RR1 available from the American Type Culture Collection (ATCC) of Rockville, MD, USA (No ATCC 31343). Preferred eukaryotic host cells include yeast, insect and mammalian cells, preferably vertebrate cells such as those from a mouse, rat, monkey or human fibroblastic and colon cell lines. Yeast host cells include YPH499, YPH500 and YPH501, which are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA. Preferred mammalian host cells include Chinese hamster ovary (CHO) cells available from the ATCC as CCL61, NIH Swiss mouse embryo cells NIH/3T3 available from the ATCC as CRL 1658, monkey kidney-derived COS-1 cells available from the ATCC as CRL 1650 and 293 cells which are human embryonic kidney cells. Preferred insect cells are Sf9 cells which can be transfected with baculovirus expression vectors. An overview regarding the choice of suitable host cells for expression can be found in, for example, the textbook of Paulina Balbás and Argelia Lorence “Methods in Molecular Biology Recombinant Gene Expression, Reviews and Protocols,” Part One, Second Edition, ISBN 978-1-58829-262-9, and other literature known to the person of skill. Transformation of appropriate cell hosts with a DNA construct of the present invention is accomplished by well-known methods that typically depend on the type of vector used. With regard to transformation of prokaryotic host cells, see, for example, Cohen et al. (Cohen et al., 1972) and (Green and Sambrook, 2012). Transformation of yeast cells is described in Sherman et al. (Sherman et al., 1986). The method of Beggs (Beggs, 1978) is also useful. With regard to vertebrate cells, reagents useful in transfecting such cells, for example calcium phosphate and DEAE-dextran or liposome formulations, are available from Stratagene Cloning Systems, or Life Technologies Inc., Gaithersburg, MD 20877, USA. Electroporation is also useful for transforming and/or transfecting cells and is well known in the art for transforming yeast cell, bacterial cells, insect cells and vertebrate cells. Successfully transformed cells, i.e. cells that contain a DNA construct of the present invention, can be identified by well-known techniques such as PCR. Alternatively, the presence of the protein in the supernatant can be detected using antibodies. It will be appreciated that certain host cells of the invention are useful in the preparation of the peptides of the invention, for example bacterial, yeast and insect cells. However, other host cells may be useful in certain therapeutic methods. For example, antigen-presenting cells, such as dendritic cells, may usefully be used to express the peptides of the invention such that they may be loaded into appropriate MHC molecules. Thus, the current invention provides a host cell comprising a nucleic acid or an expression vector according to the invention. In a preferred embodiment the host cell is an antigen presenting cell, in particular a dendritic cell or antigen presenting cell. APCs loaded with a recombinant fusion protein containing prostatic acid phosphatase (PAP) were approved by the U.S. Food and Drug Administration (FDA) on Apr. 29, 2010, to treat asymptomatic or minimally symptomatic metastatic HRPC (Sipuleucel-T) (Rini et al., 2006; Small et al., 2006). A further aspect of the invention provides a method of producing a peptide or its variant, the method comprising culturing a host cell and isolating the peptide from the host cell or its culture medium. In another embodiment, the peptide, the nucleic acid or the expression vector of the invention are used in medicine. For example, the peptide or its variant may be prepared for intravenous (i.v.) injection, sub-cutaneous (s.c.) injection, intradermal (i.d.) injection, intraperitoneal (i.p.) injection, intramuscular (i.m.) injection. Preferred methods of peptide injection include s.c., i.d., i.p., i.m., and i.v. Preferred methods of DNA injection include i.d., i.m., s.c., i.p. and i.v. Doses of e.g. between 50 μg and 1.5 mg, preferably 125 μg to 500 μg, of peptide or DNA may be given and will depend on the respective peptide or DNA. Dosages of this range were successfully used in previous trials (Walter et al., 2012). The polynucleotide used for active vaccination may be substantially pure, or contained in a suitable vector or delivery system. The nucleic acid may be DNA, cDNA, PNA, RNA or a combination thereof. Methods for designing and introducing such a nucleic acid are well known in the art. An overview is provided by e.g. Teufel et al. (Teufel et al., 2005). Polynucleotide vaccines are easy to prepare, but the mode of action of these vectors in inducing an immune response is not fully understood. Suitable vectors and delivery systems include viral DNA and/or RNA, such as systems based on adenovirus, vaccinia virus, retroviruses, herpes virus, adeno-associated virus or hybrids containing elements of more than one virus. Non-viral delivery systems include cationic lipids and cationic polymers and are well known in the art of DNA delivery. Physical delivery, such as via a “gene-gun” may also be used. The peptide or peptides encoded by the nucleic acid may be a fusion protein, for example with an epitope that stimulates T cells for the respective opposite CDR as noted above. The medicament of the invention may also include one or more adjuvants. Adjuvants are substances that non-specifically enhance or potentiate the immune response (e.g., immune responses mediated by CD8-positive T cells and helper-T (TH) cells to an antigen, and would thus be considered useful in the medicament of the present invention. Suitable adjuvants include, but are not limited to, 1018 ISS, aluminum salts, AMPLIVAX®, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, flagellin or TLR5 ligands derived from flagellin, FLT3 ligand, GM-CSF, IC30, IC31, Imiquimod (ALDARA®), resiquimod, ImuFact IMP321, Interleukins as IL-2, IL-13, IL-21, Interferon-alpha or -beta, or pegylated derivatives thereof, IS Patch, ISS, ISCOMATRIX, ISCOMs, JuvImmune®, LipoVac, MALP2, MF59, monophosphoryl lipid A, Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, water-in-oil and oil-in-water emulsions, OK-432, OM-174, OM-197-MP-EC, ONTAK, OspA, PepTel® vector system, poly(lactid co-glycolid) [PLG]-based and dextran microparticles, talactoferrin SRL172, Virosomes and other Virus-like particles, YF-17D, VEGF trap, R848, beta-glucan, Pam3Cys, Aquila's QS21 stimulon, which is derived from saponin, mycobacterial extracts and synthetic bacterial cell wall mimics, and other proprietary adjuvants such as Ribi's Detox, Quil, or Superfos. Adjuvants such as Freund's or GM-CSF are preferred. Several immunological adjuvants (e.g., MF59) specific for dendritic cells and their preparation have been described previously (Allison and Krummel, 1995). Also cytokines may be used. Several cytokines have been directly linked to influencing dendritic cell migration to lymphoid tissues (e.g., TNF-), accelerating the maturation of dendritic cells into efficient antigen-presenting cells for T-lymphocytes (e.g., GM-CSF, IL-1 and IL-4) (U.S. Pat. No. 5,849,589, specifically incorporated herein by reference in its entirety) and acting as immunoadjuvants (e.g., IL-12, IL-15, IL-23, IL-7, IFN-alpha. IFN-beta) (Gabrilovich et al., 1996). CpG immunostimulatory oligonucleotides have also been reported to enhance the effects of adjuvants in a vaccine setting. Without being bound by theory, CpG oligonucleotides act by activating the innate (non-adaptive) immune system via Toll-like receptors (TLR), mainly TLR9. CpG triggered TLR9 activation enhances antigen-specific humoral and cellular responses to a wide variety of antigens, including peptide or protein antigens, live or killed viruses, dendritic cell vaccines, autologous cellular vaccines and polysaccharide conjugates in both prophylactic and therapeutic vaccines. More importantly it enhances dendritic cell maturation and differentiation, resulting in enhanced activation of TH1 cells and strong cytotoxic T-lymphocyte (CTL) generation, even in the absence of CD4 T cell help. The TH1 bias induced by TLR9 stimulation is maintained even in the presence of vaccine adjuvants such as alum or incomplete Freund's adjuvant (IFA) that normally promote a TH2 bias. CpG oligonucleotides show even greater adjuvant activity when formulated or co-administered with other adjuvants or in formulations such as microparticles, nanoparticles, lipid emulsions or similar formulations, which are especially necessary for inducing a strong response when the antigen is relatively weak. They also accelerate the immune response and enable the antigen doses to be reduced by approximately two orders of magnitude, with comparable antibody responses to the full-dose vaccine without CpG in some experiments (Krieg, 2006). U.S. Pat. No. 6,406,705 B1 describes the combined use of CpG oligonucleotides, non-nucleic acid adjuvants and an antigen to induce an antigen-specific immune response. A CpG TLR9 antagonist is dSLIM (double Stem Loop Immunomodulator) by Mologen (Berlin, Germany) which is a preferred component of the pharmaceutical composition of the present invention. Other TLR binding molecules such as RNA binding TLR 7, TLR 8 and/or TLR 9 may also be used. Other examples for useful adjuvants include, but are not limited to chemically modified CpGs (e.g. CpR, Idera), dsRNA analogues such as Poly(I:C) and derivates thereof (e.g. AmpliGen®, Hiltonol®, poly-(ICLC), poly(IC-R), poly(I:C12U), non-CpG bacterial DNA or RNA as well as immunoactive small molecules and antibodies such as cyclophosphamide, sunitinib, Bevacizumab®, celebrex, NCX-4016, sildenafil, tadalafil, vardenafil, sorafenib, temozolomide, temsirolimus, XL-999, CP-547632, pazopanib, VEGF Trap, ZD2171, AZD2171, anti-CTLA4, other antibodies targeting key structures of the immune system (e.g. anti-CD40, anti-TGFbeta, anti-TNFalpha receptor) and SC58175, which may act therapeutically and/or as an adjuvant. The amounts and concentrations of adjuvants and additives useful in the context of the present invention can readily be determined by the skilled artisan without undue experimentation. Preferred adjuvants are anti-CD40, imiquimod, resiquimod, GM-CSF, cyclophosphamide, sunitinib, bevacizumab, interferon-alpha, CpG oligonucleotides and derivates, poly-(I:C) and derivates, RNA, sildenafil, and particulate formulations with PLG or virosomes. In a preferred embodiment, the pharmaceutical composition according to the invention the adjuvant is selected from the group consisting of colony-stimulating factors, such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim), cyclophosphamide, imiquimod, resiquimod, and interferon-alpha. In a preferred embodiment, the pharmaceutical composition according to the invention the adjuvant is selected from the group consisting of colony-stimulating factors, such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim), cyclophosphamide, imiquimod and resiquimod. In a preferred embodiment of the pharmaceutical composition according to the invention, the adjuvant is cyclophosphamide, imiquimod or resiquimod. Even more preferred adjuvants are Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, poly-ICLC (Hiltonol®) and anti-CD40 mAB, or combinations thereof. This composition is used for parenteral administration, such as subcutaneous, intradermal, intramuscular or oral administration. For this, the peptides and optionally other molecules are dissolved or suspended in a pharmaceutically acceptable, preferably aqueous carrier. In addition, the composition can contain excipients, such as buffers, binding agents, blasting agents, diluents, flavors, lubricants, etc. The peptides can also be administered together with immune stimulating substances, such as cytokines. An extensive listing of excipients that can be used in such a composition, can be, for example, taken from A. Kibbe, Handbook of Pharmaceutical Excipients (Kibbe, 2000). The composition can be used for a prevention, prophylaxis and/or therapy of adenomatous or cancerous diseases. Exemplary formulations can be found in, for example, EP2112253. It is important to realize that the immune response triggered by the vaccine according to the invention attacks the cancer in different cell-stages and different stages of development. Furthermore different cancer associated signaling pathways are attacked. This is an advantage over vaccines that address only one or few targets, which may cause the tumor to easily adapt to the attack (tumor escape). Furthermore, not all individual tumors express the same pattern of antigens. Therefore, a combination of several tumor-associated peptides ensures that every single tumor bears at least some of the targets. The composition is designed in such a way that each tumor is expected to express several of the antigens and cover several independent pathways necessary for tumor growth and maintenance. Thus, the vaccine can easily be used “off-the-shelf” for a larger patient population. This means that a pre-selection of patients to be treated with the vaccine can be restricted to HLA typing, does not require any additional biomarker assessments for antigen expression, but it is still ensured that several targets are simultaneously attacked by the induced immune response, which is important for efficacy (Banchereau et al., 2001; Walter et al., 2012). As used herein, the term “scaffold” refers to a molecule that specifically binds to an (e.g. antigenic) determinant. In one embodiment, a scaffold is able to direct the entity to which it is attached (e.g. a (second) antigen binding moiety) to a target site, for example to a specific type of tumor cell or tumor stroma bearing the antigenic determinant (e.g. the complex of a peptide with MHC, according to the application at hand). In another embodiment a scaffold is able to activate signaling through its target antigen, for example a T cell receptor complex antigen. Scaffolds include but are not limited to antibodies and fragments thereof, antigen binding domains of an antibody, comprising an antibody heavy chain variable region and an antibody light chain variable region, binding proteins comprising at least one ankyrin repeat motif and single domain antigen binding (SDAB) molecules, aptamers, (soluble) TCRs and (modified) cells such as allogenic or autologous T cells. To assess whether a molecule is a scaffold binding to a target, binding assays can be performed. “Specific” binding means that the scaffold binds the peptide-MHC-complex of interest better than other naturally occurring peptide-MHC-complexes, to an extent that a scaffold armed with an active molecule that is able to kill a cell bearing the specific target is not able to kill another cell without the specific target but presenting other peptide-MHC complex(es). Binding to other peptide-MHC complexes is irrelevant if the peptide of the cross-reactive peptide-MHC is not naturally occurring, i.e. not derived from the human HLA-peptidome. Tests to assess target cell killing are well known in the art. They should be performed using target cells (primary cells or cell lines) with unaltered peptide-MHC presentation, or cells loaded with peptides such that naturally occurring peptide-MHC levels are reached. Each scaffold can comprise a labelling which provides that the bound scaffold can be detected by determining the presence or absence of a signal provided by the label. For example, the scaffold can be labelled with a fluorescent dye or any other applicable cellular marker molecule. Such marker molecules are well known in the art. For example a fluorescence-labelling, for example provided by a fluorescence dye, can provide a visualization of the bound aptamer by fluorescence or laser scanning microscopy or flow cytometry. Each scaffold can be conjugated with a second active molecule such as for example IL-21, anti-CD3, and anti-CD28. For further information on polypeptide scaffolds see for example the background section of WO 2014/071978A1 and the references cited therein. The present invention further relates to aptamers. Aptamers (see for example WO 2014/191359 and the literature as cited therein) are short single-stranded nucleic acid molecules, which can fold into defined three-dimensional structures and recognize specific target structures. They have appeared to be suitable alternatives for developing targeted therapies. Aptamers have been shown to selectively bind to a variety of complex targets with high affinity and specificity. Aptamers recognizing cell surface located molecules have been identified within the past decade and provide means for developing diagnostic and therapeutic approaches. Since aptamers have been shown to possess almost no toxicity and immunogenicity they are promising candidates for biomedical applications. Indeed aptamers, for example prostate-specific membrane-antigen recognizing aptamers, have been successfully employed for targeted therapies and shown to be functional in xenograft in vivo models. Furthermore, aptamers recognizing specific tumor cell lines have been identified. DNA aptamers can be selected to reveal broad-spectrum recognition properties for various cancer cells, and particularly those derived from solid tumors, while non-tumorigenic and primary healthy cells are not recognized. If the identified aptamers recognize not only a specific tumor sub-type but rather interact with a series of tumors, this renders the aptamers applicable as so-called broad-spectrum diagnostics and therapeutics. Further, investigation of cell-binding behavior with flow cytometry showed that the aptamers revealed very good apparent affinities that are within the nanomolar range. Aptamers are useful for diagnostic and therapeutic purposes. Further, it could be shown that some of the aptamers are taken up by tumor cells and thus can function as molecular vehicles for the targeted delivery of anti-cancer agents such as siRNA into tumor cells. Aptamers can be selected against complex targets such as cells and tissues and complexes of the peptides comprising, preferably consisting of, a sequence according to any of SEQ ID NO 1 to SEQ ID NO 772, according to the invention at hand with the MHC molecule, using the cell-SELEX (Systematic Evolution of Ligands by Exponential enrichment) technique. The peptides of the present invention can be used to generate and develop specific antibodies against MHC/peptide complexes. These can be used for therapy, targeting toxins or radioactive substances to the diseased tissue. Another use of these antibodies can be targeting radionuclides to the diseased tissue for imaging purposes such as PET. This use can help to detect small metastases or to determine the size and precise localization of diseased tissues. Therefore, it is a further aspect of the invention to provide a method for producing a recombinant antibody specifically binding to a human major histocompatibility complex (MHC) class I or II being complexed with a HLA-restricted antigen (preferably a peptide according to the present invention), the method comprising: immunizing a genetically engineered non-human mammal comprising cells expressing said human major histocompatibility complex (MHC) class I or II with a soluble form of a MHC class I or II molecule being complexed with said HLA-restricted antigen; isolating mRNA molecules from antibody producing cells of said non-human mammal; producing a phage display library displaying protein molecules encoded by said mRNA molecules; and isolating at least one phage from said phage display library, said at least one phage displaying said antibody specifically binding to said human major histocompatibility complex (MHC) class I or II being complexed with said HLA-restricted antigen. It is thus a further aspect of the invention to provide an antibody that specifically binds to a human major histocompatibility complex (MHC) class I or II being complexed with a HLA-restricted antigen, wherein the antibody preferably is a polyclonal antibody, monoclonal antibody, bi-specific antibody and/or a chimeric antibody. Respective methods for producing such antibodies and single chain class I major histocompatibility complexes, as well as other tools for the production of these antibodies are disclosed in WO 03/068201, WO 2004/084798, WO 01/72768, WO 03/070752, and in publications (Cohen et al., 2003a; Cohen et al., 2003b; Denkberg et al., 2003), which for the purposes of the present invention are all explicitly incorporated by reference in their entireties. Preferably, the antibody is binding with a binding affinity of below 20 nanomolar, preferably of below 10 nanomolar, to the complex, which is also regarded as “specific” in the context of the present invention. The present invention relates to a peptide comprising a sequence that is selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 772, or a variant thereof which is at least 88% homologous (preferably identical) to SEQ ID NO: 1 to SEQ ID NO: 772 or a variant thereof that induces T cells cross-reacting with said peptide, wherein said peptide is not the underlying full-length polypeptide. The present invention further relates to a peptide comprising a sequence that is selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 772 or a variant thereof which is at least 88% homologous (preferably identical) to SEQ ID NO: 1 to SEQ ID NO: 772, wherein said peptide or variant has an overall length of between 8 and 100, preferably between 8 and 30, and most preferred between 8 and 14 amino acids. The present invention further relates to the peptides according to the invention that have the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I or -II. The present invention further relates to the peptides according to the invention wherein the peptide consists or consists essentially of an amino acid sequence according to SEQ ID NO: 1 to SEQ ID NO: 772. The present invention further relates to the peptides according to the invention, wherein the peptide is (chemically) modified and/or includes non-peptide bonds. The present invention further relates to the peptides according to the invention, wherein the peptide is part of a fusion protein, in particular comprising N-terminal amino acids of the HLA-DR antigen-associated invariant chain (Ii), or wherein the peptide is fused to (or into) an antibody, such as, for example, an antibody that is specific for dendritic cells. The present invention further relates to a nucleic acid, encoding the peptides according to the invention, provided that the peptide is not the complete (full) human protein. The present invention further relates to the nucleic acid according to the invention that is DNA, cDNA, PNA, RNA or combinations thereof. The present invention further relates to an expression vector capable of expressing a nucleic acid according to the present invention. The present invention further relates to a peptide according to the present invention, a nucleic acid according to the present invention or an expression vector according to the present invention for use in medicine, in particular in the treatment of ovarian cancer. The present invention further relates to a host cell comprising a nucleic acid according to the invention or an expression vector according to the invention. The present invention further relates to the host cell according to the present invention that is an antigen presenting cell, and preferably a dendritic cell. The present invention further relates to a method of producing a peptide according to the present invention, said method comprising culturing the host cell according to the present invention, and isolating the peptide from said host cell or its culture medium. The present invention further relates to the method according to the present invention, where-in the antigen is loaded onto class I or II MHC molecules expressed on the surface of a suitable antigen-presenting cell by contacting a sufficient amount of the antigen with an antigen-presenting cell. The present invention further relates to the method according to the invention, wherein the antigen-presenting cell comprises an expression vector capable of expressing said peptide containing SEQ ID NO: 1 to SEQ ID NO: 772 or said variant amino acid sequence. The present invention further relates to activated T cells, produced by the method according to the present invention, wherein said T cells selectively recognizes a cell which aberrantly expresses a polypeptide comprising an amino acid sequence according to the present invention. The present invention further relates to a method of killing target cells in a patient which target cells aberrantly express a polypeptide comprising any amino acid sequence according to the present invention, the method comprising administering to the patient an effective number of T cells as according to the present invention. The present invention further relates to the use of any peptide described, a nucleic acid according to the present invention, an expression vector according to the present invention, a cell according to the present invention, or an activated cytotoxic T lymphocyte according to the present invention as a medicament or in the manufacture of a medicament. The present invention further relates to a use according to the present invention, wherein the medicament is active against cancer. The present invention further relates to a use according to the invention, wherein the medicament is a vaccine. The present invention further relates to a use according to the invention, wherein the medicament is active against cancer. The present invention further relates to a use according to the invention, wherein said cancer cells are ovarian cancer cells or other solid or hematological tumor cells such as hepatocellular carcinoma, colorectal carcinoma, glioblastoma, gastric cancer, esophageal cancer, non-small cell lung cancer, small cell lung cancer, pancreatic cancer, renal cell carcinoma, prostate cancer, melanoma, breast cancer, chronic lymphocytic leukemia, Non-Hodgkin lymphoma, acute myeloid leukemia, gallbladder cancer and cholangiocarcinoma, urinary bladder cancer, uterine cancer, head and neck squamous cell carcinoma, mesothelioma. The present invention further relates to particular marker proteins and biomarkers based on the peptides according to the present invention, herein called “targets” that can be used in the diagnosis and/or prognosis of ovarian cancer. The present invention also relates to the use of these novel targets for cancer treatment. The term “antibody” or “antibodies” is used herein in a broad sense and includes both polyclonal and monoclonal antibodies. In addition to intact or “full” immunoglobulin molecules, also included in the term “antibodies” are fragments (e.g. CDRs, Fv, Fab and Fc fragments) or polymers of those immunoglobulin molecules and humanized versions of immunoglobulin molecules, as long as they exhibit any of the desired properties (e.g., specific binding of a ovarian cancer marker (poly)peptide, delivery of a toxin to a ovarian cancer cell expressing a cancer marker gene at an increased level, and/or inhibiting the activity of a ovarian cancer marker polypeptide) according to the invention. Whenever possible, the antibodies of the invention may be purchased from commercial sources. The antibodies of the invention may also be generated using well-known methods. The skilled artisan will understand that either full length ovarian cancer marker polypeptides or fragments thereof may be used to generate the antibodies of the invention. A polypeptide to be used for generating an antibody of the invention may be partially or fully purified from a natural source, or may be produced using recombinant DNA techniques. For example, a cDNA encoding a peptide according to the present invention, such as a peptide according to SEQ ID NO: 1 to SEQ ID NO: 772 polypeptide, or a variant or fragment thereof, can be expressed in prokaryotic cells (e.g., bacteria) or eukaryotic cells (e.g., yeast, insect, or mammalian cells), after which the recombinant protein can be purified and used to generate a monoclonal or polyclonal antibody preparation that specifically bind the ovarian cancer marker polypeptide used to generate the antibody according to the invention. One of skill in the art will realize that the generation of two or more different sets of monoclonal or polyclonal antibodies maximizes the likelihood of obtaining an antibody with the specificity and affinity required for its intended use (e.g., ELISA, immunohistochemistry, in vivo imaging, immunotoxin therapy). The antibodies are tested for their desired activity by known methods, in accordance with the purpose for which the antibodies are to be used (e.g., ELISA, immunohistochemistry, immunotherapy, etc.; for further guidance on the generation and testing of antibodies, see, e.g., Greenfield, 2014 (Greenfield, 2014)). For example, the antibodies may be tested in ELISA assays or, Western blots, immunohistochemical staining of formalin-fixed cancers or frozen tissue sections. After their initial in vitro characterization, antibodies intended for therapeutic or in vivo diagnostic use are tested according to known clinical testing methods. The term “monoclonal antibody” as used herein refers to an antibody obtained from a substantially homogeneous population of antibodies, i.e.; the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. The monoclonal antibodies herein specifically include “chimeric” antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired antagonistic activity (U.S. Pat. No. 4,816,567, which is hereby incorporated in its entirety). Monoclonal antibodies of the invention may be prepared using hybridoma methods. In a hybridoma method, a mouse or other appropriate host animal is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes may be immunized in vitro. The monoclonal antibodies may also be made by recombinant DNA methods, such as those described in U.S. Pat. No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). In vitro methods are also suitable for preparing monovalent antibodies. Digestion of antibodies to produce fragments thereof, particularly Fab fragments, can be accomplished using routine techniques known in the art. For instance, digestion can be performed using papain. Examples of papain digestion are described in WO 94/29348 and U.S. Pat. No. 4,342,566. Papain digestion of antibodies typically produces two identical antigen binding fragments, called Fab fragments, each with a single antigen binding site, and a residual Fc fragment. Pepsin treatment yields a F(ab′)2 fragment and a pFc′ fragment. The antibody fragments, whether attached to other sequences or not, can also include insertions, deletions, substitutions, or other selected modifications of particular regions or specific amino acids residues, provided the activity of the fragment is not significantly altered or impaired compared to the non-modified antibody or antibody fragment. These modifications can provide for some additional property, such as to remove/add amino acids capable of disulfide bonding, to increase its bio-longevity, to alter its secretory characteristics, etc. In any case, the antibody fragment must possess a bioactive property, such as binding activity, regulation of binding at the binding domain, etc. Functional or active regions of the antibody may be identified by mutagenesis of a specific region of the protein, followed by expression and testing of the expressed polypeptide. Such methods are readily apparent to a skilled practitioner in the art and can include site-specific mutagenesis of the nucleic acid encoding the antibody fragment. The antibodies of the invention may further comprise humanized antibodies or human antibodies. Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′ or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In some instances, Fv framework (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such “humanized” antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies. Transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production can be employed. For example, it has been described that the homozygous deletion of the antibody heavy chain joining region gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production. Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice will result in the production of human antibodies upon antigen challenge. Human antibodies can also be produced in phage display libraries. Antibodies of the invention are preferably administered to a subject in a pharmaceutically acceptable carrier. Typically, an appropriate amount of a pharmaceutically-acceptable salt is used in the formulation to render the formulation isotonic. Examples of the pharmaceutically-acceptable carrier include saline, Ringer's solution and dextrose solution. The pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5. Further carriers include sustained release preparations such as semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, liposomes or microparticles. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentration of antibody being administered. The antibodies can be administered to the subject, patient, or cell by injection (e.g., intravenous, intraperitoneal, subcutaneous, intramuscular), or by other methods such as infusion that ensure its delivery to the bloodstream in an effective form. The antibodies may also be administered by intratumoral or peritumoral routes, to exert local as well as systemic therapeutic effects. Local or intravenous injection is preferred. Effective dosages and schedules for administering the antibodies may be determined empirically, and making such determinations is within the skill in the art. Those skilled in the art will understand that the dosage of antibodies that must be administered will vary depending on, for example, the subject that will receive the antibody, the route of administration, the particular type of antibody used and other drugs being administered. A typical daily dosage of the antibody used alone might range from about 1 (μg/kg to up to 100 mg/kg of body weight or more per day, depending on the factors mentioned above. Following administration of an antibody, preferably for treating ovarian cancer, the efficacy of the therapeutic antibody can be assessed in various ways well known to the skilled practitioner. For instance, the size, number, and/or distribution of cancer in a subject receiving treatment may be monitored using standard tumor imaging techniques. A therapeutically-administered antibody that arrests tumor growth, results in tumor shrinkage, and/or prevents the development of new tumors, compared to the disease course that would occurs in the absence of antibody administration, is an efficacious antibody for treatment of cancer. It is a further aspect of the invention to provide a method for producing a soluble T-cell receptor (sTCR) recognizing a specific peptide-MHC complex. Such soluble T-cell receptors can be generated from specific T-cell clones, and their affinity can be increased by mutagenesis targeting the complementarity-determining regions. For the purpose of T-cell receptor selection, phage display can be used (US 2010/0113300, (Liddy et al., 2012)). For the purpose of stabilization of T-cell receptors during phage display and in case of practical use as drug, alpha and beta chain can be linked e.g. by non-native disulfide bonds, other covalent bonds (single-chain T-cell receptor), or by dimerization domains (Boulter et al., 2003; Card et al., 2004; Willcox et al., 1999). The T-cell receptor can be linked to toxins, drugs, cytokines (see, for example, US 2013/0115191), and domains recruiting effector cells such as an anti-CD3 domain, etc., in order to execute particular functions on target cells. Moreover, it could be expressed in T cells used for adoptive transfer. Further information can be found in WO 2004/033685A1 and WO 2004/074322A1. A combination of sTCRs is described in WO 2012/056407A1. Further methods for the production are disclosed in WO 2013/057586A1. In addition, the peptides and/or the TCRs or antibodies or other binding molecules of the present invention can be used to verify a pathologist's diagnosis of a cancer based on a biopsied sample. The antibodies or TCRs may also be used for in vivo diagnostic assays. Generally, the antibody is labeled with a radionucleotide (such as111In,99Tc,14C,131I,3H,32P or35S) so that the tumor can be localized using immunoscintiography. In one embodiment, antibodies or fragments thereof bind to the extracellular domains of two or more targets of a protein selected from the group consisting of the above-mentioned proteins, and the affinity value (Kd) is less than 1×10 μM. Antibodies for diagnostic use may be labeled with probes suitable for detection by various imaging methods. Methods for detection of probes include, but are not limited to, fluorescence, light, confocal and electron microscopy; magnetic resonance imaging and spectroscopy; fluoroscopy, computed tomography and positron emission tomography. Suitable probes include, but are not limited to, fluorescein, rhodamine, eosin and other fluorophores, radioisotopes, gold, gadolinium and other lanthanides, paramagnetic iron, fluorine-18 and other positron-emitting radionuclides. Additionally, probes may be bi- or multi-functional and be detectable by more than one of the methods listed. These antibodies may be directly or indirectly labeled with said probes. Attachment of probes to the antibodies includes covalent attachment of the probe, incorporation of the probe into the antibody, and the covalent attachment of a chelating compound for binding of probe, amongst others well recognized in the art. For immunohistochemistry, the disease tissue sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin. The fixed or embedded section contains the sample are contacted with a labeled primary antibody and secondary antibody, wherein the antibody is used to detect the expression of the proteins in situ. Another aspect of the present invention includes an in vitro method for producing activated T cells, the method comprising contacting in vitro T cells with antigen loaded human MHC molecules expressed on the surface of a suitable antigen-presenting cell for a period of time sufficient to activate the T cell in an antigen specific manner, wherein the antigen is a peptide according to the invention. Preferably a sufficient amount of the antigen is used with an antigen-presenting cell. Preferably the mammalian cell lacks or has a reduced level or function of the TAP peptide transporter. Suitable cells that lack the TAP peptide transporter include T2, RMA-S andDrosophilacells. TAP is the transporter associated with antigen processing. The human peptide loading deficient cell line T2 is available from the American Type Culture Collection, 12301 Parklawn Drive, Rockville, MD 20852, USA under Catalogue No CRL 1992; theDrosophilacell line Schneider line 2 is available from the ATCC under Catalogue No CRL 19863; the mouse RMA-S cell line is described in Ljunggren et al. (Ljunggren and Karre, 1985). Preferably, before transfection the host cell expresses substantially no MHC class I molecules. It is also preferred that the stimulator cell expresses a molecule important for providing a co-stimulatory signal for T-cells such as any of B7.1, B7.2, ICAM-1 and LFA 3. The nucleic acid sequences of numerous MHC class I molecules and of the co-stimulator molecules are publicly available from the GenBank and EMBL databases. In case of a MHC class I epitope being used as an antigen, the T cells are CD8-positive T cells. If an antigen-presenting cell is transfected to express such an epitope, preferably the cell comprises an expression vector capable of expressing a peptide containing SEQ ID NO: 1 to SEQ ID NO: 772, or a variant amino acid sequence thereof. A number of other methods may be used for generating T cells in vitro. For example, autologous tumor-infiltrating lymphocytes can be used in the generation of CTL. Plebanski et al. (Plebanski et al., 1995) made use of autologous peripheral blood lymphocytes (PLBs) in the preparation of T cells. Furthermore, the production of autologous T cells by pulsing dendritic cells with peptide or polypeptide, or via infection with recombinant virus is possible. Also, B cells can be used in the production of autologous T cells. In addition, macrophages pulsed with peptide or polypeptide, or infected with recombinant virus, may be used in the preparation of autologous T cells. S. Walter et al. (Walter et al., 2003) describe the in vitro priming of T cells by using artificial antigen presenting cells (aAPCs), which is also a suitable way for generating T cells against the peptide of choice. In the present invention, aAPCs were generated by the coupling of preformed MHC:peptide complexes to the surface of polystyrene particles (microbeads) by biotin:streptavidin biochemistry. This system permits the exact control of the MHC density on aAPCs, which allows to selectively eliciting high- or low-avidity antigen-specific T cell responses with high efficiency from blood samples. Apart from MHC:peptide complexes, aAPCs should carry other proteins with co-stimulatory activity like anti-CD28 antibodies coupled to their surface. Furthermore such aAPC-based systems often require the addition of appropriate soluble factors, e.g. cytokines, like interleukin-12. Allogeneic cells may also be used in the preparation of T cells and a method is described in detail in WO 97/26328, incorporated herein by reference. For example, in addition toDrosophilacells and T2 cells, other cells may be used to present antigens such as CHO cells, baculovirus-infected insect cells, bacteria, yeast, and vaccinia-infected target cells. In addition plant viruses may be used (see, for example, Porta et al. (Porta et al., 1994) which describes the development of cowpea mosaic virus as a high-yielding system for the presentation of foreign peptides. The activated T cells that are directed against the peptides of the invention are useful in therapy. Thus, a further aspect of the invention provides activated T cells obtainable by the foregoing methods of the invention. Activated T cells, which are produced by the above method, will selectively recognize a cell that aberrantly expresses a polypeptide that comprises an amino acid sequence of SEQ ID NO: 1 to SEQ ID NO 772. Preferably, the T cell recognizes the cell by interacting through its TCR with the HLA/peptide-complex (for example, binding). The T cells are useful in a method of killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention wherein the patient is administered an effective number of the activated T cells. The T cells that are administered to the patient may be derived from the patient and activated as described above (i.e. they are autologous T cells). Alternatively, the T cells are not from the patient but are from another individual. Of course, it is preferred if the individual is a healthy individual. By “healthy individual” the inventors mean that the individual is generally in good health, preferably has a competent immune system and, more preferably, is not suffering from any disease that can be readily tested for, and detected. In vivo, the target cells for the CD8-positive T cells according to the present invention can be cells of the tumor (which sometimes express MHC class II) and/or stromal cells surrounding the tumor (tumor cells) (which sometimes also express MHC class II; (Dengjel et al., 2006)). The T cells of the present invention may be used as active ingredients of a therapeutic composition. Thus, the invention also provides a method of killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention, the method comprising administering to the patient an effective number of T cells as defined above. By “aberrantly expressed” the inventors also mean that the polypeptide is over-expressed compared to levels of expression in normal tissues or that the gene is silent in the tissue from which the tumor is derived but in the tumor it is expressed. By “over-expressed” the inventors mean that the polypeptide is present at a level at least 1.2-fold of that present in normal tissue; preferably at least 2-fold, and more preferably at least 5-fold or 10-fold the level present in normal tissue. T cells may be obtained by methods known in the art, e.g. those described above. Protocols for this so-called adoptive transfer of T cells are well known in the art. Reviews can be found in: Gattioni et al. and Morgan et al. (Gattinoni et al., 2006; Morgan et al., 2006). Another aspect of the present invention includes the use of the peptides complexed with MHC to generate a T-cell receptor whose nucleic acid is cloned and is introduced into a host cell, preferably a T cell. This engineered T cell can then be transferred to a patient for therapy of cancer. Any molecule of the invention, i.e. the peptide, nucleic acid, antibody, expression vector, cell, activated T cell, T-cell receptor or the nucleic acid encoding it, is useful for the treatment of disorders, characterized by cells escaping an immune response. Therefore any molecule of the present invention may be used as medicament or in the manufacture of a medicament. The molecule may be used by itself or combined with other molecule(s) of the invention or (a) known molecule(s). The present invention is further directed at a kit comprising:(a) a container containing a pharmaceutical composition as described above, in solution or in lyophilized form;(b) optionally a second container containing a diluent or reconstituting solution for the lyophilized formulation; and(c) optionally, instructions for (i) use of the solution or (ii) reconstitution and/or use of the lyophilized formulation. The kit may further comprise one or more of (iii) a buffer, (iv) a diluent, (v) a filter, (vi) a needle, or (v) a syringe. The container is preferably a bottle, a vial, a syringe or test tube; and it may be a multi-use container. The pharmaceutical composition is preferably lyophilized. Kits of the present invention preferably comprise a lyophilized formulation of the present invention in a suitable container and instructions for its reconstitution and/or use. Suitable containers include, for example, bottles, vials (e.g. dual chamber vials), syringes (such as dual chamber syringes) and test tubes. The container may be formed from a variety of materials such as glass or plastic. Preferably the kit and/or container contain/s instructions on or associated with the container that indicates directions for reconstitution and/or use. For example, the label may indicate that the lyophilized formulation is to be reconstituted to peptide concentrations as described above. The label may further indicate that the formulation is useful or intended for subcutaneous administration. The container holding the formulation may be a multi-use vial, which allows for repeat administrations (e.g., from 2-6 administrations) of the reconstituted formulation. The kit may further comprise a second container comprising a suitable diluent (e.g., sodium bicarbonate solution). Upon mixing of the diluent and the lyophilized formulation, the final peptide concentration in the reconstituted formulation is preferably at least 0.15 mg/mL/peptide (=75 μg) and preferably not more than 3 mg/mL/peptide (=1500 μg). The kit may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. Kits of the present invention may have a single container that contains the formulation of the pharmaceutical compositions according to the present invention with or without other components (e.g., other compounds or pharmaceutical compositions of these other compounds) or may have distinct container for each component. Preferably, kits of the invention include a formulation of the invention packaged for use in combination with the co-administration of a second compound (such as adjuvants (e.g. GM-CSF), a chemotherapeutic agent, a natural product, a hormone or antagonist, an anti-angiogenesis agent or inhibitor, an apoptosis-inducing agent or a chelator) or a pharmaceutical composition thereof. The components of the kit may be pre-complexed or each component may be in a separate distinct container prior to administration to a patient. The components of the kit may be provided in one or more liquid solutions, preferably, an aqueous solution, more preferably, a sterile aqueous solution. The components of the kit may also be provided as solids, which may be converted into liquids by addition of suitable solvents, which are preferably provided in another distinct container. The container of a therapeutic kit may be a vial, test tube, flask, bottle, syringe, or any other means of enclosing a solid or liquid. Usually, when there is more than one component, the kit will contain a second vial or other container, which allows for separate dosing. The kit may also contain another container for a pharmaceutically acceptable liquid. Preferably, a therapeutic kit will contain an apparatus (e.g., one or more needles, syringes, eye droppers, pipette, etc.), which enables administration of the agents of the invention that are components of the present kit. The present formulation is one that is suitable for administration of the peptides by any acceptable route such as oral (enteral), nasal, ophthal, subcutaneous, intradermal, intramuscular, intravenous or transdermal. Preferably, the administration is s.c., and most preferably i.d. administration may be by infusion pump. Since the peptides of the invention were isolated from ovarian cancer, the medicament of the invention is preferably used to treat ovarian cancer. The present invention further relates to a method for producing a personalized pharmaceutical for an individual patient comprising manufacturing a pharmaceutical composition comprising at least one peptide selected from a warehouse of pre-screened TUMAPs, wherein the at least one peptide used in the pharmaceutical composition is selected for suitability in the individual patient. In one embodiment, the pharmaceutical composition is a vaccine. The method could also be adapted to produce T cell clones for down-stream applications, such as TCR isolations, or soluble antibodies, and other treatment options. A “personalized pharmaceutical” shall mean specifically tailored therapies for one individual patient that will only be used for therapy in such individual patient, including actively personalized cancer vaccines and adoptive cellular therapies using autologous patient tissue. As used herein, the term “warehouse” shall refer to a group or set of peptides that have been pre-screened for immunogenicity and/or over-presentation in a particular tumor type. The term “warehouse” is not intended to imply that the particular peptides included in the vaccine have been pre-manufactured and stored in a physical facility, although that possibility is contemplated. It is expressly contemplated that the peptides may be manufactured de novo for each individualized vaccine produced, or may be pre-manufactured and stored. The warehouse (e.g. in the form of a database) is composed of tumor-associated peptides which were highly overexpressed in the tumor tissue of ovarian cancer patients with various HLA-A HLA-B and HLA-C alleles. It may contain MHC class I and MHC class II peptides or elongated MHC class I peptides. In addition to the tumor associated peptides collected from several ovarian cancer tissues, the warehouse may contain HLA-A*02, HLA-A*01, HLA-A*03, HLA-A*24, HLA-B*07, HLA-B*08 and HLA-B*44 marker peptides. These peptides allow comparison of the magnitude of T-cell immunity induced by TUMAPS in a quantitative manner and hence allow important conclusion to be drawn on the capacity of the vaccine to elicit anti-tumor responses. Secondly, they function as important positive control peptides derived from a “non-self” antigen in the case that any vaccine-induced T-cell responses to TUMAPs derived from “self” antigens in a patient are not observed. And thirdly, it may allow conclusions to be drawn, regarding the status of immunocompetence of the patient. TUMAPs for the warehouse are identified by using an integrated functional genomics approach combining gene expression analysis, mass spectrometry, and T-cell immunology (XPresident®). The approach assures that only TUMAPs truly present on a high percentage of tumors but not or only minimally expressed on normal tissue, are chosen for further analysis. For initial peptide selection, ovarian cancer samples from patients and blood from healthy donors were analyzed in a stepwise approach:1. HLA ligands from the malignant material were identified by mass spectrometry2. Genome-wide messenger ribonucleic acid (mRNA) expression analysis was used to identify genes over-expressed in the malignant tissue (ovarian cancer) compared with a range of normal organs and tissues3. Identified HLA ligands were compared to gene expression data. Peptides presented on tumor tissue, preferably encoded by selectively expressed or over-expressed genes as detected in step 2 were considered suitable TUMAP candidates for a multi-peptide vaccine.4. Literature research was performed in order to identify additional evidence supporting the relevance of the identified peptides as TUMAPs5. The relevance of over-expression at the mRNA level was confirmed by redetection of selected TUMAPs from step 3 on tumor tissue and lack of (or infrequent) detection on healthy tissues.6. In order to assess, whether an induction of in vivo T-cell responses by the selected peptides may be feasible, in vitro immunogenicity assays were performed using human T cells from healthy donors as well as from ovarian cancer patients. In an aspect, the peptides are pre-screened for immunogenicity before being included in the warehouse. By way of example, and not limitation, the immunogenicity of the peptides included in the warehouse is determined by a method comprising in vitro T-cell priming through repeated stimulations of CD8+ T cells from healthy donors with artificial antigen presenting cells loaded with peptide/MHC complexes and anti-CD28 antibody. This method is preferred for rare cancers and patients with a rare expression profile. In contrast to multi-peptide cocktails with a fixed composition as currently developed, the warehouse allows a significantly higher matching of the actual expression of antigens in the tumor with the vaccine. Selected single or combinations of several “off-the-shelf” peptides will be used for each patient in a multitarget approach. In theory, an approach based on selection of e.g. 5 different antigenic peptides from a library of 50 would already lead to approximately 17 million possible drug product (DP) compositions. In an aspect, the peptides are selected for inclusion in the vaccine based on their suitability for the individual patient based on the method according to the present invention as described herein, or as below. The HLA phenotype, transcriptomic and peptidomic data is gathered from the patient's tumor material, and blood samples to identify the most suitable peptides for each patient containing “warehouse” and patient-unique (i.e. mutated) TUMAPs. Those peptides will be chosen, which are selectively or over-expressed in the patients' tumor and, where possible, show strong in vitro immunogenicity if tested with the patients' individual PBMCs. Preferably, the peptides included in the vaccine are identified by a method comprising: (a) identifying tumor-associated peptides (TUMAPs) presented by a tumor sample from the individual patient; (b) comparing the peptides identified in (a) with a warehouse (database) of peptides as described above; and (c) selecting at least one peptide from the warehouse (database) that correlates with a tumor-associated peptide identified in the patient. For example, the TUMAPs presented by the tumor sample are identified by: (a1) comparing expression data from the tumor sample to expression data from a sample of normal tissue corresponding to the tissue type of the tumor sample to identify proteins that are over-expressed or aberrantly expressed in the tumor sample; and (a2) correlating the expression data with sequences of MHC ligands bound to MHC class I and/or class II molecules in the tumor sample to identify MHC ligands derived from proteins over-expressed or aberrantly expressed by the tumor. Preferably, the sequences of MHC ligands are identified by eluting bound peptides from MHC molecules isolated from the tumor sample, and sequencing the eluted ligands. Preferably, the tumor sample and the normal tissue are obtained from the same patient. In addition to, or as an alternative to, selecting peptides using a warehousing (database) model, TUMAPs may be identified in the patient de novo, and then included in the vaccine. As one example, candidate TUMAPs may be identified in the patient by (a1) comparing expression data from the tumor sample to expression data from a sample of normal tissue corresponding to the tissue type of the tumor sample to identify proteins that are over-expressed or aberrantly expressed in the tumor sample; and (a2) correlating the expression data with sequences of MHC ligands bound to MHC class I and/or class II molecules in the tumor sample to identify MHC ligands derived from proteins over-expressed or aberrantly expressed by the tumor. As another example, proteins may be identified containing mutations that are unique to the tumor sample relative to normal corresponding tissue from the individual patient, and TUMAPs can be identified that specifically target the mutation. For example, the genome of the tumor and of corresponding normal tissue can be sequenced by whole genome sequencing: For discovery of non-synonymous mutations in the protein-coding regions of genes, genomic DNA and RNA are extracted from tumor tissues and normal non-mutated genomic germline DNA is extracted from peripheral blood mononuclear cells (PBMCs). The applied NGS approach is confined to the re-sequencing of protein coding regions (exome re-sequencing). For this purpose, exonic DNA from human samples is captured using vendor-supplied target enrichment kits, followed by sequencing with e.g. a HiSeq2000 (Illumina). Additionally, tumor mRNA is sequenced for direct quantification of gene expression and validation that mutated genes are expressed in the patients' tumors. The resultant millions of sequence reads are processed through software algorithms. The output list contains mutations and gene expression. Tumor-specific somatic mutations are determined by comparison with the PBMC-derived germline variations and prioritized. The de novo identified peptides can then be tested for immunogenicity as described above for the warehouse, and candidate TUMAPs possessing suitable immunogenicity are selected for inclusion in the vaccine. In one exemplary embodiment, the peptides included in the vaccine are identified by: (a) identifying tumor-associated peptides (TUMAPs) presented by a tumor sample from the individual patient by the method as described above; (b) comparing the peptides identified in a) with a warehouse of peptides that have been prescreened for immunogenicity and overpresentation in tumors as compared to corresponding normal tissue; (c) selecting at least one peptide from the warehouse that correlates with a tumor-associated peptide identified in the patient; and (d) optionally, selecting at least one peptide identified de novo in (a) confirming its immunogenicity. In one exemplary embodiment, the peptides included in the vaccine are identified by: (a) identifying tumor-associated peptides (TUMAPs) presented by a tumor sample from the individual patient; and (b) selecting at least one peptide identified de novo in (a) and confirming its immunogenicity. Once the peptides for a personalized peptide based vaccine are selected, the vaccine is produced. The vaccine preferably is a liquid formulation consisting of the individual peptides dissolved in between 20-40% DMSO, preferably about 30-35% DMSO, such as about 33% DMSO. Each peptide to be included into a product is dissolved in DMSO. The concentration of the single peptide solutions has to be chosen depending on the number of peptides to be included into the product. The single peptide-DMSO solutions are mixed in equal parts to achieve a solution containing all peptides to be included in the product with a concentration of ˜2.5 mg/ml per peptide. The mixed solution is then diluted 1:3 with water for injection to achieve a concentration of 0.826 mg/ml per peptide in 33% DMSO. The diluted solution is filtered through a 0.22 μm sterile filter. The final bulk solution is obtained. Final bulk solution is filled into vials and stored at −20° C. until use. One vial contains 700 μL solution, containing 0.578 mg of each peptide. Of this, 500 μL (approx. 400 μg per peptide) will be applied for intradermal injection. In addition to being useful for treating cancer, the peptides of the present invention are also useful as diagnostics. Since the peptides were generated from ovarian cancer cells and since it was determined that these peptides are not or at lower levels present in normal tissues, these peptides can be used to diagnose the presence of a cancer. The presence of claimed peptides on tissue biopsies in blood samples can assist a pathologist in diagnosis of cancer. Detection of certain peptides by means of antibodies, mass spectrometry or other methods known in the art can tell the pathologist that the tissue sample is malignant or inflamed or generally diseased, or can be used as a biomarker for ovarian cancer. Presence of groups of peptides can enable classification or sub-classification of diseased tissues. The detection of peptides on diseased tissue specimen can enable the decision about the benefit of therapies involving the immune system, especially if T-lymphocytes are known or expected to be involved in the mechanism of action. Loss of MHC expression is a well described mechanism by which infected of malignant cells escape immuno-surveillance. Thus, presence of peptides shows that this mechanism is not exploited by the analyzed cells. The peptides of the present invention might be used to analyze lymphocyte responses against those peptides such as T cell responses or antibody responses against the peptide or the peptide complexed to MHC molecules. These lymphocyte responses can be used as prognostic markers for decision on further therapy steps. These responses can also be used as surrogate response markers in immunotherapy approaches aiming to induce lymphocyte responses by different means, e.g. vaccination of protein, nucleic acids, autologous materials, adoptive transfer of lymphocytes. In gene therapy settings, lymphocyte responses against peptides can be considered in the assessment of side effects. Monitoring of lymphocyte responses might also be a valuable tool for follow-up examinations of transplantation therapies, e.g. for the detection of graft versus host and host versus graft diseases. The present invention will now be described in the following examples which describe preferred embodiments thereof, and with reference to the accompanying figures, nevertheless, without being limited thereto. For the purposes of the present invention, all references as cited herein are incorporated by reference in their entireties. FIGURES FIGS.1A through1Sshow exemplary expression profile of source genes of the present invention that are over-expressed in different cancer samples. Tumor (black dots) and normal (grey dots) samples are grouped according to organ of origin, and box-and-whisker plots represent median, 25th and 75th percentile (box), and minimum and maximum (whiskers) RPKM values. Normal organs are ordered according to risk categories. RPKM=reads per kilobase per million mapped reads. Normal samples: blood cells; blood vessel; brain; heart; liver; lung; adipose: adipose tissue; adren.gl.: adrenal gland; bile duct; bladder; BM: bone marrow; cartilage; esoph: esophagus; eye; gallb: gallbladder; head and neck; kidney; large_int: large intestine; LN: lymph node; nerve; pancreas; parathyr: parathyroid; pituit: pituitary; skel.mus: skeletal muscle; skin; small_int: small intestine; spleen; stomach; thyroid; trachea; bladder; breast; ovary; placenta; prostate; testis; thymus; uterus. Tumor samples: AML: acute myeloid leukemia; BRCA: breast cancer; CLL: chronic lymphocytic leukemia; CRC: colorectal cancer; GALB: gallbladder cancer; GB: glioblastoma; GC: gastric cancer; HCC: hepatocellular carcinoma; HNSCC: head-and-neck cancer; MEL: melanoma; NHL: non-hodgkin lymphoma; NSCLC: non-small cell lung cancer; OC: ovarian cancer; OSC_GC: esophageal/gastric cancer; OSCAR: esophageal cancer; PC: pancreatic cancer; PCA: prostate cancer; RCC: renal cell carcinoma; SCLC: small cell lung cancer; UBC: urinary bladder carcinoma; UEC: uterine and endometrial cancer.FIG.1A) Gene symbol: CT45A2, Peptide: KYEKIFEML (SEQ ID No.: 12),FIG.1B) Gene symbol: NLRP2, Peptide: VLYGPAGLGK (SEQ ID No.: 27),FIG.1C) Gene symbol: NLRP7, Peptide: LLDEGAMLLY (SEQ ID No.: 31),FIG.1D) Gene symbol: HTR3A, Peptide: GLLQELSSI (SEQ ID No.: 66),FIG.1E) Gene symbol: VTCN1, Peptide: KVVSVLYNV (SEQ ID No.: 75),FIG.1F) Gene symbol: CYP2W1, Peptide: RYGPVFTV (SEQ ID No.: 98),FIG.1G) Gene symbol: MMP11, Peptide: LLQPPPLLAR (SEQ ID No.: 98),FIG.1H) Gene symbol: MMP12, Peptide: FVDNQYWRY (SEQ ID No.: 115),FIG.1I) Gene symbol: CTAG2, Peptide: APLPRPGAVL (SEQ ID No.: 119),FIG.1J) Gene symbol: FAM111B, Peptide: KPSESIYSAL (SEQ ID No.: 123),FIG.1K) Gene symbol: CCNA1, Peptide: HLLLKVLAF (SEQ ID No.: 151),FIG.1L) Gene symbol: FAM83H, Peptide: HVKEKFLL (SEQ ID No.: 156),FIG.1M) Gene symbol: MAGEA11, Peptide: KEVDPTSHSY (SEQ ID No.: 194),FIG.1N) Gene symbol: MMP11, Peptide: YTFRYPLSL (SEQ ID No.: 227),FIG.1O) Gene symbol: ZNF560, Peptide: VFVSFSSLF (SEQ ID No.: 255),FIG.1P) Gene symbol: IGF2BP1, Peptide: ISYSGQFLVK (SEQ ID No.: 266),FIG.1Q) Gene symbol: CLDN6, Peptide: LPMWKVTAF (SEQ ID No.: 303),FIG.1R) Gene symbol: IGF2BP3, Peptide: IEALSGKIEL (SEQ ID No.: 413),FIG.1S) Gene symbol: PRAME, Peptide: EEQYIAQF (SEQ ID No.: 432). FIGS.1T through1Vshow exemplary expression profiles of source genes of the present invention, that are over-expressed in different cancer samples. Tumor (black dots) and normal (grey dots) samples are grouped according to organ of origin. Box-and-whisker plots represent median FPKM value, 25th and 75th percentile (box) plus whiskers that extend to the lowest data point still within 1.5 interquartile range (IQR) of the lower quartile and the highest data point still within 1.5 IQR of the upper quartile. Normal organs are ordered according to risk categories. FPKM: fragments per kilobase per million mapped reads. Normal samples: blood cells; bloodvess (blood vessels); brain; heart; liver; lung; adipose (adipose tissue); adrenal gl (adrenal gland); bile duct; bladder; bone marrow; cartilage; esoph (esophagus); eye; gall bl (gallbladder); head&neck; intest. la (large intestine); intest. sm (small intestine); kidney; lymph node; nerve perith (peripheral nerve); pancreas; parathyr (parathyroid gland); perit (peritoneum); pituit (pituitary); pleura; skel. mus (skeletal muscle); skin; spleen; stomach; thyroid; trachea; ureter; breast; ovary; placenta; prostate; testis; thymus; uterus. Tumor samples: AML (acute myeloid leukemia); BRCA (breast cancer); CCC (cholangiocellular carcinoma); CLL (chronic lymphocytic leukemia); CRC (colorectal cancer); GBC (gallbladder cancer); GBM (glioblastoma); GC (gastric cancer); HCC (hepatocellular carcinoma); HNSCC (head and neck squamous cell carcinoma); MEL (melanoma); NHL (non-hodgkin lymphoma); NSCLCadeno (non-small cell lung cancer adenocarcinoma); NSCLCother (NSCLC samples that could not unambiguously be assigned to NSCLCadeno or NSCLCsquam); NSCLCsquam (squamous cell non-small cell lung cancer); OC (ovarian cancer); OSCAR (esophageal cancer); PACA (pancreatic cancer); PRCA (prostate cancer); RCC (renal cell carcinoma); SCLC (small cell lung cancer); UBC (urinary bladder carcinoma); UEC (uterine and endometrial cancer).FIG.1T) Gene symbol: MAGEA4, Peptide: SPDAESLFREALSNKVDEL (SEQ ID No.: 597),FIG.1U) Gene symbol: MAGEA4, Peptide: LSNKVDELAHFLLRK (SEQ ID No.: 601),FIG.1V) Gene symbol: MAGEB3, Peptide: KLITQDLVKLKYLEYRQ (SEQ ID No.: 604). FIG.2shows exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-A*02+ donor. CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-A*02 in complex with SeqID No 773 peptide (ALYGKLLKL, Seq ID NO: 773) (left panel. After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with A*02/SeqID No 773. Right panel shows control staining of cells stimulated with irrelevant A*02/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated. FIG.3shows exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-A*24+ donor. CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-A*24 in complex with SeqID No 774 peptide (left panel). After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with A*24/SeqID No 774 (VYVDDIYVI, Seq ID NO: 774). Right panel shows control staining of cells stimulated with irrelevant A*24/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated. FIGS.4A and4Bshow exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-A*02+ donor. CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-A*02 in complex with SeqID No 67 peptide SLLLPSIFL (FIG.4A, left panel) and SeqID No 75 peptide KVVSVLYNV (FIG.4B, left panel), respectively. After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with A*02/SeqID No 67 (FIG.4A) or A*02/SeqID No 75 (FIG.4B). Right panels (FIGS.4A and4B) show control staining of cells stimulated with irrelevant A*02/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated. FIGS.5A and5Bshow exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-A*24+ donor. CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-A*24 in complex with SeqID No 11 peptide SYSDLHYGF (FIG.5A, left panel) and SeqID No 79 peptide SYNEHWNYL (FIG.5B, left panel), respectively. After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with A*24/SeqID No 11 (FIG.5A) or A*24/SeqID No 79 (FIG.5B). Right panels (FIGS.5A and5B) show control staining of cells stimulated with irrelevant A*24/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated. FIGS.6A and6Bshow exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-B*07+ donor. CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-B*07 in complex with SeqID No 33 peptide SPTFHLTL (FIG.6A, left panel) and SeqID No 40 peptide KPGTSYRVTL (FIG.6B, left panel), respectively. After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with B*07/SeqID No 33 (FIG.6A) or B*07/SeqID No 40 (FIG.6B). Right panels (FIGS.6A and6B) show control staining of cells stimulated with irrelevant B*07/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated. FIGS.7A and7Bshow exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-A*01+ donor. CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-A*01 in complex with SeqID No 113 peptide QLDSNRLTY (FIG.7A, left panel) and SeqID No 115 peptide FVDNQYWRY (FIG.7B, left panel), respectively. After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with A*01/SeqID No 113 (FIG.7A) or A*01/SeqID No 115 (FIG.7B). Right panels (FIGS.7A and7B) show control staining of cells stimulated with irrelevant A*01/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated. FIGS.8A and8Bshow exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-A*03+ donor. CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-A*03 in complex with SeqID No 23 peptide GMMKGGIRK (FIG.8A, left panel) and SeqID No 90 peptide KVAGERYVYK (FIG.8B, left panel), respectively. After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with A*03/SeqID No 23 (FIG.8A) or A*03/SeqID No 90 (FIG.8B). Right panels (FIGS.8A and8B) show control staining of cells stimulated with irrelevant A*03/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated. FIGS.9A and9Bshow exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-B*44+ donor. CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-B*44 in complex with SeqID No 200 peptide AESIPTVSF (FIG.9A, left panel) and SeqID No 211 peptide EEKVFPSPLW (FIG.9B, left panel), respectively. After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with B*44/SeqID No 200 (FIG.9A) or B*44/SeqID No 211 (FIG.9B). Right panels (FIGS.9A and9B) show control staining of cells stimulated with irrelevant B*44/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated. EXAMPLES Example 1 Identification of Tumor Associated Peptides Presented on the Cell Surface Tissue Samples Patients' tumor tissues and normal tissues were obtained from the University Hospital Tübingen (Tübingen, Germany). Written informed consents of all patients had been given before surgery or autopsy. Tissues were shock-frozen immediately after excision and stored until isolation of TUMAPs at −70° C. or below. Isolation of HLA Peptides from Tissue Samples HLA peptide pools from shock-frozen tissue samples were obtained by immune precipitation from solid tissues according to a slightly modified protocol (Falk et al., 1991; Seeger et al., 1999) using the HLA-A*02-specific antibody BB7.2, the HLA-A, —B, C-specific antibody W6/32, the HLA-DR specific antibody L243 and the pan-HLA class II specific antibody TU39, CNBr-activated sepharose, acid treatment, and ultrafiltration. Mass Spectrometry Analyses The HLA peptide pools as obtained were separated according to their hydrophobicity by reversed-phase chromatography (Ultimate 3000 RSLC Nano UHPLC System, Dionex)) and the eluting peptides were analyzed in LTQ-Orbitrap and Fusion Lumos hybrid mass spectrometers (ThermoElectron) equipped with an ESI source. Peptide samples were loaded with 3% of solvent B (20% H2O, 80% acetonitrile and 0.04% formic acid) on a 2 cm PepMap 100 C18 Nanotrap column (Dionex) at a flowrate of 4 μI/min for 10 min. Separation was performed on either 25 cm or 50 cm PepMap C18 columns with a particle size of 2 μm (Dionex) mounted in a column oven running at 50° C. The applied gradient ranged from 3 to 32% solvent B within 90 min at a flow rate of 300 nl/min (for 25 cm columns) or 140 min at a flow rate of 175 nl/min (for 50 cm columns). (Solvent A: 99% H2O, 1% ACN and 0.1% formic acid; Solvent B: 20% H2O, 80% ACN and 0.1% formic acid). Mass spectrometry analysis was performed in data dependent acquisition mode employing a top five method (i.e. during each survey scan the five most abundant precursor ions were selected for fragmentation). Alternatively, a TopSpeed method was employed for analysis on Fusion Lumos instruments, Survey scans were recorded in the Orbitrap at a resolution of 60,000 (for Orbitrap XL) or 120,000 (for Orbitrap Fusion Lumos). MS/MS analysis was performed by collision induced dissociation (CID, normalized collision energy 35%, activation time 30 ms, isolation width 1.3 m/z) with subsequent analysis in the linear trap quadrupole (LTQ). Mass range for HLA class I ligands was limited to 400-650 m/z with possible charge states 2+ and 3+ selected for fragmentation. For HLA class II mass range was set to 300-1500 m/z allowing for fragmentation with all positive charge states ≥2. Tandem mass spectra were interpreted by MASCOT or SEQUEST at a fixed false discovery rate (q≤0.05) and additional manual control. In cases where the identified peptide sequence was uncertain it was additionally validated by comparison of the generated natural peptide fragmentation pattern with the fragmentation pattern of a synthetic sequence-identical reference peptide. Table 19 shows the presentation on various cancer entities for selected peptides, and thus the particular relevance of the peptides as mentioned for the diagnosis and/or treatment of the cancers as indicated (e.g. peptide SEQ ID No. 1 for colorectal cancer, gallbladder cancer, non-hodgkin lymphoma, non-small cell lung cancer, and uterine and endometrial cancer, peptide SEQ ID No. 2 for breast cancer, cholangiocellular carcinoma, colorectal cancer, gallbladder cancer, gastric cancer, head and neck squamous cell carcinoma, melanoma, non-hodgkin lymphoma, non-small cell lung cancer, esophageal cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, small cell lung cancer, and uterine and endometrial cancer). TABLE 19Overview of presentation of selected tumor-associatedpeptides of the present invention across tumor types.AML: acute myeloid leukemia; BRCA: breast cancer; CCC:cholangiocellular carcinoma; CLL: chronic lymphocyticleukemia; CRC: colorectal cancer; GBC: gallbladder cancer;GBM: glioblastoma; GC: gastric cancer; GEJC: gastro-esophagealjunction cancer; HCC: hepatocellular carcinoma; HNSCC: headand neck squamous cell carcinoma; MEL: melanoma; NHL: non-hodgkinlymphoma; NSCLC: non-small cell lung cancer; OC: ovarian cancer;OSCAR: esophageal cancer; PACA: pancreatic cancer; PRCA: prostatecancer; RCC: renal cell carcinoma; SCLC: small cell lung cancer;UBC: urinary bladder carcinoma; UEC: uterine and endometrial cancerSeq IDNoSequencePeptide Presentation on tumor types1MIPTFTALLCRC, GBC, NHL, NSCLC, UEC2TLLKALLEIBRCA, CCC, CRC, GBC, GC, HNSCC,MEL, NHL, NSCLC, OSCAR, PACA,PRCA, RCC, SCLC, UEC3ALIYNLVGIHCC4ALFKAWALAML, BRCA, CLL, CRC, GBC, GC, HCC,HNSCC, MEL, NHL, NSCLC, OSCAR,RCC, SCLC, UBC, UEC5RLLDFINVLUECGC, GEJC, HNSCC, NSCLC, PACA,7ALQAFEFRVSCLC, UBC8YLVTKVVAVAML, BRCA, CCC, CLL, CRC, GBC,GBM, GC, GEJC, HCC, HNSCC, MEL,NHL, NSCLC, OSCAR, PACA, PRCA,RCC, SCLC, UBC, UEC10RYSDSVGRVSFBRCA, CRC, GBC, GC, NSCLC, SCLC,UBC, UEC11SYSDLHYGFGC, NSCLC, UEC12KYEKIFEMLAML, NSCLC13VYTFLSSTLNSCLC14FYFPTPTVLGBC, NSCLC15VYHDDKQPTFGBM, GC, NSCLC, OSCAR, UECBRCA, NHL, NSCLC, OSCAR, UBC,16IYSPQFSRLUEC18KYPVHIYRLAML, BRCA, GBC, GC, HNSCC, MEL,NHL, NSCLC, OSCAR, PACA, RCC,UBC, UEC19KYVKVFHQFAML, BRCA, CLL, CRC, GBC, GBM, GC,HCC, MEL, NHL, NSCLC, OSCAR,PACA, PRCA, RCC, SCLC, UBC, UEC20RMASPVNVKCLL21AVRKPIVLKAML, BRCA, CCC, CRC, GBC, GBM,GC, HCC, HNSCC, MEL, NHL, NSCLC,OSCAR, PACA, RCC, SCLC, UBC, UEC22SLKERNPLKNSCLC24SMYYPLQLKBRCA, CRC, GBM, HCC, NHL, RCC25GTSPPSVEKUEC27VLYGPAGLGKHCC, HNSCC, NSCLC, OSCAR, PACA,SCLC, UBC, UEC28KTYETNLEIKKNSCLC, UBC29QQFLTALFYPACA, PRCA31LLDEGAMLLYGBC, HNSCC, NSCLC, SCLC, UBC32SPNKGTLSVNSCLC33SPTFHLTLNSCLC, PRCA, SCLC, UBC, UEC34LPRGPLASLLHNSCC, NSCLC, OSCAR, PACA, SCLC35FPDNQRPALBRCA, CRC, GBC, MEL, NSCLC, PACA,UBC, UEC36APAAWLRSABRCA, CCC, CRC, GBC, GC, HCC,HNSCC, NSCLC, OSCAR, PACA, SCLC,UBC, UEC38SPHPVTALLTLPACA, UEC40KPGTSYRVTLGBM43ALKARTVTFBRCA, CCC, GBM, HCC, HNSCC, MEL,NHL, NSCLC, OSCAR, PRCA, SCLC,UBC, UEC48DVKKKIKEVNSCLC, RCC, SCLC53MEHPGKLLFUEC56SEPDTTASWNSCLC, UEC57QESDLRLFLBRCA, CLL, CRC, GC, GEJC, HNSCC,NHL, NSCLC, PACA, UBC, UEC59SENVTMKVVUEC60GLLSLTSTLYLBRCA62KVLGVNVMLBRCA, HNSCC, MEL, NSCLC, SCLC63MMEEMIFNLUBC64FLDPDRHFLBRCA, CCC, CRC, GBC, GC, GEJC,HCC, HNSCC, MEL, NSCLC, OSCAR,PACA, PRCA, RCC, SCLC, UBC, UEC65TMFLRETSLMEL, NHL, NSCLC, PRCA, SCLC68KLFDTQQFLAML, BRCA, CRC, HCC, HNSCC, MEL,NHL, NSCLC, OSCAR, RCC69TTYEGSITVNSCLC, UEC71YLEDTDRNLAML, BRCA, CCC, CRC, GBC, GC,GEJC, HCC, HNSCC, MEL, NHL,NSCLC, OSCAR, PACA, PRCA, RCC,SCLC, UBC, UEC72YLTDLQVSLAML, BRCA, CCC, CLL, CRC, GBC,GBM, GC, GEJC, HCC, HNSCC, MEL,NHL, NSCLC, OSCAR, PACA, PRCA,RCC, SCLC, UBC, UEC74SQSPSVSQLUEC75KVVSVLYNVBRCA, UEC77RYGPVFTVCCC, GC78SFAPRSAVFSCLC79SYNEHWNYLBRCA, CCC, CRC, GBC, GC, HCC,HNSCC, MEL, NHL, NSCLC, OSCAR,PACA, PRCA, RCC, SCLC, UBC, UEC81VYNHTTRPLOSCAR85VLLGSLFSRKAML, CRC, HCC, MEL, NHL, NSCLC,RCC, UEC86VVLLGSLFSRKAML, CRC, GC, HCC, PACA, RCC87AVAPPTPASKAML, CRC, GBC, MEL, NSCLC, OSCAR,RCC, SCLC, UEC90KVAGERYVYKCCC, UEC92SVFPIENIYUEC94ATFERVLLRBRCA, NSCLC96TAFGGFLKYOSCAR, RCC97TMLDVEGLFYGC99KVVDRWNEKCRC, NHL, RCC101RVFTSSIKTKNSCLC, PACA, UEC106AAFVPLLLKAML, BRCA, NHL, NSCLC, SCLC108VLYPVPLESYAML, MEL, NHL, NSCLC, RCC, SCLC,UEC109KTFTIKRFLAKBRCA, CCC, MEL, NHL, NSCLC,OSCAR, SCLC, UEC110SAAPPSYFRRCC, UEC113QLDSNRLTYHCC115FVDNQYWRYBRCA, GBC, GC, GEJC,NSCLC,OSCAR, PACA, SCLC116VLLDEGAMLLYNSCLC, PACA117APRLLLLAVLBRCA, CRC, HNSCC, MEL, NSCLC,OSCAR, PRCA, RCC, SCLC, UBC, UEC118SPASRSISLNHL, OSCAR, RCC119APLPRPGAVLMEL, OSCAR120RPAMNYDKLCRC123KPSESIYSALBRCA, CRC, HNSCC, MEL, NHL,NSCLC, OSCAR, SCLC, UBC124LPSDSHFKITFCRC, HNSCC, NHL, OSCAR, SCLC125VPVYILLDEMCCC, GC, HNSCC, UEC127APRAGSQVVAML, BRCA, CRC, GBM, HCC, HNSCC,MEL, NSCLC, OSCAR, PACA, PRCA,RCC, SCLC, UBC, UEC129APRPASSLBRCA, CRC, NSCLC, OSCAR, SCLC,UEC133MPNLPSTTSLUEC141SPMTSLLTSGLUEC146IPRPEVQALAML, CRC, GC, HNSCC, MEL147APRWFPQPTVVBRCA148KPYGGSGPLAML, BRCA, NHL, RCC149GPREALSRLAML, BRCA, CCC, CRC, HCC, MEL,NHL, NSCLC, OSCAR, PACA, PRCA,RCC, SCLC, UEC150MAAVKQALCCC, NSCLC, PACA151HLLLKVLAFHNSCC152MGSARVAELHNSCC156HVKEKFLLCCC, HNSCC157EAMKRLSYICCC, HNSCC, PACA174AEATARLNVFNSCLC176AEIEPKADGSWCCC, NSCLC, PRCA178NELFRDGVNWAML, BRCA, CCC, CLL, CRC, HCC,HNSCC, MEL, NHL, NSCLC, OSCAR,PACA, PRCA, SCLC, UBC, UEC179REAGDEFELCCC, NSCLC, SCLC180REAGDEFELRYCRC, HCC, MEL, NSCLC, OSCAR,PACA, RCC, UEC181GEGPKTSWNSCLC182KEATEAQSLNSCLC184AELEALTDLWNHL, NSCLC, NSCLC186REGPEEPGLGC188AEFAKKQPWWCCC, CLL, CRC, MEL, NHL, NSCLC191EEDAALFKAWAML, BRCA, CCC, CLL, CRC, GC, HCC,HNSCC, MEL, NHL, NSCLC, OSCAR,PACA, PRCA, RCC, SCLC, UBC, UEC192YEFKFPNRLBRCA, HCC, NSCLC, OSCAR, UEC196REMPGGPVWBRCA, CCC, CRC, GC, HCC, HNSCC,MEL, NHL, NSCLC, OSCAR, PACA,SCLC, UBC, UEC197AEVLLPRLVNSCLC, PACA, UEC199REIDESLIFYNSCLC200AESIPTVSFNSCLC208TEVSRTEAINSCLC, UEC211EEKVFPSPLWNHL215SEDGLPEGIHLCLL, GC, GEJC, HNSCC, NHL, NSCLC,PACA216IMFDDAIERAUEC217VSSSLTLKVBRCA, RCC224SLPRFQVTLBRCA, HCC, NHL, NSCLC, OSCAR,UBC, UEC225SVFAHPRKLBRCA, OSCAR226QVDPKKRISMBRCA, NHL, NSCLC, SCLC227YTFRYPLSLCCC, CRC, GBC, GC, HCC, HNSCC,NSCLC, OSCAR, PACA, SCLC, UBC,UEC228RLWDWVPLAAML, NHL235SAIETSAVLNSCLC, UEC237SAMGTISIMUEC240FSTDTSIVLPACA241RQPNILVHLUEC243YASEGVKQVUEC245LAVEGGQSLAML, BRCA, CCC, CRC, GBC, GBM,GC, GEJC, HCC, HNSCC, MEL, NHL,NSCLC, OSCAR, PACA, PRCA, RCC,SCLC, UBC, UEC246RYLAVVHAVFHCC, NHL, NSCLC, PACA, SCLC247ARPPWMWVLBRCA, GBC, HNSCC, OSCAR251KQRQVLIFFGBM, NSCLC, OSCAR, PACA, RCC252LYQPRASEMNHL256RTEEVLLTFKRCC, SCLC, UEC257VTADHSHVFUEC259KTLELRVAYGBC, HNSCC260GTNTVILEYMEL, PACA, UEC262RSRLNPLVQRHNSCC, NSCLC264AIKVIPTVFKHNSCC, MEL, NSCLC, RCC, UEC268GLLGLSLRYPRCA269RLKGDAWVYKMEL, NHL, OSCAR, UEC271RMFADDLHNLNKNSCLC273RVNAIPFTYGBC275STTFPTLTKUEC277TTALKTTSRNSCLC279SVSSETTKIKRUEC280SVSGVKTTFHCC, UEC281RAKELEATFCLL, GC, NSCLC283IVQEPTEEKHCC, NHL, NSCLC286TVAPPQGVVKHCC288SPVTSVHGGTYNHL289RWEKTDLTYCRC, UEC291ETIRSVGYYGBM, NSCLC, UBC295YPLRGSSIFGLUEC296YPLRGSSIUEC299HPGSSALHYAML, CCC, CRC, GC, HNSCC, MEL,NHL, NSCLC, OSCAR, PACA, PRCA,RCC, UEC300IPMAAVKQALAML, BRCA, CLL, CRC, GC, HCC,HNSCC, MEL, NSCLC, OSCAR, PACA,RCC, UEC302RVEEVRALLBRCA, CRC, GBM, UBC306APVIFSHSAAML, CCC, HCC, MEL, NSCLC, UBC307LPYGPGSEAAAFBRCA, UEC308YPEGAAYEFPRCA, UEC314VPDQPHPEIPACA315SPRENFPDTLHNSCC317FPFQPGSVAML, BRCA, CLL, CRC, GBC, GBM, GC,HCC, HNSCC, MEL, NHL, NSCLC,OSCAR, PACA, PRCA, RCC, SCLC,UBC, UEC318FPNRLNLEACCC, CLL, GC, HNSCC, MEL, NSCLC,PRCA, RCC, SCLC, UBC319SPAEPSVYATLBRCA, GC, NSCLC, OSCAR320FPMSPVTSVAML, BRCA, CCC, CRC, GBC, GBM,GC, HCC, HNSCC, MEL, NHL, NSCLC,OSCAR, PACA, PRCA, RCC, SCLC,UBC, UEC321SPMDTFLLIAML, BRCA, CLL, CRC, GBC, GBM, GC,HCC, HNSCC, MEL, NHL, NSCLC,OSCAR, PACA, PRCA, RCC, SCLC,UBC, UEC322SPDPSKHLLNHL, NSCLC, PRCA, RCC324VPYRVVGLCLL, CRC, GC, MEL, NHL, NSCLC,PRCA, SCLC325GPRNAQRVLCRC, GBC, NHL, NSCLC326VPSEIDAAFBRCA, CCC, CRC, GBC, GC, NSCLC,OSCAR, PACA, RCC, SCLC, UEC330FPFVTGSTEMUEC331FPHPEMTTSMUEC332FPHSEMTTLNSCLC, PACA333FPHSEMTTVMNSCLC, SCLC, UEC334FPYSEVTTLNSCLC, SCLC, UEC335HPDPVGPGLNSCLC, UEC337HPVETSSALUEC355SPLVTSHIMUEC363TAKTPDATFCCC369FPHSEMTTVPACA, UEC371LYVDGFTHWNSCLC, UEC376RPRSPAGQVAPACA378RPRSPAGQVAANHL, PACA, SCLC385SPALHIGSVBRCA, GBM, HCC, NSCLC, PRCA,SCLC, UBC, UEC386FPFNPLDFGC, NHL388SPAPLKLSRTPAMEL389SPGAQRTFFQLAML, MEL391APSTPRITTFHCC, NHL392KPIESTLVAGBM, MEL, NSCLC, UEC393ASKPHVEICRC395VLLPRLVSCNSCLC399RELLHLVTLNSCLC, SCLC, UEC403EEAQWVRKYBRCA, CLL, NHL404NEAIMHQYBRCA, CCC, CLL, CRC, GBC, GC, HCC,MEL, NHL, NSCLC, OSCAR, SCLC,UBC, UEC405NEIWTHSYNSCLC, UEC407AEHEGVSVLNSCLC, UEC408LEKALQVFCRC, GC, HNSCC, OSCAR, UEC409REFVLSKGDAGLGBC, GC, GEJC, HNSCC, NSCLC410SEDPSKLEABRCA, HNSCC, NSCLC, OSCAR, SCLC,UEC411LELPPILVYBRCA, CRC, GBC, GBM, GC, NHL,NSCLC, OSCAR, PRCA, SCLC, UBC,UEC414EDAALFKAWCLL, CRC, MEL, NHL415REEDAALFKAWBRCA, CLL, CRC, HCC, HNSCC, MEL,NHL, NSCLC, OSCAR, PRCA, UBC,UEC416SEEETRVVFAML, CRC, HNSCC, NSCLC, UEC417AEHFSMIRAAML, BRCA, CRC, GBM, GC, HNSCC,NHL, NSCLC, OSCAR, PACA, PRCA,RCC, UEC418FEDAQGHIWBRCA, CCC, CRC, HCC, NSCLC,OSCAR, PACA, UBC, UEC419HEFGHVLGLBRCA, CCC, CRC, GC, HNSCC, MEL,NSCLC, OSCAR, PACA, UEC420FESHSTVSAUEC423SEVPTGTTAGBC, GBM425SEVPLPMAINSCLC, UEC429REKFIASVIUEC430DEKILYPEFUEC431AEQDPDELNKACRC, OSCAR, SCLC, UEC432EEQYIAQFOSCAR, SCLC433SDSQVRAFGBM, GC, HCC, HNSCC, NSCLC,OSCAR, RCC, SCLC, UEC436REPGDIFSELCRC437TEAVVTNELCRC, GC, NSCLC, SCLC, UEC438SEVDSPNVLCCC, CLL, CRC, GC, HNSCC, MEL,NHL, NSCLC, SCLC, UBC442ILSKLTDIQYBRCA, GBM, NHL, NSCLC443GTFNPVSLWBRCA, GBC, GBM, HNSCC, MEL, NHL,NSCLC, OSCAR, PACA, SCLC444KLSQKGYSWBRCA, CCC, CRC, GBM, HCC, HNSCC,MEL, NHL, NSCLC, OSCAR, PACA,PRCA, SCLC, UBC445LHITPGTAYHCC, PRCA446GRIVAFFSFAML, BRCA, CRC, HCC, HNSCC, MEL,NHL, NSCLC, OSCAR, PACA, PRCA,SCLC, UBC, UEC447MQVLVSRIGC, NSCLC, PACA, PRCA, RCC, SCLC448LSQKGYSWNHL, NSCLC, UBC451DYLNEWGSRFNSCLC, OSCAR, UEC454AQTDPTTGYGBM, GC, NSCLC455AAAANAQVYBRCA, UEC456IPLERPLGEVYBRCA, UEC457NAAAAANAQVYBRCA, NSCLC, UEC458TDTLIHLMUEC459KVAGERYVYBRCA, CCC, CRC, GBM, HNSCC, MEL,NSCLC, OSCAR, PACA, PRCA, SCLC,UBC460RLSSATANALYGBC461AQRMTTQLLCRC, MEL, NSCLC, RCC462QRMTTQLLLNSCLC, RCC, UEC466DLIESGQLRERUEC467MQMQERDTLGEJC, HNSCC, NHL, NSCLC, OSCAR471AQRLDPVYFCCC, CRC, GBC, GEJC, NSCLC,OSCAR, PACA, SCLC, UBC472MRLLVAPLSCLC, UEC474AADGGLRASVTLBRCA, NSCLC, OSCAR477RIQQQTNTYGBM, SCLC479TEGSHFVEABRCA, SCLC, UEC480GRADIMIDFBRCA, CRC, HNSCC, MEL, NSCLC,OSCAR, SCLC, UEC481GRWEKTDLTYBRCA, GBC, HNSCC, MEL, NSCLC,OSCAR, PACA, SCLC, UBC, UEC482GRWEKTDLTYRHNSCC, NSCLC, OSCAR, PACA, SCLC,UEC484AWLRSAAACCC485VRFPVHAALMEL, NSCLC, OSCAR486DRFFWLKVNSCLC, SCLC487GMADILVVFNSCLC488RSFSLGVPRAML, CLL, GC, HCC, HNSCC, NHL,NSCLC, PRCA, SCLC, UEC490AEVQKLLGPHNSCC, NSCLC, OSCAR, UEC491EAYSSTSSWGBC, UEC493DTNLEPVTRUEC495EVPSGATTEVSRUEC496EVPTGTTAEVSRUEC498EVYPELGTQGRUEC503TVFDKAFTAANSCLC507TSIFSGQSLUEC508TVAKTTTTFUEC509GRGPGGVSWNSCLC518TSDFPTITVPACA520THSAMTHGFNHL527QSTPYVNSVUEC528TRTGLFLRFHNSCC, NSCLC, UEC533GQHLHLETFAML, CCC, GBC, GC, HCC, MEL, NHL,NSCLC, OSCAR, RCC, SCLC, UBC,UEC537IRRLKELKDQNSCLC539IPIPSTGSVEMCCC, GC, HNSCC, NSCLC, OSCAR,PRCA, SCLC, UBC, UEC540AGIPAVALWHCC, NSCLC, OSCAR541RLSPAPLKLGBM, NSCLC544LRNPSIQKLGBM545RVGPPLLIBRCA, CRC, NSCLC, OSCAR, UEC546GRAFFAAAFCRC, GBM, HNSCC, MEL, NSCLC,OSCAR, PACA, SCLC, UBC, UEC547EVNKPGVYTRHCC, UEC549ARSKLQQGLMEL550RRFKEPWFLBRCA, HCC, MEL, NSCLC, PRCA,SCLC, UBC, UEC563PNFSGNWKIIRSENFEELNSCLC589APDAKSFVLNLGKDSNNLNSCLC590RVRGEVAPDAKSFVLNLGNSCLC591VRGEVAPDAKSFVLNLNSCLC, RCC592VRGEVAPDAKSFVLNLGNSCLC, RCC593GEVAPDAKSFVLNLGNSCLC, RCC594VRGEVAPDAKSFVLNNSCLC, RCC598AESLFREALSNKVDELNSCLC599AESLFREALSNKVDENSCLC607LTVAEVQKLLGPHVEGLKAEENSCLC608LTVAEVQKLLGPHVEGLKAENSCLC609LTVAEVQKLLGPHVEGLKANSCLC610LTVAEVQKLLGPHVEGLKNSCLC611LTVAEVQKLLGPHVEGLNSCLC612TVAEVQKLLGPHVEGLKNSCLC613LTVAEVQKLLGPHVEGNSCLC614TVAEVQKLLGPHVEGLNSCLC615VAEVQKLLGPHVEGLKNSCLC616TVAEVQKLLGPHVEGNSCLC617VAEVQKLLGPHVEGLNSCLC618VAEVQKLLGPHVEGNSCLC619VAEVQKLLGPHVENSCLC620EVQKLLGPHVEGNSCLC625DALRGLLPVLGQPIIRSIPQGNSCLC628DALRGLLPVLGQPIIRSIPQNSCLC629GLLPVLGQPIIRSIPQGIVANSCLC630ALRGLLPVLGQPIIRSIPQNSCLC633LRGLLPVLGQPIIRSIPQNSCLC634DALRGLLPVLGQPIIRSNSCLC635ALRGLLPVLGQPIIRSNSCLC637ALRGLLPVLGQPIIRNSCLC638LRGLLPVLGQPIIRSNSCLC639ALRGLLPVLGQPIINSCLC646GLLPVLGQPIIRSIPQGIVAAWRQNSCLC648GLLPVLGQPIIRSIPQGIVAANSCLC651LPVLGQPIIRSIPQGIVAANSCLC653LPVLGQPIIRSIPQGIVANSCLC654PVLGQPIIRSIPQGIVAGC, NSCLC656VLGQPIIRSIPQGIVANSCLC661LRGLLPVLGQPIIRSIPQGNSCLC666LPLTVAEVQKLLGPHVEGNSCLC668AVLPLTVAEVQKBRCA, CRC, GBC, GC, NSCLC, PACA,UEC677IWAVRPQDLDTCDPRNSCLC680GVRGSLLSEADVRALGGLANSCLC682GVRGSLLSEADVRALGGLNSCLC686VRGSLLSEADVRALGGLNSCLC694GSLLSEADVRALGGNSCLC695RGSLLSEADVRALGNSCLC697GSLLSEADVRALGNSCLC717IPQGIVAAWRQRSSRDPSGC730LPGRFVAESAEVLNSCLC Example 2 Expression Profiling of Genes Encoding the Peptides of the Invention Over-presentation or specific presentation of a peptide on tumor cells compared to normal cells is sufficient for its usefulness in immunotherapy, and some peptides are tumor-specific despite their source protein occurring also in normal tissues. Still, mRNA expression profiling adds an additional level of safety in selection of peptide targets for immunotherapies. Especially for therapeutic options with high safety risks, such as affinity-matured TCRs, the ideal target peptide will be derived from a protein that is unique to the tumor and not found on normal tissues. RNA Sources and Preparation Surgically removed tissue specimens were provided as indicated above (see Example 1) after written informed consent had been obtained from each patient. Tumor tissue specimens were snap-frozen immediately after surgery and later homogenized with mortar and pestle under liquid nitrogen. Total RNA was prepared from these samples using TRI Reagent (Ambion, Darmstadt, Germany) followed by a cleanup with RNeasy (QIAGEN, Hilden, Germany); both methods were performed according to the manufacturer's protocol. Total RNA from healthy human tissues for RNASeq experiments was obtained from: Asterand (Detroit, MI, USA & Royston, Herts, UK); Bio-Options Inc. (Brea, CA, USA); Geneticist Inc. (Glendale, CA, USA); ProteoGenex Inc. (Culver City, CA, USA); Tissue Solutions Ltd (Glasgow, UK). Total RNA from tumor tissues for RNASeq experiments was obtained from: Asterand (Detroit, MI, USA & Royston, Herts, UK); BioCat GmbH (Heidelberg, Germany); BioServe (Beltsville, MD, USA); Geneticist Inc. (Glendale, CA, USA); Istituto Nazionale Tumori “Pascale” (Naples, Italy); ProteoGenex Inc. (Culver City, CA, USA); University Hospital Heidelberg (Heidelberg, Germany). Quality and quantity of all RNA samples were assessed on an Agilent 2100 Bioanalyzer (Agilent, Waldbronn, Germany) using the RNA 6000 Pico LabChip Kit (Agilent). RNAseq Experiments Gene expression analysis of—tumor and normal tissue RNA samples was performed by next generation sequencing (RNAseq) by CeGaT (Tübingen, Germany). Briefly, sequencing libraries are prepared using the Illumina HiSeq v4 reagent kit according to the provider's protocol (Illumina Inc., San Diego, CA, USA), which includes RNA fragmentation, cDNA conversion and addition of sequencing adaptors. Libraries derived from multiple samples are mixed equimolar and sequenced on the Illumina HiSeq 2500 sequencer according to the manufacturer's instructions, generating 50 bp single end reads. Processed reads are mapped to the human genome (GRCh38) using the STAR software. Expression data are provided on transcript level as RPKM (Reads Per Kilobase per Million mapped reads, generated by the software Cufflinks) and on exon level (total reads, generated by the software Bedtools), based on annotations of the ensembl sequence database (Ensembl77). Exon reads are normalized for exon length and alignment size to obtain RPKM values. Exemplary expression profiles of source genes of the present invention that are highly over-expressed or exclusively expressed in ovarian cancer are shown inFIGS.1A through1V. Expression scores for further exemplary genes are shown in Table 10. TABLE 10Expression scores. The table lists peptides fromgenes that are very highly over-expressed in OCtumors compared to a panel of normal tissues (+++),highly over-expressed in OC tumors compared to apanel of normal tissues (++) or over-expressedin OC tumors compared to a panel of normal tissues(+). The baseline for this score was calculatedfrom measurements of the following relevant normaltissues: adipose tissue, adrenal gland, bile duct,blood cells, blood vessels, bone marrow, brain,cartilage, esophagus, eye, gallbladder, heart,head&neck, kidney, large intestine, liver, lung,lymph node, nerve, parathyroid, pancreas,pituitary, skeletal muscle, skin, small intestine,spleen, stomach, thyroid gland, trachea, urinarybladder. In case expression data for severalsamples of the same tissue type were available,the arithmetic mean of all respective samples wasused for the calculation.Seq IDNoSequenceGene Expression1MIPTFTALL++5RLLDFINVL+++6SLGKHTVAL+++10RYSDSVGRVSF+11SYSDLHYGF++12KYEKIFEML+++13VYTFLSSTL+++14FYFPTPTVL++16IYSPQFSRL+++17RFTTMLSTF+++18KYPVHIYRL+20RMASPVNVK++21AVRKPIVLK+22SLKERNPLK++23GMMKGGIRK+++25GTSPPSVEK++26RISEYLLEK+++27VLYGPAGLGK+++28KTYETNLEIKK+++29QQFLTALFY+++30ALEVAHRLK+31LLDEGAMLLY+++32SPNKGTLSV+33SPTFHLTL+34LPRGPLASLL++35FPDNQRPAL++36APAAWLRSA+++37RPLFQKSSM+++38SPHPVTALLTL++39RPAPFEVVF++40KPGTSYRVTL+++42TLKVTSAL+43ALKARTVTF+47MPNLRSVDL+++51SLRLKNVQL+++52AEFLLRIFL+53MEHPGKLLF+++54AEITITTQTGY++55HETETRTTW++56SEPDTTASW++57QESDLRLFL+++58GEMEQKQL+++59SENVTMKVV+++60GLLSLTSTLYL+61YMVHIQVTL++62KVLGVNVML++63MMEEMIFNL++64FLDPDRHFL++66GLLQELSSI+++67SLLLPSIFL+++69TTYEGSITV++70VLQGLLRSL+++71YLEDTDRNL+72YLTDLQVSL+73FLIEELLFA+++74SQSPSVSQL+++75KVVSVLYNV+++76KYVAELSLL+++77RYGPVFTV+++78SFAPRSAVF++82SYFRGFTLI+++83GTYAHTVNR+++84KLQPAQTAAK+++87AVAPPTPASK++88VVHAVFALK+89RVAELLLLH+++90KVAGERYVYK++91RSLRYYYEK++92SVFPIENIY++96TAFGGFLKY+++97TMLDVEGLFY++98LLQPPPLLAR+++100RLFTSPIMTK++101RVFTSSIKTK++102SVLTSSLVK++103TSRSVDEAY++104VLADSVTTK++107RLQEWKALK+++108VLYPVPLESY+++110SAAPPSYFR+++111TLPQFRELGY++112TVTGAEQIQY++113QLDSNRLTY++114VMEQSAGIMY+++115FVDNQYWRY+++116VLLDEGAMLLY+++117APRLLLLAVL++118SPASRSISL++119APLPRPGAVL+++120RPAMNYDKL++121VPNQSSESL+++122YPGFPQSQY+++123KPSESIYSAL+++124LPSDSHFKITF+++125VPVYILLDEM++126KPGPEDKL++128YPRTITPGM+129APRPASSL++130FPRLVGPDF+131APTEDLKAL++132IPGPAQSTI++133MPNLPSTTSL++135RVRSTISSL++136SPFSAEEANSL++137SPGATSRGTL++138SPMATTSTL++139SPQSMSNTL++140SPRTEASSAVL++141SPMTSLLTSGL++142TPGLRETSI++143SPAMTSTSF++144SPSPVSSTL++145SPSSPMSTF++147APRWFPQPTVV+++151HLLLKVLAF+++152MGSARVAEL+++154MLRKIAVAA++155NKKMMKRLM+++156HVKEKFLL++157EAMKRLSYI+159VLKHKLDEL++160YPKARLAF++161ALKTTTTAL++162QAKTHSTL++163QGLLRPVF+++164SIKTKSAEM++165SPRFKTGL++166TPKLRETSI++167TSHERLTTL++168TSHERLTTY++169TSMPRSSAM++170YLLEKSRVI+++171FAFRKEAL+++172KLKERNREL+++173AEAQVGDERDY+174AEATARLNVF+175AEIEPKADG+176AEIEPKADGSW+177TEVGTMNLF++181GEGPKTSW+183YEKGIMQKV++184AELEALTDLW++185AERQPGAASL++186REGPEEPGL++187GEAQTRIAW++189KEFLFNMY++190YEVARILNL++193LEAQQEAL++194KEVDPTSHSY+++195AEDKRHYSV+196REMPGGPVW+++197AEVLLPRLV++198QEAARAAL++199REIDESLIFY++200AESIPTVSF++201AETILTFHAF++202HESEATASW++203IEHSTQAQDTL++204RETSTSEETSL++205SEITRIEM++206SESVTSRTSY+++207TEARATSDSW++208TEVSRTEAI++209TEVSRTEL++210VEAADIFQNF++211EEKVFPSPLW+++212MEQKQLQKRF+++214VEQTRAGSLL++216IMFDDAIERA+++217VSSSLTLKV+218TIASQRLTPL++219PLPRPGAVL+++220RMTTQLLLL++225SVFAHPRKL++226QVDPKKRISM++227YTFRYPLSL++229ISVPAKTSL++230SAFREGTSL++231SVTESTHHL++232TISSLTHEL++233GSDTSSKSL++234GVATRVDAI+++235SAIETSAVL++236SAIPFSMTL++237SAMGTISIM++238PLLVLFTI+++239FAVPTGISM++240FSTDTSIVL++241RQPNILVHL++242STIPALHEI++243YASEGVKQV++244DTDSSVHVQV++246RYLAVVHAVF+247ARPPWMWVL+++248SVIQHLGY++249VYTPTLGTL++250HFPEKTTHSF++252LYQPRASEM+++254IIQHLTEQF+++255VFVSFSSLF+++256RTEEVLLTFK++257VTADHSHVF+++258GAYAHTVNR+++259KTLELRVAY+260GTNTVILEY++261HTFGLFYQR++262RSRLNPLVQR++263SSSSATISK++266ISYSGQFLVK+++267VTDLISPRK+++268GLLGLSLRY+++269RLKGDAWVYK++270AVFNPRFYRTY+++272RQPERTILRPR++273RVNAIPFTY++274KTFPASTVF++275STTFPTLTK++276VSKTTGMEF++277TTALKTTSR++278NLSSITHER++279SVSSETTKIKR++280SVSGVKTTF++281RAKELEATF+++282CLTRTGLFLRF+++285GTVNPTVGK++286TVAPPQGVVK+287RRIHTGEKPYK++288SPVTSVHGGTY+289RWEKTDLTY++290DMDEEIEAEY+++291ETIRSVGYY++292NVTMKVVSVLY+++293VPDSGATATAY+++294YPLRGSSIF+++295YPLRGSSIFGL+++296YPLRGSSI+++297TVREASGLL+298YPTEHVQF+299HPGSSALHY++301SPRRSPRISF+302RVEEVRALL+++303LPMWKVTAF+++304LPRPGAVL+++305TPWAESSTKF++306APVIFSHSA++307LPYGPGSEAAAF+++308YPEGAAYEF+++309FPQSQYPQY+++310RPNPITIIL+++311RPLFYVVSL+++312LPYFREFSM+++313KVKSDRSVF+++315SPRENFPDTL+++316EPKTATVL++320FPMSPVTSV+321SPMDTFLLI+322SPDPSKHLL+323RPMPNLRSV+++324VPYRVVGL+++326VPSEIDAAF++327SPLPVTSLI++328EPVTSSLPNF++329FPAMTESGGMIL++330FPFVTGSTEM++331FPHPEMTTSM++332FPHSEMTTL++333FPHSEMTTVM++334FPYSEVTTL++335HPDPVGPGL+++336HPKTESATPAAY++337HPVETSSAL++338HVTKTQATF++339LPAGTTGSLVF++340LPEISTRTM++341LPLDTSTTL++342LPLGTSMTF++343LPSVSGVKTTF++344LPTQTTSSL++345LPTSESLVSF++346LPWDTSTTLF++347MPLTTGSQGM++348MPNSAIPFSM++349MPSLSEAMTSF++350NPSSTTTEF++351NVLTSTPAF++352SPAETSTNM++353SPAMTTPSL++354SPLPVTSLL++355SPLVTSHIM++356SPNEFYFTV++357SPSPVPTTL++358SPSPVTSTL++359SPSTIKLTM++360SPSVSSNTY++361SPTHVTQSL++362SPVPVTSLF++363TAKTPDATF++364TPLATTQRF++365TPLATTQRFTY++366TPLTTTGSAEM++367TPSVVTEGF++368VPTPVFPTM++369FPHSEMTTV++370PGGTRQSL++372IPRNPPPTLL+++373RPRALRDLRIL+++374NPIGDTGVKF+++375AAASPLLLL+++376RPRSPAGQVA+++377RPRSPAGQVAAA+++378RPRSPAGQVAA+++379GPFPLVYVL+++380IPTYGRTF+++381LPEQTPLAF+++382SPMHDRWTF+++383TPTKETVSL+++384YPGLRGSPM+++387APLKLSRTPA+++388SPAPLKLSRTPA+++389SPGAQRTFFQL++395VLLPRLVSC++396REASGLLSL+397REGDTVQLL+399RELLHLVTL+400GEIEIHLL+403EEAQWVRKY++404NEAIMHQY++405NEIWTHSY++411LELPPILVY+412QEILTQVKQ+++413IEALSGKIEL+++416SEEETRVVF++417AEHFSMIRA+++418FEDAQGHIW++419HEFGHVLGL++420FESHSTVSA++421GEPATTVSL++422SETTFSLIF++423SEVPTGTTA++424TEFPLFSAA++425SEVPLPMAI++426PEKTTHSF++427HESSSHHDL+428LDLGLNHI++429REKFIASVI+++430DEKILYPEF+++432EEQYIAQF+++433SDSQVRAF+++435REEFVSIDHL+++436REPGDIFSEL+++437TEAVVTNEL+ Example 3 In Vitro Immunogenicity for MHC Class I Presented Peptides In order to obtain information regarding the immunogenicity of the TUMAPs of the present invention, the inventors performed investigations using an in vitro T-cell priming assay based on repeated stimulations of CD8+ T cells with artificial antigen presenting cells (aAPCs) loaded with peptide/MHC complexes and anti-CD28 antibody. This way the inventors could show immunogenicity for HLA-A*0201, HLA-A*24:02, HLA-A*01:01, HLA-A*03:01, HLA-B*07:02 and HLA-B*44:02 restricted TUMAPs of the invention, demonstrating that these peptides are T-cell epitopes against which CD8+ precursor T cells exist in humans (Table 11). In Vitro Priming of CD8+ T Cells In order to perform in vitro stimulations by artificial antigen presenting cells loaded with peptide-MHC complex (pMHC) and anti-CD28 antibody, the inventors first isolated CD8+ T cells from fresh HLA-A*02, HLA-A*24, HLA-A*01, HLA-A*03, HLA-B*07 or HLA-B*44 leukapheresis products via positive selection using CD8 microbeads (Miltenyi Biotec, Bergisch-Gladbach, Germany) of healthy donors obtained from the University clinics Mannheim, Germany, after informed consent. PBMCs and isolated CD8+ lymphocytes were incubated in T-cell medium (TCM) until use consisting of RPMI-Glutamax (Invitrogen, Karlsruhe, Germany) supplemented with 10% heat inactivated human AB serum (PAN-Biotech, Aidenbach, Germany), 100 U/ml Penicillin/100 μg/ml Streptomycin (Cambrex, Cologne, Germany), 1 mM sodium pyruvate (CC Pro, Oberdorla, Germany), 20 μg/ml Gentamycin (Cambrex). 2.5 ng/ml IL-7 (PromoCell, Heidelberg, Germany) and 10 U/ml IL-2 (Novartis Pharma, Nürnberg, Germany) were also added to the TCM at this step. Generation of pMHC/anti-CD28 coated beads, T-cell stimulations and readout was performed in a highly defined in vitro system using four different pMHC molecules per stimulation condition and 8 different pMHC molecules per readout condition. The purified co-stimulatory mouse IgG2a anti human CD28 Ab 9.3 (Jung et al., 1987) was chemically biotinylated using sulfo-N-hydroxysuccinimidobiotin as recommended by the manufacturer (Perbio, Bonn, Germany). Beads used were 5.6 μm diameter streptavidin coated polystyrene particles (Bangs Laboratories, Illinois, USA). pMHC used for positive and negative control stimulations were A*0201/MLA-001 (peptide ELAGIGILTV (SEQ ID NO. 775) from modified Melan-A/MART-1) and A*0201/DDX5-001 (YLLPAIVHI from DDX5, SEQ ID NO. 776), respectively. 800.000 beads/200 μl were coated in 96-well plates in the presence of 4×12.5 ng different biotin-pMHC, washed and 600 ng biotin anti-CD28 were added subsequently in a volume of 200 μl. Stimulations were initiated in 96-well plates by co-incubating 1×106CD8+ T cells with 2×106washed coated beads in 200 μl TCM supplemented with 5 ng/ml IL-12 (PromoCell) for 3 days at 37° C. Half of the medium was then exchanged by fresh TCM supplemented with 80 U/ml IL-2 and incubating was continued for 4 days at 37° C. This stimulation cycle was performed for a total of three times. For the pMHC multimer readout using 8 different pMHC molecules per condition, a two-dimensional combinatorial coding approach was used as previously described (Andersen et al., 2012) with minor modifications encompassing coupling to 5 different fluorochromes. Finally, multimeric analyses were performed by staining the cells with Live/dead near IR dye (Invitrogen, Karlsruhe, Germany), CD8-FITC antibody clone SK1 (BD, Heidelberg, Germany) and fluorescent pMHC multimers. For analysis, a BD LSRII SORP cytometer equipped with appropriate lasers and filters was used. Peptide specific cells were calculated as percentage of total CD8+ cells. Evaluation of multimeric analysis was done using the FlowJo software (Tree Star, Oregon, USA). In vitro priming of specific multimer+ CD8+ lymphocytes was detected by comparing to negative control stimulations. Immunogenicity for a given antigen was detected if at least one evaluable in vitro stimulated well of one healthy donor was found to contain a specific CD8+ T-cell line after in vitro stimulation (i.e. this well contained at least 1% of specific multimer+ among CD8+ T-cells and the percentage of specific multimer+ cells was at least 10× the median of the negative control stimulations). In Vitro Immunogenicity for Ovarian Cancer Peptides For tested HLA class I peptides, in vitro immunogenicity could be demonstrated by generation of peptide specific T-cell lines. Exemplary flow cytometry results after TUMAP-specific multimer staining for 14 peptides of the invention are shown inFIGS.2through9B together with corresponding negative controls. Results for 118 peptides from the invention are summarized in Table 11a and Table 11b. TABLE 11ain vitro immunogenicity of HLA class I peptidesof the invention. Exemplary results of in vitroimmunogenicity experiments conducted by theapplicant for the peptides of the invention.<20% = +; 20%-49% = ++; 50%-69% = +++;>=70% = ++++Seq IDSequenceWells positive [%]773ALYGKLLKL+++774VYVDDIYVI+++ TABLE 11bin vitro immunogenicity of HLA class I peptidesof the invention. Exemplary results of in vitroimmunogenicity experiments conducted by theapplicant for the peptides of the invention.<20% = +; 20%-49% = ++; 50%-69% = +++;>=70% = ++++Wells positiveSeq ID NoSequence[%]HLA2TLLKALLEI++A*023ALIYNLVGI++A*024ALFKAWAL++++A*025RLLDFINVL++A*027ALQAFEFRV++++A*0260GLLSLTSTLYL+A*0262KVLGVNVML++A*0264FLDPDRHFL+++A*0266GLLQELSSI+A*0267SLLLPSIFL+++A*0271YLEDTDRNL+A*0273FLIEELLFA+++A*0275KVVSVLYNV+++A*0211SYSDLHYGF+++A*2412KYEKIFEML+A*2413VYTFLSSTL+A*2416IYSPQFSRL+A*2418KYPVHIYRL+A*2479SYNEHWNYL+A*2480TAYMVSVAAF+A*2482SYFRGFTLI+A*24113QLDSNRLTY+A*01115FVDNQYWRY+A*0120RMASPVNVK+A*0321AVRKPIVLK+A*0322SLKERNPLK+A*0323GMMKGGIRK++A*0324SMYYPLQLK+A*0325GTSPPSVEK+++A*0326RISEYLLEK+A*0327VLYGPAGLGK+A*0328KTYETNLEIKK+A*0330ALEVAHRLK++A*0383GTYAHTVNR+A*0384KLQPAQTAAK+A*0385VLLGSLFSRK+A*0386VVLLGSLFSRK+A*0387AVAPPTPASK+A*0390KVAGERYVYK+++A*0391RSLRYYYEK++A*0394ATFERVLLR+A*0395QSMYYPLQLK+A*0399KVVDRWNEK++A*03100RLFTSPIMTK+A*03102SVLTSSLVK+A*03106AAFVPLLLK+++A*03109KTFTIKRFLAK+A*03110SAAPPSYFR++A*0332SPNKGTLSV+B*0733SPTFHLTL++++B*0734LPRGPLASLL+B*0735FPDNQRPAL+B*0736APAAWLRSA+++B*0737RPLFQKSSM+B*0738SPHPVTALLTL+B*0739RPAPFEVVF+++B*0740KPGTSYRVTL++++B*0741RVRSRISNL+B*07118SPASRSISL+B*07119APLPRPGAVL++B*07120RPAMNYDKL+B*07121VPNQSSESL+B*07123KPSESIYSAL++B*07124LPSDSHFKITF++B*07128YPRTITPGM+B*07129APRPASSL+B*07130FPRLVGPDF+++B*07131APTEDLKAL++B*07133MPNLPSTTSL++++B*07134RPIVPGPLL++B*07139SPQSMSNTL+B*07140SPRTEASSAVL+B*07141SPMTSLLTSGL++B*07146IPRPEVQAL+++B*07147APRWFPQPTVV++B*07148KPYGGSGPL+B*07149GPREALSRL++B*0752AEFLLRIFL+B*4453MEHPGKLLF+B*4455HETETRTTW+++B*4457QESDLRLFL+B*4458GEMEQKQL++++B*4459SENVTMKVV++B*44174AEATARLNVF+B*44175AEIEPKADG++++B*44177TEVGTMNLF++B*44178NELFRDGVNW+B*44179REAGDEFEL+B*44180REAGDEFELRY++++B*44181GEGPKTSW+B*44182KEATEAQSL+B*44183YEKGIMQKV++++B*44184AELEALTDLW+B*44186REGPEEPGL+B*44187GEAQTRIAW++B*44188AEFAKKQPWW++B*44189KEFLFNMY++++B*44190YEVARILNL++++B*44191EEDAALFKAW+++B*44192YEFKFPNRL+B*44195AEDKRHYSV+++B*44197AEVLLPRLV++B*44198QEAARAAL++B*44199REIDESLIFY+B*44200AESIPTVSF+++B*44201AETILTFHAF+++B*44202HESEATASW++B*44203IEHSTQAQDTL++++B*44205SEITRIEM++++B*44207TEARATSDSW+B*44208TEVSRTEAI+B*44209TEVSRTEL++++B*44210VEAADIFQNF+B*44211EEKVFPSPLW+++B*44212MEQKQLQKRF++B*44213KESIPRWYY+B*44 Example 4 Synthesis of Peptides All peptides were synthesized using standard and well-established solid phase peptide synthesis using the Fmoc-strategy. Identity and purity of each individual peptide have been determined by mass spectrometry and analytical RP-HPLC. The peptides were obtained as white to off-white lyophilizes (trifluoro acetate salt) in purities of >50%. All TUMAPs are preferably administered as trifluoro-acetate salts or acetate salts, other salt-forms are also possible. Example 5 MHC Binding Assays Candidate peptides for T cell based therapies according to the present invention were further tested for their MHC binding capacity (affinity). The individual peptide-MHC complexes were produced by UV-ligand exchange, where a UV-sensitive peptide is cleaved upon UV-irradiation, and exchanged with the peptide of interest as analyzed. Only peptide candidates that can effectively bind and stabilize the peptide-receptive MHC molecules prevent dissociation of the MHC complexes. To determine the yield of the exchange reaction, an ELISA was performed based on the detection of the light chain (β2m) of stabilized MHC complexes. The assay was performed as generally described in Rodenko et al. (Rodenko et al., 2006). 96 well MAXISorp plates (NUNC) were coated over night with 2 ug/ml streptavidin in PBS at room temperature, washed 4× and blocked for 1 h at 37° C. in 2% BSA containing blocking buffer. Refolded HLA-A*02:01/MLA-001 monomers served as standards, covering the range of 15-500 ng/ml. Peptide-MHC monomers of the UV-exchange reaction were diluted 100-fold in blocking buffer. Samples were incubated for 1 h at 37° C., washed four times, incubated with 2 ug/ml HRP conjugated anti-β2m for 1 h at 37° C., washed again and detected with TMB solution that is stopped with NH2SO4. Absorption was measured at 450 nm. Candidate peptides that show a high exchange yield (preferably higher than 50%, most preferred higher than 75%) are generally preferred for a generation and production of antibodies or fragments thereof, and/or T cell receptors or fragments thereof, as they show sufficient avidity to the MHC molecules and prevent dissociation of the MHC complexes. TABLE 12MHC class I binding scores. Binding ofHLA-class I restricted peptides to HLA-A*02:01was ranged by peptide exchange yield:>10% = +; >20% = ++; >50 = +++; >75% = ++++PeptideSeq ID NoSequenceexchange1MIPTFTALL+++2TLLKALLEI++++3ALIYNLVGI++++4ALFKAWAL++++5RLLDFINVL++++6SLGKHTVAL+++7ALQAFEFRV++++8YLVTKVVAV++++9VLLAGFKPPL+60GLLSLTSTLYL++++61YMVHIQVTL++++62KVLGVNVML++++63MMEEMIFNL++++64FLDPDRHFL++++66GLLQELSSI++++67SLLLPSIFL++++68KLFDTQQFL++++69TTYEGSITV++++70VLQGLLRSL++++71YLEDTDRNL++++72YLTDLQVSL++++73FLIEELLFA++++75KVVSVLYNV++++216IMFDDAIERA++++217VSSSLTLKV+219PLPRPGAVL+220RMTTQLLLL+++221SLLDLYQL++222ALMRLIGCPL++++223FAHHGRSL+224SLPRFQVTL++++225SVFAHPRKL+++227YTFRYPLSL+++228RLWDWVPLA++++229ISVPAKTSL+231SVTESTHHL+++232TISSLTHEL++++234GVATRVDAI++236SAIPFSMTL+++241RQPNILVHL++242STIPALHEI+++243YASEGVKQV+++244DTDSSVHVQV+ TABLE 13MHC class I binding scores. Binding ofHLA-class I restricted peptides to HLA-A*24:02was ranged by peptide exchange yield:>10% = +; >20% = ++; >50 = +++; >75% = ++++Seq IDPeptideNoSequenceexchange10RYSDSVGRVSF++++11SYSDLHYGF++++12KYEKIFEML++++13VYTFLSSTL++++14FYFPTPTVL++++15VYHDDKQPTF++++16IYSPQFSRL++++17RFTTMLSTF++++18KYPVHIYRL++++19KYVKVFHQF++++76KYVAELSLL++++77RYGPVFTV++++78SFAPRSAVF++++79SYNEHWNYL++++80TAYMVSVAAF+++81VYNHTTRPL++++82SYFRGFTLI++++246RYLAVVHAVF++++249VYTPTLGTL++++252LYQPRASEM+++255VFVSFSSLF+++ TABLE 14MHC class I binding scores. Binding ofHLA-class I restricted peptides to HLA-A*01:01was ranged by peptide exchange yield:>10% = +; >20% = ++; >50 = +++; >75% = ++++Seq IDPeptideNoSequenceexchange31LLDEGAMLLY++++112TVTGAEQIQY++113QLDSNRLTY+++114VMEQSAGIMY++115FVDNQYWRY+++116VLLDEGAMLLY++288SPVTSVHGGTY++289RWEKTDLTY++290DMDEEIEAEY++291ETIRSVGYY+++292NVTMKVVSVLY+++ TABLE 15MHC class I binding scores. Binding ofHLA-class I restricted peptides to HLA-A*03:01was ranged by peptide exchange yield:>10% = +; >20% = ++; >50 = +++; >75% = ++++Seq IDPeptideNoSequenceexchange20RMASPVNVK++++21AVRKPIVLK+++22SLKERNPLK++23GMMKGGIRK+++24SMYYPLQLK+++25GTSPPSVEK++26RISEYLLEK++27VLYGPAGLGK+++28KTYETNLEIKK+++30ALEVAHRLK++83GTYAHTVNR+++84KLQPAQTAAK++85VLLGSLFSRK++86VVLLGSLFSRK++87AVAPPTPASK++88VVHAVFALK+++89RVAELLLLH++90KVAGERYVYK+++91RSLRYYYEK++93KILEEHTNK++94ATFERVLLR+++95QSMYYPLQLK++98LLQPPPLLAR++99KVVDRWNEK++100RLFTSPIMTK+++101RVFTSSIKTK++102SVLTSSLVK++104VLADSVTTK++105RLFSWLVNR+++106AAFVPLLLK++107RLQEWKALK+++109KTFTIKRFLAK++110SAAPPSYFR++256RTEEVLLTFK++257VTADHSHVF+258GAYAHTVNR+++259KTLELRVAY++260GTNTVILEY+++261HTFGLFYQR++262RSRLNPLVQR++263SSSSATISK++264AIKVIPTVFK++265QIHDHVNPK++266ISYSGQFLVK+++267VTDLISPRK++269RLKGDAWVYK+++270AVFNPRFYRTY++271RMFADDLHNLNK+++272RQPERTILRPR++273RVNAIPFTY+++274KTFPASTVF+275STTFPTLTK++276VSKTTGMEF+277TTALKTTSR+278NLSSITHER++279SVSSETTKIKR++280SVSGVKTTF++281RAKELEATF+283IVQEPTEEK++284KSLIKSWKK++285GTVNPTVGK++286TVAPPQGVVK++287RRIHTGEKPYK++ TABLE 16MHC class I binding scores. Binding ofHLA-class I restricted peptides to HLA-B*07:02was ranged by peptide exchange yield:>10% = +; >20% = ++; >50 = +++; >75% = ++++PeptideSeq ID NoSequenceexchange32SPNKGTLSV“+++”33SPTFHLTL“+++”34LPRGPLASLL“+++”35FPDNQRPAL“+++”36APAAWLRSA“++”37RPLFQKSSM“+++”38SPHPVTALLTL“+++”39RPAPFEVVF“+++”40KPGTSYRVTL“+++”41RVRSRISNL“+++”118SPASRSISL“+++”119APLPRPGAVL“+++”120RPAMNYDKL“++”121VPNQSSESL“+++”122YPGFPQSQY“++”123KPSESIYSAL“+++”124LPSDSHFKITF“+++”125VPVYILLDEM“++”126KPGPEDKL“++”127APRAGSQVV“+++”128YPRTITPGM“+++”129APRPASSL“+++”130FPRLVGPDF“+++”131APTEDLKAL“+++”132IPGPAQSTI“++”133MPNLPSTTSL“+++”134RPIVPGPLL“+++”135RVRSTISSL“+++”136SPFSAEEANSL“+++”137SPGATSRGTL“+++”138SPMATTSTL“+++”139SPQSMSNTL“+++”140SPRTEASSAVL“+++”141SPMTSLLTSGL“+++”142TPGLRETSI“++”143SPAMTSTSF“++”144SPSPVSSTL“+++”145SPSSPMSTF“++”146IPRPEVQAL“+++”147APRWFPQPTVV“+++”148KPYGGSGPL“+++”149GPREALSRL“+++”293VPDSGATATAY“++”294YPLRGSSIF“+++”295YPLRGSSIFGL“+++”296YPLRGSSI“++”297TVREASGLL“+++”298YPTEHVQF“++”299HPGSSALHY“++”300IPMAAVKQAL“+++”301SPRRSPRISF“++”302RVEEVRALL“+++”303LPMWKVTAF“+++”304LPRPGAVL“+++”305TPWAESSTKF“++”306APVIFSHSA“++”307LPYGPGSEAAAF“+++”308YPEGAAYEF“++”309FPQSQYPQY“++++”311RPLFYVVSL“++”312LPYFREFSM“+++”313KVKSDRSVF“+”314VPDQPHPEI“+++”315SPRENFPDTL“+++”316EPKTATVL“++”317FPFQPGSV“+++”318FPNRLNLEA“+++”319SPAEPSVYATL“++++”320FPMSPVTSV“+++”321SPMDTFLLI“++”322SPDPSKHLL“++”323RPMPNLRSV“+++”324VPYRVVGL“++”325GPRNAQRVL“+++”326VPSEIDAAF“++”327SPLPVTSLI“+++”328EPVTSSLPNF“++”329FPAMTESGGMIL“+++”330FPFVTGSTEM“++”331FPHPEMTTSM“+++”332FPHSEMTTL“+++”333FPHSEMTTVM“+++”334FPYSEVTTL“+++”335HPDPVGPGL“++”336HPKTESATPAAY“++”337HPVETSSAL“+++”338HVTKTQATF“++”339LPAGTTGSLVF“+++”340LPEISTRTM“++”341LPLDTSTTL“+++”342LPLGTSMTF“+++”343LPSVSGVKTTF“++”344LPTQTTSSL“+++”345LPTSESLVSF“++”346LPWDTSTTLF“+++”347MPLTTGSQGM“++”348MPNSAIPFSM“+++”349MPSLSEAMTSF“+++”350NPSSTTTEF“+++”351NVLTSTPAF“++”352SPAETSTNM“+++”353SPAMTTPSL“+++”354SPLPVTSLL“+++”355SPLVTSHIM“+++”356SPNEFYFTV“+++”357SPSPVPTTL“+++”358SPSPVTSTL“+++”359SPSTIKLTM“+++”360SPSVSSNTY“++”361SPTHVTQSL“+++”362SPVPVTSLF“+++”363TAKTPDATF“++”364TPLATTQRF“++”365TPLATTQRFTY“++”367TPSVVTEGF“++”368VPTPVFPTM“++”369FPHSEMTTV“+++”370PGGTRQSL“+”371LYVDGFTHW“++”372IPRNPPPTLL“+++”373RPRALRDLRIL“+++”374NPIGDTGVKF“+++”375AAASPLLLL“++”376RPRSPAGQVA“+++”377RPRSPAGQVAAA“+++”378RPRSPAGQVAA“+++”379GPFPLVYVL“+++”380IPTYGRTF“+++”381LPEQTPLAF“++”382SPMHDRWTF“+++”383TPTKETVSL“+++”384YPGLRGSPM“++++”385SPALHIGSV“+++”386FPFNPLDF“++”387APLKLSRTPA“+++”388SPAPLKLSRTPA“++”389SPGAQRTFFQL“+++”390NPDLRRNVL“+++”391APSTPRITTF“+++”392KPIESTLVA“+++” TABLE 17MHC class I binding scores. Binding ofHLA-class I restricted peptides to HLA-B*44:02was measured by peptide exchange yield:>10% = +; >20% = ++; >50 =+++; >75% = ++++PeptideSeq ID NoSequenceexchange52AEFLLRIFL“++”53MEHPGKLLF“++++”54AEITITTQTGY“+++”55HETETRTTW“+++”56SEPDTTASW“+++”57QESDLRLFL“+++”58GEMEQKQL“++”59SENVTMKVV“+++”173AEAQVGDERDY“+++”174AEATARLNVF“++++”175AEIEPKADG“++”176AEIEPKADGSW“+++”177TEVGTMNLF“+++”178NELFRDGVNW“+++”179REAGDEFEL“++”180REAGDEFELRY“++”181GEGPKTSW“++”182KEATEAQSL“+++”183YEKGIMQKV“++”184AELEALTDLW“+++”185AERQPGAASL“++”186REGPEEPGL“++”187GEAQTRIAW“+++”188AEFAKKQPWW“+++”189KEFLFNMY“++”190YEVARILNL“++”191EEDAALFKAW“+++”192YEFKFPNRL“+++”193LEAQQEAL“++”194KEVDPTSHSY“++”195AEDKRHYSV“++”196REMPGGPVW“+++”197AEVLLPRLV“+++”198QEAARAAL“++”199REIDESLIFY“+++”200AESIPTVSF“+++”201AETILTFHAF“+++”202HESEATASW“+++”203IEHSTQAQDTL“++”204RETSTSEETSL“+++”205SEITRIEM“++”206SESVTSRTSY“+++”207TEARATSDSW“+++”208TEVSRTEAI“++”209TEVSRTEL“++”210VEAADIFQNF“+++”211EEKVFPSPLW“+++”212MEQKQLQKRF“+++”213KESIPRWYY“++”214VEQTRAGSLL“++”215SEDGLPEGIHL“++”396REASGLLSL“+++”397REGDTVQLL“++”398SFEQVVNELF“++”399RELLHLVTL“+++”400GEIEIHLL“+”402RELANDELIL“++”403EEAQWVRKY“++”404NEAIMHQY“++”405NEIWTHSY“+”406EDGRLVIEF“+”407AEHEGVSVL“++”408LEKALQVF“++”409REFVLSKGDAGL“+++”410SEDPSKLEA“+”411LELPPILVY“+”412QEILTQVKQ“++”413lEALSGKIEL“++”414EDAALFKAW“++”415REEDAALFKAW“+++”416SEEETRVVF“+++”417AEHFSMIRA“++”418FEDAQGHIW“+++”419HEFGHVLGL“++”420FESHSTVSA“+”421GEPATTVSL“++”422SETTFSLIF“+++”423SEVPTGTTA“++”424TEFPLFSAA“+”425SEVPLPMAI“+++”426PEKTTHSF“+”427HESSSHHDL“++”429REKFIASVI“++”431AEQDPDELNKA“++”432EEQYIAQF“+”433SDSQVRAF“+”434KEAIREHQM“++”435REEFVSIDHL“++”436REPGDIFSEL“++”437TEAVVTNEL“++”438SEVDSPNVL“+++” Example 6 Stability of Peptide-MHC Class I Complexes Peptide-MHC stability assays for HLA-B*08:01 peptides were performed. The data were obtained using a proximity based, homogenous, real-time assay in order to measure the dissociation of peptides from HLA class I molecules. First, human recombinant HLA-B*08:01 and b2m were expressed inE. coliand purified in a series of liquid chromatography based steps (Ferre et al., 2003; Ostergaard et al., 2001). Then, the stability of a peptide-MHC complex (pMHC) was determined by measuring the amount of b2m associated with the MHC heavy chain over time at 37° C. (Harndahl et al., 2012). The stability of each pMHC, expressed as the half life of b2m associated with the respective heavy chain, was calculated by fitting the data to a one-phase dissociation equation. The pMHC stabilities were measured in three independent experiments with the peptides in question, and for HLA-B*08:01 were found to span the range from weak-binders (+) to very stable binders (++++). The mean half-life (T½) is shown in Table 18. TABLE 18Mean half-life (T½) based on three individualmeasurements. T½ > 2 h = +; T½ > 4 h = ++;T½ > 6 h = +++; T½ > 10 h = ++++Seq IDMean Half-lifeNoSequence(T½)43ALKARTVTF+++44LNKQKVTF++++45VGREKKLAL++46DMKKAKEQL+47MPNLRSVDL++48DVKKKIKEV+49LPRLKAFMI++50DMKYKNRV+51SLRLKNVQL+150MAAVKQAL++151HLLLKVLAF++152MGSARVAEL++153NAMLRKVAV+154MLRKIAVAA+156HVKEKFLL++157EAMKRLSYI+158LPKLAGLL+159VLKHKLDEL+160YPKARLAF+++161ALKTTTTAL+162QAKTHSTL+163QGLLRPVF++164SIKTKSAEM+++166TPKLRETSI++167TSHERLTTL++169TSMPRSSAM+++170YLLEKSRVI++171FAFRKEAL++172KLKERNREL+++394MYKMKKPI+395VLLPRLVSC+ Example 7 Binding Scores of Selected Peptides for HLA Class II Allotypes Major histocompatibility complex class II (MHC-II) molecules are predominantly expressed on the surface of professional antigen presenting cells, where they display peptides to T helper cells, which orchestrate the onset and outcome of many host immune responses. Understanding which peptides will be presented by the MHC-II molecule is therefore important for understanding the activation of T helper cells and can be used to identify T-cell epitopes. Peptides presented by the MHC class II molecule bind to a binding groove formed by residues of the MHC α- and β-chain. The peptide-MHC binding affinity is primarily determined by the amino acid sequence of the peptide-binding core. HLA class II binding prediction algorithms are only available for the most important class II alleles and have been tested using the SYFPEITHI algorithm (Rammensee et al., 1999). The algorithm has already been successfully used to identify class I and class II epitopes from a wide range of antigens, e.g. from the human tumor-associated antigens TRP2 (class I) (Sun et al., 2000) and SSX2 (class II) (Neumann et al., 2004). Table 20 shows the HLA class II allotypes which are likely to bind the selected peptides. The peptide was considered as binding to an HLA molecule if the SYFPEITHI score was equal to or higher than 18. TABLE 20Binding of the class II peptides to various HLA class II allotypes. Based on theprediction by the SYFPEITHI algorithm, the selected peptides are likelyto bind to at least 4 of the HLA class II allotpyes with known binding motif. Listedare all HLA class II alleles for which a SYFPEITHI prediction matrix is available.No of HLASeqBest HLA class IIClass IIID NoSequencebindersHLA Class II bindersbinder552GVNAMLRKVAVAAASKPHDRB1*11:04DQA1*05:01/DQB1*02:01 (DQ2),15VEQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*13:01, DRB1*13:02,DRB1*15:01560PNFSGNWKIIRSENFEELLDRB1*07:01DQA1*05:01/DQB1*02:01 (DQ2),14KDRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:02, DRB1*08:03, DRB1*09:01,DRB1*11:01, DRB1*13:02, DRB1*15:01,DRB1*15:02574LPDFYNDWMFIAKHLPDLDRB1*11:01DQA1*05:01/DQB1*02:01 (DQ2),15DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*08:02, DRB1*08:03,DRB1*11:01, DRB1*11:04, DRB1*13:02,DRB1*15:01, DRB1*15:02575VGDDHLLLLQGEQLRRTDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),8DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*09:01, DRB1*15:01,DRB1*15:02579SGGPLVCDETLQGILSDQA1*0501/DQB1*0201DQA1*05:01/DQB1*02:01 (DQ2),8(DQ2)DQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*15:01582GSQPWQVSLFNGLSFHDRB1*15:01DQA1*05:01/DQB1*02:01 (DQ2),10DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:04, DRB1*07:01,DRB1*09:01, DRB1*15:01, DRB1*15:02583LTVKLPDGYEFKFPNRLNLDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),14EAINY(DQ2)DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:02, DRB1*08:03, DRB1*09:01,DRB1*11:01, DRB1*13:02, DRB1*15:01,DRB1*15:02587DQANLTVKLPDGYEFKFPDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),13NRLNL(DQ2)DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:02, DRB1*08:03, DRB1*11:01,DRB1*13:02, DRB1*15:01, DRB1*15:02588VAPDAKSFVLNLGKDSNNDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),16LDQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*08:02, DRB1*08:03,DRB1*09:01, DRB1*11:01, DRB1*11:04,DRB1*13:01, DRB1*13:02590RVRGEVAPDAKSFVLNLGDRB1*03:01DQA1*05:01/DQB1*02:01 (DQ2),10DQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*07:01, DRB1*09:01,DRB1*11:04, DRB1*15:01596MAADGDFKIKCVAFDDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),10(DQ2);DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*03:01;DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*07:01DRB1*08:03, DRB1*09:01, DRB1*15:01597SPDAESLFREALSNKVDELDRB1*07:01DQA1*05:01/DQB1*02:01 (DQ2),8DQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*04:01, DRB1*04:04,DRB1*04:05, DRB1*07:01, DRB1*09:01601LSNKVDELAHFLLRKDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),14(DQ2)DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*08:02, DRB1*08:03,DRB1*11:01, DRB1*11:04, DRB1*15:01,DRB1*15:02604KLITQDLVKLKYLEYRQDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),9(DQ2)DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*08:02, DRB1*13:01,DRB1*13:02, DRB1*15:01605LTVAEVQKLLGPHVEGLKADRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),15EERHRPDQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*13:01, DRB1*15:01,DRB1*15:02622MDALRGLLPVLGQPIIRSIPDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),15QGIVADQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:02, DRB1*04:04,DRB1*04:05, DRB1*07:01, DRB1*09:01,DRB1*11:01, DRB1*11:04, DRB1*15:01,DRB1*15:02645RGLLPVLGQPIIRSIPQGIVDRB1*01:01;DQA1*05:01/DQB1*02:01 (DQ2),14AAWRQDRB1*09:01DQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:02, DRB1*04:04,DRB1*07:01, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*15:01, DRB1*15:02658VSTMDALRGLLPVLGQPIIDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),14RSIPQGDQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:02, DRB1*04:04,DRB1*04:05, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*15:01, DRB1*15:02662LRTDAVLPLTVAEVQKLLGDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),15PHVEGDQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*13:01, DRB1*15:01,DRB1*15:02669VLPLTVAEVQKLLGPHVEGDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),15LKAEEDQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*13:01, DRB1*15:01,DRB1*15:02672LRGLLPVLGQPIIRSIPQGIDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),14VAADQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:02, DRB1*04:04,,DRB1*07:01, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*15:01, DRB1*15:02673IPFTYEQLDVLKHKLDELYDRB1*08:03DQA1*05:01/DQB1*02:01 (DQ2),15PQDRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:02, DRB1*08:03, DRB1*09:01,DRB1*11:01, DRB1*11:04, DRB1*13:02,DRB1*15:01, DRB1*15:02676VPPSSIWAVRPQDLDTCDDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),10PR(DQ2)DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:03, DRB1*09:01, DRB1*15:01679WGVRGSLLSEADVRALGGDRB1*09:01DQA1*05:01/DQB1*02:01 (DQ2),12LADQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*09:01, DRB1*11:01,DRB1*15:01706LSTERVRELAVALAQKNVKDQA1*05:01/DQB1*03:01DQA1*05:01/DQB1*02:01 (DQ2),15(DQ7)DQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:02, DRB1*04:04,DRB1*04:05, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*13:01, DRB1*15:01,DRB1*15:02714AIPFTYEQLDVLKHKLDEDRB1*08:03DQA1*05:01/DQB1*02:01 (DQ2),15DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:02, DRB1*08:03, DRB1*09:01,DRB1*11:01, DRB1*11:04, DRB1*13:02,DRB1*15:01, DRB1*15:02715GLSTERVRELAVALAQKNDQA1*05:01/DQB1*03:01DQA1*05:01/DQB1*02:01 (DQ2),15(DQ7)DQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:02, DRB1*04:04,DRB1*04:05, DRB1*09:01, DRB1*11:01,DRB1*11:04, DRB1*13:01, DRB1*15:01,DRB1*15:02717IPQGIVAAWRQRSSRDPSDRB1*11:04DQA1*05:01/DQB1*02:01 (DQ2),13DQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*11:01, DRB1*11:04,DRB1*13:01, DRB1*15:01720ALGGLACDLPGRFVAESDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),8(DQ2)DQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:05, DRB1*11:01, DRB1*11:04721RELAVALAQKNVKLSTEDQA1*05:01/DQB1*03:01DQA1*05:01/DQB1*03:01 (DQ7),11(DQ7)DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:04, DRB1*04:05,DRB1*09:01, DRB1*11:04, DRB1*13:01,DRB1*15:01722LKALLEVNKGHEMSPQDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),13(DQ2);DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*01:01;DRB1*04:01, DRB1*04:02, DRB1*04:04,DRB1*04:05;DRB1*04:05, DRB1*08:03, DRB1*11:01,DRB1*08:03;DRB1*11:04, DRB1*13:01, DRB1*15:01DRB1*11:04723TFMKLRTDAVLPLTVADRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),13DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:02, DRB1*08:03, DRB1*09:01,DRB1*13:02, DRB1*15:01, DRB1*15:02727TLGLGLQGGIPNGYLVDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),9(DQ2);DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*01:01;DRB1*04:02, DRB1*04:04, DRB1*07:01,DRB1*15:01DRB1*09:01, DRB1*15:01728DLPGRFVAESAEVLLDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),12(DQ2)DQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*03:01,DRB1*04:01, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*09:01, DRB1*15:01,DRB1*15:02732ERHRPVRDWILRQRQDRB1*15:01DQA1*05:01/DQB1*02:01 (DQ2),4DRB1*04:01, DRB1*04:04, DRB1*15:01733SPRQLLGFPCAEVSGDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),8DQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*04:01,DRB1*04:04, DRB1*15:01, DRB1*15:02734SRTLAGETGQEAAPLDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),6DRB1*01:01, DRB1*04:01, DRB1*04:04,DRB1*04:05, DRB1*15:01735VTSLETLKALLEVNKDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),15DQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*08:03, DRB1*11:01,DRB1*11:04, DRB1*13:01, DRB1*15:01,DRB1*15:02745WELSQLTNSVTELGPYTLDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),13DRD(DQ2);DQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*09:01, DRB1*13:01,DRB1*15:01, DRB1*15:02746EITITTQTGYSLATSQVTLPDRB1*01:01DQA1*05:01/DQB1*02:01 (DQ2),10DQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*09:01, DRB1*15:01747ATTPSWVETHSIVIQGFPHDRB1*07:01DQA1*05:01/DQB1*02:01 (DQ2),9DQA1*05:01/DQB1*03:01 (DQ7),DRB1*01:01, DRB1*04:01, DRB1*04:04,DRB1*04:05, DRB1*07:01, DRB1*15:01,DRB1*15:02748GIKELGPYTLDRNSLYVNGDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),13(DQ2);DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*01:01DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*08:02, DRB1*09:01,DRB1*13:02, DRB1*15:01, DRB1*15:02755IELGPYLLDRGSLYVNGDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),14(DQ2)DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*08:02, DRB1*09:01,DRB1*11:04, DRB1*13:02, DRB1*15:01,DRB1*15:02759EELGPYTLDRNSLYVNGDRB1*03:01DQA1*05:01/DQB1*02:01 (DQ2),12DRB1*03:01, DRB1*04:01, DRB1*04:02,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:02, DRB1*09:01, DRB1*13:02,DRB1*15:01, DRB1*15:02760LKPLFKSTSVGPLYSGDRB1*11:04DQA1*05:01/DQB1*02:01 (DQ2),16DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*08:02, DRB1*08:03,DRB1*09:01, DRB1*11:01, DRB1*11:04,DRB1*13:01, DRB1*13:02, DRB1*15:01764FDKAFTAATTEVSRTEDQA1*05:01/DQB1*03:01DQA1*05:01/DQB1*03:01 (DQ7),9(DQ7)DRB1*01:01, DRB1*04:01, DRB1*04:04,DRB1*04:05, DRB1*07:01, DRB1*09:01,DRB1*11:01, DRB1*15:01765ELGPYTLDRDSLYVNDQA1*05:01/DQB1*02:01DQA1*05:01/DQB1*02:01 (DQ2),11(DQ2)DRB1*03:01, DRB1*04:01, DRB1*04:02,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*08:02, DRB1*13:02, DRB1*15:01,DRB1*15:02766GLLKPLFKSTSVGPLDRB1*11:04DQA1*05:01/DQB1*02:01 (DQ2),16DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:02, DRB1*04:04, DRB1*04:05,DRB1*07:01, DRB1*08:02, DRB1*08:03,DRB1*09:01, DRB1*11:01, DRB1*11:04,DRB1*13:01, DRB1*13:02, DRB1*15:01768SDPYKATSAVVITSTDQA1*05:01/DQB1*03:01DQA1*05:01/DQB1*02:01 (DQ2),10(DQ7)DQA1*05:01/DQB1*03:01 (DQ7),DQB1*06:02, DRB1*01:01, DRB1*04:01,DRB1*04:04, DRB1*07:01, DRB1*09:01,DRB1*15:01, DRB1*15:02770SRKFNTMESVLQGLLDRB1*09:01DQA1*05:01/DQB1*03:01 (DQ7),10DRB1*01:01, DRB1*03:01, DRB1*04:01,DRB1*04:04, DRB1*04:05, DRB1*07:01,DRB1*09:01, DRB1*13:02, DRB1*15:01 REFERENCE LIST The Molecular Taxonomy of Primary Prostate Cancer. Cell 163 (2015): 1011-1025Abba, M. C. et al., Mol. Cancer Res 5 (2007): 881-890Abdelmalak, C. A. et al., Clin Lab 60 (2014): 55-61Abe, A. et al., Genes Chromosomes. Cancer 55 (2016): 242-250Aghajanova, L. et al., Hum. Reprod. 30 (2015): 232-238Agherbi, H. et al., PLoS. One. 4 (2009): e5622Aguilo, F. et al., Curr. Top. Microbiol. Immunol. 394 (2016): 29-39Akagi, T. et al., J Biol Chem 290 (2015): 22460-22473Al, Zeyadi M. et al., Biotechnol. Biotechnol. Equip. 29 (2015): 111-118Albergaria, A. et al., Int. J Dev. Biol 55 (2011): 811-822Alentorn, A. et al., Presse Med 42 (2013): 806-813Alhosin, M. et al., J Exp. Clin Cancer Res 35 (2016): 174Allison, J. P. et al., Science 270 (1995): 932-933Alrawi, S. J. et al., Anticancer Res 26 (2006): 107-119Alvarado-Ruiz, L. et al., Asian Pac. J Cancer Prev. 17 (2016): 1037-1047Alzrigat, M. et al., Oncotarget. (2016)Amsterdam, A. et al., Acta Histochem. 116 (2014): 781-787Andersen, R. S. et al., Nat. Protoc. 7 (2012): 891-902Andrade, V. C. et al., Cancer Immun. 8 (2008): 2Andrade, V. C. et al., Exp. Hematol. 37 (2009): 446-449Angelopoulou, K. et al., Mamm. Genome 21 (2010): 516-524Angulo, J. C. et al., J Urol. 195 (2016): 619-626Ansari, K. I. et al., J Mol. Endocrinol. 48 (2012): 61-75Appay, V. et al., Eur. J Immunol. 36 (2006): 1805-1814Aprelikova, O. et al., Cancer Res 69 (2009): 616-624Arsenic, R. et al., BMC. Cancer 15 (2015): 784Askew, E. B. et al., J Biol Chem 284 (2009): 34793-34808Aung, P. P. et al., Oncogene 25 (2006): 2546-2557Avasarala, S. et al., Biol Open. 2 (2013): 675-685Avgustinova, A. et al., Nat Commun. 7 (2016): 10305Aytes, A. et al., Proc. Natl. Acad. Sci. U.S.A. 110 (2013): E3506-E3515Bahnassy, A. A. et al., World J Gastroenterol. 20 (2014): 18240-18248Bailey, V. J. et al., Methods 52 (2010): 237-241Balaz, P. et al., Ann. Surg. 235 (2002): 519-527Banchereau, J. et al., Cell 106 (2001): 271-274Band, A. M. et al., J Mammary. Gland. Biol Neoplasia. 16 (2011): 109-115Bao, L. et al., Cell Biol Toxicol. 32 (2016): 419-435Baroy, T. et al., Mol. Cancer 13 (2014): 93Bartolini, A. et al., Clin Cancer Res 22 (2016): 4923-4933Baty, F. et al., J Biomed. Inform. 58 (2015): 175-185Beard, R. E. et al., Clin Cancer Res 19 (2013): 4941-4950Beatty, G. et al., J Immunol 166 (2001): 2276-2282Beggs, J. D., Nature 275 (1978): 104-109Beke, L. et al., Biosci. Rep. 35 (2015)Bell, J. L. et al., J Clin Oncol 33 (2015): 1285-1293Bell, J. L. et al., Cell Mol Life Sci. 70 (2013): 2657-2675Benz, C. C. et al., Oncogene 15 (1997): 1513-1525Bergamini, A. et al., Expert. Opin. Investig. Drugs 25 (2016): 1405-1412Berger, C. et al., Curr. Mol. Med. 13 (2013): 1229-1240Bernstein, M. B. et al., Cancer Biother. Radiopharm. 29 (2014): 153-161Beyranvand, Nejad E. et al., Cancer Res 76 (2016): 6017-6029Bhan, S. et al., Oncol Rep. 28 (2012): 1498-1502Bierkens, M. et al., Genes Chromosomes. Cancer 52 (2013): 56-68Bikkavilli, R. K. et al., Oncogene 34 (2015): 5317-5328Bisig, B. et al., Best. Pract. Res Clin Haematol. 25 (2012): 13-28Bode, P. K. et al., Mod. Pathol. 27 (2014): 899-905Boeva, V. et al., PLoS. One. 8 (2013): e72182Bogush, T. A. et al., Antibiot. Khimioter. 54 (2009): 41-49Bonitsis, N. et al., Exp. Oncol 28 (2006): 187-193Borgono, C. A. et al., Cancer Res 63 (2003): 9032-9041Borgono, C. A. et al., Clin Cancer Res 12 (2006): 1487-1493Borgono, C. A. et al., J Biol Chem 282 (2007): 2405-2422Borowicz, S. et al., J Vis. Exp. (2014): e51998Boulter, J. M. et al., Protein Eng 16 (2003): 707-711Braumuller, H. et al., Nature (2013)Brinkmann, U. et al., Proc. Natl. Acad. Sci. U.S.A. 95 (1998): 10757-10762Brossart, P. et al., Blood 90 (1997): 1594-1599Bruckdorfer, T. et al., Curr. Pharm. Biotechnol. 5 (2004): 29-43Bruey, J. M. et al., J Biol Chem 279 (2004): 51897-51907Bryan, R. T., Philos. Trans. R Soc. Lond B Biol Sci. 370 (2015): 20140042Bryan, R. T. et al., J Urol. 184 (2010): 423-431Buchet-Poyau, K. et al., Nucleic Acids Res 35 (2007): 1289-1300Bundela, S. et al., PLoS. One. 9 (2014): e102610Burnett, R. M. et al., Oncotarget. 6 (2015): 12682-12696Cano, A. et al., Future. Oncol 8 (2012): 1095-1108Caputi, F. F. et al., J Mol. Neurosci. 51 (2013): 532-538Card, K. F. et al., Cancer Immunol Immunother. 53 (2004): 345-357Carrera, M. et al., Int. J Clin Exp. Pathol. 8 (2015): 3613-3623Cerveira, N. et al., BMC. Cancer 10 (2010): 518Chakrabarty, S. et al., J Cell Physiol 186 (2001): 47-52Chan, M. H. et al., Pediatr. Blood Cancer 59 (2012): 1173-1179Chandra, V. et al., Cell Death. Dis. 5 (2014): e1380Chang, H. et al., Biochem. Biophys. Res Commun. 464 (2015a): 45-50Chang, Q. et al., Oncotarget. 6 (2015b): 42838-42853Chen, H. S. et al., Zhonghua Gan Zang. Bing. Za Zhi. 11 (2003): 145-148Chen, S. T. et al., Cancer Sci. 102 (2011a): 2191-2198Chen, Y. et al., Cancer Biol Ther. 11 (2011 b): 497-511Chen, Y. et al., Onco. Targets. Ther. 7 (2014): 1465-1472Chen, Y. C. et al., Taiwan. J Obstet. Gynecol. 54 (2015): 572-579Chen, Y. L. et al., Int J Surg. 11 (2013): 85-91Chen, Y. T. et al., Int. J Cancer 124 (2009): 2893-2898Chen, Y. T. et al., Proc. Natl. Acad. Sci. U.S.A. 102 (2005): 7940-7945Cheng, Y. C. et al., J Neurochem. 110 (2009): 947-955Cheung, A. et al., Oncotarget. 7 (2016): 52553-52574Chevillard, G. et al., Blood 117 (2011): 2005-2008Choi, M. R. et al., APMIS 123 (2015): 65-71Choijamts, B. et al., Stem Cells 29 (2011): 1485-1495Chung, H. et al., Biol Chem 393 (2012a): 413-420Chung, S. et al., Oncotarget. 3 (2012b): 1629-1640Ciruelos Gil, E. M., Cancer Treat. Rev 40 (2014): 862-871Clancy, A. A. et al., Ann. Oncol 27 (2016): 1696-1705Clermont, P. L. et al., Clin Epigenetics. 8 (2016): 16Clermont, P. L. et al., Clin Epigenetics. 7 (2015): 40Clermont, P. L. et al., Br. J Cancer 111 (2014): 1663-1672Cohen, C. J. et al., J Mol Recognit. 16 (2003a): 324-332Cohen, C. J. et al., J Immunol 170 (2003b): 4349-4361Cohen, S. N. et al., Proc. Natl. Acad. Sci. U.S.A. 69 (1972): 2110-2114Colas, E. et al., Clin Transl. Oncol 14 (2012): 715-720Coligan, J. E. et al., Current Protocols in Protein Science (1995)Colombetti, S. et al., J Immunol. 176 (2006): 2730-2738Colovai, A. I. et al., Cytometry B Clin Cytom. 72 (2007): 354-362Cortesini, R., JOP. 8 (2007): 697-703Coulie, P. G. et al., Immunol. Rev 188 (2002): 33-42Coutte, L. et al., Gene 240 (1999): 201-207Cui, X. P. et al., Dig. Dis. Sci. 59 (2014): 1442-1451Curran, K. J. et al., Mol. Ther. 23 (2015): 769-778Dai, W. et al., Am. J Cancer Res 5 (2015): 2697-2707Dalerba, P. et al., Int. J Cancer 93 (2001): 85-90Dannenmann, S. R. et al., Cancer Immunol. Res. 1 (2013): 288-295Darda, L. et al., PLoS. One. 10 (2015): e0122285Darling, M. R. et al., Head Neck Pathol. 2 (2008): 169-174Dat, le T. et al., Int. J Oncol 40 (2012): 1455-1469Davidson, B. et al., J Cell Mol. Med. 15 (2011): 535-544Davis, M. P., Cleve. Clin J Med 79 Electronic Suppl 1 (2012): eS51-eS55de Goeje, P. L. et al., Oncoimmunology 4 (2015): e1014242de Matos, Simoes R. et al., BMC. Syst. Biol 9 (2015): 21De, Meulenaere A. et al., Pathobiology 83 (2016): 327-333De, Plaen E. et al., Immunogenetics 40 (1994): 360-369Dengjel, J. et al., Clin Cancer Res 12 (2006): 4163-4170Denkberg, G. et al., J Immunol 171 (2003): 2197-2207Devetzi, M. et al., Thromb. Haemost. 109 (2013): 716-725Dewar, R. et al., Arch. Pathol. Lab Med 135 (2011): 422-429Dias, R. P. et al., Epigenomics. 5 (2013): 331-340Dobrowolska, H. et al., Cytometry B Clin Cytom. 84 (2013): 21-29Dong, Q. et al., Biomed. Res Int. 2015 (2015): 156432Dorn, J. et al., Crit Rev Clin Lab Sci. 51 (2014): 63-84Du, T. et al., Mol. Cancer 13 (2014): 100Dua, P. et al., Cancer Res 73 (2013): 1934-1945Duan, Z. et al., Clin Cancer Res 9 (2003): 2778-2785Dufour, C. et al., Cancer 118 (2012): 3812-3821Dyrskjot, L. et al., Br. J Cancer 107 (2012): 116-122Ek, S. et al., Cancer Res 62 (2002): 4398-4405Emmrich, S. et al., Genes Dev. 28 (2014): 858-874Fabbri, C. et al., Dig. Endosc. (2017)Falk, K. et al., Nature 351 (1991): 290-296Fan, M. et al., Int. J Clin Exp. Pathol. 7 (2014): 6768-6775Fang, F. et al., Clin Cancer Res 20 (2014a): 6504-6516Fang, L. et al., Biochem. Biophys. Res Commun. 446 (2014b): 272-279Faure, A. et al., Nat Rev Urol. 13 (2016): 141-150Feng, X. et al., Mol. Biosyst. 11 (2015): 2946-2954Ferre, H. et al., Protein Sci. 12 (2003): 551-559Findeis-Hosey, J. J. et al., Biotech. Histochem. 87 (2012): 24-29Fishman, W. H., Tumour. Biol 16 (1995): 394-402Follenzi, A. et al., Nat Genet. 25 (2000): 217-222Fong, L. et al., Proc. Natl. Acad. Sci. U.S.A 98 (2001): 8809-8814Fontalba, A. et al., J Immunol. 179 (2007): 8519-8524Forghanifard, M. M. et al., Cancer Biol Ther. 12 (2011): 191-197Frasor, J. et al., Mol. Cell Endocrinol. 418 Pt 3 (2015): 235-239Fritzsche, F. et al., Br. J Cancer 94 (2006): 540-547Fry, E. A. et al., Int. J Cancer 140 (2017): 495-503Fujiyama, T. et al., J Dermatol. Sci. 75 (2014): 43-48Fukumoto, I. et al., J Hum. Genet. 61 (2016): 109-118Fuqua, S. A. et al., Breast Cancer Res Treat. 144 (2014): 11-19Gabrilovich, D. I. et al., Nat Med. 2 (1996): 1096-1103Gan, X. et al., Dis. Markers 2016 (2016): 5259602Ganguly, R. et al., Mol. Cancer Ther. 13 (2014): 1393-1398Garg, M. et al., J Clin Endocrinol. Metab 99 (2014): E62-E72Gattinoni, L. et al., Nat Rev. Immunol 6 (2006): 383-393Ge, L. et al., J Biomed. Res 29 (2015a)Ge, L. et al., Eur. Rev Med Pharmacol. Sci. 19 (2015b): 2703-2710Geyer, C. R., Epigenetics. 5 (2010): 696-703Ghodsi, M. et al., Int. J Surg. 13 (2015): 193-197Gibbs, P. et al., Melanoma Res 10 (2000): 259-264Gleize, V. et al., Ann. Neurol. 78 (2015): 355-374Gnjatic, S. et al., Proc Natl. Acad. Sci. U.S.A 100 (2003): 8862-8867Godkin, A. et al., Int. Immunol 9 (1997): 905-911Gomez, A. et al., Mol. Pharmacol. 78 (2010): 1004-1011Gong, Y. et al., Adv. Anat. Pathol. 21 (2014): 191-200Gottlieb, H. B. et al., J Neuroendocrinol. 19 (2007): 531-542Gragert, L. et al., Hum. Immunol. 74 (2013): 1313-1320Grah, J. J. et al., Tumori 100 (2014): 60-68Gratio, V. et al., Am. J Pathol. 179 (2011): 2625-2636Green, M. R. et al., Molecular Cloning, A Laboratory Manual 4th (2012)Greenfield, E. A., Antibodies: A Laboratory Manual 2nd (2014)Gu, C. et al., Stem Cells 31 (2013): 870-881Gu, Z. D. et al., Zhonghua Wei Chang Wai Ke. Za Zhi. 10 (2007): 365-367Gualco, G. et al., Appl. Immunohistochem. Mol. Morphol. 18 (2010): 301-310Guerrero, K. et al., Gynecol. Oncol 125 (2012): 720-726Guo, J. T. et al., Zhonghua Zhong. Liu Za Zhi. 31 (2009): 528-531Gupta, A. K. et al., Med J Armed. Forces. India 72 (2016): S37-S42Gustafsson, C. et al., Trends Biotechnol. 22 (2004): 346-353Haass, N. K. et al., Pigment Cell Res 18 (2005): 150-159Haeberle, H. et al., Neoplasia. 14 (2012): 666-669Hall, R. D. et al., Cancer Control 20 (2013): 22-31Hamilton, K. E. et al., Mol. Cancer Res 13 (2015): 1478-1486Han, J. et al., World J Surg. Oncol 10 (2012): 37Han, Y. et al., Eur. J Cell Biol 94 (2015): 642-652Han, Y. D. et al., Oncotarget. 8 (2017): 1871-1883Hanafusa, H. et al., Seikagaku 83 (2011): 1127-1131Harndahl, M. et al., Eur. J Immunol. 42 (2012): 1405-1416Hase, H. et al., Mol. Cancer Res 12 (2014): 1807-1817Hasegawa, H. et al., Arch. Pathol. Lab Med. 122 (1998): 551-554Hashem, N. N. et al., Int. J Biol Markers 25 (2010): 32-37Hashimoto, Y. et al., Oncotarget. (2017)Hayashi, S. I. et al., Endocr. Relat Cancer 10 (2003): 193-202Hayashida, T. et al., Proc. Natl. Acad. Sci. U.S.A 107 (2010): 1100-1105Heeg, S. et al., Gastroenterology 151 (2016): 540-553Heerma van Voss, M. R. et al., Histopathology 65 (2014): 814-827Hemminger, J. A. et al., Mod. Pathol. 27 (2014): 1238-1245Hennard, C. et al., J Pathol. 209 (2006): 430-435Heubach, J. et al., Mol. Cancer 14 (2015): 108Higgins, J. et al., Horm. Cancer 6 (2015): 67-86Hildebrandt, M. O. et al., Bone Marrow Transplant. 22 (1998): 771-775Hiramoto, T. et al., Oncogene 18 (1999): 3422-3426Hirata, T. et al., Oncol Rep. 33 (2015): 2052-2060Hiroumi, H. et al., Int. J Cancer 93 (2001): 786-791Hoff, P. M. et al., Surg. Oncol Clin N. Am. 26 (2017): 57-71Hoffmann, N. E. et al., Cancer 112 (2008): 1471-1479Hofmann, M. C. et al., Eur. Urol. 23 (1993): 38-44Holm, C. et al., Leuk. Res 30 (2006): 254-261Honrado, E. et al., Crit Rev Oncol Hematol. 59 (2006): 27-39Horiuchi, S. et al., J Pathol. 200 (2003): 568-576Horvath, A. et al., World J Gastroenterol. 10 (2004): 152-154Hoshino, Y. et al., Mol. Cancer 13 (2014): 102Hou, Z. et al., Mol. Cancer Ther. (2017)Hu, S. et al., J Cancer Res Clin Oncol 140 (2014): 883-893Hu, X. T. et al., Cell Prolif. 47 (2014): 200-210Huang, K. et al., Chin J Cancer Res 26 (2014): 72-80Huang, X. et al., Cell Prolif. 48 (2015): 593-599Hudolin, T. et al., J Transl. Med 11 (2013): 123Hur, H. et al., J Cancer 7 (2016): 768-773Hussein, Y. M. et al., Med. Oncol 29 (2012): 3055-3062Hwang, M. L. et al., J Immunol. 179 (2007): 5829-5838lacobuzio-Donahue, C. A. et al., Cancer Res 63 (2003): 8614-8622Idbaih, A., Rev Neurol. (Paris) 167 (2011): 691-698Ingaramo, P. I. et al., Mol. Cell Endocrinol. 425 (2016): 37-47Ioannidis, P. et al., Anticancer Res 23 (2003): 2179-2183Ishida, S. et al., Biochem. Biophys. Res Commun. 339 (2006): 325-330Ishikawa, K. et al., Mol. Biol Cell 23 (2012): 1294-1306Ishikawa, S. et al., Cell Tissue Res 337 (2009): 381-391Itesako, T. et al., PLoS. One. 9 (2014): e84311Jacobs, J. et al., Pharmacol. Ther. 155 (2015a): 1-10Jacobs, J. et al., Oncotarget. 6 (2015b): 13462-13475James, S. R. et al., Epigenetics. 8 (2013): 849-863Jang, B. G. et al., Virchows Arch. 467 (2015): 393-403Jeng, Y. M. et al., Br. J Surg. 96 (2009): 66-73Ji, P. et al., Oncogene 24 (2005): 2739-2744Jiang, D. et al., PLoS. One. 9 (2014): e96822Jiang, Y. et al., Mol. Oncol 10 (2016): 292-302Jiang, Y. et al., Oncotarget. 6 (2015): 39865-39876Jiao, T. T. et al., Int. J Clin Exp. Pathol. 6 (2013): 3036-3041Jin, H. et al., Tumour. Biol 27 (2006): 274-282Jnawali, H. N. et al., J Nat Prod. 77 (2014): 258-263John, T. et al., Clin Cancer Res 14 (2008): 3291-3298Jung, D. B. et al., Oncotarget. 6 (2015): 4992-5004Jung, G. et al., Proc Natl Acad Sci USA 84 (1987): 4611-4615Kalejs, M. et al., BMC. Cancer 6 (2006): 6Kannan, K. et al., Proc. Natl. Acad. Sci. U.S.A 112 (2015): E1272-E1277Karlgren, M. et al., Expert. Opin. Ther. Targets. 11 (2007): 61-67Karpf, A. R. et al., Mol. Cancer Res 7 (2009): 523-535Kasashima, H. et al., Cancer Lett. 354 (2014): 438-446Kaufman, L. et al., J Am. Soc. Nephrol. 15 (2004): 1721-1730Kazi, J. A. et al., Neuropeptides 41 (2007): 227-231Kedage, V. et al., Cell Rep. 17 (2016): 1289-1301Keld, R. et al., Br. J Cancer 105 (2011): 124-130Keld, R. et al., Mol. Cancer 9 (2010): 313Kelemen, L. E. et al., Nat Genet. 47 (2015): 888-897Kelly, Z. et al., Int. J Cancer 139 (2016): 1608-1617Kerns, S. L. et al., J Urol. 190 (2013): 102-108Kevans, D. et al., Int J Surg. Pathol. 19 (2011): 751-760Khan, F. S. et al., Hepatol. Int. 11 (2017): 45-53Khan, M. F. et al., Transl. Oncol 5 (2012): 85-91Kibbe, A. H., Handbook of Pharmaceutical Excipients rd (2000)Kikkawa, Y. et al., Exp. Cell Res 328 (2014): 197-206Kikkawa, Y. et al., J Biol Chem 288 (2013): 30990-31001Kikkawa, Y. et al., Exp. Cell Res 314 (2008): 2579-2590Kim, B. K. et al., J Dermatol. Sci. 79 (2015a): 137-147Kim, B. R. et al., Cell Signal. 26 (2014): 1765-1773Kim, T. D. et al., J Clin Invest 126 (2016): 706-720Kim, T. H. et al., J Korean Med Sci. 30 (2015b): 155-161Kim, Y. D. et al., Int. J Mol. Med. 29 (2012): 656-662Kim, Y. H. et al., Ann. Surg. Oncol 18 (2011): 2338-2347King, M. L. et al., Oncogene 34 (2015): 3452-3462Kinoshita, T. et al., Int. Immunol. 18 (2006): 1701-1706Kinoshita, T. et al., J Biol Chem 280 (2005): 21720-21725Kirkova, M. et al., Pharmacol. Rep. 61 (2009): 1163-1172Klatka, J. et al., Eur. Arch. Otorhinolaryngol. 270 (2013): 2683-2693Klauke, K. et al., Nat Cell Biol 15 (2013): 353-362Kleeff, J. et al., Nat Rev Dis. Primers. 2 (2016): 16022Knudsen, K. A. et al., J Cell Biochem. 95 (2005): 488-496Ko, A. et al., BMB. Rep. 49 (2016): 598-606Kocak, H. et al., Cell Death. Dis. 4 (2013): e586Kohrt, D. et al., Cell Cycle 13 (2014): 62-71Kontos, C. K. et al., Clin Chem Lab Med 54 (2016): 315-324Koonrungsesomboon, N. et al., Cancer Epidemiol. 39 (2015): 487-496Korr, D. et al., Cell Signal. 18 (2006): 910-920Kountourakis, P. et al., Thromb. Haemost. 101 (2009): 541-546Krepischi, A. C. et al., Mol. Cytogenet. 9 (2016): 20Krieg, A. M., Nat Rev. Drug Discov. 5 (2006): 471-484Kruhlak, M. et al., Nature 447 (2007): 730-734Kuball, J. et al., Blood 109 (2007): 2331-2338Kuga, T. et al., J Cell Sci. 126 (2013): 4721-4731Kuga, T. et al., Sci. Rep. 6 (2016): 26557Kumar-Sinha, C. et al., Genome Med 7 (2015): 129Kumari, A. et al., BMC. Res Notes 9 (2016): 92Kuner, R. et al., J Mol. Med (Berl) 91 (2013): 237-248Kuo, C. T. et al., Cancer Lett. 378 (2016): 104-110Kuppers, R. et al., J Clin Invest 111 (2003): 529-537Kuraishi, Y., Yakugaku Zasshi 134 (2014): 1125-1142Kurscheid, S. et al., Genome Biol 16 (2015): 16Kwon, O. S. et al., Oncotarget. 6 (2015): 41916-41928La, Vecchia C., Eur. J Cancer Prev. 10 (2001): 125-129Lally, K. M. et al., Int. J Cancer 93 (2001): 841-847Lapin, V. et al., Oncogenesis. 3 (2014): e133Latini, F. R. et al., Blood Cells Mol. Dis. 50 (2013): 161-165Lawrenson, K. et al., Int. J Cancer 136 (2015a): 1390-1401Lawrenson, K. et al., Nat Commun. 6 (2015b): 8234Lazaro-lbanez, E. et al., BMC. Cancer 17 (2017): 92Lederer, M. et al., Semin. Cancer Biol 29 (2014): 3-12Lee, E. et al., BMB. Rep. 46 (2013): 594-599Lee, O. H. et al., Mol. Cell Proteomics. 10 (2011): M110Leong, S. R. et al., Mol. Pharm. 12 (2015): 1717-1729Leung, F. et al., Cancer Epidemiol. Biomarkers Prev. 25 (2016): 1333-1340Li, B. et al., Cell Biochem. Biophys. 70 (2014a): 1363-1368Li, B. et al., Oncotarget. (2016a)Li, H. et al., Gynecol. Oncol 84 (2002): 216-221Li, L. et al., Cytokine 89 (2017): 173-178Li, L. et al., Zhongguo Shi Yan. Xue. Ye. Xue. Za Zhi. 24 (2016b): 326-331Li, M. et al., Oncotarget. 7 (2016c): 51503-51514Li, M. et al., Clin Cancer Res 11 (2005): 1809-1814Li, Q. et al., J Gastroenterol. Hepatol. 29 (2014b): 835-842Li, W. M. et al., J Surg. Oncol (2016d)Li, Z. et al., Cancer Res 76 (2016e): 619-629Liang, X. et al., Oncotarget. 7 (2016): 52207-52217Liddy, N. et al., Nat Med. 18 (2012): 980-987Lilja-Maula, L. et al., J Comp Pathol. 150 (2014): 399-407Lim, J. Y. et al., World J Gastroenterol. 19 (2013): 7078-7088Lin, J. et al., Clin Cancer Res 10 (2004): 5708-5716Lin, L. et al., Oncol Lett. 6 (2013a): 740-744Lin, Q. et al., J Cancer Res Clin Oncol 135 (2009): 1675-1684Lin, Z. et al., Diagn. Pathol. 8 (2013b): 133Linn, D. E. et al., PLoS. One. 10 (2015): e0120628Linz, K. et al., J Pharmacol. Exp. Ther. 349 (2014): 535-548Liu, D. B. et al., World J Gastroenterol. 11 (2005): 1562-1566Liu, M. et al., Cell Mol. Neurobiol. 34 (2014a): 913-923Liu, Q. et al., J Biol Chem 286 (2011): 29951-29963Liu, R. et al., Cancer Biol Ther. 16 (2015a): 317-324Liu, W. et al., Mol. Clin Oncol 2 (2014b): 219-225Liu, X. et al., Biomed. Pharmacother. 88 (2017): 595-602Liu, X. et al., Int. Immunopharmacol. 25 (2015b): 416-424Liu, Y. et al., Int. J Gynecol. Cancer 23 (2013): 304-311Ljunggren, H. G. et al., J Exp. Med. 162 (1985): 1745-1759Llaurado, M. et al., Int. J Cancer 130 (2012a): 1532-1543Llaurado, M. et al., Mol. Cancer Res 10 (2012b): 914-924Longenecker, B. M. et al., Ann N.Y. Acad. Sci. 690 (1993): 276-291Lopez, R. et al., Int. J Gynecol. Cancer 16 (2006): 1289-1296Lopez-Romero, R. et al., Rev Med Inst. Mex. Seguro. Soc. 53 Suppl 2 (2015): S188-S193Lorincz, A. T., Acta Cytol. 60 (2016): 501-512Lose, F. et al., Biol Chem 393 (2012): 403-412Low, G. M. et al., Oral Oncol 61 (2016): 27-30Lukas, T. J. et al., Proc. Natl. Acad. Sci. U.S.A 78 (1981): 2791-2795Lunardi, A. et al., Cancer Discov 5 (2015): 550-563Lundblad, R. L., Chemical Reagents for Protein Modification 3rd (2004)Luo, H. et al., Int. J Clin Exp. Med 7 (2014): 1244-1254Luo, P. et al., Oncol Rep. (2017)Luostari, K. et al., PLoS. One. 9 (2014): e102519Lv, G. Q. et al., Biochem. Cell Biol 92 (2014): 379-389Lv, L. et al., Cancer Lett. 357 (2015a): 105-113Lv, X. et al., J Exp. Clin Cancer Res 34 (2015b): 133Ma, K. et al., Clin Epigenetics. 8 (2016a): 43Ma, Y. et al., Biosens. Bioelectron. 85 (2016b): 641-648Ma, Z. et al., Int. J Clin Exp. Pathol. 8 (2015): 5071-5079MacLean, J. A. et al., PLoS. One. 11 (2016): e0156109Mahajan, A., Hum. Pathol. 51 (2016): 64-74Maine, E. A. et al., Oncotarget. 7 (2016): 14708-14726Maines-Bandiera, S. et al., Int. J Gynecol. Cancer 20 (2010): 16-22Mamane, Y. et al., J Interferon Cytokine Res 22 (2002): 135-143Mantia-Smaldone, G. M. et al., Hum. Vaccin. Immunother. 8 (2012): 1179-1191Manzella, L. et al., Curr. Cancer Drug Targets. 16 (2016): 594-605Mao, L. et al., Cancer Res 71 (2011): 4314-4324Marcinkiewicz, K. M. et al., Exp. Cell Res 320 (2014a): 128-143Marcinkiewicz, K. M. et al., J Cell Physiol 229 (2014b): 1405-1416Marlow, L. A. et al., J Cell Sci. 125 (2012): 4253-4263Maruta, S. et al., APMIS 117 (2009): 791-796Marzese, D. M. et al., Hum. Mol. Genet. 23 (2014): 226-238Masamoto, I. et al., Leuk. Lymphoma 57 (2016): 685-691Mason, J. M. et al., Nucleic Acids Res. 43 (2015): 3180-3196Mawrin, C. et al., J Neurooncol. 99 (2010): 379-391McCormack, E. et al., Cancer Immunol. Immunother. 62 (2013): 773-785Medina-Aguilar, R. et al., Data Brief. 11 (2017): 169-182Mehta, A. et al., Breast 23 (2014): 2-9Melin, B., Curr. Opin. Oncol 23 (2011): 643-647Menezes, J. et al., Leukemia 28 (2014): 823-829Mercer, K. E. et al., Adv. Exp. Med Biol 815 (2015): 185-195Mercer, K. E. et al., Cancer Prev. Res (Phila) 7 (2014): 675-685Mesquita, D. et al., Oncotarget. 6 (2015): 5217-5236Meziere, C. et al., J Immunol 159 (1997): 3230-3237Michaelidou, K. et al., Breast Cancer Res Treat. 152 (2015): 323-336Millan, J. L. et al., Crit Rev Clin Lab Sci. 32 (1995): 1-39Mitsuhashi, K. et al., Int. J Hematol. 100 (2014): 88-95Miyazaki, M. et al., Immunity. 28 (2008): 231-245Miyoshi, Y. et al., Med. Mol. Morphol. 43 (2010): 193-196Mizuno, K. et al., Int. J Oncol 48 (2016): 450-460Moon, H. J. et al., Bioorg. Chem 57 (2014): 231-241Moore, K. N. et al., J Clin Oncol (2016): JCO2016699538Morgan, R. A. et al., Science 314 (2006): 126-129Moroy, T. et al., Semin. Immunol. 23 (2011): 379-387Mortara, L. et al., Clin Cancer Res. 12 (2006): 3435-3443Moskvina, L. V. et al., Arkh. Patol. 72 (2010): 58-61Moussa, O. et al., J Cell Biochem. 108 (2009): 1389-1398Mumberg, D. et al., Proc. Natl. Acad. Sci. U.S.A 96 (1999): 8633-8638Nabeshima, K. et al., Diagn. Cytopathol. 44 (2016): 774-780Nalla, A. K. et al., Mol. Carcinog 55 (2016): 1761-1771Nawaz, I. et al., Oncotarget. 6 (2015): 31493-31507Nishida, C. R. et al., Mol. Pharmacol. 78 (2010): 497-502Niskakoski, A. et al., Epigenetics. 9 (2014): 1577-1587Noah, T. K. et al., Gastroenterology 144 (2013): 1012-1023Notaridou, M. et al., Int. J Cancer 128 (2011): 2063-2074Notaro, S. et al., BMC. Cancer 16 (2016): 589Noubissi, F. K. et al., J Invest Dermatol. 134 (2014): 1718-1724Obiezu, C. V. et al., Cancer Lett. 224 (2005): 1-22Oh, S. et al., Biochim. Biophys. Acta 1826 (2012): 1-12Ohno, S. et al., Anticancer Res 28 (2008): 2493-2497Okada, K. et al., Cancer Sci. 95 (2004): 949-954Okamoto, O. K. et al., Biochim. Biophys. Acta 1769 (2007): 437-442Oliveira-Costa, J. P. et al., Oncotarget. 6 (2015): 20902-20920Orsini, G. et al., Pathol. Oncol Res 20 (2014): 267-276Orwat, D. E. et al., Arch. Pathol. Lab Med 136 (2012): 333-338Ostergaard, Pedersen L. et al., Eur. J Immunol. 31 (2001): 2986-2996Ottaviani, S. et al., Cancer Immunol. Immunother. 55 (2006): 867-872Otte, M. et al., Cancer Res 61 (2001): 6682-6687Oue, N. et al., Cancer Sci. 106 (2015): 951-958Pacholczyk, M. et al., Med Pr 67 (2016): 255-266Pagnotta, S. M. et al., PLoS. One. 8 (2013): e72638Pai, V. P. et al., Breast Cancer Res 11 (2009): R81Pal, M. et al., J Biol Chem 288 (2013): 12222-12231Palacios, J. et al., Pathobiology 75 (2008): 85-94Paliouras, M. et al., Breast Cancer Res Treat. 102 (2007): 7-18Paliouras, M. et al., Mol. Oncol 1 (2008a): 413-424Paliouras, M. et al., Tumour. Biol 29 (2008b): 63-75Palma, M. et al., BMC. Clin Pathol. 12 (2012): 2Papachristopoulou, G. et al., Tumour. Biol 34 (2013): 369-378Papadakis, A. I. et al., Cell Res 25 (2015): 445-458Papadopoulou-Boutis, A. et al., Cancer Detect. Prev. 8 (1985): 141-150Paredes, J. et al., Breast Cancer Res 9 (2007): 214Paredes, J. et al., Biochim. Biophys. Acta 1826 (2012): 297-311Parris, T. Z. et al., BMC. Cancer 14 (2014): 324Parris, T. Z. et al., Clin Cancer Res 16 (2010): 3860-3874Pathiraja, T. N. et al., Sci. Transl. Med 6 (2014): 229ra41Patrick, A. N. et al., Nat Struct. Mol. Biol 20 (2013): 447-453Pattani, K. M. et al., PLoS. One. 7 (2012): e45534Peeters, M. C. et al., Cell Signal. 27 (2015): 2579-2588Pelkonen, M. et al., BMC. Cancer 15 (2015): 431Peng, H. X. et al., Biomed. Res Int. 2015 (2015): 326981Pereira, B. et al., Nucleic Acids Res 41 (2013): 3986-3999Perez, C. A. et al., Expert. Rev Anticancer Ther. 11 (2011): 1599-1605Petersen, G. M., Semin. Oncol 43 (2016): 548-553Petrau, C. et al., J Cancer 5 (2014): 761-764Pich, C. et al., Br. J Cancer 114 (2016a): 63-70Pich, C. et al., PLoS. One. 11 (2016b): e0148095Pickard, M. R. et al., Breast Cancer Res 11 (2009): R60Pineda, C. T. et al., Cell 160 (2015): 715-728Piura, B. et al., Harefuah 144 (2005): 261-5, 303, 302Planaguma, J. et al., Hum. Pathol. 42 (2011): 57-67Planque, C. et al., Biol Chem 389 (2008a): 781-786Planque, C. et al., Clin Chem 56 (2010): 987-997Planque, C. et al., Clin Cancer Res 14 (2008b): 1355-1362Plebanski, M. et al., Eur. J Immunol 25 (1995): 1783-1787Pollack, S. M. et al., PLoS. One. 7 (2012): e32165Pollard, C. et al., Expert. Rev Mol. Med 12 (2010): e10Ponte, J. F. et al., Neoplasia. 18 (2016): 775-784Ponzoni, M. et al., Ann. Oncol 25 (2014): 316-322Popov, N. et al., Epigenetics. 5 (2010): 685-690Porta, C. et al., Virology 202 (1994): 949-955Power, P. F. et al., J Orthop. Res 31 (2013): 493-501Prasad, M. L. et al., Head Neck 26 (2004): 1053-1057Pu, X. et al., Clin Pharmacol. Ther. 96 (2014): 609-615Pyle-Chenault, R. A. et al., Tumour. Biol 26 (2005): 245-257Qin, Y. et al., Chin Med. J (Engl.) 127 (2014): 1666-1671Rabien, A. et al., Tumour. Biol 29 (2008): 1-8Raeisossadati, R. et al., Tumour. Biol 35 (2014): 5299-5305Rahmatpanah, F. B. et al., Leukemia 20 (2006): 1855-1862Rajapakse, S. et al., Zoolog. Sci. 24 (2007): 774-780Rammensee, H. et al., Immunogenetics 50 (1999): 213-219Ramos-Solano, M. et al., Exp. Cell Res 335 (2015): 39-50Rapoport, A. P. et al., Nat Med 21 (2015): 914-921Rasmussen, S. L. et al., Colorectal Dis. 18 (2016): 549-561Rastelli, F. et al., Tumori 96 (2010): 875-888Rauscher, G. H. et al., BMC. Cancer 15 (2015): 816Ravenni, N. et al., MAbs. 6 (2014): 86-94Reck, M., Ann. Oncol 23 Suppl 8 (2012): viii28-viii34RefSeq, The NCBI handbook [Internet], Chapter 18, (2002)Resende, C. et al., Helicobacter. 16 Suppl 1 (2011): 38-44Reyes, C. et al., Appl. Immunohistochem. Mol. Morphol. 21 (2013): 283-286Reyniers, L. et al., J Neurochem. 131 (2014): 239-250Ribeiro, A. S. et al., Front Oncol 4 (2014): 371Ricketts, C. J. et al., PLoS. One. 9 (2014): e85621Riedel, S. S. et al., J Clin Invest 126 (2016): 1438-1450Ries, J. et al., Int. J Oncol 26 (2005): 817-824Rinaldi, A. et al., Pathobiology 77 (2010): 129-135Rini, B. I. et al., Cancer 107 (2006): 67-74Risch, H. A. et al., J Natl. Cancer Inst. 98 (2006): 1694-1706Rock, K. L. et al., Science 249 (1990): 918-921Rodenko, B. et al., Nat Protoc. 1 (2006): 1120-1132Rodriguez-Rodero, S. et al., J Clin Endocrinol. Metab 98 (2013): 2811-2821Roy, A. et al., J Cell Physiol 230 (2015): 504-509Royce, L. S. et al., Invasion Metastasis 12 (1992): 149-155Ruf, M. et al., Clin Cancer Res 21 (2015a): 889-898Ruf, M. et al., Oncoimmunology 4 (2015b): e1049805Rui, X. et al., Int. J Clin Exp. Pathol. 8 (2015): 5435-5442S3-Leitlinie maligne Ovarialtumore, 032-0350L, (2013)Sadik, H. et al., Cancer Res 76 (2016): 4443-4456Saiki, R. K. et al., Science 239 (1988): 487-491Salazar, M. D. et al., Breast Cancer Res 13 (2011): R18Samaan, S. et al., Biol Chem 395 (2014): 991-1001Sanchez, W. Y. et al., Endocrinology 153 (2012): 3179-3189Savone, D. et al., Tumori 102 (2016): 450-458Sawasaki, T. et al., Tumour. Biol 25 (2004): 141-148Schaefer, J. S. et al., J Biol Chem 285 (2010): 11258-11269Schmitt, T. M. et al., Hum. Gene Ther. 20 (2009): 1240-1248Scholten, K. B. et al., Clin Immunol. 119 (2006): 135-145Schulte, I. et al., BMC. Genomics 13 (2012): 719Scrideli, C. A. et al., J Neurooncol. 88 (2008): 281-291Sedgwick, A. E. et al., Cancers (Basel) 8 (2016)Seeger, F. H. et al., Immunogenetics 49 (1999): 571-576Seifi-Alan, M. et al., Asian Pac. J Cancer Prev. 14 (2014): 6625-6629Seki, H. et al., Ann. Surg. Oncol 19 (2012): 1831-1840Seliger, B., Methods Mol. Biol 1102 (2014): 367-380Sha, L. et al., Clin Exp. Med 15 (2015): 55-64Sha, S. et al., Dig. Liver Dis. 45 (2013): 422-429Shabestarian, H. et al., Asian Pac. J Cancer Prev. 16 (2015): 8461-8465Shaffer, A. L. et al., Clin Cancer Res 15 (2009): 2954-2961Shang, B. et al., Cell Death. Dis. 5 (2014): e1285Shantha Kumara, H. M. et al., Cancer Immun. 12 (2012): 16Sharpe, D. J. et al., Oncotarget. 5 (2014): 8803-8815Sher, Y. P. et al., Cancer Res 66 (2006): 11763-11770Sherman, F. et al., Laboratory Course Manual for Methods in Yeast Genetics (1986)Shi, H. et al., World J Surg. Oncol 12 (2014): 188Shi, X. et al., Cancer Lett. 339 (2013): 159-166Shima, H. et al., Int. J Hematol. 99 (2014): 21-31Shrestha, B. et al., FEBS J 279 (2012): 3715-3726Si, Y. Q. et al., Transplant. Proc. 44 (2012): 1407-1411Simeone, A. et al., Mech. Dev. 33 (1991): 215-227Simonova, O. A. et al., Mol. Biol (Mosk) 49 (2015): 667-677Singh-Jasuja, H. et al., Cancer Immunol. Immunother. 53 (2004): 187-195Skandalis, S. S. et al., Matrix Biol 35 (2014): 182-193Skotheim, R. I. et al., Cancer Res 65 (2005): 5588-5598Slim, R. et al., Mol. Hum. Reprod. 18 (2012): 52-56Small, E. J. et al., J Clin Oncol. 24 (2006): 3089-3094Smith, J. B. et al., Gynecol. Oncol 134 (2014): 181-189Snijders, A. M. et al., Mol. Oncol 11 (2017): 167-179Sohal, D. P. et al., Crit Rev Oncol Hematol. 107 (2016): 111-118Song, D. G. et al., J Hematol. Oncol 9 (2016): 56Sontakke, P. et al., PLoS. One. 11 (2016): e0153226Spadaro, A. et al., World J Gastroenterol. 12 (2006): 4716-4720Sriraksa, R. et al., Cancer Prev. Res (Phila) 6 (2013): 1348-1355Stamer, U. M. et al., Br. J Anaesth. 106 (2011): 566-572Steffan, J. J. et al., Cancer Lett. 310 (2011): 109-117Stornaiuolo, A. et al., Cell Differ. Dev. 31 (1990): 119-127Su, S. et al., J Biol Chem 287 (2012): 34809-34824Suciu-Foca, N. et al., J Immunol. 178 (2007): 7432-7441Sun, H. et al., J BUON. 20 (2015): 296-308Sun, S. et al., Dig. Dis. Sci. 61 (2016): 2535-2544Suzuki, N. et al., J Orthop. Res 32 (2014): 915-922Sykes, D. B. et al., Cell 167 (2016): 171-186Szajnik, M. et al., Gynecol. Obstet. (Sunnyvale.) Suppl 4 (2013): 3Szalay, F. et al., World J Gastroenterol. 10 (2004): 42-45Szarvas, T. et al., Int J Cancer 135 (2014): 1596-1604Szczepanski, M. J. et al., Oral Oncol 49 (2013): 144-151Szczepanski, M. J. et al., Biomark. Med. 7 (2013): 575-578Ta, L. et al., Mol. Med Rep. 14 (2016): 1371-1378Tabuse, M. et al., Mol. Cancer 10 (2011): 60Talieri, M. et al., Br. J Cancer 100 (2009): 1659-1665Tan, A. C. et al., Cancer Biol Ther. 7 (2008): 135-144Tan, P. et al., Biochem. Biophys. Res Commun. 419 (2012): 801-808Tanaka, F. et al., Int. J Oncol 10 (1997): 1113-1117Tatarian, T. et al., Surg. Clin North Am. 96 (2016): 1207-1221Tchabo, N. E. et al., Cancer Immun. 9 (2009): 6Teufel, R. et al., Cell Mol Life Sci. 62 (2005): 1755-1762Thomsen, H. et al., BMC. Cancer 16 (2016): 227Thorne, A. et al., Cytotherapy. 17 (2015): 633-646Tian, X. et al., J Transl. Med. 13 (2015): 337Titz, B. et al., Oncogene 29 (2010): 5895-5910Tjalma, W. A., Eur. J Obstet. Gynecol. Reprod. Biol 210 (2017): 275-280Tomioka, N. et al., Cancer Genet. Cytogenet. 201 (2010): 6-14Torres, S. et al., Clin Cancer Res 21 (2015): 4892-4902Tran, E. et al., Science 344 (2014): 641-645Trojandt, S. et al., Hum. Immunol. 77 (2016): 1223-1231Tsukihara, H. et al., PLoS. One. 11 (2016): e0163961Tung, P. Y. et al., Stem Cells 31 (2013): 2330-2342Uehiro, N. et al., Breast Cancer Res 18 (2016): 129Ulker, V. et al., Eur. J Obstet. Gynecol. Reprod. Biol 170 (2013): 188-192Underwood, L. J. et al., Biochim. Biophys. Acta 1502 (2000): 337-350Ushiku, T. et al., Histopathology 61 (2012): 1043-1056van den Hurk, K. et al., Biochim. Biophys. Acta 1826 (2012): 89-102van der Bruggen, P. et al., Immunol. Rev 188 (2002): 51-64van, Duin M. et al., Haematologica 96 (2011): 1662-1669Vanderstraeten, A. et al., Cancer Immunol. Immunother. 63 (2014): 545-557Vardhini, N. V. et al., Tumour. Biol 35 (2014): 10855-10860Vater, I. et al., Leukemia 29 (2015): 677-685Vieira, A. F. et al., Mol. Cancer 14 (2015): 178Vincent, A. et al., Oncotarget. 5 (2014): 2575-2587Vlad, G. et al., Exp. Mol. Pathol. 93 (2012): 294-301Vukovic, M. et al., J Exp. Med 212 (2015): 2223-2234Walker, F. et al., Biol Chem 395 (2014): 1075-1086Wallrapp, C. et al., Cancer Res 60 (2000): 2602-2606Walter, S. et al., J Immunol 171 (2003): 4974-4978Walter, S. et al., Nat Med. 18 (2012): 1254-1261Wang, H. et al., Int. J Cancer 124 (2009): 1349-1357Wang, J. et al., Cancer Epidemiol. Biomarkers Prev. 24 (2015a): 1332-1340Wang, L. et al., Diagn. Pathol. 8 (2013): 190Wang, Q. et al., Onco. Targets. Ther. 8 (2015b): 1971-1977Wang, Q. J. et al., Clin Cancer Res (2016a)Wang, X. et al., Med Oncol 28 (2011): 1225-1254Wang, X. et al., Hum. Immunol. 75 (2014): 1203-1209Wang, X. Z. et al., Oncogene 18 (1999): 5718-5721Wang, Y. et al., BMC. Genomics 17 Suppl 7 (2016b): 515Wang, Y. Y. et al., World J Surg. Oncol 13 (2015c): 259Wanli, H. et al., Yi. Chuan 37 (2015): 1095-1104Watabe, T., J Biochem. 152 (2012): 1-3Wegiel, B. et al., J Natl. Cancer Inst. 100 (2008): 1022-1036Wen, Y. et al., Zhongguo Fei. Ai. Za Zhi. 17 (2014): 30-33Western, P. S. et al., PLoS. One. 6 (2011): e20736Wielscher, M. et al., EBioMedicine 2 (2015): 929-936Willcox, B. E. et al., Protein Sci. 8 (1999): 2418-2423Wilson, E. M., Ther. Adv. Urol. 2 (2010): 105-117Wilson, E. M., Methods Mol. Biol 776 (2011): 113-129Wolff, L. et al., Blood Cells Mol. Dis. 50 (2013): 227-231Wong, C. C. et al., Hepatology 60 (2014a): 1645-1658Wong, N. A. et al., J Clin Pathol. 67 (2014b): 105-111Woo, M. M. et al., Clin Cancer Res 10 (2004): 7958-7964Wu, G. Q. et al., Plasmid 64 (2010): 41-50Wu, L. et al., Int. J Mol. Med. 36 (2015): 1200-1204Wu, P. et al., Nano. Lett. 13 (2013): 4632-4641Wu, S. Y. et al., Nat Commun. 7 (2016): 11169Wu, Z. Y. et al., Scand. J Immunol. 74 (2011): 561-567Xiao, Z. D. et al., Int. J Clin Exp. Pathol. 7 (2014): 4039-4044Xu, C. et al., Biomarkers 20 (2015a): 271-274Xu, C. Q. et al., Alcohol Clin Exp. Res 39 (2015b): 969-979Xu, D. et al., Mol. Biosyst. 12 (2016): 3067-3087Xu, J. et al., Dig. Liver Dis. 46 (2014a): 750-757Xu, L. et al., Zhongguo Fei. Ai. Za Zhi. 14 (2011): 727-732Xu, Y. et al., Oncol Lett. 7 (2014b): 1474-1478Xuan, F. et al., Neuro. Oncol 18 (2016): 819-829Xylinas, E. et al., Biomolecules. 6 (2016)Yakimchuk, K. et al., Mol. Cell Endocrinol. 375 (2013): 121-129Yamada, R. et al., Tissue Antigens 81 (2013): 428-434Yamagata, M. et al., J Neurosci. 32 (2012): 14402-14414Yamamoto, H. et al., Carcinogenesis 25 (2004): 325-332Yang, B. et al., Asian Pac. J Trop. Med 9 (2016a): 1105-1110Yang, F. et al., Onco. Targets. Ther. 9 (2016b): 7039-7045Yang, H. et al., Oncol Rep. 34 (2015a): 1681-1691Yang, L. et al., Oncol Lett. 12 (2016c): 4068-4074Yang, P. et al., Curr. Pharm. Des 21 (2015b): 1292-1300Yang, W. et al., Cancer 91 (2001): 1277-1283Yang, X. S. et al., Asian Pac. J Cancer Prev. 13 (2012): 1657-1662Yao, J. et al., Cancer Immunol. Res. 2 (2014): 371-379Yi, J. M. et al., Tumour. Biol 33 (2012): 363-372Yi, Y. J. et al., Int. J Mol. Med 37 (2016): 1405-1411Yilmaz-Ozcan, S. et al., PLoS. One. 9 (2014): e107905Yin, B. et al., Int. J Clin Exp. Pathol. 7 (2014): 2934-2941Yin, X. Y. et al., Oncogene 20 (2001): 2908-2917Yin, X. Y. et al., Oncogene 18 (1999): 6621-6634Yoon, H. et al., Tohoku J Exp. Med 224 (2011): 41-46Yu, H. et al., Blood 124 (2014): 1737-1747Yu, Y. et al., J Pharmacol. Sci. 112 (2010): 83-88Yuan, R. et al., Cancer Res 74 (2014): 5287-5300Yuan, R. H. et al., Ann Surg. Oncol 16 (2009): 1711-1719Yue, W. et al., Sci. Rep. 5 (2015): 13390Zamuner, F. T. et al., Mol. Cancer Ther. 14 (2015): 828-834Zanaruddin, S. N. et al., Hum. Pathol. 44 (2013): 417-426Zaremba, S. et al., Cancer Res. 57 (1997): 4570-4577Zha, Y. et al., PLoS. One. 7 (2012): e40728Zhai, Y. et al., Cancer Res 67 (2007): 10163-10172Zhan, J. et al., Br. J Cancer 111 (2014): 883-893Zhan, J. et al., Cancer Lett. 361 (2015): 75-85Zhang, C. et al., J Surg. Res 197 (2015a): 301-306Zhang, D. et al., Oncol Rep. 35 (2016a): 81-88Zhang, G. et al., Int. J Biochem. Cell Biol 36 (2004): 1613-1623Zhang, H. et al., J Exp. Clin Cancer Res 26 (2007): 361-366Zhang, H. Y. et al., Oncol Rep. 34 (2015b): 1193-1202Zhang, J. et al., Oncotarget. 6 (2015c): 42040-42052Zhang, J. et al., Sci. Rep. 7 (2017): 42819Zhang, K. et al., Tumour. Biol 35 (2014a): 7669-7673Zhang, L. et al., Med Oncol 31 (2014b): 52Zhang, L. et al., Cancer Res 65 (2005): 925-932Zhang, M. et al., Onco. Targets. Ther. 9 (2016b): 2717-2723Zhang, R. et al., Mol. Med Rep. 5 (2012a): 256-259Zhang, W. et al., Epigenetics. 10 (2015d): 736-748Zhang, W. et al., Acta Haematol. 130 (2013): 297-304Zhang, X. et al., Int. J Oncol (2016c)Zhang, X. et al., Med. Oncol 32 (2015e): 148Zhang, Y. et al., Mol. Med. Rep. 5 (2012b): 910-916Zhang, Z. et al., Tumour. Biol (2015f)Zhao, H. et al., Zhonghua Gan Zang. Bing. Za Zhi. 10 (2002): 100-102Zhao, L. et al., Oncogene (2017)Zhao, R. et al., EBioMedicine 8 (2016): 30-39Zheng, J. et al., J Surg. Oncol 107 (2013): 746-751Zhou, B. et al., Biochim. Biophys. Acta 1784 (2008): 747-752Zhou, Y. et al., Mol. Cell Endocrinol. 386 (2014): 16-33Zhu, J. et al., Asian Pac. J Cancer Prev. 14 (2013): 3011-3015Zhu, N. et al., J Clin Invest 126 (2016): 997-1011Zhussupova, A. et al., PLoS. One. 9 (2014): e105285Zou, C. et al., Cancer 118 (2012): 1845-1855Zufferey, R. et al., J Virol. 73 (1999): 2886-2892Neumann, F. et al., Cancer Immunol. Immunother. 53 (2004): 589-599Rammensee, H. et al., Immunogenetics 50 (1999): 213-219Sun, Y. et al., Int. J. Cancer 87 (2000): 399-404
367,292
11858979
DETAILED DESCRIPTION OF THE INVENTION Definitions By a “polypeptide” is meant any sequence of two or more amino acids, regardless of length, post-translation modification, or function. “Polypeptide,” “peptide,” and “protein” are used interchangeably herein. Polypeptides can include natural amino acids and non-natural amino acids such as those described in U.S. Pat. No. 6,559,126. Polypeptides can also be modified in any of a variety of standard chemical ways (e.g., an amino acid can be modified with a protecting group; the carboxy-terminal amino acid can be made into a terminal amide group; the amino-terminal residue can be modified with groups to, e.g., enhance lipophilicity; or the polypeptide can be chemically glycosylated or otherwise modified to increase stability or in vivo half-life). Polypeptide modifications can include the attachment of another structure such as a cyclic compound or other molecule to the polypeptide and can also include polypeptides that contain one or more amino acids in an altered configuration (i.e., R or S; or, L or D). The peptides of the invention are proteins derived from the tenth type III domain of fibronectin that have been modified to bind specifically to PCSK9 and are referred to herein as, “anti-PCSK9 Adnectin” or “PCSK9 Adnectin”. The term “PK” is an acronym for “pharmacokinetic” and encompasses properties of a compound including, by way of example, absorption, distribution, metabolism, and elimination by a subject. A “PK modulation protein” or “PK moiety” refers to any protein, peptide, or moiety that affects the pharmacokinetic properties of a biologically active molecule when fused to or administered together with the biologically active molecule. Examples of a PK modulation protein or PK moiety include PEG, human serum albumin (HSA) binders (as disclosed in U.S. Publication Nos. 2005/0287153 and 2007/0003549, PCT Publication Nos. WO 2009/083804 and WO 2009/133208, and SABA molecules as described herein), human serum albumin, Fc or Fc fragments and variants thereof, and sugars (e.g., sialic acid). “Percent (%) amino acid sequence identity” herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in a selected sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR®) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared. An “isolated” polypeptide is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. In preferred embodiments, the polypeptide will be purified (1) to greater than 95% by weight of polypeptide as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing condition using Coomassie blue or, preferably, silver stain. Isolated polypeptide includes the polypeptide in situ within recombinant cells since at least one component of the polypeptide's natural environment will not be present. Ordinarily, however, isolated polypeptide will be prepared by at least one purification step. The notations “mpk”, “mg/kg”, or “mg per kg” refer to milligrams per kilogram. All notations are used interchangeably throughout the present disclosure. The “half-life” of an amino acid sequence or compound can generally be defined as the time taken for the serum concentration of the polypeptide to be reduced by 50%, in vivo, for example due to degradation of the sequence or compound and/or clearance or sequestration of the sequence or compound by natural mechanisms. The half-life can be determined in any manner known per se, such as by pharmacokinetic analysis. Suitable techniques will be clear to the person skilled in the art, and may for example generally involve the steps of suitably administering to the primate a suitable dose of the amino acid sequence or compound of the invention; collecting blood samples or other samples from said primate at regular intervals; determining the level or concentration of the amino acid sequence or compound of the invention in said blood sample; and calculating, from (a plot of) the data thus obtained, the time until the level or concentration of the amino acid sequence or compound of the invention has been reduced by 50% compared to the initial level upon dosing. Reference is, for example, made to the standard handbooks, such as Kenneth, A. et al.,Chemical Stability of Pharmaceuticals: A Handbook for Pharmacistsand in Peters et al.,Pharmacokinete Analysis: A Practical Approach(1996). Reference is also made to Gibaldi, M. et al.,Pharmacokinetics,2nd Rev. Edition, Marcel Dekker (1982). Half-life can be expressed using parameters such as the t1/2-alpha, t1/2-beta, HL_Lambda_z, and the area under the curve (AUC). In the present specification, an “increase in half-life” refers to an increase in any one of these parameters, any two of these parameters, any three of these parameters or all four of these parameters. An “increase in half-life” in particular refers to an increase in the t1/2-beta and/or HL_Lambda_z, either with or without an increase in the t1/2-alpha and/or the AUC or both. Overview This application provides Adnectins against human PCSK9. In order to identify PCSK9 specific antagonists, PCSK9 was presented to large synthetic libraries of Adnectins. Adnectins that bound to PCSK9 were screened for PCSK9 binding, for biophysical properties, and for PCSK9 inhibitory activity. The anti-PCSK9 Adnectins were mutated and subjected to further selective pressure by lowering the target concentration and selecting for anti-PCSK9 Adnectins with slow off-rates. From this optimization process, a family of Adnectins was identified as PCSK9 specific inhibitors with favorable biochemical and biophysical properties. Fibronectin Based Scaffolds One aspect of the application provides for polypeptides comprising Fn3 domain in which one or more of the solvent accessible loops has been randomized or mutated. The Fn3 domain is an Fn3 domain derived from the wild-type tenth module of the human fibronectin type III domain (10Fn3): VSDVPRDLEVVAATPTSLLISWDAPAVTVRYYRITYGETGGNSPVQEFTVPGSKSTATIS GLKPGVDYTITVYAVTGRGDSPASSKPISINYRT (SEQ ID NO: 1). In the10Fn3 sequence, the BC, DE and FG loops are underlined. As described herein, non-ligand binding sequences of10Fn3, i.e., the “10Fn3 scaffold”, may be altered provided that the10Fn3 retains ligand binding function and/or structural stability. A variety of mutant10Fn3 scaffolds have been reported. In one aspect, one or more of Asp 7, Glu 9, and Asp 23 is replaced by another amino acid, such as, for example, a non-negatively charged amino acid residue (e.g., Asn, Lys, etc.). These mutations have been reported to have the effect of promoting greater stability of the mutant10Fn3 at neutral pH as compared to the wild-type form (See, PCT Publication No. WO 02/04523). A variety of additional alterations in the10Fn3 scaffold that are either beneficial or neutral have been disclosed. See, for example, Batori et al.,Protein Eng.,15(12):1015-1020 (December 2002); Koide et al.,Biochemistry,40(34):10326-10333 (Aug. 28, 2001). Both variant and wild-type10Fn3 proteins are characterized by the same structure, namely seven beta-strand domain sequences designated A through G and six loop regions (AB loop, BC loop, CD loop, DE loop, EF loop, and FG loop) which connect the seven beta-strand domain sequences. The beta strands positioned closest to the N- and C-termini may adopt a beta-like conformation in solution. In SEQ ID NO:1, the AB loop corresponds to residues 15-16, the BC loop corresponds to residues 21-30, the CD loop corresponds to residues 39-45, the DE loop corresponds to residues 51-56, the EF loop corresponds to residues 60-66, and the FG loop corresponds to residues 76-87 (Xu et al.,Chemistry&Biology,9:933-942 (2002)). In some embodiments, the10Fn3 polypeptide may be at least 40%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% identical to the human10Fn3 domain, shown in SEQ ID NO:1. Much of the variability will generally occur in one or more of the loops. Each of the beta or beta-like strands of a10Fn3 polypeptide may consist essentially of an amino acid sequence that is at least 80%, 85%, 90%, 95% or 100% identical to the sequence of a corresponding beta or beta-like strand of SEQ ID NO:1, provided that such variation does not disrupt the stability of the polypeptide in physiological conditions. The disclosure provides polypeptides comprising a tenth fibronectin type III (10Fn3) domain, wherein the10Fn3 domain comprises a loop, AB; a loop, BC; a loop, CD; a loop, DE; a loop EF; and a loop FG; and has at least one loop selected from loop BC, DE, and FG with an altered amino acid sequence relative to the sequence of the corresponding loop of the human10Fn3 domain. In some embodiments, the BC and FG loops are altered, and in some embodiments, the BC, DE, and FG loops are altered, i.e., the Fn3 domains comprise non-naturally occurring loops. In some embodiments, the AB, CD and/or the EF loops are altered. By “altered” is meant one or more amino acid sequence alterations relative to a template sequence (corresponding human fibronectin domain) and includes amino acid additions, deletions, and substitutions. Altering an amino acid sequence may be accomplished through intentional, blind, or spontaneous sequence variation, generally of a nucleic acid coding sequence, and may occur by any technique, for example, PCR, error-prone PCR, or chemical DNA synthesis. In some embodiments, one or more loops selected from BC, DE, and FG may be extended or shortened in length relative to the corresponding human fibronectin loop. In some embodiments, the length of the loop may be extended by 2-25 amino acids. In some embodiments, the length of the loop may be decreased by 1-11 amino acids. To optimize antigen binding, therefore, the length of a loop of10Fn3 may be altered in length as well as in sequence to obtain the greatest possible flexibility and affinity in antigen binding. In some embodiments, the polypeptide comprises a Fn3 domain that comprises an amino acid sequence at least 80, 85, 90, 95, 98, 99 or 100% identical to the non-loop regions of SEQ ID NO:1, wherein at least one loop selected from BC, DE, and FG is altered. In some embodiments, the altered BC loop has up to 10 amino acid substitutions, up to 4 amino acid deletions, up to 10 amino acid insertions, or a combination thereof. In some embodiments, the altered DE loop has up to 6 amino acid substitutions, up to 4 amino acid deletions, up to 13 amino acid insertions or a combination thereof. In some embodiments, the FG loop has up to 12 amino acid substitutions, up to 11 amino acid deletions, up to 25 amino acid insertions or a combination thereof. As described above, amino acid residues corresponding to residues 21-30, 51-56, and 76-87 of SEQ ID NO: 1 define the BC, DE, and FG loops, respectively. However, it should be understood that not every residue within the loop region needs to be modified in order to achieve a10Fn3 binder having strong affinity for a desired target (e.g., PCSK9). For example, residues 21 (S) and 22 (W) of the BC loop as shown in SEQ ID NO: 1 do not need to be modified for binding PCSK9. That is,10Fn3 domains with high affinity binding to PCSK9 may be obtained by modifying only residues 23-30 of loop BC as shown in SEQ ID NO: 1. This is demonstrated in the BC loops exemplified in Table 3, which indicates that only the residues spanning the italicized positions were altered. Therefore, in some embodiments, a BC loop according to this designation comprises the italicized portion of any one of SEQ ID NOs: 2-17, 106-135, and 301-303 as shown in Table 3. For example, in one embodiment, a BC loop may comprise the sequence PPPSHGYG (residues 3-10 of SEQ ID NO: 2), DAPAHAYG (residues 3-10 of SEQ ID NO: 5), EPFSRLPGGGE (residues 3-13 of SEQ ID NO: 106), or DAPADGGYG (residues 3-11 of SEQ ID NO: 107). Similarly, positions 51 (P) and 56 (T) of loop DE as shown in SEQ ID NO: 1 do not need to be modified for binding PCSK9. That is,10Fn3 domains with high affinity binding to PCSK9 may be obtained by modifying only residues 52-55 of loop DE as shown in SEQ ID NO: 1. This is demonstrated in the DE loops exemplified in Table 3, which indicates that only the residues spanning the italicized positions were altered. Therefore, in some embodiments, a DE loop according to this designation comprises the italicized portion of any one of SEQ ID NOs: 18-27 and 136-141, as shown in Table 3. For example, in one embodiment, a DE loop may comprise the sequence PGKG (residues 2-5 of SEQ ID NO: 18), VGVG (residues 2-5 SEQ ID NO: 27), or VSKS (residues 2-5 of SEQ ID NO: 137). Likewise, position 87 (P) of the FG loop as shown in SEQ ID NO: 1 does not need to be modified for binding PCSK9. That is,10Fn3 domains with high affinity binding to PCSK9 may be obtained by modifying only residues 76-86 of the FG loop as shown in SEQ ID NO: 1. This is demonstrated in the FG loops exemplified in Table 3, which indicates that only the residues spanning the italicized positions were altered. Therefore, in some embodiments, an FG loop according to this designation comprises the italicized portion of any one of SEQ ID NOs: 28-38 and 142-172, as shown in Table 3. For example, in one embodiment, an FG loop may comprise the sequence EYPYKHSGYYHR (residues 1-12 in SEQ ID NO: 28), EYPYDYSGYYHR (residues 1-12 in SEQ ID NO: 142), or EFDFVGAGYYHR (residues 1-12 in SEQ ID NO: 167). In some embodiments, the present application demonstrates that the BC, DE, and FG loop regions can be generally described according to consensus sequences. For example, the BC loop can be generally defined by the consensus sequence SW(X1)ZX2G (SEQ ID NO: 323) where X1is any amino acid, Z is a number from 6-9, and X2is Y or H. This consensus sequence is exemplified by BC loops shown in Table 3 except for the BC loop defined by SEQ ID NO: 106. In other embodiments, Z is a number selected from 2-5. In certain embodiments, Z is a number selected from 10-15. In some embodiments, X2is any aromatic residue (i.e., Y, F, W, or H). In another embodiment, the DE loop can be generally defined by the consensus sequence PX1X1X1X3T, (SEQ ID NO: 324) where X1is any amino acid and X3is G or S. This consensus is exemplified by the DE loops shown in Table 3. In another embodiment, the FG loop can be generally defined by the consensus sequence EX4X1X5X1X1X6GYX4HRP (SEQ ID NO: 325), where X1is any amino acid; X4is Y or F; X5is Y, F, or W; and X6is S or A. This consensus is exemplified by the FG loops shown in Table 3. Accordingly, in certain embodiments, the present disclosure provides a PCSK9 binding Adnectin comprising a BC loop having the sequence SW(X1)ZX2G (SEQ ID NO: 323), a DE loop having the sequence PX1X1X1X3T (SEQ ID NO: 324), and an FG loop having the sequence EX4X1X5X1X1X6GYX4HRP (SEQ ID NO: 325), as defined above. In another embodiment, the present disclosure provides a PCSK9 binding Adnectin comprising a BC loop having the sequence SWEPFSRLPGGGE (SEQ ID NO: 106), a DE loop having the sequence PX1X1X1X3T (SEQ ID NO: 324), and an FG loop having the sequence EX4X1X5X1X1X6GYX4HRP (SEQ ID NO: 325), as defined above. In certain embodiments, a BC loop can be generally defined by the consensus sequence (X1)ZX2G (SEQ ID NO: 449) where X1is any amino acid, Z is a number from 6-9, and X2is Y or H. This consensus sequence is exemplified by BC loops shown in Table 3 except for the BC loop defined by SEQ ID NO: 106. In other embodiments, Z is a number selected from 2-5. In certain embodiments, Z is a number selected from 10-15. In some embodiments, X2is any aromatic residue (i.e., Y, F, W, or H). In certain embodiments, the DE loop can be generally defined by the consensus sequence X1X1X1X3, (SEQ ID NO: 450) where X1is any amino acid and X3is G or S. This consensus is demonstrated by the DE loops shown in Table 3. The FG loop is defined by the consensus sequence EX4X1X5X1X1X6GYX4HR (SEQ ID NO: 451), where X1is any amino acid; X4is Y or F; X5is Y, F, or W; and X6is S or A. This consensus is exemplified by the FG loops shown in Table 3. Accordingly, in one embodiment, the present disclosure provides a PCSK9 binding Adnectin having a BC loop comprising the sequence (X1)ZX2G (SEQ ID NO: 449), a DE loop comprising the sequence X1X1X1X3(SEQ ID NO: 450), and an FG loop comprising the sequence EX4X1X5X1X1X6GYX4HR (SEQ ID NO: 451), as defined above. In certain embodiments, antibody-like proteins based on the10Fn3 scaffold can be defined generally by the following sequence: (SEQ ID NO: 328)EVVAAT(X)aSLLI(X)xYYRITYGE(X)bQEFTV(X)yATI(X)cDYTITVYAV(X)zISINYRT In SEQ ID NO:328, the AB loop is represented by Xa, the CD loop is represented by Xb, the EF loop is represented by Xc, the BC loop is represented by Xx, the DE loop is represented by Xy, and the FG loop is represented by Xz. X represents any amino acid and the subscript following the X represents an integer of the number of amino acids. In particular, a may be anywhere from 1-15, 2-15, 1-10, 2-10, 1-8, 2-8, 1-5, 2-5, 1-4, 2-4, 1-3, 2-3, or 1-2 amino acids; and b, c, x, y and z may each independently be anywhere from 2-20, 2-15, 2-10, 2-8, 5-20, 5-15, 5-10, 5-8, 6-20, 6-15, 6-10, 6-8, 2-7, 5-7, or 6-7 amino acids. In preferred embodiments, a is 2 amino acids, b is 7 amino acids, c is 7 amino acids, x is 9 amino acids, y is 6 amino acids, and z is 12 amino acids. The sequences of the beta strands may have anywhere from 0 to 10, from 0 to 8, from 0 to 6, from 0 to 5, from 0 to 4, from 0 to 3, from 0 to 2, or from 0 to 1 substitutions, deletions or additions across all 7 scaffold regions relative to the corresponding amino acids shown in SEQ ID NO: 1. In an exemplary embodiment, the sequences of the beta strands may have anywhere from 0 to 10, from 0 to 8, from 0 to 6, from 0 to 5, from 0 to 4, from 0 to 3, from 0 to 2, or from 0 to 1 conservative substitutions across all 7 scaffold regions relative to the corresponding amino acids shown in SEQ ID NO: 1. In certain embodiments, the core amino acid residues are fixed and any substitutions, conservative substitutions, deletions or additions occur at residues other than the core amino acid residues. In exemplary embodiments, the BC, DE, and FG loops as represented by (X)x, (X)y, and (X)z, respectively, are replaced with polypeptides comprising the BC, DE and FG loop sequences from any of the PCSK9 binders shown in Table 3, or the italicized portions thereof, or the consensus sequences 323-325 or 449-451. In certain embodiments, Antibody-like proteins based on the10Fn3 scaffold can be defined generally by the sequence: (SEQ ID NO: 329)EVVAATPTSLLI(X)xYYRITYGETGGNSPVQEFTV(X)yATISGLKPGVDYTITVYAV(X)zISINYRT In SEQ ID NO:329, the BC loop is represented by Xx, the DE loop is represented by Xy, and the FG loop is represented by Xz. X represents any amino acid and the subscript following the X represents an integer of the number of amino acids. In particular, x, y and z may each independently be anywhere from 2-20, 2-15, 2-10, 2-8, 5-20, 5-15, 5-10, 5-8, 6-20, 6-15, 6-10, 6-8, 2-7, 5-7, or 6-7 amino acids. In preferred embodiments, x is 9 amino acids, y is 6 amino acids, and z is 12 amino acids. The sequences of the beta strands may have anywhere from 0 to 10, from 0 to 8, from 0 to 6, from 0 to 5, from 0 to 4, from 0 to 3, from 0 to 2, or from 0 to 1 substitutions, deletions or additions across all 7 scaffold regions relative to the corresponding amino acids shown in SEQ ID NO: 1. In an exemplary embodiment, the sequences of the beta strands may have anywhere from 0 to 10, from 0 to 8, from 0 to 6, from 0 to 5, from 0 to 4, from 0 to 3, from 0 to 2, or from 0 to 1 conservative substitutions across all 7 scaffold regions relative to the corresponding amino acids shown in SEQ ID NO: 1. In certain embodiments, the core amino acid residues are fixed and any substitutions, conservative substitutions, deletions or additions occur at residues other than the core amino acid residues. In exemplary embodiments, the BC, DE, and FG loops as represented by (X)x, (X)y, and (X)z, respectively, are replaced with polypeptides comprising the BC, DE and FG loop sequences from any of the PCSK9 binders shown in Table 3, or the italicized portions thereof, or the consensus sequences 323-325 or 449-451. In certain embodiments, an anti-PCSK9 Adnectin described herein may comprise the sequence as set forth in SEQ ID NO: 328 or 329, wherein the BC, DE, and FG loops as represented by (X)x, (X)y, and (X)z, respectively, are replaced with a respective set of specified BC, DE, and FG loops from any of the clones in Table 3, or sequences at least 75%, 80%, 85%, 90%, 95%, 97%, 98%, or 99% identical to the BC, DE or FG loop sequences of the clones listed in Table 3. In exemplary embodiments, an anti-PCSK9 Adnectin as described herein is defined by SEQ ID NO: 329 and has a respective set of BC, DE and FG loop sequences from any of the clones listed in Table 3. For example, clone 1459D05 in Table 3 comprises BC, DE, and FG loops as set forth in SEQ ID NOs: 2, 18, and 28, respectively. Therefore, an anti-PCSK9 Adnectin based on these loops may comprise SEQ ID NO: 328 or 329, wherein (X)xcomprises SEQ ID NO: 2, (X)ycomprises SEQ ID NO: 18, and (X)zcomprises SEQ ID NO: 28. Similar constructs are contemplated utilizing the set of BC, DE and FG loops from the other clones in Table 3, or the consensus sequences 323-325 or 449-451. The scaffold regions of such anti-PCSK9 Adnectin may comprise anywhere from 0 to 20, from 0 to 15, from 0 to 10, from 0 to 8, from 0 to 6, from 0 to 5, from 0 to 4, from 0 to 3, from 0 to 2, or from 0 to 1 substitutions, conservative substitutions, deletions or additions relative to the scaffold amino acids residues of SEQ ID NO: 1. Such scaffold modifications may be made, so long as the anti-PCSK9 Adnectin is capable of binding PCSK9 with a desired KD. In some embodiments, the BC loop of the protein of the invention comprises an amino acid sequence selected from the group consisting of SWPPPSHGYG (SEQ ID NO: 2), SWRPPIHAYG (SEQ ID NO: 3), SWDAPIHAYG (SEQ ID NO:4), SWDAPAHAYG (SEQ ID NO:5) and SWDAPAVTYG (SEQ ID NO:6), SWSPPANGYG (SEQ ID NO:7), SWTPPPKGYG (SEQ ID NO:8), SWRPPSHAYG (SEQ ID NO:9), SWDPPSHAYG (SEQ ID NO:10), SWEPPSHAYG (SEQ ID NO:11), SWSPPSHAYG (SEQ ID NO:12), SWRPPSNGHG (SEQ ID NO:13), SWVPPSDDYG (SEQ ID NO:14), SWVPSSHAYG (SEQ ID NO:15), SWDPSSHAYG (SEQ ID NO:16), and SWEPSSHAYG (SEQ ID NO:17). In further embodiments, the BC loop of the protein of the invention comprises an amino acid sequence selected from SEQ ID NOs: 106-135 and 301-303. In other embodiments, the BC loop of the protein of the invention comprises the italicized portion of any one of SEQ ID NOs: 2-17, 106-135, and 301-303 as shown in Table 3. For example, in one embodiment, a BC loop comprises the sequence PPPSHGYG (residues 3-10 of SEQ ID NO: 2), DAPAHAYG (residues 3-10 of SEQ ID NO: 5), EPFSRLPGGGE (residues 3-13 of SEQ ID NO: 106), or DAPADGGYG (residues 3-11 of SEQ ID NO: 107). In some embodiments, the DE loop of the protein of the invention comprises an amino acid sequence selected from the group consisting of PPGKGT (SEQ ID NO:18), PIVEGT (SEQ ID NO:19), PGSEGT (SEQ ID NO:20), PGSKGT (SEQ ID NO:21), PGSKST (SEQ ID NO:22), PVGRGT (SEQ ID NO:23), PVGEGT (SEQ ID NO:24), PIGKGT (SEQ ID NO:25), PVNEGT (SEQ ID NO:26), and PVGVGT (SEQ ID NO:27). In further embodiments, the DE loop of the protein of the invention comprises an amino acid sequence selected from SEQ ID NOs: 136-141. In other embodiments, the DE loop of the protein of the invention comprises the italicized portion of any one of SEQ ID NOs: 18-27 and 136-141 as shown in Table 3. For example, in one embodiment, a DE loop comprises the sequence PGKG (residues 2-5 of SEQ ID NO: 18), VGVG (residues 2-5 SEQ ID NO: 27), or VSKS (residues 2-5 of SEQ ID NO: 137). In some embodiments, the FG loop of the protein of the invention comprises an amino acid sequence selected from the group consisting of EYPYKHSGYYHRP (SEQ ID NO:28), EYTFKHSGYYHRP (SEQ ID NO:29), EYTYKGSGYYHRP (SEQ ID NO:30), EYTYNGAGYYHRP (SEQ ID NO:31), EYTYIGAGYYHRP (SEQ ID NO:32), EYTYEGAGYYHRP (SEQ ID NO:33), EYAYNGAGYYHRP (SEQ ID NO:34), EYPWKGSGYYHRP (SEQ ID NO:35), EFPFKWSGYYHRP (SEQ ID NO:36), EFPWPHAGYYHRP (SEQ ID NO:37) and EYAFEGAGYYHRP (SEQ ID NO:38). In further embodiments, the FG loop of the protein of the invention comprises an amino acid sequence selected from SEQ ID NOs: 142-172. In other embodiments, the FG loop of the protein of the invention comprises the italicized portion of any one of SEQ ID NOs: 28-38 and 142-172 as shown in Table 3. For example, in one embodiment, an FG loop comprises the sequence EYPYKHSGYYHR (residues 1-12 in SEQ ID NO: 28), EYPYDYSGYYHR (residues 1-12 in SEQ ID NO: 142), or EFDFVGAGYYHR (residues 1-12 in SEQ ID NO: 167). In some embodiments, the protein of the invention comprises one BC loop sequence selected from the BC loop sequences having SEQ ID NOs: 2-17, 106-135, and 301-303, or the italicized portion of any one of SEQ ID NOS: 2-17, 106-135, and 301-303, as shown in Table 3; one DE loop sequence selected from the DE loop sequences having SEQ ID NOs:18-27 and 136-141, or the italicized portion of any one of SEQ ID NOS:18-27 and 136-141 as shown in Table 3; and one FG loop sequence selected from the FG loop sequences having SEQ ID NOS: 28-38 and 142-172, or the italicized portion of any one of SEQ ID NOS: 28-38 and 142-172 as shown in Table 3. In some embodiments, the protein of the invention comprises a BC, DE and FG loop amino acid sequence at least 70, 75, 80, 85, 90, 95, 98, 99 or 100% identical to of any one of SEQ ID NOS: 2-38, 106-172, 301-303. In other embodiments, the protein of the invention comprises a BC, DE and FG loop amino acid sequence at least 70, 75, 80, 85, 90, 95, 98, 99 or 100% identical to the italicized portion of any one of SEQ ID NOS: 2-38, 106-172, 301-303 as shown in Table 3. In some embodiments, the anti-PCSK9 Adnectin comprises the amino acid sequence of any one of SEQ ID NOS: 39-76, 173-290, and 304-309. In some embodiments, the anti-PCSK9 Adnectin comprises the Fn3 domain amino acid sequence from position 3-96 of any one of SEQ ID NOS: 39-76, 173-290, and 304-309. In some embodiments, the anti-PCSK9 Adnectin comprises an amino acid sequence at least 70, 75, 80, 85, 90, 95, 98, 99 or 100% identical to any one of SEQ ID NOS:39-76, 173-290, and 304-309. Fibronectin naturally binds certain types of integrins through its integrin-binding motif, “arginine-glycine-aspartic acid” (RGD). In some embodiments, the polypeptide comprises a10Fn3 domain that lacks the (RGD) integrin binding motif. The integrin binding domain may be removed by altering the RGD sequence by amino acid substitution, deletion or insertion. In certain embodiments, the anti-PCSK9 Adnectin molecules of the present invention may be modified to comprise an N-terminal extension sequence and/or a C-terminal extension. For example, an MG sequence may be placed at the N-terminus of the10Fn3 defined by SEQ ID NO: 1. The M will usually be cleaved off, leaving a G at the N-terminus. Alternatively, the first 10 amino acids of the anti-PCSK9 Adnectins shown in Table 4 may be replaced with an alternative N-terminal sequence, referred to herein as N-terminal extensions, as shown in Table 6 (i.e., SEQ ID NOs: 371-379). In addition, an M, G or MG may also be placed N-terminal to any of the N-terminal extensions having SEQ ID NOs: 371-379. The anti-PCSK9 Adnectins described herein may also comprise alternative C-terminal tail sequences, referred to herein as C-terminal extension sequences. For example, the anti-PCSK9 Adnectin sequences shown in Table 4 may be truncated at the threonine corresponding to T94 of SEQ ID NO: 1 (i.e., truncated after the INYRT (SEQ ID NO: 636) portion of the sequence). Such truncated version may be used as therapeutic molecules in the truncated form, or alternative C-terminal extensions may be added after the threonine residue. Exemplary C-terminal extension sequences are shown in Table 6 as SEQ ID NOs: 380-395. Exemplary anti-PCSK9 Adnectins comprising C-terminal extension sequences are shown in Table 4. For example, SEQ ID NO: 49 (clone 1813E02) comprises the naturally occurring C-terminal extension EIDKPSQ (SEQ ID NO: 380) followed by a His6 tag (SEQ ID NO: 637). However, it should be understood that the His6 tag is completely optional. In certain embodiments, the C-terminal extension sequences (also called “tails”), comprise E and D residues, and may be between 8 and 50, 10 and 30, 10 and 20, 5 and 10, and 2 and 4 amino acids in length. In some embodiments, tail sequences include ED-based linkers in which the sequence comprises tandem repeats of ED. In exemplary embodiments, the tail sequence comprises 2-10, 2-7, 2-5, 3-10, 3-7, 3-5, 3, 4 or 5 ED repeats. In certain embodiments, the ED-based tail sequences may also include additional amino acid residues, such as, for example: EI (SEQ ID NO: 385), EID, ES, EC, EGS, and EGC. Such sequences are based, in part, on known Adnectin tail sequences, such as EIDKPSQ (SEQ ID NO: 380), in which residues D and K have been removed. In exemplary embodiments, the ED-based tail comprises an E, I or EI (SEQ ID NO: 385) residues before the ED repeats. In other embodiments, the N- or C-terminal sequences may be combined with other known linker sequences (e.g., SEQ ID NO: 396-419 in Table 6) as necessary when designing an anti-PCSK9 Adnectin fusion molecule. Exemplary anti-PCSK9 Adnectin comprising linker sequences are shown in Table 4 (e.g., SEQ ID NOs: 53, 55, and 57). In some embodiments, sequences may be placed at the C-terminus of the10Fn3 domain to facilitate attachment of a pharmacokinetic moiety. For example, a cysteine containing linker such as GSGC (SEQ ID NO:77) may be added to the C-terminus to facilitate site directed PEGylation on the cysteine residue. Pharmacokinetic Moieties In one aspect, the application provides for anti-PCSK9 Adnectins further comprising a pharmacokinetic (PK) moiety. Improved pharmacokinetics may be assessed according to the perceived therapeutic need. Often it is desirable to increase bioavailability and/or increase the time between doses, possibly by increasing the time that a protein remains available in the serum after dosing. In some instances, it is desirable to improve the continuity of the serum concentration of the protein over time (e.g., decrease the difference in serum concentration of the protein shortly after administration and shortly before the next administration). The anti-PCSK9 Adnectin may be attached to a moiety that reduces the clearance rate of the polypeptide in a mammal (e.g., mouse, rat, or human) by greater than three-fold relative to the unmodified anti-PCSK9 Adnectin. Other measures of improved pharmacokinetics may include serum half-life, which is often divided into an alpha phase and a beta phase. Either or both phases may be improved significantly by addition of an appropriate moiety. Moieties that tend to slow clearance of a protein from the blood, herein referred to as “PK moieties”, include polyoxyalkylene moieties, e.g., polyethylene glycol, sugars (e.g., sialic acid), and well-tolerated protein moieties (e.g., Fc and fragments and variants thereof, transferrin, or serum albumin). The anti-PCSK9 Adnectin may be fused to albumin or a fragment (portion) or variant of albumin as described in U.S. Publication No. 2007/0048282. In some embodiments, the PCSK9 Adnectin may be fused to one or more serum albumin binding Adnectin, as described herein. In some embodiments, the PK moiety is a serum albumin binding protein such as those described in U.S. Publication Nos. 2007/0178082 and 2007/0269422. In some embodiments, the PK moiety is a serum immunoglobulin binding protein such as those described in U.S. Publication No. 2007/0178082 In some embodiments, the anti-PCSK9 Adnectin comprises polyethylene glycol (PEG). One or more PEG molecules may be attached at different positions on the protein, and such attachment may be achieved by reaction with amines, thiols or other suitable reactive groups. The amine moiety may be, for example, a primary amine found at the N-terminus of a polypeptide or an amine group present in an amino acid, such as lysine or arginine. In some embodiments, the PEG moiety is attached at a position on the polypeptide selected from the group consisting of: a) the N-terminus; b) between the N-terminus and the most N-terminal beta strand or beta-like strand; c) a loop positioned on a face of the polypeptide opposite the target-binding site; d) between the C-terminus and the most C-terminal beta strand or beta-like strand; and e) at the C-terminus. Pegylation may be achieved by site-directed pegylation, wherein a suitable reactive group is introduced into the protein to create a site where pegylation preferentially occurs. In some embodiments, the protein is modified to introduce a cysteine residue at a desired position, permitting site directed pegylation on the cysteine. PEG may vary widely in molecular weight and may be branched or linear. In one embodiment the PEG has two branches. In another embodiment the PEG has four branches. In some embodiments, the anti-PCSK9 Adnectin is fused to an immunoglobulin Fc domain, or a fragment or variant thereof. In an exemplary embodiment, the Fc domain is derived from an IgG1 subclass, however, other subclasses (e.g., IgG2, IgG3, and IgG4) may also be used. Shown below is the sequence of a human IgG1 immunoglobulin Fc domain, and the relative position of each region within the Fc domain are indicated based on the EU numbering format: (SEQ ID NO: 315)ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK The core hinge sequence is underlined, and the CH1 region is italicized; the CH2 and CH3 regions are in regular text. It should be understood that the C-terminal lysine is optional. The fusion may be formed by attaching an anti-PCSK9 Adnectin to either end of the Fc molecule, i.e., Fc-anti-PCSK9 Adnectin or anti-PCSK9 Adnectin-Fc arrangements. In certain embodiments, the Fc and anti-PCSK9 Adnectin are fused via a linker. Exemplary linker sequences include AGGGGSG (SEQ ID NO: 310), GSGSGSGSGSGS (SEQ ID NO: 311), QPDEPGGS (SEQ ID NO: 312), ELQLEESAAEAQDGELD (SEQ ID NO: 313), TVAAPS (SEQ ID NO: 314), KAGGGGSG (SEQ ID NO: 620), KGSGSGSGSGSGS (SEQ ID NO: 621), KQPDEPGGS (SEQ ID NO: 622), KELQLEESAAEAQDGELD (SEQ ID NO: 623), KTVAAPS (SEQ ID NO: 624), KAGGGGSGG (SEQ ID NO: 625), KGSGSGSGSGSGSG (SEQ ID NO: 626), KQPDEPGGSG (SEQ ID NO: 627), KELQLEESAAEAQDGELDG (SEQ ID NO: 628), KTVAAPSG (SEQ ID NO: 629) AGGGGSGG (SEQ ID NO: 630), GSGSGSGSGSGSG (SEQ ID NO: 631), QPDEPGGSG (SEQ ID NO: 632), ELQLEESAAEAQDGELDG (SEQ ID NO: 633), and TVAAPSG (SEQ ID NO: 634). In some embodiments, the Fc region used in the anti-PCSK9 Adnectin fusion comprises the hinge region of an Fc molecule. As used herein, the “hinge” region comprises the core hinge residues spanning positions 104-119 of SEQ ID NO: 315 (DKTHTCPPCPAPELLG; SEQ ID NO: 316) of IgG1, which corresponds to positions 221-236 according to EU numbering. In certain embodiments, the anti-PCSK9 Adnectin-Fc fusion adopts a multimeric structure (e.g., dimer) owing, in part, to the cysteine residues at positions 109 and 112 of SEQ ID NO: 315 (EU numbering 226 and 229, respectively) within the hinge region. In other embodiments, the hinge region as used herein, may further include residues derived from the CH1 and CH2 regions that flank the core hinge sequence, as shown in SEQ ID NO: 315. In some embodiments, the hinge sequence may include substitutions that confer desirable pharmacokinetic, biophysical, and/or biological properties. Some exemplary hinge sequences include EPKSSDKTHTCPPCPAPELLGGPS (SEQ ID NO: 317; core hinge region underlined), EPKSSDKTHTCPPCPAPELLGGSS (SEQ ID NO: 318; core hinge region underlined), EPKSSGSTHTCPPCPAPELLGGSS (SEQ ID NO: 319; core hinge region underlined),DKTHTCPPCPAPELLGGPS (SEQ ID NO: 320; core hinge region underlined), andDKTHTCPPCPAPELLGGSS (SEQ ID NO: 321, core hinge region underlined). In one embodiment, the residue P at position 122 (EU numbering 238) of SEQ ID NO: 315 has been replaced with S to ablate Fc effector function; this replacement is exemplified in hinges having any one of SEQ ID NOs: 318, 319, and 321. In another embodiment, the residues DK at positions 104-105 of SEQ ID NO: 315 (EU numbering 221-222) have been replaced with GS to remove a potential clip site; this replacement is exemplified in SEQ ID NO: 319. In another embodiment, the C at position 103 of SEQ ID NO: 315 (EU numbering 220) has been replaced with S to prevent improper cystine bond formation in the absence of a light chain; this replacement is exemplified in SEQ ID NOs: 317-319. In certain embodiments, an antiPCSK9 Adnectin-Fc fusion may have the following configurations: 1) anti-PCSK9 Adnectin-hinge-Fc or 2) hinge-Fc-anti-PCSK9 Adnectin. Therefore, any anti-PCSK9 Adnectin of the present invention can be fused to an Fc region comprising a hinge sequence according to these configurations. In some embodiments, a linker may be used to join the anti-PCSK9 Adnectin to the hinge-Fc moiety, for example, an exemplary fusion protein may have the configuration hinge-anti-PCSK9 Adnectin-linker-Fc. Additionally, depending on the system in which the fusion polypeptide is produced, a leader sequence may placed at the N-terminus of the fusion polypeptide. For example, if the fusion is produced in a mammalian system, a leader sequence such as METDTLLLWVLLLWVPGSTG (SEQ ID NO: 326) may be added to the N-terminus of the fusion molecule. If the fusion is produced inE. coli, the fusion sequence will be preceded by a methionine. The following sequence exemplifies an anti-PCSK9 Adnectin-hinge-Fc construct produced in a mammalian system: METDTLLLWVLLLWVPGSTGGVSDVPRDLEVVAATPTSLLISWVPPSDDYGYYRITYGET GGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEFPWPHAGYYHRPISINYRTEIEPKSSGS THTCPPCPAPELLGGSSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKA KGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 322). Here, the Fc domain comprises the human IgG1 CH2 and CH3 regions as follows: VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRD ELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 448) and the hinge sequence of SEQ ID NO:319. In SEQ ID NO: 322, the leader sequence is in bold, the anti-PCSK9 Adnectin sequence is in italics, and the hinge region is underlined. It should be understood that the lysine at the end of SEQ ID NO: 322 is optional. The efficacy of the polypeptide fusion as set forth in SEQ ID NO: 322 (also described herein as PRD460) is demonstrated in Example 4. Exemplary PCSK9 Adnectin-Fc fusions are shown in Table 1. All sequences may begin with a methionine or a mammalian leader sequence (e.g., SEQ ID NO: 326). TABLE 1Exemplary Anti-PCSK9 Adnectin-Fc Fusion ProteinsSEQClone orIDNameDescriptionSequencePCSK9 Adnectin-X1-Fc C-Terminal Fusions4521459D05-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWPPPSHGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPPGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPYKHSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4531784F03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWRPPIHAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIVEGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYTFKHSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4541784F03-m1-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPIHAFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPGSEGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYTFKHSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4551784F03-m2-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAHAFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPGSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYTFKHSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4561784F03-m3-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAVTFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPGSKSTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYTFKHSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4571813E02-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWSPPANGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVGRGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYTYKGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4581923B02-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWTPPPKGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVGEGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYTYNGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK4591923B02(N82I)-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWTPPPKGFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVGEGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYTYIGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4601923B02(N82E)-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWTPPPKGFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVGEGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYTYEGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4611923B02(T80A)-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWTPPPKGFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVGEGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYAYNGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4621922G04-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWRPPSHAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPWKGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4631922G04(R25D)-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDPPSHAFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPWKGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4641922G04(R25E)-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPPSHAFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPWKGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKENWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4651922G04(R25S)-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWSPPSHAFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPWKGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;ALPAPIEKTISKAKGQPREPQVYTLPPSRthe Fc may optionallySTYRVVSVLTVLHQDWLNGKEYKCKVSNKinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4662012A04-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWRPPSNGfusionpresent can be selected fromHGYYRITYGETGGNSPVQEFTVPVNEGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFPFKWSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4672013E01-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWVPPSDDfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFPWPHAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4682011H05-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWVPSSHAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVGVGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYAFEGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4692011H05(V23D)-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDPSSHAFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVGVGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYAFEGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4702011H05(V23E)-X1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPSSHAFc fusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVGVGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYAFEGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4712381B02-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPFSRLfusionpresent can be selected fromPGGGEYYRITYGETGGNSPLQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEYPYDYSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKENWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNinclude a C-terminal KGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNV FSCSVMHEALHNHYTQKSLSLSPG4722381B04-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPFSRLfusionpresent can be selected fromPGGGEYYRITYGETGGNSPLQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEYPYEHSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4732381B06-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPFSRLfusionpresent can be selected fromPGGGEYYRITYGETGGNSPLQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEYPYPHSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4742381B08-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPSSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEYTFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4752381D02-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPFSRLfusionpresent can be selected fromPGGGEYYRITYGETGGNSPLQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEYPYDHSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4762381D04-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPFSRLfusionpresent can be selected fromPGGGEYYRITYGETGGNSPLQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEFPYDHSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4772381F11-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKSTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEYTFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4782381G03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPFSRLfusionpresent can be selected fromPGGGEYYRITYGETGGNSPLQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEFPYAHSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4792381G09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGDfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFTFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4802381H03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPFSRLfusionpresent can be selected fromPGGGEYYRITYGETGGNSPLQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEYPYAHSGNO: 384), EIE, and EIEKYFHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4812382A01-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWAAPAGGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEYDFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4822382B10-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPSSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4832382B09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDYPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4842382C05-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromAYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEYSFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4852382C09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAEGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4862382D03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADEfusionpresent can be selected fromAYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFDFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4872382D05-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFDFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4882382D08-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFPFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4892382D09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAEGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDFPGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4902382F02-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFDFPGSGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4912382F03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAADfusionpresent can be selected fromAYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFNFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4922382F05-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAEAfusionpresent can be selected fromGKHYGYYRITYGETGGNSPVQEFTVPVSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEFDFPGAGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKENWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4932382F08-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAEAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFTYPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4942382F09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAAAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4952382G04-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPSSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFDFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4962382H10-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4972382H11-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVFKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDYPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKENWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4982382H04-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAAGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPSSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEYDFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG4992382H07-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPGSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5002382H09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAAAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPSSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5012451A02-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAAGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFPFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5022451B05-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPSSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDYPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5032451B06-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGT(equivalent toE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFDFPGAGYY2382D05)NO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5042451C06-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGAfusionpresent can be selected fromASYGYYRITYGETGGNSPVQEFTVPVSKGE, EI, EID, EIDK (SEQ IDTATISGLKPGVDYTITVYAVEFPFPGAGYNO: 384), EIE, and EIEKYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5052451D05-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5062451F03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDPPAEGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFNFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKENWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5072451G01-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPSSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5082451H07-FcX1 is optional and whenGITDVPRDLEVVAATPTSLLISWNPPDVNfusionpresent can be selected fromYGYYRITYGETGGNSPLQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPYAHAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5092382E03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGDfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFDFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5102382E04-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFTFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5112382E05-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAEGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFDFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5122382E09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAEAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5132381A04-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWEPFSRLfusionpresent can be selected fromPGGGEYYRITYGETGGNSPLQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEYPYPFSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5142381A08-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPGSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEYDFPGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5152381B10-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEYNFIGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5162381C08-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADGfusionpresent can be selected fromAYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFPYPFAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5172381G06-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWSEKLDGfusionpresent can be selected fromKARRGYYRITYGETGGNSPVQQFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEFPYDHSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5182381H01-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWSPRDSTfusionpresent can be selected fromGLVRRGYYRITYGETGGNSPVQQFTVPGSE, EI, EID, EIDK (SEQ IDKGTATISGLKPGVDYTITVYAVEYPYDHSNO: 384), EIE, and EIEKGYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5192381H06-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWGDVRTNfusionpresent can be selected fromEARQGYYRITYGETGGNSPLQGFTVPGSKE, EI, EID, EIDK (SEQ IDGTATISGLKPGVDYTITVYAVEYTYEHSGNO: 384), EIE, and EIEKYYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5202381H09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromGYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEFDFVGAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5212382B11-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAAAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYDFAGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5222382B08-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPSSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFAFPGAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5232382C11-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAGGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYDFAGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5242382G03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAEAfusionpresent can be selected fromEAYGYYRITYGETGGNSPVQEFTVPVSKGE, EI, EID, EIDK (SEQ IDTATISGLKPGVDYTITVYAVEYVFPGAGYNO: 384), EIE, and EIEKYHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5252382H03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAEGfusionpresent can be selected fromAYGYYRITYGETGGNSPVQEFTVPVSKGTE, EI, EID, EIDK (SEQ IDATISGLKPGVDYTITVYAVEYPYPFAGYYNO: 384), EIE, and EIEKHRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5262451A10-FcX1 is optional and whenGVTDVPRDMEVVAATPTSLLISWQPPAVTfusionpresent can be selected fromYGYYRITYGETGGNSTLQQFTVPVYKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPYDHSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKENWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5272451B02-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAAAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFDYPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5282451C11-FcX1 is optional and whenGIVDVPRDLEVVAATPTSLLISWDPPAGAfusionpresent can be selected fromYGYYRITYGETGGNSPKQQFTVPGYKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPYDHSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK5292451H01-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPAAGfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPVSKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYDFPGSGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5302011B11-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWAPPSDAfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEYPYSHAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5312971A03-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDPPSDDfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFPWPHAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5322971A09-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPADDfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFPWPHAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG5332971E02-FcX1 is optional and whenGVSDVPRDLEVVAATPTSLLISWDAPSDDfusionpresent can be selected fromYGYYRITYGETGGNSPVQEFTVPIGKGTAE, EI, EID, EIDK (SEQ IDTISGLKPGVDYTITVYAVEFPWPHAGYYHNO: 384), EIE, and EIEKRPISINYRT-X1-X2-(SEQ ID NO: 635); X2 isVFLFPPKPKDTLMISRTPEVTCVVVDVSHselected from hingeEDPEVKFNWYVDGVEVHNAKTKPREEQYNsequences SEQ ID NOs: 317-321;STYRVVSVLTVLHQDWLNGKEYKCKVSNKthe Fc may optionallyALPAPIEKTISKAKGQPREPQVYTLPPSRinclude a C-terminal KDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGX1-Fc-X2-PCSK9 Adnectin N-Terminal Fusions534Fc-1459D05X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWPPPSHGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPPGKGTATISGLKPGVDYTITVYAVEYPYKHSGYYHRPISINYRT-X3535Fc-1784F03X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWRPPIHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIVEGTATISGLKPGVDYTITVYAVEYTFKHSGYYHRPISINYRT-X3536Fc-1784F03-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHm1 fusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPIHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPGSEGTATISGLKPGVDYTITVYAVEYTFKHSGYYHRPISINYRT-X3537Fc-1784F03-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHm2 fusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOS: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPGSKGTATISGLKPGVDYTITVYAVEYTFKHSGYYHRPISINYRT-X3538Fc-1784F03-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHm3 fusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAVTYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYTITVYAVEYTFKHSGYYHRPISINYRT-X3539Fc-1813E02X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWSPPANGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVGRGTATISGLKPGVDYTITVYAVEYTYKGSGYYHRPISINYRT-X3540Fc-1923B02X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWTPPPKGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVGEGTATISGLKPGVDYTITVYAVEYTYNGAGYYHRPISINYRT-X3541Fc-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSH1923B02(N82I)sequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNfusionX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWTPPPKGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVGEGTATISGLKPGVDYTITVYAVEYTYIGAGYYHRPISINYRT-X3542Fc-X1 is selected from hingeX1-1923B02(N82E)sequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWTPPPKGYGYYRITYGETGGNSPVQEFTVPVGEGTATISGLKPGVDYTITVYAVEYTYEGAGYYHRPISINYRT-X3543Fc-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSH1923B02(T80A)sequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNfusionX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWTPPPKGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVGEGTATISGLKPGVDYTITVYAVEYAYNGAGYYHRPISINYRT-X3544Fc-1922G04X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOS: 380-395 andVSDVPRDLEVVAATPTSLLISWRPPSHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEYPWKGSGYYHRPISINYRT-X3545Fc-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSH1922G04(R25D)sequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNfusionX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDPPSHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEYPWKGSGYYHRPISINYRT-X3546Fc-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSH1922G04(R25E)sequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNfusionX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOS: 380-395 andVSDVPRDLEVVAATPTSLLISWEPPSHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEYPWKGSGYYHRPISINYRT-X3547Fc-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSH1922G04(R25S)sequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNfusionX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWSPPSHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEYPWKGSGYYHRPISINYRT-X3548Fc-2012A04X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWRPPSNGHEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVNEGTATISGLKPGVDYTITVYAVEFPFKWSGYYHRPISINYRT-X3549Fc-2013E01X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWVPPSDDYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEFPWPHAGYYHRPISINYRT-X3550Fc-2011H05X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWVPSSHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVGVGTATISGLKPGVDYTITVYAVEYAFEGAGYYHRPISINYRT-X3551Fc-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSH2011H05(V23D)sequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNfusionX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDPSSHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVGVGTATISGLKPGVDYTITVYAVEYAFEGAGYYHRPISINYRT-X3552Fc-X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSH2011H05(V23E)sequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNfusionX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWEPSSHAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVGVGTATISGLKPGVDYTITVYAVEYAFEGAGYYHRPISINYRT-X3553Fc-2381B02X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKENWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWEPFSRLPEIEK (SEQ ID NO: 635)GGGEYYRITYGETGGNSPLQQFTVPGSKGTATISGLKPGVDYTITVYAVEYPYDYSGYYHRPISINYRT-X3554Fc-2381B04X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWEPFSRLPEIEK (SEQ ID NO: 635)GGGEYYRITYGETGGNSPLQQFTVPGSKGTATISGLKPGVDYTITVYAVEYPYEHSGYYHRPISINYRT-X3555Fc-2381B06X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWEPFSRLPGGGEYYRITYGETGGNSPLQQFTVPGSKGTATISGLKPGVDYTITVYAVEYPYPHSGYYHRPISINYRT-X3556Fc-2381B08X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKENWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPSSKGTATISGLKPGVDYTITVYAVEYTFPGAGYYHRPISINYRT-X3557Fc-2381D02X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWEPFSRLPEIEK (SEQ ID NO: 635)GGGEYYRITYGETGGNSPLQQFTVPGSKGTATISGLKPGVDYTITVYAVEYPYDHSGYYHRPISINYRT-X3558Fc-2381D04X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWEPFSRLPEIEK (SEQ ID NO: 635)GGGEYYRITYGETGGNSPLQQFTVPGSKGTATISGLKPGVDYTITVYAVEFPYDHSGYYHRPISINYRT-X3559Fc-2381F11X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKSTATISGLKPGVDYTITVYAVEYTFPGAGYYHRPISINYRT-X3560Fc-2381G03X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWEPFSRLPEIEK (SEQ ID NO: 635)GGGEYYRITYGETGGNSPLQQFTVPGSKGTATISGLKPGVDYTITVYAVEFPYAHSGYYHRPISINYRT-X3561Fc-2381G09X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOS: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPAGDGYGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFTFPGAGYYHRPISINYRT-X3562Fc-2381H03X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWEPFSRLPGGGEYYRITYGETGGNSPLQQFTVPGSKGTATISGLKPGVDYTITVYAVEYPYAHSGYFHRPISINYRT-X3563Fc-2382A01X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWAAPAGGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYDFPGAGYYHRPISINYRT-X3564Fc-2382B10X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPADAYGYYRITYGETGGNSPVQEFTVPSSKGTATISGLKPGVDYTITVYAVEYDFPGSGYYHRPISINYRT-X3565Fc-2382B09X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDYPGSGYYHRPISINYRT-X3566Fc-2382C05X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPADGAYGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYSFPGAGYYHRPISINYRT-X3567Fc-2382C09X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAEGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGSGYYHRPISINYRT-X3568Fc-2382D03X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADEAEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGAGYYHRPISINYRT-X3569Fc-2382D05X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPADGGYGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGAGYYHRPISINYRT-X3570Fc-2382D08X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFPFPGSGYYHRPISINYRT-X3571Fc-2382D09X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOS: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAEGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGAGYYHRPISINYRT-X3572Fc-2382F02X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGSGYYHRPISINYRT-X3573Fc-2382F03X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAADAEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFNFPGAGYYHRPISINYRT-X3574Fc-2382F05X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAEAGEIEK (SEQ ID NO: 635)KHYGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGAGYYHRPISINYRT-X3575Fc-2382F08X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAEAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFTYPGSGYYHRPISINYRT-X3576Fc-2382F09X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAAAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYDFPGSGYYHRPISINYRT-X3577Fc-2382G04X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPSSKGTATISGLKPGVDYTITVYAVEFDFPGAGYYHRPISINYRT-X3578Fc-2382H10X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOS: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGSGYYHRPISINYRT-X3579Fc-2382H11X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVFKGTATISGLKPGVDYTITVYAVEFDYPGSGYYHRPISINYRT-X3580Fc-2382H04X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOS: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAAGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPSSKGTATISGLKPGVDYTITVYAVEYDFPGAGYYHRPISINYRT-X3581Fc-2382H07X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOS: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPADAYGYYRITYGETGGNSPVQEFTVPGSKGTATISGLKPGVDYTITVYAVEFDFPGSGYYHRPISINYRT-X3582Fc-2382H09X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKENWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAAAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPSSKGTATISGLKPGVDYTITVYAVEFDFPGSGYYHRPISINYRT-X3583Fc-2451A02X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAAGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFPFPGSGYYHRPISINYRT-X3584Fc-2451B05X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKENWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPSSKGTATISGLKPGVDYTITVYAVEFDYPGSGYYHRPISINYRT-X3585Fc-2451B06X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYN(equivalent toX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNK2382D05)linker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGAGYYHRPISINYRT-X3586Fc-2451C06X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGAAEIEK (SEQ ID NO: 635)SYGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFPFPGAGYYHRPISINYRT-X3587Fc-2451D05X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGSGYYHRPISINYRT-X3588Fc-2451F03X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKENWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDPPAEGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFNFPGSGYYHRPISINYRT-X3589Fc-2451G01X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPSSKGTATISGLKPGVDYTITVYAVEFDFPGSGYYHRPISINYRT-X3590Fc-2451H07X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andITDVPRDLEVVAATPTSLLISWNPPDVNYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPLQEFTVPVSKGTATISGLKPGVDYTITVYAVEYPYAHAGYYHRPISINYRT-X3591Fc-2382E03X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPAGDGYGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGAGYYHRPISINYRT-X3592Fc-2382E04X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFTFPGAGYYHRPISINYRT-X3593Fc-2382E05X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAEGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFPGAGYYHRPISINYRT-X3594Fc-2382E09X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAEAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYDFPGSGYYHRPISINYRT-X3595Fc-2381A04X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKENWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWEPFSRLPEIEK (SEQ ID NO: 635)GGGEYYRITYGETGGNSPLQQFTVPGSKGTATISGLKPGVDYTITVYAVEYPYPFSGYYHRPISINYRT-X3596Fc-2381A08X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPADGGYGYYRITYGETGGNSPVQEFTVPGSKGTATISGLKPGVDYTITVYAVEYDFPGAGYYHRPISINYRT-X3597Fc-2381B10X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGGGEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYNFIGAGYYHRPISINYRT-X3598Fc-2381C08X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHFusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADGAEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFPYPFAGYYHRPISINYRT-X3599Fc-2381G06X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWSEKLDGKEIEK (SEQ ID NO: 635)ARRGYYRITYGETGGNSPVQQFTVPGSKGTATISGLKPGVDYTITVYAVEFPYDHSGYYHRPISINYRT-X3600Fc-2381H01X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWSPRDSTGEIEK (SEQ ID NO: 635)LVRRGYYRITYGETGGNSPVQQFTVPGSKGTATISGLKPGVDYTITVYAVEYPYDHSGYYHRPISINYRT-X3601Fc-2381H06X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWGDVRTNEARQGYYRITYGETGGNSPLQGFTVPGSKGTATISGLKPGVDYTITVYAVEYTYEHSGYYHRPISINYRT-X3602Fc-2381H09X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPAGGGYGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDFVGAGYYHRPISINYRT-X3603Fc-2382B11X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAAAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYDFAGSGYYHRPISINYRT-X3604Fc-2382B08X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPADAYGYYRITYGETGGNSPVQEFTVPSSKGTATISGLKPGVDYTITVYAVEFAFPGAGYYHRPISINYRT-X3605Fc-2382C11X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHFusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAGGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYDFAGSGYYHRPISINYRT-X3606Fc-2382G03X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAEAEEIEK (SEQ ID NO: 635)AYGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYVFPGAGYYHRPISINYRT-X3607Fc-2382H03X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAEGAEIEK (SEQ ID NO: 635)YGYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYPYPFAGYYHRPISINYRT-X3608Fc-2451A10X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVTDVPRDMEVVAATPTSLLISWQPPAVTYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSTLQQFTVPVYKGTATISGLKPGVDYTITVYAVEYPYDHSGYYHRPISINYRT-X3609Fc-2451B02X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAAAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEFDYPGSGYYHRPISINYRT-X3610Fc-2451C11X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andIVDVPRDLEVVAATPTSLLISWDPPAGAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPKQQFTVPGYKGTATISGLKPGVDYTITVYAVEYPYDHSGYYHRPISINYRT-X3611Fc-2451H01X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPAAGYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPVSKGTATISGLKPGVDYTITVYAVEYDFPGSGYYHRPISINYRT-X3612Fc-2011B11X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWAPPSDAYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEYPYSHAGYYHRPISINYRT-X3613Fc-2971A03X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDPPSDDYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEFPWPHAGYYHRPISINYRT-X3614Fc-2971A09X1 is selected from hingeX1-VFLFPPKPKDTLMISRTPEVTCVVVDVSHfusionsequences SEQ ID NOs: 317-321;EDPEVKFNWYVDGVEVHNAKTKPREEQYNX2 is selected fromSTYRVVSVLTVLHQDWLNGKEYKCKVSNKlinker sequences SEQ IDALPAPIEKTISKAKGQPREPQVYTLPPSRNOs: 310-314 and 620-634;DELTKNQVSLTCLVKGFYPSDIAVEWESNX3 is optional and whenGQPENNYKTTPPVLDSDGSFFLYSKLTVDpresent can be a C-terminalKSRWQQGNVFSCSVMHEALHNHYTQKSLStail sequence selected fromLSPG-X2-SEQ ID NOs: 380-395 andVSDVPRDLEVVAATPTSLLISWDAPADDYEIEK (SEQ ID NO: 635)GYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEFPWPHAGYYHRPISINYRT-X3615Fc-2971E02X1 is selected from hingeX1-fusionsequences SEQ ID NOs: 317-321;VFLFPPKPKDTLMISRTPEVTCVVVDVSHX2 is selected fromEDPEVKFNWYVDGVEVHNAKTKPREEQYNlinker sequences SEQ IDSTYRVVSVLTVLHQDWLNGKEYKCKVSNKNOs: 310-314 and 620-634;ALPAPIEKTISKAKGQPREPQVYTLPPSRX3 is optional and whenDELTKNQVSLTCLVKGFYPSDIAVEWESNpresent can be a C-terminalGQPENNYKTTPPVLDSDGSFFLYSKLTVDtail sequence selected fromKSRWQQGNVFSCSVMHEALHNHYTQKSLSSEQ ID NOs: 380-395 andLSPG-X2-EIEK (SEQ ID NO: 635)VSDVPRDLEVVAATPTSLLISWDAPSDDYGYYRITYGETGGNSPVQEFTVPIGKGTATISGLKPGVDYTITVYAVEFPWPHAGYYHRPISINYRT-X3 In some embodiments, the anti-PCSK9 Adnectin comprises an Fn3 domain and a PK moiety. In some embodiments, the Fn3 domain is a10Fn3 domain. In some embodiments, the PK moiety increases the serum half-life of the polypeptide by more than 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 150, 200, 400, 600, 800, 1000% or more relative to the Fn3 domain alone. In some embodiments, the PK moiety is a polymeric sugar. In some embodiments, the PK moiety is a polyethylene glycol moiety. In some embodiments the PK moiety is a serum albumin binding protein. In some embodiments the PK moiety is human serum albumin. In some embodiments the PK moiety is a serum immunoglobulin binding protein. In some embodiments, the PK moiety is transferrin. In some embodiments the PK moiety is another Adnectin specific for a serum protein, e.g., HSA. The present application provides specific serum albumin binding Adnectin molecules (or SABA), as described herein. In certain embodiments, a PCSK9 Adnectin fused to a SABA can be defined generally as follows: X1-PCSK9 Adnectin core-X2-X3-X4-SABA core-X5(SEQ ID NO: 618), or X1-SABA core-X2-X3-X4-PCSK9 Adnectin core-X5(SEQ ID NO: 619), wherein X1and X4represent optional N-terminal extension sequences, X2and X5represent optional C-terminal extension sequences, and X3is a linker. In one embodiment, the Adnectins (either PCSK9 or serum albumin binding) of SEQ ID NOs: 618 and 619 comprise the “core” region of Adnectin, i.e., a PCSK9 Adnectin core sequence may be any one of the PCSK9 Adnectin sequences shown in Table 4, wherein the sequence begins at the amino acid residue corresponding to E8 of SEQ ID NO:1 and ends at the amino acid corresponding to residue T94 of SEQ ID NO:1; and a SABA core sequence may be selected from any of the SABA core sequences shown in Table 6. In some embodiments, X1and X4are independently optional, and when present are independently selected from SEQ ID NOs: 371-379 listed in Table 6, and may optionally comprise an M, G or MG sequence at the N-terminus when such residues are not already present. For expression in a mammalian system, the fusion proteins may further comprise a leader sequence at the N-terminus, such as METDTLLLWVLLLWVPGSTG (SEQ ID NO: 326). In some embodiments, X2and X5are independently optional, and when present are independently selected from SEQ ID NOs: 380-395 listed in Table 6. In certain embodiments, X3is a linker sequence selected from SEQ ID NOs: 396-419 listed in Table 6. The sequences shown in Table 2 represent exemplary fusions of anti-PCSK9 Adnectin and SABA. It should be understood that any PCSK9 Adnectin and SABA sequence described in the present application may be incorporated into these configurations. TABLE 2Exemplary PCSK9 Adnectin - SABA Fusion SequencesSEQCloneIDor NameDescriptionSequence616PCSK9PCSK9 Adnectin sequence isX1-Adnectin-underlined; SABA sequence isEVVAATPTSLLISWVPPSDDYGYYRITYSABA fusionin bold. PCSK9 AdnectinGETGGNSPVQEFTVPIGKGTATISGLKPsequence is the core regionGVDYTITVYAVEFPWPHAGYYHRPISINderived from clone 2013E01;YRT-X2-X3-X4-SABA sequence is derivedEVVAATPTSLLISWHSYYEQNSYYRITYfrom SABA 1 (SEQ ID NO:GETGGNSPVQEFTVPYSQTTATISGLKP330)GVDYTITVYAVYGSKYYYPISINYRT-X5617SABA-PCSK9SABA sequence is in bold;X1-AdnectinPCSK9 Adnectin sequence isEVVAATPTSLLISWHSYYEQNSYYRITYfusionunderlined. SABA sequenceGETGGNSPVQEFTVPYSQTTATISGLKPis derived from SABA 1 (SEQGVDYTITVYAVYGSKYYYPISINYRT-ID NO: 330); PCSK9X2-X3-X4-Adnectin sequence is the coreEVVAATPTSLLISWVPPSDDYGYYRITYregion derived from cloneGETGGNSPVQEFTVPIGKGTATISGLKP2013E01GVDYTITVYAVEFPWPHAGYYHRPISINYRT-X5 Biophysical and Biochemical Characterization The application provides Adnectin comprising a Fn3 domain that binds to PCSK9. Polypeptide binding to a target molecule may be assessed in terms of equilibrium constants (e.g., dissociation, KD) and in terms of kinetic constants (e.g., on-rate constant, Konand off-rate constant, koff). An Adnectin will generally bind to a target molecule with a KDof less than 500 nM, 100 nM, 10 nM, 1 nM, 500 pM, 200 pM, 100 pM, although higher KDvalues may be tolerated where the Koffis sufficiently low or the Kon, is sufficiently high. The SEQ ID NOS of the BC, DE and FG loops of the anti-PCSK9 Adnectins of the invention are presented in italic in Table 3. TABLE 3Anti-PCSK9 Adnectin BC, DE and FG LoopsLDLRDepletionPCSK9-(% inhibitionAffinityEGFA FRETat 75 nM, EC50SEQ IDSEQ IDSEQ IDClone ID(KD, nM)(EC50, nM)(nM))BC LoopNODE LoopNOFG loopNO1459D0514.4†466.8, >200SWPPPSHGYG2PPGKGT18EYPYKHSGYYHRP281.58*26^1784F033.8^2150.2, 26 ± 13SWRPPIHAYG3PIVEGT19EYTFKHSGYYHRP291784F03-m1ndndnd, >2000SWDAPIHAYG4PGSEGT20EYTFKHSGYYHRP291784F03-m2ndndnd, >2000SWDAPAHAYG5PGSKGT21EYTFKHSGYYHRP291784F03-m3ndndnd, >2000SWDAPAVTYG6PGSKST22EYTFKHSGYYHRP291813E02<2^1.3nd, 16SWSPPANGYG7PVGRGT23EYTYKGSGYYHRP301923B020.173*2.3178.0, 23 ± 7SWTPPPKGYG8PVGEGT24EYTYNGAGYYHRP311923B02(N82I)ndndnd, 14SWTPPPKGYG8PVGEGT24EYTYIGAGYYHRP321923B02(N82E)ndndnd, 28SWTPPPKGYG8PVGEGT24EYTYEGAGYYHRP331923B02(T80A)ndndnd, 42SWTPPPKGYG8PVGEGT24EYAYNGAGYYHRP341922G040.09*1.2105.1, 10 ± 2SWRPPSHAYG9PIGKGT25EYPWKGSGYYHRP351922G04(R25D)nd2.5nd, 29 ± 8SWDPPSHAYG10PIGKGT25EYPWKGSGYYHRP351922G04(R25E)nd3.5nd, 29 ± 18SWEPPSHAYG11PIGKGT25EYPWKGSGYYHRP351922G04(R25S)ndndnd, 21SWSPPSHAYG12PIGKGT25EYPWKGSGYYHRP352012A040.25*2.1144.5, 12 ± 6SWRPPSNGHG13PVNEGT26EFPFKWSGYYHRP362013E011.51†1.6165.5, 10 ± 4SWVPPSDDYG14PIGKGT25EFPWPHAGYYHRP370.29*2011H050.08*2.7197.6, 12 ± 5SWVPSSHAYG15PVGVGT27EYAFEGAGYYHRP382011H05(V23D)nd5.5nd, 18 ± 3SWDPSSHAYG16PVGVGT27EYAFEGAGYYHRP382011H05(V23E)nd7.4nd, 12 ± 3SWEPSSHAYG17PVGVGT27EYAFEGAGYYHRP382381B02(1)3.29*2.5125.4, ndSWEPFSRLPGGGE106PGSKGT21EYPYDYSGYYHRP1422381B04(1)0.527†2.4121.6, ndSWEPFSRLPGGGE106PGSKGT21EYPYEHSGYYHRP1432381B06(1)nd3.5119.7, ndSWEPFSRLPGGGE106PGSKGT21EYPYPHSGYYHRP1442381B084.11†2.6124.8, ndSWDAPADGGYG107PSSKGT136EYTFPGAGYYHRP1452381D02(1)nd3.1185.0, ndSWEPFSRLPGGGE106PGSKGT21EYPYDHSGYYHRP1462381D04(1)0.237†2.9119.2, ndSWEPFSRLPGGGE106PGSKGT21EFPYDHSGYYHRP1472381F111.59*4110.2, ndSWDAPADGGYG107PVSKST137EYTFPGAGYYHRP1452381G03(1)nd3.470.2, ndSWEPFSRLPGGGE106PGSKGT21EFPYAHSGYYHRP1482381G091.12†3.1133.0, ndSWDAPAGDGYG108PVSKGT138EFTFPGAGYYHRP1492381H03(1)nd3.489.8, ndSWEPFSRLPGGGE106PGSKGT21EYPYAHSGYFHRP1502382A01nd12.9119.8, ndSWAAPAGGGYG109PVSKGT138EYDFPGAGYYHRP1512382B102.35†3100.2, ndSWDAPADAYG110PSSKGT136EYDFPGSGYYHRP1522382B090.656†3.8105.0, ndSWDAPADAYG110PVSKGT138EFDYPGSGYYHRP1532382C052.49†4105.3, ndSWDAPADGAYG111PVSKGT138EYSFPGAGYYHRP1542382C090.757†3.5121.7, ndSWDAPAEGYG112PVSKGT138EFDFPGSGYYHRP1552382D031.53*3.380.4, ndSWDAPADEAYG113PVSKGT138EFDFPGAGYYHRP1562382D050.314†2.6140.5, ndSWDAPADGGYG107PVSKGT138EFDFPGAGYYHRP1562382D08nd3.1106.6, ndSWDAPADGYG114PVSKGT138EFPFPGSGYYHRP1572382D090.304†2.6109.1, ndSWDAPAEGYG112PVSKGT138EFDFPGAGYYHRP1562382F02nd2.6−6.3, ndSWDAPAGGGYG115PVSKGT138EFDFPGSGYYHRP1552382F03nd2.788.6, ndSWDAPAADAYG116PVSKGT138EFNFPGAGYYHRP1582382F054.54†2.472.2, ndSWDAPAEAGKHYG117PVSKGT138EFDFPGAGYYHRP1562382F08nd2.5105.0, ndSWDAPAEAYG118PVSKGT138EFTYPGSGYYHRP1592382F09nd3.1109.7, ndSWDAPAAAYG119PVSKGT138EYDFPGSGYYHRP1522382G041.11†2.9146.1, ndSWDAPAGGGYG115PSSKGT136EFDFPGAGYYHRP1562382H101.40†2.6118.6, ndSWDAPAGGYG120PVSKGT138EFDFPGSGYYHRP1552382H11nd2.9117.2, ndSWDAPADGYG114PVFKGT139EFDYPGSGYYHRP1532382H04nd3.268.2, ndSWDAPAAGGYG121PSSKGT136EYDFPGAGYYHRP1512382H07nd2.786.2, ndSWDAPADAYG110PGSKGT21EFDFPGSGYYHRP1552382H091.86*0.9101.2, ndSWDAPAAAYG119PSSKGT136EFDFPGSGYYHRP1552451A02nd3.2106.4, ndSWDAPAAGYG122PVSKGT138EFPFPGSGYYHRP1572451B05nd6.391.7, ndSWDAPAGGYG120PSSKGT136EFDYPGSGYYHRP1532451B06nd4.592.2, ndSWDAPADGGYG107PVSKGT138EFDFPGAGYYHRP1562451C061.27†1.289.4, ndSWDAPAGAASYG123PVSKGT138EFPFPGAGYYHRP1602451D05nd2.8115.0, ndSWDAPAGAYG124PVSKGT138EFDFPGSGYYHRP1552451F03nd2.8113.2, ndSWDPPAEGYG125PVSKGT138EFNFPGSGYYHRP1612451G01nd3.890.8, ndSWDAPAGGYG120PSSKGT136EFDFPGSGYYHRP1552451H07(2)2.08†0.288.8, ndSWNPPDVNYG126PVSKGT138EYPYAHAGYYHRP1622382E032.94†2.489.5, ndSWDAPAGDGYG108PVSKGT138EFDFPGAGYYHRP1562382E04nd361.5, ndSWDAPAGGGYG115PVSKGT138EFTFPGAGYYHRP1492382E050.604†2.8103.5, ndSWDAPAEGGYG127PVSKGT138EFDFPGAGYYHRP1562382E09nd6.297.2, ndSWDAPAEAYG118PVSKGT138EYDFPGSGYYHRP1522381A04(1)nd3.3100.1, ndSWEPFSRLPGGGE106PGSKGT21EYPYPFSGYYHRP1632381A08nd3.691.4, ndSWDAPADGGYG107PGSKGT21EYDFPGAGYYHRP1512381B10nd7.396.4, ndSWDAPAGGGYG115PVSKGT138EYNFIGAGYYHRP1642381C08nd0.715.3, ndSWDAPADGAYG111PVSKGT138EFPYPFAGYYHRP1652381G06(3)nd957.7, ndSWSEKLDGKARRG128PGSKGT21EFPYDHSGYYHRP1472381H01(3)nd422.2, ndSWSPRDSTGLVRRG129PGSKGT21EYPYDHSGYYHRP1462381H06(4)nd553.4, ndSWGDVRTNEARQG130PGSKGT21EYTYEHSGYYHRP1662381H093.23†3.494.4, ndSWDAPAGGGYG115PVSKGT138EFDFVGAGYYHRP1672382B11nd2.988.8, ndSWDAPAAAYG119PVSKGT138EYDFAGSGYYHRP1682382B08nd2.9107.2, ndSWDAPADAYG110PSSKGT136EFAFPGAGYYHRP1692382C11nd3.782.9, ndSWDAPAGGYG120PVSKGT138EYDFAGSGYYHRP1682382G03nd2.777.8, ndSWDAPAEAEAYG131PVSKGT138EYVFPGAGYYHRP1702382H030.677†3.4102.1, ndSWDAPAEGAYG132PVSKGT138EYPYPFAGYYHRP1712451A10(5)nd10.953.7, ndSWQPPAVTYG133PVYKGT140EYPYDHSGYYHRP1462451B02nd5.371.4, ndSWDAPAAAYG119PVSKGT138EFDYPGSGYYHRP1532451C11(6)nd9.770.3, ndSWDPPAGAYG134PGYKGT141EYPYDHSGYYHRP1462451H01nd2.895.8, ndSWDAPAAGYG122PVSKGT138EYDFPGSGYYHRP1522011B11nd1.7144.5, ndSWAPPSDAYG135PIGKGT25EYPYSHAGYYHRP1722971A030.806†nd120.1, ndSWDPPSDDYG301PIGKGT25EFPWPHAGYYHRP372971A092.79†nd132.3, ndSWDAPADDYG302PIGKGT25EFPWPHAGYYHRP372971E021.78*nd126.2, ndSWDAPSDDYG303PIGKGT25EFPWPHAGYYHRP37†KDdetermined using Octet Red at 37° C.; *KDdetermined using ProteOn at 25° C.; ^KDdetermined using ITC at 37° C.; (1)In addition to mutations in the loops, these clones also have the mutations V45L and E47Q; (2)In addition to mutations in the loops, this clone also has the mutations VII, S2T, and V45L; (3)In addition to mutations in the loops, these clones also have the mutation E47Q; (4)In addition to mutations in the loops, this clone also has the mutations V45L and E47G; (5)In addition to mutations in the loops, this clone also has the mutations S2T, L8M, P44T, V45L, and E47Q; (6)In addition to mutations in the loops, this clone also has the mutations VII, S2V, V45K, and E47Q. The SEQ ID NOS of the family of anti-PCSK9 Adnectin of the invention are presented in Table 4. TABLE 4Anti-PCSK9 Adnectin FamilySequenceCloneAmino AcidNucleic Acid1459D05 alsoMGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAreferred to asSWPPPSHGYGYYRITYGETGGNTCAGCTGGCCGCCGCCGTCTCATGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGATI000891 orSPVQEFTVPPGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCGCCTGGTAAAGGTACAGCTACCATCAGCGGCCTTATI-891GVDYTITVYAVEYPYKHSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACAAACATTCTGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 39)CCACCACCAC (SEQ ID NO: 40)1784F03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWRPPIHAYGYYRITYGETGGNTCAGCTGGAGGCCGCCGATTCATGCTTACGGGTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPIVEGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGTTGAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYTFKHSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATATACATTTAAACATTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 41)CCACCACCAC (SEQ ID NO: 42)1784F03-mlMGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPIHAYGYYRITYGETGGNTCAGCTGGGACGCTCCGATTCATGCTTACGGGTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPGSEGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGGTTCTGAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYTFKHSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATATACATTTAAACATTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 43)CCACCACCAC (SEQ ID NO: 44)1784F03-m2MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAHAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTCATGCTTACGGGTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPGSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYTFKHSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATATACATTTAAACATTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 45)CCACCACCAC (SEQ ID NO: 46)1784F03-m3MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAVTYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGTTACTTACGGGTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPGSKSTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGGTTCTAAATCTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYTFKHSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATATACATTTAAACATTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 47)CCACCACCAC (SEQ ID NO: 48)1813E02MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWSPPANGYGYYRITYGETGGNTCAGCTGGTCCCCACCGGCTAACGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVGRGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTGGTAGAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYTYKGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTATACCTACAAAGGCTCTGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGCCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 49)CCACCACCAC (SEQ ID NO: 50)1923B02MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWTPPPKGYGYYRITYGETGGNTCAGCTGGACGCCTCCCCCTAAAGGGTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVGEGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTGGTGAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYTYNGAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACACGTACAACGGTGCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCACCGGCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 51)CCACCACCAC (SEQ ID NO: 52)1923B02(N82I)MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWTPPPKGYGYYRITYGETGGNTCAGCTGGACGCCTCCCCCTAAAGGGTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVGEGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTGGTGAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYTYIGAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACACGTACATTGGTGCCGGTTRPISINYRTGSGSHHHHHHACTACCACCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTCCCACCATCACCACCATCA(SEQ ID NO: 53)C (SEQ ID NO: 54)1923B02(N82E)MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWTPPPKGYGYYRITYGETGGNTCAGCTGGACGCCTCCCCCTAAAGGGTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVGEGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTGGTGAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYTYEGAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACACGTACGAAGGTGCCGGTTRPISINYRTGSGSHHHHHHACTACCACCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTCCCACCATCACCACCATCA(SEQ ID NO: 55)C (SEQ ID NO: 56)1923B02(T80A)MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWTPPPKGYGYYRITYGETGGNTCAGCTGGACGCCTCCCCCTAAAGGGTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVGEGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTGGTGAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYAYNGAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGCGTACAACGGTGCCGGTTRPISINYRTGSGSHHHHHHACTACCACCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTCCCACCATCACCACCATCA(SEQ ID NO: 57)C (SEQ ID NO: 58)1922G04 alsoMGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAreferred toSWRPPSHAYGYYRITYGETGGNTCAGCTGGCGGCCGCCATCTCATGCTTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGherein asSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGGAAAGGTACAGCTACCATCAGCGGCCTTATI001057 orGVDYTITVYAVEYPWKGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTGGAAAGGTTCTGGTTATI-1057RPISINYRTEIDKPSQHHHHHHACTACCATCGGCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 59)CCACCACCAC (SEQ ID NO: 60)1922G04(R25D)MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDPPSHAYGYYRITYGETGGNTCAGCTGGGACCCGCCATCTCATGCTTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGGAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYPWKGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTGGAAAGGTTCTGGTTRPISINYRTGSGSHHHHHHACTACCATCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTCCCACCATCACCACCATCA(SEQ ID NO: 61)C (SEQ ID NO: 62)1922G04(R25E)MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPPSHAYGYYRITYGETGGNTCAGCTGGGAACCGCCATCTCATGCTTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGGAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYPWKGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTGGAAAGGTTCTGGTTRPISINYRTGSGSHHHHHHACTACCATCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTCCCACCATCACCACCATCA(SEQ ID NO: 63)C (SEQ ID NO: 64)1922G04(R25S)MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWSPPSHAYGYYRITYGETGGNTCAGCTGGAGCCCGCCATCTCATGCTTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGGAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYPWKGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTGGAAAGGTTCTGGTTRPISINYRTGSGSHHHHHHACTACCATCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTCCCACCATCACCACCATCA(SEQ ID NO: 65)C (SEQ ID NO: 66)2012A04MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWRPPSNGHGYYRITYGETGGNTCAGCTGGCGGCCCCCCTCTAATGGTCACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVNEGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTAATGAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFPFKWSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCCTTCAAGTGGTCGGGCTRPISINYRTEIDKPSQHHHHHHACTACCATCGACCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 67)CCACCACCAC (SEQ ID NO: 68)2013E01 alsoMGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAreferred to asSWVPPSDDYGYYRITYGETGGNTCAGCTGGGTCCCGCCTTCAGATGATTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGATI001081 orSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGAACAGCTACCATCAGCGGCCTTATI-1081GVDYTITVYAVEFPWPHAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTTTCCGTGGCCACATGCTGGTTRPISINYRTEIDKPSQHHHHHHACTATCATCGGCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 69)CCACCACCAC (SEQ ID NO: 70)2011H05 alsoMGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAreferred to asSWVPSSHAYGYYRITYGETGGNTCAGCTGGGTTCCGTCGTCTCATGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGATI001091 orSPVQEFTVPVGVGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTGGGGGTAGGTACAGCTACCATCAGCGGCCTTATI-1091GVDYTITVYAVEYAFEGAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGCTTTCGAAGGGGCTGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 71)CCACCACCAC (SEQ ID NO: 72)2011H05(V23D)MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDPSSHAYGYYRITYGETGGNTCAGCTGGGACCCGTCGTCTCATGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVGVGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTGGGGGTAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYAFEGAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGCTTTCGAAGGGGCTGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 73)CCACCACCAC (SEQ ID NO: 74)2011H05(V23E)MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPSSHAYGYYRITYGETGGNTCAGCTGGGAACCGTCGTCTCATGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVGVGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTGGGGGTAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYAFEGAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGCTTTCGAAGGGGCTGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 75)CCACCACCAC (SEQ ID NO: 76)2381B02MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPFSRLPGGGEYYRITYGETTCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAGGNSPLQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEYPYDYSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACGACTYYHRPISINYRTEIDKPSQHHHATTCTGGTTACTACCATCGCCCCATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 173)GCACCATCACCACCACCAC (SEQ ID NO: 174)2381B04MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPFSRLPGGGEYYRITYGETTCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAGGNSPLQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEYPYEHSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACGAGCYYHRPISINYRTEIDKPSQHHHATTCTGGGTACTATCATCGTCCGATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 175)GCACCATCACCACCACCAC (SEQ ID NO: 176)2381B06MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPFSRLPGGGEYYRITYGETTCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAGGNSPLQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEYPYPHSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACCCGCYYHRPISINYRTEIDKPSQHHHATTCTGGTTACTACCATCGACCGATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO : 177)GCACCATCACCACCACCAC (SEQ ID NO: 178)2381B08MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGATGGAGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPSSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTAGTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEYTFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACACCTTCCCGGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO : 179)TCACCACCACCAC (SEQ ID NO: 180)2381D02MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPFSRLPGGGEYYRITYGETTCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAGGNSPLQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEYPYDHSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACGACCYYHRPISINYRTEIDKPSQHHHATTCTGGTTACTACCATCGTCCCATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 181)GCACCATCACCACCACCAC (SEQ ID NO: 182)2381D04MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPFSRLPGGGEYYRITYGETTCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAGGNSPLQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEFPYDHSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCGTACGACCYYHRPISINYRTEIDKPSQHHHATTCTGGTTACTACCATCGGCCCATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 183)GCACCATCACCACCACCAC (SEQ ID NO: 184)2381F11MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGACGGGGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKSTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAAGTACAGCTACCATCAGCGGCPGVDYTITVYAVEYTFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACACCTTCCCCGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 185)TCACCACCACCAC (SEQ ID NO : 186)2381G03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPFSRLPGGGEYYRITYGETTCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAGGNSPLQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEFPYAHSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCGTACGCGCYYHRPISINYRTEIDKPSQHHHATTCTGGGTACTACCATCGTCCGATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 187)GCACCATCACCACCACCAC (SEQ ID NO: 188)2381G09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGDGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGGGGACGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCCGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFTFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCACCTTCCCGGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO : 189)TCACCACCACCAC (SEQ ID NO: 190)2381H03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPFSRLPGGGEYYRITYGETTCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAGGNSPLQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEYPYAHSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACGCGCYFHRPISINYRTEIDKPSQHHHATTCTGGTTACTTCCATCGTCCGATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO : 191)GCACCATCACCACCACCAC (SEQ ID NO: 192)2382A01MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWAAPAGGGYGYYRITYGETGGTCAGCTGGGCCGCTCCGGCTGGTGGTGGCTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEYDFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGACTTCCCGGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 193)TCACCACCACCAC (SEQ ID NO: 194)2382B10MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADAYGYYRITYGETGGNTAAGCTGGGACGCTCCGGCTGACGCGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPSSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTAGTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGACTTCCCCGGCAGCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO : 195)CCACCACCAC (SEQ ID NO: 196)2382B09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGACGCGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDYPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTACCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 197)CCACCACCAC (SEQ ID NO: 198)2382C05MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADGAYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGATGGGGCATACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAGGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEYSFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACTCCTTCCCCGGCGCCGHRPISINYRTEIDKPSQHI HGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 199)TCACCACCACCAC (SEQ ID NO: 200)2382C09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAEGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGAGGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 201)CCACCACCAC (SEQ ID NO: 202)2382D03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADEAYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGACGAGGCGTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFDFPGAGYYCTTAAACCTGGTGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 203)TCACCACCACCAC (SEQ ID NO: 204)2382D05MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGATGGTGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFDFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCGGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 205)TCACCACCACCAC (SEQ ID NO: 206)2382D08MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGATGGCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFPFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCCTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 207)CCACCACCAC (SEQ ID NO: 208)2382D09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAEGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGAAGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDFPGAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCGCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 209)CCACCACCAC (SEQ ID NO: 210)2382F02MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGGCGGGGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFDFPGSGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCGGGCTCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 211)TCACCACCACCAC (SEQ ID NO: 212)2382F03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAADAYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGCCGATGCTTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFNFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCAACTTCCCGGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 213)TCACCACCACCAC (SEQ ID NO: 214)2382F05MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAEAGKHYGYYRITYGETTCAGCTGGGACGCTCCGGCTGAAGCAGGTAAGCACTACGGTTATTACCGCATCACTTACGGCGAGGNSPVQEFTVPVSKGTATISGAACAGGAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEFDFPGAGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCGGYYHRPISINYRTEIDKPSQHHHGCGCCGGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 215)GCACCATCACCACCACCAC (SEQ ID NO: 216)2382F08MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAEAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGAAGCATACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFTYPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCACCTACCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 217)CCACCACCAC (SEQ ID NO: 218)2382F09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAAAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGCAGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 219)CCACCACCAC (SEQ ID NO: 220)2382G04MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGGTGGGGGATACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPSSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTAGTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFDFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCGGGCGCCGHRPISINYRTEIDKPSQHEGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 221)TCACCACCACCAC (SEQ ID NO: 222)2382H10MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGGGGGCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO:223)CCACCACCAC (SEQ ID NO: 224)2382H11MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGATGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVFKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTTTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDYPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTACCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 225)CCACCACCAC (SEQ ID NO: 226)2382H04MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAAGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGCGGGGGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPSSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTAGTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEYDFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTATATGCTGTCGAATACGACTTCCCCGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 227)TCACCACCACCAC (SEQ ID NO: 228)2382H07MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGATGCTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPGSKGTATISGLKPCAATAGCCCAGTCCAGGAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 229)CCACCACCAC (SEQ ID NO: 230)2382H09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAAAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGCGGCTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPSSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTAGTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGCCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 231)CCACCACCAC (SEQ ID NO: 232)2451A02MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAAGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGCGGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFPFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCCTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 233)CCACCACCAC (SEQ ID NO: 234)2451B05MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGGGGGATACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPSSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTAGTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDYPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTACCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 235)CCACCACCAC (SEQ ID NO: 236)2451B06MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGA(equivalent toSWDAPADGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGATGGTGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGG2382D05)NSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFDFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCGGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 205)TCACCACCACCAC (SEQ ID NO: 206)2451C06MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGAASYGYYRITYGETGTCAGCTGGGACGCTCCGGCTGGGGCAGCGTCCTACGGTTATTACCGCATCACTTACGGCGAAACGNSPVQEFTVPVSKGTATISGLAGGAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCKPGVDYTITVYAVEFPFPGAGYGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCCTTCCCCGGCGYHRPISINYRTEIDKPSQHHHHCCGGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCAHH (SEQ ID NO: 237)CCATCACCACCACCAC (SEQ ID NO:238)2451D05MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGGCGCGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 239)CCACCACCAC (SEQ ID NO: 240)2451F03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDPPAEGYGYYRITYGETGGNTCAGCTGGGACCCTCCGGCTGAAGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFNFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCAACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 241)CCACCACCAC (SEQ ID NO: 242)2451G01MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGGGGGCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPSSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTAGTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCGGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 243)CCACCACCAC (SEQ ID NO: 244)2451H07MGITDVPRDLEVVAATPTSLLIATGGGTATCACGGATGTGCCGCGAGACTTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWNPPDVNYGYYRITYGETGGNTCAGCTGGAACCCGCCGGATGTGAATTACGGTTATTATCGCATCACTTACGGGGAAACAGGAGGSPLQEFTVPVSKGTATISGLKPCAATAGCCCTTTGCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYPYAHAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATATCCGTACGCGCACGCTGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCGATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 245)CCACCACCAC (SEQ ID NO: 246)2382E03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGDGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGGGGACGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFDFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 247)TCACCACCACCAC (SEQ ID NO: 248)2382E04MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGGTGGTGGATACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFTFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCACCTTCCCGGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 249)TCACCACCACCAC (SEQ ID NO: 250)2382E05MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAEGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGAGGGCGGCTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFDFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 251)TCACCACCACCAC (SEQ ID NO: 252)2382E09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAEAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGAGGCTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 253)CCACCACCAC (SEQ ID NO: 254)2381A04MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWEPFSRLPGGGEYYRITYGETTCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAGGNSPLQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEYPYPFSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACCCGTYYHRPISINYRTEIDKPSQHHHTTTCTGGTTACTACCATCGTCCCATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 255)GCACCATCACCACCACCAC (SEQ ID NO: 256)2381A08MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGACGGCGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPGSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEYDFPGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGACTTCCCGGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 257)TCACCACCACCAC (SEQ ID NO: 258)2381B10MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGGGGGTGGATACGGTTATTACCGSATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEYNFIGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACAACTTCATCGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 259)TCACCACCACCAC (SEQ ID NO: 260)2381C08MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADGAYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGACGGTGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFPYPFAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCCTACCCCTTCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 261)TCACCACCACCAC (SEQ ID NO: 262)2381G06MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWSEKLDGKARRGYYRITYGETTCAGCTGGTCGGAGAAGTTGGACGGGAAGGCGCGCCGCGGGTATTACCGCATCACATACGGCGAGGNSPVQQFTVPGSKGTATISGAACAGGAGGCAATAGCCCTGTCCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEFPYDHSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCGTACGACCYYHRPISINYRTEIDKPSQHHHATTCTGGTTACTACCATCGTCCCATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 263)GCACCATCACCACCACCAC (SEQ ID NO: 264)2381H01MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWSPRDSTGLVRRGYYRITYGETCAGCTGGAGCCCGCGGGACTCCACCGGCTTGGTGAGGCGGGGGTATTACCGCATCACTTACGGTGGNSPVQQFTVPGSKGTATISCGAAACAGGAGGCAATAGCCCTGTTCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCGLKPGVDYTITVYAVEYPYDHSATCAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACGGYYHRPISINYRTEIDKPSQHHACCATTCTGGTTACTACCATCGGCCCATTTCCATTAATTACCGCACAGAAATTGACAAACCATCHHHH (SEQ ID NO: 265)CCAGCACCATCACCACCACCAC (SEQ ID NO: 266)2381H06MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWGDVRTNEARQGYYRITYGETTCAGCTGGGGCGACGTCCGGACGAACGAGGCGCGGCAGGGCTATTACCGGATCACTTACGGCGAGGNSPLQGFTVPGSKGTATISGAACAGGAGGCAATAGCCCTCTCCAGGGGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCLKPGVDYTITVYAVEYTYEHSGAGCGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTATACGTACGAGCYYHRPISINYRTEIDKPSQHHHATTCTGGTTACTACCATCGTCCGATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAHHH (SEQ ID NO: 267)GCACCATCACCACCACCAC (SEQ ID NO: 268)2381H09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGGYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGGGGGGGGCTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEFDFVGAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCGTCGGCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 269)TCACCACCACCAC (SEQ ID NO: 270)2382B11MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAAAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGCGGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYDFAGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGACTTCGCGGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 271)CCACCACCAC (SEQ ID NO: 272)2382B08MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGACGCGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPSSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTAGTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFAFPGAGYYHAAACCTGGCGTTGATTATACCATCACTGTATATGCTGTCGAATTCGCCTTCCCCGGCGCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 273)CCACCACCAC (SEQ ID NO: 274)2382C11MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAGGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGGAGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYDFAGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGACTTCGCGGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 275)CCACCACCAC (SEQ ID NO: 276)2382G03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAEAEAYGYYRITYGETGTCAGCTGGGACGCTCCGGCTGAAGCAGAAGCGTACGGTTATTACCGCATCACTTACGGCGAAACGNSPVQEFTVPVSKGTATISGLAGGAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCKPGVDYTITVYAVEYVFPGAGYGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGTCTTCCCCGGCGYHRPISINYRTEIDKPSQHHHHCCGGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCAHH (SEQ ID NO: 277)CCATCACCACCACCAC (SEQ ID NO: 278)2382H03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAEGAYGYYRITYGETGGTCAGCTGGGACGCTCCGGCTGAGGGCGCTTACGGTTATTACCGCATCACTTACGGCGAAACAGGNSPVQEFTVPVSKGTATISGLKAGGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCPGVDYTITVYAVEYPYPFAGYYCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCCTACCCCTTCGCCGHRPISINYRTEIDKPSQHHHHHGTTACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCAH (SEQ ID NO: 279)TCACCACCACCAC (SEQ ID NO: 280)2451A10MGVTDVPRDMEVVAATPTSLLIATGGGTGTCACCGATGTGCCGCGCGACATGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWQPPAVTYGYYRITYGETGGNTCAGCTGGCAGCCGCCGGCTGTTACTTACGGTTATTATCGCATCACTTACGGCGAAACAGGAGGSTLQQFTVPVYKGTATISGLKPCAATAGCACTCTCCAGCAGTTCACTGTGCCTGTTTATAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYPYDHSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACGACCATTCTGGGTRPISINYRTEIDKPSQHHHHHHACTACCATCGGCCGATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 281)CCACCACCAC (SEQ ID NO: 282)2451B02MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAAAYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGCTGCTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFDYPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTACCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 283)CCACCACCAC (SEQ ID NO: 284)2451C11MGIVDVPRDLEVVAATPTSLLIATGGGTATCGTGGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDPPAGAYGYYRITYGETGGNTCAGCTGGGACCCGCCGGCTGGTGCTTACGGTTATTATCGCATCACTTACGGCGAAACAGGAGGSPKQQFTVPGYKGTATISGLKPCAATAGCCCAAAGCAGCAGTTCACTGTGCCTGGTTATAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYPYDHSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACGACCATTCTGGTTRPISINYRTEIDKPSQHHHACTACCATCGGCCGATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 285)CCACCACCAC (SEQ ID NO: 286)2451H01MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPAAGYGYYRITYGETGGNTCAGCTGGGACGCTCCGGCTGCGGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPVSKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYDFPGSGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGACTTCCCCGGCTCCGGTTRPISINYRTEIDKPSQHHHHHHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 287)CCACCACCAC (SEQ ID NO: 288)2011B11MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWAPPSDAYGYYRITYGETGGNTCAGCTGGGCGCCGCCTTCTGATGCGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGTACAGCTACCATCAGCGGCCTTGVDYTITVYAVEYPYSHAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTATTCACATGCTGGTTRPISINYRTEIDKPSQH HHACTACCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 289)CCACCACCAC (SEQ ID NO: 290)2971A03MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDPPSDDYGYYRITYGETGGNTCAGCTGGGACCCGCCTTCGGATGATTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGAACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFPWPHAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTTTCCGTGGCCACATGCTGGTTRPISINYRTEIDKPSQHHHHHHACTATCATCGGCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 304)CCACCACCAC (SEQ ID NO: 305)2971A09MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPADDYGYYRITYGETGGNTCAGCTGGGACGCGCCTGCGGATGATTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGAACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFPWPHAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTTTCCGTGGCCACATGCTGGTTRPISINYRTEIDKPSQHHHHHHACTATCATCGGCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 306)CCACCACCAC (SEQ ID NO: 307)2971E02MGVSDVPRDLEVVAATPTSLLIATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGASWDAPSDDYGYYRITYGETGGNTCAGCTGGGACGCGCCTTCGGATGATTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGSPVQEFTVPIGKGTATISGLKPCAATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGAACAGCTACCATCAGCGGCCTTGVDYTITVYAVEFPWPHAGYYHAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTTTCCGTGGCCACATGCTGGTTRPISINYRTEIDKPSQHHHHHHACTATCATCGGCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATCCCAGCACCATCA(SEQ ID NO: 308)CCACCACCAC (SEQ ID NO: 309) The SEQ ID NOS of the family of the pegylated anti-PCSK9 Adnectins of the invention are presented in Table 5. TABLE 5Anti-PCSK9 Adnectin Family Cysteine Mutants to Enable PegylationATI#/Clone#Sequence[Description]AANTATI001170MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[2013E01-non HisLISWVPPSDDYGYYRITYGECAGCTGGGTCCCGCCTTCAGATGATTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCAtagged Cys mut]TGGNSPVQEFTVPIGKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGAACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEFPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTTTCCGTGGCCACATGCTGGTTACTAWPHAGYYHRPISINYRTEIDTCATCGGCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATGCCAGTG (SEQ IDKPCQ (SEQ ID NO: 78)NO: 79)ATI001172MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[2013E01-non HisLISWVPPSDDYGYYRITYGECAGCTGGGTCCCGCCTTCAGATGATTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCAtagged Cys mut]TGGNSPVQEFTVPIGKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGAACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEFPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTTTCCGTGGCCACATGCTGGTTACTAWPHAGYYHRPISINYRTEGSTCATCGGCCAATTTCCATTAATTACCGAACAGAAGGTAGCGGTTGCTG (SEQ ID NO: 81)GC (SEQ ID NO: 80)ATI001174*MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[2013E01-non HisLISWVPPSDDYGYYRITYGECAGCTGGGTCCCGCCTTCAGATGATTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCAtagged Cys mut]TGGNSPVQEFTVPIGKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGAACAGCTACCATCAGCGGCCTTAAAalso referred toISGLKPGVDYTITVYAVEFPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTTTCCGTGGCCACATGCTGGTTACTAas ATI-1174WPHAGYYHRPISINYRTEIETCATCGGCCAATTTCCATTAATTACCGCACAGAAATTGAGAAACCATGCCAGTG (SEQ IDKPCQ (SEQ ID NO: 82)NO: 83)ATI001114*MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[2013E01cys mut]LISWVPPSDDYGYYRITYGECAGCTGGGTCCCGCCTTCAGATGATTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCAalso referred toTGGNSPVQEFTVPIGKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGTAAAGGAACAGCTACCATCAGCGGCCTTAAAas ATI-1114ISGLKPGVDYTITVYAVEFPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAGTTTCCGTGGCCACATGCTGGTTACTAWPHAGYYHRPISINYRTGSGTCATCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACCHHHHHH (SEQ ID(SEQ ID NO: 85)NO: 84)ATI000959*MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[1459D05 cys mut]LISWPPPSHGYGYYRITYGECAGCTGGCCGCCGCCGTCTCATGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPPGKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCGCCTGGTAAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTACAAACATTCTGGTTACTAYKHSGYYHRPISINYRTEIDCCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATGCCAGCACCATCACCACCKPCQHHHHHH (SEQ IDACCAC (SEQ ID NO: 87)NO: 86)ATI001063*MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[1784F03 Cys mut]LISWRPPIHAYGYYRITYGECAGCTGGAGGCCGCCGATTCATGCTTACGGGTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPIVEGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGTTGAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYTCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATATACATTTAAACATTCCGGTTACTAFKHSGYYHRPISINYRTEIDCCATCGTCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATGCCAGCACCATCACCACCKPCQHHHHHH (SEQ IDACCAC (SEQ ID NO: 89)NO: 88)ATI001119*MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[2012A04 Cys mut]LISWRPPSNGHGYYRITYGECAGCTGGCGGCCCCCCTCTAATGGTCACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPVNEGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTAATGAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEFPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCCTTCAAGTGGTCGGGCTACTAFKWSGYYHRPISINYRTGSGCCATCGACCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACCHHHHHH (SEQ ID(SEQ ID NO: 91)NO: 90)ATI001117*MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[2011H05 Cys mut]LISWVPSSHAYGYYRITYGECAGCTGGGTTCCGTCGTCTCATGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPVGVGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTGTGGGGGTAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGCTTTCGAAGGGGCTGGTTACTAFEGAGYYHRPISINYRTGSGCCATCGTCCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACCHHHHHH (SEQ ID(SEQ ID NO: 93)NO: 92)ATI001194*MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[2011H05(V23D)LISWDPSSHAYGYYRITYGECAGCTGGGACCCGTCGTCTCATGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCACys mut]TGGNSPVQEFTVPVGVGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTGTGGGGGTAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGCTTTCGAAGGGGCTGGTTACTAFEGAGYYHRPISINYRTEGSCCATCGTCCAATTTCCATTAATTACCGCACAGAAGGTAGCGGTTGCCACCATCACCACCATCACGCHHHHHH (SEQ ID(SEQ ID NO: 95)NO: 94)2011H05 (V23E)-MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGATCys mutLISWEPSSHAYGYYRITYGECAGCTGGGAACCGTCGTCTCATGCCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPVGVGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTGTGGGGGTAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACGCTTTCGAAGGGGCTGGTTACTAFEGAGYYHRPISINYRTEGSCCATCGTCCAATTTCCATTAATTACCGCACAGAAGGTAGCGGTTGCCACCATCACCACCATCACGCHHHHHH (SEQ ID(SEQ ID NO: 97)NO: 96)ATI001112MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[1923B02 Cys mut]LISWTPPPKGYGYYRITYGECAGCTGGACGCCTCCCCCTAAAGGGTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPVGEGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTGGTGAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYTCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACACGTACAACGGTGCCGGTTACTAYNGAGYYHRPISINYRTGSGCCACCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACCHHHHHH (SEQ ID(SEQ ID NO: 99)NO: 98)ATI001110MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[1922G04 Cys mut]LISWRPPSHAYGYYRITYGECAGCTGGCGGCCGCCATCTCATGCTTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPIGKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGGAAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTGGAAAGGTTCTGGTTACTAWKGSGYYHRPISINYRTGSGCCATCGGCCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACCHHHHHH (SEQ ID(SEQ ID NO: 101)NO: 100)ATI001128MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[1922G04 Cys mut]LISWRPPSHAYGYYRITYGECAGCTGGCGGCCGCCATCTCATGCTTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPIGKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGGAAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTGGAAAGGTTCTGGTTACTAWKGSGYYHRPISINYRTEIDCCATCGGCCAATTTCCATTAATTACCGCACAGAAATTGACAAACCATGCCAGCACCACCACCACCKPCQH HHH (SEQ IDACCAC (SEQ ID NO: 103)NO: 102)ATI001184 *MGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGAT[1922G04(R23E)LISWEPPSHAYGYYRITYGECAGCTGGGAACCGCCATCTCATGCTTATGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCACys mut]TGGNSPVQEFTVPIGKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTATTGGGAAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEYPCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATACCCGTGGAAAGGTTCTGGTTACTAWKGSGYYHRPISINYRTEGSCCATCGGCCAATTTCCATTAATTACCGCACAGAAGGTAGCGGTTGCCACCATCACCACCATCACGCHHHHHH (SEQ ID(SEQ ID NO: 105)NO: 104)2381D04-CysMGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGATLISWEPFSRLPGGGEYYRITCAGCTGGGAGCCGTTCAGCCGGTTGCCCGGGGGCGGCGAGTATTACCGGATCACTTACGGCGAAAYGETGGNSPLQQFTVPGSKGCAGGAGGCAATAGCCCTCTGCAGCAGTTCACTGTGCCTGGTTCTAAAGGTACAGCTACCATCAGCTATISGLKPGVDYTITVYAVGGCCTTAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCCCGTACGACCATTCEFPYDHSGYYHRPISINYRTTGGTTACTACCATCGGCCCATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCGSGCHHHHHH (SEQ IDATCAC (SEQ ID NO: 292)NO:291)2382D09-CysMGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGATLISWDAPAEGYGYYRITYGECAGCTGGGACGCTCCGGCTGAAGGGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPVSKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEFDCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCGCCGGTTACTAFPGAGYYHRPISINYRTGSGCCATCGTCCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACCHHHHHH (SEQ ID(SEQ ID NO: 294)NO: 293)2451B06-CysMGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGATLISWDAPADGGYGYYRITYGCAGCTGGGACGCTCCGGCTGATGGTGGTTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGETGGNSPVQEFTVPVSKGTAGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTTISGLKPGVDYTITVYAVEFAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCGGGCGCCGGTTADFPGAGYYHRPISINYRTGSCTACCATCGTCCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACGCHHHHHH (SEQ ID(SEQ ID NO: 296)NO: 295)2382E05-CysMGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGATLISWDAPAEGGYGYYRITYGCAGCTGGGACGCTCCGGCTGAGGGCGGCTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGETGGNSPVQEFTVPVSKGTAGCAATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTTISGLKPGVDYTITVYAVEFAAACCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTTCCCCGGCGCCGGTTADFPGAGYYHRPISINYRTGSCTACCATCGTCCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACGCHHHHHH (SEQ ID(SEQ ID NO: 298)NO: 297)2382B09-CysMGVSDVPRDLEVVAATPTSLATGGGAGTTTCTGATGTGCCGCGCGACCTGGAAGTGGTTGCTGCCACCCCCACCAGCCTGCTGATLISWDAPADAYGYYRITYGECAGCTGGGACGCTCCGGCTGACGCGTACGGTTATTACCGCATCACTTACGGCGAAACAGGAGGCATGGNSPVQEFTVPVSKGTATATAGCCCTGTCCAGGAGTTCACTGTGCCTGTTTCTAAAGGTACAGCTACCATCAGCGGCCTTAAAISGLKPGVDYTITVYAVEFDCCTGGCGTTGATTATACCATCACTGTGTATGCTGTCGAATTCGACTACCCCGGCTCCGGTTACTAYPGSGYYHRPISINYRTGSGCCATCGTCCAATTTCCATTAATTACCGCACAGGTAGCGGTTGCCACCATCACCACCATCACCHHHHHH (SEQ ID(SEQ ID NO: 300)NO: 299)*Note:Some proteins listed have not yet been pegylated but are enabled to be pegylated via the cysteine mutation. Proteins that have been pegylated are indicated by asterisk. Nucleic Acid-Protein Fusion Technology In one aspect, the application provides an Adnectin comprising fibronectin type III domains that binds PCSK9. One way to rapidly make and test Fn3 domains with specific binding properties is the nucleic acid-protein fusion technology of Adnexus, a Bristol-Myers Squibb R&D Company. This disclosure utilizes the in vitro expression and tagging technology, termed PROfusion which exploits nucleic acid-protein fusions (RNA- and DNA-protein fusions) to identify novel polypeptides and amino acid motifs that are important for binding to proteins. Nucleic acid-protein fusion technology is a technology that covalently couples a protein to its encoding genetic information. For a detailed description of the RNA-protein fusion technology and fibronectin-based scaffold protein library screening methods see Szostak et al., U.S. Pat. Nos. 6,258,558, 6,261,804, 6,214,553, 6,281,344, 6,207,446, 6,518,018 and 6,818,418; and Roberts et al.,Proc. Natl. Acad. Sci.,94:12297-12302 (1997). Vectors and Polynucleotide Embodiments Nucleic acids encoding any of the various proteins or polypeptides disclosed herein may be synthesized chemically. Codon usage may be selected so as to improve expression in a cell. Such codon usage will depend on the cell type selected. Specialized codon usage patterns have been developed forE. coliand other bacteria, as well as mammalian cells, plant cells, yeast cells and insect cells. See for example: Mayfield et al.,Proc. Natl. Acad. Sci. USA,100(2):438-442 (Jan. 21, 2003); Sinclair et al.,Protein Expr. Puri.26 (496-105 (October 2002); Connell, N. D.,Curr. Opin. Biotechnol.,12(5):446-449 (October 2001); Makrides et al.,Microbiol. Rev.,60(3):512-538 (September 1996); and Sharp et al.,Yeast,7(7):657-678 (October 1991). General techniques for nucleic acid manipulation are described for example in Sambrook et al.,Molecular Cloning: A Laboratory Manual,2nd Edition, Vols. 1-3, Cold Spring Harbor Laboratory Press (1989), or Ausubel, F. et al.,Current Protocols in Molecular Biology, Green Publishing and Wiley-Interscience, New York (1987) and periodic updates. Generally, the DNA encoding the polypeptide is operably linked to suitable transcriptional or translational regulatory elements derived from mammalian, viral, or insect genes. Such regulatory elements include a transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites, and sequences that control the termination of transcription and translation. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants is additionally incorporated. The proteins described herein may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide. The heterologous signal sequence selected preferably is one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell. An exemplary N-terminal leader sequence for production of polypeptides in a mammalian system is METDTLLLWVLLLWVPGSTG (SEQ ID NO: 326), which is removed by the host cell following expression. For prokaryotic host cells that do not recognize and process a native signal sequence, the signal sequence is substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, lpp, or heat-stable enterotoxin II leaders. For yeast secretion the native signal sequence may be substituted by, e.g., the yeast invertase leader, a factor leader (includingSaccharomycesandKluyveromycesalpha-factor leaders), or acid phosphatase leader, theC. albicansglucoamylase leader, or the signal described in U.S. Pat. No. 5,631,144. In mammalian cell expression, mammalian signal sequences as well as viral secretory leaders, for example, the herpes simplex gD signal, are available. The DNA for such precursor regions may be ligated in reading frame to DNA encoding the protein. Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Generally, in cloning vectors this sequence is one that enables the vector to replicate independently of the host chromosomal DNA, and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells. Generally, the origin of replication component is not needed for mammalian expression vectors (the SV40 origin may typically be used only because it contains the early promoter). Expression and cloning vectors may contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli. Expression and cloning vectors usually contain a promoter that is recognized by the host organism and is operably linked to the nucleic acid encoding the protein of the invention, e.g., a fibronectin-based scaffold protein. Promoters suitable for use with prokaryotic hosts include the phoA promoter, beta-lactamase and lactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, and hybrid promoters such as the tan promoter. However, other known bacterial promoters are suitable. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the protein of the invention. Promoter sequences are known for eukaryotes. Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region where N may be any nucleotide. At the 3′ end of most eukaryotic genes is an AATAAA sequence that may be the signal for addition of the poly A tail to the 3′ end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vectors. Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase or other glycolytic enzymes, such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. Transcription from vectors in mammalian host cells can be controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, from heat-shock promoters, provided such promoters are compatible with the host cell systems. Transcription of a DNA encoding proteins of the invention by higher eukaryotes is often increased by inserting an enhancer sequence into the vector. Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, α-fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. See also Yaniv,Nature,297:17-18 (1982) on enhancing elements for activation of eukaryotic promoters. The enhancer may be spliced into the vector at a position 5′ or 3′ to the peptide-encoding sequence, but is preferably located at a site 5′ from the promoter. Expression vectors used in eukaryotic host cells (e.g., yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5′ and, occasionally 3′, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of mRNA encoding the protein of the invention. One useful transcription termination component is the bovine growth hormone polyadenylation region. See WO 94/11026 and the expression vector disclosed therein. The recombinant DNA can also include any type of protein tag sequence that may be useful for purifying the protein. Examples of protein tags include but are not limited to a histidine tag, a FLAG tag, a myc tag, an HA tag, or a GST tag. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts can be found inCloning Vectors: A Laboratory Manual, (Elsevier, New York (1985)). The expression construct is introduced into the host cell using a method appropriate to the host cell, as will be apparent to one of skill in the art. A variety of methods for introducing nucleic acids into host cells are known in the art, including, but not limited to, electroporation; transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (where the vector is an infectious agent). Suitable host cells include prokaryotes, yeast, mammalian cells, or bacterial cells. Suitable bacteria include gram negative or gram positive organisms, for example,E. coliorBacillusspp. Yeast, preferably from theSaccharomycesspecies, such asS. cerevisiae, may also be used for production of polypeptides. Various mammalian or insect cell culture systems can also be employed to express recombinant proteins. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow et al. (Bio/Technology,6:47 (1988)). Examples of suitable mammalian host cell lines include endothelial cells, COS-7 monkey kidney cells, CV-1, L cells, C127, 3T3, Chinese hamster ovary (CHO), human embryonic kidney cells, HeLa, 293, 293T, and BHK cell lines. Purified polypeptides are prepared by culturing suitable host/vector systems to express the recombinant proteins. For many applications, the small size of many of the polypeptides disclosed herein would make expression inE. colias the preferred method for expression. The protein is then purified from culture media or cell extracts. Protein Production Host cells are transformed with the herein-described expression or cloning vectors for protein production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences. In the examples shown here, the host cells used for high-throughput protein production (HTPP) and mid-scale production was the HMS174-bacterial strain. The host cells used to produce the proteins of this invention may be cultured in a variety of media. Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium ((MEM), (Sigma)), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma)) are suitable for culturing the host cells. In addition, many of the media described in Ham et al.,Meth. Enzymol.,58:44 (1979), Barites et al.,Anal. Biochem.,102:255 (1980), U.S. Pat. Nos. 4,767,704, 4,657,866, 4,927,762, 4,560,655, 5,122,469, 6,048,728, 5,672,502, or U.S. Pat. No. RE 30,985 may be used as culture media for the host cells. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as Gentamycin drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan. Proteins disclosed herein can also be produced using cell-translation systems. For such purposes the nucleic acids encoding the polypeptide must be modified to allow in vitro transcription to produce mRNA and to allow cell-free translation of the mRNA in the particular cell-free system being utilized (eukaryotic such as a mammalian or yeast cell-free translation system or prokaryotic such as a bacterial cell-free translation system. Proteins of the invention can also be produced by chemical synthesis (e.g., by the methods described inSolid Phase Peptide Synthesis,2nd Edition, The Pierce Chemical Co., Rockford, Ill. (1984)). Modifications to the protein can also be produced by chemical synthesis. The proteins of the present invention can be purified by isolation/purification methods for proteins generally known in the field of protein chemistry. Non-limiting examples include extraction, recrystallization, salting out (e.g., with ammonium sulfate or sodium sulfate), centrifugation, dialysis, ultrafiltration, adsorption chromatography, ion exchange chromatography, hydrophobic chromatography, normal phase chromatography, reversed-phase chromatography, get filtration, gel permeation chromatography, affinity chromatography, electrophoresis, countercurrant distribution or any combinations of these. After purification, polypeptides may be exchanged into different buffers and/or concentrated by any of a variety of methods known to the art, including, but not limited to, filtration and dialysis. The purified polypeptide is preferably at least 85% pure, or preferably at least 95% pure, and most preferably at least 98% pure. Regardless of the exact numerical value of the purity, the polypeptide is sufficiently pure for use as a pharmaceutical product. A platform manufacturing process was used to prepare anti-PCSK9 Adnectin. The anti-PCSK9 Adnectin is produced inEscherichia coli(E. coli).E. coliBLR (DE3) cells were transformed with expression vector (pET9d/ATI001173) which produces the protein in a soluble form intracellularly. The recombinant strain is grown in stirred tank fermentors. At the end of fermentation the cells are collected, lysed, and clarified in preparation for purification. ATI001173 is a non-his tagged version of ATI001114. The purified anti-PCSK9 Adnectin is conjugated to a 40 kDa branched methoxyPEG using a maleimide linker. The conjugated material is subsequently repurified to remove free PEG, free anti-PCSK9 Adnectin and product related impurities. Quality control testing is performed on the bulk drug substance. Therapeutic In Vivo Uses The application describes anti-PCSK9 Adnectin useful in the treatment of atherosclerosis, hypercholesterolemia and other cholesterol related diseases. The application also describes methods for administering anti-PCSK9 Adnectin to a subject. The subject can be a human. The anti-PCSK9 Adnectin can be pharmaceutically acceptable to a mammal, in particular a human. A “pharmaceutically acceptable” polypeptide refers to a polypeptide that is administered to an animal without significant adverse medical consequences, such as essentially endotoxin free, or very low endotoxin levels. Formulation and Administration The application further provides pharmaceutically acceptable compositions comprising the anti-PCSK9 Adnectin or fusion proteins thereof described herein, wherein the composition is essentially endotoxin free. Therapeutic formulations comprising anti-PCSK9 Adnectin or fusions thereof are prepared for storage by mixing the described polypeptide having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Osol, A.,Remington's Pharmaceutical Sciences,16th Edition (1980)), in the form of aqueous solutions, lyophilized or other dried formulations. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyidimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrans; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as Tween, PLURONIC® or polyethylene glycol (PEG). The formulations herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. Such molecules are suitably present in combination in amounts that are effective for the purpose intended. The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes. The skilled artisan will understand that the dosage of each therapeutic agent will be dependent on the identity of the agent. For therapeutic applications, the anti-PCSK9 Adnectin or a fusion protein comprising an anti-PCSK9 Adnectin is administered to a subject, in a pharmaceutically acceptable dosage form. They can be administered intravenously as a bolus or by continuous infusion over a period of time, or by subcutaneous routes. Suitable pharmaceutically acceptable carriers, diluents, and excipients are well known and can be determined by those of skill in the art as the clinical situation warrants. Examples of suitable carriers, diluents and/or excipients include: (1) Dulbecco's phosphate buffered saline, (2) 0.9% saline (0.9% w/v NaCl), and (3) 5% (w/v) dextrose. The method described herein can be practiced in vitro, in vivo, or ex vivo. Administration of anti-PCSK9 Adnectin or a fusion thereof, and one or more additional therapeutic agents, whether co-administered or administered sequentially, may occur as described above for therapeutic applications. Suitable pharmaceutically acceptable carriers, diluents, and excipients for co-administration will be understood by the skilled artisan to depend on the identity of the particular therapeutic agent being administered. When present in an aqueous dosage form, rather than being lyophilized, the protein typically will be formulated at a concentration of about 0.1 mg/ml to 100 mg/ml, although wide variation outside of these ranges is permitted. For the treatment of disease, the appropriate dosage of anti-PCSK9 Adnectin or a fusion thereof will depend on the type of disease to be treated, the severity and course of the disease, whether the protein is administered for preventive or therapeutic purposes, the course of previous therapy, the patient's clinical history and response to the protein, and the discretion of the attending physician. The protein is suitably administered to the patient at one time or over a series of treatments. Fusions of Serum Albumin Binding Adnectin (SABA) In certain aspects, the application provides fusion proteins comprising anti-PCSK9 Adnectin fused to a10Fn3 domain that binds to human serum albumin (a Serum Albumin Binding Adnectin (10Fn3 domain) or SABA). Such fusion proteins have extended serum half lives in the presence of albumin relative to the anti-PCSK9 Adnectin alone (e.g., not conjugated to a PK moiety). 10Fn3 domains are cleared rapidly from circulation via renal filtration and degradation due to their small size of ˜10 kDa (t1/2=15-45 minutes in mice; 1-3 hours in monkeys). Fusion of a10Fn3 domain, such as an anti-PCSK9 Adnectin, to a second polypeptide comprising a10Fn3 domain that binds specifically to serum albumin, e.g., human serum albumin (HSA), may be used to prolong the t1/2of the anti-PCSK9 Adnectin. In certain embodiments, the serum half-life of the anti-PCSK9 Adnectin fused to the SABA is increased relative to the serum half-life of the anti-PCSK9 Adnectin when not conjugated to the SABA. In certain embodiments, the serum half-life of the SABA fusion is at least 20, 40, 60, 80, 100, 120, 150, 180, 200, 400, 600, 800, 1000, 1200, 1500, 1800, 1900, 2000, 2500, or 3000% longer relative to the serum half-life of the anti-PCSK9 Adnectin when not fused to the SABA. In other embodiments, the serum half-life of the SABA fusion is at least 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5 fold, 4-fold, 4.5-fold, 5-fold, 6-fold, 7-fold, 8-fold, 10-fold, 12-fold, 13-fold, 15-fold, 17-fold, 20-fold, 22-fold, 25-fold, 27-fold, 30-fold, 35-fold, 40-fold, or 50-fold greater than the serum half-life of the anti-PCSK9 Adnectin when not fused to the SABA. In some embodiments, the serum half-life of the SABA fusion is at least 10 hours, 15 hours, 20 hours, 25 hours, 30 hours, 35 hours, 40 hours, 50 hours, 60 hours, 70 hours, 80 hours, 90 hours, 100 hours, 110 hours, 120 hours, 130 hours, 135 hours, 140 hours, 150 hours, 160 hours, or 200 hours. In certain embodiments, the serum albumin binding portion of the SABA fusion protein binds to HSA with a KDof less than 3 uM, 2.5 uM, 2 uM, 1.5 uM, 1 uM, 500 nM, 100 nM, 50 nM, 10 nM, 1 nM, 500 pM, 100 pM, 50 pM or 10 pM. In certain embodiments, the serum albumin binding portion of the SABA fusion proteins bind to HSA with a KDof less than 3 uM, 2.5 uM, 2 uM, 1.5 uM, 1 uM, 500 nM, 100 nM, 50 nM, 10 nM, 1 nM, 500 pM, 100 pM, 50 pM or 10 pM at a pH range of 5.5 to 7.4 at 25° C. or 37° C. In some embodiments, the serum albumin binding portion of the SABA fusion proteins bind more tightly to HSA at a pH less than 7.4 as compared to binding at pH 7.4. Accordingly, the SABA fusion molecules described herein are useful for increasing the half-life of anti-PCSK9 Adnectin by creating a fusion between anti-PCSK9 Adnectin and the SABA. Such fusion molecules may be used to treat conditions which respond to the biological activity of PCSK9. The use of the SABA fusion molecules in diseases caused by the dysregulation of PCSK9 is contemplated. The fusion may be formed by attaching anti-PCSK9 Adnectin to either end of the SABA molecule, i.e., SABA-anti-PCSK9 Adnectin or anti-PCSK9 Adnectin-SABA arrangements. HSA has a serum concentration of 600 μM and a t1/2of 19 days in humans. The extended t1/2of HSA has been attributed, in part, to its recycling via the neonatal Fc receptor (FcRn). HSA binds FcRn in a pH-dependent manner after endosomal uptake into endothelial cells; this interaction recycles HSA back into the bloodstream, thereby shunting it away from lysosomal degradation. FcRn is widely expressed and the recycling pathway is thought to be constitutive. In the majority of cell types, most FcRn resides in the intracellular sorting endosome. HSA is readily internalized by a nonspecific mechanism of fluid-phase pinocytosis and rescued from degradation in the lysosome by FcRn. At the acidic pH found in the endosome, HSA's affinity for FcRn increases (5 μM at pH 6.0). Once bound to FcRn, HSA is shunted away from the lysosomal degradation pathway, transcytosed to and released at the cell surface. In certain embodiments, the serum albumin binding portion of the SABA fusion proteins described herein may also bind serum albumin from one or more of monkey, rat, or mouse. In certain embodiments, the HSA binding portion of the SABA fusion proteins described herein bind to rhesus serum albumin (RhSA) or cynomolgus monkey serum albumin (CySA) with a KDof less than 3 uM, 2.5 uM, 2 uM, 1.5 uM, 1 uM, 500 nM, 100 nM, 50 nM, 10 nM, 1 nM, 500 pM or 100 pM. In certain embodiments, the serum albumin binding portion of the SABA fusion proteins described herein bind to domain I and/or domain II of HSA. In one embodiment, the HSA binding portion of the SABA fusion proteins described herein do not bind to domain III of HSA. In certain embodiments, the serum albumin binding portion of the SABA fusion proteins comprises a sequence having at least 40%, 50%, 60%, 70%, 75%, 80% or 85% identity to the wild-type10Fn3 domain (SEQ ID NO: 1). In one embodiment, at least one of the BC, DE, or FG loops is modified relative to the wild-type10Fn3 domain. In another embodiment, at least two of the BC, DE, or FG loops are modified relative to the wild-type10Fn3 domain. In another embodiment, all three of the BC, DE, and FG loops are modified relative to the wild-type10Fn3 domain. In other embodiments, a SABA comprises a sequence having at least 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, or 95% identity to any one of the 26 core SABA sequences shown in Table 6 (i.e., SEQ ID NO: 334, 338, 342, 346, and 348-370) or any one of the extended SABA sequences shown in Table 6 (i.e., SEQ ID NO: 420-447, minus the 6×HIS tag). In certain embodiments, the serum binding Adnectins based on the10Fn3 scaffold can be defined generally by the following sequence: (SEQ ID NO: 328)EVVAAT(X)aSLLI(X)xYYRITYGE(X)bQEFTV(X)yATI(X)cDYTITVYAV(X)zISINYRT In certain embodiments, the serum binding Adnectins based on the10Fn3 scaffold can be defined generally by the sequence: (SEQ ID NO: 329)EVVAATPTSLLI(X)xYYRITYGETGGNSPVQEFTV(X)yATISGLKPGVDYTITVYAV(X)zISINYRT As described herein for anti-PCSK9 Adnectins, SEQ ID NOs: 328 and 329 can be defined and applied to SABA molecules in the same way. In exemplary embodiments, the BC, DE, and FG loops as represented by (X)x, (X)y, and (X)z, respectively, are replaced with polypeptides comprising the BC, DE and FG loop sequences from any of the HSA binders shown in Table 6 below (i.e., SEQ ID NOs: 330, 334, 338, 342, 346, and 348-370 in Table 6). In certain embodiments, the BC, DE, or FG loop sequences shown in Table 6 may contain one or more additional residues flanking the N- and/or C-termini. In particular, the BC loop may contain an SW at the N-terminus of the BC loop sequences shown in Table 6 when replacing (X)xin SEQ ID NO: 328. Similarly, the DE loop may contain a P preceding loop DE and the residue T following loop DE when replacing (X)yin SEQ ID NO: 328. The FG loop may contain a P following the FG loop when replacing (X)zin SEQ ID NO: 328. For example, SEQ ID NO: 330 indicates that the BC, DE, and FG loops comprise HSYYEQNS (SEQ ID NO: 638), YSQT (SEQ ID NO: 639), and YGSKYYY (SEQ ID NO: 640), respectively. However, when replacing (X)x, (X)y, and (X)zin SEQ ID NO: 328, i.e., the BC, DE and FG loops, the (X)xsequence may be SWHSYYEQNS (SEQ ID NO: 641), the (X)ysequence may be PYSQTT (SEQ ID NO: 642), and the (X)zsequence may be YGSKYYYP (SEQ ID NO: 643). In certain embodiments, a SABA for use in a fusion as described herein may comprise the sequence as set forth in SEQ ID NO: 328 or 329, wherein the BC, DE, and FG loops as represented by (X)x, (X)y, and (X)z, respectively, are replaced with a respective set of specified BC, DE, and FG loops from any of the 26 core SABA sequences (i.e., SEQ ID NOs: 330, 334, 338, 342, 346, and 348-370 in Table 6), or sequences at least 75%, 80%, 85%, 90%, 95%, 97%, 98%, or 99% identical to the BC, DE and FG loop sequences of the 26 core SABA sequences. In exemplary embodiments, a SABA as described herein is defined by SEQ ID NO: 329 and has a set of BC, DE and FG loop sequences from any of the 26 core SABA sequences (i.e., SEQ ID NOs: 330, 334, 338, 342, 346, and 348-370 in Table 6), optionally with the N- and/or C-terminal additions to the loop sequences as described above. For example, SABA1 has the core sequence set forth in SEQ ID NO: 330 and comprises BC, DE, and FG loops as set forth in SEQ ID NO: 331-333, respectively. Therefore, a SABA based on the SABA1 core may comprise SEQ ID NO: 328 or 329, wherein (X)xcomprises SEQ ID NO: 331, (X)ycomprises SEQ ID NO: 332, and (X)zcomprises SEQ ID NO: 333. In some embodiments, the sequences that replace (X)x, (X)y, and (X)zcomprise additional residue(s) on either or both ends of the loops as described above. Similar constructs are contemplated utilizing the set of BC, DE and FG loops from the other SABA core sequences. The scaffold regions of such SABA may comprise anywhere from 0 to 20, from 0 to 15, from 0 to 10, from 0 to 8, from 0 to 6, from 0 to 5, from 0 to 4, from 0 to 3, from 0 to 2, or from 0 to 1 substitutions, conservative substitutions, deletions or additions relative to the scaffold amino acids residues of SEQ ID NO: 1. Such scaffold modifications may be made, so long as the SABA is capable of binding serum albumin, e.g., HSA, with a desired KD. In certain embodiments, a SABA (e.g., a SABA core sequence or a sequence based thereon as described above) may be modified to comprise an N-terminal extension sequence and/or a C-terminal extension sequence. Exemplary extension sequences are shown in Table 6. For example, SEQ ID NO: 420 designated as SABA1.1 comprises the core SABA 1 sequence (SEQ ID NO: 330) with an N-terminal sequence MGVSDVPRDLE (SEQ ID NO: 371, designated as AdNT1), and a C-terminal sequence EIDKPSQ (SEQ ID NO: 380, designated as AdCT1). SABA1.1 further comprises a His6 tag at the C-terminus, however, it should be understood that the His6 tag is completely optional and may be placed anywhere within the N- or C-terminal extension sequences, or may be absent from the sequence all together. Further, any of the exemplary N- or C-terminal extension sequences provided in Table 6 (SEQ ID NO: 371-395), and any variants thereof, can be used to modify any given SABA core sequence provided in Table 6. In certain embodiments, the C-terminal extension sequences (also called “tails”), comprise E and D residues, and may be between 8 and 50, 10 and 30, 10 and 20, 5 and 10, and 2 and 4 amino acids in length. In some embodiments, tail sequences include ED-based linkers in which the sequence comprises tandem repeats of ED. In exemplary embodiments, the tail sequence comprises 2-10, 2-7, 2-5, 3-10, 3-7, 3-5, 3, 4 or 5 ED repeats. In certain embodiments, the ED-based tail sequences may also include additional amino acid residues, such as, for example: EI, EID, ES, EC, EGS, and EGC. Such sequences are based, in part, on known Adnectin tail sequences, such as EIDKPSQ (SEQ ID NO: 380), in which residues D and K have been removed. In exemplary embodiments, the ED-based tail comprises an E, I or EI residues before the ED repeats. In other embodiments, the tail sequences may be combined with other known linker sequences (e.g., SEQ ID NO: 396-419 in Table 6) as necessary when designing a SABA fusion molecule. Conjugation/Linkers SABA fusions may be covalently or non-covalently linked. In some embodiments, a serum albumin binding10Fn3 may be directly or indirectly linked to an anti-PCSK9 Adnectin via a polypeptide linker. Suitable linkers for joining Fn3 domains are those which allow the separate domains to fold independently of each other forming a three dimensional structure that permits high affinity binding to a target molecule. The disclosure provides a number of suitable linkers that meet these requirements, including glycine-serine based linkers, glycine-proline based linkers, as well as the linker having the amino acid sequence PSTSTST (SEQ ID NO: 416). The Examples described herein demonstrate that Fn3 domains joined via polypeptide linkers retain their target binding function. In some embodiments, the linker is a glycine-serine based linker. These linkers comprise glycine and serine residues and may be between 8 and 50, 10 and 30, and 10 and 20 amino acids in length. Examples include linkers having an amino acid sequence (GS)7(SEQ ID NO: 403), G(GS)6(SEQ ID NO: 398), and G(GS)7G (SEQ ID NO: 400). Other linkers contain glutamic acid, and include, for example, (GSE)5(SEQ ID NO: 405) and GGSE GGSE (SEQ ID NO: 409). Other exemplary glycine-serine linkers include (GS)4(SEQ ID NO: 402), (GGGGS)7(SEQ ID NO: 411), (GGGGS)5(SEQ ID NO: 412), and (GGGGS)3G (SEQ ID NO: 413). In some embodiments, the linker is a glycine-proline based linker. These linkers comprise glycine and proline residues and may be between 3 and 30, 10 and 30, and 3 and 20 amino acids in length. Examples include linkers having an amino acid sequence (GP)3G (SEQ ID NO: 414), (GP)5G (SEQ ID NO: 415), and GPG. In other embodiments, the linker may be a proline-alanine based linker having between 3 and 30, 10 and 30, and 3 and 20 amino acids in length. Examples of proline alanine based linkers include, for example, (PA)3(SEQ ID NO: 417), (PA)6(SEQ ID NO: 418) and (PA)9(SEQ ID NO: 419). It is contemplated, that the optimal linker length and amino acid composition may be determined by routine experimentation in view of the teachings provided herein. In some embodiments, an anti-PCSK9 Adnectin is linked to a SABA via a polypeptide linker having a protease site that is cleavable by a protease in the blood or target tissue. Such embodiments can be used to release an anti-PCSK9 Adnectin for better delivery or therapeutic properties or more efficient production. Additional linkers or spacers, may be introduced at the C-terminus of a Fn3 domain between the Fn3 domain and the polypeptide linker. Additional linkers or spacers may be introduced at the N-terminus of a Fn3 domain between the Fn3 domain and the polypeptide linker. In some embodiments, an anti-PCSK9 Adnectin may be directly or indirectly linked to a SABA via a polymeric linker. Polymeric linkers can be used to optimally vary the distance between each component of the fusion to create a protein fusion with one or more of the following characteristics: 1) reduced or increased steric hindrance of binding of one or more protein domains when binding to a protein of interest, 2) increased protein stability or solubility, 3) decreased protein aggregation, and 4) increased overall avidity or affinity of the protein. In some embodiments, an anti-PCSK9 Adnectin is linked to a SABA via a biocompatible polymer such as a polymeric sugar. The polymeric sugar can include an enzymatic cleavage site that is cleavable by an enzyme in the blood or target tissue. Such embodiments can be used to release an anti-PCSK9 Adnectin for better delivery or therapeutic properties or more efficient production. Summary of Sequences Many of the sequences referenced in “Fusions of Serum Albumin Binding Adnectin (SABA)” and “Conjugation/Linkers” sections above are summarized in Table 6 below. Unless otherwise specified, all N-terminal extensions are indicated with a single underline, all C-terminal tails/extensions are indicated with a double underline, and linker sequences are boxed. Loop regions BC, DE and FG are italicized for each core SABA sequence. As described further above, the modification sequences (e.g., N or C terminal extensions and linkers) can also be used to modify anti-PCSK9 Adnectin molecules. TABLE 6Summary of Exemplary SequencesSEQSequenceIDNameDescriptionSequenceExemplary Serum Albumin-Binding Adnectins (SABA)32710Fn3WTWT core human10Fn3EVVAATPTSLLISWDAPAVTVRYYRITYGETcoredomainGGNSPVQEFTVPGSKSTATISGLKPGVDYTITVYAVTGRGDSPASSKPISINYRT32810Fn3v6Generic10Fn3 having 6EVVAAT(X)aSLLI(X)xYYRITYGE(X)bQEvariable loopsFTV(X)yATI(X)cDYTITVYAV(X)zISINYRT32910Fn3v3Generic10Fn3 having 3EVVAATPTSLLI(X)xYYRITYGETGGNSPVvariable loopsQEFTV(X)yATISGLKPGVDYTITVYAV(X)zISINYRT330SABA1Core 1 AdnectinEVVAATPTSLLISWHSYYEQNSYYRITYGETGGNSPVQEFTVPYSQTTATISGLKPGVDYTITVYAVYGSKYYYPISINYRT331SABA1BCCore 1 BC LoopHSYYEQNS332SABA1DECore 1 DE LoopYSQT333SABA1FGCore 1 FG LoopYGSKYYY334SABA2Core 2 AdnectinEVVAATPTSLLISWPKYDKTGHYYRITYGETGGNSPVQEFTVPTRQTTATISGLKPGVDYTITVYAVSKDDYYPHEHRPISINYRT335SABA2BCCore 2 BC LoopPKYDKTGH336SABA2DECore 2 DE LoopTRQT337SABA2FGCore 2 FG LoopSKDDYYPHEHR338SABA3Core 3 AdnectinEVVAATPTSLLISWSNDGPGLSYYRITYGETGGNSPVQEFTVPSSQTTATISGLKPGVDYTITVYAVSYYTKKAYSAGPISINYRT339SABA3BCCore 3 BC LoopSNDGPGLS340SABA3DECore 3 DE LoopSSQT341SABA3FGCore 3 FG LoopSYYTKKAYSAG342SABA4Core 4 Adnectin;EMVAATPTSLLISWEDDSYYSRYYRITYGETcontains a scaffoldGGNSPVQEFTVPSDLYTATISGLKPGVDYTImutation (bolded);TVYAVTYDVTDLIMHEPISINYRTscaffold-perfect versionis SABA5343SABA4BCCore 4 BC LoopEDDSYYSR344SABA4DECore 4 DE LoopSDLY345SABA4FGCore 4 FG LoopYDVTDLIMHE346SABA5Core 5 Adnectin; seeEVVAATPTSLLISWEDDSYYSRYYRITYGETdescription for SABA4;GGNSPVQEFTVPSDLYTATISGLKPGVDYTIcorrected residue isTVYAVTYDVTDLIMHEPISINYRTbolded347SABA5BCCore 5 BC LoopEDDSYYSR348SABA5DECore 5 DE LoopSDLY349SABA5FGCore 5 FG LoopYDVTDLIMHE350SABA6Core 6 AdnectinEVVAATPTSLLISWYMDEYDVRYYRITYGETGGNSPVQEFTVPNYYNTATISGLKPGVDYTITVYAVTRIKANNYMYGPISINYRT351SABA7Core 7 AdnectinEVVAATPTSLLISWNHLEHVARYYRITYGETGGNSPVQEFTVPEYPTTATISGLKPGVDYTITVYAVTITMLKYPTQSPISINYRT352SABA8Core 8 AdnectinEVVAATPTSLLISWGHYRRSGHYYRITYGETGGNSPVQEFTVDPSSYTATISGLKPGVDYTITVYAVSKDDYYPHEHRPISINYRT353SABA9Core 9 AdnectinEVVAATPTSLLISWDASHYERRYYRITYGETGGNSPVQEFTVPRYHHTATISGLKPGVDYTITVYAVTQAQEHYQPPISINYRT354SABA10Core 10 AdnectinEVVAATPTSLLISWNSYYHSADYYRITYGETGGNSPVQEFTVPYPPTTATISGLKPGVDYTITVYAVYSAKSYYPISINYRT355SABA11Core 11 AdnectinEVVAATPTSLLISWSKYSKHGHYYRITYGETGGNSPVQEFTVPSGNATATISGLKPGVDYTITVYAVEDTNDYPHTHRPISINYRT356SABA12Core 12 AdnectinEVVAATPTSLLISWHGEPDQTRYYRITYGETGGNSPVQEFTVPPYRRTATISGLKPGVDYTITVYAVTSGYTGHYQPISINYRT357SABA13Core 13 AdnectinEVVAATPTSLLISWSKYSKHGHYYRITYGETGGNSPVQEFTVDPSSYTATISGLKPGVDYTITVYAVSKDDYYPHEHRPISINYRT358SABA14Core 14 AdnectinEVVAATPTSLLISWYEPYTPIHYYRITYGETGGNSPVQEFTVPGYYGTATISGLKPGVDYTITVYAVYGYYQYTPISINYRT359SABA15Core 15 AdnectinEVVAATPTSLLISWSKYSKHGHYYRITYGETGGNSPVQEFTVPSGNATATISGLKPGVDYTITVYAVSDDNKYYHQHRPISINYRT360SABA16Core 16 AdnectinEVVAATPTSLLISWGHYRRSGHYYRITYGETGGNSPVQEFTVDPSSYTATISGLKPGVDYTITVYAVSKDDYYPHEHRPISINYRT361SABA17Core 17 AdnectinEVVAATPTSLLISWSKYSKHGHYYRITYGETGGNSPVQEFTVPSGNATATISGLKPGVDYTITVYAVEDTNDYPHTHRPISINYRT362SABA18Core 18 AdnectinEVVAATPTSLLISWYEPGASVYYYRITYGETGGNSPVQEFTVPSYYHTATISGLKPGVDYTITVYAVYGYYEYEPISINYRT363SABA19Core 19 AdnectinEVVAATPTSLLISWQSYYAHSDYYRITYGETGGNSPVQEFTVPYPPQTATISGLKPGVDYTITVYAVYAGSSYYPISINYRT364SABA20Core 20 AdnectinEVVAATPTSLLISWGHYRRSGHYYRITYGETGGNSPVQEFTVDPSSYTATISGLKPGVDYTITVYAVSKDDYYPHEHRPISINYRT365SABA21Core 21 AdnectinEVVAATPTSLLISWPEPGTPVYYYRITYGETGGNSPVQEFTVPAYYGTATISGLKPGVDYTITVYAVYGYYDYSPISINYRT366SABA22Core 22 AdnectinEVVAATPTSLLISWYRYEKTQHYYRITYGETGGNSPVQEFTVPPESGTATISGLKPGVDYTITVYAVYAGYEYPHTHRPISINYRT367SABA23Core 23 AdnectinEVVAATPTSLLISWVKSEEYYRYYRITYGETGGNSPVQEFTVPYYVHTATISGLKPGVDYTITVYAVTEYYYAGAVVSVPISINYRT368SABA24Core 24 AdnectinEVVAATPTSLLISWYDPYTYGSYYRITYGETGGNSPVQEFTVGPYTTTATISGLKPGVDYTITVYAVSYYYSTQPISINYRT369SABA25Core 25 AdnectinEVVAATPTSLLISWSNDGPGLSYYRITYGETGGNSPVQEFTVPSSQTTATISGLKPGVDYTITVYAVSYYTKKAYSAGPISINYRT370SABA26Core 26 AdnectinEVVAATPTSLLISWPDPYYKPDYYRITYGETGGNSPVQEFTVPRDYTTATISGLKPGVDYTITVYAVYSYYGYYPISINYRTExemplary Adnectin N-Terminal Extension Sequences371AdNT1Exemplary leaderMGVSDVPRDL372AdNT2Exemplary leaderGVSDVPRDL373AdNT3Exemplary leaderVSDVPRDL374AdNT4Exemplary leaderSDVPRDL375AdNT5Exemplary leaderDVPRDL376AdNT6Exemplary leaderVPRDL377AdNT7Exemplary leaderPRDL378AdNT8Exemplary leaderRDL379AdNT9Exemplary leaderDLExemplary Adnectin C-Terminal Extension Sequences380AdCT1Exemplary tailEIDKPSQ381AdCT2Exemplary tailEIDKPS382AdCT3Exemplary tailEIDKPC383AdCT4Exemplary tailEIDKP384AdCT5Exemplary tailEIDK385AdCT6Exemplary tailEI386AdCT7Exemplary tailEIEKPSQ387AdCT8Exemplary tailEIDKPSQLE388AdCT9Exemplary tailEIEDEDEDEDED389AdCT10Exemplary tailEIEKPSQEDEDEDEDED390AdCT11Exemplary tailEGSGS391AdCT12Exemplary tailEIDKPCQ392AdCT13Exemplary tailEIEKPCQ393AdCT14Exemplary tailGSGC394AdCT15Exemplary tailEGSGC395AdCT16Exemplary tailEIDKPCQLE396L1G(GS)2GGSGS397L2G(GS)4GGSGSGSGS398L3G(GS)6GGSGSGSGSGSGS399L4G(GS)7GGSGSGSGSGSGSGS400L5G(GS)7GGGSGSGSGSGSGSGSG401L6GSGSGSGS402L7(GS)4GSGSGSGS403L7(GS)7GSGSGSGSGSGSGS404L9GS(A)9GSGSAAAAAAAAAGS405L10(GSE)5GSEGSEGSEGSEGSE406L11(PAS)5PASPASPASPASPAS407L12(GSP)5GSPGSPGSPGSPGSP408L13GS(TVAAPS)2GSTVAAPSTVAAPS409L14(GGSE)2GGSEGGSE410L15(ST)3GSTSTSTG411L16(GGGGS)7GGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS412L17(GGGGS)5GGGGSGGGGSGGGGSGGGGSGGGGSGGGGS413L18(GGGGS)3GGGGGSGGGGSGGGGSG414L19(GP)3GGPGPGPG415L20(GP)5GGPGPGPGPGPG416L21P(ST)3PSTSTST417L22(PA)3PAPAPA418L23(PA)6PAPAPAPAPAPA419L24(PA)9PAPAPAPAPAPAPAPAPAExemplary Extensions to Adnectin Core Sequences420SABA1.1Adnectin core 1MGVSDVPRDLEVVAATPTSLLISWHSYYEQNsequence having AdNT1SYYRITYGETGGNSPVQEFTVPYSQTTATISand AdCT1 terminalGLKPGVDYTITVYAVYGSKYYYPISINYRTEsequences with His6 tagIDKPSQHHHHHH421SABA1.2Adnectin core 1MGVSDVPRDLEVVAATPTSLLISWHSYYEQNsequence having AdNT1SYYRITYGETGGNSPVQEFTVPYSQTTATISand AdCT8 terminalGLKPGVDYTITVYAVYGSKYYYPISINYRTEsequencesIEDEDEDEDED422SABA1.3Adnectin core 1MGVSDVPRDLEVVAATPTSLLISWHSYYEQNand AdCT9 terminalSYYRITYGETGGNSPVQEFTVPYSQTTATISsequences with His6 tagGLKPGVDYTITVYAVYGSKYYYPISINYRTEsequence having AdNT1IEDEDEDEDEDHHHHHH423SABA2.1Adnectin core 2MGVSDVPRDLEVVAATPTSLLISWPKYDKTGsequence having AdNT1HYYRITYGETGGNSPVQEFTVPTROTTATISand AdCT1 terminalGLKPGVDYTITVYAVSKDDYYPHEHRPISINsequences with His6 tagYRTEIDKPSQHHHHHH424SABA3.1Adnectin core 3MGVSDVPRDLEVVAATPTSLLISWSNDGPGLand AdCT1 terminalSYYRITYGETGGNSPVQEFTVPSSQTTATISsequences with His6 tagGLKPGVDYTITVYAVSYYTKKAYSAGPISINsequence having AdNT1YRTEIDKPSQHHHHHH425SABA4.1Adnectin core 4MGVSDVPRDLEMVAATPTSLLISWEDDSYYSsequence having AdNT1RYYRITYGETGGNSPVQEFTVPSDLYTATISand AdCT1 terminalGLKPGVDYTITVYAVTYDVTDLIMHEPISINsequences with His6 tagYRTEIDKPSQHHHHHH426SABA5.1Adnectin core 5MGVSDVPRDLEVVAATPTSLLISWEDDSYYSand AdCT1 terminalRYYRITYGETGGNSPVQEFTVPSDLYTATISsequences with His6 tagGLKPGVDYTITVYAVTYDVTDLIMHEPISINsequence having AdNT1YRTEIDKPSQHHHHHH427SABA6.1Adnectin core 6MGVSDVPRDLEVVAATPTSLLISWYMDEYDVand AdCT1 terminalRYYRITYGETGGNSPVQEFTVPNYYNTATISsequences with His6 tagGLKPGVDYTITVYAVTRIKANNYMYGPISINsequence having AdNT1YRTEIDKPSQHHHHHH428SABA7.1Adnectin core 7MGVSDVPRDLEVVAATPTSLLISWNHLEHVAsequence having AdNT1RYYRITYGETGGNSPVQEFTVPEYPTTATISand AdCT1 terminalGLKPGVDYTITVYAVTITMLKYPTQSPISINsequences with His6 tagYRTEIDKPSQHHHHHH429SABA8.1Adnectin core 8MGVSDVPRDLEVVAATPTSLLISWGHYRRSGsequence having AdNT1HYYRITYGETGGNSPVQEFTVDPSSYTATISand AdCT1 terminalGLKPGVDYTITVYAVSKDDYYPHEHRPISINsequences with His6 tagYRTEIDKPSQHHHHHH430SABA9.1Adnectin core 9MGVSDVPRDLEVVAATPTSLLISWDASHYERand AdCT1 terminalRYYRITYGETGGNSPVQEFTVPRYHHTATISsequences with His6 tagGLKPGVDYTITVYAVTQAQEHYQPPISINYRsequence having AdNT1TEIDKPSQHHHHHH431SABA10.1Adnectin core 10MGVSDVPRDLEVVAATPTSLLISWNSYYHSAsequence having AdNT1DYYRITYGETGGNSPVQEFTVPYPPTTATISand AdCT1 terminalGLKPGVDYTITVYAVYSAKSYYPISINYRTEsequences with His6 tagIDKPSQHHHHHH432SABA11.1Adnectin core 11MGVSDVPRDLEVVAATPTSLLISWSKYSKHGand AdCT1 terminalHYYRITYGETGGNSPVQEFTVPSGNATATISsequences with His6 tagGLKPGVDYTITVYAVEDTNDYPHTHRPISINsequence having AdNT1YRTEIDKPSQHHHHHH433SABA12.1Adnectin core 12MGVSDVPRDLEVVAATPTSLLISWHGEPDQTsequence having AdNT1RYYRITYGETGGNSPVQEFTVPPYRRTATISand AdCT1 terminalGLKPGVDYTITVYAVTSGYTGHYQPISINYRsequences with His6 tagTEIDKPSQHHHHHH434SABA13.1Adnectin core 13MGVSDVPRDLEVVAATPTSLLISWSKYSKHGsequence having AdNT1HYYRITYGETGGNSPVQEFTVDPSSYTATISand AdCT1 terminalGLKPGVDYTITVYAVSKDDYYPHEHRPISINsequences with His6 tagYRTEIDKPSQHHHHHH435SABA14.1Adnectin core 14MGVSDVPRDLEVVAATPTSLLISWYEPYTPIand AdCT1 terminalHYYRITYGETGGNSPVQEFTVPGYYGTATISsequences with His6 tagGLKPGVDYTITVYAVYGYYQYTPISINYRTEsequence having AdNT1IDKPSQHHHHHH436SABA15.1Adnectin core 15MGVSDVPRDLEVVAATPTSLLISWSKYSKHGsequence having AdNT1HYYRITYGETGGNSPVQEFTVPSGNATATISand AdCT1 terminalGLKPGVDYTITVYAVSDDNKYYHQHRPISINsequences with His6 tagYRTEIDKPSQHHHHHH437SABA16.1Adnectin core 16MGVSDVPRDLEVVAATPTSLLISWGHYRRSGsequence having AdNT1HYYRITYGETGGNSPVQEFTVDPSSYTATISand AdCT1 terminalGLKPGVDYTITVYAVSKDDYYPHEHRPISINsequences with His6 tagYRTEIDKPSQHHHHHH438SABA17.1Adnectin core 17MGVSDVPRDLEVVAATPTSLLISWSKYSKHGsequence having AdNT1HYYRITYGETGGNSPVQEFTVPSGNATATISand AdCT1 terminalGLKPGVDYTITVYAVEDTNDYPHTHRPISINsequences with His6 tagYRTEIDKPSQHHHHHH439SABA18.1Adnectin core 18MGVSDVPRDLEVVAATPTSLLISWYEPGASVsequence having AdNT1YYYRITYGETGGNSPVQEFTVPSYYHTATISand AdCT1 terminalGLKPGVDYTITVYAVYGYYEYEPISINYRTEsequences with His6 tagIDKPSQHHHHHH440SABA19.1Adnectin core 19MGVSDVPRDLEVVAATPTSLLISWQSYYAHSand AdCT1 terminalDYYRITYGETGGNSPVQEFTVPYPPQTATISsequences with His6 tagGLKPGVDYTITVYAVYAGSSYYPISINYRTEsequence having AdNT1IDKPSQHHHHHH441SABA20.1Adnectin core 20MGVSDVPRDLEVVAATPTSLLISWGHYRRSGsequence having AdNT1HYYRITYGETGGNSPVQEFTVDPSSYTATISand AdCT1 terminalGLKPGVDYTITVYAVSKDDYYPHEHRPISINsequences with His6 tagYRTEIDKPSQHHHHHH442SABA21.1Adnectin core 21MGVSDVPRDLEVVAATPTSLLISWPEPGTPVand AdCT1 terminalYYYRITYGETGGNSPVQEFTVPAYYGTATISsequences with His6 tagGLKPGVDYTITVYAVYGYYDYSPISINYRTEsequence having AdNT1IDKPSQHHHHHH443SABA22.1Adnectin core 22MGVSDVPRDLEVVAATPTSLLISWYRYEKTQand AdCT1 terminalHYYRITYGETGGNSPVQEFTVPPESGTATISsequences with His6 tagGLKPGVDYTITVYAVYAGYEYPHTHRPISINsequence having AdNT1YRTEIDKPSQHHHHHH444SABA23.1Adnectin core 23MGVSDVPRDLEVVAATPTSLLISWVKSEEYYsequence having AdNT1RYYRITYGETGGNSPVQEFTVPYYVHTATISand AdCT1 terminalGLKPGVDYTITVYAVTEYYYAGAVVSVPISIsequences with His6 tagNYRTEIDKPSQHHHHHH445SABA24.1Adnectin core 24MGVSDVPRDLEVVAATPTSLLISWYDPYTYGand AdCT1 terminalSYYRITYGETGGNSPVQEFTVGPYTTTATISsequences with His6 tagGLKPGVDYTITVYAVSYYYSTQPISINYRTEsequence having AdNT1IDKPSQHHHHHH446SABA25.1Adnectin core 25MGVSDVPRDLEVVAATPTSLLISWSNDGPGLand AdCT1 terminalSYYRITYGETGGNSPVQEFTVPSSQTTATISsequences with His6 tagGLKPGVDYTITVYAVSYYTKKAYSAGPISINsequence having AdNT1YRTEIDKPSQHHHHHH447SABA26.1Adnectin core 26MGVSDVPRDLEVVAATPTSLLISWPDPYYKPsequence having AdNT1DYYRITYGETGGNSPVQEFTVPRDYTTATISand AdCT1 terminalGLKPGVDYTITVYAVYSYYGYYPISINYRTEsequences with His6 tagIDKPSQHHHHHH EXAMPLES Example 1 Material and Methods Used Herein High Throughput Protein Production (HTPP) Selected binders cloned into pET9d vector and transformed intoE. coliHMS174 cells were inoculated in 5 ml LB medium containing 50 μg/mL kanamycin in a 24-well format and grown at 37° C. overnight. Fresh 5 ml LB medium (50 μg/mL kanamycin) cultures were prepared for inducible expression by aspiration 200 μl from the overnight culture and dispensing it into the appropriate well. The cultures were grown at 37° C. until A6000.6-0.9. After induction with 1 mM isopropyl-β-thiogalactoside (IPTG) the culture was expressed for 6 hours at 30° C. and harvested by centrifugation for 10 minutes at 2750 g at 4° C. Cell pellets (in 24-well format) were lysed by resuspension in 450 μl of Lysis buffer (50 mM NaH2PO4, 0.5 M NaCl, 1× Complete Protease Inhibitor Cocktail-EDTA free (Roche), 1 mM PMSF, 10 mM CHAPS, 40 mM Imidazole, 1 mg/ml lysozyme, 30 μg/ml DNAse, 2 μg/ml aprotonin, pH 8.0) and shaken at room temperature for 1-3 hours. Lysates were clarified and re-racked into a 96-well format by transfer into a 96-well Whatman GF/D UNIFILTER® fitted with a 96-well, 1.2 ml catch plate and filtered by positive pressure. The clarified lysates were transferred to a 96-well Ni-Chelating Plate that had been equilibrated with equilibration buffer (50 mM NaH2PO4, 0.5 M NaCl, 40 mM Imidazole, pH 8.0) and was incubated for 5 min. Unbound material was removed by vacuum. The resin was washed 2×0.3 ml/well with Wash buffer #1 (50 mM NaH2PO4, 0.5 M NaCl, 5 mM CHAPS, 40 mM Imidazole, pH 8.0) with each wash removed by vacuum. Prior to elution each well was washed with 50 μl Elution buffer (PBS+20 mM EDTA), incubated for 5 min and this wash was discarded by vacuum. Protein was eluted by applying an additional 100 μl of Elution buffer to each well. After a 30 minute incubation at room temperature the plate(s) were centrifuged for 5 minutes at 200 g and eluted protein is collected in 96-well catch plates containing 5 μl of 0.5M MgCl2added to the bottom of elution catch plate prior to elution. Eluted protein was quantified using a BCA assay with SGE as the protein standard. Midscale Expression and Purification of Insoluble Fibronectin-Based Scaffold Protein Binders For expression of insoluble clones, the clone(s), followed by the HIS6tag, are cloned into a pET9d (EMD Bioscience, San Diego, Calif.) vector and are expressed inE. coliHMS174 cells. Twenty ml of an inoculum culture (generated from a single plated colony) is used to inoculate 1 liter of LB medium containing 50 μg/ml carbenicillin and 34 μg/ml chloramphenicol. The culture is grown at 37° C. until A6000.6-1.0. After induction with 1 mM isopropyl-β-thiogalactoside (IPTG) the culture is grown for 4 hours at 30° C. and is harvested by centrifugation for 30 minutes at 10,000 g at 4° C. Cell pellets are frozen at −80° C. The cell pellet is resuspended in 25 ml of lysis buffer (20 mM NaH2PO4, 0.5 M NaCl, 1× Complete Protease Inhibitor Cocktail-EDTA free (Roche), 1 mM PMSF, pH 7.4) using an ULTRA-TURRAX® homogenizer (IKA works) on ice. Cell lysis is achieved by high pressure homogenization (≥18,000 psi) using a Model M-110S MICROFLUIDIZER® (Microfluidics). The insoluble fraction is separated by centrifugation for 30 minutes at 23,300 g at 4° C. The insoluble pellet recovered from centrifugation of the lysate is washed with 20 mM sodiumphosphate/500 mM NaCl, pH7.4. The pellet is resolubilized in 6.0M guanidine hydrochloride in 20 mM sodium phosphate/500M NaCl pH 7.4 with sonication followed by incubation at 37 degrees for 1-2 hours. The resolubilized pellet is filtered to 0.45 μm and loaded onto a Histrap column equilibrated with the 20 mM sodium phosphate/500M NaCl/6.0M guanidine pH 7.4 buffer. After loading, the column is washed for an additional 25 CV with the same buffer. Bound protein is eluted with 50 mM Imidazole in 20 mM sodium phosphate/500 mM NaCl/6.0M guan-HCl pH7.4. The purified protein is refolded by dialysis against 50 mM sodium acetate/150 mM NaCl pH 4.5. Midscale Expression and Purification of Soluble Fibronectin-Base Scaffold Protein Binders For expression of soluble clones, the clone(s), followed by the HIS6tag, were cloned into a pET9d (EMD Bioscience, San Diego, Calif.) vector and were expressed inE. coliHMS174 cells. Twenty ml of an inoculum culture (generated from a single plated colony) was used to inoculate 1 liter of LB medium containing 50 μg/ml carbenicillin and 34 μg/ml chloramphenicol. The culture was grown at 37° C. until A6000.6-1.0. After induction with 1 mM isopropyl-β-thiogalactoside (IPTG), the culture was grown for 4 hours at 30° C. and was harvested by centrifugation for 30 minutes at 10,000 g at 4° C. Cell pellets were frozen at −80° C. The cell pellet was resuspended in 25 ml of lysis buffer (20 mM NaH2PO4, 0.5 M NaCl, 1× Complete Protease Inhibitor Cocktail-EDTA free (Roche), 1 mM PMSF, pH 7.4) using an ULTRA-TURRAX® homogenizer (IKA works) on ice. Cell lysis was achieved by high pressure homogenization (≥18,000 psi) using a Model M-110S MICROFLUIDIZER® (Microfluidics). The soluble fraction was separated by centrifugation for 30 minutes at 23,300 g at 4° C. The supernatant was clarified via 0.45 μm filter. The clarified lysate was loaded onto a Histrap column (GE) pre-equilibrated with the 20 mM sodium phosphate/500M NaCl pH 7.4. The column was then washed with 25 column volumes of the same buffer, followed by 20 column volumes of 20 mM sodium phosphate/500M NaCl/25 mM Imidazole, pH 7.4 and then 35 column volumes of 20 mM sodium phosphate/500M NaCl/40 mM Imidazole, pH 7.4. Protein was eluted with 15 column volumes of 20 mM sodium phosphate/500M NaCl/500 mM Imidazole, pH 7.4, fractions were pooled based on absorbance at A280and were dialyzed against 1×PBS, 50 mM Tris, 150 mM NaCl, pH 8.5 or 50 mM NaOAc, 150 mM NaCl, pH4.5. Any precipitate was removed by filtering at 0.22 μm. Example 2 In Vitro Nonclinical Pharmacology KDby SPR The binding characteristics were characterized by Surface Plasmon Resonance (SPR). Human PCSK9 and Cynomolgus PCSK9 were immobilized on separate channels on one dimension of a ProteOn XPR (Bio-Rad) chip surfaces and exposed to 6 different concentrations of 2013E01 in the other dimension of the same SPR chip surface. This allowed kinetic determination in the absence of regeneration. Duplicate chips were used for kinetic determinations of the Human and Cynomolgus PCSK9 at 25° C. Evaluation of the kinetic parameters was performed using the Langmuir interaction model and constant parameter fitting with the ProteOn Manager software. Under these conditions, anti-PCSK9 Adnectins bound to human PCSK9 with dissociation constants (KD) ranging from 80 pM to 1.6 nM and to the cyno PCSK9 with dissociation constants (KD) ranging from 8 nM to 24 nM (Table 7). Association rates were approximately 105M−1s−1, coupled with dissociations that were typically 10−3-10−5s1. For some Adnectins, the off-rates from human PCSK9 were slow (on the order of 10−5s1), which is close to the limit of detection for SPR technologies so it is possible that these dissociation constant measurements from human PCSK9 are under-estimates. TABLE 7SPR-Determined Kinetic Parameters for Anti-PCSK9 AdnectinsAgainst Directly Immobilized Human and Cyno PCSK9PCSK9konkoffKDClone IDSpecies(M−1s−1)(s−1)(nM)1459D05human1.13E+051.80E−041.58 ± 0.1762013E01human7.03 ± 0.1E+052.42 ± 0.3E−050.292 ± 0.0082013E01cyno2.19E+051.77E−038.11922G04human5.41E+055.08E−050.094 ± 0.0091922G04cyno4.65E+057.00E−0315.032011H05human1.18E+059.76E−060.079 ± 0.0382011H05cyno1.90E+054.40E−0323.12012A04human2.59E+056.47E−050.251 ± 0.0112012A04cyno1.95E+054.75E−0324.32 KDby BLI The binding characteristics of Adnectins and human PCSK9 were also determined by Bio-Layer Interferometry (BLI). Biotinylated human PCSK9 was immobilized onto superstreptavidin sensor tips which were subsequently immersed into wells containing diluted Adnectin for the duration of the association phase. Tips were then immersed into a buffer-only well for observation of Adnectin dissociation. Experiments were performed in a temperature controlled environment at either 25 or 37° C., and the oscillation speed was set at 1500 rpm. Binding interaction analysis was performed using proprietary software from Fortebio (Fortebio Data Analysis Software version 6.3.0.36). Global fits were performed for all samples, using a 1:1 binding model. The nature of these global fits constrained all values of concentration to a single pair of association and dissociation rates that were themselves constrained to each other. Affinities (KD), association and dissociation rates were averaged over the various loading levels used in the analysis. Under these conditions, Adnectins bound human PCSK9 with affinities ranging from 200 pM to 7.5 nM, as shown in Table 8 below. Association rates ranged from 104-105M−1s−1, and coupled with dissociations that were typically 10−3-10−5s−1. TABLE 8BLI-Determined Kinetic Parameters forPCSK9 Adnectins Against Human PCSK9Clone IDkonkoffKDor ATI#(M−1s−1)(s−1)(nM)2381D042.66E+05 ± 6.4E+046.90E−05 ± 6.9E−050.237 ± 0.082382D095.33E+051.64E−040.3042382D055.05E+05 ± 2.0E+051.57E−04 ± 4.2E−050.314 ± 0.062381B043.09E+05 ± 2.1E+041.60E−04 ± 1.6E−040.527 ± 0.112382E054.36E+05 ± 1.6E+052.50E−04 ± 1.1E−050.604 ± 0.212382B094.51E+05 ± 4.7E+042.91E−04 ± 4.0E−050.656 ± 0.022382H034.71E+05 ± 1.4E+055.57E−04 ± 2.3E−040.677 ± 0.022382C093.49E+052.33E−040.7572971A035.74E+054.20E−040.8062382G044.19E+054.64E−041.112381G092.71E+052.91E−041.122451C063.47E+05 ± 5.4E+044.35E−04 ± 5.4E+041.27 ± 0.142382H102.88E+053.82E−041.402013E015.46E+058.37E−041.512382D033.54E+056.63E−041.532381F113.23E+055.22E−041.592971E023.55E+055.55E−041.782382H092.51E+054.83E−041.862451H074.48E+059.40E−042.082382B103.94E+059.77E−042.352382C053.68E+058.79E−042.492971A093.99E+051.05E−032.792382E032.20E+055.96E−042.942381H091.36E+054.60E−043.232381B022.65E+058.31E−043.292381B082.13E+059.40E−044.112382F052.44E+051.09E−034.541459D051.49E+05 ± 2.5E+042.02E−03 ± 2.0E−0314.4 ± 0.2ATI 10912.863E+058.201E−050.293ATI0011171.451E+055.074E−050.554ATI0010575.325E+051.403E−040.255ATI0011198.190E+044.450E−045.296ATI00116815.829E+053.335E−040.586ATI00117526.558E+053.522E−040.543ATI 10818.29E+05 ± 8.1E+053.49E−04 ± 3E−040.479 ± 0.11ATI8911.08E+054.03E−043.56ATI11148.072E+043.952E−044.876ATI11741.265E+059.012E−047.3971ATI001168 is a deimmunized version of 1922G04 having an R23E substitution.2ATI001175 is a deimmunized version of 1922G04 having an R23D substitution. Solution Phase Affinity KinExA The solution affinities of ATI001081 and ATI001174 for human PCSK9 were measured using a Kinetic Exclusion Assay (KinExA). The relative unbound Adnectin concentrations were measured by capture on a hPCSK9 solid matrix followed by detection with a fluorescently labeled antibody that recognizes the Adnectin scaffold. Due to technical limitations, the lowest concentration of Adnectins that could be tested was 1 nM. The global KDanalyses estimate a KD=70 pM (28-127 pM within a 95% confidence interval) for ATI001081 and a KD=223 pM for ATI001174 (range of 54-585 pM within 95% confidence interval). TABLE 9Solution Phase Affinity Measurements for PCSK9 AdnectinsATI001081ATI001174KD69.5 pM223 pM95% confidence interval:Kd high127 pM585 pMKd low28 pM54 pM The thermodynamics and stoichiometry of binding of Adnectins ATI001174 and ATI001081 to human PCSK9 were characterized by isothermal titration calorimetry (ITC). Solution phase binding was measured in 25 mM HEPES, pH 7.41, 150 mM NaCl at 37° C. An average unimolecular binding constant of 1.3±0.2 nM was observed for ATI001174 and 1.4±0.4 nM for ATI001081. Detailed thermodynamic analyses are shown in Table 10 andFIG.13. The difference in observed enthalpies (−3.3 kcal/mol) for the two Adnectins suggests that ATI001174 incurs an order of magnitude reduction in its affinity for PCSK9 due to PEGylation that is at least partially offset by the corresponding difference in entropy (−10.4 cal/mol·K). TABLE 10StoichiometryKAKDΔHΔSAdnectin(N)(M−1)(nM)(kcal/mol)(cal/mol ° C.)ATI0010810.9267.1E8 ± 2.1E81.4 ± 0.4−26.6 ± 0.2−45.4(±30%)ATI001174*0.8577.5E8 ± 1.5E81.3 ± 0.2−29.9 ± 0.1−55.8*Average of 3 experiments Fluorescence Resonance Energy Transfer (FRET) Assay Three FRET based assays were developed to determine the binding affinity and potency of PCSK9-binding Adnectins, adapted from the general method described previously by Maio et al. (See, Miao, B. et al.,Meth. Enzymol.,357:180-188 (2002)). The PCSK9:EGFA FRET assay (FIGS.2and3) measured the inhibition of PCSK9 binding to the low density lipoprotein receptor (LDLR) epidermal growth factor precursor homology domain (EGFA domain), using recombinant human PCSK9 expressed in baculovirus and a synthetic 40-mer EGFA peptide (biotinylated). EGFA has been shown to represent the key interacting domain of LDLR with PCSK9 (Kwon, H. J. et al.,Proc. Natl. Acad. Sci. USA,105(6):1820-1825 (2008)). This assay used a PCSK9 C-terminal domain binding mAb (mAb 4H5) labeled with Eu-chelate to provide FRET interaction with biotinylated EGFA through the streptavidin/allophycocyanin fluorophore complex. Two other related, PCSK9-dependent FRET assays were also constructed. In one of these assays, competitive displacement by Adnectins of biotinylated Adnectins—ATI000972 or ATI001125 is quantified (ATI000972 results shown inFIG.4). ATI000972 is a biotinylated version of 1459D05 and ATI001125 is a biotinylated version of ATI001081. In another assay, direct binding of an Adnectin (his-tagged) to PCSK9 is assayed using anti-his6 antibody (FIG.5). In each of the FRET assays, human PCSK9 concentration was either 1 or 5 nM. In some cases cynomolgus monkey PCSK9 replaced hPCSK9. Table 11 summarizes the overall data from these three FRET assays. TABLE 11Summary of Adnectin Testing Data for 3 FRET Assays Using Human PCSK9 and 1 Assay for Cyno PCSK9Direct bindinghPCSK9:EGFAhPCSK9:ATI000972hPCSK9:ATI001125hPCSK9cPCSK9:EGFAIC50IC50IC50EC50IC50Clone ID(nM)(nM)(nM)(nM)(nM)1459D054.014nd3.923001784F0321.9nd1.61061813E021.32.4nd2.31181923B022.33.2nd3.253.51922G041.22.0nd2.526.62012A042.11.2nd1.470.32011H052.71.4nd2.017.22013E011.61.8nd0.9012.51922G04 (R25D)2.5ndndnd48.11922G04 (R25E)3.5ndndnd58.52382E032.4nd0.53.710.82382E052.8nd0.43.812.62381B082.6nd14.127.12381B022.5nd0.58.49.82381B042.4nd0.54.217.32451H070.2nd0.213.227.12381D042.9nd0.53.7122381F114nd0.8<126.52381G093.1nd1.14.525.22381H093.4nd0.54.120.22382B093.8nd0.64.037.22382B103nd0.53.118.12382C054nd0.93.427.22382C093.5nd0.62.923.92382D033.3nd0.63.311.82382D092.6nd0.43.713.92382F052.4nd0.93.814.62382G042.9nd0.53.620.12382H033.4nd0.33.819.82382H090.9nd0.33.617.92382H102.6nd0.44.618.22382D024.5nd0.55.357.82451C061.2nd0.45.524.7 Cell-Based Inhibition of PCSK9 Activity by PCSK9 Adnectins DiI-LDL Uptake Assay Cell culture methods were developed to assay the ability of Adnectins to inhibit PCSK9 activity on the LDLR. An effective means of measuring cellular LDLR activity is through an assay for uptake of labeled LDL, as shown by Lagace, T. A. et al. (J. Clin. Invest.,116(11):2995-3005 (2006)). The work further adapted a method for LDLR functional activity using fluorescent-labeled LDL (DiI-LDL) uptake adapted from a method originally shown by Teupser et al. (Biochim. Biophys. Acta,1303(3):193-198 (1996)). Cells were first preincubated with recombinant human PCSK9 protein (10 ug/mL, 135 nM) in the presence and absence of Adnectins as shown. After 2 hours, the remaining LDLR activity was assayed by incubation with DiI-LDL (5 ug) for 2 hours followed by an assessment of accumulated DiI-LDL inside the cells using high content fluorescent micrscopy and image analysis (Cellomics).FIG.6shows the effect of several Adnectins to inhibit PCSK9 activity and restore LDLR functional activity in HepG2 cells. In this assay, the Adnectins inhibited PCSK9 and restored DiI-LDL uptake with the following EC50values: 1459D05, EC50=190 nM; 1784F03, 210 nM; 2012A04, 130 nM; 2013E01, 160 nM. LDLR Depletion Assay HepG2 cells were grown in complete media, Eagle's Minimum Essential Medium (EMEM, ATCC®) with 10% FBS (Hyclone), and split twice a week with Trypsin 0.25% (Invitrogen). To induce upregulation of the LDL receptor, cells were incubated overnight in LPDS media [RPMI (ATCC) with 5% lipoprotein deficient serum (Intracel), 100 nM superstatin (BMS) and 50 uM Sodium Mevalonate (Sigma)]. The following day, cells were trypsinized briefly with Trypsin 0.05% (Invitrogen) and resuspended at 2×10{circumflex over ( )}6 cells per ml then aliquoted at 100 ul per well in a V-bottom 96 well plate. In the meantime, Adnectins were pre-incubated with PCSK9 in LPDS media for an hour at 3TC. After an hour, cells were centrifuged and resuspended in 100 ul of Adnectin/PCSK9 mix and incubated overnight at 37° C. The following day cells were labeled with an antibody for LDL receptor (BAF 2148 from R&D), followed by a phycoerythrin (PE)-streptavidin conjugated secondary antibody (BD554061 from BD Pharmingen) and analyzed by FACS on the FACS CantoII (BD). 10 nM of PCSK9 was pre-incubated for an hour with increasing concentration of Adnectin candidates before being added to HepG2 cells. After overnight incubation LDLR level were measured by FACS and the percentage of inhibition of PCSK9-induced LDLR depletion was graphed and EC50 determined using PRISM. 1459D05 appears to be the least potent candidate among those tested and did not reach the maximum inhibition whereas the other clones reach 150-200% maximum inhibition of PCSK9 (FIG.7). A summary of the PCSK9 Adnectin in vitro pharmacology data is shown below in Table 12. TABLE 12Cyno cross-reactivityKD hPCSK9KDhPCSK9Cyno PCSK9:Tm(37° C.,(25° C.,KDcPCSK9EGFAPCSK9:LDLR Depletion% monomer(° C.,Octet Red)ProteOn)(25° C., ProteOn)FRETGFA FRET% inhibitionEC50Clone ID(SEC-HPLC)DSC)(nM)(nM)(nM)(EC50, nM)(EC50, nM)at 75 nM(nM)1459D05≥956314.41.58>1000>10005.866.8>2001813E02≥9570ndndnd118.52.7nd161784F03≥9565ndndnd106.52.01150.226 ± 131923B02≥9573nd0.17nd53.52.317823 ± 71922G0410083nd0.0915.026.61.2105.110 ± 22013E0110081nd0.298.112.51.6165.510 ± 42012A0410084nd0.2524.370.32.1144.512 ± 62011H0510076nd0.0823.117.22.7197.612 ± 52382D0597860.314ndnd57.82.6140.5nd2382E0396832.94ndnd10.82.489.5nd2382E0595840.604ndnd12.62.8103.5nd2381B0296683.29ndnd9.82.5125.4nd2381B0498770.527ndnd17.32.4121.6nd2381B0897784.11ndnd27.12.6124.8nd2381D0498700.237ndnd122.9119.2nd2381F1195791.59ndnd26.54110.2nd2381G0996781.12ndnd25.23.1133.0nd2381H0977633.23ndnd20.23.494.4nd2382B0999880.656ndnd37.23.8105.0nd2382B1099822.35ndnd18.13100.2nd2382C0597852.49ndnd27.24105.3nd2382C0996850.757ndnd23.93.5121.7nd2382D0397841.53ndnd11.83.380.4nd2382D0993840.304ndnd13.92.6109.1nd2382F0599834.54ndnd14.62.472.2nd2382G0495811.11ndnd20.12.9146.1nd2382H0396850.677ndnd19.83.4102.1nd2382H0995811.86ndnd17.90.9101.2nd2382H1097871.40ndnd18.22.6118.6nd2451B06ndndndndnd57.84.592.2nd2451C0696871.27ndnd24.71.289.4nd2451H0797872.08ndnd27.10.288.8nd Example 3 In Vivo Pharmacodynamic Effects of PCSK9 Adnectins Human PCSK9 Transgenic Mouse Models PCSK9 Adnectins exhibited pharmacodynamic effects in vivo in two different human transgenic mouse models. One mouse model overexpresses human PCSK9 levels markedly and exhibits hypercholesterolemia as a result (Lagace, T. A. et al., J. Clin. Invest., 116(11):2995-3005 (2006)). The other mouse model is a genomic hPCSK9 transgenic (BAC-transgenic) which is regulated in liver similarly to mouse PCSK9 and which expresses near human-normal levels of hPCSK9 in plasma. For these studies, ELISA assays using species-specific, site-specific labeled antibodies and Adnectins were developed to measure plasma unbound human PCSK9 levels (i.e., hPCSK9 not complexed with the administered Adnectin) as an index of target engagement. Single doses of PCSK9 Adnectins in PEGylated form were injected intraperitoneally into the overexpresser hPCSK9 transgenic mouse model at the doses shown inFIGS.8-9. PBS or dialysate samples were also injected as controls. Adnectin 1459D05-PEG (100 mg/kg intraperitoneal) treatment rapidly decreased plasma total cholesterol (FIG.8A) and LDL-C (not shown) to >35% below baseline in 4 hr. Cholesterol levels in Adnectin treated mice remained below control levels throughout the 48 hr test period. This was accompanied by a sharp decrease in circulating levels of unbound hPCSK9 in the Adnectin treated transgenic mice (FIG.8B). Western blots of liver taken at 6 hours in parallel studies showed that LDLR protein levels were increased ˜2-fold in Adnectin-treated mice (not shown). In further studies in this transgenic mouse model, ATI001114 was administered at 10 or 60 mg/kg. A marked, dose-dependent, rapid lowering of plasma cholesterol was seen, concomitant with a dose-related reduction in unbound hPCSK9 levels (FIG.9). These studies represent in vivo proof-of-concept for PCSK9 Adnectins as effective cholesterol lowering agents in a hypercholesterolemic human transgenic PCSK9 mouse model. In vivo studies were conducted in the normal expresser hPCSK9 transgenic mouse model. Injection of single doses of 1459D05-PEG or ATI001114 (5 mg/kg) resulted in rapid and strong decreases in unbound hPCSK9 levels in plasma (FIG.10). This pharmacodynamic effect on unbound hPCSK9 was more pronounced following ATI001114 compared to 1459D05-PEG, with greater magnitude and duration of effect observed for the higher affinity/potency Adnectin. A further study of dose dependency revealed that that the 50% inhibitory dose (ED50) was less than 0.1 mg/kg for ATI001114 at time points from 3 to 48 hours post-dose, as seen inFIG.11. These findings in a normal expresser transgenic mouse model show that PCSK9 inhibitory Adnectins exhibit marked, affinity-dependent and dose-dependent effects on pharmacodynamic endpoints which are correlated with LDLR regulation and LDL cholesterol lowering. Cynomolgus Monkeys A pharmacodynamic study was conducted in normal lean cynomolgus monkeys. Adnectin ATI001114 was administered to cynos intravenously at 5 mg/kg, and plasma samples were collected at time intervals for LDL-C assay and pharmacokinetic assessment. A single dose of ATI001114 rapidly lowered plasma LDL-C levels to >50% vs. baseline (or vs. PBS control group) within 48 hours (FIG.12). The duration of effect on LDL-C continued for more than a week with eventual return to baseline by 3 wk. This effect was observed with both two-branched and four-branched 40 kDa PEGylated forms of the anti-PCSK9 Adnectin (ATI001114 and ATI001211, respectively). ATI001211 is ATI001081 with a 40 kDa 4-branched NOF PEG moiety. Total cholesterol showed a similar pattern but no effect on HDL or other metabolic parameter was observed (not shown). Pharmacokinetic analysis revealed that the plasma half-life was approximately 80-120 hrs, consistent with the pharmacodynamics of LDL lowering in the cynos. These findings indicate that a PCSK9 Adnectin is efficacious and fast-acting with rapid, robust, specific effects on LDL-C lowering in cynomolgus monkey model. Example 4 In Vitro and In Vivo Pharmacological Evaluation of the PCSK9 Adnectin-Fc Fusion Protein, PRD460 Production of PRD460 A vector encoding PRD460 was transfected into HEK-293 6E cells using polyethylenimine (PEI). The cells were grown at 37° C. for 5 days with 80% humidification and 5% CO2. The cells were then pelleted, the supernatant was passed through a 0.22 um filter and then loaded onto a ProteinA column. The column was washed with PBS and the protein was eluted with 20 mM Glycine, 150 mM NaCl pH 2.8. The eluted protein was concentrated and passed over a superdex200 column in 50 mM MES, 100 mM NaCl pH 5.8. PRD460 KDby SPR The binding characteristics were characterized by Surface Plasmon Resonance (SPR). Anti-human antibody was immobilized on a BIACORE® chip, and PRD460 (sequence as set forth in SEQ ID NO: 322) was captured on the chip surface. Varying concentrations of hPCSK9 were placed into the flow solution using MgCl2(3 M) for chip regeneration between cycles. For comparison, ATI-1081 was captured on an anti-His antibody immobilized on a BIACORE® chip. Duplicate experiments for PRD460 were performed on different days. Kinetic determinations were performed at 25° C. Evaluation of the kinetic parameters was performed using the 1:1 Binding algorithm on the BIACORE® Evaluation software. Under these conditions, ATI-1081 bound to human PCSK9 with a dissociation constant (KD) of 6.7 nM at 25° C. and PRD460 bound to human PCSK9 with a dissociation constant (KD) of 3.29+/−0.55 nM at 25° C. (Table 13). The off-rate determinations using this assay format may be artificially limited by the off-rate of the captured ligand from the immobilized capture antibody, thus the assay format using direct immobilization of PCSK9 is a more accurate reflection of dissociation constant (KD) for ATI-1081. TABLE 13Kinetic Parameters for PRD460 and ATI-1081 Against Captured Human PCSK9kakdKD(1/Ms)(1/s)(nM)PRD4603.75 +/− 0.7E+041.21 +/− 0.05E−043.29 +/− 0.55ATI-10813.65E+042.45E−046.7 PCSK9 Binding FRET Assays Two fluorescence resonance energy transfer (FRET) based assays were used to determine the competitive binding potency of PRD460 and other Adnectins to hPCSK9. The PCSK9:EGFA FRET assay measures the binding of PCSK9 to the LDLR, using a soluble epidermal growth factor precursor homology domain-A (EGFA) peptide and recombinant human PCSK9. The PCSK9:ATI972 FRET assay measures competitive displacement by Adnectins of the biotinylated Adnectin, ATI-972, from PCSK9. In the PCSK9:EGFA FRET assay (at 5 nM PCSK9), PRD460 completely and potently displaced EGFA from the PCSK9 binding site with EC50=0.7 nM (FIG.1, left panel). PRD460 was more potent in this assay than either ATI-1174 (EC50=1.9 nM) or ATI-1081 (EC50=3.7 nM) (FIG.14). The greater apparent potency of PRD460 in this assay may be explained by bivalent (2:1) binding of Adnectin PRD460 to PCSK9 (theoretically) compared to monovalent (1:1) binding by ATI-1081 and ATI-1174. Using the PCSK9:ATI-972 FRET assay (at 5 nM human PCSK9), PRD460 inhibited with EC50=0.3 nM, compared to 0.8 nM for ATI-1114 and 2.8 nm for ATI-1081 (FIG.15). These findings indicate that PRD460 potently displaced the biotinylated Adnectin ATI-972 from its binding site on PCSK9. The higher potency of PRD460 relative to ATI-1081 and ATI-1174 is consistent with bivalent binding by PRD460. Inhibition of PCSK9-Induced LDLR Depletion in HepG2 Cells Human PCSK9 promotes the depletion of LDLR from the surface of HepG2 cells. Pre-incubation of PCSK9 with PCSK9 Adnectins inhibits PCSK9 binding to LDLR and prevents the depletion of LDLR from the cell surface. This assay was used to measure the potency of ATI-1081, ATI-1174 and PRD460 to inhibit PCSK9 induced depletion of LDLR from the cell surface. A dilution series of PCSK9 Adnectins were pre-incubated with 10 nM human PCSK9 for 1 hr at 37 degrees, the pre-incubated mixture was added to HepG2 cells, and the cells were incubated for 24 hours. Following this incubation, the level of LDLR on HepG2 cells was measured using FACS analysis. The percentage of inhibition of PCSK9-induced LDLR depletion was calculated and graphed (FIG.15). In this assay ATI-1081, ATI-1174, and PRD460 inhibited PCSK9 with comparable EC50's (9 nM, 8 nM and 6 nM respectively) although a leftward-shift of the response curve was consistently observed for PRD460. These EC50's represent the limit of the assay. PCSK9 Cell Entry Assay in HepG2 Cells PCSK9 binding to the LDLR on the surface of hepatocytes results in co-internalization of the LDLR-PCSK9 complex during LDLR endocytosis, leading to enhanced degradation of the LDLR. A cell-based assay was developed to measure LDLR-dependent cellular entry of fluorescent PCSK9. Human PCSK9 was covalently labeled using the fluorophore ALEXA FLUOR®-647 (AF647). PCSK9-AF647 was incubated with HepG2 cells with or without PCSK9-Adnectins and the intracellular fluorescence was quantified by high content fluorescent microscopy and image analysis (Cellomics). Dependence of PCSK9-AF647 cell entry on LDLR endocytosis was established in preliminary experiments. HepG2 cells were incubated with 10 nM PCSK9-AF647 and varying levels of Adnectins for 4 hrs at 37 degrees. In this assay, potent inhibition of PCSK9-AF647 intracellular fluorescence was observed for PRD460 (EC50=6 nM) as well as for ATI-1174 (EC50=10 nM) (FIG.16). These findings indicate that Adnectin PRD460 and ATI-1174 effectively blocked the binding of PCSK9 to cell surface LDLR in a human hepatic-derived cell line in culture, thereby reducing the internalization of PCSK9-AF647 during LDLR endocytosis. In Vivo Transgenic Mouse Study In vivo studies were conducted in the normal expresser hPCSK9 transgenic mouse model. Binding of Adnectins to PCSK9 in the plasma is predicted to result in a decrease in the measured amount of unbound (free) circulating PCSK9. The decrease in unbound PCSK9 is the initial pharmacodynamic event which results in inhibition of the PCSK9-LDLR interaction and in LDL cholesterol lowering. Administration of single doses of PRD460 (i.p. doses from 0.6 to 18 mg/kg) to the transgenic mice resulted in rapid, strong decreases in plasma unbound hPCSK9 levels (FIG.17). Dose-dependent decreases in unbound PCSK9 were observed with ED50<0.6 mg/kg at the 3 hr time point. These findings in the normal expresser human PCSK9 transgenic mouse model show that PRD460 binds strongly and potently to circulating hPCSK9 in vivo. In Vivo Pharmacodynamics in Cynomolgus Monkeys The pharmacodynamic effects of PCSK9 Adnectin PRD460 were evaluated in normal lean cynomolgus monkeys. PRD460 was administered to monkeys by i.v. dosing at 15 mg/kg, and plasma samples were collected at time intervals over 4 wks for the assay of LDL-C and free PCSK9 levels. A single dose of PRD460 rapidly lowered plasma LDL-C levels in the monkeys, reaching an average maximum effect of 42% of baseline LDL-C (58% reduction; n=3 monkeys) by day 3 after dosing (FIG.18). LDL-C levels were reduced by 50% or more for a week at this dose, remaining significantly below baseline for 3 wks and returning to baseline by 4 wks. Total cholesterol showed a similar pattern but no effect on HDL was observed (not shown). Treatment with PRD460 caused an immediate drop to near zero (below the lower limit of quantitation) in the unbound, free form of plasma PCSK9 (FIG.18). The free PCSK9 levels remained near the lower limits of detection for several days then gradually returned to baseline levels by the end of 4 wks, consistent with a cause/effect relationship with plasma LDL-C. The data indicate that plasma LDL lowering mirrored the drop in free PCSK9 levels, consistent with PCSK9 inhibition regulating LDLR function following treatment with PRD460 in vivo. Pharmacokinetic analysis revealed that the plasma half-life of Adnectin PRD460 was approximately 70 hrs in this cynomolgus monkey study. These findings indicate that a PCSK9 Adnectin-Fc fusion protein is highly efficacious and fast-acting with robust, specific, and long-lasting effects on LDL-C lowering in the cynomolgus monkey model. In Vivo Pharmacological Evaluation of the Unmodified PCSK9 Adnectin, ATI-1081 In addition to modified Adnectins containing a PK moiety (e.g., PEGylated and Fc-fusion Adnectins), an unmodified (“naked”) PCSK9 Adnectin can also be administered. Strategies for unmodified PCSK9 Adnectin treatment include more frequent dosing to accommodate the shorter PK half-life, or using an extended release subcutaneous formulation to increase the length of the absorption phase and extend the pharmacodynamic effect. Many such formulations can be envisioned including, as a simple example, propylene glycol/PBS solutions to delay the rate of absorption and increase the time of exposure to the Adnectin in the circulation. The unmodified Adnectin ATI-1081 was administered to cynomolgus monkeys i.v. at 10 mg/kg in PBS vehicle. ATI-1114 (PEGylated version of the same Adnectin) was also administered at 1 mg/kg in PBS as a comparator. ATI-1081 elicited a rapid, transient inhibition of unbound circulating PCSK9 levels. Within 30 minutes the extent of initial inhibition approached 100% (below the limits of quantitation) before returning to baseline several hours later (FIG.19). Concurrently, a trend to lower LDL-C levels was observed over the first 24 hrs in the ATI-1081 treated monkeys (FIG.20). The unmodified Adnectin ATI-1081 was also administered to the normal expresser hPCSK9 transgenic mice in a simple extended release subcutaneous formulation using 50:50 propylene glycol:PBS vehicle (PG vehicle) compared to PBS vehicle. This formulation is expected to modestly delay the rate of Adnectin absorption and increase the exposure time to ATI-1081 in the circulation, thus improving the pharmacodynamic response. Administration (intraperitoneal) of ATI-1081 at 1 mg/kg in PBS vehicle resulted in ˜50% lowering of unbound plasma PCSK9 at 3 hr, compared to >85% lowering for ATI-1114 at 0.3 mg/kg (FIG.21). In a second experiment in the transgenic mice, ATI-1081 administered by subcutaneous injection in PG vehicle resulted in nearly equivalent decreases in unbound LDL compared to ATI-1174, with an improved duration of effect over the first 6 hrs of the study (FIG.22). These findings indicate that the unmodified PCSK9 Adnectin ATI-1081 when administered subcutaneously in a simple extended release vehicle bound to the target human PCSK9 in vivo and elicited the initial pharmacodynamic response. The time dependency of the response was consistent with extended release prolonging the duration of effect for the unmodified PCSK9 Adnectin. Example 5 Serum Albumin-Binding Adnectins (SABA) Example 5A. Screening and Selection of Candidate Serum Albumin-Binding Adnectin Overview A selection technique known as PROfusion (see e.g., Roberts et al.,Proc. Natl. Acad. Sci. USA,94(23):12297-12302 (1997) and WO 2008/066752) was applied to a DNA library with variable regions designed into the BC, DE and FG loops of10Fn3. A random library of greater than 1013molecules was created from this design, and selection pressure was applied against a biotinylated form of HSA to isolate candidate serum albumin-binding Adnectin (SABA) with desirable binding properties. High Throughput Protein Production (HTTP) Process The various HSA binding Adnectins were purified using a high throughput protein production process (HTPP). Selected binders were cloned into pET9d vector containing a HIS6 tag and transformed intoE. coliBL21(DE3)pLysS cells. Transformed cells were inoculated in 5 ml LB medium containing 50 μg/mL Kanamycin in a 24-well format and grown at 37° C. overnight. Fresh 5 ml LB medium (50 μg/mL Kanamycin) cultures were prepared for inducible expression by aspirating 200 μl from the overnight culture and dispensing it into the appropriate well. The cultures were grown at 37° C. until A6000.6-0.9. After induction with 1 mM isopropyl-β-thiogalactoside (IPTG), the culture was grown for another 4 hours at 30° C. and harvested by centrifugation for 10 minutes at 3220×g at 4° C. Cell Pellets were frozen at −80° C. Cell pellets (in 24-well format) were lysed by resuspension in 450 μl of Lysis buffer (50 mM NaH2PO4, 0.5 M NaCl, 1× Complete Protease Inhibitor Cocktail-EDTA free (Roche), 1 mM PMSF, 10 mM CHAPS, 40 mM Imidazole, 1 mg/ml lysozyme, 30 ug/ml DNAse, 2 ug/ml aprotonin, pH 8.0) and shaken at room temperature for 1 hour. Lysates were clarified and re-racked into a 96-well format by transfer into a 96-well Whatman GF/D UNIFILTER® fitted with a 96-well, 650 μl catch plate and centrifuged for 5 minutes at 200×g. The clarified lysates were transferred to a 96-well Ni-Chelating Plate that had been equilibrated with equilibration buffer (50 mM NaH2PO4, 0.5 M NaCl, 10 mM CHAPS, 40 mM Imidazole, pH 8.0) and incubated for 5 min. Unbound material was removed. The resin was washed 2×0.3 ml/well with Wash buffer #1 (50 mM NaH2PO4, 0.5 M NaCl, 5 mM CHAPS, 40 mM Imidazole, pH 8.0). Next the resin was washed with 3×0.3 ml/well with PBS. Prior to elution each well was washed with 50 μl Elution buffer (PBS+20 mM EDTA), incubated for 5 min and this wash discarded by vacuum. Protein was eluted by applying an additional 100 ul of Elution buffer to each well. After 30 minute incubation at room temperature the plate(s) were centrifuged for 5 minutes at 200×g and eluted protein collected in 96-well catch plates containing 5 μl of 0.5M MgCl2affixed to the bottom of the Ni-plates. Eluted protein was quantified using a BCA Protein assay with SGE (control Adnectin) as the protein standard. The SGE Adnectin is a wild-type10Fn3 domain (SEQ ID NO: 1) in which integrin binding domain (amino acids RGD at positions 78-80) have been replaced with SGE. HSA, RhSA and MuSA Direct Binding ELISA For assaying direct binders to HSA, MaxiSorp plates (Nunc International, Rochester, N.Y.) were coated with 10 ug/mL HSA (Sigma, St. Louis, Mo.) in PBS at 4° C. overnight followed by blocking in casein block buffer (Thermo Scientific, Rockford, Ill.) for 1-3 hours at room temperature. For single-point screening assays, purified HTPP Adnectin were diluted 1:20 in casein block buffer and allowed to bind to HSA in each well for 1 hour at room temperature. For dose response assays, concentrations ranging from 0.1 nM up to 1 μM were used. After washing in PBST to remove unbound Adnectins, anti-His mAb-HRP conjugate (R&D Systems, MN) diluted 1:2500 in casein block buffer was added to the bound His-tagged Adnectin for 1 hour at room temperature. Excess conjugate was removed by washing with PBST and bound Adnectins detected using TMB detection reagents (BD Biosciences) according to the manufacturer's instructions. Aggregation Measurement by Analytical Size Exclusion Chromatography Size exclusion chromatography (SEC) was performed on the SABAs resulting from the HTPP. SEC of HTPP derived material was performed using a SUPERDEX® 200 5/150 or SUPERDEX® 75 5/150 column (GE Healthcare) on an Agilent 1100 or 1200 HPLC system with UV detection at A214nm and A280nm and with fluorescence detection (excitation=280 nm, emission=350 nm). A buffer of 100 mM sodium sulfate, 100 mM sodium phosphate, 150 mM sodium chloride, pH 6.8 at appropriate flow rate of the SEC column employed. Gel filtration standards (Bio-Rad Laboratories, Hercules, Calif.) were used for molecular weight calibration. The results of the SEC on the HTPP purified SABAs were shown to be predominantly monomeric and eluted in the approximate range of 10 kDa vs. globular Gel Filtration standards (BioRad). Identification of Candidate Serum Albumin-Binding Adnectin (SABA) As a result of the screening for HSA/RhSA/MuSA binding and biophysical criteria, four unique serum albumin-binding Adnectins (SABA) were identified and chosen to have their half-lives evaluated in mice. In order to carry out in vitro and in vivo characterization, midscales were undertaken for the four SABAs. Table 6 provides the sequences of twenty-six unique SABA core sequences identified from PROfusion, designated as SABA 1-26. SABA4 had a scaffold mutation that was fixed prior to midscaling. The scaffold-perfect version of SABA4 is SABAS. SABA4 and SABAS have identical sequences in the BC, DE, and FG loops. Example 5B. Production and Formulation of Candidate SABAs Midscale Protein Production of SABAs The selected SABAs described in Example 5A, followed by the His6 tag, were cloned into a pET 9d vector and expressed inE. coliBL21(DE3)pLysS cells (see Table 6 for each His-tagged SABA sequence designated SABA1.1, SABA2.1, SABA3.1, and SABA5.1). 20 ml of an inoculum culture (generated from a single plated colony) was used to inoculate 1 liter of LB medium containing 50 μg/mL Kanamycin. The culture was grown at 37° C. until A6000.6-1.0. After induction with 1 mM isopropyl-β-thiogalactoside (IPTG) the culture was grown for another 4 hours at 30° C. and harvested by centrifugation for 30 minutes at ≥10,000×g at 4° C. Cell Pellets were frozen at −80° C. The cell pellet was resuspended in 25 mL of lysis buffer (20 mM NaH2PO4, 0.5 M NaCl, 1× Complete Protease Inhibitor Cocktail-EDTA free (Roche), pH 7.4) using an ULTRA-TURRAX® homogenizer (IKA works) on ice. Cell lysis was achieved by high pressure homogenization (≥18,000 psi) using a Model M-110S MICROFLUIDIZER® (Microfluidics). The soluble fraction was separated by centrifugation for 30 minutes at 23,300×g at 4° C. The supernatant was clarified via 0.45 μm filter. The clarified lysate was loaded onto a HISTRAP® column (GE) pre-equilibrated with 20 mM NaH2PO4, 0.5 M NaCl, pH 7.4. The column was then washed with 25 column volumes of 20 mM NaH2PO4, 0.5 M NaCl, pH 7.4, followed by 20 column volumes of 20 mM NaH2PO4, 0.5 M NaCl, 25 mM imidazole pH 7.4, and then 35 column volumes of 20 mM NaH2PO4, 0.5 M NaCl, 40 mM imidazole pH 7.4. Protein was eluted with 15 column volumes of 20 mM NaH2PO4, 0.5 M NaCl, 500 mM imidazole pH 7.4, fractions pooled based on absorbance at A280and dialyzed against 1×PBS, 50 mM Tris, 150 mM NaCl pH 8.5 or 50 mM NaOAc; 150 mM NaCl; pH 4.5. Any precipitate was removed by filtering at 0.22 μm. Midscale expression and purification yielded highly pure and active Adnectins that were expressed in a soluble form and purified from the soluble fraction of the bacterial cytosol. SEC analysis on a SUPERDEX® 200 or SUPERDEX® 75 10/30GL in a mobile phase of 100 mM NaPO4, 100 mM NaSO4, 150 mM NaCl, pH 6.8 (GE Healthcare) demonstrated predominantly monomeric Adnectins. Formulation of SABA1.2 One specific SABA, SABA1.2 (SEQ ID NO: 411), was chosen for a preliminary formulation screen. SABA1.2 comprises an (ED)5extension on the “core 1” sequence of10Fn3 (see SEQ ID NO: 421 in Table 6). For SABA1.2, a stable formulation of 10 mM succinic acid, 8% sorbitol, 5% glycine at pH 6.0 and at a product concentration of 5 mg/mL was identified. In this formulation the protein melting temperature was 75° C. as determined by Differential Scanning calorimetry (DSC) using a protein concentration of 1.25 mg/mL. The formulation provided satisfactory physical and chemical stability at 4° C. and 25° C., with an initial aggregate level at 1.2%. After one month of stability, the level of aggregation was very low (1.6% at 4° C. and 3.8% at 25° C.). The protein was also stable in this formulation after five cycles of freeze-thaw as transitioned from −80° C. and −20° C. to ambient temperature. In addition, in this formulation SABA1.2 was soluble to at least 20 mg/mL protein concentration at 4° C. and ambient temperature with no precipitation or increase in aggregation. Example 5C. Biophysical Characterization of Candidate SABAs Size Exclusion Chromatography Standard size exclusion chromatography (SEC) was performed on the candidate SABAs resulting from the midscale process. SEC of midscaled material was performed using a SUPERDEX® 200 10/30 or on a SUPERDEX® 75 10/30 column (GE Healthcare) on an Agilent 1100 or 1200 HPLC system with UV detection at A214nm and A280nm and with fluorescence detection (excitation=280 nm, emission=350 nm). A buffer of 100 mM sodium sulfate, 100 mM sodium phosphate, 150 mM sodium chloride, pH 6.8 at appropriate flow rate of the SEC column employed. Gel filtration standards (Bio-Rad Laboratories, Hercules, Calif.) were used for molecular weight calibration. The results of the SEC on the midscaled purified SABAs showed predominantly monomeric Adnectin and elution in the approximate range of 10 kDa vs. globular Gel Filtration standards (BioRad) as showed. Thermostability Differential Scanning calorimetry (DSC) analyses of the midscaled SABAs were performed to determine their respective Tm's. A 1 mg/ml solution was scanned in a N-DSC II calorimeter (calorimetry Sciences Corp) by ramping the temperature from 5° C. to 95° C. at a rate of 1 degree per minute under 3 atm pressure. The data was analyzed vs. a control run of the appropriate buffer using a best fit using Orgin Software (OrginLab Corp). The results of the SEC and DSC analyses are summarized in Table 14. TABLE 14Summary of SEC and DSC Analyses on Candidate SABAsSECMonomerDimerDSCClone(%)(%)(Tm)SABA1.192.37.763.9° C.SABA5.1881270.1° C.SABA2.191958.5° C./78.2° C.SABA3.199BLD65.2° C. Example 5D. Characterization of Candidate SABA1 Binding to Serum Albumin The kinetics of selected SABA clones purified from HTPP and/or midscaled material described in Examples 5A and 5B were determined by capturing the respective serum albumin (HSA/RhSA/MuSA) on the surface of a Biasensor CMS chip and flowing a concentration series of SABAs over both the reference flow cell and the captured albumins. In addition, binding to albumin was carried out under various pH conditions ranging from pH 5.5 to pH 7.4. HSA-binding Adnectins SABA2.1, SABA3.1, SABA4.1, and SABA1.1 cross reacted with RhSA but did not cross react with MuSA. SABA2 and SABA4 binding is pH sensitive whereas clone SABA3 demonstrated pH resistance binding to HSA down to pH 6.0. SABA1.1 fits biochemical criteria for pH resistance and affinity/kinetics down to pH 5.5. Domain mapping was determined by BIACORE®. Selected SABA clones purified from HTPP and/or midscaled material were determined by capturing HSA or a construct consisting of just HSA-domain I & II or HSA-domain III on the surface of a Biasensor CMS chip and flowing a concentration series of the SABAs over both the reference flow cell and the captured albumins. Clones SABA2 & SABA1 bound to HSA and the HSA-domain I-II construct but not the HSA-domain III construct. Clones SABA3 & SABA4 bound to HSA but not to either the HSA-domain I-II or HSA-domain III constructs. The results are summarized in Table 15. TABLE 15Binding Affinity and Kinetics of Candidate SABAs (SABA1.1, 2.1, 3.1 and 4.1)KDKoffResistant toAdnectinTarget(nM)(s−1)pH 7.4→5.5?Epitope on HSASABA2HSA33.8 +/− 20.5 (n = 6)1.71E−04−−−Domain I-IIRhSA63.64.42E−04SABA3HSA8636.82E−02+++Neither domain I-RhSA4313.37E−02(down to pH 6.0)II nor III(interfacial?)SABA4HSA412 +/− 8 (n = 4)7.82E−04−−Neither domain I-RhSA>10003.83E−03II nor III(interfacial?)SABA1HSA47.2 +/− 18.2 (n = 9)4.57E−04+++Domain I-IIRhSA778 +/− 313 (n = 4)5.45E−03 Example 5E. Examination of the In Vivo t1/2of Candidate SABAs The half-life of HSA in mice was determined to allow for evaluation of HSA-binding Adnectins in mice as the HSA-binding Adnectins do not cross react with MuSA. HSA was injected into the tail vein of approximately 6 week old Ncr nude female mice at a 20 mg/kg (FIG.23A) and 50 mg/kg dose (FIG.23B), and the concentration of HSA in blood samples taken at intervals post-injection was determined by ELISA. The t1/2of HSA injected into mice at 20 mg/kg and 50 mg/kg were determined to be ˜24 hrs and ˜20 hrs, respectively. Half-Life Determination of SABA1-4 in Mice One literE. coligrowth of HSA binding clones SABA1.1, SABA2.1, SABA3.1, and SABA4.1 were prepared, purified and endotoxin removed. Each SABA variant was injected into the tail vein of mice, and the concentration in blood samples taken at intervals post-injection was determined by ELISA. The pharmacokinetic profiles of each SABA were compared in the presence or absence of HSA in approximately 6 week old Ncr nude female mice. The mice that were co-injected with HSA had the HSA premixed with each SABA (HSA in a 3-4 molar excess) because the binding clone was selective for HSA and RhSA and did not bind the mouse serum albumin. The half-life of SABA1.1 (clone 1318H04) in mice plasma was 0.56 hours whereas the half-life of SABA1.1 co-injected with HSA was 5.6 hours, a˜10-fold increase in half life (FIG.24A). The half-life of SABA2.1 in mice plasma was 0.24 hours whereas the half-life of SABA2.1 co-injected with HSA was 2.8 hours, a˜12-fold increase in half life (FIG.24B). The half-life of SABA3.1 (clone 1245H07) in mice plasma was 0.28 hours whereas the half-life of SABA3.1 co-injected with HSA was 0.53 hours, a˜2-fold increase in half life (FIG.24C). The half-life of SABA4.1 in mice plasma was 0.66 hours whereas the half-life of SABA4 co-injected with HSA was 4.6 hours, a˜7-fold increase in half life (FIG.24D). A summary of the present example is shown inFIG.25. Table 16 summarizes similar data for SABA1.1, SABA2.1, SABA3.1, and SABA5.1; comparison is made to half life in cyno, where available. TABLE 16PK (T½)CLONEMiceCynoCommentsSABA1.15.6 hrs96-137 hrsT½ = 96-137 hrsSABA5.14.6 hrs12 hrsPoor binding affinityfor RhSA.2-fold decrease in KDobserved at pH <6.0SABA2.12.8 hrsNALoss of binding at pH <6.5SABA3.132 minNAPoor T½ observed in mice Half-Life Determination of SABA1.1 and SABA5.1 in Cynomolgus Monkeys A three week single dose proof of concept study of SABA1.1 (FIG.26A) and SABA5.1 (FIG.26B) was conducted in cynomolgus monkeys to assess pharmacokinetics at a 1 mg per kg (mpk) dose IV in 2 cynomolgus monkeys. The pharmacokinetics were evaluated using a quantitative ELISA-based assay that was developed to detect the Adnectin in plasma samples. SABA1.1 has a half-life in the range of 96-137 hours (FIG.26Aand Table 17A). SABA5.1 has a half-life of approximately 12 hours and was only measureable in the ELISA up to 120 hours (FIG.26B). Table 17A summarizes data for SABA1.1; Table 17B summarizes data for SABA5.1. TABLE 17ASABA1.1t½CmaxAUCallCl_obsVz_obsMonkey(hrs)(μg/mL)(hr*μg/mL)(mL/hr/kg)(mL/kg)#195.89.03673.71.45200.8#2136.67.24625.11.60315.2 TABLE 17BSABA5.1HL_Lambda_zCmaxAUCallCl_obsVz_obs(hr)(μg/mL)(hr*μg/mL)(mL/hr/kg)(mL/kg)N22222Mean12.18617.358246.8824.08972.507SD1.4513.0836.2450.59619.045Min11.1615.18221.253.6759.04Max13.2119.54272.514.5185.97CV %11.917.714.714.626.3 Example 5F. Characterization of SABA1 Binding to Serum Albumin SABA1.1 and 1.2 Binds to HSA and RhSA SABA1.2, a “core 1”10Fn3 comprising an (ED)5extension (SEQ ID NO: 421 in Table 6) bound to human serum albumin (HSA) at neutral pH and 25° C. with an average association rate constant (ka) of 8.21E+03 M−1s−1, and an average dissociation rate constant (kd) of 4.43E-04 s−1, for a calculated average Kdof 55.3 nM (Table 18). For rhesus serum albumin (RhSA), the measured average association rate constant was 6.6E+03 M−1s−1, and the dissociation rate constant was 3.78E-03 s−1, giving a calculated average Kdof 580 nM. No measurable interaction between SABA1.2 and mouse or rat serum albumin could be observed up to 1 μM (Table 18 andFIG.27). At 37° C., the ka and kd increased between 2 to 5-fold, leading to a ˜2-fold increase in affinity for HSA and ½ the affinity for RhSA (Table 18). TABLE 18Kinetic parameters for SABA1.2 bindingto albumins, in HBS-P buffer.TempkakdKDAlbumin(° C.)(1/Ms)(1/s)(nM)Human258.21 ± 1.19E+034.43 ± 0.65E−0455.3 ± 13.7Rhesus6.60 ± 1.18E+033.78 ± 0.45E−03580 ± 62.6Mouseno observable bindingHuman373.38E+048.15E−0424.1Rhesus1.89E+041.85E−02977.4Mouseno observable binding Additionally, a calorimetric titration was performed to determine the stoichiometry between SABA1 and HSA. For this study, SABA1.1, a “core 1”10Fn3 comprising a His6 extension (SEQ ID NO: 420 in Table 6), was used. HSA (10 μl per injection of 115 μM protein solution) was injected into the calorimetric cell containing SABA1.1 at a concentration of 8.1 μM. The experiment was performed at 37° C. in PBS buffer pH 7.4.FIG.28shows that SABA1.1 binds to HSA with 1:1 stoichiometry. SABA1.2 Binds Potently to HSA at Low pH The long half-life of albumins (e.g., t1/2of HSA is 19 days) is due in large part to the fact that they are recycled from an endocytic pathway by binding to the neonatal Fc receptor, FcRn, under the low pH conditions that exist inside the endosome. As shown in Table 19 SABA1.2 potently bound HSA at the endosomal pH of 5.5, suggesting that the t1/2of SABA1, once bound to HSA, would also benefit from the FcRn recycling mechanism. TABLE 19Comparison of Albumin Binding Kineticsat pH 7.4 and 5.5, in MES BufferkakdKDalbuminpH(1/Ms)(1/s)(nM)Human7.49.26E+033.88E−0441.95.59.44E+032.70E−0428.6Rhesus7.46.16E+032.95E−034795.57.57E+032.72E−03359 SABA1.2 Binds to Domains I and II of HSA, but not Domain III The binding site SABA1.2 on albumin was mapped to the N-terminal domains I or II using recombinant HSA fragments and has no detectable binding to domain III (FIG.29). Because domain III is the domain of HSA that primarily interacts with FcRn, it is less likely that SABA1.2 would compete for HSA binding to FcRn, again increasing the possibility of fully leveraging the recycling mechanism for enhanced half-life. Example 5G. In Vivo Pharmacology of SABA1.2 A four week single dose pre-toxicology study of SABA1.2 was conducted in cynomolgus monkeys to assess pharmacokinetics and immunogenicity at two different dose levels. The pharmacokinetics and immunogenicity were also evaluated in a three-week, single-dose pre-toxicology study that included both intravenous and subcutaneous administration arms. Additionally, the pharmacokinetics of SABA1.2 was evaluated in two separate, single dose pre-toxicology studies in cynomolgus monkeys using a quantitative ELISA-based assay that was developed to detect SABA1.2 in plasma samples. SABA1.2 was administered to monkeys at 1 mpk and 10 mpk IV. Non-compartmental analyses using WINNONLIN® software were performed to evaluate pharmacokinetic parameters. As shown inFIG.30and the parameters described below, SABA1.2 exhibited dose-dependent pharmacokinetics in this study as determined by area under the concentration-time curve (AUC) evaluation. The clearance (CL) for SABA1.2 at 10 mpk was 0.15 ml/hr/kg, the beta phase half-life (t1/2) was 143 hours, the volume of distribution (Vz) was 30 mL/kg, and total drug exposure (AUCall) was 5,609,457 hr*nmol/L (Table 20). The clearance (CL) for SABA1.2 at 1 mpk was 0.4 ml/hr/kg, the half-life (t1/2) was 124 hours, the volume of distribution (Vz) was 72 mL/kg, and total drug exposure (AUCall) was 214,636 hr*nmol/L (Table 20). After SC or IV administration of SABA1.2, the beta-phase pharmacokinetic profiles were similar (FIG.31). The clearance (CL) for SABA1.2 at 1 mpk IV was 0.22 ml/hr/kg, the beta phase half-life (t1/2) was 125 hours, the volume of distribution (Vz) was 40 mL/kg, and total drug exposure (AUCall) was 357,993 hr*nmol/L (Table 20). The clearance (CL) for SABA1.2 at 1 mpk SC was 0.32 ml/hr/kg, the beta phase half-life (t1/2) was 134 hours, the volume of distribution (Vz) was 62 mL/kg, and total drug exposure (AUCall) was 251,339 hr*nmol/L (Table 20). The SC relative bioavailability (F) compared to IV was 0.7. TABLE 20Pharmacokinetic Parameters for SABA1.2 in MonkeysStudy #12Dose (mg/kg)11011Route ofi.v.i.v.i.v.s.c.administrationN3312CL (mL/hr/kg)0.40.150.220.32Vz (mL/kg)72304062AUCall214,6365,609,457357,993251,339(hr*nmol/L)beta T1/2(h)124143125134Bioavailabilityn/an/an/a0.7(F)
294,348
11858980
DETAILED DESCRIPTION Disclosed herein are polypeptides (e.g., antibody molecules or fusion proteins) that bind to a target molecule or cell, e.g., a human protein or cell, with high affinity and specificity. Advantageously, several of the polypeptides (e.g., antibody molecules or fusion proteins) describe herein have one or more improved or desired pharmacokinetic properties, such as circulating half-life. Without wishing to be bound theory, it is believed that polypeptides can have a range of circulating half-lives in humans, and circulating half-life can affect, e.g., interaction with serum and cell components, rate of fluid phase pinocytosis, interaction with FcRn, receptor mediated endocytosis, drug doses, and generation of anti-drug antibodies. Nucleic acid molecules encoding the polypeptides (e.g., antibody molecules or fusion proteins), expression vectors, host cells, compositions (e.g., pharmaceutical compositions), kits, containers, and methods for making the polypeptides (e.g., antibody molecules or fusion proteins), are also provided. The polypeptides (e.g., antibody molecules or fusion proteins) and pharmaceutical compositions disclosed herein can be used (alone or in combination with other agents or therapeutic modalities) to treat, prevent, and/or diagnose disorders and conditions, e.g., disorders and conditions associated with a target molecule (e.g., protein) or cell, e.g., a disorder or condition described herein. Without wishing to be bound by theory, it is believed that in some embodiments, the long circulating half-life of IgGs is attributed to its ability to minimize its endosomal degradation by associating with the neonatal Fc receptor (FcRn). FcRn plays an important role in placental transfer of IgG molecules from mother to fetus and in serum IgG homeostasis (Leach et al.,J Immunol,1996. 157(8): 3317-22; Simister et al.,Eur J Immunol,1996. 26(7): 1527-31; Kristoffersen,APMISSuppl, 1996. 64: 5-36; Roopenian et al.,J Immunol,2003. 170(7): 3528-33; Junghans and Anderson,Proc Natl Acad Sci USA,1996. 93(11): 5512-6). The acidic environment of the early endosome can allow for binding of IgG and albumin to FcRn, which protects the IgG from undergoing degradation and helps trafficking the IgG back to the extracellular environment, where, upon exposure to physiological pH, the molecules are released back into circulation. This pathway can be largely responsible for the prolonged serum half-life of both IgG and albumin (Junghans and Anderson,Proc Natl Acad Sci USA,1996. 93(11): 5512-6; Chaudhury et al.,J Exp Med,2003. 197(3): 315-22). The Fc domain of an antibody is primarily responsible for binding to FcRn to facilitate antibody recycling. In some embodiments, antibodies with identical Fc regions can have different circulating half-lives, at least in part, because a number of factors such as thermal stability, interaction with serum and cell components, presence of anti-drug antibodies, high dose, receptor mediated endocytosis, and fluid phase pinocytosis, can promote antibody degradation and negatively influence its half-life. The interaction of IgG with FcRn can serve to protect the antibody from endosomal degradation and extend the half-life of the antibody. Modification of Fc can be used to promote FcRn interaction and therefore extend the half-life of antibodies (Ghetie et al.,Nat Biotechnol,1997. 15(7): p. 637-40; Dall'Acqua et al.,J Biol Chem,2006. 281(33): 23514-24; Hinton et al.,J Biol Chem,2004. 279(8): 6213-6; Vaccaro et al.,Nat Biotechnol,2005. 23(10): 1283-8; Zalevsky et al.,Nat Biotechnol,2010. 28(2): 157-9; Dall'Acqua et al.,J Immunol,2002. 169(9): 5171-80; Monnet et al.,MAbs,2014. 6(2): 422-36; Monnet et al.,Front Immunol,2015. 6: 39; Shields et al.,J Biol Chem,2001. 276(9): 6591-604; Robbie et al.,Antimicrob Agents Chemother,2013. 57(12): 6147-53). The Fc of IgG can also bind to various other receptors such as FcγRI, FcγRIIa, FcγRIIb, FcγRIII, C1q, and TRIM21, and these interactions mediate various effector functions such as antibody dependent cellular cytotoxicity (ADCC), complement dependent cytotoxicity (CDC), antibody dependent cellular phagocytosis (ADCP), and antibody dependent intracellular neutralization (ADIN). Traditional approaches used to identify the Fc variants have largely relied on random mutagenesis and display formats and often compromise certain important attributes of the antibody, be it effector functions or biophysical stability. Without wishing to be bound by theory, it is believed that in some embodiment, the engineering of Fc for FcRn binding or half-life extension as disclosed herein is performed in the context of the various effector functions mediated by Fc. For example, a structural and network based framework can be used to interrogate the interaction of Fc with FcRn at neutral and acidic pH. Using this framework, different pathways for improving FcRn binding, e.g., decreasing the koffof interaction, can be identified. The interaction networks of mutations can be mapped and mutations can be combined and assessed for binding to FcRn and other Fc receptors. For example, Fc variants that confer enhancement in half-life and retain and in some cases enhance effector functions such as ADCC and CDC can be identified. With the increasing use of antibodies and fusion proteins as therapeutics for prevention and treatment of different diseases, there have been greater needs to develop antibodies and fusion proteins with long half-life, e.g., to treat or prevent chronic diseases. Definitions As used herein, the articles “a” and “an” refer to one or to more than one (e.g., to at least one) of the grammatical object of the article. The term “or” is used herein to mean, and is used interchangeably with, the term “and/or”, unless context clearly indicates otherwise. “About” and “approximately” shall generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, and more typically, within 5% of a given value or range of values. The compositions and methods disclosed herein encompass polypeptides and nucleic acids having the sequences specified, or sequences substantially identical or similar thereto, e.g., sequences at least 85%, 90%, 95% identical or higher to the sequence specified. In the context of an amino acid sequence, the term “substantially identical” is used herein to refer to a first amino acid that contains a sufficient or minimum number of amino acid residues that are i) identical to, or ii) conservative substitutions of aligned amino acid residues in a second amino acid sequence such that the first and second amino acid sequences can have a common structural domain and/or common functional activity. For example, amino acid sequences that contain a common structural domain having at least about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to a reference sequence, e.g., a sequence provided herein. In the context of nucleotide sequence, the term “substantially identical” is used herein to refer to a first nucleic acid sequence that contains a sufficient or minimum number of nucleotides that are identical to aligned nucleotides in a second nucleic acid sequence such that the first and second nucleotide sequences encode a polypeptide having common functional activity, or encode a common structural polypeptide domain or a common functional polypeptide activity. For example, nucleotide sequences having at least about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to a reference sequence, e.g., a sequence provided herein. The term “functional variant” refers polypeptides that have a substantially identical amino acid sequence to the naturally-occurring sequence, or are encoded by a substantially identical nucleotide sequence, and are capable of having one or more activities of the naturally-occurring sequence. Calculations of homology or sequence identity between sequences (the terms are used interchangeably herein) are performed as follows. To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). In a typical embodiment, the length of a reference sequence aligned for comparison purposes is at least 30%, e.g., at least 40%, 50%, 60%, e.g., at least 70%, 80%, 90%, 100% of the length of the reference sequence. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. In some embodiments, the percent identity between two amino acid sequences is determined using the Needleman and Wunsch ((1970)J. Mol. Biol.48:444-453) algorithm which has been incorporated into the GAP program in the GCG software package (available at www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. In certain embodiments, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6. One suitable set of parameters (and the one that should be used unless otherwise specified) are a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5. The percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of E. Meyers and W. Miller ((1989) CABIOS, 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The nucleic acid and protein sequences described herein can be used as a “query sequence” to perform a search against public databases to, for example, identify other family members or related sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990)J. Mol. Biol.215:403-10. BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to a nucleic acid as described herein. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to protein molecules described herein. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997)Nucleic Acids Res.25:3389-3402. When utilizing BLAST and gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See www.ncbi.nlm.nih.gov. As used herein, the term “hybridizes under low stringency, medium stringency, high stringency, or very high stringency conditions” describes conditions for hybridization and washing. Guidance for performing hybridization reactions can be found inCurrent Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is incorporated by reference. Aqueous and nonaqueous methods are described in that reference and either can be used. Specific hybridization conditions referred to herein are as follows: 1) low stringency hybridization conditions in 6× sodium chloride/sodium citrate (SSC) at about 45° C., followed by two washes in 0.2×SSC, 0.1% SDS at least at 50° C. (the temperature of the washes can be increased to 55° C. for low stringency conditions); 2) medium stringency hybridization conditions in 6×SSC at about 45° C., followed by one or more washes in 0.2×SSC, 0.1% SDS at 60° C.; 3) high stringency hybridization conditions in 6×SSC at about 45° C., followed by one or more washes in 0.2×SSC, 0.1% SDS at 65° C.; and preferably 4) very high stringency hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65° C., followed by one or more washes at 0.2×SSC, 1% SDS at 65° C. Very high stringency conditions 4) are suitable conditions and the ones that should be used unless otherwise specified. It is understood that the molecules described herein may have additional conservative or non-essential amino acid substitutions, which do not have a substantial effect on their functions. The term “amino acid” is intended to embrace all molecules, whether natural or synthetic, which include both an amino functionality and an acid functionality and capable of being included in a polymer of naturally-occurring amino acids. Exemplary amino acids include naturally-occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and all stereoisomers of any of any of the foregoing. As used herein the term “amino acid” includes both the D- or L-optical isomers and peptidomimetics. A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). The terms “polypeptide,” “peptide” and “protein” (if single chain) are used interchangeably herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component. The polypeptide can be isolated from natural sources, can be a produced by recombinant techniques from a eukaryotic or prokaryotic host, or can be a product of synthetic procedures. In an embodiment, the polypeptide is an antibody molecule. In another embodiment, the polypeptide is a fusion protein. The terms “nucleic acid,” “nucleic acid sequence,” “nucleotide sequence,” or “polynucleotide sequence,” and “polynucleotide” are used interchangeably. They refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. The polynucleotide may be either single-stranded or double-stranded, and if single-stranded may be the coding strand or non-coding (antisense) strand. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. The nucleic acid may be a recombinant polynucleotide, or a polynucleotide of genomic, cDNA, semisynthetic, or synthetic origin which either does not occur in nature or is linked to another polynucleotide in a non-natural arrangement. The term “isolated,” as used herein, refers to material that is removed from its original or native environment (e.g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated by human intervention from some or all of the co-existing materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of the environment in which it is found in nature. As used herein, the term “treat,” e.g., a disorder described herein, means that a subject (e.g., a human) who has a disorder, e.g., a disorder described herein, and/or experiences a symptom of a disorder, e.g., a disorder described herein, will, in an embodiment, suffer less a severe symptom and/or recover faster when an antibody molecule is administered than if the antibody molecule were never administered. Treatment can, e.g., partially or completely, alleviate, ameliorate, relieve, inhibit, or reduce the severity of, and/or reduce incidence, and optionally, delay onset of, one or more manifestations of the effects or symptoms, features, and/or causes of the disorder. In an embodiment, treatment is of a subject who does not exhibit certain signs of the disorder, and/or of a subject who exhibits only early signs of the disorder. In an embodiment, treatment is of a subject who exhibits one or more established signs of a disorder. In an embodiment, treatment is of a subject diagnosed as suffering from a disorder. As used herein, the term “prevent,” a disorder, means that a subject (e.g., a human) is less likely to have the disorder, if the subject receives a polypeptide (e.g., antibody molecule). Various aspects of the compositions and methods herein are described in further detail below. Additional definitions are set out throughout the specification. Antibody Molecules Disclosed herein are antibody molecules, e.g., antibody molecules comprising an Fc region, e.g. an Fc region having one or more mutations described herein, and/or having one or more structural or functional properties described herein. In an embodiment, the antibody molecule is engineered or derived from an antibody molecule (e.g., a parental antibody molecule) that contains an Fc region. For example, the engineered antibody molecule, or antibody molecule derivative, can have a different Fc region than the parental antibody molecule. In another embodiment, the antibody molecule is engineered or derived from an antibody molecule (e.g., a parental antibody molecule) that does not contain an Fc region. For example, the engineered antibody molecule, or antibody molecule derivative, can have an Fc region, directly or indirectly, fused to the parental antibody molecule or a functional fragment thereof. As used herein, the term “antibody molecule” refers to a protein, e.g., an immunoglobulin chain or a fragment thereof, comprising at least one immunoglobulin variable domain sequence. The term “antibody molecule” includes, for example, full-length, mature antibodies and antigen-binding fragments of an antibody. For example, an antibody molecule can include a heavy (H) chain variable domain sequence (abbreviated herein as VH), and a light (L) chain variable domain sequence (abbreviated herein as VL). In another example, an antibody molecule includes two heavy (H) chain variable domain sequences and two light (L) chain variable domain sequence, thereby forming two antigen binding sites, such as Fab, Fab′, F(ab′)2, Fc, Fd, Fd′, Fv, single chain antibodies (scFv for example), single variable domain antibodies, diabodies (Dab) (bivalent and bispecific), and chimeric (e.g., humanized) antibodies, which may be produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA technologies. These functional antibody fragments retain the ability to selectively bind with their respective antigen or receptor. Antibodies and antibody fragments can be from any class of antibodies including, but not limited to, IgG, IgA, IgM, IgD, and IgE, and from any subclass (e.g., IgG1, IgG2, IgG3, and IgG4) of antibodies. The antibody molecules can be monoclonal or polyclonal. The antibody molecule can also be a human, humanized, CDR-grafted, or in vitro generated antibody. The antibody molecule can have a heavy chain constant region chosen from, e.g., IgG1, IgG2, IgG3, or IgG4. The antibody molecule can also have a light chain chosen from, e.g., kappa or lambda. The term “immunoglobulin” (Ig) is used interchangeably with the term “antibody” herein. Examples of antigen-binding fragments include: (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a diabody (dAb) fragment, which consists of a VH domain; (vi) a camelid or camelized variable domain; (vii) a single chain Fv (scFv), see e.g., Bird et al. (1988)Science242:423-426; and Huston et al. (1988)Proc. Natl. Acad. Sci. USA85:5879-5883); (viii) a single domain antibody. These antibody fragments may be obtained using any suitable method, including several conventional techniques known to those with skill in the art, and the fragments can be screened for utility in the same manner as are intact antibodies. The term “antibody” includes intact molecules as well as functional fragments thereof. Constant regions of the antibodies can be altered, e.g., mutated, to modify the properties of the antibody (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function). The antibody molecule can be a single chain antibody. A single-chain antibody (scFV) may be engineered (see, for example, Colcher, D. et al. (1999)Ann N Y Acad Sci880:263-80; and Reiter, Y. (1996)Clin Cancer Res2:245-52). The single chain antibody can be dimerized or multimerized to generate multivalent antibodies having specificities for different epitopes of the same target protein. The antibody molecules disclosed herein can also be single domain antibodies. Single domain antibodies can include antibodies whose complementary determining regions are part of a single domain polypeptide. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies. Single domain antibodies may be any of the art, or any future single domain antibodies. Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, llama, fish, shark, goat, rabbit, and bovine. According to some aspects, a single domain antibody is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains. Such single domain antibodies are disclosed in WO 94/04678, for example. For clarity reasons, this variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins. Such a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, llama, dromedary, alpaca and guanaco. Other species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are also contemplated. The VH and VL regions can be subdivided into regions of hypervariability, termed “complementarity determining regions” (CDR), interspersed with regions that are more conserved, termed “framework regions” (FR or FW). The terms “complementarity determining region,” and “CDR,” as used herein refer to the sequences of amino acids within antibody variable regions which confer antigen specificity and binding affinity. As used herein, the terms “framework,” “FW” and “FR” are used interchangeably. The extent of the framework region and CDRs has been precisely defined by a number of methods (see, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; Chothia, C. et al. (1987)J. Mol. Biol.196:901-917; and the AbM definition used by Oxford Molecular's AbM antibody modeling software. See, generally, e.g., Protein Sequence and Structure Analysis of Antibody Variable Domains. In: Antibody Engineering Lab Manual (Ed.: Duebel, S. and Kontermann, R., Springer-Verlag, Heidelberg). In an embodiment, the following definitions are used: AbM definition of CDR1 of the heavy chain variable domain and Kabat definitions for the other CDRs. In an embodiment, Kabat definitions are used for all CDRs. In addition, embodiments described with respect to Kabat or AbM CDRs may also be implemented using Chothia hypervariable loops. Each VH and VL typically includes three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. As used herein, an “immunoglobulin variable domain sequence” refers to an amino acid sequence which can form the structure of an immunoglobulin variable domain. For example, the sequence may include all or part of the amino acid sequence of a naturally-occurring variable domain. For example, the sequence may or may not include one, two, or more N- or C-terminal amino acids, or may include other alterations that are compatible with formation of the protein structure. The term “antigen-binding region” refers to the part of an antibody molecule that comprises determinants that form an interface that binds to an antigen, or an epitope thereof. With respect to proteins (or protein mimetics), the antigen-binding region typically includes one or more loops (of at least, e.g., four amino acids or amino acid mimics) that form an interface that binds to the antigen. Typically, the antigen-binding region of an antibody molecule includes at least one or two CDRs and/or hypervariable loops, or more typically at least three, four, five or six CDRs and/or hypervariable loops. The terms “compete” or “cross-compete” are used interchangeably herein to refer to the ability of an antibody molecule to interfere with binding of another antibody molecule, to a target. The interference with binding can be direct or indirect (e.g., through an allosteric modulation of the antibody molecule or the target). The extent to which an antibody molecule is able to interfere with the binding of another antibody molecule to the target, and therefore whether it can be said to compete, can be determined using a competition binding assay, for example, a FACS assay, an ELISA or BIACORE assay. In an embodiment, a competition binding assay is a quantitative competition assay. In an embodiment, a first antibody molecule is said to compete for binding to the target with a second antibody molecule when the binding of the first antibody molecule to the target is reduced by 10% or more, e.g., 20% or more, 30% or more, 40% or more, 50% or more, 55% or more, 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or more, 99% or more in a competition binding assay (e.g., a competition assay described herein). The terms “monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. A monoclonal antibody can be made by hybridoma technology or by methods that do not use hybridoma technology (e.g., recombinant methods). An “effectively human” protein is a protein that does not evoke a neutralizing antibody response, e.g., the human anti-murine antibody (HAMA) response. HAMA can be problematic in a number of circumstances, e.g., if the antibody molecule is administered repeatedly, e.g., in treatment of a chronic or recurrent disease condition. A HAMA response can make repeated antibody administration potentially ineffective because of an increased antibody clearance from the serum (see, e.g., Saleh et al.,Cancer Immunol. Immunother.32:180-190 (1990)) and also because of potential allergic reactions (see, e.g., LoBuglio et al.,Hybridoma,5:5117-5123 (1986)). The antibody molecule can be a polyclonal or a monoclonal antibody. In some embodiments, the antibody can be recombinantly produced, e.g., produced by any suitable phage display or combinatorial methods. Various phage display and combinatorial methods for generating antibodies are known in the art (as described in, e.g., Ladner et al. U.S. Pat. No. 5,223,409; Kang et al. International Publication No. WO 92/18619; Dower et al. International Publication No. WO 91/17271; Winter et al. International Publication WO 92/20791; Markland et al. International Publication No. WO 92/15679; Breitling et al. International Publication WO 93/01288; McCafferty et al. International Publication No. WO 92/01047; Garrard et al. International Publication No. WO 92/09690; Ladner et al. International Publication No. WO 90/02809; Fuchs et al. (1991)Bio/Technology9:1370-1372; Hay et al. (1992)Hum Antibod Hybridomas3:81-85; Huse et al. (1989)Science246:1275-1281; Griffths et al. (1993)EMBO J12:725-734; Hawkins et al. (1992)J Mol Biol226:889-896; Clackson et al. (1991)Nature352:624-628; Gram et al. (1992)PNAS89:3576-3580; Garrad et al. (1991)Bio/Technology9:1373-1377; Hoogenboom et al. (1991)Nuc Acid Res19:4133-4137; and Barbas et al. (1991)PNAS88:7978-7982, the contents of all of which are incorporated by reference herein). In an embodiment, the antibody molecule is a fully human antibody (e.g., an antibody made in a mouse which has been genetically engineered to produce an antibody from a human immunoglobulin sequence), or a non-human antibody, e.g., a rodent (mouse or rat), goat, primate (e.g., monkey), camel antibody. In an embodiment, the non-human antibody is a rodent (mouse or rat antibody). Methods of producing rodent antibodies are known in the art. Human monoclonal antibodies can be generated using transgenic mice carrying the human immunoglobulin genes rather than the mouse system. Splenocytes from these transgenic mice immunized with the antigen of interest are used to produce hybridomas that secrete human mAbs with specific affinities for epitopes from a human protein (see e.g., Wood et al. International Application WO 91/00906, Kucherlapati et al. PCT publication WO 91/10741; Lonberg et al. International Application WO 92/03918; Kay et al. International Application 92/03917; Lonberg et al. 1994Nature368:856-859; Green, L. L. et al. 1994Nature Genet.7:13-21; Morrison, S. L. et al. 1994Proc. Natl. Acad. Sci. USA81:6851-6855; Bruggeman et al. 1993Year Immunol7:33-40; Tuaillon et al. 1993PNAS90:3720-3724; Bruggeman et al. 1991Eur J Immunol21:1323-1326). An antibody can be one in which the variable region, or a portion thereof, e.g., the CDRs, are generated in a non-human organism, e.g., a rat or mouse. Chimeric, CDR-grafted, and humanized antibodies are within the invention. Antibodies generated in a non-human organism, e.g., a rat or mouse, and then modified, e.g., in the variable framework or constant region, to decrease antigenicity in a human are within the invention. Chimeric antibodies can be produced by any suitable recombinant DNA technique. Several are known in the art (see Robinson et al., International Patent Publication PCT/US86/02269; Akira, et al., European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al., European Patent Application 173,494; Neuberger et al., International Application WO 86/01533; Cabilly et al. U.S. Pat. No. 4,816,567; Cabilly et al., European Patent Application 125,023; Better et al. (1988Science240:1041-1043); Liu et al. (1987)PNAS84:3439-3443; Liu et al., 1987, J. Immunol.139:3521-3526; Sun et al. (1987)PNAS84:214-218; Nishimura et al., 1987, Canc. Res.47:999-1005; Wood et al. (1985)Nature314:446-449; and Shaw et al., 1988, J. Natl Cancer Inst.80:1553-1559). A humanized or CDR-grafted antibody will have at least one or two but generally all three recipient CDRs (of heavy and or light immunoglobulin chains) replaced with a donor CDR. The antibody may be replaced with at least a portion of a non-human CDR or only some of the CDRs may be replaced with non-human CDRs. It is only necessary to replace the number of CDRs required for binding of the humanized antibody to lipopolysaccharide. In an embodiment, the donor will be a rodent antibody, e.g., a rat or mouse antibody, and the recipient will be a human framework or a human consensus framework. Typically, the immunoglobulin providing the CDRs is called the “donor” and the immunoglobulin providing the framework is called the “acceptor.” In some embodiments, the donor immunoglobulin is a non-human (e.g., rodent). The acceptor framework is typically a naturally-occurring (e.g., a human) framework or a consensus framework, or a sequence about 85% or higher, e.g., 90%, 95%, 99% or higher identical thereto. As used herein, the term “consensus sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of proteins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence. A “consensus framework” refers to the framework region in the consensus immunoglobulin sequence. An antibody can be humanized by any suitable method, and several such methods known in the art (see e.g., Morrison, S. L., 1985, Science229:1202-1207, by Oi et al., 1986, BioTechniques4:214, and by Queen et al. U.S. Pat. Nos. 5,585,089, 5,693,761 and 5,693,762, the contents of all of which are hereby incorporated by reference). Humanized or CDR-grafted antibodies can be produced by CDR-grafting or CDR substitution, wherein one, two, or all CDRs of an immunoglobulin chain can be replaced. See e.g., U.S. Pat. No. 5,225,539; Jones et al. 1986Nature321:552-525; Verhoeyan et al. 1988Science239:1534; Beidler et al. 1988J. Immunol.141:4053-4060; Winter U.S. Pat. No. 5,225,539, the contents of all of which are hereby expressly incorporated by reference. Winter describes a CDR-grafting method which may be used to prepare humanized antibodies (UK Patent Application GB 2188638A, filed on Mar. 26, 1987; Winter U.S. Pat. No. 5,225,539), the contents of which is expressly incorporated by reference. Also provided are humanized antibodies in which specific amino acids have been substituted, deleted or added. Criteria for selecting amino acids from the donor are described in, e.g., U.S. Pat. No. 5,585,089, e.g., columns 12-16 of U.S. Pat. No. 5,585,089, the contents of which are hereby incorporated by reference. Other techniques for humanizing antibodies are described in Padlan et al. EP 519596 A1, published on Dec. 23, 1992. In an embodiment, the antibody molecule has a heavy chain constant region chosen from, e.g., the heavy chain constant regions of IgG1, IgG2 (e.g., IgG2a), IgG3, IgG4, IgM, IgA1, IgA2, IgD, and IgE; particularly, chosen from, e.g., the (e.g., human) heavy chain constant regions of IgG1, IgG2, IgG3, and IgG4. In another embodiment, the antibody molecule has a light chain constant region chosen from, e.g., the (e.g., human) light chain constant regions of kappa or lambda. The constant region can be altered, e.g., mutated, to modify the properties of the antibody molecule (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, and/or complement function). In an embodiment, the antibody molecule has effector function and can fix complement. In another embodiment, the antibody molecule does not recruit effector cells or fix complement. In certain embodiments, the antibody molecule has reduced or no ability to bind an Fc receptor. For example, it may be an isotype or subtype, fragment or other mutant, which does not support binding to an Fc receptor, e.g., it has a mutagenized or deleted Fc receptor binding region. In an embodiment, a constant region of the antibody molecule is altered. Methods for altering an antibody constant region are known in the art. Antibody molecules s with altered function, e.g. altered affinity for an effector ligand, such as FcR on a cell, or the C1 component of complement can be produced by replacing at least one amino acid residue in the constant portion of the antibody with a different residue (see e.g., EP 388,151 A1, U.S. Pat. Nos. 5,624,821 and 5,648,260, the contents of all of which are hereby incorporated by reference) Amino acid mutations which stabilize antibody structure, such as S228P (EU nomenclature, S241P in Kabat nomenclature) in human IgG4 are also contemplated. Similar type of alterations could be described which if applied to the murine, or other species immunoglobulin would reduce or eliminate these functions. In an embodiment, the only amino acids in the antibody molecule are canonical amino acids. In an embodiment, the antibody molecule comprises naturally-occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and/or all stereoisomers of any of any of the foregoing. The antibody molecule may comprise the D- or L-optical isomers of amino acids and peptidomimetics. A polypeptide of an antibody molecule described herein may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The antibody molecule may also be modified; for example, by disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component. The polypeptide can be isolated from natural sources, can be a produced by recombinant techniques from a eukaryotic or prokaryotic host, or can be a product of synthetic procedures. The antibody molecule described herein can be used alone in unconjugated form, or can be bound to a substance, e.g., a toxin or moiety (e.g., a therapeutic drug; a compound emitting radiation; molecules of plant, fungal, or bacterial origin; or a biological protein (e.g., a protein toxin) or particle (e.g., a recombinant viral particle, e.g., via a viral coat protein). For example, the antibody molecule can be coupled to a radioactive isotope such as an α-, β-, or γ-emitter, or a β- and γ-emitter. An antibody molecule can be derivatized or linked to another functional molecule (e.g., another peptide or protein). As used herein, a “derivatized” antibody molecule is one that has been modified. Methods of derivatization include but are not limited to the addition of a fluorescent moiety, a radionucleotide, a toxin, an enzyme or an affinity ligand such as biotin. Accordingly, the antibody molecules are intended to include derivatized and otherwise modified forms of the antibodies described herein, including immunoadhesion molecules. For example, an antibody molecule can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a toxin, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag). Some types of derivatized antibody molecule are produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies). Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate). Such linkers are available from Pierce Chemical Company, Rockford, Ill. Useful detectable agents with which an anti-dengue antibody molecule may be derivatized (or labeled) to include fluorescent compounds, various enzymes, prosthetic groups, luminescent materials, bioluminescent materials, fluorescent emitting metal atoms, e.g., europium (Eu), and other anthanides, and radioactive materials (described below). Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like. An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, β-galactosidase, acetylcholinesterase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody molecule may also be derivatized with a prosthetic group (e.g., streptavidin/biotin and avidin/biotin). For example, an antibody may be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding. Examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of bioluminescent materials include luciferase, luciferin, and aequorin. Labeled antibody molecules can be used, for example, diagnostically and/or experimentally in a number of contexts, including (i) to isolate a predetermined antigen by standard techniques, such as affinity chromatography or immunoprecipitation; (ii) to detect a predetermined antigen (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the protein; (iii) to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to determine the efficacy of a given treatment regimen. An antibody molecule may be conjugated to another molecular entity, typically a label or a therapeutic (e.g., antimicrobial (e.g., antibacterial or bactericidal), immunomodulatory, immunostimularoty, cytotoxic, or cytostatic) agent or moiety. Radioactive isotopes can be used in diagnostic or therapeutic applications. Radioactive isotopes that can be coupled to the antibody molecules include, but are not limited to α-, β-, or γ-emitters, or β- and γ-emitters. Such radioactive isotopes include, but are not limited to iodine (131I or125I), yttrium (90Y), lutetium (177Lu), actinium (225Ac), praseodymium, astatine (211At), rhenium (186Re), bismuth (212Bi or213Bi), indium (111In), technetium (99mTc), phosphorus (32P), rhodium (188Rh), sulfur (35S), carbon (14C), tritium (3H), chromium (51Cr), chlorine (36Cl), cobalt (57Co or58Co), iron (59Fe), selenium (75Se), or gallium (67Ga). Radioisotopes useful as therapeutic agents include yttrium (90Y), lutetium (177Lu), actinium (225Ac), praseodymium, astatine (211At), rhenium (186Re), bismuth (212Bi or213Bi), and rhodium (188Rh). Radioisotopes useful as labels, e.g., for use in diagnostics, include iodine (131I or125I), indium (111In), technetium (99mTc), phosphorus (32P), carbon (14C), and tritium (3H), or one or more of the therapeutic isotopes listed above. The present disclosure provides radiolabeled antibody molecules and methods of labeling the same. In an embodiment, a method of labeling an antibody molecule is disclosed. The method includes contacting an antibody molecule, with a chelating agent, to thereby produce a conjugated antibody. The conjugated antibody is radiolabeled with a radioisotope, e.g.111Indium,90Yttrium and177Lutetium, to thereby produce a labeled antibody molecule. In some aspects, this disclosure provides a method of making an antibody molecule disclosed herein. The method includes: providing an antigen, or a fragment thereof; obtaining an antibody molecule that specifically binds to the antigen; evaluating efficacy of the antibody molecule in modulating activity of the antigen and/or organism expressing the antigen. The method can further include administering the antibody molecule, including a derivative thereof (e.g., a humanized antibody molecule) to a subject, e.g., a human. This disclosure provides an isolated nucleic acid molecule encoding the above antibody molecule, vectors and host cells thereof. The nucleic acid molecule includes, but is not limited to, RNA, genomic DNA and cDNA. Fusion Proteins Disclosed herein are fusion proteins, e.g., fusion proteins comprising an Fc region, e.g. an Fc region having one or more mutations described herein, and/or having one or more structural or functional properties described herein. In an embodiment, the fusion protein is engineered or derived from a polypeptide (e.g., a fusion protein) that contains an Fc region (e.g., a parental polypeptide). For example, the engineered polypeptide, or derivative, can have a different Fc region than the parental polypeptide. In another embodiment, the fusion protein is engineered or derived from a polypeptide that does not contain an Fc region (e.g., a parental polypeptide). For example, the engineered antibody molecule, or antibody molecule derivative, can have an Fc region, directly or indirectly, fused to the parental polypeptide. As used herein, the term “fusion protein” refers to a protein, comprising two or more protein or peptide components. The two or more protein or peptide components can be obtained from different sources or encoded by different genes. A fusion protein is sometimes also referred to as a chimeric protein. An Fc-fusion protein (also known as Fc chimeric fusion protein, Fc-Ig, Ig-based chimeric fusion protein, or Fc-tag protein) can include an Fc region of an immunoglobulin (e.g., an Fc region described herein) linked (e.g., fused) to a protein or peptide. The Fc region can be linked (e.g., fused genetically) to the protein or peptide directly, or indirectly, e.g., through a linker In an embodiment, the Fc region is derived from the Fc region of IgG, e.g., human IgG, e.g., IgG1, IgG2, IgG3, or IgG4. In an embodiment, the Fc region is derived from the Fc region of IgG1, e.g., human IgG1. The Fc-fused binding partner can include a variety of proteins or peptides, or fragments thereof. For example, the Fc region can be fused to a peptide (e.g., a therapeutic peptide), a ligand (e.g., a ligand that activates upon binding with a cell surface receptor), a signaling molecule, the extracellular domain of a receptor, or a bait protein (e.g., used to identify a binding partner, e.g., in a protein microarray). In an embodiment, the Fc fusion protein comprises an extracellular domain of a receptor or a soluble receptor, or a ligand binding portion thereof. In an embodiment, the receptor is a growth factor receptor. In an embodiment, the receptor is a cytokine receptor. In an embodiment, the receptor is an immune checkpoint molecule. In an embodiment, the fusion protein comprises a vascular endothelia growth factor (VEGF)-binding portion from the extracellular domain of human VEGF receptors 1 and 2, fused to the Fc region of human IgG1. In an embodiment, the fusion protein is aflibercept. In an embodiment, the fusion protein (e.g., aflibercept) is used to treat a disorder described herein, e.g., an eye disorder (e.g., wet macular degeneration) or a cancer (e.g., a colorectal cancer). In another embodiment, the fusion protein comprises a soluble tumor necrosis factor (TNF) receptor 2 fused to the Fc region of the human IgG1. In an embodiment, the fusion protein is etanercept. In an embodiment, the Fc fusion protein (e.g., etanercept) is used to treat a disorder described herein, e.g., an autoimmune disorder (e.g., rheumatoid arthritis). In yet another embodiment, the fusion protein comprises ligand-binding domains of the extracellular portions of human interleukin-1 receptor 1 (IL-1R1) and IL-1 receptor accessory protein (IL-1RAcP) fused to the Fc region of human IgG1. In an embodiment, the fusion protein is rilonacept. In an embodiment, the fusion protein (e.g., rilonacept) is used to treat a disorder described herein, e.g., a cryopyrin-associated periodic syndrome (CAPS), e.g., familial cold autoinflammatory syndrome, Muckle-Wells syndrome, or a neonatal onset multisystem inflammatory disease. In still another embodiment, the fusion protein comprises the extracellular domain of CTLA-4 fused to the Fc region of human IgG1. In an embodiment, the fusion protein is abatacept or belatacept. In an embodiment, the fusion protein (e.g., abatacept or belatacept) is used to treat a disorder described herein, e.g., an organ rejection, an autoimmune disorder (e.g., rheumatoid arthritis), or a cancer. In an embodiment, the Fc fusion protein comprises a peptide, e.g., a therapeutic peptide. In an embodiment, the fusion protein comprises a thrombopoietin-binding peptide fused to the Fc region of human IgG1. In an embodiment, the fusion protein is romiplostim. In an embodiment, the fusion protein (e.g., romiplostim) is used to treat a disorder described herein, e.g., chronic idiopathic (immune) thrombocytopenic purpura (ITP). In an embodiment, the fusion protein comprises the extracellular CD2-binding portion of the human leukocyte function antigen-3 (LFA-3) fused to the Fc region of human IgG1. In an embodiment, the fusion protein is alefacept. In an embodiment, the fusion protein (e.g., alefacept) is used to treat a disorder described herein, e.g., an autoimmune disorder (e.g., psoriasis) or a cancer (e.g., a cutaneous T-cell lymphoma or a T-cell non-Hodgkin lymphoma). In an embodiment, the fusion protein comprises a coagulation factor. In an embodiment, the fusion protein comprises Factor IX fused with the Fc region of IgG1. In another embodiment, the fusion protein comprises Factor VIII fused with the Fc region of IgG1. In an embodiment the fusion protein (e.g., the FIX-Fc fusion or FVIII-Fc fusion) is used to treat a disorder described herein, e.g., hemophilia A or hemophilia B. In an embodiment, the fusion protein comprises one or more glycosylation sites, or is glycosylated. In another embodiment, the fusion protein does not have a glycosylation site, or is not glycosylated. In an embodiment, the only amino acids in the fusion protein are canonical amino acids. In an embodiment, the fusion protein comprises naturally-occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and/or all stereoisomers of any of any of the foregoing. The fusion protein may comprise the D- or L-optical isomers of amino acids and peptidomimetics. In an aspect, this disclosure provides a method of making a fusion protein disclosed herein. The fusion proteins described herein can be produced by any suitable recombinant DNA technique. In an embodiment, the method includes culturing a cell containing a nucleic acid encoding the fusion protein under conditions that allow production of the fusion protein. In another embodiment, the method further includes isolating or purifying the fusion protein. In yet another embodiment, the method further includes evaluating efficacy of the fusion protein in a cell-based assay or in an animal model. In still another embodiment, the method further includes administering the fusion protein to a subject, e.g., a human. This disclosure provides an isolated nucleic acid molecule encoding the above fusion proteins, vectors and host cells thereof. The nucleic acid molecule includes, but is not limited to, RNA, genomic DNA and cDNA. Fc Region A fragment crystallizable region, or Fc region, refers to a region of an immunoglobulin that is capable of interacting with an Fc receptor. In an embodiment, the Fc region is also capable of interacting with a protein of the complement system. While without wishing to be bound by theory, it is believed that in an embodiment, the interaction between the Fc region with an Fc receptor, allows for activation of the immune system. In IgG, IgA and IgD antibody isotypes, the naturally-occurring Fc region generally comprises two identical protein fragments, derived from the second and third constant domains of the antibody's two heavy chains. Naturally-occurring IgM and IgE Fc regions generally comprise three heavy chain constant domains (CHdomains 2-4) in each polypeptide chain. The Fc regions of IgGs can contain a highly conserved N-glycosylation site (Stadlmann et al. (2008).Proteomics8 (14): 2858-2871; Stadlmann (2009)Proteomics9 (17): 4143-4153). While not wishing to be bound by theory, it is believed that in an embodiment, glycosylation of the Fc fragment contributes to Fc receptor-mediated activities (Peipp et al. (2008)Blood112 (6): 2390-2399). In an embodiment, the N-glycans attached to this site are predominantly core-fucosylated diantennary structures of the complex type. In another embodiment, small amounts of these N-glycans also contain bisecting GlcNAc and/or α-2,6 linked sialic acid residues. An exemplary Fc region amino acid sequence is shown below. (SEQ ID NO: 1)ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK In SEQ ID NO: 1, the first amino acid residue in this sequence is referred to as position 118 herein. The three Histidines in bold and underlined are positions 310, 433 and 435, respectively. A polypeptide (e.g., an antibody molecule or fusion protein) described herein can have one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more) of mutations or combinations of mutations described in Table 1. TABLE 1Exemplary Fc mutationsNameMutationFcMut001I253MFcMut002L309H_D312A_N315DFcMut003L309NFcMut004M252E_S254RFcMut005M252E_S254R_R255YFcMut006S254HFcMut007S254MFcMut008T256D_T307RFcMut009T256L_N286I_T307IFcMut010T256I_N286I_T307IFcMut011K248S_D376QFcMut012K248S_D376NFcMut013D376Q_E380AFcMut014D376N_E380AFcMut015D376Q_M428LFcMut016K248S_A378IFcMut017L314KFcMut018T250Q_M428LFcMut019M428L_N434AFcMut020N434AFcMut021T307A_E380A_N434AFcMut022M252WFcMut023V308FFcMut024V308F_N434YFcMut026T256D_T307R_D376NFcMut027L309R_D312EFcMut028L309R_Q311P_D312EFcMut029K246N_P247AFcMut030K246N_P247A_D376NFcMut031T256E_T307RFcMut032T256R_T307DFcMut033T256R_T307EFcMut034Q311PFcMut035D376QFcMut036L234A_L235AFcMut037L235V_G236AFcMut038L234P_L235PFcMut039L235PFcMut040P329GFcMut041P329EFcMut042E233KFcMut043T256D_N286D_A287S_T307RFcMut044T256D_P257L_T307RFcMut045T256D_T307R_Q311VFcMut046P247D_T256D_T307RFcMut047P247D_N286D_A287S_Q311VFcMut048P257M_V308NFcMut049V279I_Q311L_N315TFcMut050M428L_N434SFcMut051N434SFcMut052H433G_N434PFcMut053V259I_V308F_M428LFcMut067T256D_N286D_T307RFcMut068T256D_N286E_T307RFcMut069T256D_N286Q_T307RFcMut070T256D_P257T_T307RFcMut071T256D_P257V_T307RFcMut072T256D_T307R_Q311IFcMut073T256D_T307R_Q311LFcMut074T256D_T307R_Q311MFcMut075T256D_P257L_N286D_T307R_Q311VFcMut076T256D_T307R_M428LFcMut077M428LFcMut078M252Y_S254T_T256QFcMut079M252Y_S254T_T256E_K288EFcMut080T256K_K288EFcMut081T256D_E258TFcMut082E283Q_H285EFcMut083R344D_D401RFcMut084K248E_E380KFcMut085K248E_E380RFcMut086K246HFcMut087K248HFcMut088T250IFcMut089T250VFcMut090L251FFcMut091L251MFcMut093P257VFcMut094N276DFcMut095H285NFcMut096H285DFcMut097K288HFcMut098K288QFcMut099K288EFcMut100T307EFcMut101T307QFcMut102V308PFcMut103V308IFcMut104V308LFcMut105L309HFcMut106L309MFcMut107Q311HFcMut108L314FFcMut109Y319HFcMut110I336TFcMut111P343DFcMut112P343VFcMut113E345QFcMut114P346VFcMut115P374TFcMut116D376NFcMut117A378SFcMut118A431TFcMut119A431PFcMut120A431GFcMut121L432VFcMut122L432IFcMut123L432QFcMut124N434TFcMut125H435NFcMut126Y436HFcMut127K439QFcMut128T256DFcMut129T307RFcMut130A378TFcMut131A378DFcMut132A378HFcMut133A378YFcMut134A378VFcMut135D376RFcMut136D376FFcMut137D376WFcMut138L314HFcMut139L432E_T437QFcMut140D376Q_A378TFcMut141D376Q_I377M_A378TFcMut142P244Q_D376QFcMut143P247T_A378TFcMut144P247N_A378TFcMut145T256D_T307R_L309TFcMut146A339T_S375E_F404YFcMut147L235V_G236A_T256D_T307RFcMut148L235V_G236A_D376Q_M428LFcMut149L314NFcMut150N315DFcMut151A378TFcMut152T437QFcMut153L432EFcMut154Y436RFcMut155L314MFcMut156L234A_L235A_T256D_T307R_Q311VFcMut157L234A_L235A_T256D_P257V_T307RFcMut158L234A_L235A_T256D_P257L_N286D_T307R_Q311VFcMut159L235V_G236A_T256D_T307R_Q311VFcMut160L235V_G236A_T256D_P257V_T307RFcMut161L235V_G236A_T256D_P257L_N286D_T307R_Q311VFcMut162S267T_A327N_A330MFcMut163S267T_A327NFcMut164L235V_G236A_S267T_A327N_A330MFcMut165L235V_G236A_S267T_A327NFcMut166M252Y_S254TFcMut167T256EFcMut168G236A_I332EFcMut169S239D_I332EFcMut170G236A_S239D_I332EFcMut171T256D_N286D_T307R_Q311VFcMut172T256D_E258T_T307RFcMut173T256D_E258T_T307R_Q311VFcMut174T256D_P257V_E258T_T307RFcMut175T256D_P257L_E258T_N286D_T307R_Q311VFcMut176T256D_E258T_N286D_T307R_Q311VFcMut177A378V_M428LFcMut178A378V_M428IFcMut179A378V_M428VFcMut180T256D_N286DFcMut181T256D_A378VFcMut182T256D_Q311VFcMut183T256D_Q311V_A378VFcMut184T256D_T307R_A378VFcMut185T256D_N286D_T307R_A378VFcMut186T256D_T307R_Q311V_A378VFcMut187H285D_A378VFcMut188H285D_Q311VFcMut189T256D_H285DFcMut190T256D_H285D_Q311VFcMut191T256D_H285D_T307RFcMut192T256D_H285D_T307R_A378VFcMut193H285D_L314M_A378VFcMut194T256D_E258T_H285D_Q311HFcMut195T256D_E258T_H285DFcMut196H285D_N315DFcMut197H285N_T307Q_N315DFcMut198H285D_L432E_T437QFcMut199T256D_E258T_N315DFcMut200P257V_H285NFcMut201H285N_L432FFcMut202H285N_T437IFcMut203T256D_E258T_L314MFcMut204T256D_E258T_T307QFcMut205T256D_E258T_A378VFcMut206V308P_A378VFcMut207P257V_A378TFcMut208P257V_V308P_A378VFcMut209N315D_A378TFcMut210H285N_L314MFcMut211L314M_L432E_T437QFcMut212T307Q_N315DFcMut213H285D_T307Q_A378VFcMut214L314M_N315DFcMut215T307Q_Q311V_A378VFcMut216H285D_Q311V_A378VFcMut217Q311V_N315D_A378VFcMut218T256D_E258T_Q311VFcMut219T256D_N315D_A378VFcMut220T256D_Q311V_N315DFcMut221T256D_T307Q_A378VFcMut222T256D_T307Q_Q311VFcMut223T256D_H285D_A378VFcMut224T256D_H285D_T307R_Q311VFcMut225T256D_H285D_N286D_T307RFcMut226T256D_H285D_N286D_T307R_Q311VFcMut227T256D_H285D_N286D_T307R_A378VFcMut228T256D_N286D_T307R_Q311V_A378VFcMut229T256D_H285D_T307R_Q311V_A378VFcMut230V308P_Q311V_A378VFcMut231T256D_V308P_A378VFcMut232T256D_V308P_Q311VFcMut233T256D_E258T_V308PFcMut234H285D_V308P_Q311VFcMut242E258TFcMut243N286DFcMut244Q311VYTEM252Y_S254T_T256E In an embodiment, the Fc region comprises FcMut001. In an embodiment, the Fc region comprises FcMut002. In an embodiment, the Fc region comprises FcMut003. In an embodiment, the Fc region comprises FcMut004. In an embodiment, the Fc region comprises FcMut005. In an embodiment, the Fc region comprises FcMut006. In an embodiment, the Fc region comprises FcMut007. In an embodiment, the Fc region comprises FcMut008. In an embodiment, the Fc region comprises FcMut009. In an embodiment, the Fc region comprises FcMut010. In an embodiment, the Fc region comprises FcMut011. In an embodiment, the Fc region comprises FcMut012. In an embodiment, the Fc region comprises FcMut013. In an embodiment, the Fc region comprises FcMut014. In an embodiment, the Fc region comprises FcMut015. In an embodiment, the Fc region comprises FcMut016. In an embodiment, the Fc region comprises FcMut017. In an embodiment, the Fc region comprises FcMut018. In an embodiment, the Fc region comprises FcMut019. In an embodiment, the Fc region comprises FcMut020. In an embodiment, the Fc region comprises FcMut021. In an embodiment, the Fc region comprises FcMut022. In an embodiment, the Fc region comprises FcMut023. In an embodiment, the Fc region comprises FcMut024. In an embodiment, the Fc region comprises FcMut026. In an embodiment, the Fc region comprises FcMut027. In an embodiment, the Fc region comprises FcMut028. In an embodiment, the Fc region comprises FcMut029. In an embodiment, the Fc region comprises FcMut030. In an embodiment, the Fc region comprises FcMut031. In an embodiment, the Fc region comprises FcMut032. In an embodiment, the Fc region comprises FcMut033. In an embodiment, the Fc region comprises FcMut034. In an embodiment, the Fc region comprises FcMut035. In an embodiment, the Fc region comprises FcMut036. In an embodiment, the Fc region comprises FcMut037. In an embodiment, the Fc region comprises FcMut038. In an embodiment, the Fc region comprises FcMut039. In an embodiment, the Fc region comprises FcMut040. In an embodiment, the Fc region comprises FcMut041. In an embodiment, the Fc region comprises FcMut042. In an embodiment, the Fc region comprises FcMut043. In an embodiment, the Fc region comprises FcMut044. In an embodiment, the Fc region comprises FcMut045. In an embodiment, the Fc region comprises FcMut046. In an embodiment, the Fc region comprises FcMut047. In an embodiment, the Fc region comprises FcMut048. In an embodiment, the Fc region comprises FcMut049. In an embodiment, the Fc region comprises FcMut050. In an embodiment, the Fc region comprises FcMut051. In an embodiment, the Fc region comprises FcMut052. In an embodiment, the Fc region comprises FcMut053. In an embodiment, the Fc region comprises FcMut067. In an embodiment, the Fc region comprises FcMut068. In an embodiment, the Fc region comprises FcMut069. In an embodiment, the Fc region comprises FcMut070. In an embodiment, the Fc region comprises FcMut071. In an embodiment, the Fc region comprises FcMut072. In an embodiment, the Fc region comprises FcMut073. In an embodiment, the Fc region comprises FcMut074. In an embodiment, the Fc region comprises FcMut075. In an embodiment, the Fc region comprises FcMut076. In an embodiment, the Fc region comprises FcMut077. In an embodiment, the Fc region comprises FcMut078. In an embodiment, the Fc region comprises FcMut079. In an embodiment, the Fc region comprises FcMut080. In an embodiment, the Fc region comprises FcMut081. In an embodiment, the Fc region comprises FcMut082. In an embodiment, the Fc region comprises FcMut083. In an embodiment, the Fc region comprises FcMut084. In an embodiment, the Fc region comprises FcMut085. In an embodiment, the Fc region comprises FcMut086. In an embodiment, the Fc region comprises FcMut087. In an embodiment, the Fc region comprises FcMut088. In an embodiment, the Fc region comprises FcMut089. In an embodiment, the Fc region comprises FcMut090. In an embodiment, the Fc region comprises FcMut091. In an embodiment, the Fc region comprises FcMut093. In an embodiment, the Fc region comprises FcMut094. In an embodiment, the Fc region comprises FcMut095. In an embodiment, the Fc region comprises FcMut096. In an embodiment, the Fc region comprises FcMut097. In an embodiment, the Fc region comprises FcMut098. In an embodiment, the Fc region comprises FcMut099. In an embodiment, the Fc region comprises FcMut100. In an embodiment, the Fc region comprises FcMut101. In an embodiment, the Fc region comprises FcMut102. In an embodiment, the Fc region comprises FcMut103. In an embodiment, the Fc region comprises FcMut104. In an embodiment, the Fc region comprises FcMut105. In an embodiment, the Fc region comprises FcMut106. In an embodiment, the Fc region comprises FcMut107. In an embodiment, the Fc region comprises FcMut108. In an embodiment, the Fc region comprises FcMut109. In an embodiment, the Fc region comprises FcMut110. In an embodiment, the Fc region comprises FcMut111. In an embodiment, the Fc region comprises FcMut112. In an embodiment, the Fc region comprises FcMut113. In an embodiment, the Fc region comprises FcMut114. In an embodiment, the Fc region comprises FcMut115. In an embodiment, the Fc region comprises FcMut116. In an embodiment, the Fc region comprises FcMut117. In an embodiment, the Fc region comprises FcMut118. In an embodiment, the Fc region comprises FcMut119. In an embodiment, the Fc region comprises FcMut120. In an embodiment, the Fc region comprises FcMut121. In an embodiment, the Fc region comprises FcMut122. In an embodiment, the Fc region comprises FcMut123. In an embodiment, the Fc region comprises FcMut124. In an embodiment, the Fc region comprises FcMut125. In an embodiment, the Fc region comprises FcMut126. In an embodiment, the Fc region comprises FcMut127. In an embodiment, the Fc region comprises FcMut128. In an embodiment, the Fc region comprises FcMut129. In an embodiment, the Fc region comprises FcMut130. In an embodiment, the Fc region comprises FcMut131. In an embodiment, the Fc region comprises FcMut132. In an embodiment, the Fc region comprises FcMut133. In an embodiment, the Fc region comprises FcMut134. In an embodiment, the Fc region comprises FcMut135. In an embodiment, the Fc region comprises FcMut136. In an embodiment, the Fc region comprises FcMut137. In an embodiment, the Fc region comprises FcMut138. In an embodiment, the Fc region comprises FcMut139. In an embodiment, the Fc region comprises FcMut140. In an embodiment, the Fc region comprises FcMut141. In an embodiment, the Fc region comprises FcMut142. In an embodiment, the Fc region comprises FcMut143. In an embodiment, the Fc region comprises FcMut144. In an embodiment, the Fc region comprises FcMut145. In an embodiment, the Fc region comprises FcMut146. In an embodiment, the Fc region comprises FcMut147. In an embodiment, the Fc region comprises FcMut148. In an embodiment, the Fc region comprises FcMut149. In an embodiment, the Fc region comprises FcMut150. In an embodiment, the Fc region comprises FcMut151. In an embodiment, the Fc region comprises FcMut152. In an embodiment, the Fc region comprises FcMut153. In an embodiment, the Fc region comprises FcMut154. In an embodiment, the Fc region comprises FcMut155. In an embodiment, the Fc region comprises FcMut156. In an embodiment, the Fc region comprises FcMut157. In an embodiment, the Fc region comprises FcMut158. In an embodiment, the Fc region comprises FcMut159. In an embodiment, the Fc region comprises FcMut160. In an embodiment, the Fc region comprises FcMut161. In an embodiment, the Fc region comprises FcMut162. In an embodiment, the Fc region comprises FcMut163. In an embodiment, the Fc region comprises FcMut164. In an embodiment, the Fc region comprises FcMut165. In an embodiment, the Fc region comprises FcMut166. In an embodiment, the Fc region comprises FcMut167. In an embodiment, the Fc region comprises FcMut168. In an embodiment, the Fc region comprises FcMut169. In an embodiment, the Fc region comprises FcMut170. In an embodiment, the Fc region comprises FcMut171. In an embodiment, the Fc region comprises FcMut172. In an embodiment, the Fc region comprises FcMut173. In an embodiment, the Fc region comprises FcMut174. In an embodiment, the Fc region comprises FcMut175. In an embodiment, the Fc region comprises FcMut176. In an embodiment, the Fc region comprises FcMut177. In an embodiment, the Fc region comprises FcMut178. In an embodiment, the Fc region comprises FcMut179. In an embodiment, the Fc region comprises FcMut180. In an embodiment, the Fc region comprises FcMut181. In an embodiment, the Fc region comprises FcMut182. In an embodiment, the Fc region comprises FcMut183. In an embodiment, the Fc region comprises FcMut184. In an embodiment, the Fc region comprises FcMut185. In an embodiment, the Fc region comprises FcMut186. In an embodiment, the Fc region comprises FcMut187. In an embodiment, the Fc region comprises FcMut188. In an embodiment, the Fc region comprises FcMut189. In an embodiment, the Fc region comprises FcMut190. In an embodiment, the Fc region comprises FcMut191. In an embodiment, the Fc region comprises FcMut192. In an embodiment, the Fc region comprises FcMut193. In an embodiment, the Fc region comprises FcMut194. In an embodiment, the Fc region comprises FcMut195. In an embodiment, the Fc region comprises FcMut196. In an embodiment, the Fc region comprises FcMut197. In an embodiment, the Fc region comprises FcMut198. In an embodiment, the Fc region comprises FcMut199. In an embodiment, the Fc region comprises FcMut200. In an embodiment, the Fc region comprises FcMut201. In an embodiment, the Fc region comprises FcMut202. In an embodiment, the Fc region comprises FcMut203. In an embodiment, the Fc region comprises FcMut204. In an embodiment, the Fc region comprises FcMut205. In an embodiment, the Fc region comprises FcMut206. In an embodiment, the Fc region comprises FcMut207. In an embodiment, the Fc region comprises FcMut208. In an embodiment, the Fc region comprises FcMut209. In an embodiment, the Fc region comprises FcMut210. In an embodiment, the Fc region comprises FcMut211. In an embodiment, the Fc region comprises FcMut212. In an embodiment, the Fc region comprises FcMut213. In an embodiment, the Fc region comprises FcMut214. In an embodiment, the Fc region comprises FcMut215. In an embodiment, the Fc region comprises FcMut216. In an embodiment, the Fc region comprises FcMut217. In an embodiment, the Fc region comprises FcMut218. In an embodiment, the Fc region comprises FcMut219. In an embodiment, the Fc region comprises FcMut220. In an embodiment, the Fc region comprises FcMut221. In an embodiment, the Fc region comprises FcMut222. In an embodiment, the Fc region comprises FcMut223. In an embodiment, the Fc region comprises FcMut224. In an embodiment, the Fc region comprises FcMut225. In an embodiment, the Fc region comprises FcMut226. In an embodiment, the Fc region comprises FcMut227. In an embodiment, the Fc region comprises FcMut228. In an embodiment, the Fc region comprises FcMut229. In an embodiment, the Fc region comprises FcMut230. In an embodiment, the Fc region comprises FcMut231. In an embodiment, the Fc region comprises FcMut232. In an embodiment, the Fc region comprises FcMut233. In an embodiment, the Fc region comprises FcMut234. In an embodiment, the Fc region comprises FcMut242. In an embodiment, the Fc region comprises FcMut243. In an embodiment, the Fc region comprises FcMut244. In an embodiment, the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, or more) of mutations or combinations of mutations chosen from FcMut045, FcMut171, FcMut183, FcMut186, FcMut190, FcMut197, FcMut213, FcMut215, FcMut216, FcMut219, FcMut222, FcMut223, FcMut224, FcMut226, FcMut227, FcMut228, or FcMut229. In an embodiment, the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, or all) of mutations or combinations of mutations chosen from FcMut045, FcMut183, FcMut197, FcMut213, FcMut215, FcMut228, or FcMut156. In another embodiment, the Fc region comprises one or more (e.g., 2, 3, 4, 5, or all) of mutations or combinations of mutations chosen from FcMut183, FcMut197, FcMut213, FcMut215, FcMut228, or FcMut229. In an embodiment, the Fc region does not comprise one or more (e.g., 2, 3, 4, or all) of mutations or combinations of mutations chosen from FcMut018, FcMut021, FcMut050, FcMut102, or YTE. In an embodiment, the Fc region comprises one or more (e.g., 2, 3, 4, or all) of mutations or combinations of mutations chosen from FcMut018, FcMut021, FcMut050, FcMut102, or YTE, and one or more other mutations or combinations of mutations described in Table 1. In an embodiment, the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more) of mutations or combinations of mutations described in Table 1 that result in a synergistic effect (e.g., binding affinity or circulating half-life) as described herein. In an embodiment, the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, or 7) mutations in residues chosen from T256, H285, N286, T307, Q311, N315, or A378. In an embodiment, the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, or 7) mutations chosen from T256D, H285N, N286D, T307Q, Q311V, N315D, or A378V. In an embodiment, the Fc region comprises a half-life enhancing mutation, a mutation that is capable of disrupting an Fc effector function, or both. In an embodiment, the Fc region comprises one or more mutations or combinations of mutations described herein, e.g., chosen from M252W, V308F/N434Y, R255Y, P257L/N434Y, V308F, P257N/M252Y, G385N, P257N/V308Y, N434Y, M252Y/S254T/T256E (“YTE”), M428L/N434S (“LS”), or any combination thereof. Alternatively, or additionally, in an embodiment, the Fc region comprises (a) one or more (e.g., 2, 3, 4, 5, or all) combinations of mutations chosen from: T256D/Q311V/A378V, H285N/T307Q/N315D, H285D/T307Q/A378V, T307Q/Q311V/A378V, T256D/N286D/T307R/Q311V/A378V, or T256D/T307R/Q311V; (b) a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., L234A/L235A (also known as “LALA” mutation), or (c) both (a) and (b). In an embodiment, the Fc region comprises mutations T256D/Q311V/A378V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., L234A/L235A. In an embodiment, the Fc region comprises mutations H285N/T307Q/N315D and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., L234A/L235A. In an embodiment, the Fc region comprises mutations H285D/T307Q/A378V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., L234A/L235A. In an embodiment, the Fc region comprises mutations T307Q/Q311V/A378V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., L234A/L235A. In an embodiment, the Fc region comprises mutations T256D/N286D/T307R/Q311V/A378V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., L234A/L235A. In an embodiment, the Fc region comprises mutations T256D/T307R/Q311V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., L234A/L235A. A reference Fc region amino acid sequence (including the numbering used herein) is provided below (e.g., for identification of the mutation positions described herein). The CH2 domain sequence is underlined; the hinge region sequence is in italics, and the CH3 domain sequence is in bold. (SEQ ID NO: 1)118ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL175QSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPA232PELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHN287AKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQP344REPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS401DGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK Any of the half-life extension mutations described herein can be used in combination with any Fc mutation capable of enhancing or disrupting an Fc effector function. The Fc region can bind to various cell receptors (e.g., Fc receptors) and complement proteins. The Fc region can also mediate different physiological effects of antibody molecules, e.g., detection of opsonized particles; cell lysis; degranulation of mast cells, basophils, and eosinophils; and other processes. There are several different types of Fc receptors (FcR), which can be classified based on the type of antibody that they recognize. Fcγ receptors (FcγR) belong to the immunoglobulin superfamily, and are involved, e.g., in inducing phagocytosis of opsonized microbes. This family includes several members, FcγRI (CD64), FcγRIIA (CD32), FcγRIIB (CD32), FcγRIIIA (CD16a), FcγRIIIB (CD16b), which differ in their antibody affinities due to their different molecular structure. For instance, FcγRI can bind to IgG more strongly than FcγRII or FcγRIII does. FcγRI also has an extracellular portion comprising three immunoglobulin (Ig)-like domains, one more domain than FcγRII or FcγRIII has. This property allows FcγRI to bind a sole IgG molecule (or monomer), but Fcγ receptors generally need to bind multiple IgG molecules within an immune complex to be activated. The Fcγ receptors differ in their affinity for IgG and the different IgG subclasses can have unique affinities for each of the Fcγ receptors. These interactions can be further tuned by the glycan (oligosaccharide) at certain position of IgG. For example, by creating steric hindrance, fucose containing CH2-84.4 glycans reduce IgG affinity for FcγRIIIA, whereas GO glycans, which lack galactose and terminate instead with GlcNAc moieties, have increased affinity for FcγRIIIA (Maverakis et al. (2015)Journal of Autoimmunity57 (6): 1-13) The neonatal Fc receptor (FcRn) is expressed on multiple cell types and is similar in structure to MHC class I. This receptor also binds IgG and is involved in preservation of this antibody (Zhu et al. (2001).Journal of Immunology166 (5): 3266-76.). FcRn is also involved in transferring IgG from a mother either via the placenta to her fetus or in milk to her suckling infant. This receptor may also play a role in the homeostasis of IgG serum levels. FcαRI (or CD89) belongs to the FcαR subgroup. FcαRI is found on the surface of neutrophils, eosinophils, monocytes, macrophages (including Kupffer cells), and dendritic cells. It comprises two extracellular Ig-like domains, and is a member of both the immunoglobulin superfamily and the multi-chain immune recognition receptor (MIRR) family. It signals by associating with two FcRγ signaling chains. Fc-alpha/mu receptor (Fcα/μR) is a type I transmembrane protein. It can bind IgA, although it has higher affinity for IgM (Shibuya and Honda (2006)Springer Seminars in Immunopathology28 (4): 377-82). With one Ig-like domain in its extracellular portion, this Fc receptor is also a member of the immunoglobulin superfamily. There are two known types of FcεR. The high-affinity receptor FcεRI is a member of the immunoglobulin superfamily (it has two Ig-like domains). FcεRI is found on epidermal Langerhans cells, eosinophils, mast cells and basophils. This receptor can play a role in controlling allergic responses. FcεRI is also expressed on antigen-presenting cells, and controls the production of immune mediators, e.g., cytokines that promote inflammation (von Bubnoff et al. (2003)Clinical and Experimental Dermatology28 (2): 184-7). The low-affinity receptor FcεRII (CD23) is a C-type lectin. FcεRII has multiple functions as a membrane-bound or soluble receptor. It can also control B cell growth and differentiation and blocks IgE-binding of eosinophils, monocytes, and basophils (Kikutani et al. (1989)Ciba Foundation Symposium147: 23-31). In an embodiment, the Fc region can be engineered to contain an antigen-binding site to generate an Fcab fragment (Wozniak-Knopp et al. (2010)Protein Eng Des23 (4): 289-297). Fcab fragments can be inserted into a full immunoglobulin by swapping the Fc region, thus obtaining a bispecific antibody (with both Fab and Fcab regions containing distinct binding sites). The binding and recycling of FcRn can be illustrated below. For example, IgG and albumin are internalized into vascular endothelial cells through pinocytosis. The pH of the endosome is 6.0, facilitating association with membrane-bound FcRn. The contents of endosomes can be processed in one of two ways: either recycling back to the apical cell membrane or transcytosis from the apical to the basolateral side. IgG not associated with FcRn is degraded by lysosomes. While not wishing to be bound by theory, it is believed that FcRn interaction with IgG is mediated through Fc. The binding of Fc to FcRn is pH specific, e.g., no significant binding at pH 7.4 and strong binding in acidic environment. Structure of FcRn in complex with Fc domain of IgG1 molecule is known inFIG.1. Each FcRn molecule generally binds to an Fc-monomer. In an embodiment, Fab domains can also influence binding of IgG to FcRn, e.g., have either a negative or no influence on the affinity of the IgG for FcRn. There can be multiple considerations when an Fc region is engineered to enhance half-life of a polypeptide. For example, prolonging half-life and efficient recirculation of antibody molecules or fusion proteins often requires pH specific affinity enhancement (e.g., only at low pH of the endosome). FcRn binds proximal to the linker region between CH2 and CH3 domains of a Fc region. Modifications to the linker can impact Fc engagement with Fcγ receptors. Modifications on the Fc region can impact thermal stability and aggregation properties of the polypeptide. FcRn Binding Optimization: Reducing Impact on Other Effector Functions In an embodiment, the polypeptide (e.g., antibody molecule or fusion protein) described herein has the same affinity function, or does not substantially alter (e.g., decrease by more than 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%) an effector function (e.g., an effector function described herein). In an embodiment, the effector function is not associated with the binding between an Fc region and an FcRn. The amino acid residues to be mutated can be selected, at least in part, based on the structural or functional properties of one or more binding sites on the Fc region. These binding sites include, but are not limited to, a Protein A binding site, a C1q binding site, an FcγRI binding site, an FcγRIIa binding site, an FcγRIIIa binding site, or an FcRn binding site, e.g., as shown inFIG.2. The binding sites can also include a TRIM21 binding site, e.g., one or more residues chosen from loop 308-316, loop 252-256, or loop 429-436 of an IgG. In an embodiment, the linker region between the CH2 and CH3 domain can influence the dynamics of the CH2 domain which impinges on FcγR binding. Structural Basis for pH Specific Engagement of FcRn Without wishing to be bound by theory, it is believed that low pH of the endosome leads to protonation of surface histidines on the CH2 and CH3 domains. For example, protonation of residue H310 on CH2 and/or H433 on CH3 can be important for FcRn engagement, e.g., at low pH (e.g., at pH 6.0). Protonation can also lead to change in the conformational dynamics of the region, such as better exposure or shielding of the linker region for solvent or ligand molecule binding. Accordingly, in an embodiment, the polypeptide (e.g., antibody molecule or fusion protein) comprises a mutation in residue H310, a mutation in residue H433, or both. One or more residues adjacent to residues H310 and/or H433 can also be mutated. The polypeptide can also include a compensating or beneficial mutation, e.g., a mutation that compensates, or beneficial, for any of the aforesaid mutations, e.g., to reduce a negative consequence of that mutation (e.g., polar vs. non-polar, charged vs. no charge, positively-charged (basic) vs. negatively charged (acidic), or hydrophobic vs. hydrophilic). For example, P247D can be a compensating or beneficial mutation. In an embodiment, protonation of histidine can result in additional conformational changes including, e.g., movement/displacement of the linker/CH2/CH3 interface residues. Crystal structures of Fc fragments have been crystallized at different pHs. As shown inFIG.3, an analysis of two high resolution crystal structures of Fc fragments crystallized and pH 6.5 (cyan) and 5.0 (green) indicated potential differences. Mapping of Fc-FcRn Interaction Interface In an embodiment, the polypeptide (e.g., antibody molecule or fusion protein) described herein comprises a mutation that can alter the interaction between an Fc region and an FcRn. In an embodiment, the mutation is selected based, at least in part, on a structural feature of the Fc-FcRn interaction interface. In an embodiment, the Fc region of an immunoglobulin monomer can have the structure shown inFIG.4. As shown inFIG.4, the black dotted line indicates the Fc-FcRn interaction interface. The structure includes FcRn contact residues and FcRn affinity enhancing Fc residues, e.g., as described herein. Residues H310 and H435, which are located in the CH2 and CH3 domains, respectively, are primarily responsible for the pH dependent Fc-FcRn interactions. In an embodiment, the FcRn binds the Fc region between its CH2 and CH3 domains. In another embodiment, the Fc-FcRn binding site is located across both the CH2 and CH3 domains of the Fc monomer. Network View of Fc-FcRn Engagement Provides Insights on Co-Substitutions of Amino Acids In an embodiment, the polypeptide (e.g., antibody molecule or fusion protein) described herein comprises a plurality of mutations that can alter the interaction between an Fc region and an FcRn. In an embodiment, the mutation is selected based, at least in part, on a network view of the Fc-FcRn interaction. Without wishing to be bound by theory, it is believed that in an embodiment, residue H310 plays a central role in engagement with the FcRn, e.g., as determined by a network analysis of the Fc-FcRn complex. As shown inFIG.5, residue H310 is highly interconnected to multiple other highly networked residues. In an embodiment, a mutation in the H310 cluster, and neighboring (connected nodes), can strengthen the H310 network. Analysis of sub-networks informs introduction of synergistic mutations for favorable FcRn interaction; with reduced or minimal impact on other Fc residues. Design Considerations for Optimizing FcRn Binding In an embodiment, the polypeptide (e.g., antibody molecule or fusion protein) described herein can be designed for optimizing Fc-FcRn binding. In an embodiment, the polypeptide having a mutation in the Fc region has a pH-specific affinity enhancement, compared to a reference polypeptide (e.g., an otherwise identical polypeptide without the mutation). In an embodiment, affinity enhancement is achieved by increasing van der Waal interaction. In an embodiment, affinity enhancement is not achieved by introduction of hydrogen bonds and/or electrostatic interaction. In an embodiment, the mutation does not alter, or has reduced or minimal perturbation to, the conformation of the linker region between the CH2 and CH3 domains. In an embodiment, the polypeptide comprises a plurality of mutations across both domains (four quadrants). In an embodiment, the polypeptide does not contain a large cluster of hydrophobic or aromatic residues on the surface. In an embodiment, the polypeptide comprises a mutation that enhances the strength of interaction between an Fc region and FcRn or reduces the dissociation constant (Kd) for FcRn, e.g., at an acidic pH. In an embodiment, the polypeptide comprises a mutation that reduces the rate of dissociation (koff) for FcRn, e.g., at an acidic pH. In an embodiment, the polypeptide comprises a mutation that increases the rate of association (kon) for FcRn, e.g., at an acidic pH. In an embodiment, the polypeptide comprises a mutation that reduces the rate of dissociation (koff) for FcRn, and increases the rate of association (kon) for FcRn, e.g., at an acidic pH. In an embodiment, the polypeptide comprises a mutation that reduces the rate of dissociation (koff) for FcRn, and does not, or does not significantly, affect the rate of association (kon) for FcRn, e.g., at an acidic pH. Without wishing to be bound by theory, it is believed that in an embodiment, the reduction of the dissociation constant Kdfor FcRn is primarily resulted from the reduction of the rate of dissociation (koff) for FcRn, rather than the increase of the rate of association (kon). Experimental Evaluation: Fc Mutations Assessed Along Multiple Dimensions and by Different Assay Platforms The polypeptides (e.g., antibody molecules or fusion proteins) described herein can be evaluated by a number of methods. The pH-specific Fc-FcRn binding (e.g., binding at pH6.0 and pH7.4) can be determined, e.g., by an Octet-based assay, a competition assay (e.g., flow cytometry), or surface plasmon resonance (SPR). Biophysical characterization (e.g., thermal stability, aggregation, or expression) can be performed. Thermal stability can be determined, e.g., by SYPRO orange. Aggregation can be measured, e.g., by SEC or RP-HPLC. Effector functions, e.g., relating to FcγRI (e.g., by an Octet-based assay), FcγRIIIa, FcγRIIa, FcγRIIb (e.g., by ELISA), C1q, ADCC, or CDC, can be tested. Tripartite Motif-Containing Protein 21 (TRIM21) binding can be tested. TRIM21 is a cytosolic receptor that binds with Fc of IgG. TRIM21 plays a role in mediating intracellular recognition and neutralization of Fc bound pathogens such as viruses, bacteria and fungus. For example, TRIM21 plays an important role in the neutralization of non-enveloped viruses (McEwan et al.Nat Immunol.2013; 14(4):327-36). Its role has been further expanded to include directing of immune complexes for degradation (McEwan et al.Proc Natl Acad Sci USA.2017; 114(3):574-579). TRIM21 binds to the CH2:CH3 interface of the antibody Fc region, which overlaps with the FcRn binding site. Some Fc mutations that increase FcRn affinity decrease TRIM21 affinity (Foss et al.J Immunol.2016; 196(8):3452-3459). Key contact residues include, e.g., positions 253, 433, 434, and 435. The LS variant (M428L/N434S) contains a mutation at position 434, and it has been shown that the N434S mutation causes a 10 fold decrease in TRIM21 binding. TRIM21-mediated neutralization is known as antibody dependent intracellular neutralization (ADIN). Mucosal uptake can be tested. FcRn transports IgG across different cellular barriers such as the mucosal epithelium lining the intestine and the alveolar surfaces. Modification of FcRn binding provides a mechanism to enhance mucosal localization that confers immune protection. The half-life of the polypeptide can be measured, e.g., using transgenic mice (e.g., Tg32 and Tg276 mice from Jackson's lab), or in primates (e.g., cynomolgus monkeys). Exemplary assays are described in more detail as follows: 1. FcRn Binding Assays a. Octet Assay with Immobilization of FcRn to NiNTA Biosensors Immobilization of FcRn to a NiNTA biosensor via a 6× histidine tag (SEQ ID NO: 2) allows for subsequent interrogation of binding to IgG molecules under acidic (pH 6.0) and physiological (pH 7.4) conditions. This strategy has been previously described (1) and this method details an adaptation of the referenced protocol. Briefly, recombinant human FcRn at 5 μg/mL is loaded onto a NiNTA biosensor for 180 seconds. After a 60 second baseline step in 1×PBS pH 6.0, the FcRn loaded tip is exposed to IgG at a concentration of 250 nM (37.5 μg/mL) for 60 seconds, followed by dissociation for 60 seconds in PBS pH 6.0, and an additional 30 seconds in PBS pH 7.4. After assay completion, a quantitative assessment of the affinity constant (KD) at pH 6.0 is performed using the ForteBio octet software and a qualitative assessment is performed by plotting the response rate over time, allowing for visualization of the association of IgG to FcRn at pH 6.0 and the subsequent dissociation at pH 6.0 and pH 7.4. b. Octet Assay with Immobilization of IgG to Anti-CH1 Biosensors Immobilization of IgG to an anti-CH1 biosensor allows for subsequent interrogation of binding to FcRn molecules under acidic (pH 6.0) and physiological (pH 7.4) conditions. This strategy has been previously described (2) and this method details an adaptation of the referenced protocol. Briefly, purified IgG at 5 μg/mL is loaded onto an anti-CH1 biosensor for 180 seconds. After a 60 second baseline step in 1×PBS pH 6.0, the IgG loaded tip is exposed to FcRn at a concentration of 50 μg/mL for 60 seconds, followed by dissociation for 60 seconds in PBS pH 6.0, and an additional 30 seconds in PBS pH 7.4. After assay completion, a quantitative assessment of the affinity constant (KD) at pH 6.0 is performed using the ForteBio octet software and a qualitative assessment is performed by plotting the response rate over time, allowing for visualization of the association of IgG to FcRn at pH 6.0 and the subsequent dissociation at pH 6.0 and pH 7.4. c. Cell Based Assay Cell-based assays are also used to analyze the interactions between FcRn and IgG (Ref: PMID: 23384837). Expression of membrane-anchored FcRn on the cell surface closely represents the physiological presentation of FcRn, where the plasma membrane and molecular orientations influence interactions between FcRn and IgG. The assay used here is a competition assay in which IgGs of interest are evaluated for their ability to compete with cell binding of a fluorescently-labeled, high affinity Fc competitor reagent (Fc-A488). Expi293 cells expressing the full-length, membrane-bound FcRn heterodimer are incubated with mixtures of a static concentration of Fc-A488 (0.5 ug/ml) and varying concentrations of IgG of interest (0.001-10 μM). Cell-bound fluorescence is detected by flow cytometry. IgGs with improved binding to FcRn will compete off the Fc competitor at lower IgG concentrations. This assay is robust, linear, and specific and can be used to show differences in relative binding of IgG/Fc variants to FcRn. 2. Thermal Stability Assay The stability of the IgG variants was assessed by Differential Scanning Fluorimetry (DSF) is an assay using SYPRO® Orange dye to monitor protein unfolding under thermal stress. SYPRO® Orange is a fluorescent dye that non-specifically binds to hydrophobic surfaces and its fluorescence is quenched in aqueous environments. Proteins begin to lose their secondary structure and unfold with increasing temperatures thus exposing their hydrophobic core residues, and allowing the dye to fluoresce. Maximum fluorescent signal is attained at complete unfolding, after which the protein begins to aggregate, reducing the exposed hydrophobic residues, and thus reducing the fluorescent signal. At the midpoint between native state and fully unfolded protein is the transition temperature, or melt temperature (Tm), which can be used to directly compare protein constructs or formulation conditions for relative stability. In an embodiment, factors other than FcRn binding can also affect the observed half-life of a polypeptide (e.g., an antibody molecule or fusion protein). These factors can include, e.g., aggregation propensity, non-specific binding, stability, or Fab composition. In an embodiment, a mutation in a non-Fc region is introduced, e.g., to provide a substantial improvement in half-life in the context of a suboptimal template. For example, polypeptides (e.g., antibody molecules or fusion proteins) with lower starting half-life may indicate the presence of suboptimal properties. Engineering efforts can also focus on mitigating the root cause of lower half-life, e.g., to maintain requisite binding profile (e.g., affinity and/or specificity), or to obtain suitable developability characteristics (e.g., stability, solubility, expression level, or aggregation). In an embodiment, additional engineering is performed for polypeptide (e.g., antibody molecules or fusion proteins) with suboptimal biophysical properties to realize maximal half-life extension. Pharmacokinetics The polypeptides (e.g., antibody molecules or fusion proteins) described herein can have one or more desired pharmacokinetic properties, e.g., one or more (e.g., 2, 3, 4, 5, or more) of the pharmacokinetic properties described herein. Pharmacokinetics (PK) can be used to determine the fate of substances administered to a living organism. PK studies can be used to analyze drug metabolism and to identify the fate of a drug from the moment that it is administered up to the point at which it is completely eliminated from the body. Pharmacokinetics describes, e.g., how the body affects a specific drug after administration through the mechanisms of absorption and distribution, the chemical changes of the substance in the body (e.g. by metabolic enzymes such as cytochrome P450 or glucuronosyltransferase enzymes), or the effects and routes of excretion of the metabolites of the drug. Pharmacokinetic properties of drugs may be affected by elements such as the site of administration and the dose of administered drug, which may affect the absorption rate. Pharmacokinetics can be analyzed in conjunction with pharmacodynamics (e.g., the study of the biochemical and physiologic effects of drugs). A number of different models have been developed for pharmacokinetics. For example, pharmacokinetic modelling can be performed by noncompartmental or compartmental methods. Noncompartmental methods estimate the exposure to a drug by estimating the area under the curve of a concentration-time graph. Noncompartmental PK analysis is highly dependent on estimation of total drug exposure. Total drug exposure is often estimated by area under the curve (AUC) methods, with the trapezoidal rule (numerical integration) the most common method. Due to the dependence on the length of ‘x’ in the trapezoidal rule, the area estimation is highly dependent on the blood/plasma sampling schedule. That is, the closer time points are, the closer the trapezoids reflect the actual shape of the concentration-time curve. Compartmental methods estimate the concentration-time graph using kinetic models. Compartmental PK analysis uses kinetic models to describe and predict the concentration-time curve. PK compartmental models are often similar to kinetic models used in other scientific disciplines such as chemical kinetics and thermodynamics. The advantage of compartmental over some noncompartmental analyses is the ability to predict the concentration at any time. Compartment-free modelling based on curve stripping does not suffer this limitation. The simplest PK compartmental model is the one-compartmental PK model with IV bolus administration and first-order elimination. The more complex PK models (e.g., PBPK models) rely on the use of physiological information to ease development and validation. In a single-compartment model, the graph of the relationship between the various factors involved (e.g., dose, blood plasma concentrations, or elimination) gives a straight line or an approximation to one (i.e., linear pharmacokinetics). For drugs to be effective they need to be able to move rapidly from blood plasma to other body fluids and tissues. In multi-compartmental models, the graph for the non-linear relationship between the various factors is represented by a curve, the relationships between the factors can then be found by calculating the dimensions of different areas under the curve. The models used in non-linear pharmacokinetics are largely based on Michaelis-Menten kinetics. The various compartments that the model is divided into are commonly referred to as the ADME scheme (also referred to as LADME if liberation is included as a separate step from absorption): liberation (e.g., the process of release of a drug from the pharmaceutical formulation); absorption (e.g., the process of a substance entering the blood circulation); distribution (e.g., the dispersion or dissemination of substances throughout the fluids and tissues of the body); metabolism (or biotransformation, or inactivation) (e.g., the recognition by the organism that a foreign substance is present and the irreversible transformation of parent compounds into daughter metabolites); and excretion (e.g., the removal of the substances from the body). In rare cases, some drugs irreversibly accumulate in body tissue. The two phases of metabolism and excretion can also be grouped together under the title elimination. All these parameters can be represented through mathematical formulas that have a corresponding graphical representation. The use of these models allows an understanding of the characteristics of a molecule, as well as how a particular drug will behave given information regarding some of its basic characteristics such as its acid dissociation constant (pKa), bioavailability and solubility, absorption capacity and distribution in the organism. The model outputs for a drug can be used in industry (for example, in calculating bioequivalence when designing generic drugs) or in the clinical application of pharmacokinetic concepts. Clinical pharmacokinetics provides a number of performance guidelines for effective and efficient use of drugs for human-health professionals and in veterinary medicine. Exemplary pharmacokinetic properties include, but are not limited to, dose (e.g., the amount of drug administered), dosing interval (e.g., the time between drug dose administrations), Cmax(e.g., the peak plasma concentration of a drug after administration), tmax(e.g., the time to reach Cmax), (e.g., the lowest concentration that a drug reaches before the next dose is administered), volume of distribution (e.g., the apparent volume in which a drug is distributed, e.g., relating drug concentration to drug amount in the body), concentration (e.g., the amount of drug in a given volume of plasma), half-life or elimination half-life (e.g., the time required for the concentration of the drug to reach half of its original value), elimination rate constant (e.g., the rate at which a drug is removed from the body), infusion rate (e.g., the rate of infusion required to balance elimination), area under the curve (e.g., the integral of the concentration-time curve, e.g., after a single dose or in steady state), clearance (e.g., the volume of plasma cleared of the drug per unit time), bioavailability (e.g., the systemically available fraction of a drug), or fluctuation (e.g., the peak trough fluctuation within one dosing interval at steady state). Pharmacokinetic properties can be measured by various methods. For example, bioanalytical methods can be used to construct a concentration-time profile. Chemical techniques can be employed to measure the concentration of drugs in biological matrix, e.g., plasma. Proper bioanalytical methods should be selective and sensitive. For example, microscale thermophoresis can be used to quantify how the biological matrix/liquid affects the affinity of a drug to its target (Baaske et al. (2010).Angew. Chem. Int. Ed.49 (12): 1-5; Wienken et al. (2010).Nature Communications1 (7): 100). Pharmacokinetic properties can also be studied using mass spectrometry, e.g., when there is a need for high sensitivity to observe concentrations after a low dose and a long time period. A common instrumentation used in this application is LC-MS with a triple quadrupole mass spectrometer. Tandem mass spectrometry can be employed for added specificity. Standard curves and internal standards can be used for quantitation of a pharmaceutical in the samples. The samples represent different time points as a pharmaceutical is administered and then metabolized or cleared from the body. Blank samples taken before administration are used in determining background and ensuring data integrity with such complex sample matrices. The standard curve can be linear, or curve fitting can be used with more complex functions such as quadratics since the response of most mass spectrometers is less than linear across large concentration ranges (Hsieh and Korfmacher (2006) Current Drug Metabolism 7 (5): 479-89; Covey et al. (1986) Anal. Chem. 58 (12): 2453-60; Covey et al. (1985). Anal. Chem. 57 (2): 474-81). Exemplary Antibody Molecules The methods described herein can be used to engineer a variety of antibody molecules, e.g., any antibody molecule containing an Fc region. In an embodiment, the antibody molecule is a chimeric antibody molecule, a humanized antibody molecule, or a human antibody molecule. In an embodiment, the antibody molecule is a whole monoclonal antibody. In another embodiment, the antibody molecule is an Fc region-containing derivative of an antibody molecule that does not contain an Fc region (e.g., an antigen-binding fragment described herein). Exemplary antigen-binding fragments include, but are not limited to, Fab, F(ab′)2, Fab′, scFv, di-scFv, or sdAb. In another embodiment, the antibody molecule is a bispecific monoclonal antibody, e.g., a trifunctional antibody (3funct) or bi-specific T-cell engager (BiTE). In an embodiment, the antibody molecule targets a molecule (e.g., a protein) associated with an infectious disease (e.g., a viral infection, a bacterial infection, or a fungal infection). In another embodiment, the antibody molecule targets a molecule (e.g., a protein) or cell associated with a cancer. In another embodiment, the antibody molecule targets a molecule (e.g., a protein) or cell associated with an immune disorder. In another embodiment, the antibody molecule targets a molecule (e.g., a protein) or cell associated with a cardiovascular disorder. In another embodiment, the antibody molecule targets a molecule (e.g., a protein) or cell associated with a metabolic disorder. In another embodiment, the antibody molecule targets a molecule (e.g., a protein) or cell associated with a neurological disorder. Exemplary antibody molecules include, but are not limited to, antibody molecules that target one or more (e.g., 2) of the following molecules or cells: β-amyloid, 4-1BB, SAC, 5T4, ACF9, ACFIX, activin receptor-like kinase 1, ACVR2B, an adenocarcinoma antigen, AGS-22M6, alpha-fetoprotein, angiopoietin 2, angiopoietin 3, a protective antigen of anthrax toxin, AOC3 (VAP-1), B7-H3,bacillusanthracisanthrax, BAFF, B-lymphoma cell, C242 antigen, C5, CA-125 (imitation), calcitonin,canis lupus familiarisIL31, carbonic anhydrase 9 (CA-IX), cardiac myosin, CCL11 (eotaxin-1), CCR2, CCR4, CCR5, CD11, CD18, CD125, CD140a, CD147 (basigin), CD15, CD152, CD154 (CD40L), CD19, CD2, CD20, CD200, CD22, CD221, CD23 (IgE receptor), CD25 (a chain of IL-2 receptor), CD27, CD274, CD276, CD28, CD3, CD3 epsilon, CD30 (TNFRSF8), CD33, CD37, CD38 (cyclic ADP ribose hydrolase), CD4, CD40, CD40 ligand, CD41 (integrin alpha-IIb), CD44 v6, CD5, CD51, CD52, CD56, CD6, CD70, CD74, CD79B, CD80, CEA, a CEA-related antigen, CFD, CGRP, ch4D5, CLDN18.2,Clostridium difficile, clumping factor A, coagulation factor III, CSF1R, CSF2, CTGF, CTLA-4, C—X—C chemokine receptor type 4, cytomegalovirus, cytomegalovirus glycoprotein B, dabigatran, DLL3, DLL4, DPP4, DRS,E. colishiga toxintype-1,E. colishiga toxintype-2, EGFL7, EGFR, endoglin, endotoxin, EpCAM, ephrin receptor A3, episialin, ERBB3,Escherichia coli, F protein of respiratory syncytial virus, FAP, fibrin II beta chain, fibronectin extra domain-B, folate hydrolase, folate receptor 1, folate receptor alpha, Frizzled receptor, ganglioside GD2, GCGR, GD2, GD3 ganglioside, GDF-8, glypican 3, GMCSF receptor α-chain, GPNMB, growth differentiation factor8, GUCY2C, hemagglutinin, hepatitis B surface antigen, hepatitis B virus, HER1, HER2/neu, HER3, HGF, HHGFR, histone complex, HIV-1, HLA-DR, HNGF, Hsp90, human scatter factor receptor kinase, human TNF, human beta-amyloid, ICAM-1 (CD54), ICOSL, IFN-α, IFN-γ, IgE, IgE Fc region, IGF-1 receptor, IGF-I, IGHE, IL 20, IL-1, IL-12, IL-13, IL-17, IL-17A, IL-17F, IL-1β, IL2, IL-22, IL-23, IL23A, IL31RA, IL-4, IL-4, IL-5, IL6, IL-6 receptor (IL6R), IL-9, ILGF2, influenza A virus hemagglutinin (HA), insulin-like growth factor I receptor, integrin α4, integrin α4β7, integrin α5β1, integrin α7β7, integrin αIIbβ3, integrin αvβ3, interferon receptor, interferon α/β receptor, interferon gamma-induced protein, ITGA2, ITGB2 (CD18), kallikrein, KIR2D, KLRC1, Lewis-Y antigen, LFA-1 (CD11a), LFA-1 (CD11a), LINGO-1, lipoteichoic acid, LOXL2, L-selectin (CD62L), LTA, MCP-1, mesothelin, MIF, MS4A1, MSLN, MUC1, mucin CanAg, myelin-associated glycoprotein, myostatin, NCA-90 (granulocyte antigen), NCA-90 (granulocyte antigen), neural apoptosis-regulated proteinase 1, neural apoptosis-regulated proteinase 1, NGF, NGF, N-glycolylneuraminic acid, NOGO-A, Notch 1, Notch receptor, NRP1, Oryctolagus cuniculus, OX-40, oxLDL, PCSK9, PD-1, PD-1, PDCD1, PDGF-R α, phosphate-sodium co-transporter, phosphatidylserine, platelet-derived growth factor receptor beta, prostatic carcinoma cells,Pseudomonas aeruginosa, Pseudomonas aeruginosatype III secretion system, rabies virus glycoprotein, rabies virus glycoprotein, RANKL, respiratory syncytial virus, respiratory syncytial virus, RHD, Rhesus factor, Rhesus factor, RON, RTN4, sclerostin, SDC1, selectin P, SLAMF7, SOST, sphingosine-1-phosphate,Staphylococcus aureus, STEAP1, TAG-72, T-cell receptor, TEM1, tenascin C, TFPI, TGF beta 1, TGF beta 2, TGF-β, TNFR superfamily member 4, TNF-α, TRAIL-R1, TRAIL-R2, TSLP, tumor antigen CTAA16.88, tumor specific glycosylation of MUC1, tumor-associated calcium signal transducer 2, TWEAK receptor, TYRP1(glycoprotein 75), VEGFA, VEGFR-1, VEGFR2, vimentin, or VWF. Exemplary antibody molecules include, but are not limited to, antibody molecules that target one or more (e.g., 2) of the following pathogens (e.g., bacteria, viruses, or fungi):Actinomyces gerencseriae, Actinomyces israelii, Actinomycetomaspecies, Alphavirus,Anaplasma phagocytophilum, Anaplasma species, Ancylostoma braziliense, Ancylostoma duodenale, Angiostrongylus, Anisakis, Arcanobacterium haemolyticum, Ascaris lumbricoides, Aspergillusspecies, Astroviridae family,Babesiaspecies,Bacillus anthracis, Bacillus cereus, bacterial vaginosis microbiota,Bacteroidesspecies,Balantidium coli, Bartonella, Bartonella bacilliformis, Bartonella henselae, Batrachochytrium dendrabatidis, Baylisascarisspecies, BK virus,Blastocystisspecies,Blastomyces dermatitidis, Bordetella pertussis, Borrelia afzelii, Borrelia burgdorferi, Borrelia garinii, Borrelia hermsii, Borrelia recurrentis, Borrelia species, Brucella species, Brugia malayi, Bunyaviridae family, Burkholderia cepacia, Burkholderia mallei, Burkholderia pseudomallei, Burkholderiaspecies, Caliciviridae family,Campylobacterspecies,Candida albicans, Candidaspecies,Capillaria aerophila, Capillaria hepatica, Capillaria philippinensis, Chlamydia trachomatis, Chlamydia trachomatis, Chlamydia trachomatis, Chlamydophila pneumoniae, Chlamydophila psittaci, Clonorchis sinensis, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, Clostridium perfringens, Clostridium tetani, Clostridiumspecies,Coccidioides immitis, Coccidioides immitis, Coccidioides posadasii, Coccidioides posadasii, Colorado tick fever virus (CTFV), coronaviruses,Corynebacterium diphtheriae, Coxiella burnetii, Coxsackie A virus and Enterovirus 71 (EV71), Crimean-Congo hemorrhagic fever virus,Cryptococcus neoformans, Cryptosporidium species, Cyclospora cayetanensis, Cytomegalovirus, Dengue viruses (DEN-1, DEN-2, DEN-3 or DEN-4),Dientamoeba fragilis, Diphyllobothrium, Dracunculus medinensis, Ebolavirus (EBOV),Echinococcusspecies,Ehrlichia chaffeensis, Ehrlichia ewingii, Ehrlichia species, Entamoeba histolytica, Enterobacteriaceae family, Enterobius vermicularis, Enterococcusspecies, Enterovirus species, Enteroviruses, Entomophthorales order (Entomophthoramycosis),Epidermophyton floccosum, Epstein-Barr Virus (EBV),Escherichia coliO157:H7, Eumycetomaspecies,Fasciola hepaticaandFasciola gigantica, Fasciolopsis buski, Filarioidea superfamily, Flavivirus,Fonsecaea pedrosoi, Francisella tularensis, Fusobacterium species, Geotrichum candidum, Giardia lamblia, Gnathostoma hispidum, Gnathostoma spinigerum, Green algaeDesmodesmus armatus, Group AStreptococcus, Guanarito virus,Haemophilus ducreyi, Haemophilus influenzae, Heartland virus, Helicobacter pylori, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, Hepatitis D Virus, Hepatitis E virus, Herpes simplex virus 1 and 2 (HSV-1 and HSV-2),Histoplasma capsulatum, HIV (Human immunodeficiency virus),Hortaea werneckii, Human bocavirus (HBoV), Human herpesvirus 6 (HHV-6) and Human herpesvirus 7 (HHV-7), Human metapneumovirus (hMPV), Human papillomavirus (HPV), Human parainfluenza viruses (HPIV),Hymenolepis diminuta, Hymenolepis nana, Isospora belli, JC virus, Junin virus,Kingella kingae, Klebsiella granulomatis, Lassa virus, Legionella pneumophila, Leishmaniaspecies,Leptospiraspecies,Listeria monocytogenes, Lymphocytic choriomeningitis virus (LCMV), Machupo virus,Malasseziaspecies, Marburg virus, Measles virus, Measles virus,Metagonimus yokagawai, Microsporidia phylum, Middle East respiratory syndrome coronavirus,Molluscum contagiosumvirus (MCV), Monkeypox virus, Mucorales order (Mucormycosis), Mumps virus,Mycobacterium leprae, Mycobacterium lepromatosis, Mycobacterium tuberculosis, Mycobacterium ulcerans, Mycoplasma pneumoniae, Naegleria fowleri, Necator americanus, Neisseria gonorrhoeae, Neisseria gonorrhoeae, Neisseria meningitidis, Nocardia asteroids, Nocardiaspecies, O111 and O104:H4,Onchocerca volvulus, Opisthorchis felineus, Opisthorchis viverrini, Orthomyxoviridae family, Paracoccidioides brasiliensis, Paragonimus westermani, Paragonimusspecies, parasitic dipterous fly larvae, Parvovirus B19,Pasteurellaspecies,Pediculus humanus capitis, Pediculus humanus corporis, Phthirus pubis, Piedraia hortae, Plasmodiumspecies,Pneumocystis jirovecii, Poliovirus,Prevotellaspecies, PRNP,Propionibacterium propionicus, Rabies virus, Respiratory syncytial virus (RSV),Rhinosporidium seeberi, Rhinovirus, rhinoviruses,Rickettsia, Rickettsia akari, Rickettsia prowazekii, Rickettsia rickettsii, Rickettsia typhi, Rickettsiaspecies, Rift Valley fever virus, Rotavirus, Rubella virus, Sabia,Salmonella entericasubsp.enterica, Salmonella species, Sarcoptes scabiei, SARS coronavirus,Schistosomaspecies, serovartyphi, Shigellaspecies, Sin Nombre virus,Sporothrix schenckii, Staphylococcus, Staphylococcusspecies,Staphylococcusspecies,Streptococcus agalactiae, Streptococcus pneumoniae, Streptococcus pyogenes, Strongyloides stercoralis, Taenia solium, Taenia species, Toxocara canis, Toxocara cati, Toxoplasma gondii, Treponema pallidum, Trichinella spiralis, Trichomonas vaginalis, Trichophyton mentagrophytes, Trichophyton rubrum, Trichophyton rubrum, Trichophyton tonsurans, Trichophyton species, Trichosporon beigelii, Trichuris trichiura, Trypanosoma brucei, Trypanosoma cruzi, Ureaplasma urealyticum, Varicella zoster virus (VZV), Variola major, Variola minor, Venezuelan equine encephalitis virus,Vibrio cholerae, Vibrio parahaemolyticus, Vibrio vulnificus, West Nile virus,Wuchereria bancrofti, Yellow fever virus,Yersinia enterocolitica, Yersinia pestis, Yersinia pseudotuberculosis, or Zika virus, Exemplary antibody molecules include, but are not limited to, 3f8, 8h9, abagovomab, abciximab, abituzumab, abrilumab, actoxumab, adalimumab, adecatumumab, aducanumab, afasevikumab, afelimomab, afutuzumab, alacizumab pegol, ald518, alemtuzumab, alirocumab, altumomab pentetate, amatuximab, anatumomab mafenatox, anetumab ravtansine, anifrolumab, anrukinzumab (ima-638), apolizumab, arcitumomab, ascrinvacumab, aselizumab, atezolizumab, atinumab, atlizumab (tocilizumab), atorolimumab, avelumab, bapineuzumab, basiliximab, bavituximab, bectumomab, begelomab, belimumab, benralizumab, bertilimumab, besilesomab, bevacizumab, bezlotoxumab, biciromab, bimagrumab, bimekizumab, bivatuzumab mertansine, bleselumab, blinatumomab, blontuvetmab, blosozumab, bococizumab, brazikumab, brentuximab vedotin, briakinumab, brodalumab, brolucizumab, brontictuzumab, cabiralizumab, canakinumab, cantuzumab mertansine, cantuzumab ravtansine, caplacizumab, capromab pendetide, carlumab, carotuximab, catumaxomab, cbr96-doxorubicin immunoconjugate, cedelizumab, cergutuzumab amunaleukin, certolizumab pegol, cetuximab, ch.14.18, citatuzumab bogatox, cixutumumab, clazakizumab, clenoliximab, clivatuzumab tetraxetan, codrituzumab, coltuximab ravtansine, conatumumab, concizumab, crenezumab, crotedumab, cr6261, dacetuzumab, daclizumab, dalotuzumab, dapirolizumab pegol, daratumumab, dectrekumab, demcizumab, denintuzumab mafodotin, denosumab, derlotuximab biotin, detumomab, dinutuximab, diridavumab, domagrozumab, dorlimomab aritox, drozitumab, duligotumab, dupilumab, durvalumab, dusigitumab, ecromeximab, eculizumab, edobacomab, edrecolomab, efalizumab, efungumab, eldelumab, elgemtumab, elotuzumab, elsilimomab, emactuzumab, emibetuzumab, emicizumab, enavatuzumab, enfortumab vedotin, enlimomab pegol, enoblituzumab, enokizumab, enoticumab, ensituximab, epitumomab cituxetan, epratuzumab, erenumab, erlizumab, ertumaxomab, etaracizumab, etrolizumab, evinacumab, evolocumab, exbivirumab, fanolesomab, faralimomab, farletuzumab, fasinumab, fbta05, felvizumab, fezakinumab, fibatuzumab, ficlatuzumab, figitumumab, firivumab, flanvotumab, fletikumab, fontolizumab, foralumab, foravirumab, fresolimumab, fulranumab, futuximab, galcanezumab, galiximab, ganitumab, gantenerumab, gavilimomab, gemtuzumab ozogamicin, gevokizumab, girentuximab, glembatumumab vedotin, golimumab, gomiliximab, guselkumab, ibalizumab, ibritumomab tiuxetan, icrucumab, idarucizumab, igovomab, imab362, imalumab, imciromab, imgatuzumab, inclacumab, indatuximab ravtansine, indusatumab vedotin, inebilizumab, infliximab, intetumumab, inolimomab, inotuzumab ozogamicin, ipilimumab, iratumumab, isatuximab, itolizumab, ixekizumab, keliximab, labetuzumab, lambrolizumab, lampalizumab, lanadelumab, landogrozumab, laprituximab emtansine, lebrikizumab, lemalesomab, lendalizumab, lenzilumab, lerdelimumab, lexatumumab, libivirumab, lifastuzumab vedotin, ligelizumab, lilotomab satetraxetan, lintuzumab, lirilumab, lodelcizumab, lokivetmab, lorvotuzumab mertansine, lucatumumab, lulizumab pegol, lumiliximab, lumretuzumab, mapatumumab, margetuximab, maslimomab, mavrilimumab, matuzumab, mepolizumab, metelimumab, milatuzumab, minretumomab, mirvetuximab soravtansine, mitumomab, mogamulizumab, monalizumab, morolimumab, motavizumab, moxetumomab pasudotox, muromonab-cd3, nacolomab tafenatox, namilumab, naptumomab estafenatox, naratuximab emtansine, narnatumab, natalizumab, navicixizumab, navivumab, nebacumab, necitumumab, nemolizumab, nerelimomab, nesvacumab, nimotuzumab, nivolumab, nofetumomab merpentan, obiltoxaximab, obinutuzumab, ocaratuzumab, ocrelizumab, odulimomab, ofatumumab, olaratumab, olokizumab, omalizumab, onartuzumab, ontuxizumab, opicinumab, oportuzumab monatox, oregovomab, orticumab, otelixizumab, otlertuzumab, oxelumab, ozanezumab, ozoralizumab, pagibaximab, palivizumab, pamrevlumab, panitumumab, pankomab, panobacumab, parsatuzumab, pascolizumab, pasotuxizumab, pateclizumab, patritumab, pembrolizumab, pemtumomab, perakizumab, pertuzumab, pexelizumab, pidilizumab, pinatuzumab vedotin, pintumomab, placulumab, plozalizumab, pogalizumab, polatuzumab vedotin, ponezumab, prezalizumab, priliximab, pritoxaximab, pritumumab, pro 140, quilizumab, racotumomab, radretumab, rafivirumab, ralpancizumab, ramucirumab, ranibizumab, raxibacumab, refanezumab, regavirumab, reslizumab, rilotumumab, rinucumab, risankizumab, rituximab, rivabazumab pegol, robatumumab, roledumab, romosozumab, rontalizumab, rovalpituzumab tesirine, rovelizumab, ruplizumab, sacituzumab govitecan, samalizumab, sapelizumab, sarilumab, satumomab pendetide, secukinumab, seribantumab, setoxaximab, sevirumab, sibrotuzumab, sgn-cd19a, sgn-cd33a, sifalimumab, siltuximab, simtuzumab, siplizumab, sirukumab, sofituzumab vedotin, solanezumab, solitomab, sonepcizumab, sontuzumab, stamulumab, sulesomab, suvizumab, tabalumab, tacatuzumab tetraxetan, tadocizumab, talizumab, tamtuvetmab, tanezumab, taplitumomab paptox, tarextumab, tefibazumab, telimomab aritox, tenatumomab, teneliximab, teplizumab, teprotumumab, tesidolumab, tetulomab, tezepelumab, tgn1412, ticilimumab (tremelimumab), tildrakizumab, tigatuzumab, timolumab, tisotumab vedotin, tnx-650, tocilizumab (atlizumab), toralizumab, tosatoxumab, tositumomab, tovetumab, tralokinumab, trastuzumab, trastuzumab emtansine, trbs07, tregalizumab, tremelimumab, trevogrumab, tucotuzumab celmoleukin, tuvirumab, ublituximab, ulocuplumab, urelumab, urtoxazumab, ustekinumab, utomilumab, vadastuximab talirine, vandortuzumab vedotin, vantictumab, vanucizumab, vapaliximab, varlilumab, vatelizumab, vedolizumab, veltuzumab, vepalimomab, vesencumab, visilizumab, vobarilizumab, volociximab, vorsetuzumab mafodotin, votumumab, xentuzumab, zalutumumab, zanolimumab, zatuximab, ziralimumab, zolimomab aritox, or derivative thereof. In an embodiment, the antibody molecule comprises one, two, or three CDRs of the VH region of an antibody molecule described herein, using the Kabat or Chothia definitions of CDRs. In an embodiment, the antibody molecule comprises one, two, or three CDRs of the VL region of an antibody molecule described herein, using the Kabat or Chothia definitions of CDRs. In an embodiment, the antibody molecule comprises one or more (e.g., two or three) CDRs of the VH region and/or one or more (e.g., two or three) CDRs of the VL region of an antibody molecule described herein, using the Kabat or Chothia definitions of CDRs. In an embodiment, the antibody molecule comprises one, two, three, or four frameworks of the VH region of an antibody molecule described herein. In an embodiment, the antibody molecule comprises one, two, three, or four frameworks of the VL region of an antibody molecule described herein. In an embodiment, the antibody molecule comprises one or more (e.g., two, three, or four) frameworks of the VH region and/or one or more (e.g., two, three, or four) frameworks of the VL region of an antibody molecule described herein. In an embodiment, the antibody molecule comprises a heavy chain variable region of an antibody molecule described herein, or a heavy chain variable region having an amino acid sequence substantially identical thereto (e.g., an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues). In an embodiment, the antibody molecule comprises a light chain variable region of an antibody molecule described herein, or a light chain variable region having an amino acid sequence substantially identical thereto (e.g., an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues). In an embodiment, the antibody molecule comprises a heavy chain variable region, or a heavy chain variable region having an amino acid sequence substantially identical thereto (e.g., an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues), and a light chain variable region of an antibody molecule described herein, or a light chain variable region having an amino acid sequence substantially identical thereto (e.g., an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues). In an embodiment, the antibody molecule further comprises a heavy chain constant region, e.g., a heavy chain constant region of an antibody molecule described herein, or a heavy chain constant region having an amino acid sequence substantially identical thereto (e.g., an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues). In an embodiment, the antibody molecule further comprises a light chain constant region, e.g., a light chain constant region of an antibody molecule described herein, or a light chain constant region having an amino acid sequence substantially identical thereto (e.g., an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues). In an embodiment, the antibody molecule further comprises a heavy chain constant region, or a heavy chain constant region having an amino acid sequence substantially identical thereto (e.g., an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues), and a light chain constant region, or a light chain constant region having an amino acid sequence substantially identical thereto (e.g., an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues). In an embodiment, the antibody molecule comprises a heavy chain constant region, a light chain constant region, and heavy and light chain variable regions of an antibody molecule, as described herein. The antibody molecules described herein can have several advantageous properties, including, but limited to, a desired (e.g., increased) half-life. For example, the antibody molecules can be used to effectively treat, prevent or diagnose a disorder described herein. In an embodiment, the antibody molecule is capable of binding to a target molecule or cell. For example, the engineered antibody molecule is capable of binding to the target molecule or cell, with the same, or substantially the same, binding specificity and/or affinity, as compared to the parental antibody molecule. In an embodiment, the antibody molecule binds to the target molecule or cell with high affinity, e.g., with a dissociation constant (Kd) of less than about 100 nM, typically about 10 nM, and more typically, about 10-0.01 nM, about 5-0.01 nM, about 3-0.05 nM, about 1-0.1 nM, or stronger, e.g., less than about 80, 70, 60, 50, 40, 30, 20, 10, 8, 6, 4, 3, 2, 1, 0.5, 0.2, 0.1, 0.05, or 0.01 nM. In an embodiment, the antibody molecule binds to the target molecule or cell with a Koffslower than 1×10−4, 5×10−5, or 1×10−5s−1. In an embodiment, the antibody molecule binds to the target molecule or cell with a Konfaster than 1×104, 5×104, 1×105, or 5×105M−1s−1. In an embodiment, the antibody molecule is capable of inhibiting or activating a biological function of a target molecule or cell. For example, the engineered antibody molecule is capable of inhibiting or activating a biological function of the target molecule or cell, with the same, or substantially the same, level of effectiveness, as compared to the parental antibody molecule, e.g., as determined by IC50, EC50, or LD50. In an embodiment, the antibody molecule is capable of binding to an epitope on a target molecule or cell. For example, the engineered antibody molecule is capable of binding to the same, or substantially the same, epitope on the target molecule or cell, as compared to the parental antibody molecule. Exemplary Fusion Proteins The methods described herein can be used to engineer a variety of fusion proteins, e.g., any Fc fusion protein containing an Fc region. Exemplary Fc fusion proteins include, but are not limited to, a CTLA-4 Fc fusion protein (e.g., belatacept or abatacept), a vascular endothelial growth factor receptor (VEGFR) Fc fusion protein (e.g., a VEGFR1/VEGFR2 Fc fusion protein, e.g., aflibercept or KH902), an IL-1R Fc fusion protein (e.g., (rilonacept), a thrombopoietin-binding peptide Fc fusion protein (e.g., romiplostim), an LFA-3 Fc fusion protein (e.g., alefacept), an anti-CD40L Fc fusion protein (e.g., a dimeric fusion protein comprising the C-terminus of the domain antibody (dAb) targeting the CD40 ligand (CD40L or CD154) linked to an Fc fragment of IgG1, e.g., BMS-986004 or letolizumab), an TNF receptor (TNFR) Fc fusion protein (e.g., a recombinant TNF receptor 2 (TNFR2) fused to an IgG1 Fc domain, e.g., OPRX-106 or etanercept), a coagulation Factor VIII-Fc fusion protein (e.g., BIIB031, efraloctocog-α, or rFVIIIFc), a coagulation Factor IX-Fc Fusion Protein (e.g., BIIB029 or eftrenonacog-α), a Factor IX Fc fusion protein (e.g., rFIXFc), a granulocyte colony-stimulating factor Fc fusion protein (e.g., F-627), a follicle stimulating hormone (FSH) Fc fusion protein (e.g., KN015), an activin type 2B receptor Fc fusion protein (e.g., STM 434), an activin receptor-like kinase 1 (ALK-1) inhibitor receptor Fc fusion protein (e.g., dalantercept), an RNase Fc fusion (e.g., RSLV-132), an anti-angiopoietin peptibody (e.g., a peptide with angiopoietin-binding properties that is fused to the Fc region, e.g., AMG 386), a tissue nonspecific alkaline phosphatase (TNSALP) Fc fusion protein (e.g., asfotase alfa or ENB-0040), a CD24 Fc fusion protein, a BAFF-Fc fusion protein (e.g., blisibimod), a GLP1 peptide analog Fc fusion protein (e.g., dulaglutide or LY2189265), an erythropoietin-mimetic peptide Fc fusion protein (e.g., an erythropoietin-mimetic peptide-IgG1 Fc mimetibody (e.g., CNTO 528), or an erythropoietin-mimetic peptide-IgG4 Fc fusion protein mimetibody (e.g., CNTO 530)), or a CD95 Fc fusion (e.g., APG 101 or apocept). Animal Models The polypeptides (e.g., antibody molecules or fusion proteins) described herein can be evaluated in vivo, e.g., using various animal models. For example, an animal model can be used to test the efficacy of a polypeptide (e.g., antibody molecule or fusion proteins) described herein in modulating a biological function of a target molecule or cell. As another example, an animal model can also be used to test the efficacy of a polypeptide (e.g., antibody molecule) described herein in in treating, preventing, or diagnosing a disorder described herein. Animal models can also be used, e.g., to investigate for side effects, measure concentrations of antibody molecules in situ, demonstrate correlations between a function of a target molecule or cell and a disorder described herein. Exemplary animal models for other disorders described herein are also known in the art. Exemplary types of animals that can be used to evaluate the antibody molecules described herein include, but are not limited to, mice, rats, rabbits, guinea pigs, and monkeys. Non-human primates and transgenic mice expressing human FcRn are typically used as the model of choice for PK analysis (Avery et al.MAbs.2016; 8(6):1064-78; Fan et al.MAbs.2016; 8(5):848-53; Tam et al.MAbs.2013; 5(3):397-405). For example, humanized FcRn mice can be established on the C57BL/6J background in a sequential manner, including the creation of a mouse strain carrying a deletion in the mouse FcRn gene, followed by introduction of the human FcRn gene. Exemplary mouse lines include, e.g., Tg276 and Tg32 (The Jackson Laboratory stock number 004919 and 014565). With further backcrossing and sequential alterations, additional lines can be made. Exemplary mouse models that can be used to evaluate the polypeptides described herein include, but are not limited to, FcRn-null mice, humanized Tg276 FcRn mice (e.g., B6.Cg-Fcgrt<tm1Dcr>Tg(CAG-FCGRT)276Dcr/DcrJ with the Jackson Laboratory stock number 004919), humanized Tg32 FcRn mice (e.g., B6.Cg-Fcgrt<tm1Dcr>Tg(FCGRT)32Dcr/DcrJ with the Jackson Laboratory stock number 014565), immunodeficient hFcRn mice (e.g., B6.Cg-Fcgrt<tm1Dcr>Prkdc<scid>Tg(CAG-FCGRT)276Dcr/DcrJ with the Jackson Laboratory stock number 021146), B6.Cg-Fcgrt<tm1Dcr>Prkdc <scid>Tg(FCGRT)32Dcr/DcrJ with the Jackson Laboratory stock number 018441, and B6.Cg-Rag1<tm>Mom<Fcgrt>tm1Dcr[Tg(CAG-FCGRT)276Dcr/DcrJ with the Jackson Laboratory stock number 16919), e.g., as described in Proetzel et al.BioDrugs.2014; 28(2): 171-180. Pharmaceutical Compositions and Kits In some aspects, this disclosure provides compositions, e.g., pharmaceutically acceptable compositions, which include a polypeptide (e.g., an antibody molecule or fusion protein) described herein, formulated together with a pharmaceutically acceptable carrier. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, isotonic and absorption delaying agents, and the like that are physiologically compatible. The carrier can be suitable for intravenous, intramuscular, subcutaneous, parenteral, rectal, spinal or epidermal administration (e.g., by injection or infusion). In certain embodiments, less than about 5%, e.g., less than about 4%, 3%, 2%, or 1% of the antibody molecules in the pharmaceutical composition are present as aggregates. In other embodiments, at least about 95%, e.g., at least about 96%, 97%, 98%, 98.5%, 99%, 99.5%, 99.8%, or more of the antibody molecules in the pharmaceutical composition are present as monomers. In some embodiments, the level of aggregates or monomers is determined by chromatography, e.g., high performance liquid chromatography size exclusion chromatography (HPLC-SEC). The compositions set out herein may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, liposomes, and suppositories. A suitable form depends on the intended mode of administration and therapeutic application. Typical suitable compositions are in the form of injectable or infusible solutions. One suitable mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). In an embodiment, the polypeptide (e.g., antibody molecule or fusion proteins) is administered by intravenous infusion or injection. In another embodiment, the polypeptide (e.g., antibody molecule or fusion proteins) is administered by intramuscular or subcutaneous injection. The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Therapeutic compositions typically should be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high antibody concentration. Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody or antibody portion) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin. The polypeptides (e.g., antibody molecules or fusion proteins) described herein can be administered by a variety of methods. Several are known in the art, and for many therapeutic, prophylactic, or diagnostic applications, an appropriate route/mode of administration is intravenous injection or infusion. For example, the antibody molecules can be administered by intravenous infusion at a rate of less than 10 mg/min; preferably less than or equal to 5 mg/min to reach a dose of about 1 to 100 mg/m2, preferably about 5 to 50 mg/m2, about 7 to 25 mg/m2and more preferably, about 10 mg/m2. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. In certain embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g.,Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. In certain embodiments, a polypeptide (e.g., an antibody molecule or fusion protein) can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The antibody molecule (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the antibody molecule may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a polypeptide (e.g., an antibody molecule) by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. Therapeutic, prophylactic, or diagnostic compositions can also be administered with medical devices, and several are known in the art. Dosage regimens are adjusted to provide the desired response (e.g., a therapeutic, prophylactic, or diagnostic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the antibody molecule and the particular therapeutic, prophylactic, or diagnostic effect to be achieved, and (b) the limitations inherent in the art of compounding such an antibody molecule for the treatment of sensitivity in individuals. An exemplary, non-limiting range for a therapeutically, prophylactically, or diagnostically effective amount of an antibody molecule is about 0.1-50 mg/kg, e.g., about 0.1-30 mg/kg, e.g., about 1-30, 1-15, 1-10, 1-5, 5-10, or 1-3 mg/kg, e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 mg/kg. The antibody molecule can be administered by intravenous infusion at a rate of less than 10 mg/min, e.g., less than or equal to 5 mg/min to reach a dose of about 1 to 100 mg/m2, e.g., about 5 to 50 mg/m2, about 7 to 25 mg/m2, e.g., about 10 mg/m2. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions. The pharmaceutical compositions herein may include a “therapeutically effective amount,” “prophylactically effective amount,” or “diagnostically effectively amount” of a polypeptide (e.g., an antibody molecule) described herein. A “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the polypeptide (e.g., antibody molecule or fusion protein) may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effect of the antibody molecule is outweighed by the therapeutically beneficial effects. A “therapeutically effective dosage” typically inhibits a measurable parameter by at least about 20%, e.g., by at least about 40%, by at least about 60%, or by at least about 80% relative to untreated subjects. The measurable parameter may be, e.g., hematuria, colored urine, foamy urine, pain, swelling (edema) in the hands and feet, or high blood pressure. The ability of an antibody molecule to inhibit a measurable parameter can be evaluated in an animal model system predictive of efficacy in treating or preventing a disorder described herein. Alternatively, this property of a composition can be evaluated by examining the ability of the polypeptide (e.g., antibody molecule or fusion proteins) to modulate a biological function of a target molecule or cell, e.g., by an in vitro assay. A “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount. A “diagnostically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired diagnostic result. Typically, a diagnostically effective amount is one in which a disorder, e.g., a disorder described herein, can be diagnosed in vitro, ex vivo, or in vivo. Also within this disclosure is a kit that comprises a polypeptide (e.g., an antibody molecule or fusion protein), described herein. The kit can include one or more other elements including: instructions for use; other reagents, e.g., a label, a therapeutic agent, or an agent useful for chelating, or otherwise coupling, an antibody molecule to a label or therapeutic agent, or a radioprotective composition; devices or other materials for preparing the polypeptide (e.g., antibody molecule or fusion protein) for administration; pharmaceutically acceptable carriers; and devices or other materials for administration to a subject. Nucleic Acids The present disclosure also features nucleic acids comprising nucleotide sequences that encode polypeptides (e.g., antibody molecules or fusion proteins), e.g., Fc regions of the polypeptides, as described herein. For example, the present disclosure features a nucleic acid encoding an Fc region described herein, e.g., an Fc region comprising one or more of the mutations described herein. The Fc region can be engineered from an Fc region of an existing polypeptide (e.g., an antibody molecule or fusion protein) described herein. The nucleic acid can comprise a nucleotide sequence encoding an amino acid sequence of an Fc region of a polypeptide (e.g., antibody molecule or fusion protein) described herein, or a nucleotide sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In an embodiment, the nucleic acid further comprises a nucleotide sequence encoding a heavy chain variable region of a polypeptide (e.g., an antibody molecule or fusion protein) described herein, or having a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In another embodiment, the nucleic acid further comprises a nucleotide sequence encoding a light chain variable region of a polypeptide (e.g., an antibody molecule or fusion protein) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In yet another embodiment, the nucleic acid further comprises a nucleotide sequence encoding a heavy chain variable region and a light chain variable region of a polypeptide (e.g., an antibody molecule or fusion protein) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In an embodiment, the nucleic acid further comprises a nucleotide sequence encoding at least one, two, or three CDRs from a heavy chain variable region of a polypeptide (e.g., an antibody molecule or fusion protein) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In another embodiment, the nucleic acid further comprises a nucleotide sequence encoding at least one, two, or three CDRs from a light chain variable region of a polypeptide (e.g., an antibody molecule or fusion protein) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In yet another embodiment, the nucleic acid comprises a nucleotide sequence encoding at least one, two, three, four, five, or six CDRs from heavy and light chain variable regions of a polypeptide (e.g., an antibody molecule or fusion protein) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In an embodiment, the nucleic acid comprises a portion of a nucleotide sequence described herein. The portion may encode, for example, an Fc region, a variable region (e.g., VH or VL); one, two, or three or more (e.g., four, five, or six) CDRs; or one, two, three, or four or more framework regions. The nucleic acids disclosed herein include deoxyribonucleotides or ribonucleotides, or analogs thereof. The polynucleotide may be either single-stranded or double-stranded, and if single-stranded may be the coding strand or non-coding (antisense) strand. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. The nucleic acid may be a recombinant polynucleotide, or a polynucleotide of genomic, cDNA, semisynthetic, or synthetic origin which either does not occur in nature or is linked to another polynucleotide in a non-natural arrangement. In some aspects, the application features host cells and vectors containing the nucleic acids described herein. The nucleic acids may be present in a single vector or separate vectors present in the same host cell or separate host cell, as described in more detail below. Vectors The present disclosure features vectors that comprise nucleotide sequences encoding polypeptides (e.g., an antibody molecules or fusion proteins), e.g., Fc regions of the polypeptides, as described herein. For example, the present disclosure features a vector comprising a nucleotide sequence encoding an Fc region described herein, e.g., an Fc region comprising one or more of the mutations described herein. The Fc region can be engineered from an Fc region of an existing polypeptide (e.g., an antibody molecule or fusion protein) described herein. The vector can comprise a nucleotide sequence encoding an amino acid sequence of an Fc region of a polypeptide (e.g., antibody molecule or fusion protein) described herein, or a nucleotide sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). The vectors include, but are not limited to, a virus, plasmid, cosmid, lambda phage or a yeast artificial chromosome (YAC). Numerous vector systems can be employed. For example, one class of vectors utilizes DNA elements which are derived from animal viruses such as, for example, bovine papilloma virus, polyoma virus, adenovirus, vaccinia virus, baculovirus, retroviruses (Rous Sarcoma Virus, MMTV or MOMLV) or SV40 virus. Another class of vectors utilizes RNA elements derived from RNA viruses such as Semliki Forest virus, Eastern Equine Encephalitis virus and Flaviviruses. Additionally, cells which have stably integrated the DNA into their chromosomes may be selected by introducing one or more markers which allow for the selection of transfected host cells. The marker may provide, for example, prototropy to an auxotrophic host, biocide resistance (e.g., antibiotics), or resistance to heavy metals such as copper, or the like. The selectable marker gene can be either directly linked to the DNA sequences to be expressed, or introduced into the same cell by cotransformation. Additional elements may also be needed for optimal synthesis of mRNA. These elements may include splice signals, as well as transcriptional promoters, enhancers, and termination signals. Once the expression vector or DNA sequence containing the constructs has been prepared for expression, the expression vectors may be transfected or introduced into an appropriate host cell. Various techniques may be employed to achieve this, such as, for example, protoplast fusion, calcium phosphate precipitation, electroporation, retroviral transduction, viral transfection, gene gun, lipid based transfection or other conventional techniques. In the case of protoplast fusion, the cells are grown in media and screened for the appropriate activity. Methods and conditions for culturing the resulting transfected cells and for recovering the polypeptide (e.g., antibody molecule) produced are known to those skilled in the art, and may be varied or optimized depending upon the specific expression vector and mammalian host cell employed, based upon the present description. Cells The present disclosure also provides host cells comprising a nucleic acid encoding a polypeptide (e.g., an antibody molecule or fusion protein) as described herein. The polypeptide (e.g., antibody molecule or fusion protein) can be engineered in accordance with a method described herein. For example, the host cells may comprise a nucleic acid molecule having a nucleotide sequence of a polypeptide described herein (e.g., an antibody molecule or fusion protein described herein), a sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein), or a portion of one of said nucleic acids. In some embodiments, the host cells are genetically engineered to comprise nucleic acids encoding the polypeptide (e.g., antibody molecule or fusion protein) described herein. In certain embodiments, the host cells are genetically engineered by using an expression cassette. The phrase “expression cassette,” refers to nucleotide sequences, which are capable of affecting expression of a gene in hosts compatible with such sequences. Such cassettes may include a promoter, an open reading frame with or without introns, and a termination signal. Additional factors necessary or helpful in effecting expression may also be used, such as, for example, an inducible promoter. The disclosure also provides host cells comprising the vectors described herein. The cell can be, but is not limited to, a eukaryotic cell, a bacterial cell, an insect cell, or a human cell. Suitable eukaryotic cells include, but are not limited to, Vero cells, HeLa cells, COS cells, CHO cells, HEK293 cells, BHK cells and MDCKII cells. Suitable insect cells include, but are not limited to, Sf9 cells. Uses of Polypeptides The polypeptides (e.g., antibody molecules or fusion proteins) disclosed herein, as well as the pharmaceutical compositions disclosed herein, have in vitro, ex vivo, and in vivo therapeutic, prophylactic, and/or diagnostic utilities. In an embodiment, the polypeptide (e.g., antibody molecule or fusion protein) modulates (e.g., reduces (e.g., inhibits, blocks, or neutralizes) or increases (e.g., activates, initiates, or enhances)) one or more biological activities of a target molecule (e.g., protein) or cell. For example, these polypeptides (e.g., antibody molecules or fusion proteins) can be administered to cells in culture, in vitro or ex vivo, or to a subject, e.g., a human subject, e.g., in vivo, to modulate one or more biological activities of the target molecule or cell. Accordingly, in an aspect, the disclosure provides a method of treating, preventing, or diagnosing a disorder, e.g., a disorder described herein, in a subject, comprising administering to the subject a polypeptide (e.g., an antibody molecule or fusion protein) described herein, such that the disorder is treated, prevented, or diagnosed. For example, the disclosure provides a method comprising contacting the polypeptide (e.g., antibody molecule or fusion protein) described herein with cells in culture, e.g. in vitro or ex vivo, or administering the polypeptide (e.g., antibody molecule or fusion protein) described herein to a subject, e.g., in vivo, to treat, prevent, or diagnose a disorder, e.g., a disorder associated with a target molecule or cell (e.g., a disorder described herein). As used herein, the term “subject” is intended to include human and non-human animals. In some embodiments, the subject is a human subject, e.g., a human patient having a disorder described herein, or at risk of having a disorder described herein. The term “non-human animals” includes mammals and non-mammals, such as non-human primates. In an embodiment, the subject is a human. The methods and compositions described herein are suitable for treating human patients for a disorder described herein. Patients having a disorder described herein include those who have developed a disorder described herein but are (at least temporarily) asymptomatic, patients who have exhibited a symptom of a disorder described herein, or patients having a disorder related to or associated with a disorder described herein. Methods of Treating or Preventing Disorders The polypeptides (e.g., antibody molecules or fusion proteins) described herein can be used to treat or prevent disorders or conditions. In an embodiment, the polypeptide has an optimal or improved half-life, which can be desirable for treating or preventing the disorder or condition. While not wishing to be bound by theory, it is believed that in an embodiment, the polypeptide described herein (e.g., the polypeptide having an optimal or improved half-life) can provide one or more benefits over another polypeptide having the same or similar binding affinity and/or specificity (e.g., a polypeptide that does not have, or has not been engineered to have, an optimal or improved half-life). These benefits can include, but are not limited to, an increased therapeutic or preventive efficacy, a reduced dosage regimen, or an improved pharmacokinetic property. In an embodiment, the polypeptide includes a mutated Fc region as described herein. Exemplary disorders or conditions that can be treated or prevented by the polypeptides described herein include, but are not limited to, a cancer (e.g., a solid tumor or a hematologic cancer), an infectious disease (e.g., a bacterial infection or a viral infection), an immune disorder (e.g., an autoimmune disorder), an inflammatory disorder, a metabolic disorder (e.g., diabetes), a cardiovascular disorder, an organ transplant rejection. In an embodiment, the disorder is a chronic disorder. Exemplary cancers that can be treated or prevented by the polypeptides described herein include, but are not limited to, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), adrenocortical carcinoma, Kaposi sarcoma, an AIDS-related lymphoma, primary central nervous system (CNS) lymphoma, anal cancer, appendix cancer, astrocytoma, atypical teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer (e.g., Ewing sarcoma or osteosarcoma and malignant fibrous histiocytoma), brain tumor (e.g., astrocytomas, brain stem glioma, central nervous system atypical teratoid/rhabdoid tumor, central nervous system embryonal tumor, central nervous system germ cell tumor, craniopharyngioma, or ependymoma), breast cancer, bronchial tumor, Burkitt lymphoma, carcinoid tumor (e.g., gastrointestinal carcinoid tumor), cardiac (heart) tumor, embryonal tumor, germ cell tumor, lymphoma, cervical cancer, cholangiocarcinoma, chordoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myeloproliferative neoplasm, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, ductal carcinoma in situ (DCIS), endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, Ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer (e.g., intraocular melanoma or retinoblastoma), fallopian tube cancer, fibrous histiocytoma of bone, osteosarcoma, gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor (e.g., central nervous system tumor, extracranial tumor, extragonadal tumor, ovarian cancer, or testicular cancer), gestational trophoblastic disease, glioma, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumor, pancreatic neuroendocrine tumor, Kaposi sarcoma, kidney cancer (e.g., renal cell cancer or Wilms tumor), Langerhans cell histiocytosis (LCH), laryngeal cancer, leukemia (e.g., acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), or hairy cell leukemia), lip and oral cavity cancer, liver cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC) or small cell lung cancer), lymphoma (e.g., aids-related, Burkitt lymphoma, cutaneous T-cell lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, or primary central nervous system (CNS) lymphoma), Waldenström macroglobulinemia, male breast cancer, malignant fibrous histiocytoma of bone and osteosarcoma, melanoma (e.g., intraocular (eye) melanoma), Merkel cell carcinoma, mesothelioma, metastatic squamous neck cancer, midline tract carcinoma, mouth cancer, multiple endocrine neoplasia syndrome, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndrome, myelodysplastic/myeloproliferative neoplasm, chronic myeloproliferative neoplasm, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, oral cancer, lip and oral cavity cancer, oropharyngeal cancer, osteosarcoma and malignant fibrous histiocytoma of bone, ovarian cancer (e.g., epithelial ovarian cancer or germ cell ovarian tumor), pancreatic cancer, pancreatic neuroendocrine tumors (islet cell tumors), papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pituitary tumor, pleuropulmonary blastoma, peritoneal cancer, prostate cancer, rectal cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma (e.g., Ewing sarcoma, Kaposi sarcoma, osteosarcoma, rhabdomyosarcoma, soft tissue sarcoma, or uterine sarcoma), Sézary syndrome, skin cancer (e.g., melanoma, Merkel cell carcinoma, or nonmelanoma skin cancer), small intestine cancer, squamous cell carcinoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, urethral cancer, endometrial uterine cancer, vaginal cancer, vulvar cancer, or a metastatic lesion thereof. Exemplary infectious diseases that can be treated or prevented by the polypeptides described herein include, but are not limited to,Acinetobacterinfections, actinomycosis, African sleeping sickness (African trypanosomiasis), AIDS (acquired immunodeficiency syndrome), amebiasis, anaplasmosis, angiostrongyliasis, anisakiasis, anthrax,arcanobacterium haemolyticuminfection, argentine hemorrhagic fever, ascariasis, aspergillosis, astrovirus infection, babesiosis,Bacillus cereusinfection, bacterial pneumonia, bacterial vaginosis,bacteroidesinfection, balantidiasis, bartonellosis,Baylisascarisinfection, bk virus infection, black piedra, blastocystosis, blastomycosis, bolivian hemorrhagic fever, botulism (and infant botulism), brazilian hemorrhagic fever, brucellosis, bubonic plague,burkholderiainfection, buruli ulcer, calicivirus infection (norovirus and sapovirus), campylobacteriosis, candidiasis (moniliasis; thrush), capillariasis, carrion's disease, cat-scratch disease, cellulitis, chagas disease (american trypanosomiasis), chancroid, chickenpox, chikungunya,chlamydia, chlamydophila pneumoniaeinfection (taiwan acute respiratory agent or twar), cholera, chromoblastomycosis, chytridiomycosis, clonorchiasis,Clostridium difficilecolitis, coccidioidomycosis, colorado tick fever (CTF), common cold (Acute viral rhinopharyngitis; Acute coryza), Creutzfeldt-Jakob disease (CJD), Crimean-Congo hemorrhagic fever (CCHF), cryptococcosis, cryptosporidiosis, cutaneous larva migrans (CLM), cyclosporiasis, cysticercosis, cytomegalovirus infection, dengue fever, desmodesmus infection, dientamoebiasis, diphtheria, diphyllobothriasis, dracunculiasis, ebola hemorrhagic fever, echinococcosis, ehrlichiosis, enterobiasis (pinworm infection),enterococcusinfection, enterovirus infection, epidemic typhus, erythema infectiosum (fifth disease), exanthem subitum (sixth disease), fasciolasis, fasciolopsiasis, fatal familial insomnia (FFI), filariasis, food poisoning byClostridium perfringens, free-living amebic infection,fusobacteriuminfection, gas gangrene (clostridial myonecrosis), geotrichosis, gerstmann-straussler-scheinker syndrome (GSS), giardiasis, glanders, gnathostomiasis, gonorrhea, granuloma inguinale (donovanosis), Group A streptococcal infection, Group B streptococcal infection,Haemophilus influenzaeinfection, hand, foot and mouth disease (HFMD), Hantavirus Pulmonary Syndrome (HPS), heartland virus disease,Helicobacter pyloriinfection, hemolytic-uremic syndrome (HUS), hemorrhagic fever with renal syndrome (HFRS), hepatitis A, hepatitis B, hepatitis C, hepatitis D, hepatitis E, herpes simplex, histoplasmosis, hookworm infection, human bocavirus infection, humanewingiiehrlichiosis, human granulocytic anaplasmosis (HGA), human metapneumovirus infection, Human monocytic ehrlichiosis, human papillomavirus (HPV) infection, Human parainfluenza virus infection, Hymenolepiasis, Epstein-Barr Virus Infectious Mononucleosis (Mono), influenza (flu), isosporiasis, kawasaki disease, keratitis, kingella kingae infection, kuru, lassa fever, legionellosis (legionnaires' disease), legionellosis (pontiac fever), leishmaniasis, leprosy, leptospirosis, listeriosis, lyme disease (lyme borreliosis), lymphatic filariasis (Elephantiasis), Lymphocytic choriomeningitis, Malaria, Marburg hemorrhagic fever (MHF), Measles, Middle East respiratory syndrome (MERS), melioidosis (Whitmore's disease), meningitis, meningococcal disease, metagonimiasis, microsporidiosis, molluscum contagiosum (MC), Monkeypox, Mumps, Murine typhus (Endemic typhus),Mycoplasmapneumonia, Mycetoma (disambiguation), Myiasis, Neonatal conjunctivitis (Ophthalmia neonatorum), (New) Variant Creutzfeldt-Jakob disease (vCJD, nvCJD), nocardiosis, onchocerciasis (River blindness), opisthorchiasis, paracoccidioidomycosis (South American blastomycosis), paragonimiasis, pasteurellosis, pediculosis capitis (head lice), pediculosis corporis (body lice), pediculosis pubis (pubic lice, crab lice), pelvic inflammatory disease (PID), pertussis (Whooping cough), plague, pneumococcal infection,pneumocystispneumonia (PCP), pneumonia, poliomyelitis,prevotellainfection, primary amoebic meningoencephalitis (PAM), progressive multifocal leukoencephalopathy, psittacosis, Q fever, rabies, relapsing fever, respiratory syncytial virus infection, rhinosporidiosis, rhinovirus infection, rickettsial infection, rickettsialpox, Rift Valley fever (RVF), Rocky Mountain spotted fever (RMSF), rotavirus infection, rubella, salmonellosis, SARS (Severe Acute Respiratory Syndrome), scabies, schistosomiasis, sepsis, shigellosis (Bacillary dysentery), shingles (Herpes zoster), smallpox (Variola), sporotrichosis, staphylococcal food poisoning, staphylococcal infection, strongyloidiasis, subacute sclerosing panencephalitis, syphilis, Taeniasis, Tetanus (Lockjaw), Tinea barbae (Barber's itch), Tinea capitis (Ringworm of the Scalp), Tinea corporis (Ringworm of the Body), Tinea cruris (Jock itch), Tinea manum (Ringworm of the Hand), Tinea nigra, Tinea pedis (Athlete's foot), Tinea unguium (Onychomycosis),Tinea versicolor(Pityriasis versicolor), Toxocariasis (Ocular Larva Migrans (OLM)), Toxocariasis (Visceral Larva Migrans (VLM)), Trachoma, Toxoplasmosis, Trichinosis, Trichomoniasis, Trichuriasis (Whipworm infection), Tuberculosis, Tularemia, Typhoid fever, Typhus fever,Ureaplasma urealyticuminfection, Valley fever, Venezuelan equine encephalitis, Venezuelan hemorrhagic fever,Vibrio vulnificusinfection,Vibrio parahaemolyticusenteritis, viral pneumonia, West Nile Fever, white piedra (Tinea blanca),Yersinia pseudotuberculosisinfection, yersiniosis, yellow fever, Zika fever, or zygomycosis. Exemplary immune disorders or conditions that can be treated or prevented by the polypeptides described herein include, but are not limited to, Addison's disease, agammaglobulinemia, alopecia areata, amyloidosis, ankylosing spondylitis, anti-GBM/anti-TBM nephritis, antiphospholipid syndrome (APS), autoimmune hepatitis, autoimmune inner ear disease (AIED), axonal & neuronal neuropathy (AMAN), Behcet's disease, Bullous pemphigoid, Castleman disease (CD), Celiac disease, Chagas disease, chronic inflammatory demyelinating polyneuropathy (CIDP), chronic recurrent multifocal osteomyelitis (CRMO), Churg-Strauss, Cicatricial pemphigoid/benign mucosal pemphigoid, Cogan's syndrome, Cold agglutinin disease, Congenital heart block, Coxsackie myocarditis, CREST syndrome, Crohn's disease, dermatitis herpetiformis, dermatomyositis, Devic's disease (neuromyelitis optica), Discoid lupus, Dressler's syndrome, endometriosis, eosinophilic esophagitis (EoE), eosinophilic fasciitis, erythema nodosum, essential mixed cryoglobulinemia, Evans syndrome, fibromyalgia, fibrosing alveolitis, giant cell arteritis (temporal arteritis), giant cell myocarditis, Glomerulonephritis, Goodpasture's syndrome, Granulomatosis with Polyangiitis, Graves' disease, Guillain-Barre syndrome, Hashimoto's thyroiditis, hemolytic anemia, Henoch-Schonlein purpura (HSP), herpes gestationis or pemphigoid gestationis (PG), hypogammalglobulinemia, IgA nephropathy, IgG4-related sclerosing disease, inclusion body myositis (IBM), interstitial cystitis (IC), juvenile arthritis, juvenile diabetes (Type 1 diabetes), juvenile myositis (JM), Kawasaki disease, Lambert-Eaton syndrome, leukocytoclastic vasculitis, Lichen planus, Lichen sclerosus, Ligneous conjunctivitis, linear IgA disease (LAD), lupus, Lyme disease chronic, Meniere's disease, microscopic polyangiitis (MPA), mixed connective tissue disease (MCTD), Mooren's ulcer, Mucha-Habermann disease, multiple sclerosis (MS), Myasthenia gravis, Myositis, Narcolepsy, Neuromyelitis optica, neutropenia, ocular cicatricial pemphigoid, optic neuritis, palindromic rheumatism (PR), PANDAS (Pediatric Autoimmune Neuropsychiatric Disorders Associated withStreptococcus), paraneoplastic cerebellar degeneration (PCD), Paroxysmal nocturnal hemoglobinuria (PNH), Parry Romberg syndrome, Pars planitis (peripheral uveitis), Parsonnage-Turner syndrome, Pemphigus, peripheral neuropathy, Perivenous encephalomyelitis, pernicious anemia (PA), POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, skin changes), polyarteritis nodosa, polymyalgia rheumatica, polymyositis, postmyocardial infarction syndrome, postpericardiotomy syndrome, primary biliary cirrhosis, primary sclerosing cholangitis, progesterone dermatitis, psoriasis, psoriatic arthritis, pure red cell aplasia (PRCA), pyoderma gangrenosum, Raynaud's phenomenon, Reactive Arthritis, Reflex sympathetic dystrophy, Reiter's syndrome, relapsing polychondritis, restless legs syndrome (RLS), retroperitoneal fibrosis, rheumatic fever, rheumatoid arthritis (RA), sarcoidosis, Schmidt syndrome, scleritis, scleroderma, Sjogren's syndrome, sperm & testicular autoimmunity, Stiff person syndrome (SPS), subacute bacterial endocarditis (SBE), Susac's syndrome, sympathetic ophthalmia (SO), Takayasu's arteritis, temporal arteritis/Giant cell arteritis, thrombocytopenic purpura (TTP), Tolosa-Hunt syndrome (THS), transverse myelitis, type 1 diabetes, ulcerative colitis (UC), undifferentiated connective tissue disease (UCTD), uveitis, vasculitis, vitiligo, or Wegener's granulomatosis (Granulomatosis with Polyangiitis (GPA)). The polypeptides (e.g., antibody molecules or fusion proteins) described herein are typically administered at a frequency that keeps a therapeutically effective level of polypeptides in the patient's system until the patient recovers. For example, the polypeptides may be administered at a frequency that achieves a serum concentration sufficient for at least about 1, 2, 5, 10, 20, 30, or 40 polypeptides to bind each target molecule or cell. In an embodiment, the polypeptides are administered every 1, 2, 3, 4, 5, 6, or 7 days, every 1, 2, 3, 4, 5, or 6 weeks, or every 1, 2, 3, 4, 5, or 6 months. Methods of administering various polypeptides (e.g., antibody molecules or fusion proteins) are known in the art and are described below. Suitable dosages of the polypeptides used will depend on the age and weight of the subject and the particular drug used. The polypeptides can be used by themselves or conjugated to a second agent, e.g., an bacterial agent, toxin, or protein, e.g., a second polypeptide. This method includes: administering the polypeptide, alone or conjugated to a second agent, to a subject requiring such treatment. The polypeptides can be used to deliver a variety of therapeutic agents, e.g., a toxin, or mixtures thereof. Combination Therapies The polypeptides (e.g., antibody molecules or fusion proteins) can be used in combination with other therapies. For example, the combination therapy can include a polypeptide co-formulated with, and/or co-administered with, one or more additional therapeutic agents, e.g., one or more additional therapeutic agents described herein. In other embodiments, the polypeptides are administered in combination with other therapeutic treatment modalities, e.g., other therapeutic treatment modalities described herein. Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies. Administered “in combination”, as used herein, means that two (or more) different treatments are delivered to the subject before, or during the course of the subject's affliction with a disorder. In an embodiment, two or more treatments are delivered prophylactically, e.g., before the subject has the disorder or is diagnosed with the disorder. In another embodiment, the two or more treatments are delivered after the subject has developed or diagnosed with the disorder. In some embodiments, the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap. This is sometimes referred to herein as “simultaneous” or “concurrent delivery.” In other embodiments, the delivery of one treatment ends before the delivery of the other treatment begins. In some embodiments of either case, the treatment is more effective because of combined administration. For example, the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment. In some embodiments, delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive. The delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered. In an embodiment, the polypeptide is administered in combination with a second therapy (e.g., an additional agent) to treat or prevent a disorder described herein. In an embodiment, the additional agent is a second polypeptide (e.g., antibody molecule), e.g., a polypeptide (e.g., an antibody molecule) different from a first polypeptide (e.g., antibody molecule). Exemplary polypeptides (e.g., antibody molecules) that can be used in combination include, but are not limited to, any combination of the polypeptides (e.g., antibody molecules) described herein. In another embodiment, the additional agent is other than a polypeptide (e.g., antibody molecule). For example, the additional agent can be a small molecule or a nucleic acid molecule. In yet another embodiment, the second therapy is chosen from a surgery, a radiation therapy, a cell therapy (e.g., a stem cell therapy), or an organ or tissue transplantation. In an embodiment, the second therapy comprises a therapy chosen from one or more of: an androgen replacement therapy, an antihormone therapy, an antiserum therapy, an autologous immune enhancement therapy, a biotherapy, a blood irradiation therapy, a brachytherapy, a cardiac resynchronization therapy, a cell therapy, a cell transfer therapy, a chelation therapy, a chemotherapy, a chrysotherapy, a cobalt therapy, a cold compression therapy, a cryotherapy, an electroconvulsive therapy, an electromagnetic therapy, an electron therapy, an electrotherapy, an enzyme replacement therapy, an epigenetic therapy, an estrogen replacement therapy, an extracorporeal shockwave therapy, a fast neutron therapy, a fluoride therapy, a gene therapy, a heat therapy, a helminthic therapy, a hormone therapy, a hormone replacement therapy, a host modulatory therapy, a hyperbaric oxygen therapy, a hyperthermia therapy, an immunosuppressive therapy, an immunotherapy, an intraoperative electron radiation therapy, an intraoperative radiation therapy, an inversion therapy, a laser therapy, a light therapy, a lithium therapy, a low level laser therapy, a magnet therapy, a magnetic resonance therapy, a medical gas therapy, a medical nutrition therapy, a molecular chaperone therapy, a molecular therapy, a monoclonal antibody therapy, a negative air ionization therapy, a neutron capture therapy, a neutron therapy, an oral rehydration therapy, an osmotherapy, an oxygen therapy, an ozone therapy, a palliative therapy, a particle therapy, a phage therapy, a phonemic neurological hypochromium therapy, a photodynamic therapy, a phototherapy, a photothermal therapy, a physical therapy, a prolotherapy, a protein therapy, a proton therapy, a pulsed electromagnetic field therapy, a PUVA therapy, a radiation therapy, a rehydration therapy, a respiratory therapy, salvage therapy, a serotherapy, a stem cell therapy, a stereotactic radiation therapy, a targeted therapy, a thermotherapy, a TK cell therapy, a tolerogenic therapy, a transdermal continuous oxygen therapy, an ultraviolet light therapy, or a virotherapy. Exemplary therapies that can be used in combination with a polypeptide or composition described herein to treat or prevent other disorders are also described in the section of “Methods of Treating or Preventing Disorders” herein. Methods of Diagnosis In some aspects, the present disclosure provides a diagnostic method for detecting the presence of a target molecule (e.g., a protein) or cell in vitro (e.g., in a biological sample, such as a biopsy or body fluid (e.g., blood, urine, or cerebrospinal fluid) sample) or in vivo (e.g., in vivo imaging in a subject). The method includes: (i) contacting the sample with a polypeptide described herein (e.g., an antibody molecule described herein), or administering to the subject, the polypeptide (e.g., antibody molecule); (optionally) (ii) contacting a reference sample, e.g., a control sample (e.g., a control biological sample, such as a biopsy or body fluid (e.g., blood, urine, or cerebrospinal fluid) sample) or a control subject with a polypeptide described herein (e.g., an antibody molecule described herein); and (iii) detecting formation of a complex between the polypeptide (e.g., antibody molecule) and the target molecule or cell in the sample or subject, or the control sample or subject, wherein a change, e.g., a statistically significant change, in the formation of the complex in the sample or subject relative to the control sample or subject is indicative of the presence of the target molecule or cell in the sample. The polypeptide (e.g., antibody molecule) can be directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound polypeptide (e.g., antibody molecule). Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials, as described herein. The term “sample,” as it refers to samples used for detecting bacteria includes, but is not limited to, cells, cell lysates, proteins or membrane extracts of cells, body fluids such as blood, urine, or CSF, or tissue samples such as biopsies. Complex formation between the polypeptide (e.g., antibody molecule), and the target molecule or cell, can be detected by measuring or visualizing either the polypeptide (e.g., antibody molecule) bound to the target molecule or cell, or unbound polypeptide (e.g., antibody molecule). Any suitable detection assays can be used, and conventional detection assays include an enzyme-linked immunosorbent assays (ELISA), a radioimmunoassay (RIA) or tissue immunohistochemistry. Alternative to labeling the polypeptide, the presence of the target molecule or cell can be assayed in a sample by a competition immunoassay utilizing standards labeled with a detectable substance and an unlabeled polypeptide. In this assay, the biological sample, the labeled standards and the polypeptide are combined and the amount of labeled standard bound to the unlabeled binding molecule is determined. The amount of the target molecule or cell in the sample is inversely proportional to the amount of labeled standard bound to the polypeptide (e.g., antibody molecule). The polypeptides (e.g., antibody molecules) described herein can be used to diagnose disorders that can be treated or prevented by the polypeptides described herein. The detection or diagnostic methods described herein can be used in combination with other methods described herein to treat or prevent disorders described herein. The present disclosure also includes any of the following numbered paragraphs: 1. A polypeptide comprising an Fc region, wherein the Fc region comprises a mutation, wherein the polypeptide has 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all of the following properties: a) has an increased binding affinity for a neonatal Fc receptor (FcRn) at a pH between 6.0 and 6.5, compared to a reference polypeptide; b) has a higher binding affinity for FcRn at a pH between 6.0 and 6.5 than the binding affinity at a pH between 7.0 and 7.4; c) binds to an FcRn at a pH between 6.0 and 6.5 with a dissociation constant (Kd) of 300 nM or less; d) binds to the FcRn at a pH between 7.0 and 7.4 with a Kdof 50 nM or more; e) has the same, substantially the same, or increased binding affinity for an Fcγ receptor, compared to a reference polypeptide; f) has the same, or substantially the same, thermal stability, compared to a reference polypeptide; g) has the same, substantially the same, or increased binding affinity for C1q, compared to a reference polypeptide; h) has the same, substantially the same, or increased binding affinity for TRIM21, compared to a reference polypeptide. i) has an effector function that is the same, substantially the same, or increased, compared to a reference polypeptide; j) has an increased half-life in vivo, compared to a reference polypeptide; k) has a biological function, in vitro, ex vivo, or in vivo, that is the same, substantially the same, or increased, compared to a reference polypeptide; l) has a developability characteristic that is the same or substantially the same, compared to a reference polypeptide; m) has the same, substantially the same, or increased binding affinity, specificity, or both, for an epitope, compared to a reference polypeptide; orn) increases mucosal uptake, compared to a reference polypeptide, and wherein the polypeptide has at least properties a), b), and one, two, three, four, or all of properties e), f), g), h), or i). 2. The polypeptide of paragraph 1, which has at least properties a), b), c), d), and one, two, three, four, or all of properties e), f), g), h), or i). 3. The polypeptide of paragraph 1 or 2, which has at least properties a), b), one, two, three, four, or all of properties e), f), g), h), or i), and one, two, three, four, five, six, or all of properties c), d), j), k), l), m), or n). 4. The polypeptide of any of paragraphs 1-3, which has at least 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50-fold increase in the binding affinity for the FcRn at pH 6.0, compared to a reference polypeptide, as determined by an octet-based assay or a cell-based assay. 5. The polypeptide of any of paragraphs 1-4, which has at least 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 50-fold higher in the binding affinity for the FcRn at pH 6.0 than that at pH 7.4, as determined by an octet-based assay or a cell-based assay. 6. The polypeptide of any of paragraphs 1-5, which binds to the FcRn at pH 6.0 with a dissociation constant (Kd) of 250 nM or less, 200 nM or less, 150 nM or less, 100 nM or less, 50 nM or less, 25 nM or less, 10 nM or less, 5 nM or less, 2 nM or less, 1 nM or less, 0.5 nM or less, 0.2 nM or less, 0.1 nM or less, 0.05 nM or less, 0.02 nM or less, 0.01 nM or less, between 25 nM and 0.1 nM, between 20 nM and 0.5 nM, between 15 nM and 1 nM, between 10 nM and 5 nM, or between 20 nM and 10 nM, as determined by an octet-based assay or a cell-based assay. 7. The polypeptide of any of paragraphs 1-6, which binds to the FcRn at pH 7.4 with a Kdof 60 nM or more, 80 nM or more, 100 nM or more, 150 nM or more, 200 nM or more, 500 nM or more, between 50 nM and 500 nM, or between 100 nM and 250 nM, as determined by an octet-based assay or a cell-based assay. 8. The polypeptide of any of paragraphs 1-7, which decreases the binding affinity for one, two, or, all of FcγRI, FcγRIIa/b, or FcγRIII by no more than 10%, 20%, 30%, 40%, or 50%, or increases the binding affinity for one, two, or all of FcγRI, FcγRIIa/b, or FcγRIII by at least 1.5, 2, 3, 4, or 5-fold, compared to a reference polypeptide, as determined by an octet-based assay or a cell-based assay. 9. The polypeptide of any of paragraphs 1-8, which increases or decreases the melting temperature by no more than 1° C., 2° C., 3° C., 4° C., 5° C., 6° C., 7° C., 8° C., 9° C., or 10° C., compared to a reference polypeptide, as determined by a sypro orange assay. 10. The polypeptide of any of paragraphs 1-9, which decreases the binding affinity for C1q by no more than 10%, 20%, 30%, 40%, or 50%, or increases the binding affinity for C1q by at least 1.5, 2, 3, 4, or 5-fold, compared to a reference polypeptide, as determined by ELISA. 11. The polypeptide of any of paragraphs 1-10, which decreases the binding affinity for TRIM21 by no more than 10%, 20%, 30%, 40%, or 50%, or increases the binding affinity for TRIM21 by at least 1.5, 2, 3, 4, or 5-fold, compared to a reference polypeptide, as determined by ELISA. 12. The polypeptide of any of paragraphs 1-11, which decreases one, two, three, or all of a complement dependent cytotoxicity (CDC), an antibody dependent cell mediated cytotoxicity (ADCC), an antibody dependent cell mediated phagocytosis (ADCP), or an antibody dependent intracellular neutralization (ADIN) by no more than 10%, 20%, 30%, 40%, or 50%, or increases one, two, three, or all of a complement dependent cytotoxicity (CDC), an antibody dependent cell mediated cytotoxicity (ADCC), an antibody dependent cell mediated phagocytosis (ADCP), or an antibody dependent intracellular neutralization (ADIN) by at least 1.5, 2, 3, 4, or 5-fold, compared to a reference polypeptide. 13. The polypeptide of any of paragraphs 1-12, which has at least 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10-fold increase in the half-life in vivo, compared to a reference polypeptide, as determined in an animal model. 14. The polypeptide of any of paragraphs 1-13, which decreases the biological function, in vitro, ex vivo, or in vivo by no more than 10%, 20%, 30%, 40%, or 50%, or increases the biological function, in vitro, ex vivo, or in vivo by at least 1.5, 2, 3, 4, or 5-fold, compared to a reference polypeptide. 15. The polypeptide of any of paragraphs 1-14, which alters one, two, three, or all of stability, solubility, aggregation, or expression level by no more than 10%, 20%, 30%, 40%, or 50%, compared to a reference polypeptide. 16. The polypeptide of any of paragraphs 1-15, which decreases the binding affinity, specificity, or both by no more than 10%, 20%, 30%, 40%, or 50%, or increases the binding affinity, specificity, or both by at least 1.5, 2, 3, 4, or 5-fold, compared to a reference polypeptide. 17. The polypeptide of any of paragraphs 1-16, which increases mucosal uptake by at least 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10-fold, compared to a reference polypeptide, as determined by a transcytosis assay. 18. The polypeptide of any of paragraphs 1-4 or 8-17, wherein the reference polypeptide comprises a wild-type Fc region, an Fc region comprising the amino acid sequence of SEQ ID NO: 1, or an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues. 19. The polypeptide of any of paragraphs 1-18, wherein the mutation is in a residue in a CH2 domain. 20. The polypeptide of any of paragraphs 1-18, wherein the mutation is in a residue in a CH3 domain. 21. The polypeptide of any of paragraphs 1-20, comprising at least one mutation in a residue in a CH2 domain and at least one mutation in a residue in a CH3 domain. 22. The polypeptide of any of paragraphs 1-21, further comprising a mutation in a residue in a region other than a CH2 domain and/or a CH3 domain. 23. The polypeptide of any of paragraphs 1-22, wherein the mutation does not alter, or does not substantially alter, the conformation of the linker region between a CH2 domain and a CH3 domain. 24. The polypeptide of any of paragraphs 1-23, wherein the mutation does not introduce 3, 4, 5, 6, 7, 8, 9, 10, or more consecutive hydrophobic or aromatic residues on a surface region. 25. The polypeptide of any of paragraphs 1-24, which comprises an antibody molecule. 26. The polypeptide of any of paragraphs 1-25, which comprises an IgG. 27. The polypeptide of any of paragraphs 1-26, which comprises an IgG1, IgG2, IgG3, or IgG4. 28. The polypeptide of any of paragraphs 1-27, which comprises a heavy chain immunoglobulin variable region, a light chain immunoglobulin variable region, or both. 29. The polypeptide of any of paragraphs 1-28, which comprises a tetramer of two heavy chain immunoglobulin variable regions and two light chain immunoglobulin variable regions. 30. The polypeptide of any of paragraphs 1-29, which comprises a full length antibody molecule. 31. The polypeptide of any of paragraphs 1-30, which comprises a fragment of an antibody molecule. 32. The polypeptide of any of paragraphs 1-31, which comprises a chimeric antibody molecule or a murine antibody molecule. 33. The polypeptide of any of paragraphs 1-32, which comprises a human antibody molecule or a humanized antibody molecule. 34. The polypeptide of any of paragraphs 1-24, which comprises a fusion protein. 35. The polypeptide of any of paragraphs 1-34, comprising 1, 2, 3, 4, or all of the following:(i) a mutation in a residue in a surface region that interacts with FcRn;(ii) a mutation in a residue that is a peripheral residue along the Fc-FcRn interface; (iii) a mutation is in a residue that is non-contact residue in Fc-FcRn binding;(iv) a mutation in a residue which is a helix contact reside that enhances the conformational dynamics of a helix comprising 1, 2, 3, 4, 5, or all of P247, K248, D249, T250, L251, or M252; or(v) a mutation, which modulates pK of a histidine or is an introduction of a histidine along the Fc-FcRn interface. 36. The polypeptide of any of paragraphs 1-35, comprising a mutation in a residue in a surface region that interacts with FcRn. 37. The polypeptide of paragraph 36, wherein the mutation is in a residue chosen from: L251, 1253, R255, P257, H285, N286, K288, T307, V308, L309, Q311, L314, H310, H433, N434, H435, or Y436. 38. The polypeptide of any of paragraphs 1-37, comprising a plurality of mutations in 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or all of the residues chosen from L251, I253, R255, P257, H285, N286, K288, T307, V308, L309, Q311, L314, H310, H433, N434, H435, or Y436. 39. The polypeptide of any of paragraphs 1-38, comprising a mutation in a residue that is a peripheral residue along the Fc-FcRn interface. 40. The polypeptide of paragraph 39, wherein the mutation is in a residue chosen from M252, T256, T307, L309, Q311, H433, N434, Y436, N286, or K288. 41. The polypeptide of any of paragraphs 1-40, comprising a plurality of mutations in 2, 3, 4, 5, 6, 7, 8, 9, or all of the residues chosen from M252, T256, T307, L309, Q311, H433, N434, Y436, N286, or K288. 42. The polypeptide of any of paragraphs 1-41, comprising a mutation is in a residue that is non-contact residue in Fc-FcRn binding. 43. The polypeptide of paragraph 42, wherein the mutation is in a residue chosen from A287, V308, N315, L314, L432, H429, E430, or A431. 44. The polypeptide of any of paragraphs 1-43, comprising a plurality of mutations in 2, 3, 4, 5, 6, 7, or all of the residues chosen from A287, V308, N315, L314, L432, H429, E430, or A431. 45. The polypeptide of any of paragraphs 1-44, comprising a mutation in a residue which is a helix contact reside that enhances the conformational dynamics of a helix comprising 1, 2, 3, 4, 5, or all of P247, K248, D249, T250, L251, or M252. 46. The polypeptide of paragraph 45, wherein the mutation is in a residue chosen from P244, P245, T250, L251, P247, E380, M428, A378, D376, P257, V308, A287, L306, or H427. 47. The polypeptide of any of paragraphs 1-46, comprising a plurality of mutations in 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all of the residues chosen from P244, P245, T250, L251, P247, E380, M428, A378, D376, P257, V308, A287, L306, or H427. 48. The polypeptide of any of paragraphs 1-47, comprising a mutation which is the introduction of a histidine along the Fc-FcRn interface. 49. The polypeptide of any of paragraphs 1-48, comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations, or one or more combination of mutations, as described in Table 1. 50. The polypeptide of any of paragraphs 1-49, wherein the mutation is other than M252Y, S254T, T256E, L309N, T250Q, M428L, N434S, N434A, T307A, E380A, N434A, M252Y, S254T, T256E, or a combination thereof. 51. The polypeptide of any of paragraphs 1-50, wherein the mutation is in a residue other than residue M252Y, S254T, T256E, L309N, T250, M428, N434, N434, T307, E380, N434, M252, S254, T256, or a combination thereof. 52. The polypeptide of any of paragraphs 1-51, which does not have 1, 2, 3, 4, 5, 6, 7, 8, 9, or all of the following mutation or mutations: (i) M252Y, S254T, and T256E; (ii) L309N; (iii) T250Q and M428L; (iv) M428L and N434A; (v) N434A; (vi) T307A, E380A, and N434A; (vii) M252W; (viii) V308F; (ix) V308F and N434Y; or (x) H435A. 53. The polypeptide of any of paragraphs 1-52, comprising a first mutation chosen from M252Y, S254T, T256E, L309N, T250Q, M428L, N434S, N434A, T307A, E380A, N434A, M252Y, S254T, or T256E, and a second mutation chosen from a mutation in Table 1 other than M252Y, S254T, T256E, L309N, T250Q, M428L, N434S, N434A, T307A, E380A, N434A, M252Y, S254T, and T256E. 54. The polypeptide of any of paragraphs 1-53, further comprising a mutation in the Fc region that increases an effector function. 55. The polypeptide of paragraph 54, wherein the mutation is in a residue chosen from S239, A330, I332, F243, G236, or a combination thereof. 56. The polypeptide of any of paragraphs 1-55, further comprising a mutation in the Fc region that decreases an effector function. 57. The polypeptide of paragraph 56, wherein the mutation is in a residue chosen from K322, L234, L235, P331, N297, or a combination thereof. 58. The polypeptide of any of paragraphs 1-57, wherein the Fc region comprises: (a) 1, 2, 3, 4, 5, or all of the combinations of mutations chosen from: T256D/Q311V/A378V, H285N/T307Q/N315D, H285D/T307Q/A378V, T307Q/Q311V/A378V, T256D/N286D/T307R/Q311V/A378V, or T256D/T307R/Q311V; (b) a mutation or a combination of mutations capable of disrupting an Fc effector function, or (c) both (a) and (b). 59. The polypeptide of any of paragraphs 1-58, which comprises mutations in residues chosen from: (i) T256, Q311, and A378; (ii) H285, T307, and N315; (iii) H285, T307, and A378; (iv) T307, Q311, and A378; (v) T256, N286, T307, Q311, and A378; or (vi) T256, H285, T307, Q311, and A378. 60. The polypeptide of any of paragraphs 1-59, which comprises mutations chosen from: (i) T256D, Q311V, and A378V; (ii) H285N, T307Q, and N315D; (iii) H285D, T307Q, and A378V; (iv) T307Q, Q311V, and A378V; (v) T256D, N286D, T307R, Q311V, and A378V; or (vi) T256D, H285D, T307R, Q311V, and A378V. 61. The polypeptide of any of paragraphs 1-60, further comprising a mutation in a region other than the Fc region. 62. The polypeptide of any of paragraphs 1-61, comprising a plurality of mutations, wherein at least one mutation is a compensating or beneficial mutation. 63. The polypeptide of any of paragraphs 1-62, which is an isolated polypeptide. 64. The polypeptide of any of paragraphs 1-62, which is a synthetic polypeptide. 65. A composition comprising a polypeptide of any of paragraphs 1-64. 66. The composition of paragraph 65, further comprising a pharmaceutical acceptable carrier. 67. A nucleic acid molecule encoding a polypeptide of any of paragraphs 1-64. 68. A vector comprising a nucleic acid molecule of paragraph 67. 69. A cell comprising a nucleic acid molecule of paragraph 67 or a vector of paragraph 68. 70. A kit comprising a polypeptide of any of paragraphs 1-64 and instructions to use of the polypeptide. 71. A container comprising a polypeptide of any of paragraphs 1-64. 72. A method of producing a polypeptide, the method comprising culturing a cell of paragraph 69 under conditions that allow production of an antibody molecule, thereby producing the polypeptide. 73. The method of paragraph 72, further comprising isolating or purifying the polypeptide. 74. A method of treating a disorder, the method comprising administering to a subject in need thereof an effective amount of a polypeptide of any of paragraphs 1-64 or a composition of paragraph 65 or 66, thereby treating the disorder. 75. A polypeptide of any of paragraphs 1-64, or a composition of paragraph 65 or 66, for use in treating a disorder in a subject. 76. Use of a polypeptide of any of paragraphs 1-64, or a composition of paragraph 65 or 66, in the manufacture of a medicament for the treatment of a disorder in a subject. 77. A method of detecting a molecule, the method comprising contacting a cell or a sample from a subject with a polypeptide of any of paragraphs 1-64, thereby detecting the molecule. EXAMPLES A structure and network based framework was developed to interrogate the engagement of IgG with multiple Fc receptors (e.g., FcRn, C1q, TRIM21, FcγRI, FcγRIIa/b, and FcγRIIIa). Using this framework, features that govern Fc-FcRn interaction and multiple distinct pathways for enhancing FcRn binding in a pH specific manner were identified. Network analysis provided a lens to study the allosteric impact of FcRn optimization mutations on the distal FcγR engagement. Applying these principles, a panel of distinct Fc variants that enhance FcRn binding with robust biophysical properties and wild-type like binding to activating receptors were engineered. Control polypeptides with these Fc variants have shown a half-life improvement of >9 fold and robust effector functions such as ADCC, ADCP, CDC and ADIN mediated by TRIM21. Example 1: Structure Characterization and Molecular Features of Fc-FcRn Interaction A co-crystal structure of the human FcRn protein in complex with the Fc domain of a human IgG1 and human serum albumin was recently reported (Oganesyan et al.,J Biol Chem,2014. 289 (11): 7812-2). Inspection of the structure (FIG.20A) shows that both the alpha and beta subunits of the FcRn molecule participate in binding to the Fc-domain, making contact with both CH2 and CH3 domains of the Fc. The primary interaction is mediated by the alpha-subunit on the FcRn side and CH2 domain on the Fc side. The pH specific nature of binding is driven by the histidine residues at positions 310 and 435 (Kabat numbering) which undergo protonation at acidic pH and make critical contacts with FcRn glutamate at position 115 and aspartate at position 130. Mutation of either of the two histidine residues significantly reduces the binding affinity of Fc with FcRn (Oganesyan et al.,J Biol Chem,2014. 289(43): 29874-80; Raghavan et al.,Biochemistry,1995. 34(45): 14649-57). In addition to the H310 and H435, a number of other Fc residues (L251, I253, R255, P257, H285, N286, K288, T307, V308, L309, Q311, L314, H433, N434, and Y436) are involved in making molecular contacts with FcRn. The structure of Fc has been solved at different pHs (Crispin et al.,Proc Natl Acad Sci USA,2013. 110(38): E3544-6; Ahmed et al.,J Mol Biol,2014. 426(18): 3166-79; Chen et al.,ACS Chem Biol,2016. 11(7): 1852-61). Superposition of the crystal structure of Fc at pH 4.0 (pdb id 4BYH) and 6.5 (pdb id 4Q7D) reveals a number of subtle changes, including lateral displacement of the 250-helix and differences in the relative orientation of CH2 (FIG.20B). Given that FcRn binding amino acid residues such as M252 and 1253 are located on the 250-helix, the observed displacement of the helix is expected to influence the FcRn binding (Oganesyan et al.,J Biol Chem,2014. 289 (11): 7812-2). Further, the difference in the relative orientation of CH2 with respect to CH3 highlights the conformational flexibility of the CH2 region (FIG.20B) (Frank et al.,J Mol Biol,2014. 426(8): 1799-811). Given that the FcRn-Fc interface is found across both the CH2/CH3 domains of the Fc, the conformation dynamics of the CH2 domain is also expected to influence the FcRn binding. Deuterium exchange studies on the Fc residues in the presence and absence of FcRn at acidic and neutral pHs showed that FcRn binding to Fc molecule offers protection to its binding Fc residues; however, in the absence of FcRn molecule the 250-helix residues at acidic pH showed enhanced Deuterium exchange suggesting that pH change induces conformational change for this region (Walters et al.,J Biol Chem,2016. 291(4): 1817-25; Jensen et al.,Mol Cell Proteomics,2017. 16(3): 451-456). Analysis of the strength of engagement of Fc with FcRn at acidic pH reveals that human FcRn binds to human Fc domain with weak affinity (>600 nM). The kinetic parameters further show that while the rate of association (kon˜105M−1s−1) of Fc-FcRn interaction is comparable to a typical antibody-antigen interaction, the lower binding affinity is primarily due to the fast dissociation rate (koff˜0.1 s−1) of Fc-FcRn interaction (Suzuki et al.,J Immunol,2010. 184(4): 1968-76). The conformational flexibility of the CH2 subdomain is thought to contribute to the poor koffof FcRn binding. Improving the off rate of Fc-FcRn interaction can serve as a key facet for improved half-life (Datta-Mannan et al.,J Biol Chem,2007. 282(3): 1709-17). Towards that, four sets of Fc residues, mutations to which could impinge on FcRn interaction and dissociation rate (koff), were identified (FIG.21A). This includes residues that make direct contact with FcRn as well as peripheral and non-surface exposed residues that have the potential to modify the interaction surface and residues that have the potential to influence 250-helix dynamics. Indeed, some of these positions have been previously interrogated and half-life enhancing mutants such as M252Y/S254T/T256E (YTE), M428L/N434S (LS), T307A/E380A/N434A (AAA), T250Q/M428L (QL), V308P, include combinations of mutations at the listed sites. In the analysis, the role of each residue was first investigated in silico, and the network of interactions mediated by the residue was identified (FIG.21B). Specific set of mutations were designed to enhance hydrophobic interactions in the Fc-FcRn interface and to enhance polar and electrostatic interactions at the periphery of the Fc-FcRn interface. Mutations of non-contacting residues near the interface can enhance the electrostatic charge complementarity and the affinity by reducing the koffrate (Lee and Tidor,Protein Sci,2001. 10(2): 362-77; Whitehead et al.,Nat Biotechnol,2012. 30(6): 543-8). The FcRn binding site on Fc has overlaps with the binding site of intracellular receptor TRIM21, protein A used for antibody purification and the Fc-Fc interaction interface formed during hexamerization of IgG for CDC activity. Substitution of at each position was assessed for impact on these binding to TRIM21, protein A and Fc. More than 30 distinct positions were chosen for substitutions. Combination of the individual mutations were designed based on the following guiding principles (a) avoiding introduction of large clusters of charge or hydrophobicity to minimize binding to solvent ions and impact on thermal stability of the IgG and (b) incorporate diversity in the types of interaction, not just electrostatic or hydrophobic (c) include mutations across CH2 and CH3 domains. Based on these considerations, more than 150 unique mutant combinations were designed and experimentally evaluated. IgG1s incorporating the designed Fc variants on either actoxumab or motavizumab Fab were recombinantly expressed and evaluated for binding to human FcRn by biolayer interferometry using two different protocols: FcRn on NiNTA biosensor with IgG as the analyte, and IgG on an anti-CH1 biosensor with FcRn as the analyte. The assays were used to quantify binding at pH 6.0 followed by dissociation at pH 6.0 and pH 7.4, as well as binding and dissociation at pH 7.4 (FIG.11A). Fc variants incorporating mutations at P257 and V308 had high affinity at pH 6.0 but displayed markedly slower off-rates at pH 7.4 and were not considered for further analyses. More than 10 distinct variants had greater than 5-fold decrease in Kdas compared to IgG with WT Fc. Further, these variants decreased the koffby more than 2.5-fold (FIG.22). The FcRn binding site on the Fc domain considerably overlaps with the protein A and Trim21 binding sites as determined from their complex crystal structures (FIG.27A). The Fc domain also binds to C1q to mediate CDC, however the C1q binding site does note overlap with FcRn binding site on Fc. C1q is naturally found as hexamer and a homo-hexamer assembly of Fc domains is expected to not only have a better C1q binding and also CDC. The Fc-Fc interface for formation of homo-hexamer overlaps with the FcRn binding site (FIG.27A). Superposition of crystal structures of Fc domain shows that the orientation of the CH2 domain in these structures varies with respect to the CH3 domain illustrating the conformational flexibility of the CH2 domain (FIG.27B). Since the FcRn binding site is at the interface across both the CH2 and CH3 domains therefore this conformational flexibility may be important for its binding. Example 2: Effect of Fc Region Mutations on pH Specific Fc-FcRn Binding The FcRn binding affinity of Fc variants FcMut008 and FcMut015 was assessed by Octet. Antibody was immobilized on the Octet tip by anti-CH1 antibody and FcRn was in solution. As shown inFIG.6, FcMut008 and FcMut015 had increased affinity to FcRn. The binding is pH specific and increased binding at pH 6.0 was observed. Example 3: Cell-Based FcRn Binding Competition Assay Cell-based competition assay provides a robust, specific, linear assay to show differences in relative binding of Fc variants. FcRn-expressing cells were obtained by transient transfection of FcRn alpha and β2m. Cells were incubated at pH6.0 with dilutions of IgG and fixed concentration of fluorescently-labeled Fc (Fc-A488). Cell-bound fluorescence was read by FACS. The results are shown inFIG.7. FcMut008 and FcMut015 showed improved FcRn binding at pH6.0. Example 4: Effect of Fc Region Mutations on FcγR Engagement The binding of exemplary mutant antibody molecules to FcγRI and FcγRIIIa was determined. As shown inFIG.8, FcMut008 and FcMut015 retained and in some instance enhanced FcγR engagement. Example 5: Effect of Fc Region Mutations on Thermal Stability and Biophysical Characterization of Fc Variants The thermal stability of exemplary mutant antibody molecules was determined. The thermal stability was measured by SYPRO orange. As shown inFIG.9, FcMut008 and FcMut015 retained high melting temperature. The impact of incorporating exemplary Fc variants on biophysical attributes was experimentally assessed. IgGs incorporating the Fc variants on motavizumab Fab were tested on SE-HPLC. All samples eluted at similar retention times as wild-type Fc, and displayed clean monomeric profile, and no aggregates were detected (FIG.23). The IgGs were also assessed for the thermal stability of the CH2 and CH3 domains by Differential Scanning Fluorimetry (DSF). The melting temperature (Tm) of the wild type human CH2 and CH3 domain, as measured differential scanning calorimetry, is approximately 70° C. and 81.5° C., respectively (Ionescu et al.,J Pharm Sci,2008. 97(4): 1414-26). The DSF experimental results in this Example yielded similar results with a CH2 and CH3 TMof 68.8° C. and 80.8° C., respectively. The half-life extending Fc variant YTE has been reported to decrease the TMof the CH2 domain by 6.7° C. (Majumdar et al.,MAbs,2015. 7(1): p. 84-95.). In the experiments described in this Example, the TMof the CH2 domain of YTE was 7.2° C. lower than WT. Additionally, mutations at 247, 257, and 308 significantly impacted the TMof CH2. The exemplary Fc variants (FcMut183, FcMut197, FcMut213, FcMut215, FcMut228, FcMut229) were thermally stable with the TMof the CH2 domain >64° C. (FIG.23). Example 6: Evaluation of Fc Variants in Transgenic Mice Model Tg32 mice were homozygous, 8 week old, males. There were 4 mice per test article group. The test articles included CDA1-WT, CDA1-FcMut008, and CDA1-FcMut015. The mice were dosed at 10 mg/Kg by IV administration. Data were collected at thirteen time points (1 h, 8 h, 1 d, 2 d, 3 d, 4 d, 6 d, 8 d, 10 d, 13 d, 16 d, 19 d, and 22 d). Human IgG was quantified by ELISA using an anti-hIgG polyclonal antibody. Tg32 is a human FcRn transgenic mouse model that can be used in drug discovery for early assessment and prediction of human pharmacokinetics of monoclonal antibodies. Monoclonal antibody clearance in Tg32 homozygous mice has the strongest correlation to monoclonal antibody clearance in humans (Avery et al.MAbs.2016; 8(6):1064-78). CDA1 (actoxumab) is known to have a half-life of >25 days in human. In vivo evaluation with additional mAbs in Tg32 model was performed. The different constructs can also be evaluated on Tg276 mice which are reported to have increased half-life differences between IgG variants. The results are shown in Table 2 andFIG.10. FcMut015 increased the half-life of CDA1 in Tg32 mice. TABLE 2Half-Lives of Exemplary Antibody Moleculesin Tg32 Homozygous MiceCmaxClastAUCinfGroupt1/2(hr)(ug/ml)(ug/ml)(hr * ug/ml)RsqWT261.17116.0315.4024108.030.99FcMut008231.92131.3315.7425687.390.99FCMut015436.69151.8227.6942735.90.93 Example 7: Fc Engineering of Exemplary Antibodies FcRn interaction with IgG is believed to be mediated through Fc. The binding of Fc to FcRn is generally pH specific (typically little or no binding at pH7.4 and strong binding in acidic environment). Structure of FcRn in complex with Fc domain of IgG1 is known and each FcRn molecule binds to an Fc-monomer. Fab domains can also influence binding of IgG to FcRn. Network analysis of Fc-FcRn complex highlights the centrality of H310 in engagement with FcRn. H310 is highly interconnected to multiple other highly networked residues. Mutations in the H310 cluster, and neighboring (connected nodes) may strengthen the H310 network. Analysis of sub-networks informs introduction of synergistic mutations for favorable FcRn interaction, with minimal impact on other Fc residues. To identify Fc variants with improved binding to FcRn at pH 6.0, various Fc mutations were engineered into IgG1 Motazivumab (WT). All antibodies were assessed for binding to FcRn using biolayer interferometry. Briefly, anti-CH1 biosensors (ForteBio) were loaded with each antibody of interest. Loaded biosensors were exposed to recombinant FcRn protein at pH 6.0 to detect binding. After saturation, biosensors were exposed to buffer alone at pH 6.0 to measure dissociation of the FcRn from each antibody. The result is a response curve representing on rate and off rate of the antibody. These rates were calculated using the ForteBio software and compared to the rate of wild type Motavizumab. The fold increase in on-rate and the fold decrease in off-rate as compared to wild type are listed in the table. Numerous antibody mutations significantly increased and decreased on and off rate, respectively (FIG.11A). Also shown is the binding of a representative Fc variant to FcRn at a pH range of 6.0 to 7.4. It is important that the affinity of the Fc mutant antibodies for FcRn is improved at pH 6.0 but not significantly enhanced at a higher pH such as pH 7.4.FIG.11Bdemonstrates that this representative antibody still shows poor binding to FcRn at pH 7.4, a desirable feature. FIG.11Cshows the interaction of the CH2 domain of the antibody Fc with the FcRn molecule. The engineering efforts described herein attempted to improve shape complementarity (SC), electrostatic charge complementarity (CC), and hydrophobic complementarity (HC). A few positions on the Fc are noted as important for the interaction. Also noted is the 250 helix. This helix is dynamic and moves depending on the pH of the environment. This is important in the binding of the Fc domain to FcRn at pH 6.0 but not pH 7.4. Exemplary Fc variants were tested in developability assays, the results of which are summarized inFIG.12. Assays, including Sypro orange, SDS PAGE and SEC-HPLC, were performed. For expression determination, constructs were transfected into Expi293 cells in 96 well culture dishes using ExpiFectamine as described by the manufacturer. After 5-7 days, supernatants were quantified using biolayer interferometry (Octet) equipped with anti-human CH1 biosensors using a motavizumab standard curve. For protein A binding assessment the following was performed. Constructs were transfected into Expi293 cells in 30 mL cultures using ExpiFectamine as described by the manufacturer. After 5-7 days, supernatants were harvested and antibodies purified. Using biolayer interferometry with protein A biosensors, Fc-modified antibody affinity to protein A was measured and compared to antibody containing a wild type Fc domain. All Fc variants, expressed in the context of the Motavizumab antibody, were separated by SDS-PAGE under reducing and non-reducing conditions. Briefly, 2 μg of antibody in 5 μL of water was mixed with 5 μL of Laemmli Sample Buffer (BioRad Catalog #161-0737), with and of f3-mercaptoethanol (BioRad Catalog #161-0710). The samples were boiled at 95° C. for ten minutes and then briefly centrifuged. Samples were then run on a 4-12% Bis-Tris NuPAGE gel (Thermo Scientific #NP0321BOX) in 1×MES running buffer in an XCell SureLock Gel Electrophoresis Cell (Novex Catalog #090403-839) alongside the SeeBlue Plus2 molecular weight standard (Invitrogen Catalog #LC5925). The samples were run at 200V for 35 minutes. The gels were stained with SimplyBlue SafeStain (Novex #LC6065) following manufacturer's protocol and imaged on the BioRad ChemiDoc MP Imaging System. HPLC based size exclusion chromatography (HPLC-SEC) is an analytical tool used to determine the apparent size of a protein, monomeric purity, and the apparent level non-specific column adsorption between the protein and the silica based sizing resin. The impact of Fc mutations on the IgG elution profile was assessed on a Phenomenex Biosep 3000s column. Briefly the IgGs with various Fc variants were diluted to 1 mg/ml in PBS pH 7.4 and 20 μL was injected into the column. The elution time and percentage purity was recorded. The thermal stability of the mAbs was determined by differential scanning fluorescence (DSF). DSF monitors the conformational stability of a protein as it is exposed to increasing thermal stress. The dye, SYPRO ORANGE®, fluoresces in a hydrophobic environment, such as hydrophobic core residues that are exposed during thermally triggered protein unfolding, or denaturation. By monitoring the fluorescent signal, the unfolding of CH2, CH3 and Fab can be monitored. Various Fc variants with Motavizumab Fab were evaluated in a SYPRO orange assay. Briefly 15 μL of IgG at ˜0.5 mg/mL was mixed with 15 μL of 1:500 diluted SYPRO Orange dye and assessed by a thermocycler with fluorescent read capabilities using a 1° C. ramp from 40° C. to 99° C. The midpoint between native state and first unfolding event was reported as the transition temperature or melt temperature (Tm). All Fc mutations were introduced in IgG1 (m3 allotype) heavy chain gene and cloned into pcDNA3.1(C). The light chain genes were cloned into pcDNA3.1(A). In all cases, the native signal peptide was replaced with an osteonectin signal peptide (GenBank accession # AAA60993). Co-expression of the heavy and light chain vectors was performed by transient transfection in Expi293 cells using the Expi293 transfection kit (Thermo Fisher catalogue # A14524) following the manufacturer's protocol. The heavy and light chain vectors were co-transfected at a 1:2 ratio. Cell culture supernatant was harvested 5 to 7 days post transfection and purified on the AKTA 10 FPLC system (GE) using HiTrap MabSelect SuRe protein A columns (GE) following manufacturer's instructions. All antibodies were purified from cell culture supernatant using 1 mL columns packed with mAb select sure protein A resin (GE catalogue #17543801) using the AKTA purifier 10 FPLC system. Briefly, sterile filtered cell culture supernatant was loaded onto the columns at a flow rate of 2 mL/minute. Columns were washed with 10 column volumes of PBSN (1×PBS with 0.05% sodium azide). Antibodies were eluted with 10 column volumes of elution buffer (100 mM glycine pH 2.5) and neutralized by addition 17.5% v/v of neutralization buffer (1M Tris, 1M NaCl, pH 8.0) and collated in 1 mL fractions. The chromatogram for absorbance at 280 nm was used to identify elution fractions containing the antibody. All antibodies were then dialyzed into 1×PBS using 10,000 dalton molecular weight cut-off cassette (Thermo Fisher catalogue #66380). Protein A binding was functionally determined by the ability of all antibodies to be purified by FPLC using protein A columns. After purification, protein A binding was further assessed by quantifying a known amount of antibody using the Octet QKesystem and protein A biosensors (Pall catalogue #18-5012) following the standard quantitation protocol provided with the Octet data acquisition software. Expi293 cells were co-transfected with a plasmid encoding the human α-FcRn with a 6× histidine tag on the C-terminus and a plasmid encoding human β2M. Cell culture supernatant was harvested 4 days post transfection. FcRn was purified from cell culture supernatant using the AKTA pure FPLC system with a HisTrap HP column (GE catalogue #17-5247-01). Post purification the protein was dialyzed into 1×PBS pH 6.0. Screening assays were performed using anti-CH1 Fab biosensors on the Octet QKesystem. Briefly, purified IgG at 10 μg/mL is loaded onto an anti-CH1 biosensor for 180 seconds. After a 60 second baseline step in 1×PBS pH 6.0, the IgG loaded tip is exposed to FcRn at a concentration of 50 μg/mL for 60 seconds, followed by dissociation for 60 seconds in PBS pH 6.0, and an additional 30 seconds in PBS pH 7.4. In addition, FcRn binding was performed using NiNTA biosensors. Briefly, recombinant human FcRn at 5 μg/mL is loaded onto a NiNTA biosensor for 180 seconds. After a 60 second baseline step in 1×PBS pH 6.0, the FcRn loaded tip is exposed to IgG at a concentration of 250 nM (37.5 μg/mL) for 60 seconds, followed by dissociation for 60 seconds in PBS pH 6.0, and an additional 30 seconds in PBS pH 7.4. After assay completion of each assay, a quantitative assessment of the affinity constant (KD) at pH 6.0 is performed using the ForteBio octet software and a qualitative assessment is performed by plotting the response rate over time, allowing for visualization of the association of IgG to FcRn at pH 6.0 and the subsequent dissociation at pH 6.0 and pH 7.4. As shown inFIG.12, all Fc variants performed comparably to the WT antibody in all of these experiments. Example 8: In Vivo Assessment of Half-Life and Pharmacokinetic Analysis of Engineered Antibodies Motavizumab wildtype was compared to Motavizumab containing three of Fc engineering mutations in the in vivo assessment of engineered antibody half-life. Antibodies (2-5 mg/kg) were administered to mice transgenic for human FcRn and daily samples of mouse serum were obtained (day 0-day 4). ELISA was performed on serum to quantify the amount of Motazivumab in the serum. The amount of human IgG present in mouse serum was determined using a human IgG quantitation ELISA kit (Bethyl Labs catalogue # E80-104) following the manufacturer's protocol. All serum samples were titrated in a twofold serial dilution starting at 1:50 dilution and ending at a 1:6400 dilution. Each ELISA plate included a human reference standard curve provided with the kit in duplicate or triplicate. The standard curve contains the following concentrations: 500.0, 250.0, 125.0, 62.5, 31.3, 15.6, 7.8, and 3.9 ng/mL. The lower limit of detection was considered to be the A450 nm value obtained for the second to last point on the reference standard, which was 7.8 ng/mL. Because the starting dilution of the serum was 1:50, this puts the level of detection the assay at ˜0.4 μg/mL (7.812 ng/mL×50 fold dilution). The following procedures were followed to calculate the IgG levels: (1) perform a four parameter logistic regression (4PL) on the standard curve; (2) set the maximum acceptable A450 nm signal as the reading for the third titration point on the standard curve; (3) set the minimum acceptable A450 nm signal as the reading for the seventh titration point on the standard curve; (4) mask all samples titration points that fall above the maximum acceptable signal and below the minimum acceptable signal; (5) user the reference standard 4PL curve fit to calculate the concentration for each titration point with an acceptable A450 signal and multiply that value by the dilution factor of that titration point; (6) for each sample titration, calculate the mean value for the calculated concentration across titration series. ELISA results were converted to percent of antibody remaining based on the day 0 timepoint representing 100%. As shown inFIGS.13A-13B, all three antibody variants demonstrated extended half-life in this well-established mouse model of antibody half-life. Motavizumab wild-type has a half-life of 32 hours. FcMut043 and FcMut045 mutants built on FcMut008 show significant half-life improvement. FcMut045 mutant enhanced half-life 5.2 fold (about 166 hours half-life). The half-life as well as the other parameters inFIG.13Bwere calculated using the Winonlin software. Similar experiments were conducted with later stage Fc variants in the context of Motazivumab. When the Motavizumab variants were administered at a dose of 5 mg/kg, next generation variants (FcMut171, FcMut183, FcMut186, and FcMut197) demonstrated further enhanced half-life with more than nine fold increase in half-life observed with FcMut213 (FIGS.14A-14B). One of the early variants (FcMut045) was included to demonstrate the improved half-life seen with the later stage designs as compared to the early stage designs. Similar results were observed when the Motavizumab variants were administered at a dose of 2 mg/kg (FIGS.14C-14D). To evaluate if the enhanced binding of Fc variants to human FcRn translated to increased serum persistence and longer circulating half-life life, a pharmacokinetic study of IgGs containing the different Fc variants was performed in Tg276 transgenic mice. Transgenic mouse models expressing human FcRn have been developed by the Jackson laboratory (JAX) to study PK of human Fc-containing biotherapeutics. The Tg276 mice are null for mouse FcRn alpha chain and express the human FcRn alpha transgene under the control of a constitutive promoter (actin) and use the mouse β2 microglobulin. The Tg276 homozygous or hemizygous mice have been widely used to differentiate half-lives of antibody variants. For the in vivo study, Tg276hemi FcRn transgenic mice were dosed with 2 or 5 mg/kg of mAb intravenously. Each mAb group had 4 mice/group. Blood was collected at several time points between 1 hour and 21 days, and IgG titers were determined by quantitative ELISA as described herein. PK parameters were determined for each group of mice with a non-compartmental model using Phoenix WinNonlin version 7.0 (Certera). The results are shown inFIGS.14A-14D. Example 9: In Vivo Assessment of Half-Life of Engineered Antibodies Two Zika antibodies (ZVA and ZVB) as well as wild type Motavizumab (MVZ) were administered to mice transgenic for human FcRn at a dose of 2-5 mg/kg, and daily samples of mouse serum were obtained (day 0-day 4). ELISA was performed on serum to quantify the amount of Motavizumab. ELISA results were converted to percent of antibody remaining based on the day 0 timepoint representing 100%. ZVA antibody represents the A series antibody A-3/2. The ZVB antibody represents the A series antibody A-5/1 containing the affinity enhancing light chain modification S92Y. As shown inFIG.15, ZVB had a much longer half-life than either Motavizumab or ZVA (both ZVA and ZVB had longer half-life than Motavizumab). These antibodies contained wild-type Fc regions so the extended half-life of the ZVB antibody is a property of the antibody itself, and not any Fc engineering. In the next experiment, Motavizumab wild type (MVZ WT) was tested against the ZVB antibody (ZB-1/4, ZKB in the graph) and the ZVB antibody containing Fc modification 156 (L234A/L235A (“LALA”) and T256D/T307R/Q311V (half-life extension), ZKB-156 in the graph) as well as the “LS” half-life extension Fc mutation (ZKB-LS). Both LS and Fc modification 156 extended half-life significantly compared to the wild-type ZVB (FIG.16). These data demonstrate that the Visterra mutation is comparable to the literature derived LS mutation, and also demonstrates that the FC engineering efforts described herein can extend the half-life of an antibody that already has a very long half-life in this transgenic mouse model. “ZVB” is indicated as “ZKB” inFIG.16. Example 10: Binding to Fc Receptors and Impact on Effector Functions Exemplary Fc variants were assessed on multiple assays for FcγRI, FcγRIIA (mediator of opsonophagocytosis), FcγRIIB, FcγRIIIA (mediator of ADCC), and C1q (mediator of CDC). Binding to Fcγ receptors I, IIa, IIb, IIIc, and IIIc V176F (R&D Systems catalogue #1257-FC-050, 1330-CD-050, 1875-CD-050, 4325-FC-050, and 8894-FC-050) was measured by ELISA. All Fc variants were tested in the context of the Motavizumab, Rituximab, or Actoxumab antibody. Briefly, Fc receptors were coated on an ELISA plate (VWR catalogue #62409-002) at 1 μg/mL (0.1 μg/well) in PBS and stored at 4° C. overnight. Plates were washed three times with PBST (1×PBS 0.05% Tween20). Antibodies were titrated threefold in PBST-BSA from 100 μg/mL to 0.05 μg/mL and 100 μL was added to each well of the ELISA plate and incubated for 1 hour at room temperature. Plates were washed three times with PBST. Goat anti-human Fc (Jackson catalogue #109-035-098) was diluted 1:5000 in PBST-BSA and 100 μL was added to each well and incubated for 1 hour at 4 C. Plates were washed six times with PBST. Plates were developed using the TMB Microwell Peroxidase Substrate Kit (VWR catalogue #95059-156). The reaction was stopped after 10 minutes by the addition of 1N sulfuric acid and absorbance at 450 nm was measured. The values of antibody concentration (x-axis) and absorbance at 450 nm (y-axis) were fit to a four parameter logistic regression (4PL) curve. The curve fit was then used to determine the EC50 (the midpoint of the 4PL) for each Fc variant. Binding to Fcγ receptors IIa and IIb was measured by BioLayer Interferometry using the Octet QKe system. Briefly, FcγIIa and FcγIIb (R&D Systems catalogue #1330-CD-050 and 1875-CD-050) were diluted to 5 μg/mL in PBS. The receptors were immobilized via a C-terminal 6× histidine tag (SEQ ID NO: 2) to Ni-NTA biosensors (Pall catalogue #18-5101) for 180 seconds followed by a 60 second baseline step in PBS. The biosensors were then exposed to the various Fc variants at a concentration of 50 μg/mL in PBS for 120 seconds followed by a dissociation step in PBS for an additional 120 seconds. The max binding response during the association step for each variant was reported and compared to the wild type Fc response. Binding to C1q was measured by ELISA. All Fc variants were tested in the context of the Motavizumab antibody. Briefly, antibodies were coated on an ELISA plate (VWR catalogue #62409-002) at 25 μg/mL (2.5 μg/well) in PBS and stored at 4° C. overnight. Plates were washed three times with PBST (1×PBS 0.05% Tween 20). Purified C1q (Quidel Corporation catalogue # A400) was titrated threefold in PBST-BSA (1×PBS 0.05% Tween20 1% BSA) from 12.5 μg/mL to 0.02 μg/mL and incubated for 90 minutes at room temperature. Liquid was aspirated from wells and polyclonal rabbit anti-human C1q (Agilent catalogue # A013602-1) was diluted in PBST-BSA to a final concentration of 1 μg/mL and 100 μL was added to each well and incubated for 1 hour at room temperature. Plates were washed three times with PBST. Polyclonal swine anti-rabbit-HRP (Agilent catalogue # P021702-2) was diluted to 0.5 μg/mL in PBST-BSA and 100 μL was added to each well and incubated for 1 hour at room temperature. Plates were washed six times with PBST. Plates were developed using the TMB Microwell Peroxidase Substrate Kit (VWR catalogue #95059-156). The reaction was stopped after 10 minutes by the addition of 1N sulfuric acid and absorbance at 450 nm was measured. The values of antibody concentration (x-axis) and absorbance at 450 nm (y-axis) were fit to a four parameter logistic regression (4PL) curve. The curve fit was then used to determine the EC50 (the midpoint of the 4PL) for each Fc variant. The results are shown inFIGS.17A-17D. Example 11: CDC Activity of Engineered Antibodies Complement dependent cytotoxicity (CDC) activity of exemplary Fc variants (Rituximab Fab) was examined. CDC assays were performed using CD20+ Raji cells and low toxicity guinea pig complement (Cedarlane Laboratories Product # CL4051). Complement induced cell lysis was measured using the CYTOTOX 96® Non-Radioactive Cytotoxicity Assay from Promega (catalogue # G1780) following the manufacturer's protocol. All Fc variants were tested in the context of the anti-CD20 antibody, Rituximab. Briefly, antibody concentrations were titrated fourfold ranging from 20 μg/mL to 0.005 μg/mL and incubated with 20,000 target cells per well at 37° C. for 30 minutes. Complement was then added to the cells and incubated an additional 2 hours at 37° C. Additionally, cell lysis buffer provided with the CYTOTOX kit was added to control wells to measure the maximum cell lysis. A negative control antibody, Motavizumab, was used to measure the background signal of an irrelevant antibody. The background signal and maximum lysis signal were used to calculate the percent of cell lysis for each Fc variant. The values of antibody concentration (x-axis) and percent lysis (y-axis) were fit to a four parameter logistic regression curve. The curve fit was then used to determine the EC50 (concentration needed to obtain 50% lysis) and the maximum lysis for each Fc variant. The results are shown inFIG.18. Example 12: ADCC Activity of Engineered Antibodies Antibody dependent cellular cytotoxicity (ADCC) activity of exemplary Fc variants (Rituximab Fab) was examined. ADCC assays were performed using the ADCC Reporter Bioassay with CD20+WIL2-S target cells from Promega (catalogue # G7014) following the manufacturer's protocol. All Fc variants were tested in the context of the anti-CD20 antibody, Rituximab. Antibody concentrations were titrated fivefold ranging from 5 μg/mL to 0.0016 μg/mL. The values of antibody concentration (x-axis) and fold induction of the luminescent reporter gene (y-axis) were fit to a four parameter logistic regression (4PL) curve. The curve fit was then used to determine the EC50 (the midpoint of the 4PL) and the maximum induction for each Fc variant. The results are shown inFIGS.19A-19B. Exemplary Fc variants retain and in some cases enhance ADCC activity. Example 13: ADIN Activity of Engineered Antibodies TRIM21 is a cytosolic receptor that binds with Fc of IgG. TRIM21 plays a role in mediating intracellular recognition and neutralization of Fc bound viruses. TRIM21-mediated neutralization is known as antibody dependent intracellular neutralization (ADIN). Binding to TRIM21 was measured by ELISA. All Fc variants were tested in the context of the Motavizumab or Actoxumab antibody. Briefly, antibodies were coated on an ELISA plate (VWR catalogue #62409-002) at 25 μg/mL (2.5 μg/well) in PBS and stored at 4° C. overnight. Plates were washed three times with PBST (1×PBS 0.05% Tween20). TRIM21-GST (Antibodies Online catalogue # ABIN1323621) was titrated threefold in PBST-BSA (1×PBS 0.05% Tween20 1% BSA) from 12.5 μg/mL to 0.02 μg/mL and incubated for 90 minutes at room temperature. Liquid was aspirated from wells and two rabbit anti-TRIM21 antibodies (AbCam catalogue # ab91423 and ab96800) were mix together in PBST-BSA at a final concentration of 1 μg/mL each and 100 μL was added to each well and incubated for 1 hour at room temperature. Plates were washed three times with PBST. Polyclonal swine anti-rabbit-HRP (Agilent catalogue # P021702-2) was diluted to 0.5 μg/mL in PBST-BSA and 100 μL was added to each well and incubated for 1 hour at room temperature. Plates were washed six times with PBST. Plates were developed using the TMB Microwell Peroxidase Substrate Kit (VWR catalogue #95059-156). The reaction was stopped after 10 minutes by the addition of 1N sulfuric acid and absorbance at 450 nm was measured. The values of antibody concentration (x-axis) and absorbance at 450 nm (y-axis) were fit to a four parameter logistic regression (4PL) curve. The curve fit was then used to determine the EC50 (the midpoint of the 4PL) for each Fc variant. A TRIM21 binding ELISA was performed to evaluate if the Fc variants impacted binding to TRIM21. The results are shown inFIG.26. Fc variants FcMut045, FcMut183, FcMut197, FcMut213, FcMut215, FcMut228 had TRIM21 binding EC50s that were within 1.5 fold of WT EC50. Example 14: Enhancement of Mucosal Uptake by Fc Mutations FcRn transports IgG across different cellular barriers such as the mucosal epithelium lining the intestine and the alveolar surfaces. Modification of FcRn binding provides a mechanism to enhance mucosal localization that confers immune protection. Exemplary Fc mutants are expected to enhance mucosal uptake in a similar fashion Example 15: Impact of FcRn Affinity Enhancing Mutations on Engagement with Fc Receptors and Effector Functions The impact of FcRn affinity enhancing mutations on binding of Fc to other receptors was experimentally evaluated. The Fc region of IgG is capable of binding to many different Fc receptors and the mechanism of action for many therapeutic antibodies relies on engagement with Fc receptors. Mutations, even at positions distant from the Fc receptor binding site, can impact engagement of Fc with receptors such as FcγRI, FcγRIIa/b, FcγRIIIa, C1q, TRIM21. While Fc receptor TRIM21 binds to a site that overlaps with FcRn binding site on CH2-CH3, other receptors such Fcγ receptors and C1q engage at the interface formed by dimeric CH2. As such any mutations introduced for enhancing half-life should be assessed for its impact on binding to other receptors. Mutations introduced to enhance half-life can The Fc variants (FcMut183, FcMut197, FcMut213, FcMut215, FcMut228, FcMut229, YTE, LS) were incorporated into rituximab Fab and together with WT rituximab, were evaluated for binding to FcγRI, FcγRIIa, FcγRIIb, FcγRIIIa, C1q and TRIM21. As shown inFIG.24, introduction of Fc variants FcMut183, FcMut197, FcMut213, FcMut215, FcMut228 and FcMut229 did not significantly impact engagement with the tested Fc receptors. While binding to Fc receptors is necessary for activities such as ADCC (FcγRIIIa), CDC (C1q) and ADIN (TRIM21), it may not be sufficient in mediating these activities. For example, a hexameric assembly of Fc is required to elicit CDC activity. The Fc hexamer assembly involves formation of an Fc-Fc interface, and residues involved in formation of this interface partially overlaps with FcRn binding site. As such, it was important to evaluate the impact of the Fc mutations on not just binding to Fc receptors but also activities such as CDC and ADCC. Rituximab with various Fc variants (FcMut183, FcMut197, FcMut213, FcMut215, FcMut228, FcMut229, YTE, LS, WT) were evaluated for ADCC and CDC activities as described in the methods section. The results confirmed that Fc variants FcMut183, FcMut197, FcMut213, FcMut215, FcMut228 and FcMut229 did not negatively impact ADCC activity and in fact showed a slight enhancement in CDC activity. Rituximab containing YTE had no detectable CDC activity and significantly reduced ADCC activity. Similarly another mutant FcMut197 had reduced ADCC and CDC activity. Inspects of the network map of these mutations provides an insight into the possible reason for loss of ADCC and CDC activity. Example 16: Persistence of Optimized Fc Variants in Sera of Transgenic Mice To evaluate whether the enhanced binding of Fc variants to human FcRn translated to increased serum persistence and longer circulating half-life life, a pharmacokinetic study of IgGs containing the different Fc variants was performed in Tg276 transgenic mice. Transgenic mouse models expressing human FcRn can be used to study PK of human Fc-containing biotherapeutics (Roopenian et al.,Methods Mol Biol,2010. 602: 93-104; Petkova et al.,Int Immunol,2006. 18(12): 1759-69). The Tg276 mice are null for mouse FcRn alpha chain and express the human FcRn alpha transgene under the control of a constitutive promoter (actin) and use the mouse β2microglobuli. The Tg276 homozygous and hemizygous mice can be used to differentiate half-lives of antibody variants. For the PK study, Tg276hemi FcRn transgenic mice were intravenously administered with 5 mg/kg of IgG (motavizumab Fab on WT or modified Fc). Retro-orbital blood collection was performed at several time points, and IgG titers were determined by quantitative ELISA. IgGs containing the engineered Fc variants persisted in serum much longer than wild-type Motavizumab (FIG.25A). Pharmacokinetic parameters from non-compartmental analysis indicates Motavizumab with Fc variants have greater than two fold slower clearance rate and significant increases in β-phase elimination half-life and area under the curve (AUC) measurements (FIG.25B). INCORPORATION BY REFERENCE All publications, patents, and Accession numbers mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. EQUIVALENTS While specific embodiments of the subject invention have been discussed, the above specification is illustrative and not restrictive. Many variations of the invention will become apparent to those skilled in the art upon review of this specification and the claims below. The full scope of the invention should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.
239,678
11858981
DETAILED DESCRIPTION OF THE INVENTION Definitions In order that the present invention may be more readily understood, certain terms will be defined as follows. Additional definitions are set forth throughout the detailed description. The term “antibody” refers to whole antibodies and any antigen binding fragment. The term “antigen binding polypeptide” and “immunobinder” are used simultaneously herein. An “antibody” refers to a protein, optionally glycosylated, comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VHand VLregions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VHand VLis composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system. The term “antigen-binding portion” of an antibody (or simply “antibody portion”) refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., TNF). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VHand CH1 domains; (iv) a Fv fragment consisting of the VLand VHdomains of a single arm of an antibody, (v) a single domain or dAb fragment (Ward et al., (1989)Nature341:544-546), which consists of a VHdomain; and (vi) an isolated complementarity determining region (CDR) or (vii) a combination of two or more isolated CDRs which may optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VLand VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VLand VHregions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988)Science242:423-426; and Huston et al. (1988)Proc. Natl. Acad. Sci. USA85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins. Antibodies can be of different isotype, for example, an IgG (e.g., an IgG1, IgG2, IgG3, or IgG4 subtype), IgA1, IgA2, IgD, IgE, or IgM antibody. The term “immunobinder” refers to a molecule that contains all or a part of the antigen binding site of an antibody, e.g. all or part of the heavy and/or light chain variable domain, such that the immunobinder specifically recognizes a target antigen. Non-limiting examples of immunobinders include full-length immunoglobulin molecules and scFvs, as well as antibody fragments, including but not limited to (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CLand CH1 domains; (ii) a F(ab′)2fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fab′ fragment, which is essentially a Fab with part of the hinge region (see, FUNDAMENTAL IMMUNOLOGY (Paul ed., 3.sup.rd ed. 1993); (iv) a Fd fragment consisting of the VHand CH1 domains; (v) a Fv fragment consisting of the VLand VHdomains of a single arm of an antibody, (vi) a single domain antibody such as a Dab fragment (Ward et al., (1989)Nature341:544-546), which consists of a VHor VLdomain, a Camelid (see Hamers-Casterman, et al., Nature 363:446-448 (1993), and Dumoulin, et al., Protein Science 11:500-515 (2002)) or a Shark antibody (e.g., shark Ig-NARs Nanobodies®; and (vii) a nanobody, a heavy chain variable region containing a single variable domain and two constant domains. The term “single chain antibody”, “single chain Fv” or “scFv” is refers to a molecule comprising an antibody heavy chain variable domain (or region; VH) and an antibody light chain variable domain (or region; VL) connected by a linker. Such scFv molecules can have the general structures: NH2-VL-linker-VH-COOH or NH2-VH-linker-VL-COOH. A suitable state of the art linker consists of repeated GGGGS (SEQ ID NO: 26) amino acid sequences or variants thereof. In a preferred embodiment of the present invention a (GGGGS)4(SEQ ID NO: 8) linker is used, but variants of 1-3 repeats are also possible (Holliger et al. (1993), Proc. Natl. Acad. Sci. USA 90:6444-6448). Other linkers that can be used for the present invention are described by Alfthan et al. (1995), Protein Eng. 8:725-731, Choi et al. (2001), Eur. J. Immunol. 31:94-106, Hu et al. (1996), Cancer Res. 56:3055-3061, Kipriyanov et al. (1999), J. Mol. Biol. 293:41-56 and Roovers et al. (2001), Cancer Immunol. As used herein, the term “functional property” is a property of a polypeptide (e.g., an immunobinder) for which an improvement (e.g., relative to a conventional polypeptide) is desirable and/or advantageous to one of skill in the art, e.g., in order to improve the manufacturing properties or therapeutic efficacy of the polypeptide. In one embodiment, the functional property is stability (e.g., thermal stability). In another embodiment, the functional property is solubility (e.g., under cellular conditions). In yet another embodiment, the functional property is aggregation behavior. In still another embodiment, the functional property is protein expression (e.g., in a prokaryotic cell). In yet another embodiment the functional property is refolding behavior following inclusion body solubilization in a manufacturing process. In certain embodiments, the functional property is not an improvement in antigen binding affinity. In another preferred embodiment, the improvement of one or more functional properties has no substantial effect on the binding affinity of the immunobinder. The term “CDR” refers to one of the six hypervariable regions within the variable domains of an antibody that mainly contribute to antigen binding. One of the most commonly used definitions for the six CDRs was provided by Kabat E. A. et al., (1991) Sequences of proteins of immunological interest. NIH Publication 91-3242). As used herein, Kabat's definition of CDRs only apply for CDR1, CDR2 and CDR3 of the light chain variable domain (CDR L1, CDR L2, CDR L3, or L1, L2, L3), as well as for CDR2 and CDR3 of the heavy chain variable domain (CDR H2, CDR H3, or H2, H3). CDR1 of the heavy chain variable domain (CDR H1 or H1), however, as used herein is defined by the residue positions (Kabat numbering) starting with position 26 and ending prior to position 36. This definition is basically a fusion of CDR H1 as differently defined by Kabat and Chotia (see alsoFIG.1for illustration). The term “antibody framework” as used herein refers to the part of the variable domain, either VL or VH, which serves as a scaffold for the antigen binding loops (CDRs) of this variable domain. In essence it is the variable domain without the CDRs. The term “epitope” or “antigenic determinant” refers to a site on an antigen to which an immunoglobulin or antibody specifically binds (e.g., a specific site on the TNF molecule). An epitope typically includes at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 consecutive or non-consecutive amino acids in a unique spatial conformation. See, e.g.,Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996). The terms “specific binding,” “selective binding,” “selectively binds,” and “specifically binds,” refer to antibody binding to an epitope on a predetermined antigen. Typically, the antibody binds with an affinity (KD) of approximately less than 10−7M, such as approximately less than 10−8M, 10−9M or 10−10M or even lower. The term “KD” or “Kd” refers to the dissociation equilibrium constant of a particular antibody-antigen interaction. Typically, the antibodies of the invention bind to TNF with a dissociation equilibrium constant (KD) of less than approximately 10−7M, such as less than approximately 10−8M, 10−9M or 10−10M or even lower, for example, as determined using surface plasmon resonance (SPR) technology in a BIACORE instrument. The term “nucleic acid molecule,” as used herein refers to DNA molecules and RNA molecules. A nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA. A nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence. The term “vector,” refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. In one embodiment, the vector is a “plasmid,” which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. In another embodiment, the vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. The vectors disclosed herein can be capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors) or can be can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome (e.g., non-episomal mammalian vectors). The term “host cell” refers to a cell into which and expression vector has been introduced. Host cells include bacterial, microbial, plant or animal cells, preferably,Escherichia coli, Bacillus subtilis; Saccharomyces cerevisiae, Pichia pastoris, CHO (Chinese Hamster Ovary lines) or NSO cells. The term “lagomorphs” refers to members of the taxonomic order Lagomorpha, comprising the families Leporidae (e.g. hares and rabbits), and the Ochotonidae (pikas). In a most preferred embodiment, the lagomorphs is a rabbit. The term “rabbit” as used herein refers to an animal belonging to the family of the leporidae. As used herein, “identity” refers to the sequence matching between two polypeptides, molecules or between two nucleic acids. When a position in both of the two compared sequences is occupied by the same base or amino acid monomer subunit (for instance, if a position in each of two polypeptides is occupied by a lysine), then the respective molecules are identical at that position. The “percentage identity” between two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. Generally, a comparison is made when two sequences are aligned to give maximum identity. Such alignment can be provided using, for instance, the method of the Needleman and Wunsch (J. Mol. Biol. (48):444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package, using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. Various aspects of the invention are described in further detail in the following subsections. It is understood that the various embodiments, preferences and ranges may be combined at will. Further, depending of the specific embodiment, selected definitions, embodiments or ranges may not apply. If not otherwise stated, the amino acid positions are indicated according to the AHo numbering scheme. The AHo numbering system is described further in Honegger, A. and Pluckthun, A. (2001)J. Mol. Biol.309:657-670). Alternatively, the Kabat numbering system as described further in Kabat et al. (Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) may be used. Conversion tables for the two different numbering systems used to identify amino acid residue positions in antibody heavy and light chain variable regions are provided in A. Honegger, J. Mol. Biol. 309 (2001) 657-670. In a first aspect, the present invention provides a universal acceptor framework for the grafting of CDRs from other animal species, for example, from rabbit. It has previously been described that antibodies or antibody derivatives comprising the human frameworks identified in the so called “Quality Control” screen (WO0148017) are characterised by a generally high stability and/or solubility. Although the human single-chain framework FW1.4 (a combination of SEQ ID NO: 1 (named a43 in WO03/097697) and SEQ ID NO: 2 (named KI27 in WO03/097697)) clearly underperformed in the Quality Control assay, it was surprisingly found that it has a high intrinsic thermodynamic stability and is well producible, also in combination with a variety of different CDRs. The stability of this molecule can be attributed mostly to its framework regions. It has further been shown that FW1.4 is in essence highly compatible with the antigen-binding sites of rabbit antibodies. Therefore, the FW1.4 represents a suitable scaffold to construct stable humanized scFv antibody fragments derived from grafting of rabbit loops. Thus, in one aspect, the invention provides an immunobinder acceptor framework, comprising a VH sequence having at least 90% identity to SEQ ID No. 1 and/or a VL sequence having at least 85% identity to SEQ ID No. 2, more preferably comprising the sequence of FW1.4 (SEQ ID NO: 3) for the grafting of rabbit CDRs, or a sequence having at least 60%, more preferably at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, identity to SEQ ID NO: 3. Moreover, it was found that FW1.4 could be optimized by substituting several residue positions in the heavy chain of FW1.4 and/or by substituting 1 position in the light chain of FW1.4. Thereby, it was surprisingly found that loop conformation of a large variety of rabbit CDRs in the VH could be fully maintained, largely independent of the sequence of the donor framework. Said residues in the heavy chain as well as the 1 position in the light chain of FW1.4 are conserved in rabbit antibodies. The consensus residue for the positions in the heavy chain as well as the one position in the light chain was deduced from the rabbit repertoire and introduced into the sequence of the human acceptor framework. As a result, the modified framework 1.4 (hereinafter referred to as rFW1.4) is compatible with virtually any rabbit CDRs. Moreover, rFW1.4 containing different rabbit CDRs is well expressed and good produced contrary to the rabbit wild type single chains and still almost fully retains the affinity of the original donor rabbit antibodies. Thus, the present invention provides the variable heavy chain framework of SEQ ID No. 1, further comprising one or more amino acid residues that generally support conformation of CDRs derived from a rabbit immunobinder. In particular, said residues are present at one or more amino acid positions selected from the group consisting of 24H, 25H, 56H, 82H, 84H, 89H and 108H (AHo numbering). These positions are prove to affect CDR conformation and are therefore contemplated for mutation to accommodate donor CDRs. Preferably, said one or more residues are selected from the group consisting of: Threonine (T) at position 24, Valine (V) at position 25, Glycine or Alanine (G or A) at position 56, Lysine (K) at position 82, Threonine (T) at position 84, Valine (V) at position 89 and Arginine (R) at position 108 (AHo numbering). Preferably, at least three, more preferably, four, five, six and most preferably all seven residues are present. Surprisingly, it has been found that the presence of the mentioned residues improves the stability of the immunobinder. In a preferred embodiment, the invention provides an immunobinder acceptor framework comprising a VH having at least 50%, more preferably at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% and eve more preferably 100% identity to SEQ ID No. 4, with the provision that at least one, more preferably at least three, more preferably, four, five, six and most preferably seven residues of the group consisting of threonine (T) at position 24, valine (V) at position 25, alanine (A) or glycine (G) at position 56, threonine (T) at position 84, lysine (K) at position 82, valine (V) at position 89 and arginine (R) at position 108 (AHo numbering) are present. In a preferred embodiment, the immunobinder acceptor framework is an immunobinder acceptor framework for rabbit CDRs. In a preferred embodiment, said variable heavy chain framework is or comprises SEQ ID No. 4 or SEQ ID No. 6. Both of said variable heavy chain frameworks may for example be combined with any suitable light chain framework. Accordingly, the present invention provides an immunobinder acceptor framework comprising(i) a variable heavy chain framework having at least 70% identity, preferably at least 75%, 80%, 85%, 90%, more preferably at least 95% identity, to SEQ ID No. 4; and/or(ii) a variable light chain framework having at least 70% identity, preferably at least 75%, 80%, 85%, 90%, more preferably at least 95% identity, to SEQ ID No. 2. In a much preferred embodiment, the variable heavy chain framework comprises threonine (T) at position 24, glycine (G) at position 56, threonine (T) at position 84, valine (V) at position 89 and arginine (R) at position 108 (AHo numbering). In a preferred embodiment, the variable light chain comprises Threonine (T) at position 87 (AHo numbering). In a preferred embodiment, said immunobinder acceptor framework comprises(i) a variable heavy chain framework selected from the group consisting of SEQ ID No. 1, SEQ ID No. 4 and SEQ ID No. 6; and/or(ii) a variable light chain framework of SEQ ID No. 2 or SEQ ID No. 9. In a preferred embodiment, the variable heavy chain framework is linked to a variable light chain framework via a linker. The linker may be any suitable linker, for example a linker comprising 1 to 4 repeats of the sequence GGGGS (SEQ ID NO: 26), preferably a (GGGGS)4peptide (SEQ ID No. 8), or a linker as disclosed in Alfthan et al. (1995) Protein Eng. 8:725-731. In another preferred embodiment, the immunobinder acceptor framework is a sequence having at least 70%, 75%, 80%, 85%, 90% more preferably at least 95% identity, to SEQ ID No. 5, whereas the sequence, preferably, is not SEQ ID No. 3. More preferably, the immunobinder acceptor framework comprises or is SEQ ID No. 5. In another preferred embodiment, the immunobinder acceptor framework is a sequence having at least 70%, 75%, 80%, 85%, 90%, more preferably at least 95% identity, to SEQ ID No. 7, whereas the sequence, preferably, is not SEQ ID No. 3. More preferably, the immunobinder acceptor framework comprises or is SEQ ID No. 7. Moreover, it was surprisingly found that the presence of the above described amino acid motif renders a framework, preferably a human framework, particularly suitable for the accommodation of CDRs from other non-human animal species, in particular rabbit CDRs. Said motif has no negative impact on the stability of an immunobinder. The CDRs are presented in a conformation similar to their native spatial orientation in the rabbit immunobinder; thus, no structurally relevant positions need to be grafted onto the acceptor framework. Accordingly, the human or humanized immunobinder acceptor framework comprises at least three amino acids, preferably four, five, six and more preferably seven amino acids of the group consisting of threonine (T) at position 24, valine (V) at position 25, alanine (A) or glycine (G) at position 56, lysine (K) at position 82, threonine (T) at position 84, valine (V) at position 89 and arginine (R) at position 108 (AHo numbering). The immunobinder acceptor frameworks as described herein may comprise solubility enhancing substitution in the heavy chain framework, preferably at positions 12, 103 and 144 (AHo numbering). Preferably, a hydrophobic amino acid is substituted by a more hydrophilic amino acid. Hydrophilic amino acids are e.g. Arginine (R), Asparagine (N), Aspartic acid (D), Glutamine (Q), Glycine (G), Histidine (H), Lysine (K), Serine (S) and Threonine (T). More preferably, the heavy chain framework comprises (a) Serine (S) at position 12; (b) Serine (S) or Threonine (T) at position 103 and/or (c) Serine (S) or Threonine (T) at position 144. Moreover, stability enhancing amino acids may be present at one or more positions 1, 3, 4, 10, 47, 57, 91 and 103 of the variable light chain framework (AHo numbering). More preferably, the variable light chain framework comprises glutamic acid (E) at position 1, valine (V) at position 3, leucine (L) at position 4, Serine (S) at position 10; Arginine (R) at position 47, Serine (S) at position 57, phenylalanine (F) at position 91 and/or Valine (V) at position 103. As glutamine (Q) is prone to deamination, in another preferred embodiment, the VH comprises at position 141 a glycine (G). This substitution may improve long-term storage of the protein. For example, the acceptor frameworks disclosed herein can be used to generate a human or humanized antibody which retains the binding properties of the non-human antibody from which the non-human CDRs are derived. Accordingly, in a preferred embodiment the invention encompasses an immunobinder acceptor framework as disclosed herein, further comprising heavy chain CDR1, CDR2 and CDR3 and/or light chain CDR1, CDR2 and CDR3 from a donor immunobinder, preferably from a mammalian immunobinder, more preferably from a lagomorph immunobinder and most preferably from a rabbit. Thus, in one embodiment, the invention provides an immunobinder specific to a desired antigen comprising(i) variable light chain CDRs of a lagomorph; and(ii) a human variable heavy chain framework having at least 50% identity to SEQ ID NO. 4. In one preferred embodiment, there is the provision that at least one amino acid of the group consisting of threonine (T) at position 24, valine (V) at position 25, alanine (A) or glycine (G) at position 56, threonine (T) at position 84, lysine (K) at position 82, valine (V) at position 89 and arginine (R) at position 108 (AHo numbering) is present in said human variable heavy chain framework sequence. Preferably, the lagomorph is a rabbit. More preferably, the immunobinder comprises heavy chain CDR1, CDR2 and CDR3 and light chain CDR1, CDR2 and CDR3 from the donor immunobinder. As known in the art, many rabbit VH chains have extra paired cysteines relative to the murine and human counterparts. In addition to the conserved disulfide bridge formed between cys22 and cys92, there is also a cys21-cys79 bridge as well as an interCDR S—S bridge formed between the last residue of CDRH1 and the first residue of CDR H2 in some rabbit chains. Besides, pairs of cysteine residues in the CDR-L3 are often found. Besides, many rabbit antibody CDRs do not belong to any previously known canonical structure. In particular the CDR-L3 is often much longer than the CDR-L3 of a human or murine counterpart. As stated before, the grafting of the non-human CDRs onto the frameworks disclosed herein yields a molecule wherein the CDRs are displayed in a proper conformation. If required, the affinity of the immunobinder may be improved by grafting antigen interacting framework residues of the non-human donor immunobinder. These positions may e.g. be identified by(i) identifying the respective germ line progenitor sequence or, alternatively, by using the consensus sequences in case of highly homologous framework sequences;(ii) generating a sequence alignment of donor variable domain sequences with germ line progenitor sequence or consensus sequence of step (i); and(iii) identifying differing residues. Differing residues on the surface of the molecule were in many cases mutated during the affinity generation process in vivo, presumably to generate affinity to the antigen. In another aspect, the present invention provides an immunobinder which comprises the immunobinder acceptor framework described herein. Said immunobinder may e.g. be a scFv antibody, a full-length immunoglobulin, a Fab fragment, a Dab or a Nanobody. In a preferred embodiment, the immunobinder is attached to one or more molecules, for example a therapeutic agent such as a cytotoxic agent, a cytokine, a chemokine, a growth factor or other signaling molecule, an imaging agent or a second protein such as a transcriptional activator or a DNA-binding domain. The immunobinder as disclosed herein may e.g. be used in diagnostic applications, therapeutic application, target validation or gene therapy. The invention further provides an isolated nucleic acid encoding the immunobinder acceptor framework disclosed herein or the immunobinder(s) as disclosed herein. In another embodiment, a vector is provided which comprises the nucleic acid disclosed herein. The nucleic acid or the vector as disclosed herein can e.g. be used in gene therapy. The invention further encompasses a host cell comprising the vector and/or the nucleic acid disclosed herein. Moreover, a composition is provided, comprising the immunobinder acceptor framework as disclosed herein, the immunobinder as disclosed herein, the isolated nucleic acid as disclosed herein or the vector as disclosed herein. The sequences disclosed herein are the following (X residues are CDR insertion sites): SEQ ID NO. 1:variable heavy chain framework of FW1.4 (a43)EVQLVESGGGLVQPGGSLRLSCAAS(X)n=1−50WVRQAPGKGLEWVS (X)n=1−50RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK(X)n=1−50WGQGTLVTVSSSEQ ID NO. 2:variable light chain framework of FW1.4 (KI27)EIVMTQSPSTLSASVGDRVIITC(X)n=1−50WYQQKPGKAPKLLIY(X)n=1−50GVPSRFSGSGSGAEFTLTISSLQPDDFATYYC(X)n=1−50FGQGTKLTVLGSEQ ID NO. 3:framework of FW1.4EIVMTQSPSTLSASVGDRVIITC(X)n=1−50WYQQKPGKAPKLLIY(X)n=1−50GVPSRFSGSGSGAEFTLTISSLQPDDFATYYC(X)n=1−50FGQGTKLTVLGGGGGSGGGGSGGGGSGGGGS EVQLVESGGGLVQPGGSLRLSCAAS(X)n=1−50WVRQAPGKGLEWVS(X)n=1−50RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK(X)n=1−50WGQGTLVTVSSSEQ ID NO. 4:variable heavy chain framework of rFW1.4EVQLVESGGGLVQPGGSLRLSCTAS(X)n=1−50WVRQAPGKGLEWVG(X)n=1−50RFTISRDTSKNTVYLQMNSLRAEDTAVYYCAR(X)n=1−50WGQGTLVTVSSSEQ ID NO. 5:framework of rFW1.4EIVMTQSPSTLSASVGDRVIITC(X)n=1−50WYQQKPGKAPKLLIY(X)n=1−50GVPSRFSGSGSGTEFTLTISSLQPDDFATYYC(X)n=1−50FGQGTKLTVLGGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQPGGSLRLSCTAS(X)n=1−50WVRQAPGKGLEWVG(X)n=1−50RFTISRDTSKNTVYLQMNSLRAEDTAVYYCAR(X)n=1−50WGQGTLVTVSSSEQ ID NO. 6:variable heavy chain framework of rFW1.4(V2)EVQLVESGGGLVQPGGSLRLSCTVS(X)n=1−50WVRQAPGKGLEWVG(X)n=1−50RFTISKDTSKNTVYLQMNSLRAEDTAVYYCAR(X)n=1−50WGQGTLVTVSSSEQ ID NO. 7:framework of rFW1.4(V2)EIVMTQSPSTLSASVGDRVIITC(X)n=1−50WYQQKPGKAPKLLIY(X)n=1−50GVPSRFSGSGSGTEFTLTISSLQPDDFATYYC(X)n=1−50FGQGTKLTVLGGGGGSGGGGSGGGGSGGGGS EVQLVESGGGLVQPGGSLRLSCTVS(X)n=1−50WVRQAPGKGLEWVG(X)n=1−50RFTISKDTSKNTVYLQMNSLRAEDTAVYYCAR(X)n=1−50WGQGTLVTVSSSEQ ID NO. 8:linkerGGGGSGGGGSGGGGSGGGGSSEQ ID NO. 9:substituted variable light chain framework of FW1.4EIVMTQSPSTLSASVGDRVIITC(X)n=1−50WYQQKPGKAPKLLIY(X)n=1−50GVPSRFSGSGSGTEFTLTISSLQPDDFATYYC(X)n=1−50FGQGTKLTVLG In another aspect, the invention provides methods for the humanization of non-human antibodies by grafting CDRs of non-human donor antibodies onto stable and soluble antibody frameworks. In a particularly preferred embodiment, the CDRs stem from rabbit antibodies and the frameworks are those described above. A general method for grafting CDRs into human acceptor frameworks has been disclosed by Winter in U.S. Pat. No. 5,225,539 and by Queen et al. in WO9007861A1, which are hereby incorporated by reference in their entirety. The general strategy for grafting CDRs from rabbit monoclonal antibodies onto selected frameworks is related to that of Winter et al. and Queen et al., but diverges in certain key respects. In particular, the methods of the invention diverge from the typical Winter and Queen methodology known in the art in that the human antibody frameworks as disclosed herein are particularly suitable as universal acceptors for human or non-human donor antibodies. Thus, unlike the general method of Winter and Queen, the framework sequence used for the humanization methods of the invention is not necessarily the framework sequence which exhibits the greatest sequence similarity to the sequence of the non-human (e.g., rabbit) antibody from which the donor CDRs are derived. In addition, framework residue grafting from the donor sequence to support CDR conformation is not required. At most, antigen binding amino acids located in the framework or other mutations that occurred during somatic hypermutation may be introduced. Particular details of the grafting methods to generate humanized rabbit-derived antibodies with high solubility and stability are described below. Accordingly, the invention provides a method of humanizing a rabbit CDR donor immunobinder which comprises heavy chain CDR1, CDR2 and CDR3 sequences and/or light chain CDR1, CDR2 and CDR3 sequences. The method comprises the steps of:(i) grafting onto the heavy chain at least one, preferably two, more preferably three CDRs of the group consisting of CDR1, CDR2 and CDR3 sequences into a human heavy chain acceptor framework having at least 50%, preferably at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, more preferably at least 95% identity to SEQ ID NO:1; and/or(ii) grafting onto the light chain at least one, preferably two, more preferably three CDRs of the group consisting of CDR1, CDR2 and CDR3 sequences into a human light chain acceptor framework, the human light chain framework having at least 50%, preferably at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, more preferably at least 95% identity to SEQ ID NO:2. In a preferred embodiment, the variable chain acceptor framework comprises (i) a human heavy chain framework comprising a framework amino acid sequence selected from the group consisting of SEQ ID NO:1, SEQ ID NO:4 and SEQ ID NO:6 and (ii) a human light chain framework comprising the framework amino acid sequence of SEQ ID NO:2 or SEQ ID NO:9. In a much preferred embodiment, the method comprises the step of (i) grafting the heavy chain CDR1, CDR2 and CDR3 sequences into the heavy chain and (ii) grafting the light chain CDR1, CDR2 and CDR3 sequences into the light chain of an immunobinder having at least 75%, 80%, 85%, 90%, more preferably at least 95% identity to SEQ ID No. 3, SEQ ID No. 5 or SEQ ID No. 7. More preferably, the immunobinder is or comprises SEQ ID No. 3, SEQ ID No. 5 or SEQ ID No. 7. In another embodiment, in order to improve antigen binding, the method may further comprise the step of substituting acceptor framework residues by donor residues which are involved in antigen binding. In exemplary embodiments of the methods of the invention, the amino acid sequence of the CDR donor antibody is first identified and the sequences aligned using conventional sequence alignment tools (e.g., Needleman-Wunsch algorithm and Blossum matrices). The introduction of gaps and nomenclature of residue positions may be done using a conventional antibody numbering system. For example, the AHo numbering system for immunoglobulin variable domains may be used. The Kabat numbering scheme may also be applied since it is the most widely adopted standard for numbering the residues in an antibody. Kabat numbering may e.g. be assigned using the SUBIM program. This program analyses variable regions of an antibody sequence and numbers the sequence according to the system established by Kabat and co-workers (Deret et al 1995). The definition of framework and CDR regions is generally done following the Kabat definition which is based on sequence variability and is the most commonly used. However, for CDR-H1, the designation is preferably a combination of the definitions of Kabat's, mean contact data generated by analysis of contacts between antibody and antigen of a subset of 3D complex structures (MacCallum et al., 1996) and Chotia's which is based on the location of the structural loop regions (see alsoFIG.1). Conversion tables for the two different numbering systems used to identify amino acid residue positions in antibody heavy and light chain variable regions are provided in A. Honegger, J. Mol. Biol. 309 (2001) 657-670. The Kabat numbering system is described further in Kabat et al. (Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). The AHo numbering system is described further in Honegger, A. and Pluckthun, A. (2001)J. Mol. Biol.309:657-670). The variable domains of the rabbit monoclonal antibodies may e.g. be classified into corresponding human sub-groups using e.g. an EXCEL implementation of sequence analysis algorithms and classification methods based on analysis of the human antibody repertoire (Knappik et al., 2000,J Mol Biol. February 11; 296(1):57-86). CDR conformations may be assigned to the donor antigen binding regions, subsequently residue positions required to maintain the different canonical structures can also be identified. The CDR canonical structures for five of the six antibody hypervariable regions of rabbit antibodies (L1, L2, L3, H1 and H2) are determined using Chothia's (1989) definition. In a preferred embodiment, the CDRs are generated, identified and isolated according to the following method: B-cells, preferably rabbit B-cells, are incubated with (i) target antigens (preferably purified) or (ii) with cells expressing the target antigen on their surface. In case (ii), said cells expressing the target antigen may e.g. be mammalian cells, preferably CHO or HEK293 cells, yeast cells, preferably yeast spheroblasts, or bacterial cells which naturally express the target of choice or are transformed to express the target protein on their surface. Upon expression, the target antigen may be expressed on the cell surface either integrated or attached to the cell membrane. The cells may e.g. be cultivated as isolated strains in cell culture or be isolated from their natural environment, e.g. a tissue, an organ or an organism. Providing the target antigen expressed on the surface of cells, i.e. case (ii), is especially preferred for transmembrane proteins, even more preferably for multi-membrane-spanning proteins, such as GPCRs (G protein-coupled receptors) or ion channels or any other protein of which the native conformation is difficult to maintain upon recombinant expression and purification. Traditional immunization with the recombinant protein is in these cases inadvisable or impossible due to loss of native conformation of integral membrane proteins/complexes during the purification process or due to insufficient amounts of pure protein. In a preferred embodiment of the invention, a mammal, more preferably a rabbit, is immunized with DNA instead of recombinant protein e.g. by a DNA vaccination protocol as disclosed in WO/2004/087216. DNA vaccination induces a rapid immune response against a native antigen. Since no recombinant protein is needed, this technology is on one hand very cost-effective, on the other hand, and more importantly, this method allows for native expression of integral membrane complexes and/or multi-membrane-spanning membrane proteins. The B-cells may be isolated from said immunized mammal, preferably of said rabbit, or alternatively be naive B-cells. In a subsequent step of said method B-cells, preferably memory B-cells, are isolated from lymphatic organs of the immunized animal (such as spleen or lymph nodes), preferably of immunized rabbits. The B-cells are incubated in a mixture with either cells expressing the antigen on their surface or with fluorescence-labeled soluble antigen. B-cells that express target specific antibodies on their surface and consequently bind to the target antigen or to the target antigen expressed on the cell surface are isolated. In a much preferred embodiment, the B-cells and/or the target cells are stained to allow for isolation via flow cytometry based sorting of B-cell/target cell or B-cell/antigen complexes. Flow cytometry normally measures the fluorescence emitted by single cells when they cross a laser beam. However, some researchers have already used cytometers to investigate cell-cell interactions, for example adhesion mediated by cadherins (Panorchan et al, 2006, J. Cell Science, 119, 66-74; Leong et Hibma, 2005, J. Immunol. Methods, 302, 116-124) or integrins (Gawaz et al, 1991, J. Clin. Invest, 88, 1128-1134). Thus, in case (ii), cells expressing the target of choice are stained with an intracellular fluorescent dye (for example calcein). B-cells are stained with fluorescent antibodies binding to cell surface specific markers. Thus, bi-color “events” may be selected, consisting in two cells adhering to each other through B-cell receptor-target interactions (seeFIG.2). As IgG have generally a higher affinity as IgMs, preferably, positive B-cells expressing IgG but not IgM on their surface (which is characteristic for memory B-cells) are selected. For said purpose, multicolor staining is preferably used, where antibodies specific for IgG and IgM are differentially labeled, e.g. with APC and FITC, respectively. In one particular embodiment, a read-out for B-cell sorting can also select for the ability of this interaction to functionally block/activate receptor signaling. For example, B-cells could be incubated with cells that functionally express a GPCR (G protein-coupled receptor). An agonist that signals through a GPCR can be added to the mixture to induce GPCR mediated Ca2+ efflux from the endoplasmic reticulum. In case an antibody presented on a B-cell would functionally block agonist signaling, Ca2+ efflux would consequently also be blocked by this cell-cell interaction. Ca2+ efflux can be quantitatively measured by flow-cytometry. Therefore, only B-cell/target cell conglomerates that either show increase or decrease in Ca2+ efflux would be sorted. The selection step is followed by the cultivation of the B-cells under suitable conditions so that antibodies are secreted into the culture medium. The produced antibodies are monoclonal antibodies. The cultivation may involve the use of a helper cell line, such as a thymoma helper cell line (e.g. EL4-B5, see Zubler et al, 1985, J. Immunol., 134(6): 3662-3668). Preferably, a validation step is performed testing the generated antibodies for specific binding to the target, e.g. for excluding antibodies which are directed against a protein being expressed on the cell surface other than the target protein. For example, CELISA, i.e. a modified enzyme-linked immunosorbent assay (ELISA), where coating step is performed with entire cells, is suitable for said purpose. Said method allows for the evaluation of the selectivity and the ability of antibodies to compete with the ligand. The antibodies generated in the above mentioned step are then analyzed to identify the CDRs of said antibodies. This may involve steps such as purifying the antibodies, elucidating their amino acid sequence and/or nucleic acid sequence. Finally, the CDRs may then be grafted onto acceptor frameworks e.g. by gene synthesis with the oligo extension method, preferably onto the acceptor frameworks described above. In one embodiment, the art recognized process of CDR grafting can be used to transfer donor CDRs into acceptor frameworks. In most cases, all three CDRs from the heavy chain are transplanted from the donor antibody to a single acceptor framework and all three CDRs from the light chain are transplanted to a different acceptor framework. It is expected that it should not always be necessary to transplant all the CDRs, as some CDRs may not be involved in binding to antigen, and CDRs with different sequences (and the same length) can have the same folding (and therefore contacts from antigen to the main chain contacts could be retained despite the different sequences). Indeed single domains (Ward et al, 1989, Nature 341, pp. 544-546) or even single CDRs (R. Taub et al, 1989, J. Biol Chem 264, pp. 259-265) can have antigen binding activities alone. However, whether all or only some of the CDRs are transplanted, the intention of CDR grafting is to transplant the same, or much the same antigen binding site, from animal to human antibodies (see, e.g., U.S. Pat. No. 5,225,539 (Winter)). In another embodiment, the CDRs of the donor antibody can be altered prior to or after their incorporation into the acceptor framework. Alternatively, characterization of the antibodies would be performed only in their final immunobinder format. For this approach, CDR sequences of antibodies expressed on sorted B-cells are retrieved by RT-PCR from either the cultured sorted B-cells or from single sorted B-cells directly. For said purpose, multiplication of B-cells and/or the validation step described above and/or the analyzation step as described above may not be necessary. Combination of two pools of partially overlapping oligonucleotides in which one oligonucleotide pool is coding for the CDRs and a second pool encodes the framework regions of a suitable immunobinder scaffold would allow to generate a humanized immunobinders in a one-step PCR procedure. Highthroughput sequencing, cloning and production would allow to perform clone selection based on the performance of the purified humanized immunobinders, instead of characterizing secreted IgG in the cell culture supernatant. In a preferred embodiment thereof, the immunobinder is a scFv. However, grafting of CDRs may result in an impaired affinity of the generated immunobinder to the antigen due to framework residues which are in contact with the antigen. Such interactions may be a result of somatic hypermutation. Therefore, it may still be required to graft such donor framework amino acids onto the framework of the humanized antibody. Amino acid residues from the non-human immunobinder involved in antigen binding may be identified by examination of rabbit monoclonal antibody variable region sequences and structures. Each residue in the CDR donor framework that differs from the germline may be considered as relevant. If the closest germline cannot be established, the sequence can be compared against the subgroup consensus or the consensus of rabbit sequences with a high percentage of similarity. Rare framework residues are considered as possible result of somatic hypermutation and therefore as playing a role in binding. The antibodies of the invention may be further optimized to show enhanced functional properties, e.g., enhanced solubility and/or stability. In certain embodiments, the antibodies of the invention are optimized according to the “functional consensus” methodology disclosed in PCT Application Serial No. PCT/EP2008/001958, entitled “Sequence Based Engineering and Optimization of Single Chain Antibodies”, filed on Mar. 12, 2008, which is incorporated herein by reference. For example, the immunobinders of the invention can be compared with a database of functionally-selected scFvs is used to identify amino acid residue positions that are either more or less tolerant of variability than the corresponding positions in immunobinder, thereby indicating that such identified residue positions may be suitable for engineering to improve functionality such as stability and/or solubility. Exemplary framework residue positions for substitution and exemplary framework substitutions are described in PCT Application No. PCT/CH2008/000285, entitled “Methods of Modifying Antibodies, and Modified Antibodies with Improved Functional Properties”, filed on Jun. 25, 2008, and PCT Application No. PCT/CH2008/000284, entitled “Sequence Based Engineering and Optimization of Single Chain Antibodies”, filed on Jun. 25, 2008. For example, one or more of the following substitutions may be introduced at an amino acid position (AHo numbering is referenced for each of the amino acid position listed below) in the heavy chain variable region of an immunobinder of the invention:(a) Q or E at amino acid position 1;(b) Q or E at amino acid position 6;(c) T, S or A at amino acid position 7, more preferably T or A, even more preferably T;(d) A, T, P, V or D, more preferably T, P, V or D, at amino acid position 10,(e) L or V, more preferably L, at amino acid position 12,(f) V, R, Q, M or K, more preferably V, R, Q or M at amino acid position 13;(g) R, M, E, Q or K, more preferably R, M, E or Q, even more preferably R or E, at amino acid position 14;(h) L or V, more preferably L, at amino acid position 19;(i) R, T, K or N, more preferably R, T or N, even more preferably N, at amino acid position 20;(j) I, F, L or V, more preferably I, F or L, even more preferably I or L, at amino acid position 21;(k) R or K, more preferably K, at amino acid position 45;(l) T, P, V, A or R, more preferably T, P, V or R, even more preferably R, at amino acid position 47;(m) K, Q, H or E, more preferably K, H or E, even more preferably K, at amino acid position 50;(n) M or I, more preferably I, at amino acid position 55;(o) K or R, more preferably K, at amino acid position 77;(p) A, V, L or I, more preferably A, L or I, even more preferably A, at amino acid position 78;(q) E, R, T or A, more preferably E, T or A, even more preferably E, at amino acid position 82;(r) T, S, I or L, more preferably T, S or L, even more preferably T, at amino acid position 86;(s) D, S, N or G, more preferably D, N or G, even more preferably N, at amino acid position 87;(t) A, V, L or F, more preferably A, V or F, even more preferably V, at amino acid position 89;(u) F, S, H, D or Y, more preferably F, S, H or D, at amino acid position 90;(v) D, Q or E, more preferably D or Q, even more preferably D, at amino acid position 92;(w) G, N, T or S, more preferably G, N or T, even more preferably G, at amino acid position 95;(x) T, A, P, F or S, more preferably T, A, P or F, even more preferably F, at amino acid position 98;(y) R, Q, V, I, M, F, or L, more preferably R, Q, I, M, F or L, even more preferably Y, even more preferably L, at amino acid position 103; and(z) N, S or A, more preferably N or S, even more preferably N, at amino acid position 107. Additionally or alternatively, one or more of the following substitutions can be introduced into the light chain variable region of an immunobinder of the invention:(aa) Q, D, L, E, S, or I, more preferably L, E, S or I, even more preferably L or E, at amino acid position 1;(bb) S, A, Y, I, P or T, more preferably A, Y, I, P or T, even more preferably P or T at amino acid position 2;(cc) Q, V, T or I, more preferably V, T or I, even more preferably V or T, at amino acid position 3;(dd) V, L, I or M, more preferably V or L, at amino acid position 4;(ee) S, E or P, more preferably S or E, even more preferably S, at amino acid position 7;(ff) T or I, more preferably I, at amino acid position 10;(gg) A or V, more preferably A, at amino acid position 11;(hh) S or Y, more preferably Y, at amino acid position 12;(ii) T, S or A, more preferably T or S, even more preferably T, at amino acid position 14;(jj) S or R, more preferably S, at amino acid position 18;(kk) T or R, more preferably R, at amino acid position 20;(ll) R or Q, more preferably Q, at amino acid position 24;(mm) H or Q, more preferably H, at amino acid position 46;(nn) K, R or I, more preferably R or I, even more preferably R, at amino acid position 47;(oo) R, Q, K, E, T, or M, more preferably Q, K, E, T or M, at amino acid position 50;(pp) K, T, S, N, Q or P, more preferably T, S, N, Q or P, at amino acid position 53;(qq) I or M, more preferably M, at amino acid position 56;(rr) H, S, F or Y, more preferably H, S or F, at amino acid position 57;(ss) I, V or T, more preferably V or T, R, even more preferably T, at amino acid position 74;(tt) R, Q or K, more preferably R or Q, even more preferably R, at amino acid position 82;(uu) L or F, more preferably F, at amino acid position 91;(vv) G, D, T or A, more preferably G, D or T, even more preferably T, at amino acid position 92;(xx) S or N, more preferably N, at amino acid position 94;(yy) F, Y or S, more preferably Y or S, even more preferably S, at amino acid position 101; and(zz) D, F, H, E, L, A, T, V, S, G or I, more preferably H, E, L, A, T, V, S, G or I, even more preferably A or V, at amino acid position 103. In other embodiments, the immunobinders of the invention comprise one or more of the stability enhancing mutations described in U.S. Provisional Application Ser. No. 61/075,692, entitled “Solubility Optimization of Immunobinders”, filed on Jun. 25, 2008. In certain preferred embodiments, the immunobinder comprises a solubility enhancing mutation at an amino acid position selected from the group of heavy chain amino acid positions consisting of 12, 103 and 144 (AHo Numbering convention). In one preferred embodiment, the immunobinder comprises one or more substitutions selected from the group consisting of: (a) Serine (S) at heavy chain amino acid position 12; (b) Serine (S) or Threonine (T) at heavy chain amino acid position 103; and (c) Serine (S) or Threonine (T) at heavy chain amino acid position 144. In another embodiment, the immunobinder comprises the following substitutions: (a) Serine (S) at heavy chain amino acid position 12; (b) Serine (S) or Threonine (T) at heavy chain amino acid position 103; and (c) Serine (S) or Threonine (T) at heavy chain amino acid position 144. In certain preferred embodiments, the immunobinder comprises stability enhancing mutations at a framework residue of the light chain acceptor framework in at least one of positions 1, 3, 4, 10, 47, 57, 91 and 103 of the light chain variable region according to the AHo numbering system. In a preferred embodiment, the light chain acceptor framework comprises one or more substitutions selected from the group consisting of (a) glutamic acid (E) at position 1, (b) valine (V) at position 3, (c) leucine (L) at position 4; (d) Serine (S) at position 10; (e) Arginine (R) at position 47; (e) Serine (S) at position 57; (f) phenylalanine (F) at position 91; and (g) Valine (V) at position 103. One can use any of a variety of available methods to produce a humanized antibody comprising a mutation as described above. Accordingly, the present invention provides an immunobinder humanized according to the method described herein. In certain preferred embodiments, the target antigen of said immunobinder is VEGF or TNFα. The polypeptides described in the present invention or generated by a method of the present invention can, for example, be synthesized using techniques known in the art. Alternatively nucleic acid molecules encoding the desired variable regions can be synthesized and the polypeptides produced by recombinant methods. For example, once the sequence of a humanized variable region has been decided upon, that variable region or a polypeptide comprising it can be made by techniques well known in the art of molecular biology. More specifically, recombinant DNA techniques can be used to produce a wide range of polypeptides by transforming a host cell with a nucleic acid sequence (e.g., a DNA sequence that encodes the desired variable region (e.g., a modified heavy or light chain; the variable domains thereof, or other antigen-binding fragments thereof)). In one embodiment, one can prepare an expression vector including a promoter that is operably linked to a DNA sequence that encodes at least VHor VL. If necessary, or desired, one can prepare a second expression vector including a promoter that is operably linked to a DNA sequence that encodes the complementary variable domain (i.e., where the parent expression vector encodes VH, the second expression vector encodes VLand vice versa). A cell line (e.g., an immortalized mammalian cell line) can then be transformed with one or both of the expression vectors and cultured under conditions that permit expression of the chimeric variable domain or chimeric antibody (see, e.g., International Patent Application No. PCT/GB85/00392 to Neuberger et. al.). In one embodiment, variable regions comprising donor CDRs and acceptor FR amino acid sequences can be made and then changes introduced into the nucleic acid molecules to effect the CDR amino acid substitution. Exemplary art recognized methods for making a nucleic acid molecule encoding an amino acid sequence variant of a polypeptide include, but are not limited to, preparation by site-directed (or oligonucleotide-mediated) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared DNA encoding the polypeptide. Site-directed mutagenesis is a preferred method for preparing substitution variants. This technique is well known in the art (see, e.g., Carter et al. Nucleic Acids Res. 13:4431-4443 (1985) and Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488 (1987)). Briefly, in carrying out site-directed mutagenesis of DNA, the parent DNA is altered by first hybridizing an oligonucleotide encoding the desired mutation to a single strand of such parent DNA. After hybridization, a DNA polymerase is used to synthesize an entire second strand, using the hybridized oligonucleotide as a primer, and using the single strand of the parent DNA as a template. Thus, the oligonucleotide encoding the desired mutation is incorporated in the resulting double-stranded DNA. PCR mutagenesis is also suitable for making amino acid sequence variants of polypeptides. See Higuchi, in PCR Protocols, pp. 177-183 (Academic Press, 1990); and Vallette et al., Nuc. Acids Res. 17:723-733 (1989). Briefly, when small amounts of template DNA are used as starting material in a PCR, primers that differ slightly in sequence from the corresponding region in a template DNA can be used to generate relatively large quantities of a specific DNA fragment that differs from the template sequence only at the positions where the primers differ from the template. Another method for preparing variants, cassette mutagenesis, is based on the technique described by Wells et al., Gene 34:315-323 (1985). The starting material is the plasmid (or other vector) comprising the DNA to be mutated. The codon(s) in the parent DNA to be mutated are identified. There must be a unique restriction endonuclease site on each side of the identified mutation site(s). If no such restriction sites exist, they may be generated using the above-described oligonucleotide-mediated mutagenesis method to introduce them at appropriate locations in the DNA encoding the polypeptide. The plasmid DNA is cut at these sites to linearize it. A double-stranded oligonucleotide encoding the sequence of the DNA between the restriction sites but containing the desired mutation(s) is synthesized using standard procedures, wherein the two strands of the oligonucleotide are synthesized separately and then hybridized together using standard techniques. This double-stranded oligonucleotide is referred to as the cassette. This cassette is designed to have 5′ and 3′ ends that are compatible with the ends of the linearized plasmid, such that it can be directly ligated to the plasmid. This plasmid now contains the mutated DNA sequence. A variable region generated by the methods of the invention can be re-modeled and further altered to further increase antigen binding. Thus, the steps described above can be preceded or followed by additional steps, including, e.g. affinity maturation. In addition, empirical binding data can be used for further optimization. It will be understood by one of ordinary skill in the art that the polypeptides of the invention may further be modified such that they vary in amino acid sequence, but not in desired activity. For example, additional nucleotide substitutions leading to amino acid substitutions at “non-essential” amino acid residues may be made to the protein For example, a nonessential amino acid residue in an immunoglobulin polypeptide may be replaced with another amino acid residue from the same side chain family. In another embodiment, a string of amino acids can be replaced with a structurally similar string that differs in order and/or composition of side chain family members, i.e., a conservative substitution, in which an amino acid residue is replaced with an amino acid residue having a similar side chain, may be made. Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Aside from amino acid substitutions, the present invention contemplates other modifications, e.g., to Fc region amino acid sequences in order to generate an Fc region variant with altered effector function. One may, for example, delete one or more amino acid residues of the Fc region in order to reduce or enhance binding to an FcR. In one embodiment, one or more of the Fc region residues can be modified in order to generate such an Fc region variant. Generally, no more than one to about ten Fc region residues will be deleted according to this embodiment of the invention. The Fc region herein comprising one or more amino acid deletions will preferably retain at least about 80%, and preferably at least about 90%, and most preferably at least about 95%, of the starting Fc region or of a native sequence human Fc region. One may also make amino acid insertion Fc region variants, which variants have altered effector function. For example, one may introduce at least one amino acid residue (e.g. one to two amino acid residues and generally no more than ten residues) adjacent to one or more of the Fc region positions identified herein as impacting FcR binding. By “adjacent” is meant within one to two amino acid residues of an Fc region residue identified herein. Such Fc region variants may display enhanced or diminished FcRn binding. Such Fc region variants will generally comprise at least one amino acid modification in the Fc region. In one embodiment amino acid modifications may be combined. For example, the variant Fc region may include two, three, four, five, etc substitutions therein, e.g. of the specific Fc region positions identified herein. In another embodiment, a polypeptide may have altered binding to FcRn and to another Fc receptor. In one embodiment, the polypeptides described in the present invention or generated by a method of the present invention, e.g., humanized Ig variable regions and/or polypeptides comprising humanized Ig variable regions may be produced by recombinant methods. For example, a polynucleotide sequence encoding a polypeptide can be inserted in a suitable expression vector for recombinant expression. Where the polypeptide is an antibody, polynucleotides encoding additional light and heavy chain variable regions, optionally linked to constant regions, may be inserted into the same or different expression vector. An affinity tag sequence (e.g. a His(6) tag) may optionally be attached or included within the polypeptide sequence to facilitate downstream purification. The DNA segments encoding immunoglobulin chains are the operably linked to control sequences in the expression vector(s) that ensure the expression of immunoglobulin polypeptides. Expression control sequences include, but are not limited to, promoters (e.g., naturally-associated or heterologous promoters), signal sequences, enhancer elements, and transcription termination sequences. Preferably, the expression control sequences are eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and the collection and purification of the polypeptide. These expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA. Commonly, expression vectors contain selection markers (e.g., ampicillin-resistance, hygromycin-resistance, tetracycline resistance or neomycin resistance) to permit detection of those cells transformed with the desired DNA sequences (see, e.g., U.S. Pat. No. 4,704,362). E. coliis one prokaryotic host particularly useful for cloning the polynucleotides (e.g., DNA sequences) of the present invention. Other microbial hosts suitable for use include bacilli, such asBacillus subtilus, and other enterobacteriaceae, such asSalmonella, Serratia, and variousPseudomonasspecies. Other microbes, such as yeast, are also useful for expression.SaccharomycesandPichiaare exemplary yeast hosts, with suitable vectors having expression control sequences (e.g., promoters), an origin of replication, termination sequences and the like as desired. Typical promoters include 3-phosphoglycerate kinase and other glycolytic enzymes. Inducible yeast promoters include, among others, promoters from alcohol dehydrogenase, isocytochrome C, and enzymes responsible for methanol, maltose, and galactose utilization. Within the scope of the present invention,E. coliandS. cerevisiaeare preferred host cells. In addition to microorganisms, mammalian tissue culture may also be used to express and produce the polypeptides of the present invention (e.g., polynucleotides encoding immunoglobulins or fragments thereof). See Winnacker, From Genes to Clones, VCH Publishers, N.Y., N.Y. (1987). Eukaryotic cells are actually preferred, because a number of suitable host cell lines capable of secreting heterologous proteins (e.g., intact immunoglobulins) have been developed in the art, and include CHO cell lines, various Cos cell lines, HeLa cells, 293 cells, myeloma cell lines, transformed B-cells, and hybridomas. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, and an enhancer (Queen et al.,Immunol. Rev.89:49 (1986)), and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Preferred expression control sequences are promoters derived from immunoglobulin genes, SV40, adenovirus, bovine papilloma virus, cytomegalovirus and the like. See Co et al.,J. Immunol.148:1149 (1992). The vectors containing the polynucleotide sequences of interest (e.g., the heavy and light chain encoding sequences and expression control sequences) can be transferred into the host cell by well-known methods, which vary depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment, electroporation, lipofection, biolistics or viral-based transfection may be used for other cellular hosts. (See generally Sambrook et al.,Molecular Cloning: A Laboratory Manual(Cold Spring Harbor Press, 2nd ed., 1989). Other methods used to transform mammalian cells include the use of polybrene, protoplast fusion, liposomes, electroporation, and microinjection (see generally, Sambrook et al., supra). For production of transgenic animals, transgenes can be microinjected into fertilized oocytes, or can be incorporated into the genome of embryonic stem cells, and the nuclei of such cells transferred into enucleated oocytes. The subject polypeptide can also be incorporated in transgenes for introduction into the genome of a transgenic animal and subsequent expression, e.g., in the milk of a transgenic animal (see, e.g., Deboer et al. U.S. Pat. No. 5,741,957; Rosen U.S. Pat. No. 5,304,489; and Meade U.S. Pat. No. 5,849,992. Suitable transgenes include coding sequences for light and/or heavy chains in operable linkage with a promoter and enhancer from a mammary gland specific gene, such as casein or beta lactoglobulin. Polypeptides can be expressed using a single vector or two vectors. For example, antibody heavy and light chains may be cloned on separate expression vectors and co-transfected into cells. In one embodiment, signal sequences may be used to facilitate expression of polypeptides of the invention. Once expressed, the polypeptides can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns (e.g., protein A or protein G), column chromatography, HPLC purification, gel electrophoresis and the like (see generally Scopes, Protein Purification (Springer-Verlag, N.Y., (1982)). Either the humanized Ig variable regions or polypeptides comprising them can be expressed by host cells or cell lines in culture. They can also be expressed in cells in vivo. The cell line that is transformed (e.g., transfected) to produce the altered antibody can be an immortalized mammalian cell line, such as those of lymphoid origin (e.g., a myeloma, hybridoma, trioma or quadroma cell line). The cell line can also include normal lymphoid cells, such as B-cells, that have been immortalized by transformation with a virus (e.g., the Epstein-Barr virus). Although typically the cell line used to produce the polypeptide is a mammalian cell line, cell lines from other sources (such as bacteria and yeast) can also be used. In particular,E. coli-derived bacterial strains can be used, especially, e.g., phage display. Some immortalized lymphoid cell lines, such as myeloma cell lines, in their normal state, secrete isolated Ig light or heavy chains. If such a cell line is transformed with a vector that expresses an altered antibody, prepared during the process of the invention, it will not be necessary to carry out the remaining steps of the process, provided that the normally secreted chain is complementary to the variable domain of the Ig chain encoded by the vector prepared earlier. If the immortalized cell line does not secrete or does not secrete a complementary chain, it will be necessary to introduce into the cells a vector that encodes the appropriate complementary chain or fragment thereof. In the case where the immortalized cell line secretes a complementary light or heavy chain, the transformed cell line may be produced for example by transforming a suitable bacterial cell with the vector and then fusing the bacterial cell with the immortalized cell line (e.g., by spheroplast fusion). Alternatively, the DNA may be directly introduced into the immortalized cell line by electroporation. In one embodiment, a humanized Ig variable region as described in the present invention or generated by a method of the present invention can be present in an antigen-binding fragment of any antibody. The fragments can be recombinantly produced and engineered, synthesized, or produced by digesting an antibody with a proteolytic enzyme. For example, the fragment can be a Fab fragment; digestion with papain breaks the antibody at the region, before the inter-chain (i.e., VH-VH) disulphide bond, that joins the two heavy chains. This results in the formation of two identical fragments that contain the light chain and the VHand CH1 domains of the heavy chain. Alternatively, the fragment can be an F(ab′)2fragment. These fragments can be created by digesting an antibody with pepsin, which cleaves the heavy chain after the inter-chain disulfide bond, and results in a fragment that contains both antigen-binding sites. Yet another alternative is to use a “single chain” antibody. Single-chain Fv (scFv) fragments can be constructed in a variety of ways. For example, the C-terminus of VHcan be linked to the N-terminus of VL. Typically, a linker (e.g., (GGGGS)4; SEQ ID NO: 8) is placed between VHand VL. However, the order in which the chains can be linked can be reversed, and tags that facilitate detection or purification (e.g., Myc-, His-, or FLAG-tags) can be included (tags such as these can be appended to any antibody or antibody fragment of the invention; their use is not restricted to scFv). Accordingly, and as noted below, tagged antibodies are within the scope of the present invention. In alternative embodiments, the antibodies described herein, or generated by the methods described herein, can be heavy chain dimers or light chain dimers. Still further, an antibody light or heavy chain, or portions thereof, for example, a single domain antibody (DAb), can be used. In another embodiment, a humanized Ig variable region as described in the present invention or generated by a method of the present invention is present in a single chain antibody (ScFv) or a minibody (see e.g., U.S. Pat. No. 5,837,821 or WO 94/09817A1). Minibodies are dimeric molecules made up of two polypeptide chains each comprising an ScFv molecule (a single polypeptide comprising one or more antigen binding sites, e.g., a VLdomain linked by a flexible linker to a VHdomain fused to a CH3 domain via a connecting peptide). ScFv molecules can be constructed in a VH-linker-VLorientation or VL-linker-VHorientation. The flexible hinge that links the VLand VHdomains that make up the antigen binding site preferably comprises from about 10 to about 50 amino acid residues. An exemplary connecting peptide for this purpose is (Gly4Ser)3 (SEQ ID NO: 27) (Huston et al. (1988).PNAS,85:5879). Other connecting peptides are known in the art. Methods of making single chain antibodies are well known in the art, e.g., Ho et al. (1989),Gene,77:51; Bird et al. (1988),Science242:423; Pantoliano et al. (1991),Biochemistry30:10117; Milenic et al. (1991),Cancer Research,51:6363; Takkinen et al. (1991),Protein Engineering4:837. Minibodies can be made by constructing an ScFv component and connecting peptide-CH3component using methods described in the art (see, e.g., U.S. Pat. No. 5,837,821 or WO 94/09817A1). These components can be isolated from separate plasmids as restriction fragments and then ligated and recloned into an appropriate vector. Appropriate assembly can be verified by restriction digestion and DNA sequence analysis. In one embodiment, a minibody of the invention comprises a connecting peptide. In one embodiment, the connecting peptide comprises a Gly/Ser linker, e.g., GGGSSGGGSGG (SEQ ID NO: 28). In another embodiment, a tetravalent minibody can be constructed. Tetravalent minibodies can be constructed in the same manner as minibodies, except that two ScFv molecules are linked using a flexible linker, e.g., having an amino acid sequence (G4S)4G3AS (SEQ ID NO: 29). In another embodiment, a humanized variable region as described in the present invention or generated by a method of the present invention can be present in a diabody. Diabodies are similar to scFv molecules, but usually have a short (less than 10 and preferably 1-5) amino acid residue linker connecting both variable domains, such that the VLand VHdomains on the same polypeptide chain can not interact. Instead, the VLand VHdomain of one polypeptide chain interact with the VHand VLdomain (respectively) on a second polypeptide chain (WO 02/02781). In another embodiment, a humanized variable region of the invention can be present in an immunoreactive fragment or portion of an antibody (e.g. an scFv molecule, a minibody, a tetravalent minibody, or a diabody) operably linked to an FcR binding portion. In an exemplary embodiment, the FcR binding portion is a complete Fc region. Preferably, the humanization methods described herein result in Ig variable regions in which the affinity for antigen is not substantially changed compared to the donor antibody. In one embodiment, polypeptides comprising the variable domains of the instant invention bind to antigens with a binding affinity greater than (or equal to) an association constant Ka of about 105M−1, 106M−1, 107M−1, 108M−1, 109M−1, 1010M−1, 1011M−1, or 1012M−1, (including affinities intermediate of these values). Affinity, avidity, and/or specificity can be measured in a variety of ways. Generally, and regardless of the precise manner in which affinity is defined or measured, the methods of the invention improve antibody affinity when they generate an antibody that is superior in any aspect of its clinical application to the antibody (or antibodies) from which it was made (for example, the methods of the invention are considered effective or successful when a modified antibody can be administered at a lower dose or less frequently or by a more convenient route of administration than an antibody (or antibodies) from which it was made). More specifically, the affinity between an antibody and an antigen to which it binds can be measured by various assays, including, e.g., an ELISA assay, a BiaCore assay or the KinExA™ 3000 assay (available from Sapidyne Instruments (Boise, ID)). Briefly, sepharose beads are coated with antigen (the antigen used in the methods of the invention can be any antigen of interest (e.g., a cancer antigen; a cell surface protein or secreted protein; an antigen of a pathogen (e.g., a bacterial or viral antigen (e.g., an HIV antigen, an influenza antigen, or a hepatitis antigen)), or an allergen) by covalent attachment. Dilutions of antibody to be tested are prepared and each dilution is added to the designated wells on a plate. A detection antibody (e.g. goat anti-human IgG-HRP conjugate) is then added to each well followed by a chromagenic substrate (e.g. HRP). The plate is then read in ELISA plate reader at 450 nM, and EC50 values are calculated. (It is understood, however, that the methods described here are generally applicable; they are not limited to the production of antibodies that bind any particular antigen or class of antigens.) Those of ordinary skill in the art will recognize that determining affinity is not always as simple as looking at a single figure. Since antibodies have two arms, their apparent affinity is usually much higher than the intrinsic affinity between the variable region and the antigen (this is believed to be due to avidity). Intrinsic affinity can be measured using scFv or Fab fragments. In another aspect, the present invention features bispecific molecules comprising a humanized rabbit antibody, or a fragment thereof, of the invention. An antibody of the invention, or antigen-binding portions thereof, can be derivatized or linked to another functional molecule, e.g., another peptide or protein (e.g., another antibody or ligand for a receptor) to generate a bispecific molecule that binds to at least two different binding sites or target molecules. The antibody of the invention may be derivatized or linked to more than one other functional molecule to generate multispecific molecules that bind to more than two different binding sites and/or target molecules; such multispecific molecules are also intended to be encompassed by the term “bispecific molecule” as used herein. To create a bispecific molecule of the invention, an antibody of the invention can be functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other binding molecules, such as another antibody, antibody fragment, tumor specific or pathogen specific antigens, peptide or binding mimetic, such that a bispecific molecule results. Accordingly, the present invention includes bispecific molecules comprising at least one first binding molecule having specificity for a first target and a second binding molecule having specificity for one or more additional target epitope. In one embodiment, the bispecific molecules of the invention comprise as a binding specificity at least one antibody, or an antibody fragment thereof, including, e.g., an Fab, Fab′, F(ab′)2, Fv, or a single chain Fv. The antibody may also be a light chain or heavy chain dimer, or any minimal fragment thereof such as a Fv or a single chain construct as described in Ladner et al. U.S. Pat. No. 4,946,778, the contents of which is expressly incorporated by reference. While human monoclonal antibodies are preferred, other antibodies which can be employed in the bispecific molecules of the invention are murine, chimeric and humanized monoclonal antibodies. The bispecific molecules of the present invention can be prepared by conjugating the constituent binding specificities using methods known in the art. For example, each binding specificity of the bispecific molecule can be generated separately and then conjugated to one another. When the binding specificities are proteins or peptides, a variety of coupling or cross-linking agents can be used for covalent conjugation. Examples of cross-linking agents include protein A, carbodiimide, N-succinimidyl-S-acetyl-thioacetate (SATA), 5,5′-dithiobis(2-nitrobenzoic acid) (DTNB), o-phenylenedimaleimide (oPDM), N-succinimidyl-3-(2-pyridyldithio)propionate (SPDP), and sulfosuccinimidyl 4-(N-maleimidomethyl) cyclohaxane-1-carboxylate (sulfo-SMCC) (see e.g., Karpovsky et al. (1984)J. Exp. Med.160:1686; Liu, M A et al. (1985)Proc. Natl. Acad. Sci. USA82:8648). Other methods include those described in Paulus (1985) Behring Ins. Mitt. No. 78, 118-132; Brennan et al. (1985)Science229:81-83), and Glennie et al. (1987)J. Immunol.139: 2367-2375). Preferred conjugating agents are SATA and sulfo-SMCC, both available from Pierce Chemical Co. (Rockford, IL). When the binding specificities are antibodies, they can be conjugated via sulfhydryl bonding of the C-terminus hinge regions of the two heavy chains. In a particularly preferred embodiment, the hinge region is modified to contain an odd number of sulfhydryl residues, preferably one, prior to conjugation. Alternatively, both binding specificities can be encoded in the same vector and expressed and assembled in the same host cell. This method is particularly useful where the bispecific molecule is a mAb×mAb, mAb×Fab, Fab×F(ab′)2or ligand×Fab fusion protein. A bispecific molecule of the invention can be a single chain molecule comprising one single chain antibody and a binding determinant, or a single chain bispecific molecule comprising two binding determinants. Bispecific molecules may comprise at least two single chain molecules. Methods for preparing bispecific molecules are described for example in U.S. Pat. Nos. 5,260,203; 5,455,030; 4,881,175; 5,132,405; 5,091,513; 5,476,786; 5,013,653; 5,258,498; and 5,482,858. Binding of the bispecific molecules to their specific targets can be confirmed by, for example, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (MA), flow cytometry based single cell sorting (e.g. FACS analysis), bioassay (e.g., growth inhibition), or Western Blot assay. Each of these assays generally detects the presence of protein-antibody complexes of particular interest by employing a labeled reagent (e.g., an antibody) specific for the complex of interest. For example, the antibody complexes can be detected using e.g., an enzyme-linked antibody or antibody fragment which recognizes and specifically binds to the antibody-VEGF complexes. Alternatively, the complexes can be detected using any of a variety of other immunoassays. For example, the antibody can be radioactively labeled and used in a radioimmunoassay (RIA) (see, for example, Weintraub, B.,Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986, which is incorporated by reference herein). The radioactive isotope can be detected by such means as the use of a γ counter or a scintillation counter or by autoradiography. In another aspect, the present invention features a humanized rabbit antibody, or a fragment thereof, conjugated to a therapeutic moiety, such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radiotoxin. Such conjugates are referred to herein as “immunoconjugates”. Immunoconjugates that include one or more cytotoxins are referred to as “immunotoxins.” A cytotoxin or cytotoxic agent includes any agent that is detrimental to (e.g., kills) cells. Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, l-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents also include, for example, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine). Other preferred examples of therapeutic cytotoxins that can be conjugated to an antibody of the invention include duocarmycins, calicheamicins, maytansines and auristatins, and derivatives thereof. An example of a calicheamicin antibody conjugate is commercially available (MYLOTARG™, a calicheamicin antibody conjugate; Wyeth-Ayerst). Cytoxins can be conjugated to antibodies of the invention using linker technology available in the art. Examples of linker types that have been used to conjugate a cytotoxin to an antibody include, but are not limited to, hydrazones, thioethers, esters, disulfides and peptide-containing linkers. A linker can be chosen that is, for example, susceptible to cleavage by low pH within the lysosomal compartment or susceptible to cleavage by proteases, such as proteases preferentially expressed in tumor tissue such as cathepsins (e.g., cathepsins B, C, D). For further discussion of types of cytotoxins, linkers and methods for conjugating therapeutic agents to antibodies, see also Saito, G. et al. (2003)Adv. Drug Deliv. Rev.55:199-215; Trail, P. A. et al. (2003)Cancer Immunol. Immunother.52:328-337; Payne, G. (2003)Cancer Cell3:207-212; Allen, T. M. (2002)Nat. Rev. Cancer2:750-763; Pastan, I. and Kreitman, R. J. (2002)Curr. Opin. Investig. Drugs3:1089-1091; Senter, P. D. and Springer, C. J. (2001)Adv. Drug Deliv. Rev.53:247-264. Antibodies of the present invention also can be conjugated to a radioactive isotope to generate cytotoxic radiopharmaceuticals, also referred to as radioimmunoconjugates. Examples of radioactive isotopes that can be conjugated to antibodies for use diagnostically or therapeutically include, but are not limited to, iodine131, indium111, yttrium90and lutetium177. Method for preparing radioimmunoconjugates are established in the art. Examples of radioimmunoconjugates are commercially available, including ZEVALIN™, a radioimmunoconjugate (IDEC Pharmaceuticals) and BEXXAR™, a radioimmunoconjugate (Corixa Pharmaceuticals), and similar methods can be used to prepare radioimmunoconjugates using the antibodies of the invention. The antibody conjugates of the invention can be used to modify a given biological response, and the drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, an enzymatically active toxin, or active fragment thereof, such as abrin, ricin A,pseudomonasexotoxin, or diphtheria toxin; a protein such as tumor necrosis factor or interferon-γ; or, biological response modifiers such as, for example, lymphokines, interleukin-1 (“IL-1”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors. Techniques for conjugating such therapeutic moiety to antibodies are well known, see, e.g., Arnon et al., “Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy”, inMonoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al., “Antibodies For Drug Delivery”, inControlled Drug Delivery(2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thorpe, “Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review”, inMonoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); “Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy”, inMonoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., “The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates”,Immunol. Rev.,62:119-58 (1982). In one aspect the invention provides pharmaceutical formulations comprising humanized rabbit antibodies for the treatment disease. The term “pharmaceutical formulation” refers to preparations which are in such form as to permit the biological activity of the antibody or antibody derivative to be unequivocally effective, and which contain no additional components which are toxic to the subjects to which the formulation would be administered. “Pharmaceutically acceptable” excipients (vehicles, additives) are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed. A “stable” formulation is one in which the antibody or antibody derivative therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage. Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones, A. Adv. Drug Delivery Rev. 10: 29-90 (1993), for example. Stability can be measured at a selected temperature for a selected time period. Preferably, the formulation is stable at room temperature (about 30° C.) or at 40° C. for at least 1 month and/or stable at about 2-8° C. for at least 1 year for at least 2 years. Furthermore, the formulation is preferably stable following freezing (to, e.g., −70° C.) and thawing of the formulation. An antibody or antibody derivative “retains its physical stability” in a pharmaceutical formulation if it shows no signs of aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography. An antibody or antibody derivative “retains its chemical stability” in a pharmaceutical formulation, if the chemical stability at a given time is such that the protein is considered to still retain its biological activity as defined below. Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein. Chemical alteration may involve size modification (e.g. clipping) which can be evaluated using size exclusion chromatography, SDS-PAGE and/or matrix-assisted laser desorption ionization/time-of-flight mass spectrometry (MALDI/TOF MS), for example. Other types of chemical alteration include charge alteration (e.g. occurring as a result of deamidation) which can be evaluated by ion-exchange chromatography, for example. An antibody or antibody derivative “retains its biological activity” in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within about 10% (within the errors of the assay) of the biological activity exhibited at the time the pharmaceutical formulation was prepared as determined in an antigen binding assay, for example. Other “biological activity” assays for antibodies are elaborated herein below. By “isotonic” is meant that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 250 to 350 mOsm. Isotonicity can be measured using a vapor pressure or ice-freezing type osmometer, for example. A “polyol” is a substance with multiple hydroxyl groups, and includes sugars (reducing and non-reducing sugars), sugar alcohols and sugar acids. Preferred polyols herein have a molecular weight which is less than about 600 kD (e.g. in the range from about 120 to about 400 kD). A “reducing sugar” is one which contains a hemiacetal group that can reduce metal ions or react covalently with lysine and other amino groups in proteins and a “non-reducing sugar” is one which does not have these properties of a reducing sugar. Examples of reducing sugars are fructose, mannose, maltose, lactose, arabinose, xylose, ribose, rhamnose, galactose and glucose. Non-reducing sugars include sucrose, trehalose, sorbose, melezitose and raffinose. Mannitol, xylitol, erythritol, threitol, sorbitol and glycerol are examples of sugar alcohols. As to sugar acids, these include L-gluconate and metallic salts thereof. Where it is desired that the formulation is freeze-thaw stable, the polyol is preferably one which does not crystallize at freezing temperatures (e.g. −20° C.) such that it destabilizes the antibody in the formulation. Non-reducing sugars such as sucrose and trehalose are the preferred polyols herein, with trehalose being preferred over sucrose, because of the superior solution stability of trehalose. As used herein, “buffer” refers to a buffered solution that resists changes in pH by the action of its acid-base conjugate components. The buffer of this invention has a pH in the range from about 4.5 to about 6.0; preferably from about 4.8 to about 5.5; and most preferably has a pH of about 5.0. Examples of buffers that will control the pH in this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers. Where a freeze-thaw stable formulation is desired, the buffer is preferably not phosphate. In a pharmacological sense, in the context of the present invention, a “therapeutically effective amount” of an antibody or antibody derivative refers to an amount effective in the prevention or treatment of a disorder for the treatment of which the antibody or antibody derivative is effective. A “disease/disorder” is any condition that would benefit from treatment with the antibody or antibody derivative. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question. A “preservative” is a compound which can be included in the formulation to essentially reduce bacterial action therein, thus facilitating the production of a multi-use formulation, for example. Examples of potential preservatives include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride (a mixture of alkylbenzyldimethylammonium chlorides in which the alkyl groups are long-chain compounds), and benzethonium chloride. Other types of preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol. The most preferred preservative herein is benzyl alcohol. The present invention also provides pharmaceutical compositions comprising one or more antibodies or antibody derivative compounds, together with at least one physiologically acceptable carrier or excipient. Pharmaceutical compositions may comprise, for example, one or more of water, buffers (e.g., neutral buffered saline or phosphate buffered saline), ethanol, mineral oil, vegetable oil, dimethylsulfoxide, carbohydrates (e.g., glucose, mannose, sucrose or dextrans), mannitol, proteins, adjuvants, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione and/or preservatives. As noted above, other active ingredients may (but need not) be included in the pharmaceutical compositions provided herein. A carrier is a substance that may be associated with an antibody or antibody derivative prior to administration to a patient, often for the purpose of controlling stability or bioavailability of the compound. Carriers for use within such formulations are generally biocompatible, and may also be biodegradable. Carriers include, for example, monovalent or multivalent molecules such as serum albumin (e.g., human or bovine), egg albumin, peptides, polylysine and polysaccharides such as aminodextran and polyamidoamines. Carriers also include solid support materials such as beads and microparticles comprising, for example, polylactate polyglycolate, poly(lactide-co-glycolide), polyacrylate, latex, starch, cellulose or dextran. A carrier may bear the compounds in a variety of ways, including covalent bonding (either directly or via a linker group), noncovalent interaction or admixture. Pharmaceutical compositions may be formulated for any appropriate manner of administration, including, for example, topical, oral, nasal, rectal or parenteral administration. In certain embodiments, compositions in a form suitable for oral use are preferred. Such forms include, for example, pills, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Within yet other embodiments, compositions provided herein may be formulated as a lyophilizate. The term parenteral as used herein includes subcutaneous, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, intracranial, intrathecal and intraperitoneal injection, as well as any similar injection or infusion technique. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and may contain one or more agents, such as sweetening agents, flavoring agents, coloring agent, and preserving agents in order to provide appealing and palatable preparations. Tablets contain the active ingredient in admixture with physiologically acceptable excipients that are suitable for the manufacture of tablets. Such excipients include, for example, inert diluents (e.g., calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate), granulating and disintegrating agents (e.g., corn starch or alginic acid), binding agents (e.g., starch, gelatin or acacia) and lubricating agents (e.g., magnesium stearate, stearic acid or talc). The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monosterate or glyceryl distearate may be employed. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium (e.g., peanut oil, liquid paraffin or olive oil). Aqueous suspensions contain the antibody or antibody derivative in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia); and dispersing or wetting agents (e.g., naturally-occurring phosphatides such as lecithin, condensation products of an alkylene oxide with fatty acids such as polyoxyethylene stearate, condensation products of ethylene oxide with long chain aliphatic alcohols such as heptadecaethyleneoxycetanol, condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides such as polyethylene sorbitan monooleate). Aqueous suspensions may also comprise one or more preservatives, for example ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, propylene glycol, sorbitol, or sucrose. Such formulations may also comprise one or more demulcents, preservatives, flavoring agents, and/or coloring agents. Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil (e.g.,arachisoil, olive oil, sesame oil, or coconut oil) or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent such as beeswax, hard paraffin, or cetyl alcohol. Sweetening agents, such as those set forth above, and/or flavoring agents may be added to provide palatable oral preparations. Such suspensions may be preserved by the addition of an anti-oxidant such as ascorbic acid. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. Pharmaceutical compositions may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil (e.g., olive oil orarachisoil), a mineral oil (e.g., liquid paraffin), or a mixture thereof. Suitable emulsifying agents include naturally-occurring gums (e.g., gum acacia or gum tragacanth), naturally-occurring phosphatides (e.g., soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol), anhydrides (e.g., sorbitan monooleate), and condensation products of partial esters derived from fatty acids and hexitol with ethylene oxide (e.g., polyoxyethylene sorbitan monoleate). An emulsion may also comprise one or more sweetening and/or flavoring agents. The pharmaceutical composition may be prepared as a sterile injectable aqueous or oleaginous suspension in which the modulator, depending on the vehicle and concentration used, is either suspended or dissolved in the vehicle. Such a composition may be formulated according to the known art using suitable dispersing, wetting agents and/or suspending agents such as those mentioned above. Among the acceptable vehicles and solvents that may be employed are water, 1,3-butanediol, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may be used in the preparation of injectable compositions, and adjuvants such as local anesthetics, preservatives and/or buffering agents can be dissolved in the vehicle. Pharmaceutical compositions may be formulated as sustained release formulations (i.e., a formulation such as a capsule that effects a slow release of modulator following administration). Such formulations may generally be prepared using well known technology and administered by, for example, oral, rectal, or subcutaneous implantation, or by implantation at the desired target site. Carriers for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of modulator release. The amount of an antibody or antibody derivative contained within a sustained release formulation depends upon, for example, the site of implantation, the rate and expected duration of release and the nature of the disease/disorder to be treated or prevented. Antibody or antibody derivatives provided herein are generally administered in an amount that achieves a concentration in a body fluid (e.g., blood, plasma, serum, CSF, synovial fluid, lymph, cellular interstitial fluid, tears or urine) that is sufficient to detectably bind to a target such as e.g. VEGF and prevent or inhibit such target mediated diseases/disorders, e.g. VEGF-mediated diseases/disorders. A dose is considered to be effective if it results in a discernible patient benefit as described herein. Preferred systemic doses range from about 0.1 mg to about 140 mg per kilogram of body weight per day (about 0.5 mg to about 7 g per patient per day), with oral doses generally being about 5-20 fold higher than intravenous doses. The amount of antibody or antibody derivative that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Dosage unit forms will generally contain between from about 1 mg to about 500 mg of an active ingredient. Pharmaceutical compositions may be packaged for treating conditions responsive to an antibody or antibody derivative directed e.g. to VEGF. Packaged pharmaceutical compositions may include a container holding a effective amount of at least one antibody or antibody derivative as described herein and instructions (e.g., labeling) indicating that the contained composition is to be used for treating a disease/disorder responsive to one antibody or antibody derivative following administration in the patient. The antibodies or antibody derivatives of the present invention can also be chemically modified. Preferred modifying groups are polymers, for example an optionally substituted straight or branched chain polyalkene, polyalkenylene, or polyoxyalkylene polymer or a branched or unbranched polysaccharide. Such effector group may increase the half-live of the antibody in vivo. Particular examples of synthetic polymers include optionally substituted straight or branched chain poly(ethyleneglycol) (PEG), poly(propyleneglycol), poly(vinylalcohol) or derivatives thereof. Particular naturally occurring polymers include lactose, amylose, dextran, glycogen or derivatives thereof. The size of the polymer may be varied as desired, but will generally be in an average molecular weight range from 500 Da to 50000 Da. For local application where the antibody is designed to penetrate tissue, a preferred molecular weight of the polymer is around 5000 Da. The polymer molecule can be attached to the antibody, in particular to the C-terminal end of the Fab fragment heavy chain via a covalently linked hinge peptide as described in WO0194585. Regarding the attachment of PEG moieties, reference is made to “Poly(ethyleneglycol) Chemistry, Biotechnological and Biomedical Applications”, 1992, J. Milton Harris (ed), Plenum Press, New York and “Bioconjugation Protein Coupling Techniques for the Biomedical Sciences”, 1998, M. Aslam and A. Dent, Grove Publishers, New York. After preparation of the antibody or antibody derivative of interest as described above, the pharmaceutical formulation comprising it is prepared. The antibody to be formulated has not been subjected to prior lyophilization and the formulation of interest herein is an aqueous formulation. Preferably the antibody or antibody derivative in the formulation is an antibody fragment, such as an scFv. The therapeutically effective amount of antibody present in the formulation is determined by taking into account the desired dose volumes and mode(s) of administration, for example. From about 0.1 mg/ml to about 50 mg/ml, preferably from about 0.5 mg/ml to about 25 mg/ml and most preferably from about 2 mg/ml to about 10 mg/ml is an exemplary antibody concentration in the formulation. An aqueous formulation is prepared comprising the antibody or antibody derivative in a pH-buffered solution. The buffer of this invention has a pH in the range from about 4.5 to about 6.0, preferably from about 4.8 to about 5.5, and most preferably has a pH of about 5.0. Examples of buffers that will control the pH within this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers. The buffer concentration can be from about 1 mM to about 50 mM, preferably from about 5 mM to about 30 mM, depending, for example, on the buffer and the desired isotonicity of the formulation. The preferred buffer is sodium acetate (about 10 mM), pH 5.0. A polyol, which acts as a tonicifier and may stabilize the antibody, is included in the formulation. In preferred embodiments, the formulation does not contain a tonicifying amount of a salt such as sodium chloride, as this may cause the antibody or antibody derivative to precipitate and/or may result in oxidation at low pH. In preferred embodiments, the polyol is a non-reducing sugar, such as sucrose or trehalose. The polyol is added to the formulation in an amount which may vary with respect to the desired isotonicity of the formulation. Preferably the aqueous formulation is isotonic, in which case suitable concentrations of the polyol in the formulation are in the range from about 1% to about 15% w/v, preferably in the range from about 2% to about 10% whv, for example. However, hypertonic or hypotonic formulations may also be suitable. The amount of polyol added may also alter with respect to the molecular weight of the polyol. For example, a lower amount of a monosaccharide (e.g. mannitol) may be added, compared to a disaccharide (such as trehalose). A surfactant is also added to the antibody or antibody derivative formulation. Exemplary surfactants include nonionic surfactants such as polysorbates (e.g. polysorbates 20, 80 etc) or poloxamers (e.g. poloxamer 188). The amount of surfactant added is such that it reduces aggregation of the formulated antibody/antibody derivative and/or minimizes the formation of particulates in the formulation and/or reduces adsorption. For example, the surfactant may be present in the formulation in an amount from about 0.001% to about 0.5%, preferably from about 0.005% to about 0.2% and most preferably from about 0.01% to about 0.1%. In one embodiment, the formulation contains the above-identified agents (i.e. antibody or antibody derivative, buffer, polyol and surfactant) and is essentially free of one or more preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and benzethonium Cl. In another embodiment, a preservative may be included in the formulation, particularly where the formulation is a multidose formulation. The concentration of preservative may be in the range from about 0.1% to about 2%, most preferably from about 0.5% to about 1%. One or more other pharmaceutically acceptable carriers, excipients or stabilizers such as those described in Remington's Pharmaceutical Sciences 21st edition, Osol, A. Ed. (2006) may be included in the formulation provided that they do not adversely affect the desired characteristics of the formulation. Acceptable carriers, excipients or stabilizers are non-toxic to recipients at the dosages and concentrations employed and include; additional buffering agents; co-solvents; antioxidants including ascorbic acid and methionine; chelating agents such as EDTA; metal complexes (e.g. Zn-protein complexes); biodegradable polymers such as polyesters; and/or salt-forming counterions such as sodium. The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to, or following, preparation of the formulation. The formulation is administered to a mammal in need of treatment with the antibody, preferably a human, in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, intrathecal, oral, topical, or inhalation routes. In preferred embodiments, the formulation is administered to the mammal by intravenous administration. For such purposes, the formulation may be injected using a syringe or via an IV line, for example. The appropriate dosage (“therapeutically effective amount”) of the antibody will depend, for example, on the condition to be treated, the severity and course of the condition, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, the type of antibody used, and the discretion of the attending physician. The antibody or antibody derivative is suitably administered to the patent at one time or over a series of treatments and may be administered to the patent at any time from diagnosis onwards. The antibody or antibody derivative may be administered as the sole treatment or in conjunction with other drugs or therapies useful in treating the condition in question. As a general proposition, the therapeutically effective amount of the antibody or antibody derivative administered will be in the range of about 0.1 to about 50 mg/kg of patent body weight whether by one or more administrations, with the typical range of antibody used being about 0.3 to about 20 mg/kg, more preferably about 0.3 to about 15 mg/kg, administered daily, for example. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques. In another embodiment of the invention, an article of manufacture is provided comprising a container which holds the pharmaceutical formulation of the present invention, preferably an aqueous formulation, and optionally provides instructions for its use. Suitable containers include, for example, bottles, vials and syringes. The container may be formed from a variety of materials such as glass or plastic. An exemplary container is a 3-20 cc single use glass vial. Alternatively, for a multidose formulation, the container may be 3-100 cc glass vial. The container holds the formulation and the label on, or associated with, the container may indicate directions for use. The article of manufacture may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. In certain preferred embodiments, the article of manufacture comprises a lyophilized immunobinder as described herein or generated by the methods described herein. EXEMPLIFICATION The present disclosure is further illustrated by the following examples, which should not be construed as further limiting. The contents of all figures and all references, patents and published patent applications cited throughout this application are expressly incorporated herein by reference in their entireties. Materials and Methods In general, the practice of the present invention employs, unless otherwise indicated, conventional techniques of chemistry, molecular biology, recombinant DNA technology, immunology (especially, e.g., antibody technology), and standard techniques of polypeptide preparation. See, e.g., Sambrook, Fritsch and Maniatis, Molecular Cloning: Cold Spring Harbor Laboratory Press (1989); Antibody Engineering Protocols (Methods in Molecular Biology), 510, Paul, S., Humana Pr (1996); Antibody Engineering: A Practical Approach (Practical Approach Series, 169), McCafferty, Ed., Irl Pr (1996); Antibodies: A Laboratory Manual, Harlow et al., C.S.H.L. Press, Pub. (1999); and Current Protocols in Molecular Biology, eds. Ausubel et al., John Wiley & Sons (1992). Methods of grafting CDRs from rabbit and other non-human monoclonal antibodies onto selected human antibody frameworks is described in detail above. Examples of such grafting experiments are set forth below. For purposes of better understanding, grafts denominated “min” are those where CDRs were grafted onto framework 1.4 or a variable domain thereof, whereas grafts named “max” are those where CDRs were grafted onto framework 1.4 or a variable domain thereof and wherein the framework further comprises donor framework residues which interact with the antigen. Example 1: Design of rFW1.4 1.1. Primary Sequence Analysis and Database Searching 1.1.1. Collection of Rabbit Immunoglobulin Sequences Sequences of variable domains of rabbit mature antibodies and germlines were collected from different open source databases (e.g. Kabat database and IMGT) and entered into a customized database as one letter code amino acid sequences. For the entire analysis we used only the amino acid portion corresponding to the V (variable) region. Sequences in the KDB database less than 70% complete or containing multiple undetermined residues in the framework regions were discarded. Sequences with more than 95% identity to any other sequence within the database were also excluded to avoid random noise in the analysis. 1.1.2. Alignments and Numbering of Rabbit Sequences Rabbit antibody sequences were aligned using conventional sequence alignment tools based on the Needleman-Wunsch algorithm and Blossum matrices. The introduction of gaps and nomenclature of residue positions were done following AHo's numbering system for immunoglobulin variable domains (Honegger and Pluckthun, 2001). The Kabat numbering scheme was also applied in parallel since it is the most widely adopted standard for numbering the residues in an antibody. Kabat numbering was assigned using the SUBIM program. This program analyses variable regions of an antibody sequence and numbers the sequence according to the system established by Kabat and co-workers (Deret et al 1995). The definition of framework and CDR regions was done following the Kabat definition which is based on sequence variability and is the most commonly used. Nevertheless, CDR-H1 designation was a compromise between different definitions including, AbM's, kabat's, mean contact data generated by analysis of contacts between antibody and antigen of a subset of 3D complex structures (MacCallum et al., 1996) and Chotia's which is based on the location of the structural loop regions (described above and shown inFIG.1). 1.1.3. Frequency and Conservation of Residue Positions Amino acid sequence diversity was analyzed using a set of 423 rabbit sequences from the kabat database. The residue frequency, f(r), for each position, i, in the mature rabbit sequences was calculated by the number of times that particular residue is observed within the data set divided by the total number of sequences. The degree of conservation for each position, i, was calculated using the Simpson's index, which takes into account the number of different amino acids present, as well as the relative abundance of each residue. D=∑i=1r⁢n⁡(n-1)N⁡(N-1) where: N Total number of amino acids, r is the number of different amino acids present at each position and n is the number of residues of a particular amino acid type. 1.1.4. Lineage Analysis of the Rabbit V Region Phylogeny analysis tools were used to study the rabbit repertoire. Amino acids sequences of the V region were clustered using both cluster and topological algorithms. The distance matrix was calculated for the whole array and used as indication of the germline usage. Consensus sequence of each cluster was calculated and the nearest rabbit germline sequence counterpart identified. Also the overall consensus sequence was derived for the whole set of sequences. 1.1.5. Assignment of Human Subgroup. For each rabbit representative sequence of the different clusters, the most homologous human sub-group was identified using an EXCEL implementation of sequence analysis algorithms and classification methods based on analysis of the human antibody repertoire (Knappik et al., 2000). 1.2. Design of the Human Acceptor Framework With the blueprint of the rabbit repertoire from the sequence analysis described above, residues in the framework generally involved in the positioning of rabbit CDRs were identified. Among the frameworks having high homology relative to the rabbit repertoire and the respective clusters, one having good biophysical properties was selected from a pool of fully human sequences. The selected framework to serve as acceptor framework belongs to variable light chain subgroup kappa 1 and heavy variable chain subgroup III with the ESBATech's ID KI 27, a43 correspondingly. This stable and soluble antibody framework has been identified by screening of a human spleen scFv library using a yeast-based screening method named “Quality Control” system (Auf der Maur et al., 2004) and was designated “FW1.4”. Although the stable and soluble framework sequence FW1.4 exhibits high homology, it was not the most homologous sequence available. The identified residues were incorporated in said acceptor framework to generate rFW1.4. With the information for amino acid sequence diversity, germline usage and the structural features of the rabbit antibodies, we analyzed the FW1.4 for compatibility of residue positions required to preserve the CDR conformation in the new human framework. We examined the variable regions of FW1.4 for compatibility of the following characteristics:i. Residues that are part of the canonical sequences for loop structures.ii. Framework residues located at the VL/VH interface.iii. The platform of residues directly underneath the CDRsiv. Upper and lower core residuesv. Framework residues defining the subtype 1.3. Grafting of Rabbit CDRs Grafts were generated by simply combining the CDR sequences (according above definition) from one antibody with the framework sequence of FW1.4 or the rFW1.4. Residues potentially involved in binding were identified. For each rabbit variable domain sequence, the nearest rabbit germline counterpart was identified. If the closest germline could not be established, the sequence was compared against the subgroup consensus or the consensus of rabbit sequences with a high percentage of similarity. Rare framework residues were considered as possible result of somatic hypermutation and therefore playing a role in binding. Consequently, such residues were grafted onto the acceptor framework. 1.4 Results By analyzing the rabbit antibody repertoire in terms of structure, amino acid sequence diversity and germline usage, 5 residue positions in the light chain of FW1.4 were found which were modified to maintain loop conformation of rabbit CDRs. These positions are highly conserved in rabbit antibodies. The consensus residue for these 5 positions was deduced from the rabbit repertoire and introduced into the human acceptor framework 1.4. With the modification of these conserved positions, said framework became virtually compatible with all six complementarity determining regions (CDRs) of any rabbit CDRs. The master rFW1.4 containing different rabbit CDRs is well expressed and good produced contrary to the wild type single chains. 16 members derived from the combination of this framework and rabbit CDRs were created detailed characterization showed functionality. Example 2: B Cell Screening System A FACS (flow cytometry based single cell sorting)-based screening system has been established at ESBATech in order to select B cells that bind to a target of interest via their B cell receptors (BCR). One target was for example a soluble protein, namely a single-chain antibody (ESBA903) labeled with a fluorescent dye (PE and PerCP). Lymphocyte suspension was prepared from the spleen of rabbits immunized with the recombinant target. Cells were then incubated with PE and PerCP labeled ESBA903 as well as with antibodies specific for IgG (APC-labeled) or IgM (FITC labeled). ESBA903 positive B-cells that express IgG but not IgM on their surface were sorted in 96-well plates (FIG.3; table 2). By means of a thymoma helper cell line (EL4-B5: see Zubler et al, 1985, J. Immunol, 134(6): 3662-3668), selected B cells proliferated, differentiated into plasma cells and secreted antibodies. The affinity of these IgGs for the target was verified by ELISA and Biacore measurements. Kinetic parameters are depicted in table 1 for seven selected clones. These clones, from a pool of ˜200 sorted cells, show binding affinities in the low nanomolar to picomolar range. Finally, mRNA was isolated from 6 clones of interest and CDRs were grafted on the single-chain framework FW1.4. TABLE 1Kinetic values for 7 B cell culture supernatants.B-cell cloneka [Ms−1]kd [s−1]KD[M]SG22.91E+062.95E−041.01E−10SE113.63E+053.81E−041.05E−092E-038.34E+053.53E−044.23E−109E-038.66E+056.47E−047.47E−107D-033.97E+053.04E−047.65E−1012B-021.08E+061.10E−041.01E−10 TABLE 2Sorting statistics.Population#events% parent% totalAll events100.000####100.0Lymphocytes86.58586.686.6Single Lymphocytes 186.01399.386.0Single Lymphocytes 285.52399.485.5Memory B cells?5.4506.45.4Sorted cells160.30.0903-binding cells1602.90.2 Example 3: Detection of the Interaction Between Beads Coated with Anti-TNFalpha Antibody and CHO Cells Expressing Membrane-Bound TNFalpha In order to evaluate whether or not the high pressure in flow-cytometry stream breaks non covalent binding between two cells, the following experiment was performed. CHO cells stably transfected with membrane-bound TNFalpha (B-220 cells) were incubated with beads coated with a PE-labeled anti-TNFalpha antibody. In this set-up the beads mimic memory B cells (they have more or less the same size). As negative controls, non-transfected CHO cells were used, as well as beads coated with an APC labeled unrelated antibody (anti-CD19). After 2 hours incubation at 4° C. with agitation, the cell-bead suspension was analyzed by FACS (using a 130 um nozzle).FIG.4shows that a specific binding between anti-TNFalpha beads and TNFalpha-transfected CHO cells is clearly detectable with FACS. Indeed, in this sample (upper panel) about two thirds of the beads are bound to cells (585 bound against 267 unbound). In contrast, in the control samples (middle and lower panels), almost no bead binds to CHO cells. Further, both bead populations (anti-TNFalpha-PE and anti-CD19-APC) were mixed together with TNFalpha-transfected CHO cells.FIG.5and table 4 shows that about half of the anti-TNFalpha beads bind to CHO cells, whereas the vast majority of the anti-CD19 beads stay unbound. The percentage of beads binding to the cell in each sample is detailed in table 5. Thus, the demonstration is made that the specific selection of single B cells that bind to an integral membrane target protein through their B cell receptor is possible using flow-cytometry. TABLE 3aSorting statistics (see also FIG. 4a)Population# Events% Parent% TotalAll events10.000###100.0P19.69296.996.9P35856.05.9P410.00.0P22672.72.7 TABLE 3bSorting statistics (see also FIG. 4b)Population# Events% Parent% TotalAll events10.000###100.0P19.39994.094.0P330.00.0P460.10.1P25505.65.6 TABLE 3cSorting statistics (see also FIG. 4c)Population# Events% Parent% TotalAll events10.000###100.0P19.00190.090.0P3130.10.1P470.10.1P28118.18.1 TABLE 4Sorting statistics (see also FIG. 5)Population# Events% Parent% TotalAll events10.000###100.0P19.09691.091.0P34014.44.0P420.00.0P28568.68.6 TABLE 5Percentage of beads bound to CHO cells in each sampleCellsmAb on beads% bound beadsSample 1CHO-TNFα (B220)anti-TNFα68.0Sample 2CHO-TNFα (B220)anti-CD190.9Sample 3CHO wtanti-TNFα1.5Sample 4CHO-TNFα (B220)anti-TNFα47.0anti-CD190.4 Example 4: CDR Grafting and Functional Humanization of Anti-TNFα Rabbit Donor Antibodies Four anti-TNFα rabbit antibodies “Rabmabs” (EPI-1, EPI-15, EP-34, EP-35 and EP-42) were selected for CDR grafting. The general experimental scheme for the CDR grafting, humanization, and preliminary characterization of humanized rabbit donor antibodies was done as outlined in the description. Unlike traditional humanization methods which employ the human antibody acceptor framework that shares the greatest sequence homology with the non-human donor antibody, the rabbit CDRs were grafted into a human framework (FW 1.4) that was preselected for desirable functional properties (solubility and stability) using a Quality Control assay (WO0148017). These stable and soluble framework sequence exhibited high homology with the RabMabs. CDR grafts were generated for each of the RabMabs using the methodology described herein. In “Min” grafts, only the rabbit CDRs were transplanted from the VL and VH domains of the rabbit donor antibody to the human acceptor framework FW1.4. “Max” grafts refer to the grafting of the rabbit CDRs to rFW1.4. The scFvs described and characterized herein were produced as follows. The humanized VL sequences were connected to humanized VH sequences via the linker of SEQ ID NO:8 to yield an scFv of the following orientation: NH2-VL-linker-VH-COOH. In many cases DNA sequences encoding for the various scFvs were de novo synthesized at the service provider Entelechon GmbH. The resulting DNA inserts were cloned into the bacterial expression vector pGMP002 via NcoI and HindIII restriction sites introduced at the 5′ and 3′ end of the scFv DNA sequence, respectively. Between the DNA sequence of the VL domain and the VH domain, a BamHI restriction site is located. In some cases the scFv encoding DNA was not de novo synthesized, but the scFv expressing constructs were cloned by domain shuffling. Accordingly, the VL domains were excised and introduced into the new constructs via NcoI and BamHI restriction sites, the VH domains via BamHI and HindIII restriction sites. In other cases, point mutations were introduced into the VH and/or VL domain using state of the art assembling PCR methods. The cloning of GMP002 is described in Example 1 of WO2008006235. The production of the scFvs was done analogue as for ESBA105 as described in Example 1 of WO2008006235. Table 3 depicts a summary of the detailed characterization data for the four rabbit monoclonals (EP6, EP19, EP34, EP35 and EP43) and their CDR grafted variants. Although the CDR grafts exhibited a broad range of activities in BIACore binding assays and L929, TNFα-mediated cytotoxicity assays, 3 of the 4 maximal (“max”) grafts exhibited therapeutically relevant activities. EP43max exhibited the most favorable binding affinity (Kd of 0.25 nM) and an excellent EC50 in the cytotoxicity assay. This data show that FW1.4 (SEQ ID No: 1 and 2) is an exemplary soluble and stable human acceptor framework region for grafting rabbit CDRs. Potency Assay The neutralizing activity of anti-TNFα binders was assessed in a L929 TNFα-mediated cytotoxicity assay. Toxicity of Mouse L929 fibroblast cells treated with Actinomycin was induced with recombinant human TNF (hTNF). 90% of maximal hTNF-induced cytoxicity was determined to be at a TNF concentration of 1000 pg/ml. All L929 cells were cultured in RPMI 1640 with phenolred, with L-Glutamine medium supplemented with fetal calf serum (10% v/v). The neutralizing activity of anti-TNFα binders was assessed in RPMI 1640 without phenolred and 5% fetal calf serum. Different concentrations (0-374 ng/mL) of anti-TNF binders are added to L929 cells in presence of 1000 pg/ml hTNF in order to determine the concentration at which the antagonistic effect reaches half-maximal inhibition (EC50%) The dose response curve was fitted with nonlinear sigmoidal regression with variable slope and the EC50 was calculated. Biacore Binding Analysis of Anti-TNF scFvs For binding affinity measurements, surface Plasmon resonance measurements with BIAcore™-T100 were employed using a NTA sensor chip and His-tagged TNF (produced at ESBATech). The surface of the NTA sensor chip consists of a carboxymethylated dextran matrix pre-immobilized with nitrilotriacetic acid (NTA) for capture of histidine tagged molecules via Ni2+NTA chelation. Human TNFα N-his trimers (5 nM) are captured by the nickel via their N-terminal his-tags and ESBA105 (analyte) is injected at several concentrations ranging from 30 nM to 0.014 nM in 3 fold serial dilution steps. At the regeneration step, the complex formed by nickel, ligand and analyte is washed away. This allows the use of the same regeneration conditions for different samples. The response signal is generated by surface Plasmon resonance (SPR) technology and measured in resonance units (RU). All the measurements are performed at 25° C. Sensorgrams were generated for each anti-TNF scFv sample after in-line reference cell correction followed by buffer sample subtraction. The apparent dissociation rate constant (kd), the apparent association rate constant (ka) and the apparent dissociation equilibrium constant (KD) were calculated using one-to-one Langmuir binding model with BIAcore T100 evaluation Software version 1.1. TABLE 3TNFalpha second generation of bindersDescriptionIDL929*konkoffKDFT-IR TM ° C.RF yield**EP1_min1071ND***————2EP6_min673ND***4.67E+044.94E−031.06E−0750.235EP15_min1073ND***1.57E+054.10E−022.62E−07—41.5EP19_min616ND***—————EP34_min643ND***—————EP35_min1075ND***————1EP42_min1076ND***1.42E+058.35E−035.87E−08—3EP43_min705ND***5.38E+032.98E−025.54E−0670.230.0EP1_max1072ND***1.11E+046.30E−045.69E−08—44EP6_max6741.12.84E+051.45E−045.12E−1048.112EP15_max10740.391.53E+062.26E−031.48E−0968.657.8EP19_max10070.62.25E+046.54E−052.91E−0953.552EP34_max79110.55.86E+051.68E−052.86E−1172.44.05EP35_max10895.207.72E+051.50E−041.94E−10—0.66EP42_max1077ND***1.21E+054.19E−043.46E−09—47.6EP43_max6766.41.78E+054.48E−052.51E−1074.321.73EP34min_C-His7900.2EP19max_C-His7891.9*L929 [EC50-E105/EC50-X], compared in mass units [ng/ml] relative to the performance of ESBA105(WO06/131013)**(mg/L refolding solution);***Not Determined Example 5: CDR Grafting and Functional Humanization of Anti-VEGF Rabbit Donor Antibodies Eight anti-VEGF Rabmabs (375, 435, 509, 511, 534, 567, 578 and 610) were selected for CDR grafting. Unlike traditional humanization methods which employ the human antibody acceptor framework that shares the greatest sequence homology with the non-human donor antibody, the rabbit CDRs were grafted into a human acceptor framework FW1.4 (SEQ ID No: 1 and 2) that was preselected for desirable functional properties (solubility and stability) using a Quality Control assay (WO0148017). A number of CDR grafts were generated for each of the RabMabs (rabbit antibodies) using the methodology described herein (see Example 4). “Min” grafts comprised a minimal graft wherein only the rabbit CDRs were transplanted from the VL and VH domains of the rabbit donor antibody to the human acceptor framework FW1.4 (SEQ ID No: 1). “Max” grafts comprised not only the rabbit CDRs for the VL and VH, but also some additional framework residues from the rabbit donor that were predicted to be important for antigen binding. In the case of 578max, the heavy chain variable domain framework region of FW1.4 has additional amino acid alterations at Kabat positions 23H, 49H, 73H, 78H, and 94H. Table 4 shows a summary of the detailed characterization data for the “Min” and “Max” CDR grafted variants. Their potency as VEGF inhibitors, which is measured using VEGFR competition ELISA and/or HUVEC assay are described. This data shows that FW1.4 (SEQ ID No: 1 and 2) is an exemplary soluble and stable human acceptor framework region for grafting rabbit CDRs. Biacore Binding Analysis of Anti-VEGF scFvs The Biacore-binding ability of scFvs was tested and the binding affinity was measured using the exemplary surface plasmon resonance method with BIACORE™-T100, a plasmon resonance method (GE Healthcare). The VEGF proteins, tested for binding by these scFv candidates, in this example and later examples include purifiedEscherichia coli-expressed recombinant human VEGF165(PeproTech EC Ltd.), recombinant human VEGF121(PeproTech EC Ltd.), recombinant human VEGF110(ESBATech AG), recombinant murine VEGF164(PeproTech EC Ltd.), recombinant rat VEGF164(Biovision), recombinant rabbit VEGF110(ESBATech AG), and recombinant human PLGF (PeproTech EC Ltd.). For the surface plasmon resonance experiment, carboxymethylated dextran biosensor chips (CM4, GE Healthcare) were activated with N-ethyl-N′-(3-dimethylaminopropyl) carbodiimide hydrochloride and N-hydroxysuccinimide according to the supplier's instructions. Each of the 6 different VEGF forms, as exemplified above, was coupled to 1 of the 4 different flow cells on a CM4 sensor chip using a standard amine-coupling procedure. The range of responses obtained with these immobilized VEGF molecules after coupling and blocking were ˜250-500 response units (RU) for hVEGF165, ˜200 RU for hVEGF110, hVEGF121, murine VEGF164, rat VEGF164and rabbit VEGF110and ˜400 RU for PLGF. The 4th flow cell of each chip was treated similarly except no proteins were immobilized prior to blocking, and the flow cell was used as in-line reference. Various concentrations of anti-VEGF scFvs (e.g., 90 nM, 30 nM, 10 nM, 3.33 nM, 1.11 nM, 0.37 nM, 0.12 nM and 0.04 nM) in HBS-EP buffer (0.01 M HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% surfactant P20) were injected into the flow cells at a flow rate of 30 μl/min for 5 min. Dissociation of the anti-VEGF scFv from the VEGF on the CM4 chip was allowed to proceed for 10 min at 25° C. Sensorgrams were generated for each anti-VEGF scFv sample after in-line reference cell correction followed by buffer sample subtraction. The apparent dissociation rate constant (kd), the apparent association rate constant (ka) and the apparent dissociation equilibrium constant (KD) were calculated using one-to-one Langmuir binding model with BIACORE™-T100, a plasmon resonance method (GE Healthcare) evaluation Software version 1.1. HUVEC Assay of VEGF Inhibition The HUVEC assay is a method to measure the potency of the disclosed anti-VEGF scFv candidates as VEGF inhibitors. Human umbilical vein endothelial cells (HUVECs) (PromoCell GmbH), pooled from several donors, were used at passage 2 to passage 14. Cells were seeded at 1000 cells/well in 50 μl complete endothelial cell growth medium (ECGM) (PromoCell GmbH), that contained 0.4% ECGS/H, 2% Fetal Calf Serum, 0.1 ng/ml Epidermal Growth Factor, 1 μg/ml Hydrocortisone, 1 ng/ml basic Fibroblast Factor and 1% penicillin/streptomycin (Gibco). 7 to 8 h later, 50 μl starving medium (ECGM without supplements containing 0.5% heat inactivated FCS and 1% penicillin/streptomycin) was added to the cells and the cells were starved for 15 to 16 hours. 3 fold Serial dilutions of anti-VEGF scFvs (0.023-150 nM) and one of the following—recombinant human VEGF165(0.08 nM), recombinant mouse VEGF164(0.08 nM), or recombinant rat VEGF164(0.3 nM) —were prepared in starving medium and preincubated for 30-60 min at room temperature. The different concentrations of VEGFs were used to compensate for their different relative biological activities. Concentrations that stimulate submaximal VEGF induced proliferation (EC90) were used. 100 μl of the mixtures were added to the 96-well tissue-culture plates containing the HUVEC suspension and incubated for 4 days in a 37° C./5% CO2humified incubator. Proliferation of HUVECs was assessed by measuring absorbance at 450 nm (620 nm used as reference wavelength) after addition of 20 μl/well WST-1 cell proliferation reagent (Roche) using a Sunrise microplate reader (Tecan). Data were analyzed using a 4-parameter logistic curve-fit, and the concentration of anti-VEGF scFvs required to inhibit HUVEC proliferation by 50% (EC50) was derived from inhibition curves. TABLE 4Rel. activityRel. activityhVEGR2 comp.hVEGR1 comp.ELISAELISABiacore MeasurementsProtein(EC50Luc[nM]/(EC50Luc[nM]/hVEGF165IDNr.EC50test[nM])EC50test[nM])ka (1/Ms)kd (1/s)KD (M)375-min8570.3ND9.27E+055.01E−035.41E−09375-max8730.6ND2.44E+066.55E−032.68E−09375-max C-His8770.4ND2.93E+058.75E−042.98E−09509-min8541.02.96.23E+051.14E−031.82E−09509-max8554.1132.26E+062.72E−031.21E−09509-maxII8560.60.098.38E+052.82E−033.37E−09511-min8014.90.75.05E+051.28E−032.53E−09511-max8028.786.59E+054.40E−056.67E−11534-min C-His8070.1ND2.71E+059.21E−033.41E−08534-max7931.1ND1.88E+061.73E−029.21E−09567-min8849.7572.01E+064.61E−042.30E−10567-max8744.115.7/54.51.20E+062.26E−041.88E−10578-min8204.14.81.14E+061.03E−029.01E−09578-max8219.635.5/51.67.00E+053.07E−044.39E−10610-min8820.1ND2.51E+052.65E−031.06E−08610-max8830.4ND5.09E+056.01E−041.18E−09435-min944NDNDNDNDND435-max9457.6ND1.67E+057.55E−044.53E−09 EQUIVALENTS Numerous modifications and alternative embodiments of the present invention will be apparent to those skilled in the art in view of the foregoing description. Accordingly, this description is to be construed as illustrative only and is for the purpose of teaching those skilled in the art the best mode for carrying out the present invention. Details of the structure may vary substantially without departing from the spirit of the invention, and exclusive use of all modifications that come within the scope of the appended claims is reserved. It is intended that the present invention be limited only to the extent required by the appended claims and the applicable rules of law. All literature and similar material cited in this application, including, patents, patent applications, articles, books, treatises, dissertations, web pages, figures and/or appendices, regardless of the format of such literature and similar materials, are expressly incorporated by reference in their entirety. In the event that one or more of the incorporated literature and similar materials differs from or contradicts this application, including defined terms, term usage, described techniques, or the like, this application controls.
140,130
11858982
EXAMPLES Example 1—Effect of Antibodies on Cell Proliferation Materials The cell lines MCF7, MCF-7/TAMR7, A2780, A2780cis, A2780ADR, HCT116, Caco-2, SW480, COR-L23, COR-L23.5010, MIA-PaCa-2, PANC-1 and BxPC-3 were obtained from Public Health England Culture Collections. The HCC1806 cell line was obtained from the ATCC. MCF7 is a human breast adenocarcinoma cell line positive for the oestrogen receptor and progesterone receptor; MCF-7/TAMR7 is a tamoxifen-resistant derivative of MCF7; HCC1806 is a triple-negative human breast cancer cell line; A2780 is a human ovarian carcinoma cell line; A2780cis is a cisplatin-resistant human ovarian carcinoma cell line (derived from A2780); A2780ADR is an adriamycin-resistant human ovarian carcinoma cell line (derived from A2780); MIA PaCa-2 is a human pancreatic carcinoma cell line; BxPC-3 is a human pancreatic adenocarcinoma cell line; PANC-1 is a human pancreatic epithelioid carcinoma cell line; Caco-2 is a human colorectal adenocarcinoma cell line; HCT116 is a human colorectal carcinoma cell line; SW480 is a human colorectal adenocarcinoma cell line; COR-L23 is a human lung large cell carcinoma cell line; COR-L23.5010 is an adriamycin-resistant derivative of COR-L23. The L1M2H4 and L2M2H2 anti-Anx-A1 antibodies are disclosed in WO 2018/146230 with sequences as described herein. The L1M2H4 antibody has a light chain with the amino acid sequence set forth in SEQ ID NO: 13 and a heavy chain with the amino acid sequence set forth in SEQ ID NO: 14; the L2M2H2 antibody has a light chain with the amino acid sequence set forth in SEQ ID NO: 15 and a heavy chain with the amino acid sequence set forth in SEQ ID NO: 16. The anti-Anx-A1 antibody ab65844 was obtained from Abcam (UK). The antibody is a polyclonal rabbit antibody that binds human Anx-A1 amino acids 3-24 (SEQ ID NO: 30). This epitope sequence forms part of the N-terminal region of Anx-A1 which, in the absence of Ca2+binds within a pocket in the Anx-A1 core, discussed above. Methods Cell Culture In the initial proliferation assays cells were cultured in the following media: DMEM+ Glutamax, 10% FBS+pen/strep (MCF7, MIA PaCa-2, HCC1806), RPMI 1640+2 mM L-glu, 10% FBS+pen/strep (BxPc-3, A2780). A2780cis and A2780ADR were cultured in same growth media as A2780 but included the respective drug at various stages of culture to maintain drug resistance (i.e. cisplatin for A2780cis and adriamycin for A2780ADR). In the further proliferation assays, cells were cultured in the following media under the following conditions:MCF7, HCC1806, MIA PaCa-2 and PANC-1 in DMEM containing 10% FBS, 1% pen/strep and 1% L-glutamine;MCF7/TAMR7 in phenol red-free DMEM/F12 containing 1% FBS, 1% pen/strep, 1% L-glutamine and 1% insulin;A2780, COR-L23, COR-L23.5010 and BxPC-3 in RPMI containing 10% FBS, 1% pen/strep and 1% L-glutamine;HCT116 in McCoy's 5A containing 10% FBS, 1% pen/strep and 1% L-glutamine;SW480 in L-15 containing 10% FBS, 1% pen/strep and 1% L-glutamine;Caco-2 in MEM containing 10% FBS, 1% pen/strep, 1% L-glutamine and 1% non-essential amino acid solution. Additionally, COR-L23.5010 was cultured in the presence of adriamycin, to maintain drug resistance. All cell lines were cultured at 37° C. in an atmosphere containing 5% CO2. Cell Proliferation Assay Cell proliferation was measured using the MTT colorimetric assay to measure cell metabolic activity. In the assay, NADPH-dependent cellular oxidoreductase enzymes reduce the yellow tetrazolium dye, MTT, to an insoluble purple formazan product, quantified by measuring absorbance at 500-600 nm using a spectrophotometer. The quantity of the formazan is proportional to the level of cell proliferation with rapidly dividing cells reducing a higher level of MTT. Assays were performed in triplicate. Cells were seeded in a final volume of 100 μL. In the initial proliferation assays cells were seeded at the following densities: 1×105/mL (MCF7, MIA PaCa-2 and n=2 of A2780cis), 2×105/mL (A2780, A2780ADR and n=1 of A2780cis), 5×104/mL (HCC1806) and 2.5×104/mL (BxPC-3). In the further proliferation assays cells were seeded at the following densities: MCF7, HCC1806, A2780, A2780ADR, A2780cis, COR-L23 and HCT116 at 5×103cells per well; MCF7/TAMR7, COR-L23.5010, SW480, Caco-2, MIA PaCa-2, BxPC-3 and PANC-1 at 1×104per well. Cells were then cultured for 24 hr prior to assay, then cell proliferation was measured. Cell proliferation was measured following one of the following protocols:(a) 48 hr culture in the absence of antibody (as control), with 1 μM antibody (either L1M2H4 or L2M2H2) or 10 μM antibody (either L1M2H4 or L2M2H2). MTT assays were performed on the various cancer cell lines on up to three separate occasions (initial proliferation assays); or(b) 72 hr culture in the absence of antibody, or in the presence of antibody at a concentration of 2.5, 5, 7.5 or 10 μM. The antibodies used in this protocol were L1M2H4 (also referred to herein as MDX-124), the commercially-available anti-Anx-A1 antibody ab65844, and a non-Anx-A1-specific IgG as isotype control (Thermo Fisher Scientific, USA, catalogue number 31154) (further proliferation assays). Cell number of the control culture was defined as the baseline count for the proliferation assay. Cell counts for cultures in which an antibody was present were normalised to the baseline, and presented as a percentage of the baseline value (referred to as “viability”). Statistical analysis of cell proliferation assay results was performed using the Mann-Whitney U test. ELISA ELISA was performed by The Antibody Company (UK) using standard ELISA techniques. ELISA plates were coated with 25 μg/ml full-length Anx-A1 or Anx-A1 N-terminal peptide (corresponding to Anx-A1 amino acids 2-26, SEQ ID NO: 31) and coating buffer (45 mM Na2CO3, pH 9.6 supplemented with 1 mM CaCl2) for 17 hr at 4° C. Plates were then blocked for 1.5 hr at room temperature with blocking buffer (1 mM CaCl2, 10 mM HEPES, 2% w/v BSA). Primary antibody (ab65844) was then applied to the plates. The antibody was applied in duplicate in four-fold dilutions made across the plate, starting at a concentration of 1 μg/ml and ending at a concentration of 2.38×107μg/ml. The antibody was diluted in wash buffer (10 mM HEPES, 150 mM NaCl, 0.05% (v/v) TWEEN-20 and 1 mM CaCl2) supplemented with 0.1 BSA. The primary antibody was applied to the plate for 1 hr at room temperature, and the plate then washed with wash buffer. The detection antibody was then applied. For detection a horseradish peroxidase (HRP)-conjugated goat anti-rabbit antibody was used (Merck KGaA, Germany; catalogue number AP156P) at a dilution of 1:3000. This was applied to the ELISA plate for 1 hour at room temperature. The ELISA plate was then washed again with wash buffer. The colorimetric substrate OPD (o-phenylenediamine dihydrochloride, Sigma-Aldrich P4664) was then applied to the plate. OPD solution was made up according to the manufacturer's instructions to yield a 0.4 mg/ml OPD solution in phosphate-citrate buffer, pH 5. 40 μl of 30% H2O2was added per 100 ml OPD solution immediately prior to use. 100 μl of the resultant OPD solution was then added to each well of the plate. The plate was incubated for 20 mins in the dark at room temperature, after which 50 μl of 3 M H2SO4was added to stop the reaction. Immediately after addition of H2SO4the absorbances of the plate were read at 492 nm. Results The commercially-acquired anti-Anx-A1 antibody_ab65844 was tested by ELISA to confirm binding to its reported epitope. The assay demonstrated that antibody ab65844 binds both to full-length Anx-A1 and an N-terminal Anx-A1 peptide (data not shown), indicating that the reported epitope of amino acids 3-24 of human Anx-A1 (SEQ ID NO: 30) is correct. Initial Proliferation Assays The first proliferation assays carried out measured proliferation over 48 hours, and compared the effect of the two antibodies L1M2H4 and L2M2H2 on the cell lines of interest, relative to incubation without any antibody (i.e. using protocol (a) as described above). The results of these proliferation assays with the breast cancer cell lines are shown inFIGS.1&2. As shown, the L1M2H4 antibody had a statistically significant effect (p<0.001) at 10 μM, reducing proliferation of the MCF7 cells although this was only n=1. In the HCC1806 cell line, the L2M2H2 antibody also showed a statistically significant decrease in proliferation (p<0.05) at 10 μM (n=2). The results of the proliferation assay with the ovarian cancer cell line, A2780, are shown inFIG.3. As shown, a statistically significant reduction in proliferation was seen following incubation with the L1M2H4 antibody at 10 μM, p<0.01) (n=2). In the cisplatin resistant ovarian cancer cell line, A2780cis (FIG.4), a statistically significant reduction in proliferation was seen following incubation with the L1M2H4 antibody at 1 μM (p<0.001) and 10 μM (p<0.01) (n=2) with a statistically significant decrease in proliferation observed with the L2M2H2 antibody at 10 μM (p<0.05) (n=3). The results of the proliferation assays with the adriamycin resistant ovarian cancer cell line, A2780ADR are shown inFIG.5. The L2M2H2 antibody had a significant effect on proliferation of these cells. The results of the proliferation assays with the pancreatic cancer cell lines are shown inFIGS.6&7. Further Proliferation Assays The proliferation assays were repeated, measuring proliferation over 72 hours. These assays compared the effect of one antibody of the invention (L1M2H4, also known as MDX-124) with the effect on proliferation of a non-specific IgG control and, where indicated, the commercially-available anti-Anx-A1 antibody ab65844. Comparison to proliferation in the absence of any antibody was also performed and used as the baseline. All experiments were performed in triplicate (for MDX-124 and the IgG control, and where ab65844 was also tested, experiments using this antibody were performed in duplicate). MDX-124 was found to have a significant effect on proliferation of the HCC1806 breast cancer cell line, causing an almost two-thirds reduction (63%) in viability relative to the baseline (FIG.8). The non-specific IgG control had no effect on viability. Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in HCC1806 cell viability at all tested concentrations, with a P value of <0.001 (at an antibody concentration of 2.5 μM) or <0.0001 (at all other antibody concentrations). Notably, the polyclonal anti-Anx-A1 antibody ab65844 actually caused an increase in cell viability (and thus proliferation). Indeed, at all antibody concentrations MDX-124 was found to cause a statistically significant reduction in HCC1806 cell viability relative to ab65844, with a P value of <0.01 (at an antibody concentration of 2.5 μM) or <0.001 (at all other antibody concentrations). This result demonstrates that MDX-124 inhibits the proliferation of HCC1806 cells (causing a significant reduction in viability). This effect is not, however, seen for all anti-Anx-A1 antibodies, in that ab65844 has the opposite effect on cell viability. MDX-124 was also found to have significant effects on proliferation of the breast cancer cell line MCF7, and its tamoxifen-resistant derivative (FIGS.9&10, respectively). In both instances, the non-specific IgG control reduces proliferation by up to 26%. At its maximum concentration MDX-124 causes a significant 76% reduction in viability of MCF7 cells, and also a significant (though lower) 47% reduction in the viability of tamoxifen-resistant MCF7 cells. Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in MCF7 cell viability at all tested concentrations with a P value of <0.0001. MDX-124 was also found to cause statistically significant reductions in MCD7/TAMR7 cell viability, relative to the non-specific IgG control, at concentrations of 5 and 7.5 μM (P<0.01) and 10 μM (P<0.05). These results show that MDX-124 is highly effective in inhibiting the proliferation of breast cancer cells, of both triple-negative and hormone receptor positive cell lines. The antibody is also effective against drug-resistant breast cancer. This effect is specific to MDX-124, and is not seen in all anti-Anx-A1 antibodies. Similarly, MDX-124 was found to have a significant effect on proliferation of the ovarian cancer cell line A2780 (FIG.11). While the non-specific IgG reduces proliferation by up to 30% (at the maximum concentration), MDX-124 has more than double the effect on proliferation (causing a 61% reduction in proliferation at the maximum concentration). Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in A2780 cell viability at concentrations of 5 and 7.5 μM (P<0.05) and 10 μM (P<0.01). Again, this is not seen for the polyclonal anti-Anx-A1 antibody ab65844, which caused no significant impact on proliferation. For this antibody, slightly increased proliferation was seen at low concentrations of this antibody (up to 5 μM), while at the maximum concentration a modest reduction in proliferation of 5% was seen. Indeed, at antibody concentrations of 5, 7.5 and 10 μM MDX-124 was found to cause a statistically significant reduction in A2780 cell viability relative to ab65844, with a P value of <0.001. MDX-124 was also found to have a substantial impact on proliferation of colorectal cancer cells (FIGS.12-14). The results on the proliferation of the HCT116 cell line are shown inFIG.12. MDX-124 more than halves proliferation of these cells (reducing proliferation by up to 54% at the maximum concentration). The non-specific IgG control has no real impact on proliferation, and the polyclonal anti-Anx-A1 antibody ab65844 had varying impacts at the different concentrations. Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in HCT116 cell viability at all tested concentrations with a P value of <0.001 (at antibody concentrations of 2.5 and 7.5 μM) or <0.0001 (at antibody concentrations of 5 and 10 μM). While the data for ab65844 is slightly inconsistent, there is a clear general trend of the antibody again driving increased proliferation of the cancer cells, and relative to ab65844, MDX-124 was found to cause statistically significant reductions in HCT116 cell viability at antibody concentrations of 2.5 μM (P<0.01) and 5 and 10 μM (both P<0.001). No data point suggests that ab65844 causes a reduction in proliferation. The results using the cell lines Caco-2 (FIG.13) and SW480 (FIG.14) tell a similar story, in that MDX-124 causes a significant reduction in proliferation, while the non-specific IgG control has at most minimal impact. Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in Caco-2 cell viability at all tested concentrations, with a P value of <0.05 (at antibody concentrations of 2.5 and 5 μM) or <0.001 (at antibody concentrations of 7.5 and 10 μM). Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in SW480 cell viability at all tested concentrations, with a P value of <0.01 (at an antibody concentration of 2.5 μM) or <0.0001 (at all other tested antibody concentrations). These results demonstrate that MDX-124 is highly effective in inhibiting the proliferation of colorectal cancer cells. Again, this effect is specific to MDX-124, not being seen in all anti-Anx-A1 antibodies. The impact of MDX-124 on pancreatic cancer cell lines is shown inFIGS.15-17. The impact of MDX-124 on the cell line BxPC-3 is shown inFIG.15. Again, MDX-124 has a significant impact on cell proliferation, reducing proliferation by half at the maximum concentration. The non-specific IgG control had minimal impact on proliferation, while again the polyclonal anti-Anx-A1 antibody ab65844 drove a significant increase in proliferation. Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in BxPC-3 cell viability at all tested concentrations, with a P value of <0.001 (at an antibody concentration of 2.5 μM) or <0.0001 (at all other tested antibody concentrations). Relative to ab65844, MDX-124 was found to cause a statistically significant reduction in BxPC-3 cell viability with a P value of <0.001 at all tested antibody concentrations. The impact of MDX-124 on the cell lines MIA PaCa-2 and PANC-1 is shown inFIGS.16and17, respectively. In both instances, MDX-124 causes a significant reduction in proliferation of almost half, while the non-specific IgG causes much lesser reductions in viability. Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in MIA PaCa-2 cell viability at antibody concentrations of 5 and 10 μM, with a P value of <0.05. Relative to the non-specific IgG control, MDX-124 was found to cause a statistically significant reduction in PANC-1 cell viability at all antibody concentrations, with a P value of <0.05 (at an antibody concentration of 2.5 μM) or <0.001 (at all other tested antibody concentrations). The impact of MDX-124 on the lung cancer cell lines COR-L23 and COR-L23.5010 is shown inFIGS.18&19, respectively. These results show that the MDX-124 antibody has a modest but negative effect on proliferation, whereas the non-specific IgG control and the polyclonal anti-Anx-A1 antibody ab65844 showed very little effect on proliferation. Similar results were obtained with other lung cancer cell lines (data not shown). Conclusions Exposure to MDX-124 causes a significant reduction in proliferation of cell lines from breast cancer (including triple negative, hormone receptor positive and drug-resistant cell lines), colorectal cancer, ovarian cancer, lung cancer and pancreatic cancer. The impact of MDX-124 on cancer cell proliferation is antibody-specific, i.e. not all antibodies against the same target (Anx-A1) have the same impact. This is demonstrated by the fact that the ab65844 failed to significantly reduce proliferation of any of the cell lines it was tested against, and indeed increased proliferation in the majority of cases. The non-specific IgG control also did not cause the significant reduction in proliferation seen using MDX-124. Example 2—Epitope Determination HDX analysis was performed at the Natural and Medical Sciences Institute (NMI), University of Tübingen, Germany, using the L1M2H4 antibody. Sample Preparation and Analysis Antibody-Antigen Complex Formation and Hydrogen-Deuterium Exchange Five aliquots of antibody-antigen samples and five aliquots of antigen without antibody were prepared as follows: 0.8 μL Anx-A1 (41 μM), 1.8 μL antibody (38.7 μM) or HEPES buffer (10 mM HEPES, 1 mM CaCl2, 150 mM NaCl pH 7.4) respectively, 1 μL HEPES buffer and 0.5 μL CaCl2(8 mM) were mixed and incubated for 10 minutes at 20° C. 8.5 μL HEPES buffer was added to adjust the salt content. The antibody-antigen complex was lyophilized over night at 0° C. and subsequently at 15° C. for 2 h to remove as much water as possible. The ten lyophilized aliquots were frozen at −20° C. until HDX-exchange and LC-MS analysis. One aliquot of each of the antibody-antigen complex and the antigen without antibody was solubilized in 12.5 μl H2O, the others in 12.5 μL D2O. Aliquots were incubated for the following times, whereby each aliquot was prepared separately right before analysis:0 minutes (H2O reference samples);5, 70, 360 minutes and 24 hours (D2O deuterium exchange kinetic samples). The exchange was quenched by addition of 12.5 μL of freshly prepared quenching solution (guanidine hydrochloride 0.8 M with TCEP 0.4 M in 100 mM ammonium formate buffer pH 2.5). Peptic Digest Immediately after addition of the quenching solution, 0.35 μL pepsin (100 μM) were added and digestion performed for 2 minutes at 20° C. Aliquots were placed immediately in −20° C. pre-cooled autosampler vials and injected via a pre-cooled injection syringe into the LC-MS. LC-MS The peptide mixture obtained was injected and separated without pretreatment using reverse phase HPLC (RSLC3000 LC, Thermo Scientific Dionex, Idstein, Germany). An LC column (ACQUITY UPLC BEH300 C18 1.7 μm 1×50 mm Thermo Scientific Dionex, Idstein, Germany) was used for separation of the sample. Blank runs and column wash runs were performed within consecutive sample runs. Chromatographic separation was achieved by using a nearly isocratic gradient for 31 minutes. Eluent A was water with 0.1% formic acid and eluent B was acetonitrile with 0.1% formic acid. An optimized 20-minute linear gradient with varying slopes was applied at −0° C. as follows (minute/% B): 0/8, 3/8, 11.9/20, 31.9/20, 33/99, 34/99, 35/8. Manual injection was performed. The injection amount was 25.35 μL using a sample loop of 20 μL volume. Flow rate was 40 μL/min. The HPLC eluate was directly infused into a QTOF-type mass spectrometer (MaXis HD, Bruker). The mass spectrometer operated in positive ion mode, the spray voltage was 1.9 kV, the capillary temperature was 275° C. and the S-Lens RF voltage was 55 V. Data Analysis The data was analysed using the software HDExaminer 2.40 beta 1 64 bit (Sierra Analytics, Modest, CA, USA). Briefly, a raw dataset containing different exchange time points, and for each time point the analysis of Anx-A1 with and without antibody was examined. Using the Anx-A1 sequence information and a sequence list of peptic peptides with corresponding retention times and charge, the software identifies the peptides with and without deuterium exchange and calculates the deuterium uptake per peptide as being the difference between the centroid mass of the deuterated versus the non-deuterated peptide. By using overlapping peptide information (mass shift of individual overlapping peptides) the epitope region was manually further limited. Results After the initial data evaluation using HDExaminer the individual peptic peptides were manually verified for a statistically relevant uptake of deuterium. In case of several overlapping peptides, the epitope region was further limited using HDX-data without deuterium uptake covering the N- and C-terminal parts of the peptide with deuterium uptake. The whole experiment was repeated twice. In the first experiment identified a potential epitope region was identified but a statistically relevant deuterium uptake was also observed in a very long peptide containing the N-terminus, which from a structural point of view is rather flexible. In the second experiment it was possible to confirm the epitope region, while the N-terminus showed no deuterium uptake. The underlined regions in the sequence indicate the epitope bound by the antibody: (SEQ ID NO: 17)MAMVSEFLKQAWFIENEEQEYVQTVKSSKGGPGSAVSPYPTFNPSSDVAALHKAIMVKGVDEATIIDILTKRNNAQRQQIKAAYLQETGKPLDETLKKALTGHLEEVVLALLKTPAQFDADELRAAMKGLGTDEDTLIEILASRTNKEIRDINRVYREELKRDLAKDITSDTSGDFRNALLSLAKGDRSEDFGVNEDLADSDARALYEAGERRKGTDVNVFNTILTTRSYPQLRRVFQKYTKYSKHDMNKVLDLELKGDIEKCLTAIVKCATSKPAFFAEKLHQAMKGVGTRHKALIRIMVSRSEIDMNDIKAFYQKMYGISLCQAILDETKGDYEKILVALCGGN The identified epitope regions are not in the Anx-A1 self-interaction region, but peptides from the self-interaction region show a slight tendency to more deuterium uptake in the antibody-antigen complex samples, which might by due to slight differences in local Anx-A1 concentration when two Anx-A1 molecules are bound to the two arms of the antibody. Example 3—In Vivo Anti-Cancer Activity of MDX-124 Methods Tolerability Study The tolerability study was performed by Crown Bioscience (USA). Mice were dosed once per week for 2 weeks with MDX-124 at 1 mg/kg, 10 mg/kg or 29 mg/kg. Body weight of each mouse was measured daily for the duration of the study. A reduction in body weight would be taken as an indication of toxicity of the antibody to the mice. Murine Breast Cancer Model Mouse work was performed by Crown Bioscience. The mice used were 8-9 week old female BALB/c mice. The breast cancer model used utilised the luciferase expressing murine breast cancer cell line 4T1-Luc. The cell line was obtained from the ATCC and cultured in RPMI medium containing 10% FBS, 2 mM L-glutamine and 2 μg/ml puromycin. Mice were shaved, then 72 hours later a transponder chip implanted for the purposes of individual mouse identification. Bepanthen cream was applied immediately following shaving and then daily until tumour inoculation. Each mouse was first inoculated with 5×1044T1-Luc cells, suspended in 100 μl PBS. Inoculation was performed on day 0 into a mammary fat pad (lower left side, 2ndpad from bottom) while mice were under gaseous anaesthesia. The skin at the inoculation site was cleaned with 70% ethanol prior to inoculation. Tumour size was measured three times a week starting from day 5, using an IVIS Spectrum In Vivo Imaging System (PerkinElmer, USA). Bioluminescent imaging was used to measure each tumour in 2 dimensions, using electronic callipers. Tumour volumes were estimated using the formula 0.5(L×W2), where L=tumour length and W=tumour width. Treatment began when the mean tumour volume reached 50-60 mm3. After the first tumour measurement, bepanthen cream was again applied to the area around the tumour. Bepanthen cream was subsequently applied daily. The mice were split into 4 groups of 12 mice each, with uniform mean tumour volume between groups. Treatment was administered weekly. Of the four mouse groups, a control group was dosed with vehicle only (PBS). The three experimental groups received doses of 1, 10 or 25 mg/kg MDX-124, in PBS. Each dose was given intravenously in a volume of 10 ml/kg. Treatment was to be continued for up to 3 weeks. Thrice weekly tumour measurement continued following the commencement of treatment. Mice were weighed three times a week prior to treatment commencing, and daily thereafter. Results To check that MDX-124 was not inherently toxic to mice a tolerability study was performed. Mice were administered the antibody and their body weights monitored. No body weight loss was evident in the mice (data not shown), indicating that the antibody was not toxic to them at any of the tested doses. The anti-cancer effect of MDX-124 was then tested in a murine model of breast cancer. Average tumour volumes for each group of mice tested are shown inFIG.20. Following tumour cell inoculation on day 0, the first treatment dose was administered to all groups on day 12. As shown, by day 17 of the study, mice treated with MDX-124 had significantly lower tumour volumes than the control mice treated with a vehicle. The pattern of increased tumour growth in the control group continued to day 19. The lower tumour volumes seen in the groups treated with MDX-124 also corresponded to lower relative tumour volumes in these groups (seeFIG.21). The tumour volume at day 12 was defined as the baseline tumour volume (i.e. 100% tumour volume). By day 19 the tumours of the mice treated with MDX-124 had increased in size approximately 2.5-fold. The tumours of the control mice had increased in size approximately 3.3-fold. This means that treatment with MDX-124 resulted in an approximately one-third reduction in tumour growth relative to the control by day 19, demonstrating the anti-cancer effect of the antibody. While the antibody was administered to the mice at three different concentrations (1 mg/kg, 10 mg/kg and 25 mg/kg), each of these treatment regimes had a similar effect on tumour growth (i.e. increasing the amount of antibody administered did not seem to increase the effect of the treatment).
27,716
11858983
DETAILED DESCRIPTION The disclosure provides an antibody that specifically binds to sclerostin, wherein the antibody comprises a heavy chain comprising a sequence of amino acids comprising Pro-Ala-Arg-Gly (SEQ ID NO: 8) at the C-terminus of the heavy chain. In some embodiments, the antibody comprises a first heavy comprising a sequence of amino acids comprising Pro-Ala-Arg-Gly (SEQ ID NO: 8) at the C-terminus of the heavy chain and a second heavy chain comprising a wild-type heavy chain amino acid sequence. In some embodiments, the antibody comprises a sequence of amino acids comprising Pro-Ala-Arg-Gly-Lys (SEQ ID NO: 11) at the C-terminus of the heavy chain. Pharmaceutical compositions comprising the antibody (or mixture of antibodies) and methods of using the antibody are also provided. An “anti-sclerostin antibody” or an “antibody that binds to sclerostin” is an antibody that binds to sclerostin of SEQ ID NO: 1 or portions thereof. Recombinant human sclerostin/SOST is commercially available from, e.g., R&D Systems (Minneapolis, Minn., USA; 2006 Catalog #1406-ST-025). U.S. Pat. Nos. 6,395,511 and 6,803,453, and U.S. Patent Publication Nos. 2004/0009535 and 2005/0106683 refer to anti-sclerostin antibodies generally. Examples of sclerostin antibodies suitable for use in the context of the invention also are described in U.S. Patent Publication Nos. 2007/0110747 and 2007/0072797, which are hereby incorporated by reference. Additional information regarding materials and methods for generating sclerostin antibodies can be found in U.S. Patent Publication No. 20040158045 (hereby incorporated by reference). The term “antibody” refers to an intact immunoglobulin molecule (including polyclonal, monoclonal, chimeric, humanized, and/or human versions having full length heavy and/or light chains). “Specifically binds” as used herein means that the antibody preferentially binds the antigen over other proteins. In some embodiments, “specifically binds” means the antibody has a higher affinity for the antigen than for other proteins. Antibodies that specifically bind an antigen may have a binding affinity for the antigen of less than or equal to 1×10−7M, less than or equal to 2×10−7M, less than or equal to 3×10−7M, less than or equal to 4×10−7M, less than or equal to 5×10−7M, less than or equal to 6×10−7M, less than or equal to 7×10−7M, less than or equal to 8×10−7M, less than or equal to 9×10−7M, less than or equal to 1×−8M, less than or equal to 2×10−8M, less than or equal to 3×10−8M, less than or equal to 4×10−8M, less than or equal to 5×10−8M, less than or equal to 6×10−8M, less than or equal to 7×10−8M, less than or equal to 8×10−8M, less than or equal to 9×10−8M, less than or equal to 1×10−9M, less than or equal to 2×10−9M, less than or equal to 3×10−9M, less than or equal to 4×10−9M, less than or equal to 5×10−9M, less than or equal to 6×10−9M, less than or equal to 7×10−9M, less than or equal to 8×10−9M, less than or equal to 9×10−9M, less than or equal to 1×10−10M, less than or equal to 2×10−10M, less than or equal to 3×10−10M, less than or equal to 4×10−10M, less than or equal to 5×10−10M, less than or equal to 6×10−10M, less than or equal to 7×10−10M, less than or equal to 8×10−10M, less than or equal to 9×10−10M, less than or equal to 1×10−11M, less than or equal to 2×10−11M, less than or equal to 3×10−11M, less than or equal to 4×10−11M, less than or equal to 5×10−11M, less than or equal to 6×10−11M, less than or equal to 7×10−11M, less than or equal to 8×10−11M, less than or equal to 9×10−11M, less than or equal to 1×10−12M, less than or equal to 2×10−12M, less than or equal to 3×10−12M, less than or equal to 4×10−12M, less than or equal to 5×10−12M, less than or equal to 6×10−12M, less than or equal to 7×10−12M, less than or equal to 8×10−12M, or less than or equal to 9×10−12M. In some or any embodiments, the antibody binds to sclerostin of SEQ ID NO: 1, or a naturally occurring variant thereof, with an affinity (Kd) of less than or equal to 1×10−7M, less than or equal to 1×10−8M, less than or equal to 1×10−9M, less than or equal to 1×10−10M, less than or equal to 1×10−11M, or less than or equal to 1×10−12M. Affinity is determined using a variety of techniques, an example of which is an affinity ELISA assay. In various embodiments, affinity is determined by a BIAcore assay. In various embodiments, affinity is determined by a kinetic method. In various embodiments, affinity is determined by an equilibrium/solution method. U.S. Patent Publication No. 2007/0110747 (the disclosure of which is incorporated herein by reference) contains additional description of affinity assays suitable for determining the affinity (Kd) of an antibody for sclerostin. In some or any embodiments, the antibody (or antibody fragments thereof) binds to a sclerostin polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 1 and binds a region of sclerostin comprising the sequence of SEQ ID NO: 5 (CGPARLLPNAIGRGKWWRPSGPDFRC; corresponding to amino acids 86-111 of SEQ ID NO: 1). This region is also referred to herein as the “loop 2” region of sclerostin. Regions of sclerostin outside of the loop 2 region are defined herein as “non-loop 2 regions.” Alternatively or in addition, the anti-sclerostin antibody binds to a sclerostin polypeptide comprising amino acids 57-146 of SEQ ID NO: 1. Alternatively or in addition, the anti-sclerostin antibody binds to a sclerostin polypeptide comprising amino acids 89-103 of SEQ ID NO: 1 and/or amino acids 137-151 of SEQ ID NO: 1. In some or any embodiments, the sclerostin polypeptide that is a fragment of full length sclerostin retains the tertiary structure of the corresponding polypeptide region of human sclerostin of SEQ ID NO: 1. In some or any embodiments, the anti-sclerostin antibody described herein preferably modulates sclerostin function in the cell-based assay described in U.S. Patent Publication No. 2007/0110747 and/or the in vivo assay described in U.S. Patent Publication No. 20070110747 and/or bind to one or more of the epitopes described in U.S. Patent Publication No. 2007/0110747 and/or cross-block the binding of one of the antibodies described in U.S. Patent Publication No. 2007/0110747 and/or are cross-blocked from binding sclerostin by one of the antibodies described in U.S. Patent Publication No. 2007/0110747 (incorporated by reference in its entirety and for its description of assays for characterizing an anti-sclerostin antibody). “CDR” refers to the complementarity determining region within antibody variable sequences. There are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDR1, CDR2 and CDR3, for each of the variable regions. The term “set of six CDRs” as used herein refers to a group of three CDRs that occur in the light chain variable region and heavy chain variable region, which are capable of binding the antigen. The exact boundaries of CDRs have been defined differently according to different systems. The system described by Kabat (Kabat et al., Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987) and (1991)) not only provides an unambiguous residue numbering system applicable to any variable region of an antibody, but also provides precise residue boundaries defining the three CDRs. These CDRs may be referred to as Kabat CDRs. Chothia and coworkers (Chothia & Lesk, J. Mol. Biol. 196:901-917 (1987) and Chothia et al., Nature 342:877-883 (1989)) found that certain sub-portions within Kabat CDRs adopt nearly identical peptide backbone conformations, despite having great diversity at the level of amino acid sequence. These sub-portions were designated as L1, L2 and L3 or H1, H2 and H3 where the “L” and the “H” designates the light chain and the heavy chains regions, respectively. These regions may be referred to as Chothia CDRs, which have boundaries that overlap with Kabat CDRs. Other boundaries defining CDRs overlapping with the Kabat CDRs have been described by Padlan (FASEB J. 9:133-139 (1995)) and MacCallum (J Mol Biol 262(5):73245 (1996)). Still other CDR boundary definitions may not strictly follow one of the above systems, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. The methods used herein may utilize CDRs defined according to any of these systems, although preferred embodiments use Kabat or Chothia defined CDRs. CDRs are obtained by, e.g., constructing polynucleotides that encode the CDR of interest. Such polynucleotides are prepared, for example, by using the polymerase chain reaction to synthesize the variable region using mRNA of antibody-producing cells as a template (see, for example, Larrick et al.,Methods: A Companion to Methods in Enzymology,2:106 (1991); Courtenay-Luck, “Genetic Manipulation of Monoclonal Antibodies,” inMonoclonal Antibodies Production, Engineering and Clinical Application, Ritter et al. (eds.), page 166, Cambridge University Press (1995); and Ward et al., “Genetic Manipulation and Expression of Antibodies,” inMonoclonal Antibodies: Principles and Applications, Birch et al., (eds.), page 137, Wiley-Liss, Inc. (1995)). In various aspects, the antibody comprises at least one CDR sequence having at least 75% identity (e.g., at least 75%, 80%, 85%, 90%, 95% or 100% identity) to a CDR selected from CDR-H1, CDR-H2, CDR-H3, CDR-L1, CDR-L2, and CDR-L3 wherein CDR-H1 has the sequence given in SEQ ID NO: 2, CDR-H2 has the sequence given in SEQ ID NO: 3, CDR-H3 has the sequence given in SEQ ID NO: 4, CDR-L1 has the sequence given in SEQ ID NO: 5, CDR-L2 has the sequence given in SEQ ID NO: 6 and CDR-L3 has the sequence given in SEQ ID NO: 7. The anti-sclerostin antibody, in various aspects, comprises two of the CDRs or six of the CDRs. In a preferred embodiment, the anti-sclerostin antibody comprise a set of six CDRs as follows: CDR-H1 of SEQ ID NO: 2, CDR-H2 of SEQ ID NO: 3, CDR-H3 of SEQ ID NO: 4, CDR-L1 of SEQ ID NO: 5, CDR-L2 of SEQ ID NO: 6 and CDR-L3 of SEQ ID NO: 7. In some or any embodiments, the antibody comprises a light chain variable region comprising an amino acid sequence having at least 75% identity (e.g., at least 75%, 80%, 85%, 90%, 95% or 100% identity) to the amino acid sequence set forth in SEQ ID NO: 9 and a heavy chain variable region comprising an amino acid sequence having at least 75% identity (e.g., at least 75%, 80%, 85%, 90%, 95% or 100% identity) to the amino acid sequence set forth in SEQ ID NO: 10. In various aspects, the difference in the sequence compared to SEQ ID NO: 9 or 10 lies outside the CDR region in the corresponding sequences. In some or any embodiments, the antibody comprises a light chain variable region comprising an amino acid sequence set forth in SEQ ID NO: 9 and a heavy chain variable region comprising an amino acid sequence set forth in SEQ ID NO: 10. In some or any embodiments the anti-sclerostin antibody comprises all or part of a heavy chain (e.g., two heavy chains) comprising an amino acid sequence having at least 75% identity (e.g., at least 75%, 80%, 85%, 90%, 95% or 100% identity) to the amino acid sequence set forth in SEQ ID NO: 16 and all or part of a light chain (e.g., two light chains) comprising an amino acid sequence having at least 75% identity (e.g., at least 75%, 80%, 85%, 90%, 95% or 100% identity) to the amino acid sequence set forth in SEQ ID NO 12. The antibody comprises a heavy chain comprising the amino acid sequence Pro-Ala-Arg-Gly (SEQ ID NO: 8) at the C-terminus of the heavy chains. In some embodiments, the C-terminus of both heavy chains of the antibody comprises the amino acid sequence Pro-Ala-Arg-Gly (SEQ ID NO: 8). In some embodiments, the antibody comprises a first heavy chain comprising the amino acid sequence Pro-Ala-Arg-Gly (SEQ ID NO: 8) and a second heavy chain comprising a wild-type amino acid sequence. The antibody, in various aspects, comprises the light chain amino acid sequence set forth in SEQ ID NO: 12 and the heavy chain amino acid sequence set forth in SEQ ID NO: 13. Alternatively, in some or any embodiments, the antibody comprises a sequence of amino acids comprising Pro-Ala-Arg-Gly-Lys (SEQ ID NO: 11) at the C-terminus of a heavy chain, optionally at the C-terminus of both heavy chains. In some embodiments, the antibody comprises a first heavy chain comprising the amino acid sequence Pro-Ala-Arg-Gly-Lys (SEQ ID NO: 11) and a second heavy chain comprising a wild-type amino acid sequence (i.e., without the C-terminal Pro-Ala-Arg-Gly-Lys (SEQ ID NO: 11)). The antibody, in various aspects, comprises the light chain amino acid sequence set forth in SEQ ID NO: 12 and the heavy chain amino acid sequence set forth in SEQ ID NO: 14. Examples of other anti-sclerostin antibodies include, but are not limited to, the anti-sclerostin antibodies disclosed in International Patent Publication Nos. WO 2008/092894, WO 2008/115732, WO 2009/056634, WO 2009/047356, WO 2010/100200, WO 2010/100179, WO 2010/115932, and WO 2010/130830 (each of which is incorporated by reference herein in its entirety). It will be understood by one skilled in the art that some proteins, such as antibodies, may undergo a variety of posttranslational modifications. The type and extent of these modifications often depends on the host cell line used to express the protein as well as the culture conditions. Such modifications may include variations in glycosylation, methionine oxidation, diketopiperizine formation, aspartate isomerization and asparagine deamidation. A frequent modification is the loss of a carboxy-terminal basic residue (such as lysine or arginine) due to the action of carboxypeptidases (as described in Harris, R J.Journal of Chromatography705:129-134, 1995). Other modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the α-amino groups of lysine, arginine, and histidine side chains (T. E. Creighton, Proteins: Structure and Molecular Properties, W. H. Freeman & Co., San Francisco, pp. 79-86 [1983], entirely incorporated by reference), acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group. In some or any embodiments, the C-terminus of the heavy chain of the antibody, comprising the amino acid sequence Pro-Ala-Arg-Gly (SEQ ID NO: 8), is amidated. In some or any embodiments, both heavy chains of the antibody comprise the amino acid sequence Pro-Ala-Arg-Gly (SEQ ID NO: 8) and both heavy chains are amidated. In some embodiments, the glycine is amidated. Amidation can occur, e.g., as described in Prigg, S. T. et al., “New insights into copper monooxygenases and peptide amidation: structure, mechanism and function”, Cell. Mol. Life Sci. 57 (2000) 1236-1259. The enzyme peptidylglycine α-amidating monooxygenase (PAM) can catalyze the amidation of glycine. PAM has two active domains, peptidylglycine α-hydroxylating monooxygenase (PHM) and peptidyl-α-hydroxylglycine α-amidating lyase (PAL). PHM catalyzes the conversion of peptidylglycine (along with ascorbate and oxygen) to peptidyl α-hydroxylglycine (along with semidehydrogenascorbate and water). In turn, PAL catalyzes the conversion of peptidyl α-hydroxylglycine to an amidated peptide (and glyoxylate). Amidation of an antibody can be controlled by altering certain conditions during the cell culture process. For example, copper (e.g., in ferric ammonium citrate) and/or oxygen levels may be used to influence amidation levels. It is contemplated that increasing copper concentration (e.g., in the media) or oxygen availability (e.g., during culturing) may increase amidation by impacting the activity of an enzyme such as PHM. Pharmaceutical Compositions The disclosure provides a pharmaceutical composition comprising a population of the antibody described herein together with a pharmaceutically effective diluent, carrier, solubilizer, emulsifier, preservative, and/or adjuvant. Pharmaceutical compositions of the invention include, but are not limited to, liquid, frozen, and lyophilized compositions. The disclosure also provides a pharmaceutical composition comprising a mixture of antibodies that specifically bind to sclerostin of SEQ ID NO: 1 and a pharmaceutically acceptable carrier, wherein about 3-5% of the antibodies in the composition are a population of antibodies described herein (e.g., antibodies comprising set of six CDRs set forth in SEQ ID NOs: 2-7 and having a heavy chain (or two heavy chains) comprising the amino acid sequence Pro-Ala-Arg-Gly (SEQ ID NO: 8) at the C-terminus of the heavy chain(s)). The disclosure also contemplates compositions comprising alternative amounts (e.g., 5-10%, 1-3%, 3-15%, 2-10%, 4-20%, 1-5%) of the population of antibodies described herein (e.g., antibodies comprising set of six CDRs set forth in SEQ ID NOs: 2-7 and having a heavy chain (or two heavy chains) comprising the amino acid sequence Pro-Ala-Arg-Gly (SEQ ID NO: 8) at the C-terminus of the heavy chain(s)). In some embodiments, less than 70% of the antibodies of the population (e.g., about 69%, about 68%, about 67%, about 66%, about 65%, about 64%, about 63%, about 62%, about 61%, about 60%, about 59%, about 58%, about 57%, about 56%, about 55%, about 54%, about 53%, about 52%, about 51%, about 50%, about 49%, about 48%, about 47%, about 46%, about 45%, about 44%, about 43%, about 42%, about 41%, about 40%, about 39%, about 38%, about 37%, about 36%, about 35%, about 34%, about 33%, about 32%, about 31%, about 30%, about 29%, about 28%, about 27%, about 26%, about 25%, about 24%, about 23%, about 22%, about 21%, about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, bout 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1% or less) comprise a heavy chain comprising a C-terminal Pro-Ala-Arg-Gly (SEQ ID NO: 8) sequence, which is optionally amidated. In some embodiments, less than 35% (e.g., about 34%, about 33%, about 32%, about 31%, about 30%, about 29%, about 28%, about 27%, about 26%, about 25%, about 24%, about 23%, about 22%, about 21%, about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, bout 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1% or less) of the antibodies of the population comprise a C-terminal Pro-Ala-Arg-Gly (SEQ ID NO: 8) sequence on both heavy chains, where both heavy chains are optionally amidated. It is also contemplated that both heavy chains comprise the C-terminal Pro-Ala-Arg-Gly (SEQ ID NO: 8) sequence but only one of the chains is amidated. In some embodiments, less than 35% (e.g., about 34%, about 33%, about 32%, about 31%, about 30%, about 29%, about 28%, about 27%, about 26%, about 25%, about 24%, about 23%, about 22%, about 21%, about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, bout 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1% or less) of the antibodies in the composition comprise a C-terminal Pro-Ala-Arg-Gly (SEQ ID NO: 8) sequence that is not amidated. In some embodiments, about 33% of antibodies of the population comprise a C-terminal Pro-Ala-Arg-Gly (SEQ ID NO: 8) sequence that is amidated, about 33% of the antibodies of the population comprise C-terminal Pro-Ala-Arg-Gly (SEQ ID NO: 8) sequences on both heavy chains which are both amidated, and about 33% of the antibodies of the population comprise heavy chain(s) with a C-terminal Pro-Ala-Arg-Gly (SEQ ID NO: 8) sequence but which are not amidated. In some embodiments, the pharmaceutical composition contains formulation materials for modifying, maintaining or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition. In such embodiments, suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine, proline, or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides; disaccharides; and other carbohydrates (such as glucose, mannose or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring, flavoring and diluting agents; emulsifying agents; hydrophilic polymers (such as polyvinylpyrrolidone); low molecular weight polypeptides; salt-forming counterions (such as sodium); preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine, sorbic acid or hydrogen peroxide); solvents (such as glycerin, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); suspending agents; surfactants or wetting agents (such as pluronics, PEG, sorbitan esters, polysorbates such as polysorbate 20, polysorbate, triton, tromethamine, lecithin, cholesterol, tyloxapal); stability enhancing agents (such as sucrose or sorbitol); tonicity enhancing agents (such as alkali metal halides, preferably sodium or potassium chloride, mannitol sorbitol); delivery vehicles; diluents; excipients and/or pharmaceutical adjuvants. See, REMINGTON'S PHARMACEUTICAL SCIENCES, 18″ Edition, (A. R. Genrmo, ed.), 1990, Mack Publishing Company. Selection of the particular formulation materials described herein may be driven by, for example, the intended route of administration, delivery format and desired dosage. See, for example, REMINGTON'S PHARMACEUTICAL SCIENCES, supra. The primary vehicle or carrier in a pharmaceutical composition may be either aqueous or non-aqueous in nature. For example, a suitable vehicle or carrier may be water for injection, physiological saline solution or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. In specific embodiments, pharmaceutical compositions comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, and may further include sorbitol or a suitable substitute therefor. In certain embodiments, the composition may be prepared for storage by mixing the selected composition having the desired degree of purity with optional formulation agents (REMINGTON'S PHARMACEUTICAL SCIENCES, supra) in the form of a lyophilized cake or an aqueous solution. Further, in some embodiments, the antibody or fragment may be formulated as a lyophilizate using appropriate excipients such as sucrose. The pharmaceutical compositions of the invention can be selected for parenteral delivery. Alternatively, the compositions may be selected for inhalation or for delivery through the digestive tract, such as orally. Preparation of such pharmaceutically acceptable compositions is within the skill of the art. The formulation components are present preferably in concentrations that are acceptable to the site of administration. In certain embodiments, buffers are used to maintain the composition at physiological pH or at a slightly lower pH, typically within a pH range of from about 5 to about 8. When parenteral administration is contemplated, the therapeutic compositions for use in this invention may be provided in the form of a pyrogen-free, parenterally acceptable aqueous solution comprising the desired antibody or fragment in a pharmaceutically acceptable vehicle. A particularly suitable vehicle for parenteral injection is sterile distilled water in which the antibody or fragment is formulated as a sterile, isotonic solution, properly preserved. In certain embodiments, implantable drug delivery devices may be used to introduce the desired antibody or fragment. In some or any embodiments, the pharmaceutical composition described herein comprises a calcium salt, an acetate buffer, a polyol and a surfactant. Exemplary calcium salts include, but are not limited to, calcium acetate, calcium carbonate and calcium chloride. In some embodiments, the calcium salt is at a concentration of at least 0.5 mM, at least 1 mM, at least 2 mM, at least 3 mM, at least 4 mM, at least 5 mM, at least 6 mM, at least 7 mM, at least 8 mM, at least 9 mM or at least 10 mM. In certain embodiments, the concentration of calcium salt is not greater than 11 mM, no greater than 12 mM, no greater than 13 mM, no greater than 14 mM, no greater than 15 mM, no greater than 16 mM, no greater than 17 mM, no greater than 18 mM, no greater than 19 mM, no greater than 20 mM, no greater than 21 mM, no greater than 22 mM, no greater than 23 mM, no greater than 24 mM, or no greater than 25 mM. Any range featuring a combination of the foregoing endpoints is contemplated, including but not limited to from about 0.5 mM to about 10 mM, about 5 mM to about 10 mM, or about 5 mM to about 15 mM. In some embodiments, the pharmaceutical composition comprises an acetate buffer (e.g., sodium acetate) having a concentration ranging from about 0.1 mM to about 1000 mM (1 M). In some embodiments, the concentration of the acetate buffer is at least 5 mM, at least 6 mM, at least 7 mM, at least 8 mM, at least 9 mM, at least 10 mM, at least 15 mM, at least mM, at least 70 mM, at least 80 mM, at least 90 mM, at least 100 mM, at least 200 mM, at least 500 mM, at least 700 mM, or at least 900 mM. In some embodiments, the concentration of the acetate buffer is no greater than 10 mM, no greater than 15 mM, no greater than 20 mM, no greater than 25 mM, no greater than 30 mM, no greater than 35 mM, no greater than mM, no greater than 45 mM, no greater than 50 mM, no greater than 55 mM, no greater than 60 mM, no greater than 65 mM, no greater than 70 mM, no greater than 75 mM, no greater than 80 mM, no greater than 85 mM, no greater than 90 mM, no greater than 95 mM or no greater than 100 mM. Any range featuring a combination of the foregoing endpoints is contemplated, including but not limited to from about 5 mM to about 15 mM, or from about 5 mM to about 10 mM or from about 10 mM to about 25 mM. The buffer is preferably added to a concentration that maintains pH around 5-6 or 5-5.5 or 4.5-5.5. When the calcium salt in the formulation is calcium acetate, in some embodiments, the total concentration of acetate is about 10 mM to about 55 mM, or about 20 mM to about 40 mM. In some aspects, the pharmaceutical composition comprises a total concentration of acetate that is at least about 10 mM, at least about 15 mM, at least about 20 mM, at least about 25 mM, at least about 30 mM, at least about 35 mM, at least about 40 mM, 45 mM, or mM. In some embodiments, the concentration of acetate is no greater than about 30 mM, mM, 40 mM, 45 mM, 50 mM, 55 mM, 60 mM, 65 mM, 70 mM, 75 mM, 80 mM, 85 mM, or 90 mM. Any range featuring a combination of the foregoing endpoints is contemplated, including but not limited to: about 10 mM to about 50 mM, about 20 mM to about 50 mM, about 20 mM to about 40 mM, about 30 mM to about 50 mM, or about 30 mM to about 75 mM. In some embodiments, the calcium salt is calcium acetate and the acetate buffer is sodium acetate. By way of nonlimiting example, a solution containing 10 mM calcium acetate will have 20 mM acetate anion and 10 mM of calcium cation, because of the divalent nature of the calcium cation, while a solution containing 10 mM sodium acetate will have 10 mM sodium cation and 10 mM acetate anion. In some embodiments, the total concentration of ions (cations and anions) in solution is at least 10 mM, at least about 15 mM, at least about 20 mM, at least about 25 mM, at least about 30 mM, at least about 35 mM, at least about 40 mM, at least about 45 mM, at least about 50 mM, at least about 55 mM, at least about 60 mM, at least about 65 mM, at least about 70 mM, at least about 75 mM, at least about 80 mM, or at least about 85 mM. In some embodiments, the total concentration of ions is no greater than about 30 mM, no greater than about 35 mM, no greater than about 40 mM, no greater than about 45 mM, no greater than about 50 mM, no greater than about 55 mM, no greater than about 60 mM, no greater than about 65 mM, no greater than about 70 mM, no greater than about 75 mM, no greater than about 80 mM, no greater than about 85 mM, no greater than about 90 mM, no greater than about 95 mM, no greater than about 100 mM, no greater than about 110 mM, no greater than about 120 mM, no greater than about 130 mM, no greater than about 140 mM, no greater than about 150 mM, no greater than about 160 mM, no greater than about 170 mM, no greater than about 180 mM, no greater than about 190 mM or no greater than about 200 mM. Any range featuring a combination of the foregoing endpoints is contemplated, including but not limited to: about 30 mM to about 60 mM, or about 30 mM to about 70 mM, or about 30 mM to about 80 mM, or about 40 mM to about 150 mM, or about 50 mM to about 150 mM. By way of nonlimiting example, a solution of 10 mM calcium acetate will have a 30 mM total concentration of ions (10 mM cations and 20 mM anions). In some or any embodiments, the pharmaceutical composition comprises a polyol. Polyols encompass a class of excipients that includes sugars (e.g. mannitol, sucrose, sorbitol) and other polyhydric alcohols (e.g., glycerol and propylene glycol). Exemplary polyols include, but are not limited to, propylene glycol, glycerin (glycerol), threose, threitol, erythrose, erythritol, ribose, arabinose, arabitol, lyxose, maltitol, sorbitol, sorbose, glucose, mannose, mannitol, levulose, dextrose, maltose, trehalose, fructose, xylitol, inositol, galactose, xylose, fructose, sucrose, 1,2,6-hexanetriol and the like. Higher order sugars include, but are not limited to, dextran, propylene glycol, or polyethylene glycol. Reducing sugars such as fructose, maltose or galactose oxidize more readily than do non-reducing sugars. Additional examples of sugar alcohols are glucitol, maltitol, lactitol or iso-maltulose. Additional exemplary lyoprotectants include glycerin and gelatin, and the sugars mellibiose, melezitose, raffinose, mannotriose, and stachyose. Examples of reducing sugars include glucose, maltose, lactose, maltulose, iso-maltulose and lactulose. Examples of non-reducing sugars include non-reducing glycosides of polyhydroxy compounds selected from sugar alcohols and other straight chain polyalcohols. Monoglycosides include compounds obtained by reduction of disaccharides such as lactose, maltose, lactulose and maltulose. In some or any embodiments, the pharmaceutical composition comprises a polyol at a concentration ranging from about 0% to about 40% w/v. In some or any embodiments, the compositions comprise a polyol at concentration of at least 0.5, at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 30, or at least 40% w/v. In some or any embodiments, the composition comprises a polyol at a concentration of about 1, 2, 3, 4, 5, 6, 7, 8, 9% to about 10% w/v. In some or any embodiments, the composition comprises a polyol at a concentration of about 2% to about 6% w/v. In some or any embodiments, the composition comprises a polyol at a concentration of about 4% w/v. In some or any embodiments, the composition comprises a polyol at about 6% w/v. In some or any embodiments, the pharmaceutical composition comprises a surfactant. Exemplary surfactants include, but are not limited to, anionic, cationic, nonionic, zwitterionic, and amphoteric surfactants including surfactants derived from naturally-occurring amino acids. Anionic surfactants include, but are not limited to, sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate, chenodeoxycholic acid, N-lauroylsarcosine sodium salt, lithium dodecyl sulfate, 1-octanesulfonic acid sodium salt, sodium cholate hydrate, sodium deoxycholate, and glycodeoxycholic acid sodium salt. Cationic surfactants include, but are not limited to, benzalkonium chloride or benzethonium chloride, cetylpyridinium chloride monohydrate, and hexadecyltrimethylammonium bromide. Zwitterionic surfactants include, but are not limited to, CHAPS, CHAPSO, SB3-10, and SB3-12. Non-ionic surfactants include, but are not limited to, digitonin, Triton X-100, Triton X-114, TWEEN-20, and TWEEN-80. In another embodiment, surfactants include, but are not limited to, lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 40, 50 and 60, glycerol monostearate, polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 65 and polysorbate 80, soy lecithin and other phospholipids such as DOPC, DMPG, DMPC, and DOPG; sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose. In some or any embodiments, the surfactant is polysorbate 20. Surfactants may be included in the compositions either individually or as a mixture in different ratios. In some or any embodiments, the composition comprises a surfactant at a concentration of about 0% to about 5% w/v (e.g., about 0.001, about 0.002, about 0.005, about 0.007, about 0.01, about 0.05, about 0.1, about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1.0, about 1.5, about 2.0, about 2.5, about 3.0, about 3.5, about 4.0, or about 4.5% w/v). In some or any embodiments, the composition comprises a surfactant at a concentration of about 0.001% to about 0.5% w/v. In some or any embodiments, the composition comprises a surfactant at a a concentration of about 0.004, about 0.005, about 0.007, about 0.01, about 0.05, or about 0.1% w/v to about 0.2% w/v. In some or any embodiments, the composition comprises a surfactant at a concentration of about 0.01% to about 0.1% w/v. In some or any embodiments, the pharmaceutical composition comprises 55 mM acetate, 13 mm calcium, 6.0% (w/v) sucrose, 0.006% (w/v) polysorbate 20, pH 5.2. Additional pharmaceutical compositions will be evident to those skilled in the art, including formulations involving antigen binding proteins in sustained- or controlled-delivery formulations. Techniques for formulating a variety of other sustained- or controlled-delivery means, such as liposome carriers, bio-erodible microparticles or porous beads and depot injections, are also known to those skilled in the art. See, for example, International Patent Application No. PCT/US93/00829, which is incorporated by reference and describes controlled release of porous polymeric microparticles for delivery of pharmaceutical compositions. Sustained-release preparations may include semipermeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules. Sustained release matrices may include polyesters, hydrogels, polylactides (as disclosed in U.S. Pat. No. 3,773,919 and European Patent Application Publication No. EP058481, each of which is incorporated by reference), copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et al., 1983, Biopolymers 2:547-556), poly (2-hydroxyethyl-methacrylate) (Langer et al., 1981, J. Biomed. Mater. Res. 15:167-277 and Langer, 1982, Chem. Tech. 12:98-105), ethylene vinyl acetate (Langer et al., 1981, supra) or poly-D(−)-3-hydroxybutyric acid (European Patent Application Publication No. EP133988). Sustained release compositions may also include liposomes that can be prepared by any of several methods known in the art. See, e.g., Eppstein et al., 1985, Proc. Natl. Acad. Sci. U.S.A. 82:3688-3692; European Patent Application Publication Nos. EP036676; EP088046 and EP143949, incorporated by reference. Pharmaceutical compositions used for in vivo administration are typically provided as sterile preparations. Sterilization can be accomplished by filtration through sterile filtration membranes. When the composition is lyophilized, sterilization using this method may be conducted either prior to or following lyophilization and reconstitution. Compositions for parenteral administration can be stored in lyophilized form or in a solution. Parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. Free amino acids can be used in antibody or fragment formulations in accordance with various embodiments of the invention as bulking agents, stabilizers, and antioxidants, as well as other standard uses. Lysine, proline, serine, and alanine can be used for stabilizing proteins in a formulation. Glycine is useful in lyophilization to ensure correct cake structure and properties. Arginine may be useful to inhibit protein aggregation, in both liquid and lyophilized formulations. Methionine is useful as an antioxidant. Embodiments of antibody formulations may further comprise one or more antioxidants. To some extent deleterious oxidation of proteins can be prevented in pharmaceutical formulations by maintaining proper levels of ambient oxygen and temperature and by avoiding exposure to light. Antioxidant excipients can be used as well to prevent oxidative degradation of proteins. Among useful antioxidants in this regard are reducing agents, oxygen/free-radical scavengers, and chelating agents. Antioxidants for use in therapeutic protein formulations in accordance with the invention preferably are water-soluble and maintain their activity throughout the shelf life of a product. EDTA is a preferred antioxidant in accordance with the invention in this regard. Formulations in accordance with the invention may include metal ions that are protein co-factors and that are necessary to form protein coordination complexes, such as zinc necessary to form certain insulin suspensions. Metal ions also can inhibit some processes that degrade proteins. However, metal ions also catalyze physical and chemical processes that degrade proteins. Magnesium ions (10-120 mM) can be used to inhibit isomerization of aspartic acid to isoaspartic acid. Ca+2ions (up to 100 mM) can increase the stability of human deoxyribonuclease. Mg+2, Mn+2, and Zn+2, however, can destabilize rhDNase. Similarly, Ca+2and Sr+2can stabilize Factor VIII, it can be destabilized by Mg+2, Mn+2and Zn+2, Cu+2and Fe+2, and its aggregation can be increased by Al+3ions. Embodiments of the antibody formulations can further comprise one or more preservatives. Once the pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, crystal, or as a dehydrated or lyophilized powder. Such formulations may be stored either in a ready-to-use form or in a form (e.g., lyophilized) that is reconstituted prior to administration. The invention also provides kits for producing a single-dose administration unit. The kits of the invention may each contain both a first container having a dried protein and a second container having an aqueous formulation. In certain embodiments of this invention, kits containing single and multi-chambered pre-filled syringes (e.g., liquid syringes and lyosyringes) are provided. The therapeutically effective amount of an antibody-containing pharmaceutical composition to be employed will depend, for example, upon the therapeutic context and objectives. One skilled in the art will appreciate that the appropriate dosage levels for treatment will vary depending, in part, upon the molecule delivered, the indication(s) for which the antibody is being used, the route of administration, and the size (body weight, body surface or organ size) and/or condition (the age and general health) of the patient. Stability The terms “stability” and “stable” as used herein in the context of a composition comprising an antibody (or antigen binding fragment thereof) refer to the resistance of the antibody (or antigen binding fragment thereof) in the composition to aggregation, degradation or fragmentation under given manufacture, preparation, transportation and/or storage conditions. Antibody formulations comprising a high degree of stability demonstrate enhanced reliability and safety and, as such, are advantageous for clinical use. Antibody stability in a composition is optionally assessed by examining a desired parameter of the antibody in the composition (e.g., aggregation, degradation of heavy and/or light chains, chemical modification, etc.) over time. In this regard, a parameter is typically examined at an initial time point (T0) and an assessment time point (T1), optionally while exposing the antibody to any of a number of environmental conditions, and compared. An initial time point can be, for instance, the time that the antibody is first formulated in a composition or first examined for quality (i.e., examined to determine whether the antibody composition meets regulatory or manufacturing specifications with respect to aggregation or degradation). An initial time point also can be the time at which the antibody is reformulated in a composition (e.g., reformulated at a higher or lower concentration compared to an initial preparation). An assessment time point is, in various embodiments, about 1 week (or about 2 weeks, or about 3 weeks, or about 4 weeks, or about 5 weeks, or about 6 weeks, or about 7 weeks, or about 8 weeks, or about 10 weeks, or about 3 months, or about 6 months or about 1 year) after the initial time point. The desired parameter (e.g., aggregation or degradation) of the antibody or fragment thereof in the composition can be assessed under a variety of storage conditions, such as temperatures of −30° C., 4° C., 20° C. or 40° C., shaking, pH, storage in different container materials (e.g., glass vials, pre-filled syringes, etc.), and the like. Exemplary methods for determining the degree of aggregation, and/or types and/or sizes of aggregates present in a composition comprising the antibody include, but are not limited to, size exclusion chromatography (SEC), high performance size exclusion chromatography (HPSEC), static light scattering (SLS), Fourier Transform Infrared Spectroscopy (FTIR), circular dichroism (CD), urea-induced protein unfolding techniques, intrinsic tryptophan fluorescence, differential scanning calorimetry, and 1-anilino-8-naphthalenesulfonic acid (ANS) protein binding techniques. Size exclusion chromatography (SEC) may be performed to separate molecules on the basis of their size, by passing the molecules over a column packed with the appropriate resin, the larger molecules (e.g. aggregates) will elute before smaller molecules (e.g. monomers). The molecules are generally detected by UV absorbance at 280 nm and may be collected for further characterization. High pressure liquid chromatographic columns are often utilized for SEC analysis (HP-SEC). Alternatively, analytical ultracentrifugation (AUC) may be utilized. AUC is an orthogonal technique which determines the sedimentation coefficients of macromolecules in a liquid sample. Like SEC, AUC is capable of separating and detecting antibody fragments/aggregates from monomers and is further able to provide information on molecular mass. Antibody aggregation in a composition may also be characterized by particle counter analysis using a coulter counter or by turbidity measurements using a turbidimeter. Turbidity is a measure of the amount by which the particles in a solution scatter light and, thus, may be used as a general indicator of protein aggregation. In addition, non-reducing polyacrylamide gel electrophoresis (PAGE) or capillary gel electrophoresis (CGE) may be used to characterize the aggregation and/or fragmentation state of antibodies or antibody fragments in a composition. Exemplary methods for determining antibody degradation include, but are not limited to, size-exclusion chromatography (SEC), sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and capillary electrophoresis with SDS (CE-SDS) and reversed phase HPLC with in-line MS detection. In various embodiments, less than 5% of the antibody described herein in the composition is in aggregate form under conditions of interest. For instance, less than 4%, or less than 3%, or less than 2%, or less than 1% of the antibody in the composition is in aggregate form after storage at −30° C., 4° C., 20° C. or 40° C. for a period of about 1 week (or about 2 weeks, or about 3 weeks, or about 4 weeks, or about 5 weeks, or about 6 weeks, or about 7 weeks, or about 8 weeks, or about 10 weeks, or about 3 months, or about 6 months or about 1 year). In some embodiments, less than 5% (or less than 4% or less than 3% or less than 2% or less than 1% or less) of the antibody described herein in the composition is in aggregate form after storage for two weeks at about 4° C. For example at least 85% (or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99%) of antibody in a composition optionally is present in non-aggregate (i.e., monomeric) form after storage at −30° C., 4° C., 20° C. or 40° C. for a period of about 1 week (or about 2 weeks, or about 3 weeks, or about 4 weeks, or about 5 weeks, or about 6 weeks, or about 7 weeks, or about 8 weeks, or about 10 weeks, or about 3 months, or about 6 months or about 1 year). In some embodiments, at least 85% (or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99% or more) of the antibody is present in the composition in non-aggregate form after two weeks of storage at about 4° C. In some embodiments, at least 99% of the antibody is present in the composition in non-aggregate form after storage for two weeks at about 4° C. for two weeks and/or at least 95% of antibody present in the composition is in non-aggregate form after storage for two weeks at 40° C. In various embodiments, less than 5% of the antibody described herein in the composition is degraded. For instance, less than 4%, or less than 3%, or less than 2%, or less than 1% or less of the antibody in the composition is degraded under conditions of interest. For example, optionally at least 85% (or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99%) of the antibody is intact (i.e., not degraded) in a composition stored at about −30° C., about 4° C., about 20° C. or about 40° C. for a period of about 1 week (or about 2 weeks, or about 3 weeks, or about 4 weeks, or about 5 weeks, or about 6 weeks, or about 7 weeks, or about 8 weeks, or about 10 weeks, or about 3 months, or about 6 months or about 1 year). In some aspects, at least 85% (or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99% or more) of the antibody is intact (i.e., non-degraded) after storage in a composition at about 4° C. for a period of two weeks. In some embodiments, at least 99% of the antibody remains intact when stored in a composition at about 4° C. for two weeks and/or at least 95% remains intact when stored in a composition at about 40° C. for two weeks. Functional or activity stability of the antibody in a composition also is contemplated herein. Assays for detecting and/or quantifying, e.g., antibody binding to a target or sclerostin neutralization are known in the art. Optionally, the antibody demonstrates about 50-100% activity under conditions of interest compared to the activity of the antibody at the initial time point. For example, the antibody retains a level of activity of between about 60-90% or 70-80% compared to the activity the initial time point. Accordingly, functional stability of the antibody includes retention of activity of at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% and can include activity measurements greater than 100% such as 105%, 110%, 115%, 120%, 125% or 150% or more compared to the activity at the initial time point. Viscosity In some embodiments, the viscosity of a composition comprising one or more of the antibodies described herein is determined. The term “viscosity” as used herein refers to “absolute viscosity.” Absolute viscosity, sometimes called dynamic or simple viscosity, is the product of kinematic viscosity and fluid density (Absolute Viscosity=Kinematic Viscosity×Density). The dimension of kinematic viscosity is L2/T where L is a length and T is a time. Commonly, kinematic viscosity is expressed in centistokes (cSt). The SI unit of kinematic viscosity is mm 2/s, which is 1 cSt. Absolute viscosity is expressed in units of centipoise (cP). The SI unit of absolute viscosity is the millipascal-second (mPa-s), where 1 cP=1 mPa-s. The viscosity of a composition can be measured hours (e.g., 1-23 hours), days (e.g., 1-10 days), weeks (e.g., 1-5 weeks), months (e.g., 1-12 months), or years (e.g., 1-2 years, 1-3 years) after the addition of the antibody to the composition. Viscosity measurements may be made at a storage or administration temperature, e.g. 2-8° C. or 25° C. (room temperature). In some embodiments, absolute viscosity of the liquid or reconstituted liquid composition at the storage and/or administration temperature is 15 cP or less, or 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, or 4 cP or less. In some embodiments, absolute viscosity of the liquid or reconstituted liquid composition is 6 cP or less. In some embodiments, the viscosity of the antibody composition is measured prior to and after the addition of antibody. Methods of measuring viscosity are well known in the art and include, for example, using a capillary viscometer, or a cone-plate rheometer. Any method may be used provided the same method is used to compare the test and reference formulations. Therapeutic Methods The antibody and pharmaceutical compositions described herein are useful for treating or preventing bone-related disorders, such as bone-related disorders associated with abnormal osteoblast or osteoclast activity. In some embodiments, the antibody is administered to a subject suffering from a bone related disorder selected from the group consisting of achondroplasia, cleidocranial dysostosis, enchondromatosis, fibrous dysplasia, Gaucher's Disease, hypophosphatemic rickets, Marfan's syndrome, multiple hereditary exotoses, neurofibromatosis, osteogenesis imperfecta, osteopetrosis, osteopoikilosis, sclerotic lesions, pseudoarthrosis, pyogenic osteomyelitis, periodontal disease, anti-epileptic drug induced bone loss, primary and secondary hyperparathyroidism, familial hyperparathyroidism syndromes, weightlessness induced bone loss, osteoporosis in men, postmenopausal bone loss, osteoarthritis, renal osteodystrophy, infiltrative disorders of bone, oral bone loss, osteonecrosis of the jaw, juvenile Paget's disease, melorheostosis, metabolic bone diseases, mastocytosis, sickle cell anemia/disease, organ transplant related bone loss, kidney transplant related bone loss, systemic lupus erythematosus, ankylosing spondylitis, epilepsy, juvenile arthritides, thalassemia, mucopolysaccharidoses, Fabry Disease, Turner Syndrome, Down Syndrome, Klinefelter Syndrome, leprosy, Perthe's Disease, adolescent idiopathic scoliosis, infantile onset multi-system inflammatory disease, Winchester Syndrome, Menkes Disease, Wilson's Disease, ischemic bone disease (such as Legg-Calve-Perthes disease and regional migratory osteoporosis), anemic states, conditions caused by steroids, glucocorticoid-induced bone loss, heparin-induced bone loss, bone marrow disorders, scurvy, malnutrition, calcium deficiency, osteoporosis, osteopenia, alcoholism, chronic liver disease, postmenopausal state, chronic inflammatory conditions, rheumatoid arthritis, inflammatory bowel disease, ulcerative colitis, inflammatory colitis, Crohn's disease, oligomenorrhea, amenorrhea, pregnancy-related bone loss, diabetes mellitus, hyperthyroidism, thyroid disorders, parathyroid disorders, Cushing's disease, acromegaly, hypogonadism, immobilization or disuse, reflex sympathetic dystrophy syndrome, regional osteoporosis, osteomalacia, bone loss associated with joint replacement, HIV associated bone loss, bone loss associated with loss of growth hormone, bone loss associated with cystic fibrosis, chemotherapy-associated bone loss, tumor-induced bone loss, cancer-related bone loss, hormone ablative bone loss, multiple myeloma, drug-induced bone loss, anorexia nervosa, disease-associated facial bone loss, disease-associated cranial bone loss, disease-associated bone loss of the jaw, disease-associated bone loss of the skull, bone loss associated with aging, facial bone loss associated with aging, cranial bone loss associated with aging, jaw bone loss associated with aging, skull bone loss associated with aging, and bone loss associated with space travel. In some embodiments, the antibodies described herein are useful for improving outcomes in orthopedic procedures, dental procedures, implant surgery, joint replacement, bone grafting, bone cosmetic surgery and bone repair such as fracture healing, nonunion healing, delayed union healing and facial reconstruction. A composition comprising one or more antibodies may be administered before, during and/or after the procedure, replacement, graft, surgery or repair. In some embodiments, the antibodies described herein are useful for the treatment of any fracture comprising a gap between two segments of bone (e.g., a gap of at least about 1 mm between two segments of bone). In some or any embodiments, the gap is at least about 2 mm, at least about 3 mm, at least about 4 mm, at least about 5 mm, at least about 6 mm, at least about 7 mm, at least about 8 mm, at least about 9 mm, or at least about 1 cm or more. In some or any embodiments, the gap is about 5 mm to 1 cm, or up to 1 cm. The terms “bone gap defect” and “segmental skeletal defect” are used synonymously herein and refer to a gap between two segments of bone (e.g., a gap of at least 1 mm). Exemplary bone gap defects include, but are not limited to, a comminuted fracture, a non-union fracture, a segmental skeletal defect, surgically created bone defects, surgically treated bone defects, and bone defects created from traumatic injury to the bone or disease (including, but not limited to, arthritis, tumor removal (resection) or infection removal). In some or any embodiments, the bone gap defect is produced by removal of infected sections of bone or the removal of cancer from the bone due to bone cancers including, but not limited to, osteosarcoma, Ewing's sarcoma, chondrosarcoma, malignant fibrous histiocytoma, fibrosarcoma, and chordoma. In some or any embodiments, the bone gap defect is a developmental deformity, e.g., due to a genetic defect. In some or any embodiments, the bone gap defect is produced by removal of sections of bone containing a benign tumor. Exemplary benign bone tumors include, but are not limited to, osteoma, osteoid osteoma, osteoblastoma, osteochondroma, enchondroma, chonrdomyxoid fibroma, aneurysmal bone cyst, unicameral bone cyst, fibrous dysplasia of bone and giant cell tumor of the bone. Administration of the antibody enhances or accelerates bone gap defect healing, thereby “treating” the bone gap defect. “Enhancing” bone healing means mediating a level of bone healing beyond (i.e., greater than) the level of bone healing experienced in subjects (e.g., mammals, such as humans) not administered the sclerostin inhibitor (i.e., control subjects). Bone healing is evidenced by, for example, bridging status, improved bone volume, improved bone mineral content and density within the fracture gap (i.e., formation of bridging bone), mature bone callus, improved bone strength (optionally accompanied by a medically-acceptable level of bone stiffness), or improved patient use of the affected area. By “improved” is meant an increase or decrease (as desired) in the measured parameter. The increase can be a return, in whole or in part, of the measured parameter to baseline level (e.g., the level prior to the bone gap defect), to values provided in normative databases used in the art, or to the contralateral functional level (e.g., return, in whole or in part, to the functional capabilities of, for example, the contralateral limb). In some cases, the increase can be an improvement beyond baseline level. If desired, the measured parameters in patients administered one or more doses of the antibody can be compared to the same parameters in fracture patients (optionally age and gender matched) not administered the antibody to further analyze the efficacy of the methods described herein. Formation of bridging bone, bone mineral content and bone density, and/or mature boney callus at the site of bone defect may be measured using radiography (e.g., radiographic absorptometry), single- and/or dual-energy X-ray absorptometry, quantitative computed tomography (QCT), ultrasonography, radiography (e.g., radiographic absorptometry), and magnetic resonance imaging. In some embodiments, the antibody may be administered at a dose and for a time period effective to increase bridging bone formation, formation of bony callus, or bone density (or volume) at the defect site by at least about 5% (about 6%, about 7%, about 8%, or about 9%). In some embodiments, bridging bone formation, formation of bony callus, or bone density at the defect site is increased by at least about 10% (e.g., at least about 10%, at least about 12%, at least about 15%, at least about 18%, at least about 20%, or at least about 22%). In other embodiments, bridging bone formation, formation of bony callus, or bone density at the defect site is increased by the sclerostin inhibitor at least about 25% (e.g., at least about 26% or at least about 28%). In yet other embodiments, bridging bone formation, formation of bony callus, or bone density at the defect site is increased at least about 30% (e.g., at least about 32%, at least about 35%, at least about 38%, or at least about 40%) or at least about 50% (e.g., at least about 60%, at least about 70%, at least about 80%, at least about 90%, or about 100%). The increase or re-establishment of bridging bone formation can be determined at 1 week, 2 weeks, 3 weeks, or 4 weeks following the initial administration of antibody. Alternatively, the bone density level can be determined after the treatment period ends (e.g., 1 week, 2 weeks, 3 weeks, or 4 weeks after the treatment period ends). In one aspect, the method reduces the amount of time required to establish a desired level of bone formation, bone volume, bony callus, or bone density (e.g., any percent increase in bone formation, bone mineral density, bony callus, or bone volume described herein) compared to age and gender-matched patients that do not receive the antibody, thereby reducing recovery time for a subject. For example, in one embodiment, the antibody reduces the amount of time required to increase bone density or volume at the defect site at least about 10% (e.g., at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, or at least about 50%). The antibody need not cure the subject of the disorder or completely protect against the onset of a bone-related disorder to achieve a beneficial biological response. The antibody may be used prophylactically, meaning to protect, in whole or in part, against a bone-related disorder or symptom thereof. The antibody also may be used therapeutically to ameliorate, in whole or in part, a bone-related disorder or symptom thereof, or to protect, in whole or in part, against further progression of a bone-related disorder or symptom thereof. Indeed, the materials and methods of the invention are particularly useful for increasing bone mineral density, and optionally maintaining the increased bone mineral density over a period of time. In some embodiments, one or more administrations of an antibody described herein are carried out over a therapeutic period of, for example, about 1 week to about 18 months (e.g., about 1 month to about 12 months, about 1 month to about 9 months or about 1 month to about 6 months or about 1 month to about 3 months). In some embodiments, a subject is administered one or more doses of a antibody described herein over a therapeutic period of, for example about 1 month to about 12 months (52 weeks) (e.g., about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, or about 11 months). In addition, it may be advantageous to administer multiple doses of the antibody or space out the administration of doses, depending on the therapeutic regimen selected for a particular subject. In some embodiments, the antibody or fragment thereof is administered periodically over a time period of one year (12 months, 52 weeks) or less (e.g., 9 months or less, 6 months or less, or 3 months or less). In this regard, the antibody or fragment thereof is administered to the human once every about 3 days, or about 7 days, or 2 weeks, or 3 weeks, or 4 weeks, or 5 weeks, or 6 weeks, or 7 weeks, or 8 weeks, or 9 weeks, or 10 weeks, or 11 weeks, or 12 weeks, or 13 weeks, or 14 weeks, or 15 weeks, or 16 weeks, or 17 weeks, or 18 weeks, or 19 weeks, or 20 weeks, or 21 weeks, or 22 weeks, or 23 weeks, or 6 months, or 12 months. In some embodiments, one or more doses of the antibody are administered in an amount and for a time effective to increase bone mineral density or treat a bone disorder associated with decreased bone mineral density. In various embodiments, one or more doses comprising from about 50 milligrams to about 1,000 milligrams of the antibody are administered per week to a subject (e.g., a human subject). For example, a dose of antibody can comprise at least about 5 mg, 15 mg, 25 mg, 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 120 mg, about 150 mg, about 200 mg, about 210 mg, about 240 mg, about 250 mg, about 280 mg, about 300 mg, about 350 mg, about 400 mg, about 420 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg or up to about 1,000 mg of antibody. Ranges between any and all of these endpoints are also contemplated, e.g. about 50 mg to about 80 mg, about 70 mg to about 140 mg, about 70 mg to about 270 mg, about 75 mg to about 100 mg, about 100 mg to about 150 mg, about 140 mg to about 210 mg, or about 150 mg to about 200 mg, or about 180 mg to about 270 mg, or about 280 to about 410 mg. The dose is administered at any interval, such as multiple times a week (e.g., twice or three times per week), once a week, once every two weeks, once every three weeks, or once every four weeks. In some or any embodiments, a dose of antibody ranging from about 120 mg to about 210 mg is administered twice a week. In some or any embodiments, a dose of about 140 mg of the antibody is administered twice a week. In various aspects, a dose of about 210 mg of antibody is administered once a month. In some embodiments, the one or more doses of antibody can comprise between about 0.1 to about 50 milligrams (e.g., between about 5 and about 50 milligrams), or about 1 to about 100 milligrams, of antibody per kilogram of body weight (mg/kg). For example, the dose of antibody may comprise at least about 0.1 mg/kg, 0.5 mg/kg, 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, about 10 mg/kg, about 20 mg/kg, about 25 mg/kg, about 26 mg/kg, about 27 mg/kg, about 28 mg/kg, about 29 mg/kg, about 30 mg/kg, about 31 mg/kg, about 32 mg/kg, about 33 mg/kg, about 34 mg/kg, about 35 mg/kg, about 36 mg/kg, about 37 mg/kg, about 38 mg/kg, about 39 mg/kg, about 40 mg/kg, about 41 mg/kg, about 42 mg/kg, about 43 mg/kg, about 44 mg/kg, about 45 mg/kg, about 46 mg/kg, about 47 mg/kg, about 48 mg/kg, or about 49 mg/kg, or about 50 mg/kg, about 55 mg/kg, about 60 mg/kg, about 65 mg/kg, about 70 mg/kg, about 75 mg/kg, about 80 mg/kg, about 85 mg/kg, about 90 mg/kg, about 95 mg/kg, or up to about 100 mg/kg. Ranges between any and all of these endpoints are also contemplated, e.g., about 1 mg/kg to about 3 mg/kg, about 1 mg/kg to about 5 mg/kg, about 1 mg/kg to about 8 mg/kb, about 3 mg/kg to about 8 mg·kg, about 1 mg/kg to about 10 mg/kg, about 1 mg/kg to about 20 mg/kg, about 1 mg/kg to about 40 mg/kg, about 5 mg/kg to about 30 mg/kg, or about 5 mg/kg to about 20 mg/kg. Monitoring Therapy Antibody-mediated increases in bone mineral content or bone density may be measured using single- and dual-energy X-ray absorptometry, ultrasound, computed tomography, radiography, and magnetic resonance imaging. The amount of bone mass may also be calculated from body weights or by using other methods (see Guinness-Hey, Metab. Bone Dis. Relat. Res., 5:177-181 (1984)). Animal models are used in the art for testing the effect of the pharmaceutical compositions and methods on, for example, parameters of bone loss, bone resorption, bone formation, bone strength, or bone mineralization that mimic conditions of human disease such as osteoporosis and osteopenia. Examples of such models include the ovariectomized rat model (Kalu, Bone and Mineral, 15:175-192 (1991); Frost and Jee, Bone and Mineral, 18:227-236 (1992); and Jee and Yao, J. Musculoskel. Neuron. Interact., 1:193-207 (2001)). The methods for measuring antibody activity described herein also may be used to determine the efficacy of other sclerostin inhibitors. In humans, bone mineral density can be determined clinically using dual x-ray absorptiometry (DXA) of, for example, the hip and spine. Other techniques include quantitative computed tomography (QCT), ultrasonography, single-energy x-ray absorptiometry (SXA), and radiographic absorptiometry. Common central skeletal sites for measurement include the spine and hip; peripheral sites include the forearm, finger, wrist and heel. Except for ultrasonography, the American Medical Association notes that BMD techniques typically involve the use of x-rays and are based on the principle that attenuation of the radiation depends on thickness and composition of the tissues in the radiation path. All techniques involve the comparison of results to a normative database. Alternatively, a physiological response to one or more anti-sclerostin antibodies can be gauged by monitoring bone marker levels. Bone markers are products created during the bone remodeling process and are released by bone, osteoblasts, and/or osteoclasts. Fluctuations in bone resorption and/or bone formation “marker” levels imply changes in bone remodeling/modeling. The International Osteoporosis Foundation (IOF) recommends using bone markers to monitor bone density therapies (see, e.g., Delmas et al., Osteoporos Int., Suppl. 6:S2-17 (2000), incorporated herein by reference). Markers indicative of bone resorption (or osteoclast activity) include, for example, C-telopeptide (e.g., C-terminal telopeptide of type 1 collagen (CTX) or serum cross-linked C-telopeptide), N-telopeptide (N-terminal telopeptide of type 1 collagen (NTX)), deoxypyridinoline (DPD), pyridinoline, urinary hydroxyproline, galactosyl hydroxylysine, and tartrate-resistant acid phosphatase (e.g., serum tartrate-resistant acid phosphatase isoform 5b). Bone formation/mineralization markers include, but are not limited to, bone-specific alkaline phosphatase (BSAP), peptides released from N- and C-terminal extension of type I procollagen (P1NP, PICP), and osteocalcin (OstCa). Several kits are commercially-available to detect and quantify markers in clinical samples, such as urine and blood. Combination Therapy Treatment of a pathology by combining two or more agents that target the same pathogen or biochemical pathway or biological process sometimes results in greater efficacy and diminished side effects relative to the use of a therapeutically relevant dose of each agent alone. In some cases, the efficacy of the drug combination is additive (the efficacy of the combination is approximately equal to the sum of the effects of each drug alone), but in other cases the effect is synergistic (the efficacy of the combination is greater than the sum of the effects of each drug given alone). As used herein, the term “combination therapy” means that two or more agents are delivered in a simultaneous manner, e.g., concurrently, or wherein one of the agents is administered first, followed by the second agent, e.g., sequentially. In some embodiments, the antibody is administered along with a standard of care therapeutic for the treatment of decreased bone mineral density (i.e., the antibody and standard of care therapeutic are part of the same treatment plan). As used herein, the term “standard of care” refers to a treatment that is generally accepted by clinicians for a certain type of patient diagnosed with a type of illness. In some embodiments, the antibody is administered along with a second bone-enhancing agent useful for the treatment of decreased bone mineral density or bone defect. In some embodiments, the bone-enhancing agent is selected from the group consisting of an anti-resorptive agent, a bone-forming agent (i.e., anabolic), an estrogen receptor modulator (including, but not limited to, raloxifene, bazedoxifene and lasofoxifene) and a drug that has an inhibitory effect on osteoclasts. In some embodiments, the second bone-enhancing agent is selected from the group consisting of a bisphosphonate (including, but not limited to, alendronate sodium (FOSAMAX®), risedronate, ibandronate sodium (BONIVA®) and zoledronic acid (RECLAST®)); an estrogen or estrogen analogue; an anti-RANK ligand (RANKL) inhibitor, such as an anti-RANKL antibody (e.g., denosumab, PROLIA®); vitamin D, or a vitamin D derivative or mimic thereof; a calcium source, a cathepsin-K (cat-K) inhibitor (e.g. odanacatib), Tibolone, calcitonin or a calcitriol; and hormone replacement therapy. In some embodiments, the second bone-enhancing agent includes, but is not limited to, parathyroid hormone (PTH) or a peptide fragment thereof, PTH-related protein (PTHrp), bone morphogenetic protein, osteogenin, NaF, a PGE2 agonist, a statin, strontium ranelate, and a sclerostin inhibitor (e.g., an anti-sclerostin antibody described in, for example, U.S. Pat. Nos. 7,592,429 or 7,872,106). In some embodiments, the second bone-enhancing agent is Forteo® (Teriparatide), Preotact®, or Protelos®. In some embodiments, the second bone-enhaiving agent comprises a bone morphogenetic protein (e.g., BMP-1, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8, BMP-9, BMP-10, BMP-11, BMP-12, BMP-13, BMP-14 and/or BMP-15). In some embodiments, the combination therapy employing an antibody described herein may precede or follow administration of additional therapeutic(s) (e.g., second bone-enhancing agent) by intervals ranging from minutes to weeks to months. For example, separate modalities are administered within about 24 hours of each other, e.g., within about 6-12 hours of each other, or within about 1-2 hours of each other, or within about 10-30 minutes of each other. In some situations, it may be desirable to extend the time period for treatment significantly, where several days (2, 3, 4, 5, 6 or 7 days) to several weeks (1, 2, 3, 4, 5, 6, 7 or 8 weeks) lapse between the respective administrations of different modalities. Repeated treatments with one or both agents/therapies of the combination therapy is specifically contemplated. Maintenance Therapeutic Regimen Also contemplated is the use of a second bone-enhancing agent and/or antibody described herein in a maintenance regimen to, e.g., prevent or slow the loss of bone mineral density. In this regard, a method or use described herein optionally comprises administering one or more amounts of a second bone-enhancing agent effective to maintain bone mineral density for a maintenance period of about 1 week to about 5 years after the treatment period with the antibody has ended. For example, in some embodiments, a method or use described herein comprises the administration of a second bone-enhancing agent to the subject for a maintenance period of about at least about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 12 weeks, about 3 months, about 13 weeks, about 14 weeks, about 15 weeks, about 16 weeks, about 4 months, about 17 weeks, about 18 weeks, about 19 weeks, about 20 weeks, about 5 months, about 21 weeks, about 22 weeks, about 23 weeks, about 24 weeks, about 6 months, about 25 weeks, about 26 weeks, about 27 weeks, about 28 weeks, about 7 months, about 29 weeks, about 30 weeks, about 31 weeks or longer (e.g., about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 15 months, about 18 months, about 2 years, about 3 years, about 4 years, about 5 years or longer (e.g., over the lifetime of the subject). In some embodiments, the maintenance period is about 6-12 weeks. In some embodiments, the maintenance period is about 4-12 weeks, or about 1-3 months. In some embodiments, the maintenance period is about 12-20 weeks, or about 3-5 months. In some embodiments, the maintenance period is about 20-32 weeks, or about 5-8 months. In some embodiments, the maintenance period is about 24-36 weeks, or about 6-9 months. In some embodiments, the maintenance period is about 1 year, about 2 years, about 3 years, about 4 years, about 5 years or longer. “Maintaining” bone mineral density includes maintaining similar levels of bone mineral density parameters experienced in the subject that received the antibody treatment. Kits A pharmaceutical composition comprising one or more antibodies described herein may be placed within containers (e.g., vials or syringes), along with packaging material that provides instructions regarding the use of such pharmaceutical compositions. Generally, such instructions will include a tangible expression describing the antibody concentration, as well as within certain embodiments, relative amounts of excipient ingredients or diluents (e.g., water, saline or PBS) that may be necessary to reconstitute the pharmaceutical composition. EXAMPLES Example 1—Analysis of Romosozumab PARG (SEQ ID NO: 8) C-Terminal Variant Wild-type romosozumab and a romosozumab PARG (SEQ ID NO: 8) C-terminal variant were digested by Lys-C and analyzed by LC/MS peptide mapping. The UV profiles of these two constructs were compared side by side (FIG.1). It was determined that wild-type romosozumab and the romosozumab PARG (SEQ ID NO: 8) C-terminal variant have a similar peak eluting at 37.7 minutes but wild-type romosozumab was determined to have a mass of 659.3 Da and the romosozumab PARG (SEQ ID NO: 8) C-terminal variant was determined to have a mass of 886.7 Da. The majority of lysine (K) variants of romosozumab (PGK) was thought to be removed from the process. The presence of a significant amount of the amidated form of the romosozumab PARG (SEQ ID NO: 8) C-terminal variant (828.6 Da peak) confirms that the amidation efficiency is sequence dependent when compared to the wild-type romosozumab PG sequence. Next, the PARG (SEQ ID NO: 8) C-terminal variant was then treated with carboxypeptidase (CP-B), analyzed by CEX-HPLC method and compared with the PARG (SEQ ID NO: 8) C-terminal variant control which was not treated by the CP-B. There is a significant shift post treatment for the peaks eluting at 17.5 mins and 21 mins, but not for the peak at 24 mins (FIG.2). It is contemplated that the 24 min peak is the doubly amidated form, which is protected from proteolytic degradation. Example 2—C-Terminal Variant Enrichment Purification or enrichment for different romosozumab species from a composition comprising wild-type romosozumab and the romosozumab PARG LSEQ ID NO: 8) C-terminal variant is achieved by Cation Exchange Chromatography (CEX) fractionation. CEX separates proteins based on differences in their surface charges. At a set pH, positively charged variants of wild-type romosozumab are separated on a cation-exchange column (e.g., Dionex Pro Pac WCX-10 analytical column, 2.0 mm×250 mm) and eluted using a salt gradient (e.g., Mobile Phase A: 10:90 (v/v) ACN, 19 mM MES pH 6.2; Mobile Phase B: 10:90 (v/v) ACN, 19 mM MES, 250 mM NaCl, pH 6.2). The different C-terminal variants of romosozumab are charged differently and the more positively charged variant elutes later in CEX. Thus, the elution order is: PG (wild-type), P-amide (amidated proline of wild-type), PARG (SEQ ID NO: 8) variant, and PAR-amide. The fraction collector can be programmed to collect CEX eluents containing different variants at different elution times. Example 3—Analysis of Romosozumab PARG (SEQ ID NO: 8) C-Terminal Variant Aggregation Without being bound to any particular theory, it is contemplated that because the PARG (SEQ ID NO: 8) C-terminal variant is highly charged, such forms would repel non-amidated forms in the compositions, thus reducing aggregation in the composition. Romosozumab PARG (SEQ ID NO: 8) C-terminal variant protein A pool was analyzed side by side with wild-type romosozumab protein A pool using SEC-HPLC, a size exclusion HPLC method that separates protein based on differences in their hydrodynamic volume (Table 1). TABLE 1Molecule% HMWAMG785 ARG ProA pool3.4%AMG785 WT ProA pool7.2% The data demonstrated that romosozumab PARG (SEQ ID NO: 8) C-terminal variant has less high molecular weight species as compared to the wild-type romosozumab. Example 4—Viscosity Analysis of Romosozumab PARG (SEQ ID NO: 8) C-Terminal Variant Antibody solutions containing romosozumab PARG (SEQ ID NO: 8) C-terminal variant or wild-type romosozumab are measured using a cone and plate. The solutions are concentrated up to 120 mg/mL according to approximate volume depletion, and final concentrations are determined (±10%) using the proteins absorbance at 280 nm (after dilution to end up within 0.1-1 absorbance units (AU)) and a protein specific extinction coefficient. Viscosity analysis is performed on a Brookfield LV-DVIII cone and plate instrument (Brookfield Engineering, Middleboro, MA, USA) using a CP-40 spindle and sample cup or an ARES-G2 rheometer (TA Instruments, New Castle, DE, USA) using a TA Smart Swap 2 degree cone/plate spindle. All measurements are performed at 25° C. and controlled by a water bath attached to the sample cup. Multiple viscosity measurements were collected, manually within a defined torque range (10-90%) by increasing the RPM of the spindle. Measurements are averaged in order to report one viscosity value per sample to simplify the resulting comparison chart. Example 5—Solubility Analysis of Romosozumb PARG (SEQ ID NO: 8) C-Terminal Variant To determine the impact of the amino acid variation of romosozumab PARG (SEQ ID NO: 8) variant as compared to the wild type romosozumab on solubility upon subcutaneous (SC) injection, a dialysis solubility assay was performed on both wild type and PARG (SEQ ID NO: 8) C-terminal variant romosozumab in parallel. This screen entails dialyzing a sample of the romosozumab PARG (SEQ ID NO: 8) C-terminal variant and a sample of the wild-type romosozumab into a solution that simulates the pH and ionic strength of the SC space and monitoring the solubility and physical stability of the antibody in these conditions over a short time period. Samples were formulated at ˜63 mg/mL in formulation buffer (pH 5.2). Then each sample was injected into a dialysis cassette and dialyzed into a PBS buffer to mimic the SC space. Visual observations were made 24 hours after initial dialysis. Wild-type romosozumab typically shows precipitation after 24 hours. The results show that both molecules precipitate in this analysis but the PARG (SEQ ID NO: 8) C-terminal variant precipitates less and at a slower rate. This suggests that the variant is more resistant to precipitation than wild type, although the variant does not abolish precipitation completely. Example 6—Diffusion Analysis of Romosozumab PARG (SEQ ID NO: 8) C-Terminal Variant To determine the impact of the amino acid variation of romosozumab PARG (SEQ ID NO: 8) C-terminal variant as compared to the wild type romosozumab on diffusion from the subcutaneous (SC) space, an assay was performed using Scissor (Pion Inc., Billerica, MA). This assay entails injecting the samples (the romosozumab PARG (SEQ ID NO: 8) C-terminal variant or wild-type romosozumab) at ˜70 mg/mL into a simulated SC space comprised of a collagen and hyaluronic acid matrix. The antibody is able to diffuse out of this matrix through a dialysis membrane into a reservoir of carbonate buffer at pH 7.4. Time points were collected for up to 3 days and each time point was assayed for protein concentration by RP-HPLC. The protein concentration vs. time curves generated simulate the diffusion rates from the SC space. In addition, precipitation in the SC matrix is monitored with visual inspection. Both the wild type and PARG (SEQ ID NO: 8) C-terminal variant romosozumab were tested in the Scissor as described above. The results shown inFIG.3indicate that wild type romosozumab diffuses from the simulated SC space at a much lower rate and more wild type romosozumab is retained at the simulated injection site than PARG (SEQ ID NO: 8) C-terminal variant romosozumab. Example 7—FcRn Binding FcRn, the neonatal Fc receptor, is an MHC class I-like heterodimer composed of a transmembrane a chain (homologous to MHC class-I like molecules) and a (32 microglobulin light chain. FcRn binds to the interface between CH2 and CH3 domains of IgG heavy chains in the Fc region of the IgG molecule under mildly acidic conditions (˜pH 6) and releases it at neutral pH (˜7.4). By this highly pH-dependent interaction, FcRn mediates IgG homeostasis in human adults by maintaining serum IgG levels. A competitive binding assay, the AlphaScreen® binding assay (PerkinElmer, San Jose, CA), was used to assess the binding of the Fc domain of wild-type romosozumab and romosozumab PARG (SEQ ID NO: 8) C-terminal variant to FcRn. The assay is a bead based amplified luminescent proximity homogeneous assay (“Alpha”) that detects bimolecular interactions. The assay contains two bead types, an acceptor bead and a donor bead. The acceptor beads are coated with a hydrogel that contains thioxene derivatives, as well as nickel chelate which binds to the histidine domain of histidine labeled FcRn (FcRn-His). The donor beads are coated with a hydrogel that contains phthalocyanine, a photosensitizer, and streptavidin, which binds to biotinylated CHO derived human Fc. When FcRn-His and the biotinylated human Fc bind together, they bring the acceptor and donor beads into close proximity. When laser light is applied to this complex, ambient oxygen is converted to singlet oxygen by the donor bead. If the beads are in close proximity, an energy transfer to the acceptor bead occurs, resulting in light production (luminescence), which is measured in a plate reader equipped for AlphaScreen® signal detection. When an antibody is present at sufficient concentrations to inhibit the binding of FcRn-His to the biotinylated human Fc domain, a dose dependent decrease in emission at 570 nm is observed. The test sample binding relative to the antibody reference standard is determined and reported as % relative binding and can be used to demonstrate the integrity of the Fc domain of the antibody. It is contemplated that compositions having the PARG (SEQ ID NO: 8) C-terminal variant will have a similar or better dose response curve than the wild type antibody. The results are shown inFIG.4. It was observed that both wild type romosozumab and PARG (SEQ ID NO: 8) C-terminal variant romosozumab bound FcRn similarly and FcRn binding was not affected by the PARG (SEQ ID NO: 8) mutation. Example 8—FcγRIIa Binding FcγRIIa is an activating Fc receptor expressed on monocytes, certain dendritic cells, neutrophils, B cells, platelets and NK cells. FcγRIIa (CD32a) is the most widely distributed FcγR with two extracellular Ig-like domains and low binding affinity for monomeric IgG. There are two common allelic variants in humans that are known to exist for FcγRIIa, expressing either histidine or arginine at position 131 (131H and 131R, respectively). A competitive binding assay was developed to assess the binding of wild-type romosozumab and romosozumab PARG (SEQ ID NO: 8)_C-terminal variant to FcγRIIa (131H). The FcγRIIa (131H) binding assay is a bead-based amplified luminescent proximity homogeneous assay (AlphaScreen® binding assay (PerkinElmer, San Jose, CA) that detects bimolecular interactions. The assay contains 2 bead types, an acceptor bead and a donor bead. The acceptor beads contain the fluorophore europium chelate and are coated with a hydrogel that contains glutathione, which binds recombinant human FcγRIIa (131H)-glutathione-S-transferase (FcγRIIa (131H)-GST). The donor beads are coated with a hydrogel that contains phthalocyanine, a photosensitizer, and streptavidin, which binds to biotinylated human IgG1. When FcγRIIa (131H)-GST and the biotinylated human IgG1 bind together, they bring the acceptor and donor beads into proximity. When a laser is applied to this complex, ambient oxygen is converted to singlet oxygen by the donor bead. When the acceptor and donor beads are near, the singlet oxygen diffuses within the acceptor beads resulting in light production (luminescence), which is measured in a plate reader equipped for luminescence signal detection. When antibody is present at sufficient concentrations to inhibit the binding of FcγRIIa (131H)-GST to the biotinylated human IgG1, a dose-dependent decrease in emission at 570 nm is measured. The test sample binding relative to the antibody reference standard is determined and reported as % relative binding and can be used to demonstrate the integrity of the Fc domain of the antibody. The results are shown inFIG.5. It was observed that the relative binding of PARG (SEQ ID NO: 8) C-terminal variant romosozumab to FcγRIIa (131H) was much higher than wild-type romosozumab. Example 9—Mouse Pharmacokinetic Study To evaluate in vivo drug exposure and bioavailability, a single dose pharmacokinetic study in mice is performed. Romosozumab PARG (SEQ ID NO: 8) C-terminal variant is injected either intravenously (via tail vein) or subcutaneously at a dose of 1 mg/kg. Using nine animals per group, staggered sampling permits collection of data at a large number of time points without exceeding the maximum volume of blood that can be drawn from an individual animal. At each time point, 0.05 ml of blood is drawn. Animals 1 to 3 are sampled at 0.083, 24, 96 and 192 hours post-dose. Animals 4-6 are sampled at 1, 48, 168 and 240 hours. Animals 7-9 are sampled at 6, 72 and 192 hours. Serum is collected from the whole blood sample and test article concentration is determined by a binding immunoassay such as an ELISA (Enzyme-Linked ImmunoSorbant Assay). Changes in test article concentration over time can be used to calculate pharmacokinetic parameters via two compartment analysis. Parameters of interest include, but not limited to, area under the plasma concentration-time curve (AUC), half-life (t1/2) and clearance (CL) for each dose group. Bioavailability can be determined as the ratio of AUC for the subcutaneous dose to the AUC for the intravenous dose.
88,682
11858984
DESCRIPTION OF EMBODIMENTS The present invention relates to an antibody or an antibody fragment thereof having antigen-binding activity for an S100A8/A9 heterodimer. The present invention also relates to a pharmaceutical composition containing the anti-S100A8/A9 antibody or the antibody fragment thereof as an active ingredient. The antibody having antigen-binding activity for an S100A8/A9 heterodimer is hereinafter referred to as “anti-S100A8/A9 antibody”. The present invention relates to an anti-S100A8/A9 antibody or an antibody fragment thereof capable of effectively suppressing cancer metastasis, or effective against an inflammatory disease. The anti-S100A8/A9 antibody or the antibody fragment thereof of the present invention is based on an antibody generated using the S100A8/A9 heterodimer as an antigen, and has antigen-binding activity for the S100A8/A9 heterodimer. More specifically, the anti-S100A8/A9 antibody or the antibody fragment thereof of the present invention is an antibody or an antibody fragment thereof that undergoes an antigen-antibody reaction with the S100A8/A9 heterodimer, or with the S100A8/A9 heterodimer and an S100A8 monomer or an S100A9 monomer. Herein, the term “antibody” is used in its broadest sense, and encompasses monoclonal antibodies, polyclonal antibodies, chimeric antibodies, and multispecific antibodies as long as those antibodies each show antigen-binding activity for the S100A8/A9 heterodimer. Further, the present invention encompasses various antibody structures including antibody fragments thereof. An example of the antibody fragments is an antigen-binding fragment of the antibody. The anti-S100A8/A9 antibody or the antibody fragment thereof of the present invention may contain a heavy chain variable region (VH-CDR) and/or a light chain variable region (VL-CDR), or a fragment thereof. The class of the antibody refers to the type of constant domain or constant region included in a heavy chain (H chain) of the antibody, and examples thereof include IgA, IgD, IgE, IgG, and IgM. Herein, the class of the antibody is not particularly limited, but is most suitably IgG. As subclasses of IgG, there are given, for example, IgG1, IgG2, IgG3, and IgG4, among which IgG1or IgG2is suitable. Examples of the antibody fragment may include Fv, Fab, Fab′, Fab′-SH, F(ab′)2, and combinations thereof. The anti-S100A8/A9 antibody or the antibody fragment thereof of the present invention may be a human antibody or a humanized antibody. The human antibody refers to: an antibody produced by a human or human cells; or an antibody including an amino acid sequence corresponding to the amino acid sequence of an antibody derived from a nonhuman supply source using a human antibody repertoire or other human antibody-coding sequences. The humanized antibody may be a chimeric antibody. The amino acid sequences of VH-CDR and/or VL-CDR contained in the anti-S100A8/A9 antibody or the antibody fragment thereof of the present invention may contain, for example, amino acid sequences identified by the following SEQ ID NOs. For example, a heavy chain variable region 1 (CDR H1) may contain any one amino acid sequence set forth in SEQ ID NO: 7, SEQ ID NO: 10, SEQ ID NO: 13, SEQ ID NO: 16, or SEQ ID NO: 19. A heavy chain variable region 2 (CDR H2) may contain any one amino acid sequence set forth in SEQ ID NO: 8, SEQ ID NO: 11, SEQ ID NO: 14, SEQ ID NO: 17, or SEQ ID NO: 20. A heavy chain variable region 3 (CDR H3) may contain any one amino acid sequence set forth in SEQ ID NO: 9, SEQ ID NO: 12, SEQ ID NO: 15, SEQ ID NO: 18, or SEQ ID NO: 21. For example, a light chain variable region 1 (CDR L1) may contain any one amino acid sequence set forth in SEQ ID NO: 22, SEQ ID NO: 25, SEQ ID NO: 28, SEQ ID NO: 31, or SEQ ID NO: 34. A light chain variable region 2 (CDR L2) region may contain any one amino acid sequence set forth in SEQ ID NO: 23, SEQ ID NO: 26, SEQ ID NO: 29, SEQ ID NO: 32, or SEQ ID NO: 35. A light chain variable region 3 (CDR L3) may contain any one amino acid sequence set forth in SEQ ID NO: 24, SEQ ID NO: 27, SEQ ID NO: 30, SEQ ID NO: 33, or SEQ ID NO: 36. In the present invention, amino acid sequence information on each of the above-mentioned regions is also encompassed in the scope of rights. In addition to the above-mentioned amino acid sequences, even when one or a plurality of amino acids are substituted, deleted, added, or inserted in each of the sequences, anti-S100A8/A9 antibodies or antibody fragments thereof containing such amino acid sequences are also encompassed in the scope of rights of the present invention as long as those antibodies or antibody fragments each show antigen-binding activity for the S100A8/A9 heterodimer. The anti-S100A8/A9 antibody of the present invention may be generated by a method known per se or any method to be developed in the future, through use of the above-mentioned S100A8/A9 heterodimer as an antigen. For example, the anti-S100A8/A9 antibody may be generated by immunizing a mammal, such as a mouse or a rat, with an antigen. The animal may be immunized using, as an immunogen, a mixture of the S100A8/A9 heterodimer antigen and an adjuvant. The adjuvant is not particularly limited, but examples thereof include Freund's complete adjuvant and Freund's incomplete adjuvant. A method of administering the immunogen at the time of the immunization may be any of the methods known per se, such as subcutaneous injection, intraperitoneal injection, intravenous injection, and intramuscular injection. Of those, subcutaneous injection or intraperitoneal injection is preferred. The immunization may be performed once or a plurality of times at an appropriate interval, preferably a plurality of times at an interval of from 1 week to 5 weeks. Through use of the S100A8/A9 heterodimer antigen, a monoclonal antibody may also be generated in accordance with a conventional method. Hybridomas that produce the anti-S100A8/A9 antibody may be obtained by immunizing a mammal, such as a mouse or a rat, with the S100A8/A9 heterodimer antigen, collecting lymphocytes from the animal, and fusing myeloma cells thereto in accordance with a conventional method to generate hybridomas. Cells that produce the monoclonal antibody of interest may be obtained by investigating a binding property to the S100A8/A9 heterodimer by an ELISA method or the like for a culture supernatant or the like of the generated hybridomas, and repeating operation of cloning antibody-producing hybridomas. A method known per se or the like may be applied as a method of generating a humanized antibody. From the antibody-producing hybridoma cells, purification of total RNA and subsequent synthesis of cDNA may be performed in accordance with conventional methods. Through amplification of antibody genes for a full-length heavy chain (H chain) and light chain (L chain) from the resultant cDNA by PCR using respective primers, respective gene fragments may be obtained. Through ligation of the resultant gene fragments to an expression vector, the antibody genes may be cloned. With regard to the amino acid sequences of the H chain and L chain of the antibody, the base sequence of a plasmid vector encoding the amino acid sequences may be identified to determine the amino acid sequence of the antibody. On the basis of the obtained information on the amino acid sequence and the base sequence, the antibody may be generated by a gene recombination technique, or the antibody may be generated by a synthesis method. When the antibody is generated by a gene recombination technique, the antibody may be generated by, for example, a method described in WO 2017/061354 A1. When the antibody is generated by a gene recombination technique, for example, information on genes encoding respective amino acids that identify CDR H1, CDR H2, CDR H3, CDR L1, CDR L2, and CDR L3 may be utilized. As a specific amino acid sequence, for example, for CDR H1, there is given any one amino acid sequence set forth in SEQ ID NO: 7, SEQ ID NO: 10, SEQ ID NO: 13, SEQ ID NO: 16, or SEQ ID NO: 19. For CDR H2, there is given any one amino acid sequence set forth in SEQ ID NO: 8, SEQ ID NO: 11, SEQ ID NO: 14, SEQ ID NO: 17, or SEQ ID NO: 20. For CDR H3, there is given any one amino acid sequence set forth in SEQ ID NO: 9, SEQ ID NO: 12, SEQ ID NO: 15, SEQ ID NO: 18, or SEQ ID NO: 21. For example, for CDR L1, there is given any one amino acid sequence set forth in SEQ ID NO: 22, SEQ ID NO: 25, SEQ ID NO: 28, SEQ ID NO: 31, or SEQ ID NO: 34. For CDR L2, there is given any one amino acid sequence set forth in SEQ ID NO: 23, SEQ ID NO: 26, SEQ ID NO: 29, SEQ ID NO: 32, or SEQ ID NO: 35. For CDR L3, there is given any one amino acid sequence set forth in SEQ ID NO: 24, SEQ ID NO: 27, SEQ ID NO: 30, SEQ ID NO: 33, or SEQ ID NO: 36. The present invention also encompasses base sequence information encoding respective amino acids that identify the above-identified CDR H1, CDR H2, CDR H3, CDR L1, CDR L2, and CDR L3 and base sequence information on strands complementary thereto. In the present invention, in addition to the above-mentioned base sequence information, even when a base sequence has one to a plurality of nucleotides substituted, deleted, added, or inserted, such base acid sequence information is also encompassed in the scope of rights of the present invention as long as the base sequence allows the anti-S100A8/A9 antibody of the present invention to be generated. A screening method for the anti-S100A8/A9 antibody of the present invention and investigation methods for evaluating the antibody are specifically described in, for example, Reference Example, Examples, and experimental examples to be described later, but for example, the following methods may also be applied. Among the above-mentioned antibody-producing hybridomas, hybridomas expressing a plurality of kinds of S100A8/A9 neutralizing antibody candidates may be adapted to serum-free culture and prepared in large amounts for an in vitro or in vivo experiment. A culture supernatant of each clone may be recovered and subjected to the purification of the antibody. Methods known per se or any method to be developed in the future may be applied to the purification of the antibody. For example, the antibody may be recovered by performing affinity chromatography. Specifically, affinity purification using Protein A/G is generally employed, and a column suitable for each animal species or antibody subclass may be used. A purity test for the purified antibody may be performed by a method known per se, and may be performed, for example, by CBB staining. For evaluation of the anti-S100A8/A9 antibody of the present invention, S100A8/A9-binding decoy protein formulations (exEMMPRIN-Fc, exNPTNβ-Fc, exMCAM-Fc, exRAGE-Fc, and exALCAM-Fc) serving as receptors for S100A8/A9 may be appropriately prepared. The present invention relates to a pharmaceutical composition, particularly an anticancer agent and/or an anti-inflammatory agent, containing the anti-S100A8/A9 antibody or the antibody fragment thereof as an active ingredient. The “anticancer agent containing the anti-S100A8/A9 antibody or the antibody fragment thereof as an active ingredient” of the present invention is specifically used as a cancer metastasis suppressor and/or a cancer therapeutic agent. Cancer to be targeted by the anticancer agent of the present invention only needs to be, for example, cancer that may metastasize to a site different from primary cancer, and is not particularly limited, but specific examples thereof include one kind or a plurality of kinds of cancers selected from skin cancer (melanoma), lung cancer, stomach cancer, colon cancer, pancreatic cancer, liver cancer, lung cancer, kidney cancer, breast cancer, uterine cancer, bile duct cancer, esophageal cancer, pharyngeal cancer, biliary tract cancer, bladder cancer, blood cancer, lymphoma, ovarian cancer, prostate cancer, brain tumor, and thyroid cancer. Particularly suitable examples thereof include skin cancer (melanoma), lung cancer, and breast cancer. The site to which the cancer metastasizes is also not particularly limited, but examples thereof include lung, liver, brain, and bone. In particular, metastasis to lung is given. For example, among cancers (malignant tumors) formed in the lung, for example, cancer derived from the lung or bronchial cells is referred to as “primary lung cancer”, and cancer formed by “leaping flame” to the lung from any other site in the body, such as skin cancer, breast cancer, or colon cancer, is referred to as “metastatic lung cancer”. The primary cancer and the metastatic cancer differ from each other in terms of therapeutic strategies, therapeutic methods, and the like. It is conceivable that lung metastasis of melanoma is strongly induced in response to S100A8/A9 secreted by the lungs. As the group of receptors for S100A8/A9, as described in the “Background Art” section, there are known, for example, EMMPRIN, NPTNα, NPTNβ, MCAM, and ALCAM. Those receptors are expressed on the cancer cell side, and have a function of catching an S100A8/A9 signal, leading to, for example, lung metastasis of melanoma. Profiling of the S100A8/A9 receptor group in human melanoma, lung cancer, and breast cancer was performed, and found high expressions of EMMPRIN and MCAM in human melanoma, a high expression of NPTNβ in lung cancer, and a high expression of MCAM in breast cancer. For evaluation of the anti-S100A8/A9 antibody or the antibody fragment thereof of the present invention, an animal model of cancer cell metastasis may be generated. For example, for the metastasis model, for example, B16-BL6 (melanoma), A549 (lung cancer), or MDA-MB-231 (breast cancer) may be used as a cancer cell line reported to undergo lung metastasis in mice. For melanoma, the presence or absence of metastasis can be easily judged by its black color, but in the case of cells for which judgment is difficult, it is also suitable to generate, for example, a line stably expressing a reporter element, such as GFP. For example, in a lung metastasis model of B16-BL6 cells, S100A8/A9-binding decoy protein formulations (exEMMPRIN-Fc, exNPTNβ-Fc, exMCAM-Fc, exRAGE-Fc, and exALCAM-Fc) each show an excellent ability to suppress metastasis. Examples of the inflammatory disease to be targeted by the “anti-inflammatory agent containing the anti-S100A8/A9 antibody or the antibody fragment thereof as an active ingredient” of the present invention include one kind or a plurality of kinds of inflammatory diseases selected from pulmonary fibrosis, lung injury (including acute lung injury and chronic lung injury), systemic inflammatory response syndrome, chronic obstructive pulmonary disease, elderly-onset rheumatoid arthritis, juvenile rheumatoid arthritis, juvenile idiopathic arthritis, inflammatory arthritis, reactive arthritis, uveitis-associated arthritis, inflammatory bowel disease-associated arthritis, inflammatory bowel disease, skin stress, insulitis, nephritis (including glomerulonephritis and pyelonephritis), cystic fibrosis, periodontitis, cervicitis, peritonitis, cancerous peritonitis, diabetic angiopathy, infectious disease, cardiovascular disease, autoimmune disease, autoinflammatory disease, pneumonia (including interstitial pneumonia and cryptogenic organizing pneumonia), pulmonary tuberculosis, pulmonary nontuberculous mycobacteriosis, pneumomycosis, pyothorax, endometritis, metritis, adnexitis, tubo-ovarian abscess, pelvic peritonitis, ankylosing spondylitis, psoriasis, psoriatic arthritis, esophagitis, gastroesophageal reflux disease, esophageal ulcer, gastric ulcer, duodenal ulcer, stress ulcer, steroid ulcer, acute gastritis, chronic gastritis, infectious enteritis, acute colitis, appendicitis, chronic enteritis, irritable bowel syndrome, ulcerative colitis, Crohn's disease, nonalcoholic steatohepatitis (NASH), ischemic colitis, acute pancreatitis, chronic pancreatitis, acute cholecystitis, chronic cholecystitis, cholangitis, hepatitis, collagenosis, mucosal injury, small-intestinal mucosal injury, undifferentiated spondyloarthritis, sepsis, cerebral ischemic infarction, cerebral infarction, brain trauma, brain injury caused by brain surgery, spinal cord injury, arteriosclerosis, acute respiratory distress syndrome, lung injury caused by hemorrhagic shock, multiple organ failure, neuropathic pain, cerebral vasospasm after subarachnoid hemorrhage, burn, polytrauma, idiopathic interstitial pulmonary fibrosis, epilepsy, status epilepticus, viral encephalitis, influenza encephalopathy, inflammatory bowel disease, Kawasaki disease, multiple sclerosis, bronchial asthma, chronic bronchitis, pulmonary emphysema, organ injury after surgery, organ injury after radiotherapy, nephrotic syndrome, acute kidney injury, acute/chronic rejection after organ transplantation, SLE, rheumatoid arthritis, Behcet's disease, myocarditis, endocarditis, ischemia-reperfusion injury, myocardial infarction, congestive heart failure, adipose tissue inflammation, neutrophilic dermatosis, Sweet's disease, and Stevens-Johnson syndrome. Particularly preferred examples thereof include pulmonary fibrosis, acute lung injury, chronic obstructive pulmonary disease, pneumonia (including interstitial pneumonia and cryptogenic organizing pneumonia), pulmonary tuberculosis, pulmonary nontuberculous mycobacteriosis, and pneumomycosis. The “pharmaceutical composition containing the anti-S100A8/A9 antibody or the antibody fragment thereof as an active ingredient” of the present invention may be locally administered, or may be systemically administered. A formulation of the antibody to be used in accordance with the present invention is optionally prepared in a freeze-dried formulation or water-soluble form for storage by mixing the antibody having a desired purity with a pharmaceutically acceptable carrier, excipient, or stabilizer. Formulations for parenteral administration may include sterilized, aqueous or nonaqueous solutions, suspensions, and emulsions. Examples of nonaqueous diluents are propylene glycol, polyethylene glycol, plant oils, such as olive oil, and organic ester compositions, such as ethyl oleate, which are suitable for injection. Aqueous carriers may include water, alcoholic/aqueous solutions, emulsions, suspensions, saline, and buffered media. Parenteral carriers may include sodium chloride solution, Ringer's dextrose, dextrose, and sodium chloride, lactated Ringer's, and fixed oils. Intravenous carriers may include, for example, fluid replenishers, and nutrient and electrolyte replenishers (such as those based on Ringer's dextrose). A therapeutic drug for a disease caused by neutrophil activation and/or an inflammatory disease accompanied by neutrophil activation of the present invention may further contain a preservative and other additives, such as an antimicrobial compound, an antioxidant, a chelating agent, and an inert gas. The “pharmaceutical composition containing the anti-S100A8/A9 antibody or the antibody fragment thereof as an active ingredient” of the present invention may contain two or more active compounds as required for a specific indication to be treated. When the pharmaceutical composition is an anticancer agent, an anticancer agent known per se, an anticancer agent to be developed in the future, and for example, any other medicaments, capable of alleviating a side effect that preferably have complementary activities that do not adversely affect each other, may be used in combination. When the pharmaceutical composition is an anti-inflammatory agent, an anti-inflammatory agent known per se, an anti-inflammatory agent to be developed in the future, and for example, any other medicaments, capable of alleviating a side effect that preferably have complementary activities that do not adversely affect each other may be used in combination. The “pharmaceutical composition containing the anti-S100A8/A9 antibody or the antibody fragment thereof as an active ingredient” of the present invention contains a therapeutically effective amount of the anti-S100A8/A9 antibody or the antibody fragment thereof. The “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result. The therapeutically effective amount may vary depending on factors such as the disease state, age, sex, and body weight of an individual, and the ability of the pharmaceutical to elicit a desired response in the individual. The pharmaceutical composition of the present invention may be used in the following manner: a single dose or divided doses thereof are used generally every 24 hours, 12 hours, 8 hours, 6 hours, 4 hours, or 2 hours or any combination thereof, generally at least once on day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 after the start of treatment, or at least once in week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, or any combination thereof, at a daily dose in terms of daily antibody amount of from about 0.1 mg/kg body weight to about 100 mg/kg body weight, for example, 0.5 mg/kg body weight, 0.9 mg/kg body weight, 1.0 mg/kg body weight, 1.1 mg/kg body weight, 1.5 mg/kg body weight, 2 mg/kg body weight, 3 mg/kg body weight, 4 mg/kg body weight, 5 mg/kg body weight, 6 mg/kg body weight, 7 mg/kg body weight, 8 mg/kg body weight, 9 mg/kg body weight, 10 mg/kg body weight, 11 mg/kg body weight, 12 mg/kg body weight, 13 mg/kg body weight, 14 mg/kg body weight, 15 mg/kg body weight, 16 mg/kg body weight, 17 mg/kg body weight, 18 mg/kg body weight, 19 mg/kg body weight, 20 mg/kg body weight, 21 mg/kg body weight, 22 mg/kg body weight, 23 mg/kg body weight, 24 mg/kg body weight, 25 mg/kg body weight, 26 mg/kg body weight, 27 mg/kg body weight, 28 mg/kg body weight, 29 mg/kg body weight, 30 mg/kg body weight, 40 mg/kg body weight, 45 mg/kg body weight, 50 mg/kg body weight, 60 mg/kg body weight, 70 mg/kg body weight, 80 mg/kg body weight, 90 mg/kg body weight, or 100 mg/kg body weight. EXAMPLES Now, the results of experiments performed to complete the present invention are shown as Reference Example, and the present invention is more specifically described in Examples. However, the present invention is not limited thereto, and various applications are possible without departing from the technical concept of the present invention. (Reference Example 1) Preparation of S100A8/A9 Heterodimer for Generating Anti-S100A8/A9 Antibodies In this Reference Example, the preparation of an S100A8/A9 heterodimer serving as an antigen for the generation of anti-S100A8/A9 antibodies shown in subsequent Examples is described. The S100A8/A9 heterodimer was generated withEscherichia coliusing an expression vector obtained by incorporating full-length S100A8 and full-length S100A9 into pET21 (seeFIG.1), and was purified (see Non Patent Literature 8). For comparative examples, full-length S100A8 or full-length S100A9 was incorporated into pET21, generated withEscherichia coliby the same technique as above, and purified (see Non Patent Literature 8). The purified S100A8/A9 heterodimer, and S100A8 monomer and S100A9 monomer serving as comparative examples were subjected to SDS-PAGE, followed by CBB staining. The results are shown inFIG.2. The S100A8/A9 heterodimer had nearly equal amounts of S100A8 and S100A9, and hence was recognized to have been purified to a high purity. Further, the S100A8/A9 heterodimer was subjected to HPLC analysis. As a result, the results of comparison among the structures of S100A8, S100A9, and S100A8/A9 were as follows: only S100A8/A9 had no monomer presence recognized and mostly had a dimer structure (seeFIG.3). Meanwhile, S100A8 and S100A9 generated as comparative examples were each a mixture of a monomer and a dimer (seeFIG.3). InFIG.4, it is illustrated that a naturally occurring S100A8/A9 heterodimer (abbreviated simply as “A8-A9 heterodimer”) is thermodynamically stable, but S100A8 (abbreviated simply as “A8”) and S100A9 (abbreviated simply as “A9”) each form a homodimer, and hence it is difficult to generate a stable S100A8/A9 heterodimer by mixing S100A8 and S100A9. The S100A8/A9 heterodimer prepared by the method of this Reference Example has high stability, and can be used as an S100A8/A9 heterodimer antigen for generating antibodies in subsequent Examples. (Example 1) Generation of Anti-S100A8/A9 Monoclonal Antibodies In this Example, the generation of anti-S100A8/A9 monoclonal antibodies to be used in the following Examples and experimental examples is described. The anti-S100A8/A9 monoclonal antibodies of this Example were generated using S100A8/A9 prepared in the foregoing (Reference Example 1) as an antigen. (1) Generation of Hybridomas The anti-S100A8/A9 monoclonal antibodies of this Example were generated through use of S100A8/A9 prepared in the foregoing (Reference Example 1) as an antigen and through utilization of a monoclonal antibody on-contract service, GenoStaff (Nippon Genetics). Mice (Balb/c) were used as immunized animals, and Titer-MAX was used as an adjuvant in immunization with the antigen. In accordance with a conventional method, the spleen of the immunized animals and mouse myeloma cells (P3U1) were fused using polyethylene glycol (PEG1500) to generate hybridomas, affording 160 clones. (2) Cloning of Hybridomas and Generation of Antibodies The 160 clones of hybridomas obtained above were subjected to ELISA screening by immobilizing the S100A8/A9 heterodimer, S100A8, or S100A9. Thus, 10 clones shown inFIG.5were selected. Hybridomas expressing the selected S100A8/A9 neutralizing antibody (“α-S100A8/A9 antibody” shown inFIG.5) candidates were adapted to serum-free culture and prepared in large amounts for in vitro and in vivo experiments. A culture supernatant of each clone was recovered and purified with a Protein G column to prepare several milligrams of protein for each of all the clones. A purity test by CBB staining was performed, and as a result, no band other than that of the protein of interest was detected at all. Thus, it was recognized that purified antibodies were prepared at high purities. (3) Reactivity of Monoclonal Antibodies The 10 clones selected in (2) above were each investigated for its reactivity against S100A8/A9 heterodimer, S100A8, or S100A9 and subclass, which are shown in Table 1. TABLE 1ReactivityClone No.S100A8/A9S100A8S100A9Subclass26∘x∘IgG1 κ42∘xxIgG2b κ45∘xxIgG1 κ85∘xxIgG2b κ108∘x∘IgG1 κ213∘∘xIgG2b κ219∘xxIgG2b κ235∘x∘IgG2b κ258∘∘xIgG2b κ260∘x∘IgG1 κ (Example 2) Screening for Neutralizing Antibodies In this Example, for the monoclonal antibodies produced from the 160 clones of hybridomas generated and selected in Example 1, their influences on the production amounts of S100A8/A9-induced inflammatory cytokines were investigated. Through use of human keratinocytes in which inflammatory cytokines were strongly induced by S100A8/A9, the S100A8/A9 signal-suppressing effect of each antibody was evaluated with the mRNA expression amounts of the inflammatory cytokines serving as indicators. Specifically, 30 ng/mL of purified S100A8/A9 and each anti-S100A8/A9 monoclonal antibody purified with the Protein G column from 1 mL of the culture supernatant of each of the 160 clones of hybridomas were added to keratinocytes (NHK), and after culture at 37° C. for 3 hours, the cells were recovered, followed by real-time quantitative PCR (qPCR) analysis for the respective mRNA expression amounts of TNF-α, IL-6, and IL-8. The real-time quantitative PCR (qPCR) analysis was performed using a LightCycler rapid thermal cycler system (ABI 7900HT; Applied Biosystems). Measurement was performed using forward (Fwd) and reverse (Rev) primers having the following base sequences. For TNFα measurementFwd:(SEQ ID NO: 1)GACAAGCCTGTAGCCCATGTRev:(SEQ ID NO: 2)TCTCAGCTCCACGCCATTFor IL-6 measurementFwd:(SEQ ID NO: 3)CTTCCCTGCCCCAGTACCRev:(SEQ ID NO: 4)CTGAAGAGGTGAGTGGCTGTCFor IL-8 measurementFwd:(SEQ ID NO: 5)AGACAGCAGAGCACACAAGCRev:(SEQ ID NO: 6)AGGAAGGCTGCCAAGAGAG As the results of the foregoing, measurement results of the S100A8/A9 (abbreviated simply as “A8/A9”)-induced inflammatory cytokines (TNF-α, IL-6, and IL-8) in the presence of the 10 selected clones (Clone Nos.: 26, 42, 45, 85, 108, 213, 219, 235, 258, and 260) are shown inFIG.6. On the basis of the results, five kinds of antibodies having particularly high suppressive capacities (one kind of antibody reactive to S100A8 (abbreviated simply as “A8”), two kinds of antibodies reactive to S100A9 (abbreviated simply as “A9”), and two kinds of antibodies reactive only to an S100A8/A9 complex (abbreviated simply as “A8/A9”)) were selected. In addition, “α-S100A8/A9 antibody” inFIG.6means anti-S100A8/A9 monoclonal antibody. (Example 3) Evaluation of Chemotaxis of S100A8/A9-induced Cancer Cells In this Example, for the monoclonal antibodies produced from the 160 clones of hybridomas generated and selected in Example 1, their influences on the chemotaxis of cancer cells induced by S100A8/A9 were investigated using a minute cell chemotaxis measurement apparatus TAXiScan™ (GE Healthcare). Profiling of the S100A8/A9 receptor group in human melanoma, lung cancer, and breast cancer was performed, and found high expressions of EMMPRIN and MCAM in human melanoma, a high expression of NPTNβ, in lung cancer, and a high expression of MCAM in breast cancer. Five kinds of S100A8/A9-binding decoy protein formulations (exEMMPRIN-Fc, exNPTNβ-Fc, exMCAM-Fc, exRAGE-Fc, and exALCAM-Fc) each excellently suppress the migration of S100A8/A9-induced cancer cells. In particular, exEMMPRIN-Fc and exNPTNβ-Fc each show high effects on all of the three kinds of cancer cells. In view of this, for respective cells of B16-BL6 (melanoma), A549 (lung cancer), and MDA-MB-231 (breast cancer), the anti-S100A8/A9 antibody of the present invention was also similarly investigated for its influences on the chemotaxis of cancer cells induced by S100A8/A9. Respective cancer cells of B16-BL6 (melanoma), A549 (lung cancer), and MDA-MB-231 (breast cancer) were cultured using, for example, a medium containing 10% FBS in D/F medium (Thermo Fisher Scientific). For measurement, the cells were suspended at 2×106cells/ml in an assay buffer (0.1% mouse serum/RPMI1640/25 mM HEPES). One chamber was loaded with a ligand (S100A8/A9 and monoclonal antibodies generated in Example 1), the other chamber was loaded with cells, and the chemotaxis of each type of cells was measured. The outline of the measurement of the chemotaxis is illustrated inFIG.7. The migration ability of each type of cells in the presence of each of the 5 selected clones (Clone Nos.: 45, 85, 235, 258, and 260) was investigated in terms of velocity and directionality of cell chemotaxis (FIG.8). InFIG.8, “α-S100A8/A9 antibody” means anti-S100A8/A9 monoclonal antibody. As a result, Clone No. 45 showed a particularly strong migration property-suppressing action. (Example 4) Lung Metastasis-Suppressing Effect in Tail Vein Injection of Mouse Breast Cancer 4T1 Cells Through use of a lung metastasis model of mouse breast cancer 4T1 cells, the lung metastasis-suppressing effects of anti-S100A8/A9 monoclonal antibodies were investigated. In accordance with a protocol illustrated inFIG.9, 1×105mouse breast cancer 4T1 cells and 50 μg of each anti-S100A8/A9 monoclonal antibody (Clone Nos.: 45, 85, 235, 258, and 260) were simultaneously injected into the tail vein of five Balb/c nu/nu mice per group, and 2 weeks later, CT scans were performed.FIG.10shows the results for comparing typical CT images and the areas of tumor cells calculated from the CT images to those of a negative control group. As a result, it was recognized that Clone No. 45 showed a significant lung metastasis-suppressing effect. (Example 5) Lung Metastasis-Suppressing Effect in Tail Vein Injection of Human Breast Cancer MDA-MB-231 Cells In this Example, the lung metastasis-suppressing effects of anti-S100A8/A9 monoclonal antibodies were investigated. Through use of a lung metastasis model of human breast cancer MDA-MB-231 cells, the lung metastasis-suppressing effects of anti-S100A8/A9 monoclonal antibodies were investigated. For the MDA-MB-231 cells, a line stably expressing GFP was generated. In accordance with a protocol illustrated inFIG.11, 1×105human breast cancer MDA-MB-231 cells and 50 μg of each anti-S100A8/A9 monoclonal antibodies (Clone Nos.: 45, 85, 235, 258, and 260) were simultaneously injected into the tail vein of each five Balb/c nu/nu mice per group, and 1 month later, CT scans were performed.FIG.12shows the results for comparing typical CT images and the areas of tumor cells calculated from the CT images to those of a negative control group. As a result, it was recognized that Clone Nos. 85, 258, and 260 showed significant lung metastasis-suppressing effects. For the MDA-MB-231 cells, mouse lung metastasis was hardly found, suggesting a need for a further investigation on the generation of a metastasis model. (Example 6) Lung Metastasis-Suppressing Effect in Tail Vein Injection of Mouse Melanoma B16-BL6 Through use of a lung metastasis model of mouse melanoma B16-BL6 cells, the lung metastasis-suppressing effects of anti-S100A8/A9 monoclonal antibodies were investigated. For melanoma, the presence or absence of metastasis can be easily judged by its black color. In accordance with a protocol illustrated inFIG.13, 1×105mouse melanoma B16-BL6 cells and 50 μg of each anti-S100A8/A9 monoclonal antibodies (Clone Nos.: 45, 85, 235, 258, and 260) were simultaneously injected into the tail vein of five Balb/c nu/nu mice per group, and 1 month later, CT scans were performed.FIG.14shows the results for comparing typical mouse lung and CT images and areas calculated from the CT images to those of a negative control group. As a result, it was recognized that Clone Nos. 45, 85, 235, and 258 showed significant lung metastasis-suppressing effects. In particular, Clone No. 45 was found to have a strong metastasis-suppressing effect. (Example 7) Amino Acid Sequences of Variable Regions of Selected Antibodies For the five kinds of anti-S100A8/A9 monoclonal antibodies (Clone Nos.: 45, 85, 235, 258, and 260) selected by the screening described above, the sequences of the variable regions of their heavy chains and light chains were analyzed. VH-CDRClone No. 45:CDR H1:(SEQ ID NO: 7)SYWMQClone No. 45:CDR H2:(SEQ ID NO: 8)AIYPGDGDTRDTQKFKGClone No. 45:CDR H3:(SEQ ID NO: 9)MAGYNYDNDYClone No. 85:CDR H1:(SEQ ID NO: 10)SGYNWHClone No. 85:CDR H2:(SEQ ID NO: 11)YIQYSGSTNYNPSLKSClone No. 85:CDR H3:(SEQ ID NO: 12)ALRYDYSWFAYClone No. 235:CDR H1:(SEQ ID NO: 13)NFWMNClone No. 235:CDR H2:(SEQ ID NO: 14)QIYPGKSDTNYNGKFKGClone No. 235:CDR H3:(SEQ ID NO: 15)WGAYYKYGGSYFDYClone No. 258:CDR H1:(SEQ ID NO: 16)TASMGVSClone No. 258:CDR H2:(SEQ ID NO: 17)HIYWDDDKRYNPSLKSClone No. 258:CDR H3:(SEQ ID NO: 18)RPLGYFDVClone No. 260:CDR H1:(SEQ ID NO: 19)NYGVHClone No. 260:CDR H2:(SEQ ID NO: 20)VVWAGGSTNYNSALMSClone No. 260:CDR H3:(SEQ ID NO: 21)ARDYYGYDGYFGAVL-CDRClone No. 45:CDR L1:(SEQ ID NO: 22)KASQDINKYIAClone No. 45:CDR L2:(SEQ ID NO: 23)YTSTLQPClone No. 45:CDR L3:(SEQ ID NO: 24)LQYDNLRTClone No. 85:CDR L1:(SEQ ID NO: 25)KASQDVSTAVAClone No. 85:CDR L2:(SEQ ID NO: 26)SASYRYTClone No. 85:CDR L3:(SEQ ID NO: 27)QQHYSTPLTClone No. 235:CDR L1:(SEQ ID NO: 28)SASQGISNYLNClone No. 235:CDR L2:(SEQ ID NO: 29)YTSSLHSClone No. 235:CDR L3:(SEQ ID NO: 30)QQYSKFPYTClone No. 258:CDR L1:(SEQ ID NO: 31)KASQDINNYISClone No. 258:CDR L2:(SEQ ID NO: 32)YTSTLQPClone No. 258:CDR L3:(SEQ ID NO: 33)LQYDNLLWTClone No. 260:CDR L1:(SEQ ID NO: 34)KASQDINSYLTClone No. 260:CDR L2:(SEQ ID NO: 35)RANRLVDClone No. 260:CDR L3:(SEQ ID NO: 36)LQYDEFPLT (Example 8) Generation of Anti-S100A8/A9 Chimeric Antibody In this Example, a chimeric antibody having the Fc portion of human IgG2fused to the Fab domain of the S100A8/A9 monoclonal antibody (Clone No. 45) was generated. Sequence analysis and CDR analysis of the variable regions of the heavy chain and light chain of the S100A8/A9 monoclonal antibody (Clone No. 45) were performed, and a stable expression vector for CHO cells having incorporated therein sequences recombined with variable regions of human IgG2was generated and transduced into CHO cells in combination with a gene for the Fc portion of human IgG2. Thus, the anti-S100A8/A9 chimeric antibody was stably generated (FIG.15). The antibody was generated by a method described in WO 2017/061354 A1. (Example 9) Lung Metastasis-Suppressing Effect in Tail Vein Injection of Mouse Melanoma B16-BL6 Cells Through use of a lung metastasis model of mouse melanoma B16-BL6 cells, the lung metastasis-suppressing effect of the anti-S100A8/A9 chimeric antibody generated in Example 8 was investigated. In accordance with a protocol illustrated inFIG.16, 1×105mouse melanoma B16-BL6 cells and 50 μg of the anti-S100A8/A9 chimeric antibody were simultaneously injected into the tail vein of five Balb/c nu/nu mice per group, and 2 weeks later, CT scans were performed.FIG.17shows the results for comparing typical mouse lung and CT images and areas calculated from the CT images to those of a negative control group. As a result, it was recognized that the antibody of Clone No. 45 significantly suppressed lung metastasis also as the chimeric antibody obtained by fusing the Fc portion of human IgG2 thereto, demonstrating its usefulness as a lung metastasis suppressor for melanoma. (Example 10) Lung Metastasis-Suppressing Effect in Local Injection of Mouse Melanoma B16-BL6 Cells In this Example, the lung metastasis-suppressing effect of the anti-S100A8/A9 monoclonal antibody (Clone No. 45) generated in Example 1 was investigated. In accordance with a protocol illustrated inFIG.18, 1×105mouse melanoma B16-BL6 cells were intradermally injected into the right ear of two Balb/c nu/nu mice per group. After a lapse of 13 days, at a time point when a tumor measuring from about 4 mm to about 5 mm was observed, 0 μg, 10 μg, 50 μg, or 100 μg of the anti-S100A8/A9 monoclonal antibody was injected into the tail vein. After 1 day from the antibody injection, the tumor in the right ear was partially resected to induce metastasis of the melanoma B16-BL6 cells. After a lapse of 20 days from the antibody injection, metastasis to the lungs was observed. As a result, it was observed that lung metastasis was suppressed in an injection concentration-dependent manner (FIG.19). It was recognized that even antibody injection after tumor formation suppressed lung metastasis in a dose-dependent manner. InFIG.19, “α-S100A8/A9 antibody (Ab45)” means the anti-S100A8/A9 monoclonal antibody (Clone No. 45). In accordance with the protocol illustrated inFIG.18, three Balb/c nu/nu mice per group were injected with 50 μg of the anti-S100A8/A9 monoclonal antibody (Clone No. 45) or IgG serving as a control, and after a lapse of 20 days, lung foci were checked. As a result, a metastasis-suppressing effect was clearly observed in the Clone No. 45-injected group (FIG.20). Lung foci were observed in each of the groups injected with 0 μg, 10 μg, 50 μg, and 100 μg of the anti-S100A8/A9 monoclonal antibody (Clone No. 45). As a result, in the 10 μg-injected group, a significant reduction in number of lung foci was found (Table 2). In Table 2 below, “α-S100A8/A9 antibody (Ab45)” means the anti-S100A8/A9 monoclonal antibody (Clone No. 45). TABLE 2α-S100A8/A9 antibody (Ab45)0 μg10 μg50 μg100 μgThe Number of Total Lung FociNo. 1221321(≥1 mm in diameter)No. 22121630The Number of Lung FociNo. 12000(≥5 mm in diameter)No. 20000 (Example 11) Lung Injury-Suppressing Effect in Pulmonary Fibrosis Model Intratracheally Injected with Bleomycin As shown inFIG.21, it has been recognized that S100A8 and S100A9 proteins are expressed in a human lung tissue of idiopathic pulmonary fibrosis. In this Example, the lung injury-suppressing effect of the anti-S100A8/A9 monoclonal antibody (Clone No. 45) generated in Example 1 in a pulmonary fibrosis model intratracheally injected with bleomycin was investigated. In accordance with a protocol illustrated inFIG.22, six or seven female C57BL/6J (11-week-old) mice per group were intratracheally injected with 50 μl of PBS containing 20 μg/mouse of bleomycin to generate acute lung injury model mice. As a result of the bleomycin injection, abrupt increases in S100A8 and S100A9, which are proteins involved in inflammation, were observed in the lung tissue (FIG.22). At from 2 hours to 3 hours after the bleomycin injection, 50 μg of the anti-S100A8/A9 monoclonal antibody (Clone No. 45) was injected into the tail vein. As a control, IgG was injected. Changes in body weight of the acute lung injury model mice until a lapse of 21 days after the bleomycin injection were observed, and as a result, a mouse body weight reduction-suppressing effect was found in the anti-S100A8/A9 monoclonal antibody (Clone No. 45)-injected group as compared to the IgG-injected group (FIG.23). Further, in the anti-S100A8/A9 monoclonal antibody (Clone No. 45)-injected group, a suppressing effect on lung injury on day 21 after the bleomycin injection was observed (FIG.24). The lung injury was observed by CT scanning. As a result of an investigation by pathological observation of a tissue slice, a suppressing effect on the injury/fibrosis of the lung tissue was observed (FIG.25). FIG.26is a schematic illustration of: a situation in which the production of S100A8 and S100A9 is enhanced in the lungs of a patient subjected to various stresses due to, for example, systemic inflammatory response syndrome, medication, radiation irradiation, operation, and ischemic reperfusion injury, with the result that lung injury progresses; and the preventing or ameliorating effect of anti-S100A8/A9 antibody injection on the inflammation/tissue injury of the lung tissue. INDUSTRIAL APPLICABILITY As described in detail above, the anti-S100A8/A9 antibody of the present invention has an action of suppressing the metastasis of cancer cells. In recent years, the survival rate of cancer patients has been presumably improved by virtue of improvements in, for example, prevention, diagnosis, and treatment of cancer. Also in anticancer agent treatment, effective treatment has been developed by, for example, using an anticancer agent having a high therapeutic effect and having reduced side effects, or combining a plurality of medicaments, to thereby improve a treatment outcome. However, there still remains a problem in that cancer metastasis is difficult to treat for the purpose of cure. Under such circumstances, the anti-S100A8/A9 antibody of the present invention can effectively suppress the metastasis of cancer cells, thereby making a great contribution to improving a cancer treatment outcome, and hence the antibody is industrially extremely useful. Further, the anti-S100A8/A9 antibody of the present invention also takes effect on various inflammatory diseases, and hence the industrial usefulness of the present invention as such is immeasurable.
43,319
11858986
DETAILED DESCRIPTION OF THE INVENTION 1. Anti-IFNβ Antibodies A. Interferon Beta (IFNβ) Interferon beta (IFNβ), also known as fibroblast IFN, is a glycosylated, secreted, and approximately 22 kDa member of the type I interferon family of molecules. The sequence of human IFNβ precursor is shown as SEQ ID NO: 40. A signal peptide (residues 1-21 of SEQ ID NO: 40) of the precursor is cleaved to produce mature IFNβ (SEQ ID NO: 41), which shares 47% and 46% amino acid sequence identity with the mouse and rat proteins, respectively. Alignments of IFNβ from various species are shown inFIG.15B. The signal peptide is underlined in the sequence below. MTNKCLLQIA LLLCFSTTAL SMSYNLLGFL QRSSNFQCQKLLWQLNGRLE YCLKDRMNFD IPEEIKQLQQ FQKEDAALTIYEMLQNIFAI FRQDSSSTGW NETIVENLLA NVYHQINHLKTVLEEKLEKE DFTRGKLMSS LHLKRYYGRI LHYLKAKEYSHCAWTIVRVE ILRNEYFINR LTGYLRN (Human IFNβprecursor, SEQ ID NO: 40) The structure of IFNβ contains 5 α-helices, designated A (YNLLGFLQRSSNFQCQKLL; SEQ ID NO:153 or residues 3-21 of SEQ ID NO:41), B (KEDAALTIYEMLQNIFAIF; SEQ ID NO:154 or residues 52-70 of SEQ ID NO:41), C (ETIVENLLANVYHQINHLKTVLEEKL; SEQ ID NO:155 or residues 81-106 of SEQ ID NO:41), D (SLHLKRYYGRILHYLKA; SEQ ID NO:156 or residues 119-135 of SEQ ID NO:41), and E (HCAWTIVRVEILRNFYFINRLT; SEQ ID NO:157 or residues 140-161 of SEQ ID NO:41). The five α-helices are interconnected by loops of 2 to 28 residues designated AB, BC, CD, and DE loops (FIG.15A). It has been reported that the A helix, the AB loop, and the E helix are involved in binding of IFNβ to its receptor, IFNAR. B. Anti-IFNβ Antibodies One potential drawback of an anti-IFNAR antibody (e.g., anifrolimab) is that both IFNα and IFNβ cytokines bind to IFNAR. Although these two types of IFN cytokines elicit similar biological activities to a similar degree, there are significant differences in potency and cell type specific activities between these two types of IFNs. For example, IFNβ elicits a markedly higher anti-proliferative response in some cell types, such as embryonal carcinoma, melanoma and melanocytes, than does IFNα. Higher potency of IFNβ in treatment of multiple sclerosis and certain cancers has also been observed. Blocking the activity of IFNAR, however, does not selectively modulate the activities of IFNβ. Significantly, IFNα is an important cytokine in response to viral infections, such that blocking its activity may have unwanted effects. Accordingly, an antibody that specially binds IFNβ, but not IFNα, would fulfill a significant unmet need for treatment of diseases that are primarily driven by IFNβ. In one aspect, the invention provides an isolated antibody, or antigen-binding fragment thereof, that specifically binds human IFNβ. Sequences of exemplary antibodies of the invention are shown in Table 11. As shown in the Examples, in certain embodiments, the antibody of the invention inhibits the binding of IFNβ to its receptor, and is hence referred to as a “neutralizing” antibody. Without wishing to be bound by any particular theory, the data indicate that the antibody, or antigen-binding fragment thereof, blocks, or partially blocks, the receptor binding sites of IFNβ, either by competing for the same or overlapping residues from IFNAR, or by creating steric hindrance. For example, residues from helix A, AB loop, and helix E of IFNβ are believed to be involved in binding of IFNβ to its receptor. Accordingly, in certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention binds an epitope comprising one more residues selected from the group consisting of: residues 3-21 (helix A), 22-51 (AB loop); and 140-161 (helix E), according to the numbering of SEQ ID NO: 41. In certain embodiments, the antibody, or antigen-binding fragment thereof, bind to human IFNβ with a binding affinity (KD) value that is at least 100 fold less, than its KDvalue for a human IFNα under substantially the same assay conditions. For example, the ratio of KDfor IFNβ versus KDfor IFNα can be 1:100 or less, 1:250 or less, 1:500 or less, 1:1000 or less, 1:2500 or less, 1:5000 or less, or 1:10,000 or less. Mutagenesis studies and crystal structure studies also identified epitope residues in human IFNβ that are recognized by anti-IFNβ antibodies disclosed herein. In particular, among all IFNβ residues that are within 3.8 Å from a heavy atom of the antibody (“potential” epitope residues), three different types have been identified: (i) “primary” epitope residues that are characterized as highly buried residues at the of antibody-antigen interface and zero-to-low sequence tolerance to any other amino acid substitutions at this position; (ii) “secondary” epitope residues that are characterized as residues with medium buried surface area at the interface and medium sequence tolerance to amino acid substitutions at these positions; and (iii) “Optional” epitope residues are characterized as residues with low buried surface area at the interface and high sequence tolerance to amino acid substitutions at these positions. Accordingly, in certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention specifically binds an epitope in human IFNβ, wherein said epitope comprises one or more residues selected from the group consisting of Ala89, Tyr 92, His93, and His97, according to the numbering of SEQ ID NO:41 (“primary” epitope residues). In certain embodiments, the epitope further comprises one or more residues selected from the group consisting of Phe8, Leu9, Ser12, Gln16, Asn86, Asn90, Asp96, and Thr100, according to the numbering of SEQ ID NO:41 (“secondary” epitope residues). In certain embodiments, the epitope further comprises one or more residues selected from the group consisting of Leu5, Leu6, Ser13, Phe15, and Thr82, according to the numbering of SEQ ID NO:41 (“optional” epitope residues). In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention also specifically binds cynomolgus monkey IFNβ, in addition to human IFNβ.In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention specifically binds an epitope in cynomolgus monkey IFNβ, wherein said epitope comprises one or more residues selected from the group consisting of Ala89, Tyr 92, His93, and His97, according to the numbering of SEQ ID NO:44 (“primary” epitope residues). In certain embodiments, the epitope further comprises one or more residues selected from the group consisting of Phe8, Leu9, Ser12, Gln16, Asn86, Asn90, Asp96, Thr100 and Tyr67, according to the numbering of SEQ ID NO:44 (“secondary” epitope residues). In certain embodiments, the epitope further comprises one or more residues selected from the group consisting of Leu5, Leu6, Ser13, Phe15, and Thr82, according to the numbering of SEQ ID NO:44 (“optional” epitope residues). Provided herein are antibody CTI-AF1 and variants thereof. Accordingly, in certain embodiments, the antibody or antigen-binding fragment thereof comprises the following heavy chain CDR sequences: (i) CDR-H1 comprising SEQ ID NO: 37, CDR-H2 comprising SEQ ID NO: 38, and CDR-H3 comprising SEQ ID NO: 39; and/or (ii) the following light chain CDR sequences: CDR-L1 comprising SEQ ID NO: 34, CDR-L2 comprising SEQ ID NO: 35, and CDR-L3 comprising SEQ ID NO: 36. As demonstrated from the crystal structure studies, not all residues in CDRs contribute to antibody-antigen binding. As shown in Example 7 and Table 14, only limited number of CDR residues are within 3.8 Å from a heavy atom of the antigen, and are considered as potential paratope residues. Among these potential paratope residues, (i) “primary” paratope residues are those characterized as highly buried residues at the antibody-antigen interface and low sequence tolerance to any other amino acid substitutions at this position; and (ii) “secondary” paratope residues are characterized as residues with lower buried surface area at the interface and higher sequence tolerance to amino acid substitutions at these positions. Accordingly, in certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention comprises a VH chain that comprises one or more paratope residues selected from the group consisting of: Trp33 in CDR-H1, Tyr56 in CDR-H2, Tyr58 in CDR-H2, and Tyr97 in CDR-H3, according to Kabat numbering (“primary” paratope residues). In certain embodiments, the VH further comprises one or more paratope residues selected from the group consisting of: Asp54 in CDR-H2, Gln61 in CDR-H2, Gly98 in CDR-H3, and Leu100 in CDR-H3, according to Kabat numbering (“secondary” paratope residues). In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention comprises a VL that comprises one or more paratope residues selected from the group consisting of: Tyr32 in CDR-L1, Ile92 in CDR-L3, and Leu94 in CDR-L3, according to Kabat numbering (“primary” paratope residues). In certain embodiments, the VH further comprises one or more paratope residues selected from the group consisting of: Gln27 in CDR-L1, Asp28 in CDR-L1, Ile29 in CDR-L1, Gly30 in CDR-L1, and Ile93 in CDR-L3, according to Kabat numbering (“secondary” paratope residues). The antibody, or antigen binding fragment thereof, of the invention may also comprise any combination of the paratope residues disclosed herein. In certain embodiments, the antibody, or antigen-binding fragment thereof, described herein comprises the following heavy chain CDR sequences: (i) a CDR-H1 sharing at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, or at least 95% identical to SEQ ID NO: 37, a CDR-H2 sharing at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, or at least 95% identity with SEQ ID NO: 38, and a CDR-H3 sharing at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, or at least 95% identity with SEQ ID NO: 39; and/or (ii) the following light chain CDR sequences: a CDR-L1 sharing at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, or at least 95% identity with SEQ ID NO: 34, a CDR-L2 sharing at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, or at least 95% identity with SEQ ID NO: 35, and a CDR-L3 sharing at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, or at least 95% identity with SEQ ID NO: 36. In certain embodiments, the amino acid differences, as compared to SEQ ID NOs. 37, 38, 39, 34, 35, and 36, respectively, are not one of the primary or secondary paratope residues as shown in Table 14. In certain embodiments, no more than 10, no more than 9, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 3, no more than 2, or no more than one substitution is made in the sequence of CDR-L1, relative to SEQ ID NO. 34. In certain embodiments, no more than 6, no more than 5, no more than 4, no more than 3, no more than 3, no more than 2, or no more than one substitution is made in the sequence of CDR-L2, relative to SEQ ID NO. 35. In certain embodiments, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 3, no more than 2, or no more than one substitution is made in the sequence of CDR-L3, relative to SEQ ID NO. 36. In certain embodiments, no more than 9, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 3, no more than 2, or no more than one substitution is made in the sequence of CDR-H1, relative to SEQ ID NO. 37. In certain embodiments, no more than 16, no more than 15, no more than 14, no more than 13, no more than 12, no more than 11, no more than 10, no more than 9, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 3, no more than 2, or no more than one substitution is made in the sequence of CDR-H2, relative to SEQ ID NO. 38. In certain embodiments, no more than 9, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 3, no more than 2, or no more than one substitution is made in the sequence of CDR-H3, relative to SEQ ID NO. 39. In certain embodiments, the substitution does not change binding affinity (KD) value by more than 3 orders of magnitude, more than 2 orders of magnitude, or 1 order of magnitude, as compared with the KDof the antibody, or antigen-binding fragment thereof, without the substitution. In certain embodiments, the substitution is not one of the primary or secondary paratope residues as shown in Table 14. In certain embodiments, the substitution is a conservative substitution as provided by Table 1. TABLE 1Exemplary Conservative SubstitutionsConservativeConservativeResiduesubstitutionResiduesubstitutionAlaSerLeuIle, ValArgLysLysArg, GlnAsnGln; HisMetLeu, IleAspGluPheMet, Leu, TyrCysSerSerThr; GlyGlnAsnThrSer, ValGluAspTrpTyrGlyProTyrTrp, PheHisAsn, GlnValIle, LeuIleLeu, ValPro— In certain embodiments, when an antibody is derived from a non-human species, such as a humanized antibody in which murine CDRs are grafted to a human framework, the substitution is human germline substitution in which a non-human CDR residue is replaced with the corresponding human germline residue. One benefit of such substitution is to increase the human amino acid content, and to reduce potential immunogenicity of an antibody derived from a non-human species. For example, if human germline DPK9 framework is used and the exemplary antibody CTI-AF1, then the alignment of the CDR-L1 of CTI-AF1 antibody and human germline DPK9 is as follows: TABLE 2Position2425262728293031323334Human GermlineRASQSISSYLNDPK9(SEQ ID NO: 46)CTI-AF1 antibodyRTSQDIGNYLN(SEQ ID NO: 34) For positions 24, 26, 27, 29, 32, 33, and 34, the human germline residue and the corresponding CTI-AF1 residue are the same, and no substitution is needed at these positions. For positions 25, 28, 30, and 31 (in bold), the human germline residue and the corresponding CTI-AF1 murine residue are different. Murine residues of CTI-AF1 at these positions may be replaced with the corresponding human germline DPK9 residue to further increase the human amino acid residue content. Methods and libraries for introducing human germline residues in antibody CDRs are described in detail in Townsend et al.,Augmented Binary Substitution: Single-pass CDR germlining and stabilization of therapeutic antibodies, PNAS, vol. 112, 15354-15359 (2015), and United States Patent Application Number 2017-0073395 A1 (published Mar. 16, 2017) and are herein incorporated by reference in their entirety. In certain embodiments, the antibody, or antigen-binding fragment thereof, described herein comprises a human framework sequence. For example, a heavy chain framework sequence can be derived from a human VH3 germline, a VH1 germline, a VH5 germline, or a VH4 germline sequence. Preferred human germline heavy chain frameworks are frameworks derived from VH1, VH3, or VH5 germline sequences. For example, VH frameworks from the following well-known germline sequences may be used: IGHV3-23, IGHV3-7, or IGHV1-69, where germline names are based on IMGT germline definition. Preferred human germline light chain frameworks are frameworks derived from VK or Vλ germline sequences. For example, VL frameworks from the following germlines may be used: IGKV1-39 or IGKV3-20, where germline names are based on IMGT germline definition. Alternatively or in addition, the framework sequence may be a human germline consensus framework sequence, such as the framework of human Vλ1 consensus sequence, VK1 consensus sequence, VK2 consensus sequence, VK3 consensus sequence, VH3 germline consensus sequence, VH1 germline consensus sequence, VH5 germline consensus sequence, or VH4 germline consensus sequence. Sequences of human germline frameworks are available from various public databases, such as V-base, IMGT, NCBI, or Abysis. In certain embodiments, the human germline VL framework is the framework of DPK9 (IMGT name: IGKV1-39), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK9 germline residues as shown in Table 3 (SEQ ID NOs.:46, 47, 48). TABLE 3SEQIDLight Chain46DPK9 CDR-L1RASQSISSYLN47DPK9 CDR-L2AASSLQS48DPK9 CDR-L3QQSYSTP49DPK12 CDR-L1KSSQSLLHSDGKTYLY50DPK12 CDR-L2EVSNRFS51DPK12 CDR-L3MQSIQLP52DPK18 CDR-L1RSSQSLVYSDGNTYLN53DPK18 CDR-L2KVSNRDS54DPK18 CDR-L3MQGTHWP55DPK24 CDR-L1KSSQSVLYSSNNKNYLA56DPK24 CDR-L2WASTRES57DPK24 CDR-L3QQYYSTP58HK102_V1 CDR-L1RASQSISSWLA59HK102_V1 CDR-L2DASSLES60HK102_V1 CDR-L3QQYNSYS61DPK1 CDR-L1QASQDISNYLN62DPK1 CDR-L2DASNLET63DPK1 CDR-L3QQYDNLP64DPK8 CDR-L1RASQGISSYLA65DPK8 CDR-L2AASTLQS66DPK8 CDR-L3QQLNSYP67DPK21 CDR-L1RASQSVSSNLA68DPK21 CDR-L2GASTRAT69DPK21 CDR-L3QQYNNWP70Vg_38K CDR-L1RASQSVSSYLA71Vg_38K CDR-L2DASNRAT72Vg_38K CDR-L3QQRSNWP73DPK22 CDR-L1RASQSVSSSYLA74DPK22 CDR-L2GASSRAT75DPK22 CDR-L3QQYGSSP76DPK15 CDR-L1RSSQSLLHSNGYNYLD77DPK15 CDR-L2LGSNRAS78DPK15 CDR-L3MQALQTP79DPL16 CDR-L1QGDSLRSYYAS80DPL16 CDR-L2GKNNRPS81DPL16 CDR-L3NSRDSSGNH82DPL8 CDR-L1TGSSSNIGAGYDVH83DPL8 CDR-L2GNSNRPS84DPL8 CDR-L3QSYDSSLSG85V1-22 CDR-L1TRSSGSIASNYVQ86V1-22 CDR-L2EDNQRPS87V1-22 CDR-L3QSYDSSN88Vλ consensus CDR-L1TGSSSGGSYYVS or89TGSSSDVGGSYYVS90Vλ consensus CDR-L2ENDSNRPS or91EDSNR(S/D)K(Q/G)QKPS92Vλ consensus CDR-L3QSWDSSA(N/T) or93QSWDSSA(N/T)F(F/V)(G/V)94Vλ1 consensus CDR-L1SGSSSNIGNN(A/Y)V(N/H/S)or95SGSSSNIIGNN(A/Y)V(N/H/S)96Vλ1 consensusCDR-L2GNN(K/N/Q)RPS97Vλ1 consensusCDR-L3AAWDDSL(N/S)G98Vλ3 consensusCDR-L1CSGD(A/V)LG(K/S)KYAH99Vλ3 consensusCDR-L2KDSERPS100Vλ3 consensusCDR-L3QSWDSSG(N/D/T/A) or101QSWDSSG(N/D/T/A)H102Vκ consensus CDR-L1RASQSLLHSDGISSYLA or103RASQGISSYLA104Vκ consensus CDR-L2AASSRAS105Vκ consensus CDR-L3QQYNSYP106Vκ1 consensus CDR-L1RASQGIS(N/S)YLA107Vκ1 consensus CDR-L2AASSLQS108Vκ1 consensus CDR-L3QQYNSYP109Vκ2 consensus CDR-L1RSSQSLLHSDGNTYLD or110RSSQSLLHSDDGNTYLD111Vκ2 consensus CDR-L2(K/T)(V/I)SNR(A/F)S112Vκ2 consensus CDR-L3MQATQFP113Vκ3 consensus CDR-L1RASQS(S/V)(S/V)SSYLA114Vκ3 consensus CDR-L2GASTRAT115Vκ3 consensus CDR-L3QW(S/N/G/H)NWP In certain embodiments, the human germline VL framework is the framework of DPK12 (IMGT name: IGKV2D-29), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK12 germline residues as shown in Table 3 (SEQ ID NOs.:49, 50, 51). In certain embodiments, the human germline VL framework is the framework of DPK18 (IMGT name: IGKV2-30), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK18 germline residues as shown in Table 3 (SEQ ID NOs.:52, 53, 54). In certain embodiments, the human germline VL framework is the framework of DPK24 (IMGT name: IGKV4-1), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK24 germline residues as shown in Table 3 (SEQ ID NOs.:55, 56, 57). In certain embodiments, the human germline VL framework is the framework of HK102_V1 (IMGT name: IGKV1-5), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding HK102_V1 germline residues as shown in Table 3 (SEQ ID NOs.:58, 59, 60). In certain embodiments, the human germline VL framework is the framework of DPK1 (IMGT name: IGKV1-33), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK1 germline residues as shown in Table 3 (SEQ ID NOs.:61, 62, 63). In certain embodiments, the human germline VL framework is the framework of DPK8 (IMGT name: IGKV1-9), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK8 germline residues as shown in Table 3 (SEQ ID NOs.:64, 65, 66). In certain embodiments, the human germline VL framework is the framework of DPK21 (IMGT name: IGKV3-15), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK21 germline residues as shown in Table 3 (SEQ ID NOs.:67, 68, 69). In certain embodiments, the human germline VL framework is the framework of Vg_38K (IMGT name: IGKV3-11), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding Vg_38K germline residues as shown in Table 3 (SEQ ID NOs.:70, 71, 72). In certain embodiments, the human germline VL framework is the framework of DPK22 (IMGT name: IGKV3-20), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK22 germline residues as shown in Table 3 (SEQ ID NOs.:73, 74, 75). In certain embodiments, the human germline VL framework is the framework of DPK15 (IMGT name: IGKV2-28), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPK15 germline residues as shown in Table 3 (SEQ ID NOs.:76, 77, 78). In certain embodiments, the human germline VL framework is the framework of DPL16 (IMGT name: IGLV3-19), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPL16 germline residues as shown in Table 3 (SEQ ID NOs.:79, 80, 81). In certain embodiments, the human germline VL framework is the framework of DPL8 (IMGT name: IGLV1-40), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DPL8 germline residues as shown in Table 3 (SEQ ID NOs.:82, 83, 84). In certain embodiments, the human germline VL framework is the framework of V1-22 (IMGT name: IGLV6-57), and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding V1-22 germline residues as shown in Table 3 (SEQ ID NOs.:85, 86, 87). In certain embodiments, the human germline VL framework is the framework of human VA, consensus sequence, and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding VA, germline consensus residues as shown in Table 3 (SEQ ID NOs.:88, 89, 90, 91, 92, 93). Alternative sequences are provided for the consensus sequence with and without gaps. At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VL framework is the framework of human Vλ1 consensus sequence, and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding Vλ1 germline consensus residues as shown in Table 3 (SEQ ID NOs.:94, 95, 96, 97) Alternative sequences are provided for the consensus sequence with and without gaps. At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VL framework is the framework of human Vλ3 consensus sequence, and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding Vλ3 germline consensus residues as shown in Table 3 (SEQ ID NOs.: 98, 99, 100, 101). Alternative sequences are provided for the consensus sequence with and without gaps. At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VL framework is the framework of human Vκ consensus sequence and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding Vκ germline consensus residues as shown in Table 3 (SEQ ID NOs.:102, 103, 104, 105). Alternative sequences are provided for the consensus sequence with and without gaps. In certain embodiments, the human germline VL framework is the framework of human Vκ1 consensus sequence, and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding Vκ1 germline consensus residues as shown in Table 3 (SEQ ID NOs.:106, 107, 108). At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VL framework is the framework of human Vκ2 consensus sequence, and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding Vκ2 germline consensus residues as shown in Table 3 (SEQ ID NOs.:109, 110, 111, 112). Alternative sequences are provided for the consensus sequence with and without gaps. At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VL framework is the framework of human VK3 consensus sequence, and one or more residues in CDR-L1, CDR-L2, and CDR-L3 of the antibodies (and fragments) of the invention may be substituted with the corresponding germline residues as shown in Table 3 (SEQ ID NOs.:113, 114, 115). At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VH framework is the framework of DP54 (IMGT name: IGHV3-7), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding germline residues as shown in Table 4 SEQ ID NOs.:116, 117). TABLE 4SEQIDHeavy Chain116DP54 CDR-H1GFTFSSYWMS117DP54 CDR-H2ANIKQDGSEKYYVDSVKG118DP47 CDR-H1GFTFSSYAMS119DP47 CDR-H2AISGSGGSTYYADSVKG120DP71 CDR-H1GGSISSYYWS121DP71 CDR-H2GYIYYSGSTNYNPSLKS122DP75 CDR-H1GYTFTGYYMH123DP75 CDR-H2GWINPNSGGTNYAQKFQG124DP10 CDR-H1GGTFSSYAIS125DP10 CDR-H2GGIIPIFGTANYAQKFQG126DP7 CDR-H1GYTGTSYYMH127DP7 CDR-H2GIINPSGGSTSYAQKFQG128DP49 CDR-H1GFTFSSYGMH129DP49 CDR-H2AVISYDGSNKYYADSVKG130DP51 CDR-H1GFTFSSYSMN131DP51 CDR-H2SYISSSSSTIYYADSVKG132DP38 CDR-H1GFTFSNAWMS133DP38 CDR-H2GRIKSKTDGGTTDYAAPVKG134DP79 CDR-H1GGSISSSSYYWG135DP79 CDR-H2GSIYYSGSTYYNPSLKS136DP78 CDR-H1GGSISSGDYYWS137DP78 CDR-H2GYIYYSGSTYYNPSLKS138DP73 CDR-H1GYSFTSYWIG139DP73 CDR-H2GIIYPGDSDTRYSPSFQG140VH consensus CDR-H1GFTFSSYAM(H/S) or141GFTFSSYAM(H/S)WS142VH consensus CDR-H2GWISPNGGSTYYADSVKG or143GWISPKANGGSTYYADSVKG144VH3 consensus CDR-H1GFTFSSYAMS145VH3 consensus CDR-H2SVISSDG(G/S)STYYADSVKGor146SVISSKADG(G/S)STYYADSVKG147VH5 consensus CDR-H1GYSFTSYWI(S/G/H)148VH5 consensus CDR-H2G(R/I/S)IYPGDSDTRYSPSFQG149VH1 consensus CDR-H1GYTFTSY(A/Y)(I/M)H150VH1 consensus CDR-H2GWINP(G/Y)NGNTNYAQKFQ151VH4 consensus CDR-H1GGSISSG(N/Y)YYWS152VH4 consensus CDR-H2GYIYYSGSTYYNPSLKS In certain embodiments, the human germline VH framework is the framework of DP47 (IMGT name: IGHV3-23), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP47 germline residues as shown in Table 4 (SEQ ID NOs.:118, 119). In certain embodiments, the human germline VH framework is the framework of DP71 (IMGT name: IGHV4-59), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP71 germline residues as shown in Table 4 (SEQ ID NOs.:120, 121). In certain embodiments, the human germline VH framework is the framework of DP75 (IMGT name: IGHV1-2_02), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP75 germline residues as shown in Table 4 (SEQ ID NOs.:122, 123). In certain embodiments, the human germline VH framework is the framework of DP10 (IMGT name: IGHV1-69), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP10 germline residues as shown in Table 4 (SEQ ID NOs.:124, 125). In certain embodiments, the human germline VH framework is the framework of DP7 (IMGT name: IGHV1-46), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP7 germline residues as shown in Table 4 (SEQ ID NOs.:126, 127). In certain embodiments, the human germline VH framework is the framework of DP49 (IMGT name: IGHV3-30), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP49 germline residues as shown in Table 4 (SEQ ID NOs.:128, 129). In certain embodiments, the human germline VH framework is the framework of DP51 (IMGT name: IGHV3-48), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP51 germline residues as shown in Table 4 (SEQ ID NOs.:130, 131). In certain embodiments, the human germline VH framework is the framework of DP38 (IMGT name: IGHV3-15), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP38 germline residues as shown in Table 4 (SEQ ID NOs.:132, 133). In certain embodiments, the human germline VH framework is the framework of DP79 (IMGT name: IGHV4-39), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP79 germline residues as shown in Table 4 (SEQ ID NOs.:134, 135). In certain embodiments, the human germline VH framework is the framework of DP78 (IMGT name: IGHV4-30-4), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP78 germline residues as shown in Table 4 (SEQ ID NOs.:136, 137). In certain embodiments, the human germline VH framework is the framework of DP73 (IMGT name: IGHV5-51), and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding DP73 germline residues as shown in Table 4 (SEQ ID NOs.:138, 139). In certain embodiments, the human germline VH framework is the framework of human VH germline consensus sequence, and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding VH germline consensus residues as shown in Table 4 (SEQ ID NOs.:140, 141, 142, 143). Alternative sequences are provided for the consensus sequence with and without gaps. At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VH framework is the framework of human VH3 germline consensus sequence, and r one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding VH3 germline consensus residues as shown in Table 4 (SEQ ID NOs.:144, 145, 146). Alternative sequences are provided for the consensus sequence with and without gaps. At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VH framework is the framework of human VH5 germline consensus sequence, and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding VH5 germline consensus residues as shown in Table 4 (SEQ ID NOs.:147, 148). At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VH framework is the framework of human VH1 germline consensus sequence, and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding VH1 germline consensus residues as shown in Table 4 (SEQ ID NOs.:149, 150). At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the human germline VH framework is the framework of human VH4 germline consensus sequence, and one or more residues in CDR-H1 and CDR-H2 of the antibody, or antigen-binding fragment thereof, of the invention may be substituted with the corresponding VH4 germline consensus residues as shown in Table 4 (SEQ ID NOs.:151, 152). At positions where there is no consensus, residues within parenthesis ( ) are those that are tied for the most frequent residues present in human antibodies. In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention comprises (numbering according to Kabat):(i) a VH that comprises: (a) a CDR-H1 comprising Trp33, and three or fewer amino acid differences as compared to SEQ ID NO: 37, (b) a CDR-H2 comprising Asp54, Tyr56, Tyr58, and Gln61, and three or fewer amino acid differences as compared to ID NO: 38; and (c) a CDR-H3 comprising Tyr97, Gly98, and Leu100; and three or fewer amino acid differences as compared to SEQ ID NO: 39; and(ii) a VL that comprises: (a) a CDR-L1 comprising Gln27, Asp28, Ile29, Gly30, Tyr32; and three or fewer amino acid differences as compared to SEQ ID NO: 34, (b) a CDR-L2 comprising a sequence that comprises three or fewer amino acid differences as compared to SEQ ID NO: 35; and (c) a CDR-L3 comprising Ile92, Ile93, and Leu94; and three or fewer amino acid differences as compared to of SEQ ID NO: 36. In certain embodiments, the amino acid differences in CDR-H1, CDR-H2, CDR-L1, CDR-L2, and CDR-L3 are human germline substitutions in which a non-human CDR residue is replaced with a corresponding human germline residue (such as those human germline residues as shown in Tables 3 and 4). In certain embodiments, the antibody or antigen-binding fragment thereof described herein comprises (i) a VH comprising an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 28, and/or (ii) a VL comprising an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 1. Any combination of these VL and VH sequences is also encompassed by the invention. In certain embodiments, the VH framework is DP10. Other similar framework regions are also predicted to deliver advantageous antibodies or antibody fragments of the invention comprising CDRs of SEQ ID NOs. 37, 38, and 39 include: DP-88, DP-25, DP-73, IGHV5-10-1*01, IGHV5-10-1*04, DP-14, DP-75, DP15, DP-8, DP-7 and IGHV7-4-1*02, which share 99%, 93%, 75%, 73%, 73%, 92%, 90%, 90%, 89%, 93%, and 79% sequence identity, respectively, with the FW region of DP10, and comprise four or fewer amino acid differences in the common structural features: (A) residues directly underneath CDR (Vernier Zone), H2, H47, H48, and H49, H67, H69, H71, H73, H93, H94; (B) VH/VL chain packing residues: H37, H39, H45, H47, H91, H93; and (C) canonical CDR Structural support residues H24, H71, H94 (all Kabat numbering). Particularly preferred are framework regions of DP-88, DP-25, DP-73, IGHV5-10-1*01, and IGFV-10-1*04, sharing 99%, 93%, 75%, 73%, and 73% sequence identity with DP10, respectively, and have two or fewer amino acid differences in these common structural features. In certain embodiments, the VL framework is DPK9. Other similar framework regions are also predicted to deliver advantageous antibodies of the invention comprising CDRs of SEQ ID NOs. 34, 35, and 36 include: DPK5, DPK4, DPK1, IGKV1-5*01, DPK24, DPK21, DPK15, IGKV1-13*02, IGKV1-17*01, DPK8, IGKV3-11*01, and DPK22, which share 99%, 97%, 97%, 96%, 80%, 76%, 66%, 97%, 97%, 96%, 76%, and 74% sequence identity, respectively, with the FW region of DPK-9, and comprise one or fewer amino acid difference in common structural features: (A) residues directly underneath CDR (Vernier Zone), L2, L4, L35, L36, L46, L47, L48, L49, L64, L66, L68, L69, L71; (B) VH/VL Chain packing Residues: L36, L38, L44, L46, L87; and (C) canonical CDR Structural support residues L2, L48, L64, L71 (all Kabat numbering). Particularly preferred are framework regions of DPK5, DPK4, DPK1, IGKV1-5*01, DPK24, DPK21, and DPK15, which share 99%, 97%, 97%, 96%, 80%, 76%, and 66% sequence identity with DPK9, respectively, and have no amino acid difference in these common structural features. In certain embodiments, the antibody or antigen-binding fragment thereof described herein comprises (i) a CDR-H1 comprising SEQ ID NO:37, a CDR-H2 comprising SEQ ID NO:38, a CDR-H3 comprising SEQ ID NO:39, a CDR-L1 comprising SEQ ID NO:34; a CDR-L2 comprising SEQ ID NO:35, and a CDR-L3 comprising SEQ ID NO:36; and (ii) a VL framework comprising a sequence that is at least 66%, at least 74%, at least 76%, at least 80%, at least 96%, at least 97%, or at least 99% identical to the framework sequence of human germline DPK9, and a VH framework comprising a sequence that is at least 73%, at least 75%, at least 79%, at least 89%, at least 90%, at least 92%, at least 93%, or at least 99% identical to the framework sequence of human germline DP10. In certain embodiments, the antibody or antigen-binding fragment thereof described herein comprises (i) a CH comprising an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 29; and/or (ii) a CL comprising an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 30. Any combination of these CH and CL sequences is also encompassed by the invention. In certain embodiments, the antibody or antigen-binding fragment thereof described herein comprises an Fc domain. The Fc domain can be derived from IgA (e.g., IgA1or IgA2), IgG, IgE, or IgG (e.g., IgG1, IgG2, IgG3, or IgG4). In certain embodiments, the antibody or antigen-binding fragment thereof described herein comprises (i) a heavy chain comprising an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 33, and/or (ii) a light chain comprising an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 32. Any combination of these heavy chain and light chain sequences is also encompassed by the invention. Additional antibodies (e.g., CTI-AF2 through CTI-AF27), antigen-binding fragments thereof, and antigen-binding variants thereof, are also provided by the invention. CTI-AF2 to CTI-AF27 share the same VH sequence but have different VL sequences. Accordingly, in certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention comprises (i) a VH comprising an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 28, and/or (ii) a VL comprising an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to any of SEQ ID NOs: 2-27. Any combination of these VL and VH sequences is also encompassed by the invention. Also provided by the invention is an antibody, or antigen-binding fragment thereof, that competes for binding to human IFNβ with any of the antibody or antigen-binding fragment thereof described herein, such as any one of the antibodies listed in Table 11, or antigen-binding fragments thereof. For example, if the binding of an antibody, or an antigen-binding portion thereof, to human IFNβ reduces the subsequent binding to human IFNβ by CTI-AF1, the antibody, or an antigen-binding portion thereof, is deemed as competing with CTI-AF1 for human IFNβ binding. Also provided by the invention is an antibody, or antigen-binding fragment thereof, that binds the same epitope of human IFNβ as any antibody, or antigen-binding fragment thereof, described herein, such as any antibody listed in Table 11, or antigen-binding fragments thereof. For example, an antibody competition assay (and overlapping epitope analysis) can be assessed using SPR, as described in detail herein, or any art-recognized competitive binding assay. The SPR binding assay described herein is the preferred, not exclusive method for assessing binding of the antibody of the invention, and any other test antibodies. The antibodies, and antigen-binding fragments thereof, of the invention include monoclonal antibodies, polyclonal antibodies, antibody fragments (e.g., Fab, Fab′, F(ab′)2, Fv, Fc, etc.), chimeric antibodies, bispecific antibodies, heteroconjugate antibodies, single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion, domain antibodies (dAbs), humanized antibodies, and any other configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies. The antibodies and antigen-binding fragments may be murine, rat, human, or any other origin (including chimeric or humanized antibodies). In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a chimeric, humanized or human antibody. In certain embodiments, the antibody is a fully human antibody. In certain embodiments, the antibody is a humanized antibody. The binding affinity of an antibody can be expressed as a KDvalue, which refers to the dissociation rate of a particular antigen-antibody interaction. KDis the ratio of the rate of dissociation, also called the “off-rate (koff)”, to the association rate, or “on-rate (kon)”. Thus, KDequals koff/kon(dissociation/association) and is expressed as a molar concentration (M), and the smaller the KD, the stronger the affinity of binding. KDvalues for antibodies can be determined using methods well established in the art. Unless otherwise specified, “binding affinity” refers to monovalent interactions (intrinsic activity; e.g., binding of an antibody to an antigen through a monovalent interaction). In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention has an affinity (KD) value of not more than about 1×10−7M, such as not more than about 1×10−7M, not more than about 9×10−8M, not more than about 8×10−8M, not more than about 7×10−8M, not more than about 6×10−8M, not more than about 5×10−8M, not more than about 4×10−8M, not more than about 3×10−8M, not more than about 2×10−8M, not more than about 1×10−8M, not more than about 9×10−9M, not more than about 8×10−9M, not more than about 7×10−9M, not more than about 6×10−9M, not more than about 5×10−9M, not more than about 4×10−9M, not more than about 3×10−9M, not more than about 2×10−9M, not more than about 1×10−9M, not more than about 9×10−10M, not more than about 8×10−10M, not more than about 7×10−10M, not more than about 6×10−10M, not more than about 5×10−10M, not more than about 4×10−10M, not more than about 3×10−10M, not more than about 2×10−10M, not more than about 1×10−10M, not more than about 9×10−11M, not more than about 8×10−11M, not more than about 7×10−11M, not more than about 6×10−11M, not more than about 5×10−11M, not more than about 4×10−11M, not more than about 3×10−11M, not more than about 2×10−11M, not more than about 1×10−11M, not more than about 9×10−12M not more than about 8×10−12M, not more than about 7×10−12M, not more than about 6×10−12M, not more than about 5×10−12M, not more than about 4×10−12M, not more than about 3×10−12M, not more than about 2×10−12M, not more than about 1×10−12M, not more than about 9×10−13M, not more than about 8×10−13M, not more than about 7×10−13M, not more than about 6×10−13M, not more than about 5×10−13M, not more than about 4×10−13M, not more than about 3×10−13M, not more than about 2×10−13M, not more than about 1×10−13M, from about 1×10−7M to about 1×10−14M, from about 9×10−8M to about 1×10−14M, from about 8×10−8M to about 1×10−14M, from about 7×10−8M to about 1×10−14M, from about 6×10−8M to about 1×10−14M, from about 5×10−8M to about 1×10−14M, from about 4×10−8M to about 1×10−14M, from about 3×10−8M to about 1×10−14M, from about 2×10−8M to about 1×10−14M, from about 1×10−8M to about 1×10−14M, from about 9×10−9M to about 1×10−14M, from about 8×10−9M to about 1×10−14M, from about 7×10−9M to about 1×10−14M, from about 6×10−9M to about 1×10−14M, from about 5×10−9M to about 1×10−14M, from about 4×10−9M to about 1×10−14M, from about 3×10−9M to about 1×10−14M, from about 2×10−9M to about 1×10−14M, from about 1×10−9M to about 1×10−14M, from about 1×10−7M to about 1×10−13M, from about 9×10−8M to about 1×10−13M, from about 8×10−8M to about 1×10−13M, from about 7×10−8M to about 1×10−13M, from about 6×10−8M to about 1×10−13M, from about 5×10−8M to about 1×10−13M, from about 4×10−8M to about 1×10−13M, from about 3×10−8M to about 1×10−13M, from about 2×10−8M to about 1×10−13M, from about 1×10−8M to about 1×10−13M, from about 9×10−9M to about 1×10−13M, from about 8×10−9M to about 1×10−13M, from about 7×10−9M to about 1×10−13M, from about 6×10−9M to about 1×10−13M, from about 5×10−9M to about 1×10−13M, from about 4×10−9M to about 1×10−13M, from about 3×10−9M to about 1×10−13M, from about 2×10−9M to about 1×10−13M, or from about 1×10−9M to about 1×10−13M. The value of KDcan be determined directly by well-known methods, and can be computed even for complex mixtures by methods such as those, for example, set forth in Caceci et al. (1984, Byte 9: 340-362). For example, the KDmay be established using a double-filter nitrocellulose filter binding assay such as that disclosed by Wong & Lohman (1993, Proc. Natl. Acad. Sci. USA 90: 5428-5432). Other standard assays to evaluate the binding ability of ligands such as antibodies towards target antigens are known in the art, including for example, ELISAs, Western blots, RIAs, and flow cytometry analysis, and other assays exemplified elsewhere herein. One exemplary method for measuring binding affinity (KD) value is surface plasmon resonance (SPR), typically using a biosensor system such as a BIACORE® system. SPR refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIACORE® system. BIAcore kinetic analysis comprises analyzing the binding and dissociation of an antigen from a chip with an immobilized molecule (e.g., a molecule comprising an antigen-binding domain), on their surface; or the dissociation of an antibody, or antigen-binding fragment thereof, from a chip with an immobilized antigen. In certain embodiments, the SPR measurement is conducted using a BIACORE® T100 or T200 instrument. For example, a standard assay condition for surface plasmon resonance can be based on antibody immobilization of approximately 100-500 Response Units (RU) of IgG on the SPR chip. Purified target proteins are diluted in buffer to a range of final concentrations and injected at a requisite flow rate (e.g. 10-100 μl/min) to allow the calculation of Ka. Dissociation is allowed to proceed to establish off-rate, followed by 3 M MgCl2(or 20 mM NaOH) for regeneration of the chip surface. Sensorgrams are then analyzed using a kinetics evaluation software package. In an exemplary embodiment, the SPR assay is according to the conditions as set forth in Example 1. In certain embodiments, the binding affinity (KD) value is measured using solution-based kinetic exclusion assay (KinExA™). In a particular embodiment, the KinExA measurement is conducted using a KinExA™ 3200 instrument (Sapidyne). The Kinetic Exclusion Assay (KinExA™) is a general purpose immunoassay platform (basically a flow spectrofluorimeter) that is capable of measuring equilibrium dissociation constants, and association and dissociation rate constants for antigen/antibody interactions. Since KinExA™ is performed after equilibrium has been obtained it is an advantageous technique to use for measuring the KDof high affinity interactions where the off-rate of the interaction may be very slow. The KinExA™ methodology can be conducted generally as described in Drake et al (2004) Analytical Biochemistry 328, 35-43. Another method for determining the KDof an antibody is by using Bio-Layer Interferometry, typically using OCTET® technology (Octet QKe system, ForteBio). In general, an anti-IFNβ antibody should bind to IFNβ with high affinity, in order to effectively block the activities of IFNβ. IFNβ binds IFNAR1 at a KDof about 50 nM, and to IFNAR2 at a KDof about 100 μM. Accordingly, it is desirable that the IFNβ antibody have binding affinities (KD) in nanomolar and picomolar range, such as about 1×10−9M or lower. Activity Assays In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention is a neutralizing antibody that reduces at least one activity of IFNβ. Such activity of IFNβ includes, but it not limited to, binding to IFNAR, increasing expression of an IFNβ-dependent gene, and/or inducing phosphorylation of, e.g., STAT1, and/or STAT2, among other IFNβ activities known in the art. Whether an antibody, or antigen-binding fragment thereof, reduces an activity of IFNβ can be assessed by a number of assays. For example, assays can be used to determine whether the antibody, or antigen-binding fragment thereof: (a) inhibits the binding of IFNβ to IFNAR; (b) reduces the expression level of an IFNβ-dependent gene; and/or (c) inhibit IFNβ-induced phosphorylation, such as phosphorylation of STAT1, and/or STAT2. In certain embodiments, the antibody, or antigen-binding fragment thereof, inhibits the binding of IFNβ to IFNAR (e.g., can be assessed by competitive binding to IFNβ). For example, an assay may compare (i) the binding of IFNβ to IFNAR in the presence of the antibody, or antigen-binding fragment thereof, with (ii) the binding of IFNβ to IFNAR in the absence of the antibody, or antigen-binding fragment thereof. The reduction in binding of IFNβ to IFNAR can be at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%, in the presence of the anti-IFNβ antibody, or antigen-binding fragment thereof. The expected binding of IFNβ to IFNAR in the absence of the antibody, or antigen-binding fragment thereof, can be set as 100%. In certain embodiments, the antibody, or antigen-binding fragment thereof, inhibits the binding of IFNβ to IFNAR, with a half maximal inhibitory concentration (IC50) of not more than about 1×10−7M, not more than about 1×10−8M, not more than about 1×10−9M, not more than about 1×10−10M, not more than about 1×10−11M, not more than about 1×10−12M, not more than about 1×10−13M, not more than about 1×10−14M, not more than about 1×10−15M, from about 1×10−7M to about 5×10−14M, from about 1×10−7M to about 1×10−14M, from about 1×10−7M to about 5×10−13M, from about 1×10−7M to about 1×10−13M, from about 1×10−7M to about 5×10−12M, or from about 1×10−7M to about 1×10−12M. The activities of an antibody, or antigen-binding fragment thereof, of the invention can also be assessed by measuring the expression level of an IFNβ-dependent gene. For example, the gene can be a downstream component in the IFNβ-mediated signal pathway (such as CMPK2, IFIT1, IF127, IFIH1, IF144, IF144L, IF16, ISG15, LY6E, HERC5, MX1, OAS1, OAS2, OAS3, RSAD2, XAF1, CXCL10, or any combination thereof). Alternatively, the gene can be a reporter gene (e.g., the luciferase reporter gene as used in the examples) where the expression level of the reporter gene correlates with IFNβ activity (e.g., the reporter gene is operably linked to an IFNβ-dependent response element). The expression level of the downstream gene or reporter gene can be assessed by a variety of methods, such as measuring the RNA level, protein level, or activity level of a protein. The assay can compare (i) the expression level of the IFNβ dependent gene in the presence of the antibody, or antigen-binding fragment thereof, with (ii) the expression level of the IFNβ dependent gene in the absence of the antibody, or antigen-binding fragment thereof. The reduction in expression level of a downstream gene or reporter gene can be at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%, in the presence of the anti-IFNβ antibody, or antigen-binding fragment thereof. The baseline expression level in the absence of the antibody, or antigen-binding fragment thereof, can be set as 100%. In certain embodiments, the antibody, or antigen-binding fragment thereof, inhibits the expression of an IFNβ-dependent gene, with a half maximal inhibitory concentration (IC50) of not more than about 1×10−7M, not more than about 1×10−8M, not more than about 1×10−9M, not more than about 1×10−10M, not more than about 1×10−11M, not more than about 1×10−12M, not more than about 1×10−13M, not more than about 1×10−14M, not more than about 1×10−15M, from about 1×10−7M to about 5×10−14M, from about 1×10−7M to about 1×10−14M, from about 1×10−7M to about 5×10−13M, from about 1×10−7M to about 1×10−13M, from about 1×10−7M to about 5×10−12M, or from about 1×10−7M to about 1×10−12M. In certain embodiments, IC50of from about 1×10−10M to about 1×10−13M is preferred. In certain embodiments, IC50of from about 5×10−11M to about 5×10−12M is preferred. The inhibitory activity of an antibody, or antigen-binding fragment thereof, can also be assessed by measuring the level of IFNβ-induced phosphorylation, such as STAT1 phosphorylation, and/or STAT2 phosphorylation level. The assay can compare (i) the phosphorylation level of STAT1 and/or STAT2 in the presence of the antibody, or antigen-binding fragment thereof, with (ii) the phosphorylation level of STAT1 and/or STAT2 in the absence of the antibody, or antigen-binding fragment thereof. The reduction in phosphorylation level can be at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%, in the presence of the anti-IFNβ antibody, or antigen-binding fragment thereof. The baseline STAT1 phosphorylation and/or STAT2 phosphorylation level in the absence of the antibody, or antigen-binding fragment thereof, can be set as 100%. In certain embodiments, the antibody, or antigen-binding fragment thereof, inhibits IFNβ-induced phosphorylation (such as STAT1 phosphorylation, and/or STAT2 phosphorylation), with a half maximal inhibitory concentration (IC50) of not more than about 1×10−7M, not more than about 1×10−8M, not more than about 1×10−9M, not more than about 1×10−10M, not more than about 1×10−11M, not more than about 1×10−12M, not more than about 1×10−13M, not more than about 1×10−14M, not more than about 1×10−15M, from about 1×10−7M to about 5×10−14M, from about 1×10−7M to about 1×10−14M, from about 1×10−7M to about 5×10−13M, from about 1×10−7M to about 1×10−13M, from about 1×10−7M to about 5×10−12M, or from about 1×10−7M to about 1×10−12M. In certain embodiments, IC50of from about 1×10−10M to about 1×10−13M is preferred. In certain embodiments, IC50of from about 5×10−11M to about 5×10−12M is preferred. In certain embodiments, the characteristics of the antibody, or antigen-binding fragment thereof, of the invention is further assessed using other biological activity assays, e.g., in order to evaluate its potency, pharmacological activity, and potential efficacy as a therapeutic agent. Such assays are known in the art and depend on the intended use for the antibody. Examples include e.g., toxicity assays, immunogenicity assays, stability assays, and/or PK/PD profiling. C. Nucleic Acids and Methods of Producing Anti-IFNβ Antibodies The invention also provides polynucleotides encoding any of the antibodies, including antibody portions and modified antibodies described herein. The invention also provides a method of making any of the polynucleotides described herein. Polynucleotides can be made and expressed by procedures known in the art. The sequence of a desired antibody, or antigen-binding fragment thereof, and nucleic acid encoding such antibody, or antigen-binding fragment thereof, can be determined using standard sequencing techniques. A nucleic acid sequence encoding a desired antibody, or antigen-binding fragment thereof, may be inserted into various vectors (such as cloning and expression vectors) for recombinant production and characterization. A nucleic acid encoding the heavy chain, or an antigen-binding fragment of the heavy chain, and a nucleic acid encoding the light chain, or an antigen-binding fragment of the light chain, can be cloned into the same vector, or different vectors. In one aspect, the invention provides polynucleotides encoding the amino acid sequence of any of the following anti-IFNβ antibodies and antigen-binding portions thereof: CTI-AF1, CTI-AF2, CTI-AF3, CTI-AF4, CTI-AFS, CTI-AF6, CTI-AF7, CTI-AF8, CTI-AF9, CTI-AF10, CTI-AF11, CTI-AF12, CTI-AF13, CTI-AF14, CTI-AF15, CTI-AF16, CTI-AF17, CTI-AF18, CTI-AF19, CTI-AF20, CTI-AF21, CTI-AF22, CTI-AF23, CTI-AF24, CTI-AF25, CTI-AF26, and CTI-AF27. The invention also provides polynucleotides encoding an antibody, or antigen-binding fragment thereof, that binds substantial the same epitope as an antibody selected from the group consisting of: CTI-AF1, CTI-AF2, CTI-AF3, CTI-AF4, CTI-AF5, CTI-AF6, CTI-AF7, CTI-AF8, CTI-AF9, CTI-AF10, CTI-AF11, CTI-AF12, CTI-AF13, CTI-AF14, CTI-AF15, CTI-AF16, CTI-AF17, CTI-AF18, CTI-AF19, CTI-AF20, CTI-AF21, CTI-AF22, CTI-AF23, CTI-AF24, CTI-AF25, CTI-AF26, and CTI-AF27. The invention also provides polynucleotides encoding an antibody, or antigen-binding fragment thereof, that competes for binding to IFNβ with an antibody selected from the group consisting of: CTI-AF1, CTI-AF2, CTI-AF3, CTI-AF4, CTI-AF5, CTI-AF6, CTI-AF7, CTI-AF8, CTI-AF9, CTI-AF10, CTI-AF11, CTI-AF12, CTI-AF13, CTI-AF14, CTI-AF15, CTI-AF16, CTI-AF17, CTI-AF18, CTI-AF19, CTI-AF20, CTI-AF21, CTI-AF22, CTI-AF23, CTI-AF24, CTI-AF25, CTI-AF26, and CTI-AF27. The invention also provides polynucleotides comprising a sequence encoding a protein comprising the amino acid sequence selected from the group consisting of: (i) SEQ ID NOs:1-27, (ii) SEQ ID NO:28, and (iii) any combination thereof. The invention also provides polynucleotides comprising the nucleic acid sequence set forth as SEQ ID NOs: 166 or 167. The invention also provides polynucleotides comprising the nucleic acid sequence of the DNA insert of the plasmid deposited with the ATCC and having Accession No. PTA-122727 or the DNA insert of the plasmid deposited with the ATCC and having Accession No. PTA-122726. In another aspect, the invention provides polynucleotides and variants thereof encoding an anti-IFNβ antibody, wherein such variant polynucleotides share at least 70%, at least 75%, at least 80%, at least 85%, at least 87%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to any of the specific nucleic acid sequences disclosed herein. These amounts are not meant to be limiting, and increments between the recited percentages are specifically envisioned as part of the disclosure. In one embodiment, the VH and VL domains, or antigen-binding portion thereof, or full length HC or LC, are encoded by separate polynucleotides. Alternatively, both VH and VL, or antigen-binding portion thereof, or HC and LC, are encoded by a single polynucleotide. Polynucleotides complementary to any such sequences are also encompassed by the present disclosure. Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules. RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present disclosure, and a polynucleotide may, but need not, be linked to other molecules and/or support materials. Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence. Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule. The effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein. In some embodiments, variants exhibit at least about 70% identity, in some embodiments, at least about 80% identity, in some embodiments, at least about 90% identity, and in some embodiments, at least about 95% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof. These amounts are not meant to be limiting, and increments between the recited percentages are specifically envisioned as part of the disclosure. Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A “comparison window” as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, or 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Optimal alignment of sequences for comparison may be conducted using the MegAlign® program in the Lasergene® suite of bioinformatics software (DNASTAR®, Inc., Madison, WI), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M. O., 1978, A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins, D. G. and Sharp, P. M., 1989, CABIOS 5:151-153; Myers, E. W. and Muller W., 1988, CABIOS 4:11-17; Robinson, E. D., 1971, Comb. Theor. 11:105; Santou, N., Nes, M., 1987, Mol. Biol. Evol. 4:406-425; Sneath, P. H. A. and Sokal, R. R., 1973, Numerical Taxonomy the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, CA; Wilbur, W. J. and Lipman, D. J., 1983, Proc. Natl. Acad. Sci. USA 80:726-730. In some embodiments, the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity. Variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof. Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementary sequence). Suitable “moderately stringent conditions” include prewashing in a solution of 5×SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50° C.-65° C., 5×SSC, overnight; followed by washing twice at 65° C. for 20 minutes with each of 2×, 0.5× and 0.2×SSC containing 0.1% SDS. As used herein, “highly stringent conditions” or “high stringency conditions” are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5×SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5×Denhardt's solution, sonicated salmon sperm DNA (50 μg/mL), 0.1% SDS, and 10% dextran sulfate at 42° C., with washes at 42° C. in 0.2×SSC (sodium chloride/sodium citrate) and 50% formamide at 55° C., followed by a high-stringency wash consisting of 0.1×SSC containing EDTA at 55° C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like. It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present disclosure. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present disclosure. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison). The polynucleotides of this disclosure can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence. For preparing polynucleotides using recombinant methods, a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein. Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome. The polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al., 1989. Alternatively, PCR allows reproduction of DNA sequences. PCR technology is well known in the art and is described in U.S. Pat. Nos. 4,683,195, 4,800,159, 4,754,065 and 4,683,202, as well as PCR: The Polymerase Chain Reaction, Mullis et al. eds., Birkauswer Press, Boston, 1994. RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., 1989, for example. Suitable cloning and expression vectors can include a variety of components, such as promoter, enhancer, and other transcriptional regulatory sequences. The vector may also be constructed to allow for subsequent cloning of an antibody variable domain into different vectors. Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector. Suitable examples include plasmids and bacterial viruses, e.g., pUC18, pUC19, Bluescript (e.g., pBS SK+) and its derivatives, mp18, mp19, pBR322, μMB9, ColE1, pCR1, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28. These and many other cloning vectors are available from commercial vendors such as BioRad, Strategene, and Invitrogen. Expression vectors are further provided. Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the disclosure. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462. Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons. The vectors containing the polynucleotides of interest and/or the polynucleotides themselves, can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus). The choice of introducing vectors or polynucleotides will often depend on features of the host cell. The antibody, or antigen-binding fragment thereof, may be made recombinantly using a suitable host cell. A nucleic acid encoding the antibody or antigen-binding fragment thereof can be cloned into an expression vector, which can then be introduced into a host cell, such asE. colicell, a yeast cell, an insect cell, a simian COS cell, a Chinese hamster ovary (CHO) cell, or a myeloma cell where the cell does not otherwise produce an immunoglobulin protein, to obtain the synthesis of an antibody in the recombinant host cell. Preferred host cells include a CHO cell, a Human embryonic kidney (HEK) 293 cell, or an Sp2.0 cell, among many cells well-known in the art. An antibody fragment can be produced by proteolytic or other degradation of a full-length antibody, by recombinant methods, or by chemical synthesis. A polypeptide fragment of an antibody, especially shorter polypeptides up to about 50 amino acids, can be conveniently made by chemical synthesis. Methods of chemical synthesis for proteins and peptides are known in the art and are commercially available. The antibody, or antigen-binding fragment thereof, of the invention may be affinity matured. For example, an affinity matured antibody can be produced by procedures known in the art (Marks et al., 1992, Bio/Technology, 10:779-783; Barbas et al., 1994, Proc Nat. Acad. Sci, USA 91:3809-3813; Schier et al., 1995, Gene, 169:147-155; Yelton et al., 1995, J. Immunol., 155:1994-2004; Jackson et al., 1995, J. Immunol., 154(7):3310-9; Hawkins et al., 1992, J. Mol. Biol., 226:889-896; and WO2004/058184). 2. Formulations and Uses The antibody, or antigen-binding fragment thereof, of the invention can be formulated as a pharmaceutical composition. The pharmaceutical composition may further comprise a pharmaceutically acceptable carrier, excipient, and/or stabilizer (Remington: The Science and practice of Pharmacy 20th Ed., 2000, Lippincott Williams and Wilkins, Ed. K. E. Hoover), in the form of lyophilized formulation or aqueous solution. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations, and may comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrans; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). Pharmaceutically acceptable excipients are further described herein. The antibody, or antigen-binding fragment thereof, of the invention can be used for various therapeutic or diagnostic purposes. For example, the antibody, or antigen-binding fragment thereof, of the invention may be used as an affinity purification agents (e.g., for in vitro purification of IFNβ), as a diagnostic agent (e.g., for detecting expression of IFNβ in specific cells, tissues, or serum). Exemplary therapeutic uses of the antibody, or antigen-binding fragment thereof, of the invention include treating a rheumatic disease (such as SLE or DM) or an interferonopathy. The antibody, or antigen-binding fragment thereof, of the invention may also be used in prophylactic treatment (e.g., administering to a subject who has not exhibited a disease symptom but is susceptible to a rheumatic disease or an interferonopathy). For therapeutic applications, the antibody, or antigen-binding fragment thereof, of the invention can be administered to a mammal, especially a human by conventional techniques, such as intravenously (as a bolus or by continuous infusion over a period of time), intramuscularly, intraperitoneally, intra-cerebrospinally, subcutaneously, intra-articularly, intrasynovially, intrathecally, orally, topically, or by inhalation. The antibody, or antigen-binding fragment thereof, of the invention also is suitably administered by intra-tumoral, peri-tumoral, intra-lesional, or peri-lesional routes. Accordingly, in one aspect, the invention provides a method of reducing the activity of IFNβ, comprising administering to a subject (e.g., a human) in need thereof a therapeutically effective amount of the antibody, or antigen-binding fragment thereof, of the invention. In certain embodiments, the subject suffers from or is susceptible to a rheumatic disease. In certain embodiments, the rheumatic disease is SLE. In certain embodiments, the rheumatic disease is DM. In certain embodiments, the subject suffers from or is susceptible to an interferonopathy. In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention is administered subcutaneously. In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention is administered intravenously. The pharmaceutical compositions may be administered to a subject in need thereof at a frequency that may vary with the severity of the rheumatic disease or interferonopathy. In the case of prophylactic therapy, the frequency may vary depending on the subject's susceptibility or predisposition to a rheumatic disease or an interferonopathy. The compositions may be administered to patients in need as a bolus or by continuous infusion. For example, a bolus administration of an antibody present as a Fab fragment may be in an amount of from 0.0025 to 100 mg/kg body weight, 0.025 to 0.25 mg/kg, 0.010 to 0.10 mg/kg or 0.10-0.50 mg/kg. For continuous infusion, an antibody present as an Fab fragment may be administered at 0.001 to 100 mg/kg body weight/minute, 0.0125 to 1.25 mg/kg/min, 0.010 to 0.75 mg/kg/min, 0.010 to 1.0 mg/kg/min. or 0.10-0.50 mg/kg/min fora period of 1-24 hours, 1-12 hours, 2-12 hours, 6-12 hours, 2-8 hours, or 1-2 hours. For administration of an antibody present as a full-length antibody (with full constant regions), dosage amounts may be from about 1 mg/kg to about 10 mg/kg, from about 2 mg/kg to about 10 mg/kg, from about 3 mg/kg to about 10 mg/kg, from about 4 mg/kg to about 10 mg/kg, from about 5 mg/kg to about 10 mg/kg, from about 1 mg/kg to about 20 mg/kg, from about 2 mg/kg to about 20 mg/kg, from about 3 mg/kg to about 20 mg/kg, from about 4 mg/kg to about 20 mg/kg, from about 5 mg/kg to about 20 mg/kg, about 1 mg/kg or more, about 2 mg/kg or more, about 3 mg/kg or more, about 4 mg/kg or more, about 5 mg/kg or more, about 6 mg/kg or more, about 7 mg/kg or more, about 8 mg/kg or more, about 9 mg/kg or more, about 10 mg/kg or more, about 11 mg/kg or more, about 12 mg/kg or more, about 13 mg/kg or more, about 14 mg/kg or more, about 15 mg/kg or more, about 16 mg/kg or more, about 17 mg/kg or more, about 19 mg/kg or more, or about 20 mg/kg or more. The frequency of the administration would depend upon the severity of the condition. Frequency could range from three times per week to once every two or three weeks. Additionally, the compositions may be administered to patients via subcutaneous injection. For example, a dose of 1 to 100 mg anti-IFNβ antibody can be administered to patients via subcutaneous or intravenous injection administered twice a week, once a week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, once every six weeks, once every seven weeks, once every eight weeks, once every nine weeks, once every ten weeks, twice a month, once a month, once every two months, or once every three months. For example, antibody CTI-AF1 has an estimated half-life of about 19 days. This half-life supports subcutaneous or intravenous injection at every 2-6 weeks, such as once every 2 weeks or once every 4 weeks. In certain embodiments, the half-life of the anti-IFNβ antibody in human is about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, about 14 days, about 15 days, about 16 days, about 17 days, about 18 days, about 19 days, about 20 days, about 21 days, about 22 days, about 23 days, about 24 days, about 25 days, about 26 days, about 27 days, about 28 days, about 29 days, about 30 days, from about 5 days to about 40 days, from about 5 days to about 35 days, from about 5 days to about 30 days, from about 5 days to about 25 days, from about 10 days to about 40 days, from about 10 days to about 35 days, from about 10 days to about 30 days, from about 10 days to about 25 days, from about 15 days to about 40 days, from about 15 days to about 35 days, from about 15 days to about 30 days, or from about 15 days to about 25 days, In certain embodiments, the pharmaceutical composition is administered subcutaneously or intravenously at every 2-6 weeks, with a dose from about 0.1 mg/kg to about 10 mg/kg, from about 0.5 mg/kg to about 10 mg/kg, from about 1 mg/kg to about 10 mg/kg, from about 1.5 mg/kg to about 10 mg/kg, from about 2 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 8 mg/kg, from about 0.5 mg/kg to about 8 mg/kg, from about 1 mg/kg to about 8 mg/kg, from about 1.5 mg/kg to about 8 mg/kg, from about 2 mg/kg to about 8 mg/kg, from about 0.1 mg/kg to about 5 mg/kg, from about 0.5 mg/kg to about 5 mg/kg, from about 1 mg/kg to about 5 mg/kg, from about 1.5 mg/kg to about 5 mg/kg, from about 2 mg/kg to about 5 mg/kg, about 0.5 mg/kg, about 1.0 mg/kg, about 1.5 mg/kg, about 2.0 mg/kg, about 2.5 mg/kg, about 3.0 mg/kg, about 3.5 mg/kg, about 4.0 mg/kg, about 4.5 mg/kg, about 5.0 mg/kg, about 5.5 mg/kg, about 6.0 mg/kg, about 6.5 mg/kg, about 7.0 mg/kg, about 7.5 mg/kg, about 8.0 mg/kg, about 8.5 mg/kg, about 9.0 mg/kg, about 9.5 mg/kg, or about 10.0 mg/kg. In certain embodiments, the pharmaceutical composition is administered subcutaneously or intravenously at every 2-6 weeks, with a dose of about 2.0 mg/kg. In certain embodiments, the pharmaceutical composition is administered subcutaneous or intravenously every 2-6 weeks, with a dose of from about 2.0 mg/kg to about 10.0 mg/kg. In one exemplary embodiment, pharmaceutical composition is administered subcutaneously every 2 weeks. The antibody, or antigen-binding fragment thereof, of the invention can be used as monotherapy or in combination with other therapies to treat a rheumatic disease. 3. Definitions Unless otherwise defined herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. Generally, nomenclatures used in connection with, and techniques of, cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well-known and commonly used in the art. An “antigen-binding fragment” of an antibody refers to a fragment of a full-length antibody that retains the ability to specifically bind to an antigen (preferably with substantially the same binding affinity). Examples of an antigen-binding fragment includes (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR), disulfide-linked Fvs (dsFv), and anti-idiotypic (anti-Id) antibody and intrabody. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv)); see e.g., Bird et al. Science 242:423-426 (1988) and Huston et al., Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988)). Other forms of single chain antibodies, such as diabodies, are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen-binding sites (see e.g., Holliger et al. Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993); Poljak et al., 1994, Structure 2:1121-1123). An antibody “variable domain” refers to the variable region of the antibody light chain (VL) or the variable region of the antibody heavy chain (VH), either alone or in combination. As known in the art, the variable regions of the heavy and light chains each consist of three complementarity determining regions (CDRs), and connected by four framework regions (FR),and contribute to the formation of the antigen-binding site of antibodies. Residues in a variable domain are numbered according Kabat, which is a numbering system used for heavy chain variable domains or light chain variable domains of the compilation of antibodies. See, Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991)). Using this numbering system, the actual linear amino acid sequence may contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FR or CDR of the variable domain. For example, a heavy chain variable domain may include a single amino acid insert (residue 52a according to Kabat) after residue 52 of CDR-H2 and inserted residues (e.g. residues 82a, 82b, and 82c, according to Kabat) after heavy chain FR residue 82. The Kabat numbering of residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a “standard” Kabat numbered sequence. Various algorithms for assigning Kabat numbering are available. The algorithm implemented in the 2012 release of Abysis (www.abysis.org) is used herein to assign Kabat numbering to variable regions unless otherwise noted. Specific amino acid residue positions in an antibody (such as paratope residues) are also numbered according to Kabat. “Complementarity Determining Regions” (CDRs) can be identified according to the definitions of the Kabat, Chothia, the accumulation of both Kabat and Chothia, AbM, contact, and/or conformational definitions or any method of CDR determination well known in the art. See, e.g., Kabat et al., 1991, Sequences of Proteins of Immunological Interest, 5th ed. (hypervariable regions); Chothia et al., 1989, Nature 342:877-883 (structural loop structures). AbM definition of CDRs is a compromise between Kabat and Chothia and uses Oxford Molecular's AbM antibody modeling software (ACCELRYS®).The “contact” definition of CDRs is based on observed antigen contacts, set forth in MacCallum et al., 1996, J. Mol. Biol., 262:732-745. The “conformational” definition of CDRs is based on residues that make enthalpic contributions to antigen binding (see, e.g., Makabe et al., 2008, Journal of Biological Chemistry, 283:1156-1166). Still other CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. As used herein, a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches. In the Examples (see Table 11), the CDRs are defined as follows (numbering according to Kabat; H: heavy chain; L: light chain):CDR-H1: H26-H35B; CDR-H2: H50-H65; CDR-H3: H95-H102CDR-L1: L24-L34; CDR-L2: L50-L56; CDR-L3: L89-L97 “Framework” (FR) residues are antibody variable domain residues other than the CDR residues. A VH or VL domain framework comprises four framework sub-regions, FR1, FR2, FR3 and FR4, interspersed with CDRs in the following structure: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4. In the Examples (see Table 11), FR residues include the following (numbering according to Kabat; H: heavy chain; L: light chain): TABLE 5FR1FR2FR3FR4Heavy ChainH1-H25H36-H49H66-H94H103-H113Light ChainL1-L23L35-L49L57-L88L98-L107 An “epitope” refers to the area or region of an antigen (Ag) to which an antibody specifically binds, e.g., an area or region comprising amino acid residues that interact with the antibody (Ab). Epitopes can be linear or non-linear (e.g., conformational). An antibody, or antigen-binding fragment thereof, binds substantially the same epitope as another antibody, or antigen-binding fragment thereof, when binding of the corresponding antibodies, or antigen-binding fragments thereof, are mutually exclusive. That is, binding of one antibody, or antigen-binding fragment thereof, excludes simultaneous or consecutive binding of the other antibody, or antigen-binding fragment thereof. Epitopes are said to be unique, or not substantially the same, if the antigen is able to accommodate binding of both corresponding antibodies, or antigen-binding fragments thereof, simultaneously. The term “paratope” is derived from the above definition of “epitope” by reversing the perspective, and refers to the area or region of an antibody molecule which is involved in binding of an antigen, e.g., an area or region comprising residues that interacts with the antigen. A paratope may be linear or conformational (such as discontinuous residues in CDRs). The epitope/paratope for a given antibody/antigen binding pair can be defined and characterized at different levels of detail using a variety of experimental and computational epitope mapping methods. The experimental methods include mutagenesis, X-ray crystallography, Nuclear Magnetic Resonance (NMR) spectroscopy, Hydrogen/deuterium exchange Mass Spectrometry (HX-MS) and various competition binding methods. As each method relies on a unique principle, the description of an epitope is intimately linked to the method by which it has been determined. Thus, the epitope/paratope for a given antibody/antigen pair will be defined differently depending on the mapping method employed. At its most detailed level, the epitope/paratope for the interaction between an antibody (Ab) and antigen (Ag) can be defined by the spatial coordinates defining the atomic contacts present in the Ag-Ab interaction, as well as information about their relative contributions to the binding thermodynamics. At one level, an epitope/paratope residue can be characterized by the spatial coordinates defining the atomic contacts between the Ag and Ab. In one aspect, the epitope/paratope residue can be defined by a specific criterion, e.g., distance between atoms in the Ab and the Ag (e.g., a distance of equal to or less than about 4 Å (such as 3.8 Å used in the Examples here) from a heavy atom of the cognate antibody and a heavy atom of the antigen. In another aspect, an epitope/paratope residue can be characterized as participating in a hydrogen bond interaction with the cognate antibody/antigen, or with a water molecule that is also hydrogen bonded to the cognate antibody/antigen (water-mediated hydrogen bonding). In another aspect, an epitope/paratope residue can be characterized as forming a salt bridge with a residue of the cognate antibody/antigen. In yet another aspect, an epitope/paratope residue can be characterized as a residue having a non-zero change in buried surface area (BSA) due to interaction with the cognate antibody/antigen. At a less detailed level, epitope/paratope can be characterized through function, e.g., by competition binding with other Abs. The epitope/paratope can also be defined more generically as comprising amino acid residues for which substitution by another amino acid will alter the characteristics of the interaction between the Ab and Ag (e.g. alanine scanning). In the context of an X-ray derived crystal structure defined by spatial coordinates of a complex between an antibody, e.g., a Fab fragment or two Fab fragments, and its antigen, unless otherwise specified, an epitope residue refers to an IFNβ residue (i) having a heavy atom (i.e., a non-hydrogen atom) that is within a distance of about 4 Å (e.g., 3.8 Å) from a heavy atom of the cognate antibody; (ii) participating in a hydrogen bond with a residue of the cognate antibody, or with a water molecule that is also hydrogen bonded to the cognate antibody (water-mediated hydrogen bonding), (iii) participating in a salt bridge to a residue of the cognate antibody, and/or (iv) having a non-zero change in buried surface area (BSA) due to interaction with the cognate antibody. In general, a cutoff is imposed for BSA to avoid inclusion of residues that have minimal interactions. Therefore, unless otherwise specified, epitope residues under category (iv) are selected if it has a BSA of 20 Å2or greater, or is involved in electrostatic interactions when the antibody binds to IFNβ. Similarly, in the context of an X-ray derived crystal structure, unless otherwise specified or contradicted by context, a paratope residue, refers to an antibody residue (i) having a heavy atom (i.e., a non-hydrogen atom) that is within a distance of about 4 Å from a heavy atom of IFNβ, (ii) participating in a hydrogen bond with an IFNβ residue, or with a water molecule that is also hydrogen bonded to IFNβ (water-mediated hydrogen bonding), (iii) participating in a salt bridge to a residue of IFNβ, and/or (iv) having a non-zero change in buried surface area due to interaction with IFNβ. Again, unless otherwise specified, paratope residues under category (iv) are selected if it has a BSA of 20 Å2or greater, or is involved in electrostatic interactions when antibody binds to IFNβ. Residues identified by (i) distance or (iv) BSA are often referred to as “contact” residues. From the fact that descriptions and definitions of epitopes, dependent on the epitope mapping method used, and obtained at different levels of detail, it follows that comparison of epitopes for different Abs on the same Ag can similarly be conducted at different levels of detail. For example, epitopes described on the amino acid level, e.g., determined from an X-ray structure, are said to be identical if they contain the same set of amino acid residues. Epitopes characterized by competition binding are said to be overlapping if the binding of the corresponding antibodies are mutually exclusive, i.e., binding of one antibody excludes simultaneous or consecutive binding of the other antibody; and epitopes are said to be separate (unique) if the antigen is able to accommodate binding of both corresponding antibodies simultaneously. The epitope and paratope for a given antibody/antigen pair may be identified by routine methods. For example, the general location of an epitope may be determined by assessing the ability of an antibody to bind to different fragments or variant IFNβ polypeptides as more fully described previously elsewhere herein. Specific residues within IFNβ that make contact with specific residues within an antibody may also be determined using routine methods, such as those described in the examples. For example, antibody/antigen complex may be crystallized. The crystal structure may be determined and used to identify specific sites of interaction between the antibody and antigen. The terms “specifically binds” and “specific binding” are terms well-understood in the art, and methods to determine such specific binding are also well known in the art. A molecule is said to exhibit “specific binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance, than it does with alternative cells or substances. An antibody, or antigen-binding fragment thereof, “specifically binds” to a target (e.g., IFNβ) if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds other substances. For example, an antibody, or antigen-binding fragment thereof, that specifically binds IFNβ is an antibody that binds its cognate antigen (IFNβ) with greater affinity, avidity, more readily, and/or with greater duration than it binds other antigens, such as other members of the IFN superfamily (e.g., INFα, IFNγ, IFNω), or other unrelated molecules. For example, an anti-IFNβ antibody can specifically binds human IFNβ in a sample, but does not substantially recognize or bind other molecules in the sample under a standard binding assay condition. It is also understood that an antibody, or antigen-binding fragment thereof, which specifically binds a first target may or may not specifically bind to a second target. As such, “specific binding” does not necessarily require (although it can include) exclusive binding. Generally, but not necessarily, reference to “binding” means specific binding. A variety of assay formats may be used to select an antibody, or antigen-binding fragment thereof, that specifically binds a molecule of interest. For example, solid-phase ELISA immunoassay, immunoprecipitation, BIACORE™ (GE Healthcare), KinExA, fluorescence-activated cell sorting (FACS), OCTET™ (FortéBio, Inc.) and Western blot analysis are among many assays that may be used to identify an antibody, or antigen-binding fragment thereof, that specifically binds an antigen. Typically, a specific binding will be at least twice of the background signal or noise, more typically at least 10 times of background, at least 50 times of background, at least 100 times of background, at least 500 times of background, at least 1000 of times background, or at least 10,000 times of background. The specificity of an antibody binding may be assessed by determining and comparing the KDvalues of a specific binding between an antibody and IFNβ, with the KDvalue of a control antibody that is known not to bind to IFNβ. In general, an antibody is said to “specifically bind” an antigen when the KDis about ×10−5M or less. An antibody, or antigen-binding fragment thereof, “does not substantially bind” to an antigen when it does not bind to said antigen with greater affinity, avidity, more readily, and/or with greater duration than it binds other antigens. Typically, the binding will be no greater than twice of the background signal or noise. In general, it binds the antigen with a KDof 1×10−4M or more, 1×10−3M or more, 1×10−2M or more, or 1×10−1M or more. The term “compete”, as used herein with regard to an antibody, means that binding of a first antibody, or an antigen-binding portion thereof, to an antigen reduces the subsequent binding of the same antigen by a second antibody or an antigen-binding portion thereof. In general, binding of a first antibody creates steric hindrance, conformational change, or binding to a common epitope (or portion thereof), such that the binding of the second antibody to the same antigen is reduced. Standard competitive binding assays may be used to determine whether two antibodies compete with each other. One suitable assay for antibody competition involves the use of the Biacore technology, which can measure the extent of interactions using surface plasmon resonance (SPR) technology, typically using a biosensor system (such as a BIACORE® system). For example, SPR can be used in an in vitro competitive binding inhibition assay to determine the ability of one antibody to inhibit the binding of a second antibody. Another assay for measuring antibody competition uses an ELISA-based approach. Furthermore, a high throughput process for “binning” antibodies based upon their competition is described in WO2003/48731. Competition is present if one antibody, or antigen-binding fragment thereof, reduces the binding of another antibody, or antigen-binding fragment thereof, to IFNβ. For example, a sequential binding competition assay may be used, with different antibodies being added sequentially. The first antibody may be added to reach binding that is close to saturation. Then, the second antibody is added. If the binding of second antibody to IFNβ is not detected, or is significantly reduced (e.g., at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90% reduction) as compared to a parallel assay in the absence of the first antibody (which value can be set as 100%), the two antibodies are considered as competing with each other. An exemplary antibody competition assay (and overlapping epitope analysis) by SPR is provided in Example 1. A competitive binding assay can also be conducted in which the binding of the antibody to the antigen is compared to the binding of the target by another binding partner of that target, such as another antibody or a soluble receptor that otherwise binds the target. The concentration at which 50% inhibition occurs is known as the Ki. Under ideal conditions, the Kiis equivalent to KD. Thus, in general, measurement of Kican conveniently be substituted to provide an upper limit for KD. Binding affinities associated with different molecular interactions, e.g., comparison of the binding affinity of different antibodies for a given antigen, may be compared by comparison of the KDvalues for the individual antibody/antigen complexes. KDvalues for antibodies or other binding partners can be determined using methods well established in the art. An “Fc fusion” protein is a protein wherein one or more polypeptides are operably linked to an Fc polypeptide. An Fc fusion combines the Fc region of an immunoglobulin with a fusion partner. The “Fc region” may be a native sequence Fc region or a variant Fc region. Although the boundaries of the Fc region of an immunoglobulin heavy chain might vary, the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof. The numbering of the residues in the Fc region is that of the EU index as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991. The Fc region of an immunoglobulin generally comprises two constant domains, CH2and CH3. As is known in the art, an Fc region can be present in dimer or monomeric form. The term “therapeutically effective amount” means an amount of an anti-IFNβ antibody, or an antigen-binding fragment thereof, or a combination comprising such antibody, or antigen-binding fragment thereof, that is of sufficient quantity to achieve the intended purpose, such as decreased binding of IFNβ to IFNAR, the decreased phosphorylation of STAT1 and/or STAT2, the decreased expression of IFNβ-dependent gene, or otherwise causing a measurable benefit in vivo to a subject in need. The precise amount will depend upon numerous factors, including, but not limited to the components and physical characteristics of the therapeutic composition, intended patient population, individual patient considerations, and the like, and can be determined by one skilled in the art. The term “treatment” includes prophylactic and/or therapeutic treatments. If it is administered prior to clinical manifestation of a disease, disorder, or condition, the treatment is considered prophylactic. Therapeutic treatment includes, e.g., ameliorating or reducing the severity of a disease, disorder, or condition, or shortening the length of the disease, disorder, or condition. Preferably, the disease, disorder, or condition is mediated by or related to IFNβ binding to IFNAR. The term “about”, as used herein, refers to +/−10% of a value. Biological Deposit Representative materials of the present invention were deposited in the American Type Culture Collection, 10801 University Boulevard, Manassas, VA 20110-2209, USA, on Dec. 18, 2015. Vector CTI-AF1-VH, having ATCC Accession No. PTA-122727, comprises a DNA insert encoding the heavy chain variable region of antibody CTI-AF1, and vector CTI-AF1-VL, having ATCC Accession No. PTA-122726, comprises a DNA insert encoding the light chain variable region of antibody CTI-AF1. The deposits were made under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and Regulations thereunder (Budapest Treaty). This assures maintenance of a viable culture of the deposit for 30 years from the date of deposit. The deposit will be made available by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Pfizer Inc. and ATCC, which assures permanent and unrestricted availability of the progeny of the culture of the deposit to the public upon issuance of the pertinent U.S. patent or upon laying open to the public of any U. S. or foreign patent application, whichever comes first, and assures availability of the progeny to one determined by the U. S. Commissioner of Patents and Trademarks to be entitled thereto according to 35 U.S.C. Section 122 and the Commissioner's rules pursuant thereto (including 37 C.F.R. Section 1.14 with particular reference to 886 OG 638). The owner of the present application has agreed that if a culture of the materials on deposit should die or be lost or destroyed when cultivated under suitable conditions, the materials will be promptly replaced on notification with another of the same. Availability of the deposited material is not to be construed as a license to practice the invention in contravention of the rights granted under the authority of any government in accordance with its patent laws. EXAMPLES The invention is further described in detail by reference to the following experimental examples. These examples are provided for purposes of illustration only, and are not intended to be limiting unless otherwise specified. Thus, the invention should in no way be construed as being limited to the following examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein. Example 1. Generation Of Anti-IFNβ Antibodies Antibody CTI-AF1 is a humanized IgG1 antibody against the soluble cytokine interferon beta (IFNβ). A mouse monoclonal antibody (mouse mAb) against human IFNβ was generated by standard immunizations of female BALB/c mice with human IFNβ, and subsequent hybridoma screening. Two hybridoma clones were selected for humanization based on kinetic binding profile. The clones showed a KDvalue of about 20 nM and an IC50of about 20 nM. Hybridoma clones were humanized by using human germline frameworks sequences from IGKV1-39 (DPK9 light chain variable domain; Gene Bank Accession No. X59315) and IGHV1-69 (DP10 heavy chain variable domain; Gene Bank Accession No. L22582). Multiple rounds of affinity maturation were used to increase the affinity of the antibody. The sequences of VL region of these antibodies are shown in Table 11. All antibodies in Table 11 have the same VH sequence. In particular, CTI-AF1 showed a decrease in KDvalue from 25 nM to 29 μM by introducing the following mutations in the light chain variable domain: S to G mutation in position 30, H to I and T to I mutations at position 92 and 93 respectively, and L to I mutation in position 96. No mutations were introduced in the heavy chain variable domain. The affinities of CTI-AF antibodies to human interferon beta (IFNβ) were determined by SPR as follows, using a Biacore T200 instrument. Antibodies were directly immobilized on the surface of a CM5 sensor chip at room temperature, using standard amine-coupling technique. Immobilization levels covered a range from 49 to 375 resonance units (RU). The analyte, recombinant human IFNβ, was then injected in a series of dilutions ranging from 10 nM down to 0.078 nM (2-fold dilution), at a flow rate of 30 to 50 μL per minute for an association time ranging from 65 to 300 seconds, followed by a dissociation phase of 10 minutes. Each concentration was evaluated in duplicate. The analyte was removed by regeneration of the CM5 sensor chip surface between each cycle using 3 M MgCl2at pH 3.0 or 10 mM glycine-HCl at pH 1.5, followed by a buffer rinse. This regeneration step removed the bound analyte and returned the response signal to baseline. Data from the reference flow cell (without analyte) were subtracted from the antigen binding responses to remove systematic artifacts. The apparent binding affinity was determined with a 1:1 interaction model using Biacore T200 evaluation software version 2.0. The equilibrium constant KDwas determined as the ratio of the kinetic rate constants, kd/ka. Binding was validated by repeating the binding experiments over multiple days, using two separate instruments and different flow cells on the CM5 sensor chip. The results are shown in Table 6. TABLE 6summaries of biological activities of the antibodies in Table 11ResponseIC50 (pM)IC50 (pM)KD(M)—rankISREpSTAT1Ab Namebiacore(Octet)NeutralizationInhibitionCTI-AF13.6E−11124CTI-AF2————CTI-AF3————CTI-AF4—4——CTI-AF5———10CTI-AF6————CTI-AF7————CTI-AF8—6460—CTI-AF9—1275—CTI-AF10—5——CTI-AF11————CTI-AF12————CTI-AF13————CTI-AF14—7—30CTI-AF15—3—80CTI-AF16—214—CTI-AF17————CTI-AF18————CTI-AF19————CTI-AF203.35E−108—20CTI-AF21—11——CTI-AF22————CTI-AF23—9——CTI-AF24————CTI-AF25————CTI-AF26————CTI-AF27—10—70 Example 2. Biophysical Properties of Anti-IFNβ Antibodies CTI-AF1 was dialyzed and concentrated to 150 mg/mL in MOD1 buffer with 10K MWCO regenerated cellulose membrane. The cynomolgus monkey ETS material was ultrafiltrated/diafiltrated into the same buffer to a final concentration of 72 mg/mL with minimal losses of product. When formulated in PBS, pH 7.2 at ˜50 mg/mL, CTI-AF1 phase-separated at 2-8° C. and formed a stable milky emulsion. Upon warming up to room temperature, the solution becomes clear again. In MOD1 buffer, no phase-separation occurred. Viscosity was measured at 22° C. using the mVROC viscometer. Injections were performed at 100 μL/min using a 100 μL Hamilton syringe. The dependence of viscosity on concentration is shown inFIG.1. Even at the maximum concentration the viscosity is still below 10 cP. Thermal stability was evaluated using MicroCal VP-DSC (Malvern). CTI-AF1 was scanned at 1 mg/mL protein in MOD1 buffer at 1 deg/min. As shown inFIG.2, the first melting transition of this molecule occurs at 69.4° C., which is well above the known required stability threshold for commercial scale manufacturability. Low-pH stability was evaluated by titrating protein A pool with citric acid down to pH 2.8, 3.0 and 3.4 and incubating for 5 hours at room temperature before neutralizing to pH 7.0. As shown inFIG.3, the formation of HMMS occurs only at pH 2.8, while at higher pH levels the product is stable. This stability will enable inactivation of enveloped viruses at low pH, as required for commercial manufacture. Freeze/thaw stability was performed at 72 mg/mL in MOD1 buffer by placing an Eppendorf tube containing 1 mL of product at −80° C. for 10 min, followed by thawing at room temperature. No significant aggregation was observed after 3 cycles of freeze-thaw. Stability studies were performed at 100 mg/mL in MOD1 buffer for 6 weeks at 2-8° C. (FIG.4A) and ambient temperature (22° C.,FIG.4C); in MOD1 buffer at 5 mg/mL for 4 weeks at 40° C. (FIG.4B); in 20 mM buffer (glutamic acid pH 4.0, histidine pH 5.8, tris pH 8.0) at 4 mg/mL for 5 or 11 days at 37° C. (FIG.4D). Testing of the time points was performed by SE-HPLC. No significant increase in HMW was detected in any of the studies. Similarly analysis by CGE did not show any significant differences between the time points. Charge heterogeneity was assayed by iCE (Table 7), which showed an increase in acidic species at 37° C. (particularly at pH 8.0) and 40° C., indicating some degree of deamidation and/or oxidation. However, no major changes were detected to trigger a liquid chromatography (LS)/mass spectrometry (MS) investigation. Other stability series (2-8° C. and ambient temperature) did not show significant changes in % acidic and % basic species by iCE. The stability time points from the 40° C. series were tested in the cell-based assay measuring the neutralization of IFNβ activity (FIGS.5A-D). On day 1, 20,000 HEK293 ISRE-Luc (IFNβ responsive luciferase reporter) cells were plated in 100 μL of DMEM containing 10% fetal bovine serum (FBS) per well in tissue culture treated 96 well plates. Antibody solutions were prepared as 2× stocks starting at a top concentration of 1 μM in DMEM/10% FBS, and then an 11 point, 10-fold dilution series was made with media. A 20× stock of IFNβ (0.625 ng/mL) was prepared in media and added to the antibody titration stocks to a final 2× concentration. The antibody:IFNβ solutions were incubated for 2 hours at 37° C., then 100 μL of the solution was added per well and plates were cultured overnight at 37° C. On day 3, a 150 μg/mL solution of Beetle Luciferine, potassium salt was prepared and 20 μL/well was added and plates were incubated for 15 minutes at 37° C. Luminesence was read on an EnVision multilabel plate reader. No changes in neutralizing activity were detected. CTI-AF1 is compatible with a formulation buffer (20 mM His, 8.5% Sucrose, 0.05 mg/mL EDTA, pH 5.8) and maintains solubility up to 150 mg/mL with acceptable viscosity. TABLE 7Charge heterogeneity in the stability samplesSampleNameplAcidicMainBasicHC_T08.7417.379.53.2HC_1wk4C8.7417.479.73HC_2wk4C8.7517.579.13.3HC_3wk4C8.7417.778.93.4HC_4wk4C8.7518.178.93HC_5wk4C8.7419.177.13.8HC_6wk4C8.7417.879.23HC_1wk25C8.7417.479.33.4HC_2wk25C8.7417.978.93.2HC_3wk_25C8.7318.278.53.4HC_4wk25C8.7319.276.93.9HC_5wk25C8.7319.876.73.5HC_6wk25C8.7220.376.43.440C_1wk8.7123.970.85.240C_2wk8.732.860.86.440C_3wk8.737.456.75.940C_4wk8.742.152.15.7pH4_T08.718.778.52.8pH4_5d8.72274.93.1pH4_11d8.6925.967.46.7PH5_8_T08.7419.377.73pH5_8_5d8.7321.375.63.2pH5_8_11d8.7424.470.84.8pH8_T08.732176.32.7pH8_5d8.7427.570.12.4pH8_11d8.7434.163.62.3 Example 3. Pharmacology Brief Summary CTI-AF1 is a potent and highly selective humanized IgG1 antibody against the soluble cytokine interferon beta (IFNβ). In vitro, CTI-AF1 showed high affinity for human IFNβ (KDof 36.7±12.4 μM). The antibody showed similar EC50binding for human and cynomolgus monkey IFNβ (15.28±2.11 μM and 25.04±5.11 μM, respectively). In human cell-based functional assays, CTI-AF1 showed potent neutralization of IFNβ induced STAT1 phosphorylation (IC507.7±5.0 to 29.8±6.9 μM) and expression of a type I interferon stimulated luciferase reporter in cultured human cells (ISRE assay; IC5028.8±7.6 μM). CTI-AF1 also inhibited the IFNβ-driven expression of MxA (Mx1) in gene expression assays (IC5029.4±23.5 μM) and was able to inhibit IFNβ endogenously expressed by human dermal fibroblasts, a disease relevant cell type, after polyinosinic:polycytidylic acid (poly I:C) stimulation. Primary Pharmacology, In Vitro During the initial hybridoma screening, antibodies were selected based upon their ability to block the binding of IFNβ to IFNAR2, the high affinity component of the type I IFN receptor (FIG.6). In subsequent screenings post humanization and affinity maturation, antibody selection was based upon functional neutralization of IFNβ in cell based assays. SPR was used to determine the KDof CTI-AF1 to human IFNβ; binding experiments were performed using a Biacore T200 optical biosensor equipped with research-grade CM5 sensor chip and human IFNβ (Peprotech). Regeneration of the chip was performed using stripping buffer (3 M MgCl2at pH 3.0 or 10 mM glycine at pH 1.5) followed by a buffer rinse. CTI-AF1 was immobilized on the surface of a CM5 sensor chip at room temperature. The capture level covered a range of 50 to 375 resonance units (RU). The analyte, human IFNβ, was then injected at a flow rate of 30-50 μL per minute for an association time ranging from 65-300 seconds, followed by a dissociation phase of 10 minutes. The kinetic characterization of the interactions was performed using the traditional multi-cycle method, using a series of human IFNβ concentrations from 10 nM down to 0.078125 nM in a series of 2-fold dilutions. Each concentration was evaluated in duplicate. The analyte was removed by regeneration of the array surface between each cycle using 3 M MgCl2at pH 3.0 or 10 mM glycine at pH 1.5, followed by a buffer rinse. This regeneration step removed the bound analyte and returned the response signal to baseline. Data from the reference flow cell (without analyte) were subtracted from the antigen binding responses to remove systematic artifacts. The apparent binding affinity was determined using a simple 1:1 interaction model and the equilibrium constant KDwas determined as the ratio of the kinetic rate constants. The apparent binding affinity of CTI-AF1 for human IFNβ was determined to be 36.7±12.4 μM (FIG.7). Binding of CTI-AF1 to human IFNβ along with cynomolgus monkey, rabbit, rat and mouse orthologs and three of the nearest type I human homologs and IFNγ (type II) were evaluated in plate-based ELISAs. ELISA plates were coated overnight at 4° C. with 5 μg/mL of one of the following cytokines: human IFNβ, cynomolgus monkey IFNβ, rat IFNβ, human IFNα2, IFNγ, human IFNω; mouse IFNβ or human IFNα14(H2) were coated at 1 μg/mL, and rabbit IFNβ was coated at 10 ng/mL. All proteins were diluted in calcium and magnesium-free phosphate buffered saline. Coated plates were washed with phosphate buffered saline containing 0.05% Tween-20 (PBST) and blocked for 1 hour at room temperature with blocking buffer (PBST+0.5% BSA). Plates were washed again with PBST and primary antibodies were added to the plate at 30 nM starting concentration, followed by 1:3 dilutions in blocking buffer. For the anti-rabbit IFNβ, 1:10 dilutions were performed. Plates were incubated for 1 hour at room temperature and then washed with PBST. Binding was detected with species-specific peroxidase-linked secondary antibodies and tetramethylbenzidine (TMB1) substrate. The reaction was stopped with 0.18 M sulfuric acid (H2SO4) and absorbance was read at 450 nm in an EnVision multilabel reader (PerkinElmer). Table 8 shows similar reactivity for human and cynomolgus monkey IFNβ, while reactivity to rabbit IFNβ is 200 times lower. There was no detectable binding to rat or mouse IFNβ, or to the three nearest human homologs or IFNγ (type II). TABLE 8Reactivity of CTI-AFI to IFNβ orthologs and nearest type Ihomologs and IFNγ as measured by ELISATargetHomology (%)CTI-AF1 EC50(pM)Cross-species bindingHuman IFNβ10015.14Cynomolgus monkey IFNβ95.724.67Rabbit IFNβ56.12948Rat IFNβ48.6No bindingMouse IFNβ47.5No bindingCross-reactivityHuman IFNα230.2No bindingHuman IFNα1738.2No bindingHuman IFNω29.0No bindingHuman IFNγ13.2No binding“No binding”: when the absorbance at 450 nm was < 2× the absorbance of the blank control wells. Two in vitro assays were used to demonstrate CTI-AF1 dependent inhibition of IFNβ induced signals. Firstly, HEK293 cells stably transduced with a human ISRE luciferase reporter were used as a measure of IFNβ dependent gene expression; on day 1, 20,000 HEK293 ISRE-Luc (IFNβ responsive luciferase reporter) cells were plated in 100 μL of DMEM containing 10% fetal bovine serum (FBS) per well in tissue culture treated 96 well plates. Antibody solutions were prepared as 2× stocks starting at a top concentration of 1 μM in DMEM/10% FBS. An 11 point, 10-fold dilution series was made with media. A 20× stock of IFNβ (28 nM, final assay concentration was 1.4 nM, the EC50) was prepared in media and added to the antibody titration stocks to a final 2× concentration. The antibody:IFNβ solutions were incubated for 2 hours at 37° C., then 100 μL was added per well and plates were cultured overnight at 37° C. On day 3, a 150 μg/mL solution of Beetle Luciferine, potassium salt was prepared and 20 μL/well was added and plates were incubated for 15 minutes at 37° C. Luminesence was read on an EnVision multilabel plate reader.FIG.8Ashows CTI-AF1 dose-dependent inhibition of IFNβ induced luciferase activity with an IC50of 28.8±7.6 μM. Secondly, CTI-AF1 mediated inhibition of IFNβ induced STAT1 phosphorylation was evaluated by phosflow. U937 cells, a human monocytic cell line, were grown in RPMI 1640 containing 10% FBS and 2 mM Glutamax (cRPMI). Antibody stocks were made at 4×, with a top concentration of 4 μM (final top concentration was 1 μM) and a 12 point, 10-fold dilution series was made in cRPMI; 25 μL was added/well in a u-bottom 96 well tissue culture plate. An equal volume of 4×IFNβ (200 μM, final concentration was 50 μM, EC90) was added to the antibody stocks and incubated for 2 hours at 37° C. Control wells included media alone (no stimulation background pSTAT1 expression) and 50 μM IFNβ only (maximum pSTAT1 signal). U937 cells were harvested, centrifuged for 5 min at 1500 rpm, room temperature and then resuspended at a concentration of 2×106/mL in cRPMI warmed to 37° C.; 50 μL of cell suspension was added per well and plates placed at 37° C. for 15 minutes. Next, 100 μL of pre-warmed cytofix buffer was added and plates were placed back at 37° C. for 15 minutes. Plates were removed and centrifuged as described above. Media was removed from the plates, cells resuspended and washed in 200 μL of PBS and centrifuged again. Media was removed again, then cells were resuspended in 100 μL of permeabilization buffer IV and incubated at room temperature for 15 minutes. At the end of the incubation, cells were centrifuged and washed as described above. After the PBS wash, cells were resuspended in 100 μL of PBS/5% FBS; 5 μL of TruStain FcX/well was added and plates were incubated for 10 min at 4° C. Ten microliters of Alexa Fluor 674 (AF647) conjugated anti-phospho STAT1 antibody was added per well and incubated for 20 min at 4° C. After incubation, 120 μL of FACS buffer was added per well and plates were centrifuged as described above. The wash was repeated with 220 μL of FACS buffer and cells were resuspended in 120 uL of FACS buffer. A Fortessa cytometer was used to acquire the data and analysis was performed using FlowJo software. The geometric mean fluorescence intensity (Geo MFI) in the AF647 channel was calculated and prism software was used to calculate the IC50. CTI-AF1 is a potent neutralizer of human IFNβ with an IC50of 29.8±6.9 μM (FIG.8B). To evaluate the ability of CTI-AF1 to neutralize recombinant IFNβ induced MxA (Mx1) gene expression normal human dermal fibroblasts (HDF) were plated in a T-150 flask in fibroblast culture medium. To set up the assay, cells were dislodged from the flask using trypsin/EDTA solution and plated in a 48 well plate with three wells assigned per experimental condition. On day 3, the cells were stimulated for 5 hours with culture medium spiked with 0.15 μM IFNβ that was pre-incubated for 2 hours with or without dilutions of CTI-AF1 ranging from 10 nM to 0.016 nM. A combination of 0.15 μM IFNβ and 50 nM of isotype control antibody was used as a negative control for the experiment. After 5 hours, cells were harvested, RNA was isolated using RNeasy micro kit and cDNA synthesized using high capacity cDNA reverse transcription kit. Taqman real time PCR analyses were performed in a Vii A7 system (Thermo Fisher) using human gene specific primer probes for Mx1 and 82 M. The relative quantification (fold change) was calculated from the resultant Ctvalues using the ΔΔCt method as follows: for each condition, Ctvalues of the endogenous control gene (82 M) were subtracted from respective Ctvalues for target gene (Mx1). This was followed by normalization against the untreated sample to calculate the ΔΔCt values, which were subsequently used to calculate the fold change (2−ΔΔCt). The isotype negative control antibody had no impact on MxA (Mx1) expression; however, in the presence of CTI-AF1, a dose-dependent inhibition of gene transcription was seen with an IC50of 29.4±23.5 pM (FIG.9). The specificity of CTI-AF1 neutralization was evaluated by using the same pSTAT assay as described earlier forFIG.8B, however, U937 cells were stimulated with either a final concentration of 20 pM IFNβ or 50 pM IFNα. The different concentrations of type I IFNs were selected to provide a similar level of STAT1 phosphorylation as IFNα is a less potent activator of IFNAR signaling. A similar 12 point, 10-fold dilution series was made with sifalimumab (SIF) as a positive control for IFNα neutralization. As can be seen, CTI-AF1 specifically inhibited IFNβ induced STAT1 phosphorylation, but did not inhibit phosphorylation induced by IFNα (FIGS.10A and B, respectively). A single experiment was conducted using either IFNω (at 100 μM) or IFNα14 (at 4 pM) and CTI-AF1 had no effect on IFNω or IFNα14 induced STAT1 phosphorylation. To ensure that CTI-AF1 neutralized endogenously expressed IFNβ, normal human dermal fibroblasts were seeded in a 48 well plate with three wells assigned per experimental condition. On day 3, cells were stimulated with or without a combination of 1 μg/mL poly I:C and dilutions of CTI-AF1 (dose range: 50 pM-100 nM) or 100 nM sifalumumab. After 2.5 and 24 hours, cells were harvested, RNA isolated using RNeasy micro kit and cDNA synthesized using high capacity cDNA reverse transcription kit. Taqman real time PCR and fold change calculations were performed as described above (FIG.9). While the amount of IFNβ induced by poly I:C stimulation was unknown, a dose-dependent inhibition of MxA (Mx1) expression was seen in the presence of CTI-AF1 (FIG.11). Example 4. Translational Pharmacology The PK/PD relationship for IFNβ in dermatomyositis (DM) has not been defined. There are no relevant translatable preclinical models available for DM and the preclinical efficacious concentration (Ceff) is not understood. A type 1 Interferon gene signature will be used clinically as a mechanistic biomarker of pharmacology modulation. Type 1 Interferon genes are typically elevated in DM and SLE patients and the mean fold-change of the type 1 Interferon gene signature has been used previously in clinical studies for anti-IFNα (sifalimumab and rontalizumab) and anti-IFNAR (anifrolumab) mAbs. However, a quantitative understanding of the gene signature modulation has not been established and the relationship between in vivo exposure, target engagement, downstream pharmacology and efficacy over time is not understood. Human efficacious dose feasibility projections are based on the ability of CTI-AF1 to neutralize >95% of IFNβ in skin. An LC\MS\MS assay is used to measure total IFNβ in clinical serum and tissue biopsies, and in combination with CTI-AF1 clinical PK and KD, is used to assess and confirm target engagement. Type 1 IFN gene signature in blood and skin, as well as IP-10 (CXCL10), are assessed as mechanistic biomarkers. In a subsequent Proof of Mechanism (PoM)/Early Signal of Efficacy (ESoE) study in DM patients, cutaneous dermatomyositis disease area and severity index (CDASI) is used as the primary endpoint (outcome biomarker) in addition to any relevant mechanistic biomarkers. Pharmacokinetics-Pharmacodynamics Relationship and Human Dose The pharmacokinetic and pharmacodynamic (PK/PD) relationships between antibody drug exposure and IFNβ for CTI-AF1 have been simulated using reported PK parameters for typical IgG1therapeutics, IFNβ-CTI-AF1 equilibrium binding constant, IFNβ concentrations in skin and serum, and IFNβ turnover half-life. A “Site-of-Action” PK/PD model was used to predict the coverage of IFNβ in DM patients. An IFNβ coverage of >95% at trough was considered necessary to achieve efficacy. Skin interstitial concentrations of CTI-AF1 were assumed to be 30% of serum concentrations. The binding affinity of CTI-AF1 to IFNβ determined by SPR (Biacore KD=36.7 μM) was used for PK/PD modeling. Consistent with this, in cell-based functional assays, CTI-AF1 showed potent neutralization of IFNβ-induced STAT1 phosphorylation (IC5029.8 μM). The median IFNβ concentration in DM patient serum was 3 pg/mL (N=26); however, the IFNβ concentration in DM patient skin is not known. Therefore, in the model, the impact of IFNβ skin:plasma ratio was investigated at ratios of 10 and 100. Since this is a sensitive parameter for the model, these ratios were used as proposed boundary conditions to demonstrate the impact of the skin:plasma ratio on target coverage. The in vivo half-life of IFNβ turnover was estimated by fitting a 3-compartmental model to the human PK data for IFNβ1α, which included 3 IV doses. This fitting resulted in two different half-lives for IFNβ turnover which are considered most relevant, depending on the phase and compartments considered ranging from 3 minutes (based on the initial phase) to 126 minutes (based on the effective half-life). To increase confidence in this model parameter, an IFNβ assay for cynomolgus monkey serum was developed for use in cynomolgus monkey. The IFNβ skin:plasma ratio and the IFNβ turnover rate are sensitive parameters for the PK/PD model. Thus, the human efficacious dose feasibility assessment was performed using the ranges described above for both IFNβ skin:plasma ratio and IFNβ turnover rate. Example assessments for two likely clinical ESoE dose regimens are shown inFIGS.12A-D (IV Q4W) andFIGS.13A-D(SC Q1W). CTI-AF1 solubility of 150 mg/mL would enable a clinical dose of 2 mg/kg, as it can be delivered via a 1 mL injection pen. Hence a dose of 2 mg/kg was used for the dose feasibility assessments below. FIGS.12A-Dshow that at a dose of 2 mg/kg IV Q4W, irrespective of IFNβ skin:plasma ratio, only the 126 min half-life for IFNβ predicts >95% IFNβ coverage in skin. If the half-life of IFNβ was 3 min, >95% IFNβ coverage in skin is predicted to require doses higher than 2 mg/kg.FIGS.13A-Dshow that at a dose of 2 mg/kg SC Q1W, irrespective of IFNβ skin:plasma ratio, the 126 min half-life for IFNβ predicts >95% IFNβ coverage in skin. If the half-life of IFNβ was 3 min, then only the IFNβ skin:plasma ratio of 100 will result in >95% IFNβ coverage in skin at 2 mg/kg. By contrast, if IFNβ skin:plasma ratio is 10, achieving >95% coverage will require doses higher than 2 mg/kg. Human PK/Exposure Based on the pharmacokinetic profiles of CTI-AF1 in cynomolgus monkey, the pharmacokinetics of CTI-AF1 in human are expected to be similar to the reported values for a typical IgG1therapeutic. The 2-compartment pharmacokinetic parameter values are summarized in Table 9. Simulated concentration-time profiles of CTI-AF1 at projected efficacious dose levels are depicted in the top panels ofFIGS.12A-Dand13A-D. TABLE 9Projected Pharmacokinetic Parameters of CTI-AF1 in HumanParameterDefinitionProjectionCLcentral clearance0.00258mL/min/kgV1central volume43.7mL/kgCLDdistribution clearance0.00565mL/min/kgV2peripheral volume44.3mL/kgKaabsorption rate constant for SC0.000181/mindosingF_scSC bioavailability60%Vdsssteady-state volume of distribution88mL/kgT1/2terminal half life19days Nonclinical Pharmacokinetics IV and SC pharmacokinetics of CTI-AF1 have been assessed in cynomolgus monkeys using data from a single-dose exploratory toxicity study. Mean serum pharmacokinetic parameter values for cynomolgus monkeys are summarized in Table 10 and mean serum concentrations of CTI-AF1 are shown inFIG.14. TABLE 10Summary Table of CTI-AF1 Pharmacokinetics in Cynomolgus MonkeysDoseCmaxAUCinfCLVssT1/2F(mg/kg)Route(μg/mL)(μg*hr/mL)(mL/h/kg)(L/kg)(h)(%)10SC97.750000n/an/a37987.310IV248549000.1830.0823337n/a200IV498010000000.2090.0747273n/aMean N = 2 monkeys/group, 1 male and 1 female Example 5. IFNβ as a Target for SLE and DM There is increasing evidence that IFN production is linked to SLE and other rheumatic diseases, such as DM. Moreover, the perpetuation of the SLE disease process likely involves further production of type I IFNs and a vicious pathogenic cycle. DM is a rare autoimmune disease (about 20,000 patients in the U. S.) characterized by inflammation of skeletal muscle and skin, and, concomitantly, skeletal muscle weakness and skin rash. DM is typically associated with autoantibodies, and the pathogenesis of the disease may involve sequential binding of these autoantibodies to an endothelial autoantigen, triggering complement activation and vascular inflammation, ultimately leading to perifascicular atrophy. As shown inFIGS.16A-B, data indicated an association of type I interferon-regulated gene (IRG) transcript “signature” in DM blood with skin rash activity, as measured by the cutaneous dermatomyositis disease area and severity index (CDASI). The highly IFNβ-inducible gene MxA (Mx1) is expressed in DM perifascicular myofibers and capillaries, and blood serum IFNβ—but not IFNα or IFNω—is associated with DM, but not with other inflammatory myopathies or normal sera. These data support the notion that injury to capillaries, myofibers and skin in DM results from a pathogenic overproduction of IFNβ message and protein. Data have also demonstrated an association between CDASI scores and serum levels of IFNβ protein (FIG.17). Analyses of paired skin biopsies indicate the presence of both IFNβ mRNA and upregulation of an IRG signature only in affected tissue (FIGS.18A-B). Taken together, these data strongly suggest that DM is an IFNβ-driven disease. Given that in many tissue contexts IFNβ production may precede IFNα production and initiate a pathogenic elevation of IRG signature expression, together with the notion that DM may be a largely IFNβ-driven disease, it is believed that DM and SLE share many pathogenic features and attributes. Indeed, skin lesions of DM are difficult if not impossible to distinguish histologically from those of SLE, and a diagnosis of DM skin lesions typically requires clinical determination of increased CD4+ and CXCR3+ cell types and endothelial expression of Mx1. Moreover, both DM and SLE are characterized by B cell activation and autoantibody mediated inflammation and tissue destruction. TABLE 11Sequences of anti-IFN antibodiesSequencesSeqAb(CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3IDNameunderlined when applicable))1CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIGNYLNWYQQKPGKAFKLLIYSTSRLHSGAF1VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK(CDR-L1, CDR-L2, CDR-L3: SEQ ID NOs 34, 35, and 36,respectively)2CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIGNYLNWYQQKPGKAFKLLIYSTSRLHSGAF2VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK3CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIFSTSRLHSGAF3VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK4CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISSYLNWYQQKPGKAFKLLIYSTSRLHSGAF4VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK5CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIYTTSRLRSGAF5VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK6CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIDNFLQWYQQKPGKAFKLLIYSTSRLHSGAF6VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK7CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIYSTSKLHSGAF7VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK8CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIGNYLNWYQQKPGKAFKLLIYSTSRLHSGAF8VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK9CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIFSTSRLHSGAF9VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK10CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISSYLNWYQQKPGKAFKLLIYSTSRLHSGAF10VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK11CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIYTTSRLRSGAF11VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK12CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIDNFLQWYQQKPGKAFKLLIYSTSRLHSGAF12VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK13CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIYSTSKLHSGAF13VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK14CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIFSTSRLHSGAF14VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK15CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISSYLNWYQQKPGKAFKLLIYSTSRLHSGAF15VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK16CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIYTTSRLRSGAF16VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK17CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIDNFLQWYQQKPGKAFKLLIYSTSRLHSGAF17VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK18CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISNYLNWYQQKPGKAFKLLIYSTSKLHSGAF18VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK19CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIGNYLNWYQQKPGKAFKLLIFSTSRLHSGAF19VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK20CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISSYLNWYQQKPGKAFKLLIYTTSRLRSGAF20VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK21CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIDNFLQWYQQKPGKAFKLLIFSTSKLHSGAF21VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIVLPITFGGGTKVEIK22CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIGNYLNWYQQKPGKAFKLLIFSTSRLHSGAF22VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK23CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISSYLNWYQQKPGKAFKLLIYTTSRLRSGAF23VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK24CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIDNFLQWYQQKPGKAFKLLIFSTSKLHSGAF24VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSTILPLTFGGGTKVEIK25CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIGNYLNWYQQKPGKAFKLLIFSTSRLHSGAF25VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK26CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDISSYLNWYQQKPGKAFKLLIYTTSRLRSGAF26VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK27CTI-VLDIQMTQSPSSLSASVGDRVTITCRTSQDIDNFLQWYQQKPGKAFKLLIFSTSKLHSGAF27VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIK28CTI-VHQVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWMHWVRQAPGQGLEWMGHIDPSDSYAF1TYYNQKFKGRVTITADESTSTAYMELSSLRSEDTAVYYCARWDYGNLLFEYWGQGTLtoVTVSSACT-(CDR-H1, CDR-H2, CDR-H3: SEQ ID NOs 37, 38, and 39,AF27respectively)29AllCHASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLCTI-QSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAAFsPEAAGAPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG(K)30AllCLRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTCTI-EQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECAFs32CTI-LightDIQMTQSPSSLSASVGDRVTITCRTSQDIGNYLNWYQQKPGKAFKLLIYSTSRLHSGVAF1chainPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQGIILPITFGGGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC33CTI-HeavyQVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWMHWVRQAPGQGLEWMGHIDPSDSYAF1chainTYYNQKFKGRVTITADESTSTAYMELSSLRSEDTAVYYCARWDYGNLLFEYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGAPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG(K)34CTI-CDR-L1RTSQDIGNYLNAF135CTI-CDR-L2STSRLHSAF136CTI-CDR-L3QQGIILPITAF137CTI-CDR-H1GYTFSRYWMHAF138CTI-CDR-H2HIDPSDSYTYYNQKFKGAF139CTI-CDR-H3WDYGNLLFEYAF1166CTI-VHCAGGTGCAGCTGGTGCAGAGCGGCGCCGAGGTGAAGAAGCCCGGCAGCAGCGTGAAGAF1nucleicGTGAGCTGCAAGGCCAGCGGCTACACCTTCAGCCGGTACTGGATGCACTGGGTGCGGacidCAGGCCCCCGGCCAGGGCCTGGAGTGGATGGGCCACATCGACCCCAGCGACAGCTACACCTACTACAACCAGAAGTTCAAGGGCCGGGTGACCATCACCGCCGACGAGAGCACCAGCACCGCCTACATGGAGCTGAGCAGCCTGCGGAGCGAGGACACCGCCGTGTACTACTGCGCCCGGTGGGACTACGGCAACCTGCTGTTCGAGTACTGGGGCCAGGGCACCCTGGTGACCGTCTCGAGC167CTI-VLGACATCCAGATGACCCAGAGCCCCAGCAGCCTGAGCGCCAGCGTGGGCGACCGGGTGAF1nucleicACCATCACCTGCCGGACCAGCCAGGACATCGGCAACTACCTGAACTGGTACCAGCAGacidAAGCCCGGCAAGGCCTTCAAGCTGCTGATCTACAGCACCAGCCGGCTGCACAGCGGCGTGCCCAGCCGGTTCAGCGGCAGCGGCAGCGGCACCGACTTCACCCTGACCATCAGCAGCCTGCAGCCCGAGGACTTCGCCACCTACTACTGCCAGCAGGGGATTATTTTGCCCATTACCTTCGGCGGCGGCACCAAGGTGGAGATCAAG Example 6. Epitope Mapping To elucidate the epitope recognized by CTI-AF1, hybrid IFNβ proteins were made where selected portions of IFNβ sequences were replaced with IFNα sequence. CTI-AF1 specifically neutralizes IFNβ but not IFNα, therefore the inability of CTI-AF1 to neutralize a given hybrid protein would indicate loss of the epitope. Hybrid proteins were produced, purified and ability to induce STAT1 phosphorylation was confirmed (Table 12). TABLE 12sequences of hybrid IFN proteinsSeqHybridSequencesIDIFN name(mutated residues underlined)41HumanMSYNLLGFLQ RSSNFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQLQQFIFNβQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAN VYHQINHLKTVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKAKEYSH CAWTIVRVEILRNFYFINRL TGYLRN158CID1276MSYNLLGFLQ RSSNRRCLML LAQLNGRLEY CLKDRMNFDI PEEIKQLQQFQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAN VYHQINHLKTVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKAKEYSH CAWTIVRVEILRNFYFINRL TGYLRN159CID1277MSYNLLGFLQ RSSNFQCQKL LWQLNGRLEYCLKDRHDFGI PQEIKQLQQFQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAN VYHQINHLKTVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKAKEYSH CAWTIVRVEILRNFYFINRL TGYLRN160CID1280MSYNLLGFLQ RSSNFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQLQQFQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVDKLLTN VYHQINHLKTVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKAKEYSH CAWTIVRVEILRNFYFINRL TGYLRN161CID1281MSYNLLGFLQ RSSNFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQLQQFQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAEVYQQINDLEAVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKAKEYSH CAWTIVRVEILRNFYFINRL TGYLRN162CID1283MSYNLLGFLQ RSSNFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQLQQFQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAN VYHQINHLKTVLEEKLEKED FTRGKLMSIL HLRKYYGRIL HYLKAKEYSH CAWTIVRVEILRNFYFINRL TGYLRN163CID1285MSYNLLGFLQ RSSNFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQLQQFQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAN VYHQINHLKTVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKEKKYSH CAWTIVRVEILRNFYFINRL TGYLRN164CID1286MSYNLLGFLQ RSSNFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQLQQFQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAN VYHQINHLKTVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKAKEYSPCAWTIVRVEILRNFYFINRL TGYLRN165CID1287MSYNLLGFLQ RSSNFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQLQQFQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAN VYHQINHLKTVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKAKEYSH CAWTIVRAEILRNFSLITRL TGYLRN All purified hybrid proteins were able to induce STAT1 phosphorylation, however, there were differences in the biological activity. Each hybrid protein was used at the EC80concentration in the following phospho-STAT1 assay (pSTAT1). U937 cells, a human monocytic cell line, were grown in RPMI 1640 containing 10% FBS (cRPMI). Antibody stocks were made at 4×, with a top concentration of 4-400 μM (final top concentration was 1-100 μM) and a 11 point, 10-fold dilution series was made in cRPMI; 25 μl was added/well in a u-bottom 96 well tissue culture plate. An equal volume of 4× hybrid or control IFNβ was added at the appropriate EC80concentration to the antibody stocks and incubated for 2 hours at 37° C. Control wells included media alone (no stimulation background pSTAT1 expression) or no addition of antibody (maximum pSTAT1 signal). U937 cells were harvested, centrifuged for 5 min at 1500 rpm at room temperature and then resuspended at a concentration of 2×106/ml in cRPMI warmed to 37° C.; 50 μl of cell suspension was added per well and plates placed at 37° C. for 15 minutes. Next 100 μl of pre-warmed cytofix buffer (BD Biosciences, catalog #554655) was added and plates were placed back at 37° C. for 15 minutes. Plates were removed and centrifuged as described above. Media was removed from the plates, cells resuspended and washed in 200 μl of PBS and centrifuged again. Media was removed again, cells were resuspended in 100 μl of permeabilization buffer IV (BD Biosciences) and incubated at room temperature for 15 minutes. At the end of the incubation, cells were centrifuged and washed as described above. After the PBS wash, cells were resuspended in 100 μl of PBS/5% FBS (FACS buffer); 5 μl of TruStain FcX/well (BioLegend) was added and plates were incubated for 10 min at 4° C. Ten microliters of Alexa Fluor 674 (AF647) conjugated anti-phospho STAT1 Ab (BD Biosciences) was added per well and incubated 20 min at 4° C. After incubation, 120 μl of FACS buffer was added per well and plates were centrifuged as described above. The wash was repeated with 220 μl of FACS buffer and cells were resuspended in 120 ul of FACS buffer; a Fortessa cytometer (BD Biosciences) was used to acquire the data and analysis was performed using FlowJo software (TreeStar). The geometric mean fluorescence intensity (Geo MFI) in the AF647 channel was calculated and prism software was used to calculate the IC50. Data was normalized as the ratio of antibody concentration/IFN concentration and the percentage of the maximum signal was determined after subtracting the background. U937 cells were stimulated with IFNα/IFNβ hybrid proteins for 15 minutes in the presence of CTI-AF1 after which the presence of phosphorylated STAT1 was assessed by intracellular flow cytometry. CTI-AF1 did not inhibit CID1280-dependent STAT1 phosphorylation and the potency for CID1281-induced STAT1 phosphorylation neutralization was greatly reduced. CTI-AF1 neutralized STAT1 phosphorylation of all other hybrid IFN proteins with equal potency relative to human IFNβ. SeeFIG.19and Table 13. These data combined indicate that the epitope residues recognized by CTI-AF1 are contained within the constructs CID1280 and CID1281, in which the IFNα sequence substitutions span amino acids 85-89 and 90-100, respectively (see Table 12). TABLE 13IC50and fold change of CTI-AF1 mediated neutralizationof type I IFN-induced STAT1 phosphorylationIFN proteinIC50(nM)Fold difference from IFNβHuman IFNβ0.3CID12760.20.7CID12770.30.9CID128047.7161.8CID12813281.011137.1CID12830.41.2CID12850.41.4CID12860.41.4CID12870.31.0 Example 7. Crystal Structure of Anti-IFNβ Antibodies The co-crystals of the complex between Cynomolgus monkey IFNβ and CTI-AF1 Fab were grown using the following solution as a precipitant: 19% PEG 3350, 250 mM sodium Citrate, 100 mM Bis-Tris propane pH 8.5. The crystals belong to space group P21 (unit cell parameters a=49.58 Å; b=91.76 Å; c=162.52 Å; b=94.86 deg) and contain two copies of complex per crystal asymmetric unit. The structure has been determined at 3.2 Å resolution using Molecular Replacement method and the refinement was performed using autoBUSTER. CTI-AF1 Fab binds to IFNβ on the side formed by two α-helices, A and C, which define the binding epitope of CTI-AF1 (Table 13) TABLE 13Epitope analysiscyno-IFNβhuman IFNβStructureAminoPrimarySecondaryOptionalAminoPrimarySecondaryOptionalelementsAcidsepitopeepitopeepitopeStructuralAcidsepitopeepitopeepitopeHelix ALeu 5Leu 5Helix ALeu 5Leu 5Leu 6Leu 6Leu 6Leu 6Phe 8Phe 8Phe 8Phe 8Leu 9Leu 9Leu 9Leu 9Ser 12Ser 12Ser 12Ser 12Ser 13Ser 13Ser 13Ser 13Phe 15Phe 15Phe 15Phe 15Gln 16Gln 16Gln 16Gln 16Helix CThr 82Thr 82Helix CThr 82Thr 82Asn 86Asn 86Asn 86Asn 86Ala 89Ala 89Ala 89Ala 89Asn 90Asn 90Asn 90Asn 90Tyr 92Tyr 92Tyr 92Tyr 92His 93His 93His 93His 93Asp 96Asp 96Asn 96Asn 96His 97His 97His 97His 97Thr 100Thr 100Thr 100Thr 100Helix BTyr 67Tyr 67Helix BPhe 67 is not part of theepitope on human IFNβ All amino acids that are within 3.8 Å from of CTI-AF1 were selected as “potential” epitope residues. “Primary” epitope residues are characterized as highly buried residues at the of CTI-AF1-IFNβ interface and zero-to-low sequence tolerance to any other amino acid substitutions at this position. “Secondary” epitope residues are characterized as residues with medium buried surface area at the interface and medium sequence tolerance to amino acid substitutions at these positions. “Optional” epitope residues are characterized as residues with low buried surface area at the interface and high sequence tolerance to amino acid substitutions at these positions. The binding paratope is made up by five CDR-variable regions: CDR-H1, -H2, -H3 and CDR-L1, -L3 (Table 14). The total surface area buried under the binding interface is 1,920 Å2. Analysis of CTI-AF1-IFNβ binding mode reveals that the neutralizing effect of CTI-AF1 is achieved through direct blockage on the IFNAR1 binding site. TABLE 14Paratope analysisAminoPrimarySecondaryCDRsAcids*ParatopeparatopeCDR-H1Trp 33HTrp 33HCDR-H2Asp 54HAsp 54HTyr 56HTyr 56HTyr 58HTyr 58HGln 61HGln 61HCDR-H3Tyr 97HTyr 97HGly 98HGly 98HLeu 100HLeu 100HCDR-L1Gln 27LGln 27LAsp 28LAsp 28LIle 29LIle 29LGly 30LGly 30LTyr 32LTyr 32LCDR-L3Ile 92LIle 92LIle 93LIle 93LLeu 94LLeu 94L All amino acids that are within 3.8 Å from IFNβ were selected as “potential” binding paratope. “Primary” paratope residues are characterized as highly buried residues at the CTI-AF1-IFNβ interface and low sequence tolerance to any other amino acid substitutions at this position. “Secondary” paratope residues are characterized as residues with lower buried surface area at the interface and higher sequence tolerance to amino acid substitutions at these positions. Table 15 summarizes the epitope-paratope interaction pairs. Table 16 summarizes epitope and paratope analysis based on BSA. TABLE 15Epitope-paratope interaction pairsHuman IFNβepitope residueCTI-AF1 paratope residue(s)Type of interaction5 Leu32L TyrH-bond6 Leu32L TyrH-bond8 Phe28L Asp, 29L Ile, 30L Gly, 32L Tyrvan der Waals9 Leu32L Tyr, 92L Ilevan der Waals12 Ser28L AspH-bond92L Ilevan der Waals13 Ser92L Ilevan der Waals15 Phe27L Glnvan der Waals16 Gln27L GlnH-bond28L Asp, 93L Ilevan der Waals82 Thr61H Glnvan der Waals86 Asn58H Tyr, 94L Leuvan der Waals89 Ala58H Tyr, 94L Leuvan der Waals90 Asn93L Ile,van der Waals94L LeuH-bond92 Tyr33H Trp, 56H Tyrvan der Waals93 His97H Tyr,H-bond100H Leu,van der Waals92L IleH-bond96 Asp97H Tyr,van der Waals33H TrpH-bond97 His97H Tyr, 98H Gly, 100H Leuvan der Waals100 Thr97H TyrH-bond TABLE 16Epitope and paratope analysis based on BSAPotential IFNβepitope residuesBSA (Å2)5 Leu89.86 Leu3.58 Phe72.49 Leu51.812 Ser30.113 Ser18.916 Gln77.482 Thr40.286 Asn51.889 Ala52.090 Asn53.192 Tyr75.793 His119.4Potential paratopeAminoBSAresiduesAcids*(Å2)CDR-H1Trp 33H34.5CDR-H2Asp 54H18.7Tyr 56H67.6Tyr 58H69.9Gln 61H52.1CDR-H3Tyr 97H101.7Gly 98H31.7Leu 100H31.3CDR-L1Gln 27L54.4Asp 28L39.1Ile 29L7.8Gly 30L16.8Tyr 32L91.9CDR-L3Ile 92L80.3Ile 93L55.2Leu 94L79.7 Example 8. Type I Interferon Expression Profiles In this example, we studied type I IFN expression profiles of 4 disease relevant cell lines in response to toll-like receptor ligand stimulation. Four types of cells were used: PBMCs, a dermal fibroblast cell line, a muscle cell line and a kidney cell line, which were stimulated with a TLR3, TLR4, TLR7/8 and TLR9 agonist in the presence and absence of anti-IFNβ antibody. Gene expression levels of Type I IFN and Mx1 in different primary human cell types was measured using quantitative-PCR. Primary cells were cultured in the relevant media as follows: normal human dermal fibroblasts in FGM-2 bulletkit media, normal human mesangial in MsGM bulletkit media, and primary human skeletal muscle derived cells in Myotonic growth medium. Peripheral blood mononuclear cells (PBMC) were isolated by centrifugation over Ficoll-Paque Plus. Mononuclear cells were cultured in RPM11640 supplemented with 10% FBS and penicillin-streptomycin. To measure the type I IFN gene expression, cells were seeded then stimulated with the relevant TLR ligand for 1, 2.5, 5, 8 and 24 hours. After culture, cells were harvested, RNA was isolated and cDNA was synthesized. Expression of the following genes was assessed by Taqman PCR: IFNβ, Mx1, IFNα1, IFNα2, IFNα4, IFNα5, IFNα6, IFNα7, IFNα8, IFNα14, IFNα16, IFNα17, and B2m. Taqman real time PCR and fold change calculations were performed as described above (FIG.9). Table 17A shows that IFNβ is the predominant Type I IFN produced by various tissue resident primary human cell types upon Toll like receptor (TLR) ligand stimulation. Dermal fibroblasts, skeletal muscle cells, glomerular mesangial cells and PBMCs from normal human donors were stimulated with poly I:C (TLR3 ligand), LPS (TLR4 ligand), R848 (TLR7/8 ligand) and ODN2216 (TLR9 ligand) in a time and dose-dependent manner. Relative expression levels of IFNβ, Mx1, IFNα (1, 2, 4, 5, 6, 7, 8, 14, 16, and 17) were measured via quantitative-PCR using B2 M as the control. Relative expression of each gene is indicated as strong (+), weak (+/−) or no expression (−). CTI-AF1 was shown to be a potent neutralizer of endogenously produced IFNβ from primary human cells stimulated with TLR ligands (poly I:C, LPS, R848 or ODN2216). Cells were stimulated with the various TLR ligands in the absence or presence of titrated amounts of CTI-AF1. Expression of Mx1 was measured 24 hours post stimulation, with the exception of PBMCs stimulated with LPS, which was measured at 6 hours. RNA isolation, cDNA synthesis and quantitative PCR were performed as described above (FIG.9). While the amount of IFNβ induced by any cell type upon TLR stimulation was unknown, a dose-dependent inhibition of Mx1 expression was seen in the presence of CTI-AF1. Table 17B shows that CTI-AF1 is a potent inhibitor of endogenous IFNβ secreted by primary human cells after poly I:C and LPS stimulation. Cells were stimulated with the indicated TLR ligand and quantitative-PCR was performed to determine the level of Mx1 expression using B2 M as the control. Dose-dependent inhibition of Mx1 gene expression by CTI-AF1 is indicated by “+” while the absence of CTI-AF1 dependent Mx1 expression inhibition is indicated by “−”. Conditions where Type I IFN expression was insufficient to drive any meaningful increase in Mx1 expression that could potentially be neutralized by CTI-AF1 is indicated as NA. TABLE 17AGeneDermal FibroblastsSkeletal Muscle Cellstran-In vitro stimulationscriptPolyI:CLPSR848ODN2216PolyI:CLPSR848ODN2216IFNβ++−−++−−Mx1++−−++−−IFNα1−−+/−+/−−+/−−−IFNα2−−−−−−−−IFNα4−−−−−−−−IFNα5+/−−−−−−−−IFNα6−−−−−−−−IFNα7−−−−−−−−IFNα8−−−−−−−−IFNα14−−−−−−−−IFNα16−−−−−−−−IFNα17−−−−−−−−GeneGlomerular Mesangial CellsPBMCstran-In vitro stimulationscriptPolyI:CLPSR848ODN2216PolyI:CLPSR848ODN2216IFNβ++−−++++Mx1++−−++++IFNα1−+/−+/−−+/−+/−++IFNα2−−−−+/−−++IFNα4−−−−+/−−++IFNα5−−−−−−++IFNα6−−−−+/−−++IFNα7−−−−+/−−++IFNα8−−−−+/−+/−++IFNα14−−−−+/−+/−++IFNα16−−−−+/−−++IFNα17−−−−−−+++ = strong expression;+/− = relatively weak expression;− = not detected TABLE178GeneDermal FibroblastsSkeletal Muscle CellsGlomerular Mesangial CellsPBMCstran-In vitro stimulationscriptPolyI:CLPSR848ODN2216PolyI:CLPSR848ODN2216PolyI:CLPSR848ODN2216PolyI:CLPSR848ODN2216Mx1++NANA++NANA++NANA++−−+ = dose-dependent Inhibition of Mx1 gene expression by CTI-AF1;− = no dose-dependent Inhibition of Mx1 gene expression;NA = not applicable, Insufficient type I IFN expression to drive Mx1 expression TABLE 18Sequences of interferon 13 proteinsSEQIDNameSequence40Human IFNβMTNKCLLQIA LLLCFSTTAL SMSYNLLGFL QRSSNFQCQK LLWQLNGRLEprecursorYCLKDRMNFD IPEEIKQLQQ FQKEDAALTI YEMLQNIFAI FRQDSSSTGWNETIVENLLA NVYHQINHLK TVLEEKLEKE DFTRGKLMSS LHLKRYYGRILHYLKAKEYS HCAWTIVRVE ILRNFYFINR LTGYLRN41Mature humanMSYNLLGFLQ RSSNFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQLQQFIFNβQKEDAALTIY EMLQNIFAIF RQDSSSTGWN ETIVENLLAN VYHQINHLKTVLEEKLEKED FTRGKLMSSL HLKRYYGRIL HYLKAKEYSH CAWTIVRVEILRNFYFINRL TGYLRN42Mature mouseINYKQLQLQE RTNIRKCQEL LEQLNGKINL TYRADFKIPM EMTEKMQKSYIFNβTAFAIQEMLQ NVFLVFRNNF SSTGWNETIV VRLLDELHQQ TVFLKTVLEEKQEERLTWEM SSTALHLKSY YWRVQRYLKL MKYNSYAWMV VRAEIFRNFLIIRRLTRNFQ N43Mature ratIDYKQLQFRQ STSIRTCQKL LRQLNGRLNL SYRTDFKIPM EVMHPSQMEKIFNβSYTAFAIQVM LQNVFLVFRS NFSSTGWNET IVESLLDELH QQTELLEIILKEKQEERLTW VTSTTTLGLK SYYWRVQRYL KDKKYNSYAW MVVRAEVFRNFSIILRLNRN FQN44MatureMSYNLLGFLQ RSSSFQCQKL LWQLNGRLEY CLKDRMNFDI PEEIKQPQQFCynomolgusQKEDAALTIY EMLQNIYAIF RQDLSSTGWN ETIVENLLAN VYHQIDHLKTmonkey IFNβILEEKLEKED FTRGKFVSSL HLKRYYGRIL HYLKAKEYSH CAWTIVRVEILRNFFFINKL TGYLRN45Mature rabbitMSYNSLQIQL WHGSLTCAKL LLQLNGTTED CLNERINFKV PKEIKEPQQLIFNβQKEDTTLVIF EMLNNIFDIF RKNFSSTGWN ETLVENLLGE THLQIHHLKSKINKKVTLES IRMNLRLKSY YWRIMDYLET KQYSNCAWKI VQLEIFRNFSFIIMLIDYL The various features and embodiments of the present invention, referred to in individual sections above apply, as appropriate, to other sections, mutatis mutandis. Consequently features specified in one section may be combined with features specified in other sections, as appropriate. All references cited herein, including patents, patent applications, papers, text books, and cited sequence Accession numbers, and the references cited therein are hereby incorporated by reference in their entirety. In the event that one or more of the incorporated literature and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls.
160,124
11858987
MODE FOR CARRYING OUT THE INVENTION The present invention relates to cell surface molecules that are specifically expressed on cancer stem cells, and pharmaceutical compositions (anti-cancer agents, etc.) and reagents for detecting cancer stem cells, which use antibodies against the cell surface molecules. Herein, “cancer” refers to the physiological condition in mammals, which is typically characterized by unregulated cell growth, or such a physiological condition. Herein, cancer types are not particularly limited, and include those listed below. Carcinomas (epithelial cancers) include pancreatic cancer, prostatic cancer, breast cancer, skin cancer, cancers of the digestive tract, lung cancer, hepatocellular carcinoma, cervical cancer, uterine cancer, ovary cancer, fallopian tube cancer, vaginal cancer, liver cancer, bile duct cancer, bladder cancer, ureter cancer, thyroid cancer, adrenal cancer, kidney cancer, and cancers of other glandular tissues. Sarcomas (non-epithelial tumors) include liposarcoma, leiomyosarcoma, rhabdomyosarcoma, synovial sarcoma, angiosarcoma, fibrosarcoma, malignant peripheral nerve sheath tumor, gastrointestinal stromal tumor, desmoid tumor, Ewing's sarcoma, osteosarcoma, chondrosarcoma, leukemia, lymphoma, myeloma, tumors of other parenchymal organs, for example, melanoma and brain tumor (Kumar V, Abbas A K, Fausio N. Robbins and Cotran Pathologic Basis of Disease. 7th Ed. Unit I: General Pathology, 7: Neoplasia, Biology of tumor growth: Benign and malignant neoplasms. 269-342, 2005). Herein, “tumor” refers to arbitrary benign (non-cancerous) and malignant (cancerous) tissue masses, including pre-cancerous lesions, which result from overgrowth or overexpansion of cells. Herein, cancer stem cell (CSC) refers to cells having the abilities described in (i) and/or (ii) below. (i) The ability to self-renew. The self-renewal ability refers to the ability of either or both of the divided daughter cells to produce cells which maintain the same capacity and the degree of differentiation as the parental cell in terms of cell lineage. (ii) The ability to differentiate into various types of cancer cells that constitute a cancer cell mass. Like normal stem cells, various types of cancer cells differentiated from cancer stem cells generate a hierarchical organization with cancer stem cells at the top in terms of cell lineage. Various types of cancer cells are generated in a sequential manner from cancer stem cells. This results in the formation of a cancer cell mass that exhibits a variety of features. Cancer stem cell refers to a cancer cell that has the ability to form cancers as well as, like normal stem cell, pluripotency and self-renewal ability. Cancer stem cells generate a hierarchical organization with cancer stem cells at the top. Various types of cancer cells are generated in a sequential manner from cancer stem cells. This results in the formation of a cancer cell mass that exhibits a variety of features. Cancer cell mass refers to, not a group of individual cells, but a mass formed by the adhesion of cells etc. as in human tumor tissue, which is built with cancer cells, and other cells such as stromal cells and blood cells, extracellular matrix such as collagen and laminin, and so on. The origin of cancer stem cells, which are the target in therapy using pharmaceutical compositions of the present invention, is not particularly limited; it is possible to use cancer stem cells derived from mammals such as humans, monkeys, chimpanzees, dogs, bovines, pigs, rabbits, rats, and mice. However, cancer stem cells derived from humans are preferred, and those derived from human tumor tissues are more preferred. Cancer stem cells to be detected using the present invention are preferably those which reconstitute the hierarchical structure of cancer tissues. For example, cancer cell lines are prepared by grafting cancer tissues from which the detected cancer stem cells have been collected, into, preferably, nonhuman animals, and passaging them in such animals, and one can test whether the established cancer cell lines reconstitute the hierarchical structure of the cancer tissues. One can test whether the hierarchical structure of cancer tissues is reproduced by NOG-established cancer cell lines prepared by grafting and passaging cancer tissues in nonhuman animals, more preferably immunodeficient animals, and still more preferably NOG mice which lack functional T cells, B cells, and natural killer cells. Alternatively, cancer stem cells to be detected using the present invention can be a spheroid (cell mass) formed by spheroid culture. “Spheroid culture” means that cancer stem cells are inoculated in a culture vessel such as non-adherent or low-adherent cell culture flasks, plates, or dishes using a medium capable of culturing cancer stem cells, and then the cells are cultured under a three-dimensionally floating condition. A cell mass formed by this method is called “spheroid”. NOG-established cancer cell lines can be generated by a method known to those skilled in the art, for example, the method described in Fujii E. et al., Pathol Int. 2008; 58: 559-567. Human colorectal cancer, stomach cancer, lung cancer, breast cancer, pancreatic cancer, or the like is resected surgically. After mechanically mincing it with scissors, the cancer is grafted subcutaneously in NOG mice and passaged to establish cell lines. Even after passages, NOG-established cancer cell lines maintain the properties of the original human cancer tissues. In the present invention, cancer stem cells can be selected by using cell markers. Cell markers used in the present invention include, for example, leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5), CD133, CD44, EpCAM, CD166, CD24, CD26, and CD29. The present invention relates to molecules expressed in cancer stem cells that are positive for the expression of the cell marker Lgr5, and are adherent and highly proliferative under serum-free culture conditions. Hereinafter, such cancer stem cells are also referred to as “high proliferative Lgr5-positive cancer stem cells”. The present invention also relates to molecules expressed in cancer stem cells that are negative for the expression of the cell marker Lgr5, and non-adherent and poorly proliferative under serum-free culture conditions. Hereinafter, such cancer stem cells are also referred to as “low proliferative Lgr5-negative cancer stem cells”. Any culture media or liquids can be used to culture cancer stem cells of the present invention as long as they are serum-free media and capable of culturing cancer stem cells. There is no particular limitation on the culture media or liquids. For example, it is possible to use conventional basal media or mixtures thereof that are supplemented with EGF, bFGF, hLIF, HGF, NGF, NSF-1, TGF β, TNFα, heparin, BSA, insulin, transferrin, putrescine, selenite, progesterone, hydrocortisone, D-(+)-glucose, sodium bicarbonate, HEPES, L-glutamine, or N-acetylcysteine. The concentration of EGF is not particularly limited; however, it ranges from 0.1 to 100 ng/ml, preferably from 0.5 to 50 ng/ml, and more preferably from 1 to 20 ng/ml. The concentration of bFGF is not particularly limited; however, it ranges from 0.1 to 100 ng/ml, preferably from 0.5 to 50 ng/ml, and more preferably from 1 to 20 ng/ml. The concentration of hLIF is not particularly limited; however, it ranges from 0.1 to 100 ng/ml, preferably from 0.5 to 50 ng/ml, and more preferably from 1 to 20 ng/ml. The concentration of HGF is not particularly limited; however, it ranges from 0.1 to 100 ng/ml, preferably from 1 to 50 ng/ml. The concentration of NGF is not particularly limited; however, it ranges from 0.1 to 100 ng/ml, preferably from 1 to 50 ng/ml. The concentration of NSF-1 is not particularly limited; however, it ranges from 0.1 to 100 ng/ml, preferably from 1 to 50 ng/ml. The concentration of TGFβ is not particularly limited; however, it ranges from 0.1 to 100 ng/ml, preferably from 1 to 50 ng/ml. The concentration of TNFα is not particularly limited; however, it ranges from 0.1 to 100 ng/ml, preferably from 1 to 50 ng/ml. The concentration of heparin is not particularly limited; however, it ranges from 10 ng/ml to 10 μg/ml, preferably from 2 to 5 μg/ml. The concentration of BSA is not particularly limited; however, it ranges from 0.1 to 10 mg/ml, preferably from 1 to 8 mg/ml. The concentration of insulin is not particularly limited; however, it ranges from 1 to 100 μg/ml, preferably from 10 to 50 μg/ml. The concentration of transferrin is not particularly limited; however, it ranges from 10 to 500 μg/ml, preferably from 50 to 200 μg/ml. The concentration of putrescine is not particularly limited; however, it ranges from 1 to 50 μg/ml, preferably from 10 to 20 μg/ml. The concentration of selenite is not particularly limited; however, it ranges from 1 to 50 nM, preferably from 20 to 40 nM. The concentration of progesterone is not particularly limited; however, it ranges from 1 to 50 nM, preferably from 10 to 30 nM. The concentration of hydrocortisone is not particularly limited; however, it ranges from 10 ng/ml to 10 μg/ml, preferably from 100 ng/ml to 1 μg/ml. The concentration of D-(+)-glucose is not particularly limited; however, it ranges from 1 to 20 mg/ml, preferably from 5 to 10 mg/ml. The concentration of sodium bicarbonate is not particularly limited; however, it ranges from 0.1 to 5 mg/ml, preferably from 0.5 to 2 mg/ml. The concentration of HEPES is not particularly limited; however, it ranges from 0.1 to 50 mM, preferably from 1 to 20 mM. The concentration of L-glutamine is not particularly limited; however, it ranges from 0.1 to 10 mM, preferably from 1 to 5 mM. The concentration of N-acetylcysteine is not particularly limited; however, it ranges from 1 to 200 μg/ml, preferably from 10 to 100 μg/ml. Known basal culture liquids, which are not particularly limited as long as they are suitable for culturing cancer cells from which cancer stem cells are derived, include, for example, DMEM/F12, DMEM, F10, F12, IMDM, EMEM, RPMI-1640, MEM, BME, Mocoy's 5A, and MCDB131. Of them, DMEM/F12 is preferred. The most preferred stem cell media include DMEM/F12 medium supplemented with 20 ng/ml human EGF, 10 ng/ml human bFGF, 4 μg/ml heparin, 4 mg/ml BSA, 25 μg/ml human insulin, and 2.9 mg/ml glucose where each concentration a final concentration. As described herein, high proliferative Lgr5-positive cancer stem cells have the properties of mesenchymal cells. Meanwhile, as described herein, low proliferative Lgr5-negative cancer stem cells have the properties of epithelial cells. Herein, “epithelial cells” refers to cells that constitute epithelial tissues in the living body. In the present invention, the origin of cancer stem cells is not particularly limited; however, they are preferably derived from a solid cancer, more preferably a gastrointestinal cancer. Gastrointestinal cancers include, for example, esophageal cancer, stomach cancer, duodenal cancer, pancreatic cancer, bile duct cancer, gallbladder cancer, biliary tract cancer, colorectal cancer, colon cancer, and rectal cancer. A preferred gastrointestinal cancer is colorectal cancer. Furthermore, in the present invention, cancer stem cells are preferably positive for one or more of the cell markers CD133, CD44, EpCAM, CD166, CD24, CD26, and CD29, more preferably positive for CD133, CD44, EpCAM, CD166, CD24, CD26, and CD29. In addition, in the present invention, acetaldehyde dehydrogenase (ALDH) activity can be used as a cell marker. In the present invention, Lgr5-positive adherent cancer stem cells are positive for the ALDH activity cell marker, whereas Lgr5-negative cancer stem cells are negative for ALDH activity. In the present invention, one or more of HLA-DMA, TMEM173, ZMAT3, and GPR110 can also be used as cell markers. Lgr5-positive adherent cancer stem cells are negative for any of the cell markers HLA-DMA, TMEM173, ZMAT3, and GPR110, while Lgr5-negative cancer stem cells are positive for any of the cell markers HLA-DMA, TMEM173, ZMAT3, and GPR110. In the present invention, cancer stem cells preferably have the feature of reconstituting the hierarchical structure of cancer tissues. Herein, “hierarchical structure” means that some of the unique and characteristic structures observed in a normal tissue are detected histopathologically in the structure of a tumor originated from the tissue. In general, highly-differentiated cancers reconstitute the hierarchical structure to a high degree. For example, lumen formation and mucous cells are observed in the case of tumors of glandular lumen-forming organs (stomach cancer, colorectal cancer, pancreatic cancer, liver cancer, bile duct cancer, breast cancer, lung adenocarcinoma, prostatic cancer, etc.). In the case of tumors that form squamous epithelial structures (squamous cell carcinoma of lung, skin, vaginal mucosa, etc.), layer structure formation, the tendency to keratosis, and such are observed in the epithelium. On the other hand, poorly-differentiated cancers insufficiently reconstitute the hierarchical structure, and they are said to be highly atypical (Kumar V, Abbas A K, Fausio N. Robbins and Cotran Pathologic Basis of Disease. 7th Ed. Unit I: General Pathology, 7: Neoplasia, Biology of tumor growth: Benign and malignant neoplasms. 272-281, 2005). Since the hierarchical structure is considered to be reconstituted as a result of various biological reactions, cancer stem cells that reconstitute it are thought to be highly useful. “Reconstitution of the hierarchical structure” means that the unique and characteristic structure possessed by the original cancer stem cells is also observed even after isolation or induction of cancer stem cells. Furthermore, in the present invention, cancer stem cells preferably have the ability of epithelial-mesenchymal transition (EMT). Herein, the ability of epithelial-mesenchymal transition means both that epithelial cells transition into mesenchymal cells by obtaining their characteristics, and that mesenchymal cells transition into epithelial cells by obtaining their characteristics. EMT does not occur in normal cells except during the process of embryogenesis. Epithelial cells, which are bound together tightly and exhibit polarity, change into mesenchymal cells that are bound together more loosely, exhibit a loss of polarity, and have the ability to move. These mesenchymal cells can spread into tissues around the primary tumor, and also separate from the tumor, invade blood and lymph vessels, and move to new locations where they divide and form additional tumors. Drug resistance, metastasis, or recurrence of cancer can be explained by such additional tumor formation. Furthermore, the present invention provides pharmaceutical compositions comprising as an active ingredient an antibody that binds to a molecule expressed in a substantially homogeneous cancer stem cell population comprising the above cancer stem cells of the present invention. “Substantially homogeneous” means that, when immunodeficient animals are grafted with 1000 cells, 100 cells, or 10 cells and analyzed for the frequency of formation of cancer cell populations using Extreme Limiting Dilution Analysis (Hu Y & Smyth G K., J Immunol Methods. 2009 Aug. 15; 347(1-2): 70-8) utilizing, for example, the method described in Hu Y & Smyth G K., J Immunol Methods. 2009 Aug. 15; 347 (1-2):70-8 or Ishizawa K & Rasheed Z A. et al., Cell Stem Cell. 2010 Sep. 3; 7(3):279-82, the frequency of cancer stem cells is 1/20 or more, preferably 1/10 or more, more preferably 1/5 or more, even more preferably 1/3 or more, still more preferably 1/2 or more, and yet more preferably 1/1. In the present invention, cancer stem cell populations can be prepared, for example, by culturing cells or a group of cells containing the cancer stem cells described herein. Herein, “adherent culture” means that, after seeding cells into culture vessels for adherent culture, the adhered cells are cultured and passaged while non-adherent cells are removed. The cells grown to confluency are detached with Accutase and passaged into fresh adherent culture flasks, adherent culture plates, or adherent culture dishes for further culture. Culture vessels for adherent culture are not particularly limited as long as they are used for adherent culture. It is possible to appropriately select and use flasks for adherent culture or highly adherent flasks, plates for adherent culture or highly adherent plates, flat-bottomed plates for adherent culture or highly adherent flat-bottomed plates, dishes for adherent culture or highly adherent dishes, etc. Media used for adherent culture are not particularly limited; however, it is preferable to use serum-free stem cell culture media. Herein, “adherent” refers to the property of cells to adhere to culture vessels for adherent culture when they are cultured in the vessels. Herein, “suspension culture” means that, after seeding cells into culture vessels for suspension culture, the floating cells are cultured and passaged while adherent cells are removed. The cells grown to confluency are passaged into fresh low adherent cell culture flasks, ultra-low adherent cell culture flasks, low adherent plates, ultra-low adherent plates, low adherent dishes, or ultra-low adherent dishes for further culture. Culture vessels for suspension culture are not particularly limited as long as they are used for suspension culture. It is possible to appropriately select and use low adherent cell culture flasks, ultra-low adherent cell culture flasks, low adherent plates, ultra-low adherent plates, low adherent dishes, ultra-low adherent dishes, etc. Media used for suspension culture are not particularly limited; however, it is preferable to use serum-free stem cell culture media. A cell group containing cancer stem cells are preferably expanded before performing adherent or suspension culture. Herein, “non-adherent” refers to the property of cells to be cultured in a floating state without adherence to culture vessels for suspension culture when the cells are cultured in the vessels. “Expansion of a cell group” means, for example, proliferation by spheroid culture or grafting and passaging in nonhuman animals, but is not particularly limited thereto. As described herein, for nonhuman animals, immunodeficient animals can be used for grafting since they are unlikely to have rejection reactions. Immunodeficient animals preferably used include nonhuman animals that lack functional T cells, for example, nude mice and nude rats, and nonhuman animals that lack both functional T and B cells, for example, SCID mice and NOD-SCID mice. It is more preferably to use mice that lack T, B, and NK cells and have excellent transplantability, including, for example, NOG mice. Regarding the weekly age of nonhuman animals, for example, 4 to 100-week-old athymic nude mice, SCID mice, NOD-SCID mice, or NOG mice are preferably used. NOG mice can be prepared, for example, by the method described in WO 2002/043477, and are available from the Central Institute for Experimental Animals or the Jackson Laboratory (NSG mice). Cells to be grafted may be any cells, including cell masses, tissue fragments, individually dispersed cells, cells cultured after isolation, and cells isolated from a different animal into which the cells have been grafted; however, dispersed cells are preferred. The number of grafted cells may be 106or less; however, it is acceptable to graft more cells. With respect to the grafting site, subcutaneous grafting is preferred because the graft technique is simple. The grafting site is not particularly limited, and it is preferable to select an appropriate grafting site depending on the animal used. There is no particular limitation on the grafting operation of NOG-established cancer cell lines, and the cells can be grafted by conventional grafting operations. Cancer stem cells or a cancer stem cell population can be prepared, for example, by collecting cancer tissues from patients and culturing the tissues in a serum-free stem cell culture medium under adherent or floating culture conditions. Alternatively, cancer tissues collected from patients can be spheroid-cultured, and then cultured in a serum-free stem cell culture medium under adherent or floating culture conditions to prepare cancer stem cells or a cancer stem cell population Alternatively, cancer tissues collected from patients can be grafted and passaged in nonhuman animals, and then cultured in a serum-free stem cell culture medium under adherent or floating culture conditions to prepare cancer stem cells or a cancer stem cell population. Alternatively, it is possible to use a method in which cancer tissues collected from patients are grafted and passaged in NOG mice to prepare NOG-established cancer cell lines, and they are cultured in a serum-free stem cell culture medium under adherent or suspension culture conditions. Cancer stem cells and cancer stem cell populations of the present invention can be used in methods of screening for pharmaceutical agents, anti-cancer agents, or the like. In an embodiment of methods of screening for pharmaceutical agents, the present invention provides methods comprising the steps of: (a) preparing a substantially homogeneous cancer stem cell population comprising an Lgr5-positive adherent cancer stem cell; (b) contacting a test substance with the cancer stem cell population or a cancer stem cell comprised in the cancer stem cell population; and (c) detecting a change in a biological property of the cancer stem cell population or cancer stem cell contacted with the test substance. In these methods, first, a substantially homogeneous cancer stem cell population containing Lgr5-positive adherent cancer stem cells or Lgr5-negative cancer stem cells is prepared. Then, a test substance is contacted with the prepared cancer stem cell population or cancer stem cells contained in the cancer stem cell population. In these methods, there is no particular limitation on the method for contacting a test substance with a cancer stem cell population or cancer stem cells contained in the cancer stem cell population. For example, a test substance may be contacted with cultured cells of a cancer stem cell population or cancer stem cells contained in the cancer stem cell population. This treatment can be carried out by adding a test substance to a cell culture medium or cell extract. When a test substance is a protein, this treatment can be performed, for example, as follows: a vector comprising a DNA encoding the protein is introduced into a cancer stem cell population or cancer stem cells contained in the cancer stem cell population; or the vector is added to a cell extract of a cancer stem cell population or cancer stem cells contained in the cancer stem cell population. Alternatively, it is possible, for example, to use the two-hybrid method utilizing yeast, animal cells, or the like. In these methods, then, a change in a biological property of the cancer stem cell population or cancer stem cells treated with the test substance is detected. Such a change in a biological property includes, for example, a change in the proliferation ability, a change in the viable cell count, a change in a tissue structure characteristic of the process of cancer progression of the cancer stem cell population or cancer stem cells, and a change in the expression of a DNA, RNA, protein, or metabolite in the cancer stem cell population or cancer stem cells. A change in a biological property can be detected, for example, by the methods described below. There is no particular limitation on the assessment of the expression of DNAs, RNAs, proteins, peptides, and metabolites; the expression can be assessed by conventional expression assessment methods. RNAs include microRNAs, siRNAs, tRNAs, snRNAs, mRNAs, and non-coding RNAs. For example, mRNAs of a gene are extracted according to a conventional method. Using the mRNAs as a template, the transcriptional level of the gene can be determined using the Northern hybridization or RT-PCR method. DNA array techniques can also be used to determine the expression level of the gene. Alternatively, fractions containing a protein encoded by a gene are collected according to a conventional method. The translational level of the gene can be determined by detecting the protein expression by an electrophoresis method such as SDS-PAGE. The translational level of a gene can also be determined by performing the Western blotting method using an antibody against a protein and detecting the protein expression. These methods can be used to screen for pharmaceutical agents (pharmaceutical compositions). The DNAs, RNAs, and proteins that are contained in a cancer stem cell population or cancer stem cells and which are characteristic of the process of cancer progression of the cancer stem cell population or cancer stem cells, preferably include the proteins or polypeptides of any one of SEQ ID NOs: 1 to 3 and 5 to 7, and polynucleotides encoding the proteins or polypeptides. For example, when there is no change in a biological property of a cancer stem cell population or cancer stem cells, or the degree of the change is reduced after treatment with a test substance compared to before the treatment, the test substance is expected to be useful as a pharmaceutical agent (pharmaceutical composition) that has the activity of suppressing cancer recurrence or metastasis (for example, an agent for suppressing cancer recurrence, an agent for post-chemotherapy adjuvant therapy, an agent for postoperative adjuvant therapy, an anti-cancer agent, or an agent for suppressing cancer metastasis). Such test substances can be selected as effective substances that have the therapeutic or preventive effect against cancerous diseases. Such pharmaceutical agents (pharmaceutical compositions) having the activity of suppressing cancer progression are used as an agent for suppressing cancer recurrence, an agent for post-chemotherapy adjuvant therapy, an agent for postoperative adjuvant therapy, an anti-cancer agent, or an agent for suppressing cancer metastasis. Anti-cancer agents of the present invention may be used against, for example, cancers resistant to pharmaceutical agents or chemotherapeutic agents. Specifically, pharmaceutical agents (pharmaceutical compositions) of the present invention also include therapeutic agents against drug-resistant or chemotherapeutic agent-resistant cancers. In the present invention, the above pharmaceutical agents (pharmaceutical compositions) are not particularly limited to anti-cancer agents or agents for suppressing metastasis or recurrence, and they can also be used as an agent for inhibiting angiogenesis or cell growth. Furthermore, the pharmaceutical agents (pharmaceutical compositions) of the present invention may be used simultaneously with chemotherapeutic agents or after treatment with chemotherapeutic agents. The pharmaceutical agents are not particularly limited, and they include proteinaceous agents, nucleic acid agents, low-molecular-weight agents, and cellular agents. In another embodiment of the screening methods, the present invention provides methods of screening for pharmaceutical agents (pharmaceutical compositions), which comprise the steps of: (a) preparing a substantially homogeneous cancer stem cell population comprising an Lgr5-negative non-adherent cancer stem cell; (b) contacting a test substance with the cancer stem cell population or a cancer stem cell comprised in the cancer stem cell population; and (c) detecting a change in a biological property of the cancer stem cell population or cancer stem cell contacted with the test substance. In these methods, first, a substantially homogeneous cancer stem cell population containing Lgr5-negative non-adherent cancer stem cells is prepared. Then, the prepared cancer stem cell population or cancer stem cells contained in the cancer stem cell population are treated with a test substance. Next, a change in a biological property of the cancer stem cell population or cancer stem cells treated with the test substance is detected. The DNAs, RNAs, and proteins that are contained in such a cancer stem cell population or cancer stem cells, and which are characteristic of the process of cancer progression of the cancer stem cell population or cancer stem cells, preferably include the proteins or polypeptides of any one of SEQ ID NOs: 1 to 3 and 5 to 9, and polynucleotides encoding the proteins or polypeptides. In a non-limiting embodiment of the present invention, the proteins or polypeptides of SEQ ID NOs: 1 to 3 and 5 to 7, and polynucleotides encoding the proteins or polypeptides may be used. In another non-limiting embodiment of the present invention, the protein or polypeptide of SEQ ID NO: 7 or 8, and polynucleotides encoding the protein or polypeptide may be used. Pharmaceutical agents (pharmaceutical compositions) that are obtained by the screening methods are not particularly limited, and they can be used as anti-cancer agents. Specifically, when there is no change in a biological property of a cancer stem cell population or cancer stem cells, or the degree of the change is reduced after treatment with a test substance compared to before the treatment, the test substance is expected to be useful as a pharmaceutical agent that has the activity of suppressing cancer recurrence or metastasis (for example, an agent for suppressing cancer recurrence, an agent for post-chemotherapy adjuvant therapy, an agent for postoperative adjuvant therapy, an anti-cancer agent, or an agent for suppressing cancer metastasis). Such test substances may be selected as effective substances that have the therapeutic or preventive effect against cancerous diseases. Such pharmaceutical agents (pharmaceutical compositions) having the activity of suppressing cancer progression are used as an agent for suppressing cancer recurrence, an agent for post-chemotherapy adjuvant therapy, an agent for postoperative adjuvant therapy, an anti-cancer agent, or an agent for suppressing cancer metastasis. Furthermore, pharmaceutical agents (pharmaceutical compositions) of the present invention include cancer therapeutic agents against Lgr5-negative cancers, which contain as an active ingredient at least an antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 9. The Lgr5-negative cancers include those resistant to drugs or chemotherapeutic agents. Still another embodiment of the screening methods of the present invention includes methods that use nonhuman animals administered with a test substance, and a cancer stem cell population of the present invention or cancer stem cells contained in the cancer stem cell population. Specifically, the present invention provides methods of screening for pharmaceutical agents (pharmaceutical compositions), which comprise the steps of: (a) preparing a substantially homogeneous cancer stem cell population comprising an Lgr5-positive adherent cancer stem cell; (b) administering a nonhuman animal with a test substance and the cell population or a cancer stem cell comprised in the cancer stem cell population; and (c) detecting tumor formation in the nonhuman animal. In these methods, first, a substantially homogeneous cancer stem cell population containing Lgr5-positive adherent cancer stem cells is prepared. Then, nonhuman animals are administered with a test substance, and the cancer stem cell population prepared or cancer stem cells contained in the cancer stem cell population. In these methods, the method for administering a test substance to nonhuman animals is not particularly limited. Oral administration, or parenteral administration such as subcutaneous, intravenous, local, transdermal, or transintestinal (transrectal) administration can be appropriately selected depending on the type of a test substance to be administered. Furthermore, in these methods, there is no particular limitation on the method for administering a cancer stem cell population or cancer stem cells to nonhuman animals, and an appropriate method can be selected depending on the cell population to be administered. The preferred method is subcutaneous or intravenous administration. In these methods, then, tumor formation is detected in the nonhuman animals. The assessment of a test substance can be performed as follows: tissues administered with a test substance and a cancer stem cell population or cancer stem cells are excised from nonhuman animals, and then histological features of the tissues are observed to determine the presence or absence of tumor formation. When tumor formation is not detected, the test substance is expected to be useful as a pharmaceutical agent having the activity of suppressing cancer progression or metastasis (for example, an anti-cancer agent or an agent for suppressing cancer metastasis or recurrence), and the test substance can be selected as an effective substance that has the therapeutic or preventive effect against cancerous diseases. That is, pharmaceutical agents (pharmaceutical compositions) obtained by the screening methods are not particularly limited, and can be used as an anti-cancer agent, or an agent for suppressing cancer metastasis or recurrence. “Test substances” used in the methods of the present invention are not particularly limited, and include, for example, single compounds such as natural compounds, organic compounds, inorganic compounds, proteins, antibodies, peptides, and amino acids, as well as compound libraries, expression products of gene libraries, cell extracts, cell culture supernatants, products of fermenting microorganisms, extracts of marine organisms, plant extracts, prokaryotic cell extracts, unicellular eukaryote extracts, and animal cell extracts. These may be purified products or crude purified products such as extracts of plants, animals, and microorganisms. Also, methods for producing test substances are not particularly limited; test substances may be isolated from natural materials, synthesized chemically or biochemically, or prepared by genetic engineering. It is also possible to appropriately use antisense and RNAi molecules that are designed by known methods based on partial sequences of polynucleotides encoding the protein of any one of SEQ ID NOs: 1 to 3 and 5 to 9. If needed, the above test substances can be appropriately labeled and used. Labels include, for example, radiolabels and fluorescent labels. Mixtures of an above-mentioned test substance and multiple kinds of such labels are included in the test substances of the present invention. Furthermore, the present invention provides pharmaceutical agents such as vaccines comprising a partial peptide of the protein of any one of SEQ ID NOs: 1 to 3 and 5 to 9, and methods of screening for vaccines. Such screening methods preferably include methods for determining the cytotoxic activity targeted to cancer stem cells disclosed in the present application using cytotoxic T cells (CTL) or the like induced with a cancer vaccine of the present invention in vitro. Specifically, adherent and non-adherent cells are separated from peripheral blood mononuclear cells (PBMCs) collected by centrifugation of human peripheral blood in Ficoll-Conray density gradient. The adherent cells are incubated with 100 ng/ml GM-CSF (Novartis) and 10 IU/ml IL-4 (GIBCO-BRL) in AIM-V (GIBCO), and then the cells are used as antigen-presenting cells (APC). Meanwhile, the non-adherent cells are incubated with 30 to 100 IU/ml recombinant IL-4 (Ajinomoto) in AIM-V. On day 7 to 10, a partial peptide of the protein of any one of SEQ ID NOs: 1 to 3 and 5 to 9 provided by the present invention is added (at a final concentration of 30 μg/ml) to APC. On the following day, recombinant TNF-α and IFN-α (Sumitomo Pharma Co.) are added for APC maturation. Then, CD8-positive cells isolated from autologous non-adherent cells are mixed with irradiated APC in IL-2-free AIM-V. After two days of incubation, IL-2 (Takeda Pharmaceutical Company) is added at a final concentration 100 IU/ml to the culture. The CD8-positive cells are stimulated every seven days using, as APC, autologous PHA blasts (PHA-stimulated T cells) that have been stimulated with the T cell mitogen PHA. A fresh medium containing 100 IU/ml IL-2 is added to the culture at every time point of stimulation. CTL on day 28 is used for the activity assay. High proliferative Lgr5-positive cancer stem cells and low proliferative Lgr5-negative cancer stem cells that are provided by the present invention can be used as target cells of CTL. The cytotoxic activity can be assessed by determining sCr-sodium chromate uptake activity by a measurement method similar to that of ADCC activity. Furthermore, pharmaceutical agents selected by the screening methods of the present invention may be further screened as necessary for more effective and practical preventive or therapeutic active substances by conducting additional drug effectiveness tests and safety tests, and further conducting clinical tests in human cancer patients. Based on results of structural analysis of pharmaceutical agents thus selected, they can be industrially manufactured by chemical synthesis, biochemical synthesis (fermentation), or genetic engineering. “High proliferative ability” means that the doubling time is 6 days or less, preferably 4 days or less, and more preferably 3 days or less when cells are cultured in a serum-free medium supplemented with EGF and FGF using the method described herein. “Low proliferative ability” means that the doubling time is 7 days or more, preferably 14 days or more, and more preferably there is no significant proliferation when cells are cultured in a serum-free medium supplemented with EGF and FGF using the method described herein. When preparing such high proliferative Lgr5-positive cancer stem cells and low proliferative Lgr5-negative cancer stem cells, the cells can be separated using the cell marker Lgr5. The separation methods include the following: methods in which a cell population containing cancer stem cells is isolated by using an anti-Lgr5 antibody; methods in which a substantially homogeneous cancer stem cell population is first prepared by culturing a population containing cancer stem cells under adherent or suspension culture conditions, and then the population is isolated by using an anti-Lgr5 antibody; and methods in which a substantially homogeneous cancer stem cell population is first prepared by culturing a population containing cancer stem cells in a medium with or without a growth inhibitor under adherent culture conditions, and then the population is isolated by using an anti-Lgr5 antibody. Any of the above methods may be used in the present invention. Preferably, cells are isolated from cancer tissues after three or more passages in NOG mice, and cultured in a serum-free stem cell culture media under adherent culture conditions to prepare high proliferative Lgr5-positive cancer stem cells. Then, low proliferative Lgr5-negative cancer stem cells can be prepared as follows. The resulting Lgr5-positive cancer stem cells are maintained under various stresses such as a contact with a growth inhibitor, for example, treatment with irinotecan (culture for three days in a serum-free stem cell medium supplemented with 10 μg/ml irinotecan). Furthermore, the present invention provides methods of screening for pharmaceutical agents, which comprise contacting a test substance with cancer stem cells that differ in the proliferation ability, which are induced by the methods provided by the present invention. Specifically, the present invention provides methods of screening for pharmaceutical agents, which comprises detecting a change in a biological property of cancer stem cells by contacting a test substance with high or low proliferative cancer stem cells induced by a method for converting low proliferative cancer stem cells to high proliferative cancer stem cells, or converting high proliferative cancer stem cells to low proliferative cancer stem cells. Specifically, as described herein, low proliferative cancer stem cells can be prepared by maintaining high proliferative cancer stem cells under various stresses such as a suspension culture or in contact with a growth inhibitor. For example, high proliferative cancer stem cells can be converted to low proliferative cancer stem cells by culturing high proliferative cancer stem cells under suspension culture conditions. Alternatively, high proliferative cancer stem cells can be converted to low proliferative cancer stem cells by culturing high proliferative cancer stem cells in low adherent or ultra-low adherent cell culture vessels such as low adherent plates, ultra-low adherent plates, low adherent dishes, ultra-low adherent dishes, low adherent flasks, or ultra-low adherent cell culture flasks. In other words, low proliferative cancer stem cells can be prepared by culturing high proliferative cancer stem cells in low adherent or ultra-low adherent cell culture vessels such as low adherent plates, ultra-low adherent plates, low adherent dishes, ultra-low adherent dishes, low adherent flasks, or ultra-low adherent cell culture flasks. In a non-limiting embodiment, high proliferative cancer stem cells can be converted to low proliferative cancer stem cells by using a growth inhibitor such as 5-FU or irinotecan. Specifically, low proliferative cancer stem cells can be produced by exposing high proliferative cancer stem cells to a growth inhibitor such as 5-FU or irinotecan. Exposure to a growth inhibitor can be achieved under any condition such as in vitro culture or inside the body of grafted nonhuman animals. In this case, those skilled in the art can select an appropriate exposure dose of a cell growth inhibitor for cancer stem cells. Alternatively, high proliferative cancer stem cells can be prepared by re-seeding low proliferative cancer stem cells into a medium without a growth inhibitor such as 5-FU or irinotecan. In another non-limiting embodiment, high proliferative cancer stem cells can be produced by discontinuing administration of a growth inhibitor to nonhuman animals having low proliferative cancer stem cells. Furthermore, low proliferative cancer stem cells can be cultured under adherent culture conditions to convert them to high proliferative cancer stem cells. Alternatively, low proliferative cancer stem cells can be converted to high proliferative cancer stem cells by culturing low proliferative cancer stem cells in a non-low-adherent but highly adherent cell culture vessel such as a flat-bottomed plate, plate, adherent culture plate, adherent culture flask, dish, or adherent culture dish. That is, high proliferative cancer stem cells can be produced by culturing low proliferative cancer stem cells in a non-low-adherent but highly adherent cell culture vessel such as a flat-bottomed plate, plate, adherent culture plate, adherent culture flask, dish, or adherent culture dish. The present invention also relates to cancer cell detection reagents. Cancer cell detection reagents of the present invention preferably contain as an active ingredient at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 9 (protein composed of the amino acid sequence of any one of SEQ ID NOs: 1 to 3 and 5 to 9). In another embodiment, the reagents of the present invention include reagents for detecting Lgr5-positive cancer cells, which preferably contain at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 7 (protein composed of the amino acid sequence of any one of SEQ ID NOs: 1 to 3 and 5 to 7). In still another embodiment, the reagents of the present invention include reagents for detecting Lgr5-negative cancer cells, which preferably contain at least one antibody that binds to the protein of any one of SEQ ID NOs: 1 to 3 and 5 to 9 (protein composed of the amino acid sequence of any one of SEQ ID NOs: 1 to 3 and 5 to 9). Growth Inhibitors In a non-limiting embodiment, preferred growth inhibitors include DNA-damaging agents, antimitotic agents, and/or anti-metabolites. Such a DNA-damaging agent may be an alkylating reagent, a topoisomerase inhibitor, and/or a DNA intercalator. Examples of preferred growth inhibitors include, but are not limited to, carboplatin (DNA alkylating reagent), etoposide (topoisomerase II inhibitor), doxorubicin (DNA intercalator), docetaxel (antimitotic agent), and Gemzar (gemcitabine; anti-metabolite). Alkylating reagents can be at least one reagent selected from the following. Specifically, it is possible to use at least one alkylating reagent selected from: chlorambucil, cyclophosphamide, ifosfamide, mechlorethamine, melphalan, uracil mustard, thiotepa, busulfan, carmustine, lomustine, streptozocin, carboplatin, cisplatin, satraplatin, oxaliplatin, altretamine, ET-743, XL119 (becatecarin), dacarbazine, chlormethine, bendamustine, trofosfamide, uramustine, fotemustine, nimustine, prednimustine, ranimustine, semustine, nedaplatin, triplatin tetranitrate, mannosulfan, treosulfan, temozolomide, carboquone, triaziquone, triethylene melamine, procarbazine, etc. Topoisomerase inhibitors can be at least one inhibitor selected from the following. Specifically, it is possible to use at least one topoisomerase inhibitor selected from: doxorubicin (Doxil), daunorubicin, epirubicin, idarubicin, anthracenedione (Novantrone), mitoxantrone, mitomycin C, bleomycin, dactinomycin, plicatomycin, irinotecan (Camptosar), camptothecin, rubitecan, belotecan, etoposide, teniposide, topotecan (Hycamptin), etc. At least one topoisomerase inhibitor selected from the following can be used as a DNA intercalator: proflavin, doxorubicin (adriamycin), daunorubicin, dactinomycin, thalidomide, etc. Antimitotic agents can be at least one agent selected from the following. Specifically, it is possible to use at least one topoisomerase inhibitor selected from: paclitaxel (Abraxane)/Taxol, docetaxel (Taxotere), BMS-275183, Xyotax, Tocosal, vinorlebine, vincristine, vinblastine, vindesine, vinzolidine, etoposide (VP-16), teniposide (VM-26), ixavepilone, larotaxel, ortataxel, tesetaxel, ispinesib, etc. Anti-metabolites can be at least one inhibitor selected from the following. Specifically, it is possible to use at least one topoisomerase inhibitor selected from: fluorouracil (5-FU), floxuridine (5-FUdR), methotrexate, Xeloda, Arranon, leucovorin, hydroxyurea, thioguanine (6-TG), mercaptopurine (6-MP), cytarabine, pentostatin, fludarabine phosphate, cladribine (2-CDA), asparaginase, gemcitabine, pemetrexed, bortezomib, aminopterin, raltitrexed, clofarabine, enocitabine, sapacitabine, azacytidine, etc. Furthermore, the present invention relates to methods of screening for anti-cancer drugs, which use cancer stem cells isolated or induced by the above methods of the present invention. The present invention also relates to methods for assessing compounds, which use cancer stem cells isolated or induced by the above methods of the present invention. Method for Detecting Cancer Stem Cells Furthermore, the present invention provides methods for detecting, identifying, or quantifying the presence of cancer stem cells of the present invention. Specifically, the present invention provides methods for detecting, identifying, or quantifying the presence of cancer stem cells or substantially homogeneous cancer stem cell populations of the present invention, which comprise the steps of:(a) preparing a sample obtained from a cancer patient; and(b) contacting a sample with an anti-Lgr5 antibody. In these methods, first, samples obtained from cancer patients are prepared. In the present invention, a “sample” is not particularly limited as long as it is preferably an organ or tissue derived from a cancer patient. It is possible to use a frozen or unfrozen organ or tissue. Such samples include, for example, cancer (tumor) tissues isolated from cancer patients. In these methods, a sample is then contacted with an anti-Lgr5 antibody. Methods for detecting, identifying, or quantifying the presence of above-described cancer stem cells or substantially homogeneous cancer stem cell populations of the present invention can be used in, for example, cancer diagnosis, selection of cancer patients, prediction or assessment of the effectiveness of an agent (pharmaceutical composition), treatment monitoring, and cancer imaging. Specifically, for example, organs or tissues are isolated from cancer patients, and specimens are prepared. The specimens can be used to detect, identify, or quantify the presence of cancer stem cells. Specimens can be appropriately prepared by using known methods, for example, the PFA-AMeX-Paraffin method (WO 09/078386). The samples include, for example, frozen or unfrozen organs or tissues. First, samples from cancer patients are fixed in a PFA solution. “PFA solution” refers to a cell fixation solution which is an aqueous solution of 1 to 6% paraformaldehyde combined with a buffer such as phosphate buffer. It is preferable to use 4% PFA fixation solution (4% paraformaldehyde/0.01 M PBS (pH7.4)). For fixation with a PFA fixation solution, organs or tissues of interest are immersed in a PFA solution containing 1 to 6%, preferably 4% paraformaldehyde, at 0 to 8° C., preferably at about 4° C., for 2 to 40 hours, preferably for 6 to 30 hours. Then, fixed organs or tissues are washed with phosphate buffered saline or such. Washing may be carried out after excising portions from the observed organs or tissues. Organs or tissues thus prepared are then embedded in paraffin by the AMeX method. The AMeX method is a paraffin embedding method with a series of the following steps: cold acetone fixation, dehydration with acetone, clearing in methylbenzoate and xylene, and paraffin embedding. Specifically, tissues are immersed in acetone at −25 to 8° C., preferably at −20 to 6° C., for 2 to 24 hours, preferably for 4 to 16 hours. Then, the tissues in acetone are warmed to room temperature. Alternatively, organs or tissues are transferred into acetone at room temperature. Then, dehydration is performed for 0.5 to 5 hours, preferably 1 to 4 hours at room temperature. Subsequently, the organs or tissues are cleared by immersion in methylbenzoate at room temperature for 0.5 to 3 hours, preferably for 0.5 to 2 hours, followed by immersion in xylene at room temperature for 0.5 to 3 hours, preferably 0.5 to 2 hours. Next, the organs or tissues are embedded in paraffin by penetration at 55 to 65° C., preferably at 58 to 62° C. for 1 to 4 hours, preferably for 1 to 3 hours. The paraffin blocks of organs or tissues prepared by the PFA-AMeX method are stored at low temperature before use. At the time of use, the paraffin blocks thus prepared are sliced into thin sections using a microtome or the like. Then, the thin sections are deparaffinized and rehydrated. Deparaffinization and rehydration can be performed by known methods. For example, deparaffinization can be performed using xylene and toluene, while rehydration can be carried out using alcohol and acetone. The resulting thin sections are stained, for example, by histochemistry, immunohistochemistry, or enzyme histochemistry for detection, identification, or quantitation. When the prepared samples are stained by histochemistry (special staining), it is possible to use any staining method commonly available for paraffin-embedded sections (for example, PAS staining, giemsa staining, and toluidine blue staining). For staining by enzyme histochemistry, the sections may be stained by any staining method available for sections (for example, various staining such as with ALP, ACP, TRAP, or esterase). In addition, histopathological tissues can be stained by the following: hematoxylin-eosin staining for general staining; van Gieson staining, azan staining, and Masson Trichrome staining for collagen fiber staining; Weigert staining andElasticavan Gieson staining for elastic fiber staining; Watanabe's silver impregnation staining and PAM staining (periodic acid methenamine silver stain) for reticular fibers/basal membrane staining, etc. Staining with immunohistochemistry and enzyme histochemistry can be performed by direct methods using primary antibodies labeled with an enzyme or labeling substance, or indirect methods using non-labeled primary antibodies and labeled secondary antibodies. However, such methods are not limited thereto. Antibodies can be labeled by conventional methods. Labeling substances include, for example, radioisotopes, enzymes, fluorescent substances, and biotin/avidin. The labeling substances may be those commercially available. Radioisotopes include, for example,32P,33P,131I,125I,3H,14C, and35S. Enzymes include, for example, alkaline phosphatase, horse radish peroxidase, β-galactosidase, and β-glucosidase. Fluorescent substances include, for example, fluorescein isothiocyanate (FITC) and rhodamine. These may be commercially available. Labeling can be carried out by known methods. Thin sections are stained, for example, by histochemistry, immunohistochemistry, or enzyme histochemistry for detection, identification, or quantitation. Alternatively, detection, identification, or quantitation can be carried out by quantifying DNA or RNA in cells in organ/tissue samples. Assessment of the expression is not particularly limited, and conventional expression assessment methods can be used. RNAs include microRNAs, siRNAs, tRNAs, snRNAs, mRNAs, and non-coding RNAs. For example, Lgr5 mRNA is extracted according to conventional methods. Using the mRNA as a template, the transcriptional level of each gene can be determined by the Northern hybridization or RT-PCR method. DNA array techniques can also be used to determine the expression level of Lgr5. Desired tissues, cells, or such can be collected from samples by the microdissection method, in particular, laser microdissection (LMD) method. The LMD method can collect a group of target cells from living tissues, and thus accurately determine which cells express a specific gene among various cells that constitute a tissue, and at what level the cells express the gene. Devices used for microdissection include, for example, the AS-LMD system (Leica Microsystems). Furthermore, the present invention provides methods for diagnosing cancer, detecting cancer stem cells, or selecting cancer patients, which comprise using at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 9 to detect the presence of at least one of the proteins in a sample isolated from a cancer patient. In order to detect the presence of cancer stem cells, it is possible to use, instead of the anti-Lgr5 antibody described above, at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 9. In a non-limiting embodiment, the present invention provides methods for diagnosing cancer, detecting cancer stem cells, or selecting cancer patients, which comprise using at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 7 to detect the presence of at least one of the proteins in a sample isolated from a cancer patient. In order to detect the presence of Lgr5-positive cancer stem cells, it is possible to use, instead of the anti-Lgr5 antibody described above, at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 7. The presence of Lgr5-positive cancer stem cells can be detected by detecting the presence of the protein. However, the present invention does not exclude detection of Lgr5 in addition to the protein. In a non-limiting embodiment, the present invention provides methods for diagnosing cancer, detecting cancer stem cells, or selecting cancer patients, which comprise using at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 9 to detect the presence of at least one of the proteins in a sample isolated from a cancer patient. In order to detect the presence of Lgr5-negative cancer stem cells, it is possible to use, instead of the anti-Lgr5 antibody described above, at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 9. The presence of Lgr5-negative cancer stem cells can be detected by detecting the presence of the protein. However, the present invention does not exclude detection of Lgr5 in addition to the protein. Furthermore, the present invention provides methods for assessing the effectiveness of a pharmaceutical composition comprising at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 9, which comprise detecting one or more of the proteins of SEQ ID NOs: 1 to 3 and 5 to 9 and/or polynucleotides encoding the proteins in a sample isolated from a subject administered with the pharmaceutical composition. In these methods, detection may be carried out using at least one antibody that binds to a protein of SEQ ID NOs: 1 to 3 and 5 to 9, or a portion of a polynucleotide encoding the protein of any one of SEQ ID NOs: 1 to 3 and 5 to 9 and/or a complementary strand thereof. In another non-limiting embodiment, the present invention provides methods for assessing the effectiveness of a cancer treatment in a test subject, which comprise comparing the expression of at least one of the proteins of SEQ ID NOs: 1 to 3 and 5 to 9 and/or polynucleotides encoding the proteins in a first sample obtained from a test subject before providing the subject with at least part of the treatment, to the expression of at least one of the proteins of SEQ ID NOs: 1 to 3 and 5 to 9 and/or polynucleotides encoding the proteins in a second sample obtained from the subject after providing the part of the treatment, wherein a significantly lower level of the protein and/or polynucleotide in the second sample than in the first sample is an indicator showing that the treatment is effective for inhibiting cancer in the test subject. In a non-limiting embodiment, the present invention provides methods of monitoring in a test subject the effectiveness of a treatment with an antibody provided by the present invention, which comprise the steps of:(i) collecting a pre-administration sample from the subject before administration of the antibody;(ii) determining the expression level of at least one marker protein selected from the proteins of SEQ ID NOs: 1 to 3 and 5 to 9, or an mRNA or genomic DNA thereof in the pre-administration sample;(iii) collecting one or more post-administration sample(s) from the subject;(iv) determining the expression or activity level of at least one marker protein selected from the proteins of SEQ ID NOs: 1 to 3 and 5 to 9, or an mRNA or genomic DNA thereof in the post-administration sample(s);(v) comparing the expression or activity level of the marker protein, mRNA, or genomic DNA in the pre-administration sample to that of the marker protein, mRNA, or genomic DNA in the post-administration sample(s); and(vi) modifying the antibody administration to the test subject according to the comparison. For example, an increased dosage of an antibody of the present invention can be used to reduce the expression or activity of a marker towards a level higher than the detected level (the expression or activity level of the marker protein, mRNA, or genomic DNA in the sample before administration), i.e., to increase the effectiveness of the antibody. In a different non-limiting embodiment, the present invention provides methods of monitoring in a test subject the effectiveness of a treatment with an antibody provided by the present invention, which comprise the steps of:(i) detecting an Lgr5-positive cancer stem cell in a pre-administration sample collected from the subject before administration of the antibody;(ii) determining the expression level of at least one marker protein selected from the proteins of SEQ ID NOs: 1 to 3 and 5 to 7, or mRNA or genomic DNA thereof in the pre-administration sample;(iii) collecting one or more post-administration sample(s) from the subject;(iv) determining the expression or activity level of at least one marker protein selected from the proteins of SEQ ID NOs: 1 to 3 and 5 to 7, or mRNA or genomic DNA thereof in the post-administration sample(s);(v) comparing the expression or activity level of the marker protein, mRNA, or genomic DNA in the pre-administration sample to that of the marker protein, mRNA, or genomic DNA in the post-administration sample(s); and(vi) modifying the antibody administration to the test subject according to the comparison. For example, an increased dosage of an antibody of the present invention can be used to reduce the expression or activity of a marker towards a level higher than the detected level (the expression or activity level of the marker protein, mRNA, or genomic DNA in the sample before administration), i.e., to increase the effectiveness of the antibody. In another non-limiting embodiment, the present invention provides methods of monitoring in a test subject the effectiveness of a treatment with an antibody provided by the present invention, which comprise the steps of:(i) detecting an Lgr5-negative cancer stem cell in a pre-administration sample collected from the subject before administration of the antibody;(ii) determining the expression level of at least one marker protein selected from the proteins of SEQ ID NOs: 1 to 3 and 5 to 9, or mRNA or genomic DNA thereof in the pre-administration sample;(iii) collecting one or more post-administration sample(s) from the subject;(iv) determining the expression or activity level of at least one marker protein selected from the proteins of SEQ ID NOs: 1 to 3 and 5 to 9, or mRNA or genomic DNA thereof in the post-administration sample(s);(v) comparing the expression or activity level of the marker protein, mRNA, or genomic DNA in the pre-administration sample to that of the marker protein, mRNA, or genomic DNA in the post-administration sample(s); and(vi) modifying the antibody administration to the test subject according to the comparison. For example, an increased dosage of an antibody of the present invention can be used to reduce the expression or activity of a marker towards a level higher than the detected level (the expression or activity level of the marker protein, mRNA, or genomic DNA in the sample before administration), i.e., to increase the effectiveness of the antibody. Cancer Stem Cell Inhibitors “Cancer stem cell inhibitor” refers to, for example, an agent having the effect of suppressing the proliferation of cancer stem cells, suppressing the metastasis or recurrence of cancer stem cells, killing cancer stem cells, etc. It may have the effect of suppressing the proliferation of cancer cells, suppressing the metastasis or recurrence of cancer cells, killing cancer cells, etc. When used in connection with a biological activity, whose non-limiting examples include the proliferation or metastasis of cancer stem cells, the terms “suppress” and “suppressing”, and synonymous expressions refer to the down-regulation of the biological activity. This can reduce or eliminate a target function such as protein production and phosphorylation of a molecule, etc. In a specific embodiment, the suppression means a decrease in a target activity by about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95%. When used in connection with a disorder or disease, the terms refer to the prevention of development of a symptom, relief of a symptom, or successful alleviation of a disease, condition, or disorder. “Metastasis” refers to a process where cancer spreads or moves from the primary site to another site in the body, resulting in the development of similar cancer lesions at the new site. “Metastatic cell” or “metastasizing cell” refers to a cell that loses the adhesive contact with adjacent cells, and leaves the primary site of the disease and invades a nearby body structure via blood or lymphatic circulation. “Recurrence” means that the same malignant tumor reappears in a remaining organ after partial resection of an organ for removing a malignant tumor from a cancer patient, or after postoperative chemotherapy following the resection. Proteins Proteins for use in the present invention can be easily prepared by any method known to those skilled in the art as follows. An expression vector containing a gene comprising a DNA encoding a protein is constructed. The protein is produced and accumulated by culturing transformants transformed with the expression vector. The transformants are harvested to prepare the protein. Such an expression vector can be constructed according to methods known in the art, for example, by the following:(1) excising a DNA fragment that comprises a gene comprising a DNA encoding a protein; and(2) ligating the DNA fragment downstream of a promoter in an appropriate expression vector. Such vectors used includeE. coli-derived plasmids (for example, pBR322, pBR325, pUC18, and pUC118),Bacillus subtilis-derived plasmids (for example, pUB110, pTP5, and pC194), yeast-derived plasmids (for example, pSH19 and pSH15), bacteriophages such as λ phage, and animal viruses such as retroviruses, vaccinia viruses, and Baculoviruses. Promoters for use in the present invention may be any promoters as long as they are appropriate and compatible with a host to be used for gene expression. For example, when the host isE. coli, preferred promoters include the trp promoter, lac promoter, recA promoter, kPL promoter, and lpp promoter. When the host isBacillus subtilis, preferred promoters include the SPO1 promoter, SPO2 promoter, and penP promoter. When the host is yeast, preferred promoters include the PHO5 promoter, PGK promoter, GAP promoter, and ADH promoter. When the host is an animal cell, promoters include the SRa promoter, SV40 promoter, LTR promoter, CMV promoter, and HSV-TK promoter. In addition to those described above, if desired, enhancers, splicing signals, poly-A addition signals, selection markers, SV40 replication origins, or such known in the art can be added to expression vectors. As necessary, a protein for use in the present invention can be expressed as a fusion protein with another protein (for example, glutathione-S-transferase or Protein A). Such a fusion protein can be cleaved into individual proteins by using an appropriate protease. Host cells include, for example, bacteria of the genusEscherichia, bacteria of the genusBacillus, yeasts, insect cells, insects, and animal cells. Specific examples of bacteria of the genusEscherichiaincludeEscherichia coliK12 DH1 (Proc. Natl. Acad. Sci, USA, 60, 160 (1968)), JM103 (Nucleic Acids Research, 9, 309 (1981)), JA221 (Journal of Molecular Biology, 120, 517 (1978)), and HB101 (Journal of Molecular Biology, 41, 459 (1969)). Bacteria of the genusBacillusinclude, for example,Bacillus subtilisMI114 (Gene, 24, 255 (1983)) and 207-21 (Journal of Biochemistry, 95, 87 (1984)). Yeasts include, for example,Saccharomyces cerevisiaeAH22, AH22R-, NA87-11A, DKD-5D, and 20B-12; Schizosaccaromyces pombeNCYC1913 and NCYC2036; andPichia pastoris. Animal cells include, for example, monkey COS-7 cells, Vero cells, Chinese hamster CHO cells (hereinafter abbreviated as CHO cells), dhfr gene-deficient CHO cells, mouse L cells, mouse AtT-20 cells, mouse myeloma cells, rat GH3 cells, and human FL cells. These host cells can be transformed according to methods known in the art. See, for example, the following references. Proc. Natl. Acad. Sci. USA, Vol. 69, 2110 (1972); Gene, Vol. 17, 107 (1982); Molecular & General Genetics, Vol. 168, 111 (1979); Methods in Enzymology, Vol. 194, 182-187 (1991); Proc. Natl. Acad. Sci. USA), Vol. 75, 1929 (1978); and Virology, Vol. 52, 456 (1973). Transformants thus prepared can be cultured according to methods known in the art. For example, when hosts were bacteria of the genusEscherichia, in general, they are cultured at about 15 to 43° C. for about 3 to 24 hours. Aeration or stirring is performed as necessary. When hosts are bacteria of the genusBacillus, in general, they are cultured at about 30 to 40° C. for about 6 to 24 hours. Aeration or stirring is performed as necessary When hosts are yeasts, in general, transformants are cultured at about 20° C. to 35° C. for about 24 to 72 hours in a medium adjusted to about pH 5 to 8. Aeration or stirring is performed as necessary. When hosts are animal cells, in general, transformants are cultured at about 30° C. to 40° C. for about 15 to 60 hours in a medium adjusted to about pH 6 to 8. Aeration or stirring is performed as necessary. To isolate and purify a protein for use in the present invention from the above culture, for example, cells or bacteria are harvested after culture by a known method, and this is suspended in an appropriate buffer. After disrupting the cells or bacteria by sonication, lysozyme, and/or freeze-thawing, a crude protein extract is prepared by centrifugation or filtration. The buffer may contain protein denaturants such as urea and guanidine hydrochloride, and detergents such as Triton X-100™. When the protein is secreted to the culture medium, the supernatant is separated from the cells or bacteria after culture by a known method, and the supernatant is collected. A protein contained in the resulting culture supernatant or extract can be purified by appropriately combining known isolation/purification methods. According to known or equivalent methods, a protein prepared as described above can be arbitrarily modified or a polypeptide can be partially removed from the protein by treating the protein produced by recombinants with an appropriate protein modification enzyme such as trypsin and chymotrypsin before or after purification. The presence of a protein for use in the present invention can be assessed by various binding assays, enzyme immunoassays using specific antibodies, etc. Antibodies Antibodies for use in the present invention are not particularly limited as long as they bind to proteins for use in the present invention. The antibodies may be obtained as polyclonal or monoclonal antibodies using known methods. Particularly preferred antibodies for use in the present invention include monoclonal antibodies derived from mammals. Monoclonal antibodies derived from mammals include those produced by hybridomas and those produced by hosts transformed with expression vectors carrying antibody genes using gene engineering technologies. It is preferable that antibodies for use in the present invention specifically bind to proteins for use in the present invention. Basically, hybridomas producing monoclonal antibodies can be prepared using known techniques by the following procedure. Specifically, immunization is carried out using as a sensitizing antigen a protein for use in the present invention according to conventional immunization methods. The resulting immune cells are fused with known parental cells by conventional cell fusion methods. Monoclonal antibody-producing cells are screened using conventional screening methods. More specifically, monoclonal antibodies can be prepared by the following procedure. A gene sequence encoding the protein is inserted into a known expression vector system, and this is transformed into appropriate host cells. Then, the protein is purified from the host cells or culture supernatant by known methods. Next, the protein is used as a sensitizing antigen. Alternatively, a partial peptide of the protein is used as a sensitizing antigen. In this case, the partial peptide can be prepared by chemical synthesis based on the amino acid sequence of the protein according to common methods known to those skilled in the art. Such a partial polypeptide of the protein has, for example, at least 10 or more amino acids, preferably 50 or more amino acids, more preferably 70 or more amino acids, still more preferably 100 or more amino acids, and yet more preferably 200 or more amino acids of the amino acid sequence constituting the protein, and has, for example, a biological activity substantially equivalent to the function of the protein. The C terminus of the partial peptide is generally a carboxyl group (—COH) or carboxylate (—COO—); however, the C terminus may also be amide (—CONH2) or ester (—COOR). In addition, the partial peptides include those in which the amino group of the N-terminal methionine residue is protected with a protecting group, those in which a glutamyl residue resulting from in vivo N-terminal cleavage is pyroglutamine-oxidized, those in which a substituent group in the side chain of an amino acid in the molecule is protected with an appropriate protecting group, and conjugated peptides such as so-called glycopeptides linked with sugar chains. Mammals that are immunized with a sensitizing antigen are not particularly limited, though it is preferable to take into consideration compatibility with the parent cell used for cell fusion. Thus, rodents such as mice, rats, or hamsters are generally selected. Immunization of animals with a sensitizing antigen is performed according to known methods. For example, standard methods of delivering sensitizing antigen to mammals involve intraperitoneal or subcutaneous injection. More specifically, a sensitizing antigen is diluted to be an appropriate volume with PBS (phosphate-buffered saline), physiological saline, or the like. If desired, this may be mixed with an appropriate amount of a typical adjuvant, for example, Freund's complete adjuvant, made into an emulsion, and then administered to mammals several times every 4 to 21 days. An appropriate carrier may also be used for immunization with sensitizing antigens. After the mammals are immunized as described above, an increase in the level of desired antibody in the serum is confirmed, immunocytes are collected from the mammals for cell fusion. Immunocytes that are preferably subjected to cell fusion are splenocytes in particular. Regarding the other parent cell to be fused with the above-mentioned immunocytes, mammalian myeloma cells are used. For myeloma cells, it is preferable to use various known cell lines, for example, P3 (P3×63Ag8.653) (J. Immnol. (1979) 123, 1548-1550), P3×63Ag8U.1 (Current Topics in Microbiology and Immunology (1978) 81, 1-7), NS-1 (Kohler, G. and Milstein, C. Eur. J. Immunol. (1976) 6, 511-519), MPC-11 (Margulies, D. H. et al., Cell (1976) 8, 405-415), SP2/0 (Shulman, M. et al., Nature (1978) 276, 269-270), FO (de St. Groth, S. F. et al., J. Immunol. Methods (1980) 35, 1-21), S194 (Trowbridge, I. S. J. Exp. Med. (1978) 148, 313-323), and R210 (Galfre, G. et al., Nature (1979) 277, 131-133). In general, the above-described immunocytes and myeloma cells can be fused according to known methods, examples of which are described by Kohler and Milstein et al. (Kohler, G. and Milstein, C., Methods Enzymol. (1981) 73, 3-46). More specifically, the above-described cell fusion is carried out, for example, in a typical nutrient culture medium in the presence of a cell fusion promoting agent. For example, polyethylene glycol (PEG), Sendai virus (HVJ), or such can be used as the fusion promoting agent. If desired, adjuvants such as dimethylsulfoxide can additionally be used to increase fusion efficiency. It is possible to arbitrarily determine the proportion of immunocytes and myeloma cells used. The preferred ratio of myeloma cells to immunocytes is, for example, from 1:1 to 1:10. The culture medium used for the above-described cell fusion may be, for example, RPMI1640 medium, MEM medium, which are suitable for proliferation of the above-described myeloma cell lines, or other kinds of culture medium commonly used for culturing such cells. Furthermore, serum supplements such as fetal calf serum (FCS) may be used in combination. The cell fusion is carried out by thoroughly mixing prescribed amounts of the above-described immunocytes and myeloma cells in the aforementioned culture medium, adding to the medium a PEG solution preheated to about 37° C. generally at a concentration of 30% to 60% (w/v), wherein the PEG has an average molecular weight of about 1,000 to 6,000, for example, and mixing them to form the desired fusion cells (hybridomas). An appropriate culture medium is then successively added. Cell fusing agents and such that are undesirable for the proliferation of hybridomas are removed by repeatedly removing the supernatant by centrifugation. The hybridomas obtained in this manner are selected by culturing them in a common selection culture medium, for example, the HAT medium (a culture medium containing hypoxanthine, aminopterin, and thymidine). Culture in the HAT medium described above is continued for a sufficient time, usually from a few days to a few weeks, to allow death of all cells but the target hybridomas (the non-fused cells). The usual limiting dilution method is then performed to screen and clone hybridomas producing antibodies used the present invention. In addition to methods obtaining the above-described hybridomas by immunizing non-human animals with an antigen, desired human antibodies having an activity of binding to the protein can also be obtained by in vitro sensitizing human lymphocytes with the protein and fusing the sensitized lymphocytes with human-derived myeloma cells having permanent cell division ability (see Japanese Patent Application Kokoku Publication No. (JP-B) HO1-59878 (examined, approved Japanese patent application published for opposition)). Furthermore, human antibodies against a protein may be obtained from immortalized antibody-producing cells that are prepared by administering the protein as an antigen to a transgenic animal having a full repertoire of human antibody genes (see, International Patent Applications WO 94/25585, WO 93/12227, WO 92/03918, and WO 94/02602). There are known techniques for obtaining human antibodies by panning using a human antibody library. For example, the V regions of human antibodies can be expressed as single-chain antibodies (scFvs) on the surface of phages using a phage display methods, from which phages presenting scFv that binds to an antigen can be selected. The DNA sequences encoding the V regions of human antibodies that bind to the antigen can be determined by analyzing the genes of selected phages. After identifying the DNA sequences of scFvs that bind to the antigen, the V region sequences are fused in frame with the C region sequences of a desired human antibody. Then, the resulting DNA is inserted into an appropriate expression vector to construct an expression vector. The expression vector is introduced into suitable cells for expression, such as those described above. The human antibody can be obtained by expressing the gene encoding the human antibody. These methods are already known (see WO 1992/001047, WO 1992/020791, WO 1993/006213, WO 1993/011236, WO 1993/019172, WO 1995/001438, and WO 1995/015388). The hybridomas prepared in this manner that produce monoclonal antibodies can be passaged in a common culture medium and stored for a long time in liquid nitrogen. Monoclonal antibodies may be obtained from the hybridomas using common techniques; for example, the hybridomas are cultured according to standard methods and the antibodies may be obtained from the culture supernatants. Alternatively, the hybridomas are administered to a compatible mammal for proliferation and then the antibodies may be obtained from the ascites fluid. The former method is suitable for obtaining highly pure antibodies, while the latter method is suitable for mass production of antibodies. Monoclonal antibodies used in the present invention may be recombinant antibodies produced by genetic engineering techniques. They can be produced, for example, by cloning an antibody gene from a hybridoma, incorporating the antibody gene into an appropriate vector, and introducing the resulting vector into a host (see, for example, Vandamme, A. M. et al., Eur. J. Biochem., (1990) 192, p. 767-775, 1990). Specifically, mRNAs encoding antibody variable (V) regions are isolated from hybridomas producing the antibodies. mRNAs can be isolated by preparing total RNAs using known methods, for example, guanidine-ultracentrifugation method (Chirgwin, J. M. et al., Biochemistry (1979) 18, 5294-5299), AGPC method (Chomczynski, P. et al., Anal. Biochem. (1987) 162, 156-159), or such. mRNAs of interest are prepared using the mRNA Purification Kit (Pharmacia) or such. The mRNAs can be prepared directly by using the QuickPrep mRNA Purification Kit (Pharmacia). The obtained mRNAs are used to synthesize cDNAs of the antibody V regions using reverse transcriptase. cDNAs are synthesized using the AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Co.) or such. Alternatively, cDNA may be synthesized and amplified following the 5′-RACE method (Frohman, M. A. et al., Proc. Natl. Acad. Sci. USA (1988) 85, 8998-9002; Belyavsky, A. et al., Nucleic Acids Res. (1989) 17, 2919-2932) using the 5′-Ampli FINDER RACE Kit (Clontech) and PCR, and such. DNA fragments of interest are purified from the resulting PCR products, and ligated to vector DNAs. From this, a recombinant vector is produced. The recombinant vector is then introduced intoE. colior such, and the desired recombinant vector is prepared from a selected colony. The nucleotide sequences of DNAs of interest are then determined by known methods, for example, the dideoxynucleotide chain termination method. A DNA encoding the antibody V region of interest is obtained, and then incorporated into an expression vector carrying a DNA that encodes a desired antibody constant region (C region). To produce an antibody used in the present invention, the antibody gene is incorporated into an expression vector so that the gene will be expressed under the control of an expression regulatory region, for example, an enhancer and a promoter. Then, host cells are transformed with the resulting expression vector to express the antibody. When expressing antibody genes, a DNA encoding an antibody heavy chain (H chain) or light chain (L chain) can be each separately incorporated into an expression vector to simultaneously transform the host cell, or alternatively DNAs encoding H and L chains can be incorporated into a single expression vector to transform the host cells (see, WO 94/11523). Besides the above-described host cells, transgenic animals can also be used to produce recombinant antibodies. For example, an antibody gene is prepared as a fusion gene by inserting the antibody gene into a gene encoding a protein that is specifically produced in milk, such as goat casein. DNA fragments containing the fusion gene to which the antibody gene has been inserted is injected into goat embryos, which are then introduced into female goats. The desired antibody is then obtained from the milk produced by the transgenic goats, which are born from the goats that received the embryos, or from their offspring. Hormones may be suitably given to the transgenic goat to increase the production of milk containing the antibody of interests (Ebert, K. M. et al., Bio/Technology (1994) 12, 699-702). In the present invention, in addition to the antibodies described above, artificially modified genetically-recombinant antibodies such as chimeric, humanized, and human antibodies can be used to reduce heterologous antigenicity against humans and such. Such modified antibodies can be produced using known methods. Monoclonal antibodies of the present invention include not only those derived from animals described above but also artificially modified genetically-recombinant antibodies such as chimeric antibodies, humanized antibodies, and bispecific antibodies. A chimeric antibody can be obtained by linking a DNA encoding the antibody V region obtained as described above to a DNA encoding the human antibody C region, incorporating this into an expression vector, and then introducing it into a host for production. Useful chimeric antibodies can be obtained using this known method. Humanized antibodies are also referred to as “reshaped human antibodies”, which are antibodies obtained by grafting the complementarity determining regions (CDRs) of an antibody from a non-human mammal (e.g., mouse antibody) to the complementarity determining regions of a human antibody. General gene recombination procedures are also known (see, European Patent Application Publication No. EP 125023; International Patent Application Publication No. WO 96/02576). Specifically, a DNA sequence designed to link a mouse antibody CDR to the framework region (FR) of a human antibody is synthesized by PCR, using as primers several oligonucleotides prepared to contain overlapping portions in both CDR and FR terminal regions (see methods described in WO 98/13388). The human antibody framework region to be linked via CDR is selected such that complementarity determining region forms a favorable antigen-binding site. As necessary, amino acids of the framework region in the antibody variable region may be substituted so that the complementarity determining region of the reshaped human antibody forms a suitable antigen-binding site (Sato, K. et al., 1993, Cancer Res. 53, 851-856). Human antibody C-regions are used as the C-regions of chimeric antibodies or humanized antibodies. For example, CH1, CH2, CH3, and CH4 can be used for the H chain, while CK and Ck can be used for the L chain. The human antibody C-region may be modified in order to improve stability of the antibody or its production. A chimeric antibody is composed of the variable region of an antibody derived from a non-human mammal and the constant region derived from a human antibody. On the other hand, a humanized antibody is composed of the complementarity determining region of an antibody derived from a non-human mammal, and the framework region and C region derived from a human antibody. Since the antigenicity of humanized antibodies is low in the human body, and humanized antibodies are useful as an active ingredient in therapeutic agents of the present invention. Antibodies used in the present invention are not limited to whole antibody molecules, and as long as they bind to proteins used in the present invention, antibody fragments and modification products thereof as well as divalent and monovalent antibodies are also included. Antibody fragments include, for example, Fab, F(ab′)2, Fv, Fab/c having an Fab and the whole Fc, single chain Fv (scFv) in which Fv fragments from H and L chains are ligated via an appropriate linker, and Diabody. Specifically, antibody fragments are prepared by treating antibodies with an enzyme, for example, papain or pepsin. Alternatively, after genes encoding such antibody fragments are constructed and introduced into an expression vector, the antibody fragments are expressed in appropriate host cells using the vector (see, for example, Co, M. S. et al., J. Immunol. (1994) 152, 2968-2976; Better, M. & Horwitz, A. H. Methods in Enzymology (1989) 178, 476-496, Academic Press, Inc.; Plueckthun, A. & Skerra, A. Methods in Enzymology (1989) 178, 476-496, Academic Press, Inc.; Lamoyi, E., Methods in Enzymology (1989) 121, 652-663; Rousseaux, J. et al., Methods in Enzymology (1989) 121, 663-669; Bird, R. E. et al., TIBTECH (1991) 9, 132-137). scFv is obtained by ligating antibody H-chain V region with an antibody L-chain V region. In this scFv, the H-chain and L-chain V regions are ligated via a linker, preferably via a peptide linker (Huston, J. S. et al., Proc. Natl. Acad. Sci. U.S.A. (1988) 85, 5879-5883). The H-chain V region and L-chain V region of an scFv may be derived from any of the antibodies described herein. For example, any single-chain peptides consisting of 12 to 19 amino acid residues such as (GGGGS)n may be used as a peptide linker for ligating the V regions. A DNA encoding an scFv can be obtained by using, among DNAs encoding the antibody H chain or H chain V region and the antibody L chain or L chain V region mentioned above, all or DNA portion encoding amino acid sequence of interest as a template, amplifying by PCR using a primer pair that defines its two ends; and then carrying out a subsequent amplification using a combination of a DNA encoding the peptide linker portion, and primer pairs that define both ends of the linker DNA to be ligated to the H chain and L chain, respectively. Once DNAs encoding scFvs are constructed, expression vectors carrying the DNAs and hosts transformed with the expression vectors can be obtained according to conventional methods. Furthermore, scFvs can be obtained using these hosts according to conventional methods. Diabodies are dimers formed by linking two fragments (for example, scFv) in which a variable region is linked to another variable region via a linker or such, and typically have two VLs and two VHs (P. Holliger et al., Proc. Natl. Acad. Sci. USA, 90, 6444-6448 (1993); EP 404097; WO 93/11161; Johnson et al., Method in Enzymology, 203, 88-98, (1991); Holliger et al., Protein Engineering, 9, 299-305 (1996); Perisic et al., Structure, 2, 1217-1226 (1994); John et al., Protein Engineering, 12(7), 597-604 (1999); Holliger et al., Proc. Natl. Acad. Sci. USA., 90, 6444-6448 (1993); Atwell et al., Mol. Immunol. 33, 1301-1312 (1996); and such). These antibody fragments can be produced, in a similar manner as described above, by obtaining their genes and expressing them in hosts. Herein, “antibody” comprises such antibody fragments. As modified antibodies, antibodies of the present invention linked to various molecules such as polyethylene glycol (PEG) may be used. Moreover, antibodies can also be linked to radioisotopes, chemotherapeutic agents, cytotoxic substances such as bacteria-derived toxins, or such. Herein, “antibody” includes such modified antibodies. Modified antibodies can be obtained by chemically modifying the prepared antibodies. Such antibody modification methods are already established in the art. Furthermore, antibodies used in the present invention may be bispecific antibodies. Bispecific antibodies of the present invention may be those having antigen-binding sites that each recognizes different epitopes in the protein used in the present invention or those which recognize the protein used in the present invention and a different protein. Alternatively, bispecific antibodies of the present invention may be those in which one antigen-binding domain recognizes the protein used in the present invention and the other recognizes a chemotherapeutic agent or a cytotoxic substance such as a cell-derived toxin. In this case, proliferation of cancer stem cells can be suppressed by allowing a cytotoxic substance to act directly on cancer stem cells expressing a protein used in the present invention and specifically damaging the cancer stem cells. It is also possible to use bispecific antibodies in which one antigen-binding domain recognizes a molecule that constitutes the T cell receptor complex such as CD3, expressing on cytotoxic T cells, and the other recognizes an epitope in the protein of any one of SEQ ID NOs: 1 to 3 and 5 to 9 of the present invention. The bispecific antibodies may be prepared by linking pairs of H and L chains from two types of antibodies, or by fusing hybridomas that produce different monoclonal antibodies to yield a fusion cell producing bispecific antibodies. Furthermore, the bispecific antibodies can be prepared using genetic engineering techniques. Antibody genes constructed as described above can be expressed and obtained according to known methods. When mammalian cells are used, antibody genes can be expressed using a DNA in which a common useful promoter, an antibody gene to be expressed, and a poly A signal positioned downstream of the antibody gene on the 3′ side are operably linked. Promoter/enhancer includes, for example, human cytomegalovirus immediate early promoter/enhancer. Furthermore, other promoter/enhancers that can be used to express the antibody used in the present invention include viral promoter/enhancers of retroviruses, polyoma viruses, adenoviruses, simian virus 40 (SV40), and such; and mammalian cell-derived promoter/enhancers such as human elongation factor 1α (HEF1α). When SV40 promoter/enhancer and HEF1α promoter/enhancer is used, gene expression can be easily carried out by the method of Mulligan et al. (Nature (1979) 277, 108) and the method by Mizushima et al. (Nucleic Acids Res. (1990) 18, 5322), respectively. Replication origin derived from SV40, polyoma viruses, adenoviruses, bovine papilloma viruses (BPV), and such can be used. Furthermore, to increase the gene copy number in a host cell system, the expression vector may include, as a selection marker, the aminoglycoside transferase (APH) gene, thymidine kinase (TK) gene,E. colixanthine-guanine phosphoribosyltransferase (Ecogpt) gene, dihydrofolate reductase (dhfr) gene, and such. In the case ofE. coli, the antibody gene can be expressed by an operably linked common useful promoter, a signal sequence for antibody secretion, and the antibody gene to be expressed. Such promoters include, for example, the lacz promoter and araB promoter. When the lacz promoter or araB promoter is used, the gene can be expressed by the method of Ward et al. (Nature (1098) 341, 544-546; FASEB J. (1992) 6, 2422-2427) or the method of Better et al. (Science (1988) 240, 1041-1043), respectively. When an antibody is produced into the periplasm ofE. coli, the pel B signal sequence (Lei, S. P. et al., J. Bacteriol. (1987) 169, 4379) may be used as a signal sequence for antibody secretion. After antibodies produced into the periplasm is separated, the antibody structure is appropriately refolded and then used. Any expression system that uses, for example, eukaryotic cells or prokaryotic cells may be used to produce antibodies used in the present invention. Eukaryotic cells include, for example, animal cells such as established mammalian cell systems, insect cell systems, cells of filamentous fungi, and yeast cells. Prokaryotic cells include, for example, bacterial cells such asE. colicells. Antibodies used in the present invention are preferably expressed in mammalian cells, for example, CHO, COS, myeloma, BHK, Vero, and HeLa cells. Then, transformed host cells are cultured in vitro or in vivo to produce antibodies of interest. Host cells are cultured according to known methods. For example, DMEM, MEM, RPMI1640, or IMDM may be used as a culture medium, and this may also be used with serum supplements such as fetal calf serum (FCS). Antibodies expressed and produced as described above can be isolated from cells or host animals and purified to be homogeneous. Antibodies used in the present invention can be isolated/purified by using affinity columns. For example, Protein A columns include Hyper D, POROS, and Sepharose F. F. (Pharmacia). It is also possible to use other common protein isolation/purification methods. Such methods are not particularly limited. For example, antibodies may be isolated/purified by appropriately selecting/combining chromatography columns other than the above-described affinity columns, filters, ultrafiltration, salting-out, dialysis, and such (Antibodies A Laboratory Manual. Ed Harlow, David Lane, Cold Spring Harbor Laboratory, 1988). The antigen-binding activity (Antibodies A Laboratory Manual. Ed Harlow, David Lane, Cold Spring Harbor Laboratory, 1988) and ligand-receptor binding-inhibitory activity (Harada, A. et al., International Immunology (1993) 5, 681-690) of an antibody used in the present invention can be determined by using known methods. Enzyme-linked immunosorbent assays (ELISAs), enzyme immunoassays (EIAs), radioimmunoassays (RIAs), and fluorescent antibody methods can be used to determine the antigen-binding activity of the antibody of the present invention. For example, when an enzyme immunoassay is used, samples containing an antibody of the present invention such as a culture supernatant of cells producing the antibody or the purified antibody are added to plates coated with a protein used in the present invention. A secondary antibody labeled with an enzyme such as alkaline phosphatase is added, and the plates are incubated. After washing, an enzyme substrate such as p-nitrophenyl phosphate is added and the absorbance is measured to evaluate the antigen-binding activity. An antibody used in the present invention may appropriately be linked to a cytotoxic substance described above such as a proliferation inhibitor, toxic peptide, or radioactive chemical substance. Such modified antibodies (hereinafter referred to as antibody conjugates) can be obtained by chemically modifying the obtained antibodies. Specifically, a linker molecule links a growth inhibitor to an antibody via chemical bonding so that the antibody and growth inhibitor or cytotoxic substance can chemically conjugate with each other (for example, can bind covalently). Preferred binders (linkers) are cleavable linkers. It is more preferable that the linkers are cleaved under mild conditions (specifically, intracellular conditions that do not affect the activity of inhibitors). Examples of suitable cleavable linkers include disulfide linkers, acid-labile linkers, photo-labile linkers, peptidase-labile linkers, and esterase-labile linkers. Disulfide-containing linkers can be cleaved via disulfide exchange, which can occur under physiological conditions. Acid-labile linkers can be cleaved at acid pH. For example, certain intracellular compartments such as endosomes and lysosomes have an acidic pH (pH 4 to 5), and provide conditions suitable for cleaving acid-labile linkers. Photo-labile linkers are useful on the body surface and in many body cavities, which can be exposed to light. Furthermore, infrared light can penetrate tissues. Peptidase-labile linkers can be used to cleave certain peptides inside or outside cells (for example, see Trouet et al., Proc. Natl. Acad. Sci. USA (1982) 79, 626-629; Umemoto et al., Int. J. Cancer (1989) 43, 677-684). Such modified antibodies can be prepared not only by chemical modification as described above, but also in a molecular form such as a bispecific antibody designed to recognize a growth inhibitor, toxic peptide, radioactive chemical substance, or the like using genetic recombination techniques. “Antibody” of the present invention also comprises such antibodies. Examples of modified antibodies that are provided by the present invention also include those modified with a toxic peptide such as ricin, abrin, ribonuclease, onconase, DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin,Pseudomonasexotoxin,Pseudomonasendotoxin, L-asparaginase, or PEG L-Asparaginase. In another embodiment, antibodies may be modified by the combined use of one or more growth inhibitors and toxic peptides. As described above, the linkage between the antibody of the present invention that binds to at least one protein described in SEQ ID NOs: 1 to 3 and 5 to 9 and an above-described growth inhibitor, toxic peptide, or radioactive chemical substance may be a covalent or non-covalent bond. Methods for preparing modified antibodies linked to such chemotherapeutic agents are known. Furthermore, proteinaceous pharmaceutical agents and toxins can be linked to an antibody by using a genetic engineering procedure. Specifically, for example, a recombinant vector into which a DNA encoding a toxic peptide described above and a DNA encoding an antibody that binds to any of proteins of at least one of SEQ ID NOs: 1 to 3 and 5 to 9 of the present invention are fused in frame and incorporated into an expression vector is constructed. Transformed cells obtained by introducing the vector into appropriate host cells are cultured to express the incorporated DNA. The modified antibody linked to the toxic peptide is obtained as a fusion protein. When a fusion protein with an antibody is prepared, in general, a proteinaceous pharmaceutical agent or toxin is placed at the C terminus of the antibody. A peptide linker may be interposed between the antibody and a proteinaceous pharmaceutical agent or toxin. Antibodies used in the present invention may have a cytotoxic activity. Herein, the cytotoxic activity includes, for example, complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). Herein, CDC refers to a cytotoxic activity mediated by the complement system, while ADCC refers to an activity of damaging target cells, which is caused by binding of Fcγ receptor-carrying cells (immunocytes, etc.) via Fcγ receptor to the Fc portion of specific antibody upon binding of the antibody to cell-surface antigens on target cells. Whether an antibody used in the present invention has ADCC or CDC can be measured by known methods (see, for example, Current protocols in Immunology, Chapter 7. Immunologic studies in humans, Editor, John E, Coligan et al., John Wiley & Sons, Inc., (1993)). Specifically, cytotoxicity can be measured, for example, by the following method. Preparation of Effector Cells Spleen is removed from a CBA/N mouse or the like, and spleen cells are dispersed in RPMI1640 medium (GIBCO). After washing with the same medium containing 10% fetal bovine serum (FBS, HyClone), effector cells with a cell concentration adjusted to 5×106cells/ml were prepared. Preparation of Complement Solution Baby Rabbit Complement (CEDARLANE) is diluted 10-fold with a medium (GIBCO) containing 10% FBS to prepare a complement solution. Preparation of Target Cells Cells expressing a protein used in the present invention (cancer stem cells, etc.) are radiolabeled by incubating them with 0.2 mCi of51Cr-sodium chromate (Amersham Pharmacia Biotech) in DMEM medium containing 10% FBS for one hour at 37° C. After radiolabeled, the cells are washed three times with RPMI1640 medium containing 10% FBS, and the target cells with a cell concentration adjusted to 2×105cells/ml were prepared. ADCC Measurement 50 μl the target cells and 50 μl of the antibody used in the present invention are each added to a 96-well U-bottom plate (Becton Dickinson), and reacted for 15 minutes on ice. Thereafter, 100 μl of effector cells are added and incubated in a carbon dioxide incubator for four hours. The final antibody concentration is adjusted to 0 or 10 μg/ml. After incubation, 100 μl of the supernatant is collected and the radioactivity is measured with a gamma counter (COBRAIIAUTO-GMMA, MODEL D5005, Packard Instrument Company). The cytotoxic activity (%) can be calculated according to: (A−C)/(B−C)×100. A represents the radioactivity (cpm) of each sample, B represents the radioactivity (cpm) of a sample where 1% NP-40 (nacalai tesque) has been added, and C represents the radioactivity (cpm) of a sample containing the target cells alone. CDC Measurement 50 μl of the target cells and 50 of the antibody used in the present invention are each added to a 96-well flat-bottom plate (Becton Dickinson), and reacted for 15 minutes on ice. Thereafter, 100 μl of the complement solution is added, and incubated in a carbon dioxide incubator for four hours. The antibody final concentration is adjusted to 0 or 3 μg/ml. After incubation, 100 μl of the supernatant is collected to measure the radioactivity with a gamma counter. The cytotoxic activity can be calculated by the similar way as in the ADCC determination. Antibodies with modified sugar chains can appropriately be used in the antibodies provided by the present invention. It is known that cytotoxic activity of antibodies can be increased by modifying its sugar chains. Known antibodies with modified sugar chains include, for example: glycosylated antibodies (WO 1999/054342 and such); antibodies with defucosylated sugar chain (WO 2000/061739, WO 2002/031140, and such); and antibodies having a sugar chain with bisecting GlcNAc (bisecting N-acetylglucosamine) (WO 2002/079255). Antibodies of the present invention preferably include antibodies with modified sugar chains whose sugar chain composition has been altered to increase the ratio of defucosylated antibody or to increase the ratio of antibody attached with bisecting N-acetylglucosamine. Antibodies having a neutralizing activity can also be used appropriately in the present invention. In general, “neutralizing activity” refers to the activity of a foreign molecule such as a toxin or virus, or an internal molecule such as a hormone or cytokine to inhibit a ligand's biological activity on cells. Specifically, substance having a neutralizing activity refers to a substance that binds to a ligand or a receptor to which the ligand binds, thereby inhibiting the ligand-receptor binding. The receptor whose ligand binding is inhibited by the neutralizing activity cannot exert their receptor-mediated biological activity. When the antigen-binding molecule is an antibody, in general, such an antibody with a neutralizing activity is called a neutralizing antibody. The neutralizing activity of a test substance can be assessed by comparing biological activities in the presence of a ligand, in the condition of when the test substance is present or not present. EREG, which is a target of EP27 antibody described later on in the Examples, is exemplified below. EGF receptor, which is believed to be a main receptor for the EREG represented by SEQ ID NO: 3, dimerizes upon ligand binding and activates its own cytoplasmic tyrosine kinase domain. The activated tyrosine kinase causes a peptide having phosphotyrosine by autophosphorylation, which allows association of various signal transduction accessory molecules. The molecules are mainly phospholipase Cγ (PLCγ), She, Grb2, and such. Of these accessory molecules, the former two are further phosphorylated by the tyrosine kinase of the EGF receptor. The main signaling pathway from the EGF receptor is the one in which phosphorylation occurs in order of She, Grb2, Sos, Ras, and Raf/MAPK kinase/MAP kinase. It is believed that there is also an alternative pathway from PLCγ to PKC. Since such intracellular signal cascades vary depending on cell type, a target molecule can appropriately be selected for each target cell type of interest and is not limited to the factors described above. It is possible to use an appropriate in vivo signal activation assay kit available on the market (for example, protein kinase C activation measurement system (GE Healthcare Bioscience, etc.)). Alternatively, the in vivo signal activation can be detected by using as an indicator the transcriptional induction of a target gene downstream in the in vivo signal cascade. Changes in the transcriptional activity can be detected based on the principle of reporter assay. Specifically, a reporter gene such as GFP (Green Fluorescence Protein) or luciferase is placed downstream of the transcriptional factor or promoter region of the target gene. A change in the transcriptional activity can be determined as reporter activity by measuring the reporter activity. In addition, the EGF receptor typically functions to promote cell proliferation, and thus the activation of in vivo signal transduction can be assessed by measuring the proliferative activity of the target cell. In the present invention, the neutralizing activities of neutralizing antibodies of the present invention are assessed by measuring the cell proliferative activity. However, methods are not limited thereto, and methods described above can suitably be used to assess the activity depending on the type of selected target cells. Specifically, the neutralizing activity of an anti-EREG antibody can be assessed or determined by measuring the cell proliferative activity, for example, using the method described below. For example, the method where the incorporation of [3H]-labeled thymidine, which is added to a culture medium, into viable cells is measured as an indicator for the DNA replication ability is used. Simpler methods include the MTT method and dye exclusion tests in which the ability of cells to exclude dyes such as Trypan Blue outside is measured under a microscope. The MTT method utilizes the ability of viable cells to convert MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide), a tetrazolium salt, into a blue formazan product. More specifically, the MTT method is performed as follows: a test antibody is added to a culture medium containing test cells; after a certain period, an MTT solution is added to the culture medium; the mixture is allowed to stand for a certain period so that MMT is incorporated into the cells. As a result, a yellow compound MTT is converted into a blue compound by succinate dehydrogenase in cytoplasmic mitochondria. After the blue product is dissolved for color development, the absorbance is measured as an indicator for the viable cell count. In addition to MTT, other commercially available reagents such as MTS, XTT, WST-1, and WST-8 (nacalai tesque, etc.) can preferably be used. There are also known methods for assessing the cell proliferative activity using as an indicator an intracellular ATP content or impedance of cell culture. When assessing the activity, a control antibody that is of the same isotype as the anti-EREG antibody of interest but does not have the neutralizing activity is used in the same manner as for the anti-EREG antibody. The activity can be assessed whether the anti-EREG antibody exhibits the neutralizing activity greater than that of the control antibody. Cells whose proliferation is inhibited by the anti-EREG antibody are not particularly limited, as long as they express EREG protein. Examples of preferred EREG-expressing cells include cancer cells. Specifically, cells derived from colorectal cancer, lung adenocarcinoma, pancreatic cancer, stomach cancer, or kidney cancer are preferable EREG-expressing cells in the present invention. A cell proliferation-inhibitory effect against both primary and metastatic lesions of all these cancers can be achieved by the present invention. More preferred cancer cells include those of primary colorectal cancer, metastatic colorectal cancer, lung adenocarcinoma, pancreatic cancer, stomach cancer, and kidney cancer. Thus, anti-EREG antibodies can be used to treat/prevent diseases caused by cell proliferation, for example, colorectal cancer, lung adenocarcinoma, pancreatic cancer, stomach cancer, and kidney cancer. These cancers can be targets of treatment or prevention, regardless of primary or metastatic lesions. More preferably, anti-EREG antibodies are used to treat and/or prevent primary colorectal cancer, metastatic colorectal cancer, and pancreatic cancer. Furthermore, among these cancers, those which grow in an EREG-dependent manner are preferred as a target of treatment and/or prevention in the present invention. In the description and Tables herein, when nucleotides and amino acids are represented by abbreviations, these abbreviations are based on the abbreviations by IUPAC-IUB Commission on Biochemical Nomenclature, or the conventional abbreviations in the art. Regarding amino acids, when an optical isomer exists, it represents L form, unless otherwise specified. Cancer Stem Cell Inhibitors of the Present Invention The effective dosage of cancer stem cell inhibitors of the present invention is selected within the range of 0.001 to 1,000 mg/kg weight for each administration. Alternatively, the dosage may be selected within the range of 0.01 to 100,000 mg/body for each patient. However, the dosage of the inhibitors of the present invention is not limited to these doses. Meanwhile, with respect to the timing of administration, an inhibitor of the present invention may be administered before or after manifestation of clinical symptoms of diseases. The inhibitors of the present invention can be formulated according to conventional methods (Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Easton, US), and may contain both pharmaceutically acceptable carriers and additives. Such carriers and medical additives include, for example, water, pharmaceutically acceptable organic solvents, collagen, polyvinyl alcohol, polyvinylpyrrolidone, carboxyvinyl polymer, carboxymethylcellulose sodium, sodium polyacrylate, sodium alginate, water-soluble dextran, carboxymethyl starch sodium, pectin, methyl cellulose, ethyl cellulose, xanthan gum, gum arabic, casein, agar, polyethylene glycol, diglycerin, glycerin, propylene glycol, vaseline, paraffin, stearyl alcohol, stearic acid, human serum albumin (HSA), mannitol, sorbitol, lactose, and surfactants that are acceptable as a medical additive. In practice, additives are selected alone or in appropriate combination from those listed above depending on the dosage form of an inhibitor of the present invention; but are not limited thereto. For example, when used as a preparation for injection, it can be used in a form in which the inhibitor is dissolved in a medium such as physiological saline, buffer, or glucose solution and an adsorption inhibitor such as Tween80, Tween20, gelatin, or human serum albumin is added; alternatively, the inhibitor of the preset invention may be in a lyophilized form for dissolution and reconstitution before use. As excipients for lyophilization, for example, sugar alcohols and saccharides such as mannitol and glucose can be used. The inhibitors of the present invention are generally administered by a parenteral route, for example, via injection (subcutaneous, intravenous, intramuscular, intraperitoneal, etc.), transdermal, transmucosal, intranasal, or pulmonary administration; however, the inhibitor can be administered orally. Herein, “combined use” of a cancer stem cell inhibitor and an anti-cancer agent means that these agents may be administered at the same time or in succession; alternatively, one is administered at an interval after administration of the other. Herein, cancer stem cell inhibitors can be used as various embodiments such as, for example, prevention of cancer recurrence, suppression of cancer recurrence, prevention of cancer metastasis, suppression of cancer metastasis, and adjuvant therapy for preventing postoperative recurrence for application. When used in the above-described embodiments, any cancer stem cell inhibitor can be used as a cancer stem cell inhibitor of the present invention. However, non-limiting preferred examples include agents for inhibiting cancer stem cell proliferation or agents for disrupting cancer stem cell. As long as agents for inhibiting cancer stem cell proliferation that are provided by the present invention can suppress the proliferation of target cancer stem cells, mechanism of suppressing cancer stem cell proliferation is not relevant. As a non-limiting example, such agents for inhibiting cancer stem cell proliferation include those comprising as an active ingredient an antibody having neutralizing activity against cancer stem cell proliferation or growth or an antibody having cytotoxicity against cancer stem cells. Similarly, as long as agents for disrupting cancer stem cells that are provided by the present invention can destroy target cancer stem cells, mechanism of disrupting cancer stem cells is not relevant. As a non-limiting example, such agents for disrupting cancer stem cells include agents for inhibiting cancer stem cell proliferation comprising as an active ingredient an antibody having cytotoxicity or apoptotic activity against cancer stem cells. Those skilled in the art can determine whether a test agent for disrupting cancer stem cells has apoptotic activity by using known methods including terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling (TUNEL) assay, caspase activity (in particular, caspase-3) assay, fas ligand assay, and Annexin V assay as apoptotic activity assay methods. Non-limiting preferred examples include cancer stem cell differentiation enhancers. Non-limiting examples of differentiation enhancers include BMP4, i.e., the polypeptide of SEQ ID NO: 4, or polypeptide equivalents having one or several amino acid addition(s), deletion(s), and/or substitution(s) among amino acids of the polypeptide. Such polypeptide equivalents preferably have a CSC differentiation-inducing activity equivalent to that of the polypeptide of SEQ ID NO: 4. The equivalency of the differentiation-inducing activity can be defined, for example, by whether the CK20-inducing activity for CSCs is 10%, preferably 20%, more preferably 30%, even more preferably 40%, and still more preferably 50% of that of the polypeptide of SEQ ID NO: 4. In another non-limiting embodiment, the equivalency of the differentiation-inducing activity can be defined, for example, by whether the CK20-inducing activity for CSCs is 60%, preferably 70%, more preferably 80%, even preferably 90%, and still preferably 95% of that of the polypeptide of SEQ ID NO: 4. Anti-cancer agents that are used in combination with a cancer stem cell inhibitor of the present invention include alkylating agents, metabolic antagonists, natural products, platinum complexes, and other pharmaceutical agents. Alkylating agents include nitrogen mustards, ethylenimines, methylmelamines, alkyl sulfonates, nitrosoureas, and triazens. Nitrogen mustards include, for example, mechlorethamine, cyclophosphamide, ifosfamide, melphalan, and chlorambucil. Ethylenimines and methylmelamines include, for example, hexamethylmelamine and thiotepa. Alkyl Sulfonates include busulfan. Nitrosoureas include, for example, carmustine (BCNU), lomustine (CCNU), semustine (methyl-CCNU), and streptozocin. Triazens include dacarbazine (DTIC). Metabolic antagonists include folic acid analogs, pyrimidine analogs, and purine analogs. Folic acid analogs include methotrexate. Pyrimidine analogs include, for example, fluorouracil (5-FU), doxifluridine (5′-DFUR; trade name: FURTULON), capecitabine (trade name: Xeloda), floxuridine (FudR), and cytarabine. Purine analogs include, for example, mercaptopurine (6-MP), thioguanine (TG), and pentostatin. Natural products includevincaalkaloids, epipodophyllotoxins, and antibiotics.Vincaalkaloids include, for example, vinblastine (VLB) and vincristine (VCR). Epipodophyllotoxins include, for example, etoposide and teniposide. Antibiotics include, for example, dactinomycin (actinomycin D), daunorubicin, doxorubicin, bleomycin, plicamycin, and mitomycin. Platinum complex refers to platinum coordination complex, and includes, for example, cisplatin (CDDP) and carboplatin. Other pharmaceutical agents include: topoisomerase inhibitors such as irinotecan and camptothecin; taxols, for example, paclitaxel, docetaxel; anthracenediones, for example, mitoxantrone; urea-substituted derivatives, for example, hydroxyurea; methyl hydrazines, for example, procarbazine hydrochloride (trade name: Natulan), vitamin A metabolites, for example, tretinoin (trade name: VESANOID), as well as include rituximab, alemtuzumab, trastuzumab, bevacizumab, cetuximab, panitumumab, trastuzumab, and gemutuzumab. All prior-art documents cited herein are incorporated herein by reference. EXAMPLES Herein below, the present invention will be specifically described with reference to examples, however, it is not to be construed as being limited thereto. Establishment of Human Colorectal Cancer Cell Lines with Immunodeficient NOG Mice Colorectal cancer specimens were obtained from patients with their consent under approval of the ethical committees of PharmaLogicals Research (Singapore) and Parkway Laboratory Services (Singapore). Tumor blocks were cut into small pieces with a razor blade, and grafted in the lateral region of NOG mice. Human colorectal cancer xenografts were maintained by passaging them in NOG mice provided by the Central Institute for Experimental Animals (Japan). Mice used in this experiment were treated in accordance with the animal experiment guidelines of PharmaLogicals Research. For histopathological examination, small blocks of xenograft tumors and surgical human tissue samples were fixed with 4% paraformaldehyde at 4° C. for 16 to 24 hours, and embedded in paraffin by the AMeX method (Sato Y, et al., (1986) Am J Pathol, 125: 431-435; Sato Y, et al., (1992) Am J Pathol, 140: 775-779; Suzuki M, et al. (2002) J Toxicol Sci, 27: 165-172). The thin sections were stained with eosin and hematoxylin and assessed by microscopic observation. Isolation and In Vitro Culture of Large Intestine CSCs Tissues of xenograft were cut with a razor blade in order to prepare single cancer cell suspensions. After the suspensions were incubated at 37° C. for 3 hours in DPBS containing collagenase/dispase (Roche) and DNaseI (Roche), the suspensions were filtered through 40-μm Cell Strainers (BD Biosciences). The cells were suspended in the lysis buffer (BD Biosciences) to remove erythrocytes. The prepared xenograft-derived cells (such cells are referred to as primary cells) were cultured under 5% CO2atmosphere at 37° C. in DMEM/F12 (Invitrogen) containing N-2 Supplement (Invitrogen), 20 ng/ml human EGF (Invitrogen), 10 ng/ml human basic fibroblast growth factor (Sigma), 4 μg/ml heparin (Sigma), 4 mg/ml BSA (Invitrogen), 20 μg/ml human insulin zinc solution (Invitrogen), and 2.9 mg/ml glucose (Sigma) (Todaro M, et al. (2007) Cell Stem Cell 1: 389-402). Adherent and floating CSCs were cultured using conventional polystyrene-treated cell culture flasks (BD Biosciences) and Ultra-low attachment cell culture flasks (Corning), respectively. In Vivo Tumor Formation Analysis Cell suspensions were prepared by serial dilution. 100 μl of cancer cell suspensions in Hanks' balanced salt solution (Invitrogen) were subcutaneously inoculated into lateral region of mice using 50% matrigel (BD Bioscience). The tumor development was monitored over seven weeks. In order to inoculate single cell, cells were labeled using an FITC-labeled mouse anti-human CD326 (EpCAM) antibody (Miltenyi Biotec), and plated in a terasaki plate (Termo Fisher Scientific). Cell singularity was confirmed under a microscope. Single cells were inoculated into lateral region of mice using 50 μl of 50% matrigel. The tumor development was monitored over 10 weeks. Establishment of Cells Expressing Full-Length Human Lgr4, Lgr5, and Lgr6 Full-length human Lgr4, Lgr5, and Lgr6 cDNAs were cloned by PCR based on the sequences of NM_018490 (Lgr4), NM_001017403 (Lgr6), and NM_003667 (Lgr5). The cloned genes were expressed with or without adding HA tag to their N termini. Cells of Chinese hamster ovary cell line CHO DG44 (Invitrogen) were transfected with expression plasmids using Gene Pulser (BioRad). Stable cell lines HA-Lgr4/DG, HA-Lgr5/DG, and HA-Lgr6/DG were selected using G418. Preparation of Soluble Lgr5-Fc Protein Soluble Lgr5 protein (amino acids 1 to 555) was expressed as a fusion protein with the Fc domain of mouse IgG2a in CHO DG44. Transfectants were screened by sandwich ELISA using a goat anti-mouse IgG2a (Bethyl laboratories) and HRP-rat anti-mouse IgG2a mAb (Serotec). A clone that produces sLgr5-Fc at the highest level was named 2D3. A culture supernatant of 2D3 was collected, and Lgr5-Fc protein was affinity-purified using a Protein A-Sepharose column (Pharmacia). Lgr5-Fc was used as an antigen in protein immunization and ELISA screening. Generation of Anti-Lgr5 Monoclonal Antibody by Immunization with Lgr5-Fc Protein Balb/c mice (Charles River Japan) were immunized subcutaneously with 50 g of Lgr5-Fc emulsified in Freund's complete adjuvant. After two weeks, the mice were injected with the same amount of Lgr5-Fc in Freund's incomplete adjuvant once a week over two weeks. Three days before cell fusion, 25 μg of Lgr5Fc was intravenously injected to the mice. Spleen lymphocytes derived from the immunized mice were fused with cells of mouse myeloma line P3-X63Ag8U1 (ATCC) using a conventional method (Kremer L and Marquez G (2004) Methods Mol Biol., 239: 243-260). Hybridoma culture supernatants were screened for antibodies reactive to sLgr5-Fc by ELISA to establish Lgr5-specific mouse mAb 2T15E-2 and 2U2E-2. Flow Cytometry Analysis Large intestine CSCs were incubated with labeled antibodies and analyzed using the EPICS ALTRA (Beckman Coulter) and FACSCalibur (Becton Dickinson)). Antibodies used were: PE-labeled mouse anti-human CD133 antibody (Miltenyi Biotec), PE-labeled mouse anti-human CD44 antibody (BD Pharmingen), FITC-labeled mouse anti-human CD326 (EpCAM) antibody (Miltenyi Biotec), PE-labeled mouse anti-human CD166 antibody (R&D Systems), PE-labeled mouse anti-human CD24 antibody (BD Pharmingen), PE-labeled mouse anti-human CD26 antibody (BD Pharmingen), and PE-labeled mouse anti-human CD29 antibody (BD Pharmingen). Large intestine CSCs were incubated with mouse anti-human Lgr5 antibody (2T15E-2) and then with PR-labeled rat anti-mouse IgG antibody (Invitrogen) to stain Lgr5. The aldehyde dehydrogenase activity was measured using the AldeFluor Kit (Stemcell Technologies). Mouse cells were discriminated from human large intestine CSCs by staining with anti-mouse MHC class I antibody (Abcam), and PE- or APC-labeled goat anti-human IgG2a antibody (BioLegend). Dead cells were also removed using the 7-AAD Viability Dye (Beckman Coulter). Western Blot Analysis Protein was extracted using RIPA buffer (Sigma) supplemented with the Complete Mini Protease Inhibitor Cocktail (Roche). Proteins were fractionated by the NuPAGE Gel (Invitrogen) and transferred onto PVDF membrane. After blocking with PBS containing 1% skimmed milk, the membrane was probed with rabbit anti-human β-catenin antibody (Sigma), rabbit anti-human phospho-c-JUN antibody (Sigma), rabbit anti-human TCF1 antibody (Cell Signaling), rabbit anti-human TCF3 antibody (Cell Signaling), rabbit anti-human TCF4 antibody (Cell Signaling), rabbit anti-human Lgr5 antibody (Abcam), mouse anti-human E-cadherin antibody (Abcam), rabbit anti-human Snail antibody (Abeam), and mouse anti-human GAPDH antibody (Santa Cruz). Reactive bands were detected using BCIP/NBT substrate (KPL). Quantitative Real-Time Polymerase Chain Reaction Total RNAs were isolated using the RNeasy Mini Kit including DNAase treatment (Qiagen). cDNAs were synthesized using the First-Strand cDNA Synthesis Kit (SABiosciences). Quantitative real-time PCR (QRT-PCR) analysis was performed with the SYBR Green/Rox qPCR (SABiosciences) using the Mx3005P Real-Time PCR System (Stratagene). The fold induction value was calculated according to the 2-ΔΔCt method. GAPDH and ACTB were used as a reference. All experiments were performed in triplicate. Primers for Quantitative Real-Time PCR Analysis The following primers were used to amplify reactive transcripts. Lgr5:forward primer:(SEQ ID NO: 10)5′-AGTTTATCCTTCTGGTGGTAGTCC-3′;reverse primer:(SEQ ID NO: 11)5′-CAAGATGTAGAGAAGGGGATTGA-3′;GAPDH:forward primer:(SEQ ID NO: 12)5′-CTCTGCTCCTCCTGTTCGAC-3′;reverse primer:(SEQ ID NO: 13)5′-ACGACCAAATCCGTTGACTC-3′;ACTB:forward primer:(SEQ ID NO: 14)5′-AAGTCCCTTGCCATCCTAAAA-3′;reverse primer:(SEQ ID NO: 15)5′-ATGCTATCACCTCCCCTGTG-3′ Cell Proliferation Assay Floating and adherent CSCs were plated at about 100 and 1×104cells/well in 96-well plates, respectively. On days 0 and 3, viable cell counts were determined by the Cell Counting Kit-8 Assay (Doujindo) according to the manufacturer's protocol. Average absorbance on day 0 was taken as 100%. For chemosensitivity assay, floating and adherent CSCs were plated at about 100 and 1×104cells/well in 96-well plates, respectively. After 24 hours of incubation, 10 μg/ml 5-FU (Hospira), 10 μg/ml irinotecan (Hospira), 50 mM TCF inhibitor FH535 (Merck), or 50 mM β-catenin inhibitor Cardamonin (Merck) were added to the plates. After three days of culture in the presence of the agents, the Cell Counting Kit-8 was added to the cells. The average absorbance of cells exposed to DMSO or medium alone was taken as 100%. All experiments were performed in triplicate. Immunofluorescent Staining of Cultured Cells and Xenograft Tissues For immunofluorescent cytochemistry, cells were fixed with 4% paraformaldehyde and methanol, and incubated with a mouse anti-human E-cadherin antibody (Abcam), rabbit anti-human Snail antibody (Abcam), or rabbit anti-human β-catenin antibody (Sigma)). Then, the cells were visualized using the AlexaFluor 488-labeled goat anti-mouse IgG antibody or goat anti-rabbit IgG antibody. For immunofluorescent cytochemistry, thin sections derived from paraffin blocks of xenograft tumors described above were incubated with a mouse anti-human Lgr5 antibody (2U2E-2) or rabbit anti-human Snail antibody (Abcam). After incubation with a primary antibody, Lgr5 protein was detected with a goat anti-mouse antibody conjugated with the polymer-HRP (DAKO) and visualized with the AlexaFluor 488-labeled tyramide (Invitrogen), while Snail protein was detected with biotinylated goat anti-rabbit antibody (VECTOR) and visualized with the AlexaFluor 568-labeled streptavidin (Invitrogen). These cells and samples were also stained with DAPI (Invitrogen). Example 1: Establishment of Colorectal Cancer Xenografts As described in a previous report (Fujii E. et al., (2008) Establishment and characterization of in vivo human tumor models in the NOD/SCID/gamma(c)(null) mouse. Pathol Int 58: 559-567), the present inventors established 11 types of human colorectal cancer xenografts using NOD/Shi-scid, IL-2Rγnull (NOG) mice (Table 1; the number of human colorectal cancer cell lines established with immunodeficient NOG mice). TABLE 1AdenocarcinomaG1G2G3TotalNumber of cases446353Established010111Impracticable*0606EBV lymphoma216119Aggravated animal112114condition †No viable cancer1203 In the above Table 1, asterisk indicates cases established but unsuitable for experiment, and dagger indicates cases with infection and such. As shown in Table 1, 17 types of colorectal cancer xenografts were established from samples of 53 human colorectal cancer patients. Except for the 17 types of xenografts, associated EBV-infected lymphoma cells occurred in 19 cases (which aggravated the condition of NOG mice); other infections were found in 14 cases; and no tumor growth was observed in three cases. Of the 17 types, 11 xenografts survived even after freeze-thawing, and had the capacity to reconstitute tumor, and showed similar histopathological features as those of the original tumors. Of the 11 types, 10 xenografts were derived from grade-2 moderately-differentiated adenocarcinomas, and the remaining one was derived from a grade-3 poorly-differentiated adenocarcinoma. Of the 11 types, 10 xenografts were derived from moderately-differentiated colorectal cancer (MDCC), and the remaining one was derived from poorly-differentiated colorectal cancer (PDCC) (Table 2; histopathological classification of the original human colorectal cancers that were used to establish the 11 xenografts). TABLE 2Histopathological classification of original human tumorAJCCDukes'Line No.TypeGradeTNMstagestagePLR30AdenocarcinomaG2pT3N0MXIIBPLR59AdenocarcinomaG2pT3N2MXIIIC2PLR123AdenocarcinomaG2pT4N1MXIIIC1PLR168AdenocarcinomaG2pT3N0MXIIBPLR215AdenocarcinomaG2pT3N0MXIIBPLR241AdenocarcinomaG2pT4N3M1IVDPLR254AdenocarcinomaG2pT4N2MXIIIC2PLR261AdenocarcinomaG2pT3N0MXIIBPLR325AdenocarcinomaG3pT4N1M1IIIC1PLR379AdenocarcinomaG2pT4N2MXIIICC1PLR423AdenocarcinomaG2pT3N0M1IVD Both MDCC and PDCC xenografts reconstituted histopathological morphologies almost equivalent to those of the original tumors. MDCC xenografts formed specific epithelial ducts which contained goblet cells, and small budding clusters (may undergo epithelial-mesenchymal transition (EMT)). In contrast, PDCC xenografts did not form such specific epithelial duct structures (FIGS.1and16). Example 2: Isolation of Large Intestine CSCs In order to isolate large intestine CSCs, the present inventors used two types of MDCC xenografts, i.e., PLR59 and PLR123. These xenografts were chosen by the present inventors because they grew rapidly even after 10 passages in NOG mice while maintaining the capacity to reconstitute tumors with epithelial ducts and small budding clusters (FIG.1). Thus, the present inventors predicted that stable CSCs can be obtained from the xenografts. Flow cytometry analysis of primary cells passaged in NOG mice derived from PLR59 and PLR123 showed that signal level of CD44, ALDH, CD26, and Lgr5 were lower than that of CD133, EpCAM, CD166, CD24, and CD29. This suggests the existence of a small population of CSCs (FIG.2). Indeed, when primary cells derived from PLR59 and PLR123 were grafted subcutaneously to NOG mice at 100 cells/injection site, tumors were formed at half of the injection sites (five of 12 injection sites; Table 3), and the histopathological morphology of tumors is highly similar to that of original tumors in that they had hierarchical organization (FIG.3). However, primary cells derived from PLR59 and PLR23 were injected subcutaneously at 10 cells/site, any tumor was not formed in NOG mice (Table 3). Table 3 shows the cancer formation ability 49 days after inoculation. TABLE 3CellNumber of cells/inoculation siteline*Specimen †1,00010010PLR59Primary12+/12‡(100)5/12(42)0/12(0)Non-adherent6/6(100)6/6(100)1/6(17)(Lgr5−)Adherent (Lgr5+)6/6(100)6/6(100)6/6(100)PLR123Primary12/12(100)5/12(42)0/12(0)Non-adherent6/6(100)5/6(83)2/6(33)(Lgr5−)Adherent (Lgr5+)6/6(100)6/6(100)6/6(100) In Table 3 shown above, asterisk indicates tumor xenografts established with NOG mice, and dagger indicates cell preparations. Primary indicates cells (primary cells) prepared by harvesting xenograft tumor tissues grown in NOG mice and removing erythrocytes and mouse cells. Floating indicates cells prepared by in vitro culturing primary cells under a non-adherent culture condition. Adherent indicates cells prepared by in vitro culturing primary cells under adherent culture conditions. Plus symbol (single) indicates the number of formed tumors, while plus symbol (double) indicates the total number of inoculation sites. Parenthesis indicates percent tumor (cancer) formation. Lgr5+represents Lgr5 positive, and Lgr5−represents Lgr5 negative. Primary cells derived from PLR59 and PLR123 were cultured in serum-free media supplemented with EGF and FGF. This yielded adherent and non-adherent cells. The present inventors harvested the adherent and non-adherent cells and cultured them separately. The adherent cells were grown with a doubling time of about 2.5 days and exhibited mesenchymal cell-like morphology. The non-adherent cells, on the other hand, formed spheroid-like cell clusters but did not proliferate significantly (FIGS.4,5,18, and19). After one-week or longer culture, the cells were assessed for large intestine CSC markers. This demonstrated that both adherent and non-adherent cells were highly homogeneous. The adherent cells were of Lgr5+, ALDH+, CD133+, CD44+, EpCAM+, CD166+, CD24+, CD26+, and CD29+. Meanwhile, the non-adherent cells were of Lgr5−and ALDH−, and thus were different from the adherent cells (FIGS.6and20). Lgr5 mRNA was detected at a significant level in the adherent cells, while it was undetectable in non-adherent cells (FIG.27). Example 3: Analysis of Lgr5 Protein Expression In order to assess the expression of Lgr5 protein, the present inventors prepared two types of Lgr5-specific monoclonal antibodies (2L36, 2T15E-2, and 2U2E-2) respectively for immunohistochemistry and flow cytometry analysis. The antibodies produced by the present inventors were highly specific to Lgr5 without any cross-reactivity to Lgr4 or Lgr6, both of which are highly homologous to Lgr5 (FIGS.28and29). Using the antibodies, the present inventors demonstrated the expression of Lgr5 in the adherent CSCs. Lgr5-positive cells were detected in tumor tissues that were the origins of PLR59 and PLR123 as well as in xenograft cancer tissues therefrom through all passages (FIG.39). The frequency of Lgr5-positive cells was low in the original tumor tissues (0.01% and 0.04% for PLR59 and PLR123, respectively). Regarding the xenograft cancer tissues, the frequency of Lgr5-positive cells was increased as passage number increased; however, there was no further change after tenth generation (FIG.39). On the other hand, the tumor-reconstituting capacity of primary cells from PLR123 xenograft model was also potentiated as passage number increased. The ratio of CSCs in the primary cells, which was estimated based on the capacity to reconstitute tumor, was about 0.1% after 5 passages, and was increased to about 0.4% after 14 passages. Example 4: Tumor-Reconstituting Capacities of Lgr5-Positive and Lgr5-Negative Colorectal CSCs If a group of colorectal cancer stem cells is characterized by Wnt signaling, Lgr5-positive adherent cells alone can form tumors in vivo. To confirm whether this prediction is true, the present inventors assessed the tumor-forming capacities of Lgr5-positive adherent cells and Lgr5-negative non-adherent cells. The result showed that Lgr5-positive adherent cells were more potent than Lgr5-negative non-adherent cells in the capacity to form tumors. However, both Lgr5-positive and Lgr5-negative cells had the capacity to form tumors in NOG mice. Subcutaneous injection of ten Lgr5-positive cells caused tumor formation at every injection site (six of six sites), while Lgr5-negative cells formed tumors at two of six injection sites (PLR123-derived cells) or at a single site (PLR59-derived cells) (Table 3). Lgr5-positive cells, even when injected at only one cell per inoculation site, reconstituted tumors at two of 12 injection sites (PLR123-derived cells) or at a single site (PLR59-derived cells) (FIG.7). The histopathological morphologies of tumors derived from the Lgr5-positive and Lgr5-negative cells were almost the same as those of the original tumors (FIGS.17and40). Furthermore, there was no change in the expression of cell surface markers and tumor-initiating activity of the Lgr5-positive CSCs even after one month of passage culture (FIGS.30and31). Under adherent culture conditions, the Lgr5-positive cells underwent symmetric cell division (FIG.41). Meanwhile, in the presence of matrigel and serum, Lgr5 protein was distributed to one of two daughter cells under the same culture conditions (FIGS.42C and42D), demonstrating that the Lgr5-positive cells undergo asymmetrical cell division. One of CSC's properties is symmetrical cell division for self-renewal, and another characteristic property is asymmetrical cell division. The Lgr5-positive adherent cells divided symmetrically under adherent culture conditions (FIG.41), whereas, in the presence of matrigel and FBS, as seen from the fact that Lgr5 protein was distributed to one of daughter cells (FIG.42), the Lgr5-positive cells underwent asymmetrical cell division, which resulted in two distinct progenies. The results described above demonstrate that Lgr5-positive and Lgr5-negative cells derived from PLR59 and PLR123 are highly pure large intestine CSCs, and that the Lgr5-positive and Lgr5-negative cells correspond to two independent CSC states in colorectal cancer. Example 5: Effect of TCF and β-catenin In the Lgr5-positive cells, the levels of β-catenin, TCF1, TCF3, and TCF4 proteins were upregulated in accordance with the expression of Lgr5. This was not observed in the Lgr5-negative cells (FIGS.7and21). On the other hand, the N-terminal phosphorylation of c-Jun was undetectable in the Lgr5-positive CSCs as compared to the Lgr5-negative CSCs (FIGS.7and21). To answer the question of whether Wnt signaling promotes the proliferation of large intestine CSCs, the present inventors assessed the effects of β-catenin/TCF inhibitor FH535 and Wnt/β-catenin inhibitor cardamonin (which induces β-catenin degradation) on the proliferation of large intestine CSCs. The result showed that 50 M FH535 significantly reduced the proliferation of Lgr5-positive large intestine CSCs but had no effect on the proliferation of Lgr5-negative large intestine CSCs (FIGS.8and22). Meanwhile, 50 M cardamonin reduced the viable cell count to 70% for the Lgr5-positive large intestine CSCs and to about 50% for the Lgr5-negative large intestine CSCs (FIGS.8and22). This result suggests that TCF mediates the proliferation of Lgr5-positive cells and that β-catenin is involved in the survival of large intestine CSCs. Interestingly, the Lgr5-positive cells proliferated even without supplement of EGF and FGF (FIGS.9and23). This finding shows that large intestine CSCs have an intrinsic/autocrine mechanism for activating the Wnt signaling for their proliferation. Example 6: Ability of Large Intestine CSCs to Convert from Lgr5-Positive to Lgr5-Negative State One of CSC's properties is resistance to chemotherapeutic agents. Thus, the present inventors tested large intestine CSCs for the sensitivity to 5-FU and irinotecan. As described above, the Lgr5-positive cells proliferated with a doubling time of about 2.5 days. Meanwhile, the Lgr5-negative CSCs were in a quiescent state in terms of growth. Both 5-FU (10 μg/ml) and irinotecan (10 μg/ml) treatments significantly inhibited the proliferation of Lgr5-positive large intestine CSCs, while they did not affect the proliferation and survival of Lgr5-negative large intestine CSCs (FIGS.10and24). Three-day exposure of the Lgr5-positive large intestine CSCs to 5-FU (10 μg/ml) or irinotecan (10 μg/ml) caused the appearance of cells resistant to the chemotherapeutic agents. Surprisingly, the drug-resistant cells were negative for Lgr5 and had changed in morphology (FIGS.11,32, and25). This finding demonstrates the transition from the Lgr5-positive to Lgr5-negative state. HLA-DMA, TMEM173, ZMAT3, and GPR110 were chosen as markers for use in specific detection of such CSCs stably negative for Lgr5. Immunostaining performed using specific antibodies against the above molecules yielded a specific staining pattern with colon CSCs that converted to negative for Lgr5 after three days of irinotecan exposure (FIG.43). Furthermore, this immunostaining method was demonstrated to be applicable to tissue sections prepared from paraffin blocks, which are used commonly (FIG.43). These findings suggest that HLA-DMA, TMEM173, ZMAT3, GPR110 can serve as specific markers for CSCs that converted to negative for Lgr5. The fluorescence representing Lgr5 positivity (FIG.44A), which had been observed before irinotecan treatment, disappeared after the treatment (FIG.44B). From Lgr5-negative cells again inoculated and cultured in the absence of irinotecan, Lgr5-positive cells appeared four days after the inoculation (FIG.44C), and expanded by eight days after the inoculation (FIG.44D). All the Lgr5-negative drug-resistant cells are negative for Lgr5 (FIGS.44and45) and remained negative for CK20 (FIG.46). This suggests that the transition of colon CSCs from the actively proliferating state to a quiescent state is correlated with the disappearance of Lgr5 molecule. The correlation was also verified by in vitro growth inhibitor-resistance assay (FIG.47). In addition, the ALDH activity was reduced, while there was no alteration in other CSC markers (FIG.48). The Lgr5-negative cells prepared via irinotecan treatment were assessed for the tumor-forming activity. Subcutaneous injection of ten cells derived from PLR59 and PLR123 resulted in formation of tumors in two and one NOG mice (Table 4), respectively. Table 4 shows the tumor-forming activity of Lgr5-negative CSCs 49 days after inoculation. In Table 4 shown below, asterisk indicates tumor xenografts established in NOG mice; plus symbol (single) indicates the number of animals bearing tumors, and plus symbol (double) indicates the total number of animals. TABLE 4CellNumber of cells/inoculation siteline*1,00010010PLR596†/6‡6/62/6PLR1236/66/61/6 In addition, to assess whether Lgr5-negative large intestine CSCs undergo a transition into an Lgr5-positive state, the present inventors cultured adherently Lgr5-negative large intestine CSCs prepared via irinotecan treatment again in a serum-free stem cell culture medium. The cells became positive for Lgr5 and exhibited mesenchymal cell-like morphology (FIGS.12and33), and started to proliferate. On the other hand, when Lgr5-positive adherent large intestine CSCs were cultured in an ultra-low adherent plate, the present inventors observed that some of the cells halted their growth and formed a spheroid-like structure and that the Lgr5 mRNA level was very low (FIGS.12and33). The transition from the Lgr5-positive to Lgr5-negative state (and the reverse) was also confirmed by observations using a single cell in culture. When single Lgr5-positive cells were individually cultured in a multi-well plate, the cells underwent a transition into the Lgr5-negative state within three days after irinotecan treatment. When single Lgr5-negative cells prepared via irinotecan treatment were individually cultured in a multi-well plate in the absence of irinotecan, 19 to 43% of the cells underwent a transition into the Lgr5-positive state within four days (FIG.49and Table 5). TABLE 5Transition ofNumber of cellsstateCell lineLgr5 positiveLgr5 negativeTotalLgr5 positivePLR590(0%)132(100%)132to negativePLR1231(1%)173(99%)174Lgr5 negativePLR5918(19%)78(81%)96to positivePLR12329(43%)39(57%)68 Table 5 shows cell count ratios of Lgr5-positive and -negative cells stained by 5 immunocytochemistry using an anti-Lgr5 antibody (antibody 2L36). Number in parenthesis represents the ratio of Lgr5-positive or -negative cell count. Thus, the present inventors concluded that large intestine CSCs underwent interconversion between the Lgr5-positive and -negative states and the transition does not require any exogenous factor and/or niche environment. Example 7: In Vitro and In Vivo EMT of Lgr5-Positive Large Intestine CSCs Mesenchymal-like cells expressing nuclear β-catenin are considered migratory CSCs and metastatic CSCs that undergo EMT (Brabletz T, Jung A, Spaderna S, Hlubek F, Kirchner T (2005) Opinion: migrating cancer stem cells—an integrated concept of malignant tumor progression. Nat Rev Cancer 5:744-749). Since the morphology of Lgr5-positive large intestine CSCs was similar to that of mesenchymal cells, the present inventors tested whether Lgr5-positive large intestine CSCs correspond to migratory CSCs. Western blot analysis revealed low level expression of cell-surface E-cadherin, high level expression of Snail, and nuclear β-catenin (which is characteristic of EMT) in the Lgr5-positive large intestine CSCs (FIGS.13, and14, and26). In contrast, the Lgr5-negative colorectal CSCs did not show any evidence of EMT. Specifically, cell-surface E-cadherin was expressed at a high level; Snail was expressed at a low level, and there was no nuclear localization of β-catenin. Furthermore, concomitant expression of Snail and Lgr5 was observed in cells that underwent EMT in budding areas of xenograft tumor tissues (FIG.15). This finding supports the view that the Lgr5-positive large intestine CSCs correspond to migratory stem cells. In addition, the present inventors demonstrated that the Lgr5-positive large intestine CSCs formed tumors in multiple tissues including lung, liver, lymph node, and subcutaneous tissues. Interestingly, in the liver, lymph node, and subcutaneous tissues, tumors with epithelial ductal structures were reconstituted by at least 40 days after intravenous injection of tumor cells, whereas such structures were not reconstituted in the lung (FIGS.34and35). Next, the present inventors examined whether Lgr5-negative CSCs directly form the hierarchical organization of cancer or first undergo transition to Lgr5-positive cells in vivo. To find markers for detecting Lgr5-negative CSCs, gene expression profiling was carried out using Lgr5-positive cells, Lgr5-negative cells, and primary cells from xenograft tumors. As a result, HLA-DMA was selected from molecules whose expression can be detected at high level in the Lgr5-negative CSCs as compared to the Lgr5-positive CSC and primary cells (FIG.50). By immunohistochemistry using anti-Lgr5 antibody, anti-HLA-DMA antibody, and anti-EREG antibody, HLA-DMA was demonstrated to be specifically expressed in the Lgr5-negative CSCs (FIG.51). HLA-DMA is also expressed in macrophages. Then, to rule out the possibility that cells stained by immunohistochemistry using the anti-HLA-DMA antibody are macrophages, the present inventors tested not only HLA-DMA but also other markers expressed in CSCs. Immunohistochemistry using an antibody against EREG expressed in both Lgr5-positive and -negative CSCs (FIG.50) confirmed that EREG was expressed in both of Lgr5-positive and Lgr5-negative CSCs (FIG.51). It was demonstrated that Lgr5-negative CSCs could be identified as cells that are positive for both HLA-DMA and EREG by detection using both markers in combination. After injection of a homogeneous population of Lgr5-negative CSCs to NOG mice, cells expressing Lgr5 only weakly for one day after the injection, which however remained positive for HLA-DMA and EREG, appeared. Then, cells that are negative for HLA-DMA but remain positive for Lgr5 and EREG appeared by five days after the injection (FIG.52). Tumors derived from Lgr5-negative CSCs had specific ductal structures and included Lgr5-positive cells (FIG.53). To probe the possibility of in vivo transition to a growth inhibitor-resistant state, irinotecan was administered at the maximum tolerated dose (MTD) (120 mg/kg) into the peritoneal cavities of NOG mice bearing tumors derived from Lgr5-positive CSCs. Tumor growth was inhibited almost completely (FIG.55), and the ductal structures were collapsed extensively (FIG.54). This condition resulted in a dramatic decrease of Lgr5-positive cells (FIGS.54and56). The number of Lgr5-negative and HLA-DMA-positive cells increased significantly after irinotecan treatment. By contrast, in vehicle-treated control mice, about one third of cancer cells were positive for Lgr5 in both ductal and budding areas (FIG.54). Both Lgr5-positive cells and HLA-DMA-positive and Lgr5-negative cells were positive for EREG, and were identified to be CSCs (FIG.54). After irinotecan treatment, Lgr5-positive cells appeared again (FIG.54). The results described above, when considered together, suggest that Lgr5-negative CSCs can be the origin of colorectal cancer after growth inhibitor treatment and reconstitute cancer hierarchy via transition to Lgr5-positive cells. Example 8: Identification of Molecules Specifically Expressed in Cancer Stem Cells 1. Preparation of Lgr5-Negative Adherent Cells by Irinotecan Treatment Using a stem cell medium, Lgr5-positive adherent cells were seeded at 3×105cells/well in a 6-well plate (BD, Cat. No. 353046). On the following day, irinotecan (Hospira, 61703-349-09) was added to cells at a final concentration of 10 μg/ml. After three-day culture, irinotecan-resistant cells were detected. The cells were harvested using Accutase, and suspended in FACS buffer. Then, the cells were incubated at 4° C. for 30 minutes with 7-AAD Viability Dye as dead cell staining and each of the following antibodies as cancer stem cell markers: FITC-labeled mouse mAb to human CD326 (EpCAM), PE-labeled mouse mAb to human CD133/1 (AC133), PE-labeled mouse mAb to human CD44, PE-labeled mouse mAb to human CD166, PE-labeled mouse mAb to human CD24, PE-labeled mouse mAb to human CD26, or PE-labeled mouse mAb to human CD29. To detect Lgr5, the cells were incubated with the mouse mAb to human Lgr5 at 4° C. for 30 minutes. After washing once with FACS buffer, the cells were incubated with the PE-labeled goat Ab to mouse IgG2a at 4° C. for 30 minutes. Then, after washing once with FACS buffer, the cells were subjected to flow cytometry analysis. The ALDH activity was detected using AldeFluor Kit according to the procedure recommended by the manufacturer. Flow cytometry analysis was performed using EPICS ALTRA. Cells negative for 7-AAD Viability Dye were analyzed for cancer stem cell markers. The irinotecan-resistant cells were demonstrated to change from positive to negative for Lgr5. 2. Identification of Molecules Specifically Expressed in Cancer Stem Cells Primary cells from PLR59 and PLR123, high proliferative Lgr5-positive cancer stem cells prepared by adherent culture of primary cells, and low proliferative Lgr5-negative cancer stem cells prepared by irinotecan treatment of the cells as described above were homogenized mechanically with QIAshredder (Qiagen, Cat. No. 79654), and RNAs were extracted from them using RNeasy Mini Kit (Qiagen, Cat. No. 74104) and RNase-Free DNase Set (Qiagen, Cat. No. 79254) according to the procedure recommended by the manufacturer. The extracted RNAs were analyzed for the purity and quality using Agilent 2100 Bioanalyzer. Following cRNA synthesis, gene expression information was obtained using GeneChip (HG-U133 plus2) of Affymetrix. Data analysis was performed with Microsoft Excel and Statistics software R. The three types of cells (primary cells, Lgr5-positive cells, and Lgr5-negative cells) were compared to each other to make a list of genes whose expression levels are significantly increased in each cell type. Specifically, raw data from GeneChip were normalized and log 2 transformed by GCRMA to calculate differences in the expression level between distinct sample types (three types: primary cells and Lgr5-positive cells, Lgr5-positive cells and Lgr5-negative cells, and Lgr5-negative cells and primary cells). The criteria used for selecting differently expressed genes were:(1) genes showing a twofold or more change in Lgr5-positive cells as compared to primary cells and a twofold or more change in Lgr5-negative cells as compared to primary cells (expressed at high levels in both Lgr5-positive and -negative cancer stem cells) (Table 6-1 to 6-10) (partial amino acid sequences of proteins encoded by the genes are shown in SEQ ID NOs: 1 to 7);(2) genes showing a twofold or more change in Lgr5-positive cells as compared to primary cells and a less than twofold change in Lgr5-negative cells as compared to primary cells and (expressed at a high level in Lgr5-positive cancer stem cells alone) (Table 7-1 to 7-5);(3) genes showing a less than twofold change in Lgr5-positive cells as compared to primary cells and a twofold or more change in Lgr5-negative cells as compared to primary cells (expressed at a high level in Lgr5-negative cancer stem cells alone) (Table 8-1 and 8-2) (partial amino acid sequences of proteins encoded by the genes are shown in SEQ ID NOs: 8 and 9). Furthermore, to identify genes encoding proteins that are presented specifically on cell membrane of cancer stem cells, genes of GO:0005886 [plasma membrane] were extracted from GeneOntology (GO). Then, the present inventors extracted genes with GO:0005576 [extracellular region], GO:0009986 [cell surface], and GO:0016020 [membrane], or genes which are predicted to have a transmembrane region by membrane protein prediction software TMHMM and to have a signal peptide by signal peptide prediction software SignalP, and which do not have GO:0031090 [organelle membrane]. Furthermore, with the aid of GeneChip data from normal colorectal tissues, the present inventors exclude genes whose expression levels are relatively high in normal tissues or primary cells as well as gene only showing a small fold-change in Lgr5-positive or Lgr5-negative cells. TABLE 6-1DBPLR59PLR123PLR59PLR123SEQaccessionprimaryprimaryprimaryprimary-IDNO.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NO.NP_036438.2FAIM2Fas apoptotic inhibitory molecule 28.47.05.44.1—NP_116265.1JUBjub, ajuba homolog (Xenopus laevis)5.85.76.25.5—NP_116281.2FRMD5FERM domain containing 54.06.72.54.8—NP_071731.1EDARectodysplasin A receptor7.07.65.34.61NP_001240622.1MCOLN3mucolipin 33.75.10.82.5—NP_001007098.1NTRK2neurotrophic tyrosine kinase, receptor, type 23.65.21.02.6—NP_001243.1CD70CD70 molecule4.04.94.14.72NP_062818.1SLCO1B3solute carrier organic anion transporter family, member 1B34.94.43.03.9—NP_003606.3SLC4A7solute carrier family 4, sodium bicarbonate2.94.01.63.2—cotransporter, member 7NP_005836.2ABCC4ATP-binding cassette, sub-family C (CFTR/MRP),4.04.01.82.4—member 4NP_003920.1RAB7L1RAB7, member RAS oncogene family-like 15.55.35.66.0—NP_009162.1SLC6A14solute carrier family 6 (amino acid transporter), member1.93.74.76.3—14NP_872631.1EFNA4ephrin-A42.63.61.42.5—NP_001423.1EREGepiregulin2.53.32.63.13NP_001127839.1SLC6A6solute carrier family 6 (neurotransmitter transporter,3.33.30.81.6—taurine), member 6NP_003497.2FZD6frizzled homolog 6 (Drosophila)3.73.24.64.0—NP_003264.2TM7SF2transmembrane 7 superfamily member 24.04.26.86.2—NP_001172024.1AIF1Lallograft inflammatory factor 1-like3.52.92.32.0—NP_060033.3IL17RDinterleukin 17 receptor D3.12.82.43.0—NP_000013.2ADAadenosine deaminase2.12.72.82.9—NP_004834.1IL27RAinterleukin 27 receptor, alpha3.62.62.21.8— Table 6-2 is a continuation of Table 6-1. TABLE 6-2DBPLR59PLR123PLR59PLR123SEQaccessionprimaryprimaryprimaryprimaryIDNO.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_001731.2CALB2calbindin 23.82.52.40.5—NP_065209.2ANK1ankyrin 1, erythrocytic5.82.54.92.4—NP_001157412.1PYGLphosphorylase, glycogen, liver1.22.51.72.1—NP_001097.2ACVR2Bactivin A receptor, type IIB2.42.51.51.2—NP_001129141.1XPR1xenotropic and polytropic retrovirus receptor 13.42.53.52.4—NP_001523.2SLC29A2solute carrier family 29 (nucleoside transporters),1.62.40.51.7—member 2NP_563615.3DCBLD2discoidin, CUB and LCCL domain containing 22.95.35.24.9—NP_689522.2PIK3AP1phosphoinositide-3-kinase adaptor protein 13.52.41.31.0—NP_054772.1FLVCR1feline leukemia virus subgroup C cellular receptor 13.52.41.71.2—NP_006849.1TMED1transmembrane emp24 protein transport domain2.62.32.41.9—containing 1NP_116254.4TNS4tensin 41.92.31.91.5—NP_001193874.1CSPG5chondroitin sulfate proteoglycan 5 (neuroglycan C)3.92.21.50.5—NP_000667.1ADORA2Badenosine A2b receptor1.52.10.71.5—NP_064423.2ACCN2amiloride-sensitive cation channel 2, neuronal1.81.91.71.3—NP_001018000.1KAZkazrin2.51.82.01.8—NP_004773.1SNAP29synaptosomal-associated protein, 29 kDa0.31.70.81.9—NP_066292.2KCNJ12potassium inwardly-rectifying channel,3.43.54.03.4—subfamily J, member 12NP_938205.1FLRT3fibronectin leucine rich transmembrane protein 32.81.57.15.0—NP_115899.1PARD6Gpar-6 partitioning defective 6 homolog gamma (C.1.81.02.71.9—elegans)NP_066924.1CLDN1claudin 11.40.73.43.5—NP_066939.1ADCY1adenylate cyclase 1 (brain)2.92.22.11.4— Table 6-3 is a continuation of Table 6-2. TABLE 6-3DBPLR59PLR123PLR59PLR123SEQaccessionprimaryprimaryprimaryprimaryIDNO.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_001127807.1AIMP2aminoacyl tRNA synthetase complex-interacting1.00.61.20.7—multifunctional protein 2NP_619539.1AKAP7A kinase (PRKA) anchor protein 72.01.31.41.2—NP_064715.1ANKMY2ankyrin repeat and MYND domain containing 22.72.53.23.4—NP_112591.2APH1Banterior pharynx defective 1 homolog B (C. elegans)0.81.61.13.1—NP_658985.2AP0A1BPapolipoprotein A-I binding protein1.81.32.21.6—NP_940852.3APOOLapolipoprotein O-like1.61.62.32.3—NP_647537.1ATRNattractin1.52.11.11.8—NP_001193.2BMP4bone morphogenetic protein 42.94.21.92.84NP_001720.1BTCbetacellulin1.42.51.73.2—NP_001224.1CAV2caveolin 21.92.93.74.2—NP_001788.2CDH11cadherin 11, type 2, OB-cadherin (osteoblast)6.22.43.80.6—NP_857592.1CKLFchemokine-like factor0.91.6-0.81.1—NP_612419.1CMTM7CKLF-like MARVEL transmembrane domain2.84.22.33.2—containing 7NP_054860.1CNTNAP2contactin associated protein-like 24.64.33.83.7—NP_004738.3DLG5discs, large homolog 5 (Drosophila)1.51.01.51.2—NP_001926.2DPP4dipeptidyl-peptidase 40.11.32.03.8—NP_690611.1FASFas (TNF receptor superfamily, member 6)1.40.13.42.1—NP_001099043.1FBX045F-box protein 451.01.30.31.2—NP_001138390.1FGFR2fibroblast growth factor receptor 23.91.73.01.0—NP_001457.1FZD2frizzled homolog 2 (Drosophila)5.25.42.62.9—NP_031379.2GNA12guanine nucleotide binding protein (G protein) alpha 121.20.51.10.3— Table 6-4 is a continuation of Table 6-3. TABLE 6-4DBPLR59PLR123PLR59PLR123SEQaccessionprimaryprimaryprimaryprimaryIDNO.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_001243343.1GNAI1guanine nucleotide binding protein (G protein), alpha4.04.67.68.2—inhibiting activity polypeptide 1NP_002766.1HTRA1HtrA serine peptidase 15.17.05.06.3—NP_001543.2IGEBP4insulin-like growth factor binding protein 43.42.70.51.1—NP_001002915.2IGFL2IGF-like family member 23.03.32.32.5—NP_001034659.2KREMEN1kringle containing transmembrane protein 13.03.71.32.3—NP_005597.3LGMNlegumain1.11.80.51.4—NP_002303.2LIG4ligase IV, DNA, ATP-dependent3.73.75.65.4—NP_006024.1LIPGlipase, endothelial2.12.60.11.1—NP_001392.2LPAR1lysophosphatidic acid receptor 12.93.41.32.4—NP_036284.1LPAR3lysophosphatidic acid receptor 36.74.93.72.9—NP_002327.2LRP6low density lipoprotein receptor-related protein 62.81.81.50.6—NP_055414.2MAGED2melanoma antigen family D, 21.11.82.72.9—NP_005922.2MICBMHC class I polypeptide-reiated sequence B2.63.62.33.7—NP_001182555.1MLLT10myeloid/lymphoid or mixed-lineage leukemia (trithorax2.52.01.01.3—homolog, Drosophila); translocated to, 10NP_002435,1MSNmoesin3.01.72.20.0—NP_001018169.1NAE1NEDD8 activating enzyme El subunit 11.81.71.21.8—NP_056146.1NCSTNnicastrin1.50.42.30.8—NP_060562.3NETO2neuropilin (NRP) and tolloid (TLL)-like 27.36.18.06.9—NP_009014.2NUDT6nudix (nucleoside diphosphate linked moiety X)-type3.21.94.22.9—motif 6NP_689501.1ORAI3ORAI calcium release-activated calcium modulator 31.51.14.32.2—NP_002605.2PDZK1PDZ domain containing 18.18.34.93.6— Table 6-5 is a continuation of Table 6-4. TABLE 6-5DBPLR59PLR123PLR59PLR123SEQaccessionprimaryprimaryprimaryprimaryIDNO.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_055824.1PDZRN3PDZ domain containing ring finger 32.62.51.40.6—phospholipase A2, group IVA (cytosolic, calcium-NP_077734.1PLA2G4Adependent)7.78.25.05.9—NP_001123508.1PLEKHB1pleckstrin homology domain containing, family B4.05.35.16.3—(evectins) member 1NP_079501.2PNPLA3patatin-like phospholipase domain containing 34.44.91.73.5—NP_004641.1PNPLA4patatin-like phospholipase domain containing 42.31.92.82.2—NP_006395.2PROCRprotein C receptor, endothelial1.31.54.23.65NP_001159449.1PROM2prominin 25.44.610.510.46NP_077748.3PSTPIP2proline-serine-threonine phosphatase interacting protein2.22.75.14.9—2NP_002834.3PTPRJprotein tyrosine phosphatase, receptor type, J2.93.01.52.2—NP_002861.1RAB13RAB13, member RAS oncogene family1.30.92.01.4—NP_066361.1RAP2ARAP2A, member of RAS oncogene family2.01.72.41.7—NP_001094058.1RC3H2ring finger and CCCH-type zinc finger domains 21.20.32.00.0—NP_002897.1RDXradixin4.75.34.54.6—NP_006502.1RSC1A1regulatory solute carrier protein, family 1, member 11.10.01.20.4—(DDI2)(DNA-damage inducible 1 homolog 2)NP_004162.2SLC1A2solute carrier family 1 (glial high affinity7.47.32.11.9—glutamate transporter), member 2NP_006349.1SLC25A17solute carrier family 25 (mitochondrial2.41.92.41.5—carrier; peroxisomal membrane protein, 34 kDa), member 17NP_075053.2SLC30A5solute carrier family 30 (zinc transporter), member 51.30.91.20.8—NP_001070253.1SLC7A6solute carrier family 7 (cationic amino acid transporter,1.00.21.50.8—y+ system), member 6NP_071420.1SMOC1SPARC related modular calcium binding 12.65.4-0.12.4—NP_001159884.1SMOC2SPARC related modular calcium binding 27.28.4-0.92.2—NP_054730.1SOCS5suppressor of cytokine signaling 52.31.72.51.9— Table 6-6 is a continuation of Table 6-5. TABLE 6-6DBPLR59PLR123PLR59PLR123SEQaccessionprimaryprimaryprimaryprimaryIDNO.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_001030127.1SORBS1sorbin and SH3 domain containing 16.44.31.1−0.2—NP_003095.2SORDsorbitol dehydrogenase1.51.81.02.1—NP_003705.1STC2stanniocalcin 23.63.11.22.3—NP_005810.1STX6syntaxin 63.42.54.43.2—NP_003229.1TGFB2transforming growth factor, beta 25.35.14.23.6—NP_001124388.1TGFBR1transforming growth factor, beta receptor 11.91.81.51.0—NP_057635.1TM7SF3transmembrane 7 superfamily member 31.91.62.72.2—NP_653233.3TMEM182transmembrane protein 1823.33.44.64.6—NP_003802.1TNFSF9tumor necrosis factor (ligand) superfamiiy, member 94.54.45.24.97NP_005714.2TSPAN5tetraspanin 51.82.90.01.5—NP_068835.1UTS2urotensin 22.41.72.52.3— Table 6-7 is a continuation of Table 6-6. TABLE 6-7PLR59PLR123PLR59PLR123SEQDB accessionprimaryprimaryprimaryprimaryIDNo.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_056222.2ABHD14Aabhydrolase domain containing 14A1.30.92.71.9—NP_004449.1ACSL4acyl-CoA synthetase long-chain family member 44.54.53.64.2—NP_665812.1AlFM1apoptosis-inducing factor, mitochondrion-associated, 11.91.62.01.9—aldo-keto reductase family 1, member C1 (dihydrodiolNP_001344.2AKR1C1dehydrogenase 1; 20-alpha (3-alpha)-hydroxysteroid9.47.88.12.7—dehydrogenase)NP_940683.1ANKRD46ankyrin repeat domain 462.61.83.52.2—NP_077027.1APOOapolipoprotein O1.41.51.21.0—NP_004766.2B4GALT6UDP-Gal:betaGlcNAc beta 1,4-galactosyltransferase,2.01.50.31.1—polypeptide 6NP_004326.1BST2bone marrow stromal cell antigen 2−3.94.4−3.95.9—NP_001185983.1C16orf5chromosome 16 open reading frame 53.12.66.95.4—NP_115700.1C1orf57chromosome 1 open reading frame 571.51.33.03.0—NP_653181.1C1orf85chromosome 1 open reading frame 851.10.82.92.4—NP_001074293.1C2orf89chromosome 2 open reading frame 891.91.70.81.1—NP_775823.1C3orf58chromosome 3 open reading frame 582.00.31.6−0.4—NP_439896.1C6orf192chromosome 6 open reading frame 1924.02.54.53.3—NP_001135942.1C6orf203chromosome 6 open reading frame 2031.21.02.01.8—NP_620140.1C6orf72chromosome 6 open reading frame 721.31.81.21.7—NP_001243894.1CCDC51coiled-coil domain containing 511.10.93.63.1—NP_001157882.1CDK5cyclin-dependent kinase 51.92.03.53.3—NP_055061.1CELSR1cadherin, EGF LAG seven-pass G-type receptor 10.51.50.61.1—(flamingo homolog, Drosophila)NP_004077.1COCHcoagulation factor C homolog, cochlin (Limulus2.12.30.52.2—polyphemus)NP_001896.2CTPSCTP synthase2.01.01.40.7—NP_055191.2CYFIP2cytoplasmic FMR1 interacting protein 22.63.95.05.3—NP_004393.1DFFBDNA fragmentation factor, 40kDa, beta polypeptide2.31.81.71.5—(caspase-activated DNase)NP_001077058.1E2F5E2F transcription factor 5, p130-binding2.82.22.72.2— Table 6-8 is a continuation of Table 6-7. TABLE 6-8PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_056067.2EHBP1EH domain binding protein 12.31.91.10.4—NP_060682.2ENAHenabled homolog (Drosophila)4.43.51.51.2—NP_001180289.1EXD2exonuclease 3′-5′ domain containing 22.31.22.71.9—NP_660323.3FAM119Afamily with sequence similarity 119, member A5.45.13.03.7—NP_620775.2FAM175Afamily with sequence similarity 175, member A3.42.83.82.8—NP_937995.1FAM189Bfamily with sequence similarity 189, member B1.71.71.31.1—NP_055679.1FAM2OBfamily with sequence similarity 20, member B1.60.81.50.8—NP_942600.1FIBPfibroblast growth factor (acidic) intracellular binding protein1.01.22.01.7—NP_000139.1FUT1fucosyltransferase 1 (galactoside 2-alpha-L-7.05.63.14.0—fucosyltransferase, H blood group)NP_000135.2FXNfrataxin1.71.12.01.6—NP_000393.4G6PDglucose-6-phosphate dehydrogenase0.81.02.01.4—NP_000143.2GAAglucosidase, alpha; acid1.21.43.32.2—NP_000160.1GLAgalactosidase, alpha2.01.11.50.9—NP_002072.2GPC1glypican 11.01.22.60.5—NP_001008398.2GPX8glutathione peroxidase 8 (putative)5.05.57.17.0—NP_005329.3HIP1huntingtin interacting protein 12.62.72.62.5—NP_254274.1IL33interleukin 33−0.51.23.63.4—NP_002262.3IPO5importin 53.62.31.90.4—NP_060573.2LRRC8Dleucine rich repeat containing 8 family, member D2.01.01.91.2—NP_067679.6MFAP3Lmicrofibrillar-associated protein 3-like1.50.23.31.2—NP_612440.1MFSD3major facilitator superfamily domain containing 30.91.32.02.4—NP_066014.1MOV10Mov10, Moloney leukemia virus 10, homolog (mouse)0.51.41.32.0—NP_036351.3MRASmuscle RAS oncogene homolog1.10.64.31.3—NP_057034.2MRPL2mitochondrial ribosomal protein L21.30.80.80.0—NP_065972.3NINninein (GSK3B interacting protein)2.03.12.73.0— Table 6-9 is a continuation of Table 6-8. TABLE 6-9PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_065181.1NIPAL3NIPA-like domain containing 31.71.33.02.3—NP_002504.2NME3non-metastatic cells 3, protein expressed in1.12.11.51.9—NP_057672.1NRN1neuritin 18.55.77.76.3—NP_689643.2OR51 Elolfactory receptor, family 51, subfamily E, member 16.99.80.33.0—NP_071748.2OSGEPL1O-sialoglycoprotein endopeptidase-like 14.14.73.14.4—NP_079431.1PAAF1proteasomal ATPase-associated factor 11.92.11.51.9—NP_000523.2PCCBpropionyl CoA carboxylase, beta polypeptide1.41.61.42.0—NP_061757.1PCDHB14protocadherin beta 141.40.51.40.1—NP_002622.2PGDphosphogluconate dehydrogenase1.21.41.21.2—NP_003550.1PIP4K2Bphosphatidylinositol-5-phosphate 4-kinase, type II, beta1.81.31.81.1—NP_056530.2PLA2G3phospholipase A2, group III2.64.30.13.2—NP_005038.1PSMD5proteasome (prosome, macropain) 26S subunit,1.50.71.70.7—non-ATPase, 5NP_006255.1PTPN13protein tyrosine phosphatase, non-receptor type 130.11.10.72.2—(APO-1/CD95 (Fas)-associated phosphatase)NP_057161.1PTRH2peptidyl-tRNA hydrolase 21.41.41.51.8—NP_037390.2PYCARDPYD and CARD domain containing0.21.31.12.1—NP_055113.2QPRTquinolinate phosphoribosyltransferase2.53.63.14.5—NP_060233.3RNF43ring finger protein 432.72.71.21.5—NP_060616.1RNMTL1RNA methyltransferase like 11.50.91.71.1—NP_003698.1RUVBL1RuvB-like 1 (E. coli)1.92.21.32.2—NP_002949.2RYKRYK receptor-like tyrosine kinase1.51.01.21.2—NP_116250.3SERAC1serine active site containing 11.80.82.71.5—NP_005016.1SERPINI1serpin peptidase inhibitor, clade I (neuroserpin), member 15.58.72.14.2—NP_008927.1SLC19A2solute carrier family 19 (thiamine transporter), member 23.82.72.01.2—NP_065075.1SLC39A10solute carrier family 39 (zinc transporter), member 102.93.12.62.7—NP_060306.3SLC41A3solute carrier family 41, member 31.91.51.30.8— Table 6-10 is a continuation of Table 6-9. TABLE 6-10PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_005692.1SNUPNsnurportin 12.22.42.32.5—NP_055563.1SNX17sorting nexin 170.81.11.01.0—NP_003156.1STXBP1syntaxin binding protein 14.35.74.95.3—NP_003192.1TFAMtranscription factor A, mitochondrial2.11.61.11.6—NP_444283.2THEM4thioesterase superfamily member 41.6−0.52.80.4—NP_612472.1TLCD1TLC domain containing 12.72.13.94.2—NP_057548,1TMEM138transmembrane protein 1381.82.22.02.7—NP_085054,2TMEM177transmembrane protein 1773.13.23.54.1—NP_056236.2TMEM186transmembrane protein 1862.42.02.02.3—NP_078863.2TMEM53transmembrane protein 531.21.31.21.0—NP_001008495.2TMEM64transmembrane protein 646.75.78.38.0—NP_669630.1TMEM68transmembrane protein 681.81.83.42.9—NP_861448.2TMTC3transmembrane and tetratricopeptide repeat containing 32.51.83.82.3—NP_775107.1TRIM59tripartite motif-containing 593.74.33.04.0—NP_003293.2TRIP6thyroid hormone receptor interactor 62.71.35.13.3—NP_009215.1TWF2twinfilin, actin-binding protein, homolog 2 (Drosophila)0.61.51.41.8—NP_079094.1UBA5ubiquitin-like modifier activating enzyme 51.82.12.42.4—NP_060769.4UBE2Wubiquitin-conjugating enzyme E2W (putative)1.31.72.42.4—NP_001017980.1VMA21VMA21 vacuolar H+-ATPase homolog (S. cerevisiae)3.02.62.92.6—NP_660295.2ZG16Bzymogen granule protein 16 homolog B (rat)1.12.01.41.9—NP_115549.2ZNRF3zinc and ring finger 34.74.12.22.6— (The values in Tables 6-1 to 6-10 shown above represent the expression difference (log 2 ratio).) TABLE 7-1PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_003658.1LGR5leucine-rich repeat-containing G protein-coupled receptor 53.03.8−1.50.4—NP_066301.1SNTB1syntrophin, beta 1 (dystrophin-associated protein A1,3.13.10.00.4—59kDa, basic component 1)NP_001123575.1COL13A1collagen, type XIII, alpha 12.02.9−0.20.3—NP_001167538.1FGER1fibroblast growth factor receptor 11.32.3−0.70.6—NP_004432.1EPHB1EPH receptor B11.02.3−0.30.7—NP_002002.3FGFR4fibroblast growth factor receptor 42.02.2−1.9-0.5—NP_004622.2LRP8low density lipoprotein receptor-related protein 8,2.32.10.50.7—apolipoprotein e receptorNP_002832.3PTPRGprotein tyrosine phosphatase, receptor type, G1.82.1−5.0−5.4—NP_004727.2XPR1xenotropic and polytropic retrovirus receptor 11.52.10.70.9—NP_859052.3QSOX2quiescin Q6 sulfhydryl oxidase 21.82.00.70.8—NP_003876.1CDK5R1cyclin-dependent kinase 5, regulatory subunit 1 (p35)3.13.30.32.0—NP_002821.1PTPN4protein tyrosine phosphatase, non-receptor type 41.71.90.30.7—(megakaryocyte)NP_001457.1FZD2frizzled homolog 2 (Drosophila)5.25.42.62.9—NP_000674.2ADRA2Cadrenergic, alpha-2C-, receptor1.81.60.1−0.1—NP_000334.1SLC5A1solute carrier tam ly 5 (sodium/glucose cotransporter),2.21.60.20.6—member1NP_005901.2MAPTmicrotubule-associated protein tau1.91.50.40.6—NP_598328.1SYN2synapsin II0.31.70.20.5—NP_005496.4SCARB1scavenger receptor class B, member 11.11.2−0.3−0.3—NP_004434,2EPHB3EPH receptor B30.71.0−0.5−0.4—NP_000259.1NF2neurofibromin 2 (merlin)1.20.90.60.1—NP_003477.4SLC7A5solute carrier family 7 (cationic amino acid transporter,2.00.8−0.4−0.7—y+ system), member 5NP_054740.3SSX2IPsynovial sarcoma, X breakpoint 2 interacting protein1.80.70.9−0.3—NP_055736.2LPHN1latrophilin 11.40.60.90.3—NP_004435.3EPHB4EPH receptor B41.30.50.7−0.1— Table 7-2 is a continuation of Table 7-1. TABLE 7-2PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_116197.4LINGO1leucine rich repeat and Ig domain containing 11.20.00.6−1.3—NP_004987.2ABCC1ATP-binding cassette, sub-family C (CFTR/MRP),1.10.90.0−0.2—member 1NP_003174.3ADAM17ADAM metallopeptidase domain 171.00.3−0.3−0.4—NP_620686.1ADAMTS15ADAM metallopeptidase with thrombospondin6.14.6−3.6−4.2—type 1 motif, 15NP_005091.2AKAP12A kinase (PRKA) anchor protein 122.43.7−1.20.3—NP_001618.2ALCAMactivated leukocyte cell adhesion molecule1.30.90.30.6—NP_001648.1AREGBamphiregulin B1.70.40.0−0.5—NP_001164.2ARHGAP5Rho GTPase activating protein 51.20.40.80.3—NP_542172.2B3GALT6UDP-Gal:betaGal beta 1,3-galactosyltransferase1.10.60.60.3—polypeptide 6NP_001711.2BMP8Bbone morphogenetic protein 8b0.91.30.40.3—NP_006560.3CGREF1cell growth regulator with EF-hand domain 10.91.00.30.8—NP_058647.1CKLFchemokine-like factor0.91.5−0.50.9—NP_849199.2CMTM8CKLF-like MARVEL transmembrane domain1.31.40.60.7—containing 8NP_001422.1EPB41L2erythrocyte membrane protein band 4.1-like 21.30.9−0.4−0.4—NP_004433.2EPHB2EPH receptor 621.00.5−3.1−2.8—NP_000496.2F12coagulation factor XII (Hageman factor)1.21.60.21.0—NP_057133.2FAM158Afamily with sequence similarity 158, member A0.81.20.10.4—NP_001990.2FBN2fibrillin 22.62.5−0.40.1—NP 068656,21FGGfibrinogen gamma chain1.30.60.60.5—(OSMR)(oncostatin M receptor)NP_001439.2GPC4glypican 42.02.1−0.90.0—NP_065857.1GPHNgephyrin3.22.9−3.4−2.6—NP_057399.1GULP1GULP, engulfment adaptor PTB domain containing 11.42.00.40.9—NP_036616.2HMMRhyaluronan-mediated motility receptor (RHAMM)5.04.5−1.7−1.5—NP_0008661IGF1Rinsulin-like growth factor 1 receptor0.71.1−1.9−1.2— Table 7-3 is a continuation of Table 7-2. TABLE 7-3PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_000588.2IGFBP2insulin-like growth factor binding protein 2, 36kDa1.40.70.3−0.4—NP_0611952IL17RBinterleukin 17 receptor B1.01.2−2.7−1.9—NP_002326.2LRP5low density lipoprotein receptor-related protein 51.21.7−0.8−0.2—NP_006491.21MCAMmelanoma cell adhesion molecule2.11.5−0.2−0.4—NP_055456.2MDC1mediator of DNA-damage checkpoint 12.62.2−1.3−1.7—NP_055606.1MELKmaternal embryonic leucine zipper kinase1.51.6−2.0−1.1—NP_000236.2METmet proto-oncogene (hepatocyte growth factor receptor)1.21.20.60.4—NP_065825.1MIB1mindbomb homolog 1 (Drosophila)1.31.30.70.5—NP_005952.2MUC6mucin 6, oligomeric mucus/gel-forming1.13.0−0.11.0—NP_777596.2PCSK9proprotein convertase subtilisin/kexin type 91.51.8−1.6−0.4—NP_003619.2PKP4plakophilin 41.51.40.0−0.1—NP_003042.3SLC16A1solute carrier family 16, member 11.70.60.30.3—(monocarboxylic acid transporter 1)NP_057438.3SLCO4A1solute carrier organic anion transporter family,0.51.2−1.4−0.2—member 4A1NP_003093.2SOD3superoxide dismutase 3, extracellular2.21.70.6−1.2—NP_006425.2SORBS1sorbin and SH3 domain containing 13.23.4−0.90.2—NP_003095.2SORDsorbitol dehydrogenase1.10.70.90.8—NP_000342.2STSsteroid sulfatase (microsomal), isozyme S1.82.2−0.5−0.7—NP_003234.2TGFBR3transforming growth factor, beta receptor III1.41.8−0.2−0.2—NP_055388.2TMEM97transmembrane protein 972.62.11.0−0.3—CAA26435.1TRACT cell receptor alpha constant0.01.10.00.0— Table 7-4 is a continuation of Table 7-3. TABLE 7-4PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_076417.2AACSacetoacetyl-CoA synthetase1.11.60.20.6—NP_653191.2ANKRD22ankyrin repeat domain 222.01.5−1.8−1.7—NP_056020.2ATP11AATPase, class VI, type 11A1.31.00.70.6—NP_001159.2BIRC5baculoviral IAP repeat-containing 52.72.4−4.8−4.4—NP_061189.2CDCA7Lcell division cycle associated 7-like2.11.8−0.60.4—NP_005183.2CDKN3cyclin-dependent kinase inhibitor 34.85.3−3.1−1.8—NP_004077.1COCHcoagulation factor C homolog, cochlin (Limulus1.21.2−0.40.7—polyphemus)NP_0053023GRB10growth factor receptor-bound protein 101.10.4−1.2−1.5—NP_6717043HS6ST2heparan sulfate 6-O-sulfotransferase 21.81.6−1.1−0.2—NP_002262.3IP05importin 52.61.70.5−0.3—NP_114428.1ITFG3integrin alpha FG-GAP repeat containing 31.00.90.0−1.1—NP_002241.1KCNN4potassium intermediate/small conductance calcium-1.20.9−0.7−0.7—activated channel, subfamily N, member 4NP_061159.1KIAA1199KIAA11991.11.1−1.5−2.4—NP_115940.2KISS1RKISS1 receptor1.11.8−3.00.2—NP_057034.2MRPL2mitochondrial ribosomal protein L21.30.80.80.0—NP_000242.1MSH2mutS homolog 2, colon cancer, nonpolyposis type 1 (E.2.52.8−0.8−0.4—coli)NP_055452.3MTFR1mitochondrial fission regulator 11.60.90.2−0.1—NP_005947.3MTHFD1methylenetetrahydrofolate dehydrogenase (NADP+1.71.60.30.1—dependent) 1, methenyltetrahydrofolate cyclohydrolase,formyitetrahydrofolate synthetaseNP_005366.2MYBv-myb myeloblastosis viral oncogene homolog (avian)2.72.3−4.40.2—NP_078938.2NAT10N-acetyltransferase 10 (GCN5-related)1.91.30.1−0.1—NP_777549.1NDUFAF2NADH dehydrogenase (ubiguinone) 1 alpha1.01.4−0.90.1—subcomplex, assembly factor 2NP_004280.5NFE2L3nuclear factor (erythroid-derived 2)-like 31.21.2−0.9−0.1—NP_006672.1NMUneuromedin U0.21.6−4.70.2—NP_055950.1NUP205nucleoporin 205kDa1.91.7−0.3−0.2— Table 7-5 is a continuation of Table 7-4. TABLE 7-5PLR59PLR123PLR59PLR123SEQDB accessionprimaryprimaryprimaryprimaryIDNo.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_942590.1NUP43nucleoporin 43kDa1.30.90.00.0—NP 0554843NUP93nucleoporin 93kDa2.12.40.10.8—NP_006614.2PHGDHphosphoglycerate dehydrogenase3.03.4−2.0−0.4—NP_060904.2RNF130ring finger protein 1300.91.00.40.3—NP_060259.4SEMA4Csema domain, immunoglobulin domain (Ig),1.92.30.20.5—transmembrane domain (TM) and shortcytoplasmic domain, (semaphorin) 40NP_005857.1SIGMAR1sigma non-opioid intracellular receptor 11.01.10.80.9—NP_055413.1SOCS7suppressor of cytokine signaling 71.00.60.70.1—NP_005554.1STMN1stathmin 11.92.1−3.1−1.4—NP_054897,4STXBP6syntaxin binding protein 6 (amisyn)2.12.1−3.4−0.1—NP_001070884.1TMEM231transmembrane protein 2310.51.5−2.2−0.8—NP_002537.3TNFRSF11Btumor necrosis factor receptor superfamily,1.76.3−2.9−0.9—member 11bNP_443195.1TOP1MTtopoisomerase (DNA) I, mitochondrial1.10.40.5−0.1—NP_001058.2TOP2Atopoisomerase (DNA) II alpha 170kDa2.22.7−6.1−3.1—NP_006364.2VAT1vesicle amine transport protein 1 homolog1.01.50.51.0—(T. californica)NP_612471.1ZMYND19zinc finger, MYND-type containing 191.20.70.70.3— (The values in Tables 7-1 to 7-5 shown above represent the expression difference (log 2 ratio).) TABLE 8-1PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_076404.3GPR87G protein-coupled receptor 870.00.07.17.4—NP_005109.2TNFSF15tumor necrosis factor (ligand) superfamily, member 15−2.4−2.33.53.3—NP_001926.2DPP4dipeptidyl-peptidase 40.11.32.03.8—NP_149017.2BBS4Bardet-Biedl syndrome 40.20.61.72.6—NP_542386.1C9orf30chromosome 9 open reading frame 300.20.21.51.7—NP_001447.2FLNAfilamin A, alpha−0.40.80.71.7—NP_000425.1NEU1sialidase 1 (lysosomal sialidase)0.30.41.91.7—NP_002125.3HMOX2hems oxygenase (decycling) 20.30.41.51.6—NP_001078.2AAMPangio-associated, migratory cell protein0.10.31.21.6—NP_061985.2ABCA7ATP-binding cassette, sub-family A (ABC1), member 7−1.30.42.11.6—NP_000401.1HFEhemochromatosis2.22.01.22.8—NP_001142.2SLC25A4solute carrier family 25 (mitochondrial carrier: adenine0.60.81.31.5—nucleotide translocator), member 4NP_006569.1GNB5guanine nucleotide binding protein (G protein), beta 50.20.80.91.4—NP_005846.1RAMP1receptor (G protein-coupled) activity modifying protein 1−0.5−0.51.11.4—NP_598378.3RHOVras homolog gene family, member V0.1−0.20.51.4—NP_112178.2PVRL4poliovirus receptor-related 4−0.3−2.86.11.87NP_003686.1LY6Dlymphocyte antigen 6 complex, locus D−3.8−1.8−0.52.7—NP_775876.1KCNRGpotassium channel regulator1.00.22.61.9—NP_005063.19LC12A4solute carrier family 12 (potassium/chloride−0.2−0.11.21.2—transporters), member 4NP_005292.2GPR35G protein-coupled receptor 35−0.8−1.41.1−0.8—NP_001607.1ACVR2Aactivin A receptor, type IIA0.2−0.51.41.0—NP_001687.1ATP6V1E1ATPase, H+ transporting, lysosomal 31kDa, V1 subunit E1−0.10.31.11.2—NP_036474.1BAMBIBMP and activin membrane-bound inhibitor homolog−0.80.71.32.4—(Xenopus laevis)NP_005177.2CAPN1calpain 1, (mu/l) large subunit−1.3−1.31.3−0.2— Table 8-2 is a continuation of Table 8-1. TABLE 8-2PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryIDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−NONP_001798.2CELcarboxyl ester lipase (bile salt-stimulated lipase)0.00.07.45.4—NP_056034.2EXOC7exocyst complex component 7−0.2−0.21.91.2—NP_000034.1FASFas (TNF receptor superfamily, member 6)0.80.42.82.39NP_005802.1GDF11growth differentiation factor 110.70.91.31.9—NP_060456.3GPR172BG protein-coupled receptor 17280.00.05.64.3—NP_665895.1KLK10kallikrein-related peptidase 10−0.90.41.21.6—NP_055414.2MAGED2melanoma antigen family D, 20.00.32.13.0—NP_000519.2MAN2B1mannosidase, alpha, class 2B, member 1−0.2−0.22.31.5—NP_002434.1MSMBmicroseminoprotein, beta-0.00.05.64.7—NP_009014.2NUDT6nudix (nucleoside diphosphate linked moiety X)-type0.00.01.30.8—motif 6NP_036528.1PHLDA3pleckstrin homology-like domain, family A, member 3−0.3−0.22.91.0—NP_002629.1PI3peptidase inhibitor 3, skin-derived−7.6−7.91.4−0.6—NP_597998.1SAT2spermidine/spermine N1-acetyltransferase family0.70.91.71.2—member 2NP_006207.1SERPINE2serpin peptidase inhibitor, clade E (nexin,−3.2− 2.51.30.7—plasminogen activator inhibitor type 1), member 2NP_036303.1TSPAN17tetraspanin 170.60.41.71.3— (The values in Tables 8-1 and 8-2 shown above represent the expression difference (log 2 ratio).) Furthermore, genes that meet a criterion described below and have GO:0005886 [plasma membrane] from GeneOntology (GO) (Tables 9 and 10) were extracted in order to obtain genes encoding proteins that are specifically presented on cell membrane of cancer stem cells. Markers common for both proliferating and quiescent CSCs: genes whose expression levels are in average greater than 64 in Lgr5-negative and Lgr5-positive cells; which show a greater than four-fold change in both Lgr5-negative and Lgr5-positive cells relative to primary cells; and which show a significant difference by t-test (Table 9). TABLE 9PLR59PLR123PLR59PLR123PLR59PLR123SEQprimaryprimaryprimaryprimaryLgr5+Lgr5+IDDB accession No.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−Lgr5−Lgr5−NONP_001423.1EREGepiregulin2.23.42.53.10.3−0.33NP_001986.2ACSL1acyl-CoA synthetase long-chain family3.03.92.93.7−0.2−0.2—member 1NP_005922.2MICBMHC class I polypeptide-related sequence B2.33.82.23.7−0.1−0.1—NP_000296.2PON2paraoxonase 22.34.62.94.60.70.0—NP_003458.1CXCR4chemokine (C-X-C motif) receptor 410.29.58.57.8−1.7−1.7—NP_062818.1SLCO1B3solute carrier organic anion transporter family,4.45.13.04.1−1.4−1.0—member 183NP_000280.1PF KMphosphofructokinase, muscle3.32.33.32.10.0−0.2—NP_057672.1NRN1neuritin 18.88.57.86.5−1.0−2.0—NP_060369.3TESCtescalcin3.35.02.036−1.3−1.3—NP_000139.1FUT1fucosyltransferase 1 (galactoside 2-alpha-L-6.95.93.14.3−3.9−1.6—fucosyltransferase, H blood group)NP_003802.1TNFSF9tumor necrosis factor (ligand) superfamily,4.54.45.45.10.90.77member 9NP_059108.1FZD3frizzled homolog 3 (Drosophila)2.82.62.63.2−0.20.6—NP_005810.1STX6syntaxin 62.82.74.23.51.40.9—NP_057635.1TM7SF3transmembrane 7 superfamily member 33.02.53.93.00.90.5—NP_000013.2ADAadenosine deaminase2.02.72.82.90.80.2—NP_071731.1EDARectodysplasin A receptor6.26.85.03.7−1.2−3.11NP_003264.2TM7SF2transmembrane 7 superfamily member 23.64.36.96.73.22.4—NP_116265.1JUBjab, ajuba homolog (Xenopus laevis)3.74.34.73.81.0−0.5—NP_689740.2SLC16A14solute carrier family 16, member 145.46.65.86.10.3−0.4—(monocarboxylic acid transporter 14)NP_065209.2ANK1ankyrin 1, erythrocytic6.63.05.32.7−1.3−0.3— Quiescent CSC-specific markers: genes whose expression levels are in average greater than 64 in Lgr5-negative cells and are in average less than 64 in both primary cells and Lgr5-positive cells; which show a greater than 20 fold change in Lgr5-negative cells relative to Lgr5-positive cells; and which show a significant difference by t-test (Table 10). TABLE 10PLR59PLR123PLR59PLR123PLR59PLR123SEQDB accessionprimaryprimaryprimaryprimaryLgr5+Lgr5+IDNo.AbbreviationMolecule nameLgr5+Lgr5+Lgr5−Lgr5−Lgr5−Lgr5−NONP_006111.2HLA-DMAmajor histocompatibility complex,0.00.28.08.78.08.5—class II, DM alphaNP_862830.1AMIGO2adhesion molecule with Ig-like domain 20.00.07.07.57.07.5—NP_001159449.1PROM2prominin 23.32.910.110.56.97.66NP_076404.3GPR87G protein-coupled receptor 870.00.07.07.47.07.4—NP_722582.2GPR110G protein-coupled receptor 1100.00.06.07.06.07.0—NP_112178.2PVRL4poliovirus receptor-related 4−0.6−2.75.81.76.44.48NP_938205.1FLRT3fibronectin leucine rich transmembrane1.30.26.85.45.55.2—protein 3 3. Expression Analysis by Flow Cytometry Analysis 3.1. Flow Cytometry Analysis of NOG-Established Cancer Cell Lines After suspending in FACS buffer, cells of NOG-established cancer lines collected from mice were incubated at 4° C. for 30 minutes with rat mAb to mouse MHC I (Abcam; ab15680) and mAb to human EREG (EP27; WO2008/047723). Then, following washing once with FACS buffer, the cells were incubated at 4° C. for 30 minutes with 7-AAD Viability Dye (Beckman Coulter; A07704) as dead cell staining and secondary antibodies: PE-labeled goat F(ab′)2 fragment to mouse IgG (H+L) (Beckman Coulter; IM0855) and APC-labeled goat Ab to rat IgG (BioLegend; 405406). After washing once with FACS buffer, the cells were subjected to flow cytometry analysis. Flow cytometry analysis was performed using EPICS ALTRA. Cells negative for 7-AAD Viability Dye and mouse MHC were analyzed for EREG expression. 3.2. Flow Cytometry Analysis of In Vitro Cultured Cancer Cell Lines Lgr5-positive adherent cells and Lgr5-negative adherent cells resulting from induction by irinotecan treatment were harvested using Accutase. The cells were suspended in FACS buffer, and then incubated at 4° C. for 30 minutes with mouse mAb to human EREG. After the cells were washed once with FACS buffer, 7-AAD Viability Dye as dead cell staining and a PE-labeled goat F(ab′)2 fragment to mouse IgG (H+L) as a secondary antibody were added thereto. The cells were incubated at 4° C. for 30 minutes. Then, the cells were washed once with FACS buffer, and subjected to flow cytometry analysis. Flow cytometry analysis was performed using EPICS ALTRA. Cells negative for 7-AAD Viability Dye were analyzed for EREG expression. The result showed that the corresponding protein was expressed at a high level on cell membrane surface. The result obtained by EREG flow cytometry analysis of primary cells from PLR59 and PLR123, and Lgr5+and Lgr5−cancer stem cells is shown inFIG.37. The cells were stained using an antibody against EREG and analyzed by flow cytometry. It was demonstrated that primary cells were negative for EREG while Lgr5+and Lgr5−cancer stem cells were homogeneous EREG-positive cell populations. Gray indicates fluorescence intensity after cell staining with an indicated antibody; and white indicates fluorescence intensity after cell staining with a control isotype antibody. 4. In Vitro Assessment of Drug Efficacy by ADCC Activity Measurement 4.1. Preparation of Effector Cell Suspension A mononuclear cell fraction collected from human peripheral blood was used as human effector cells. Fifty ml of peripheral blood was collected from a healthy volunteer (adult male) of the inventors' company using a syringe loaded in advance with 200 μl of 1000 units/ml heparin solution (Novo-Heparin 5,000 units/5 ml for Injection; Novo Nordisk). The peripheral blood was diluted twofold with PBS(−), and then introduced into a Leucosep lymphocyte separation tube (Greiner bio-one) in advance loaded with Ficoll-Paque PLUS and subjected to centrifugation. After centrifugation (2150 rpm, room temperature, 10 minutes), the monocyte fraction layer was collected from the tube. The cells were washed once with 10% FBS/D-MEM, and then suspended at a cell density of 5×106/ml in 10% FBS/D-MEM. The suspension was used as an effector cell suspension. 4.2. Preparation of Target Cell Suspensions Target cell suspensions were prepared at the time of use. One×106cells of cancer lines were centrifuged (1200 rpm, room temperature, 5 minutes). The cell pellets were suspended in 200 μl of 0.2 mg/ml calcein-AM (Nacalai Tesque)/DMEM (10% FBS) medium. Cell suspensions in calcein-AM solution were incubated for two hours in a CO2incubator set to 37° C. and to a CO2concentration of 5%. After washing once with 10% FBS/D-MEM, the cells were adjusted to a cell density of 2×105/ml with 10% FBS/D-MEM to prepare target cell suspensions. 4.3. ADCC Activity Measurement Anti-EREG antibody was prepared at a concentration of 0.5 mg/ml, which was further diluted with 10% FBS/D-MEM to give antibody solutions. The final concentration was adjusted to 0.4, 4, and 40 μg/ml. The antibody solutions of respective concentrations or 10% FBS/D-MEM were each added at 50 μl/well to a 96-well round-bottomed plate. Then, the target cell suspensions were added at 50 μl/well to every well. The plate was incubated at room temperature for 15 minutes. Next, 100 μl of the effector cell suspension was added to each well containing target cell suspension, and antibody solution or 10% FBS/D-MEM. One-hundred μl of 10% FBS/D-MEM or 2% NP-40 solution (NP-40 substitute; Wako Pure Chemical Industries) was added to each of other wells containing 10% FBS/D-MEM and target cell suspension. The plate was centrifuged (1200 rpm, room temperature, 5 minutes) and incubated for 4 hours in a CO2incubator set to 37° C. and to a CO2concentration of 5%. The plate was centrifuged (1200 rpm, room temperature, 5 minutes), and a 100-μl aliquot of supernatant was collected from each well. The fluorescence intensity (λex=490 nm, λem=515 nm) was determined using a spectrofluorometer. The specific calcein release rate (cytotoxicity (%)) was determined according to the following formula. cytotoxicity(%)=(A−C)×100/(B−C)  Formula 1: where A represents the fluorescence intensity in each well; B represents the mean value of fluorescence intensity in a well where 50 μl of target cell suspension and 100 μl of NP-40 solution were added to 50 μl of 10% FBS/D-MEM; and C represents the mean value of fluorescence intensity in a well where 50 μl of target cell suspension and 100 μl of 10% FBS/D-MEM were added to 50 μl of 10% FBS/D-MEM. This assay was carried out in triplicate, and the cytotoxicity (%) at each antibody concentration was determined using Microsoft Office Excel 2007. The anti-EREG antibody-mediated ADCC activities against Lgr5-positive and -negative cells derived from PLR59 cells, and those against Lgr5-positive and -negative cells derived from PLR123 cells are shown inFIG.38. The result showed that the anti-EREG antibody exerted cytotoxic activity against both Lgr5-positive and -negative cancer stem cells from PRL59 or PLR123 in a dose dependent manner whereas the control antibody had no cytotoxic activity. To assess in vivo EREG expression, the Lgr5-positive cells were administered into the peritoneal cavities of NOG mice. In the early stage of tumor generation, EREG was expressed at a high level. In the late stage where the tumor formed specific ductal structures, EREG expression was somewhat localized to the budding clusters rather than ductal structures. EREG-positive cells were detected even after irinotecan administration to tumor-bearing mice (FIG.54). The anti-EREG antibody was assessed for anti-tumor activity after irinotecan treatment. Effector cells are essential for the anti-EREG antibody to mediate ADCC activity. Thus, SCID mice were used as a model to assess the pharmacological efficacy of the anti-EREG antibody. Tumor growth was suppressed when the antibody was administered at the time points of days 4 and 11 after the final irinotecan administration (FIG.57). As a first step to assess the pharmacological efficacy based on the metastasis model, it was tested whether EREG is expressed in the metastasis model. When Lgr5-positive cells were intravenously injected into NOG mice, tumors were formed in multiple tissues including lung. Cells of the tumors formed in lungs are mostly positive for EREG (FIG.58A). The pharmacological efficacy of the anti-EREG antibody was assessed using SCID-Beige mice where macrophages and mononuclear cells can serve as effector cells to mediate ADCC. The anti-EREG antibody was administered to mice once a week for a total of five times starting at three days after the injection of Lgr5-positive cells. The number of tumor cells in distal locations was demonstrated to be markedly reduced as compared to that in control mice (FIG.58B). In addition, the size of each tumor was also shown to be remarkably reduced in mice administered with the antibody (FIGS.58C and58D). Example 9: Presence of Lgr5-Negative and -Positive CSCs in Clinical Tumor Specimens Proliferating and quiescent CSCs were identified by immunohistochemistry using anti-Lgr5 antibody (2U2E-2), anti-HLA-DMA antibody, and anti-EREG antibody (FIG.59and Table 11). Proliferative CSC represents Lgr5-positive cell, while quiescent CSC represents HLA-DMA-positive and EREG-positive cell (Table 11). Lgr5-positive cells which are positive for both HLA-DMA and EREG, and Lgr5-negative cells which are positive for both HLA-DMA and EREG were present in a very small number in primary and metastatic colorectal cancer specimens isolated from colorectal cancer patients (FIG.59). Both Lgr5-positive and -negative cells were detected in eight of 12 specimens of human colorectal cancer tissues. Meanwhile, either Lgr5-positive or Lgr5-negative cells were observed in the remaining four specimens. Throughout all specimens, Lgr5-positive cells accounted for 0.003 to 1.864%, and Lgr5-negative cells accounted for 0.001 to 10.243% (Table 11). TABLE 11Property ofCase numberCSC123456789101112DuctProliferativeP†PPPPPPPPNPNQuiescentNPNPPPPPPPNPBuddingProliferativePNNPPNNPPPNNareaQuiescentNPNPNPNPNNNPFrequencyProliferative1.8640.7860.1360.1210.1190.0950.0630.0540.0180.0100.0030.000Quiescent0.0000.2430.0000.1870.0010.2280.0450.0650.0030.0030.0000.073(P†indicates that proliferating or quiescent CSCs were detected; N indicates that proliferating or quiescent CSCs were undetectable)(Frequency indicates cell percentage) Both Lgr5-positive and -negative CSCs were detected in the ductal and budding areas (FIG.59). Furthermore, in ducts, Lgr5-positive and -negative CSCs were not limited to particular areas but distributed at random over the entire ducts. Example 10: Anti-Tumor Effect of Various Antibodies Used in Combination with Mab-ZAP and Rat-ZAP The present inventors tested whether an anti-tumor effect can be expected with target therapy using a membrane protein expressed at a high level as the target in irinotecan-treated or non-treated PLR59 and PLR123. Commercially available antibodies shown in Table 12 were assessed by flow cytometry (FCM) for the binding activity to antigens expressed on the cell surface of irinotecan-treated or non-treated PLR59 or PLR123. The result is summarized in Table 13. TABLE 12Antigen nameSubtypeManufacturerCD70mIgG3BD PharmingenEDARmIgG1MBLFASmIgG1BD PharmingenPROM2mIgG2bR & D SystemsPVRL4mIgG2bR & D SystemsTNFSF9mIgG1BioLegendPROCRratIgG1BD PharmingenEPCAMmIgG2aABGENT TABLE 13Irinotecan non-treatedIrinotecan-treatedCSCPLR59PLR123PLR59PLR123Antibody +/−−+−+−+−+CD704117741843NTNTNTNTEDAR737761NTNTNTNTFAS45194230221871251480PROM2NTNTNTNT2939119298PVRL4484192210925129TNFSF94864332218325104PROCR6195132251925127EPCAM7756377701NTNTNTNT(NT indicates not tested.) Using Mab-ZAP and Rat-ZAP, various antibodies that had been demonstrated to have binding activity were assessed for the activity of internalization (into cells). Mab-ZAP and Rat-ZAP are anti-mouse IgG antibody and anti-rat IgG antibody, respectively, conjugated with saporin, a toxin that inhibits protein synthesis (Advanced Targeting Systems). To assess the activity of internalization into irinotecan-non-treated cells, PLR59 and PLR123 cells were seeded at a cell density of 30000 cells/80 μl/well to respective wells of a 96-well plate. Following day, each antibody solution was added at a final concentration of 0.01, 0.1, or 1 μg/ml to the respective wells. Then, Mab-ZAP or Rat-ZAP was added at a final concentration 1 μg/ml to the respective wells, and the plate was incubated at 37° C. for 72 hours in a CO2incubator. To assess the activity of internalization into irinotecan-treated cells, PLR59 and PLR123 cells were seeded in a 96-well plate, and irinotecan was added at a final concentration of 15 M to each well. The plate was incubated at 37° C. for 72 hours in a CO2incubator. Various antibodies were assessed for the activity of internalization to cells cultured as described above in the presence or absence of irinotecan. For assay, the various antibodies were each assessed for the internalization activity into cells contained in each well where the medium was replaced with the same irinotecan-free medium as used for irinotecan-non-treated cells. Seventy-two hours after addition of antibodies and Mab-ZAP or Rat-ZAP, 10 μl of 3% SDS (Nacalai Tesque) was added to each well of the plate. The cells in the plate were lysed thoroughly by stirring the plate using a plate mixer. Then, 100 μl of CellTiter-Glo™ Luminescent Cell Viability Assay (Promega) was added to each well. The mixture in each well was assayed to determine its luminescent signal. The determined anti-tumor activity is shown in Table 14 andFIGS.60to72. InFIGS.60to72, the percent suppression of cell proliferation indicated by the vertical axis represents a relative value for the difference in the luminescence signal value between the mixtures in wells, one of which contained a test antibody alone (without Mab-ZAP and Rat-ZAP) and the other contained a test antibody, and Mab-ZAP or Rat-ZAP, when taking as 100% the difference in the luminescence signal value between the mixtures in wells, one of which contained a test antibody alone (without Mab-ZAP and Rat-ZAP) and the other did not contain any cells. In Table 14, symbols, −, +, ++, and +++, represent relative values for the internalization activity when a test antibody was assayed at a concentration of 1 μg/μl. The relative value refers to a relative value for the difference in the luminescence signal value between the mixtures in wells, one of which contained a test antibody alone (without Mab-ZAP and Rat-ZAP) and the other contained a test antibody, and Mab-ZAP or Rat-ZAP, when taking as 100% the difference in the luminescence signal value between the mixtures in wells, one of which contained a test antibody alone (without Mab-ZAP and Rat-ZAP) and the other did not contain any cells. Symbols, −, +, ++, and +++, indicate that the relative value is less than 5%, 5% or more and less than 15%, 15% or more and less than 25%, and 25% or more, respectively. TABLE 14PLR59PLR123AntigenIrinotecanIrinotecan-IrinotecanIrinotecan-namenon-treatedtreatednon-treatedtreatedCD70+++NT+++NTEDAR++NT−NTFAS++++++++++++PROM2NT+NT+PVRL4++++−+TNFSF9−+++−+++PROCR+++−+EPCAM++++++++++++ As shown inFIG.66, regarding irinotecan-non-treated PLR59 and PLR123, anti-CD70 antibody and anti-FAS antibody showed the internalization activity level of 25% or more under a condition where the anti-EPCAM antibody used as a positive control had been demonstrated to exhibit a sufficient anti-tumor activity (FIGS.60and62). Furthermore, regarding PLR59, anti-EDAR antibody showed the internalization activity level of 15 to 25%, and anti-PVRL4 antibody and anti-PROCR antibody exhibited the internalization activity level of 5 to 15% (FIGS.61,63, and65). Meanwhile, regarding irinotecan-treated PLR59 and PLR123, anti-FAS antibody and anti-TNFRSF9 antibody showed the internalization activity level of 25% or more, and anti-PROM2 antibody exhibited the internalization activity level of 5 to 15% (FIGS.67,70, and68). Both anti-PVRL4 antibody and anti-PROCR antibody showed the internalization activity for irinotecan-treated PLR59 and PLR123, and the activity for PLR59 was greater than that for PLR123 (FIGS.69and71). The result described above demonstrates that all antibodies tested have an anti-tumor effect against PLR59 and PLR123. BMP4 was assessed for the differentiation-enhancing effect on irinotecan-non-treated and irinotecan-treated PLR59 and PLR123. To assess the differentiation-enhancing effect on irinotecan-non-treated cells, PLR59 and PLR123 cells suspended in the media where BMP4 (R&D Systems; a final concentration of 20 nM) or a control buffer was added to culture media were seeded at a cell density of 5×105cells/1.5 ml/well to each well of a 12-well plate. The cells were passaged while changing the culture media with the same type of medium 2, 4, and 7 days after seeding. To assess the differentiation-enhancing effect on irinotecan-treated cells, PLR59 or PLR123 was seeded at a cell density of 17×105cells/5 ml/flask to a 12.5-ml culture flask. Following day, irinotecan was added at a final concentration of 15 M. The flask was incubated at 37° C. for 72 hours in a CO2incubator. Then, the medium in the flask was changed with a medium containing BMP4 or a control buffer. The medium was further changed with the same type of medium 2, 4, and 7 days after the initial medium change. From cells isolated 4 and 9 days after the initial medium change, RNAs were extracted using RNeasy Plus Mini Kit and RNase-Free DNase Set (QIAGEN). cDNAs were synthesized with ThermoScript RT-PCR System (Invitrogen) using the extracted RNAs as a template. Quantitative real-time PCR was carried out using the cDNAs isolated as described above. As shown inFIG.73, elevated CK20 levels were observed in PLR59 and PLR123 cells cultured in the presence of BMP4. INDUSTRIAL APPLICABILITY The present inventors identified cell surface molecules that are expressed specifically on cancer stem cells. The present invention provides novel anti-cancer drugs and reagents for detecting cancer stem cells, which use antibodies against the cell surface molecules.
203,817
11858988
DEFINITION OF TERMS In order that the present invention may be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the detailed description. Throughout this specification and the claims which follow, unless the context requires otherwise, the word “comprise”, and variations such as “comprises” and “comprising”, will be understood to imply the inclusion of a stated member, integer or step or group of members, integers or steps but not the exclusion of any other member, integer or step or group of members, integers or steps. The terms “a” and “an” and “the” and similar reference used in the context of describing the invention (especially in the context of the claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”), provided herein is intended merely to better illustrate the invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention. Claudins are a family of proteins that are the most important components of tight junctions, where they establish the paracellular barrier that controls the flow of molecules in the intercellular space between cells of an epithelium. Claudins are transmembrane proteins spanning the membrane 4 times with the N-terminal and the C-terminal end both located in the cytoplasm. The first extracellular loop consists on average of 53 amino acids and the second one of around 24 amino acids. CLDN6 and CLDN9 are the most similar members of the CLDN family. The term “CLDN” as used herein means claudin and includes CLDN6, CLDN9, CLDN4 and CLDN3. Preferably, a CLDN is a human CLDN. The term “CLDN6” preferably relates to human CLDN6, and, in particular, to (i) a nucleic acid comprising a nucleic acid sequence encoding the amino sequence of SEQ ID NO: 2 or encoding the amino sequence of SEQ ID NO: 8 such as a nucleic acid comprising the nucleic acid sequence of SEQ ID NO: 1 or (ii) a protein comprising the amino acid sequence of SEQ ID NO: 2 or comprising the amino acid sequence of SEQ ID NO: 8. The first extracellular loop of CLDN6 preferably comprises amino acids 28 to 80, more preferably amino acids 28 to 76 of the amino acid sequence shown in SEQ ID NO: 2 or the amino acid sequence shown in SEQ ID NO: 8, such as the amino acid sequence shown in SEQ ID NO: 7. The second extracellular loop of CLDN6 preferably comprises amino acids 138 to 160, preferably amino acids 141 to 159, more preferably amino acids 145 to 157 of the amino acid sequence shown in SEQ ID NO: 2 or the amino acid sequence shown in SEQ ID NO: 8, such as the amino acid sequence shown in SEQ ID NO: 6. Said first and second extracellular loops preferably form the extracellular portion of CLDN6. The term “CLDN9” preferably relates to human CLDN9, and, in particular, to (i) a nucleic acid comprising a nucleic acid sequence encoding the amino sequence of SEQ ID NO: 9 or (ii) a protein comprising the amino acid sequence of SEQ ID NO: 9. The first extracellular loop of CLDN9 preferably comprises amino acids 28 to 76 of the amino acid sequence shown in SEQ ID NO: 9. The second extracellular loop of CLDN9 preferably comprises amino acids 141 to 159 of the amino acid sequence shown in SEQ ID NO: 9. Said first and second extracellular loops preferably form the extracellular portion of CLDN9. The term “CLDN4” preferably relates to human CLDN4, and, in particular, to (i) a nucleic acid comprising a nucleic acid sequence encoding the amino sequence of SEQ ID NO: 10 or (ii) a protein comprising the amino acid sequence of SEQ ID NO: 10. The first extracellular loop of CLDN4 preferably comprises amino acids 28 to 76 of the amino acid sequence shown in SEQ ID NO: 10. The second extracellular loop of CLDN4 preferably comprises amino acids 141 to 159 of the amino acid sequence shown in SEQ ID NO: 10. Said first and second extracellular loops preferably form the extracellular portion of CLDN4. The term “CLDN3” preferably relates to human CLDN3, and, in particular, to (i) a nucleic acid comprising a nucleic acid sequence encoding the amino sequence of SEQ ID NO: 11 or (ii) a protein comprising the amino acid sequence of SEQ ID NO: 11. The first extracellular loop of CLDN3 preferably comprises amino acids 27 to 75 of the amino acid sequence shown in SEQ ID NO: 11. The second extracellular loop of CLDN3 preferably comprises amino acids 140 to 158 of the amino acid sequence shown in SEQ ID NO: 11. Said first and second extracellular loops preferably form the extracellular portion of CLDN3. The above described CLDN sequences include any variants of said sequences, in particular mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present. An allelic variant relates to an alteration in the normal sequence of a gene, the significance of which is often unclear. Complete gene sequencing often identifies numerous allelic variants for a given gene. A species homolog is a nucleic acid or amino acid sequence with a different species of origin from that of a given nucleic acid or amino acid sequence. The term “CLDN” shall encompass (i) CLDN splice variants, (ii) CLDN-posttranslationally modified variants, particularly including variants with different glycosylation such as N-glycosylation status, (iii) CLDN conformation variants, (iv) CLDN cancer related and CLDN non-cancer related variants. Preferably, a CLDN is present in its native conformation. CLDN6 has been found to be expressed, for example, in ovarian cancer, lung cancer, gastric cancer, breast cancer, hepatic cancer, pancreatic cancer, skin cancer, melanomas, head neck cancer, sarcomas, bile duct cancer, renal cell cancer, and urinary bladder cancer. CLDN6 is a particularly preferred target for the prevention and/or treatment of ovarian cancer, in particular ovarian adenocarcinoma and ovarian teratocarcinoma, lung cancer, including small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), in particular squamous cell lung carcinoma and adenocarcinoma, gastric cancer, breast cancer, hepatic cancer, pancreatic cancer, skin cancer, in particular basal cell carcinoma and squamous cell carcinoma, malignant melanoma, head and neck cancer, in particular malignant pleomorphic adenoma, sarcoma, in particular synovial sarcoma and carcinosarcoma, bile duct cancer, cancer of the urinary bladder, in particular transitional cell carcinoma and papillary carcinoma, kidney cancer, in particular renal cell carcinoma including clear cell renal cell carcinoma and papillary renal cell carcinoma, colon cancer, small bowel cancer, including cancer of the ileum, in particular small bowel adenocarcinoma and adenocarcinoma of the ileum, testicular embryonal carcinoma, placental choriocarcinoma, cervical cancer, testicular cancer, in particular testicular seminoma, testicular teratoma and embryonic testicular cancer, uterine cancer, a germ cell tumor such as a teratocarcinoma or an embryonal carcinoma, in particular a germ cell tumor of the testis, and the metastatic forms thereof. In one embodiment, the cancer disease associated with CLDN6 expression is selected from the group consisting of ovarian cancer, lung cancer, metastatic ovarian cancer and metastatic lung cancer. Preferably, the ovarian cancer is a carcinoma or an adenocarcinoma. Preferably, the lung cancer is a carcinoma or an adenocarcinoma, and preferably is bronchiolar cancer such as a bronchiolar carcinoma or bronchiolar adenocarcinoma. In one embodiment, the tumor cell associated with CLDN6 expression is a cell of such a cancer. The term “portion” refers to a fraction. With respect to a particular structure such as an amino acid sequence or protein the term “portion” thereof may designate a continuous or a discontinuous fraction of said structure. Preferably, a portion of an amino acid sequence comprises at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, preferably at least 40%, preferably at least 50%, more preferably at least 60%, more preferably at least 70%, even more preferably at least 80%, and most preferably at least 90% of the amino acids of said amino acid sequence. Preferably, if the portion is a discontinuous fraction said discontinuous fraction is composed of 2, 3, 4, 5, 6, 7, 8, or more parts of a structure, each part being a continuous element of the structure. For example, a discontinuous fraction of an amino acid sequence may be composed of 2, 3, 4, 5, 6, 7, 8, or more, preferably not more than 4 parts of said amino acid sequence, wherein each part preferably comprises at least 5 continuous amino acids, at least 10 continuous amino acids, preferably at least 20 continuous amino acids, preferably at least 30 continuous amino acids of the amino acid sequence. The terms “part” and “fragment” are used interchangeably herein and refer to a continuous element. For example, a part of a structure such as an amino acid sequence or protein refers to a continuous element of said structure. A portion, a part or a fragment of a structure preferably comprises one or more functional properties of said structure. For example, a portion, a part or a fragment of an epitope or peptide is preferably immunologically equivalent to the epitope or peptide it is derived from. The term “an extracellular portion of a CLDN” in the context of the present invention refers to a part of a CLDN facing the extracellular space of a cell and preferably being accessible from the outside of said cell, e.g., by antibodies located outside the cell. Preferably, the term refers to one or more extracellular loops or a part thereof or any other extracellular part of a CLDN which is preferably specific for said CLDN. Preferably, said part comprises at least 5, at least 8, at least 10, at least 15, at least 20, at least 30, or at least 50 amino acids or more. The term “CLDN associated with the surface of a cell” is to be understood to relate to native CLDN, i.e. CLDN in its non-denatured, preferably naturally folded state. Preferably, the term “CLDN associated with the surface of a cell” means that the CLDN is associated with and located at the plasma membrane of said cell, wherein at least a part of the CLDN, preferably the extracellular portion, faces the extracellular space of said cell and is accessible from the outside of said cell, e.g., by antibodies located outside the cell. The association may be direct or indirect. For example, the association may be by one or more transmembrane domains, one or more lipid anchors, and/or by the interaction with any other protein, lipid, saccharide, or other structure that can be found on the outer leaflet of the plasma membrane of a cell. For example, a CLDN associated with the surface of a cell may be a transmembrane protein, i.e. an integral membrane protein, having an extracellular portion or may be a protein associated with the surface of a cell by interacting with another protein that is a transmembrane protein. CLDN6 is associated with the surface of a cell if it is located at the surface of said cell and is accessible to binding by CLDN6-specific antibodies added to the cell. In preferred embodiments, a cell being characterized by association of CLDN6 with its cell surface is a cell expressing CLDN6. It is to be understood that in the case where CLDN6 is expressed by cells, the CLDN6 associated with the surface of said cells may only be a portion of the expressed CLDN6. The term “a cell carrying a CLDN” preferably means that said cell carries a CLDN on its surface, i.e., that the CLDN is associated with the surface of said cell. “Cell surface” or “surface of a cell” is used in accordance with its normal meaning in the art, and thus includes the outside of the cell which is accessible to binding by proteins and other molecules. The expression “CLDN expressed on the surface of a cell” means that the CLDN expressed by a cell is found in association with the surface of said cell. According to the invention CLDN6 is not substantially expressed in a cell and is not substantially associated with a cell surface if the level of expression and association is lower compared to expression and association in placenta cells or placenta tissue. Preferably, the level of expression and association is less than 10%, preferably less than 5%, 3%, 2%, 1%, 0.5%, 0.1% or 0.05% of the expression and association in placenta cells or placenta tissue or even lower. Preferably, CLDN6 is not substantially expressed in a cell and is not substantially associated with a cell surface if the level of expression and association exceeds the level of expression and association in non-tumorigenic, non-cancerous tissue other than placenta tissue by no more than 2-fold, preferably 1.5-fold, and preferably does not exceed the level of expression and association in said non-tumorigenic, non-cancerous tissue. Preferably, CLDN6 is not substantially expressed in a cell and is not substantially associated with a cell surface if the level of expression or association is below the detection limit and/or if the level of expression or association is too low to allow binding by CLDN6-specific antibodies added to the cells. According to the invention CLDN6 is expressed in a cell and is associated with a cell surface if the level of expression and association exceeds the level of expression and association in non-tumorigenic, non-cancerous tissue other than placenta tissue, preferably by more than 2-fold, preferably 10-fold, 100-fold, 1000-fold, or 10000-fold. Preferably, CLDN6 is expressed in a cell and is associated with a cell surface if the level of expression and association is above the detection limit and/or if the level of expression and association is high enough to allow binding by CLDN6-specific antibodies added to the cells. Preferably, CLDN6 expressed in a cell is expressed or exposed on the surface of said cell. The term “raft” refers to the sphingolipid- and cholesterol-rich membrane microdomains located in the outer leaflet area of the plasma membrane of a cell. The ability of certain proteins to associate within such domains and their ability of forming “aggregates” or “focal aggregates” can effect the protein's function. For example, the translocation of CLDN6 molecules into such structures, after being bound by antibodies of the present invention, creates a high density of CLDN6 antigen-antibody complexes in the plasma membranes. Such a high density of CLDN6 antigen-antibody complexes can enable efficient activation of the complement system during CDC. According to the invention, the term “disease” refers to any pathological state, including cancer, in particular those forms of cancer described herein. “Diseases involving cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface” means according to the invention that expression and association in cells of a diseased tissue or organ is preferably increased compared to the state in a healthy tissue or organ. An increase refers to an increase by at least 10%, in particular at least 20%, at least 50%, at least 100%, at least 200%, at least 500%, at least 1000%, at least 10000% or even more. In one embodiment, expression and association with the cell surface is only found in a diseased tissue, while expression in a healthy tissue is repressed. According to the invention, diseases associated with cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface include tumor diseases such as cancer diseases. Furthermore, according to the invention, tumor diseases such as cancer diseases preferably are those wherein the tumor cells or cancer cells express CLDN6 and are characterized by association of CLDN6 with their cell surface. As used herein, a “tumor disease”, “tumor-related disease” or “tumorigenic disease” includes a disease characterized by aberrantly regulated cellular growth, proliferation, differentiation, adhesion, and/or migration, which may result in the production of or tendency to produce tumors and/or tumor metastasis. By “tumor cell” is meant an abnormal cell that grows by a rapid, uncontrolled cellular proliferation and continues to grow after the stimuli that initiated the new growth cease. By “tumor” is meant an abnormal group of cells or a tissue growing by a rapid, uncontrolled cellular proliferation and continues to grow after the stimuli that initiated the new growth cease. Tumors show partial or complete lack of structural organization and functional coordination with the normal tissue, and usually form a distinct mass of tissue, which may be either benign, pre-malignant or malignant. Preferably, a “tumor disease”, “tumor-related disease” or “tumorigenic disease” according to the invention is a cancer disease, i.e. a malignant disease and a tumor cell is a cancer cell. Preferably, a “tumor disease”, “tumor-related disease” or “tumorigenic disease” is characterized by cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface and a tumor cell expresses CLDN6 and is characterized by association of CLDN6 with its cell surface. A cell expressing CLDN6 and being characterized by association of CLDN6 with its cell surface preferably is a tumor cell or cancer cell, preferably of the tumors and cancers described herein. Preferably, such cell is a cell other than a placental cell. Preferred cancer diseases or cancers according to the invention are selected from the group consisting of ovarian cancer, in particular ovarian adenocarcinoma and ovarian teratocarcinoma, lung cancer, including small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), in particular squamous cell lung carcinoma and adenocarcinoma, gastric cancer, breast cancer, hepatic cancer, pancreatic cancer, skin cancer, in particular basal cell carcinoma and squamous cell carcinoma, malignant melanoma, head and neck cancer, in particular malignant pleomorphic adenoma, sarcoma, in particular synovial sarcoma and carcinosarcoma, bile duct cancer, cancer of the urinary bladder, in particular transitional cell carcinoma and papillary carcinoma, kidney cancer, in particular renal cell carcinoma including clear cell renal cell carcinoma and papillary renal cell carcinoma, colon cancer, small bowel cancer, including cancer of the ileum, in particular small bowel adenocarcinoma and adenocarcinoma of the ileum, testicular embryonal carcinoma, placental choriocarcinoma, cervical cancer, testicular cancer, in particular testicular seminoma, testicular teratoma and embryonic testicular cancer, uterine cancer, a germ cell tumor such as a teratocarcinoma or an embryonal carcinoma, in particular a germ cell tumor of the testis, and the metastatic forms thereof. The main types of lung cancer are small cell lung carcinoma (SCLC) and non-small cell lung carcinoma (NSCLC). There are three main sub-types of the non-small cell lung carcinomas: squamous cell lung carcinoma, adenocarcinoma, and large cell lung carcinoma. Adenocarcinomas account for approximately 10% of lung cancers. This cancer usually is seen peripherally in the lungs, as opposed to small cell lung cancer and squamous cell lung cancer, which both tend to be more centrally located. Skin cancer is a malignant growth on the skin. The most common skin cancers are basal cell cancer, squamous cell cancer, and melanoma. Malignant melanoma is a serious type of skin cancer. It is due to uncontrolled growth of pigment cells, called melanocytes. According to the invention, a “carcinoma” is a cancer that begins in the lining layer (epithelial cells) of organs. “Bronchiolar carcinoma” is a carcinoma of the lung, thought to be derived from epithelium of terminal bronchioles, in which the neoplastic tissue extends along the alveolar walls and grows in small masses within the alveoli. Mucin may be demonstrated in some of the cells and in the material in the alveoli, which also includes denuded cells. “Adenocarcinoma” is a cancer that originates in glandular tissue. This tissue is also part of a larger tissue category known as epithelial tissue. Epithelial tissue includes skin, glands and a variety of other tissue that lines the cavities and organs of the body. Epithelium is derived embryologically from ectoderm, endoderm and mesoderm. To be classified as adenocarcinoma, the cells do not necessarily need to be part of a gland, as long as they have secretory properties. This form of carcinoma can occur in some higher mammals, including humans. Well differentiated adenocarcinomas tend to resemble the glandular tissue that they are derived from, while poorly differentiated may not. By staining the cells from a biopsy, a pathologist will determine whether the tumor is an adenocarcinoma or some other type of cancer. Adenocarcinomas can arise in many tissues of the body due to the ubiquitous nature of glands within the body. While each gland may not be secreting the same substance, as long as there is an exocrine function to the cell, it is considered glandular and its malignant form is therefore named adenocarcinoma. Malignant adenocarcinomas invade other tissues and often metastasize given enough time to do so. Ovarian adenocarcinoma is the most common type of ovarian carcinoma. It includes the serous and mucinous adenocarcinomas, the clear cell adenocarcinoma and the endometrioid adenocarcinoma. “Cystadenocarcinoma” is a malignant form of a surface epithelial-stromal tumor, a type of ovarian cancer. Surface epithelial-stromal tumors are a class of ovarian neoplasms that are thought to be derived from the ovarian surface epithelium (modified peritoneum) or from ectopic endometrial or Fallopian tube (tubal) tissue. This group of tumors accounts for the majority of all ovarian tumors. Teratocarcinoma refers to a germ cell tumor that is a mixture of teratoma with embryonal carcinoma, or with choriocarcinoma, or with both. Choriocarcinoma is a malignant, trophoblastic and aggressive cancer, usually of the placenta. It is characterized by early hematogenous spread to the lungs. A sarcoma is a cancer of the connective tissue (bone, cartilage, fat) resulting in mesoderm proliferation. This is in contrast to carcinomas, which are of epithelial origin. A synovial sarcoma is a rare form of cancer which usually occurs near to the joints of the arm or leg. It is one of the soft tissue sarcomas. Renal cell carcinoma also known as renal cell cancer or renal cell adenocarcinoma is a kidney cancer that originates in the lining of the proximal convoluted tubule, the very small tubes in the kidney that filter the blood and remove waste products. Renal cell carcinoma is by far the most common type of kidney cancer in adults and the most lethal of all the genitorurinary tumors. Distinct subtypes of renal cell carcinoma are clear cell renal cell carcinoma and papillary renal cell carcinoma. Clear cell renal cell carcinoma is the most common form of renal cell carcinoma. When seen under a microscope, the cells that make up clear cell renal cell carcinoma appear very pale or clear. Papillary renal cell carcinoma is the second most common subtype. These cancers form little finger-like projections (called papillae) in some, if not most, of the tumors. A germ cell tumor is a neoplasm derived from germ cells. Germ cell tumors can be cancerous or non-cancerous tumors. Germ cells normally occur inside the gonads (ovary and testis). Germ cell tumors that originate outside the gonads (e.g. in head, inside the mouth, neck, pelvis; in fetuses, babies, and young children most often found on the body midline, particularly at the tip of the tailbone) may be birth defects resulting from errors during development of the embryo. The two major classes of germ cell tumors are the seminomas and non-seminomas, wherein non-seminomas include: teratocarcinoma, embryonal carcinoma, yolk sac tumors, choriocarcinoma and differentiated teratoma. Most cell lines from non-seminomas are equivalent to embryonal carcinomas, that is, they are composed almost entirely of stem cells which do not differentiate under basal conditions, though some may respond to inducers of differentiation such as retinoic acid. By “metastasis” is meant the spread of cancer cells from its original site to another part of the body. The formation of metastasis is a very complex process and depends on detachment of malignant cells from the primary tumor, invasion of the extracellular matrix, penetration of the endothelial basement membranes to enter the body cavity and vessels, and then, after being transported by the blood, infiltration of target organs. Finally, the growth of a new tumor at the target site depends on angiogenesis. Tumor metastasis often occurs even after the removal of the primary tumor because tumor cells or components may remain and develop metastatic potential. In one embodiment, the term “metastasis” according to the invention relates to “distant metastasis” which relates to a metastasis which is remote from the primary tumor and the regional lymph node system. The cells of a secondary or metastatic tumor are like those in the original tumor. This means, for example, that, if ovarian cancer metastasizes to the liver, the secondary tumor is made up of abnormal ovarian cells, not of abnormal liver cells. The tumor in the liver is then called metastatic ovarian cancer, not liver cancer. By “treat” is meant to administer a compound or composition as described herein to a subject in order to prevent or eliminate a disease, including reducing the size of a tumor or the number of tumors in a subject; arrest or slow a disease in a subject; inhibit or slow the development of a new disease in a subject; decrease the frequency or severity of symptoms and/or recurrences in a subject who currently has or who previously has had a disease; and/or prolong, i.e. increase the lifespan of the subject. The term “treatment of a disease” includes curing, shortening the duration, ameliorating, preventing, slowing down or inhibiting progression or worsening, or preventing or delaying the onset of a disease or the symptoms thereof. By “being at risk” is meant a subject, i.e. a patient, that is identified as having a higher than normal chance of developing a disease, in particular cancer, compared to the general population. In addition, a subject who has had, or who currently has, a disease, in particular cancer is a subject who has an increased risk for developing a disease, as such a subject may continue to develop a disease. Subjects who currently have, or who have had, a cancer also have an increased risk for cancer metastases. The term “immunotherapy” relates to a treatment involving a specific immune reaction. In the context of the present invention, terms such as “protect”, “prevent”, “prophylactic”, “preventive”, or “protective” relate to the prevention or treatment or both of the occurrence and/or the propagation of a tumor in an individual. The term “immunotherapy” in the context of the present invention preferably refers to active tumor immunization or tumor vaccination. A prophylactic administration of an immunotherapy, for example, a prophylactic administration of the composition of the invention, preferably protects the recipient from the development of tumor growth. A therapeutic administration of an immunotherapy, for example, a therapeutic administration of the composition of the invention, may lead to the inhibition of the progress/growth of the tumor. This comprises the deceleration of the progress/growth of the tumor, in particular a disruption of the progression of the tumor, which preferably leads to elimination of the tumor. A therapeutic administration of an immunotherapy may protect the individual, for example, from the dissemination or metastasis of existing tumors. The term “immunization” or “vaccination” describes the process of administering antigen to a subject with the purpose of inducing an immune response for therapeutic or prophylactic reasons. The terms “subject”, “individual”, “organism” or “patient” are used interchangeably and relate to vertebrates, preferably mammals. For example, mammals in the context of the present invention are humans, non-human primates, domesticated animals such as dogs, cats, sheep, cattle, goats, pigs, horses etc., laboratory animals such as mice, rats, rabbits, guinea pigs, etc. as well as animals in captivity such as animals of zoos. The term “animal” as used herein also includes humans. The term “subject” may also include a patient, i.e., an animal, preferably a human having a disease, preferably a disease associated with expression of CLDN6, preferably a tumorigenic disease such as a cancer. The term “adjuvant” relates to compounds which prolongs or enhances or accelerates an immune response. The composition of the present invention preferably exerts its effect without addition of adjuvants. Still, the composition of the present application may contain any known adjuvant. Adjuvants comprise a heterogeneous group of compounds such as oil emulsions (e.g., Freund's adjuvants), mineral compounds (such as alum), bacterial products (such asBordetella pertussistoxin), liposomes, and immune-stimulating complexes. Examples for adjuvants are monophosphoryl-lipid-A (MPL SmithKline Beecham). Saponins such as QS21 (SmithKline Beecham), DQS21 (SmithKline Beecham; WO 96/33739), QS7, QS17, QS18, and QS-L1 (So et al., 1997, Mol. Cells 7: 178-186), incomplete Freund's adjuvants, complete Freund's adjuvants, vitamin E, montanid, alum, CpG oligonucleotides (Krieg et al., 1995, Nature 374: 546-549), and various water-in-oil emulsions which are prepared from biologically degradable oils such as squalene and/or tocopherol. According to the invention, a sample may be any sample useful according to the present invention, in particular a biological sample such a tissue sample, including bodily fluids, and/or a cellular sample and may be obtained in the conventional manner such as by tissue biopsy, including punch biopsy, and by taking blood, bronchial aspirate, sputum, urine, feces or other body fluids. According to the invention, the term “biological sample” also includes fractions of biological samples. The term “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, and includes any molecule comprising an antigen binding portion thereof. The term “antibody” includes monoclonal antibodies and fragments or derivatives thereof, including, without limitation, human monoclonal antibodies, humanized monoclonal antibodies, chimeric monoclonal antibodies, single chain antibodies, e.g., scFv's and antigen-binding antibody fragments such as Fab and Fab′ fragments and also includes all recombinant forms of antibodies, e.g., antibodies expressed in prokaryotes, unglycosylated antibodies, and any antigen-binding antibody fragments and derivatives as described herein. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system. According to the invention, the term “at least one of the CDR sequences” preferably means at least the CDR3 sequence. The term “CDR sequences of an antibody chain” preferably relates to CDR1, CDR2 and CDR3 of the heavy chain or light chain of an antibody. According to the invention, a reference to an antibody chain comprising a particular CDR sequence such as a particular CDR3 sequence means that said particular CDR sequence either forms the CDR region such as the CDR3 region of said antibody chain, i.e. the CDR region consists of said particular CDR sequence, or forms a part of the CDR region such as the CDR3 region of said antibody chain, i.e. the CDR region comprises said particular CDR sequence. If according to the invention reference is made to an antibody comprising a particular antibody heavy chain and/or a particular antibody light chain, such as a chain comprising particular CDR sequences, it is preferred that both heavy chains and/or both light chains of the antibody are each composed of the particular antibody heavy chain and/or the particular antibody light chain. The term “humanized antibody” refers to a molecule having an antigen binding site that is substantially derived from an immunoglobulin from a non-human species, wherein the remaining immunoglobulin structure of the molecule is based upon the structure and/or sequence of a human immunoglobulin. The antigen binding site may either comprise complete variable domains fused onto constant domains or only the complementarity determining regions (CDR) grafted onto appropriate framework regions in the variable domains. Antigen binding sites may be wild-type or modified by one or more amino acid substitutions, e.g. modified to resemble human immunoglobulins more closely. Some forms of humanized antibodies preserve all CDR sequences (for example a humanized mouse antibody which contains all six CDRs from the mouse antibody). Other forms have one or more CDRs which are altered with respect to the original antibody. The term “chimeric antibody” refers to those antibodies wherein one portion of each of the amino acid sequences of heavy and light chains is homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular class, while the remaining segment of the chain is homologous to corresponding sequences in another. Typically the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals, while the constant portions are homologous to sequences of antibodies derived from another. One clear advantage to such chimeric forms is that the variable region can conveniently be derived from presently known sources using readily available B-cells or hybridomas from non-human host organisms in combination with constant regions derived from, for example, human cell preparations. While the variable region has the advantage of ease of preparation and the specificity is not affected by the source, the constant region being human, is less likely to elicit an immune response from a human subject when the antibodies are injected than would the constant region from a non human source. However the definition is not limited to this particular example. The term “antigen-binding portion” of an antibody (or simply “binding portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) Fab fragments, monovalent fragments consisting of the VL, VH, CL and CH domains; (ii) F(ab′)2fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) Fd fragments consisting of the VH and CH domains; (iv) Fv fragments consisting of the VL and VH domains of a single arm of an antibody, (v) dAb fragments (Ward et al., (1989) Nature 341: 544-546), which consist of a VH domain; (vi) isolated complementarity determining regions (CDR), and (vii) combinations of two or more isolated CDRs which may optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242: 423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85: 5879-5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. A further example is binding-domain immunoglobulin fusion proteins comprising (i) a binding domain polypeptide that is fused to an immunoglobulin hinge region polypeptide, (ii) an immunoglobulin heavy chain CH2 constant region fused to the hinge region, and (iii) an immunoglobulin heavy chain CH3 constant region fused to the CH2 constant region. The binding domain polypeptide can be a heavy chain variable region or a light chain variable region. The binding-domain immunoglobulin fusion proteins are further disclosed in US 2003/0118592 and US 2003/0133939. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. The term “epitope” refers to an antigenic determinant in a molecule, i.e., to the part in a molecule that is recognized by the immune system, for example, that is recognized by an antibody. For example, epitopes are the discrete, three-dimensional sites on an antigen, which are recognized by the immune system. In the context of the present invention, the epitope is preferably derived from a CLDN protein. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents. An epitope of a protein such as a CLDN preferably comprises a continuous or discontinuous portion of said protein and is preferably between 5 and 100, preferably between 5 and 50, more preferably between 8 and 30, most preferably between 10 and 25 amino acids in length, for example, the epitope may be preferably 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acids in length. The term “discontinuous epitope” as used herein, means a conformational epitope on a protein antigen which is formed from at least two separate regions in the primary sequence of the protein. The term “bispecific molecule” is intended to include any agent, e.g., a protein, peptide, or protein or peptide complex, which has two different binding specificities. For example, the molecule may bind to, or interact with (a) a cell surface antigen, and (b) an Fc receptor on the surface of an effector cell. The term “multispecific molecule” or “heterospecific molecule” is intended to include any agent, e.g., a protein, peptide, or protein or peptide complex, which has more than two different binding specificities. For example, the molecule may bind to, or interact with (a) a cell surface antigen, (b) an Fc receptor on the surface of an effector cell, and (c) at least one other component. Accordingly, the invention includes, but is not limited to, bispecific, trispecific, tetraspecific, and other multispecific molecules which are directed to CLDN6, and to other targets, such as Fc receptors on effector cells. The term “bispecific antibodies” also includes diabodies. Diabodies are bivalent, bispecific antibodies in which the VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90: 6444-6448; Poljak, R. J., et al. (1994) Structure 2: 1121-1123). As used herein, the term “heteroantibodies” refers to two or more antibodies, derivatives thereof, or antigen binding regions linked together, at least two of which have different specificities. These different specificities include a binding specificity for an Fc receptor on an effector cell, and a binding specificity for an antigen or epitope on a target cell, e.g., a tumor cell. The antibodies described herein may be human antibodies. The term “human antibody”, as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). The term “monoclonal antibody” as used herein refers to a preparation of antibody molecules of single molecular composition. A monoclonal antibody displays a single binding specificity and affinity for a particular epitope. In one embodiment, the monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a non-human animal, e.g., mouse, fused to an immortalized cell. The term “recombinant antibody”, as used herein, includes all antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal with respect to the immunoglobulin genes or a hybridoma prepared therefrom, (b) antibodies isolated from a host cell transformed to express the antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences. The term “transfectoma”, as used herein, includes recombinant eukaryotic host cells expressing an antibody, such as CHO cells, NS/0 cells, HEK293 cells, HEK293T cells, plant cells, or fungi, including yeast cells. As used herein, a “heterologous antibody” is defined in relation to a transgenic organism producing such an antibody. This term refers to an antibody having an amino acid sequence or an encoding nucleic acid sequence corresponding to that found in an organism not consisting of the transgenic organism, and being generally derived from a species other than the transgenic organism. As used herein, a “heterohybrid antibody” refers to an antibody having light and heavy chains of different organismal origins. For example, an antibody having a human heavy chain associated with a murine light chain is a heterohybrid antibody. The invention includes all antibodies and derivatives of antibodies as described herein which for the purposes of the invention are encompassed by the term “antibody”. The term “antibody derivatives” refers to any modified form of an antibody, e.g., a conjugate of the antibody and another agent or antibody, or an antibody fragment. The antibodies described herein are preferably isolated. An “isolated antibody” as used herein, is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds to CLDN6 is substantially free of antibodies that specifically bind antigens other than CLDN6). An isolated antibody that specifically binds to an epitope, isoform or variant of human CLDN6 may, however, have cross-reactivity to other related antigens, e.g., from other species (e.g., CLDN6 species homologs). Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals. In one embodiment of the invention, a combination of “isolated” monoclonal antibodies relates to antibodies having different specificities and being combined in a well defined composition. According to the present invention, an antibody is capable of binding to a predetermined target if it has a significant affinity for said predetermined target and binds to said predetermined target in standard assays such as the assays described herein. Preferably, an antibody is capable of binding to a target if it detectably binds to said target in a flow cytometry analysis (FACS analysis) wherein binding of said antibody to said target expressed on the surface of intact cells is determined. Preferably, the antibody detectably binds to said target if present in a concentration of 10 μg/ml or lower, 5 μg/ml or lower or 2 μg/ml or lower. Preferably, the antibody detectably binds to said target if present in a concentration of 50 nM or lower, 30 nM or lower or 15 nM or lower. “Affinity” or “binding affinity” is often measured by equilibrium dissociation constant (KD). Preferably, the term “significant affinity” refers to the binding to a predetermined target with a dissociation constant (KD) of 10−5M or lower, 10−6M or lower, 10−7M or lower, 10−8M or lower, 10−9M or lower, 10−10M or lower, 10−11M or lower, or 10−12M or lower. Antibodies of the present invention preferably have EC50 values for binding to CLDN6 of 6500 ng/ml or lower, 3000 ng/ml or lower, 2500 ng/ml or lower, 2000 ng/ml or lower, 1500 ng/ml or lower, 1000 ng/ml or lower, 500 ng/ml or lower, 400 ng/ml or lower, 300 ng/ml or lower, 200 ng/ml or lower, or 100 ng/ml or lower. An antibody is not (substantially) capable of binding to a target if it has no significant affinity for said target and does not bind significantly to said target in standard assays. Preferably, an antibody is not (substantially) capable of binding to a target if it does not detectably bind to said target in a flow cytometry analysis (FACS analysis) wherein binding of said antibody to said target expressed on the surface of intact cells is determined. Preferably, the antibody does not detectably bind to said target if present in a concentration of up to 2 μg/ml, preferably up to 5 μg/ml, preferably up to 10 μg/ml, preferably up to 20 μg/ml, more preferably up to 50 μg/ml, in particular up to 100 μg/ml, or up to 150 μg/ml, up to 200 μg/ml or higher. Preferably, the antibody does not detectably bind to said target if present in a concentration of up to 15 nM, preferably up to 30 nM, preferably up to 50 nM, preferably up to 100 nM, preferably up to 150 nM, or up to 170 nM, up to 300 mM, up to 600 nM, up to 1000 nM, up to 1300 nM or higher. Preferably, the antibody does not detectably bind to said target if present in a concentration that saturates binding to the target to which the antibody binds, i.e. CLDN6. Preferably, an antibody has no significant affinity for a target if it binds to said target with a KDthat is at least 10-fold, 100-fold, 103-fold, 104-fold, 105-fold, or 106-fold higher than the KDfor binding to the predetermined target to which the antibody is capable of binding. For example, if the KDfor binding of an antibody to the target to which the antibody is capable of binding is 10−7M, the KDfor binding to a target for which the antibody has no significant affinity would be is at least 10−6M, 10−5M, 10−4M, 10−1M, 10−2M, or 10−1M. An antibody is specific for a predetermined target if it is capable of binding to said predetermined target while it is not capable of binding to other targets, i.e. has no significant affinity for other targets and does not significantly bind to other targets in standard assays. According to the invention, an antibody is specific for CLDN6 if it is capable of binding to CLDN6 but is not capable of binding to other targets, in particular claudin proteins other than CLDN6 such as CLDN9, CLDN4, CLDN3 and CLDN1. Preferably, an antibody is specific for CLDN6 if the affinity for and the binding to a claudin protein other than CLDN6 such as CLDN9, CLDN4, CLDN3 and CLDN1 does not significantly exceed the affinity for or binding to claudin-unrelated proteins such as bovine serum albumin (BSA), casein, human serum albumin (HSA) or non-claudin transmembrane proteins such as MHC molecules or transferrin receptor or any other specified polypeptide. Preferably, an antibody is specific for a predetermined target if it binds to said target with a KDthat is at least 10-fold, 100-fold, 103-fold, 104-fold, 105-fold, or 106-fold lower than the KDfor binding to a target for which it is not specific. For example, if the KDfor binding of an antibody to the target for which it is specific is 10−7M, the KDfor binding to a target for which it is not specific would be at least 10−6M, 10−5M, 10−4M, 10−3M, 10−2M, or 10−1M. Binding of an antibody to a target can be determined experimentally using any suitable method; see, for example, Berzofsky et al., “Antibody-Antigen Interactions” In Fundamental Immunology, Paul, W. E., Ed., Raven Press New York, N Y (1984), Kuby, Janis Immunology, W. H. Freeman and Company New York, N Y (1992), and methods described herein. Affinities may be readily determined using conventional techniques, such as by equilibrium dialysis; by using the BIAcore 2000 instrument, using general procedures outlined by the manufacturer; by radioimmunoassay using radiolabeled target antigen; or by another method known to the skilled artisan. The affinity data may be analyzed, for example, by the method of Scatchard et al., Ann N.Y. Acad. ScL, 51:660 (1949). The measured affinity of a particular antibody-antigen interaction can vary if measured under different conditions, e.g., salt concentration, pH. Thus, measurements of affinity and other antigen-binding parameters, e.g., KD, IC50, are preferably made with standardized solutions of antibody and antigen, and a standardized buffer. A unique feature of the antibody of the present invention is the ability to bind cell surface claudin 6. This is demonstrated by flow cytometry analysis of cells expressing claudin 6. To test the binding of monoclonal antibodies to live cells expressing claudins, flow cytometry can be used. Briefly, cell lines expressing membrane-associated claudins (grown under standard growth conditions) are mixed with various concentrations of antibodies in PBS containing 2% heat inactivated FCS and 0.1% NaN3at 4° C. for 30 min. After washing, the cells are reacted with a fluorescently labeled secondary antibody under the same conditions as the primary antibody staining. The samples can be analyzed by FACS using light and side scatter properties to gate on single cells and binding of the labeled antibodies is determined. The term “binding” according to the invention preferably relates to a specific binding as defined herein. As used herein, “isotype” refers to the antibody class (e.g., IgM or IgG1) that is encoded by heavy chain constant region genes. As used herein, “isotype switching” refers to the phenomenon by which the class, or isotype, of an antibody changes from one Ig class to one of the other Ig classes. The term “naturally occurring” as used herein as applied to an object refers to the fact that an object can be found in nature. For example, a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally occurring. The term “rearranged” as used herein refers to a configuration of a heavy chain or light chain immunoglobulin locus wherein a V segment is positioned immediately adjacent to a D-J or J segment in a conformation encoding essentially a complete VH or VL domain, respectively. A rearranged immunoglobulin (antibody) gene locus can be identified by comparison to germline DNA; a rearranged locus will have at least one recombined heptamer/nonamer homology element. The term “unrearranged” or “germline configuration” as used herein in reference to a V segment refers to the configuration wherein the V segment is not recombined so as to be immediately adjacent to a D or J segment. The term “nucleic acid molecule”, as used herein, is intended to include DNA molecules and RNA molecules. A nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA. A nucleic acid molecule can be employed for introduction into, i.e. transfection of, cells, for example, in the form of RNA which can be prepared by in vitro transcription from a DNA template. The RNA can moreover be modified before application by stabilizing sequences, capping, and polyadenylation. The nucleic acids described according to the invention have preferably been isolated. The term “isolated nucleic acid” means according to the invention that the nucleic acid was (i) amplified in vitro, for example by polymerase chain reaction (PCR), (ii) recombinantly produced by cloning, (iii) purified, for example by cleavage and gel-electrophoretic fractionation, or (iv) synthesized, for example by chemical synthesis. An isolated nucleic acid is a nucleic acid which is available for manipulation by recombinant DNA techniques. Nucleic acids may, according to the invention, be present alone or in combination with other nucleic acids, which may be homologous or heterologous. In preferred embodiments, a nucleic acid is functionally linked to expression control sequences which may be homologous or heterologous with respect to said nucleic acid wherein the term “homologous” means that the nucleic acid is also functionally linked to the expression control sequence naturally and the term “heterologous” means that the nucleic acid is not functionally linked to the expression control sequence naturally. A nucleic acid, such as a nucleic acid expressing RNA and/or protein or peptide, and an expression control sequence are “functionally” linked to one another, if they are covalently linked to one another in such a way that expression or transcription of said nucleic acid is under the control or under the influence of said expression control sequence. If the nucleic acid is to be translated into a functional protein, then, with an expression control sequence functionally linked to a coding sequence, induction of said expression control sequence results in transcription of said nucleic acid, without causing a frame shift in the coding sequence or said coding sequence not being capable of being translated into the desired protein or peptide. The term “expression control sequence” comprises according to the invention promoters, ribosome binding sites, enhancers and other control elements which regulate transcription of a gene or translation of a mRNA. In particular embodiments of the invention, the expression control sequences can be regulated. The exact structure of expression control sequences may vary as a function of the species or cell type, but generally comprises 5′-untranscribed and 5′- and 3′-untranslated sequences which are involved in initiation of transcription and translation, respectively, such as TATA box, capping sequence, CAAT sequence, and the like. More specifically, 5′-untranscribed expression control sequences comprise a promoter region which includes a promoter sequence for transcriptional control of the functionally linked nucleic acid. Expression control sequences may also comprise enhancer sequences or upstream activator sequences. According to the invention the term “promoter” or “promoter region” relates to a nucleic acid sequence which is located upstream (5′) to the nucleic acid sequence being expressed and controls expression of the sequence by providing a recognition and binding site for RNA-polymerase. The “promoter region” may include further recognition and binding sites for further factors which are involved in the regulation of transcription of a gene. A promoter may control the transcription of a prokaryotic or eukaryotic gene. Furthermore, a promoter may be “inducible” and may initiate transcription in response to an inducing agent or may be “constitutive” if transcription is not controlled by an inducing agent. A gene which is under the control of an inducible promoter is not expressed or only expressed to a small extent if an inducing agent is absent. In the presence of the inducing agent the gene is switched on or the level of transcription is increased. This is mediated, in general, by binding of a specific transcription factor. Promoters which are preferred according to the invention include promoters for SP6, T3 and T7 polymerase, human U6 RNA promoter, CMV promoter, and artificial hybrid promoters thereof (e.g. CMV) where a part or parts are fused to a part or parts of promoters of genes of other cellular proteins such as e.g. human GAPDH (glyceraldehyde-3-phosphate dehydrogenase), and including or not including (an) additional intron(s). According to the invention, the term “expression” is used in its most general meaning and comprises the production of RNA or of RNA and protein/peptide. It also comprises partial expression of nucleic acids. Furthermore, expression may be carried out transiently or stably. According to the invention, the term expression also includes an “aberrant expression” or “abnormal expression”. “Aberrant expression” or “abnormal expression” means according to the invention that expression is altered, preferably increased, compared to a reference, preferably compared to the state in a non-tumorigenic normal cell or a healthy individual. An increase in expression refers to an increase by at least 10%, in particular at least 20%, at least 50% or at least 100%. In one embodiment, expression is only found in a diseased tissue, while expression in a healthy tissue is repressed. In a preferred embodiment, a nucleic acid molecule is according to the invention present in a vector, where appropriate with a promoter, which controls expression of the nucleic acid. The term “vector” is used here in its most general meaning and comprises any intermediary vehicle for a nucleic acid which enables said nucleic acid, for example, to be introduced into prokaryotic and/or eukaryotic cells and, where appropriate, to be integrated into a genome. Vectors of this kind are preferably replicated and/or expressed in the cells. Vectors comprise plasmids, phagemids, bacteriophages or viral genomes. The term “plasmid” as used herein generally relates to a construct of extrachromosomal genetic material, usually a circular DNA duplex, which can replicate independently of chromosomal DNA. As the vector for expression of an antibody, either of a vector type in which the antibody heavy chain and light chain are present in different vectors or a vector type in which the heavy chain and light chain are present in the same vector can be used. The teaching given herein with respect to specific nucleic acid and amino acid sequences, e.g. those shown in the sequence listing, is to be construed so as to also relate to modifications, i.e. variants, of said specific sequences resulting in sequences which are functionally equivalent to said specific sequences, e.g. amino acid sequences exhibiting properties identical or similar to those of the specific amino acid sequences and nucleic acid sequences encoding amino acid sequences exhibiting properties identical or similar to those of the amino acid sequences encoded by the specific nucleic acid sequences. One important property is to retain binding of an antibody to its target or to sustain effector functions of an antibody such as CDC and/or ADCC. Preferably, a sequence modified with respect to a specific sequence, when it replaces the specific sequence in an antibody retains binding of said antibody to the target and preferably functions of said antibody as described herein. Similarly, the teaching given herein with respect to specific antibodies or hybridomas producing specific antibodies is to be construed so as to also relate to antibodies characterized by an amino acid sequence and/or nucleic acid sequence which is modified compared to the amino acid sequence and/or nucleic acid sequence of the specific antibodies but being functionally equivalent. One important property is to retain binding of an antibody to its target or to sustain effector functions of an antibody. Preferably, a sequence modified with respect to a specific sequence, when it replaces the specific sequence in an antibody retains binding of said antibody to the target and preferably functions of said antibody as described herein, e.g. CDC mediated lysis or ADCC mediated lysis. It will be appreciated by those skilled in the art that in particular the sequences of the CDR, hypervariable and variable regions can be modified without losing the ability to bind to a target. For example, CDR regions will be either identical or highly homologous to the regions of antibodies specified herein. By “highly homologous” it is contemplated that from 1 to 5, preferably from 1 to 4, such as 1 to 3 or 1 or 2 substitutions may be made in the CDRs. In addition, the hypervariable and variable regions may be modified so that they show substantial homology with the regions of antibodies specifically disclosed herein. It is to be understood that the specific nucleic acids described herein also include nucleic acids modified for the sake of optimizing the codon usage in a particular host cell or organism. Differences in codon usage among organisms can lead to a variety of problems concerning heterologous gene expression. Codon optimization by changing one or more nucleotides of the original sequence can result in an optimization of the expression of a nucleic acid, in particular in optimization of translation efficacy, in a homologous or heterologous host in which said nucleic acid is to be expressed. According to the invention, a variant, derivative, modified form or fragment of a nucleic acid sequence, amino acid sequence, or peptide preferably has a functional property of the nucleic acid sequence, amino acid sequence, or peptide, respectively, from which it has been derived. Such functional properties comprise the interaction with or binding to other molecules. In one embodiment, a variant, derivative, modified form or fragment of a nucleic acid sequence, amino acid sequence, or peptide is immunologically equivalent to the nucleic acid sequence, amino acid sequence, or peptide, respectively, from which it has been derived. Preferably the degree of identity between a specific nucleic acid sequence and a nucleic acid sequence which is modified with respect to or which is a variant of said specific nucleic acid sequence will be at least 70%, preferably at least 75%, more preferably at least 80%, even more preferably at least 90% or most preferably at least 95%, 96%, 97%, 98% or 99%. Regarding CLDN6 nucleic acid variants, the degree of identity is preferably given for a region of at least about 300, at least about 400, at least about 450, at least about 500, at least about 550, at least about 600 or at least about 630 nucleotides. In preferred embodiments, the degree of identity is given for the entire length of the reference nucleic acid sequence, such as the nucleic acid sequences given in the sequence listing. Preferably, the two sequences are capable of hybridizing and forming a stable duplex with one another, with hybridization preferably being carried out under conditions which allow specific hybridization between polynucleotides (stringent conditions). Stringent conditions are described, for example, in Molecular Cloning: A Laboratory Manual, J. Sambrook et al., Editors, 2nd Edition, Cold Spring Harbor Laboratory press, Cold Spring Harbor, New York, 1989 or Current Protocols in Molecular Biology, F. M. Ausubel et al., Editors, John Wiley & Sons, Inc., New York and refer, for example, to hybridization at 65° C. in hybridization buffer (3.5×SSC, 0.02% Ficoll, 0.02% polyvinylpyrrolidone, 0.02% bovine serum albumin, 2.5 mM NaH2PO4(pH 7), 0.5% SDS, 2 mM EDTA). SSC is 0.15 M sodium chloride/0.15 M sodium citrate, pH 7. After hybridization, the membrane to which the DNA has been transferred is washed, for example, in 2×SSC at room temperature and then in 0.1-0.5×SSC/0.1×SDS at temperatures of up to 68° C. The term “variant” according to the invention also includes mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present. An allelic variant relates to an alteration in the normal sequence of a gene, the significance of which is often unclear. Complete gene sequencing often identifies numerous allelic variants for a given gene. A species homolog is a nucleic acid or amino acid sequence with a different species of origin from that of a given nucleic acid or amino acid sequence. For the purposes of the present invention, “variants” of an amino acid sequence comprise amino acid insertion variants, amino acid addition variants, amino acid deletion variants and/or amino acid substitution variants. Amino acid deletion variants that comprise the deletion at the N-terminal and/or C-terminal end of the protein are also called N-terminal and/or C-terminal truncation variants. Amino acid insertion variants comprise insertions of single or two or more amino acids in a particular amino acid sequence. In the case of amino acid sequence variants having an insertion, one or more amino acid residues are inserted into a particular site in an amino acid sequence, although random insertion with appropriate screening of the resulting product is also possible. Amino acid addition variants comprise amino- and/or carboxy-terminal fusions of one or more amino acids, such as 1, 2, 3, 5, 10, 20, 30, 50, or more amino acids. Amino acid deletion variants are characterized by the removal of one or more amino acids from the sequence, such as by removal of 1, 2, 3, 5, 10, 20, 30, 50, or more amino acids. The deletions may be in any position of the protein. Amino acid substitution variants are characterized by at least one residue in the sequence being removed and another residue being inserted in its place. Preference is given to the modifications being in positions in the amino acid sequence which are not conserved between homologous proteins or peptides and/or to replacing amino acids with other ones having similar properties. Preferably, amino acid changes in protein variants are conservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids. A conservative amino acid change involves substitution of one of a family of amino acids which are related in their side chains. Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate), basic (lysine, arginine, histidine), non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and uncharged polar (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine) amino acids. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids. Preferably the degree of similarity, preferably identity between a specific amino acid sequence and an amino acid sequence which is modified with respect to or which is a variant of said specific amino acid sequence such as between amino acid sequences showing substantial homology will be at least 70%, preferably at least 80%, even more preferably at least 90% or most preferably at least 95%, 96%, 97%, 98% or 99%. The degree of similarity or identity is given preferably for an amino acid region which is at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or about 100% of the entire length of the reference amino acid sequence. For example, if the reference amino acid sequence consists of 200 amino acids, the degree of similarity or identity is given preferably for at least about 20, at least about 40, at least about 60, at least about 80, at least about 100, at least about 120, at least about 140, at least about 160, at least about 180, or about 200 amino acids, preferably continuous amino acids. Regarding CLDN6 polypeptide variants, the degree of similarity or identity is given preferably for a region of at least about 100, at least about 120, at least about 140, at least about 160, at least about 180, at least about 200, or at least about 210 amino acids. In preferred embodiments, the degree of similarity or identity is given for the entire length of the reference amino acid sequence such as the amino acid sequences given in the sequence listing. The alignment for determining sequence similarity, preferably sequence identity can be done with art known tools, preferably using the best sequence alignment, for example, using Align, using standard settings, preferably EMBOSS::needle, Matrix: Blosum62, Gap Open 10.0, Gap Extend 0.5. “Sequence similarity” indicates the percentage of amino acids that either are identical or that represent conservative amino acid substitutions. “Sequence identity” between two polypeptide or nucleic acid sequences indicates the percentage of amino acids or nucleotides that are identical between the sequences. The “percentage identity” is obtained after the best alignment, this percentage being purely statistical and the differences between the two sequences being distributed randomly and over their entire length. Sequence comparisons between two nucleotide or amino acid sequences are conventionally carried out by comparing these sequences after having aligned them optimally, said comparison being carried out by segment or by “window of comparison” in order to identify and compare local regions of sequence similarity. The optimal alignment of the sequences for comparison may be produced, besides manually, by means of the local homology algorithm of Smith and Waterman, 1981, Ads App. Math. 2, 482, by means of the local homology algorithm of Neddleman and Wunsch, 1970, J. Mol. Biol. 48, 443, by means of the similarity search method of Pearson and Lipman, 1988, Proc. Natl Acad. Sci. USA 85, 2444, or by means of computer programs which use these algorithms (GAP, BESTFIT, FASTA, BLAST P, BLAST N and TFASTA in Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Drive, Madison, Wis.). The percentage identity is calculated by determining the number of identical positions between the two sequences being compared, dividing this number by the number of positions compared and multiplying the result obtained by 100 so as to obtain the percentage identity between these two sequences. “Conservative substitutions,” may be made, for instance, on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. For example: (a) nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; (b) polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; (c) positively charged (basic) amino acids include arginine, lysine, and histidine; and (d) negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Substitutions typically may be made within groups (a)-(d). In addition, glycine and proline may be substituted for one another based on their ability to disrupt α-helices. Some preferred substitutions may be made among the following groups: (i) S and T; (ii) P and G; and (iii) A, V, L and I. Given the known genetic code, and recombinant and synthetic DNA techniques, the skilled scientist readily can construct DNAs encoding the conservative amino acid variants. The present invention comprises antibodies in which alterations have been made in the Fc region in order to change the functional or pharmacokinetic properties of the antibodies. Such alterations may result in a decrease or increase of C1q binding and CDC or of FcγR binding and ADCC. Substitutions can, for example, be made in one or more of the amino acid residues of the heavy chain constant region, thereby causing an alteration in an effector function while retaining the ability to bind to the antigen as compared with the modified antibody, cf. U.S. Pat. Nos. 5,624,821 and 5,648,260. The in vivo half-life of antibodies can be improved by modifying the salvage receptor epitope of the Ig constant domain or an Ig-like constant domain such that the molecule does not comprise an intact CH2 domain or an intact Ig Fc region, cf U.S. Pat. Nos. 6,121,022 and 6,194,551. The in vivo half-life can furthermore be increased by making mutations in the Fc region, e.g., by substituting threonine for leucine at position 252, by substituting threonine for serine at position 254, or by substituting threonine for phenylalanine at position 256, cf. U.S. Pat. No. 6,277,375. Furthermore, the glycosylation pattern of antibodies can be modified in order to change the effector function of the antibodies. For example, the antibodies can be expressed in a transfectoma which does not add the fucose unit normally attached to Asn at position 297 of the Fc region in order to enhance the affinity of the Fc region for Fc-Receptors which, in turn, will result in an increased ADCC of the antibodies in the presence of NK cells, cf. Shield et al. (2002) JBC, 277: 26733. Furthermore, modification of galactosylation can be made in order to modify CDC. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of a anti-CLDN6 antibody coding sequence, such as by saturation mutagenesis, and the resulting modified anti-CLDN6 antibodies can be screened for binding activity. According to the invention the term “cell” or “host cell” preferably relates to an intact cell, i.e. a cell with an intact membrane that has not released its normal intracellular components such as enzymes, organelles, or genetic material. An intact cell preferably is a viable cell, i.e. a living cell capable of carrying out its normal metabolic functions. Preferably said term relates according to the invention to any cell which can be transformed or transfected with an exogenous nucleic acid. The term “cell” includes according to the invention prokaryotic cells (e.g.,E. coli) or eukaryotic cells (e.g., dendritic cells, B cells, CHO cells, COS cells, K562 cells, HEK293 cells, HELA cells, yeast cells, and insect cells). The exogenous nucleic acid may be found inside the cell (i) freely dispersed as such, (ii) incorporated in a recombinant vector, or (iii) integrated into the host cell genome or mitochondrial DNA. Mammalian cells are particularly preferred, such as cells from humans, mice, hamsters, pigs, goats, and primates. The cells may be derived from a large number of tissue types and include primary cells and cell lines. Specific examples include keratinocytes, peripheral blood leukocytes, bone marrow stem cells, and embryonic stem cells. In further embodiments, the cell is an antigen-presenting cell, in particular a dendritic cell, a monocyte, or macrophage. The term “host cell”, as used herein, preferably is intended to refer to a cell into which a recombinant expression vector has been introduced. A cell which comprises a nucleic acid molecule preferably express the peptide or protein encoded by the nucleic acid. The terms “transgenic animal” refers to an animal having a genome comprising one or more transgenes, preferably heavy and/or light chain transgenes, or transchromosomes (either integrated or non-integrated into the animal's natural genomic DNA) and which is preferably capable of expressing the transgenes. For example, a transgenic mouse can have a human light chain transgene and either a human heavy chain transgene or human heavy chain transchromosome, such that the mouse produces human anti-CLDN6 antibodies when immunized with CLDN6 antigen and/or cells expressing CLDN6. The human heavy chain transgene can be integrated into the chromosomal DNA of the mouse, as is the case for transgenic mice, e.g., HuMAb mice, such as HCo7 or HCol2 mice, or the human heavy chain transgene can be maintained extrachromosomally, as is the case for transchromosomal (e.g., KM) mice as described in WO 02/43478. Such transgenic and transchromosomal mice may be capable of producing multiple isotypes of human monoclonal antibodies to CLDN6 (e.g., IgG, IgA and/or IgE) by undergoing V-D-J recombination and isotype switching. “Reduce” or “inhibit” as used herein means the ability to cause an overall decrease, preferably of 5% or greater, 10% or greater, 20% or greater, more preferably of 50% or greater, and most preferably of 75% or greater, in the level, e.g. in the level of proliferation of cells. The term “inhibit” or similar phrases includes a complete or essentially complete inhibition, i.e. a reduction to zero or essentially to zero. Terms such as “increasing” or “enhancing” preferably relate to an increase or enhancement by about at least 10%, preferably at least 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 80%, and most preferably at least 100%. These terms may also relate to circumstances, wherein at time zero there is no detectable signal for a certain compound or condition and at a particular time point later than time zero there is a detectable signal for a certain compound or condition. The term “immunologically equivalent” means that the immunologically equivalent molecule such as the immunologically equivalent amino acid sequence exhibits the same or essentially the same immunological properties and/or exerts the same or essentially the same immunological effects, e.g., with respect to the type of the immunological effect such as induction of a humoral and/or cellular immune response, the strength and/or duration of the induced immune reaction, or the specificity of the induced immune reaction. In the context of the present invention, the term “immunologically equivalent” is preferably used with respect to the immunological effects or properties of a peptide or peptide variant used for immunization. A particular immunological property is the ability to bind to antibodies and, where appropriate, generate an immune response, preferably by stimulating the generation of antibodies. For example, an amino acid sequence is immunologically equivalent to a reference amino acid sequence if said amino acid sequence when exposed to the immune system of a subject induces an immune reaction, preferably antibodies, having a specificity of reacting with the reference amino acid sequence, such as the reference amino acid sequence forming part of CLDN6. The term “immune effector functions” in the context of the present invention includes any functions mediated by components of the immune system that result in the inhibition of tumor growth and/or inhibition of tumor development, including inhibition of tumor dissemination and metastasis. Preferably, immune effector functions result in killing of tumor cells. Preferably, the immune effector functions in the context of the present invention are antibody-mediated effector functions. Such functions comprise complement dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), induction of apoptosis in the cells carrying the tumor-associated antigen, for example, by binding of the antibody to a surface antigen, and/or inhibition of proliferation of the cells carrying the tumor-associated antigen, preferably ADCC and/or CDC. Thus, antibodies that are capable of mediating one or more immune effector functions are preferably able to mediate killing of cells by inducing CDC-mediated lysis, ADCC-mediated lysis, apoptosis, homotypic adhesion, and/or phagocytosis, preferably by inducing CDC-mediated lysis and/or ADCC-mediated lysis. Antibodies may also exert an effect simply by binding to tumor-associated antigens on the surface of a tumor cell. For example, antibodies may block the function of the tumor-associated antigen or induce apoptosis just by binding to the tumor-associated antigen on the surface of a tumor cell. DETAILED DESCRIPTION OF THE INVENTION Mechanisms of mAb Action Although the following provides considerations regarding the mechanism underlying the therapeutic efficacy of antibodies of the invention it is not to be considered as limiting to the invention in any way. The antibodies described herein may interact with components of the immune system, preferably through ADCC or CDC. Antibodies of the invention can also be used to target payloads (e.g., radioisotopes, drugs or toxins) to directly kill tumor cells or can be used synergistically with traditional chemotherapeutic agents, attacking tumors through complementary mechanisms of action that may include anti-tumor immune responses that may have been compromised owing to a chemotherapeutic's cytotoxic side effects on T lymphocytes. However, antibodies of the invention may also exert an effect simply by binding to CLDN6 on the cell surface, thus, e.g. blocking proliferation of the cells. Antibody-Dependent Cell-Mediated Cytotoxicity ADCC describes the cell-killing ability of effector cells as described herein, in particular lymphocytes, which preferably requires the target cell being marked by an antibody. ADCC preferably occurs when antibodies bind to antigens on tumor cells and the antibody Fc domains engage Fc receptors (FcR) on the surface of immune effector cells. Several families of Fc receptors have been identified, and specific cell populations characteristically express defined Fc receptors. ADCC can be viewed as a mechanism to directly induce a variable degree of immediate tumor destruction that leads to antigen presentation and the induction of tumor-directed T-cell responses. Preferably, in vivo induction of ADCC will lead to tumor-directed T-cell responses and host-derived antibody responses. Complement-Dependent Cytotoxicity CDC is another cell-killing method that can be directed by antibodies. IgM is the most effective isotype for complement activation. IgG1 and IgG3 are also both very effective at directing CDC via the classical complement-activation pathway. Preferably, in this cascade, the formation of antigen-antibody complexes results in the uncloaking of multiple C1q binding sites in close proximity on the CH2 domains of participating antibody molecules such as IgG molecules (C1q is one of three subcomponents of complement C1). Preferably these uncloaked C1q binding sites convert the previously low-affinity C1q-IgG interaction to one of high avidity, which triggers a cascade of events involving a series of other complement proteins and leads to the proteolytic release of the effector-cell chemotactic/activating agents C3a and C5a. Preferably, the complement cascade ends in the formation of a membrane attack complex, which creates pores in the cell membrane that facilitate free passage of water and solutes into and out of the cell. Production of Antibodies Antibodies of the invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology, e.g., the standard somatic cell hybridization technique of Kohler and Milstein, Nature 256: 495 (1975). Although somatic cell hybridization procedures are preferred, in principle, other techniques for producing monoclonal antibodies can be employed, e.g., viral or oncogenic transformation of B-lymphocytes or phage display techniques using libraries of antibody genes. The preferred animal system for preparing hybridomas that secrete monoclonal antibodies is the murine system. Hybridoma production in the mouse is a very well established procedure. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also known. Other preferred animal systems for preparing hybridomas that secrete monoclonal antibodies are the rat and the rabbit system (e.g. described in Spieker-Polet et al., Proc. Natl. Acad. Sci. U.S.A. 92:9348 (1995), see also Rossi et al., Am. J. Clin. Pathol. 124: 295 (2005)). In yet another preferred embodiment, human monoclonal antibodies directed against CLDN6 can be generated using transgenic or transchromosomal mice carrying parts of the human immune system rather than the mouse system. These transgenic and transchromosomic mice include mice known as HuMAb mice and KM mice, respectively, and are collectively referred to herein as “transgenic mice.” The production of human antibodies in such transgenic mice can be performed as described in detail for CD20 in WO2004 035607 Yet another strategy for generating monoclonal antibodies is to directly isolate genes encoding antibodies from lymphocytes producing antibodies of defined strategy e.g. see Babcock et al., 1996; A novel strategy for generating monoclonal antibodies from single, isolated lymphocytes producing antibodies of defined strategy. For details of recombinant antibody engineering see also Welschof and Kraus, Recombinant antibodies for cancer therapy ISBN-0-89603-918-8 and Benny K. C. Lo Antibody Engineering ISBN 1-58829-092-1. Immunizations To generate antibodies to CLDN6, mice can be immunized with carrier-conjugated peptides derived from the CLDN6 sequence, an enriched preparation of recombinantly expressed CLDN6 antigen or fragments thereof and/or cells expressing CLDN6 or fragments thereof, as described. Alternatively, mice can be immunized with DNA encoding full length human CLDN6 or fragments thereof. In the event that immunizations using a purified or enriched preparation of the CLDN6 antigen do not result in antibodies, mice can also be immunized with cells expressing CLDN6, e.g., a cell line, to promote immune responses. The immune response can be monitored over the course of the immunization protocol with plasma and serum samples being obtained by tail vein or retroorbital bleeds. Mice with sufficient titers of anti-CLDN6 immunoglobulin can be used for fusions. Mice can be boosted intraperitonealy or intravenously with CLDN6 expressing cells 3-5 days before sacrifice and removal of the spleen to increase the rate of specific antibody secreting hybridomas. Generation of Hybridomas Producing Monoclonal Antibodies To generate hybridomas producing monoclonal antibodies to CLDN6, cells from lymph nodes or spleens obtained from immunized mice can be isolated and fused to an appropriate immortalized cell line, such as a mouse myeloma cell line. The resulting hybridomas can then be screened for the production of antigen-specific antibodies. Individual wells can then be screened by ELISA for antibody secreting hybridomas. By Immunofluorescence and FACS analysis using CLDN6 expressing cells, antibodies with specificity for CLDN6 can be identified. The antibody secreting hybridomas can be replated, screened again, and if still positive for anti-CLDN6 monoclonal antibodies can be subcloned by limiting dilution. The stable subclones can then be cultured in vitro to generate antibody in tissue culture medium for characterization. Generation of Transfectomas Producing Monoclonal Antibodies Antibodies of the invention also can be produced in a host cell transfectoma using, for example, a combination of recombinant DNA techniques and gene transfection methods as are well known in the art (Morrison, S. (1985) Science 229: 1202). For example, in one embodiment, the gene(s) of interest, e.g., antibody genes, can be ligated into an expression vector such as a eukaryotic expression plasmid such as used by the GS gene expression system disclosed in WO 87/04462, WO 89/01036 and EP 338 841 or other expression systems well known in the art. The purified plasmid with the cloned antibody genes can be introduced in eukaryotic host cells such as CHO cells, NS/0 cells, HEK293T cells or HEK293 cells or alternatively other eukaryotic cells like plant derived cells, fungal or yeast cells. The method used to introduce these genes can be methods described in the art such as electroporation, lipofectine, lipofectamine or others. After introduction of these antibody genes in the host cells, cells expressing the antibody can be identified and selected. These cells represent the transfectomas which can then be amplified for their expression level and upscaled to produce antibodies. Recombinant antibodies can be isolated and purified from these culture supernatants and/or cells. Alternatively, the cloned antibody genes can be expressed in other expression systems, including prokaryotic cells, such as microorganisms, e.g.E. coli. Furthermore, the antibodies can be produced in transgenic non-human animals, such as in milk from sheep and rabbits or in eggs from hens, or in transgenic plants; see e.g. Verma, R., et al. (1998) J. Immunol. Meth. 216: 165-181; Pollock, et al. (1999) J. Immunol. Meth. 231: 147-157; and Fischer, R., et al. (1999) Biol. Chem. 380: 825-839. Use of Partial Antibody Sequences to Express Intact Antibodies (i.e. Humanization and Chimerization). a) Chimerization Murine monoclonal antibodies can be used as therapeutic antibodies in humans when labeled with toxins or radioactive isotopes. Nonlabeled murine antibodies are highly immunogenic in man when repetitively applied leading to reduction of the therapeutic effect. The main immunogenicity is mediated by the heavy chain constant regions. The immunogenicity of murine antibodies in man can be reduced or completely avoided if respective antibodies are chimerized or humanized. Chimeric antibodies are antibodies, the different portions of which are derived from different animal species, such as those having a variable region derived from a murine antibody and a human immunoglobulin constant region. Chimerization of antibodies is achieved by joining of the variable regions of the murine antibody heavy and light chain with the constant region of human heavy and light chain (e.g. as described by Kraus et al., in Methods in Molecular Biology series, Recombinant antibodies for cancer therapy ISBN-0-89603-918-8). In a preferred embodiment chimeric antibodies are generated by joining human kappa-light chain constant region to murine light chain variable region. In an also preferred embodiment chimeric antibodies can be generated by joining human lambda-light chain constant region to murine light chain variable region. The preferred heavy chain constant regions for generation of chimeric antibodies are IgG1, IgG3 and IgG4. Other preferred heavy chain constant regions for generation of chimeric antibodies are IgG2, IgA, IgD and IgM. b) Humanization Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto framework sequences from a different antibody with different properties (see, e.g., Riechmann, L. et al. (1998) Nature 332: 323-327; Jones, P. et al. (1986) Nature 321: 522-525; and Queen, C. et al. (1989) Proc. Natl. Acad. Sci. U.S.A 86: 10029-10033). Such framework sequences can be obtained from public DNA databases that include germline antibody gene sequences. These germline sequences will differ from mature antibody gene sequences because they will not include completely assembled variable genes, which are formed by V (D) J joining during B cell maturation. Germline gene sequences will also differ from the sequences of a high affinity secondary repertoire antibody at individual evenly across the variable region. For example, somatic mutations are relatively infrequent in the amino terminal portion of framework region 1 and in the carboxy-terminal portion of framework region 4. Furthermore, many somatic mutations do not significantly alter the binding properties of the antibody. For this reason, it is not necessary to obtain the entire DNA sequence of a particular antibody in order to recreate an intact recombinant antibody having binding properties similar to those of the original antibody (see WO 99/45962). Partial heavy and light chain sequences spanning the CDR regions are typically sufficient for this purpose. The partial sequence is used to determine which germline variable and joining gene segments contributed to the recombined antibody variable genes. The germline sequence is then used to fill in missing portions of the variable regions. Heavy and light chain leader sequences are cleaved during protein maturation and do not contribute to the properties of the final antibody. To add missing sequences, cloned cDNA sequences can be combined with synthetic oligonucleotides by ligation or PCR amplification. Alternatively, the entire variable region can be synthesized as a set of short, overlapping, oligonucleotides and combined by PCR amplification to create an entirely synthetic variable region clone. This process has certain advantages such as elimination or inclusion or particular restriction sites, or optimization of particular codons. The nucleotide sequences of heavy and light chain transcripts from hybridomas are used to design an overlapping set of synthetic oligonucleotides to create synthetic V sequences with identical amino acid coding capacities as the natural sequences. The synthetic heavy and kappa chain sequences can differ from the natural sequences in three ways: strings of repeated nucleotide bases are interrupted to facilitate oligonucleotide synthesis and PCR amplification; optimal translation initiation sites are incorporated according to Kozak's rules (Kozak, 1991, J. Biol. Chem. 266: 19867-19870); and HindIII sites are engineered upstream of the translation initiation sites. For both the heavy and light chain variable regions, the optimized coding and corresponding non-coding, strand sequences are broken down into 30-50 nucleotides approximately at the midpoint of the corresponding non-coding oligonucleotide. Thus, for each chain, the oligonucleotides can be assembled into overlapping double stranded sets that span segments of 150-400 nucleotides. The pools are then used as templates to produce PCR amplification products of 150-400 nucleotides. Typically, a single variable region oligonucleotide set will be broken down into two pools which are separately amplified to generate two overlapping PCR products. These overlapping products are then combined by PCR amplification to form the complete variable region. It may also be desirable to include an overlapping fragment of the heavy or light chain constant region in the PCR amplification to generate fragments that can easily be cloned into the expression vector constructs. The reconstructed chimerized or humanized heavy and light chain variable regions are then combined with cloned promoter, leader, translation initiation, constant region, 3′ untranslated, polyadenylation, and transcription termination sequences to form expression vector constructs. The heavy and light chain expression constructs can be combined into a single vector, co-transfected, serially transfected, or separately transfected into host cells which are then fused to form a host cell expressing both chains. Plasmids for use in construction of expression vectors for human IgGκ are described. The plasmids can be constructed so that PCR amplified V heavy and V kappa light chain cDNA sequences can be used to reconstruct complete heavy and light chain minigenes. These plasmids can be used to express completely human, or chimeric IgG1, Kappa or IgG4, Kappa antibodies. Similar plasmids can be constructed for expression of other heavy chain isotypes, or for expression of antibodies comprising lambda light chains. Thus, in another aspect of the invention, the structural features of the anti-CLDN6 antibodies of the invention, are used to create structurally related humanized anti-CLDN6 antibodies that retain at least one functional property of the antibodies of the invention, such as binding to CLDN6. More specifically, one or more CDR regions of mouse monoclonal antibodies can be combined recombinantly with known human framework regions and CDRs to create additional, recombinantly-engineered, humanized anti-CLDN6 antibodies of the invention. Binding to Antigen Expressing Cells The ability of the antibody to bind CLDN6 can be determined using standard binding assays, such as those set forth in the examples (e.g., ELISA, Western Blot, Immunofluorescence and flow cytometric analysis) Isolation and Characterization of Antibodies To purify anti-CLDN6 antibodies, selected hybridomas can be grown in two-liter spinner-flasks for monoclonal antibody purification. Alternatively, anti-CLDN6 antibodies can be produced in dialysis based bioreactors. Supernatants can be filtered and, if necessary, concentrated before affinity chromatography with protein G-sepharose or protein A-sepharose. Eluted IgG can be checked by gel electrophoresis and high performance liquid chromatography to ensure purity. The buffer solution can be exchanged into PBS, and the concentration can be determined by OD280 using 1.43 extinction coefficient. The monoclonal antibodies can be aliquoted and stored at −80° C. To determine if the selected anti-CLDN6 monoclonal antibodies bind to unique epitopes, site-directed or multi-site directed mutagenesis can be used. Isotype Determination To determine the isotype of purified antibodies, isotype ELISAs with various commercial kits (e.g. Zymed, Roche Diagnostics) can be performed. Wells of microtiter plates can be coated with anti-mouse Ig. After blocking, the plates are reacted with monoclonal antibodies or purified isotype controls, at ambient temperature for two hours. The wells can then be reacted with either mouse IgG1, IgG2a, IgG2b or IgG3, IgA or mouse IgM-specific peroxidase-conjugated probes. After washing, the plates can be developed with ABTS substrate (1 mg/ml) and analyzed at OD of 405-650. Alternatively, the IsoStrip Mouse Monoclonal Antibody Isotyping Kit (Roche, Cat. No. 1493027) may be used as described by the manufacturer. Flow Cytometric Analysis In order to demonstrate presence of anti-CLDN6 antibodies in sera of immunized mice or binding of monoclonal antibodies to living cells expressing CLDN6, flow cytometry can be used. Cell lines expressing naturally or after transfection CLDN6 and negative controls lacking CLDN6 expression (grown under standard growth conditions) can be mixed with various concentrations of monoclonal antibodies in hybridoma supernatants or in PBS containing 1% FBS, and can be incubated at 4° C. for 30 min. After washing, the APC- or Alexa647-labeled anti IgG antibody can bind to CLDN6-bound monoclonal antibody under the same conditions as the primary antibody staining. The samples can be analyzed by flow cytometry with a FACS instrument using light and side scatter properties to gate on single, living cells. In order to distinguish CLDN6-specific monoclonal antibodies from non-specific binders in a single measurement, the method of co-transfection can be employed. Cells transiently transfected with plasmids encoding CLDN6 and a fluorescent marker can be stained as described above. Transfected cells can be detected in a different fluorescence channel than antibody-stained cells. As the majority of transfected cells express both transgenes, CLDN6-specific monoclonal antibodies bind preferentially to fluorescence marker expressing cells, whereas non-specific antibodies bind in a comparable ratio to non-transfected cells. An alternative assay using fluorescence microscopy may be used in addition to or instead of the flow cytometry assay. Cells can be stained exactly as described above and examined by fluorescence microscopy. Immunofluorescence Microscopy In order to demonstrate presence of anti-CLDN6 antibodies in sera of immunized mice or binding of monoclonal antibodies to living cells expressing CLDN6, immunofluorescence microscopy analysis can be used. For example, cell lines expressing either spontaneously or after transfection CLDN6 and negative controls lacking CLDN6 expression are grown in chamber slides under standard growth conditions in DMEM/F12 medium, supplemented with 10% fetal calf serum (FCS), 2 mM L-glutamine, 100 IU/ml penicillin and 100 μg/ml streptomycin. Cells can then be fixed with methanol or paraformaldehyde or left untreated. Cells can then be reacted with monoclonal antibodies against CLDN6 for 30 min. at 25° C. After washing, cells can be reacted with an Alexa555-labelled anti-mouse IgG secondary antibody (Molecular Probes) under the same conditions. Cells can then be examined by fluorescence microscopy. Total CLDN6 levels in cells can be observed when cells are methanol fixed or paraformaldehyde fixed and permeabilized with Triton X-100. In living cells and non-permeabilized, paraformaldehyde fixed cells surface localization of CLDN6 can be examined. Additionally targeting of CLDN6 to tight junctions can be analyzed by co-staining with tight junction markers such as ZO-1. Furthermore, effects of antibody binding and CLDN6 localization within the cell membrane can be examined. Western Blot Anti-CLDN6 IgG can be further tested for reactivity with CLDN6 antigen by Western Blotting. Briefly, cell extracts from cells expressing CLDN6 and appropriate negative controls can be prepared and subjected to sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis. After electrophoresis, the separated antigens will be transferred to nitrocellulose membranes, blocked, and probed with the monoclonal antibodies to be tested. IgG binding can be detected using anti-mouse IgG peroxidase and developed with ECL substrate. Immunohistochemistry Anti-CLDN6 mouse IgGs can be further tested for reactivity with CLDN6 antigen by Immunohistochemistry in a manner well known to the skilled person, e.g. using paraformaldehyde or acetone fixed cryosections or paraffin embedded tissue sections fixed with paraformaldehyde from non-cancer tissue or cancer tissue samples obtained from patients during routine surgical procedures or from mice carrying xenografted tumors inoculated with cell lines expressing spontaneously or after transfection CLDN6. For immunostaining, antibodies reactive to CLDN6 can be incubated followed by horseradish-peroxidase conjugated goat anti-mouse or goat anti-rabbit antibodies (DAKO) according to the vendor's instructions. Phagocytic and Cell Killing Activities of Antibodies In Vitro In addition to binding specifically to CLDN6, anti-CLDN6 antibodies can be tested for their ability to mediate phagocytosis and killing of cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface. The testing of monoclonal antibody activity in vitro will provide an initial screening prior to testing in vivo models. Antibody Dependent Cell-Mediated Cytotoxicity (ADCC): Briefly, polymorphonuclear cells (PMNs), NK cells, monocytes, mononuclear cells or other effector cells, from healthy donors can be purified by Ficoll Hypaque density centrifugation, followed by lysis of contaminating erythrocytes. Washed effector cells can be suspended in RPMI supplemented with 10% heat-inactivated fetal calf serum or, alternatively with 5% heat-inactivated human serum and mixed with51Cr labeled target cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface, at various ratios of effector cells to target cells. Alternatively, the target cells may be labeled with a fluorescence enhancing ligand (BATDA). A highly fluorescent chelate of Europium with the enhancing ligand which is released from dead cells can be measured by a fluorometer. Another alternative technique may utilize the transfection of target cells with luciferase. Added lucifer yellow may then be oxidated by viable cells only. Purified anti-CLDN6 IgGs can then be added at various concentrations. Irrelevant human IgG can be used as negative control. Assays can be carried out for 4 to 20 hours at 37° C. depending on the effector cell type used. Samples can be assayed for cytolysis by measuring51Cr release or the presence of the EuTDA chelate in the culture supernatant. Alternatively, luminescence resulting from the oxidation of lucifer yellow can be a measure of viable cells. Anti-CLDN6 monoclonal antibodies can also be tested in various combinations to determine whether cytolysis is enhanced with multiple monoclonal antibodies. Complement Dependent Cytotoxicity (CDC): Monoclonal anti-CLDN6 antibodies can be tested for their ability to mediate CDC using a variety of known techniques. For example, serum for complement can be obtained from blood in a manner known to the skilled person. To determine the CDC activity of mAbs, different methods can be used.51Cr release can for example be measured or elevated membrane permeability can be assessed using a propidium iodide (PI) exclusion assay. Briefly, target cells can be washed and 5×105/ml can be incubated with various concentrations of mAb for 10-30 min. at room temperature or at 37° C. Serum or plasma can then be added to a final concentration of 20% (v/v) and the cells incubated at 37° C. for 20-30 min. All cells from each sample can be added to the PI solution in a FACS tube. The mixture can then be analyzed immediately by flow cytometry analysis using FACSArray. In an alternative assay, induction of CDC can be determined on adherent cells. In one embodiment of this assay, cells are seeded 24 h before the assay with a density of 3×104/well in tissue-culture flat-bottom microtiter plates. The next day growth medium is removed and the cells are incubated in triplicates with antibodies. Control cells are incubated with growth medium or growth medium containing 0.2% saponin for the determination of background lysis and maximal lysis, respectively. After incubation for 20 min. at room temperature supernatant is removed and 20% (v/v) human plasma or serum in DMEM (prewarmed to 37° C.) is added to the cells and incubated for another 20 min. at 37° C. All cells from each sample are added to propidium iodide solution (10 μg/ml). Then, supernatants are replaced by PBS containing 2.5 μg/ml ethidium bromide and fluorescence emission upon excitation at 520 nm is measured at 600 nm using a Tecan Safire. The percentage specific lysis is calculated as follows: % specific lysis=(fluorescence sample-fluorescence background)/(fluorescence maximal lysis-fluorescence background)×100. Inhibition of Cell Proliferation by Monoclonal Antibodies: To test for the ability to initiate apoptosis, monoclonal anti-CLDN6 antibodies can, for example, be incubated with CLDN6 positive tumor cells or CLDN6 transfected tumor cells at 37° C. for about 20 hours. The cells can be harvested, washed in Annexin-V binding buffer (BD biosciences), and incubated with Annexin V conjugated with FITC or APC (BD biosciences) for 15 min. in the dark. All cells from each sample can be added to PI solution (10 μg/ml in PBS) in a FACS tube and assessed immediately by flow cytometry (as above). Alternatively, a general inhibition of cell-proliferation by monoclonal antibodies can be detected with commercially available kits. The DELFIA Cell Proliferation Kit (Perkin-Elmer, Cat. No. AD0200) is a non-isotopic immunoassay based on the measurement of 5-bromo-2′-deoxyuridine (BrdU) incorporation during DNA synthesis of proliferating cells in microplates. Incorporated BrdU is detected using europium labelled monoclonal antibody. To allow antibody detection, cells are fixed and DNA denatured using Fix solution. Unbound antibody is washed away and DELFIA inducer is added to dissociate europium ions from the labelled antibody into solution, where they form highly fluorescent chelates with components of the DELFIA Inducer. The fluorescence measured—utilizing time-resolved fluorometry in the detection—is proportional to the DNA synthesis in the cell of each well. Preclinical Studies Monoclonal antibodies which bind to CLDN6 also can be tested in an in vivo model (e.g. in immune deficient mice carrying xenografted tumors inoculated with cell lines expressing CLDN6, possibly after transfection) to determine their efficacy in controlling growth of CLDN6-expressing tumor cells. In vivo studies after xenografting CLDN6 expressing tumor cells into immunocompromised mice or other animals can be performed using antibodies of the invention. Antibodies can be adminstered to tumor free mice followed by injection of tumor cells to measure the effects of the antibodies to prevent formation of tumors or tumor-related symptoms. Antibodies can be adminstered to tumor-bearing mice to determine the therapeutic efficacy of respective antibodies to reduce tumor growth, metastasis or tumor related symptoms. Antibody application can be combined with application of other substances as cystostatic drugs, growth factor inhibitors, cell cycle blockers, angiogenesis inhibitors or other antibodies to determine synergistic efficacy and potential toxicity of combinations. To analyze toxic side effects mediated by antibodies of the invention animals can be inoculated with antibodies or control reagents and thoroughly investigated for symptoms possibly related to CLDN6-antibody therapy. Possible side effects of in vivo application of CLDN6 antibodies particularly include toxicity at CLDN6 expressing tissues including placenta. Antibodies recognizing CLDN6 in human and in other species, e.g. mice, are particularly useful to predict potential side effects mediated by application of monoclonal CLDN6 antibodies in humans. Epitope Mapping Mapping of epitopes recognized by antibodies of invention can be performed as described in detail in “Epitope Mapping Protocols (Methods in Molecular Biology) by Glenn E. Morris ISBN-089603-375-9 and in “Epitope Mapping: A Practical Approach” Practical Approach Series, 248 by Olwyn M. R. Westwood, Frank C. Hay. I. Bispecific/Multispecific Molecules which Bind to CLDN6 In yet another embodiment of the invention, antibodies to CLDN6 can be derivatized or linked to another functional molecule, e.g., another peptide or protein (e.g., an Fab′ fragment) to generate a bispecific or multispecific molecule which binds to multiple binding sites or target epitopes. For example, an antibody of the invention can be functionally linked (e.g. by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other binding molecules, such as another antibody, peptide or binding mimetic. Accordingly, the present invention includes bispecific and multispecific molecules comprising at least one first binding specificity for CLDN6 and a second binding specificity for a second target epitope. In a particular embodiment of the invention, the second target epitope is an Fc receptor, e.g. human Fc-gammaRI (CD64) or a human Fc-alpha receptor (CD89), or a T cell receptor, e.g. CD3. Therefore, the invention includes bispecific and multispecific molecules capable of binding both to Fc-gammaR, Fc-alphaR or Fc-epsilonR expressing effector cells (e.g. monocytes, macrophagesor polymorphonuclear cells (PMNs)), and to target cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface. These bispecific and multispecific molecules may target cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface to effector cells and may trigger Fc receptor-mediated effector cell activities, such as phagocytosis of cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface, antibody dependent cellular cytotoxicity (ADCC), cytokine release, or generation of superoxide anion. Bispecific and multispecific molecules of the invention can further include a third binding specificity, in addition to an anti-Fc binding specificity and an anti-CLDN6 binding specificity. In one embodiment, the third binding specificity is an anti-enhancement factor (EF) portion, e.g. a molecule which binds to a surface protein involved in cytotoxic activity and thereby increases the immune response against the target cell. The “anti-enhancement factor portion” can be an antibody, functional antibody fragment or a ligand that binds to a given molecule, e.g., an antigen or a receptor, and thereby results in an enhancement of the effect of the binding determinants for the Fc receptor or target cell antigen. The “anti-enhancement factor portion” can bind an Fc receptor or a target cell antigen. Alternatively, the anti-enhancement factor portion can bind to an entity that is different from the entity to which the first and second binding specificities bind. For example, the anti-enhancement factor portion can bind a cytotoxic T cell (e.g., via CD2, CD3, CD8, CD28, CD4, CD40, ICAM-1 or other immune cell that results in an increased immune response against the target cell). In one embodiment, the bispecific and multispecific molecules of the invention comprise as a binding specificity at least one antibody, including, e.g., an Fab, Fab′, F(ab′)2, Fv, or a single chain Fv. The antibody may also be a light chain or heavy chain dimer, or any minimal fragment thereof such as a Fv or a single chain construct as described in Ladner et al., U.S. Pat. No. 4,946,778. The antibody may also be a binding-domain immunoglobulin fusion protein as disclosed in US2003/0118592 and US 2003/0133939. In one embodiment bispecific and multispecific molecules of the invention comprise a binding specificity for an Fc-gammaR or an Fc-alphaR present on the surface of an effector cell, and a second binding specificity for a target cell antigen, e.g., CLDN6. In one embodiment, the binding specificity for an Fc receptor is provided by a monoclonal antibody, the binding of which is not blocked by human immunoglobulin G (IgG). As used herein, the term “IgG receptor” refers to any of the eight gamma-chain genes located on chromosome 1. These genes encode a total of twelve transmembrane or soluble receptor isoforms which are grouped into three Fc-gamma receptor classes: Fc-gammaRI (CD64), Fc-gammaRII (CD32), and Fc-gammaRIII (CD16). In one preferred embodiment, the Fc-gamma receptor is a human high affinity Fc-gammaRI. In still other preferred embodiments, the binding specificity for an Fc receptor is provided by an antibody that binds to a human IgA receptor, e.g., an Fc-alpha receptor (Fc-alphaRI (CD89)), the binding of which is preferably not blocked by human immunoglobulin A (IgA). The term “IgA receptor” is intended to include the gene product of one alpha-gene (Fc-alphaRI) located on chromosome 19. This gene is known to encode several alternatively spliced transmembrane isoforms of 55 to 110 kDa. Fc-alphaRI (CD89) is constitutively expressed on monocytes/macrophages, eosinophilic and neutrophilic granulocytes, but not on non-effector cell populations. Fc-alphaRI has medium affinity for both IgA1 and IgA2, which is increased upon exposure to cytokines such as G-CSF or GM-CSF (Morton, H. C. et al. (1996) Critical Reviews in Immunology 16: 423-440). Four Fc-alphaRI-specific monoclonal antibodies, identified as A3, A59, A62 and A77, which bind Fc-alphaRI outside the IgA ligand binding domain, have been described (Monteiro, R. C. et al. (1992) J. Immunol. 148: 1764). In another embodiment the bispecific molecule is comprised of two monoclonal antibodies according to the invention which have complementary functional activities, such as one antibody predominately working by inducing CDC and the other antibody predominately working by inducing apoptosis. An “effector cell specific antibody” as used herein refers to an antibody or functional antibody fragment that binds the Fc receptor of effector cells. Preferred antibodies for use in the subject invention bind the Fc receptor of effector cells at a site which is not bound by endogenous immunoglobulin. As used herein, the term “effector cell” refers to an immune cell which is involved in the effector phase of an immune response, as opposed to the cognitive and activation phases of an immune response. Exemplary immune cells include cells of myeloid or lymphoid origin, e.g, lymphocytes (e.g., B cells and T cells including cytolytic T cells (CTLs), killer cells, natural killer cells, macrophages, monocytes, eosinophils, neutrophils, polymorphonuclear cells, granulocytes, mast cells, and basophils. Some effector cells express specific Fc receptors and carry out specific immune functions. In preferred embodiments, an effector cell is capable of inducing antibody-dependent cellular cytotoxicity (ADCC), e.g., a neutrophil capable of inducing ADCC. For example, monocytes, macrophages, which express FcR are involved in specific killing of target cells and presenting antigens to other components of the immune system, or binding to cells that present antigens. In other embodiments, an effector cell can phagocytose a target antigen, target cell, or microorganism. The expression of a particular FcR on an effector cell can be regulated by humoral factors such as cytokines. For example, expression of Fc-gammaRI has been found to be up-regulated by interferon gamma (IFN-γ). This enhanced expression increases the cytotoxic activity of Fc-gammaRI-bearing cells against targets. An effector cell can phagocytose or lyse a target antigen or a target cell. “Target cell” shall mean any undesirable cell in a subject (e.g., a human or animal) that can be targeted by an antibody of the invention. In preferred embodiments, the target cell is a cell expressing or overexpressing CLDN6 and being characterized by association of CLDN6 with its cell surface. Cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface typically include tumor cells. II. Immunoconjugates In another aspect, the present invention features an anti-CLDN6 antibody conjugated to a therapeutic moiety or agent, such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radioisotope. Such conjugates are referred to herein as “immunoconjugates”. Immunoconjugates which include one or more cytotoxins are referred to as “immunotoxins”. A cytotoxin or cytotoxic agent includes any agent that is detrimental to and, in particular, kills cells. Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Suitable therapeutic agents for forming immunoconjugates of the invention include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, fludarabin, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC), and anti-mitotic agents (e.g., vincristine and vinblastine). In a preferred embodiment, the therapeutic agent is a cytotoxic agent or a radiotoxic agent. In another embodiment, the therapeutic agent is an immunosuppressant. In yet another embodiment, the therapeutic agent is GM-CSF. In a preferred embodiment, the therapeutic agent is doxorubicin, cisplatin, bleomycin, sulfate, carmustine, chlorambucil, cyclophosphamide or ricin A. Antibodies of the present invention also can be conjugated to a radioisotope, e.g., iodine-131, yttrium-90 or indium-111, to generate cytotoxic radiopharmaceuticals for treating a CLDN6-related disorder, such as a cancer. The antibody conjugates of the invention can be used to modify a given biological response, and the drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, an enzymatically active toxin, or active fragment thereof, such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor or interferon-γ; or, biological response modifiers such as, for example, lymphokines, interleukin-1 (“IL-1”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors. Techniques for conjugating such therapeutic moiety to antibodies are well known, see, e.g., Arnon et al., “Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy”, in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al., “Antibodies For Drug Delivery”, in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thorpe, “Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review”, in Monoclonal Antibodies '84: Biological And Clinical Applications, Pincheraet al. (eds.), pp. 475-506 (1985); “Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy”, in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., “The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates”, Immunol. Rev., 62: 119-58 (1982). In a further embodiment, the antibodies according to the invention are attached to a linker-chelator, e.g., tiuxetan, which allows for the antibody to be conjugated to a radioisotope. III. Pharmaceutical Compositions In another aspect, the present invention provides a composition, e.g., a pharmaceutical composition, containing one or a combination of antibodies of the present invention. The pharmaceutical compositions may be formulated with pharmaceutically acceptable carriers or diluents as well as any other known adjuvants and excipients in accordance with conventional techniques such as those disclosed in Remington: The Science and Practice of Pharmacy, 19th Edition, Gennaro, Ed., Mack Publishing Co., Easton, P A, 1995. In one embodiment, the compositions include a combination of multiple (e.g., two or more) isolated antibodies of the invention which act by different mechanisms, e.g., one antibody which predominately acts by inducing CDC in combination with another antibody which predominately acts by inducing apoptosis. Pharmaceutical compositions of the invention also can be administered in combination therapy, i.e., combined with other agents. For example, the combination therapy can include a composition of the present invention with at least one anti-inflammatory agent or at least one immunosuppressive agent. In one embodiment such therapeutic agents include one or more anti-inflammatory agents, such as a steroidal drug or a NSAID (nonsteroidal anti-inflammatory drug). Preferred agents include, for example, aspirin and other salicylates, Cox-2 inhibitors, such as rofecoxib (Vioxx) and celecoxib (Celebrex), NSAIDs such as ibuprofen (Motrin, Advil), fenoprofen (Nalfon), naproxen (Naprosyn), sulindac (Clinoril), diclofenac (Voltaren), piroxicam (Feldene), ketoprofen (Orudis), diflunisal (Dolobid), nabumetone (Relafen), etodolac (Lodine), oxaprozin (Daypro), and indomethacin (Indocin). In another embodiment, such therapeutic agents include agents leading to the depletion or functional inactivation of regulatory T cells like low dose cyclophosphamid, anti-CTLA4 antibodies, anti-IL2 or anti-IL2-receptor antibodies. In yet another embodiment, such therapeutic agents include one or more chemotherapeutics, such as Taxol derivatives, taxotere, gemcitabin, 5-Fluoruracil, doxorubicin (Adriamycin), cisplatin (Platinol), cyclophosphamide (Cytoxan, Procytox, Neosar). In another embodiment, antibodies of the present invention may be administered in combination with chemotherapeutic agents, which preferably show therapeutic efficacy in patients suffering from cancer, e.g. cancer types as described herein. In yet another embodiment, the antibodies of the invention may be administered in conjunction with radiotherapy and/or autologous peripheral stem cell or bone marrow transplantation. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion). Depending on the route of administration, the active compound, e.g., antibody, bispecific and multispecific molecule, may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound. A “pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge, S. M., et al. (1977) J. Pharm. Sci. 66: 1-19). Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like. Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as N,N′-dibenzylethylenediamine, N-methylglucamine, chloroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like. A composition of the present invention can be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. The active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for the preparation of such formulations are generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. To administer a compound of the invention by certain routes of administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. For example, the compound may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Liposomes include water-in-oil-in-water CGF emulsions as well as conventional liposomes (Strejan et al. (1984) J. Neuroimmunol. 7: 27). Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. The use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions. Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin. Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. For the therapeutic compositions, formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, and the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect. Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate. Dosage forms for the topical or transdermal administration of compositions of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required. The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the presence of microorganisms may be ensured both by sterilization procedures, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In general, a suitable daily dose of a composition of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. It is preferred that administration be intravenous, intramuscular, intraperitoneal, or subcutaneous, preferably administered proximal to the site of the target. If desired, the effective daily dose of a therapeutic composition may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition). In one embodiment, the antibodies of the invention may be administered by infusion, preferably slow continuous infusion over a long period, such as more than 24 hours, in order to reduce toxic side effects. The administration may also be performed by continuous infusion over a period of from 2 to 24 hours, such as of from 2 to 12 hours. Such regimen may be repeated one or more times as necessary, for example, after 6 months or 12 months. The dosage can be determined or adjusted by measuring the amount of circulating monoclonal anti-CLDN6 antibodies upon administration in a biological sample by using anti-idiotypic antibodies which target the anti-CLDN6 antibodies. In yet another embodiment, the antibodies are administered by maintenance therapy, such as, e.g., once a week for a period of 6 months or more. In still another embodiment, the antibodies according to the invention may be administered by a regimen including one infusion of an antibody against CLDN6 followed by an infusion of an antibody against CLDN6 conjugated to a radioisotope. The regimen may be repeated, e.g., 7 to 9 days later. In one embodiment of the invention, the therapeutic compounds of the invention are formulated in liposomes. In a more preferred embodiment, the liposomes include a targeting moiety. In a most preferred embodiment, the therapeutic compounds in the liposomes are delivered by bolus injection to a site proximal to the desired area, e.g., the site of a tumor. The composition must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. In a further embodiment, antibodies of the invention can be formulated to prevent or reduce their transport across the placenta. This can be done by methods known in the art, e.g., by PEGylation of the antibodies or by use of F(ab)2′ fragments. Further references can be made to “Cunningham-Rundles C, Zhuo Z, Griffith B, Keenan J. (1992) Biological activities of polyethylene-glycol immunoglobulin conjugates. Resistance to enzymatic degradation. J. Immunol. Methods, 152: 177-190; and to “Landor M. (1995) Maternal-fetal transfer of immunoglobulins, Ann. Allergy Asthma Immunol. 74: 279-283. A “therapeutically effective dosage” for tumor therapy can be measured by objective tumor responses which can either be complete or partial. A complete response (CR) is defined as no clinical, radiological or other evidence of disease. A partial response (PR) results from a reduction in aggregate tumor size of greater than 50%. Median time to progression is a measure that characterizes the durability of the objective tumor response. A “therapeutically effective dosage” for tumor therapy can also be measured by its ability to stabilize the progression of disease. The ability of a compound to inhibit cancer can be evaluated in an animal model system predictive of efficacy in human tumors. Alternatively, this property of a composition can be evaluated by examining the ability of the compound to inhibit cell growth or apoptosis by in vitro assays known to the skilled practitioner. A therapeutically effective amount of a therapeutic compound can decrease tumor size, or otherwise ameliorate symptoms in a subject. One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected. The composition must be sterile and fluid to the extent that the composition is deliverable by syringe. In addition to water, the carrier can be an isotonic buffered saline solution, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. Proper fluidity can be maintained, for example, by use of coating such as lecithin, by maintenance of required particle size in the case of dispersion and by use of surfactants. In many cases, it is preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol or sorbitol, and sodium chloride in the composition. Long-term absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin. When the active compound is suitably protected, as described above, the compound may be orally administered, for example, with an inert diluent or an assimilable edible carrier. IV. Uses and Methods of the Invention The antibodies (including immunoconjugates, bispecifics/multispecifics, compositions and other derivatives described herein) of the present invention have numerous therapeutic utilities involving the treatment of disorders involving cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface. For example, the antibodies can be administered to cells in culture, e.g., in vitro or ex vivo, or to human subjects, e.g., in vivo, to treat or prevent a variety of disorders such as those described herein. Preferred subjects include human patients having disorders that can be corrected or ameliorated by killing diseased cells, in particular cells characterized by an altered expression pattern of CLDN6 and/or an altered pattern of association of CLDN6 with their cell surface compared to normal cells. For example, in one embodiment, antibodies of the present invention can be used to treat a subject with a tumorigenic disorder, e.g., a disorder characterized by the presence of tumor cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface. Examples of tumorigenic diseases which can be treated and/or prevented encompass all CLDN6 expressing cancers and tumor entities including those described herein. The pharmaceutical compositions and methods of treatment described according to the invention may also be used for immunization or vaccination to prevent a disease described herein. In another embodiment, antibodies of the invention can be used to detect levels of CLDN6 or particular forms of CLDN6, or levels of cells which contain CLDN6 on their membrane surface, which levels can then be linked to certain diseases or disease symptoms such as described above. Alternatively, the antibodies can be used to deplete or interact with the function of cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface, thereby implicating these cells as important mediators of the disease. This can be achieved by contacting a sample and a control sample with the anti-CLDN6 antibody under conditions that allow for the formation of a complex between the antibody and CLDN6. Any complexes formed between the antibody and CLDN6 are detected and compared in the sample and a control sample, i.e. a reference sample. Antibodies of the invention can be initially tested for their binding activity associated with therapeutic or diagnostic uses in vitro. For example, the antibodies can be tested using flow cytometric assays as described herein. The antibodies of the invention can be used to elicit in vivo or in vitro one or more of the following biological activities: to inhibit the growth of and/or differentiation of a cell expressing CLDN6 and being characterized by association of CLDN6 with its cell surface; to kill a cell expressing CLDN6 and being characterized by association of CLDN6 with its cell surface; to mediate phagocytosis or ADCC of a cell expressing CLDN6 and being characterized by association of CLDN6 with its cell surface in the presence of effector cells; to mediate CDC of a cell expressing CLDN6 and being characterized by association of CLDN6 with its cell surface in the presence of complement; to mediate apoptosis of a cell expressing CLDN6 and being characterized by association of CLDN6 with its cell surface; to induce homotypic adhesion; and/or to induce translocation into lipid rafts upon binding CLDN6. In a particular embodiment, the antibodies are used in vivo or in vitro to treat, prevent or diagnose a variety of CLDN6-related diseases. Examples of CLDN6-related diseases include, among others, cancers such as those described herein. As described above, anti-CLDN6 antibodies of the invention can be co-administered with one or other more therapeutic agents, e.g., a cytotoxic agent, a radiotoxic agent, antiangiogeneic agent or and immunosuppressive agent to reduce the induction of immune responses against the antibodies of invention. The antibody can be linked to the agent (as an immunocomplex) or can be administered separate from the agent. In the latter case (separate administration), the antibody can be administered before, after or concurrently with the agent or can be co-administered with other known therapies, e.g., an anti-cancer therapy, e.g., radiation. Such therapeutic agents include, among others, anti-neoplastic agents such as listed above. Co-administration of the anti-CLDN6 antibodies of the present invention with chemotherapeutic agents provides two anti-cancer agents which operate via different mechanisms yielding a cytotoxic effect to tumor cells. Such co-administration can solve problems due to development of resistance to drugs or a change in the antigenicity of the tumor cells which would render them unreactive with the antibody. The compositions (e.g., antibodies, multispecific and bispecific molecules and immunoconjugates) of the invention which have complement binding sites, such as portions from IgG1, -2, or -3 or IgM which bind complement, can also be used in the presence of complement. In one embodiment, ex vivo treatment of a population of cells comprising target cells with a binding agent of the invention and appropriate effector cells can be supplemented by the addition of complement or serum containing complement. Phagocytosis of target cells coated with a binding agent of the invention can be improved by binding of complement proteins. In another embodiment target cells coated with the compositions of the invention can also be lysed by complement. In yet another embodiment, the compositions of the invention do not activate complement. The compositions of the invention can also be administered together with complement. Accordingly, within the scope of the invention are compositions comprising antibodies, multispecific or bispecific molecules and serum or complement. These compositions are advantageous in that the complement is located in close proximity to the antibodies, multispecific or bispecific molecules. Alternatively, the antibodies, multispecific or bispecific molecules of the invention and the complement or serum can be administered separately. Binding of the compositions of the present invention to target cells may cause translocation of the CLDN6 antigen-antibody complex into lipid rafts of the cell membrane. Such translocation creates a high density of antigen-antibody complexes which may efficiently activate and/or enhance CDC. Also within the scope of the present invention are kits comprising the antibody compositions of the invention (e.g., antibodies and immunoconjugates) and instructions for use. The kit can further contain one or more additional reagents, such as an immunosuppressive reagent, a cytotoxic agent or a radiotoxic agent, or one or more additional antibodies of the invention (e.g., an antibody having a complementary activity). Accordingly, patients treated with antibody compositions of the invention can be additionally administered (prior to, simultaneously with, or following administration of a antibody of the invention) with another therapeutic agent, such as a cytotoxic or radiotoxic agent, which enhances or augments the therapeutic effect of the antibodies of the invention. In other embodiments, the subject can be additionally treated with an agent that modulates, e.g., enhances or inhibits, the expression or activity of Fc-gamma or Fc-alpha receptors by, for example, treating the subject with a cytokine. Preferred cytokines include granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), interferon-γ (IFN-γ), and tumor necrosis factor (TNF). Other important agents for increasing the therapeutic efficacy of the antibodies and pharmaceutical compositions described herein are β-glucans which are homopolysaccharides of branched glucose residues and are produced by a variety of plants and microorganisms, for example, bacteria, algae, fungi, yeast and grains. Fragments of β-glucans produced by organisms may be also be used. Preferably, the β-glucan is a polymer of β(1,3) glucose wherein at least some of the backbone glucose units, e.g. 3-6% of the backbone glucose units, possess branches such as β(1,6) branches. In a particular embodiment, the invention provides methods for detecting the presence of CLDN6 antigen in a sample, or measuring the amount of CLDN6 antigen, comprising contacting the sample, and a control sample, with an antibody which specifically binds to CLDN6, under conditions that allow for formation of a complex between the antibody or portion thereof and CLDN6. The formation of a complex is then detected, wherein a difference complex formation between the sample compared to the control sample is indicative for the presence of CLDN6 antigen in the sample. In still another embodiment, the invention provides a method for detecting the presence or quantifying the amount of cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface in vivo or in vitro. The method comprises (i) administering to a subject a composition of the invention conjugated to a detectable marker; and (ii) exposing the subject to a means for detecting said detectable marker to identify areas containing cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface. Methods as described above are useful, in particular, for diagnosing CLDN6-related diseases and/or the localization of CLDN6-related diseases such as cancer diseases. Preferably an amount of CLDN6 in a sample which is higher than the amount of CLDN6 in a control sample is indicative for the presence of a CLDN6-related disease in a subject, in particular a human, from which the sample is derived. When used in methods as described above, an antibody described herein may be provided with a label that functions to: (i) provide a detectable signal; (ii) interact with a second label to modify the detectable signal provided by the first or second label, e.g. FRET (Fluorescence Resonance Energy Transfer); (iii) affect mobility, e.g. electrophoretic mobility, by charge, hydrophobicity, shape, or other physical parameters, or (iv) provide a capture moiety, e.g., affinity, antibody/antigen, or ionic complexation. Suitable as label are structures, such as fluorescent labels, luminescent labels, chromophore labels, radioisotopic labels, isotopic labels, preferably stable isotopic labels, isobaric labels, enzyme labels, particle labels, in particular metal particle labels, magnetic particle labels, polymer particle labels, small organic molecules such as biotin, ligands of receptors or binding molecules such as cell adhesion proteins or lectins, label-sequences comprising nucleic acids and/or amino acid residues which can be detected by use of binding agents, etc. Labels comprise, in a nonlimiting manner, barium sulfate, iocetamic acid, iopanoic acid, calcium ipodate, sodium diatrizoate, meglumine diatrizoate, metrizamide, sodium tyropanoate and radio diagnostic, including positron emitters such as fluorine-18 and carbon-11, gamma emitters such as iodine-123, technetium-99m, iodine-131 and indium-111, nuclides for nuclear magnetic resonance, such as fluorine and gadolinium. In yet another embodiment immunoconjugates of the invention can be used to target compounds (e.g., therapeutic agents, labels, cytotoxins, radiotoxins immunosuppressants, etc.) to cells which have CLDN6 associated with their surface by linking such compounds to the antibody. Thus, the invention also provides methods for localizing ex vivo or in vitro cells expressing CLDN6 and being characterized by association of CLDN6 with their cell surface, such as circulating tumor cells. The present invention is further illustrated by the following examples which are not be construed as limiting the scope of the invention. EXAMPLES The techniques and methods used herein are described herein or carried out in a manner known per se and as described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. All methods including the use of kits and reagents are carried out according to the manufacturer's information unless specifically indicated. Example 1: Quantification of CLDN6 Expression in Normal Tissues, Cancerous Tissues and Cell Lines Using Real-Time RT-PCR Total cellular RNA was extracted from frozen tissue specimens and cancer cell lines using RNeasy Mini Kit (Qiagen), primed with a dT18oligonucleotide and reverse-transcribed with Superscript II (GIBCO/Lifetech) according to the manufacturer's instructions. Integrity of the obtained cDNA was tested by amplification of p53 transcripts in a 30 cycle PCR. After normalization to HPRT expression of CLDN6 was quantified using ΔΔCT calculation. Tissues from three individuals were tested for each normal tissue type. Only trace amounts of CLDN6 transcripts could be detected in normal tissues after 40 cycles of RT-PCR. The only normal tissue slightly exceeding the expression cutoff was placenta. In contrast to normal tissues, we found high expression of CLDN6 in samples from ovarian cancer (adenocarcinomas), lung cancer (NSCLC, with highest frequency and expression levels in adenocarcinomas), gastric cancer, breast cancer, hepatic cancer, pancreatic cancer, skin cancer (basal cell carcinoma and squamous cell carcinoma), malignant melanoma, head and neck cancer (malignant pleomorphic adenoma), sarcoma (synovial sarcoma and carcinosarcoma), bile duct cancer, renal cell cancer (clear cell carcinoma and papillary carcinoma), uterine cancer and cancer cell lines A2780 (ovarian cancer), NIH-OVCAR3 (ovarian cancer), HCT-116 (colon cancer), EFO-27 (ovarian cancer), CPC-N(SCLC), NCI-H552 (NSCLC), SNU-1 (gastric cancer), KATOIII (gastric cancer), YAPC (pancreatic cancer), AGS (gastric cancer), FU97 (gastric cancer), MKN7 (gastric cancer). Example 2: Quantification of CLDN6 Expression in Normal Tissues, Cancerous Tissues and Cell Lines Using Western Blot Analysis For Western blot analysis 20 μg of total protein extracted from cells lyzed with Laemmli-lysis buffer was used. Extracts were diluted in reducing sample buffer (Roth), subjected to SDS-PAGE and subsequently electrotransferred onto PVDF membrane (Pall). Immunostaining was performed with polyclonal antibodies reactive to CLDN6 (ARP) and beta-Actin (Abcam) followed by detection of primary antibodies with horseradish-peroxidase conjugated goat anti-mouse and goat anti-rabbit secondary antibodies (Dako). Tissue lysates from up to five individuals were tested for each normal tissue type. No CLDN6 protein expression was detected in any of the normal tissues analyzed. In contrast to normal tissues, high expression of CLDN6 protein was detected in samples from ovarian cancer and lung cancer. CLDN6 expression was detected in NIH-OVCAR3 (ovarian cancer), MKN7 (gastric cancer), AGS (gastric cancer), CPC-N(SCLC), HCT-116 (colon cancer), FU97 (gastric cancer), NEC8 (testicular embryonal carcinoma), JAR (placental choriocarcinoma), JEG3 (placental choriocarcinoma), BEWO (placental choriocarcinoma), and PA-1 (ovarian teratocarcinoma). Example 3: Immunohistochemical (IHC) Analysis of CLDN6 Expression in Normal Tissues and Cancerous Tissues Paraffin-embedded tissue sections (4 μm) were incubated for 1 hour at 58° C. on a heating plate (HI 1220, Leica). Paraffin was removed from the sections by incubating the slides in Roticlear (Roth) for 2×10 min at RT. Afterwards the sections were rehydrated in graded alcohol (99%, 2×96%, 80% and 70%, 5 min each). Antigen retrieval was performed by boiling slides at 120° C. (15 psi) for 15 min in 10 mM citrate buffer (pH 6.0)+0.05% Tween-20. Directly after boiling slides were incubated in PBS for 5 min. Endogenous peroxidase activity was blocked with 0.3% hydrogen peroxide in MeOH for 15 min at RT. To avoid non-specific binding the slides were blocked with 10% goat serum in PBS for 30 min at RT. Thereafter, the slides were incubated with CLDN6-specific polyclonal antibody (1 μg/ml) (ARP) overnight at 4° C. On the next day the slides were washed with PBS at RT (3×5 min) and incubated with 100 μl of the secondary antibodies (PowerVision poly HRP-Anti-Rabbit IgG ready-to-use (ImmunoLogic)) for one hour at RT. Afterwards, slides were washed with PBS at RT (3×5 min). Final staining was performed by using the VECTOR NovaRED Substrate Kit SK-4800 from Vector Laboratories (Burlingame). Sections were counterstained with haematoxylin for 90 sec at RT. After dehydration with graded alcohol (70%, 80%, 2×96% and 99%, 5 min each) and 10 min incubation in xylol slides were mounted with X-tra Kit (Medite Histotechnic). No CLDN6 protein expression was detectable in normal tissues from lung, ovary, stomach, colon, pancreas, liver, duodenum or kidney. In contrast to normal tissues, strong or at least significant staining was observed on tissue sections from ovarian cancer, lung cancer, skin cancer, pancreatic cancer, gastric cancer, breast cancer, urinary bladder cancer (transitional cell carcinoma), cervical cancer, testicular cancer (seminoma) and uterine cancer. Staining was clearly accentuated at the plasma membrane of the malignant epithelial cell populations, whereas adjacent stromal and non-malignant epithelial cells were negative. These results indicate that CLDN6 protein is localized at the plasma membrane of malignant cells. Example 4: Generation of Murine Antibodies Against CLDN6 a. Generation of Expression Vectors Encoding Full Length CLDN6 and CLDN6 Fragments A non-natural, codon-optimized DNA sequence (SEQ ID NO: 3) encoding full length CLDN6 (NCBI accession number NP_067018.2, SEQ ID NO: 2) was prepared by chemical synthesis (GENEART AG, Germany) and cloned into the pcDNA3.1/myc-His vector (Invitrogen, USA) yielding the vector p3953. Insertion of a stop codon allowed the expression of CLDN6 protein without being fused to the vector encoded myc-His tag. Expression of CLDN6 was tested by Western blot, flow cytometry and immunofluorescence analyzes using commercially available anti-CLDN6 antibodies (ARP, 01-8865; R&D Systems, MAB3656). In addition, a codon-optimized DNA sequence (SEQ ID NO: 4) coding for the putative extracellular domain 2 (EC2) fragment of CLDN6 (SEQ ID NO: 6) as a fusion with an N-terminal Ig kappa leader derived signal peptide followed by 4 additional amino acids to ensure a correct signal peptidase cleavage site (SEQ ID NO: 5) was prepared and cloned into the pcDNA3.1/myc-His vector yielding the vector p3974. Prior to immunization, expression of the EC2 fragment was confirmed by immunofluorescence microscopy on transiently transfected and paraformaldehyde (PFA)-fixed CHO-K1 cells using a commercially available anti-myc antibody (Cell Signaling, MAB 2276). b. Generation of Cell Lines Stably Expressing CLDN6 HEK293 and P3X63Ag8U.1 cell lines stably expressing CLDN6 were generated by standard techniques using the vector p3953. c. Immunizations Balb/c mice were immunized with 25 μg of p3974 plasmid DNA together with 4 μl PEI-mannose (PEI-Man; in vivo-jetPEI™-Man from PolyPlus Transfection) (150 mM PEI-Man in H2O with 5% glucose) by intraperitoneal injection on days 0, 16 and 36. On days 48 and 62 mice were immunized by intraperitoneal injection with P3X63Ag8U.1 myeloma cells transfected with p3953 vector to stably express CLDN6. The cells administered on day 62 had been irradiated with 3000 rad prior to injection. The presence of antibodies directed against CLDN6 in sera of mice was monitored by immunofluorescence microscopy between days 20 and 70 using CHO-K1 cells co-transfected with nucleic acids encoding CLDN6 and GFP. To this end, 24 h following transfection, PFA-fixed or non-fixed cells were incubated with a 1:100 dilution of sera from immunized mice for 45 min at room temperature (RT). Cells were washed, incubated with an Alexa555-labeled anti-mouse Ig antibody (Molecular Probes) and subjected to fluorescence microscopy. Anti-CLDN6 specific antibodies were detected in serum samples obtained from a mouse on the basis of which the hybridoma F3-6C3-H8 was produced; seeFIG.2. For generation of monoclonal antibodies, mice with detectable anti-CLDN6 immune responses were boosted four days prior to splenectomy by intraperitonal injection of 2×107HEK293 cells stably transfected with p3953 vector. d. Generation of Hybridomas Producing Murine Monoclonal Antibodies Against CLDN6 6×107splenocytes isolated from an immunized mouse were fused with 3×107cells of the mouse myeloma cell line P3X63Ag8.653 (ATCC, CRL 1580) using PEG 1500 (Roche, CRL 10783641001). Cells were seeded at approximately 5×104cells per well in flat bottom microtiter plates and cultivated for about two weeks in RPMI selective medium containing 10% heat inactivated fetal bovine serum, 1% hybridoma fusion and cloning supplement (HFVCS, Roche, CRL 11363735), 10 mM HEPES, 1 mM sodium pyruvate, 4.5% glucose, 0.1 mM 2-mercaptoethanol, 1×penicillin/streptomycin and 1×HAT supplement (Invitrogen, CRL 21060). After 10 to 14 days, individual wells were screened by flow cytometry for anti-CLDN6 monoclonal antibodies. Antibody secreting hybridomas were subcloned by limiting dilution and again tested for anti-CLDN6 monoclonal antibodies. The stable subclones were cultured to generate small amounts of antibody in tissue culture medium for characterization. At least one clone from each hybridoma which retained the reactivity of the parent cells (tested by flow cytometry) was selected. Nine-vial-cell banks were generated for each clone and stored in liquid nitrogen. Example 5: Binding Characteristics of Hybridoma Supernatants and Monoclonal Antibodies a. Quality Control of Transiently Transfected HEK293T Cells by (i) Western Blot and (ii) Flow Cytometry Analyzes (i) HEK293T cells were transfected with nucleic acids encoding CLDN3, CLDN4, CLDN6, and CLDN9, respectively, or mock-transfected. Expression of CLDN3, CLDN4, CLDN6 or CLDN9 in HEK293T cells was determined by Western blotting. To this end, cells were harvested 24 hours post transfection and subjected to lysis. The lysate was subjected to SDS-PAGE, blotted onto nitrocellulose membrane and stained with anti-CLDN3(A) (Invitrogen, 34-1700), anti-CLDN4(A) (Zymed, 32-9400), anti-CDLN6(A) (ARP, 01-8865) or anti-CLDN9(A) (Santa Cruz, sc-17672) antibodies which specifically bind to the C-terminus of the corresponding claudin under denaturing conditions. Following incubation with a peroxidase-labeled secondary antibody and developing with ECL reagent, a LAS-3000 imager (Fuji) was used for visualization. Bands of the expected molecular weights of CLDN3, CLDN4, CLDN6 and CLDN9, respectively, were observed only in the transfected cells but not in the control cells (FIG.3) demonstrating that HEK293T cells do not endogenously express any of the claudins investigated and thus, are a suitable tool for determining the cross reactivity of CLDN6 antibodies. (ii) The HEK293T cells of (i) were further analyzed by flow cytometry using anti-CLDN antibodies recognizing native epitopes (mouse anti-CLDN3 IgG2a (R&D, MAB4620), mouse anti-CLDN4 IgG2a (R&D, MAB4219), mouse anti-CLDN6 IgG2b (R&D, MAB3656)). The antibodies obtainable from Sigma under the product numbers M9144 and M8894 served as isotype controls. Specificity of these anti-CLDN antibodies was analyzed using HEK293T cells transiently transfected with nucleic acids encoding CLDN3, CLDN4, CLDN6, and CLDN9, respectively. The anti-CLDN6 antibody shows cross-reactivity with CLDN3, CLDN4 and CLDN9. The anti-CLDN4 antibody shows cross-reactivity with CLDN3, CLDN6 and CLDN9. The anti-CLDN3 antibody binds specifically to CLDN3 (FIG.4). b. Determination of the Specificity of Monoclonal Antibodies Produced According to the Invention Using Flow Cytometry HEK293T cells were co-transfected with a vector encoding different CLDN proteins and a vector encoding a fluorescence marker. 24 h post transfection cells were harvested using 0.05% trypsin/EDTA solution and washed with FACS buffer (PBS containing 2% FCS and 0.1% sodium azide). Cells were transferred into U-bottom microtiter plates at 2×105cells per well and incubated for 60 min at 4° C. with hybridoma supernatants. Following washing three times with FACS buffer, cells were incubated with an allophycocyanin (APC)-conjugated anti-mouse IgG 1+2a+2b+3 specific secondary antibody (Dianova, 115-135-164). Thereafter, cells were washed twice and binding was assessed by flow cytometry using a BD FACSArray (FIG.5). The expression of the fluorescence marker is plotted on the horizontal axis against the antibody binding on the vertical axis. A commercially available mouse anti-CLDN6 IgG2b antibody (R&D, MAB3656) served as a positive control and the antibody obtainable from Sigma under the product number M8894 served as an isotype control. Antibodies in the supernatants from the monoclonal hybridoma subclones F3-6C3-H2, F3-6C3-H8, F3-6C3-H9, F3-6C3-D8 and F3-6C3-G4, all derived from hybridoma F3-6C3, were specific for CLDN6 and did not bind to CLDN9, CLDN3 and CLDN4.FIG.5Aexemplarily shows the results for the monoclonal hybridoma subclone F3-6C3-H8. Antibodies in the supernatant from the monoclonal hybridoma subclone F3-6C3-H8 also bind to cells transfected with the (1143V)-SNP variant of CLDN6. Antibodies in the supernatant from the monoclonal hybridoma subclone F4-4F7-F2 bind to both CLDN6 and CLDN9 (FIG.5A). Antibodies in the supernatant from the monoclonal hybridoma subclone F3-7B3-B4 bind to CLDN6, CLDN3 and CLDN9 (FIG.5B). Antibodies in the supernatant from the monoclonal hybridoma subclone F3-3F7-A5 bind to CLDN6, CLDN4 and CLDN9 (FIG.5B). Example 6: Generation and Testing of Monoclonal Antibodies Against CLDN6 a. Generation of Expression Vectors Encoding the Extracellular Domain 1 of CLDN6 A codon-optimized DNA sequence (SEQ ID NO: 12) coding for the putative extracellular domain 1 (EC1) fragment of CLDN6 (SEQ ID NO: 7) as a fusion with an N-terminal Ig kappa leader derived signal peptide followed by 4 additional amino acids to ensure a correct signal peptidase cleavage site (SEQ ID NO: 13) was prepared and cloned into the pcDNA3.1/myc-His vector yielding the vector p3973. Prior to immunization, expression of the EC1 fragment was confirmed by immunofluorescence microscopy on transiently transfected and paraformaldehyde (PFA)-fixed CHO-K1 cells using a commercially available anti-myc antibody (Cell Signaling, MAB 2276). b. Immunization Balb/c mice were immunized with 25 μg of p3973 plasmid DNA together with 4 μl PEI-mannose (PEI-Man; in vivo-jetPEI™-Man from PolyPlus Transfection) (150 mM PEI-Man in H2O with 5% glucose) by intraperitoneal injection on days 0 and 14. On days 28 and 44 mice were immunized subcutaneously with KLH-conjugated peptides SEQ ID NO: 14 and SEQ ID NO: 15 (100 μg each in PBS, JPT Peptide Technologies GmbH, Germany) together with HPLC-purified PTO-CpG-ODN (25 μg in PBS; 5′-TCCATGACGTTCCTGACGTT; Eurofins MWG Operon, Germany). On days 64, 77 and 97 mice were immunized by intraperitoneal injection with 2×107P3X63Ag8U.1 myeloma cells transfected with p3953 vector to stably express CLDN6. Prior to administration, cells were treated with mitomycin-C (2.5 μg/ml, Sigma-Aldrich, M4287). On days 64 and 97 cells were administered together with HPLC-purified PTO-CpG-ODN (50 μg in PBS), on day 77 together with incomplete Freund's adjuvant. For generation of monoclonal antibodies, mice with detectable anti-CLDN6 immune responses were boosted four days prior to splenectomy by intraperitonal injection of 2×107HEK293 cells stably transfected with p3953 vector. c. Testing of Monoclonal Antibodies Against CLDN6 Flow Cytometry To test the binding of monoclonal antibodies to CLDN6 and its homologous HEK293T cells were transiently transfected with the corresponding claudin-coding plasmid and the expression was analyzed by flow cytometry. In order to differentiate between transfected and non-transfected cells, HEK293T cells were co-transfected with a fluorescence marker as a reporter. 24 h post transfection cells were harvested with 0.05% trypsin/EDTA, washed with FACS buffer (PBS containing 2% FCS and 0.1% sodium azide) and resuspended in FACS buffer at a concentration of 2×106cells/ml. 100 μl of the cell suspension were incubated with the appropriate antibody at indicated concentrations for 30 min at 4° C. A cross-reactive antibody was used to detect CLDN6 and CLDN9 expression. The commercially available mouse anti-claudin antibodies anti-CLDN3 (R&D, MAB4620) and anti-CLDN4 (R&D, MAB4219) served as positive controls, whereas mouse IgG2a (Sigma, M9144) and IgG2b (Sigma, M8894), respectively, served as isotype control. The cells were washed three times with FACS buffer and incubated with an APC-conjugated anti-mouse IgG 1+2a+2b+3a specific secondary antibody (Dianova, 115-135-164) for 30 min at 4° C. The cells were washed twice and resuspended in FACS buffer. The binding was analyzed by flow cytometry using a BD FACSArray. The expression of the fluorescence marker was plotted on the horizontal axis against the antibody binding on the vertical axis. CDC The complement dependent cytotoxicity (CDC) was determined by measuring the content of intracellular ATP in non-lysed cells after the addition of human complement to the target cells incubated with anti-CLDN6 antibodies. As a very sensitive analytical method the luminescent reaction of luciferase was used for measuring ATP. CHO-K1 cells stably transfected with CLDN6 (CHO-K1-CLDN6) were harvested with 0.05% trypsin/EDTA, washed twice with X-Vivo 15 medium (Lonza, BE04-418Q) and suspended at a concentration of 1×107cells/ml in X-Vivo 15 medium. 250 μl of the cell suspension were transferred into a 0.4 cm electroporation cuvette and mixed with 7 μg of in vitro transcribed RNA encoding for luciferase (luciferase IVT RNA). The cells were electroporated at 200 V and 300 μF using a Gene Pulser Xcell (Bio Rad). After electroporation, the cells were suspended in 2.4 ml prewarmed D-MEM/F12 (1:1) with GlutaMax-I medium (Invitrogen, 31331-093) containing 10% (v/v) FCS, 1% (v/v) penicillin/streptomycin and 1.5 mg/ml G418. 50 μl of the cell suspension per well were seeded into a white 96-well PP-plate and incubated at 37° C. and 7.5% CO2. 24 h post electroporation 50 μl monoclonal murine anti-CLDN6 antibodies in 60% RPMI (containing 20 mM HEPES) and 40% human serum (serum pool obtained from six healthy donors) were added to the cells at indicated concentrations. 10 μl 8% (v/v) Triton X-100 in PBS per well were added to total lysis controls, whereas 10 μl PBS per well were added to max viable cells controls and to the actual samples. After an incubation of 80 min at 37° C. and 7.5% CO250 μl luciferin mix (3.84 mg/ml D-luciferin, 0.64 U/ml ATPase and 160 mM HEPES in ddH2O) were added per well. The plate was incubated in the dark for 45 min at RT. The luminescence was measured using a luminometer (Infinite M200, TECAN). Results are given as integrated digital relative light units (RLU). NEC8 cells were electroporated at 200 V and 400 μF and cultivated in RPMI 1640 with GlutaMAX-I medium (Invitrogen, 61870) containing 10% (v/v) FCS. The specific lysis is calculated as: specific⁢⁢lysis⁢[%]=100-[(sample-total⁢⁢lysis)(max⁢⁢viable⁢⁢cells-total⁢⁢lysis)×100]max viable cells: 10 μl PBS, without antibodytotal lysis: 10 μl 8% (v/v) Triton X-100 in PBS, without antibody Early Treatment For early antibody treatments 2×107NEC8 cells in 200 μl PBS were subcutaneously inoculated into the flank of athymic Nude-Foxn1numice. Each experimental group consisted of ten 6-8 week-old female mice. Three days after inoculation 200 μg of purified murine monoclonal antibodies muMAB 59A, 60A, 61D, 64A, 65A, 66B and 67A were applied for 46 days by alternating intravenous and intraperitoneal injections twice a week. Experimental groups treated with PBS served as a negative controls. The tumor volume (TV=(length×width2)/2) was monitored bi-weekly. TV is expressed in mm3, allowing construction of tumor growth curves over time. When the tumor reached a volume greater than 1500 mm3mice were killed. d. Results Murine monoclonal antibodies muMAB 59A, 60A, 61D, 64A, 65A, 66B and 67A showed strong binding to human CLDN6 and the CLDN6 SNP (single nucleotide polymorphism) variant I143V while no binding to CLDN3, 4, and 9 was observed (FIG.6). MuMAB 59A, 60A, 61D, 64A, 65A, 66B and 67A exhibited very low EC50 values (EC50 200-500 ng/ml) and saturation of binding was achieved at low concentrations (FIG.7). MuMAB 59A, 60A, 61D, 64A, 65A, 66B and 67A exhibited dose-dependent CDC activity and induced CDC at low concentrations (FIG.8). The anti-CLDN6 antibodies muMAB 65A and 66B induced CDC on NEC8 cells in a dose dependent manner (FIG.9). Target specificity of muMAB 65A and 66B was proved by using NEC8 LVTS2 54 cells (CLDN6 knock-down). Furthermore, muMAB 59A, 60A, 61D, 64A, 65A, 66B and 67A showed tumor growth inhibition in mice engrafted with NEC8 cells (FIG.10). Example 7: Generation and Testing of Chimeric Monoclonal Antibodies Against CLDN6 a. Generation of Mouse/Human Chimeric Monoclonal Antibodies For chimerization, the murine heavy chain and light chain variable region including leader sequences were amplified by PCR using primers listed in the table below. The murine heavy chains were fused by an ApaI restriction site (5′-GGGCCC-3′) to the N-terminal part of the human Fcgamma1 chain, which was encoded by the expression vector. Variable domains of the murine kappa chain including leader sequences were cloned in front of the constant region using a BsiWI restriction site. The correct orientation of the constant region in the vector, i.e. suitable for the preceeding promoter of the vector, was verified by sequencing. Due to the position of the ApaI restriction site, any amplification of a variable region including leader sequence for this purpose has to include the first 11 nucleotides of the sequence of the human gamma-1 constant region in addition to the sequence of the ApaI site. The nucleotide sequence of human gamma-1 heavy chain constant region is listed as SEQ ID NO: 24, the amino acid sequence of the thus expressed human gamma-1 constant region is listed as SEQ ID NO: 25. The nucleotide sequence encoding the constant part of the kappa light chain is listed as SEQ ID NO: 26, the respective amino acid sequence is listed as SEQ ID NO: 27. TABLE 1Mouse hybridoma cell lines used for antibody cloningPrimermuMABIsotypeSEQ ID NOs:heavey chain64AIgG2a17, 1889AIgG2a17, 1961DIgG2a17, 2067AIgG2a17, 20light chain64AIgK21, 2289AIgK21, 2361DIgK21, 2267AIgK21, 22 Corresponding to their murine counterparts the chimeric monoclonal antibodies were named adding the prefix “chim”, e.g. chimAB 64A. Amplification of the murine variable regions of light and heavy chains including leader sequences was carried out according to the “step-out PCR” method described in Matz et al. (Nucleic Acids Research, 1999, Vol. 27, No. 6). For this, total RNA was prepared from monoclonal hybridoma cell lines (see Tab. 1) by standard methods known to those skilled in the art, for example with the use of RNeasy Mini Kit (Qiagen). Single stranded cDNA was prepared according to the “template-switch” method also described in Matz et al. (Nucleic Acids Research, 1999, Vol. 27, No. 6, 1558). In addition to a (dT)30 oligomer (SEQ ID NO: 28), it included a DNA/RNA hybrid oligomer (SEQ ID NO: 29) serving as an 5′ adaptor for template switching during polymerization of the cDNA strand. In this adaptor oligomer the last three nucleotides were ribo- instead of deoxyribonucleotides. The subsequent “step-out PCR” used an antisense oligomer targeted to the constant region of the mouse kappa chain or to the constant region of the subclass 2a of the gamma chain (SEQ ID NO: 30 and 31, respectively). The IgG subclass of the murine monoclonal antibody produced by the hybridoma cell lines was afore immunologically analyzed with IsoStrip (Roche), and the appropriate antisense oligomer was chosen accordingly (see Tab. 1). A primer mix served as the sense oligomer in the “step-out PCR”, comprising the two oligomers listed in SEQ ID NO: 32 and 33. The identified murine variable regions including leader sequences were then amplified by PCR omitting the 5′ UTR and the 3′ mouse constant region, adding restriction sites to the ends which allowed subcloning into the prepared expression vectors carrying the human constant regions. In addition, the sense oligomers provided a consensus Kozak sequence (5′-GCCGCCACC-3′) and the antisense oligomers for heavy chain variable regions included the first 11 nucleotides of the human gamma-1 constant region in addition to the ApaI restriction site (see Tab. 1, SEQ ID NOs: 17 to 23). Kappa light chain variable regions including leader sequences were cloned using HindIII and BsiWI restriction enzymes, gamma heavy chain variable regions demanded HindIII and ApaI restriction enzymes. Further murine variable regions of light and heavy chains including leader sequences were amplified and further chimeric monoclonal antibodies against CLDN6 generated in accordance to the protocol disclosed above. b. Production of Chimeric Monoclonal Anti-CLDN6 Antibodies Chimeric monclonal antibodies were transiently expressed in HEK293T cells (ATCC CRL-11268) transfected with plasmid DNA coding for the light and heavy chains of the corresponding antibody. 24 h before transfection 8×107cells were seeded on 145 mm cell culture plates and cultivated in 25 ml HEK293T-medium (DMEM/F12+GlutaMAX-I, 10% FCS, 1% penicillin/streptomycin). 20 μg plasmid DNA were dissolved in 5 ml HEK293T-medium without supplements per cell culture plate. After adding 75 μl linear polyethylenimine (PEI) (1 mg/ml) (Polyscience, 23966) the (DNA:PEI)-mixture was incubated 15 min at RT. Thereafter, the transfection-mix was added dropwise to the cells. 24 h post transfection the HEK293T-medium was replaced with Pro293a-medium (Lonza, BE12-764Q) containing 1% penicillin/streptomycin. For optimal expression, the transfected cells were cultivated at 37° C. and 7.5% CO2for additional 96 to 120 h. The supernatant was harvested and the chimeric antibody was purified by FPLC using protein-A columns. The concentration of the antibody was determined and quality was tested by SDS-PAGE. c. Testing of Chimeric Monoclonal Antibodies Against CLDN6 Flow Cytometry To test the specificities and affinities of CLDN6-specific chimeric monoclonal antibodies binding to HEK293 cells stably transfected with CLDN3, 4, 6 or 9, respectively, and tumor cell lines that endogenously express CLDN6 was analyzed by flow cytometry. Therefore, cells were harvested with 0.05% Trypsin/EDTA, washed with FACS buffer (PBS containing 2% FCS and 0.1% sodium azide) and resuspended in FACS buffer at a concentration of 2×106cells/ml. 100 μl of the cell suspension were incubated with the appropriate antibody at indicated concentrations for 60 min at 4° C. A chimeric cross-reactive antibody (chimAB 5F2D2) was used to detect CLDN6 and CLDN9 expression. The commercially available mouse anti-claudin antibodies anti-CLDN3 (R&D, MAB4620) and anti-CLDN4 (R&D, MAB4219) served as positive controls, whereas human IgG1-kappa (Sigma, 15154) served as a negative control. The cells were washed three times with FACS buffer and incubated for 30 min at 4° C. with an APC-conjugated goat anti-human IgG Fc-gamma (Dianova, 109-136-170) or an APC-conjugated anti-mouse IgG 1+2a+2b+3a (Dianova, 115-135-164) specific secondary antibody, respectively. The cells were washed twice and resuspended in FACS buffer. The binding was analyzed by flow cytometry using a BD FACSArray. CDC The complement dependent cytotoxicity (CDC) was determined by measuring the content of intracellular ATP in non-lysed cells after the addition of human complement to the target cells incubated with anti-CLDN6 antibodies. As a very sensitive analytical method the bioluminescent reaction of luciferase is used for measuring ATP. In this assay, NEC8 wildtype cells (CLDN6 positive) and NEC8 CLDN6 knock-down cells (CLDN6 negative) were used which both were stably transduced with luciferase expression construct. The cells were harvested with 0.05% Trypsin/EDTA and adjusted to a concentration of 2×105cells/ml in RPMI with GlutaMax-I medium (Invitrogen, 61870-010) containing 10% (v/v) FCS. 1×104cells were seeded into a white 96-well PP-plate and incubated for 24 h at 37° C. and 5% CO2. After incubation, 50 μl monoclonal chimeric anti-CLDN6 antibodies in 60% RPMI (containing 20 mM HEPES) and 40% human serum (serum pool obtained from six healthy donors) were added to the cells at indicated concentrations. 10 μl 8% (v/v) Triton X-100 in PBS per well were added to total lysis controls, whereas 10 μl PBS per well were added to max viable cells controls and to the actual samples. After a further incubation of 80 min at 37° C. and 5% CO250 μl luciferin mix (3.84 mg/ml D-luciferin, 0.64 U/ml ATPase and 160 mM HEPES in ddH2O) was added per well. The plate was incubated in the dark at RT for 45 min. The bioluminescence was measured using a luminometer (Infinite M200, TECAN). Results are given as integrated digital relative light units (RLU). The specific lysis is calculated as: specific⁢⁢lysis⁢[%]=100-[(sample-total⁢⁢lysis)(max⁢⁢viable⁢⁢cells-total⁢⁢lysis)×100]max viable cells: 10 μl PBS, without antibodytotal lysis: 10 μl 8% (v/v) Triton X-100 in PBS, without antibody ADCC The antibody dependent cellular cytotoxicity (ADCC) was determined by measuring the content of intracellular ATP in non-lysed cells after the addition of human PBMC to the target cells incubated with anti-CLDN6 antibodies. As a very sensitive analytical method the bioluminescent reaction of luciferase is used for measuring ATP. In this assay, NEC-8 wildtype cells (CLDN6 positive) and NEC-8 CLDN6 knock-down cells (CLDN6 negative) were used which both were stably transduced with luciferase expression construct. The cells were harvested with 0.05% Trypsin/EDTA and adjusted to a concentration of 2×105cells/ml in RPMI with GlutaMax-I medium (Invitrogen, 61870-010) containing 10% (v/v) FCS and 20 mM Hepes. 1×104cells were seeded into a white 96-well PP-plate and incubated 4 h at 37° C. and 5% CO2. PBMC were isolated from human donor blood samples by density gradient centrifugation using Ficoll Hypaque (GE Healthcare, 17144003). The PMBC containing interphase was isolated and cells were washed twice with PBS/EDTA (2 mM). 1×10′ PBMC were seeded in 50 ml X-Vivo 15 medium (Lonza, BE04-418Q) containing 5% heat-inactivated human serum (Lonza, US14-402E) and incubated for 2 h at 37° C. and 5% C02. 4 h post seeding of the target cells (NEC-8) 25 μl monoclonal chimeric anti-CLDN6 antibodies in PBS were added to the cells at indicated concentrations. Nonadherent PBMC, which separated within the 2 h incubation from adherent monocytes, were harvested and adjusted to 8×106cells/ml in X-vivo 15 medium. 25 μl of this cell suspension was added to the target cells and the monoclonal chimeric anti-CLDN6 antibodies. The plates were incubated for 24h at 37° C. and 5% CO2. After the 24 h incubation 10 μl 8% (v/v) Triton X-100 in PBS per well were added to total lysis controls, whereas 10 μl PBS per well were added to max viable cells controls and to the actual samples. 50 μl luciferin mix (3.84 mg/ml D-luciferin, 0.64 U/ml ATPase and 160 mM HEPES in ddH2O) was added per well. The plate was incubated in the dark at RT for 30 min. The bioluminescence was measured using a luminometer (Infinite M200, TECAN). Results are given as integrated digital relative light units (RLU). The specific lysis is calculated as: specific⁢⁢lysis⁢[%]=100-[(sample-total⁢⁢lysis)(max⁢⁢viable⁢⁢cells-total⁢⁢lysis)×100]max viable cells: 10 μl PBS, without antibodytotal lysis: 10 μl 8% (v/v) Triton X-100 in PBS, without antibody d. Results Anti-CLDN6 chimeric monoclonal antibodies chimAB 61D, 64A, 67A and 89A showed strong binding to human CLDN6 while no binding to CLDN3, 4, and 9 was observed (FIG.11). Regarding binding to human CLDN6 stably expressed on the surface of HEK293 cells, anti-CLDN6 chimeric monoclonal antibodies chimAB 64A and 89A exhibit very low EC50 values (EC50 450-600 ng/ml) and saturation of binding was achieved at low concentrations. ChimAB 67A and 61D showed low (EC50 1000 ng/ml) and medium (EC50 2300 ng/ml) EC50 values, respectively (FIG.12). Regarding binding to CLDN6 endogenously expressed in NEC8 cells, anti-CLDN6 chimeric monoclonal antibodies chimAB 64A and 89A exhibited very low EC50 values (EC50 600-650 ng/ml) and saturation of binding was achieved at low concentrations, whereas chimAB 61D and 67A showed medium (EC50 1700 ng/ml) and high (EC50 6100 ng/ml) EC50 values, respectively (FIG.13). Regarding binding to CLDN6 endogenously expressed in OV90 cells, anti-CLDN6 chimeric monoclonal antibodies chimAB 64A and 89A exhibited very low EC50 values (EC50 550-600 ng/ml) and saturation of binding was achieved at low concentrations. ChimAB 61D and 67A showed medium EC50 values (EC50 1500 ng/ml and EC50 2300 ng/ml, respectively) (FIG.14). Anti-CLDN6 chimeric monoclonal antibodies chimAB 61D, 64A, 67A and 89A exhibited CDC activity in a dose-dependent manner on NEC-8 cells (FIG.15). Anti-CLDN6 chimeric monoclonal antibodies chimAB 61D, 64A, 67A and 89A exhibited dose-dependent ADCC activity on NEC-8 cells and induced ADCC even at low antibody concentrations (FIG.16). These results clearly show the specificity of these chimeric monoclonal antibodies for CLDN6. Example 8: Treatment Using Monoclonal Antibodies Against CLDN6 Early Treatment For early antibody treatments 2×107NEC8 cells in 200 μl RPMI medium (Gibco) were subcutaneously inoculated into the flank of athymic Nude-Foxn1numice. Each experimental group consisted of ten 6-8 week-old female mice. Three days after tumor cell inoculation 200 μg of purified murine monoclonal antibody muMAB 89A was applied for seven weeks by alternating intravenous and intraperitoneal injections twice a week. Experimental group treated with PBS served as negative control. The tumor volume (TV=(length x width2)/2) was monitored bi-weekly. TV is expressed in mm3, allowing construction of tumor growth curves over time. When the tumors reached a volume greater than 1500 mm3mice were sacrificed. Advanced Treatments For antibody treatments of advanced xenograft tumors 2×107NEC8 cells in 200 μl RPMI medium (Gibco) were subcutaneously inoculated into the flank of 6-8 week-old female athymic Nude-Foxn1numice. The tumor volume (TV=(length x width2)/2) was monitored bi-weekly. TV is expressed in mm3, allowing construction of tumor growth curves over time. 15 to 17 days after tumor cell inoculation mice were divided into treatment groups of eight animals per cohorte with homogenous tumor sizes of above 80 mm3. 200 μg of purified murine monoclonal antibodies muMAB 61D, 64A, 67A and 89A were applied for five weeks by alternating intravenous and intraperitoneal injections twice a week. Experimental groups treated with PBS and an unspecific antibody served as negative controls. When the tumors reached a volume bigger than 1500 mm3mice were sacrificed. In an early treatment xenograft model using mice engrafted with the tumor cell line NEC8, mice treated with murine monoclonal antibodies muMAB 61D, 64A and 67A did not show any tumor growth even after stopping the immunotherapy (FIG.17). In an early treatment xenograft model using mice engrafted with the tumor cell line NEC8, muMAB 89A showed tumor growth inhibition and no tumors were detectable in mice treated with muMAB89A at the end of the study (FIG.18). In an advanced treatment xenograft model using mice engrafted with the tumor cell line NEC8, muMAB 64A showed an inhibition of tumor growth (FIG.19). In an advanced treatment xenograft model using mice engrafted with the tumor cell line NEC8, mice treated with muMAB 64A showed prolonged survival (FIG.20). In an advanced treatment xenograft model using mice engrafted with the tumor cell line NEC8, inhibition of tumor growth was achieved with the murine monoclonal anti-CLDN6 antibodies muMAB 61D, 67A and 89A (FIG.21). In an advanced treatment xenograft model using mice engrafted with the tumor cell line NEC8, mice treated with the CLDN6 specific antibody muMAB 61D or 67A showed prolonged survival (FIG.22). In an advanced treatment xenograft model using mice engrafted with NEC8 wildtype and NEC8 cells with a stable CLDN6 knock-down, muMAB 64A and 89A only show a therapeutic effect in mice engrafted with NEC8 wildtype but not in mice engrafted with NEC8 CLDN6 knock-down cells demonstrating CLDN6-specificity of the antibody in vivo (FIG.23). Example 9: High-Resolution Eitope Mapping of Monoclonal Antibodies Against CLDN6 The CLDN6 specific monoclonal antibodies only show very weak (if any) binding to linear peptides in ELISA epitope-mapping studies, implying that their epitopes are conformational. To analyze the interaction between antibodies described herein and CLDN6 in its native conformation site-directed mutagenesis in mammalian cell culture was used as an epitope-mapping technique. Alanine scanning mutagenesis of amino acids 27-81 and 137-161 within the first and second extracellular domain, respectively, was performed. Following transient expression in HEK293T cells, CLDN6 mutants were assessed for their ability to be bound by specific monoclonal antibodies. Impaired binding of a specific monoclonal antibody to a CLDN6 mutant suggest that the mutated amino acid is an important contact and/or conformational residue. The binding was analyzed by flow cytometry. To discriminate between transfected and non-transfected cell populations, cells were co-transfected with a fluorescence marker. The amino acid residues of CLDN6 that are important for the interaction with CLDN6 specific chimeric antibodies have been systematically identified by alanine-scanning. Alanine and glycine mutations were generated by site-directed mutagenesis (GENEART AG, Germany). To test the binding of monoclonal chimeric antibodies to wild-type CLDN6 and its mutants HEK293T cells were transiently transfected with the corresponding claudin-coding plasmid and the expression was analyzed by flow cytometry. In order to differentiate between transfected and non-transfected cells, HEK293T cells were co-transfected with a fluorescence marker as a reporter. 24 h post transfection cells were harvested with 0.05% Trypsin/EDTA, washed with FACS buffer (PBS containing 2% FCS and 0.1% sodium azide) and resuspended in FACS buffer at a concentration of 2×106cells/ml. 100 μl of the cell suspension were incubated with 10 μg/ml antibody for 45 min at 4° C. The commercially available mouse anti-CLDN6 (R&D, MAB3656) was used as a control to detect cell-surface expression of CLDN6 mutants. The cells were washed three times with FACS buffer and incubated with an APC-conjugated goat anti-human IgG Fc-gamma (Dianova, 109-136-170) or an APC-conjugated anti-mouse IgG 1+2a+2b+3a specific secondary antibody (Dianova, 115-135-164) for 30 min at 4° C. The cells were washed twice and resuspended in FACS buffer. The binding within the transfected cell population was analyzed by flow cytometry using a BD FACSArray. Therefore, the expression of the fluorescence marker was plotted on the horizontal axis against the antibody binding on the vertical axis. The average signal intensity of a monoclonal chimeric CLDN6 specific antibody bound to mutant CLDN6 was expressed as the percentage of wild-type binding. Amino acids that are essential for binding showed no binding after being mutated whereas amino acids that support binding only showed reduced binding compared to wild-type. High resolution epitope-mapping demonstrated that the amino acids F35, G37, S39 and possibly T33 of the first extracellular domain of CLDN6 are important for the interaction with the CLDN6 specific chimeric antibodies chimAB 61D, 64A, 67A and 89A. Residue 140 is essential for the binding of chimAB 89A and it contributes to the binding of chimAB 61D and 67A. In addition, L151 of the second extracellular domain of CLDN6 is important for the interaction with chimAB 67A (FIG.24).
195,794
11858989
DETAILED DESCRIPTION OF THE INVENTION The present disclosures are in part based on the development of monoclonal antibodies that target AQP4. As may be understood from the earlier discussion, the present invention includes a variety of aspects, which may be combined in different ways. The following descriptions are provided to list elements and describe some of the embodiments of the present invention. These elements are listed with initial embodiments, however it should be understood that they may be combined in any manner and in any number to create additional embodiments. The variously described examples and preferred embodiments should not be construed to limit the present invention to only the explicitly described systems, techniques, and applications. Further, this description should be understood to support and encompass descriptions and claims of all the various embodiments, systems, techniques, methods, devices, and applications with any number of the disclosed elements, with each element alone, and also with any and all various permutations and combinations of all elements in this or any subsequent application. The terms “a”, “an” and “the” as used herein are defined to mean “one or more” and include the plural unless the context is inappropriate. As used herein, the term “epitope” refers to any antigenic determinant on an antigen to which the paratope of an antibody binds. As used herein, the term “amino acid” includes all of the naturally occurring amino acids as well as modified amino acids. As used herein, the terms “polypeptide”, “peptide”, and “protein”, as used herein, are interchangeable and are defined to mean a biomolecule composed of amino acids linked by a peptide bond. As used herein, the term “hybridoma” is art recognized and is understood by those of ordinary skill in the art to refer to a cell produced by the fusion of an antibody-producing cell and an immortal cell, e.g. a multiple myeloma cell. Such a hybrid cell is capable of producing a continuous supply of antibody. See the definition of “monoclonal antibody” above and the Examples below for a more detailed description of one art known method of fusion. As used herein, the term “antibody” is intended to include monoclonal antibodies, polyclonal antibodies, and chimeric antibodies. The antibody may be from recombinant sources and/or produced in transgenic animals. The term “antibody fragment” as used herein is intended to include without limitations Fab, Fab′, F(ab′).sub.2, scFv, dsFv, ds-scFv, dimers, minibodies, diabodies, and multimers thereof, multispecific antibody fragments and Domain Antibodies. Antibodies can be fragmented using conventional techniques. As used herein, the term “monoclonal antibody” is also well recognized in the art and refers to an antibody that is mass produced in the laboratory from a single clone and that recognizes only one antigen. As used herein, the term “therapeutically effective amount” refers to the amount of antibody which, when administered to a human or animal, elicits an immune response which is sufficient to result in a therapeutic effect in said human or animal. The effective amount is readily determined by one of ordinary skill in the art following routine procedures. The present disclosure uses extracellular domains of AQP4 peptide to induce immune response and establish B cell clone by cell fusion to produce high specificity and affinity AQP4 antibody. The present disclosure successfully selects B cell clone that can produces peptide-specific AQP4 antibodies having high specificity and affinity. These peptide-specific AQP4 antibodies play a role to create a NMO model and contribute for investigating the NMO disease mechanisms and developing the strategy of the treatment. The present disclosure provides a peptide epitope, comprising a peptide having an amino acid sequence TPPSVVGGLGVTTVHGNLTC (SEQ ID NO: 1) or CSMNPARSFGPAVIMGNWANH (SEQ ID NO:2). The epitope can further links to KLH through —SH— bond. Particularly, the epitope has an amino acid sequence of CKLH-(SH)-TPPSVVGGLGVTTVHGNLTC (CKLH-(SH)-SEQ ID NO: 1; mAQP4-Loop) or EKLH-(SH)-CSMNPARSFGPAVIMGNWANH (EKLH-(SH-SEQ ID NO: 2; mAQP4-Loop). An immunogenic epitope of a polypeptide is a part of the polypeptide, which elicits an immune response in an animal or a human being, and/or in a biological sample determined by any of the biological assays. A common feature of the polypeptides of the present disclosures is their capability to induce an immunological response as illustrated in the examples. It is understood that a variant of a polypeptide of the invention produced by substitution, insertion, addition or deletion is also immunogenic determined by any of the assays. The present disclosures provide antibodies or antigen-binding fragments or molecules that specifically bind to AQP4 peptide and extracellular domains thereof. These anti-AQP4 agents are capable of treating and/or preventing NMO disease. Examples of the antibodies include, but are not limited to, polyclonal antibody, monoclonal antibody or chimeric antibody or antigen-binding fragments thereof. In one embodiment, the antibody is an monoclonal antibody produced by the hybridoma cell line AQP002 deposited at National Institute of Technology and Evaluation (NITE), Tokyo, Japan under the deposit number NITE BP-02882. The antibody specifically binds to the peptide epitope with the sequence of SEQ ID NO:2. Some embodiments of the invention are directed to modified antibodies that are based on or modified from the mouse anti-AQP4 antibodies exemplified herein. These include, e.g., chimeric, humanized and human anti-AQP4 antibodies. Relative to the exemplified antibody, these modified antibodies can have similar binding specificity, as well as improved binding affinity. They also have substantially reduced antigenicity when used in vivo in a non-mouse subject, e.g., a human subject. Some of the modified antibodies are chimeric antibodies which contain partial human immunoglobulin sequences (e.g., constant regions) and partial non-human immunoglobulin sequences. Some other modified antibodies are humanized antibodies. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source that is non-human. The methods can be readily employed to generate humanized anti-AQP4 antibodies of the invention by substituting at least a portion of a CDR from a non-human anti-AQP4 antibody for the corresponding regions of a human antibody. In some embodiments, the humanized anti-AQP4 antibodies of the invention have all three CDRs in each immunoglobulin chain from the exemplified mouse anti-AQP4 antibody grafted into corresponding human framework regions. Various monoclonal antibodies or antigen-binding fragments with similar binding activities to that of the anti-AQP4 antibodies exemplified herein can be produced. General methods for preparation of monoclonal or polyclonal antibodies are well known in the art. See, e.g., Harlow & Lane, Using Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1998. The anti-AQP4 antibodies of the present disclosures can be generated by any technique for producing monoclonal antibody well known in the ar. One animal system for preparing hybridomas is the murine system. Hybridoma production in the mouse is a very well-established procedure. After immunization an animal with an appropriate antigen, B cells isolated from the animal are then fused to myeloma cells to generate antibody-producing hybridomas. Monoclonal mouse anti-AQP4 antibodies can be obtained by screening the hybridomas in an ELISA assay using an AQP polypeptide or fusion protein. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also well known in the art. Hybridomas secreting anti-AQP4 monoclonal antibodies, or recombinant monoclonal antibodies, can be prepared using methods known in the art. Once a monoclonal antibody specific for the AQP4 protein is identified (e.g., either a hybridoma-derived monoclonal antibody or a recombinant antibody from a combinatorial library), DNAs encoding the light and heavy chains of the monoclonal antibody are isolated by standard molecular biology techniques. For hybridoma derived antibodies, light and heavy chain cDNAs can be obtained, for example, by PCR amplification or cDNA library screening. For recombinant antibodies, such as from a phage display library, cDNA encoding the light and heavy chains can be recovered from the display package (e.g., phage) isolated during the library screening process and the nucleotide sequences of antibody light and heavy chain genes are determined. For example, many such sequences are disclosed in Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242. Once obtained, the antibody light and heavy chain sequences are cloned into a recombinant expression vector using standard methods. The invention provides substantially purified polynucleotides (DNA or RNA) which encode polypeptides comprising segments or domains of the anti-AQP4 antibody chains or antigen-binding molecules described above. Some of the polynucleotides of the invention comprise the nucleotide sequence encoding the heavy chain variable region of exemplified mouse anti-AQP4 antibody. They can alternatively or additionally comprise the nucleotide sequence encoding the light chain variable region of the described anti-AQP4 antibody. Some other polynucleotides of the invention comprise nucleotide sequences that are substantially identical (e.g., at least 80%, 95%, or 99%) to the nucleotide sequence encoding the heavy chain variable region or light chain variable region of an exemplified anti-AQP4 antibody. Also provided in the present disclosures are expression vectors and host cells for producing the anti-AQP4 antibodies described above. Various expression vectors can be employed to express the polynucleotides encoding the anti-AQP4 antibody chains or binding fragments. As used herein, an expression vector refers to any nucleic acid construct which contains the necessary elements for the transcription and translation of an inserted coding sequence, or in the case of an RNA viral vector, the necessary elements for replication and translation, when introduced into an appropriate host cell. Expression vectors can include plasmids, phagemids, viruses, and derivatives thereof. Expression vectors of the disclosure can include polynucleotides encoding the antibody or antigen binding porting thereof described herein. In some embodiments, the coding sequences for the antibody or antigen binding porting thereof is operably linked to an expression control sequence. A coding sequence and a gene expression control sequence are said to be operably linked when they are covalently linked in such a way as to place the expression or transcription and/or translation of the coding sequence under the influence or control of the gene expression control sequence. The anti-AQP4 antibodies described herein can be employed in treating or preventing neuromyelitis optica (NMO). Accordingly, the present disclosures provide a method for treating and/or preventing NMO, comprising administering an effective amount of an antibody of the invention to a subject. Some embodiments of the invention employ a pharmaceutical composition containing the above-described anti-AQP4 antibody for administration to a subject already affected by a disease or condition caused by or associated with AQP4 (e.g., NMO). The composition contains the antibody or antigen-binding molecules in an amount sufficient to cure, partially arrest, or detectably slow the progression of the condition, and its complications. In prophylactic applications, compositions containing the anti-AQP4 antibodies or antigen-binding molecules are administered to a subject not already suffering from NMO. Rather, they are directed to a subject who is at the risk of, or has a predisposition, to developing such a disorder. Such applications allow the subject to enhance the subject's resistance or to retard the progression of a disorder mediated by AQP4. The invention provides pharmaceutical compositions comprising the anti-AQP4 antibodies or antigen-binding molecules formulated together with a pharmaceutically acceptable carrier. The compositions can additionally contain other therapeutic agents that are suitable for treating or preventing a given disorder. Pharmaceutically carriers enhance or stabilize the composition, or to facilitate preparation of the composition. Pharmaceutically acceptable carriers include solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. A pharmaceutical composition of the present invention can be administered by a variety of methods known in the art. The route and/or mode of administration vary depending upon the desired results. It is preferred that administration be intravenous, intramuscular, intraperitoneal, or subcutaneous, or administered proximal to the site of the target. The pharmaceutically acceptable carrier should be suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion). Depending on the route of administration, the active compound, i.e., antibody, may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound. The following examples will help describe how the invention is practiced and will illustrate the characteristics of the claimed antibodies and methods. Examples Materials and Methods Animals Used in Study The wild type C57BL/6J (B6) mice in 6-8 weeks old wee used in animal study. The mice were purchased from National Laboratory Animal Center and hosted according to the regulations of Institutional Animal Care and Use Committee, IACUC. Cell Line Used in Cell Fusion NS-1 myeloma cells used in the cell fusion with splenic lymphocyte were cultured in RPMI-1640 medium with 10% FBS. After culturing for several generations, the stabilized cells were freezing stored and used in experiments. Synthesis of Peptide Epitope The peptide epitopes, mAQP4-Loop C (mouse AQP4137-155-KLH) and mAQP-Loop E (mouse AQP4211-230-KLH), were synthesized and used to as antigen to induce immune reaction. Peptide Epitope mAQP4-LoopCKLH-(SH)-TPPSVVGGLGVTTVHGNLTC(CKLH-(SH)-SEQ ID NO: 1)mAQP4-LoopEKLH-(SH)-CSMNPARSFGPAVIMGNWANH(EKLH-(SH)-SEQ ID NO: 2) Intrasplenic Immunization The mice were anesthetized with 30 mg/kg of zoletil 50 and 2% rompun by intraperitoneal injection. The abdominal cavity was open by surgical procedure. 20 μg of peptide epitope mAQP4-Loop C or mAQP4-Loop E was injected to spleen of the mouse. After injection, the abdominal cavity was sutured. Evaluation of Immune Response At the 4thday and the 7thday after immunization, blood of the mouse was taken by cardiac puncture. The resulting blood was centrifugated at 3,000 rpm for 10 minutes to collect serum. The serum was subjected to enzyme immunoassay (EIA). Before the EIA was conducted, 100 μL of the antigen (2.5 μg/mL), mAQP4-Loop C or mAQP4-Loop E in coating buffer, was added to each well of the 96-well plate. The solution was discarded and then the plate was washed with washing buffer for three times to remove the antigens unbound to the bottom of the wells. Then, blocking buffer was added to each well and reacted at the room temperature for one hour. After removing the blocking buffer, the plate was washed with washing buffer for three times. The blocking buffer as negative control, AQP4 antibody as positive control and serum with appropriate dilution were added to the wells, respectively, and placed at the room temperature for 2 hours for reaction. The wells were washed by washing buffer 4 times. Then, 50 μL of anti-IgG-HRP was added to each well and reacted for one hour. The wells were washed with washing buffer three times. 50 μL of TMB substrate solution was added each well and reacted 30 minutes under light shade. After color reaction, 50 μL of stop solution was added to each well to quench the reaction. Then, the absorbances were measured at 450 nm by ELISA reader. Isolation of Spleen Lymphocytes At the 4thday and the 7thday after immunization, the mice were sacrificed and the spleen was removed from the body. The spleen was mixed with 10 mL of T cell medium and grinded for mixing. The resulting solution was centrifugated at 1,300 rpm for 5 minutes. The supernatant was discarded and then 4 mL of T cell medium was added. After homogenization, 4 mL of cell suspension was slowly added to 3 mL of Ficol-Paque (GE Healthcare, Sweden) and then centrifugated Example 1 Induction of Immune Response Six to eight weeks old of wild type C57BL/6J (B6) female mice were used in intrasplenic injection for immunization. At 4thand 7thdays after immunization, blood was taken from heart of the mice and then centrifugated at 3,000 rpm for 10 minutes to collect serums. The serum samples were subjected to enzyme immunoassay (EIA). The serum samples were added to 96-well plate with or without antigen and then EIA was conducted for the serum samples and the absorbances were detected. The absorbance of the serum sample in the plate with the antigen divides by that without the antigen to obtain OD ratio. The OD ratio greater than 2 represents the presence of antibody in serum sample and the induction of immune response. As shown inFIG.1, the OD ratios determined at 4t h day after immunization for LoopC-4 and LoopE-4 are 2.892 and 2.692, respectively. The OD ratios determined at 7th day after immunization for LoopC-4 and LoopE-4 are 5.474 and 4.519, respectively. Example 2 B Cell Lymphocyte Cell Line Secreting Anti-Mouse AQP4 Antibody After the immunization with mAQP4-Loop C or mAQP4-Loop E for 4 days or 7 days, the mouse was sacrificed and the spleen was taken out from the mouse. Then, lymphocytes were isolated from the spleen. The resulting lymphocytes were fused with NS-1 myeloma cells and the medium containing 1X HAT was used to select cell hybridoma. The fused hybridoma was cultured in 1X HAT at 37° C., 5% CO2for 3 weeks. The supernatant was collected and subjected to enzyme immunoassay (EIA) to select B cell lymphocytes secreting anti-mouse AQP4 antibody. The results are shown inFIG.2(4-day immunization) andFIG.3(7-day immunization). Four B cell lymphocyte cell lines which secret antibody with highest concentration after immunization of 4 days and 7 days were selected. The anti-mouse AQP4 antibodies were obtained by using protein G spin column and indirect immunofluorescence assay (IFA) to purify the supernatants of cell culture medium. The purified antibodies reacted with HEK293 expressing eGFP-mAQP4. The purified antibody linked to the secondary antibody with fluorescent Cy3 to confirm the specificity and affinity of the antibody to the mouse AQP4. The results show that the anti-mouse AQP4 antibodies (A001 produced from the hybridoma cell line AQP001 and A002 produced from the hybridoma cell line AQP002) produced from the four B cell lymphocyte cell lines have specificity and affinity to the mouse AQP4 protein (seeFIG.4). Example 2 Competitive Test of Anti-AQP4 Antibody to NMO-IgG Comparative studies of binding affinity on human AQP4 were conducted in dead cells or living cells using anti-mouse AQP4 antibody and NMO-IgG. For study conducted in dead cells, eGFP-hAQP4-HEK293 cells were fixed using 4% paraformaldehyde and then 1% BSA of blocking buffer was added to the cells. After one hour, 1X PBS was added for washing for 3 times. After washing, A001 and A002 were added to the cells and reacted at 4° C. under dark for 24 hours and then the resulting cells were washed by PB ST for 4 times. Then, NMO-IgG was added to the resulting cells for reaction. After 2 hours, the cells were washed by PB ST for 4 times. Then, anti-human IgG-Cy3 (Jackson ImmunoResearch, 709-165-149) and anti-mouse IgG-DyLight633 (Invitrogen, 35512) were added to the cells, respectively. After reaction for 45 minutes, the resulting cells were washed by PB ST for 3 times and then Fluoromount-G (eBioscienc, USA) was added for mounting. The resulting cells were observed by co-focal microscope (LSM510, Zeiss, Göttingen, Germany). For study conducted in living cells, NMO-IgG was added to eGFP-hAQP4-HEK293 cells. After a reaction at 37° C. under 5% CO2for 2 hours, A001 and A002 were added to the resulting cells were reacted at 37° C. under 5% CO2for 24 hours and then washed by 1X PBS for 3 times. After the resulting cells were fixed with 4% paraformaldehyde, permeabilized buffer with 0.1% Triton-X was added to the cells and reacted at room temperature for 10 minutes. Then, blocking buffer with 1% BSA was added to the cells and reacted at room temperature under dark for 1 hour. The resulting cells were washed with 1×PBS for 3 times and then anti-human IgG-Cy3 (Jackson ImmunoResearch, 709-165-149) and nti-mouse IgG-DyLight633 (Invitrogen, 35512) were added to cells and reacted at room temperature under dark for 45 minutes. The resulting cells were washed by PB ST for 3 times. Then, Fluoromount-G (eBioscienc, USA) was added for mounting. The resulting cells were observed by co-focal microscope (LSM510, Zeiss, Gottingen, Germany). The results of the above-mentioned comparative studies were shown inFIGS.5to7. Example 3 CDC Assay of A002 Monoclonal Ab After HEK293 cells were transfected with hAQP4-plasmid DNA for 24 hours, the cells were incubated with NMO-IgG or A002, or with normal human serum (1:20) or anti-beta actin mouse mAb with 5% human complement for 90 min at 37° C. After washed by 1X PBS, cells were incubated with 10 nM Calcein-AM for 15 min at 37° C. Then cells were incubated with 10 μM Propidium Iodide for 15 min at 37° C. after washed. After washed, cells were incubated with 4% formaldehyde for 10 min at room temperature. The images were obtained by microscopy and the results were shown inFIG.8. Example 4 Antibody-Dependent Cell-Mediated Cytotoxicity Assay HEK293-hAQP4-GFP and LPS-stimulated RAW264.7 were co-cultured with CD4 antibody, commercial AQP4 antibody, A002 antibody (ten-fold serial dilution) or culture medium only at 37° C. and 5% CO 2 for 6 hours. Cells were stain with Fixable Far Red-labeled anti-amine, PE-labeled anti-mouse CD11 b then analyzed the % of amine in CD11b-/GFP+ cells. Cell death(%) increased in ADCC=(% cell death in presence of IgG-% cell death in absence of IgG)/(% Cell death in maximum lysis-% cell death in absence of IgG)×100. Antibody-dependent cell-mediated cytotoxicity Assay (ADCC) of A002 antibody by immunofluorescent stain. HEK293-hAQP4-GFP and LPS-stimulated RAW264.7 were co-cultured with CD4 antibody, commercial AQP4 antibody, A002 antibody (ten-fold serial dilution) or culture medium only at 37° C. and 5% CO2for 6 hours. Then cells were stain with Propidium Iodide (PI). Histograms show quantification of Propidium Iodide (PI) of cells co-cultured with CD4 antibody, commercial AQP4 antibody or A002 antibody (ten-fold serial dilution) (FIG.9). PI intensity was adjusted by Subtracting PI intensity of cells co-cultured with culture medium only. Example 5 Mouse Ig Isotyping Assay Mouse Ig Isotyping of A002 antibody. A002 Ab was diluted to 100 ng/μl as working concentration and analyzed by Mouse Ig Isotyping Instant ELISA Kit (Invitrogen). After reading absorbance of 450 nm by ELISA reader (BioTek, USA), A002 antibody was classified to mouse IgG subtype, IgG3 (FIG.10).
23,889
11858990
DETAILED DESCRIPTION OF THE INVENTION Introduction The antibodies of the disclosure are able to directly and specifically target KIR3DL2-expressing cells, notably CD4+, KIR3DL2+ T cells, without targeting other cells such as KIR3DL1+ cells (or KIR3DL2+ KIR3DL1+ cells, KIR3DS1+ cells; or KIR3DS1 KIR3DL2+ cells), and do not internalize into KIR3DL2+ cells. Also provided are antibodies that do or not inhibit binding of natural ligands of KIR3DL2 (or ligand-induced KIR3DL2 signaling). The disclosure provides a number of antibodies having such properties, and which compete with each other for binding to a region of KIR3DL2+ that includes domains 0 and 2 defined by amino acid residues 1-98 and residues 193-292, respectively, of the mature KIR3DL2 polypeptides of SEQ ID NO: 1. KIR3DL2 (CD158k) is a disulphide-linked homodimer of three-Ig domain molecules of about 140 kD, described in Pende et al. (1996) J. Exp. Med. 184: 505-518, the disclosure of which is incorporated herein by reference. KIR3DL1 (CD158e1) is a monomeric molecule of about 70 kD, described in Colonna and Samaridis (1995) Science 268 (5209), 405-408; the HLA binding pocket has been described in Vivian et al. (2011) Nature 479: 401-405. Natural ligands of KIR3DL2 include, inter alia, HLA-A and HLA-B polypeptides, notably HLA-A3 and HLA-A11 (see Hansasuta et al. (2004) Eur. J. Immunol. 34: 1673-1679 and HLA-B27. HLA-B27 (see, e.g., Weiss et al. (1985) Immunobiology 170(5):367-380 for organization, sequence and expression of the HLA-B27 gene, and for HLA-B27 multimers and HLA-B272 homodimers see Allen et al. (1999) J. Immunol. 162: 5045-5048 and Kollnberger et al (2007) Eur. J. Immunol. 37: 1313-1322. The disclosures of all of the above references are incorporated herein by reference. As used herein, “KIR3D” refers to any KIR3D receptor (e.g. KIR3DL1, KIR3DL2, KIR3DS1) individually or collectively, and the term “KIR3D” may be substituted by the term “KIR3DL1, KIR3DL2 and/or KIR3DS1”. Similarly, “KIR3DL” refers to any KIR3DL receptor (e.g. KIR3DL1, KIR3DL2) individually or collectively, and the term “KIR3DL” may be substituted by the term “KIR3DL1 and/or KIR3DL2”. The terms “KIR3D”, “KIR3DL”, “KIR3DL1”, “KIR3DL2”, “KIR3DS1” each furthermore include any variant, derivative, or isoform of the KIR3D gene or encoded protein(s) to which they refer. Several allelic variants have been reported for KIR3D polypeptides (e.g. KIR3DL2), each of these are encompassed by the respective terms. The amino acid sequence of the mature human KIR3DL2 (allele *002) is shown in SEQ ID NO: 1, corresponding to Genbank accession no. AAB52520 in which the 21 amino acid residue leader sequence has been omitted, and corresponding to IPD KIR database (published by the EMBL-EBI, European Bioinformatics Institute, United Kingdom) accession no. KIR00066. The cDNA of KIR3DL2 (allele *002) is shown in Genbank accession no. U30272. The precursor amino acid sequence (including leader sequence) of a human KIR3DL2 allele *002 is shown in SEQ ID NO: 159, corresponding to Genbank accession no. AAB52520. The amino acid sequence of a human KIR3DL2 allele *001 is shown in SEQ ID NO: 160, corresponding to IPD KIR database accession no. KIR00065. The amino acid sequence of a human KIR3DL2 allele *003 is shown in SEQ ID NO: 161, corresponding to Genbank accession no. AAB36593 and IPD KIR database accession no. KIR00067. The amino acid sequence of a human KIR3DL2 allele *004 is shown in SEQ ID NO: 162, corresponding to IPD KIR database accession no. KIR00068. The amino acid sequence of a human KIR3DL2 allele *005 is shown in SEQ ID NO: 163, corresponding to IPD KIR database accession no. KIR00069. The amino acid sequence of a human KIR3DL2 allele *006 (mature) is shown in SEQ ID NO: 164, corresponding to Genbank accession no. AAK30053 and IPD KIR database accession no. KIR00070. The amino acid sequence of a human KIR3DL2 allele *007 (mature) is shown in SEQ ID NO: 165, corresponding to Genbank accession no. AAK30052 and IPD KIR database accession no. KIR00071. The amino acid sequence of a human KIR3DL2 allele *008 is shown in SEQ ID NO: 166, corresponding to Genbank accession no. AAK30054 and IPD KIR database accession no. KIR00072. The amino acid sequence of a human KIR3DL2 allele *009 is shown in SEQ ID NO: 167, corresponding to IPD KIR database accession no. KIR00457. The amino acid sequence of a human KIR3DL2 allele *011 is shown in SEQ ID NO: 168, corresponding to IPD KIR database accession no. KIR00544. The cDNA encoding a KIR3DL1 (CD158e2) polypeptide (allele *00101) is shown in Genbank accession no. L41269; the encoded amino acid sequence is shown in SEQ ID NO: 169, corresponding to Genbank accession no. AAA69870. Where a leader sequence is present in a particular SEQ ID NO describing a KIR3DL2 polypeptide sequence (e.g. SEQ ID NOS: 1 and 159 to 168), any reference to amino acid residue positions herein will be to the mature KIR3DL polypeptide. Provided are methods of using the antigen-binding compounds; for example, a method for inhibiting cell proliferation or activity, for delivering a molecule into a cell (e.g. a toxic molecule, a detectable marker, etc.), for targeting, identifying or purifying a cell, for depleting, killing or eliminating a cell, for reducing cell proliferation, the method comprising exposing a cell, such as a T cell which expresses a KIR3DL polypeptide, to an antigen-binding compound of the disclosure that binds a KIR3DL2 polypeptide. It will be appreciated that for the purposes of the present disclosure, “cell proliferation” can refer to any aspect of the growth or proliferation of cells, e.g., cell growth, cell division, or any aspect of the cell cycle. The cell may be in cell culture (in vitro) or in a mammal (in vivo), e.g. a mammal suffering from a KIR3DL2-expressing pathology. Also provided is a method for inducing the death of a cell or inhibiting the proliferation or activity of a cell which expresses a KIR3DL2 polypeptide, comprising exposing the cell to an antigen-binding compound that binds a KIR3DL2 polypeptide linked to a toxic agent, in an amount effective to induce death and/or inhibit the proliferation of the cell. Thus, provided is a method for treating a mammal suffering from a proliferative disease, and any condition characterized by a pathogenic expansion or activation of cells expressing of a KIR3DL2 polypeptide, the method comprising administering a pharmaceutically effective amount of an antigen-binding compound disclosed herein to the mammal. Examples of such conditions include Sézary Syndrome, Mycosis Fungoides, CTCL, and autoimmune or inflammatory conditions, e.g. arthritis, cardiovascular disease. Preferably such pathogenically expanded cells express KIR3DL2 but do not prominently express KIR3DL1 (e.g. no more than 20%, 40%, 50% or 60% of pathogenic cells express KIR3DL1, these conditions benefitting particularly from selective antibodies. Several KIR3DL2-expressing disorders, particularly T and NK cell mediated disorders can be treated or diagnosed using the methods and compositions of the disclosure. The disorders may be for example CD4+ T cell malignancies such as CTCL, MF or SS, or autoimmune or inflammatory disorders where the elimination or inhibiting the activity and/or proliferation of T and/or NK cells would be useful. CD4+ T cells includes for example activated CD4+ T cells, Th17 T cells, CD4+ T cells expressing or not one or more other markers (e.g. CD2+, CD3+, CD5+, CD8−, CD28−, CD28−, CD45R0+ and TCRαβ+). CD4+CD28− T cells, for example, are known to be capable of expressing KIR3DL2 and are present in high frequencies of clonally expanded cells in some autoimmune and inflammatory disorders but are rare in healthy individuals. These T cells can be cytotoxic, secrete large amounts of IFN-gamma, and proliferate upon stimulation with autologous adherent mononuclear cells. The antibodies of the disclosure have the advantage of binding across different KIR3DL2 alleles permitting a broad use to treat, characterize and diagnose diseases. Cutaneous and circulating MF/SS cells have been reported to not express preferential alleles among nine KIR3DL2 alleles tested. Thirteen alleles have also been described to date. Whereas the p140-KIR3DL2 receptor is expressed on a minor subset of NK cells and on rare CD8+ T cells in healthy persons, it appears to be restricted to CTCL tumor CD4+ T cells in MF/SS patients. Other receptors that are usually observed at the surface of NK cells (such as p58.1, p58.2, p70KIRs, CD94/NKG2A) are not found at the surface of malignant CD4+ T cells (Bahler D. W. et al., (2008) Cytometry B Clin Cytom. 74(3):156-62). SS cells are also typically characterized, in addition to CD4+, by having a mature T lymphocyte phenotype, CD2+, CD3+, CD5+, CD8−, CD28+, CD45RO+ and TCRαβ+. The methods and compositions of the disclosure can be used in the treatment of autoimmune and inflammatory conditions characterized by KIR3DL2 expression, by eliminating KIR3DL2-expressing cells and/or by inhibiting the biological activity KIR3DL2-expressing cells (i.e. by blocking KIR3DL2 signaling induced by its natural ligands). Inhibiting the biological activity KIR3DL2-expressing cells can comprise for example decreasing the proliferation of KIR3DL2-expressing cells, decreasing the reactivity or cytotoxicity of KIR3DL2-expressing cells toward target cells, decreasing activation, activation markers (e.g. CD107 expression) and/or cytokine production (e.g., IFNγ production) by a KIR3DL2-expressing cell, and/or decreasing the frequency in vivo of such activated, reactive, cytotoxic and/or activated KIR3DL2-expressing cells. For example, it has been shown that several such disorders are mediated at least in part by CD4+ T cells, including particular CD4+CD28null T cells. Activation of CD4+ T cells is generally thought to be governed by interplay between stimulatory and inhibitory receptors, where a predominance of stimulatory signals favors autoimmune reactions. Chan et al. ((2005) Arthrit. Rheumatism 52(11): 3586-3595 report that increased number of peripheral blood and synovial fluid CD4+ T cells and NK cells express KIR3DL2 in spondyloarthritis. In patients with rheumatoid arthritis, expression of the critical costimulatory molecule, CD28, is frequently lost. Instead, a CD4+T cell population which lacks CD28 (CD4+CD28−T cells) express killer immunoglobulin-like receptors (KIRs). CD4+CD28nullT cells in particular have been reported to express KIR3D polypeptides. Compared with their CD28+counterparts, CD4+CD28− cells produce significantly higher levels of IFN-γ giving them the ability to function as proinflammatory cells. CD4−CD28nullT cell clones persist for years in circulation. These T cells are known to differ from CD28+T cells by being resistant to Fas-mediated apoptosis upon cross-linking of CD3. CD28nullT cells progress through the cell cycle, and cells at all stages of the cell cycle are resistant to apoptosis, unlike their CD28+counterparts. Dysregulation of apoptotic pathways in CD4+CD28nullT cells has been shown to favor their clonal outgrowth and maintenance in vivo. Namekawa et al. ((2000) J. Immunol. 165:1138-1145 report that KIR, including KIR3DL2, was present on CD4+CD28null T cells expanded in rheumatoid arthritis. Rheumatoid arthritis involves lymphocyte infiltrates, inflammatory mediators, and synovial hyperplasia resulting from aggressive proliferation of fibroblast-like synoviocytes and macrophages. Prognoses of joint erosions and disease severity correlate with high frequencies of clonally expanded CD4+CD28−T cells. Lamprecht et al. (2001) Thorax 56:751-757 report recruitment of CD4+CD28−T cells in Wegener's granulomatosis. Markovic-Plese et al. (2001) J Clin Invest. 108: 1185-1194 report the presence of CD4+CD28− costimulation-independent T cells in the CNS, and their associate with multiple sclerosis. The methods and compositions can therefore be used in the treatment or prevention of Wegener's granulomatosis, multiple sclerosis or other central nervous system inflammatory or autoimmune disorders, arthritis, or other rheumatic disorders characterized by inflammation. CD4+CD28−T cells have also been associated with cardiovascular disorders. Betjes et al. (2008) Kidney International 74, 760-767 report that the increased risk for atherosclerotic disease in patients with Cytomegalovirus (CMV) seropositivity is associated with age-dependent increase of CD4+CD28−T cells, which can comprise over half of the circulating CD4 T cells in individuals. Patients over 50 years of age were reported to have a 50-fold higher percentage of CD4+CD28−T cells compared to CMV seronegative patients and a 5-fold higher percentage when compared to seropositive healthy controls. Nakajima et al. ((2003) Circ. Res. 93:106-113) report de novo expression of KIR in acute coronary syndrome, where CD4+ T cells from patients with acute coronary syndrome (ACS) express multiple KIR whereas normal CD4+CD28null T cells from healthy donors do not express KIR. Yen et al. Journal of Experimental Medicine, Volume 193, Number 10, May 21, 2001 1159-1168 studied CD4+CD28nullT cell clones established from patients with rheumatoid vasculitis for the expression of inhibitory and stimulatory KIR by RT-PCR. In patients with rheumatoid arthritis and a patient with ACS, the expression patterns favored the inhibitory KIR, including KIR3DL2, whereas expression of stimulatory receptors was highly restricted to KIR2DS2. The methods and compositions can therefore be used in the treatment or prevention of cardiovascular disorders, e.g. ACS, atherosclerotic disease, rheumatoid vasculitis, characterized by inflammation. Bowness et al (2011) J. Immunol. 186: 2672-2680 report that KIR3DL2+ CD4 T cells account for the majority of IL-23R expression by peripheral blood CD4 T cells, and that such KIR3DL2+ cells of the Th17 type produce more IL-17 in the presence of IL-23. Despite KIR3DL2+ cells comprising a mean of just 15% of CD4 T in the peripheral blood of SpA patients, this subset accounted for 70% of the observed increase in Th17 numbers in SpA patients compared with control subjects. TCR-stimulated peripheral blood KIR3DL2+ CD4 T cell lines from SpA patients secreted 4-fold more IL-17 than KIR3DL2+ lines from controls or KIR3DL2−CD4 T cells. Provided are methods for producing and using antibodies and other compounds suitable for the treatment of disorders (e.g. cancers, inflammatory and autoimmune disorders) where eliminating KIR3DL2-expressing cells would be useful. Antibodies, antibody derivatives, antibody fragments, and cell producing them are encompassed, as are methods of producing the same and methods of treating patients using the antibodies and compounds. Since the present antibodies are specific for KIR3DL2, they can be used for a range of purposes, including purifying KIR3DL2 or KIR3DL2-expressing cells, modulating (e.g. activating or inhibiting) KIR3DL2 receptors in vitro, ex vivo, or in vivo, targeting KIR3DL2-expressing cells for destruction in vivo, or specifically labeling/binding KIR3DL2 in vivo, ex vivo, or in vitro, including for methods such as immunoblotting, IHC analysis, i.e. on frozen biopsies, FACS analysis, and immunoprecipitation. Definitions As used in the specification, “a” or “an” may mean one or more. As used in the claim(s), when used in conjunction with the word “comprising”, the words “a” or “an” may mean one or more than one. As used herein “another” may mean at least a second or more. Where “comprising” is used, this can preferably be replaced by “consisting essentially of”, more preferably by “consisting of”. “Treatment of a proliferative disease” or “treatment of a tumor”, or “treatment of cancer” or the like, with reference to anti-KIR3DL2 binding agent (e.g. antibody), includes, but is not limited to: (a) method of treatment of a proliferative disease, said method comprising the step of administering (for at least one treatment) an anti-KIR3DL2 binding agent, (e.g., in a pharmaceutically acceptable carrier material) to a warm-blooded animal, especially a human, in need of such treatment, in a dose that allows for the treatment of said disease (a therapeutically effective amount), e.g., in a dose (amount) as specified hereinabove and herein below; (b) the use of an anti-KIR3DL2 binding agent for the treatment of a proliferative disease, or an anti-KIR3DL2 binding agent, for use in said treatment (especially in a human); (c) the use of an anti-KIR3DL2 binding agent, for the manufacture of a pharmaceutical preparation for the treatment of a proliferative disease, a method of using an anti-KIR3DL2 binding agent for the manufacture of a pharmaceutical preparation for the treatment of a proliferative disease, comprising admixing an anti-KIR3DL2 binding agent with a pharmaceutically acceptable carrier, or a pharmaceutical preparation comprising an effective dose of an anti-KIR3DL2 binding agent that is appropriate for the treatment of a proliferative disease; or (d) any combination of a), b), and c), in accordance with the subject matter allowable for patenting in a country where this application is filed. In cases where a particular disease (e.g., inflammatory or autoimmune disease) or a specific tumor (e.g. CTCL) are mentioned instead of “proliferative disease”, categories a) to e) are also encompassed, meaning that the respective disease can be filled in under a) to e) above instead of “proliferative disease”, in accordance with the patentable subject matter. The terms “cancer” and “tumor” as used herein are defined as a new growth of cells or tissue comprising uncontrolled and progressive multiplication. In a specific embodiment, upon a natural course the cancer is fatal. In specific embodiments, a cancer is invasive, metastatic, and/or anaplastic (loss of differentiation and of orientation to one another and to their axial framework). “Autoimmune” disorders include any disorder, condition, or disease in which the immune system mounts a reaction against self cells or tissues, due to a breakdown in the ability to distinguish self from non-self or otherwise. Examples of autoimmune disorders include rheumatoid arthritis, rheumatoid vasculitis, systemic lupus erythematosus, multiple sclerosis, Wegener's granulomatosis, spondyloarthritis, and others. An “inflammatory disorder” includes any disorder characterized by an unwanted immune response. Autoimmune and inflammatory disorders can involve any component of the immune system, and can target any cell or tissue type in the body. The term “biopsy” as used herein is defined as removal of a tissue from an organ (e.g., a joint) for the purpose of examination, such as to establish diagnosis. Examples of types of biopsies include by application of suction, such as through a needle attached to a syringe; by instrumental removal of a fragment of tissue; by removal with appropriate instruments through an endoscope; by surgical excision, such as of the whole lesion; and the like. The term “antibody,” as used herein, refers to polyclonal and monoclonal antibodies. Depending on the type of constant domain in the heavy chains, antibodies are assigned to one of five major classes: IgA, IgD, IgE, IgG, and IgM. Several of these are further divided into subclasses or isotypes, such as IgG1, IgG2, IgG3, IgG4, and the like. An exemplary immunoglobulin (antibody) structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids that is primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains, respectively. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are termed “alpha,” “delta,” “epsilon,” “gamma” and “mu,” respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. IgG and/or IgM are the preferred classes of antibodies employed herein, with IgG being particularly preferred, because they are the most common antibodies in the physiological situation and because they are most easily made in a laboratory setting. Preferably the antibody is a monoclonal antibody. Particularly preferred are humanized, chimeric, human, or otherwise-human-suitable antibodies. “Antibodies” also includes any fragment or derivative of any of the herein described antibodies. The term “specifically binds to” means that an antibody can bind preferably in a competitive binding assay to the binding partner, e.g. KIR3DL2, as assessed using either recombinant forms of the proteins, epitopes therein, or native proteins present on the surface of isolated target cells. Competitive binding assays and other methods for determining specific binding are further described below and are well known in the art. When an antibody is said to “compete with” a particular monoclonal antibody (e.g. 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9), it means that the antibody competes with the monoclonal antibody in a binding assay using either recombinant KIR3DL2 molecules or surface expressed KIR3DL2 molecules. For example, if a test antibody reduces the binding of 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9 to a KIR3DL2 polypeptide or KIR3DL2-expressing cell in a binding assay, the antibody is said to “compete” respectively with 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9. The term “affinity”, as used herein, means the strength of the binding of an antibody to an epitope. The affinity of an antibody is given by the dissociation constant Kd, defined as [Ab]×[Ag]/[Ab-Ag], where [Ab-Ag] is the molar concentration of the antibody-antigen complex, [Ab] is the molar concentration of the unbound antibody and [Ag] is the molar concentration of the unbound antigen. The affinity constant Kais defined by 1/Kd. Examples of methods for determining the affinity of mAbs can be found in Harlow, et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988), Coligan et al., eds., Current Protocols in Immunology, Greene Publishing Assoc. and Wiley Interscience, N.Y., (1992, 1993), and Muller, Meth. Enzymol. 92:589-601 (1983), which references are entirely incorporated herein by reference. One standard method well known in the art for determining the affinity of mAbs is the use of surface plasmon resonance (SPR) screening (such as by analysis with a BIAcore™ SPR analytical device). As used herein, a “determinant” designates a site of interaction or binding on a polypeptide. The term “epitope” refers to an antigenic determinant, and is the area or region on an antigen to which an antibody binds. A protein epitope may comprise amino acid residues directly involved in the binding as well as amino acid residues which are effectively blocked by the specific antigen binding antibody or peptide, i.e., amino acid residues within the “footprint” of the antibody. It is the simplest form or smallest structural area on a complex antigen molecule that can combine with e.g., an antibody or a receptor. Epitopes can be linear or conformational/structural. The term “linear epitope” is defined as an epitope composed of amino acid residues that are contiguous on the linear sequence of amino acids (primary structure). The term “conformational or structural epitope” is defined as an epitope composed of amino acid residues that are not all contiguous and thus represent separated parts of the linear sequence of amino acids that are brought into proximity to one another by folding of the molecule (secondary, tertiary and/or quaternary structures). A conformational epitope is dependent on the 3-dimensional structure. The term ‘conformational’ is therefore often used interchangeably with ‘structural’. The term “intracellular internalization”, or “internalization” when referring to a KIR3DL2 polypeptide and/or antibody that binds such, refers to the molecular, biochemical and cellular events associated with the process of translocating a molecule from the extracellular surface of a cell to the intracellular surface of a cell. The processes responsible for intracellular internalization of molecules are well-known and can involve, inter alfa, the internalization of extracellular molecules (such as hormones, antibodies, and small organic molecules); membrane-associated molecules (such as cell-surface receptors); and complexes of membrane-associated molecules bound to extracellular molecules (for example, a ligand bound to a transmembrane receptor or an antibody bound to a membrane-associated molecule). Thus, “inducing and/or increasing intracellular internalization” comprises events wherein intracellular internalization is initiated and/or the rate and/or extent of intracellular internalization is increased. The term “depleting”, with respect to KIR3DL2-expressing cells means a process, method, or compound that can kill, eliminate, lyse or induce such killing, elimination or lysis, so as to negatively affect the number of KIR3DL2-expressing cells present in a sample or in a subject. The term “agent” is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials. The term “therapeutic agent” refers to an agent that has biological activity. The terms “toxic agent” and “cytotoxic agent” encompass any compound that can slow down, halt, or reverse the proliferation of cells, decrease their activity in any detectable way, or directly or indirectly kill them. Preferably, cytotoxic agents cause cell death primarily by interfering directly with the cell's functioning, and include, but are not limited to, alkylating agents, tumor necrosis factor inhibitors, intercalators, microtubule inhibitors, kinase inhibitors, proteasome inhibitors and topoisomerase inhibitors. A “toxic payload” as used herein refers to a sufficient amount of cytotoxic agent which, when delivered to a cell results in cell death. Delivery of a toxic payload may be accomplished by administration of a sufficient amount of immunoconjugate comprising an antibody or antigen binding fragment and a cytotoxic agent. Delivery of a toxic payload may also be accomplished by administration of a sufficient amount of an immunoconjugate comprising a cytotoxic agent, wherein the immunoconjugate comprises a secondary antibody or antigen binding fragment thereof which recognizes and binds an antibody or antigen binding fragment. For the purposes herein, a “humanized” or “human” antibody refers to an antibody in which the constant and variable framework region of one or more human immunoglobulins is fused with the binding region, e.g. the CDR, of an animal immunoglobulin. Such antibodies are designed to maintain the binding specificity of the non-human antibody from which the binding regions are derived, but to avoid an immune reaction against the non-human antibody. Such antibodies can be obtained from transgenic mice or other animals that have been “engineered” to produce specific human antibodies in response to antigenic challenge (see, e.g., Green et al. (1994) Nature Genet 7:13; Lonberg et al. (1994) Nature 368:856; Taylor et al. (1994) Int Immun 6:579, the entire teachings of which are herein incorporated by reference). A fully human antibody also can be constructed by genetic or chromosomal transfection methods, as well as phage display technology, all of which are known in the art (see, e.g., McCafferty et al. (1990) Nature 348:552-553). Human antibodies may also be generated by in vitro activated B cells (see, e.g., U.S. Pat. Nos. 5,567,610 and 5,229,275, which are incorporated in their entirety by reference). A “chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity. The terms “Fc domain,” “Fc portion,” and “Fc region” refer to a C-terminal fragment of an antibody heavy chain, e.g., from about amino acid (aa) 230 to about aa 450 of human γ (gamma) heavy chain or its counterpart sequence in other types of antibody heavy chains (e.g., α, δ, ε and μ for human antibodies), or a naturally occurring allotype thereof. Unless otherwise specified, the commonly accepted Kabat amino acid numbering for immunoglobulins is used throughout this disclosure (see Kabat et al. (1991) Sequences of Protein of Immunological Interest, 5th ed., United States Public Health Service, National Institute of Health, Bethesda, MD). The term “antibody-dependent cell-mediated cytotoxicity” or “ADCC” is a term well understood in the art, and refers to a cell-mediated reaction in which non-specific cytotoxic cells that express Fc receptors (FcRs) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. Non-specific cytotoxic cells that mediate ADCC include natural killer (NK) cells, macrophages, monocytes, neutrophils, and eosinophils. The terms “isolated”, “purified” or “biologically pure” refer to material that is substantially or essentially free from components which normally accompany it as found in its native state. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified. The terms “polypeptide,” “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer. The term “recombinant” when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (nonrecombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all. The term “modification” when referring to a sequence of amino acids (e.g., “amino acid modification”), is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence. By “modification” or “amino acid modification” is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence. By “amino acid substitution” or “substitution” herein is meant the replacement of an amino acid at a given position in a protein sequence with another amino acid. For example, the substitution P14S refers to a variant of a parent polypeptide, in which the proline at position 14 is replaced with serine. A “variant” of a polypeptide refers to a polypeptide having an amino acid sequence that is substantially identical to a reference polypeptide, typically a native or “parent” polypeptide. The polypeptide variant may possess one or more amino acid substitutions, deletions, and/or insertions at certain positions within the native amino acid sequence. As used herein, “T cells” refers to a sub-population of lymphocytes that mature in the thymus, and which display, among other molecules T cell receptors on their surface. T cells can be identified by virtue of certain characteristics and biological properties, such as the expression of specific surface antigens including the TCR, CD4 or CD8, optionally CD4 and IL-23R, the ability of certain T cells to kill tumor or infected cells, the ability of certain T cells to activate other cells of the immune system, and the ability to release protein molecules called cytokines that stimulate or inhibit the immune response. Any of these characteristics and activities can be used to identify T cells, using methods well known in the art. As used herein, “active” or “activated” T cells designate biologically active T cells, more particularly T cells having the capacity of cytolysis or of stimulating an immune response by, e.g., secreting cytokines. Active cells can be detected in any of a number of well-known methods, including functional assays and expression-based assays such as the expression of cytokines such as TNF-alpha or IL-17A. As used herein, the term antibody that “binds” a polypeptide or epitope designates an antibody that binds said determinant with specificity and/or affinity. Antibodies and Epitopes The antibodies disclosed are antibodies that bind human KIR3DL2. In an embodiment, the antibodies selectively bind KIR3DL2 (e.g. the 1, 2, 3, 4 or more most predominant KIR3DL2 alleles) and do not bind KIR3DL1 (e.g. the 1, 2, 3, 4 or more most predominant KIR3DL1 alleles). In one embodiment, the antibodies bind the D0 domain of KIR3DL2 corresponding to amino acid residues 1-98 of the KIR3DL2 polypeptide of SEQ ID NO: 1. In one embodiment, the antibodies bind the D2 domain of KIR3DL2, or to a region spanning both the D1 and D2 domains (at the border of the D1 and D2 domains), of the KIR3DL2 polypeptide of SEQ ID NO: 1. In one embodiment, the antibodies have an affinity for human KIR3DL2 characterized by a KDof less than 10−9M, preferably less than 10−10M. In another embodiment, the antibodies bind substantially the same epitope as antibody 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9. In another embodiment, the antibodies at least partially overlaps, or includes at least one residue in the segment corresponding to residues 1-98 or residues 193-292 of the KIR3DL2 polypeptide of SEQ ID NO: 1 (or a subsequence thereof. In one embodiment, all key residues of the epitope are in a segment corresponding to residues 1-98. In one embodiment, the antibody binds a residue present in the D1 domain as well as a residue present in in the D2 domain; optionally one or more key residues is at the border of the D1 (residues 99-192) and D2 domains (residues 193-292). In one embodiment, the antibodies bind an epitope comprising 1, 2, 3, 4, 5, 6, 7 or more residues in the segment corresponding to residues 1-98, 99-292, 99-192, or 193-292 of the KIR3DL2 polypeptide of SEQ ID NO: 1. Preferably the residues bound by the antibody are present on the surface of the of the KIR3DL2 polypeptide. In one embodiment, the antibodies bind an epitope comprising residues R13, A25, and/or Q27. Optionally, the antibodies bind an epitope comprising residues R13, A25, and/or Q27, as well residues I60 and/or G62. Optionally, the antibodies do not bind residues H32 and/or H33. Optionally, the antibodies further bind residues Q56 and/or E57. In one embodiment, the antibodies bind an epitope comprising residues I60 and/or G62. Optionally, the antibodies bind an epitope comprising one or more of residues I60 and/or G62, but not residues R13, A25, and/or Q27. In one embodiment, the antibodies bind an epitope comprising one or more of residues I60 and/or G62 as well as one or more of residues P14, S15 and/or H23. Optionally, the antibodies bind an epitope comprising 1, 2, 3, 4, 5, 6 or 7 of residues G21, G22, H23, E57, S58, F59, P63 and/or H68. Optionally, the antibodies bind an epitope comprising one or more of residues R78 and/or L82. Optionally, the antibodies bind an epitope comprising 1, 2, 3, 4, 5, 6 or 7 of residues K7, Y30, R31, P79, H80, S81, T83, G84, W85, S86 and/or A87. Optionally, the antibodies bind an epitope comprising residue W226. Optionally, the antibodies bind an epitope comprising 1, 2, 3, 4, 5, 6 or 7 of residues Q201, K202, P203, S204, S224, S225, S227, S228, N252, R253 and/or T254. Optionally, the antibodies bind an epitope comprising one or more of residues I231 and/or R246. Optionally, the antibodies bind an epitope comprising residues I231 and/or R246 as well as to an epitope comprising residue W226. Optionally, the antibodies bind an epitope comprising 1, 2, 3, 4, 5, 6 or 7 of residues D230, I231, R244, L245, R246, A247, V248, S275, R277 and/or P280. Optionally, the antibodies bind an epitope comprising residue E239. Optionally, the antibodies further bind one or more of residues I231 and/or R246. Optionally, the antibodies further bind residue W226. The Examples section herein describes the construction of a series of mutant human KIR3DL2 polypeptides. Binding of anti-KIR3DL2 antibody to cells transfected with the KIR3DL2 mutants was measured and compared to the ability of anti-KIR3DL2 antibody to bind wild-type KIR3DL2 polypeptide (SEQ ID NO:1). A reduction in binding between an anti-KIR3DL2 antibody and a mutant KIR3DL2 polypeptide as used herein means that there is a reduction in binding affinity (e.g., as measured by known methods such FACS testing of cells expressing a particular mutant, or by Biacore testing of binding to mutant polypeptides) and/or a reduction in the total binding capacity of the anti-KIR3DL2 antibody (e.g., as evidenced by a decrease in Bmax in a plot of anti-KIR3DL2 antibody concentration versus polypeptide concentration). A significant reduction in binding indicates that the mutated residue is directly involved in binding to the anti-KIR3DL2 antibody or is in close proximity to the binding protein when the anti-KIR3DL2 antibody is bound to KIR3DL2. An antibody epitope will thus preferably include such residue and may include additional residues adjacent to such residue. In some embodiments, a significant reduction in binding means that the binding affinity and/or capacity between an anti-KIR3DL2 antibody and a mutant KIR3DL2 polypeptide is reduced by greater than 40%, greater than 50%, greater than 55%, greater than 60%, greater than 65%, greater than 70%, greater than 75%, greater than 80%, greater than 85%, greater than 90% or greater than 95% relative to binding between the antibody and a wild type KIR3DL2 polypeptide (e.g., the polypeptide shown in SEQ ID NO:1). In certain embodiments, binding is reduced below detectable limits. In some embodiments, a significant reduction in binding is evidenced when binding of an anti-KIR3DL2 antibody to a mutant KIR3DL2 polypeptide is less than 50% (e.g., less than 45%, 40%, 35%, 30%, 25%, 20%, 15% or 10%) of the binding observed between the anti-KIR3DL2 antibody and a wild-type KIR3DL2 polypeptide (e.g., the extracellular domain shown in SEQ ID NO:1). Such binding measurements can be made using a variety of binding assays known in the art. A specific example of one such assay is described in the Example section. In some embodiments, anti-KIR3DL2 antibodies are provided that exhibit significantly lower binding for a mutant KIR3DL2 polypeptide in which a residue in a wild-type KIR3DL2 polypeptide (e.g., SEQ ID NO:1) is substituted. In the shorthand notation used here, the format is: Wild type residue: Position in polypeptide: Mutant residue, with the numbering of the residues as indicated in SEQ ID NO: 1. In some embodiments, an anti-KIR3DL2 antibody binds a wild-type KIR3DL2 polypeptide but has decreased binding to a mutant KIR3DL2 polypeptide having any one or more of the following mutations (with reference to SEQ ID NO:1):R13W, A25T and/or G25R;I60N and/or G62S;P14S, S 15A and/or H23S;one or more of R13W, A25T and/or G25R, and one or more of I60N and/or G62S;one or more of P14S, S15A and/or H23S, and one or more of I60N and/or G62S;one or more of R13W, A25T and/or G25R, one or more of I60N and/or G62S; and one or more of P14S, S15A and/or H23S;one or more of P14S, S15A and/or H23S, and one or more of I60N and/or G62S;R78H and/or L82P;W226A;I231M and/or R246P;one or more of I231M and/or R246P, and additionally W226A; orone or more of I231M and/or R246P, but wherein the antibody does not have decreased binding to a mutant have a mutation in W226A. Preferably binding to the particular mutant(s) of KIR3DL2 is significantly reduced compared to binding to the wild-type KIR3DL2. Producing Anti-KIR3DL2 Antibodies The antibodies may be produced by a variety of techniques known in the art. Typically, they are produced by immunization of a non-human animal, preferably a mouse, with an immunogen comprising a KIR3DL2 polypeptide, preferably a human KIR3DL2 polypeptide. The KIR3DL2 polypeptide may comprise the full length sequence of a human KIR3DL2 polypeptide, or a fragment or derivative thereof, typically an immunogenic fragment, i.e., a portion of the polypeptide comprising an epitope exposed on the surface of cells expressing a KIR3DL2 polypeptide, preferably the epitope recognized by the 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9 antibody. Such fragments typically contain at least about 7 consecutive amino acids of the mature polypeptide sequence, even more preferably at least about 10 consecutive amino acids thereof. Fragments typically are essentially derived from the extra-cellular domain of the receptor. In one embodiment, the immunogen comprises a wild-type human KIR3DL2 polypeptide in a lipid membrane, typically at the surface of a cell. In a specific embodiment, the immunogen comprises intact cells, particularly intact human cells, optionally treated or lysed. In another embodiment, the polypeptide is a recombinant KIR3DL2 polypeptide. In a specific embodiment, the immunogen comprises intact SS or MF cells, particularly intact human malignant CD4+ T cells, or CD4+CD28− T cells, optionally treated or lysed. In another embodiment, the polypeptide is a recombinant dimeric KIR3DL2 polypeptide. The step of immunizing a non-human mammal with an antigen may be carried out in any manner well known in the art for stimulating the production of antibodies in a mouse (see, for example, E. Harlow and D. Lane, Antibodies: A Laboratory Manual., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (1988), the entire disclosure of which is herein incorporated by reference). The immunogen is suspended or dissolved in a buffer, optionally with an adjuvant, such as complete or incomplete Freund's adjuvant. Methods for determining the amount of immunogen, types of buffers and amounts of adjuvant are well known to those of skill in the art. These parameters may be different for different immunogens, but are easily elucidated. Similarly, the location and frequency of immunization sufficient to stimulate the production of antibodies is also well known in the art. In a typical immunization protocol, the non-human animals are injected intraperitoneally with antigen on day 1 and again about a week later. This is followed by recall injections of the antigen around day 20, optionally with an adjuvant such as incomplete Freund's adjuvant. The recall injections are performed intravenously and may be repeated for several consecutive days. This is followed by a booster injection at day 40, either intravenously or intraperitoneally, typically without adjuvant. This protocol results in the production of antigen-specific antibody-producing B cells after about 40 days. Other protocols may also be used as long as they result in the production of B cells expressing an antibody directed to the antigen used in immunization. For polyclonal antibody preparation, serum is obtained from an immunized non-human animal and the antibodies present therein isolated by well-known techniques. The serum may be affinity purified using any of the immunogens set forth above linked to a solid support so as to obtain antibodies that react with KIR3DL2 polypeptides. In an alternate embodiment, lymphocytes from a non-immunized non-human mammal are isolated, grown in vitro, and then exposed to the immunogen in cell culture. The lymphocytes are then harvested and the fusion step described below is carried out. For monoclonal antibodies, the next step is the isolation of splenocytes from the immunized non-human mammal and the subsequent fusion of those splenocytes with an immortalized cell in order to form an antibody-producing hybridoma. The isolation of splenocytes from a non-human mammal is well-known in the art and typically involves removing the spleen from an anesthetized non-human mammal, cutting it into small pieces and squeezing the splenocytes from the splenic capsule through a nylon mesh of a cell strainer into an appropriate buffer so as to produce a single cell suspension. The cells are washed, centrifuged and resuspended in a buffer that lyses any red blood cells. The solution is again centrifuged and remaining lymphocytes in the pellet are finally resuspended in fresh buffer. Once isolated and present in single cell suspension, the lymphocytes can be fused to an immortal cell line. This is typically a mouse myeloma cell line, although many other immortal cell lines useful for creating hybridomas are known in the art. Murine myeloma lines include, but are not limited to, those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, U.S.A., X63 Ag8653 and SP-2 cells available from the American Type Culture Collection, Rockville, Maryland U.S.A. The fusion is effected using polyethylene glycol or the like. The resulting hybridomas are then grown in selective media that contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells. Hybridomas are typically grown on a feeder layer of macrophages. The macrophages are preferably from littermates of the non-human mammal used to isolate splenocytes and are typically primed with incomplete Freund's adjuvant or the like several days before plating the hybridomas. Fusion methods are described in Goding, “Monoclonal Antibodies: Principles and Practice,” pp. 59-103 (Academic Press, 1986), the disclosure of which is herein incorporated by reference. The cells are allowed to grow in the selection media for sufficient time for colony formation and antibody production. This is usually between about 7 and about 14 days. The hybridoma colonies are then assayed for the production of antibodies that specifically bind to KIR3DL2 polypeptide gene products, optionally the epitope specifically recognized by antibody 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9. The assay is typically a colorimetric ELISA-type assay, although any assay may be employed that can be adapted to the wells that the hybridomas are grown in. Other assays include radioimmunoassays or fluorescence activated cell sorting. The wells positive for the desired antibody production are examined to determine if one or more distinct colonies are present. If more than one colony is present, the cells may be re-cloned and grown to ensure that only a single cell has given rise to the colony producing the desired antibody. Hybridomas that are confirmed to produce a suitable monoclonal antibody can be grown up in larger amounts in an appropriate medium, such as DMEM or RPMI-1640. Alternatively, the hybridoma cells can be grown in vivo as ascites tumors in an animal. After sufficient growth to produce the desired monoclonal antibody, the growth media containing monoclonal antibody (or the ascites fluid) is separated away from the cells and the monoclonal antibody present therein is purified. Purification is typically achieved by gel electrophoresis, dialysis, chromatography using protein A or protein G-Sepharose, or an anti-mouse Ig linked to a solid support such as agarose or Sepharose beads (all described, for example, in the Antibody Purification Handbook, Biosciences, publication No. 18-1037-46, Edition AC, the disclosure of which is hereby incorporated by reference). The bound antibody is typically eluted from protein A/protein G columns by using low pH buffers (glycine or acetate buffers of pH 3.0 or less) with immediate neutralization of antibody-containing fractions. These fractions are pooled, dialyzed, and concentrated as needed. Positive wells with a single apparent colony are typically re-cloned and re-assayed to insure only one monoclonal antibody is being detected and produced. Antibodies may also be produced by selection of combinatorial libraries of immunoglobulins, as disclosed for instance in (Ward et al. Nature, 341 (1989) p. 544, the entire disclosure of which is herein incorporated by reference). The identification of one or more antibodies that bind(s) to KIR3DL2, particularly substantially or essentially the same epitope as monoclonal antibody 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9, can be readily determined using any one of a variety of immunological screening assays in which antibody competition can be assessed. Many such assays are routinely practiced and are well known in the art (see, e. g., U.S. Pat. No. 5,660,827, which is specifically incorporated herein by reference). It will be understood that actually determining the epitope to which an antibody described herein binds is not in any way required to identify an antibody that binds to the same or substantially the same epitope as the monoclonal antibody described herein. For example, where the test antibodies to be examined are obtained from different source animals, or are even of a different Ig isotype, a simple competition assay may be employed in which the control (10F6, for example for purposes of illustration, or any other antibody such as 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9) and test antibodies are admixed (or pre-adsorbed) and applied to a sample containing KIR3DL2 polypeptides. Protocols based upon western blotting and the use of BIACORE analysis are suitable for use in such competition studies. In certain embodiments, one pre-mixes the control antibodies (10F6, for example, although any other of antibodies) with varying amounts of the test antibodies (e.g., about 1:10 or about 1:100) for a period of time prior to applying to the KIR3DL2 antigen sample. In other embodiments, the control and varying amounts of test antibodies can simply be admixed during exposure to the KIR3DL2 antigen sample. As long as one can distinguish bound from free antibodies (e. g., by using separation or washing techniques to eliminate unbound antibodies) and (10F6 from the test antibodies (e. g., by using species-specific or isotype-specific secondary antibodies or by specifically labeling 10F6 with a detectable label) one can determine if the test antibodies reduce the binding of 10F6 to the antigens, indicating that the test antibody recognizes substantially the same epitope as 10F6. The binding of the (labeled) control antibodies in the absence of a completely irrelevant antibody can serve as the control high value. The control low value can be obtained by incubating the labeled (10F6) antibodies with unlabelled antibodies of exactly the same type (10F6), where competition would occur and reduce binding of the labeled antibodies. In a test assay, a significant reduction in labeled antibody reactivity in the presence of a test antibody is indicative of a test antibody that recognizes substantially the same epitope, i.e., one that “cross-reacts” or competes with the labeled (10F6) antibody. Any test antibody that reduces the binding of 10F6 to KIR3DL2 antigens by at least about 50%, such as at least about 60%, or more preferably at least about 80% or 90% (e. g., about 65-100%), at any ratio of 10F6:test antibody between about 1:10 and about 1:100 is considered to be an antibody that binds to substantially the same epitope or determinant as 10F6. Preferably, such test antibody will reduce the binding of 10F6 to the KIR3DL2 antigen by at least about 90% (e.g., about 95%). Competition can also be assessed by, for example, a flow cytometry test. In such a test, cells bearing a given KIR3DL2 polypeptide can be incubated first with 10F6, for example, and then with the test antibody labeled with a fluorochrome or biotin. The antibody is said to compete with 10F6 if the binding obtained upon preincubation with a saturating amount of 10F6 is about 80%, preferably about 50%, about 40% or less (e.g., about 30%, 20% or 10%) of the binding (as measured by mean of fluorescence) obtained by the antibody without preincubation with 10F6. Alternatively, an antibody is said to compete with 10F6 if the binding obtained with a labeled 10F6 antibody (by a fluorochrome or biotin) on cells preincubated with a saturating amount of test antibody is about 80%, preferably about 50%, about 40%, or less (e. g., about 30%, 20% or 10%) of the binding obtained without preincubation with the test antibody. A simple competition assay in which a test antibody is pre-adsorbed and applied at saturating concentration to a surface onto which a KIR3DL2 antigen is immobilized may also be employed. The surface in the simple competition assay is preferably a BIACORE chip (or other media suitable for surface plasmon resonance analysis). The control antibody (e.g., 10F6) is then brought into contact with the surface at a KIR3DL2-saturating concentration and the KIR3DL2 and surface binding of the control antibody is measured. This binding of the control antibody is compared with the binding of the control antibody to the KIR3DL2-containing surface in the absence of test antibody. In a test assay, a significant reduction in binding of the KIR3DL2-containing surface by the control antibody in the presence of a test antibody indicates that the test antibody recognizes substantially the same epitope as the control antibody such that the test antibody “cross-reacts” with the control antibody. Any test antibody that reduces the binding of control (such as 10F6) antibody to a KIR3DL2 antigen by at least about 30% or more, preferably about 40%, can be considered to be an antibody that binds to substantially the same epitope or determinant as a control (e.g., 10F6). Preferably, such a test antibody will reduce the binding of the control antibody (e.g., 10F6) to the KIR3DL2 antigen by at least about 50% (e. g., at least about 60%, at least about 70%, or more). It will be appreciated that the order of control and test antibodies can be reversed: that is, the control antibody can be first bound to the surface and the test antibody is brought into contact with the surface thereafter in a competition assay. Preferably, the antibody having higher affinity for the KIR3DL2 antigen is bound to the surface first, as it will be expected that the decrease in binding seen for the second antibody (assuming the antibodies are cross-reacting) will be of greater magnitude. Further examples of such assays are provided in, e.g., Saunal (1995) J. Immunol. Methods 183: 33-41, the disclosure of which is incorporated herein by reference. Preferably, monoclonal antibodies that recognize a KIR3DL2 epitope will react with an epitope that is present on a substantial percentage of or even all relevant cells, e.g., malignant CD4+ T cells, cells from a SS or MF patient, but will not significantly react with other cells, i.e., cells that do not express KIR3DL2. In one aspect, the anti-KIR3DL2 antibodies bind KIR3DL2 but do not bind KIR3DL1 and/or KIR3DS1. In some embodiments, the antibodies will bind to KIR3DL2-expressing cells from an individual or individuals with a disease characterized by expression of KIR3DL2-positive cells, i.e. an individual that is a candidate for treatment with one of the herein-described methods using an anti-KIR3DL2 antibody. Accordingly, once an antibody that specifically recognizes KIR3DL2 on cells is obtained, it can be tested for its ability to bind to KIR3DL2-positive cells (e.g. malignant CD4+ T cells) taken from a patient with a disorder such as SS or MF. In particular, prior to treating a patient with one of the present antibodies, it will be beneficial to test the ability of the antibody to bind malignant cells taken from the patient, e.g. in a blood sample, to maximize the likelihood that the therapy will be beneficial in the patient. In one embodiment, the antibodies are validated in an immunoassay to test their ability to bind to KIR3DL2-expressing cells, e.g. malignant CD4+ T cells, pro-inflammatory CD4+ cells. For example, peripheral blood lymphocytes (PBLs) are taken from a plurality of patients, and CD4+ T cells are enriched from the PBLs, e.g., by flow cytometry using relevant antibodies (for malignant CD4+ cells see, e.g., Bagot et al. (2001) Blood 97:1388-1391, the disclosure of which is incorporated herein by reference), or CD4+CD28− cell fractions are isolated by magnetic separation on a MACS column (Miltenyi Biotec). The ability of a given antibody to bind to the cells is then assessed using standard methods well known to those in the art. Antibodies that are found to bind to a substantial proportion (e.g., 20%, 30%, 40%, 50%, 60%, 70%, 80% or more) of cells known to express KIR3DL2, e.g. T cells, from a significant percentage of individuals or patients (e.g., 5%, 10%, 20%, 30%, 40%, 50% or more) are suitable for use herein, both for diagnostic purposes to determine the presence or level of malignant T cells in a patient or for use in the herein-described therapeutic methods, e.g., for use to increase or decrease malignant T cell number or activity. To assess the binding of the antibodies to the cells, the antibodies can either be directly or indirectly labeled. When indirectly labeled, a secondary, labeled antibody is typically added. The binding of the antibodies to the cells can then be detected using, e.g., cytofluorometric analysis (e.g. FACScan). Such methods are well known to those of skill in the art. Determination of whether an antibody binds within an epitope region can be carried out in ways known to the person skilled in the art. As one example of such mapping/characterization methods, an epitope region for an anti-KIR3DL2 antibody may be determined by epitope “foot-printing” using chemical modification of the exposed amines/carboxyls in the KIR3DL2 protein. One specific example of such a foot-printing technique is the use of HXMS (hydrogen-deuterium exchange detected by mass spectrometry) wherein a hydrogen/deuterium exchange of receptor and ligand protein amide protons, binding, and back exchange occurs, wherein the backbone amide groups participating in protein binding are protected from back exchange and therefore will remain deuterated. Relevant regions can be identified at this point by peptic proteolysis, fast microbore high-performance liquid chromatography separation, and/or electrospray ionization mass spectrometry. See, e. g., Ehring H, Analytical Biochemistry, Vol. 267 (2) pp. 252-259 (1999) Engen, J. R. and Smith, D. L. (2001) Anal. Chem. 73, 256A-265A. Another example of a suitable epitope identification technique is nuclear magnetic resonance epitope mapping (NMR), where typically the position of the signals in two-dimensional NMR spectra of the free antigen and the antigen complexed with the antigen binding peptide, such as an antibody, are compared. The antigen typically is selectively isotopically labeled with 15N so that only signals corresponding to the antigen and no signals from the antigen binding peptide are seen in the NMR-spectrum. Antigen signals originating from amino acids involved in the interaction with the antigen binding peptide typically will shift position in the spectrum of the complex compared to the spectrum of the free antigen, and the amino acids involved in the binding can be identified that way. See, e. g., Ernst Schering Res Found Workshop. 2004; (44): 149-67; Huang et al. Journal of Molecular Biology, Vol. 281 (1) pp. 61-67 (1998); and Saito and Patterson, Methods. 1996 June; 9 (3): 516-24. Epitope mapping/characterization also can be performed using mass spectrometry methods. See, e.g., Downward, J Mass Spectrom. 2000 April; 35 (4): 493-503 and Kiselar and Downard, Anal Chem. 1999 May 1; 71 (9): 1792-801. Protease digestion techniques also can be useful in the context of epitope mapping and identification. Antigenic determinant-relevant regions/sequences can be determined by protease digestion, e.g. by using trypsin in a ratio of about 1:50 to KIR3DL2 or o/n digestion at and pH 7-8, followed by mass spectrometry (MS) analysis for peptide identification. The peptides protected from trypsin cleavage by the anti-KIR3DL2 binder can subsequently be identified by comparison of samples subjected to trypsin digestion and samples incubated with antibody and then subjected to digestion by e.g. trypsin (thereby revealing a footprint for the binder). Other enzymes like chymotrypsin, pepsin, etc., also or alternatively can be used in similar epitope characterization methods. Moreover, enzymatic digestion can provide a quick method for analyzing whether a potential antigenic determinant sequence is within a region of the KIR3DL2 polypeptide that is not surface exposed and, accordingly, most likely not relevant in terms of immunogenicity/antigenicity. See, e. g., Manca, Ann Ist Super Sanita. 1991; 27: 15-9 for a discussion of similar techniques. Site-directed mutagenesis is another technique useful for elucidation of a binding epitope. For example, in “alanine-scanning”, each residue within a protein segment is re-placed with an alanine residue, and the consequences for binding affinity measured. If the mutation leads to a significant reduction in binding affinity, it is most likely involved in binding. Monoclonal antibodies specific for structural epitopes (i.e., antibodies which do not bind the unfolded protein) can be used to verify that the alanine-replacement does not influence over-all fold of the protein. See, e.g., Clackson and Wells, Science 1995; 267:383-386; and Wells, Proc Natl Acad Sci USA 1996; 93:1-6. Electron microscopy can also be used for epitope “foot-printing”. For example, Wang et al., Nature 1992; 355:275-278 used coordinated application of cryoelectron micros-copy, three-dimensional image reconstruction, and X-ray crystallography to determine the physical footprint of a Fab-fragment on the capsid surface of native cowpea mosaic virus. Other forms of “label-free” assay for epitope evaluation include surface plasmon resonance (SPR, BIACORE) and reflectometric interference spectroscopy (RifS). See, e.g., Fagerstam et al., Journal Of Molecular Recognition 1990; 3:208-14; Nice et al., J. Chromatogr. 1993; 646:159-168; Leipert et al., Angew. Chem. Int. Ed. 1998; 37:3308-3311; Kroger et al., Biosensors and Bioelectronics 2002; 17:937-944. It should also be noted that an antibody binding the same or substantially the same epitope as an antibody described herein can be identified in one or more of the exemplary competition assays described herein. Optionally, cellular uptake or localization is assessed in order to select an antibody that is readily taken up into the cell and/or into the cellular compartment where it KIR3DL2 is present. Cellular uptake or localization will generally be measured in the cells in which the antibody is sought or believed to exert its activity. Cellular uptake or localization can be assessed by standard methods, such as by confocal staining using an antibody marked with a detectable moiety (e.g. a fluorescent moiety). Upon immunization and production of antibodies in a vertebrate or cell, particular selection steps may be performed to isolate antibodies as claimed. In this regard, in a specific embodiment, provided are methods of producing such antibodies, comprising: (a) immunizing a non-human mammal with an immunogen comprising a KIR3DL2 polypeptide; and (b) preparing antibodies from said immunized animal; and (c) selecting antibodies from step (b) that are capable of binding KIR3DL2. Typically, an anti-KIR3DL2 antibody herein has an affinity for a KIR3DL2 polypeptide in the range of about 104to about 1011M−1(e.g., about 108to about 1010M−1). For example, an antibody can have an average disassociation constant (Kd) of less than 1×10−9M with respect to KIR3DL2, as determined by, e.g., surface plasmon resonance (SPR) screening (such as by analysis with a BIAcore™ SPR analytical device). In a more particular exemplary aspect, an antibody can have a Kdof about 1×10−8M to about 1×10−10M, or about 1×10−9M to about 1×10−11M, for KIR3DL2. Antibodies can be characterized for example by a mean Kdof no more than about (i.e. better affinity than) 100, 60, 10, 5, or 1 nanomolar, preferably sub-nanomolar or optionally no more than about 500, 200, 100 or 10 picomolar. Kdcan be determined for example for example by immobilizing recombinantly produced human KIR3DL2 proteins on a chip surface, followed by application of the antibody to be tested in solution. In one embodiment, the method further comprises a step (d), selecting antibodies from (b) that are capable of competing for binding to KIR3DL2 with antibody 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9. In one aspect of any of the embodiments, the antibodies prepared according to the present methods are monoclonal antibodies. In another aspect, the non-human animal used to produce antibodies according to the methods of the invention is a mammal, such as a rodent, bovine, porcine, fowl, horse, rabbit, goat, or sheep. The antibodies encompass 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9. Additionally, antibodies of can optionally be specified to be antibodies other than any of antibodies Q241 and Q66 (Pende, et al. (1996) J Exp Med 184:505-518), clone 5.133 (Miltenyi Biotec), “AZ158” (Parolini, S., et al. (2002) In Leucocyte typing VII. D. Mason, editor. Oxford University Press, Oxford. 415-417 and WO2010/081890 (e.g. antibodies having the heavy and light chain variable region of SEQ ID NOS: 8 and 10 of WO2010/081890), or derivatives of the foregoing, e.g. that comprise the antigen binding region in whole or in part. According to an alternate embodiment, the DNA encoding an antibody that binds an epitope present on KIR3DL2 polypeptides is isolated from the hybridoma and placed in an appropriate expression vector for transfection into an appropriate host. The host is then used for the recombinant production of the antibody, or variants thereof, such as a humanized version of that monoclonal antibody, active fragments of the antibody, chimeric antibodies comprising the antigen recognition portion of the antibody, or versions comprising a detectable moiety. DNA encoding the monoclonal antibodies, e.g., antibody 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9, can be readily isolated and sequenced using conventional procedures (e. g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). Once isolated, the DNA can be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. As described elsewhere in the present specification, such DNA sequences can be modified for any of a large number of purposes, e.g., for humanizing antibodies, producing fragments or derivatives, or for modifying the sequence of the antibody, e.g., in the antigen binding site in order to optimize the binding specificity of the antibody. Recombinant expression in bacteria of DNA encoding the antibody is well known in the art (see, for example, Skerra et al., Curr. Opinion in Immunol., 5, pp. 256 (1993); and Pluckthun, Immunol. 130, p. 151 (1992). Assessing Activity Once an antigen-binding compound is obtained it will generally be assessed for its ability to internalize into KIR3DL2-expressing target cells or cause KIR3DL2 internalization into KIR3DL2-expressing target cells, to increase the number of KIR3DL2 polypeptides at the surface of a cell, to induce ADCC or CDC towards, to inhibit the pro-inflammatory activity and/or proliferation of and/or cause the elimination of KIR3DL2-expressing target cells. Assessing the antigen-binding compound's ability to internalize or to induce ADCC, CDC or generally lead to the elimination or inhibition of activity of KIR3DL2-expressing target cells, can be carried out at any suitable stage of the method, e.g. as in the examples are provided herein. This assessment can be useful at one or more of the various steps involved in the identification, production and/or development of an antibody (or other compound) destined for therapeutic use. For example, activity may be assessed in the context of a screening method to identify candidate antigen-binding compounds, or in methods where an antigen-binding compound is selected and made human suitable (e.g. made chimeric or humanized in the case of an antibody), where a cell expressing the antigen-binding compound (e.g. a host cell expressing a recombinant antigen-binding compound) has been obtained and is assessed for its ability to produce functional antibodies (or other compounds), and/or where a quantity of antigen-binding compound has been produced and is to be assessed for activity (e.g. to test batches or lots of product). Generally the antigen-binding compound will be known to specifically bind to a KIR3DL2 polypeptide. The step may involve testing a plurality (e.g., a very large number using high throughput screening methods or a smaller number) of antigen-binding compounds. As used herein, an anti-KIR3DL2 antibody that is not “internalized” or that does not “internalize” is one that is not substantially taken up by (i.e., enters) the cell upon binding to KIR3DL2 on a mammalian cell (i.e. cell surface KIR3DL2). The non-internalizing antibody will of course include antibody fragments, human or humanized antibody and antibody conjugate. Whether an anti-KIR3DL2 antibody internalizes upon binding KIR3DL2 on a mammalian cell, or whether a KIR3DL2 polypeptide undergoes intracellular internalization (e.g. upon being bound by an antibody) can be determined by various assays including those described in the experimental examples herein. For example, to test internalization in vivo, the test antibody is labeled and introduced into an animal known to have KIR3DL2 expressed on the surface of certain cells. The antibody can be radiolabeled or labeled with fluorescent or gold particles, for instance. Animals suitable for this assay include a mammal such as a nude mouse that contains a human KIR3DL2-expressing tumor transplant or xenograft, or a mouse into which cells transfected with human KIR3DL2 have been introduced, or a transgenic mouse expressing the human KIR3DL2 transgene. Appropriate controls include animals that did not receive the test antibody or that received an unrelated antibody, and animals that received an antibody to another antigen on the cells of interest, which antibody is known to be internalized upon binding to the antigen. The antibody can be administered to the animal, e.g., by intravenous injection. At suitable time intervals, tissue sections of the animal can be prepared using known methods or as described in the experimental examples below, and analyzed by light microscopy or electron microscopy, for internalization as well as the location of the internalized antibody in the cell. For internalization in vitro, the cells can be incubated in tissue culture dishes in the presence or absence of the relevant antibodies added to the culture media and processed for microscopic analysis at desired time points. The presence of an internalized, labeled antibody in the cells can be directly visualized by microscopy or by autoradiography if radiolabeled antibody is used. Optionally, in microscopy, co-localization with a known polypeptide or other cellular component can be assessed; for example co-localization with endosomal/lysosomal marker LAMP-1 (CD107a) can provide information about the subcellular localization of the internalized antibody. Alternatively, in a quantitative biochemical assay, a population of cells comprising KIR3DL2-expressing cells are contacted in vitro or in vivo with a radiolabeled test antibody and the cells (if contacted in vivo, cells are then isolated after a suitable amount of time) are treated with a protease or subjected to an acid wash to remove uninternalized antibody on the cell surface. The cells are ground up and the amount of protease resistant, radioactive counts per minute (cpm) associated with each batch of cells is measured by passing the homogenate through a scintillation counter. Based on the known specific activity of the radiolabeled antibody, the number of antibody molecules internalized per cell can be deduced from the scintillation counts of the ground-up cells. Cells are “contacted” with antibody in vitro preferably in solution form such as by adding the cells to the cell culture media in the culture dish or flask and mixing the antibody well with the media to ensure uniform exposure of the cells to the antibody. Testing whether an antibody is capable of increasing the number of KIR3DL2 polypeptides at the surface of a cell can be carried out by incubating the test antibody with a KIR3DL2-expressing cell (e.g. a T cell lymphoma) and detecting KIR3DL2 polypeptides at the surface of the cell after the incubation period. KIR3DL2 polypeptides can be carried out using a suitable affinity regent, e.g. one or more antibodies. Exemplary assays are shown in Example 4. For example, an antibody may induce an increase of at least 20%, 50%, 75% or 100% of the number of KIR3DL2 polypeptides detectable at the surface of cells after incubation (e.g. for at least 1, 3, 6, 12, 24 or 48 hours) in the presence of test antibody, compared to a control antibody (e.g. an antibody not binding to KIR3DL2, a different anti-KIR3DL2 antibody). Optionally, the number of KIR3DL2 polypeptides detectable at the surface of cells after incubation is the number detectable using the test antibody. Optionally, the number of KIR3DL2 polypeptides detectable at the surface of cells after incubation is the number detectable using a second anti-KIR3DL2 antibody that does not compete with the test antibody for binding to KIR3DL2. Testing CDC and ADCC can be carried out can be determined by various assays including those described in the experimental examples herein (see Examples 4 and 5). Testing ADCC typically involves assessing cell-mediated cytotoxicity in which a KIR3DL2-expressing target cell (e.g. a Cou-L cell, Sézary Syndrome cell or other KIR3DL2-expressing cell) with bound anti-KIR3DL2 antibody is recognized by an effector cell bearing Fc receptors, without the involvement of complement. A cell which does not express a KIR3DL2 antigen can optionally be used as a control. Activation of NK cell cytotoxicity is assessed by measuring an increase in cytokine production (e.g. IFN-γ production) or cytotoxicity markers (e.g. CD107 mobilization). Preferably the antibody will induce an increase in cytokine production, expression of cytotoxicity markers, or target cell lysis of at least 20%, 50%, 80%, 100%, 200% or 500% in the presence of target cells, compared to a control antibody (e.g. an antibody not binding to KIR3DL2, a KIR3DL2 antibody having murine constant regions). In another example, lysis of target cells is detected, e.g. in a chromium release assay, preferably the antibody will induce lysis of at least 10%, 20%, 30%, 40% or 50% of target cells. Where an antigen-binding compound is tested for both its ability to (a) induce both ADCC and (b) internalize into KIR3DL2-expressing cells and/or induce KIR3DL2 internalization, the assays of (a) and (b) can be carried out in any order. However, greater the extent and speed of internalization will generally be expected to be associated with a decrease of the extent of CDC and ADCC activity. Antibody 10F6 The amino acid sequence of the heavy chain variable region of antibody 10F6 is listed as SEQ ID NO: 2, the amino acid sequence of the light chain variable region is listed as SEQ ID NO: 3. In a specific embodiment, provided is an antibody that binds essentially the same epitope or determinant as monoclonal antibodies 10F6; optionally the antibody comprises an antigen binding region of antibody 10F6. In any of the embodiments herein, antibody 10F6 can be characterized by its amino acid sequence and/or nucleic acid sequence encoding it. In one embodiment, the monoclonal antibody comprises the Fab or F(ab′)2portion of 10F6. Also provided is a monoclonal antibody that comprises the heavy chain variable region of 10F6. According to one embodiment, the monoclonal antibody comprises the three CDRs of the heavy chain variable region of 10F6 Also provided is a monoclonal antibody that further comprises the variable light chain variable region of 10F6 or one, two or three of the CDRs of the light chain variable region of 10F6. Optionally any one or more of said light or heavy chain CDRs may contain one, two, three, four or five or more amino acid modifications (e.g. substitutions, insertions or deletions). Optionally, provided is an antibody where any of the light and/or heavy chain variable regions comprising part or all of an antigen binding region of antibody 10F6 are fused to an immunoglobulin constant region of the human IgG type, optionally a human constant region, optionally a human IgG1 or IgG3 isotype. In another aspect, provided is a purified polypeptide which encodes an antibody, wherein the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFTIAGMQ as set forth in SEQ ID NO: 6, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. IAGMQ (SEQ ID NO: 4), GYTFTI (SEQ ID NO: 5)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence WINTHSGVPKYAEDFKG as set forth in SEQ ID NO: 7, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. WINTHSGVPK (SEQ ID NO: 8)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence GGDEGVMDY as set forth in SEQ ID NO: 9, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence KASQDVSTAVA as set forth in SEQ ID NO: 10, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence WASTRHT as set forth in SEQ ID NO: 11, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region comprising an amino acid sequence QQHYNTPWT as set forth in SEQ ID NO: 12, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid. In another aspect, provided is an antibody that binds human KIR3DL2, comprising:(a) the heavy chain variable region of SEQ ID NO: 2, optionally wherein one, two, three or more residues may be substituted by a different amino acid; and/or(b) the light chain variable region of SEQ ID NO: 3, optionally wherein one, two, three or more residues may be substituted by a different amino acid; and/or(c) the heavy chain variable region of SEQ ID NO: 2, wherein one or more of these amino acids may be substituted by a different amino acid; and the light chain variable region of SEQ ID NO: 3, optionally wherein one, two, three or more residues may be substituted by a different amino acid; and/or(d) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 4-6, 7-8 and 9, respectively, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and/or(e) the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 10, 11 and 12, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and/or(f) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 4, 7 and 9, respectively, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and the light chain CDRs 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 10, 11 and 12, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid. In another aspect of any of the embodiments herein, any of the CDRs 1, 2 and 3 of the heavy and light chains may be characterized by a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, and/or as having an amino acid sequence that shares at least 50%, 60%, 70%, 80%, 85%, 90% or 95% sequence identity with the particular CDR or set of CDRs listed in the corresponding SEQ ID NO. In another aspect, provided is an antibody that competes for KIR3DL2 binding with a monoclonal antibody of (a) to (f), above. Antibody 2B12 The amino acid sequence of the heavy chain variable region of antibody 2B12 is listed in SEQ ID NO: 13, the amino acid sequence of the light chain variable region is listed as SEQ ID NO: 14. In one embodiment, provided is an antibody that binds essentially the same epitope or determinant as monoclonal antibodies 2B12; optionally the antibody comprises an antigen binding region of antibody 2B12. In any of the embodiments herein, antibody 2B12 can be characterized by its amino acid sequence and/or nucleic acid sequence encoding it. In one embodiment, the monoclonal antibody comprises the Fab or F(ab′)2portion of 2B12. Also provided is a monoclonal antibody that comprises the heavy chain variable region of 2B12. According to one embodiment, the monoclonal antibody comprises the three CDRs of the heavy chain variable region of 2B12. Also provided is a monoclonal antibody that further comprises the variable light chain variable region of 2B12 or one, two or three of the CDRs of the light chain variable region of 2B12. Optionally any one or more of said light or heavy chain CDRs may contain one, two, three, four or five amino acid modifications (e.g. substitutions, insertions or deletions). Optionally, provided is an antibody where any of the light and/or heavy chain variable regions comprising part or all of an antigen binding region of antibody 2B12 are fused to an immunoglobulin constant region of the IgG type, optionally a human constant region, optionally an IgG1 or IgG4 isotype. In another aspect, provided is a purified polypeptide which encodes an antibody, wherein the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFTTAGMQ as set forth in SEQ ID NO: 17, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g., TAGMQ (SEQ ID NO: 15), GYTFTT (SEQ ID NO: 16)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence WINSHSGVPKYAEDFK as set forth in SEQ ID NO: 18, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. WINSHSGVP (SEQ ID NO: 19)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence GGDEGVMDYW as set forth in SEQ ID NO: 20, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence KASQDVSTAVA as set forth in SEQ ID NO: 10, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence WTSTRHT as set forth in SEQ ID NO: 21, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; and/or a LCDR3 region comprising an amino acid sequence QQHYSTPWT as set forth in SEQ ID NO: 22, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid, or where the sequence may comprise an insertion of one or more amino acids. In another aspect, provided is an antibody that binds human KIR3DL2, comprising:(a) the heavy chain variable region of SEQ ID NO: 13, optionally wherein one, two, three or more of amino acid residues may be substituted by a different amino acid; and/or(b) the light chain variable region of SEQ ID NO: 14, optionally wherein one, two, three or more of amino acid residues may be substituted by a different amino acid; and/or(c) the heavy chain variable region of SEQ ID NO: 13, optionally wherein one, two, three or more of amino acid residues may be substituted by a different amino acid; and the light chain variable region of SEQ ID NO: 14, optionally wherein one or more of amino acid residues may be substituted by a different amino acid; and/or(d) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 15-17, 18-19 and 20, respectively, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and/or(e) the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 10, 21 and 22, respectively, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and/or(f) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 15, 18 and 20, respectively, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 10, 21 and 22, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid. In another aspect of any of the embodiments herein, any of the CDRs 1, 2 and 3 of the heavy and light chains may be characterized by a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, and/or as having an amino acid sequence that shares at least 50%, 60%, 70%, 80%, 85%, 90% or 95% sequence identity with the particular CDR or set of CDRs listed in the corresponding SEQ ID NO. In another aspect, provided is an antibody that competes for KIR3DL2 binding with a monoclonal antibody of (a) to (f), above. Antibody 10G5 The amino acid sequence of the heavy chain variable region of antibody 10G5 is listed as SEQ ID NO: 23, the amino acid sequence of the light chain variable region is listed as SEQ ID NO: 24. In a specific embodiment, provided is an antibody that binds essentially the same epitope or determinant as monoclonal antibodies 10G5; optionally the antibody comprises an antigen binding region of antibody 10G5. In any of the embodiments herein, antibody 10G5 can be characterized by its amino acid sequence and/or nucleic acid sequence encoding it. In one embodiment, the monoclonal antibody comprises the Fab or F(ab′)2portion of 10G5. Also provided is a monoclonal antibody that comprises the heavy chain variable region of 10G5. According to one embodiment, the monoclonal antibody comprises the three CDRs of the heavy chain variable region of 10G5 Also provided is a monoclonal antibody that further comprises the variable light chain variable region of 10G5 or one, two or three of the CDRs of the light chain variable region of 10G5. Optionally any one or more of said light or heavy chain CDRs may contain one, two, three, four or five or more amino acid modifications (e.g. substitutions, insertions or deletions). Optionally, provided is an antibody where any of the light and/or heavy chain variable regions comprising part or all of an antigen binding region of antibody 10G5 are fused to an immunoglobulin constant region of the human IgG type, optionally a human constant region, optionally a human IgG1 or IgG3 isotype. In another aspect, provided is a purified polypeptide which encodes an antibody, wherein the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFTSYTMH as set forth in SEQ ID NO: 27, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. SYTMH (SEQ ID NO: 25), GYTFTS (SEQ ID NO: 26)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence YINPSSGYTENNRKF as set forth in SEQ ID NO: 28, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. YINPSSGY (SEQ ID NO: 29)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence RLGKGLLPPFDY as set forth in SEQ ID NO: 30, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence RASENIYSNLA as set forth in SEQ ID NO: 31, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence AATNLAD as set forth in SEQ ID NO: 32, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region comprising an amino acid sequence QHFWGTPYT as set forth in SEQ ID NO: 33, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid. In another aspect, provided is an antibody that binds human KIR3DL2, comprising:(a) the heavy chain variable region of SEQ ID NO: 23, optionally wherein one, two, three or more residues may be substituted by a different amino acid; and/or(b) the light chain variable region of SEQ ID NO: 24, optionally wherein one, two, three or more residues may be substituted by a different amino acid; and/or(c) the heavy chain variable region of SEQ ID NO: 23, optionally wherein one or more residues may be substituted by a different amino acid; and the light chain variable region of SEQ ID NO: 24 wherein one, two, three or more residues may be substituted by a different amino acid; and/or(d) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NO: 25-27, 28-29 and 30, respectively, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and/or(e) the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 31, 32, and 33, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and/or(f) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 25, 28 and 30, respectively, optionally wherein one or more residues of any CDR may be substituted by a different amino acid; and the light chain CDRs 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 31, 32, and 33, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid. In another aspect of any of the embodiments herein, any of the CDRs 1, 2 and 3 of the heavy and light chains may be characterized by a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, and/or as having an amino acid sequence that shares at least 50%, 60%, 70%, 80%, 85%, 90% or 95% sequence identity with the particular CDR or set of CDRs listed in the corresponding SEQ ID NO. In another aspect, provided is an antibody that competes for KIR3DL2 binding with a monoclonal antibody of (a) to (f), above. Antibody 13H1 The amino acid sequence of the heavy chain variable region of antibody 13H1 is listed as SEQ ID NO: 34, the amino acid sequence of the light chain variable region is listed as SEQ ID NO: 35. In a specific embodiment, provided is an antibody that binds essentially the same epitope or determinant as monoclonal antibodies 13H1; optionally the antibody comprises an antigen binding region of antibody 13H1. In any of the embodiments herein, antibody 13H1 can be characterized by its amino acid sequence and/or nucleic acid sequence encoding it. In one embodiment, the monoclonal antibody comprises the Fab or F(ab′)2portion of 13H1. Also provided is a monoclonal antibody that comprises the heavy chain variable region of 13H1. According to one embodiment, the monoclonal antibody comprises the three CDRs of the heavy chain variable region of 13H1. Also provided is a monoclonal antibody that further comprises the variable light chain variable region of 13H1 or one, two or three of the CDRs of the light chain variable region of 13H1. Optionally any one or more of said light or heavy chain CDRs may contain one, two, three, four or five or more amino acid modifications (e.g. substitutions, insertions or deletions). Optionally, provided is an antibody where any of the light and/or heavy chain variable regions comprising part or all of an antigen binding region of antibody 13H1 are fused to an immunoglobulin constant region of the human IgG type, optionally a human constant region, optionally a human IgG1 or IgG3 isotype. In another aspect, provided is a purified polypeptide which encodes an antibody, wherein the antibody comprises: a HCDR1 region comprising an amino acid sequence HYSFIGYTM as set forth in SEQ ID NO: 38, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. GYTMN (SEQ ID NO: 36), HYSFIG (SEQ ID NO: 37)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence LINPYNGDTTYNQKFKG as set forth in SEQ ID NO: 39, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. LINPYNGDTT (SEQ ID NO: 40)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence ENWGYPYAMDY as set forth in SEQ ID NO: 41, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence RASESVDNFGISFMN as set forth in SEQ ID NO: 42, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence AASNQGS as set forth in SEQ ID NO: 43, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region comprising an amino acid sequence QQSKEVPYT as set forth in SEQ ID NO: 44, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid. In another aspect, provided is an antibody that binds human KIR3DL2, comprising:(a) the heavy chain variable region of SEQ ID NO: 34, optionally wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or(b) the light chain variable region of SEQ ID NO: 35, optionally wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or(c) the heavy chain variable region of SEQ ID NO: 34, optionally wherein one or more amino acid residues may be substituted by a different amino acid; and the light chain variable region of SEQ ID NO: 35, wherein one, two, three or more of these amino acids may be substituted by a different amino acid; and/or(d) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 36-38, 39-40 and 41, respectively, optionally wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and/or(e) the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 42, 43 and 44, optionally wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and/or(f) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 36, 39 and 41, respectively, optionally wherein one or more amino acid residues of any CDR may be substituted by a different amino acid; and the light chain CDRs 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 42, 43 and 44, optionally wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid. In another aspect of any of the embodiments herein, any of the CDRs 1, 2 and 3 of the heavy and light chains may be characterized by a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, and/or as having an amino acid sequence that shares at least 50%, 60%, 70%, 80%, 85%, 90% or 95% sequence identity with the particular CDR or set of CDRs listed in the corresponding SEQ ID NO. In another aspect, provided is an antibody that competes for KIR3DL2 binding with a monoclonal antibody of (a) to (f), above. Antibody 1E2 The amino acid sequence of the heavy chain variable region of antibody 1E2 is listed as SEQ ID NO: 45, the amino acid sequence of the light chain variable region is listed as SEQ ID NO: 46. In a specific embodiment, provided is an antibody that binds essentially the same epitope or determinant as monoclonal antibodies 1E2; optionally the antibody comprises an antigen binding region of antibody 1E2. In any of the embodiments herein, antibody 1E2 can be characterized by its amino acid sequence and/or nucleic acid sequence encoding it. In one embodiment, the monoclonal antibody comprises the Fab or F(ab′)2portion of 1E2. Also provided is a monoclonal antibody that comprises the heavy chain variable region of 1E2. According to one embodiment, the monoclonal antibody comprises the three CDRs of the heavy chain variable region of 1E2 Also provided is a monoclonal antibody that further comprises the variable light chain variable region of 1E2 or one, two or three of the CDRs of the light chain variable region of 1E2. Optionally any one or more of said light or heavy chain CDRs may contain one, two, three, four or five or more amino acid modifications (e.g. substitutions, insertions or deletions). Optionally, provided is an antibody where any of the light and/or heavy chain variable regions comprising part or all of an antigen binding region of antibody 1E2 are fused to an immunoglobulin constant region of the human IgG type, optionally a human constant region, optionally a human IgG1 or IgG3 isotype. In another aspect, provided is a purified polypeptide which encodes an antibody, wherein the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFTDYAMN as set forth in SEQ ID NO: 49, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. DYAMN (SEQ ID NO: 47), GYTFTD (SEQ ID NO: 48)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence VISTYYGDANYNQKFKG as set forth in SEQ ID NO: 50, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. VISTYYGDAN (SEQ ID NO: 51)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence IYYDYDGSY as set forth in SEQ ID NO: 52, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence RSSQSLVHSNGNTYLH as set forth in SEQ ID NO: 53, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence KVSNRFS as set forth in SEQ ID NO: 54, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region comprising an amino acid sequence SQSTHVPPYT as set forth in SEQ ID NO: 55, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid. In another aspect, provided is an antibody that binds human KIR3DL2, comprising:(a) the heavy chain variable region of SEQ ID NO: 42, optionally wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or(b) the light chain variable region of SEQ ID NO: 43, optionally wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or(c) the heavy chain variable region of SEQ ID NO: 42, optionally wherein one or more amino acid residues may be substituted by a different amino acid; and the light chain variable region of SEQ ID NO: 43, optionally wherein one, two, three or more of these amino acids may be substituted by a different amino acid; and/or(d) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 47-49, 50-51 and 52, respectively, optionally wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and/or(e) the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 53, 54 and 55, optionally wherein one, two, three amino acid residues of any CDR may be substituted by a different amino acid; and/or(f) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 47, 50 and 52, optionally wherein one or more amino acid residues of any CDR may be substituted by a different amino acid; and the light chain CDRs 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 53, 54 and 55, optionally wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid. In another aspect of any of the embodiments herein, any of the CDRs 1, 2 and 3 of the heavy and light chains may be characterized by a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, and/or as having an amino acid sequence that shares at least 50%, 60%, 70%, 80%, 85%, 90% or 95% sequence identity with the particular CDR or set of CDRs listed in the corresponding SEQ ID NO. In another aspect, provided is an antibody that competes for KIR3DL2 binding with a monoclonal antibody of (a) to (f), above. Antibody 9E10 The amino acid sequence of the heavy chain variable region of 9E10 is listed as SEQ ID NO: 56, the amino acid sequence of the light chain variable regions (two alternative light chains available) of 9E10 are listed as SEQ ID NOS: 57 and 67. In a specific embodiment, provided is an antibody that binds essentially the same epitope or determinant as monoclonal antibodies 9E10; optionally the antibody comprises an antigen binding region of antibody 9E10. In any of the embodiments herein, antibody 9E10 can be characterized by its amino acid sequence and/or nucleic acid sequence encoding it. In one embodiment, the monoclonal antibody comprises the Fab or F(ab′)2 portion of 9E10. Also provided is a monoclonal antibody that comprises the heavy chain variable region of 9E10. According to one embodiment, the monoclonal antibody comprises the three CDRs of the heavy chain variable region of 9E10. Also provided is a monoclonal antibody that further comprises the variable light chain variable region of 9E10 or one, two or three of the CDRs of the light chain variable region of 9E10. Optionally any one or more of said light or heavy chain CDRs may contain one, two, three, four or five amino acid modifications (e.g. substitutions, insertions or deletions). Optionally, provided is an antibody where any of the light and/or heavy chain variable regions comprising part or all of an antigen binding region of antibody 9E10 are fused to an immunoglobulin constant region of the IgG type, optionally a human constant region, optionally a human IgG1 or IgG4 isotype. In another aspect, provided is a purified polypeptide which encodes an antibody, wherein the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFTSYTMH as set forth in SEQ ID NO: 60, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g., SYTMH (SEQ ID NO: 58), GYTFTS (SEQ ID NO: 59)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence YINPSSGYTDYNQKFKD as set forth in SEQ ID NO: 61, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. YINPSSGYTD (SEQ ID NO: 62)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence LGKGLLPPFDY as set forth in SEQ ID NO: 63, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence KSNQNLLWSGNQRYCLV as set forth in SEQ ID NO: 64, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence WTSDRYS as set forth in SEQ ID NO: 65, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region comprising an amino acid sequence QQHLHIPYT as set forth in SEQ ID NO: 66, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid, or where the sequence may comprise an insertion of one or more amino acids. In another aspect, provided is an antibody that binds human KIR3DL2, comprising:(a) the heavy chain variable region of SEQ ID NO: 56, optionally wherein one, two, three or amino acid residues may be substituted by a different amino acid; and/or(b) the light chain variable region of SEQ ID NOS: 57 or 67, optionally wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or(c) the heavy chain variable region of SEQ ID NO: 56, optionally wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and the light chain variable region of SEQ ID NOS: 57 or 67, optionally wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or(d) the heavy chain CDR 1 and 2 (HCDR1, HCDR2) amino acid sequences as shown in SEQ ID NOS: 58, 59 or 60, 61-62 and 63, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and/or(e) the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 64, 65 and 66, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid; and/or(f) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 58, 61 and 63, optionally wherein one, two, three residues of any CDR may be substituted by a different amino acid; and the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 64, 65 and 66, optionally wherein one, two, three or more residues of any CDR may be substituted by a different amino acid. In another embodiment, provided is antibody 1C3 (anti-D2 domain), its variable region and CDRs. In one embodiment, provided is an antibody having respectively a VH and VL region of SEQ ID NOS: 170 and 171 (1C3). In one embodiment, provided is an antibody having a heavy chain comprising CDRs 1, 2 and 3 (HCDR1, HCDR2, HCDR3) comprising a sequence of SEQ ID NO: 172, 173 or 174 (HCDR1), SEQ ID NO: 175 or 176 (HCDR2) and SEQ ID NO: 177 (HCDR3), respectively, wherein each CDR may optionally comprise 1, 2, 3 or 4 amino acid substitutions, deletions or insertions. In one embodiment, provided is an antibody having (i) a heavy chain comprising CDRs 1, 2 and 3 (HCDR1, HCDR2, HCDR3) comprising a sequence of SEQ ID NO: 172, 173 or 174 (HCDR1), SEQ ID NO: 175 or 176 (HCDR2) and SEQ ID NO: 177 (HCDR3), respectively, and (ii) a light chain comprising CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) comprising a sequence of SEQ ID NO: 178, 179 or 180, respectively, wherein each CDR may optionally comprise 1, 2, 3 or 4 amino acid substitutions, deletions or insertions. In another embodiment, provided is antibody 20E9 (anti-D2 domain), its variable region and CDRs. In one embodiment, provided is an antibody having respectively a VH and VL region of SEQ ID NOS: 181 and 182 (20E9). In one embodiment, provided is an antibody having a heavy chain comprising CDRs 1, 2 and 3 (HCDR1, HCDR2, HCDR3) comprising a sequence of SEQ ID NO: 183, 184 or 185 (HCDR1), SEQ ID NO: 186 or 187 (HCDR2) and SEQ ID NO: 188 (HCDR3), respectively, wherein each CDR may optionally comprise 1, 2, 3 or 4 amino acid substitutions, deletions or insertions. In one embodiment, provided is an antibody having (i) a heavy chain comprising CDRs 1, 2 and 3 (HCDR1, HCDR2, HCDR3) comprising a sequence of SEQ ID NO: 183, 184 or 185 (HCDR1), SEQ ID NO: 186 or 187 (HCDR2) and SEQ ID NO: 188 (HCDR3), respectively, and (ii) a light chain comprising CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) comprising a sequence of SEQ ID NO: 189, 190 or 191, respectively, wherein each CDR may optionally comprise 1, 2, 3 or 4 amino acid substitutions, deletions or insertions. In another aspect of any of the embodiments herein, any of the CDRs 1, 2 and 3 of the heavy and light chains may be characterized by a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, and/or as having an amino acid sequence that shares at least 50%, 60%, 70%, 80%, 85%, 90% or 95% sequence identity with the particular CDR or set of CDRs listed in the corresponding SEQ ID NO. In another aspect, provided is an antibody that competes for KIR3DL2 binding with a monoclonal antibody above. In any of the antibodies, the specified variable region and CDR sequences may comprise one, two, three, four, five or more conservative sequence modifications. Conservative sequence modifications refers to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are typically those in which an amino acid residue is replaced with an amino acid residue having a side chain with similar physicochemical properties. Specified variable region and CDR sequences may comprise one, two, three, four or more amino acid insertions, deletions or substitutions. Where substitutions are made, substitutions can optionally be conservative modifications. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g. threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, one or more amino acid residues within the CDR regions of an antibody can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for retained function (i.e., the properties set forth herein) using the assays described herein. The term “identity” or “identical”, when used in a relationship between the sequences of two or more polypeptides, refers to the degree of sequence relatedness between polypeptides, as determined by the number of matches between strings of two or more amino acid residues. “Identity” measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., “algorithms”). Identity of related polypeptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M ed Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M. Stockton Press, New York, 1991; and Carillo et al., SIAM J. Applied Math. 48, 1073 (1988). Preferred methods for determining identity are designed to give the largest match between the sequences tested. Methods of determining identity are described in publicly available computer programs. Preferred computer program methods for determining identity between two sequences include the GCG program package, including GAP (Devereux et al., Nucl. Acid. Res. 12, 387 (1984); Genetics Computer Group, University of Wisconsin, Madison, Wis.), BLASTP, BLASTN, and FASTA (Altschul et al., J. Mol. Biol. 215, 403-410 (1990)). The BLASTX program is publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST Manual, Altschul et al. NCB/NLM/NIH Bethesda, Md. 20894; Altschul et al., supra). The well known Smith Waterman algorithm may also be used to determine identity. The sequences of the CDRs of antibodies, according to AbM (Oxford Molecular's AbM antibody modelling software definition), Kabat and Chothia definitions systems, have been summarized in Table 1 for heavy chain CDRs, and in Table 2 below for light chain CDRs (light chain CDRs are the same for each of AbM, Kabat and Chothia definitions). The amino acids sequences described herein are numbered according to Abm, Kabat and Chothia numbering systems. While any suitable numbering system may be used to designated CDR regions, in the absence of any other indication, Abm numbering can be used. Such numbering has been established using the following indications: CDR-L1: Start: approx. residue 24, residue before: always a Cys, residue after: always a Trp (typically Trp-Tyr-Gln, but also, Trp-Leu-Gln, Trp-Phe-Gln, Trp-Tyr-Leu), length: 10 to 17 residues; CDR-L2: Start: always 16 residues after the end of L1, Residues before: generally Ile-Tyr (but also, Val-Tyr, Ile-Lys, Ile-Phe), Length: always 7 residues; CDR-L3, Start: always 33 residues after end of L2, Residue before: always Cys, Residues after: always Phe-Gly-Xaa-Gly, Length: 7 to 11 residues; CDR-H1, Start: approx. residue 26 (always 4 after a Cys) (Chothia/AbM definition, the Kabat definition starts 5 residues later), Residues before: always Cys-Xaa-Xaa-Xaa, Residues after: always a Trp (typically Trp-Val, but also, Trp-Ile, Trp-Ala), Length: 10 to 12 residues (AbM definition, Chothia definition excludes the last 4 residues); CDR-H2, Start: always 15 residues after the end of Kabat/AbM definition of CDR-H1, Residues before: typically Leu-Glu-Trp-Ile-Gly (SEQ ID NO: 192) (but a number of variations, Residues after Lys/Arg-Leu/Ile/Val/Phe/Thr/Ala-Thr/Ser/Ile/Ala), Length: Kabat definition 16 to 19 residues; AbM (and Chothia) definition ends 7 residues earlier; CDR-H3, Start: always 33 residues after end of CDR-H2 (always 2 after a Cys), Residues before: always Cys-Xaa-Xaa (typically Cys-Ala-Arg), Residues after: always Trp-Gly-Xaa-Gly, Length: 3 to 25 residues. In one embodiment, the antibodies are of the human or mouse IgG1 isotype. In another embodiment, the antibodies are of the human IgG1 isotype In an embodiment, the antibodies are antibody fragments that retain their binding and/or functional properties. In one embodiment, the antibody is an antibody having the Kabat, Chotia or AbM heavy and light chain CDR1, CDR2 and CDR3 of any of the antibodies as shown in Table 1 below. TABLE 1CDRHCDR1HCDR2HCDR3defini-SEQSEQSEQmAbtionIDSequenceIDSequenceIDSequence10F6Kabat4IAGMQ7WINTHSG9GGDEGVPKYAEDVMDYFKGChotia5GYTFTI8WINTHSG9GGDEGVPKVMDYAbM6GYTFT8WINTHSG9GGDEGIAGMQVPKVMDY2B12Kabat15TAGMQ18WINSHSG20GGDEGVPKYAEDVMDYWFKChotia16GYTFTT19WINSHSG20GGDEGVPVMDYWAbM17GYTFT19WINSHSG20GGDEGTAGMQVPVMDYW10G5Kabat25SYTMH28YINPSSGY30RLGKGLTENNRKFLPPFDYChotia26GYTFTS29YINPSSGY30RLGKGLLPPFDYAbM27GYTFT29YINPSSGY30RLGKGLSYTMHLPPFDY13H1Kabat36GYTMN39LINPYNG41ENWGYPDTTYNQKYAMDYFKGChotia37HYSFIG40LINPYNG41ENWGYPDTTYAMDYAbM38HYSFI40LINPYNG41ENWGYPGYTMNDTTYAMDY1E2Kabat47DYAMN50VISTYYG52IYYDYDANYNQKDGSYFKGChotia48GYTFTD51VISTYYG52IYYDYDANDGSYAbM49GYTFT51VISTYYG52IYYDYDYAMNDANDGSY9E10Kabat58SYTMH61YINPSSG63LGKGLLYTDYNQKPPFDYFKDChotia59GYTFTS62YINPSSG63LGKGLLYTDPPFDYAbM60GYTFT62YINPSSG63LGKGLLSYTMHYTDPPFDY1C3Kabat172SYWMQ175AIYPGDG177RYDGYDTRYTQKYHFDYFKGChotia173GYTFTS176AIYPGDG177RYDGYDTRYHFDYAbM174GYTFT176AIYPGDG177RYDGYSYWMQDTRYHFDY20E9Kabat183TYWMQ186AIYPGDG188RGDYGNDTRYTQKYGMDYFKGChotia184GFTFTT187AIYPGDG188RGDYGNDTRYGMDYAbM185GFTFT187AIYPGDG188RGDYGNTYWMQDTRYGMDY TABLE 2LCDR1LCDR2LCDR3SEQSEQSEQmAbIDSequenceIDSequenceIDSequence10F610KASQDVSTAVA11WASTRHT12QQHYNTPWT2B1210KASQDVSTAVA21WTSTRHT22QQHYSTPWT10G531RASENIYSNLA32AATNLAD33QHFWGTPYT13H142RASESVDNFGI43AASNQGS44QQSKEVPYTSFMN1E253RSSQSLVHSNG54KVSNRFS55SQSTHVPPYTNTYLH9E1064KSNQNLLWSGN65WTSDRYS66QQHLHIPYTQRYCLV1C3178KSSQSLLWSVN179GASIRES180QHNHGSFLPLTQKNYLS20E9189RSSQSIVHSNG190KVSNHFS191FQGSHVPPTNTYLE The sequences of the variable chains of the antibodies are listed in Table 3 below, with the CDRs underlined. In any embodiment herein, a VL or VH sequence can be specified or numbered so as to contain or lack a signal peptide or any part thereof. TABLE 3SEQAntibodyIDportionNO:10F6 VH2QIQLVQSGPELKKPGETVRISCKASGYTFTIAGMQWVQKMPGKGLKWIGWINTHSGVPKYAEDFKGRFAFSLETSANIAYLQISNLKNEDTATYFCARGGDEGVMDYWGQGTSVTVS10F6 VL3DIVMTQSHKFMSTSVGDRVSITCKASQDVSTAVAWYHQKPGQSPKLLIYWASTRHTGVPDRFSGSGSGTDYTLTISALQAEDLALYYCQQHYNTPWTFGGGTKLEIK2B12 VH13QIQLVQSGPELKKPGETVRISCKASGYTFTTAGMQWVQKTPGKGLKWIGWINSHSGVPKYAEDFKGRFAFSLETSASTAYLQISTLKNEDTATYFCARGGDEGVMDYWGQGTSVTVS2B12 VL14DIVMTQSHKFMSTSLGDRVSFTCKASQDVSTAVAWYQQKPGQSPKLLIYWTSTRHTGVPDRFTGSGSGTDYTLTISSVQAEDLALYYCQQHYSTPWTFGGGTKLEIK10G5 VH23QVQLQQSAAELARPGASVKSCKASGYTFTSYTHWVKQRPGQGLEWIGYINPSSGYTENNRKFKDKTTLTADKSSSTAYQLSSLTSEDSAVYYCARLGKGLLPPFDYWGQGTTLTVSSAKTTPPSVYPLAPGSAAQT10G5 VL24DIQMTQSPASLSVSVGETVTITCRASENIYSNLAWYQQKQGKSPQLLVYAATNLADGVPSRFSGSGSGTQYSLKINSLQSEDFGSYYCQHFWGTPYTFGGGTKLEIK13H1 VH34EVQLQQSGPELVKPGASMKISCKASHYSFIGYTMNWVKQRHGKNLEWIGLINPYNGDTTYNQKFKGKASLTVDKSSSTAYMEILSLTSEDSAVYYCARENWGYPYAMDYWGQGTSVTVS13H1 VL35DIVLTQSPASLAVSLGQRATISCRASESVDNFGISFMNWFQQKPGQPPKLLIYAASNQGSGVPARFSGSRSGTDFSLNIHPMEEDDTAMYFCQQSKEVPYTFGGGTKLEIK1E2 VH45QVQLQQSGAELVRPGVSVKISCKGSGYTFTDYAMNWVKQSHAKSLEWIGVISTYYGDANYNQKFKGKATMTVDKSSSTAYMELARLTSEDSAIYYCALIYYDYDGSYWGQGTTLTVS1E2 VL46DVVMTQTPLSLPVSLGDQASISCRSSQSLVHSNGNTYLHWYLQKPGQSPKLLIYKVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYFCSQSTHVPPYTFGGGTKLEIK9E10 VH56QVQLQQSAAELARPGASVKMSCKASGYTFTSYTMHWVKQRPGQGLEWIGYINPSSGYTDYNQKFKDKTTLTADRSSSTAYMQLSSLTSEDSAVYYCARLGKGLLPPFDYWGQGSTLTVSS9E10 VL157EIVLTQSIPSLTVSAGERVTISCKSNQNLLWSGNQRYCLVWHQWKPGQTPTPLITWTSDRYSGVPDRFIGSGSVTDFTLTISSVQAEDVAVYFCQQHLHIPYTFGGGTKLEIK9E10 VL267DIQMTQSPASLSVSVGETVTITCRASENIYSNLAWYQQKQGKSPQLLVYAATNLADGVPSRFSGSGSGTQYSLKINSLQSEDFGSYYCQHFWGTPYTFGGGTKLEIK1C3 VH170QVQLQQSGAELARPGASVKLSCKASGYTFTSYWMQWVKQRPGQGLEWIGAIYPGDGDTRYTQKFKGKATLTADKSSSTAYMQLSSLASEDSAVYYCARRYDGYYHFDYWGQGTTLTVS1C3 VL171DIVMTQSPSSLAVTAGEKVTMSCKSSQSLLWSVNQKNYLSWYQQKQRQPPKLLIYGASIRESWVPDRFTGSGSGTDFTLTISNVHAEDLAVYYCQHNHGSFLPLTFGSGTKLEIK20E9 VH181QVQLQQSGAEVARPGASVKLSCKSSGFTFTTYWMQWVKQRPGQGLEWIGAIYPGDGDTRYTQKFKGKATLTADKSSITAYMQLSSLASEDSAVYYCARRGDYGNYGMDYWGQGTSVTVSS29E9 VL182DVLMTQTPLSLPVSLGDQASISCRSSQSIVHSNGNTYLEWYLQKPGQSPKLLIYKVSNHFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYYCFQGSHVPPTFGGGTKLEIK Fragments and Derivatives Fragments and derivatives of antibodies (which are encompassed by the term “antibody” or “antibodies” as used in this application, unless otherwise stated or clearly contradicted by context), preferably a 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9-like antibody, can be produced by techniques that are known in the art. “Fragments” comprise a portion of the intact antibody, generally the antigen binding site or variable region. Examples of antibody fragments include Fab, Fab′, Fab'-SH, F (ab′) 2, and Fv fragments; diabodies; any antibody fragment that is a polypeptide having a primary structure consisting of one uninterrupted sequence of contiguous amino acid residues (referred to herein as a “single-chain antibody fragment” or “single chain polypeptide”), including without limitation (1) single-chain Fv molecules (2) single chain polypeptides containing only one light chain variable domain, or a fragment thereof that contains the three CDRs of the light chain variable domain, without an associated heavy chain moiety and (3) single chain polypeptides containing only one heavy chain variable region, or a fragment thereof containing the three CDRs of the heavy chain variable region, without an associated light chain moiety; and multispecific antibodies formed from antibody fragments. Included, inter alfa, are a nanobody, domain antibody, single domain antibody or a “dAb”. Fragments of the present antibodies can be obtained using standard methods. For instance, Fab or F (ab′) 2 fragments may be produced by protease digestion of the isolated antibodies, according to conventional techniques. It will be appreciated that immunoreactive fragments can be modified using known methods, for example to slow clearance in vivo and obtain a more desirable pharmacokinetic profile the fragment may be modified with polyethylene glycol (PEG). Methods for coupling and site-specifically conjugating PEG to a Fab' fragment are described in, for example, Leong et al, 16 (3): 106-119 (2001) and Delgado et al, Br. J. Cancer 73 (2): 175-182 (1996), the disclosures of which are incorporated herein by reference. Alternatively, the DNA of a hybridoma producing an antibody, preferably a 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9-like antibody, may be modified so as to encode a fragment. The modified DNA is then inserted into an expression vector and used to transform or transfect an appropriate cell, which then expresses the desired fragment. In certain embodiments, the DNA of a hybridoma producing an antibody, preferably a 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9-like antibody, can be modified prior to insertion into an expression vector, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous non-human sequences (e.g., Morrison et al., PNAS pp. 6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. In that manner, “chimeric” or “hybrid” antibodies are prepared that have the binding specificity of the original antibody. Typically, such non-immunoglobulin polypeptides are substituted for the constant domains of an antibody. Thus, according to another embodiment, the antibody, preferably a 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9-like antibody, is humanized. “Humanized” forms of antibodies are specific chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F (ab′) 2, or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from the murine immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary-determining region (CDR) of the recipient are replaced by residues from a CDR of the original antibody (donor antibody) while maintaining the desired specificity, affinity, and capacity of the original antibody. In some instances, Fv framework residues of the human immunoglobulin may be replaced by corresponding non-human residues. Furthermore, humanized antibodies can comprise residues that are not found in either the recipient antibody or in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of the original antibody and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details see Jones et al., Nature, 321, pp. 522 (1986); Reichmann et al, Nature, 332, pp. 323 (1988); Presta, Curr. Op. Struct. Biol., 2, pp. 593 (1992); Verhoeyen et Science, 239, pp. 1534; and U.S. Pat. No. 4,816,567, the entire disclosures of which are herein incorporated by reference.) The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity. According to the so-called “best-fit” method, the sequence of the variable domain of an antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of the mouse is then accepted as the human framework (FR) for the humanized antibody (Sims et al., J. Immunol. 151, pp. 2296 (1993); Chothia and Lesk, J. Mol. 196, 1987, pp. 901). Another method uses a particular framework from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework can be used for several different humanized antibodies (Carter et. al.., PNAS 89, pp. 4285 (1992); Presta et al., J. Immunol., 151, p. 2623 (1993)). It is further important that antibodies be humanized with retention of high affinity for KIR3DL2 receptors and other favorable biological properties. To achieve this goal, according to an exemplary method, humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the consensus and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen (s), is achieved. In general, the CDR residues are directly and most substantially involved in influencing antigen binding. Another method of making “humanized” monoclonal antibodies is to use a murine host according that has had its immunoglobulin genes replaced by functional human immunoglobulin genes (see, e.g., U.S. Pat. No. 6,162,963, which is herein incorporated in its entirety by reference). Human antibodies may also be produced according to various other techniques, such as by using, for immunization, other transgenic animals that have been engineered to express a human antibody repertoire (Jakobovitz et al., Nature 362 (1993) 255), or by selection of antibody repertoires using phage display methods. Such techniques are known to the skilled person and can be implemented starting from monoclonal antibodies as disclosed herein. The antibodies, optionally a 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 5H1, 1E2, 1C3 or 20E9-like antibody, may also be derivatized to “chimeric” antibodies (immunoglobulins) in which a portion of the heavy/light chain(s) is identical with or homologous to corresponding sequences in the original antibody, while the remainder of the chain (s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity and binding specificity (Cabilly et al., supra; Morrison et al., Proc. Natl. Acad. Sci. U.S.A., pp. 6851 (1984)). Various forms of the humanized antibody or affinity-matured antibody are contemplated. For example, the humanized antibody or affinity-matured antibody may be an antibody fragment, such as a Fab. Alternatively, the humanized antibody or affinity-matured antibody may be a full-length or intact antibody, such as a full-length or intact IgG1 or IgG4 antibody. In one embodiment, the humanized antibody is a full-length IgG4 antibody or a fragment thereof. To produce such antibodies, humanized VH and VL regions, or variant versions thereof, can be cloned into expression vectors encoding full-length or truncated constant regions from a human antibody according to standard recombinant methods (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, 2ndEd., Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989). The result is a transfected cell line that expresses and secretes the humanized antibody molecule of interest, comprising the selected VH and VL regions and constant regions. cDNA sequences encoding the constant regions of human antibodies are known. The constant region may further be modified according to known methods. For example, in an IgG4 constant region, residue S241 may be mutated to a proline (P) residue to allow complete disulphide bridge formation at the hinge (see, e.g., Angal et al., Mol Immunol. 1993; 30:105-8). Modified Constant Regions In view of the ability of the anti-KIR3DL2 antibodies (particularly the non-internalizing antibodies) to induce ADCC and CDC, the antibodies can also be made with modifications that increase their ability to bind Fc receptors which can affect effector functions such as antibody-dependent cytotoxicity, mast cell degranulation, and phagocytosis, as well as immunomodulatory signals such as regulation of lymphocyte proliferation and antibody secretion. Typical modifications include modified human IgG1 constant regions comprising at least one amino acid modification (e.g. substitution, deletions, insertions), and/or altered types of glycosylation, e.g., hypofucosylation. Such modifications can affect interaction with Fc receptors: FcγRI (CD64), FcγRII (CD32), and FcγRIII (CD 16). FcγRI (CD64), FcγRIIA (CD32A) and FcγRIII (CD 16) are activating (i.e., immune system enhancing) receptors while FcγRIIB (CD32B) is an inhibiting (i.e., immune system dampening) receptor. A modification may, for example, increase binding of the Fc domain to FcγRIIIa on effector (e.g. NK) cells. Anti-KIR3DL2 antibodies preferably comprise an Fc domain (or portion thereof) of human IgG1 or IgG3 isotype, optionally modified. Residues 230-341 (Kabat EU) are the Fc CH2 region. Residues 342-447 (Kabat EU) are the Fc CH3 region. Anti-KIR3DL2 antibodies may comprise a variant Fc region having one or more amino acid modifications (e.g., substitutions, deletions, insertions) in one or more portions, which modifications increase the affinity and avidity of the variant Fc region for an FcγR (including activating and inhibitory FcγRs). In some embodiments, said one or more amino acid modifications increase the affinity of the variant Fc region for FcγRIIIA and/or FcγRIIA. In another embodiment, the variant Fc region further specifically binds FcγRIIB with a lower affinity than does the Fc region of the comparable parent antibody (e.g., an antibody having the same amino acid sequence as the antibody except for the one or more amino acid modifications in the Fc region). For example, the one or both of the histidine residues at amino acid positions 310 and 435 may be substituted, for example by lysine, alanine, glycine, valine, leucine, isoleucine, proline, methionine, tryptophan, phenylalanine, serine or threonine (see, e.g. PCT publication no. WO 2007/080277); such substituted constant regions provide decreased binding to the inhibitory FcγRIIB without decreasing binding to the activatory FcγRIIIA. In some embodiments, such modifications increase the affinity of the variant Fc region for FcγRIIIA and/or FcγRIIA and also enhance the affinity of the variant Fc region for FcγyRIIB relative to the parent antibody. In other embodiments, said one or more amino acid modifications increase the affinity of the variant Fc region for FcγRIIIA and/or FcγRIIA but do not alter the affinity of the variant Fc regions for FcγRIIB relative to the Fc region of the parent antibody. In another embodiment, said one or more amino acid modifications enhance the affinity of the variant Fc region for FcγRIIIA and FcγRIIA but reduce the affinity for FcγRIIB relative to the parent antibody. Increased affinity and/or avidity results in detectable binding to the FcγR or FcγR-related activity in cells that express low levels of the FcγR when binding activity of the parent molecule (without the modified Fc region) cannot be detected in the cells. The affinities and binding properties of the antibodies for an FcγR can be determined using in vitro assays (biochemical or immunological based assays) known in the art for determining antibody-antigen or Fc-FcγR interactions, i.e., specific binding of an antigen to an antibody or specific binding of an Fc region to an FcγR, respectively, including but not limited to ELISA assay, surface plasmon resonance assay, immunoprecipitation assays. In some embodiments, the antibodies comprising a variant Fc region comprise at least one amino acid modification (for example, possessing 1, 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) in the CH3 domain of the Fc region. In other embodiments, the antibodies comprising a variant Fc region comprise at least one amino acid modification (for example, possessing 1, 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) in the CH2 domain of the Fc region, which is defined as extending from amino acids 231-341. In some embodiments, antibodies comprise at least two amino acid modifications (for example, possessing 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications), wherein at least one such modification is in the CH3 region and at least one such modification is in the CH2 region. Encompasses also are amino acid modification in the hinge region. In one embodiment, encompassed are amino acid modification in the CH1 domain of the Fc region, which is defined as extending from amino acids 216-230. Any combination of Fc modifications can be made, for example any combination of different modifications disclosed in United States Patents Nos. U.S. Pat. Nos. 7,632,497; 7,521,542; 7,425,619; 7,416,727; 7,371,826; 7,355,008; 7,335,742; 7,332,581; 7,183,387; 7,122,637; 6,821,505 and 6,737,056; in PCT Publications Nos. WO2011/109400; WO 2008/105886; WO 2008/002933; WO 2007/021841; WO 2007/106707; WO 06/088494; WO 05/115452; WO 05/110474; WO 04/1032269; WO 00/42072; WO 06/088494; WO 07/024249; WO 05/047327; WO 04/099249 and WO 04/063351; and in Presta, L. G. et al. (2002) Biochem. Soc. Trans. 30(4):487-490; Shields, R. L. et al. (2002) J. Biol. Chem. 26; 277(30):26733-26740 and Shields, R. L. et al. (2001) J. Biol. Chem. 276(9):6591-6604). Anti-KIR3DL2 antibodies may comprise a variant Fc region, wherein the variant Fc region comprises at least one amino acid modification (for example, possessing 1, 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) relative to a wild-type Fc region, such that the molecule has an enhanced effector function relative to a molecule comprising a wild-type Fc region, optionally wherein the variant Fc region comprises a substitution at any one or more of positions 221, 239, 243, 247, 255, 256, 258, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 300, 301, 303, 305, 307, 308, 309, 310, 311, 312, 316, 320, 322, 326, 329, 330, 332, 331, 332, 333, 334, 335, 337, 338, 339, 340, 359, 360, 370, 373, 376, 378, 392, 396, 399, 402, 404, 416, 419, 421, 430, 434, 435, 437, 438 and/or 439. In one embodiment, anti-KIR3DL2 antibodies may comprise a variant Fc region, wherein the variant Fc region comprises at least one amino acid modification (for example, possessing 1, 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) relative to a wild-type Fc region, such that the molecule has an enhanced effector function relative to a molecule comprising a wild-type Fc region, optionally wherein the variant Fc region comprises a substitution at any one or more of positions 329, 298, 330, 332, 333 and/or 334 (e.g. S239D, S298A, A330L, 1332E, E333A and/or K334A substitutions). In one embodiment, antibodies having variant or wild-type Fc regions may have altered glycosylation patterns that increase Fc receptor binding ability of antibodies. Such carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies to thereby produce an antibody with altered glycosylation. See, for example, Shields, R. L. et al. (2002) J. Biol. Chem. 277:26733-26740; Umana et al. (1999) Nat. Biotech. 17:176-1, as well as, European Patent No: EP 1,176,195; PCT Publications WO 06/133148; WO 03/035835; WO 99/54342, each of which is incorporated herein by reference in its entirety. Generally, such antibodies with altered glycosylation are “glyco-optimized” such that the antibody has a particular N-glycan structure that produces certain desirable properties, including but not limited to, enhanced ADCC and effector cell receptor binding activity when compared to non-modified antibodies or antibodies having a naturally occurring constant region and produced by murine myeloma NSO and Chinese Hamster Ovary (CHO) cells (Chu and Robinson, Current Opinion Biotechnol. 2001, 12: 180-7), HEK293T-expressed antibodies as produced herein in the Examples section, or other mammalian host cell lines commonly used to produce recombinant therapeutic antibodies. Monoclonal antibodies produced in mammalian host cells contain an N-linked glycosylation site at Asn297 of each heavy chain. Glycans on antibodies are typically complex biatennary structures with very low or no bisecting N-acetylglucosamine (bisecting GlcNAc) and high levels of core fucosylation. Glycan temini contain very low or no terminal sialic acid and variable amounts of galactose. For a review of effects of glycosylation on antibody function, see, e.g., Wright & Morrison, Trend Biotechnol. 15:26-31(1997). Considerable work shows that changes to the sugar composition of the antibody glycan structure can alter Fc effector functions. The important carbohydrate structures contributing to antibody activity are believed to be the fucose residues attached via alpha-1,6 linkage to the innermost N-acetylglucosamine (GlacNAc) residues of the Fc region N-linked oligosaccharides (Shields et al., 2002). Antibodies having lowered fucose content on N-linked glycans (hypofucosylated N-linked glycans) can therefore be produced. FcγR binding requires the presence of oligosaccharides covalently attached at the conserved Asn297 in the Fc region of human IgG1, IgG2 or IgG3 type. Non-fucosylated oligosaccharides structures have recently been associated with dramatically increased in vitro ADCC activity. “Asn 297” refers to the amino acid asparagine located at about position 297 in the Fc region; based on minor sequence variations of antibodies, Asn297 can also be located some amino acids (usually not more than +3 amino acids) upstream or downstream. Historically, antibodies produced in CHO cells contain about 2 to 6% in the population that are nonfucosylated. YB2/0 (rat myeloma) and Lec13 cell line (a lectin mutant of CHO line which has a deficient GDP-mannose 4,6-dehydratase leading to the deficiency of GDP-fucose or GDP sugar intermediates that are the substrate of alpha6-fucosyltransferase have been reported to produce antibodies with 78 to 98% non-fucosylated species. In other examples, RNA interference (RNAi) or knock-out techniques can be employed to engineer cells to either decrease the FUT8 mRNA transcript levels or knock out gene expression entirely, and such antibodies have been reported to contain up to 70% non-fucosylated glycan. An antibody binding to KIR3DL2 may be glycosylated with a sugar chain at Asn297, said antibody showing increased binding affinity via its Fc portion to FcγRIII. In one embodiment of the invention, an antibody will comprise a constant region comprising at least one amino acid alteration in the Fc region that improves antibody binding to FcγRIIIa and/or ADCC. In one aspect, the antibodies are hypofucosylated in their constant region. Such antibodies may comprise an amino acid alteration or may not comprise an amino acid alteration but be produced or treated under conditions so as to yield such hypofucosylation. In one aspect, an antibody composition comprises a chimeric, human or humanized antibody described herein, wherein at least 20, 30, 40, 50, 60, 75, 85, 90, 95% or substantially all of the antibody species in the composition have a constant region comprising a core carbohydrate structure (e.g. complex, hybrid and high mannose structures) which lacks fucose. In one embodiment, provided is an antibody composition which is free of antibodies comprising a core carbohydrate structure having fucose. The core carbohydrate will preferably be a sugar chain at Asn297. In one embodiment, provided is an antibody composition, e.g. a composition comprising antibodies which bind to KIR3DL2, are glycosylated with a sugar chain at Asn297, wherein the antibodies are partially fucosylated. Partially fucosylated antibodies are characterized in that the proportion of anti-KIR3DL2 antibodies in the composition that lack fucose within the sugar chain at Asn297 is between 20% and 90%, preferably between 20% and 80%, preferably between 20% and 50%, 55%, 60%, 70% or 75%, between 35% and 50%, 55%, 60%, 70% or 75%, or between 45% and 50%, 55%, 60%, 70% or 75%. Preferably the antibody is of human IgG1 or IgG3 type. The sugar chain show can further show any characteristics (e.g. presence and proportion of complex, hybrid and high mannose structures), including the characteristics of N-linked glycans attached to Asn297 of an antibody from a human cell, or of an antibody recombinantly expressed in a rodent cell, murine cell (e.g. CHO cell) or in an avian cell. In one embodiment, the antibody is expressed in a cell that is lacking in a fucosyltransferase enzyme such that the cell line produces proteins lacking fucose in their core carbohydrates. For example, the cell lines Ms704, Ms705, and Ms709 lack the fucosyltransferase gene, FUT8 (alpha (1,6) fucosyltransferase), such that antibodies expressed in the Ms704, Ms705, and Ms709 cell lines lack fucose on their core carbohydrates. These cell lines were created by the targeted disruption of the FUT8 gene in CHO/DG44 cells using two replacement vectors (see U.S. Patent Publication No. 20040110704 by Yamane et al.; and Yamane-Ohnuki et al. (2004) Biotechnol Bioeng 87:614-22, the disclosures of which are incorporated herein by reference). Other examples have included use of antisense suppression, double-stranded RNA (dsRNA) interference, hairpin RNA (hpRNA) interference or intron-containing hairpin RNA (ihpRNA) interference to functionally disrupt the FUT8 gene. In one embodiment, the antibody is expressed in a cell line with a functionally disrupted FUT8 gene, which encodes a fucosyl transferase, such that antibodies expressed in such a cell line exhibit hypofucosylation by reducing or eliminating the alpha 1,6 bond-related enzyme. In one embodiment, the antibody is expressed in cell lines engineered to express glycoprotem-modifying glycosyl transferases (e.g., beta(1,4)-N-acetylglucosaminyl-transferase III (GnTHI)) such that antibodies expressed in the engineered cell lines exhibit increased bisecting GlcNac structures which results in increased ADCC activity of the antibodies (PCT Publication WO 99/54342 by Umana et al.; and Umana et al. (1999) Nat. Biotech. 17:176-180, the disclosures of which are incorporated herein by reference). In another embodiment, the antibody is expressed and the fucosyl residue(s) is cleaved using a fucosidase enzyme. For example, the fucosidase alpha-L-fucosidase removes fucosyl residues from antibodies (Tarentino, et al. (1975) Biochem. 14:5516-5523). In other examples, a cell line producing an antibody can be treated with a glycosylation inhibitor; Zhou et al. Biotech. and Bioengin. 99: 652-665 (2008) described treatment of CHO cells with the alpha-mannosidase I inhibitor, kifunensine, resulting in the production of antibodies with non-fucosylated oligomannose-type N-glucans. In one embodiment, the antibody is expressed in a cell line which naturally has a low enzyme activity for adding fucosyl to the N-acetylglucosamine that binds to the Fc region of the antibody or does not have the enzyme activity, for example the rat myeloma cell line YB2/0 (ATCC CRL 1662). Other example of cell lines include a variant CHO cell line, Led 3 cells, with reduced ability to attach fucosyl to Asn(297)-linked carbohydrates, also resulting in hypofucosylation of antibodies expressed in that host cell (WO 03/035835 (Presta et al); and Shields, RX. et al. (2002) J. Biol. Chem. 277:26733-26740, the disclosures of which are incorporated herein by reference). In another embodiment, the antibody is expressed in an avian cell, preferably a EBx® cell (Vivalis, France) which naturally yields antibodies with low fucose content e.g. WO2008/142124. Hypofucosylated glycans can also be produced in cell lines of plant origin, e.g. WO 07/084926A2 (Biolex Inc.), WO 08/006554 (Greenovation Biotech GMBH), the disclosures of which are incorporated herein by reference. Uses in Diagnostics and Therapy In certain embodiments, the present antibodies are used to purify or identify KIR3DL2 positive cells from a biological sample. Biological samples can be obtained from a patient, e.g. for diagnostic or ex vivo therapeutic purposes, or from individuals or non-human primates to obtain a source of such cells for research purposes. KIR3DL2 positive cells can be purified or identified using the present antibodies with any of a number of standard methods. For example, peripheral blood cells can be sorted using a FACS scanner using labeled antibodies specific for KIR3DL2, and optionally to other cell surface molecules typically present on cells, e.g., CD4, CD8 or CD30 for T cell; CD4 CD2+, CD3+, CD5+, CD8−, CD28+, CD45RO+ and/or TCRαβ+ for malignant cells in Sézary Syndrome; CD4+ (optionally CD4+ and CD28−) in inflammatory, autoimmune or cardiovascular diseases. In addition, the antibodies can be conjugated or covalently linked to a solid support and used to purify or identify KIR3DL2 positive cells or any cells expressing KIR3DL2 from a biological sample, e.g., from a blood sample or mucosal tissue biopsy from a patient or other individual. Specifically, the biological sample is placed into contact with the antibodies under conditions that allow cells within the sample to bind to the antibody, and then the cells are eluted from the solid-support-bound antibody. Regardless of the method used to isolate, purify or identify the KIR3DL2 positive cells, the ability to do so is useful for numerous purposes, e.g. to diagnose a disorder characterized by a pathogenic expansion of KIR3DL2-expressing cells, by assessing the number or activity or other characteristics of KIR3DL2 positive cells obtained from a patient, or to evaluate the ability of the antibodies, or fragments or derivatives thereof, to modulate the activity or behavior of the cells of a patient prior, e.g., to one of the herein-described treatments using the antibodies. Further, purified KIR3DL2 positive cells are useful in a research context, e.g., to better characterize the cells and their various properties and behaviors, as well as to identify compounds or methods that can be used to modulate their behavior, activity, or proliferation. The antibodies can also be useful in diagnostic methods, for example in methods of detecting KIR polypeptides on cells, e.g. disease cells from a patient. The present disclosure also provides pharmaceutical compositions that comprise an antibody which specifically binds to KIR3DL2 polypeptides on the surface of cells. The antibody preferably inhibits the growth or activity (e.g. cytokine production) of the cells and/or leads to the elimination of the KIR3DL2 positive cells, preferably via induction of CDC and/or ADCC. The composition further comprises a pharmaceutically acceptable carrier. The disclosure further provides a method of inhibiting the growth or activity of, and/or depleting, KIR3DL2-positive cells, in a patient in need thereof, comprising the step of administering to said patient a composition described herein. Such treatment methods can be used for a number of disorders, including, but not limited to CTCL, SS and MF, inflammatory, autoimmune and cardiovascular disorders. Regardless of the form of CD4+ CTCL, there are malignant CD4+ T cells which express KIR3DL2 at their surface. KIR3DL2 thus covers the range of CD4+ CTCL, and notably the Sézary Syndrome (“SS”), transformed Mycosis Fungoides (“transformed MF”), Lymphomatoide Papulosis (“LP”), and CD30+ lymphomas. A diagnosis (e.g. a CTCL diagnosis) may be based on the analysis of the presence of KIR3DL2 at the surface of CD4+ cells collected from the suspected body area (e.g. sample of skin erythroderma when transformed MF is suspected, or sample of peripheral blood when a more aggressive CTCL form, such as SS, is suspected). It can typically be concluded that a CD4+ T cell is tumoral as soon as there are KIR3DL2 polypeptides detected at the surface of these CD4+ T cells. The percentage of CD4+ KIR3DL2+ T cells can measured in a sample of peripheral blood collected from a patient for whom a SS is suspected, and such percentage will substantially correspond to the percentage of malignant SS cells that are actually present in the peripheral blood of this patient (generally within a ±10% range or even a ±5% range for KIR3DL2+, CD4+ cells. KIR3DL2 and the anti-KIR3DL2 antibodies described herein therefore can be used in the staging of disease, particularly SS. Insofar as KIR3DL2 is a universal marker for CTCL, the antibodies can be used in combination with other treatments or diagnostic markers for CTCL. For example, CD30 of which presence at the surface of malignant CD4+ T cells indicates that the patient has a particular form of CD4+ CTCL which is referred to in the art as CD30+ lymphoma. CD30 is therefore a CTCL marker for a particular form of CTCL (CD30+ lymphomas), however CD30 does not cover every form of CD4+ CTCL since for CD4+ CTCL such as SS, transformed MF, or LP, there does not necessarily exist a malignant CD4+ T cell which would express CD30 at its surface. CD30 can therefore be used in addition to KIR3DL2 as a marker in CTCL diagnosis and therapy. Furthermore, a finding that a patient has CD4+ CTCL which expresses CD30 can indicate that the patient is suitable for treatment with an anti-KIR3DL2 antibody and an anti-CD30 antibody; optionally the patient can then be treated anti-KIR3DL2 antibody and an anti-CD30 antibody. In some embodiments, prior to the administration of the anti-KIR3DL2 antibody or composition, the presence of CD2, CD3, CD4, CD5, CD8, CD28, CD30, CD45RO and/or TCRαβ will be assessed on cells (e.g. pathogenic cells) from a patient. A patient whose cells express (or do not express, in accordance with the particular disorder and cells sought to be targeted) a marker can then be treated with an anti-KIR3DL2 antibody or composition. In some embodiments, prior to the administration of the anti-KIR3DL2 antibody or composition, the presence of KIR3DL2 on cells of the patient will be assessed, e.g., to determine the relative level and activity of KIR3DL2-positive cells in the patient as well as to confirm the binding efficacy of the antibodies to the cells of the patient. A patient whose cells express KIR3DL2 can then be treated with an anti-KIR3DL2 antibody or composition. This can be accomplished by obtaining a sample of PBLs or cells from the site of the disorder, and testing e.g., using immunoassays, to determine the relative prominence of markers such as CD4, CD8, CD30 or KIR3DL2 on the cells. In one embodiment, where it is sought to inhibit the activity or growth of, or deplete, a patient's KIR3DL2-positive cells, the ability of the anti-KIR3DL2 antibody to inhibit proliferation of or deplete a patient's KIR3DL2-positive cells is assessed. If the KIR3DL2-positive cells are depleted by the anti-KIR3DL2 antibody or composition, the patient is determined to be responsive to therapy with an anti-KIR3DL2 antibody or composition, and optionally the patient is treated with an anti-KIR3DL2 antibody or composition. In some embodiments, the method may comprise the additional step of administering to said patient an appropriate additional (second) therapeutic agent selected from an immunomodulatory agent, an immunosuppressive agent, a hormonal agent, a chemotherapeutic agent, a second antibody (e.g. a depleting antibody) that binds to a polypeptide present on a KIR3DL2-expressing cell. Such additional agents can be administered to said patient as a single dosage form together with said antibody, or as a separate dosage form. The dosage of the antibody (or antibody and the dosage of the additional therapeutic agent collectively) are sufficient to detectably induce, promote, and/or enhance a therapeutic response in the patient. Where administered separately, the antibody, fragment, or derivative and the additional therapeutic agent are desirably administered under conditions (e.g., with respect to timing, number of doses, etc.) that result in a detectable combined therapeutic benefit to the patient. Mycosis fungoides and the more aggressive Sézary syndrome represent the most common forms of CTCL. The clinical course of MF/SS is usually indolent, with pruritic erythematous areas slowly developing over long periods. Eventually, however, the erythematous patches become progressively infiltrated, developing into plaques and finally to ulcerating tumors. The prognosis of MF/SS is based on the extent of disease at presentation. Patients with stage I disease have a median survival of 20 years or more, in comparison with a median survival of approximately 3 to 4 years for patients with stage III/IV disease. The compositions described herein can be used for treatment in combination with any agent known to be useful in the treatment of the particular T cell malignancy. Various treatments for CTCL are in use, including corticosteroids, nitrogen mustard, carmustine, topical tacrolimus (Protopic®), imiquimod (Aldara®; 3M Inc.), topical retinoids, and rexinoids (bexarotene; Targretin®; Ligand Pharmaceuticals, San Diego, CA)), as well as ultraviolet light therapy (Psoralen+UVA (PUVA), narrowband UVB, and UVB), Photodynamic therapy (PDT) and body irradiation. Treatments also include histone deacetylase inhibitors such as vorinostat (suberoylanilide hydroxamic acid, Zolinza®) and Romidepsin (depsipeptide, FK-228, Istodax®), a cyclic peptide that selectively inhibits histone deacetylase isotypes 1, 2, 4 and 6. Chemotherapy or combination chemotherapy are also used. Examples include gemcitabine, antifolate analogues such as Pralatrexate (Folotyn®). Further therapies include IMiDs (immunomodulatory drugs), analogs derived from thalidomide that have a wide range of effects, including both immune and non-immune related effects. Representatives of the IMiD class include CC-5013 (lenalidomide; Revlimid®), CC-4047 (Actimid), and ENMD-0995. Further treatments include proteosome inhibitors such as bortezomib (Velcade®), a reversible 26S proteasome inhibitor. Stem cell transplantation is also used. Although there is no current standard of care for MF/SS, there is a general tendency to rely on topical interventions for early disease delaying systemic and more toxic therapy until the development of extensive symptoms. Psoralen and ultraviolet A radiation (PUVA), combined or not with low doses of interferon-α, is effective in early-stage MF/SS, inducing complete remission (CR) in most patients. Local radiotherapy or total-skin electron-beam irradiation (TSEB) has been used with success to control advanced skin disease. Extra corporeal photopheresis may also be used successfully but is not generally available. Once the disease becomes refractory to topical therapy, interferon-α, the rexinoid bexarotene (Targretin®, Ligand Pharmaceuticals, San Diego, CA), a synthetic retinoid analog targeting the retinoid X receptor, single-agent chemotherapy or combination chemotherapy may be given. Treatments, particularly skin-directed therapies, include, e.g., corticosteroids, nitrogen mustard, carmustine, topical tacrolimus (Protopic®) and imiquimod (Aldara®; 3M Inc.). The duration of response is however often less than 1 year, and ultimately all patients have relapses and the disease becomes refractory. The recombinant IL2-diphteria toxin denileukin diftitox (DAB389IL-2, ONTAK®, Ligand Pharmaceuticals, San Diego, CA) is active in patients with stage Ib to stage IV CTCL refractory to previous treatments (overall objective response in 30% of 71 patients with a median duration response of 7 months) and appears to have a beneficial effect in symptoms relief and quality of life. More recently, denileukin diftitox have been tested in a Phase I trial in combination with bexarotene, since it induces CD25 up regulation in vitro. The combination was well tolerated and induced objective response in 67% of 14 patients. The most significant adverse events were those already reported with bexarotene alone (hypertriglyceridemia and suppression of thyroid function due to decreased TSH production) and grade 3 or 4 lymphopenia but resolving within one month of cessation of therapy. The time to treatment failure was not reported in this study. In other studies, anti-CD4 antibodies that deplete CD4 expressing cells have been developed. Examples include the fully human IgG1 anti-CD4 antibody zanolimumab (HuMax-CD4; Genmab A/S and TenX BioPharma Inc.), and the chimeric monoclonal anti-CD4 (cM-T412, Centocor, Malvern, PA) was administered to 8 patients with MF and induced objective response in 7 of them but with a median response duration of only 5 months. Uvadex® (methoxsalen, Therakos Inc. Exton, PA) in extra corporal photopheresis, has also shown signs of efficacy. The humanized monoclonal antibody alemtuzumab (hu-IgG1anti-CD52 mAb, Campath®, Millennium Pharmaceuticals, Inc. and ILEX Oncology, Inc., marketed and distributed in the US by Berlex Laboratories, Inc., Montville, NJ) is indicated for the treatment of B-cell chronic lymphocytic leukemia (B-CLL) in patients who have been treated with alkylating agents and who have failed fludarabine therapy. It has been tested in patients with advanced MF/SS (stage III or IV disease) and led to objective responses in at least half of cases (55% of 22 patients). Its side effect profile consists mainly of immunosuppression and infusion reactions. An independent retrospective study described also significant cardiac toxicity in 4 out of 8 patients. With long lasting remissions observed (median time to treatment failure 12 months, range 5 to 32+ months), alemtuzumab therapy appears to be the treatment with the more favorable median response duration compared to all treatments reported to date. Other agents that may be useful include anti-CCR4 (C-C chemokine receptor 4; CD194) antibodies. One example is mogamulizumab (KW-0761; AMG-761; trade name Poteligeo, Kyowa Hakko Kirin Ltd., Japan and Amgen, USA), and humanized anti-CCR4 antibody. Other agents that may be useful include anti-CD30 antibodies. One example is SGN-35 is an antibody-drug conjugate (ADC) containing the potent antimitotic drug, monomethylauristatin E (MMAE), linked to the anti-CD30 monoclonal antibody, cAC10 (Okeley et al. (2010) Clin. Cancer Res. 16(3): 888-897); another examples is the human anti-CD30 immunoglobulin (Ig) Glκ monoclonal antibody MDX-060 (Medarex Inc. and Bristol Myers Squibb; Ansell et al. (2007) J. Clin. Oncol. 25: 2767-2769). Each of these treatments can be used in combination with the antibodies of the disclosure. The antibodies produced using the present methods are particularly effective at treating autoimmune and inflammatory disorders, as well as cardiovascular disorders most particularly acute coronary syndrome, arthritis, rheumatoid arthritis, rheumatoid vasculitis, systemic lupus erythematosus, multiple sclerosis, Wegener's granulomatosis, and spondyloarthritis. In general, the present methods can be used to treat any disorder caused at least in part by the presence or activity of KIR3DL-expressing cells, e.g., NK cells or T cells, proinflammatory T or NK cells producing IL-17A, T cells such as Th17 cells or CD4+CD28−cells expressing KIR3DL2, and which can therefore be effectively treated by selectively killing or inhibiting the proliferation or activation of KIR3DL2-expressing cells. In some embodiments, prior to the administration of the anti-KIR3DL2 antibody, the expression of KIR3DL2 on cells underlying the particular disorder will be assessed. This can be accomplished by obtaining a sample of PBLs or cells from the site of the disorder (e.g., from the synovium in RA patients), and testing e.g., using immunoassays, to determine the relative prominence of markers such as CD4, CD28, etc., as well as KIR3DL2 on the cells. Other methods can also be used to detect expression of KIR3DL2 and other genes, such as RNA-based methods, e.g., RT-PCR or Northern blotting. The treatment may involve multiple rounds of antibody or compound administration. For example, following an initial round of administration, the level and/or activity of KIR3DL-expressing T or NK cells (e.g., CD4+CD28−T cells, malignant CD4+ T cells), in the patient will generally be re-measured, and, if still elevated, an additional round of administration can be performed. In this way, multiple rounds of receptor detection and antibody or compound administration can be performed, e.g., until the disorder is brought under control. When used for the treatment of autoimmune or inflammatory disorders, the anti-KIR3DL2 antibodies of the disclosure can be used for treatment in combination with any agent known to be useful in the treatment of the particular inflammatory disorder, autoimmune disorder, or cardiovascular disorder. Anti-KIR3DL2 antibodies can be combined for example with steroidal anti-inflammatory agents, non-steroidal anti-inflammatory agents, anti-metabolites and other agents used in treating cardiovascular, inflammatory or autoimmune diseases. In some embodiments, anti-inflammatory agents comprise steroidal anti-inflammatory agents, which include glucocorticosteroids and mineralocorticosteroids. These may be administered by any methods suitable for treating the inflammatory disorders, including, among others, oral, intravenous, intramuscular, dermal, or nasal routes. In some embodiments, the anti-inflammatory agents comprise non-steroidal anti-inflammatory agents. These agents generally act by inhibiting the action of cyclooxygenase and lipoxygenase enzymes, or receptors for mediators generated by these enzymes. The non-steroidal anti-inflammatory compounds include non-selective COX inhibitors, selective COX inhibitors, as well as FLAP antagonists and 5-lipoxygenase antagonists. In some embodiments, the anti-inflammatory agents can comprise anti-metabolites that affect proliferation of cells involved in the immune response. Suitable anti-metabolites include folate analogs, such as methotrexate; inosine monophosphate dehydrogenase (IMPDH) inhibitors, such as mycophenolate mofetil; and azathiopurine. Compounds of this group generally affect production of the substrates necessary for DNA replication, thereby inhibiting the proliferation of cells involved or activated in response to an inflammatory reaction. In some embodiments, the anti-inflammatory agent is an agent that blocks the action of TNF-alpha, the major cytokine implicated in inflammatory disorders. In some embodiments, the anti-TNF is an antibody that blocks the action of TNF alpha. An exemplary anti-TNF antibody is infliximab (Remicade®). In other embodiments, the anti-TNF alpha agent is a receptor construct that binds TNF alpha and prevents its interaction with TNF receptors on present on cells, e.g. entanercept (Enbrel®). In other embodiments, the anti-inflammatory agent is any other agent (e.g. an antibody agent) having immunosuppressive properties and useful in the treatment of the disorder being treated with the KIR3DL2 antibody described herein. Pharmaceutical Formulations Pharmaceutically acceptable carriers that may be used in these compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. The antibodies described herein may be employed in a method of modulating, e.g. inhibiting, the activity of KIR3DL2-expressing cells in a patient. This method comprises the step of contacting said composition with said patient. Such method will be useful for both prophylaxis and therapeutic purposes. For use in administration to a patient, the composition will be formulated for administration to the patient. The compositions described herein may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. The antibody can be present in a single dose in an amount, for example, of between about 25 mg and 500 mg. Sterile injectable forms of the compositions described herein may be aqueous or an oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation. The compositions described herein may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include, e.g., lactose. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. Alternatively, the compositions described herein may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols. The compositions described herein may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs. For topical applications, the compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds described herein include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. The present antibodies can be included in kits. The kits may optionally further contain any number of antibodies and/or other compounds, e.g., 1, 2, 3, 4, or any other number of therapeutic antibodies and/or compounds. It will be appreciated that this description of the contents of the kits is not limiting in any way. For example, the kit may contain other types of therapeutic compounds. Preferably, the kits also include instructions for using the antibodies, e.g., detailing the herein-described methods. Dosage Forms Therapeutic formulations of the antibodies are prepared for storage by mixing the antibodies having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions. For general information concerning formulations, see, e.g., Gilman et al. (eds.), The Pharmacological Bases of Therapeutics, 8th Ed. (Pergamon Press, 1990); Gennaro (ed.), Remington's Pharmaceutical Sciences, 18thEdition (Mack Publishing Co., Easton, Pa., 1990); Avis et al. (eds.), Pharmaceutical Dosage Forms: Parenteral Medications (Dekker, N.Y., 1993); Lieberman et al. (eds.), Pharmaceutical Dosage Forms: Tablets (Dekker, N.Y., 1990); Lieberman et al. (eds.) Pharmaceutical Dosage Forms: Disperse Systems (Dekker, N.Y., 1990); and Walters (ed.), Dermatological and Transdermal Formulations (Drugs and the Pharmaceutical Sciences), Vol. 119 (Dekker, N.Y., 2002). Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low-molecular-weight (less than about 10 residues) polypeptides; proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as ethylenediaminetetraacetic acid (EDTA); sugars such as sucrose, mannitol, trehalose, or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™, or PEG. Exemplary antibody formulations are described for instance in WO 1998/56418, which describes a liquid multidose formulation for an anti-CD20 antibody, comprising 40 mg/mL rituximab, 25 mM acetate, 150 mM trehalose, 0.9% benzyl alcohol, and 0.02% polysorbate20™ at pH 5.0 that has a minimum shelf life of two years storage at 2-8° C. Another anti-CD20 formulation of interest comprises 10 mg/mL rituximab in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium citrate dihydrate, 0.7 mg/mL polysorbate80™, and Sterile Water for Injection, pH 6.5. Lyophilized formulations adapted for subcutaneous administration are described, for example, in U.S. Pat. No. 6,267,958 (Andya et al.). Such lyophilized formulations may be reconstituted with a suitable diluent to a high protein concentration and the reconstituted formulation may be administered subcutaneously to the mammal to be treated herein. The formulation herein may also contain more than one active compound (a second medicament as noted above), preferably those with complementary activities that do not adversely affect each other. The type and effective amounts of such medicaments depend, for example, on the amount and type of B-cell antagonist present in the formulation, and clinical parameters of the subjects. Exemplary second medicaments are noted above. The active ingredients may also be entrapped in microcapsules prepared, e.g., by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, supra, for example. Sustained-release formulations may be prepared. Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antagonist, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and γ ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the Lupron Depot™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(−)-3-hydroxybutyric acid. The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes. Pharmaceutically acceptable carriers that may be used in these compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. Further aspects and advantages will be disclosed in the following experimental section, which should be regarded as illustrative and not limiting the scope of this application. EXAMPLES Example 1 Generation of Anti-KIR3DL2 Antibodies Materials and Methods Primary and Secondary Flow Cytometry Screenings Anti-KIR3DL2 mAbs were primarily screened in flow cytometry for binding to KIR3DL2-expressing Sézary cell lines (HUT78 and COU-L) and to KIR3DL2-transfected tumor cell lines (HEK-293T). Flow cytometry devices include: FACSarray (BD Biosciences, primary screen), FACSCanto II No. 1 and No. 2 (BD Biosciences) (secondary screens) and FC500 (Beckman Coulter) (secondary screens). The KIR3DL2+ and other tumor cell lines used included:HUT-78 (KIR3DL2 positive Sézary cell line) grown in complete IMDM;HEK-293T (human kidney cancer)/KIR3DL2 and HEK-293T/KIR3DL2 Domain 0— eGFP cell lines (grown in complete DMEM);COU-L (KIR3DL2 positive Sézary cell line) (grown in complete RPMI complemented with 10% human serum AB);HEK-293T/KIR3DL1 and HEK-293T/KIR3DL1— eGFP cell lines (grown in complete DMEM);B221 (B-lymphoblastoid, CD20 positive human cell line)/KIR3DL2 cell line (grown in complete RPMI containing FCS serum); andRAJI (Burkitt's lymphoma CD20 positive human cell line)/KIR3DL2 cell line (grown in complete RPMI containing FCS serum). Whereas none of the Sézary cell lines used grow after IV or SC transfer to immune compromised mice, KIR3DL2-transfected B221 or RAJI cells grow as disseminated (IV) or solid (SC) tumors after injection to mice. Based on the information available in Gardiner et al,Journal of Immunology2001 (Vol 166, p2992-3001), the KIR3DL2 gene alleles present in the tumor cell lines used were determined. We established that the Sézary cell line COU-L is heterozygous for alleles 3DL2*003 and 3DL2*008 and HUT-78 is heterozygous for alleles 3DL2*002 and 3DL2*007. All 4 alleles 3DL2*003, 3DL2*008, 3DL2*002 and 3DL2*007 encode KIR3DL2 protein variants bearing differences in their extracellular domains. Of note, the recombinant KIR3DL2-Fc fusion protein that was used to immunize mice is encoded by different KIR3DL2 gene alleles 3DL2*006 and 3DL2*007 (clone 1.1, both alleles encoding the same extracellular domain protein sequence). KIR3DL2 Domains 0, 1 and 2 Cell Lines HEK293T/17 cells were cultured in DMEM (Gibco) supplemented with sodium pyruvate (1 mM), penicillin (100 U/ml), streptomycin (100 μg/ml) and 10% heat inactivated FCS (PAN biotech). Lipofectamine 2000 reagent, Trizol, SuperScript II reverse Transcriptase, pcDNA3.1 vector and anti-V5-FITC antibodies were purchased from Invitrogen. Goat anti-mouse (H+L)−PE was purchased from Beckman Coulter. PBMC (5×106cells) from Homo Sapiens were re-suspended into 1 ml of Trizol reagent. RNA extraction was performed by adding 200 μl chloroform. After centrifugation (15 min, 13,000 rpm), RNA was precipitated from aqueous phase with 500 μl isopropanol. After incubation (10 min, RT) and centrifugation (10 min, 13,000), RNA was washed with 70% ethanol and re-centrifugated (5 min, 13,000 rpm). RNA was re-suspended in H2Od Rnase free water. cDNA was obtained using SuperScript II reverse Transcriptase using 2 μg of specific RNA and following manufacturer instructions. Human KIR3DL2 (accession number U30272, KIR3DL2 allele *002) domain 0, domain 1 and domain 2 sequences are shown in Table 4. TABLE 4Ig-likeSEQdomainIDof KIR3DL2NO:Amino acid sequenceDomain 068PLMGGQDKPF LSARPSTVVP RGGHVALQCHYRRGFNNFML YKEDRSHVPI FHGRIFQESFIMGPVTPAHA GTYRCRGSRP HSLTGWSAPSNPLVIMVTGN HRKPSLLAHP GPLLKSGDomain 169TVILQCWSDV MFEHFFLHRE GISEDPSRLVGQIHDGVSKA NFSIGPLMPV LAGTYRCYGSVPHSPYQLSA PSDPLDIVIT GLYEKPSLSAQPGPTVQAGEDomain 270NVTLSCSSWS SYDIYHLSRE GEAHERRLRAVPKVNRTFQA DFPLGPATHG GTYRCFGSFRALPCVWSNSS DPLLVSVTGN PSSSWPSPTEPSSKSGICRH LH Homo Sapiens KIR3DL2 (accession number U30272) domain 0, domain 1 and domain 2 sequences were amplified by PCR reaction from cDNA using 5′ AA GCT AGC GGT AAG CCT ATC CCT AAC CCT CTC CTC GGT CTC GAT TCT ACG CTC ATG GGT GGT CAG GAC AAA C (SEQ ID NO: 71) (forward) and 3′ AA GGA TCC CTC TCC TGA TTT CAG CAG GGT (SEQ ID NO: 72) (reverse); 5′ AA GCT AGC GGT AAG CCT ATC CCT AAC CCT CTC CTC GGT CTC GAT TCT ACG ACA GTC ATC CTG CAA TGT TGG (SEQ ID NO: 73) (forward) and 3′ AA GGA TCC CTC TCC TGC CTG AAC CGT GGG (SEQ ID NO: 74) (reverse); 5′ AA GCT AGC GGT AAG CCT ATC CCT AAC CCT CTC CTC GGT CTC GAT TCT ACG AAC GTG ACC TTG TCC TGT AGC (SEQ ID NO: 75) (forward) and 3′ AA GGA TCC ATG CAG GTG TCT GCA GAT ACC (SEQ ID NO: 76) (reverse) oligonucleotides, respectively. After TA-cloning and sequencing, sequences were cloned into pcDNA3.1 vector between NheI and BamHI restriction sites. These constructs were inserted between the CD33 peptide leader and the CD24 GPI anchor (CD24 GPI anchor DNA and amino acid sequences are shown in SEQ ID NOS: 77 and 78, respectively) synthesized by MWG Biotech (inserted between BamHI and HindIII restriction sites). HEK-293T/17 cells were seeded 24 hours prior to transfection into 6 wells plates (5·105cells/well) in DMEM without antibiotics. Transfections were performed using 5 μg of the different pcDNA3.1/KIR3DL2 domain 0, pcDNA3.1/KIR3DL2 domain 1 or pcDNA3.1/KIR3DL2 domain 2 constructs using Lipofectamine 2000 according to manufacturer instructions. To ensure DNA purity for transfection, Maxi-prep endotoxin free kit from Qiagen was used. The Lipofectamine/DNA ratio used was fixed at 2/1. Cells were harvested 48 hours after transfection for flow cytometry experiments. Immunization Mice were immunized with recombinant KIR3DL2-Fc fusion protein (allele *006). Supernatant (SN) of the growing hybridomas were tested by flow cytometry on HUT78, COU-L and HEK-293T/KIR3DL2 Domain 0—eGFP. Potentially interesting hybridomas selected from the initial screening were cloned by limiting dilution techniques in 96-wells plates. The secondary screen involved selection of hybridomas of interest by testing supernatants of the subclones by flow cytometry on HUT78, COU-L, HEK-293T/KIR3DL1 Domain 0—eGFP and HEK-293T/KIR3DL2 Domain 0—eGFP. Positive subclones were injected into mice to produce ascitis and antibodies of interest were purified before being tested in a Biacore assay using rec KIR3DL2 chips, followed by various assays formats based on binding to human KIR3DL2-expressing cells. Among the clones selected were supernatants for antibodies 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 4B5, 5H1, 1E2, 1C3 and 20E9. Based on the screen that permitted selection among D0 or D1/2 domain binding, antibodies 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 4B5, 5H1 and 1E2 bind to KIR3DL2 present on extracellular domain 0 (D0) while 1C3 and 20E9 bind to an epitope present on domain 1/2 (D2). Sequences of the variable domains of heavy (VH) and light (VL) chain of selected antibodies were amplified by PCR from the cDNA of each antibody. Sequences amplified were run on agarose gel then purified using the Qiagen Gel Extraction kit. VH and VL sequences were then sub-cloned into the Lonza expression vectors (Double-Gene Vectors) using the InFusion system (Clontech) according to the manufacturer's instructions. After sequencing, vectors containing the VH and VL sequences were prepared as Maxiprep using the Promega PureYield™ Plasmid Maxiprep System. Vectors were then used for HEK-293T cell transfection using Invitrogen's Lipofectamine 2000 according to the manufacturer instructions. Example 2 Antibodies that do not Induce KIR3DL2 Internalization Briefly, either no antibody or 20 μg/mL of an anti-KIR3DL2 domain 0 antibody, or antibody 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 4B5, 5H1, 1E2, 1C3 or 20E9 were incubated with fresh Sézary Syndrome cells from 5 different human donors, for 24h at 37° C. Cells were then washed, fixed and permeabilized using IntraPrep permeabilization reagent from Beckman Coulter. Presence of KIR3DL2-bound 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 4B5, 5H1, 1E2, 1C3 or 20E9 Ab is revealed with a goat anti-mouse Ab, labelled with GAM-PE. Table 6 shows an example of an anti-KIR3DL2 domain 0 antibody 13H1, after 24h incubation, respectively. Table 5 shows a strong decrease in fluorescence for 13H1 in each of the different donors, confirming that the binding of this antibody down-modulates the expression of KIR3DL2 on SS cells. Similar results were obtained for anti-D0 antibody 4B5 as well as a range of anti-D 1 antibodies. Conversely, the anti-KIR3DL2 domain 0 or domain 2 antibodies 10F6, 2B12, 18C6, 9E10, 10G5, 5H1, 1E2, 1C3 and 20E9 did not result in a decrease in fluorescence indicating that this antibody did not down-modulate the expression of KIR3DL2 on SS cells. Table 6 shows an representative example for antibody 10G5. TABLE 5KIR3DL2 mfi after 24 hKIR3DL2 mfi after 24 hPatientincubation without mAbincubation with 10 μg/ml mAbKLU1426592HAE2676871STA1095544CER475197 TABLE 6KIR3DL2 mfi after 24 hKIR3DL2 mfi after 24 hPatientincubation without mAbincubation with 10 μg/ml mAbKLU22374015HAE35874909STA15582786CER462733 Example 3 Antibodies that do not Internalize into Sezary Syndrome Cell Line Internalization of antibodies 10F6, 2B12, 18C6, 9E10, 10G5, 13H1, 4B5, 5H1, 1E2, 1C3 and 20E9, as well as antibody AZ158 (an anti-domain 0 mAb) and other anti-D1 antibodies were assessed by fluoro-microscopy using the HUT78 SS cell line. Materials and Methods: Hut-78 cells were incubated during 1H at 4° C. with 10 μg/ml of the different antibodies. After this incubation cells were either fixed (t=OH) or incubated for 2H at 37° C. Cells incubated for 2H were then fixed and stained. Antibodies were stained using goat anti-mouse antibodies coupled to Alexa594 (Invitrogen, A11032). LAMP-1 compartments were stained using rabbit anti-LAMP-1 antibodies (Abcam, ab24170) revealed by goat anti-rabbit polyclonal antibodies coupled to FITC (Abcam ab6717). Pictures were acquired using an Apotome device (Zeiss) and analyzed using the Axiovision software. Results: Anti-KIR3DL2 mAbs were visible in red while LAMP-1 compartments were visible in green. At the time of addition of antibodies, KIR3DL2 staining in red was visible at the cell surface while green LAMP-1 were visible intracellularly in green. However, at 2 hours following the addition of antibodies, each of antibodies AZ158, 13H1 and 4B5, and anti-D1 antibodies caused red staining to be colocalized with green staining, along with a decrease in red staining at the cell surface, indicating that AZ158, 13H1 and 4B5, and anti-D1 antibodies were rapidly internalized. Antibodies 10F6, 2B12, 18C6, 9E10, 10G5, 5H1, 1E2, 1C3 and 20E9, however was not internalized, and at 2 hours following the addition of antibody, red staining remained entirely on the cell surface. Example 4 Antibodies that Increase the Number of Available Cell Surface KIR3DL2 Receptors Part 1: Impact of Staining Conditions on 2B12 Labelling of KIR3DL2-Expressing Cells This study aimed to evaluate the impact of staining conditions on 2B12 labelling of KIR3DL2-expressing cells, gated on total cells, at 4° C. or 37° C., and with incubation times of 2 hours, 4 hours or 24 hours. Briefly, 100,000 HUT78 cells per well were incubated with a dose range of 2B12 antibody starting from 0.0005 μg/ml to 30 μg/ml (serial dilution 1/3 in complete medium). The protocol used was as follows: incubation 2H; 4H and overnight at 4° C. and 37°; staining in RPMI 10% with or without PFA fixation; 2 washes in SB (150 μl/w); addition of anti-human-Fc PE for 30 min at 4° C.; 2 washes in SB (100 μl/w); and detection using FACS CANTO II. Results are shown inFIG.6A. While incubation at 4° C. which inhibits receptor internalization/cycling was expected to result in an at least equal level of available cell-surface KIR3DL2, staining with antibody 2B12 (human IgG1) was higher at 37° C. than at 4° C. Furthermore, higher median fluorescence was observed with increasing duration of incubation, the greatest KIR3DL2 expression was observed after 24 hours of incubation. Part 2: Total, Free and 2B12-Bound KIR3DL2 Detection on HUT78 Tumor Cells after Overnight Incubation This study aimed to evaluate the impact of a 20 hour incubation with antibody 2B12 on cell surface KIR3DL2 level by observing the amount of bound 2B12 (human IgG1), free (non-antibody bound) cell surface KIR3DL2 polypeptide, and total cell surface KIR3DL2 polypeptide. Briefly, HUT78 (100,000 cells/well) were incubated for 20h at 37° C. with a dose range of 2B12 antibody starting from (decreasing) 8.88 μg/ml, 1/3 serial dilution, 11 concentrations. Dose ranges were made in duplicate in order to perform the following 2 staining conditions:1. Total KIR3DL2+bound KIR3DL2 (GaH IgG Fc-PE+mAb2-APC (a non-competing anti-KIR3LD2 mAb) (10 μg/ml)2. Free KIR3DL2: 2B12-PE (10 μg/ml) Staining was performed at 4° C. in staining buffer for 1h and analyzed with a FACS Canto II HTS. Results are shown inFIG.6B. The dark line/squares represents antibody 2B12 while the light line/circles represents isotype control. It can be seen that free KIR3DL2 receptors can be detected when incubating cells with 10 μg/ml of 2B12-PE and detecting free receptors with non-competing anti-KIR3DL2 antibody. It can be seen that 2B12-bound KIR3DL2 receptors can be detected by incubating cells with goat anti-human IgG Fc-PE secondary Ab. Both read-outs were correlated, with similar EC50. The rightmost panel shows that a 20 hour incubation with 2B12 increases KIR3DL2 receptor level at cell surface as detected by the non-competing anti-KIR3DL2 antibody mAb2-APC. Antibody 2B12 may be causing conformational change upon binding, receptor stabilization/accumulation at cell surface and/or internalization/recycling blockade. Part 3: Total, Free and 2B12-Bound KIR3DL2 Detection on HUT78 Tumor Cells after 1, 24 or 48 Hours This study aimed to evaluate the dynamics of KIR3DL2 receptor expression by observing by observing the amount of total, free and 2B12-bound KIR3DL2 after different periods of incubation with antibody 2B12. Briefly, HUT78 cells (50,000 cells/well) were incubated for 1h, 24h or 48h at 37° C. in complete medium with 2B12 (human IgG1), dose range starting from 10 μg/ml (decreasing), 1/3 serial dilution, 11 concentrations, or with isotype control (IC), dose range starting (decreasing) from 10 μg/ml, 1/3 serial dilution, 11 concentrations. Dose ranges were made in triplicate in order to perform 3 staining conditions:Bound KIR3DL2 (30 min at 4° C.): GaH IgG Fc-PE,Free KIR3DL2+total KIR3DL2 (1h at 4° C.): 2B12-PE (10 μg/ml)+mAb2-APC (non-competing anti-KIR3DL2) (10 μg/ml),Total KIR3DL2 (1h+30 min at 4° C.): 2B12 (10 μg/ml)+GaH IgG Fc-PE. Staining was performed at 4° C. in staining buffer, and analysis conducted with HTFC Intellicyt. Results are shown inFIG.6C. In these culture conditions (96 well-plates, 50,000 HUT78/well at T0), KIR3DL2 detection at cell surface decreases in the absence of any Ab (as detected by mAb2-APC, 2B12-PE or 2B12+GaH-PE, points on the Y-axis). Incubation with 2B12 at 37° C. increases surface expression of KIR3DL2 (as detected by non-competing mAb2 or by 2B12 itself+secondary Ab), in a dose-dependent manner. Isotype control did not give rise to any change in KIR3DL2. This increase is already observed after 1h at 37° C., and seems to reach its maximum after 24h. Staining is optimal after 24h (in terms of total staining and of detected Ab-bound receptors). Example 5 Antibodies are Able to Kill KIR3DL2 Expressing Targets Via Complement-Dependent Mechanism (CDC) Briefly, 50 μl of 20 μg/ml antibodies (2× concentrated) diluted were provided in standard medium a White clear bottom P96 wells (Ref 655098—Greiner), to which were added 50 μl of a cell suspension at 2 million per ml (100,000 cells per well) in standard medium, and incubated for 30 min at 4° C. 5 μl per well of freshly reconstituted complement (Ref CL3441—Cedarlan) was added, followed by incubation 1H at 37° C. 100 μl per well of Cell Titer Glo (Ref G7572—Promega) was added followed by incubation 10 min at room temperature protect from light. Results were read using a luminometer (VICTOR). Using complement purified from rabbit blood, the ability of our anti-KIR3DL2 mAbs to recruit complement and lyse KIR3DL2-transfected B221 cells was addressed in vitro. FIG.7Ashows ability of antibodies to mediate CDC; anti-KIR3DL2 mAbs that bind the D0 domain are in gray, those that bind the D1 domain are in black. With the parental murine mAbs, the isotype of the mAb has the most prominent influence on the result of this assay as IgG2b murine mAbs bind complement more efficiently than any other isotype (mouse IgG1 do not bind complement at all). To address the impact on complement-mediated target cell death of KIR3DL2 internalization upon binding with anti-KIR3DL2 mAbs, we used mo19H12, an anti-KIR3DL2 antibody that induces rapid internalization of KIR3DL2 into HUT78 Sézary cell line and B221-KIR3DL2. Before incubation with complement, we pre-incubated the target B221-KIR3DL2 targets with mo19H12 either at 4° C. (internalization is blocked) or 37° C. (that allows optimal internalization). Then, complement was added, incubated and CDC measured as above. In this experiment, the internalization of KIR3DL2 upon binding totally abrogates the ability of mo19H12 to kill B221-KIR3DL2 with complement recruitment, whereas in temperature conditions that limit internalization, CDC activity of mo19H12 is clearly observed (FIG.7B). Anti-CD20 rituximab is used as a control that mediates CDC against CD20+ targets but does not induce CD20 internalization. Selected mAbs were chimerized into human IgG1 to render them able of mediating effector functions (ADCC and CDC).FIG.8shows the ability of chimeric anti-KIR3DL2 mAbs to mediate CDC against B221-KIR3DL2 in vitro. Certain mAb clones like 1E2 and 10G5 have, after chimerization, acquired the ability to kill KIR3DL2 positive targets through a CDC mechanism. In this experiment, for anti-DO mAbs, potent internalization (such as that induced by 13H1, in black), might prevent optimal efficacy as observed for 1E2 and 10G5 in particular. Example 6 Antibodies are Able to Kill KIR3DL2 Expressing Targets Via Antibody Dependent Cellular Cytotoxicity (ADCC) Cell lysis through an ADCC mechanism was monitored in a radioactivity-based51Cr release experiment (the level of radioactivity released from the preloaded target cells being proportional to their death). One million target cells were loaded with51Cr for 1 hour at 37° C. and washed 3 times. 3,000 cells were seeded per well (U-shaped bottom 96-well plates) and test mAbs are added at 10 or 20 μg/ml final concentration (or increasing concentrations if dose-response relationship is studied). Effector cells were added at a defined effector:target ratio (in general 10:1) and the mixture was incubated at 37° C. for 4 h. Supernatant is analyzed on a Lumaplate apparatus. When chimeric huIgG1 mAbs are used, effector cells were allogeneic human NK cells purified from PBMCs taken from a healthy volunteer donor. For optimal assessment, ADCC experiments were performed generally using chimerized huIgG1 mAbs generated from various parental murine anti-KIR3DL2 mAbs.FIG.9shows the ability of a series of anti-KIR3DL2 mAbs, tested at the same final concentration (10 μg/ml), to kill the prototypical Sézary cell line HUT78 through an ADCC-mediated mechanism. FIG.10shows a similar experiment in which target cells used are KIR3DL2-transfected B221 which are overall more sensitive to ADCC-mediated killing by anti-KIR3DL2 mAbs. The mAbs shown in gray induce internalization of the receptor and seem to be less efficient than the 4 other mAbs that do not induce KIR3DL2 internalization. FIG.11shows a comparison of antibodies in a dose-ranging experiment the ability of chimerized huIgG1 anti-KIR3DL2 mAbs to mediate ADCC against KIR3DL2-expressing B221 targets, the efficacy profile of mAbs that do not induce internalization of the target (10F6, 2B12 and 10G5) is better than that of mAbs inducing KIR3DL2 internalization (13H1 and anti-DO mAbs 15C11 and 18B10). Example 7 Activity in Mouse Xenograft Models of KIR3DL2 Expressing Human Tumors Materials & Methods Immune compromised mice used for B221-KIR3DL2 and RAJI-KIR3DL2 models were NOD-SCID purchased from Charles River Laboratories. In the following models, 5 million human tumor cells (in 100 μl PBS as vehicle) were engrafted IV on Day 0 (DO), i.e. 1 day before treatment initiation (D1). From D1, mice were treated IV with different doses of mAbs (doses were adapted to mouse body weight) diluted in PBS, 2 injections per week for the duration of the whole experiment. Control groups included, depending on the experiment:PBS/placebo-treated mice as a control of normal/unaffected tumor growth;mice injected with the same dose of isotype control-matched mAbs directed against an irrelevant antigen. Mice were weighed and observed for clinical signs every 2 to 5 days depending on the model. Percent of body weight changes were calculated as compared to body weight at DO before tumor engraftment or to the highest body weight reached during the experiment. Mouse deaths or important weight losses were recorded and used to draw survival Kaplan-Meier curves and calculate improvement in survival as compared to control groups of mice. Results FIG.12shows the results of an experiment (n=6 NOD-SCID mice per group) in which the efficacy of 3 IgG2b isotype murine anti-KIR3DL2 9E10 and 19H12 (both given at 300 μg/mouse, twice a week) was tested against SC B221-KIR3DL2 xenografts. Non-internalizing anti-D0 antibody 9E10 showed increased survival compared to both PBS and internalizing anti-D1/D2 antibody 19H12. FIG.13shows the results of another experiment (n=6 NOD-SCID mice per group) in which the efficacy of murine anti-KIR3DL2 19H12 (given at 300 μg/mouse, twice a week) was tested against SC RAJI-KIR3DL2 xenografts. In vitro, KIR3DL2-transfected RAJI cells showed less internalization upon mAb binding than B221-KIR3DL2 or Sézary cell lines. In the RAJI-KIR3DL2 xenograft model, mo19H12 mAb was more efficient than in the B221-KIR3DL2 model. This is due to less potent internalization of the target in vivo. Example 8 Ligand Blockade Materials & Methods Antibody Inhibition of Tetramer Staining B27 dimer and HLA-A3 tetramer preparation and FACS staining have been described previously (Kollnberger, et al. (2007)Eur J Immunol37:1313-1322). HLA-A3 tetramers were prepared with For antibody inhibition experiments Baf3 cells transduced with KIR3DL2 were stained with 5 μg antibody or IgG1/IgG2a isotype control (Biolegend UK Ltd) at 4° C. for 20 minutes before staining with tetramer at room temperature for 20 minutes. Stained cells were then washed and fixed as described previously before FACS analysis on a BD Fortessa FACS machine. FACS analysis was performed using Flowjo software (Tree star Inc US). Antibody Inhibition of Jurkat KIR3DL2 CD3ε Reporter Cells Jurkat reporter cells transduced to express KIR3DL2CD3ε fusion protein have been previously described (Payeli, et al. (2012)Arthritis Rheum.). For antibody inhibition experiments reporter cells 100,000/well in RPM1640 (Sigma, supplemented with 10% FCS and penicillin and streptomycin) were first stained at 4° C. with 10 ug antibody or isotype control antibody for 20 minutes. Subsequently reporter cells were stimulated with 200,000 parental LCL.LBL.721.221 cells (hereafter referred to as 221 cells) or 221 cells transfected with HLA-B27 or control HLA-class 1 in a final volume of 200 Supernatants were harvested after overnight stimulation for IL-2 assay by ELISA (Ebiosciences UK Ltd) performed according to the manufacturer's instructions. Results D0 Domain-Specific Antibodies Inhibit HLA-A3 and B27dimer (B272) Tetramer Staining of KIR3DL2 Transduced Cells First we studied the ability of D0 and D 1/D2 domain-specific anti-KIR3DL2 antibodies to inhibit HLA-A3 and B27 dimer tetramer staining of KIR3DL2. The D0-domain specific anti-KIR3DL2 antibodies 1E2 and 13H1 consistently inhibited HLA-A3 and B27 heavy chain (B272) staining of KIR3DL2 transduced Baf3 cells (FIGS.14and16A). 2B12 inhibited B272while demonstrating negligible effects on HLA-A3 tetramer staining of KIR3DL2. By contrast 10G5 did not inhibit staining of KIR3DL2 with either HLA-A3 or B272tetramers. The D2 Domain Specific Antibody 1C3 Inhibits HLA A3 but not B27 Dimer (B272) Tetramer Staining of KIR3DL2 The D2 specific antibody 1C3 consistently inhibited HLA-A3 tetramer staining of KIR3DL2 transduced cells (FIGS.15and16B). By contrast 1C3 MAb did not affect B272staining of KIR3DL2 Baf3 cells and D1 specific antibodies did not significantly affect neither HLA-A3 nor B272tetramer staining of KIR3DL2 (FIGS.15and16B). D0 Domain but not D 1/D2 Specific Anti-KIR3DL2 MAbs Inhibit KIR3DL2 Reporter Cell Interactions with HLA-Class 1. We next determined the effect of KIR3DL2 specific antibodies on KIR3DL2 recognition of HLA-B27 and other HLA-class 1 by studying the effect of antibodies on IL-2 production KIR3DL2CD3ε transduced Jurkat reporter cells stimulated with 221 transfectants. In agreement with our previous findings 221 cells expressing HLA-B27 consistently stimulated 6 fold greater IL-2 production by KIR3DL2 reporter cells compared to stimulation with cells expressing control HLA class 1 (FIG.17). D0 domain-specific antibodies 2B12, 1E2, 10G5 and 13H1 all inhibited IL-2 production by KIR3DL2 reporter cells stimulated with HLA-B27 transfected cells to some degree (FIG.4) with 2B12 and 1E2 demonstrating the greatest inhibitory effect. By contrast the D2 specific antibody 1C3 had no significant effect on reporter cell recognition of HLA-B27. D0 domain-specific antibodies also inhibited IL-2 production by KIR3DL2 reporter cells stimulated with 221 cells transfected with HLA-B7, HLA-B35 and HLA-A2 and HLA-A3, although effects were less pronounced than those observed when cells were stimulated with HLA-B27. By contrast the D2 specific antibody 1C3 had no significant effect on IL-2 production by KIR3DL2 reporter cells stimulated with 221 cells transfected with HLA-B7, HLA-B35 and HLA-A2 and HLA-A3. Summary Here we show that monoclonal antibodies against the D0 domain of KIR3DL2 (2B12, 1E2, 13H1) inhibit binding to β2m-free B27 heavy chain dimers (B272) and β2m-associated ligands such as HLA-A3. By contrast antibodies against the D2 domain of KIR3DL2 (1C3) only inhibit interactions with HLA-A3 and have little effect on B272tetramer binding. Although KIR3DL2 reporter T cells produce IL-2 when stimulated with HLA-B27, HLA-B7, HLA-B35, HLA-A2 and HLA-A3 transfected LBL.721.221 B cells, reporter cells consistently produce 6 fold higher IL-2 in response to HLA-B27. KIR3DL2 interactions with B cells expressing HLA-B27 and other HLA-class 1 are consistently inhibited with the D0 domain specific antibodies 2B12 and 1E2 and to a lesser extent 13H1. This suggests that the D0 domain of KIR3DL2 may have some affinity for common shared features of different HLA-class 1. The KIR3DL2 D0 domain may bind at least in part to a region in HLA-B27 which is shared between different HLA class 1. The increased avidity of KIR3DL2 for HLA-B27 may result from dimerization of B27 heavy chains. It has been reported that the three immunoglobulin-like domains D0, D1 and D2 of KIR3DL2 are involved in binding ligand. The results from the antibody inhibition studies suggest that the dominant contact of KIR3DL2 with HLA-class 1 is via the D0 domain. Notably the D2 antibody 1C3 only inhibited HLA-A3 binding to KIR3DL2 and not B27 dimer. We therefore propose that the D0 domain contacts residues are conserved between different HLA-class 1 and the D1 and D2 domains contact polymorphic regions and peptide in the peptide MHC complex. The antibodies identified can be used for therapeutic, diagnostic and other research applications depending on the particular application, to selectively block different ligands, or to block multiple ligands, or to not compete with ligands for binding to KIR3DL2. Example 9 Epitope Mapping KIR3DL2 mutants were developed to identify KIR3DL2-specific antibodies that had desired binding properties. Antibodies will advantageously have binding to most or all of the major KIR3DL2 alleles in the population (in terms of allele frequency) while not binding to the major KIR3DL1 alleles (e.g. allele *00101). Mutations were generated that corresponded to residues that differ between KIR3DL1 and KIR3DL2. A first set of mutations were generated in which the amino acid in KIR3DL1 were substituted into KIR3DL2. However, many of these mutated proteins failed to express at the cell surface, suggesting that incorporating the KIR3DL1 deeply impacted the folding of the entire KIR3DL2 molecule. In particular, mutants in clusters D21, D22, D23, D26 and D27 shown below in Table 7A did not express at the cell surface. TABLE 7AClusterResidues in KIR3DL1Residues in KIR3DL2D21V196; P199; D285; P286I196; L199; N285; S286D22K212; S218T212; N218D23R226W226D26R249P249D27H278; S279; E282; Y281A278; L279; V282; C281 Further mutants were redesigned and tested, with the final set of mutations shown in Table 7B below. Antibodies were tested for binding KIR3DL2 to various KIR3DL2 mutants. KIR3DL2 mutants were generated by PCR (see Table 7B below). All the Mx-R primers were used with the following 5′ primer ACCCAAGCTGGCTAGCATGTCGCTCACGGTCGTCAGCATG (SEQ ID NO: 79). All the Mx-F primers were used with the following 3′ primer AGCACAGTGGCGGCCGCCTAGAAAA CCCCCTCAAGACC (SEQ ID NO: 80). The sequences amplified were run on agarose gel then purified using the Qiagen Gel Extraction kit. To create mutants 12 and 21, it was necessary to do a third PCR. Primers used for these PCR were: M12a-F primer (5′-GCCACAGGTGCATATGAGAAACCTTCTCTCTCAGCC-3′) (SEQ ID NO: 81) with the M12b-R primer (5′-TGGGTCACTTGCGGCTGACCACACGCAGGGCAGGG-3′) (SEQ ID NO: 82) and M21a-F primer (5′-CGTGCCCTGCCCTACGTGTGGTCAAACTCAAGTGAC-3′) (SEQ ID NO: 83) with the M21b-R primer (5′-ATG CAGGTGTCTGGGGATACCAGATTTGGAGCTTGGTTC-3′) (SEQ ID NO: 84). The two or three PCR products generated for each mutant were then ligated into a pcDNA3.1 vector, digested with the restriction enzyme NheI and NotI, with the InFusion system (Clontech) according to the manufacturer's instructions. After sequencing, the vectors containing the mutated sequences were prepared as Maxiprep using the Promega PureYield™ Plasmid Maxiprep System. Vectors were then used for HEK-293T cell transfection using Invitrogen's Lipofectamine 2000 according to the manufacturer instructions. TABLE 7BMutantsReverse primersForward primersNumber 1M1-RM1-FR13W +5′-ccgaagagtcacgtgtcctcctcgaggcaccac5′-cacgtgactcttcggtgtcactatcgtcgA25T +agtgctgggccaggcaca-3′tggg-3′Q27R(SEQ ID NO: 85)(SEQ ID NO: 86)Number 2M2-RM2-FI60N +5′-cacagggctcatgttgaagctctcctggaatattc-3′5′-aacatgagccctgtgaccccagcacatg-3′G62S(SEQ ID NO: 87)(SEQ ID NO: 88)Number 3M3-RM3-FR32H +5′-attgttaaacctatgacgatagtgacactgaagag-3′5′-cataggtttaacaatttcatgctgtac-3′G33R(SEQ ID NO: 89)(SEQ ID NO: 90)Number 4M4-RM4-FS45I +5′-gatgggaatgtggattctgtcttctttgtacagca5′-atccacattcccatcttccacggcagaatV45Itg-3′attc-3′(SEQ ID NO: 91)(SEQ ID NO: 92)Number 5M5-RM5-FP66T5′-atgtgctgtggtcacagggcccatgatgaag-3′5′-gtgaccacagcacatgcagggacctacag-3′(SEQ ID NO: 93)(SEQ ID NO: 94)Number 6M6-RM6-FR78H +5′-gggggagtgtgggtgtgaaccccgacatctgtag-3′5′-cacccacactcccccactgggtggtcggcac-3′L82P(SEQ ID NO: 95)(SEQ ID NO: 96)Number 7M7-RM7-FL113V +5′-ttgcaggatgactctctctcctgatttcaccagggg-3′5′-agagtcatcctgcaatgttggtcagatgtc-3′T118R(SEQ ID NO: 97)(SEQ ID NO: 98)Number 8M8-RM8-FV127I5′-ctcaaacatgatatctgaccaacattgcaggatga5′-gatatcatgtttgagcacttctttctgcac-3′(SEQ ID NO: 98)(SEQ ID NO: 100)Number 9M9-RM9-FL164M +5′-aagggcaagcatcatgggaccgatggagaagttgg5′-atgatgcttgcccttgcaggaacctacagP166L +ccttg-3′atgttat gg-3′V167A(SEQ ID NO: 101)(SEQ ID NO: 102)NumberM10-RM10-F105′-tagagatcccatctttgtgcagaaagaagtgctca5′-aagatgggatctctaaggacccctcacgcR136K +aacat-3′ctcgttgg-3′E141K(SEQ ID NO: 103)(SEQ ID NO: 104)NumberM11-RM11-F115′-gggggtgtgagtaacagaaccataacatctgtagg-3′5′-gttactcacaccccctatcagttgtcagcP179T +(SEQ ID NO: 105)tc-3′S181T(SEQ ID NO: 106)NumberM12a-RM12b-F125′-atatgcacctgtggccacgatgtccagggggtcac5′-gccgcaagtgacccactgcttgtttctgtI196A +tgg-3′c-3′L199A +(SEQ ID NO: 107)(SEQ ID NO: 108)N285A +S286ANumberM13-RM13-F135′-ggcctctcctgcctgaaccgcggggcccggctggg5′-caggcaggagaggccgtgaccttgtcctgT212A +ctgag-3′tagctcc-3′N218A(SEQ ID NO: 109)(SEQ ID NO: 110)NumberM14-RM14-F145′-ataggagctcgcggagctacaggacaaggtcac-3′5′-tccgcgagctcctatgacatctaccatctW226A(SEQ ID NO: 111)gtcc-3′(SEQ ID NO: 112)NumberM15-RM15-F155′-atgggcctccccttccctggacagatggtacatgt5′-gaaggggaggcccatgaacgtaggctcccI231M +catagga-3′tgcagtg-3′R246P(SEQ ID NO: 113)(SEQ ID NO: 114)NumberM16-RM16-F165′-atgtgctccaccttccctggacagatggtagatgt5′-gaaggtggagcacatgaacgtaggctccgE239Gc-3′tgcagtg-3′(SEQ ID NO: 115)(SEQ ID NO: 116)NumberM17-RM17-F175′-tctgttgaccttggccactgcacggagcctacgtt5′-gccaaggtcaacagaacattccaggcagaP249Ac-3′c-3′(SEQ ID NO: 117)(SEQ ID NO: 118)NumberM18-RM18-F185′-cgcggcgggcgcgtgacggaaagagccgaagcatc5′-cacgcgcccgccgcgtggtcaaactcaagA278H +tg-3′tgaccc-3′L279A +(SEQ ID NO: 119)(SEQ ID NO: 120)C281A +V282ANumberM19-RM19-F195′-ctcgcagggcgagtgacggaaagagccgaagcatc5′-cactcgccctgcgagtggtcaaactcaagA278H +tgtag-3′tgaccc-3′L279S +(SEQ ID NO: 121)(SEQ ID NO: 122)V282ENumberM21a-RM21b-F215′-gtagggcagggcacggaaagagccgaagca-3′5′-cccagacacctgcatgttctgattg-3′C281Y +(SEQ ID NO: 123)(SEQ ID NO: 124)C315PNumberM22a-RM22a-F225′-tgtggt cac agg gcc cat gat gaa5′-ggc cct gtg accAca gca(5 + 11)gct ctc ctg gaa tat tc-3′cat gca ggg acc tac aga-3′P66T +(SEQ ID NO: 125)(SEQ ID NO: 126)P179T;S181TNumberM22b-RM22b-F225′-gtc act ggg agc tga caa ctg ata5′-tca gct ccc agt gac ccc(5 + 11)ggg ggTgtg agTaac-3′ctg gac atc gtg atc aca gg-3′(SEQ ID NO: 127)(SEQ ID NO: 128)NumberM23a-RM23a-F235′-gaTatc tga cca aca ttg cag gat5′-tgt tgg tca gatAtc atg(8 + 11)gac tgt ctc tcc-3′ttt gag cac ttc ttt ctg-3′V127I +(SEQ ID NO: 129)(SEQ ID NO: 130)P179T +S181TNumberSame primers as M22b-RSame primers as M22b-F23(8 + 11)V127I +P179T +S181TNumberM24-RM24-F245′-gga gGAagg aGc aga acc ata aca5′-tct gCt cctTCc Tcc ccc(11A1)tct gta ggt tcc-3′tat cag ttg tca gct ccc-3′V178A +(SEQ ID NO: 131)(SEQ ID NO: 132)H180SNumberM26a-RM26a-F265′-gat cac cag ggg gtt gct ggg5′-aac ccc ctg gtg atc atg(11A3)agc cga cca ccc-3′gtc aca gga aGcTCc AGA AAAQ184A +(SEQ ID NO: 133)CCT TCC-3′H100S +(SEQ ID NO: 134)N99SNumberM26b-RM26b-F265′-gtc cag ggg gtc act ccc agc5′agt gac ccc ctg gac atc(11A3)tga caa cGCata ggg gga gtg agg-3′gtg atc aca ggt c-3′Q184A +(SEQ ID NO: 135)(SEQ ID NO: 136)H100S +N99SNumberM27-RM27-F275′-aga gat ccc atc tct gtg cag5′-aga gat ggg atc tct Gag(11A4)aaa gaa gGAcGAaaa c-3′gac ccc tcaAgc ctc-3′E130S +(SEQ ID NO: 137)(SEQ ID NO: 138)H131S +R145SNumberM28-RM28-F285′-atg gat cGAtcc aGc gag gcg tga5′-gCt ggaTCg atc cat gat(11A5)ggg gtc ctc-3′ggg gtc tcc aag gcc-3′V147A +(SEQ ID NO: 139)(SEQ ID NO: 140)Q149SNumberM29-RM29-F295′-ctc aga gat ccc atc tct gtg cag5′-gat ggg atc tct Gag gac(11A6)aaa gaa gtg ctc aaa cGCgac-3′ccc tca cgc ctc gtt gga cagI150A +(SEQ ID NO: 141)GCc cat g-3′M128A(SEQ ID NO: 142)NumberM30a-RM30a-F305′-tcc tcc tcg agg cac cac agt5′-gtg cct cga Gga gga cac(1 + 2)gct ggg ccAggc ag-3′gtgAct ctt cGg tgt cac tatR13W +(SEQ ID NO: 143)cg-3′A25T +(SEQ ID NO: 144)Q27R +I60N +G62SNumberM30b-RM30b-F305′-tgg ggt cac agg gcTcat gTt5′-Agc cct gtg acc cca gca(1 + 2)gaa gct ctc ctg g-3′cat gca ggg acc tac aga tgtR13W +(SEQ ID NO: 145)cg-3′A25T +(SEQ ID NO: 146)Q27R +I60N +G62SNumberM31-RM31-F315′-ggc agCcag gGa ggg ttt gtc ctg5′-ccc tCc ctgGct gcc cgg(D0-acc acc cat g-3′ccc agc act gtg gtg cc-3′HLA1)(SEQ ID NO: 147)(SEQ ID NO: 148)F9S +S11ANumberM32-RM32-F325′-ccc acg acg ata gGAaca ctg aag5′-TCc tat cgt cgt gggGCt(D0-agc cac gtg tcc-3′aac aat ttc atg ctg tac-3′HLA2)(SEQ ID NO: 149)(SEQ ID NO: 150)H29S +F34ANumberM33-RM33-F335′-tatGct gcc gtg gGCgat ggg aac5′-GCc cac ggc agCata ttc(1 + 2gtg gct tct g-3′cag gag agc ttc atc-3′A1)(SEQ ID NO: 151)(SEQ ID NO: 152)F50A +R53SNumberM34-RM34-F345′-gaa gct cGc cGAgaa tat tct gcc5′-ttcTCg gCg agc ttc atc(1 + 2gtg gaa gat gg-3′Atg ggc cct gtg acc-3′A2)(SEQ ID NO: 153)(SEQ ID NO: 154)Q56S +E57ANumberM35-RM35-F355′-tcc tcg agg cac cac agt gGCggA5′-gtg gtg cct cga Gga gga(1 + 2ccg ggc aga cag-3′TCc gtg gct ctt cag tgt c-3′A3)(SEQ ID NO: 155)(SEQ ID NO: 156)P14S +S15A +H23SNumberM37-RM37-F375′-gtc ctcTTGgat ccc atc tct gtg5′-ggg atcCAAGag gac ccc(1 + 2cag aaa g-3′tca cgc ctc gtt gg-3′A5)(SEQ ID NO: 157)(SEQ ID NO: 158)S140Q Each antibody was tested for binding to wild-type KIR3DL2 and to each of the D0, D1 and D2 domain mutants. Antibodies did not show any loss of binding to unmutated wild type KIR3DL2 (WTaKIR3DL2) but lost binding to one or more mutants, thereby identifying several epitopes. A summary is shown in Tables 7C and 7D (“+” indicates no significant loss of binding, “+/−” indicates a decrease in binding (or partial loss of binding) and “−” indicates substantially complete loss of binding). Most non-internalizing D0 antibodies lost substantially all binding to mutant 2 (four antibodies: 10F6, 2B12, 18C6, 10G5). All of these antibodies also had at least partial loss of binding to mutant 2A3. One non-internalizing D0 antibody showed loss of binding to only mutant 1 (9E10). One non-internalizing D0 antibody (1E2) lost binding only to mutant 2A3. One antibody (5H1) lost binding to mutant 6. Natural ligand blocking and internalizing antibody 13H1 additionally showed decreased binding to mutant 2A2 and MD0/HLA1, in addition to mutants 1 and 2. As to the antibodies that bound domain D2 of KIR3DL2 (both non-internalizing) antibodies 1C3 and 20E9 lost binding to mutant 14, as well as partial loss of binding to mutant 15 and mutant 16. Antibodies 10F6, 2B12, 18C6 and 10G5 had loss of binding to mutant 2 having I60N and G62S substitutions and decrease in binding to mutant 2A3 having P14S, S 15A and H23S substitutions, but did not lose binding to any other mutants. The principal epitope of these antibodies therefore includes residues I60 and/or G62 (and the epitope optionally further includes one or more of P14, S15, and H23). Residues 60 and 62 are within the D0 domain of KIR3DL2. Antibody 13H1 had loss of binding to both mutant 1 having R13W, A25T and Q27R and to mutant 2 having I60N and G62S substitutions. 13H1 also had decreased binding to mutant 2A2 (Q56S, E57A) and mutant MD0/HLA1 (F9S, S11A). The epitope of 13H1 therefore includes residues F9, S11, Q56 and/or E57. These residues are within the D0 domain. Antibody 9E10 had decreased binding to mutant 1 having R13W, A25T and Q27R substitutions, but not to any other mutants. The epitope of 9E10 and 10G5 therefore includes residues R13, A25 and/or Q27. FIG.1shows a view of the KIR3DL2 polypeptide, including portions within the D0 domain, showing amino acid residues mutated indicated as “Mutant 1”, “Mutant 2”, “Mutant 3” and “Mutant 6” which resulted (in different combinations) in loss of binding by antibodies.FIG.2shows a view of the KIR3DL2 polypeptide, including portions within the D0 domain, showing amino acid residues mutated indicated as “Mutant 1”, “Mutant 2” and “Mutant 3” which resulted (in different combinations) in loss of binding by antibodies, with shading of residues adjacent to residues (F9, S11, P14, F34 and/or S140 adjacent to mutant 2, and G21, G22, H23, E57, S58, F59, P63 and/or H68 adjacent to mutant 1). Antibody 5H1 had loss of binding to mutant 6 having R78H and L82P substitutions, but did not lose binding to any other mutants. The principal epitope of 5H1 therefore includes residues R78 and/or L82. Residues R78 and L82 are within the D0 domain of KIR3DL2. Surface-exposed residues adjacent to these mutated residues can also contribute to the epitopes of the antibodies.FIG.3shows a view of each face of the KIR3DL2 polypeptide, including portions within the D0 domain, showing amino acid residues mutated indicated as “Mutant 6” which resulted (in different combinations) in loss of binding by antibodies, with “Mutant 3” that did not result in loss of binding shown. Also shown in shading are residues adjacent to residues adjacent to mutant 6 that may also be bound by the antibodies (K7, Y30, R31, P79, H80, S81, T83, G84, W85, S86 and/or A87). Antibodies 1C3 and 20E9 had loss of binding to mutant 14 having a W226A substitution. The antibodies additionally had decreased binding to mutant 15 having I231M and R246P substitutions and to mutant 16 having an E239G substitution. The principal epitope of 1C3 therefore includes residues W226. The principal epitope of 20E9 may include residues I231M and/or R246P, and/or may additionally include E239. Residues W226, I231 and R246 are in the region of the junction of the D1 and D2 domains of KIR3DL2. Surface-exposed residues adjacent to the mutated residues can also contribute to the epitopes of the antibodies, including for example residues Q201, K202, P203, S204, S224, S225, S227, S228, N252, R253 and/or T254 (reference to SEQ ID NO: 1) located at the surface of KIR3DL2 in the region of the W226 epitope but outside of the region of the KIR3DL2 mutations which did not result in loss of binding of the antibodies (e.g. mutants 12 and 17). Surface-exposed residues adjacent to the mutated residues I231 and R246 can also contribute to the epitopes of the antibodies, including for example residues D230, I231, R244, L245, R246, A247, V248, S275, R277 and/or P280 (reference to SEQ ID NO: 1) located at the surface of KIR3DL2 in the region of the I231/R246 epitope but outside of the region of the KIR3DL2 mutations which did not result in loss of binding of the antibodies (e.g. mutants 12 and 17). FIG.4shows a view of the KIR3DL2 polypeptide, including portions within the D2 domain (D1/D2 junction), showing amino acid residues mutated indicated as “Mutant 14” to which antibodies lost binding, and “Mutant 12” and “Mutant 17” which did not cause loss of binding by antibodies; also shown in shading are residues adjacent to residues (Q201, K202, P203, S204, S224, S225, S227, S228, N252, R253 and/or T254 adjacent to mutant 14) that may also be bound. FIG.5shows a view of the KIR3DL2 polypeptide, including portions within the D2 domain (D1/D2 junction), showing amino acid residues mutated indicated as “Mutant 15” to which antibodies lost binding; also shown in shading are residues adjacent to residues (D230, I231, R244, L245, R246, A247, V248, S275, R277 and/or P280) adjacent to mutant 14) that may also be bound. FIGS.18,19and20show views of the KIR3DL2 polypeptide, allele *001, with the binding site of mutant 2 shown, and showing amino acid differences seen in different KIR3DL2 alleles having highest frequency (studies of populations in the United States). It can be seen that the antibody binding site is at a site that is conserved across KIR3DL2 alleles, which is consistent with the ability of the antibody to bind cells from all individuals tested. TABLE 7CDomain 0 mutantsKIR3DL2 D0 AntibodiesMutantsMutations1E210F62B1218C69E1010G513H15H1M1R13W; A25T; Q27R+++++/−+/−−+M2I60N; G62S+−−−++/−−+M3R32H; G33R++++++++M4S45I; V47I++++++++M5P66T++++++++M6R78H; L82P+++++++−M1 + 2A1F50A; R53S+++++++M1 + 2A2Q56S; E57A+++++++/−M1 + 2A3P14S; S15A; H23S−+/−+/−−++/−+M1 + 2A5S140Q+++++++M1 + 2R13W; A25T;+−−−++/−−Q27R; I60N; G62SMD0/HLA1F9S; S11A+++++++/−MD0/HLA2H29S; F34A++++++ TABLE 7DDomain 2 mutantsKIR3DL2 D2AntibodiesMutantsMutations1C320E9M12I196A; L199A; N285A; S286A++M13T212A; N218A++M14W226A−−M15I231M; R246P+/−+/−M16E239G+/−+/−M17P249A++M18A278H; L279A; C281A; V282A++M19A278H; L279S; V282E++M21C281Y; C315P++ All headings and sub-headings are used herein for convenience only and should not be construed as limiting the invention in any way. Any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. Unless otherwise stated, all exact values provided herein are representative of corresponding approximate values (e. g., all exact exemplary values provided with respect to a particular factor or measurement can be considered to also provide a corresponding approximate measurement, modified by “about,” where appropriate). All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise indicated. No language in the specification should be construed as indicating any element is essential to the practice of the invention unless as much is explicitly stated. The citation and incorporation of patent documents herein is done for convenience only and does not reflect any view of the validity, patentability and/or enforceability of such patent documents, The description herein of any aspect or embodiment of the invention using terms such as reference to an element or elements is intended to provide support for a similar aspect or embodiment of the invention that “consists of,” “consists essentially of” or “substantially comprises” that particular element or elements, unless otherwise stated or clearly contradicted by context (e.g., a composition described herein as comprising a particular element should be understood as also describing a composition consisting of that element, unless otherwise stated or clearly contradicted by context). This invention includes all modifications and equivalents of the subject matter recited in the aspects or claims presented herein to the maximum extent permitted by applicable law. All publications and patent applications cited in this specification are herein incorporated by reference in their entireties as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to one of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.
226,537
11858991
DETAILED DESCRIPTION OF THE INVENTION The present invention is directed to LAG-3 binding molecules that comprise the LAG-3-binding domain of selected anti-LAG-3 antibodies: LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 that are capable of binding to both cynomolgus monkey LAG-3 and to human LAG-3. The invention particularly concerns LAG-3 binding molecules that are humanized or chimeric versions of such antibodies, or that comprise LAG-3 binding-fragments of such anti-LAG-3 antibodies (especially immunocongugates, diabodies, BiTEs, bispecific antibodies, etc.). The invention particularly concerns such LAG-3-binding molecules that are additionally capable of binding an epitope of a molecule involved in regulating an immune check point that is present on the surface of an immune cell. The present invention also pertains to methods of using such LAG-3 binding molecules to detect LAG-3 or to stimulate an immune response. The present invention also pertains to methods of combination therapy in which a LAG-3-binding molecule that comprises one or more LAG-3-binding domain(s) of such selected anti-LAG-3 antibodies is administered in combination with one or more additional molecules that are effective in stimulating an immune response to thereby further enhance, stimulate or upregulate such immune response in a subject. I. Antibodies and their Binding Domains The antibodies of the present invention are immunoglobulin molecules capable of immunospecific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one epitope recognition site, located in the Variable Domain of the immunoglobulin molecule. As used herein, the term “antibody” refers to an immunoglobulin molecule capable of immunospecific binding to a polypeptide or protein or a non-protein molecule due to the presence on such molecule of a particular domain or moiety or conformation (an “epitope”). An epitope-containing molecule may have immunogenic activity, such that it elicits an antibody production response in an animal; such molecules are termed “antigens”). Epitope-containing molecules need not necessarily be immunogenic. The binding domains of the present invention bind to epitopes in an “immunospecific” manner. As used herein, an antibody, diabody or other epitope-binding molecule is said to “immunospecifically” bind (or to exhibit “specific” binding to) a region of another molecule (i.e., an epitope) if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with that epitope relative to alternative epitopes. For example, an antibody that specifically binds to a LAG-3 epitope is an antibody that binds such LAG-3 epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other LAG-3 epitopes or to a non-LAG-3 epitope. It is also understood by reading this definition that, for example, an antibody (or moiety or epitope) that immunospecifically binds to a first target may or may not specifically or preferentially bind to a second target. As such, “immunospecific binding” does not necessarily require (although it can include) exclusive binding. Generally, but not necessarily, reference to binding means “specific” binding. Two molecules are said to be capable of binding to one another in a “physiospecific” manner, if such binding exhibits the specificity with which receptors bind to their respective ligands. The ability of an antibody to immunospecifically bind to an epitope may be determined by, for example, an immunoassay. Natural antibodies (such as IgG antibodies) are composed of two Light Chains complexed with two Heavy Chains. Each light chain contains a Variable Domain (VL) and a Constant Domain (CL). Each heavy chain contains a Variable Domain (VH), three Constant Domains (CH1, CH2 and CH3), and a “Hinge” Domain (“H”) located between the CH1 and CH2 Domains. The basic structural unit of naturally occurring immunoglobulins (e.g., IgG) is thus a tetramer having two light chains and two heavy chains, usually expressed as a glycoprotein of about 150,000 Da. The amino-terminal (“N-terminal”) portion of each chain includes a Variable Domain of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal (“C-terminal”) portion of each chain defines a constant region, with light chains having a single Constant Domain and heavy chains usually having three Constant Domains and a Hinge Domain. Thus, the structure of the light chains of an IgG molecule is n-VL-CL-c and the structure of the IgG heavy chains is n-VH-CH1-H-CH2-CH3-c (where n and c represent, respectively, the N-terminus and the C-terminus of the polypeptide). The ability of an antibody to bind an epitope of an antigen depends upon the presence and amino acid sequence of the antibody's VL and VH Domains. Interaction of an antibody light chain and an antibody heavy chain and, in particular, interaction of its VL and VH Domains forms one of the two epitope-binding sites of a natural antibody. Natural antibodies are capable of binding to only one epitope species (i.e., they are monospecific), although they can bind multiple copies of that species (i.e., exhibiting bivalency or multivalency). The Variable Domains of an IgG molecule consist of the complementarity determining regions (CDR), which contain the residues in contact with epitope, and non-CDR segments, referred to as framework segments (FR), which in general maintain the structure and determine the positioning of the CDR loops so as to permit such contacting (although certain framework residues may also contact antigen). Thus, the VLand VH Domains have the structure n-FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4-c. Polypeptides that are (or may serve as) the first, second and third CDR of an antibody Light Chain are herein respectively designated CDRL1 Domain, CDRL2 Domain, and CDRL3 Domain. Similarly, polypeptides that are (or may serve as) the first, second and third CDR of an antibody heavy chain are herein respectively designated CDR1Domain, CDRH2 Domain, and CDRH3 Domain. Thus, the terms CDRL1 Domain, CDRL2 Domain, CDRL3 Domain, CDRH1 Domain, CDRH2 Domain, and CDRH3 Domain are directed to polypeptides that when incorporated into a protein cause that protein to be able to bind to a specific epitope regardless of whether such protein is an antibody having light and heavy chains or a diabody or a single-chain binding molecule (e.g., an scFv, a BiTe, etc.), or is another type of protein. Accordingly, as used herein, the term “Epitope-Binding Domain” refers to that portion of an epitope-binding molecule that is responsible for the ability of such molecule to immunospecifically bind an epitope. An epitope-binding fragment may contain 1, 2, 3, 4, 5 or all 6 of the CDR Domains of such antibody and, although capable of immunospecifically binding to such epitope, may exhibit an immunospecificity, affinity or selectivity toward such epitope that differs from that of such antibody. Preferably, however, an epitope-binding fragment will contain all 6 of the CDR Domains of such antibody. An epitope-binding fragment of an antibody may be a single polypeptide chain (e.g., an scFv), or may comprise two or more polypeptide chains, each having an amino terminus and a carboxy terminus (e.g., a diabody, a Fab fragment, an F(ab′)2 fragment, etc.). As used herein, the term “antibody” encompasses monoclonal antibodies, multispecific antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, polyclonal antibodies, camelized antibodies, single-chain Fvs (scFv), single-chain antibodies, immunologically active antibody fragments (e.g., antibody fragments capable of binding to an epitope, e.g., Fab fragments, Fab′ fragments, F(ab′)2 fragments, Fv fragments, fragments containing a VL and/or VH domain, or that contain 1, 2, or 3 of the complementary determining regions (CDRs) of such VL domain (i.e., CDRL1, CDRL2, and/or CDRL3) or VH domain (i.e., CDRH1, CDRH2, and/or CDRH3)) that specifically bind an antigen, etc., bi-functional or multi-functional antibodies, disulfide-linked bispecific Fvs (sdFv), intrabodies, and diabodies, and epitope binding fragments of any of the above. In particular, the term “antibody” is intended to encompass immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain an epitope-binding site. Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1and IgA2) or subclass (see, e.g., United States Patent Publication Nos.: 20040185045; 20050037000; 20050064514; 20050215767; 20070004909; 20070036799; 20070077246; and 20070244303). The last few decades have seen a revival of interest in the therapeutic potential of antibodies, and antibodies have become one of the leading classes of biotechnology-derived drugs (Chan, C. E. et al. (2009) “The Use Of Antibodies In The Treatment Of Infectious Diseases,” Singapore Med. J. 50(7):663-666). Over 200 antibody-based drugs have been approved for use or are under development. The anti-LAG-3 antibodies of the present invention include humanized, chimeric or caninized variants of antibodies LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6. The term “chimeric antibody” refers to an antibody in which a portion of a heavy and/or light chain is identical to or homologous with an antibody from one species (e.g., mouse) or antibody class or subclass, while the remaining portion is identical to or homologous with an antibody of another species (e.g., human) or antibody class or subclass, so long as they exhibit the desired biological activity. Chimeric antibodies of interest herein include “primatized” antibodies comprising variable domain antigen binding sequences derived from a non-human primate (e.g., Old World Monkey, Ape, etc.) and human constant region sequences. The term “monoclonal antibody” as used herein refers to an antibody of a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible antibodies possessing naturally occurring mutations that may be present in minor amounts, and the term “polyclonal antibody” as used herein refers to an antibody obtained from a population of heterogeneous antibodies. The term “monoclonal” indicates the character of the antibody as being a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method (e.g., by hybridoma, phage selection, recombinant expression, transgenic animals, etc.). The term includes whole immunoglobulins as well as the fragments etc. described above under the definition of “antibody.” Methods of making monoclonal antibodies are known in the art. One method which may be employed is the method of Kohler, G. et al. (1975) “Continuous Cultures Of Fused Cells Secreting Antibody Of Predefined Specificity,” Nature 256:495-497 or a modification thereof. Typically, monoclonal antibodies are developed in mice, rats or rabbits. The antibodies are produced by immunizing an animal with an immunogenic amount of cells, cell extracts, or protein preparations that contain the desired epitope. The immunogen can be, but is not limited to, primary cells, cultured cell lines, cancerous cells, proteins, peptides, nucleic acids, or tissue. Cells used for immunization may be cultured for a period of time (e.g., at least 24 hours) prior to their use as an immunogen. Cells may be used as immunogens by themselves or in combination with a non-denaturing adjuvant, such as Ribi (see, e.g., Jennings, V. M. (1995) “Review of Selected Adjuvants Used in Antibody Production,” ILAR J. 37(3):119-125). In general, cells should be kept intact and preferably viable when used as immunogens. Intact cells may allow antigens to be better detected than ruptured cells by the immunized animal. Use of denaturing or harsh adjuvants, e.g., Freud's adjuvant, may rupture cells and therefore is discouraged. The immunogen may be administered multiple times at periodic intervals such as, bi-weekly, or weekly, or may be administered in such a way as to maintain viability in the animal (e.g., in a tissue recombinant). Alternatively, existing monoclonal antibodies and any other equivalent antibodies that are immunospecific for a desired pathogenic epitope can be sequenced and produced recombinantly by any means known in the art. In one embodiment, such an antibody is sequenced and the polynucleotide sequence is then cloned into a vector for expression or propagation. The sequence encoding the antibody of interest may be maintained in a vector in a host cell and the host cell can then be expanded and frozen for future use. The polynucleotide sequence of such antibodies may be used for genetic manipulation to generate the monospecific or multispecific (e.g., bispecific, trispecific and tetraspecific) molecules of the invention as well as an affinity optimized, a chimeric antibody, a humanized antibody, and/or a caninized antibody, to improve the affinity, or other characteristics of the antibody. The term “scFv” refers to single-chain Variable Domain fragments. scFv molecules are made by linking Light and/or Heavy Chain Variable Domain using a short linking peptide. Bird et al. (1988) (“Single-Chain Antigen-Binding Proteins,” Science 242:423-426) describes example of linking peptides which bridge approximately 3.5 nm between the carboxy terminus of one Variable Domain and the amino terminus of the other Variable Domain. Linkers of other sequences have been designed and used (Bird et al. (1988) “Single-Chain Antigen-Binding Proteins,” Science 242:423-426). Linkers can in turn be modified for additional functions, such as attachment of drugs or attachment to solid supports. The single-chain variants can be produced either recombinantly or synthetically. For synthetic production of scFv, an automated synthesizer can be used. For recombinant production of scFv, a suitable plasmid containing polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such asE. coli. Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides. The resultant scFv can be isolated using standard protein purification techniques known in the art. The invention particularly encompasses humanized variants of the anti-LAG-3 antibodies of the invention and multispecific binding molecules comprising the same. The term “humanized” antibody refers to a chimeric molecule, generally prepared using recombinant techniques, having an epitope-binding site of an immunoglobulin from a non-human species and a remaining immunoglobulin structure of the molecule that is based upon the structure and/or sequence of a human immunoglobulin. The epitope-binding site may comprise either complete variable domains fused onto constant domains or only the CDRs grafted onto appropriate framework regions in the variable domains. Epitope-binding sites may be wild-type or modified by one or more amino acid substitutions. This eliminates the constant region as an immunogen in human individuals, but the possibility of an immune response to the foreign variable region remains (LoBuglio, A. F. et al. (1989) “Mouse/Human Chimeric Monoclonal Antibody In Man: Kinetics And Immune Response,” Proc. Natl. Acad. Sci. (U.S.A.) 86:4220-4224). Another approach focuses not only on providing human-derived constant regions, but modifying the variable regions as well so as to reshape them as closely as possible to human form. It is known that the variable regions of both heavy and light chains contain three CDRs which vary in response to the antigens in question and determine binding capability, flanked by four framework regions (FRs) which are relatively conserved in a given species and which putatively provide a scaffolding for the CDRs. When non-human antibodies are prepared with respect to a particular antigen, the variable regions can be “reshaped” or “humanized” by grafting CDRs derived from a non-human antibody on the FRs present in the human antibody to be modified. Application of this approach to various antibodies has been reported by Sato, K. et al. (1993) “Reshaping A Human Antibody To Inhibit The Interleukin6-Dependent Tumor Cell Growth,” Cancer Res 53:851-856. Riechmann, L. et al. (1988) “Reshaping Human Antibodies for Therapy,” Nature 332:323-327; Verhoeyen, M. et al. (1988) “Reshaping Human Antibodies: Grafting An Antilysozyme Activity,” Science 239:1534-1536; Kettleborough, C. A. et al. (1991) “Humanization Of A Mouse Monoclonal Antibody By CDR-Grafting: The Importance Of Framework Residues On Loop Conformation,” Protein Engineering 4:773-3783; Maeda, H. et al. (1991) “Construction Of Reshaped Human Antibodies With HIV-Neutralizing Activity,” Human Antibodies Hybridoma 2:124-134; Gorman, S. D. et al. (1991) “Reshaping A Therapeutic CD4Antibody,” Proc. Natl. Acad. Sci. (U.S.A.) 88:4181-4185; Tempest, P. R. et al. (1991) “Reshaping A Human Monoclonal Antibody To Inhibit Human Respiratory Syncytial Virus Infection in vivo,” Bio/Technology 9:266-271; Co, M. S. et al. (1991) “Humanized Antibodies For Antiviral Therapy,” Proc. Natl. Acad. Sci. (U.S.A.) 88:2869-2873; Carter, P. et al. (1992) “Humanization Of An Anti-p185her2Antibody For Human Cancer Therapy,” Proc. Natl. Acad. Sci. (U.S.A.) 89:4285-4289; and Co, M. S. et al. (1992) “Chimeric And Humanized Antibodies With Specificity For The CD33Antigen,” J. Immunol. 148:1149-1154. In some embodiments, humanized antibodies preserve all CDR sequences (for example, a humanized mouse antibody which contains all six CDRs from the mouse antibodies). In other embodiments, humanized antibodies have one or more CDRs (one, two, three, four, five or six) that are altered in their amino acid sequence(s) relative to the original antibody, which are also termed one or more CDRs “derived from” one or more CDRs from the original antibody (i.e., derived from such CDRs, derived from knowledge of the amino acid sequences of such CDRs, etc.). A polynucleotide sequence that encodes the variable domain of an antibody may be used to generate such derivatives and to improve the affinity, or other characteristics of such antibodies. The general principle in humanizing an antibody involves retaining the basic sequence of the epitope-binding portion of the antibody, while swapping the non-human remainder of the antibody with human antibody sequences. There are four general steps to humanize a monoclonal antibody. These are: (1) determining the nucleotide and predicted amino acid sequence of the starting antibody light and heavy variable domains (2) designing the humanized antibody or caninized antibody, i.e., deciding which antibody framework region to use during the humanizing or canonizing process (3) the actual humanizing or caninizing methodologies/techniques and (4) the transfection and expression of the humanized antibody. See, for example, U.S. Pat. Nos. 4,816,567; 5,807,715; 5,866,692; and 6,331,415. The epitope-binding site of the molecules of the present invention may comprise a complete Variable Domain fused to a Constant Domain or only the complementarity determining regions (CDRs) of such Variable Domain grafted to appropriate framework regions. Epitope-binding sites may be wild-type or modified by one or more amino acid substitutions. This eliminates the constant region as an immunogen in human individuals, but the possibility of an immune response to the foreign variable domain remains (LoBuglio, A. F. et al. (1989) “Mouse/Human Chimeric Monoclonal Antibody In Man: Kinetics And Immune Response,” Proc. Natl. Acad. Sci. (U.S.A.) 86:4220-4224). Another approach focuses not only on providing human-derived constant regions, but modifying the variable domains as well so as to reshape them as closely as possible to human form. It is known that the variable domains of both heavy and light chains contain three complementarity determining regions (CDRs) which vary in response to the antigens in question and determine binding capability, flanked by four framework regions (FRs) which are relatively conserved in a given species and which putatively provide a scaffolding for the CDRs. When non-human antibodies are prepared with respect to a particular antigen, the variable domains can be “reshaped” or “humanized” by grafting CDRs derived from non-human antibody on the FRs present in the human antibody to be modified. Application of this approach to various antibodies has been reported by Sato, K. et al. (1993) Cancer Res 53:851-856. Riechmann, L. et al. (1988) “Reshaping Human Antibodies for Therapy,” Nature 332:323-327; Verhoeyen, M. et al. (1988) “Reshaping Human Antibodies: Grafting An Antilysozyme Activity,” Science 239:1534-1536; Kettleborough, C. A. et al. (1991) “Humanization Of A Mouse Monoclonal Antibody By CDR-Grafting: The Importance Of Framework Residues On Loop Conformation,” Protein Engineering 4:773-3783; Maeda, H. et al. (1991) “Construction Of Reshaped Human Antibodies With HIV-Neutralizing Activity,” Human Antibodies Hybridoma 2:124-134; Gorman, S. D. et al. (1991) “Reshaping A Therapeutic CD4Antibody,” Proc. Natl. Acad. Sci. (U.S.A.) 88:4181-4185; Tempest, P. R. et al. (1991) “Reshaping A Human Monoclonal Antibody To Inhibit Human Respiratory Syncytial Virus Infection in vivo,” Bio/Technology 9:266-271; Co, M. S. et al. (1991) “Humanized Antibodies For Antiviral Therapy,” Proc. Natl. Acad. Sci. (U.S.A.) 88:2869-2873; Carter, P. et al. (1992) “Humanization Of An Anti-p185her2 Antibody For Human Cancer Therapy,” Proc. Natl. Acad. Sci. (U.S.A.) 89:4285-4289; and Co, M. S. et al. (1992) “Chimeric And Humanized Antibodies With Specificity For The CD33Antigen,” J. Immunol. 148:1149-1154. In some embodiments, humanized antibodies preserve all CDR sequences (for example, a humanized mouse antibody which contains all six CDRs from the mouse antibodies). In other embodiments, humanized antibodies have one or more CDRs (one, two, three, four, five, or six) which differ in sequence relative to the original antibody. A number of “humanized” antibody molecules comprising an epitope-binding site derived from a non-human immunoglobulin have been described, including chimeric antibodies having rodent or modified rodent Variable Domain and their associated complementarity determining regions (CDRs) fused to human Constant Domains (see, for example, Winter et al. (1991) “Man-made Antibodies,” Nature 349:293-299; Lobuglio et al. (1989) “Mouse/Human Chimeric Monoclonal Antibody In Man: Kinetics And Immune Response,” Proc. Natl. Acad. Sci. (U.S.A.) 86:4220-4224 (1989), Shaw et al. (1987) “Characterization Of A Mouse/Human Chimeric Monoclonal Antibody(17-1A)To A Colon Cancer Tumor-Associated Antigen,” J. Immunol. 138:4534-4538, and Brown et al. (1987) “Tumor-Specific Genetically Engineered Murine/Human Chimeric Monoclonal Antibody,” Cancer Res. 47:3577-3583). Other references describe rodent CDRs grafted into a human supporting framework region (FR) prior to fusion with an appropriate human antibody Constant Domain (see, for example, Riechmann, L. et al. (1988) “Reshaping Human Antibodies for Therapy,” Nature 332:323-327; Verhoeyen, M. et al. (1988) “Reshaping Human Antibodies: Grafting An Antilysozyme Activity,” Science 239:1534-1536; and Jones et al. (1986) “Replacing The Complementarity-Determining Regions In A Human Antibody With Those From A Mouse,” Nature 321:522-525). Another reference describes rodent CDRs supported by recombinantly veneered rodent framework regions. See, for example, European Patent Publication No. 519,596. These “humanized” molecules are designed to minimize unwanted immunological response towards rodent anti-human antibody molecules, which limits the duration and effectiveness of therapeutic applications of those moieties in human recipients. Other methods of humanizing antibodies that may also be utilized are disclosed by Daugherty et al. (1991) “Polymerase Chain Reaction Facilitates The Cloning, CDR-Grafting, And Rapid Expression Of A Murine Monoclonal Antibody Directed Against The CD18Component Of Leukocyte Integrins,” Nucl. Acids Res. 19:2471-2476 and in U.S. Pat. Nos. 6,180,377; 6,054,297; 5,997,867; and 5,866,692. II. Fcγ Receptors (FcγRs) The CH2 and CH3 Domains of the two heavy chains interact to form the Fc Region, which is a domain that is recognized by cellular Fc Receptors including but not limited to Fc gamma Receptors (FcγRs). As used herein, the term “Fc Region” is used to define a C-terminal region of an IgG heavy chain. The amino acid sequence of the CH2-CH3 Domain of an exemplary human IgG1 is (SEQ ID NO:1): 231      240          250          260          270          280APELLGGPSV   FLFPPKPKDT   LMISRTPEVT   CVVVDVSHED   PEVKFNWYVD290          300          310          320          330GVEVHNAKTK   PREEQYNSTY   RVVSVLTVLH   QDWLNGKEYK   CKVSNKALPA340          350          360          370          380PIEKTISKAK   GQPREPQVYT   LPPSREEMTK   NQVSLTCLVK   GFYPSDIAVE390          400          410          420          430WESNGQPENN   YKTTPPVLDS   DGSFFLYSKL   TVDKSRWQQG   NVFSCSVMHE440       447ALHNHYTQKS   LSLSPGX as numbered by the EU index according to Kabat, wherein, X is a lysine (K) or is absent. The amino acid sequence of the CH2-CH3 Domain of an exemplary human IgG2 is (SEQ ID NO:2): 231      240          250          260          270          280APPVA-GPSV   FLFPPKPKDT   LMISRTPEVT   CVVVDVSHED   PEVQFNWYVD290          300          310          320          330GVEVHNAKTK   PREEQFNSTF   RVVSVLTVVH   QDWLNGKEYK   CKVSNKGLPA340          350          360          370          380PIEKTISKTK   GQPREPQVYT   LPPSREEMTK   NQVSLTCLVK   GFYPSDISVE390          400          410          420          430WESNGQPENN   YKTTPPMLDS   DGSFFLYSKL   TVDKSRWQQG   NVFSCSVMHE440       447ALHNHYTQKS   LSLSPGX as numbered by the EU index according to Kabat, wherein, X is a lysine (K) or is absent. The amino acid sequence of the CH2-CH3 Domain of an exemplary human IgG3 is (SEQ ID NO:3): 231      240          250          260          270          280APELLGGPSV   FLFPPKPKDT   LMISRTPEVT   CVVVDVSHED   PEVQFKWYVD290          300          310          320          330GVEVHNAKTK   PREEQYNSTF   RVVSVLTVLH   QDWLNGKEYK   CKVSNKALPA340          350          360          370          380PIEKTISKTK   GQPREPQVYT   LPPSREEMTK   NQVSLTCLVK   GFYPSDIAVE390          400          410          420          430WESSGQPENN   YNTTPPMLDS   DGSFFLYSKL   TVDKSRWQQG   NIFSCSVMHE440       447ALHNRFTQKS   LSLSPGX as numbered by the EU index according to Kabat, wherein, X is a lysine (K) or is absent. The amino acid sequence of the CH2-CH3 Domain of an exemplary human IgG4 is (SEQ ID NO:4): 231      240          250          260          270          280APEFLGGPSV   FLFPPKPKDT   LMISRTPEVT   CVVVDVSQED   PEVQFNWYVD290          300          310          320          330GVEVHNAKTK   PREEQFNSTY   RVVSVLTVLH   QDWINGKEYK   CKVSNKGLPS340          350          360          370          380SIEKTISKAK   GQPREPQVYT   LPPSQEEMTK   NQVSLTCLVK   GFYPSDIAVE390          400          410          420          430WESNGQPENN   YKTTPPVLDS   DGSFFLYSRL   TVDKSRWQEG   NVFSCSVMHE440       447ALHNHYTQKS   LSLSLGX as numbered by the EU index according to Kabat, wherein, X is a lysine (K) or is absent. Throughout the present specification, the numbering of the residues in the constant region of an IgG heavy chain is that of the EU index according to Kabat et al., SEQUENCES OFPROTEINS OFIMMUNOLOGICALINTEREST, 5th Ed. Public Health Service, NH1, MD (1991) (“Kabat”), expressly incorporated herein by references. The “EU index according to Kabat” refers to the numbering of the human IgG1 EU antibody. Amino acids from the Variable Domains of the mature heavy and light chains of immunoglobulins are designated by the position of an amino acid in the chain, and the CDRs are identified as defined by Kabat (it will be understood that CDRH1 as defined by Chothia, C. & Lesk, A. M. ((1987) “Canonical structures for the hypervariable regions of immunoglobulins,”. J. Mol. Biol. 196:901-917) begins five residues earlier). Kabat described numerous amino acid sequences for antibodies, identified an amino acid consensus sequence for each subgroup, and assigned a residue number to each amino acid. Kabat's numbering scheme is extendible to antibodies not included in his compendium by aligning the antibody in question with one of the consensus sequences in Kabat by reference to conserved amino acids. This method for assigning residue numbers has become standard in the field and readily identifies amino acids at equivalent positions in different antibodies, including chimeric or humanized variants. For example, an amino acid at position 50 of a human antibody light chain occupies the equivalent position to an amino acid at position 50 of a mouse antibody light chain. Polymorphisms have been observed at a number of different positions within antibody constant regions (e.g., Fc positions, including but not limited to positions 270, 272, 312, 315, 356, and 358 as numbered by the EU index according to Kabat), and thus slight differences between the presented sequence and sequences in the prior art can exist. Polymorphic forms of human immunoglobulins have been well-characterized. At present, 18 Gm allotypes are known: G1m (1, 2, 3, 17) or G1m (a, x, f, z), G2m (23) or G2m (n), G3m (5, 6, 10, 11, 13, 14, 15, 16, 21, 24, 26, 27, 28) or G3m (b1, c3, b3, b0, b3, b4, s, t, g1, c5, u, v, g5) (Lefranc, et al., “The Human IgG Subclasses: Molecular Analysis Of Structure, Function And Regulation.” Pergamon, Oxford, pp. 43-78 (1990); Lefranc, G. et al., 1979, Hum. Genet.: 50, 199-211). It is specifically contemplated that the antibodies of the present invention may be incorporate any allotype, isoallotype, or haplotype of any immunoglobulin gene, and are not limited to the allotype, isoallotype or haplotype of the sequences provided herein. Furthermore, in some expression systems the C-terminal amino acid residue (bolded above) of the CH3 Domain may be post-translationally removed. Accordingly, the C-terminal residue of the CH3 Domain is an optional amino acid residue in the LAG-3-binding molecules of the invention. Specifically encompassed by the instant invention are LAG-3-binding molecules lacking the C-terminal residue of the CH3 Domain. Also specifically encompassed by the instant invention are such constructs comprising the C-terminal lysine residue of the CH3 Domain. Activating and inhibitory signals are transduced through the ligation of an Fc Region to a cellular Fc Receptor (FcγR). The ability of such ligation to result in diametrically opposing functions results from structural differences among the different receptor isoforms. Two distinct domains within the cytoplasmic signaling domains of the receptor called immunoreceptor tyrosine-based activation motifs (ITAMs) and immunoreceptor tyrosine-based inhibitory motifs (ITIMS) account for the different responses. The recruitment of different cytoplasmic enzymes to these structures dictates the outcome of the FcγR-mediated cellular responses. ITAM-containing FcγR complexes include FcγRI, FcγRIIA, FcγRIIIA, whereas ITIM-containing complexes only include FcγRIIB. Human neutrophils express the FcγRIIA gene. FcγRIIA clustering via immune complexes or specific antibody cross-linking serves to aggregate ITAMs along with receptor-associated kinases which facilitate ITAM phosphorylation. ITAM phosphorylation serves as a docking site for Syk kinase, activation of which results in activation of downstream substrates (e.g., PI3K). Cellular activation leads to release of proinflammatory mediators. The FcγRIIB gene is expressed on B lymphocytes; its extracellular domain is 96% identical to FcγRIIA and binds IgG complexes in an indistinguishable manner. The presence of an ITIM in the cytoplasmic domain of FcγRIIB defines this inhibitory subclass of FcγR. Recently the molecular basis of this inhibition was established. When co-ligated along with an activating FcγR, the ITIM in FcγRIIB becomes phosphorylated and attracts the SH2 domain of the inositol polyphosphate 5′-phosphatase (SHIP), which hydrolyzes phosphoinositol messengers released as a consequence of ITAM-containing FcγR-mediated tyrosine kinase activation, consequently preventing the influx of intracellular Ca++. Thus cross-linking of FcγRIIB dampens the activating response to FcγR ligation and inhibits cellular responsiveness. B-cell activation, B-cell proliferation and antibody secretion is thus aborted. III. Bispecific Antibodies, Multispecific Diabodies and DART® Diabodies The functionality of antibodies can be enhanced by generating multispecific antibody-based molecules that can simultaneously bind two separate and distinct antigens (or different epitopes of the same antigen) and/or by generating antibody-based molecule having higher valency (i.e., more than two binding sites) for the same epitope and/or antigen. In order to provide molecules having greater capability than natural antibodies, a wide variety of recombinant bispecific antibody formats have been developed (see, e.g., PCT Publication Nos. WO 2008/003116, WO 2009/132876, WO 2008/003103, WO 2007/146968, WO 2009/018386, WO 2012/009544, WO 2013/070565), most of which use linker peptides either to fuse a further epitope-binding fragment (e.g., an scFv, VL, VH, etc.) to, or within the antibody core (IgA, IgD, IgE, IgG or IgM), or to fuse multiple epitope-binding fragments (e.g., two Fab fragments or scFvs). Alternative formats use linker peptides to fuse an epitope-binding fragment (e.g., an scFv, VL, VH, etc.) to a dimerization domain such as the CH2-CH3 Domain or alternative polypeptides (WO 2005/070966, WO 2006/107786A WO 2006/107617A, WO 2007/046893). Typically, such approaches involve compromises and trade-offs. For example, PCT Publications Nos. WO 2013/174873, WO 2011/133886 and WO 2010/136172 disclose that the use of linkers may cause problems in therapeutic settings, and teaches a trispecific antibody in which the CL and CH1 Domains are switched from their respective natural positions and the VL and VH Domains have been diversified (WO 2008/027236; WO 2010/108127) to allow them to bind to more than one antigen. Thus, the molecules disclosed in these documents trade binding specificity for the ability to bind additional antigen species. PCT Publications Nos. WO 2013/163427 and WO 2013/119903 disclose modifying the CH2 Domain to contain a fusion protein adduct comprising a binding domain. The document notes that the CH2 Domain likely plays only a minimal role in mediating effector function. PCT Publications Nos. WO 2010/028797, WO2010028796 and WO 2010/028795 disclose recombinant antibodies whose Fc Regions have been replaced with additional VL and VH Domains, so as to form trivalent binding molecules. PCT Publications Nos. WO 2003/025018 and WO2003012069 disclose recombinant diabodies whose individual chains contain scFv Domains. PCT Publications No. WO 2013/006544 discloses multivalent Fab molecules that are synthesized as a single polypeptide chain and then subjected to proteolysis to yield heterodimeric structures. Thus, the molecules disclosed in these documents trade all or some of the capability of mediating effector function for the ability to bind additional antigen species. PCT Publications Nos. WO 2014/022540, WO 2013/003652, WO 2012/162583, WO 2012/156430, WO 2011/086091, WO 2008/024188, WO 2007/024715, WO 2007/075270, WO 1998/002463, WO 1992/022583 and WO 1991/003493 disclose adding additional binding domains or functional groups to an antibody or an antibody portion (e.g., adding a diabody to the antibody's light chain, or adding additional VL and VH Domains to the antibody's light and heavy chains, or adding a heterologous fusion protein or chaining multiple Fab Domains to one another). Thus, the molecules disclosed in these documents trade native antibody structure for the ability to bind additional antigen species. The art has additionally noted the capability to produce diabodies that differ from such natural antibodies in being capable of binding two or more different epitope species (i.e., exhibiting bispecificity or multispecificity in addition to bivalency or multivalency) (see, e.g., Holliger et al. (1993) “‘Diabodies’: Small Bivalent And Bispecific Antibody Fragments,” Proc. Natl. Acad. Sci. (U.S.A.) 90:6444-6448; US 2004/0058400 (Hollinger et al.); US 2004/0220388/WO 02/02781 (Mertens et al.); Alt et al. (1999) FEBS Lett. 454(1-2):90-94; Lu, D. et al. (2005) “A Fully Human Recombinant IgG-Like Bispecific Antibody To Both The Epidermal Growth Factor Receptor And The Insulin-Like Growth Factor Receptor For Enhanced Antitumor Activity,” J. Biol. Chem. 280(20):19665-19672; WO 02/02781 (Mertens et al.); Olafsen, T. et al. (2004) “Covalent Disulfide-Linked Anti-CEA Diabody Allows Site-Specific Conjugation And Radiolabeling For Tumor Targeting Applications,” Protein Eng. Des. Sel. 17(1):21-27; Wu, A. et al. (2001) “Multimerization Of A Chimeric Anti-CD20Single Chain Fv-Fv Fusion Protein Is Mediated Through Variable Domain Exchange,” Protein Engineering 14(2):1025-1033; Asano et al. (2004) “A Diabody For Cancer Immunotherapy And Its Functional Enhancement By Fusion Of Human Fc Domain,” Abstract 3P-683, J. Biochem. 76(8):992; Takemura, S. et al. (2000) “Construction Of A Diabody(Small Recombinant Bispecific Antibody)Using A Refolding System,” Protein Eng. 13(8):583-588; Baeuerle, P. A. et al. (2009) “Bispecific T-Cell Engaging Antibodies For Cancer Therapy,” Cancer Res. 69(12):4941-4944). The design of a diabody is based on the antibody derivative known as a single-chain Variable Domain fragment (scFv). Such molecules are made by linking Light and/or Heavy chain Variable Domains by using a short linking peptide. Linkers can in turn be modified for additional functions, such as attachment of drugs or attachment to solid supports. The single-chain variants can be produced either recombinantly or synthetically. For synthetic production of scFv, an automated synthesizer can be used. For recombinant production of scFv, a suitable plasmid containing polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such asE. coli. Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides. The resultant scFv can be isolated using standard protein purification techniques known in the art. The provision of non-monospecific diabodies provides a significant advantage over antibodies, including but not limited to, the capacity to co-ligate and co-localize cells that express different epitopes. Bispecific diabodies thus have wide-ranging applications including therapy and immunodiagnosis. Bispecificity allows for great flexibility in the design and engineering of the diabody in various applications, providing enhanced avidity to multimeric antigens, the cross-linking of differing antigens, and directed targeting to specific cell types relying on the presence of both target antigens. Due to their increased valency, low dissociation rates and rapid clearance from the circulation (for diabodies of small size, at or below ˜50 kDa), diabody molecules known in the art have also shown particular use in the field of tumor imaging (Fitzgerald et al. (1997) “Improved Tumour Targeting By Disulphide Stabilized Diabodies Expressed In Pichia pastoris,” Protein Eng. 10:1221). The bispecificity of diabodies has led to their use for co-ligating differing cells, for example, the cross-linking of cytotoxic T-cells to tumor cells (Staerz et al. (1985) “Hybrid Antibodies Can Target Sites For Attack By T Cells,” Nature 314:628-631, and Holliger et al. (1996) “Specific Killing Of Lymphoma Cells By Cytotoxic T-Cells Mediated By A Bispecific Diabody,” Protein Eng. 9:299-305; Marvin et al. (2005) “Recombinant Approaches To IgG-Like Bispecific Antibodies,” Acta Pharmacol. Sin. 26:649-658). Alternatively, or additionally, bispecific diabodies can be used to co-ligate receptors on the surface of different cells or on a single cell. Co-ligation of different cells and/or receptors is useful to modulation effector functions and/or immune cell signaling. Multispecific molecules (e.g., bispecific diabodies) comprising epitope-binding sites may be directed to a surface determinant of any immune cell such as B7-H3 (CD276), B7-H4 (VTCN1), BTLA (CD272), CD3, CD8, CD16, CD27, CD32, CD40, CD40L, CD47, CD64, CD70 (CD27L), CD80 (B7-1), CD86 (B7-2), CD94 (KLRD1), CD137 (4-1BB), CD137L (4-1BBL), CD226, CTLA-4 (CD152), Galectin-9, GITR, GITRL, HHLA2, ICOS (CD278), ICOSL (CD275), Killer Activation Receptor (KIR), LAG-3 (CD223), LIGHT (TNFSF14, CD258), MHC class I or II, NKG2a, NKG2d, OX40 (CD134), OX40L (CD134L), PD1H, PD-1 (CD279), PD-L1 (B7-H1, CD274), PD-L2 (B7-CD, CD273), PVR (NECL5, CD155), SIRPa, TCR, TIGIT, TIM-3 (HAVCR2), and/or VISTA (PD-1H), which are expressed on T lymphocytes, Natural Killer (NK) cells, Antigen-presenting cells or other mononuclear cell. In particular, epitope-binding sites directed to a cell surface receptor that is involved in regulating an immune checkpoint (or the ligand thereof) are useful in the generation of bispecific or multispecific binding molecules which antagonize or block the inhibitory signaling of immune checkpoint molecules and thereby stimulate, upregulate or enhance, immune responses in a subject. Molecules involved in regulating immune checkpoints include, but are not limited to B7-H3, B7-H4, BTLA, CD40, CD40L, CD47, CD70, CD80, CD86, CD94, CD137, CD137L, CD226, CTLA-4, Galectin-9, GITR, GITRL, HHLA2, ICOS, ICOSL, KIR, LAG-3, LIGHT, MHC class I or II, NKG2a, NKG2d, OX40, OX40L, PD1H, PD-1, PD-L1, PD-L2, PVR, SIRPa, TCR, TIGIT, TIM-3 and/or VISTA. However, the above advantages come at a salient cost. The formation of such non-monospecific diabodies requires the successful assembly of two or more distinct and different polypeptides (i.e., such formation requires that the diabodies be formed through the heterodimerization of different polypeptide chain species). This fact is in contrast to monospecific diabodies, which are formed through the homodimerization of identical polypeptide chains. Because at least two dissimilar polypeptides (i.e., two polypeptide species) must be provided in order to form a non-monospecific diabody, and because homodimerization of such polypeptides leads to inactive molecules (Takemura, S. et al. (2000) “Construction Of A Diabody(Small Recombinant Bispecific Antibody)Using A Refolding System,” Protein Eng. 13(8):583-588), the production of such polypeptides must be accomplished in such a way as to prevent covalent bonding between polypeptides of the same species (i.e., so as to prevent homodimerization) (Takemura, S. et al. (2000) “Construction Of A Diabody(Small Recombinant Bispecific Antibody)Using A Refolding System,” Protein Eng. 13(8):583-588). The art has therefore taught the non-covalent association of such polypeptides (see, e.g., Olafsen et al. (2004) “Covalent Disulfide-Linked Anti-CEA Diabody Allows Site-Specific Conjugation And Radiolabeling For Tumor Targeting Applications,” Prot. Engr. Des. Sel. 17:21-27; Asano et al. (2004) “A Diabody For Cancer Immunotherapy And Its Functional Enhancement By Fusion Of Human Fc Domain,” Abstract 3P-683, J. Biochem. 76(8):992; Takemura, S. et al. (2000) “Construction Of A Diabody(Small Recombinant Bispecific Antibody)Using A Refolding System,” Protein Eng. 13(8):583-588; Lu, D. et al. (2005) “A Fully Human Recombinant IgG-Like Bispecific Antibody To Both The Epidermal Growth Factor Receptor And The Insulin-Like Growth Factor Receptor For Enhanced Antitumor Activity,” J. Biol. Chem. 280(20):19665-19672). However, the art has recognized that bispecific diabodies composed of non-covalently associated polypeptides are unstable and readily dissociate into non-functional monomers (see, e.g., Lu, D. et al. (2005) “A Fully Human Recombinant IgG-Like Bispecific Antibody To Both The Epidermal Growth Factor Receptor And The Insulin-Like Growth Factor Receptor For Enhanced Antitumor Activity,” J. Biol. Chem. 280(20):19665-19672). In the face of this challenge, the art has succeeded in developing stable, covalently bonded heterodimeric non-monospecific diabodies, termed DART® (Dual Affinity Re-Targeting Reagents) diabodies; see, e.g., United States Patent Publications No. 2013-0295121; 2010-0174053 and 2009-0060910; European Patent Publication No. EP 2714079; EP 2601216; EP 2376109; EP 2158221 and PCT Publications No. WO 2012/162068; WO 2012/018687; WO 2010/080538; WO 2008/157379; WO 2006/113665 and Sloan, D. D. et al. (2015) “Targeting HIV Reservoir in Infected CD4T Cells by Dual-Affinity Re-targeting Molecules(DARTs)that Bind HIV Envelope and Recruit Cytotoxic T Cells,” PLoS Pathog. 11(11):e1005233. doi: 10.1371/journal.ppat.1005233; Al Hussaini, M. et al. (2015) “Targeting CD123In AML Using A T-Cell Directed Dual-Affinity Re-Targeting(DARTR)Platform,” Blood 127(1):122-131; Chichili, G. R. et al. (2015) “A CD3xCD123Bispecific DART For Redirecting Host T Cells To Myelogenous Leukemia: Preclinical Activity And Safety In Nonhuman Primates,” Sci. Transl. Med. 7(289):289ra82; Moore, P. A. et al. (2011) “Application Of Dual Affinity Retargeting Molecules To Achieve Optimal Redirected T-Cell Killing Of B-Cell Lymphoma,” Blood 117(17):4542-4551; Veri, M. C. et al. (2010) “Therapeutic Control Of B Cell Activation Via Recruitment Of Fcgamma Receptor IIb(CD32B)Inhibitory Function With A Novel Bispecific Antibody Scaffold,” Arthritis Rheum. 62(7):1933-1943; Johnson, S. et al. (2010) “Effector Cell Recruitment With Novel Fv-Based Dual-Affinity Re-Targeting Protein Leads To Potent Tumor Cytolysis And in vivo B-Cell Depletion,” J. Mol. Biol. 399(3):436-449; Marvin, J. S. et al. (2005) “Recombinant Approaches To IgG-Like Bispecific Antibodies,” Acta Pharmacol. Sin. 26:649-658; Olafsen, T. et al. (2004) “Covalent Disulfide-Linked Anti-CEA Diabody Allows Site-Specific Conjugation And Radiolabeling For Tumor Targeting Applications,” Prot. Engr. Des. Sel. 17:21-27; Holliger, P. et al. (1993) “‘Diabodies’: Small Bivalent And Bispecific Antibody Fragments,” Proc. Natl. Acad. Sci. (U.S.A.) 90:6444-6448. Such diabodies comprise two or more covalently complexed polypeptides and involve engineering one or more cysteine residues into each of the employed polypeptide species that permit disulfide bonds to form and thereby covalently bond two polypeptide chains. For example, the addition of a cysteine residue to the C-terminus of such constructs has been shown to allow disulfide bonding between the polypeptide chains, stabilizing the resulting heterodimer without interfering with the binding characteristics of the bivalent molecule. Each of the two polypeptides of the simplest bispecific DART® diabody comprises three domains. The first polypeptide comprises (in the N-terminal to C-terminal direction): (i) a First Domain that comprises a binding region of a Light Chain Variable Domain of a first immunoglobulin (VL1), (ii) a Second Domain that comprises a binding region of a Heavy Chain Variable Domain of a second immunoglobulin (VH2), and (iii) a Third Domain that contains a cysteine residue (or a cysteine-containing domain) and a Heterodimer-Promoting Domain that serves to promote heterodimerization with the second polypeptide of the diabody and to covalently bond the diabody's first and second polypeptides to one another. The second polypeptide contains (in the N-terminal to C-terminal direction): (i) a First Domain that comprises a binding region of a Light Chain Variable Domain of the second immunoglobulin (VL2), (ii) a Second Domain that comprises a binding region of a Heavy Chain Variable Domain of the first immunoglobulin (VH1), and (iii) a Third Domain that contains a cysteine residue (or a cysteine-containing domain) and a complementary Heterodimer-Promoting Domain that complexes with the Heterodimer-Promoting Domain of the first polypeptide chain in order to promote heterodimerization with the first polypeptide chain. The cysteine residue (or a cysteine-containing domain) of the third domain of the second polypeptide chain serves to promote the covalent bonding of the second polypeptide chain to the first polypeptide chain of the diabody. Such molecules are stable, potent and have the ability to simultaneously bind two or more antigens. In one embodiment, the Third Domains of the first and second polypeptides each contain a cysteine residue, which serves to bind the polypeptides together via a disulfide bond.FIG.1provides a schematic of such a diabody, which utilizes E-coil/K-coil Heterodimer-Promoting domains and a cysteine containing linker for covalent bonding. As provided inFIG.2andFIGS.3A-3C, one or both of the polypeptides may additionally possesses the sequence of a CH2-CH3 Domain, such that complexing between the two diabody polypeptides forms an Fc Region that is capable of binding to the Fc receptor of cells (such as B lymphocytes, dendritic cells, natural killer cells, macrophages, neutrophils, eosinophils, basophils and mast cells). As provided in more detail below, the CH2 and/or CH3 Domains of such polypeptide chains need not be identical in sequence, and advantageously are modified to foster complexing between the two polypeptide chains. Many variations of such molecules have been described (see, e.g., United States Patent Publications No. 2015/0175697; 2014/0255407; 2014/0099318; 2013/0295121; 2010/0174053 and 2009/0060910; European Patent Publication No. EP 2714079; EP 2601216; EP 2376109; EP 2158221 and PCT Publications No. WO 2012/162068; WO 2012/018687; WO 2010/080538). These Fc Region-containing DART® diabodies may comprise two pairs of polypeptide chains. The first polypeptide chain comprises (in the N-terminal to C-terminal direction): (i) a First Domain that comprises a binding region of a Light Chain Variable Domain of a first immunoglobulin (VL), (ii) a Second Domain that comprises a binding region of a Heavy Chain Variable Domain of a second immunoglobulin (VH2), (iii) a Third Domain that contains a cysteine residue (or a cysteine-containing domain) and a serves to promote heterodimerization with the second polypeptide of the diabody and to covalently bond the diabody's first and second polypeptides to one another, and (iv) a CH2-CH3 Domain. The second polypeptide contains (in the N-terminal to C-terminal direction): (i) a First Domain that comprises a binding region of a Light Chain Variable Domain of the second immunoglobulin (VL2), (ii) a Second Domain that comprises a binding region of a Heavy Chain Variable Domain of the first immunoglobulin (VH1), and (iii)) a Third Domain that contains a cysteine residue (or a cysteine-containing domain) and a Heterodimer-Promoting Domain that promotes heterodimerization with the first polypeptide chain. Here, two first polypeptides complex with each other to form an Fc Region.FIGS.3A-3Cprovide schematics of three variations of such diabodies utilizing different Heterodimer-Promoting Domains. Other Fc-Region-containing DART® diabodies may comprise three polypeptide chains. The first polypeptide of such DART® diabodies contains three domains: (i) a VL1-containing Domain, (ii) a VH2-containing Domain and (iii) a Domain containing a CH2-CH3 sequence. The second polypeptide of such DART® diabodies contains: (i) a VL2-containing Domain, (ii) a VH-containing Domain and (iii) a Domain that promotes heterodimerization and covalent bonding with the diabody's first polypeptide chain. The third polypeptide of such DART® diabodies comprises a CH2-CH3 sequence. Thus, the first and second polypeptide chains of such DART® diabodies associate together to form a VL1/VH1 binding site that is capable of binding to the epitope, as well as a VL2/VH2 binding site that is capable of binding to the second epitope. Such more complex DART® molecules also possess cysteine-containing domains which function to form a covalently bonded complex. Thus, the first and second polypeptides are bonded to one another through a disulfide bond involving cysteine residues in their respective Third Domains. Notably, the first and third polypeptide chains complex with one another to form an Fc Region that is stabilized via a disulfide bond.FIGS.4A-4Bprovide schematics of such diabodies comprising three polypeptide chains. Still other Fc-Region-containing DART® diabodies may comprise five polypeptide chains which may comprise the binding regions from the Light and Heavy Chain Variable Domains of up to three different immunoglobulins (referred to as VL1/VH1, VL2/VH2 and VL3/VH3). For example, the first polypeptide chain of such diabodies may contain: (i) a VH1-containing domain, (ii) a CH-containing domain, and (iii) a Domain containing a CH2-CH3 sequence. The second and fifth polypeptide chains of such diabodies may contain: (i) a VL1-containing domain, and (ii) a CL-containing domain. The third polypeptide chain of such diabodies may contain: (i) a VH1-containing domain, (ii) a CH1-containing domain, (iii) a Domain containing a CH2-CH3 sequence, (iv) a VL2-containing Domain, (v) a VH3-containing Domain and (vi) a Heterodimer-Promoting Domain, where the Heterodimer-Promoting Domains promote the dimerization of the third chain with the fourth chain. The fourth polypeptide of such diabodies may contain: (i) a VL3-containing Domain, (ii) a VH2-containing Domain and (iii) a Domain that promotes heterodimerization and covalent bonding with the diabody's third polypeptide chain. Here, the first and third polypeptides complex with each other to form an Fc Region. Such more complex DART® molecules also possess cysteine-containing domains which function to form a covalently bonded complex, such that each polypeptide chain is bonded to at least one addition polypeptide chain through a disulfide bond involving cysteine residues. Preferably, such domains are ordered in the N-terminal to C-terminal direction.FIG.5provides schematics of such diabodies comprising five polypeptide chains Alternative constructs are known in the art for applications where a tetravalent molecule is desirable but an Fc is not required including, but not limited to, tetravalent tandem antibodies, also referred to as “TandAbs” (see, e.g. United States Patent Publications Nos. 2005-0079170, 2007-0031436, 2010-0099853, 2011-020667 2013-0189263; European Patent Publication Nos. EP 1078004, EP 2371866, EP 2361936 and EP 1293514; PCT Publications Nos. WO 1999/057150, WO 2003/025018, and WO 2013/013700) which are formed by the homo-dimerization of two identical chains each possessing a VH1, VL2, VH2, and VL2 Domain. Recently, trivalent structures incorporating two diabody-type binding domains and one non-diabody-type domain and an Fc Region have been described (see, e.g., PCT Application No: PCT/US15/33076, titled “Tri-Specific Binding Molecules and Methods of Use Thereof,” filed May 29, 2015; and PCT/US15/33081, titled “Tri-Specific Binding Molecules That Specifically Bind to Multiple Cancer Antigens and Methods of Use Thereof,” filed May 29, 2015). Such trivalent molecules may be utilized to generate monospecific, bispecific or trispecific molecules.FIGS.6A-6Fprovide schematics of such trivalent molecules comprising 3 or 4 polypeptide chains. IV. The LAG-3-Binding Molecules of the Present Invention The preferred LAG-3-binding molecules of the present invention include antibodies, diabodies, BiTEs, etc. and are capable of binding to a continuous or discontinuous (e.g., conformational) portion (epitope) of human LAG-3. The LAG-3-binding molecules of the present invention will preferably also exhibit the ability to bind to LAG-3 molecules of one or more non-human species, in particular, primate species (and especially a primate species, such as cynomolgus monkey). A representative human LAG-3 polypeptide (NCBI Sequence NP_002277.4; including a 22 amino acid residue signal sequence (shown underlined) and the 503 amino acid residue mature protein) has the amino acid sequence (SEQ ID NO:5): MWEAQFLGLLFLQPLWVAPVKPLQPGAEVP VVWAQEGAPAQDLSLLRRAG VTWQHQPDSG PPAAAPGHPL APGPHPAAPSVLSVGPGGLR SGRLPLQPRV QLDERGRQRG DFSLWLRPARVHLRDRALSC RLRLRLGQAS MTASPPGSLR ASDWVILNCSQLPCSPTIPL SWGPRPRRYT RADAGEYRAA FSRPDRPASVHWFRNRGQGR VPVRESPHHH LAESFLFLPQ VSPMDSGPWGVSIMYNLTVL GLEPPTPLTV YAGAGSRVGL PCRLPAGVGTPGGGPDLLVT GDNGDFTLRL EDVSQAQAGT YTCHIHLQEQITVTPKSFGS PGSLGKLLCE VTPVSGQERF VWSSLDTPSQCILTYRDGFN RSFLTAKWTP QLNATVTLAI RSFSGPWLEAQEAQLLSQPW QCOLYQGERL LGAAVYFTEL SSPGAQRSGRLLFLILGVLS LLLLVTGAFG FHLWRRQWRP RRFSALEQGIAPGALPAGHL HPPQAQSKIE ELEQEPEPEP EPEPEPEPEPEPEQL In certain embodiments the LAG-3-binding molecules of the invention are characterized by any (one or more) of the following criteria:(1) specifically binds human LAG-3 as endogenously expressed on the surface of a stimulated human T-cell;(2) specifically binds human LAG-3 with an equilibrium binding constant (KD) of 40 nM or less;(3) specifically binds human LAG-3 with an equilibrium binding constant (KD) of 0.5 nM or less;(4) specifically binds non-human primate LAG-3 (e.g., LAG-3 of cynomolgus monkey);(5) specifically binds non-human primate LAG-3 with an equilibrium binding constant (KD) of 50 nM or less;(6) specifically binds non-human primate LAG-3 with an equilibrium binding constant (KD) of 5 nM or less;(7) inhibits (i.e., blocks or interferes with) the binding of LAG-3 to MHC class II;(8) stimulates an immune response;(9) stimulates antigen specific T-cell response as a single agent;(10) synergizes with an anti-PD-1 antibody to stimulate an antigen specific T-cell response;(11) binds the same epitope of LAG-3 as the anti-LAG-3 antibody LAG-3 mAb 1 or LAG-3 mAb 6 and/or(12) does not compete with the anti-LAG-3 antibody 257F (BMS 986016, Medarex/BMS) for LAG-3 binding (e.g., as measured by Biacore Analysis). As used here the term “antigen specific T-cell response” refers to responses by a T-cell that result from stimulation of the T-cell with the antigen for which the T-cell is specific. Non-limiting examples of responses by a T-cell upon antigen specific stimulation include proliferation and cytokine production (e.g., TNF-α, IFN-γ production). The ability of a molecule to stimulate an antigen specific T-cell response may be determined, for example, using theStaphylococcus aureusEnterotoxin type B antigen (“SEB”)-stimulated PBMC assay described herein. The preferred LAG-3-binding molecules of the present invention possess the VH and/or VL Domains of murine anti-LAG-3 monoclonal antibodies “LAG-3 mAb 1,” “LAG-3 mAb 2,” “LAG-3 mAb 3,” “LAG-3 mAb 4,” “LAG-3 mAb 5,” and/or “LAG-3 mAb 6” and more preferably possess 1, 2 or all 3 of the CDRHs of the VH Domain and/or 1, 2 or all 3 of the CDRLs of the VL Domain of such anti-LAG-3 monoclonal antibodies. Such preferred LAG-3-binding molecules include bispecific (or multispecific) antibodies, chimeric or humanized antibodies, BiTes, diabodies, etc, and such binding molecules having variant Fc Regions. The invention particularly relates to LAG-3-binding molecules comprising a LAG-3 binding domain that possess:(A) (1) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 1, or hLAG-3 mAb 1 VL4;(2) the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 1;(3) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 1 and the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 1;(4) the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 1;(5) the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 1;(6) the VH and VL Domains of the anti-LAG-3 antibody LAG-3 mAb 1;(B) (1) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 2;(2) the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 2;(3) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 2 and the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 2;(4) the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 2;(5) the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 2;(6) the VH and VL Domains of the anti-LAG-3 antibody LAG-3 mAb 2;(C) (1) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 3;(2) the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 3;(3) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 3 and the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 3;(4) the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 3;(5) the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 3;(6) the VH and VL Domains of the anti-LAG-3 antibody LAG-3 mAb 3;(D) (1) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 4;(2) the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 4;(3) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 4 and the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 4;(4) the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 4;(5) the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 4;(6) the VH and VL Domains of the anti-LAG-3 antibody LAG-3 mAb 4;(E) (1) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 5;(2) the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 5;(3) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 5 and the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 5;(4) the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 5;(5) the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 5;(6) the VH and VL Domains of the anti-LAG-3 antibody LAG-3 mAb 5;(F) (1) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 6;(2) the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 6;(3) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 6 and the three CDRLs of the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 6;(4) the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 6;(5) the VL Domain of the anti-LAG-3 antibody LAG-3 mAb 6;(6) the VH and VL Domains of the anti-LAG-3 antibody LAG-3 mAb 6,(G) (1) the three CDRLs of the VL Domain of the anti-LAG-3 antibody hLAG-3 mAb 1 VL4;(2) the three CDRHs of the VH Domain of the anti-LAG-3 antibody LAG-3 mAb 1 and the three CDRs of the VL Domain of the anti-LAG-3 antibody hLAG-3 mAb 1 VL4;or that binds, or competes for binding with, the epitope that LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5 or LAG-3 mAb 6 immunospecifically binds. A. The Anti-LAG-3 Antibody LAG-3 mAb 1 1. Murine Anti-Human Antibody LAG-3 mAb 1 The amino acid sequence of the VH Domain of LAG-3 mAb 1 (SEQ ID NO:6) is shown below (CDRHresidues are shown underlined). QIQLVQSGPE LKKPGETVKI SCKASGYTFRNYGMNWVKQAPGKVLKWMGWINTYTGESTY ADDFEGRFAF SLGTSASTAYLQINILKNED TATYFCARESLYDYYSMDYW GQGTSVTVSSCDRH1 of LAG-3 mAb 1 (SEQ ID NO: 8):NYGMNCDRH2 of LAG-3 mAb 1 (SEQ ID NO: 9):WINTYTGESTYADDFEGCDRH3 of LAG-3 mAb 1 (SEQ ID NO: 10):ESLYDYYSMDY An exemplary polynucleotide that encodes the VH Domain of LAG-3 mAb 1 is SEQ ID NO:7 (nucleotides encoding the CDRHresidues are shown underlined): cagatccagt tggtgcagtc tggacctgag ctgaagaagcctggagagac agtcaagatc tcctgcaagg cttctgggtataccttcagaaactatggaatgaactgggt gaagcaggctccaggaaagg ttttaaagtg gatgggctggataaacacctacactggaga gtcaacatat gctgatgact tcgagggacggtttgccttc tctttgggaa cctctgccag cactgcctatttgcagatca acatcctcaa aaatgaggac acggctacatatttctgtgc aagagaatccctctatgatt actattctatggactactgg ggtcaaggaa cctcagtcac cgtctcctca The amino acid sequence of the VL Domain of LAG-3 mAb 1 (SEQ ID NO:11) is shown below (CDRLresidues are shown underlined): DVVVTQTPLT LSVTIGQPAS ISCKSSQSLL HSDGKTYLNWLLQRPGQSPE RLIYLVSELD SGVPDRFTGS GSGTDFTLKISRVEAEDLGV YYCWQGTHFPYTFGGGTKLE IKCDRL1 of LAG-3 mAb 1 (SEQ ID NO: 13):KSSQSLLHSDGKTYLNCDRL2 of LAG-3 mAb 1 (SEQ ID NO: 14):LVSELDSCDRL3 of LAG-3 mAb 1 (SEQ ID NO: 15):WQGTHFPYT An exemplary polynucleotide that encodes the VL Domain of LAG-3 mAb 1 is SEQ ID NO:12 (nucleotides encoding the CDRLresidues are shown underlined): gatgttgtgg tgacccagac tccactcact ttgtcggttaccattggaca accagcctcc atctcttgca agtcaagtcagagcctctta catagtgatggaaagacata tttgaattggttgttacaga ggccaggcca gtctccagag cgcctaatctatctggtgtc tgaactggac tctggagtcc ctgacaggttcactggcagt ggatcaggga cagatttcac actgaaaatcagcagagtgg aggctgagga tttgggagtt tattattgctggcaaggtac acattttccgtacacgttcg gaggggggaccaagctggaa ataaaa 2. Humanization of the Anti-LAG-3 Antibody LAG-3 mAb 1 to Form “hLAG-3 mAb 1” The above-described murine anti-LAG-3 antibody LAG-3mAb 1 was humanized in order to demonstrate the capability of humanizing an anti-LAG-3 antibody so as to decrease its antigenicity upon administration to a human recipient. The humanization yielded two humanized VH Domains, designated herein as “hLAG-3 mAb 1 VH-1,” and “hLAG-3 mAb 1 VH-2,” and four humanized VL Domains designated herein as “hLAG-3 mAb 1 VL-1,” “hLAG-3 mAb 1 VL-2,” “hLAG-3 mAb 1 VL-3,” and “hLAG-3 mAb 1 VL-4.” Any of the humanized VL Domains may be paired with the humanized VH Domains. Accordingly, any antibody comprising one of the humanized VL Domains paired with the humanized VH Domain is referred to generically as “hLAG-3 mAb 1,” and particular combinations of humanized VH/VL Domains are referred to by reference to the specific VH/VL Domains, for example a humanized antibody comprising hLAG-3 mAb 1 VH-1 and hLAG-3 mAb 1 VL-2 is specifically referred to as “hLAG-3 mAb 1(1.2).” The amino acid sequence of the VH Domain of hLAG-3 mAb 1 VH-1 (SEQ ID NO:16) is shown below (CDRHresidues are shown underlined): QVQLVQSGAE VKKPGASVKV SCKASGYTFTNYGMNWVRQAPGQGLEWMGWINTYTGESTY ADDFEGRFVF SMDTSASTAYLQISSLKAED TAVYYCARESLYDYYSMDYW GQGTTVTVSS An exemplary polynucleotide that encodes hLAG-3 mAb 1 VH-1 is SEQ ID NO:17 (nucleotides encoding the CDRHresidues are shown underlined): caggtgcaac tggttcaatc cggcgccgag gtgaaaaagcctggcgcctc cgtgaaagtg tcctgtaagg catctgggtatacgttcacaaattatggtatgaactgggt gcgacaggcaccagggcagg gactggaatg gatggggtggatcaatacttatacaggcga gagtacttat gctgacgatt tcgagggcagatttgtcttc tccatggaca ccagcgctag taccgcttatctccagatta gttctctcaa ggcggaggac acagctgtttattattgtgc ccgcgagagtttgtatgact actatagcatggattactgg ggacaaggta caaccgtgac agtgagttcc The amino acid sequence of the VH Domain of hLAG-3 mAb 1 VH-2 (SEQ ID NO:18) is shown below (CDRHresidues are shown underlined): QVQLVQSGAE VKKPGASVKV SCKASGYTFTNYGMNWVRQAPGQGLEWMGWINTYTGESTY ADDFEGRFVF SMDTSASTAYLQISSLKAED TAVYFCARESLYDYYSMDYW GQGTTVTVSS An exemplary polynucleotide that encodes hLAG-3 mAb 1 VH-2 is SEQ ID NO:19 (nucleotides encoding the CDRHresidues are shown underlined): caggtgcaac tggttcaatc cggcgccgag gtgaaaaagcctggcgcctc cgtgaaagtg tcctgtaagg catctgggtatacgttcacaaattatggtatgaactgggt gcgacaggcaccagggcagg gactggaatg gatggggtggatcaatacttatacaggcga gagtacttat gctgacgatt tcgagggcagatttgtcttc tccatggaca ccagcgctag taccgcttatctccagatta gttctctcaa ggcggaggac acagctgtttatttctgtgc ccgcgagagtttgtatgact actatagcatggattactgg ggacaaggta caaccgtgac agtgagttcc The amino acid sequence of the VL Domain of hLAG-3 mAb 1 VL-1 (SEQ ID NO:20) is shown below (CDRLresidues are shown underlined): DIVMTQTPLS LSVTPGQPAS ISCKSSQSLL HSDGKTYLNWLLQKPGQSPE RLIYLVSELD SGVPDRFSGS GSGTDFTLKISRVEAEDVGV YYCWQGTHFP YTFGGGTKVE IK An exemplary polynucleotide that encodes hLAG-3 mAb 1 VL-1 is SEQ ID NO:21 (nucleotides encoding the CDRLresidues are shown underlined): gatatcgtta tgactcagac accactgtca ctgagtgtgaccccaggtca gcccgctagt atttcctgta aatcatcccagtccctcctg catagcgatg gaaagaccta tttgaactggcttctgcaga aaccaggcca aagtccagag agattgatctacctcgtttc agaactcgac agtggagtgc ccgatcgcttctcagggtcc ggctctggga ctgattttac tctcaagatctcaagagtgg aggccgagga cgtcggggtt tactactgttggcagggtac ccacttccct tatacatttg gcggaggcacaaaagtggag attaaa The amino acid sequence of the VL Domain of hLAG-3 mAb 1 VL-2 (SEQ ID NO:22) is shown below (CDRLresidues are shown underlined): DIVMTQTPLS LSVTPGQPAS ISCKSSQSLL HSDGKTYLNWLLQRPGQSPE RLIYLVSELD SGVPDRFSGS GSGTDFTLKISRVEAEDVGV YYCWQGTHFPYTFGGGTKVE IK An exemplary polynucleotide that encodes hLAG-3 mAb 1 VL-2 is SEQ ID NO:23 (nucleotides encoding the CDRLresidues are shown underlined): gatatcgtta tgactcagac accactgtca ctgagtgtgaccccaggtca gcccgctagt atttcctgta aatcatcccagtccctcctg catagcgatggaaagaccta tttgaactggcttctgcaga gaccaggcca aagtccagag agattgatctacctcgtttc agaactcgac agtggagtgc ccgatcgcttctcagggtcc ggctctggga ctgattttac tctcaagatctcaagagtgg aggccgagga cgtcggggtt tactactgttggcagggtac ccacttcccttatacatttg gcggaggcacaaaagtggag attaaa The amino acid sequence of the VL Domain of hLAG-3 mAb 1 VL-3 (SEQ ID NO:24) is shown below (CDRLresidues are shown underlined): DIVMTQTPLS LSVTPGQPAS ISCKSSQSLL HSDGKTYLNWLLQKPGQPPE RLIYLVSELD SGVPDRFSGS GSGTDFTLKISRVEAEDVGV YYCWQGTHFPYTFGGGTKVE IK An exemplary polynucleotide that encodes hLAG-3 mAb 1 VL-3 is SEQ ID NO:25 (nucleotides encoding the CDRLresidues are shown underlined): gatatcgtta tgactcagac accactgtca ctgagtgtgaccccaggtca gcccgctagt atttcctgta aatcatcccagtccctcctg catagcgatggaaagaccta tttgaactggcttctgcaga aaccaggcca accgccagag agattgatctacctcgtttc agaactcgac agtggagtgc ccgatcgcttctcagggtcc ggctctggga ctgattttac tctcaagatctcaagagtgg aggccgagga cgtcggggtt tactactgttggcagggtac ccacttcccttatacatttg gcggaggcacaaaagtggag attaaa The amino acid sequence of the VL Domain of hLAG-3 mAb 1 VL-4 (SEQ ID NO:26) is shown below (CDRLresidues are shown underlined): DIVMTQTPLS LSVTPGQPAS ISCKSSQSLL HSDAKTYLNWLLQKPGQPPE RLIYLVSELD SGVPDRFSGS GSGTDFTLKISRVEAEDVGV YYCWQGTHFPYTFGGGTKVE IK An exemplary polynucleotide that encodes hLAG-3 mAb 1 VL-4 is SEQ ID NO:27 (nucleotides encoding the CDRLresidues are shown underlined): gatatcgtta tgactcagac accactgtca ctgagtgtgaccccaggtca gcccgctagt atttcctgta aatcatcccagtccctcctg catagcgatgcaaagaccta tttgaactggcttctgcaga aaccaggcca accgccagag agattgatctacctcgtttc agaactcgac agtggagtgc ccgatcgcttctcagggtcc ggctctggga ctgattttac tctcaagatctcaagagtgg aggccgagga cgtcggggtt tactactgttggcagggtac ccacttcccttatacatttg gcggaggcacaaaagtggag attaaa The CDRL1 of the VL Domain of hLAG-3 mAb 1 VL-4 comprises an glycine to alanine amino acid substitution and has the amino acid sequence: KSSQSLLHSDAKTYLN (SEQ ID NO:28), the substituted alanine is shown underlined). It is contemplated that a similar substitution may be incorporated into any of the LAG-3 mAb 1 CDRL1 Domains described above. B. The Anti-LAG-3 Antibody LAG-3 mAb 2 The amino acid sequence of the VH Domain of LAG-3 mAb 2 (SEQ ID NO:29) is shown below (CDRHresidues are shown underlined): EVQLQQSGPE LVKPGASVKI SCKTSGYTFTDYNIHWLRQSHGESLEWIGYIYPYSGDIGY NQKFKNRATL TVDNSSSTAYMDLRSLTSED SAVFYCARWHRNYFGPWFAYWGQGTPVTVS ACDRH1 of LAG-3 mAb 2 (SEQ ID NO: 31):DYNIHVH CDRH2 of LAG-3 mAb 2 (SEQ ID NO: 32):YIYPYSGDIGYNQKFKNVH CDRH3 of LAG-3 mAb 2 (SEQ ID NO: 33):WHRNYFGPWFAY An exemplary polynucleotide that encodes the VH Domain of LAG-3 mAb 2 is SEQ ID NO:30 (nucleotides encoding the CDRHresidues are shown underlined): gaggtccagc ttcagcagtc aggacctgag ctggtgaaacctggggcctc agtgaagatt tcctgcaaga cttctggatacacatttactgactacaacatacactggtt gaggcagagccatggagaga gccttgagtg gattggatatatttatccttacagtggtga tattggatac aaccagaagt tcaagaacagggccacattg atctgaagac attcctccag cacagcctacatggatctcc gcagcctgac actgtagaca tctgcagtcttttactgtgc aagatggcacaggaactact ttggcccctggtttgcttactggggccaag ggactccggt cactgtctct gca The amino acid sequence of the VL Domain of LAG-3 mAb 2 (SEQ ID NO:34) is shown below (CDRLresidues are shown underlined): DIVLTQSPAS LAVSLGQRAT ISCKASQSVD YDGESYMNWYQQKPGQPPKL LIYVVSNLESGIPARFSGSG SGTDFTLNIHPVEEEDAATY YCQQSSEDPLTFGAGTKLEL KCDRL1 of LAG-3 mAb 2 (SEQ ID NO: 36):KASQSVDYDGESYMNCDRL2 of LAG-3 mAb 2 (SEQ ID NO: 37):VVSNLESCDRL3 of LAG-3 mAb 2 (SEQ ID NO: 38):QQSSEDPLT An exemplary polynucleotide that encodes the VL Domain of LAG-3 mAb 2 is SEQ ID NO:35 (nucleotides encoding the CDRLresidues are shown underlined): gacattgtgc tgacccaatc tccagcttct ttggctgtgtctctagggca gagggccacc atctcctgca aggccagccaaagtgttgat tatgatggtgaaagttatat gaactggtaccaacagaaac caggacagcc acccaaactc ctcatttatgttgtatccaa tctagaatctgggatcccag ccaggtttagtggcagtggg tctgggacag acttcaccct caacatccatcctgtggagg aggaggatgc tgcaacctat tactgtcagcaaagtagtga ggatccgctcacgttcggtg ctgggaccaagctggagctg aaa C. The Anti-LAG-3 Antibody LAG-3 mAb 3 The amino acid sequence of the VH Domain of LAG-3 mAb 3 (SEQ ID NO:39) is shown below (CDRHresidues are shown underlined): EVRLQQSGPE LVKPGASVKI SCKASGYTFTDYNIHWVRQSHGQSLEWIGYIYPYNGDTGY NQKFKTKATL TVDNSSNTAYMELRSLASED SAVYYCTRWSRNYFGPWFAYWGQGTLVTVS ACDRH1 of LAG-3 mAb 3 (SEQ ID NO: 41):DYNIHCDRH2 of LAG-3 mAb 3 (SEQ ID NO: 42):YTYPYNGDTGYNQKFKTCDRH3 of LAG-3 mAb 3 (SEQ ID NO: 43):WSRNYFGPWFAY An exemplary polynucleotide that encodes the VH Domain of LAG-3 mAb 3 is SEQ ID NO:40 (nucleotides encoding the CDRHresidues are shown underlined): gaggtccggc ttcagcagtc aggacctgag ctggtgaaacctggggcctc agtgaagata tcctgcaagg cttctggatacacattcactgactacaacattcactgggt gaggcagagccatggacaga gccttgagtg gattggatatatttatccttataatggtga tactggctac aaccagaagt tcaagaccaaggccacattg actgtagaca attcctccaa cacagcctacatggaactcc gcagcctggc atctgaagac tctgcagtctattactgtac aagatggagcaggaactact ttggcccctggtttgcttactggggccaag ggactctggt cactgtctct gca The amino acid sequence of the VL Domain of LAG-3 mAb 3 (SEQ ID NO:44) is shown below (CDRLresidues are shown underlined): DIVLTQSPTS LAVSLGQRAT ISCKASQSVD YDGDSYMNWYQQKPGQPPKL LIYAASNLESGIPARFSGSG SGTDFTLNIHPVEEEDAATY YCQQSSEDPLTFGAGTKLEL KCDRL1 of LAG-3 mAb 3 (SEQ ID NO: 46):KASQSVDYDGDSYMNCDRL2 of LAG-3 mAb 3 (SEQ ID NO: 47):AASNLESCDRL3 of LAG-3 mAb 3 (SEQ ID NO: 48):QQSSEDPLT An exemplary polynucleotide that encodes the VL Domain of LAG-3 mAb 3 is SEQ ID NO:45 (nucleotides encoding the CDRHresidues are shown underlined): gacattgtgc tgacccaatc tccaacttct ttggctgtgtctctagggca gagggccacc atctcctgca aggccagccaaagtgttgat tatgatggtgatagttatat gaactggtatcaacagaaac caggacagcc acccaaactc ctcatctatgctgcatccaa tctagaatctgggatcccag ccaggtttagtggcagtggg tctgggacag acttcaccct caacatccatcctgtggagg aggaggatgc tgcaacctat tactgtcagcaaagtagtga ggatccgctcacgttcggtg ctgggaccaagctggagctg aaa D. The Anti-LAG-3 Antibody LAG-3 mAb 4 The amino acid sequence of the VH Domain of LAG-3 mAb 4 (SEQ ID NO:49) is shown below (CDRHresidues are shown underlined): EVQLHQSGPE LVKPGASVKI SCKTSGYTFTDYNIHWVKQSHGKSLEWIGYIYPYNGDAGY NQNFKTKATL TVDNSSSTAYMELRSLTSED SAVYYCARWNMNYFGPWFAYWGQGTLVTVS ACDRH1 of LAG-3 mAb 4 (SEQ ID NO: 51):DYNIHCDRH2 of LAG-3 mAb 4 (SEQ ID NO: 52):YTYPYNGDAGYNQNFKTCDRH3 of LAG-3 mAb 4 (SEQ ID NO: 53):WNMNYFGPWFAY An exemplary polynucleotide that encodes the VH Domain of LAG-4 mAb 4 is SEQ ID NO:50 (nucleotides encoding the CDRHresidues are shown underlined): gaggtccagc ttcaccagtc aggacctgag ctggtgaaacctggggcctc agtgaagata tcctgcaaga cttctggatacactttcactgactacaacatacactgggt gaagcagagccatggaaaga gccttgagtg gattggatatatttatccttacaatggtga tgctggctac aaccagaact tcaagaccaaggccacattg actgtagaca attcctccag cacagcctacatggagctcc gcagcctgac atctgaggac tctgcagtctattactgtgc aagatggaacatgaactact ttggcccctggtttgcttactggggccaag ggactctggt cactgtctct gcg The amino acid sequence of the VL Domain of LAG-3 mAb 4 (SEQ ID NO:54) is shown below (CDRLresidues are shown underlined): DIVLTQSPAS LAVSLGQRAT ISCKASQSVD YDGVTYINWYQQKPGQPPKL LIFAASNLESGIPARFSGSG SGTDFTLNIHPVEEEDAATY YCQQSNEDPLTFGAGTKLEL KCDRL1 of LAG-3 mAb 4 (SEQ ID NO: 56):KASQSVDYDGVTYINCDRL2 of LAG-3 mAb 4 (SEQ ID NO: 7570):AASNLESCDRL3 of LAG-3 mAb 4 (SEQ ID NO: 58):QQSNEDPLT An exemplary polynucleotide that encodes the VL Domain of LAG-3 mAb 4 is SEQ ID NO:55 (nucleotides encoding the CDRLresidues are shown underlined): gacattgtgc tgacccaatc tccagcttct ttggctgtgtctctagggca gagggccacc atctcctgca aggccagccaaagtgttgat tatgatggtgttacttatat caactggtaccaacagaaac caggacagcc acccaaactc ctcatctttgctgcatccaa tctagaatctgggatcccag ccaggtttagtggcagtggg tctgggacag acttcaccct caacatccatcctgtggagg aggaggatgc tgcaacctat tactgtcagcaaagtaatga ggatccgctcacgttcggtg ctgggaccaagctggagctg aaa E. The Anti-LAG-3 Antibody LAG-3 mAb 5 The amino acid sequence of the VH Domain of LAG-3 mAb 5 (SEQ ID NO:59) is shown below (CDRHresidues are shown underlined): EVQLQQSGPE LVKPGASVKI SCKASGYTFTDYNIHWVKQSPGKSLEWIGYIYPYSGDFGY NQKFKSKATL TVDNSSSTAYMDLRSLTSED SAVFYCARWHRNYFGPWFAYWGQGTLVTVS ACDRH1 of LAG-3 mAb 5 (SEQ ID NO: 61):DYNIHCDRH2 of LAG-3 mAb 5 (SEQ ID NO: 62):YTYPYSGDFGYNQKFKSCDRH3 of LAG-3 mAb 5 (SEQ ID NO: 63):WHRNYFGPWFAY An exemplary polynucleotide that encodes the VH Domain of LAG-3 mAb 5 is SEQ ID NO:60 (nucleotides encoding the CDRHresidues are shown underlined): gaggtccagc ttcagcagtc aggacctgag ctggtgaaacctggggcctc agtgaagatt tcctgcaaag cttctggatacacatttactgactacaacatacactgggt gaagcagagccctggaaaga gccttgaatg gattggatatatttatccttacagtggtga ttttggatac aaccagaagt tcaagagcaaggccacattg actgtagaca attcctccag cacagcctacatggatctcc gcagcctgac atctgaggac tctgcagtcttttactgtgc aagatggcacaggaactact ttggcccctggtttgcttactggggccaag ggactctggt cactgtctct gca The amino acid sequence of the VL Domain of LAG-3 mAb 5 (SEQ ID NO:64) is shown below (CDRLresidues are shown underlined): DIVLTQSPAS LAVSLGQRAT ISCKASQSVD YDGESYMNWYQQKPGQPPKL LIYVVSNLESGIPARFSGSG SGTDFTLNIHPVEEEDAATY YCQQSSEDPLTFGAGTKLEL KCDRL1 of LAG-3 mAb 5 (SEQ ID NO: 66):KASQSVDYDGESYMNCDRL2 of LAG-3 mAb 5 (SEQ ID NO: 67):VVSNLESCDRL3 of LAG-3 mAb 5 (SEQ ID NO: 68):QQSSEDPLT An exemplary polynucleotide that encodes the VL Domain of LAG-3 mAb 5 is SEQ ID NO:65 (nucleotides encoding the CDRLresidues are shown underlined): gacattgtgc tgacccaatc tccagcttct ttggctgtgtctctagggca gagggccacc atctcctgca aggccagccaaagtgttgat tatgatggtgaaagttatat gaactggtaccaacagaaac caggacagcc acccaaactc ctcatttatgttgtttccaa tctagaatctgggatcccag ccaggtttagtggcagtggg tctgggacag acttcaccct caacatccatcctgtggagg aggaggatgc tgcaacctat tactgtcagcaaagtagtga ggatccgctcacgttcggtg ctgggaccaagctggagctg aaa F. The Anti-LAG-3 Antibody LAG-3 mAb 6 1. Murine Anti-Human Antibody LAG-3 mAb 6 The amino acid sequence of the VH Domain of LAG-3 mAb 6 (SEQ ID NO:69 is shown below CDRHresidues are shown underlined): The amino acid sequence of the VH Domain of LAG-3 mAb 6 (SEQ ID NO:69) is shown below (CDRHresidues are shown underlined): EVLLQQSGPE LVKPGASVKI PCKASGYTFTDYNMDWVKQSHGESLEWIGD INPDNGVTIY NQKFEGKATL TVDKSSSTAYMELRSLTSED TAVYYCAREA DYFYFDYWGQ GTTLTVSSCDRH1 of LAG-3 mAb 6 (SEQ ID NO: 71):DYNMDCDRH2 of LAG-3 mAb 6 (SEQ ID NO: 72):DINPDNGVTIYNQKFEGCDRH3 of LAG-3 mAb 6 (SEQ ID NO: 73):EADYFYFDY An exemplary polynucleotide that encodes the VH Domain of LAG-3 mAb 6 is SEQ ID NO:70 (nucleotides encoding the CDRHresidues are shown underlined): gaggtcctgc tgcaacagtc tggacctgag ctggtgaagcctggggcttc agtgaagata ccctgcaagg cttctggatacacattcactgactacaacatggactgggt gaagcagagccatggagaga gccttgagtg gattggagatattaatcctgacaatggtgt tactatctac aaccagaagt ttgagggcaaggccacactg actgtagaca agtcctccag tacagcctacatggagctcc gcagcctgac atctgaggac actgcagtctattactgtgc aagagaggcggattacttct actttgactactggggccaa ggcaccactc tcacagtctc ctca The amino acid sequence of the VL Domain of LAG-3 mAb 6 (SEQ ID NO:74) is shown below (CDRLresidues are shown underlined): DIVMTQSHRF MSTSVGDRVS ITCKASQDVS SVVAWYQQKPGQSPKLLIFSASYRYTGVPD RFTGSGSGTD FTFTISSVQAADLAVYYCQQ HYSTPWTFGG GTKLEIKCDRL1 of LAG-3 mAb 6 (SEQ ID NO: 76):KASQDVSSVVACDRL2 of LAG-3 mAb 6 (SEQ ID NO: 77):SASYRYTCDRL3 of LAG-3 mAb 6 (SEQ ID NO: 78):QQHYSTPWT An exemplary polynucleotide that encodes the VL Domain of LAG-3 mAb 6 is SEQ ID NO:75 (nucleotides encoding the CDRs are shown underlined): gacattgtga tgacccagtc tcacagattc atgtccacatcagttggaga cagggtcagc atcacctgca aggccagtcaggatgtgagt tctgttgtagcctggtatca acagaaaccaggacaatctc ctaaattact gattttttcggcatcctaccggtacactgg agtccctgat cgcttcactg gcagtggatctgggacggat ttcactttca ccatcagcag tgtgcaggctgcagacctgg cagtttatta ctgtcagcaa cattatagtactccgtggac gttcggtgga ggcaccaagc tggaaatcaa a 2. Humanization of the Anti-LAG-3 Antibody LAG-3 mAb 6 to Form “hLAG-3 mAb 6” The above-described murine anti-LAG-3 antibody LAG-3mAb 6 was humanized in order to demonstrate the capability of humanizing an anti-LAG-3 antibody so as to decrease its antigenicity upon administration to a human recipient. The humanization yielded two humanized VH Domains, designated herein as “hLAG-3 mAb 6 VH-1,” and “hLAG-3 mAb 6 VH-2,” and two humanized VL Domains designated herein as “hLAG-3 mAb 6 VL-1,” and “hLAG-3 mAb 6 VL-2.” Any of the humanized VL Domains may be paired with either of the humanized VH Domains. Accordingly, any antibody comprising one of the humanized VL Domains paired with the humanized VH Domain is referred to generically as “hLAG-3 mAb 6,” and particular combinations of humanized VH/VL Domains are referred to by reference to the specific VH/VL Domains, for example a humanized antibody comprising hLAG-3 mAb 6 VH-1 and hLAG-3 mAb 6 VL-2 is specifically referred to as “hLAG-3 mAb 6(1.2).” The amino acid sequence of the VH Domain of hLAG-3 mAb 6 VH-1 (SEQ ID NO:79) is shown below (CDRHresidues are shown underlined): QVQLVQSGAE VKKPGASVKV SCKASGYTFTDYNMDWVRQAPGQGLEWMGDINPDNGVTIY NQKFEGRVTM TTDTSTSTAYMELRSLRSDD TAVYYCAREADYFYFDYWGQ GTTLTVSS An exemplary polynucleotide that encodes hLAG-3 mAb 6 VH-1 is SEQ ID NO:80 (nucleotides encoding the CDRHresidues are shown underlined): caggtccagc tggtgcagtc tggcgccgaa gtgaagaaacctggcgcaag cgtgaaggtg tcctgcaagg ccagcggctacaccttcaccgactacaacatggactgggt ccgacaggccccaggacagg gcctggaatg gatgggcgacatcaaccccgacaacggcgt gaccatctac aaccagaaat tcgagggcagagtgaccatg accaccgaca ccagcaccag caccgcctacatggaactgc ggtccctgcg gagcgacgac accgccgtgtactactgcgc cagagaggccgactacttct acttcgactactggggccag ggcaccaccc tgaccgtgtc ctcc An amino acid sequence of the VH Domain of hLAG-3 mAb 6 VH-2 (SEQ ID NO:81) is shown below (CDRHresidues are shown underlined): EVQLVESGGG LVKPGGSLRL SCAASGFTFSDYNMDWVRQAPGKGLEWVSDINPDNGVTIY NQKFEGRFTI SRDNAKNSLYLQMNSLRAED TAVYYCAREADYFYFDYWGQ GTTLTVSS An exemplary polynucleotide that encodes hLAG-3 mAb 6 VH-2 is SEQ ID NO:82 (nucleotides encoding the CDRHresidues are shown underlined): gaggtccagc tggtggaatc tggcggcgga ctggtcaagcctggcggcag cctgagactg agctgcgctg ccagcggcttcaccttcagcgactacaacatggactgggt ccgacaggcccctggcaagg gcctggaatg ggtgtccgacatcaaccccgacaacggcgt gaccatctac aaccagaagt tcgagggccggttcaccatc agccgggaca acgccaagaa cagcctgtacctgcagatga acagcctgcg ggccgaggac accgccgtgtactactgcgc cagagaggccgactacttct acttcgactactggggccag ggcaccaccc tgaccgtgtc ctcc The amino acid sequence of the VL Domain of hLAG-3 mAb 6 VL-1 (SEQ ID NO:83) is shown below (CDRLresidues are shown underlined): DIQMTQSPSS LSASVGDRVT ITCRASQDVS SVVAWYQQKPGKAPKLLIYSASYRYTGVPS RFSGSGSGTD FTLTISSLQPEDFATYYCQQ HYSTPWTFGG GTKLEIK An exemplary polynucleotide that encodes hLAG-3 mAb 6 VL-1 is SEQ ID NO:84 (nucleotides encoding the CDRLresidues are shown underlined): gacatccaga tgacccagag ccccagcagc ctgagcgccagcgtgggcga cagagtgacc atcacctgtc gggccagccaggatgtgtcc agcgtggtggcctggtatca gcagaagcccggcaaggccc ccaagctgct gatctacagcgccagctaccggtacacagg cgtgcccagc agattcagcg gcagcggctccggcaccgac ttcaccctga ccatcagcag cctgcagcccgaggacttcg ccacctacta ctgccagcag cactacagcaccccctggac cttcggcgga ggcaccaagc tggaaatcaa g The amino acid sequence of the VL Domain of hLAG-3 mAb 6 VL-2 (SEQ ID NO:85) is shown below (CDRLresidues are shown underlined): DIVMTQSPSS LSASVGDRVT ITCRASQDVS SVVAWYQQKPGKAPKLLIYSASYRYTGVPD RFSGSGSGTD FTFTISSLQPEDIAVYYCQQ HYSTPWTFGG GTKLEIK An exemplary polynucleotide that encodes hLAG-3 mAb 6 VL-2 is SEQ ID NO:86 (nucleotides encoding the CDRLresidues are shown underlined): gacatcgtga tgacccagag ccccagcagc ctgagcgccagcgtgggcga cagagtgacc atcacctgtc gggccagccaggatgtgtcc agcgtggtggcctggtatca gcagaagcccggcaaggccc ccaagctgct gatctacagcgccagctaccggtacacagg cgtgcccgat agattcagcg gcagcggctccggcaccgac ttcaccttca ccatcagcag cctgcagcccgaggacatcg ccgtttacta ctgccagcag cactacagcaccccctggac cttcggcgga ggcaccaagc tagaaatcaa a The CDRL1 of the VL Domain of hLAG-3 mAb 2 VL-1 and VL-2 comprises an lysine to arginine amino acid substitution and has the amino acid sequence:RASQDVSSVVA (SEQ ID NO:87), the substituted arginine is shown underlined). It is contemplated that a similar substitution may be incorporated into any of the LAG-3 mAb 6 CDRL1 Domains described above. Minor changes to the amino acid sequence of the VH and/or VL Domains provided herein are contemplated. For example, the C-terminal amino acid residue of any of the VH and/or VL Domains described herein may be substituted to facilitate sub-cloning. V. Anti-LAG-3 Antibodies LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, and/or LAG-3 mAb 6 and their Derivatives Having an Engineered Fc Region In traditional immune function, the interaction of antibody-antigen complexes with cells of the immune system results in a wide array of responses, ranging from effector functions such as antibody dependent cytotoxicity, mast cell degranulation, and phagocytosis to immunomodulatory signals such as regulating lymphocyte proliferation and antibody secretion. All of these interactions are initiated through the binding of the Fc Region of antibodies or immune complexes to specialized cell surface receptors on hematopoietic cells. The diversity of cellular responses triggered by antibodies and immune complexes results from the structural heterogeneity of the three Fc receptors: FcγRI (CD64), FcγRII (CD32), and FcγRIII (CD16). FcγRI (CD64), FcγRIIA (CD32A) and FcγRIII (CD16) are activating (i.e., immune system enhancing) receptors; FcγRIIB (CD32B) is an inhibiting (i.e., immune system dampening) receptor. In addition, interaction with the neonatual Fc Receptor (FcRn) mediates the recycling of IgG molecules from the endosome to the cell surface and release into the blood. The amino acid sequence of exemplary IgG1 (SEQ ID NO:1), IgG2 (SEQ ID NO:2), IgG3 (SEQ ID NO: 3), and IgG4 (SEQ ID NO:4) are presented above. Modification of the Fc Region normally leads to an altered phenotype, for example altered serum half-life, altered stability, altered susceptibility to cellular enzymes or altered effector function. It may be desirable to modify an antibody or other binding molecule of the present invention with respect to effector function, for example, so as to enhance the effectiveness of such molecule in treating cancer. Reduction or elimination of effector function is desirable in certain cases, for example in the case of antibodies whose mechanism of action involves blocking or antagonism, but not killing of the cells bearing a target antigen. Increased effector function is generally desirable when directed to undesirable cells, such as tumor and foreign cells, where the FcγRs are expressed at low levels, for example, tumor-specific B cells with low levels of FcγRIIB (e.g., non-Hodgkin's lymphoma, CLL, and Burkitt's lymphoma). In said embodiments, molecules of the invention with conferred or altered effector function activity are useful for the treatment and/or prevention of a disease, disorder or infection where an enhanced efficacy of effector function activity is desired. In certain embodiments, the LAG-3-binding molecules of the present invention comprise an Fc Region that possesses one or more modifications (e.g., substitutions, deletions, or insertions) to the sequence of amino acids of a wild-type Fc Region (e.g., SEQ ID NO:1), which reduce the affinity and avidity of the Fc Region and, thus, the molecule of the invention, for one or more FcγR receptors. In other embodiments, the molecules of the invention comprise an Fc Region that possesses one or more modifications to the amino acids of the wild-type Fc Region, which increase the affinity and avidity of the Fc Region and, thus, the molecule of the invention, for one or more FcγR receptors. In other embodiments, the molecules comprise a variant Fc Region wherein said variant confers or mediates increased antibody dependent cell mediated cytotoxicity (ADCC) activity and/or an increased binding to FcγRIIA, relative to a molecule comprising no Fc Region or comprising a wild-type Fc Region. In alternate embodiments, the molecules comprise a variant Fc Region wherein said variant confers or mediates decreased ADCC activity (or other effector function) and/or an increased binding to FcγRIIB, relative to a molecule comprising no Fc Region or comprising a wild-type Fc Region. In some embodiments, the invention encompasses LAG-3-binding molecules comprising a variant Fc Region, which variant Fc Region does not show a detectable binding to any FcγR, relative to a comparable molecule comprising the wild-type Fc Region. In other embodiments, the invention encompasses LAG-3-binding molecules comprising a variant Fc Region, which variant Fc Region only binds a single FcγR, preferably one of FcγRIIA, FcγRIIB, or FcγRIIIA. Any such increased affinity and/or avidity is preferably assessed by measuring in vitro the extent of detectable binding to the FcγR or FcγR-related activity in cells that express low levels of the FcγR when binding activity of the parent molecule (without the modified Fc Region) cannot be detected in the cells, or in cells which express non-FcγR receptor target antigens at a density of 30,000 to 20,000 molecules/cell, at a density of 20,000 to 10,000 molecules/cell, at a density of 10,000 to 5,000 molecules/cell, at a density of 5,000 to 1,000 molecules/cell, at a density of 1,000 to 200 molecules/cell or at a density of 200 molecules/cell or less (but at least 10, 50, 100 or 150 molecules/cell). The LAG-3-binding molecules of the present invention may comprise a variant Fc Region having altered affinities for an activating and/or inhibitory Fcγ receptor. In one embodiment, the LAG-3-binding molecule comprises a variant Fc Region that has increased affinity for FcγRIIB and decreased affinity for FcγRIIIA and/or FcγRIIA, relative to a comparable molecule with a wild-type Fc Region. In another embodiment, the LAG-3-binding molecule of the present invention comprise a variant Fc Region, which has decreased affinity for FcγRIIB and increased affinity for FcγRIIIA and/or FcγRIIA, relative to a comparable molecule with a wild-type Fc Region. In yet another embodiment, the LAG-3-binding molecules of the present invention comprise a variant Fc Region that has decreased affinity for FcγRIIB and decreased affinity for FcγRIIIA and/or FcγRIIA, relative to a comparable molecule with a wild-type Fc Region. In still another embodiment, the LAG-3-binding molecules of the present invention comprise a variant Fc Region, which has unchanged affinity for FcγRIIB and decreased (or increased) affinity for FcγRIIIA and/or FcγRIIA, relative to a comparable molecule with a wild-type Fc Region. In certain embodiments, the LAG-3-binding molecules of the present invention comprise a variant Fc Region having an altered affinity for FcγRIIIA and/or FcγRIIA such that the immunoglobulin has an enhanced effector function. Non-limiting examples of effector cell functions include antibody dependent cell mediated cytotoxicity, antibody dependent phagocytosis, phagocytosis, opsonization, opsonophagocytosis, cell binding, rosetting, C1q binding, and complement dependent cell mediated cytotoxicity. In a preferred embodiment, the alteration in affinity or effector function is at least 2-fold, preferably at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 50-fold, or at least 100-fold, relative to a comparable molecule comprising a wild-type Fc Region. In other embodiments of the invention, the variant Fc Region immunospecifically binds one or more FcRs with at least 65%, preferably at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 125%, at least 150%, at least 175%, at least 200%, at least 225%, or at least 250% greater affinity relative to a molecule comprising a wild-type Fc Region. Such measurements can be in vivo or in vitro assays, and in a preferred embodiment are in vitro assays such as ELISA or surface plasmon resonance assays. In different embodiments, the LAG-3-binding molecules of the present invention comprise a variant Fc Region wherein said variant agonizes at least one activity of an FcγR receptor, or antagonizes at least one activity of an FcγR receptor. In a preferred embodiment, the molecules comprise a variant that antagonizes one or more activities of FcγRIIB, for example, B-cell receptor-mediated signaling, activation of B-cells, B-cell proliferation, antibody production, intracellular calcium influx of B cells, cell cycle progression, FcγRIIB-mediated inhibition of FcεRI signaling, phosphorylation of FcγRIIB, SHIP recruitment, SHIP phosphorylation and association with Shc, or activity of one or more downstream molecules (e.g., MAP kinase, INK, p38, or Akt) in the FcγRIIB signal transduction pathway. In another embodiment, the LAG-3-binding molecules of the present invention comprise a variant that agonizes one or more activities of FcεRI, for example, mast cell activation, calcium mobilization, degranulation, cytokine production, or serotonin release. In certain embodiments, the molecules comprise an Fc Region comprising regions from two or more IgG isotypes (e.g., IgG1, IgG2, IgG3 and IgG4). As used herein, an Fc Region is said to be of a particular IgG isotype if its amino acid sequence is most homologous to that isotype relative to other IgG isotypes. The various IgG isotypes exhibit differing physical and functional properties including serum half-life, complement fixation, FcγR binding affinities and effector function activities (e.g., ADCC, CDC, etc.) due to differences in the amino acid sequences of their hinge and/or Fc Regions, for example as described in Flesch and Neppert (1999) J. Clin. Lab. Anal. 14:141-156; Chappel et al. (1993) J. Biol. Chem. 33:25124-25131; Chappel et al. (1991) Proc. Natl. Acad. Sci. (U.S.A.) 88:9036-9040; or Bruggemann et al. (1987) J. Exp. Med 166:1351-1361. This type of variant Fe Region may be used alone, or in combination with an amino acid modification, to affect Fc-mediated effector function and/or binding activity. In combination, the amino acid modification and IgG hinge/Fc Region may display similar functionality (e.g., increased affinity for FcγRIIA) and may act additively or, more preferably, synergistically to modify the effector functionality in the molecule of the invention, relative to a molecule of the invention comprising a wild-type Fc Region. In other embodiments, the amino acid modification and IgG Fc Region may display opposite functionality (e.g., increased and decreased affinity for FcγRIIA, respectively) and may act to selectively temper or reduce a specific functionality in the molecule of the invention, relative to a molecule of the invention not comprising an Fc Region or comprising a wild-type Fc Region of the same isotype. In a preferred specific embodiment, the LAG-3-binding molecules of the present invention comprise a variant Fc Region, wherein said variant Fc Region comprises at least one amino acid modification relative to a wild-type Fc Region, such that said molecule has an altered affinity for an FcR, provided that said variant Fc Region does not have a substitution at positions that make a direct contact with FcγR based on crystallographic and structural analysis of Fc-FcR interactions such as those disclosed by Sondermann et al. (2000) Nature 406:267-73. Examples of positions within the Fc Region that make a direct contact with FcγR are amino acid residues 234-239 (hinge region), amino acid residues 265-269 (B/C loop), amino acid residues 297-299 (C′/E loop), and amino acid residues 327-332 (F/G loop). In some embodiments, the molecules of the invention comprise variant Fc Regions comprise modification of at least one residue that does not make a direct contact with an FcγR based on structural and crystallographic analysis, e.g., is not within the Fc-FcγR binding site. Variant Fc Regions are well known in the art, and any known variant Fc Region may be used in the present invention to confer or modify the effector function exhibited by a molecule of the invention comprising an Fc Region (or portion thereof) as functionally assayed, e.g., in an NK dependent or macrophage dependent assay. For example, Fc Region variants identified as altering effector function are disclosed in PCT Publications No. WO 04/063351; WO 06/088494; WO 07/024249; WO 06/113665; WO 07/021841; WO 07/106707; and WO 2008/140603, and any suitable variant disclosed therein may be used in the present molecules. In certain embodiments, the LAG-3-binding molecules of the present invention comprise a variant Fc Region, having one or more amino acid modifications in one or more regions, which modification(s) alter (relative to a wild-type Fc Region) the Ratio of Affinities of the variant Fc Region to an activating FcγR (such as FcγRIIA or FcγRIIIA) relative to an inhibiting FcγR (such as FcγRIIB): Ratio⁢of⁢Affinities=Wild⁢‐⁢Type⁢to⁢Variant⁢Change⁢in⁢Affinity⁢to⁢Fc⁢γ⁢RActivatingWild⁢‐⁢Type⁢to⁢Variant⁢Change⁢in⁢Affinity⁢to⁢Fc⁢γ⁢RInhibiting Particularly preferred are LAG-3-binding molecules of the present invention that possess a variant Fc Region (relative to the wild-type Fc Region) in which the variant Fc Region has a Ratio of Affinities greater than 1. Such molecules have particular use in providing a therapeutic or prophylactic treatment of a disease, disorder, or infection, or the amelioration of a symptom thereof, where an enhanced efficacy of effector cell function (e.g., ADCC) mediated by FcγR is desired, e.g., cancer or infectious disease. In contrast, a variant Fc Region having a Ratio of Affinities less than 1 mediates decreased efficacy of effector cell function. Table 1 lists exemplary single, double, triple, quadruple and quintuple mutations by whether their Ratio of Affinities is greater than or less than 1. TABLE 1Exemplary Single and Multiple Mutations Listed by Ratio of AffinitiesSingleDoubleTripleQuadrupleQuintupleRatio of Affinities > 1F243LF243L &F243L, P247L &L234F, F243L,L235V, F243L,R292PN421KR292P &R292P,Y300LY300L &P396LD270EF243L &F243L, R292P &L235I, F243L,L235P, F243L,Y300LY300LR292P &R292P,Y300LY300L &P396LR292GF243L &F243L, R292P &L235Q, F243L,F243L, R292P,P396LV305IR292P &V305I,Y300LY300L &P396LR292PD270E &F243L, R292P &F243L, P247L,P396LP396LD270E &N421KR292P &F243L, Y300L &F243L, R255L,Y300LP396LD270E &P396LR292P &P247L, D270E &F243L, D270E,V305IN421KG316D &R416GR292P &R255L, D270E &F243L, D270E,P396LP396LK392T &P396LY300L &D270E, G316D &F243L, D270E,P396LR416GP396L &Q419HP396L &D270E, K392T &F243L, R292P,Q419HP396LY300L, &P396LD270E, P396L &F243L, R292P,Q419HV305I & P396LV284M, R292LP247L, D270E,& K370NY300L &N421KR292P, Y300L &R255L, D270E,P396LR292G &P396LR255L, D270E,Y300L &P396LD270E, G316D,P396L &R416GRatio of Affinities < 1Y300LF243L &F243L, R292P &P396LV305IP396LP247L &N421KR255L &P396LR292P &V305IK392T &P396LP396L &Q419H In a specific embodiment, in variant Fe Regions, any amino acid modifications (e.g., substitutions) at any of positions 235, 240, 241, 243, 244, 247, 262, 263, 269, 298, 328, or 330 and preferably one or more of the following residues: A240, I240, L241, L243, H244, N298, I328 or V330. In a different specific embodiment, in variant Fc Regions, any amino acid modifications (e.g., substitutions) at any of positions 268, 269, 270, 272, 276, 278, 283, 285, 286, 289, 292, 293, 301, 303, 305, 307, 309, 331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 416, 419, 430, 434, 435, 437, 438 or 439 and preferably one or more of the following residues: H280, Q280, Y280, G290, S290, T290, Y290, N294, K295, P296, D298, N298, P298, V298, I300 or L300. In a preferred embodiment, in variant Fc Regions that bind an FcγR with an altered affinity, any amino acid modifications (e.g., substitutions) at any of positions 255, 256, 258, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 300, 301, 303, 305, 307, 309, 312, 320, 322, 326, 329, 330, 332, 331, 333, 334, 335, 337, 338, 339, 340, 359, 360, 373, 376, 416, 419, 430, 434, 435, 437, 438 or 439. Preferably, the variant Fc Region has any of the following residues: A256, N268, Q272, D286, Q286, S286, A290, S290, A298, M301, A312, E320, M320, Q320, R320, E322, A326, D326, E326, N326, S326, K330, T339, A333, A334, E334, H334, L334, M334, Q334, V334, K335, Q335, A359, A360 or A430. In a different embodiment, in variant Fe Regions that bind an FcγR (via its Fe Region) with a reduced affinity, any amino acid modifications (e.g., substitutions) at any of positions 252, 254, 265, 268, 269, 270, 278, 289, 292, 293, 294, 295, 296, 298, 300, 301, 303, 322, 324, 327, 329, 333, 335, 338, 340, 373, 376, 382, 388, 389, 414, 416, 419, 434, 435, 437, 438 or 439. In a different embodiment, in variant Fc Regions that bind an FcγR (via its Fc Region) with an enhanced affinity, any amino acid modifications (e.g., substitutions) at any of positions 280, 283, 285, 286, 290, 294, 295, 298, 300, 301, 305, 307, 309, 312, 315, 331, 333, 334, 337, 340, 360, 378, 398 or 430. In a different embodiment, in variant Fc Regions that binds FcγRIIA with an enhanced affinity, any of the following residues: A255, A256, A258, A267, A268, N268, A272, Q272, A276, A280, A283, A285, A286, D286, Q286, S286, A290, S290, M301, E320, M320, Q320, R320, E322, A326, D326, E326, S326, K330, A331, Q335, A337 or A430. Preferred variants include one or more modifications at any of positions: 228, 230, 231, 232, 233, 234, 235, 239, 240, 241, 243, 244, 245, 247, 262, 263, 264, 265, 266, 271, 273, 275, 281, 284, 291, 296, 297, 298, 299, 302, 304, 305, 313, 323, 325, 326, 328, 330 or 332. Particularly preferred variants include one or more modifications selected from groups A-AI: A228E, 228K, 228Y or 228G;B230A, 230E, 230Y or 230G;C231E, 231K, 231Y, 231P or 231G;D232E, 232K, 232Y, 232G;E233D;F234I or 234F;G235D, 235Q, 235P, 235I or 235V;H239D, 239E, 239N or 239Q;I240A, 240I, 240M or 240T;J243R, 243, 243Y, 243L, 243Q, 243W, 243H or 243I;K244H;L245A;M247G, 247V or 247L;N262A, 262E, 262I, 262T, 262E or 262F;O263A, 263I, 263M or 263T;P264F, 264E, 264R, 264I, 264A, 264T or 264W;Q265F, 265Y, 265H, 265I, 265L, 265T, 265V, 265N or 265Q;R266A, 266I, 266M or 266T;S271D, 271E, 271N, 271Q, 271K, 271R, 271S, 271T, 271H, 271A,271V, 271L, 271I, 271F, 271M, 271Y, 271W or 271G;T273I;U275L or 275W;V281D, 281K, 281Y or 281P;W284E, 284N, 284T, 284L, 284Y or284M;X291D, 291E, 291Q, 291T, 291H, 291I or 291G;Y299A, 299D, 299E, 299F, 299G, 299H, 299I, 299K, 299L, 299M,299N, 299P, 299Q, 299R, 299S, 299V, 299W or 299Y;Z302I;AA304D, 304N, 304T, 304H or 304LAB305I;AC313F;AD323I;AE325A, 325D, 325E, 325G, 325H, 325I, 325L, 325K, 325R, 325S,325F, 325M, 325T, 325V, 325Y, 325W or 325P;AF328D, 328Q, 328K, 328R, 328S, 328T, 328V, 328I, 328Y, 328W,328P, 328G, 328A, 328E, 328F, 328H, 328M or 328N;AG330L, 330Y, 330I or 330V;AH332A, 332D, 332E, 332H, 332N, 332Q, 332T, 332K, 332R, 332S,332V, 332L, 332F, 332M, 332W, 332P, 332G or 332Y; andAI336E, 336K or 336Y Still more particularly preferred variants include one or more modifications selected from Groups 1-105: GroupVariant1A330L/I332E2D265F/N297E/I332E3D265Y/N297D/I332E4D265Y/N297D/T299L/I332E5F241E/F243Q/V262T/V264F6F241E/F243Q/V262T/V264E/I332E7F241E/F243R/V262E/V264R8F241E/F243R/V262E/V264R/I332E9F241E/F243Y/V262T/V264R10F241E/F243Y/V262T/V264R/I332E11F241L/F243L/V262I/V264I12F241L/V262I13F241R/F243Q/V262T/V264R14F241R/F243Q/V262T/V264R/I332E15F241W/F243W/V262A/V264A16F241Y/F243Y/V262T/V264T17F241Y/F243Y/V262T/V264T/N297D/I332E18F243L/V262I/V264W19P243L/V264I20L328D/I332E21L328E/I332E22L328H/I332E23L3281/I332E24L328M/I332E25L328N/I332E26L328Q/I332E27L328T/I332E28L328V/I332E29N297D/A330Y/I332E30N297D/I332E31N297D/I332E/S239D/A330L32N297D/S298A/A330Y/I332E33N297D/T299L/I332E34N297D/T299F/I332E/N297D/T299H/I332E35N297D/T2991/I332E36N297D/T299L/I332E37N297D/T299V/I332E38N297E/I332E39N297S/I332E40P230A/E233D/I332E41P244H/P245A/P247V42S239D/A330L/I332E43S239D/A330Y/I332E44S239D/A330Y/I332E/K326E45S239D/A330Y/I332E/K326T46S239D/A330Y/I332E/L234I47S239D/A330Y/I332E/L235D48S239D/A330Y/I332E/V240I49S239D/A330Y/I332E/V264T50S239D/A330Y/I332E/V266I51S239D/D265F/N297D/I332E52S239D/D265H/N297D/I332E53S239D/D265I/N297D/I332E54S239D/D265L/N297D/I332E55S239D/D265T/N297D/I332E56S239D/D265V/N297D/I332E57S239D/D265Y/N297D/I332E58S239D/I332D59S239D/I332E60S239D/I332E/A330I61S239D/I332N62S239D/I332Q63S239D/N297D/I332E64S239D/N297D/I332E/A330Y65S239D/N297D/I332E/A330Y/F241S/F243H/V262T/V264T66S239D/N297D/I332E/K326E67S239D/N297D/I332E/L235D68S239D/S298A/I332E69S239D/V2641/A330L/I332E70S239D/V2641/I332E71S239D/V264I/S298A/I332E72S239E/D265N73S239E/D265Q74S239E/I332D75S239E/I332E76S239E/I332N77S239E/I332Q78S239E/N297D/I332E79S239E/V264I/A330Y/I332E80S239E/V2641/I332E81S239E/V264I/S298A/A330Y/I332E82S239N/A330L/I332E83S239N/A330Y/I332E84S239N/I332D85S239N/I332E86S239N/I332N87S239N/I332Q88S239N1S298A/I332E89S239Q/I332D90S239Q/I332E91S239Q/I332N92S239Q/I332Q93S239Q/V2641/I332E94S298A/I332E95V264E/N297D/I332E96V2641/A330L/I332E97V2641/A330Y/I332E98V2641/I332E99V264I/S298A/I332E100Y296D/N297D/I332E101Y296E/N297D/I332 E102Y296H/N297D/I332E103Y296N/N297D/I332E104Y296Q/N2971/I332E105Y296T/N297D/I332E In one embodiment, a LAG-3 binding molecule of the invention will comprise a variant Fc Region having at least one modification in the Fc Region. In certain embodiments, the variant Fc Region comprises at least one substitution selected from the group consisting of L235V, F243L, R292P, Y300L, V305I, and P396L, wherein said numbering is that of the EU index according to Kabat. In a specific embodiment, the variant Fc Region comprises:(A) at least one substitution selected from the group consisting of F243L, R292P, Y300L, V305I, and P396L;(B) at least two substitutions selected from the group consisting of:(1) F243L and P396L;(2) F243L and R292P; and(3) R292P and V305I;(C) at least three substitutions selected from the group consisting of:(1) F243L, R292P and Y300L;(2) F243L, R292P and V305I;(3) F243L, R292P and P396L; and(4) R292P, V305I and P396L;(D) at least four substitutions selected from the group consisting of:(1) F243L, R292P, Y300L and P396L; and(2) F243L, R292P, V305I and P396L; or(E) at least the five substitutions selected from the group consisting of:(1) F243L, R292P, Y300L, V305I and P396L; and(2) L235V, F243L, R292P, Y300L and P396L. In another specific embodiment, the variant Fc Region comprises substitutions of:(A) F243L, R292P, and Y300L;(B) L235V, F243L, R292P, Y300L, and P396L; or(C) F243L, R292P, Y300L, V305I, and P396L. In one embodiment, a LAG-3-binding molecule of the invention comprises a variant Fc Region that exhibits decreased (or substantially no) binding to FcγRIA (CD64), FcγRIIA (CD32A), FcγRIIB (CD32B), FcγRIIIA (CD16a) or FcγRIIIB (CD16b) (relative to the binding exhibited by the wild-type IgG1 Fc Region (SEQ ID NO:1)). In one embodiment, a LAG-3-binding molecule of the invention will comprise a variant Fc Region that exhibits reduced (or substantially no) binding to an FcγR (e.g., FcγRIIIA) and reduced (or substantially no) ADCC effector function. In certain embodiments, the variant Fc Region comprises at least one substitution selected from the group consisting of L234A, L235A, D265A, N297Q, and N297G. In a specific embodiment, the variant Fc Region comprises the substitution of L234A; L235A; L234A and L235A; D265A; N297Q, or N297G. In a different embodiment, a LAG-3-binding molecule of the invention comprises an Fc Region which inherently exhibits decreased (or substantially no) binding to FcγRIIIA (CD16a) and/or reduced effector function (relative to the binding exhibited by the wild-type IgG1 Fc Region (SEQ ID NO:1)). In a specific embodiment, a LAG-3-binding molecule of the present invention comprises an IgG2 Fc Region (SEQ ID NO:2) or an IgG4 Fe Region (SEQ ID NO:4). When an IgG4 Fe Region in utilized, the instant invention also encompasses the introduction of a stabilizing mutation, such the IgG4hinge region S228P substitution (see, e.g., SEQ ID NO:117: ESKYGPPCPPCP, (Lu et al., (2008) “The Effect Of A Point Mutation On The Stability Of Igg4As Monitored By Analytical Ultracentrifugation,” J. Pharmaceutical Sciences 97:960-969) to reduce the incidence of strand exchange. Other stabilizing mutations known in the art may be introduced into an IgG4 Fe Region (Peters, P et al., (2012) “Engineering an Improved IgG4Molecule with Reduced Disulfide Bond Heterogeneity and Increased Fab Domain Thermal Stability,” J. Biol. Chem., 287:24525-24533; PCT Patent Publication No: WO 2008/145142). In other embodiments, the invention encompasses the use of any Fc variant known in the art, such as those disclosed in Jefferis, B. J. et al. (2002) “Interaction Sites On Human IgG-Fc For FcgammaR: Current Models,” Immunol. Lett. 82:57-65; Presta, L. G. et al. (2002) “Engineering Therapeutic Antibodies For Improved Function,” Biochem. Soc. Trans. 30:487-90; Idusogie, E. E. et al. (2001) “Engineered Antibodies With Increased Activity To Recruit Complement,” J. Immunol. 166:2571-75; Shields, R. L. et al. (2001) “High Resolution Mapping Of The Binding Site On Human IgG1For Fc Gamma RI, Fc Gamma RII, Fc Gamma RII, And FcRn And Design Of IgG1Variants With Improved Binding To The Fc gamma R,” J. Biol. Chem. 276:6591-6604; Idusogie, E. E. et al. (2000) “Mapping Of The C1q Binding Site On Rituxan, A Chimeric Antibody With A Human IgG Fc,” J. Immunol. 164:4178-84; Reddy, M. P. et al. (2000) “Elimination Of Fc Receptor-Dependent Effector Functions Of A Modified IgG4Monoclonal Antibody To Human CD4,” J. Immunol. 164:1925-1933; Xu, D. et al. (2000) “In Vitro Characterization of Five Humanized OKT3Effector Function Variant Antibodies,” Cell. Immunol. 200:16-26; Armour, K. L. et al. (1999) “Recombinant human IgG Molecules Lacking Fcgamma Receptor I Binding And Monocyte Triggering Activities,” Eur. J. Immunol. 29:2613-24; Jefferis, R. et al. (1996) “Modulation Of Fc(Gamma)R And Human Complement Activation By IgG3-Core Oligosaccharide Interactions,” Immunol. Lett. 54:101-04; Lund, J. et al. (1996) “Multiple Interactions Of IgG With Its Core Oligosaccharide Can Modulate Recognition By Complement And Human Fc Gamma Receptor I And Influence The Synthesis Of Its Oligosaccharide Chains,” J. Immunol. 157:4963-4969; Hutchins et al. (1995) “Improved Biodistribution, Tumor Targeting, And Reduced Immunogenicity In Mice With A Gamma4Variant Of Campath-1H,” Proc. Natl. Acad. Sci. (U.S.A.) 92:11980-84; Jefferis, R. et al. (1995) “Recognition Sites On Human IgG For Fc Gamma Receptors: The Role Of Glycosylation,” Immunol. Lett. 44:111-17; Lund, J. et al. (1995) “Oligosaccharide-Protein Interactions In IgG Can Modulate Recognition By Fc Gamma Receptors,” FASEB J. 9:115-19; Alegre, M. L. et al. (1994) “A Non-Activating “Humanized” Anti-CD3Monoclonal Antibody Retains Immunosuppressive Properties In Vivo,” Transplantation 57:1537-1543; Lund et al. (1992) “Multiple Binding Sites On The CH2Domain Of IgG For Mouse Fc Gamma RII,” Mol. Immunol. 29:53-59; Lund et al. (1991) “Human Fc Gamma RI And Fc Gamma RII Interact With Distinct But Overlapping Sites On Human IgG,” J. Immunol. 147:2657-2662; Duncan, A. R. et al. (1988) “Localization Of The Binding Site For The Human High-Affinity Fc Receptor On IgG,” Nature 332:563-564; U.S. Pat. Nos. 5,624,821; 5,885,573; 6,194,551; 7,276,586; and 7,317,091; and PCT Publications WO 00/42072 and PCT WO 99/58572. In some embodiments, the molecules of the invention further comprise one or more glycosylation sites, so that one or more carbohydrate moieties are covalently attached to the molecule. Preferably, the molecules of the invention with one or more glycosylation sites and/or one or more modifications in the Fc Region confer or have an enhanced antibody mediated effector function, e.g., enhanced ADCC activity, compared to the unmodified antibody. In some embodiments, the invention further comprises molecules comprising one or more modifications of amino acids that are directly or indirectly known to interact with a carbohydrate moiety of the Fc Region, including but not limited to amino acids at positions 241, 243, 244, 245, 245, 249, 256, 258, 260, 262, 264, 265, 296, 299, or 301. Amino acids that directly or indirectly interact with a carbohydrate moiety of an Fc Region are known in the art, see, e.g., Jefferis et al., 1995Immunology Letters,44: 111-7, which is incorporated herein by reference in its entirety. In another embodiment, the invention encompasses molecules that have been modified by introducing one or more glycosylation sites into one or more sites of the molecules, preferably without altering the functionality of the molecules, e.g., binding activity to target antigen or FcγR. Glycosylation sites may be introduced into the variable and/or constant region of the molecules of the invention. As used herein, “glycosylation sites” include any specific amino acid sequence in an antibody to which an oligosaccharide (i.e., carbohydrates containing two or more simple sugars linked together) will specifically and covalently attach. Oligosaccharide side chains are typically linked to the backbone of an antibody via either N- or O-linkages. N-linked glycosylation refers to the attachment of an oligosaccharide moiety to the side chain of an asparagine residue. O-linked glycosylation refers to the attachment of an oligosaccharide moiety to a hydroxyamino acid, e.g., serine, threonine. The molecules of the invention may comprise one or more glycosylation sites, including N-linked and O-linked glycosylation sites. Any glycosylation site for N-linked or O-linked glycosylation known in the art may be used in accordance with the instant invention. An exemplary N-linked glycosylation site that is useful in accordance with the methods of the present invention is the amino acid sequence: Asn-X-Thr/Ser, wherein X may be any amino acid and Thr/Ser indicates a threonine or a serine. Such a site or sites may be introduced into a molecule of the invention using methods well known in the art to which this invention pertains (see for example, INVITROMUTAGENESIS, RECOMBINANTDNA: A SHORTCOURSE, J. D. Watson, et al. W. H. Freeman and Company, New York, 1983, chapter 8, pp. 106-116, which is incorporated herein by reference in its entirety. An exemplary method for introducing a glycosylation site into a molecule of the invention may comprise: modifying or mutating an amino acid sequence of the molecule so that the desired Asn-X-Thr/Ser sequence is obtained. In some embodiments, the invention encompasses methods of modifying the carbohydrate content of a molecule of the invention by adding or deleting a glycosylation site. Methods for modifying the carbohydrate content of antibodies (and molecules comprising antibody domains, e.g., Fc Region) are well known in the art and encompassed within the invention, see, e.g., U.S. Pat. No. 6,218,149; EP 0 359 096 B1; U.S. Publication No. US 2002/0028486; WO 03/035835; U.S. Publication No. 2003/0115614; U.S. Pat. Nos. 6,218,149; 6,472,511; all of which are incorporated herein by reference in their entirety. In other embodiments, the invention encompasses methods of modifying the carbohydrate content of a molecule of the invention by deleting one or more endogenous carbohydrate moieties of the molecule. In a specific embodiment, the invention encompasses shifting the glycosylation site of the Fc Region of an antibody, by modifying positions adjacent to 297. In a specific embodiment, the invention encompasses modifying position 296 so that position 296 and not position 297 is glycosylated. Effector function can also be modified by techniques such as by introducing one or more cysteine residues into the Fc Region, thereby allowing interchain disulfide bond formation in this region to occur, resulting in the generation of a homodimeric antibody that may have improved internalization capability and/or increased complement-mediated cell killing and ADCC (Caron, P. C. et al. (1992) “Engineered Humanized Dimeric Forms Of IgG Are More Effective Antibodies,” J. Exp. Med. 176:1191-1195; Shopes, B. (1992) “A Genetically Engineered Human IgG Mutant With Enhanced Cytolytic Activity,” J. Immunol. 148(9):2918-2922. Homodimeric antibodies with enhanced antitumor activity may also be prepared using heterobifunctional cross-linkers as described in Wolff, E. A. et al. (1993) “Monoclonal Antibody Homodimers: Enhanced Antitumor Activity In Nude Mice,” Cancer Research 53:2560-2565. Alternatively, an antibody can be engineered which has dual Fc Regions and may thereby have enhanced complement lysis and ADCC capabilities (Stevenson, G. T. et al. (1989) “A Chimeric Antibody With Dual Fc Regions(bisFabFc)Prepared By Manipulations At The IgG Hinge,” Anti-Cancer Drug Design 3:219-230). The serum half-life of the molecules of the present invention comprising Fc Regions may be increased by increasing the binding affinity of the Fc Region for FcRn. The term “half-life” as used herein means a pharmacokinetic property of a molecule that is a measure of the mean survival time of the molecules following their administration. Half-life can be expressed as the time required to eliminate fifty percent (50%) of a known quantity of the molecule from a subject's body (e.g., human patient or other mammal) or a specific compartment thereof, for example, as measured in serum, i.e., circulating half-life, or in other tissues. In general, an increase in half-life results in an increase in mean residence time (MRT) in circulation for the molecule administered. In some embodiments, the LAG-3-binding molecules of the present invention comprise a variant Fe Region that comprises at least one amino acid modification relative to a wild-type Fc Region and that exhibit an increased half-life (relative to a wild-type Fc Region). In some embodiments, the LAG-3-binding molecules of the present invention comprise a variant Fc Region that comprises a half-life-extending amino acid substitution at one or more positions selected from the group consisting of 238, 250, 252, 254, 256, 257, 256, 265, 272, 286, 288, 303, 305, 307, 308, 309, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424, 428, 433, 434, 435, and 436, as numbered by the EU index according to Kabat. Numerous specific mutations capable of increasing the half-life of an Fc Region-containing molecule are known in the art and include, for example M252Y, S254T, T256E, and combinations thereof. For example, see the mutations described in U.S. Pat. Nos. 6,277,375, 7,083,784; 7,217,797, 8,088,376; U.S. Publication Nos. 2002/0147311; 2007/0148164; and International Publication Nos. WO 98/23289 WO 2009/058492, and WO 2010/033279, which are herein incorporated by reference in their entireties. Fc Region-containing molecules with enhanced half-life also include those with substitutions at two or more of Fc Region residues 250, 252, 254, 256, 257, 288, 307, 308, 309, 311, 378, 428, 433, 434, 435 and 436. In particular, two or more substitutions selected from: T250Q, M252Y, S254T, T256E, K288D, T307Q, V308P, A378V, M428L, N434A, H435K, and Y436I. In a specific embodiment, the variant Fc Region comprises substitutions of:(A) 252Y, 254T and 256E;(B) M252Y and S254T;(C) M252Y and T256E;(D) 250Q and 428L;(E) T307Q and N434A;(F) A378V and N434A;(G) N434A and Y436I;(H) V308P and N434A; or(I) K288D and H435K. The instant invention further encompasses variant Fe Regions comprising:(A) one or more mutations which alter effector function and/or FcγR; and(B) one or more mutations which extend serum half-life. VI. Bispecific LAG-3-Binding Molecules of the Present Invention One embodiment of the present invention relates to bispecific binding molecules that are capable of binding to a “first epitope” and a “second epitope,” wherein the first epitope is an epitope of human LAG-3 and the second epitope is the same or a different epitope of LAG-3, or is an epitope of another molecule that is present on the surface of an immune cell (such as a T lymphocyte) and is involved in regulating an immune checkpoint. In certain embodiments, the second epitope is preferably not an epitope of LAG-3. In one embodiment, the second epitope is an epitope of B7-H3, B7-H4, BTLA, CD3, CD8, CD16, CD27, CD32, CD40, CD40L, CD47, CD64, CD70, CD80, CD86, CD94, CD137, CD137L, CD226, CTLA-4, Galectin-9, GITR, GITRL, HHLA2, ICOS, ICOSL, KIR, LAG-3, LIGHT, MHC class I or II, NKG2a, NKG2d, OX40, OX40L, PD1H, PD-1, PD-L1, PD-L2, PVR, SIRPa, TCR, TIGIT, TIM-3 or VISTA. In a specific embodiment, the second epitope is CD137, PD-1, OX40, TIGIT, or TIM-3. In certain embodiments, such bispecific molecules comprise more than two epitope-binding sites. Such bispecific molecules may, for example, bind two or more different epitopes of LAG-3 and at least one epitope of a molecule that is not LAG-3. The instant invention encompasses bispecific antibodies capable of simultaneously binding to LAG-3 and the second epitope (e.g. B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, MHC class I or II, OX40, PD-1, PD-L1, TCR, TIM-3, etc.). In some embodiments, the bispecific antibody capable of simultaneously binding to PD-1 and the second epitope is produced using any of the methods described in PCT Publication Nos. WO 1998/002463, WO 2005/070966, WO 2006/107786 WO 2007/024715, WO 2007/075270, WO 2006/107617, WO 2007/046893, WO 2007/146968, WO 2008/003103, WO 2008/003116, WO 2008/027236, WO 2008/024188, WO 2009/132876, WO 2009/018386, WO 2010/028797, WO2010028796, WO 2010/028795, WO 2010/108127, WO 2010/136172, WO 2011/086091, WO 2011/133886, WO 2012/009544, WO 2013/003652, WO 2013/070565, WO 2012/162583, WO 2012/156430, WO 2013/174873, and WO 2014/022540, each of which is hereby incorporated herein by reference in its entirety. 1. Bispecific Diabodies Lacking Fc Regions One embodiment of the present invention relates to bispecific monovalent diabodies that comprise, and most preferably consist of, a first polypeptide chain and a second polypeptide chain, whose sequences permit the polypeptide chains to covalently bind to each other to form a covalently associated diabody that is capable of simultaneously binding to a first epitope (“Epitope 1”) and a second epitope (Epitope 2”), such epitopes not being identical to one another. Such bispecific diabodies thus comprise “VL1”/“VH1” domains that are capable of binding to the first epitope (VLEpitope 1/VHEpitope 1) and “VL2”/“VH2” domains that are capable of binding to the second epitope (VLEpitope 2/VHEpitope 2). The notation “VL1” and “VH1” denote respectively, the Variable Light Chain Domain and Variable Heavy Chain Domain that bind the “first” epitope of such bispecific diabody. Similarly, the notation “VL2” and “VH2” denote respectively, the Variable Light Chain Domain and Variable Heavy Chain Domain that bind the “second” epitope of such bispecific diabody. In one embodiment, Epitope 1 of such diabody molecules is an epitope of LAG-3 and Epitope 2 of such diabody molecules is not an epitope of LAG-3 (for example, it is an epitope of B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, MHC class I or II, OX40, PD-1, PD-L1, TCR, TIM-3, etc.). The VL Domain of the first polypeptide chain of such LAG-2 binding diabodies interacts with the VH Domain of the second polypeptide chain to form a first functional epitope-binding site that is specific for a first antigen (i.e., either LAG-3 or an antigen that contains the second epitope). Likewise, the VL Domain of the second polypeptide chain interacts with the VH Domain of the first polypeptide chain in order to form a second functional epitope-binding site that is specific for a second antigen (i.e., either an antigen that contains the second epitope or LAG-3). Thus, the selection of the VL and VH Domains of the first and second polypeptide chains is coordinated, such that the two polypeptide chains of the diabody collectively comprise VL and VH Domains capable of binding to both an epitope of LAG-3 and to the second epitope (i.e., they collectively comprise VLLAG-3/VHLAG-3and VLEpitope 2/VHEpitope 2Domains). It is irrelevant whether a particular pair of binding domains (i.e., VLEpitope 1/VHEpitope 1or VLEpitope 2/VHEpitope 2) an epitope of an antigen having Epitope 1 or an epitope of an antigen having Epitope 2) is designated as the first vs. the second epitope of the diabody; such notation having relevance only with respect to the presence and orientation of domains of the polypeptide chains of the binding molecules of the present invention The first polypeptide chain of an embodiment of such bispecific monovalent diabodies comprises, in the N-terminal to C-terminal direction, an N-terminus, the VL Domain of a monoclonal antibody capable of binding to either the first epitope or the VL Domain of a monoclonal antibody capable of binding to the second epitope (i.e., either VLLAG-3or VLEpitope 2), a first intervening spacer peptide (Linker 1), a VH Domain of a monoclonal antibody capable of binding to either the second epitope (if such first polypeptide chain contains VLLAG-3) or a VH Domain of a monoclonal antibody capable of binding to the first epitope (if such first polypeptide chain contains VLEpitope 2), a second intervening spacer peptide (Linker 2), optionally comprising a cysteine residue, a Heterodimer-Promoting Domain and a C-terminus (FIG.1). The second polypeptide chain of this embodiment of bispecific monovalent diabodies comprises, in the N-terminal to C-terminal direction, an N-terminus, a VL Domain of a monoclonal antibody capable of binding to LAG-3 or a VL Domain of a monoclonal antibody capable of binding to the second epitope (i.e., either VLLAG-3or VLEpitope 2, and being the VL Domain not selected for inclusion in the first polypeptide chain of the diabody), an intervening linker peptide (Linker 1), a VH Domain of a monoclonal antibody capable of binding to either the second epitope (if such second polypeptide chain contains VLLAG-3) or a VH Domain of a monoclonal antibody capable of binding to LAG-3 (if such second polypeptide chain contains VLEpitope 2), a second intervening spacer peptide (Linker 2) optionally containing a cysteine residue, a Heterodimer-Promoting Domain, and a C-terminus (FIG.1). Most preferably, the length of the intervening linker peptide (e.g., Linker 1) that separates such VL and VH Domains) is selected to substantially or completely prevent the VL and VH Domains of the polypeptide chain from binding to one another. Thus the VL and VH Domains of the first polypeptide chain are substantially or completely incapable of binding to one another. Likewise, the VL and VH Domains of the second polypeptide chain are substantially or completely incapable of binding to one another. A preferred intervening spacer peptide (Linker 1) has the sequence (SEQ ID NO:88): GGGSGGGG. The length and composition of the second intervening linker peptide (Linker 2) is selected based on the choice of heterodimer-promoting domains. Typically, the second intervening linker peptide (Linker 2) will comprise 3-20 amino acid residues. In particular, where the heterodimer-promoting domains do not comprise a cysteine residue a cysteine-containing second intervening linker peptide (Linker 2) is utilized. The cysteine-containing second intervening spacer peptide (Linker 2) will contain 1, 2, 3 or more than 3 cysteine residue(s). A preferred cysteine-containing spacer peptide (Linker 2) has the sequence is SEQ ID NO:89: GGCGGG. Alternatively, Linker 2 does not comprise a cysteine (e.g., GGG, GGGS (SEQ ID NO:90), LGGGSG (SEQ ID NO:91), GGGSGGGSGGG (SEQ ID NO:92), ASTKG (SEQ ID NO:93), LEPKSS (SEQ ID NO:94), APSSS (SEQ ID NO:95), etc.) and a Cysteine-Containing Heterodimer-Promoting Domain, as described below is used. Optionally, both a cysteine-containing Linker 2 and a cysteine-containing Heterodimer-Promoting Domain are used. The Heterodimer-Promoting Domains may be GVEPKSC (SEQ ID NO:96) or VEPKSC (SEQ ID NO:97) or AEPKSC (SEQ ID NO:98) on one polypeptide chain and GFNRGEC (SEQ ID NO:99) or FNRGEC (SEQ ID NO:100) on the other polypeptide chain (US2007/0004909). More preferably, however, the Heterodimer-Promoting Domains of such diabodies are formed from one, two, three or four tandemly repeated coil domains of opposing charge that comprise a sequence of at least six, at least seven or at least eight amino acid residues such that the Heterodimer-Promoting Domain possesses a net charge (Apostolovic, B. et al. (2008) “pH-Sensitivity of the E3/K3Heterodimeric Coiled Coil,” Biomacromolecules 9:3173-3180; Arndt, K. M. et al. (2001) “Helix-stabilized Fv(hsFv)Antibody Fragments: Substituting the Constant Domains of a Fab Fragment for a Heterodimeric Coiled-coil Domain,” J. Molec. Biol. 312:221-228; Arndt, K. M. et al. (2002) “Comparison of In Vivo Selection and Rational Design of Heterodimeric Coiled Coils,” Structure 10:1235-1248; Boucher, C. et al. (2010) “Protein Detection By Western Blot Via Coiled-Coil Interactions,” Analytical Biochemistry 399:138-140; Cachia, P. J. et al. (2004) “Synthetic Peptide Vaccine Development: Measurement Of Polyclonal Antibody Affinity And Cross-Reactivity Using A New Peptide Capture And Release System For Surface Plasmon Resonance Spectroscopy,” J. Mol. Recognit. 17:540-557; De Crescenzo, G. D. et al. (2003) “Real-Time Monitoring of the Interactions of Two-Stranded de novo Designed Coiled-Coils: Effect of Chain Length on the Kinetic and Thermodynamic Constants of Binding,” Biochemistry 42:1754-1763; Fernandez-Rodriquez, J. et al. (2012) “Induced Heterodimerization And Purification Of Two Target Proteins By A Synthetic Coiled-Coil Tag,” Protein Science 21:511-519; Ghosh, T. S. et al. (2009) “End-To-End And End-To-Middle Interhelical Interactions: New Classes Of Interacting Helix Pairs In Protein Structures,” Acta Crystallographica D65:1032-1041; Grigoryan, G. et al. (2008) “Structural Specificity In Coiled-Coil Interactions,” Curr. Opin. Struc. Biol. 18:477-483; Litowski, J. R. et al. (2002) “Designing Heterodimeric Two-Strandedα-Helical Coiled-Coils: The Effects Of Hydrophobicity And a-Helical Propensity On Protein Folding, Stability, And Specificity,” J. Biol. Chem. 277:37272-37279; Steinkruger, J. D. et al. (2012) “The d′--d--d′ Vertical Triad is Less Discriminating Than the a′--a--a′ Vertical Triad in the Antiparallel Coiled-coil Dimer Motif,” J. Amer. Chem. Soc. 134(5):2626-2633; Straussman, R. et al. (2007) “Kinking the Coiled Coil—Negatively Charged Residues at the Coiled-coil Interface,” J. Molec. Biol. 366:1232-1242; Tripet, B. et al. (2002) “Kinetic Analysis of the Interactions between Troponin C and the C-terminal Troponin I Regulatory Region and Validation of a New Peptide Delivery/Capture System used for Surface Plasmon Resonance,” J. Molec. Biol. 323:345-362; Woolfson, D. N. (2005) “The Design Of Coiled-Coil Structures And Assemblies,” Adv. Prot. Chem. 70:79-112; Zeng, Y. et al. (2008) “A Ligand-Pseudoreceptor System Based On de novo Designed Peptides For The Generation Of Adenoviral Vectors With Altered Tropism,” J. Gene Med. 10:355-367). Such repeated coil domains may be exact repeats or may have substitutions. For example, the coil domain of the Heterodimer-Promoting Domain of the first polypeptide chain may comprise a sequence of eight amino acid residues selected to confer a negative charge to such Heterodimer-Promoting Domain, and the coil domain of the Heterodimer-Promoting Domain of the second polypeptide chain may comprise a sequence of eight amino acid residues selected to confer a positive charge to such Heterodimer-Promoting Domain. It is immaterial which coil is provided to the first or second polypeptide chains, provided that, when both polypeptide chains are provided with such Heterodimer-Promoting Domains, a coil of opposite charge is used for the other polypeptide chain. The positively charged amino acid may be lysine, arginine, histidine, etc. and/or the negatively charged amino acid may be glutamic acid, aspartic acid, etc. The positively charged amino acid is preferably lysine and/or the negatively charged amino acid is preferably glutamic acid. It is possible for only a single Heterodimer-Promoting Domain to be employed (since such domain will inhibit homodimerization and thereby promote heterodimerization), however, it is preferred for both the first and second polypeptide chains of the diabodies of the present invention to contain Heterodimer-Promoting Domains. In a preferred embodiment, one of the Heterodimer-Promoting Domains will comprise four tandem “E-coil” helical domains (SEQ ID NO:101:EVAALEK-EVAALEK-EVAALEK-EVAALEK), whose glutamate residues will form a negative charge at pH 7, while the other of the Heterodimer-Promoting Domains will comprise four tandem “K-coil” helical domains (SEQ ID NO:102:KVAALKE-KVAALKE-KVAALKE-KVAALKE), whose lysine residues will forma positive charge at pH 7. The presence of such charged domains promotes association between the first and second polypeptides, and thus fosters heterodimer formation. Especially preferred is a Heterodimer-Promoting Domain in which one of the four tandem “E-coil” helical domains of SEQ ID NO:101 has been modified to contain a cysteine residue:EVAACEK-EVAALEK-EVAALEK-EVAALEK (SEQ ID NO:103). Likewise, especially preferred is a Heterodimer-Promoting Domain in which one of the four tandem “K-coil” helical domains of SEQ ID NO:102 has been modified to contain a cysteine residue:KVAACKE-KVAALKE-KVAALKE-KVAALKE (SEQ ID NO:104). As disclosed in WO 2012/018687, in order to improve the in vivo pharmacokinetic properties of diabodies, a diabody may be modified to contain a polypeptide portion of a serum-binding protein at one or more of the termini of the diabody. Most preferably, such polypeptide portion of a serum-binding protein will be installed at the C-terminus of the diabody. Albumin is the most abundant protein in plasma and has a half-life of 19 days in humans. Albumin possesses several small molecule binding sites that permit it to non-covalently bind to other proteins and thereby extend their serum half-lives. The Albumin-Binding Domain 3 (ABD3) of protein G ofStreptococcusstrain G148 consists of 46 amino acid residues forming a stable three-helix bundle and has broad albumin-binding specificity (Johansson, M. U. et al. (2002) “Structure, Specificity, And Mode Of Interaction For Bacterial Albumin-Binding Modules,” J. Biol. Chem. 277(10):8114-8120. Thus, a particularly preferred polypeptide portion of a serum-binding protein for improving the in vivo pharmacokinetic properties of a diabody is the Albumin-Binding Domain (ABD) from streptococcal protein G, and more preferably, the Albumin-Binding Domain 3 (ABD3) of protein G ofStreptococcusstrain G148 (SEQ ID NO:105): LAEAKVLANR ELDKYGVSKY YKNLIDNAKS AEGVKALIDE ILAALP. As disclosed in WO 2012/162068 (herein incorporated by reference), “deimmunized” variants of SEQ ID NO:105 have the ability to attenuate or eliminate MHC class II binding. Based on combinational mutation results, the following combinations of substitutions are considered to be preferred substitutions for forming such a deimmunized ABD: 66D/70S+71A; 66S/70S+71A; 66S/70S+79A; 64A/65A/71A; 64A/65A/71A+66S; 64A/65A/71A+66D; 64A/65A/71A+66E; 64A/65A/79A+66S; 64A/65A/79A+66D; 64A/65A/79A+66E. Variant ABDs having the modifications L64A, I65A and D79A or the modifications N66S, T70S and D79A. Variant deimmunized ABD having the amino acid sequence: (SEQ ID NO: 106)LAEAKVLANR ELDKYGVSDY YKNLID66NAKS70A71EGVKALIDEILAALP, or the amino acid sequence: (SEQ ID NO: 107)LAEAKVLANR ELDKYGVSDY YKNA64A65NNAKT VEGVKALIA79EILAALP, or the amino acid sequence: (SEQ ID NO: 108)LAEAKVLANR ELDKYGVSDY YKNLIS66NAKS70VEGVKALIA79EILAALP, are particularly preferred as such deimmunized ABD exhibit substantially wild-type binding while providing attenuated MHC class II binding. Thus, the first polypeptide chain of such a diabody having an ABD contains a third linker (Linker 3) preferably positioned C-terminally to the E-coil (or K-coil) Domain of such polypeptide chain so as to intervene between the E-coil (or K-coil) Domain and the ABD (which is preferably a deimmunized ABD). A preferred sequence for such Linker 3 is SEQ ID NO:90: GGGS. 2. Bispecific Diabodies Containing Fc Regions One embodiment of the present invention relates to bispecific diabodies comprising an Fc Region capable of simultaneously binding to LAG-3 and a second epitope (e.g. B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, MHC class I or II, OX40, PD-1, PD-L1, TCR, TIM-3, etc.). The addition of an IgG CH2-CH3 Domain to one or both of the diabody polypeptide chains, such that the complexing of the diabody chains results in the formation of an Fc Region, increases the biological half-life and/or alters the valency of the diabody. Incorporating an IgG CH2-CH3 Domain onto both of the diabody polypeptides will permit a two-chain bispecific Fc-Region-containing diabody to form (FIG.2). Alternatively, incorporating an IgG CH2-CH3 Domain onto only one of the diabody polypeptides will permit a more complex four-chain bispecific Fc Region-containing diabody to form (FIGS.3A-3C).FIG.3Cshows a representative four-chain diabody possessing the Constant Light (CL) Domain and the Constant Heavy CH1 Domain, however fragments of such domains as well as other polypeptides may alternatively be employed (see, e.g.,FIGS.3A and3B, United States Patent Publications No. 2013-0295121; 2010-0174053 and 2009-0060910; European Patent Publication No. EP 2714079; EP 2601216; EP 2376109; EP 2158221 and PCT Publications No. WO 2012/162068; WO 2012/018687; WO 2010/080538). Thus, for example, in lieu of the CH1 Domain, one may employ a peptide having the amino acid sequence GVEPKSC (SEQ ID NO:96)VEPKSC (SEQ ID NO:97), or AEPKSC (SEQ ID NO:98), derived from the hinge domain of a human IgG, and in lieu of the CL Domain, one may employ the C-terminal 6 amino acids of the human kappa light chain, GFNRGEC (SEQ ID NO:99) or FNRGEC (SEQ ID NO:100). A representative peptide containing four-chain diabody is shown inFIG.3A. Alternatively, or in addition, one may employ a peptide comprising tandem coil domains of opposing charge such as the “E-coil” helical domains (SEQ ID NO:101:EVAALEK-EVAALEK-EVAALEK-EVAALEK or SEQ ID NO:103:EVAACEK-EVAALEK-EVAALEK-EVAALEK); and the “K-coil” domains (SEQ ID NO:102:KVAALKE-KVAALKE-KVAALKE-KVAALKE or SEQ ID NO:104:KVAACKE-KVAALKE-KVAALKE-KVAALKE). A representative coil domain containing four-chain diabody is shown inFIG.3B. The Fc Region-containing diabody molecules of the present invention generally include additional intervening linker peptides (Linkers). Typically, the additional Linkers will comprise 3-20 amino acid residues. Additional or alternative linkers that may be employed in the Fc Region-containing diabody molecules of the present invention include: GGGS (SEQ ID NO:90), LGGGSG (SEQ ID NO:91), GGGSGGGSGGG (SEQ ID NO:92), ASTKG (SEQ ID NO:93), DKTHTCPPCP (SEQ IDNO:109), LEPKSS (SEQ IDNO:94), APSSS (SEQ ID NO:95), and APSSSPME (SEQ ID NO:110), LEPKSADKTHTCPPC (SEQ ID NO:111), GGC, and GGG. SEQ ID NO:94 may be used in lieu of GGG or GGC for ease of cloning. Additionally, the amino acid GGG, or SEQ ID NO:94 may be immediately followed by SEQ ID NO:109 to form the alternate linkers: GGGDKTHTCPPCP (SEQ ID NO:112); and LEPKSSDKTHTCPPCP; (SEQ ID NO:113). Fc Region-containing diabody molecule of the present invention may incorporate an IgG hinge region in addition to or in place of a linker. Exemplary hinge regions include: EPKSCDKTHTCPPCP (SEQ ID NO:114) from IgG1, ERKCCVECPPCP (SEQ ID NO:115) from IgG2, ESKYGPPCPSCP (SEQ ID NO:116) from IgG4, and ESKYGPPCPPCP (SEQ ID NO:117) an IgG4 hinge variant comprising a stabilizing substitute to reduce strand exchange. As provided inFIG.3A-3C, diabodies of the invention may comprise four different chains. The first and third polypeptide chains of such a diabody contain three domains: (i) a VL1-containing Domain, (ii) a VH2-containing Domain, (iii) Heterodimer-Promoting Domain and (iv) a Domain containing a CH2-CH3 sequence. The second and fourth polypeptide chains contain: (i) a VL2-containing Domain, (ii) a VH1-containing Domain and (iii) a Heterodimer-Promoting Domain, where the Heterodimer-Promoting Domains promote the dimerization of the first/third polypeptide chains with the second/fourth polypeptide chains. The VL and/or VH Domains of the third and fourth polypeptide chains, and VL and/or VH Domains of the first and second polypeptide chains may be the same or different so as to permit tetravalent binding that is either monospecific, bispecific or tetraspecific. The notation “VL3” and “VH3” denote respectively, the Variable Light Chain Domain and Variable Heavy Chain Domain that bind the “third” epitope of such diabody (“Epitope 3”). Similarly, the notation “VL4” and “VH4” denote respectively, the Variable Light Chain Domain and Variable Heavy Chain Domain that bind the “fourth” epitope of such diabody (“Epitope 4”). The general structure of the polypeptide chains of a representative four-chain Fc Region-containing diabodies of invention is provided in Table 2: TABLE 2Bispecific2ndChainNH2-VL2-VH1-HPD-COOH1stChainNH2-VL1-VH2-HPD-CH2-CH3-COOH1stChainNH2-VL1-VH2-HPD-CH2-CH3-COOH2ndChainNH2-VL2-VH1-HPD-COOHTetra-2ndChainNH2-VL2-VH1-HPD-COOHspecific1stChainNH2-VL1-VH2-HPD-CH2-CH3-COOH3rdChainNH2-VL3-VH4-HPD-CH2-CH3-COOH4thChainNH2-VL4-VH3-HPD-COOHHPD = Heterodimer-Promoting Domain In a specific embodiment, diabodies of the present invention are bispecific, tetravalent (i.e., possess four epitope-binding sites), Fc-containing diabodies (FIGS.3A-3C) that are composed of four total polypeptide chains. The bispecific, tetravalent, Fc-containing diabodies of the invention comprise two epitope-binding sites immunospecific for LAG-3 (which may be capable of binding to the same epitope of LAG-3 or to different epitopes of LAG-3), and two epitope-binding sites specific for a second epitope (e.g., B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, MHC class I or II, OX40, PD-1, PD-L1, TCR, TIM-3, etc.). In a further embodiment, the bispecific Fc Region-containing diabodies may comprise three polypeptide chains. The first polypeptide of such a diabody contains three domains: (i) a VL1-containing Domain, (ii) a VH2-containing Domain and (iii) a Domain containing a CH2-CH3 sequence. The second polypeptide of such diabodies contains: (i) a VL2-containing Domain, (ii) a VH1-containing Domain and (iii) a Domain that promotes heterodimerization and covalent bonding with the diabody's first polypeptide chain. The third polypeptide of such diabodies comprises a CH2-CH3 sequence. Thus, the first and second polypeptide chains of such diabodies associate together to form a VL1/VH1 binding site that is capable of binding to the first epitope, as well as a VL2/VH2 binding site that is capable of binding to the second epitope. The first and second polypeptides are bonded to one another through a disulfide bond involving cysteine residues in their respective Third Domains. Notably, the first and third polypeptide chains complex with one another to form an Fc Region that is stabilized via a disulfide bond. Such diabodies have enhanced potency.FIGS.4A and4Billustrate the structures of such diabodies. Such Fc-Region-containing bispecific diabodies may have either of two orientations (Table 3): TABLE 3First3rdChainNH2-CH2-CH3-COOHOrientation1stChainNH2-VL1-VH2-HPD-CH2-CH3-COOH2ndChainNH2-VL2-VH1-HPD-COOHSecond3rdChainNH2-CH2-CH3-COOHOrientation1stChainNH2-CH2-CH3-VL1-VH2-HPD-COOH2ndChainNH2-VL2-VH1-HPD-COOHHPD = Heterodimer-Promoting Domain In a specific embodiment, diabodies of the present invention are bispecific, bivalent (i.e., possess two epitope-binding sites), Fc-containing diabodies (FIGS.4A-4B) that are composed of three total polypeptide chains. The bispecific, bivalent Fc-containing diabodies of the invention comprise one epitope-binding site immunospecific for LAG-3, and one epitope-binding site specific for a second epitope (e.g., B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3 MHC class I or II, OX40, PD-L1, TCR, TIM-3, etc.). In a further embodiment, the bispecific Fc Region-containing diabodies may comprise a total of five polypeptide chains. In a particular embodiment, two of said five polypeptide chains have the same amino acid sequence. The first polypeptide chain of such diabodies contains: (i) a VH1-containing domain, (ii) a CH-containing domain, and (iii) a Domain containing a CH2-CH3 sequence. The first polypeptide chain may be the heavy chain of an antibody that contains a VH1 and a heavy chain constant region. The second and fifth polypeptide chains of such diabodies contain: (i) a VL1-containing domain, and (ii) a CL-containing domain. The second and/or fifth polypeptide chains of such diabodies may be light chains of an antibody that contains a VL1 complementary to the VH1 of the first/third polypeptide chain. The first, second and/or fifth polypeptide chains may be isolated from naturally occurring antibodies. Alternatively, they may be constructed recombinantly. The third polypeptide chain of such diabodies contains: (i) a VH1-containing domain, (ii) a CH-containing domain, (iii) a Domain containing a CH2-CH3 sequence, (iv) a VL2-containing Domain, (v) a VH3-containing Domain and (vi) a Heterodimer-Promoting Domain, where the Heterodimer-Promoting Domains promote the dimerization of the third chain with the fourth chain. The fourth polypeptide of such diabodies contains: (i) a VL3-containing Domain, (ii) a V12-containing Domain and (iii) a Domain that promotes heterodimerization and covalent bonding with the diabody's third polypeptide chain. Thus, the first and second, and the third and fifth, polypeptide chains of such diabodies associate together to form two VL1/VH1 binding sites capable of binding a first epitope. The third and fourth polypeptide chains of such diabodies associate together to form a VL2/VH2 binding site that is capable of binding to a second epitope, as well as a VL3/VH3 binding site that is capable of binding to a third epitope. The first and third polypeptides are bonded to one another through a disulfide bond involving cysteine residues in their respective constant regions. Notably, the first and third polypeptide chains complex with one another to form an Fc Region. Such diabodies have enhanced potency.FIG.5illustrates the structure of such diabodies. It will be understood that the VL1/VH1, VL2/VH2, and VL3/VH3 Domains may be the same or different so as to permit binding that is monospecific, bispecific or trispecific. However, as provided herein, these domains are preferably selected so as to bind LAG-3 and a second epitope (or a second and a third epitope (e.g., B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3 MHC class I or II, OX40, PD-L1, TCR, TIM-3, etc.). The second and third epitope may be different epitopes of the same antigen molecule, or may be epitopes of different antigen molecules. Such aspects of the invention are discussed in detail below. Thus, the VL and VH Domains of the polypeptide chains are selected so as to form VL/VH binding sites specific for the desired epitopes. The VL/VH binding sites formed by the association of the polypeptide chains may be the same or different so as to permit tetravalent binding that is monospecific, bispecific, trispecific or tetraspecific. In particular, the VL and VH Domains may be selected such that a bispecific diabody may comprise two binding sites for a first epitope and two binding sites for a second epitope, or three binding sites for a first epitope and one binding site for a second epitope, or two binding sites for a first epitope, one binding site for a second epitope and one binding site for a third epitope (as depicted inFIG.5). The general structure of the polypeptide chains of representative five-chain Fc Region-containing diabodies of invention is provided in Table 4: TABLE 4Bispecific2ndChainNH2-VL1-CL-COOH(2 × 2)1stChainNH2-VH1-CH1-CH2-CH3-COOH3rdChainNH2-VH1-CH1-CH2-CH3-VL2-VH2-HPD-COOH5ndChainNH2-VL1-CL-COOH4thChainNH2-VL2-VH2-HPD-COOHBispecific2ndChainNH2-VL1-CL-COOH(3 × 1)1stChainNH2-VH1-CH1-CH2-CH3-COOH3rdChainNH2-VH1-CH1-CH2-CH3-VL1-VH2-HPD-COOH5ndChainNH2-VL1-CL-COOH4thChainNH2-VL2-VH1-HPD-COOHTrispecific2ndChainNH2-VL1-CL-COOH(2 × 1 × 1)1stChainNH2-VH1-CH1-CH2-CH3-COOH3rdChainNH2-VH1-CH1-CH2-CH3-VL2-VH3-HPD-COOH5ndChainNH2-VL1-CL-COOH4thChainNH2-VL3-VH2-HPD-COOHHPD = Heterodimer-Promoting Domain In a specific embodiment, diabodies of the present invention are bispecific, tetravalent (i.e., possess four epitope-binding sites), Fc-containing diabodies that are composed of five total polypeptide chains having two binding sites for a first epitope and two binding sites for a second epitope. In one embodiment, the bispecific, tetravalent, Fc-containing diabodies of the invention comprise two epitope-binding sites immunospecific for LAG-3 (which may be capable of binding to the same epitope of LAG-3 or to different epitopes of LAG-3), and two epitope-binding sites specific for a second epitope (e.g., B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3 MIC class I or II, OX40, PD-L1, TCR, TIM-3, etc.). In another embodiment, the bispecific, tetravalent, Fc-containing diabodies of the invention comprise three epitope-binding sites immunospecific for LAG-3 (which may be capable of binding to the same epitope of LAG-3 or to different epitopes of LAG-3), and one epitope-binding sites specific for a second epitope (e.g., B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3 MHC class I or II, OX40, PD-L1, TCR, TIM-3, etc.). In another embodiment, the bispecific, tetravalent, Fc-containing diabodies of the invention comprise one epitope-binding sites immunospecific for LAG-3, and three epitope-binding sites specific for a second epitope (e.g., B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3 MHC class I or II, OX40, PD-L1, TCR, TIM-3, etc.). 3. Bispecific Trivalent Binding Molecules Containing Fc Regions A further embodiment of the present invention relates to bispecific, trivalent binding molecules, comprising an Fc Region, and being capable of simultaneously binding to a first epitope, a second epitope and a third epitope, wherein at least one of such epitopes is not identical to another of such epitopes. Such bispecific diabodies thus comprise “VL1”/“VH1” domains that are capable of binding to the first epitope, “VL2”/“VH2” domains that are capable of binding to the second epitope and “VL3”/“VH3” domains that are capable of binding to the third epitope. In one embodiment, one or two of such epitopes is an epitope of LAG-3 and another (or the other) of such epitopes is not an epitope of LAG-3 (for example, an epitope of B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3, MHC class I or II, OX40, PD-1, PD-L1, TCR, TIM-3, etc.). Such bispecific trivalent binding molecules comprise three epitope-binding sites, two of which are diabody-type binding domains, which provide binding Site A and binding Site B, and one of which is a non-diabody-type binding domain, which provides binding Site C (see, e.g.,FIGS.6A-6F, and PCT Application No: PCT/US15/33081; and PCT/US15/33076). Typically, the trivalent binding molecules of the present invention will comprise four different polypeptide chains (seeFIGS.6A-6B), however, the molecules may comprise fewer or greater numbers of polypeptide chains, for example, by fusing such polypeptide chains to one another (e.g., via a peptide bond) or by “dividing” such polypeptide chains to form additional polypeptide chains, or by associating fewer or additional polypeptide chains via disulfide bonds.FIGS.6B-6Fillustrate this aspect of the present invention by schematically depicting such molecules having three polypeptide chains. As provided inFIGS.6A-6F, the trivalent binding molecules of the present invention may have alternative orientations in which the diabody-type binding domains are N-terminal (FIGS.6A,6C and6D) or C-terminal (FIGS.6B,6E and6F) to an Fc Region. In certain embodiments, the first polypeptide chain of such trivalent binding molecules of the present invention contains: (i) a VL1-containing Domain, (ii) a VH2-containing Domain, (iii) a Heterodimer-Promoting Domain, and (iv) a Domain containing a CH2-CH3 sequence. The VL1 and VL2 Domains are located N-terminal or C-terminal to the CH2-CH3-containing domain as presented in Table 5 (FIGS.6A and6B). The second polypeptide chain of such embodiments contains: (i) a VL2-containing Domain, (ii) a VH1-containing Domain, and (iii) a Heterodimer-Promoting Domain. The third polypeptide chain of such embodiments contains: (i) a VH3-containing Domain, (ii) a CH1-containing Domain and (iii) a Domain containing a CH2-CH3 sequence. The third polypeptide chain may be the heavy chain of an antibody that contains a VH3 and a heavy chain constant region. The fourth polypeptide of such embodiments contains: (i) a VL3-containing Domain and (ii) a CL-containing Domain. The fourth polypeptide chains may be light chain of an antibody that contains a VL3 complementary to the VH3 of the third polypeptide chain. The third or fourth polypeptide chains may be isolated from naturally occurring antibodies. Alternatively, they may be constructed recombinantly, synthetically or by other means. The Variable Light Chain Domain of the first and second polypeptide chains are separated from the Variable Heavy Chain Domains of such polypeptide chains by an intervening spacer linker having a length that is too short to permit their VL1/VH2 (or their VL2/VH1) domains to associate together to form epitope-binding site capable of binding to either the first or second epitope. A preferred intervening spacer peptide (Linker 1) for this purpose has the sequence (SEQ ID NO:14): GGGSGGGG. Other Domains of the trivalent binding molecules may be separated by one or more intervening spacer peptides, optionally comprising a cysteine residue. Exemplary linkers useful for the generation of trivalent binding molecules are provided herein and are also provided in PCT Application Nos: PCT/US15/33081; and PCT/US15/33076. Thus, the first and second polypeptide chains of such trivalent binding molecules associate together to form a VL1/VH1 binding site capable of binding a first epitope, as well as a VL2/VH2 binding site that is capable of binding to a second epitope. The third and fourth polypeptide chains of such trivalent binding molecules associate together to form a VL3/VH3 binding site that is capable of binding to a third epitope. As described above, the trivalent binding molecules of the present invention may comprise three polypeptides. Trivalent binding molecules comprising three polypeptide chains may be obtained by linking the domains of the fourth polypeptide N-terminal to the VH3-containing Domain of the third polypeptide. Alternatively, a third polypeptide chain of a trivalent binding molecule of the invention containing the following three domains is utilized: (i) a VL3-containing Domain, (ii) a VH3-containing Domain, and (iii) a Domain containing a CH2-CH3 sequence, wherein the VL3 and VH3 are spaced apart from one another by an intervening spacer peptide that is sufficiently long (at least 9 or more amino acid residues) so as to allow the association of these domains to form an epitope-binding site. It will be understood that the VL1/VH1, VL2/VH2, and VL3/VH3 Domains of such diabody molecules may be the same or different so as to permit binding that is monospecific, bispecific or trispecific. However, as provided herein, these domains are preferably selected so as to bind LAG-3 and a second epitope (or a second and third epitope) (preferably, such epitopes are epitopes of B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3 MHC class I or II, OX40, PD-L1, TCR, TIM-3, etc.). In particular, the VL and VH Domains may be selected such that a trivalent binding molecule comprises two binding sites for a first epitope and one binding sites for a second epitope, or one binding site for a first epitope and two binding sites for a second epitope, or one binding site for a first epitope, one binding site for a second epitope and one binding site for a third epitope. The general structure of the polypeptide chains of representative trivalent binding molecules of invention is provided inFIGS.6A-6Fand in Table 5: TABLE 5Four Chain2ndChainNH2-VL2-VH1-HPD-COOH1st1stChainNH2-VL1-VH2-HPD-CH2-CH3-COOHOrientation3rdChainNH2-VH3-CH1-CH2-CH3-COOH4thChainNH2-VL3-CL-COOHFour Chain2ndChainNH2-VL2-VH1-HPD-COOH2nd1stChainNH2-CH2-CH3-VL1-VH2-HPD COOHOrientation3rdChainNH2-VH3-CH1-CH2-CH3-COOH4thChainNH2-VL3-CL-COOHThree Chain2ndChainNH2-VL2-VH1-HPD-COOH1st1stChainNH2-VL1-VH2-HPD-CH2-CH3-COOHOrientation3rdChainNH2-VL3-VH3-HPD-CH2-CH3-COOHThree Chain2ndChainNH2-VL2-VH1-HPD-COOH2nd1stChainNH2-CH2-CH3-VL1-VH2-HPD COOHOrientation3rdChainNH2-VL3-VH3-HPD-CH2-CH3-COOHHPD = Heterodimer-Promoting Domain One embodiment of the present invention relates to bispecific trivalent binding molecules that comprise two epitope-binding sites for LAG-3 and one epitope-binding site for the second epitope present on a molecule other than LAG-3 (e.g. B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3, MHC class I or II, OX40, PD-L1, TCR, TIM-3, etc.). The two epitope-binding sites for LAG-3 may bind the same epitope or different epitopes. Another embodiment of the present invention relates to bispecific trivalent binding molecules that comprise, one epitope-binding site for LAG-3 and two epitope-binding sites that bind a second antigen present on a molecule other than LAG-3 (e.g. B7-H3, B7-H4, BTLA, CD40, CD80, CD86, CD137, CTLA-4, ICOS, KIR, LAG-3, MHC class I or II, OX40, PD-L1, TCR, TIM-3, etc.). The two epitope-binding sites for the second antigen may bind the same epitope or different epitopes of the antigen (e.g., the same or different epitopes of LAG-3). As provided above, such bispecific trivalent binding molecules may comprise three or four polypeptide chains. VII. Constant Domains and Fc Regions Provided herein are antibody Constant Domains useful in the generation of LAG-3-binding molecules (e.g., antibodies, diabodies, trivalent binding molecules, etc.) of the invention. A preferred CL Domain is a human IgG CL Kappa Domain. The amino acid sequence of an exemplary human CL Kappa Domain is (SEQ ID NO:118): RTVAAPSVFI FPPSDEQLKS GTASVVCLLN NFYPREAKVQWKVDNALQSG NSQESVTEQD SKDSTYSLSS TLTLSKADYEKHKVYACEVT HQGLSSPVTK SFNRGEC Alternatively, an exemplary CL Domain is a human IgG CL Lambda Domain. The amino acid sequence of an exemplary human CL Kappa Domain is (SEQ ID NO:119): QPKAAPSVTL FPPSSEELQA NKATLVCLIS DFYPGAVTVAWKADSSPVKA GVETTPSKQS NNKYAASSYL SLTPEQWKSHRSYSCQVTHE GSTVEKTVAP TECS As provided herein, the LAG-3-binding molecules of the invention may comprise an Fc Region. The Fc Region of such molecules of the invention may be of any isotype (e.g., IgG1, IgG2, IgG3, or IgG4). The LAG-3-binding molecules of the invention may further comprise a CH1 Domain and/or a hinge region. When present, the CH1 Domain and/or hinge region may be of any isotype (e.g., IgG1, IgG2, IgG3, or IgG4), and is preferably of the same isotype as the desired Fc Region. An exemplary CH1 Domain is a human IgG1 CH Domain. The amino acid sequence of an exemplary human IgG1 CH Domain is (SEQ ID NO:120): ASTKGPSVFP LAPSSKSTSG GTAALGCLVK DYFPEPVTVSWNSGALTSGV HTFPAVLQSS GLYSLSSVVT VPSSSLGTQTYICNVNHKPS NTKVDKRV An exemplary CH1 Domain is a human IgG2CH Domain. The amino acid sequence of an exemplary human IgG2CH Domain is (SEQ ID NO:121): ASTKGPSVFP LAPCSRSTSE STAALGCLVK DYFPEPVTVSWNSGALTSGV HTFPAVLQSS GLYSLSSVVT VPSSNFGTQTYTCNVDHKPS NTKVDKTV An exemplary CH1 Domain is a human IgG4CH1 Domain. The amino acid sequence of an exemplary human IgG4CH1 Domain is (SEQ ID NO:122): ASTKGPSVFP LAPCSRSTSE STAALGCLVK DYFPEPVTVSWNSGALTSGV HTFPAVLQSS GLYSLSSVVT VPSSSLGTKTYTCNVDHKPS NTKVDKRV One exemplary hinge region is a human IgG1 hinge region. The amino acid sequence of an exemplary human IgG1 hinge region is (SEQ ID NO:114): EPKSCDKTHTCPPCP. Another exemplary hinge region is a human IgG2 hinge region. The amino acid sequence of an exemplary human IgG2 hinge region is (SEQ ID NO:115): ERKCCVECPPCP. Another exemplary hinge region is a human IgG4 hinge region. The amino acid sequence of an exemplary human IgG4 hinge region is (SEQ ID NO:116): ESKYGPPCPSCP. As described herein, an IgG4 hinge region may comprise a stabilizing mutation such as the S228P substitution. The amino acid sequence of an exemplary stabilized IgG4 hinge region is (SEQ ID NO:117): ESKYGPPCPPCP. The Fc Region of the Fc Region-containing molecules (e.g., antibodies, diabodies, and trivalent molecules) of the present invention may be either a complete Fc Region (e.g., a complete IgG Fc Region) or only a fragment of an Fc Region. Optionally, the Fc Region of the Fc Region-containing molecules of the present invention lacks the C-terminal lysine amino acid residue. In particular, the Fc Region of the Fc Region-containing molecules of the present invention may be an engineered variant Fc Region. Although the Fc Region of the bispecific Fc Region-containing molecules of the present invention may possess the ability to bind to one or more Fc receptors (e.g., FcγR(s)), more preferably such variant Fc Region will have altered binding to FcγRIA (CD64), FcγRIIA (CD32A), FcγRIIB (CD32B), FcγRIIIA (CD16a) or FcγRIIIB (CD16b) (relative to the binding exhibited by a wild-type Fc Region) or will have substantially reduced or no ability to bind to inhibitory receptor(s). Thus, the Fc Region of the Fc Region-containing molecules of the present invention may include some or all of the CH2 Domain and/or some or all of the CH3 Domain of a complete Fc Region, or may comprise a variant CH2 and/or a variant CH3 sequence (that may include, for example, one or more insertions and/or one or more deletions with respect to the CH2 or CH3 domains of a complete Fc Region). Such Fc Regions may comprise non-Fc polypeptide portions, or may comprise portions of non-naturally complete Fc Regions, or may comprise non-naturally occurring orientations of CH2 and/or CH3 Domains (such as, for example, two CH2 domains or two CH3 domains, or in the N-terminal to C-terminal direction, a CH3 Domain linked to a CH2 Domain, etc.). Fe Region modifications identified as altering effector function are known in the art, including modifications that increase binding to activating receptors (e.g., FcγRIIA (CD16A) and reduce binding to inhibitory receptors (e.g., FcγRIIB (CD32B) (see, e.g., Stavenhagen, J. B. et al. (2007) “Fc Optimization Of Therapeutic Antibodies Enhances Their Ability To Kill Tumor Cells In Vitro And Controls Tumor Expansion In Vivo Via Low-Affinity Activating Fcgamma Receptors,” Cancer Res. 57(18):8882-8890). Exemplary variants of human IgG1 Fc Regions with reduced binding to CD32B and/or increased binding to CD16A contain F243L, R292P, Y300L, V305I or P296L substitutions. These amino acid substitutions may be present in a human IgG1 Fc Region in any combination or subcombination. In one embodiment, the human IgG1 Fc Region variant contains a F243L, R292P and Y300L substitution. In another embodiment, the human IgG1 Fc Region variant contains F243L, R292P, Y300L, V305I and P296L substitutions. In particular, it is preferred for the CH2-CH3 Domains of the polypeptide chains of the Fc Region-containing molecules of the present invention to exhibit decreased (or substantially no) binding to FcγRIA (CD64), FcγRIIA (CD32A), FcγRIIB (CD32B), FcγRIIIA (CD16a) or FcγRIIIB (CD16b) (relative to the binding exhibited by the wild-type IgG1 Fc Region (SEQ ID NO:1). Variant Fc Regions and mutant forms capable of mediating such altered binding are described above. In a specific embodiment, the Fc Region-containing molecules of the present invention comprise an IgG Fc Region that exhibits reduced ADCC effector function. In a preferred embodiment the CH2-CH3 Domain of the first and/or third polypeptide chains of such molecules include any 1, 2, or 3, of the substitutions: L234A, L235A, N297Q, and N297G. In another embodiment, the human IgG Fc Region variant contains an N297Q substitution, an N297G substitution, L234A and L235A substitutions or a D265A substitution, as these mutations abolish FcR binding. Alternatively, a CH2-CH3 Domain of an Fc Region which inherently exhibits decreased (or substantially no) binding to FcγRIIIA (CD16a) and/or reduced effector function (relative to the binding exhibited by the wild-type IgG1 Fc Region (SEQ ID NO:1)) is utilized. In a specific embodiment, the Fc Region-containing molecules of the present invention comprise an IgG2 Fc Region (SEQ ID NO:2) or an IgG4 Fc Region (SEQ ID NO:4). When an IgG4 Fc Region in utilized, the instant invention also encompasses the introduction of a stabilizing mutation, such as the hinge region S228P substitution described above (see, e.g., SEQ ID NO:117). Since the N297G, N297Q, L234A, L235A and D265A substitutions abolish effector function, in circumstances in which effector function is desired, these substitutions would preferably not be employed. A preferred IgG1 sequence for the CH2 and CH3 Domains of the LAG-3-binding molecules of the invention will have the L234A/L235A substitutions (SEQ ID NO:123): APEAAGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHEDPEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLHQDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYTLPPSREEMTK NQVSLTCLVK GFYPSDIAVE WESNGQPENNYKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHEALHNHYTQKS LSLSPGX wherein, X is a lysine (K) or is absent. In particular, it is preferred for the Fc Regions of the polypeptide chains of the Fc Region-containing molecules of the present invention to exhibit increased serum half-life (relative to the half-life exhibited by the corresponding wild-type Fc). Variant Fc Regions and mutant forms exhibiting extended serum half-life are described above. In a preferred embodiment the CH2-CH3 Domain of the first and/or third polypeptide chains of such Fc Region-containing molecules include any 1, 2, or 3, of the substitutions: M252Y, S254T and T256E. The invention further encompasses Fc Region-containing molecules of the present invention comprising variant Fc Regions comprising:(A) one or more mutations which alter effector function and/or FcγR; and(B) one or more mutations which extend serum half-life. A preferred IgG1 sequence for the CH2 and CH3 Domains of the Fc Region-containing molecules of the present invention will comprise the substitutions L234A/L235A/M252Y/S254T/T256E (SEQ ID NO:124): APEAAGGPSV FLFPPKPKDT LYITREPEVT CVVVDVSHEDPEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLHQDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYTLPPSREEMTK NQVSLTCLVK GFYPSDIAVE WESNGQPENNYKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHEALHNHYTQKS LSLSPGX wherein, X is a lysine (K) or is absent A preferred IgG4 sequence for the CH2 and CH3 Domains of the Fe Region-containing molecules of the present invention will comprise the M252Y/S254T/T256E substitutions (SEQ ID NO:125): APEFLGGPSV FLFPPKPKDT LYITREPEVT CVVVDVSQEDPEVQFNWYVD GVEVHNAKTK PREEQFNSTY RVVSVLTVLHQDWLNGKEYK CKVSNKGLPS SIEKTISKAK GQPREPQVYTLPPSQEEMTK NQVSLTCLVK GFYPSDIAVE WESNGQPENNYKTTPPVLDS DGSFFLYSRL TVDKSRWQEG NVFSCSVMHEALHNHYTQKS LSLSLGX wherein, X is a lysine (K) or is absent For diabodies and trivalent binding molecules whose first and third polypeptide chains are not identical), it is desirable to reduce or prevent homodimerization from occurring between the CH2-CH3 Domains of two first polypeptide chains or between the CH2-CH3 Domains of two third polypeptide chains. The CH2 and/or CH3 Domains of such polypeptide chains need not be identical in sequence, and advantageously are modified to foster complexing between the two polypeptide chains. For example, an amino acid substitution (preferably a substitution with an amino acid comprising a bulky side group forming a “knob”, e.g., tryptophan) can be introduced into the CH2 or CH3 Domain such that steric interference will prevent interaction with a similarly mutated domain and will obligate the mutated domain to pair with a domain into which a complementary, or accommodating mutation has been engineered, i.e., “the hole” (e.g., a substitution with glycine). Such sets of mutations can be engineered into any pair of polypeptides comprising CH2-CH3 Domains that forms an Fc Region. Methods of protein engineering to favor heterodimerization over homodimerization are well known in the art, in particular with respect to the engineering of immunoglobulin-like molecules, and are encompassed herein (see e.g., Ridgway et al. (1996) “Knobs-Into-Holes' Engineering Of Antibody CH3Domains For Heavy Chain Heterodimerization,” Protein Engr. 9:617-621, Atwell et al. (1997) “Stable Heterodimers From Remodeling The Domain Interface Of A Homodimer Using A Phage Display Library,” J. Mol. Biol. 270: 26-35, and Xie et al. (2005) “A New Format Of Bispecific Antibody: Highly Efficient Heterodimerization, Expression And Tumor Cell Lysis,” J. Immunol. Methods 296:95-101; each of which is hereby incorporated herein by reference in its entirety). Preferably the “knob” is engineered into the CH2-CH3 Domains of the first polypeptide chain and the “hole” is engineered into the CH2-CH3 Domains of the third polypeptide chain of diabodies comprising three polypeptide chains. Thus, the “knob” will help in preventing the first polypeptide chain from homodimerizing via its CH2 and/or CH3 Domains. As the third polypeptide chain preferably contains the “hole” substitution it will heterodimerize with the first polypeptide chain as well as homodimerize with itself. This strategy may be utilized for diabodies and trivalent binding molecules comprising three, four or five chains as detailed above, where the “knob” is engineered into the CH2-CH3 Domains of the first polypeptide chain and the “hole” is engineered into the CH2-CH3 Domains of the third polypeptide chain. A preferred knob is created by modifying an IgG Fc Region to contain the modification T366W. A preferred hole is created by modifying an IgG Fc Region to contain the modification T366S, L368A and Y407V. To aid in purifying the hole-bearing third polypeptide chain homodimer from the final bispecific heterodimeric Fc Region-containing molecule, the protein A binding site of the CH2 and CH3 Domains of the third polypeptide chain is preferably mutated by amino acid substitution at position 435 (H435R). Thus, the hole-bearing third polypeptide chain homodimer will not bind to protein A, whereas the bispecific heterodimer will retain its ability to bind protein A via the protein A binding site on the first polypeptide chain. In an alternative embodiment, the hole-bearing third polypeptide chain may incorporate amino acid substitutions at positions 434 and 435 (N434A/N435K). A preferred IgG1 amino acid sequence for the CH2 and CH3 Domains of the first polypeptide chain of an Fc Region-containing molecule of the present invention will have the “knob-bearing” sequence (SEQ ID NO:126): APEAAGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHEDPEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLHQDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYTLPPSREEMTK NQVSLWCLVK GFYPSDIAVE WESNGQPENNYKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHEALHNHYTQKS LSLSPGX wherein, X is a lysine (K) or is absent A preferred IgG1 amino acid sequence for the CH2 and CH3 Domains of the second polypeptide chain of an Fc Region-containing molecule of the present invention having two polypeptide chains (or the third polypeptide chain of an Fc Region-containing molecule having three, four, or five polypeptide chains) will have the “hole-bearing” sequence (SEQ ID NO:127): APEAAGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHEDPEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLHQDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYTLPPSREEMTK NQVSLSCAVK GFYPSDIAVE WESNGQPENNYKTTPPVLDS DGSFFLVSKL TVDKSRWQQG NVFSCSVMHEALHNRYTQKS LSLSPGX wherein, X is a lysine (K) or is absent As will be noted, the CH2-CH3 Domains of SEQ ID NO:126 and SEQ ID NO:127 include a substitution at position 234 with alanine and 235 with alanine, and thus form an Fc Region exhibit decreased (or substantially no) binding to FcγRIA (CD64), FcγRIIA (CD32A), FcγRIIB (CD32B), FcγRIIIA (CD16a) or FcγRIIIB (CD16b) (relative to the binding exhibited by the wild-type Fc Region (SEQ ID NO:1). The invention also encompasses such CH2-CH3 Domains, which comprise alanine residues at positions 234 and/or 235 and/or alternative and/or additional substitutions which modify effector function and/or FγR binding activity of the Fc Region. The invention also encompasses such CH2-CH3 Domains, which further comprise one or more half-live extending amino acid substitutions. In particular, the invention encompasses such hole-bearing and such knob-bearing CH2-CH3 Domains which further comprise the M252Y/S254T/T256E. It is preferred that the first polypeptide chain will have a “knob-bearing” CH2-CH3 sequence, such as that of SEQ ID NO:126. However, as will be recognized, a “hole-bearing” CH2-CH3 Domain (e.g., SEQ ID NO:127) could be employed in the first polypeptide chain, in which case, a “knob-bearing” CH2-CH3 Domain (e.g., SEQ ID NO:126) would be employed in the second polypeptide chain of an Fc Region-containing molecule of the present invention having two polypeptide chains (or the third polypeptide chain of an Fc Region-containing molecule having three, four, or five polypeptide chains). As detailed above the invention encompasses Fe Region-containing molecules (e.g., antibodies and Fc Region-containing diabodies) having wild type CH2 and CH3 Domains, or having CH2 and CH3 Domains comprising combinations of the substitutions described above. An exemplary amino acid sequence of an IgG1 CH2-CH3 Domain encompassing such variations is (SEQ ID NO:128): APEX1X2GGPSV FLFPPKPKDT LX3IX4RX5PEVT CVVVDVSHEDPEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLHQDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYTLPPSREEMTK NQVSLX6CX7VK GFYPSDIAVE WESNGQPENNYKTTPPVLDS DGSFFLX8SKL TVDKSRWQQG NVFSCSVMHEALHX9X10YTQKS LSLSPGX11 wherein:(a) X1and X2are both L (wild type), or are both A (decreased FcγR binding);(b) X3, X4, and X5respectively are M, S and T (wild type), or are Y, T and E (extended half-life),(C) X6, X7, and X8respectively are T, L and Y (wild type), or are W, L and Y (knob), or S, A and V (hole);(d) X9and X10respectively are N and H (wild type), or are N and R (no protein A binding), or A and K (no protein A binding); and(e) X11is K or is absent In other embodiments, the invention encompasses LAG-3-binding molecules comprising CH2 and/or CH3 Domains that have been engineered to favor heterodimerization over homodimerization using mutations known in the art, such as those disclosed in PCT Publication No. WO 2007/110205; WO 2011/143545; WO 2012/058768; WO 2013/06867, all of which are incorporated herein by reference in their entirety. VIII. Reference Antibodies A. Reference Anti-LAG-3 Antibody In order to assess and characterize the novel anti-LAG-3-binding molecules of the present invention, the following reference antibody was employed: 25F7 (BMS-986016, Medarex/BMS, designated herein as “LAG-3 mAb A”). 1. 25F7 (“LAG-3 mAb A”) The amino acid sequence of the VH Domain of 25F7 (“LAG-3 mAb A”) (SEQ ID NO:129) is shown below (CDRHresidues are shown underlined): QVQLQQWGAG LLKPSETLSL TCAVYGGSFSDYYWNWIRQPPGKGLEWIGEINHNGNTNSNPSLKSRVTLS LDTSKNQFSLKLRSVTAADT AVYYCAFGYSDYEYNWFDPW GQGTLVTVSS The amino acid sequence of the VL Domain of 25F7 (“LAG-3 mAb A”) (SEQ ID NO:130) is shown below (CDRLresidues are shown underlined): EIVLTQSPAT LSLSPGERAT LSCRASQSISSYLAWYQQKPGQAPRLLIYDASNRATGIPA RFSGSGSGTD FTLTISSLEPEDFAVYYCQQRSNWPLTFGQ GTNLEIK B. Reference Anti-PD-1 Antibodies In order to assess and characterize the activity of the novel LAG-3-binding molecules of the present invention in combination with an anti-PD-1 antibody a reference antibody may be used. Antibodies that are immunospecific for PD-1 are known (see, e.g., U.S. Pat. Nos. 8,008,449; 8,552,154; PCT Patent Publications WO 2012/135408; WO 2012/145549; and WO 2013/014668) and include: nivolumab (also known as 5C4, BMS-936558, ONO-4538, MDX-1106, and marketed as OPDIVO® by Bristol-Myers Squibb) designated herein as “PD-1 mAb 1;” pembrolizumab (formerly known as lambrolizumab, also known as MK-3475, SCH-900475, and marketed as KEYTRUDA® by Merck) designated herein as “PD-1 mAb 2”; EH12.2H7 (Dana Farber) designated herein as “PD-1 mAb 3”; pidilizumab (also known as CT-011, CureTech) designated herein as “PD-1 mAb 4.” 1. Nivolumab (“PD-1 mAb 1”) The amino acid sequence of the Heavy Chain Variable Domain of PD-1 mAb 1 has the amino acid sequence (SEQ ID NO:131) (CDRHresidues are shown underlined): QVQLVESGGG VVQPGRSLRL DCKASGITFSNSGMHWVRQAPGKGLEWVAVIWYDGSKRYYADSVKGRFTI SRDNSKNTLFLQMNSLRAED TAVYYCATNDDYWGQGTLVT VSS The amino acid sequence of the Light Chain Variable Domain of PD-1 mAb 1 has the amino acid sequence (SEQ ID NO:132) (CDRLresidues are shown underlined): EIVLTQSPAT LSLSPGERAT LSCRASQSVSSYLAWYQQKPGQAPRLLIYDASNRATGIPA RESGSGSGTD FTLTISSLEPEDFAVYYCQQSSNWPRTFGQ GTKVEIK 2. Pembrolizumab (“PD-1 mAb 2”) The amino acid sequence of the Heavy Chain Variable Domain of PD-1 mAb 2 has the amino acid sequence (SEQ ID NO:133) (CDRHresidues are shown underlined): QVQLVQSGVE VKKPGASVKV SCKASGYTFTNYYMYWVRQAPGQGLEWMGGINPSNGGTNFNEKFKNRVTL TTDSSTTTAYMELKSLQFDD TAVYYCARRDYRFDMGFDYW GQGTTVTVSS The amino acid sequence of the Light Chain Variable Domain of PD-1 mAb 2 has the amino acid sequence (SEQ ID NO:134) (CDRLresidues are shown underlined): EIVLTQSPAT LSLSPGERAT LSCRASKGVSTSGYSYLHWYQQKPGQAPRL LIYLASYLESGVPARFSGSG SGTDFTLTISSLEPEDFAVY YCQHSRDLPLTFGGGTKVEIK 3. EH12.2H7 (“PD-1 mAb 3”) The amino acid sequence of the Heavy Chain Variable Domain of PD-1 mAb 3 has the amino acid sequence (SEQ ID NO:135) (CDRHresidues are shown underlined): QVQLQQSGAE LAKPGASVQM SCKASGYSFTSSWIHWVKQRPGQGLEWIGYTYPSTGFTEYNQKFKDKATL TADKSSSTAYMQLSSLTSED SAVYYCARWRDSSGYHAMDYWGQGTSVTVSS The amino acid sequence of the Light Chain Variable Domain of PD-1 mAb 3 has the amino acid sequence (SEQ ID NO:136) (CDRLresidues are shown underlined): DIVLTQSPAS LTVSLGQRAT ISCRASQSVSTSGYSYMHWYQQKPGQPPKL LIKFGSNLESGIPARFSGSG SGTDFTLNIHPVEEEDTATY YCQHSWEIPYTFGGGTKLEI K 4. Pidilizumab (“PD-1 mAb 4”) The amino acid sequence of the Heavy Chain Variable Domain of PD-1 mAb 4 has the amino acid sequence (SEQ ID NO:137) (CDRHresidues are shown underlined): QVQLVQSGSE LKKPGASVKI SCKASGYTFTNYGMNWVRQAPGQGLQWMGWINTDSGESTYAEEFKGRFVF SLDTSVNTAYLQITSLTAED TGMYFCVRVGYDALDYWGQG TLVTVSS The amino acid sequence of the Light Chain Variable Domain of PD-1 mAb 4 has the amino acid sequence (SEQ ID NO:138) (CDRLresidues are shown underlined): EIVLTQSPSS LSASVGDRVT ITCSARSSVSYMHWFQQKPGKAPKLWIYRTSNLASGVPSR FSGSGSGTSY CLTINSLQPEDFATYYCQQRSSFPLTFGGG IX. Methods of Production An anti-LAG-3 polypeptide, and other LAG-3 agonists, antagonists and modulators can be created from the polynucleotides and/or sequences of the LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 antibodies by methods known in the art, for example, synthetically or recombinantly. One method of producing such peptide agonists, antagonists and modulators involves chemical synthesis of the polypeptide, followed by treatment under oxidizing conditions appropriate to obtain the native conformation, that is, the correct disulfide bond linkages. This can be accomplished using methodologies well known to those skilled in the art (see, e.g., Kelley, R. F. et al. (1990) In: GENETICENGINEERINGPRINCIPLES ANDMETHODS, Setlow, J. K. Ed., Plenum Press, N.Y., vol. 12, pp 1-19; Stewart, J. M et al. (1984) SOLIDPHASEPEPTIDESYNTHESIS, Pierce Chemical Co., Rockford, Ill.; see also U.S. Pat. Nos. 4,105,603; 3,972,859; 3,842,067; and 3,862,925). Polypeptides of the invention may be conveniently prepared using solid phase peptide synthesis (Merrifield, B. (1986) “Solid Phase Synthesis,” Science 232(4748):341-347; Houghten, R. A. (1985) “General Method For The Rapid Solid-Phase Synthesis Of Large Numbers Of Peptides: Specificity Of Antigen-Antibody Interaction At The Level Of Individual Amino Acids,” Proc. Natl. Acad. Sci. (U.S.A.) 82(15):5131-5135; Ganesan, A. (2006) “Solid-Phase Synthesis In The Twenty-First Century,” Mini Rev. Med. Chem. 6(1):3-10). In yet another alternative, fully human antibodies having one or more of the CDRs of LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 or which compete with LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 for binding to human LAG-3 or a soluble form thereof may be obtained through the use of commercially available mice that have been engineered to express specific human immunoglobulin proteins. Transgenic animals that are designed to produce a more desirable (e.g., fully human antibodies) or more robust immune response may also be used for generation of humanized or human antibodies. Examples of such technology are XENOMOUSE™ (Abgenix, Inc., Fremont, Calif.) and HUMAB-MOUSE® and TC MOUSE™ (both from Medarex, Inc., Princeton, N.J.). In an alternative, antibodies may be made recombinantly and expressed using any method known in the art. Antibodies may be made recombinantly by first isolating the antibodies made from host animals, obtaining the gene sequence, and using the gene sequence to express the antibody recombinantly in host cells (e.g., CHO cells). Another method that may be employed is to express the antibody sequence in plants {e.g., tobacco) or transgenic milk. Suitable methods for expressing antibodies recombinantly in plants or milk have been disclosed (see, for example, Peeters et al. (2001) “Production Of Antibodies And Antibody Fragments In Plants,” Vaccine 19:2756; Lonberg, N. et al. (1995) “Human Antibodies From Transgenic Mice,” Int. Rev. Immunol 13:65-93; and Pollock et al. (1999) “Transgenic Milk As A Method For The Production Of Recombinant Antibodies,” J. Immunol Methods 231:147-157). Suitable methods for making derivatives of antibodies, e.g., humanized, single-chain, etc. are known in the art. In another alternative, antibodies may be made recombinantly by phage display technology (see, for example, U.S. Pat. Nos. 5,565,332; 5,580,717; 5,733,743; 6,265,150; and Winter, G. et al. (1994) “Making Antibodies By Phage Display Technology,” Annu. Rev. Immunol. 12.433-455). The antibodies or protein of interest may be subjected to sequencing by Edman degradation, which is well known to those of skill in the art. The peptide information generated from mass spectrometry or Edman degradation can be used to design probes or primers that are used to clone the protein of interest. An alternative method of cloning the protein of interest is by “panning” using purified LAG-3 or portions thereof for cells expressing an antibody or protein of interest that possesses one or more of the CDRs LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 or that competes with LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 for binding to human LAG-3. The “panning” procedure may be conducted by obtaining a cDNA library from tissues or cells that express LAG-3, overexpressing the cDNAs in a second cell type, and screening the transfected cells of the second cell type for a specific binding to LAG-3 in the presence or absence of LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6. Detailed descriptions of the methods used in cloning mammalian genes coding for cell surface proteins by “panning” can be found in the art (see, for example, Aruffo, A. et al. (1987) “Molecular Cloning Of A CD28cDNA By A High-Efficiency COS Cell Expression System,” Proc. Natl. Acad. Sci. (U.S.A.) 84:8573-8577 and Stephan, J. et al. (1999) “Selective Cloning Of Cell Surface Proteins Involved In Organ Development: Epithelial Glycoprotein Is Involved In Normal Epithelial Differentiation,” Endocrinol. 140:5841-5854). Vectors containing polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus). The choice of introducing vectors or polynucleotides will often depend on features of the host cell. Any host cell capable of overexpressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest. Non-limiting examples of suitable mammalian host cells include but are not limited to COS, HeLa, and CHO cells. Preferably, the host cells express the cDNAs at a level of about 5-fold higher, more preferably 10-fold higher, even more preferably 20-fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells. Screening the host cells for a specific binding to LAG-3 is effected by an immunoassay or FACS. A cell overexpressing the antibody or protein of interest can be identified. The invention includes polypeptides comprising an amino acid sequence of the antibodies of this invention. The polypeptides of this invention can be made by procedures known in the art. The polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis. Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available. For example, an anti-LAG-3 polypeptide could be produced by an automated polypeptide synthesizer employing the solid phase method. The invention includes variants of LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 antibodies and their polypeptide fragments that bind to LAG-3, including functionally equivalent antibodies and fusion polypeptides that do not significantly affect the properties of such molecules as well as variants that have enhanced or decreased activity. Modification of polypeptides is routine practice in the art and need not be described in detail herein. Examples of modified polypeptides include polypeptides with conservative substitutions of amino acid residues, one or more deletions or additions of amino acids which do not significantly deleteriously change the functional activity, or use of chemical analogs. Amino acid residues that can be conservatively substituted for one another include but are not limited to: glycine/alanine; serine/threonine; valine/isoleucine/leucine; asparagine/glutamine; aspartic acid/glutamic acid; lysine/arginine; and phenylalanine/tyrosine. These polypeptides also include glycosylated and non-glycosylated polypeptides, as well as polypeptides with other post-translational modifications, such as, for example, glycosylation with different sugars, acetylation, and phosphorylation. Preferably, the amino acid substitutions would be conservative, i.e., the substituted amino acid would possess similar chemical properties as that of the original amino acid. Such conservative substitutions are known in the art, and examples have been provided above. Amino acid modifications can range from changing or modifying one or more amino acids to complete redesign of a region, such as the Variable Domain. Changes in the Variable Domain can alter binding affinity and/or specificity. Other methods of modification include using coupling techniques known in the art, including, but not limited to, enzymatic means, oxidative substitution and chelation. Modifications can be used, for example, for attachment of labels for immunoassay, such as the attachment of radioactive moieties for radioimmunoassay. Modified polypeptides are made using established procedures in the art and can be screened using standard assays known in the art. The invention encompasses fusion proteins comprising one or more of the polypeptides or LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 antibodies of this invention. In one embodiment, a fusion polypeptide is provided that comprises a light chain, a heavy chain or both a light and heavy chain. In another embodiment, the fusion polypeptide contains a heterologous immunoglobulin constant region. In another embodiment, the fusion polypeptide contains a Light Chain Variable Domain and a Heavy Chain Variable Domain of an antibody produced from a publicly-deposited hybridoma. For purposes of this invention, an antibody fusion protein contains one or more polypeptide domains that specifically bind to LAG-3 and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region. X. Uses of the LAG-3-Binding Molecules of the Present Invention The present invention encompasses compositions, including pharmaceutical compositions, comprising the LAG-3-binding molecules of the present invention (e.g., anti-LAG-3 antibodies, anti-LAG-3 bispecific diabodies, etc.), polypeptides derived from such molecules, polynucleotides comprising sequences encoding such molecules or polypeptides, and other agents as described herein. As discussed above, LAG-3 plays an important role in negatively regulating T-cell proliferation, function and homeostasis. The LAG-3-binding molecules of the present invention have the ability to inhibit LAG-3 function, and thus reverse the LAG-3-mediated immune system inhibition. As such, LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, and LAG-3 mAb 6, their humanized derivatives, and molecules comprising their LAG-3-binding fragments (e.g., bispecific diabodies, etc.), or that compete for binding with such antibodies, may be used to block LAG-3-mediated immune system inhibition, and thereby promote the activation of the immune system. Bispecific LAG-3-binding molecules of the present invention that bind to LAG-3 and another molecule involved in regulating an immune check point present on the cell surface (e.g., PD-1) augment the immune system by blocking immune system inhibition mediated by LAG-3 and such immune check point molecules. Thus, the LAG-3-binding molecules of the invention are useful for augmenting an immune response (e.g., the T-cell mediated immune response) of a subject. In particular, the LAG-3-binding molecules of the invention and may be used to treat any disease or condition associated with an undesirably suppressed immune system, including cancer and diseases that are associated with the presence of a pathogen (e.g., a bacterial, fungal, viral or protozoan infection). The cancers that may be treated by the LAG-3-binding molecules of the present invention include cancers characterized by the presence of a cancer cell selected from the group consisting of a cell of: an adrenal gland tumor, an AIDS-associated cancer, an alveolar soft part sarcoma, an astrocytic tumor, bladder cancer, bone cancer, a brain and spinal cord cancer, a metastatic brain tumor, a breast cancer, a carotid body tumors, a cervical cancer, a chondrosarcoma, a chordoma, a chromophobe renal cell carcinoma, a clear cell carcinoma, a colon cancer, a colorectal cancer, a cutaneous benign fibrous histiocytoma, a desmoplastic small round cell tumor, an ependymoma, a Ewing's tumor, an extraskeletal myxoid chondrosarcoma, a fibrogenesis imperfecta ossium, a fibrous dysplasia of the bone, a gallbladder or bile duct cancer, gastric cancer, a gestational trophoblastic disease, a germ cell tumor, a head and neck cancer, hepatocellular carcinoma, an islet cell tumor, a Kaposi's Sarcoma, a kidney cancer, a leukemia, a lipoma/benign lipomatous tumor, a liposarcoma/malignant lipomatous tumor, a liver cancer, a lymphoma, a lung cancer, a medulloblastoma, a melanoma, a meningioma, a multiple endocrine neoplasia, a multiple myeloma, a myelodysplastic syndrome, a neuroblastoma, a neuroendocrine tumors, an ovarian cancer, a pancreatic cancer, a papillary thyroid carcinoma, a parathyroid tumor, a pediatric cancer, a peripheral nerve sheath tumor, a phaeochromocytoma, a pituitary tumor, a prostate cancer, a posterious uveal melanoma, a rare hematologic disorder, a renal metastatic cancer, a rhabdoid tumor, a rhabdomysarcoma, a sarcoma, a skin cancer, a soft-tissue sarcoma, a squamous cell cancer, a stomach cancer, a synovial sarcoma, a testicular cancer, a thymic carcinoma, a thymoma, a thyroid metastatic cancer, and a uterine cancer. In particular, the LAG-3-binding molecules of the present invention may be used in the treatment of colorectal cancer, hepatocellular carcinoma, glioma, kidney cancer, breast cancer, multiple myeloma, bladder cancer, neuroblastoma; sarcoma, non-Hodgkin's lymphoma, non-small cell lung cancer, ovarian cancer, pancreatic cancer and rectal cancer. Pathogen-associated diseases that may be treated by the LAG-3-binding molecules of the present invention include chronic viral, bacterial, fungal and parasitic infections. Chronic infections that may be treated by the LAG-3-binding molecules of the present invention include Epstein Barr virus, Hepatitis A Virus (HAV); Hepatitis B Virus (HBV); Hepatitis C Virus (HCV); herpes viruses (e.g. HSV-1, HSV-2, HHV-6, CMV), Human Immunodeficiency Virus (HIV), Vesicular Stomatitis Virus (VSV), Bacilli,Citrobacter, Cholera, Diphtheria,Enterobacter, Gonococci,Helicobacter pylori, Klebsiella, Legionella, Meningococci, mycobacteria, Pseudomonas, Pneumonococci, rickettsiabacteria,Salmonella, Serratia, Staphylococci, Streptococci,Tetanus, Aspergillus(A. fumigatus, A. niger, etc.),Blastomyces dermatitidis, Candida(C. albicans, C. krusei, C. glabrata, C. tropicalis, etc.),Cryptococcus neoformans, Genus Mucorales (mucor, absidia, rhizopus),Sporothrix schenkii, Paracoccidioides brasiliensis, Coccidioides immitis, Histoplasma capsulatum, Leptospirosis,Borrelia burgdorferi, helminth parasite (hookworm, tapeworms, flukes, flatworms (e.g. Schistosomia),Giardia lambia, trichinella, Dientamoeba Fragilis, Trypanosoma brucei, Trypanosoma cruzi, andLeishmania donovani. The LAG-3-binding molecules of the invention can be combined with an immunogenic agent such as a tumor vaccine. Such vaccines may comprise purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), autologous or allogeneic tumor cells. A number of tumor vaccine strategies have been described (see for example, Palena, C., et al., (2006) “Cancer vaccines: preclinical studies and novel strategies,” Adv. Cancer Res. 95, 115-145; Mellman, I., et al. (2011) “Cancer immunotherapy comes of age,” Nature 480, 480-489; Zhang, X. M. et al. (2008) “The Anti-Tumor Immune Response Induced By A Combination of MAGE-3/MAGE-n-Derived Peptides,” Oncol. Rep. 20, 245-252; Disis, M. L. et al. (2002) “Generation of T-cell Immunity to the HER-2ne Protein After Active Immunization with HER-2/neu Peptide-Based Vaccines,” J Clin. Oncol. 20:2624-2632; Vermeij, R. et al. (2012) “Potentiation a p53-SLP Vaccine By Cyclophosphamide In Ovarian Cancer: A Single-Arm Phase II Study.” MI J. Cancer 131:B670-E680). The LAG-3-binding molecules of the invention can be combined with chemotherapeutic regimes In these instances, it may be possible to reduce the dose of chemotherapeutic reagent administered (Mokyr. M. B. et al. (1998) “Realization Of The Therapeutic Potential Of CTLA-4 Blockade In Low-Dose Chemotherapy-Treated Tumor-Bearing Mice,” Cancer Research 58: 530-5304). The LAG-3-binding molecules of the invention can be combined with other immunostimulatory molecules such as antibodies which activate host immune responsiveness to provide for increased levels of T-cell activation. In particular, anti-PD-1 antibodies, anti-PD-L1 antibodies and/or an anti-CTLA-4 antibodies have been demonstrated to active the immune system (see, e.g., del Rio, M-L. et al. (2005) “Antibody-Mediated Signaling Through PD-1Costimulates T Cells And Enhances CD28-Dependent Proliferation,” Eur. J. Immunol 35:3545-3560; Barber, D. L. et al. (2006) “Restoring Function In Exhausted CD8T Cells During Chronic Viral Infection,” Nature 439, 682-687; Iwai, Y. et al. (2002) “Involvement of PD-L1On Tumor Cells In The Escape From Host Immune System And Tumor Immunotherapy by PD-L1blockade,” Proc. Natl Acad. Sci. USA 99, 12293-12297; Leach, D. R., et al., (1996) “Enhancement Of Antitumor Immunity By CTLA-4Blockade,” Science 271, 1734-1736). Additional immunostimulatory molecules that may be combined with the LAG-3-binding molecules of the invention include antibodies to molecules on the surface of dendritic cells that activate dendritic cell (DC) function and antigen presentation, anti-CD40 antibodies able to substitute for T-cell helper activity, and activating antibodies to T-cell costimulatory molecules such as PD-L1, CTLA-4, OX-40 4-1BB, and ICOS (see, for example, Ito et al. (2000) “Effective Priming Of Cytotoxic T Lymphocyte Precursors By Subcutaneous Administration Of Peptide Antigens In Liposomes Accompanied By Anti-CD40And Anti-CTLA-4Antibodies,” Immunobiology 201:527-40; U.S. Pat. No. 5,811,097; Weinberg et al. (2000) “Engagement of the OX-40Receptor In Vivo Enhances Antitumor Immunity,” Immunol 164:2160-2169; Melero et al. (1997) “Monoclonal Antibodies Against The4-1BB T-Cell Activation Molecule Eradicate Established Tumors,” Nature Medicine 3: 682-685; Hutloff et al. (1999) “ICOS is An Inducible T-Cell Co-Stimulator Structurally And Functionally Related to CD28,” Nature 397:263-266; and Moran, A. E. et al. (2013) “The TNFRs OX40, 4-1BB, and CD40As Targets For Cancer Immunotherapy,” Curr Opin Immunol. 2013 April; 25(2): 10.1016/j.coi.2013.01.004), and/or stimulatory Chimeric Antigen Receptors (CARs) comprising an antigen binding domain directed against a disease antigen fused to one or more intracellular signaling domains from various costimulatory protein receptors (e.g., CD28, 4-1BB, ICOS, OX40, etc.) which serve to stimulate T-cells upon antigen binding (see, for example, Tettamanti, S. et al. (2013) “Targeting Of Acute Myeloid Leukaemia By Cytokine-Induced Killer Cells Redirected With A Novel CD123-Specific Chimeric Antigen Receptor,” Br. J. Haematol. 161:389-401; Gill, S. et al. (2014) “Efficacy Against Human Acute Myeloid Leukemia And Myeloablation Of Normal Hematopoiesis In A Mouse Model Using Chimeric Antigen Receptor-Modified T Cells,” Blood 123(15): 2343-2354; Mardiros, A. et al. (2013) “T Cells Expressing CD123-Specific Chimeric Antigen Receptors Exhibit Specific Cytolytic Effector Functions And Antitumor Effects Against Human Acute Myeloid Leukemia,” Blood 122:3138-3148; Pizzitola, I. et al. (2014) “Chimeric Antigen Receptors Against CD331CD123Antigens Efficiently Target Primary Acute Myeloid Leukemia Cells in vivo,” Leukemia 28(8):1596-1605). LAG-3-binding molecules of the invention can be combined with inhibitory Chimeric Antigen Receptors (iCARs) to divert off target immunotherapy responses. iCARs an antigen binding domain directed against a disease antigen fused to one or more intracellular signaling domains from various inhibitory protein receptors (e.g., CTLA-4, PD-1, etc.) which serve to constrain T-cell responses upon antigen binding (see, for example, Fedorov V. D. (2013) “PD-1-and CTLA-4-Based Inhibitory Chimeric Antigen Receptors(iCARs)Divert Off-Target Immunotherapy Responses,” Sci Tranl Med. 5(215):215ra172). In particular, the anti-LAG-3 antibodies of the invention are used in combination with an anti-CD137 antibody, an anti-OX40 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIGIT antibody, an anti-TIM-3 antibody and/or a cancer vaccine. In addition to their utility in therapy, the LAG-3-binding molecules of the present invention may be detectably labeled and used in the detection of LAG-3 in samples or in the imaging of LAG-3 on cells. X. Pharmaceutical Compositions The compositions of the invention include bulk drug compositions useful in the manufacture of pharmaceutical compositions (e.g., impure or non-sterile compositions) and pharmaceutical compositions (i.e., compositions that are suitable for administration to a subject or patient) that can be used in the preparation of unit dosage forms. Such compositions comprise a prophylactically or therapeutically effective amount of the LAG-3-binding molecules of the present invention, or a combination of such agents and a pharmaceutically acceptable carrier. Preferably, compositions of the invention comprise a prophylactically or therapeutically effective amount of the LAG-3-binding molecules of the present invention and a pharmaceutically acceptable carrier. The invention particularly encompasses such pharmaceutical compositions in which the LAG-3-binding molecule is: a LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 antibody; a humanized LAG-3 mAb 1; LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4 LAG-3 mAb 5, or LAG-3 mAb 6 antibody; a LAG-3-binding fragment of any such antibody; or in which the LAG-3-binding molecule is a bispecific LAG-3 diabody (e.g., a LAG-3×PD-1 bispecific diabody). Especially encompassed are such molecules that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 1; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 2; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 3, or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 4, or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 5, or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 6. The invention also encompasses such pharmaceutical compositions that additionally include a second therapeutic antibody (e.g., tumor-specific monoclonal antibody) that is specific for a particular cancer antigen, and a pharmaceutically acceptable carrier. In a specific embodiment, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete), excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. Generally, the ingredients of compositions of the invention are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration. The compositions of the invention can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include, but are not limited to those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc. The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with a LAG-3-binding molecule of the present invention (and more preferably, a LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 antibody; a humanized LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 antibody; a LAG-3-binding fragment of any such antibody; or in which the LAG-3-binding molecule is a bispecific LAG-3 diabody (e.g., a LAG-3×PD-1 bispecific diabody)). Especially encompassed are such molecules that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 1; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 2; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 3; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 4; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 5; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 6, alone or with such pharmaceutically acceptable carrier. Additionally, one or more other prophylactic or therapeutic agents useful for the treatment of a disease can also be included in the pharmaceutical pack or kit. The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. The present invention provides kits that can be used in the above methods. A kit can comprise any of the LAG-3-binding molecules of the present invention. The kit can further comprise one or more other prophylactic and/or therapeutic agents useful for the treatment of cancer, in one or more containers; and/or the kit can further comprise one or more cytotoxic antibodies that bind one or more cancer antigens associated with cancer. In certain embodiments, the other prophylactic or therapeutic agent is a chemotherapeutic. In other embodiments, the prophylactic or therapeutic agent is a biological or hormonal therapeutic. XII. Methods of Administration The compositions of the present invention may be provided for the treatment, prophylaxis, and amelioration of one or more symptoms associated with a disease, disorder or infection by administering to a subject an effective amount of a fusion protein or a conjugated molecule of the invention, or a pharmaceutical composition comprising a fusion protein or a conjugated molecule of the invention. In a preferred aspect, such compositions are substantially purified (i.e., substantially free from substances that limit its effect or produce undesired side effects). In a specific embodiment, the subject is an animal, preferably a mammal such as non-primate (e.g., bovine, equine, feline, canine, rodent, etc.) or a primate (e.g., monkey such as, a cynomolgus monkey, human, etc.). In a preferred embodiment, the subject is a human. Various delivery systems are known and can be used to administer the compositions of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or fusion protein, receptor-mediated endocytosis (See, e.g., Wu et al. (1987) “Receptor-Mediated In Vitro Gene Transformation By A Soluble DNA Carrier System,” J. Biol. Chem. 262:4429-4432), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of administering a molecule of the invention include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous), epidural, and mucosal (e.g., intranasal and oral routes). In a specific embodiment, the LAG-3-binding molecules of the present invention are administered intramuscularly, intravenously, or subcutaneously. The compositions may be administered by any convenient route, for example, by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. See, e.g., U.S. Pat. Nos. 6,019,968; 5,985,320; 5,985,309; 5,934,272; 5,874,064; 5,855,913; 5,290,540; and 4,880,078; and PCT Publication Nos. WO 92/19244; WO 97/32572; WO 97/44013; WO 98/31346; and WO 99/66903, each of which is incorporated herein by reference in its entirety. The invention also provides that the LAG-3-binding molecules of the present invention are packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of the molecule. In one embodiment, such molecules are supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted, e.g., with water or saline to the appropriate concentration for administration to a subject. Preferably, the LAG-3-binding molecules of the present invention are supplied as a dry sterile lyophilized powder in a hermetically sealed container. The lyophilized LAG-3-binding molecules of the present invention should be stored at between 2° C. and 8° C. in their original container and the molecules should be administered within 12 hours, preferably within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted. In an alternative embodiment, such molecules are supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the molecule, fusion protein, or conjugated molecule. Preferably, such LAG-3-binding molecules when provided in liquid form are supplied in a hermetically sealed container. The amount of the composition of the invention which will be effective in the treatment, prevention or amelioration of one or more symptoms associated with a disorder can be determined by standard clinical techniques. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. As used herein, an “effective amount” of a pharmaceutical composition, in one embodiment, is an amount sufficient to effect beneficial or desired results including, without limitation, clinical results such as decreasing symptoms resulting from the disease attenuating a symptom of infection (e.g., viral load, fever, pain, sepsis, etc.) or a symptom of cancer (e.g., the proliferation, of cancer cells, tumor presence, tumor metastases, etc.), thereby increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing the effect of another medication such as via targeting and/or internalization, delaying the progression of the disease, and/or prolonging survival of individuals. An effective amount can be administered in one or more administrations. For purposes of this invention, an effective amount of drug, compound, or pharmaceutical composition is an amount sufficient to reduce the proliferation of (or the effect of) viral presence and to reduce and/or delay the development of the viral disease, either directly or indirectly. In some embodiments, an effective amount of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective amount” may be considered in the context of administering one or more chemotherapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art. For the LAG-3-binding molecules encompassed by the invention (e.g., antibodies, diabodies, etc.), the dosage administered to a patient is preferably determined based upon the body weight (kg) of the recipient subject. For the LAG-binding molecules encompassed by the invention, the dosage administered to a patient is typically from at least about 0.01 μg/kg body weight, at least about 0.05 μg/kg body weight, at least about 0.1 μg/kg body weight, at least about 0.2 μg/kg body weight, at least about 0.5 μg/kg body weight, at least about 1 g/kg body weight, at least about 2 g/kg body weight, at least about 3 g/kg body weight, at least about 5 g/kg body weight, at least about 10 μg/kg body weight, at least about 20 μg/kg body weight, at least about 30 μg/kg body weight, at least about 50 μg/kg body weight, at least about 100 μg/kg body weight, at least about 250 μg/kg body weight, at least about 750 μg/kg body weight, at least about 1.5 mg/kg body weight, at least about 3 mg/kg body weight, at least about 5 mg/kg body weight, or at least about 10 mg/kg, at least about 30 mg/kg, at least about 50 mg/kg, at least about 75 mg/kg, at least about 100 mg/kg, at least about 125 mg/kg, at least about 150 mg/kg or more body weight. The calculated dose will be administered based on the patient's body weight at baseline. Significant (>10%) change in body weight from baseline or established plateau weight should prompt recalculation of dose. In some embodiments, the LAG-3-binding bispecific molecules (e.g., diabodies and trivalent binding molecules) encompassed by the invention, the dosage administered to a patient is typically from at least about 0.3 ng/kg per day to about 0.9 ng/kg per day, from at least about 1 ng/kg per day to about 3 ng/kg per day, from at least about 3 ng/kg per day to about 9 ng/kg per day, from at least about 10 ng/kg per day to about 30 ng/kg per day, from at least about 30 ng/kg per day to about 90 ng/kg per day, from at least about 100 ng/kg per day to about 300 ng/kg per day, from at least about 200 ng/kg per day to about 600 ng/kg per day, from at least about 300 ng/kg per day to about 900 ng/kg per day, from at least about 400 ng/kg per day to about 800 ng/kg per day, from at least about 500 ng/kg per day to about 1000 ng/kg per day, from at least about 600 ng/kg per day to about 1000 ng/kg per day, from at least about 700 ng/kg per day to about 1000 ng/kg per day, from at least about 800 ng/kg per day to about 1000 ng/kg per day, from at least about 900 ng/kg per day to about 1000 ng/kg per day, or at least about 1,000 ng/kg per day. The calculated dose will be administered based on the patient's body weight at baseline. Significant (>10%) change in body weight from baseline or established plateau weight should prompt recalculation of dose. In another embodiment, the patient is administered a treatment regimen comprising one or more doses of such prophylactically or therapeutically effective amount of a LAG-3-binding molecule of the present invention, wherein the treatment regimen is administered over 2 days, 3 days, 4 days, 5 days, 6 days or 7 days. In certain embodiments, the treatment regimen comprises intermittently administering doses of the prophylactically or therapeutically effective amount of the LAG-3-binding molecules of the present invention (for example, administering a dose on day 1, day 2, day 3 and day 4 of a given week and not administering doses of the prophylactically or therapeutically effective amount of the LAG-3-binding molecule (and particularly, a LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, LAG-3 mAb 6 antibody; a humanized LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, or LAG-3 mAb 6 antibody; a LAG-3-binding fragment of any such antibody; or in which the LAG-3-binding molecule is a bispecific LAG-3 diabody (e.g., a LAG-3×PD-1 bispecific Fc diabody). Especially encompassed is the administration (on day 5, day 6 and day 7 of the same week) of molecules that comprise the the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 1; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 2; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 3; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 4; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 5; or that comprise the 3 CDRLs and the 3 CDRHs of LAG-3 mAb 6. Typically, there are 1, 2, 3, 4, 5 or more courses of treatment. Each course may be the same regimen or a different regimen. In another embodiment, the administered dose escalates over the first quarter, first half or first two-thirds or three-quarters of the regimen(s) (e.g., over the first, second, or third regimens of a 4 course treatment) until the daily prophylactically or therapeutically effective amount of the LAG-3-binding molecule is achieved. Table 6 provides 5 examples of different dosing regimens described above for a typical course of treatment with a diabody. TABLE 6Diabody DosageRegimenDay(ng diabody per kg subject weight per day)11, 2, 3, 41001001001001005, 6, 7nonenonenonenonenone21, 2, 3, 43005007009001,0005, 6, 7nonenonenonenonenone31, 2, 3, 43005007009001,0005, 6, 7nonenonenonenonenone41, 2, 3, 43005007009001,0005, 6, 7nonenonenonenonenone The dosage and frequency of administration of a LAG-3-binding molecule of the present invention may be reduced or altered by enhancing uptake and tissue penetration of the molecule by modifications such as, for example, lipidation. The dosage of a LAG-3-binding molecule of the invention administered to a patient may be calculated for use as a single agent therapy. Alternatively, the molecule may be used in combination with other therapeutic compositions and the dosage administered to a patient are lower than when said molecules are used as a single agent therapy. The pharmaceutical compositions of the invention may be administered locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering a molecule of the invention, care must be taken to use materials to which the molecule does not absorb. The compositions of the invention can be delivered in a vesicle, in particular a liposome (See Langer (1990) “New Methods Of Drug Delivery,” Science 249:1527-1533); Treat et al., in LIPOSOMES IN THETHERAPY OFINFECTIOUSDISEASE ANDCANCER, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 3 17-327). The compositions of the invention can be delivered in a controlled-release or sustained-release system. Any technique known to one of skill in the art can be used to produce sustained-release formulations comprising one or more of the LAG-3-binding molecule(s) of the invention. See, e.g., U.S. Pat. No. 4,526,938; PCT publication WO 91/05548; PCT publication WO 96/20698; Ning et al. (1996) “Intratumoral Radioimmunotheraphy Of A Human Colon Cancer Xenograft Using A Sustained-Release Gel,” Radiotherapy & Oncology 39:179-189, Song et al. (1995) “Antibody Mediated Lung Targeting Of Long-Circulating Emulsions,” PDA Journal of Pharmaceutical Science & Technology 50:372-397; Cleek et al. (1997) “Biodegradable Polymeric Carriers For A bFGF Antibody For Cardiovascular Application,” Pro. Int'l. Symp. Control. Rel. Bioact. Mater. 24:853-854; and Lam et al. (1997) “Microencapsulation Of Recombinant Humanized Monoclonal Antibody For Local Delivery,” Proc. Int'l. Symp. Control Rel. Bioact. Mater. 24:759-760, each of which is incorporated herein by reference in its entirety. In one embodiment, a pump may be used in a controlled-release system (See Langer, supra; Sefton, (1987) “Implantable Pumps,” CRC Crit. Rev. Biomed. Eng. 14:201-240; Buchwald et al. (1980) “Long-Term, Continuous Intravenous Heparin Administration By An Implantable Infusion Pump In Ambulatory Patients With Recurrent Venous Thrombosis,” Surgery 88:507-516; and Saudek et al. (1989) “A Preliminary Trial Of The Programmable Implantable Medication System For Insulin Delivery,” N. Engl. J. Med. 321:574-579). In another embodiment, polymeric materials can be used to achieve controlled-release of the molecules (see e.g., MEDICALAPPLICATIONS OFCONTROLLEDRELEASE, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); CONTROLLEDDRUGBIOAVAILABILITY, DRUGPRODUCTDESIGN ANDPERFORMANCE, Smolen and Ball (eds.), Wiley, New York (1984); Levy et al. (1985) “Inhibition Of Calcification Of Bioprosthetic Heart Valves By Local Controlled-Release Diphosphonate,” Science 228:190-192; During et al. (1989) “Controlled Release Of Dopamine From A Polymeric Brain Implant: In Vivo Characterization,” Ann. Neurol. 25:351-356; Howard et al. (1989) “Intracerebral Drug Delivery In Rats With Lesion-Induced Memory Deficits,” J. Neurosurg. 7(1):105-112); U.S. Pat. Nos. 5,679,377; 5,916,597; 5,912,015; 5,989,463; 5,128,326; PCT Publication No. WO 99/15154; and PCT Publication No. WO 99/20253). Examples of polymers used in sustained-release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters. A controlled-release system can be placed in proximity of the therapeutic target (e.g., the lungs), thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in MEDICALAPPLICATIONS OFCONTROLLEDRELEASE, supra, vol. 2, pp. 115-138 (1984)). Polymeric compositions useful as controlled-release implants can be used according to Dunn et al. (See U.S. Pat. No. 5,945,155). This particular method is based upon the therapeutic effect of the in situ controlled-release of the bioactive material from the polymer system. The implantation can generally occur anywhere within the body of the patient in need of therapeutic treatment. A non-polymeric sustained delivery system can be used, whereby a non-polymeric implant in the body of the subject is used as a drug delivery system. Upon implantation in the body, the organic solvent of the implant will dissipate, disperse, or leach from the composition into surrounding tissue fluid, and the non-polymeric material will gradually coagulate or precipitate to form a solid, microporous matrix (See U.S. Pat. No. 5,888,533). Controlled-release systems are discussed in the review by Langer (1990, “New Methods Of Drug Delivery,” Science 249:1527-1533). Any technique known to one of skill in the art can be used to produce sustained-release formulations comprising one or more therapeutic agents of the invention. See, e.g., U.S. Pat. No. 4,526,938; International Publication Nos. WO 91/05548 and WO 96/20698; Ning et al. (1996) “Intratumoral Radioimmunotheraphy Of A Human Colon Cancer Xenograft Using A Sustained-Release Gel,” Radiotherapy & Oncology 39:179-189, Song et al. (1995) “Antibody Mediated Lung Targeting Of Long-Circulating Emulsions,” PDA Journal of Pharmaceutical Science & Technology 50:372-397; Cleek et al. (1997) “Biodegradable Polymeric Carriers For A bFGF Antibody For Cardiovascular Application,” Pro. Int'l. Symp. Control. Rel. Bioact. Mater. 24:853-854; and Lam et al. (1997) “Microencapsulation Of Recombinant Humanized Monoclonal Antibody For Local Delivery,” Proc. Int'l. Symp. Control Rel. Bioact. Mater. 24:759-760, each of which is incorporated herein by reference in its entirety. Where the composition of the invention is a nucleic acid encoding a LAG-3-binding molecule of the present invention, the nucleic acid can be administered in vivo to promote expression of its encoded LAG-3-binding molecule by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (See U.S. Pat. No. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (See e.g., Joliot et al. (1991) “Antennapedia Homeobox Peptide Regulates Neural Morphogenesis,” Proc. Natl. Acad. Sci. (U.S.A.) 88:1864-1868), etc. Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression by homologous recombination. Treatment of a subject with a therapeutically or prophylactically effective amount of a LAG-3-binding molecule of the present invention can include a single treatment or, preferably, can include a series of treatments. In a preferred example, a subject is treated with such a diabody one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks. The pharmaceutical compositions of the invention can be administered once a day, twice a day, or three times a day. Alternatively, the pharmaceutical compositions can be administered once a week, twice a week, once every two weeks, once a month, once every six weeks, once every two months, twice a year or once per year. It will also be appreciated that the effective dosage of the molecules used for treatment may increase or decrease over the course of a particular treatment. EXAMPLES Having now generally described the invention, the same will be more readily understood through reference to the following examples, which are provided by way of illustration and are not intended to be limiting of the present invention unless specified. It will be apparent to those skilled in the art that many modifications, both to materials and methods, can be practiced without departing from the scope of the present disclosure. Example 1: Characterization of Anti-LAG-3 Monoclonal Antibodies LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, and LAG-3 mAb 6 Six murine monoclonal antibodies were isolated as being capable of immunospecifically binding to both human and cynomolgus monkey LAG-3, and accorded the designations “LAG-3 mAb 1,” “LAG-3 mAb 2,” “LAG-3 mAb 3,” “LAG-3 mAb 4,” “LAG-3 mAb 5,” and “LAG-3 mAb 6.” The CDRs of these antibodies were found to differ and are provided above. LAG-3 mAb 1 was humanized yielded two humanized VH Domains, designated herein as “hLAG-3 mAb 1 VH-1,” and “hLAG-3 mAb 1 VH-2,” and four humanized VL Domains designated herein as “hLAG-3 mAb 1 VL-1,” “hLAG-3 mAb 1 VL-2,” “hLAG-3 mAb 1 VL-3,” and “hLAG-3 mAb 1 VL-4.” LAG-3 mAb 6 was also humanized yielded two humanized VH Domains, designated herein as “hLAG-3 mAb 6 VH-1,” and “hLAG-3 mAb 6 VH-2,” and two humanized VL Domains designated herein as “hLAG-3 mAb 6 VL-1,” and “hLAG-3 mAb 6 VL-2.” As provided above, the humanized heavy and light Variable Domains of a given antibody may be used in any combination and particular combinations of humanized Variable Domains are referred to by reference to the specific VH/VL Domains, for example a humanized antibody comprising hLAG-3 mAb 1 VH-1 and hLAG-3 mAb 1 VL-2 is specifically referred to as “hLAG-3 mAb 1(1.2).” Full length humanized mAbs were constructed as follows: the C-terminus of a humanized VL Domain was fused to the N-terminus of a human light chain kappa region to generate a light chain and each light chain is paired with a heavy chain comprising a humanized VH Domain of the same antibody fused to the N-terminus of either a human IgG1 Constant Region comprising the L234A/L235A (AA) substitutions or a human IgG4 Constant Region comprising the S228P substitution. The amino acid sequence of an exemplary human IgG1 Constant Region comprising the L234A/L235A (AA) substitutions (SEQ ID NO:139): ASTKGPSVFP LAPSSKSTSG GTAALGCLVK DYFPEPVTVSWNSGALTSGV HTFPAVLQSS GLYSLSSVVT VPSSSLGTQTYICNVNHKPS NTKVDKRVEP KSCDKTHTCP PCPAPEAAGGPSVFLFPPKP KDTLMISRTP EVTCVVVDVS HEDPEVKFNWYVDGVEVHNA KTKPREEQYN STYRVVSVLT VLHQDWLNGKEYKCKVSNKA LPAPIEKTIS KAKGQPREPQ VYTLPPSREEMTKNQVSLTC LVKGFYPSDI AVEWESNGQP ENNYKTTPPVLDSDGSFFLY SKLTVDKSRW QQGNVFSCSV MHEALHNHYTQKSLSLSPGX wherein, X is a lysine (K) or is absent In SEQ ID NO:139, amino acid residues 1-98 correspond to the IgG1 CH1 Domain (SEQ ID NO:120), amino acid residues 99-113 correspond to the IgG1 hinge region (SEQ ID NO: 114) and amino acid residues 114-329 correspond to the IgG1 CH2-CH3 Domain comprising the L234A/L235A substitutions (underlined) (SEQ ID NO:123). The amino acid sequence of an exemplary human human IgG4 Constant Region comprising the S228P substitution (SEQ ID NO:140): ASTKGPSVFP LAPCSRSTSE STAALGCLVK DYFPEPVTVSWNSGALTSGV HTFPAVLQSS GLYSLSSVVT VPSSSLGTKTYTCNVDHKPS NTKVDKRVES KYGPPCPPCP APEFLGGPSVFLFPPKPKDT LMISRTPEVT CVVVDVSQED PEVQFNWYVDGVEVHNAKTK PREEQFNSTY RVVSVLTVLH QDWLNGKEYKCKVSNKGLPS SIEKTISKAK GQPREPQVYT LPPSQEEMTKNQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDSDGSFFLYSRL TVDKSRWQEG NVFSCSVMHE ALHNHYTQKSLSLSLGX wherein, X is a lysine (K) or is absent In SEQ ID NO:140, amino acid residues 1-98 correspond to the IgG4 CH1 Domain (SEQ ID NO:122), amino acid residues 99-110 correspond to the stabilized IgG4 hinge region comprising the S228P substitutions (underlined) (SEQ ID NO: 117) and amino acid residues 111-326 correspond to the IgG4 CH2-CH3 Domain (SEQ ID NO:4). The binding kinetics of the antibodies LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, LAG-3 mAb 6, several humanized and a reference antibody LAG-3 mAb A was investigated using Biacore analysis. The anti-LAG-3 antibodies were captured and were incubated with His-tagged soluble human LAG-3 (shLAG-3-His) and the kinetics of binding was determined via Biacore analysis. The calculated ka, kd and KDare presented in Table 7. TABLE 7Anti-LAG-3 Antibodyka(×105)kd(×10−4)KD (nM)LAG-3 mAb A8.75.40.6LAG-3 mAb 1ª200.260.013LAG-3 mAb 1b310.270.01LAG-3 mAb 2ª11211.9LAG-3 mAb 3ª7.7344.4LAG-3 mAb 4ª129.30.8LAG-3 mAb 5ª14130.9LAG-3 mAb 6ª420.840.02hLAG-3 mAb 1 (1.2)d,e230.130.01hLAG-3 mAb 1 (2.2)d,e8.22.60.3hLAG-3 mAb 1 (1.1)d,e170.740.04hLAG-3 mAb 1 (1.4)c,e160.590.04hLAG-3 mAb 1 (1.4)c,f170.860.05a= captured on immobilized Fab2 goat-anti-mouse Fcb= captured on immobilized Protein Gc= captured on immobilized Fab2 goat anti-human Fcd= captured on immobilized Protein Ae= human IgG1 (AA)f= IgG4 (S228P) In additional studies, the binding kinetics of the humanized antibodies hLAG-3 mAb 1 (1.4) and hLAG-3 mAb 6 (1.1), and a reference antibody LAG-3 mAb A to both human and cynomolgus monkey LAG-3 was investigated using Biacore analysis. In these studies, a soluble LAG-3 fusion protein (the extracellular domain of human or cynomolgus monkey LAG-3 fused to murine IgG2a) was captured on a Fab2 goat-anti mouse Fc surface and incubated with the anti-LAG-3 antibody and the kinetics of binding was determined via Biacore analysis. The binding curves for LAG-3 mAb A, hLAG-3 mAb 1 (1.4) and hLAG-3 mAb 6 (1.1) binding to cynomolgus monkey LAG-3 are shown inFIGS.7A-7Crespectively, and the calculated ka, kd and KD are presented in Table 8. In a separate study, the binding of a bispecific Fc Region-containing diabody comprising hLAG-3 mAb 6 (1.1) to both human and cynomolgus monkey LAG-3 was investigated using Biacore analysis. In this study, the hLAG-3 mAb 6 (1.1) containing diabody was captured on a Fab2 goat-anti human Fc surface and incubated with soluble LAG-3 fusion protein (the extracellular domain of human or cynomolgus monkey LAG-3 fused to a His tag) and the kinetics of binding was determined via Biacore analysis. The calculated ka, kd and KD are presented in Table 8. TABLE 8HumanCynomolgus MonkeyAnti-LAG-3 Antibodyka(×104)kd(×10−5)KD (nM)ka(×104)kd(×10−5)KD (nM)LAG-3 mAb A6.21.00.162.41100458hLAG-3 mAb 1 (1.4)ª3.4<1.0<0.291.9<1.0<0.53hLAG-3 mAb 6 (1.1)ª9.9<1.0<0.18.2303.7hLAG-3 mAb 6 (1.1)b480160.03359780.13a= immobilized human or cynomolgus LAG-3- murine IgG2a fusion proteinb= immobilized Fc Region-containing diabody comprising a hLAG-3 mAb 6 (1.1) epitope-binding domain The results demonstrate that LAG-3 mAb 1 and LAG-3 mAb 6 exhibit better binding kinetics than reference antibody LAG-3 mAb A. In addition, humanized LAG-3 mAb 6 exhibits better cross-reactive binding kinetics with cynomolgus monkey LAG-3. The epitope specificity of LAG-3 mAb 1, LAG-3 mAb 6 and the reference antibody LAG-3 mAb A was examined using Biacore analysis. In order to determine whether the antibodies bound to different LAG-3 epitopes, shLAG-3-His was captured by mouse anti-PentaHis antibody immobilized on the CM5 sensor chip according to the procedure recommended by the manufacturer. Briefly, the carboxyl groups on the sensor chip surface were activated with an injection of a solution containing 0.2 M N-ethyl-N-(3dietylamino-propyl) carbodimide and 0.05 M N-hydroxy-succinimide. Anti-PentaHis antibody was injected over the activated CM5 surface in 10 mM sodium-acetate, pH 5.0, at a flow rate 5 L/min, followed by 1 M ethanolamine for deactivation of remaining amine-reactive groups. Binding experiments were performed in HBS-EP buffer, which contains 10 mM HEPES, pH 7.4, 150 mM NaCl, 3 mM EDTA, and 0.005% P20 surfactant. Each antibody (LAG-3 mAb 1, LAG-3 mAb 6 and LAG-2 mAb A) was preinjected over captured hLAG3-His for 180 seconds at a flow rate of 5 μL/min at concentration of 1 μM followed by running buffer and injection of competing antibody at the same conditions. Binding response of competing antibody was compared to its binding response to hLAG3-His preinjected with buffer to identify antibodies competing for the same epitope. Regeneration of the immobilized anti-PentaHis surface was performed by pulse injection of 10 mM glycine, pH 1.5. Reference curves were obtained by injection of analytes over the treated surface with no immobilized protein. Binding curves were generated by BIAevaluation software v4.1 from real-time sensogram data. The results of this experiment are shown inFIGS.8A-8D. The results of this experiment indicate that the biotinylated antibody LAG3 mAb A was capable of binding to shLAG-3-His even in the presence of excess amounts of the non-biotinylated antibodies LAG-3 mAb 1 and LAG-3 mAb 6. In contrast, LAG-3 mAb 1 blocked the binding of LAG-3 mAb 6. Thus, the results show that LAG-3 mAb 1 and LAG-3 mAb 6 likely bind to the same, or over lapping epitopes of LAG-3, and compete with one another for binding to LAG-3. Both LAG-3 mAb 1 and LAG-3 mAb 6 were found to bind to an epitope that is distinct from that bound by LAG-3 mAb A. In order to further characterize the anti-LAG3 antibodies, their ability to block binding between LAG-3 and MHC class II was assessed in two different assays. In one assay, the ability of the antibodies to block the binding of a soluble human LAG3-Fc fusion protein to MHC class II immobilized on a surface was examined. For this assay, LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, and LAG-3 mAb 5 each (at 0-67 nM, 2 fold serial dilutions) were mixed with a soluble human LAG-3-Fc fusion protein, (at 5 μg/mL) and were separately incubated with immobilized MHC class II (1 μg/mL). The amount of LAG-3 binding to the immobilized MHC class II was assessed using a goat anti-human Fc gamma-HRP secondary antibody. In additional experiments LAG-3 mAb A and the humanized antibodies, hLAG-3 mAb 1 (1.4) and hLAG-3 mAb 6 (1.1) (at 0.0096-7.0 nM, three fold serial dilutions) were mixed with soluble human LAG-3-His fusion protein (0.2 μg/ml) and assayed for binding to immobilized MHC class II as described above. The results of these experiments are shown inFIG.9AandFIG.9B. In another assay, the ability of the anti-LAG-3 antibodies of the present invention to block the binding of a soluble human LAG3-Fc fusion protein (shLAG-3-Fc) to native MHC class II on a cell surface was examined. For this assay, LAG-3 mAb 1, LAG-3 mAb 2, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 5, and the reference antibody LAG-3 mAb A each (at 0.1-26.7 ng/ml, 2 fold serial dilutions) were mixed with a biotinylated-soluble human LAG-3-Fc fusion protein, (at 0.5 μg/ml) and were separately incubated with MHC II-positive Daudi cells. The amount of LAG-3 binding to the surface of the Daudi cells was determined using a PE-conjugated Streptavidin secondary antibody by FACS analysis. In additional separate experiments LAG-3 mAb 1, LAG-3 mAb 6 and LAG-3 mAb A; or LAG-3 mAb 1 and the humanized antibodies, hLAG-3 mAb 1 (1.4), hLAG-3 mAb 1 (1.2), hLAG-3 mAb 1 (2.2), and hLAG-3 mAb 1 (1.1) were assayed as described above. The results of these experiments are shown inFIGS.10A-10C, respectively. The results of these inhibition assays (FIGS.9A-9BandFIGS.10A-10C) show that all the anti-LAG-3 antibodies tested blocked the binding of a shLAG-3-Fc fusion protein to bind MHC class II. Example 2: Flow-Cytometry Methodology Experiments to determine expression levels of checkpoint inhibitors: PD-1 and LAG-3 on cells in the experiments described below used the following appropriately fluorescent labeled commercial antibodies (phycoerythrin-cyanine7 (PE-Cy7)-conjugated anti-CD4 [clone SK3] or fluorescein isothiocyanate (FITC)-conjugated anti-CD4 [clone RPA-T4], phycoerythrin (PE)-conjugated anti-LAG-3 [clone 3DS223H], phycoerythrin (PE)-conjugated anti-PD-1 [clone EH12.2H7] or allophycocyanin (APC)-conjugated (eBiosciences, or BioLegend)) and the appropriate isotype controls. All antibodies were used at the manufacturer's recommended concentrations. Cell staining was performed in FACS buffer (10% FCS in PBS) on ice for 30 minutes in the dark for the addition of primary antibodies. After two washes, cells were either stained with the appropriate secondary reagent on ice for 30 minutes in the dark or immediately analyzed on a flow cytometer. To exclude dead cells, all samples were co-stained with a viability dye: 7-Aminoactinomycin D (7-AAD) (BD Biosceinces, or BioLegend) or 4′,6-Diamidino-2-Phenylindole, Dihydrochloride (DAPI) (Life Technologies). All samples were analyzed on either a FACS Calibur or Fortessa Flow Cytometer (BD Biosciences) and analyzed using FlowJo Software (TreeStar, Ashland, Oreg.). Example 3: LAG-3 Expression and Antibody Binding to Stimulated T-Cells LAG-3 expression and the ability of the isolated LAG-3 antibodies to specifically bind LAG-3 on the surface of CD3/CD28-stimulated T-cells was examined. T-cells were obtained from peripheral blood mononuclear cell (PBMCs), briefly, PBMCs were purified using the Ficoll-Paque Plus (GE Healthcare) density gradient centrifugation method according to manufacturer's instructions from whole blood obtained under informed consent from healthy donors (Biological Specialty Corporation) and T cells were then purified using the Dynabeads® Untouched Human T Cells Kit (Life Technologies) according to manufacturer's instructions. For stimulation, isolated T cells were cultured for 10-14 days in the presence of recombinant human IL-2 30 U/ml] (Peprotech) and Dynabeads® Human T cell Activator beads (Life Technologies) according to manufacturer's instructions. LAG-3 expression on freshly isolated unstimulated CD4+ T cells, and stimulated CD4+ T cells (taken from culture at day 11 or 14), was examined by flow cytometry as described above, using FITC-conjugated anti-CD4 and PE-conjugated anti-LAG-3. The results of these studies are shown inFIG.11A-11C. No LAG-3 expression was observed on unstimulated CD4+ T-cells (FIG.11A). However, CD3/CD8 stimulated CD4+ T-cells from two different donors (D:58468 and D:43530) exhibited a dramatic increase in LAG-3 expression (FIGS.11B and11C). The ability of LAG-3 mAb 1 (FIG.12, Panels A and D), LAG-3 mAb 2 (FIG.12, Panels B and E), LAG-3 mAb 3 (FIG.12, Panels C and F), LAG-3 mAb 4 (FIG.13, Panels A and C), and LAG-3 mAb 5 (FIG.13, Panels B and D), to specifically bind to stimulated CD4+ T cells was investigated. Stimulated T cells (prepared as described above from donor D:58468) taken from culture at day 14, and fresh unstimulated cells (prepared as described above from donor D:43530) were subjected to flow cytometry using the isolated anti-LAG-3 antibodies and the following appropriately fluorescent labeled secondary reagent (PC-conjugated anti-mouse-IgG (H+L) (Jackson ImmunoResearch Labs)) and FITC-conjugated anti-CD4. As shown inFIG.12, Panels A-F andFIG.13, Panels A-D, each of the anti-LAG-3 antibodies examined bound only to stimulated, but not to unstimulated CD4+ T-cells. The results of these studies demonstrate that LAG-3 is upregulated on stimulated T-cells and that the anti-LAG-3 antibodies of the present invention specifically bind only stimulated T-cells. Example 4: Functional Activity of Anti-LAG Antibodies Staphylococcus aureusenterotoxin type B (SEB) is a microbial superantigen capable of activating a large proportion of T-cells (5-30%). SEB binds to MHC II outside the peptide binding grove and thus is MHC II dependent, but unrestricted and TCR mediated. SEB-stimulation of T-cells results in oligoclonal T-cell proliferation and cytokine production (although donor variability may be observed). The expression of anti-LAG-3 and anti-PD-1 antibodies alone and in combination on SEB-stimulated PMBCs was examined. PBMCs purified as described above were cultured in RPMI-media+10% heat inactivated FBS+1% Penicillin/Streptomycin in T-25 bulk flasks for 2-3 days alone or with SEB (Sigma-Aldrich) at 0.1 ng/ml (primary stimulation). At the end of the first round of SEB-stimulation, PBMCs were washed twice with PBS and immediately plated in 96-well tissue culture plates at a concentration of 1-5×105cells/well in media alone, media with SEB at 0.1 ng/ml (secondary stimulation) and no antibody, or media with SEB and a control IgG antibody, and cultured for an additional 2-3 days. At 48 hours post-primary bulk SEB-stimulation, cells were examined for PD-1 and LAG-3 expression by flow cytometry using PE-conjugated anti-LAG-3 and FITC-conjugated anti-CD3; or APC-conjugated anti-PD-1 and FITC-conjugated anti-CD3. At day 5, post-secondary culture in 96-well plate with SEB-stimulation, wells treated with no antibody or with control antibody were examined using flow cytometry for PD-1 and LAG-3 expression using PE-conjugated anti-LAG-3 and APC-conjugated anti-PD-1. Flow cytometry results from two representative donors (D:34765 and D:53724) are shown inFIG.14, Panels A-D (D:34765) andFIG.15, Panels A-D (D:53724). These results demonstrate that LAG-3 and PD-1 are upregulated by 48 hours post-SEB-stimulation with a further enhancement seen at day 5 post culture with SEB-stimulation. In these studies, Donor 1 had more LAG-3/PD-1 double positive cells after SEB-stimulation. The addition of a control antibody post-SEB-stimulation did not alter LAG-3 or PD-1 expression (compareFIG.14, Panels C and D, andFIG.15, Panels C and D). Upregulation of the immune check point proteins LAG-3 and PD-1 following SEB-stimulation of PBMCs limits cytokine release upon restimulation. The ability of LAG-3 mAb 1, LAG-3 mAb 3, LAG-3 mAb 4, LAG-3 mAb 6, a PD-1 monoclonal antibody designated “PD-1 mAb 5”, and the reference antibodies PD-1 mAb 1 (comprising the 235A/235A Fc variant (AA)), PD-1 mAb 2, LAG-3 mAb A, and the commercial anti-LAG3 antibody 17B4 (#LS-C18692, LS Bio, designated “LAG-3 mAb B”) to enhance cytokine release through checkpoint inhibition was examined. PBMCs were stimulated with SEB as described above except during the secondary stimulations cells were plated with no antibody, with an isotype control antibody, or with anti-LAG-3 and anti-PD-1 antibodies alone or in combination. At the end of the second stimulation, supernatants were harvested to measure cytokine secretion using human DuoSet ELISA Kits for IFNγ, TNFα, IL-10, and IL-4 (R&D Systems) according to the manufacture's instructions.FIGS.16A-16Bshows the IFNγ (FIG.16A) and TNFα (FIG.16B), secretion profiles from SEB-stimulated PBMCs from a representative donor (D:38166), treated with no antibody or one of the following antibodies: isotype control, PD-1 mAb 1, PD-1 mAb 2, LAG-3 mAb A, LAG-3 mAb B, LAG-3 mAb 1, LAG-3 mAb 3, LAG-3 mAb 4, or LAG-3 mAb 6. For this study the antibodies were utilized at 0.009, 0.039, 0.156, 0.625, 2.5, 10, and 40 μg/ml.FIG.17shows the IFNγ secretion profiles from SEB-stimulated PBMCs from another representative donor (D:58108), treated with: no antibody; isotype control antibody; PD-1 mAb 2 and/or LAG-3 mAb A; PD-1 mAb 5 and/or LAG-3 mAb 1; or PD-1 mAb 5 and/or LAG-3 mAb 3. For this study the antibodies were used at 10 μg/ml. The results of these studies demonstrate that anti-PD-1 antibodies dramatically enhanced immune system function as evidenced by increased IFNγ (FIG.16AandFIG.17), and TNFα (FIG.16B) production from SEB-stimulated PBMCs upon restimulation. Surprisingly, LAG-3 mAb 1 was also seen to increase cytokine production across multiple donors to levels comparable to the anti-PD-1 antibodies while the reference anti-LAG-3 mAbs, LAG-3 mAb A and LAG-3 mAb B and several of the isolated antibodies (LAG-3 mAb 3, LAG-4, and LAG-6) provided only a slight enhancement of cytokine release. In addition, the combination of anti-LAG-3 antibodies with anti-PD-1 resulted in a further enhancement of cytokine release (FIG.17) from SEB-stimulated PBMCs upon restimulation. LAG-3 mAb 1 provided the largest enhancement in cytokine release when combined with an anti-PD-1 antibody as compared to LAG-3 mAb 3 and the reference antibody LAG-3 mAb A. Example 5: Binding to Endogenous Cynomolgus Monkey LAG-3 The ability of the humanized antibody hLAG-3 mAb 6 (1.1), and the reference antibody LAG-3 mAb A to bind endogenous LAG-3 expressed on cynomolgus monkey PBMCs by was investigated FACS. For this study PBMCs were isolated from donor cynomolgus monkey whole blood and cultured alone or with SEB stimulation (500 ng/mL) essentially as described above. At 66 hours post-SEB-stimulation cells (unstimulated and stimulated) were stained with hLAG-3 mAb 6 (1.1), LAG-3 mAb A, or PD-1 mAb 1 antibodies (10 fold serial dilutions). The antibodies were detected with goat-anti human Fc-APC labeled secondary antibody. Binding is plotted inFIG.18A-18Bfor two cynomolgus monkey donors. SEB stimulation was confirmed by enhanced PD-1 expression as detected with the anti-PD-1 antibody PD-1 mAb 1. The results of these studies demonstrate that hLAG-3 mAb 6 (1.1) binds endogenous LAG-3 expressed on the surface of stimulated cynomolgous monkey PBMCs. All publications and patents mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference in its entirety. While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth.
249,713
11858992
BRIEF DESCRIPTION OF THE SEQUENCES SEQ ID NO: 1 represents the amino acid sequence of wild type (wt) human TREM-1. SEQ ID NO: 2 represents the amino acid sequence of the variable heavy chain of the m14F69 antibody. SEQ ID NO: 3 represents the amino acid sequence of the variable light chain of the m14F69 antibody. SEQ ID NO: 4 represents the amino acid sequence of the heavy chain of a first humanised TREM-1 antibody (mAb 0170). SEQ ID NO: 5 represents the amino acid sequence of the light chain of a first humanised TREM-1 antibody (mAB 0170). SEQ ID NO: 6 represents the amino acid sequence of the heavy chain of a second humanised TREM-1 antibody (mAb 0122). SEQ ID NO: 7 represents the amino acid sequence of the light chain of a second humanised TREM-1 antibody (mAb 0122). SEQ ID NO: 8 represents the amino acid sequence of the heavy chain of the m14F128 antibody. SEQ ID NO: 9 represents the amino acid sequence of the light chain of the m14F128 antibody. SEQ ID NO: 10 represents the amino acid sequence of the heavy chain of the m14F113 antibody. SEQ ID NO: 11 represents the amino acid sequence of the light chain of the m14F113 antibody. SEQ ID NO: 12 represents the amino acid sequence of the extracellular domain of wild type (wt) cynomolgus monkey (c) TREM-1, when expressed inE. coli. SEQ ID NO: 13 represents the amino acid sequence of K20A-hTREM-1-Cmyc2-His6 (construct 0222). SEQ ID NO: 14 represents the amino acid sequence of A24T/Y28F/N30S/R32Q/P70H-cTREM-1-Cmyc2-His6 (construct 0244). SEQ ID NO: 15 represents the amino acid sequence of A24T/Y28F/N30S/R32Q/ES4K-cTREM-1-Cmyc2-His6 (construct 0245). SEQ ID NO: 16 represents the nucleic acid sequence of a primer SEQ ID NO: 17 represents the nucleic acid sequence of a primer. SEQ ID NO: 18 represents the amino acid sequence of human (h) TREM-1(1-134)-His6. SEQ ID NO: 19 represents the amino acid sequence of cTREM-1-Cmyc2-His6 (construct 0238). SEQ ID NO: 20 represents the amino acid sequence of hTREM-1-Cmyc2-His6 (construct 0247). SEQ ID NO: 21 represents the amino acid sequence of full length cTREM-1. SEQ ID NO: 22 represents the amino acid sequence of full length murine (m) TREM-1. SEQ ID NO: 23 represents the amino acid sequence of full length hPGLYRP1. SEQ ID NO: 24 represents the amino acid sequence of the extracellular IgV-like domain of human TREM-1, Met-human (h) TREM-1 (21-134). SEQ ID NO: 25 represents the amino acid sequence of the heavy chain of the Fab region of monoclonal antibody (mAb)0170. SEQ ID NO: 26 represents the amino acid sequence of the light chain of the Fab region of monoclonal antibody (mAb)0170. DESCRIPTION TREM-1 is a transmembrane protein that consists of 234 amino acids, including a single extracellular immunoglobulin domain and a short cytoplasmic tail with no apparent signaling motif. When activated, TREM-1 associates with the ITAM-containing signaling adaptor protein, DAP12. Downstream signalling may include activation of the NFAT transcription factor, causing an upregulation of pro-inflammatory cytokine production. The present invention relates to antibodies that are capable of specifically binding and blocking the function of TREM-1. Antibodies of the invention may block TREM-1 function by reducing/blocking TREM-1 activation and downstream signalling. Antibodies according to the invention may block TREM-1 by means of one of one or a combination of several different mechanisms, blocking TREM-1 directly or indirectly. For example, antibodies of the invention may prevent the natural ligand of TREM-1, peptidoglycan recognition protein 1 (PGLYRP1), from creating a functional complex with TREM-1 and/or antibodies of the invention may block TREM-1 by preventing individual TREM-1 molecules from forming dimers or multimers. TREM-1 dimerisation or multimerisation may be reduced or prevented by TREM-1 antibodies that are capable of binding to a portion of TREM-1 that would otherwise reside in the interface of a TREM-1 dimer, thus preventing individual TREM-1 molecules from associating with one another. TREM-1 dimerisation or multimerisation may be reduced or prevented by TREM-1 antibodies that interfere with the interaction of TREM-1 with its ligand. Antibodies according to the current invention may block PGLYRP1-induced activation of TREM-1. PGLYRP1, a highly conserved, 196 amino acid long protein consisting of a signal peptide and a peptidoglycan binding domain, is expressed in neutrophils and released upon their activation. Antibodies according to the current invention may down-regulate pro-inflammatory cytokine release from myeloid cells. Antibodies according to the current invention may block the release of TNFalpha, MIP-1beta, MCP-1, IL-1beta, GM.CSF, IL-6 and/or IL-8 from macrophages, neutrophils, synovial tissue cells and/or a reporter cell, as disclosed herein. Antibodies of the invention may be capable of binding both human TREM-1 and TREM-1 from another species than a human being. The term “TREM-1”, as used herein, thus encompasses any naturally occurring form of TREM-1 which may be derived from any suitable organism. For example, TREM-1 for use as described herein may be vertebrate TREM-1, such as mammalian TREM-1, such as TREM-1 from a primate (such as a human, a chimpanzee, a cynomolgus monkey or a rhesus monkey); a rodent (such as a mouse or a rat), a lagomorph (such as a rabbit), or an artiodactyl (such a cow, sheep, pig or camel). Preferably, the TREM-1 is SEQ ID NO: 1 (human TREM-1). The TREM-1 may be a mature form of TREM-1 such as a TREM-1 protein that has undergone post-translational processing within a suitable cell. Such a mature TREM-1 protein may, for example, be glycosylated. The TREM-1 may be a full length TREM-1 protein. Antibodies of the invention may be monoclonal antibodies, in the sense that they are directly or indirectly derived from a single clone of a B lymphocyte. TREM-1 antibodies may be produced, screened and purified using, for example, the methods described in the Examples. In brief, a suitable mouse such as a TREM-1 or TREM-1REM-3 knock-out (KO) mouse may be immunised with TREM-1, a cell expressing TREM-1 or a combination of both. Antibodies of the invention may be polyclonal in the sense of being a mixture of monoclonal antibodies according to the current invention. Primary screening of hybridoma supernatants may be performed using direct ELISA or FMAT and secondary screening may be performed using flow cytometry. Positive hybridoma supernatants may then be screened in a reporter gene assay. Antibodies may be recombinantly expressed in prokaryotic or eukaryotic cells. The prokaryotic cell may beE. coli. The eukaryotic cell may be a yeast, insect or mammalian cell, such as a cell derived from an organism that is a primate (such as a human, a chimpanzee, a cynomolgus monkey or a rhesus monkey), a rodent (such as a mouse or a rat), a lagomorph (such as a rabbit) or an artiodactyl (such a cow, sheep, pig or camel). Suitable mammalian cell lines include, but are not limited to, HEK293 cells, CHO cells and HELA cells. TREM-1 antibodies may also be produced by means of other methods known to the person skilled in the art, such as a phage display or a yeast display. Once produced, antibodies may be screened for binding to, for example, full length TREM-1 or mutants thereof using the methods described in the Examples. One embodiment of the current invention is a method of identifying a functional TREM-1 antibody. Antibodies that are capable of specifically binding TREM-1 and that have any effect upon TREM-1 activation and downstream signalling are herein referred to as “functional TREM-1 antibodies”. A “functional” TREM-1 antibody herein refers to an antibody that is capable of blocking or stimulating TREM-1. The method of identifying a functional TREM-1 antibody comprises (a) culturing a first cell expressing TREM-1, a signalling protein and a reporter construct; (b) measuring the activity of the first cell when said cell is incubated with a TREM-1 modifying agent; (c) contacting the co-culture of (b) with a TREM-1 antibody; and (d) measuring that the activity of the first cell is less than or more than the activity measured in (b). The “first cell” of (a) may be a cell of haematopoetic origin, such as a myeloid cell, such as a T-cell. The signalling protein of (a) may be any signalling protein that is capable of forming a complex with TREM-1. Suitable signalling proteins include DAP10, DAP12, TCR zeta, Fc gamma RIII and an Fc receptor, or part thereof. The reporter construct of (a) may be any construct that is capable of being activated via the signalling protein and generating a recognisable signal. Suitable reporter constructs comprise a transcription factor and a reporter gene. The signalling protein may signal via a transcription factor selected from the group consisting of the NFAT and NFkB. The reporter gene is a gene that is not natively expressed in said first cell and may be a gene that encodes β-galactosidase, luciferase, green fluorescent protein (GFP) or chloramphenicol transferase. Said first cell may be transfected with a transcription factor and a reporter gene using methods that are well known in the art. The “BWZ/hTREM-1 reporter cell” described in the Examples is one example of a “first cell”. The modifying agent of (b) may be a TREM-1 ligand or an activated neutrophil. The “TREM-1 antibody” of (c) may be a TREM-1 specific hybridoma supernatant or a purified antibody. The activity measured in (d) is the signal produced by the reporter construct. An example of such signalling is the luminescence caused by NFAT-driven LacZ β-lactamase luciferase) production. The method may be tailored to identify a blocking TREM-1 antibody. The method of identifying a blocking TREM-1 antibody comprises (a) culturing a first cell expressing TREM-1, a signalling protein and a reporter construct; (b) measuring the activity of the first cell when said cell is incubated with an activated neutrophil; (c) contacting the co-culture of the first cell and the activated neutrophil with a TREM-1 antibody; and (d) measuring that the activity of the first cell is less than the activity measured in (b). The method may also be tailored to identify a stimulating TREM-1 antibody. The method of identifying a stimulating TREM-1 antibody comprises (a) culturing a first cell expressing TREM-1, a signalling protein and a reporter construct; (b) measuring the activity of the first cell; (c) contacting/incubating said cell with a TREM-1 antibody, and (d) measuring that the activity of the first cell is more than the activity of the measured in (b). The present invention relates to blocking TREM-1 antibodies that may be identified by means of the method, herein disclosed, of identifying a blocking antibody. When tested using the method described above and in the Examples, an antibody according to the current invention may, at a concentration of less than 100 μg/ml—such as less than 90 μg/ml, such as less than 80 μg/ml, such as less than 70 μg/ml, such as less than 60 μg/ml, such as less than 50 μg/ml, such as less than 40 μg/ml, such as less than 30 μg/ml, such as less than 20 μg/ml, such as less than 10 μg/ml, such as less than 5 μg/ml, such as less than 1 μg/ml—be capable of reducing the activity of said first cell by 50%, such as 60%, such as 70%, such as 80%, such as 90%, such as 95%, such as 100%. An antibody according to the invention may be capable of completely extinguishing the activity of the first cell. When tested using the method described above and in the Examples, an antibody according to the current invention may, at a concentration of less than 1 μg/ml—such as less than 0.9 μg/ml, such as less than 0.8 μg/m, such as less than 0.7 μg/ml, such as less than 0.6 μg/ml, such as less than 0.5 μg/ml, such as less than 0.4 μg/ml, such as less than 0.3 μg/ml, such as less than 0.2 μg/ml—be capable of extinguishing the activity of the first cell. The present invention also relates to blocking TREM-1 antibodies that may be identified by other means than the method herein disclosed. The term “antibody” herein refers to a protein, derived from a germline immunoglobulin sequence, which is capable of specifically binding to an antigen (TREM-1) or a portion thereof. The term includes full length antibodies of any class or isotype (that is, IgA, IgE, IgG, IgM and/or IgY) and any single chain or fragment thereof. An antibody that specifically binds to an antigen, or portion thereof, may bind exclusively to that antigen, or portion thereof, or it may bind to a limited number of homologous antigens, or portions thereof. Full-length antibodies usually comprise at least four polypeptide chains: two heavy (H) chains and two light (L) chains that are interconnected by disulfide bonds. One immunoglobulin sub-class of particular pharmaceutical interest is the IgG family. In humans, the IgG class may be sub-divided into 4 sub-classes: IgG1, IgG2, IgG3 and IgG4, based on the sequence of their heavy chain constant regions. The light chains can be divided into two types, kappa and lambda, based on differences in their sequence composition. IgG molecules are composed of two heavy chains, interlinked by two or more disulfide bonds, and two light chains, each attached to a heavy chain by a disulfide bond. A heavy chain may comprise a heavy chain variable region (VH) and up to three heavy chain constant (CH) regions: CH1, CH2 and CH3. A light chain may comprise a light chain variable region (VL) and a light chain constant region (CL). VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR). VH and VL regions are typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The hypervariable regions of the heavy and light chains form a [binding] domain that is capable of interacting with an antigen, whilst the constant region of an antibody may mediate binding of the immunoglobulin to host tissues or factors, including but not limited to various cells of the immune system (effector cells), Fc receptors and the first component (Clq) of the classical complement system. Antibodies of the current invention may be isolated. The term “isolated antibody” refers to an antibody that has been separated and/or recovered from (an) other component(s) in the environment in which it was produced and/or that has been purified from a mixture of components present in the environment in which it was produced. Certain antigen-binding fragments of antibodies may be suitable in the context of the current invention, as it has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. The term “antigen-binding fragment” of an antibody refers to one or more fragment(s) of an antibody that retain the ability to specifically bind to an antigen, such as TREM-1, as described herein. Examples of antigen-binding fragments include Fab, Fab′, F(ab)2, F(ab′)2, F(ab)S, Fv (typically the VL and VH domains of a single arm of an antibody), single-chain Fv (scFv; see e.g., Bird et al., Science 1988; 242:42S-426; and Huston et al. PNAS 1988; 85:5879-5883), dsFv, Fd (typically the VH and CHI domain), and dAb (typically a VH domain) fragments; VH, VL, VhH, and V-NAR domains, monovalent molecules comprising a single VH and a single VL chain; minibodies, diabodies, triabodies, tetrabodies, and kappa bodies (see, e.g., Ill et al., Protein Eng 1997; 10:949-57); camel IgG; IgNAR; as well as one or more isolated CDRs or a functional paratope, where the isolated CDRs or antigen-binding residues or polypeptides can be associated or linked together so as to form a functional antibody fragment. Various types of antibody fragments have been described or reviewed in, e.g., Holliger and Hudson, Nat Biotechnol 2005; 2S:1126-1136; WO2005040219, and published U.S. Patent Applications 20050238646 and 20020161201. These antibody fragments may be obtained using conventional techniques known to those of skill in the art, and the fragments may be screened for utility in the same manner as intact antibodies. An antibody of the invention may be a human antibody or a humanised antibody. The term “human antibody”, as used herein, is intended to include antibodies having variable regions in which at least a portion of a framework region and/or at least a portion of a CDR region are derived from human germline immunoglobulin sequences. (For example, a human antibody may have variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences.) Furthermore, if the antibody contains a constant region, the constant region is also derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g. mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). Such a human antibody may be a human monoclonal antibody. Such a human monoclonal antibody may be produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell. Human antibodies may be isolated from sequence libraries built on selections of human germline sequences, further diversified with natural and synthetic sequence diversity. Human antibodies may be prepared by in vitro immunisation of human lymphocytes followed by transformation of the lymphocytes with Epstein-Barr virus. The term “human antibody derivative” refers to any modified form of the human antibody, such as a conjugate of the antibody and another agent or antibody. The term “humanised antibody”, as used herein, refers to a human/non-human chimeric antibody that contains one or more sequences (CDR regions or parts thereof) that are derived from a non-human immunoglobulin. A humanised antibody is, thus, a human immunoglobulin (recipient antibody) in which at least residues from a hyper-variable region of the recipient are replaced by residues from a hyper-variable region of an antibody from a non-human species (donor antibody) such as from a mouse, rat, rabbit or non-human primate, which have the desired specificity, affinity, sequence composition and functionality. In some instances. FR residues of the human immunoglobulin are replaced by corresponding non-human residues. An example of such a modification is the introduction of one or more so-called back-mutations, which are typically amino acid residues derived from the donor antibody. Humanisation of an antibody may be carried out using recombinant techniques known to the person skilled in the art (see, e.g., Antibody Engineering, Methods in Molecular Biology, vol. 248, edited by Benny K. C. Lo). A suitable human recipient framework for both the light and heavy chain variable domain may be identified by, for example, sequence or structural homology. Alternatively, fixed recipient frameworks may be used, e.g., based on knowledge of structure, biophysical and biochemical properties. The recipient frameworks can be germline derived or derived from a mature antibody sequence CDR regions from the donor antibody can be transferred by CDR grafting. The CDR grafted humanised antibody can be further optimised for e.g. affinity, functionality and biophysical properties by identification of critical framework positions where re-introduction (backmutation) of the amino acid residue from the donor antibody has beneficial impact on the properties of the humanised antibody. In addition to donor antibody derived backmutations, the humanised antibody can be engineered by introduction of germline residues in the CDR or framework regions, elimination of immunogenic epitopes, site-directed mutagenesis, affinity maturation, etc. Furthermore, humanised antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, a humanised antibody will comprise at least one—typically two—variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and in which all or substantially all of the FR residues are those of a human immunoglobulin sequence. The humanised antibody can, optionally, also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. The term “humanised antibody derivative” refers to any modified form of the humanised antibody, such as a conjugate of the antibody and another agent or antibody. The term “chimeric antibody”, as used herein, refers to an antibody whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin variable and constant region genes that originate from different species. For example, the variable segments of genes from a mouse monoclonal antibody may be joined to human constant segments. The fragment crystallisable region (“Fc region”/“Fc domain”) of an antibody is the N-terminal region of an antibody, which comprises the constant CH2 and CH3 domains. The Fc domain may interact with cell surface receptors called Fc receptors, as well as some proteins of the complement system. The Fc region enables antibodies to interact with the immune system. In one aspect of the invention, antibodies may be engineered to include modifications within the Fc region, typically to alter one or more of its functional properties, such as serum half-life, complement fixation. Fc-receptor binding, protein stability and/or antigen-dependent cellular cytotoxicity, or lack thereof, among others. Furthermore, an antibody of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody. An IgG1 antibody may carry a modified Fc domain comprises one or more, and perhaps all of the following mutations that will result in decreased affinity to certain Fc receptors (L234A, L235E, and G237A) and in reduced C1q-mediated complement fixation (A330S and P331S), respectively (residue numbering according to the EU index). The isotype of an antibody of the invention may be IgG, such as IgG1, such as IgG2, such as IgG4. If desired, the class of an antibody may be “switched” by known techniques. For example, an antibody that was originally produced as an IgM molecule may be class switched to an IgG antibody. Class switching techniques also may be used to convert one IgG subclass to another, for example: from IgG1 to IgG2 or IgG4; from IgG2 to IgG1 or IgG4; or from IgG4 to IgG1 or IgG2. Engineering of antibodies to generate constant region chimeric molecules, by combination of regions from different IgG subclasses, can also be performed. In one embodiment, the hinge region of CH1 is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further for instance in U.S. Pat. No. 5,677,425 by Bodmer et al. The constant region may be modified to stabilize the antibody, e.g., to reduce the risk of a bivalent antibody separating into two monovalent VH-VL fragments. For example, in an IgG4 constant region, residue S228 (residue numbering according to the EU index) may be mutated to a proline (P) residue to stabilise inter heavy chain disulphide bridge formation at the hinge (see, e.g., Angal et al., Mol Immunol. 1995; 30: 105-8). Antibodies or fragments thereof may also be defined in terms of their complementarity-determining regions (CDRs). The term “complementarity-determining region” or “hypervariable region”, when used herein, refers to the regions of an antibody in which amino acid residues involved in antigen binding are situated. The region of hypervariability or CDRs can be identified as the regions with the highest variability in amino acid alignments of antibody variable domains. Databases can be used for CDR identification such as the Kabat database, the CDRs e.g. being defined as comprising amino acid residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) of the light-chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy-chain variable domain; (Kabat et al. (1991) Sequences of Proteins of Immunological Interest. Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) Alternatively CDRs can be defined as those residues from a “hypervariable loop” (residues 26-33 (L1), 50-52 (L2) and 91-96 (L3) in the light-chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy-chain variable domain; Chothia and Lesk, J. Mol Biol 1987; 196: 901-917). Typically, the numbering of amino acid residues in this region is performed by the method described in Kabat et at, supra. Phrases such as “Kabat position”, “Kabat residue”, and “according to Kabat” herein refer to this numbering system for heavy chain variable domains or light chain variable domains. Using the Kabat numbering system, the actual linear amino acid sequence of a peptide may contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a framework (FR) or CDR of the variable domain. For example, a heavy chain variable domain may include amino acid insertions (residue 52a, 52b and 52c according to Kabat) after residue 52 of CDR H2 and inserted residues (e.g. residues 82a, 82b, and 82c, etc. according to Kabat) after heavy chain FR residue 82. The Kabat numbering of residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a “standard” Kabat numbered sequence. The term “framework region” or “FR” residues refer to those VH or VL amino acid residues that are not within the CDRs, as defined herein. The m14F69 antibody has a variable heavy chain sequence as shown in SEQ ID NO: 2 and a variable light chain sequence as shown in SEQ ID NO: 3. An antibody of the invention may comprise this variable heavy chain sequence and/or this variable light chain sequence. The m14F69 antibody has the CDR sequences shown at amino acids 31 to 35, 50 to 68 and 101 to 110 of SEQ ID NO: 2 and amino acids 24 to 38, 54 to 60 and 93 to 101 of SEQ ID NO: 3. An antibody of the invention may comprise 1, 2, 3, 4, 5 or all 6 of these CDR sequences. An antibody of the invention may comprise amino acids 101 to 110 of SEQ ID NO: 2. The heavy chain of an antibody according to the invention may comprise a CDR1 sequence of amino acids 31 to 35 (TYAMH) of SEQ ID NO: 2, wherein one of these amino acids may be substituted by a different amino acid. The heavy chain of an antibody according to the invention may comprise a CDR2 sequence of amino acids 50 to 68 (RIRTKSSNYATYYADSVKD) of SEQ ID NO: 2, wherein one, two or three of these amino acids may be substituted by a different amino acid. The heavy chain of an antibody according to the invention may comprise a CDR3 sequence of amino acids 101 to 110 (DMGQRRQFAY) of SEQ ID NO: 2, wherein one, two or three of these amino acids may be substituted by a different amino acid. The light chain of an antibody according to the invention may comprise a CDR1 sequence of amino acids 24 to 38 (RASESVDTFDYSFLH) of SEQ ID NO: 3, wherein one, two or three of these amino acids may be substituted with a different amino acid. The light chain of an antibody according to the invention may comprise a CDR2 sequence of amino acids 54 to 60 (RASNLES) of SEQ ID NO: 3, wherein one or two of these amino acids may be substituted with a different amino acid. The light chain of an antibody according to the invention may comprise a CDR3 sequence of amino acids 93 to 101 (QQSNEDPYT) of SEQ ID NO: 3, wherein one or two of these amino acids may be substituted with a different amino acid. The mAb 0170 antibody has a heavy chain sequence as shown in SEQ ID NO: 4 and a light chain sequence as shown in SEQ ID NO: 5. An antibody of the invention may comprise this heavy chain sequence and/or this light chain sequence. The mAb 0170 antibody has the CDR sequences shown at amino acids 31 to 35, 50 to 68 and 101 to 110 of SEQ ID NO: 4 and amino acids 24 to 38, 54 to 60 and 93 to 101 of SEQ ID NO. S. An antibody of the invention may comprise 1, 2, 3, 4, or all 6 of these CDR sequences. The mAb 0122 antibody has a heavy chain sequence as shown in SEQ ID NO: 6 and a light chain sequence as shown in SEQ ID NO: 7. An antibody of the invention may comprise this heavy chain sequence and/or this light chain sequence. The mAb 0122 antibody has the CDR sequences shown at amino acids 31 to 35, 50 to 68 and 101 to 110 of SEQ ID NO: 6 and amino acids 24 to 38, 54 to 60 and 93 to 101 of SEQ ID NO: 7. An antibody of the invention may comprise 1, 2, 3, 4, 5 or all 6 of these CDR sequences. The heavy chain of an antibody according to the invention may comprise a CDRH1 sequence of amino acids 31 to 35 (TYAMH) of SEQ ID NO: 4 or SEQ ID NO: 6, wherein one of these amino acids may be substituted by a different amino acid residue. The heavy chain of an antibody according to the invention may comprise a CDRH2 sequence of amino acids 50 to 68 (RIRTKSSNYATYYAASVKG) of SEQ ID NO: 4 or SEQ ID NO: 6, wherein one, two or three of these amino acids may be substituted by a different amino acid. The heavy chain of an antibody according to the invention may comprise a CDRH3 sequence of amino acids 101 to 110 (DMGIRRQFAY) of SEQ ID NO: 4, wherein one, two or three of these amino acids may be substituted by a different amino acid. The heavy chain of an antibody according to the invention may comprise a CDRH3 sequence of amino acids 101 to 110 (DMGQRRQFAY) of SEQ ID NO: 6, wherein one, two or three of these amino acids may be substituted by a different amino acid. The light chain of an antibody according to the invention may comprise a CDRL1 sequence of amino acids 24 to 38 (RASESVDTFDYSFLH) of SEQ ID NO: 5 or SEQ ID NO: 7, wherein one, two or three of these amino acids may be substituted with a different amino acid. The light chain of an antibody according to the invention may comprise a CDRL2 sequence of amino acids 54 to 60 (RASNLES) of SEQ ID NO: 5 or SEQ ID NO: 7, wherein one or two of these amino acids may be substituted with a different amino acid. The light chain of an antibody according to the invention may comprise a CDRL3 sequence of amino acids 93 to 101 (QQSNEDPYT) of SEQ ID NO: 5 or SEQ ID NO: 7, wherein one or two of these amino acids may be substituted with a different amino acid. The m14F128 antibody has a heavy chain as shown in SEQ ID NO: 8 and a light chain as shown in SEQ ID NO: 9. An antibody of the invention may comprise this variable heavy chain sequence and/or this variable light chain sequence. The m14F128 antibody has the CDR sequences shown at amino acids 31 to 35, 50 to 68 and 101 to 110 of SEQ ID NO: 8 and amino acids 24 to 38, 54 to 60 and 93 to 101 of SEQ ID NO: 9. An antibody of the invention may comprise 1, 2, 3, 4, 5 or all 6 of these CDR sequences. The m14F113 antibody has a heavy chain as shown in SEQ ID NO. 10 and a light chain as shown in SEQ ID NO: 11. An antibody of the invention may comprise this variable heavy chain sequence and/or this variable light chain sequence. The m14F113 antibody has the CDR sequences shown at amino acids 31 to 35, 50 to 68 and 101 to 110 of SEQ ID NO: 10 and amino acids 24 to 38, 54 to 60 and 93 to 101 of SEQ ID NO: 11. An antibody of the invention may comprise 1, 2, 3, 4, 5 or all 6 of these CDR sequences. An antibody of the invention may comprise amino acids 101 to 110 of SEQ ID NO: 10. The term “antigen” (Ag) refers to the molecular entity used for immunization of an immunocompetent vertebrate to produce the antibody (Ab) that recognizes the Ag. Herein, Ag is termed more broadly and is generally intended to include target molecules that are specifically recognized by the Ab, thus including fragments or mimics of the molecule used in the immunization process, or other process, e.g. phage display, used for generating the Ab. The term “epitope”, as used herein, is defined in the context of a molecular interaction between an “antigen binding polypeptide”, such as an antibody (Ab), and its cot-responding antigen (Ag). Generally, “epitope” refers to the area or region on an Ag to which an Ab specifically binds, i.e. the area or region in physical contact with the Ab. Physical contact may be defined through various criteria (e.g. a distance cut-off of 2-6 Å, such as 3 Å, such as 4 Å, such as 5 Å; or solvent accessibility) for atoms in the Ab and Ag molecules. A protein epitope may comprise amino acid residues in the Ag that are directly involved in binding to a Ab (also called the immunodominant component of the epitope) and other amino acid residues, which are not directly involved in binding, such as amino acid residues of the Ag which are effectively blocked by the Ab, i.e. amino acid residues within the “solvent-excluded surface” and/or the “footprint” of the Ab. The term epitope herein comprises both types of binding region in any particular region of TREM-1 that specifically binds to a TREM-1 antibody. TREM-1 may comprise a number of different epitopes, which may include, without limitation, conformational epitopes which consist of one or more non-contiguous amino acids located near each other in the mature TREM-1 conformation and post-translational epitopes which consist, either in whole or par, of molecular structures covalently attached to TREM-1, such as carbohydrate groups. The epitope for a given antibody (Ab)/antigen (Ag) pair can be described and characterized at different levels of detail using a variety of experimental and computational epitope mapping methods. The experimental methods include mutagenesis, X-ray crystallography, Nuclear Magnetic Resonance (NMR) spectroscopy, Hydrogen deuterium eXchange Mass Spectrometry (HX-MS) and various competition binding methods; methods that are known in the art. As each method relies on a unique principle, the description of an epitope is intimately linked to the method by which it has been determined. Thus, depending on the epitope mapping method employed, the epitope for a given Ab/Ag pair may be described differently. At its most detailed level, the epitope for the interaction between the Ag and the Ab can be described by the spatial coordinates defining the atomic contacts present in the Ag-Ab interaction, as well as information about their relative contributions to the binding thermodynamics. At a less detailed level, the epitope can be characterized by the spatial coordinates defining the atomic contacts between the Ag and Ab. At an even less detailed level the epitope can be characterized by the amino acid residues that it comprises as defined by a specific criteria such as the distance between or solvent accessibility of atoms in the Ab:Ag complex. At a further less detailed level the epitope can be characterized through function, e.g. by competition binding with other Abs. The epitope can also be defined more generically as comprising amino acid residues for which substitution by another amino acid will alter the characteristics of the interaction between the Ab and Ag. In the context of an X-ray derived crystal structure defined by spatial coordinates of a complex between an Ab, e.g. a Fab fragment, and its Ag, the term epitope is herein, unless otherwise specified or contradicted by context, specifically defined as TREM-1 residues characterized by having a heavy atom (i.e. a non-hydrogen atom) within a distance of, eg., 4 Å from a heavy atom in the Ab. From the fact that descriptions and definitions of epitopes, dependant on the epitope mapping method used, are obtained at different levels of detail, it follows that comparison of epitopes for different Abs on the same Ag can similarly be conducted at different levels of detail. Epitopes described on the amino acid level, e.g. determined from an X-ray structure, are said to be identical if they contain the same set of amino acid residues. Epitopes are said to overlap if at least one amino acid is shared by the epitopes. Epitopes are said to be separate (unique) if no amino acid residue are shared by the epitopes. Epitopes may also be defined indirectly, by means of comparing the binding kinetics of antibodies to wild type human TREM-1 with those of human TREM-1 variants that have alanine mutations in anticipated epitopes. Decreased affinity or abrogated binding of an antibody to variants of human TREM-1 in which an amino acid residue has been replaced with an alanine residue indicates that the mutated amino acid contributes to the interaction between said antibody and wild type human TREM-1. This approach provides a negative identification of the epitope. The method is compromised in effectively defining the epitope by the fact that protein misfolding or unfolding would give similar results as abrogation of interaction. The analysis can be complemented by comparative gain of function mutational analyses of an orthologous target protein (eg., cynomolgus monkey TREM-1), if a cross-reactive antibody exists. The comparison will define the epitope differences between the antibody that does not cross-react with, eg., cynomolgus monkey TREM-1 and the cross-reactive antibody. Indirect identification of the epitope can also be provided by means of measuring antibody (or antibody fragment) binding to variants of the wild type antigen (TREM-1). If an antibody or fragment thereof binds, eg., human but not cynomolgus monkey TREM-1 and if said antibody or fragment thereof is capable of binding a partly humanised variant of cynomolgus monkey TREM-1 then this regained binding indicates that the substituted amino acid residue(s) is/are important for the interaction of the antibody with the antigen. In the same way, increased affinity for humanized variants of cynomolgus monkey TREM-1, of an anti-human TREM-1 antibody (or its Fab fragment) that has a weaker binding to cynomolgus monkey TREM-1 compared to human TREM-1, can provide information on the identity of residues composing the binding epitope. The effect of the same mutations on any given cross-reactive antibody makes it possible to discriminate between possible protein misfolding (abrogated binding to both antibodies) and loss of interaction in human TREM-1 (binding to one of the antibodies and abrogated binding to the other antibody), whilst unambiguously providing information on the epitope differences between the antibody that does not cross-react and the cross reactive antibody on an amino acid level. Antibodies of the current invention may be capable of binding variants of human TREM-1. Antibodies of the invention may be capable of binding K20A-hTREM-1-Cmyc2-His6 (SEQ ID NO: 13), as determined using, eg., surface plasmon resonance. Antibodies of the current invention may be capable of binding variants of cynomolgus monkey TREM-1. Antibodies of the invention may be capable of binding A24T/Y28F/N30S/R32Q/P70H-cTREM-1-Cmyc2-His6 (SEQ ID NO: 14), as determined using, eg., surface plasmon resonance. Antibodies of the invention may be capable of binding A24T/Y28F/N30S/R32Q/E54K-cTREM-1-Cmyc2-His6 (SEQ ID NO: 15), as determined using, eg., surface plasmon resonance. An antibody of the invention may be capable of specifically binding TREM-1, wherein said antibody is capable of specifically binding (i) at least one amino acid residue selected from the group consisting of the A21, T22, K23, L24, T25, E26, and (ii) at least one amino acid residue selected from the group consisting of the A49, S50, S51, Q52, K53, A54, W55, Q56, I57, I58, R59, D60, G61, E62, M63, P64, K65, T66, L67, A68, C69, T70, E71, R72, P73, S74, K75, N76, S77, H78, P79, V80, Q81, V82, G83, R84, I85 and (iii) at least one amino acid residue selected from the group consisting of the C113, V114, I115, Y116, Q117, P118 and P119 of human TREM-1. An antibody of the invention may be capable of specifically binding a polypeptide comprising amino acids D38 to F48 of SEQ ID NO: 1 (human TREM-1), as determined using, eg., HX-MS. An antibody of the invention may have an epitope comprising one, two, three, four, five, six, seven or all of the amino acid residues D38, V39, K40, C41, D42, Y43, T44 and L45 of SEQ ID NO: 1 (human TREM-1) and one, two or all of the amino acid residues selected from the group consisting of the E46, K47 and F48 of SEQ ID NO: 1 (human TREM-1) as determined using, eg., HX-MS. An antibody of the invention may have an epitope comprising one, two, three or all of the amino acid residues selected from the group consisting of the D42, E46, D92 and H93 of SEQ ID NO: 1 (human TREM-1), as determined using variants of TREM-1 and surface plasmon resonance. An antibody of the invention may have an epitope comprising at least the amino acid residues E46 and/or D92 of SEQ ID NO: 1 (human TREM-1), as determined using variants of TREM-1 and surface plasmon resonance. An antibody of the invention may further comprise one, two or all of the amino acid residues selected from the group consisting of the L31, I86 and V101 of SEQ ID NO: 1 (human TREM-1). An antibody of the invention may be capable of specifically binding a polypeptide comprising amino acid residues E19 to L26 of cynomolgus monkey TREM-1 (SEQ ID NO: 12), or the corresponding amino acids of SEQ ID NO: 21, as determined using, eg., HX-MS. An antibody of the invention may be capable of specifically binding human TREM-1, wherein the epitope of said antibody comprises one, two, three, four, five, six, seven, eight, nine or all of the amino acid residues selected from the group consisting of the V39, K40, C41, D42, Y43, L45, E46, K47, F48 and A49 of SEQ ID NO: 1. An antibody of the invention may be capable of specifically binding human TREM-1, wherein the epitope of said antibody comprises one, two, three, four, five, six, seven, eight, nine or all of the amino acid residues K40, D42, T44, L45, E46, K47, Y90, H91, D92, H93, G94, L95 and R97 of SEQ ID NO: 1. An antibody of the invention may be capable of specifically binding human TREM-1, wherein the epitope of said antibody comprises the D42 of SEQ ID NO: 1. An antibody of the invention may be capable of specifically binding human TREM-1, wherein the epitope of said antibody comprises the E46 of SEQ ID NO: 1. The epitope of said antibody may comprise the V39, C41, D42, Y43, L45 of SEQ ID NO: 1. The epitope of said antibody may comprise the E46, K47 and A49 of SEQ ID NO: 1. The epitope of said antibody may further comprise the F48 of SEQ ID NO: 1. The epitope of said antibody may comprise the K40, 142T44, L45, E46, K47, Y90, H91, D92, H93, G94, L95 and R97 of SEQ ID NO: 1. The definition of the term “paratope” is derived from the above definition of “epitope” by reversing the perspective. Thus, the term “paratope” refers to the area or region on the Ab to which an Ag specifically binds, i.e. with which it makes physical contact to the Ag. In the context of an X-ray derived crystal structure, defined by spatial coordinates of a complex between an Ab, such as a Fab fragment, and its Ag, the term paratope is herein, unless otherwise specified or contradicted by context, specifically defined as Ag residues characterized by having a heavy atom (i.e. a non-hydrogen atom) within a distance of 4 Å from a heavy atom in TREM-1. The epitope and paratope for a given antibody (Ab)/antigen (Ag) pair may be identified by routine methods. For example, the general location of an epitope may be determined by assessing the ability of an antibody to bind to different fragments or variant TREM-1 polypeptides. The specific amino acids within TREM-1 that make contact with an antibody (epitope) and the specific amino acids in an antibody that make contact with TREM-1 (paratope) may also be determined using routine methods. For example, the antibody and target molecule may be combined and the Ab:Ag complex may be crystallised. The crystal structure of the complex may be determined and used to identify specific sites of interaction between the antibody and its target. Antibodies that bind to the same antigen can be characterised with respect to their ability to bind to their common antigen simultaneously and may be subjected to “competition binding”/“binning”. In the present context, the term “binning” refers to a method of grouping antibodies that bind to the same antigen. “Binning” of antibodies may be based on competition binding of two antibodies to their common antigen in assays based on standard techniques such as surface plasmon resonance (SPR), ELISA or flow cytometry. An antibody's “bin” is defined using a reference antibody. If a second antibody is unable to bind to an antigen at the same time as the reference antibody, the second antibody is said to belong to the same “bin” as the reference antibody. In this case, the reference and the second antibody competitively bind the same part of an antigen and are coined “competing antibodies”. If a second antibody is capable of binding to an antigen at the same time as the reference antibody, the second antibody is said to belong to a separate “bin”. In this case, the reference and the second antibody do not competitively bind the same part of an antigen and are coined “non-competing antibodies”. Antibody “binning” does not provide direct information about the epitope. Competing antibodies, i.e. antibodies belonging to the same “bin” may have identical epitopes, overlapping epitopes or even separate epitopes. The latter is the case if the reference antibody bound to its epitope on the antigen takes up the space required for the second antibody to contact its epitope on the antigen (“steric hindrance”). Non-competing antibodies generally have separate epitopes. An antibody of the invention may compete with mAb 0170 for binding to human TREM-1. An antibody of the invention may compete with mAb 0170 for binding to cynomolgus monkey TREM-1. In other words, an antibody of the invention may belong to the same “bin” as mAb 0170. The term “binding affinity” herein refers to a measurement of the strength of a non-covalent interaction between two molecules, e.g. an antibody, or fragment thereof, and an antigen. The term “binding affinity” is used to describe monovalent interactions (intrinsic activity). Binding affinity between two molecules, e.g. an antibody, or fragment thereof, and an antigen, through a monovalent interaction may be quantified by determination of the equilibrium dissociation constant (KD). In turn, KDcan be determined by measurement of the kinetics of complex formation and dissociation, e.g. by the SPR method. The rate constants corresponding to the association and the dissociation of a monovalent complex are referred to as the association rate constant ka(or kon) and dissociation rate constant kd(or koff), respectively. KDis related to kaand kdthrough the equation KD=kd/ka. Following the above definition, binding affinities associated with different molecular interactions, such as comparison of the binding affinity of different antibodies for a given antigen, may be compared by comparison of the KDvalues for the individual antibody/antigen complexes. An antibody of the invention may bind human TREM-1 with an affinity (KD) that is 1×10−7M or less, 1×10−8M or less, or 1×10−9M or less, or 1×10−10M or less, 1×10−11M or less, 1×10−12M or lessor 1×10−13M or less, as determined using surface plasmon resonance. An antibody of the invention may bind cynomolgus monkey TREM-1 with an affinity (KD) that is 1×10−7M or less, 1×10−8M or less, or 1×10−9M or less, or 1×10−10M or less, 1×10−11M or less, 1×10−12M or less or 1×10−13M or less, as determined using surface plasmon resonance. The term “binding specificity” herein refers to the interaction of a molecule such as an antibody, or fragment thereof, with a single exclusive antigen, or with a limited number of highly homologous antigens (or epitopes). In contrast, antibodies that are capable of specifically binding to TREM-1 are not capable of binding dissimilar molecules. Antibodies according to the invention may not be capable of binding Nkp44. The specificity of an interaction and the value of an equilibrium binding constant can be determined directly by well-known methods. Standard assays to evaluate the ability of ligands (such as antibodies) to bind their targets are known in the art and include, for example, ELISAs, Western blots, RIAs, and flow cytometry analysis. The binding kinetics and binding affinity of the antibody also can be assessed by standard assays known in the art, such as SPR. A competitive binding assay can be conducted in which the binding of the antibody to the target is compared to the binding of the target by another ligand of that target, such as another antibody. In another aspect, the present invention provides compositions and formulations comprising molecules of the invention, such as the TREM-1 antibodies, polynucleotides, vectors and cells described herein. For example, the invention provides a pharmaceutical composition that comprises one or more TREM-1 antibodies of the invention, formulated together with a pharmaceutically acceptable carrier. Accordingly, one object of the invention is to provide a pharmaceutical formulation comprising such a TREM-1 antibody which is present in a concentration from 0.25 mg/ml to 250 mg/ml, such as a concentration of from 10 to 200 mg/ml, and wherein said formulation has a pH from 2.0 to 10.0, such as a pH of from 40 to 8.0. The formulation may further comprise one or more of a buffer system, a preservative, a tonicity agent, a chelating agent, a stabilizer and/or a surfactant, as well as various combinations thereof. The use of preservatives, isotonic agents, chelating agents, stabilizers and surfactants in pharmaceutical compositions is well-known to the skilled person. Reference may be made to Remington: The Science and Practice of Pharmacy, 19thedition, 1995. In one embodiment, the pharmaceutical formulation is an aqueous formulation. Such a formulation is typically a solution or a suspension, but may also include colloids, dispersions, emulsions, and multi-phase materials. The term “aqueous formulation” is defined as a formulation comprising at least 50% w/w water. Likewise, the term “aqueous solution” is defined as a solution comprising at least 50% w/w water, and the term “aqueous suspension” is defined as a suspension comprising at least 50% w/w water. In another embodiment, the pharmaceutical formulation is a freeze-dried formulation, to which the physician or the patient adds solvents and/or diluents prior to use. In a further aspect, the pharmaceutical formulation comprises an aqueous solution of such an antibody, and a buffer, wherein the antibody is present in a concentration from 1 mg/ml or above, and wherein said formulation has a pH from about 2.0 to about 10.0. The TREM-1 antibodies of the present invention and pharmaceutical compositions comprising such antibodies may be used for the treatment of inflammatory diseases such as the following: inflammatory bowel disease (IBD), Crohns disease (CD), ulcerative colitis (UC), irritable bowel syndrome, rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), psoriasis, psoriatic arthritis, systemic lupus erythematosus (SLE), lupus nephritis, type I diabetes, Grave's disease, multiple sclerosis (MS), autoimmune myocarditis, Kawasaki disease, coronary artery disease, chronic obstructive pulmonary disease, interstitial lung disease, autoimmune thyroiditis, scleroderma, systemic sclerosis, osteoarthritis, atoptic dermatitis, vitiligo, graft versus host disease, Sjogrens's syndrome, autoimmune nephritis, Goodpasture's syndrome, chronic inflammatory demyelinating polyneuropathy, allergy, asthma and other autoimmune diseases that are a result of either acute or chronic inflammation. TREM-1 antibodies of the invention may be suitable for use in the treatment of individuals with inflammatory bowel disease. Inflammatory Bowel Disease (IBD) is a disease that may affect any part of the gastrointestinal tract from mouth to anus, causing a wide variety of symptoms. IBD primarily causes abdominal pain, diarrhea (which may be bloody), vomiting or weight loss, but may also cause complications outside of the gastrointestinal tract such as skin rashes, arthritis, inflammation of the eye, fatigue and lack of concentration. Patients with IBD can be divided into two major classes, those with ulcerative colitis (UC) and those with Crohn's disease (CD). CD generally involves the ileum and colon, it can affect any region of the intestine but is often discontinuous (focused areas of disease spread throughout the intestine). UC always involves the rectum (colonic) and is more continuous. In CD, the inflammation is transmural, resulting in abscesses, fistulas and strictures, whereas in UC, the inflammation is typically confined to the mucosa. There is no known pharmaceutical or surgical cure for Crohn's disease, whereas some patients with UC can be cured by surgical removal of the colon. Treatment options are restricted to controlling symptoms, maintaining remission and preventing relapse. Efficacy in inflammatory bowel disease in the clinic may be measured as a reduction in the Crohn's Disease Activity Index (CDAI) score for CD which is scoring scale based on laboratory tests and a quality of life questionnaire. In animal models, efficacy is mostly measured by increase in weight and also a disease activity index (DAI), which is a combination of stool consistency, weight and blood in stool. TREM-1 antibodies of the invention may be suitable for use in the treatment of individuals with rheumatoid arthritis. Rheumatoid arthritis (RA) is a systemic disease that affects nearly if not all of the body and is one of the most common forms of arthritis. It is characterized by inflammation of the joint, which causes pain, stiffness, warmth, redness and swelling. This inflammation is a consequence of inflammatory cells invading the joints, and these inflammatory cells release enzymes that may digest bone and cartilage. As a result, this inflammation can lead to severe bone and cartilage damage and to joint deterioration and severe pain, among other physiologic effects. The involved joint can lose its shape and alignment, resulting in pain and loss of movement. There are several animal models for rheumatoid arthritis known in the art. For example, in the collagen-induced arthritis (CIA) model, mice develop an inflammatory arthritis that resembles human rheumatoid arthritis. Since CIA shares similar immunological and pathological features with RA, this makes it a suitable model for screening potential human anti-inflammatory compounds. Efficacy in this model is measured by decrease in joint swelling. Efficacy in RA in the clinic is measured by the ability to reduce symptoms in patients which is measured as a combination of joint swelling, eythrocyte sedimentation rate, C-reactive protein levels and levels of serum factors, such as anti-citrullinated protein antibodies. TREM-1 antibodies of the invention may be suitable for use in the treatment of individuals with psoriasis. Psoriasis is a T-cell mediated inflammatory disorder of the skin that can cause considerable discomfort. It is a disease for which there is currently no cure and it affects people of all ages. Although individuals with mild psoriasis can often control their disease with topical agents, more than one million patients worldwide require ultraviolet light treatments or systemic immunosuppressive therapy. Unfortunately, the inconvenience and risks of ultraviolet radiation and the toxicities of many therapies limit their long-term use. Moreover, patients usually have recurrence of psoriasis, and in some cases rebound shortly after stopping immunosuppressive therapy. A recently developed model of psoriasis based on the transfer of CD4+ T cells mimics many aspects of human psoriasis and therefore can be used to identify compounds suitable for use in treatment of psoriasis (Davenport et al., Internat. Immunopharmacol 2:653-672, 2002). Efficacy in this model is a measured by reduction in skin pathology using a scoring system. Similarly, efficacy in patients is measured by a decrease in skin pathology. TREM-1 antibodies of the invention may be suitable for use in the treatment of individuals with psoriatic arthritis. Psoriatic arthritis (PA) is a type of inflammatory arthritis that occurs in a subset of patients with psoriasis. In these patients, the skin pathology/symptoms are accompanied by a joint swelling similar to that seen in rheumatoid arthritis. It features patchy, raised, red areas of skin inflammation with scaling. Psoriasis often affects the tips of the elbows and knees, the scalp, the navel and around the genital areas or anus. Approximately 10% of patients who have psoriasis also develop an associated inflammation of their joints. The term “treatment”, as used herein, refers to the medical therapy of any human or other animal subject in need thereof. Said subject is expected to have undergone physical examination by a medical or veterinary medical practitioner, who has given a tentative or definitive diagnosis which would indicate that the use of said treatment is beneficial to the health of said human or other animal subject. The timing and purpose of said treatment may vary from one individual to another, according to many factors, such as the status quo of the subject's health. Thus, said treatment may be prophylactic, palliative, symptomatic and/or curative. In terms of the present invention, prophylactic, palliative, symptomatic and/or curative treatments may represent separate aspects of the invention. An antibody of the invention may be administered parenterally, such as intravenously, such as intramuscularly, such as subcutaneously. Alternatively, an antibody of the invention may be administered via a non-parenteral route, such as perorally or topically. An antibody of the invention may be administered prophylactically. An antibody of the invention may be administered therapeutically (on demand). EXEMPLARY EMBODIMENTS 1. A method of identifying a functional TREM-1 antibody, comprising (a) culturing a first cell expressing TREM-1, a signalling protein and a reporter construct; (b) measuring the activity of the first cell when said cell is incubated with a TREM-1 modifying agent; (c) contacting the culture of (b) with a TREM-1 antibody; and (d) measuring that the activity of the first cell is less than or more than the activity measured in (b).2. A method of identifying a blocking TREM-1 antibody, comprising (a) culturing a first cell expressing TREM-1, a signalling protein and a reporter construct; (b) measuring the activity of the first cell when said cell is incubated with an activated neutrophil; (c) contacting the culture of the first cell and the activated neutrophil with a TREM-1 antibody; and (d) measuring that the activity of the first cell is less than the activity measured in (b).3. The method of any one of embodiments 1-2, wherein the modifying agent of (b) is an activated neutrophil or a TREM-1 ligand.4. A method of identifying a stimulating TREM-1 antibody, comprising (a) culturing a first cell expressing TREM-1, a signalling protein and a reporter construct; (b) measuring the activity of the first cell; (c) contacting/incubating said cell with a TREM-1 antibody, and (d) measuring that the activity of the first cell is more than the activity measured in (b).5. The method of any one of embodiments 1-4, wherein the first cell is of haematopoetic origin.6. The method according to embodiment 5, wherein the cell of haematopoetic origin is a myeloid cell.7. The method according to embodiment 5, wherein the cell of haematopoetic origin is a T-cell.8. The method according to any one of embodiments 1-7, wherein the signalling protein is DAP10.9. The method according to any one of embodiments 1-7, wherein the signalling protein is DAP12.10. The method according to any one of embodiments 1-7, wherein the signalling protein is TCR zeta.11. The method according to any one of embodiments 1-7, wherein the signalling protein is Fc gamma RIII.12. The method according to any one of embodiments 1-7, wherein the signalling protein is a Fc receptor.13. The method according to any one of embodiments 1-6, wherein the reporter construct comprises a transcription factor and a reporter gene.14. The method according to embodiment 13, wherein said transcription factor is NFAT.15. The method according to embodiment 14, wherein said transcription factor is NFkB.16. The method according to any one of embodiments 13-15, wherein said reporter gene encodes β-galactosidase.17. The method according to any one of embodiments 13-15, wherein said reporter gene encodes luciferase.18. The method according to any one of embodiments 13-15, wherein said reporter gene encodes green fluorescent protein (GFP).19. The method according to any one of embodiments 13-15, wherein said reporter gene is a gene that encodes chloramphenicol transferase.20. A method of identifying a blocking TREM-1 antibody, comprising (a) culturing a T-cell expressing TREM-1, DAP12 and a gene that encodes luciferase; (b) measuring the luminescence of the T-cell when it is incubated with an activated neutrophil; (c) contacting the co-culture of (b) with a TREM-1 antibody; and (d) measuring that the luminescence of the T-cell is less than the activity measured in (b).21. The method according to embodiment 7, wherein said cell is a BWZ.36/hTREM-1:DAP12:NFAT-LacZ T-cell line.22. The antibody identified by the method of any one of embodiments 1-3 and 5-21.23. An antibody that is capable of specifically binding to TREM-1 and that is capable of blocking TREM-1 function.24. The antibody according to any one of embodiments 22-23, wherein said antibody is capable of preventing or reducing the dimerisation/multimerisation of TREM-1.25. The antibody according to any one of embodiments 22-24, wherein said antibody is capable of blocking the interaction between TREM-1 and its ligand.26. The antibody according to any one of embodiments 22-25, wherein said antibody is capable of blocking PGLYRP1-induced TREM-1 function.27. The antibody according to any one of embodiments 22-26, wherein the TREM-1 is human TREM-1.28. The antibody according to embodiment 27, wherein said antibody is also capable of specifically binding to and blocking the function of TREM-1 from another species than a human.29. The antibody according to embodiment 28, wherein the TREM-1 from another species is cynomolgus monkey TREM-1.30. The antibody according to embodiment 28, wherein the TREM-1 from another species is rhesus monkey TREM-1.31. The antibody according to any one of embodiments 22-30, which is capable of specifically binding K20A-hTREM-1-Cmyc2-His6 (SEQ ID NO: 13).32. The antibody according to any one of embodiments 22-31, which is capable of specifically binding A24T/Y28F/N30S/R32Q/P70H-cTREM-1-Cmyc2-His6 (SEQ ID NO: 14).33. The antibody according to any one of embodiments 22-32, which is capable of specifically binding A24T/Y28F/N30S/R320/E54K-cTREM-1-Cmyc2-His6 (SEQ ID NO: 15).34. The antibody according to any one of embodiments 22-33, which competes with mAb 0170 for binding to human TREM-1.35. The antibody according to any one of embodiments 22-34, which competes with mAb 0170 for binding to cynomolgus monkey TREM-1.36. The antibody according to any one of embodiments 22-35, which is capable of specifically binding a polypeptide comprising amino acids D38 to F48 of SEQ ID NO: 1 (human TREM-1), as determined using, eg., HX-MS.37. The antibody according to any one of embodiments 22-36, which has an epitope comprising one, two, three, four, five, six, seven or all of the amino acid residues selected from the group consisting of the D38, V39, K40, C41, D42, Y43, T44 and L45 of SEQ ID NO: 1 (human TREM-1) and one, two or all of the amino acid residues selected from the group consisting of the E46, K47 and F48 of SEQ ID NO: 1 (human TREM-1), as determined using, eg., HX-MS.38. The antibody according to any one of embodiments 22-37, which is capable of specifically binding a polypeptide comprising amino acid residues E38 to L45 of cynomolgus monkey TREM-1, as determined using, eg., HX-MS.39. The antibody according to any one of embodiments 22-38 which has an epitope comprising at least the amino acid residues selected from the group consisting of the D42, E46, D92 and H93 of SEQ ID NO: 1 (human TREM-1), as determined using surface plasmon resonance.40. The antibody according to any one of embodiments 22-39 which has an epitope comprising at least the amino acid residues E46 and/or D92 of SEQ ID NO: 1 (human TREM-1), as determined using surface plasmon resonance.41. The antibody according to any one of embodiments 22-40, wherein said antibody is capable of specifically binding (i) at least one amino acid residue selected from the group consisting of the A21, T22, K23, L24, T25, E26, and (ii) at least one amino acid residue selected from the group consisting of the A49, S50, S51, Q52, K53, A54, W55, Q56, I57, I58, R59, D60, G61, E62, M63, P64, K65, T66, L67, A68, C69, T70, E71, R72, P73, S74, K75, N76, S77, H78, P79, V80, Q81, V82, G83, R84, I85 and (iii) at least one amino acid residue selected from the group consisting of the C113, V114, I115, Y116, Q117, P118 and P119 of human TREM-1.42. The antibody according to any one of embodiments 22-40, wherein said antibody is capable of specifically binding (i) at least one amino acid residue selected from the group consisting of the V39, K40, C41, D42, Y43, T44, L45, E46, K47, F48, A49, S50, S51, Q52, K53, A54, W55, Q56, and (ii) at least one amino acid residue selected from the group consisting of the T70, E71, R72, P73, S74, K75, N76, S77, H78, P79, V80, Q81, V82, G83, R84, I85 and (iii) at least one amino acid residue selected from the group consisting of the and C113, V114, I115, Y116, Q117, P118, P119.43. The antibody according to any one of embodiments 22-35, wherein said antibody is capable of specifically binding human TREM-1, wherein the epitope of said antibody comprises one, two, three, four, five, six, seven, eight, nine or all of the amino acid residues K40, D42, T44, L45, E46, K47, Y90, I191, D92, H93, G94, L95 and R97 of SEQ ID NO: 1.44. The antibody according to any one of embodiments 22-35, wherein said antibody is capable of specifically binding human TREM-1, wherein the epitope of said antibody comprises the K40, D42, T44, L45, E46, K47, Y90, H91, D92, H93, G94, L95 and R97 of SEQ ID NO: 1.45. The antibody according to any one of embodiments 22-44, the heavy chain of which comprises a CDRH3 sequence corresponding to amino acid residues 101 to 110 (DMGIRRQFAY) of SEQ ID NO: 4, wherein one, two or three of said amino acid residues may be substituted by a different amino acid.46. The antibody according to any one of embodiments 22-44, the heavy chain of which comprises a CDRH3 sequence corresponding to amino acid residues 101 to 110 (DMGQRRQFAY) of SEQ ID NO: 6, wherein one, two or three amino acid residues may be substituted by a different amino acid.47. The antibody according to any one of embodiments 45-46, further comprising a CDRH1 sequence corresponding to amino acid residues 31 to 35 (TYAMH) of SEQ ID NO: 4 or SEQ ID NO: 6, wherein one of these amino acid residues may be substituted by a different amino acid residue; and/or a CDRH2 sequence corresponding to amino acids 50 to 68 (RIRTKSSNYATYYAASVKG) of SEQ ID NO: 4 or SEQ ID NO: 6, wherein one, two or three of said amino acids may be substituted by a different amino acid residue.48. The antibody according to any one of embodiments 45-47, the light chain of which comprises: a CDRL1 sequence corresponding to amino acid residues 24 to 38 (RASESVDTFDYSFLH) of SEQ ID NO: 5 or SEQ ID NO: 7, wherein one, two or three of these amino acid residues may be substituted with a different amino acid; and/or a CDRL2 sequence corresponding to amino acid residues 54 to 60 (RASNLES) of SEQ ID NO: 5 or SEQ ID NO: 7, wherein one or two of these amino acid residues may be substituted with a different amino acid; and/or a CDRL3 sequence corresponding to amino acid residues 93 to 101 (QQSNEDPYT) of SEQ ID NO: 5 or SEQ ID NO: 7, wherein one or two of these amino acid residues may be substituted with a different amino acid.49. The antibody according to any one of embodiments 45-48, comprising SEQ ID NO. 4 or SEQ ID NO: 6 and/or SEQ ID NO: 5 or SEQ ID NO: 7.50. The antibody according to any one of embodiments 22-49, which binds human TREM-1 with a binding affinity (KD) that is 1×10−7M or less, 1×10−8M or less, or 1×10−9M or less, or 1×10−10M or less, 1×10−11M or less, 1×10−12M or less or 1×10−13M or less, as determined using surface plasmon resonance.51. The antibody according to embodiment 50, wherein said binding affinity (KD) is 1×10−10M or less.52. The antibody according to any one of embodiments 22-51, which binds cynomolgus monkey TREM-1 with a binding affinity (KD) that is 1×10−7M or less, 1×10−8M or less, or 1×10−9M or less, or 1×10−10M or less, 1×10−11M or less, 1×10−12M or less or 1×103M or less, as determined using surface plasmon resonance.53. The antibody according to embodiments 52, wherein said binding affinity is 1×10−9M or less.54. The antibody according to any one of embodiments 22-53, which is an IgG.55. The antibody according to any one of embodiments 22-54 for use as a medicament.56. The antibody according to any one of embodiments 22-55 for the treatment of an autoimmune disease and/or chronic inflammation.57. The antibody according to any one of embodiments 22-54 for the manufacture of a medicament for the treatment of an autoimmune disease and/or a chronic inflammation.58. A method of treating an autoimmune disease and/or chronic inflammation comprising administering an antibody according to any one of embodiments 22-54 to a subject in need thereof.59. The use according to any one of embodiments 55-57 or the method according to embodiment 58, wherein said autoimmune disease is rheumatoid arthritis.60. The use according to any one of embodiments 55-57 or the method according to embodiment 58, wherein said autoimmune disease is Crohn's disease.61. The use according to any one of embodiments 55-57 or the method according to embodiment 58, wherein said autoimmune disease is ulcerative colitis.62. The use according to any one of embodiments 55-57 or the method according to embodiment 58, wherein said autoimmune disease is psoriatic arthritis.63. The use according to any one of embodiments 55-57 or the method according to embodiment 58, wherein said autoimmune disease is juvenile idiopathic arthritis. The present invention is further illustrated by the following examples which should not be construed as further limiting. The contents of all figures and all references, patents and published patent applications cited throughout this application are expressly incorporated herein by reference. EXAMPLES Example 1: Generation of a BWZ.36 humanTREM-1:DAP12 Stable Cell Line The BWZ.36/hTREM-1:DAP12:NFAT-LacZ cell line (herein also referred to as the “BWZ/hTREM-1 reporter cell”) was derived from BW5147 T cells (Mus musculusthymus lymphoma cell line. ATCC TIB-47, LGC Standards, Middelsex, UK) and contains a LacZ reporter construct regulated by four copies of the NFAT promoter element (see Karttunen, J. & Shastri, N. (1991) Proc. Natl. Acad. Sci. USA 88, 3972-3976 and Fiering, S., Northrop, J. P., Nolan, G. P., Matilla, P., Crabtree, G. R. & Herzenberg, L. A. (1990) Genes Dev. 4, 1823-1834). TREM/DAP12/pMX-IRES vector (encoding 786 bp of TREM-1 from a SmaI site to BamHI site using TREM-1 cDNA (Gene Bank Ref. ID: NM_018643.2, Sino Biological Inc., Beijing, China) as template and oligo 5′ TAGTAGGGATCCGCTGGTGCACAGGAAGG (SEQ ID NO: 16) and 5′ TAGTAGGCGGCCGCTTCGTGGCCTAGGGAC (SEQ ID NO: 17) as primers cloned into pREShyg vector GenBank Accession #U89672 (Cat. no. 6061-1, Clontech Laboratories, CA, USA) was transfected in PLAT-E packaging cell line (provided by W. Yokoyama, Washington University; alternatively, Cat. no. RV-101, Cell Biolabs Inc, Bio-Mediator KY, Vantaa, Finland) using Superfect transfection reagent (Cat. no. 301305, Qiagen Nordic, Denmark). PLAT-E supernatants containing TREM/DAP12/pMX-IRES viral particles were used to infect BWZ.36 cells as follows: 2×105BWZ.36 cells were cultured in 6 well plates and the medium was replaced with 1.5 ml of supernatant containing the viral particles+8 mg/ml of polybrene. After 6-8 hours, 1.5 ml of normal medium was added to the plate and the cells were incubated for an additional 24 hours. BWZ.36 cell lines stably expressing TREM-1 were stained with anti TREM-1 monoclonal antibody (clone 21C7) and isolated by cell sorting. Example 2: Cultivation of a BWZ.36 humanTREM-1:DAP12 Stable Cell Line BWZ/hTREM-1 reporter cell were cultured in RPMI 1640 w/o phenol red (Cat #11835, Gibco, Carlsbad Calif., USA), supplemented with 10% FCS (Cat #16140-071, Gibco, New York, USA), 1% Pen/Strep (Cat #15070-06, Gibco), 1 mM Sodium Pyruvate (Cat #11360, Gibco), 5 μM-2ME (Cat #31350-010, Gibco) and 2 mM L-Glutamine (Cat #25030, Gibco). No special plates or coating was required. 10 ml Versene (Cat #15040, Gibco) was added to detach the cells which then were transferred to tubes, centrifuged 1200 rpm 5 min and washed in fresh RPMI 1640 w/o phenol red. These cell were then ready to use in an assay or re-culture for further propagation. Example 3: Immunisation of Mice and Identification of mAbs In order to generate antibodies that bind human TREM-1, both wild type Balb/C mice and TREM-1 knock-out (KO) mice (C57BL/6 background) were immunised with either human (h) TREM-1 (SEQ ID NO:1), cells expressing hTREM-1 (BWZ.36 cells), or a combination of both. Primary screening was done either by means of direct ELISA on hTREM-1 protein or by means of FMAT, using BWZ.36 cells expressing hTREM-1. Secondary screening was done by flow cytometry on HEK293 cells expressing hTREM-1. Positive hybridoma supernatants were then screened in the BWZ/hTREM-1 reporter assay described in Example 4. The highest number of blocking antibodies was obtained from KO mice immunised with hTREM-1 protein six times at two weeks intervals, followed by a booster injection. In total, over 200 hTREM-1 antibodies were isolated, of which approximately 70 were subsequently found to have a blocking effect. All TREM-1 specific hybridoma supernatants were tested in the BWZ/hTREM-1 reporter assay first as supernatants and later as purified antibodies, in full titration from 5000 ng/ml down to 7 ng/ml, both as soluble and as platebound antibodies. Blood from a range of different donors was used as a source of fresh neutrophils. As an example,FIG.4shows antibodies from one fusion where the activity in the reporter assay as blocking activity is on the x-axis and the agonistic activity when the antibody is plate-bound is on the y-axis. Example 4: Identification of PGLYRP1 as a Neutrophil-Expressed TREM-1 Ligand PGLYRP1 was identified as a TREM-1 ligand through the use of immunoprecipitation coupled with mass spectroscopy (IP-MS). Soluble TREM-1 tetramer was used as an affinity “bait” molecule to identify a ligand. Briefly, TREM-1-tetramer-Fc (SEQ ID NO: 2) and separately CD83-Fc (SEQ ID NO: 5) were each incubated at final concentrations of 100 μg/ml with 270 million human neutrophils, purified by dextran sedimentation as described above, in 1 mL PBS at 4° C., 90 minutes with mild shaking. After pelleting these cells, the cells were resuspended in 1 mL PBS buffer with the inclusion of the crosslinker 3,3′-Dithiobis[sulfosuccinimidylpropionate] (DTSSP) (Thermo Scientific: 21578, Rockford, Ill., USA), at a concentration of 2 mM and incubated 30 minutes at room temperature, Cells were washed 3× with 1 mL PBS followed by lysis in 1 mL RIPA buffer (Thermo Scientific, 89901, Rockford, Ill., USA). The lysate was centrifuged at 15,000×g for 10 minutes at 4° C. to remove insoluble materials. Neutrophil proteins cross-linked to Fc coupled probes were immunoprecipitated from the supernatant using Protein A Mag Sepharose™ beads (GE Healthcare Life Sciences, 28-9670-56, Piscataway, N.J., USA). Briefly, 50 μL of beads were first washed with 200 μL PBS, then resuspended in 1 mL of cell lysate, incubated 60 minutes at 4° C., magnetically captured, and sequentially washed 2× with 200 μl RIPA buffer then 3× with 200 μL PBS. Upon removing PBS from the final magnetic capture, proteins were eluted from the magnetic beads using 200 μL buffer containing 8 M Urea, 100 mM Tris (pH 8.0), and 15 mM TCEP (Thermo Scientific, 77720, Rockford, Ill., USA) and incubated at room temperature for 30 minutes, beads were captured and supernatant was transferred to a Microcon Ultracel YM-30 filter (Millipore, 42410, Billerica, Mass., USA). Samples were spun at 14, 000×g, 20° C., 30-60 minutes until no liquid remained on the top of the filter membrane. The retained proteins were then alkylated with 100 μL 50 mM IAA (iodoacetamide) in 8 M Urea for 30 minutes in dark at room temperature. The filter was washed 2× with 100 μL 50 mM NH4HCO3and then transferred to a new collection tube. 1 μg trypsin (Promega, V5111, Madison, Wis.) in 60 μL 50 mM NH4HCO3was added followed by incubation at 37° C. overnight. The tryptic digest was collected by centrifugation at 14,000×g for 30 minutes followed by washing the filter with 50 μL 50 mM NH4HCO3. 10 μL of the digest was analyzed by LC/MS/MS using an LTQ-Orbitrap-XL mass spectrometer (Thermo Scientific, Waltham, Mass., USA). The data was searched against 1PI human database (v3.81) using SEQUEST-Sorcerer engine (4.0.4 build) (SageN, Milpitas, Calif., USA) and then post processed with Scaffold 3 (Proteome Software, Portland, Oreg., USA) to filter protein IDs with a false discovery rate of 1%. After negative control subtraction, PGLYRP1 was found to be a high-confidence protein specifically associated with hTREM-1 tetramer. The immunoprecipitation in the neutrophils showed that out of the 148 identified proteins, 72 proteins were immunoprecipitated by the control construct (CD83) alone, 73 of the proteins were identical for TREM-1 and CD83, whereas only three were TREM-1 specific (FIG.3). The experiment was subsequently repeated using neutrophils from a different donor and PGLYRP1 was again identified as specifically interacting with hTREM-1. Example 5: Refolding and Purification of Human PGLYRP1 Expressed fromE. coli Human PGLYRP1 was expressed as inclusion bodies inEscherichia coliBL21 (DE3) cells. Bacteria were harvested by centrifugation, resuspended in 50 mM Tris-HCl pH8.0, 500 mM NaCl, 5 mM EDTA, 0.5% Triton X-100 and disrupted by sonication. The insoluble pellet was washed three times with 50 mM Tris, pH 8.0, 1% TritonX-100, 2 M urea and once with 50 mM Tris pH 8.0, then solubilized in 50 mM Tris-HCl, 6M guanidine hydrochloride, pH7.4, 1 mM DTT (final protein concentration 20 mg/ml). For in vitro folding, solubilized human PGLYRP1 inclusion bodies were diluted into 50 mM Tris, pH 8.0, 2 mM EDTA, 5 mM cysteamine, 0.5 mM cystamine, 0.4 M arginine (final protein concentration 1 mg/ml). After overnight at 4° C., the folding mixture was cleared by centrifugation/filtration and then diluted 12 fold into 10 mM MES pH 3.5 to lower the conductivity and pH (final pH˜5.8, conductivity˜6 mS/cm). The diluted folding mixture was then applied to a Hitrap SP HP 5 ml column (17-1151-01 GE Healthcare, Uppsala, Sweden), followed by a 5 column volume wash with 50 mM MES pH 5.8. The bound human PGLYRP1 was then eluted with a 0-60% linear gradient of 50 mM MES pH 5.8, 1 M NaCl in 20 column volume. The fractions containing refolded human PGLYRP1 were pooled and concentrated to less than 4 ml by Amicon ultra 15 centrifugal units ((UFC800324 3,000 kDa MWCO, Millipore, Hellerup, Denmark). A Hiload 26/60 Superdex 75 318 ml column ((17-1070-01 GE Healthcare, Uppsala, Sweden) was then used to polish and buffer-exchange the proteins to Phosphate Buffered Saline (PBS). Majority of refolded human PGLYRP1 proteins was in monomer form. After concentrating, the final protein concentration was determined by measuring 280 nm absorbance with a NANODROP UV spectrometer. Protein purity was assessed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). Example 6: Creation of a TREM-1 Responsive Reporter Assay The TREM-1 reporter cell line was generated by transfecting the BWZ.36 cell line with a NFAT-LacZ reporter construct, as well as hTREM-1 and DAP12 (as described in Example 1). Neutrophils of healthy donors were purified by means of Dextran sedimentation. Blood was stratified on FicollPaque (17-0840-03, GE Healthcare, Piscataway, N.J., USA) gradient with a rate of 3 parts of Ficoll and 4 parts of blood in a 50 ml tube, then centrifuged at 400×g for 30 minutes at 22° C., without brake. The intermediate PBMC band was gently removed by aspiration. The neutrophils stratified on the packed RBC were aspirated and transferred to a 50 ml polypropylene tube. The neutrophils and contaminating RBCs were diluted to 40 ml with 1×PBS and followed by addition of 10 ml 4% DEXTRAN 500 (Sigma, 31392, St Louis, Mo., USA) in PBS solution. After mixing by gentle inversion, the tubes were left at 22° C. for 20-30 min. A granulocyte rich supernatant was then transferred into a fresh tube and centrifuged at 250×g. 5 min, 22° C.; the supernatant was aspirated and discarded. Contaminating RBCs were removed with an osmotic lysis, briefly, the cell pellet was resuspended in 7.5 mi of 0.2% NaCl; gently mixed for 55-60 seconds and 17.5 ml of a 1.2% NaCl solution was added. The volume was then brought to 50 ml with PBS and spun at 250×g for 5 min, the pellet was resuspended in 7.5 ml of 0.2% NaCl to repeat the lysis a second time. The final granulocyte pellet was resuspended in RPMI/10% FBS. These neutrophils were stimulated with PGN (InVivogen, tlrl-pgnsa, SanDiego, Calif., USA) overnight to generate activated neutrophils able to stimulate TREM-1. The BWZ/hTREM-1 reporter cells were then added to the PGN activated neutrophil cultures in a 1:3 ratio of reporter cell:neutrophils. Instead of activated neutrophils, a TREM-1 ligand complex consisting of PGLYRP1 (SEQ ID NO: 23) and PGN could be used to stimulate TREM-1. The assay was run in Poly-D-Lysine coated Black cell culture plates (no. 356640 from BD Biosciences, San Jose, Calif., USA). TREM-1 activation was read out after 24 hours of culture using the BetaGlo reagent (E4720 from Promega, Madison, Wis., USA) and luminescence measured using a TopCount Luminescence counter from Perkin Elmer. As positive control TREM-1 could be activated by a plate-bound TREM-1 antibody (R&D MAB1278, Minneapolis, Minn., USA) able to agonise TREM-1. Plates were coated with isotype control or TREM-1 antibody MAB1278 (3 ug/ml in PBS, 100 ul/well) in the fridge O/N or for 2 hr at 37° C., 5% CO2 before the BWZ/hTREM-1 reporter cells were added. After 6-24 hours incubation TREM-1 activation could be read using the BetaGlo reagent (E4720 from Promega, Madison, Wis., USA) and luminescence measured using a TopCount Luminescence counter from Perkin Elmer. This BWZ.36/hTREM-1:DAP12:NFAT-LacZ cell line (the “BWZ/hTREM-1 reporter cell”) showed to be highly responsive to antibody-mediated cross linking of TREM-1, giving a ˜40-fold induction of the NFAT-driven LacZ production when stimulated with 1-10 μg/ml plate bound commercially available anti-TREM-1 antibody, as compared to the isotype control (FIG.1). When stimulated with a toll-like receptor cocktail (tlrl-kit2hm, Invivogen, Sigma-Aldrich Denmark) alone (BWZ+TLR) no increase in signal was observed. Furthermore, unactivated neutrophils could not stimulate TREM-1, whereas TLR agonist cocktail (tlrl-kit2hm, Invivogen, Sigma-Aldrich Denmark) activated neutrophils could stimulate the BW7/hTREM-1 reporter cell. Table 1, below, shows that TREM-1 antibodies disclosed herein are able to block the ligand-induced TREM-1 activation in such BWZ/hTREM-1 reporter cell assay. TABLE 1AntibodyLuminescence x10E6BWZ+8.8activated neutrophilsMAB127810HPA7.9TREM269.9TREM376.114F1280.314F690.414F1160.714F110.414F1130.3 None of the tested commercial available antibodies: MAB1278 (cat. no. MAB1278, R&D Systems, Minneapolis, Minn. 55413, USA), anti-TREM-1 HPA (cat. no. HPA005563, Sigma, St Louis, Mo., USA), TREM26 (cat. no. 314902, Biolegend, San Diego, Calif. 92121, USA) and TREM37 (cat. no. 316102, Biolegend, San Diego, Calif. 92121, USA) were able to block the TREM-1 signal. Example 7: Epitope Mapping Using HX-MS Materials Protein Batches Used were: hTREM-1: human recombinant TREM-1, non-glycosylated, produced inE. coli. (cat. no. PRO-457, ProSpec-Tany TechnoGene Ltd., Rehovot, Israel). TABLE 2mAbs usedAntibodySuppliermAb 0023—mAb 0024—mAb 0025—mAb 0026—MAB1278RnD SystemsTREM26BioLegend All proteins were buffer exchanged to PBS pH 7.4 before experiments. Methods: HX-MS Experiments Instrumentation and Data Recording The HX experiments were automated by a Leap robot (H/D-x PAL: Leap Technologies Inc.) operated by the LeapShell software (Leap Technologies Inc.), which performed initiation of the deuterium exchange reaction, reaction time control, quench reaction, injection onto the UPLC system and digestion time control. The Leap robot was equipped with two temperature controlled stacks maintained at 20° C. for buffer storage and HX reactions and maintained at 2° C. for storage of protein and quench solution, respectively. The Leap robot furthermore contained a cooled Trio VS unit (Leap Technologies Inc.) holding the pre- and analytical columns as well as the pepsin column, the LC tubing and switching valves at 1° C. The switching valves of the Trio VS unit have been upgraded from HPLC to Microbore UHPLC switch valves (Cheminert, VICI AG). For the inline pepsin digestion, 100 μL quenched sample containing 200 pmol hTREM-1 was loaded and passed over the Poroszyme® Immobilised Pepsin Cartridge (2.1×30 mm (Applied Biosystems)) using a isocratic flow rate of 200 μL/min (0.1% formic acid:CH3CN 95:5). The resulting peptides were trapped and desalted on a VanGuard pre-column BEH C18 1.7 μm (2.1×5 mm (Waters Inc.)). Subsequently, the valves were switched to place the pre-column inline with the analytical column, UPLC-BEH(C18 1.7 μm (2.1×100 mm (Waters Inc.)), and the peptides separated using a 9 min gradient of 15-35% B delivered at 200 μl/min from an AQUITY UPLC system (Waters Inc.). The mobile phases consisted of A: 0.1% formic acid and B: 0.1% formic acid in CH3CN. The ESI MS data, and the separate data dependent MS/MS acquisitions (CID) and elevated energy (MSE) experiments were acquired in positive ion mode using a Q-TOF Premier MS (Waters Inc.). Leucine-enkephalin was used as the lock mass ([M+H]+ion at m. 556.2771) and data was collected in continuum mode (For further description of the set-up, see Andersen and Faber, Int. J. Mass Spec., 302, 139-148(2011)). Data Analysis Peptic peptides were identified in separate experiments using standard CID MS/MS or MSEmethods (Waters Inc.). MSEdata were processed using BiopharmaLynx 1.2 (version 017). CID data-dependent MS/MS acquisition was analyzed using the MassLynx software and in-house MASCOT database. HX-MS raw data files were subjected to continuous lock mass-correction. Data analysis, i.e., centroid determination of deuterated peptides and plotting of in-exchange curves, was performed using prototype custom software (HDX browser, Waters Inc.) and HX-Express ((Version Beta): Weis et al., J. Am. Soc. Mass Spectrom. 17, 1700 (2006)). All data were also visually evaluated to ensure only resolved peptide isotopic envelopes were subjected to analysis. Epitope Mapping Experiment Amide hydrogen/deuterium exchange (HX) was initiated by a 6-8 fold dilution of hTREM-1 in the presence or absence of mAb into the corresponding deuterated buffer (i.e. PBS prepared in D20, 96% 20 final, pH 7.4 (uncorrected value)). All HX reactions were carried out at 20° C. and contained 4 μM hTREM-1 in the absence or presence of 4 μM mAb thus giving a 2 fold molar excess of mAb binding sites. At appropriate time intervals ranging from 10 sec to 10000 sec, 50 μl aliquots of the HX reaction were quenched by 50 μl ice-cold quenching buffer (1.35M TCEP) resulting in a final pH of 2.5 (uncorrected value), Results and Discussion This experiment maps the epitopes of mAbs 0023, 0024, 0025, 0026 and the commercial mAbs MAB1278 (RnD Systems) and Clone26 (Biolegend) on hTREM-1. The HX time-course of 43 peptides, covering 94% of the primary sequence of hTREM-1, were monitored in the absence or presence of the eight different mAbs for 10 to 10000 sec. Exchange protection observed in the early time-points, e.g. <300 sec, relate to surface exposed amide protons and thus also relate to protein interfaces. In contrast, effects observed late in the time course are related to slow exchanging amide hydrogens and thus related to the structural core of the protein. Therefore, epitope effects appear in the early time points whereas structural stabilization effects will manifest as exchange reduction in late time points (Garcia, Pantazatos and Villareal. Assay and Drug Dev Tech. 2, 81 (2004); Mandell, Falick and Komives, Proc. Natl. Acad. Sci. USA, 95, 14705 (1998)). The observed exchange pattern in the early timepoints in the presence or absence of a given mAb can be divided into two different groups: One group of peptides display an exchange pattern that is unaffected by mAb binding. In contrast, another group of peptides in hTREM-1 show protection from exchange upon mAb binding. For example at 100 sec exchange with D2O, approx than 2 amides are protected from exchange in the region Y111-F126 of mAb 0023. Regions displaying such protection effects are assigned to the epitope region. Epitope Mapping of mAbs 0123 and 0026 mAbs 0023 and 0026 both induce identical alterations in the exchange profile of hTREM-1 and will be described together here. The regions displaying protection upon 0023/0026 binding encompass peptides covering residues T22-L96 and Y111-D127. However, by comparing the relative amounts of exchange protection within each peptide upon binding mAb 0023/0026 and the lack of epitope effects in peptides T25-F48, R84-Q112 and peptides starting at P118, the epitope can be narrowed to residues A21-E26, A49-I85 and C113-P119. Although distant in sequence, these regions are close in the 3D structure of hTREM-1. Epitope Mapping of mAb 0024 and Biolegend Clone 26 mAb 0024 and Clone26 from Biolegend both induce identical alterations in the exchange profile of hTREM-1 and will be described together here. The regions displaying protection upon mAb 0024 binding encompass peptides covering residues V101-Q112. By comparing the relative amounts of exchange protection within each peptide upon binding mAb 0024 and the lack of epitope effects in surrounding peptides, the epitope can be narrowed to residues Q104-Q112 (FIG.7B). Epitope Mapping of NNC mAb 0025 The regions displaying protection upon 0025 binding encompass peptides covering residues D38-M63, T70-L96 and Y111-D127 (FIG.7C). However, by comparing the relative amounts of exchange protection within each peptide upon binding 0254-0025 and the lack of epitope effects in peptides in surrounding regions, the epitope can be narrowed to residues V39-Q56, T70-85 and C113-P119. Although distant in sequence, these regions are close in the 3D structure of hTREM-1 (FIG.8). Eptiope Mapping of MAB1278 The regions displaying protection upon MAB1278 binding encompass peptides covering residues T70-L96 and V101-Q112 (FIG.7D). However, by comparing the relative amounts of exchange protection within each peptide upon binding MAB1278 and the lack of epitope effects in peptides in surrounding regions, the epitope can be narrowed to residues T70-185 and Q104-Q112. Although distant in sequence, these regions are close in the 3D structure of hTREM-1. The structural position of the epitopes of mAbs 0023/0026 and mAb 0025 are shown inFIG.7A. The epitope of mAbs 0023 and 0026 seems to reside primarily in β-sheets in the dimer interface of the hTREM-1 crystal structure dimer. The antagonism of these mAbs could be a result of preventing hTREM-1 dimerisation and thus signalling. Example 8: Determination of the Interaction Interface Between TREM-1 and mAb 0170 Epitopes were mapped on both recombinant human and cynomolgus monkey TREM-1 (hTREM-1 and cTREM-1, respectively). The hTREM-1 construct used in this example comprises the residues M1-H140 (SEQ ID NO: 18) and the cTREM-1 construct comprises the residues M1-R180 of (SEQ ID NO: 12) with six histidine residues added to the C-terminus and using the amino acid numbering from wild-type hTREM-1. Throughout this example the amino acids of cTREM-1 are numbered according to the analogous residue in hTREM-1, as illustrated inFIG.11. The numbering used in this example can be converted to the numbering in SEQ ID NO: 12 by subtracting 19 if the residue number is 58 or less and by subtracting 20 if the residue number is 60 or greater. As an example, the residue number E46 on cTREM-1 in this example corresponds to residue (46−19=27) E27 in SEQ ID NO: 12. The residue number on L96 on cTREM-1 in this example corresponds to residue (96−20=76) L76 in SEQ ID NO. 12. Solutions of TREM-1, alone or in the presence of mAb 0170, were diluted 25-fold in 97% deuterated hepes buffer (20 mM hepes, 150 mM sodium chloride, pH 7.4) Non-deuterated controls were prepared by diluting into protiated hepes buffer. The hydrogen exchange experiments were performed on a waters HDX nanoAcquity ultra-high performance liquid chromatography (UPLC) system (Waters Corporation, Milford, Mass., USA) which included the HD-x PAL auto sampler (LEAP Technologies Inc., Carrboro, N.C., USA) for automated sample preparation. The LC tubing, pre- and analytical columns and switching valves were located in a chamber cooled to 0.3° C. The trypsin digestion column was stored at 15° C. Hydrogen exchange reactions were performed at 20° C. Mass analysis was performed online using a Waters SYNAPT G2 HDMS mass spectrometer. A volume containing 100 pmol of human or cynomolgus TREM-1 (1.54-1.98 μl) with or without 120 pmol mAb 0170 was diluted into deuterated hepes buffer to a final volume of 50 μl. At the appropriate time intervals the entire volume was transferred to and quenched in 50 μl 1.35 mM Tris(2-carboxyethyl)phosphine adjusted to pH 2.4 and held at 3° C. 99 μl of the quenched solution was immediately injected and passed over a Porozyme immobilised pepsin column (2.1 mm×30 mm)(Applied Biosystems, Life Technologies Corporation, Carlsbad, Calif., USA) and trapped on a Waters VanGuard BEH C18 1.7 μm (2.1 mm×5 mm) column at 100 μl/min flowrate using a 5% (vol/vol) methanol and 0.1% formic acid mobile fase. The peptides were separated on a Waters UPLC BEH C18 1.7 μm (1.0 mm×100 mm) column using a 10-40% acetonitrile gradient containing 0.1% formic acid at a 40 μl/min flow-rate. The mass spectrometer was operated in positive ion mode with ion mobility separation enabled. The electrospray conditions were 3.2 kV capillary, 25 V sample cone, and 4 V extraction cone offsets, 850 ml/min flow of nitrogen desolvation gas heated to 350° C. and 50 ml/min cone gas flow. The source block was heated to 120° C. Lock-mass correction data was acquired using the I+ ion of Leucine-enkephalin (m/z 556.2771) as reference compound and applied during data analysis. For peptide identification MSE-type experiments using trap collision offsets of 6 V (low-energy) and 50 V (elevated energy) were performed. Deuterated samples were analysed using the 6 V low energy trap collision offset only. For further details see Andersen, M. D., Faber, J. H.,Int. J. Mass Spectrom. (2011), 302, 139-148. The MSE-data was analysed using Waters ProteinLynx Global Server 2.5 and peptides of hTREM-1 were identified that covered 80% of the protein sequence (Table 3) and peptides of cTREM-1 were identified that covered 100% of the protein sequence (Table 4). The HX-MS data files were analysed using Waters DynamX 1.0 that automatically applies lock-mass correction and determines the degree of deuterium incorporation in each peptide. In addition, all data was manually inspected to ensure correct peak assignment and calculation of deuterium incorporation Results A list of the peptides and their exchange patterns is provided in Table 3. When mAb 0170 bound hTREM-1, protection from exchange was observed in peptides covering the sequence from A21 to L96 and the epitope was consequently determined to be within this region. When taking into account peptides that show no protection from exchange upon binding of mAb 0170, the epitope could be narrowed to the regions D38-F48. The region from R84-L96 showed little to no exchange in the presence or the absence of mAb 0170 and it was not possible to conclude whether this region was part of the mAb 0170 binding epitope. The peptide K47-A68 didn't show protection from exchange upon binding of mAb 0170, but the peptide T44-C69 was protected when mAb 0170 was bound. The first two residues of a peptide back-exchanges quickly and exchange information for those residues is lost. It was concluded that at least one of the residues E46, K47, and F48 was important for the binding of mAb 0170. TABLE 3Results from HXMS epitope mappingof mAb 0170 on human TREM-1PeptidemAb 0170A21-L37NA21-D38NA21-V39NA21-C69EXT22-D38ND38-Q56EXD38-M63EXD38-L67EXD38-A68EXD38-C69EXV39-A68EXV39-C69EXD42-C69EXT44-C69EXK47-A68NA49-C69NI57-A68NI57-C69NI57-L87NL67-L87NA68-E88NA68-L96NC69-L87NC69-L96NT70-L87NT70-E88NT70-L96NR84-L96LEE88-L96LEQ104-L110NY111-M124NY111-F126NY111-D127NC113-F126NV114-F126NV114-D127NI115-F126NI115-D127NEX: Epitope region indicated by hydrogen exchange protection upon antibody binding. (Exhange difference (EX) > 0.8 deuterons)W: Weak exchange due to structural effects (0.1 < EX < 0.8).N: No protection from exchange upon antibody binding. (EX < 0.1)LE: Low intrinsic exchangemAb 0170 epitope on cTREM-1 mAb 0170 Epitope on cTREM-1 A list of the peptides and their exchange patterns is given in Table 4. When mAb 0170 bound to cTREM-1, protection from exchange was observed in peptides covering the sequence from E38 to A68 and the epitope was consequently determined to be within this region. When taking into account peptides that shown no protection from exchange upon binding of mAb 0170, the epitope could be narrowed to the regions E38-L45. This epitope corresponded well with the mAb 0170 epitope on hTREM-1 but was truncated by three residues. The peptide C44-T69 in hTREM-1 was protected upon binding of mAb 0170, but the peptides A44-L67 and A44-A68 that cover the corresponding sequence in cTREM-1 were not protected. Thus, whereas at least one of the residues 46, K47, and F48 in hTREM-1 contributed to the binding epitope the corresponding residues E46, K47, and Y48 were not involved in binding of mAb 0170 to cTREM-1. TABLE 4HXMS epitope mapping of mAb 0170on cynomolgus TREM-1PeptidemAb 0170T21-L37WL24-L37WT25-L37WT25-E38WE38-L67EXE38-A68EXV39-L67EXV39-A68EXK40-L67EXA44-L67WA44-A68WE46-L67NK47-L67NA68-L87WA68-L96WA68-Q97WK69-L87WK69-L96WV82-L96WE88-L96LEE88-Q97LEQ97-L103NQ104-L110NY111-C130WY111-L131WV114-L131WI115-C130WI115-L131WL131-T180WL13I-V182WV132-T180WV132-V182WY152-T180WV181-E189NV181-H206WI190-H206NT193-H206NV195-H206NT196-H206ND197-H206NEX: Epitope region indicated by hydrogen exchange protection upon antibody binding. (Exhange difference (EX) > 0.8 deuterons)W: Weak exchange due to structural effects (0.1 < EX < 0.8).N: No protection from exchange upon antibody binding. (EX < 0.1)LE: Low intrinsic exchange Example 9: Study of Interaction Kinetics for Anti TREM-1 Antibodies to Human and Cynomolgus TREM-1 by Surface Plasmon Resonance (SPR) Binding studies were performed on a ProteOn Analyzer (BioRad) that measures molecular interactions in real time through surface plasmon resonance. Experiments were run at 25° C. and the samples were stored at 15° C. in the sample compartment. The signal (RU, response units) reported by the ProteOn is directly correlated to the mass on the individual sensor chip surfaces in six parallel flow cells. Anti-human Fc monoclonal or anti-murine Fc polyclonal antibody from Biacore human or mouse Fc capture kits were immobilized in horizontal direction onto flow cells of a GLM sensor chip according to the manufacturer's instructions. The final immobilization level of capture antibody was approximately 2600-6000 RU in different experiments. Capture of purified monoclonal mouse or recombinant expressed anti-hTREM-1 antibodies was conducted by diluting the antibodies to 5-10 nM into running buffer (10 mM Hepes 0.15 M NaCl, 5 mM EDTA, 0.05% surfactant P20, pH 7.4) and injected in vertical direction at 30 μl/min for 60 s, creating reference interspots adjacent to all flow cells with only anti-Fc antibody immobilized. This typically resulted in final capture levels of test antibodies of approximately 100-300 RU and Rmax values of analyte of 30-90 RU. Binding of hTREM-1 or cTREM-1 proteins was conducted by injecting analyte (antigen) over all flow cells in horizontal direction to allow for comparative analyses of binding to different captured anti-TREM-1 antibodies relative to binding to the reference interspot. hTREM-1 or cTREM-1 proteins was diluted serially 1:3 to 1.2-100 nM or into running buffer, injected at 100 μl/min for 250 s and allowed to dissociate for 600 s. The GLM surface was regenerated after each injection cycle of analyte via two 18 s injections of 10 mM Glycine, pH 1.7 and 50 mM NaOH at 100 μl/min. This regeneration step removed the anti-TREM-1 antibody and any bound TREM-1 protein from the immobilized capture antibody surface, and allowed for the subsequent binding of the next interaction sample pair. The regeneration procedure did not remove the directly immobilized anti-Fc capture antibody from the chip surface. Binding affinity between antibodies and the antigen was quantified by determination of the equilibrium dissociation constant (KD) determined by measurement of the kinetics of complex formation and dissociation. The rate constants corresponding to the association and the dissociation of a monovalent complex such as ka(association rate) and kd(dissociation rate) were retrieved by fitting data to 1:1 Langmuir model using the ProteOn evaluation software for data analysis. KDis related to kaand kdthrough the equation KD=kd/ka. Binding curves were processed by double referencing (subtraction of reference surface signals as well as blank buffer injections over captured anti-TREM-1 antibodies) prior to data analysis. This allowed correction for instrument noise, bulk shift and drift during sample injections. TABLE 5Results from measurements of binding constants ka (association rate),kd (dissociation rate) and KD (equilibrium dissociation constant)for the interaction of human TREM-1 to different anti-TREM-1 monoclonal antibodies.mAbCloneOriginka (1/(Ms)kd (1/s)KD (M)mAb 0023SF8A1Hybridoma1.0E+052.2E−042.1E−09mAb 0024SF28A1Hybridoma7.8E+043.6E−044.6E−09mAb 0025SF19A1Hybridoma8.8E+058.5E−049.7E−10mAb 00265F27A1Hybridoma6.2E+049.7E−051.5E−09mAb 00305F17A1B2Hybridoma1.2E+054.2E−043.6E−09mAb 003113F6A1Hybridoma1.2E+063.7E−033.1E−09mAb 003213F10A1Hybridoma1.1E+062.1E−031.9E−09mAb 00339F11A1Hybridoma7.8E+041.1E−031.5E−08mAb 003414F1741Hybridoma1.9E+051.9E−041.0E−09mAb 00395F8A1 on mIgG2aaRecombinant8.9E+041.6E−041.8E−09mAb 00405F8A1 on mIgG1Recombinant9.3E+042.0E−042.2E−09mAb 00415F27 on mIgG2aaRecombinant7.9E+041.8E−042.3E−09mAb 0042SF27A1 on mIgG1Recombinant8.5E+042.5E−042.9E−09mAb 004414F69A1Hybridoma1.5E+062.9E−042.0E−10mAb 004513F14A1Hybridoma1.3E+063.2E−032.5E−09mAb 004614F70A1Hybridoma9.3E+051.9E−042.1E−10mAb 004814F11A1B1Hybridoma1.7E+063.9E−032.4E−09mAb 004914F86A1Hybridoma8.8E+051.3E−031.5E−09mAb 00515F24 HC645/LC647Recombinant6.8E+053.0E−034.5E−09mAb 005414F128A1Hybridoma1.6E+063.8E−032.4E−09mAb 005914F113/14F69Recombinant1.7E+065.2E−043.2E−10mAb 006314F116A1B1Hybridoma1.0E+061.4E−031.4E−09mAb 006414F20A1B1Hybridoma8.9E+051.3E−031.5E−09mAb 006714F11A1B1Recombinant2.2E+063.9E−032.0E−09mAb 006814F128/14F11Recombinant2.7E+063.9E−031.7E−09mAb 007014F113/14F69Recombinant2.1E+064.3E−042.5E−10mAb 007814F106A2Hybridoma9.8E+051.4E−031.5E−09mAb 007914F29A1B1Hybridoma2.6E+051.2E−034.6E−09mAb 008014F17A1B1Hybridoma2.7E+051.9E−047.1E−10mAb 008314F116A1B1Recombinant1.0E+061.7E−031.7E−09mAb 009014F113 fully bumanizedRecombinant2.1E+061.5E−037.7E−10mAb 011514F113HC/14F128LC mixedRecombinant1.8E+062.4E−021.6E−08and humanizedmAb 012014F113/14F69 hz variantRecombinant2.2E+061.5E−036.7E−10HC A78LmAb 012114F113/14F69 hz variantRecombinant2.8E+061.7E−036.3E−10HC T93VmAb 012214FT13/14F69 hz variantRecombinant2.5E+061.4E−036.0E−10LC M4LmAb 012414F113/14F69 hz variantRecombinant2.0E+061.2E−036.1E−10LC G68RmAb 017014F113/14F69 bz variantRecombinant2.9E+065.4E−041.9E−10LC Q981 TABLE 6Results from measurements of binding constants ka (association rate).kd (dissociation rate) and KD (equilibrium dissociation constant) for theinteraction of cynomolgus TREM-1 to different anti-TREM-I monoclonal antibodies.mAbCloneOriginka (1/(Ms)kd (1/s)KD (M)mAb 004814F11A1B1Hybridoma1.48+051.7E−041.2E−09mAb 005914F113A1B1C1Recombinant1.8E+051.2E−046.7E−10mAb 006714F11A1B1Recombinant3.8E+051.7E−034.7E−09mAb 00684F128/14F11Recombinant2.88+053.4E−031.2E−08mAb 008314F116A1B1RecombinantNo bindingNo bindingNo bindingmAb 009014F113 fully humanizedRecombinant:1.4E+053.0E−042.1E−09mAb 012114F113 hz variant HCRecombinant1.3E+051.3E−049.4E−10T93VmAb 012214F113 hz variant LCRecombinant1.7E+051.6E−049.7E−10M4LmAb 012414F113 hz variant LCRecombinant1.7E+052.0E−041.1E−09G68RmAb 017014F113 hz variant LCRecombinant2.5E+0S8.9E−043.6E−09Q981 Example 10: Humanisation of the Blocking TREM-1 mAb 14F69 The variable regions of two lead antibodies were obtained from cloning of hybridomas 14F128A1 and 14F113A1B1C1. Both antibodies were cloned using the SMARTER-RACE technique (Clontech). The humanization effort was performed as an iterative loop where CDR grafted antibodies were first affinity evaluated and then re-engineered to include more backmutations until an acceptable affinity was retained, using hybridoma purified antibodies as a benchmark. The CDR grafted antibodies were designed in silico and ordered from a commercial vendor (www.genscript.com). Subsequent re-engineering of antibodies was performed using site directed mutagenesis (Stratagene). All antibodies were expressed in HEK293-6E cells in preparation for affinity testing. Below is a description of the main considerations for selection of appropriate human germline and test of backmutations. All numbering of variable regions used in this example refers to the Kabat numbering scheme. >m14F128A1_H (CDRs marked with bold and underlined) (SEQ ID NO: 8) >m14F128A1_L (CDRs marked with bold) (SEQ ID NO: 9) >m14F113A1B1C1_H (CDRs marked with bold) (SEQ ID NO: 10) >m14F113A1B1C1_L (CDRs marked with bold) (SEQ ID NO: 11) From an analysis of the sequences, the CDRs for m14F128A1 according to Kabats definition are: >CDR_H1 TYAMH >CDR_H2 RIRTKS[N/S]NYATYY[V/A]DSVKD >CDR_H3 DMG[I/A]RRQFAY >CDR_L1 RASESVD[S/T]F[G/D][I/Y]SF[M/L]H >CDR_L2 RASNLES >CDR_L3 QQSNEDPYT With the differences between m14F128A1 and m4F113A1B1C1 given as [m14F128A1/m14F113A1B1C1]. A 3D model of m14F128A 1 was build using standard techniques in MOE [available from www.chemcomp.com] and all residues within 4.5 Å of the effective CDR regions (VH: 31-35B, 50-58, 95-102; VL: 24-34, 50-56, 89-97) were defined as mask residues. Mask residues are all potentially important for sustaining the binding in the CDRs. The mask residues included positions 1-2, 4, 27-37, 47, 49-59, 69, 71, 73, 78, 92-103 for the heavy chain and positions 1-5, 7, 23-36, 46, 48-56, 58, 62, 67-71, 88-98 for the light chain. Using germline searches of m14F128A1 and manual inspection, VH3_73 and JH4 were identified as being an appropriate human germline combination for the heavy chain and VKIV_B3 and JK2 were identified as the appropriate human germline combination for the light chain. >VH3_13/JH4 >VH3_13/JH4EVQLVESGGGLVQPGGSLKLSCAASGFTFSGSAMHWVRQASGKGLEWVGRIRSKANSYATAYAASVKGRFTISRDDSKNTAYLQMNSLKTEDTAVYYCTR/YFDYWGQGTLVTVSS>VKIV B3/JK2DIVMTQSPDSLAVSLGERATINCKSSQSVLYSSNNKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYSTP/YTFGQGTKLEIKRResidues outside the mask were taken as human.Residues inside the mask and inside the Kabat CDR were taken as murine.Residues inside the mask and outside the Kabat CDR with mouse/germline consensus were taken as the consensus sequence.Residues inside the mask and outside the Kabat CDR with mouse/germline difference were subject to potential back mutations. Grafting the effective CDR regions of m14F128A1 into the germlines formed the basic humanisation construct of m14F128A1, hz14F128A1. >hz14F128A1_H >hz14F128A1_HEVQLVESGGGLVQPGGSLKLSCAASGFTFSTYAMHWVRQASGKGLEWVGRIRTKSNNYATYYAASVKGRFTISPDDSKNTAYLQMNSLKTEDTAVYYCTRDMGIRRQFAYWGQGTLVTVSS>hz14F128A1_LDIVMTQSPDSLAVSLGERATINCRASESVDSFGISFMHWYQQKPGQPPKLLIYRASNLESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSNEDPYTFGQGTKLEIK>CDR_H1TYAMH>CDR_H2RIRTKSNNYATYYAASVKG>CDR_H3DMGIRRQFAY>CDR_L1RASESVDSFGISFMH>CDR_L2RASNLES>CDR_L3QQSNEDPYT >CDR_H1 TYAMH CDR_H2 RIRTKSNNYATYYAASVKG >CDR_H3 DMGIRRQFAY >CDR_L1 RASESVDSFGISFMH >CDR_L2 RASNLES >CDR_L3 QQSNEDPYT The only differences compared to the murine CDRs were in CDR_H2 (shown in bold). Any discrepancy between m14F128A and hz14F128A1 in a mask residue will create a potential backmutation and the list includes: hz14F128A1_H: S30N, G49A, A78L, V93T hz14F128A1_L: M4L, M58I, G68R Furthermore, the close homology of m14F128A1 and m14F113A1B1C1 was used to suggest residues that could impact the affinity of hz14F128A1. hz14F128A1_H: N53S, 198Q hz14F128A1_L: S27D_T, G29D, I30Y, M33L In order to investigate all potentially humanised mAbs all combinations of the above mutants were produced and tested. The final humanised anti hTREM1 antibody (mAb 0170) derived from hybridoma 14F113 contains one LC framework-backmutation (M4L) and one HC CDR3 mutation (Q98I). The mutation in HC CDR3 was introduced based on an affinity-synergy-study with a highly homologous antibody named 14F128. The rationale for including both mutations is described below. Affinity-Synergy Study of Antibody 14F128 and 14F113 The hybridoma antibodies 14F128 and 14F113 are highly homologous and derived from the same somatic recombination event. The two antibodies compete in hTREM1 binding with 14F113 having the highest affinity. In total the CDR grafted versions of the two antibodies differ in their CDR compositions by only six amino acids (four in LC CDR, and two in HC CDR). The six mutations, when comparing 14F113 to 14F128, are LC T27dS, D29G, Y30I, L33M and HC S54N, Q98I. Although CDR grafted 14F128 had an affinity inferior to CDR grafted 14F113 it was investigated if a beneficial affinity effect from one or more of the six mutations was suppressed by the overall effect when all six mutations were present. All six mutations (except HC S54N) were therefore individually introduced in the CDR grafted 14F113 antibody and the antibodies were ranked by affinity. Two mutations (LC L33M and HC Q98I) were individually capable of improving the affinity of CDR grafted 14F113. A mutation of the HC at position Q98I gave rise to a particularly good affinity of the resultant antibody (mAb 0170). Framework Backmutation Affinity Analysis The mouse version of the 14F113 antibody had seven mutations that were potentially necessary to include as backmutations during humanisation. The potential backmutations in the HC and LC were S30N, G49A, A78L and T93V M4L, V58I, G68R, respectively. The seven backmutations were introduced individually in CDR grafted 14F113 and then ranked by affinity. Although several of the mutations were capable of improving affinity, only LC mutation M4L was selected for mAb 0170 The decision to include mutations was balanced against expression titer (HEK293 6E), affinity, and the total number of mutations. Example 11: Study of Interaction Kinetics for TREM-1 Antibodies to hTREM-1 by Surface Plasmon Resonance (SPR): Comparison Between mAb 0170 and Commercially Available TREM-1 Antibodies Binding studies were performed on a ProteOn Analyzer (BioRad) that measures molecular interactions in real time through surface plasmon resonance. Experiments were run at 25° C. and the samples were stored at 15° C. in the sample compartment. The signal (RU, response units) reported by the ProteOn is directly correlated to the mass on the individual sensor chip surfaces in six parallel flow cells. Commercially available antibodies included were Biolegend #314907, Biolegend #316102 (Biolegend, USA), Hycult Biotech H11M2252 (Hycult Biotech, Netherlands), R&D #MAB1278 (R&D systems. United Kingdom), SC98Z12 (Santa Cruz Biotechnology, USA), Sigma #WH0054210m4, Sigma #SAB1405121 (Sigma-Aldrich Danmark A/S) Anti-human Fc monoclonal or anti-murine Fc polyclonal antibody from Biacore human or mouse Fc capture kits were immobilized in horizontal direction onto flow cells of a GLM sensor chip according to the manufacturer's instructions. The final immobilization level of capture antibody was approximately 2600-6000 RU in different experiments. Capture of purified monoclonal mouse or recombinant expressed humanized anti-hTREM-1 antibodies was conducted by diluting the antibodies to 5-10 nM into running buffer (10 mM Hepes 0.15 M NaCl, 5 mM EDTA, 0.05% surfactant P20, pH 7.4) and injected in vertical direction at 30 μl/min for 60 s, creating reference interspots adjacent to all flow cells with only anti-Fc antibody immobilized. This typically resulted in final capture levels of test antibodies of approximately 100-300 RU and Rmax values of analyte of 30-90 RU. Binding of hTREM-1 or cTREM-1 proteins was conducted by injecting analyte over all flow cells in horizontal direction to allow for comparative analyses of binding to different captured anti-TREM-1 antibodies relative to binding to the reference interspot. hTREM-1 or cTREM-1 proteins was diluted serially 1:3 to 1.2-100 nM or into running buffer, injected at 100 μl/min for 210 s and allowed to dissociate for 600 s. The GLM surface was regenerated after each injection cycle of analyte via two injections of 10 mM Glycine, pH 1.7 and 50 mM NaOH at 100 μl/min. This regeneration step removed the anti-TREM-1 antibody and any bound TREM-1 protein from the immobilized capture antibody surface, and allowed for the subsequent binding of the next interaction sample pair. The regeneration procedure did not remove the directly immobilized anti-Fc capture antibody from the chip surface. Binding affinity between antibodies and the antigen was quantified by determination of the equilibrium dissociation constant (KD) determined by measurement of the kinetics of complex formation and dissociation. The rate constants corresponding to the association and the dissociation of a monovalent complex such as ka(association rate) and kd(dissociation rate) were retrieved by fitting data to 1:1 Langmuir model using the ProteOn evaluation software 3.1.0.6 for data analysis. KDis related to kaand kdthrough the equation KD=kd/ka. Binding curves were processed by double referencing (subtraction of reference surface signals as well as blank buffer injections over captured anti-REM-1 antibodies) prior to data analysis. This allowed correction for instrument noise, bulk shift and drift during sample injections. TABLE 7Results from measurements of KDdissociation constant) for the interaction of human cynomolgusTREM-1 to different anti-TREM-1 monoclonal antibodies.human TREM-1cyno TREM-1AntibodyKD (M)KD (M)mAb 01702E−103E−09Biolegend #3149079E−104E−09Biolegend #3161028E−10No bindingHycult Biotech HM22523E−09No bindingR&D #MAB12788E−09No bindingSC98Z123E−08No bindingSigma #WH0054210m42E−08No bindingSigma #SAB1405121No bindingNo binding Example 12: Competition Binding Studies of Anti-Human TREM-1 Monoclonal Antibodies by Surface Plasmon Resonance SPR binding competition studies were performed with monoclonal mouse or recombinant expressed humanised anti-hTREM-1 antibodies in order to discriminate between different binding sites (epitopes). Commercially available antibodies included were Biolegend #4314907 (Biolegend, USA) and SC98Z12 (Santa Cruz Biotechnology, USA). Anti hTREM-1 monoclonal antibodies that compete for the same or an overlapping binding site (epitope) on the antigen are not able to bind simultaneously to the antigen and are therefore assigned to the same “bin”. Anti-TREM-1 monoclonal antibodies that do not compete for the same or overlapping binding site on the antigen are able to bind simultaneously and are thus assigned to different “bins”. Experiments were performed with soluble, human TREM-1 extracellular domain as antigen. All studies were run at 25° C., and the samples were stored at 15° C. in the sample compartment. Individual anti-TREM-1 monoclonal antibodies and an unrelated control monoclonal antibody were immobilised onto separate flow cells of a GLC sensor chip using a 1:1 mixture of 0.4 M EDAC [1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride] and 0.1 M Sulfo-NHS [N-hydroxysulfosuccinimide]. Each antibody was diluted in 10 mM sodium acetate pH 5.0 to a concentration of 25 or 10 (SC98Z12 (80394)) μg/ml, and was immobilised to an individual flow cell at 30 μl/min for 240 s. The antibodies were immobilised to flow cells L1-L6 (including control). After immobilisation of the antibody, the active sites on the flow cell were blocked with 1 M ethanolamine. Immobilisations were performed with activation and deactivation in a horizontal direction creating interspot reference points without immobilised protein. The final immobilisation level of test antibodies ranged from approximately 1100 to 1300 RU in one experiment, except for one antibody (SC98Z12) where only 390 RU was immobilised. Recombinant human TREM-1 was diluted to 100 nM into running buffer (10 mM Hepes 0.15 M NaCl, 5 mM EDTA, 0.05% surfactant P20, pH 7.4). The antigen was injected over immobilised antibodies in horizontal direction at 30 μl/min for 300 s, allowing control of potential unspecific binding both to interspot references and immobilised control antibodies resulting in 150-600 RU captured TREM-1, except for to one antibody (SC98Z12) with low immobilisation level where only 4 RU was captured. Each antibody (the same ones that had been immobilised) was injected over parallel flow cells in a horizontal direction to allow for comparative analysis of binding to hTREM-1 captured by the primary antibodies relative to binding to both the interspot references and the immobilised control antibodies. Each competing antibody was diluted to 100 nM and injected at 100 μl/min for 250 s. The GLC chip was regenerated after each injection cycle of analyte via two 18 s injections of 1M Formic acid pH 3.5, 3M MgCl2 and 50 mM NaOH at 100 μl/min. This regeneration step removed the TREM-1 antigen and any bound secondary antibody from the immobilised antibody surface, and allowed for the subsequent binding of the next test sample. The regeneration procedure did not remove the directly immobilised anti-TREM-1 test antibody (primary antibody) from the chip surface. Data analysis was performed with the ProteOn Manager™ 3.1.0.6 Software. Capture levels were assessed to ensure that the regeneration step provided a consistent binding surface throughout the sequence of injections. No significant non-specific binding of human TREM-1 neither to the interspot control surfaces nor to immobilised control antibody was observed. Binding curves were processed by subtraction of interspot control surface signals. This allowed correction for instrument noise and bulk shift during sample injections. The competition results were reported as either positive or negative binding (Table 8). Positive (+) binding indicates that the competing antibody was capable of binding the hTREM-1 simultaneously with the primary antibody (i.e. they do not compete), and the primary and competing antibodies were consequently assigned to different epitope bins. Negative binding indicates that the competing antibody was unable to bind the hTREM-1 simultaneously with the primary antibody (i.e. they do compete), and the primary and competing antibodies were thus assigned to the same epitope bin. The response values in these experiments were significant and allowed for an unambiguous determination of epitope bins of the anti-TREM-1 monoclonal antibodies. TABLE 8Ability to bind (+) or to compete (−) for antibodies tested inSPR competition assay. SC98Z12 did not give high enoughcapture of TREM-1 to evaluate as primary antibody (*).PrimaryBiolegendmAbmAbSecondary#314907SC98Z1200480170Biolegend #314907−*++SC98Z12−*++mAb 0048+*−−mAb 0170+*−− MAb 0170 and mAb 0048 (purified from hybridoma 14F11, which is identical to 14F128) were shown to compete for binding to human TREM-1. Biolegend #314907 and SC98Z12 did not compete with any of these for human TREM-1 binding but competed with each other. These findings conclude that the first two (mAb 0048 and mAb 0170) belong to the same bin (Bin1) while Biolegend #314907 and SC98Z12 belong to another bin (Bin2). Example 13: Kinetic Analysis of the Interaction Between Fab 0011 and Fab 0170 Binding to Mutated Versions of Human and Cynomolgus TREM-1 Interaction studies were performed by SPR to define differences in the epitopes for 0011 and 0170 anti-humanTREM-1 antibodies on human TREM-1. By comparing binding kinetics to human M-variants with introduced Alanine mutations in known epitopes, as well as partly“humanised” variants of cynomolgus TREM-1, the latter since only mAb 0170 cross reacts with cynomolgus TREM-1 amino acid residues unique for respective epitope were identified. The hTREM-1 extracellular domain alanine mutant constructs and partly humanised cynomolgus mutant constructs used in this study are summarised in Table 9. All constructs used were variants either of SEQ ID NO:19 (cynomolgus TREM-1 variants) or SEQ ID NO: 20 (human TREM-1 variants) and included a C-terminal-cmyc2-His6tag for capture in SPR binding kinetics assay. Unless otherwise stated, sequences referred to in this example are numbered according toFIG.11. TABLE 9TREM-1variantProteinMutations0221human TREM-1 Ala mut 1D38A0222human TREM-1 Ala mut 2K40A0223human TREM-1 Ala mut 3D42A0224human TREM-1 Ala mut 4T44A0225human TREM-1 Ala mut 5E46A0226human TREM-1 Ala mut 6K47A0227human TREM-1 Ala mut 7S50A0228human TREM-1 Ala mut 8R84A0229human TREM-1 Ala mut 9Y90A0230human TREM-1 Ala mut 10H91A0231human TREM-1 Ala mut 11D92A0232human TREM-1 Ala mut 12H93A0233human TREM-1 Ala mut 13R97A0234human TREM-1 Ala mut 14R99A0235human TREM-1 Ala mut 15D127A0236human TREM-1 Ala mut 16R128A0237human TREM-1 Ala mut 17R130A0238cynomolgus TREM-1 wt—0239cynomolgus TREM-1 “partlyA44T, Y48F, N50S,humanised”R52Q, E75K, P91H0240cynomolgus TREM-1 “partlyA44A, Y48F, N50S,humanised with back mutation 1”R52Q, E75K, P91H0241cynomolgus TREM-1 “partlyA44T, Y48Y, N50S,humanised with back mutation 2”R52Q, E75K, P91H0242cynomolgus TREM-1 “partlyA44T, Y48F, N50N,humanised with back mutation 3”R52Q, E75K, P91H0243cynomolgus TREM-1 “partlyA44T, Y48F, N50S,humanised with back mutation 4”R52R, E75K, P91H0244 (SEQcynomolgus TREM-1 “partlyA44T, Y48F, N50S,ID NO: 14)humanised with back mutation 5”R52Q, E75E, P91H0245 (SEQcynomolgus TREM-1 “partlyA44T, Y48F, N50S,ID NO: 15)bumanised with back mutation 6”R52Q, E75K, P91P0247human TREM-1 wt— Binding studies were performed on a ProteOn Analyzer that measures molecular interactions in real time through surface plasmon resonance. Experiments were run at 25° C. and the samples were stored at 15° C. in the sample compartment. The signal (RU, response units) reported by the ProteOn is directly correlated to the mass on the individual sensor chip surfaces in six parallel flow cells. Anti-His monoclonal antibody was immobilised onto 6 parallel flow cells of a GLM sensor chip using a 1:1 mixture of 0.4 MEDAC [1-ethyl-3-(3-dimethylaminopropyl) carbodimide hydrochloride] and 0.1 M Sulfo-NHS [N-hydroxysulfosuccinimide]. Antibody was diluted in 10 mM sodium acetate pH 5.0 to a concentration of 25 μg/ml, and was immobilised onto individual flow cells at 30 μl/min for 240 s. After immobilisation of the antibody, the active sites on the flow cells were blocked with 1 M ethanolamine. Immobilisation was performed with all steps in horizontal direction. The final immobilisation level of capture antibody was approximately 8000 RU in one experiment. Cell culture medium from HEK 293 cells expressing wild type or different mutated variants of human or cynomolgus TREM-1 ECD was diluted 40-60 times in running buffer (10 mM HEPES, 150 mM NaCl, 3 mM EDTA, 0.05% surfactant P20, pH 7.4). TREM-1 proteins were injected over immobilised anti-His capture antibody in the vertical direction at 30 μl/min for 60 s. This resulted in 50-250 RU captured TREM-1 and created interspot references with only immobilised capture antibodies but no captured TREM-1 in the horizontal direction. Each Fab was injected over parallel flow cells in the horizontal direction to allow for kinetic analysis of binding to TREM-1 variants captured by anti-His antibody. Prior to injection each Fab was diluted to 0, 5.5 (in one experiment), 16.7 and 50 nM in running buffer, and injected at 100 μl/min for 250 s (association time). The dissociation time following these injections was monitored for 10 minutes. The GLM chip was regenerated after each interaction cycle of TREM-1 and Fab via two 18 s injections of 10 mM Glycine and 50 mM NaOH at 100 μl/min. This regeneration step removed the TREM-1 variants and any bound Fab from the anti-His antibody surface, and allowed for the subsequent binding of the next interaction pair. The regeneration procedure did not remove the directly immobilised anti-His capture antibody from the chip surface. In order to obtain kinetic data, such as ka (association rate), kd (dissociation rate) and KD (equilibrium dissociation constant), data analysis was performed using the ProteOn Manager™ 3.1.0.6 Software. Capture and binding levels of samples run in duplicates or triplicates were assessed to ensure that the regeneration step provided a consistent binding surface throughout the sequence of injections. No significant unspecific binding to interspot references with only immobilised capture antibody was observed. Binding curves were processed by subtraction of interspot control surface signals, as well as injection of running buffer. This allowed correction for instrument noise and bulk shift during sample injections. The affinity of 0170 Fab and 0011 Fab to different TREM-1 ECD variants where compared to the affinity to wild type human or cynomolgus TREM-1 ECD. The level of binding 10 s after end of injection of Fab, normalised to level of captured TREM-1 variant, was also assessed in order to identify mutated versions with abrogated or very low binding. A decrease in affinity combined with significantly lower normalised binding level can indicate a disrupted folding due to introduced mutations. It should however be noted that changes in kinetics will also affect this value. Each binding curve was therefore visually inspected for conclusion (data not shown). Results for human TREM-1 with alanine single mutations (Table 10) show that two positions (T46 and D92) were unique in that the decreased binding more than two-fold to 0170 Fab. TABLE 10Affinity of 0170 Fab and 0011 Fab to human TREM-1with alanine mutations relative to buman TREM-1 wt. Bindinglevel of 0170 Fab and 0011 Fab 10 s after end of association,expressed as degree of theoretical maximum binding level.Normalised bindingKD relative to 0247(% Rmax theor)ConstructMutation0170 Fab0011 Fab0170 Fab0011 Fab0247—1144410221D38A0.715.02950222K40A0.9No binding4310223D42A132.926.91330224T44A1.21.846400225E46A2.70.944440226K47A1.01.140380227S50A0.70.945410228R84A0.51.31580229Y90A0.52.940310230H91A0.92.541330231D92A9.21.134410232H93A36.45.222290233R97A1.16.236220234R99A0.81.015110235D127A1.10.843410236R128A0.91.239350237R130A1.00.83534 mAb-0170 was, in contrast to mAb-0011, able to bind cynomolgus TREM-1. Humanisation of cynomolgus TREM-1 in the selected area did not result in regained affinity of 0177 compared to human TREM-1 (0247) indicating that other residues or combinations of residues are important for the differences in affinity for human and cynomolgus TREM-1.0243 shows no binding to 0170 Fab or 0011 Fab. For this construct it cannot be excluded that mutations have affected the overall structure and therefore not be concluded if Q52 is involved in binding of the studied Fabs to human TREM-1. TABLE 11Affinity of 0170 Fab and 0011 Fab to wt cynomolgusTREM-1 ECD, partly humanised cynomolgus TREM-1ECD variants and wt human TREM-1 ECD. Binding levelof 0170 Fab and 0011 Fab 10 s after end of association,expressed as degree of theoretical maximum binding level.Normalised bindingKD (M)(% Rmax theor)Construct0170 Fab0011 Fab0170 Fab0011 Fabwt cTREM-11E−09No binding39002392E−09>1 uM19102402E−09>1 uM28002412E−09>1 uM26202421E−09>1 uM2110243No bindingNo binding5102441E−09No binding7002451E−09No binding34002472E−101E−094542 Example 14: mAb 0170 Efficiently Blocks TREM-1 Activation in the BWZ/hTREM-1 Reporter Cell Assay The BWZ/hTREM-1 reporter cell assay described in Example 6 was used to calculate the potency of mAb 0170 in blocking TREM-1. BWZ/hTREM-1 reporter cells were stimulated with TREM-1 ligand complex and mAb 0170 added at various concentrations.FIG.9shows a dose-dependent blocking of the TREM-1 signal resulting in total block of the signal at concentrations higher than 0.2 ug/ml. The IC50 value was determined to be 2.4 nM using the Graphpad Prism software, equation: log(inhibitor) vs. response—Variable slope. Example 15: Commercially Available TREM-1 Antibodies do not Block TREM-1 Activation in the BWZ/hTREM-1 Reporter Cell Assay Multiple doses of antibody were included in the BWZ/hTREM-1 reporter cell assay, as described previously. SAB1405121 (clone 3F5 from Sigma Aldrich, St. Louis, Mo., USA), WH0054210M4 (clone 2E2 from Sigma Aldrich, St. Louis, Mo., USA), sc-80394 (clone 98Z12 from Santa Cruz, Calif., USA), HM2252 (clone 611 from Hycult biotech, 5405 PB UDEN, The Netherlands). 316102 (clone TREM-37 from Biolegend, San Diego, Calif. 92121, USA), and 314907 (clone TREM-26 from Biolegend, San Diego, Calif. 92121, USA), were not able to block the TREM-1 activity significantly, whereas mAb 0170 discloses herein could block TREM-1 activity with more than 99% at 0.3 ug/ml. Isotype controls had >95% remaining reactivity even at 3 ug/ml. TABLE 12%%%%remainingremainingremainingremainingAnti TREM-1activity atactivity atactivity atactivity atclone0 μg/ml0.003 μg/ml0.03 μg/ml0.3 μg/mlSAB140512110010410197WHO054210M41009810486sc-80394100120134131HM2252100961221133161021009810390(0.09% azide)314907100100124108mAb 017010096800 Example 16: mAb 0170 Blocked Cynomolgus TREM-1 In order to test for functionality against TREM-1 from other species, mouse and cynomolgus monkey TREM-1 was transfected together with human and mouse DAP12, respectively, to generate a reporter cell assay as the one for human. This was essentially done as described for the human system (Examples 1, 2 and 6) but replacing humanTREM-1 with full length murine (m)TREM-1 (SEQ ID NO: 22) or full length cynomolgus monkey (c) TREM-1 (SEQ ID NO: 21). cDNA encoding cTREM-1 (SEQ ID NO: 22) was synthesized at GeneArt and cloned into pHLEF38 (licensed from CMC ICOS), in the XhoI-XbaI orientation.TE alpha NFAT Luc cells was co-transfected with pHLEF38.cynoTrem1 and pNEF38 NFlag hDAP12 using 10 ug of each plasmid and electroporated 8e6 cells in approx. 500 ul total volume (400 ul growth medium and 100 ul DNA) using the BTX electoporator (260 V, 1050 uF, 720 ohms; time constant was 23 msec). Cells were plated for 48 hours in a 10 cm plate, in 10 ml of 50% conditioned medium and plated directly into selection at 8e3 cells/well of a flat bottomed 96W plates (5 plates) in 200 ul/well of 30% conditioned medium with 800 ug/ml G418 and 0.5 mM L-histidinol. After 2 weeks of incubation, 40 single colonies were identified using the Genetix Clone Select Imager. The only commercially available TREM-1 antibody able to cross react with cynomolgus monkey TREM-1 was 314907 (clone TREM-26 from Biolegend, San Diego, Calif. 92121, USA)(see Example 11). None of the commercially available antibodies tested in Example 15 were able to block the function of cynoTREM-1, not even the one that could bind to cynomolgus monkey TREM-1. TABLE 13% remaining cyno TREM-1activity [mAb amount]mAb 01703149070 ug/ml100.00100.000.74 ug/ml7.97103.9720 ug/ml087.72 Likewise, a reporter cell line with mouse TREM-1 was generated. None of the antibodies able to bind to human TREM-1 or to cynomolgus and human TREM-1 could cross-bind to mouseTREM-1. Thus, antibodies against mouse TREM-1 were generated essentially as described for generating human TREM-1 antibodies but with mouse TREM-1 as the antigen. These antibodies were screened for mouse TREM-1 binding and blocking function in the murine reporter gene assay. One such antibody (mAb 0174) was able to bind and block the mouse TREM-1 function. Example 17: TNFalpha Release from M2 Macrophages that were Stimulated by PGLYRP-1 was Blocked by TREM-1 Antibodies Those skilled in the art will recognize the value of establishing a freezer bank collection of primary cells from multiple donors thus providing for convenient replication of experiments. In vitro derived macrophages were produced from peripheral blood monocytes as follows. Negatively enriched monocytes were isolated from a peripheral blood “leukopak” obtained from Research Blood Components (Brighton, Mass., USA) using a Rosette Sep kit (cat. no. 15068) from Stem Cell Technologies (Vancouver, BC, Canada) following the manufacture instructions. Isolated monocytes were suspended in 10% DMSO/FBS aliquots of 50e6 cell/ml and gradually cooled to −80 C. To produce macrophage cells, one or more frozen vials of monocytes were rapidly thawed in a 37 C water bath, diluted to 10 ml with growth media [RPMI 1640 (Gibco, Carlsbad Calif., USA) cat. no. 72400-047) with 10% FBS (Fisher Scientific cat no 03-600-511] and centrifuged 5 minutes at 250 g. Cells were suspended to 2e6 cells/ml in growth media supplemented with 50 ng/ml human MCSF (Gibco cat. no. PHC9501), placed into tissue culture treated, petri style tissue culture plates and into a humidified incubator programmed to maintain a “hypoxic” atmosphere of 5% CO2, 2% O2. On the third day in culture, the cells were fed with the addition of an equal volume of growth media supplemented with 50 ng/ml human MCSF. After 6 days in culture the monocytes had differentiated into M0 macrophages. M0 cells were further differentiated by changing the media to growth media supplemented with 50 ng/ml human IFNg (Gibco cat no PHC4031) for M1 macrophages or 40 ng/ml human IL-4 (Gibco cat no PHC0045) for M2 macrophages and returning to the incubator for an additional 22 hours. On the seventh day, macrophages were suitably differentiated to be used in a bioassay. Briefly, macrophages were recovered from the petri plates by washing with 1×PBS, followed by 5 mM EDTA in PBS. The plates were then returned to 37 C for 30 minutes and cells were “power washed” off the plate using a 10 ml syringe and 22G needle. Cells were then diluted into growth media, centrifuged at 250 g for 5 minutes after which the cell pellet was suspended to a final concentration of 1e6/ml. Macrophage cells prepared as above were used in bioassays where cytokines such as TNF-alpha produced in response to stimulation of the cells with TREM-1 ligand were measured in the conditioned media by ELISA. Such a bioassay was further utilized to measure blockade of such TREM-1 ligand stimulation by TREM-1 specific antibodies. TREM ligand or negative controls were prepared at 4× concentrations and 50 microliters/well in growth media were added to 96 well microtiter dishes. Final concentrations of TREM-1 ligand consisted of 7.5 ng/ml recombinant human PGLYRP1 (see Example 5) and 3 μg/ml PGN-BS (Invivogen, tlrl-pgnbs, SanDiego Calif., USA). Cells were cultured under humidified hypoxic conditions as described above for 22 hours after which conditioned media was collected and TNF-alpha levels were measured by ELISA, following manufacturer's instructions (R&D Systems, catalogue DY210, MN, USA).FIG.10shows that the TREM-1 antibodies decrease TNFalpha release from these stimulated M2 macrophages. Table 12, below, shows the IC50 values from such experiment indicating that the antibodies disclosed herein are very potent in blocking the TREM-1 dependent cytokine release. TABLE 14Antibody (mAb)R squareIC50, ng/ml01220.991401700.991200900.992101150.8892 Example 18: TNFalpha Release from Cynomolgus Macrophages can be Blocked by mAb 0170 Peripheral blood derived macrophage serve as an excellent in vitro model in the study of innate immune modulation and activation. TREM-1 is known to play a key role in controlling this process. The use of the non-human primate speciesMacaca fascicularis, commonly known as Cynomolgus monkey is critical to understanding the in vivo effects of modulating TREM1 signalling. In this example anti-TREM-1 antibodies are tested for their ability to block production of cytokines in cynomolgus M2 macrophage cultures. Macrophage cells were generated as follows. Whole blood was harvested from healthy male adult animals (SNBL, Everett Wash., USA) by venipuncture using sodium heparin vacutainer tubes (Cat No 3664870, Bectin Dickinson Franklin Lakes N.J., USA). Whole blood was diluted 30% with PBS, then 30 ml was carefully layered on to 15 ml of Ficoll-Paque (Cat No 17-1440-03 GE Healthcare, Uppsala Sweden) prediluted to 96% with PBS in a 50 ml conical tube. After centrifugation: 30 min, room temperature, 400 g with low acceleration and no brake; peripheral blood mononuclear cells (PBMC) were harvested from the Ficoll/plasma interphase, diluted 3× with PBS and centrifuged 7 minutes, room temperature, 200 g. Supernatant containing contaminating platelets was aspirated and discarded and the cell pellet was resuspended in 30 ml of PBS+0.2% FBS (fetal bovine serum). This cell wash process was repeated 2 additional cycles after which the PBMC cell pellet was resuspended in RPMI culture media (Cat No. 61870-036, Life Technologies, Grand Island N.Y., USA) plus 10% FBS to 2E6 cells/ml and dispensed onto 15 cm petri dishes (Cat No 430599 Corning, Tewksbury Mass., USA) at 20 ml/dish. Monocytes were allowed to adhere to plastic by incubating overnight at 37 C, 5% CO2, 100% humidity after which non adherent cells were removed by gently swirling and rocking the plates for 20 seconds followed by aspiration. 20 ml of fresh growth media supplemented with 50 ng/ml hMCSF (Cat No. PHC9501, Life Technologies, Grand Island N.Y., USA) was added to each plate and placed into hypoxic culture conditions of 37 C, 5% CO2, 2% O2, 100% humidity for 7 days. On the third day in culture, the cells were fed with the addition of an equal volume of growth media supplemented with 50 ng/ml human MCSF. After 6 days in culture the monocytes had differentiated into M0 macrophages. M0 cells were further differentiated by changing the media to growth media supplemented with 50 ng/ml human IFNg (Gibco cat no PHC4031) for M1 macrophages or 40 ng/ml human IL-4 (Cat No. PHC0045, Life Technologies, Grand Island N.Y., USA) for M2 macrophages and returning to the incubator for an additional 22 hours. On the seventh day, macrophages were suitably differentiated to be used in a bioassay. Briefly, macrophages were recovered from the petri plates by washing with 1×PBS, followed by 5 mM EDTA in PBS. The plates were then returned to 37 C for 30 minutes and cells were “power washed” off the plate using a 10 ml syringe and 22G needle. Cells were then diluted into growth media, centrifuged at 250 g for 5 minutes after which the cell pellet was suspended to a final concentration of 1e6/ml. Macrophage cells prepared as above were used in bioassays where cytokines such as TNF-alpha produced in response to stimulation of the cells with TREM-1 ligand were measured in the conditioned media by ELISA. Such a bioassay was further utilized to measure blockade of such TREM-1 ligand stimulation by TREM-1 specific antibodies. TREM-1 ligand or negative controls were prepared at 4× concentrations and 50 microliters/well in growth media were added to 96 well microtiter dishes. Final concentrations of TREM-1 ligand consisted of 7.5 ng/ml recombinant human PGLYRP1 (generated as described in Example 5) and 3 μg/ml PGN-BS (Cat No. tlrl-pgnbs, Invivogen SanDiego Calif., USA). Antibodies were added in 50 microliters/well of growth media followed by cells in 50 microliters/well of macrophage. Cells were cultured under humidified hypoxic conditions as described above for 22 hours after which conditioned media was collected and TNF-alpha levels were measured by EISA, following manufacturer's instructions (Cat No. DY1070 R&D Systems, Minneapolis Minn., USA). The table following table shows TNFα values of M2 macrophage cultures stimulated with TREM-1 ligand (PGLYRP1+PGN) in the presence of control antibody or anti-TREM-1 antibody. M2Donor 1Donor 2macrophagesTNF-α, pg/mlTNF-α, pg/mlwith:AvgSDAvgSDCells only0.519.012.0PGRP-S only3.513.06.3PGN BS only5.72.822.43.8PGN + PGRP-S295.349.3307.475.8+Fc4 neg −3248.554.0285.057.6+Fc4 neg −1215.641.5295.059.0+Fc4 neg −.33217.549.7357.7118.6+Fc4 neg −.11193.940.4348.475.7+Fe4 neg −.037172.657.6370.788.6+Fc4 neg −.012177.638.1286.771.5+Fc4 neg −.0041275.467.9253.345.0+0170 −333.38.857.23.2+0170 −141.38.547.010.5+0170 −.3345.314.380.421.6+00170 −.1142.212.4103.832.8+0170 −.03754.418.5142.343.5+0170 −.01286.224.4203.855.6+0170 −.0041186.721.0249.392.5+0170 −.0014231.256.0286.1112.8+0170 −.00046246.422.7254.270.5 This example illustrates that the anti TREM-1 Ab-0170 can effectively block TREM dependent cytokine production in macrophage cells derived from cynomolgus monkeys. The efficacy of this Ab supports its use in cynomolgus monkey in in vivo toxicology and disease treatment models. Example 19: TNFalpha Release from Stimulated Peripheral Blood Mononuclear Cells can be Blocked by TREM-1 Antibodies PBMC's, from a buffy coat and frozen in RPMI 1640 (Cat. no. 61870, Gibco, New York, USA), 20% FBS (Cat #16140-071, Gibco, New York, USA, 10% DMSO (Cat #D2650, Sigma, Steinheim, Germany), were thawed and washed twice in RPMI, 10% FBS, 1% Pen/Strep (Cat. no. 15070-06, Gibco, New York, USA), and resuspended in same medium to 4×10E6/ml. Cells were then distributed with 400.000 cells/well. 10 μg/ml PGN-SA (Cat #tlrl-pgnsa, Invivogen, San Diego, USA) and 0.2 μg/ml PGLYRP1 were added to the wells for stimulating the cells. Subsequently, the relevant isotype and TREM-1 antibodies were diluted in RPMI and added at 1.34 nM, and 0.167 nM respectively. TNFalpha release were measured by diaplex (Cat #880.090.001, Genprobe, Besancon, France) according to manufacturers protocol after 20 hrs incubation at 37° C., 5% CO2. As shown in Table 13, below, the TREM-1 antibodies disclosed herein (mAbs 0044, 0070 and 0059) are all able to decrease the TNFalpha release from PBMC cells. TABLE 15% Inhibition comparedIsotype atto isotype1.34 nM0 nM0.167 nMa-Trem-1 0044 (mIgG2a)10010068a-Trem-1 0070 ((hIgG4)10011776a-Trem-1 0059 (hzIgG1.1)10010561 The approximately 70% inhibition of TNFalpha release in PBMCs using a blocking TREM-1 antibody indicates a significant impact on cytokine levels in a stimulated cell culture. Example 20: Anti-TREM-1 mAb can Inhibit PGN+PGLYRP1-Induced TNFα Production in Normoxic Macrophages Stimulation of macrophages using PGN-BS+human PGLYRP1 as a stimulant of the TREM-1 receptor can be blocked by anti-TREM-1 antibodies. Monocytes were differentiated into M2 macrophages as in Example 15. All steps of the differentiation and stimulation of the cells were done in a 37° C., 5% CO2 incubator under normal atmospheric oxygen levels (normoxia). The differentiated M2 macrophages were resuspended in RPMI/10% FBS and plated out at 5×10E5 cells/ml in triplicate (unless otherwise noted) test wells. The cells were then stimulated for 24 hours with the following stimulations: no addition. PGLYRP1, PGN-BS (InVivogen, tlrl-pgnbs)(two sets of triplicates), PGN-BS+PGLYRP1 (three sets of triplicates), or PGN-BS+PGLYRP1 in the presence of anti-TREM-1 or isotype control antibody. Supernatants were then harvested and analysed for TNFα using BioPlex (Bio-Rad, 171-85026M). Antibodies (0.1 μg/mi) mAb-0122 and -0170 directed against TREM-1 were able to lower the TNF-alpha release. TABLE 16Donor 1Donor 2MacrophagesTNFa pg/mlTNFa pg/mlstimulated with:AvgSDAvgSDNo addition0000PGLYRP10000PGN-BS4278638185PGN-BS + PGLYRP1728293771220PGN-BS + PGLYRP1 +9871838844hIgG4 isotype controlPGN-BS + PGLYRP1 +2431436716mAb 0122PGN-BS + PGLYRP1 +12210622454mAb 0170 This example illustrates that anti-TREM-1 mAb-0122 and -0170 can inhibit TNFa production from macrophages grown under normoxic conditions. Example 21: TREM-1 Antibody Specifically Inhibits Rheumatoid Arthritis Synovial Fluid-Induced Response The RA synovial fluid samples from patients suffering from rheumatoid arthritis were assayed for TREM-1 ligand activity in the BWZ reporter assay as described in Example 6. Briefly, synovial fluid was thawed, vortexed, and serially diluted, assayed in duplicate +/− 10 μg/ml PGNECndi (Invivogen SanDiego, Calif., USA) with the addition of TREM-1 antibodies or a negative isotype control. The synovial fluid from a rheumatoid arthritis patient is able to trigger the BWZ/hTREM-1 reporter cell assay in a TREM-1 dependent manner which is further enhanced by adding MAB1278 (R&D Systems, Minneapolis, Minn. 55413, USA: Cat. no. MAB1278)) whereas the blocking TREM-1 antibodies disclosed herein are able to decrease this activation. The antibodies were tested in two assays indicated by the two columns below, each antibody in concentrations ranging from 0.1 to 10 ug/ml. MAB1278 clearly enhanced the signal, whereas mAbs 0122 and 0170 decreased the signal compared to the isotype control. TABLE 17+IgG4 isotypeConcentration+mAb 0122+mAb 0170+mAb 1278control10ug/ml80877797315206450304107900010540003069063975561ug/ml74191664584825446978119200010230003984313632670.1ug/ml133900163521246695169483828866691379293831313445 Example 22: Cytokine Release from Synovial Tissue Cells from Rheumatoid Arthritis Patients Upon Stimulation with PGLYRP-1 can be Blocked by mAb 0170 Synovial tissue samples were obtained from RA patients during total knee replacement. Single suspension of synovial tissue cells was isolated by a digestion via 4 mg/ml of collagenase (cat #11088793001, Roche, Mannheim, Germany) and 0.1 mg/ml of DNase (cat #11284932001, Roche, Mannheim, Germany) for 1 h at 37 degrees C. Synovial tissue cells at 1×10{circumflex over ( )}5/well in culture medium RPMI (cat #R0883, St Louis, Mo., USA)+10% FCS (cat #S0115, BioChrom AG, Grand Island, N.Y. 14072, USA) were stimulated with 4 ug/ml of PGLYRP1 and 1 ug/ml of PGN-ECNDi (cat #tlrl-kipgn, Invivogen, San Diego, Calif. 92121, USA) under hypoxic condition in the presence or absence of various concentrations of mAb 0170 or an isotype hIgG4 control. After 24 h incubation, cell supernatants were harvested, and cytokines were measured by either ELISA (TNFa (cat #DY210, R&D, Minneapolis, Minn. 55413 USA), IL-1b (88-7010-88, eBioscience, San Diego, Calif. 92121, USA), GM-CSF (cat #88-7339-88, eBioscience)) or Flowcytomix (TNFa, IL-1b, MIP-1b, MCP-1, IL-6, and IL-8 (cat #BMS, eBioscience). The cytokines were secreted from the synovial tissue cells upon stimulation with the TREM-1 ligand and specifically blocked by TREM-1 antibody mAb 0170. Below is an example of such experiment, where either 4 ng/ml or 10 ng/ml mAb was used resulting in a decrease of cytokine release when treated with TREM-1 antibody mAb 0170. TABLE 18PGN +PGN +PGLYRP1 +PGLYRP1 +CytokinePGN +10 ng/ml10 ng/ml(pg/ml)PGNPGLYRP1controlmAb 0170TNFalpha62414451034429MIP-24584395379123211betaMCP-1273471391210 TABLE 19PGN +PGN +CytokinePGN +PGLYRPH +PGLYRP1 +(pg/ml)PGNPGLYRP14 ng/ml control4 ng/ml 0170IL-1beta2419377334772308GM-CSF182616656431IL-62057418934751632IL-82575550944992112 This example shows that cells from synovial tissue from rheumatoid arthritis patients will respond to stimulation by the TREM-1 ligand, PGLYRP1, by secreting numerous cytokines which can be inhibited by mAb 0170. Example 23: Type II PGLYRP1 Induced TNFalpha Release in Synovial Tissue Cells from Rheumatoid Arthritis Patients can be Blocked by TREM-1 Antibody mAb 0170 Synovial tissue samples were obtained from RA patients during total knee replacement. Single suspension of synovial tissue cells was isolated by a digestion via 4 mg/ml of collagenase (cat. no. 11088793001, Roche, Mannheim, Germany) and 0.1 mg/m of DNase (cat. no. 11284932001 Roche, Mannheim, Germany) for 1 h at 37 degree. The synovial tissue cells (1×10{circumflex over ( )}5/well in culture medium RPMI (cat. no. 22400105, Gibco, NY 14072, USA)+10% FCS (cat. no. S0115, BioChrom AG, Berlin, Germany)) were co-cultured with various doses of HEK cells transiently transfected with type II PGLYRP1 under hypoxic condition in the presence or absence of 1 ug/ml of mAb 0170 or IgG4 isotype control. After 24 h incubation, cell supernatants were harvested, and cytokines were measured by TNFa ELISA (cat. no. DY210, R&D, Minneapolis, Minn. 55413 USA). TABLE 20TNF-a (pg/ml) releaseType II PGLYRP11 × 10{circumflex over ( )}53 × 10{circumflex over ( )}41 × 10{circumflex over ( )}43 × 10{circumflex over ( )}31 × 10{circumflex over ( )}30(HEK transfected)/Control HEKIgG4+ Type II121.17114.0895.0254.5657.8733.47cellsIgG4+ Control55.6563.7357.9933.7836.4036.32cellsmAb 0170+44.0544.6744.4039.4544.2931.40Type II cellsmAb 0170+54.5057.06$3.1042.1031.9927.82Control cells This example shows that the TREM-1 ligand (type II cells) induced TNF-alpha release in a dose-dependent manner in synovial tissue cells from rheumatoid arthritis patients compared to control cells (mock transfected). This TNFalpha response was blocked by mAb 0170 but not with isotype IgG4. The control cells were not affected. Example 24: Platebound MAB1278 Induced IL-6 and TNFalpha Response in Macrophages, Showing Agonistic Features, Whereas mAbs 0122 and 0170 Did not Stimulation of macrophages on platebound agonistic anti-TREM-1 antibodies induced production of IL-6 and TNFa. Monocytes were purified from healthy donor buffy coats using RosetteSep (StemCell Technologies, 15068) and differentiated into macrophages by culturing for 6 days in RPMI/10% FBS in the presence of 40 ng/ml human MCSF. The macrophages were then further differentiated to M2 macrophages by changing the media to growth media supplemented with 50 ng/ml human IL-4 and returning to the incubator for an additional 24 hours. On the seventh day, macrophages were recovered from the culture plates by washing with 1×PBS, followed by 5 mM EDTA in PBS. The plates were then returned to 37° C. for 30 minutes before the macrophages were washed off the plates. The macrophages were washed in RPM/10% FBS before resuspending and plating out. The test wells had been pre-coated with the specified antibodies by incubating them overnight with antibody diluted in PBS, followed by washing ×3 in PBS. The resuspended macrophages were plated out at 5×10E5 cells/ml in triplicate test wells followed by incubation for 24 hours. (All steps of the differentiation and stimulation of the cells were done in a 37° C., 5% CO2 incubator under normal atmospheric oxygen levels (normoxia)). Supernatants were then harvested and analysed for IL-6 and TNFa using BioPlex (Bio-Rad, 171-B5006M and 171-B5026M). Antibodies mAb-0122 and -0170 showed very low agonism whereas the MAB1278 antibody (RnD Systems, MAB1278) showed potent induction of both IL-6 and TNFa. TABLE 21PlateboundIL-6 pg/mlTNFa pg/mlmAb stimulation:AvgSDAvgSDNo antibody130.30.8mIgG1 isotype cntr00002 μg/mlmIgG1 isotype cntr00006 μg/mlmIgG1 isotype cntr14245520 μg/mlMAB1278 2 μg/ml412712004451MAB1278 6 μg/ml877386454278MAB1278 20 μg/ml1352767753555hIgG4 isotype cntr55212 μg/mlhIgG4 isotype cntr1815996 μg/mlhIgG4 isotype cntr37334120 μg/mlmAb 0122 2 μg/ml93485918mAb 0122 6 μg/ml126810517mAb 0122 20 μg/ml2341030426mAb 0170 2 μg/ml137297621mAb 0170 6 μg/ml1811218728mAb 0170 20 μg/ml27715629108 This example illustrates that mAb-0122 and -0170 only show very low agonistic activity in macrophages and indicates true blocking features of these mAbs. Example 25: Blocking TREM-1 in a Mouse Arthritis Model Reduces Disease The experiments outlined in Table 22 were obtained in the DTH-arthritis model, which is a single paw arthritis model. Single paw arthritis was induced in female C57BL/6 mice by eliciting a classical delayed-type hypersensitivity (DTH) reaction in the right hind paw by immunisation and subsequent challenge with methylated bovine serum albumin (mBSA), with the modification that a cocktail of type II collagen monoclonal antibodies (anti-C) was administered IV between the immunisation and challenge steps. The left hind paw received PBS challenge and functioned as an intra-animal control. Mice (10 mice/group) were treated 3 times/week with a TREM monoclonal antibody that specifically binds and blocks murine TREM-1, as determined using a murine version of the reporter assay described in Example 6. The first dose was administered on the day of immunization. Mice (9-10 mice/group) were treated with either a control antibody or PBS as a control. Paw swelling was measured from the day of arthritis induction and 11 days onwards. Results are presented as a mean area under the curve (AUC)±SEM. Statistical significance was tested by using a two sided unpaired t-test, 95% confidence interval. TABLE 22Effect of TREM-1 treatmenton paw swelling (AUC-mm) in a mouse arthritis model.EffectTREM-1ControlmeasureExperimentmAb§mAb§PBSAUC-mm#18.03 +/−12.26 +/−12.53 +/−paw swelling1.01**0.840.87AUC-mm#28.45 +/−11.71 +/−12.89 +/−paw swelling0.94**0.220.36Means +/− SEM.§mice treated with 5 mg/kg, 3 times week for 3 weeks**P ≤ 0.005, two sided unpaired t-test, 95% confidence interval vs. control mAb and vs. PBS Example 26: Activated Neutrophils Release IL-8 which can be Blocked by TREM-1 mAbs Neutrophils express TREM-1 and neutrophils also express the TREM-1 ligand. To test whether TREM-1 is involved in an autocrine stimulation loop in neutrophils, isolated neutrophils were stimulated with PGN-SA (InVivogen, tlrl-pgnsa), and the release of IL-8 into the culture medium was measured. TREM-1 antibodies mAb-0059, -0067, -0122, and -0170 were able to decrease the PGN-SA-induced IL-8 release. Neutrophils were isolated from human healthy donor whole blood and resuspended in RPMI/10% FBS at. 5×10E6 cells/ml, and plated out into triplicate test wells pre-coated with Fibrinogen (pre-coated with 50□l of 1 mg/ml Fibrinogen (Sigma, F3879) in PBS for 2 hours at 37° C., followed by washing ×3 in PBS). The cells were tested under the following conditions: no added stimulation, 10□g/ml PGN-SA only, or 10 □g/ml PGN-SA in the presence of mAb-0059, -0067, -0122, -0170 or isotype control antibody at 0.25□g/ml. The samples were cultured 24 hours in a 37° C., 5% CO2incubator. Supernatants were then harvested and analysed for IL-8 using the Bio-plex Pro Human Cytokine IL-8 set (BioRad, 171-B5008M). TABLE 23Experiment 1Experiment 2NeutrophilsIL-8, pg/mlNeutrophilsIL-8, pg/mlstimulated with:AvgSDstimulated with:AvgSDNo additionND.N.D.No addition857214PGN-SA56254PGN-SA6116191PGN-SA + hIgG1.146180PGN-SA + hIgG465301962isotype contr.isotype contr.PGN-SA + mAb 005916555PGN-SA + mAb 01222466437PGN-SA + mAb 006718377PGN-SA + mAb 01702171480 This example illustrates that IL-8 release from neutrophils induced by stimulation with the bacterially derived PGN can be reduced by TREM-1 antibodies. Thus demonstrating that TREM-1 is involved in an autrocrine activation loop in neutrophils, and the TREM-1 antibodies are potentially useful in downregulating neutrophil responses. Example 27: Activated Neutrophils can Stimulate Monocytes, which can be Blocked by Anti-TREM-1 mAbs Activated neutrophils express the TREM-1 ligand. To test if activated neutrophils can stimulate other immune cells in a TREM-1-dependent manner, activated neutrophils were used to stimulate isolated monocytes and the release of TNFα into the culture medium was measured. TREM-1 antibodies mAb-0059 and -0170 were able to decrease the neutrophil-induced TNFα release from the monocytes. Neutrophils were isolated from human healthy donor whole blood and resuspended in RPMI/10% FBS, and plated at 1.5×10E5 cells/well in poly-D-Lysine coated tissue culture 96-well plates (Corning, 3841). The neutrophils were then stimulated with 1 ng/ml PMA (Sigma, P1585)+20□g/ml PGN-SA (InVivogen, tlrl-pgnsa) for 24 hours in a 37° C., 5% CO2 incubator. The cells were then washed gently ×3 with media before adding in freshly isolated monocytes. The monocytes were purified from healthy donor buffy coats using an EasySep kit (Stem cell technologies, 19059), and were plated out with 5×10E4 cells/well in the wells already containing activated, washed neutrophils. The following antibodies were added at 1 g/ml: mAb-0059, mAb-0170, or hIgG4 isotype control. The cells were then cultured for another 24 hours before harvesting the supernatant. The supernatant was diluted 1:10 in RPMI/10% FNS before measuring TNFa by ELISA (eBioscience, BMS223INST). TABLE 24TNFa pg/mlMonocytes stimulated with:AvgSDNo neutrophils3485Activated neutrophils29967Activated neutrophils + isotype control23232Activated neutrophils + mAb 00597214Activated neutrophils + mAb 01701299 This example illustrated that activated neutrophils can stimulate monocytes in a TREM-1-dependent manner to produce TNFa, and this can be blocked by mAb-0059 and mAb-0170 anti-TREM-1 antibodies. Anti-TREM-1 antibodies are therefore potentially useful for downregulating monocyte responses. Example 28: Epitope Mapping by Protein Crystallography Materials The IgV-like domain of human TREM-1 (SEQ ID NO: 24) in a buffer consisting of 20 mM 2-(N-morpholino)ethanesulfonic acid, 150 mM NaCl, 5% (v/v) glycerol, pH 5.5 at a protein concentration of 11.7 mg/mL The Fab region of mAb 0170 (SEQ ID NO: 25 and SEQ ID NO: 26) in a buffer consisting of 10 mM phosphate, 2.68 mM KCl, 140 mM NaCl, pH 7.4 at a protein concentration of 8.5 mg/mL. Methods: Protein Complex Formation and Crystallization The IgV-like domain of human TREM-1 was mixed with the Fab region of mAb 0170 in their original buffers in a 1:1 molar ratio, giving a final concentration of Fab of 7.1 mg/mL. The proteins were co-crystallized in a hanging drop vapour diffusion experiment by equilibration of droplets consisting of 2 μL protein solution mixed with 2 μL reservoir solution against a 0.5 mL reservoir composed of 0.01 M MgCl2, 0.005 M NiCl2, 0.1 M 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid, 13 (w/v) % polyethylene glycol 3350, pH 7.0. The crystals appeared as clusters of crystals, which were separated into single crystals and transferred to a drop consisting of 35 (w/v) % polyethylene glycol 3350, 0.01 M MgCl2, 0.005 M NiCl2and left to equilibrate in the drop for 15 seconds. The crystal was mounted in a 0.2 mm diameter litholoop (Molecular Dimensions Limited) and flash-cooled in liquid nitrogen. X-Ray Diffraction Data Collection, Structure Determination and Refinement Diffraction data were collected at a micromax-007HF Cu X-ray generator (Rigaku Europe) operated at 40 kV, 30 mAmp and equipped with Varimax HF optics (Cu K□, =0.15418 nm)(Rigaku Europe), a Cryonix165 CCD (Rigaku Europe) and a Cryo-stream (Rigaku Europe) operated at 100 K. The raw data images were indexed, integrated and scaled using the XDS program package (Kabsch, Acta Crystallogr. D66, 133-144 (2010)). The space group of the crystal was P2(1), with unit cell parameters, a=62.3 Å, b=64.9 Å, c=74.4 Å, α=90°, β=75.2°, γ=90°. Data were collected to a resolution of 1.99 Å. The structure was solved by molecular replacement using the Phenix software (Adams et al., Acta Crystallogr. D66, 213-221 (2010)) as implemented in the CCP4i program suite (Potterton et al., Acta Crystallogr. D59, 1131-1137 (2003)). The search models were the structure of the mAb 0170 Fab fragment and the IgV-like domain from human TREM-1 (Kelker et al., J. Mol. Biol. 342, 1237-1248 (2004)). One copy of each molecule was located in the asymmetric unit. Structure refinement was carried out using Refmac5 (Murshudov er al., Acta Crystallogr. D53, 240-255 (1997)) from the CCP4i program suite. Coot version 7 (Emsley et al., Acta Crystallogr. D66, 486-501 (2010)) was used for manual structure rebuilding and validation. Results and Discussion The crystal structure of the complex between the Fab fragment of mAb 0170 and the IgV-like domain of human TREM-1 included E1-V219 (SEQ ID NO: 25), D1-C218 (SEQ ID NO: 26) and L24-T134 of SEQ ID NO: 24. The quality parameters of the structure were good with overall R-factor of the structure=21.1% and the Free R-factor=; 27.1%. The overall correlation coefficient was 0.93 and the diffraction-component precision index, DPI=0.2 Å (Cruickshank, Acta Crystallogr. D55, 583-601 (1999)). The root-mean-square deviation of the bond lengths in the structure from ideal bond lengths=0.0164 Å and the root-mean-square deviation from ideal bond angles=1.8661° (Engh and Huber, Acta Crystallogr. A47, 392-400 (1991)). Regions displaying intermolecular distances of less than or equal to 4 Å between the IgV-like domain of human TREM-1 and the Fab fragment of mAb 0170 were assigned to the epitope region of human TREM-1 (SEQ ID NO: 1). The analysis of intermolecular distances was carried out using the program NCONT in the CCP4 program suite (Potterton et al., Acta Crystallogr. D59, 1131-1137 (2003)). The analysis showed that human TREM-1 amino acid residues K40, D42, T44-K47 and Y90-L95, R97 defined the epitope. While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now be apparent to those of ordinary skill in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.
163,855
11858993
DEFINITIONS The scope of present invention is defined by the claims appended hereto and is not limited by particular embodiments described herein; those skilled in the art, reading the present disclosure, will be aware of various modifications that may be equivalent to such described embodiments, or otherwise within the scope of the claims. In general, terminology used herein is in accordance with its understood meaning in the art, unless clearly indicated otherwise. Explicit definitions of certain terms are provided below; meanings of these and other terms in particular instances throughout this specification will be clear to those skilled in the art from context. References cited within this specification, or relevant portions thereof, are incorporated herein by reference. In order that the present invention may be more readily understood, certain terms are first defined below. Additional definitions for the following terms and other terms are set forth throughout the specification. “Affinity”: As is known in the art, “affinity” is a measure of the tightness with a particular ligand binds to its partner. Affinities can be measured in different ways. In some embodiments, affinity is measured by a quantitative assay. In some such embodiments, binding partner concentration may be fixed to be in excess of ligand concentration so as to mimic physiological conditions. Alternatively or additionally, in some embodiments, binding partner concentration and/or ligand concentration may be varied. In some such embodiments, affinity may be compared to a reference under comparable conditions (e.g., concentrations). “Affinity matured” (or “affinity matured antibody”), as used herein, refers to an antibody with one or more alterations in one or more CDRs thereof which result an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s). In some embodiments, affinity matured antibodies will have nanomolar or even picomolar affinities for a target antigen. Affinity matured antibodies may be produced by any of a variety of procedures known in the art. Marks et al., 1992, BioTechnology 10:779-783 describes affinity maturation by VHand VLdomain shuffling. Random mutagenesis of CDR and/or framework residues is described by: Barbas et al., 1994, Proc. Nat. Acad. Sci. U.S.A 91:3809-3813; Schier et al., 1995, Gene 169: 147-155; Yelton et al., 1995, J. Immunol. 155: 1994-2004; Jackson et al., 1995, J. Immunol. 154(7):3310-9; and Hawkins et al., 1992, J. Mol. Biol. 226:889-896. “Amelioration”, as used herein, refers to the prevention, reduction or palliation of a state, or improvement of the state of a subject. Amelioration includes, but does not require complete recovery or complete prevention of a disease, disorder or condition (e.g., radiation injury). “Animal”, as used herein refers to any member of the animal kingdom. In some embodiments, “animal” refers to humans, of either sex and at any stage of development. In some embodiments, “animal” refers to non-human animals, at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, and/or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, insects, and/or worms. In certain embodiments, the animal is susceptible to infection by DV. In some embodiments, an animal may be a transgenic animal, genetically engineered animal, and/or a clone. “Antibody”, as used herein, has its art understood meaning and refers to an immunoglobulin (Ig) that binds specifically to a particular antigen. As is known by those of ordinary skill in the art, antibodies produced in nature are typically comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains. Each heavy and light chain is comprised of a variable region (abbreviated herein as HCVR or VHand LCVR or VL, respectively) and a constant region. The constant region of a heavy chain comprises a CH1, CH2 and CH3 domain (and optionally a CH4 domain in the case of IgM and IgE). The constant region of a light chain is comprised of one domain, CL. The VHand VLregions further contain regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, which are termed framework regions (FR). Each VHand VLis composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. Immunoglobulin molecules can be of any type (e.g., IgM, IgD, IgG, IgA and IgE), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass. Antibody agent: As used herein, the term “antibody agent” refers to an agent that specifically binds to a particular antigen. In some embodiments, the term encompasses any polypeptide with immunoglobulin structural elements sufficient to confer specific binding. In various embodiments, suitable antibody agents may include, but are not limited to, monoclonal antibodies, polyclonal antibodies, humanized antibodies, primatized antibodies, chimeric antibodies, human antibodies, bi-specific or multi-specific antibodies, single domain antibodies (e.g., shark single domain antibodies (e.g., IgNAR or fragments thereof)), conjugated antibodies (i.e., antibodies conjugated or fused to other proteins, radiolabels, cytotoxins), Small Modular ImmunoPharmaceuticals (“SMIPs™”), single chain antibodies, cameloid antibodies, antibody fragments, etc. In some embodiments, the term can refer to a stapled peptide. In some embodiments, the term can refer to an antibody-like binding peptidomimetic. In some embodiments, the term can refer to an antibody-like binding scaffold protein. In some embodiments, the term can refer to monobodies or adnectins. In many embodiments, an antibody agent is or comprises a polypeptide whose amino acid sequence includes one or more structural elements recognized by those skilled in the art as a complementarity determining region (CDR); in some embodiments an antibody agent is or comprises a polypeptide whose amino acid sequence includes at least one CDR (e.g., at least one heavy chain CDR and/or at least one light chain CDR) that is substantially identical to one found in a reference antibody. In some embodiments, an included CDR is substantially identical to a reference CDR in that it is either identical in sequence or contains between 1-5 amino acid substitutions as compared with the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that it shows at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity with the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that it shows at least 96%, 96%, 97%, 98%, 99%, or 100% sequence identity with the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that at least one amino acid within the included CDR is deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical with that of the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that 1-5 amino acids within the included CDR are deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical to the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that at least one amino acid within the included CDR is substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical with that of the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that 1-5 amino acids within the included CDR are deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical to the reference CDR. In some embodiments, an antibody agent is or comprises a polypeptide whose amino acid sequence includes structural elements recognized by those skilled in the art as an immunoglobulin variable domain. In some embodiments, an antibody agent is a polypeptide protein having a binding domain, which is homologous or largely homologous to an immunoglobulin-binding domain. In some embodiments, an antibody agent is or comprises a polypeptide that includes all CDRs found in a particular reference antibody chain or chains (e.g., heavy chain and/or light chain). “Antibody component”, as used herein, refers to a polypeptide element (that may be a complete polypeptide, or a portion of a larger polypeptide, such as for example a fusion polypeptide as described herein) that specifically binds to an epitope or antigen and includes one or more immunoglobulin structural features. In general, an antibody component is any polypeptide whose amino acid sequence includes elements characteristic of an antibody-binding region (e.g., an antibody light chain or variable region or one or more complementarity determining regions (“CDRs”) thereof, or an antibody heavy chain or variable region or one more CDRs thereof, optionally in presence of one or more framework regions). In some embodiments, an antibody component is or comprises a full-length antibody. In some embodiments, an antibody component is less than full-length but includes at least one binding site (comprising at least one, and preferably at least two sequences with structure of known antibody “variable regions”). In some embodiments, the term “antibody component” encompasses any protein having a binding domain, which is homologous or largely homologous to an immunoglobulin-binding domain. In particular embodiments, an included “antibody component” encompasses polypeptides having a binding domain that shows at least 99% identity with an immunoglobulin binding domain. In some embodiments, an included “antibody component” is any polypeptide having a binding domain that shows at least 70%, 75%, 80%, 85%, 90%, 95% or 98% identity with an immunoglobulin binding domain, for example a reference immunoglobulin binding domain. An included “antibody component” may have an amino acid sequence identical to that of an antibody (or a portion thereof, e.g., an antigen-binding portion thereof) that is found in a natural source. An antibody component may be monospecific, bi-specific, or multi-specific. An antibody component may include structural elements characteristic of any immunoglobulin class, including any of the human classes: IgG, IgM, IgA, IgD, and IgE. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Such antibody embodiments may also be bispecific, dual-specific, or multi-specific formats specifically binding to two or more different antigens. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VH, VL, CH1 and CLdomains; (ii) a F(ab′)2fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VHand CH1 domains; (iv) a Fv fragment consisting of the VHand VLdomains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which comprises a single variable domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VHand VL, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VHand VLregions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al., 1988, Science 242:423-426; and Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883). In some embodiments, an “antibody component”, as described herein, is or comprises such a single chain antibody. In some embodiments, an “antibody component” is or comprises a diabody. Diabodies are bivalent, bispecific antibodies in which VHand VLdomains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al., 1993, Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., 1994, Structure 2(12):1121-1123). Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5). In some embodiments, an antibody component is or comprises a single chain “linear antibody” comprising a pair of tandem Fv segments (VH-CH1-VH-CH1) which, together with complementary light chain polypeptides, form a pair of antigen binding regions (Zapata et al., 1995, Protein Eng. 8(10): 1057-1062; and U.S. Pat. No. 5,641,870). In some embodiments, an antibody component may have structural elements characteristic of chimeric or humanized antibodies. In general, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary-determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity. In some embodiments, an antibody component may have structural elements characteristic of a human antibody. “Biological activity”, as used herein, refers to an observable biological effect or result achieved by an agent or entity of interest. For example, in some embodiments, a specific binding interaction is a biological activity. In some embodiments, modulation (e.g., induction, enhancement, or inhibition) of a biological pathway or event is a biological activity. In some embodiments, presence or extent of a biological activity is assessed through detection of a direct or indirect product produced by a biological pathway or event of interest. “Bispecific antibody”, as used herein, refers to a bispecific binding agent in which at least one, and typically both, of the binding moieties is or comprises an antibody component. A variety of different bi-specific antibody structures are known in the art. In some embodiments, each binding moiety in a bispecific antibody that is or comprises an antibody component includes VHand/or VLregions; in some such embodiments, the VHand/or VLregions are those found in a particular monoclonal antibody. In some embodiments, where the bispecific antibody contains two antibody component-binding moieties, each includes VHand/or VLregions from different monoclonal antibodies. In some embodiments, where the bispecific antibody contains two antibody component binding moieties, wherein one of the two antibody component binding moieties includes an immunoglobulin molecule having VHand/or VLregions that contain CDRs from a first monoclonal antibody, and one of the two antibody component binding moieties includes an antibody fragment (e.g., Fab, F(ab′), F(ab′)2, Fd, Fv, dAB, scFv, etc.) having VHand/or VLregions that contain CDRs from a second monoclonal antibody. “Bispecific binding agent”, as used herein, refers to a polypeptide agent with two discrete binding moieties, each of which binds with a distinct target. In some embodiments, a bispecific binding agent is or comprises a single polypeptide; in some embodiments, a bispecific binding agent is or comprises a plurality of peptides which, in some such embodiments may be covalently associated with one another, for example by cross-linking. In some embodiments, the two binding moieties of a bispecific binding agent recognize different sites (e.g., epitopes) the same target (e.g., antigen); in some embodiments, they recognize different targets. In some embodiments, a bispecific binding agent is capable of binding simultaneously to two targets that are of different structure. “Carrier”, as used herein, refers to a diluent, adjuvant, excipient, or vehicle with which a composition is administered. In some exemplary embodiments, carriers can include sterile liquids, such as, for example, water and oils, including oils of petroleum, animal, vegetable or synthetic origin, such as, for example, peanut oil, soybean oil, mineral oil, sesame oil and the like. In some embodiments, carriers are or include one or more solid components. “CDR”, as used herein, refers to a complementarity determining region within an antibody variable region. There are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDR1, CDR2 and CDR3, for each of the variable regions. A “set of CDRs” or “CDR set” refers to a group of three or six CDRs that occur in either a single variable region capable of binding the antigen or the CDRs of cognate heavy and light chain variable regions capable of binding the antigen. Certain systems have been established in the art for defining CDR boundaries (e.g., Kabat, Chothia, etc.); those skilled in the art appreciate the differences between and among these systems and are capable of understanding CDR boundaries to the extent required to understand and to practice the claimed invention. “CDR-grafted antibody”, as used herein, refers to an antibody whose amino acid sequence comprises heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VHand/or VLare replaced with CDR sequences of another species, such as antibodies having murine VHand VLregions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences. Likewise, a “CDR-grafted antibody” may also refer to antibodies having human VHand VLregions in which one or more of the human CDRs (e.g., CDR3) has been replaced with mouse CDR sequences. “Chimeric antibody”, as used herein, refers to an antibody whose amino acid sequence includes VHand VLregion sequences that are found in a first species and constant region sequences that are found in a second species, different from the first species. In many embodiments, a chimeric antibody has murine VHand VLregions linked to human constant regions. In some embodiments, an antibody with human VHand VLregions linked to non-human constant regions (e.g., a mouse constant region) is referred to as a “reverse chimeric antibody”. “Combination therapy”: As used herein, the term “combination therapy” refers to those situations in which a subject is simultaneously exposed to two or more therapeutic regimens (e.g., two or more therapeutic agents). In some embodiments, two or more agents or may be administered simultaneously; in some embodiments, such agents may be administered sequentially; in some embodiments, such agents are administered in overlapping dosing regimens. “Comparable”, as used herein, refers to two or more agents, entities, situations, sets of conditions, etc. that may not be identical to one another but that are sufficiently similar to permit comparison there between so that conclusions may reasonably be drawn based on differences or similarities observed. Those of ordinary skill in the art will understand, in context, what degree of identity is required in any given circumstance for two or more such agents, entities, situations, sets of conditions, etc. to be considered comparable. “Corresponding to”, as used herein designates the position/identity of an amino acid residue in a polypeptide of interest. Those of ordinary skill will appreciate that, for purposes of simplicity, residues in a polypeptide are often designated using a canonical numbering system based on a reference related polypeptide, so that an amino acid “corresponding to” a residue at position 190, for example, need not actually be the 190thamino acid in a particular amino acid chain but rather corresponds to the residue found at 190 in the reference polypeptide; those of ordinary skill in the art readily appreciate how to identify “corresponding” amino acids. “Detection Agents”, as described herein, refer to moieties or agents that are amenable to detection, for example, due to their specific structural and/or chemical characteristics, and/or their functional properties. Non-limiting examples of such agents include enzymes, radiolabels, haptens, fluorescent labels, phosphorescent molecules, chemiluminescent molecules, chromophores, luminescent molecules, photoaffinity molecules, colored particles or ligands, such as biotin. Many detection agents are known in the art, as are systems for their attachment to antibodies (see, for e.g., U.S. Pat. Nos. 5,021,236; 4,938,948; and 4,472,509, each incorporated herein by reference). Particular examples may include paramagnetic ions, radioactive isotopes, fluorochromes, NMR-detectable substances, X-ray imaging agents, among others. In some embodiments of the present invention, the conjugated detection agent is a diagnostic or imaging agent. “Dosage form” and “unit dosage form”, as used herein, the term “dosage form” refers to physically discrete unit of a therapeutic agent for a subject (e.g., a human patient) to be treated. Each unit contains a predetermined quantity of active material calculated or demonstrated to produce a desired therapeutic effect when administered to a relevant population according to an appropriate dosing regimen. For example, in some embodiments, such quantity is a unit dosage amount (or a whole fraction thereof) appropriate for administration in accordance with a dosing regimen that has been determined to correlate with a desired or beneficial outcome when administered to a relevant population (i.e., with a therapeutic dosing regimen). It will be understood, however, that the total dosage administered to any particular patient will be selected by a medical professional (e.g., a medical doctor) within the scope of sound medical judgment. “Dosing regimen” (or “therapeutic regimen”), as used herein is a set of unit doses (typically more than one) that are administered individually to a subject, typically separated by periods of time. In some embodiments, a given therapeutic agent has a recommended dosing regimen, which may involve one or more doses. In some embodiments, a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regimen comprises a plurality of doses and at least two different time periods separating individual doses. In some embodiments, the therapeutic agent is administered continuously (e.g., by infusion) over a predetermined period. In some embodiments, a therapeutic agent is administered once a day (QD) or twice a day (BID). In some embodiments, a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regimen comprises a plurality of doses and at least two different time periods separating individual doses. In some embodiments, all doses within a dosing regimen are of the same unit dose amount. In some embodiments, different doses within a dosing regimen are of different amounts. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount different from the first dose amount. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount same as the first dose amount. In some embodiments, a dosing regimen is correlated with a desired or beneficial outcome when administered across a relevant population (i.e., is a therapeutic dosing regimen). “Effector function” as used herein refers a biochemical event that results from the interaction of an antibody Fc region with an Fc receptor or ligand. Effector functions include but are not limited to antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), and complement-mediated cytotoxicity (CMC). In some embodiments, an effector function is one that operates after the binding of an antigen, one that operates independent of antigen binding, or both. “Effector cell” as used herein refers to a cell of the immune system that expresses one or more Fc receptors and mediates one or more effector functions. In some embodiments, effector cells may include, but may not be limited to, one or more of monocytes, macrophages, neutrophils, dendritic cells, eosinophils, mast cells, platelets, large granular lymphocytes, Langerhans' cells, natural killer (NK) cells, T-lymphocytes, B-lymphocytes and may be from any organism including but not limited to humans, mice, rats, rabbits, and monkeys. “Engineered” as used herein refers, in general, to the aspect of having been manipulated by the hand of man. For example, in some embodiments, a polynucleotide may be considered to be “engineered” when two or more sequences, that are not linked together in that order in nature, are manipulated by the hand of man to be directly linked to one another in the engineered polynucleotide. In some particular such embodiments, an engineered polynucleotide may comprise a regulatory sequence that is found in nature in operative association with a first coding sequence but not in operative association with a second coding sequence, is linked by the hand of man so that it is operatively associated with the second coding sequence. Alternatively or additionally, in some embodiments, first and second nucleic acid sequences that each encode polypeptide elements or domains that in nature are not linked to one another may be linked to one another in a single engineered polynucleotide. Comparably, in some embodiments, a cell or organism may be considered to be “engineered” if it has been manipulated so that its genetic information is altered (e.g., new genetic material not previously present has been introduced, or previously present genetic material has been altered or removed). As is common practice and is understood by those in the art, progeny of an engineered polynucleotide or cell are typically still referred to as “engineered” even though the actual manipulation was performed on a prior entity. Furthermore, as will be appreciated by those skilled in the art, a variety of methodologies are available through which “engineering” as described herein may be achieved. For example, in some embodiments, “engineering” may involve selection or design (e.g., of nucleic acid sequences, polypeptide sequences, cells, tissues, and/or organisms) through use of computer systems programmed to perform analysis or comparison, or otherwise to analyze, recommend, and/or select sequences, alterations, etc). Alternatively or additionally, in some embodiments, “engineering” may involve use of in vitro chemical synthesis methodologies and/or recombinant nucleic acid technologies such as, for example, for example, nucleic acid amplification [e.g., via the polymerase chain reaction], hybridization, mutation, transformation, transfection, etc, and/or any of a variety of controlled mating methodologies). As will be appreciated by those skilled in the art, a variety of established such techniques (e.g., for for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation [e.g., electroporation, lipofection, etc] are well known in the art and described in various general and more specific references that are cited and/or discussed throughout the present specification. See e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. [1989]), which is incorporated herein by reference for any purpose. “Epitope”, as used herein, includes any moiety that is specifically recognized by an immunoglobulin (e.g., antibody or receptor) binding component. In some embodiments, an epitope is comprised of a plurality of chemical atoms or groups on an antigen. In some embodiments, such chemical atoms or groups are surface-exposed when the antigen adopts a relevant three-dimensional conformation. In some embodiments, such chemical atoms or groups are physically near to each other in space when the antigen adopts such a conformation. In some embodiments, at least some such chemical atoms are groups are physically separated from one another when the antigen adopts an alternative conformation (e.g., is linearized). “Excipient”, as used herein, refers to a non-therapeutic agent that may be included in a pharmaceutical composition, for example to provide or contribute to a desired consistency or stabilizing effect. Suitable pharmaceutical excipients include, for example, starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. “Fc ligand” as used herein refers to a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an antibody to form an Fc-ligand complex. Fc ligands include but are not limited to FcγRIIA (CD32A), FcγRIIB (CD32B), FcγRIIIA (CD16A), FcγRIIIB (CD16B), FcγRI (CD64), FccRII (CD23), FcRn, C1q, C3, staphylococcal protein A, streptococcal protein G, and viral FcγR. Fc ligands may include undiscovered molecules that bind Fc. “Fluorescent Label”, as is understood in the art, is a moiety or entity that has fluorescent character and, in some embodiments, may be detectable based on such fluorescence. In some embodiments, a fluorescent label may be or may comprise one or more of Alexa 350, Alexa 430, AMCA, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, Cascade Blue, Cy3, Cy5,6-FAM, Fluorescein Isothiocyanate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rhodamine Green, Rhodamine Red, Renographin, ROX, TAMRA, TET, Tetramethylrhodamine, and/or Texas Red, among others. “Framework” or “framework region”, as used herein, refers to the sequences of a variable region minus the CDRs. Because a CDR sequence can be determined by different systems, likewise a framework sequence is subject to correspondingly different interpretations. The six CDRs divide the framework regions on the heavy and light chains into four sub-regions (FR1, FR2, FR3 and FR4) on each chain, in which CDR1 is positioned between FR1 and FR2, CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4. Without specifying the particular sub-regions as FR1, FR2, FR3 or FR4, a framework region, as referred by others, represents the combined FRs within the variable region of a single, naturally occurring immunoglobulin chain. As used herein, a FR represents one of the four sub-regions, FR1, for example, represents the first framework region closest to the amino terminal end of the variable region and 5′ with respect to CDR1, and FRs represents two or more of the sub-regions constituting a framework region. “Host cell”, as used herein, refers to a cell into which exogenous DNA (recombinant or otherwise) has been introduced. Persons of skill upon reading this disclosure will understand that such terms refer not only to the particular subject cell, but also to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein. In some embodiments, host cells include prokaryotic and eukaryotic cells selected from any of the Kingdoms of life that are suitable for expressing an exogenous DNA (e.g., a recombinant nucleic acid sequence). Exemplary cells include those of prokaryotes and eukaryotes (single-cell or multiple-cell), bacterial cells (e.g., strains ofE. coli, Bacillusspp.,Streptomycesspp., etc.), mycobacteria cells, fungal cells, yeast cells (e.g.,S. cerevisiae, S. pombe, P. pastoris, P. methanolica, etc.), plant cells, insect cells (e.g., SF-9, SF-21, baculovirus-infected insect cells,Trichoplusia ni, etc.), non-human animal cells, human cells, or cell fusions such as, for example, hybridomas or quadromas. In some embodiments, the cell is a human, monkey, ape, hamster, rat, or mouse cell. In some embodiments, the cell is eukaryotic and is selected from the following cells: CHO (e.g., CHO Kl, DXB-1 1 CHO, Veggie-CHO), COS (e.g., COS-7), retinal cell, Vero, CV1, kidney (e.g., HEK293, 293 EBNA, MSR 293, MDCK, HaK, BHK), HeLa, HepG2, WI38, MRC 5, Colo205, HB 8065, HL-60, (e.g., BHK21), Jurkat, Daudi, A431 (epidermal), CV-1, U937, 3T3, L cell, C127 cell, SP2/0, NS-0, MMT 060562, Sertoli cell, BRL 3 A cell, HT1080 cell, myeloma cell, tumor cell, and a cell line derived from an aforementioned cell. In some embodiments, the cell comprises one or more viral genes, e.g., a retinal cell that expresses a viral gene (e.g., a PER.C6™ cell). “Human antibody”, as used herein, is intended to include antibodies having variable and constant regions generated (or assembled) from human immunoglobulin sequences. In some embodiments, antibodies (or antibody components) may be considered to be “human” even though their amino acid sequences include residues or elements not encoded by human germline immunoglobulin sequences (e.g., include sequence variations, for example that may (originally) have been introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in one or more CDRs and in particular CDR3. “Humanized”, as is known in the art, the term “humanized” is commonly used to refer to antibodies (or antibody components) whose amino acid sequence includes VHand VLregion sequences from a reference antibody raised in a non-human species (e.g., a mouse), but also includes modifications in those sequences relative to the reference antibody intended to render them more “human-like”, i.e., more similar to human germline variable sequences. In some embodiments, a “humanized” antibody (or antibody component) is one that immunospecifically binds to an antigen of interest and that has a framework (FR) region having substantially the amino acid sequence as that of a human antibody, and a complementary determining region (CDR) having substantially the amino acid sequence as that of a non-human antibody. A humanized antibody comprises substantially all of at least one, and typically two, variable domains (Fab, Fab′, F(ab′)2, FabC, Fv) in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor immunoglobulin) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. In some embodiments, a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin constant region. In some embodiments, a humanized antibody contains both the light chain as well as at least the variable domain of a heavy chain. The antibody also may include a CH1, hinge, CH2, CH3, and, optionally, a CH4 region of a heavy chain constant region. In some embodiments, a humanized antibody only contains a humanized VLregion. In some embodiments, a humanized antibody only contains a humanized VHregion. In some certain embodiments, a humanized antibody contains humanized VHand VLregions. “Improve,” “increase” or “reduce,” as used herein or grammatical equivalents thereof, indicate values that are relative to a baseline measurement, such as a measurement in the same individual prior to initiation of a treatment described herein, or a measurement in a control individual (or multiple control individuals) in the absence of the treatment described herein. A “control individual” is an individual afflicted with the same form of disease or injury as the individual being treated. “In vitro”, as used herein refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, etc., rather than within a multi-cellular organism. “In vivo”, as used herein refers to events that occur within a multi-cellular organism, such as a human and a non-human animal. In the context of cell-based systems, the term may be used to refer to events that occur within a living cell (as opposed to, for example, in vitro systems). “Isolated”, as used herein, refers to a substance and/or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature and/or in an experimental setting), and/or (2) designed, produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% of the other components with which they were initially associated. In some embodiments, isolated agents are about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. As used herein, a substance is “pure” if it is substantially free of other components. In some embodiments, as will be understood by those skilled in the art, a substance may still be considered “isolated” or even “pure”, after having been combined with certain other components such as, for example, one or more carriers or excipients (e.g., buffer, solvent, water, etc.); in such embodiments, percent isolation or purity of the substance is calculated without including such carriers or excipients. To give but one example, in some embodiments, a biological polymer such as a polypeptide or polynucleotide that occurs in nature is considered to be “isolated” when, a) by virtue of its origin or source of derivation is not associated with some or all of the components that accompany it in its native state in nature; b) it is substantially free of other polypeptides or nucleic acids of the same species from the species that produces it in nature; c) is expressed by or is otherwise in association with components from a cell or other expression system that is not of the species that produces it in nature. Thus, for instance, in some embodiments, a polypeptide that is chemically synthesized or is synthesized in a cellular system different from that which produces it in nature is considered to be an “isolated” polypeptide. Alternatively or additionally, in some embodiments, a polypeptide that has been subjected to one or more purification techniques may be considered to be an “isolated” polypeptide to the extent that it has been separated from other components a) with which it is associated in nature; and/or b) with which it was associated when initially produced. “KD”, as used herein, refers to the dissociation constant of a binding agent (e.g., an antibody or binding component thereof) from a complex with its partner (e.g., the epitope to which the antibody or binding component thereof binds). “koff”, as used herein, refers to the off rate constant for dissociation of a binding agent (e.g., an antibody or binding component thereof) from a complex with its partner (e.g., the epitope to which the antibody or binding component thereof binds). “kon”, as used herein, refers to the on rate constant for association of a binding agent (e.g., an antibody or binding component thereof) with its partner (e.g., the epitope to which the antibody or binding component thereof binds). “Linker”, as used herein, typically refers to a portion of a molecule or entity that connects two or more different regions of interest (e.g., particular structural and/or functional domains or moieties of interest). In some embodiments, a linker does not participate significantly in the relevant function of interest (e.g., so that presence or absence of the linker, in association with the relevant domain or moiety of interest does not materially alter the relevant function of the domain or moiety). In some embodiments, a linker in characterized by lack of defined or rigid structure. In some embodiments, particularly when one or more domains or moieties of interest is/are comprised of a polypeptide, a linker is or comprises a polypeptide. In some particular embodiments, a polypeptide (e.g., an engineered polypeptide) as described herein may have general structure S1-L-S2, wherein S1 and S2 are the moieties or domains of interest. In some embodiments, one or both of S1 and S2 may be or comprise a binding element (e.g., an antibody component) as described herein. In some embodiments, a polypeptide linker may be 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more amino acids long. In some embodiments, a polypeptide linker may have an amino acid sequence that is or comprises a sequence as described in Holliger, P., et al., 1993, Proc. Natl. Acad. Sci. USA 90:6444-6448 or Poljak, R. J., et al., 1994, Structure 2: 1121-1123. In some embodiments, a polypeptide linker may have an amino acid sequence that is or comprises GGGGSGGGGSGGGGS (i.e., [G4S]3) (SEQ ID NO: 8) or GGGGSGGGGSGGGGSGGGGSGGGGSGGGGS (i.e., [G4S]6) (SEQ ID NO: 9). “Multivalent binding agent”, as used herein, refers a binding agent capable of binding to two or more antigens, which can be on the same molecule or on different molecules. Multivalent binding agents as described herein are, in some embodiments, engineered to have the three or more antigen binding sites, and are typically not naturally occurring proteins. Multivalent binding agents as described herein refer to binding agents capable of binding two or more related or unrelated targets. Multivalent binding agents may be composed of multiple copies of a single antibody component or multiple copies of different antibody components. Such binding agents are capable of binding to two or more antigens and are tetravalent or multivalent binding agents. Multivalent binding agents may additionally comprise a therapeutic agent, such as, for example, an immunomodulator, toxin or an RNase. Multivalent binding agents as described herein are, in some embodiments, capable of binding simultaneously to at least two targets that are of different structure, e.g., two different antigens, two different epitopes on the same antigen, or a hapten and/or an antigen or epitope. In many embodiments, multivalent binding agents of the present invention are proteins engineered to have characteristics of multivalent binding agents as described herein. Multivalent binding agents of the present invention may be monospecific (capable of binding one antigen) or multispecific (capable of binding two or more antigens), and may be composed of two heavy chain polypeptides and two light chain polypeptides. Each binding site, in some embodiments, is composed of a heavy chain variable domain and a light chain variable domain with a total of six CDRs involved in antigen binding per antigen binding site. “Nucleic acid”, as used herein, in its broadest sense, refers to any compound and/or substance that is or can be incorporated into an oligonucleotide chain. In some embodiments, a nucleic acid is a compound and/or substance that is or can be incorporated into an oligonucleotide chain via a phosphodiester linkage. As will be clear from context, in some embodiments, “nucleic acid” refers to individual nucleic acid residues (e.g., nucleotides and/or nucleosides); in some embodiments, “nucleic acid” refers to an oligonucleotide chain comprising individual nucleic acid residues. In some embodiments, a “nucleic acid” is or comprises RNA; in some embodiments, a “nucleic acid” is or comprises DNA. In some embodiments, a nucleic acid is, comprises, or consists of one or more natural nucleic acid residues. In some embodiments, a nucleic acid is, comprises, or consists of one or more nucleic acid analogs. In some embodiments, a nucleic acid analog differs from a nucleic acid in that it does not utilize a phosphodiester backbone. For example, in some embodiments, a nucleic acid is, comprises, or consists of one or more “peptide nucleic acids”, which are known in the art and have peptide bonds instead of phosphodiester bonds in the backbone, are considered within the scope of the present invention. Alternatively or additionally, in some embodiments, a nucleic acid has one or more phosphorothioate and/or 5′-N-phosphoramidite linkages rather than phosphodiester bonds. In some embodiments, a nucleic acid is, comprises, or consists of one or more natural nucleosides (e.g., adenosine, thymidine, guanosine, cytidine, uridine, deoxyadenosine, deoxythymidine, deoxy guanosine, and deoxycytidine). In some embodiments, a nucleic acid is, comprises, or consists of one or more nucleoside analogs (e.g., 2-aminoadenosine, 2-thiothymidine, inosine, pyrrolo-pyrimidine, 3-methyl adenosine, 5-methylcytidine, C-5 propynyl-cytidine, C-5 propynyl-uridine, 2-aminoadenosine, C5-bromouridine, C5-fluorouridine, C5-iodouridine, C5-propynyl-uridine, C5-propynyl-cytidine, C5-methylcytidine, 2-aminoadenosine, 7-deazaadenosine, 7-deazaguanosine, 8-oxoadenosine, 8-oxoguanosine, 0(6)-methylguanine, 2-thiocytidine, methylated bases, intercalated bases, and combinations thereof). In some embodiments, a nucleic acid comprises one or more modified sugars (e.g., 2′-fluororibose, ribose, 2′-deoxyribose, arabinose, and hexose) as compared with those in natural nucleic acids. In some embodiments, a nucleic acid has a nucleotide sequence that encodes a functional gene product such as an RNA or protein. In some embodiments, a nucleic acid includes one or more introns. In some embodiments, nucleic acids are prepared by one or more of isolation from a natural source, enzymatic synthesis by polymerization based on a complementary template (in vivo or in vitro), reproduction in a recombinant cell or system, and chemical synthesis. In some embodiments, a nucleic acid is at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 20, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000 or more residues long. In some embodiments, a nucleic acid is single stranded; in some embodiments, a nucleic acid is double stranded. In some embodiments a nucleic acid has a nucleotide sequence comprising at least one element that encodes, or is the complement of a sequence that encodes, a polypeptide. In some embodiments, a nucleic acid has enzymatic activity. “Operably linked”, as used herein, refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences. “Operably linked” sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest. The term “expression control sequence” as used herein refers to polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion. The nature of such control sequences differs depending upon the host organism. For example, in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence, while in eukaryotes, typically, such control sequences include promoters and transcription termination sequence. The term “control sequences” is intended to include components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences. “Paramagnetic Ion”, as is understood in the art, refers to an ion with paramagnetic character. In some embodiments, a paramagnetic ion is one or more of chromium (III), manganese (II), iron (III), iron (II), cobalt (II), nickel (II), copper (II), neodymium (III), samarium (III), ytterbium (III), gadolinium (III), vanadium (II), terbium (III), dysprosium (III), holmium (III), erbium (III), lanthanum (III), gold (III), lead (II), and/or bismuth (III). “Payload”, as used herein, refers to a moiety or entity that is delivered to a site of interest (e.g., to a cell, tissue, tumor, or organism) by association with another entity. In some embodiments, a payload is or comprises a detection agent. In some embodiments, a payload entity is or comprises a therapeutic agent. In some embodiments, a payload entity is or comprises a catalytic agent. Those of ordinary skill in the art will appreciate that a payload entity may be of any chemical class. For example, in some embodiments, a payload entity may be or comprise a carbohydrate, an isotope, a lipid, a nucleic acid, a metal, a nanoparticle (e.g., a ceramic or polymer nanoparticle), polypeptide, a small molecule, a virus, etc. To give but a few examples, in some embodiments, a therapeutic agent payload may be or comprise a toxin (e.g., a toxic peptide, small molecule, or isotope [e.g., radioisotope]); in some embodiments, a detection agent payload may be or comprise a fluorescent entity or agent, a radioactive entity or agent, an agent or entity detectable by binding (e.g., a tag, a hapten, a ligand, etc), a catalytic agent, etc. “Physiological conditions”, as used herein, has its art-understood meaning referencing conditions under which cells or organisms live and/or reproduce. In some embodiments, the term refers to conditions of the external or internal milieu that may occur in nature for an organism or cell system. In some embodiments, physiological conditions are those conditions present within the body of a human or non-human animal, especially those conditions present at and/or within a surgical site. Physiological conditions typically include, e.g., a temperature range of 20 to 40° C., atmospheric pressure of 1, pH of 6 to 8, glucose concentration of 1 to 20 mM, oxygen concentration at atmospheric levels, and gravity as it is encountered on earth. In some embodiments, conditions in a laboratory are manipulated and/or maintained at physiologic conditions. In some embodiments, physiological conditions are encountered in an organism. “Polypeptide”, as used herein, refers to any polymeric chain of amino acids. In some embodiments, a polypeptide has an amino acid sequence that occurs in nature. In some embodiments, a polypeptide has an amino acid sequence that does not occur in nature. In some embodiments, a polypeptide has an amino acid sequence that is engineered in that it is designed and/or produced through action of the hand of man. In some embodiments, a polypeptide may comprise or consist of natural amino acids, non-natural amino acids, or both. In some embodiments, a polypeptide may comprise or consist of only natural amino acids or only non-natural amino acids. In some embodiments, a polypeptide may comprise D-amino acids, L-amino acids, or both. In some embodiments, a polypeptide may comprise only D-amino acids. In some embodiments, a polypeptide may comprise only L-amino acids. In some embodiments, a polypeptide may include one or more pendant groups or other modifications, e.g., modifying or attached to one or more amino acid side chains, at the polypeptide's N-terminus, at the polypeptide's C-terminus, or any combination thereof. In some embodiments, such pendant groups or modifications may be selected from the group consisting of acetylation, amidation, lipidation, methylation, pegylation, etc., including combinations thereof. In some embodiments, a polypeptide may be cyclic, and/or may comprise a cyclic portion. In some embodiments, a polypeptide is not cyclic and/or does not comprise any cyclic portion. In some embodiments, a polypeptide is linear. In some embodiments, a polypeptide may be or comprise a stapled polypeptide. In some embodiments, the term “polypeptide” may be appended to a name of a reference polypeptide, activity, or structure; in such instances it is used herein to refer to polypeptides that share the relevant activity or structure and thus can be considered to be members of the same class or family of polypeptides. For each such class, the present specification provides and/or those skilled in the art will be aware of exemplary polypeptides within the class whose amino acid sequences and/or functions are known; in some embodiments, such exemplary polypeptides are reference polypeptides for the polypeptide class. In some embodiments, a member of a polypeptide class or family shows significant sequence homology or identity with, shares a common sequence motif (e.g., a characteristic sequence element) with, and/or shares a common activity (in some embodiments at a comparable level or within a designated range) with a reference polypeptide of the class; in some embodiments with all polypeptides within the class). For example, in some embodiments, a member polypeptide shows an overall degree of sequence homology or identity with a reference polypeptide that is at least about 30 to 40%, and is often greater than about 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more and/or includes at least one region (i.e., a conserved region that may in some embodiments may be or comprise a characteristic sequence element) that shows very high sequence identity, often greater than 90% or even 95%, 96%, 97%, 98%, or 99%. Such a conserved region usually encompasses at least three to four and often up to 20 or more amino acids; in some embodiments, a conserved region encompasses at least one stretch of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more contiguous amino acids. In some embodiments, a useful polypeptide may comprise or consist of a fragment of a parent polypeptide. In some embodiments, a useful polypeptide as may comprise or consist of a plurality of fragments, each of which is found in the same parent polypeptide in a different spatial arrangement relative to one another than is found in the polypeptide of interest (e.g., fragments that are directly linked in the parent may be spatially separated in the polypeptide of interest or vice-versa, and/or fragments may be present in a different order in the polypeptide of interest than in the parent), so that the polypeptide of interest is a derivative of its parent polypeptide “Prevent” or “prevention”, as used herein when used in connection with the occurrence of a disease, disorder, and/or condition, refers to reducing the risk of developing the disease, disorder and/or condition and/or to delaying onset of one or more characteristics or symptoms of the disease, disorder or condition. Prevention may be considered complete when onset of a disease, disorder or condition has been delayed for a predefined period of time. “Radioactive Isotope”: The term “radioactive isotope” as used herein has its art-understood meaning referring to an isotope that undergoes radioactive decay. In some embodiments, a radioactive isotope may be or comprise one or more of actinium-225, astatine-211, bismuth-212, carbon-14, chromium-51, chlorine-36, cobalt-57, cobalt-58, copper-67, Europium-152, gallium-67, hydrogen-3, iodine-123, iodine-124, iodine-125, iodine-131, indium-111, iron-59, lead-212, lutetium-177, phosphorus-32, radium-223, radium-224, rhenium-186, rhenium-188, selenium-75, sulphur-35, technicium-99m, thorium-227, yttrium-90, and zirconium-89. “Recombinant”, as used herein, is intended to refer to polypeptides (e.g., antibodies or antibody components, or multispecific binding agents as described herein) that are designed, engineered, prepared, expressed, created or isolated by recombinant means, such as polypeptides expressed using a recombinant expression vector transfected into a host cell, polypeptides isolated from a recombinant, combinatorial human polypeptide library (Hoogenboom H. R., 1997, TIB Tech. 15:62-70; Azzazy H., and Highsmith W. E., 2002, Clin. Biochem. 35:425-445; Gavilondo, J. V. and Larrick, J. W., 2002, BioTechniques 29: 128-145; Hoogenboom H., and Chames, P., 2000, Immunology Today 21:371-378), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor, L. D. et al., 1992, Nucl. Acids Res. 20:6287-6295; Little M. et al., 2000, Immunology Today 21:364-370; Kellermann S-A., and Green L. L., 2002, Current Opinion in Biotechnology 13:593-597; Murphy, A. J. et al., 2014, Proc. Natl. Acad. Sci. U.S.A. 111(14):5153-5158) or polypeptides prepared, expressed, created or isolated by any other means that involves splicing selected sequence elements to one another. In some embodiments, one or more of such selected sequence elements is found in nature. In some embodiments, one or more of such selected sequence elements is designed in silico. In some embodiments, one or more such selected sequence elements results from mutagenesis (e.g., in vivo or in vitro) of a known sequence element, e.g., from a natural or synthetic source. For example, in some embodiments, a recombinant antibody polypeptide is comprised of sequences found in the germline of a source organism of interest (e.g., human, mouse, etc.). In some embodiments, a recombinant antibody has an amino acid sequence that resulted from mutagenesis (e.g., in vitro or in vivo, for example in a transgenic animal), so that the amino acid sequences of the VHand VLregions of the recombinant antibodies are sequences that, while originating from and related to germline VHand VLsequences, may not naturally exist within the germline antibody repertoire in vivo. “Recovering”, as used herein, refers to the process of rendering an agent or entity substantially free of other previously-associated components, for example by isolation, e.g., using purification techniques known in the art. In some embodiments, an agent or entity is recovered from a natural source and/or a source comprising cells. “Reference”, as used herein describes a standard, control, or other appropriate reference against which a comparison is made as described herein. For example, in some embodiments, a reference is a standard or control agent, animal, individual, population, sample, sequence, series of steps, set of conditions, or value against which an agent, animal, individual, population, sample, sequence, series of steps, set of conditions, or value of interest is compared. In some embodiments, a reference is tested and/or determined substantially simultaneously with the testing or determination of interest. In some embodiments, a reference is a historical reference, optionally embodied in a tangible medium. Typically, as would be understood by those skilled in the art, a reference is determined or characterized under conditions comparable to those utilized in the assessment of interest. “Risk”, as will be understood from context, “risk” of a disease, disorder, and/or condition comprises likelihood that a particular individual will develop a disease, disorder, and/or condition (e.g., a radiation injury). In some embodiments, risk is expressed as a percentage. In some embodiments, risk is from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90 and up to 100%. In some embodiments risk is expressed as a risk relative to a risk associated with a reference sample or group of reference samples. In some embodiments, a reference sample or group of reference samples have a known risk of a disease, disorder, condition and/or event (e.g., a radiation injury). In some embodiments a reference sample or group of reference samples are from individuals comparable to a particular individual. In some embodiments, relative risk is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more. “Specific binding”, as used herein, refers to a binding agent's ability to discriminate between possible partners in the environment in which binding is to occur. A binding agent that interacts with one particular target when other potential targets are present is said to “bind specifically” to the target with which it interacts. In some embodiments, specific binding is assessed by detecting or determining degree of association between the binding agent and its partner; in some embodiments, specific binding is assessed by detecting or determining degree of dissociation of a binding agent-partner complex; in some embodiments, specific binding is assessed by detecting or determining ability of the binding agent to compete an alternative interaction between its partner and another entity. In some embodiments, specific binding is assessed by performing such detections or determinations across a range of concentrations. “Subject”, as used herein, means any mammal, including humans. In certain embodiments of the present invention the subject is an adult, an adolescent or an infant. In some embodiments, terms “individual” or “patient” are used and are intended to be interchangeable with “subject”. Also contemplated by the present invention are the administration of the pharmaceutical compositions and/or performance of the methods of treatment in-utero. “Substantially”: As used herein, the term “substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term “substantially” is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena. “Substantial sequence homology”, as used herein refers to a comparison between amino acid or nucleic acid sequences. As will be appreciated by those of ordinary skill in the art, two sequences are generally considered to be “substantially homologous” if they contain homologous residues in corresponding positions. Homologous residues may be identical residues. Alternatively, homologous residues may be non-identical residues will appropriately similar structural and/or functional characteristics. For example, as is well known by those of ordinary skill in the art, certain amino acids are typically classified as “hydrophobic” or “hydrophilic” amino acids, and/or as having “polar” or “non-polar” side chains. Substitution of one amino acid for another of the same type may often be considered a “homologous” substitution. Typical amino acid categorizations are summarized in Table 1 and 2. TABLE 1AlanineAlaANonpolarNeutral1.8ArginineArgRPolarPositive−4.5AsparagineAsnNPolarNeutral−3.5Aspartic acidAspDPolarNegative−3.5CysteineCysCNonpolarNeutral2.5Glutamic acidGluEPolarNegative−3.5GlutamineGlnQPolarNeutral−3.5GlycineGlyGNonpolarNeutral−0.4HistidineHisHPolarPositive−3.2IsoleucineIleINonpolarNeutral4.5LeucineLeuLNonpolarNeutral3.8LysineLysKPolarPositive−3.9MethionineMetMNonpolarNeutral1.9PhenylalaninePheFNonpolarNeutral2.8ProlineProPNonpolarNeutral−1.6SerineSerSPolarNeutral−0.8ThreonineThrTPolarNeutral−0.7TryptophanTrpWNonpolarNeutral−0.9TyrosineTyrYPolarNeutral−1.3ValineValVNonpolarNeutral4.2 TABLE 2Ambiguous Amino Acids3-Letter1-LetterAsparagine or aspartic acidAsxBGlutamine or glutamic acidGlxZLeucine or IsoleucineXleJUnspecified or unknown amino acidXaaX As is well known in this art, amino acid or nucleic acid sequences may be compared using any of a variety of algorithms, including those available in commercial computer programs such as BLASTN for nucleotide sequences and BLASTP, gapped BLAST, and PSI-BLAST for amino acid sequences. Exemplary such programs are described in Altschul et al., 1990, J. Mol. Biol., 215(3): 403-410; Altschul et al., 1996, Methods in Enzymology 266:460-80; Altschul et al., 1997, Nucleic Acids Res. 25:3389-3402; Baxevanis et al., 1998, Bioinformatics: A Practical Guide to the Analysis of Genes and Proteins, Wiley; and Misener et al., (eds.), Bioinformatics Methods and Protocols (Methods in Molecular Biology, Vol. 132), Humana Press, 1999; all of the foregoing of which are incorporated herein by reference. In addition to identifying homologous sequences, the programs mentioned above typically provide an indication of the degree of homology. In some embodiments, two sequences are considered to be substantially homologous if at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or more of their corresponding residues are homologous over a relevant stretch of residues. In some embodiments, the relevant stretch is a complete sequence. In some embodiments, the relevant stretch is at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 125, at least 150, at least 175, at least 200, at least 225, at least 250, at least 275, at least 300, at least 325, at least 350, at least 375, at least 400, at least 425, at least 450, at least 475, at least 500 or more residues. “Substantial identity”, as used herein refers to a comparison between amino acid or nucleic acid sequences. As will be appreciated by those of ordinary skill in the art, two sequences are generally considered to be “substantially identical” if they contain identical residues in corresponding positions. As is well known in this art, amino acid or nucleic acid sequences may be compared using any of a variety of algorithms, including those available in commercial computer programs such as BLASTN for nucleotide sequences and BLASTP, gapped BLAST, and PSI-BLAST for amino acid sequences. Exemplary such programs are described in Altschul et al., 1990, J. Mol. Biol., 215(3): 403-410; Altschul et al., 1996, Methods in Enzymology 266:460-80; Altschul et al., 1997, Nucleic Acids Res. 25:3389-3402; Baxevanis et al., 1998, Bioinformatics: A Practical Guide to the Analysis of Genes and Proteins, Wiley; and Misener et al., (eds.), Bioinformatics Methods and Protocols (Methods in Molecular Biology, Vol. 132), Humana Press, 1999. In addition to identifying identical sequences, the programs mentioned above typically provide an indication of the degree of identity. In some embodiments, two sequences are considered to be substantially identical if at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more of their corresponding residues are identical over a relevant stretch of residues. In some embodiments, the relevant stretch is a complete sequence. In some embodiments, the relevant stretch is at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500 or more residues. In the context of a CDR, reference to “substantial identity” typically refers to a CDR having an amino acid sequence at least 80%, preferably at least 85%, at least 90%, at least 95%, at least 98% or at least 99% identical to that of a reference CDR. “Surface plasmon resonance”, as used herein, refers to an optical phenomenon that allows for the analysis of specific binding interactions in real-time, for example through detection of alterations in protein concentrations within a biosensor matrix, such as by using a BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.). For further descriptions, see Jonsson, U., et al., 1993, Ann. Biol. Clin. 51:19-26; Jonsson, U., et al., 1991, Biotechniques 11:620-627; Johnsson, B., et al., 1995, J. Mol. Recognit. 8:125-131; and Johnnson, B., et al., 1991, Anal. Biochem. 198:268-277. “Therapeutically effective amount”, as used herein, is meant an amount that produces the desired effect for which it is administered. In some embodiments, the term refers to an amount that is sufficient, when administered to a population suffering from or susceptible to a disease, disorder, and/or condition in accordance with a therapeutic dosing regimen, to treat the disease, disorder, and/or condition. In some embodiments, a therapeutically effective amount is one that reduces the incidence and/or severity of, and/or delays onset of, one or more symptoms of the disease, disorder, and/or condition. Those of ordinary skill in the art will appreciate that the term “therapeutically effective amount” does not in fact require successful treatment be achieved in a particular individual. Rather, a therapeutically effective amount may be that amount that provides a particular desired pharmacological response in a significant number of subjects when administered to patients in need of such treatment. In some embodiments, reference to a therapeutically effective amount may be a reference to an amount as measured in one or more specific tissues (e.g., a tissue affected by the disease, disorder or condition) or fluids (e.g., blood, saliva, serum, sweat, tears, urine, etc.). Those of ordinary skill in the art will appreciate that, in some embodiments, a therapeutically effective amount of a particular agent or therapy may be formulated and/or administered in a single dose. In some embodiments, a therapeutically effective agent may be formulated and/or administered in a plurality of doses, for example, as part of a dosing regimen. “Transformation”, as used herein, refers to any process by which exogenous DNA is introduced into a host cell. Transformation may occur under natural or artificial conditions using various methods well known in the art. Transformation may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. In some embodiments, a particular transformation methodology is selected based on the host cell being transformed and may include, but is not limited to, viral infection, electroporation, mating, lipofection. In some embodiments, a “transformed” cell is stably transformed in that the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome. In some embodiments, a transformed cell transiently expresses introduced nucleic acid for limited periods of time. “Vector”, as used herein, refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “expression vectors.” DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS The present invention demonstrates the successful construction of a multi-specific binding agent (e.g., bispecific antibody) that binds an established antigen on human colorectal cancers. In particular, the present disclosure specifically demonstrates the successful targeting of radioimmunotherapy in colorectal cancer using a bispecific antibody that binds to human A33 glycoprotein antigen and Benzyl-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA-Bn), provides a specific such bispecific antibody, and demonstrates its surprising usefulness and/or effectiveness. Among other things, the present invention specifically provides the first successful therapeutic use of a bispecific antibody that targets the human A33 antigen, and furthermore provides an improved therapeutic methodology for a pretargeted radioimmunotherapy regimen for treatment of A33-expressing tumors. The present invention also provides “theranostic” (i.e., therapeutic and diagnostic) agents for the simultaneous scintigraphic imaging and radioimmunotherapy of A33-positive cancers, and specifically demonstrates surprising usefulness and/or effectiveness thereof. A33, a glycoprotein antigen on human colorectal cancers with restricted normal tissue expression, is retained on the tumor cell surface after antibody binding for extended periods of time, in contrast to the rapid physiologic turnover of normal gut epithelium—a therapeutic index based on tissue retention unique to gut antigens. Radioimmunoscintigraphy and radioimmunotherapy (RIT) of advanced colorectal cancer (“CRC”) using directly conjugated antibodies (e.g.131I-huA33) has yielded suboptimal tumor dose and therapeutic index (Welt et al., 1994, J. Clin. Oncol. 12:1561-1571). The present invention encompasses the recognition that both of these deficiencies can be overcome using a multi-step pretargeted RIT (PRIT) approach where a bispecific tetravalent huA33-C825 bispecific antibody construct with high affinity for Benzyl-1,4,7,10-tetraazacyclododecane-N,N′,N″,N′″-tetraacetic acid (DOTA-Bn)-radiometal complexes is first targeted to the tumor. The present disclosure specifically demonstrates that subsequent to clearing unbound huA33-C825 bispecific antibody from circulation,177Lu-radiolabeled DOTA-Bn hapten is injected to deliver the tumorcidal dose of PRIT to the A33-positive tumor. To give one specific example, the present disclosure demonstrates that in human colorectal tumor models of SW1222, mice with established subcutaneous tumors can be cured with minimal toxicity to normal tissues including bone marrow and kidney. Without wishing to be bound by theory, we note that data provided herein demonstrate that, in some embodiments, a PRIT regimen that employs a dual-cycle dose of a huA33-C825 bispecific antibody resulted in an tremendous effect on tumor volume as compared to the same PRIT regimen that employed a single-cycle dose. Moreover, the present disclosure demonstrates, among other things, that such dual-cycle dosing of a huA33-C825 bispecific antibody as described herein yielded a complete response in approximately 80% of test subjects. Also demonstrated herein are treatments with additional cycles, which showed highly efficient responses, for example a 3 cycle dosing regimin was curative for 10/10 mice without detectable toxicity in target organs (marrow spleen and kidney). Thus, the present disclosure, in at least some embodiments, embraces the development of an improved PRIT regimen using a bispecific antibody format that effectively targets the human A33 glycoprotein antigen to achieve enhanced tumor targeting and/or tumor ablation with minimal to no clinical or histological radiation toxicity. Human Colorectal Cancer The human A33 antigen is a transmembrane glycoprotein having a molecular weight of 43 kD (213 amino acid polypeptide), and is expressed in more than 95% of human colon cancers with restricted normal expression (colon and bowel epithelium) and minimal shedding into circulation. Initially a murine monoclonal antibody (A33) and later a humanized version (huA33) was developed (King et al., 1995, British J. Cancer 72:1364-1372), and found to have ideal specificity, affinity, and antibody-antigen uptake and internalization properties for use as a targeting agent for radioisotopes for diagnosis and therapy. In a clinical study of124I-huA33 imaging in colorectal cancer patients, differential clearance between antigen-positive tumor and intestine led the authors to conclude that an alternative multi-step approach including initial administration with a non-radioactive bispecific A33 antibody form (or “pretargeting”), followed with a radiolabeled hapten may be preferred (O'Donoghue et al., 2011, J. Nucl. Med. 52:1878-1885). The high tumor persistence of radioiodine forms of A33 (e.g.,1251-A33) prompted extensive investigation of the internalization properties of the A33 antibody-antigen complex, showing that anti-A33 antibodies reside on the surface for extended periods of time, making such an antibody, in some embodiments, particularly well suited for a pretargeting approach (Ackerman et al., 2008, Mol. Cancer Ther. 7(7):2233-2240), in particular, when the normal expression in the gut is allowed to turnover before a last ligand step. The unique physiology of the gut epithelium to shed over one to three days carrying with it antigens and bound antibodies is critical if the target antigen is expressed on these normal cells (Scott et al., 2005, Clin. Cancer Res. 11:4810-4817). As described herein, in PRIT, unbound antibodies are cleared from the blood using a clearing agent (CA) before a last cytotoxic ligand step. The natural shedding of normal gut cells is functionally equivalent to a clearing step in the gut. Pretargeted radioimmunotherapy (PRIT) directed at a variety of other tumor-associated antigens has been investigated for colorectal cancer, including CEA (hMN-14-anti-DTPA-indium+131I-di-DTPA-indium hapten and recently, anti-CEACAMS-anti-histamine-succinyl-glycine “TF2” with177Lu-IMP288 hapten), TAG-72 (CC49 scFv-streptavidin+90Y-DOTA-biotin), Ep-CAM (NR-LU-10-SA with90Y-DOTA-biotin). Using antibodies to target poisons to tumors, e.g., radioimmunotherapy (RIT) with directly conjugated antibodies, has so far been met with limited success due in part to suboptimal tumor dose and therapeutic index (TI). Further, because of normal tissue bystander toxicity, dose escalation is not feasible and therefore such therapy results in limited anti-tumor effect. Thus, the present invention is based on the recognition that because the human A33 glycoprotein antigen is present in colorectal cancers and possesses unique retention properties, a PRIT methodology that achieves log-fold higher TI and complete remissions of established xenografts without toxicity to any major organs could be developed to effectively target human A33 on tumor cells using a bispecific antibody (referred to herein as huA33-C825) having a first antigen-binding site that binds human A33 and a second antigen-binding site with high affinity for DOTA-Bn (metal) complex (e.g., specificity through the single chain Fv (scFv) referred to as C825). As described herein, a PRIT methodology was improved in vivo by titrating doses of huA33-C825, a dextran-based clearing agent (dextran-CA), and a177Lu-radiolabeled DOTA-Bn hapten (177Lu-DOTA-Bn) using a subcutaneous colorectal cancer xenograft model of SW1222. As described herein, bispecific binding agents of the present invention offer dual functionality in diagnostic imaging/dosimetry and therapeutic applications. Targeted radiation therapy, called radioimmunotherapy (RIT), can deliver sufficient radiation to overcome any tumor resistance, as long as the TI is favorable. Current radiolabeled IgG drugs (e.g.90Y-Zevalin) have suboptimal TI of 3:1, borderline for curative therapy where hematological toxicity is dose liming. The present invention is encompasses the recognition that in pretargeted RIT (PRIT) an antibody targeting step is separate from the payload step. The present disclosure appreciates one potential additional advantage of PRIT over conventional MT in that in some embodiments, PRIT may facilitate patient care. In some embodiments, an initial infusion of cold antibody, and in some embodiments an administration of a clearing agent, can be performed in a physician's office (e.g., in the office of a managing physician). In some embodiments, only a step of radiolabeled DOTA-Bn (typically performed subsequent to one or more, and in some embodiments, all, other steps), would need to be done by a nuclear medicine trained physician, and the patient may then be returned to his or her physician's care, once radioactivity has cleared from the body (<24 hours). Therefore, by taking advantage of the unique pharmacokinetics of large and small ligands, the present inventors demonstrate herein that PRIT can be highly effective. The present invention specifically demonstrates that using a fully humanized PRIT system that exploits DOTA-Bn (Bn=benzyl) has significant curative potential in a mouse xenograft model. As TIs improve by >10-fold, no clinical or histologic toxicities are observed. As theranostics, PRIT dosimetry using either PET or SPECT has yielded highly reproducible dose estimates. While radioisotopes provide initial proof of principle, PRIT may be applicable to any payloads linked to DOTA-Bn, including nanoparticles, peptides, toxins, drugs and viruses. The present inventors have applied their PRIT method to target the human A33 antigen because of its high mortality in the United States. For example, A33-positive tumors are involved in high mortality in colorectal cancer (49700 annual deaths), gastric cancer (10720 annual deaths), and pancreatic cancer (39590 annual deaths). Currently, no curative therapy is available for any of these metastatic cancers. As described herein, the present inventors have developed a bispecific antibody termed huA33-C825 using the variable region sequences of humanized antibody A33 (King et al., 1995, Brit. J. Cancer 72:1364-1372) and C825, a murine scFv antibody with high affinity for benzyl-1,4,7,10-tetraazacyclododecane-N,N′,N″,N′″-tetraacetic acid (DOTA-Bn)-radiometal complexes (Orcutt el al., 2011, Nucl. Med. Biol. 38:223-233), to demonstrate an improved PRIT methodology in mice bearing established s.c. SW1222 human colorectal carcinoma xenografts. We note that data provided herein specifically demonstrates that using such an improved PRIT as described herein provides tumor-to-normal tissues ratios of 105:1 (blood) and 18:1 (kidney) at 24 hours (h) post-injection (p.i.). Further, as described herein, biodistribution of177Lu-DOTA-Bn from 2-120 h p.i., estimated absorbed doses (cGy/MBq) to tumor, blood, liver, spleen, and kidney for PRIT were 65.8, 0.9 (therapeutic index (TI): 73), 6.3 (TI: 10), 6.6 (TI: 10), and 5.3 (TI: 12), respectively. Thus, in some embodiments, the PRIT regimen employing the huA33-C825 bispecific antibody described herein provides an improved therapeutic index and optimal tumor dose in the treatment of a human colorectal xenograft. We also note that data provided herein specifically demonstrates that dual-cycle PRIT treatment (66.6 or 111 MBq177Lu-DOTA-Bn, see Table 7) of established tumors produced 9/9 complete responses and 2/9 alive without recurrence at more than 140 d. Further, in the other 7, the time to reach tumor size of 500 mm3were 27±26 d for 66.6 MBq and 40±6 d for 111 MBq, compared to 13±2 d for non-treated mice. There were no clinical or histologic evidence of radiation induced toxicities. Thus, the data provided herein confirms that bispecific antibodies described herein represent cancer therapeutics characterized by improved efficacy and safety profiles, and a multi-step PRIT approach, as described herein, could deliver safe and effective radiation using the 0-emitting isotope177Lu to ablate established colorectal tumors. Humanized Antibodies In some embodiments, antibodies for use in accordance with the present invention are monoclonal antibodies, and/or in some embodiments may be humanized versions of cognate anti-A33 antibodies that were prepared in other species. In some embodiments, a humanized antibody is one which some or all of the amino acids of a human immunoglobulin light or heavy chain that are not required for antigen binding (e.g., the constant regions and the framework regions of the variable domains) are used to substitute for the corresponding amino acids from the light or heavy chain of a cognate, nonhuman antibody. By way of example, a humanized version of a murine antibody to a given antigen has on both of its heavy and light chains (1) constant regions of a human antibody; (2) framework regions from the variable domains of a human antibody; and (3) CDRs from the murine antibody. In some embodiments, one or more residues in the human framework regions can be changed to residues at the corresponding positions in the murine antibody so as to preserve the binding affinity of the humanized antibody to the antigen. Such a change is sometimes called “back mutation.” Similarly, forward mutations may be made to revert back to murine sequence for a desired reason, e.g. stability or affinity to antigen. Humanized antibodies generally are less likely to elicit an immune response in humans as compared to chimeric human antibodies because the former contain considerably fewer non-human components. In some embodiments, a humanized antibody is produced by recombinant DNA technology. Alternatively or additionally, suitable methods for making humanized antibodies of the present invention are described in, e.g., EP0239400; Jones et al., 1986, Nature 321:522-525; Riechmann et al., 1988, Nature 332:323-327; Verhoeyen et al., 1988, Science 239:1534-1536; Queen et al., 1989, Proc. Nat. Acad. Sci. U.S.A. 86:10029; U.S. Pat. No. 6,180,370; and Orlandi et al., 1989, Proc. Natl. Acad. Sci. U.S.A. 86:3833; the disclosures of all of which are incorporated by reference herein in their entireties. Generally, the transplantation of murine (or other non-human) CDRs onto a human antibody is achieved as follows. The cDNAs encoding heavy and light chain variable domains are isolated from a hybridoma. The DNA sequences of the variable domains, including the CDRs, are determined by sequencing. The DNAs, encoding the CDRs are inserted into the corresponding regions of a human antibody heavy or light chain variable domain coding sequences, attached to human constant region gene segments of a desired isotype (e.g., γ1 for CHand κ for CL), are gene synthesized. The humanized heavy and light chain genes are co-expressed in mammalian host cells (e.g., CHO or NSO cells) to produce soluble humanized antibody. To facilitate large-scale production of antibodies, it is often desirable to select for a high expressor using a DHFR gene or GS gene in the producer line. These producer cell lines are cultured in bioreactors, or hollow fiber culture system, or WAVE technology, to produce bulk cultures of soluble antibody, or to produce transgenic mammals (e.g., goats, cows, or sheep) that express the antibody in milk (see, e.g., U.S. Pat. No. 5,827,690). As described herein, multi-specific binding agents (e.g., bispecific antibodies) were engineered utilizing sequences and/or components found in the humanized antibody A33 described in King et al., 1995 (supra). Other murine anti-A33 antibodies may be humanized (e.g., as described herein) and may be employed in the engineering of multi-specific binding agents as described herein. For example, cDNAs encoding variable regions of light and/or heavy chains of one or more (typically only one) candidate murine anti-A33 antibody(ies) are used to construct vectors for expression of murine-human chimeras in which the murine anti-A33 antibody variable regions are linked to human IgG1 (for heavy chain) and human kappa (for light chain) constant regions, as described previously. Alternatively or additionally, in some embodiments, novel forms of humanized anti-A33 antibodies with variant glycosylation can be created, for example in order to enhance binding to the Fc receptor and enhance antigen affinity if so desired. In some embodiments, in order to produce humanized anti-A33 antibodies, human acceptor framework domains can be chosen by homology matching to human germline sequences. Using such chosen human acceptor frameworks, the light and heavy chain variable domains are designed and a number of variants/versions of each can be generated and expressed. Completely human antibodies are particularly desirable for therapeutic treatment of human patients. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also, U.S. Pat. Nos. 4,444,887 and 4,716,111; and International Patent Application Publications WO 98/46645, WO 98/60433, WO 98/24893, WO 98/16664, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirety. The techniques of Cole et al. (1985, Monoclonal Antibodies and Cancer Therapy, ed. R. A. Reisfeld & S. Sell, pp. 77-96, New York, Alan R. Liss) and Boerder et al. (1991) J. Immunol, 147(1):86-95), are also available for the preparation of human monoclonal antibodies. Human antibodies produced using other techniques but retaining the variable regions of the anti-A33 antibody of the present invention are included herein. Alternatively or additionally, human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous mouse immunoglobulins, but which can express human immunoglobulin genes (e.g., see Lonberg and Huszar, 1995, Int. Rev. Immunol. 13:65-93; Taylor, L. D., et al., 1992, Nucl. Acids Res. 20:6287-6295; Kellermann S-A., and Green L. L., 2002, Current Opinion in Biotechnology 13:593-597; Little M. et al., 2000, Immunol. Today 21:364-370; Murphy, A. J. et al., 2014, Proc. Natl. Acad. Sci. U.S.A. 111(14):5153-5158). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., International Patent Application Publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No. 0 598 877; U.S. Pat. Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825; 5,661,016; 5,545,806; 5,814,318; 5,886,793; 5,916,771; 5,939,598; and 8,502,018, which are incorporated by reference herein in their entirety. Still further, human monoclonal antibodies could be made by immunizing mice transplanted with human peripheral blood leukocytes, splenocytes or bone marrows (e.g., Trioma techniques of XTL). Completely human antibodies that recognize a selected epitope can be generated using a technique referred to as “guided selection.” In this approach a selected non-human monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope (Jespers et al., 1988, Biotechnol. 12:899-903). As used herein, an “anti-A33 antibody”, “anti-A33 antibody portion,” or “anti-A33 antibody fragment” and/or “anti-A33 antibody variant” and the like may, in some embodiments, refer to a polypeptide-containing entity that comprises at least a portion of an immunoglobulin that binds to A33, and in particular refers to an entity including a polypeptide that at least one complementarity determining region (CDR) of a heavy or light chain or a ligand binding portion thereof (and typically containing all CDRs found in a relevant chain or portion thereof) found in any of the particular monoclonal antibodies described herein that to A33. In some embodiments, the term refers to an entity that includes such a polypeptide that includes not only such CDRs, but also other sequences found in a heavy chain or light chain variable region, a heavy chain or light chain constant region, a framework region, or any portion thereof, of non-murine origin, preferably of human origin, which can be incorporated into an antibody of the present invention. In some particular embodiments, the term “anti-A33 antibody”, as will be clear from context, is used to refer collectively or individually to huA33, hA33, A33, humanized antibody A33, humanized A33, and combinations thereof, and/or relevant fragments or components, domains, or regions thereof, such as single chain variable fragments (e.g., huA33 scFv, hA33 scFv, A33 scFv, and combinations thereof). In some embodiments, a humanized antibody is capable of modulating, decreasing, antagonizing, mitigating, alleviating, blocking, inhibiting, abrogating and/or interfering with at least one cell function in vitro, in situ and/or in vivo, wherein said cell expresses human A33. As a non-limiting example, a suitable anti-A33 antibody, specified portion or variant binds with high affinity to an epitope, in particular a peptide epitope, of human A33. Antibody fragments can be produced by enzymatic cleavage, synthetic or recombinant techniques, as known in the art and/or as described herein. Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site. For example, a combination gene encoding a F(ab′)2 heavy chain portion can be designed to include DNA sequences encoding the CH1 domain and/or hinge region of the heavy chain. Various portions of antibodies can be joined together chemically by conventional techniques, or can be prepared as a contiguous protein using genetic engineering techniques. In some embodiments, chimeric or humanized antibodies for use in accordance with the present invention include those wherein the CDRs are found in one or more of the anti-A33 antibodies described herein and at least a portion, or the remainder of the antibody is found in or derived from one or more human antibodies. Thus, for example, in some embodiments, the human part of the antibody may include the framework, CL, CHdomains (e.g., CH1, CH2, CH3), hinge, VL, VHregions which are substantially non-immunogenic in humans. Those skilled in the art, reading the present disclosure, will appreciate that, in some embodiments, a “human part” of an antibody utilized as described herein, may in some embodiment may not show 100% identity with the corresponding sequence found in a relevant source human antibody. In some embodiments, as many of the human amino acid residues as possible found in the source human antibody are retained in order for the immunogenicity to be negligible, however, in various embodiments, the human residues may be modified as necessary or otherwise desired to support the antigen binding site formed by the CDRs while simultaneously maximizing the humanization of the antibody. Such changes or variations, in some embodiments, retain or reduce the immunogenicity in humans or other species relative to non-modified antibodies. Those of ordinary skill in the art, reading the present disclosure, will appreciate that an antibody agent provided by the present invention, including one that is a humanized antibody and/or that utilizes a humanized antibody sequence elements as described herein, can be produced by a non-human animal or prokaryotic or eukaryotic cell that is capable of expressing functionally rearranged human immunoglobulin (e.g., heavy chain and/or light chain) genes. Further, when the antibody agent is a single chain antibody, it can comprise a linker peptide that is not found in native human antibodies. For example, an Fv can comprise a linker peptide, such as two to about twenty glycine or other amino acid residues, preferably 8-15 glycine or other amino acid residues, which connects the variable region of the heavy chain and the variable region of the light chain. Such linker peptides are considered to be of human origin. Antibody humanization can be performed by, for example, by synthesizing a combinatorial library comprising the six CDRs of a non-human target monoclonal antibody fused in frame to a pool of individual human frameworks. A human framework library that contains genes representative of all known heavy and light chain human germline genes can be utilized. Resulting combinatorial libraries can be screened for binding to antigens of interest. Such an approach can allow for screening and/or selection of particularly favorable (e.g., in terms of maintaining the binding activity to the parental antibody) combinations of fully human frameworks. Humanized antibodies can then be further optimized by a variety of techniques. Antibody humanization can be used to evolve mouse or other non-human antibodies into “fully human” antibodies. Resulting antibody(ies) may contain only human sequence and no mouse or non-human antibody sequence, while maintaining similar binding affinity and specificity as the starting antibody. In some embodiments, anti-A33 humanized antibodies for use in accordance with the present invention comprise a variant Fc region, wherein said variant Fc region comprises at least one amino acid modification relative to a wild-type Fc region (or the parental Fc region), such that said molecule has an altered affinity for an Fc receptor (e.g., an FcγR), provided that said variant Fc region does not have a substitution at positions that make a direct contact with Fc receptor based on crystallographic and structural analysis of Fc-Fc receptor interactions such as those disclosed by Sondermann et al. (2000, Nature, 406:267-273, which is incorporated herein by reference in its entirety). Examples of positions within the Fc region that make a direct contact with an Fc receptor such as an FcγR are amino acids 234-239 (hinge region), amino acids 265-269 (B/C loop), amino acids 297-299 (C′/E loop), and amino acids 327-332 (F/G) loop. In some embodiments, the anti-A33 antibodies of the present invention comprising variant Fc regions comprise modification of at least one residue that makes a direct contact with an FcγR based on structural and crystallographic analysis. In some embodiments, an anti-A33 antibody for use in accordance with the present invention is a humanized A33 antibody with an altered affinity for activating and/or inhibitory receptors, having variant Fc regions with one or more amino acid modifications, wherein said one or more amino acid modification is a substitution at position 297 with alanine; in some embodiments, a substitution at 239D, 330L, 332E to enhance FcR affinity; in some embodiments, a substitution at 322K to reduce or eliminate FcR binding. In some embodiments, anti-A33 antibodies for use in accordance with the present invention have an Fc region with variant glycosylation as compared to a parent Fc region; in some embodiments, variant glycosylation includes absence of fucose; in some embodiments, variant glycosylation results from expression in GnT1-deficient CHO cells. In some embodiments, the present invention provides bispecific binding agents having a humanized A33 antibody component that comprises a variant Fc region characterized by a K322A substitution. In some embodiments a provided bispecific binding agent includes an antibody component that shows variant glycosylation (e.g., is aglycosylated) as compared with a parent antibody from which the component may be derived; in some such embodiments, such a variant may be or comprise a variant Fc region characterized by the K322A substitution. In some embodiments, such variant components (e.g., variant Fc regions) result in a complete elimination of complement activation and FcR binding, which otherwise may damage tumor cell membrane prior to addition of a clearing agent in pre-targeted radioimmunotherapy as described herein. In some embodiments, the present invention provides and/or utilizes antibodies or antibody agents comprising a variant Fc region (i.e., an Fc region includes one or more additions, deletions, and/or substitutions relative to an appropriate reference Fc) that is characterized in that its alter effector function altered and/or its affinity for an FcR is enhanced or diminished relative to the reference Fc. These variations are within the skill of a person in the art. Therefore, among other things, the present invention provides multi-specific binding agents (e.g., antibody agents) comprising variant Fc regions that bind with a greater affinity to one or more FcγRs. Such agents preferably mediate effector function more effectively as discussed infra. In some embodiments, the present invention provides multi-specific binding agents (e.g., antibody agents) comprising a variant Fc region that bind with a weaker affinity to one or more FcγRs. Reduction or elimination of effector function is desirable in certain cases for example in the case of antibodies whose mechanism of action involves blocking or antagonism but not killing of the cells bearing a target antigen. Further, elimination of effector function is desirable, in some embodiments, when making bispecific antibodies as discussed infra. Reduction or elimination of effector function would be desirable in cases of autoimmune disease where one would block FcγR activating receptors in effector cells (This type of function would be present in the host cells). Generally, increased effector function may be directed to tumor and foreign cells; in some embodiments, effector function may be directed away from tumor cells. Fc variants for use in accordance present invention may be combined with other Fc modifications, including but not limited to modifications that alter effector function. The invention encompasses combining an Fc variant as described herein with other Fc modifications to provide additive, synergistic, or novel properties in antibodies or Fc fusions. In some such embodiments, Fc variants may enhance the phenotype of the modification with which they are combined. For example, if an Fc variant is combined with a mutant known to bind FcγRIIIA with a higher affinity than a comparable molecule comprising a wild type Fc region, the combination with the mutant results in a greater fold enhancement in FcγRIIIA affinity. In some embodiments, in accordance with the present invention Fc variants as described herein are incorporated into an antibody or Fc fusion to generate an engineered agent that comprises one or more Fc glycoforms (i.e., one or more Fc polypeptides to which one or more carbohydrates is covalently attached) to a molecule comprising an Fc region wherein the carbohydrate composition of the glycoform differs chemically from that of a parent molecule comprising an Fc region. In some embodiments, a multi-specific binding agent (e.g., an antibody agent) as described herein may include an Fc variant that shows variant glycosylation and/or may be expressed in a glycosylation deficient cell line (e.g., a GnT1-deficient CHO cell) such an Fc region of the agent is produced lacking glycosylation as compared to an appropriate reference Fc region (e.g., a wild type), or an Fc region expressed in a cell line not deficient in glycosylation. In some embodiments, antibodies utilized in accordance with the present invention, may have a modified glycosylation site relative to an appropriate reference antibody that binds to an antigen of interest (e.g., A33), preferably without altering the functionality of the antibody, e.g., binding activity to the antigen. As used herein, “glycosylation sites” include any specific amino acid sequence in an antibody to which an oligosaccharide (i.e., carbohydrates containing two or more simple sugars linked together) will specifically and covalently attach. Oligosaccharide side chains are typically linked to the backbone of an antibody via either N- or O-linkages. N-linked glycosylation refers to the attachment of an oligosaccharide moiety to the side chain of an asparagine residue. O-linked glycosylation refers to the attachment of an oligosaccharide moiety to a hydroxyamino acid, e.g., serine, threonine. For example, an Fc-glycoform (huA33-IgG1n) that lacks certain oligosaccharides including fucose and terminal N-acetylglucosamine may be produced in special CHO cells and exhibit enhanced ADCC effector function. In some embodiments, the present invention encompasses methods of modifying the carbohydrate content of an antibody of the invention by adding or deleting a glycosylation site. Methods for modifying the carbohydrate content of antibodies are well known in the art and are included within the present invention, see, e.g., U.S. Pat. No. 6,218,149; EP0359096B1; U.S. Patent Publication No. US 2002/0028486; International Patent Application Publication WO 03/035835; U.S. Patent Publication No. 2003/0115614; U.S. Pat. Nos. 6,218,149; 6,472,511; all of which are incorporated herein by reference in their entirety. In some embodiments, the present invention includes methods of modifying the carbohydrate content of an antibody (or relevant portion or component thereof) by deleting one or more endogenous carbohydrate moieties of the antibody. In some certain embodiments, the present invention includes deleting the glycosylation site of the Fc region of an antibody, by modifying position 297 from asparagine to alanine. Engineered glycoforms may be useful for a variety of purposes, including but not limited to enhancing or reducing effector function. Engineered glycoforms may be generated by any method known to one skilled in the art, for example by using engineered or variant expression strains, by co-expression with one or more enzymes, for example DI N-acetylglucosaminyltransferase III (GnTIII), by expressing a molecule comprising an Fc region in various organisms or cell lines from various organisms, or by modifying carbohydrate(s) after the molecule comprising Fc region has been expressed. Methods for generating engineered glycoforms are known in the art, and include but are not limited to those described in Umana et al., 1999, Nat. Biotechnol. 17:176-180; Davies et al., 2001, Biotechnol. Bioeng. 74:288-294; Shields et al., 2002, J. Biol. Chem. 277:26733-26740; Shinkawa et al., 2003, J. Biol. Chem. 278:3466-3473; U.S. Pat. No. 6,602,684; U.S. patent application Ser. No. 10/277,370; U.S. patent application Ser. No. 10/113,929; International Patent Application Publications WO 00/61739A1; WO 01/292246A1; WO 02/311140A1; WO 02/30954A1; POTILLEGENT™ technology (Biowa, Inc. Princeton, N.J.); GLYCOMAB™ glycosylation engineering technology (GLYCART biotechnology AG, Zurich, Switzerland); each of which is incorporated herein by reference in its entirety. See, e.g., International Patent Application Publication WO 00/061739; EA01229125; U.S. Patent Application Publication No. 2003/0115614; Okazaki et al., 2004, JMB, 336:1239-49, each of which is incorporated herein by reference in its entirety. Multivalent Binding Agents As those skilled in the art are aware, a multivalent binding agent is a molecular entity or complex that includes binding components that bind specifically to two or more targets (e.g., epitopes). Such multivalent binding agents find a variety of uses in the art, including therapeutic uses. To give but one example, as those skilled in the art are aware, multivalent binding agents have been engineered to facilitate killing of tumor cells by directing (or recruiting) cytotoxic T cells to a tumor site. Examples of tumor antigens include, but are not limited to, alpha fetoprotein (AFP), CA15-3, CA27-29, CA19-9, CA-125, calretinin, carcinoembryonic antigen, CD34, CD99, CD117, chromogranin, cytokeratin, desmin, epithelial membrane protein (EMA), Factor VIII, CD31 FL1, glial fibrillary acidic protein (GFAP), gross cystic disease fluid protein (GCDFP-15), HMB-45, human chorionic gonadotropin (hCG), inhibin, keratin, CD45, a lymphocyte marker, MART-1 (Melan-A), Myo Dl, muscle-specific actin (MSA), neurofilament, neuron-specific enolase (NSE), placental alkaline phosphatase (PLAP), prostate-specific antigen, S100 protein, smooth muscle actin (SMA), synaptophysin, thyroglobulin, thyroid transcription factor-1, tumor M2-PK, and vimentin. In some embodiments, multivalent binding agents for use in accordance with the present invention are bispecific binding agents. In many embodiments, such bispecific binding agents are capable of binding to tumor cells. In many embodiments, such bispecific binding agents are capable of binding to human colorectal cancer cells via an A33 antigen expressed in the tumor cell surface. In some embodiments, multivalent binding agents (e.g., bispecific binding agents) provided by the present invention are or comprise antibody components. A variety of technologies are known in the art for designing, constructing, and/or producing multispecific binding agents comprising antibody components. For example, multivalent binding agents have been constructed that either utilize the full immunoglobulin framework (e.g., IgG), single chain variable fragment (scFv), or combinations thereof. Bispecific binding agents composed of two scFv units in tandem has been shown to be a clinically successful bispecific antibody format. In the case of anti-tumor immunotherapy, bispecific binding agents that comprise two single chain variable fragments (scFvs) in tandem have been designed such that an scFv that binds a tumor antigen is linked with an scFv that engages T cells by binding CD3. In this way, T cells are recruited to a tumor site in the hope that they can mediate killing of the tumor cells making up the tumor by the cytotoxic properties that certain T cells have. An example of such a bispecific binding agent has been made that targets CD19 and CD3 for lymphoma (termed Bispecific T cell Engaging, or BiTE; e.g., see Dreier et al., 2003, J. Immunol. 170:4397-4402; Bargou et al., 2008, Science 321:974-977), which has been successful in preventing tumor growth in animal xenograft studies. In human studies, this bispecific binding agent demonstrated objective tumor response, including five partial and two complete remissions. Exemplary bispecific binding agents include those with a first antibody component specific for a tumor antigen and a second antibody component specific for a small molecule hapten (e.g., DTPA, IMP288, DOTA, DOTA-Bn, DOTA-desferrioxamine, Biotin, fluorescein, or those disclosed in Goodwin, D. A. et al., 1994, Cancer Res. 54(22):5937-5946, herein incorporated by reference). Bispecific binding agents can be made, for example, by combining heavy chains and/or light chains that recognize different epitopes of the same or different antigen. In some embodiments, by molecular function, a bispecific binding agent binds one antigen (or epitope) on one of its two binding arms (one VH/VLpair), and binds a different antigen (or epitope) on its second arm (a different VH/VLpair). By this definition, a bispecific binding agent has two distinct antigen binding arms (in both specificity and CDR sequences), and is monovalent for each antigen to which it binds. In some embodiments, bispecific binding agents of the present invention are characterized by the ability to bind simultaneously to two targets that are of different structure. In some embodiments, bispecific binding agents of the present invention have at least one component that specifically binds to, for example, a B-cell, T-cell, myeloid, plasma, or a mast cell antigen or epitope and at least one other component that specifically binds to a targetable conjugate that bears a therapeutic or diagnostic agent. Bispecific binding agents (e.g., bispecific antibodies) of the present invention are based on the particular insight that certain formats may be more beneficial for certain targets (e.g., a tumor antigen) when employed in multi-step pretargeted radioimmunotherapy (PRIT) methodology that targets human A33 antigen. For example, bispecific antibodies provided herein utilize a combination of a full IgG and an scFv. Such bispecific antibodies demonstrate bivalent binding via the IgG component (e.g., anti-A33) and bivalent binding via the scFv component (e.g., anti-DOTA-Bn). As described herein, bispecific antibodies having this format first bind to an A33-positive tumor cell via the IgG component (e.g., anti-A33) and excess antibody is cleared from the blood via a clearing agent (CA; e.g., a dextran-based clearing agent). This is followed by a step that includes the use of a radiolabeled small molecule hapten (e.g.,177Lu-DOTA-Bn). Exemplary radiolabeled small molecules include radiolanthanides, e.g., yttrium and lutetium (e.g.,86Y,90Y and177Lu) as well as124I and131I. Further, bispecific antibodies of the present invention provide both diagnostic and therapeutic tumor targeting features. In various embodiments, a bispecific binding agent (e.g., a bispecific antibody) according to the present invention is composed of a first binding component and a second binding component. In many embodiments, first and second binding components of a bispecific binding agent as described herein are each composed of antibody components characterized by different specificities. In many embodiments, antibody components are selected from Table 8. In various embodiments, a bispecific binding agent according to the present invention comprises a first binding component, a second binding component. In various embodiments, a bispecific binding agent according to the present invention comprises a first binding component, a second binding component and a linker that is connected to both the first and second binding component (e.g., positioned between the first and second binding components). In various embodiments, first and/or second binding components as described herein comprise or are antibody components. In various embodiments, first and/or second binding components as described herein comprise a linker sequence. In some embodiments, a linker is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more amino acids in length. In some embodiments, a linker is characterized in that it tends not to adopt a rigid three-dimensional structure, but rather provides flexibility to the polypeptide (e.g., first and/or second binding components). In some embodiments, a linker is employed in a bispecific binding agent described herein based on specific properties imparted to the bispecific binding agent such as, for example, a reduction in aggregation and/or an increase in stability. In some embodiments, a bispecific binding agent of the present invention comprises a G4S linker. In some certain embodiments, a bispecific binding agent of the present invention comprises a (GIS), linker, wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more. In various embodiments, first and/or second binding components as described herein comprise or are immunoglobulins (e.g., IgGs). In various embodiments, first and/or second binding components binding components as described herein comprise or are antibody fragments (e.g., scFvs). In various embodiments, first binding components as described herein comprise or are immunoglobulins and second binding components comprise or are antibody fragments. In some certain embodiments, first binding components are immunoglobulins and second binding components are antibody fragments. In some certain embodiments, first binding components are IgGs and second binding components are scFvs. In some certain embodiments, a bispecific binding agent according to the present invention comprises an immunoglobulin, which immunoglobulin comprises a heavy chain and a light chain, and an scFv. In some certain embodiments, scFvs are linked to the C-terminal end of the heavy chain of the immunoglobulin. In some certain embodiments, scFvs are linked to the C-terminal end of the light chain of the immunoglobulin. In various embodiments, scFvs are linked to heavy or light chains via a linker sequence. In some embodiments, a bispecific binding agent of the present invention comprises one or more sequences that are at least about 50% (e.g., at least about 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%) identical to one or more sequences that appear in Table 8. In some embodiments, a bispecific binding agent of the present invention comprises one or more sequences that are substantially identical to one or more sequences that appears in Table 8. In some embodiments, a bispecific binding agent of the present invention comprises one or more sequences that are identical to one or more sequences that appears in Table 8. In some embodiments, a bispecific binding agent of the present invention is selected from one or more sequences that appear in Table 8. In some certain embodiments, a bispecific binding agent of the present invention is selected from two sequences that appear in Table 8, for example, a heavy chain and a light chain sequence. In various embodiments, a first binding component of a bispecific binding agent as described herein comprises an antibody component having a sequence at least 50% (e.g., 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more) identical to an antibody component that appears in Table 8. In various embodiments, a first binding component of a bispecific binding agent as described herein comprises an antibody component having a sequence that is substantially identical to an antibody component that appears in Table 8. In various embodiments, a first binding component of a bispecific binding agent as described herein comprises an antibody component having a sequence that is identical to an antibody component that appears in Table 8. In various embodiments, a second binding component of a bispecific binding agent as described herein comprises an antibody component having a sequence at least 50% (e.g., 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more) identical to an antibody component that appears in Table 8. In various embodiments, a second binding component of a bispecific binding agent as described herein comprises an antibody component having a sequence that is substantially identical to an antibody component that appears in Table 8. In various embodiments, a second binding component of a bispecific binding agent as described herein comprises an antibody component having a sequence that is identical to an antibody component that appears in Table 8. As described herein, the present inventors provide an improved multi-step PRIT method in immunocompromised mice bearing s.c. A33-positive human colorectal tumors (SW1222) using a bispecific antibody termed huA33-C825. Such methodology includes i.v. injection of various doses of huA33-C825 and a dextran-based CA, followed with injection of177Lu-DOTA-Bn theranostic (i.e., diagnostic and therapeutic) hapten for simultaneous scintigraphic imaging and radioimmunotherapy. The present invention specifically describes biodistribution studies that provide optimum huA33-C825 and CA doses, followed with a series of additional biodistribution studies to determine the tumor uptake as a function of177Lu-DOTA-Bn doses (˜2.0-111.0 MBq) to serve as a practical and dosimetric guide for PRIT studies. Also, as described herein, tumors and kidneys were excised at 24 hours post injection of177Lu-DOTA-Bn at three different dose levels (11.1, 55.0, and 111.0 MBq) to examine ex vivo the177Lu-activity microdistribution via autoradiography, as well as correlate the177Lu-activity with the xenograft and tissue morphology via hematoxylin and eosin staining. Further, the estimated absolute SW1222 tumor uptake of huA33-C825 24 h p.i. of 0.25 mg/mouse based on radioactive tracer studies with131I-huA33-C825 was ˜90 pmol/g of tumor. Therefore, the present invention demonstrates that if a single huA33-C825 molecule has the capacity to bind two molecules of177Lu-DOTA-Bn (thus maximum177Lu-DOTA-Bn binding capacity of 180 pmol/g tumor), the estimated maximum occupancy is (11 pmol177Lu-DOTA-Bn/180 pmol=0.061 or −6%). Thus, the present invention specifically demonstrates that the improved PRIT method was effective without any associated adverse radiation response. Further, the present invention specifically demonstrates that, at least in some embodiments, immunocompromised mice with established s.c. human colorectal xenografts of could be cured with minimal toxicity to normal tissues including bone marrow and kidney using an improved multi-step PRIT method employing an anti-A33/anti-DOTA-Bn (metal) bispecific antibody termed huA33-C825, a dextran-based CA, and177Lu-DOTA-Bn. SW1222 stands out among commonly investigated human colorectal carcinomas (e.g., LS174T) as a relatively well-differentiated and vascularized tumor. While permitting homogeneous distribution of targeted antibodies (Emir et al., 2007, Cancer Res. 15; 67(24):11896-11905), these tumors are also relatively radioresistant. As described herein, the A33 antigen is highly expressed in more than 95% of human colon cancers with restricted normal expression and minimal shedding into circulation. There has been no successful clinical therapeutic targeting of the A33 antigen in human colorectal cancer. Bispecific antibodies as described herein demonstrate affinity to A33 and a DOTA-Bn (metal), which facilitates tumor uptake of a radiolabeled lutetium (e.g.,177Lu) and successful delivery of targeted radioimmunotherapy to A33-positive tumors. Also, bispecific binding proteins employing humanized A33 antibodies as described herein are capable of bivalent binding to A33 and bivalent binding to DOTA-Bn which results in enhanced potency for killing A33+tumors and increased safety from a lack of catastrophic radiation response. As such, the PRIT strategy employing the format of the bispecific binding proteins described herein represents a unique approach for enhanced tumor killing, reduced adverse effects, and demonstrates a potent therapeutic for the treatment of several A33-positive cancers. Targets Among other things, the present invention encompasses the recognition that multispecific binding agents, and particularly bispecific binding agents such as bispecific antibodies, are particularly useful and/or effective to facilitate cell killing. In particular, the present invention demonstrates that activity of multivalent binding agents that bind specifically to both a target-cell-associated epitope (e.g., a tumor antigen) and a small molecule hapten (e.g., a DOTA-Bn [metal]) can be an effective immunotherapy for colon cancers. For example, in some embodiments of the present invention, a multivalent binding agent binds specifically to a tumor-cell-associated epitope and a small molecule hapten. In accordance with such embodiments, the multivalent binding agent can facilitate binding of the agent to one or both of its target epitopes and/or can enhance killing of the target tumor cell as mediated by radioimmunotherapy via the small molecule hapten. In some embodiments, target cells to be killed include, for example, cells that express a tumor antigen (e.g., a A33-positive tumor). Those of ordinary skill in the art will be aware of appropriate target epitopes on such cells to which multivalent binding agents as described herein desirably bind. Nucleic Acid Construction and Expression Humanized antibodies and multispecific binding agents (e.g., bispecific antibodies) as described herein may be produced from nucleic acid molecules using molecular biological methods known to the art. Nucleic acid molecules are inserted into a vector that is able to express the fusion proteins in when introduced into an appropriate host cell. Appropriate host cells include, but are not limited to, bacterial, yeast, insect, and mammalian cells. Any of the methods known to one skilled in the art for the insertion of DNA fragments into a vector may be used to construct expression vectors encoding the fusion proteins of the present invention under control of transcriptional/translational control signals. These methods may include in vitro recombinant DNA and synthetic techniques and in vivo recombination (See Sambrook et al. Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory; Current Protocols in Molecular Biology, Eds. Ausubel, et al, Greene Publ. Assoc., Wiley-Interscience, NY). Expression of nucleic acid molecules in accordance with the present invention may be regulated by a second nucleic acid sequence so that the molecule is expressed in a host transformed with the recombinant DNA molecule. For example, expression of the nucleic acid molecules of the invention may be controlled by a promoter and/or enhancer element that are known in the art. Nucleic acid constructs include regions that encode multispecific binding proteins generated from antibodies and/or antibody components. Typically, such multispecific binding proteins will be generated from VHand/or VLregions. After identification and selection of antibodies exhibiting desired binding and/or functional properties, variable regions of each antibody are isolated, amplified, cloned and sequenced. Modifications may be made to the VHand VLnucleotide sequences, including additions of nucleotide sequences encoding amino acids and/or carrying restriction sites, deletions of nucleotide sequences encoding amino acids, or substitutions of nucleotide sequences encoding amino acids. The antibodies and/or antibody components may be generated from human, humanized or chimeric antibodies. Nucleic acid constructs of the present invention are inserted into an expression vector or viral vector by methods known to the art, and nucleic acid molecules are operatively linked to an expression control sequence. Where appropriate, nucleic acid sequences that encode humanized antibodies and multi-specific binding agents as described herein may be modified to include codons that are optimized for expression in a particular cell type or organism (e.g., see U.S. Pat. Nos. 5,670,356 and 5,874,304). Codon optimized sequences are synthetic sequences, and preferably encode the identical polypeptide (or a biologically active fragment of a full length polypeptide which has substantially the same activity as the full length polypeptide) encoded by the non-codon optimized parent polynucleotide. In some embodiments, the coding region of the genetic material encoding antibody components, in whole or in part, may include an altered sequence to optimize codon usage for a particular cell type (e.g., a eukaryotic or prokaryotic cell). For example, the coding sequence for a humanized heavy (or light) chain variable region as described herein may be optimized for expression in a bacterial cells. Alternatively, the coding sequence may be optimized for expression in a mammalian cell (e.g., a CHO). Such a sequence may be described as a codon-optimized sequence. An expression vector containing a nucleic acid molecule is transformed into a suitable host cell to allow for production of the protein encoded by the nucleic acid constructs. Exemplary host cells include prokaryotes (e.g.,E. coli) and eukaryotes (e.g., a COS or CHO cell). Host cells transformed with an expression vector are grown under conditions permitting production of a humanized antibody or multispecific binding agent of the present invention followed by recovery of the humanized antibody or multispecific binding agent. Humanized antibodies and/or multispecific binding agents of the present invention may be purified by any technique, which allows for the subsequent formation of a stable antibody or binding agent molecule. For example, not wishing to be bound by theory, antibodies and/or multispecific binding agents may be recovered from cells either as soluble polypeptides or as inclusion bodies, from which they may be extracted quantitatively by 8M guanidinium hydrochloride and dialysis. In order to further purify antibodies and/or multispecific binding agents of the present invention, conventional ion exchange chromatography, hydrophobic interaction chromatography, reverse phase chromatography or gel filtration may be used. Humanized antibodies and/or multispecific binding agents of the present invention may also be recovered from conditioned media following secretion from eukaryotic or prokaryotic cells. Screening and Detection Methods Humanized antibodies and/or multispecific binding agents of the present invention may also be used in in vitro or in vivo screening methods where it is desirable to detect and/or measure one or more activities of a cell or cells (e.g., apoptosis or cell growth). Screening methods are well known to the art and include cell-free, cell-based, and animal assays. In vitro assays can be either solid state or soluble target molecule detection may be achieved in a number of ways known to the art, including the use of a label or detectable group capable of identifying a humanized antibody or a multispecific binding agent which is bound to a target molecule (e.g., cell surface antigen). Detectable labels may be used in conjunction with assays using humanized antibodies or multispecific binding agents of the present invention. Therapeutic Agents Humanized antibodies and/or multivalent binding agents of the present invention may be utilized as therapeutic agents. In some embodiments, as will be understood in the art, they are utilized without further modification. In some embodiments, they may be incorporated into a composition or formulation as described herein. In some embodiments, they may be chemically associated or linked (e.g., conjugated) with one or more other agents or entities, e.g., with a payload. A variety of technologies for conjugating antibody agents, or components thereof, with other moieties or entities are well known in the art and may be utilized in accordance with the practice of the present invention. To give but one example, radioactively-labeled antibody agents may be produced according to well-known technologies in the art. For instance, monoclonal antibodies can be iodinated by contact with sodium and/or potassium iodide and a chemical oxidizing agent such as sodium hypochlorite, or an enzymatic oxidizing agent, such as lactoperoxidase. Antibody agents may be labeled with technetium-99m by ligand exchange process, for example, by reducing pertechnate with stannous solution, chelating the reduced technetium onto a Sephadex column and applying the antibody to this column. In some embodiments, provided antibody agents are labeled using direct labeling techniques, e.g., by incubating pertechnate, a reducing agent such as SNC12, a buffer solution such as sodium-potassium phthalate solution, and the antibody. Intermediary functional groups which are often used to bind radioisotopes which exist as metallic ions to antibody are diethylenetriaminepentaacetic acid (DTPA), or ethylene diaminetetracetic acid (EDTA), or 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA), or p-aminobenzyl-DOTA (DOTA-Bn). Radioactive isotopes may be detected by, for example, dosimetry. Therapeutic Methods The ability of humanized antibodies and/or multi-specific binding agents of the present invention to exhibit high affinity binding for one of the target antigens makes them therapeutically useful for efficiently targeting cells expressing the target antigen. Thus, it some embodiments, it may be desirable to increase the affinity of a humanized antibody or multi-specific binding agent for one target antigen and not the other target antigen that is also bound by the multi-specific binding agent (or an Fc receptor in the case of a humanized antibody). For example, in the context of tumor killing, certain conditions may benefit from an increase or decrease in affinity to a tumor antigen but not to a second antigen. Thus, it may be beneficial to increase the binding affinity of a humanized antibody or multi-specific binding agent to a tumor antigen in a patient having a tumor that expresses the tumor antigen through the use of a humanized antibody or multi-specific binding agent as described herein. The present invention provides a humanized antibody and/or multi-specific binding agent as described herein as a therapeutic for the treatment of patients having a tumor that expresses an antigen that is capable of being bound by such a multi-specific binding agent. Such humanized antibodies and/or multi-specific binding agents may be used in a method of treatment of the human or animal body, or in a method of diagnosis. Administration The present invention provides methods of administering an effective amount of a therapeutic active described herein (e.g., a humanized antibody or multi-specific binding agent) to a subject in need of treatment. Humanized antibodies or multi-specific binding agents as described herein may be administered through various methods known in the art for the therapeutic delivery of agents, such as proteins or nucleic acids can be used for the therapeutic delivery of a humanized antibody or multi-specific binding agent or a nucleic acid encoding a humanized antibody or multi-specific binding agent of the present invention for killing or inhibiting growth of target cells in a subject, e.g., cellular transfection, gene therapy, direct administration with a delivery vehicle or pharmaceutically acceptable carrier, indirect delivery by providing recombinant cells comprising a nucleic acid encoding a multi-specific binding agent of the present invention. Various delivery systems are known and can be used to administer a humanized antibody or multi-specific binding agent of the present invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), construction of a nucleic acid as part of a retroviral or other vector, etc. Routes of administration can be enteral or parenteral and include, but are not limited to, intravenous, subcutaneous, intramuscular, parenteral, transdermal, or transmucosal (e.g., oral or nasal). In some embodiments, multi-specific binding agents of the present invention are administered intravenously. In some embodiments, multispecific binding agents of the present invention are administered subcutaneously. In some embodiments, multi-specific binding agents are administered together with other biologically active agents. Those of ordinary skill in the art, reading the present disclosure, will readily appreciate that therapy with a therapeutic active described herein (e.g., with a humanized antibody or multi-specific binding agent), as described herein, may in certain embodiments be combined with other therapies, and particularly including other anti-tumor therapies. In some embodiments, such other anti-tumor therapies may be or comprise, for example administration of one or more chemotherapeutic agents, immunomodulatory agents, radiation therapy, high-frequency ultrasound therapy, surgery, etc. In some embodiments, relative timing of administration of a therapeutic active described herein (e.g., a humanized antibody or multi-specific binding agent) and another therapy with which it is combined may be selected to optimize effect. To give but a few examples, in some embodiments, a therapeutic active as described herein is administered under conditions and for a period of time (e.g., according to a dosing regimen) sufficient for it to saturate tumor cells. In some embodiments, unbound therapeutic active is removed from the blood stream after administration; in some such embodiments, such removal occurs (e.g., is permitted to occur) prior to administration of another agent. In some particular embodiments, a therapeutic active as described herein is administered in combination with another agent that targets DOTA-Bn. In some such embodiments, the another agent carries a payload. In some embodiments, the payload may be or comprise a therapeutic agent payload (e.g., a toxic payload). In some embodiments the payload may be or comprise a detection agent payload. In some particular embodiments, a therapeutic active described herein (e.g., a humanized antibody or multi-specific binding agent) as described herein is administered so that tumor cells are saturated, and subsequently a second agent, that targets DOTA-Bn (and may carry a payload) is administered. Optionally, at least one third agent that targets DOTA-Bn (e.g., and may carry a different payload) is administered. In some embodiments, second and, optionally, third agents are administered a period of time after administration of a therapeutic active described herein, which period of time may be sufficient to permit clearance of unbound therapeutic agent. In some embodiments, second and, optionally third agents are administered without further administration of the therapeutic agent. For example, in some embodiments, a therapeutic active as described herein is administered according to a regimen that includes at least one cycle of: (i) administration of the therapeutic agent (optionally so that relevant tumor cells are saturated); (ii) administration of a second and, optionally at least one third agent (e.g., that targets DOTA-Bn, and may optionally carry a payload); (iii) optional additional administration of the second and/or third agents, without additional administration of the therapeutic agent. In some embodiments, a therapeutic regimen may comprise multiple such cycles; in some embodiments, a regimen may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more cycles. In some embodiments, a therapeutic regimen comprises only a single cycle that includes administration of the therapeutic agent; in some embodiments such a therapeutic regimen may comprise one or more cycles that include steps (ii) and, optionally, (iii) but do not include additional administrations of the therapeutic agent. In some embodiments, prior administration of a therapeutic agent as described herein permits combination therapy in which the agent with which the therapeutic agent is combined shows a broader therapeutic index than it does when administered alone (i.e., without the prior administration of a therapeutic agent as described herein). In some embodiments, such a broader therapeutic index is at least a log fold improved. Pharmaceutical Compositions The present invention further provides pharmaceutical compositions comprising humanized antibodies or multi-specific binding agents of the present invention and a pharmaceutically acceptable carrier or excipient. The composition, if desired, can also contain one or more additional therapeutically active substances. Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions that are suitable for ethical administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with a diluent or another excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit. A pharmaceutical composition in accordance with the present invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a “unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient that would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage. Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100% (w/w) active ingredient. Pharmaceutical formulations may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro (Lippincott, Williams & Wilkins, Baltimore, Md., 2006; incorporated herein by reference) discloses various excipients used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Except insofar as any conventional excipient medium is incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention. In some embodiments, a pharmaceutically acceptable excipient is at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure. In some embodiments, an excipient is approved for use in humans and for veterinary use. In some embodiments, an excipient is approved by the United States Food and Drug Administration. In some embodiments, an excipient is pharmaceutical grade. In some embodiments, an excipient meets the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia. Pharmaceutically acceptable excipients used in the manufacture of pharmaceutical compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in pharmaceutical formulations. Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents can be present in the composition, according to the judgment of the formulator. General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference). Kits The present invention further provides a pharmaceutical pack or kit comprising one or more containers filled with at least one humanized antibody or multi-specific binding agent (e.g., a bispecific antibody) as described herein. Kits may be used in any applicable method, including, for example, therapeutically or diagnostically. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects (a) approval by the agency of manufacture, use or sale for human administration, (b) directions for use, or both. Other features of the invention will become apparent in the course of the following descriptions of exemplary embodiments, which are given for illustration of the invention and are not intended to be limiting thereof. EXAMPLES The following examples are provided so as to describe to those of ordinary skill in the art how to make and use methods and compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Unless indicated otherwise, temperature is indicated in Celsius, and pressure is at or near atmospheric. Example 1. In Vitro Characterization of huA33-C825 Among other things, the present invention encompasses the insight that the humanized antibody A33 (huA33) was of particular interest for constructing multi-specific binding agents (e.g., a bispecific antibody). Without wishing to be bound by any particular theory, the present inventors proposed that suboptimal tumor dose and therapeutic index observed for radiolabeled monospecific huA33 (e.g.,131I-huA33) could be overcome by employing huA33 in a multi-specific format. This Example describes production of bispecific antibodies composed of a first antigen-binding site based on humanized antibody A33 and a second antigen-binding site that binds to a small molecule hapten (e.g., benzyl-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid [DOTA-Bn]). The data presented herein describes the successful production of bispecific antibodies (termed huA33-C825) to target colorectal cancer cells. As described herein, an anti-DOTA-Bn single chain Fv fragment (ScFv) based on a affinity matured 2D12.5 antibody was linked to the carboxyl end of a humanized A33 light chain. A major drawback in the development of antibody agents for pretargeted radioimmunotherapy (PRIT) has been radiation overexposure in normal tissues, immunogenicity, suboptimal tumor dose and a low therapeutic index. As demonstrated below, bispecific antibodies of the present invention overcome such deficiencies and provide for effective PRIT possibilities for cancers expressing the human A33 antigen such as colorectal cancer. Exemplary biochemical purity analysis of huA33-C825 by SE-HPLC is set forth inFIG.1. SE-HPLC showed a major peak (90% by UV analysis) with an approximate MW of 210 KDa, as well as some minor peaks assumed to be aggregates removable by gel filtration. The bispecific antibody remained stable by SE-HPLC and Biacore after multiple freeze and thaw cycles. Binding affinity was measured by Biacore T100. Exemplary results are set forth in Table 3. Exemplary sensorgrams are set forth inFIG.2. TABLE 3AntigenAgentkon(1/Ms)koff(1/s)KD(M)Human A33huA33-IgG16.14E+051.05E−031.71E−09huA33-C8259.15E+045.81E−036.35E−08BSA-DOTA-Bn(Y)hu3F8-C8251.60E+043.37E−042.12E−08huA33-C8251.90E+042.20E−041.16E−08 As shown in this Example, huA33-C825 demonstrated a lower affinity for the human A33 antigen as compared to the monospecific huA33 antibody (KDof 63.5 nM v. 1.71 nM,FIG.2A). HuA33-C825 retained high binding affinity for BSA-(Y)-DOTA-Bn (KDof 11.6 nM) as compared to a control bispecific antibody having a first antigen-binding site that does not bind the A33 antigen and a second antigen-binding site that binds DOTA-Bn (metal) (KDof 21.2 nM,FIG.2B). Taken together, this Example demonstrates the construction of a bispecific antibody that binds the human A33 antigen and a small-molecule hapten (e.g., DOTA-Bn) that retains high affinity for both targets. Further, the reduction in affinity to the human A33 antigen observed in huA33-C825 provides for a faster clearance as compared to parental huA33-IgG1 antibody. Example 2. Optimization of PRIT with huA33-C825, Dextran-Clearing Agent (Dextran-CA) and177Lu-DOTA-Bn This Example demonstrates the efficacy of a multi-specific binding agent (e.g., a bispecific antibody) for pretargeted radioimmunotherapy (PRIT) for A33-positive tumors. In particular, this Example describes the optimization of tumor targeting in a pretargeted radioimmunotherapy (PRIT) protocol in SW1222-tumor bearing rodents employing the bispecific antibody described in Example 1 as a function of the amount of a clearing agent (CA). As shown below, with increasing doses a progressive increase in therapeutic index is observed, but also a reduction in absolute tumor uptake. A 0.25 mg/mouse dose of huA33-C825 was selected based on pilot biodistribution studies in SW1222-tumor bearing mice at 24 h p.i. of177Lu-DOTA-Bn using 0.1-0.6 mg of huA33-C825 (0.48-2.86 nmol), and fixed ratios of CA and177Lu-DOTA-Bn (5.6 MBq), showing a plateau of −15-18% ID/g for the177Lu-activity concentration in tumor at 0.25-0.6 mg huA33-C825. Next, additional biodistribution experiments were performed to optimize the CA dose during PRIT with 0.25 mg (1.19 nmol) as the huA33-C825 dose. Groups of tumor-bearing mice (n=3 to 4 per group) with were injected with huA33-C825, followed 24 h later with either: saline (i.e. vehicle), 2.4% (w/w, with respect to 0.25 mg huA33-C825 dose), 5% (w/w), 10% (w/w), or 25% (w/w) CA doses (0-62.5 μg/mouse). After an additional 4 h, mice were injected with 5.6 MBq of177Lu-DOTA-Bn, and sacrificed 24 h later for biodistribution analysis. Exemplary optimization of clearing agent is shown inFIGS.3A-3D. Exemplary177Lu-DOTA-Bn activities in SW1222 tumor and various normal tissues for the groups of mice given 25% (w/w) dose of CA is shown inFIGS.4A-4D. As expected, the CA dose had a significant impact on the circulating (i.e., blood)177Lu-activity (from ˜8 to 0.1% ID/g for saline (no CA) to 25% (w/w), respectively). In addition, the CA dose appeared to reduce the capacity for subsequent177Lu-DOTA-Bn uptake at the tumor. The highest CA dose tested (25% (w/w)) was considered optimum since the tumor-to-normal organ ratios for the tissues with the highest radiosensitivity (blood and kidney) were highest compared to lower CA doses, although at the expense of a reduction in the177Lu-DOTA-Bn tumor uptake compared with PRIT with saline (˜50% less uptake). Specifically, the tumor uptake (as % ID/g, average±standard error of the mean) of177Lu-DOTA-Bn was 17.51±0.90 (n=3) and 8.46±3.74 (n=4), for saline and at the 65 μg dose level (25% (w/w)) for CA, respectively. For saline, the tumor-to-organ ratios for blood, kidney, and muscle were 2.2±0.4, 4.9±0.6, and 23.2±3.8, respectively. At the 65 μg CA dose level (25% (w/w)) for CA, the tumor-to organ ratios for blood, kidney, and muscle were 105.8±52.3, 18.4±13.4, and 282.1±208.7, respectively. Next,177Lu-DOTA-Bn dose titration studies were performed using the optimized PRIT doses for huA33-C825 and CA. For177Lu-DOTA-Bn dose titration studies, the177Lu-activity biodistribution data for tumor and critical select tissues (blood, liver, spleen, and kidneys) was compared between177Lu-DOTA-Bn dose groups as both % ID/g and absolute uptake (kBq/g; seeFIGS.4A,4B). Finally, a single-time point biodistribution experiment at 24 h p.i. of131I-trace labeled huA33-C825 (0.39-0.40 MBq with cold huA33-C825 added to 1.19 nmol) was performed in SW1222-tumor bearing mice to estimate the absolute antibody uptake of huA33-C825 in tumor (as pmol/g) during PRIT. Exemplary tabulated values are set forth in Table 4 (data is presented as mean±SD). Exemplary tumor uptake calculations are shown inFIG.4D. As shown in this Example, the131I-huA33-C825 uptake in tumor (average±standard deviation) was 3.71±0.97% ID/g. This corresponds to an absolute huA33-C825 uptake of 44 pmol/g (taking into account ˜50% immunoreactive fraction, then 88 pmol/g). TABLE 4Biodistribution study at 24 h p.i. followingi.v. injection of131I-A33-C825(0.39-0.40 MBq, 0.25 mg/1.19 nmol)into SW1222 tumor-bearing mice (n = 5).Tissues24 hrBlood5.05 ± 0.97Heart2.10 ± 0.31Lungs2.35 ± 0.52Liver2.04 ± 0.53Spleen1.36 ± 0.30Stomach6.92 ± 3.06Small Intestine0.98 ± 0.35Large Intestine0.83 ± 0.40Kidneys1.77 ± 0.31Muscle0.49 ± 0.06Bone0.71 ± 0.20Tumor3.71 ± 0.97Tumor size (g)0.86 ± 0.34 Example 3. Biodistribution and Absorbed Dose Calculation HuA33-C825 described in the prior Examples was tested for its in vivo efficacy. Biodistribution of radiolabeled DOTA-Bn and estimates of absorbed doses in mice implanted with SW1222 tumor cells were determined. In this Example, PRIT was carried out in groups of A33-positive SW1222 tumor-bearing mice with the optimum doses of huA33-C825 and CA, followed with 2.0 MBq (˜10 pmol) of177Lu-DOTA-Bn and biodistribution studies were carried out from 2-120 h p.i. of177Lu-DOTA-Bn to determine the177Lu-activity residence time in tumor and various normal tissues. Briefly,177Lu-activity in tumor and various normal tissues determined using a biodistribution assay following PRIT with optimum A33-C825 (0.25 mg/mouse) and dextran-clearing agent doses (25% (w/w), 62.5 μg) and 2.0 MBq (˜10 pmol) of177Lu-DOTA-Bn. Groups of SW1222 tumor-bearing mice (n=4 to 5) were given 250 μg of huA33-C825, followed 24 h later with 25% (w/w) (62.5 μg) dextran-clearing agent, and after an additional 4 h, 2.0 MBq (˜10 pmol) of 177Lu-DOTA-Bn. A single group of animals was sacrificed at 2, 24, and 120 h p.i. of177Lu-DOTA-Bn for biodistribution analysis. These data were used as described in the Materials and Methods to estimate absorbed doses for radioimmunotherapy with177Lu-DOTA-Bn (Table 5). For tumor,177Lu-uptake occurred very rapidly following administration, with an average of 7.0% ID/g at 2 h p.i. Maximum tumor uptake was 8.5% ID/g at 24 h p.i. and decreased by approximately half to 4.0% ID/g over the next 96 h at 120 h p.i. Peak kidney, liver, spleen and blood uptake was observed at 2 h p.i. (0.87, 0.70, 0.92, and 0.75% ID/g, respectively; average values), and decreased (also average values) to 0.27 (3.2-fold reduction compared to peak uptake), 0.30 (2.3-fold reduction), 0.32 (2.9-fold reduction), and 0.02 (37.5-fold reduction) % ID/g (also average values), respectively. Exemplary estimates of absorbed doses for tumor and select normal tissue in female athymic mice carrying s.c. A33 positive-colorectal cancer tumors for PRIT including the optimum huA33-C825 and dextran-clearing agent doses are set forth in Table 6. For each target region, the absorbed dose was calculated as the product of the177Lu equilibrium dose constant for non-penetrating radiations (i.e. beta rays) and the target regions177Lu cumulated activity, assuming complete local absorption of the177Lu beta rays and ignoring the gamma ray and non-self dose contributions. TABLE 5Tissue2 hr (n = 5)24 hr (n = 4)120 hr (n = 5)Blood0.75 ± 0.160.08 ± 0.020.02 ± 0.01Heart0.30 ± 0.050.11 ± 0.030.05 ± 0.01Lungs0.59 ± 0.100.21 ± 0.070.06 ± 0.02Liver0.70 ± 0.140.43 ± 0.090.30 ± 0.03Spleen0.92 ± 0.150.47 ± 0.140.32 ± 0.12Stomach0.15 ± 0.030.04 ± 0.010.02 ± 0.01Small Intestine0.14 ± 0.020.05 ± 0.010.02 ± 0.00Large Intestine0.17 ± 0.030.05 ± 0.010.04 ± 0.02Kidneys0.87 ± 0.090.46 ± 0.270.27 ± 0.09Muscle0.12 ± 0.010.03 ± 0.020.02 ± 0.00Bone0.09 ± 0.020.03 ± 0.020.03 ± 0.00Tumor6.99 ± 1.248.46 ± 3.743.99 ± 0.44Tumor size (g)1.31 ± 0.501.08 ± 0.450.98 ± 0.32Tumor-to-tissue ratios2 hr (n = 5)24 hr (n = 4)120 hr (n = 5)Blood9.3 ± 2.6107.2 ± 54.0181.0 ± 62.1Heart23.7 ± 6.178.1 ± 41.378.6 ± 22.3Lungs11.8 ± 3.040.2 ± 22.464.4 ± 25.0Liver10.0 ± 2.719.5 ± 9.513.5 ± 2.2Spleen7.6 ± 1.818.0 ± 9.712.5 ± 4.8Stomach46.6 ± 13.5225.0 ± 113.5166.2 ± 50.5Small Intestine49.7 ± 11.6182.0 ± 91.8210.5 ± 43.9Large Intestine41.5 ± 11.2177.8 ± 89.8104.3 ± 49.2Kidneys8.1 ± 1.718.3 ± 13.414.8 ± 4.9Muscle58.6 ± 12.4285.3 ± 212.8249.4 ± 58.6Bone78.0 ± 22.4293.8 ± 211.5149.5 ± 31.0 TABLE 6TherapeuticTissuescGy/MBqIndexBlood0.973Tumor65.8Heart1.447Lung1.837Liver6.310Spleen6.610Stomach0.6110Small Intestine0.5132Large Intestine0.882Kidneys5.312Muscle0.3219Bone0.6110 As shown in Table 6, the estimated absorbed doses of177Lu-DOTA-Bn (as cGy/MBq) for blood, tumor, liver, spleen, and kidneys were 0.9, 65.8, 6.3, 6.6, and 5.3 respectively. Further, for a single-cycle treatment, a therapeutic index of 73 for tumor to blood, and 12 for kidney, indicates curative ranges for tumor targeting, with no major toxicity expected. Indeed, no toxicity was observed out to 140 days in the subjects with durable responses. Tumor uptake after PRIT assessed by PET imaging demonstrated similar results as the biodistribution assay above with both the86Y-DOTA-Bn and177Lu-DOTA-Bn isotypes (data not shown). Example 4. In Vivo Therapy Study This Example illustrates the in vivo efficacy of a huA33-C825 bispecific antibody in pretargeted radioimmunotherapy to mediate a reduction in tumor burden in mice bearing A33-positive cancer cells. In particular, this Example describes effect of single- and dual-cycle therapy on tumor burden in SW1222-tumor bearing mice. During the first therapy study, 5 groups of tumor-bearing mice (n=6 to 8 per group) were treated with either: vehicle (i.e., untreated, n=8, TV7: 76±15 mm3), 33.3 MBq177Lu-DOTA-Bn alone (vehicle given during bispecific antibody and CA injections, n=6, TV7: 116±23 mm3), single-cycle IgG-C825 PRIT+33.3 MBq177Lu-DOTA-Bn (n.s. IgG-C825 given in place of huA33-C825, n=8 TV7: 100±10 mm3), or single-cycle huA33-C825 PRIT+either 11.1 MBq or 33.3 MBq177Lu-DOTA-Bn (both n=8, TV7: 103±17 mm3and TV7: 93±15 mm3, respectively). The estimated absorbed doses to tumor for single-cycle huA33-C825 PRIT+either 11.1 MBq or 33.3 MBq177Lu-DOTA-Bn were 730 and 2190 cGy, respectively (based on absorbed dose estimates from Table 6). The inventors observed that the relative tumor uptake decreased as the177Lu-DOTA-Bn dose was increased during treatment, which may indicate approaching possible saturation at the tumor. This may impact estimated absorbed tumor dose. If an estimated 7% ID/g is used for peak tumor uptake (i.e., to account for reduced relative tumor uptake with the higher177Lu-DOTA-Bn dose) following PRIT+33.3 MBq177Lu-DOTA-Bn dose, then an estimated tumor absorbed dose of −1800 cGy may be more accurate, overall suggesting an effective dose range of 1800-2200 cGy. Exemplary tumor response (represented as tumor volume [mm3]) among mice from each177Lu-DOTA-Bn treatment group is set forth inFIG.5. The groups of tumor-bearing mice receiving either no treatment, treatment consisting of either 33.3 MBq177Lu-DOTA-Bn alone, or single-cycle IgG-C825 PRIT+33.3 MBq177Lu-DOTA-Bn showed no tumor responses. Scintigraphy of the two latter groups given177Lu-DOTA-Bn showed minimal activity in the tumor region. In contrast, groups treated with single-cycle huA33-C825 PRIT+either 11.1 MBq or 33.3 MBq177Lu-DOTA-Bn showed a slight growth delay of the tumors up to ˜15 days following treatment, but produced no CR. For comparison, on day 23 post tumor-inoculation (16 days following177Lu-DOTA-Bn injection), the tumor volumes (as average±SEM) were 1398±206 (n=8), 1051±167 (n=5), 877±109 (n=7), 694±138 (n=8), and 495±76 (n=8) for no treatment, 33.3 MBq177Lu-DOTA-Bn alone, single-cycle IgG-C825 PRIT+33.3 MBq177Lu-DOTA-Bn, or single-cycle huA33-C825 PRIT+either 11.1 MBq or 33.3 MBq177Lu-DOTA-Bn, respectively. Within 30 days post tumor-inoculation, the average tumor size of all groups was >1250 mm3, and the study was terminated. Similar results were observed with a higher dose single cycle huA33-C825 PRIT+177Lu-DOTA-Bn treatment with 111.1 MBq177Lu-DOTA-Bn (data not shown). In a second therapy study, dual-cycle huA33-C825 PRIT treatment was investigated. Exemplary tumor response (represented as tumor volume [mm3]) among mice receiving dual-cycle treatment is set forth inFIGS.6A-6D. When mice were given either no treatment (n=5/TV10: 314±77 mm3), all mice required sacrifice within 30 days due to excessive tumor burden, and the time to reach 500 mm3was 13±2 d. Treatment with two cycles of PRIT+11.1 MBq177Lu-DOTA-Bn (total177Lu-DOTA-Bn dose 22.2 MBq; estimated tumor dose 1460 cGy) (n=5/TV10: 462±179 mm3), 2/5 animals showed CR (FIG.6A). In the recurrent tumors, the time to reach 500 mm3was 9 d (TV10: 391 mm3) or 36 d (TV10: 712 mm3). Treatment with 2 cycles of PRIT+33.3 MBq177Lu-DOTA-Bn (total177Lu-DOTA-Bn dose 66.6 MBq; estimated tumor dose 3600-4400 cGy) (n=5/344±105 mm3) produced CR (FIG.6B) in 5/5 animals. In these recurrent tumors, the time to reach 500 mm3was 12 d (TV10: 325 mm3), 65 d (TV10: 502 mm3), 7 d (TV10: 341 mm3), and 23 d (TV10: 345 mm3), and a single mouse had a tumor size of <10 mm3at time of sacrifice. Treatment with two cycles of PRIT+55.5 mCi177Lu-DOTA-Bn (total177Lu-DOTA-Bn dose 111.0 MBq; estimated tumor dose: 2580 cGy, based on peak tumor uptake of 3% ID/g) (n=4/236±54 mm3) produced CR in 4/4 animals (FIG.6C). In these recurrent tumors, the time to reach 500 mm3was 34 d (TV10: 295 mm3), 45 d (TV10: 263 mm3), and 42 d (TV10: 175 mm3), and a single mouse had a tumor size of 44 mm3at time of sacrifice. Following treatment with two cycles of PRIT+33.3 mCi177Lu-DOTA-Bn (total177Lu-DOTA-Bn dose: 66.6 MBq), the average recurrence time to 500 mm3was 27±26 d. For treatment with two cycles of PRIT+55.5 MBq177Lu-DOTA-Bn (total177Lu-DOTA-Bn dose: 111 MBq), the average recurrence time to 500 mm3was 40±6 d. Exemplary estimates of absorbed radiation doses (represented in Gy units) for each treatment regimen is set forth in Table 7. TABLE 7Cures atComplete40 d post-Treatment GroupTumorBloodKidneyResponsetreatmentControlsnon-pretargeted 11.1 MBq0/50/5non-pretargeted 33.3 MBq0/60/6IgG-C825 + 11.1 MBq0/50/5IgG-C825 + 33.3 MBq0/70/7Single-cyclehuA33-C825 + 11.1 MBq7.30.10.60/80/8huA33-C825 + 33.3 MBq21.90.31.80/80/8Dual-cyclehuA33-C825 + 11.1 MBq (×2); 22.2 MBq14.60.21.22/51/5huA33-C825 + 33.3 MBq (×2); 66.6 MBq43.80.63.55/52/5huA33-C825 + 55.5 MBq (×2); 111.0 MBq73.01.05.94/42/4Triple-cyclehuA33-C825 + 55.5 MBq (×3); 165.0 MBq1401.58.810/1010/10 Similar trends for survival were observed in animals at 140 days post-treatment (data not shown). Example 5. Toxicity This Example illustrates the in vivo toxicity of humanized A33 bispecific antibodies described in the prior Examples. Briefly, a total of six mice treated with either two cycles of PRIT+11.1 MBq of177Lu-DOTA-Bn (n=3) or two cycles of PRIT+1.5 mCi of177Lu-DOTA-Bn (n=3) were submitted for anatomic pathology assessment of kidney, bone marrow, liver, and spleen up to 9 weeks following treatment. The 3/5 mice that showed no CR during after treatment with two cycles of PRIT+0.3 mCi of177Lu-DOTA-Bn were submitted five days following injection of the second177Lu-DOTA-Bn dose (i.e., following treatment). These mice did not show any reduction in tumor size following treatment, and required sacrifice due to excessive tumor burden. In 3/3 mice, the kidney and bone marrow were normal, suggesting no radiation-induced toxicity. For 1/3 mice, the liver showed extramedullary hematopoiesis, and the liver was normal for the other two within the group. For 1/3 mice, the spleen (white pulp) showed follicular lymphoid hyperplasia, and the spleen was normal for the other two within the group. For the mice treated with two cycles of PRIT+55.5 MBq, a single mouse was submitted seven weeks following treatment, while the other two were submitted nine weeks following treatment. All three of these mice showed a CR, followed by reoccurrence of tumor, and required sacrifice due to excessive tumor burden. For 3/3 mice, the kidney, bone marrow, and liver were normal. For 1/3 mice, the spleen (white pulp) showed follicular lymphoid hyperplasia, and the spleen was normal for the other two within the group. This Example just confirms, among other things, that huA33-C825 effectively reduce tumor burden (i.e., reduce tumor growth) in vivo and provide for effective PRIT. Example 6. Curative Theranostic Prit This Example documents use of humanized A33 bispecific antibodies described in prior Examples, and, among other things, demonstrates treatments using these antibodies can be curative. Specifically, it demonstrates theranostic curative treatment regimens that included additional treatment cycles with increased total amounts of administered activity. Nude mice bearing established SW1222 s.c. xenografts (n=20; tumor volume=102±40 mm3; average±standard deviation (SD)) underwent treatment (n=5-10/group) with either: no treatment (n=5),177Lu-DOTA-Bn only (n=5), or a three-cycle PRIT regimen consisting of anti-GPA33 PRIT+55 MBq of177Lu-DOTA-Bn (n=10; total: 165 MBq). Serial nanoSPECT/CT imaging was conducted on five randomly selected mice undergoing DPRIT up to 160 hours post-injection of the first cycle of177Lu-DOTA-Bn for dosimetry calculations. DPRIT induced complete tumor response in 10/10 mice (controls: 10/10 dead at 21 days post-tumor inoculation), with tumor-free survival of all treated animals at 100 days and no obvious toxicities. Necropsy of 5/10 mice at 100 days verified cures, as well as showed no remarkable histopathologic findings of evaluation of kidney, liver, spleen, and bone/marrow (data not shown). Dosimetry estimates of177Lu-radiation exposure to tumor following cycle 1 was 4556±637 rads (n=5, average±SD). Based on these data, a first-order approximation of the total177Lu-radiation exposure to tumor following curative DPRIT (i.e., 3 cycles) was 14000 rads (with radiation doses to blood and kidney of 150 rads (therapeutic index (TI): 93) and 875 rads (TI: 16), respectively). Lutetium-177 nanoSPECT/CT imaging of three-cycle PRIT regimen treated animals showed high contrast with visible uptake in tumors and minimal tissue background (data not shown). TI˜70:1. Detection of tumors of 10 mg or less, based on non-invasive in vivo cross-sectional imaging in living mice was observed. This Example just confirms, among other things, that huA33-C825 effectively reduce tumor burden in vivo and that a PRIT-based theranostic may have curative effects and/or be used to detect small tumors. Example 7. Theranostic “Real-Time” Simultaneous Treatment and Image-Guided Dosimetry This Example documents in vivo response to a theranostic DOTA-PRIT regimen using humanized A33 bispecific antibodies described in the prior Examples and demonstrates treatment efficacy via simultaneous treatment and image-guided dosimetry. Specifically, nanoSPECT/CT was utilized for high-resolution quantitative imaging of mice undergoing177Lu-DPRIT treatment for “real-time” dosimetry. A SW1222-tumor bearing nude mouse (volume: 100 mm3according to Vernier caliper measurement) treated with a single cycle of anti-GPA33 PRIT+55 MBq of177Lu-DOTA-Bn and imaged by nanoSPECT/CT at three times following injection of177Lu-DOTA-Bn: at 1, 24, and 160 hours post-injection. Shown inFIG.8is the maximum intensity nanoSPECT/CT images of the lower flank region where the tumor is located. The images were decay corrected to the time of injection and calibrated using known activity standards. The activity concentration in tumor was determined using region-of-interest analysis of the calibrated images. This Example just confirms, among other things, that huA33-C825 effectively reduce tumor burden in vivo and that high-resolution quantitative imaging is one method that can be used to measure efficacy. Materials and Methods for Examples Tumor Cell Lines and Cell Culture Reagents The human colorectal cancer cell line SW1222 was obtained by the Ludwig Institute for Cancer Immunotherapy (New York, N.Y.) and maintained by serial passage. The cells were cultured in Minimal Essential Medium supplemented with 10% heat inactivated fetal calf serum, 2.0 mM glutamine, 100 units/mL penicillin, and 100 units/mL streptomycin in a 37° C. environment containing 5% CO2. Upon receipt of the cell line, cultures were established and cryopreserved in small aliquots to limit passages to less than three months, and periodically tested formycoplasmaaccording to manufacturer's specifications using a commercial kit (Lonza). For trypsinization during passage and harvesting of cells, a solution of 0.25% trypsin/0.53 mM EDTA in Hanks Buffered Salt Solution without calcium and magnesium was used. Cloning and Expression of huA33-C825 HuA33-C825 was made using the platform previously described in Cheal, S. M. et al. (2014, Mol. Cancer Ther. 13(7), 10 pages) using the variable regions (VHand VL) of humanized antibody A33 (huA33; King, D. J. et al., 1995, British J. Cancer 72:1364-1372). HuA33-C825 was produced in CHO cells in a mammalian expression vector and purified by protein A affinity chromatography as described (Cheal et al., supra). Exemplary bispecific antibodies of the present invention are presented in Table 8 (huA33-C825: humanized A33 IgG1-murine C825 scFv; huA33-huC825: humanized A33 IgG1-humanized C825 scFv). For DNA sequences, leader sequences are presented as underlined text. For amino acid sequences, leader sequences are presented as underlined text, linker sequences are presented as bold text and variable region sequences are presented as italicized text. TABLE 8huA33-C825 light chain DNAATGGGCTGGTCCTGCATCATCCTGTTTCTGGTGGCTACCGCCACCGGCGACATCCAGATGACCCAGTCCCCCTCCTCCCTGTCCGTGTCTGTGGGCGACAGAGTGACCATCACATGCAAGGCCTCCCAGAACGTGCGGACCGTGGTGGCCTGGTATCAGCAGAAGCCTGGCCTGGCCCCCAAGACCCTGATCTACCTGGCCTCTAACCGGCACACCGGCGTGCCCTCCAGATTCTCCGGATCTGGCTCTGGCACCGACTTTACCTTCACCATCTCCAGCCTGCAGCCCGAGGATATCGCCACCTACTTTTGCCAGCAGCACTGGTCCTACCCCCTGACCTTTGGCCAGGGCACCAAGGTGGAAGTGAAGAGAACCGTGGCCGCTCCCTCCGTGTTCATCTTCCCACCTTCCGACGAGCAGCTGAAGTCCGGCACCGCTTCTGTCGTGTGCCTGCTGAACAACTTCTACCCCCGCGAGGCCAAGGTGCAGTGGAAGGTGGACAACGCCCTGCAGTCCGGCAACTCCCAGGAATCCGTGACCGAGCAGGACTCCAAGGACAGCACCTACAGCCTGTCCTCCACCCTGACCCTGTCCAAGGCCGACTACGAGAAGCACAAGGTGTACGCCTGCGAAGTGACCCACCAGGGCCTGTCTAGCCCCGTGACCAAGTCTTTCAACCGGGGCGAATGTGGCGGCGGAGGATCTGGCGGAGGCGGCTCTGCTTCTCACGTGAAGCTGCAGGAAAGCGGCCCTGGACTGGTGCAGCCTTCCCAGTCTCTGTCCCTGACCTGCACCGTGTCCGGCTTCTCCCTGACCGATTACGGCGTGCACTGGGTGCGACAGTCTCCAGGCAAGGGCCTGGAATGGCTGGGAGTGATTTGGAGCGGTGGCGGAACCGCCTACAACACCGCCCTGATCTCCCGGCTGAACATCTACCGGGACAACTCCAAGAACCAGGTGTTCCTGGAAATGAACTCCCTGCAGGCAGAGGACACCGCCATGTACTACTGCGCCAGACGGGGCTCCTACCCCTACAACTACTTCGACGCTTGGGGCTGCGGCACCACCGTGACAGTGTCTAGCGGAGGTGGTGGATCTGGGGGCGGAGGTAGCGGAGGGGGAGGTTCTCAGGCTGTCGTGATCCAGGAATCTGCCCTGACCACCCCCCCTGGCGAGACAGTGACACTGACCTGCGGATCTTCCACCGGCGCTGTGACCGCCTCCAACTACGCCAACTGGGTGCAGGAAAAGCCCGACCACTGCTTCACCGGCCTGATCGGCGGCCACAACAACAGACCTCCAGGCGTGCCAGCCCGGTTCTCCGGCTCTCTGATCGGAGATAAGGCCGCCCTGACAATCGCCGGCACCCAGACAGAGGACGAGGCTATCTACTTCTGCGCCCTGTGGTACAGCGACCACTGGGTCATCGGCGGAGGCACCAGACTGACCGTGCTGGGATAG (SEQ IDNO: 1)huA33-C825 light chain amino acidMGWSCIILFLVATATGDIQMTQSPSSLSVSVGDRVTITCKASQNVRTVVAWYQQKPGLAPKTLIYLASNRHTGVPSRFSGSGSGTDFTFTISSLQPEDIATYFCQQHWSYPLTFGQGTKVEVKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECGGGGSGGGGSASHVKLQESGPGLVQPSQSLSLTCTVSGFSLTDYGVHWVRQSPGKGLEWLGVIWSGGGTAYNTALISRLNIYRDNSKNQVFLEMNSLQAEDTAMYYCARRGSYPYNYFDAWGCGTTVTVSSGGGGSGGGGSGGGGSQAVVIQESALTTPPGETVTLTCGSSTGAVTASNYANWVQEKPDHCFTGLIGGHNNRPPGVPARFSGSLIGDKAALTIAGTQTEDEAIYFCALWYSDHWVIGGGTRLTVLG(SEQID NO: 2)huA33-huC825 light chain amino acid (15 aa linker)MGWSCIILFLVATATGDIQMTQSPSSLSVSVGDRVTITCKASQNVRTVVAWYQQKPGLAPKTLIYLASNRHTGVPSRFSGSGSGTDFTFTISSLQPEDIATYFCQQHWSYPLTFGQGTKVEVKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECTSGGGGSGGGGSGGGGSHVQLVESGGGLVQPGGSLRLSCAASGFSLTDYGVHWVRQAPGKGLEWLGVIWSGGGTAYNTALISRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRGSYPYNYFDAWGCGTLVTVSSGGGGSGGGGSGGGGSQAVVTQEPSLTVSPGGTVTLTCGSSTGAVTASNYANWVQQKPGQCPRGLIGGHNNRPPGVPARFSGSLLGGKAALTLLGAQPEDEAEYYCALWYSDHWVIGGGTKLTVLG(SEQ ID NO: 3)huA33-huC825 light chain amino acid (30 aa linker)MGWSCIILFLVATATGDIQMTQSPSSLSVSVGDRVTITCKASQNVRTVVAWYQQKPGLAPKTLIYLASNRHTGVPSRFSGSGSGTDFTFTISSLQPEDIATYFCQQHWSYPLTFGQGTKVEVKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECTSGGGGSGGGGSGGGGSHVQLVESGGGLVQPGGSLRLSCAASGFSLTDYGVHWVRQAPGKGLEWLGVIWSGGGTAYNTALISRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRGSYPYNYFDAWGCGTLVTVSSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSQAVVTQEPSLIVSPGGTVTLTCGSSTGAVTASNYANWVQQKPGQCPRGLIGGHNNRPPGVPARFSGSLLGGKAALTLLGAQPEDEAEYYCALWYSDHWVIGGGTKLTVLG(SEQ ID NO: 4)huA33-C825 heavy chain IgG1 DNA (aglycosylated)ATGGGCTGGTCCTGCATCATCCTGTTTCTGGTGGCTACCGCCACCGGCGAGGTGCAGCTGCTGGAATCTGGCGGAGGACTGGTGCAGCCTGGCGGCTCTCTGAGACTGTCTTGTGCCGCCTCTGGCTTCGCCTTCTCCACCTACGACATGTCCTGGGTGCGACAGGCTCCTGGCAAGGGCCTGGAATGGGTGGCCACAATCTCTTCCGGCGGCTCCTACACCTACTACCTGGACTCTGTGAAGGGCCGGTTCACCATCTCCCGGGACTCCTCCAAGAACACCCTGTACCTGCAGATGAACTCCCTGCAGGCCGAGGACTCCGCCATCTACTACTGTGCCCCTACCACCGTGGTGCCCTTCGCTTATTGGGGCCAGGGCACCCTCGTGACCGTGTCCTCTGCTTCTACCAAGGGCCCATCGGTCTTCCCCCTGGCACCCTCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCACACCTTCCCGGCCGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCCTCCAGCAGCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGCCCAGCAACACCAAGGTGGACAAGAGAGTTGAGCCCAAATCTTGTGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAACTCCTGGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACGCCAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCCCAGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAATGA (SEQ ID NO: 5)huA33-C825 heavy chain IgG1 amino acid (aglycosylated)MGWSCHILFLVATATGEVQLLESGGGLVQPGGSLRLSCAASGFAFSTYDMSWVRQAPGKGLEWVATISSGGSYTYYLDSVKGRFTISRDSSKNTLYLQMNSLQAEDSAIYYCAPTTVVPFAYWGQGTLVIVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 6)huA33-C825 heavy chain IgG1 amino acid (aglycosylated, K322A)MGWSCIILFLVATATGEVQLLESGGGLVQPGGSLRLSCAASGFAFSTYDMSWVRQAPGKGLEWVATISSGGSYTYYLDSVKGRFTISRDSSKNTLYLQMNSLQAEDSAIYYCAPTIVVPFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCAVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 7) Surface Plasmon Resonance Studies Biacore T100 Biosensor, CM5 sensor chip, and related reagents were purchased from GE Healthcare. Recombinant human A33 protein was purchased from Novoprotein. A BSA-(Y)-DOTA-Bn conjugate was prepared as described (Cheal et al., supra). A33 and DOTA antigens were immobilized using the Amino Coupling kit (GE Healthcare). Purified bispecific antibodies and control antibodies were analyzed, and data were fit to a bivalent analyte model using the Biacore T100 evaluation software as described (Cheal et al., supra). PRIT Reagents, Protocol and Xenograft Studies All animal experiments were approved by the Institutional Animal Care and Use Committee of Memorial Sloan Kettering Cancer Center and institutional guidelines for the proper and humane use of animals in research were followed. Athymic nu/nu female mice (6-8 weeks old; Harlan Sprague Dawley) were allowed to acclimate in the vivarium for at least one week. Groups of animals were injected s.c. with A33-positive SW1222 in the left flank with 5×106cells formulated 1:1 with Matrigel (BD Biosciences), and established tumors (100-900 mm2) were observed in 7-10 days using the formula for the volume of an ellipsoid V=4/3π(length/2×width/2×height/2). All reagents were given intravenously (i.v.) via the lateral tail vein. PRIT protocol included injections of: huA33-C825 [t=−28 h], followed 24 h later by CA (the CA is a 500 KDa dextran-(Y)-DOTA-Bn conjugate, prepared according to Orcutt et al. (2011, Nucl. Med. Biol. 38:223-233) and formulated in saline for injection; the substitution ratio of moles of (Y)-DOTA-Bn per moles of dextran was 61(Y)-DOTA-Bn/dextran) [t=−4 h], and177Lu-DOTA-Bn (prepared as previously described by incubating aminobenzyl-DOTA(p-NH2-Bn-DOTA) from Macrocyclics and177LuCl3(specific activity ˜30 Ci/mg; Perkin Elmer) and formulating in saline for injection) after 4 h [t=0 h]. In addition, huA33-C825 was trace radiolabeled with I-131 to estimate tumor uptake during PRIT. The IODOGEN method (Cheal, S. et al., 2014, Mol. Cancer Ther. 13(7):1-10) was used to prepare131I-huA33-C825 (final specific activity 95.5 MBq/mg, with cold huA33-C825 added to achieve desired mg dose, radiochemical purity >98% using size-exclusion high pressure liquid chromatography), and the in vitro cell binding immunoreactivity was evaluated using SW1222 cells essentially as described by Lindmo method. (Lindmo, T. et al., 1990, J. Immunol. Meth. 126(2):183-189). For PRIT with non-specific IgG-C825, an equivalent mg dose of a GD2-targeted bispecific antibody (hu3F8-C825) was used in place of huA33-C825. For ex vivo biodistribution analysis, mice were euthanized by CO2(g) asphyxiation, and tumor and selected organs were harvested, rinsed with water and allowed to air-dry, weighed and radioassayed by gamma scintillation counting (Perkin Elmer Wallac Wizard 3″). Count rates were background and decay corrected, converted to activities using a system calibration factor, normalized to the administered activity, and expressed as percent injected dose per gram (% ID/g). Differences in177Lu-activity concentration in tumor and various tissues were analyzed by Student's unpaired t test when appropriate. Estimation of Absorbed Doses Groups of A33-positive SW1222 tumor-bearing mice (n=4-5) were given 0.25 mg of huA33-C825, CA (62.5 μg; 25% (w/w)), and 1.85-2.0 MBq (˜10 pmol) of177Lu-DOTA-Bn, and sacrificed at 2, 24, and 120 h p.i. For each tissue the non-decay-corrected time-activity concentration data were fit using Excel to a 1-component, a 2-component, or a more complex exponential function as appropriate, and analytically integrated to yield the cumulated activity concentration per unit administered activity (MBq-h/g per MBq). The177Lu equilibrium dose constant for non-penetrating radiations (8.49 g-cGy/MBq-h) was used to estimate the tumor-to-tumor and select organ-to-organ self-absorbed doses, assuming complete local absorption of the177Lu beta rays only and ignoring the gamma ray and non-self dose contributions. To determine the effect of the177Lu-DOTA-Bn dose on the relative uptake of177Lu-DOTA-Bn in tumor and select tissues with the highest absorbed doses (i.e., blood, liver, spleen, and kidneys), groups of SW1222 tumor-bearing female athymic nude mice (n=5/group) were given 0.25 mg (1.19 nmol) of huA33-C825 at t=−28 h and 62.5 μg of CA at t=−4 h, followed with either 11.1 MBq (11.14-11.40), 55.5 MBq (54.61-55.06 MBq), or 111 MBq (109.52-112.5 MBq). All groups were sacrificed at 24 h p.i. of177Lu-DOTA-Bn (i.e., time of maximum tumor uptake) for biodistribution analysis of177Lu-activity. PET Imaging of PRIT+86Y-DOTA-Bn A single group of mice bearing A33-positive SW1222 tumors in the shoulder (n=5) were given 0.25 mg of huA33-C825, CA (62.5 μg; 25% (w/w)), and 8.6-8.8 MBq (˜50 pmol) of86Y-DOTA-Bn, and non-invasively imaged using a microPET Focus 120 (CTI Molecular Imaging, Inc. Knoxville, Tenn.) at approximately 2 and 20 h p.i. The following imaging acquisition parameters were used: energy window of 350-750 keV, coincidence timing window of 6 nsec, and an acquisition time of 20 min. The resulting list-mode data were sorted into 2D histograms by Fourier re-binning and transverse images reconstructed by filtered back-projection into a 128×128×95 matrix (reconstructed spatial resolution is 2.6 mm full-width half maximum (FWHM)). The image data were corrected for non-uniformity of response of the scanner, deadtime count losses, physical decay (to the time of injection), and the86Y positron branching ratio. No attenuation, scatter, or partial-volume averaging correction was applied. An empirically determined system calibration factor (i.e. μCi/mL/cps/voxel) for mice was used to convert voxel count rates to activity concentrations. The resulting image data were then normalized to the administered activity to determine by region-of-interest analysis the percent of the injected dose per gram (% ID/g) of tissue corrected for radioactive decay to the time of injection. AsiPRO VM 5.0 software (Concorde Microsystems, Knoxville, Tenn.) was used to perform image and region of interest (ROI) analyses (as ROI maximum, % ID/g). The animals were sacrificed at 24 h p.i. for ex vivo biodistribution analysis. Autoradiography and Immunohistochemistry Frozen and OCT-embedded tumor and kidney from select mice administered huA33-C825 PRIT followed with either 11.1 (11.14-11.40), 55.5 (54.61-55.06), or 111 MBq (109.52-112.5 MBq) of177Lu-DOTA-Bn (time of sacrifice: 24 hours p.i.) were cut into 10 μm sections using a cryostat (Avantik, Springfield, NJ), and immediately exposed to imaging plate (Fuji Photo Film, Kanagawa, Japan) for 72 h and subsequently scanned using Typhoon FLA 7000 scanner (GE, Pittsburgh, Pa.). The same sections underwent hematoxylin and eosin staining and were scanned under Olympus BX60 microscope equipped with controlled moving stage (Olympus, Central Valley, Pa.). Both autoradiogram and microscope images were processed and analyzed using ImageJ (NIH). Therapy and Scintigraphy Studies Groups of mice bearing established s.c. A33-positive SW1222 xenografts were injected with either huA33-C825 or non-specific (n.s.) IgG-C825 PRIT (i.e., single-cycle treatment,177Lu-DOTA-Bn injection on day 7 post tumor-inoculation) or two cycles of PRIT (i.e., dual-cycle treatment study,177Lu-DOTA-Bn injections given on day 10 and day 17 post tumor-inoculation). For the dual-cycle treatment study, the tumor volume on day 10-post tumor inoculation (TV10) is described (i.e., day of first177Lu-DOTA-Bn injection) and expressed when appropriate as average±SD. The following definitions were used to describe treatment response: a complete response (CR) is defined as tumor shrinkage to <100 mm3. A durable response (DR) was defined as survival at 140 days post treatment. Excessive tumor burden is defined as >2000 mm3. For scintigraphy studies, select groups of A33-positive SW1222 tumor-bearing mice undergoing treatment were placed under anesthesia by gas inhalation before scanning in a nanoSPECT (Bioscan, Washington D.C.) at 20 hours p.i. for 30 minutes (˜105counts per image) using a low-energy high-resolution collimator and a window set at 208 keV. Images were reconstructed to a 256×256 matrix using Bioscan HiSPECT software and uploaded into ASIPro VM for analysis. EQUIVALENTS Use of ordinal terms such as “first,” “second,” “third,” etc., in the claims to modify a claim element does not by itself connote any priority, precedence, or order of one claim element over another or the temporal order in which acts of a method are performed, but are used merely as labels to distinguish one claim element having a certain name from another element having a same name (but for use of the ordinal term) to distinguish the claim elements. The articles “a” and “an” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to include the plural referents. Claims or descriptions that include “or” between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The invention also includes embodiments in which more than one, or the entire group members are present in, employed in, or otherwise relevant to a given product or process. Furthermore, it is to be understood that the invention encompasses all variations, combinations, and permutations in which one or more limitations, elements, clauses, descriptive terms, etc., from one or more of the listed claims is introduced into another claim dependent on the same base claim (or, as relevant, any other claim) unless otherwise indicated or unless it would be evident to one of ordinary skill in the art that a contradiction or inconsistency would arise. Where elements are presented as lists, (e.g., in Markush group or similar format) it is to be understood that each subgroup of the elements is also disclosed, and any element(s) can be removed from the group. It should be understood that, in general, where the invention, or aspects of the invention, is/are referred to as comprising particular elements, features, etc., certain embodiments of the invention or aspects of the invention consist, or consist essentially of, such elements, features, etc. For purposes of simplicity those embodiments have not in every case been specifically set forth in so many words herein. It should also be understood that any embodiment or aspect of the invention can be explicitly excluded from the claims, regardless of whether the specific exclusion is recited in the specification. The publications, websites and other reference materials referenced herein to describe the background of the invention and to provide additional detail regarding its practice are hereby incorporated by reference. Having thus described several aspects of at least one embodiment of this invention, it is to be appreciated that various alterations, modifications, and improvements will readily be apparent to those skilled in the art. Such alterations, modifications, and improvements are intended to be part of this disclosure, and are intended to be within the spirit and scope of the invention. Accordingly, the foregoing description and drawings are by way of example only and the invention is described in detail by the claims that follow. REFERENCES Ackerman, M. E. et al., 2008, A33 antigen displays persistent surface expression, Cancer Immunol. Immunother. 57(7):1017-1027.Ackerman, M. E. et al., 2008, Effect of antigen turnover rate and expression level on antibody penetration into tumor spheroids, Mol. Cancer Ther. 7(7):2233-2240.Barendswaard, E. C. et al., 1998, Rapid and specific targeting of monoclonal antibody A33 to a colon cancer xenograft in nude mice, International J. Oncol. 12:45-53.Carrasquillo, J. A. et al., 2011,124I-huA33 Antibody PET of Colorectal Cancer, J. Nucl. Med. 52:1173-1180.Cheal, S. M. et al., 2014, Preclinical Evaluation of Multistep Targeting of Diasialoganglioside GD2 Using an IgG-scFv Bispecific Antibody with High Affinity for GD2 and DOTA Metal Complex, Mol. Cancer Ther. 13(7):1-10.Cheal, S. M. et al., 2014, Evaluation of glycodendron and synthetically-modified dextran clearing agents for mult-step targeting of radioisotopes for molecular imaging and radioimmunotherapy, Mol. Pharm. 11(2):400-416.El Emir, E. et al., 2007, Predicting Response to Radioimmunotherapy from the Tumor Microenvironment of Colorectal Carcinomas, Cancer Res. 67(24):11896-11905.Goodwin, D. A. et al., 1994, Pharmacokinetics of pretargeted monoclonal antibody 2D12.5 and88Y-Janus-2-(p-nitrobenzyl)-1,4,7,10-tetraazacyclododecanetetraacetic acid (DOTA) in BALB/c mice with KHJJ mouse adenocarcinoma: a model for90Y radioimmunotherapy, Cancer Res. 54(22):5937-5946.King, D. J. et al., 1995, Preparation and preclinical evaluation of humanised a33 immunoconjugates for radioimmunotherapy, British J. Cancer 72:1364-1372.Lindmo, T. et al., 1990, Immunometric assay by flow cytometry using mixtures of two particle types of different affinity, J. Immunol. Meth. 126(2):183-189.Orcutt, K. D. et al., 2010, A modular IgG-scFv bispecific antibody topology, Protein Engineering Design & Selection 23(4):221-228.Orcutt, K. D. et al., 2011, Engineering an antibody with picomolar affinity to DOTA chelates of multiple radionuclides for pretargeted radioimmunotherapy and imaging, Nucl. Med. Biol. 38(2):223-233.O'Donoghue, J. A. et al., 2011,124I-huA33 antibody uptake is driven by a33 antigen concentration in tissues from colorectal cancer patients imaged by immuno-pet. J. Nucl. Med. 52:1878-1885.Scott, A. M. et al., 2005, A phase I trial of humanized monoclonal antibody A33 in patients with colorectal carcinoma: biodistribution, pharmacokinetics, and quantitative tumor uptake, Clin. Cancer Res. 11(13):4810-4817.Welt, S. et al., 1994, Phase I/II study of iodine 131-labeled monoclonal antibody A33 in patients with advanced colon cancer, J. Clin. Oncol. 12(8):1561-71.Welt, S. et al., 2003, Phase I study of anticolon cancer humanized antibody A33, Clin. Cancer Res. 9:1338-1346.
190,207
11858994
DESCRIPTION OF EMBODIMENTS The present invention is explained hereinafter while showing the best mode of the invention. Throughout the entire specification, a singular expression should be understood as encompassing the concept thereof in the plural form, unless specifically noted otherwise. Thus, singular articles (e.g., “a”, “an”, “the”, and the like in the case of English) should also be understood as encompassing the concept thereof in the plural form, unless specifically noted otherwise. Further, the terms used herein should be understood as being used in the meaning that is commonly used in the art, unless specifically noted otherwise. Thus, unless defined otherwise, all terminologies and scientific technical terms that are used herein have the same meaning as the general understanding of those skilled in the art to which the present invention pertains. In case of a contradiction, the present specification (including the definitions) takes precedence. Hereinafter, the definitions and/or details of the basic technical details that are especially used herein are explained herein as appropriate. (Cancer Immunotherapy) As used herein, “cancer immunotherapy” refers to a method of treating cancer using the immune mechanism of an organism. Cancer immunotherapy is roughly categorized into cancer immunotherapy by strengthening the immune function against cancer and cancer immunotherapy by inhibiting the immune evasion function of cancer. Cancer immunotherapy also includes active immunotherapy for activating the immune function in the body and passive immunotherapy for activating the immune function outside the body, or returning grown immune cells into the body. It was discovered that responsiveness to a therapeutic effect of such cancer immunotherapy can be predicted with diversity of a TCR repertoire as an indicator by the method described in the present invention. Examples of cancer immunotherapy include non-specific immunopotentiators, cytokine therapy, cancer vaccine therapy, dendritic cell therapy, adoptive immunotherapy, non-specific lymphocyte therapy, cancer antigen specific T cell therapy, antibody therapy, immune checkpoint inhibition therapy, CAR-T therapy, and the like. Immune checkpoint (inhibition) therapy using an immune checkpoint inhibitor has recently drawn significant attention (Pardoll D M. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012 Mar. 22; 12(4): 252-64.). Cancer cells express various proteins on the surface, but this leads to evasion from attacks by immune cells such as T cells, so that cancer tissue cannot be eliminated only by the biological immune function in a normal state. An immune checkpoint inhibitor inhibits the ligand-receptor interaction or the like, which is responsible for the transmission of a suppression signal from such cancer tissue to the immune function to enable efficient cancer elimination by the biological immune function. One embodiment of the present invention is a method of using T cell receptor (TCR) diversity of T cells (e.g., CD8+PD-1+T cells) as an indicator for predicting responsiveness of a subject to the immune checkpoint inhibitors described below. Another embodiment of the present invention is a method of administering the immune checkpoint inhibitor shown below to a (responsive) subject who has been selected based on T cell receptor (TCR) diversity. Another embodiment provides a method of suspending, discontinuing, or avoiding the administration of an immune checkpoint inhibitor to a subject who has been determined to be non-responsive based on T cell receptor (TCR) diversity. A representative example of immune checkpoint inhibitors is a PD-1 inhibitor. Examples of PD-1 inhibitors include, but are not limited to, anti-PD-1 antibodies nivolumab (sold as Opdivo™) and pembrolizumab (sold as Keytruda™). In one preferred embodiment, nivolumab can be selected as such an inhibitor. Although not wishing to be bound by any theory, one reason that a therapy using nivolumab is preferred is because the Examples have demonstrated that responsive subjects and non-responsive subjects can be clearly distinguished by using a diversity index calculated by the large-scale high efficiency TCR repertoire analysis of the present invention, and it is elucidated that responsiveness and non-responsiveness can be clearly distinguished by a specific threshold value using a DE50 index in particular. Of course, it is understood that a diversity index can also be used to the same extent for other PD-1 inhibitors It is understood that an anti-PD-1 antibody exerts an anticancer effect by releasing the suppression of T cell activation by a PD-1 signal. It is understood that an interaction between PD-1 (programmed death 1) and PD-L1 or PD-L2 recruits SHP-2, which is one type of protein tyrosine phosphatase, to the cytoplasmic domain of PD-1 and inactivates ZAP70, which is a T cell receptor signaling protein, to suppress T cell activation (Okazaki, T., Chikuma, S., Iwai, Y. et al.: A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat. Immunol., 14, 1212-1218 (2013)). It is understood that PD-L1 also interacts with CD80 to suppress T cell activation (Butte, M. J., Keir, M. E., Phamduy, T. B. et al.: PD-L1 interacts specifically with B7-1 to inhibit T cell proliferation. Immunity, 27, 111-122 (2007)). It is understood that PD-1 is highly expressed in killer T cells and natural killer cells that infiltrate cancer tissue, and the immune response is attenuated by PD-L1 on the tumor. If such attenuation of an immune response due to a PD-1 signal is inhibited by an anti-PD-1 antibody, an effect of enhancing antitumor immune responses is attained. Other examples of immune checkpoint inhibitors include PD-L1 inhibitors (e.g., anti-PD-L1 antibodies avelumab, durvalumab, and atezolizumab). A PD-L1 inhibitor inhibits the PD-1 pathway by binding to the PD-L1 side, resulting in an antitumor immune response. Other examples of immune checkpoint inhibitors include CTLA-4 inhibitors (e.g., anti-CTLA-4 antibodies ipilimumab and tremelimumab). A CTLA-4 inhibitor activates T cells in a pathway that is different from PD-1 inhibition, resulting in an antitumor immune response. T cells are activated by the interaction of the surface CD28 with CD80 or CD86. However, it is understood that activation of even T cells which have been activated once is suppressed by surface expressed CTLA-4 (cytotoxic T-lymphocyte-associated antigen 4) preferentially interacting with CD80 or CD86 with higher affinity than with CD20. A CTLA-4 inhibitor inhibits CTLA-4 to prevent inhibition of interaction between CD20 and CD80 or CD86, resulting in an antitumor immune response. In another embodiment, an immune checkpoint inhibitor may target an immune checkpoint protein such as TIM-3 (T-cell immunoglobulin and mucin containing protein-3), LAG-3 (lymphocyte activation gene-3), B7-H3, B7-H4, B7-H5 (VISTA), or TIGIT (T cell immunoreceptor with Ig and ITIM domain). It is understood that such an immune checkpoint suppresses an immune response to autologous tissue, but immune checkpoints also increase in T cells when an antigen such as a virus remains within the body for a long period of time. It is understood that tumor tissue evades antitumor immunity by these immune checkpoints because an antigen remains in the body for a long period of time. Such an immune checkpoint inhibitor disables such an evasion function to achieve an antitumor effect. One embodiment of the present invention provides an indicator for predicting responsiveness of a subject with cancer to cancer immunotherapy. Example of target cancer in the present invention include, but are not limited to, lung cancer, non-small cell cancer, renal (renal cell) cancer, prostate cancer, gastric cancer, testicular cancer, liver cancer (hepatoma), skin cancer, esophageal cancer, melanoma, pancreatic cancer, pancreatic carcinoma, bone tumor/osteosarcoma, colon cancer, soft tissue tumor, biliary tract cancer, multiple myeloma, malignant lymphoma (Hodgkin's lymphoma, non-Hodgkin's lymphoma), bladder cancer, laryngeal cancer, uterine cancer (endometrial, cervical), head and neck cancer, ovarian cancer, breast cancer, and the like. One embodiment of the present invention provides a method of using TCR diversity of a subject with lung cancer as an indicator of responsiveness of the subject to cancer immunotherapy. (TCR Diversity) The biological defense mechanism using the immune system is heavily dependent on the specific immunity provided mainly by T cells and B cells. T cells and B cells can specifically recognize and attack exogenous pathogens such as viruses or bacteria without reacting to autologous cells or molecules. For this reason, T cells and B cells have a mechanism that can recognize and distinguish various antigens from other organisms in addition to autoantigens by a receptor molecule expressed on the cell surface. In T cells, T cell receptors (TCR) function as an antigen receptor. An intracellular signal is transmitted by a stimulation from such antigen receptors. Production of inflammatory cytokines, chemokines or the like are promoted, cell proliferation increases, and various immune responses are initiated. TCR recognizes a peptide bound to a peptide binding cleft of a major histocompatibility complex (MHC) expressed on antigen presenting cells (peptide-MHC complex, pMHC) to distinguish autologous and heterologous and recognizes an antigen peptide (Cell 1994, 76, 287-299). TCRs are heterodimer receptor molecules consisting of two TCR polypeptide chains. There are αβ TCRs expressed by normal T cells and γδ TCRs with a special function. α and β chain TCR molecules form a complex with a plurality of CD3 molecules (CD3ζ chain, CD3ε chain, CD3γ chain, and CD3δ chain), transmit an intracellular signal after antigen recognition, and initiate various immune responses. With a viral infection, an endogenous antigen such as a cancer antigen derived from a cancer cell or a viral antigen proliferated in a cell is presented as an antigen peptide on an MHC class I molecule. Further, an antigen derived from an exogenous microorganism is taken up and processed by an antigen-presenting cell by endocytosis, and then presented on an MHC class II molecule. Such antigens are recognized by TCRs expressed by each of CD8+ T cell and CD4+ T cell. It is also known that a costimulatory molecule such as a CD28, ICOS, or OX40 molecule is important for stimulation via a TCR molecule. A TCR gene consists of numerous V regions (variable region, V), J regions (joining region, J), D regions (diversity region, D), and C regions (constant regions, C) encoded by different regions in the genome. In a T cell differentiation process, such gene fragments are genetically rearranged in various combinations. α chain and γ chain TCRs express genes consisting of V-J-C, and β chain and δ chain TCRs express genes consisting of V-D-J-C. Diversity is created by rearrangement of such gene fragments. In addition, insertion or deletion of one or more bases between V and D or D and J gene fragments leads to the formation of a random amino acid sequence to create a more diverse TCR gene sequence. A region where a TCR molecule directly binds to a pMHC complex surface (TCR footprint) is composed of diverse complementarity determining regions (CDR) within the V region, CDR1, CDR2, and CDR3 regions. The CDR3 region in particular comprises a part of a V region, a part of J region and a V-D-J region formed by a random sequence, forming the most diverse antigen recognition site. Meanwhile, the other regions are called FRs (framework region) serving the role of forming a backbone structure of a TCR molecule. In a differentiation and maturation process of a T cell in the thymus gland, a β chain TCR is genetically rearranged initially, and conjugates with a pTα molecule to form a pre-TCR complex molecule. An α chain TCR is then rearranged to form an αβ TCR molecule, and when a functional αβ TCR is not formed, rearrangement occurs in the other α chain TCR gene allele. It is known that after undergoing positive/negative selection in the thymus gland, a TCR with a suitable affinity is selected to acquire antigen specificity (Annual Review Immunology, 1993, 6, 309-326). T cells produce one type of TCR with high specificity to a specific antigen. With numerous antigen specific T cells in the living body, a diverse TCR repertoire can be formed to effectively function as a defense mechanism against various pathogens. As used herein, “TCR diversity” refers to diversity of the repertoire of T cell receptors of a subject. Those skilled in the art can measure TCR diversity using various means known in the art. An index indicating TCR diversity is referred to as a “TCR diversity index”. Any TCR diversity index that is known in the art can be used. Diversity indices such as Shannon-Weaver index, Simpson index, inverse Simpson index, normalized Shannon-Weaver index, DE index (e.g., DE50 index, DE30 index, or DE80 index), or Unique index (e.g., Unique50 index, Unique30 index, or Unique80 index) can be applied to and used for TCRs. One of the methods is a method of analyzing how much of individual V chains are used by T cells in a sample by analyzing the ratio of T cells expressing individual Vβ chains using a specific Vβ chain specific antibody with flow cytometry (FACS analysis). In addition thereto, TCR repertoire analysis using a molecular biological technique has been designed based on information on TCR genes obtained from the human genomic sequence. This is a method of extracting an RNA from a cell sample to synthesize a complementary DNA and then amplifying and quantifying a TCR gene by PCR. Nucleic acids can be extracted from cell samples using a tool that is known in the art, such as RNeasy Plus Universal Mini Kit (QIAGEN). Whole RNA can be extracted and purified from cells dissolved in a TRIzol LS reagent using RNeasy Plus Universal Mini Kit (QIAGEN). A complementary DNA can be synthesized from an extracted RNA using any reverse transcriptase that is known in the art, such as Superscript III™ (Invitrogen). Those skilled in the art can appropriately perform PCR amplification of a TCR gene using any polymerase that is known in the art. However, “unbiased” amplification can have an advantageous effect for accurate measurement in amplification of a gene with large variation such as a TCR gene. It has been conventional to use a method of designing numerous individual TCR V chain specific primers to separately quantify by real-time PCR or the like, or a method of simultaneously amplifying such specific primers (Multiple PCR) for primers used of PCR amplification. However, even in quantification using an endogenous control for each V chain, accurate analysis is not possible when a large number of primers are used. Furthermore, multiple PCR has a disadvantage in that a difference in efficiency of amplification among primers results in a bias in PCR amplification. In order to overcome such a disadvantage of multiple PCR, Tsuruta et al reported Adaptor-ligation PCR, which adds an adaptor to the 5′ terminal of a double stranded complementary DNA of a TCR gene and then amplifies all γδ TCR genes with a common adaptor primer and a C region specific primer (Journal of Immunological Methods, 1994, 169, 17-23). Furthermore, methods applied to amplification of αβ TCR genes for quantification with oligoprobes specific to individual V chains were developed, i.e., Reverse dot blot (Journal of Immunological Methods, 1997, 201, 145-15.) and Microplate hybridization assay (Human Immunology, 1997, 56, 57-69). In a preferred embodiment of the present invention, TCR genes including all isotype and subtype genes are amplified with a set of primers consisting of one type of forward primer and one type of reverse primer without changing the frequency of presence to determine TCR diversity as described in WO 2015/075939 (Repertoire Genesis Inc.) The following primer design is advantageous for unbiased amplification. Focus was placed on the genetic structure of a TCR or BCR gene. An adaptor sequence is added, without setting a primer to highly diverse V regions, to a 5′ terminal thereof to amplify all V region comprising genes. Such an adaptor can have any length or sequence in a base sequence. About 20 base pairs are optimal, but a sequence from 10 bases to 100 bases can be used. An adaptor added to the 3′ terminal is removed with a restriction enzyme. In addition, all TCR genes are amplified by amplifying with a reverse primer specific to a C region which has a common sequence with an adaptor primer with the same sequence as a 20 base pair adaptor. A complementary strand DNA is synthesized with a reverse transcriptase from a TCR or BCR gene messenger RNA and then a double stranded complementary DNA is synthesized. A double stranded complementary DNA comprising V regions with different lengths is synthesized by a reverse transcription reaction or a double strand synthesizing reaction. Adaptors consisting of 20 base pairs and 10 base pairs are added to the 5′ terminal section of such genes by a DNA ligase reaction. The genes can be amplified by setting a reverse primer in a C region of an α chain, β chain, γ chain or δ chain of TCRs. As reverse primers set in a C region, primers are set which match the sequences of each of Cβ, Cα, Cγ and Cδ of TCRs and have a mismatch to an extent that other C region sequences are not primed. A reverse primer of a C region is optimally made while considering the base sequence, base composition, DNA melting temperature (Tm), or presence of a self-complementary sequence, such that amplification with an adaptor primer is possible. A primer can be set in a region other than the base sequence that is different among allelic sequences in a C region sequence to uniformly amplify all alleles. A plurality of stages of nested PCR are performed in order to enhance the specificity of an amplification reaction. The length (number of bases) of a primer candidate sequence is not particularly limited for a sequence not comprising a sequence that is different among allelic sequences for each primer. However, the number of bases is 10 to 100, preferably 15 to 50, and more preferably 20 to 30. Use of such unbiased amplification is advantageous and preferred in identifying a low frequency (1/10,000 to 1/100,000 or less) gene. TCR diversity can be determined from read data that is obtained by sequencing TCR gene amplified in this manner. Large-scale high efficiency TCR repertoire analysis can now be materialized by obtaining a more detailed gene information at a clone level from conventional TCR repertoire analysis obtaining small scale information limited to V chain usage frequency or the like by applying PCR amplification on a TCR gene from a human sample and utilizing next generation sequence analysis techniques. The sequencing approach is not limited as long as a sequence of a nucleic acid sample can be determined. While any approach known in the art can be utilized, it is preferable to use next generation sequencing (NGS). Examples of next generation sequencing include, but are not limited to, pyrosequencing, sequencing by synthesis, sequencing by ligation, ion semiconductor sequencing, and the like. The obtained read data can be mapped to a reference sequence comprising V, D, and J genes to derive the unique number of reads and determine TCR diversity. One embodiment prepares a reference database to be used for each of V, D, and J gene regions. Typically, a nucleic acid sequence data set for each allele or each region published by the IMGT is used, but is not limited thereto. Any data set with a unique ID assigned to each sequence can be used. The obtained read data (including those subjected to appropriate processing such as trimming as needed) is used as the input sequence set to search for homology with a reference database for each gene region, and an alignment with the closest reference allele and the sequence thereof are recorded. In this regard, an algorithm with high tolerance for a mismatch except for C is used for homology search. When a common homology search program such as BLAST is used, setting such as shortening of the window size, reduction in mismatch penalty, or reduction in gap penalty is set for each region. The closest reference allele is selected by using a homology score, alignment length, kernel length (length of consecutively matching base sequence) and number of matching bases as indicators, which are applied in accordance with a defined order or priority. For an input sequence with determined V and J used in the present invention, a CDR3 sequence is extracted with the front of CDR3 on reference V and end of CDR3 on reference J as guides. This is translated into an amino acid sequence for use in classification of a D region. When a reference database of a D region is prepared, a combination of results of homology search and results of amino acid sequence translation is used as a classification result. In view of the above, each allele of V, D and J is assigned for each sequence in an input set. The frequency of appearance by each of V, D and J or frequency of appearance of a combination thereof is subsequently calculated in the entire input set to derive a TCR repertoire. The frequency of appearance is calculated in a unit of allele or unit of gene name depending on the precision required in classification. The latter is possible by translating each allele into a gene name. After a V region, J region, and C region are assigned to read data, matching reads can be added to calculate the number of reads detected in a sample and the ratio to the total number of reads (frequency) for each unique read (read without a same sequence). A diversity index or similarly index can be calculated with a statistical analysis software such as ESTIMATES or R (vegan) by using data such as number of samples, read type, or the number of reads. In a preferred embodiment, TCR repertoire analysis software (Repertoire Genesis Inc.) is used. A diversity index can be found from read number data for each unique read obtained in the above manner. For example, the Shannon-Weaver index (also denoted simply as Shannon index), Simpson index, normalized Shannon-Weaver index, and DE50 index can be calculated according to the following mathematical equations. N: total number of reads, ni: number of reads of the ith unique read, S: number of unique reads, S50: number of top unique reads accounting for 50% of total reads. Simpson′⁢s⁢index⁢(1-λ)1-λ=1-∑t=1S(ni(ni-1)N⁡(N-1))[Numeral⁢1]Shannon-Weaver⁢index⁢(H′)H′=-∑i=1SniN⁢ln⁢niN[Numeral⁢2]Normalized⁢Shannon-Weaver⁢index⁢(H′)H′=-∑i=1SniN⁢ln⁢niN/ln⁢N[Numeral⁢3]DE⁢50⁢(D)D=S50S[Numeral⁢4]Unique⁢50⁢(U)U=S50[Numeral⁢5] Other diversity indices that may be used include inverse Simpson index (1/λ), Morisita's β index, McIntosh's evenness index, McNaughton's dominance index, Motomura's 1/α, Fisher's diversity index, Sheldon's eH′, Pielou's evenness index, Preston's 1/σ2, Morisita's prosperity index Nβ, Pielou's H′N, and the like. DE indices including the DE50 index can be denoted by a ratio or percentage (%). Those skilled in the art can clearly and suitably understand the meaning of the denoted numerical value and practice the present invention by converting a threshold value or the like. DE indices can be calculated as the number of unique reads/number of top unique reads accounting for any ratio (1 to 99%) of total reads and can be used as a diversity index in the present invention. In addition to the DE50 index, DEX indices based on Sx(x=any numerical value from 0 to 100) instead of S50(number of top unique reads accounting for 50% of total reads) can be used as a DE index. For example, a DE30 index and DE80 index using S30(number of top unique reads accounting for 30% of total reads) and S80(number of top unique reads accounting for 80% of total reads) can also be used. A DE index can use a value that is normalized with respect to the number of reads (e.g., normalized with respect to 80000 reads, 30000 reads, 10000 reads, or the like). UniqueX indices that directly use Sx, which is a molecule of a DE index, can also be used. Examples of Unique indices include Unique30, Unique50, Unique80, and the like. (Large-Scale High Efficiency TCR Repertoire Analysis) A preferred embodiment of the present invention measures TCR diversity using large-scale high efficiency TCR repertoire analysis. As used herein, “large-scale high efficiency repertoire analysis” is described in WO 2015/075939 (the entire disclosure thereof is incorporated herein by reference as needed) and is referred to as “large-scale high efficiency TCR repertoire analysis” when targeting TCR. Large-scale high efficiency repertoire analysis is a method of quantitatively analyzing a repertoire (variable region of a T cell receptor (TCR) or B cell receptor (BCR)) of a subject by using a database, comprising (1) providing a nucleic acid sample comprising a nucleic acid sequence of the T cell receptor (TCR) or the B cell receptor (BCR) which is amplified from the subject in an unbiased manner; (2) determining the nucleic acid sequence comprised in the nucleic acid sample; and (3) calculating a frequency of appearance of each gene or a combination thereof based on the determined nucleic acid sequence to derive a repertoire of the subject, wherein (1) comprises the following steps: (1-1) synthesizing a complementary DNA by using an RNA sample derived from a target cell as a template; (1-2) synthesizing a double stranded complementary DNA by using the complementary DNA as a template; (1-3) synthesizing an adaptor-added double stranded complementary DNA by adding a common adaptor primer sequence to the double stranded complementary DNA; (1-4) performing a first PCR amplification reaction by using the adaptor-added double stranded complementary DNA, a common adaptor primer consisting of the common adaptor primer sequence, and a first TCR or BCR C region specific primer, wherein the first TCR or BCR C region specific primer is designed to comprise a sequence that is sufficiently specific to a C region of interest of the TCR or BCR and not homologous with other genetic sequences, and comprise a mismatching base between subtypes downstream when amplified; (1-5) performing a second PCR amplification reaction by using a PCR amplicon of (1-4), the common adaptor primer, and a second TCR or BCR C region specific primer, wherein the second TCR or BCR C region specific primer is designed to have a sequence that is a complete match with the TCR or BCR C region in a sequence downstream the sequence of the first TCR C region specific primer, but comprise a sequence that is not homologous with other genetic sequences, and comprise a mismatching base between subtypes downstream when amplified; and (1-6) performing a third PCR amplification reaction by using a PCR amplicon of (1-5), an added common adaptor primer in which a nucleic acid sequence of the common adaptor primer comprises a first additional adaptor nucleic acid sequence, and an adaptor-added third TCR C region specific primer in which a second additional adaptor nucleic acid sequence and a molecule identification (MID Tag) sequence are added to a third TCR or BCR C region specific sequence; wherein the third TCR C region specific primer is designed to have a sequence that is a complete match with the TCR or BCR C region in a sequence downstream to the sequence of the second TCR or BCR C region specific primer, but comprise a sequence that is not homologous with other genetic sequences, and comprise a mismatching base between subtypes downstream when amplified, the first additional adaptor nucleic acid sequence is a sequence suitable for binding to a DNA capturing bead and for an emPCR reaction, the second additional adaptor nucleic acid sequence is a sequence suitable for an emPCR reaction, and the molecule identification (MID Tag) sequence is a sequence for imparting uniqueness such that an amplicon can be identified. The specific detail of this method is described in WO 2015/075939. Those skilled in the art can practice analysis by appropriately referring to this document and the Examples of the present specification and the like. One embodiment of the present invention provides a method of using TCR diversity of a subpopulation of T cells. In one embodiment, T cells that are positive for a T cell suppression-related cell surface marker can be used as the T cells. Alternatively in another embodiment, T cells that are positive for a T cell stimulation-related cell surface marker can be used as the T cells. For example, TCR diversity of a subpopulation of T cells that are positive for one or more cell surface markers selected from the group consisting of CD8, PD-1, CD28, CD154 (CD40L), CD134 (OX40), CD137 (4-1BB), CD278 (ICOS), CD27, CD152 (CTLA-4), CD366 (TIM-3), CD223 (LAG-3), CD272 (BILA), CD226 (DNAM-1), TIGIT, and CD367 (GITR) can be used. In one embodiment, T cells are selected from the group consisting of CD8+PD1+, CD8+4-1BB+, CD8+TIM3+, CD8+OX40+, CD8+TIGIT+, and CD8+CTLA4+T cells. As used herein, “T cell stimulation-related cell surface marker” refers to a cell surface molecule that transmits a signal for activating T cells. Examples of “T cell stimulation-related cell surface marker” include, but are not limited to, CD28, CD154 (CD40L), CD134 (OX40), CD137 (4-1BB), CD278 (ICOS), CD27, and the like. As used herein, “T cell suppression-related cell surface marker” refers to a cell surface molecule that transmits a signal for suppressing T cells. Examples of “T cell suppression-related cell surface marker” include, but are not limited to, PD-1, CD152 (CTLA-4), CD366 (TIM-3), CD223 (LAG-3), CD272 (BTLA), CD226 (DNAM-1), TIGIT, CD367 (GITR), and the like. Although not wishing to be bound by any theory, high TCR diversity of a T cell subpopulation expressing such a cell surface marker can be understood as more likely to benefit from therapy with an immune checkpoint inhibitor because the subpopulation would definitely have a TCR that recognizes a surface antigen of cancer tissue. A subpopulation of T cells is, for example, a population of CD8+T cells, preferably a T cell subpopulation, which is CD8+and expresses one or more immune checkpoint molecules, such as a subpopulation of T cells that are positive for CD8+and one or more cell surface markers selected from the group consisting of PD-1, CD28, CD154 (CD40L), CD134 (OX40), CD137 (4-1BB), CD278 (ICOS), CD27, CD152 (CTLA-4), CD366 (TIM-3), CD223 (LAG-3), CD272 (BTLA), CD226 (DNAM-1), TIGIT and CD367 (GITR). In one embodiment, T cells are CD8+. Some embodiments can use TCR diversity of a subpopulation of T cells that are positive for a T cell stimulation-related cell surface marker, TCR diversity of a subpopulation of T cells that are positive for a T cell suppression-related cell surface marker, or TCR diversity of a subpopulation of T cells that are positive for a T cell stimulation-related cell surface marker and a T cell suppression-related cell surface marker. In some cases, a subpopulation of T cells can be a population of PD-1+T cells. TCR diversity can be determined for each subpopulation of T cells. In a preferred embodiment of the present invention, a subpopulation of T cells is a population of CD8+PD-1+T cells. TCR diversity of a suitable subpopulation can be used in some cases as a more accurate indicator when used as an indicator of a medical condition of a subject. A method of separating such a subpopulation of T cells is known in the art and can be performed using a suitable cell sorter (e.g., BD FACSAria III cell sorter (BD Bioscience)). Those skilled in the art can appropriately use a labeled antibody for a cell surface marker distinguishing a subpopulation to be separated. TCR diversity of a specific T cell subpopulation can be determined by TCR repertoire analysis discussed above using a nucleic acid sample that is extracted from a separated subpopulation. In a report studying TCR diversity in PBMCs by a method that does not fractionate specific cells (https://meetinglibrary.asco.org/record/126066/abstract), it is reported that a responder and a non-responder to an anti-PD-1 antibody could not be significantly distinguished by TCR analysis when a specific cell is not fractionated. As demonstrated herein, the finding that TCR diversity in a specific cell population can be used to distinguish responders from non-responders to cancer immunotherapy was unexpected. T cells obtained from any tissue can be used. T cells can be obtained from, for example, peripheral blood, tumor site, inside normal tissue, bone marrow, thymus gland, or the like. In a preferred embodiment, TCR diversity of T cells in peripheral blood of a subject is determined. Collection of T cells from peripheral blood is non-invasive and simple. TCR chains for measuring TCR are α chain, β chain, γ chain, and/or δ chain. In one embodiment, diversity of TCRα is used. In another embodiment, TCRβ is used. (Diagnosis) Responses to cancer immunotherapy can be determined based on RECIST v1.1 (New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1)). Based on a change in tumor size or the like, the effect of cancer therapy can be determined as Complete Response (CR), Partial Response (PR), Progressive Disease (PD), or Stable Disease (SD). As used herein, “responder” refers to a subject exhibiting complete response or partial response to cancer therapy. As used herein, “non-responder” refers to a subject exhibiting progressive disease or stable disease to cancer therapy. The responsiveness of a subject to cancer therapy includes a subject being a “responder” or a subject being a “non-responder”. Therefore, determination of responsiveness of a subject to cancer therapy includes determining whether a subject is a responder or a non-responder. One aspect of the present invention predicts or determines that a subject is a “responder” or a subject is a “non-responder” using TCR diversity. As for the timing of determination, prediction before the start of therapy is preferred, but the timing may be after the start of therapy. This is because determination of whether the ongoing therapy is suitable is also medically useful. Alternatively, the prognosis can be determined using TCR diversity of the present invention. For example, TCR diversity of the present invention can be used to predict that a responder becomes a non-responder, i.e., predict a recurrence. As for the timing of determination, repertoire analysis can be performed sequentially after applying cancer immunotherapy (e.g., after administration of an immune checkpoint inhibitor) to determine the prognosis from a diversity index. (Preferred Embodiments) The preferred embodiments of the present invention are disclosed hereinafter. It is understood that the embodiments provided hereinafter are provided to better facilitate the understanding of the present invention, so that the scope of the present invention should not be limited by the following description. Thus, it is apparent that those skilled in the art can refer to the descriptions herein to appropriately make modifications within the scope of the present invention. It is also understood that the following embodiments of the present invention can be used alone or as a combination. (Indicator for Responsiveness) In one aspect, the present invention provides a method of using T cell receptor (TCR) diversity of T cells of a subject as an indicator of responsiveness of the subject to cancer immunotherapy or diagnosis of the responsiveness. In this regard, TCR diversity can be provided as a diversity index. T cells can be CD8+PD-1+in peripheral blood. In another aspect, the present invention provides a method of diagnosing responsiveness of a subject to cancer immunotherapy, comprising: measuring TCR diversity of T cells of the subject in vitro; and if the TCR diversity is high, determining the subject as having good responsiveness to cancer immunotherapy. Alternatively, if the TCR diversity is low, it is also possible to determine the subject as having poor responsiveness to cancer therapy. T cells can be CD8+PD-1+T cells in peripheral blood. In yet another aspect, the present invention is a method of diagnosing responsiveness of a subject to cancer immunotherapy, comprising: obtaining a peripheral blood sample from the subject; measuring TCR diversity of T cells in peripheral blood of the subject by a method comprising large-scale high efficiency TCR repertoire analysis; and if the TCR diversity is high, determining the subject as having good responsiveness to cancer immunotherapy. Alternatively, if the TCR diversity is low, it is also possible to determine the subject as having poor responsiveness to cancer therapy. T cells can be CD8+PD-1+T cells in peripheral blood. Although not wishing to be bound by any theory, the inventors have found that high TCR diversity of a subject, i.e., high value of a diversity index, correlates with better responsiveness to cancer immunotherapy. In particular, TCR diversity of CD8+PD-1+T cells is useful as an advantageous indicator of responsiveness to therapy with an immune checkpoint inhibitor, especially PD-1 inhibitor (e.g., nivolumab or pembrolizumab). High Shannon-Weaver index, inverse Simpson index, Simpson index, normalized Shannon-Weaver index, DEX index (X is 0 to 100, e.g., DE30 index, DE50 index, DE80 index, or the like), and/or UniqueX index (X is 0 to 100, e.g., Unique30 index, Unique50 index, Unique80 index, or the like) of a subject can be used as an indicator of better responsiveness to cancer immunotherapy. Although not wishing to be bound by any therapy, it is understood in view of the results in the Examples of the present invention that inhibition of immune checkpoints does not achieve much antitumor effect when a population of cancer attacking CD8+T cells (killer T cells or cytotoxic T cells (CTL)) have few or no cells with TCRs recognizing a surface antigen of cancer tissue. A subject with high TCR diversity can be understood as more likely to benefit from therapy with an immune checkpoint inhibitor because the subject would definitely have cells with TCR that recognizes a surface antigen of cancer tissue. Furthermore, cancer is known to be comprised of multiple cell populations instead of a uniform cell population. It is understood that such diverse cancer cells express different cancer antigens for each cell. Thus, in view of the results of the Examples in the present invention, it is expected that suppression of cancer cells requires immune cells, which recognize more diverse antigens. It can be understood that an immune checkpoint inhibitor is likely to exert an effect on a patient with diverse T cells. One embodiment of the present invention determines that a subject is a responder, not a responder, a non-responder, or not a non-responder to any cancer immunotherapy described herein, based on a value of the Shannon-Weaver index, inverse Simpson index, Simpson index, normalized Shannon-Weaver index, DEX index (X is 0 to 100, e.g., DE30 index, DE50 index, DE80 index, or the like), and/or UniqueX index (X is 0 to 100, e.g., Unique30 index, Unique50 index, Unique80 index, or the like) of TCR of the subject. In one embodiment of the present invention, a numerical value based on multiple analyses described herein can be used as a threshold value. The threshold values described herein are only exemplifications. Those skilled in the art can determine and use other threshold values based on other results of determination in a subject population. A method of determining a threshold value is disclosed herein. The method of the present invention may comprise a step of determining a threshold value or use a threshold value that is determined in advance in accordance with such a method. A threshold value of a diversity index can be set by calculating a diversity index of a certain number of responders and non-responders and determining a numerical value that differentiates the responders from non-responders. Examples of a method of determining a differentiating numerical value include use of a minimum value of a responder population (ensures that a responder is determined as a responder; sensitivity is 100%), use of a maximum value of a non-responder population (non-responder is not determined as a responder; specificity is 100%), and use of ROC analysis (maximizes the validity of determination by balancing sensitivity and specificity). As another approach, a reference range (reference value) can be found from numerical values of non-responder or responder groups to differentiate by an abnormal value exceeding such reference range. A reference range can be, for example, mean value±standard deviation (SD), mean value±2 SD, or the like, where the upper limit or the lower limit of the reference range can be the reference value. In some cases, a threshold value can be determined by calculating mean value±SD, mean value±2 SD, or the like from the numerical values of the non-responder group. In some cases, a threshold value can be determined by calculating mean value−SD, mean value−2 SD, or the like from the numerical values of the responder group. When a diversity index or threshold value is affected by the number of reads, the number of reads is normalized to set a threshold value, or a threshold value in a different number of reads is calculated by resampling, and a prediction formula (which can be represented, for example, by a general formula Y=aX{circumflex over ( )}b, wherein number of reads: X and threshold value: Y) derived from regression analysis of the number of reads and threshold value or the like is used to determine a threshold value. In one embodiment, a Shannon-Weaver index for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 3.2 to 4.4, and preferably about 3.3 to about 4.2. A specific threshold value can be for example about 3.3, about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4.0, about 4.1, about 4.2 or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, an inverse Simpson index for TCRα of CD8+TD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be set by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 9 to about 19, and preferably about 10 to about 18. A specific threshold value can be for example about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, or the like (any other specific value between these specific numerical values may be used). In one embodiment, a Simpson index for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 0.86 to about 0.96, and preferably about 0.88 to about 0.94. A specific threshold value can be for example about 0.86, about 0.87, about 0.88, about 0.89, about 0.90, about 0.91, about 0.92, about 0.93, about 0.94, about 0.95, about 0.96, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a normalized Shannon-Weaver index for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 0.41 to about 0.51, and preferably about 0.42 to about 0.49. A specific threshold value can be for example about 0.42, about 0.43, about 0.44, about 0.45, about 0.46, about 0.47, about 0.48, about 0.49, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a DE50 index for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 0.0007 to about 0.0015, and preferably about 0.0008 to about 0.0014, about 0.0009 to about 0.0013, about 0.0010 to about 0.0011, or the like as appropriate. A specific threshold value can be for example about 0.0007, about 0.0008, about 0.0009, about 0.0010, about 0.0011, about 0.0012, about 0.0013, about 0.0014, about 0.0015, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a Shannon-Weaver index for TCRβ of CD8+TD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 3.2 to about 4.3, and preferably about 3.4 to about 4.1. A specific numerical value can be for example about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4.0, about 4.1, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, an inverse Simpson index for TCRβ of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold value can be set in the range of about 8 to 32, and preferably about 10 to about 30. A specific numerical value can be for example about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a Simpson index for TCRβ of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 0.90 to about 0.96, and preferably about 0.92 to about 0.95. A specific threshold value can be for example about 0.90, about 0.91, about 0.92, about 0.93, about 0.94, about 0.95, about 0.96, or the like (any other specific numerical value between these specific numerical values may be used) In one embodiment, a normalized Shannon-Weaver index for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold value can be set in the range of about 0.37 to about 0.48, and preferably about 0.38 to about 0.47. A specific numerical value can be for example about 0.38, about 0.39, about 0.40, about 0.41, about 0.42, about 0.43, about 0.44, about 0.45, about 0.46, about 0.47, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a DE50 index for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold value can be set in the range of about 0.0004 to about 0.0012, and preferably about 0.0005 to about 0.0012. A specific numerical value can be for example about 0.0005, about 0.0006, about 0.0007, about 0.0008, about 0.0009, about 0.0010, about 0.0011, about 0.0012, or the like (any other specific numerical value between these specific numerical values may be used). Thus, the method of diagnosing responsiveness of a subject to cancer immunotherapy of the present invention can comprise a step of identifying a threshold value for determining that the subject has good responsiveness to cancer immunotherapy. Such an identification method can be identified by performing a clinical trial exemplified in the Examples or the like, calculating a diversity index, and optionally applying statistical processing. A value of an index described above, which was calculated by measuring TCR diversity of a subject, can be appropriately rounded up or down (e.g., for DE50, rounding up or down the 5th decimal) and comparing with a threshold value. Two or one significant figures can be used while considering the detection limit. It is possible to use a threshold value which is calculated from ROC analysis and numerical value equal to or greater than mean value-SD or mean value-2 SD of a responder group as the lower limit value from the results of the Examples in the present specification. For example, the following are mean value-SD of a responder group for some of the diversity indices in the Examples herein.Shannon-Weaver index for TCRα: 3.37Inverse Simpson index for TCRα: 9.83Normalized Shannon-Weaver index for TCRα: 0.422DE50 index for TCRα: 0.0013605Shannon-Weaver index for TCRβ: 3.69Inverse Simpson index for TCRβ: 15.84Normalized Shannon-Weaver index for TCRβ: 0.433DE50 index for TCRβ: 0.0009545 The following are the mean value-2 SD of a responder group.Shannon-Weaver index for TCRα: 2.66Inverse Simpson index for TCRα: −4.19Normalized Shannon-Weaver index for TCRα: 0.366DE50 index for TCRα: 0.0007697Shannon-Weaver index for TCRβ: 3.03Inverse Simpson index for TCRβ: 1.43Normalized Shannon-Weaver index for TCRβ: 0.385DE50 index for TCRβ: 0.0005064. ROC analysis (Receiver Operating Characteristic analysis) can be used to set a threshold value (cutoff value). A cutoff value determined by using ROC analysis can be utilized to, for example, predict an effect prior to therapy in an anti-PD-1 antibody therapy patient. In ROC analysis, ROC curves are created by plotting the positive rate to the vertical axis as sensitivity and false-positive rate (1-specificity) to the horizontal axis when changing the cutoff value. Methods of setting a cutoff value include a method of using a point where the distance to the top left corner of a ROC curve is minimized as a cutoff value, and a method of calculating the furthest point from a diagonal dashed line where the area under the curve (AUC) is 0.500 on a ROC curve, i.e., (sensitivity+specificity−1) and using the point at the maximum value thereof (Youden index) as the cutoff value.FIG.7shows ROC curves in each diversity index described herein. Table 12 shows cutoff values calculated from Youden index based on Example 1 herein. Such exemplified value can be used as a threshold value. As used herein, “sensitivity” refers to the probability of correctly determining what should be determined as positive as positive, with high sensitivity related to reduced false-positive. High sensitivity is useful in rule-out diagnosis. As used herein, “specificity” refers to the probability of correctly determining what is negative as negative, with high specificity related to reduced false-positive. High sensitivity is useful in rule-in diagnosis. For example, about 3.7 can be used as the cutoff value of a Shannon-Weaver index for TCRα, about 13 can be used as the cutoff value of an Inverse Simpson index for TCRα, about 0.43 can be used as the cutoff value of a normalized Shannon-Weaver index for TCRα, about 0.0012 can be used as the cutoff value of a DE50 index for TCRα, 3.8 can be used as the cutoff value of a Shannon-Weaver index for TCRβ, about 17 can be used as the cutoff value of an inverse Simpson index for TCRβ, about 0.42 can be used as the cutoff value of a normalized Shannon-Weaver index for TCRβ, and about 0.0007 can be used as the cutoff value of a DE50 index for TCRβ. However, these exemplified values are not limited. Those skilled in the art can adjust a threshold value in accordance with desired sensitivity and/or specificity based on ROC analysis. The values are also not limited to those exemplified in the Examples herein. A cutoff value can be determined by performing ROC analysis using additional information on subject. One embodiment can predict that a high therapeutic effect cannot be expected by an anti-PD-1 antibody at less than these cutoff values. A diversity index can be affected by the sampled size. In other words, some TCR diversity indices vary depending on the number of sequencing reads. When using such a diversity index, a more accurate evaluation of responsiveness is possible by normalization corresponding to a specific number of reads. It is understood that Shannon, Simpson, and inverse Simpson indices have a mostly constant value regardless of the number of reads, and are hardly affected especially at the number of reads of 10000 reads or greater, which is a level in actual analysis. In such a case, normalization of an index is not necessarily required. DE indices generally have a tendency to decrease with an increase in the number of reads. When there is a large difference in the number of sequencing reads between subjects or between a subject and a control subject (e.g., when there is a variation of 10-fold or greater), it is understood that a more accurate evaluation of responsiveness is possible using a DE index which is normalized with respect to a certain number of reads. As one method of normalization, a DE index can be approximated to those with a linear relationship with respect to both logarithmic axes and the number of reads. An index can be normalized based on this relationship. Therefore, a DE index at a certain number of reads can be compared to a threshold value that has been adjusted in accordance with the linear relationship to evaluate responsiveness. As one example of a linear relationship, the linear relationship between the number of reads and threshold value of a DE index described in the Examples herein is represented by the following equations: DE50,TCRα:y=1892.344x{circumflex over ( )}(−0.8239) DE50,TCRβ:y=993.116x{circumflex over ( )}(−0.8072) DE30,TCRα:y=260.0x{circumflex over ( )}(−0.7008) DE30,TCRβ:y=476.4x{circumflex over ( )}(−0.8032) DE80,TCRα:y=4275.6x{circumflex over ( )}(−0.7905) DE80,TCRβ:y=6151.8x{circumflex over ( )}(−0.8406)  [Numeral 6] wherein y is the threshold value and x is the number of read. Those skilled in the art can perform normalization using this relationship. In other words, a DE index obtained based on the number of reads x can be compared to y to evaluate responsiveness. Those skilled in the art can also newly derive a linear relationship from multiple sequencing results for use in normalization. The linear relationship of a threshold value can be considered as having a width. For example, this can be represented as a band when a value of a diversity index is the vertical axis and the number of reads is the horizontal axis. An index can be used such that a value at or above the upper limit thereof is determined as a responder, and a value at or below the lower limit is determined as a non-responder, and administration can be determined by the discretion of a physician if the value is in the middle. For example, the 95% confidence interval of a fitting curve can be used as the width of variation. An example is shown in the Examples herein. Such calculation can maximize the specificity or sensitivity. As another normalization method, a certain number of reads can be resampled from reads obtained by sequencing to calculate a diversity index based on the resampled reads for normalization. Resampling can be performed by randomly obtaining reads from the obtained reads. Resampling can also be performed multiple times, in which case a representative value of a diversity index for each trial (median, mean, or the like) can be used as a normalized diversity index. The reference number of reads for normalization is not limited, but can be for example 1000, 10000, 20000, 40000, 80000, 100000, 200000, or the like (any other specific numerical value between these specific numerical values may be used). In some embodiments, a DE50 index that is normalized with respect to 30000 reads is used. In one embodiment, a Shannon-Weaver index, which has been normalized with respect to 30000 reads, for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 2.8 to 4.1, and preferably about 3.9 to about 4.1. A specific threshold value can be for example about 2.8, about 2.9, about 3.0, about 3.1, about 3.2, about 3.3, about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4.0, about 4.1, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, an inverse Simpson index, which has been normalized with respect to 30000 reads, for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 8 to about 16, and preferably about 13 to about 15. A specific threshold value can be for example about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a Simpson index, which has been normalized with respect to 30000 reads, for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 0.89 to about 0.94, and preferably about 0.92 to about 0.94. A specific threshold value can be for example about 0.89, about 0.90, about 0.91, about 0.92, about 0.93, about 0.94, or the like (any other specific value between these specific numerical values may be used). In one embodiment, a normalized Shannon-Weaver index, which has been normalized with respect to 30000 reads, for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 0.41 to about 0.54, and preferably about 0.50 to about 0.52. A specific threshold value can be for example about 0.41, about 0.42, about 0.43, about 0.44, about 0.45, about 0.46, about 0.47, about 0.48, about 0.49, about 0.50, about 0.51, about 0.52, about 0.53, about 0.54, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a DE50 index (%), which has been normalized with respect to 30000 reads, for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be determined in the range of about 0.36 to about 0.40 or the like as appropriate. A specific threshold value can be for example about 0.36, about 0.37, about 0.38, about 0.39, about 0.40, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a Shannon-Weaver index, which has been normalized with respect to 30000 reads, for TCRβ of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 3.2 to about 4.0, and preferably about 3.7 to about 3.9. A specific numerical value can be for example about 3.2, about 3.3, about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4.0, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, an inverse Simpson index, which has been normalized with respect to 30000 reads, for TCR of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold value can be set in the range of about 10 to 23, and preferably about 12 to about 22. A specific numerical value can be for example about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a Simpson index, which has been normalized with respect to 30000 reads, for TCRβ of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold can be determined by a forward-looking or backward looking clinical trial on subject patients. In one specific embodiment, a threshold value can be set in the range of about 0.90 to about 0.97, and preferably about 0.92 to about 0.96. A specific threshold value can be for example about 0.90, about 0.91, about 0.92, about 0.93, about 0.94, about 0.95, about 0.96, about 0.97, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a normalized Shannon-Weaver index, which has been normalized with respect to 30000 reads, for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold value can be set in the range of about 0.42 to about 0.53, and preferably about 0.47 to about 0.52. A specific numerical value can be for example about 0.42, about 0.43, about 0.44, about 0.45, about 0.46, about 0.47, about 0.48, about 0.49, about 0.50, about 0.51, about 0.52, about 0.53, or the like (any other specific numerical value between these specific numerical values may be used). In one embodiment, a DE50 index (%), which has been normalized with respect to 30000 reads, for TCRα of CD8+PD-1+T cells of a subject equal to or greater than a threshold value indicates that the subject is a responder, and a threshold value can be set in the range of about 0.22 to about 0.26, and preferably about 0.23 to about 0.25. A specific numerical value can be for example about 0.22, about 0.23, about 0.24, about 0.25, about 0.26, or the like (any other specific numerical value between these specific numerical values may be used). It is possible to use a threshold value calculated based on ROC analysis for a normalized diversity index as discussed above. For example, a threshold value exemplified herein can be used for a diversity index that has been normalized with respect to 30000 reads. For example, about 3.9 can be used as the cutoff value of a Shannon-Weaver index, which has been normalized with respect to 30000 reads, for TCRα, about 14 can be used as the cutoff value of an Inverse Simpson index, which has been normalized with respect to 30000 reads, for TCRα, about 0.92 can be used as the cutoff value of a Simpson index, which has been normalized with respect to 30000 reads, for TCRα, about 0.51 can be used as the cutoff value of a normalized Shannon-Weaver index, which has been normalized with respect to 30000 reads, for TCRα, about 0.39 can be used as the cutoff value of a DE50 index, which has been normalized with respect to 30000 reads, for TCRα, 3.8 can be used as the cutoff value of a Shannon-Weaver index, which has been normalized with respect to 30000 reads, for TCRβ, about 22 can be used as the cutoff value of an inverse Simpson index, which has been normalized with respect to 30000 reads, for TCRβ, about 0.95 can be used as the cutoff value of a Simpson index, which has been normalized with respect to 30000 reads, for TCRβ, about 0.51 can be used as the cutoff value of a normalized Shannon-Weaver index, which has been normalized with respect to 30000 reads, for TCRβ, and about 0.24 can be used as the cutoff value of a DE50 index, which has been normalized with respect to 30000 reads, for TCRβ. A cutoff value can be selected after adjusting for the objective. For example, a cutoff value can be determined in accordance with an objective such as (i) rule out non-responders (from the viewpoint of social security cost) or (ii) eliminate missing a responder (from the viewpoint of a physician/therapy). (i) can be accomplished by using a higher value than the maximum line of non-responder as a cutoff value. (ii) can be accomplished by using a lower value than the minimum line of responder as a cutoff value. Those skilled in the art can determine these values based on a diversity index of a subject population. A threshold value can also be set based on an example of a maximum value or minimum value of a diversity index exhibited by a non-responder or responder, which is described herein. In general, biological markers yield data with variance, so that two groups being compared can rarely be separated clearly. Generally, if a threshold value of a normal value can be determined from a large amount of data to distinguish whether a value is an abnormal value based thereupon, a marker is usable. For example, highly positive PD-1 is used as a marker for application of Keytruda (PD-L1 antibody), but the actual response rate is about 50%. While there is sufficient value without being able to predict 100% as a marker, a marker that can separate two groups at 100% for both sensitivity and specificity (e.g., DE50 index of TCR diversity) can be extremely advantageous. In a preferred embodiment, TCR is TCRα. In another preferred embodiment, TCR is TCRβ. TCRβ can be more preferable. Although not wishing to be bound by any theory, this is because a diversity index for TCRβ appears to lack overlap in the numerical values exhibited by responsive and non-responsive subjects. However, the present invention is not limited thereto, where TCR may be TCRα. Although not wishing to be bound by any theory, this is because it is demonstrated that subjects can be distinguished by using a DE50 index. In one embodiment, diversity utilized in the present invention can be calculated using the steps of isolating CD8+PD-1+T cells from a peripheral blood sample of a subject; and measuring, determining, or calculating TCR diversity of the CD8+PD-1+T cells. One embodiment of the present invention is a method of diagnosing responsiveness of a subject to cancer immunotherapy, comprising if the TCR diversity is high, determining the subject as having good responsiveness to cancer immunotherapy. Preferably, TCR diversity calculated in this regard advantageously uses large-scale high efficiency TCR repertoire analysis (WO 2015/075939) disclosed in detail herein. Although not wishing to be bound by any theory, when other TCR repertoire analysis is used, some of the unique reads that can be detected by large-scale high efficiency TCR repertoire analysis cannot be detected. For this reason, diversity indices calculated by large-scale high efficiency TCR repertoire analysis are more precise and more accurately reflect the status of a subject. Although not wishing to be bound by any theory, it is understood that diversity indices in large-scale high efficiency TCR repertoire analysis can indeed clearly distinguish responders from non-responders, but conventional repertoire analysis other than large-scale high efficiency TCR repertoire analysis does not sufficiently distinguish responders from non-responders. Therefore, use of TCR diversity measured using large-scale high efficiency repertoire analysis can result in a more accurate evaluation result than conventional analysis. When TCR diversity of T cells of a subject is high after measuring the TCR diversity by a method comprising large-scale high efficiency TCR repertoire analysis in the method of diagnosing responsiveness of the subject to cancer immunotherapy of the present invention, the subject is determined to have good responsiveness to cancer immunotherapy. Whether TCR diversity is high can be determined relatively or by determining whether diversity is high compared to a predetermined threshold value of a diversity index (e.g., those described herein). If a diversity index is high, a subject can be determined to have or is responsiveness to cancer immunotherapy and appropriately undergo therapy thereafter as needed. T cells that can be used can be one or more T cells of any type described herein, preferably CD8+PD-1+T cells in peripheral blood. Still another embodiment of the present invention is a method of diagnosing responsiveness of a subject to cancer immunotherapy to treat cancer of the subject, comprising: measuring TCR diversity of T cells of the subject; and if the TCR diversity is higher than a reference value, applying the cancer immunotherapy to the subject (so-called companion diagnosis or companion therapy). A reference value or threshold value of TCR diversity can be appropriately determined by those skilled in the art based on the descriptions herein. Specific numerical values of diversity indices exemplified herein can be appropriately used. T cells that can be used can be one or more T cells of any type described herein, preferably CD8+TD-1+T cells in peripheral blood. (Companion Application of Immune Checkpoint Inhibitor) Still another aspect of the present invention provides a composition for treating cancer in a subject with high TCR diversity of T cells, comprising an immune checkpoint inhibitor. The inventors found that such an immune checkpoint inhibitor is advantageously administered to a subject with high TCR diversity of T cells. In addition, a subject with low TCR diversity of T cells can be determined as a non-responder, and a decision can made to not administer an immune checkpoint inhibitor, or suspend or discontinue administration. T cells on which TCR diversity is measured can be one or more T cells of any type described herein, preferably CD8+PD-1+T cells in peripheral blood. The composition of the present invention is preferably a pharmaceutical composition. Examples of an immune checkpoint inhibitor contained as an active ingredient thereof include PD-1 inhibitors. Examples of PD-1 inhibitors include anti-PD-1 antibodies nivolumab and pembrolizumab. A composition can be formulated in any dosage form such as aerosol, liquid agent, extract, elixir, capsule, granule, pill, ointment, powder, tablet, solution, suspension, or emulsion. A composition may comprise any pharmaceutically acceptable additive and/or excipient that are known in the art. The composition of the present invention can be administered through any suitable route determined by those skilled in the art. Examples thereof include, but are not limited to, intravenous injection, intravenous drip, oral administration, parenteral administration, transdermal administration, and the like. One embodiment provides a composition for treating cancer in a subject with a high Shannon index, Simpson index, normalized Shannon index, or DE50 index for TCR of T cells. A preferred embodiment provides a composition for treating cancer in a subject with a high DE50 index for TCR of T cells. A composition for treating cancer in a patient with a DE50 index, which is normalized with respect to 30000 reads, of 0.39% or greater for TCRα of CD8+PD-1+T cells in peripheral blood is provided. A composition for treating cancer in a patient with a DE50 index, which is normalized with respect to 30000 reads, of 0.24% or greater for TCRβ of CD8+PD-1+T cells in peripheral blood is provided. (Novel Application of Large-Scale High Efficiency TCR Repertoire Analysis) One aspect provides a method of using diversity of a repertoire determined by a method comprising large-scale high efficiency TCR repertoire analysis as an indicator of responsiveness of a subject to therapy. This approach determines the diversity of a repertoire by amplifying a TCR gene or BCR gene including all isotype and subtype genes with a set of primers consisting of one type of forward primer and one type of reverse primer, without changing the frequency of presence. As described herein and in WO 2015/075939, this primer design is advantageous for unbiased amplification. When another repertoire analysis is used, some of the unique reads that can be detected by large-scale high efficiency repertoire analysis cannot be detected. For this reason, diversity indices calculated by large-scale high efficiency repertoire analysis are more precise and more accurately reflect the status of a subject. Although not wishing to be bound by any theory, it is understood that diversity indices in large-scale high efficiency repertoire analysis can indeed clearly distinguish responders from non-responders to therapy, but conventional repertoire analysis other than large-scale high efficiency repertoire analysis does not sufficiently distinguish responders from non-responders to the therapy. In one embodiment, target therapy is therapy associated with immune responses. In another preferred embodiment, the repertoire analysis used is TCR repertoire analysis. (Note) As used herein, “or” is used when “at least one or more” of the listed matters in the sentence can be employed. When explicitly described herein as “within the range” of “two values”, the range also includes the two values themselves. Reference literatures such as scientific literatures, patents, and patent applications cited herein are incorporated herein by reference to the same extent that the entirety of each document is specifically described. The present invention has been described while showing preferred embodiments to facilitate understanding. While the present invention is described hereinafter based on Examples, the above descriptions and the following Examples are for the sole purpose of exemplification, but not limitation of the present invention. Thus, the scope of the present invention is not limited to the embodiments and Examples that are specifically described herein and is limited only by the scope of claims. EXAMPLES The Examples are described hereinafter. In the following Examples, all experiments were conducted in accordance with the guidelines approved by the Independent Ethics Committee of the Hyogo College of Medicine as needed. The experiments were also conducted in compliance with the guidelines in “Ethical Guidelines for Medical and Health Research Involving Human Subjects” prepared by the Ministry of Education, Culture, Sports, Science and Technology, the Ministry of Health, Labour and Welfare, and the Ministry of Economy, Trade and Industry (Dec. 22, 2014, 26 Notice No. 475 of Ministry of Education, Culture, Sports, Science and Technology, Research Promotion Bureau, Notice No. 1222-1 of Ministry of Health, Labour and Welfare, Notice No. 1222-1 of Health Policy Bureau). The experiments were also conducted in compliance with the ethical guidelines for human genetic analysis study. The experiments were conducted with approval after a review by the Independent Ethics Committee of the Hyogo College of Medicine. Reagents that are specifically described in the Examples were used, but the reagents can be substituted with an equivalent product from another manufacturer (Sigma-Aldrich, Wako Pure Chemical, Nacalai Tesque, R & D Systems, USCN Life Science INC, or the like). Example 1: TCR Diversity of Patients Receiving Anti-PD-1 Antibody Therapy (1. Materials and Methods) 1.1 Separation of Peripheral Blood Mononuclear Cells (PBMC) 8 mL of whole blood was collected into a heparin containing blood collection tube prior to the start of anti-PD-1 antibody (nivolumab, Opdivo) therapy in 12 lung cancer patients. PBMCs were separated from the whole blood by Ficoll-Hypaque density gradient centrifugation. The cells were counted with a hemocytometer. The isolated PBMCs were directly subjected to immunocytochemistry, or suspended in a cell cryopreservation solution STEM-CELLBANKER and stored in liquid nitrogen. 1.2. Double Staining with Anti-CD8 Antibody and Anti-PD-1 Antibody PBMCs were immunostained according to the following procedure.1. To remove the STEM-CELLBANKER, the cryopreserved cells were washed twice after the cells were suspended in Stain Buffer (PBS, 0.1% BSA, 0.1% Sodium Azide) and centrifuged, and the supernatant was disposed.2. Fresh or stored PBMCs were suspended in Stain buffer at 1×106/tube, and centrifuged at 4° C. for 5 minutes at 1000 rpm.3. The cells were stained while shaded at room temperature for 30 minutes in 100 μL of antibody diluent (5 μl/test anti-human CD8 antibody, 2.0 μg anti-human PD-1 antibody).4. After antibody staining, the cells washed twice after the cells were suspended in 2 mL of Stain buffer and centrifuged at 4° C. for 5 minutes at 1000 rpm, and then the supernatant was disposed.5. After washing, the cells were suspended in 100 μL of Stain buffer. 5 μL of 7-AAD was added and reacted while shading for 10 minutes at room temperature.6. The cells were supplemented with 500 μL of Stain buffer and filtered.7. The stained cells were sorted to separate a 7AAD-CD8+PD-1+cell population using a BD FACSAria III cell sorter (BD Bioscience).8. The sorted cells were transferred into a 1.5 mL Eppendorf tube and centrifuged at 4° C. for 5 minutes at 1000 rpm.9. After removing the Stain buffer while keeping 50 μL of the supernatant, 750 μL of TRIzol LS reagent (Invitrogen) was added and pipetted to dissolve the cells.10. The TRIzol solution to which the cells were dissolved was supplemented with 200 μL of DEPC Water to be adjusted to 1000 μL. The mixture was admixed with a vortex, and cryopreserved at −80° C. 1.3. RNA Extraction Whole RNA was extracted and purified from cells dissolved into the TRIzol LS reagent using RNeasy Plus Universal Mini Kit (QIAGEN). The purified RNA was quantified using Nanodrop absorption spectrometer (Thermo Scientific) or TapeStation 2200 (Agilent). 1.4. Synthesis of Complementary DNA and Double Stranded Complementary DNA In order to synthesize a complementary DNA using the extracted RNA, 1.25 μL of BSL-18E primer (Table 1) and 3.75 μL of RNA were admixed and annealed for 8 minutes at 70° C. TABLE 1Primer sequencesPrimersSequencesBSL-18EAAAGCGGCCGCATGCTTTTTTTTTTTTTTTTTTVN(SEQ ID NO: 1)P20EATAATACGACTCCGAATTCCC (SEQ ID NO: 2)P10EAGGGAATTCGG (SEQ ID NO: 3)P22EA-ST1-GTCTCGTGGGCTCGGAGATGTGTATAAGAGACAGCTAARTACGACTCCGAATTCCC (SEQ ID NO: 4)CA1TGTTGAAGGCGTTTGCACATGCA (SEQ ID NO: 5)CA2GTGCATAGACCTCATGTCTAGCA (SEQ ID NO: 6)CA-ST1-RTCGTCGGCAGCGTCAGATGTGTATAAGAGACAGGAGGGTCAGGGTTCTGGA (SEQ ID NO: 7)CB1GAACTGGACTTGACAGCGGAACT (SEQ ID NO: 8)CB2AGGCAGTATCTGGAGTCATTGAG (SEQ ID NO: 9)CB-ST1-RTCGTCGGCAGCGTCAGATGTGTATAAGAGACAGGCTCAAACACAGCGACCTC (SEQ ID NO: 10) After cooling on ice, a reverse transcription reaction was performed in the presence of an RNase inhibitor (RNAsin) to synthesize a complementary DNA with the following composition. TABLE 2Synthesis of cDNAReagentContent (μL)Final concentrationRNA solution3.75200 μM BSL-18E1.2550 μMTotal 570° C., 8 minutes5x First strand buffer250 mM Tris-HCl, pH8.3,75 mM KCl, 3 mM MgCl20.1M DTT110 mM10 mM dNTPs0.5500 μMRNasin (Promega)0.52 U/μLSuperscript III ™, 200 U/μL120 U/μL(Invitrogen)Total 1050° C., 1 hour The complementary DNA was subsequently incubated for 2 hours at 16° C. in the following double-stranded DNA synthesis buffer in the presence ofE. coliDNA polymerase I,E. coliDNA Ligase, and RNase H to synthesize a double stranded complementary DNA. Furthermore, T4 DNA polymerase was reacted for 5 minutes at 16° C. to perform a 5′ terminal blunting reaction. TABLE 3Synthesis of double-stranded complementary DNAReagentContent (μL)Final concentrationComplementary DNA reaction solution9Sterilized water46.55x Second strand buffer1525 mM Tris-HCl, pH7.5, 100 mMKCl, 5 mM MgCl2,10 mM (NH4)SO4, 0.15 mMβ·NAD+, 1.2 mM DTT10 mM dNTPs1.50.2 mME. coliDNA ligase, 10 U/μL (Invitrogen)0.50.067 U/μLE. coliDNA polymerase, 10 U/μL20.27 U/μL(Invitrogen)RNaseH, 2 U/μL (Invitrogen)0.50.013 U/μLTotal 75 μL16° C., 2 hoursT4 DNA polymerase, 5 U/μL10.067 U/μL(Invitrogen)16° C., 5 minutes A double stranded DNA, after column purification with a MiniElute Reaction Cleanup Kit (QIAGEN), was incubated all night at 16° C. in the presence of a P20EA/10EA adaptor (Table 4) and T4 ligase in the following T4 ligase buffer for a ligation reaction. TABLE 4Adaptor adding reactionContentReagent(μL)Final concentrationDouble-stranded complementary14DNA solution1 × Quick Ligation Reaction Buffer2566 mM Tris-HCl, pH7.6,(NEB)10 mM MgCl2, 1 mMATP, 7.5% PEG6000,1 mM DTT50 μM P20EA/10EA adaptor1010 μMQuick Ligase, 2,000 U/μL (NEB)140 U/μLTotal 25Room temperature,30 minutes An adaptor added double stranded DNA purified by a column as discussed above was digested by a Not I restriction enzyme (50 U/μL, Takara) with the following composition in order to remove an adaptor added to the 3′ terminal. TABLE 5Restriction enzyme treatmentReagentContent (μL)Final concentrationDouble-stranded34complementaryDNA solution10 × H restriction550 mM Tris-HCl, pH7.5, 10 mMenzyme bufferMgCl2, 1 mM, 1 mMDTT, 100 mM NaCl0.1% BSA50.01%0.1% Triton X-10050.01%Not I, 50 U/μL (Takara)11 U/μLTotal 5037° C., 2 hours The digestion time can be appropriately changed. 1.5. PCR 1stPCR amplification was performed using a C region specific primer (CG1, CK1, or CL1) and a common adaptor primer P20EA shown in Table 2 from a double stranded complementary DNA. PCR was performed for 20 cycles, where each cycle consisted of 20 seconds at 95° C., 30 seconds at 60° C., and one minute at 72° C. with the following composition. TABLE 61stPCR amplification reaction compositionContent (μL)Final concentration2x KAPA HiFi Ready mix (KAPA10Biosystems)10 mM P20EA primer0.2100 nM10 mM CA1 or CA2 primer0.2100 nMDouble-stranded complementary2DNASterilized water7.6 A 1stPCR amplicon was then used to perform 2ndPCR with the reaction composition shown below using a P20EA primer and a C region specific primer (CA2 or CB2). 20 cycles of PCR were performed, where each cycle consisted of 20 seconds at 95° C., 30 seconds at 60° C., and one minute at 72° C. with the following composition. TABLE 72ndPCR amplification reaction compositionContent (μL)Final concentration2x KAPA HiFi Ready mix10(KAPA Biosystems)10 mM P20EA primer1500 nM10 mM CA2 or CB2 primer1500 nM1stPCR amplicon2Sterilized water6 10 μL of the resulting 2nd PCR amplicon was purified using Agencourt AMPure XP (Beckman Coulter). Tag-adding PCR was performed with 5 μL of 30 μL of the final eluate as the template. The amplification used P22EA-ST1-R and mCG-ST1-R, mCK-ST1-R or mCL-ST1-R primers shown inFIG.1. 20 cycles of PCR were performed, where each cycle consisted of 20 seconds at 95° C., 30 seconds at 60° C., and one minute at 72° C. TABLE 8Tag PCR amplification reaction compositionContentFinal(μL)concentration2x KAPA HiFi Ready mix (KAPA Biosystems)1010 mM P22EA-ST1-R primer0.4200 nM10 mM CA-ST1-R or mCB-ST1-R primer0.4200 nM2ndPCR purification product5Sterilized water4.2 10 μL of the resulting Tag PCR amplicon was purified using Agencourt AMPure XP (Beckman Coulter). INDEX was added using Nextera XT Index Kit v2 SetA (Illumina) with 2 μL of 30 μL of the final eluate as the template. 12 cycles of PCR were performed, where each cycle consisted of 20 seconds at 95° C., 30 seconds at 55° C., and 30 seconds at 72° C. To confirm PCR amplification, 10 μL of amplicon was examined by 2% agarose gel electrophoresis. The resulting INDEX-ed PCR amplicon was quantified using Qubit 2.0 Fluorometer (Thermo Fisher Scientific). The amplicon was sequenced using an MiSeq sequencer (Illumina) after being diluted to a suitable concentration. The sequencer was operated by a procedure in accordance with the MiSeq user guide and instruction manual. Fastq data obtained from sequences was used to collate with known reference sequences and V, D, J, and C regions and determine the CDR3 amino acid sequences using repertoire analysis software (Repertoire Genesis) available from Repertoire Genesis Inc. Sequences that are identical are considered a unique read, and the number of copies thereof was counted to find the ratio of the number with respect to the whole. 1.6. Calculation of Diversity Index Diversity index was found from the resulting read number data for each unique read. The Shannon-Weaver index, Simpson index, inverse Simpson index (1/λ), and DE50 index were calculated by the following mathematical equations. N: total number of reads, ni: number of reads of the ith unique read, S: number of unique reads, S50: number of top unique reads accounting for 50% of total reads. Simpson′⁢s⁢index⁢(1-λ)1-λ=1-∑t=1S(ni(ni-1)N⁡(N-1))[Numeral⁢7]Shannon-Weaver⁢index⁢(H′)H′=-∑i=1SniN⁢ln⁢niN[Numeral⁢8]Normalized⁢Shannon-Weaver⁢index⁢(H′)H′=-∑i=1SniN⁢ln⁢niN/ln⁢N[Numeral⁢9]DE⁢50⁢(D)D=S50S[Numeral⁢10] Calculations were performed as a part of an analysis program of repertoire analysis program in an analysis server of Repertoire Genesis Inc. (http://www.repertoire.co.jp). In addition, a DE30 index and DE80 index using S30(number of top unique reads accounting for 30% of total reads) and S80(number of top unique reads accounting for 80% of total reads) instead of S50(number of top unique reads accounting for 50% of total reads), and Unique indices that directly use the S part were also calculated. (2. Results) 2.1. Clinical Evaluation Patient chest CT or PET image diagnosis was performed before the start of anti-PD-1 antibody therapy and 3 months after therapy to evaluate the therapeutic effect based on morphological evaluation of tumor size and change in tumor size (FIG.1). Table 9 shows the results of determining the effect (complete remission, partial remission, stable disease, and progressive disease) and main therapeutic progress for some of the patients.FIG.2shows chest CT images of some of the patients. 2 out of the 4 patients shown inFIG.2were determined as partial remission (PR) and the other two patients were determined as non-responders at 3 months after therapy. TABLE 9Lung cancer patients and therapeutic progressDeterminedPatientsClassificationeffectTherapeutic progress1Non-responderSDMultiple metastases in bothlungs, lump in the trachea2ResponderPRTwo metastatic lesions inthe left lung, bonemetastasis in thoracicvertebra, loss of twometastatic lesions in theleft lung and reduction inFDG in the thoracic vertebrain half after 3 months3Non-responderSDMetastasis in mediastinallymph nodes4ResponderPRBone metastasis in seventhrib, reduction in FDGaccumulation in the seventhright ribDetermination of therapeutic effect on solid cancer: In the order of effect observed, complete remission (CR) → partial remission (PR) → stable disease (SD) → progressive disease (PD) 2.2. FACS Sorting FACS analysis was performed using PBMCs collected before therapy of 12 lung cancer patients. PBMCs were stained with PE-Cy7-labeled anti-human CD8 antibodies and FITC labeled anti-PD-1 antibodies. The lymphocyte fraction was removed by FSC/SSC gating and dead cells were removed by 7AAD. 7AAD−CD8+PD-1+T cells were collected with a FACS Aria III cell sorter and dissolved into a TRIzol LS RNA extraction reagent.FIG.3shows some of the results of FACS analysis. Lymph gate by FSC/SSC (top row) and double staining of CD8 antibodies and PD-1 antibodies (bottom row) are shown. For the lymphocyte fraction, 7AAD−CD8+PD-1+cell fraction (P3) was fractionated by FACS sorting (FIG.3). The ratio of CD8+PD-1+ cells in the lymphocytes and the ratio of CD8+PD-1+ cells in CD8+ T cells in each patient were compared between anti-PD-1 antibody therapy responders (n=6) and non-responders (n=6) (FIG.4). The former tended to be higher for non-responders compared to that in responders, while a clear difference was not observed for the latter. 2.3. TCR Repertoire Analysis of CD8+PD-1+ T Cells CD8+PD-1+ T cells collected by FACS sorting were used to determine a comprehensive base sequence of a TCR gene with a next generation sequencer in accordance with a method described herein. Table 10 shows the amount of RNA and the number of cells collected by FACS sorting. Table 11 shows the number of TCR reads, number of assigned reads, number of in-frame reads, and number of unique reads acquired from each sample. TABLE 10Amount of RNA and number of CD8+PD-1+ cellsobtained from lung cancer patientsCD8+PD-1+ cellPatient% CD8+PD-1+countRNA (ng)18.6354000121.528.00120002739.04120002747.801300029.2557.17405009166.49600014711.541100025812.08240005497.381050024109.101100025119.5222500511217.2857000128 TABLE 11Acquired TCR read numberTCR αTCR βNo. ofNo. ofNo. of in-No. ofNo. ofNo. ofNo. of in-No. oftotalassignedframeuniquetotalassignedframeuniqueNo.readsreadsreadsreadsreadsreadsreadsreads1192256165832149153757817341913676813415750342217649185372171316744822228416193915998461813167936151540113273636740208824632924197312611420722814967013603697833864633236893055011954551624781473991302155113206399158466150697101956932449081090049170213859411089910972234677918278108359873222910454980042732994524818665616597314321812362226485146742141974121479148281142782139427733023618217966217306712207102146861982461636827547246771210379207223100671112624111193510989073892478061431891364281275512163409146095140299607924465717344316754910808 The acquired TCR read number and the number of unique reads were highest in four non-responder patients. A relationship between the number of TCR reads or the number of unique reads and therapeutic effect was not observed. 2.4. Comparison of Diversity in CD8+PD-1+ T Cells between Therapy Patients Using Diversity Index To compare the diversity of CD8+PD-1+ T cells between anti-PD-1 antibody therapy patients, diversity indices were calculated and compared using TCR read data sequenced from lung cancer patient samples. Diversity indices were calculated in accordance with the mathematical equation described herein using individual unique reads and the number of reads thereof (number of copies). Shannon-Weaver index, normalized Shannon-Weaver index, Simpson index, inverse Simpson index, and DE50 index were used as the diversity indices (FIGS.5and6). Non-parametric Mann-Whitney test (two-tailed test) was used as the significance test. For the TCRα chain, the Shannon-Weaver index exhibited a significantly higher value in responders compared to non-responders (mean±standard deviation, non-responder vs. responder, 2.796±0.9519 vs. 4.081±0.7124, P=0.0411). Similarly, the diversity indices of normalized Shannon-Weaver index, inverse-Simpson index, and DE50 were 0.3327±0.1018 vs. 0.4771±0.05547 (P=0.0260), 7.530±4.906 vs. 23.85±14.02 (P=0.0152), and 0.0006220±0.0003472 vs. 0.001951±0.0005909 (P=0.0022), respectively, which all exhibited a significantly higher value in responders compared to non-responders. Similarly for TCRβ chain, all indices exhibited a significantly higher value in responders compared to non-responders. Mean±standard deviation was 3.129±0.6742 vs. 4.345±0.6555, P=0.0087 (Shannon-Weaver index), 0.3528±0.0612 vs. 0.4815±0.04832, P=0.0087 (Normalized Shannon-Weaver index), 8.198±3.551 vs. 30.25±14.41, P=0.0087 (inverse Simpson index), and 0.0003910±0.00007243 vs. 0.001403±0.0004480, P=0.0022 (DE50). These results elucidated that diversity in CD8+PD-1+ T cells is clearly higher for responders than non-responders. 2.5. Determination of Cutoff Value To predict the effect in an anti-PD-1 antibody therapy patient before therapy, ROC analysis (Receiver Operating Characteristic analysis) was used to set a cutoff value of each diversity index. In ROC analysis, ROC curves are created by plotting the positive rate to the vertical axis as sensitivity and false-positive rate (1-specificity) to the horizontal axis when changing the cutoff value. Methods of determining a cutoff value include a method of using a point where the distance to the top left corner of a ROC curve is minimized as a cutoff value, and a method of calculating the furthest point from a diagonal dashed line where the area under the curve (AUC) is 0.500 on a ROC curves, i.e., (sensitivity+specificity−1) and using the point at the maximum value thereof (Youden index) as the cutoff value.FIG.7shows ROC curves in each diversity index. DE50 shows the highest AUC value for both TCRα and TCRβ relative to other diversity indices, suggesting that DE50 has the best prediction capability. The cutoff value for each diversity index was calculated with a Youden index using R program (ROCR package) and is shown in Table 12. It is predicted that a high therapeutic effect due to anti-PD-1 antibodies cannot be expected with a value less than these cutoff values. TABLE 12Provisional cutoff value in each diversity indexTCRIndexCutoff valueTCRαShannon-Weaver index3.695TCRαInverse Simpson index12.56TCRαNormalized Shannon index0.4345TCRαDE50 index0.001175TCRβShannon-Weaver index3.804TCRβInverse Simpson index17.04TCRβNormalized Shannon index0.4275TCRβDE50 index0.0006853 Although not wishing to be bound by any theory, it is observed that TCRβ repertoire diversity tends to be more clearly distinguishable than TCRα repertoire diversity. It is also observed that a DE50 index tends to be more clearly distinguishable in either repertoire. (3. Discussion) CD8+PD-1+ T cells are known to exert an antitumor effect by releasing immunosuppression with anti-PD-1 antibodies. This experiment found that lung cancer patients with higher diversity in CD8+PD-1+ T cells in peripheral blood of the patients have a higher therapeutic effect from anti-PD-1 antibodies. Tumor infiltrating T cells recognize tumor specific antigens to exert an antitumor effect. Tumor cells accumulate many genetic mutations in the tumorigenic process to produce neoantigens that are not expressed in normal cells. It is known that immunotherapy with an immune checkpoint inhibitor or the like has a high effect on tumor that accumulates more genetic mutations. It is understood that more neoantigens becoming a target of T cells is important for suppressing tumor. Patients on whom anti-PD-1 antibodies are effective are presumed to have a variety of T cells reacting to many neoantigens that are immunosuppressed prior to therapy. It is presumed that the suppression thereof is released by anti-PD-1 antibodies, resulting in a higher therapeutic effect. Lung cancer patients on whom anti-PD-1 antibodies (Nivolumab) are effective are 20 to 30%. If effective patients can be predicted prior to anti-PD-1 antibody therapy, a more effective therapy can be materialized to eliminate wasted medical cost. If TCR repertoire analysis is performed on peripheral blood cells whose sample is readily collected and a diversity index is used as a biomarker, it is expected that this will enable prediction of an effect of anti-PD-1 antibody therapy, which had been impossible up to this point. Example 2: Examination of Change in Diversity Index Due to Number of Reads Diversity indices can be affected and varied by the number of samples, i.e., number of reads obtained by sequencing. For this reason, a certain number of reads (100, 300, 1000, 3000, 10000, 30000, and 80000) were obtained by random sampling from data of each subject obtained in Example 1, and diversity indices (Shannon-Weaver index, Simpson index, normalized Shannon index, inverse Simpson index, DE30 index, DE50 index, DE80 index, Unique30 index, Unique50 index, and Unique80 index) based on said reads were each calculated and plotted. Resampling was carried out 100 times, and the median value of each diversity index was used as a normalized value with respect to each number of reads. A change in the diversity index for TCRα and TCRβ due to the number of reads is shown in each ofFIGS.8and9. It is understood that Shannon, Simpson, and inverse Simpson indices have a mostly constant value regardless of the number of reads, and are hardly affected especially by the number of reads of 10000 reads or greater, which is a level in actual analysis. Meanwhile, it is observed that DE indices have a tendency to be affected by the number of reads, and decrease with an increase in the number of reads. The same tendency is exhibited by DE indices other than DE50, i.e., DE30 and DE80. Thus, it is understood that a threshold value is advantageously set while considering the effect of the number of reads when using a specific value as a threshold value for a DE index. FIGS.10and11show the results of comparing TCR diversity indices of a responder group and a non-responder group in Example 1, which are normalized with respect to 30000 reads. The following Tables 13 to 33 show values of each diversity index normalized with respect to 100, 300, 1000, 3000, 10000, 30000, and 80000 reads for each subject. The section of Clinical shows the therapeutic effect on each subject. Res_min shows the minimum value of index in the responder group. Non_max shows the maximum value of the index in the non-responder group. In Tables 13 to 33, Discrim shows whether the minimum value of index in the responder group is greater than the maximum value of index in the non-responder group. The section of test shows the t-statistic of diversity indices between the responder group and the non-responder group. TABLE 13Parameters in different number of resampled reads (TCRalpha)IndexParameters10030010003000100003000080000ShannonMean values in3.243.543.733.843.944.014.06RespondersMean values in2.192.382.512.602.692.742.78Non-RespondersMinimum values2.392.522.612.672.722.762.78in RespondersMaximum values3.123.513.753.883.994.064.11in Non-RespondersCleardiscriminationttest0.01310.01670.020.02150.0240.02450.0251 TABLE 14IndexParameters10030010003000100003000080000NormalizedMean values in0.890.840.770.690.610.540.50ShannonRespondersMean values in0.730.640.540.480.420.380.35Non-RespondersMinimum values0.840.770.660.560.470.420.38in RespondersMaximum values0.850.790.730.670.600.550.50in Non-RespondersCleardiscriminationttest0.01490.01050.00930.01130.01630.01980.0214 TABLE 15IndexParameters10030010003000100003000080000SimpsonMean values in0.930.940.940.940.940.940.94IndexRespondersMean values in0.800.810.810.810.810.810.81Non-RespondersMinimum values0.880.880.880.890.890.890.89in RespondersMaximum values0.920.930.940.940.940.940.94in Non-RespondersCleardiscriminationttest0.04280.04390.04590.04450.04480.04470.0447 TABLE 16IndexParameters10030010003000100003000080000InverseMean values in18.6721.7923.0023.5523.7523.8423.86SimpsonRespondersIndexMean values in6.857.317.507.517.537.537.53Non-RespondersMinimum values8.158.458.688.878.828.858.84in RespondersMaximum values13.1314.9015.4215.4315.5415.4915.49in Non-RespondersCleardiscriminationttest0.01870.03060.03430.0340.0350.0350.0348 TABLE 17IndexParameters10030010003000100003000080000DE30Mean values in9.155.763.271.630.650.270.12RespondersMean values in7.133.551.420.640.260.120.06Non-RespondersMinimum values5.132.991.450.700.300.130.06in RespondersMaximum values12.505.882.171.180.500.230.12in Non-RespondersCleardiscriminationttest0.39150.14260.04650.0540.06150.07060.0717 TABLE 18IndexParameters10030010003000100003000080000DE50Mean values in20.7713.687.934.061.630.680.32RespondersMean values in13.576.882.861.260.510.230.11Non-RespondersMinimum values17.9511.115.732.580.950.390.18in RespondersMaximum values16.678.703.771.800.810.360.17in Non-RespondersClearYesYesYesYesYesYesYesdiscriminationttest0.00360.00160.00350.00790.01110.01060.0089 TABLE 19IndexParameters10030010003000100003000080000DE80Mean values in57.1441.2724.4212.635.152.161.01RespondersMean values in36.7120.859.704.621.980.890.43Non-RespondersMinimum values44.4433.3316.677.732.921.240.57in RespondersMaximum values56.4138.1021.5911.825.462.511.20in Non-RespondersCleardiscriminationttest0.00550.00190.0020.00560.01430.02430.0283 TABLE 20IndexParameters10030010003000100003000080000Unique30Mean values in3.54.23.73.83.84.04.0RespondersMean values in1.51.81.81.71.71.81.8Non-RespondersMinimum values2222222in RespondersMaximum values3333333in Non-RespondersCleardiscriminationttest0.03090.04530.06740.05660.05660.04740.0474 TABLE 21IndexParameters10030010003000100003000080000Unique50Mean values in8.510.310.011.011.011.211.2RespondersMean values in3.23.23.23.23.03.03.0Non-RespondersMinimum values3433333in RespondersMaximum values6677666in Non-RespondersCleardiscriminationttest0.01080.00380.01250.01040.00830.00930.0093 TABLE 22IndexParameters10030010003000100003000080000Unique80Mean values in23.231.033.836.335.837.036.7RespondersMean values in8.511.012.712.512.312.712.8Non-RespondersMinimum values7999999in RespondersMaximum values22334342404142in Non-RespondersCleardiscriminationttest0.01550.01850.05430.04070.03330.03670.0392 TABLE 23Parameters in different number of resampled reads (TCRbeta)IndexParameters10030010003000100003000080000ShannonMean values in3.383.693.904.034.154.244.31RespondersMean values in2.442.622.782.892.983.053.10Non-RespondersMinimum values in2.652.842.973.093.173.243.29RespondersMaximum values in2.883.093.313.443.553.633.69Non-RespondersCleardiscriminationttest0.00540.0050.00680.00740.00820.00850.0087 TABLE 24IndexParameters10030010003000100003000080000NormalizedMean values in0.910.850.770.690.610.550.51ShannonRespondersMean values in0.760.670.580.510.450.410.38Non-RespondersMinimum values0.830.740.630.550.490.440.40in RespondersMaximum values0.840.750.660.580.520.470.44in Non-RespondersCleardiscriminationttest0.00890.00420.00320.00390.00520.00610.0066 TABLE 25IndexParameters10030010003000100003000080000SimpsonMean values in0.950.950.960.960.960.960.96IndexRespondersMean values in0.840.850.850.850.850.850.85Non-RespondersMinimum values0.900.900.900.900.900.900.90in RespondersMaximum values0.910.920.920.920.920.920.92in Non-RespondersCleardiscriminationttest0.02320.02080.02210.02230.02210.02220.0223 TABLE 26IndexParameters10030010003000100003000080000InverseMean values in22.8427.3029.2729.8430.1430.2330.26SimpsonRespondersIndexMean values in7.697.948.168.178.198.198.20Non-RespondersMinimum values9.6310.1610.2710.3110.3310.3210.31in RespondersMaximum values11.2111.7712.4412.3612.4412.4112.44in Non-RespondersCleardiscriminationttest0.00720.01020.0110.0120.0120.01220.0122 TABLE 27IndexParameters10030010003000100003000080000DE30Mean values in10.116.093.161.440.540.230.10RespondersMean values in6.283.291.350.560.210.090.04Non-RespondersMinimum values8.174.261.850.760.290.120.06in RespondersMaximum values10.006.452.541.020.390.170.08in Non-RespondersCleardiscriminationttest0.04380.02530.01040.01340.01250.01130.0109 TABLE 28IndexParameters10030010003000100003000080000DE50Mean values in21.5113.777.313.421.310.540.25RespondersMean values in13.856.692.751.150.450.190.09Non-RespondersMinimum values16.008.703.721.520.580.250.11in RespondersMaximum values19.059.763.751.430.540.240.11in Non-RespondersClearYesYesYesdiscriminationttest0.00570.00170.00450.00910.01050.01080.0098 TABLE 29IndexParameters10030010003000100003000080000DE80Mean values in56.7738.4420.669.843.791.570.72RespondersMean values in36.5819.408.623.811.520.650.30Non-RespondersMinimum values40.0022.349.904.091.570.680.31in RespondersMaximum values44.6426.0912.385.612.491.100.51in Non-RespondersCleardiscriminationttest0.00250.00280.00560.01160.01650.01940.0202 TABLE 30IndexParameters10030010003000100003000080000Unique30Mean values in4.34.75.25.35.25.25.2RespondersMean values in1.81.81.81.81.81.81.8Non-RespondersMinimum values2222222in RespondersMaximum values3333333in Non-RespondersCleardiscriminationttest0.0120.02210.01290.01290.01290.01290.0129 TABLE 31IndexParameters10030010003000100003000080000Unique50Mean values in10.011.012.212.512.212.312.3RespondersMean values in3.33.53.53.53.33.53.3Non-RespondersMinimum values4444444in RespondersMaximum values5555555in Non-RespondersCleardiscriminationttest0.01070.01110.0130.01330.01310.01210.0128 TABLE 32IndexParameters10030010003000100003000080000Unique80Mean values in25.832.236.337.536.237.337.2RespondersMean values in10.211.011.311.711.311.711.5Non-RespondersMinimum values991110101111in RespondersMaximum values17191820181919in Non-RespondersCleardiscriminationttest0.01830.03060.02770.02830.02530.02750.0271 The diversity indices normalized in this manner were tested to find out whether a significant difference between the responder group (n=6) and non-responder group (n=6) detected in Example 1 is similarly detected for any number of reads. Significance test was performed using unequal variance t-test. Tables 13 to 32 show the results. All diversity indices used in Example 1 such as Shannon, Simpson, and DE50 exhibited a significant difference without being affected by the number of reads. It is understood that the number of reads affects the absolute value of a DE index such as a DE50 index, but not the significant difference, so that the significance of predicting the effect by a diversity index does not change. Therefore, a diversity index can be normalized with respect to the number of reads for comparison. A DE index, whose absolute value changes, is advantageously normalized with respect to the number of reads for comparison with a specific threshold value. A DE50 value is known to separate responder and non-responder groups well in view of the data in Example 1. This Example investigated whether DE50 also distinguishes the groups well when normalized with respect to a certain number of reads (100, 300, 1000, 3000, 10000, 30000, or 80000). Indices and number of reads that show a “clear” separation where the minimum value of the responder group exceeds the maximum value of the non-responder group (AUC of ROC curve is 1) are searched, and shown as Yes in the section of Discrim in Tables 13 to 32. It was demonstrated that such a complete separation is possible with DE50. It was also found that there may not be a significant difference with DE30, a DE50 index exhibits almost the same degree of distinguishability from 10000 reads to 80000 reads. It was demonstrated that a DE50 value is the best among the DE indices, and use of DE50 can achieve unexpected distinguishability. Such normalized diversity indices were used to examine threshold values used in evaluation of responsiveness. First, ROC analysis was performed based on a value obtained by normalizing each diversity index with respect to each number of reads to find a threshold value. The cutoff values of each diversity index that were calculated based on ROC analysis are shown in the following Table 33 (TCRα) and Table 34 (TCRβ). For example, the calculated threshold value for each number of reads can be used when evaluating responsiveness using an index normalized with respect to each number of reads. TABLE 33Cutoff values calculated by ROC analysis (TCRalpha)The number of resampled reads10030010003000100003000080000Shannon3.193.433.613.723.833.913.97Norm_Shannon0.850.810.730.650.570.510.47Simpson0.920.920.930.930.930.930.93Inv_Simpson12.3913.0313.7613.7913.7613.9113.87% DE3011.767.694.000.950.420.180.09% DE5017.1411.045.802.580.960.390.18% DE8045.0031.5217.397.632.941.240.57Unique302332222Unique506798888Unique8020232525272626 TABLE 34Cutoff values calculated by ROC analysis (TCRbeta)The number of resampled reads10030010003000100003000080000Shannon3.233.443.583.663.753.813.85Norm_Shannon0.900.840.750.670.580.520.48Simpson0.950.950.950.950.950.950.95Inv_Simpson18.5720.1221.1121.5421.6621.5921.65% DE307.894.301.850.770.290.120.06% DE5019.0511.633.641.550.580.250.11% DE8051.6735.5317.077.352.661.070.48Unique303344444Unique506999999Unique8016222222212222 Furthermore, to determine a threshold value to be used in accordance with an objective such as (i) rule out non-responder (from the viewpoint of social security cost) or (ii) eliminate missing a responder (from the viewpoint of a physician/therapy), a threshold value of each diversity index when normalized with respect to 30000 reads was further examined. The following Table 35 (TCRα) and Table 36 (TCRβ) show threshold values based on ROC analysis and examples of maximum value or minimum value of diversity indices exhibited by non-responsive and responsive subjects normalized with respect to 30000 reads. Responsiveness can be evaluated to achieve the aforementioned objective by setting a threshold value based on such exemplary values. TABLE 35Cutoff values, minimum values in Responders, and maximumvalues in Non-Responders (TCRalpha)NonCutoff valueResponder_minResponder_maxShannon3.9142.7574.061Norm_Shannon0.5130.4180.547Simpson0.9280.8870.935Inv_Simpson13.9058.85015.486DE300.1820.1310.234DE500.3870.3870.361DE801.2361.2362.513Unique30223Unique50836Unique8026942 TABLE 36Cutoff values, minimum values in Responders, and maximumvalues in Non-Responders (TCRbeta)NonCutoff valueResponder_minResponder_maxShannon3.8083.2423.632Norm_Shannon0.5180.4390.475Simpson0.9540.9030.919Inv_Simpson21.58910.32212.407DE300.1230.1230.168DE500.2460.2470.237DE801.0650.6791.100Unique30423Unique50945Unique80221119 Furthermore, variation in the threshold value of a diversity index due to random sampling was studied. Threshold values calculated from ROC analysis based on a value that is normalized with respect to each number of reads were plotted for each diversity index (FIGS.12and13). Threshold values for Shannon, Simpson, inverse Simpson, and Unique indices are nearly constant regardless of the number of reads. Threshold values of a DE index, which show a tendency to decrease with an increase in the number of reads, were found to be approximated with a linear function in a log-log plot (both logarithmic) (FIGS.12and13). In particular, the correlation coefficient is very high for plots with 3000 reads or more, which is highly likely to be used in actual analysis. For a DE50 index, an α chain can be approximated with y=1892.344x{circumflex over ( )}(−0.8239), and β chain can be approximated with y=993.116x{circumflex over ( )}(−0.8072) (x=number of reads, y=threshold value of DE50 index) (FIG.14). Therefore, it is understood that responsiveness of a subject can be evaluated by comparing a DE50 index calculated with respect to a certain number of reads with a threshold value of a DE50 index for said number of reads derived from the above relationship. Other DE indices can be approximated as follows. DE30,TCRα:y=260.0x{circumflex over ( )}(−0.7008) DE30,TCRβ:y=476.4x{circumflex over ( )}(−0.8032) DE80,TCRα:y=4275.6x{circumflex over ( )}(−0.7905) DE80,TCRβ:y=6151.8x{circumflex over ( )}(−0.8406)  [Numeral 11] (x=number of reads, y=threshold value of DE50 index) To study the range in which such a linear relationship is applicable, 95% confidence intervals were calculated for these fitting curves. The following Table 37 shows the calculated 95% confidence intervals. TABLE 37Best-fit95%IndexGeneParametersvaluesConfidence IntervalsDE30TCRaYinterept2.4152.1642.666Slope (b)−0.7008−0.7703−0.631210{circumflex over ( )}Yintercept(a)260.0145.9463.4TCRbYinterept2.6782.5882.767Slope−0.8032−0.8282−0.778210{circumflex over ( )}Yintercept(a)476.4387.3584.8DE50TCRaYinterept3.2773.2483.307Slope (b)−0.8239−0.8322−0.815710{circumflex over ( )}Yintercept(a)1892.31770.12027.7TCRbYinterept2.9972.9023.092Slope−0.8072−0.8337−0.780710{circumflex over ( )}Yintercept(a)993.1798.01235.9DE80TCRaYinterept3.6313.6183.644Slope (b)−0.7905−0.7942−0.786810{circumflex over ( )}Yintercept(a)4275.64149.54405.5TCRbYinterept3.7893.7483.831Slope−0.8406−0.8522−0.82910{circumflex over ( )}Yintercept(a)6151.85597.66776.4 The degree to which the slope and intercept change in maximum and minimum values when each data point (value) changes 10% was found. The results are shown in the following Table 38. TABLE 38Maximum and minimum values of the slope and Y-interceptwhen the size of change in each data point is 10% (Y = aX{circumflex over ( )}b)TCRalphaRes_MinNonRes_MaxTCRbetaRes_MinNonRes_MaxYintercept3.2773.5092.9972.9973.2132.701Slope (b)−0.8239−0.8787−0.7565−0.8072−0.8574−0.735210{circumflex over ( )}Yintercept (a)1892.33228.5993.1993.11633.1502.3 For determining a responder, the minimum value of a responder and/or maximum value of a non-responder can be used as a threshold value. A linear change in such a value with respect to the number of reads was also studied. It can also be used so that Res_Min or greater is a responder, and NonRes_Max or less is a non-responder. The following Table 39 shows the results of change. TABLE 39Slope and Y-intercept of responder minimum value and non-responder maximum value (Y = aX{circumflex over ( )}b)TCRalphaRes_MinNonRes_MaxTCRbetaRes_MinNonRes_MaxYintercept3.2773.2742.6372.9972.9482.901Slope (b)−0.8239−0.8235−0.685−0.8072−0.7955−0.789810{circumflex over ( )}Yintercept (a)1892.31879.3433.5993.1887.2796.2 Example 3: TCR Diversity in Immunosuppressive Molecule Expressing T Cell Fraction (1. Materials and Methods) 1. Separation of Peripheral Blood Mononuclear Cells (PBMC) 20 mL of whole blood of one anti-PD-1 antibody (nivolumab, Opdivo) therapy responder was collected into a heparin containing blood collection tube. PBMCs were separated by density gradient centrifugation using Ficoll-Paque PLUS (GE Healthcare). The cells were counted with a hemocytometer. The isolated PBMCs were suspended in a cell cryopreservation solution STEM-CELLBANKER (TaKaRa Bio) and stored in a −80° C. deep freezer. 2. Antibody Staining of PBMCs PBMCs were immunostained in accordance with the following procedure.2.1 Cryopreserved PBMCs were dissolved, and the number of cells shown in Table 41 was suspended in Stain Buffer2.2 To remove STEM-CELLBANKER, the cells, after suspended in the Stain Buffer, were centrifuged at 4° C. for 5 minutes at 800×g, and washed twice.2.3 The cells were suspended in Stain buffer. Antibodies in the following Table 40 were added in accordance with the package insert and reacted with the cells while shaded at room temperature for 30 minutes. TABLE 40Sorted cell fraction and antibodyNo.Sorted fractionAntibody1CD8+PD1+7AAD−PECy7 anti-human CD8 (BD)AF488 anti-human PD-1 (R&D)7AAD2CD8+4-188+7AAD−PECy7 anti-human CD8 (BD)PE anti-human CD137 (BD)7AAD3CD8+TIM3+7AAD−PECy7 anti-human CD8 (BD)PE anti-human TIM3 (CD366) (BD)7AAD4CD8+OX40+7AAD−FITC anti-human CD8 (BD)PE/Cy7 anti-human CD134(BioLegend)7AAD5CD8+TIGIT+7AAD−PECy7 anti-human CD8 (BD)FITC anti-human TIGHT(ThermoFisher)7AAD6CD8+CTLA-4+7AAD−FITC anti-human CD8 (BD)PE/Cy7 anti-human CD152(BioLegend)7AAD2.4 After washing, the cells were suspended in 100 μL of Stain buffer. 5 μL of 7-AAD was added and reacted while shading for 10 minutes at room temperature.2.5 The cells were supplemented with 500 μL of Stain buffer and filtered. The above sorted fractions were then sorted and separated using a BD FACSAria III cell sorter (BD Bioscience) or FACSMelody cell sorter (BD Bioscience).2.6 The sorted fraction was collected by centrifugation at 4° C. for 5 minutes at 800×g.2.7 After removal while keeping 250 μL of the supernatant, 750 μL of TRIzol LS reagent (Invitrogen) was added and pipetted to dissolve the cells.2.8 TCR repertoire analysis after RNA extraction was performed in accordance with the method in “1.3. RNA extraction”, “1.4. Synthesis of complementary DNA and double stranded complementary DNA”, and “1.5. PCR” of Example 1.2.9 After antibody staining, the cells were suspended into 2 mL of Stain buffer and centrifuged at 4° C. for 5 minutes at 800×g, and then the supernatant was disposed, and the cells were washed twice. (2. Results) Table 41 shows the percentage of CD8+PD1+, CD8+4-1BB+, CD8+TIM3+, CD8+OX40+, CD8+TIGIT+, and CD8+CTLA4+ T cell fractions in the lymphocytes (%) and the number of cells collected by FACS sorting. About 1×104to 1×105T cells coexpressing CD8 and each molecular marker were collected. RNA was extracted from these T cell fractions, and TCR repertoire analysis was performed in accordance with a conventional method. Table 42a and Table 42b show the results of TCR sequence analysis, the number of total reads, number of unique reads, and number of in-frame reads obtained from each sample. 100000 or more reads were able to be acquired for each sample. TABLE 41Percentage of sorted cells and sorted cell countNumber of% positivecollectedFractionCell countcellscellsCD8+PD1+4.0 × 1061.0%14,922CD8+4-1BB+4.0 × 1061.2%21,337CD8+TIM3+2.7 × 10611.4%83,027CD8+OX40+6.0 × 1062.4%31,072CD8+TIGIT+2.0 × 1068.4%43,694CD8+CTLA4+8.0 × 1060.76%20,340 TABLE 42aNumber of acquired reads (TCRα chain)Number ofNumber ofNumber ofin-frameCell fractiontotal readsunique readsunique readsCD8+PD1+17049663154705CD8+4-1BB+20944675925429CD8+TIM3+18613267855486CD8+OX40+2668021363010392CD8+TIGIT+20474574565694CD8+CTLA4+15769284006107CD8+2149661393810489 TABLE 42bNumber of acquired reads (TCRβ chain)Number ofNumber ofNumber ofin-frameCell fractiontotal readsunique readsunique readsCD8+PD1+14081562805157CD8+4-1BB+16869663785118CD8+TIM3+5608438912978CD8+OX40+1998281211410026CD8+TIGIT+10690275135969CD8+CTLA4+14722386567146CD8+1687321198410064 (Commonality in TCR Repertoire Among Each T Cell Fraction) Clones that are in common among CD8P+PD1+, CD8+4-1BB+, CD8+TIM3+, CD8+OX40+, CD8+TIGIT+, and CD8+CTLA4+ T cell fractions were compared for sequences of TCR clones acquired by TCR repertoire analysis. Table 43 shows TCR clones that are present in common in all fractions or in multiple fractions. It was elucidated that TCR clones that are present at a high frequency in the CD8+PD-1+ fraction are also present at a high frequency in the CD8+4-1BB+, CD8+TIM3+, CD8+OX40+, CD8+TIGIT+, or CD8+CTLA4+ fraction. This suggests the possibility that CD8+PD-1+ T cells coexpress 4-1BB, TIM3, OX40, TIGIT, or CTLA4 molecules. The correlation was studied for the number of reads for TCR clones among each T cell fraction (FIG.15). TCR clones that were present at a high frequency were also present in each T cell fraction in common, exhibiting a high correlation. The ratio of reads of TCR clones in common between CD8+PD-1+ T cells and CD8+4-1BB+, CD8+TIM3+, CD8+OX40+, CD8+TIGIT+, or CD8+CTLA4+ T cells was studied (Table 44): The ratio of reads of CD8+PD-1+ TCR clones in each fraction was significantly higher in CD8+4-1BB+, CDB+TIM3+, CD8+OX40+, CD8+TIGIT+, or CD8+CTLA4+ T cells than the control CD8+ T cells. This suggested that tumor specific TCR of tumor specific T cells contained in CD8+PD-1+ T cells is also included at a high frequency in 4-1BB, TIM3, OX40, TIGIT, or CTLA-4 positive T cell fraction. In view of the above, each of T cell fractions of CD8+4-1BB+, CD8+TIM3+, CD8+OX40+, CD8+TIGIT+, and CD8+CTLA4+ in peripheral blood is expected to be usable as a biomarker by TCR diversity. TABLE 43TCRβ chain clone present in common in each T cell fraction(SEQ ID NOS: 11-60)CD8+CD8+CD8+CD8+CD8+CD8+PD-4-TIM3OX40TIGITCTLA4No.TRVTRJCDR31+1BB+++++1TRBV11-TRBJ2-CASSPVPTFLGSYNEQFF556512961101441056381937112TRBV7-9TRBJ2-CASSPLAGVAYNEQFF5229023812443117564013TRBV6-1TRBJ2-CASSEEAGGVETQYF4983015601107654TRBV29-TRBJ1-CSVLMWTGDLNYGYTF405318135534386125TRBV30TRBJ2-CAWSVPSRETQYF3989013010056TRBV4-1TRBJ2-CASSQGTFYEQYF29240112657177TRBV28TRBJ2-CASSLYPPGGANVLTF24241576924478518689863068TRBV30TRBJ2-CAWTFSNEQYF20413695278902205372879TRBV13TRBJ2-CASSLGPGTSGRVSYEQY186512410233990908747F10TRBV29-TRBJ2-CSVVTSNSDTQYF1708180039555501311TRBV29-TRBJ2-CSVEEGDTGELFF870014708301212TRBV15TRBJ2-CATSRDFGGSYEQYF74578291487880713TRBV28TRBJ2-CASSLYPPGGVNVLTF6484352813614TRBV28TRBJ2-CASRSSGDNEQFF5720016281074244115TRBV11-TRBJ2-CANSPVPTFLGSYNEQFF4728121351116TRBV11-TRBJ2-CVSSPVPTFLGSYNEQFF284030881117TRBV7-9TRBJ2-CASSPVPTFLGSYNEQFF21711140118TRBV7-9TRBJ2-CASSTLAGVAYNEQFF1502632119TRBV28TRBJ2-CVSSLYPPGGANVLTF143274124620TRBV7-6TRBJ2-CASSPLAGVAYNEQFF1301110121TRBV30TRBJ2-CAWTLSNEQYF10261133722TRBV7-9TRBJ2-CASRPLAGVAYNEQFF1002110123TRBV30TRBJ2-CAWGVPSRETQYF1001110524TRBV11-TRBJ2-WASSPVPTFLGSYNEQFF922502261125TRBV11-TRBJ2-CASSTVPTFLGSYNEQFF91100711126TRBV11-TRBJ2-CASSPVPTFLGSYKEQFF8911511127TRBV28TRBJ2-CASSRYPPGGANVLTF81075222628TRBV11-TRBJ2-CASSPVPTFLGSYNGQFF81410711129TRBV7-9TRBJ2-CASSPLAGVAYDEQFF801220130TRBV11-TRBJ2-CASSPVPTCLGSYNEQFF8900921131TRBV11-TRBJ2-CASSPGPTFLGSYNEQFF715121731132TRBV28TRBJ2-CASSLYPPGGANVLSF7105105633TRBV7-9TRBJ2-CASSPLAGVAYNEQFF700511734TRBV11-TRBJ2-CASSPVPTFRGSYNEQFF712201001135TRBV11-TRBJ2-CASSPVTTFLGSYNEQFF61010741136TRBV11-TRBJ2-CASSPVPTLLGSYNEQFF6601511137TRBV11-TRBJ2-CASSPVPTFLGAYNEQFF612011011138TRBV7-9TRBJ2-CASSPLVGVAYNEQFF604410139TRBV7-9TRBJ2-CASSPLAGVDYNEQFF604110140TRBV7-9TRBJ2-CASSPLAGVAYNEQFL600211141TRBV29-TRBJ2-CSVVTSNSDAQYF6101101342TRBV13TRBJ2-RASSLGPGTSGRVSYEQY6003527F43TRBV11-TRB72-CASSPAPTFLGSYNEQFF6900711144TRBV11-TRBJ2-CASNPVPTFLGSYNEQFF52021411145TRBV7-9TRBJ2-CANSPLAGVAYNEQFF503113146TRBV11-TRBJ2-CASSPVSTFLGSYNEQFF5610631147TRBV11-TRBJ2-CASSPVPTFPGSYNEQFF5201311148TRBV11-TRBJ2-CASSPDPTFLGSYNEQFF5810511149TRBV7-9TRBJ2-CASSPLAGVAYNGQFF503220150TRBV7-9TRBJ2-CASSPLAGGAYNEQFF5024101 TABLE 44Percentage of reads accounted for by CD8+PD-1+TCR clones% CD8+PD-1+TCR cloneT cell subpopulationTCRαTCRβCD8+4-1BB+38.9220.63CD8+TIM3+17.9818.41CD8+OX40+9.019.31CD8+TIGIT+31.9050.13CD8+CTLA4+8.7013.81Control (CD8+)3.795.98 TCR repertoire analysis was performed on CD8+PD1+, CD8+4-1BB+, CD8+TIM3+, CD8+OX40+, CD8+TIGIT+, and CD8+CTLA4+ fractions separated by FACS sorting from PBMCs of therapy responders to calculate the Shannon index, normalized Shannon index, inverse Simpson index, and DE50 index. As a result, the diversity indices of CD8+PD1+ T cells exhibited the same degree of diversity as CD8+4-1BB+, CD8+TIM3+, CD8+OX40+, CD8+TIGIT+, or CD8+CTLA4+ T cells of the same patient. All fractions exhibited diversity indices that were about the same as CD8+PD-1+ T cells of therapy responders (N=6) and were clearly higher than CD8+PD-1+ T cells of non-responders (N=6) (FIGS.16and17). In view of the above, it is expected that not only CD8+PD1+ T cells, but also CD8+ T cells with a T cell surface marker such as 4-1BB+, TIM3+, OX40+, TIGIT+, or CTLA4+ can be analyzed and used as a biomarker for predicting a therapeutic effect of an immune checkpoint inhibitor. (Notes) As disclosed above, the present invention is exemplified by the use of its preferred embodiments. However, it is understood that the scope of the present invention should be interpreted based solely on the Claims. It is also understood that any patent, any patent application, and any references cited herein should be incorporated herein by reference in the same manner as the contents are specifically described herein. INDUSTRIAL APPLICABILITY A diversity index obtained by TCR repertoire analysis in peripheral blood cells whose sample is readily collected can be used as a biomarker for predicting the effect of cancer immunotherapy. SEQUENCE LISTING FREE TEXT SEQ ID NO: 1 BSL-18E primerSEQ ID NO: 2 P20EA primerSEQ ID NO: 3 P10EA primerSEQ ID NO: 4 P22EA-ST1-R primerSEQ ID NO: 5 CA1 primerSEQ ID NO: 6 CA2 primerSEQ ID NO: 7 CA-ST1-R primerSEQ ID NO: 8 CB1 primerSEQ ID NO: 9 CB2 primerSEQ ID NO: 10 CB-ST1-R primerSEQ ID NO: 11 to 60 CDR3 sequence of each TCRβ chain clone in Example 3
124,595